Download The amoebae plate test implicates a paralogue of lpxB in the

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

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

Document related concepts

Magnesium transporter wikipedia , lookup

Signal transduction wikipedia , lookup

JADE1 wikipedia , lookup

List of types of proteins wikipedia , lookup

Transcript
Microbiology (2005), 151, 167–182
DOI 10.1099/mic.0.27563-0
The amoebae plate test implicates a paralogue of
lpxB in the interaction of Legionella pneumophila
with Acanthamoeba castellanii
Urs Albers,1 Katrin Reus,1 Howard A. Shuman2 and Hubert Hilbi1
Correspondence
Hubert Hilbi
[email protected]
Received 18 August 2004
Revised
13 October 2004
Accepted 14 October 2004
1
Institute of Microbiology, Swiss Federal Institute of Technology (ETH), Wolfgang-Pauli Str. 10,
HCI G405, 8093 Zürich, Switzerland
2
Department of Microbiology, College of Physicians and Surgeons, Columbia University,
701 West 168th Street, New York, NY 10032, USA
Legionella pneumophila is a bacterial parasite of freshwater 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 Icm/Dot type IV secretion system. This paper reports the development of an assay, the
amoebae plate test (APT), to analyse growth of L. pneumophila wild-type and icm/dot mutant
strains spotted on agar plates in the presence of Acanthamoeba castellanii. In the APT, wild-type
L. 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 L. pneumophila chromosomal library for genes that suppress the
growth defect in the presence of the amoebae. An icmS suppressor plasmid was isolated that
harboured the icmS and flanking icm genes, indicating that this plasmid complements the
intracellular growth defect of the mutant. In contrast, 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 as 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 paralogue of the lipid A
disaccharide synthase LpxB, increased cytotoxicity of an icmG mutant but not that of other icm/dot
or rpoS mutant strains against A. castellanii. Based on sequence comparison and chromosomal
location, lcsB and lcsC probably 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.
INTRODUCTION
The Gram-negative bacterium Legionella pneumophila is
a natural parasite of freshwater 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, L. pneumophila establishes a pH-neutral vacuole
that does not fuse with lysosomes (Horwitz & Maxfield,
1984; Horwitz, 1983a). This unique, replication-permissive
Abbreviations: APT, amoebae plate test; CYE, charcoal-yeast extract;
FCS, fetal calf serum; icm/dot, intracellular multiplication/defective
organelle trafficking; lcs, Legionella cytotoxic suppressor; PI, propidium
iodide; PYG, proteose-yeast extract-glucose.
0002-7563 G 2005 SGM
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
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Printed in Great Britain
167
U. Albers and others
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 icm/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 IcmR/IcmQ directly
bind to each other (Coers et al., 2000). IcmR functions as a
chaperone of IcmQ, preventing and reversing its aggregation into high-molecular-mass complexes that form pores
in lipid membranes (Duménil & Isberg, 2001; Duménil
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, Icm/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 ARF1 to the L. pneumophila 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 Icm/
Dot-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 which comprise families of up to five
paralogues (Luo & Isberg, 2004). Secretion into culture
supernatants of the polytopic membrane protein DotA
requires the Icm/Dot secretion system but neither IcmS
nor IcmW, and it might lead to the formation of pores in
target membranes (Nagai & Roy, 2001).
The Icm/Dot-dependent establishment of the L. pneumophila vacuole eventually leads to host cell death due to
intracellular replication and lysis. Additionally, the icm/
dot genes mediate cytotoxic events such as (i) contactdependent 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 of the RTX (repeats in toxin)
family of cytotoxic adhesins (Cirillo et al., 2001).
In this report, we describe the amoebae plate test (APT), a
novel assay to analyse growth of L. pneumophila wild-type
168
and icm/dot mutants on agar plates in the presence of A.
castellanii. The APT was used to screen an L. 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
probably involved in peptidoglycan metabolism were
identified.
METHODS
Bacterial strains, cell culture and reagents. The bacterial
strains and plasmids used in this study are listed in Table 1.
Escherichia coli was cultured on LB agar plates or in LB broth,
supplemented with chloramphenicol (Cm, 30 mg ml21), sodium
ampicillin (Ap, 100 mg ml21) or kanamycin sulfate (Km, 50 mg
ml21) if required. Legionella pneumophila was grown on charcoalyeast extract (CYE) agar plates (Feeley et al., 1979) or in ACESbuffered-yeast extract broth (AYE) containing 0?5 % bovine serum
albumin (BSA) (Horwitz & Silverstein, 1983). Supplements for
L. pneumophila were used at the following concentrations: Cm,
5 mg ml21; Km, 50 mg ml21; IPTG, 0?5 mM. Acanthamoeba castellanii (ATCC 30234) was grown in proteose-yeast extract-glucose
medium (PYG) at 30 uC (Moffat & Tompkins, 1992; Segal &
Shuman, 1999b) and subcultured once or twice a week. RAW264.7
macrophages (kindly supplied by Dr David Underhill, University of
Washington, Seattle, WA, USA) were cultivated in RPMI1640
medium supplemented with 10 % fetal calf serum (FCS) and 2 mM
L-glutamine at 37 uC in 5 % CO2. High-gel-strength agar was from
Serva, proteose peptone from Becton Dickinson Biosciences and
Bacto yeast extract from Difco. RPMI medium, glutamine and FCS
were from Omnilab. All other reagents were from Sigma.
Construction of plasmids. Plasmids were isolated using commer-
cially 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, pUA11, pUA17 and pUA27 (Table 1; see
also 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 oligonucleotides listed in
Table 2 were used. The PCR fragments were phosphorylated, circularized, transformed into E. coli TOP10, 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, ORF1-3 was deleted from pG39 or pG54
by digestion with HindIII or SphI (see Fig. 5a), respectively, and
religated, thus yielding pUA20 and pUA22. To overexpress lcsA, -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 pMMB207-RBS, yielding
pMMB207-RBSlcsA–D. pMMB207-RBS harbours the Ptac promoter
and the ribosome-binding site from the T7 gene10 taken from pGSGFP-04 (Hilbi et al., 2001). The sequence of all PCR fragments was
confirmed by sequencing.
The amoebae plate test (APT). In this test, A. castellanii is
spread on CYE agar plates prior to spotting bacteria in serial dilutions onto these plates. Thus, the APT allows growth of bacteria to
be determined on solid medium in the presence of amoebae. A. castellanii cultures were fed with PYG 2 days prior to an experiment.
