1471-2164-12-5361471-2164 Research article <p>Extensive recombination events and horizontal gene transfer shaped the <it>Legionella pneumophila </it>genomes</p> Gomez-ValeroLauralgomez@pasteur.fr RusniokChristopherunsiok@pasteur.fr JarraudSophiesophie.jarraud@univ-lyon1.fr VacherieBenoitbvacheri@genoscope.cns.fr RouyZoézrouy@genoscope.cns.fr BarbeValerievbarbe@genoscope.cns.fr MedigueClaudinecmedigue@genoscope.cns.fr EtienneJeromejetienne@univ-lyon1.fr BuchrieserCarmencarmen.buchrieser@pasteur.fr

Institut Pasteur, Biologie des Bactéries Intracellulaires, 75724, Paris, France

CNRS URA 2171, 75724, Paris, France

Université de Lyon, Lyon, France, Centre National de Référence des Legionella, Lyon, France

INSERM, U851, 69007 Lyon, France

Hospices Civils de Lyon, Lyon, France

CEA/DSV/FAR/IG/Genoscope Laboratoire de Génomique Comparative, Evry Cedex, France

CNRS UMR8030 Laboratoire d'Analyses Bioinformatiques en Métabolisme et Génomique, Evry, France

BMC Genomics 1471-2164 2011 12 1 536 http://www.biomedcentral.com/1471-2164/12/536 10.1186/1471-2164-12-53622044686
297201111120111112011 2011Gomez-Valero et al; licensee BioMed Central Ltd.This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Background

Legionella pneumophila is an intracellular pathogen of environmental protozoa. When humans inhale contaminated aerosols this bacterium may cause a severe pneumonia called Legionnaires' disease. Despite the abundance of dozens of Legionella species in aquatic reservoirs, the vast majority of human disease is caused by a single serogroup (Sg) of a single species, namely L. pneumophila Sg1. To get further insights into genome dynamics and evolution of Sg1 strains, we sequenced strains Lorraine and HL 0604 1035 (Sg1) and compared them to the available sequences of Sg1 strains Paris, Lens, Corby and Philadelphia, resulting in a comprehensive multigenome analysis.

Results

We show that L. pneumophila Sg1 has a highly conserved and syntenic core genome that comprises the many eukaryotic like proteins and a conserved repertoire of over 200 Dot/Icm type IV secreted substrates. However, recombination events and horizontal gene transfer are frequent. In particular the analyses of the distribution of nucleotide polymorphisms suggests that large chromosomal fragments of over 200 kbs are exchanged between L. pneumophila strains and contribute to the genome dynamics in the natural population. The many secretion systems present might be implicated in exchange of these fragments by conjugal transfer. Plasmids also play a role in genome diversification and are exchanged among strains and circulate between different Legionella species.

Conclusion

Horizontal gene transfer among bacteria and from eukaryotes to L. pneumophila as well as recombination between strains allows different clones to evolve into predominant disease clones and others to replace them subsequently within relatively short periods of time.

Background

Legionella pneumophila is the etiologic agent of Legionnaires' disease, an atypical pneumonia, which is often fatal if not treated promptly. However, it is principally an environmental bacterium that inhabits fresh water reservoirs worldwide where it parasitizes within free-living protozoa but also survives in biofilms 1 2 3 . Since L. pneumophila does not spread from person-to-person, humans have been inconsequential for the evolution of this pathogen. Instead, the virulence strategies of L. pneumophila have been shaped by selective pressures in aquatic ecosystems. Indeed, the co-evolution of L. pneumophila with fresh-water amoebae is reflected in its genome sequence. The analysis of two L. pneumophila genomes identified the presence of an unexpected high number and variety of eukaryotic-like proteins and proteins containing motifs mainly found in eukaryotes 4 . These proteins were predicted to interfere in different steps of the infectious cycle by mimicking functions of eukaryotic proteins 4 . For several of these eukaryotic like proteins it has been shown recently that they are secreted effectors that help L. pneumophila to subvert host functions to allow intracellular replication 5 6 . The possibility that L. pneumophila has acquired at least some of these genes through horizontal gene transfer from eukaryotes has been suggested by two studies 7 8 .

Plasticity is another specific feature of the L. pneumophila genomes as integrative plasmids, putative conjugation elements and genomic islands were identified. In addition to DNA interchange between different bacterial genera and even domains of life, horizontal gene transfer within the genus Legionella and within the species L. pneumophila has been reported. For example a 65-kb pathogenicity island described first in L. pneumophila strain Philadelphia 9 is present in several L. pneumophila strains and also in other Legionella species like L. anisa 10 . Another example is the particular lipopolysaccharide cluster of serogroup 1 strains that has been detected in L. pneumophila strains of different lineages and genetic backgrounds 10 . L. pneumophila has all necessary features for incorporating foreign DNA, as these bacteria are naturally competent and possess an intact recombination machinery 11 12 . These findings suggest that the L. pneumophila genomes are very dynamic and one would expect that horizontal gene transfer and recombination events play an important role in their evolution.

However, different analyses like early studies applying multilocus enzyme electrophoreses (MEE) supported a clonal population structure of L. pneumophila 13 . Two recent reports using genetic profiling based on six or three genetic loci, respectively concluded also that L. pneumophila shows a clonal populations structure 14 15 although the presence of few recombination events was not ruled out. Later the analysis of the dotA, mip and rpoB genes in different isolates suggested for the first time that recombination may play some role in L. pneumophila evolution 16 17 18 and a more in depth analysis using over 20 loci suggested that recombination events might be more frequent than was previously thought 19 . However, comparisons of these studies are difficult due to different sampling and different analysis methods used. Furthermore there may be a bias associated with some of the genes selected in these studies like intergenic spacer regions or genes under positive selection that may lead to artefactual effects in detecting recombination. To solve these problem efforts have been undertaken recently to homogenize the results obtained for different species to allow comparisons 20 . These authors report for L. pneumophila a low recombination rate like for the obligate pathogens Bordetella pertussis or Bartonella henselae. In contrast Coscolla and colleagues suggest a more important role for recombination at the intergenic level 21 .

These different results and the fact that a globally distributed L. pneumophila clone implicated in Legionnaires' disease has been described 10 may suggest that the role of recombination is not relevant. However, the description of clonal complexes is not incompatible with high recombination rates. Transient clones may appear within a recombining population 22 , in particular if clones with high disease prevalence appear, as this seems to be the case for some L. pneumophila strains. These clones are often vastly over-sampled due to their clinical importance and show strong clonality. Thus, this may be correct for this subgroup, but it may not be representative for the population. Indeed when analyzing over 200 clinical and environmental L. pneumophila strains, significantly less diversity was found among the clinical isolates 23 .

In this study we investigated the genome dynamics and evolution of the species L. pneumophila by analyzing horizontal gene transfer, mobile genetic elements and recombination on a genome-wide level. We undertook this analysis based on six complete genome sequences four of which are the previously published reference genomes of L. pneumophila Paris, Lens 4 , Corby 24 and Philadelphia 25 and two that were sequenced in this study. The newly sequenced strains were selected according to epidemiological features that might be reflected in their genomes and should thus allow to study genome dynamics with respect to virulence. Strain Lorraine is rarely isolated from the environment but its prevalence in human disease is increasing considerably in the last years 26 . In contrast, L. pneumophila strain HL 0604 1035 has been frequently isolated from a hospital water system since over 10 years but has never caused disease. Analysis of these six strains identified a highly conserved and syntenic core genome and a diverse accessory genome. Furthermore, it showed that recombination events and horizontal gene transfer are frequent in L. pneumophila. Horizontal gene transfer from eukaryotes as well as recombination between strains were identified suggesting that L. pneumophila genomes are highly dynamic, a feature allowing different clones to evolve into predominant disease clones and others to replace them subsequently within relatively short periods of time.

Results and discussion

The L. pneumophila core genome comprises over 2400 conserved genes that are highly syntenic

To get comprehensive insight into the genetic basis, evolution and genome dynamics of L. pneumophila Sg1, the strains responsible for over 90% of disease worldwide, we analyzed six completely sequenced genomes. The strains selected are all of Sg1, have endemic and/or epidemic character (e.g. Paris, Lorraine or Philadelphia) were isolated in different countries (France, England, Spain, US) and in different years. Two strains were newly sequenced for this study (Lorraine and HL 0604 1035), the other four L. pneumophila genomes (Paris, Lens, Philadelphia, Corby) have been published previously 4 24 25 . The genomes of L. pneumophila Lorraine and HL 0604 1035 consist each of a single circular chromosome of 3.4 Mb. Strain Lorraine also contains a plasmid. As shown in Table 1, the main features of the six L. pneumophila genomes analyzed (e.g. genome size, GC content and coding density), are highly conserved. The core genome of the six L. pneumophila genomes comprises 2434 genes, which represents about 80% of the predicted genes in each genome. Furthermore, the gene order is highly conserved as the 260 kb inversion in strain Lens with respect to the other strains is the only exception. When comparing the strains two by two, in average 90% of the genes are present in both strains (Figure 1). However, when determining the non-orthologous genes specific of each genome and not present in the remaining 5 strains, each strain contains between 136 (strain HL 0604 1035) and 222 (strain Corby) strain specific genes mainly encoded on mobile genetic elements. Taken together, the L. pneumophila genomes have a highly conserved and syntenic backbone and a highly dynamic accessory genome of about 300 genes each mainly formed by mobile genetic elements, genomic islands and genes of unknown function. The complete annotation of these six genomes is available in a new data base resource that we have set up, LegionellaScope https://www.genoscope.cns.fr/agc/microscope/about/collabprojects.php?P_id = 27 and at the Institut Pasteur, LegioList http://genolist.pasteur.fr/LegioList/.

<p>Table 1</p>

General features of the 6 L. pneumophila strains analyzed

L. pneumophila strains

Philadelphia

Paris

Lens

Corby

HL06041035

Lorraine


Chromosome size (bp)

3397754

3503610

3345687

3576469

3492535

3467254

G+C content (%)

38.27

38.37

38.42

38.48

38.35

38.36

N° of genes

3031

3123

2980

3237

3132

3117

N° of protein coding genes

2999

3078

2921

3193

3079

3080

Pseudogenes

55

71

84

59

73

48

tRNA

43

43

43

44

43

44

16S/23S/5S

3/3/3

3/3/3

3/3/3

3/3/3

3/3/3

3/3/3

Average length CDS (nts)

1082.47

1000.85

1008.76

984.35

995.47

988.54

Average length ig (nts)

147.72

154

152.36

149.24

155.12

155.28

Coding density (%)

88.22

86.93

87.07

87.25

86.94

87.26

Plasmids

0

1

1

0

0

1

bp, base pairs; nts, nucleotides; CDS, coding sequence; ig, intergenic region

<p>Figure 1</p>

Shared and specific gene content of 6 L. pneumophila genomes

Shared and specific gene content of 6 L. pneumophila genomes. Each petal represents a genome with an associated color. The number in the center of the diagram represents the orthologous genes shared by all the genomes. The number inside of each individual petal corresponds to the specific genes of each genome with non-orthologous genes in any of the other genomes. The small circles inside of each petal represent the percentage of shared genes (total number divided by the number of genes in the smallest genome) between the genome of this petal and the genome represented by the color of the small circle. Yellow circle inside orange petal means that there are 88% of genes shared among Corby and Lorraine.

The species L. pneumophila has a highly conserved core genome

a) Most eukaryotic like proteins are conserved in all L. pneumophila genomes

The presence of proteins with high similarity to eukaryotic proteins or proteins with domains preferentially or only present in eukaryotic genomes are a particular feature of L. pneumophila 4 . However, the criteria for identifying these proteins were never clearly defined. To analyze their evolution and possible origin in depth we have thus developed an automatic and systematic method to identify eukaryotic like proteins according to defined criteria. Previously we had identified eukaryotic like proteins in L. pneumophila as proteins with the highest similarity score to eukaryotic proteins according to BLAST results, or by identifying eukaryotic domains 4 . However, due to constantly growing databases BLAST results are changing. Furthermore, recent analyses of amoeba-associated bacteria, in particular symbionts of amoeba have shown that they also contain eukaryotic like proteins, suggesting multiple origins of these proteins in prokaryotes 27 . To get a more complete picture of eukaryotic like proteins of L. pneumophila and also to include those proteins that might have been transferred independently to different amoeba associated bacteria we defined a eukaryotic like protein as i) a protein having a better normalized blast score against eukaryotic sequences than against prokaryotic ones and ii) a protein that did not show BLAST results against neither Legionella spp. nor other bacterial species for which resistance to amoeba infection has been demonstrated (see material and methods). Applying these criteria we identified 46 proteins with putative eukaryotic origin, of which 17 are described here for the first time (Table 2). Given the fact that these proteins were probably acquired by HGT one would expect high diversity in the repertoire. However, our analyses revealed a considerable conservation as more than 50% (26) are conserved in all six L. pneumophila strains, indicating an ancient transfer. Furthermore, they show 89-99% nucleotide identity, probably due to high selection pressure for their maintenance. Thus most of these proteins belong to the core genome, indicating that their acquisition has taken place before the speciation of L. pneumophila. These 26 proteins might have allowed a common Legionella ancestor to colonize an intracellular niche or to adapt better to the intercellular environment of a specific protozoan species leading to the evolution of the species L. pneumophila. Interestingly, 19 of these 26 proteins are also conserved in L. longbeachae, which might thus be those indispensible for intracellular replication of Legionellae (Table 2) 28 .

