dspace cover page - research collection29018/... · cytotoxic legionella pneumophila lipid a...

123
Research Collection Doctoral Thesis Identification, characterization and differential expression of a cytotoxic Legionella pneumophila lipid A disaccharide synthase paralogue Author(s): Albers, Urs Publication Date: 2006 Permanent Link: https://doi.org/10.3929/ethz-a-005270639 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection . For more information please consult the Terms of use . ETH Library

Upload: hoangtruc

Post on 21-May-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Research Collection

Doctoral Thesis

Identification, characterization and differential expression of acytotoxic Legionella pneumophila lipid A disaccharide synthaseparalogue

Author(s): Albers, Urs

Publication Date: 2006

Permanent Link: https://doi.org/10.3929/ethz-a-005270639

Rights / License: In Copyright - Non-Commercial Use Permitted

This page was generated automatically upon download from the ETH Zurich Research Collection. For moreinformation please consult the Terms of use.

ETH Library

Page 2: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Diss. ETH No. 16773

Identification, characterization and differential expression

of a cytotoxic Legionella pneumophila lipid A disaccharide

synthase paralogue

A dissertation submitted to the

SWISS FEDERAL INSTITUTE OF TECHNOLOGY ZORICH

for the degree of

DOCTOR OF NATURAL SCIENCES

presented by

Urs Albers

Diplom - Biotechnologe,Technical University Braunschweig

born on April 29, 1971from Kleve, Germany

accepted on the recommendation of

Prof. Dr. H. Hilbi, examinerProf. Dr. W.-D. Hardt, co-examiner

Zurich, 2006

Page 3: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Abstract

Abstract

Legionella pneumophila replicates intracellularly in a specific vacuole in phagocytic cells.

The major virulence determinant is the !cm/Dot type IV secretion system which is required

for intracellular growth. An icmG mutant strain shows a partial growth defect in presence of

the amoeba Acanthamoeba castellanii on agar plates and was used to screen a genomic

L. pneumophila library to identify proteins involved in virulence. Thus, suppressors of the

growth defect of the icmG mutant strain were selected. The genes responsible for the

suppression were identified on the library plasmid by deletion analysis. Increased survival in

presence of amoebae was found not to be caused by more efficient intracellular growth but by

an enhanced cytotoxicity for amoebae. Therefore, the genes were termed Legionella

cytotoxic suppressors (les). Cytotoxic genes identified showed similarities to hydrolases

(lcsA), NlpD-related metalloproteases (lcsB), lipid A disaccharide synthases (lesC), and ABC

transporters (lcsD). Overexpression of lcsC strongly increased cytotoxicity of /iicmG, but not

of other icm/dot mutant strains, on amoebae.

The L. pneumophila genome contains two paralogues of the lipid A disaccharide synthase:

lpxBl/lcsC and lpxB2. The corresponding proteins are 34% or 43%, respectively,

homologuous to the E. coli lipid A disaccharide synthase LpxB. The genome of

L. pneumophila contains not only two paralogues of lpxB, but also the acyl transferases lpxA,

lpxD and lpxL are present in more than one copy. Presence of multiple copies of lipid A

biosynthesis genes was only found in L. pneumophila but not in the genomes of any other

bacterial species. Genomic localization of the lpxB paralogues and their ability to

complement a temperature-sensitive E. coli lpxB mutant indicate that both paralogues

function as a lipid A disaccharide synthase.

Expression of the lpxA and lpxB paralogues under different conditions was examined using

reverse transcription and PCR. lpxAl and lpxBl/lesC are more strongly expressed compared

to their paralogues lpxA2 and lpxB2 during early stages of infection of A. castellanii and are

upregulated upon transfer of a stationary phase culture from nutrient broth into distilled

water.

Structural modifications of lipid A affect the protective functions of the outer membrane and

the degree of virulence. The presence of multiple acyl transferase genes of the lipid A

biosynthesis pathway and the expression of lpxA and lpxB paralogues depending on different

environmental conditions suggest that L. pneumophila adjusts the structure of lipid A, most

2

Page 4: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Abstract

likely the fatty acid composition, to adapt to the diverse environmental niches inside and

outside of host cells.

The screen for virulence genes was performed in the icmG mutant strain, and overexpression

of lcsC rendered AicmG cytotoxic, but not other partially virulence-defective mutants.

Therefore, another project adressed the function of the IcmG protein. IcmG contains a

transmembrane domain, and an N-terminal domain was shown to bind to RalF and the Sid

effector proteins. A C-terminal coiled-coil region was predicted to function as a SNARE

domain. However, we could neither detect secretion of IcmG into the supematant nor

translocation into the host cell. Since translocation is expected to be required for a SNARE

protein, it is unlikely that IcmG acts as a SNARE. An icmG mutant strain is still able to

secrete the type IV effector protein SidC, but shows a decreased recruitment of ER-derived

vesicles to the Legionella containing vacuole. We therefore propose that IcmG acts as a

coupling protein increasing the efficiency of type IV effector translocation into the host rather

than acting as a SNARE protein.

3

Page 5: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Zusammenfassung

Zusammenfassung

Legionella pneumophila vennehrt sich intrazelluUir in einer spezifischen Vacuole in

phagozytierenden Zellen. Wichtigster Virulenzfaktor ist das Icm/Dot-Typ IV

Sekretionssystem, dessen Funktion notwendig fur intrazelluHires Wachstum ist. In

Anwesenheit der Amobe A. castellanii zeigt ein Stamm mit einer Deletion in icmG einen

partiellen Wachstumsdefekt auf Agar-Platten. Dieser Stamm wurde benutzt, um eine

genomische L. pneumophila-Bibliothek nach Genen zu screenen, die zur Virulenz beitragen.

Diejenigen Klone wurden isoliert, bei denen der Wachstumsdefekt der icmG Mutanten

supprimiert war. Die fur die Suppression verantwortlichen Gene wurden mittels

Deletionsanalyse auf den Bibliotheksplasmiden identifiziert. Das verbesserte Uberleben der

Bakterien in Anwesenheit von Amoben wurde nicht durch verstarktes intrazellulares

Wachstum, sondern durch eine verstarkte zytotoxische Wirkung auf die Amoben verursacht.

Die Gene wurden daher Legionella cytotoxic suppressor (lcs) genannt. Die identifizierten

Gene sind homolog zu Hydrolasen (lcsA), NlpD-artigen Metalloproteasen (lcsB), Lipid A

Disaccharidsynthasen (lcsC) und ABC-Transportern (lcsD). Uberexpression von lcsC in

6.icmG, nicht aber in anderen partiell virulenten Mutanten, erhohte die zytotoxische Wirkung

auf Amoben betrachtlich.

Das Genom von L. pneumophila enthalt zwei Paraloge der Lipid A Disaccharidsynthase:

lpxBI/lcsC und lpxB2. Die entsprechenden Proteine sind 34% beziehungsweise 43%

homolog zur Lipid A Disaccharidsynthase LpxB von E. coli. Im Genom von L. pneumophila

sind nicht nur lpxB, sondem auch die Acyltransferasen lpxA, lpxD and lpxL mehrfach

vorhanden. Das Vorhandensein mehrfacher Kopien von Lipid A Biosynthesegenen ist

spezifisch fur 1. pneumophila und konnte in den Genomen anderer Bakterien nicht gefunden

werden. Die Lage der lpxB Paraloge im Genom und ihre Fahigkeit zur Komplementation

einer temperatursensitiven E. coli lpxB Mutante zeigen, dass beide Paraloge Lipid A

Disaccharidsynthase-Aktivitat besitzen.

Die Expressionsstarke der lpxA und lpxB Paraloge wurde mittels reverser Transkription und

PCR untersucht. Im Vergleich mit ihren Paralogen lpxA2 und lpxB2 sind lpxAl und

lpxBlllcsC in fiiihen Stadien der Infektion von A. castellanii starker exprimiert und werden

beim Transfer einer stationaren Kultur von Flussigmedium in destilliertes Wasser

heraufreguliert.

4

Page 6: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Zusammenfassung

Strukturelle Anderungen von Lipid A beeinflussen die protektiven Funktionen der ausseren

Membran und das Ausmass der Virulenz. Das Vorhandensein mehrerer Acyltransferasen des

Lipid A Biosyntheseweges und die differentielle Expression der IpxA und IpxB Paraloge

deuten darauf hin, dass 1. pneumophila die Struktur von Lipid A, insbesonders die

Fettsaurezusammensetzung, an die verschiedenen Nischen innerhalb und ausserhalb der

Wirtszelle anpasst.

Der Screen zur Identifizierung von Virulenzgenen wurde in icmG Mutanten durchgefiihrt.

Uberexpression von IcsC erhohte die Zytotoxizitat von tlicmG, nicht aber von anderen

Mutanten mit eingeschrankter Virulenz. In einem weiteren Projekt wurde die Funktion von

IcmG untersucht. IcmG enthalt eine Transmembrandomane, und eine N-terminale Domane,

die an der Bindung von RalF und den Sid Effektorproteinen beteiligt ist. Eine Funktion als

SNARE-Domane wurde fur einen C-terminalen coiled-coil Bereich von IcmG vorhergesagt.

Wir konnten jedoch weder Sekretion von IcmG in den Kulturliberstand noch Translokation in

die Wirtszelle feststellen. Da zu erwarten ist, dass die Translokation Voraussetzung fur ein

SNARE-Protein ist, ist eine SNARE-artige Funktion von IcmG unwahrscheinlich. Die icmG

Mutante kann das Typ IV-Effektorprotein SidC sekretieren, ist aber beeintrachtigt in der

Rekrutierung von ER-Vesikeln zur Legionella-enthaltenden Vakuole. Wir vermuten daher,

dass IcmG kein SNARE-Protein ist sondem als "coupling protein" agiert, das die Effizienz

der Typ IV-Sekretion erhoht.

5

Page 7: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Table of contents

Table of contents

ABSTRACT 2

ZUSAMMENFASSUNG 4

TABLE OF CONTENTS 6

1 GENERAL INTRODUCTION 8

1.1 ENVIRONMENTAL GROWTH AND DIFFERENTIATION OF LEGIONELLAE 81.2 LEGIONELLA - THE ETIOLOGICAL AGENT OF LEGIONNAIRES' DISEASE 91.3 INTRACELLULAR REPLICATION AND VIRULENCE FACTORS OF L. PNEUMOPHILA 101.4 THE ICM/DOT TYPE IV SECRETION SYSTEM 111.5 LEGIONELLA LPS 14

/.5./ Modifications oflipid A 161. 5.2 Interaction oflipid A with components ofthe innate immune system 19

1.6 LITERATURE 20

2 THE AMOEBAE PLATE TEST IMPLICATES A PARALOG OF LPXB IN THE INTERACTIONOF LEGIONELLA PNEUMOPHlLA WITH ACANTHAMOEBA CASTELLANII 25

2.1 ABSTRACT 262.2 INTRODUCTION 262.3 METHODS 28

2.3.1 Bacterial strains, cell culture and reagents 282.3.2 Construction ofplasmids 302.3.3 The amoebae plate test 3/2.3.4 Screening ofan 1. pneumophila chromosomal library for icm/dot suppressors using the APT. .. 322.3.5 Analysis ofsuppressor plasmids 332.3.6 1ntracellular growth inA. castellanii and RAW264. 7 macrophages 332.3.7 Cytotoxicity assay 332.3.8 Computational and statistical analysis 34

2.4 RESULTS 342.4.1 Growth of1. pneumophila on CYE agar plates in presence ofA. castellanii-

the amoeba plate test 342.4.2 1solation of1. pneumophila icm/dot suppressor strains using the amoebae plate test.. 362.4.3 Analysis ofsuppressor plasmid inserts 382.4.4 Intracellular growth oficmG suppressor strains in A. castellanii and macrophages 402.4.5 icmG suppressor strains are cytotoxicfor A. castellanii 422.4.6 Identification ofcytotoxic suppressor genes by deletion analysis 442.4. 7 Overexpression ofsuppressor genes 46

2.5 DISCUSSION 492.5.1 Screen for icm suppressors using the amoebae plate test... 492.5.2 LcsB, a homolog ofNlpD-like membrane-bound metalloproteases 502.5.3 LcsC, aparalog ofthe lipidA disaccharide synthase LpxB 522.5.4 Mechanism ofLcsC cytotoxicity 53

2.6 ACKNOWLEDGEMENTS 542.7 REFERENCES 54

3 THE CYTOTOXIC LEGIONELLA PNEUMOPHlLA LPXB PARALOGUE LCSC IS INVOLVEDIN LIPID A BIOSYNTHESIS AND UPREGULATED DURING BACTERIAL GROWTH INAMOEBAE 60

3.1 ABSTRACT 613.2 INTRODUCTION 613.3 METHODS 63

3.3.1 Bacteria, amoebae and reagents 633.3.2 Identification ofhomologues of1. pneumophila lipid A biosynthesis genes 643.3.3 Construction ofplasmids 643.3.4 Complementation ofa conditional E. coli IpxB mutant strain with 1. pneumophila

IpxB paralogues 65

6

Page 8: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Table of contents

3.3.5 Cytotoxicity assay 653.3.6 Expression analysis ofLPS biosynthesis genes by RT-PCR 66

3.4 RESULTS 683.4.1 L. pneumophilaparalogues ofLPS biosynthesis genes 683.4.2 Complementation ofa conditional E. coli IpxB mutant strain with L. pneumophila

IpxB paralogues 703.4.3 Cytotoxicity ofL. pneumophila IpxB paraloguesfor A. castellanii 713.4.4 Expression ofL. pneumophila lpx genes during growth in a complex medium 733.4.5 Expression ofL. pneumophila lpx genes under osmotic stress and nutrient deprivation 753.4.6 Expression ofL. pneumophila lpx genes during intracellular growth in A. castellanii 76

3.5 DISCUSSION 773.5.1 Lipid A biosynthesis paralogues and their functions 773.5.2 Differential regulation ofL. pneumophila lpx paralogues 783.5.3 Biological implications oflipid A and LPS modifications 79

3.6 ACKNOWLEDGEMENTS 803.7 REFERENCES 81

4 L. PNEUMOPHILA ICMG INCREASES THE EFFICIENCY OF EFFECTOR PROTEINSECRETION 85

4.1 INTRODUCTION 854.2 MATERIALS AND METHODS 87

4.2.1 Cultivation ofcells 874.2.2 Sequence analysis ofIcmG 874.2.3 Construction of icmG expression vectors 874.2.4 Pull-down assays using GSTfusion proteins 884.2.5 Copurification oflcmG and binding proteins using His6-tagged IcmG 904.2.6 Generation ofantibodies and Western blot 904.2.7 Immunofluorescence microscopy 91

4.3 RESULTS 914.3.1 IcmG is present in the bacteria in low amounts 914.3.2 lcmG is not translocated during infection 934.3.3 L. pneumophila .dicmG is able to translocate SidC but is impaired in recruiting ER-derived

vesicles to the LCV 964.3.4 Identification ofIcmG bindingproteins using a pull down assay with GSTfusion proteins 984.3.5 Copurification oflcmG binding proteins using His~tagged IcmG 101

4.4 DISCUSSION 1034.4.1 Expression level and localization ofIcmG 1044.4.2 Phenotypes ofthe icmG mutant 1044.4.3 Identification ofIcmG binding proteins in pull down assays 1054.4.4 Hypotheses on the function ofIcmG 106

4.5 REFERENCES 108

5 GENERAL DISCUSSION 111

5.1 LcsC SHOWS LIPID A DISACCHARIDE SYNTHASE ACTIVITy 1115.2 L. PNEUMOPHILA: LIFE IN DIFFERENT NICHES AND ADAPTATION OF LIPID A STRUCTURE 1125.3 INTRACELLULAR LPS INFLUENCES HOST CELL FUNCTIONS 1135.4 POSSIBLE INFLUENCE OF LIPID A ON OUTER MEMBRANE VESICLES 1145.5 CYTOTOXICITY OF THE LPxB PARALOGUES 1155.6 PUTATIVE MECHANISMS OF LcsC MEDIATED CyTOTOXICITy 1165.7 SPECIFITY OF LCSC-MEDIATED CYTOTOXICITY FOR aIcMG 1175.8 CONCLUSION 1185.9 LITERATURE 118

CURRICULUM VITAE 120

PUBLICATIONS 121

7

Page 9: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

1 General introduction

1.1 Environmental growth and differentiation of Legionellae

Legionella spp. are Gram-negative, aerobic, monopolarly flagellated rod-shaped bacteria

belonging to the gamma subdivision of proteobacteria. More than 50 Legionella species have

been described so far (http://www.bacterio.cict.fr). Legionellae can be found ubiquitously in

aquatic habitats and soil. In the environment, 1. pneumophila parasitizes within a large

variety of amoebae and protozoa rather than multiplying planctonically. However, in the

laboratory, 1. pneumophila can grow in nutrient rich medium. High numbers of

1. pneumophila are often found in biofilms, where they can persist for long times and infect

amoebae grazing on bacteria (Fields, 1996; Steinert et al., 2002).

Growth of 1. pneumophila can be divided into two phases; replication inside of a host cell

after infection, and transmission to a new host, during which the bacteria have to survive in

the environment. Both phases expose the bacteria to highly diverse conditions.

1. pneumophila adapts to the two phases, and the differentiation state of the bacterium can be

distinguished by changes in, e.g., stress resistance, flagellum expression, evasion of lysosome

fusion within a host cell, cytotoxicity and sodium sensitivity (Hammer et al., 2002). The

switch between these developmental stages is coordinated by a regulatory circuit that is

controled by amino acid concentration and probably also other signals. During the replicative

phase, when nutrients are abundant, the post-transcriptional regulator CsrA represses

transmission traits. When amino acids become limiting, RelA produces the alarmone ppGpp

that leads to expression of transmissive traits via the two component system LetAlS, the

enhancer protein LetE, the alternative sigrna factor RpoS and probably the master flagellar

regulator RpoN (Molofsky and Swanson, 2004). The physiological changes of

1. pneumophila during infection of amoebae closely resemble those during growth in broth

cultures. During intracellular replication and in exponential growth phase in broth, traits that

promote transmission (stress resistance, motility, ability to evade phagosome-lysosome

fusion, cytotoxicity) are repressed. As nutrients become limiting either inside the host or in

spent nutrient broth during entry in stationary phase, the bacteria switch to the transmissive

phase and derepress the traits increasing survival and infection of a new host.

Despite of a generally low number of Legionellae in aquatic habitats and soils, the abundance

of bacteria in a habitat is strongly increased by the presence of protozoa, reflecting the

requirement for intracellular replication. Furthermore, protozoa can represent a shelter when

8

Page 10: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 1

environmental conditions become unfavourable. The bacteria can survive high temperature,

disinfection and drying if enclosed within an Acanthamoeba cyst (Heuner et aI., 2002).

Replication in amoebae also influences the physiology of the bacteria. Compared to cultures

grown in broth, cells released from A. castellanii enter human monocytes more efficiently

and are more resistant to biocides and antibiotics (Swanson and Hammer, 2000).

1.2 Legionella - the etiological agent of Legionnaires' disease

The ubiquitous presence of Legionellae can lead to contamination of man-made technical

systems. Elevated temperatures and the presence of biofilms and protozoa play a key role for

growth, spreading and persistance of Legionella in these devices. Human infection occurs

exclusively by inhalation of contaminated aerosols, which can be produced by air

conditioning systems, cooling towers, whirlpools, fountains, dental devices or shower heads

(Steinert et al., 2002). Inhaled bacteria infect alveolar macrophages in a way very similar to

amobae and may cause the severe pneumoniae known as Legionnaires' disease. 90% of the

isolates associated with Legionaire's disease are identified as L. pneumophila, most of them

belonging to serogroup 1. In Switzerland, the incidence of Legionaire's disease since 2000 is

2 cases per 100000 inhabitants with a mortality rate of6.5% (www.bag.ch)

L. pneumophila is relatively resistant to innate and humoral immune responses. The bacteria

resist complement-mediated lysis and are not killed by polymorphonuclear cells, even when

opsonized with complement or specific antibodies. In a mouse model of Legionnaires'

disease, lung infection is controlled by cell-mediated immunity with interferon-gamma

release by natural killer cells being a key element (Sporri et al., 2006; Swanson and Hammer,

2000). A robust immune response is sufficient to clear L. pneumophila infections. Main risk

factors for legionellosis are immunosuppression and lung diseases.

Despite a high number of reported cases, not a single human to human transmission has been

identified. L. pneumophila is an opportunistic pathogen whose ability to infect humans likely

is a consequence of its coevolution with amoebae, rather than being due to adaptation to

humans. The bacteria evolved as parasites of amoebae and aquired capabilities to infect

protozoa and also mammalian cells due to the similarity of host cell mechanisms that are

manipulated during infection (Swanson and Hammer, 2000).

9

Page 11: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

1.3 Intracellular replicationL. pneumophila

and virulence factors of

Upon contact of 1. pneumophila with a monocyte or an amoeba, the bacterium is taken up by

conventional or coiling phagocytosis (Bozue and JoOOson, 1996; Horwitz, 1984). The major

outer membrane protein MOMP is bound by the complement protein C3 and can mediate

uptake via CRI and CRJ receptors of macrophages. Phagocytosis may also be enhanced by

opsonization with immunoglobulins (Swanson and Hammer, 2000). Lectins have been shown

to be involved in attachment and invasion of amoebae (Venkataraman et al., 1997). Other

proteins likely involved in adherence and invasion are type IV pili, that can mediate a

complement-independent attachment, and the 60 kDa heat shock protein Hsp60 (Hoffman et

al., 1990; Stone and Abu Kwaik, 1998).

Independent from the mechanism of phagocytosis, 1. pneumophila is able to to inhibit fusion

of the Legionella-containing vacuole (LCV) with lysosomes in an !cm/Dot-dependent manner

(see below). Instead, the phagosomes recruit early secretory vesicles and ER-derived vesicles

to generate a replication-permissive vacuole (Kagan et al., 2004; Roy and Tilney, 2002).

After a 6-10 hours lag phase in macrophages, the bacteria start to replicate. Concomitantly,

the ability to prevent phagosome-lysosome fusion decreases and the vacuole finally fuses

with lysosomes. This maturation into an acidic organelle is necessary for successful

multiplication (Sturgill-Koszycki and Swanson, 2000). Finally, amino acid depletion triggers

the switch from replicative to transmissive phase, genes necessary for motility and

transmission are expressed, and the host cell lyses and releases the bacteria.

A number of factors has been described to be involved in virulence. The presence of a

flagellum enhances the invasion capacity but is not required for intracellular growth. The Mip

protein (macrophage infectivity potentiator) is exposed on the bacterial surface, contributes to

intracellular survival and exhibits peptidyl-prolyl-cis/trans isomerase activity. Since this

activity is characteristic for eukaryotic cells, it might be that Mip targets a host protein

(Steinert et al., 2002; Swanson and Hammer, 2000). Other factors involved in virulence are

phospholipases, proteases and hydrolases, some of which are secreted via a type II secretion

system (reviewed in Cianciotto, 2001).

10

Page 12: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

!cm/dot mutant Legionella wild type

Chapter I

Figure 1: Intracellular replication of L. pneumophila. The bacteria are phagocytosed by the hostcell. Phagosomes containing non-pathogenic bacteria or L. pneumophila icrn/dot mutants fuse withlysosomes, and the bacteria are killed and degraded. Vacuoles containing wild-type L. pneumophilaevade phagosome-lysosome fusion and associate with smooth vesicles, mitochondria and ER-derivedvesicles. Within this specialized vacuole, the bacteria convert to the replicative form and multiply. Atlater stages of the infection, the vacuole merges with lysosomal compartments and acidifies. Upondepletion of the nutrient supply, the bacteria switch to the transmissive form, lyse the host and aredispersed in the environment where they can infect a new host. (Picture from Andre Tiaden andStefan Weber).

1.4 The Icm/Dot type IV secretion system

The mechanisms enabling L. pneumophila to grow intracellularly are similar both in free­

living unicellular eukaryotes like A. castellanii and in phagocytic cells of the immune system

like macrophages. Intracellular growth reqUIres the Icrn/Dot (intracellular

multiplication/defective for organelle trafficking) type IV secretion system. Type IV

secretion systems are related to conjugation systems and enable bacteria to deliver

macromolecules from the bacterial cytosol through the bacterial cell wall and the host cell

membrane directly into the host cytosol (Figure 2). The 25 L. pneumophila icm/dot genes are

located in two clusters in the genome, and most of them are predicted to encode membrane-

11

Page 13: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

spanning proteins (Segal et al., 1998; Vogel et aI., 1998). Functional characterization of the

single lcm and Dot proteins is still at the beginning, and most information comes from in

silico analysis and homology searches. DotA is an integral cytoplasmatic membrane protein

that can be secreted into the supernatant in an !cm/Dot dependent manner. Secreted DotA

polymerizes to hollow rings of unknown function. DotB, lcmO, lcmB and IcmF contain

Walker A motifs. For some of these proteins, ATP hydrolyzation was found to be important

for efficient intracellular growth. IcmO shows homologies to the TraG-like coupling protein

family, but functional studies have yet to be done. IcmG is partially required for intracellular

growth and is the only Icm/Dot protein for which binding to effector proteins has been

described so far (see Chapter 4). IcmB and IcmK were shown to associate with a fibrous

structure in the surface upon incubation of bacteria with macrophages or with macrophage

conditioned medium. IcmR possesses chaperon activity on IcmQ and prevents the formation

of IcmQ homopolymers. IcmQ is exposed on the bacterial surface and forms pores in lipid

membranes in the absence of IcmR. The data known so far about the Icm/Dot secretion

system are summarized in a recent review (Segal et al., 2005).

Host cell

Legionella , A ,,j}~

Effector protein Conjugative plasmid

Figure 2: Type IV secretion systems enable the bacterium to deliver effector proteins directlyinto the host cell cytosol. Schematic representation of a type IV secretion system (© by GunnarSchroeder, after Christie, 2001).

12

Page 14: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 1

In L. pneumophila, mutation analysis of icrn/dot genes revealed a requirement for the large

majority of Icm/Dot proteins for several steps of the infection. L. pneumophila actively

upregulates its own phagocytosis by usage of the !cm/Dot system both in macrophages and

amoebae (Hilbi et al., 2001; Watarai et al., 2001). Infection of macrophages with a high

multiplicity of infection results in increased cytotoxicity caused by osmotic lysis after

insertion of pores into the host cell membrane in an Icm/Dot dependent manner (Kirby and

Isberg, 1998; Kirby et al., 1998). Inhibition of phagosome-lysosome fusion and recruitment

of early secretory vesicles from ER exit sites also depend on a functional Icm/Dot system

(Roy et al., 1998; Wiater et al., 1998). After formation of a specialized organelle, bacterial

replication proceeds without requiring a functional Icm/Dot system (Coers et al., 1999). It has

also been suggested that the IcmIDot system is responsible for formation of a pore in the host

cell after depletion of the host to allow the bacteria to lyse the cell and exit (Molmeret and

Abu Kwaik, 2002). Furthermore, due to the relation to conjugal systems, the IcmlDot system

is cabable to conjugate plasmids. Mobilizable plasmids inhibit intracellular growth and

killing of macrophages probably by competition for the IcmIDot transporter capacity with

other substrates of the !cm/Dot system (Segal and Shuman, 1998).

Only recently, Icm/Dot-transported effector proteins have been identified. RalF is

translocated into the host cell and acts as a guanine nucleotide exchange factor that recruites

the small GTPase ARF1, a highly conserved regulator of vesicle trafficking, to the

1. pneumophila phagosome (Nagai et al., 2002). In an elegant screen, the Sid family of

secreted effector proteins was identified (Luo and Isberg, 2004). The authors used a bacterial

two-hybrid screen with RalF as bait to identify effector protein binding partners. A large

portion of the proteins obtained in the screen comprised of IcmG. Assuming that IcmG also

binds other effector proteins, it was used as bait in a second bacterial two hybrid screen to

identify further effector proteins. Translocation of the proteins identified in the second screen

was confirmed, and the newly identified effector proteins were named Sid (substrates of

Icm/Dot). Most of the Sid proteins exist as families of paralogues. SidC anchors to the LCV

by binding to phosphatidylinositol(4) phosphate, a mediator of membrane dynamics in

eukaryotic cells (Weber et al., 2006). LepA and B share sequence similarity with SNARE

proteins and seem to mediate the IcmIDot-dependent release of LCVs from amoebae (Chen et

al., 2004). Another protein translocated by the Icm/Dot system is LidA, which is involved in

recruitment of early secretory versicle to the LCV (Derre and Isberg, 2005).

13

Page 15: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

1.5 Legionella LPS

LPS makes up the majority of lipids in the outer monolayer of the outer membrane of Gram­

negative bacteria. It consists of three components: the O-chain polysaccharide, the core

oligosaccharide and lipid A. Particularly the oligo- and polysaccharide structures can vary

between different strains and species and determine to a large part the classification of a

strain to a particular serogroup. The O-chain from 1. pneumophila LPS is a homopolymer of

single legionaminic acid residues; a unique, hydrophobic carbohydrate (Knirel et al., 1994).

Some 1. pneumophila strains, among these Philadephia 1, contain the gene lag-I that encodes

an O-acetyl transferase that modifies the outer legionaminic acid units of the O-chain.

However, mutation analysis revealed that a lack of O-acetylation does not influence virulence

properties such as serum resistance or uptake and intracellular growth in amoebae and

macrophages (Luck et al., 2001). A further, lag-I-independent O-acetylation of core sugars

and proximal legionaminic acid units seems to be a common feature of L. pneumophila

serogroup 1 strains (Kooistra et al., 2001). The legionaminic acid residues proximal to the

core oligosaccharide can be N-methylated. Again, this modification did not affect serum

resistance or virulence (Kooistra et al., 2002a). The core oligosaccharide is a nonasaccharide

that lacks heptose and phosphate, contains mostly 6-deoxy sugars and is highly O-acetylated

(see Figure 3).

O-chain polysaccharide core oligosaccharide lipid A

R =H, Ac OAc OAc OAc OAc

Figure 3: Representative structure of L. pneumophila serogrouop 1 LPS. The number of repeatingmonosaccharides in the O-chain ranges from about 10 to up to 100 units. GlcN3N: 2,3-diamino-2,3­dideoxy-D-g1ucose, Kdo: 3-deoxy-D-manno-oct~2-ulosonic acid, Man: D-mannose, GlcNAc: 2~

acetamido-2~deoxy.D-glucose (N-acetyl-glucosamine), QuiNAc: 2-acetamido-2,6-dideoxy-D-glucose(N-acetyl-D-quinovosamine), Rha: L-rhamnose, Leg and 4e-Leg: derivatives of legionaminic and 4­epilegionaminic acid, respectively. OAc stands for O-acetyl group. (Picture from Kooistra et al.,2002a).

14

Page 16: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 1

Recently, it was shown that LPS-containing membrane vesicles released by 1. pneumophila

were able to inhibit phagosome-lysosome fusion temporarily for several hours in an IcmlDot­

independent manner. Inhibition occured when beads were coinfected with purified outer

membrane vesicles, or when beads coated with vesicles were used for infection. The ability to

inhibit fusion was observed only for vesicles derived from transmissive phase bacteria but not

from replicative phase bacteria. Furthermore, the ability to inhibit fusion correlated with

developmentally regulated modifications of the pathogen's surface as judged by LPS profiles

and lectin binding (Fernandez-Moreira et al., 2006).

LPS from Legionella jeonii was also shown to be involved in inhibition of phagosome­

lysosome fusion in Acanthamoeba proteus. LPS is released from intracellular bacteria and is

inserted into the membrane of the LCV. Masking of LPS in the phagosome membrane by

microinjection of an anti-LPS antibody increased the number of phagosomes that fused with

lysosomes (Kim et aI., 1994). The influence of LPS on vesicle trafficking suggests its

classification as a virulence factor.

Lipid A is the membrane anchor of LPS. In contrast to the majority of Gram-negative

bacteria, the disaccharide backbone of L. pneumophila lipid A is a derivative of 2,3-dideoxy­

2,3-diamino-D-glucose rather than glucosamine, yielding a lipid A in which all primary fatty

acids are amide linked to the sugar backbone. The fatty acid pattern of lipid A from

L. pneumophila and other Legionella species examined is complex. The acyl chains are

unusually long, partially branched and some of them contain additional hydroxyl groups

(Moll et al., 1992; Sonesson et al., 1989; Sonesson et al., 1994a; Sonesson et al., 1994b, c;

Zahringer et al., 1995). Furthermore, analysis of lipid A from a phase variant strain showed

that L. pneumophila is able to shift the profile of primary fatty acids towards shorter acyl

moieties (Kooistra, 2002b). Phase variation is based on excision and insertion of a 30 kb

unstable element that exists as high copy plasmid in the phase variant strain and is inserted in

the chromosome in the L. pneumophila wild type strain RC 1. Existence as extrachromosomal

plasmid likely affects regulatory circuits and thus influences several bacterial pathways. The

phase variant strain 811 is serum sensitive, unable to replicate intracellularly in the

macrophage cell line HL60 and lacks the N-methylation of legionaminic acid mentioned

above (Luneberg et aI., 2001).

Figure 4 shows the major lipid A structure of 1. pneumophila RCl (serogroup 1, subgroup

OLDA) and the corresponding phase variant mutant 811.

15

Page 17: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

Figure 4: Structure of the major lipid A fromL. pneumophila subgroup OLDA strain RCIand the phase variant mutant.The length of the 3-hydroxylated fatty acids atposition 2 and 2' is n = 20 at both GlcN3Nresidues in wild type. In the phase variant 811, thechain length at positions 2 and 2' is n = 16 and n =

18, respectively. Core oligosaccharides areattached to the 6MOH group of G1cN3N n. (Picturefrom Kooistra, 2002b).

OH

R"HorOHR2 • OH or CH,

OH

o

1.5.1 Modifications of lipid A

The genes and the corresponding enzymes of the lipid A biosynthesis pathway are well

characterized (Raetz and Whitfield, 2002; Sweet et al., 2004), and homologues of all of them

are present in the genome of L. pneumophila.

