K. A. Allard, V. K. Viswanathan, and N. P. Cianciotto, lbtA and lbtB are required for production of the Legionella pneumophila siderophore legiobactin, J Bacteriol, vol.188, pp.1351-63, 2006.

Z. Baharoglu and D. Mazel, SOS, the formidable strategy of bacteria against aggressions, FEMS Microbiol Rev, vol.38, pp.1126-1171, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01423593

A. L. Barry, W. A. Craig, H. Nadler, L. B. Reller, C. C. Sanders et al., Methods for Determining Bactericidal Activity of Antimicrobial Agents; Approved Guideline, 1999.

S. Boisset, Y. Caspar, V. Sutera, and M. Maurin, New therapeutic approaches for treatment of tularaemia: a review, Front Cell Infect Microbiol, vol.4, p.40, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00986421

K. E. Bonnington and M. J. Kuehn, Protein selection and export via outer membrane vesicles, Biochim Biophys Acta, vol.1843, pp.1612-1621, 2014.

A. Brauner, O. Fridman, O. Gefen, and N. Q. Balaban, Distinguishing between resistance, tolerance and persistence to antibiotic treatment, Nat Rev Microbiol, vol.14, pp.320-350, 2016.

A. Brauner, N. Shoresh, O. Fridman, and N. Q. Balaban, An Experimental Framework for Quantifying Bacterial Tolerance, Biophys J, vol.112, pp.2664-2671, 2017.

N. L. Brown, J. V. Stoyanov, S. P. Kidd, and J. L. Hobman, The MerR family of transcriptional regulators, FEMS Microbiol Rev, vol.27, pp.145-63, 2003.

E. Brudal, E. O. Lampe, L. Reubsaet, N. Roos, I. K. Hegna et al., Vaccination with outer membrane vesicles from Francisella noatunensis reduces development of francisellosis in a zebrafish model, Fish Shellfish Immunol, vol.42, pp.50-57, 2015.

M. G. Casabona, Y. Vandenbrouck, I. Attree, and Y. Coute, Proteomic characterization of Pseudomonas aeruginosa PAO1 inner membrane, Proteomics, vol.13, pp.2419-2442, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02135130

Y. Caspar, C. Siebert, V. Sutera, C. Villers, A. A. Mayer et al., Functional Characterization of the DNA Gyrases in Fluoroquinolone-Resistant Mutants of Francisella novicida, Antimicrob Agents Chemother, p.61, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02107075

T. Chance, J. Chua, R. G. Toothman, J. T. Ladner, J. E. Nuss et al., A spontaneous mutation in kdsD, a biosynthesis gene for 3 Deoxy-D-mannoOctulosonic Acid, occurred in a ciprofloxacin resistant strain of Francisella tularensis and caused a high level of attenuation in murine models of tularemia, PLoS One, vol.12, p.174106, 2017.

J. C. Chandler, M. D. Sutherland, M. R. Harton, C. R. Molins, R. V. Anderson et al., Francisella tularensis LVS surface and membrane proteins as targets of effective postexposure immunization for tularemia, J Proteome Res, vol.14, pp.664-75, 2015.

M. K. Chattopadhyay and M. V. Jaganandham, Vesicles-mediated resistance to antibiotics in bacteria, Frontiers in microbiology, vol.6, p.758, 2015.

F. Chen, G. Cui, S. Wang, M. Nair, L. He et al., Outer membrane vesicle-associated lipase FtlA enhances cellular invasion and virulence in Francisella tularensis LVS, Emerg Microbes Infect, vol.6, p.66, 2017.

J. Cox and M. Mann, MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification, Nat Biotechnol, vol.26, pp.1367-72, 2008.

S. N. Dean, M. C. Chung, and M. L. Van-hoek, Burkholderia Diffusible Signal Factor Signals to Francisella novicida To Disperse Biofilm and Increase Siderophore Production, Appl Environ Microbiol, vol.81, pp.7057-66, 2015.

T. Dorr, K. Lewis, and M. Vulic, SOS response induces persistence to fluoroquinolones in Escherichia coli, PLoS Genet, vol.5, p.1000760, 2009.

B. Ezraty and F. Barras, The 'liaisons dangereuses' between iron and antibiotics, FEMS Microbiol Rev, vol.40, pp.418-453, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01440752

A. Fabrega, S. Madurga, E. Giralt, and J. Vila, Mechanism of action of and resistance to quinolones, Microb Biotechnol, vol.2, pp.40-61, 2009.

G. Gesbert, E. Ramond, M. Rigard, E. Frapy, M. Dupuis et al., Asparagine assimilation is critical for intracellular replication and dissemination of Francisella, Cell Microbiol, vol.16, pp.434-483, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00972076

T. D. Goddard, C. C. Huang, E. C. Meng, E. F. Pettersen, G. S. Couch et al., UCSF ChimeraX: Meeting modern challenges in visualization and analysis, Protein Sci, vol.27, pp.14-25, 2018.

M. F. Haurat, J. Aduse-opoku, M. Rangarajan, L. Dorobantu, M. R. Gray et al., Selective sorting of cargo proteins into bacterial membrane vesicles, J Biol Chem, vol.286, pp.1269-76, 2011.

J. F. Huntley, P. G. Conley, K. E. Hagman, and M. V. Norgard, Characterization of Francisella tularensis outer membrane proteins, J Bacteriol, vol.189, pp.561-74, 2007.

C. J. Jaing, K. S. Mcloughlin, J. B. Thissen, A. Zemla, S. N. Gardner et al., Identification of Genome-Wide Mutations in Ciprofloxacin-Resistant F. tularensis LVS Using Whole Genome Tiling Arrays and Next Generation Sequencing, PLoS One, vol.11, p.163458, 2016.

A. T. Jan, Outer Membrane Vesicles (OMVs) of Gram-negative Bacteria: A Perspective Update, Frontiers in microbiology, vol.8, p.1053, 2017.

A. S. Juncker, H. Willenbrock, V. Heijne, G. Brunak, S. Nielsen et al., Prediction of lipoprotein signal peptides in Gram-negative bacteria, Protein Sci, vol.12, pp.1652-62, 2003.

P. Larsson, P. C. Oyston, P. Chain, M. C. Chu, M. Duffield et al., The complete genome sequence of Francisella tularensis, the causative agent of tularemia, Nat Genet, vol.37, pp.153-162, 2005.

H. Lindgren, M. Honn, I. Golovlev, K. Kadzhaev, W. Conlan et al., The 58-kilodalton major virulence factor of Francisella tularensis is required for efficient utilization of iron, Infect Immun, vol.77, pp.4429-4465, 2009.

B. M. Loveless, A. Yermakova, D. R. Christensen, J. P. Kondig, H. S. Heine et al., Identification of ciprofloxacin resistance by SimpleProbe, High Resolution Melt and Pyrosequencing nucleic acid analysis in biothreat agents: Bacillus anthracis, Yersinia pestis and Francisella tularensis, Mol Cell Probes, vol.24, pp.154-60, 2010.

E. D. Lovullo, C. R. Molins-schneekloth, H. P. Schweizer, and M. S. Pavelka, Single-copy chromosomal integration systems for Francisella tularensis, Microbiology, vol.155, pp.1152-63, 2009.

E. D. Lovullo, L. A. Sherrill, and M. S. Pavelka, Improved shuttle vectors for Francisella tularensis genetics, FEMS Microbiol Lett, vol.291, pp.95-102, 2009.