<p>Table 2</p>

Orthologous eukaryotic like proteins present in the 6 L. pneumophila strains and in L. longbeachae

Name

L. pneumophila strains

L. lo


Product

Paris

Lens

Corby

Lorraine

HL06041035

Philadelphia


Glucoamylase (Glucan 1,4-alpha-glucosidase)

lpp0489

99.31

lpl0465

98.93

lpc2921

99.38

lpo0482

99.45

lpv0523

99.14

lpg0422*

95.92

llo2801

Putative inosine-uridine nucleoside N-ribohydrolase §

lpp0208

98.81

lpl0206

98.64

lpc0223

98.94

SidE protein

lpp0304

98.46

lpl0288

95.25

lpc1602

98.35

lpo0273

97.95

lpv0315

98.28

lpg0234*

94.63

Putative methyltransferase

lpp0358

98.06

lpl0334

89.28

lpc0359

97.93

lpo0334

97.29

lpv0375

97.80

lpg0282

89.28

llo2356

Conserved exported protein of unknown function

lpp0379

99.63

lpl0354

98.53

lpc0380

99.45

lpo0358

99.45

99.82

lpg0301

99.26

Phosphatidylcholine-hydrolyzing phospholipase C §

lpp0565

99.37

lpl0541

98.66

lpc2843

99.34

lpo0571

99.21

lpv0603

99.29

lpg0502

97.87

llo1329

Phytanoyl-CoA dioxygenase domain-containing protein 1

lpp0578

99.25

lpl0554

98.60

lpc2829

99.46

lpo0586

98.60

lpv0619

99.25

lpg0515*

99.35

llo3224

Leucine-rich repeat protein

lpp1007

97.53

lpc2344

97.87

lpo1029

93.94

lpv1082

97.87

lpg0945*

97.54

ecto-ATP diphosphohydrolase II

map

lpp1033

98.95

lpl1000

98.78

lpc2316

98.78

lpo1060

98.69

lpv1110

98.86

lpg0971

98.43

llo1247

Major acid phosphatase Map §

lpp1120

99.06

lpl1124

97.92

lpc0577

98.12

lpo1121

97.93

lpv1267

99.06

lpg1119

98.59

llo1016

Pyruvate decarboxylase

lpp1157

99.70

lpl1162

98.87

lpc0618

99.70

lpo1168

98.69

lpv1308

99.70

lpg1155

98.51

SAM-dependent methyltransferase §

lpp1192

98.38

lpl1198

98.97

lpc0657

99.15

lpo1205

99.06

lpv1346

97.61

lpg1190

99.15

llo1296

Putative 2OG-Fe(II) oxygenase superfamily protein §

lpp1405

100

lpo1449

96.84

lpv1569

100,00

lpg1450

93.74

Phospholipase C §

lpp1411

100

lpl1573

93.12

lpc0870

100,00

lpo1455

97.61

lpv1576

100,00

lpg1455

97.93

llo1329

Putative mitogen-activated protein kinase

thi

lpp1439

99.12

lpl1545

98.11

lpc0898

97.61

lpo1483

98.93

lpv1609

99.12

lpg1483*

97.67

llo1682

Thiamine biosynthesis protein NMT-1

lpp1522

99.04

lpl1461

97.98

lpc0988

99.04

lpo1583

97.88

lpv1700

99.04

lpg1565

97.47

llo0920

Leucine-rich repeat-containing protein

purC

lpp1567

97.68

lpc1028

98.44

lpv1852

98.91

lpg1602*

97.98

Phosphoribosylamidoimidazole-succinocarboxamide synthase

mvaB

lpp1647

100

lpl1640

97.76

lpc1106

99.08

lpo1715

98.98

lpv1936

98.16

lpg1675

97.58

llo3277

Hydroxymethylglutaryl-CoA lyase (HMG-CoA lyase) §

lpp1793

99.34

lpl1794

97.13

lpc1274

98.01

lpo1891

99.01

lpv2102

98.68

lpg1830

99.12

llo0113

Putative apyrase

lpp1880

99.47

lpl1869

98.77

lpc1359

99.74

lpo1975

98.86

lpv2179

99.56

lpg1905

95.34

llo1247

Conserved protein of unknown function

lpp1905

Leucine-rich repeat-containing protein

lpp1940

94.44

lpo2043

93.7

lpv2255

93.88

lpg1958*

92.56

ZIP metal transporter family protein §

lpp2018

99.60

lpl2013

99.07

lpc1521

99.47

lpo2138

99.34

lpv2339

99.47

lpg2035

99.07

llo2518

Ankyrin repeat protein

lpp2058

99.2

lpl2048

90.42

lpc1566

98.80

lpo2181

98.05

Conserved protein of unknown function

lpp2061

99.6

lpl2051

95.95

lpc1569

96.80

lpo2185

95.38

Sphingosine-1-phosphate lyase I

lpp2128

98.84

lpl2102

98.29

lpc1635

99.06

lpo2245

98.62

lpv2428

98.02

lpg2176*

94.02

Conserved protein of unknown function

lpp2134

100

lpl2109

98.55

lpc1642

100

lpo2253

99.60

lpv2436

100

lpg2182

96.27

Conserved protein of unknown function

lpp2419

99.84

lpl2298

99.37

lpc2129

100

Leucine rich repeat protein

lpp2459

98.98

lpl2316

86.85

lpc2085

90.48

lpo2572

99.43

lpv2704

99.32

lpg2392*

97.28

Putative unspecific monooxygenase

lpp2468

99.47

lpl2326

99.01

lpc2075

98.88

lpo2586

98.15

Protein kinase-like

lpp2626

94.88

lpl2481

98.85

lpc1906

95.31

lpo2765

98.70

lpv2900

99.13

lpg2556*

99.13

llo2218

Putative methyltransferase

lpp2747

99.25

lpl2620

99

lpc0443

99.37

lpo2974

97.49

lpv3039

99.62

lpg2693

99.37

llo2356

Phytanoyl-CoA dioxygenase, PhyH

lpp2748

99.76

lpl2621

98.91

lpc0442

99.15

lpo2975

98.67

lpv3040

99.76

lpg2694*

95.44

Sugar kinase §

hemG

lpp2874

99.38

lpc3108

98.89

lpo3114

98.15

lpv3175

99.14

lpg2821

98.52

Protoporphyrinogen oxidase §

cysK

lpp2909

98.14

lpl2763

96.65

lpc3136

98.90

lpo3153

98.69

lpv3207

98.83

lpg2851

96.02

llo0133

Cysteine synthase A, O-acetylserine sulfhydrolase A subunit

lpp3022

99.26

lpl2880

98.95

lpc3266

95.99

lpo3279

99.16

lpv3334

99.26

lpg2951

98.52

llo0076

Putative methyltransferases §

lpp3025

98.50

lpl2883

97.06

lpc3269

99.30

lpo3282

97.62

lpv3338

97.54

lpg2954

97.76

llo0074

Flavanone 3-dioxygenase §

lpo1380

Protein of unknown function §

lpo1577

Conserved protein of unknown function with SNARE domain §

lpc2110

98.97

lpo2553

97.25

lpv2681

98.97

(S)-2-hydroxy-acid oxidase §

lpo2960

Protein of unknown function §

lpo3145

100,00

lpv3199

94.82

Putative Pyridine nucleotide-disulphide oxidoreductase

lpl2845

95.59

lpc3225

97.70

lpo3239

98.47

lpv3288

97.80

lpg2917

98.28

Regulator of chromosome condensation, rcc

lpv2481

79.24

lpg2224*

99.83

Putative metallophosphoesterase §

lpv2663

Serine carboxypeptidase

lpv3278

97.64

lpg2911

100

* Substrates of the Dot/Icm secretion system; § eukaryotic like proteins newly identified in this study; numbers, % nucleotide identity to strain Philadelphia; L.lo, Legionella longbeachae

b) Eukaryotic protein motifs are highly conserved among the L. pneumophila genomes

A second class of eukaryotic proteins of L. pneumophila is carrying domains predominantly present in eukaryotic proteins. To systematically identify these proteins we used the Interpro database comprising 10 different domain search programs 29 . This allowed to identify the L. pneumophila proteins carrying eukaryotic domains in the newly sequenced strains Lorraine and HL 0604 1035 as well as to identify previously not reported motifs. Similarly to the above described eukaryotic like proteins over half of the eukaryotic domain coding proteins are conserved in all six genomes and over 80% are conserved when two genomes are compared (e.g. 33 of the 39 proteins containing an eukaryotic motif in strain Lens are present also in strain Paris). Moreover half of them share very high nucleotide identity of in average 98%-100% (Table 3) again suggesting high selection pressure to maintain them.

<p>Table 3</p>

Orthologous proteins with eukaryotic motifs present in the 6 L. pneumophila strains and in L. longbeachae

