156
UNIVERSIDADE DE LISBOA FACULDADE DE FARMÁCIA DEPARTAMENTO DE MICROBIOLOGIA E IMUNOLOGIA Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga Maria Elviro Mestre DOUTORAMENTO EM FARMÁCIA MICROBIOLOGIA 2012

Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

UNIVERSIDADE DE LISBOA

FACULDADE DE FARMÁCIA

DEPARTAMENTO DE MICROBIOLOGIA E IMUNOLOGIA

Mycobacterium tuberculosis host adaptation and

evolution reflected by defense mechanisms

against oxidative stress

Olga Maria Elviro Mestre

DOUTORAMENTO EM FARMÁCIA

MICROBIOLOGIA

2012

Page 2: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga
Page 3: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

UNIVERSIDADE DE LISBOA

FACULDADE DE FARMÁCIA

DEPARTAMENTO DE MICROBIOLOGIA E IMUNOLOGIA

Mycobacterium tuberculosis host adaptation and

evolution reflected by defense mechanisms

against oxidative stress

Olga Maria Elviro Mestre

Tese orientada pela Prof.ª Dr.ª Brigitte Gicquel e pela Prof.ª Dr.ª Madalena

Pimentel, especialmente elaborada para a obtenção do grau de doutor em

Farmácia (Microbiologia)

2012

Page 4: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

The studies presented in this thesis were performed at Unité de Génétique Mycobacteriénne, Institut

Pasteur, Paris, under de supervision of Professor Brigitte Gicquel and Professor Madalena Pimentel.

Olga Maria Elviro Mestre was financially supported by a PhD fellowship (SFRH/BD/39079/2007) from

Fundação para a Ciência e Tecnologia (FCT), Lisboa, Portugal. The work presented in this thesis was

supported by the TB-ADAPT (LSHP-CT-2006-037919) project, funded by the European Commission

under the Health Cooperation Work Programme of the 6th Framework Programme, as well as by TB-VIR

(Grant agreement n° 200973) and NEWTBVAC (HEALTH-F3-2009-241745) projects, from the 7th

Framework Programme.

De acordo com o disposto no ponto 1 do artigo nº40 do regulamento de Estudos Pós-Graduados da

Universidade de Lisboa, deliberação nº 93/2006, publicado em Diário da Républica – II série nº 153 – 5

de Julho de 2003, a autora desta dissertação declara que participou na concepção e execução do

trabalho experimental, interpretação dos resultados obtidos e redacção dos manuscritos.

Page 5: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

III

ABSTRACT

Part of the success of Mycobacterium tuberculosis lies in its ability to thrive inside macrophages,

where it is exposed to strong antimicrobials molecules, such as reactive oxygen species (ROS).

However, the role of defense mechanisms against ROS in M. tuberculosis pathogenesis remains

unclear. We used, for the first time, a functional genomic approach to investigate M. tuberculosis

responses against ROS. By screening a transposon mutant library we identified a high number of

mutants, with increased susceptibility to H2O2, in genes related to cell envelope functions, including

mmpL9. This revealed the importance of M. tuberculosis cell envelope against oxidative stress.

However, we have also identified genes implicated in other kind of defense mechanism against

ROS, such as moaD1. Infection of human macrophages has shown that both mmpL9 and moaD1

have a role in M. tuberculosis intracellular lifestyle, and previous studies suggested the same for

several other genes identified during our screening. Therefore, these represent potential virulence

factors useful for the development of future anti-tuberculosis strategies.

DNA repair, recombination and replication (3R) are important mechanisms in maintaining genome

stability by repairing DNA damages, such as those induced by ROS, however, variations on 3R genes

potentially increase genomic variability, due to an increase in mutation rates. Thus, analysis of

polymorphisms in 3R genes could indicate which strains are more prone to adapt and evolve. We

have analyzed polymorphisms in 3R genes in a collection of strains of a successful family of M.

tuberculosis strains, the Beijing/W family. Specific 3R polymorphisms were found to characterize

particular groups of Beijing/W strains for which a phylogeny was constructed. Certain groups were

found to be predominant, suggesting that strains of these genotypes might have some selective

advantage. Therefore, particular 3R SNPs may define pathogenic features that have contributed to

the evolution of the Beijing/W family.

Keywords: Mycobacterium tuberculosis, Reactive oxygen species, macrophage, DNA repair

recombination and replication, The Beijing/W family, Polymorphisms

Page 6: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

IV

Page 7: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

V

RESUMO

Mycobacterium tuberculosis, a bactéria responsável pela tuberculose, infecta cerca de 1/3 da

população mundial e é responsável pela morte de aproximadamente 1,5 milhões de pessoas por

ano. Estes números podem ser explicados, em parte, pela capacidade desta bactéria persistir no

interior de macrófagos, num ambiente extremamente agressivo, no qual é exposta a diferentes

moléculas antimicrobianas tais como as espécies reactivas de oxigénio. Estas, sendo altamente

reativas, causam danos em todo o tipo de moléculas incluindo lípidos, proteínas e DNA. Desta

forma, os mecanismos de defesa utilizados pelo M. tuberculosis contra as espécies reativas de

oxigénio são sem dúvida importantes para este, porém, o seu papel na virulência e patogénese

desta bactéria ainda é pouco claro.

O trabalho desenvolvido nesta tese consistiu na investigação dos mecanismos de defesa

utilizados pelo M. tuberculosis contra as espécies reactivas de oxigénio. Com este objectivo,

realizou-se pela primeira vez, um screening de uma biblioteca de mutantes construída por

transposição, na estirpe clinica da família Beijing/W - GC1237, de forma a selecionar mutantes

sensíveis a espécies reativas de oxigénio. A análise de cerca de 6000 mutantes permitiu a selecção

de 18, sensíveis a peróxido de hidrogénio, para os quais a amplificação e posterior sequenciação do

sítio de inserção do transposão levaram à identificação de 11 genes. Verificou-se que grande parte

dos genes apresenta funções associadas ao invólucro celular bacteriano, o que revela que o

invólucro do M. tuberculosis representa a primeira barreira de defesa contra as espécies reactivas

de oxigénio nesta bactéria. O gene mmpL9 foi um dos identificados, cuja proteína se encontra

possivelmente envolvida no transporte de lípidos para o invólucro celular. Curiosamente, três

mutantes com diferentes mutações neste gene foram seleccionados. Identificaram-se também

alguns genes envolvidos noutro tipo de mecanismos tais como, moaD1, envolvido na síntese do

cofactor molibdénio.

Uma vez que o M. tuberculosis tem de fazer face às espécies reativas de oxigénio no interior de

macrófagos, o passo seguinte consistiu em analisar o fenótipo de alguns destes mutantes nestas

células. Para o efeito, macrófagos humanos foram infectados com um dos mutantes no gene

mmpL9 ou com o mutante no gene moaD1, uma vez que estudos anteriores demonstraram o papel

de diferentes MmpLs na virulência desta bactéria e que outros genes envolvidos na síntese do

cofactor molibdénio são importantes para a sobrevivência do M. tuberculosis no interior de

macrófagos. Em ambos os casos observou-se uma diminuição na capacidade de crescimento destas

Page 8: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

VI

estirpes no interior destas células, em comparação com a estirpe selvagem. A complementação

destas estirpes mutantes restaurou o fenótipo da estirpe selvagem, o que demonstra o papel

destes genes no crescimento de M. tuberculosis em macrófagos humanos. Porém, a

complementação restaurou apenas parcialmente a sensibilidade destes mutantes às espécies

reactivas de oxigénio in vitro, em comparação com a estirpe selvagem. No entanto, mmpL9 e

moaD1 deverão ter um papel na resposta às espécies reativas de oxigénio em M. tuberculosis, caso

contrário, a complementação não teria restaurado de todo o fenótipo selvagem, nas condições

referidas.

A comparação da sequência do gene mmpL9 da nossa estirpe selvagem, GC1237, com a estirpe

de referência laboratorial, H37Rv, revelou a presença de uma deleção de um nucleótido na posição

218. Contudo, os nossos resultados obtidos a partir da infecção de macrófagos com o mutante

mmpL9 e com a mesma estirpe após complementação da mutação sugerem que o gene mmpL9

deverá ser expresso apesar desta deleção. É possível que ocorra uma reniniação da tradução a

partir de um codão de iniciação situado a montante da deleção, na posição 258, no gene mmpL9.

Para além disso, a complementação do nosso mutante mmpL9 com o respectivo gene da estirpe

H37Rv, restaurou apenas parcialmente o fenótipo da estirpe selvagem, GC1237, durante a infecção

de macrófagos. Estas observações sugerem que a proteína MmpL9 na estirpe GC1237 poderá ser

diferente da existente na estirpe H37Rv. A análise da sequência de todos os genes mmpL existentes

no genoma de M. tuberculosis, numa coleção de estirpes que representam diferentes famílias,

sugeriu que a deleção identificada poderá realmente ter um efeito na proteína. Verificou-se ainda

que esta deleção é aparentemente específica para todas as estirpes modernas da família Beijing.

Isto indica que a deleção do nucleótido 218 poderá ter conferido alguma vantagem selectiva a

estas estirpes que contribuiu para a sua evolução.

Para além de moaD1 e mmpL9, alguns dos outros genes identificados durante o screening como

importantes para a resposta de M. tuberculosis às espécies reativas de oxigénio, foram igualmente

descritos, em estudos anteriores, como importantes para a capacidade desta bactéria sobreviver

intracelularmente. Isto indica uma correlação entre sensibilidade ao stress oxidativo e capacidade

de sobreviver ou replicar-se no interior de macrófagos. No entanto, a infecção de macrófagos

tratados com inibidores da NADPH oxidase, a enzima que produz as espécies reativas de oxigénio

nestas células, com os mutantes nos genes mmpL9 e moaD1, não permitiu confirmar esta hipótese.

É possível que estes mutantes sejam sensíveis a outras moléculas antimicrobianas encontradas no

interior do macrófago, tais como as espécies reativas de azoto, que, em conjunto com as espécies

Page 9: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

VII

reativas de oxigénio, são responsáveis pelo crescimento atenuado observado em macrófagos. Em

conclusão, este trabalho permitiu a identificação de genes envolvidos na defesa do M. tuberculosis

contra as espécies reactivas de oxigénio, os quais aparentam ter igualmente um papel na

sobrevivência desta bactéria nas suas células hospedeiras, os macrófagos. Estes genes representam

potenciais factores de virulência que poderão ser úteis para o desenvolvimento de futuros

antibióticos ou vacinas.

Numa segunda parte desta tese, realizou-se uma análise dos polimorfismos existentes em genes

de reparação, recombinação e replicação (3R) de DNA numa família específica de estirpes de M.

tuberculosis, a família Beijing. Os mecanismos de reparação, recombinação e replicação são

extremamente importantes na manutenção da estabilidade genómica, através da reparação de

danos causados no DNA, tais como os induzidos pelas espécies reativas de oxigénio. Por outro lado,

estes mecanismos podem ser igualmente responsáveis pela introdução de variabilidade genética.

Polimorfismos nos genes que codificam para as proteínas envolvidas nestes mecanismos poderão

induzir um aumento na taxa de mutação, e consequentemente, a acumulação de mutações

vantajosas para as estirpes, como por exemplo, mutações responsáveis pela resistência a

antibióticos. Deste modo, a análise de polimorfismos nos genes 3R poderá indicar que estirpes

estão mais predispostas a adaptar-se ou evoluir. A família Beijing é uma família de M. tuberculosis

que apresenta uma maior virulência e que tem sido identificada um pouco por todo o mundo como

estando frequentemente associada a resistência a antibióticos. Deste modo, analisaram-se

polimorfismos em genes 3R numa coleção de estirpes Beijing, isoladas em várias partes do mundo.

Diferentes polimorfismos foram identificados que permitiram a discriminação de vários grupos,

para os quais foi construída uma árvore filogenética. Estes resultados foram congruentes com os

resultados obtidos utilizando outros marcadores genéticos, o que significa que as mesmas

associações filogenéticas foram obtidas, para a colecção de estirpes utilizada neste estudo, usando

polimorfismos nos genes 3R ou outro tipo de marcadores genéticos.

Uma grande percentagem das estirpes foi agrupada num dos grupos, Bmyc10, que incluiu

estirpes isoladas em diferentes partes do mundo. Um estudo anterior demonstrou que estirpes

com características genéticas que definem as deste grupo são igualmente predominantes numa

outra região geográfica, não representada no nosso estudo. Em conjunto, estes resultados

sugerem que o genótipo das estirpes do grupo Bmyc10 poderá ter-lhes conferido alguma

vantagem selectiva que permitiu a sua evolução. Isto é igualmente sugerido num estudo anterior,

no qual a caracterização do efeito da mutação no gene mutT2, que caracteriza todas as estirpes do

Page 10: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

VIII

grupo Bmyc10, indica que esta induz uma alteração na função da proteína que pode ser vantajosa

para as estirpes que a possuem. Portanto, alguns dos polimorfismos identificados no nosso estudo

poderão ter um efeito na função das respectivas proteínas, o que significa que este tipo de análise

é importante para compreender o significado dos mesmos.

Em conclusão, a análise de polimorfismos em genes 3R demonstrou que diferentes grupos

podem ser identificados na família Beijing caracterizados por polimorfismos específicos. Estes

podem ter conferido alguma vantagem que permitiu a expansão de certos grupos e contribuiu para

a evolução desta família. A análise da variabilidade genética entre estirpes é importante uma vez

que estas podem reflectir diferenças patogénicas. Este estudo poderá contribuir para aprofundar

os nossos conhecimentos acerca dos mecanismos que determinam o êxito das estirpes da família

Beijing.

Palavras-Chave: Mycobacterium tuberculosis; Espécies reactivas de oxigénio; Macrófago;

Reparação, recombinação e replicação de ADN; Família Beijing; Polimorfismos

Page 11: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

IX

ACKNOWLEDGMENTS

I would like to thank, first of all, to my supervisor, Professor Brigitte Gicquel, for accepting me in her

laboratory, and for all the support, help and guidance during the last six years. Thank you for believing

in me and for giving the opportunity to learn all the things I have learnt that allowed me to evolve

professionally and personally and became the person I am today. Pour tout ça, un grand Merci.

Queria agradecer igualmente à Professora Madalena Pimentel, co-orientadora desta tese. Agradeço

todo o apoio que me deu durante estes cinco anos em que, esteve sempre disponível para nos

reunirmos e discutirmos sobre a evolução dos trabalhos, e para me ajudar e aconselhar. Obrigada pela

sua ajuda e apoio, principalmente nesta fase final, foi muito importante para mim.

Ao Tiago, por ter acreditado em mim e me ter apoiado incondicionalmente. Sem ti este trabalho nunca

teria sido possível. Obrigada por todo o apoio e carinho, que tu e a Zrinka sempre me deram. Por me

teres ouvido e aconselhado e por me teres orientado e guiado. Vou estar eternamente agradecida por

tudo.

À Helena, a minha grande amiga, a quem dificilmente conseguirei exprimir o quanto estou agradecida.

Pelo apoio incondicional e pela paciência que tiveste, sobretudo nos momentos mais difíceis. Obrigada

por todos os momentos únicos e inesquecíveis que passámos juntas no laboratório e em Paris!

Obrigada pela amizade, que vai para sempre durar, não tenho dúvidas.

À Mena, amiga do coração, foste um apoio imprescindível neste cinco anos. Este percurso não teria

sido possível se não tivesses estado presente para me ajudares, escutares e aconselhares. Por isso,

muito obrigada a ti, ao Elias, e ao Sebastian por tornarem a minha estadia em Paris mais fácil e por me

proporcionarem momentos únicos e especiais.

À Soraia pela amizade e por teres estado sempre presente e disponível para me apoiares apesar da

distância, e sobretudo pela ajuda e apoio nesta fase final, muito obrigada por tudo amiga.

Ao Bruno e à Elisabete, que estiveram sempre disponíveis para me apoiar, obrigada! Ao Hugo, muito

obrigada pela constante preocupação e apoio. Obrigada aos três pelos momentos inesquecíveis

passados juntos em Paris, nunca esquecerei!

Ao Amine, merci beaucoup pour tous les moments passés ensemble au labo, pour travailler et aussi

pour décontracter et rigoler. Pour toute ton aide et soutien et aussi pour ta patience, essentiel pendant

ces dernières années, je te remercie vraiment du fond du cœur.

Page 12: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

X

À Raquel, por todas as horas passadas no P3, foste uma ajuda importante, obrigada, pelo apoio e pela

preocupação e carinho que tu e Marco me deram.

À tous les personnes avec qui j’ai travaillé au laboratoire à l’Institut Pasteur, en particulier à Sandrine et

Véronique, pour toute votre aide, soutient et compagnie, pour les bons moments passés ensemble, je

vous remercie et je m’en souviendrais toujours!

A special thanks to Alessandro for all the advices, support, discussions, and long hours spent in the P3,

and off course for the Italian meals in the weekends!

A todos os meus amigos, em especial à Telma que sempre se preocupou e sempre me apoiou, obrigada

pela tua amizade incondicional.

À minha família, primos, tios e avós muito obrigada pela constante preocupação e apoio!

Aos meus pais, pelo amor e apoio incondicional que sempre me deram, sem eles sem dúvida alguma

que eu não teria conseguido chegar até aqui e ser o que sou hoje. Por tudo isso e pela preocupação e

ajuda, não tenho palavras que possam agradecer tudo o que já fizeram por mim, a não ser dizer que vos

adoro e que tenho muito orgulho em ser vossa filha.

À minha irmã e ao meu cunhado, e claro, às minhas sobrinhas lindas, pelo amor que me dão e por todo

o apoio que me deram ao longo destes anos, sobretudo pela paciência nesta fase final. A vossa ajuda

foi imprescindível para finalizar esta tese, muito muito obrigada, adoro-vos!

Ao meu irmão e cunhada obrigada pelo carinho, apoio e preocupação incondicional, muito obrigada!

Aos meus sogros, ao Nuno e à Inês, por me tratarem de uma forma tão especial, por se preocuparem

comigo e pelo apoio e força que sempre me deram, obrigada.

Por fim, ao Rui, pelo teu amor, apoio, ajuda, compreensão e sobretudo pela enorme paciência que

tiveste durante estes cinco anos. Sem ti seguramente eu não teria tido a força que tive para concluir

esta tese. Obrigada por tudo e por fazeres parte da minha vida. Por tudo isto e muito mais estarás

sempre no meu coração.

Page 13: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XI

ABREVIATIONS

3R DNA repair, recombination and replication 8-oxoG 7,8-dihydro-8-oxo-2'-deoxyguanosine

Ahp Alkyl hydroperoxide

AIDS Acquired Immunodeficiency Syndrome

AP sites apurinic or apyrimidinic sites

BCG Bacillus Calmette-Guérin

BER Base excision repair

CGD Chronic Granulomatous Disease

CFU Colony-Forming Unit

CMN Corynebacterium Mycobacterium Nocardia

DAT Diacyltrehaloses

DCs Dendritic Cells

DC-SIGN Dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin

DMSO Dimethyl Sulfoxide

DNA Deoxyribonucleic Acid

DosR Dormancy regulon

DPI Diphenyleneiodonium

DSBs Double Strand Breaks

DTM-PCR Deletion-Targeted Multiplex PCR

EEA1 Early endosome antigen 1

ESAT-6 Early secreted antigenic target protein 6

GMD GDP-D-Mannose-Dehydratase

HIV Human Immunodeficiency Virus

HR Homologous Recombination

iNOS inducible Nitric Oxide Synthase

IFNγ Interferon gamma

LAM Lipoarabinomannan

LM Lipomannan

LpdC Lipoamide dehydrogenase C

LSPs Large Sequence Polymorphisms

ManLAM Mannose-capped Lipoarabinomannan

MDR Multi-Drug Resistant

MIRUs Mycobacterial Interspersed Repetitive Units

MLST Multilocus Sequence Typing

MmpL Mycobacterial Membrane Protein Large

MMR Mismatch repair

MNNG N-methyl-N’-nitro-N-nitrosoguanidine

MoCo Molybdenum Cofactor

MOI Multiplicity Of Infection

Page 14: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XII

MR Mannose Receptor

Msr Methionine Sulfoxide Reductase

NER Nucleotide Excision Repair

NHEJ Non-Homologous End-Joining

nsSNPs non-synonymous SNPs

NTF Noise Transfer Function

OD Optical Density

PCR Polymerase Chain Reaction

PDIM Phthiocerol Dimycocerosate

PGLs Phenolic Glycolipids

Phox Phagocyte NADPH oxidase

PI3P phosphatidylinositol-3-phosphate

PIMs Phosphatidyl-myo-Inositol Mannosides

pks Polyketide biosynthesis

PMA Phagosome maturation arrest/ Phorbol 12-myristate 13-acetate (reagent)

PPRs Pattern Recognition Receptors

rhM-CSF Recombinant Human Macrophage Colony Stimulating Factor

RD Region of Difference

RecA-NDp RecA/LexA-independent promoter

RFLP Restriction Fragment Length Polymorphism

RLUs Relative Light Units

RND Resistance, nodulation and cell division proteins

RNS Reactive nitrogen species

ROS Reactive Oxygen Species

SLs Sulfatides

SNPs Single Nucleotide Polymorphisms

SOD Superoxide Dismutase

Spoligotyping Spacer Oligonucleotide Typing

SSBs Single Strand Breaks

sSNPs synonymous SNPs

TAT Triacyltrehaloses

TB Tuberculosis

TDM Trehalose Dimycolate

TDR Totally Drug resistant

TLRs Toll-like Receptors

TMM Trehalose Monomycolate

Trx Thioredoxin

UV Ultraviolet

VNTR Variable Number of Tandem Repeat

Vps33B Vacuolar Protein Sorting 33B

VitD3 Vitamin D3

WHO World Health Organization XDR Extensively-drug resistant

Page 15: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XIII

PUBLICATIONS

Most of the work presented in this thesis is included in the following published or submitted

manuscripts in international peer-reviewed journals:

Mestre O, Hurtado-Ortiz R, Dos Vultos T, Namouchi A, Cimino M, Pimentel M, Neyrolles O,

Gicquel B. (2012). High throughput phenotypic selection of Mycobacterium tuberculosis mutants

with impaired resistance to reactive oxygen species identifies genes important for intracellular

growth PLoS One; Submitted

Mestre O, Luo T, Dos Vultos T, Kremer K, Murray A, Namouchi A, Jackson C, Rauzier J, Bifani P,

Warren R, Rasolofo V, Mei J, Gao Q, Gicquel B. (2011). Phylogeny of Mycobacterium tuberculosis

Beijing strains constructed from polymorphisms in genes involved in DNA replication,

recombination and repair. PLoS One. 20;6(1):e16020.

The following manuscripts have also been published during the Ph. D. studies:

Dos Vultos, T., Mestre, O., Rauzier, J., Golec, M., Rastogi, N., Rasolofo, V., Tonjum, T., Sola, C.,

Matic, I., and Gicquel, B. (2008). Evolution and diversity of clonal bacteria: the paradigm of

Mycobacterium tuberculosis. PloS one 3, e1538.

Dos Vultos, T., Mestre, O., Tonjum, T., and Gicquel, B. (2009). DNA repair in Mycobacterium

tuberculosis revisited. FEMS microbiology reviews 33, 471-487.

Wang, C., Peyron, P., Mestre, O., Kaplan, G., van Soolingen, D., Gao, Q., Gicquel, B., and Neyrolles,

O. (2010). Innate immune response to Mycobacterium tuberculosis Beijing and other genotypes.

PloS one 5, e13594.

Page 16: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XIV

Page 17: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XV

TABLE OF CONTENTS

Abstract III

Resumo V

Acknowledgments IX

Abreviations XI

Publications XIII

Table of contents

XV

Chapter 1: General introduction

1

1.1 Tuberculosis na ancient disease 3

1.2 TB today’s major issues 3

1.3 The genus Mycobacterium 6

1.3.1 The Mycobacterium tuberculosis complex 7

1.3.2 Mycobacterial cell envelope 7

1.4 Molecular evolution of mycobacteria 9

1.4.1 Molecular genetic markers that define M. tuberculosis complex genetic

diversity

9

1.4.2 Phylogeny of the M. tuberculosis complex 12

1.4.3 The Beijing/W family 15

1.5 M. tuberculosis Infection 17

1.5.1 Recognition of M. tuberculosis and initiation of immune response 18

1.5.2 Granuloma formation 20

1.6 M. tuberculosis interactions with macrophages 22

1.6.1 M. tuberculosis evasion of macrophage’s antimicrobial mechanisms 23

1.6.1.1 Inhibition of phagosome maturation 24

1.6.2 Nitrosative and oxidative stress relevance in tuberculosis 26

1.6.3 M. tuberculosis responses against oxidative stress caused by ROS 27

1.6.3.1 The formation of ROS 27

1.6.3.2 ROS scavenging systems 29

1.6.3.3 Protein and lipid repair 31

1.6.3.4 Oxidative DNA damage caused by ROS 33

1.6.3.5 M. tuberculosis DNA repair 34

1.6.3.6 Oxidative stress response regulation 44

Objectives 47

Page 18: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XVI

Chapter 2: Identification of M. tuberculosis virulence factors involved in defense

mechanisms against oxidative stress

49

2.1 Abstract 51

2.2 Introduction 53

2.3 Materials and Methods 55

2.4 Results 61

2.4.1 Screening for mutants with increased susceptibility to oxidative stress 61

2.4.2 Sensitive mutants to oxidative stress are affected in cell envelope

functions

62

2.4.3 Quantification of sensitivity of mutants to oxidative stress 64

2.4.4 MmpL9 and MoaD1 are important for M. tuberculosis survival in human

macrophages

66

2.4.5 The mmpL9 deletion at position 218 is conserved in Beijing/W strains 67

2.4.6 MmpL9 and MoaD1 are required to counteract other forms of stress

existent in macrophages

69

2.5 Discussion 77

Chapter 3: Evolution and diversity of M. tuberculosis Beijing/W strains reflected by

polymorphisms in 3R genes

83

3.1 Abstract 85

3.2 Introduction 87

3.3 Materials and Methods 89

3.4 Results 93

3.4.1 Identification of polymorphisms in 3R genes in Beijing/W strains 93

3.4.2 Construction of a phylogeny for Beijing/W strains using 3R SNPs 96

3.5 Discussion 99

Chapter 4 : Concluding Remarks 103

References 111

Annex 133

Page 19: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XVII

FIGURES

Chapter 1

Figure 1 – Estimated MDR-TB cases among notified TB patients in 2010 4

Figure 2 – Mycobacterial cell envelope constitution 8

Figure 3 – Representation of M. tuberculosis genome of the H37Rv reference strain,

indicating the major genetic markers used for strain genotyping

10

Figure 4 – M. tuberculosis phylogeography 13

Figure 5 – M. tuberculosis infection and granuloma formation 21

Figure 6 – Assembly of the phagocyte NADPH oxidase NOX2 28

Figure 7 – Reactive oxygen species decomposed from superoxide produced by

phagocyte oxidase

29

Figure 8 – Cellular reactions leading to oxidative DNA damage via the Fenton

Reaction.

34

Figure 9 – Major mechanisms repairing DNA damages in bacteria 35

Figure 10 – Enzymatic systems that protect from the mutagenic effects of 8-

oxoguanine

36

Figure 11 – Early steps during the repair of double strand breaks (DSBs) 41

Chapter 2

Figure 1 – Growth curves of M. tuberculosis H37Rv, ΔmutT1, ΔmutM and ΔmutY in

increasing concentrations of hydrogen peroxide.

61

Figure 2 – Growth curves of M. tuberculosis GC1237 in increasing concentrations of

hydrogen peroxide.

62

Figure 3 – Susceptibility of transposon mutants to oxidative stress. 65

Figure 4 – Intracellular growth of mmpL9 and moaD1 mutants in human

macrophages.

67

Figure 5 – Graphical representation of the distribution of polymorphisms in mmpL9 in

several M. tuberculosis genomes.

69

Figure 6 – Survival of mmpL9 and moaD1 mutant and complemented strains exposed

to hydrogen peroxide.

70

Figure 7 – Macrophages treated with the NADPH oxidase inhibitors, apocynin and DPI. 71

Figure 8 – Intracellular growth of mmpL9 and moaD1 mutants in human macrophages

treated with Phox inhibitors.

72

Figure 9 – Macrophages activated with Vitamin D3. 73

Figure 10 – Intracellular growth of mmpL9 and moaD1 mutants in VitaminD3-

activated macrophages treated with Phox inhibitors.

74

Figure 11 – Susceptibility of mmpL9 and moaD1 mutants to acidic pH and starvation. 75

Figure 12 – Representation of the mmpL9 gene in M. tuberculosis GC1237 78

Page 20: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

XVIII

Chapter 3

Figure 1. Phylogenetic network based on SNPs discovered in the collection of 58

Beijing/W isolates.

95

Figure 2. Phylogenetic network based on SNPs characterized in the entire collection of

305 Beijing/W isolates.

97

TABLES

Chapter 2

Table 1. List of oligonucleotides (5’-3’) used in this study. 56

Table 2. List of identified sensitive mutants to oxidative stress using hydrogen

peroxide.

63

Table 3. Statistical analysis of SNP density in mmpL genes in a collection of M.

tuberculosis genomes.

68

Chapter 3

Table 1. List of oligonucleotides used for SNP analysis. 90

Table 2. List of primers used in Multiplex-PCR for Large Sequence Polymorphisms

(LSPs) analysis.

92

Table 3. List of SNPs identified by sequencing 3R genes in Beijing/W strains, which

define different sequence types (Figure 1 and 2).

94

Annex

Table A1. Description of M. tuberculosis Beijing/W strains belonging to each node

found in Figure 1 and 2, and respective country of isolation.

133

Page 21: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

CHAPTER 1

General Introduction

Page 22: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga
Page 23: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

3 Introduction

Tuberculosis an ancient disease 1.1

Tuberculosis (TB) is an ancient disease. Skeletal abnormalities, typical of TB, were found in

Egyptian mummies, dated of several thousand years ago, and DNA analyses have later confirmed

the presence of the disease-causing pathogen Mycobacterium tuberculosis. At this time TB was

probably sporadic, but, as population densities increased, the disease reached epidemic

proportions in Europe and North America in the 18th and 19th centuries (Daniel, 2006).

The development of a live attenuated vaccine, Bacillus Calmette-Guérin (BCG), by Albert

Calmette and Camille – Guérin in 1921, and the discovery of the first antibiotic against TB,

streptomycin, by Selman Waksman in 1943, led to the opinion that appropriate control measures

had become available for fighting TB. Between 1950 and 1965 TB-control programs were

established. However, these initiatives have had limited success in developing countries. In

industrialized countries there was a pronounced reduction of infection and death rates.

Consequently, TB programs became a lower priority because the disease was considered close to

elimination. In the early 1980s, cases of TB began to raise due to factors such as increases in prison

and homeless populations, intravenous drug use and especially Human Immunodeficiency

Virus/Acquired Immunodeficiency Syndrome (HIV/AIDS) epidemics. By 1993 increasing TB could no

longer be ignored so the World Health Organization (WHO) declared the disease a global

emergency (Daniel, 2006) (Harries and Dye, 2006). In 2000, the World Health assembly endorsed

the establishment of a Global Partnership to Stop TB with several priorities for the next 50 years

(WHO at www.who.int/topic/tuberculosis/en/).

TB today’s major issues 1.2

Despite the efforts to combat the disease, TB remains today a major public-health problem.

Every 15 seconds someone dies from TB, which means that about 1.5 million people die each year

from this treatable disease. The current TB epidemic is being sustained and fuelled by two

important factors: the HIV infection, and its association with active TB disease, and increasing

resistance of M. tuberculosis strains to anti-TB drugs (WHO at

www.who.int/topic/tuberculosis/en/).

Despite the availability of an effective antibiotherapy for decades, tuberculosis control has been

seriously challenged due to the appearance of drug-resistant strains. Drug resistance in M.

tuberculosis arises mainly from spontaneous mutations in the chromosome (Heym et al., 1994),

and not from mobile genetic elements such as plasmids or transposons, acquired by horizontal

Page 24: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

4 Chapter 1

gene transfer, as observed in other bacteria. Then, irregular drug supply, inappropriate prescription

and poor patient adherence to treatment allow selection of strains carrying drug-resistance

mutations. These strains can be transmitted and other mutations involved in resistance to

individual drugs can be accumulated resulting in the appearance of multidrug-resistant (MDR)

strains, defined as resistant to at least rifampin and isoniazid, the most effective first-line anti-TB

drugs (WHO at www.who.int/topic/tuberculosis/en/). The WHO estimates that there were 290 000

MDR-TB cases among notified TB cases in 2010 (Figure 1). The estimated number of prevalent

MDR-TB cases seems to reach 650 000 among the 12 million total prevalent cases of TB (WHO,

2011). The number of extensively-drug resistant strains (XDR), defined as MDR plus resistance to

any fluoroquinolone and to one of the three injectable second line drugs (capreomycin, kanamycin,

or amikacin), has been increasing every year, and, by the end of 2010, 77 countries had reported at

least one case of XDR TB to WHO (WHO at http://www.who.int/tb/challenges/mdr/en/index.html).

More recently, the appearance of totally drug-resistant tuberculosis (TDR), resistant to all

antituberculosis drugs, seriously threatens TB control (Velayati et al., 2009).

Figure 1 - Estimated MDR-TB cases among notified TB patients in 2010. From WHO at

http://www.who.int/tb/challenges/mdr/en/index.html.

More than one in ten TB cases occur among HIV-infected people, and almost one in four deaths

among people with HIV is due to TB. In 2010 350 000 people died of HIV-associated TB (WHO,

2011)(WHO at http://www.who.int/tb/publications/TBHIV_Facts_for_2011.pdf). In fact,

immunodeficiency caused by HIV infection greatly increases susceptibility to infection and the risk

of developing TB, which is 21-34 times higher in HIV co-infected individuals than in non-infected

No data

0-300

301-3,000

3,001-30,000

30,001-60,000

>60,000

Page 25: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

5 Introduction

(WHO, 2011). On the other hand, TB may accelerate the course of HIV infection, increasing the viral

load in some patients (Day et al., 2004; Whalen et al., 1995). In addition to contribute to an

increase in TB burden in general, HIV might also be contributing to the increase of MDR-TB

prevalence (Wells et al., 2007). Co-infection of TB in HIV patients further complicates treatment

because it has to be combined with antiretroviral therapy. This leads to an increased pill burden,

drug-drug interactions that may lead to suboptimal concentrations of anti-TB drugs, and finally

overlapping side effects (Swaminathan et al., 2010; Wells et al., 2007). Moreover, TB patients co-

infected with HIV are more likely to progress to disease, and thus potentially accelerate the

propagation and spread of these MDR strains (Gandhi et al., 2010). The intersection of drug-

resistance and HIV epidemics might represent a lethal combination and further threaten TB

control.

The development of an effective vaccine against TB could also have a significant impact in the

control of the disease. The only available vaccine today is BCG, a live vaccine derived from an

attenuated Mycobacterium bovis strain, obtained after serial passages (Calmette, 1931). It has

been used since 1921, yet, it has given little contribution to contain the current TB pandemic. It is

effective in protection against disseminated TB in children, like meningitis and miliary TB (Trunz et

al., 2006), but, it seems to have variable efficacy against adult pulmonary TB – the most prevalent

form of disease – being particularly limited in endemic areas (Colditz et al., 1994; Fine, 1995). TB

vaccine research has made huge progress and as a result twelve vaccine candidates have been

developed and entered clinical trials (Kaufmann, 2011). Nevertheless, we do not know with

certainty whether they will confer robust protection against TB in humans.

Finally, enormous progress has been made in the last decade resulting in the appearance of

much promising tools for diagnosing TB (Palomino, 2012). Still, no tests are available that are

universally applicable to all patients including HIV-coinfected people and children with TB, for

which diagnosis is usually more difficult. Additionally, an important gap in this field is the

development of tools to determine progression to disease in people infected with M. tuberculosis

without active disease (latent TB). This would be extremely important given that one-third of the

world’s population seem to be latently infected with M. tuberculosis (WHO at

www.who.int/topic/tuberculosis/en/).

Although the number of incident cases has decreased substantially during the last twenty-years,

Portugal has still an elevated rate of TB incidence, being the highest among western European

countries ((WHO, 2011) - Annex 3). There is also a high prevalence of MDR-TB and alarming rates

Page 26: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

6 Chapter 1

of XDR-TB, particularly in the Lisbon Health Region (Direcção Geral de Saúde (DGS) at

http://www.dgs.pt/upload/membro.id/ficheiros/i012626.pdf) (Perdigao et al., 2008).

The genus Mycobacterium 1.3

The first notion that TB could be an infectious disease was described by Benjamin Martens in

1790, followed by Jean-Antoine Villemin who demonstrated, in 1865, the transmissible and

infectious nature of TB in animals (Daniel, 2006). However, it was Robert Koch, in 1882, who first

described the tubercle bacillus, M. tuberculosis (Koch, 1882; Koch, 1982). Nevertheless, the

identification of mycobacteria, however, occurred earlier with the discovery of the leprosy bacillus

in 1873 (Hansen, 1874(Mange, 1992).

Mycobacteria are aerobic non-motile rod-shaped actinobacteria with a high G+C content in their

DNA (61-71%). These bacteria are characterized by an unusual cell envelope composed by mycolic

acids with long branched chains (60 to 90 carbons), which confer alcohol and acid-fast staining

properties (Levy-Frebault and Portaels, 1992; Shinnick and Good, 1994). Along with

Corynebacterium and Nocardia, they form a well-defined sub-group (CMN) of actinobacteria, which

share properties such as the production of mycolic acids (Embley and Stackebrandt, 1994; Ventura

et al., 2007). Other mycolic acid-containing genera include Gordonia, Rhodococcus, Tsukamurella

and Dietzia among others (Gurtler et al., 2004) being thus considered to be closely related to

mycobacteria.

The genus Mycobacterium is highly diverse and comprises more than 100 species (Dworkin et

al., 2006). It can be divided into two major groups’ – fast and slow growers, a traditional division

was found to be congruent by 16S rRNA analyses (Stahl and Urbance, 1990). This definition is based

on the time required for visible colonies to appear on a solid medium, after plating suspensions

diluted enough to give well separated colonies (Shinnick and Good, 1994):

I. Fast growers (colonies in less than seven days) – usually non-pathogenic mycobacteria

and free-living saprophytes that include M. smegmatis, commonly used as a non-

pathogenic model in the laboratory, to study essentially genetics or physiology of

mycobacteria (O'Toole, 2010). An exception would be M. abssecus, a fast grower that is

pathogenic.

II. Slow growers (colonies in more than seven days) – usually associated with human or

animal disease and include important pathogens such as M. leprae, M. ulcerans, and M.

tuberculosis complex members. There are, however, slow growers that are not

pathogenic, such as M. gordonae.

Page 27: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

7 Introduction

1.3.1 The Mycobacterium tuberculosis complex

The M. tuberculosis complex includes several closely related species, responsible for

tuberculosis, with different phenotypic properties and host preferences. M. tuberculosis and M.

africanum are almost exclusively human pathogens. M. microti causes disease in voles and M.

pinnipedii causes TB in seals and sea lions. M. caprae infects both goats and deer and, finally, M.

bovis has the largest host range although it can be mainly isolated from cattle (Brosch et al., 2002;

Mostowy et al., 2005; Smith et al., 2006). Some very rare isolates of tubercle bacilli, with the ability

to cause disease in humans, were found to belong to the same phylogenetic lineage as M.

tuberculosis complex members as shown by 16sRNA analyses (van Soolingen et al., 1997). These

bacteria, referred to as M. canetti, have shown unusual smooth colony morphology, in contrast to

the rough colonies produced by the other M. tuberculosis complex members.

1.3.2 Mycobacterial cell envelope

Mycobacteria possess a remarkably complex cell envelope consisting of three principal layers:

the cytoplasmic membrane, the cell wall core and an outer membrane (Figure 2). Cell walls are

constituted by a peptidoglycan – arabinogalactan polymer core with covalently bound mycolic

acids (the mycolyl-AG-peptidoglycan complex) that, along with non-covalently bound, extractable

lipids, form an outermost bilayer structure (Hoffmann et al., 2008; Zuber et al., 2008). Thus,

despite the classification of mycobacteria as Gram-positive bacteria it seems that the

mycobacterial cell envelope contains an outer membrane containing pore-forming proteins

(Niederweis et al., 2010), and, by inference, a periplasmic space (Hoffmann et al., 2008; Zuber et

al., 2008) (Figure 2). This complex structure represents a permeability barrier that contributes to

the intrinsic resistance of mycobacteria to antibiotics, dehydration and chemical damage (Brennan

and Nikaido, 1995; Nguyen and Thompson, 2006). An outer layer (also called capsule) surrounding

the outer membrane (Figure 2), mainly composed of polysaccharides, proteins and only minor

amounts of lipids (Ortalo-Magne et al., 1995), also seems to exist in various mycobacterial species

(Sani et al., 2010). Most of these compounds were found released in the culture fluids of in vitro-

grown bacteria but seem to coat intracellularly grown bacteria (Daffe and Draper, 1998).

Numerous extractable lipids are present in mycobacterial cell walls, being many of these

suspected to be present in the outermembrane. However, their exact arrangement and

distribution remains poorly understood. These include: phthiocerol dimycocerosate (PDIM), a

major lipid of the cell wall, important for virulence, architecture and permeability (Camacho et al.,

1999; Cox et al., 1999); phenolic glycolipids (PGLs), that share a common lipid core with PDIMs and

Page 28: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

8 Chapter 1

are also associated with virulence, but are produced by only a few groups of M. tuberculosis (Reed

et al., 2004; Tsenova et al., 2005); as well as glycolipids such as diacyltrehaloses (DAT),

triacyltrehaloses (TAT), sulfatides (SLs), and the mycolic acid derivatives trehalose monomycolate

(TMM) and trehalose dimycolate (TDM) (also known as cord factor) (Kaur et al., 2009).

Other important molecules in the mycobacterial cell envelope are mannosylated molecules such

as lipoarabinomannan (LAM), the related lipomannan (LM), and phosphatidyl-myo-inositol

mannosides (PIMs) (Kaur et al., 2009), that seem to be anchored not only to the outer membrane

but also to the cytoplasmic membrane (Pitarque et al., 2008) (Figure 2). In M. tuberculosis and

other pathogenic mycobacteria LAM is mannose-capped (ManLAM) (Brennan and Nikaido, 1995).

Figure 2 – Mycobacterial cell envelope constitution. LAM: lipoarabinomannan, PIMs: phosphatidyl-myo-

inositol mannosides, TDM: trehalose dimycolate, TMM: trehalose monomycolate. Adapted from Catalão et al.

(2012), with authors’ permission. (Catalao et al., 2012)

As most of these molecules are the first to contact with host cellular constituents, the

importance of these in facilitating host cell entry and modulating the host cell response has been

well documented (discussed in more detail in section 1.5.1).