16

Page 18: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 1

~OH 0 HAD' NADHFOH0 ]L-Gkl a-KG ~OH I-OH-C,.- ~OHA_ I-OH-C,." ~OH UUP ~OH

~ HO ACP HO ACP HO HO

~NHUf1I'~ 0 O",\'*'lKJP~ tt,N -\'*'UOP-'--';J'.'" o=\HllUOP~~i'''' IIlHUOP-L. ;J"" lNHO

-:.OHGnnA Gnn8 LpxA LpxC LpxO HO LpxH H ~

UDP-GlcNAc UOp·GlcNAc3N (2ll)

~o 0

HO.~-O~O

WaaA(kdtA)

--r---r-

LpxBt uoP

OH

LpxK

---,-ATP

Lipid IVA(GlcN3N 8n8logue)

Figure 5: Biosynthesis pathway of lipid A molecules with four amide-linked acyl chains. UDP­GlcNAc: UDP-N-acetyl-glucosamine, UDP-G1cNAc3N: UDP-2-acetamido-3-amino-2,3-dideoxy-a­D-glucopyranose, Kdo: 3-deoxy-D-manno-oct-2-ulosonic acid. (Picture from Sweet et al., 2004).

However, all acyl transferases and the lipid A disaccharide synthase IpxB are present in more

than one copy in the 1. pneumophila genome, but not in any other bacterial genome

examined. At least some paralogues are differentially expressed, suggesting that

1. pneumophila is able to extensively vary the fatty acid composition of lipid A (see Chapter

4). Besides the phase variation-dependent shortening of primary acyl chains (Kooistra,

2002b), little information is published about modifications of1. pneumophila lipid A.

Lipid A is a major component of the outermost bacterial barrier and, therefore, an important

structural element for protection against detrimental compounds from the environment. These

can be antibiotica, cationic antimicrobial peptides (CAMPs) released from other bacteria in a

biofilm, lysosomal contents during infection of host cells, or the arsenal of the innate immune

system during infection of a multicellular organism. Sensitivity against these compounds is

influenced by the lipid A structure, and adaptation of lipid A helps bacteria to survive in

different niches (Miller et al., 2005).

CAMPs are secreted by a wide variety of organisms from animals to unicellular eukaryotes

and Gram-positive bacteria. Their cationic charge allows them to interact with the bacterial

membrane, which usually, and also in the case of 1. pneumophila, comprises negatively

charged phospholipids. The putative mechanism of CAMP-mediated killing of

microorganisms is insertion into and disruption of membranes and thereby loss of barrier

function (Guo et al., 1998). Regulation of lipid A acylation has been demonstrated to

17

Page 19: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

promote resistance to CAMPs. The Salmonella protein PagP adds an additional palmitate to

lipid A, leading to an increased resistance. Thus, pagP mutant strains are more susceptible

against some CAMPs (Guo et al., 1998). Acylation of lipid A likely alters the fluidity of the

outer membrane by strengthening hydrophobic interactions between the increased number or

length of acyl residues. The stronger hydrophobic interaction forces could retard or abolish

insertion of the CAMP into the membrane, thus keeping the outer barrier intact and

preventing CAMPs from reaching the inner membrane, the most important diffusion barrier.

Similarly, the formation of membrane pores by complement proteins could be impaired by

highly acylated, rigid membranes and lead to increased resistance against complement­

mediated killing.

The acylation state of lipid A is adjusted depending on the temperature in E. coli. Under cold

shock conditions (12 QC), the acyl transferase LpxP is activated that inserts palmitoleate (C16)

in place of laurate (Cn) as secondary acyl chain, perhaps reflecting the need to adjust outer

membrane fluidity at low temperatures. Interestingly, a mutant lacking IpxP showed a 10-fold

increased sensitivity to rifampicin and vancomycin at 12°C but not at 30 QC. The increase in

acyl chain length might confer a selective advantage upon E. coli cells growing at lower

temperatures by making the outer membrane a more effective barrier to harmful chemicals

(Carty et al., 1999; Vorachek-Warren et al., 2002).

Yersinia pestis also regulates its lipid A acylation in response to temperature, reflecting its

life in different hosts. At 21°C or when residing in fleas, the natural host, lipid A is highly

acylated and more resistant to CAMPs, the major immune pressure within insects. Contrarily,

at 37°C, lipid A is only tetra-acylated and does not stimulate TLR4 signalling in humans, the

other host, thereby hampering recognition by the immune system (Miller et al., 2005). In this

case, increased temperature may also serve as environmental signal for being inside a

mammalian host, leading to counteractions against the innate immune system.

The importance of the lipid A structure for 1. pneumophila virulence is demonstrated by the

effect of a deletion of rep, a pagP like gene. In S. Typhimurium, pagP is controlled by the

PhoP-PhoQ two component regulatory system and expressed under low magnesium

concentrations as encountered inside a phagolysosome. PagP is an acyl carrier protein­

independent acyl transferase adding palmitate to the primary acyl chain at position 2, thus

increasing resistance to some CAMPs (Guo et al., 1998).1. pneumophila rep was identified

due to its homology to pagP. Corresponding to pagP, resistance to CAMPs was induced by

growth in low magnesium medium, and an rep mutant showed decreased resistance to

different classes of CAMPs. The rep mutant exhibited a decreased recovery in the amoebae

18

Page 20: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 1

Hartmanella vermiformis and U937 macrophages and was less successful in lung

colonization of A/J mice (Robey et al., 2001). Although attachment of an additional acyl

chain to lipid A by the rcp gene product has not yet been experimentally proven, it is likely

that Rcp modifies lipid A in a way similar to its homologue PagP, and the mutant phenotypes

observed are caused by changes in lipid A acylation that impair the functions of the outer

membrane as a protective barrier.

1.5.2 Interaction of lipid A with components of the innate immunesystem

Structural modifications of lipid A fatty acids may also significantly affect the course of an

infection in multicellular host organisms. Lipid A is the active part of endotoxin, the only

region of LPS to be recognized by the innate immune system and is highly stimulatory even

at low concentrations. Structure-function analyses of lipid A signalling indicate that the

length and number of acyl side chains are critical for TLR4 signalling in humans. Hexa­

acylated lipid A with chain lengths of 12 to 14 carbon atoms is maximally stimulating in

human cells, whereas altering the number or length of the fatty acids can reduce the

magnitude of the signal (Miller et al., 2005). L. pneumophila lipid A fatty acid chains vary in

length from 14 to 20 carbon atoms, and the secondary acyl chains even consist of 16 or 26

carbon atoms. Furthermore, some of the acyl moieties are branched (see Figure 4), probably

hampering recognition. Lipid A interacts with LPS binding protein (LBP) in the serum and is

delivered by LBP to CD14 receptors on the cell surface, where the complex interacts with

TLR4. TLR4 binding induces signal transduction leading to secretion of cytokines and

cationic antimicrobial peptides and, assists in activating the aquired immune response.

L. pneumophila LPS fails to bind the LPS receptor CD14, probably due to its unusual lipid A

structure, and therefore, shows a low endotoxic potential in the human monocytic cell line

MonoMac6 (Neumeister et aI., 1998). The unusual composition of L. pneumophila lipid A

fatty acids might help the bacteria to escape recognition by the innate immune system by

altering the LPS pathogen associated molecular pattern (PAMP). Further evidence for an

influence of the fatty acid composition on endotoxic activity is given by the effect of fatty

acid removal. E. coli IpxM mutant strains lacking a lipid A secondary acyl chain are greatly

attenuated in their ability to activate human macrophages, and a Salmonella IpxM mutant was

much less lethal in an animal shock model than wild type bacteria (Raetz and Whitfield,

19

Page 21: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

2002). The importance of lipid A in detection and clearance of infections is highlighted by

the observation that Tlr4-null mice lacking endotoxin-dependent activation are highly

susceptible to infection with Gram-negative microorganisms (Miller et al., 2005). Therefore,

the lipid A structure, particularly the fatty acid composition, is expected to influence

significantly the virulence ofL. pneurnophila in multicellular organisms.

1.6 Literature

Bozue, lA., and lohnson, W. (1996) Interaction of Legionella pneurnophila withAcantharnoeba castellanii: uptake by coiling phagocytosis and inhibition ofphagosome-lysosome fusion. Infect Irnrnun 64: 668-673.

Carty, S.M., Sreekumar, K.R., and Raetz, C.R. (1999) Effect of cold shock on lipid Abiosynthesis in Escherichia coli. Induction at 12 degrees C of an acyltransferasespecific for palmitoleoyl-acyl carrier protein. J BioI Chem 274: 9677-9685.

Chen, l., de Felipe, K.S., Clarke, M., Lu, H., Anderson, O.R., Segal, G., and Shurnan, H.A.(2004) Legionella effectors that promote nonlytic release from protozoa. Science 303:1358-1361.

Christie, P.J. (2001) Type IV secretion: intercellular transfer of macromolecules by systemsancestrally related to conjugation machines. Mol Microbiol 40: 294-305.

Cianciotto, N.P. (2001) Pathogenicity of Legionella pneumophila. Int J Med Microbiol291:331-343.

Coers, l., Monahan, C., and Roy, c.R. (1999) Modulation of phagosome biogenesis byLegionella pneumophila creates an organelle permissive for intracellular growth. NatCell Bioi 1: 451-453.

Derre, 1., and Isberg, R.R. (2005) LidA, a translocated substrate of the Legionellapneumophila type IV secretion system, interferes with the early secretory pathway.Infect Immun 73: 4370-4380.

Femandez-Moreira, K, Helbig, lH., and Swanson, M.S. (2006) Membrane vesicles shed byLegionella pneumophila inhibit fusion of phagosomes with lysosomes. Infect Immun74: 3285-3295.

Fields, B.S. (1996) The molecular ecology of Legionellae. Trends Microbiol4: 286-290.

Guo, L., Urn, K.B., Poduje, C.M., Daniel, M., Gunn, lS., Hackett, M., and Miller, SJ.(1998) Lipid A acylation and bacterial resistance against vertebrate antimicrobialpeptides. Cell 95: 189-198.

Hammer, B.K., Tateda, E.S., and Swanson, M.S. (2002) A two-component regulator inducesthe transmission phenotype of stationary-phase Legionella pneumophila. MolMicrobiol44: 107-118.

20

Page 22: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 1

Heuner, K, Dietrich, C., Skriwan, C., Steinert, M., and Hacker, J. (2002) Influence of thealternative sigma(28) factor on virulence and flagellum expression of Legionellapneumophila. Infect Immun 70: 1604-1608.

Hilbi, H., Segal, G., and Shuman, H.A. (2001) !cm/dot-dependent upregulation ofphagocytosis by Legionella pneumophila. Mol Microbiol42: 603-617.

Hoffman, P.S., Houston, L., and Butler, C.A. (1990) Legionella pneumophila htpAB heatshock operon: nucleotide sequence and expression of the 60-kilodalton antigen in1. pneumophila-infected HeLa cells. Infect Immun 58: 3380-3387.

Horwitz, M.A. (1984) Phagocytosis of the Legionnaires' disease bacterium (Legionellapneumophila) occurs by a novel mechanism: engulfment within a pseudopod coil.Cell 36: 27-33.

Kagan, J.C., Stein, M.P., Pypaert, M., and Roy, C.R. (2004) Legionella subvert the functionsofRab1 and Sec22b to create a replicative organelle. J Exp Med 199: 1201-1211.

Kim, KJ., Na, YK, and Jeon, KW. (1994) Bacterial endosymbiont-derivedlipopo1ysaccharides and a protein on symbiosome membranes in newly infectedamoebae and their roles in lysosome-symbiosome fusion. Infect Immun 62: 65-71.

Kirby, lE., and Isberg, R.R. (1998) Legionnaires' disease: the pore macrophage and thelegion of terror within. Trends Microbiol6: 256-258.

Kirby, J.E., Vogel, J.P., Andrews, H.L., and Isberg, R.R. (1998) Evidence for pore-formingability by Legionella pneumophila. Mol Microbiol27: 323-336.

Knirel, YA., Rietschel, KT., Marre, R., and Zahringer, D. (1994) The structure of the 0­specific chain ofLegionella pneumophila serogroup 1 lipopolysaccharide. Eur JBiochem 221: 239-245.

Kooistra, 0., Luneberg, E., Lindner, B., Knirel, Y.A., Frosch, M., and Zahringer, D. (2001)Complex a-acetylation in Legionella pneumophila serogroup 1 lipopolysaccharide.Evidence for two genes involved in 8-0-acetylation of legionaminic acid.Biochemistry 40: 7630-7640.

Kooistra, a., Luneberg, E., Knirel, Y.A., Frosch, M., and Zahringer, D. (2002a) N­Methylation in polylegionaminic acid is associated with the phase-variable epitope ofLegionella pneumophila serogroup 1 lipopolysaccharide. Identification of 5-(N,N­dimethylacetimidoyl)amino and 5-acetimidoy1(N-methyl)amino-7-acetamido-3,5,7,9­tetradeoxynon-2-u10sonic acid in the O-chain polysaccharide. Eur J Biochem 269:560-572.

Kooistra, a., Knirel, Y.A., Liineburg, K, Frosch, M., Zlihringer, D. (2002b) Phase variationin Legionella pneumophila serogroup1, subgroup OLDA, strain RC1 influences lipidA structure. In Legionella: Proceedings ofthe 5th international Conference. Marre,R. (ed). Washington, D.C.: ASM Press, pp. 68-73.

Luck, P.C., Freier, T., Steude1, C., Knirel, YA., Luneberg, E., Zahringer, D., and Helbig, J.H.(2001) A point mutation in the active site ofLegionella pneumophila 0­acetyltransferase results in modified lipopolysaccharide but does not influencevirulence. Int J Med Microbiol291: 345-352.

Luneberg, E., Mayer, B., Daryab, N., Kooistra, a., Zahringer, D., Rohde, M., Swanson, J.,and Frosch, M. (2001) Chromosomal insertion and excision of a 30 kb unstable

21

Page 23: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

genetic element is responsible for phase variation of lipopolysaccharide and othervirulence determinants in Legionella pneumophila. Mol Microbiol39: l259-127l.

Luo, Z.Q., and Isberg, R.R. (2004) Multiple substrates of the Legionella pneumophilaDot/Icm system identified by interbacterial protein transfer. Proc Natl Acad Sci USA101: 841-846.

Miller, S.I., Emst, RK., and Bader, M.W. (2005) LPS, TLR4 and infectious disease diversity.Nat Rev Microbiol3: 36-46.

Moll, H., Sonesson, A., Jantzen, E., Marre, R., and Zahringer, D. (1992) Identification of27­oxo-octacosanoic acid and heptacosane-1 ,27-dioic acid in Legionella pneumophila.FEMS Microbiol Left 76: 1-6.

Molmeret, M., and Abu Kwaik, Y. (2002) How does Legionella pneumophila exit the hostcell? Trends Microbiol10: 258-260.

Molofsky, A.B., and Swanson, M.S. (2004) Differentiate to thrive: lessons from theLegionella pneumophila life cycle. Mol Microbiol53: 29-40.

Nagai, H., Kagan, lC., Zhu, X., Kahn, R.A., and Roy, C.R. (2002) A bacterial guaninenucleotide exchange factor activates ARF on Legionella phagosomes. Science 295:679-682.

Neumeister, B., Faigle, M., Sornrner, M., Zahringer, D., Stelter, F., Menzel, R, Schutt, C.,and Northoff, H. (1998) Low endotoxic potential of Legionella pneumophilalipopolysaccharide due to failure of interaction with the monocyte lipopolysaccharidereceptor CD14. Infect Immun 66: 4151-4157.

Raetz, C.R, and Whitfield, C. (2002) Lipopolysaccharide endotoxins. Annu Rev Biochem 71:635-700.

Robey, M., O'Connell, W., and Cianciotto, N.P. (2001) Identification of Legionellapneumophila rep, a pagP-like gene that confers resistance to cationic antimicrobialpeptides and promotes intracellular infection. Infect Immun 69: 4276-4286.

Roy, C.R., Berger, K.H., and Isberg, R.R. (1998) Legionella pneumophila DotA protein isrequired for early phagosome trafficking decisions that occur within minutes ofbacterial uptake. Mol Microbiol28: 663-674.

Roy, c.R., and Tilney, L.G. (2002) The road less traveled: transport of Legionella to theendoplasmic reticulum. JCell Bioi 158: 415-419.

Segal, G., Purcell, M., and Shuman, H.A. (1998) Host cell killing and bacterial conjugationrequire overlapping sets of genes within a 22-kb region of the Legionellapneumophila genome. Proc Natl Acad Sci USA 95: 1669-1674.

Segal, G., and Shuman, H.A. (1998) Intracellular multiplication and human macrophagekilling by Legionella pneumophila are inhibited by conjugal components ofIncQplasmid RSF101O. Mol Microbiol30: 197-208.

Segal, G., Feldman, M., and Zusman, T. (2005) The Icm/Dot type-IV secretion systems ofLegionella pneumophila and Coxiella burnetii. FEMS Microbiol Rev 29: 65-81.

Sonesson, A., Jantzen, E., Bryn, K., Larsson, L., and Eng, l (1989) Chemical composition ofa lipopolysaccharide from Legionella pneumophila. Arch Microbiol153: 72-78.

22

Page 24: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 1

Sonesson, A, Jantzen, E., Bryn, K, Tangen, T., Eng, J., and Zahringer, D. (1994a)Composition of 2,3-dihydroxy fatty acid-containing lipopolysaccharides fromLegionella israelensis, Legionella maceachernii and Legionella micdadei.Microbiology 140: 1261-1271.

Sonesson, A, Jantzen, K, Tangen, T., and Zahringer, D. (1994b) Chemical characterizationof lipopolysaccharides from Legionella feeleii, Legionella hackeliae and Legionellajordanis. Microbiology 140: 2663-2671.

Sonesson, A, Jantzen, E., Tangen, T., and Zahringer, D. (1994c) Chemical composition oflipopolysaccharides from Legionella bozemanii and Legionella longbeachae. ArchMicrobiol162: 215-221.

Sporri, R, Joller, N., Albers, u., Hilbi, H., and Oxenius, A (2006) MyD88-dependent IFN­gamma production by NK cells is key for control of Legionella pneumophilainfection. J Immunol176: 6162-6171.

Steinert, M., Hentschel, D., and Hacker, l (2002) Legionella pneumophila: an aquaticmicrobe goes astray. FEMS Microbiol Rev 26: 149-162.

Stone, B.1., and Abu Kwaik, Y. (1998) Expression of multiple pili by Legionellapneumophila: identification and characterization of a type IV pilin gene and its role inadherence to mammalian and protozoan cells. Infect Immun 66: 1768-1775.

Sturgill-Koszycki, S., and Swanson, M.S. (2000) Legionella pneumophila replicationvacuoles mature into acidic, endocytic organelles. J Exp Med 192: 1261-1272.

Swanson, M.S., and Hammer, B.K. (2000) Legionella pneumophila pathogesesis: a fatefuljourney from amoebae to macrophages. Annu Rev Microbiol54: 567-613.

Sweet, C.R., Ribeiro, AA, and Raetz, C.R. (2004) Oxidation and transamination of the 3"­position ofDDP-N-acetylglucosamine by enzymes from Acidithiobacillusferrooxidans. Role in the formation of lipid a molecules with four amide-linked acylchains. J BioI Chem 279: 25400-25410.

Venkataraman, C., Haack, B.J., Bondada, S., and Abu Kwaik, Y. (1997) Identification ofaGallGalNAc lectin in the protozoan Hartmannella vermiformis as a potential receptorfor attachment and invasion by the Legionnaires' disease bacterium. J Exp Med 186:537-547.

Vogel, lP., Andrews, H.L., Wong, S.K, and Isberg, R.R (1998) Conjugative transfer by thevirulence system of Legionella pneumophila. Science 279: 873-876.

Vorachek-Warren, M.K, Carty, S.M., Lin, S., Cotter, R.J., and Raetz, C.R. (2002) AnEscherichia coli mutant lacking the cold shock-induced palmitoleoyltransferase oflipid A biosynthesis: absence of unsaturated acyl chains and antibiotichypersensitivity at 12 degrees C. J BioI Chem 277: 14186-14193.

Watarai, M., Andrews, H.L., and Isberg, R.R (2001) Formation of a fibrous structure on thesurface of Legionella pneumophila associated with exposure of DotH and DotOproteins after intracellular growth. Mol Microbiol39: 313-329.

Weber, S.S., Ragaz, C., Reus, K, Nyfeler, Y., and Hilbi, H. (2006) Legionella pneumophilaexploits PI(4)P to anchor secreted effector proteins to the replicative vacuole. PLoSPathog 2: e46.

23

Page 25: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General introduction

Wiater, L.A., Dunn, K., Maxfield, F.R., and Shuman, H.A. (1998) Early events in phagosomeestablishment are required for intracellular survival of Legionella pneumophila. InfectImmun 66: 4450-4460.

Zahringer, D., Knirel, Y.A., Lindner, 8., Helbig, J.H., Sonesson, A., Marre, R., and Rietschel,E.T. (1995) The lipopolysaccharide of Legionella pneumophila serogroup 1 (strainPhiladelphia 1): chemical structure and biological significance. Prog Clin Bioi Res392: 113-139.

24

Page 26: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

2 The amoebae plate test implicates a paralog of IpxB inthe interaction of Legionella pneumophila withAcanthamoeba castellanii

Urs Albers!, Katrin Reus!' Howard A. Shuman2 and Hubert Hilbil

IInstitute of Microbiology, Swiss Federal Institute of Technology (ETH), Wolfgang-Pauli Str.

10, HCI G405, 8093 ZUrich, Switzerland and 2Department of Microbiology, College of

Physicians and Surgeons, Columbia University, 701 West 168th Street, New York, NY

10032, USA.

Abbreviations: APT, amoebae plate test; BSA, bovine serum albumin; CYE, charcoal-yeast

extract; FCS, fetal calf serum; icm/dot, intracellular mUltiplication/defective organelle

trafficking; lcs, Legionella cytotoxic suppressor; MOl, multiplicity of infection; PI,

propidium iodide; PYG, proteose-yeast extract-glucose.

Microbiology (151): 167-182 (2005)

Page 27: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

2.1 ABSTRACT

Legionella pneumophila is a bacterial parasite of fresh water amoebae which also grows in

alveolar macrophages and thus causes the potentially fatal pneumonia Legionnaires' disease.

Intracellular growth within amoebae and macrophages is mechanistically similar and requires

the IcmIDot type IV secretion system. Here, we develop an assay, the amoebae plate test

(APT), to analyze growth of1. pneumophila wild-type and icmldot mutant strains spotted on

agar plates in presence of Acanthamoeba castellanii. In the APT, wild-type 1. pneumophila

formed robust colonies even at high dilutions, icmT, -R, -P, or dotB mutants failed to grow,

and icmS or -G mutants were partially growth defective. The icmS or icmG mutant strains

were used to screen an 1. pneumophila chromosomal library for genes that suppress the

growth defect in presence of the amoebae. An icmS suppressor plasmid was isolated that

harbored the icmS and flanking icm genes, indicating that this plasmid complements the

intracellular growth defect of the mutant. Contrarily, different icmG suppressor plasmids

rendered the icmG mutant more cytotoxic for A. castellanii without enhancing intracellular

multiplication in amoebae or RAW264.7 macrophages. Deletion of individual genes in the

suppressor plasmids inserts identified lcs (Legionella cytotoxic suppressor) A, -B, -C and -D

being required for enhanced cytotoxicity of an icmG mutant strain. The corresponding

proteins show sequence similarity to hydrolases, NlpD-related metalloproteases, lipid A

disaccharide synthases, and ABC transporters, respectively. Overexpression of LcsC, a

putative paralog of the lipid A disaccharide synthase LpxB, increased cytotoxicity of an icmG

but not other icmldot or rpoS mutant strains against A. castellanii. Based on sequence

comparison and chromosomal location, IcsB and IcsC likely encode enzymes involved in cell

wall maintenance and peptidoglycan metabolism. The APT established here may prove useful

to identify other bacterial factors relevant for interactions with amoeba hosts.

2.2 INTRODUCTION

The Gram-negative bacterium Legionella pneumophila is a natural parasite of fresh water

amoebae (Rowbotham, 1980; reviewed by Steinert et al., 2002). If inhaled via contaminated

aerosols, the bacteria may grow in human alveolar macrophages and cause a severe

pneumonia known as Legionnaires' disease. Within phagocytic host cells, 1. pneumophila

establishes a pH-neutral vacuole that does not fuse with lysosomes (Horwitz & Maxfield,

26

Page 28: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

1984; Horwitz, 1983a). This unique, replication-permissive vacuole associates with smooth

vesicles, mitochondria and endoplasmic reticulum (ER) (Horwitz, 1983b; Swanson & Isberg,

1995; Tilney et al., 2001) and recruits early secretory vesicles at ER exit sites (Kagan & Roy,

2002; Kagan et al., 2004).

Intracellular growth in macrophages and amoebae, including Acanthamoeba castellanii and

Dictyostelium discoideum, is mechanistically similar and requires the L. pneumophila

Icm/Dot transporter, a type IV secretion apparatus related to conjugation systems (Hagele et

al., 2000; Otto et al., 2004; Segal et al., 1998; Segal & Shuman, 1999b; Solomon et al., 2000;

Vogel et al., 1998). The Icm/Dot secretion system determines the initial contact of

L. pneumophila with host cells and phagosome biogenesis (Hilbi et al., 2001; Watarai et al.,

2001), is required to evade immediate endocytic maturation (Roy et al., 1998; Wiater et al.,

1998) and governs subsequent formation of the ER-derived, replicative vacuole (reviewed by

Nagai & Roy, 2003). Once L. pneumophila resides in this nutritionally rich compartment, the

vacuole may acidify (Sturgill-Koszycki & Swanson, 2000), and bacterial replication

apparently proceeds without requiring a functional Icm/Dot transporter (Coers et al., 1999).

Most of the genes of the km/dot loci are predicted to encode membrane-spanning proteins

(Segal & Shuman, 1999a). Interestingly, the IcmG protein contains a t-SNARE domain, and

thus this membrane protein might play a direct role in altering host cell vesicle trafficking

(Morozova et al., 2004). Biochemical analysis of the soluble Icm proteins revealed that

IcmS/IcmW and IcmRlIcmQ directly bind to each other (Coers et al., 2000). IcmR functions

as a chaperone of IcmQ, preventing and reversing its aggregation into high-molecular-weight

complexes that form pores in lipid membranes (Dumenil & Isberg, 2001; Dumenil et al.,

2004). Intracellular multiplication of and host cell killing by L. pneumophila is inhibited by a

functional plasmid mobilization system, suggesting that the nucleoprotein conjugal substrate

competes with virulence effectors for transport by the Icm/Dot machinery (Segal & Shuman,

1998). Only recently, !cm/Dot-transported proteins have been identified. RalF is translocated

into the host cell and acts as a guanine nucleotide exchange factor that recruits the small

GTPase ARFl to the Legionella phagosome (Nagai et al., 2002). LepA and LepB share

sequence similarity with SNAREs and seem to mediate the Icm/Dot-dependent release of

L. pneumophila-containing vesicles from amoebae (Chen et al., 2004). Other IcmIDot­

translocated proteins are LidA, which by an unknown mechanism contributes to an efficient

formation of the replication vacuole (Conover et al., 2003) and the SidA-H proteins, many of

27

Page 29: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

which comprise families of up to five paralogs (Luo & Isberg, 2004). Secretion into culture

supematants of the polytopic membrane protein DotA requires the IcmIDot secretion system

but neither IcmS nor IcmW, and might lead to the formation of pores in target membranes

(Nagai & Roy, 2001).

The Icm/Dot-dependent establishment of the Legionella vacuole eventually leads to host cell

death due to intracellular replication and lysis. Additionally, the icm/dot genes mediate

cytotoxic events such as (i) contact-dependent immediate cytotoxicity due to pore formation

(Kirby et al., 1998; Zuckman et al., 1999), (ii) induction of apoptosis (Zink et al., 2002), and

(iii) egress of the bacteria from the vacuole of the spent host cell (Molmeret et al., 2002).

Other cytotoxic factors of L. pneumophila include legiolysin (Wintermeyer et al., 1991), the

zinc metalloprotease Msp (Quinn & Tompkins, 1989; Szeto & Shuman, 1990) and RtxA, a

member ofthe RTX (repeats in toxin) family ofcytotoxic adhesins (Cirillo et al., 2001).

In this report, we describe the amoebae plate test (APT), a novel assay to analyze growth of

1. pneumophila wild-type and icm/dot mutants on agar plates in presence of A. castellanii.

The APT was used to screen an 1. pneumophila chromosomal library for multicopy

suppressors of the partial growth defect of icmS or icmG mutant strains. Possible suppressors

include genes that i) enhance intracellular bacterial growth and thus kill the amoebae, ii) are

otherwise cytotoxic for the amoebae, or iii) interfere with phagocytosis of the bacteria by the

amoebae. Among the plasmids isolated, icm/dot region II and cytotoxic genes likely involved

in peptidoglycan metabolism were identified.

2.3 METHODS

2.3.1 Bacterial strains, cell culture and reagents

The bacterial strains and plasmids used in this study are listed in Table 1. E. coli was cultured

on LB agar plates or in LB broth, supplemented with chloramphenicol (Cm, 3O/lg/ml),

sodium ampicillin (Ap, 100 /lg/ml) or kanamycin sulfate (Km, 50 /lg/ml) if required.

1. pneumophila was grown on charcoal-yeast extract (CYE) agar plates (Feeley et al., 1979)

or in ACES-buffered-yeast extract broth (AYE) containing 0.5% bovine serum albumin

(BSA) (Horwitz & Silverstein, 1983). Supplements for 1. pneumophila were used at the

following concentrations: Cm (5 /lg/ml), Km (50 /lg/ml), IPTG (0.5 mM). A. castellanii

(ATCC 30234) was grown in proteose-yeast extract-glucose medium (PYG) at 30°C (Moffat

28

Page 30: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

& Tompkins, 1992; Segal & Shuman, 1999b) and subcultured once or twice a week.

RAW264.7 macrophages (kindly obtained from Dr. David Underhill, University of

Washington, Seattle) were cultivated in RPMI1640 medium supplemented with 10% FCS

and 2 mM L-glutamine at 37°C in 5% CO2• High gel strength agar was from Serva, proteose

peptone from Becton Dickinson Biosciences and Bacto yeast extract from Difco,

respectively. RPMI medium, glutamine and fetal calf serum (FCS) were from Omnilab. All

other reagents were from Sigma.

Table 1: Bacterial strains and plasmids used in this study

Strain or plasmid Relevant propertiest Reference

E. coliLW253TOPI0

L. pneumophilaOS3001OS3002GS3003GS3007GS3011GS3012JR32LELA1275LELA2883LELA3473MW635MW645

Plasmids

MW66

pGS-BCD-05pGS-Lc-34-14pGS-Lc-36bpGS-Lc-37-14pOS-Lc-63-14pLAW344

pMMB207

pMMB207ab

F'F mcrA t1(mrr-hsdRMS-mcrBC)C1>80IacZt1MI5 MacX74 deoR recAlaraD139 t1(ara-leu)7697 galU galKrpsL (Srn) endAl nupG

JR32 icmS3001::KmJR32 icmP3002::KmJR32 icm03003::KmJR32 icmN3007::KmJR32 icmT30ll::KmJR32 icmR30l2::KmSalt-sensitive isolate of AM511JR32 icmF1275::Tn903dIIlacZJR32 dotB2883: :Tn903dIIlacZJR32 icmR3473::Tn903dIIlacZJR32 icmG::KmJR32 icmC::Km

1. pneumophila Philadelphia-lchromosomal library, partial EcoRIfragments (5-10 kb) in pMMB207dotBCD in pMMB207ab-Km-14icmPO in pMMB207ab-Km-14icmR in pMMB207abicmTS in pMMB207ab-Km-14icmGCD in pMMB207-Km-14sacB MCS oriT(RK2) Cm loxPoriR(ColEl) Ap loxPRSFlOlO derivative, IncQ, oriT,lac!\ Ptac, CmpMMB207, lacZa, MCS

Wiater et al., 1994Invitrogen

Segal & Shuman, 1997Segal & Shuman, 1997Segal & Shuman, 1997Segal et al., 1998Segal & Shuman, 1998Hilbi et al., 2001Sadosky et al., 1993Purcell & Shuman, 1998Sadosky et al., 1993Sadosky et al., 1993Purcell & Shuman, 1998Purcell & Shuman, 1998

Purcell & Shuman, 1998

Hilbi et al., 2001Segal & Shuman, 1998Segal & Shuman, 1997Segal & Shuman, 1998Segal et al., 1998Wiater et al., 1994

Morales et al., 1991

Segal & Shuman, 1997

29

Page 31: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

pMMB207ab-Km-14 pMMB207ab with mobA::KmpMMB207-RBS pMMB207 with ribosome binding

site (T7 gene 10)pUA3 pUC19-G54~lcsB

pUA5 pG54-1csB (~NlpD-like protease)pUA9 pUC19-G65McsCpUAll pUCI9-G65~fis

pUA13 pG65:fis (~Fis)

pUA15 pG65-1csC (~LpxB homolog)pUA17 pUCI9-G34~lcsA

pUA19 pG34-1csA (~Hydrolase)

pUA20 pG39-0RFI-3 (HindIII deletion)pUA22 pG54-0RFI-3 (SphI deletion)pUA26 pMMB207-RBSlcsC (pLcsC)pUA27 pUCI9-G66McsDpUA29 pG66-1csD (~ABC transporter)pUA34 pMMB207-RBSlcsA (pLcsA)pUA35 pMMB207-RBSlcsB (PLcsB)pUA36 pMMB207-RBSlcsD (PLcsD)pUA41 pG65-lcsC:fis (~LcsC, ~Fis)

pUA44 pUCI9-G65-lcsC::KmpUA46 pLAW344-G65-lcsC::KmpUC19 oriR(coIEl), MCS, AppUC4K oriR (pBR322), Ap, MCS::Km

;Abbreviations: Ap, ampici11in resistance; Cm, chloramphenicolresistance; Srn, Streptomycin resistance.