J. L. Martinez and F. Rojo, Metabolic regulation of antibiotic resistance, FEMS Microbiol Rev, vol.35, pp.768-89, 2011.

M. Maurin and M. Gyuranecz, Tularaemia: clinical aspects in Europe, Lancet Infect Dis, vol.16, pp.113-137, 2016.

W. D. Mccaig, A. Koller, and D. G. Thanassi, Production of outer membrane vesicles and outer membrane tubes by Francisella novicida, J Bacteriol, vol.195, pp.1120-1152, 2013.

H. Mcwilliam, W. Li, M. Uludag, S. Squizzato, Y. M. Park et al., Analysis Tool Web Services from the EMBL-EBI, Nucleic Acids Res, vol.41, pp.597-600, 2013.

C. Medigue, A. Calteau, S. Cruveiller, M. Gachet, G. Gautreau et al., Adaptation of mycoplasmas to antimicrobial agents: Acholeplasma laidlawii extracellular vesicles mediate the export of ciprofloxacin and a mutant gene related to the antibiotic target, Brief Bioinform Medvedeva ES, vol.2014, p.150615, 2014.

K. C. Nallaparaju, J. J. Yu, S. A. Rodriguez, X. Zogaj, S. Manam et al., Evasion of IFN-gamma signaling by Francisella novicida is dependent upon Francisella outer membrane protein C, PLoS One, vol.6, p.18201, 2011.

P. C. Oyston, A. Sjostedt, and R. W. Titball, Tularaemia: bioterrorism defence renews interest in Francisella tularensis, Nat Rev Microbiol, vol.2, pp.967-78, 2004.

S. Pasquaroli, G. Zandri, C. Vignaroli, C. Vuotto, G. Donelli et al., Antibiotic pressure can induce the viable but non-culturable state in Staphylococcus aureus growing in biofilms, J Antimicrob Chemother, vol.68, pp.1812-1819, 2013.

J. Perard, J. Coves, M. Castellan, C. Solard, M. Savard et al., New Subfamily of Tetrameric Proteins. Biochemistry, vol.55, pp.1503-1518, 2016.

T. Pierson, D. Matrakas, Y. U. Taylor, G. Manyam, V. N. Morozov et al., Proteomic characterization and functional analysis of outer membrane vesicles of Francisella novicida suggests possible role in virulence and use as a vaccine, J Proteome Res, vol.10, pp.954-67, 2011.

D. Post, B. Slutter, B. Schilling, A. T. Chande, J. A. Rasmussen et al., Characterization of Inner and Outer Membrane Proteins from Francisella tularensis Strains LVS and Schu S4 and Identification of Potential Subunit Vaccine Candidates, MBio, vol.8, 2017.

G. Ramakrishnan, Iron and Virulence in Francisella tularensis, Front Cell Infect Microbiol, vol.7, p.107, 2017.

G. Ramakrishnan, A. Meeker, and B. Dragulev, ) fslE is necessary for siderophore-mediated iron acquisition in Francisella tularensis Schu S4, J Bacteriol, vol.190, pp.5353-61, 2008.

G. Ramakrishnan and B. Sen, The FupA/B protein uniquely facilitates transport of ferrous iron and siderophore-associated ferric iron across the outer membrane of Francisella tularensis live vaccine strain, Microbiology, vol.160, pp.446-57, 2014.

G. Ramakrishnan, B. Sen, and R. Johnson, Paralogous outer membrane proteins mediate uptake of different forms of iron and synergistically govern virulence in Francisella tularensis tularensis, J Biol Chem, vol.287, pp.25191-202, 2012.

X. Robert and P. Gouet, Deciphering key features in protein structures with the new ENDscript server, Nucleic Acids Res, vol.42, pp.320-324, 2014.

L. Rohmer, M. Brittnacher, K. Svensson, D. Buckley, E. Haugen et al., Potential source of Francisella tularensis live vaccine strain attenuation determined by genome comparison, Infect Immun, vol.74, pp.6895-906, 2006.

S. Roier, F. G. Zingl, F. Cakar, S. Durakovic, P. Kohl et al., A novel mechanism for the biogenesis of outer membrane vesicles in Gram-negative bacteria, Nature communications, vol.7, p.10515, 2016.

V. Sampath, W. D. Mccaig, and D. G. Thanassi, Amino acid deprivation and central carbon metabolism regulate the production of outer membrane vesicles and tubes by Francisella, Mol Microbiol Schwechheimer C, Kuehn MJ, vol.13, pp.605-624, 2015.

B. Sen, A. Meeker, and G. Ramakrishnan, The fslE homolog, FTL_0439 (fupA/B), mediates siderophore-dependent iron uptake in Francisella tularensis LVS, Infect Immun, vol.78, pp.4276-85, 2010.

V. Sutera, M. Levert, W. P. Burmeister, D. Schneider, and M. Maurin, Evolution toward high-level fluoroquinolone resistance in Francisella species, J Antimicrob Chemother, vol.69, pp.101-111, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00961489

S. Twine, M. Bystrom, W. Chen, M. Forsman, I. Golovliov et al., A mutant of Francisella tularensis strain SCHU S4 lacking the ability to express a 58-kilodalton protein is attenuated for virulence and is an effective live vaccine, Infect Immun, vol.73, pp.8345-52, 2005.

M. L. Van-hoek, Biofilms: an advancement in our understanding of Francisella species, Virulence, vol.4, pp.833-879, 2013.

J. A. Vizcaino, A. Csordas, N. Del-toro, J. A. Dianes, J. Griss et al., 2016 update of the PRIDE database and its related tools, Nucleic Acids Res, vol.44, pp.447-56, 2016.

S. Wieczorek, F. Combes, C. Lazar, G. Gianetto, Q. Gatto et al., DAPAR & ProStaR: software to perform statistical analyses in quantitative discovery proteomics, Bioinformatics, vol.33, pp.135-136, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02083847

M. M. Wilson and H. D. Bernstein, Surface-Exposed Lipoproteins: An Emerging Secretion Phenomenon in Gram-Negative Bacteria, Trends Microbiol, vol.24, pp.198-208, 2016.

X. Wu, G. Ren, W. T. Gunning, D. A. Weaver, A. L. Kalinoski et al., FmvB: A Francisella tularensis Magnesium-Responsive Outer Membrane Protein that Plays a Role in Virulence, PLoS One, vol.11, p.160977, 2016.

N. Y. Yu, J. R. Wagner, M. R. Laird, G. Melli, S. Rey et al., PSORTb 3.0: improved protein subcellular localization prediction with refined localization subcategories and predictive capabilities for all prokaryotes, Bioinformatics, vol.26, pp.1608-1623, 2010.

A. Sjöstedt and . Tularemia, History, Epidemiology, Pathogen Physiology, and Clinical Manifestations, Ann N Y Acad Sci. 1 juin, vol.1105, issue.1, pp.1-29, 2007.

W. B. Wherry and B. H. Lamb, Infection of man with Bacterium tularense, J Infect Dis. 1 avr, vol.189, issue.7, pp.1321-1330, 1914.

F. E. , JAMA. 16 déc, vol.250, issue.23, pp.3216-3240, 1925.

J. Ellis, P. Oyston, M. Green, R. W. Titball, and . Tularemia, Clin Microbiol Rev, vol.15, issue.4, pp.631-677, 2002.

D. B. Ritter and R. K. Gerloff, Deoxyribonucleic acid hybridization among some species of the genus Pasteurella, J Bacteriol. déc, vol.92, issue.6, pp.1838-1847, 1966.