A large proportion of the M. tuberculosis genome is devoted to fatty acid and lipid metabolism

(Cole et al., 1998). The colocalization of some mmpL (mycobacterial membrane protein Large)

genes with genes involved in polyketide biosynthesis (pks) and lipid metabolism suggested that

MmpLs are generally involved in lipid synthesis and/or transport (Tekaia et al., 1999). This was

found to be indeed the case for MmpL7 (Camacho et al., 2001; Cox et al., 1999) as well as MmpL8

(Converse et al., 2003; Domenech et al., 2004), involved in the transport of PDIM and SL-1,

Page 29: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

9 Introduction

respectively. However, certain MmpLs do not colocalize with genes involved in lipid synthesis (Cole

et al., 1998) and it seems that MmpL substrate specificity is dictated, in part, by their interaction

with particular cytosolic enzymes (Jain and Cox, 2005).

Molecular evolution of mycobacteria 1.4

It is important to investigate diversity of bacterial pathogens for both epidemiological, to

elucidate prevalence of strains, and biological reasons, because specific strains or groups of strains

(lineages/families) might have specific relevant phenotypes (e.g. differences in virulence or drug

susceptibility).

M. tuberculosis complex members are genetically extremely closely related when compared to

other bacteria, being thus considered as monomorphic (Achtman, 2008). Therefore, some of the

genotyping tools applied to other pathogens can be uninformative in M. tuberculosis (Achtman,

2008). For example, early attempts at using multilocus sequence typing (MLST) to study strain

diversity in M. tuberculosis complex have shown that these bacteria are characterized by more

than 99.9% similarity at the DNA level and identical 16S rRNA sequences (Sreevatsan et al., 1997).

Despite this apparent genetic homogeneity, M. tuberculosis complex members display diverse

phenotypic characteristics and host ranges and several polymorphic molecular genetic markers

(Figure 3) have proved to be useful to identify and discriminate M. tuberculosis complex isolates.

1.4.1 Molecular genetic markers that define M. tuberculosis complex genetic diversity

One of the most widely applied molecular methods to type M. tuberculosis strains has been the

IS6110 Restriction Fragment Length Polymorphism (RFLP) typing. Individual strains vary in the

number of copies and in the sites into which IS6110 is inserted. A method using a restriction

enzyme has been developed and IS6110 Southern blots of the digested DNA separated by

electrophoresis on agarose gels provide a RFLP pattern (van Embden et al., 1993). Unfortunately,

this method has some limitations and disadvantages such as poor discrimination between isolates

with low copy numbers (<5) of IS6110 and it is even absent from certain strains. In addition, it is a

technically demanding and expensive technique and requires high amounts of DNA (Kato-Maeda et

al., 2011). This has led to the development of other techniques, frequently based in amplification

of DNA, being spacer oligonucleotide typing (Spoligotyping) and variable number of tandem repeat

(VNTR) typing (Frothingham and Meeker-O'Connell, 1998) the most frequently used.

Page 30: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

10 Chapter 1

Figure 3 - Representation of M. tuberculosis genome of the H37Rv reference strain, indicating the major

genetic markers used for strain genotyping. As an example one specific mycobacterial interspersed repetitive

unit (MIRU) locus, insertion sequence (IS6110), large sequence polymorphism (region of difference - RD) and

single nucleotide polymorphism are shown. Adapted from Nicol et al. (2008). (Nicol and Wilkinson, 2008)

Spoligotyping involves PCR amplification of the DR locus, a single region of M. tuberculosis

complex strains chromosome that contains multiple copies of a 36-bp direct repeat, separated by a

DNA spacer with various sequences (Kamerbeek et al., 1997). Strains have the same overall

arrangement of spacers but differ in terms of presence or absence of specific spacers. Comparison

with the 43 spacers identified in M. tuberculosis H37Rv, the laboratory reference strain, and M.

bovis BCG, allows discrimination and classification of strains (Van Soolingen, 2001). Forty-one

VNTRs of the mycobacterial interspersed repetitive units (MIRUs) have been identified in M.

tuberculosis genome (Supply et al., 2000). PCR amplification of each locus will reflect the number

of copies of the repeat unit and is the basis of VNTR typing. Several number and combinations of

loci have been used to genotype M. tuberculosis strains which might have similar or higher

discriminatory power than IS6110-RFLP and may also vary according to certain characteristics of

the sample that is being analyzed, such as geographic origin. Thus, the set of loci might have to be

adapted to the type of collection of isolates to analyze. Nevertheless, MIRU-VNTR is considered by

some authors as the gold standard for M. tuberculosis genotyping (Kato-Maeda et al., 2011). These

methods are, however, not reliable for evolutionary and phylogenetic studies (Comas et al., 2009)

because intragenomic recombination events could occur independently at the level of repetitive

sequences (Achtman, 2008).

Recently, systematic comparative genomic analyses have identified Single Nucleotide

Polymorphisms (SNPs) and Large Sequence Polymorphisms (LSPs) as robust markers that generate

Page 31: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

11 Introduction

portable and comparable data for studying population variation. This kind of markers have been

especially used to infer phylogenetic relations in M. tuberculosis (Baker et al., 2004; Dos Vultos et

al., 2008; Filliol et al., 2002; Fleischmann et al., 2002; Gagneux et al., 2006a; Gutacker et al., 2006;

Gutacker et al., 2002; Hershberg et al., 2008). LSPs correspond to genomic deletions that have

been suggested as an important mechanism for genetic variation in M. tuberculosis. Affected

sequences might have functional relevance in pathogenesis and immunity since deleted sequences

can include putative open reading frames as well as intergenic regions and housekeeping genes

(Alland et al., 2007). LSPs may be informative markers for discrimination of strains and have been,

therefore, proposed for genotyping and, as these are irreversible mutations, have also been used

for constructing phylogenies and to investigate M. tuberculosis complex evolution (Alland et al.,

2007; Brosch et al., 2002; Gagneux et al., 2006a; Tsolaki et al., 2005). SNPs also contribute to strain

variability and may have a functional effect. For example, drug-resistance is mainly caused by SNPs

in M. tuberculosis (Zhang and Yew, 2009). SNPs have also proved to be useful markers to

differentiate strains and to study the phylogenetic relatedness of M. tuberculosis strains (Dos

Vultos et al., 2008; Hershberg et al., 2008). A high number of such polymorphisms can be identified

by performing whole-genome sequencing which can be extremely informative, particularly for

genetically monomorphic bacteria like M. tuberculosis (Achtman, 2008).

Choosing the appropriate genetic marker will depend on the purpose of the study. For

epidemiologic analyses it is important to have markers with a high discriminatory power that

change relatively rapidly, but, are stable enough to make connections between related isolates. For

example, recognition of the regional specificity of circulating M. tuberculosis strains in Portugal

dates back to 1999 when early RFLP-IS6110 genotyping studies of an MDR cluster in the Lisbon

Health Region identified a prominent family, designated family Lisboa (Portugal et al., 1999). In

subsequent years RFLP-IS6110 and 12 loci MIRU–VNTR genotyping identified clusters of circulating

strains with similar genotypic characteristics (Perdigao et al., 2008; Perdigao et al., 2011), several of

which, including the Lisboa family, were associated with XDR–TB. The results show that active

transmission of MDR- and XDR-TB is taking place and that the high prevalence of observed XDR-TB

is due to the continued transmission of particular genetic clusters such as the Lisboa family,

identified in 1999 as strongly associated with MDR-TB (Portugal et al., 1999) and now associated

with XDR-TB (Perdigao et al., 2008; Perdigao et al., 2011).

To define phylogenetic associations unambiguously, genetic markers, need to be unique and,

ideally, irreversible. Robust phylogenetic markers will exhibit low rates of convergent evolution

Page 32: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

12 Chapter 1

(Kato-Maeda et al., 2011) providing solid phylogenetic frameworks in which specific strains can be

positioned and biologically meaningful groups defined.

1.4.2 Phylogeny of the M. tuberculosis complex

Although many molecular methods, such as the referred in the previous section, have been

used to categorize M. tuberculosis complex strains, the phylogenetic relationships among this

group have only recently started to be detailed. Analyses of genomic deletions (LSPs) have shown

that the animal-adapted organisms of the M. tuberculosis complex are part of the same

evolutionary lineage, defined by deletion of region of difference (RD) 9 (Brosch et al., 2002).

Furthermore, M. bovis, which was initially thought to cause disease in a variety of animals, can be

divided in different subspecies characterized by additional specific deletions, which include M.

caprae, M. microti and M. pinnipedii (Brosch et al., 2002; Mostowy et al., 2005). It was suggested

that the M. tuberculosis complex consists of distinct and genetically defined groups with specific

host-adapted groups called ecotypes (Smith et al., 2006). M. africanum shares the RD9 deletion

with these animal-adapted members of the M. tuberculosis complex which has led to the

speculation that it is not primarily adapted to humans but has an animal reservoir (Smith et al.,

2006). However, this remains to be confirmed and meanwhile, given that M. africanum clearly

infects and transmits among humans being responsible for up to 40% of human TB in west Africa

(de Jong et al., 2006; Kallenius et al., 1999), it is considered as human-adapted. The same analysis

of genomic regions also showed that M. canetti had almost all the RD regions intact (Brosch et al.,

2002). Although these strains share the same 16S rRNA sequence with M. tuberculosis complex

members as referred previously, they present high levels of nucleotide variation in housekeeping

genes and clear evidence of horizontal gene transfer (Gutierrez et al., 2005). This suggests that M.

canetti strains are relatives of a highly diverse progenitor of the M. tuberculosis complex, M.

prototuberculosis, that might have underwent an extreme bottleneck in a relatively recent past.

Consequently, much of the original diversity disappeared leaving only M. canetti strains among the

M. tuberculosis complex members (Gutierrez et al., 2005).

Other studies have defined the phylogeny of M. tuberculosis and M. africanum, the human-

adapted species of the M. tuberculosis complex. Gagneux et al. (2006) also used LSPs to describe

six major lineages, two of which included M. africanum. Each lineage seems to be associated with

particular human populations (Figure 4). Several studies have used SNPs for the same purpose.

Baker et al. (2004) analysed polymorphisms in seven housekeeping genes identifying 36

synonymous SNPs (sSNPs) that allowed discrimination of 4 main lineages in M. tuberculosis. These

Page 33: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

13 Introduction

four main lineages correspond to the same identified after by Gagneux et al. (2006) using LSPs

(Figure 4).

Figure 4 – M. tuberculosis phylogeography. Current major lineage assignments (lineage 1 to 6) are indicated

by coloured lines. (a) Tree originated from the study of Baker et al. (2004) based on 36 synonymous SNPs

identified by sequencing seven housekeeping genes in a collection of 225 strains. Roman designations correspond

to lineage assignments in the study. (b) Tree based on large sequence polymorphisms (LSPs) discovered by

Gagneux et al. (2006). (c) Association of M. tuberculosis lineages with specific geographic regions according to

data from Gagneux et al. (2006) and Filliol et al. (2006). Adapted from Achtman et al. (2008). (Filliol et al., 2006;

Gutacker et al., 2006)

Gutacker et al. (2006) have screened 5069 isolates for 36 sSNPs that have been identified by

comparison of two M. tuberculosis genomes and defined nine main lineages. Three of these

lineages corresponded to the ones described by Gagneux et al. (2006) and Baker et al. (2004). Filliol

et al. (2006) have used a similar approach to identify 159 sSNPs and screened 219 isolates of M.

tuberculosis and M. bovis. Ten major groups were identified, one of them being specific to M. bovis

and three corresponding to the same previously described (Baker et al., 2004; Gagneux et al.,

2006a). These studies have identified thus, four main M. tuberculosis lineages and two M.

africanum lineages, associated with particular geographic locations (Figure 4).

Page 34: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

14 Chapter 1

The fact that both LSP and SNP-based analyses of large strain collections resulted in single trees

without homoplasy indicated that horizontal gene transfer is extremely rare in M. tuberculosis

complex and it has a highly clonal population structure (Supply et al., 2003).

Although these studies have provided a picture of M. tuberculosis and M. africanum evolution,

some limitations didn’t allow a better elucidation. The number of SNPs was either too limited

(Baker et al., 2004) or resulted from comparison of a limited number of genomes which can lead to

branch collapsing (Filliol et al., 2006; Gutacker et al., 2006) and biased phylogenies. As for the

phylogeny inferred from LSPs (Gagneux et al., 2006a), genetic distances based on genomic

deletions are difficult to interpret. In conclusion, genotyping approaches that rely on LSPs or a

limited numbers of sSNPs can define deep phylogenetic population structures, but they are still

characterized by low discriminatory power.

More recently, two studies have contributed to a better knowledge of M. tuberculosis evolution

by overcoming the limitations of the previous studies (Dos Vultos et al., 2008; Hershberg et al.,

2008), providing a more detailed picture of M. tuberculosis phylogeny. These studies reveal that

the human-adapted members of M. tuberculosis complex are much more genetically diverse than

generally recognized and some of this genetic diversity is likely to have functional consequences

(Hershberg et al., 2008). The genetic diversity found among the human-adapted members of M.

tuberculosis complex was just as pronounced as that among animal-adapted strains, even though

the latter are adapted to distinct animal host species (Hershberg et al., 2008). In the study of

Vultos et al. (2008) a specific group of genes was analysed: genes involved in DNA repair,

recombination and replication (3R) genes. In M. tuberculosis the main source of genetic variation

should arise from random mutations, although it was recently suggested that recent recombination

might also occur in M. tuberculosis (Namouchi et al., 2012). Defective 3R systems can potentially

increase genomic variability due to higher mutation rates (Denamur and Matic, 2006; Tonjum and

Seeberg, 2001). Thus, polymorphisms in 3R genes seems to be an important mechanism in

evolution and adaptation of microorganisms (Taddei et al., 1997; Tonjum and Seeberg, 2001) as

strains with higher mutation rates (mutators) may, under certain conditions, have a selective

advantage. Analysis of polymorphisms in such genes in a collection of strains representative of

global M. tuberculosis complex diversity has identified high levels of polymorphisms, suggesting

that these genes may have a role in M. tuberculosis complex evolution (Dos Vultos et al., 2008). In

2010, Comas et al. (2010) used 21 whole genome sequences to generate the first whole genome –

based phylogeny of human-adapted M. tuberculosis, and confirmed the existence of six main

phylogenetic lineages. More recently whole genome analysis of M. tuberculosis complex strains

Page 35: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

15 Introduction

from major lineages has also confirmed previous phylogenetic analysis using finished and partial

genome data (Namouchi et al., 2012). It was also shown that purifying selection is acting on M.

tuberculosis genetic diversity (Namouchi et al., 2012), in contrast to what has been previously

suggested (Hershberg et al., 2008).(Comas et al., 2010)

There is increasing evidence that the observed M. tuberculosis strain variation is biologically

significant and that specific genotypes are associated with specific phenotypes. Indeed, different

M. tuberculosis genotypes show differences in terms of virulence and pathogenesis in animal

models (Lopez et al., 2003; Palanisamy et al., 2009) as well as survival and transcriptional profiles

within macrophages (Homolka et al., 2010). Although this genetic diversity seems to influence

clinical presentation of the disease in humans (Caws et al., 2008; Thwaites et al., 2008) this is still

not so clear (Coscolla and Gagneux, 2010) as several factors have a role in the development of TB.

One of the most studied M. tuberculosis genotypes due to its association with increased virulence,

as well as other characteristics that suggest that these strains have a selective advantage and are

more adapted than strains from other genotypes, is the Beijing/W family (East-Asian lineage

according to Gagneux et al. (2006)).

1.4.3 The Beijing/W family

The Beijing family was first described in 1995. A large proportion of M. tuberculosis strains in

the Beijing area of China had highly similar multi-banded IS6110 RFLP patterns and identical

spoligotyping patterns (van Soolingen et al., 1995). In the second half of the 1990’s the multidrug

resistant M. tuberculosis strain identified in New York, “W” strain (Bifani et al., 1996), was

recognized as a member of the Beijing genotype family which became the Beijing/W family (Bifani

et al., 2002; Kurepina et al., 1998). Since then, the Beijing/W genotype was found to be the most

frequently observed genotype of M. tuberculosis to cause disease globally (Glynn et al., 2002).

According to the fourth international spoligotyping database, strains from this genotype were

found to be present in the largest number of countries globally and represented 13% of global

isolates (Brudey et al., 2006). In some regions including Russia, Bangladesh, South Africa, and

Western Europe, the incidence of Beijing/W strains seems even to be increasing (Tuberculosis,

2006). This contrasts to what seems to happen with other genotypes that appear to be

predominantly restricted to defined geographical regions, and suggests that Beijing/W strains

might have some selective advantage that have allowed their global spread.

When compared to other genotypes, Beijing/W strains were found to grow significantly faster in

human macrophages (Li et al., 2002; Theus et al., 2004) and have shown increased virulence in

Page 36: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

16 Chapter 1

different animal models of infection (Lopez et al., 2003; Tsenova et al., 2005). However, it is not

possible to conclusively demonstrate an association between the Beijing/W strain genotype and

clinical presentation in humans (Coscolla and Gagneux, 2010; Parwati et al., 2010). In addition,

Beijing/W strains are often associated with drug-resistance (Tuberculosis, 2006).

Beijing/W strains share several genetic characteristics (Bifani et al., 2002) which differentiate

them from other all M. tuberculosis genotypes, including: a spoligotyping pattern containing at

least three of the nine spacers, 35 to 43, with an absence of hybridization to spacers 1 to 34, which

classically define and identify Beijing/W strains (Kremer et al., 2004); similar IS6110 RFLP patterns

(15-26 copies); IS6110 insertion between dnaA and dnaN genes encoding DNA replication proteins

and insertion of variable numbers of IS6110 in the noise transfer function (NTF) chromosomal

region (Kurepina et al., 1998) that define typical/modern (one or several IS6110 insertions) and

atypical/ancient (no IS6110 insertions) Beijing/W strains (Mokrousov et al., 2006). VNTR typing has

been the most frequently used marker to discriminate Beijing/W strains. However, special

consideration is given to VNTR typing of Beijing/W isolates once the standardized set of 15 or 24

loci used to type M. tuberculosis strains are not always sufficient to discriminate among Beijing/W

isolates (Iwamoto et al., 2007). In addition VNTR loci might have different discriminatory powers

among Beijing/W strains depending on strain collection characteristics such as geographical origin

(Kremer et al., 2005).

By comparative whole-genome hybridization of Beijing/W strains Tsolaki et al. (2005) have

shown that there are LSPs associated with this family. These correspond to genomic regions

variably deleted in Beijing/W strains in relation to M. tuberculosis H37Rv (RDs). RD105, RD181,

RD150, RD142, were found to, not only define but also subdivide this family into four different

subgroups (Tsolaki et al., 2005) that might be associated with different phenotypes. Hanekom et al.

(2007) have shown that the Beijing/W family can be divided into seven different lineages and that,

by analysis of epidemiological data, differences between these lineages in their ability to spread

and cause disease might exist. Thus, even closely related strains from the same lineage can

accumulate genomic diversity, which can lead to different pathogenic properties. Indeed, an earlier

study has shown that not all Beijing/W strains are hypervirulent in mice (Dormans et al., 2004). In

subsequent studies this difference among Beijing/W strains was shown in different animal models

(Aguilar et al., 2010; Palanisamy et al., 2009) as well as in human macrophages (Theus et al., 2007).

A very recent study has shown, in guinea pigs, that the LSP-defined Beijing/W sublineages have

indeed differences in pathogenicity (Kato-Maeda et al., 2012).

Page 37: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

17 Introduction

Several studies have focused on the possible mechanisms underlying the success of Beijing/W

strains. However, the results reflect, most of the times, this genetic variability observed among

Beijing/W strains. For example, the increased virulence observed for Beijing/W strains was initially

associated with the production of a polyketide synthase derived PGL (Tsenova et al., 2005).

However, it was shown later that PGL could be a virulence factor only for a fraction of Beijing/W

strains (Reed et al., 2007), not explaining the success of the entire family. It was shown though that

all Beijing/W strains tested, representing all LSPs-defined Beijing/W sublineages (Tsolaki et al.,

2005), accumulated large quantities of another kind of fatty acids, triacylglycerides associated with

constitutive overexpression of the dormancy regulon (DosR)(Reed et al., 2007), a transcriptional

program that is utilized by the bacterium to adapt for survival during periods of nonreplicating

persistence in vitro and it has been proposed to be associated with survival of the bacillus during

latent TB infection (Voskuil et al., 2003). Some authors believed that BCG vaccination is a risk factor

for developing TB in populations infected with Beijing/W strains and might even be a selective

force favoring the spread of Beijing/W strains (Abebe and Bjune, 2006). One epidemiological study

suggested that this association can be found in certain specific group of Beijing/W strains,

however, most epidemiological studies did not find a higher proportion of Beijing/W strains among

BCG-vaccinated individuals (Kremer et al., 2009).

Even though many studies have focused on this subject, the mechanisms responsible for the

apparent global success of Beijing/W strains remain unclear. Therefore, it is important to continue

to investigate genetic diversity of M. tuberculosis strains even among strains from the same

subgroup/family, because these might be responsible for the phenotypic variability observed, and

could explain the emergence of specific groups of strains.

M. tuberculosis infection 1.5

M. tuberculosis infects approximately one third of the world’s population. However, only 5 to

10% of these infected individuals will develop symptomatic disease at some point in their lives

(WHO at www.who.int/topic/tuberculosis/en/). In most cases (90-95%) the pathogen is contained,

but not eliminated, by the immune response mounted by the host (Kaufmann and McMichael,

2005). This is called latent infection – when a person is infected but is asymptomatic.

The mechanisms that determine progression to disease are not well understood but seem to be

determined by environmental and socio-economic factors (e.g. exposure to environmental

Page 38: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

18 Chapter 1

mycobacteria, overcrowding), host factors (e.g. malnutrition, genetic predisposition) as well as

mycobacterial factors (e.g. genetic variation of mycobacterium) (Comas and Gagneux, 2009).

Pulmonary infection due to the interaction of the bacillus with host immune cells results in the

formation of a granuloma (Flynn et al., 2011), the typical pathologic hallmark of TB. This structure

provides the conditions for the bacillus to grow, and the immunologic environment, in which cells

with antimycobacterial functions interact to prevent and control dissemination of infection.

However, this structure does not always avoid progression into disease. Therefore, understanding

the molecular mechanisms occurring inside granulomas, that are likely to contribute to host

protection as well as disease progression, is essential to better understand TB infection.

1.5.1 Recognition of M. tuberculosis and initiation of immune response

Infection starts after inhalation of aerosols containing small numbers of bacilli that have been

expulsed to the air by an infected individual. Once, bacteria reach the lung alveoli these are taken

up by phagocytes, primarily resident alveolar macrophages, although dendritic cells (DCs) can also

be infected (Kaufmann and McMichael, 2005) (Figure 5). Interaction with these innate immune

cells is the first line of host defense against pathogens. M. tuberculosis initial interactions with host

cells are greatly mediated by pattern recognition receptors (PPRs), such as Toll-like receptors (TLRs)

and C-type lectins. These receptors recognize microbial determinants and bridge innate and

adaptive immune responses by regulating pro- and anti-inflammatory cytokine production as well

as endosomal and phagosomal traffic.

TLRs play a critical role in the recognition of several pathogens (Akira et al., 2006). In M.

tuberculosis one of the most important TLRs identified so far is TLR2 (Ferwerda et al., 2005) that

recognizes mycobacterial structures expressed at the surface of the bacteria such as LAM and PIM,

as well as the 19 kDa lipoprotein (Ryffel et al., 2005). The role of TLRs in controlling M. tuberculosis

in mice remains, however, controversial (Drennan et al., 2004; Reiling et al., 2002; Sugawara et al.,

2003). Nonetheless, polymorphisms in TLR2 have been associated with increased susceptibility to

TB in humans, suggesting that these receptors do have an important role in human host defense

against mycobacteria (Velez et al., 2010).

TLRs signals result in the activation of nuclear transcription factor (NF)-kB and, consequently,

the transcription of genes producing pro-inflammatory cytokines, chemokines, as well as nitric

oxide (Ryffel et al., 2005). TLR2 was also found to be essential for M. tuberculosis-induced reactive

oxygen species (ROS) production (Yang et al., 2009). In addition, TLR-dependent ROS were found to

be important signaling molecules and priming factors in the upregulation of early inflammatory

responses during TB Infection (Lee et al., 2009a; Shin et al., 2008; Yang et al., 2008). However, it

Page 39: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

19 Introduction

has been shown that either expression of early secreted antigenic target protein 6 (ESAT-6), a

secreted antigen of M. tuberculosis associated with lower innate immune responses to infection, or

a peptide derived from it, is able to attenuate TLR signalling (Pathak et al., 2007). TLRs might have

thus a dual role during M. tuberculosis infection.

TLRs are strongly implicated in the induction of cytokines and chemokines, but they are not

strongly linked to phagocytosis (Underhill, 2007). Phagocytosis of M. tuberculosis by human

macrophages occurs primarily via the mannose receptor (MR) (Schlesinger, 1993), a C-type lectin,

expressed in high levels by alveolar macrophages (Stephenson and Shepherd, 1987). M.

tuberculosis ManLAM recognition through these receptors seems to inhibit production of the pro-

inflammatory cytokine IL-12 (Nigou et al., 2001). Furthermore, it was shown that M. tuberculosis

association with these receptors are also implicated in inhibition of phagosome fusion with

lysosomes in primary human macrophages (Kang et al., 2005; Torrelles et al., 2006), a key

mechanism for M. tuberculosis evasion of host immune responses (discussed in more detail in

section 1.6.1). Therefore, M. tuberculosis interaction with MRs inhibits the phagocytic response

and induces and anti-inflammatory response, favouring bacteria’s infection. Phagocytosis of

pathogens can also be mediated by opsonins, host factors such as immunoglobulin that attach to

the surface of the pathogen, acquiring a conformation that is recognized by phagocytic receptors.

Fcγ receptors that bind to the Fc portion of immunoglobulins, are responsible for opsonized

phagocytosis of mycobacteria leading to phagolysosomal fusion (Armstrong and Hart, 1975). Until

recently there were not studies about the relevance of such receptors during infection with M.

tuberculosis in vivo probably due to the prevailing view that humoral immunity has little or no role

in defence against this pathogen. Maglione et al. (2008) have shown that this kind of receptors are

important for M. tuberculosis recognition and host defence, as a mice lacking this receptor had

enhanced susceptibility and immunopathology to infection, associated with increased production

of immunosuppressive cytokines. (Maglione et al., 2008)

Another C-type lectin described as important to bind M. tuberculosis in dendritic cells is DC-

SIGN (dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin) that also

recognizes ManLAM and other ligands in mycobacterial cell envelope (Tailleux et al., 2003).

Interestingly, DC-SIGN is also expressed by alveolar macrophages in tuberculosis patients (Tailleux

et al., 2005) suggesting an important role for this molecule during TB in humans. Even though

different studies about SNPs in the promoter region of DC-SIGN gene CD209 have shown

contradictory results (Barreiro et al., 2006; Vannberg et al., 2008), a role for DC-SIGN in protection

against infection by M. tuberculosis seems to be the case. Mycobacteria recognized by SIGNR3, a

Page 40: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

20 Chapter 1

DC-SIGN homologue in mice, expressing phagocytes induced key inflammatory cytokines, including

TNF-α (Tanne et al., 2009) as well as phagolysosome fusion in contrast to MR receptors also

recognizing ManLAM (Kang et al., 2005). However, a previous study has suggested that binding of

mycobacterial components to human DCs DC-SIGN leads to enhanced release of the anti-

inflammatory cytokine IL-10 and impair DC maturation through ManLAM in vitro (Geijtenbeek et

al., 2003). Thus, the role of DC-SIGN in TB pathogenesis remains to be further investigated.

The precise receptor involved in M. tuberculosis recognition may have a major impact on the

survival chances of this pathogen once inside macrophages. Recognition of particles through TLRs

results in an inflammatory response and increased respiratory burst while internalization via MRs

prevents this kind of responses which occur in activated macrophages. Salmonella typhi appears to

survive inside phagosomes by entering a receptor-mediated pathway not coupled with

macrophage activation (Ishibashi and Arai, 1990), however, for mycobacteria, it is not clear if there

is selective receptor-mediated intracellular survival as pathogenic strategy. It seems more likely

that different entry routes result in different cytokine production and traffic fates leading to

differential survival of M. tuberculosis.

1.5.2 Granuloma formation

The interaction of M. tuberculosis with phagocytes results in the production of pro-

inflammatory cytokines, such as TNFα and IL-12, as well as chemokines. These molecules will drive

the recruitment of monocytes, neutrophils and B cells. Effector T cells that have been, in the

meantime, primed against mycobacterial antigens by DCs, in the draining lymph nodes, will also be

recruited to the infection sites (Figure 5). These cells produce their own cytokines and chemokines,

such as IFNγ, that will activate and enhance the antibacterial activity of macrophages, and amplify

cellular recruitment (Flynn et al., 2011; Kaufmann and McMichael, 2005; Russell, 2007). However,

an intense inflammatory response like this has propensity to tissue damage. Therefore, anti-

inflammatory cytokines like IL-10, IL-27 and TGF-β are also produced and downregulate pro-

inflammatory cytokines, as well as T cell proliferation and activation, balancing the response

between bacterial eradication and host survival and culminating in the formation of a granuloma

(Flynn et al., 2011; Russell, 2007). The granuloma contains, but does not eliminate the infection, as

some resistant bacilli, capable of surviving under stressful conditions generated by the host, escape

killing.

Page 41: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

21 Introduction

Figure 5 – M. tuberculosis infection and granuloma formation. (a) Mycobacterium tuberculosis infection starts

with inhalation of bacilli that reach the lung alveoli. (b) In the alveolus, the main host cells are alveolar

macrophages. However, bacteria can also be taken up by dendritic cells (DCs), which carry M. tuberculosis either

to the lung parenchyma, leading to initiation of the local inflammatory response, or to the draining thoracic lymph

nodes. (c) In draining lymph nodes, mycobacterial antigenic peptides are presented to the specific naïve T cells

through cross-presentation. Activated T cells proliferate, become effector T cells and leave the lymph node. (d)

Effector T cells reach the site of inflammation under the influence of chemokines and other mediators.

Extravasations of the mononuclear cells initiate the formation of a granuloma at the infection site. The classic TB

granuloma is composed of a central core of infected macrophages surrounded by epithelioid and foamy

macrophages and a peripheral rim of lymphocytes (B cells, CD4+ T cells and CD8

+ T cells) in association with a

fibrous cuff of extracellular matrix laid by fibroblasts. Adapted from Griffiths et al. (2010). (Griffiths et al., 2010)

In an early stage the granuloma is an organized, stratified, and vascularized structure composed

of mainly macrophages in the center that are surrounded by a mantle of lymphocytes enclosed

within a fibrous cuff that marks the periphery of the granuloma (Figure 5). As this structure

matures, it develops an extensive fibrous capsule, gets less vascularized and, in later stages, starts

Page 42: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

22 Chapter 1

to become necrotic in the center of the structure (Russell, 2007). Ultimately, the granuloma

caseates, ruptures and spills infectious bacilli into the airways, resulting in active pulmonary

disease (Russell, 2007). In chronic or latent TB, granulomas can become calcified and contain the

pathogen (Russell, 2007; Ulrichs and Kaufmann, 2006) for many decades. Although the precise

state of M. tuberculosis in these latent granulomas is not clear it seems that it exists mainly in a

dormant non-replicative state (Ulrichs and Kaufmann, 2006), in a hypoxic environment and with

nutrient deprivation. The dosR regulon was found to play a critical role in preparing M.

tuberculosis for this hypoxic environment and an essential step towards dormancy, involving

induction of several genes expression (Schnappinger et al., 2003; Voskuil et al., 2003). These

dormant non-replicating bacteria may resuscitate into an active and replicative state. However,

how this determines transformation into active disease remains to be determined.

Macrophages are the primary cellular components of the granuloma and the initiating cells for

the formation of the granuloma (Flynn et al., 2011). These are the cells that most frequently

interact with M. tuberculosis as well as with the other cells within the granuloma. Thus,

understanding the interactions between M. tuberculosis and the macrophage is essential.

M. tuberculosis interactions with macrophages 1.6

M. tuberculosis is extremely adapted to host macrophages and it has developed several evasion

mechanisms to allow its survival inside these cells. Optimal mycobacterial growth during infection

seems to be even dependent on the entry into macrophages. M. tuberculosis-infected mice

depleted of macrophages had decreased bacterial loads and increased survival (Leemans et al.,

2001). On the other hand, macrophages are important to contain infection. Activated macrophages

seem to be important for early infection control, when only innate immunity is operant (Clay et al.,

2007) and to contribute to mycobacterial growth restriction in infected mice (Leemans et al.,

2005). Infections using M. marinum and embryonic zebrafish model in the absence of macrophages

led rapidly to high bacterial burdens (Clay et al., 2007) contradicting the previous results on mice

(Leemans et al., 2001). Thus, macrophages seem to play a dual role during infection: they provide

the preferred hiding and replication site of M. tuberculosis and protection from extracellular host

defense mechanisms, but display, at the same time, antimicrobial functions.

Page 43: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

23 Introduction

1.6.1 M. tuberculosis evasion of macrophage’s antimicrobial mechanisms

Macrophages are professional phagocytes extremely qualified to internalize and eliminate most

invading organisms (Flannagan et al., 2009). After phagocytosis, the phagosome fuses with early

endosomes, late endosomes and lysosomes (Desjardins et al., 1994), culminating in the formation

of phagolysosomes. During the course of maturation, phagosome becomes a highly acidic,

oxidative and degradative environment with microbicidal substances. However, M. tuberculosis has

the ability to subsist inside these cells. Several studies have been done in which bacterial, as well as

host transcriptome, have been analyzed to have a better idea of the intracellular/phagosomal

environment as well as host-pathogen interactions. One of the first studies (Schnappinger et al.,

2003) has confirmed that M. tuberculosis inhabits, in mouse macrophages, a nitrosative, oxidative,

hypoxic and nutritional limited environment. It has also shown that M. tuberculosis seems to shift

to lipid metabolism, as carbon and energy source, given the limitation of nutrients, as it was

previously suggested (McKinney et al., 2000). Many of these responses seem to be different from

those described for other intracellular pathogens like E. coli and Salmonella enterica (Schnappinger

et al., 2003). Another transcriptomic analysis made using human macrophages has shown similar

results to the ones obtained by Schnappinger et al. (2003) and reveals the differences between

macrophages and DCs in response to infection with M. tuberculosis (Tailleux et al., 2008). One of

such differences was in production of superoxide, found to be much more pronounced in

macrophages than in DCs. More recently, a core intracellular transcriptome was established

through the examination of different strains representing the global phylogeographic diversity

of M. tuberculosis (Homolka et al., 2010). It was shown that genetic diversity resulted in functional

diversity, reflecting the power that genetic variation exerts on gene expression, and, consequently,

on host interactions. Among the important genes, for M. tuberculosis intracellular lifestyle, shared

by all strains there were genes involved in hypoxia, radical stress, cell wall remodeling and lipid

metabolism. All these studies represent, however, M. tuberculosis infection of macrophages in

culture which might not reflect all the environments that the bacteria face in the human host.

Interestingly, a study has analyzed the transcriptome of M. tuberculosis from clinical lung samples

(Rachman et al., 2006). Overall, this study identified the same class of genes previously identified in

the in vitro transcriptomic studies referred above. The major difference was that a higher

proportion of genes was identified reflecting a more accurate representation of M. tuberculosis

transcriptomic profile during natural infection.

M. tuberculosis has developed several strategies to evade mechanisms of antimicrobial

destruction by macrophages allowing its survival inside these cells. One of the most studied

Page 44: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

24 Chapter 1

mechanisms used by M. tuberculosis to survive inside macrophages is the inhibition of normal

phagosome maturation.

1.6.1.1 Inhibition of phagosome maturation

ManLAM recognition through MRs seem to be implicated in inhibition of phagosome fusion

with lysosomes (Astarie-Dequeker et al., 1999; Kang et al., 2005), as previously referred. This

component of M. tuberculosis cell envelope interferes with the recruitment of early endosome

antigen 1 (EEA1), required for fusion of early phagosomes with late endosomal and lysosome

compartments (Flannagan et al., 2009). This seems to be mediated by interference with calcium

flux in the macrophage cytoplasm (Malik et al., 2003) that will also prevent accumulation in the

phagosomal membrane of phosphatidylinositol-3-phosphate (PI3P) (Vergne et al., 2003), a docking

site for several effector proteins involved in fusion of phagosomes and lysosomes, including EEA1

(Flannagan et al., 2009).

Besides cell wall components, secreted proteins have also been implicated in the modulation of

M. tuberculosis phagosome, even though how these are released from bacterial cells to the

macrophage cytosol remains an intriguing question. SapM is a mycobacterial phosphatase that

specifically hydrolyses PI3P, consequently reducing its concentration (Vergne et al., 2005). Thus, M.

tuberculosis seems to possess different mechanisms that target the same protein (PI3P) in order to

arrest early endosome maturation. PtpA is a low-molecular-weight tyrosine mycobacterial

phosphatase that seems to interfere with human Vps33B (vacuolar protein sorting 33B) (Bach et

al., 2008), a host cytosolic protein that is part of a complex that regulates vesicle docking and

fusion (Flannagan et al., 2009). PtpA acts probably by dephosphorylation of Vps33B, therein,

interfering with VPS33B association with small Rab GTPases, like Rab7, required for proper

membrane fusion (Chao et al., 2010; Flannagan et al., 2009). Another mechanism of phagosome

maturation arrest (PMA) relies on prolonged retention on the phagosome by M. tuberculosis of

coronin-1 (or TACO) (Deghmane et al., 2007), a host factor involved in regulation of actin-based

dynamics (de Hostos, 1999) that specifically prevents lysosomal delivery. It seems that coronin-1

retention is mediated by LpdC (lipoamide dehydrogenase C) (Deghmane et al., 2007), a component

of the antioxidant enzyme complex peroxynitrite reductase/peroxidase (Rhee et al., 2005).

However, the exact mechanism to this retention is unclear. A nucleotide diphosphate kinase (Ndk)

inhibits phagosome maturation by deactivation of Rab5 and Rab7 GTPases (Sun et al., 2010). Rab

proteins are thought to be involved in targeting and tethering the endomembrane organelles to

form phagosomes (Flannagan et al., 2009).

Page 45: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

25 Introduction

Several other mycobacterial proteins have been proposed to play a role in PMA, although, many

times, the exact mechanism or cellular target that interferes with phagosome maturation is

unknown. It is the case of PknG, a serine/threonine protein kinase, present in the genome of all

pathogenic mycobacteria, which inactivation by gene disruption or chemical inhibition results in

maturation of phagosome containing mycobacteria (Walburger et al., 2004). It is proposed that this

protein might phosphorylate and interfere with a host protein that is involved in phagosome

maturation, therefore, preventing its activity. Many other potential factors involved in PMA have

been identified in independent screenings using M. tuberculosis mutants (Brodin et al., 2010;

MacGurn and Cox, 2007; Pethe et al., 2004). However, if these are specific for PMA remains to be

elucidated given that these studies don’t identify most of the mutants in common.

The prevention of phagosome maturation allows pathogenic mycobacteria to reside in an early

phagosome that can access to nutrients such as iron via recycling endosomes (Clemens and

Horwitz, 1996) and avoid antigen-presenting compartments, consequently altering host immune

responses (Ramachandra et al., 2001). In addition, this phagosome has a pH close to neutral (6.4),

correlated with an absence of the vesicular proton-ATPase responsible for acidification of the

phagosome (Sturgill-Koszycki et al., 1994). However, there is still a drop in pH from 7 to 6.4 that

seems to be a signal resulting in the expression of several genes in M. tuberculosis (Rohde et al.,

2007), possibly regulated by PhoP (Walters et al., 2006), the transcriptional regulator of the two-

component system, PhoPR. Indeed, PhoPR seems to be an important for M. tuberculosis to

successfully adapt to the environment found in the host. PhoP regulates a diverse regulon of more

than 150 genes (Gonzalo-Asensio et al., 2008; Walters et al., 2006) including genes involved in lipid

metabolism, general metabolism, and respiration. It also seems to control M. tuberculosis hypoxic

response, by regulating part of the genes regulated by the DosR regulon, including the dosRS genes

(Gonzalo-Asensio et al., 2008). M. tuberculosis phoP mutants are attenuated for growth in mice

(Perez et al., 2001; Walters et al., 2006) and human (Ferrer et al., 2010; Walters et al., 2006)

macrophages which could be, in part, due to their impaired ability to induce PMA (Ferrer et al.,

2010).

PMA was initially proposed as a requirement for the survival of the pathogen (Armstrong and

Hart, 1971). But years later it was shown that live M. tuberculosis can traffic to phagolysosomes, by

opsonization with specific anti-mycobacterial antibodies without affecting bacterial survival

(Armstrong and Hart, 1975). Indeed, mutants that traffic to late compartments that do not have

decreased intracellular survival have been identified (Brodin et al., 2010; MacGurn and Cox, 2007;

Pethe et al., 2004). This means that M. tuberculosis capacity to persist inside macrophages depends

Page 46: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

26 Chapter 1

also in its ability to resist against nitrosative and oxidative stress, caused by reactive nitrogen and

oxygen species, as well as acidic conditions, lysosomal hydrolases and antibacterial peptides

produced by macrophages (Flannagan et al., 2009).

1.6.2 Nitrosative and oxidative stress relevance in tuberculosis

Upon phagocytosis, the first microbicidal activity any microbe will encounter in macrophages

are ROS. The phagocyte NADPH oxidase (phox) complex assembles in the phagocytic cup early in

the process of phagocytosis and generates ROS (Minakami and Sumimotoa, 2006). Reactive

nitrogen species (RNS) are produced by inducible nitric oxide synthase (iNOS). In contrast to

NADPH oxidase, iNOS activity requires de novo protein synthesis and its transcription is triggered

after macrophage activation through PPR signaling together with pro-inflammatory cytokines, such

as IFNγ (Fang, 2004). Macrophage activation also leads to an increased production of ROS (Bedard

and Krause, 2007). Both RNS and ROS can be quite harmful to intracellular pathogens (Flannagan et

al., 2009).

The importance of nitric oxide production in control of tuberculosis is well established in the

mouse model (MacMicking et al., 1997). An early study has also shown that generation of nitrogen

species by iNOS, upon mouse macrophage activation, was responsible for M. tuberculosis growth

inhibition and killing inside these cells (Chan et al., 1992). However, the role of RNS in humans has

been discussed because it is still not clear if these molecules are mycobactericidal in human

macrophages. Human alveolar macrophages were able to kill M. bovis BCG through a NO-

dependent manner (Nozaki et al., 1997). However, another study has shown that the early

inhibition of mycobacterial growth by human alveolar macrophages is NO-independent (Aston et

al., 1998). Other authors have also suggested that killing of the tubercle bacilli is NO-independent

in human macrophages after activation trough TLRs (Thoma-Uszynski et al., 2001) and that these

cells activated with IFNγ didn’t produce detectable amounts of NO. On the other hand, it was

demonstrated that iNOS expression is up-regulated in alveolar macrophages from human

pulmonary TB lesions (Nicholson et al., 1996), associated with increased pulmonary NO exhalation

(Wang et al., 1998). The same has been observed in human granulomas caused by M. tuberculosis

(Choi et al., 2002; Facchetti et al., 1999). These observations in human lesions from people with TB

support the idea that NO must have a role in human host defense against infection with M.

tuberculosis.