2.3.2 Construction of plasmids

Segal & Shuman, 1998This study

This studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyThis studyYanisch-Perron et al., 1985Amershamresistance; Km, kanamycin

Plasmids were isolated using commercially available kits from Qiagen or Macherey-Nagel,

and DNA manipulations were performed according to standard protocols. In frame deletions

of single or several ORFs in the inserts of the suppressor plasmids pG34, pG54, pG65 and

pG66 yielded the plasmids pUA3, pUA9, pUAll, pUA17 and pUA27 (Table 1, Fig. 5a) and

were generated by PCR (Imai et al., 1991) after subcloning the inserts into pUC19 using the

EcoRI restriction sites. To generate the deletion constructs, the oligonuc1eotides listed in

Table 2 were used. The PCR fragments were phosphorylated, circularized, transformed into

E. coli TOPI0, checked by restriction digestion, and the inserts were finally cloned back into

pMMB207 using the EcoRI restriction sites, yielding pUA5, pUA13, pUA15, pUA19 and

pUA29. Alternatively, ORFI-3 was deleted from pG39 or pG54 by digestion with HindIII or

SphI (Fig. 5a), respectively, and re-ligated, thus yielding pUA20 and pUA22. To overexpress

IcsA, -B, -C or -D, the genes were amplified by PCR using pG34, pG54, pG65 or pG66 as

templates and cloned by digestion with EcoRI and BamHI or NdeI into the expression vector

30

Page 32: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

pMMB207-RBS, yielding pMMB207-RBSlcsA-D. pMMB207-RBS harbors the Ptac promoter

and the ribosome binding site from the T7 gene 10 taken from pGS-GFP-04 (Hilbi et al.,

2001). The sequence ofall PCR fragments was confirmed by sequencing.

Table 2: Oligonucleotides used in this study

Oligo Sequence Comment

3'-lcsD5'-lcsC5' pG66-McsD3' pG66-~lcsD

3'-lcsA5'-lcsB3'-lcsB5'-lcsD

5'ofMCS3' ofMCS5' KmR (out)3' KmR (out)5' pG54-McsB3' pG54-McsB5' pG65-~lcsC3' pG65-~lcsC5' pG65-~fis3' pG65-~fis5'-lcsC

GAATATGGCTCATAACACCCCTCACTTTCTGGCTGGATGATGGTCCAGCTTTTGCCTCAAAGGCTCCAAAGCGGTCAATCCGTTAGACTCCACCAGATTCTTCGCCTGTCCCAACTGGCAAAGTTAAAGGAATCTCCGTAGCTTGTGTATCAATGACATTCTCCCGGTACCAAATATTTTCGCTTAACGATCGAATTCTTTAAGAAGGAGATATACATATGCCGAATGCTAGTCGTGTTGATCGGATCCCTATGCATTTTTTTCAGGCAATTCTGC 3'-lcsCTCCTTGTTTGTCTTGATTGAGATGC 5' pG34-McsATCCTTCATTAGAGGATAAAAGAGCTG 3' pG34-McsAGATCGTACCATATGTTTCTTGTTAGCATCTCAATCAAG 5'-lcsAACGATCGGATCCTACTCAGCTCTTTTATCCTCTAATGGATCGTACCATATGCGCATCCCTAGCCTATCGATCGGATCCTTAAGACAACCAGTCTAACGGATTGGATCGTACCATATGGCAAATTTATTTAGTTTTTTGTTTCTTGGAACGATCGGATCCCTACTGATTTAGTTCATATCTACGAGCTAGCGTGCCGCTTGTTACTCCAGCCCAGTGTTCCAAGTCCGTGCTCGTAGATATGAAC

oUA28oUA29oUA30oUA31

pMMB207-5' ATTGTGTGGAATTGTGAGCGpMMB207-3' CTTCTCTCATCCGCCAAAACoKanStartoKanStopoUA3oUA4oUA5oUA6oUA7oUA8oUA19

oUA20oUA21oUA22oUA23

oUA24oUA25oUA26oUA27

2.3.3 The amoebae plate test

In the amoebae plate test (APT), A. castellanii is spread on CYE agar plates prior to spotting

bacteria in serial dilutions onto these plates. Thus, the APT allows determining growth of

bacteria on solid medium in presence of amoebae. A. castellanii cultures were fed with PYG

2 days prior to an experiment. One day before the experiment, the medium was exchanged,

and the amoebae were tapped off the tissue culture flask, spun down (5 min, 1500 rpm) and

resuspended in PYG (2.67 x 106/ml). 1.5 ml of suspended amoebae (or PYG only) were

applied on a CYE/Cm agar plate, let dry for 1-2 h in a laminar flow hood and left at room

31

Page 33: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

temperature over night. Wild-type and icm/dot mutant L pneumophila strains used for the

APT harbored the plasmid pMMB207ab, pMMB207ab-Km14 or the corresponding

complementing plasmids listed in Table 1. For the APT, stationary phase bacterial cultures

(OD6oo > 4.5) were adjusted to an identical OD600, and series of tenfold dilutions in sterile

H20 were prepared. 3 /-11 of stationary culture (approx. 108 bacteria) or 3 /-11 of each dilution

were spotted onto the CYE/Cm plates and incubated for 5-7 days at 30°C or 37°C.

2.3.4 Screening of an L. pneumophila chromosomal library for icmldotsuppressors using the APT

To perform a suppressor screen with the APT, the partially growth defective icmS and icmG

mutants were chosen as recipients for the L. pneumophila genomic library MW66 (Purcell &

Shuman, 1998). MW66 harbors 5-lOkb EcoRI fragments in the vector pMMB207. The

library was amplified in the E. coli host strain DH5a, isolated and electroporated into the

conjugation-competent E. coli strain LW253. Mating of MW66 into the L. pneumophila icm

mutant strains was done as described (Mintz & Shuman, 1987; Segal & Shuman, 1998).

Briefly, LW253/MW66 grown on LB/Cm plates was suspended in LB, spun down and

resuspended in M63 medium. Stationary phase icmS and icmG mutants grown in liquid

culture were washed and resuspended in M63 medium. Donor and recipient strains were

incubated at a ratio of 1110 on CYE plates (4 h, 37°C), resuspended in 0.2 ml M63 medium,

streaked on CYE/CmlKm plates and incubated for 4-5 days at 37°C. The icmS and icmG

mutants harboring the MW66 library were then suspended in AYE medium, spotted in serial

dilutions onto CYE/Cm plates containing 4 x 106 A. castellanii and incubated at 30°C. JR32

and the icmS and icmG mutants harboring pMMB207ab were used for comparison,

respectively. Within 7-14 days several colonies of icm mutants harboring library plasmids

appeared at dilutions where no parental icm mutant strains were detected. The APT-selected

suppressor strains were grown to stationary phase for 24 h in AYE/Cm broth and spotted

again onto CYE/Cm plates containing 4 x 106 A. castellanii. The MW66 library plasmids of

suppressor strains recovered from the second round of selection by APT were analyzed

further. In preliminary experiments, the icmS and icmG mutants harboring the MW66 library

were spread at an MOl of 1 simultaneously with 5 x 106 A. castellanWml PYG on CYE agar

plates and incubated at 30°C. Colonies that appeared using this approach were subjected to a

second round of selection as described above.

32

Page 34: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

2.3.5 Analysis of suppressor plasmids

The library plasmids of the suppressor strains were isolated, amplified in E. coli and grouped

according to their EcoRI restriction pattern. A representative of each group was

electroporated into the icmG or icmS mutant strain which it was isolated from and analyzed

again by the APT to exclude effects of chromosomal mutations. The suppressor plasmid

inserts were partially sequenced using primers complementary to vector sequences at the 5'

and 3' of the insert. The sequences obtained were mapped in the L. pneumophila genome

(Chien et al., 2004); http://genome3.cpmc.columbia.edu/~legionl) and analyzed for homologs

of the ORFs identified.

2.3.6 Intracellular growth in A. castellanii and RAW264.7 macrophages

For intracellular growth assays, A. castellanii (5 x 104/well) or RAW264.7 macrophages (2 x

104/well) were seeded onto a 96 well plate and let adhere for 3 h or over night, respectively.

The phagocytes were infected with L. pneumophila grown to stationary phase in AYE

medium for 24 h (MOl 1, 880 x g), and incubated at 30°C (A. castellanii) or 37°C

(RAW264.7). Intracellular growth was quantified by plating appropriately diluted supematant

of the infected host cells on CYE agar plates at the time points indicated. RAW264.7

macrophages are derived from Balb/c mice, which are considered less susceptible to

L. pneumophila than AlJ mice, the established murine model of Legionnaires' disease.

L. pneumophila does not replicate in peritoneal macrophages elicited from Balb/c mice

(Yamamoto et al., 1988). Yet bone marrow-derived macrophages from Balb/c mice permit

the intracellular replication of L. pneumophila (Wright et al., 2003), which corresponds to the

observed permissiveness of RAW264.7 macrophages.

2.3.7 Cytotoxicity assay

To determine cytotoxicity, 4 x 104 A. castellanii per well were seeded in PYG onto a 24 well

plate the day prior to infection. On the day of infection, the PYG was replaced with Ac buffer

(Moffat & Tompkins, 1992). Bacteria from 3 or 4 days old plates were resuspended and

diluted in sterile water to infect the amoebae at an MOl of 50 or 500. After spinning down the

bacteria (880 x g, 5 min), the plates were incubated at 30°C. Two days post infection,

33

Page 35: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

propidium iodide (PI) solution was added to the wells at a final concentration of 1 Ilg/ml.

After several minutes incubation, the amoebae were viewed in brightfield or by

epifluorescence with an inverse microscope (Zeiss Axiovert 200M, 20 x objective). The

percentage of dead (PI-positive) amoebae was determined by counting the number of total

and fluorescent amoebae.

2.3.8 Computational and statistical analysis

Translated nucleic acid data bases or the conserved domain data base (Marchler-Bauer et al.,

2003) were searched using the TBLASTX or RPSBLAST algorithms, respectively (Altschul

et al., 1997). The eOG data base is maintained by Tatusov et al. (Tatusov et al., 2001).

Multiple amino acid sequence alignments were created with the CLUSTALW algorithm.

Statistical analysis was done using the Mann-Whitney test taking values of < 0.05 as

significant.

2.4 RESULTS

2.4.1 Growth of L. pneumophila on CYE agar plates in presence ofA. castellanii - the amoeba plate test.

Intracellular growth of L. pneumophila within A. castellanii requires the icm/dot genes (Segal

& Shuman, 1999b). Here, we establish a novel assay, the amoebae plate test (APT) to analyze

growth of wild-type and icm/dot mutant L. pneumophila on eYE agar plates in presence of

A. castellanii. Stationary phase cultures of L. pneumophila wild-type strain JR32 and icm/dot

mutants harboring empty or complementing plasmids were spotted in tenfold serial dilutions

onto agar plates in presence or, as a control, absence of amoebae (Fig. 1). Within 5-7 days at

30°C, wild-type L. pneumophila strain JR32 formed robust colonies even at a 10-5 dilution

(MOl approximately 0.2). Under these conditions, the icmT, -R, -0, -P, -C and dotB mutant

strains did not grow at all. The icmS and -G mutant strains showed a partial growth defect in

presence of A. castellanii and eventually were lysed by the amoebae. An icmN mutant

initially formed colonies on amoebae plates similar to wild-type L. pneumophila (data not

shown). However, whereas JR32 survived on CYE agar plates in the presence ofthe amoebae

for a prolonged time at room temperature, the icmN mutant was lysed by 14 days. Supplying

the corresponding genes on a plasmid restored growth of the icm mutants almost to wild-type

34

Page 36: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2-_.,-------_.,_._---" _----------------_ __ , ,_ __--~--

level (icmT, -8, -P, -0, -R, dotB) or partially (icmC, -C). In absence of amoebae, all strains

grew equally well on CYE agar plates.

At 37°C, the icm8 mutant grew similar to wild-type and the icmN mutant in the APT. At this

temperature, the icmT mutant did not grow at all and the icmR, -P, -0, -C, -C mutants grew

only at the highest concentration. The degree of complementation of individual icm mutants

was the same as at 30°C (data not shown).

~~~

wild-type/pMMB

icmT-/pMMB

icmT-/picmT

icmS -/pMMB

icmS -/picmS

icmR-/pMMB

icmR -/picmR

icmP -/pMMB

icmP -/picmP

icmG -/pMMB

icmG -/picmG

datB -/pMMB

datB -/pdatB

+ Acanthamaeba - Acanthamaeba

Figure 1: Growth of wild-type L. pneumopltila and representative km/dot mutants in the amoebaeplate test (APT). 1. pneumophila wild-type strain JR32 and icmT, -S, -R, -P, -G and dotE mutantsharboring empty (pMMB) or corresponding complementing plasmids (picmT, -S, -R, -P, -G, dotE) werespotted in tenfold serial dilutions onto eYE agar plates in presence or absence of A. castellanii andincubated for 5 days at 300 e as described in Methods. The experiments were done at least three timesand results similar to those shown were obtained.

35

Page 37: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

2.4.2 Isolation of L. pneumophila icmldot suppressor strains using theamoebae plate test.

Among the km/dot mutant strains tested, only the icmS and the -G mutants showed a partial

and not complete growth defect in the APT at 30°C. A partial growth defect for the icmS and

-G mutant strains was shown before using HL-60 macrophages as host cells (Purcell &

Shuman, 1998; Segal & Shuman, 1997; Segal & Shuman, 1999b). We reasoned that this

phenotype would increase chances to identify in a multicopy suppressor screen genes that

increase cytotoxicity or enhance intracellular growth of the mutants. Therefore, the icmS, and

-G mutants were chosen as recipients for the L. pneumophila genomic library MW66 (Purcell

& Shuman, 1998). The library MW66 was moved by electroporation from E. coli DH5a into

the conjugation-competent E. coli strain LW253 and mated into the L. pneumophila icmS,

and -G mutants. The icmS and -G mutants harboring the library MW66 were then spotted in

serial dilutions onto CYE plates containing chloramphenicol (Cm) and A. castellanii. The

plates were incubated at 30°C or 37°C, respectively.

At 37°C, the selection was not promising, since at the same dilutions similar numbers of icm

mutants harboring the library and parental mutants were obtained. At 30°C, however, several

colonies of icm mutant strains harboring MW66 appeared within 7-14 d at dilutions where no

parental icm mutants were detected. These APT-selected strains were grown to stationary

phase for 24 h in AYE/Cm broth and spotted in serial dilutions onto CYE/Cm/A. castellanii

plates again. In the second round of selection, 54 icmG and 2 icmS suppressor strains were

recovered, representatives of which are shown in Fig. 2a. The suppressor strains (icmS ­

/pS37, icmG -/pG44-87) grow at 1O-100-fold higher dilutions than the corresponding icm

mutant strains harboring the empty library plasmid pMMB207ab (icmS -/pMMB, icmG ­

/pMMB), but not as vigorously as wild-type L. pneumophila (JR32/pMMB) or complemented

icm mutants (icmS )picmS, icmG -/picmG). In absence of the amoebae, all strains grew

equally well (data not shown).

The library plasmids of the suppressor strains were isolated, amplified in E. coli and grouped

according to their EcoRI restriction pattern into 18 groups (l icmS -, 17 icmG - background;

data not shown). A representative plasmid of each group was electroporated into the icmG or

-S mutant strain which it was isolated from and analyzed by APT. The strains thus obtained

grew better in the APT than the parental icm mutant harboring an empty library plasmid,

indicating that suppression of the growth defect in presence of amoebae was caused by the

36

Page 38: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

__________________""""""",,_,,_, ,, C_hap!~E~

library plasmid and not due to a second site mutation m the bacterial chromosome.

Representative strains belonging to different EcoRl restriction groups are shown in Fig. 2b.

Retransformation of an fernS mutant strain with plasmid pS37 led to growth in the APT

similar to wild~type L. pneumophila, consistent with the finding that pS37 complements the

fernS mutant strain (see below).

(a)wild-type/pMMB

icmS -/pMMB

icmS -/picmS

icmS -/pS37

icmG -/pMMB

icmG -/picmG

icmG -/pG44

icmG -/pG55

icmG -/pG67

icmG -/pG45

icmG -/pG50

icmG -/pG51

icmG -/pG53

icmG -/pG54

icmG -/pG63

icmG -/pG76

icmG -/pG87

(b)

wild-type/pMMB

icmS -/pMMB

icmS -/pS37

wild-type/pMMB

icmG -/picmG

icmG -/pMMB

icmG -/pG34

icmG -/pG54

IcmG -/pG65

Figure 2: Suppressor strains isolated by APT, (a) L. pneumophila iemS and iemG suppressorstrains harboring Legionella chromosomal library plasmids (pS37, pG44 - pG87) were isolated byAPT, grown to stationary phase and re-tested by APT. Representative suppressor strains are shown incomparison with wild~type, iemG and iemS mutant strains harboring the empty library plasmid(pMMB) and corresponding complemented strains (piemG, piemS). The APT was repeated twice withstrains isolated in a first round of selection, and similar results were obtained. (b) Growth defectsuppression in an APT by L. pneumophila iemS or iemG mutants transformed freshly with isolatedsuppressor plasmids (pS37, pG34, pG54, pG65).

37

Page 39: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

2.4.3 Analysis of suppressor plasmid inserts.

The inserts from representative suppressor plasmids were partially sequenced and mapped in

the 1. pneumophila strain Philadelphia genome (http://genome3.cpmc.columbia

.edu/~legion/). Some of the inserts were identical (pS36/pS37; pG54/pG58/pG63;

pG65/pG72; pG7l/pG79/pG83), and the inserts of the suppressor plasmids pG47 and

pG61 were independently selected twice, since they covered the same loci in the

1. pneumophila genome but were of different length (1.8 kb or 2.5 kb, respectively). The

inserts of the suppressor plasmids were analyzed by BLASTX searches for homo10gs to open

reading frames (ORFs), and thus, 12 different inserts were identified (pS37, pG34, pG39,

pG4l, pG47, pG53, pG54, pG65, pG66, pG67, pG71, and pG78), each harboring 1-10 ORFs

(Table 3).

The insert of the icmS suppressor plasmid pS37 contains a 9.0 kb fragment of icm/dot region

n. The fragment spans a region between 1.7 kb upstream of the icmT and the icmL gene and

includes the icmS gene. This result indicates that pS37 complements the icmS mutant and

thus, increased growth of the IcmS/pS37 strain in the APT is due to intracellular growth

within and killing of the amoebae. Contrarily, the ORFs identified in the inserts of the 11

different icmG suppressor plasmids did not immediately suggest a mechanism accounting for

their growth defect suppression.

Table 3: Suppressor plasmids identified in this study

Plas- Insert ORFst Similarity (E-value§) Accession Toxicmid (kb) (bp) number gene'\!

pS37 9.0 10 Hypothetical protein-icmTSRQPONML T18328ORFs !cm/Dot region n (0.0)

pG34 2.4 > 257 Hypothetical protein (4e-l 0) NP 799916455 Hypothetical protein (8e-1 0) NP 635420821 Hydrolase (5e-6l) ZP 00011307 IcsA

(lpg0163)pG39 3.4 > 1018 Hypothetical protein (5e-11) NP 819452

257, None290 None

>902 Lytic murein transglycosylase (3e-21) ZP 00129242pG41 3.4 >414 dCTP deaminase (2e-64) NP 298052

365 Integration host factor (6e-27) NP 231548731 Histone methyltransferase (7e-7) NP 740808761 None

> 367 None

38

Page 40: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

pG47 1.8 > 352864

> 278

NoneLuxR family transcription factor (5e-3)None

NP 230922

pG53pG54

pG65

pG66

1.43.8

1.9

2.9

> 1403>44631115471169

> 2811160

317> 1269

614956

Histidine ammonia lyase (le-142)Phospholipase C (6e-83)NonePhosphoglycerate mutase (le-154)NlpD-related membrane metallo­protease, M23/M37 family (ge-39)OxidoreductaseLipid A disaccharide synthase (ge-61)

Factor for inversion stimulation (3e-14)Carbamoyl phosphate synthase (le-28)Putative esterase (3e-14)ABC transporter (1 e-13)

NP 230847AAM73854

ZP 00141602NP 799213

NP 819651NP 819650

NP 716031NP 782304ZP 00133753NP 532754

lcsB(lpg0500)

lcsC(lpg1371)

lcsD(lpg0068)

pG67 6.1 > 1687 None2588 Heat shock proteinClpB (0.0) NP 2891471019 Signal peptide peptidase SppA (6e-69) NP 793611> 596 Inositol monophosphatase (2e-61) NP 820132

pG71 5.0 > 1634 Isovaleryl-CoA dehydrogenase (le-81) NP 4880981442 Propionyl-CoA carboxylase (6e-69) NP 4379871070 3-oxoacyl-(ACP) synthase III (ge-18) NP 687378> 716 Acyl-CoA synthetase (2e-40) ZP 00110863

pG78 4.0 1418 Hypothetical protein (le-61), NP 484368785 Hypothetical protein (3e-30) NP 4843701349 Kynurenine monooxygenase (2e-72) NP 636923

fA minimum ORF size of 100 amino acids was chosen. Truncated ORFs are indicated by">".§E-values are indicated for each translated ORF identified in the suppressor plasmid insert.'Cytotoxicity for A. castellanii of the plasmids and genes was determined with an icmG mutant strainharboring either the suppressor plasmid, deletion constructs derived thereof, or an expression plasmid.The gene number corresponds to the annotated L. pneumophila genome.

The suppressor strains might grow better on agar plates in presence of amoebae because of

more efficient intracellular replication and host cell killing or due to a cytotoxic activity not

related to intracellular replication. To test these possibilities, intracellular growth of the icmG

suppressor strains within A. castellanii or murine RAW264.7 macrophages and cytotoxicity

towards amoebae was determined.

39

Page 41: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

2.4.4 Intracellular growth oficmG suppressor strains in A. castellaniiand macrophages

Intracellular growth of the icmG suppressor strains was assayed by quantifying bacteria

released into the supematant from A. castellanii infected with bacteria at an MOl of 1. To

match the conditions used for the APT, the infected amoebae were incubated for 4 days at

30°C, rather than at 37°C described previously (Segal & Shuman, 1999b). At 30°C, the

number of wild-type 1. pneumophila or icmG mutant bacteria harboring the empty library

plasmid increased within 3 days about 4 or 1 orders of magnitude, respectively (Fig. 3a).

Supplying the icmG gene on a plasmid only partially complemented the intracellular growth

defect, as was observed previously (Segal & Shuman, 1999b). None of the 11 icmG

suppressor strains (harboring pG34 - pG78) grew better than the icmG mutant, ruling out that

the growth defect suppression observed in the APT was due to enhanced intracellular growth.

Rather, we noted that the suppressor strains tended to grow even worse than the icmG mutant

strain. We also performed an intracellular growth assay with a high MOl of 500. Under these

conditions, wild-type 1. pneumophila grew 2 orders of magnitude within 3 days (data not

shown). In this experiment, the relative growth of the wild-type and icmG mutant strains was

the same as above, and the 11 icmG suppressor strains tested also grew worse than the icmG

mutant. At 37°C, the suppressor strains icmG -/pG54 or icmG -/pG65 did also not grow better

in amoebae (MOl of 1) than an icmG mutant strain harboring the empty library plasmid (data

not shown).

To analyze intracellular multiplication of the 11 icmG suppressor strains in macrophages, the

murine macrophage cell line RAW264.7 was used. Within 4 days, the number of wild-type or

icmG mutant bacteria released into the supematant increased 4 or 3 orders of magnitude,

respectively (Fig. 3b). Supplying the icmG gene on a plasmid complemented growth of the

icmG mutant almost to wild-type level. In RAW264.7 macrophages, the icmG suppressor

strains (icmG -/pG34 - pG78) grew worse than the icmG mutant strain, similar to what was

observed for growth of these strains in A. castellanii. Taken together, growth defect

suppression of the icmG suppressor strains is not due to enhanced intracellular growth within

phagocytes.

40

Page 42: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

(a) 5

-- 4~::::J....

3(,)

0...CJ0::::. 2ca.;:.s(,)ca 1to

0

0 0.5 1 1.5 2 2.5 3 3.5

Time (d)

5(b)

- 4-5-.2 3(,)

0...CJ0::::. 2ca';:.s(,)ca 1to

0 ----- ------- r-

0 1 2 3 4

Time (d)

Figure 3: Intracellular multiplication of icmG suppressor strains within A. castel/anii andmacrophages. Intracellular growth of the icmG suppressor strains was assayed using (a) A. castel/aniior (b) murine RAW264.7 macrophages as host cells. The phagocytes were infected at an MOl of 1,and bacteria released into the supematant were quantified at the time indicated. Growth of suppressorstrains icmG -/pG34 - pG78 (0), an icmG mutant harboring the empty library plasmid (~), or plasmid­encoded icmG (A) and wild-type strain JR32 (.) is shown. The experiments were repeated at leasttwo times at two different MOIs each, and similar results were obtained.

41

Page 43: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification ofvirulence related genes using the amoebae plate test

2.4.5 icmG suppressor strains are cytotoxic for A. castellanii

Since compared to the parental icmG mutant strain, the icmG suppressor strains did not grow

better in amoebae or macrophages, we tested whether the suppressor plasmids pG34 - pG78

confer increased cytotoxicity to icmG mutants. A. castellanii was infected with wild-type

1. pneumophila, an icmG mutant harboring the empty library plasmid, a complemented icmG

mutant, or the icmG suppressor strains (harboring pG34 - G78). Cytotoxicity was determined

by propidium iodide (PI) uptake 2 days post infection. Under the conditions described, the

amoebae infected with either wild-type 1. pneumophila or the complemented icmG mutant

rounded up and were all dead as judged by their complete disintegration or by PI uptake

(Fig. 4a). In contrast, amoebae infected with an icmT mutant harboring the vector control all

survived and remained spread out and firmly attached. Amoebae infected with an icmG

mutant harboring the vector control rounded up, but only about 15% stained with PI. Most of

the suppressor strains were found to be significantly more cytotoxic than the icmG mutant

strain, thus providing a rationale for their isolation in the suppressor screen (Fig. 4b).

Notably, icmG mutants harboring pG34, pG39, pG41, pG54, or pG66 showed an increased

cytotoxicity in 4-5 out of 5 independent experiments. The suppressor strains icmG -;pG47,

/pG53, /pG65 and /pG71 were more cytotoxic than the icmG mutant in some experiments,

and icmG "/pG67 and /pG78 did not show increased cytotoxicity in any of the experiments

performed. At 37°C, the suppressor strains icmG -/pG54 or icmG -;pG65 were cytotoxic for

amoebae to the same extent as at 30°C (data not shown).

42

Page 44: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

(a)

wild-type/pMMB

icmr-/pMMB

icmG -/pMMB

icmG -/pG54

icmG -/pG65

Brightfield PI Merge

43

Page 45: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

(b) 100 -

- 90~0-'- 80'-r::oS 70......eCl) 60~

50oci

CD 40>E 30en0c. 20

c:: 10 -

Fig. 4. Suppressor strains are cytotoxic for A. castellanii. (a) A. castellanii was infected with wild­type L. pneumophila, icmT or icmG mutant strains harboring the empty library plasmid (pMMB) orwith suppressor strains (icmG -/pG34 - pG78). Cytotoxicity was detennined by uptake of propidiumiodide (PI) 2 days post infection as described in Methods. (b) The data shown are the means andstandard deviations of the percentage of PI-positive A. castellanii counted in 3 different wells on a 24­well plate and are representative of at least five independent experiments.

2.4.6 Identification of cytotoxic suppressor genes by deletion analysis

To identify cytotoxic suppressor genes, individual or several ORFs were deleted in the

suppressor plasmid inserts, and cytotoxicity was assessed by the PI uptake assay. For the

deletion analysis, we focused on the cytotoxic suppressor plasmids pG34, pG39, pG41,

pG47, pG54, pG65 and pG66, and only complete ORFs were considered. The suppressor

plasmid pG41 was not studied further since the encoded DNA-interacting proteins

(integration host factor, histone methyltransferase) presumably increase cytotoxicity

unspecifically. Deletion of the LuxR-type transcription factor in pG47 did not decrease

cytotoxicity (data not shown), and thus, this plasmid was not analyzed further. Deletion of all

the complete ORFs in the insert of pG39 did also not decrease cytotoxicity, but left the

fragment of an ORF encoding the C-tenninus of a putative lytic murein transglycosylase

44

Page 46: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

(data not shown; see Discussion). These findings and considerations left the cytotoxic

suppressor plasmids pG34, pG54, pG65 and pG66 to be analyzed in detail.

The inserts of the plasmids pG34, pG54, pG65 and pG66 harbor homologs of hydrolases,

NlpD-like metalloproteases, lipid A disaccharide synthases, and ABC transporters,

respectively (Table 3, Fig. 5a). Deletion of the genes encoding the hydrolase, NlpD-like

metalloprotease, lipid A disaccharide synthase, or ABC transporter by a PCR-based method

or by restriction enzyme digestion reproducibly reduced cytotoxicity, indicating that the

corresponding proteins are required for the enhanced cytotoxicity of the suppressor plasmids

(Fig. 5b). The genes were termed lcs (Legionella cytotoxic suppressors) A (hydrolase), -B

(NlpD-like metalloprotease), -C (lipid A disaccharide synthase homolog), and -D (ABC

transporter). Deletion of ORFl-3 in the insert of pG54 also decreased cytotoxicity,

presumably because lcsB forms an operon with ORF3 and consequently will not be expressed

in the pG54-0RFl-3 deletion mutant. Deletion of the DNA-binding regulator protein Fis

(factor of inversion stimulation) substantially increased cytotoxicity of plasmid pG65-fis

carrying lcsC as the only remaining complete ORF (Fig. 5b) and, as expected, deletion of

lcsC and fis in the insert of suppressor plasmid pG65 abolished cytotoxicity (data not shown).

(a)

I I pG34 (2.4 kb)

ORF-1 ORf-2 lesA

I • pG54 (3.8 kb)

ORF-1 ORF-2 ORF-3 lesS

c:::::::::J

pG65 (1.9 kb)

ORF-1 lesC fis

pG66 (2.9 kb)

ORF-1

1000I

ORF-2

2000I

lesD

3000I bp

45

Page 47: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

(b)100-:::e 900-....80....

t:.! 70-.eCl) 60C'au

50Ol:(Q) 40>E 30UJ0 20Q.

- 10a.

Figure 5: Identification of cytotoxic suppressor genes by deletion analysis. (a) Overview of thedeletions (horizontal bars) generated in suppressor plasmid pG34 (lcsA), pG54 (lcsB, ORFl-3), pG65(lcsC, fis) and pG66 (lcsD). (b) A. castellanii was infected with icmT or icmG mutant strainsharboring the empty library plasmid (pMMB), complemented icmG (icmG -/picmG), suppressorstrains (icmG -/pG34, IpG54, IpG65, IpG66), or with the deletion strains indicated. Cytotoxicity wasdetermined by uptake of propidium iodide (PI) 2 days post infection. The data shown are the meansand standard deviations of the percentage of PI-positive A. castellanii counted in 3 different wells on a24-well plate and representative of at least three independent experiments.

2.4.7 Overexpression of suppressor genes

Prompted by the finding that strain icmG -/pG65-fis showed enhanced cytotoxicity compared

to icmG -/pG65, we cloned lcsA, -B, -C, and -D into the expression vector pMMB207-RBS,

overexpressed the proteins under the control of the Ptac promoter, and quantified cytotoxicity

of icmG mutant strains harboring these plasmids by PI uptake. Interestingly, the strain icmG .

/pLcsC, overexpressing the LpxB homolog, was 9 or 4 times more cytotoxic than icmG ­

IpMMB or icmG -/pG65, respectively (Fig. 6a, b), suggesting that LcsC-induced cytotoxicity

might be dose-dependent. The icmG -/pLcsC strain did not grow more efficiently in

A. castellanii (30°C, 37°C) or in RAW264.7 macrophages compared to icmG -/pMMB (data

not shown). Contrarily, icmG mutants expressing the putative hydrolase LcsA, NlpD-like

metalloprotease LcsB or the ABC transporter LcsD were less cytotoxic than the icmG mutant

46

Page 48: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

strain. We noted, however, that induction of LcsB prevented growth of the bacteria, and

induction of LcsC and LcsD resulted in fewer and smaller bacterial colonies, indicating that

overexpression of these proteins is toxic for 1. pneumophila (data not shown). In agreement

with this assumption, the strains icmG) pLcsA-D grew well on agar plates, if IPTG was

omitted, and therefore, the genes were expressed only at low levels from the Ptac promotor.

Under these conditions, strain icmG-/ pLcsC was consistently more cytotoxic than the

parental suppressor strain icmG-/ pG65 in three independent experiments (data not shown).

Strain icmG-/ pLcsB was more cytotoxic than the negative control icmG-/ pMMB207 but not

as cytotoxic as the parental suppressor strain icmG-/ pG54. Finally, the strains icmG-/ pLcsA

and icmG"/ pLcsD were not cytotoxic for the amoebae (data not shown).

In presence of IPTG LcsC was cytotoxic for A. castellanii only if overexpressed in an icmG

but not in an icmS, -F or rpoS mutant background (Fig. 6c), even though the icmF as well as

the rpoS mutant strains harboring the empty plasmid pMMB207 were cytotoxic for the

amoebae to a similar extent as the icmG mutant. Equal to the icmS mutant strain, the icmC, ­

E, -0, -R, -T, -W, or dotB mutants overexpressing LcsC were not cytotoxic for amoebae (data

not shown). Moreover, about 30% of A. castellanii infected with wild-type 1. pneumophila

(MOl of 5) harboring either an empty plasmid or pLcsC were killed within 2 days, and thus,

LcsC apparently did not increase cytotoxicity of wild-type 1. pneumophila against the

amoebae (data not shown).

47

Page 49: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

______Id_·er:!}fication ofvirulenceE~lated genes usin'!;L~he amoebae plate test

(a)

icmG -/pMM8

icmG -/pLcsC

(b) 100

- 90~Q-..... 80.....t:~ 70

.! 60Cl)(0u 50.

oq;Cl) 40>:E 30tn0Q. 20

-Q. 10

Brightfield PI Merge

48

Page 50: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

(c)

-~0-..........r:::ca:::~Cl)ca(,)

•q:G)

>Etn0C.

-a.

50

40-

30 -

20

10

Figure 6: Overexpression of LcsC is cytotoxic for A. castellanii in an icmG but not in other icmor rpoS mutants. A. castellanii was infected with the icm (a-c) or rpaS (c) mutant strains indicatedharboring the empty library plasmid (pMMB), complementing plasmid (picmG), suppressor plasmids(pG34, pG54, pG65, pG66), or expression plasmids (pLcsA-D). Cytotoxicity was determined byuptake of propidium iodide (PI) 2 days post infection. The data shown are the means and standarddeviations of the percentage of PI-positive A. castellanii counted in 3 different wells on a 24-wellplate and representative of at least three independent experiments.

2.5 DISCUSSION

2.5.1 Screen for icm suppressors using the amoebae plate test

In this manuscript we describe a novel assay, the amoebae plate test (APT), to determine

growth of bacteria on CYE agar plates in presence of amoebae. Wild-type L. pneumophila

replicates within and kills amoebae, and thus grows on solid medium. Contrarily, km/dot

mutant strains are defective for intracellular growth, killed by the amoebae and do not form

colonies on agar plates (Fig. 1). The APT was routinely done at 30°C, allowing the amoebae

49

Page 51: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

to feed on the icm/dot mutant strains, as judged from the observation that most mutants did

not form colonies under these conditions. At 37°C, the icmS and other icm/dot mutants grew

better than at 30°C in presence of amoebae. Possible explanations for this finding include that

(i) the bacteria grow faster on agar plates at 37°C and thus gain an advantage over the

amoebae in the putative competition between growth on solid medium and being

phagocytosed, or (ii) the amoebae are stressed at a temperature far from environmental

conditions, and therefore, are less phagocytic or less bactericidal.

In screens using the partially growth defective icmS or icmG mutants, we identified icm/dot

region II complementing the icmS mutant and isolated several plasmids that conferred

increased cytotoxicity to an icmG mutant, without alleviating its intracellular growth defect

(Fig. 3, 4). Some cytotoxic icmG suppressor genes were identified by deletion analysis (lcsA­

D, Legionella cytotoxic suppressor) and encode a putative hydrolase, an NlpD-like

metalloprotease, a lipid A disaccharide synthase homolog and an ABC transporter,

respectively (Fig. 5, Table 3). To obtain clues about the cellular pathways which these genes

participate in, we inspected their vicinity in the L. pneumophila genome and searched the

conserved domain and translated nucleic acid data bases, respectively. The IcsB and lcse

genes were found to be of particular interest, since both genes are possibly involved in

peptidoglycan metabolism. Another gene possibly involved in cell wall degradation is a lytic

murein transglycosylase (COG2821), a fragment of which is encoded by the cytotoxic

suppressor plasmid pG39 (Table 3). Deletion of ORFI-3 of plasmid pG39 left only the

truncated lytic murein transglycosylase downstream of the Ptac promoter. This construct was

still cytotoxic in an icmG mutant background, possibly due to expression of a cytotoxic 242

amino acid C-terminal enzyme fragment by using the Ptac promotor and the first internal ATG

as a start codon.

2.5.2 LesB, a homolog of NlpD-like membrane-bound metalloproteases

LcsB is 26% identical to the E. coli YibP protease and 17-18% identical to the NlpD

orthologs from L. pneumophila and E. coli (Fig. 7). The C-terminal domains of the 4 proteins

belong to COG4942 (membrane-bound metallopeptidases, involved in cell division and

chromosome partitioning) and are characteristic for members of the M23/M37 family

(PfamOI551) of putative zinc metallo-endopeptidases from Gram-negative and Gram-positive

50

Page 52: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

bacteria (http://www.sanger.ac.uk/SoftwarelPfam/). The M37 protease family includes

staphylococcal glycy1glycine endopeptidases that hydrolyze the polyglycine interpeptide

bridges of peptidoglycan of Gram-positive bacteria (Ramadurai & Jayaswal, 1997;

Ramadurai et al., 1999; Recsei et al., 1987; Sugai et al., 1997) and the E. coli lipoprotein

NlpD that probably functions in cell wall formation and maintenance of Gram-negative

bacteria (Ichikawa et al., 1994; Lange & Hengge-Aronis, 1994). In E. coli as well as in

1. pneumophila, the nlpD gene is located immediately upstream of the rpoS gene encoding

the stationary growth phase transcription factor RpoS (Hales & Shuman, 1999). Among the

proteins found in the data bases, 1. pneumophila LcsB is most closely related to E. coli YibP.

Purified YibP has endoprotease activity which is inhibited by EDTA, and a fraction of the

protein is membrane-bound (Ichimura et al., 2002). A chromosomal yibP deletion mutant is

defective for cell division, FtsZ ring formation and growth at 42°C but not at 37°C. At the

non-permissive temperature, the yibP mutant forms filamentous, multi-nuc1eoided cells that

tend to lyse, indicating that YibP is required for cell wall degradation and proper cell

division.

1

1

1

1

- - -MRIISLSKSLSetSLG+vILFFCNGLE:AJeAlPNSITCIo:lMRG++VKPRRFAIRPIIYASVLSAGVLLcI~E I

-------------------------------------------- PR-MSAGslKIlvRrmILSLV+LAGCSDTSNPPAPVstmmtP~

Q .~~II~J<K Lpn-LcsBKERAVRQ'KIIImASlLA!I EAlSlATII!IE~ Eco-YibP

CIFFILSAlPCGTRSD QP~------------------ Lpn-NlpDPP+GTTST4PQI PQlQATQQ~PVQPVAQQP+Eco- NlpD