Motif

L. pneumophila strains

L. lo


Paris

Lens

Philadelphia

Lorraine

HL06041035

Corby


ANK

lpp0037

96.30

lpl0038

97.40

lpg0038*

97.04

lpo0042

97.89

lpv0043

93.66

lpc0039

97.10

ANK

lpp0126

98.94

lpl0111

98.48

lpg0112

94.83

lpo0119

98.79

lpv0127

93.03

lpc0131

92.16

llo1394

ANK

lpp0202

ANK

lpp0356

ANK

lpp0469

98.94

lpl0445

96.35

lpg0403*

95.53

lpo0463

97.64

lpv0501

98.48

lpc2941

98.67

ANK

lpp0503

98.37

lpl0479

93.86

lpg0436*

93.31

lpo0501

98.12

lpv0537

98.37

lpc2906

98.37

ANK

lpp0547

99.50

lpl0523

96.31

lpg0483*

96.82

lpo0551

99.83

lpv0585

98.16

lpc2861

99.16

llo2705

ANK

lpp0750

100.00

lpl0732

97.65

lpg0695*

100.00

lpo0775

99.84

lpv0817

100.00

lpc2599

98.44

ANK

lpp1100

ANK + SET

lpp1683

97.68

lpl1682

96.32

lpg1718*

98.41

lpo1757

97.86

lpv1985

96.91

lpc1152

97.25

ANK

lpp1905

ANK

lpp2058

99.20

lpl2048

90.42

lpo2181

98.05

lpc1566

98.80

ANK

lpp2061

99.60

lpl2051

95.95

lpo2185

95.38

lpc1569

96.80

ANK

lpp2065

99.93

lpl2055

98.56

lpo2189

98.62

lpc1573

98.03

ANK + Fbox

lpp2082

97.40

lpl2072

98.26

lpg2144*

98.84

lpo2207

99.03

lpv2392

93.99

lpc1593

99.22

ANK

lpp2166

99.25

lpl2140

99.12

lpg2215*

99.06

lpo2285

97.74

lpv2469

99.18

lpc1680

98.93

ANK

lpp2248

99.50

lpl2219

99.14

lpg2300*

99.14

lpo2371

99.43

lpv2567

98.93

lpc1765

99.21

llo0584

ANK

lpp2270

99.64

lpl2242

97.97

lpg2322*

98.34

lpo2399

98.08

lpv2591

99.53

lpc1789

99.53

llo0570

ANK

lpp2517

99.60

lpl2370

97.94

lpg2452*

98.19

lpo2642

98.95

lpv2776

99.46

lpc2026

99.46

ANK

lpp2522

98.76

lpl2375

96.90

lpg2456*

95.75

lpo2647

98.49

lpv2781

98.76

lpc2020

91.27

llo0365

ANK

plpp0098

96.00

lpop0045

96.00

ANK

lpl1681

100.00

lpc1151

97.98

ANK

lpl2058

86.17

lpo2193

95.37

lpv2375

94.96

ANK

lpl2339

98.64

lpg2416*

91.21

lpo2601

99.00

lpv2736

99.28

lpc2057

98.98

ANK

lpg0402*

100.00

lpv0500

96.01

ANK

lpv2258

ANK

lpl1681

100.00

lpc1151

97.98

ANK

lpl2344

100.00

lpo2607

97.93


F-Box

lpp0233

98.58

lpl0234

93.97

lpg0171*

96.81

lpo0202

97.87

lpv0254

98.94

F-Box

lpp2486

F-Box

lpg2224*

99.83

lpv2482

79.24

F-Box

lpv2481


RAS GEF

lpp0350

94.53

lpl0328

96.32

lpg0276*

97.33

lpo0327

97.64

lpv0368

97.64

lpc0353

llo0327

RAS GEF§

lpv2258


Sec7

lpp1932

98.41

lpl1919

97.40

lpg1950*

92.16

lpo2033

98.32

lpv2243

98.58

lpc1423

97.57

llo1397


Sel1

lpc0165

Sel1

lpl1059

100.00

lpg1062

99.61

lpv1209

100.00

lpc2212

99.61

Sel-1§

lpp0957

98.93

lpl0927

98.67

lpg0896

98.93

lpo0978

98.67

lpv1030

98.67

lpc2397

99.47

llo0844

Sel-1

lpp1174

99.39

lpl1180

98.30

lpg1172

98.32

lpo1187

99.11

lpv1327

99.39

lpc0638

99.05

Sel-1

lpp1310

97.87

lpl1307

98.40

lpg1356

98.67

lpo1345

99.02

lpv1469

97.87

lpc0770

98.76

llo1443

Sel-1

lpp2174

99.64

lpl2147

98.48

lpg2222*

99.56

lpo2292

99.47

lpv2477

99.47

lpc1689

96.27

Sel-1

lpp2692

99.25

lpl2564

98.61

lpg2639

98.39

lpo2917

99.28

lpv2979

99.39

lpc0501

98.75

llo2649

Sel-1

lpo3233


Spectrin

lpp1848§

99.18

lpl1845

98.77

lpg1884*

99.01

lpo1944§

98.93

lpv2158§

99.18

lpc1331

99.18

Spectrin

lpp2246

99.29

lpl2217

98.75

lpg2298*

99.29

lpo2369

99.29

lpv2565

98.27

lpc1763

98.75

llo1707

Spectrin

lpp1930

95.11

lpg1947*

96.65

lpo2029

97.72

Spectrin

lpp1309

100.00

lpg1355*

90.59

lpv1468

100.00

Spectrin §

lpp1002

98.01

lpl0971

91.62

lpg0940*

97.92

lpo1024

98.05

lpv1077

97.87

lpc2349

97.15

Spectrin §

lpp0471

97.79

lpl0447

97.45

lpg0405*

98.30

lpo0465§

98.28

lpv0504

98.28

lpc2939

97.70

llo2845

Spectrin §

lpp1843

95.45

lpl1840

97.57

lpv2151

100.00

lpc1323§

99.60

Spectrin §

lpp1173

98.56

lpl1179§

98.80

lpg1171*§

98.56

lpo1186

99.28

lpv1326

98.56

lpc0637

97.84

llo3114

STPK

lpp0267

96.95

lpl0262

98.72

lpg0208

93.26

lpo0242

98.92

lpv0288

95.13

lpc0283

97.26

STPK

lpp1439

99.12

lpl1545

98.11

lpg1483*

97.67

lpo1483

98.93

lpv1609

99.12

lpc0898

97.61

llo1682

STPK

lpp2626

94.88

lpl2481

98.85

lpg2556*

99.13

lpo2765

98.70

lpv2900

99.13

lpc1906

95.31

llo2218

U-box

lpp2887

99.72

lpg2830*

97.15

lpo3124

99.58

lpv3185

98.75

*Substrates of the Dot/Icm secretion system according to previous publications; orthologs proteins where the corresponding motif was not present in the other genome; § eukaryotic like proteins newly identified in this study; numbers, nucleotide identity with respect to the L. pneumophila Philadelphia gene; L.lo, Legionella longbeachae

Our approach identified also new eukaryotic domains like spectrin repeats. The spectrin repeat forms a three-helix bundle and was reported primarily in the animal kingdom 30 . These repeats act as modules building long, extended molecules that also serve as a docking surface for cytoskeletal and signal transduction proteins. In L. pneumophila it is present in up to eight proteins of each strain (Table 3) and all spectrin repeat proteins are predicted to be secreted Dot/Icm substrates 31 32 33 . Another interesting domain is the RAS GEF domain that is present in two proteins encoded by strain Paris one of which (Lpp0350) is conserved in the six strains analyzed. Ras-GEFs are small GTPases typically present in eukaryotes that are involved in numerous cellular processes like gene expression, cytoskeleton re-organization, microtubule organization and vesicular and nuclear transport 34 . GEFs (GDP-GTP exchange factors) regulate Rabs, GTP-binding proteins with conserved functions in membrane trafficking 35 . Interestingly, according to the Pfam database Ras-GEF domains in bacteria are only present in Legionella, Parachlamydia acanthamoebae and Protochlamydia amoebophila, all of which are amoeba-associated bacteria.

Coiled-coil domains have been identified previously in the L. pneumophila genomes as this motif can be found in all kingdoms of life. However extended coiled-coil domains are largely absent from bacterial genomes but are typical for archaea and eukaryotes. We thus searched the L. pneumophila genomes and 29 other genomes of bacterial pathogens or bacteria present in the aquatic environment (Table 4) for proteins with five or more coiled coil domains. Interestingly, Legionella spp, Streptococcus pneumoniae and Pseudomonas aeruginosa contain the highest percentage of proteins with extended coiled-coil domains (6-11 domains) compared to the number of predicted proteins encoded in their genome and only P. aeruginosa and L. pneumophila encode proteins containing more than 10 coiled-coil domains (Table 4). Most of these Legionella proteins are predicted substrates of the Dot/Icm secretion system 31 32 33 36 . This suggests that large coiled-coil domains are specific adaptations to the eukaryotic cell probably implicated in interactions with host proteins.

<p>Table 4</p>

Genes coding for proteins with more than 5 coiled coil domains/protein in different bacterial genomes

Organism

Coiled coil domains proteins

Gene

Product

Number of Coiled coil


B. henselae Houston-1

0


Ch. pneumoniae J138

0


Ch. trachomatis D UW-3

0


C.glutamicum ATCC 13032

0


E. coli O157:H7

1

ECH74115_2173

tail length tape measure protein

5


H. influenzae Rd KW20

0


H. pylori 26695

1

HP0527

cag pathogenicity island protein Y

10


L. pneumophila Corby

7

lpc1130

substrate of the Dot/Icm system/Icm system

5

lpc1131

substrate of the Dot/Icm system/Icm system

6

lpc1452

substrate of the Dot/Icm system/Icm system

6

lpc1611

hypothetical protein

12

lpc1987

substrate of the Dot/Icm system, effector protein B

9

lpc2349

substrate of the Dot/Icm system, LidA

6

lpc3079

substrate of the Dot/Icm system, effector protein A

5


L. pneumophila HL06041035

10

lpv1077

substrate of the Dot/Icm system, LidA

6

lpv1725

substrate of the Dot/Icm system

6

lpv1966

substrate of the Dot/Icm system

5

lpv1967

substrate of the Dot/Icm system

6

lpv2269

substrate of the Dot/Icm system

7

lpv2408

conserved protein of unknown function

5

lpv2816

substrate of the Dot/Icm system, effector protein B

10

lpv2959

chromosome segregation SMC protein

9

lpv3144

substrate of the Dot/Icm system, effector protein A

5

lpv3184

substrate of the Dot/Icm system, SidH

9


L. pneumophila Lens

7

lpl1437

substrate of the Dot/Icm system

6

lpl1660

substrate of the Dot/Icm system

7

lpl1661

substrate of the Dot/Icm system

6

lpl1941

substrate of the Dot/Icm system

5

lpl2084

substrate of the Dot/Icm system

5

lpl2411

substrate of the Dot/Icm system, effector protein B

9

lpl2708

substrate of the Dot/Icm system, effector protein A

5


L. pneumophila Lorraine

10

lpo1024

substrate of the Dot/Icm system, LidA

6

lpo1608

substrate of the Dot/Icm system

6

lpo1735

substrate of the Dot/Icm system

7

lpo1736

substrate of the Dot/Icm system

5

lpo2060

substrate of the Dot/Icm system

6

lpo2216

substrate of the Dot/Icm system, SdeC

5

lpo2680

substrate of the Dot/Icm system, effector protein B

9

lpo2896

chromosome segregation SMC protein

9

lpo3083

substrate of the Dot/Icm system, effector protein A

5

lpo3123

substrate of the Dot/Icm system

9


L. pneumophila Paris

6

lpp1002

substrate of the Dot/Icm system, LidA

6

lpp1546

substrate of the Dot/Icm system

6

lpp1666

substrate of the Dot/Icm system

7

lpp1952

substrate of the Dot/Icm system

6

lpp2555

substrate of the Dot/Icm system, effector protein B

10

lpp2883

substrate of the Dot/Icm system

6


L. pneumophila Philadelphia

8

lpg1355

substrate of the Dot/Icm system, SidG protein

5

lpg1588

substrate of the Dot/Icm system

6

lpg1701

substrate of the Dot/Icm system

5

lpg1702

substrate of the Dot/Icm system

6

lpg2156

protein of unknown function

5

lpg2490

substrate of the Dot/Icm system, effector protein B

9

lpg2793

substrate of the Dot/Icm system, effector protein A

5

lpg2829

substrate of the Dot/Icm system

8


L. monocytogenes EGD-e

3

lmo0650

hypothetical protein

5

lmo0955

hypothetical protein

5

lmo1224

hypothetical protein

5


M. tuberculosis F11

1

TBFG_12936

chromosome partitioning protein Smc

10


M. tuberculosis H37Ra

1

MRA_2947

putative chromosome segregation Smc

10


N. meningitidis MC58

0


P. aeruginosa LESB58

11

PLES_08211

putative tail length tape measure protein

7


PLES_12531

hypothetical protein

7

PLES_12541

hypothetical protein

5

PLES_13581

putative tail length tape measure protein

7

PLES_15241

electron transport complex protein RnfC

8

PLES_15871

hypothetical protein

6

PLES_36651

putative ClpA

_

PLES_38011

putative chromosome segregation protein

11

PLES_46621

putative exonuclease

13

PLES_50721

hypothetical protein

6

PLES_55491

putative outer membrane protein precursor

5


R. felis URRWXCal2

2

RF_0022

putative surface cell antigen sca1

7

RF_0725

antigenic heat-stable 120 kDa protein

5


R.prowazekii Madrid E

0


R.a typhi Wilmington

0


S. typhimurium LT2

5

STM0395

exonuclease subunit SbcC

7


STM0567

putative DNA repair ATPase

7

STM0994

chromosome partition protein mukB

10

STM1041

minor tail protein

5

STM3199

hypothetical protein

5


S. flexneri 2a 2457T

1

S0984

fused chromosome partitioning protein

10


Synechocystis sp. PCC 6803

2

sll1772

MutS2 protein

5

slr1301

hypothetical protein

6


S. pneumoniae D39

4

SPD_0126

exported protein of unknown function

6


SPD_0710

putative Septation ring formation regulator EzrA

7

SPD_1104

chromosome partition protein Smc

10

SPD_2017

exported protein of unknown function

6


W. pipientis wMel

0


X. fastidiosa 9a5c

0


Y. pestis KIM

4

y0227

hypothetical protein

6


y0976

ATP-dependent dsDNA exonuclease

12

y2765

chromosome partition protein MukB

10

yapB

autotransporter

6

c) High selection pressure acts on the Dot/Icm T4SS and its substrates

Central to the pathogenesis of L. pneumophila are the dot/icm loci, which together direct assembly of a type IV secretion apparatus 37 38 . Although all L. pneumophila strains investigated to date contain the complete dot/icm loci, sequence variations among the dot/icm genes among different L. pneumophila strains have been reported 39 . The dot/icm loci of the six strains analyzed here exhibited a very high nucleotide conservation of 98-100% among orthologs except for dotA, icmX and for icmC of strain Corby that is shorter and more divergent (84% nucleotide identity) as compared to icmC of strain Paris. These results indicate that strong negative selection acts on these genes (Table 5).