K. A. Dorofe'ev, Classification of the causative agent of tularemia, Symposium Research Works Institute Epidemiology and Microbiology Chita, pp.170-180, 1947.

C. R. Woese, O. Kandler, and M. L. Wheelis, Towards a natural system of organisms: proposal for the domains Archaea, Bacteria, and Eucarya, Proc Natl Acad Sci U S A. juin, vol.87, issue.12, pp.4576-4585, 1990.

M. A. Larson, U. Nalbantoglu, K. Sayood, E. B. Zentz, R. Z. Cer et al., Reclassification of Wolbachia persica as Francisella persica comb. nov. and emended description of the family Francisellaceae, Int J Syst Evol Microbiol. mars, vol.66, issue.3, pp.1200-1205, 2016.

M. Forsman, G. Sandström, and A. Sjöstedt, Analysis of 16S ribosomal DNA sequences of Francisella strains and utilization for determination of the phylogeny of the genus and for identification of strains by PCR, Int J Syst Bacteriol. janv, vol.44, issue.1, pp.38-46, 1994.

P. Larsson, P. Oyston, P. Chain, M. C. Chu, M. Duffield et al., The complete genome sequence of Francisella tularensis, the causative agent of tularemia, Nat Genet. févr, vol.37, issue.2, pp.153-162, 2005.

J. F. Challacombe, J. M. Petersen, L. V. Gallegos-graves, D. Hodge, S. Pillai et al., Whole-Genome Relationships among Francisella Bacteria of Diverse Origins Define New Species and Provide Specific Regions for Detection, Appl Environ Microbiol. 2 janv, vol.83, issue.3, pp.2589-2605, 2017.

N. G. Olsufiev, O. S. Emelyanova, and T. N. Dunayeva, Comparative study of strains of B. tularense in the old and new world and their taxonomy, J Hyg Epidemiol Microbiol Immunol, vol.3, pp.138-187, 1959.

R. R. Chaudhuri, C. Ren, L. Desmond, A. Vincent, G. Silman et al.,

, Genome Sequencing Shows that European Isolates of Francisella tularensis Subspecies tularensis Are Almost Identical to US Laboratory Strain Schu S4. PLoS ONE. 4 avr, vol.2, 2007.

M. Maurin and M. Gyuranecz, Tularaemia: clinical aspects in Europe, Lancet Infect Dis. janv, vol.16, issue.1, pp.113-137, 2016.

P. Keim, A. Johansson, and D. M. Wagner, Molecular Epidemiology, Evolution, and Ecology of Francisella, Ann N Y Acad Sci. 1 juin, vol.1105, issue.1, pp.30-66, 2007.

H. T. Eigelsbach and C. M. Downs, Prophylactic Effectiveness of Live and Killed Tularemia Vaccines, J Immunol, vol.87, issue.4, pp.415-440, 1961.

D. S. Burke, Immunization against tularemia: analysis of the effectiveness of live Francisella tularensis vaccine in prevention of laboratory-acquired tularemia, J Infect Dis. janv, vol.135, issue.1, pp.55-60, 1977.

E. Salomonsson, K. Kuoppa, A. Forslund, C. Zingmark, I. Golovliov et al., Reintroduction of Two Deleted Virulence Loci Restores Full Virulence to the Live Vaccine Strain of Francisella tularensis, Infect Immun. août, vol.77, issue.8, pp.3424-3455, 2009.

H. M. Rowe and J. F. Huntley, From the Outside-In: The Francisella tularensis Envelope and Virulence, Front Cell Infect Microbiol, vol.5, p.94, 2015.

B. D. Jones, M. Faron, J. A. Rasmussen, and J. R. Fletcher, Uncovering the components of the Francisella tularensis virulence stealth strategy, Front Cell Infect Microbiol, vol.4, p.32, 2014.

K. L. Elkins, S. C. Cowley, and C. M. Bosio, Innate and adaptive immunity to Francisella, Ann N Y Acad Sci. juin, vol.1105, pp.284-324, 2007.

P. Larsson, D. Elfsmark, K. Svensson, P. Wikström, M. Forsman et al., Molecular Evolutionary Consequences of Niche Restriction in Francisella tularensis, a Facultative Intracellular Pathogen, PLoS Pathog. juin, vol.5, issue.6, 2009.

B. Huber, R. Escudero, H. Busse, E. Seibold, H. C. Scholz et al., Description of Francisella hispaniensis sp. nov., isolated from human blood, reclassification of Francisella novicida (Larson et al. 1955) Olsufiev et al. 1959 as Francisella tularensis subsp. novicida comb. nov. and emended description of the genus Francisella, Int J Syst Evol Microbiol. août, vol.60, pp.1887-96, 2010.

W. I. Jensen, C. R. Owen, and W. L. Jellison, Yersinia philomiragia sp. n., a New Member of the Pasteurella Group of Bacteria, Naturally Pathogenic for the Muskrat

, J Bacteriol. déc, vol.100, issue.3, pp.1237-1278, 1969.

D. G. Hollis, R. E. Weaver, A. G. Steigerwalt, J. D. Wenger, C. W. Moss et al., Francisella philomiragia comb. nov. (formerly Yersinia philomiragia) and Francisella tularensis biogroup novicida (formerly Francisella novicida) associated with human disease, J Clin Microbiol. juill, vol.27, issue.7, pp.1601-1609, 1989.

K. F. Ottem, A. Nylund, E. Karlsbakk, A. Friis-møller, and T. Kamaishi, Elevation of Francisella philomiragia subsp. noatunensis Mikalsen et al. (2007) to Francisella noatunensis comb. nov. [syn. Francisella piscicida Ottem et al. (2008) syn. nov.] and characterization of Francisella noatunensis subsp. orientalis subsp. nov., two important fish pathogens, J Appl Microbiol. avr, vol.106, issue.4, pp.1231-1274, 2009.

B. Huber, R. Escudero, H. Busse, E. Seibold, H. C. Scholz et al., Description of Francisella hispaniensis sp. nov., isolated from human blood, reclassification of Francisella novicida (Larson et al. 1955) Olsufiev et al. 1959 as Francisella tularensis subsp. novicida comb. nov. and emended description of the genus Francisella, Int J Syst Evol Microbiol. août, vol.60, pp.1887-96, 2010.

C. Caipang, A. Kulkarni, M. F. Brinchmann, K. Korsnes, and V. Kiron, Detection of Francisella piscicida in Atlantic cod (Gadus morhua L) by the loop-mediated isothermal amplification (LAMP) reaction, Vet J Lond Engl, vol.184, issue.3, pp.357-61, 1997.

O. J. Brevik, K. F. Ottem, T. Kamaishi, K. Watanabe, and A. Nylund, Francisella halioticida sp. nov., a pathogen of farmed giant abalone (Haliotis gigantea) in Japan, J Appl Microbiol, vol.111, issue.5, pp.1044-56, 2011.

P. Qu, S. Chen, H. C. Scholz, H. Busse, Q. Gu et al., Francisella guangzhouensis sp. nov., isolated from air-conditioning systems, Int J Syst Evol Microbiol, vol.63, pp.3628-3663, 2013.