Page 47: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

27 Introduction

The biological significance of ROS is clearly demonstrated in patients with chronic

granulomatous disease (CGD), a disease characterized by total absence or low level of ROS caused

by a deficiency of Phox. These persons are highly susceptible to bacterial, fungal and yeast

infections (Heyworth et al., 2003). Increasing numbers in cases of mycobacterial diseases are being

recognized in patients with CGD. A number of patients with BCG disease, clinical disease caused by

BCG vaccine, and tuberculosis have been reported (Bustamante et al., 2007). Nevertheless, the

significance of ROS in host defence against M. tuberculosis remains controversial. Despite the early

demonstration that ROS can be mycobactericidal (Walker and Lowrie, 1981), studies have shown

later that M. tuberculosis could be in fact resistant to killing by oxidative stress caused by

macrophages (Manca et al., 1999). However, no effect on M. tuberculosis intracellular growth in

Phox-deficient mice macrophages was observed (Chan et al., 1992), even though phagocytes

collected from patients with CGD failed to destruct intracellular BCG (Lamhamedi-Cherradi et al.,

1999). In addition, studies using M. tuberculosis-infected mice defective in the production of ROS

are not always congruent (Adams et al., 1997; Cooper et al., 2000; Jung et al., 2002), but in general,

these mice seem to be relatively resistant to TB. These results might be explained by the fact that

M. tuberculosis possesses efficient resistance mechanisms against ROS, thus rendering

antimicrobial effects of these agents ineffective and masking the effects of M. tuberculosis growth

during infection in the absence of ROS.

1.6.3 M. tuberculosis responses against oxidative stress caused by ROS

1.6.3.1 The formation of ROS

ROS are constantly produced as by-products of aerobic respiration. Therefore, oxidative stress is

an unavoidable consequence of the aerobic lifestyle. Aerobic organisms have evolved in order to

maintain levels beneath toxicity avoiding oxidative stress caused by these endogenous ROS (Storz

and Imlay, 1999). Other sources might increase ROS concentrations and perturb this balance in the

redox state of the cell and oxidative stress occurs. This happens when bacteria is captured by

phagocytes, which generate ROS as part of their antimicrobial response (Flannagan et al., 2009).

Phox, expressed mainly in macrophages (including alveolar macrophages) and neutrophils,

generates ROS through the reduction of molecular oxygen (Bedard and Krause, 2007). Phox is a

multi-subunit complex, consisting of a transmembrane heterodimer composed of NOX2 (gp91phox)

and p22phox, known as cytochrome b, the catalytic part of Phox. In resting macrophages, these

transmembrane components are localized mainly in plasma membranes (Bedard and Krause, 2007;

Page 48: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

28 Chapter 1

Bylund et al., 2010). Upon phagocytosis it assembles with three cytosolic subunits, p47 phox, p67 phox

and p40 phox, as well as with Rac, and becomes activated (Minakami and Sumimotoa, 2006) (Figure

6).

Figure 6 - Assembly of the phagocyte NADPH oxidase NOX2. Upon activation, the cytosolic components

p47phox

, p67phox

, p40phox

, and Rac2 translocate to cytochrome b and form a functional NADPH oxidase. The active

enzyme complex transports electrons from cytoplasmic NADPH to extracellular or phagosomal oxygen to generate

superoxide (O2−). Adapted from Bylund et al. (2010).

The active oxidase transfers electrons from cytosolic NADPH to molecular oxygen, releasing

superoxide (O2-) into the phagosomal lumen (Bedard and Krause, 2007) (Figure 6 B). Superoxide

has a low reactivity, and, consequently, low bactericidal potential. It cannot cross bacterial lipidic

membranes at neutral pH but it may do it at acidic pH, found in the phagolysosomes (Storz and

Imlay, 1999). However, superoxide rapidly dismutates into hydrogen peroxide (H2O2)

(spontaneously especially at low pH, or catalysed by superoxide dismutase) which readily

permeates biological membranes and oxidize several molecules. Finally, H2O2 in turn can produce

hydroxyl radicals (•OH) (Figure 7), which are strong oxidants, when reacting with iron in what is

known as Fenton reaction (discussed below).

Page 49: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

29 Introduction

Figure 7 – Reactive oxygen species decomposed from

superoxide produced by phagocyte oxidase. Adapted from

Fang et al. (2004).

The contribution of NOX2 to microbial killing lies in both direct effects, due to ROS damaging

character on proteins, lipids and nucleic acids (Imlay, 2008; Storz and Imlay, 1999; Zahrt and

Deretic, 2002), as well as indirect effects through changes in the phagosomal pH and ion

concentrations (Bedard and Krause, 2007). ROS also seem to be involved in cellular signaling,

especially because of their potential for protein oxidation. For example, phosphatases inhibition by

oxidation of cysteines in their catalytic region (Denu and Tanner, 1998), might affect the

phosphorylation state of numerous other proteins involved in signal transduction. In macrophages,

ROS seems to have an important function in activation of kinases and transcription factors, gene

expression, regulation of proliferation, and apoptosis (Bedard and Krause, 2007).

Bacterial pathogens have developed strategies to counteract production of ROS by the host,

which interfere with the synthesis of these products, catabolize or repair the damage caused by

them. Most of what we know about these mechanisms is due to its study in E. coli and S.

typhimurium, the two model systems for oxidative stress responses (Zahrt and Deretic, 2002).

1.6.3.2 ROS scavenging systems

In early studies mycobacterial cell wall was found to be implicated in ROS resistance particularly

due to LAM (Chan et al., 1991) and PGL-1 (Chan et al., 1989) ability to scavenge oxygen-radicals.

Antioxidant enzymes are present in most bacteria dealing with endogenous or exogenous ROS

(Zahrt and Deretic, 2002) including: catalases that catalyze the decomposition of hydrogen

peroxide to water and oxygen, superoxidases that convert two superoxide anions into a molecule

of hydrogen peroxide and peroxidases that catalyze the reduction of hydrogen peroxide by organic

Page 50: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

30 Chapter 1

reductants, such as glutathione. M. tuberculosis has a protein with both catalase and peroxidase

activities, KatG (Heym et al., 1993; Manca et al., 1999). A katG mutant was hypersensitive to H2O2

and attenuated in wild-type mice as well as in wild-type mouse macrophages. However, it was

virulent in gp91-deficient mice and in macrophages from these phox-deficient mice, unable to

produce an oxidative burst (Ng et al., 2004). Thus, KatG contributes to M. tuberculosis virulence by

catabolizing peroxides produced by Phox. M. tuberculosis also has two superoxide dismutases:

SodA a Mn,Fe enzyme (Zhang et al., 1991) and SodC a Cu,Zn enzyme (Wu et al., 1998). SodC was

found to be important for bacteria’s resistance against exogenous superoxide as well as H2O2, as

sodC mutants had increased susceptibility to these ROS when compared to wild-type (Dussurget et

al., 2001; Piddington et al., 2001). A sodC mutant had also enhanced susceptibility to killing by

IFNγ-activated murine peritoneal macrophages, but not by Phox-deficient or non-activated murine

macrophages (Piddington et al., 2001), suggesting that SodC is important for M. tuberculosis

resistance against ROS produced by activated macrophages. However, sodC mutants were not

attenuated in mice or guinea pigs (Dussurget et al., 2001; Piddington et al., 2001). It was suggested

that SodA might compensate for SodC deletion in vivo. SodA seems to be essential for M.

tuberculosis growth as an attempted of inactivating its gene failed (Dussurget et al., 2001).

However, a M. tuberculosis mutant having diminished production of SodA, was more susceptible to

H2O2 and significantly attenuated in mice (Edwards et al., 2001), which exhibited increased

apoptosis of mononuclear cells. On the other hand, a BCG strains overexpressing SodA induced a

reduction in apoptosis in macrophages, as well as an impaired IFNγ and increased IL-10 release in

mice (Jain et al., 2011). As previously referred, ROS induce several cellular functions. These include

induction of apoptosis through the inhibition of phosphatases, and consequently, activation of

signaling molecules such as MAP kinases (Bedard and Krause, 2007). M. tuberculosis has the ability

to inhibit apoptosis which was recently found to be correlated with the inhibition of ROS

accumulation (Miller et al., 2010). Therefore, antioxidant enzymes, such as SodA and KatG, seem to

be involved in M. tuberculosis apoptosis inhibition through elimination of ROS (Edwards et al.,

2001; Miller et al., 2010). By affecting ROS levels, in the host cell, M. tuberculosis not only avoids

killing, but also interferes with other host defense mechanisms against pathogens.

Other antioxidant enzymes found in M. tuberculosis include an alkyl hydroperoxide reductase,

AhpC, involved in detoxification of organic peroxides, as shown by the increased sensitivity of an

ahpC mutant to cumene hydroperoxide, but not hydrogen peroxide (Springer et al., 2001). Even

though M. tuberculosis ahpC mutant had a decreased survival in macrophages (Master et al.,

2002), growth was not affected in mice, suggesting that AhpC doesn’t have a significant role during

Page 51: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

31 Introduction

infection in vivo (Springer et al., 2001). The ahpC gene belongs to an operon along with ahpD. AhpD

was found to be implicated in AhpC peroxidase activity, along with DlaT (SucB) and LpdC (Rv0462)

enzymes, involved, as well, in important metabolic pathways. AhpC, AhpD, DlaT and LpdC were

found to form a peroxynitrite reductase-peroxidase complex responsible for peroxynitrite

detoxification (Bryk et al., 2002).

1.6.3.3 Protein and lipid repair

Specialized enzyme systems comprising members of the oxidoreductase family are involved in

generalized protein reduction. In E. coli the thioredoxin (Trx) system comprises two thioredoxins

and a thioredoxin reductase. The glutaredoxin (Grx) system is composed of glutathione, three

glutaredoxins, and a glutathione oxidoreductase (Holmgren, 2000).

M. tuberculosis has mycothiol, instead of glutathione, and several thioredoxins, including AhpD

(referred above), involved in AhpC reduction, as well as a thioredoxin reductase (Akif et al., 2008).

Mutants in genes involved in mycothiol biosynthesis have shown increased susceptibility to

oxidative stress (Buchmeier et al., 2006; Buchmeier et al., 2003), although no significant growth

defects were observed during mice infection (Vilcheze et al., 2008). M. tuberculosis thioredoxins

are involved in protection against peroxides (Zhang et al., 1999) and trx genes are transcribed

under a variety of oxidative stress conditions, suggesting a role in M. tuberculosis oxidative stress

response. WhiB proteins were initially thought to have a role as transcriptional regulators,

however, most of these proteins were found to be disulfide reductases with properties similar to

thioredoxins (Alam et al., 2009), involved in M. tuberculosis oxidative stress response. It was shown

that each of the M. tuberculosis whiB family member has a unique transcriptional response pattern

to different stresses and two of these, whiB3 and whiB6, were found to be commonly induced by

diamide and cumene hydroxiperoxide (Geiman et al., 2006).

Sulfur assimilation is important for the biosynthesis of cysteine, methionine, and a variety of

cofactors that will then be integrated in reduced-sulfur-containing molecules, such as mycothiols

and thioredoxins. Genes involved in this pathway are induced in M. tuberculosis inside

macrophages (Schnappinger et al., 2003), showing the importance of these molecules in M.

tuberculosis intracellular lifestyle. CysH is an enzyme involved in this pathway and a deletion

mutant in the cysH gene shows an increased sensitivity to H2O2 and peroxynitrite. This mutant had

an attenuated phenotype in wild-type mice, in the persistence phase of infection, but was fully

virulent in NOX2-/- mice treated with iNOS inhibitor (Senaratne et al., 2006). The attenuated

Page 52: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

32 Chapter 1

phenotype of ΔcysH mutant in mice was, probably, due to an impaired antioxidant response

caused by a decrease in reduced-sulfur-containing molecules such as mycothiols.

Methionine sulfoxide reductase (Msr) is responsible for the conversion of methionine sulfoxide,

the product of oxidized methionine, whether in proteins or as free amino acid. Two Msr exist in

almost all living organisms, MsrA and MsrB that are responsible for the reduction of the two

stereomeric forms of methionine sulfoxide: methionine S-sulfoxide and methionine R-sulfoxide. In

E. coli or Helicobacter pylori one of these genes has been shown to be important for bacteria’s

oxidative stress response. In M. tuberculosis, only a double mutant in both msrA and msrB showed

increased susceptibility to hypochlorite but not to other oxidants such as hydrogen peroxide (Lee

et al., 2009b). Therefore, their role in protection of M. tuberculosis against oxidative stress in

macrophages remains unclear.

The membrane-associated protease Rv3671c also seems to protect M. tuberculosis against

oxidative stress probably by degrading damaged proteins, however, this has still to be confirmed

(Biswas et al., 2010).

A M. tuberculosis overexpressing Wag31, a protein that regulates polar cell wall synthesis, was

found to be attenuated in human macrophages and had increased susceptibility to hydrogen

peroxide (Mukherjee et al., 2009). Wag31 protects PBP3 protein (FtsI) (involved in synthesis of

septal peptidoglycan) from proteolysis induced in oxidative stress conditions in mycobacteria.

These results suggest that M. tuberculosis had also developed mechanisms to preserve cell shape

under oxidative stress conditions, in order to survive.

Curiously, a locus - mel2 - with homologous genes involved in bioluminescence in other bacteria

seems to protect M. tuberculosis against oxidative stress. A M. tuberculosis mutant in this locus has

increased susceptibility to ROS and a defective growth in activated mouse macrophages that was

less obvious in Phox-/- macrophages. Disease was found to be less severe in mice infected with this

mutant strain but similar to wild-type in Phox-/-. Although the mechanism by which this occurs

remains unknown, mel2 mutants have increased levels of oxidized lipids (Cirillo et al., 2009). Thus,

it might be involved in protection or repair of oxidized lipids.

The mechanisms described above show that M. tuberculosis oxidative stress response include

not only antioxidant enzymes and other molecules that scavenge ROS, but also, mechanisms

involved in the repair of the damages caused in proteins and lipids. ROS also cause damage in the

DNA (Demple and Harrison, 1994), which means that M. tuberculosis should have efficient DNA

repair mechanisms as well. Indeed, several transcriptomic analyses show that DNA repair genes are

induced after exposure to H2O2 (Boshoff et al., 2003; Cabusora et al., 2005; Schnappinger et al.,

Page 53: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

33 Introduction

2003; Voskuil et al., 2011). DNA repair mechanisms have also been identified as important for M.

tuberculosis survival during infection in mouse (Schnappinger et al., 2003) and human (Cappelli et

al., 2006) macrophages; as well as in mouse (Sassetti and Rubin, 2003) and human (Rachman et al.,

2006) lung tissues.

It was suggested, from early studies in E. coli, that most of the cell death that occurs upon H2O2

exposure is probably due to DNA damage (Imlay and Linn, 1986), emphasizing the potential killing

effect of ROS trough DNA damage. Pathogens have adapted and DNA repair mechanisms have

been shown to be essential for the survival of the intracellular pathogens Salmonella typhimurium

(Buchmeier et al., 1995) and H. pylori (O'Rourke et al., 2003) by repairing DNA damage resulting

from oxidative stress.

1.6.3.4 Oxidative DNA damage caused by ROS

Oxidative DNA damages constitute, probably, the most varied class of DNA damages (Demple

and Harrison, 1994). ROS induce a vast number of different types of DNA damages, including:

strand breaks (particularly single- although double-strand breaks might also occur), base loss

leading to abasic (apurinic, apyrimidinic or AP) sites, and base damages. Over 80 products of DNA

base damage caused by ROS are known, among which, the oxidation product of guanine, 7,8-

dihydro-8-oxo-2'-deoxyguanosine (8-oxoG) either in the DNA or nucleotide pool, is one of the most

frequent (Friedberg et al., 2006). M. tuberculosis is at greater risk of acquiring this kind of lesion

due to the high GC content of its genome (Cole et al., 1998).

The hydroxyl radical (•OH) is the most reactive of the primary ROS, oxidizing organic molecules

almost indiscriminately. In the DNA it can add or abstract hydrogen atoms and originate many

products. However, oxidation of DNA is surely not a result of free •OH. Its reactivity is so high that

it reacts with another cellular component before it reaches DNA. Consequently, to oxidize DNA,

•OH must be generated immediately next to a nucleic acid molecule (Friedberg et al., 2006).

Hydrogen peroxide, as opposite to •OH, is not as highly reactive with DNA. However, it acts as a

much stronger oxidant when decomposed by ferrous ions. In fact, the main source of DNA damage

by ROS is thought to be the formation of Fenton oxidants, such as •OH, by reaction of H2O2 with

iron-complexed DNA (Figure 8) (Friedberg et al., 2006). Another frequent base damage, induced by

H2O2, is 2,6-diamino-4-hydrixy-5-formamidopyrimidine (FaPy) (Friedberg et al., 2006) resulting also

from the oxidization of guanine. Free superoxide (O2-) is also not very reactive with DNA. However,

as referred previously, O2- dismutate into H2O2 and it has the ability to release protein-bound iron

to produce Fe2+ that will be used in the Fenton reaction (Figure 8) (Friedberg et al., 2006).

Page 54: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

34 Chapter 1

Figure 8 – Cellular reactions leading to oxidative DNA damage via the Fenton Reaction. Superoxide

dismutates into H2O2 and releases protein-bound iron, which in turn can react to form oxidants such as ⋅OH that

will cause DNA damage. Adapted from Friedberg et al. (2006).

1.6.3.5 M. tuberculosis DNA repair

To prevent DNA damage it was recently shown that M. tuberculosis seems to be physically

protected by Lsr2, a histone-like protein (Colangeli et al., 2009). An M. smegmatis mutant was

found to be hypersusceptible to H2O2 and attenuated in wild-type, but virulent in Phox-/- deficient,

macrophages. No mutants in lsr2 gene could be constructed in M. tuberculosis, highlighting the

importance of such protein for this pathogen and as a future drug target (Colangeli et al., 2009).

Due to the technical difficulty of working with such a slow growing pathogen, the study of M.

tuberculosis DNA repair systems has advanced slower than for other bacteria. As a result, most of

the assumptions are still made based on homology rather than on functional studies. Genes

necessary to perform base excision repair, nucleotide excision repair and recombinational repair

were identified. In addition, genes involved in the SOS response – a mechanism that responds to

DNA damage by inducing various genes required for DNA repair - were also identified. In contrast,

no homologs of mismatch repair genes were identified (Cole et al., 1998; Mizrahi and Andersen,

1998), a finding that should have potentially significant implications with respect to genome

stability.

Major DNA repair mechanisms that control DNA damages in bacteria and the corresponding

homologues found in M. tuberculosis are represented in Figure 9 (Dos Vultos et al., 2009).

Page 55: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

35 Introduction

Figure 9 – Major mechanisms repairing DNA damages in bacteria. Listed beside each pathway are the

homologues of DNA repair proteins found in M. tuberculosis. Mismatch repair homologs were found in M.

tuberculosis, and so this pathway is not shown in the figure. Adapted from Dos Vultos et al. (2009).

1.6.3.5..1 Base excision repair pathway

Base excision repair (BER) is probably one of the most frequently used DNA repair systems in

nature since it is responsible for the repair of the majority of DNA damages (Friedberg et al., 2006).

BER is initiated by DNA glycosylases, which excise the chemically altered or inappropriate bases

from DNA – like 8-oxoG – by cleavage of the N-glycosyl bond between the base and the

deoxyribose sugar. This forms a site in DNA without a base, called AP site. AP endonucleases

specifically recognize these sites in the DNA and produce nicks immediately 5’ to the AP site. Some

DNA glycosylases have an associated AP lyase activity that cleaves the phosphodiester backbone 3’

to the AP site. Then, the action of an exonuclease or DNA-deoxyribophosphodiesterase (dRpase)

completes the removal of the damaged nucleotide and DNA polymerase and DNA ligase conclude

the repair (Dianov and Lindahl, 1994; Friedberg et al., 2006; Seeberg et al., 1995) (Figure 9).

- Oxidative DNA damage repair by BER

Oxidative DNA damage is primarily processed by the BER pathway, given that DNA bases are

particularly susceptible to oxidation mediated by ROS (Dizdaroglu, 2005). As referred, one of the

most frequently occurring stable and abundant oxidized base lesions in DNA is 8-oxoG. This lesion

Page 56: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

36 Chapter 1

has strong promutagenic properties. During replication, 8-oxoG frequently mispairs with the

nucleotide A (Shibutani et al., 1991). This error is not detected by replicative DNA polymerases

leading to G→T/C→A transversions (Cheng et al., 1992). Enzymatic systems dealing with the 8-

oxoG are represented on Figure 10. Prior to DNA replication, at 8-oxoGC sites, repair can be

effected by conventional BER with MutM (Fpg) DNA glycosylase operating on the 8-oxoG damage

(Figure 10B, right pathway). If this base is not removed before replication, DNA polymerase can

introduce a C opposite to the lesion leading to another opportunity for MutM to catalyse the repair

(Figure 10B, left pathway). But DNA polymerase can also introduce an A opposite to 8-oxoG (Figure

10B, central pathway). In this situation, MutY DNA glycosylase excises the A that is usually replaced

by a C resulting in a 8-oxoGC base pair that is, again, repaired by the BER system involving the DNA

glycosylase MutM.

Moreover, ROS can contaminate the nucleotide pool with 8-oxod-GTP. MutT hydrolyses 8-oxo-

dGTP back to the monophosphate form, removing it from the dNTP pool (Figure 10C, right

pathway). If replication occurs with some 8-oxo-GTP in the dNTP pool, usually T is preferentially

inserted opposite to A. However, if 8-oxo-GTP is incorporated instead of T, 8-oxoG may be

removed either by MutM (Figure 10C, left pathway) or MutY (Figure 10C, central pathway), as

described above.

Figure 10 - Enzymatic systems that protect from the mutagenic effects of 8-oxoguanine. Details are discussed

in the text. Adapted from Friedberg et al. (2006).

Page 57: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

37 Introduction

This triplet of DNA repair enzymes – MutM, MutY and MutT – comprises the G° system that is

involved in handling with the potential mutagenic effects of guanine oxidation (David et al., 2007;

Michaels and Miller, 1992). Four mutT homologues have been annotated in mycobacterial

genomes, which contain a single mutY homolog (Cole et al., 1998; Mizrahi and Andersen, 1998).

The study of knockout mutants for MutT1, MutT2, MutT3 and Rv3908 (MutT4) in M. tuberculosis

has revealed that only the absence of mutT1 leads to an increase in the mutation frequency

(mutator phenotype) (Dos Vultos et al., 2006) although at a much lower level than the observed for

other bacteria. This may be due to the existence of other enzymes capable of counteracting this

kind of oxidative stress. Biochemical analysis show that M. tuberculosis MutY is involved in the

excision of A opposite to 8-oxoG in the DNA, as it happens in E. coli. However, excisions of G or T

opposite to 8-oxoG can also occur (Kurthkoti et al., 2010). M. tuberculosis genome possesses two

homologues of mutM (Cole et al., 1998), however, one of these (rv0944) may code for a protein

that is not functional (Guo et al., 2010). A study of MutM deficient M. smegmatis has shown that

this protein plays an important role in protecting bacteria from oxidative DNA damage. The

complementation of this mutant with the M. tuberculosis mutM gene, coding for the main MutM

(Rv2924c), could restore the sensitive phenotype to oxidative stress (Jain et al., 2007).

Interestingly, M. tuberculosis MutM was also found to be important for infection of non-human

primates (Dutta et al., 2010).

Another enzymes that excise oxidized bases are endonuclease VIII (Nei) and Nth. These act

primarily on oxidized pyrimidines, in contrast to Fpg that acts primarily on oxidized purines

(Dizdaroglu, 2005). Homologues of these proteins are present in M. tuberculosis (Cole et al., 1998;

Mizrahi and Andersen, 1998).

DNA glycosylases give rise to AP sites. Additionally, these sites can also arise spontaneously or

be directly induced by ROS (Demple and Harrison, 1994). Such lesions are repaired by the

ubiquitous AP endonucleases, including XthA and Nfo, which belong to the oxidative defence

regulon in E. coli (Storz and Imlay, 1999). These proteins are both encoded in the genome of M.

tuberculosis (nfo corresponds to end) (Cole et al., 1998) (Figure 9) and their biological importance is

shown by the fact that no variations are observed for xthA in clinical strains (Dos Vultos et al.,

2008). The existence of two enzymes for the same function highlight that dealing with abasic sites

is a crucial aspect of DNA repair and may be a reflection of either the high number of DNA

glycosylases that M. tuberculosis possesses, or the constant aggression by ROS in the host.

Page 58: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

38 Chapter 1

The filling of gaps that result from AP endonuclease activity is mediated mainly by polymerase I,

PolA (Dianov and Lindahl, 1994; Seeberg et al., 1995). M. tuberculosis possesses a DNA-dependent

PolA (Cole et al., 1998). In E. coli, it was demonstrated that overproduction of DnaE protein (an

subunit of DNA polymerase III) could restore viabilityy, in a conditionally unviable strain deficient in

PolA (Witkin and Roegner-Maniscalco, 1992). M. tuberculosis contains two apparently functionally

redundant replicative DNA polymerases designated DnaE1 and DnaE2 (Cole et al., 1998), but lacks

PolII. DnaE2 was found to be upregulated in vitro by several DNA damaging agents (including H2O2),

as well as during infection in mice. It seems to be an error prone polymerase that is the primary

mediator of survival through inducible mutagenesis and plays a role in the emergence of drug

resistance (Boshoff et al., 2003). Indeed, mutations in the rpoB gene that confer resistance to

rifampicin, were associated with an increased dnaE2 expression, suggesting a role for DnaE2 in

adaptation of fitness impaired, rifampicin resistant, M. tuberculosis strains. It was postulated that

by increasing expression of this error-prone polymerase the probability of a subsequent adaptation

would also be augmented (Bergval et al., 2007). Other genes, dnaZX, dnaQ and dnaN, encoding ,

and ß subunits of polymerase III, respectively, are also found in the M. tuberculosis genome as

well as the DNA ligases: LigA, LigB, LigC and LigD (Cole et al., 1998; Mizrahi and Andersen, 1998)

(Figure 9).

Although the study of M. tuberculosis repair of oxidative damage by BER needs to be deepened,

it is already clear that these bacteria are well adapted to environments where oxidative stress is

prevalent. This is illustrated by the sheer number of putative genes whose products might be

involved in dealing with this kind of aggressions and by the restricted nucleotide diversity that

these genes present (Dos Vultos et al., 2008). In addition, the analysis of rpoB mutation patterns in

both clinical (O'Sullivan et al., 2005) and laboratory (Morlock et al., 2000) strains has led to the

conclusion that oxidative damage is generally not the driving force behind mutations in the M.

tuberculosis genome. Finally, genes of the BER system were found to be essential for M.

tuberculosis survival in mice (Sassetti and Rubin, 2003) revealing the importance of this system to

resist oxidative damage during infection in vivo.

- BER Repair of DNA damages caused by other forms of stress

Several DNA glycosylases involved in the repair of other kinds of damaged bases were also

found in the genome of M. tuberculosis (Mizrahi and Andersen, 1998). It is the case of tagA and

alkA, important genes in repairing alkylative DNA damages that can be induced by RNS (Durbach et

al., 2003; Taverna and Sedgwick, 1996). The tagA gene encodes a highly specific 3-methyladenine

Page 59: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

39 Introduction

DNA glycosylase I that is constitutively expressed (Bjelland et al., 1993). AlkA encodes a 3-

methyladenine DNA glycosylase II (O'Brien and Ellenberger, 2004), which is a component of the

adaptive response controlled by ada. The ada gene encodes O6-alkylguanine-DNA alkyltransferase

I. A second type of O6-alkylguanine-DNA alkyltransferase is encoded by the ogt gene in E. coli

(Friedberg et al., 2006). In M. tuberculosis, ada and alkA are predicted to encode fused proteins

and to be part of an operon including ogt (Cole et al., 1998). M. tuberculosis mutants lacking this

operon were shown to be hypersensitive to the genotoxic effects of N-methyl-N’-nitro-N-

nitrosoguanidine (MNNG), an alkylating mutagen (Durbach et al., 2003). The exposure to the DNA

damaging agents - UV radiation, mitomycin C and H2O2, results in the upregulation of this operon

as well (Boshoff et al., 2003). However, these mutants were not attenuated in mice, suggesting

that nitrosative stress does not induce cytotoxic DNA damage in M. tuberculosis in vivo (Durbach et

al., 2003).

1.6.3.5..2 Nucleotide excision repair pathway

Nucleotide excision repair (NER) is an alternative pathway to BER, in which genomic damage is

repaired by means of incisions in regions flanking the damaged DNA, leading to the excision of an

oligonucleotide rather than a single base (Friedberg et al., 2006). In E. coli NER involves UvrA, B and

C proteins, which interact in complex. UvrA binds to DNA and leads to the formation of an UvrA-

UvrB-DNA complex that recognizes the damage. Then, UvrA dissociates and UvrB recruits UvrC that

catalyse incisions 5’ and 3’ to DNA damage. DNA helicase (UvrD), polymerase, and ligase reactions

then eliminate the damaged oligonucleotide and complete repair (de Laat et al., 1999) (Figure 9).

- Oxidative DNA repair by NER

Although this system is not pivotal for handling oxidative damage (it is the core mechanism for

repair of UV-induced damage) evidence indicates a demonstrable function as a secondary defence

against oxidative damages in E. coli and in Saccharomyces cerevisae (Demple and Harrison, 1994).

It is an extremely important DNA repair system that recognizes a wider range of damaged bases

than the BER system, due to the use of more generic endonucleases.

Homologues of uvrABC were found in M. tuberculosis (Cole et al., 1998). The expression of uvr

genes was increased in M. tuberculosis within human macrophages, which highlights the

importance of the uvr system for M. tuberculosis survival upon infection (Graham and Clark-Curtiss,

1999). Additionally, uvr genes seem to be involved in H2O2 response network (Cabusora et al.,

2005) being expression of uvrA and uvrB increased upon exposure to H2O2 (Boshoff et al., 2003).

Page 60: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

40 Chapter 1

However, inactivation of uvrB resulted in severe sensitivity to acidified nitrite and UV light but not

to ROS in M. tuberculosis. Yet, an uvrB mutant attenuated phenotype was reverted only in mice

lacking both iNOS and Phox, suggesting that UvrB is important for resistance against combined RNS

and ROS during infection in vivo (Darwin and Nathan, 2005). Importantly, uvrB was also shown to

be required for M. tuberculosis growth in primate lungs (Dutta et al., 2010). An M. smegmatis uvrB

mutant, however, has shown increased susceptibility to oxidative DNA damages (Kurthkoti et al.,

2008) and a recent study has shown that M. tuberculosis deficient in UvrA is also sensitive to

several DNA damaging agents including ROS (Rossi et al., 2011). It seems, thus, that NER proteins

are important players in the repair of oxidative DNA damages in mycobacteria.

Although most bacteria carry just one gene encoding DNA helicase II, M. tuberculosis possesses

two putative genes encoding this protein: uvrD1 and uvrD2 (Cole et al., 1998). Interestingly, uvrD1

seems to be critical for mycobacterial repair of UV damage and is upregulated upon H2O2 stress

(Boshoff et al., 2003), suggesting a role in NER. This has been recently confirmed. M. tuberculosis

uvrD1 mutant has shown an increased susceptibility to UV irradiation as well as to mitomycin C and

ROS-inducing agents (Houghton et al., 2012). The role of UvrD1 in mycobacterial DNA repair is of

significant interest given that deletion of uvrD1 attenuates M. tuberculosis growth in macrophages

and persistence in mice (Houghton et al., 2012).

1.6.3.5..3 Recombinational repair

Oxidative DNA damage includes DNA strand breaks, particularly single strand breaks (SSBs)

because two nearby attacks in each strand are very rare under physiological conditions. But, if this

happens, double strand breaks (DSBs) can occur. DSBs can also arise if a replication fork encounters

a SSB in the DNA, and SSBs may occur if the replication fork encounters another kind of lesion in

the DNA (Friedberg et al., 2006). Thus, SSBs and DSBs might occur by a direct or indirect effect of

oxidative DNA damaging agents. These lesions are potentially lethal and require repair through

homologous and non-homologous recombination mechanisms (Demple and Harrison, 1994;

Slupphaug et al., 2003). In homologous recombination (HR) a second intact copy of the broken

chromosome segment serves as template, which in prokaryotes corresponds to the newly

replicated sister chromatid (Friedberg et al., 2006). Proteins involved in HR play key roles in DNA

repair mechanisms and two major pathways can be found: the RecBCD and RecF pathways. It is

now evident that DSBs initiate recombinational events carried out by the RecBCD pathway,

whereas single strand gaps initiate recombinational events carried out by the RecF pathway

(Morimatsu and Kowalczykowski, 2003). In E. coli, RecBCD is a helicase that splits the duplex and

Page 61: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

41 Introduction

digests them until it reaches a recombinational hotspot (Singleton et al., 2004) (Figure 11A). In the

RecF pathway, RecQ and RecJ process gapped DNA before RecFOR proteins recognize the single

strand gap (Figure 11B) (Morimatsu and Kowalczykowski, 2003). Both pathways provide an ssDNA

molecule coated with RecA, facilitating the invasion of a homologous molecule (Figure 11). Strand

exchange is promoted by RecA, resulting in formation of a holliday junction that will be resolved by

the RuvABC complex or the RecG ligase (Friedberg et al., 2006). In M. tuberculosis the RecBCD

pathway seems to be intact (Mizrahi and Andersen, 1998). The RecF pathway was initially thought

to be defective given that recJ is absent. However, recR is one of the genes upregulated in M.

tuberculosis upon capreomycin exposure (Fu and Shinnick, 2007), a drug used for treatment of

MDR-TB. Furthermore, both recR and recF are upregulated upon translational inhibition induced by

drugs (Boshoff et al., 2004) suggesting that, despite of the absence of recJ, this pathway is active in

M. tuberculosis.

Figure 11 – Early steps during the repair of double strand breaks (DSBs) (A) and single strand breaks (SSBs) (B)

in the DNA by homologous recombination mechanisms. (A) In the RecBCD pathway, RecBCD binds to the end of a

DSB, unwinds and degrades the double strand DNA until it finds a DNA sequence (χ). RecA is then loaded onto the

χ-containing single-stranded DNA. (B) In the RecF pathway, RecQ and RecJ process the gapped DNA. Then RecFOR

recognize the SSB-coated GAP and lead to RecA filament assembly. Adapted from Friedberg et al. (2006).

A B

Page 62: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

42 Chapter 1

As referred, DSBs can also be repaired by another mechanism: the non-homologous end-joining

(NHEJ). NHEJ does not rely on a homologous DNA template, which means it can operate in

situations when only one chromosomal copy is available (Friedberg et al., 2006). NHEJ requires the

approximation of the broken ends, a process that is assisted by the DNA-end-binding protein Ku,

followed by the sealing of, at least, one of the broken strands by a specialized DNA ligase (Figure 9).

Unlike HR, which is generally error free, NHEJ can be either faithful, if the ends are sealed directly,

or mutagenic, if the ends are remodelled by nucleases or polymerases before sealing by DNA

ligases (Shuman and Glickman, 2007). This pathway was thought to be restricted to eukaryotes but

it was recently identified in prokaryotes, including M. tuberculosis (Gong et al., 2005), being LigD

the specialized DNA ligase involved in (Della et al., 2004). Mycobacteria possess a backup

mechanism for LigD-independent error-prone repair of blunt-end DSBs, probably involving LigC.

This is suggested by the fact that blunt-end NHEJ efficiency in LigD-deficient cells is reduced by 20-

fold when ligC is deleted (Gong et al., 2005). Another Ku-dependent protein is, UvrD1. However,

exactly how it contributes to DSB repair remains to be determined. This suggests a cross-talk

between NHEJ and NER pathways (Sinha et al., 2007).

The reason why do only certain bacterial species possess this pathway is unclear. In

mycobacteria, even though expression profiles suggest that M. tuberculosis ligD and ku

transcription is not regulated during infection in human macrophages (Cappelli et al., 2006), NHEJ

may be important for survival within these cells, where bacteria are continuously exposed to

genotoxic compounds that induce DSBs, such as ROS. This may explain why M. tuberculosis has

retained a functional NHEJ apparatus that has been lost by other species. On the other hand, this

mechanism would contribute to mutagenesis that can sometimes be a selective advantage

(Shuman and Glickman, 2007), for example for the acquisition of antibiotic resistance mutations.

- RecA – a central molecule in DNA damage response

RecA is a central component of bacterial response to DNA damage. Not only it has a direct role

in recombinational DNA repair as described above, but it also controls the expression of other

genes whose function promotes increased survival following DNA damage, in conjunction with the

repressor protein LexA (Friedberg et al., 2006; Little, 1982). This is the so-called SOS response. M.

tuberculosis has, with the exception of polB and umuD, the genes required for a functional SOS

system in E. coli (Cole et al., 1998; Mizrahi and Andersen, 1998). As in most species of bacteria, the

expression of M. tuberculosis recA has been shown to be inducible by DNA damaging agents

Page 63: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

43 Introduction

(Boshoff et al., 2004). RecX seems to be a negative regulator of RecA in M. tuberculosis (Venkatesh

et al., 2002).

Recently, the mechanisms underlying M. tuberculosis response to DNA damage were studied,

through a global analysis of gene expression following DNA damage induced by mitomycin C, in

both wt and recA- M. tuberculosis. Curiously, the majority of genes induced by DNA damage didn't

have known roles in DNA repair. However, it was noticed that most of the inducible genes with

known or predicted function in DNA repair did not depend on recA for induction (Rand et al.,

2003), a rare occurrence in bacteria, only documented for Acinetobacter calcoacetius (Rauch et al.,

1996). Among those, were genes involved in NER, BER, and recombination. As it has been

previously suggested (Brooks et al., 2001), it appears that an alternative mechanism of gene

regulation, RecA/LexA – independent, important for DNA repair, exists in M. tuberculosis (Rand et

al., 2003). A motif has been identified that seems to define the RecA/LexA-independent promoter

(RecA-NDp) (Gamulin et al., 2004).

It remains unclear why does M. tuberculosis have multiple mechanisms for DNA damage

induction. A possible explanation is that each mechanism responds to different stimuli. For

example, the RecA-NDp-mechanism may be important for survival in macrophages, given that the

majority of genes under the control of this promoter are induced in the macrophage environment

to a significant level (Gamulin et al., 2004; Schnappinger et al., 2003). Implication of the RecA-NDp-

mediated response in the regulation of a variety of genes that are involved in M. tuberculosis

oxidative stress response, such as ahpC, argues for this possibility (Gamulin et al., 2004).

1.6.3.5..4 The lack of Mismatch repair

Mismatch repair (MMR) systems are important for mutation avoidance by repairing

mismatched base pairs in the DNA (Friedberg et al., 2006). For example, MMR also contributes to

the repair of 8-oxoG as a backup for the repair of newly replicated strains (Friedberg et al., 2006).

One of the most striking characteristics of M. tuberculosis DNA repair system is the absence of

homologs for MMR, which highly suggests that these bacteria do not perform this kind of repair

(Dos Vultos et al., 2009). This should have potentially significant implications with respect to

genome stability and could lead us to regard M. tuberculosis as possible natural mutator. Even

though it has been show recently that M. tuberculosis complex strains show high levels of

polymorphisms in DNA repair, replication and recombination genes (Dos Vultos et al., 2008), M.

tuberculosis genomes are very stable when compared to other organisms such as H. pylori, where

the absence of a functional MMR system was correlated to a remarkably high level of genetic

Page 64: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

44 Chapter 1

diversity (Kang and Blaser, 2006). Nevertheless, its capacity to adapt to host conditions has been

demonstrated by the appearance of MDR as well as XDR, and, more recently, TDR strains resistant

to all available drugs against TB (Zumla et al., 2012).

The possibility of a higher fidelity in the processivity of DNA as a compensation for the lack of

MMR can be posed, as M. tuberculosis has a slower rate of DNA synthesis when compared to E. coli

(Hiriyanna and Ramakrishnan, 1986). The use of codons in a context-specific way may also

contribute to the stability of the genome in deficient MMR M. tuberculosis by reducing the number

of targets for frameshift mutations (Wanner et al., 2008). In addition, besides the important role in

counteracting directly the damages in DNA provoked by host conditions, it is possible that NER (as

well as BER) is involved in compensating for MMR deficiency, a model that has recently been

proposed (Guthlein et al., 2008).

1.6.3.6 Oxidative stress response regulation

S. typhimurium and E. coli oxidative stress response is primarily controlled by SoxR and OxyR

regulators. SoxR, in response to superoxide, induces expression of several genes including sodA.

OxyR is activated by hydrogen peroxide and stimulates transcription of genes such as katG

(catalase) and ahpCF (alkylhydroxyperoxidase reductase). Although OxyR is the most common

regulator of responses to hydrogen peroxide, other regulators might exist. PerR was identified in

Bacillus subtilis and in Staphylococcus aureus as transcriptional repressor that regulates

transcription of most of the genes regulated by OxyR (Imlay, 2008).

In M. tuberculosis and other closely related species of the M. tuberculosis complex, oxyR is a

pseudogene containing multiple frameshift mutations and deletions (Deretic et al., 1995). In some

fast-growing mycobacteria, such as M. smegmatis, oxyR seems to be absent from their genomes. In

contrast, an intact oxyR has been observed in several other mycobacterial species including M.

leprae, M. marinum and M. avium (Pagan-Ramos et al., 1998). As for soxRS, no apparent homolog

seems to exist in M. tuberculosis genome (Cole et al., 1998). This apparent absence of important

oxidative stress response regulators in M. tuberculosis would explain the very little transcriptional

response to H2O2 or menadione (a superoxide producer) (Garbe et al., 1996) when compared to

other species, like E. coli or Salmonella (Zahrt and Deretic, 2002). The same was observed inside

macrophages. Few potential antioxidant genes were induced in M. tuberculosis (Schnappinger et

al., 2003). These observations suggested a constitutive expression of the antioxidant genes usually

under the regulation of OxyR and SoxRS. Indeed this seems to be the case as shown recently by a

transcriptomic analysis of M. tuberculosis response to oxidative stress. Expression of katG and

Page 65: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

45 Introduction

ahpC was maintained at high basal levels (Voskuil et al., 2011). This might be an advantage for M.

tuberculosis and explain part of its success during host infection. However, even in the absence of

these classical regulators M. tuberculosis was capable of a large transcriptional response to H2O2

(Voskuil et al., 2011). Thus, other transcriptional regulators might be involved in the control of M.

tuberculosis oxidative stress responses.