~~~==~ ~~:~~~::SLK------------------------G-------- Lpn-NlpD

.<JGNjO'SGT~ITQAD~IKP+STVEco-NlpD

198 KLSQ~KC!+KRYHSTL1QS1

201EQ~ER E90 ---------------------1200 AVABlPT1TYSE"EQ'SANI~1NK

-------~------------- Lpn-LcsBTTYKPTESEKSLMSRTGGLG Eco -YibP

--------~-------------Lpn-NlpDEiiGAiiEiG--------------------- Eco-NlpD

Lpn-LcsBS Eco-YibP- Lpn-NlpD- Eco-NlpD

Lpn-LcsBEco-yibpLpn~NlpD

Eco-NlpD

Figure 7: Alignment of L. pneumophila LcsB, E. coli YibP and the membrane-boundmetalloproteases NlpD from L. pneumophila and E. coli.

51

Page 53: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

2.5.3 Lcse, a paralog of the lipid A disaccharide synthase LpxB

LcsC is homologous to lipid A disaccharide synthases (LpxB) and, as determined by

searching the conserved domain data base, distantly to MurG glycosyltransferases involved in

peptidoglycan synthesis (COG0707). Interestingly, the genome of L. pneumophila

Philadelphia-l harbors 2 lipid A disaccharide synthase paralogs which are 42% or 30%

identical to E. coli LpxB, respectively (Fig. 8). The gene more closely related to E. coli IpxB

is most likely the L. pneumophila IpxB ortholog since it is located within a cluster encoding

components of the lipopolysaccharide (LPS) biosynthesis pathway, including a

neuraminidase, lauroyllmyristoyl acyltransferase, LpxA, LpxD, LpxB, and the ABC

transporter WlaB. The arrangement of the lpx gene cluster is similar in E. coli, where IpxB is

located downstream of IpxA, fabZ and IpxD (Raetz, 1996).

SS! Lpn-LcsCI Lpn-LpxB

Eco-LpxB

LIlP KIFRKAF IKQHLS_ Lpn-LcsCS E REWTIN Lpn-LpxB

FGE Eco-LpxB

~ ~~Ip1 MTE

:: ~~.II I ~I.t::::.DI~~~:t~~~:~~~;:~101E~- ~~~ ~~;~~vIIIii:_Eco-LpxB

~:: Ittt:l1~1~"_~~~~~+~=~:I!I200 ~sAr:II ~~~c1ERIKAEVAP;JiL

297 YV+EfiIKF_ElSRYI298 HAIl I SITES

300 FWrI . EPQ+wil>l!l\N,

Lpn-LcsC

Lpn-LpxB

Eco-LpxB

Figure 8: Alignment of L. pneumophila LcsC with lipid A disaccharide synthase (LpxB)homologs from L. pneumophila and E. coli.

In the L. pneumophila genome, the IcsC gene appears to form an operon with an

oxidoreductase/dehydrogenase gene and is separated from the putative rodA-ftsloperon by

only one ORF. The latter genes encode the rod shape-determining protein RodA and the

peptidoglycan transglycosylase/peptidase FtsI. RodA and FtsI are membrane-bound, interact

with each other and are required for cell envelope biogenesis and cell division, respectively

(Begg et al., 1986). Analogously, in the E. coli genome, the murG glycosyltransferase gene is

flanked by genes involved in peptidoglycan biosynthesis and cell division (Heijenoort, 1996).

At present, we can not exclude that LesC participates in lipid A biosynthesis. However, the

vicinity of IcsC to rodA-ftsI, its lower homology to IpxB and its similarity to murG suggest

52

Page 54: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

that LcsC catalyzes a disaccharide synthase/glycosyltransferase reaction in peptidoglycan

biosynthesis.

The remote structural similarity of LcsC/LpxB and MurG corresponds to the fact that both

glycosyltransferases catalyze the synthesis of a p-linked lipodisaccharide moiety from an

acylated uridine 5'-diphosphate (UDP)-D-glucosamine (GleN) and a lipid, and both enzymes

liberate UDP during the reaction. LpxB catalyzes the P,l '-6 condensation ofUDP-2,3-diacyl­

GlcN with lipid X (2,2-diacyl-GlcN-1-phosphate), and MurG couples N-acetyl-GleN via an

P,l '-4 linkage to lipid I (N-acety1-muramyl-pentapeptide-phosphoryl-undecaprenol) to form

the lipodisaccharide lipid II that is the minimal subunit of peptidoglycan (Heijenoort, 1996).

2.5.4 Mechanism of Lcse cytotoxicity

Cytotoxicity of an icmG mutant strain harboring the suppressor plasmid pG65 was not only

decreased upon deletion of IcsC (Fig. 5), but overexpression of the gene in an icmG

background enhanced cell death of A. castellanii (Fig. 6), suggesting that the product(s) of

the LcsC enzyme are cytotoxic. While the structure of the toxic product of LcsC remains to

be identified, it might either associate with bacteria or be released into the supematant.

Soluble cytotoxic peptidoglycan derivatives have been described for Bordetella pertussis,

Neisseria gonorrhoeae, Haemophilus injluenzae and other bacteria (Burroughs et al., 1993;

Cloud & Dillard, 2002; Cookson et al., 1989; Luker et al., 1993; Luker et al., 1995). The

murein-derived tracheal cytotoxin (TCT; N-acetyl-GlcN-1,6-anhydro-N-acetyl-muramyl-L­

Ala-y-D-Glu-meso-diaminopimelyl-D-Ala) from B. pertussis, e.g., is released by growing

bacteria and is sufficient to reproduce the cytopathology observed during whooping cough.

The murein-derived toxins from N. gonorrhoeae or H injluenzae are also disaccharide

muramyl tetrapeptides, Le., consist of the monomeric subunit of Gram-negative bacterial

peptidoglycan.

Overexpression of LcsC apparently is cytotoxic for the amoebae specifically in an icmG

mutant but not in other icm/dot or an rpoS mutant background (Fig. 6c). One possibility to

account for this observation would be that LcsC requires a functional IcmIDot secretion

system to exert cytotoxicity. Among the strains tested, the icmG, icmS, icmF, icmW, icmR

and rpoS mutants are expected to form at least partially functional IcmIDot secretion systems,

53

Page 55: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

since these mutants can grow intracellularly in macrophage cell lines (Coers et al., 2000;

Hales & Shuman, 1999; Segal & Shuman, 1999b). Moreover, the icmG mutant is only

partially defective for intracellular growth within A. castellanii at 30°C (Fig. 4), the icmG and

icmF mutants persist in Dictyostelium (Otto et al., 2004), and the icmS and icmW mutants are

not impaired in !cm/Dot-dependent immediate cytotoxicity (Coers et al., 2000; Zuckman et

al., 1999). However, LcsC was cytotoxic specifically in an icmG mutant but neither in other

mutant strains only partially defective for intracellular growth, nor in wild-type

L. pneumophila (data not shown). Therefore, a (limited) functional !cm/Dot transporter is

apparently not sufficient for LcsC cytotoxicity. In contrast to other icm/dot mutants, the icmG

mutant grows to some extent in A. castellanii and persists in D. discoideum, suggesting that

entry and/or intracellular trafficking of the icmG mutant differs from other icm/dot mutants as

well as from wild-type L. pneumophila. It is feasible that overexpression of LcsC exerts

cytotoxic effects only in certain cellular compartments. We currently investigate entry and

intracellular trafficking of the icmG mutant and effects of Lcse on these processes to

establish a link between loss of !cmG (possibly involved in intracellular trafficking) and

cytotoxicity ofLcsC (a putative enzyme of peptidoglycan metabolism).

2.6 ACKNOWLEDGEMENTS

We wish to thank Natalie Schlegel und Judith Zaugg for help with cloning. This work was

supported by grants from the Swiss National Science Foundation (631-065952) and the Swiss

Federal Institute of Technology (17102-3).

2.7 REFERENCES

Altschul, S. F., Madden, T. L., Schaffer, A. A., Zhang, 1., Zhang, Z., Miller, W. & Lipman,D. J. (1997). Gapped BLAST and PSI-BLAST: a new generation ofprotein databasesearch programs. Nucleic Acids Res 25, 3389-3402.

Begg, K. J., Spratt, B. G. & Donachie, W. D. (1986). Interaction between membrane proteinsPBP3 and rodA is required for normal cell shape and division in Escherichia coli. JBacteriol167, 1004-1008.

BUIToughs, M., Rozdzinski, E., Geelen, S. & Tuomanen, E. (1993). A structure-activityrelationship for induction of meningeal inflammation by muramyl peptides. J ClinInvest 92,297-302.

54

Page 56: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

Chen, J., de Felipe, K. S., Clarke, M., Lu, H., Anderson, O. R., Segal, G. & Shuman, H. A.(2004). Legionella effectors that promote nonlytic release from protozoa. Science 303,1358-1361.

Chien, M., Morozova, I., Shi, S. & other authors (2004). The genomic sequence of theaccidental pathogen Legionella pneumophila. Science 305, 1966-1968.

Cirillo, S. 1., Bermudez, 1. E., EI-Etr, S. H., Duhamel, G. E. & Cirillo, J. D. (2001).Legionella pneumophila entry gene rtxA is involved in virulence. Infect Immun 69,508-517.

Cloud, K. A. & Dillard, J. P. (2002). A lytic transglycosylase ofNeisseria gonorrhoeae isinvolved in peptidoglycan-derived cytotoxin production. Infect Immun 70, 2752-2757.

Coers, J., Monahan, C. & Roy, C. R. (1999). Modulation of phagosome biogenesis byLegionella pneumophila creates an organelle permissive for intracellular growth. NatCell Bioi 1, 451-453.

Coers, J., Kagan, J. C., Matthews, M., Nagai, H., Zuckman, D. M. & Roy, C. R. (2000).Identification of Icm protein complexes that play distinct roles in the biogenesis of anorganelle permissive for Legionella pneumophila intracellular growth. Mol Microbiol38, 719-736.

Conover, G. M., Derre, I., Vogel, J. P. & Isberg, R. R. (2003). The LegionellapneumophilaLidA protein: a translocated substrate of the Dot/Icm system associated withmaintenance of bacterial integrity. Mol Microbiol48, 305-321.

Cookson, B. T., Tyler, A. N. & Goldman, W. E. (1989). Primary structure of thepeptidoglycan-derived tracheal cytotoxin of Bordetella pertussis. Biochemistry 28,1744-1749.

Dumenil, G. & Isberg, R. R. (2001). The Legionella pneumophila IcmR protein exhibitschaperone activity for IcmQ by preventing its participation in high-molecular-weightcomplexes. Mol Microbiol40, 1113-1127.

Dumenil, G., Montminy, T. P., Tang, M. & Isberg, R. R. (2004). IcmR-regulated membraneinsertion and efflux by the Legionella pneumophila IcmQ protein. J Bioi Chem 279,4686-4695.

Feeley, J. C., Gibson, R. J., Gorman, G. W., Langford, N. C., Rasheed, J. K., Mackel, D. C.& Baine, W. B. (1979). Charcoal-yeast extract agar: primary isolation medium forLegionella pneumophila. J Clin Microbiol10, 437-441.

Hagele, S., Kohler, R., Merkert, H., Schleicher, M., Hacker, J. & Steinert, M. (2000).Dictyostelium discoideum: a new host model system for intracellular pathogens of thegenus Legionella. Cell Microbiol2, 165-171.

Hales, 1. M. & Shuman, H. A. (1999). The Legionella pneumophila rpoS gene is required forgrowth within Acanthamoeba castellanii. J Bacteriol181, 4879-4889.

Heijenoort, J. v. (1996). Murein synthesis. In Escherichia coli and Salmonella typhimurium.2nd ed., Neidhardt, F.C. (ed. in chief). Washington: ASM press, pp., 1025- 1034.

Hilbi, H., Segal, G. & Shuman, H. A. (2001). Icm/dot-dependent upregulation ofphagocytosis by Legionella pneumophila. Mol Microbiol42, 603-617.

55

Page 57: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

Horwitz, M. & Maxfield, F. (1984). Legionella pneumophila inhibits acidification of itsphagosome in human monocytes. J Cell Bioi 99, 105-114.

Horwitz, M. A. (l983a). The Legionnaires' disease bacterium (Legionella pneumophilia)inhibits lysosome-phagosome fusion in human monocytes. J Exp Med 158, 2108­2126.

Horwitz, M. A. (1983b). Formation of a novel phagosome by the Legionnaires' diseasebacterium (Legionella pneumophila) in human monocytes. J Exp Med 158, 1319­1331.

Horwitz, M. A. & Silverstein, S. C. (1983). Intracellular multiplication of Legionnaires'disease bacteria (Legionella pneumophila) in human monocytes is reversibly inhibitedby erythromycin and rifampin. J Clin Invest 71, 15-26.

Ichikawa, J. K, Li, C., Fu, J. & Clarke, S. (1994). A gene at 59 minutes on the Escherichiacoli chromosome encodes a lipoprotein with unusual amino acid repeat sequences. JBacteriol176, 1630-1638.

Ichimura, T., Yamazoe, M., Maeda, M., Wada, C. & Hiraga, S. (2002). Proteolytic activity ofYibP protein in Escherichia coli. J Bacteriol184, 2595-2602.

Imai, Y., Matsushima, Y., Sugimura, T. & Terada, M. (1991). A simple and rapid method forgenerating a deletion by PCR. Nucleic Acids Res 19,2785.

Kagan, 1. C. & Roy, C. R. (2002). Legionella phagosomes intercept vesicular traffic fromendoplasmic reticulum exit sites. Nat Cell Bioi 4, 945-954.

Kagan, J. C., Stein, M. P., Pypaert, M. & Roy, C. R. (2004). Legionella subvert the functionsofrabl and sec22b to create a replicative organelle. J Exp Med 199, 1201-1211.

Kirby, J. E., Vogel, J. P., Andrews, H. L. & Isberg, R. R. (1998). Evidence for pore-formingability by Legionella pneumophila. Mol Microbiol27, 323-336.

Lange, R. & Hengge-Aronis, R. (1994). The nlpD gene is located in an operon with rpoS onthe Escherichia coli chromosome and encodes a novel lipoprotein with a potentialfunction in cell wall formation. Mol Microbiol13, 733-743.

Luker, K E., Collier, J. L., Kolodziej, E. W., Marshall, G. R. & Goldman, W. E. (1993).Bordetella pertussis tracheal cytotoxin and other muramyl peptides: distinct structure­activity relationships for respiratory epithelial cytopathology. Proc Natl Acad Sci USA 90, 2365-2369.

Luker, K E., Tyler, A. N., Marshall, G. R. & Goldman, W. E. (1995). Tracheal cytotoxinstructural requirements for respiratory epithelial damage in pertussis. Mol Microbiol16, 733-743.

Luo, Z. Q. & Isberg, R. R. (2004). Multiple substrates of the Legionella pneumophilaDot/Icm system identified by interbacterial protein transfer. Proc Natl Acad Sci USA101,841-846.

Marchler-Bauer, A., Anderson, J. B., DeWeese-Scott, C. & other authors (2003). CDD: acurated Entrez database of conserved domain alignments. Nucleic Acids Res 31,383­387.

56

Page 58: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

Mintz, C. S. & Shuman, H. A. (1987). Transposition of bacteriophage Mu in theLegionnaires' disease bacterium. Proc Natl Acad Sci USA 84,4645-4649.

Moffat,1. F. & Tompkins, L. S. (1992). A quantitative model of intracellular growth ofLegionella pneumophila in Acanthamoeba castellanii. Infect Immun 60, 296-301.

Molmeret, M., Alli, O. A., Zink, S., Flieger, A., Cianciotto, N. P. & Kwaik, Y. A. (2002).icmT is essential for pore formation-mediated egress ofLegionella pneumophila frommammalian and protozoan cells. Infect Immun 70, 69-78.

Morales, V. M., Backrnan, A. & Bagdasarian, M. (1991). A series of wide-host-range low­copy-number vectors that allow direct screening for recombinants. Gene 97, 39-47.

Morozova, 1., Qu, X., Shi, S., Asamani, G., Greenberg, J. E., Shuman, H. A. & Russo, 1. J.(2004). Comparative sequence analysis of the km/dot genes in Legionella. Plasmid51, 127-147.

Nagai, H. & Roy, C. R. (2001). The DotA protein from Legionella pneumophila is secretedby a novel process that requires the Dot/Icm transporter. Embo J 20, 5962-5970.

Nagai, H., Kagan, J. C., Zhu, x., Kahn, R. A. & Roy, C. R. (2002). A bacterial guaninenucleotide exchange factor activates ARF on Legionella phagosomes. Science 295,679-682.

Nagai, H. & Roy, C. R. (2003). Show me the substrates: modulation of host cell function bytype IV secretion systems. Cell Microbiol5, 373-383.

Otto, G. P., Wu, M. Y., Clarke, M., Lu, H., Anderson, O. R., Hilbi, H., Shuman, H. A. &Kessin, R. H. (2004). Macroautophagy is dispensable for intracellular replication ofLegionella pneumophila in Dictyostelium discoideum. Mol Microbiol51, 63-72.

Purcell, M. & Shuman, H. A. (1998). The Legionella pneumophila icmGCDJBF genes arerequired for killing of human macrophages. lrifect Immun 66, 2245-2255.

Quinn, F. D. & Tompkins, L. S. (1989). Analysis ofa cloned sequence of Legionellapneumophila encoding a 38 kD metalloprotease possessing haemolytic and cytotoxicactivities. Mol Microbiol3, 797-805.

Raetz, C. R. H. (1996). Bacteriallipopolysaccharides: a remarkable family ofbioactivemacroamphiphiles. In Escherichia coli and Salmonella typhimurium. 2nd ed.,Neidhardt, F.C. (ed. in chief). Washington: ASM press, pp., 1035-1063.

Ramadurai, L. & Jayaswal, R. K. (1997). Molecular cloning, sequencing, and expression oflytM, a unique autolytic gene of Staphylococcus aureus. J Bacteriol179, 3625-3631.

Ramadurai, L., Lockwood, K. J., Nadakavukaren, M. J. & Jayaswal, R. K. (1999).Characterization of a chromosomally encoded glycylglycine endopeptidase ofStaphylococcus aureus. Microbiology 145, 801-808.

Recsei, P. A., Gruss, A. D. & Novick, R. P. (1987). Cloning, sequence, and expression of thelysostaphin gene from Staphylococcus simulans. Proc Natl Acad Sci USA 84, 1127­1131.

Rowbotham, T. 1. (1980). Preliminary report on the pathogenicity of Legionella pneumophilafor freshwater and soil amoebae. J Clin Pathol 33, 1179-1183.

57

Page 59: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Identification of virulence related genes using the amoebae plate test

Roy, C. R., Berger, K. H. & Isberg, R. R. (1998). Legionella pneumophila DotA protein isrequired for early phagosome trafficking decisions that occur within minutes ofbacterial uptake. Mol Microbiol28, 663-674.

Sadosky, A B., Wiater, L. A & Shuman, H. A (1993). Identification of Legionellapneumophila genes required for growth within and killing of human macrophages.Infect Immun 61, 5361-5373.

Segal, G. & Shuman, H. A (1997). Characterization of a new region required for macrophagekilling by Legionella pneumophila. Infect Immun 65, 5057-5066.

Sega1, G., Purcell, M. & Shuman, H. A. (1998). Host cell killing and bacterial conjugationrequire overlapping sets of genes within a 22-kb region of the Legionellapneumophila genome. Proc Natl Acad Sci USA 95, 1669-1674.

Segal, G. & Shuman, H. A. (1998). Intracellular multiplication and human macrophagekilling by Legionella pneumophila are inhibited by conjugal components of IncQplasmid RSF1010. Mol Microbiol30, 197-208.

Segal, G. & Shuman, H. A (1999a). Possible origin of the Legionella pneumophila virulencegenes and their relation to Coxiella burnetii. Mol Microbiol 33, 669-670.

Segal, G. & Shuman, H. A (1999b). Legionella pneumophila utilizes the same genes tomultiply within Acanthamoeba castellanii and human macrophages. Infect Immun 67,2117-2124.

Solomon, J. M., Rupper, A, Cardelli, 1. A & Isberg, R. R. (2000). Intracellular growth ofLegionella pneumophila in Dictyostelium discoideum, a system for genetic analysis ofhost-pathogen interactions. Infect Immun 68, 2939-2947.

Steinert, M., Hentschel, U. & Hacker, J. (2002). Legionella pneumophila: an aquatic microbegoes astray. FEMS Microbiol Rev 26, 149-162.

Sturgill-Koszycki, S. & Swanson, M. S. (2000). Legionella pneumophila replication vacuolesmature into acidic, endocytic organelles. J Exp Med 192, 1261-1272.

Sugai, M., Fujiwara, T., Akiyama, T., Ohara, M., Komatsuzawa, H., Inoue, S. & Suginaka,H. (1997). Purification and molecular characterization of glycylg1ycine endopeptidaseproduced by Staphylococcus capitis EPK1. J Bacteriol179, 1193-1202.

Swanson, M. S. & Isberg, R. R. (1995).Association ofLegionella pneumophila with themacrophage endoplasmic reticulum. In Infect Immun, pp. 3609-3620.

Szeto, L. & Shuman, H. A (1990). The Legionella pneumophila major secretory protein, aprotease, is not required for intracellular growth or cell killing. Infect Immun 58,2585-2592.

Tatusov, R. L., Natale, D. A, Garkavtsev, 1. V. & other authors (2001). The COG database:new developments in phylogenetic classification of proteins from complete genomes.Nucleic Acids Res 29,22-28.

Ti1ney, L. G., Harb, O. S., Connelly, P. S., Robinson, C. G. & Roy, C. R. (2001). How theparasitic bacterium Legionella pneumophila modifies its phagosome and transforms itinto rough ER: implications for conversion of plasma membrane to the ER membrane.J Cell Sci 114, 4637-4650.

58

Page 60: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 2

Vogel, J. P., Andrews, H. L., Wong, S. K. & Isberg, R. R. (1998). Conjugative transfer by thevirulence system of Legionella pneumophila. Science 279, 873-876.

Watarai, M., Derre, 1., Kirby, J., Growney, J. D., Dietrich, W. F. & Isberg, R. R. (2001).Legionella pneumophila is internalized by a macropinocytotic uptake pathwaycontrolled by the Dot/Icm system and the mouse 19n1locus. J Exp Med 194, 1081­1096.

Wiater, L. A., Sadosky, A. B. & Shuman, H. A. (1994). Mutagenesis of Legionellapneumophila using Tn903dlllacZ: identification of a growth-phase-regulatedpigmentation gene. Mol Microbiolll, 641-653.

Wiater, L. A., Dunn, K., Maxfield, F. R. & Shuman, H. A. (1998). Early events in phagosomeestablishment are required for intracellular survival of Legionella pneumophila. InfectImmun 66, 4450-4460.

Wintermeyer, E., Rdest, D., Ludwig, B., Debes, A. & Hacker, J. (1991). Characterization oflegiolysin (lly), responsible for haemolytic activity, colour production andfluorescence ofLegionella pneumophila. Mol Microbiol 5, 1135-1143.

Wright, E. K., Goodart, S. A., Growney, J. D. & other authors (2003). Naip5 Affects HostSusceptibility to the Intracellular Pathogen Legionella pneumophila. Curr Bioi 13, 27­36.

Yamamoto, Y., Klein, T. W., Newton, C. A., Widen, R. & Friedman, H. (1988). Growth ofLegionella pneumophila in thioglycolate-elicited peritoneal macrophages from AlJmice. Infect Immun 56,370-375.

Yanisch-Perron, c., Vieira, J. & Messing, J. (1985). Improved M13 phage cloning vectorsand host strains: nucleotide sequences of the M13mpl8 and pDC19 vectors. Gene 33,103-119.

Zink, S. D., Pedersen, L., Cianciotto, N. P. & Abu-Kwaik, Y. (2002). The Dot/Icm type IVsecretion system of Legionella pneumophila is essential for the induction of apoptosisin human macrophages. Infect Immun 70, 1657-1663.

Zuckrnan, D. M., Hung, J. B. & Roy, C. R. (1999). Pore-forming activity is not sufficient forLegionella pneumophila phagosome trafficking and intracellular growth. MolMicrobiol 32, 990-1001.

59

Page 61: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue Lcse

3 The cytotoxic Legionella pneumophila IpxB paralogueIcsC is involved in lipid A biosynthesis and upregulatedduring bacterial growth in amoebae

Urs Albers ., Andre Tiaden ., Thomas Spirig" and Hubert Hilbi 1

1 ETH Zurich, Institute of Microbiology, Wolfgang-Pauli Strasse 10, HCI G405, 8093

Zurich, Switzerland.

Abbreviations: ACES, N-(2-acetamido)-2-aminoethanesulfonic acid; CYE, charcoal-yeast

extract; km/dot, intracellular multiplication/defective organelle trafficking; GlcN3N, 2,3­

diamino-2,3-dideoxy-a-D-glucopyranose; GlcNAc; 2-acetamido-2-deoxy-D-glucose (N­

acetyl-glucosamine); GlcNAc3N, 2-acetamido-3-amino-2,3-dideoxy-a-D-glucopyranose; !cs,

Legionella cytotoxic suppressor; MOl, multiplicity of infection; RT-PCR, reverse

transcription polymerase chain reaction; PI, propidium iodide; PYG, proteose-yeast extract­

glucose; T4SS, type IV secretion system.

Manuscript in preparation

60

Page 62: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

3.1 ABSTRACT

The facultative intracellular pathogen Legionella pneumophila replicates within amoebae and

macrophages. Intracellular growth takes place within a replicative vacuole and requires the

bacterial !cm/Dot type IV secretion system. Using Acanthamoeba castellanii as a host cell,

we previously identified IcsC (Legionella cytotoxic suppressor), a paralogue of the lipid A

disaccharide synthase IpxB, as a cytotoxic factor of L. pneumophila. Among bacteria,

L. pneumophila is unique in harboring two paralogues of IpxB and two or three paralogues of

the UDP-GlcNAc acyltransferases IpxA or IpxD, respectively. In the genome of

L. pneumophila, IcsC (lpxBl) lies adjacent to gnnA. GnnA is an NAD+-dependent oxidase,

which catalyzes the first step in the conversion of UDP-GlcNAc to the 3-amino derivative

UDP-GlcNAcN3, the precursor of G1cN3N analogues of lipid A. IpxB2 clusters with IpxD2,

IpxA2 and other genes involved in lipid A biosynthesis. We show here that IpxBl and gnnA

fonn a transcriptional unit. Moreover, IpxBl or IpxB2 complemented the growth defect of the

temperature sensitive E. coli IpxB mutant strain MN7 at the non-pennissive temperature to a

similar extent, indicating that both corresponding enzymes possess lipid A disaccharide

synthase activity. However, the two L. pneumophila IpxB paralogues are functionally not

equivalent, since expression of IpxBl but not IpxB2 in an icmG mutant was cytotoxic for

A. castellanii. Using an RT-PCR approach, we demonstrate that in AYE broth IpxBl, IpxB2,

IpxAl, and IpxA2 are expressed in exponential as well as in early stationary growth phase.

Interestingly, in A. castellanii, IpxBl and IpxAl are expressed at early time points (4 h post

infection), while IpxB2 and IpxA2 are expressed only at later time points (24 h post infection).

These results are in agreement with the hypothesis that L. pneumophila evolved and,

depending on environmental conditions, differentially regulates paralogues of UDP-GlcNAc

acyl transferases and disaccharide synthases involved in LPS biosynthesis.

3.2 INTRODUCTION

Legionella pneumophila is a Gram-negative bacterium that colonizes different niches in the

environment. The bacteria not only survive and replicate in numerous amoebae and ciliates

(Fields, 1996; Steinert et al., 2002), but also colonizes and persists in biofilms (Mampel et

al., 2006; Murga et al., 2001). Moreover, L. pneumophila is an opportunistic human pathogen

that causes the life-threatening pneumonia Legionnaires' disease and grows within

61

Page 63: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

macrophages from different sources, including human alveolar macrophages (Nash et al.,

1984), primary mouse macrophages from certain mouse strains (e.g. AlJ; Sporri et al., 2006;

Yamamoto et aI., 1988) and several macrophage-like cell lines (Fields, 1996).

Phagocytosis and intracellular replication of 1. pneumophila depends on the Icm/Dot type IV

secretion system (T4SS), a conjugation apparatus which transports more than 30 putative

"effector" proteins (Hilbi et al., 2001; Segal et aI., 1998; Vogel et al., 1998). The !cm/Dot

T4SS is also required for 1. pneumophila to survive and grow on agar plates in presence of

Acanthamoeba castellanii. This "amoebae plate test" was recently used to screen an

1. pneumophila chromosomal library for multicopy suppressors of the partial growth defect

of an icmG mutant strain (Albers et al., 2005). In this screen, a paralogue of the lipid A

disaccharide synthase IpxB was identified and termed IcsC (Legionella cytotoxic suppressor).