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151
Cytotoxic L. pneumophila lpxB paralogue
Table 1. Bacterial strains and plasmids
Strain or plasmid
E. coli
DH5a
LW253
TOP10
L. pneumophila
GS3001
GS3002
GS3003
GS3007
GS3011
GS3012
JR32
LELA1275
LELA2883
LELA3473
MW635
MW645
Plasmids
MW66
pGS-BCD-05
pGS-Lc-34-14
pGS-Lc-36b
pGS-Lc-37-14
pGS-Lc-63-14
pMMB207
pMMB207ab
pMMB207ab-Km-14
pMMB207-RBS
pUA3
pUA5
pUA9
pUA11
pUA13
pUA15
pUA17
pUA19
pUA20
pUA22
pUA26
pUA27
pUA29
pUA34
pUA35
pUA36
pUA41
pUC19
pUC4K
Relevant properties*
F9
F2 mcrA D(mrr–hsdRMS–mcrBC) w80lacZDM15 DlacX74 deoR recA1
araD139 D(ara–leu)7697 galU galK rpsL (Sm) endA1 nupG
Reference
Laboratory stock
Wiater et al. (1994)
Invitrogen
JR32 icmS3001 : : Km
JR32 icmP3002 : : Km
JR32 icmO3003 : : Km
JR32 icmN3007 : : Km
JR32 icmT3011 : : Km
JR32 icmR3012 : : Km
Salt-sensitive isolate of AM511
JR32 icmF1275 : : Tn903dIIlacZ
JR32 dotB2883 : : Tn903dIIlacZ
JR32 icmR3473 : : Tn903dIIlacZ
JR32 icmG : : Km
JR32 icmC : : Km
Segal & Shuman (1997)
Segal & Shuman (1997)
Segal & Shuman (1997)
Segal et al. (1998)
Segal & Shuman (1998)
Hilbi et al. (2001)
Sadosky et al. (1993)
Purcell & Shuman (1998)
Sadosky et al. (1993)
Sadosky et al. (1993)
Purcell & Shuman (1998)
Purcell & Shuman (1998)
L. pneumophila Philadelphia-1 chromosomal library, partial EcoRI
fragments (5–10 kb) in pMMB207
dotBCD in pMMB207ab-Km-14
icmPO in pMMB207ab-Km-14
icmR in pMMB207ab
icmTS in pMMB207ab-Km-14
icmGCD in pMMB207-Km-14
RSF1010 derivative, IncQ, oriT lacIq Ptac Cm
pMMB207, lacZa, MCS
pMMB207ab with mobA : : Km
pMMB207 with ribosome-binding site (T7 gene10)
pUC19-G54DlcsB
pG54-lcsB (DNlpD-like protease)
pUC19-G65DlcsC
pUC19-G65Dfis
pG65-fis (DFis)
pG65-lcsC (DLpxB homologue)
pUC19-G34DlcsA
pG34-lcsA (Dhydrolase)
pG39-ORF1-3 (HindIII deletion)
pG54-ORF1-3 (SphI deletion)
pMMB207-RBSlcsC (pLcsC)
pUC19-G66DlcsD
pG66-lcsD (DABC transporter)
pMMB207-RBSlcsA (pLcsA)
pMMB207-RBSlcsB (pLcsB)
pMMB207-RBSlcsD (pLcsD)
pG65-lcsC-fis (DLcsC, DFis)
oriR (ColE1), Ap, MCS
oriR (pBR322), Ap, MCS : : Km
Purcell & Shuman (1998)
Hilbi et al. (2001)
Segal & Shuman (1998)
Segal & Shuman (1997)
Segal & Shuman (1998)
Segal et al. (1998)
Morales et al. (1991)
Segal & Shuman (1997)
Segal & Shuman (1998)
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
This study
Yanisch-Perron et al. (1985)
Amersham
*Abbreviations: Ap, ampicillin resistance; Cm, chloramphenicol resistance; Km, kanamycin resistance; Sm, streptomycin resistance.
http://mic.sgmjournals.org
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
169
U. Albers and others
Table 2. Oligonucleotides
Oligo
pMMB207-59
pMMB207-39
oKanStart
oKanStop
oUA3
oUA4
oUA5
oUA6
oUA7
oUA8
oUA19
oUA20
oUA21
oUA22
oUA23
oUA24
oUA25
oUA26
oUA27
oUA28
oUA29
oUA30
oUA31
Sequence
Comment
ATTGTGTGGAATTGTGAGCG
CTTCTCTCATCCGCCAAAAC
GAATATGGCTCATAACACCCC
TCACTTTCTGGCTGGATGATGG
TCCAGCTTTTGCCTCAAAGGC
TCCAAAGCGGTCAATCCGTTAGAC
TCCACCAGATTCTTCGCCTG
TCCCAACTGGCAAAGTTAAAGGAATC
TCCGTAGCTTGTGTATCAATGACATTC
TCCCGGTACCAAATATTTTCGCTTAAC
GATCGAATTCTTTAAGAAGGAGATATACATATGCCGAATGCTAGTCGTGTT
GATCGGATCCCTATGCATTTTTTTCAGGCAATTCTGC
TCCTTGTTTGTCTTGATTGAGATGC
TCCTTCATTAGAGGATAAAAGAGCTG
GATCGTACCATATGTTTCTTGTTAGCATCTCAATCAAGAC
GATCGGATCCTACTCAGCTCTTTTATCCTCTAATG
GATCGTACCATATGCGCATCCCTAGCCTATC
GATCGGATCCTTAAGACAACCAGTCTAACGGATTG
GATCGTACCATATGGCAAATTTATTTAGTTTTTTGTTTCTTGGAAC
GATCGGATCCCTACTGATTTAGTTCATATCTACGAGC
TAGCGTGCCGCTTGTTAC
TCCAGCCCAGTGTTCCAAG
TCCGTGCTCGTAGATATGAAC
59 of MCS
39 of MCS
59 KmR (out)
39 KmR (out)
59 pG54-DlcsB
39 pG54-DlcsB
59 pG65-DlcsC
39 pG65-DlcsC
59 pG65-Dfis
39 pG65-Dfis
59-lcsC
39-lcsC
59 pG34-DlcsA
39 pG34-DlcsA
59-lcsA
39-lcsA
59-lcsB
39-lcsB
59-lcsD
39-lcsD
59-lcsC
59 pG66-DlcsD
39 pG66-DlcsD
One day before the experiment, the medium was exchanged, and the
amoebae were tapped off the tissue culture flask, spun down (5 min,
470 g) and resuspended in PYG (2?676106 ml21). Then 1?5 ml
samples of suspended amoebae (or PYG only) were applied on a
CYE/Cm agar plate, allowed to dry for 1–2 h in a laminar-flow
hood and left at room temperature overnight. Wild-type and icm/
dot mutant L. pneumophila strains used for the APT harboured the
plasmid pMMB207ab, pMMB207ab-Km14 or the corresponding
complementing plasmids listed in Table 1. For the APT, stationaryphase bacterial cultures (OD600 >4?5) were adjusted to an identical
OD600, and series of tenfold dilutions in sterile H2O were prepared;
3 ml of stationary culture (approx. 107 bacteria) or 3 ml of each dilution were spotted onto the CYE/Cm plates and incubated for
5–7 days at 30 uC or 37 uC.
Screening of an L. pneumophila chromosomal library for
icm/dot suppressors 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 harbours 5–10 kb
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 previously (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 or
icmG mutants grown in liquid culture were washed and resuspended
in M63 medium. Donor and recipient strains were incubated at a
ratio of 1/10 on CYE plates (4 h, 37 uC), resuspended in 0?2 ml M63
medium, streaked on CYE/Cm/Km plates and incubated for 4–
5 days at 37 uC. The icmS and icmG mutants harbouring the MW66
library were then suspended in AYE medium, spotted in serial
dilutions onto CYE/Cm plates containing 46106 A. castellanii and
170
incubated at 30 uC. JR32 and the icmS and icmG mutants harbouring
pMMB207ab were used for comparison. Within 7–14 days several
colonies of icm mutants harbouring 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 46106 A. castellanii. The MW66 library plasmids of
suppressor strains recovered from the second round of selection by
APT were analysed further. In preliminary experiments, the icmS
and icmG mutants harbouring the MW66 library were spread at an
m.o.i. of 1 simultaneously with 56106 A. castellanii ml21 in PYG
on CYE agar plates and incubated at 30 uC. Colonies that appeared
using this approach were subjected to a second round of selection as
described above.