IdeR, an iron-dependent transcriptional repressor, seems to play a role in M. tuberculosis

oxidative stress response (Rodriguez et al., 2002). Inactivation of IdeR in M. tuberculosis results in

increased sensitivity to oxidative stress. However, no difference in expression of genes involved in

oxidative stress protection was found between ideR mutant and the wild-type. Therefore, the

sensitive phenotype of the ideR mutant to oxidative stress might be due to an indirect effect,

caused by changes on iron levels, instead of a direct interference with gene expression. Iron

deficiency can lead to oxidative stress by decreasing the activity of iron-containing enzymes such as

superoxide dismutases and catalases. On the other hand, increased intracellular free iron,

participate in Fenton reactions, resulting in the enhanced generation of oxygen radicals and

oxidative stress.

Several sigma factors might also be involved in regulation of oxidative stress responses in

mycobacteria. M. tuberculosis sigH mutant is more susceptible than wild-type to oxidative stress

caused by H2O2 and other oxidative agents. In addition, SigH seems to regulate expression of

thioredoxin/thioredoxin reductase genes (Manganelli et al., 2002; Raman et al., 2001) as well as

sodA (Mehra and Kaushal, 2009). Other sigma factors (SigF, SigJ) might be also implicated in

mycobacterial oxidative stress responses, as mutants in these genes are more susceptible to

oxidative agents (Gebhard et al., 2008; Hu et al., 2004). However, no known antioxidant genes have

been identified as regulated by these sigma factors, suggesting that the contribution of these

transcriptional regulators might be indirect. The same happens for CarD, a recently identified

transcriptional regulator in M. tuberculosis, which expression is induced by H2O2 and depletion

leads to sensitivity to oxidative stress and other conditions (Stallings et al., 2009).

Even though the referred regulators seem to, somehow, contribute to oxidative stress response

regulation in mycobacteria, these are not specific for oxidative stress being also involved in the

regulation of other kind of stresses/responses. A very recent study has shown that OxyS might be

an oxidative stress response regulator in mycobacteria lacking OxyR (Li and He, 2012). OxyS is a

LysR-type transcriptional regulator – the most abundant type of transcriptional regulators in

prokaryotes involved in regulation of diverse functions including oxidative stress responses

(Maddocks and Oyston, 2008). In fact, it was previously shown that OxyS might play a role in M.

Page 66: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

46 Chapter 1

tuberculosis oxidative stress response, as increasing concentrations of OxyS lowered levels of AhpC

and made bacteria more sensitive to organic hydroperoxides. However, deletion of oxyS did not

affect expression of katG (Domenech et al., 2001), suggesting that this was not the main katG

regulator. Li et al. (2012) have shown though that M. tuberculosis OxyS binds to katG promoter,

and this binding is inhibited by H2O2. Furthermore, M. smegmatis overexpressing OxyS had a

reduced expression of katG and increased susceptibility to H2O2, suggesting that this was, indeed, a

transcriptional repressor of katG (Li and He, 2012). These characteristics of OxyS are similar to that

of FurA that also regulates expression of katG in M. tuberculosis (Sala et al., 2003). OxyS seems to

regulate not only katG but also ahpC (Domenech et al., 2001). Further studies need to be

performed to characterize the OxyS regulon and confirm if this could be a substitute regulator to

OxyR. In addition, RoxY was identified in M. smegmatis as a repressor of OxyS, suggesting that

oxidative stress regulation in mycobacteria that lack OxyR might be more complex than imagined.

Page 67: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

Objectives

The main goal of this thesis is to investigate M. tuberculosis defense mechanisms against

oxidative stress and their role in mycobacterial host adaptation and evolution. This particular kind

of studies are relevant given that M. tuberculosis resistance mechanisms against reactive oxygen

species (ROS) should contribute to the success this pathogen, however, their role in pathogenesis

remains poorly understood. On the other hand, DNA repair, replication and recombination (3R)

proteins are involved in the repair of the damages such as the ones induced by ROS, being

important players in maintaining genome integrity, however, 3R proteins can also be responsible

for genome variability and contribute to microbial adaptation and evolution, through allelic

variation of its genes. Given that certain M. tuberculosis families seem to have some selective

advantage over others, it is important to investigate the basis for their genetic variability.

To achieve this goal two research studies were performed:

A. Screening of a transposon mutant library for mutants with an impaired ability to resist to ROS

in order to address the following questions:

Which genes are identified as important for M. tuberculosis resistance against ROS?

How do these genes contribute to M. tuberculosis virulence by allowing its survival in host

cells?

B. Analysis of polymorphisms in 3R genes in strains belonging to a successful family of M.

tuberculosis to address the following questions:

Do we find 3R polymorphisms associated with this family?

Could some of these polymorphisms confer some selective advantage and contribute

to the evolutionary success of these strains?

The ultimate goal of this kind of studies is to identify potential virulence factors useful for the

development of new anti-tuberculosis drugs or vaccine candidates. It could serve the purpose of

identifying the strains that are more capable of evolving, and/or resisting treatment, which would

be particularly relevant, given the increase of M. tuberculosis drug-resistant strains.

Page 68: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga
Page 69: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

CHAPTER 2

Identification of M. tuberculosis virulence factors

involved in defense mechanisms against

oxidative stress

Olga Mestre1, Raquel Hurtado-Ortiz1, Tiago Dos Vultos1, Amine Namouchi1, Mena Cimino1,

Madalena Pimentel2, Olivier Neyrolles3,4, Brigitte Gicquel1

1

Unité de Génétique Mycobactérienne, Institut Pasteur, Paris, France ; 2

Centro de Patogénese Molecular,

Unidade dos Retrovírus e Infecções Associadas, Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal;

3 Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Toulouse,

France; 4

Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale,

Toulouse, France

Part of the data presented in this chapter is included in the manuscript: “High throughput phenotypic selection

of Mycobacterium tuberculosis mutants with impaired resistance to reactive oxygen species identifies genes

important for intracellular growth”. Mestre O, Hurtado-Ortiz R, Dos Vultos T, Namouchi A, Cimino M, Pimentel

M, Neyrolles O, Gicquel B (2012) PLoS One; Submitted

Page 70: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga
Page 71: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

51 M. tuberculosis resistance to ROS

ABSTRACT 2.1

Mycobacterium tuberculosis has the remarkable capacity to survive within the hostile

environment of the macrophage and to resist potent antibacterial molecules such as reactive

oxygen species (ROS). Thus, understanding mycobacterial defence mechanisms against ROS may

contribute to the development of new anti-tuberculosis therapies. Here we identified genes

involved in such mechanisms by screening a high-density transposon mutant library and showed

that most of these genes are involved in the intracellular lifestyle of the pathogen. Many of these

genes were found to play a part in cell envelope functions further strengthening the important role

of the mycobacterial cell envelope in protecting against aggressions such as the ones caused by

ROS inside host cells.

Page 72: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga
Page 73: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

53 M. tuberculosis resistance to ROS

INTRODUCTION 2.2

Part of the success of Mycobacterium tuberculosis lies in its ability to thrive inside macrophages,

in a very harsh environment, where bacteria are exposed to strong antimicrobial molecules such as

reactive oxygen species (ROS) (Flannagan et al., 2009). Therefore, defence mechanisms against ROS

are important for M. tuberculosis pathogenicity. Understanding the molecular basis of such

mechanisms will provide clues for the development of target-specific drugs or effective attenuated

strain-based vaccine candidates.

The generation of ROS is initiated by the phagocyte NADPH oxidase (Phox) that produces

superoxide anions. These anions have a low bactericidal potential, however, they rapidly dismutate

into H2O2, particularly at acid pH in the phagosome. H2O2 can produce hydroxyl radicals, the most

powerful oxidant, when reacting with iron in what is known as Fenton reaction(Friedberg et al.,

2006). These molecules are highly reactive agents, damaging proteins, lipids, carbohydrates and

nucleic acids (Bedard and Krause, 2007). The biological importance of ROS is clearly demonstrated

in patients with chronic granulomatous disease (CGD) characterized by a total absence or low level

of ROS, resulting from Phox genetic deficiency. These individuals are highly susceptible to bacterial,

fungal and yeast infections (Bedard and Krause, 2007) and increasing numbers of cases of

mycobacterial diseases, including tuberculosis, are being diagnosed in such patients (Bustamante

et al., 2007). Nonetheless, the significance of ROS in host defence against M. tuberculosis remains

controversial.

Despite the early demonstration that ROS were mycobactericidal (Walker and Lowrie, 1981),

years later it was shown that M. tuberculosis could be resistant to killing by oxidative stress in

macrophages (Manca et al., 1999). M. tuberculosis genes showing similarities with genes known to

be involved in ROS resistance in different bacterial species were identified (Cole et al., 1998; Zahrt

and Deretic, 2002). However, transcriptomic analysis showed that many of these genes are not

induced in oxidative conditions (Garbe et al., 1996; Schnappinger et al., 2003). This is explained by

a relatively high constitutive expression level of such genes (Voskuil et al., 2011) that, associated

with an absence of the classic oxidative stress response regulators (SoxR and OxyR) (Zahrt and

Deretic, 2002), leads to reduced induction ratios. This suggests that bacilli are continuously primed

for oxidative stress defense revealing once again the importance of this kind of responses in M.

tuberculosis. Still, evidence for a role in defence against ROS mainly comes from M. tuberculosis

katG mutant, with an alteration of both catalase and peroxidase activities, which was attenuated in

wild-type mice but grew normally in phox-deficient mice (Ng et al., 2004).

Page 74: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

54 Chapter 2

The construction of transposon mutant libraries in mycobacteria has enabled the newly

identification and study of several potential virulence factors (Camacho et al., 1999; Cox et al.,

1999; McAdam et al., 2002). So far, different screening-based approaches using mutant libraries

have been conducted to identify M. tuberculosis mutants sensitive to different conditions found

inside host macrophages, including acidic (Vandal et al., 2008) as well as nitric (Darwin et al., 2003)

stresses. With the aim to identify and investigate genes required for resistance against ROS in M.

tuberculosis, we screened, for the first time, a transposon mutant library to identify mutants with

impaired ability to resist oxidative stress.

Page 75: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

55 M. tuberculosis resistance to ROS

MATERIAL AND METHODS 2.3

2.3.1 Bacterial Strains and growth conditions

M. tuberculosis strains GC1237 (Caminero et al., 2001) and H37Rv were grown in Middlebrook

7H9 media (Difco) supplemented with ADC and 0,05% Tween 80, except when referred, or in

Middlebrook 7H11 media (Difco) supplemented with OADC and 0,5% glycerol for CFU analysis.

Mutant strains were grown in the presence of kanamycin 20 µg/ml and complemented strains

grew in the presence of both kanamycin and hygromycin 20 µg/ml and 50µg/ml, respectively.

Luminescent bacteria were obtained by transforming M. tuberculosis strains, by electroporation as

described (Parish and Stoker, 1998), with a luciferase-expressing plasmid derived from pSMT3

(Humphreys et al., 2006). Positive clones were selected with 50 µg/ml hygromycin and

luminescence emission from luciferase was confirmed in a luminometer.

2.3.2 Mutant library screening

Mutants of the M. tuberculosis GC1237 strain transposon library (Brodin et al., 2010) were

grown individually at 37ºC in 96-well plates in 7H9 medium for 3 weeks. Optical density (OD) was

measured, at 620nm, and mutants which presented growth defects at this phase were excluded

from subsequent analysis. For those mutants, which no growth defects were observed, aliquots

(10-20 µl) were sub-cultured in 200 µl 7H9 medium containing 9mM of H2O2 (Sigma-Aldrich). OD

was measured two to three weeks later in a plate spectrophotometer and the ratio of the OD value

of the mutant to that of the wild-type strains (growth index) was calculated. Mutants with a growth

index below 0.4 were considered as sensitive to H2O2 (Table 2).

2.3.3 Identification of transposon insertion sites

Genomic DNA from selected transposon mutants was extracted and the exact location of the

transposon insertion was determined by sequencing amplicons generated by ligation-mediated

inverse PCR (Cox et al., 1999). Briefly, 1-2 µg of DNA was digested with BssHII, a low-moderate

frequency cutting restriction enzyme that doesn’t cut the transposon, followed by heat inactivation

of the restriction enzyme and analysis in an agarose gel to check for complete digestion. An aliquot

of the digested DNA (<0,5ng) was diluted (1:10) and ligated overnight at 16ºC, followed by ethanol

precipitation. Using these products a PCR was done to amplify the DNA regions adjacent to the

transposon using TaKaRa La Taq DNA polymerase (TaKaRa, Shiga, Japan) with the primers

corresponding to the left (Tn_L) and the right (Tn_R) (Table 1) ends of the transposon. The

Page 76: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

56 Chapter 2

amplified products were sequenced using an ABI prism 3100 Genetic Analyser (Applied Biosystems)

with Big Dye Terminator v3.1 cycle sequencing Kit (Perkin Elmer Applied Biosystems, Courtaboeuf,

France). Sequences were mapped in M. tuberculosis H37Rv genome available at Tuberculist (Cole

et al., 1998). Transposon insertions within the mmpL9, moaD1, ppe54 and ppe56 genes were also

confirmed by Multiplex-PCR using a primer flaking the site of transposon insertion (genename_F)

with primers hybridizing inside the transposon (Tn_L and Tn_R2) (Table 1).

Table 1. List of oligonucleotides (5’-3’) used in this study.

Primer name Sequence

Tn_L GTCGGCCATTAGCTTCT

Tn_R CACCACCGATCCTCAT

Tn_R2 GCACCACCGATCCTCAT

mmpL9_F GCATCAGAACCTATGTCACCG

mmpL9_R GATCGCATGGTCAACATCA

moaD1_F CGCTGTCAGCGTCACTATC

ppe54_F GGTATCGGCAATGTAGGTACTCA

ppe56_F CATCCTCATCGGCGATA

pMV_F ATTCTAGAGCATCATCCTCCCACGAC

pMV_R ATAAGCTTTATCAACCCCGGTGCAGAT

2.3.4 Complementation of mmpL9 and moaD1 mutants

To genetically complement the mmpL9 mutant, the mmpL9 gene with its putative promoter

region from the wild-type strain M. tuberculosis GC1237 or H37Rv, was introduced in the

integrative vector pMV306, conferring hygromycin resistance (Lee et al., 1991). The

complementation construct was generated by amplifying the mmpL9 gene along with 629bp

upstream intergenic region from GC1237 or H37Rv chromosomal DNA, using Phusion High-fidelity

DNA polymerase (Finnzymes), with the primers pMV_F and pMV_R (Table 1), containing the

restriction sites of XbaI and HindIII, respectively. The amplified product was gel purified, digested

with XbaI and HindIII, and cloned between XbaI and HindIII sites in pMV306. The resulting plasmid

was sequenced to confirm that no mutations were present, and transformed into the mutant by

electroporation as described (Parish and Stoker, 1998). Transformed bacteria were recovered using

hygromycin and kanamycin selection (50 µg/ml both) and the presence of the wild-type gene was

confirmed by PCR using primers mmpL9_F and mmpL9_R (Table 1).

Page 77: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

57 M. tuberculosis resistance to ROS

The moaD1 mutant was complemented using the pYUB412-derived cosmid (Bange et al., 1999)

I528, which carries a DNA fragment covering the 3454 to 3485 Kb region of the M. tuberculosis

H37Rv chromosome and a hygromycin resistance cassette. Bacteria were transformed and selected

as described for mmpL9 mutant complementation.

2.3.5 Susceptibility to oxidative stress, starvation and acidic pH

Bacteria were collected from cultures, washed twice with PBS and clumps were dissociated by

needle passages, as performed for macrophage infection. To assess the killing effect of H2O2,

single-cell suspensions were adjusted to approximately 1x106 CFU/ml in 7H9 medium without ADC.

Then, bacteria were exposed to 9mM H2O2 for 4 hours at 37ºC. Control untreated bacteria were

also included. Dilutions were made, plated and CFUs were counted after 3 weeks growth at 37ºC.

Percentage of survival was calculated by dividing the number of H2O2 treated CFUs by the number

of untreated CFUs and multiplying by 100. For growth rate analysis, bacteria were grown in 7H9

medium at 37ºC in the presence of increasing concentrations of H2O2 (4mM, 6mM, 9mM, 12mM

and 15mM), or at pH 4.5, and OD values were measured, at 600nm, every three days, during 2 to 3

weeks.

Susceptibility to complete starvation was determined essentially as previously described (Parish,

2003). Briefly, single-cell suspensions were adjusted to approximately 1x106CFUs/ml in sterile

distilled water and incubated at 37ºC. Aliquots were taken 5 and 10 days after, dilutions were

made and plated, and CFUs were analysed 3 weeks later. Percentage of survival was calculated by

dividing the number of CFUs at day 5 or 10 by the number of CFUs at time zero and multiplying by

100.

2.3.6 Human monocyte isolation for subsequent macrophage generation

Freshly collected buffy coats were obtained in accordance with institutional requirements and

written consent from healthy donors. PBMC fraction was collected using a Ficoll gradient (Ficoll-

PaqueTM Plus, GE Health Care) and monocytes were purified by positive selection using anti-CD14

conjugated magnetic microbeads (Milteny Biotec) and Quadro MACS Separation System (Milteny

Biotec) according to manufacturer’s protocol. Monocytes were distributed into 6-well plates and

allowed to adhere in a 5% CO2 incubator at 37°C in 3ml of RPMI-1640 (Sigma-Aldrich) containing 2

mM of L-glutamine (Gibco), 10% of heat inactivated foetal bovine serum (PAN Biotech) and 50

ng/ml of cytokine rhM-CSF (recombinant human macrophage colony stimulating factor) (R&D

Page 78: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

58 Chapter 2

Systems) to differentiate into macrophages. Cultures were fed every 2 days with this medium

containing full doses of cytokine during 6 days.

Before infection, macrophages were recovered using a non-enzymatic cell dissociation solution

(Sigma-Aldrich), counted and adjusted to 2x105 per well in 24-well plates, where they were allowed

to adhere.

2.3.7 Macrophage Infections

Bacteria were grown in 7H9 medium without Tween at 37°C. In the infection day, bacteria were

recovered, washed two times and resuspended in 1 ml PBS. Clumps were disassociated by 30

passages through a 0,8x40 mm needle and 50 passages though a 0,45x10 mm needle (3 times). OD

was measured, at 600 nm, and correlated with bacterial number in order to adjust the multiplicity

of infection (MOI) used for infection. The medium was removed from blood-monocyte derived

macrophages and replaced with 500µl of bacterial suspensions in RPMI-1640, containing 2 mM of

L-glutamine and 10% of heat inactivated foetal bovine serum. After 4 hours of infection, medium

was removed along with non-ingested bacteria and cells were incubated with fresh medium. At

each time point post-infection, macrophages were lysed, dilutions were made and samples were

plated in 7H11 Middlebrook agar. After 3 weeks growth at 37ºC, bacteria were quantified by CFUs

analysis. When using luminescent M. tuberculosis, bacteria were quantified immediately after

macrophage lysis, in a luminometer. Each infection experiment was carried out using triplicates for

each condition/strain.

When using NADPH oxidase inhibitors, macrophages were incubated for 1 hour with 100µM

Apocynin (Calbiochem) and 10µM Diphenyleneiodonium (DPI) (Sigma-Aldrich) prior to infection.

Dimethyl sulfoxide (DMSO) was added to cultures at 0.1% (vol/vol) as a solvent control. The ROS

scavenger 4-Hydroxy-TEMPO (0.1 and 0.2 mM; Sigma-Aldrich) as well as superoxide dismutase

(55U/ml; Sigma-Aldrich) were added during the 4 hour infection period and washed away from

cells along with non-ingested bacteria. To activate macrophages, 1α,25-Dihydroxyvitamin D3 (10-8,

10-7 or 10-6 M; Sigma-Aldrich), the active form of vitamin D3, was added to cells after the 4 hour

infection period and left in the medium during the entire post-infection period. Ethanol 95% was

added to cultures at 0.1% (vol/vol) as a solvent control for Vitamin D3. In these experiments

macrophages were infected with M. tuberculosis strains expressing luciferase.

Page 79: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

59 M. tuberculosis resistance to ROS

2.3.8 Superoxide Anion detection in human macrophages

Superoxide production by macrophages was measured using the LumiMax Superoxide Anion

Detection kit (Agilent Technologies). Monocyte derived-macrophages (2x105) were incubated with

apocynin (50 and 100µM) and DPI (1 and 10 µM) for 1 hour at 37ºC, 5% CO2. Then, macrophages

were collected and resuspended in the SOA assay medium, provided with the kit. A solution

containing luminol, an enhancer, and PMA (Phorbol 12-myristate 13-acetate;Sigma), a NADPH-

oxidase stimulator, or M. tuberculosis, was added to macrophages following manufacturer’s

instructions. The chemiluminescence resulting from oxidation of luminol by the superoxide anion

was then detected using a luminometer.

2.3.9 Analysis of polymorphisms in mmpL genes in M. tuberculosis genomes

Twenty-four genomes, three recently sequenced by our lab (Namouchi et al., 2012) and 21

publicly available, from major M. tuberculosis lineages were analysed to identify SNPs in the 13

mmpL genes present in M. tuberculosis genomes (Cole et al., 1998). This was performed as

described by Namouchi et al. (2012).

2.3.10 Statistics

Statistical significance was determined using the Student’s two-tailed, unpaired, t-test. Analyses

were performed using the GraphPad Prism software (GraphPad Software, CA, USA).

2.3.11 Ethics Statement

Human monocytes were purified from buffy coats obtained from healthy blood donors

(Etablissement Français du Sang, EFS, Rungis). All donors participating to this study had given

their written consent under a EFS contract C- CPSL UNT N°09/EFS/062, following articles L1243-

4 and R1243-61 of the French Public Health Code and approved by the French Ministry of

Science and Technology. Supply and handling of human red cells followed the guidelines of the

agreement between Institut Pasteur, EFS and the regulation of blood donation in France. As a

“collection” is defined by Decree 2007-1220 as “a collection, for scientific use, of biological

samples from a group of persons who have been identified and selected on the basis of clinical

or biological characteristics of one or several members of the group”, and as the blood samples

used in our study were not obtained from individuals that have been “identified or selected on

the basis of clinical or biological characteristics”, these samples do not constitute a so-called

Page 80: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

60 Chapter 2

“collection”, which waives the need for ethical approval by the ethical committee “Comité de

Protection des Personnes”, in agreement with Decree 2007-1220, article R1243-63.

Page 81: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

61 M. tuberculosis resistance to ROS

RESULTS 2.4

2.4.1 Screening for mutants with increased susceptibility to oxidative stress

In order to optimize the conditions to screen a mutant library for mutants with increased

susceptibility to H2O2, we tested a small set of DNA repair mutant strains. These were knockout

mutants in mutT1, mutM, and mutY genes, involved in handling oxidized guanine, an important

DNA lesion caused by ROS (Demple and Harrison, 1994). Analysis of growth rates in the presence of

increasing concentrations of H2O2 show that 9 mM H2O2 totally inhibited the mutY mutant growth,

while the wild-type (H37Rv), as well as the mutT1 and mutM mutant strains, were still able to grow

(Figure 1). At 12mM H2O2 growth was totally inhibited for all strains (Figure 1). The mutY mutant

presented a slight growth defect in the absence of H2O2 that could also be due to sensitivity to

oxidative stress caused by endogenous ROS resulting from aerobic respiration (Imlay, 2008; Storz

and Imlay, 1999). These results show that the mutY mutant has an increased susceptibility to H2O2

and allowed us to define the conditions to use in our screening.

Figure 1. Growth curves of M. tuberculosis H37Rv, ΔmutT1, ΔmutM and ΔmutY in increasing

concentrations of hydrogen peroxide. Data represent means of triplicates in a representative

experiment.

Page 82: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

62 Chapter 2

The difference observed between the mutY, mutT1 and mutM mutants might be explained by

the fact that M. tuberculosis possesses multiple homologs of mutT and mutM in the genome, while

a single mutY homolog has been annotated in mycobacterial genomes (Cole et al., 1998). For

example, mycobacterial MutT1 and MutT2 seem both to have 8-oxo-GTP hydrolase activity (Dos

Vultos et al., 2006).

Next, we screened a transposon mutant library constructed in a virulent M. tuberculosis clinical

isolate (GC1237) of the Beijing/W family (Brodin et al., 2010). M. tuberculosis GC1237 growth in the

presence of H2O2 was similar to H37Rv (Figure 2). Sensitive mutants were identified by comparing

their OD values to the ones of the wild-type after 3 weeks growth in the presence of 9 mM H2O2.

The analysis of approximately 6000 mutants allowed the selection of 18 H2O2 sensitive mutants

corresponding to 11 different genes and two intergenic regions identified by sequencing the

transposon insertion sites after inverse PCR (Table 2). No significant growth defect was observed

for these mutants in the absence of H2O2, indicating that their sensitivity to H2O2 is specific (Table

2).

Figure 2 - Growth curves of M. tuberculosis GC1237 in increasing concentrations of hydrogen

peroxide. Data represent means of triplicates in a representative experiment.

2.4.2 Sensitive mutants to oxidative stress are affected in cell envelope functions

Interestingly, many of the sensitive mutants to oxidative stress we have identified harbour defects in

genes involved in M. tuberculosis cell envelope functions (Table 2). These include four different ppe

genes, that code for cell surface proteins, present almost exclusively in pathogenic mycobacteria,

thought to be involved in virulence (Akhter et al., 2012), as well as an ABC transporter (Rv0986)

implicated either in the transport of an adherence factor or in mycobacterial cell wall architecture and

integrity (Rosas-Magallanes et al., 2007).

Page 83: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

63 M. tuberculosis resistance to ROS

Table 2. List of identified sensitive mutants to oxidative stress using hydrogen peroxide.

Cell envelope associated function Insertion sequences and phages

Intermediary metabolism and respiration Unknown

Conserved hypotheticals

aAnnotations are from tuberculist http://genolist.pasteur.fr/TubercuList/.

b, c Growth index is the ratio of the optical density value of the mutant to that of the wild-type strain after 3 weeks

growth in the absence (b) or presence (c) of H2O2 9mM. Mutants with a growth index below 0.4 were selected as sensitive to H2O2.

*Tn inserted 41 bp upstream the gene start codon.

Mutant id

Gene (transposon insertion site)

Product Putative function

a Growth Index

b

Growth Index H2O2

c

Reference

118B7 Rv0305c (2701:2702)

PPE6 Unknown

0,99 0.35 This study

118F3 Rv0986 (430:431)

ABC transporter Thought to be involved in active transport of adhesion component across the membrane

1,04 0.24 (Fontan et al., 2008; Pethe et al., 2004;

Rosas-Magallanes et al., 2007)

100C5 Rv1548c (1125:1126)

PPE21 Unknown

0.85 0.38 (Voskuil et al., 2004)

98F4 Rv2339(1290:1291)

Transmembrane transport protein MmpL9 Thought to be involved in fatty acid transport

0.80 0.13 (Domenech et al., 2005; Lamichhane et al., 2005;

MacGurn and Cox, 2007)

100D7 Rv2339 (957:958)

Transmembrane transport protein MmpL9 Thought to be involved in fatty acid transport

1,02 0.31 (Domenech et al., 2005; Lamichhane et al., 2005;

MacGurn and Cox, 2007)

117C10 Rv2339 (812:813)

Transmembrane transport protein MmpL9 Thought to be involved in fatty acid transport

0,95 0.23 (Domenech et al., 2005; Lamichhane et al., 2005;

MacGurn and Cox, 2007)

104D8 Rv3343c (2758:2759)

PPE54 Unknown

1.13 0.31 (Brodin et al., 2010; Voskuil et al., 2004)

115C10 Rv3343c (5907:5908)

PPE54 Unknown

1,01 0.32 (Brodin et al., 2010; Voskuil et al., 2004)

99G11 Rv3350c (1441:1442)

PPE56 Unknown

0.93 0.23 (Voskuil et al., 2004)

115B9 Rv3350c (5059:5060)

PPE56 Unknown

1,03 0.27 (Voskuil et al., 2004)

115C9 RV3350c (5059:5060)

PPE56 Unknown

0,98 0.30 (Voskuil et al., 2004)

115C4 Rv3112 (49:50)

Molybdenum cofactor biosynthesis protein MoaD1 Involved in molybdenum cofactor biosynthesis

0,92 0.38 (Brodin et al., 2010)

100E6 Rv1507c (591:592)

Conserved hypothetical protein Unknown

0.95 0.36 (Brodin et al., 2010)

111E12 Rv3594 (774:775)

Conserved hypothetical protein Unknown

0,74 0.09 This study

96F9 Rv1509 (637:638)

Hypothetical protein Unknown

0,89 0.34 This study

100D1 Rv2337c (-41)* Hypothetical protein Unknown

1,05 0.15 This study

106H11 Rv3430c (1049:1050)

Possible transposase Involved in the transposition of the insertion sequence IS1540

0,75 0.20 (Dutta et al., 2010)

98G4 Rv2480c-Rv2481c (2786248:2786249)

Possible transposase for insertion sequence element IS6110 (fragment)-Hypothetical protein

0.76 0.14 -

Page 84: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

64 Chapter 2

Strikingly, we could independently isolate three different mutants in the mmpL9 gene, which

suggests an important role for MmpL9 in M. tuberculosis response to oxidative stress (Table 2).

MmpLs (mycobacterial membrane protein large) are thought to be involved in lipid synthesis

and/or transport (Tekaia et al., 1999).

Some genes with no apparent known function were also identified in our screen (Table 2).

However, rv1507c belongs apparently to a cluster of genes organized in an operon (rv1503c-

rv1507c), involved in di-O-acyl-trehalose (DAT) lipid synthesis (Brodin et al., 2010). The rv1509 gene

seems to form an operon along with rv1508a. BLAST analysis identified methyltransferase domains

in the Rv1509 protein. Rv1508a shows homology with GDP-D-Mannose-Dehydratase (GMD). GMD

is involved in the production of L-fucose from GDP-D-mannose. Fucose is part of PGLs found in

Beijing/W strains. However, other GMP has been identified that seems to be already involved in

this pathway (Malaga et al., 2008). Thus, whether Rv1508a along with Rv1509 are involved in lipid

synthesis is unclear. Rv3594, another protein with unknown function found in our screen, shows

similarities with an N-acetylmuramoyl-L-alanine-amidase, a peptidoglycan hydrolase.

Peptidoglycan hydrolases participate in bacterial cell wall growth and regulation as well as in

different lysis phenomena (Vollmer et al., 2008).

We have also identified a few genes not associated with M. tuberculosis cell envelope functions.

It was the case of moaD1, recently confirmed as involved in the biosynthesis of the molybdenum

cofactor (MoCo) in M. tuberculosis (Williams et al., 2011).

Curiously, a mutant in a putative transposase (Rv3430c) was selected as sensitive to oxidative

stress. A significant proportion of insertion element genes including transposases were also found

to be highly induced in M. tuberculosis after exposure to several DNA-damaging agents, including

H2O2 (Boshoff et al., 2003). However, the correlation between these mobile genetic elements and

oxidative stress or DNA damage/repair is not clear. This gene was also found to be important for

M. tuberculosis survival in the lungs of non-human primates (Dutta et al., 2010), suggesting a role

for Rv3430c in vivo.

2.4.3 Quantification of sensitivity of mutants to oxidative stress

The sensitive phenotype of our newly identified mutants to oxidative stress was further

analyzed and quantified on the individual level. This was done by analyzing the killing effect of

H2O2 in several of those mutants possibly affected in cell envelope components including, two PPE

mutants, ppe54 and ppe56, and an mmpL9 mutant, as well as for the moaD1 mutant possibly

implicated in other kind of defense mechanism against oxidative stress. As several mutants,

Page 85: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

65 M. tuberculosis resistance to ROS

carrying independent transposon insertions, were identified we have randomly chosen a ppe54

(104D8), a ppe56 (115B9) and an mmpL9 (100D7) mutant (Table 2).

The results represented in Figure 3 show that all mutant strains, except ppe54, have an

increased sensitivity to killing by H2O2 when compared to their wild-type counterpart. We observed

a decrease in survival ranging from around 40% (104D8, ppe56 mutant) to nearly 80% (100D7,

mmpL9 mutant), while the killing rate for the wild-type strain was less than 20% (Figure 3). There

was no difference in colony size or morphology of the mutants when compared to the wild-type

strain in the absence of H2O2 (data not shown). These results confirm the sensitive phenotype of

these strains to oxidative stress in the presence of H2O2 and reveal its bactericidal effects.

Figure 3. Susceptibility of transposon mutants to oxidative stress. Percentage of survival of the M.

tuberculosis 100D7 (mmpL9), 115C4 (moaD1), 104D8 (ppe54), 115B9 (ppe56) transposon mutants and

wild-type (GC1237) strains was determined by comparing bacteria treated with 9mM of hydrogen

peroxide to untreated controls. Data and error bars represent means and standard deviations of the

results from triplicates in a representative experiment carried out three times with similar results.

Statistically significant differences were calculated using the Student’s two-tailed, unpaired, t-test, and

are indicated by asterisks *P<0,05; ***P<0,001; ****P<0,0001.

The difference in sensitivity to H2O2 between the ppe54 mutant and the wild-type strain was not

statistically significant. This suggests that the effect of H2O2 on this strain is not bactericidal but

probably only bacteriostatic as we observed growth inhibition in the presence of H2O2 during the

screen (Table 2). Additionally, two different mutants, carrying independent transposon insertions

Page 86: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

66 Chapter 2

in the ppe54 gene, were selected (Table 2), confirming the sensitivity to oxidative stress of any

strain carrying a mutation in this gene.

2.4.4 MmpL9 and MoaD1 are important for M. tuberculosis survival in human

macrophages

Because bacteria have to face oxidative stress conditions inside host cells we decided to analyze

growth in human macrophages of the moaD1 and mmpL9 mutants which were the most sensitive

strains to killing by H2O2 (Figure 3). Additionally, MmpLs have been identified as important

determinants of M. tuberculosis virulence (Camacho et al., 1999; Converse et al., 2003; Cox et al.,

1999; Domenech et al., 2005), even though reports regarding mmpL9 mutant phenotype in mice

are contradictory (Cox et al., 1999; Domenech et al., 2005; Lamichhane et al., 2005; MacGurn and

Cox, 2007). Genes involved in the same biosynthetic pathway as moaD1 have been previously

shown to be important for bacterial intracellular lifestyle (Rosas-Magallanes et al., 2007),

suggesting an important role for MoCo in M. tuberculosis survival inside host cells.

Monocyte-derived macrophages were infected with mutant, wild-type and complemented

strains and after six days post-infection cells were lysed and bacteria were plated to analyze CFUs.

As shown in Figure 4, both moaD1 and mmpL9 (100D7) mutants displayed an impaired intracellular

growth when compared to their wild-type counterpart (Figure 4A and B). At day 6 post-infection,

the moaD1 mutant showed a 4-fold increase in intracellular growth while the wild-type showed a

12-fold increase, with respect to time-point zero. Complementation of the moaD1 mutant using an

integrative vector restored the wild-type phenotype inside macrophages (Figure 4A). These results

suggest that MoCo might be indeed important for mycobacterial growth inside host cells.

Infection of human macrophages with the mmpL9 mutant resulted in an increase of 2.5-fold

while the wild-type had a 15-fold increase in CFUs, after 6 days (Figure 4B). Sequence analysis of

the mmpL9 gene of the wild-type strain GC1237 revealed, when compared to H37Rv, the presence

of one nucleotide deletion at position 218. To better understand the possible effects of this

frameshift mutation, we decided to complement the mmpL9 mutant with mmpL9 of either GC1237

or H37Rv. As shown in Figure 4B, complementation restored the wild-type phenotype inside

macrophages using mmpL9 of GC1237. However, it seems to be only partially restored when

complemented with H37Rv mmpL9.

Page 87: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

67 M. tuberculosis resistance to ROS

Figure 4. Intracellular growth of mmpL9 and moaD1 mutants in human macrophages. Monocyte-

derived macrophages where infected with wild-type, moaD1 (A) or mmpL9 (B) mutant and

complemented (COMP) strains, at an MOI of 1:100 (bacteria per macrophage). At time point zero (0)

and six days of infection, cells were lysed and bacteria were enumerated. Data and error bars represent

means and standard deviations of the results from triplicates in a representative experiment carried out

three times with similar results. Statistically significant differences were calculated using the Student’s

two-tailed, unpaired, t-test, and are indicated by asterisks *P<0,05; **P<0,01;***P<0,001.

Unfortunately, we could not detect, using Western blotting, expression of either H37Rv or

GC1237 mmpL9 carrying a hemagglutinin tag, under the control of its own or of a strong promoter

(hsp60), in either M. tuberculosis or M. smegmatis, using different growth conditions and whole

cell lysate preparation methods (data not shown).

2.4.5 The mmpL9 deletion at position 218 is conserved in Beijing/W strains

To investigate the possible effects of the deletion identified in mmpL9 for the GC1237 strain, we

searched for SNPs in the 13 M. tuberculosis mmpL genes (Cole et al., 1998) in a collection of strains

representing main M. tuberculosis lineages. Statistical analysis of the identified SNPs suggests that

Page 88: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

68 Chapter 2

the mmpL9 gene does not accumulate more polymorphisms than the other mmpLs (Table 3),

excluding the hypothesis of pseudogene formation but not that of an effect, of the observed

polymorphisms, on protein’s functions.

Table 3. Statistical analysis of SNP density in mmpL genes in a collection of M. tuberculosis genomes.

Name Length (bp)

SNPs Binomial testa

Bonferroni correctionb

mmpL1 2877 3 0,926187042 12,96661859

mmpL2 2907 9 0,167599855 2,346397974

mmpL3 2835 13 0,008366836 0,117135699

mmpL4 2904 6 0,565387826 7,915429562

mmpL5 2895 7 0,406734347 5,694280858

mmpL6 1194 6 0,026461116 0,370455623

mmpL7 2763 5 0,670824515 9,391543207

mmpL8 3270 9 0,270440559 3,786167821

mmpL9 2889 7 0,404469247 5,662569455

mmpL10 3009 2 0,980458231 13,72641523

mmpL11 2901 5 0,716449242 10,03028939

mmpL12 3441 11 0,128927703 1,804987847

mmpL13a 912 4 0,071781264 1,004937698

mmpL13b 1413 1 0,862276271 12,07186779

Total 36210 88

aThe binomial test was used to identify mmpL genes that accumulate more SNPs than expected. The p-value was

calculated according to the number of SNPs and length of each gene and the total number of SNPs and genes length. bThe final p-value was estimated after Bonferroni correction.

Additionally, analysis of polymorphisms in mmpL9 in these collection of strains have shown that

the deletion in nucleotide 218 is specific to all modern and ancestral Beijing/W strains, being

absent from early ancestral Beijing/W strains, such as 94_M4241A, and strains of any other M.

tuberculosis family/lineage (Figure 5).

Page 89: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

69 M. tuberculosis resistance to ROS

Figure 5. Graphical representation of the distribution of polymorphisms in mmpL9 in several M.

tuberculosis genomes. Polymorphisms in mmpL9 were analyzed in 24 different M. tuberculosis

genomes (Namouchi et al., 2012) that correspond to the following groups/lineages according to

Gagneux’s classification (Gagneux et al., 2006a): 1-8: Beijing/W strains; 9-16: Euro-American strains; 17-

20: Indo-oceanic strains; 21-22: West-African strains. Black vertical lines correspond to SNPs. Gap

correspond to deletion in position 218 identified in all modern and ancestral (2-8) but not in early

ancestral (1) Beijing/W strains.

2.4.6 MmpL9 and MoaD1 are required to counteract other forms of stress existent in

macrophages

The wild-type phenotype was partially achieved for both moaD1 and mmpL9 complemented

strains in susceptibility tests using H2O2 (Figure 6). The reasons behind these observations are not

understood. It could be due to differences in the fine tuning of expression of these genes once they

are placed outside their normal chromosomal context. Nonetheless, the mmpL9 and moaD1 genes

should have a function in M. tuberculosis oxidative stress, otherwise complementation would not

have restored the wild-type phenotype at all. These results were unexpected once

complementation totally restored the wild-type phenotype in macrophages (Figure 4). To

investigate if the attenuated growth of the mmpL9 and moaD1 mutants in macrophages was due,

at least in part, to their sensitivity to ROS, we’ve analysed their phenotype in macrophages treated

with Phox-inhibitors.

Page 90: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

70 Chapter 2

Figure 6. Survival of mmpL9 and moaD1 mutant and complemented strains exposed to hydrogen

peroxide. Percentage of survival of M. tuberculosis wild-type (GC1237), moaD1 (A) and mmpL9 (B)

mutant and complemented (COMP) strains was determined by comparing bacteria treated with 9mM

of hydrogen peroxide to untreated controls. Data and error bars represent means and standard

deviations of the results from triplicates in a representative experiment. Statistically significant

differences were calculated using the Student’s two-tailed, unpaired, t-test, and are indicated by

asterisks ***P<0,001; ****P<0,0001.

Macrophages were treated with apocynin and diphenyleneiodonium (DPI), the most common

used NADPH inhibitors (Flannagan et al., 2009). To analyse if Phox could be effectively inhibited

using these compounds we measured superoxide production in these cells. Almost 90% reduction

in superoxide production using apocynin (100µM) and more than 70% using DPI (10µM) was

observed (Figure 7A). These compounds did not seem to significantly affect the viability of

macrophages as assessed by trypan blue exclusion (Figure 7B).

Page 91: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

71 M. tuberculosis resistance to ROS

Figure 7. Macrophages treated with the NADPH oxidase inhibitors, apocynin and DPI.

Macrophages (~2x105) were treated for 1h with DPI (1, 10µM) and apocynin (50, 100 µM). Untreated

macrophages were used as control. Superoxide production was measured by quantifying

chemiluminescence resulting from oxidation of luminol in a luminometer (RLUs - Relative light units),

after stimulation of macrophages with PMA (A). Effect on cell viability was determined by counting

cells unstained (viable) with trypan blue. DMSO was included as a solvent control. No statistical

significant differences between cells treated with inhibitors and untreated cells were observed (B).

Data and error bars represent means and standard deviations of the results from triplicates.

Macrophages were treated with the inhibitors and infected with luminescent M. tuberculosis

mmpL9 and moaD1 mutants, as well as the wild-type strain. As shown in figure 8, no statistically

significant differences were observed between untreated and inhibitor-treated macrophages

infected with either mutants or the wild-type strain (Figure 8). We realized, however, that infected

macrophages treated with DPI had a different morphology compared to untreated macrophages,

when observed under the light microscope (data not shown) even though this compound didn’t

seem to significantly affect macrophage viability (Figure 7B). This could be probably because DPI is

a nonspecific inhibitor of many different electron transporters (Bedard and Krause, 2007). Given

that we did not know which consequences this could have on intracellular bacterial survival, DPI

was excluded from the subsequent experiments.

Page 92: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

72 Chapter 2

Figure 8. Intracellular growth of mmpL9 and moaD1 mutants in human macrophages treated with

Phox inhibitors. Monocyte-derived macrophages (~2x105) where treated with apocynin (100µM) and

DPI (10µM) prior to infection with luminescent wild-type (GC1237), moaD1 or mmpL9 mutant strains, at

an MOI of 1:10 (bacteria per macrophage). DMSO was used as a solvent control. After 3 days (D3) of

infection cells were lysed and bacteria were quantified by detecting luminescence in a luminometer

(RLUs - Relative light units). Data and error bars represent means and standard deviations of the results

from triplicates in a representative experiment carried out three times with similar results.

