B. Fields, R. Benson, and R. Besser, Legionella and Legionnaires' Disease: 25 Years of Investigation, Clinical Microbiology Reviews, vol.15, issue.3, pp.506-532, 2002.
DOI : 10.1128/CMR.15.3.506-526.2002

T. Rowbotham, Preliminary report on the pathogenicity of Legionella pneumophila for freshwater and soil amoebae., Journal of Clinical Pathology, vol.33, issue.12, pp.1179-83, 1980.
DOI : 10.1136/jcp.33.12.1179

M. Steinert, U. Hentschel, and J. Hacker, : an aquatic microbe goes astray, FEMS Microbiology Reviews, vol.26, issue.2, pp.149-62, 2002.
DOI : 10.1111/j.1574-6976.2002.tb00607.x

P. Edelstein and . Legionnaires, Disease: History and clinical findings, Legionella: Molecular Microbiology. 1, pp.1-19, 2008.

M. Horwitz and S. Silverstein, Legionnaires' Disease Bacterium (Legionella pneumophila) Multiplies Intracellularly in Human Monocytes, Journal of Clinical Investigation, vol.66, issue.3, pp.441-50, 1980.
DOI : 10.1172/JCI109874

T. Nora, M. Lomma, L. Gomez-valero, and C. Buchrieser, to subvert host functions, Future Microbiology, vol.4, issue.6, pp.691-701, 2009.
DOI : 10.1371/journal.ppat.1000220

B. Byrne and M. Swanson, Expression of Legionella pneumophila virulence traits in response to growth conditions, Infect Immun, vol.66, issue.7, pp.3029-3063, 1998.

A. Molofsky and M. Swanson, Differentiate to thrive: lessons from the Legionella pneumophila life cycle, Molecular Microbiology, vol.184, issue.1, pp.29-40, 2004.
DOI : 10.1128/jb.179.14.4639-4642.1997

H. Bruggemann, A. Hagman, M. Jules, O. Sismeiro, M. Dillies et al., Virulence strategies for infecting phagocytes deduced from the in vivo transcriptional program of Legionella pneumophila, Cellular Microbiology, vol.22, issue.8, pp.1228-1268, 2006.
DOI : 10.1093/nar/30.4.e15

V. Forsbach-birk, T. Mcnealy, C. Shi, D. Lynch, and R. Marre, Reduced expression of the global regulator protein CsrA in Legionella pneumophila affects virulence-associated regulators and growth in Acanthamoeba castellanii, International Journal of Medical Microbiology, vol.294, issue.1, pp.15-25, 2004.
DOI : 10.1016/j.ijmm.2003.12.003

A. Molofsky and M. Swanson, Legionella pneumophila CsrA is a pivotal repressor of transmission traits and activator of replication, Molecular Microbiology, vol.184, issue.2, pp.445-61, 2003.
DOI : 10.1128/jb.178.4.1012-1017.1996

C. Vakulskas, A. Potts, P. Babitzke, B. Ahmer, and T. Romeo, Regulation of Bacterial Virulence by Csr (Rsm) Systems, Microbiology and Molecular Biology Reviews, vol.79, issue.2, pp.193-224, 2015.
DOI : 10.1128/MMBR.00052-14

E. Van-assche, S. Van-puyvelde, J. Vanderleyden, and H. Steenackers, RNA-binding proteins involved in post-transcriptional regulation in bacteria, Frontiers in Microbiology, vol.9, issue.e85626, pp.141-25784899, 2015.
DOI : 10.1016/S1097-2765(01)00437-3

Z. Dalebroux, R. Edwards, and M. Swanson, differentiation in host macrophages, Molecular Microbiology, vol.5, issue.3, pp.640-58, 2008.
DOI : 10.1111/j.1365-2958.2008.06555.x

Z. Dalebroux, B. Yagi, T. Sahr, C. Buchrieser, and M. Swanson, differentiation, Molecular Microbiology, vol.184, issue.5555, pp.200-219, 2010.
DOI : 10.1111/j.1365-2958.2010.07094.x

M. Rasis and G. Segal, The LetA-RsmYZ-CsrA regulatory cascade, together with RpoS and PmrA, posttranscriptionally regulates stationary phase activation of Legionella pneumophila Icm/Dot effectors, Mol Microbiol, vol.72, issue.4, 2009.