Expression of IcsC in the icmG mutant strain rendered 1. pneumophila more cytotoxic for

A. castellanii, but did not enhance intracellular replication of the bacteria.

The genomes of the 1. pneumophila strains Philadelphia, Paris and Lens (Cazalet et al., 2004;

Chien et al., 2004) encode orthologues of the E. coli lipid A biosynthetic genes, indicating

that L. pneumophila employs a lipid A biosynthesis pathway similar to the one described for

E. coli (Raetz and Whitfield, 2002). 1. pneumophila synthesizes a lipid A analogue

containing 2,3-diamino-2,3-dideoxyglucose (Zahringer et al., 1995). The initial step in the

biosynthesis of this lipid A analogue is the conversion of UDP-GlcNAc to its 3-amino

derivative UDP-GlcNAc3N, which in Acidithiobacillus ferrooxidans is catalyzed by the

NAD+-dependent oxidase GnnA and the L-glutamate-dependent aminotransferase GnnB

(Sweet et al., 2004). Lipid A of L. pneumophila is substituted with unusual long-chain,

branched and dihydroxylated fatty acids (Moll et al., 1992). The O-antigen of1. pneumophila

LPS is a homo-polymer of an unique carbohydrate unit, legionaminic acid, which can be

further O-acetylated and N-methylated (Knirel et aI., 1994; Kooistra et al., 2002a).

Structural variations of lipid A alter the physical properties of the outer membrane and

thereby influence endotoxic activity and resistance against antibiotic compounds (Raetz and

Whitfield, 2002). L. pneumophila LPS has low endotoxic activity, possibly due to the

unusually long and branched fatty acid residues (Neumeister et al., 1998). Moreover,

L. pneumophila harbors a gene similar to the Salmonella lipid A acyl transferase pagP that

confers resistance to cationic antimicrobial peptides and promotes intracellular replication

(Robey et al., 2001).

Reversible LPS phase variation is a frequently adopted strategy of pathogenic bacteria to alter

their surface carbohydrate pattern and thus adapt to changes in the environment (immune

62

Page 64: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

evasion, colonization of new niches). Phase variation of 1. pneumophila serogroup 1

subgroup OLDA strain RC1 was found to decrease virulence in a guinea pig infection model,

diminish intracellular replication within macrophage-like HL60 cells or A. castellanii, and

reduce serum resistance (Luneberg et al., 1998). Phase variation in 1. pneumophila was

shown to be due to the reversible chromosomal excision of a 30 kb genetic element

(Luneberg et al., 2001). In the virulent wild-type strain RCI the 30 kb element is integrated

into a specific site in the chromosome. Contrarily, in the phase variant strain 811, the element

is excised and replicates as a high copy number plasmid. The 30 kb element does not harbor

genes related to LPS biosynthesis, yet phase variation abolished N-linked methylation of

legionaminic acid in the O-chain polysaccharide (Kooistra et al., 2002b) and shifted the

profile of fatty acids attached to lipid A towards shorter chain moieties (Kooistra et aI.,

2002a), thus demonstrating the capability of 1. pneurnophila to alter its LPS and lipid A

structure.

In this report, we analyze in more detail the previously identified cytotoxic 1. pneurnophila

lipid A disaccharide synthase paralogue lcsC. We demonstrate that both 1. pneurnophila lpxB

paralogues are involved in lipid A biosynthesis, but only one (lcsC) is cytotoxic for amoebae.

Analysis of the lipid A biosynthesis genes present in the 1. penurnophila genome reveals that,

in contrast to other bacteria, the acyltransferases are present in multiple copies. Expression

analysis of lpxA and lpxB shows differential regulation of paralogous genes upon intracellular

replication of 1. pneumophila within A. castellanii and upon transfer from nutrient broth into

distilled water.

3.3 METHODS

3.3.1 Bacteria, amoebae and reagents

The 1. pneumophila strains used were wild-type strain JR32 (Philadelphia-I, serogroup 1;

(Sadosky et al., 1993), and an isogenic deletion mutant containing a kanamycin (Km)

resistance cassette in icmG (MW635; Purcell and Shuman, 1998).1. pneurnophila was grown

on charcoal-yeast extract (CYE) agar plates (Feeley et al., 1979) or in ACES-buffered-yeast

extract broth (AYE). Supplements were used at the following concentrations:

chloramphenicol (Cm; 5 ~g/ml), Km (50 ~g1ml), IPTG (0.25 mM or 0.05 mM to induce

expression of 1. pneurnophila lcsC or lpxB, respectively). Escherichia coli strain TOPI0

(Invitrogen) or the conditional E. coli lpxB mutant strain MN7 (Nishijima et al., 1981) were

63

Page 65: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

cultured in LB medium, supplemented with Cm (30 ~g/ml), ampicillin (Ap, 100 Ilg/ml), or

0.2% L-(+)-arabinose, if required.

A. castellanii (ATCC 30234) was grown in proteose-yeast extract-glucose medium (PYG) at

30 QC (Motrat and Tompkins, 1992; Segal and Shuman, 1999) and split once or twice a week.

High gel strength agar was from Serva, proteose peptone from Becton Dickinson Biosciences

and Bacto yeast extract from Difco, respectively. All other reagents were from Sigma.

3.3.2 Identification of homologues of L. pneumophila lipid Abiosynthesis genes

To identify proteins involved in lipid A biosynthesis, a blastp search (Altschul et al., 1997)

was performed in the genomes of 1. pneumophila strains Philadelphia-1, Lens and Paris

(Cazalet et al., 2004; Chien et al., 2004) and other bacterial genomes. The proteins from

E. coli K12 were used as query (LpxA, accession number POA722; LpxB, P10441; LpxC,

POA725; LpxD, P21645; LpxH, P43341). The A. ferrooxidans proteins GnnA (AAS48421)

and GnnB (AAS48422), catalyzing the conversion ofUDP-GlcNAc to its 3-amino derivative

UDP-GlcNAc3N, were used as query to search for the corresponding 1. pneumophila

enzymes.

3.3.3 Construction of plasmids

DNA manipulations were performed according to standard protocols, and plasmids were

isolated using commercially available kits from Qiagen or Macherey-Nagel. To construct

plasmid pMMB207C-RBS-lcsC, the IcsC gene (lpg1371) including a ribosome binding site

(RBS) was released from plasmid pUA26 (pMMB207-RBS-lcsC) by digestion with EcoRI

and BamHI and inserted into the same restriction sites of pMMB207C. Plasmid pTS-10

(pMMB207C-RBS) was constructed by cutting pMMB207C-RBS-lcsC with NdeI and

BamHI, fill in with Klenow polymerase and religation with T4 DNA polymerase. To

construct plasmid pMMB207C-RBS-lpxB the corresponding open reading frame (lpg2945)

was amplified by PCR using chromosomal DNA as template and the oligos oLpxBfo and

oLpxBre (Table 1), respectively. The PCR fragment was cut with NdeI and PstI and inserted

64

Page 66: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

into pMMB207C-RBS-IcsC cut with the same enzymes, thus replacing lcsC by lpxB. All

constructs containing PCR fragments were sequenced.

3.3.4 Complementation of a conditional E. coli !pxB mutant strain withL. pneumophi!a !pxB paralogues

The ability of the L. pneumophila lpxB paralogues to complement an lpxB mutant was

examined using the temperature sensitive E. coli lpxB mutant MN7 (kindly provided by

C.R.H. Raetz, Duke University, NC; Nishijima et al., 1981). The mutant was transformed by

electroporation with empty vectors (pUCI8, pMMB207C-RBS), with pSR8 (expressing

E. coli lpxB), or with vectors expressing the L. pneumophila lpxB paralogues (pMMB207C­

RBS-lesC, pMMB207C-RBS-IpxB).

Cultures grown over night were adjusted to an OD6oo of 1.0, and dilutions were plated on LB

agar with 80 Ilg/ml Ap (pUCI8, pSR8) or 20 Ilg/ml Cm (pMMB207C and derivatives).

Expression of lpxB in pSR8 was induced with 0.2% arabinose. Since induction of lcsC or

lpxB with IPTG is toxic for the bacteria, expression of these two genes was not induced and,

therefore, remained at a basal level due to leakiness of the Ptac promotor. The plates were

incubated at 30°C or 42 cC, respectively, and the number of colonies was determined after

24 hours.

3.3.5 Cytotoxicity assay

4 x 104 A. castellanii per well were seeded in PYG onto a 24 well plate and let grow over

night. Before an infection, the PYG medium was replaced with Ac buffer (Moffat and

Tompkins, 1992). Bacteria from 3 or 4 days old plates grown in the presence of 0.2 mM

IPTG to induce expression of L. pneumophila lcsC or lpxB, respectively, were resuspended

and diluted in sterile water to infect the amoebae at an MOl of 50. The infection was

synchronized by spinning down the bacteria (880 x g, 5 min), and the plates were incubated

at 30°C. Two days post infection, propidiurn iodide (PI) solution was added to the wells at a

final concentration of 1 Ilg/ml. After several minutes incubation, the amoebae were viewed in

brightfield or by epifluorescence microscopy with an inverse microscope (Zeiss Axiovert

65

Page 67: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

200M, 20 x objective). The percentage of dead (PI-positive) amoebae was determined by

counting the number of total and fluorescent amoebae.

3.3.6 Expression analysis of LPS biosynthesis genes by RT-peR

Expression of LPS biosynthesis genes was quantified in L. pneumophila strain JR32 grown

for 3-4 d on CYE agar plates followed by growth in AYE medium and exposure to different

conditions. Cotranscription of the gnnA and IcsC/lpxBI genes was analyzed in

L. pneumophila grown in AYE to exponential growth phase (OD600 0.8). To compare gene

expression in exponential and post-exponential growth phase, 3 ml AYE medium was

inoculated (OD600 0.1) with bacteria grown on agar plates, and at the time points indicated,

samples were processed for RNA isolation as described below. Gene expression under

osmotic stress and nutrient deprivation was determined by splitting a bacterial culture in

stationary growth phase (23 h, OD600 3.3). Half of the culture was washed once with distilled

water and resuspendend in distilled water, the other half remained in the growth medium as

control. Both cultures were kept at room temperature for 4 h before RNA was extracted.

To analyze RNA from bacteria growing intracellularly in amoebae, 2 x 106 A. castellanii

were seeded in a 10 cm dish the day before the experiment, infected with L. pneumophila in

early stationary growth phase at an Mal of 40 and incubated at 30°C. 2.5 h post infection,

50 ~g/ml gentamicin was added to kill extracellular bacteria. After additional 1.5 or 21.5 h,

the supernatant was aspirated, the plates were chilled on ice, and the amoebae were scraped

into 1.75 ml RNAprotect bacteria reagent (Qiagen). To disrupt the host cells, the suspension

was passed four times through a ball homogenizer (lsobiotec, Heidelberg) with a clearance of

6 ~m. Host cell debris were removed by centrifugation (2 min, 250 x g). To pellet bacteria

the supernatant was spun again (10 min, 5200 x g), and RNA was extracted.

RNA was extracted with the RNEasy Midi Prep kit according to the manufacturer's

instructions (Qiagen) and quantified by ODZ60• Contaminating genomic DNA was digested

using the DNAse RQl (Promega). To synthesize cDNA, M-MLV reverse transcriptase

(Promega) and the 3' primers listed in Table 1 were used. Cotranscription of gnnA and

IcsC/lpxBl was analyzed using primer oUA50 to synthesize cDNA and primers oUA50 and

oUA55 for the PCR.

The primers were annealed to the RNA for 10 min at 70°C and cooled down on ice before

the other components were added. The final reaction mixture contained 200 ng RNA (400 ng

66

Page 68: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

RNA from bacteria infecting A. castellanii), 20 pmol 3' primer, 0.5 mM of each dNTP and

80 U M-MLV reverse transcriptase in 25 III 1x reaction buffer (Promega) and was incubated

for 60 min at 42 QC. For the PCR reaction, 2.5 III reverse transcription mixture was used as

template, and 50 pmol each of3' and 5' primer (Table 1),0.2 mM of each dNTP and 1 III Taq

polymerase were added. The primers were designed to have a melting temperature of 58­

60 QC and to yield a 480-520 bp product with 40-60% GC content. The PCR conditions were:

95 QC (30 s), 55 QC (30 s), 72 QC (40 s) during the first 10 cycles, and afterwards 72 QC (45 s).

Samples were taken after the number of cycles indicated to determine the amount of the PCR

products on an agarose gel. Sample size and settings for photographic documentation were

kept identical in all assays.

P1asmidspGS-Lc-63-14 icmGCD in pMMB207-Km14 (Purcell and Shuman, 1998)pMMB207C RSF1010 derivative, IncQ, oriT, (Chen et al., 2004)

lacf4, Ptac, !1mobA, CmR

pMMB207C-RBS- Expression of1. pneumophila This worklesC IcsC/lpxBl

pMMB207C-RBS- Expression of1. pneumophila IpxB2 This workIpxB

pSR8 Expression ofE. coli IpxB, ParaB, ApR (Crowell et al., 1986)pTS-10 pMMB207C-RBS, !1mobA, RBS This workpUA26 pMMB207-RBS-lesC, MobA pos. (Albers et aI., 2005)pUC19 (Yanisch-Perron et al.,

1985)Oligos I

oLpxB2fo CGGAATTCCATATGACTATGAA 5'oflpxB2 (lpg 2945)AAGACCGACCC

oLpxB2re GTACAACTGCAGTTAAGTCAAA 3' of IpxB2 (lpg 2945)GAAGTCCCCAAAATC

oUA39 CCGCATGTAGTAATTGAAGG 5' oflpxA1 (lpg 0511)oUA40 CACGTCGCAAACAATCAATC 3' oflpxAl (lpg 0511)oUA41 CCGTCATTGGTTCTCATGTG 5' oflpxA2 (lpg 2943)oUA42 CTTGCGACAGAACTGTATCC 3' oflpxA2 (lpg 2943)oUA49 CTGGAAATTAGTGGCATTGG 5' oflcsC/lpxBl (lpg

1371)oUA50 GGCAGTAAGGCAATAATTGG 3' of lesC/lpxB1 (lpg

1371)oUA51 GTGGTCCTCAAATGCTCAAG 5' oflpxB2 (lpg 2945)oUA52 GATTTCACCTACCCGACTAC 3' of IpxB2 (lpg 2945)oUA53 TAAGACCAGCTCCTGTAAAC 5' of IpxC (lpg 2608)oUA54 GCCCTACAAACTTCTTTGAC 3' of IpxC (lpg 2608)oUA55 CACTGCGTAGCAAATGTAAC 5' ofgnnA (lpg 1372)oUA56 AGGACCATGTCAAGTTACAG 5' ofrpoA (lpg 0354)

67

Page 69: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

oUA57 CGAACAGTCAACTCCAAATC 3' ofrpoA (lpg 0354)oUA60 GCAGCAACCAGTATTAACTC 5' offlaA (lpg 1340)oUA61 TAGCAACAGTACCACCAATC 3' offlaA (lpg 1340)oUA64 ATACCGCATAATGTCTGAGG 5' of 168 rDNA (lpg 0302)oUA65 TACACCGGAAATTCCACTAC 3' of 168 rDNA (lpg 0302)

Table 1: Plasmids and oligonucleotides used. 1Restriction sites are underlined.

3.4 RESULTS

3.4.1 L. pneumophila paralogues of LPS biosynthesis genes

The L. pneumophila lcsC gene was recently identified as a gene which is cytotoxic for

amoebae upon overexpresssion (Albers et al., 2005). LcsC is 34% identical to E. coli LpxB,

the lipid A disaccharide synthase, which catalyzes the glycosyl transferase reaction between

UDP-2,3-diacylglucosamine and 2,3-diacylglucosamine-l-phosphate to fonn the tetra­

acylated lipid IVA in the course of lipid A biosynthesis (Figure 1). The L. pneumophila

genome harbors another IpxB gene (lpxB2), which is 43% identical to E. coli IpxB on an

amino acid level. The apparent paralogues LcsC/LpxB1 and LpxB2 share 31 % identity.

In order to gain more insight into lipid A biosynthesis in L. pneumophila, we examined the

genomes of L. pneumophila Philadelphia-I, Paris and Lens (Cazalet et al., 2004; Chien et aI.,

2004) for the presence of lipid A biosynthesis genes. The genome of L. pneumophila was

found to harbor orthologues of all the E. coli genes required for GlcNAc-based lipid A

biosynthesis (lpxA, -B, -C, -D, -H, -K, -L, and WaaA (KdtA)). Moreover, in agreement with

the finding that L. pneumophila synthesizes a GlcNAc3N-derivative of lipid A (Zahringer et

al., 1995), L. pneumophila also harbors orthologues ofA. ferooxidans gnnA and gnnB, which

are required for the conversion of UDP-GlcNAc into its 3-amino derivative UDP-GlcNAc3N

(Sweet et al., 2004). The proposed pathway of lipid IVA biosynthesis in L. pneumophila

based on the presence of these genes is shown in Figure 1. Interestingly, all three

L. pneumophila strains harbor two copies of IpxA and IpxB, and three copies of IpxD. LpxA

and LpxD are acyl transferases attaching primary acyl residues to the carbohydrate backbone.

68

Page 70: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

~0tI MAl)< MADH[~0tI ]L-G1u a..KG 0tI J.OH.C,.. 0tI A_ I-OH-C,.- 0tI lIMP 0tIHO 0 0 ACP HO 0 ACP HO

HO NH ~ 0 NH .J-L~-'-OH~~~~-L. OH~ l?

0-<, UOP 0-<, ID' 0=\ UOP HOl 0-<., UOI' i 0) ID' HOl1 O-f'OI1HO 0tIGl1nA GrmB LpJ<A LpJ<C LpJ<O LpJ<H

IIPQ1372 (39%) I Ipg1424 (39%) Ipg2608 (68%) (2x) Ipg1552 (38%)Ipg0766 (34%)

Ipg2943 (45%) A2 Ipg2944 (33%) 02Ipg0511 (47%) A1 Ipg0508 (36%) 01

Ipg0100 (30%) 03

:~:~ ~:~ :::~:;: :t:,~I, ~:'>l>1JtL:::o 0 J ~O

~~ --~ ~~.~~

Ipg0383 (24%) L3 Ipg2340 (43%)

Kdo,·Llpld A (GlcN3N analogue) Kdo,.Llpld IVA(GlcN3N analogue) Lipid IVA (GlcN3N analogue)

Figure 1: Proposed pathway of biosynthesis of lipid IVA in L. pneumophila. LPS biosynthesisenzymes of L. pneumophila strain Philadelphia-l homologous to E. coli or A. ferrooxidans (GnnA,GnnB) proteins are indicated. The degree of identity on protein level is indicated in parenthesis. Theacyl chains shown symbolize the fatty acids and do not correspond to the published structure ofL. pneumophila lipid A. ACP: acyl carrier protein, Kdo: 3-deoxy-D-manno-oct-2-ulosonic acid.(Scheme modified after Sweet et al., 2004).

Kdoz-lipid IVA is converted into lipid A in E. coli by addition of lauroyl and myristoyl

residues to the primary acyl chains of the Kdoz-linked sugar catalyzed by the acyl-carrier

protein-dependent acyl transferases LpxL and LpxM. The L. pneumophila genome contains

three E. coli IpxL homologues (see Figure 1). These genes also show similar degrees of

homology to the E. coli acyl transferase LpxP that inserts palmitoleate instead of laurate as

secondary acyl chain under cold shock conditions. These genes do not show significant

homology to E. coli IpxM. Furthermore, the L. pneumophila genome contains the pagP

homologue rep (lpg0025) (Robey et al., 2001), that seems to be activated by low magnesium

concentrations and most likely adds an additional third secondary acyl chain to lipid A.

The paralogues of IpxA, IpxB, and IpxD are clustered similarly in the L. pneumophila strains,

yet the genomic organization of the genes varies to some extent (Table 2). The

L. pneumophila genome is unique among 12 bacterial genomes analyzed in containing

multiple paralogues of IpxA, IpxB, and IpxD. The IpxB gene was found to be present only as a

69

Page 71: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

single copy in 25 other bacterial genomes analyzed. Among the bacteria examined were

Coxiella burnetii, the closest relative of L. pneumophila, and other pathogenic or symbiontic

species from different taxonomic groups.

Locus Philadelphia-l (lpg) 1 Paris (lpp) 1 Lens (lpl) 1

IpxBl/lcsC-gnnA 1371-1372 1325-1326 1322-1323

IpxL-A2-D2-B22940-0RF-ORF-2943- 3012-0RF-3014- 2870-0RF-2872-2873-2944-2945 (L--ADB) 3015-3016 (L-BDA) 2874 (L-BDA)

IpxDI-AI 0508-0RF-ORF-0511 0571-0RF-0573 0547-ORF-0549

IpxK-msbA 2 1818-1819 1781-1782 1782-1783

Table 2: Genomic arrangement of selected lipid A biosynthesis genes in L. pneumophila strains.1 Gene number as annotated in the Philadelphia-l (lpg), Paris (lpp) or Lens (lpl) genome. ORFindicates an open reading frame, which is not involved in lipid A biosynthesis or whose function isnot clear. The genes Ipg2941, Ipp3013 and Ipl2871 encode proteins with weak homologies to acyltransferases, and Ipg2942 shows no homology to known proteins. The genes Ipg0510, Ipp0572 andIp10548 are annotated as hydroxymyristoyl[ACP]dehydratase FabZ, and Ipg0509 is annotated as ORFbelonging to a sugar transporter family.2 MsbA is an ABC transporter involved in lipid A translocation through the inner membrane.

In summary, L. pneumophila seems to be unique in harboring multiple copies of lipid A

disaccharide synthases (LpxB) and acyl transferases (LpxA, -D, -L). A common feature of

these enzymes is that they modify the acylation state of lipid A or its precursors in the LPS

biosynthesis pathway. The paralogues might have different acyl substrate specificities and

thus determine the variable length of the lipid A acyl chains identified in the L. pneumophila

phase variant. Contrarily, in other bacterial genomes, the genes necessary for lipid A

biosynthesis are present in only one copy (Raetz and Whitfield, 2002).

3.4.2 Complementation of a conditional E. coli IpxB mutant strain withL. pneumophila IpxB paralogues

The genomic organization of the L. pneumophila IpxB genes suggests that both paralogues

are involved in lipid A biosynthesis. To test for their function as lipid A disaccharide

synthases, a complementation assay was performed. Since deletion of IpxB is lethal for

E. coli, the temperature sensitive E. coli IpxB mutant strain MN7 was used (Nishijima et al.,

1981). The strain was transformed with 1. pneumophila IcsC/lpxBI or IpxB2 expression

vectors or control vectors and grown at 30 QC (permissive temperature) or 42 QC (non

permissive temperature) for 24 h. Under these conditions, expression of E. coli IpxB (pSR8)

70

Page 72: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

restored growth at 42 QC to a level similar to that at 30 QC. Expression of 1. pneumophila

IcsC/lpxBl or IpxB2 complemented growth at the non-permissive temperature (Table 3),

indicating that both 1. pneumophila lpxB paralogues function as lipid A disaccharide

synthases during biosynthesis of lipid A. The 1. pneumophila enzymes only partially

complement E. coli IpxB. This is likely due to the structural difference between

1. pneumophila and E. coli lipid A, causing a reduced efficiency of lipid A synthesis. The

sugar backbone of 1. pneumophila lipid A consists of diamino-dideoxy-glucopyranose

instead of glucosamine, and the primary fatty acid chains differ considerably in length (C20 or

C14, respectively). Furthermore, expression of E. coli IpxB was fully induced, whereas

expression of the 1. pneumophila IpxB paralogues remained restricted to the basal level of the

Ptac promoter to avoid toxic overexpression effects. It might be that the the amount of the

1. pneumophila LpxB enzymes was not sufficient.

Plasmid Dilution Number of colonies after 24 hrs incubation

30°C 42°C

pUC18 10-6 411 0

pSR8 10-6 416 301

pMMB207C 10-1 >2000 0

pMMB207C-lcsC 10-1 >2000 220

pMMB207C-lpxB 10-1 >2000 330

Table 3: Complementation of the temperatnre sensitive E. coli lpxB mutant MN7 withL. pneumophila lcsC and lpxB. MN7 transformed with the plasmids indicated was plated in differentdilutions and grown at 30 QC (permissive temperature) and 42 QC (non permissive temperature). After24 hours incubation were the number of cfu determined. pUC18: empty vector, pSR8: expressionvector for E. coli lpxB, pMMB207C: empty vector, pMMB207C-lcsC; -lpxB: expression vectors forL. pneumophila lcsC or lpxB, respectively.

3.4.3 Cytotoxicity of L. pneumophila IpxB paralogues for A. castellanii

Since IcsC/lpxBl was identified due to its ability to render bacteria cytotoxic for

A. castellanii, we tested whether the expression of lpxB2 is also toxic for amoebae.

A. castellanii was infected with an 1. pneumophila icmG mutant strain expressing either of

the IpxB paralogues, and cytotoxicity was determined by propidium iodide (PI) uptake 2 d

71

Page 73: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

post infection. Under these conditions, overexpression of IcsC, but not IpxB, was cytotoxic

for A. castellanii, without enhancing intracellular replication of the bacteria within the

amoebae (data not shown; Albers et al., 2005). While the expression of IcsCllpxBl in an

icmG mutant background killed approximately 35% of the amoebae within 2 d, expression of

IpxB2 did not increase cytotoxicity above background level (Figure 2). This finding suggests

that the two L. pneumophila IpxB paralogues are functionally not equivalent.

100

90'-'-r:: 80.!-.e 70fJCJ 60

oc;(50

CD>:.= 40';0 30c.Q: 20:::e0

10

0icmG-/pBCR icmG-/plcsC icmG-/plpxB icmG-/picmGCD

Figure 2: Cytotoxicity of lpxB paralogues on A. castellanii. Amoebae were infected with an MOl of50 and cytotoxicity of the different strains was assayed 2 days p.i. by propidium iodide staining.icmG-/pBCR: .:iicmG/pMMB207C-RBS, icmG-/plcsC: .:iicmG/pMMB207C-RBS-lcsC, icmG-/plpxB:.:iicmG/pMMB207C-RBS-lpxB, icmG-/pGCD: McmG/pGS-Lc-63-14 (genomic fragment containingicmGCD in pMMB207C-Km14).

Upon induction in exponential growth phase with IPTG, IcsCllpxBl but not IpxB2 expression

reduced the growth rate of liquid cultures. On agar plates, overexpression of IpxB2 was

apparently more cytotoxic than IcsCllpxBI, as lower concentrations ofIPTG were required to

inhibit bacterial growth on plates (data not shown). The distinct effects of overexpression

give further evidence for functional differences between LcsC/LpxB1 and LpxB2.

72

Page 74: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

3.4.4 Expression of L. pneumophila Ipx genes during growth in acomplex medium

Both 1. pneumophila IpxB paralogues are located adjacent to other lipid A biosynthesis genes

in the genome: IpxB2 (lpg2945) lies next to IpxD2 (lpg2944) and IpxA2 (lpg2943), but is

encoded on the opposite strand and, therefore, does not form an operon with other genes.

IcsC/lpxBl (lpg1371) is located immediately downstream of gnnA (lpgI372), encoding the

putative first enzyme of the pathway. To examine whether the two genes are cotranscribed,

RNA was isolated, and a RT-PCR was performed using primers hybridizing to gnnA and to

IcsC/lpxBl, respectively. The presence of a PCR product demonstrates that gnnA and

IcsC/lpxBl are cotranscribed (Figure 3), supporting the idea that both enzymes belong to the

same biosynthetic pathway.

genomic MW ~ RT + RT

Figure 3: peR analysis of cotranscription of IcsC and gnnA. RNA from bacteria in exponentialgrowth phase was assayed by reverse transkription and peR with primers binding in gnnA and lesC.Genomic: control with genomic DNA as template, MW: molecular weight marker, -/+ RT: absence orpresence of reverse transcriptase during cDNA synthesis.

The existence of multiple paralogues of genes that are predicted to modulate the acylation

state of lipid A suggests that L. pneumophila has the potential to synthesize structurally

different lipid A molecules in response to different environmental conditions. To test whether

1. pneumophila lipid A biosynthesis genes are differentially transcribed, we examined the

expression levels of the genes by RT-PCR under different conditions. Samples of the PCR

reaction were taken after different cycle numbers and the amount of product was assessed on

an agarose gel. Thereby, the time point of appearance and the strength of a band correspond

to the amount of mRNA in a sample.

The method was validated using genes with known expression profiles during different

growth phases in liquid medium: rpoA encodes the a-subunit of RNA polymerase, which is

maximally expressed in logarithmic growth phase and slightly downregulated in stationary

phase (Bruggemann et al., 2006). 16S ribosomal RNA was used as a control for RNA present

constitutively at high levels.jlaA encodes the major flagellum component (flagellin), which is

strongly upregulated during stationary growth phase, corresponding to the observation that

73

Page 75: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

___________T_h_e_c"---yt_o_to_xi~~!ipidA disaccharide synthase E't,~alogue LcsC

L. pneumophila becomes motile upon entry into stationary phase (Byrne and Swanson, 1998;

Heuner et al., 1999). As expected, rpoA was expressed constitutively in all growth phases,

but slightly downregulated in late stationary phase, 16S rRNA was abundant and expressed

constitutively, and the expression level offlaA was lower than that of rpoA, but upregulated

at late stationary phase (Figure 4). The expected regulation patterns demonstrate that the

method is suitable for gene expression analysis in 1. pneumophila. In absence of reverse

transcriptase, faint bands of the target genes were visible only after 32 PCR cycles, indicating

that the amount of contaminating DNA in the assay was low.

Exp- +

rpoAES LS

- + - +

168 rRNAG Exp ES LS G Exp

-+-+ -+ -+

flaAES LS- + - +

Greverse transcriptase

20 cycles

26 cycles

32 cycles

Figure 4: RT-peR analysis of expression in different growth phases in liquid cultures. Geneswith known expression profiles were examined to validate the method. rpoA, the a subunit of theRNA polymerase, is maximally expressed in logarithmic growth phase and moderatelydownregulated at late stationary growth phase, 16S rRNA is constitutively expressed at very highlevels and flaA is upregulated during stationary phase. Exp: exponential phase (OD600 0.8), ES: earlystationary phase (OD600 2.4), LS: late stationary phase (00600 3.4), G: genomic DNA as peRtemplate.

The expression levels of the IpxB and IpxA paralogues were examined in exponential, early

stationary and late stationary growth phase, respectively (Figure 5). The amount of

IcsCllpxBI and IpxB2 RNA was low in all growth phases, and expression of IcsC/lpxBl was

further reduced in late stationary growth phase. The two IpxA paralogues were expressed at

comparable levels, preferentially in exponential growth phase and more strongly than the

IpxB paralogues. Since the IpxB and IpxA paralogues are synchronously expressed during

growth of L. pneumophila in a rich medium, the genes seem to be used in parallel under these

conditions.

74

Page 76: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

___________"""~hapter3

IcsC IpxB2

Exp ES LS G Exp ES LSrev. Tr.: - + - + - + ~ + ~ +-+

IpxA1

G Exp ES LS• + " + - +

IpxA2G Exp ES LS G

_ + ~ + M +

20 cycles

26 cycles

32 cycles

Figure 5: Gene expression of IpxB and [pxA paralogues in different growth phases in liquidmedium. RNA was extracted from bacteria grown in liquid medium to different growth phases andexpression levels were assayed by reverse transkription and peR. Exp: exponential growth phase,(OD600 0.8, 6 hours incubation), ES: early stationary phase (OD600 2.4, 16 hours), LS: late stationaryphase (OD600 3.4, 26 hours), G: peR from genomic DNA.

3.4.5 Expression of L. pneumophila Ipx genes under osmotic stress andnutrient deprivation

1. pneumophila might adapt to osmotic stress or nutrient deprivation by modulating the

expression of lipid A biosynthesis genes, and consequently, altering its lipid A structure. To

test this hypothesis, an 1. pneumophila liquid culture in stationary growth phase (OD600 3.3)

was split and either suspended in distilled water or kept in AYE medium. 1. pneumophila

tolerates exposure to distilled water, and differences in gene expression were examined four

hours after incubation at room temperature. To assay effects of osmotic stress or nutrient

deprivation on the expression of lpx genes, bacteria in stationary growth phase were chosen,

since incubation of exponentially growing bacteria in water induces a transition from

exponential to stationary phase, leading to downregulation of lpx genes (Figure 5). The

effects of growth phase transition would likely mask effects caused by osmotic stress or

nutrient deprivation.

While expression of flaA was not affected upon transition of 1. pneumophila from rich

medium to distilled water, IcsCllpxBI and IpxAI were upregulated and IpxB2 was slightly

downregulated under these conditions (Figure 6). The expression of IpxA2 and IpxC in

stationary growth phase was very low upon incubation of 1. pneumophila in distilled water.

The inverse regulation of IcsCllpxBI and IpxB2 and the upregulation of IpxA 1 suggest that

under osmotic stress or nutrient deprivation L. pneumophila might employ IcsCllpxB1 and

IpxAI to modulate its lipid A structure.

75

Page 77: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

168 rRNA

AYE H20 Grev. Tr.. - + - +

168 rRNA

AYE H20 Grev, Tr.; - + - +

flaA

IpxA1

AYE H20 G~ + - +

lesC

IpxA2

AYE H20 G- + - +

IpxB2

AYE H20 G+ - +

IpxC

AYE H20 Gw... _ +

20 cycles

26 cycles

32 cycles

20 cycles

26 cycles

32 cycles

Figure 6: Gene expression under osmotic stress and nutrient deprivation. L. pneumophila fromstationary phase liquid cultures were split and either incubated in A YE medium or in distilled waterfor 4 hours at room temperature. RNA was extracted and expression levels were assayed by reversetranscription and PCR. G: PCR from genomic DNA.

3.4.6 Expression of L. pneumophila Ipx genes during intracellulargrowth in A. castellanii

Pathogenic bacteria respond to the challenges of the intracellular environment of phagocytes

by modulating their lipid A (Miller et al., 2005). To examine lpx gene expression during

residence in a phagocyte, A. castellanii amoebae were infected with 1. pneumophila, and

bacterial gene expression was analyzed shortly after infection (4 h) and after 24 h. As RNA

was extracted from infected amoeba, it is from both bacteria and amoeba. Due to intracellular

bacterial replication, the proportion of bacterial RNA is higher in the 24 h sample compared

to the 4 h sample. Therefore, the method used here allows comparison of different genes

within the same RNA preparation (at a given time point), but does not allow direct

comparison of expression levels at different time points of sampling.