Vitamin D3-activated macrophages have an increased antimycobacterial activity that seems to

be mediated, at least in part, by superoxide produced by Phox (Sly et al., 2001). By increasing ROS

production we could possibly increase the difference between treated and untreated macrophages

with Phox inhibitors, in case there is one. We did indeed observe a reduction in bacterial load

(Figure 9A) as well as an increased superoxide production (Figure 9B) in macrophages activated

with 1α,25-Dihydroxyvitamin D3, the active form of vitamin D3, infected with M. tuberculosis

GC1237. Thus, we decided to use vitamin D3-activated macrophages to analyse the phenotype of

the mmpL9 and moaD1 mutants in the presence of phox inhibitors.

Page 93: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

73 M. tuberculosis resistance to ROS

Figure 9. Macrophages activated with Vitamin D3. Monocyte-derived macrophages were treated with

1α,25-Dihydroxyvitamin D3 (10-6M), the active form of vitamin D3, and superoxide production was

measured by quantifying chemiluminescence resulting from oxidation of luminol in a luminometer

(RLUs - Relative light units), after stimulation with M. tuberculosis GC1237 (A). Macrophages were

infected with luminescent M. tuberculosis GC1237 at an MOI of 1:10 (bacteria per macrophage), and

incubated with 1α,25-Dihydroxyvitamin D3 (10-8, 10-7,10-6M). After 3 days of infection, cells were lysed

and bacteria were quantified by detecting luminescence in a luminometer (RLUs - Relative light units).

Data and error bars represent means and standard deviations of the results from triplicates in a

representative experiment carried out two times with similar results. Statistically significant differences

were calculated using the Student’s two-tailed, unpaired, t-test, and are indicated by asterisks *P<0,05;

***P<0,001; ****P<0,0001 (B). Untreated macrophages were used as control. Ethanol was used as a

solvent control.

Given that the ROS scavengers TEMPO, and the antioxidant enzyme superoxide dismutase (SOD)

were found to abrogate Vitamin D3 antimycobacterial effect (Sly et al., 2001), we used these

compounds, along with apocynin, to inhibit ROS production. Infected macrophages treated with

apocynin, TEMPO, as well as SOD, were incubated with vitamin D3 and bacteria were quantified by

detecting luminescence. The results presented in Figure 10 show a bacterial growth augmentation,

for the mutants as well as the wild-type strain, in macrophages treated with TEMPO, when

compared to untreated cells. However, this increase was similar in all strains, which means no

difference between mutants and wild-type. No statistically significant difference was observed in

bacterial growth, for any of the strains, in apocynin or SOD treated macrophages.

Page 94: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

74 Chapter 2

Figure 10. Intracellular growth of mmpL9 and moaD1 mutants in VitaminD3-activated

macrophages treated with Phox inhibitors. Monocyte-derived macrophages (~2x105) where treated

with apocynin (100µM), superoxide dismutase (SOD) (55U/ml) and TEMPO (T ) (0.1 and 0.2 mM),

infected with luminescent M. tuberculosis wild-type (GC1237), moaD1 or mmpL9 mutant strains, at an

MOI of 1:10 (bacteria per macrophage), and incubated with 1α,25-Dihydroxyvitamin D3 (5x10-7M).

Untreated macrophages were used as control. Ethanol and DMSO were used as a solvent control for

vitaminD3 and apocynin, respectively. After 3 days of infection, cells were lysed and bacteria were

quantified by detecting luminescence in a luminometer (RLUs - Relative light units). Data and error bars

represent means and standard deviations of the results from triplicates in a representative experiment

carried out two times with similar results. Statistically significant differences were calculated using the

Student’s two-tailed, unpaired, t-test, and are indicated by asterisks *P<0,05; **P<0,01; ***P<0,001.

Page 95: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

75 M. tuberculosis resistance to ROS

These results suggest that mmpL9 and moaD1 genes are required to protect M. tuberculosis

against other kind of stress found inside macrophages. Therefore, we tested susceptibility of these

mutants to other conditions such as starvation and acidic pH. Due to technical issues we couldn’t

obtain results for the moaD1 mutant in acidic conditions. Preliminary results showed no

statistically significant difference in mutant’s fitness, when compared to the wild-type, in the

conditions tested (Figure 11). Unfortunately, we didn’t have the opportunity to test for

susceptibility to reactive nitrogen species (RNS), another group of antimicrobial molecules

produced by macrophages to destroy intracellular bacteria (Flannagan et al., 2009).

Figure 11. Susceptibility of mmpL9 and moaD1 mutants to acidic pH and starvation. Growth curves

of M. tuberculosis GC1237, and mmpL9 mutant in 7H9 medium at pH7 or 4.5 (A). Susceptibility of

mmpL9 and moaD1 mutants to starvation was determined by quantifying CFUs after 5 and 10 days of

incubation in sterile water at 37ºC. Percentage of survival was determined by comparing CFUs at the

referred time points with CFUs at time-point zero, before incubation (B).

Page 96: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

76 Chapter 2

Page 97: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

77 M. tuberculosis resistance to ROS

DISCUSSION 2.5

The role of ROS during infection with M. tuberculosis remains unclear. Therefore, we decided to

look for genes involved in M. tuberculosis oxidative stress response using, for the first time, a

functional genomic approach. By screening a transposon mutant library we found a high number of

mutants sensitive to H2O2 that were affected in genes involved in cell envelope functions. M.

tuberculosis cell envelope is an important permeability barrier involved in bacterial protection

against different kinds of aggression (Brennan and Nikaido, 1995). This has been continuously

shown. For example, a high percentage of genes identified as responsible for M. tuberculosis

resistance to acidic stress during a genetic screening, were also found to be involved in bacteria’s

cell wall functions (Vandal et al., 2008). However, few studies have previously described cell

envelope components/mechanisms as involved in M. tuberculosis defense against ROS. Our results

reveal the important role of M. tuberculosis cell envelope in protection against ROS.

One of the genes identified during our study as associated with cell envelope functions was

mmpL9. We identified three mutants carrying independent transposon insertions is this gene. Even

though it is possible that mmpL9 represents a hotspot for transposon insertion, our results suggest

that it seems to be indeed important for M. tuberculosis survival under oxidative stress conditions

as well as inside host cells. MmpLs belong to the RND (resistance, nodulation and cell division

proteins) family of pumps that recognize and mediate the transport of a range of substrates. In M.

tuberculosis, the colocalization of some mmpL genes with genes involved in polyketide biosynthesis

(pks genes) and lipid metabolism, suggested that MmpLs are involved in lipid synthesis and/or

transport (Tekaia et al., 1999). However, their substrate specificity seems to be dictated, in part, by

their interaction with particular cytosolic enzymes (Jain and Cox, 2005). Thus, mmpL9 might be

involved in the transport of lipids, even though, it does not colocalize with known genes involved in

lipid synthesis (Cole et al., 1998). This seems also to be the case for MmpL3 found to be involved in

the transport of trehalose mycolate in mycobacteria (Grzegorzewicz et al., 2012). Curiously,

MmpL3 has been previously described as involved in heme iron acquisition as well (Tullius et al.,

2011), suggesting that MmpL transporters might be involved in more than one physiological

process in M. tuberculosis. Therefore, MmpL9 could be involved in the transport of other kind of

substances, related to detoxification processes responsible for maintaining an oxidative balance

within the cell. A previous study has shown that mycobacterial P55 efflux pump, besides being

involved in drug resistance, is important for different processes in the cell, including oxidative

stress response (Ramon-Garcia et al., 2009). A Δp55 mutant had increased sensitivity to redox

compounds including diamide, a thiol-oxidizing agent, suggesting that this pump may be involved

Page 98: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

78 Chapter 2

in the extrusion of toxic compounds generated by a thiol reductant such as mycothiol (Ramon-

Garcia et al., 2009). Curiously, this mutant was also highly sensitive to drugs targeting the

peptidoglycan layer of the cell wall, suggesting that P55 is also involved in the transport of some

important substrate to the cell wall (Ramon-Garcia et al., 2009). A similar scenario might be

happening for MmpL9.

Strikingly, we’ve identified a frameshift mutation in the mmpL9 gene of the Beijing/W GC1237

strain. Nonetheless, the fact that a transposon mutant had an attenuated phenotype inside

macrophages that was restored after complementation with the wild-type gene suggests that the

GC1237 mmpL9 gene might be functional. Translation of mmpL9 in GC1237 might occur due to a

recoding mechanism (Baranov et al., 2001), for example through the reinitiation of translation from

a second initiation codon, at position 259, in the same reading frame (Figure 12).

Figure 12. Representation of the mmpL9 gene in M. tuberculosis GC1237. The mmpL9 gene in M.

tuberculosis GC1237 contains a deletion (Δ) in position 218. Reinitiaton of translation might occur from

a second initiation codon situated upstream the deletion, at position 259, which would explain the

attenuated phenotype observed in macrophages of the mmpL9 (100D7) mutant carrying a transposon

inserted at position 957:958. The initial part of the gene’s sequence is shown where both deletion (-)

and the putative second initiation codon, which in the same reading frame, are highlighted in bold.

In this case the Beijing/W MmpL9 would be a modified protein, what would explain the partial

complementation by H37Rv mmpL9 in macrophages. Statistical analysis of polymorphisms in mmpL

genes showed that this deletion could have indeed an effect on protein’s functions and seems to

be specific to all modern and ancestral Beijing/W strains. These results suggest that this

polymorphism might have contributed to the evolution of these strains by conferring some

selective advantage. This is important because the Beijing/W family is one of the most successful

groups of M. tuberculosis strains as it has been frequently associated with drug resistance and

found to be distributed throughout the world (Parwati et al., 2010).

Page 99: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

79 M. tuberculosis resistance to ROS

Curiously, we did not identify during our screen known antioxidant genes, such as katG or sodC.

It is possible that these mutants were not represented in our library given that we have screened

around 6000 mutants, which may not cover the entire 4000 ORFs of the M. tuberculosis genome

(Cole et al., 1998). On the other hand, some of these mutants might be sensitive to endogenous

ROS resulting from aerobic respiration (Imlay, 2008; Storz and Imlay, 1999), which can result in

some in vitro growth defect in the absence of H2O2. In this case, they could have been excluded

from the study before the screening with H2O2. Indeed, an exhaustive study of M. tuberculosis

whole genome expression in response to oxidative stress caused by H2O2 has shown that katG,

sodA, sodC, ahpD, ahpE and ahpC have high basal levels of expression (Voskuil et al., 2011).

However, most of these are not highly induced by H2O2. In addition these genes are not

consistently induced in M. tuberculosis in host cells (Fontan et al., 2008; Schnappinger et al., 2003).

Thus, these genes might be important to counteract low or basal levels of ROS, however, other

pathways are induced when bacteria are exposed to additional levels of oxidative stress, such as

those produced during the phagocytic oxidative burst (Imlay, 2008; Storz and Imlay, 1999). On the

other hand, M. tuberculosis oxidative stress response is concentration-dependent (Voskuil et al.,

2011), as previously shown for E. coli (Imlay and Linn, 1986). Therefore, using different ROS levels

induce different kinds of responses. Finally, it is also possible that some of these genes have

redundant functions and some mutants might not be sensitive to H2O2 but to other forms of

oxidative stress.

Our results have shown that the molybdenum cofactor (MoCo) seems to be important for M.

tuberculosis growth inside macrophages and resistance to oxidative stress. The same has also been

suggested in previous studies. Several genes involved in MoCo biosynthetic pathway have been

identified as important for M. tuberculosis to counteract oxidative stress conditions (Manganelli et

al., 2002) and to parasitize macrophages (Rosas-Magallanes et al., 2007). Additionally, MoCo also

appears to be important for M. tuberculosis to grow in primate lungs (Dutta et al., 2010), which

reveals the significance of this molecule during infection in vivo. MoCo seems to be required for

the activity of several M. tuberculosis enzymes that present recognizable MoCo-associated

domains, including Nitrate reductase (NarGHJI) (Williams et al., 2011). Nitrate reductase seems to

have both nitrate respiratory (Sohaskey and Wayne, 2003) and assimilatory functions (Malm et al.,

2009). It also seems that nitrate reductase is involved in protection of hypoxic M. tuberculosis from

acid killing, as well as against RNS, conditions encountered by this pathogen during host infection

(Tan et al., 2010). This would explain the importance of MoCo in M. tuberculosis survival in host

cells. However, the role of this molecule in protection against oxidative stress is less clear. Biotin

sulfoxide reductase is also a MoCo-enzyme in M. tuberculosis (Williams et al., 2011) that seems

Page 100: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

80 Chapter 2

important for protection from oxidative stress in E. coli, by reducing biotin sulfoxide, the product of

oxidized biotin, into biotin (Ezraty et al., 2005). However, whether this enzyme is functional and

whether it has the same role in M. tuberculosis remains to be determined.

Several genes identified in our screen seem to also have a role in bacterial survival inside host

cells. In a previous study, a ppe54 mutant, identified in a screening using the same transposon

mutant library, exhibited an attenuated growth in mouse macrophages (Brodin et al., 2010). In

addition, M. tuberculosis ppe54 along with ppe56 and ppe21 expression was found to be induced in

IFNγ-activated macrophages (Voskuil et al., 2004). The Rv0986 transporter was also previously

identified as important for M. tuberculosis to bind (Rosas-Magallanes et al., 2007) and replicate

(Pethe et al., 2004) inside macrophages. We have shown that moaD1 and mmpL9 are important for

M. tuberculosis growth in human macrophages as well. Strikingly, some of these genes, rv0986,

ppe54, moaD1 and mmpL9, were also found to be involved in inhibition of phagosome maturation

by M. tuberculosis in previous studies (Brodin et al., 2010; MacGurn and Cox, 2007; Pethe et al.,

2004). We hypothesize that mutants in these genes are delivered to late phagosome

compartments as a consequence of being more susceptible to intracellular killing. Any strain

impaired in intracellular survival will traffic to late endosomes as a secondary effect (Philips, 2008).

Taken together, these observations, suggest a correlation between resistance to oxidative stress

and intracellular growth in macrophages. However, we could not provide experimental proof for a

direct link between increased susceptibility to ROS and reduced intracellular mycobacterial growth.

There was no significant change in mmpL9 and moaD1 mutants phenotype, when compared to the

wild-type, in macrophages treated with ROS inhibitors/scavengers, even when cells where

activated with Vitamin D3, which triggers antimicrobial responses involving ROS production (Sly et

al., 2001)(Sly et al., 2001; Yang et al., 2009). Vitamin D3 activated macrophages might be able to

control mmpL9 and moaD1 mutants growth in a ROS-independent manner, for example through

the induction of autophagy, an apparently central mechanism of vitamin D3 antimicrobial activity

(Shin et al., 2010; Yuk et al., 2009). On the other hand, there are no potent and specific NOX

inhibitors, so, apocynin or DPI, may not be effectively inhibiting Phox throughout the infection

period and could have NOX-unrelated effects (Bedard and Krause, 2007). It is also possible that the

mmpL9 and moaD1 genes are required to protect M. tuberculosis against other forms of stress

existent in macrophages. Indeed, several acid-sensitive mutants, with defects in their cell wall,

were also found to be hypersensitive to oxidative stress (Vandal et al., 2009). However, the mmpL9

mutant did not appear to have an increased susceptibility to acidic pH. Furthermore, neither

mmpL9 nor the moaD1 mutants were more susceptible to starvation. Thus, these genes might be

important to protect M. tuberculosis against RNS or some product resulting from the combination

Page 101: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

81 M. tuberculosis resistance to ROS

of ROS with RNS inside macrophages, such as peroxynitrite, one of the most potent natural

oxidants in biological systems (Zahrt and Deretic, 2002). Indeed, few bacterial genes seem to

exclusively participate in resistance to ROS in vivo (Ehrt and Schnappinger, 2009). Finally, another

possible explanation for the results obtained using the phox inhibitors is that M. tuberculosis

oxidative stress resistance mechanisms are extremely efficient thus masking the antimicrobial

effects of ROS on this pathogen. Consequently, M. tuberculosis infection in the absence of ROS is

not particularly affected. This also seems to be an explanation for the results observed during M.

tuberculosis infection in phox-deficient mice (Jung et al., 2002) and macrophages (Chan et al.,

1992).

Most of the genes identified during our screen have not been described in previous genetic

screenings as required for M. tuberculosis survival in macrophages (Rengarajan et al., 2005) or in

animal models such as mice and non-human primates (Dutta et al., 2010; Lamichhane et al., 2005;

Sassetti and Rubin, 2003). They have also not been previously reported as upregulated in M.

tuberculosis inside host cells (Fontan et al., 2008; Schnappinger et al., 2003). However, we cannot

exclude that the genes identified in our and previous reports are involved in the same metabolic

pathway (as observed for MoaD1), or involved in the synthesis of the same factor. For example,

MmpL9 and Rv0986 are transporters of a yet unidentified substrate and some of the genes we

have identified have unknown annotated functions (Table 2). Similarly, numerous genes identified

during analysis of M. tuberculosis whole genome expression in response to oxidative stress are

annotated as having unknown functions (Voskuil et al., 2011). We also have to consider that

differences regarding M. tuberculosis strains and host cells (human or mice cells; primary cell

cultures or cell lines) employed might also explain the only modest overlap of our results with

those of previous studies.

In conclusion, genes involved in M. tuberculosis oxidative stress response are important for this

pathogen and seem to contribute to its intracellular growth in host cells, which make the products

of these genes attractive targets for the development of new drugs or vaccine candidates.

Page 102: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

82 Chapter 2

Page 103: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

CHAPTER 3

Evolution and diversity of M. tuberculosis Beijing/W strains

reflected by polymorphisms in 3R genes

Olga Mestre1, Tao Luo2, Tiago Dos Vultos1†, Kristin Kremer3, Alan Murray1,4, Amine

Namouchi1, Céline Jackson1, Jean Rauzier1, Pablo Bifani5, Rob Warren6, Voahangy Rasolofo7,

Jian Mei8, Qian Gao2, Brigitte Gicquel1

1 Unité de Génétique Mycobactérienne, Institut Pasteur, Paris, France ;

2Key Laboratory of Medical Molecular

Virology, Fudan University, Shanghai, China; 3

Tuberculosis Reference Laboratory, National Institute for Public

Health and the Environment, Bilthoven, The Netherlands; 4 Institute of Veterinary, Animal and Biomedical Science,

Massey University, Palmerston North, New Zealand;5 Mycobacterial Immunology, Scientific Institute of Public

Health, Brussels, Belgium;6 NRF Centre of Excellence in Biomedical Tuberculosis Research/ MRC Centre for

Molecular and Cellular Biology, Stellenbosch University, Tygerberg, Cape Town, South Africa;7 Unité de la

Tuberculose et des Mycobactéries, Institut Pasteur de Madagascar, Antananarivo, Madagascar;8 Department of TB

Control, Shanghai Municipal CDC, Shanghai, China

The data presented in this chapter is included in the manuscript: “Phylogeny of

Mycobacterium tuberculosis Beijing strains constructed from polymorphisms in genes involved in DNA

replication, recombination and repair.” Phylogeny of Mycobacterium tuberculosis Beijing strains constructed

from polymorphisms in genes involved in DNA replication, recombination and repair.

Mestre O, Luo T, Dos Vultos T, Kremer K, Murray A, Namouchi A, Jackson C, Rauzier J, Bifani P, Warren R,

Rasolofo V, Mei J, Gao Q, Gicquel B. (2011) PLoS One Jan 20;6(1):e16020.

Page 104: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga
Page 105: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

85 Polymorphisms in 3R genes in Beijing/W strains

ABSTRACT 3.1

Investigating the genetic variability among Mycobacterium tuberculosis is important as it can

provide a basis for understanding pathogenesis and host adaptation, as well as the evolution of this

pathogen. Polymorphic genetic markers have been used to determine this genetic diversity. We

have recently shown that SNPs in DNA repair, recombination and replication (3R) genes are a

useful high resolution tool for strain discrimination and can be used to study M. tuberculosis

evolution.

The Beijing/W family is a successful group of M. tuberculosis strains, often associated with drug

resistance and widely distributed throughout the world. To investigate the genetic diversity and

evolution of the Beijing/W family, polymorphisms in 3R genes were analyzed in a collection of

Beijing/W isolates from different geographic origins. We found SNPs in 3R genes associated with

the Beijing/W family, which enabled discrimination of different groups and the proposal of a

phylogeny. One of these groups appeared to be predominant, representing over 60% of all isolates

in distinct parts of the world. Our results show that the Beijing/W family can be divided into

different groups, characterized by particular SNPs in 3R genes that may define their pathogenic

features and contribute to the evolution of this family. These results may contribute to better

understand the success of the Beijing/W family.

Page 106: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

86 Chapter 3

Page 107: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

87 Polymorphisms in 3R genes in Beijing/W strains

INTRODUCTION 3.2

Mycobacterium tuberculosis is considered as one of the most successful human pathogens,

infecting nearly one third of the world’s population. However, only 5 to 10% of these infected

persons will develop the disease (TB) and become sick at a certain point during their lifetimes

(WHO at www.who.int/topic/tuberculosis/en/). One of the main questions that has occupied

researchers is what defines who becomes infected and who develops TB. The genetic variability of

M. tuberculosis seems to have a significant role in the outcome of the disease. Epidemiological data

suggested that differences in transmissibility and virulence among M. tuberculosis strains are

related to the genetic background of the organisms (Caminero et al., 2001; Valway et al., 1998).

Years later, studies using animal models have shown that different M. tuberculosis genotypes

display indeed differences in pathogenesis and virulence (Dormans et al., 2004; Lopez et al., 2003;

Palanisamy et al., 2009).

Polymorphic molecular genetic markers have been used to investigate M. tuberculosis genetic

diversity. IS6110 restriction fragment length polymorphism typing is one of the most widely used

methods, however, this technique is time consuming, technically demanding and insufficiently

discriminatory for isolates containing less than five copies of IS6110 (Kato-Maeda et al., 2011). This

has led to the development of other methods based on the polymorphism of repetitive sequences,

either the direct repeat (DR) region (spoligotyping) (Kamerbeek et al., 1997) or mini satellites

(variable numbers of tandem repeats (VNTR) typing) (Frothingham and Meeker-O'Connell, 1998).

In the recent years, the availability of whole-genome sequences has enabled comparative genomic

analysis to identify single nucleotide polymorphisms (SNPs). SNPs have been used to differentiate

between clinical isolates and are preferred over the use of repeats for the construction of

phylogenetic trees, because recombination events that could occur, independently, at the level of

repetitive sequences, are avoided (Achtman, 2008). Large numbers of SNPs have been identified

and used to genotype worldwide strain collections and investigate M. tuberculosis evolutionary

history (Baker et al., 2004; Filliol et al., 2006; Gutacker et al., 2006; Hershberg et al., 2008). On the

other hand, SNPs might have a role in pathogenesis by affecting expression of genes. We have

recently analyzed SNPs in DNA replication, recombination and repair (3R) genes (Dos Vultos et al.,

2008) in a set of geographically diverse M. tuberculosis strains. Pathogens are constantly exposed

to stressful environments and aggressions to the DNA, such as reactive oxygen species (ROS),

produced by host macrophages (Flannagan et al., 2009). DNA replication, recombination and repair

are responsible for the repair of such damages to the DNA, and, therefore, important mechanisms

in genome dynamics and stability. On the other hand, variations on 3R genes potentially increase

Page 108: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

88 Chapter 3

genomic variability, due to an increase in mutation rates, and contribute to the adaptation and

evolution of bacteria (Denamur and Matic, 2006; Taddei et al., 1997; Tonjum and Seeberg, 2001).

Our study has revealed an unexpectedly high polymorphic set of genes, useful for strain

discrimination. Additionally, analysis of polymorphism in these genes may indicate which strains

are more prone to adapt or evolve (Dos Vultos et al., 2008).

The Beijing/W family, a group of genetically related strains, has demonstrated particular

properties when compared to other M. tuberculosis families, including: increased virulence in

various in vitro (Li et al., 2002; Theus et al., 2004) and in vivo models of infection (Lopez et al.,

2003; Tsenova et al., 2005), association with drug resistance (Tuberculosis, 2006), as well as a wide

distribution throughout the world (Brudey et al., 2006; Glynn et al., 2002). Taken together this

suggests that Beijing/W strains have selective advantages over other M. tuberculosis strains and

represent a serious threat to TB control. Therefore, we decided to investigate genetic diversity and

evolution of Beijing/W strains by analyzing polymorphisms in 3R genes. This will hopefully

contribute to better understand the mechanisms underlying the phenotypic and genotypic

characteristics that explain the success of the Beijing/W family.

Page 109: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

89 Polymorphisms in 3R genes in Beijing/W strains

MATERIALS AND METHODS 3.3

3.3.1 Study isolates and SNP detection

M. tuberculosis Beijing/W clinical isolates used in this study are listed in Table A1. DNA from the

58 isolates, with a Beijing/W spoligotype, used for SNP discovery in 3R genes was provided by:

Madagascar Pasteur Institute, Madagascar (MG); RIVM, The Netherlands (NL) and the Scientific

Institute of Public Health, Belgium (BE). These DNAs were used to amplify the 22 3R genes (Table 1)

using TaKaRa LA Taq (TaKaRa, Shiga, Japan) and primers listed in Table 1. A control non-Beijing

isolate, Myc2 (DNA also provided by the Scientific Institute of Public Health, Belgium (BE)), was also

included. Myc2 is a clinical strain belonging to the Euro-American lineage according to Gagneux’s

classification (Gagneux et al., 2006a).

The amplified products were purified in a single step using exonuclease I (USB, USA) and shrimp

alkaline phosphatase (USB, USA), as previously described (Werle et al., 1994). The purified products

were then sequenced by the dideoxy chain-termination method using the Big Dye Terminator v3.1

cycle sequencing Kit (Perkin Elmer Applied Biosystems, Courtaboeuf, France), according to

manufacturer’s instructions. Sequencing products were run on an ABI prism 3100 Genetic Analyser

(Applied Biosystems).

Sequences were analyzed using the software Genalys obtained at http://software.cng.fr.

Detection of polymorphic sites was performed by comparison with the non-Beijing strain, H37Rv

(Myc1) sequences, obtained from Institute Pasteur at http://genolist.pasteur.fr. Beijing/W specific

SNPs were identified by comparison of sequences with the non-Beijing (Myc2).

Sequencing was also performed for subsequent SNP analysis of the Beijing/W isolates from

South Africa (ZA) (Table A1), which DNAs were provided from NRF Centre of Excellence in

Biomedical Tuberculosis Research/ MRC Centre for Molecular and Cellular Biology in South Africa,

and the GC1237 strain, available in our laboratory. A mismatched PCR method, using one wild-type

primer and one containing the SNP which matched/mismatched the template DNA at the 3'-end of

the primer (Table 1), was used to detect SNPs in the Beijing/W isolates from China (CN) (Table A1).

Page 110: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

90 Chapter 3

Table 1. List of oligonucleotides used for SNP analysis.

Primer name Sequence (5’-3’) Primer name (mismatch)

Sequence (5’-3’)

ligD_f GTCACGGCGAAATTCCACGCGATATTTGA ligD580_f CGGGCATTGGCGGAGGATCT

ligD_r CCCGACCAGATCCAGCAACGACACGTC ligD580_wt-f TGCGTTAGCTAGGGTTTCGAGCAG

ligD_2 TCACCAGCGGCAGCAAGGGATTGCAT ligD580_mt-r TGCGTTAGCTAGGGTTTCGAGCAA

ligD_3 GATACACACCGAGGACCACCCGCTGGAATA ligD162_f CGACGACCTATCCGATCATCG

ligB_f CCACATAGCCCCCAGGCGGTATTGGTA ligD162_wt-r GGAAGGTGACCAACCCGATAT

ligB_r CGCTTGGTCGACGAGCGTGAATCTG ligD162_mt-r GGAAGGTGACCAACCCGATAG

ligB_2 GGCACTCTACCGGGCAAAGGGTCTCAG rccR44_f AAGCGCCCCGCCCAGGACGTG

ligC_f ACCCCAGCTTCGGGAAATACATCCTGT rccR44_wt-r CGGACATCGACCGGCTGACCG

ligC_r TCGCCACACAGACGACAAGTCCCAA rccR44_mt-r CGGACATCGACCGGCTGACCT

dnaZX_f CGCCGAAATCACGCCGAACGTTCA ligD346_f ACCACCATCGCGCCGTACTCA

dnaZX_r CGAACGAAACAACCTGCAGCTACATCACG ligD346_wt-r ACCGCCCACGAGACCAGCACG

dnaZX_2 AACACCTGATCTTCATATTCGCCACCA ligD346_mt-r ACCGCCCACGAGACCAGCACA

dnaZX_3 CTGCTGCTGGAAGTGGTTTGCG uvrC388_r GGATCCCGAAGTGGCGGTAGT

recD_f GGTGTGTTCACCTGGAACCCGCCCA uvrC388_wt-f CAACAACACAAGCTGAAGCGG

recD_r GTCGCCGTGCTGTTCGTGTATGCGATGT uvrC388_mt-f CAACAACACAAGCTGAAGCGC

recD_2 TCTCGCAAGGTGTTACGGTGTTGACTGG mutT4-48_r CGCATCAAATAATGGTGGACG

recG_f CATGTGCACGACCACCATCCAGGCAC mutT4-48_wt-f CGGCAACGGCGAAGCGGTCCC

recG_r CGATGATCCCAGCGTCTGATACGCGA mutT4-48_mt-f CGGCAACGGCGAAGCGGTCCG

recG_2 CAGCACAAAAGTGCAGAGCTGGGACATCTT ogt37_f AGCTGGGCCTGCCTGCACAAC

recG_3 GATGACGGCAGGGCAGAAGAAGCAAGTTC ogt37_wt-r GGTCGGGTGTCCAGTGTGTGC

recX_f CCGACGTGGCTGACGAGATCGAGAAGAA ogt37_mt-r GGTCGGGTGTCCAGTGTGTGA

recX_r CCGCCATCAAGTCGAGGTAAATTCGTTCA uvrC166_f CCGCTACCGCGACGACAAGTC

ruvB_f GATACGGTGCTGGCCGCCAACCAT uvrC166_wt-r GCAGGCATGGACGATCGATCTG

ruvB_r GGGGTCATTGCCAACGGCTCCTTTG uvrC166_mt-r GCAGGCATGGACGATCGATCTT

uvrC_f CAATGCACCCGACCAACAGTGGGATAGC recX8_r GGCCGAGTTCGACATCCTCTA

uvrC_r CCGGACAGCCCGGTTACCAAGACGA recX8_wt-f CTTCGCGCTCAGAAGTCGACG

uvrC_2 TACATCGACAAATGTTCCGCGCCGTGT recX8_mt-f CTTCGCGCTCAGAAGTCGACA

uvrC_3 CGGTGCACCGAAACGCAGAAGATGC recX59_wt-f GGTGTCATCCACCAGGCCAAC

recR_f AAGATGGCGCAGGAACGGCTGGGT recX59_mt-f GGTGTCATCCACCAGGCCAAG

recR_r GAGATCAACATTTTGCAGGCAAGGTGCG recX59-r CTCGGCCAGGGCAAGGAGAAT

nei_f TCTGGTCGAGCGGGCCGACGGCAT recG285_r CGTGCGGCAGGTGCTCGATGT

nei_r GGTGGCAGGCAATATCTGCCCAAGGCGG recG285_wt-f TCCCGCCGTCAGCTCAAAAGG

nth_f ATGACACAAGGAGAGTAAACATGGC recG285_mt-f TCCCGCCGTCAGCTCAAAAGA

nth_r AATAGTCATGCAGTTGGGCAACCA mutT2-58_f CCGGCCATAAACGTCGGAAAC

rv2979_f GTTCGAAGGTCCACAGGGCCAGAACG mutT2-58_wt-r GAGGTCGGCGACCTCGAGTCC

rv2979_r TCCAGTTGTATGCCTTGCGACGAGCA mutT2-58_mt-r GAGGTCGGCGACCTCGAGTCG

tagA_f TGAGCTCGAGGCGCTACGCTCTCAGC ogt12_f CCGCAGGAGAAGATCGCAT

tagA_r CCCCGCCATTGGATTTCCAGCCATA ogt12_wt-r GCCCGGCCAGGGTTAATAGC

uvrD1_f CCCGCAAAAACTTGGCGGGAAAAGTG ogt12_mt-r GCCCGGCCAGGGTTAATAGT

uvrD1_r GGACTTAGCGTCGGCAATTACACCGGTTGA recR89_f CGGACGCGATCCGTGTGACGG

uvrD1_2 CAACCTGAAGAACGAGTTGATCGACCC recR89_wt-r GTGCATTGTCGAGGAACCCAAAGAC

uvrD1_3 CGAGGGTAGCGAGATCACCTACAACGAT recR89_mt-r GTGCATTGTCGAGGAACCCAAAGAT

dnaQ_f CGGGTGGTTACCACCCGGGCAGTTTAC recF269_f GGCGGAGCACGGGGCTGAACT

dnaQ_r TCTCGCAAGGTGTTACGGTGTTGACTGG recF269_wt-r CGGTGCGGACCAACTAGACAAACC

radA_f TAATGGTGCCGATCTCGGCCGGATT recF269_mt-r CGGTGCGGACCAACTAGACAAACA

radA_r GTTGCTGCATAGCGGACATCGAGGGAGAA uvrD1-462_f TCCGCGCCGGTATTCCGTACA

radA_2 GAGATCTACCTCGCCGCACAGTCCGA uvrD1-462_wt-r GACGAGCGCGTCACCGAAGCC

recF_f GGAGCGAGTGTCTTTCGGGTTTACGACTGC uvrD1-462_mt-r GACGAGCGCGTCACCGAAGCT

recF_r CGCCCTCGACCGGCGTCTTGTCC ligB77_r TGTCGGCGAGACATGCCAAGCT

mutT2_f CTGCCAGCCGTTGAGGTCGT ligB77_wt-f GGGTGGCGTCGACACCGGTGA

mutT2_f CGGGCATGCAAACCCAAGTTA ligB77_mt-f GGGTGGCGTCGACACCGGTGG

mutT4_f TCGAAGGTGGGCAAATCGTG dnaQ161_f GGACCAGCGGGCGGCCCTGGA

mutT4_r TGGGGTTCGCTGGAAGTGG dnaQ161_wt-r CAACGGCCGCACGATGCATTC

ogt_f CAGCGCTCGCTGGCGCC dnaQ161_mt-r CAACGGCCGCACGATGCATTT

ogt_r GACTCAGCCGCTCGCGA nth122_f CCCGCCGTCCGTGAAAGATCA

alkA_f AGCCGCGTAGGTAACCT nth122_wt-r GCCGGCCACCATGGACAAGTT

alkA_r TGCTCGAGCATCCGCAG nth122_mt-r GCCGGCCACCATGGACAAGTG

alkA_2 CGCATGCAGACCGCCCG nth34_f TCACCGCCAAACCGCTCAA

alkA_3 CACTGCACGTTGCCGAC nth34_wt-r ATTTCCGCACGTATACTGAGA

alkA_4 GCTGACGATGCCGTTGCC nth_34_mt-r ATTTCCGCACGTATACTGAGC

dnaZX92_f GCGAGCAACGCCCGCATAGT

dnaZX92_wt-r GCAGCATCGACGTGGTAGAGC

Page 111: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

91 Polymorphisms in 3R genes in Beijing/W strains

Primer name (mismatch)

Sequence (5’-3’)

dnaZX92_mt-r GCAGCATCGACGTGGTAGAGT

alkA11_f ATCGCCCGCGCCACGACGTCA

alkA11_wt-r ACTTCGAACGCTGCTACCGGG

alkA11_mt-r ACTTCGAACGCTGCTACCGGA

mutT4-99_f GGCGGCGCGCTGCGGCTACAG

mutT4-99_wt-r CAACTCGATGTGCCCCTTGGGTAGC

mutT4-99_mt-r CAACTCGATGTGCCCCTTGGGTAGT

recX153_f GCGGGCGAACGCAGCAAAGAG

recX153_wt-r CCTCGCACGCCAAGGTCTGGC

recX153_mt-r CCTCGCACGCCAAGGTCTGGT

recD277_r CGGGCCTGGCACCGGGAAGAC

recD277_wt-f TCGGCCAGCCGGGCCATCAGC

recD277_mt-f TCGGCCAGCCGGGCCATCAGT

radA186_f TCCGGACGGCGCGCGCTCTAT

radA186_wt-r CCTGCGTGACCCCGCCGGTGA

radA186_mt-r CCTGCGTGACCCCGCCGGTGG

tagA179_f GTGCGCAACCGCGCCAAGATT

tagA179_wt-r CCAGCATGCTTGGATATGGTCGTCG

tagA179_mt-r CCAGCATGCTTGGATATGGTCGTCA

(f) for forward and (r) for reverse oligonucleotides used for amplification and sequencing reactions.

Oligonucleotides whose name finishes in number were used for sequencing reactions. (wt) for wild-type and (mt)

for mutant oligonucleotides used for detection of SNPs by mismatched PCR.

3.3.2 LSPs detection

Tsolaki et al. (2005) have described Large Sequence Polymorphisms (LSPs) for the Beijing/W

family. These, designated regions of Difference (RDs) (RD105, RD181, RD150, RD142), correspond

to genomic regions present in M. tuberculosis H37Rv but variably deleted in Beijing/W strains. The

initial set of 58 Beijing/W isolates were tested for the presence or absence of these RDs through a

Deletion-Targeted Multiplex PCR (DTM-PCR), as previously described for RD105 detection in

Beijing/W strains (Chen et al., 2007). Briefly, two primers were designed flanking each one of the

RD regions (P1 and P2) and one primer hybridizing inside each the RD region (P3) (Table 2).

Multiplex-PCR reactions were performed using TaKaRa LA Taq (TaKaRa, Shiga, Japan) and different

sets of primers for each RD (Table 2). The presence or absence of deletions was determined,

following electrophoresis on an agarose gel, by comparison with M. tuberculosis H37Rv PCR

products, used as control.

Page 112: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

92 Chapter 3

Table 2. List of primers used in Multiplex-PCR for Large Sequence Polymorphisms (LSPs) analysis.

3.3.3 SNP Data analysis for Phylogenetic tree construction

SNPs were concatenated resulting in one character string (nucleotide sequence) for each clinical

isolate analyzed. A FASTA file was created to run in the Network software (Bandelt et al., 1999) to

build a phylogenetic tree based on the median-joining method. This software assumes that there is

no recombination between genomes.

Region of difference (RD) Primer sequence (5’ – 3’)

RD105

P1(105) - GGAGTCGTTGAGGGTGTTCATCAGCTCAGT

P2(105) - GCGCCAAGGCCGCATAGTCACGG

P3(105) - GGTTGCCCACTGGTCGATATGGTGGACTT

RD181

P1(181) - CGCAACGGCCGCGGTGAACTCT

P2(181) - CGGGCGGCTGCGGGAACCTT

P3(181) - CTGGCCTGGTTCGGCTTGGTCCC

RD150

P1(150) - TGTGGCGTGGCTCGGCAAATAG

P2(150) - CGGGACGGCAAACGGGTGAT

P3(150) - CGGCGTCATCGCGTATCTGA

RD142

P1(142) - CGTCCGCGACGACGAACAA

P2(142 - TCACTTTCCATTTCCAGCGGCAACT

P3(142) - CGTCGATGGCAACACACCGAAA

Page 113: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

93 Polymorphisms in 3R genes in Beijing/W strains

RESULTS 3.4

3.4.1 Identification of polymorphisms in 3R genes in Beijing/W strains

A collection of 58 clinical isolates with a Beijing/W spoligotype from different geographic origins

(Table A1, materials and methods), was used to search for variations in 3R genes. Of the 56

described genes encoding 3R components (Dos Vultos et al., 2008), 22 were previously

demonstrated to be polymorphic among Beijing/W strains. However, these studies analyzed either

a small number of isolates or a restricted number of genes (Dos Vultos et al., 2008; Ebrahimi-Rad et

al., 2003). Therefore, these 22 genes (Table 3) were sequenced for each of the 58 Beijing/W

isolates as well as for a non-Beijing strain (Myc2), resulting in roughly 1,6 Mbp of sequence data.

Comparative analysis with the M. tuberculosis H37Rv (Myc1) genome sequence identified 48 SNPs.

Forty-one (85%) SNPs appeared to be specific for Beijing/W strains, as these were absent from the

non-Beijing strain (Myc2), as well from the 86 non-Beijing strains included in our previous analysis

of 3R polymorphisms using a global M. tuberculosis collection (Dos Vultos et al., 2008). Almost half

of these SNPs (19/41) corresponded to new variations, not previously described in Beijing/W

strains (Dos Vultos et al., 2008; Ebrahimi-Rad et al., 2003; Hershberg et al., 2008). Thirty of the 41

Beijing/W specific SNPs enabled discrimination of 24 different sequence types for which a

preliminary phylogenetic network was constructed using the Network software (Table 3) (Figure

1A). Based on the inferred proteins, the number of non-synonymous SNPs (nsSNPs) was twice the

number of synonymous SNPs (sSNPs) (Table 3).

Analysis of large sequence polymorphisms (LSPs) revealed that these isolates included the four

different sublineages previously defined in Beijing/W strains using these markers (Tsolaki et al.,

2005)(Figure 1B).

Page 114: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

94 Chapter 3

Table 3. List of SNPs identified by sequencing 3R genes in Beijing/W strains, which define

different sequence types (Figure 1 and 2).

* Most informative SNPs (identified in more than one isolate) observed in this study.