T. Sahr, C. Rusniok, D. Dervins-ravault, O. Sismeiro, J. Coppee et al., Deep sequencing defines the transcriptional map of L. pneumophila and identifies growth phase-dependent regulated ncRNAs implicated in virulence, RNA biology, vol.2012, issue.94, pp.503-522
URL : https://hal.archives-ouvertes.fr/pasteur-01338351

P. Fettes, V. Forsbach-birk, D. Lynch, and R. Marre, Overexpresssion of a homologue of the regulator affects cell size, flagellation, and pigmentation, International Journal of Medical Microbiology, vol.291, issue.5, pp.353-60, 2001.
DOI : 10.1078/1438-4221-00141

A. Kessler, U. Schell, T. Sahr, A. Tiaden, C. Harrison et al., orphan sensor kinase LqsT regulates competence and pathogen-host interactions as a component of the LAI-1 circuit, Environmental Microbiology, vol.184, issue.2, pp.646-62, 2013.
DOI : 10.1128/JB.184.1.67-75.2002

URL : https://hal.archives-ouvertes.fr/pasteur-01336729

A. Tiaden, T. Spirig, P. Carranza, H. Bruggemann, K. Riedel et al., Synergistic Contribution of the Legionella pneumophila lqs Genes to Pathogen-Host Interactions, Journal of Bacteriology, vol.190, issue.22, pp.7532-7579, 2008.
DOI : 10.1128/JB.01002-08

A. Tiaden, T. Spirig, T. Sahr, M. Walti, K. Boucke et al., The autoinducer synthase LqsA and putative sensor kinase LqsS regulate phagocyte interactions, extracellular filaments and a genomic island of Legionella pneumophila, Environmental Microbiology, vol.63, issue.5, pp.1243-59, 2010.
DOI : 10.4161/cib.7713

A. Tiaden, T. Spirig, S. Weber, H. Bruggemann, R. Bosshard et al., The Legionella pneumophila response regulator LqsR promotes host cell interactions as an element of the virulence regulatory network controlled by RpoS and LetA, Cellular Microbiology, vol.63, issue.12, pp.2903-2923, 2007.
DOI : 10.1099/00221287-146-6-1345

T. Zusman, G. Aloni, E. Halperin, H. Kotzer, E. Degtyar et al., The response regulator PmrA is a major regulator of the icm/dot type IV secretion system in Legionella pneumophila and Coxiella burnetii, Molecular Microbiology, vol.67, issue.5, pp.1508-1531, 2007.
DOI : 10.1128/IAI.71.7.3714-3723.2003

O. Nevo, T. Zusman, M. Rasis, Z. Lifshitz, and G. Segal, Identification of Legionella pneumophila Effectors Regulated by the LetAS-RsmYZ-CsrA Regulatory Cascade, Many of Which Modulate Vesicular Trafficking, Journal of Bacteriology, vol.196, issue.3, pp.681-92, 2014.
DOI : 10.1128/JB.01175-13

S. Gudapaty, K. Suzuki, X. Wang, P. Babitzke, and T. Romeo, Regulatory interactions of Csr components: the RNA binding protein CsrA activates csrB transcription in Escherichia coli, J Bacteriol, vol.18320, issue.20, pp.6017-276017, 2001.