Interestingly, at 4 h post infection, IpxAl and IcsC/lpxBl were much stronger expressed than

IpxA2 and IpxB2 (Figure 7). The expression level of IpxAl was similar to IpxC, a gene with

no paralogues in the L. pneumophila genome. 24 hours after infection, all lpx paralogues

tested were expressed abundantly and at comparable levels. This finding suggests that while

not detectable at 4 h post infection, lpxA2 and IpxB2 become more important at later stages of

an infection. Taken together, the data suggest that during early stages of an infection IpxAl

and IcsC/lpxBl are predominantly expressed, whereas in late stages of infection lpxA2 and

76

Page 78: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

IpxB2 expression increases. The IpxA 1 and IcsC/lpxBl genes were also expressed with a

similar pattern upon incubation of the bacteria in distilled water (Figure 6), suggesting that

the genes might be co-regulated.

4h 24h G 0 4h 24h G 0

168 rRNAo 4h 24h G 0

IpxA1 IpxA2 IpxC4h 24h G

rev,Tr,: - ... - ... - ... - + • + - + ~+-+~+ - + - + ~ +

22 cycles

29 cycles

36 cycles

Figure 7: Expression of lipid A biosynthesis genes during intracellular growth in A. castel/anii.RNA from un infected A. castellanii amoebae (0) or from amoebae 4 or 24 hours post infection (4h,24h) was extracted and expression levels were assayed by reverse transkription and PCR. RNApreparations from different time points contain different proportions of RNA from amoebae andbacteria, and therefore, expression levels can be directly compared only within identical RNApreparations prepared at the same time post infection. G: PCR from genomic DNA.

3.5 DISCUSSION

3.5.1 Lipid A biosynthesis paralogues and their functions

The identification of the cytotoxic IpxB paralogue IcsC (Albers et al., 2005) prompted us to

analyze the lipid A biosynthesis pathway in 1. pneumophila. A search for genes homologous

to E. coli and A. ferrooxidans lipid A biosynthesis genes revealed that 1. pneumophila

harbors genes required for the conversion of GIcNAc to GIcNAcN3, and multiple copies of

the acyl transferases IpxA, IpxD, and IpxL, as well as the lipid A disaccharide synthase IpxB.

Both IpxB paralogues partially complement an E. coli conditionallpxB mutant, indicating that

the 1. pneumophila IpxB genes indeed function as lipid A disaccharide synthases (Table 3).

However, the enzymes are distinct, as overexpression of IcsC/lpxBl but not IpxB2 is

cytotoxic for amoebae (Figure 2). Interestingly, all of the genes existing in more than one

77

Page 79: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

copy in the 1. pneumophila genome are presumably involved in lipid A acylation, or in case

of IpxB, might process differentially acylated substrates.

In E. coli, LpxA functions as an accurate hydrocarbon ruler that incorporates 14-carbon acyl

chains two orders of magnitude faster than 12- or 16-carbon acyl chains, and LpxA from

P. aeruginosa specifically adds 10-carbon acyl chains (Raetz and Whitfield, 2002). The

1. pneumophila LpxA and LpxD paralogues likely also have different substrate specificities

and might preferentially use as substrates fatty acids with distinct chain length, branches or

other modifications. The two lipid A disaccharide synthases then presumably condense the

different substrates generated by the LpxA and LpxD paralogues, resulting in lipid IVA

moieties that vary in primary fatty acid composition. The three 1. pneumophila LpxL

paralogues possibly catalyze the transfer of different acyl chains. E. coli LpxL and the cold

shock-induced acyl transferase LpxP both catalyze the addition of secondary fatty acids to the

~-hydroxyl groups of the primary acyl chains (Raetz and Whitfield, 2002). Lipid A of

1. pneumophila contains CI6 and C28 secondary fatty acids attached to position 2' and 3'

(Kooistra et al., 2002a) and it is conceivable that the three LpxL homologues and the pagP

like gene rep enable 1. pneumophila to vary also the composition of the secondary fatty

acids. In summary, the presence of multiple lpx paralogues suggests that L. pneumophila

synthesizes lipid A molecules modified by a variety of different primary and secondary acyl

chains.

Synthesis of lipid A with acyl chains of different length has been shown for L. pneumophila

serogroup 1 subgroup OLDA. This strain undergoes phase variation based on excision and

insertion of a 30 kb unstable element (Luneberg et al., 2001). In the phase variant strain, the

length of the primary 3-hydroxylated fatty acids attached to position 2 and 2' of the GlucN3N

disaccharide is shifted from C20 to shorter chains of CI6 or C18, while the lipid A backbone

and the phosphorylation pattern remain unaffected (Kooistra et aI., 2002a). The three

different LpxD paralogues are candidate enzymes to catalyze these modifications.

3.5.2 Differential regUlation of L. pneumophila Ipx paralogues

As an adaption to different niches, 1. pneumophila might produce distinct lipid A structures

under different environmental conditions. Therefore, we examined whether transcription of

lpx paralogues is differentially regulated. During growth in nutrient broth, there was no large

difference between the expression of IpxAl and IpxA2 or between expression of lesC/lpxBl

78

Page 80: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

and IpxB2 (Figure 5). However, upon transfer of 1. pneumophila from nutrient broth into

distilled water, expression of lesC/lpxBl and IpxAl was upregulated, while expression of

IpxB2 was downregulated (Figure 6). This regulation pattern suggests a role for LcsC/LpxB1

and LpxA1 in 1. pneumophila exposed to salt- and nutrient free medium, possibly mimicking

a nutritionally poor extracellular environment.

Shortly after infection of A. eastellanii by 1. pneumophila, IpxAI and lesC/lpxBl are stronger

expressed than their paralogues IpxA2 and IpxB2, respectively (Figure 7). As expression of

lesC/lpxBI and IpxAl was also upregulated upon transfer of 1. pneumophila to water, these

paralogues might represent lipid A biosynthesis genes required during the

transmissive/infective phase of 1. pneumophila (Molofsky and Swanson, 2004). Contrarily,

24 h post infection, IpxAl and IpxA2, as well as lesC/lpxBl and IpxB2 were expressed at a

similar level, indicating a role in lipid A biosynthesis for alllpx paralogues at later stages of

the infection in the replicative phase of1. pneumophila. In agreement with this notion, the lpx

paralogues were also expressed at similar levels during growth in nutrient broth (Figure 5). In

summary, 1. pneumophila expresses distinct lpx paralogues under specific growth and

environmental conditions, suggesting that the lipid A structure is modified as an adaption to a

changing environment.

3.5.3 Biological implications of lipid A and LPS modifications

Lipid A modifications profoundly effect various interactions between pathogenic bacteria and

their environment, including host cells and organisms. A prominent example is Salmonella

enteriea serovar Typhimurium, which, dependent on PhoP-PhoQ and other two-component

regulatory systems, regulates a number of lipid A-modifying enzymes (Miller et aI., 2005).

The acyl transferase PagP is synthesized under low magnesium concentrations encountered

inside a phagosome and adds an additional palmitate to the primary acyl chain at position 2 of

lipid A, thus increasing the resistance of the bacteria to cationic antimicrobial peptides

(CAMPs) (Bishop et al., 2000; Guo et al., 1998). Interestingly, L. pneumophila harbors the

pagP-like gene rep, confering resistance to CAMPs in low magnesium medium (Robey et al.,

2001). Similar to a pagP mutant, an rep mutant showed decreased resistance to CAMPs.

Moreover, the rep mutant was defective for growth in Hartmanella vermiformis amoebae and

U937 macrophages, as well as for lung colonization of AlJ mice.

79

Page 81: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

Structural modifications of lipid A may also significantly affect the course of an infection in

multicellular host organisms. Lipid A is the active part of endotoxin, which is recognized by

the innate immune system by binding to the CD14 receptor and toll-like receptor (TLR) 4

(Miller et al., 2005; Raetz and Whitfield, 2002). 1. pneumophila LPS fails to bind CD14,

probably due to the unusually long and branched fatty acid residues of its lipid A (Neumeister

et al., 1998). The unusual composition of 1. pneumophila lipid A fatty acids, possibly

attached by the different lpx paralogues, might help the bacteria to escape recognition by the

innate immune system.

Recently, LPS-containing vesicles released from the outer membrane of growing

1. pneumophila were shown to inhibit phagosome-lysosome fusion in infected macrophages

independently of the Icm/Dot secretion system (Femandez-Moreira et al., 2006).

Interestingly, only vesicles derived from the transmissive/infective phase but not the

replicative phase inhibited phagosome-lysosome fusion. The structure of LPS was regulated

in a growth phase dependent manner, indicating that LPS modifications might be involved in

altering vesicle trafficking. Intracellular distribution of LPS during infection and an impact of

intracellular LPS on vesicle trafficking were also shown for Legionella jeonii infecting

Acanthamoeba proteus. LPS released from the bacteria inserted into the LCV membrane and

assisted in preventing fusion of the LCV with lysosomes (Kim et al., 1994). It is conceivable

that modifications of the membrane anchor lipid A affect the interactions of LPS with host

cell functions and thus modulate the course of infection.

In summary, due to the involvement of lipid A in protection, immune recognition and

probably also in intracellular life, modification of its structure contributes to the adaption of

1. pneumophila to its diverse niches. It will be of great interest to identify the biochemical

functions of the different lpx paralogues, the structure of lipid A molecules synthesized by the

paralogues and the mechanisms by which the different lipid A variants interact with host

organisms.

3.6 ACKNOWLEDGEMENTS

This work was supported by grants from the Swiss National Science Foundation (631­

065952), the ETH Zurich (17/02-3), the Swiss Commission for Technology and Innovation

(CTIIKTI, 6629.2 BTS-LS) and the Federal Office for Energy (BFE).

80

Page 82: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

3.7 REFERENCES

Albers, u., Reus, K., Shuman, H.A, and Hilbi, H. (2005) The amoebae plate test implicates aparalogue of IpxB in the interaction of Legionella pneumophila with Acanthamoebacastellanii. Microbiology 151: 167-182.

Altschul, S.F., Madden, T.L., Schaffer, AA., Zhang, J., Zhang, Z., Miller, W., and Lipman,DJ. (1997) Gapped BLAST and PSI-BLAST: a new generation of protein databasesearch programs. Nucleic Acids Res 25: 3389-3402.

Bishop, R.E., Gibbons, H.S., Guina, T., Trent, M.S., Miller, S.L, and Raetz, C.R. (2000)Transfer of palmitate from phospholipids to lipid A in outer membranes of gram­negative bacteria. Embo J 19: 5071-5080.

Bruggemann, H., Hagrnan, A., Jule, M., Sismeiro, 0., Dillies, M., Gouyette, C., Kunst, F.,Steinert, M., Heuner, K., Coppee, J., and Buchrieser, C. (2006) Virulence strategiesfor infecting phagocytes deduced from the in vivo transcriptional program ofLegionella pneumophila. Cellular Microbiology 0: O.

Byme, B., and Swanson, M.S. (1998) Expression of Legionella pneumophila virulence traitsin response to growth conditions. Infect Immun 66: 3029-3034.

Cazalet, e., Rusniok, C., Bruggemann, H., Zidane, N., Magnier, A., Ma, L., Tichit, M.,Jarraud, S., Bouchier, C., Vandenesch, F., Kunst, F., Etienne, J., Glaser, P., andBuchrieser, C. (2004) Evidence in the Legionella pneumophila genome forexploitation of host cell functions and high genome plasticity. Nat Genet 36: 1165­1173.

Chen, 1., de Felipe, K.S., Clarke, M., Lu, H., Anderson, O.R., Segal, G., and Shuman, H.A.(2004) Legionella effectors that promote nonlytic release from protozoa. Science 303:1358-1361.

Chien, M., Morozova, 1., Shi, S., Sheng, H., Chen, 1., Gomez, S.M., Asamani, G., Hill, K.,Nuara, J., Feder, M., Rineer, 1., Greenberg, JJ., Steshenko, V., Park, S.H., Zhao, B.,Teplitskaya, E., Edwards, 1.R., Pampou, S., Georghiou, A, Chou, Le., Iannuccilli,W., VIz, M.E., Kim, D.H., Geringer-Sameth, A., Goldsberry, C., Morozov, P.,Fischer, S.G., Segal, G., Qu, x., Rzhetsky, A., Zhang, P., Cayanis, K, De Jong, PJ.,Ju, J., Kalachikov, S., Shuman, H.A., and Russo, J.J. (2004) The genomic sequence ofthe accidental pathogen Legionella pneumophila. Science 305: 1966-1968.

Crowell, D.N., Anderson, M.S., and Raetz, C.R. (1986) Molecular cloning of the genes forlipid A disaccharide synthase and UDP-N-acetylglucosamine acyltransferase inEscherichia coli. J Bacteriol168: 152-159.

Feeley, J.C., Gibson, RJ., Gorrnan, G.W., Langford, N.C., Rasheed, J.K., Mackel, D.C., andBaine, W.B. (1979) Charcoal-yeast extract agar: primary isolation medium forLegionella pneumophila. J Clin Microbioll0: 437-441.

Femandez-Moreira, E., Helbig, 1.H., and Swanson, M.S. (2006) Membrane vesicles shed byLegionella pneumophila inhibit fusion of phagosomes with lysosomes. Infect Immun74: 3285-3295.

Fields, B.S. (1996) The molecular ecology of Legionellae. Trends Microbiol4: 286-290.

81

Page 83: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

Guo, L., Lim, K.R, Poduje, C.M., Daniel, M., Gunn, J.S., Hackett, M., and Miller, S.L(1998) Lipid A acylation and bacterial resistance against vertebrate antimicrobialpeptides. Cell 95: 189-198.

Heuner, K., Brand, RC., and Hacker, J. (1999) The expression ofthe flagellum of Legionellapneumophila is modulated by different environmental factors. FEMS Microbiol Left175: 69-77.

Hilbi, H., Segal, G., and Shuman, H.A. (2001) Icm/Dot-dependent upregulation ofphagocytosis by Legionella pneumophila. Mol Microbiol42: 603-617.

Kim, KJ., Na, YE., and Jeon, K.W. (1994) Bacterial endosymbiont-derivedlipopolysaccharides and a protein on symbiosome membranes in newly infectedamoebae and their roles in lysosome-symbiosome fusion. Infect Immun 62: 65-71.

Knirel, Y.A., Rietschel, KT., Marre, R, and Zahringer, U. (1994) The structure of the 0­specific chain of Legionella pneumophila serogroup 1 lipopolysaccharide. Eur JBiochem 221: 239-245.

Kooistra, 0., Knirel, YA., LUneberg, E., Frosch, M., and Zahringer, U. (2002a) Phasevariation in Legionella pneumophila serogroup I, subgroup OLDA, strain RClinfluences lipid A structure. In LegionelZa: Proceedings ofthe 5th internationalConference. Marre, R (ed)., Washington, D.C.: ASM Press.

Kooistra, 0., Luneberg, E., Knirel, YA., Frosch, M., and Zahringer, U. (2002b) N­Methylation in polylegionaminic acid is associated with the phase-variable epitope ofLegionella pneumophila serogroup 1 lipopolysaccharide. Identification of 5-(N,N­dimethylacetimidoyl)amino and 5-acetimidoyl(N-methy1)amino-7-acetamido-3 ,5,7,9­tetradeoxynon-2-ulosonic acid in the O-chain polysaccharide. Eur J Biochem 269:560-572.

Luneberg, E., Zahringer, U., Knirel, YA., Steinmann, D., Hartmann, M., Steinmetz, 1.,Rohde, M., Kohl, J., and Frosch, M. (1998) Phase-variable expression oflipopolysaccharide contributes to the virulence of LegionelZa pneumophila. J Exp Med188: 49-60.

Luneberg, E., Mayer, B., Daryab, N., Kooistra, 0., Zahringer, u., Rohde, M., Swanson, l,and Frosch, M. (2001) Chromosomal insertion and excision ofa 30 kb unstablegenetic element is responsible for phase variation of lipopolysaccharide and othervirulence determinants in Legionella pneumophila. Mol Microbioi 39: 1259-1271.

Mampel, J., Spirig, T., Weber, S.S., Haagensen, lA., Mo1in, S., and Hilbi, H. (2006)Planktonic replication is essential for biofilm formation by Legionella pneumophila ina complex medium under static and dynamic flow conditions. Appl Environ Microbiol72: 2885-2895.

Miller, S.L, Ernst, RK., and Bader, M.W. (2005) LPS, TLR4 and infectious disease diversity.Nat Rev Microbiol3: 36-46.

Moffat, IF., and Tompkins, L.S. (1992) A quantitative model of intracellular growth ofLegionella pneumophila in Acanthamoeba castellanii. Infect Immun 60: 296-301.

Moll, H., Sonesson, A., Jantzen, E., Marre, R., and Zahringer, U. (1992) Identification of27­oxo-octacosanoic acid and heptacosane-1,27-dioic acid in Legionella pneumophila.FEMS Microbiol Left 76: 1-6.

82

Page 84: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 3

Molofsky, A.B., and Swanson, M.S. (2004) Differentiate to thrive: lessons from theLegionella pneumophila life cycle. Mol Microbiol53: 29-40.

Murga, R, Forster, T.S., Brown, E., Pruckler, J.M., Fields, B.S., and Donlan, RM. (2001)Role of biofilms in the survival ofLegionella pneumophila in a model potable-watersystem. Microbiology 147: 3121-3126.

Nash, T.W., Libby, D.M., and Horwitz, M.A. (1984) Interaction between the Legionnaires'disease bacterium (Legionella pneumophila) and human alveolar macrophages.Influence of antibody, lymphokines, and hydrocortisone. JClin Invest 74: 771-782.

Neumeister, B., Faigle, M., Sommer, M., Zahringer, u., Stelter, F., Menzel, R, Schutt, C,and Northoff, H. (1998) Low endotoxic potential of Legionella pneumophilalipopolysaccharide due to failure of interaction with the monocyte lipopolysaccharidereceptor CD14. Infect Immun 66: 4151-4157.

Nishijima, M., Bulawa, C.E., and Raetz, C.R (1981) Two interacting mutations causingtemperature-sensitive phosphatidylglycerol synthesis in Escherichia coli membranes.J Bacterioll45: 113-121.

Purcell, M., and Shuman, RA (1998) The Legionella pneumophila icmGCDJBF genes arerequired for killing of human macrophages. Infect Immun 66: 2245-2255.

Raetz, CR., and Whitfield, C. (2002) Lipopolysaccharide endotoxins. Annu Rev Biochem 71:635-700.

Robey, M., O'Connell, W., and Cianciotto, N.P. (2001) Identification of Legionellapneumophila rep, a pagP-like gene that confers resistance to cationic antimicrobialpeptides and promotes intracellular infection. Infect Immun 69: 4276-4286.

Sadosky, AB., Wiater, L.A, and Shuman, H.A. (1993) Identification of Legionellapneumophila genes required for growth within and killing of human macrophages.Infect Immun 61: 5361-5373.

Segal, G., Purcell, M., and Shuman, H.A. (1998) Host cell killing and bacterial conjugationrequire overlapping sets of genes within a 22-kb region of the Legionellapneumophila genome. Proc Natl Acad Sci USA 95: 1669-1674.

Segal, G., and Shuman, H.A (1999) Legionella pneumophila utilizes the same genes tomultiply within Acanthamoeba castellanii and human macrophages. Infect Immun 67:2117-2124.

Sporri, R., Joller, N., A1bers, D., Hilbi, H., and Oxenius, A. (2006) MyD88-dependent IFN­gamma production by NK cells is key for control of Legionella pneumophilainfection. J Immunoll76: 6162-6171.

Steinert, M., Hentsche1, D., and Hacker, J. (2002) Legionella pneumophila: an aquaticmicrobe goes astray. FEMS Microbiol Rev 26: 149-162.

Sweet, CR., Ribeiro, AA., and Raetz, C.R. (2004) Oxidation and transamination of the 3"­position ofDDP-N-acety1g1ucosamine by enzymes from Acidithiobacillusferrooxidans. Role in the formation of lipid a molecules with four amide-linked acylchains. J Biol Chem 279: 25400-25410.

Vogel, J.P., Andrews, H.L., Wong, S.K., and Isberg, R.R. (1998) Conjugative transfer by thevirulence system ofLegionella pneumophila. Science 279: 873-876.

83

Page 85: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

The cytotoxic lipid A disaccharide synthase paralogue LcsC

Yamamoto, Y., Klein, T.W., Newton, c.A., Widen, R., and Friedman, H. (1988) Growth ofLegionella pneumophila in thioglycolate-elicited peritoneal macrophages from A/Jmice. Infect Immun 56: 370-375.

Yanisch-Perron, C., Vieira, J., and Messing, J. (1985) Improved Ml3 phage cloning vectorsand host strains: nucleotide sequences of the Ml3mp18 and pUC19 vectors. Gene 33:103-119.

Zahringer, u., Knirel, Y.A., Lindner, B., Helbig, J.H., Sonesson, A., Marre, R., and Rietschel,E.T. (1995) The lipopolysaccharide of Legionella pneumophila serogroup 1 (strainPhiladelphia 1): chemical structure and biological significance. Prog Clin Bioi Res392: 113-139.

84

Page 86: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

4 L. pneumophila IcmG increases the efficiency ofeffector protein secretion

4. 1 Introduction

1. pneumophila is a Gram-negative bacterium that can be found ubiquitously in the

environment. It survives and multiplies mainly intracellularly in various protozoa, where it

manipulates the host cell vesicular machinery to prevent phagosome-lysosome fusion and to

establish a replication permissive vacuole (Fields, 1996; Roy and Tilney, 2002). Upon

transmission into the human lung, the bacteria are phagocytosed by alveolar macrophages,

replicate within the cells and finally kill the macrophages, thereby causing the life­

threatening pneumonia known as Legionnaire's disease (Nash et al., 1984).

Efficient phagocytosis and intracellular replication of 1. pneumophila depend on a type IV

secretion system encoded by the icm/dot (intracellular multiplication/defective in organelle

trafficking) genes (Hilbi et aI., 2001; Segal et aI., 1998; Vogel et al., 1998). The icm/dot

genes are located within the Legionella genome in two clusters: Region I contains the

dotDCB and dotA-icmVWX genes, and Region II comprises 18 genes, including icmG. The

gene clusters were identified by transposon mutagenesis (Purcell and Shuman, 1998; Segal

and Shuman, 1999), and most of the icm/dot genes are absolutely required for intracellular

growth and killing of human macrophages and the protozoan host Acanthamoeba castellanii.

Mutation of the icmG, icmS or icmF gene led to only slight defects in the ability to grow

within and kill macrophage-like HL-60 cells but abolished growth in amoebae (Segal and

Shuman, 1999). In contrast to the majority of icm/dot mutants, the icmG and icmS mutant are

capable to survive and grow on agar plates in presence of amoebae, albeit with a strong

growth defect compared to wild type bacteria. The icmG mutant also shows a partial

phenotype with respect to cytotoxicity for A. castellanii (Albers et al., 2005).

The IcmG protein contains a predicted transmembrane and a coiled-coil domain (Figure 1). A

bacterial two-hybrid screen revealed that the part of IcmG spanning amino acids 28 to 123

specifically interacts with 1. pneumophila effector proteins, namely RalF and the Sid effector

proteins (Luo and Isberg, 2004).

Morozova and coworkers performed a comparative sequence analysis of icm/dot genes and

predicted a putative SNARE domain in IcmG at amino acids 142 to 210 (Morozova et al.,

2004). SNARE proteins (soluble NSF attachment receptor; NSF: N-ethyl-maleimide-

85

Page 87: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

sensitive fusion protein) form a family of proteins that mediate intracellular membrane fusion

events in eukaryotic cells (Chen and Scheller, 200 I). Modification of vesicular trafficking is

a prerequisite for 1. pneumophila to generate a replication permissive vacuole, and interfering

with SNARE-mediated vesicle fusion could be a method to alter host cell vesicle trafficking.

A SNARE domain was also predicted in the corresponding region of Coxiella burnetii IcmG,

the only known protein showing sequence similarities to Legionella lemG.

~,,-----r'---'""""T'-I=•••d!"ilrlld11111@IIt·r'T-r-r-'---""'--T'----hydrophilic TM coiled-coil

Effector binding SNARE homology

50I I

100I I

150

I I

200

I I

250

I I aa

IcmG (269 aa)Figure 1: In silico aualysis of IcmG. The protein sequence was analyzed as described in Materialand Methods. The binding region for effector proteins was described in Luo and Isberg, 2004. Thepresence of a putative SNARE domain was stated by Morozova et al., 2004. TM: Transmembranedomain.

The icmG mutant was recently used in a screen to identify genes involved in virulence of

1. pneumophila (Albers et al., 2005). In the "amoebae plate test", the ability of bacteria was

examined to grow and survive on agar plates in the presence of the phagocytic amoeba

A. castellanii. An 1. pneumophila chromosomal library was introduced into !i.icmG and

screened in the amoebae plate test for plasmids suppressing the partial growth defect of the

icmG mutant. In this screen, a paralogue of the E. coli lipid A disaccharide synthase IpxB was

identified and termed lcsC (Legionella cytotoxic suppressor), since overexpression of lcsC in

!i.icmG rendered the bacteria more cytotoxic for the amoebae. Surprisingly, overexpression of

IcsC in other 1. pneumophila mutants showing partial growth defects in the amoebae plate

test or in intracellular growth assays (e.g., !i.icmS, !i.icmF, !i.rpoS) did not render the bacteria

more cytotoxic (see Chapter 2, Albers et al., 2005).

The restriction of lcsC mediated cytotoxicity to the icmG mutant and the possibility of IcmG

acting as a SNARE protein prompted us to examine the function of IcmG in more detail to

better understand its role in infection and LcsC-mediated cytotoxicity. We show that IcmG is

expressed at low levels in 1. pneumophila, is most likely not translocated during infection

and that an icmG mutant strain retains the capability to translocate effector proteins but is

impaired in the ability to recruit ER derived vesicles. The data suggest that IcmG is part of

86

Page 88: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

the lem/Dot secretion system and acts as a coupling protein supporting the release of effector

proteins.

4.2 Materials and methods

4.2.1 Cultivation of cells

The 1. pneumophila wild type strain used was Philadelphia-l serogroup I JR32 (Sadosky et

al., 1993). The icmG mutant strain MW635 (Purcell and Shuman, 1998) is an isogenic mutant

containing a kanamycin resistance gene inserted in the icmG gene, and GS3011 is an

avirulent mutant containing a kanamycin resistance gene insertion in the icm/dot transporter

gene icmT (Segal and Shuman, 1998). 1. pneumophila was grown on charcoal yeast extract

(CYE) plates (Feeley et al., 1979) and in AYE liquid medium (Horwitz and Silverstein,

1983). Plasmids were maintained by addition of 5 Ilg/ml chloramphenicol. E. coli TOPlO

(lnvitrogen) was used for cloning and E. coli BL21 (DE3) (Novagen) was used for expression

of GST and GST fusion proteins. D. discoideum amoebae (Hagele et al., 2000; Solomon et

al., 2000) were grown axenically at 23 QC in culture flasks in HL5 medium (Weber et al.,

2006). RAW246.7 macrophages were cultivated in RPMI1640 supplemented with 10% FCS

and 2 mM L-glutamine at 37 QC in 5% CO2.

4.2.2 Sequence analysis of IcmG

Hydrophobicity was analyzed with the ProtScale software (www.expasy.org). The

transmembrane region was identified by TMHMM 2.0 (www.cbs.dtu.dk). coiled coil

structures were searched using Coils 2.1 (www.embl.de). Protein family and domain search

was performed with lnterProScan (www.ebLac.uk) and Pfam (pfam.wustl.edu) algorithms.

4.2.3 Construction of icmG expression vectors

To generate an N-terminal fusion of GST to lemG, the icmG gene was amplified using

genomic DNA from JR32 as template and the primers oUA32

(GATCGTACGGATCCATGATGGCAGAGCACG) and oVA33 (GATCGTACGTCGA

CTCAACTATCTTCTTGACTAAAC), digested with BamHl and Sall and ligated into the

87

Page 89: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

BamHI-SalI-digested pGEX-4T-1 (Amersham Biosciences) to generate pUA47 (pGEX­

IcmG).

The same cloning strategy was employed to generate N-termina1 fusions of GST to the N­

terminal part of IcmG (amino acids 1-123) and the C-termina1 part (amino acids 124-269).

For PCR amplification of the N-termina1 part of IcmG, oUA32 and oUA37

(GTACGATCGTCGACTCAATCGGGGTCATCTTCAATAATAG) were used. The C­

terminal part was amplified usmg oUA38 (GATCGTACGGATCCCT

AAAGAAGAAAGTTTCAGCAATAGAAATG) and oUA33. BamHI-SalI digest and

1igation into pGEX-4T-1 yielded pUA52 (pGEX-icmG-N123) and pUA53 (pGEX-IcmG­

C146).

An N-termina1 His6 tag was attached to IcmG by amplifying the icmG gene from genomic

DNA from JR32 with oUA34 (GTACGTACATATGAT GGCAGAGCACGATC) and

oUA33, digestion of the PCR product with NdeI-SalI and ligation into the NdeI-SalI-digested

pET28a(+) (Novagen), yielding pUA48 (pET-icmG-HisN).

For inducible expression of icmG and N-terminal1y His6-tagged icmG in 1. pneumophila, the

genes were transferred into the broad host plasmid pMMB207C (Chen et al., 2004). The

NdeI-HindIII insert from pUA48 was cloned into the NdeI-HindlII-digested pMMB207C to

yield pUA50 (pMMB207C-icmG). The XbaI-HindIII insert from pUA48 was cloned into the

XbaI-HindIII-digested pMMB207C to yield pUA51 (pMMB207C-IcmG-HisN).

For inducible expression of N-terminal1y M45-tagged icmG in 1. pneumophila, the insert

from the BamHI-SalI digested pUA47 (pGEX-IcmG) was 1igated into the BamHI-SalI­

digested pCR33 (pMMB207C-RBS-M45-G1Y6; Weber et al., 2006) to yield pUA54

(pMMB207C-M45-IcmG).

4.2.4 Pull-down assays using GST fusion proteins

N-termina1 GST-IcmG fusion proteins were used to search for IcmG binding proteins in pull

down assays. GST-IcmG, GST-IcmG-N123 (GST fused N-terminally to amino acids 1-123

of IcmG) and GST-IcmG-C146 (GST fused N-terminally to amino acids 124-169) and GST

alone as a control were expressed in E. coli BL21 (DE3) (Novagen). The BL21 (DE3) strains

were diluted to an OD6oo of 0.1 and grown at 37°C until the OD600 reached 0.6-0.8.

Expression was induced with 0.5 mM IPTG, cells were grown for additiona14 hours at 30°C

and pelleted (6000 x g, 10 min, 4°C). 25 m1 of the bacterial culture expressing GST were

88

Page 90: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

used per batch. All three GST-IcmG derivatives are poorly soluble and required a volume of

150 ml culture per batch to obtain sufficient soluble protein. The bacterial pellets were

resuspended in 0.5 ml (GST-expressing bacteria) or 1.2 ml (GST-IcmG-fusions-expressing

bacteria) lysis buffer A (50 mM Tris, 150 mM NaCl, 0.5 % Triton XI00, 1 mM CaCh,

100 IlM PMSF, 1 mg DNAse per 100 ml, pH 7.4). GST-expressing bacteria were lysed by

three times 10 seconds sonication on ice. GST-IcmG fusion proteins were lysed by three

passages through a French pressure cell and supplemented with 2 mM EDTA. Cell debris

was removed by two centrifugation steps (21000 x g, 15 min, 4 C).

Glutathione sepharose 4B (Amersham Biosciences) was equilibrated in lysis buffer A. GST

or the GST-IcmG derivatives were bound to 100 III glutathione sepharose at 4°C for 4 hours,

washed four times on a rotating wheel at 4°C with 5 ml of the lysis buffer A and two times

with lysis buffer B (50 mM Tris, 50 mM NaCl, 0.1 % Triton XlOO, 1 mM CaCh, IOOIlM

PMSF, 1 spatula tip DNAse per 100 ml, pH 7.4).

To search for Legionella proteins binding to IcmG, 200 ml AYE medium per batch were

inoculated with JR32 at an OD6oo of 0.15 and grown for 22 hours at 37°C. The bacteria in

early stationary phase were pelleted, resuspended in 2 ml lysis buffer B, lysed by three

passages in a French pressure cell and supplemented with 2 mM EDTA. Cell debris was

removed by two centrifugations (21000 x g, 15 min, 4°C) to obtain the JR32 lysate.

To search for host cell proteins binding to IcmG, seven T150 flasks (1050 cm2) were

inoculated with RAW264.7 macrophages. The cells were grown to 90% confluency, washed

with ice cold PBS and scraped off in 4 mllysis buffer B. The macrophages were lysed by 10

passages through a 26 GA needle, and the lysate was supplemented with 2 mM EDTA. Cell

debris was removed by two centrifugation steps (21000 x g, 15 min, 4°C) to obtain the clear

macrophage lysate.

The glutathione sepharose beads loaded with GST or the GST-IcmG derivatives were

incubated over night at 4°C with the JR32 lysate, the RAW264.7 lysate or lysis buffer as a

control. The beads were washed extensively five times with 5 mllysis buffer B at 4 °C and

boiled for 5 minutes after addition of Laemmli sample buffer to release proteins. Proteins

bound to 100 III beads were separated on a 8%-18% gradient SDS gel, transferred onto PVDF

membranes, stained on the membrane with Coomassie brilliant blue, and bands were cut out

and analyzed by Edman degradation at the ETH protein service laboratory. In some cases, the

protein bands were stained with Coomassie brilliant blue within the gel, cut out and analyzed

by tryptic digest and MALDI-MS at the protein service laboratory.