SNP number Gene Codon position SNP type

1* ligD 580 (CTG > TTG) Synonymous

2 ligD 162 (GAT > GCT) Non-synonymous

3* recR 44 (GGT > TGT) Non-synonymous

4 ligD 346 (GGC > GGT) Synonymous

5 uvrC 388 (CGG > CGC) Synonymous

6* mutT4 48 (CGG > GGG) Non-synonymous

7* ogt 37 (CGC > CTC) Non-synonymous

8 uvrC 166 (CAG > AAG) Non-synonymous

9 recX 8 (CCG > CTG) Non-synonymous

10* recX 59 (GTT > CTT) Non-synonymous

11 recG 285 (CCT > TCT) Non-synonymous

12* muT2 58 (GGA > CGA) Non-synonymous

13* ogt 12 (GGG > GGA) Synonymous

14 recR 89 (GAC > GAT) Synonymous

15* recF 269 (GGG > GGT) Synonymous

16* uvrD1 462 (GGC > AGC) Non-synonymous

17 ligB 77 (GTC > GCC) Non-synonymous

18 dnaQ 161 (TTC > TTT) Synonymous

19 nth 122 (TTG > TGG) Non-synonymous

20* dnaZX 92 (CTG > TTG) Synonymous

21 nth 34 (GAG > GCG) Non-synonymous

22 alkA 11 (GCG > ACG) Non-synonymous

23* mutT4 99 (TCG > TCA) Synonymous

24* tagA 129 (GCG > ACG) Non-synonymous

25* recX 153 (GGC > GAC) Non-synonymous

26* radA 276 (ATC > ACC) Non-synonymous

27 recD 139 (GTA > TTA) Non-synonymous

28 recD 277 (ACG > ACA) Non-synonymous

29 radA 186 (GTC > GCC) Non-synonymous

30 tagA 179 (GTC > GTT) Synonymous

Page 115: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

95 Polymorphisms in 3R genes in Beijing/W strains

Figure 1. Phylogenetic network based on SNPs discovered in the collection of 58 Beijing/W isolates. This

phylogenetic network was constructed using the median-joining algorithm with the final set of 48 SNPs

characterized by sequencing 22 3R genes in 58 Beijing/W isolates plus one non-Beijing isolate (Myc2). Isolates are

Page 116: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

96 Chapter 3

color coded according to their geographic origin (A), large sequence polymorphisms (LSPs) (B) and, SNPs in mutT2

mutT4 and ogt genes (C). The reference strain M. tuberculosis H37Rv (Myc1) was also included. The numbers in

each branch correspond to SNPs (Table 3) that enabled discrimination of sequence types. Node sizes are

proportional to the number of isolates belonging to the same sequence type: Bmyc4 node (2); Bmyc12 node (3);

Bmyc13 node (3); Bmyc19 (2); Bmyc16 node (7); Bmyc10 node (23). Table A1 describes the strains that belong to

each node. Mv represents a median vector created by the software and can be interpreted as possibly extant

unsampled sequences or extinct ancestral sequences. The evolutionary pathways proposed previously for

Beijing/W strains using LSPs (Tsolaki et al., 2005) and SNPs in mutT2, mutT4 and ogt (Ebrahimi-Rad et al., 2003)

have been included for comparison (B and C right bottom).

The same SNP patterns were found in isolates from different geographic regions (Figure 1A),

suggesting that the present collection of Beijing/W isolates is representative of this family.

A color code was attributed to each node according to SNPs previously described in mutT2,

mutT4 and ogt DNA repair genes (Ebrahimi-Rad et al., 2003), as well as according to LSPs (Tsolaki et

al., 2005) (Figure 1B and C). Phylogenetic relationships established by 3R SNPs were in agreement

with evolutionary pathways suggested previously using these genetic markers (Ebrahimi-Rad et al.,

2003; Tsolaki et al., 2005) (Figure 1B and 1C). However, sequencing of 3R genes was more

discriminatory than LSPs; 24 sequence types versus four sublineages defined by the LSPs.

3.4.2 Construction of a phylogeny for Beijing/W strains using 3R SNPs

Next, we investigated the set of 30 polymorphic SNPs (Table 3), discovered by sequence analysis

of the 3R genes, in a larger collection of Beijing/W strains including: 192 Beijing/W clinical isolates

from China and 55 Beijing/W strains isolated in South Africa (Table A1). The M. tuberculosis

Beijing/W strain, GC1237, responsible for a tuberculosis epidemic in Gran Canaria (Caminero et al.,

2001), was also included.

A phylogenetic network was constructed including this larger set of isolates (Figure 2). Certain

SNPs that were previously found in a single isolate were confirmed with this larger sample. Overall,

fourteen SNPs were found in more than one isolate and were therefore informative (Table 1). Two

new sequence types (Bmyc25 and Bmyc26) were identified (Figure 2). Strikingly, sixty-two percent

of the isolates belonged to a single group, Bmyc10, which included strains from distinct parts of the

world. The second most represented sequence-type was Bmyc25 that included the Gran Canaria TB

outbreak strain GC1237 (Caminero et al., 2001).

Page 117: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

97 Polymorphisms in 3R genes in Beijing/W strains

Figure 2. Phylogenetic network based on SNPs characterized in the entire collection of 305

Beijing/W isolates. This phylogenetic network was constructed using the median-joining algorithm with

the set of SNPs identified in the 3R genes analyzed on the final collection of 305 Beijing/W isolates.

Isolates are color coded according to their geographic origin. M. tuberculosis strains Myc1 (H37Rv) and

Myc2 are included as non-Beijing strains. The numbers in each branch correspond to SNPs (Table 3) that

enabled discrimination of sequence types. Node sizes are proportional to the number of isolates

belonging to the same SNP type: Bmyc1 node (2); Bmyc2 node (14); Bmyc4 node (13); Bmyc6 node (7);

Bmyc25 node (28); Bmyc26 node (13); Bmyc12 node (3); Bmyc13 node (13); Bmyc16 node (7); Bmyc19

node (2); Bmyc10 node (188). Table A1 describes the strains that belong to each node. Mv represents a

median vector created by the software and can be interpreted as possibly extant unsampled sequences

or extinct ancestral sequences. The relative proportion of isolates in each node, of a given geographic

origin, may not reflect the population structure of the Beijing/W family of that geographic region.

Page 118: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

98 Chapter 3

Page 119: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

99 Polymorphisms in 3R genes in Beijing/W strains

DISCUSSION 3.5

M. tuberculosis complex strains have been considered as highly clonal, when compared to other

pathogens (Achtman, 2008). Initial reports have even suggested that these strains share 99%

similarity at the nucleotide level (Sreevatsan et al., 1997). However, in the recent years, large

numbers of SNPs have been identified and used in order to get a more detailed insight into the

diversity and evolutionary history of this bacteria (Baker et al., 2004; Dos Vultos et al., 2008; Filliol

et al., 2006; Hershberg et al., 2008). SNPs analysis is a simple and relatively fast way to compare

organisms and trace back the evolutionary history of strains, as some SNPs are highly informative.

Therefore, SNPs analyses are becoming more and more attractive with the increasing number of

genome sequencing projects. This will provide data particularly relevant to understand the genetic

basis for strain differences in pathogenesis.

Allelic variation in 3R genes seems to be an important mechanism in evolution and adaptation

of microorganisms. Defective 3R systems could potentially increase genomic variability, due to an

increase in mutation rates. Strains with high mutations rates (mutators), may have, in certain

conditions, a selective advantage, as they produce more adaptive mutations (Denamur and Matic,

2006; Taddei et al., 1997; Tonjum and Seeberg, 2001). The Beijing/W family is a group of strains

that seems to have a selective advantage when compared to other M. tuberculosis families.

Therefore, polymorphisms in 3R genes were analyzed in a collection of Beijing/W isolates from

different geographic origins. SNPs in 3R genes associated with the Beijing/W family were identified

that enabled the discrimination of 26 different sequence types for which a phylogeny was

constructed. Phylogenetic relationships established by sequence types were in agreement with

evolutionary pathways suggested by other genetic markers, such as Large Sequence

Polymorphisms (LSPs). The remarkable clonality of M. tuberculosis implies that different genetic

markers reveal similar phylogenies (Supply et al., 2003), if not, these are not robust markers

appropriate to evolutionary analyses.

Polymorphisms in 3R genes show that the Beijing/W family can be subdivided in different

groups, as previously shown using other genetic markers (Hanekom et al., 2007; Tsolaki et al.,

2005). We did find however, a recently expanded group, Bmyc10, which seems to be predominant

as it includes over 60% of strains. This sequence type was identified in isolates from China, where

the Beijing/W family is highly prevalent, but also in other countries, where the Beijing/W family is

less prevalent, such as Madagascar and The Netherlands. In a previous study, a group of Beijing/W

strains characterized by the RD181 deletion and polymorphisms in mutT4 and mutT2 appears to be

predominant in a collection of strains isolated in Italy (Rindi et al., 2009). Strains belonging to

Page 120: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

100 Chapter 3

Bmyc10 node also had the RD181 deletion and the same SNPs in mutT4 and mutT2 genes (SNP6

and SNP12). This seems, though, to be a prevalent group in different parts of the world, suggesting

that Beijing/W strains of this genotype might have some selective advantage. The Bmyc25 group

that includes the Gran Canaria TB outbreak strain GC 1237 (Caminero et al., 2001), might represent

another predominant group among Beijing/W strains. Taken together, these observations suggest

that polymorphisms in 3R genes may confer advantageous phenotypes on certain Beijing/W

genotypes. Indeed, different Beijing/W strains show different pathogenic characteristics in human

macrophages (Theus et al., 2007), as well as in mice (Aguilar et al., 2010; Dormans et al., 2004), or

guinea pigs (Kato-Maeda et al., 2011; Palanisamy et al., 2009). However, host cell transcriptome

responses were found to be similar in Beijing/W strains from different genotypes defined by 3R

SNPs (Wu et al., 2012). Similarly, different Beijing/W genotypes, including Bmyc10 and Bmyc25

strains, do not seem to induce a differential cytokine modulation in macrophages (Wang et al.,

2010). Therefore, other pathogenic characteristics might explain the increased adaptability of

strains with the Bmyc10 and Bmyc25 genotypes (Wang et al., 2010; Wu et al., 2012). For example,

Beijing/W strains belonging to different sublineages defined by LSPs (Tsolaki et al., 2005) seem to

present differences in transmissibility in a mouse model of infection (Aguilar et al., 2010).

Beijing/W strains have also shown variability in the expression levels of the dormancy regulon

(DosR) (Fallow et al., 2010). Therefore, it could be interesting to evaluate differences in Beijing/W

strains ability to persist in a dormant state inside host cells. It should also be interesting to analyze

if these strains have different capacity to resist macrophage antimicrobial mechanisms during

hypoxia, a granuloma characteristic condition that determines DosR activation in M. tuberculosis

(Nickel et al., 2012). Hypoxia doesn’t change cytokine release but triggers an antimicrobial pathway

associated with growth inhibition of the bacilli (Nickel et al., 2012).

Analysis of the effect on enzyme characteristics of the variation in the mutT2 gene, a

characteristic of all Bmyc10 isolates (SNP12, Figure 2), has revealed significant changes in enzyme

properties, caused by a single amino acid substitution, that leads to protein destabilization. It was

suggested that this altered MutT2 enzyme may contribute to the success of strains due to an

increase in nucleotide-dependent reactions (Moreland et al., 2009). Therefore, some of the SNPs

identified in this study might have an effect on protein’s function. The analysis of the functional

effects of the polymorphisms observed in 3R genes might be crucial to understand the real

meaning of such variations. In our previous analysis we have shown that the observed variations in

3R might be slightly deleterious, resulting in suboptimal 3R system activity (Dos Vultos et al., 2008).

Therefore, some of these strains might have a mutator phenotype, increasing the probability of

acquiring mutations in genes conferring some selective advantage. For example, mutator strains

Page 121: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

101 Polymorphisms in 3R genes in Beijing/W strains

may generate mutations that confer antibiotic-resistance at a higher rate than non-mutator strains

(Denamur and Matic, 2006). When antibiotic-resistance is mainly determined by the acquisition of

mutations in the chromosome, as is the case of M. tuberculosis (Heym et al., 1994), the advantage

of being a mutator increases significantly (Tenaillon et al., 1999). A study tried to evaluate

contribution of polymorphism on the DNA repair genes, mutT, to resistance to antibiotics. Even

though no statistically significant association of missense mutations on these genes with and

increased prevalence of antibiotic-resistance was detected, the authors couldn’t rule out the

hypothesis that mutator phenotypes might increase the rate of drug resistance (Lari et al., 2006).

Beijing/W strains have been often associated with drug resistance, even though, the underlying

mechanisms for this association remain unknown (Tuberculosis, 2006). Beijing/W strains have

shown similar rates of mutation-conferring resistance to rifampin compared to non-Beijing strains

(Werngren and Hoffner, 2003). Therefore, another explanation would be that drug-resistance in

Beijing/W strains has a lower or no cost on fitness, given that resistance-conferring mutations are

often associated with a fitness cost (Andersson and Levin, 1999). The fitness of non-Beijing drug

resistant strains was found to be slightly reduced when compared to drug-susceptible M.

tuberculosis. However, drug resistant Beijing/W strains presented some variability: some strains

showed loss of fitness while others did not (Toungoussova et al., 2004). Given that the fitness

impact of resistance-conferring mutations in M. tuberculosis depends on the genetic background of

the strain (Fenner et al., 2012; Gagneux et al., 2006b), the fitness cost of these mutations may vary

among different Beijing/W sublineages. Indeed, different sublineages of the Beijing/W family may

differ in their mechanisms of adaptation to drug selection pressures (Iwamoto et al., 2008;

Mokrousov et al., 2006). It is also possible that the accumulation of compensatory mutations will

mitigate the biological cost of drug-resistance mutations (Gagneux et al., 2006b). Thus, certain

Beijing/W sublineages could have an advantage due to the occurrence of compensatory mutations.

In mutator strains, the probability for this to occur is much higher. However, the concept of a

mutator phenotype in Beijing/W strains was not supported when the frequency of spontaneous

mutations was calculated using the Luria–Delbruck fluctuation test. The rate of spontaneous

mutations was the same in Beijing/W, non-Beijing and laboratory strains (Werngren and Hoffner,

2003). In mutator strains, reduction in mutation rates should occur, for example, by the acquisition

of suppressor/compensatory mutations, before the load of deleterious mutations becomes too

high (Denamur and Matic, 2006; Taddei et al., 1997). Thus, polymorphisms found in 3R genes may

be related to transient mutator phenotypes in Beijing/W strains.

In conclusion, analysis of polymorphisms in 3R genes has shown that different Beijing/W groups

can be identified and characterized by specific 3R SNPs. These might have induced some selective

Page 122: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

102 Chapter 3

advantage that allowed the expansion of certain genotypes, therefore contributing to the evolution

of the Beijing/W family. We hope that our study will contribute to better understand the molecular

basis for the enhanced pathogenicity attributed to the Beijing/W family.

Page 123: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

CHAPTER 4

Concluding Remarks

Page 124: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

104 Chapter 4

Page 125: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

105 Concluding Remarks

Mycobacterium tuberculosis is one of the most successful pathogens, being extremely adapted

to its host. It is estimated that one-third of the world’s population is infected with this pathogen

(WHO at www.who.int/topic/tuberculosis/en/) and, even though in 90-95% of these cases the

infection is contained, M. tuberculosis is not eliminated and is able to persist in its host for years,

until, disease develops due to not always well-understood reasons (Kaufmann and McMichael,

2005). Understanding the mechanisms used by M. tuberculosis to successfully persist in its host,

particularly inside macrophages, will lead to insights into possible targets for new effective anti-TB

strategies.

M. tuberculosis cell envelope constitution explains part of its resistance against host

antimicrobial mechanisms. It forms a permeability barrier and is, thus, the first line of defense

against toxic molecules (Brennan and Nikaido, 1995). However, the contribution of M. tuberculosis

envelope components to this remains poorly documented. We have shown, by screening, for the

first time, a transposon mutant library for mutants with increased susceptibility to reactive oxygen

species (ROS), that M. tuberculosis cell envelope is the first line of defense against ROS,

antimicrobial molecules produced by macrophages to destroy intracellular pathogens such as M.

tuberculosis (Flannagan et al., 2009). Certain cell wall lipids, such as LAM, have been previously

identified as ROS scavengers (Chan et al., 1991). Even though we did not identify any known genes

involved in the biosynthesis of mycobacterial lipids, such as pks (polyketide synthases), we have

identified transporters possibly involved in transport of lipids or other molecules to the cell

envelop (MmpL9 and Rv0986). We have identified as well, proteins that seem to be involved in the

synthesis of components of the cell envelope, although no known function has been assigned so

far (Rv1507c, Rv1508a, Rv1509, Rv3594). Several PPE coding genes were also identified. However,

how these proteins protect mycobacteria against ROS is not clear once not much is known about

the molecular function of PE/PPEs (Akhter et al., 2012). Nonetheless, PE_PGRS11 was shown to be

exposed to the cell surface and to be involved in mycobacterial resistance against H2O2-induced

oxidative stress. This protein was found to be a phosphoglycerate mutase, a protein involved in

glycolysis that has been previously demonstrated to decrease the accumulation of ROS in cells

(Chaturvedi et al., 2010). In addition, a transcriptomic analysis has also shown that several PE/PPE

genes are induced by H2O2, but not by NO, in M. tuberculosis (Voskuil et al., 2011). These results

provide proof that PE/PPEs have indeed a role in protecting mycobacteria against oxidative stress.

The highly impermeable cell wall of M. tuberculosis also contributes to its intrinsic resistance

against chemotherapeutic agents (Jarlier and Nikaido, 1994; Nikaido, 2001). Therefore, cell wall

components are good targets for developing new chemotherapeutic agents against TB. Indeed,

these are currently the target of several antimycobacterial agents, including the two important first

Page 126: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

106 Chapter 4

line drugs, isoniazid and ethambutol (Sarkar and Suresh, 2011). Nonetheless, there is an ongoing

search for new cell wall targets. Certain bacterial antibiotics are responsible for an increase in ROS

production within cells via the fenton reaction (Kohanski et al., 2007). Tolerance to these

antibiotics may, therefore, depend on the ability of the cell to defend itself against ROS. Thus,

targeting molecules involved in resistance to ROS, such as those identified in our study, could

enhance the efficacy of mycobacterial drugs known to generate ROS, such as isoniazid, kanamycin

or ofloxacin (Kohanski et al., 2007; Mukherjee et al., 2009). Indeed, a mutant overexpressing

Wag31, a protein involved in regulation of polar cell wall synthesis, had increased susceptibility to

hydrogen peroxide as well as to isoniazid and ofloxacin (Mukherjee et al., 2009). This kind of

approach could significantly contribute to TB treatment accomplishment. On the other hand, it

could also avoid the appearance of drug-resistant bacteria. It was shown recently that bacterial

death induced by ROS might eliminate persisters (Grant et al., 2012), small populations that resist

to high doses of antibiotics and contribute to the rise in MDR- and XDR-TB (Keren et al., 2011).

Furthermore, these drug-tolerant, persistent bacteria have, in M. tuberculosis, the physiological

state of dormant bacilli, typically found in latent TB (Keren et al., 2011; Ulrichs and Kaufmann,

2006). Therefore, targeting M. tuberculosis molecules involved in resistance to ROS may contribute

to TB treatment improvement and could be suitable to treat patients latently infected with M.

tuberculosis, which represent today a huge reservoir for the disease, without increasing the burden

of drug-resistant TB.

Usually, basal oxidative defences are sufficient to protect bacteria from endogenous ROS that

result from aerobic respiration. However, these defences are inadequate if rates of ROS are

accelerated, for example during the phagocytic oxidative burst. Most microbes induce additional

responses when this happens (Imlay, 2008; Storz and Imlay, 1999). Thus, it is important to

discriminate the effects of phagocytic and endogenous ROS (Craig and Slauch, 2009; Storz and

Imlay, 1999). The fact that most of the genes identified in our study are components of the cell

envelop suggests that these are relevant to counteract ROS generated exogenously, such as those

originated during the phagocytic burst. However, extracellular ROS that penetrate bacterial

membranes will increase intracellular ROS to superior levels to those induced by endogenous ROS

(Imlay, 2008; Storz and Imlay, 1999). Therefore, mechanisms against high intracellular levels of ROS

are also important for bacteria to counteract oxidative stress induced by phagocytes. This would

explain the identification of genes in our screen that do not code for envelope components, such

as moaD1. One might hypothesise that these genes are not essential for sustaining endogenous

ROS as no significant growth defects were observed in vitro without H2O2 for these mutants (Table

1). Additionally, many of the genes identified in our screening seem to have a role in the

Page 127: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

107 Concluding Remarks

intracellular lifestyle of M. tuberculosis, suggesting that these are important to resist against ROS

produced by host cells. However, to confirm that these proteins are specific for resistance against

phagocytic ROS, mutants’ growth in phox-deficient macrophages should be investigated.

The proteins identified in our study might thus contribute to M. tuberculosis virulence. Putative

virulence factors could be useful for designing attenuated vaccine candidates. Indeed, PPEs and

MmpL proteins have been frequently associated with virulence (Akhter et al., 2012; Domenech et

al., 2005). In a recent study, the comparative genomic analysis of free-living soil mycobacteria and

obligate parasites of the M. tuberculosis complex has shown that, along with the known gene

families related to pathogenesis such as PE/PPE genes, gene families related to biosynthesis of

Molybdenum Cofactor (MoCo) were found to be expanded in pathogenic mycobacteria. These

results suggest an important role for MoCo in the adaptation of environmental mycobacteria to

obligate pathogenesis (McGuire et al., 2012). Proteins involved in MoCo biosynthesis, such as

MoaD1, seem to represent potential virulence factors as well.

In the study of McGuire et al. (2012) it was also shown that genes related to DNA repair are

associated with adaptation of environmental mycobacteria to obligate pathogenesis. Surprisingly,

we did not select any mutant in known DNA repair genes during our screen. In E. coli, at low

concentrations (1mM) of H2O2, the main mechanism of ROS-dependent antibacterial activity is

DNA damage (Imlay and Linn, 1986). In M. tuberculosis induction of DNA repair gene expression

indicates that DNA damage occurs only at 5-10 mM H2O2 (Voskuil et al., 2011). However, bacteria

are not killed at these concentrations, suggesting that the damage does not result in DNA-

dependent killing, like in E. coli. These results suggest that M. tuberculosis DNA repair mechanisms

are highly effective in repairing the damages caused by H2O2. In addition, M. tuberculosis possesses

many genes with redundant functions, particularly in the BER pathway, one of the major

mechanisms involved in the repair of oxidative DNA damage (Dos Vultos et al., 2009; Friedberg et

al., 2006). These observations could explain why we did not identify any H2O2 sensitive mutant in

genes playing a role in DNA repair mechanisms. DNA repair mechanisms have also been suggested

to be essential for the survival of the intracellular pathogen Salmonella typhimurium (Buchmeier et

al., 1995). However, a recent study has shown that phagocytic superoxide primarily damages an

extracytoplasmic target to inhibit or kill this pathogen, suggesting that the dogma that DNA is a

primary bacterial target of the phagocytic oxidative burst needs to be re-evaluated (Craig and

Slauch, 2009).

In conclusion, our study has allowed us to identify genes involved in M. tuberculosis defence

against oxidative stress. However, there are still many genes with apparently unknown functions

that seem to have a role in such kind of mechanisms. This means that further studies are required

Page 128: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

108 Chapter 4

to expand our knowledge about the molecular mechanisms used by M. tuberculosis to counteract

oxidative stress, particularly during macrophage infection.

Genome variability is required for bacteria to successfully adapt to constantly changing and

stressful environments, such as the ones found in human hosts or those induced by antibiotic

treatment. DNA repair, recombination and replication (3R) are important mechanisms in

generating this genomic variability (Denamur and Matic, 2006; Tonjum and Seeberg, 2001).

Therefore, analysis of polymorphisms in 3R genes can provide clues into which strains are/were

more prone to adapt and evolve. Our previous analysis of polymorphisms in 3R genes have shown

that these genes may indeed play a role in M. tuberculosis evolution and could be a useful tool for

strain discrimination (Dos Vultos et al., 2008). These results prompt us to investigate

polymorphisms in a successful group of M. tuberculosis strains, the Beijing/W family. We have

shown that different groups carrying specific polymorphisms in 3R genes exist within the Beijing/W

family (Chapter 3). It has been previously, shown that the Beijing/W family can be divided into

several groups/sublineages using other genetic markers (Hanekom et al., 2007; Tsolaki et al., 2005).

3R mechanisms might have contributed to this genetic diversity observed in Beijing/W strains.

However, an important question needs to be asked: what is the meaning of these polymorphisms?

Polymorphisms in 3R genes might have induced the accumulation of mutations conferring a

selective advantage (Denamur and Matic, 2006). Indeed, certain mutations could have conferred

some advantage to M. tuberculosis Beijing/W strains. For example, it was shown that typical

Beijing/W strains carry a high number of non-synonymous SNPs in genes coding for the regulatory

network (Schurch et al., 2011) that could result in changes in cellular processes controlled by

transcriptional regulation. These variations may play an important role in adaptation of these

bacteria to different environments and explain why typical Beijing/W strains are an emerging

phenotype (Schurch et al., 2011). A frameshift mutation in the gene encoding the DosT sensor

kinase, which senses the environmental stimuli that trigger dosR expression, was found to

correlate with the appearance of constitutive overexpression of the DosR regulon in the most

recently evolved Beijing/W sublineages (Fallow et al., 2010). Even though mutation in dosT doesn’t

seem to be directly responsible for DosR constitutive expression, its inactivation might have

induced for selection of this phenotype, which is associated with M. tuberculosis adaptation and

survival to conditions found in human lungs (Voskuil et al., 2003). Another frameshift mutation that

might have induced a selective advantage to Beijing/W strains was the one identified by us in the

mmpL9 gene given that all publicly available modern Beijing/W genomes were found to carry it.

Despite this mutation, MmpL9 seems to be functional in Beijing/W strains and to have a role in

bacterial response to oxidative stress as well as in survival inside host cells (Chapter 2). This

Page 129: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

109 Concluding Remarks

mutation might have induced a protein change that confers a better protection to bacteria in these

conditions.

Studies in vitro (Firmani and Riley, 2002; O'Brien et al., 1994) and in animal models (Aguilar et

al., 2010; Dormans et al., 2004; Lopez et al., 2003; Palanisamy et al., 2009; Tsenova et al., 2005),

have demonstrated strain-dependent variation in key aspects of virulence such as stress survival,

transmission, pathology and lethality. On the other hand, it is also important to consider the

genetic diversity of any pathogen when identifying drug targets, vaccine antigens and developing

tools for molecular diagnostics. The analysis of 3R genes in Beijing/W strains might indicate which

strains are more prone to develop mutations conferring-drug resistance, or are more able to adapt

and persist in host cells. This is important considering that Beijing/W strains have been frequently

associated with drug resistance (Tuberculosis, 2006), and seem to have increased virulence when

compared to strains from other M. tuberculosis families (Lopez et al., 2003; Tsenova et al., 2005).

Page 130: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

110 Chapter 4

Page 131: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

REFERENCES

Abebe, F., and Bjune, G. (2006). The emergence of Beijing family genotypes of Mycobacterium tuberculosis and low-level protection by bacille Calmette-Guerin (BCG) vaccines: is there a link? Clinical and experimental immunology 145, 389-397. Achtman, M. (2008). Evolution, population structure, and phylogeography of genetically monomorphic bacterial pathogens. Annual review of microbiology 62, 53-70. Adams, L.B., Dinauer, M.C., Morgenstern, D.E., and Krahenbuhl, J.L. (1997). Comparison of the roles of reactive oxygen and nitrogen intermediates in the host response to Mycobacterium tuberculosis using transgenic mice. Tubercle and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease 78, 237-246. Aguilar, D., Hanekom, M., Mata, D., Gey van Pittius, N.C., van Helden, P.D., Warren, R.M., and Hernandez-Pando, R. (2010). Mycobacterium tuberculosis strains with the Beijing genotype demonstrate variability in virulence associated with transmission. Tuberculosis 90, 319-325. Akhter, Y., Ehebauer, M.T., Mukhopadhyay, S., and Hasnain, S.E. (2012). The PE/PPE multigene family codes for virulence factors and is a possible source of mycobacterial antigenic variation: perhaps more? Biochimie 94, 110-116. Akif, M., Khare, G., Tyagi, A.K., Mande, S.C., and Sardesai, A.A. (2008). Functional studies of multiple thioredoxins from Mycobacterium tuberculosis. Journal of bacteriology 190, 7087-7095. Akira, S., Uematsu, S., and Takeuchi, O. (2006). Pathogen recognition and innate immunity. Cell 124, 783-801. Alam, M.S., Garg, S.K., and Agrawal, P. (2009). Studies on structural and functional divergence among seven WhiB proteins of Mycobacterium tuberculosis H37Rv. The FEBS journal 276, 76-93. Alland, D., Lacher, D.W., Hazbon, M.H., Motiwala, A.S., Qi, W., Fleischmann, R.D., and Whittam, T.S. (2007). Role of large sequence polymorphisms (LSPs) in generating genomic diversity among clinical isolates of Mycobacterium tuberculosis and the utility of LSPs in phylogenetic analysis. Journal of clinical microbiology 45, 39-46. Andersson, D.I., and Levin, B.R. (1999). The biological cost of antibiotic resistance. Current opinion in microbiology 2, 489-493. Armstrong, J.A., and Hart, P.D. (1971). Response of cultured macrophages to Mycobacterium tuberculosis, with observations on fusion of lysosomes with phagosomes. The Journal of experimental medicine 134, 713-740. Armstrong, J.A., and Hart, P.D. (1975). Phagosome-lysosome interactions in cultured macrophages infected with virulent tubercle bacilli. Reversal of the usual nonfusion pattern and observations on bacterial survival. The Journal of experimental medicine 142, 1-16. Astarie-Dequeker, C., N'Diaye, E.N., Le Cabec, V., Rittig, M.G., Prandi, J., and Maridonneau-Parini, I. (1999). The mannose receptor mediates uptake of pathogenic and nonpathogenic mycobacteria and bypasses bactericidal responses in human macrophages. Infection and immunity 67, 469-477. Aston, C., Rom, W.N., Talbot, A.T., and Reibman, J. (1998). Early inhibition of mycobacterial growth by human alveolar macrophages is not due to nitric oxide. American journal of respiratory and critical care medicine 157, 1943-1950. Bach, H., Papavinasasundaram, K.G., Wong, D., Hmama, Z., and Av-Gay, Y. (2008). Mycobacterium tuberculosis virulence is mediated by PtpA dephosphorylation of human vacuolar protein sorting 33B. Cell host & microbe 3, 316-322. Baker, L., Brown, T., Maiden, M.C., and Drobniewski, F. (2004). Silent nucleotide polymorphisms and a phylogeny for Mycobacterium tuberculosis. Emerging infectious diseases 10, 1568-1577. Bandelt, H.J., Forster, P., and Rohl, A. (1999). Median-joining networks for inferring intraspecific phylogenies. Molecular biology and evolution 16, 37-48.

Page 132: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

112 References

Bange, F.C., Collins, F.M., and Jacobs, W.R., Jr. (1999). Survival of mice infected with Mycobacterium smegmatis containing large DNA fragments from Mycobacterium tuberculosis. Tubercle and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease 79, 171-180. Baranov, P.V., Gurvich, O.L., Fayet, O., Prere, M.F., Miller, W.A., Gesteland, R.F., Atkins, J.F., and Giddings, M.C. (2001). RECODE: a database of frameshifting, bypassing and codon redefinition utilized for gene expression. Nucleic Acids Res 29, 264-267. Barreiro, L.B., Neyrolles, O., Babb, C.L., Tailleux, L., Quach, H., McElreavey, K., Helden, P.D., Hoal, E.G., Gicquel, B., and Quintana-Murci, L. (2006). Promoter variation in the DC-SIGN-encoding gene CD209 is associated with tuberculosis. PLoS medicine 3, e20. Bedard, K., and Krause, K.H. (2007). The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiological reviews 87, 245-313. Bergval, I.L., Klatser, P.R., Schuitema, A.R., Oskam, L., and Anthony, R.M. (2007). Specific mutations in the Mycobacterium tuberculosis rpoB gene are associated with increased dnaE2 expression. FEMS microbiology letters 275, 338-343. Bifani, P.J., Mathema, B., Kurepina, N.E., and Kreiswirth, B.N. (2002). Global dissemination of the Mycobacterium tuberculosis W-Beijing family strains. Trends in microbiology 10, 45-52. Bifani, P.J., Plikaytis, B.B., Kapur, V., Stockbauer, K., Pan, X., Lutfey, M.L., Moghazeh, S.L., Eisner, W., Daniel, T.M., Kaplan, M.H., et al. (1996). Origin and interstate spread of a New York City multidrug-resistant Mycobacterium tuberculosis clone family. JAMA : the journal of the American Medical Association 275, 452-457. Biswas, T., Small, J., Vandal, O., Odaira, T., Deng, H., Ehrt, S., and Tsodikov, O.V. (2010). Structural insight into serine protease Rv3671c that Protects M. tuberculosis from oxidative and acidic stress. Structure 18, 1353-1363. Bjelland, S., Bjoras, M., and Seeberg, E. (1993). Excision of 3-methylguanine from alkylated DNA by 3-methyladenine DNA glycosylase I of Escherichia coli. Nucleic Acids Res 21, 2045-2049. Boshoff, H.I., Myers, T.G., Copp, B.R., McNeil, M.R., Wilson, M.A., and Barry, C.E., 3rd (2004). The transcriptional responses of Mycobacterium tuberculosis to inhibitors of metabolism: novel insights into drug mechanisms of action. The Journal of biological chemistry 279, 40174-40184. Boshoff, H.I., Reed, M.B., Barry, C.E., 3rd, and Mizrahi, V. (2003). DnaE2 polymerase contributes to in vivo survival and the emergence of drug resistance in Mycobacterium tuberculosis. Cell 113, 183-193. Brennan, P.J., and Nikaido, H. (1995). The envelope of mycobacteria. Annual review of biochemistry 64, 29-63. Brodin, P., Poquet, Y., Levillain, F., Peguillet, I., Larrouy-Maumus, G., Gilleron, M., Ewann, F., Christophe, T., Fenistein, D., Jang, J., et al. (2010). High content phenotypic cell-based visual screen identifies Mycobacterium tuberculosis acyltrehalose-containing glycolipids involved in phagosome remodeling. PLoS pathogens 6, e1001100. Brooks, P.C., Movahedzadeh, F., and Davis, E.O. (2001). Identification of some DNA damage-inducible genes of Mycobacterium tuberculosis: apparent lack of correlation with LexA binding. Journal of bacteriology 183, 4459-4467. Brosch, R., Gordon, S.V., Marmiesse, M., Brodin, P., Buchrieser, C., Eiglmeier, K., Garnier, T., Gutierrez, C., Hewinson, G., Kremer, K., et al. (2002). A new evolutionary scenario for the Mycobacterium tuberculosis complex. Proceedings of the National Academy of Sciences of the United States of America 99, 3684-3689. Brudey, K., Driscoll, J.R., Rigouts, L., Prodinger, W.M., Gori, A., Al-Hajoj, S.A., Allix, C., Aristimuno, L., Arora, J., Baumanis, V., et al. (2006). Mycobacterium tuberculosis complex genetic diversity: mining the fourth international spoligotyping database (SpolDB4) for classification, population genetics and epidemiology. BMC microbiology 6, 23. Bryk, R., Lima, C.D., Erdjument-Bromage, H., Tempst, P., and Nathan, C. (2002). Metabolic enzymes of mycobacteria linked to antioxidant defense by a thioredoxin-like protein. Science 295, 1073-1077.

Page 133: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

113 References

Buchmeier, N.A., Libby, S.J., Xu, Y., Loewen, P.C., Switala, J., Guiney, D.G., and Fang, F.C. (1995). DNA repair is more important than catalase for Salmonella virulence in mice. The Journal of clinical investigation 95, 1047-1053. Buchmeier, N.A., Newton, G.L., and Fahey, R.C. (2006). A mycothiol synthase mutant of Mycobacterium tuberculosis has an altered thiol-disulfide content and limited tolerance to stress. Journal of bacteriology 188, 6245-6252. Buchmeier, N.A., Newton, G.L., Koledin, T., and Fahey, R.C. (2003). Association of mycothiol with protection of Mycobacterium tuberculosis from toxic oxidants and antibiotics. Molecular microbiology 47, 1723-1732. Bustamante, J., Aksu, G., Vogt, G., de Beaucoudrey, L., Genel, F., Chapgier, A., Filipe-Santos, O., Feinberg, J., Emile, J.F., Kutukculer, N., et al. (2007). BCG-osis and tuberculosis in a child with chronic granulomatous disease. The Journal of allergy and clinical immunology 120, 32-38. Bylund, J., Brown, K.L., Movitz, C., Dahlgren, C., and Karlsson, A. (2010). Intracellular generation of superoxide by the phagocyte NADPH oxidase: how, where, and what for? Free radical biology & medicine 49, 1834-1845. Cabusora, L., Sutton, E., Fulmer, A., and Forst, C.V. (2005). Differential network expression during drug and stress response. Bioinformatics 21, 2898-2905. Calmette, A. (1931). Preventive Vaccination Against Tuberculosis with BCG. Proceedings of the Royal Society of Medicine 24, 1481-1490. Camacho, L.R., Constant, P., Raynaud, C., Laneelle, M.A., Triccas, J.A., Gicquel, B., Daffe, M., and Guilhot, C. (2001). Analysis of the phthiocerol dimycocerosate locus of Mycobacterium tuberculosis. Evidence that this lipid is involved in the cell wall permeability barrier. The Journal of biological chemistry 276, 19845-19854. Camacho, L.R., Ensergueix, D., Perez, E., Gicquel, B., and Guilhot, C. (1999). Identification of a virulence gene cluster of Mycobacterium tuberculosis by signature-tagged transposon mutagenesis. Molecular microbiology 34, 257-267. Caminero, J.A., Pena, M.J., Campos-Herrero, M.I., Rodriguez, J.C., Garcia, I., Cabrera, P., Lafoz, C., Samper, S., Takiff, H., Afonso, O., et al. (2001). Epidemiological evidence of the spread of a Mycobacterium tuberculosis strain of the Beijing genotype on Gran Canaria Island. American journal of respiratory and critical care medicine 164, 1165-1170. Cappelli, G., Volpe, E., Grassi, M., Liseo, B., Colizzi, V., and Mariani, F. (2006). Profiling of Mycobacterium tuberculosis gene expression during human macrophage infection: upregulation of the alternative sigma factor G, a group of transcriptional regulators, and proteins with unknown function. Research in microbiology 157, 445-455. Catalao, M.J., Gil, F., Moniz-Pereira, J., Sao-Jose, C., and Pimentel, M. (2012). Diversity in bacterial lysis systems: bacteriophages show the way. FEMS microbiology reviews. Caws, M., Thwaites, G., Dunstan, S., Hawn, T.R., Lan, N.T., Thuong, N.T., Stepniewska, K., Huyen, M.N., Bang, N.D., Loc, T.H., et al. (2008). The influence of host and bacterial genotype on the development of disseminated disease with Mycobacterium tuberculosis. PLoS pathogens 4, e1000034. Chan, J., Fan, X.D., Hunter, S.W., Brennan, P.J., and Bloom, B.R. (1991). Lipoarabinomannan, a possible virulence factor involved in persistence of Mycobacterium tuberculosis within macrophages. Infection and immunity 59, 1755-1761. Chan, J., Fujiwara, T., Brennan, P., McNeil, M., Turco, S.J., Sibille, J.C., Snapper, M., Aisen, P., and Bloom, B.R. (1989). Microbial glycolipids: possible virulence factors that scavenge oxygen radicals. Proceedings of the National Academy of Sciences of the United States of America 86, 2453-2457. Chan, J., Xing, Y., Magliozzo, R.S., and Bloom, B.R. (1992). Killing of virulent Mycobacterium tuberculosis by reactive nitrogen intermediates produced by activated murine macrophages. The Journal of experimental medicine 175, 1111-1122. Chao, J., Wong, D., Zheng, X., Poirier, V., Bach, H., Hmama, Z., and Av-Gay, Y. (2010). Protein kinase and phosphatase signaling in Mycobacterium tuberculosis physiology and pathogenesis. Biochimica et biophysica acta 1804, 620-627.

Page 134: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

114 References

Chaturvedi, R., Bansal, K., Narayana, Y., Kapoor, N., Sukumar, N., Togarsimalemath, S.K., Chandra, N., Mishra, S., Ajitkumar, P., Joshi, B., et al. (2010). The multifunctional PE_PGRS11 protein from Mycobacterium tuberculosis plays a role in regulating resistance to oxidative stress. The Journal of biological chemistry 285, 30389-30403. Chen, J., Tsolaki, A.G., Shen, X., Jiang, X., Mei, J., and Gao, Q. (2007). Deletion-targeted multiplex PCR (DTM-PCR) for identification of Beijing/W genotypes of Mycobacterium tuberculosis. Tuberculosis 87, 446-449. Cheng, K.C., Cahill, D.S., Kasai, H., Nishimura, S., and Loeb, L.A. (1992). 8-Hydroxyguanine, an abundant form of oxidative DNA damage, causes G----T and A----C substitutions. The Journal of biological chemistry 267, 166-172. Choi, H.S., Rai, P.R., Chu, H.W., Cool, C., and Chan, E.D. (2002). Analysis of nitric oxide synthase and nitrotyrosine expression in human pulmonary tuberculosis. American journal of respiratory and critical care medicine 166, 178-186. Cirillo, S.L., Subbian, S., Chen, B., Weisbrod, T.R., Jacobs, W.R., Jr., and Cirillo, J.D. (2009). Protection of Mycobacterium tuberculosis from reactive oxygen species conferred by the mel2 locus impacts persistence and dissemination. Infection and immunity 77, 2557-2567. Clay, H., Davis, J.M., Beery, D., Huttenlocher, A., Lyons, S.E., and Ramakrishnan, L. (2007). Dichotomous role of the macrophage in early Mycobacterium marinum infection of the zebrafish. Cell host & microbe 2, 29-39. Clemens, D.L., and Horwitz, M.A. (1996). The Mycobacterium tuberculosis phagosome interacts with early endosomes and is accessible to exogenously administered transferrin. The Journal of experimental medicine 184, 1349-1355. Colangeli, R., Haq, A., Arcus, V.L., Summers, E., Magliozzo, R.S., McBride, A., Mitra, A.K., Radjainia, M., Khajo, A., Jacobs, W.R., Jr., et al. (2009). The multifunctional histone-like protein Lsr2 protects mycobacteria against reactive oxygen intermediates. Proceedings of the National Academy of Sciences of the United States of America 106, 4414-4418. Colditz, G.A., Brewer, T.F., Berkey, C.S., Wilson, M.E., Burdick, E., Fineberg, H.V., and Mosteller, F. (1994). Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature. JAMA : the journal of the American Medical Association 271, 698-702. Cole, S.T., Brosch, R., Parkhill, J., Garnier, T., Churcher, C., Harris, D., Gordon, S.V., Eiglmeier, K., Gas, S., Barry, C.E., 3rd, et al. (1998). Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature 393, 537-544. Comas, I., Chakravartti, J., Small, P.M., Galagan, J., Niemann, S., Kremer, K., Ernst, J.D., and Gagneux, S. (2010). Human T cell epitopes of Mycobacterium tuberculosis are evolutionarily hyperconserved. Nature genetics 42, 498-503. Comas, I., and Gagneux, S. (2009). The past and future of tuberculosis research. PLoS pathogens 5, e1000600. Comas, I., Homolka, S., Niemann, S., and Gagneux, S. (2009). Genotyping of genetically monomorphic bacteria: DNA sequencing in Mycobacterium tuberculosis highlights the limitations of current methodologies. PloS one 4, e7815. Converse, S.E., Mougous, J.D., Leavell, M.D., Leary, J.A., Bertozzi, C.R., and Cox, J.S. (2003). MmpL8 is required for sulfolipid-1 biosynthesis and Mycobacterium tuberculosis virulence. Proceedings of the National Academy of Sciences of the United States of America 100, 6121-6126. Cooper, A.M., Pearl, J.E., Brooks, J.V., Ehlers, S., and Orme, I.M. (2000). Expression of the nitric oxide synthase 2 gene is not essential for early control of Mycobacterium tuberculosis in the murine lung. Infection and immunity 68, 6879-6882. Coscolla, M., and Gagneux, S. (2010). Does M. tuberculosis genomic diversity explain disease diversity? Drug discovery today Disease mechanisms 7, e43-e59. Cox, J.S., Chen, B., McNeil, M., and Jacobs, W.R., Jr. (1999). Complex lipid determines tissue-specific replication of Mycobacterium tuberculosis in mice. Nature 402, 79-83. Craig, M., and Slauch, J.M. (2009). Phagocytic superoxide specifically damages an extracytoplasmic target to inhibit or kill Salmonella. PloS one 4, e4975.