T. Romeo, M. Gong, M. Liu, and A. Brun-zinkernagel, Identification and molecular characterization of csrA, a pleiotropic gene from Escherichia coli that affects glycogen biosynthesis, gluconeogenesis, cell size, and surface properties., Journal of Bacteriology, vol.175, issue.15, pp.4744-55, 1993.
DOI : 10.1128/jb.175.15.4744-4755.1993

M. Schubert, K. Lapouge, O. Duss, F. Oberstrass, I. Jelesarov et al., Molecular basis of messenger RNA recognition by the specific bacterial repressing clamp RsmA/CsrA, Nature Structural & Molecular Biology, vol.177, issue.9, pp.807-820, 2007.
DOI : 10.1146/annurev.physchem.51.1.129

B. Hammer, E. Tateda, and M. Swanson, A two-component regulator induces the transmission phenotype of stationary-phase Legionella pneumophila, Molecular Microbiology, vol.273, issue.1, pp.107-125, 2002.
DOI : 10.1128/jb.166.1.187-193.1986

C. Albert-weissenberger, T. Sahr, O. Sismeiro, J. Hacker, K. Heuner et al., Control of Flagellar Gene Regulation in Legionella pneumophila and Its Relation to Growth Phase, Journal of Bacteriology, vol.192, issue.2, pp.446-55, 2009.
DOI : 10.1128/JB.00610-09

M. Zucker, Mfold web server for nucleic acid folding and hybridization prediction, Nucleic Acids Research, vol.31, issue.13, pp.3406-3421, 2003.
DOI : 10.1093/nar/gkg595

T. Spirig, A. Tiaden, P. Kiefer, C. Buchrieser, J. Vorholt et al., Autoinducer Synthase LqsA Produces an ??-Hydroxyketone Signaling Molecule, Journal of Biological Chemistry, vol.53, issue.26, pp.18113-18136, 2008.
DOI : 10.1046/j.1365-2958.2003.03477.x

S. Schoebel, A. Cichy, R. Goody, and A. Itzen, Protein LidA from Legionella is a Rab GTPase supereffector, Proceedings of the National Academy of Sciences, vol.60, issue.Pt 12 Pt 1, pp.17945-50, 2011.
DOI : 10.1107/S0907444904019158

E. Campodonico, L. Chesnel, and C. Roy, A yeast genetic system for the identification and characterization of substrate proteins transferred into host cells by the Legionella pneumophila Dot/Icm system, Molecular Microbiology, vol.71, issue.4, pp.918-951, 2005.
DOI : 10.1091/mbc.10.2.435

O. Brien, K. Lindsay, E. Starai, and V. , The Legionella pneumophila effector protein, LegC7, alters yeast endosomal trafficking, ):e0116824. PubMed Central PMCID: PMCPMC4314205, p.25643265, 2015.

M. Ciampi, Rho-dependent terminators and transcription termination, Microbiology, vol.152, issue.9, pp.2515-2543, 2006.
DOI : 10.1099/mic.0.28982-0

URL : http://mic.microbiologyresearch.org/deliver/fulltext/micro/152/9/2515.pdf?itemId=/content/journal/micro/10.1099/mic.0.28982-0&mimeType=pdf&isFastTrackArticle=

N. Figueroa-bossi, A. Schwartz, B. Guillemardet, D. Heygere, F. Bossi et al., RNA remodeling by bacterial global regulator CsrA promotes Rho-dependent transcription termination, Genes & Development, vol.28, issue.11, pp.1239-51, 2014.
DOI : 10.1101/gad.240192.114

URL : https://hal.archives-ouvertes.fr/hal-01178971

J. George, L. Pine, M. Reeves, and W. Harrell, Amino acid requirements of Legionella pneumophila, J Clin Microbiol, vol.11, issue.3, pp.286-91, 1980.

B. James, W. Mauchline, P. Dennis, C. Keevil, and R. Wait, Poly-3-hydroxybutyrate in Legionella pneumophila , an energy source for survival in low-nutrient environments, Appl Environ Microbiol, vol.65, issue.2, pp.822-829, 1999.

J. Kacmar, R. Carlson, S. Balogh, and F. Srienc, Staining and quantification of poly-3-hydroxybutyrate in Saccharomyces cerevisiae and Cupriavidus necator cell populations using automated flow cytometry, Cytometry A, vol.69, issue.1, pp.27-35, 2006.