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 analysed 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 59 and 39 ends of the insert. The sequences
obtained were mapped in the L. pneumophila genome (Chien et al.,
2004; http://genome3.cpmc.columbia.edu/~legion/) and analysed
for homologues of the ORFs identified.
Intracellular growth in A. castellanii and RAW264.7 macrophages. For intracellular growth assays, A. castellanii (56104 per
well) or RAW264.7 macrophages (26104 per well) were seeded onto
a 96-well plate and allowed to adhere for 3 h or overnight, respectively. The phagocytes were infected with L. pneumophila grown to
stationary phase in AYE medium for 24 h (m.o.i. 1, 880 g), and
incubated at 30 uC (A. castellanii) or 37 uC (RAW264.7). Intracellular growth was quantified by plating appropriately diluted
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151
Cytotoxic L. pneumophila lpxB paralogue
supernatant 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 A/J 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). However,
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.
Cytotoxicity assay. To determine cytotoxicity, 46104 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 plates 3 or 4 days
old were resuspended and diluted in sterile water to infect the amoebae at an m.o.i. of 50 or 500. After spinning down the bacteria
(880 g, 5 min), the plates were incubated at 30 uC. Two days postinfection, propidium iodide (PI) solution was added to the wells at
a final concentration of 1 mg ml21. After several minutes’ incubation, the amoebae were viewed in brightfield or by epifluorescence
with an inverse microscope (Zeiss Axiovert 200M, 206 objective).
The percentage of dead (PI-positive) amoebae was determined by
counting the number of total and fluorescent amoebae.
Computational and statistical analysis. Translated nucleic acid
databases or the conserved domain database (Marchler-Bauer et al.,
2003) were searched using the TBLASTX or RPSBLAST algorithms,
respectively (Altschul et al., 1997). The COG database is maintained
by 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.
RESULTS
Growth of L. pneumophila on CYE agar plates
in the presence of A. castellanii – the amoebae
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 analyse growth of wild-type and icm/dot mutant L.
pneumophila on CYE agar plates in the presence of A.
castellanii. Stationary-phase cultures of L. pneumophila
wild-type strain JR32 and icm/dot mutants harbouring
empty or complementing plasmids were spotted in tenfold
serial dilutions onto agar plates in the presence or, as a
control, absence of amoebae (Fig. 1). Within 5–7 days at
30 uC, wild-type L. pneumophila strain JR32 formed robust
colonies even at a 1025 dilution (m.o.i. approximately
0?02). Under these conditions, the icmT, -R, -O, -P, -C
and dotB mutant strains did not grow at all. The icmS and
-G mutant strains showed a partial growth defect in the
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 of the 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
Fig. 1. Growth of wild-type L. pneumophila
and representative icm/dot mutants in the
APT. L. pneumophila wild-type strain JR32
and icmT, -S, -R, -P, -G and dotB mutants
harbouring empty (pMMB) or corresponding
complementing plasmids (picmT, -S, -R, -P,
-G, dotB) were spotted in tenfold serial dilutions onto CYE agar plates in the presence
or absence of A. castellanii and incubated
for 5 days at 30 6C as described in
Methods. The experiments were done at
least three times and results similar to those
shown were obtained.
http://mic.sgmjournals.org
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
171
U. Albers and others
level (icmT, -S, -P, -O, -R, dotB) or partially (icmG, -C). In
the absence of amoebae, all strains grew equally well on
CYE agar plates.
At 37 uC, the icmS mutant grew similarly 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, -O, -G, -C
mutants grew only at the highest concentration. The degree
of complementation of individual icm mutants was the
same as at 30 uC (data not shown).
Isolation of L. pneumophila icm/dot suppressor
strains using the APT
Among the icm/dot mutant strains tested, only the icmS
and -G mutants showed a partial, rather than complete,
growth defect in the APT at 30 uC. A partial growth defect
for the icmS and -G mutant strains was shown previously
using HL-60 macrophages as host cells (Purcell & Shuman,
1998; Segal & Shuman, 1997, 1999b). We reasoned that
this phenotype would increase the 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 or -G mutants. The icmS and -G mutants harbouring the library MW66 were then spotted in serial dilutions onto CYE plates containing Cm and A. castellanii. The
plates were incubated at 30 uC or 37 uC, respectively.
At 37 uC, the selection was not promising, since at the same
dilutions similar numbers of icm mutants harbouring the
library and parental mutants were obtained. At 30 uC,
however, several colonies of icm mutant strains harbouring MW66 appeared within 7–14 days 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. 2(a). The suppressor strains (icmS2/pS37, icmG2/pG44–87) grow at
10–100-fold higher dilutions than the corresponding icm
mutant strains harbouring the empty library plasmid
pMMB207ab (icmS2/pMMB, icmG2/pMMB), but not as
vigorously as wild-type L. pneumophila (JR32/pMMB) or
complemented icm mutants (icmS2/picmS, icmG2/picmG).
In the 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 (1 icmS2, 17 icmG2
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 analysed by APT.
172
The strains thus obtained grew better in the APT than
the parental icm mutant harbouring an empty library
plasmid, indicating that suppression of the growth defect
in the presence of amoebae was caused by the library
plasmid and not due to a second-site mutation in the
bacterial chromosome. Representative strains belonging to
different EcoRI restriction groups are shown in Fig. 2(b).
Retransformation of an icmS mutant strain with plasmid
pS37 led to growth in the APT similar to that of wild-type
L. pneumophila, consistent with the finding that pS37
complements the icmS mutant strain (see below).
Analysis of suppressor plasmid inserts
The inserts from representative suppressor plasmids were
partially sequenced and mapped in the L. pneumophila
strain Philadelphia genome (http://genome3.cpmc.
columbia.edu/~legion/). Some of the inserts were identical
(pS36/pS37; pG54/pG58/pG63; pG65/pG72; pG71/pG79/
pG83), and the inserts of the suppressor plasmids pG47
and pG61 were independently selected twice, since they
covered the same loci in the L. pneumophila genome but
were of different length (1?8 kb and 2?5 kb, respectively).
The inserts of the suppressor plasmids were analysed by
BLASTX searches for homologues to ORFs, and 12 different
inserts were identified (pS37, pG34, pG39, pG41, pG47,
pG53, pG54, pG65, pG66, pG67, pG71 and pG78), each
harbouring 1–10 ORFs (Table 3).
The insert of the icmS suppressor plasmid pS37 contains a
9?0 kb fragment of icm/dot region II. 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 icmS2/pS37 strain in the APT is
due to intracellular growth within and killing of the
amoebae. In contrast, 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. The suppressor strains might
grow better on agar plates in the presence of amoebae
because of more efficient intracellular replication and
host cell killing or as a result of 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.
