P. N. Levett and . Leptospirosis, Clin Microbiol Rev, vol.14, issue.2, p.11292640, 2001.

A. R. Bharti, J. E. Nally, J. N. Ricaldi, M. A. Matthias, M. M. Diaz et al., Leptospirosis: A zoonotic disease of global importance, Lancet Infect Dis, vol.3, p.14652202, 2003.

D. A. Ashford, R. M. Kaiser, R. A. Spiegel, B. A. Perkins, R. S. Weyant et al., Asymptomatic infection and risk factors for leptospirosis in Nicaragua, Am J Trop Med Hyg, vol.63, issue.5-6, p.11421372, 2000.

C. A. Ganoza, M. A. Matthias, M. Saito, M. Cespedes, E. Gotuzzo et al., Asymptomatic renal colonization of humans in the peruvian Amazon by Leptospira, PLoS Negl Trop Dis, vol.4, issue.2, p.2826405, 2010.

H. Dikken and E. Kmety, Serological typing methods of leptospires, Methods in Microbiology. 11, pp.259-307, 1978.

D. J. Brenner, A. F. Kaufmann, K. R. Sulzer, A. G. Steigerwalt, F. C. Rogers et al., Further determination of DNA relatedness between serogroups and serovars in the family Leptospiraceae with a proposal for Leptospira alexanderi sp. nov. and four new Leptospira genomospecies, International journal of systematic bacteriology, vol.49, issue.2, pp.839-58, 1999.

D. J. Brenner, A. F. Kaufmann, K. R. Sulzer, A. G. Steigerwalt, F. C. Rogers et al., Further determination of DNA relatedness between serogroups and serovars in the family Leptospiraceae witha proposal for Leptospira alexanderi sp. nov. and four new Leptospira genomospecies, Int J Syst Bacteriol, vol.49, p.10319510, 1999.

P. Ramadass, B. Jarvis, R. J. Corner, D. Penny, and R. B. Marshall, Genetic characterization of pathogenic Leptospira species by DNA hybridization, Int J Syst Bacteriol, vol.42, p.1581182, 1992.

P. Ramadass, B. Jarvis, R. J. Corner, M. Cinco, and R. B. Marshall, DNA relatedness among strains of Leptospira biflexa, Int J Syst Bacteriol, vol.40, p.2397191, 1990.

P. H. Yasuda, A. G. Steigerwalt, K. R. Sulzer, A. F. Kaufmann, F. C. Rogers et al., Deoxyribonucleic acid relatedness between serogroups and serovars in the family Leptospiraceae with proposals for seven new Leptospira species, Int J Syst Bacteriol, vol.37, pp.407-422, 1987.

P. Bourhy, L. Collet, S. Brisse, and M. Picardeau, Leptospira mayottensis sp. nov., a pathogenic species of the genus Leptospira isolated from humans, Int J Syst Evol Microbiol, vol.64, pp.4061-4068, 2014.

M. Saito, S. Y. Villanueva, Y. Kawamura, K. Iida, J. Tomida et al., Leptospira idonii sp. nov., isolated from environmental water. International journal of systematic and evolutionary microbiology, vol.63, pp.2457-62, 2013.

A. I. Ko, C. Goarant, and M. Picardeau, Leptospira: the dawn of the molecular genetics era for an emerging zoonotic pathogen, Nat Rev Microbiol, vol.7, issue.10, pp.736-783, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00450871

A. Ahmed, M. P. Grobusch, P. R. Klatser, and R. A. Hartskeerl, Molecular approaches in the detection and characterization of Leptospira, J Bacteriol Parasitol, pp.5-7, 2011.

G. M. Cerqueira and M. Picardeau, A century of Leptospira strain typing, Infect Genet Evol, vol.9, pp.760-768, 2009.

P. N. Levett and L. Smythe, International Committee on Systematics of Prokaryotes Subcommittee on the taxonomy of Leptospiraceae. Minutes of the closed meeting, Int J Syst Evol Micr, vol.64, 2013.

A. F. Auch, M. Klenk, and H. P. , Digital DNA-DNA hybridization for microbial species delineation by means of genome-to-genome sequence comparison, Stand Genomic Sci, vol.2, pp.117-151, 2010.

J. Goris, K. T. Konstantinidis, J. A. Klappenbach, T. Coenye, P. Vandamme et al., DNA-DNA hybridization values and their relationship to whole-genome sequence similarities, Int J Syst Evol Micr, vol.57, pp.81-91, 2007.

Y. I. Wolf, I. Rogozin, N. Grishin, R. Tatusov, and E. Koonin, Genome trees constructed using five different approaches suggest new major bacterial clades, BMC Evol Biol, vol.1, 2001.

A. F. Auch, H. P. Klenk, and M. S. Göker, Standard operating procedure for calculating genome-to-genome distances based on high-scoring segment pairs, Stand Genomic Sci, vol.2, p.21304686, 2010.