<p>Table 5</p>

Percentage of nucleotide identity of orthologous dot/icm genes with respect to the L. pneumophila Philadelphia sequence

Gene name

Length (nts)

Phila

Paris

Id

Lens

Id

Lorrain

Id

HL06041035

Id

Corby

Id

L. long

Id


icmT

261

lpg0441

lpp0507

99.6

lpl0483

99.1

lpo0507

100

lpv0541

96

lpc2902

99.2

llo2795

75.2

icmS

345

lpg0442

lpp0508

98.5

lpl0484

98.8

lpo0508

99.1

lpv0542

94.4

lpc2901

98.3

llo2794

76.9

icmR

363

lpg0443

lpp0509

96.9

lpl0485

98.3

lpo0509

97.8

lpv0543

97.5

lpc2900

96.9

IcmQ

576

lpg0444

lpp0510

97

lpl0486

99

lpo0510

98

lpv0544

98

lpc2899

98

llo2792

70.7

icmP/dotM

1131

lpg0445

lpp0511

98

lpl0487

99

lpo0511

98

lpv0545

98

lpc2898

99

llo2791

74.5

icmO/dotL

2352

lpg0446

lpp0512

98.4

lpl0488

97.7

lpo0512

98.1

lpv0546

98.3

lpc2897

98.3

llo2790

77.7

IcmN/DotK

570

lpg0447

lpp0513

99.3

lpl0489

98.6

lpo0513

98.9

lpv0547

99.6

lpc2896

99.7

llo2789

67.3

icmM/dotJ

285

lpg0448

lpp0514

97.9

lpl0490

97.9

lpo0514

97.9

lpv0548

99.3

lpc2895

98.6

llo2788

61.7

icmL/dotI

639

lpg0449

lpp0515

99.8

lpl0491

99.4

lpo0515

99.4

lpv0549

99.8

lpc2894

99.5

llo2787

78.6

icmK/dotH

1083

lpg0450

lpp0516

94.8

lpl0492

94.3

lpo0516

95.2

lpv0550

94.4

lpc2893

94.7

llo2786

71.2

icmE/dotG

3147

lpg0451

lpp0517

93.7

lpl0493

94.0

lpo0517

94

lpv0551

94

lpc2892

94.3

llo2785

69.1

icmG/dotF

810

lpg0452

lpp0518

98

lpl0494

97

lpo0518

98

lpv0552

98

lpc2891

97

llo2784

55.7

icmC/dotE

585

lpg0453

lpp0519

99.6

lpl0495

99.1

lpo0519

99.7

lpv0553

99.3

lpc2890

54

llo2783

69.1

icmD/DotP

399

lpg0454

lpp0520

97

lpl0496

98

lpo0520

97

lpv0554

98

lpc2889

97

llo2782

77.3

icmJ/dotN

627

lpg0455

lpp0521

99

lpl0497

98

lpo0521

99

lpv0555

99

lpc2888

98

llo2781

79.4

IcmB/DotO

3030

lpg0456

lpp0522

98.1

lpl0498

98.3

lpo0522

98.3

lpv0556

98.2

lpc2887

97.6

llo2780

76.4

IcmF

2922

lpg0458

lpp0524

98.2

lpl0500

98.5

lpo0524

98.3

lpv0558

98.5

lpc2885

98.2

llo3075

69.5

IcmH/DotU

786

lpg0459

lpp0525

99.4

lpl0501

99.5

lpo0525

99.7

lpv0559

99

lpc2884

99

llo3074

68.8

dotD

492

lpg2674

lpp2728

98

lpl2601

98

lpo2953

98

lpv3018

98

lpc0463

99

llo0369

76.5

dotC

912

lpg2675

lpp2729

98.7

lpl2602

98.5

lpo2954

98.8

lpv3019

98.6

lpc0462

99.9

llo0368

74.8

dotB

1134

lpg2676

lpp2730

99

lpl2603

98

lpo2955

98

lpv3020

98

lpc0461

99

llo0367

76

dotA

3108

lpg2686

lpp2740

83.3

lpl2613

96.8

lpo2967

83

lpv3032

83.6

lpc0450

85.8

llo0364

51.4

icmV

456

lpg2687

lpp2741

91

lpl2614

91

lpo2968

91

lpv3033

92

lpc0449

92

llo0363

64.3

icmW

456

lpg2688

lpp2742

95.1

lpl2615

97.6

lpo2969

95.1

lpv3034

95.4

lpc0448

95.1

llo0362

79.3

icmX

1419

lpg2689

lpp2743

84.3

lpl2616

85.2

lpo2970

85.6

lpv3035

85.6

lpc0447

84.1

llo0361

46.9

Id, identity

Since the identification of RalF 40 , numerous approaches have been used to identify Dot/Icm translocated substrates. Currently 278 proteins of L. pneumophila have been described as being transloctaed by the Dot/Icm T4SS system 7 31 32 41 42 43 44 . Analysis of their distribution among the six L. pneumophila strains reveals a very high conservation, as 206 of the 278 substrates are present in all six strains. Nearly all of them show a nucleotide similarity of 95-100% and only nine are specific to strain Philadelphia (Additional file 1, Table S1). Furthermore, only 34 of the 278 substrates of strain Philadelphia are missing in strain Paris, 30 in strain Lorraine or 25 in strain HL 0604 1035 (Additional file 1, Table S1). Thus, although high redundancy seems to be present in the repertoire of Dot/Icm effectors, the strong conservation of nearly all of them in all genomes, argues for their mutual importance for the L. pneumophila life cycle,

<p>Additional file 1</p>

Table S1: Nucleotide identity of 140 selected Dot/Icm substrates of strain Philadelphia and of their orthologs in the L. pneumophila strains analyzed in this study.

Click here for file

Rare exceptions are RalF and AnkB/Lpp2028. The nucleotide sequence of ralF of strain Philadelphia is only 85% similar to the ralF genes of the other strains and is 72 nts (24aa) shorter. A similar situation is seen for lpg2144/ankB that is 54 nts (18aa) longer in strain Philadelphia and Lens than in strain Paris and Corby. This is surprising, as the C-terminal region of AnkB of strain Philadelphia contains a eukaryotic prenylation CAAX motif mediating posttranslational modification of effector proteins, important for intracellular replication of L. pneumophila. Lipidation facilitates the localization of this effector protein to host organelles and serves as a docking platform for ubiquitinated proteins 45 46 . Thus in strain Paris and Corby other proteins might take over this function. Taken together, this analysis suggests that over 200 of the Dot/Icm substrates of L. pneumophila have been present or have been acquired before the speciation and that such a large repertoire of effectors is indeed necessary for intracellular replication and adaptation to the specific protozoan hosts.

The species L. pneumophila has a highly dynamic accessory genome

a) A wide variety of T4ASSs and conjugative elements contribute to genome plasticity

Based on sequence comparisons, T4SSs are categorized according to their similarity to the A. tumefaciens VirB/D4 system into type IVA (type F and P) and type IVB secretion systems 47 . T4ASSs resemble the VirB/D4 system of A. tumefaciens, whereas T4BSS proteins are more distantly related to the VirB/D4 proteins 48 . T4SSs are involved in effector translocation, horizontal DNA transfer to other bacteria and eukaryotic cells, in DNA uptake from or release into the extracellular milieu or in the spread of conjugative plasmids 49 . Genome sequence analyses suggest that for L. pneumophila T4SSs play an important role for adaptation and virulence as each genome encodes several T4ASSs in addition to the essential T4BSS Dot/Icm discussed above. We identified in each strain either F-type or P-type T4ASSs or both. Figures 2 and Figure 3 show the organization of the structural genes encoding these systems, their organization and their localization (chromosomal or plasmid). The F-type T4ASSs are all predicted to encode a complete T4SS core as well as the essential gene products for pilus assembly and mating pair stabilization that appears to be involved in DNA transfer. They show homology and colinearity with the tra-region of the E. coli F plasmid 50 and with the recently described tra region of Rickettsia belii 51 . In L. pneumophila strain Philadelphia (Tra5) and L. longbeachae strain NSW (Tra6), where the system has a chromosomal localization, it is inserted in a tRNA gene and flanking repeats are present as well as a gene coding for an integrase, suggesting that these T4SSs are mobile (Figure 2). Furthermore, comparison of amino acid identities revealed that the Tra- region on the L. pneumophila strain Paris plasmid (Tra1) shows much higher identity with the Tra region located on the L. longbeachae plasmid (Tra4) than with those of the different L. pneumophila strains (Paris-Tra1, Lens-Tra3 or Lorraine-Tra2) (Figure 2). Thus these systems seem to be transferred horizontally via plasmids but are also able to integrate in the genome similar to what was reported for the Lvh-region 52 .

<p>Figure 2</p>

Schematic representation of F-type IV secretion systems (T4SSA) for conjugal DNA transfer of L. pneumophila

Schematic representation of F-type IV secretion systems (T4SSA) for conjugal DNA transfer of L. pneumophila. In green and orange, tra and trb genes respectively. L. long, Legionella longbeachae; P, Plasmid; C, Chromosome; ycaO, Protein of unknown function with a YcaO like-domain; tfu, Protein of unknown function with a TfuA domain; pil, Pilus assembly protein precursor; t, transposase; E. coli, Escherichia coli; R. beeli, Rickettsia beeli; pha, Phage repressor; int, integrase; pin, site-specific DNA recombinase e14 prophage; R; repeat. Yellow squares represent flanking repeats, with length and percentage of identity between repeats in parenthesis. tRNAs, position in the genome in parenthesis.

<p>Figure 3</p>

Schematic representation of P-type IV secretion systems (T4SSA) for conjugal DNA transfer of L. pneumophila

Schematic representation of P-type IV secretion systems (T4SSA) for conjugal DNA transfer of L. pneumophila. In green and orange, tra and trb genes respectively. L. long, Legionella longbeachae; P, Plasmid; C, Chromosome; ?, Protein of unknown function; A. tumefaciens, Agrobacterium tumefaciens; P. aeruginosa, Pseudomonas aeruginosa; t, transposase; pha, Phage repressor; Int, integrase; Pseudogenes are in discontinues squares; Yellow squares represent flanking repeats, with length and percentage of identity between repeats in parenthesis. tRNAs, their position in the genome is given in parenthesis.

The F-type T4SS encode long, flexible pili that allow donors to mate in liquid and on solid media with equal efficiencies 53 . In contrast P-type T4SS like described in P. aeuroginosa encode short and rigid conjugative pili that allow surface mating. Homologues to this system are also present in the Legionella genomes. They were initially described in two genomic islands of L. pneumophila strain Corby (Figure 3; Trb1 and Trb2) 54 . We show here that they are also present in the chromosomes of L. pneumophila strain Lorraine (Trb3) and L. longbeachae NSW150 (Trb4) (Figure 3). Again for all T4SS regions flanking repeats are found suggesting mobility, and protein identity values and GC-content values of the tra-trb genes are higher than the genomic average (38%), supporting again horizontal and not vertical transmission.

Another intriguing feature of these regions is that several transposases and phage related proteins are present in each of the tra clusters as well as genes coding for homologues of a putative phage repressor protein (PrpA) and for homologues of LvrA, LvrB and LvrC, first described for the Lvh region of L. pneumophila. LvrC is a homologue of CsrA, a protein crucial for the regulation of the switch between replicative and transmissive phase of L. pneumophila 55 . It is tempting to assume that these CsrA homologues are implicated in the regulation of the mobility of these islands. Possibly, dependent on the growth phase and/or on metabolic cues L. pneumophila might excise these islands as multiple copies could be advantageous in certain conditions, or perhaps allow high frequencies of DNA transfer leading to fast and efficient adaptation to new conditions. The genomic features of these islands suggest a particular mechanism of mobility, which will be interesting to investigate.

b) The L. pneumophila genomes encode systems specific for protection against invading DNA and stabilization of large genomic fragments

Bacteria have developed multiple methods of protection against mobile genetic elements or bacteriophages. An example for acquired phage specific immunity is clustered regularly interspaced short palindromic repeats (CRISPR) loci 56 . Another type of protection may be conferred by toxin-antitoxin (TA) systems. Bacterial TA systems are small genetic modules composed of a toxin and antitoxin. While toxins are always proteins, antitoxins are either RNAs (type I and III) or proteins (type II) 57 . These systems were first described for being dedicated to plasmid maintenance. Several lines of research indicate that chromosomal TA systems might serve as protection against mobile genetic elements such as plasmids and phages. However, recent studies have shown that type II systems are also involved in the stabilization of large genomic fragments and of integrative conjugative elements 57 . Interestingly, type II TA systems are thought itself to be part of the mobilome and to move from one genome to another through horizontal gene transfer 57 .

Genome analyses identified several TA and CRISPR systems. Interestingly, we identified only type II TA systems of which all except two are in a chromosomal location (Table 6). However, of the 18 chromosomal encoded TA systems identified at least 14 are located on putative genomic islands or mobile genetic elements. The two most frequently found TA systems in the L. pneumophila genomes are homologues of the HigAB and RelEB systems. HigAB was first described in the Vibrio cholerae superintegron where it encodes mRNA cleaving enzymes and can stabilize plasmids 58 . RelEB was shown, when introduced into the E. coli chromosome to prevent deletion of flanking DNA and thus to diminish large scale genome reduction 59 . The same function was shown for the ParED system of Vibrio vulinificus, homologues of which are also present in one of the L. pneumophila genomes (Table 6). Thus, the different L. pneumophila TA systems might be important for stabilization of plasmids and integrative conjugative elements and for protection against invasion of plasmids, phages, or other mobile genetic elements.