Intracellular growth of icmG suppressor strains
in A. castellanii and macrophages
Intracellular growth of the icmG suppressor strains was
assayed by quantifying bacteria released into the supernatant from A. castellanii infected with bacteria at an
m.o.i. of 1. To match the conditions used for the APT,
the infected amoebae were incubated for 4 days at 30 uC,
rather than at 37 uC as described previously (Segal &
Shuman, 1999b). At 30 uC, the number of wild-type
L. pneumophila or icmG mutant bacteria harbouring the
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151
Cytotoxic L. pneumophila lpxB paralogue
Legionella
(a)
Legionella
(b)
Wild-type/pMMB
Wild-type/pMMB
_
_
icmS /pMMB
icmS /pMMB
_
_
icmS /picmS
icmS /pS37
_
icmS /pS37
Wild-type/pMMB
_
icmG /pMMB
_
icmG /picmG
_
icmG /picmG
_
icmG /pMMB
_
_
icmG /pG44
icmG /pG34
_
icmG /pG55
_
icmG /pG54
_
icmG /pG67
_
icmG /pG65
_
icmG /pG45
_
icmG /pG50
_
icmG /pG51
_
Fig. 2. Suppressor strains isolated by APT. (a) L. pneumophila
icmS and icmG suppressor strains harbouring Legionella chromosomal library plasmids (pS37, pG44–pG87) were isolated
by APT, grown to stationary phase and retested by APT.
Representative suppressor strains are shown in comparison
with wild-type, icmG and icmS mutant strains harbouring the
empty library plasmid (pMMB) and corresponding complemented strains (picmG, picmS). The APT was repeated twice with
strains isolated in a first round of selection, and similar results
were obtained. (b) Growth defect suppression in an APT by L.
pneumophila icmS or icmG mutants transformed freshly with
isolated suppressor plasmids (pS37, pG34, pG54, pG65).
icmG /pG53
_
icmG /pG54
_
icmG /pG63
_
icmG /pG76
_
icmG /pG87
empty library plasmid increased within 3 days, by 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 (harbouring 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 m.o.i. of 500. Under these conditions, wildtype L. pneumophila grew 2 orders of magnitude within
http://mic.sgmjournals.org
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 uC, the suppressor strains icmG2/pG54 or icmG2/pG65 did also not
grow better in amoebae (m.o.i. of 1) than an icmG mutant
strain harbouring the empty library plasmid (data not
shown).
To analyse 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
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
173
U. Albers and others
Table 3. Suppressor plasmids identified in this study
Plasmid
Insert (kb)
ORFs* (bp)
Similarity (E-valueD)
pS37
9?0
10 ORFs
pG34
2?4
pG39
3?4
pG41
3?4
pG47
1?8
>257
455
821
>1018
257
290
>902
>414
365
731
761
>367
>352
864
>278
>1403
>446
311
1547
1169
Hypothetical protein-icmTSRQPONML,
Icm/Dot region II (0?0)
Hypothetical protein (4e210)
Hypothetical protein (8e210)
Hydrolase (5e261)
Hypothetical protein (5e211)
None
None
Lytic murein transglycosylase (3e221)
dCTP deaminase (2e264)
Integration host factor (6e227)
Histone methyltransferase (7e27)
None
None
None
LuxR family transcription factor (5e23)
None
Histidine ammonia lyase (1e2142)
Phospholipase C (6e283)
None
Phosphoglycerate mutase (1e2154)
NlpD-related membrane metalloprotease,
M23/M37 family (9e239)
Oxidoreductase
Lipid A disaccharide synthase (9e261)
Factor for inversion stimulation (3e214)
Carbamoyl phosphate synthase (1e228)
Putative esterase (3e214)
ABC transporter (1e213)
None
Heat-shock protein ClpB (0?0)
Signal peptide peptidase SppA (6e269)
Inositol monophosphatase (2e261)
Isovaleryl-CoA dehydrogenase (1e281)
Propionyl-CoA carboxylase (6e269)
3-Oxoacyl-(ACP) synthase III (9e218)
Acyl-CoA synthetase (2e240)
Hypothetical protein (1e261),
Hypothetical protein (3e230)
Kynurenine monooxygenase (2e272)
pG53
pG54
pG65
pG66
1?4
3?8
1?9
2?9
pG67
6?1
pG71
5?0
pG78
4?0
>281
1160
317
>1269
614
956
>1687
2588
1019
>596
>1634
1442
1070
>716
1418
785
1349
Accession no.
Toxic gened
T18328
NP_799916
NP_635420
ZP_00011307
NP_819452
lcsA (lpg0163)
ZP_00129242
NP_298052
NP_231548
NP_740808
NP_230922
NP_230847
AAM73854
ZP_00141602
NP_799213
NP_819651
NP_819650
NP_716031
NP_782304
ZP_00133753
NP_532754
NP_289147
NP_793611
NP_820132
NP_488098
NP_437987
NP_687378
ZP_00110863
NP_484368
NP_484370
NP_636923
lcsB (lpg0500)
lcsC (lpg1371)
lcsD (lpg0068)
*A minimum ORF size of 100 amino acids was chosen. Truncated ORFs are indicated by ‘>’.
DE-values are indicated for each translated ORF identified in the suppressor plasmid insert.
dCytotoxicity for A. castellanii of the plasmids and genes was determined with an icmG mutant strain harbouring either the suppressor plasmid,
deletion constructs derived therefrom, or an expression plasmid. The gene number corresponds to the annotated L. pneumophila genome.
supernatant 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 (icmG2/pG34–pG78) grew worse
than the icmG mutant strain, similar to what was observed
for growth of these strains in A. castellanii. Taken together,
the results indicate that growth defect suppression of the
174
icmG suppressor strains is not due to enhanced intracellular
growth within phagocytes.
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
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151
Cytotoxic L. pneumophila lpxB paralogue
the icmG mutant in some experiments, and icmG2/pG67
and /pG78 did not show increased cytotoxicity in any
of the experiments performed. At 37 uC, the suppressor
strains icmG2/pG54 or icmG2/pG65 were cytotoxic for
amoebae to the same extent as at 30 uC (data not shown).
Identification of cytotoxic suppressor genes by
deletion analysis
Fig. 3. Intracellular multiplication of icmG suppressor strains
within A. castellanii and macrophages. Intracellular growth of
the icmG suppressor strains was assayed using (a) A. castellanii or (b) murine RAW264.7 macrophages as host cells. The
phagocytes were infected at an m.o.i. of 1, and bacteria
released into the supernatant were quantified at the time indicated. Growth of suppressor strains icmG”/pG34–pG78 (#),
an icmG mutant harbouring the empty library plasmid (n), or
plasmid-encoded icmG (m) and wild-type strain JR32 (&) is
shown. The experiments were repeated at least twice at two
different m.o.i.’s each, and similar results were obtained.
or macrophages, we tested whether the suppressor plasmids
pG34–pG78 confer increased cytotoxicity to icmG mutants.