J. F. Petrosino, Q. Xiang, S. E. Karpathy, H. Jiang, S. Yerrapragada et al., Chromosome rearrangement and diversification of Francisella tularensis revealed by the type B (OSU18) genome sequence, J Bacteriol, vol.188, pp.6977-85, 2006.

L. Rohmer, C. Fong, S. Abmayr, M. Wasnick, L. Freeman et al., Comparison of Francisella tularensis genomes reveals evolutionary events associated with the emergence of human pathogenic strains, Genome Biol, vol.8, issue.6, p.102, 2007.

A. J. Vogler, D. Birdsell, L. B. Price, J. R. Bowers, S. M. Beckstrom-sternberg et al., Phylogeography of Francisella tularensis: global expansion of a highly fit clone, J Bacteriol. avr, vol.191, issue.8, pp.2474-84, 2009.

Y. Wang, Y. Peng, R. Hai, L. Xia, H. Li et al., Diversity of Francisella tularensis subsp. holarctica lineages, China. Emerg Infect Dis. juill, vol.20, issue.7, pp.1191-1195, 2014.

M. Maurin and M. Gyuranecz, Tularaemia: clinical aspects in Europe, Lancet Infect Dis. janv, vol.16, issue.1, pp.113-137, 2016.

M. Maurin, Francisella tularensis as a potential agent of bioterrorism?, Expert Rev Anti Infect Ther. févr, vol.13, issue.2, pp.141-145, 2015.

P. Oyston, A. Sjöstedt, and R. W. Titball, Tularaemia: bioterrorism defence renews interest in Francisella tularensis, Nat Rev Microbiol. déc, vol.2, issue.12, pp.967-78, 2004.

N. G. Olsufiev, O. S. Emelyanova, and T. N. Dunayeva, Comparative study of strains of B. tularense in the old and new world and their taxonomy, J Hyg Epidemiol Microbiol Immunol, vol.3, pp.138-187, 1959.

L. C. Kingry and J. M. Petersen, Comparative review of Francisella tularensis and Francisella novicida, Front Cell Infect Microbiol, vol.4, p.35, 2014.

E. Meunier, P. Wallet, R. F. Dreier, S. Costanzo, A. L. Rühl et al., Guanylate-binding proteins promote activation of the AIM2 inflammasome during infection with Francisella novicida, Nat Immunol. mai, vol.16, issue.5, pp.476-84, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01924091

J. Ziveri, F. Tros, I. C. Guerrera, C. Chhuon, A. M. Dupuis et al., The metabolic enzyme fructose-1,6-bisphosphate aldolase acts as a transcriptional regulator in pathogenic Francisella, Nat Commun, vol.11, issue.1, p.853, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01886430

A. Sjöstedt, Intracellular survival mechanisms of Francisella tularensis, a stealth pathogen, Microbes Infect. févr, vol.8, issue.2, pp.561-568, 2006.

P. Wallet, B. Lagrange, and T. Henry, Francisella Inflammasomes: Integrated Responses to a Cytosolic Stealth Bacterium, Curr Top Microbiol Immunol, vol.397, pp.229-56, 2016.
DOI : 10.1007/978-3-319-41171-2_12

URL : https://hal.archives-ouvertes.fr/hal-01927023

J. Ziveri, M. Barel, and A. Charbit, Importance of Metabolic Adaptations in Francisella Pathogenesis, Front Cell Infect Microbiol, vol.7, p.96, 2017.

J. Pizarro-cerdá, A. Charbit, J. Enninga, F. Lafont, and P. Cossart, Manipulation of host membranes by the bacterial pathogens Listeria, Francisella, Shigella and Yersinia, Semin Cell Dev Biol, vol.60, pp.155-67, 2016.

G. B. Moreau and B. J. Mann, Adherence and uptake of Francisella into host cells, Virulence, vol.4, issue.8, pp.826-858, 2013.

A. Balagopal, A. S. Macfarlane, N. Mohapatra, S. Soni, J. S. Gunn et al., Characterization of the receptor-ligand pathways important for entry and survival of Francisella tularensis in human macrophages, Infect Immun. sept, vol.74, issue.9, pp.5114-5139, 2006.

G. S. Schulert and L. Allen, Differential infection of mononuclear phagocytes by Francisella tularensis: role of the macrophage mannose receptor, J Leukoc Biol. sept, vol.80, issue.3, pp.563-71, 2006.

H. Geier and J. Celli, Phagocytic receptors dictate phagosomal escape and intracellular proliferation of Francisella tularensis, Infect Immun. juin, vol.79, issue.6, pp.2204-2218, 2011.

B. Nasr, A. Haithcoat, J. Masterson, J. E. Gunn, J. S. Eaves-pyles et al., Critical role for serum opsonins and complement receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) in phagocytosis of Francisella tularensis by human dendritic cells (DC): uptake of Francisella leads to activation of immature DC and intracellular survival of the bacteria, J Leukoc Biol, vol.80, issue.4, pp.774-86, 2006.

L. M. Pierini, Uptake of serum-opsonized Francisella tularensis by macrophages can be mediated by class A scavenger receptors, Cell Microbiol. août, vol.8, issue.8, pp.1361-70, 2006.

M. Santic, R. Asare, I. Skrobonja, S. Jones, A. Kwaik et al., Acquisition of the vacuolar ATPase proton pump and phagosome acidification are essential for escape of Francisella tularensis into the macrophage cytosol, Infect Immun. juin, vol.76, issue.6, pp.2671-2678, 2008.

D. L. Clemens, B. Lee, and M. A. Horwitz, Francisella tularensis phagosomal escape does not require acidification of the phagosome, Infect Immun. mai, vol.77, issue.5, pp.1757-73, 2009.

F. E. Nano, N. Zhang, S. C. Cowley, K. E. Klose, K. Cheung et al., A Francisella tularensis Pathogenicity Island Required for Intramacrophage Growth, J Bacteriol, vol.186, pp.6430-6436, 2004.

J. R. Barker, A. Chong, T. D. Wehrly, J. Yu, S. A. Rodriguez et al., The Francisella tularensis Pathogenicity Island Encodes a Secretion System that is required for Phagosome Escape and Virulence, Mol Microbiol. déc, vol.74, issue.6, pp.1459-70, 2009.

A. Chong and J. Celli, The Francisella Intracellular Life Cycle: Toward Molecular Mechanisms of Intracellular Survival and Proliferation, Front Microbiol. 28 déc, vol.1, 2010.

E. Bourdonnay and T. Henry, Catch me if you can. eLife
URL : https://hal.archives-ouvertes.fr/hal-01909006

S. Steele, L. Radlinski, S. Taft-benz, J. Brunton, and T. H. Kawula, Trogocytosis-associated cell to cell spread of intracellular bacterial pathogens, eLife. 23 janv, vol.5, 2016.

L. A. Gallagher, E. Ramage, M. A. Jacobs, R. Kaul, M. Brittnacher et al., A comprehensive transposon mutant library of Francisella novicida, a bioweapon surrogate, Proc Natl Acad Sci U S A. 16 janv, vol.104, issue.3, pp.1009-1023, 2007.

D. S. Weiss, A. Brotcke, T. Henry, J. J. Margolis, K. Chan et al., In vivo negative selection screen identifies genes required for Francisella virulence, Proc Natl Acad Sci U S A. 3 avr, vol.104, issue.14, pp.6037-6079, 2007.