V. Bunik, A. Tylicki, and N. Lukashev, Thiamin diphosphate-dependent enzymes: from enzymology to metabolic regulation, drug design and disease models, FEBS Journal, vol.40, issue.24, pp.6412-6454, 2013.
DOI : 10.1093/nar/gkr998

J. Miranda-rios, The THI-box Riboswitch, or How RNA Binds Thiamin Pyrophosphate, Structure, vol.15, issue.3, pp.259-65, 2007.
DOI : 10.1016/j.str.2007.02.001

J. Ristroph, K. Hedlund, and S. Gowda, Chemically defined medium for Legionella pneumophila growth, J Clin Microbiol, vol.13, pp.115-124, 1981.

L. Patterson-fortin, C. Vakulskas, H. Yakhnin, P. Babitzke, and T. Romeo, Dual Posttranscriptional Regulation via a Cofactor-Responsive mRNA Leader, Journal of Molecular Biology, vol.425, issue.19, pp.3662-77, 2013.
DOI : 10.1016/j.jmb.2012.12.010

M. Nairz, A. Schroll, E. Demetz, I. Tancevski, I. Theurl et al., ???Ride on the ferrous wheel??? ??? The cycle of iron in macrophages in health and disease, Immunobiology, vol.220, issue.2, pp.280-94, 2015.
DOI : 10.1016/j.imbio.2014.09.010

N. Cianciotto, : new pathways for siderophore uptake and ferric iron reduction, Future Microbiology, vol.2, issue.6, pp.841-51, 2015.
DOI : 10.1007/s10534-009-9219-2

G. Porcheron and C. Dozois, Interplay between iron homeostasis and virulence: Fur and RyhB as major regulators of bacterial pathogenicity, Veterinary Microbiology, vol.179, issue.1-2, pp.2-14, 2015.
DOI : 10.1016/j.vetmic.2015.03.024

URL : https://hal.archives-ouvertes.fr/pasteur-01352636

E. Portier, H. Zheng, T. Sahr, D. Burnside, C. Mallama et al., virulence against amoebae and macrophages, Environmental Microbiology, vol.81, issue.Part 6, pp.1338-50, 2015.
DOI : 10.1128/IAI.00858-13

URL : https://hal.archives-ouvertes.fr/pasteur-01327258

M. Liu, H. Yang, and T. Romeo, The product of the pleiotropic Escherichia coli gene csrA modulates glycogen biosynthesis via effects on mRNA stability., Journal of Bacteriology, vol.177, issue.10, pp.2663-72, 1995.
DOI : 10.1128/jb.177.10.2663-2672.1995

A. Yakhnin, C. Baker, C. Vakulskas, H. Yakhnin, I. Berezin et al., mRNA from RNase E-mediated cleavage, Molecular Microbiology, vol.31, issue.4, pp.851-66, 2013.
DOI : 10.1093/nar/gkg595

G. Hovel-miner, S. Pampou, S. Faucher, M. Clarke, I. Morozova et al., ??S Controls Multiple Pathways Associated with Intracellular Multiplication of Legionella pneumophila, Journal of Bacteriology, vol.191, issue.8, pp.2461-73, 2009.
DOI : 10.1128/JB.01578-08

H. Yakhnin, A. Yakhnin, C. Baker, E. Sineva, I. Berezin et al., Complex regulation of the global regulatory gene csrA: CsrA-mediated translational repression, transcription from five promoters by E??70 and E??S, and indirect transcriptional activation by CsrA, Molecular Microbiology, vol.64, issue.3, pp.689-704, 2011.
DOI : 10.1111/j.1365-2958.2007.05765.x

A. Edwards, L. Patterson-fortin, C. Vakulskas, J. Mercante, K. Potrykus et al., Circuitry linking the Csr and stringent response global regulatory systems, Molecular Microbiology, vol.178, issue.6, pp.1561-80, 2011.
DOI : 10.1128/jb.178.4.1012-1017.1996

J. Williams, A. Ritter, and A. Stevens, CsrA modulates luxR transcript levels in Vibrio fischeri, FEMS Microbiology Letters, vol.329, issue.1, pp.28-35, 2012.
DOI : 10.1111/j.1574-6968.2012.02499.x