A. castellanii was infected with wild-type L. pneumophila,
an icmG mutant harbouring the empty library plasmid, a
complemented icmG mutant, or the icmG suppressor
strains (harbouring pG34–G78). Cytotoxicity was determined by PI uptake 2 days post-infection. Under the
conditions described, the amoebae infected with either
wild-type L. 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 harbouring
the vector control all survived and remained spread out
and firmly attached. Amoebae infected with an icmG
mutant harbouring 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 harbouring pG34, pG39, pG41, pG54 or
pG66 showed an increased cytotoxicity in 4–5 out of 5
independent experiments. The suppressor strains icmG2/
pG47, /pG53, /pG65 and /pG71 were more cytotoxic than
http://mic.sgmjournals.org
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
therefore this plasmid was not analysed further. Deletion
of all the complete ORFs in the insert of pG39 also did
not decrease cytotoxicity, but left the fragment of an
ORF encoding the C-terminus of a putative lytic murein
transglycosylase (data not shown; see Discussion). These
findings and considerations left the cytotoxic suppressor
plasmids pG34, pG54, pG65 and pG66 to be analysed in
detail.
The inserts of the plasmids pG34, pG54, pG65 and pG66
harbour homologues 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 homologue) and -D (ABC transporter). Deletion of ORF1-3 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-ORF1-3 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).
Overexpression of suppressor genes
Prompted by the finding that strain icmG2/pG65-fis
showed enhanced cytotoxicity compared to icmG2/pG65,
we cloned lcsA, -B, -C, and -D into the expression vector
pMMB207-RBS, overexpressed the proteins under the
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
175
U. Albers and others
(a)
Wild-type/pMMB
_
icmT /pMMB
_
icmG /pMMB
_
icmG /pG54
_
icmG /pG65
Brightfield
PI
Merge
(b)
80
60
40
20
ic
ic
m
G
_
/p
M
M
B
m
G _
/p
G
34
ic
m
G _
/p
G
39
ic
m
G _
/p
G
41
ic
m
G _
/p
G
47
ic
m
G _
/p
G
53
ic
m
G _
/p
G
54
ic
m
G _
/p
G
65
ic
m
G _
/p
G
66
ic
m
G _
/p
G
67
ic
m
G _
/p
G
71
ic
m
G _
/p
G
78
PI-positive A. castellanii (%)
100
control of the Ptac promoter, and quantified cytotoxicity
of icmG mutant strains harbouring these plasmids by PI
uptake. Interestingly, the strain icmG2/pLcsC, overexpressing
the LpxB homologue, was 9 or 4 times more cytotoxic than
icmG2/pMMB or icmG2/pG65, respectively (Fig. 6a, b),
suggesting that LcsC-induced cytotoxicity might be dosedependent. The icmG2/pLcsC strain did not grow more
efficiently in A. castellanii (30 uC, 37 uC) or in RAW264.7
macrophages compared to icmG2/pMMB (data not shown).
In contrast, icmG mutants expressing the putative hydrolase LcsA, NlpD-like metalloprotease LcsB or the ABC
transporter LcsD were less cytotoxic than the icmG
mutant strain. We noted, however, that induction of LcsB
prevented growth of the bacteria, and induction of LcsC
176
Fig. 4. Suppressor strains are cytotoxic for
A. castellanii. (a) A. castellanii was infected
with wild-type L. pneumophila, icmT or
icmG mutant strains harbouring the empty
library plasmid (pMMB) or with suppressor
strains (icmG”/pG34–pG78). Cytotoxicity
was determined by PI uptake 2 days postinfection as described in Methods. (b) The
data shown are the means and standard
deviations of the percentage of PI-positive
A. castellanii counted in three different wells
on a 24-well plate and are representative of
at least five independent experiments.
and LcsD resulted in fewer and smaller bacterial colonies,
indicating that overexpression of these proteins is toxic for
L. pneumophila (data not shown). In agreement with this
assumption, the strains icmG2/pLcsA–D grew well on agar
plates, if IPTG was omitted, and therefore the genes were
expressed only at low levels from the Ptac promoter. Under
these conditions, strain icmG2/pLcsC was consistently
more cytotoxic than the parental suppressor strain icmG2/
pG65 in three independent experiments (data not shown).
Strain icmG2/pLcsB was more cytotoxic than the negative
control icmG2/pMMB207 but not as cytotoxic as the
parental suppressor strain icmG2/pG54. Finally, the strains
icmG2/pLcsA and icmG2/pLcsD were not cytotoxic for the
amoebae (data not shown).
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151
Cytotoxic L. pneumophila lpxB paralogue
(a)
(a)
_
icmG /pMMB
_
icmG /pLcsC
Brightfield
PI
Merge
http://mic.sgmjournals.org
Lc
G
m
ic
ic
ic
sD
66
_
/p
_
/p
G
m
G
m
m
ic
G
sC
65
Lc
_
/p
_
/p
G
_
/p
G
m
ic
ic
G
Lc
G
/p
_
G
m
G
m
ic
sB
54
sA
Lc
34
G
_
_
/p
/p
ic
m
m
T
G
_
ic
ic
ic
m
m
G
G
_
In the 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 harbouring the empty
plasmid pMMB207 were cytotoxic for the amoebae to a
similar extent as the icmG mutant. Like the icmS mutant
strain, the icmC, -E, -O, -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 L. pneumophila (m.o.i. of 5)
harbouring either an empty plasmid or pLcsC were killed
within 2 days, and thus LcsC apparently did not increase
cytotoxicity of wild-type L. pneumophila against the
amoebae (data not shown).
(c)
50
40
30
20
Lc
/p
_
/p
rp
oS
_
rp
oS
_
F
m
ic
sC
B
M
Lc
/p
_
F
m
ic
M
sC
B
/p
_
ic
m
S
_
S
M
Lc
/p
M
/p
/p
_
ic
m
G
_
M
B
M
sC
Lc
M
M
/p
G
m
ic
sC
10
B
PI-positive A. castellanii (%)
Fig. 5. Identification of cytotoxic suppressor genes by deletion
analysis. (a) Overview of the deletions (horizontal bars) generated in suppressor plasmid pG34 (lcsA), pG54 (lcsB, ORF13), pG65 (lcsC, fis) and pG66 (lcsD). (b) A. castellanii was
infected with icmT or icmG mutant strains harbouring the
empty library plasmid (pMMB), complemented icmG (icmG”/
picmG), suppressor strains (icmG”/pG34, /pG54, /pG65,
/pG66), or with the deletion strains indicated. Cytotoxicity was
determined by PI uptake 2 days post-infection. The data shown
are the means and standard deviations of the percentage of PIpositive A. castellanii counted in three different wells on a 24well plate and are representative of at least three independent
experiments.
M
B
/p
/p
ic
M
m
M
G
20
B
20
M
40
40
m
60
60
_
80
80
ic
100
ic
m
G _
/p
ic
ic
m
m
G _ G
/p
M
ic
M
m
T _ B
/p
M
ic
M
m
ic
G _ B
m
G _ /pG
/p
34
G
34
-lc
ic
s
m
ic
G _ A
m
G _ /pG
ic
/p
m
54
G _ G
54
/p
G
54 lcsB
-O
RF
ic
1
m
G _ -3
ic
m
/
G _ pG
/p
65
G
65
ic
m
G _ lcs
C
/p
G
65
ic
-fi
m
ic
G _ s
m
G _ /p
G
/p
66
G
66
-lc
sD
PI-positive A. castellanii (%)
(b)
100
/p
PI-positive A. castellanii (%)
(b)
Fig. 6. Overexpression of LcsC is cytotoxic for A. castellanii in
an icmG but not in other icm or rpoS mutants. A. castellanii
was infected with the icm (a–c) or rpoS (c) mutant strains indicated harbouring the empty library plasmid (pMMB), complementing plasmid (picmG), suppressor plasmids (pG34, pG54,
pG65, pG66), or expression plasmids (pLcsA–D). Cytotoxicity
was determined by PI uptake 2 days post-infection. The data
shown are the means and standard deviations of the percentage of PI-positive A. castellanii counted in three different wells
on a 24-well plate and are representative of at least three independent experiments.