S. R. Lindemann, K. Peng, M. E. Long, J. R. Hunt, M. A. Apicella et al., Francisella tularensis Schu S4 O-antigen and capsule biosynthesis gene mutants induce early cell death in human macrophages, Infect Immun. févr, vol.79, issue.2, pp.581-94, 2011.

G. Sandström, S. Löfgren, and A. Tärnvik, A capsule-deficient mutant of Francisella tularensis LVS exhibits enhanced sensitivity to killing by serum but diminished sensitivity to killing by polymorphonuclear leukocytes, Infect Immun. mai, vol.56, issue.5, pp.1194-202, 1988.

M. A. Apicella, D. Post, A. C. Fowler, B. D. Jones, J. A. Rasmussen et al., Identification, characterization and immunogenicity of an O-antigen capsular polysaccharide of Francisella tularensis, PloS One. 6 juill, vol.5, issue.7, p.11060, 2010.

J. A. Rasmussen, J. R. Fletcher, M. E. Long, L. Allen, and B. D. Jones, Characterization of Francisella tularensis Schu S4 mutants identified from a transposon library screened for O-antigen and capsule deficiencies, Front Microbiol. 5 mai, vol.6, 2015.

A. B. Bandara, A. E. Champion, X. Wang, G. Berg, M. A. Apicella et al., Isolation and mutagenesis of a capsule-like complex (CLC) from Francisella tularensis, and contribution of the CLC to F. tularensis virulence in mice, PloS One. 22 avr, vol.6, issue.4, p.19003, 2011.

J. S. Gunn and R. K. Ernst, The Structure and Function of Francisella Lipopolysaccharide, Ann N Y Acad Sci. juin, vol.1105, pp.202-220, 2007.

M. S. Trent, C. M. Stead, A. X. Tran, and J. V. Hankins, Invited review: Diversity of endotoxin and its impact on pathogenesis, Invited review: Diversity of endotoxin and its impact on pathogenesis, J Endotoxin Res. 1 août, vol.12, issue.4, pp.205-228, 2006.

N. A. Okan and D. L. Kasper, The atypical lipopolysaccharide of Francisella, Carbohydr Res. 30 août, vol.378, pp.79-83, 2013.

B. Lagrange, S. Benaoudia, P. Wallet, F. Magnotti, A. Provost et al., Human caspase-4 detects tetra-acylated LPS and cytosolic Francisella and functions differently from murine caspase-11, Nat Commun. 16 janv, vol.9, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01911326

E. N. Salomonsson, A. Forslund, and A. Forsberg, Type IV Pili in Francisella-A Virulence Trait in an Intracellular Pathogen, Front Microbiol, vol.2, p.29, 2011.

A. Forslund, K. Kuoppa, K. Svensson, E. Salomonsson, A. Johansson et al., Direct repeat-mediated deletion of a type IV pilin gene results in major virulence attenuation of Francisella tularensis, Mol Microbiol. mars, vol.59, issue.6, pp.1818-1848, 2006.

A. Eshraghi, J. Kim, A. C. Walls, H. E. Ledvina, C. N. Miller et al., Secreted Effectors Encoded within and outside of the Francisella Pathogenicity Island Promote Intramacrophage Growth, Cell Host Microbe, vol.20, issue.5, pp.573-83, 2016.

M. Rigard, J. E. Bröms, A. Mosnier, M. Hologne, A. Martin et al., Francisella tularensis IglG Belongs to a Novel Family of PAAR-Like T6SS Proteins and Harbors a Unique N-terminal Extension Required for Virulence, PLoS Pathog. 7 sept, vol.12, issue.9, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01546486

G. Ramakrishnan, Iron and Virulence in Francisella tularensis, Front Cell Infect Microbiol, vol.7, p.107, 2017.

A. Nordstoga, K. Handeland, T. B. Johansen, L. Iversen, D. Gavier-widén et al., Tularaemia in Norwegian dogs, Vet Microbiol, vol.173, issue.3-4, pp.318-340, 2014.

M. A. Larson, P. D. Fey, S. H. Hinrichs, and P. C. Iwen, Francisella tularensis Bacteria Associated with Feline Tularemia in the United States, Emerg Infect Dis. déc, vol.20, issue.12, pp.2068-71, 2014.

A. N. Weinberg and J. A. Branda, Case records of the Massachusetts General Hospital. Case 31-2010. A 29-year-old woman with fever after a cat bite, N Engl J Med, vol.363, issue.16, pp.1560-1568, 2010.

J. M. Petersen, P. S. Mead, and M. E. Schriefer, Francisella tularensis: an arthropod-borne pathogen, Vet Res. avr, vol.40, issue.2, p.7, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00903078

J. Thelaus, A. Andersson, T. Broman, S. Bäckman, M. Granberg et al., Francisella tularensis subspecies holarctica occurs in Swedish mosquitoes, persists through the developmental stages of laboratory-infected mosquitoes and is transmissible during blood feeding, Microb Ecol. janv, vol.67, issue.1, pp.96-107, 2014.

V. Výrosteková,

, Epidemiol Mikrobiol Imunol Cas Spolecnosti Epidemiol Mikrobiol Ceske Lek Spolecnosti JE Purkyne. déc, vol.43, issue.4, pp.166-70, 1994.

S. Bäckman, J. Näslund, M. Forsman, and J. Thelaus, Transmission of tularemia from a water source by transstadial maintenance in a mosquito vector, Sci Rep. 22 janv, vol.5, p.7793, 2015.

T. Broman, J. Thelaus, A. Andersson, S. Bäckman, P. Wikström et al., Molecular Detection of Persistent Francisella tularensis Subspecies holarctica in Natural Waters, Int J Microbiol, 2011.

S. Kilic, D. N. Birdsell, A. Karagöz, B. Çelebi, Z. Bakkaloglu et al., Water as Source of Francisella tularensis Infection in Humans, Turkey. Emerg Infect Dis. déc, vol.21, issue.12, pp.2213-2219, 2015.

S. H. El-etr, J. J. Margolis, D. Monack, R. A. Robison, M. Cohen et al., Francisella tularensis type A strains cause the rapid encystment of Acanthamoeba castellanii and survive in amoebal cysts for three weeks postinfection, Appl Environ Microbiol. déc, vol.75, issue.23, pp.7488-500, 2009.

M. Maurin, I. Pelloux, J. P. Brion, D. Banõ, J. Picard et al., Human tularemia in France, Clin Infect Dis Off Publ Infect Dis Soc Am, vol.53, issue.10, pp.133-141, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00993402

S. Lang and M. Kleines, Two at one blow: reemergence of tularemia in Upper Austria, New Microbiol. juill, vol.35, issue.3, pp.349-52, 2012.

W. D. Splettstoesser, I. Piechotowski, A. Buckendahl, D. Frangoulidis, P. Kaysser et al., Tularemia in Germany: the tip of the iceberg?, Epidemiol Infect. mai, vol.137, issue.5, pp.736-779, 2009.

R. Reintjes, I. Dedushaj, A. Gjini, T. R. Jorgensen, B. Cotter et al., Tularemia outbreak investigation in Kosovo: case control and environmental studies, Emerg Infect Dis. janv, vol.8, issue.1, pp.69-73, 2002.

T. Kantardjiev, I. Ivanov, T. Velinov, P. Padeshki, B. Popov et al., Emerg Infect Dis. avr, vol.12, issue.4, pp.678-80, 1997.

P. Rydén, R. Björk, M. L. Schäfer, J. O. Lundström, B. Petersén et al., Outbreaks of tularemia in a boreal forest region depends on mosquito prevalence, J Infect Dis. 15 janv, vol.205, issue.2, pp.297-304, 2012.