Page 135: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

115 References

Daffe, M., and Draper, P. (1998). The envelope layers of mycobacteria with reference to their pathogenicity. Advances in microbial physiology 39, 131-203. Daniel, T.M. (2006). The history of tuberculosis. Respiratory medicine 100, 1862-1870. Darwin, K.H., Ehrt, S., Gutierrez-Ramos, J.C., Weich, N., and Nathan, C.F. (2003). The proteasome of Mycobacterium tuberculosis is required for resistance to nitric oxide. Science 302, 1963-1966. Darwin, K.H., and Nathan, C.F. (2005). Role for nucleotide excision repair in virulence of Mycobacterium tuberculosis. Infection and immunity 73, 4581-4587. David, S.S., O'Shea, V.L., and Kundu, S. (2007). Base-excision repair of oxidative DNA damage. Nature 447, 941-950. Day, J.H., Grant, A.D., Fielding, K.L., Morris, L., Moloi, V., Charalambous, S., Puren, A.J., Chaisson, R.E., De Cock, K.M., Hayes, R.J., et al. (2004). Does tuberculosis increase HIV load? The Journal of infectious diseases 190, 1677-1684. de Hostos, E.L. (1999). The coronin family of actin-associated proteins. Trends in cell biology 9, 345-350. de Jong, B.C., Hill, P.C., Brookes, R.H., Gagneux, S., Jeffries, D.J., Otu, J.K., Donkor, S.A., Fox, A., McAdam, K.P., Small, P.M., et al. (2006). Mycobacterium africanum elicits an attenuated T cell response to early secreted antigenic target, 6 kDa, in patients with tuberculosis and their household contacts. The Journal of infectious diseases 193, 1279-1286. de Laat, W.L., Jaspers, N.G., and Hoeijmakers, J.H. (1999). Molecular mechanism of nucleotide excision repair. Genes Dev 13, 768-785. Deghmane, A.E., Soualhine, H., Bach, H., Sendide, K., Itoh, S., Tam, A., Noubir, S., Talal, A., Lo, R., Toyoshima, S., et al. (2007). Lipoamide dehydrogenase mediates retention of coronin-1 on BCG vacuoles, leading to arrest in phagosome maturation. Journal of cell science 120, 2796-2806. Della, M., Palmbos, P.L., Tseng, H.M., Tonkin, L.M., Daley, J.M., Topper, L.M., Pitcher, R.S., Tomkinson, A.E., Wilson, T.E., and Doherty, A.J. (2004). Mycobacterial Ku and ligase proteins constitute a two-component NHEJ repair machine. Science 306, 683-685. Demple, B., and Harrison, L. (1994). Repair of oxidative damage to DNA: enzymology and biology. Annual review of biochemistry 63, 915-948. Denamur, E., and Matic, I. (2006). Evolution of mutation rates in bacteria. Molecular microbiology 60, 820-827. Denu, J.M., and Tanner, K.G. (1998). Specific and reversible inactivation of protein tyrosine phosphatases by hydrogen peroxide: evidence for a sulfenic acid intermediate and implications for redox regulation. Biochemistry 37, 5633-5642. Deretic, V., Philipp, W., Dhandayuthapani, S., Mudd, M.H., Curcic, R., Garbe, T., Heym, B., Via, L.E., and Cole, S.T. (1995). Mycobacterium tuberculosis is a natural mutant with an inactivated oxidative-stress regulatory gene: implications for sensitivity to isoniazid. Molecular microbiology 17, 889-900. Desjardins, M., Huber, L.A., Parton, R.G., and Griffiths, G. (1994). Biogenesis of phagolysosomes proceeds through a sequential series of interactions with the endocytic apparatus. The Journal of cell biology 124, 677-688. Dianov, G., and Lindahl, T. (1994). Reconstitution of the DNA base excision-repair pathway. Curr Biol 4, 1069-1076. Dizdaroglu, M. (2005). Base-excision repair of oxidative DNA damage by DNA glycosylases. Mutation research 591, 45-59. Domenech, P., Honore, N., Heym, B., and Cole, S.T. (2001). Role of OxyS of Mycobacterium tuberculosis in oxidative stress: overexpression confers increased sensitivity to organic hydroperoxides. Microbes and infection / Institut Pasteur 3, 713-721. Domenech, P., Reed, M.B., and Barry, C.E., 3rd (2005). Contribution of the Mycobacterium tuberculosis MmpL protein family to virulence and drug resistance. Infection and immunity 73, 3492-3501. Domenech, P., Reed, M.B., Dowd, C.S., Manca, C., Kaplan, G., and Barry, C.E., 3rd (2004). The role of MmpL8 in sulfatide biogenesis and virulence of Mycobacterium tuberculosis. The Journal of biological chemistry 279, 21257-21265. Dormans, J., Burger, M., Aguilar, D., Hernandez-Pando, R., Kremer, K., Roholl, P., Arend, S.M., and van Soolingen, D. (2004). Correlation of virulence, lung pathology, bacterial load and delayed type

Page 136: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

116 References

hypersensitivity responses after infection with different Mycobacterium tuberculosis genotypes in a BALB/c mouse model. Clinical and experimental immunology 137, 460-468. Dos Vultos, T., Blazquez, J., Rauzier, J., Matic, I., and Gicquel, B. (2006). Identification of Nudix hydrolase family members with an antimutator role in Mycobacterium tuberculosis and Mycobacterium smegmatis. Journal of bacteriology 188, 3159-3161. Dos Vultos, T., Mestre, O., Rauzier, J., Golec, M., Rastogi, N., Rasolofo, V., Tonjum, T., Sola, C., Matic, I., and Gicquel, B. (2008). Evolution and diversity of clonal bacteria: the paradigm of Mycobacterium tuberculosis. PloS one 3, e1538. Dos Vultos, T., Mestre, O., Tonjum, T., and Gicquel, B. (2009). DNA repair in Mycobacterium tuberculosis revisited. FEMS microbiology reviews 33, 471-487. Drennan, M.B., Nicolle, D., Quesniaux, V.J., Jacobs, M., Allie, N., Mpagi, J., Fremond, C., Wagner, H., Kirschning, C., and Ryffel, B. (2004). Toll-like receptor 2-deficient mice succumb to Mycobacterium tuberculosis infection. The American journal of pathology 164, 49-57. Durbach, S.I., Springer, B., Machowski, E.E., North, R.J., Papavinasasundaram, K.G., Colston, M.J., Bottger, E.C., and Mizrahi, V. (2003). DNA alkylation damage as a sensor of nitrosative stress in Mycobacterium tuberculosis. Infection and immunity 71, 997-1000. Dussurget, O., Stewart, G., Neyrolles, O., Pescher, P., Young, D., and Marchal, G. (2001). Role of Mycobacterium tuberculosis copper-zinc superoxide dismutase. Infection and immunity 69, 529-533. Dutta, N.K., Mehra, S., Didier, P.J., Roy, C.J., Doyle, L.A., Alvarez, X., Ratterree, M., Be, N.A., Lamichhane, G., Jain, S.K., et al. (2010). Genetic requirements for the survival of tubercle bacilli in primates. The Journal of infectious diseases 201, 1743-1752. Dworkin, M., Falkow, S., Rosenberg, E., Schleifer, K.-H., and Stackebrandt, E. (2006). The Prokaryotes: Proteobacteria:gamma subclass, Vol 3 (Springer). Ebrahimi-Rad, M., Bifani, P., Martin, C., Kremer, K., Samper, S., Rauzier, J., Kreiswirth, B., Blazquez, J., Jouan, M., van Soolingen, D., et al. (2003). Mutations in putative mutator genes of Mycobacterium tuberculosis strains of the W-Beijing family. Emerging infectious diseases 9, 838-845. Edwards, K.M., Cynamon, M.H., Voladri, R.K., Hager, C.C., DeStefano, M.S., Tham, K.T., Lakey, D.L., Bochan, M.R., and Kernodle, D.S. (2001). Iron-cofactored superoxide dismutase inhibits host responses to Mycobacterium tuberculosis. American journal of respiratory and critical care medicine 164, 2213-2219. Ehrt, S., and Schnappinger, D. (2009). Mycobacterial survival strategies in the phagosome: defence against host stresses. Cellular microbiology 11, 1170-1178. Embley, T.M., and Stackebrandt, E. (1994). The molecular phylogeny and systematics of the actinomycetes. Annual review of microbiology 48, 257-289. Ezraty, B., Bos, J., Barras, F., and Aussel, L. (2005). Methionine sulfoxide reduction and assimilation in Escherichia coli: new role for the biotin sulfoxide reductase BisC. Journal of bacteriology 187, 231-237. Facchetti, F., Vermi, W., Fiorentini, S., Chilosi, M., Caruso, A., Duse, M., Notarangelo, L.D., and Badolato, R. (1999). Expression of inducible nitric oxide synthase in human granulomas and histiocytic reactions. The American journal of pathology 154, 145-152. Fallow, A., Domenech, P., and Reed, M.B. (2010). Strains of the East Asian (W/Beijing) lineage of Mycobacterium tuberculosis are DosS/DosT-DosR two-component regulatory system natural mutants. Journal of bacteriology 192, 2228-2238. Fang, F.C. (2004). Antimicrobial reactive oxygen and nitrogen species: concepts and controversies. Nature reviews Microbiology 2, 820-832. Fenner, L., Egger, M., Bodmer, T., Altpeter, E., Zwahlen, M., Jaton, K., Pfyffer, G.E., Borrell, S., Dubuis, O., Bruderer, T., et al. (2012). Effect of mutation and genetic background on drug resistance in Mycobacterium tuberculosis. Antimicrobial agents and chemotherapy 56, 3047-3053. Ferrer, N.L., Gomez, A.B., Neyrolles, O., Gicquel, B., and Martin, C. (2010). Interactions of attenuated Mycobacterium tuberculosis phoP mutant with human macrophages. PloS one 5, e12978. Ferwerda, G., Girardin, S.E., Kullberg, B.J., Le Bourhis, L., de Jong, D.J., Langenberg, D.M., van Crevel, R., Adema, G.J., Ottenhoff, T.H., Van der Meer, J.W., et al. (2005). NOD2 and toll-like receptors are nonredundant recognition systems of Mycobacterium tuberculosis. PLoS pathogens 1, 279-285.

Page 137: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

117 References

Filliol, I., Driscoll, J.R., Van Soolingen, D., Kreiswirth, B.N., Kremer, K., Valetudie, G., Anh, D.D., Barlow, R., Banerjee, D., Bifani, P.J., et al. (2002). Global distribution of Mycobacterium tuberculosis spoligotypes. Emerging infectious diseases 8, 1347-1349. Filliol, I., Motiwala, A.S., Cavatore, M., Qi, W., Hazbon, M.H., Bobadilla del Valle, M., Fyfe, J., Garcia-Garcia, L., Rastogi, N., Sola, C., et al. (2006). Global phylogeny of Mycobacterium tuberculosis based on single nucleotide polymorphism (SNP) analysis: insights into tuberculosis evolution, phylogenetic accuracy of other DNA fingerprinting systems, and recommendations for a minimal standard SNP set. Journal of bacteriology 188, 759-772. Fine, P.E. (1995). Variation in protection by BCG: implications of and for heterologous immunity. Lancet 346, 1339-1345. Firmani, M.A., and Riley, L.W. (2002). Mycobacterium tuberculosis CDC1551 is resistant to reactive nitrogen and oxygen intermediates in vitro. Infection and immunity 70, 3965-3968. Flannagan, R.S., Cosio, G., and Grinstein, S. (2009). Antimicrobial mechanisms of phagocytes and bacterial evasion strategies. Nature reviews Microbiology 7, 355-366. Fleischmann, R.D., Alland, D., Eisen, J.A., Carpenter, L., White, O., Peterson, J., DeBoy, R., Dodson, R., Gwinn, M., Haft, D., et al. (2002). Whole-genome comparison of Mycobacterium tuberculosis clinical and laboratory strains. Journal of bacteriology 184, 5479-5490. Flynn, J.L., Chan, J., and Lin, P.L. (2011). Macrophages and control of granulomatous inflammation in tuberculosis. Mucosal immunology 4, 271-278. Fontan, P., Aris, V., Ghanny, S., Soteropoulos, P., and Smith, I. (2008). Global transcriptional profile of Mycobacterium tuberculosis during THP-1 human macrophage infection. Infection and immunity 76, 717-725. Friedberg, E.C., Walker, G.C., Siede, W., Wood, R.D., Schultz, R.A., and Ellenberger, T. (2006). DNA repair and mutagenesis, 2nd ed. edn (ASM press, Washington, DC.). Frothingham, R., and Meeker-O'Connell, W.A. (1998). Genetic diversity in the Mycobacterium tuberculosis complex based on variable numbers of tandem DNA repeats. Microbiology 144 ( Pt 5), 1189-1196. Fu, L.M., and Shinnick, T.M. (2007). Genome-wide exploration of the drug action of capreomycin on Mycobacterium tuberculosis using Affymetrix oligonucleotide GeneChips. The Journal of infection 54, 277-284. Gagneux, S., DeRiemer, K., Van, T., Kato-Maeda, M., de Jong, B.C., Narayanan, S., Nicol, M., Niemann, S., Kremer, K., Gutierrez, M.C., et al. (2006a). Variable host-pathogen compatibility in Mycobacterium tuberculosis. Proceedings of the National Academy of Sciences of the United States of America 103, 2869-2873. Gagneux, S., Long, C.D., Small, P.M., Van, T., Schoolnik, G.K., and Bohannan, B.J. (2006b). The competitive cost of antibiotic resistance in Mycobacterium tuberculosis. Science 312, 1944-1946. Gamulin, V., Cetkovic, H., and Ahel, I. (2004). Identification of a promoter motif regulating the major DNA damage response mechanism of Mycobacterium tuberculosis. FEMS microbiology letters 238, 57-63. Gandhi, N.R., Nunn, P., Dheda, K., Schaaf, H.S., Zignol, M., van Soolingen, D., Jensen, P., and Bayona, J. (2010). Multidrug-resistant and extensively drug-resistant tuberculosis: a threat to global control of tuberculosis. Lancet 375, 1830-1843. Garbe, T.R., Hibler, N.S., and Deretic, V. (1996). Response of Mycobacterium tuberculosis to reactive oxygen and nitrogen intermediates. Mol Med 2, 134-142. Gebhard, S., Humpel, A., McLellan, A.D., and Cook, G.M. (2008). The alternative sigma factor SigF of Mycobacterium smegmatis is required for survival of heat shock, acidic pH and oxidative stress. Microbiology 154, 2786-2795. Geijtenbeek, T.B., Van Vliet, S.J., Koppel, E.A., Sanchez-Hernandez, M., Vandenbroucke-Grauls, C.M., Appelmelk, B., and Van Kooyk, Y. (2003). Mycobacteria target DC-SIGN to suppress dendritic cell function. The Journal of experimental medicine 197, 7-17.

Page 138: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

118 References

Geiman, D.E., Raghunand, T.R., Agarwal, N., and Bishai, W.R. (2006). Differential gene expression in response to exposure to antimycobacterial agents and other stress conditions among seven Mycobacterium tuberculosis whiB-like genes. Antimicrobial agents and chemotherapy 50, 2836-2841. Glynn, J.R., Whiteley, J., Bifani, P.J., Kremer, K., and van Soolingen, D. (2002). Worldwide occurrence of Beijing/W strains of Mycobacterium tuberculosis: a systematic review. Emerging infectious diseases 8, 843-849. Gong, C., Bongiorno, P., Martins, A., Stephanou, N.C., Zhu, H., Shuman, S., and Glickman, M.S. (2005). Mechanism of nonhomologous end-joining in mycobacteria: a low-fidelity repair system driven by Ku, ligase D and ligase C. Nat Struct Mol Biol 12, 304-312. Gonzalo-Asensio, J., Mostowy, S., Harders-Westerveen, J., Huygen, K., Hernandez-Pando, R., Thole, J., Behr, M., Gicquel, B., and Martin, C. (2008). PhoP: a missing piece in the intricate puzzle of Mycobacterium tuberculosis virulence. PloS one 3, e3496. Graham, J.E., and Clark-Curtiss, J.E. (1999). Identification of Mycobacterium tuberculosis RNAs synthesized in response to phagocytosis by human macrophages by selective capture of transcribed sequences (SCOTS). Proceedings of the National Academy of Sciences of the United States of America 96, 11554-11559. Grant, S.S., Kaufmann, B.B., Chand, N.S., Haseley, N., and Hung, D.T. (2012). Eradication of bacterial persisters with antibiotic-generated hydroxyl radicals. Proceedings of the National Academy of Sciences of the United States of America 109, 12147-12152. Griffiths, G., Nystrom, B., Sable, S.B., and Khuller, G.K. (2010). Nanobead-based interventions for the treatment and prevention of tuberculosis. Nature reviews Microbiology 8, 827-834. Grzegorzewicz, A.E., Pham, H., Gundi, V.A., Scherman, M.S., North, E.J., Hess, T., Jones, V., Gruppo, V., Born, S.E., Kordulakova, J., et al. (2012). Inhibition of mycolic acid transport across the Mycobacterium tuberculosis plasma membrane. Nature chemical biology 8, 334-341. Guo, Y., Bandaru, V., Jaruga, P., Zhao, X., Burrows, C.J., Iwai, S., Dizdaroglu, M., Bond, J.P., and Wallace, S.S. (2010). The oxidative DNA glycosylases of Mycobacterium tuberculosis exhibit different substrate preferences from their Escherichia coli counterparts. DNA repair 9, 177-190. Gurtler, V., Mayall, B.C., and Seviour, R. (2004). Can whole genome analysis refine the taxonomy of the genus Rhodococcus? FEMS microbiology reviews 28, 377-403. Gutacker, M.M., Mathema, B., Soini, H., Shashkina, E., Kreiswirth, B.N., Graviss, E.A., and Musser, J.M. (2006). Single-nucleotide polymorphism-based population genetic analysis of Mycobacterium tuberculosis strains from 4 geographic sites. The Journal of infectious diseases 193, 121-128. Gutacker, M.M., Smoot, J.C., Migliaccio, C.A., Ricklefs, S.M., Hua, S., Cousins, D.V., Graviss, E.A., Shashkina, E., Kreiswirth, B.N., and Musser, J.M. (2002). Genome-wide analysis of synonymous single nucleotide polymorphisms in Mycobacterium tuberculosis complex organisms: resolution of genetic relationships among closely related microbial strains. Genetics 162, 1533-1543. Guthlein, C., Wanner, R.M., Sander, P., Davis, E.O., Bosshard, M., Jiricny, J., Bottger, E.C., and Springer, B. (2008). Characterisation of the mycobacterial NER system reveals novel functions of uvrD1 helicase. Journal of bacteriology. Gutierrez, M.C., Brisse, S., Brosch, R., Fabre, M., Omais, B., Marmiesse, M., Supply, P., and Vincent, V. (2005). Ancient origin and gene mosaicism of the progenitor of Mycobacterium tuberculosis. PLoS pathogens 1, e5. Hanekom, M., van der Spuy, G.D., Streicher, E., Ndabambi, S.L., McEvoy, C.R., Kidd, M., Beyers, N., Victor, T.C., van Helden, P.D., and Warren, R.M. (2007). A recently evolved sublineage of the Mycobacterium tuberculosis Beijing strain family is associated with an increased ability to spread and cause disease. Journal of clinical microbiology 45, 1483-1490. Harries, A.D., and Dye, C. (2006). Tuberculosis. Ann Trop Med Parasitol 100, 415-431. Hershberg, R., Lipatov, M., Small, P.M., Sheffer, H., Niemann, S., Homolka, S., Roach, J.C., Kremer, K., Petrov, D.A., Feldman, M.W., et al. (2008). High functional diversity in Mycobacterium tuberculosis driven by genetic drift and human demography. PLoS Biol 6, e311.

Page 139: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

119 References

Heym, B., Honore, N., Truffot-Pernot, C., Banerjee, A., Schurra, C., Jacobs, W.R., Jr., van Embden, J.D., Grosset, J.H., and Cole, S.T. (1994). Implications of multidrug resistance for the future of short-course chemotherapy of tuberculosis: a molecular study. Lancet 344, 293-298. Heym, B., Zhang, Y., Poulet, S., Young, D., and Cole, S.T. (1993). Characterization of the katG gene encoding a catalase-peroxidase required for the isoniazid susceptibility of Mycobacterium tuberculosis. Journal of bacteriology 175, 4255-4259. Heyworth, P.G., Cross, A.R., and Curnutte, J.T. (2003). Chronic granulomatous disease. Current opinion in immunology 15, 578-584. Hiriyanna, K.T., and Ramakrishnan, T. (1986). Deoxyribonucleic acid replication time in Mycobacterium tuberculosis H37 Rv. Archives of microbiology 144, 105-109. Hoffmann, C., Leis, A., Niederweis, M., Plitzko, J.M., and Engelhardt, H. (2008). Disclosure of the mycobacterial outer membrane: cryo-electron tomography and vitreous sections reveal the lipid bilayer structure. Proceedings of the National Academy of Sciences of the United States of America 105, 3963-3967. Holmgren, A. (2000). Antioxidant function of thioredoxin and glutaredoxin systems. Antioxidants & redox signaling 2, 811-820. Homolka, S., Niemann, S., Russell, D.G., and Rohde, K.H. (2010). Functional genetic diversity among Mycobacterium tuberculosis complex clinical isolates: delineation of conserved core and lineage-specific transcriptomes during intracellular survival. PLoS pathogens 6, e1000988. Houghton, J., Townsend, C., Williams, A.R., Rodgers, A., Rand, L., Walker, K.B., Bottger, E.C., Springer, B., and Davis, E.O. (2012). Important role for Mycobacterium tuberculosis UvrD1 in pathogenesis and persistence apart from its function in nucleotide excision repair. Journal of bacteriology 194, 2916-2923. Hu, Y., Kendall, S., Stoker, N.G., and Coates, A.R. (2004). The Mycobacterium tuberculosis sigJ gene controls sensitivity of the bacterium to hydrogen peroxide. FEMS microbiology letters 237, 415-423. Humphreys, I.R., Stewart, G.R., Turner, D.J., Patel, J., Karamanou, D., Snelgrove, R.J., and Young, D.B. (2006). A role for dendritic cells in the dissemination of mycobacterial infection. Microbes and infection / Institut Pasteur 8, 1339-1346. Imlay, J.A. (2008). Cellular defenses against superoxide and hydrogen peroxide. Annual review of biochemistry 77, 755-776. Imlay, J.A., and Linn, S. (1986). Bimodal pattern of killing of DNA-repair-defective or anoxically grown Escherichia coli by hydrogen peroxide. Journal of bacteriology 166, 519-527. Ishibashi, Y., and Arai, T. (1990). Roles of the complement receptor type 1 (CR1) and type 3 (CR3) on phagocytosis and subsequent phagosome-lysosome fusion in Salmonella-infected murine macrophages. FEMS microbiology immunology 2, 89-96. Iwamoto, T., Yoshida, S., Suzuki, K., Tomita, M., Fujiyama, R., Tanaka, N., Kawakami, Y., and Ito, M. (2007). Hypervariable loci that enhance the discriminatory ability of newly proposed 15-loci and 24-loci variable-number tandem repeat typing method on Mycobacterium tuberculosis strains predominated by the Beijing family. FEMS microbiology letters 270, 67-74. Iwamoto, T., Yoshida, S., Suzuki, K., and Wada, T. (2008). Population structure analysis of the Mycobacterium tuberculosis Beijing family indicates an association between certain sublineages and multidrug resistance. Antimicrobial agents and chemotherapy 52, 3805-3809. Jain, M., and Cox, J.S. (2005). Interaction between polyketide synthase and transporter suggests coupled synthesis and export of virulence lipid in M. tuberculosis. PLoS pathogens 1, e2. Jain, R., Dey, B., Khera, A., Srivastav, P., Gupta, U.D., Katoch, V.M., Ramanathan, V.D., and Tyagi, A.K. (2011). Over-expression of superoxide dismutase obliterates the protective effect of BCG against tuberculosis by modulating innate and adaptive immune responses. Vaccine 29, 8118-8125. Jain, R., Kumar, P., and Varshney, U. (2007). A distinct role of formamidopyrimidine DNA glycosylase (MutM) in down-regulation of accumulation of G, C mutations and protection against oxidative stress in mycobacteria. DNA repair 6, 1774-1785. Jarlier, V., and Nikaido, H. (1994). Mycobacterial cell wall: structure and role in natural resistance to antibiotics. FEMS microbiology letters 123, 11-18.

Page 140: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

120 References

Jung, Y.J., LaCourse, R., Ryan, L., and North, R.J. (2002). Virulent but not avirulent Mycobacterium tuberculosis can evade the growth inhibitory action of a T helper 1-dependent, nitric oxide Synthase 2-independent defense in mice. The Journal of experimental medicine 196, 991-998. Kallenius, G., Koivula, T., Ghebremichael, S., Hoffner, S.E., Norberg, R., Svensson, E., Dias, F., Marklund, B.I., and Svenson, S.B. (1999). Evolution and clonal traits of Mycobacterium tuberculosis complex in Guinea-Bissau. Journal of clinical microbiology 37, 3872-3878. Kamerbeek, J., Schouls, L., Kolk, A., van Agterveld, M., van Soolingen, D., Kuijper, S., Bunschoten, A., Molhuizen, H., Shaw, R., Goyal, M., et al. (1997). Simultaneous detection and strain differentiation of Mycobacterium tuberculosis for diagnosis and epidemiology. Journal of clinical microbiology 35, 907-914. Kang, J., and Blaser, M.J. (2006). Bacterial populations as perfect gases: genomic integrity and diversification tensions in Helicobacter pylori. Nature reviews Microbiology 4, 826-836. Kang, P.B., Azad, A.K., Torrelles, J.B., Kaufman, T.M., Beharka, A., Tibesar, E., DesJardin, L.E., and Schlesinger, L.S. (2005). The human macrophage mannose receptor directs Mycobacterium tuberculosis lipoarabinomannan-mediated phagosome biogenesis. The Journal of experimental medicine 202, 987-999. Kato-Maeda, M., Metcalfe, J.Z., and Flores, L. (2011). Genotyping of Mycobacterium tuberculosis: application in epidemiologic studies. Future microbiology 6, 203-216. Kato-Maeda, M., Shanley, C.A., Ackart, D., Jarlsberg, L.G., Shang, S., Obregon-Henao, A., Harton, M., Basaraba, R.J., Henao-Tamayo, M., Barrozo, J.C., et al. (2012). Beijing Sublineages of Mycobacterium tuberculosis Differ in Pathogenicity in the Guinea Pig. Clinical and vaccine immunology : CVI 19, 1227-1237. Kaufmann, S.H. (2011). Fact and fiction in tuberculosis vaccine research: 10 years later. The Lancet infectious diseases 11, 633-640. Kaufmann, S.H., and McMichael, A.J. (2005). Annulling a dangerous liaison: vaccination strategies against AIDS and tuberculosis. Nature medicine 11, S33-44. Kaur, D., Guerin, M.E., Skovierova, H., Brennan, P.J., and Jackson, M. (2009). Chapter 2: Biogenesis of the cell wall and other glycoconjugates of Mycobacterium tuberculosis. Advances in applied microbiology 69, 23-78. Keren, I., Minami, S., Rubin, E., and Lewis, K. (2011). Characterization and transcriptome analysis of Mycobacterium tuberculosis persisters. mBio 2, e00100-00111. Koch, R. (1882). Die Aetiogie der Tuberculose. Berl Klin Wochenschr 19, 221–230. Koch, R. (1982). Classics in infectious diseases. The etiology of tuberculosis: Robert Koch. Berlin, Germany 1882. Reviews of infectious diseases 4, 1270-1274. Kohanski, M.A., Dwyer, D.J., Hayete, B., Lawrence, C.A., and Collins, J.J. (2007). A common mechanism of cellular death induced by bactericidal antibiotics. Cell 130, 797-810. Kremer, K., Au, B.K., Yip, P.C., Skuce, R., Supply, P., Kam, K.M., and van Soolingen, D. (2005). Use of variable-number tandem-repeat typing to differentiate Mycobacterium tuberculosis Beijing family isolates from Hong Kong and comparison with IS6110 restriction fragment length polymorphism typing and spoligotyping. Journal of clinical microbiology 43, 314-320. Kremer, K., Glynn, J.R., Lillebaek, T., Niemann, S., Kurepina, N.E., Kreiswirth, B.N., Bifani, P.J., and van Soolingen, D. (2004). Definition of the Beijing/W lineage of Mycobacterium tuberculosis on the basis of genetic markers. Journal of clinical microbiology 42, 4040-4049. Kremer, K., van-der-Werf, M.J., Au, B.K., Anh, D.D., Kam, K.M., van-Doorn, H.R., Borgdorff, M.W., and van-Soolingen, D. (2009). Vaccine-induced immunity circumvented by typical Mycobacterium tuberculosis Beijing strains. Emerging infectious diseases 15, 335-339. Kurepina, N.E., Sreevatsan, S., Plikaytis, B.B., Bifani, P.J., Connell, N.D., Donnelly, R.J., van Sooligen, D., Musser, J.M., and Kreiswirth, B.N. (1998). Characterization of the phylogenetic distribution and chromosomal insertion sites of five IS6110 elements in Mycobacterium tuberculosis: non-random integration in the dnaA-dnaN region. Tubercle and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease 79, 31-42.

Page 141: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

121 References

Kurthkoti, K., Kumar, P., Jain, R., and Varshney, U. (2008). Important role of the nucleotide excision repair pathway in Mycobacterium smegmatis in conferring protection against commonly encountered DNA-damaging agents. Microbiology 154, 2776-2785. Kurthkoti, K., Srinath, T., Kumar, P., Malshetty, V.S., Sang, P.B., Jain, R., Manjunath, R., and Varshney, U. (2010). A distinct physiological role of MutY in mutation prevention in mycobacteria. Microbiology 156, 88-93. Lamhamedi-Cherradi, S., de Chastellier, C., and Casanova, J.L. (1999). Growth of Mycobacterium bovis, Bacille Calmette-Guerin, within human monocytes-macrophages cultured in serum-free medium. Journal of immunological methods 225, 75-86. Lamichhane, G., Tyagi, S., and Bishai, W.R. (2005). Designer arrays for defined mutant analysis to detect genes essential for survival of Mycobacterium tuberculosis in mouse lungs. Infection and immunity 73, 2533-2540. Lari, N., Rindi, L., Bonanni, D., Tortoli, E., and Garzelli, C. (2006). Mutations in mutT genes of Mycobacterium tuberculosis isolates of Beijing genotype. Journal of medical microbiology 55, 599-603. Lee, H.M., Shin, D.M., Kim, K.K., Lee, J.S., Paik, T.H., and Jo, E.K. (2009a). Roles of reactive oxygen species in CXCL8 and CCL2 expression in response to the 30-kDa antigen of Mycobacterium tuberculosis. Journal of clinical immunology 29, 46-56. Lee, M.H., Pascopella, L., Jacobs, W.R., Jr., and Hatfull, G.F. (1991). Site-specific integration of mycobacteriophage L5: integration-proficient vectors for Mycobacterium smegmatis, Mycobacterium tuberculosis, and bacille Calmette-Guerin. Proceedings of the National Academy of Sciences of the United States of America 88, 3111-3115. Lee, W.L., Gold, B., Darby, C., Brot, N., Jiang, X., de Carvalho, L.P., Wellner, D., St John, G., Jacobs, W.R., Jr., and Nathan, C. (2009b). Mycobacterium tuberculosis expresses methionine sulphoxide reductases A and B that protect from killing by nitrite and hypochlorite. Molecular microbiology 71, 583-593. Leemans, J.C., Juffermans, N.P., Florquin, S., van Rooijen, N., Vervoordeldonk, M.J., Verbon, A., van Deventer, S.J., and van der Poll, T. (2001). Depletion of alveolar macrophages exerts protective effects in pulmonary tuberculosis in mice. Journal of immunology 166, 4604-4611. Leemans, J.C., Thepen, T., Weijer, S., Florquin, S., van Rooijen, N., van de Winkel, J.G., and van der Poll, T. (2005). Macrophages play a dual role during pulmonary tuberculosis in mice. The Journal of infectious diseases 191, 65-74. Levy-Frebault, V.V., and Portaels, F. (1992). Proposed minimal standards for the genus Mycobacterium and for description of new slowly growing Mycobacterium species. International journal of systematic bacteriology 42, 315-323. Li, Q., Whalen, C.C., Albert, J.M., Larkin, R., Zukowski, L., Cave, M.D., and Silver, R.F. (2002). Differences in rate and variability of intracellular growth of a panel of Mycobacterium tuberculosis clinical isolates within a human monocyte model. Infection and immunity 70, 6489-6493. Li, Y., and He, Z.G. (2012). The mycobacterial LysR-type regulator OxyS responds to oxidative stress and negatively regulates expression of the catalase-peroxidase gene. PloS one 7, e30186. Little, J.W. (1982). Control of the SOS regulatory system by the level of RecA protease. Biochimie 64, 585-589. Lopez, B., Aguilar, D., Orozco, H., Burger, M., Espitia, C., Ritacco, V., Barrera, L., Kremer, K., Hernandez-Pando, R., Huygen, K., et al. (2003). A marked difference in pathogenesis and immune response induced by different Mycobacterium tuberculosis genotypes. Clinical and experimental immunology 133, 30-37. MacGurn, J.A., and Cox, J.S. (2007). A genetic screen for Mycobacterium tuberculosis mutants defective for phagosome maturation arrest identifies components of the ESX-1 secretion system. Infection and immunity 75, 2668-2678. MacMicking, J.D., North, R.J., LaCourse, R., Mudgett, J.S., Shah, S.K., and Nathan, C.F. (1997). Identification of nitric oxide synthase as a protective locus against tuberculosis. Proceedings of the National Academy of Sciences of the United States of America 94, 5243-5248. Maddocks, S.E., and Oyston, P.C. (2008). Structure and function of the LysR-type transcriptional regulator (LTTR) family proteins. Microbiology 154, 3609-3623.

Page 142: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

122 References

Maglione, P.J., Xu, J., Casadevall, A., and Chan, J. (2008). Fc gamma receptors regulate immune activation and susceptibility during Mycobacterium tuberculosis infection. Journal of immunology 180, 3329-3338. Malaga, W., Constant, P., Euphrasie, D., Cataldi, A., Daffe, M., Reyrat, J.M., and Guilhot, C. (2008). Deciphering the genetic bases of the structural diversity of phenolic glycolipids in strains of the Mycobacterium tuberculosis complex. The Journal of biological chemistry 283, 15177-15184. Malik, Z.A., Thompson, C.R., Hashimi, S., Porter, B., Iyer, S.S., and Kusner, D.J. (2003). Cutting edge: Mycobacterium tuberculosis blocks Ca2+ signaling and phagosome maturation in human macrophages via specific inhibition of sphingosine kinase. Journal of immunology 170, 2811-2815. Malm, S., Tiffert, Y., Micklinghoff, J., Schultze, S., Joost, I., Weber, I., Horst, S., Ackermann, B., Schmidt, M., Wohlleben, W., et al. (2009). The roles of the nitrate reductase NarGHJI, the nitrite reductase NirBD and the response regulator GlnR in nitrate assimilation of Mycobacterium tuberculosis. Microbiology 155, 1332-1339. Manca, C., Paul, S., Barry, C.E., 3rd, Freedman, V.H., and Kaplan, G. (1999). Mycobacterium tuberculosis catalase and peroxidase activities and resistance to oxidative killing in human monocytes in vitro. Infection and immunity 67, 74-79. Manganelli, R., Voskuil, M.I., Schoolnik, G.K., Dubnau, E., Gomez, M., and Smith, I. (2002). Role of the extracytoplasmic-function sigma factor sigma(H) in Mycobacterium tuberculosis global gene expression. Molecular microbiology 45, 365-374. Mange, P.F. (1992). Hansen and his discovery of Mycobacterium leprae. New Jersey medicine : the journal of the Medical Society of New Jersey 89, 118-121. Master, S.S., Springer, B., Sander, P., Boettger, E.C., Deretic, V., and Timmins, G.S. (2002). Oxidative stress response genes in Mycobacterium tuberculosis: role of ahpC in resistance to peroxynitrite and stage-specific survival in macrophages. Microbiology 148, 3139-3144. McAdam, R.A., Quan, S., Smith, D.A., Bardarov, S., Betts, J.C., Cook, F.C., Hooker, E.U., Lewis, A.P., Woollard, P., Everett, M.J., et al. (2002). Characterization of a Mycobacterium tuberculosis H37Rv transposon library reveals insertions in 351 ORFs and mutants with altered virulence. Microbiology 148, 2975-2986. McGuire, A.M., Weiner, B., Park, S.T., Wapinski, I., Raman, S., Dolganov, G., Peterson, M., Riley, R., Zucker, J., Abeel, T., et al. (2012). Comparative analysis of Mycobacterium and related Actinomycetes yields insight into the evolution of Mycobacterium tuberculosis pathogenesis. BMC genomics 13, 120. McKinney, J.D., Honer zu Bentrup, K., Munoz-Elias, E.J., Miczak, A., Chen, B., Chan, W.T., Swenson, D., Sacchettini, J.C., Jacobs, W.R., Jr., and Russell, D.G. (2000). Persistence of Mycobacterium tuberculosis in macrophages and mice requires the glyoxylate shunt enzyme isocitrate lyase. Nature 406, 735-738. Mehra, S., and Kaushal, D. (2009). Functional genomics reveals extended roles of the Mycobacterium tuberculosis stress response factor sigmaH. Journal of bacteriology 191, 3965-3980. Michaels, M.L., and Miller, J.H. (1992). The GO system protects organisms from the mutagenic effect of the spontaneous lesion 8-hydroxyguanine (7,8-dihydro-8-oxoguanine). Journal of bacteriology 174, 6321-6325. Miller, J.L., Velmurugan, K., Cowan, M.J., and Briken, V. (2010). The type I NADH dehydrogenase of Mycobacterium tuberculosis counters phagosomal NOX2 activity to inhibit TNF-alpha-mediated host cell apoptosis. PLoS pathogens 6, e1000864. Minakami, R., and Sumimotoa, H. (2006). Phagocytosis-coupled activation of the superoxide-producing phagocyte oxidase, a member of the NADPH oxidase (nox) family. International journal of hematology 84, 193-198. Mizrahi, V., and Andersen, S.J. (1998). DNA repair in Mycobacterium tuberculosis. What have we learnt from the genome sequence? Molecular microbiology 29, 1331-1339. Mokrousov, I., Jiao, W.W., Sun, G.Z., Liu, J.W., Valcheva, V., Li, M., Narvskaya, O., and Shen, A.D. (2006). Evolution of drug resistance in different sublineages of Mycobacterium tuberculosis Beijing genotype. Antimicrobial agents and chemotherapy 50, 2820-2823.

Page 143: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

123 References

Moreland, N.J., Charlier, C., Dingley, A.J., Baker, E.N., and Lott, J.S. (2009). Making sense of a missense mutation: characterization of MutT2, a Nudix hydrolase from Mycobacterium tuberculosis, and the G58R mutant encoded in W-Beijing strains of M. tuberculosis. Biochemistry 48, 699-708. Morimatsu, K., and Kowalczykowski, S.C. (2003). RecFOR proteins load RecA protein onto gapped DNA to accelerate DNA strand exchange: a universal step of recombinational repair. Mol Cell 11, 1337-1347. Morlock, G.P., Plikaytis, B.B., and Crawford, J.T. (2000). Characterization of spontaneous, In vitro-selected, rifampin-resistant mutants of Mycobacterium tuberculosis strain H37Rv. Antimicrobial agents and chemotherapy 44, 3298-3301. Mostowy, S., Inwald, J., Gordon, S., Martin, C., Warren, R., Kremer, K., Cousins, D., and Behr, M.A. (2005). Revisiting the evolution of Mycobacterium bovis. Journal of bacteriology 187, 6386-6395. Mukherjee, P., Sureka, K., Datta, P., Hossain, T., Barik, S., Das, K.P., Kundu, M., and Basu, J. (2009). Novel role of Wag31 in protection of mycobacteria under oxidative stress. Molecular microbiology 73, 103-119. Namouchi, A., Didelot, X., Schock, U., Gicquel, B., and Rocha, E.P. (2012). After the bottleneck: Genome-wide diversification of the Mycobacterium tuberculosis complex by mutation, recombination, and natural selection. Genome research 22, 721-734. Ng, V.H., Cox, J.S., Sousa, A.O., MacMicking, J.D., and McKinney, J.D. (2004). Role of KatG catalase-peroxidase in mycobacterial pathogenesis: countering the phagocyte oxidative burst. Molecular microbiology 52, 1291-1302. Nguyen, L., and Thompson, C.J. (2006). Foundations of antibiotic resistance in bacterial physiology: the mycobacterial paradigm. Trends in microbiology 14, 304-312. Nicholson, S., Bonecini-Almeida Mda, G., Lapa e Silva, J.R., Nathan, C., Xie, Q.W., Mumford, R., Weidner, J.R., Calaycay, J., Geng, J., Boechat, N., et al. (1996). Inducible nitric oxide synthase in pulmonary alveolar macrophages from patients with tuberculosis. The Journal of experimental medicine 183, 2293-2302. Nickel, D., Busch, M., Mayer, D., Hagemann, B., Knoll, V., and Stenger, S. (2012). Hypoxia triggers the expression of human beta defensin 2 and antimicrobial activity against Mycobacterium tuberculosis in human macrophages. Journal of immunology 188, 4001-4007. Nicol, M.P., and Wilkinson, R.J. (2008). The clinical consequences of strain diversity in Mycobacterium tuberculosis. Transactions of the Royal Society of Tropical Medicine and Hygiene 102, 955-965. Niederweis, M., Danilchanka, O., Huff, J., Hoffmann, C., and Engelhardt, H. (2010). Mycobacterial outer membranes: in search of proteins. Trends in microbiology 18, 109-116. Nigou, J., Zelle-Rieser, C., Gilleron, M., Thurnher, M., and Puzo, G. (2001). Mannosylated lipoarabinomannans inhibit IL-12 production by human dendritic cells: evidence for a negative signal delivered through the mannose receptor. Journal of immunology 166, 7477-7485. Nikaido, H. (2001). Preventing drug access to targets: cell surface permeability barriers and active efflux in bacteria. Seminars in cell & developmental biology 12, 215-223. Nozaki, Y., Hasegawa, Y., Ichiyama, S., Nakashima, I., and Shimokata, K. (1997). Mechanism of nitric oxide-dependent killing of Mycobacterium bovis BCG in human alveolar macrophages. Infection and immunity 65, 3644-3647. O'Brien, L., Carmichael, J., Lowrie, D.B., and Andrew, P.W. (1994). Strains of Mycobacterium tuberculosis differ in susceptibility to reactive nitrogen intermediates in vitro. Infection and immunity 62, 5187-5190. O'Brien, P.J., and Ellenberger, T. (2004). The Escherichia coli 3-methyladenine DNA glycosylase AlkA has a remarkably versatile active site. The Journal of biological chemistry 279, 26876-26884. O'Rourke, E.J., Chevalier, C., Pinto, A.V., Thiberge, J.M., Ielpi, L., Labigne, A., and Radicella, J.P. (2003). Pathogen DNA as target for host-generated oxidative stress: role for repair of bacterial DNA damage in Helicobacter pylori colonization. Proceedings of the National Academy of Sciences of the United States of America 100, 2789-2794. O'Sullivan, D.M., McHugh, T.D., and Gillespie, S.H. (2005). Analysis of rpoB and pncA mutations in the published literature: an insight into the role of oxidative stress in Mycobacterium tuberculosis evolution? The Journal of antimicrobial chemotherapy 55, 674-679.