DISCUSSION
Screen for icm suppressors using the APT
In this paper we describe a novel assay, the amoebae plate
test (APT), to determine growth of bacteria on CYE agar
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
177
U. Albers and others
plates in the presence of amoebae. Wild-type L. pneumophila replicates within and kills amoebae, and thus grows
on solid medium. In contrast, icm/dot mutant strains are
defective for intracellular growth, are killed by the amoebae
and do not form colonies on agar plates (Fig. 1). The APT
was routinely done at 30 uC, allowing the amoebae 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 uC, the icmS and other icm/dot
mutants grew better than at 30 uC in the presence of
amoebae. Possible explanations for this finding include (i)
the bacteria grow faster on agar plates at 37 uC 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 (Figs 3
and 4). Some cytotoxic icmG suppressor genes were
identified by deletion analysis (lcsA–D, Legionella cytotoxic suppressor); these encode a putative hydrolase, an
NlpD-like metalloprotease, a lipid A disaccharide synthase
homologue 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 databases. The lcsB and
lcsC genes were found to be of particular interest, since
both genes may be 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 ORF1-3 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 promoter and the first
internal ATG as a start codon.
LcsB, a homologue of NlpD-like membranebound metalloproteases
LcsB is 26 % identical to the E. coli YibP protease and 17–
18 % identical to the NlpD orthologues from L. pneumophila and E. coli (Fig. 7). The C-terminal domains of the
four proteins belong to COG4942 (membrane-bound
metallopeptidases, involved in cell division and chromosome partitioning) and are characteristic for members of
the M23/M37 family (Pfam01551) of putative zinc metalloendopeptidases from Gram-negative and Gram-positive
bacteria (http://www.sanger.ac.uk/Software/Pfam/). The
M37 protease family includes staphylococcal glycylglycine
endopeptidases that hydrolyse 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
Fig. 7. Alignment of L. pneumophila LcsB, E. coli YibP and the membrane-bound metalloproteases NlpD from L.
pneumophila and E. coli.
178
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151
Cytotoxic L. pneumophila lpxB paralogue
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 L. 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 databases, L. 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 uC but not at 37 uC. At the non-permissive
temperature, the yibP mutant forms filamentous, multinucleoided cells that tend to lyse, indicating that YibP is
required for cell wall degradation and proper cell division.
LcsC, a paralogue of the lipid A disaccharide
synthase LpxB
LcsC is homologous to lipid A disaccharide synthases
(LpxB) and, as determined by searching the conserved
domain database, distantly to MurG glycosyltransferases
involved in peptidoglycan synthesis (COG0707). Interestingly, the genome of L. pneumophila Philadelphia-1
harbours two lipid A disaccharide synthase paralogues
which are 42 % or 30 % identical to E. coli LpxB, respectively (Fig. 8). The gene more closely related to E. coli
lpxB is most likely the L. pneumophila lpxB orthologue
since it is located within a cluster encoding components
of the lipopolysaccharide (LPS) biosynthesis pathway,
including a neuraminidase, lauroyl/myristoyl acyltransferase, LpxA, LpxD, LpxB and the ABC transporter WlaB.
The arrangement of the lpx gene cluster is similar in E. coli,
where lpxB is located downstream of lpxA, fabZ and lpxD
(Raetz, 1996).
In the L. pneumophila genome, the lcsC gene appears to
form an operon with an oxidoreductase/dehydrogenase
gene and is separated from the putative rodA–ftsI operon
by only one ORF. The latter genes encode the rodshape-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 the
possibility that LcsC participates in lipid A biosynthesis.
However, the vicinity of lcsC to rodA–ftsI, its lower homology to lpxB and its similarity to murG suggest that LcsC
catalyses a disaccharide synthase/glycosyltransferase reaction in peptidoglycan biosynthesis.
The remote structural similarity of LcsC/LpxB and MurG
corresponds to the fact that both glycosyltransferases
catalyse the synthesis of a b-linked lipodisaccharide
moiety from an acylated uridine 59-diphosphate (UDP)D-glucosamine (GlcN) and a lipid, and both enzymes
liberate UDP during the reaction. LpxB catalyses the b-19-6
condensation of UDP-2,3-diacyl-GlcN with lipid X
(2,2-diacyl-GlcN-1-phosphate), and MurG couples Nacetyl-GlcN via a b-19-4 linkage to lipid I (N-acetylmuramyl-pentapeptide-phosphoryl-undecaprenol) to form
the lipodisaccharide lipid II that is the minimal subunit
of peptidoglycan (Heijenoort, 1996).
Mechanism of LcsC cytotoxicity
Not only was cytotoxicity of an icmG mutant strain
harbouring the suppressor plasmid pG65 decreased upon
deletion of lcsC (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 supernatant. Soluble cytotoxic peptidoglycan derivatives have been described for
Bordetella pertussis, Neisseria gonorrhoeae, Haemophilus
Fig. 8. Alignment of L. pneumophila LcsC with lipid A disaccharide synthase (LpxB) homologues from L. pneumophila and
E. coli.
http://mic.sgmjournals.org
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
179
U. Albers and others
influenzae and other bacteria (Burroughs et al., 1993; Cloud
& Dillard, 2002; Cookson et al., 1989; Luker et al., 1993,
1995). For example, the murein-derived tracheal cytotoxin
(TCT; N-acetyl-GlcN-1,6-anhydro-N-acetyl-muramyl-LAla-c-D-Glu-meso-diaminopimelyl-D-Ala) from B. pertussis
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.
influenzae are also disaccharide muramyl tetrapeptides, i.e.
they consist of the monomeric subunit of Gram-negative
bacterial peptidoglycan.
generation of protein database search programs. Nucleic Acids Res 25,
3389–3402.
Overexpression of LcsC apparently is cytotoxic for the
amoebae specifically in an icmG mutant but not in other
icm/dot mutants or an rpoS mutant background (Fig. 6c).
One possibility to account for this observation would be
that LcsC requires a functional Icm/Dot 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 Icm/Dot secretion
systems, 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 uC (Fig. 4), the icmG and icmF
mutants persist in D. discoideum (Otto et al., 2004), and the
icmS and icmW mutants are not impaired in Icm/Dotdependent 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 Icm/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 that in other icm/dot mutants as well as from
that in wild-type L. pneumophila. It is feasible that overexpression of LcsC exerts cytotoxic effects only in certain
cellular compartments. We are currently investigating entry
and intracellular trafficking of the icmG mutant and effects
of LcsC on these processes to establish a link between loss of
IcmG (possibly involved in intracellular trafficking) and
cytotoxicity of LcsC (a putative enzyme of peptidoglycan
metabolism).
Chien, M., Morozova, I., Shi, S. & 34 other authors (2004). The
Begg, K. J., Spratt, B. G. & Donachie, W. D. (1986). Inter-
action between membrane proteins PBP3 and RodA is required for
normal cell shape and division in Escherichia coli. J Bacteriol 167,
1004–1008.