89

Page 91: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

4.2.5 Copurification of IcmG and binding proteins using His6~tagged

IcmG

To copurify IcmG and binding partners, icmG with an N-terminal His6 tag was overexpressed

in 1. pneumophila !:J.icmG. An over night culture of !:J.icmG/pMMB207C-IcmG-HisN was

diluted to an OD600 of 004, incubated at 37 QC until the OD6oo reached 1.0, induced with 0.5

mM IPTG and kept growing for four hours. Bacteria from 200 ml culture were used per

batch. Cells were spun down (6000 x g, 15 min, 4 QC), resuspended in 3.5 mllysis buffer HI

(10 mM imidazol, 150 mM NaCl, 50 mM NaH2P04, 0.2% Triton XlOO, 100 ~M PMSF, pH

8.0) or lysis buffer H2 (2.5 mM imidazol, 200 mM NaCl, 50 mM NaH2P04, 0.5%

Triton XI00, 100 ~M PMSF, pH 8.0), and lysed by three passages through a French pressure

cell. Cell debris was removed by two centrifugation steps (21000 x g, 20 min, 4 QC) and

added to 200 ~l Ni-NTA beads (Qiagen) that had been preequilibrated in lysis buffer. After 2

hours of incubation at 4 QC, the beads were washed five times with 5 ml lysis buffer HI

containing 10 mM imidazol, respectively, and eluted with increasing imidazol concentrations

or further washed with increasing NaCl concentrations as described in the figure legends.

Proteins were separated on an SDS gel and stained with Coomassie brilliant blue or blotted

onto nitrocellulose, and His-tagged proteins were detected with an anti-His4 antibody

(Qiagen).

4.2.6 Generation of antibodies and Western blot

An antibody against a partial amino acid sequence of IcmG (amino acids 189 to 203) was

derived by immunization of rabbits with the chemically synthesized peptide

CARTTPKKVVKVSRPI coupled to the carrier protein KLH (keyhole limpet hemocyanin)

(NeoMPS, Strasbourg, France). The antibody was used in Western blots diluted 1:2000 in

blocking buffer (PBS with 5% fat free milk powder and 0.05% Tween 20) and visualized by

a goat anti rabbit-HRP conjugate (Becton Dickinson, diluted 1:4000 in blocking buffer) and

luminescence detection (ECL kit, Amersham Biosciences). The presence of 20% or 30%

methanol and the lack of SDS in the Tris-glycine blotting buffer turned out to be crucial for

strong signals.

The anti-SidC and anti-M45 antibodies are described elsewhere (Weber et aI., 2006).

90

Page 92: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

4.2.7 Immunofluorescence microscopy

Intracellular localization of proteins was examined by immunofluorescence of infected

Dictyostelium discoideum AX3 expressing a calnexin-gfp fusion protein as described in

Weber et al., 2006. In brief, 2 x 104 amoebae were seeded onto glass cover slips in a 24-well

plate the day prior to infection. Bacteria for infection were inoculated at an OD600 of 0.1 and

grown for 21 hours at 37 QC on a rotating wheel. Expression of M45-sidC was induced upon

inoculation, expression of M45-icmG was induced after 17 hours. On the day of infection,

bacteria were added at a multiplicity of infection of 100, spun onto the amoebae (880 x g, 5

min), and the amoebae were incubated for one hour at 25 QC. After a wash with SorC

(Malchow et al., 1972), the cells were fixed for 30 minutes at 4 QC with 4% PFA in PBS,

permeabilized for 10 min at 25 QC with 0.1% Triton XI00 in SorC, and blocked over night

with 2% normal human AB serum in SorC.

M45-tagged proteins were detected by a monoclonal anti-M45 hybridoma supernatant (Obert

et al., 1994, diluted 1:4 in blocking solution). SidC was detected with an affinity-purified

rabbit anti-SidC antibody (Weber et aI., 2006, diluted 1:1000 in blocking solution). Cy5­

conjugates from Jackson ImmunoResearch were used as secondary antibodies (l :200 in

blocking solution). Bacteria were either detected with a rhodamine-labelled anti-Legionella

pneumophila Philadelphia-l Serogroup 1 antibody (m-Tech, 1:100 in blocking solution) or by

constitutive expression of the red fluorescent protein DSRedExpress from plasmid pSWOOl

(Mampel et al., 2006). The cover slips were mounted and analyzed using a Zeiss Axiovert

200 microscope and a Perkin-Elmer Ultraview confocal imaging system.

To control for similar expression levels of M45-tagged proteins, whole cell cultures were

adjusted to an OD600 of 1.0, boiled in Laemmli buffer, separated by SDS gel electrophoresis

and analyzed by Western blot using the anti-M45 antibody described above.

4.3 Results

4.3.1 IcmG is present in the bacteria in low amounts

The intracellular concentration of a protein and the regulation of its expression can give hints

about its function, and, therefore, synthesis of IcmG was analyzed by Western blot. A

polyclonal rabbit antibody against an IcmG-derived peptide was generated, and the amount of

protein in early stationary growth phase cultures, during which L. pneumophila is most

91

Page 93: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of lemG

virulent, was determined by Western blot (Figure 2). IcmG expression under control of the

Ptae promoter for 4 or 8 hours yielded strong bands even after 30 seconds exposure time,

whereas lemG from wild type (JR 32) or expressed from a plasmid containing the icmGCD

genomic region (~G/pGCD) became only visible as a weak band after 25 minutes exposure

time, indicating that lemG is present in the bacteria only at low concentrations.

JR32

____' uninduced

~G ~G/pG

_------=:I4--'--Jh'--!JinL!>d..,.u""'ce><.>d'---- ------"'8--'-h'---'-iL!>nd"--"'u~ __,~'''__~G/pGH ~G/pG ~G/pGH ~G/pG l'.G/pGH ~G/pGCD

Figure 2: Expression levels of icmG under native conditions and after 4 and 8 hoursoverexpression. Bacterial cultures were grown to early stationary phase and whole cell culturesdiluted to an OD600 of 1.0 were analyzed by Western blot using the rabbit anti-IcmG scrum. The bandswere visualized using the ECL kit (Amersham Biosciences). Exposure time was 25 minutes. Thenative IcmG position is marked with a triangle. The uppennost band is unspecific and serves as aninternal control. Abbreviations: L1G: McmG/pMMB207C, L1G/pG: L1icmG/pMMB207C-lcmG,L1G/pGH: L1icmG/pMMB207C-IcmG-HisN, L1G/pGCD: L1icmG/pGS-Lc-63-14 (genomic fragmentcontaining icmGCD in pMMB207-Km14; Segal and Shuman, 1999).

To examine whether icmG might be stronger expressed at growth phases between logarithmic

and late stationary phase, 14 to 27 hours old cultures were examined for the amount of IcmG

protein by Western blot. All of the samples showed a low IcmG expression comparable to

that one shown in Figure 2 (data not shown).

Expression of icmG could also be induced by environmental conditions inside of a host cell.

This possibility was examined using A. castellanii as host cells. The amoebae were infected

with JR32 and ~icmG, and the presence of IcmG was assayed in TeA-precipitated culture

medium supernatant and in amoeba lysates after 24 and 48 hours post infection by Western

blot. lemG was not detected in the supernatants or in amoebae infected with L1icmG. The

concentration of IcmG in amoebae infected with JR32 was compared with the concentration

in the early stationary culture used for infection. Bacterial numbers were estimated by taking

the intensitiy of the unspecific band above IcmG as a standard. There was no difference

between IcmG levels in JR32 in early stationary cultures and in amoebae infected for one or

two days, indicating that intracellular conditions do not increase expression of icmG

significantly (data not shown).

92

Page 94: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

_____~ ~_S:hapter4

Acting as a SNARE protein would possibly require secretion of IcmG. Secretion would also

decrease the intracellular concentration and could lead to the low protein levels detected in

the bacteria. No substances are known that induce secretion of Legionella effector proteins

into the culture medium. Congo Red has been shown to induce secretion of effector proteins

in Shigella .flexneri (Bahrani et al., 1997), and contact with cholesterol rich liposomes was

able to activate S. jlexnerii type III secretion (van der Goot et al., 2004). Therefore, Congo

Red and cholesterol were tested for their ability to induce secretion of effector proteins also

in Legionella. SidC has been shown to be translocated during infection (Luo and Isberg,

2004; Weber et al., 2006) and was used to determine whether secretion of effector proteins

into the culture supernatant occurs under the experimental conditions chosen. TCA

precipitated supernatant (20% TCA, over night) from 20 ml untreated or Congo Red (20 IlM)

or cholesterol (50 IlM) supplemented cultures and bacterial cells (JR32 and l1icmT) were

examined by Western blot. SidC was strongly expressed intracellulary in all cultures but

could not be detected in the untreated or induced supernatants, indicating that secretion does

not take place during growth in liquid medium and neither Congo Red nor cholesterol could

induce secretion into the supernatant (data not shown). IcmG was weakly expressed in thc

bacterial pellets and was not detected in the supernatants, indicating that the low intracellular

IcmG concentration is not due to release into the supernatant.

4.3.2 IcmG is not translocated during infection

Acting as a SNARE protein probably requires translocation of IcmG during infection into the

host cytosol and insertion of the protein into the membrane of the Legionella containing

vacuole. Thus, the SNARE domain facing the cytosol can interact with SNARE proteins of

other vesicles to direct vesicle fusion. Immunofluorescence microscopy was used to localize

IcmG during infection and to find out whether it is translocated to the LCV membrane. Since

SidC has been shown to localize to the LCV membrane (Weber et al., 2006), it was used as

positive control for vesicular staining. M45-tagged icmG and side were overexpressed,

D. discoideum amoebae were infected with early stationary growth phase bacteria, and

proteins were localized one hour post infection by immunofluorescence. Bacterial protein

expression levels were assessed by Western blot (Figure 3).

93

Page 95: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

MW M45-lcmGJR32 ~icmT

M45-SidCJR32 ~icmT

84 ­

62 ­48 _

33 -

25 -

17 -

Figure 3: Expression of M45-tagged proteins for immunofluorescence analysis. Whole cellcultures used for infection were adjusted to an identical 00600 and analysed by anti-M45 Western blotto control for similar protein expression.

The left pictures in Figure 4 show localization of M45-SidC in amoebae infected with wild

type bacteria. During the staining procedure, most bacteria get perrneabilized, and

intrabacterial M45-SidC gets stained. Comparable intrabacterial staining is also visible in

bacteria that are not located inside of or attached to host cells and thus did not induce

translocation. However, a large part of intracellular wild type bacteria show M45-SidC

signals that localize clearly outside of the bacteria and often colocalize with calnexin-GFP, a

marker for the LCV membrane. In contrast, M45-IcmG never co-localized with calnexin

(Figure 4, right images). In these preliminary experiments, we could not discriminate whether

IcmG solely localized inside the bacteria, or whether a portion of the protein was also

secreted to the bacterial surface. Similar results were obtained for infections with !1icmG

expressing M45-IcmG (data not shown). Intrabacterial concentrations of M45-IcmG in the

different strains were similar or even higher than those of M45-SidC (Figure 3), and M45­

IcmG and M45-SidC signals inside of lysed bacteria had a comparable intensity indicating

that staining intensities for both proteins are comparable. A possible interference of the M45­

tag with translocation of IcmG was not analysed in detail and can not be excluded. However,

since M45-tagged SidC was translocated efficiently, it is likely that the tag does not inhibit

translocation of IcmG either. Therefore, translocation of IcmG during infection in amounts

comparable to other effector proteins like SidC is unlikely to occur.

94

Page 96: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

JR32/pM45-SidC

___________ c;)lapte~_~_

JR32/pM45-lcmG

merged

anti-M45

anti-LPS

Figure 4: Immunofluorescence analysis of translocation of M45-tagged proteins duringinfection with L. pneumophila .JR32/pMMB207C-M45-sidC (left pictures) andJR32/pMMB207C-M45-icmG (right pictures). D. discoideum Ax3 were infccted with an m.o.i. of100 for one hour and staincd using an anti-M45 antibody (blue). L. pneumophila were staincd with ananti-L. pneumophila antibody (red). 'rhe amoebae express a calnexin-GFP fusion protein whichlocalizes to the ER. GFP signals are shown in green. Arrows indicate bacteria in which translocationofIvf45-SidC can casily be seen (left picturcs). Scale bars denote 5 ~Lm.

95

Page 97: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

4.3.3 L. pneumophila AicmG is able to translocate SidC but is impairedin recruiting ER-derived vesicles to the LCV

The Icm/Dot transporter translocates effector proteins during infection of a host cell. In order

to get a better understanding of the role of IcmG, its requirement for effector protein

translocation was examined. The Icm/Dot-secreted protein SidC was used as a model effector

protein and was expressed in L. pneumophila wild type JR32, the icmG mutant and the

Icm/Dot transport deficient icmT mutant. D. discoideum was infected, and localization of

proteins was determined one hour post infection by immunofluorescence using an affinity

purified polyclonal anti SidC antibody.

Figure 5 shows that SidC localizes to the LCV membrane in amoebae infected with wild type

bacteria and is visible only intracellularly in lysed bacteria in amoebae infected with the icmT

mutant. Translocation of SidC was also detected in infections with the icmG mutant, and the

signals localized to the membrane of the LCV in the same pattern as seen in infections with

wild type bacteria, thus proving that translocation of at least the effector protein SidC is still

possible in an icmG mutant.

However, a large difference was observed between wild type and llicmG infections in the

ratio of LCVs stained with SidC and calnexin-GFP. Calnexin is an ER marker and indicates

the recruitment of ER-derived vesicles to the LCV membrane (Derre and Isberg, 2004).

Whereas about two-thirds of the LCV membranes in an infection with wild type bacteria co­

localized with calnexin-GFP, less than ten percent of the LCVs containing the icmG mutant

showed calnexin-GFP signals. Only calnexin-GFP positive vacuoles were further examined

for the presence of SidC in their membranes. 75% of the wild type containing vesicles (27 out

of 36) but only 39% of the llicmG containing vesicles (7 out of 18) stained also positive for

SidC. No calnexin-GFP positive LCVs and no SidC translocation were found in infections

with the icmT mutant.

This data show that the icmG mutant is impaired for the recruitment of ER derived vesicles

and the translocation of effector proteins. However, IcmG is not absolutely required for

translocation of at least the effector protein SidC, as SidC is still translocated to some LCV

membranes.

96

Page 98: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

JR32

!1.icmT

!1.icmG

!1.icmG

anti-Side merged

Figure 5: Immunofluorescence analysis of translocation of Side during infection withL. pneumopltila strains. D. discoideum Ax3 were infected with an m.o.i. of 100 for one hour andstained using an anti-SidC antibody. SidC signals are shown in the left picture and localize to themembrane of the LCV with exception of l:iicmT. Bacteria are visualized by fluorescence ofDsRedExpress and are shown in red. The amoebae express a calnexin-GFP fusion protein whichlocalizes to the ER. GFP signals are shown in green. Scale bars denote 5 !lm.

97

Page 99: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

4.3.4 Identification of IcmG binding proteins using a pull down assaywith GST fusion proteins

The C-terminal part of IcmG contains a coiled-coil domain. These domains are often

involved in protein-protein interactions, such as assembly of subunits (Delahay and Frankel,

2002). A SNARE domain was predicted in IcmG at amino acids 140 to 210 (Morozova et al.,

2004). SNARE domains promote vesicle fusion by binding SNAREs on target vesicle

membranes. Furthermore, a bacterial two hybrid screen revealed that a number of effector

proteins interact with an IcmG fragment spanning amino acids 28-123 (Luo and Isberg,

2004). The identification of proteins interacting with these or other domains of IcmG could

give clues about its function. Thus, we examined protein binding to IcmG using pull down

assays.

GST was fused to full length IcmG and the N- and C-terminal part ofIcmG, respectively. The

fusion proteins were loaded onto glutathione beads and used as baits to bind proteins either

from L. pneumophila JR32 lysate to identify bacterial binding partners, or to bind proteins

from host cell lysate to identify host cell binding partners.

Figure 6 shows a pull down assay designed to identify bacterial binding partners. Solubility

of the three GST-IcmG fusion proteins was poor, and despite of a sixfold increased culture

volume, more GST (26kDa) bound to the glutathione beads than GST-IcmG (56kDa), GST­

IcmG-NI23 (apparent MW 45 kDa, calculated MW 40kDa) and GST-IcmG-CI46 (apparent

MW 38 kDa, calculated MW 42 kDa). Also, much less unspecific binding was observed for

GST-Ioaded beads, compared to beads loaded with GST-IcmG fusion proteins (Figure 7 and

Figure 8, see lanes in which beads were incubated with buffer only).

Several proteins were identified that bound specifically to the GST-IcmG fusion proteins.

The numbered bands were analyzed by Edman sequencing. However, only band 5 gave a

reliable sequence. BLAST search against the sequenced L. pneumophila genomes (strains

Philadelphia, Lens and Paris) did not yield a protein sequence fully matching the Edman

result. The best hit was 75% identical (6 out of 8 amino acids) with the N-terminus of

L. pneumophila proteins annotated as Hsp60 60 kDa chaperonin HtpB (strain Philadelphia) or

as 60 kDa chaperonin Cpn60 (GroEL) (strains Lens and Paris). To examine whether this

protein might originate from E. coli BL21, a BLAST search against E. coli proteins was

done. Again, no fully matching protein could be identified and the best hit yielded 77% (7 out

of9 amino acids) identity to E. coli hsp60 (GroEL).

98

Page 100: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter ~..

In another pull down experiment (Figure 7), band 3 was clearly identified as the 40 kDa heat

shock protein Dnal from 1. pneumophila with 10 out of 10 amino acids matching. The

remaining numbered bands shown in Figure 6 and Figure 7 were also analyzed by Edman

sequencing, but no reliable results could be obtained. Some bands gave no signals in the

amino acid detection, and in one case a band contained two proteins in similar amounts, thus

eliminating the possibility to assemble sequences.

MW JR32Ly

GSTB Ly

GST-lcmGB Ly

GST-G-N123B Ly

GST-G-C146B Ly

83-

62-

48-

33-

25-

17-

7-

Figure 6: Purification of IcmG binding proteins from L. pneumophUa wild type lysate using apull down assay. GST-fusion proteins of IcmG or the N- and C- tenninal part ofIcmG, respectively,were immobilized on glutathione sepharose beads and incubated with lysate from JR32. Unboundproteins were washed off, bound proteins were released by boiling the beads in Laemmli buffer,separated on an SOS gel, transferred onto PVOF membrane and visualized by staining withCoomassie brilliant blue. The marked bands were analyzed by Edman sequencing. Abbreviations: Ly:lysate from JR32. B: lysis buffer, GST-IcmG: GST fused to full length IcmG, GST-G-N 123: GSTfused to the N-tennillal 123 amino acids of IcmG, GST-G-CI46: GST fused to the 146 C-tenninalamino acids ofIcmG.

99

Page 101: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

Lysates MW buffer MW JR32 MW RAW MW Ecoli IysatesJR32 RAW GST GST-G GST GST-G GST GST-G GST GST-G

Figure 7: Purification of IcmG binding proteins from L. pneumopltila wild type JR32 and frommurine RAW264.7 macrophage lysate using a pull down assay. GST and a GST-IcmG fusionprotein were immobilized on glutathione sepharose beads and incubated with bacterial or eukaryoticlysate. Unbound proteins were washed off, bound proteins were released by boiling the beads inLaemmli buffer, separated on an SDS gel and visualized by staining with Coomassie brilliant blue.The marked bands were cut out and analyzed by MALDI-MS. Abbreviations: GST-G: GST-IcmGfusion, MW: molecular weight marker.

Bands obtained from pull down assays using lysate from RAW264.7 macrophages as prey

were identified using MALDI-MS. The protein fragments from analyzed bands matched with

a RI02 kinase like protein (UniProt identifier Q8BHT4), a nuclear export factor CRMl

(Q80U96) and a protein similar to GCN 1, a transcriptional regulator involved in amino acid

synthesis in yeast (Q8CHH7).

100

Page 102: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

____'0__ ' __ ••••• _

Chapter 4

4.3.5 Copurification of IcmG binding proteins using Hiss-tagged IcmG

Pull down of IcmG binding proteins using GST fusion proteins requires reconstitution of

protein complexes in vitro. An alternative approach to identify IcmG binding proteins that

avoids reconstitution in artificial buffer is copurification of IcmG together with its binding

partners. His6-tagged IcmG was overexpressed in !1icmG, the lysate was incubated with Ni­

NTA beads, unbound proteins were washed off, and His-IcmG and binding proteins were

eluted and analyzed. Since initial experiments yielded a high number of unspecifically bound

proteins, the effect of the major parameters imidazol concentration and ion strength on the

enrichment of His-IcmG was examined. Ni-NTA beads were loaded with bacterial lysate,

washed with lysis buffer HI (lOmM imidazol, 150 mM NaCl) and treated either with

increasing imidazol or NaCI concentration to determine the highest concentrations which

could be used to wash off proteins binding unspecifically without releasing His-IcmG. Figure

8 shows that after 5 washes with 5 ml lysis buffer HI large amounts of many different

proteins remain bound to Ni-NTA and are released by an increase in imidazol concentration

(left image). Only a small part of these proteins can be detached by increasing ion strenght

(right image). Bound His-IcmG appears to be firmly attached to the beads, since it can not be

eluted with 500 mM imidazol but only by boiling with EDTA (see anti-His Western blot, left

picture). Unfortunately, the portion of His-IcmG binding to the beads seems to be marginal.

1.25 Jlllysate (100 Jlg protein) were applied directly to the first two lanes in the left gel and

yielded strong bands in the Western blot. His-IcmG from 4500 Jlllysate was enriched on the

Ni-NTA beads but yielded only a signal not stronger than that from 1.25 JlI lysate after

releasing bound His-IcmG by boiling the beads in 100 JlI 500 mM EDTA in water. The

presence of His-IcmG in the lysate after incubation with Ni-NTA beads shows that the

protein was available in the lysate in excess and therefore, binding of His-IcmG to Ni-NTA

was poor.

101

Page 103: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

33

17

25

MW Lys+ Lys+ Lys- _5_0_IQbL. . 1]5 ~50 .2illL.- boiled MW wash 1 wash 3 wash 5 ~50 50.Q 1000 2000 boiledkDa before b, after b, IPTG mM Imidazol +EDTA mM Nael +EDTA

17583

62

48

a-HisWB--~

Figure 8: Binding and release of Hisdagged IcmG and other proteins bound to Ni-NTA beads.Proteins from 200 ml culture from L1icmG/pMMB207C-IcmG-HisN were extracted using lysis bufferHI, applied to 200 III Ni-NTA beads, and unbound proteins were washed off with lysis buffer HI asdescribed in Material and Methods. Proteins were separated on SDS gels and stained with Coomassiebrilliant blue (top panel) or blotted onto nitrocellulose membrane and analyzed by an anti-HisWestern blot (bottom panel). The left gel shows the protein pattern of the lysates and the proteinseluted with 200 III lysis buffer containing increasing concentrations of imidazol. The right gel showsthe proteins washed off after loading the beads (wash I to wash 5) and the proteins subsequentlyeluted by addition of 200 III lysis buffer containing 10 mM imidazol and increasing NaCIconcentrations. In both cases, the beads were boiled in Laemmli buffer containing 500 mM EDTAafter the last elution or wash step to quantitatively release all bound proteins. Abbreviations: Lys+before b.: lysate from L1icmG/pMMB207C-icmG-HisN induced with IPTG before incubation with Ni­NTA beads, Lys+ after beads: the same lysate after incubation with Ni-NTA beads, Lys-lPTG: lysatefrom L1icmG/pMMB207C-icmG-HisN without induction.

Possible reasons for poor binding of His-IcmG could be a tertiary structure which embeds the

His tag inside the protein, or aggregation of His-IcmG with itself or other proteins via the

hydrophobic transmembrane or the coiled-coil domain. This could render the His tag

inaccessible and prevent binding of the tag to the nickel ions.

To discriminate whether biochemical properties of IcmG or experimental conditions led to

poor binding of His-IcmG, the purification was repeated including His6-tagged SidC as a

control protein. One purification was performed using denaturing conditions (8 M urea)

during lysis, binding and washing to disrupt tertiary structures. Figure 9 shows a strong

enrichment of His-SidC under native conditions, whereas not enough His-IcmG has bound to

be visualized by Coomassie staining. An anti-His Western blot of the lysate proteins

confirmed that His-SidC and His-IcmG were present at similar concentrations (data not

shown). Western blot ofthe released proteins (right image) also showed that under denaturing

conditions binding of His-IcmG was increased.

102

Page 104: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

Coomassie _._~.....L'lJJ1id::l,-,-"isL.!W-"-,B,,,-----__kDa G/pG G/pG T/Sid Coli/pG G/pG G/pG T/Sid Coli/pG

nat denat nat nat nat denat nat nat

83 ­62 -

48 -

33 -

25 -

17 -

Figure 9: Enrichment of His6-tagged IcmG and Side by binding to Ni-NTA beads. Proteins from200 ml culture were extracted using lysis buffer H2 and were applied to 200 III Ni-NTA beads asdescribed in Material and Methods. For G/pG denat, both lysis and wash buffer were supplementedwith 8 M urea to examine binding under denaturing conditions. The beads were washed with lysisbuffer H2 adjusted to 50 mM imidazol and boiled in Laemmli buffer supplemented with 500 mMEDTA to release all bound proteins. Proteins were separated on an SDS gel and stained withCoomassie brilliant blue (left picture) or analyzed by Western blot with an anti-His antibody (rightpicture). The position of His-tagged IcmG and His-tagged SidC is indicated with black triangles.Abbreviations: G/pG: ~icmG/pMMB207C-icmG-HisN, T/Sid: ~icmTlpMMB207C-sidC-HisN,

Coli/pG: E. coli TOPIO/pMMB207C-icmG-HisN, nat: lysisbuffer H2 without 8 M urea, denat: lysisbuffer H2 with 8 M urea.

The successful enrichment of His-SidC shows that not the experimental conditions but the

biochemical properties of His-IcmG cause poor binding to the beads. Denaturing conditions

increase binding of His-IcmG but disrupt protein bindings and can therefore not be used in a

copurification assay. Thus, a successful copurification of IcmG binding proteins using His­

tagged IcmG is unlikely, and this approach was not further pursued.

4.4 Discussion

The presence of a SNARE domain in 1cmG was predicted by in silico analysis, and acting as

a SNARE protein would render IcmG an important player in the modification of vesicle

trafficking during infection. Furthermore, the cytotoxic effect of the IpxB paralogue IcsC was

only observed when lcsC was expressed in the icmG mutant, but not in other mutants with

partial virulence phenotypes. This prompted us to characterize IcmG in more detail to get a

better understanding of its role in infection and in the mechanism of IcsC-mediated

cytotoxicity.

103

Page 105: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

4.4.1 Expression level and localization of IcmG

The intracellular concentration of IcmG was low in all growth phases examined and also

during intracellular growth in A. castellanii. Furthermore, secretion of IcmG could not be

detected, suggesting that the icmG expression is constantly kept low. Secretion and SNARE

function would probably require expression of larger amounts of protein to ensure that

sufficient protein is available to be inserted in all parts of the LCV membrane and to come in

contact with host SNARE proteins. Neither secretion nor translocation could be detected by

Western blot of TCA-precipitated supernatants (see 4.3.1) and by immunofluorescence

localization ofN-terminally M45-tagged IcmG in infected D. disoideum (see 4.3.2). IfIcmG

would act as a SNARE, it is expected to be translocated and its transmembrane domain to be

inserted into the LCV membrane with the C-terminal SNARE domain facing the cytosol.

This localization would result in a staining pattern similar to SidC, which localizes on the

LCV membrane. Using the relatively insensitive immunofluorescence microscopy as

detection method, it can not be excluded that small amounts of IcmG are translocated. Both

the low expression level and the fact that translocation does not occur or only at levels below

the detection limit argue against a SNARE function.

The prediction of Morozova and coworkers is based upon PFAM domain searches and an

alignment of IcmG and three bacterial protein domains showing similarities to SNARE

domains from eukaryotic proteins. Our own domain search using Pfam 15.0 (pfam.wustl.edu)

yielded a SNARE domain with an expectation value of 0.36. E-values above 0.05 should not

be considered as significant. Together with the data presented here, it is likely that the

predicted SNARE domain does not function as such.

4.4.2 Phenotypes of the icmG mutant

We examined the effect of loss of IcmG on the ability to translocate effector proteins during

infection. By immunofluorescence, it could be shown that i1icmG is able to translocate SidC

to the LCV membrane and, therefore, the Icm/Dot secretion system is at least partially

functional. However, recruitment of ER derived vesicles and the portion of SidC-positive

LCVs were clearly reduced in i1icmG, indicating that IcmG is required for optimal function

of the transporter. Thus, IcmG could be part of the transporter complex rather than

functioning as a SNARE protein. If IcmG is a part of the transporter complex, one would

104

Page 106: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

expect that it binds to other Icm/Dot proteins, and it remains unclear why none of these were

identified in the pull down assays. IcmG was poorly soluble even when fused to GST and

dissolved in a detergent-containing lysis buffer. 15 out of 21 Icm/Dot proteins are predicted

to localize in the inner membrane due to the presence of one ore more transmembrane

domains. It might be that some of these Icm/Dot proteins do form a complex together with

IcmG in vivo, but they suffered from poor solubility and were not available in sufficient

amounts in the pull down assays to give a clear band in the Coomassie staining or a clear

sequence in Edman analysis.

4.4.3 Identification of IcmG binding proteins in pull down assays

Pull down assays with GST fusion proteins were performed to identify IcmG binding proteins

from L. pneumophila wild type lysate or from murine RAW264.7 macrophages.

The pull down assays with JR32 lysate clearly identified DnaJ and, less confidently, a Hsp60

like protein. DnaJ bound to full length IcmG and Hsp60 to amino acids 124-269 of IcmG.

Both DnaJ and Hsp60 are chaperons involved in folding of a large portion of bacterial

proteins after their synthesis. Since binding to many different proteins is part of their

chaperone function, copurification with IcmG or binding to the immobilized GST-fusion

proteins does not necessarily mean that binding of these chaperones to IcmG is functionally

relevant. This is supported by the observation that DnaJ is frequently copurified with

overexpressed proteins. It might be that IcmG altered its structure during bacterial lysis or

pull down, was recognised as misfolded and bound by the chaperones, thus leading to

purification of chaperones.

However, Hsp60 function is not restricted to folding proteins intracellularly and it may bind

IcmG specifically. Hsp60 has been described to localize on the bacterial surface and to

mediate invasion of L. pneumophila in HeLa cells (Garduno et al., 1998). Hsp60 also

interferes with signalling cascades, actin modification and vesicle trafficking (Garduno,

2005). Secretion of Hsp60 was dependent on an intact Icrn/Dot secretion apparatus, since

several icm/dot mutants were defective in localizing Hsp60 onto their surface and

accumulated Hsp60 intracellularly (Hoffman and Garduno, 1999). This IcrnlDot dependency

shows a connection between Hsp60 and Icm/Dot proteins, and it might be that IcmG is

involved in Hsp60 secretion. However, the sequence obtained by Edman analysis of the

isolated protein matches only 75% with L. pneumophila Hsp60. It will be necessary to

105

Page 107: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

confirm that it actually is Hsp60 that binds IcmG before further experiments regarding the

relation between the two proteins are done.

Luo and Isberg described binding of the RalF and the Sid effector proteins to IcmG (Luo and

Isberg, 2004). In this work, no IcmG-binding effector proteins were identified. Reason for

this might be the different aproaches used for identification of binding partners. Luo and

Isberg used a bacterial two hybrid assay, which identifies also temporally or weakly binding

proteins. In a pull down assay, as used in this work, the binding needs to be established in

vitro and maintained during several wash steps, and thus probably only strongly binding

proteins can be identified. Both RalF and SidC are secreted effector proteins that, since IcmG

itself is not secreted, interact only temporarily with IcmG. This is a possible reason why these

interactions were not detected in the pull down assays.

Pull down of proteins from RAW264.7 macrophages with GST fusion proteins identified a

kinase-like protein, a nuclear export factor and a putative regulatory protein similar to GCNl.

Specific binding of these proteins to IcmG in vivo requires secretion of IcmG into the host

cell. Since neither secretion nor translocation of IcmG during infection could be shown (see

4.3.1 and 4.3.2) is it likely that the binding observed here is an artefact and does not have a

biological meaning.

4.4.4 Hypotheses on the function of IcmG

The data presented here argue against the hypothesis that IcmG acts as a SNARE and suggest

that IcmG is part of the Icm/Dot transporter. The weak expression level of icmG argues for an

involvment in the transporter apparatus rather than for a SNARE function. Presumed that

every transporter complex within a bacterium contains only one or a few copies of IcmG,

only a low number of molecules would be necessary within a cell.

The binding to the translocated effectors RalF and at least eight Sid proteins shows that IcmG

interacts with a number of different effectors. It might act as a coupling protein directing

different effectors towards the translocation channel and assist in their secretion, or it could

be involved in regulation of effector translocation. Loss of a coupling protein or

misregulation of translocation in !1icmG would either reduce the translocation efficiency of

all or a subset of proteins, or it could lead to an uncontrolled release of effectors before or

during infection, hence causing the impaired recruitment of ER derived vesicles. However,

SidC was not detected in TCA precipitated supematants of a !1icmG culture (see 4.3.1),

106

Page 108: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

indicating that uncontrolled release of effectors does at least not happen during growth in

liquid medium.

It might be possible that IcmG is required for translocation only of a subset of effector

proteins. A lack of translocation of these effectors or a reduced amount of translocated

proteins would then cause the partial phenotype of the icmG mutant. The Yersinia protein

TyeA is an example for a translocation control protein required for translocation of only a

subset of Yop effector proteins (Iriarte et al., 1998).