M. Lomma, D. Dervins-ravault, R. M. Nora, T. Newton, H. Sansom et al., The Legionella pneumophila F-box protein Lpp2082 (AnkB) modulates ubiquitination of the host protein parvin B and promotes intracellular replication, Cellular Microbiology, vol.202, issue.9, pp.1272-91, 2010.
DOI : 10.1016/j.bbamcr.2004.01.006

T. Sahr and C. Buchrieser, Co-immunoprecipitation: Protein???RNA and Protein???DNA Interaction, Methods Mol Biol, vol.954, pp.583-93, 2013.
DOI : 10.1007/978-1-62703-161-5_36

URL : https://hal.archives-ouvertes.fr/pasteur-01333974

T. Sahr and C. Buchrieser, cDNA Library Construction for Next-Generation Sequencing to Determine the Transcriptional Landscape of Legionella pneumophila, Methods Mol Biol, vol.954, pp.555-66, 2013.
DOI : 10.1007/978-1-62703-161-5_34

URL : https://hal.archives-ouvertes.fr/pasteur-01334075

A. Quinlan and I. Hall, BEDTools: a flexible suite of utilities for comparing genomic features, Bioinformatics, vol.26, issue.6, pp.841-843, 2010.
DOI : 10.1093/bioinformatics/btq033

T. Carver, S. Harris, M. Berriman, J. Parkhill, and J. Mcquillan, Artemis: an integrated platform for visualization and analysis of high-throughput sequence-based experimental data, Bioinformatics, vol.28, issue.4, pp.464-473, 2012.
DOI : 10.1093/bioinformatics/btr703

R. Edgar, M. Domrachev, and A. Lash, Gene Expression Omnibus: NCBI gene expression and hybridization array data repository, Nucleic Acids Research, vol.30, issue.1, pp.207-217, 2002.
DOI : 10.1093/nar/30.1.207

E. Milohanic, P. Glaser, J. Coppee, L. Frangeul, Y. Vega et al., Transcriptome analysis of Listeria monocytogenes identifies three groups of genes differently regulated by PrfA, Molecular Microbiology, vol.171, issue.23, pp.1613-1638, 2003.
DOI : 10.1128/jb.171.5.2795-2802.1989

Y. Yang, S. Dudoit, P. Luu, D. Lin, V. Peng et al., Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation, Nucleic Acids Research, vol.30, issue.4, pp.15-11842121, 2002.
DOI : 10.1093/nar/30.4.e15

P. Delmar, R. S. Daudin, and J. , VarMixt: efficient variance modelling for the differential analysis of replicated gene expression data, Bioinformatics, vol.21, issue.4, pp.502-510, 2005.
DOI : 10.1093/bioinformatics/bti023

URL : https://hal.archives-ouvertes.fr/hal-00126119

A. Reiner, D. Yekutieli, and Y. Benjamini, Identifying differentially expressed genes using false discovery rate controlling procedures, Bioinformatics, vol.19, issue.3, pp.368-75, 2003.
DOI : 10.1093/bioinformatics/btf877

J. Cox and M. Mann, MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification, Nature Biotechnology, vol.7, issue.12, pp.1367-72, 2008.
DOI : 10.1038/nprot.2007.261

J. Cox, N. Neuhauser, A. Michalski, R. Scheltema, J. Olsen et al., Andromeda: A Peptide Search Engine Integrated into the MaxQuant Environment, Journal of Proteome Research, vol.10, issue.4, pp.1794-805, 2011.
DOI : 10.1021/pr101065j

J. Cox, M. Hein, C. Luber, I. Paron, N. Nagaraj et al., Accurate Proteome-wide Label-free Quantification by Delayed Normalization and Maximal Peptide Ratio Extraction, Termed MaxLFQ, Molecular & Cellular Proteomics, vol.10, issue.9, pp.2513-2539, 2014.
DOI : 10.1074/mcp.M111.011015

J. Vizcaino, A. Csordas, N. Toro, J. Dianes, J. Griss et al., 2016 update of the PRIDE database and its related tools, Nucleic Acids Research, vol.44, issue.D1, pp.447-56, 2016.
DOI : 10.1093/nar/gkv1145