<p>Table 6</p>

Genes encoding putative toxin-antitoxin systems in six L. pneumophila genomes

L. pneumophila strains


Toxin-antitoxin

Paris

Lens

Philadelphia

Corby

Lorraine

HL06041035


higA

lpl2833 (96)*

lpg2914 (96)

lpv3285 (96)

higB

lpl2834 (87)*

lpg2915 (103)

lpv3286 (103)

higA

lpl1092 (93)*

higB

lpl1093 (107)*

higA

lpp0064 (434)*

lpo0072 (432)*

higB

lpp0065 (79)*

lpo0073 (79)*

lpc2112 (312)

lpl2291 (102)*

lpg2369 (102)

lpc2113 (37)

lpv2676 (102)*

Similar to hipA

lpp2427 (78)*

lpl2292 (312)*

lpg2370 (312)

lpc2114 (65)

lpo2551 (115)*

lpv2677 (310)*

yhvA

lpo1074 (168)*

sohA

lpo1075 (115)*

relE

plpp0090 (83)

lpl1587 (82)*

relB

plpp0089 (95)

lpl1588 (85)*

relE

lpl1084 (84)*

lpc2177 (93)*

lpo0120 (93)*

relB

lpc2178 (88)*

lpo0119 (86)*

parE

lpe2361 (98)*

parD

lpe2360 (84)*

pemK

lpo0114 (106)

*TA systems located on putative genomic islands; In parenthesis length of the corresponding protein

The CRISPR/cas system was shown to provide resistance against invading viruses and plasmids and has been identified in many bacteria and archea 60 . CRISPR/cas loci are also present in the L. pneumophila genomes of strains Paris, Lens, Alcoy and 130 b but are absent from strains HL06041035 and Lorraine. According to the cas genes, the CRISPR locus of Paris is closely related to that of strain 130 b. In contrast the one of strain Lens located on the plasmid is closely related to the chromosomal CRISPR locus of strain Alcoy as previously described 61 . Strain Lens carries a second CRISR locus on the chromosome; however, it does not seem to be functional like the one encoded by strain Alcoy. Probably strong protection against invading phages is not extremely important, as not all L. pneumophila strains contain CRISPR loci. This may be related to their intracellular life style or that despite their widespread occurrence in aquatic environments only few bacteriophages that specifically infect Legionella seem to exist 62 .

c) Accessory genome of strains Lorraine and HL 0604 1035

In order to get insight in the genetic basis of the two newly sequenced strains, possibly implicated in their different disease frequencies (Lorraine is an newly emerging endemic clone and strain HL 0604 1035 is a L. pneumophila Sg1 strain never isolated from disease) we analyzed the specific gene content of each of these strains more in depth. Strain HL 0604 1035 contains 92 and strain Lorraine 148 genes without homology to any gene of the other five L. pneumophila strains sequenced of which the majority (60 in strain HL 0604 1035 and 73 in strain Lorraine) code for proteins of unknown function (Additional file 2, Tables S2 and additional file 3, Table S3). Among the genes in these two genomes that lack an ortholog in the other sequenced L. pneumophila genomes, about 50% are clustered on three large genomic islands. One genomic Island (GI-HL1) of 45 kb spans from lpv2637 to lpv2691. It is bordered by a Met tRNA gene and encodes a phage related integrase. A second putative mobile element (GI-HL2) of 27 kbs contains the region from lpv0193 to lpv0226. It is bordered at one side by an integrase and a reverse transcriptase (lpv0225) and on the other side by a prophage Rac integrase and a phage excisionase. Strain Lorraine contains also a large genomic island (GI-Lo1) of 69 kb that spans from lpo2442 to lpo2531. It is inserted in a Met tRNA gene, contains a phage related integrase and flanking repeats of 72 nts. Additional, smaller genomic islands seem to be present, however, their borders are difficult to define. Thus most of the strain specific genes seem to be acquired by HGT through mobility of genomic islands.

<p>Additional file 2</p>

Table S2: Genes specific of strain HL 0604 1035 with respect to strains Paris, Lens, Philadelphia, Corby and Lorraine.

Click here for file

<p>Additional file 3</p>

Table S3: Genes specific of strain Lorraine with respect to strains Paris, Lens, Philadelphia, Corby and HL0604 1035.

Click here for file

Only for few of the specific genes a putative function can be predicted like genes coding for proteins involved in sugar and nucleotide metabolism, for uridine diphosphoglucuronate 5'-epimerase or for an UDP-glucose 6-dehydrogenase. Furthermore a specific ANK motif containing protein and a leucine reach repeat protein are present in strain HL 0604 1035. In strain Lorraine we identified mainly specific metabolic enzymes like a putative flavanone 3-dioxygenase, an enzyme involved in flavonoids metabolism and in biosynthesis of phenylpropanoids, which are secondary metabolites of plants and algae. In addition, lpo2614 is predicted to encode a kynurenine-oxoglutarate transaminase, an enzyme that is part of the tryptophan metabolism and lpo2960 codes for a putative glycolate oxidase that catalyses the conversion of glycolate and oxygen to glyoxylate and hydrogen proxide. lpo2502 codes a homologue of CsbD, a general stress response protein of Bacillus subtilis 63 . However, the best BLASTp hit is with the Protochlamydia amoebophila homologue, an Acanthamoeba sp. symbiont 64 . Probably this gene has been acquired by HGT between these two bacteria within their amoeba host. Quite surprisingly, we identified a gene coding a putative methyl-accepting chemotaxis sensory transducer (lpv1770) although all L. pneumophila strains analyzed to date do not encode chemotaxis systems. This gene shares 71.34% amino acid identity with Llo3301 of L. longbeachae a protein that is part of its chemotaxis system 28 also present in L. drancourtii 65 . Probably a common ancestor encoded a chemotaxis system that was lost in L. pneumophila through a deletion and degradation process.

d) Shared genome of the epidemic strains Paris and Lorraine

A search for genes shared by the two endemic strains but absent in all other strains identified only three genes that fulfilled these criteria and for which a function could be predicted. These encode the alpha, beta and gamma subunits of a putative thiocyanate hydrolase (lpo1236, lpo1237, lpo1238 and lpp1219, lpp1220, lpp1221). Most interestingly, these strains are both common in France and strain Paris is also world-wide distributed 10 suggesting a better niche adaptation. Indeed, thiocyanate compounds are used for cleaning water circuits and these strains are thus probably able to better resist these treatments 66 . Furthermore, strain Alcoy that is responsible for several outbreaks and many cases of Legionnaires' disease in Spain, also contains these genes 61 . The genes coding the putative thiocyanate hydrolase have a GC content of 41-43%, which is significantly higher than the average G+C content of the L. pneumophila genome, which is 38%. When searching for the closest homologues according to BLAST searches we identified them in the genomes of Rhodococcus opacus strain B4 and Nocardia farcinica spp. These two are high G+C Gram-positive bacteria belonging to the Actinomycetales, which are phylogenetically not closely related to Legionella suggesting that L. pneumophila acquired these genes by horizontal gene transfer.

Taken together, the analysis of the accessory gene content showed again that L. pneumophila genomes show high plasticity due to mobile genetic elements and HGT. No specific virulence related genes explaining their different disease frequencies have been identified. However, the identification of a specific thiocyanate hydrolase might explain the wide distribution of strains Paris and Lorraine as it may allow them to better adapted to artificial water systems.

Evolutionary genomics

Phylogenetic reconstruction reveals extensive recombination

To analyze the relationship among the six different L. pneumophila strains a phylogenetic reconstruction was done based on a multilocus sequence (MLSA) approach using 31 genes selected according to Zeigler 67 (Table 7 and Additional file 4, Table S4). These 31 genes were chosen as they had been shown to be powerful for predicting the relatedness of bacterial genomes 67 . The phylogeny obtained from their concatenated alignment showed a well-resolved topology with bootstrap values over 50%. To ascertain the reliability of the obtained phylogenetic tree we established individual phylogenies for each of the 31 genes. Surprisingly, the incongruence among several gene trees was high. In addition the Consense program results did not support any node to at least 50%. To further investigate these results we undertook a second analysis using a Shimodaira-Hasegawa test and compared the topologies of the individual alignments of each gene and the concatenated alignment of the 31 genes. As shown in Additional file 5, Table S5 the likelihood-based SH test for alternative tree topologies identified striking discordances. A possible explanation for the identified incongruences among the phylogenies obtained in our study is the presence of recombination events.

<p>Table 7</p>

Characteristics of the 31 genes used for phylogenetic reconstruction

Gene Name

Product

Labela

Function

Length (nts)a


uvrB

Excinuclease ABC, subunit B

lpp0086

DNA replication, recombination, and repair

1992

pgk

Phosphoglycerate kinase

lpp0152

Glycolysis/gluconeogenesis

1191

rpoA

RNA polymerase, alpha subunit

lpp0419

Transcription

993

ffh

Signal recognition particle protein, GTPase

lpp0467

Transport and binding proteins

1377

serS

Seryl tRNA synthetase

lpp0575

tRNA aminoacylation

1281

proS

Prolyl-tRNA synthase

lpp0749

tRNA aminoacylation

1710

glyA

Serine hydroxymethyltransferase

lpp0791

Glycine/serine hydroxymethyltransferase

1254

dnaB

Replicative DNA helicase

lpp0803

DNA replication, recombination, and repair

1383

gpi

Glucose-6-phosphate isomerase

lpp0825

Glycolysis/gluconeogenesis

1500

lig

DNA ligase

lpp1020

DNA replication, recombination, and repair

2022

cysS

Cysteinyl-tRNA synthetase

lpp1271

tRNA aminoacylation

1371

trpS

Tryptophanyl tRNA synthetase

lpp1399

tRNA aminoacylation

1215

aspS

Aspartyl-tRNA synthetase

lpp1434

tRNA aminoacylation

1782

ruvB

Holliday junction DNA helicase

lpp1534

tRNA aminoacylation

1011

nrdA

Ribonucleoside-diphosphate reductase, alpha subunit

lpp1738

Deoxyribonucleotide/ribonucleoside metabolism

2829

recA

Bacterial DNA recombination protein

lpp1765

DNA replication, recombination, and repair

1047

tig

Trigger factor

lpp1830

Protein folding and stabilization

1332

lepA

GTP-binding membrane protein

lpp1837

Translation

1833

metK

S-adenosylmethionine synthetase

lpp2004

tRNA aminoacylation

1149

dnaJ

Heat shock protein

lpp2006

Protein folding and stabilization

1140

argS

Arginyl tRNA synthetase

lpp2013

tRNA aminoacylation

1770

eno

Enolase

lpp2020

Glycolysis/gluconeogenesis

1269

ftsZ

Cell division protein

lpp2662

Cell division

1197

uvrC

Excinuclease ABC, subunit C

lpp2698

DNA replication, recombination, and repair

1857

dnaX

DNA polymerase III, subunits gamma and tau

lpp2802

DNA replication, recombination, and repair

1671

recN

DNA repair protein

lpp2877

DNA replication, recombination, and repair

1668

metG

Methionyl tRNA synthetase

lpp2941

tRNA aminoacylation

2013

rho

Transcription terminator factor

lpp3002

Translation

1262

atpD

ATP synthase F1, subunit beta

lpp3053

ATP-proton motive force interconversion

1377

atpA

ATP synthase, subunit alpha

lpp3055

ATP-proton motive force interconversion

1554

thdF

GTP binding protein, thiophene oxidation

lpp3073

tRNA and rRNA base modification

1341

a with respect to strain Paris, nts nucleotides

<p>Additional file 4</p>

Table S4: Summary of genetic diversity parameters for the 31 selected L. pneumophila genes used to establish the phylogeny.

Click here for file

<p>Additional file 5</p>

Table S5: Results for the SH Test of alternative topologies for the 6 analyzed L. pneumophila strains.

Click here for file

With the aim to explore whether recombination events are present in the selected genes we undertook an in depth analysis using the program RDP 68 . Indeed, the analysis of individual genes identified intragenic recombination in 9 of the 31 genes (Table 8). Numerous additional recombination events were detected with the concatenated alignment of the 22 genes for which no intragenic recombination had been shown (Table 8). To minimize false positive recombination events only those that were supported by at least two of the six methods used in RDP were taken into account. However, except one, all were supported by at least three methods. No artifacts resulting of positive selection should be included in this analysis since all of the genes are either informational or operational (housekeeping). Most interestingly, four of the genes in which intragenic recombination was detected are housekeeping genes (pgk, atpD, ffh, metK). Housekeeping genes allow to estimate the extent of recombination within bacterial species since presence of recombination in such "normally recombination free genes" is indicative of a high rate of recombination 22 . Similarly antigen-coding genes of Legionella were reported to show recombination events 18 69 and certain other genomic regions 17 19 70 71 72 . Another example of intragenic recombination in L. pneumophila is the rtxA gene that contains a long tandem repeated domain of variable copy number and sequence 4 10 73 . rtxA of strain Lorraine and Corby share the same repeats, whereas the other strains have unique types of repeats. However, when including the newly sequenced strains Lorraine and HL 0604 1035 we found that repeats of the same type are shared by HL 0604 1035 and Philadelphia and by Lorraine and Lens (Figure 4 and Additional file 6, Table S6), further substantiating high intragenic recombination among strains.