To learn more about the function of IcmG, it would be interesting to compare the ability of

McmG to secrete not only SidC, but also other known effector proteins. This could reveal

whether IcmG acts as coupling protein for all or only for a subset of effectors. A quantitative

time course of effector translocation would give information whether translocation in !i.icmG

is delayed or the amount of translocated proteins is reduced.

lcsC encodes a lipid A disaccharide synthase (see Chapter 3) and is toxic only when

overexpressed in !i.icmG, but not in other mutants with partial phenotypes like !i.icmS, !i.icmF

and !i.rpoS. The proposed functions of IcmG need to be in accordance with these data and,

ideally, can give an explanation for the restriction of cytotoxicity to !i.icmG. Lipid A

disaccharide synthases are cytosolic enzymes that form lipid A, the active moiety of

endotoxin. Overexpression of lcsC might lead to accumulation of lipid A or other LPS

precursors. If !i.icmG is impaired in regulation of translocation, it might be that toxic products

of lcsC overexpression are translocated through the Icm/Dot apparatus in !i.icmG, but not in

other mutants with partial phenotypes. These molecules would be released into the host cell

and could interfere with host cell mechanisms leading to increased cell death. Type IV

dependent secretion of molecules other than proteins has been described for protein bound

DNA in Legionella (Segal and Shuman, 1998) and peptidoglycan in Helicobacter (Viala et

al.,2004).

Another hypothesis not presuming IcmIDot-dependent secretion of an LcsC product is that a

toxic compound can exert its cytotoxic effect only in certain intracellular compartments. It

might be that trafficking of the icmG mutant differs compared to other mutants and only

!i.icmG enters a compartment enabling the bacteria to become cytotoxic.

However, explanations for the mechanism of LcsC-mediated cytotoxicity will be speculative

unless more information are available about the translocation capabilities and the intracellular

behavior of the different mutants. It will also be necessary to more closely examine whether

107

Page 109: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

products from the LPS biosynthesis pathway accumulate during lcsC overexpression,

whether these products are secreted by the !cm/Dot secretion system and how and where they

interact with host cell pathways to cause cell death.

4.5 References

Albers, D., Reus, K, Shuman, RA., and Hilbi, H. (2005) The amoebae plate test implicates aparalogue of lpxB in the interaction of Legionella pneumophila with Acanthamoebacastellanii. Microbiology 151: 167-182.

Bahrani, F.K, Sansonetti, P.J., and Parsot, C. (1997) Secretion ofIpa proteins by Shigellaj/exneri: inducer molecules and kinetics of activation. Infect Immun 65: 4005-4010.

Chen, J., de Felipe, KS., Clarke, M., Lu, H., Anderson, O.R, Segal, G., and Shuman, H.A.(2004) Legionella effectors that promote nonlytic release from protozoa. Science 303:1358-1361.

Chen, Y.A., and Scheller, RH. (2001) SNARE-mediated membrane fusion. Nat Rev Mol CellBioI 2: 98-106.

Delahay, RM., and Frankel, G. (2002) Coiled-coil proteins associated with type III secretionsystems: a versatile domain revisited. Mol Microbiol45: 905-916.

Derre, I., and Isberg, RR. (2004) Legionella pneumophila replication vacuole formationinvolves rapid recruitment of proteins of the early secretory system. Infect Immun 72:3048-3053.

Feeley, l.C., Gibson, RJ., Gorman, G.W., Langford, N.C., Rasheed, J.K, Mackel, D.C., andBaine, W.B. (1979) Charcoal-yeast extract agar: primary isolation medium forLegionella pneumophila. J Clin Microbiol10: 437-441.

Fields, B.S. (1996) The molecular ecology of Legionellae. Trends Microbiol4: 286-290.

Garduno, RA., Garduno, E., and Hoffman, P.S. (1998) Surface-associated Hsp60 chaperoninofLegionella pneumophila mediates invasion in a HeLa cell model. Infect Immun 66:4602-4610.

Garduno, RA. (2005) The Hsp60 chaperonin ofLegionella pneumophila: an intriguingplayer in the infection of host cells. In 6th International Conference on LegionellaChicago, Illinois.

Hagele, S., Kohler, R, Merkert, H., Schleicher, M., Hacker, l., and Steinert, M. (2000)Dictyostelium discoideum: a new host model system for intracellular pathogens of thegenus Legionella. Cell Microbiol2: 165-171.

Hilbi, H., Segal, G., and Shuman, H.A. (2001) Icrn/dot-dependent upregulation ofphagocytosis by Legionella pneumophila. Mol Microbiol42: 603-617.

Hoffman, P.S., and Garduno, R.A. (1999) Surface-associated heat shock proteins ofLegionella pneumophila and Helicobacter pylori: roles in pathogenesis and immunity.Infect Dis Obstet Gynecol7: 58-63.

108

Page 110: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 4

Horwitz, M.A, and Silverstein, S.c. (1983) Intracellular multiplication of Legionnaires'disease bacteria (Legionella pneumophila) in human monocytes is reversibly inhibitedby erythromycin and rifampin. J Clin Invest 71: 15-26.

Iriarte, M., Sory, M.P., Boland, A, Boyd, AP., Mills, S.D., Lambermont, 1., and Comelis,G.R. (1998) TyeA, a protein involved in control ofYop release and in translocation ofYersinia Yop effectors. EmboJl7: 1907-1918.

Luo, Z.Q., and Isberg, R.R. (2004) Multiple substrates of the Legionella pneumophilaDot/Icm system identified by interbacterial protein transfer. Proc Natl Acad Sci USA101: 841-846.

Malchow, D., Nagele, B., Schwarz, H., and Gerisch, G. (1972) Membrane-bound cyclic AMPphosphodiesterase in chemotactically responding cells of Dictyostelium discoideum.EurJ Biochem 28: 136-142.

Mampel, J., Spirig, T., Weber, S.S., Haagensen, J.A, Molin, S., and Hilbi, H. (2006)Planktonic replication is essential for biofilm formation by Legionella pneumophila ina complex medium under static and dynamic flow conditions. Appl Environ Microbiol72: 2885-2895.

Morozova, 1., Qu, X., Shi, S., Asamani, G., Greenberg, J.E., Shuman, H.A., and Russo, J.1.(2004) Comparative sequence analysis of the icm/dot genes in Legionella. Plasmid51: 127-147.

Nash, T.W., Libby, D.M., and Horwitz, M.A (1984) Interaction between the legionnaires'disease bacterium (Legionella pneumophila) and human alveolar macrophages.Influence of antibody, lymphokines, and hydrocortisone. J Clin Invest 74: 771-782.

Obert, S., O'Connor, R.J., Schmid, S., and Hearing, P. (1994) The adenovirus E4-617 proteintransactivates the E2 promoter by inducing dimerization of a heteromeric E2Fcomplex. Mol Cell Bioi 14: 1333-1346.

Purcell, M., and Shuman, H.A (1998) The Legionella pneumophila icmGCDJBF genes arerequired for killing of human macrophages. Infect Immun 66: 2245-2255.

Roy, c.R., and Tilney, L.G. (2002) The road less traveled: transport of Legionella to theendoplasmic reticulum. J Cell Bioi 158: 415-419.

Sadosky, AB., Wiater, L.A., and Shuman, H.A. (1993) Identification of Legionellapneumophila genes required for growth within and killing of human macrophages.Infect Immun 61: 5361-5373.

Segal, G., Purcell, M., and Shuman, H.A. (1998) Host cell killing and bacterial conjugationrequire overlapping sets of genes within a 22-kb region of the Legionellapneumophila genome. Proc Natl Acad Sci USA 95: 1669-1674.

Segal, G., and Shuman, H.A (1998) Intracellular multiplication and human macrophagekilling by Legionella pneumophila are inhibited by conjugal components of IncQplasmid RSF1010. Mol Microbiol30: 197-208.

Segal, G., and Shuman, H.A (1999) Legionella pneumophila utilizes the same genes tomultiply within Acanthamoeba castellanii and human macrophages. Infect Immun 67:2117-2124.

109

Page 111: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Characterization of IcmG

Solomon, J.M., Rupper, A., Cardelli, J.A., and Isberg, R.R. (2000) Intracellular growth ofLegionella pneumophila in Dictyostelium discoideum, a system for genetic analysis ofhost-pathogen interactions. Infect Immun 68: 2939-2947.

van der Goot, F.G., Tran van Nhieu, G., Allaoui, A., Sansonetti, P., and Lafont, F. (2004)Rafts can trigger contact-mediated secretion of bacterial effectors via a lipid-basedmechanism. J Bioi Chem 279: 47792-47798.

Viala, J., Chaput, C., Boneca, LG., Cardona, A., Girardin, S.E., Moran, A.P., Athman, R.,Memet, S., Huerre, M.R., Coyle, AJ., DiStefano, P.S., Sansonetti, PJ., Labigne, A.,Bertin, J., Philpott, DJ., and Ferrero, R.L. (2004) NodI responds to peptidoglycandelivered by the Helicobacter pylori cag pathogenicity island. Nat Immunol5: 1166­1174.

Vogel, J.P., Andrews, H.L., Wong, S.K., and Isberg, R.R. (1998) Conjugative transfer by thevirulence system of Legionella pneumophila. Science 279: 873-876.

Weber, S.S., Ragaz, C., Reus, K., Nyfeler, Y., and Hilbi, H. (2006) Legionella pneumophilaexploits PI(4)P to anchor secreted effector proteins to the replicative vacuole. PLoSPathog 2: e46.

110

Page 112: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 5

5 General discussion

5. 1 LcsC shows lipid A disaccharide synthase activity

In the amoebae plate test, an L. pneumophila genomic library was screened for genes

increasing the survival of L. pneumophila on plates in presence of A. castellanii. The screen

identified IcsC, a gene conferring cytotoxicity to the bacteria (see Chapter 2, Albers et al.,

2005). Search for homologues revealed that LcsC (LpxB1) shows 34% identity on protein

level to the E. coli lipid A disaccharide synthase LpxB. Interestingly, the L. pneumophila

genome contains a second LpxB homologue (LpxB2) with 43% identity to the E. coli

enzyme. In almost all cases, the genes necessary for lipid A biosynthesis are present in only

one copy (Raetz and Whitfield, 2002). Therefore, we examined the genomic vicinity of both

IpxB paralogues to assess whether one of the genes might have another function than a lipid

A disaccharide synthase. LpxB2 is located within a cluster of other genes involved in lipid A

biosynthesis. In contrast, IpxBl/lcsC is not located in the vicinity of genes belonging to the

classical lipid A biosynthesis pathway. Instead, genes involved in peptidoglycan metabolism

were found, and a domain in LcsC is distantly related to MurG, a glycosyl transferase

involved in peptidoglycan biosynthesis. Therefore, we hypothesized that LcsC might be

involved in peptidoglycan metabolism and cytotoxicity might be caused by the release of

toxic peptidoglycan fragments, as described for a number of other bacteria (Burroughs et al.,

1993; Cloud and Dillard, 2002; Luker et al., 1993).

However, a closer inspection of lipid A biosynthesis and the genes involved revealed that the

gene immediately upstream of IcsC encodes a homologue of the Acidithiobacillus

ferrooxidans oxidase GnnA. The first step of lipid A biosynthesis in A. ferrooxidans and most

likely also in L. pneumophila is the conversion of UDP-GlcNAc to UDP-GlcNAc3N, which

is catalyzed by GnnA and GnnB (Sweet et al., 2004). Reverse transcription and PCR proved

that gnnA and IcsC are cotranscribed, suggesting that the corresponding two enzymes are

employed in the same biosynthetic pathway. Furthermore, both IpxBlIlcsC and IpxB2

complemented an E. coli IpxB mutant, demonstrating their activity as lipid A disaccharide

synthases. Therefore, it can be concluded that IcsC encodes a lipid A disaccharide synthase

rather than an enzyme involved in peptidoglycan synthesis.

111

Page 113: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General Discussion

5.2 L. pneumophila: life in different niches and adaptation of lipidA structure

L. pneumophila is able to survive under highly diverse conditions. In the environment, the

bacterium colonizes and persists in biofilms and multiplies within various protozoa (Fields,

1996). After lysing a spent host, the bacteria have to survive in the aquatic environment or in

soils, until they are phagocytosed by another host celL In macrophages, L. pneumophila

resides in a vacuole that finally fuses with lysosomes and matures into an acidic compartment

(Sturgill-Koszycki and Swanson, 2000). The drastic differences in the conditions within these

different niches require adaptation of the bacterium. To adjust their physiology, the

L. pneumophila life cycle comprises of bacterial differentiation into a replicative state,

designated to multiply within a host cell, and in a transmissive state, in which the bacteria

adjust their physiology to survive in the environment and to promote infection of a new host

cell (Molofsky and Swanson, 2004). Differentiation is directed by global regulatory circuits,

and it is tempting to speculate that also the outer membrane, the outermost protective barrier,

undergoes changes to adapt to the highly diverse environments. Lipid A is a major part of the

outer membrane. All genes in L. pneumophila encoding the lipid A acyl transferases and the

lipid A disaccharide synthase are present in more than one copy, and at least the lpxA and the

lpxB paralogues are differentially expressed (see Chapter 3). Examination of a phase variant

strain showed that L. pneumophila is able to alter the composition of the primary fatty acids

of lipid A. It is therefore conceivable, that the lipid A biosynthesis paralogues are used to

adjust the lipid A structure, most probably the fatty acid composition, by differential

expression of paralogous genes with the purpose to adapt the properties of the outer

membrane to the diverse environmental conditions.

The amoeba plate test was applied to identify genes increasing the survival of the partially

growth defective icmG mutant on agar plates in presence of amoeba (see Chapter 2, Albers et

al., 2005). The identification of the lipid A disaccharide synthase lpxBl/lcsC in this screen is

an evidence for the importance of lipid A structure for the interaction of L. pneumophila with

A. castellanii, and it is likely that a lipid A structure synthesized by LcsC is required for a

successful infection of amoebae.

The impact of lipid A structure, particularly the fatty acid composition, on virulence is further

supported by literature describing the effect of mutations of genes involved in lipid A

acylation, e.g., pagP, IpxP or IpxM in different pathogenic bacteria (Guo et al., 1998; Raetz

and Whitfield, 2002; see also Chapter 1). Deletion of these genes alters the fatty acid

112

Page 114: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 5

composition of lipid A, and thus influences sensitivity to antimicrobial peptides or antibiotics

or alters TLR4 dependent activation of the immune response. Therefore, a well regulated

adaptation of lipid A structure is expected to be a valuable tool for 1. pneumophila to survive

under diverse conditions and to multiply in a variety of host cells. Differential expression of

paralogous enzymes is an adequate means to perform such structural modifications.

5.3 Intracellular LPS influences host cell functions

Interference of Legionella LPS and material derived from the outer membrane with host cell

functions has been described in the literature, and it is conceivable that modifying the

structure of the membrane anchor lipid A will have an influence on the effect of LPS on host

cell functions.

For LPS from Legionellajeonii (Park et al., 2004), the ability to alter vesicular trafficking in

Acanthamoeba proteus has been suggested. Microinjection of an anti-LPS antibody into the

host cell increased the number of phagosomes that fused with lysosomes, indicating that LPS

assists in the prevention of phagosome-lysosome fusion (Kim et al., 1994).

Membrane-derived vesicles from 1. pneumophila have recently been shown to inhibit

phagosome-lysosome fusion in an !cm/Dot-independent manner. The ability of these outer

membrane-derived vesicles to interfere with host vesicle fusion correlated with

developmentally regulated modifications of the pathogen's surface. Bacterial surface

properties were assayed by LPS profile and binding to a lectin, suggesting that the sugar

structure of LPS is altered during the switch from replicative to transmissive phase

(Fernandez-Moreira et al., 2006). The correlation of the ability to inhibit vesicle fusion and

LPS properties and the influence of an anti-LPS antibody on vesicle fusion in amoebae

infected with 1. jeonii render LPS a candidate molecule for interference with host vesicle

trafficking.

Intracellular LPS distribution and interference with host cell mechanisms was examined for

Brucella, a persistent intracellular pathogen leading to chronic diseases in a wide variety of

mammals (Lapaque et al., 2005). Similarly to 1. pneumophila LPS, Brucella LPS has a

GlucN3N-based disaccharide backbone with unusually long acyl chains and a low endotoxic

activity. Brucella LPS impairs phagosome-lysosome fusion during the first hours of infection

(comparable to the effect of 1. pneumophila membrane vesicles, Fernandez-Moreira et al.,

2006), travels through the endocytic route and can later be found on the cell surface in

113

Page 115: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General Discussion

detergent-resistant membrane regions called macrodomains. These domains have a reduced

membrane fluidity and remain stable in macrophages for several months (Lapaque et al.,

2006). There, LPS interferes with MHC-II presentation to specific CD4+ T cells, probably to

control host immune response by prevention of Brucella-specific T cell activation.

Release and delivery of LPS to target compartments of the cell is a prerequisite for acting as a

virulence factor. LPS release and distribution within numerous host cell vesicles was

examined in detail for the infection of eukaryotic cell lines with S. Typhimurium and was

found to occur exclusively when bacteria localize inside of a cell (Garcia-del Portillo et al.,

1997). Salmonella LPS was detectable 30 minutes after infection in areas surrounding

internalized bacteria and at 6 hours p. i. in numerous vesicles throughout the cytosol,

including vesicles not containing intracellular bacteria. The occurrence of LPS depended on

intracellular presence of bacteria, since addition of purified LPS to the medium did not

produce LPS-positive vacuoles even after induction of ruffling and macropinocytosis. Also,

uninfected cells neighbouring cells containing bacteria remained unstained. Interestingly, the

ability to spread within the cell was determined by the structure of LPS, since neither E. coli

LPS added simultaneously with infecting Salmonella nor LPS from Salmonella mutants with

altered LPS structures were released to host vesicles as efficiently as wild type LPS.

Trafficking and localization of L. pneumophila LPS has not yet been examined throughout

the whole course of infection. However, data from the literature suggest an involvement of

intracellular LPS in virulence. It is not unlikely that the structure of the membrane anchor

lipid A influences the intracellular trafficking of LPS to a large extent. Modification of lipid

A could determine localization and interactions of LPS with intracellular targets and thus

direct the effects of LPS on virulence.

5.4 Possible influence of lipid A on outer membrane vesicles

Membrane-derived vesicles from L. pneumophila inhibit phagosome-lysosome fusion,

demonstrating their role in pathogenicity (Fernandez-Moreira et al., 2006). Outer membrane

vesicles (OMV) are naturally produced during growth of most Gram-negative bacteria.

£. coli packages 0.2-0.5% of outer membrane and periplasmic proteins in vesicles (Kuehn

and Kesty, 2005). The OMVs are produced by outer membrane budding during growth and

are composed of proteins and lipids of the outer membran and periplasmic material, but not

of inner membrane or cytoplasmic components. The vesicles are capable to fuse with other

114

Page 116: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 5

bacterial and eukaryotic membranes and deliver their content, which can range from enzymes

and toxins to antibiotic compounds and DNA, to the recipient. Vesicle surface molecules can

mediate adhesion to host cells and subsequent internalization of vesicle material (Kuehn and

Kesty, 2005).

Acylation of lipid A affects the physical properties of the outer membrane bilayer. It might be

that changing lipid A fatty acid composition by differential use of distinct acyl transferase

paralogues will have an impact on number, size or properties of released outer membrane

vesicles. For example, altered membrane fluidity might change the frequency of membrane

budding, the fusogenic properties of the vesicles, or affect the size of vesicles formed and

thereby the amount of lipids and vesicle content transported. Therefore, modifying outer

membrane properties by adjusting lipid A acylation might control the formation and release

of OMVs and affect their role in virulence.

5.5 Cytotoxicity of the IpxB paralogues

Overexpression of lpxBl/lcsC increases the ability of L. pneumophila to kill amoebae. We

were interested to know whether lpxB2 overexpression also shows an effect but could not

find an increase in cytotoxicity. However, this result may be compromised by the toxic effect

of lpxB2 overexpression on the bacteria. Overexpression of lpxB2 impairs growth of the icmG

mutant on plates more strongly than overexpression of lcsC. Overexpression of lcsC on plates

with 0.5 mM IPTG yields colonies with an only slightly reduced diameter, whereas induction

of lpxB2 expression with the same concentration inhibits growth of l1icmG. The cytotoxicity

assay was performed with bacteria from plates. It might be that the ability of lpxB2­

expressing bacteria to infect and kill amoebae was impaired due to the larger toxicity of

lpxB2, and thus a possible cytotoxic effect was diminished. To exclude this possibility, it will

be necessary to control the plating efficiency of the bacteria used for infection, and also to

determine whether lpxB2-overexpressing bacteria are impaired in intracellular growth

compared to lcsC/lpxBl-expressing strains.

115

Page 117: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General Discussion

5.6 Putative mechanisms of LcsC mediated cytotoxicity

The lipid A disaccharide synthase paralogue LcsC increases bacterial survival in presence of

A. castellanii due to its cytotoxic effect on amoebae (see Chapter 2, Albers et al., 2005). The

mechanism of cytotoxicity remained unclear. Likely, overexpression of IcsC alters the

composition of lipid IVA by favoring synthesis of an LcsC-derived molecule. This lipid IVA

molecule might be used to synthesize the full LPS molecule, and then exert toxic effects

during infection. However, it is not clear whether LPS with an LcsC-derived lipid A moiety

is the cytotoxic agent, or whether overexpression of IcsC leads to accumulation of lipid IVA

or other products of the LPS biosynthesis pathway which exert the cytotoxic effect.

Furthermore, it can not be excluded that indirect effects of IcsC overexpression increase

cytotoxicity. As a last speculative possibility, the enzyme itself could enter the eukaryotic

host cell and disturb cellular saccharide or lipid metabolism.

LPS can be released and distributed within the cell by intracellular bacteria and interfere with

host cell mechanisms, e.g., inhibit vesicle fusion. If Legionella LPS is distributed throughout

many different host cell vesicle membranes, it might be that LPS or LPS derivatives released

from intracellularly growing bacteria lead to a severe disturbance of host vesicle trafficking,

which could finally cause cell death. The cytotoxicity assays with amoebae infected with

IcsC-overexpressing strains were performed two days after infection, and it is reasonable to

speculate that this period of time will be sufficient to kill an infected amoebae by interruption

of vesicular trafficking.

The question remains why IcsC needs to be overexpressed to increase cell death. It might be

that IcsC overexpression leads to an increase in lipid A or LPS release, damaging the host by

elevated LPS concentrations in the membranes. Alternatively, IcsC overexpression could

enlarge the proportion of LcsC/LpxB I-derived lipid A compared to LpxB2-derived lipid A.

The putative differences in fatty acid composition of LcsC- or LpxB2-derived lipid A might

lead to different interactions with the host, e.g., the ability of LPS to persist in specific

membrane domains or to associate with host membrane factors is distinct. LcsC-derived lipid

A molecules might therefore act at other interaction sites or have a greater stability, thus

altering the place or duration of putative LPS interactions and damaging the host more

severely. Differences in release or distinct interaction sites of LcsC- and LpxB2-derived

molecules could also explain the difference in cytotoxicity between the IcsC/lpxBl and IpxB2

overexpressing strains.

116

Page 118: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 5

The cytotoxic effect of lcsC overexpression could also be connected to the fonnation of outer

membrane vesicles. Release of LPS from intracellular bacteria could occur in fonn of OMVs.

Fusion of OMVs with the membrane of the LCV could be a mechanism by which LPS is

inserted into cellular membranes, from where it is distributed throughout the eukaryotic cell.

Increasing OMV release could increase the amount ofLPS (or other outer membrane factors)

that is inserted into host cell membranes and is available to interfere with host cell functions.

Since during fusion of an OMV with the LCV membrane the contents of the OMV are

released into the host cell cytosol, an increase in OMV fonnation could also lead to a more

massive disposal of putative toxic vesicle contents into the cytosol.

Both increased LPS transfer into host membranes and increased release of toxic molecules

from OMV content could cause the lcsC-mediated cytotoxicity observed.

5.7 Specifity of LcsC-mediated cytotoxicity for AicmG

Overexpression of lcsC rendered the icmG mutant strain cytotoxic, but not other mutants with

partial phenotypes. A reliable explanation why lcsC cytotoxicity is restricted to !:J.icmG

remains difficult unless the toxic compound itself is identified and the function of lemG is

known in more detail. However, the data collected so far allow some speculations.

If an icmG mutant releases only a subset or an insufficient amount of effector proteins, the

trafficking of the icmG mutant might be different from that of other partially virulence­

defective mutants. The icmG mutant-containing vacuole could interact with other vesicles,

allowing putative toxic LcsC-derived molecules to travel from the LCV membrane to host

organelles that are not affected in strains other than !:J.icmG. There, they could interfere with

host processes in a way detrimental for the cell.

An alternative explanation presumes that lemG regulates the selective transfer of molecules

through the lern/Dot secretion system. If IcmG acts as a regulatory protein, it might be that

lack of the protein leads to leakiness and uncontrolled release of molecules. Thus, toxic LcsC

products could enter the host cell via the type IV secretion system and kill the amoebae.

117

Page 119: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

General Discussion

5.8 Conclusion

Using the amoebae plate test, we screened for genes involved in interactions of

L. pneumophila with A. castellanii. Genes were identified whose overexpression lead to an

increased killing of amoebae. Among the genes identified, IcsC showed the strongest

cytotoxic effect. It could be demonstrated that IcsC encodes a lipid A disaccharide synthase.

LcsC and the acyl transferases necessary for lipid A biosynthesis are present in more than a

single copy in the genome of 1. pneumophila, and at least the IpxA and IpxB paralogues are

differentially regulated. These findings suggest that L. pneumophila modifies its lipid A

structure by use of distinct enzymes to adapt to different environmental conditions. Many

examples in the literature show that the lipid A structure affects different aspects of bacterial

survival and bacteria-host interactions in L. pneumophila and other pathogenic and

nonpathogenic species.

Currently available data only allow speculations about the role and effects of lipid A

modifications in L. pneumophila. Detailed analysis of lipid A biosynthesis gene regulation,

biochemical functions of the enzymes, the structures of lipid A synthesized by different

paralogues and the localization of lipid A and LPS during infection will be necessary to get a

better understanding of the biological meaning and the effects of L. pneumophila lipid A

variations.

5.9 Literature

Albers, U., Reus, K., Shuman, H.A., and Hilbi, H. (2005) The amoebae plate test implicates aparalogue of IpxB in the interaction of Legionella pneumophila with Acanthamoebacastellanii. Microbiology 151: 167-182.

Burroughs, M., Rozdzinski, E., Geelen, S., and Tuomanen, E. (1993) A structure-activityrelationship for induction ofmeningeal inflammation by muramyl peptides. J ClinInvest 92: 297-302.

Cloud, K.A., and Dillard, J.P. (2002) A lytic transglycosylase ofNeisseria gonorrhoeae isinvolved in peptidoglycan-derived cytotoxin production. Infect Immun 70: 2752-2757.

Femandez-Moreira, E., Helbig, lH., and Swanson, M.S. (2006) Membrane vesicles shed byLegionella pneumophila inhibit fusion of phagosomes with lysosomes. Infect Immun74: 3285-3295.

Fields, B.S. (1996) The molecular ecology of Legionellae. Trends Microbiol4: 286-290.

118

Page 120: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Chapter 5

Garcia-del Portillo, F., Stein, M.A., and Finlay, B.B. (1997) Release oflipopolysaccharidefrom intracellular compartments containing Salmonella typhimurium to vesicles of thehost epithelial celL Infect Immun 65: 24-34.

Guo, L., Urn, K.B., Poduje, C.M., Daniel, M., Gunn, J.S., Hackett, M., and Miller, S.I.(1998) Lipid A acylation and bacterial resistance against vertebrate antimicrobialpeptides. Cell 95: 189-198.

Kim, K.J., Na, YE., and Jeon, K.W. (1994) Bacterial endosymbiont-derivedlipopolysaccharides and a protein on symbiosome membranes in newly infectedamoebae and their roles in lysosome-symbiosome fusion. Infect Immun 62: 65-71.

Kuehn, M.J., and Kesty, N.C. (2005) Bacterial outer membrane vesicles and the host­pathogen interaction. Genes Dev 19: 2645-2655.

Lapaque, N., Moriyon, I., Moreno, E., and Gorvel, lP. (2005) Brucella lipopolysaccharideacts as a virulence factor. Curr Opin Microbiol8: 60-66.

Lapaque, N., Forquet, F., de Chastellier, C., Mishal, Z., Jolly, G., Moreno, E., Moriyon, I.,Heuser, J.K, He, H.T., and Gorvel, lP. (2006) Characterization of Brucella abortuslipopolysaccharide macrodomains as mega rafts. Cell Microbiol8: 197-206.

Luker, K.E., Collier, J.L., Kolodziej, KW., Marshall, G.R., and Goldman, W.E. (1993)Bordetella pertussis tracheal cytotoxin and other muramyl peptides: distinct structure­activity relationships for respiratory epithelial cytopathology. Proc Natl Acad Sci USA 90: 2365-2369.

Molofsky, A.B., and Swanson, M.S. (2004) Differentiate to thrive: lessons from theLegionella pneumophila life cycle. Mol Microbiol53: 29-40.

Park, M., Yun, S.T., Kim, M.S., Chun, J., and Ahn, T.I. (2004) Phylogenetic characterizationof Legionella-like endosymbiotic X-bacteria in Amoeba proteus: a proposal for'Candidatus Legionellajeonii' sp. novo Environ Microhiol6: 1252-1263.

Raetz, C.R., and Whitfield, C. (2002) Lipopolysaccharide endotoxins. Annu Rev Biochem 71:635-700.

Sturgill-Koszycki, S., and Swanson, M.S. (2000) Legionella pneumophila replicationvacuoles mature into acidic, endocytic organelles. J Exp Med 192: 1261-1272.

Sweet, C.R., Ribeiro, A.A., and Raetz, C.R. (2004) Oxidation and transamination of the 3"­position ofUDP-N-acetylglucosamine by enzymes from Acidithiobacillusferrooxidans. Role in the formation oflipid a molecules with four amide-linked acylchains. J Bioi Chem 279: 25400-25410.

119

Page 121: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Curriculum Vitae

Personal Data

Date of Birth

Citizenship

Education

2002 - now

2001 - 2002

1999 - 2000

1994 - 2000

1991 - 1994

1990 - 1991

1981 - 1990

Curriculum Vitae

Urs Albers

April 29th, 1971, Kleve, Germany

German and Swiss (Baden, AG)

ETH ZUrich, Institute of Microbiology,PhD Thesis

ETH ZUrich, Institute of Biotechnology

Genentech Inc., USA,Diploma work

Technical University Braunschweig, GermanyStudy of Biotechnology

Naturwissenschaftliches Technikum Landau, GermanyTraining as Biological Technical Assistant

Military service, Germany

Freiherr-vom-Stein-Gymnasium, Kleve, Germany

120

Page 122: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Publications

2006

2006

2005

2001

1999

Publications

R. Sporri, N. Joller, U. Albers, H. Hilbi and A. Oxenius:MyD88-dependent interferon gamma production by NK cellsis key for control of Legionella pneumophila infection.J. Immunol. 176(10): 6162-71

U. Albers, K. Reus and H. Hilbi: Identification of a cytotoxicLegionella pneumophila IpxB paralogue in a multicopysuppressor screen using Acanthamoeba castellanii as aselective host. In: Cianciotto, N. et al., Proceedings of the 6th

International Conference on Legionella, Chicago, October16-20, 2005. ASM Press, Washington, DC

U. Albers, K. Reus, H. Shuman and H. Hilbi:The amoebae plate test implicates a paralogue of IpxB in theinteraction of Legionella pneumophila with Acanthamoebacastellanii. Microbiology 151 (1): 167-182

T. Larson, M. Gawlitzek, H. Evans, U. Albers and J. Cacia:Chemometric Evaluation of On-Line High PerformanceChromatography in Mammalian Cell Culture: Monitoring ofAmino Acids and Glucose. Biotechnol. Bioeng. 77(5): 553­563

N. Schulze, U. Albers, E. Grabenhorst, H.S. Conradt, M.Rohde, M. Nimtz and V. Jager: Intracellular localization ofnon-secreted recombinant p-Trace protein in vesicle-likestructures of baculovirus-infected insect cells. In: Bernard A.,Griffiths B., Noe W., Wurm F. (Eds.): Animal CellTechnology: Products from Cells, Cells as Products.Proceedings of the 16th ESACT Meeting April 1999, Lugano,Switzerland. Kluwer Academic Publishers

121

Page 123: dspace cover page - Research Collection29018/... · cytotoxic Legionella pneumophila lipid A disaccharide synthase ... ofa cytotoxic Legionella pneumophila lipid A ... dass 1. pneumophila

Acknowledgements

Danksaqunq

Meinem Doktorvater Hubert Hilbi mOchte ich sehr fUr die Betreuung dieser Arbeitdanken, fOr seinen Optimismus, die vielen Ideen, und insbesonders fOr dieBereitschaft, jederzeit die Projekte oder auftretende Schwierigkeiten zu diskutieren.

Wolf-Dietrich Hardt danke ich fOr die Obernahme des Koreferats und die vielenstimulierenden und hilfreichen Diskussionen und Anregungen wahrend dergemeinsamen Hardt-Hilbi-Gruppenseminare.

Ein ganz besonderer Dank geht an die ehemaligen und jetzigen Mitglieder der Hilbi­Gruppe fOr die tolle Atmosphare im Lab! Die freundschaftliche Stimmung, die vielenwissenschaftlichen und nichtwissenschaftlichen Diskussionen, die vielen klugen unddummen SprUche und das gemeinsame Lachen haben viel dazu beigetragen, dassich, auch wenn mal wieder ein Experiment nicht lief wie erwartet, am Morgen gerneins Institut gekommen bin und wir viel Spass zusammen hatten.Ein herzliches DankeschOn an Claudius, der durch seinen Einsatz in seinerDiplomarbeit wichtige Erfahrungen fOr diese Arbeit gesammelt hat.Vielen Dank an Claudius, Curd in, Gunnar, Holger, JOrg, Katrin, Monika, Naja,Natalie, Nicki, Rachelle, Simon, Stefan, Thomas, und Yves!Und wenn wieder etwas nicht klappt, nicht verzweifeln: It's not you, it's just the data!

Danke an die Mitarbeiter des Institutes: die bei alien vorhandene Bereitschaft, sichmit den Fragen und Problemen der anderen zu beschaftigen und gegenseitigWissen auszutauschen, habe ich als sehr positiv erlebt.Besonderer Dank gilt dabei den "Hardts": das gute Arbeitsklima und der steteAustausch von Wissen, Ideen, Materialien und Geraten hat vieI geholfen!

Der wichtigste Dank gebOhrt meinen Eltern: sie erst haben mir durch ihreUnterstotzung in aller Form den Weg bis hier ermOglicht!Danke!

122