J. Eden, K. Rose, J. Ng, M. Shi, Q. Wang et al., Francisella tularensis ssp. holarctica in Ringtail Possums, Australia. Emerg Infect Dis, vol.23, issue.7, pp.1198-201, 2017.

G. Hestvik, E. Warns-petit, L. A. Smith, N. J. Fox, H. Uhlhorn et al., The status of tularemia in Europe in a one-health context: a review, Epidemiol Infect. juill, vol.143, issue.10, pp.2137-60, 2015.

I. S. Meshcheryakova, T. V. Mikhailova, T. N. Demidova, and M. I. Kormilitsyna,

, Med Parazitol (Mosk). mars, issue.1, pp.42-48, 2016.

J. L. Pérez-castrillón, P. Bachiller-luque, M. Martín-luquero, F. J. Mena-martín, and V. Herreros, Tularemia epidemic in northwestern Spain: clinical description and therapeutic response, Clin Infect Dis Off Publ Infect Dis Soc Am. 15 août, vol.33, issue.4, pp.573-579, 2001.

A. Desvars, M. Furberg, M. Hjertqvist, L. Vidman, A. Sjöstedt et al., Epidemiology and ecology of tularemia in Sweden, Emerg Infect Dis. janv, vol.21, issue.1, pp.32-41, 1984.

M. Faber, K. Heuner, D. Jacob, and R. Grunow, Tularemia in Germany-A Re-emerging Zoonosis, Front Cell Infect Microbiol, vol.8, p.40, 2018.

A. Mailles and V. Vaillant, 10 years of surveillance of human tularaemia in France, Euro Surveill Bull Eur Sur Mal Transm Eur Commun Dis Bull, vol.19, issue.45, p.20956, 2014.

M. J. Hepburn and A. Simpson, Tularemia: current diagnosis and treatment options, Expert Rev Anti Infect Ther. avr, vol.6, issue.2, pp.231-271, 2008.

E. Prudent, L. Scola, B. Drancourt, M. Angelakis, E. Raoult et al., Molecular strategy for the diagnosis of infectious lymphadenitis, Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. juin, vol.37, issue.6, pp.1179-86, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01801664

O. Karatuna, B. Celebi, S. Can, I. Akyar, and S. Kilic, The use of Matrix-assisted laser desorption ionization-time of flight mass spectrometry in the identification of Francisella tularensis, Bosn J Basic Med Sci. 15 janv, vol.16, issue.2, pp.132-140, 2016.

N. S. Harik, Tularemia: epidemiology, diagnosis, and treatment, Pediatr Ann. juill, vol.42, issue.7, pp.288-92, 2013.

H. Yanes, A. Hennebique, I. Pelloux, S. Boisset, D. J. Bicout et al., Evaluation of In-House and Commercial Serological Tests for Diagnosis of Human Tularemia, J Clin Microbiol. janv, vol.56, issue.1, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02011114

R. Thomas, A. Johansson, B. Neeson, K. Isherwood, A. Sjöstedt et al., Discrimination of human pathogenic subspecies of Francisella tularensis by using restriction fragment length polymorphism, J Clin Microbiol. janv, vol.41, issue.1, pp.50-57, 2003.

T. Skottman, H. Piiparinen, H. Hyytiäinen, V. Myllys, M. Skurnik et al., Simultaneous real-time PCR detection of Bacillus anthracis, Francisella tularensis and Yersinia pestis, Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. mars, vol.26, issue.3, pp.207-218, 2007.

A. Sjöstedt, G. Sandström, A. Tärnvik, and B. Jaurin, Nucleotide sequence and T cell epitopes of a membrane protein of Francisella tularensis, J Immunol Baltim Md 1950. 1 juill, vol.145, issue.1, pp.311-318, 1990.

M. Fulop, D. Leslie, and R. Titball, A rapid, highly sensitive method for the detection of Francisella tularensis in clinical samples using the polymerase chain reaction, Am J Trop Med Hyg. avr, vol.54, issue.4, pp.364-370, 1996.

J. Thelaus, A. Andersson, P. Mathisen, A. Forslund, L. Noppa et al., Influence of nutrient status and grazing pressure on the fate of Francisella tularensis in lake water, FEMS Microbiol Ecol. janv, vol.67, issue.1, pp.69-80, 2009.

D. N. Birdsell, A. J. Vogler, J. Buchhagen, C. A. Kaufman, E. Naumann et al., TaqMan real-time PCR assays for single-nucleotide polymorphisms which identify Francisella tularensis and its subspecies and subpopulations, PloS One, vol.9, issue.9, p.107964, 2014.
DOI : 10.1371/journal.pone.0107964

URL : https://doi.org/10.1371/journal.pone.0107964

J. L. Versage, D. Severin, M. C. Chu, and J. M. Petersen, Development of a multitarget realtime TaqMan PCR assay for enhanced detection of Francisella tularensis in complex specimens, J Clin Microbiol. déc, vol.41, issue.12, pp.5492-5501, 2003.

K. J. Kugeler, R. Pappert, Y. Zhou, and J. M. Petersen, Real-time PCR for Francisella tularensis types A and B. Emerg Infect Dis, vol.12, pp.1799-801, 2006.

A. Tärnvik and M. C. Chu, New Approaches to Diagnosis and Therapy of Tularemia, Ann N Y Acad Sci. 1 juin, vol.1105, issue.1, pp.378-404, 2007.

D. T. Dennis, T. V. Inglesby, D. A. Henderson, J. G. Bartlett, M. S. Ascher et al., Tularemia as a biological weapon: medical and public health management, JAMA. 6 juin, vol.285, issue.21, pp.2763-73, 2001.

C. Dentan, P. Pavese, I. Pelloux, S. Boisset, J. Brion et al., Treatment of tularemia in pregnant woman, France. Emerg Infect Dis. juin, vol.19, issue.6, pp.996-1004, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00965575

S. K. Urich and J. M. Petersen, In vitro susceptibility of isolates of Francisella tularensis types A and B from North America, Antimicrob Agents Chemother. juin, vol.52, issue.6, pp.2276-2284, 2008.

Y. Caspar, A. Hennebique, and M. Maurin, Antibiotic susceptibility of Francisella tularensis subsp. holarctica strains isolated from tularaemia patients in France between 2006 and 2016, J Antimicrob Chemother. 14 déc, 2017.

P. Wayne, Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility Testing-Nineteeth Informational Supplement M100-S19. CLSI, 2009.

I. Ikäheimo, H. Syrjälä, J. Karhukorpi, R. Schildt, and M. Koskela, In vitro antibiotic susceptibility of Francisella tularensis isolated from humans and animals, J Antimicrob Chemother. août, vol.46, issue.2, pp.287-90, 2000.

E. Valade, J. Vaissaire, A. Mérens, E. Hernandez, C. Gros et al., Susceptibility of 71 French isolates of Francisella tularensis subsp. holarctica to eight antibiotics and accuracy of the Etest method, J Antimicrob Chemother. juill, vol.62, issue.1, pp.208-218, 2008.