Burroughs, M., Rozdzinski, E., Geelen, S. & Tuomanen, E. (1993). A
structure-activity relationship for induction of meningeal inflammation by muramyl peptides. J Clin Invest 92, 297–302.
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.
genomic sequence of the accidental pathogen Legionella pneumophila. Science 305, 1966–1968.
Cirillo, S. L., Bermudez, L. E., El-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 of
Neisseria gonorrhoeae is involved in peptidoglycan-derived cytotoxin
production. Infect Immun 70, 2752–2757.
Coers, J., Monahan, C. & Roy, C. R. (1999). Modulation of
phagosome biogenesis by Legionella pneumophila creates an
organelle permissive for intracellular growth. Nat Cell Biol 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 an organelle permissive
for Legionella pneumophila intracellular growth. Mol Microbiol 38,
719–736.
Conover, G. M., Derre, I., Vogel, J. P. & Isberg, R. R. (2003). The
Legionella pneumophila LidA protein: a translocated substrate of the
Dot/Icm system associated with maintenance of bacterial integrity.
Mol Microbiol 48, 305–321.
Cookson, B. T., Tyler, A. N. & Goldman, W. E. (1989). Primary
structure of the peptidoglycan-derived tracheal cytotoxin of
Bordetella pertussis. Biochemistry 28, 1744–1749.
Duménil, G. & Isberg, R. R. (2001). The Legionella pneumophila
IcmR protein exhibits chaperone activity for IcmQ by preventing its
participation in high-molecular-weight complexes. Mol Microbiol 40,
1113–1127.
Duménil, G., Montminy, T. P., Tang, M. & Isberg, R. R. (2004).
IcmR-regulated membrane insertion and efflux by the Legionella
pneumophila IcmQ protein. J Biol 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 for Legionella pneumophila. J Clin
Microbiol 10, 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 the genus Legionella. Cell
Microbiol 2, 165–171.
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 (17/02-3).
REFERENCES
Hales, L. M. & Shuman, H. A. (1999). The Legionella pneumophila
rpoS gene is required for growth within Acanthamoeba castellanii.
J Bacteriol 181, 4879–4889.
Heijenoort, J. v. (1996). Murein synthesis. In Escherichia coli and
Salmonella typhimurium, 2nd edn, pp. 1025–1034. Edited by F. C.
Neidhardt and others. Washington, DC: American Society for
Microbiology.
Hilbi, H., Segal, G. & Shuman, H. A. (2001). Icm/dot-dependent
Altschul, S. F., Madden, T. L., Schaffer, A. A., Zhang, J., Zhang, Z.,
Miller, W. & Lipman, D. J. (1997). Gapped BLAST and PSI-BLAST: a new
180
upregulation of phagocytosis by Legionella pneumophila. Mol
Microbiol 42, 603–617.
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151
Cytotoxic L. pneumophila lpxB paralogue
Horwitz, M. A. (1983a). The Legionnaires’ disease bacterium
(Legionella pneumophila) inhibits lysosome-phagosome fusion in
human monocytes. J Exp Med 158, 2108–2126.
Horwitz, M. A. (1983b). Formation of a novel phagosome by the
Legionnaires’ disease bacterium (Legionella pneumophila) in human
monocytes. J Exp Med 158, 1319–1331.
Horwitz, M. & Maxfield, F. (1984). Legionella pneumophila inhibits
acidification of its phagosome in human monocytes. J Cell Biol 99,
105–114.
Horwitz, M. A. & Silverstein, S. C. (1983). Intracellular multiplication
of Legionnaires’ disease bacteria (Legionella pneumophila) in human
monocytes is reversibly inhibited by 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 Escherichia coli chromosome encodes a lipoprotein with
unusual amino acid repeat sequences. J Bacteriol 176, 1630–1638.
Morozova, I., Qu, X., Shi, S., Asamani, G., Greenberg, J. E.,
Shuman, H. A. & Russo, J. J. (2004). Comparative sequence
Ichimura, T., Yamazoe, M., Maeda, M., Wada, C. & Hiraga, S.
(2002). Proteolytic activity of YibP protein in Escherichia coli.
Purcell, M. & Shuman, H. A. (1998). The Legionella pneumophila
analysis of the icm/dot genes in Legionella. Plasmid 51, 127–147.
Nagai, H. & Roy, C. R. (2001). The DotA protein from Legionella
pneumophila is secreted by a novel process that requires the Dot/Icm
transporter. EMBO J 20, 5962–5970.
Nagai, H. & Roy, C. R. (2003). Show me the substrates: modulation
of host cell function by type IV secretion systems. Cell Microbiol 5,
373–383.
Nagai, H., Kagan, J. C., Zhu, X., Kahn, R. A. & Roy, C. R. (2002).
A bacterial guanine nucleotide exchange factor activates ARF on
Legionella phagosomes. Science 295, 679–682.
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 of Legionella pneumophila
in Dictyostelium discoideum. Mol Microbiol 51, 63–72.
J Bacteriol 184, 2595–2602.
icmGCDJBF genes are required for killing of human macrophages.
Infect Immun 66, 2245–2255.
Imai, Y., Matsushima, Y., Sugimura, T. & Terada, M. (1991). A
Quinn, F. D. & Tompkins, L. S. (1989). Analysis of a cloned sequence
simple and rapid method for generating a deletion by PCR. Nucleic
Acids Res 19, 2785.
Kagan, J. C. & Roy, C. R. (2002). Legionella phagosomes intercept
vesicular traffic from endoplasmic reticulum exit sites. Nat Cell Biol
4, 945–954.
Kagan, J. C., Stein, M. P., Pypaert, M. & Roy, C. R. (2004). Legionella
subvert the functions of rab1 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-forming ability by Legionella pneumophila. Mol
Microbiol 27, 323–336.
Lange, R. & Hengge-Aronis, R. (1994). The nlpD gene is located
in an operon with rpoS on the Escherichia coli chromosome and
encodes a novel lipoprotein with a potential function in cell wall
formation. Mol Microbiol 13, 733–743.
Luker, K. E., Collier, J. L., Kolodziej, E. W., Marshall, G. R. &
Goldman, W. E. (1993). Bordetella pertussis tracheal cytotoxin and
of Legionella pneumophila encoding a 38 kD metalloprotease possessing haemolytic and cytotoxic activities. Mol Microbiol 3, 797–805.
Raetz, C. R. H. (1996). Bacterial lipopolysaccharides: a remarkable
family of bioactive macroamphiphiles. In Escherichia coli and
Salmonella typhimurium, 2nd edn, pp., 1035–1063. Edited by F. C.
Neidhardt and others. Washington: American Society for Microbiology.
Ramadurai, L. & Jayaswal, R. K. (1997). Molecular cloning,
sequencing, and expression of lytM, a unique autolytic gene of
Staphylococcus aureus. J Bacteriol 179, 3625–3631.
Ramadurai, L., Lockwood, K. J., Nadakavukaren, M. J. & Jayaswal,
R. K. (1999). Characterization of a chromosomally encoded glycyl-
glycine endopeptidase of Staphylococcus aureus. Microbiology 145,
801–808.
Recsei, P. A., Gruss, A. D. & Novick, R. P. (1987). Cloning, sequence,
and expression of the lysostaphin gene from Staphylococcus simulans.