<p>Table 8</p>

Intragenic and intergenic recombination in six L. pneumophila genomes predicted on individual genes and on combined data using six different methods

Detection Method


Data set

Event Number

Putative recombinant sequences

RDP

GENECONV

Boot

scan

Max

chi

Chimaera

SiSscan


metG

1

Lorraine, Lens

NS

NS

NS

Yes

Yes

Yes

dnaX

1

Philadelphia

NS

NS

Yes

Yes

Yes

Yes

2

Lens, Lorraine

NS

NS

NS

Yes

Yes

Yes

proS

1

HL06041035

Yes

Yes

Yes

Yes

Yes

Yes

2

Philadelphia

NS

Yes

NS

Yes

Yes

Yes

cysS

1

Philadelphia

NS

NS

NS

Yes

Yes

NS

lig

1

Lorraine

NS

Yes

Yes

NS

NS

NS

uvrC

1

Lens,Philadelphia, Lorraine

NS

NS

NS

Yes

Yes

Yes

flh

1

Lens

NS

NS

Yes

Yes

Yes

Yes

2

Paris, HL06041035

NS

NS

NS

Yes

Yes

Yes

pgk

1

Lens

NS

NS

NS

Yes

Yes

Yes

atpD

1

Corby

NS

NS

NS

Yes

Yes

Yes


Concatenated

1

Philadelphia

Yes

Yes

Yes

Yes

Yes

Yes

2

Philadelphia

Yes

Yes

Yes

Yes

Yes

Yes

3

HL06041035

Yes

Yes

Yes

Yes

Yes

Yes

4

HL06041035

Yes

Yes

Yes

Yes

Yes

Yes

5

Philadelphia, Corby, Lorraine

Yes

Yes

Yes

Yes

Yes

Yes

6

Lens

Yes

Yes

Yes

Yes

Yes

Yes

7

Paris, HL06041035

Yes

NS

NS

Yes

Yes

NS

8

Paris

Yes

Yes

Yes

Yes

Yes

Yes

9

Lens

Yes

Yes

NS

Yes

Yes

Yes

10

Lens

Yes

Yes

Yes

Yes

Yes

NS

11

HL06041035

Yes

Yes

NS

Yes

NS

NS

12

Paris, HL06041035

Yes

Yes

Yes

Yes

Yes

Yes

13

HL06041035, Lens

NS

Yes

NS

Yes

Yes

Yes

14

Lens, Lorraine

Yes

NS

NS

Yes

Yes

NS

15

Paris, HL06041035

Yes

Yes

NS

Yes

Yes

NS

16

Corby

Yes

NS

NS

Yes

Yes

NS

17

Lens

NS

Yes

NS

Yes

Yes

NS

18

HL06041035, Paris

Yes

NS

NS

Yes

Yes

Yes

19

Corby

Yes

NS

NS

Yes

Yes

NS

20

Lorraine

Yes

Yes

NS

NS

NS

Yes

21

Lens

Yes

NS

Yes

NS

NS

Yes

22

Corby

Yes

NS

Yes

Yes

NS

Yes

23

Lens

NS

Yes

NS

Yes

NS

NS

24

Lens

NS

Yes

NS

Yes

NS

Yes

25

Philadelphia

Yes

NS

NS

Yes

Yes

Yes

NS = non significant result. Yes = significant result with p-value ≤0.05 (where P is the highest acceptable probability value of recombination occurrence).

<p>Figure 4</p>

Schematic representation of the repeat regions present in the rtxA gene of L. pneumophila

Schematic representation of the repeat regions present in the rtxA gene of L. pneumophila. Colored squares represent repeated sequences where the same color corresponds to the same type of repeat. Discontinues lines indicate that the exact number of repeats has not been defined.

<p>Additional file 6</p>

Table S6: Conserved domains and repeats of the rtxA gene in 8 L. pneumophila strains.

Click here for file

To reconstruct the phylogenetic history of the species L. pneumophila we used thus the concatenated alignment of the 31 genes described above. It gave a topology with high bootstrap support, however recombination bias may result in high support for the wrong tree. To avoid possible bias we thus analyzed the concatenated alignment of the 31 genes using a split tree decomposition that allows a more realistic representation of the phylogenetic relationships. Furthermore we constructed a classical bifurcating tree using the highest possible number of genes [all orthologs among the six strains with (1867 genes) and without (2434 genes) L. longbeachae as outgroup]. As shown in Figure 5 the Splits Decomposition phylogeny is network-like suggesting incompatible partitions within sequence data, which commonly arise from recombination. Although the phylogeny based on the orthologous genes can also be affected by recombination, the high number of informative sites included in this data set, should allow recovering the correct history of the species as it has been shown previously for other closely related bacterial species 74 .

<p>Figure 5</p>

Phylogenetic relationships of the 6 L. pneumophila strains analyzed

Phylogenetic relationships of the 6 L. pneumophila strains analyzed. a) Neighbor-net constructed from a concatenation of 31 genes from 6 L. pneumophila strains under a GTR model, with associated bootstrap values. b) Likelihood tree topology of L. pneumophila strains and the outgroup L. longbeachae based on orthologous genes present in all strains/species concatenated.

Taken together, in contrast to previous studies, which reported that the species L. pneumophila is a clonal population 13 14 our results show clearly that a high recombination rate shapes the L. pneumophila genomes. This finding is in line with the natural competence of L. pneumophila. However, some worldwide distributed L. pneumophila clones have been described (e.g. 10 ), suggesting that L. pneumophila is able to develop a unique genetic population structure within a particular region or environment as reported recently 72 .

Recombination of large chromosomal regions of over 200 kbs among L. pneumophila strains

Our recombination analysis revealed not only intragenic recombination events but also intergenic recombination as recombination was detected when using the entire alignment even with only recombination free genes (Table 8). This finding may be explained by the recombination of fragments encompassing several genes or multiple recombination events involving smaller tracts along the genome. To test this hypothesis we used a method recently developed for the analysis of Streptococcus agalactiae genomes 75 . In order to identify patterns of recombination, nucleotide substitutions between strains were counted in sliding windows across the previously defined core chromosome representing 15 possible pair wise comparisons. Each pair wise comparison revealed highly conserved regions (<0.05% polymorphism on average) and less-conserved regions (>0.7% polymorphism), suggesting the occurrence of recombinational exchanges. When analyzing the different strains in depth we identified in each genome several regions with very low polymorphisms (below 0.05%) suggesting that DNA exchange of these fragments has occurred between the different L. pneumophila strains. Most interestingly, the two French strains Paris and HL 0604 1035 that are present since several years in France show 15 regions of a size between 10 and 99 kbs that have very low polymorphism and thus seem to have been exchanged between them (Additional file 7, Figure S1). In contrast when comparing strain Lens with the other 5 genomes analyzed here, very few regions with low polymorphism, two with strain HL 0604 1035 and one with strain Lorraine, were detected. Furthermore, no DNA exchanges seem to have occurred with strains Corby, Philadelphia or Paris. This indicates that strains that are frequent in the same environment (e.g. strain Paris and HL 0604 1035) show high rates of DNA exchange probably by conjugation as suggested for Streptococcus agalactiae 75 and Enterococcus fecalis 76 . In contrast strain Lens, which has been identified to date only twice, in Lens (France) and in Germany, very few DNA transfers with the studied L. pneumophila strains seem to have taken place. Furthermore, some regions may be transferred also between several strains. Figure 6 shows the distribution of single-nucleotide polymorphisms (SNPs) along 330 kb of the genome of L. pneumophila HL 0604 1035, Philadelphia and Lorraine as compared to the same region in the genome of strain Paris. We identified a region of 213 kbs a SNP frequency of 0.005%. Except an indel of 158 bs that shows higher polymorphism, only 11 SNPs are present in this region. This fragment may have evolved by conjugative transfer and recombination between strains Philadelphia and Paris. Among others, this region carries the genes necessary for lipopolysaccaride biosynthesis, that are also part of the smaller fragment that has been exchanged with strain HL 0604 1035. Our analyses suggest, that in addition to frequent intragenic recombination also recombination and horizontal transfer of large chromosomal fragments is taking place and shapes the chromosomes of L. pneumophila.

<p>Additional file 7</p>

Figure S1 - Distribution of single-nucleotide polymorphisms (SNPs) along the genome of L. pneumophila HL 0604 1035 as compared to strains Lens, Philadelphia, Corby and Lorraine. The number of SNPs (y axis) is plotted according to the position of the corresponding 500 bp fragment on the strain Paris chromosome (x axis). A straight blue line indicates 0 polymorphism between the two strains. Numbers on the scale bar indicate the percentage of polymorphism. Yellow blocks indicate chromosomal regions with a SNP number lower than 0,005%.

Click here for file

<p>Figure 6</p>

Distribution of single-nucleotide polymorphisms (SNPs) along 330 kb of the genomes of L. pneumophila HL 0604 1035, Philadelphia and Lorraine

Distribution of single-nucleotide polymorphisms (SNPs) along 330 kb of the genomes of L. pneumophila HL 0604 1035, Philadelphia and Lorraine. The number of SNPs (y axis) is plotted according to the position of the corresponding 500 bp fragment on the strain Paris chromosome (x axis). A straight blue line indicates 0 polymorphism between the two strains. Numbers on the scale bar indicate the percentage of polymorphism. The green (+ strand) and red (- strand) lines depict the corresponding genes.

Conclusion

Analysis of the genome sequences of six L. pneumophila strains shows that the genomes of this environmental pathogen evolve by frequent HGT and high recombination rates. Most interestingly, these events take place between eukaryotes and prokaryotes and among different strains and species of Legionella. A genome-wide map analysis of nucleotide polymorphisms among these six strains demonstrated that each chromosome is a mosaic of large chromosomal fragments from different origins suggesting that exchanges of large DNA regions of over 200 kb have contributed to the genome dynamics in the natural population. The many T4SS might be implicated in exchange of these fragments by conjugal transfer. Plasmids also play a role in genome diversification and are exchanged among strains and circulate even between different species of Legionella. Importantly, plasmids seem to excise and integrate into the genome probably depending on environmental cues. However, L. pneumophila encodes also several toxin anti-toxin that might help to stabilize certain mobile genetic elements. In the near future, the analyses of 100 s of genomes thanks to new generation sequencing combined with molecular studies should provide further clues about the genetic mechanisms and the evolutionary forces that shape the Legionella genomes.

Methods

Bacterial strains and sequence accession numbers

The strains sequenced in this study are L. pneumophila strain Lorraine [EMBL: FQ958210, EMBL:FQ958212] and L. pneumophila HL 0604 1035 [EMBL:FQ958211]. Strain Lorraine was isolated in 2004 from a patient and was recently described as a newly emerging endemic clone 26 . L. pneumophila strain HL 0604 1035 (ST 734, Bellingham subgroup of the Dresden panel) was isolated in 2006 from a water supply system in a French hospital that it is colonizing since more than 10 years.

Sequencing and assembly

The complete genome sequence of L. pneumophila subsp. pneumophila strain HL06041035 (A) and strain Lorraine (B) were determined using a Sanger/pyrosequencing hybrid approach. A shotgun library was constructed with 10kb size fragments, obtained after mechanical shearing of the total genomic DNA, and cloned into vector pCNS (pSU derived). Sequencing with vector-based primers was carried out using the ABI 3730 Applera Sequencer. A total of 20736 (A) and 21888 (B) reads (~4 fold-coverage) were analyzed and assembled with 502731 (A) and 555541 (B) reads (~15 fold-coverage) obtained with Genome Sequencer GS20 (Roche Applied Science). For the assembly, we used the Arachne "HybridAssemble" version (Broad Institute, http://www.broad.mit.edu) that combines the contigs obtained with 454 sequencing with Sanger reads. To validate the assembly, the Mekano interface (Genoscope), based on visualization of clone links inside and between contigs, was used to check the clone coverage and misassemblies. In addition, the consensus was confirmed using Consed functionalities http://www.phrap.org: the consensus quality and the high quality discrepancies. The finishing step was achieved by PCR, primer walking and in vitro transposition technology (Template Generation System™ II Kit; Finnzyme, Espoo, Finland), and a total of 930 (A) and 999 (B) sequences (109, 165 and 656 respectively for L. pneumophila subsp. pneumophila strain HL06041035 and 62, 204 and 733 respectively for L pneumophila subsp. pneumophila str. Lorraine) were needed for gap closure and quality assessment.

Sequence analysis and annotation

The two newly sequenced L. pneumophila genomes were integrated into the MicroScope platform 77 to perform automatic and expert annotation of the genes, and comparative analysis with the other L. pneumophila strains already published. In addition the annotations of the previously published genomes were updated. The system integrates, for each predicted gene, the results of multiple bioinformatics methods (Blast result on UniProt and specialized genomic data, InterPro, COG, PRIAM, synteny group computation using the complete bacterial genomes available at NCBI RefSeq, etc; more information on the syntaxic and functional annotation process is given in 78 ). In addition, many genomic and metabolic comparative tools are also available 77 . For details see https://www.genoscope.cns.fr/agc/microscope/home/index.php.

Definition of orthologous genes

To define orthologous chromosomal genes among the different L. pneumophila strains, pseudogenes and mobile elements were not taken into account due to the difficulty of ortholog assignment for these genes. Putative orthologous relations were defined as gene couples fulfilling two criteria: (i) having a bidirectional best hit (BBH) with an alignment threshold of 55% identity over at least 60% of the query sequence and target size (ii) and being in synteny. Subsequently, putative genes without any orthologous relation due to reduced identity percentage were integrated in a pre-existing orthologue group if they were flanked by orthologous genes showing gene order conservation (microsynteny). A final step of manual curation was carried out for each doubtful case.

Sequence alignments

For each gene of the selected data set, the nucleotide sequence was aligned based on the amino acid sequence using tranalign/EMBOSS package http://emboss.sourceforge.net/. Subsequently genes were concatenated in different data sets.