V. Sutera, M. Levert, W. P. Burmeister, D. Schneider, and M. Maurin, Evolution toward highlevel fluoroquinolone resistance in Francisella species, J Antimicrob Chemother. janv, vol.69, issue.1, pp.101-111, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00961489

C. J. Jaing, K. S. Mcloughlin, J. B. Thissen, A. Zemla, S. N. Gardner et al., Identification of Genome-Wide Mutations in Ciprofloxacin-Resistant F. tularensis LVS Using Whole Genome Tiling Arrays and Next Generation Sequencing, PloS One, vol.11, issue.9, p.163458, 2016.

B. M. Loveless, A. Yermakova, D. R. Christensen, J. P. Kondig, H. S. Heine et al., Identification of ciprofloxacin resistance by SimpleProbe, High Resolution Melt and Pyrosequencing nucleic acid analysis in biothreat agents: Bacillus anthracis, Yersinia pestis and Francisella tularensis, Mol Cell Probes. juin, vol.24, issue.3, pp.154-60, 2010.

M. Toth, H. Frase, N. T. Antunes, and S. B. Vakulenko, Novel aminoglycoside 2''phosphotransferase identified in a gram-negative pathogen, Antimicrob Agents Chemother. janv, vol.57, issue.1, pp.452-459, 2013.

X. R. Bina, C. Wang, M. A. Miller, and J. E. Bina, The Bla2 beta-lactamase from the live-vaccine strain of Francisella tularensis encodes a functional protein that is only active against penicillin-class beta-lactam antibiotics, Arch Microbiol. sept, vol.186, issue.3, pp.219-247, 2006.

X. R. Bina, C. L. Lavine, M. A. Miller, and J. E. Bina, The AcrAB RND efflux system from the live vaccine strain of Francisella tularensis is a multiple drug efflux system that is required for virulence in mice, FEMS Microbiol Lett. févr, vol.279, issue.2, pp.226-259, 2008.

E. Karlsson, I. Golovliov, A. Lärkeryd, M. Granberg, E. Larsson et al., Clonality of erythromycin resistance in Francisella tularensis, J Antimicrob Chemother, vol.71, issue.10, pp.2815-2838, 2016.

H. Tezer, A. Ozkaya-parlakay, H. Aykan, M. Erkocoglu, B. Gülhan et al., Emerg Infect Dis. janv, vol.21, issue.1, pp.1-7, 2009.

A. Karl?, G. ?ensoy, ?. Paksu, M. F. Korkmaz, Ö. Ertu?rul et al., Treatment-failure tularemia in children, Korean J Pediatr. févr, vol.61, issue.2, pp.49-52, 2018.

V. Sutera, G. Hoarau, P. Renesto, Y. Caspar, and M. Maurin, In vitro and in vivo evaluation of fluoroquinolone resistance associated with DNA gyrase mutations in Francisella tularensis, including in tularaemia patients with treatment failure, Int J Antimicrob Agents. sept, vol.50, issue.3, pp.377-83, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02107079

G. Y. Lesher, E. J. Froelich, M. D. Gruett, and J. H. Bailey, Brundage RP. 1,8-NAPHTHYRIDINE DERIVATIVES. A NEW CLASS OF CHEMOTHERAPEUTIC AGENTS, J Med Pharm Chem. sept, vol.91, pp.1063-1068, 1962.

M. and M. Jg, Quinolones et fluoroquinolones : des décennies de développement et d'utilisation Le point sur les molécules vétérinaires, Ann. Méd. Vét, pp.73-87, 2014.

G. Capranico, J. Marinello, and G. Chillemi, Type I DNA Topoisomerases, J Med Chem. 23 mars, vol.60, issue.6, pp.2169-92, 2017.

P. Forterre, S. Gribaldo, D. Gadelle, and M. Serre, Origin and evolution of DNA topoisomerases, Biochimie. avr, vol.89, issue.4, pp.427-473, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00194416

V. Nagaraja, A. A. Godbole, S. R. Henderson, and A. Maxwell, DNA topoisomerase I and DNA gyrase as targets for TB therapy, Drug Discov Today. 1 mars, vol.22, issue.3, pp.510-518, 2017.

T. Khan, K. Sankhe, V. Suvarna, A. Sherje, K. Patel et al., DNA gyrase inhibitors: Progress and synthesis of potent compounds as antibacterial agents, Biomed Pharmacother. 1 juill, vol.103, pp.923-961, 2018.

J. M. Berger, Type II DNA topoisomerases, Curr Opin Struct Biol. 1 févr, vol.8, issue.1, pp.26-32, 1998.

J. Piton, S. Petrella, M. Delarue, G. André-leroux, V. Jarlier et al., Structural insights into the quinolone resistance mechanism of Mycobacterium tuberculosis DNA gyrase, PloS One. 18 août, vol.5, issue.8, p.12245, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-01126607

N. G. Bush, K. Evans-roberts, and M. A. Dna-topoisomerases, EcoSal Plus, vol.6, issue.2, 2015.

M. Gellert, K. Mizuuchi, O. Dea, M. H. Nash, and H. A. , DNA gyrase: an enzyme that introduces superhelical turns into DNA, Proc Natl Acad Sci, vol.73, issue.11, pp.3872-3878, 1976.

J. J. Champoux, DNA topoisomerases: structure, function, and mechanism, Annu Rev Biochem, vol.70, pp.369-413, 2001.

K. J. Aldred, R. J. Kerns, and N. Osheroff, Mechanism of quinolone action and resistance, Biochemistry. 18 mars, vol.53, issue.10, pp.1565-74, 2014.

T. R. Blower, B. H. Williamson, R. J. Kerns, and J. M. Berger, Crystal structure and stability of gyrase-fluoroquinolone cleaved complexes from Mycobacterium tuberculosis, Proc Natl Acad Sci U S A. 16 févr, vol.113, issue.7, pp.1706-1719, 2016.

J. Piton, S. Petrella, M. Delarue, G. André-leroux, V. Jarlier et al., Structural insights into the quinolone resistance mechanism of Mycobacterium tuberculosis DNA gyrase, PloS One. 18 août, vol.5, issue.8, p.12245, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-01126607

D. C. Hooper and G. A. Jacoby, Topoisomerase Inhibitors: Fluoroquinolone Mechanisms of Action and Resistance, Cold Spring Harb Perspect Med. 1 sept, vol.6, issue.9, 2016.

L. S. Redgrave, S. B. Sutton, M. A. Webber, and L. Piddock, Fluoroquinolone resistance: mechanisms, impact on bacteria, and role in evolutionary success, Trends Microbiol. août, vol.22, issue.8, pp.438-483, 2014.

L. Scola, B. Elkarkouri, K. Li, W. Wahab, T. Fournous et al., Rapid comparative genomic analysis for clinical microbiology: the Francisella tularensis paradigm, Genome Res. mai, vol.18, issue.5, pp.742-50, 2008.

S. Correia, P. Poeta, M. Hébraud, J. L. Capelo, and G. Igrejas, Mechanisms of quinolone action and resistance: where do we stand, J Med Microbiol, vol.66, issue.5, pp.551-560, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01607127

M. D. Routh, Y. Zalucki, C. Su, F. Long, Q. Zhang et al., Efflux pumps of the resistance-nodulation-division family: A perspective of their structure, function and regulation in gram-negative bacteria, Adv Enzymol Relat Areas Mol Biol, vol.77, pp.109-155, 2011.

H. Wang, J. L. Dzink-fox, M. Chen, and S. B. Levy, Genetic Characterization of Highly Fluoroquinolone-Resistant Clinical Escherichia coli Strains from China: Role of acrR Mutations, Antimicrob Agents Chemother. mai, vol.45, issue.5, pp.1515-1536, 2001.