Proc Natl Acad Sci U S A 84, 1127–1131.
other muramyl peptides: distinct structure-activity relationships for
respiratory epithelial cytopathology. Proc Natl Acad Sci U S A 90,
2365–2369.
Rowbotham, T. J. (1980). Preliminary report on the pathogenicity of
Legionella pneumophila for freshwater and soil amoebae. J Clin Pathol
33, 1179–1183.
Luker, K. E., Tyler, A. N., Marshall, G. R. & Goldman, W. E. (1995).
Roy, C. R., Berger, K. H. & Isberg, R. R. (1998). Legionella
Tracheal cytotoxin structural requirements for respiratory epithelial
damage in pertussis. Mol Microbiol 16, 733–743.
Luo, Z. Q. & Isberg, R. R. (2004). Multiple substrates of the Legionella
pneumophila Dot/Icm system identified by interbacterial protein
transfer. Proc Natl Acad Sci U S A 101, 841–846.
Marchler-Bauer, A., Anderson, J. B., DeWeese-Scott, C. & 24 other
authors (2003). CDD: a curated Entrez database of conserved
domain alignments. Nucleic Acids Res 31, 383–387.
Mintz, C. S. & Shuman, H. A. (1987). Transposition of bacteriophage
Mu in the Legionnaires’ disease bacterium. Proc Natl Acad Sci U S A
84, 4645–4649.
Moffat, J. F. & Tompkins, L. S. (1992). A quantitative model of
intracellular growth of Legionella 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 of Legionella pneumophila from mammalian and protozoan
cells. Infect Immun 70, 69–78.
Morales, V. M., Backman, A. & Bagdasarian, M. (1991). A series of
wide-host-range low-copy-number vectors that allow direct screening for recombinants. Gene 97, 39–47.
http://mic.sgmjournals.org
pneumophila DotA protein is required for early phagosome
trafficking decisions that occur within minutes of bacterial uptake.
Mol Microbiol 28, 663–674.
Sadosky, A. B., Wiater, L. A. & Shuman, H. A. (1993). Identification
of Legionella pneumophila 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 macrophage killing by Legionella pneumophila. Infect
Immun 65, 5057–5066.
Segal, G. & Shuman, H. A. (1998). Intracellular multiplication and
human macrophage killing by Legionella pneumophila are inhibited
by conjugal components of IncQ plasmid RSF1010. Mol Microbiol
30, 197–208.
Segal, G. & Shuman, H. A. (1999a). Possible origin of the Legionella
pneumophila virulence genes and their relation to Coxiella burnetii.
Mol Microbiol 33, 669–670.
Segal, G. & Shuman, H. A. (1999b). Legionella pneumophila utilizes
the same genes to multiply within Acanthamoeba castellanii and
human macrophages. Infect Immun 67, 2117–2124.
Segal, G., Purcell, M. & Shuman, H. A. (1998). Host cell killing
and bacterial conjugation require overlapping sets of genes within
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
181
U. Albers and others
a 22-kb region of the Legionella pneumophila genome. Proc Natl Acad
Sci U S A 95, 1669–1674.
Watarai, M., Derre, I., Kirby, J., Growney, J. D., Dietrich, W. F. &
Isberg, R. R. (2001). Legionella pneumophila is internalized by a
Solomon, J. M., Rupper, A., Cardelli, J. A. & Isberg, R. R. (2000).
macropinocytotic uptake pathway controlled by the Dot/Icm system
and the mouse lgn1 locus. J Exp Med 194, 1081–1096.
Intracellular growth of Legionella pneumophila in Dictyostelium
discoideum, a system for genetic analysis of host-pathogen interactions. Infect Immun 68, 2939–2947.
Wiater, L. A., Sadosky, A. B. & Shuman, H. A. (1994). Mutagenesis
phila: an aquatic microbe goes astray. FEMS Microbiol Rev 26, 149–162.
of Legionella pneumophila using Tn903dlllacZ: identification of
a growth-phase-regulated pigmentation gene. Mol Microbiol 11,
641–653.
Sturgill-Koszycki, S. & Swanson, M. S. (2000). Legionella pneumo-
Wiater, L. A., Dunn, K., Maxfield, F. R. & Shuman, H. A. (1998).
Steinert, M., Hentschel, U. & Hacker, J. (2002). Legionella pneumo-
phila replication vacuoles mature 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 charac-
terization of glycylglycine endopeptidase produced by Staphylococcus
capitis EPK1. J Bacteriol 179, 1193–1202.
Swanson, M. S. & Isberg, R. R. (1995). Association of Legionella
pneumophila with the macrophage endoplasmic reticulum. Infect
Immun 63, 3609–3620.
Szeto, L. & Shuman, H. A. (1990). The Legionella pneumophila major
Early events in phagosome establishment are required for intracellular survival of Legionella pneumophila. Infect Immun 66,
4450–4460.
Wintermeyer, E., Rdest, U., Ludwig, B., Debes, A. & Hacker, J.
(1991). Characterization of legiolysin (lly), responsible for haemo-
lytic activity, colour production and fluorescence of Legionella
pneumophila. Mol Microbiol 5, 1135–1143.
Wright, E. K., Goodart, S. A., Growney, J. D. & 7 other authors
(2003). Naip5 affects host susceptibility to the intracellular pathogen
Legionella pneumophila. Curr Biol 13, 27–36.
secretory protein, a protease, is not required for intracellular growth
or cell killing. Infect Immun 58, 2585–2592.
Yamamoto, Y., Klein, T. W., Newton, C. A., Widen, R. & Friedman, H.
(1988). Growth of Legionella pneumophila in thioglycolate-elicited
Tatusov, R. L., Natale, D. A., Garkavtsev, I. V. & 7 other authors
(2001). The COG database: new developments in phylogenetic
Yanisch-Perron, C., Vieira, J. & Messing, J. (1985). Improved M13
classification of proteins from complete genomes. Nucleic Acids Res
29, 22–28.
phage cloning vectors and host strains: nucleotide sequences of the
M13mp18 and pUC19 vectors. Gene 33, 103–119.
Tilney, L. G., Harb, O. S., Connelly, P. S., Robinson, C. G. & Roy, C. R.
(2001). How the parasitic bacterium Legionella pneumophila modifies
Zink, S. D., Pedersen, L., Cianciotto, N. P. & Abu-Kwaik, Y. (2002).
its phagosome and transforms it into rough ER: implications for
conversion of plasma membrane to the ER membrane. J Cell Sci 114,
4637–4650.
Vogel, J. P., Andrews, H. L., Wong, S. K. & Isberg, R. R. (1998).
Conjugative transfer by the virulence system of Legionella pneumophila. Science 279, 873–876.
182
peritoneal macrophages from A/J mice. Infect Immun 56, 370–375.
The Dot/Icm type IV secretion system of Legionella pneumophila is
essential for the induction of apoptosis in human macrophages.
Infect Immun 70, 1657–1663.
Zuckman, D. M., Hung, J. B. & Roy, C. R. (1999). Pore-
forming activity is not sufficient for Legionella pneumophila
phagosome trafficking and intracellular growth. Mol Microbiol 32,
990–1001.
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Mon, 19 Jun 2017 01:44:43
Microbiology 151