Identification of eukaryotic like proteins and eukaryotic domain carrying proteins

Eukaryotic domains were identified by analyzing the results obtained for all genes using the Interpro database that is integrated in MAGE. For the identification of eukaryotic like proteins we developed a new method. First we constructed two databases, one containing all and only eukaryotic sequences retrieved from public databases and a second one containing all and only prokaryotic sequences. From the second database we excluded the proteins of bacterial genera for which eukaryotic like protein-domains have been found in high proportions (e.g. parasites of protozoa) or bacterial genera that are reported to establish a symbiotic relationship with amoeba (for a detailed list see Additional file 8, Table S7). Those proteins, that showed a better, normalized blast score against eukaryotic proteins than to those present in the prokaryotic database were retrieved as eukaryotic like proteins. Parameters established for blast were: minimum identity: 25%; minimum ratio avec query: 60%; minimum ratio avec target: 50%. The final results were manually checked.

<p>Additional file 8</p>

Tables S7 - List of bacterial genera removed from our prokaryotic database.

Click here for file

Phylogenetic Analysis

For phylogentic reconstruction of the L. pneumophila strains analyzed in this work several data sets were used: (i) 31 housekeeping genes described to be essential for all prokaryotes were selected based on the study of Zeigler 67 (Table 7 and Additional file 9, Figure S2) for a multi locus sequence analysis (MLSA) approach for which gene each was analyzed individually and as a concatenated alignment, (ii) a concatenated alignment of 2434 orthologous genes present in all analyzed L. pneumophila strains (iii) a concatenated alignment of 1867 orthologous genes present in all analyzed L. pneumophila strains and in the selected out group, Legionella longbeachae strain NSW150. An analysis of genetic divergence was performed using DNAsp vs 5.00.07 79 using the 31 selected housekeeping genes. For phylogenetic reconstruction maximum likelihood (ML) methods were used to infer phylogenetic relationships for all data sets. Prior to ML analyses, a DNA substitution model for each gene or data set was selected using Modeltest v3.06 80 and the Akaike information criterion. ML heuristic searches were performed using 500 random taxon-addition replicates with tree bisection and reconnection (TBR) and branch swapping. ML bootstrap support was determined using 1000 bootstrap replicates. The ML best trees were rooted on L. longbeachae when added. A network reconstruction was done for the same data set (i) using SplitsTree4 (version 4.10) 81 . The NeighborNet method and the GTR distance model were used to create the network.

<p>Additional file 9</p>

Figure S2: Distribution of the 31 genes selected for establishing the phylogeny of L. pneumophila species. The coordinates are given with respect to the chromosome of L. pneumophila strain Paris. Numbers next to gene names indicate the first position of the corresponding gene starting from the origin of replication.

Click here for file

Congruence test

The 31 genes selected for a MLST approach were tested for the significance of topological differences in the obtained phylogenetic trees using several methods. The first approach was based on the consensus of individual gene trees. The consensus tree was inferred using the CONSENSE program in the PHYLIP package http://evolution.genetics.washington.edu/phylip.html applying the extended majority rule. Secondly we tested the significance of topological differences in phylogenetic trees using the Shimodaira-Hasegawa (SH) test. The SH test compares the likelihood score (-lnL) of a given data set across its ML tree versus the -lnL of that data set across alternative topologies, which in this case are the ML phylogenies for other data sets. The differences in the -lnL values are evaluated for statistical significance using 1000 replicates based on resampling estimated with the log-likelihood (RELL) method (PAUP version 4.0b10; http://paup.csit.fsu.edu/. We applied the test using all the trees obtained with individual genes, with the concatenated alignment against the alignment of each individual gene and with the alignment of all the 31 genes concatenated.

Recombination analysis

The 31 genes selected for a MLST approach and its corresponding concatenated alignment, were screened for the presence of putative recombination events by using RDP 2.0b08 82 . This program identifies recombinant sequences and recombination breakpoints applying several methods. We selected six of them; two phylogenetic methods (which infer recombination when different parts of the genome result in discordant topologies): RDP 68 , 2000) and Bootscanning 83 ; and four nucleotide substitution methods (which examine the sequences either for a significant clustering of substitutions or for a fit to an expected statistical distribution): Maxchi and Chimaera 84 , GeneConv 85 and Sis-scan 86 . We considered only those recombination events in our analysis that were identified by at least two methods. The common settings for all methods were (i) to consider sequences as circular, (ii) a statistical significance of P < 0.05, and (iii) a Bonferroni correction for multiple comparisons implemented in RDP.

Abbreviations

ANK: ankyrin motif; CRISPR: Clustered regularly interspaced short palindromic repeats; HGT: horizontal gene transfer; ML: maximum likelihood; nt: nucleotide; Sg1: serogroup 1; T4SS: Type IV secretion system; T2SS: Type II secretion system;

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

LGV and CB designed the study. SJ and JE supplied material and expertise; VB and BV performed genome sequencing; LGV and CR performed the genome annotation and analysis work, CM and RZ set up the LegioScope database. LGV and CB drafted and wrote the manuscript. All authors contributed to and approved the final manuscript.

Acknowledgements

This work received financial support from the Institut Pasteur, the Centre National de la Recherche (CNRS), the Institut Carnot-Pasteur MI and from the ANR-10-PATH-004 project, in the frame of ERA-Net PathoGenoMics. The MicroScope platform got financial support from GIS IBiSA. L. Gomez-Valero was holder of a Roux postdoctoral research Fellowship financed by the Institut Pasteur and subsequently with support from the Fondation pour la Recherche Médicale (FRM). We would like to particularly thank Philippe Glaser for stimulating discussions and critical commenting of the article and Cyril Firmo for helping with the recombination analysis.