Page 144: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

124 References

O'Toole, R. (2010). Experimental models used to study human tuberculosis. Advances in applied microbiology 71, 75-89. Ortalo-Magne, A., Dupont, M.A., Lemassu, A., Andersen, A.B., Gounon, P., and Daffe, M. (1995). Molecular composition of the outermost capsular material of the tubercle bacillus. Microbiology 141 ( Pt 7), 1609-1620. Pagan-Ramos, E., Song, J., McFalone, M., Mudd, M.H., and Deretic, V. (1998). Oxidative stress response and characterization of the oxyR-ahpC and furA-katG loci in Mycobacterium marinum. Journal of bacteriology 180, 4856-4864. Palanisamy, G.S., DuTeau, N., Eisenach, K.D., Cave, D.M., Theus, S.A., Kreiswirth, B.N., Basaraba, R.J., and Orme, I.M. (2009). Clinical strains of Mycobacterium tuberculosis display a wide range of virulence in guinea pigs. Tuberculosis 89, 203-209. Palomino, J.C. (2012). Current developments and future perspectives for TB diagnostics. Future microbiology 7, 59-71. Parish, T. (2003). Starvation survival response of Mycobacterium tuberculosis. Journal of bacteriology 185, 6702-6706. Parish, T., and Stoker, N.G. (1998). Electroporation of mycobacteria. Methods Mol Biol 101, 129-144. Parwati, I., van Crevel, R., and van Soolingen, D. (2010). Possible underlying mechanisms for successful emergence of the Mycobacterium tuberculosis Beijing genotype strains. The Lancet infectious diseases 10, 103-111. Pathak, S.K., Basu, S., Basu, K.K., Banerjee, A., Pathak, S., Bhattacharyya, A., Kaisho, T., Kundu, M., and Basu, J. (2007). Direct extracellular interaction between the early secreted antigen ESAT-6 of Mycobacterium tuberculosis and TLR2 inhibits TLR signaling in macrophages. Nature immunology 8, 610-618. Perdigao, J., Macedo, R., Joao, I., Fernandes, E., Brum, L., and Portugal, I. (2008). Multidrug-resistant tuberculosis in Lisbon, Portugal: a molecular epidemiological perspective. Microb Drug Resist 14, 133-143. Perdigao, J., Macedo, R., Silva, C., Pinto, C., Furtado, C., Brum, L., and Portugal, I. (2011). Tuberculosis drug-resistance in Lisbon, Portugal: a 6-year overview. Clinical microbiology and infection : the official publication of the European Society of Clinical Microbiology and Infectious Diseases 17, 1397-1402. Perez, E., Samper, S., Bordas, Y., Guilhot, C., Gicquel, B., and Martin, C. (2001). An essential role for phoP in Mycobacterium tuberculosis virulence. Molecular microbiology 41, 179-187. Pethe, K., Swenson, D.L., Alonso, S., Anderson, J., Wang, C., and Russell, D.G. (2004). Isolation of Mycobacterium tuberculosis mutants defective in the arrest of phagosome maturation. Proceedings of the National Academy of Sciences of the United States of America 101, 13642-13647. Philips, J.A. (2008). Mycobacterial manipulation of vacuolar sorting. Cellular microbiology 10, 2408-2415. Piddington, D.L., Fang, F.C., Laessig, T., Cooper, A.M., Orme, I.M., and Buchmeier, N.A. (2001). Cu,Zn superoxide dismutase of Mycobacterium tuberculosis contributes to survival in activated macrophages that are generating an oxidative burst. Infection and immunity 69, 4980-4987. Pitarque, S., Larrouy-Maumus, G., Payre, B., Jackson, M., Puzo, G., and Nigou, J. (2008). The immunomodulatory lipoglycans, lipoarabinomannan and lipomannan, are exposed at the mycobacterial cell surface. Tuberculosis 88, 560-565. Portugal, I., Covas, M.J., Brum, L., Viveiros, M., Ferrinho, P., Moniz-Pereira, J., and David, H. (1999). Outbreak of multiple drug-resistant tuberculosis in Lisbon: detection by restriction fragment length polymorphism analysis. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease 3, 207-213. Rachman, H., Strong, M., Ulrichs, T., Grode, L., Schuchhardt, J., Mollenkopf, H., Kosmiadi, G.A., Eisenberg, D., and Kaufmann, S.H. (2006). Unique transcriptome signature of Mycobacterium tuberculosis in pulmonary tuberculosis. Infection and immunity 74, 1233-1242. Ramachandra, L., Noss, E., Boom, W.H., and Harding, C.V. (2001). Processing of Mycobacterium tuberculosis antigen 85B involves intraphagosomal formation of peptide-major histocompatibility

Page 145: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

125 References

complex II complexes and is inhibited by live bacilli that decrease phagosome maturation. The Journal of experimental medicine 194, 1421-1432. Raman, S., Song, T., Puyang, X., Bardarov, S., Jacobs, W.R., Jr., and Husson, R.N. (2001). The alternative sigma factor SigH regulates major components of oxidative and heat stress responses in Mycobacterium tuberculosis. Journal of bacteriology 183, 6119-6125. Ramon-Garcia, S., Martin, C., Thompson, C.J., and Ainsa, J.A. (2009). Role of the Mycobacterium tuberculosis P55 efflux pump in intrinsic drug resistance, oxidative stress responses, and growth. Antimicrobial agents and chemotherapy 53, 3675-3682. Rand, L., Hinds, J., Springer, B., Sander, P., Buxton, R.S., and Davis, E.O. (2003). The majority of inducible DNA repair genes in Mycobacterium tuberculosis are induced independently of RecA. Molecular microbiology 50, 1031-1042. Rauch, P.J., Palmen, R., Burds, A.A., Gregg-Jolly, L.A., van der Zee, J.R., and Hellingwerf, K.J. (1996). The expression of the Acinetobacter calcoaceticus recA gene increases in response to DNA damage independently of RecA and of development of competence for natural transformation. Microbiology 142 ( Pt 4), 1025-1032. Reed, M.B., Domenech, P., Manca, C., Su, H., Barczak, A.K., Kreiswirth, B.N., Kaplan, G., and Barry, C.E., 3rd (2004). A glycolipid of hypervirulent tuberculosis strains that inhibits the innate immune response. Nature 431, 84-87. Reed, M.B., Gagneux, S., Deriemer, K., Small, P.M., and Barry, C.E., 3rd (2007). The W-Beijing lineage of Mycobacterium tuberculosis overproduces triglycerides and has the DosR dormancy regulon constitutively upregulated. Journal of bacteriology 189, 2583-2589. Reiling, N., Holscher, C., Fehrenbach, A., Kroger, S., Kirschning, C.J., Goyert, S., and Ehlers, S. (2002). Cutting edge: Toll-like receptor (TLR)2- and TLR4-mediated pathogen recognition in resistance to airborne infection with Mycobacterium tuberculosis. Journal of immunology 169, 3480-3484. Rengarajan, J., Bloom, B.R., and Rubin, E.J. (2005). Genome-wide requirements for Mycobacterium tuberculosis adaptation and survival in macrophages. Proceedings of the National Academy of Sciences of the United States of America 102, 8327-8332. Rhee, K.Y., Erdjument-Bromage, H., Tempst, P., and Nathan, C.F. (2005). S-nitroso proteome of Mycobacterium tuberculosis: Enzymes of intermediary metabolism and antioxidant defense. Proceedings of the National Academy of Sciences of the United States of America 102, 467-472. Rindi, L., Lari, N., Cuccu, B., and Garzelli, C. (2009). Evolutionary pathway of the Beijing lineage of Mycobacterium tuberculosis based on genomic deletions and mutT genes polymorphisms. Infection, genetics and evolution : journal of molecular epidemiology and evolutionary genetics in infectious diseases 9, 48-53. Rodriguez, G.M., Voskuil, M.I., Gold, B., Schoolnik, G.K., and Smith, I. (2002). ideR, An essential gene in mycobacterium tuberculosis: role of IdeR in iron-dependent gene expression, iron metabolism, and oxidative stress response. Infection and immunity 70, 3371-3381. Rohde, K.H., Abramovitch, R.B., and Russell, D.G. (2007). Mycobacterium tuberculosis invasion of macrophages: linking bacterial gene expression to environmental cues. Cell host & microbe 2, 352-364. Rosas-Magallanes, V., Stadthagen-Gomez, G., Rauzier, J., Barreiro, L.B., Tailleux, L., Boudou, F., Griffin, R., Nigou, J., Jackson, M., Gicquel, B., et al. (2007). Signature-tagged transposon mutagenesis identifies novel Mycobacterium tuberculosis genes involved in the parasitism of human macrophages. Infection and immunity 75, 504-507. Rossi, F., Khanduja, J.S., Bortoluzzi, A., Houghton, J., Sander, P., Guthlein, C., Davis, E.O., Springer, B., Bottger, E.C., Relini, A., et al. (2011). The biological and structural characterization of Mycobacterium tuberculosis UvrA provides novel insights into its mechanism of action. Nucleic Acids Res 39, 7316-7328. Russell, D.G. (2007). Who puts the tubercle in tuberculosis? Nature reviews Microbiology 5, 39-47. Ryffel, B., Fremond, C., Jacobs, M., Parida, S., Botha, T., Schnyder, B., and Quesniaux, V. (2005). Innate immunity to mycobacterial infection in mice: critical role for toll-like receptors. Tuberculosis 85, 395-405.

Page 146: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

126 References

Sala, C., Forti, F., Di Florio, E., Canneva, F., Milano, A., Riccardi, G., and Ghisotti, D. (2003). Mycobacterium tuberculosis FurA autoregulates its own expression. Journal of bacteriology 185, 5357-5362. Sani, M., Houben, E.N., Geurtsen, J., Pierson, J., de Punder, K., van Zon, M., Wever, B., Piersma, S.R., Jimenez, C.R., Daffe, M., et al. (2010). Direct visualization by cryo-EM of the mycobacterial capsular layer: a labile structure containing ESX-1-secreted proteins. PLoS pathogens 6, e1000794. Sarkar, S., and Suresh, M.R. (2011). An overview of tuberculosis chemotherapy - a literature review. Journal of pharmacy & pharmaceutical sciences : a publication of the Canadian Society for Pharmaceutical Sciences, Societe canadienne des sciences pharmaceutiques 14, 148-161. Sassetti, C.M., and Rubin, E.J. (2003). Genetic requirements for mycobacterial survival during infection. Proceedings of the National Academy of Sciences of the United States of America 100, 12989-12994. Schlesinger, L.S. (1993). Macrophage phagocytosis of virulent but not attenuated strains of Mycobacterium tuberculosis is mediated by mannose receptors in addition to complement receptors. Journal of immunology 150, 2920-2930. Schnappinger, D., Ehrt, S., Voskuil, M.I., Liu, Y., Mangan, J.A., Monahan, I.M., Dolganov, G., Efron, B., Butcher, P.D., Nathan, C., et al. (2003). Transcriptional Adaptation of Mycobacterium tuberculosis within Macrophages: Insights into the Phagosomal Environment. The Journal of experimental medicine 198, 693-704. Schurch, A.C., Kremer, K., Warren, R.M., Hung, N.V., Zhao, Y., Wan, K., Boeree, M.J., Siezen, R.J., Smith, N.H., and van Soolingen, D. (2011). Mutations in the regulatory network underlie the recent clonal expansion of a dominant subclone of the Mycobacterium tuberculosis Beijing genotype. Infection, genetics and evolution : journal of molecular epidemiology and evolutionary genetics in infectious diseases 11, 587-597. Seeberg, E., Eide, L., and Bjoras, M. (1995). The base excision repair pathway. Trends Biochem Sci 20, 391-397. Senaratne, R.H., De Silva, A.D., Williams, S.J., Mougous, J.D., Reader, J.R., Zhang, T., Chan, S., Sidders, B., Lee, D.H., Chan, J., et al. (2006). 5'-Adenosinephosphosulphate reductase (CysH) protects Mycobacterium tuberculosis against free radicals during chronic infection phase in mice. Molecular microbiology 59, 1744-1753. Shibutani, S., Takeshita, M., and Grollman, A.P. (1991). Insertion of specific bases during DNA synthesis past the oxidation-damaged base 8-oxodG. Nature 349, 431-434. Shin, D.M., Yang, C.S., Lee, J.Y., Lee, S.J., Choi, H.H., Lee, H.M., Yuk, J.M., Harding, C.V., and Jo, E.K. (2008). Mycobacterium tuberculosis lipoprotein-induced association of TLR2 with protein kinase C zeta in lipid rafts contributes to reactive oxygen species-dependent inflammatory signalling in macrophages. Cellular microbiology 10, 1893-1905. Shin, D.M., Yuk, J.M., Lee, H.M., Lee, S.H., Son, J.W., Harding, C.V., Kim, J.M., Modlin, R.L., and Jo, E.K. (2010). Mycobacterial lipoprotein activates autophagy via TLR2/1/CD14 and a functional vitamin D receptor signalling. Cellular microbiology 12, 1648-1665. Shinnick, T.M., and Good, R.C. (1994). Mycobacterial taxonomy. European journal of clinical microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology 13, 884-901. Shuman, S., and Glickman, M.S. (2007). Bacterial DNA repair by non-homologous end joining. Nature reviews Microbiology 5, 852-861. Singleton, M.R., Dillingham, M.S., Gaudier, M., Kowalczykowski, S.C., and Wigley, D.B. (2004). Crystal structure of RecBCD enzyme reveals a machine for processing DNA breaks. Nature 432, 187-193. Sinha, K.M., Stephanou, N.C., Gao, F., Glickman, M.S., and Shuman, S. (2007). Mycobacterial UvrD1 is a Ku-dependent DNA helicase that plays a role in multiple DNA repair events, including double-strand break repair. The Journal of biological chemistry 282, 15114-15125. Slupphaug, G., Kavli, B., and Krokan, H.E. (2003). The interacting pathways for prevention and repair of oxidative DNA damage. Mutation research 531, 231-251.

Page 147: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

127 References

Sly, L.M., Lopez, M., Nauseef, W.M., and Reiner, N.E. (2001). 1alpha,25-Dihydroxyvitamin D3-induced monocyte antimycobacterial activity is regulated by phosphatidylinositol 3-kinase and mediated by the NADPH-dependent phagocyte oxidase. The Journal of biological chemistry 276, 35482-35493. Smith, N.H., Kremer, K., Inwald, J., Dale, J., Driscoll, J.R., Gordon, S.V., van Soolingen, D., Hewinson, R.G., and Smith, J.M. (2006). Ecotypes of the Mycobacterium tuberculosis complex. Journal of theoretical biology 239, 220-225. Sohaskey, C.D., and Wayne, L.G. (2003). Role of narK2X and narGHJI in hypoxic upregulation of nitrate reduction by Mycobacterium tuberculosis. Journal of bacteriology 185, 7247-7256. Springer, B., Master, S., Sander, P., Zahrt, T., McFalone, M., Song, J., Papavinasasundaram, K.G., Colston, M.J., Boettger, E., and Deretic, V. (2001). Silencing of oxidative stress response in Mycobacterium tuberculosis: expression patterns of ahpC in virulent and avirulent strains and effect of ahpC inactivation. Infection and immunity 69, 5967-5973. Sreevatsan, S., Pan, X., Stockbauer, K.E., Connell, N.D., Kreiswirth, B.N., Whittam, T.S., and Musser, J.M. (1997). Restricted structural gene polymorphism in the Mycobacterium tuberculosis complex indicates evolutionarily recent global dissemination. Proceedings of the National Academy of Sciences of the United States of America 94, 9869-9874. Stahl, D.A., and Urbance, J.W. (1990). The division between fast- and slow-growing species corresponds to natural relationships among the mycobacteria. Journal of bacteriology 172, 116-124. Stallings, C.L., Stephanou, N.C., Chu, L., Hochschild, A., Nickels, B.E., and Glickman, M.S. (2009). CarD is an essential regulator of rRNA transcription required for Mycobacterium tuberculosis persistence. Cell 138, 146-159. Stephenson, J.D., and Shepherd, V.L. (1987). Purification of the human alveolar macrophage mannose receptor. Biochemical and biophysical research communications 148, 883-889. Storz, G., and Imlay, J.A. (1999). Oxidative stress. Current opinion in microbiology 2, 188-194. Sturgill-Koszycki, S., Schlesinger, P.H., Chakraborty, P., Haddix, P.L., Collins, H.L., Fok, A.K., Allen, R.D., Gluck, S.L., Heuser, J., and Russell, D.G. (1994). Lack of acidification in Mycobacterium phagosomes produced by exclusion of the vesicular proton-ATPase. Science 263, 678-681. Sugawara, I., Yamada, H., Li, C., Mizuno, S., Takeuchi, O., and Akira, S. (2003). Mycobacterial infection in TLR2 and TLR6 knockout mice. Microbiology and immunology 47, 327-336. Sun, J., Wang, X., Lau, A., Liao, T.Y., Bucci, C., and Hmama, Z. (2010). Mycobacterial nucleoside diphosphate kinase blocks phagosome maturation in murine RAW 264.7 macrophages. PloS one 5, e8769. Supply, P., Mazars, E., Lesjean, S., Vincent, V., Gicquel, B., and Locht, C. (2000). Variable human minisatellite-like regions in the Mycobacterium tuberculosis genome. Molecular microbiology 36, 762-771. Supply, P., Warren, R.M., Banuls, A.L., Lesjean, S., Van Der Spuy, G.D., Lewis, L.A., Tibayrenc, M., Van Helden, P.D., and Locht, C. (2003). Linkage disequilibrium between minisatellite loci supports clonal evolution of Mycobacterium tuberculosis in a high tuberculosis incidence area. Molecular microbiology 47, 529-538. Swaminathan, S., Padmapriyadarsini, C., and Narendran, G. (2010). HIV-associated tuberculosis: clinical update. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 50, 1377-1386. Taddei, F., Radman, M., Maynard-Smith, J., Toupance, B., Gouyon, P.H., and Godelle, B. (1997). Role of mutator alleles in adaptive evolution. Nature 387, 700-702. Tailleux, L., Pham-Thi, N., Bergeron-Lafaurie, A., Herrmann, J.L., Charles, P., Schwartz, O., Scheinmann, P., Lagrange, P.H., de Blic, J., Tazi, A., et al. (2005). DC-SIGN induction in alveolar macrophages defines privileged target host cells for mycobacteria in patients with tuberculosis. PLoS medicine 2, e381. Tailleux, L., Schwartz, O., Herrmann, J.L., Pivert, E., Jackson, M., Amara, A., Legres, L., Dreher, D., Nicod, L.P., Gluckman, J.C., et al. (2003). DC-SIGN is the major Mycobacterium tuberculosis receptor on human dendritic cells. The Journal of experimental medicine 197, 121-127. Tailleux, L., Waddell, S.J., Pelizzola, M., Mortellaro, A., Withers, M., Tanne, A., Castagnoli, P.R., Gicquel, B., Stoker, N.G., Butcher, P.D., et al. (2008). Probing host pathogen cross-talk by transcriptional profiling

Page 148: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

128 References

of both Mycobacterium tuberculosis and infected human dendritic cells and macrophages. PloS one 3, e1403. Tan, M.P., Sequeira, P., Lin, W.W., Phong, W.Y., Cliff, P., Ng, S.H., Lee, B.H., Camacho, L., Schnappinger, D., Ehrt, S., et al. (2010). Nitrate respiration protects hypoxic Mycobacterium tuberculosis against acid- and reactive nitrogen species stresses. PloS one 5, e13356. Tanne, A., Ma, B., Boudou, F., Tailleux, L., Botella, H., Badell, E., Levillain, F., Taylor, M.E., Drickamer, K., Nigou, J., et al. (2009). A murine DC-SIGN homologue contributes to early host defense against Mycobacterium tuberculosis. The Journal of experimental medicine 206, 2205-2220. Taverna, P., and Sedgwick, B. (1996). Generation of an endogenous DNA-methylating agent by nitrosation in Escherichia coli. Journal of bacteriology 178, 5105-5111. Tekaia, F., Gordon, S.V., Garnier, T., Brosch, R., Barrell, B.G., and Cole, S.T. (1999). Analysis of the proteome of Mycobacterium tuberculosis in silico. Tubercle and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease 79, 329-342. Tenaillon, O., Toupance, B., Le Nagard, H., Taddei, F., and Godelle, B. (1999). Mutators, population size, adaptive landscape and the adaptation of asexual populations of bacteria. Genetics 152, 485-493. Theus, S., Eisenach, K., Fomukong, N., Silver, R.F., and Cave, M.D. (2007). Beijing family Mycobacterium tuberculosis strains differ in their intracellular growth in THP-1 macrophages. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease 11, 1087-1093. Theus, S.A., Cave, M.D., and Eisenach, K.D. (2004). Activated THP-1 cells: an attractive model for the assessment of intracellular growth rates of Mycobacterium tuberculosis isolates. Infection and immunity 72, 1169-1173. Thoma-Uszynski, S., Stenger, S., Takeuchi, O., Ochoa, M.T., Engele, M., Sieling, P.A., Barnes, P.F., Rollinghoff, M., Bolcskei, P.L., Wagner, M., et al. (2001). Induction of direct antimicrobial activity through mammalian toll-like receptors. Science 291, 1544-1547. Thwaites, G., Caws, M., Chau, T.T., D'Sa, A., Lan, N.T., Huyen, M.N., Gagneux, S., Anh, P.T., Tho, D.Q., Torok, E., et al. (2008). Relationship between Mycobacterium tuberculosis genotype and the clinical phenotype of pulmonary and meningeal tuberculosis. Journal of clinical microbiology 46, 1363-1368. Tonjum, T., and Seeberg, E. (2001). Microbial fitness and genome dynamics. Trends in microbiology 9, 356-358. Torrelles, J.B., Azad, A.K., and Schlesinger, L.S. (2006). Fine discrimination in the recognition of individual species of phosphatidyl-myo-inositol mannosides from Mycobacterium tuberculosis by C-type lectin pattern recognition receptors. Journal of immunology 177, 1805-1816. Toungoussova, O.S., Caugant, D.A., Sandven, P., Mariandyshev, A.O., and Bjune, G. (2004). Impact of drug resistance on fitness of Mycobacterium tuberculosis strains of the W-Beijing genotype. FEMS immunology and medical microbiology 42, 281-290. Trunz, B.B., Fine, P., and Dye, C. (2006). Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 367, 1173-1180. Tsenova, L., Ellison, E., Harbacheuski, R., Moreira, A.L., Kurepina, N., Reed, M.B., Mathema, B., Barry, C.E., 3rd, and Kaplan, G. (2005). Virulence of selected Mycobacterium tuberculosis clinical isolates in the rabbit model of meningitis is dependent on phenolic glycolipid produced by the bacilli. The Journal of infectious diseases 192, 98-106. Tsolaki, A.G., Gagneux, S., Pym, A.S., Goguet de la Salmoniere, Y.O., Kreiswirth, B.N., Van Soolingen, D., and Small, P.M. (2005). Genomic deletions classify the Beijing/W strains as a distinct genetic lineage of Mycobacterium tuberculosis. Journal of clinical microbiology 43, 3185-3191. Tuberculosis, E.C.A.o.N.G.G.M.a.T.f.t.E.a.C.o. (2006). Beijing/W genotype Mycobacterium tuberculosis and drug resistance. Emerging infectious diseases 12, 736-743. Tullius, M.V., Harmston, C.A., Owens, C.P., Chim, N., Morse, R.P., McMath, L.M., Iniguez, A., Kimmey, J.M., Sawaya, M.R., Whitelegge, J.P., et al. (2011). Discovery and characterization of a unique mycobacterial heme acquisition system. Proceedings of the National Academy of Sciences of the United States of America 108, 5051-5056.

Page 149: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

129 References

Ulrichs, T., and Kaufmann, S.H. (2006). New insights into the function of granulomas in human tuberculosis. The Journal of pathology 208, 261-269. Underhill, D.M. (2007). Collaboration between the innate immune receptors dectin-1, TLRs, and Nods. Immunological reviews 219, 75-87. Valway, S.E., Sanchez, M.P., Shinnick, T.F., Orme, I., Agerton, T., Hoy, D., Jones, J.S., Westmoreland, H., and Onorato, I.M. (1998). An outbreak involving extensive transmission of a virulent strain of Mycobacterium tuberculosis. The New England journal of medicine 338, 633-639. van Embden, J.D., Cave, M.D., Crawford, J.T., Dale, J.W., Eisenach, K.D., Gicquel, B., Hermans, P., Martin, C., McAdam, R., Shinnick, T.M., et al. (1993). Strain identification of Mycobacterium tuberculosis by DNA fingerprinting: recommendations for a standardized methodology. Journal of clinical microbiology 31, 406-409. Van Soolingen, D. (2001). Molecular epidemiology of tuberculosis and other mycobacterial infections: main methodologies and achievements. Journal of internal medicine 249, 1-26. van Soolingen, D., Hoogenboezem, T., de Haas, P.E., Hermans, P.W., Koedam, M.A., Teppema, K.S., Brennan, P.J., Besra, G.S., Portaels, F., Top, J., et al. (1997). A novel pathogenic taxon of the Mycobacterium tuberculosis complex, Canetti: characterization of an exceptional isolate from Africa. International journal of systematic bacteriology 47, 1236-1245. van Soolingen, D., Qian, L., de Haas, P.E., Douglas, J.T., Traore, H., Portaels, F., Qing, H.Z., Enkhsaikan, D., Nymadawa, P., and van Embden, J.D. (1995). Predominance of a single genotype of Mycobacterium tuberculosis in countries of east Asia. Journal of clinical microbiology 33, 3234-3238. Vandal, O.H., Pierini, L.M., Schnappinger, D., Nathan, C.F., and Ehrt, S. (2008). A membrane protein preserves intrabacterial pH in intraphagosomal Mycobacterium tuberculosis. Nature medicine 14, 849-854. Vandal, O.H., Roberts, J.A., Odaira, T., Schnappinger, D., Nathan, C.F., and Ehrt, S. (2009). Acid-susceptible mutants of Mycobacterium tuberculosis share hypersusceptibility to cell wall and oxidative stress and to the host environment. Journal of bacteriology 191, 625-631. Vannberg, F.O., Chapman, S.J., Khor, C.C., Tosh, K., Floyd, S., Jackson-Sillah, D., Crampin, A., Sichali, L., Bah, B., Gustafson, P., et al. (2008). CD209 genetic polymorphism and tuberculosis disease. PloS one 3, e1388. Velayati, A.A., Masjedi, M.R., Farnia, P., Tabarsi, P., Ghanavi, J., Ziazarifi, A.H., and Hoffner, S.E. (2009). Emergence of new forms of totally drug-resistant tuberculosis bacilli: super extensively drug-resistant tuberculosis or totally drug-resistant strains in iran. Chest 136, 420-425. Velez, D.R., Wejse, C., Stryjewski, M.E., Abbate, E., Hulme, W.F., Myers, J.L., Estevan, R., Patillo, S.G., Olesen, R., Tacconelli, A., et al. (2010). Variants in toll-like receptors 2 and 9 influence susceptibility to pulmonary tuberculosis in Caucasians, African-Americans, and West Africans. Human genetics 127, 65-73. Venkatesh, R., Ganesh, N., Guhan, N., Reddy, M.S., Chandrasekhar, T., and Muniyappa, K. (2002). RecX protein abrogates ATP hydrolysis and strand exchange promoted by RecA: insights into negative regulation of homologous recombination. Proceedings of the National Academy of Sciences of the United States of America 99, 12091-12096. Ventura, M., Canchaya, C., Tauch, A., Chandra, G., Fitzgerald, G.F., Chater, K.F., and van Sinderen, D. (2007). Genomics of Actinobacteria: tracing the evolutionary history of an ancient phylum. Microbiology and molecular biology reviews : MMBR 71, 495-548. Vergne, I., Chua, J., and Deretic, V. (2003). Tuberculosis toxin blocking phagosome maturation inhibits a novel Ca2+/calmodulin-PI3K hVPS34 cascade. The Journal of experimental medicine 198, 653-659. Vergne, I., Chua, J., Lee, H.H., Lucas, M., Belisle, J., and Deretic, V. (2005). Mechanism of phagolysosome biogenesis block by viable Mycobacterium tuberculosis. Proceedings of the National Academy of Sciences of the United States of America 102, 4033-4038. Vilcheze, C., Av-Gay, Y., Attarian, R., Liu, Z., Hazbon, M.H., Colangeli, R., Chen, B., Liu, W., Alland, D., Sacchettini, J.C., et al. (2008). Mycothiol biosynthesis is essential for ethionamide susceptibility in Mycobacterium tuberculosis. Molecular microbiology 69, 1316-1329.

Page 150: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

130 References

Vollmer, W., Joris, B., Charlier, P., and Foster, S. (2008). Bacterial peptidoglycan (murein) hydrolases. FEMS microbiology reviews 32, 259-286. Voskuil, M.I., Bartek, I.L., Visconti, K., and Schoolnik, G.K. (2011). The response of mycobacterium tuberculosis to reactive oxygen and nitrogen species. Frontiers in microbiology 2, 105. Voskuil, M.I., Schnappinger, D., Rutherford, R., Liu, Y., and Schoolnik, G.K. (2004). Regulation of the Mycobacterium tuberculosis PE/PPE genes. Tuberculosis 84, 256-262. Voskuil, M.I., Schnappinger, D., Visconti, K.C., Harrell, M.I., Dolganov, G.M., Sherman, D.R., and Schoolnik, G.K. (2003). Inhibition of respiration by nitric oxide induces a Mycobacterium tuberculosis dormancy program. The Journal of experimental medicine 198, 705-713. Walburger, A., Koul, A., Ferrari, G., Nguyen, L., Prescianotto-Baschong, C., Huygen, K., Klebl, B., Thompson, C., Bacher, G., and Pieters, J. (2004). Protein kinase G from pathogenic mycobacteria promotes survival within macrophages. Science 304, 1800-1804. Walker, L., and Lowrie, D.B. (1981). Killing of Mycobacterium microti by immunologically activated macrophages. Nature 293, 69-71. Walters, S.B., Dubnau, E., Kolesnikova, I., Laval, F., Daffe, M., and Smith, I. (2006). The Mycobacterium tuberculosis PhoPR two-component system regulates genes essential for virulence and complex lipid biosynthesis. Molecular microbiology 60, 312-330. Wang, C., Peyron, P., Mestre, O., Kaplan, G., van Soolingen, D., Gao, Q., Gicquel, B., and Neyrolles, O. (2010). Innate immune response to Mycobacterium tuberculosis Beijing and other genotypes. PloS one 5, e13594. Wang, C.H., Liu, C.Y., Lin, H.C., Yu, C.T., Chung, K.F., and Kuo, H.P. (1998). Increased exhaled nitric oxide in active pulmonary tuberculosis due to inducible NO synthase upregulation in alveolar macrophages. The European respiratory journal : official journal of the European Society for Clinical Respiratory Physiology 11, 809-815. Wanner, R.M., Guthlein, C., Springer, B., Bottger, E.C., and Ackermann, M. (2008). Stabilization of the genome of the mismatch repair deficient Mycobacterium tuberculosis by context-dependent codon choice. BMC genomics 9, 249. Wells, C.D., Cegielski, J.P., Nelson, L.J., Laserson, K.F., Holtz, T.H., Finlay, A., Castro, K.G., and Weyer, K. (2007). HIV infection and multidrug-resistant tuberculosis: the perfect storm. The Journal of infectious diseases 196 Suppl 1, S86-107. Werle, E., Schneider, C., Renner, M., Volker, M., and Fiehn, W. (1994). Convenient single-step, one tube purification of PCR products for direct sequencing. Nucleic Acids Res 22, 4354-4355. Werngren, J., and Hoffner, S.E. (2003). Drug-susceptible Mycobacterium tuberculosis Beijing genotype does not develop mutation-conferred resistance to rifampin at an elevated rate. Journal of clinical microbiology 41, 1520-1524. Whalen, C., Horsburgh, C.R., Hom, D., Lahart, C., Simberkoff, M., and Ellner, J. (1995). Accelerated course of human immunodeficiency virus infection after tuberculosis. American journal of respiratory and critical care medicine 151, 129-135. WHO (2011). Global tuberculosis control report Williams, M.J., Kana, B.D., and Mizrahi, V. (2011). Functional analysis of molybdopterin biosynthesis in mycobacteria identifies a fused molybdopterin synthase in Mycobacterium tuberculosis. Journal of bacteriology 193, 98-106. Witkin, E.M., and Roegner-Maniscalco, V. (1992). Overproduction of DnaE protein (alpha subunit of DNA polymerase III) restores viability in a conditionally inviable Escherichia coli strain deficient in DNA polymerase I. Journal of bacteriology 174, 4166-4168. Wu, C.H., Tsai-Wu, J.J., Huang, Y.T., Lin, C.Y., Lioua, G.G., and Lee, F.J. (1998). Identification and subcellular localization of a novel Cu,Zn superoxide dismutase of Mycobacterium tuberculosis. FEBS letters 439, 192-196. Wu, K., Dong, D., Fang, H., Levillain, F., Jin, W., Mei, J., Gicquel, B., Du, Y., Wang, K., Gao, Q., et al. (2012). An interferon-related signature in the transcriptional core response of human macrophages to Mycobacterium tuberculosis infection. PloS one 7, e38367.

Page 151: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

131 References

Yang, C.S., Shin, D.M., Kim, K.H., Lee, Z.W., Lee, C.H., Park, S.G., Bae, Y.S., and Jo, E.K. (2009). NADPH oxidase 2 interaction with TLR2 is required for efficient innate immune responses to mycobacteria via cathelicidin expression. Journal of immunology 182, 3696-3705. Yang, C.S., Shin, D.M., Lee, H.M., Son, J.W., Lee, S.J., Akira, S., Gougerot-Pocidalo, M.A., El-Benna, J., Ichijo, H., and Jo, E.K. (2008). ASK1-p38 MAPK-p47phox activation is essential for inflammatory responses during tuberculosis via TLR2-ROS signalling. Cellular microbiology 10, 741-754. Yuk, J.M., Shin, D.M., Lee, H.M., Yang, C.S., Jin, H.S., Kim, K.K., Lee, Z.W., Lee, S.H., Kim, J.M., and Jo, E.K. (2009). Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell host & microbe 6, 231-243. Zahrt, T.C., and Deretic, V. (2002). Reactive nitrogen and oxygen intermediates and bacterial defenses: unusual adaptations in Mycobacterium tuberculosis. Antioxidants & redox signaling 4, 141-159. Zhang, Y., Lathigra, R., Garbe, T., Catty, D., and Young, D. (1991). Genetic analysis of superoxide dismutase, the 23 kilodalton antigen of Mycobacterium tuberculosis. Molecular microbiology 5, 381-391. Zhang, Y., and Yew, W.W. (2009). Mechanisms of drug resistance in Mycobacterium tuberculosis. The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease 13, 1320-1330. Zhang, Z., Hillas, P.J., and Ortiz de Montellano, P.R. (1999). Reduction of peroxides and dinitrobenzenes by Mycobacterium tuberculosis thioredoxin and thioredoxin reductase. Archives of biochemistry and biophysics 363, 19-26. Zuber, B., Chami, M., Houssin, C., Dubochet, J., Griffiths, G., and Daffe, M. (2008). Direct visualization of the outer membrane of mycobacteria and corynebacteria in their native state. Journal of bacteriology 190, 5672-5680. Zumla, A., Abubakar, I., Raviglione, M., Hoelscher, M., Ditiu, L., McHugh, T.D., Squire, S.B., Cox, H., Ford, N., McNerney, R., et al. (2012). Drug-resistant tuberculosis--current dilemmas, unanswered questions, challenges, and priority needs. The Journal of infectious diseases 205 Suppl 2, S228-240.

Page 152: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga
Page 153: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

Annex

Page 154: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

134 Annex

Table A1. Description of M. tuberculosis Beijing/W strains belonging to each node found in Figure 1 and

2, and respective country of isolation.

Node Strain Origin of isolation Node Strain Origin of isolation

Bmyc1 NL25 USA Bmyc10 BE5 Singapore

ZA40 South Africa BE8 USA

Bmyc2 NL20 The Netherlands BE10 USA

CN1 China BE12 USA

CN2 China BE14 USA

CN3 China BE17 USA

CN4 China BE19 USA

CN5 China BE20 USA

CN6 China BE23 USA

CN7 China BE27 USA

CN8 China MG3 Madagascar

CN9 China MG10 Madagascar

ZA13 South Africa MG11 Madagascar

ZA42 South Africa MG21 Madagascar

ZA43 South Africa CN60 China

ZA51 South Africa CN61 China

Bmyc3 NL34 USA CN62 China

Bmyc4 NL17 USA CN63 China

BE26 USA CN64 China

CN10 China CN65 China

CN11 China CN66 China

CN12 China CN67 China

CN13 China CN68 China

CN14 China CN69 China

CN15 China CN70 China

CN16 China CN71 China

CN17 China CN72 China

CN18 China CN73 China

ZA45 South Africa CN74 China

ZA52 South Africa CN75 China

Bmyc5 NL33 USA CN76 China

Bmyc6 NL19 South Korea CN77 China

CN20 China CN78 China

CN21 China CN79 China

CN22 China CN80 China

CN23 China CN81 China

CN24 China CN82 China

CN25 China CN83 China

Bmyc7 BE3 South Korea CN84 China

Bmyc8 NL21 The Netherlands CN85 China

Bmyc9 MG9 Madagascar CN86 China

Bmyc10 NL1 Mongolia CN87 China

NL2 South Africa CN88 China

NL3 Malaysia CN89 China

NL7 Thailand CN90 China

NL18 The Netherlands CN91 China

NL23 The Netherlands CN92 China

NL24 The Netherlands CN93 China

NL31 The Netherlands CN94 China

BE1 USA CN95 China

Page 155: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

135 Annex

Node Strain Origin of isolation Node Strain Origin of isolation

Bmyc10 CN96 China Bmyc10 CN150 China

CN97 China CN151 China

CN98 China CN153 China

CN99 China CN154 China

CN100 China CN155 China

CN101 China CN156 China

CN102 China CN157 China

CN103 China CN158 China

CN104 China CN159 China

CN105 China CN160 China

CN106 China CN161 China

CN107 China CN162 China

CN108 China CN163 China

CN109 China CN164 China

CN110 China CN165 China

CN111 China CN166 China

CN112 China CN167 China

CN113 China CN168 China

CN114 China CN169 China

CN115 China CN170 China

CN116 China CN171 China

CN117 China CN172 China

CN118 China CN173 China

CN119 China CN174 China

CN120 China CN175 China

CN121 China CN176 China

CN122 China CN177 China

CN123 China CN178 China

CN124 China CN179 China

CN125 China CN180 China

CN126 China CN181 China

CN127 China CN182 China

CN128 China ZA1 South Africa

CN129 China ZA2 South Africa

CN130 China ZA3 South Africa

CN131 China ZA4 South Africa

CN132 China ZA5 South Africa

CN133 China ZA6 South Africa

CN134 China ZA7 South Africa

CN135 China ZA8 South Africa

CN136 China ZA10 South Africa

CN137 China ZA11 South Africa

CN138 China ZA12 South Africa

CN139 China ZA15 South Africa

CN140 China ZA16 South Africa

CN141 China ZA17 South Africa

CN142 China ZA18 South Africa

CN143 China ZA20 South Africa

CN144 China ZA21 South Africa

CN145 China ZA22 South Africa

CN146 China ZA23 South Africa

CN147 China ZA24 South Africa

CN148 China ZA25 South Africa

CN149 China ZA26 South Africa

Page 156: Mycobacterium tuberculosis host adaptation and evolution … · Mycobacterium tuberculosis host adaptation and evolution reflected by defense mechanisms against oxidative stress Olga

136 Annex

Node Strain Origin of isolation Node Strain Origin of isolation

Bmyc10 ZA28 South Africa Bmyc25 GC1237 Spain

ZA29 South Africa ZA9 South Africa

ZA31 South Africa ZA14 South Africa

ZA32 South Africa ZA19 South Africa

ZA33 South Africa ZA35 South Africa

ZA34 South Africa CN26 China

ZA36 South Africa CN27 China

ZA38 South Africa CN28 China

ZA41 South Africa CN29 China

ZA44 South Africa CN30 China

ZA46 South Africa CN31 China

ZA47 South Africa CN32 China

ZA48 South Africa CN33 China

ZA49 South Africa CN34 China

ZA50 South Africa CN35 China

ZA53 South Africa CN36 China

ZA54 South Africa CN37 China

ZA55 South Africa CN38 China

ZA56 South Africa CN39 China

ZA57 South Africa CN40 China

Bmyc11 NL4 China CN41 China

Bmyc12 NL28 South Africa CN42 China

BE6 Russia CN43 China

BE9 China CN44 China

Bmyc13 MG8 Madagascar CN45 China

MG12 Madagascar CN46 China

CN183 China CN47 China

CN184 China CN48 China

CN185 China Bmyc26 CN49 China

CN186 China CN50 China

CN187 China CN51 China

CN188 China CN52 China

CN189 China CN53 China

CN190 China CN54 China

CN191 China CN55 China

CN192 China CN56 China

Bmyc14 MG7 Madagascar CN57 China

Bmyc15 NL6 Malaysia CN58 China

Bmyc16 MG13 Madagascar CN59 China

MG14 Madagascar ZA30 South Africa

MG15 Madagascar ZA58 South Africa

MG16 Madagascar

MG17 Madagascar

MG19 Madagascar

MG20 Madagascar

Bmyc17 MG6 Madagascar

Bmyc18 BE21 USA

Bmyc19 MG4 Madagascar

MG5 Madagascar

Bmyc20 BE13 USA

Bmyc21 BE16 USA

Bmyc22 NL5 Thailand

Bmyc23 BE15 Philippines