A. Robicsek, J. Strahilevitz, G. A. Jacoby, M. Macielag, D. Abbanat et al., Fluoroquinolone-modifying enzyme: a new adaptation of a common aminoglycoside acetyltransferase, Nat Med. janv, vol.12, issue.1, pp.83-91, 2006.

K. Yamane, J. Wachino, S. Suzuki, and Y. Arakawa, Plasmid-mediated qepA gene among Escherichia coli clinical isolates from Japan, Antimicrob Agents Chemother. avr, vol.52, issue.4, pp.1564-1570, 2008.

L. H. Hansen, E. Johannesen, M. Burmølle, A. H. Sørensen, and S. J. Sørensen, Plasmid-Encoded Multidrug Efflux Pump Conferring Resistance to Olaquindox in Escherichia coli, Antimicrob Agents Chemother. sept, vol.48, issue.9, pp.3332-3339, 2004.

A. Filloux and I. Vallet, Biofilm: set-up and organization of a bacterial community

, Med Sci MS. janv, vol.19, issue.1, pp.77-83, 2003.

A. Roux and J. M. Ghigo, Les biofilms bactériens. Bulletin de l'Académie Vétérinaire de France, 2006.

D. Lebeaux and J. M. Ghigo, Les biofilms : d'intraitables adversaires, BioFutur, vol.32, pp.34-43, 2013.

M. W. Durham-colleran, A. B. Verhoeven, and M. L. Van-hoek, Francisella novicida forms in vitro biofilms mediated by an orphan response regulator, Microb Ecol. avr, vol.59, issue.3, pp.457-65, 2010.

A. Khweek, A. Amer, and A. O. , Factors Mediating Environmental Biofilm Formation by Legionella pneumophila, Front Cell Infect Microbiol, vol.8, p.38, 2018.

A. B. Verhoeven, M. W. Durham-colleran, T. Pierson, W. T. Boswell, V. Hoek et al., Francisella philomiragia biofilm formation and interaction with the aquatic protist Acanthamoeba castellanii, Biol Bull, vol.219, issue.2, pp.178-88, 2010.

J. J. Margolis, S. El-etr, L. Joubert, M. E. Robison, R. Rasley et al., Contributions of Francisella tularensis subsp. novicida chitinases and Sec secretion system to biofilm formation on chitin, Appl Environ Microbiol. janv, vol.76, issue.2, pp.596-608, 2010.

D. Lebeaux and J. Ghigo, Management of biofilm-associated infections: what can we expect from recent research on biofilm lifestyles?

, Med Sci MS. sept, vol.28, issue.8-9, pp.727-766, 2012.

S. Matrat, A. A. Mayer, C. Jarlier, V. Cambau, and E. , Mutagenesis in the alpha3alpha4 GyrA helix and in the Toprim domain of GyrB refines the contribution of Mycobacterium tuberculosis DNA gyrase to intrinsic resistance to quinolones, Antimicrob Agents Chemother. août, vol.52, issue.8, pp.2909-2923, 2008.

A. Mustaev, M. Malik, X. Zhao, N. Kurepina, G. Luan et al., Fluoroquinolone-gyrase-DNA complexes: two modes of drug binding, J Biol Chem. 2 mai, vol.289, issue.18, pp.12300-12312, 2014.

S. Twine, M. Byström, W. Chen, M. Forsman, I. Golovliov et al., A mutant of Francisella tularensis strain SCHU S4 lacking the ability to express a 58-kilodalton protein is attenuated for virulence and is an effective live vaccine, Infect Immun. déc, vol.73, issue.12, pp.8345-52, 2005.

K. C. Nallaparaju, J. Yu, S. A. Rodriguez, X. Zogaj, S. Manam et al., Evasion of IFN-? signaling by Francisella novicida is dependent upon Francisella outer membrane protein C, PloS One. 31 mars, vol.6, issue.3, p.18201, 2011.

H. Lindgren, M. Honn, I. Golovlev, K. Kadzhaev, W. Conlan et al., The 58-kilodalton major virulence factor of Francisella tularensis is required for efficient utilization of iron, Infect Immun, vol.77, issue.10, pp.4429-4465, 2009.

E. Salomonsson, K. Kuoppa, A. Forslund, C. Zingmark, I. Golovliov et al., Reintroduction of two deleted virulence loci restores full virulence to the live vaccine strain of Francisella tularensis, Infect Immun. août, vol.77, issue.8, pp.3424-3455, 2009.

B. Sen, A. Meeker, and G. Ramakrishnan, The fslE homolog, FTL_0439 (fupA/B), mediates siderophore-dependent iron uptake in Francisella tularensis LVS. Infect Immun, vol.78, pp.4276-85, 2010.

G. Ramakrishnan and B. Sen, The FupA/B protein uniquely facilitates transport of ferrous iron and siderophore-associated ferric iron across the outer membrane of Francisella tularensis live vaccine strain, Microbiol Read Engl. févr, vol.160, pp.446-57, 2014.

S. Pasquaroli, G. Zandri, C. Vignaroli, C. Vuotto, G. Donelli et al., Antibiotic pressure can induce the viable but non-culturable state in Staphylococcus aureus growing in biofilms, J Antimicrob Chemother. août, vol.68, issue.8, pp.1812-1819, 2013.

F. Szoka, F. Olson, T. Heath, W. Vail, E. Mayhew et al., Preparation of unilamellar liposomes of intermediate size (0.1-0.2 mumol) by a combination of reverse phase evaporation and extrusion through polycarbonate membranes, Biochim Biophys Acta, vol.601, issue.3, pp.559-71, 1980.

S. Devos, W. Van-putte, J. Vitse, G. Van-driessche, S. Stremersch et al., Membrane vesicle secretion and prophage induction in multidrug-resistant Stenotrophomonas maltophilia in response to ciprofloxacin stress, Environ Microbiol, vol.19, issue.10, pp.3930-3937, 2017.

A. Bauwens, L. Kunsmann, H. Karch, A. Mellmann, and M. Bielaszewska, Antibiotic-Mediated Modulations of Outer Membrane Vesicles in Enterohemorrhagic Escherichia coli O104:H4 and O157:H7, Antimicrob Agents Chemother. sept, issue.9, p.61, 2017.

D. Fleming and K. P. Rumbaugh, Approaches to Dispersing Medical Biofilms. Microorganisms, 2017.

C. B. Whitchurch, T. Tolker-nielsen, P. C. Ragas, and J. S. Mattick, Extracellular DNA Required for Bacterial Biofilm Formation, Science. 22 févr, vol.295, issue.5559, pp.1487-1487, 2002.

C. B. Waryah, K. Wells, D. Ulluwishewa, N. Chen-tan, J. Gogoi-tiwari et al., In Vitro Antimicrobial Efficacy of Tobramycin Against Staphylococcus aureus Biofilms in Combination With or Without DNase I and/or Dispersin B: A Preliminary Investigation, Microb Drug Resist Larchmt N. avr, vol.23, issue.3, pp.384-90, 2017.

D. Lebeaux, V. Leflon-guibout, J. Ghigo, and C. Beloin, In vitro activity of gentamicin, vancomycin or amikacin combined with EDTA or l-arginine as lock therapy against a wide spectrum of biofilm-forming clinical strains isolated from catheter-related infections, J Antimicrob Chemother, vol.70, issue.6, pp.1704-1716, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01378735