<p>Ecological distribution of <it>Legionella pneumophila</it></p>FliermansCBCherryWBOrrisonLHSmithSJTisonDLPopeDHAppl Environ Microbiol19814119162436337013702<p>The molecular ecology of <it>Legionellae</it></p>FieldsBSTrends Microbiol19964728629010.1016/0966-842X(96)10041-X8829338<p>Preliminary report on the pathogenicity of <it>Legionella pneumophila </it>for freshwater and soil amoebae</p>RowbothamTJJ Clin Pathol198033121179118310.1136/jcp.33.12.117911463717451664<p>Evidence in the <it>Legionella pneumophila </it>genome for exploitation of host cell functions and high genome plasticity</p>CazaletCRusniokCBruggemannHZidaneNMagnierAMaLTichitMJarraudSBouchierCVandeneschFKunstFEtienneJGlaserPBuchrieserCNat Genet200436111165117310.1038/ng144715467720<p>Modulation of host cell function by <it>Legionella pneumophila </it>type IV effectors</p>HubberARoyCRAnnu Rev Cell Dev Biol20102626128310.1146/annurev-cellbio-100109-10403420929312<p>Molecular mimicry: an important virulence strategy employed by <it>Legionella pneumophila </it>to subvert host functions</p>NoraTLommaMGomez-ValeroLBuchrieserCFuture Microbiol2009469170110.2217/fmb.09.4719659425<p>Evidence for acquisition of <it>Legionella </it>type IV secretion substrates via interdomain horizontal gene transfer</p>de FelipeKSPampouSJovanovicOSPericoneCDYeSFKalachikovSShumanHAJ Bacteriol2005187227716772610.1128/JB.187.22.7716-7726.2005128029916267296<p>The origins of eukaryotic-like proteins in <it>Legionella pneumophila</it></p>Lurie-WeinbergerMNGomez-ValeroLMeraultNGlocknerGBuchrieserCGophnaUInt J Med Microbiol2010300747048110.1016/j.ijmm.2010.04.01620537944<p>A 65-Kilobase Pathogenicity Island Is Unique to Philadelphia-1 Strains of <it>Legionella pneumophila</it></p>BrassingaAKHiltzMFSissonGRMorashMGHillNGardunoEEdelsteinPHGardunoRAHoffmanPSJ Bacteriol2003185154630363710.1128/JB.185.15.4630-4637.200316578012867476<p>Multigenome analysis identifies a worldwide distributed epidemic <it>Legionella pneumophila </it>clone that emerged within a highly diverse species</p>CazaletCJarraudSGhavi-HelmYKunstFGlaserPEtienneJBuchrieserCGenome Res200818343144110.1101/gr.7229808225910718256241<p>Regulation of hypercompetence in <it>Legionella pneumophila</it></p>SextonJAVogelJPJ Bacteriol2004186123814382510.1128/JB.186.12.3814-3825.200441997115175295<p>Natural competence for DNA transformation by <it>Legionella pneumophila </it>and its association with expression of type IV pili</p>StoneBJKwaikYAJ Bacteriol19991815139514029352610049368<p>Genetic structure of populations of <it>Legionella pneumophila</it></p>SelanderRKMcKinneyRMWhittamTSBibbWFBrennerDJNolteFSPattisonPEJ Bacteriol19851633102110372192344030689<p>Clonal population structure of <it>Legionella pneumophila </it>inferred from allelic profiling</p>EdwardsMTFryNKHarrisonTGMicrobiology2008154Pt 385286418310031<p>Genetic variability in environmental isolates of <it>Legionella pneumophila </it>from Comunidad Valenciana (Spain)</p>CoscollaMGosalbesMJCatalanVGonzalez-CandelasFEnviron Microbiol2006861056106310.1111/j.1462-2920.2006.00997.x16689726<p>Sequence polymorphism of <it>dotA </it>and <it>mip </it>alleles mediating invasion and intracellular replication of <it>Legionella pneumophila</it></p>BumbaughACMcGrawEAPageKLSelanderRKWhittamTSCurr Microbiol200244531432211927981<p>Molecular evolution of the dotA gene in <it>Legionella pneumophila</it></p>KoKSHongSKLeeHKParkMYKookYHJ Bacteriol2003185216269627710.1128/JB.185.21.6269-6277.200321940014563861<p>Population genetic structure of <it>Legionella pneumophila </it>inferred from RNA polymerase gene (<it>rpoB</it>) and DotA gene (<it>dotA</it>) sequences</p>KoKSLeeHKParkMYParkMSLeeKHWooSYYunYJKookYHJ Bacteriol200218482123213010.1128/JB.184.8.2123-2130.200213495911914343<p>Population structure and recombination in environmental isolates of <it>Legionella pneumophila</it></p>CoscollaMGonzalez-CandelasFEnviron Microbiol20079364365610.1111/j.1462-2920.2006.01184.x17298365<p>A comparison of homologous recombination rates in bacteria and archaea</p>VosMDidelotXISME J20093219920810.1038/ismej.2008.9318830278<p>Quantifying nonvertical inheritance in the evolution of <it>Legionella pneumophila</it></p>CoscollaMComasIGonzalez-CandelasFMol Biol Evol2011282985100110.1093/molbev/msq27820961962<p>Recombination and the population structures of bacterial pathogens</p>FeilEJSprattBGAnnu Rev Microbiol20015556159010.1146/annurev.micro.55.1.56111544367<p>Comparison of clinical and environmental isolates of <it>Legionella pneumophila </it>obtained in the UK over 19 years</p>HarrisonTGDoshiNFryNKJosephCAClin Microbiol Infect2007131788510.1111/j.1469-0691.2006.01558.x17184291<p><it>Legionella </it>pathogenicity: genome structure, regulatory networks and the host cell response</p>SteinertMHeunerKBuchrieserCAlbert-WeissenbergerCGlocknerGInt J Med Microbiol20072977-857758710.1016/j.ijmm.2007.03.00917467337<p>The genomic sequence of the accidental pathogen <it>Legionella pneumophila</it></p>ChienMMorozovaIShiSShengHChenJGomezSMAsamaniGHillKNuaraJFederMRineerJGreenbergJJSteshenkoVParkSHZhaoBTeplitskayaEEdwardsJRPampouSGeorghiouAChouICIannuccilliWUlzMEKimDHGeringer-SamethAGoldsberryCMorozovPFischerSGSegalGQuXRzhetskyAScience200430556921966196810.1126/science.109977615448271<p>Lorraine strain of <it>Legionella pneumophila </it>serogroup 1, France</p>GinevraCForeyFCampeseCReyrolleMCheDEtienneJJarraudSEmerg Infect Dis200814467367510.3201/eid1404.070961257094118394295<p>The genome of the amoeba symbiont "Candidatus <it>Amoebophilus asiaticus</it>" reveals common mechanisms for host cell interaction among amoeba-associated bacteria</p>Schmitz-EsserSTischlerPArnoldRMontanaroJWagnerMRatteiTHornMJ Bacteriol201019241045105710.1128/JB.01379-09281295820023027<p>Analysis of the <it>Legionella longbeachae </it>genome and transcriptome uncovers unique strategies to cause Legionnaires' disease</p>CazaletCGomez-ValeroLRusniokCLommaMDervins-RavaultDNewtonHJSansomFMJarraudSZidaneNMaL BouchierCEtienneJHartlandELBuchrieserCPLoS Genet201062e100085110.1371/journal.pgen.1000851282474720174605<p>InterPro: the integrative protein signature database</p>HunterSApweilerRAttwoodTKBairochABatemanABinnsDBorkPDasUDaughertyLDuquenneLFinnRDGoughJHaftDHuloNKahnDKellyELaugraudALetunicILonsdaleDLopezRMaderaMMaslenJMcAnullaCMcDowallJMistryJMitchellAMulderNNataleDOrengoCQuinnAFNucleic Acids Res2009D211215<p>The spectrin repeat: a structural platform for cytoskeletal protein assemblies</p>Djinovic-CarugoKGautelMYlänneJYoungPFEBS Lett2002513111912310.1016/S0014-5793(01)03304-X11911890<p><it>Legionella </it>eukaryotic-like type IV substrates interfere with organelle trafficking</p>de FelipeKSGloverRTCharpentierXAndersonORReyesMPericoneCDShumanHAPLoS Pathog200848e100011710.1371/journal.ppat.1000117247551118670632<p>Large-scale identification of <it>Legionella pneumophila </it>Dot/Icm substrates that modulate host cell vesicle trafficking pathways</p>HeidtmanMChenEJMoyMYIsbergRRCell Microbiol200911223024810.1111/j.1462-5822.2008.01249.x274495519016775<p>The <it>Legionella pneumophila </it>replication vacuole: making a cosy niche inside host cells</p>IsbergRRO'ConnorTJHeidtmanMNat Rev Microbiol200971132410.1038/nrmicro1967263140219011659<p>Structure of small G proteins and their regulators</p>PaduchMJeleńFOtlewskiJActa Biochim Pol200148882985011995995<p>Rab GEFs and GAPs</p>BarrFLambrightDGCurr Opin Cell Biol201022446147010.1016/j.ceb.2010.04.007292965720466531<p>A <it>Legionella pneumophila </it>Effector Protein Encoded in a Region of Genomic Plasticity Binds to Dot/Icm-Modified Vacuoles</p>NinioSCelliJRoyCRPLoS Pathog200951e100027810.1371/journal.ppat.1000278262134919165328<p>Two distinct defects in intracellular growth complemented by a single genetic locus in <it>Legionella pneumophila</it></p>BergerKHIsbergRRMol Microbiol19937171910.1111/j.1365-2958.1993.tb01092.x8382332<p>Characterization of a new region required for macrophage killing by <it>Legionella pneumophila</it></p>SegalGShumanHAInfect Immun19976512505750661757299393796<p>Comparative sequence analysis of the <it>icm/dot </it>genes in <it>Legionella</it></p>MorozovaIQuXShiSAsamaniGGreenbergJEShumanHARussoJJPlasmid200451212714710.1016/j.plasmid.2003.12.00415003709<p>A bacterial guanine nucleotide exchange factor activates ARF on <it>Legionella </it>phagosomes</p>NagaiHKaganJCZhuXKahnRARoyCRScience2002295555567968210.1126/science.106702511809974<p>Genome-scale identification of <it>Legionella pneumophila </it>effectors using a machine learning approach</p>BursteinDZusmanTDegtyarEVinerRSegalGPupkoTPLoS Pathog200957e100050810.1371/journal.ppat.1000508270160819593377<p>A yeast genetic system for the identification and characterization of substrate proteins transferred into host cells by the <it>Legionella pneumophila </it>Dot/Icm system</p>CampodonicoEMChesnelLRoyCRMol Microbiol200556491893310.1111/j.1365-2958.2005.04595.x15853880<p>Pathogen effector protein screening in yeast identifies <it>Legionella </it>factors that interfere with membrane trafficking</p>ShohdyNEfeJAEmrSDShumanHAProc Natl Acad Sci USA2005102134866487110.1073/pnas.050131510255570915781869<p>Comprehensive Identification of Protein Substrates of the Dot/Icm Type IV Transporter of <it>Legionella pneumophila</it></p>ZhuWBangaSTanYZhengCStephensonRGatelyJLuoZQPLoS One201163e1763810.1371/journal.pone.0017638305236021408005<p>Modulation of ubiquitin dynamics and suppression of DALIS formation by the <it>Legionella pneumophila </it>Dot/Icm system</p>IvanovSSRoyCRCell Microbiol200911226127810.1111/j.1462-5822.2008.01251.x285606819016782<p>Exploitation of conserved eukaryotic host cell farnesylation machinery by an F-box effector of <it>Legionella pneumophila</it></p>PriceCTAl-QuadanTSanticMJonesSCAbu KwaikYJ Exp Med201020781713172610.1084/jem.20100771291613120660614<p>Type IV secretion systems: tools of bacterial horizontal gene transfer and virulence</p>JuhasMCrookDWHoodDWCell Microbiol200810122377238610.1111/j.1462-5822.2008.01187.x268867318549454<p>Biogenesis, architecture, and function of bacterial type IV secretion systems</p>ChristiePJAtmakuriKKrishnamoorthyVJakubowskiSCascalesEAnnu Rev Microbiol200559451-485<p>Type IV secretion systems and their effectors in bacterial pathogenesis</p>BackertSmeyerTFCurr Opin Microbiol20069220721710.1016/j.mib.2006.02.00816529981<p>Analysis of the sequence and gene products of the transfer region of the F sex factor</p>FrostLSIppen-IhlerKSkurrayRA1994<p>Genome sequence of <it>Rickettsia bellii </it>illuminates the role of amoebae in gene exchanges between intracellular pathogens</p>OgataHLa ScolaBAudicSRenestoPBlancGRobertCFournierPEClaverieJMRaoultDPLoS Genet200625e:76<p>Growth-phase-dependent mobility of the <it>lvh</it>-encoding region in <it>Legionella pneumophila </it>strain Paris</p>Doleans-JordheimAAkermiMGinevraCCazaletCKayESchneiderDBuchrieserCAtlanDVandeneschFEtienneJJarraudSMicrobiology2006152Pt 123561356817159208<p>F factor conjugation is a true type IV secretion system</p>LawleyTDKlimkeWAGubbinsMJFrostLSFEMS Microbiol Lett2003224111510.1016/S0378-1097(03)00430-012855161<p>Identification and characterization of a new conjugation/type IVA secretion system (trb/tra) of <it>Legionella pneumophila </it>Corby localized on a mobile genomic island</p>GlöcknerGAlbert-WeissenbergerCWeinmannEJacobiSSchunderESteinertMHackerJHeunerKInt J Med Microbiol20072985-641142817888731<p><it>Legionella pneumophila </it>CsrA is a pivotal repressor of transmission traits and activator of replication</p>MolofskyABSwansonMSMol Microbiol200350244546110.1046/j.1365-2958.2003.03706.x14617170<p>CRISPR--a widespread system that provides acquired resistance against phages in bacteria and archaea</p>SorekRKuninVHugenholtzPNat Rev Microbiol20086318118610.1038/nrmicro179318157154<p>Toxin-antitoxin systems: why so many, what for?</p>Van MelderenLCurr Opin Microbiol201013678178510.1016/j.mib.2010.10.00621041110<p>Two <it>higBA </it>loci in the <it>Vibrio cholerae </it>superintegron encode mRNA cleaving enzymes and can stabilize plasmids</p>Christensen-DalsgaardMGerdesKMol Microbiol200662239741110.1111/j.1365-2958.2006.05385.x17020579<p>Chromosomal toxin-antitoxin loci can diminish large-scale genome reductions in the absence of selection</p>SzekeresSDautiMWildeCMazelDRowe-MagnusDAMol Microbiol20076361588160510.1111/j.1365-2958.2007.05613.x17367382<p>RNA in Defense: CRISPRs Protect Prokaryotes against Mobile Genetic Elements</p>JoreMMBrounsSJvan der OostJCold Spring Harb Perspect Biol2011<p><it>Legionella pneumophila </it>pangenome reveals strain-specific virulence factors</p>D'AuriaGJimenez-HernandezNPeris-BondiaFMoyaALatorreABMC Genomics201011118110.1186/1471-2164-11-181285940520236513<p>Evidence for the presence of <it>Legionella </it>bacteriophages in environmental water samples</p>LammertynEVande VoordeJMeyenEMaesLMastJAnnéJMicrob Ecol200856119119710.1007/s00248-007-9325-z17957402<p>Regulatory interactions between the Pho and sigma(B)-dependent general stress regulons of <it>Bacillus subtilis</it></p>PragaiZHarwoodCRMicrobiology2002148Pt 51593160211988534<p>'Candidatus <it>Protochlamydia amoebophila</it>', an endosymbiont of <it>Acanthamoeba </it>spp</p>CollingroAToenshoffERTaylorMWFritscheTRWagnerMHornMInt J Syst Evol Microbiol200555Pt 51863186616166679<p><it>Legionella drancourtii </it>sp. nov., a strictly intracellular amoebal pathogen</p>La ScolaBBirtlesRJGreubGHarrisonTJRatcliffRMRaoultDInt J Syst Evol Microbiol200454Pt 369970315143011<p>Literature review--efficacy of various disinfectants against <it>Legionella </it>in water systems</p>KimBRAndersonJEMuellerSAGainesWAKendallAMWater Res200236184433444410.1016/S0043-1354(02)00188-412418646<p>Gene sequences useful for predicting relatedness of whole genomes in bacteria</p>ZeiglerDRInt J Syst Evol Microbiol200353Pt 61893190014657120<p>RDP: detection of recombination amongst aligned sequences</p>MartinDRybickiEBioinformatics200016656256310.1093/bioinformatics/16.6.56210980155<p>Application of RNA polymerase beta-subunit gene (<it>rpoB</it>) sequences for the molecular differentiation of <it>Legionella </it>species</p>KoKSLeeHKParkMYLeeKHYunYJWooSYMiyamotoHKookYHJ Clin Microbiol20024072653265810.1128/JCM.40.7.2653-2658.200212060712089300<p>Comparison of clinical and environmental samples of <it>Legionella pneumophila </it>at the nucleotide sequence level</p>CoscollaMGonzalez-CandelasFInfect Genet Evol20099588288810.1016/j.meegid.2009.05.01319465160<p>Mosaic structure of pathogenicity islands in <it>Legionella pneumophila</it></p>KoKSLeeHKParkMYKookYHJ Mol Evol2003571637210.1007/s00239-002-2452-812962307<p>Genetic diversity of <it>Legionella pneumophila </it>inferred from <it>rpoB </it>and <it>dotA </it>sequences</p>KoKSMiyamotoHLeeHKParkMYFukudaKParkBJKookYHClin Microbiol Infect200612325426110.1111/j.1469-0691.2005.01338.x16451413<p>Virulence factor rtx in <it>Legionella pneumophila</it>, evidence suggesting it is a modular multifunctional protein</p>D'AuriaGJiménezNPeris-BondiaFPelazCLatorreAMoyaABMC Genomics200891410.1186/1471-2164-9-14225794118194518<p>The effect of recombination on the accuracy of phylogeny estimation</p>PosadaDCrandallKAJ Mol Evol200254339640211847565<p>Shaping a bacterial genome by large chromosomal replacements, the evolutionary history of <it>Streptococcus agalactiae</it></p>BrochetMRusniokCCouveEDramsiSPoyartCTrieu-CuotPKunstFGlaserPProc Natl Acad Sci USA200810541159611596610.1073/pnas.0803654105257295218832470<p>Mechanism of chromosomal transfer of <it>Enterococcus faecalis </it>pathogenicity island, capsule, antimicrobial resistance, and other traits</p>MansonJMHancockLEGilmoreMSProc Natl Acad Sci USA107271226912274<p>MaGe: a microbial genome annotation system supported by synteny results</p>VallenetDLabarreLRouyZBarbeVBocsSCruveillerSLajusAPascalGScarpelliCMedigueCNucleic Acids Res2006341536510.1093/nar/gkj406132623716407324<p>MicroScope: a platform for microbial genome annotation and comparative genomics</p>VallenetDEngelenSMornicoDCruveillerSFleuryLLajusARouyZRocheDSalvignolGScarpelliCMédigueCDatabase (Oxford)2009bap021<p>DnaSP, DNA polymorphism analyses by the coalescent and other methods</p>RozasJSánchez-DelBarrioJCMesseguerXRozasRBioinformatics20031918<p>MODELTEST: testing the model of DNA substitution</p>PosadaDCrandallKABioinformatics199814981781810.1093/bioinformatics/14.9.8179918953<p>Application of phylogenetic networks in evolutionary studies</p>HusonDHBryantDMol Biol Evol200623225426716221896<p>RDP2: recombination detection and analysis from sequence alignments</p>MartinDPWilliamsonCPosadaDBioinformatics200521226026210.1093/bioinformatics/bth49015377507<p>Identification of breakpoints in intergenotypic recombinants of HIV type 1 by bootscanning</p>SalminenMOCarrJKBurkeDSMcCutchanFEAIDS Res Hum Retroviruses199511111423142510.1089/aid.1995.11.14238573403<p>Analyzing the mosaic structure of genes</p>SmithJMJ Mol Evol19923421261291556748<p>A phage single-stranded DNA (ssDNA) binding protein complements ssDNA accumulation of a geminivirus and interferes with viral movement</p>PadidamMBeachyRNFauquetCMJ Virol1999732160916161039869882367<p>Sister-scanning: a Monte Carlo procedure for assessing signals in recombinant sequences</p>GibbsMJArmstrongJSGibbsAJBioinformatics200016757358210.1093/bioinformatics/16.7.57311038328