S. Boonsilp, J. Thaipadungpanit, P. Amornchai, V. Wuthiekanun, M. S. Bailey et al., A single multilocus sequence typing (MLST) scheme for seven pathogenic Leptospira species, PLoS neglected tropical diseases, vol.7, issue.1, p.3554523, 2013.

A. Ahmed, A. S. Ferreira, and R. A. Hartskeerl, Multilocus sequence typing (MLST): markers for the traceability of pathogenic Leptospira strains, Methods in molecular biology, vol.1247, pp.349-59, 2015.

K. Nalam, A. Ahmed, S. M. Devi, P. Francalacci, M. Baig et al., Genetic affinities within a large global collection of pathogenic Leptospira: implications for strain identification and molecular epidemiology, PLoS One, vol.5, issue.8, p.2929200, 2010.

E. C. Romero, R. M. Blanco, and R. L. Galloway, Analysis of multilocus sequence typing for identification of Leptospira isolates in Brazil, J Clin Microbiol, vol.49, issue.11, p.3209071, 2011.

A. Ahmed, J. Thaipadungpanit, S. Boonsilp, V. Wuthiekanun, K. Nalam et al., Comparison of two multilocus sequence based genotyping schemes for Leptospira species, PLoS Negl Trop Dis, vol.5, issue.11, p.3210738, 2011.

S. B. Agampodi, A. C. Moreno, J. M. Vinetz, and M. A. Matthias, Utility and limitations of direct multi-locus sequence typing on qPCR-positive blood to determine infecting Leptospira strain. The American journal of tropical medicine and hygiene, vol.88, p.3541733, 2013.

Y. Chiani, P. Jacob, V. Varni, N. Landolt, M. F. Schmeling et al., Isolation and clinical sample typing of human leptospirosis cases in Argentina, Infect Genet Evol, vol.37, pp.245-51, 2016.

J. S. Lehmann, M. A. Matthias, J. M. Vinetz, and D. E. Fouts, Leptospiral pathogenomics. Pathogens, vol.3, issue.2, p.4243447, 2014.

S. X. Ren, G. Fu, X. G. Jiang, R. Zeng, Y. G. Miao et al., Unique physiological and pathogenic features of Leptospira interrogans revealed by whole-genome sequencing, Nature, vol.422, issue.6934, pp.888-93, 2003.

A. L. Nascimento, A. I. Ko, E. A. Martins, C. B. Monteiro-vitorello, P. L. Ho et al., Comparative genomics of two Leptospira interrogans serovars reveals novel insights into physiology and pathogenesis, J Bacteriol, vol.186, issue.7, p.374407, 2004.

D. M. Bulach, R. L. Zuerner, P. Wilson, T. Seemann, A. Mcgrath et al., Genome reduction in Leptospira borgpetersenii reflects limited transmission potential, Proc Natl Acad Sci, vol.103, issue.39, p.1599999, 2006.

J. N. Ricaldi, D. E. Fouts, J. D. Selengut, D. M. Harkins, K. P. Patra et al., Whole Genome Analysis of Leptospira licerasiae Provides Insight into Leptospiral Evolution and Pathogenicity, PLoS Negl Trop Dis, vol.6, issue.10, p.3493377, 2012.

M. Picardeau, D. M. Bulach, C. Bouchier, R. L. Zuerner, N. Zidane et al., Genome sequence of the saprophyte Leptospira biflexa provides insights into the evolution of Leptospira and the pathogenesis of leptospirosis, PLoS One, vol.3, issue.2, p.2229662, 2008.

L. F. Chou, Y. T. Chen, C. W. Lu, Y. C. Ko, C. Y. Tang et al., Sequence of Leptospira santarosai serovar Shermani genome and prediction of virulence-associated genes, Gene, vol.511, issue.2, pp.364-70, 2012.

M. A. Matthias, M. M. Diaz, K. J. Campos, M. Calderon, M. R. Willig et al., Diversity of bat-associated Leptospira in the Peruvian Amazon inferred by bayesian phylogenetic analysis of 16S ribosomal DNA sequences, Am J Trop Med Hyg, vol.73, issue.5, p.2270400, 2005.

P. N. Levett, Systematics of leptospiraceae. Current topics in microbiology and immunology, vol.387, pp.11-20, 2015.

A. L. Nascimento, A. I. Ko, E. A. Martins, C. B. Monteiro-vitorello, P. L. Ho et al., Comparative genomics of two Leptospira interrogans serovars reveals novel insights into physiology and pathogenesis, J Bacteriol, vol.186, issue.7, p.374407, 2004.

M. A. Matthias, J. N. Ricaldi, M. Cespedes, M. M. Diaz, R. L. Galloway et al., Human leptospirosis caused by a new, antigenically unique Leptospira associated with a Rattus species reservoir in the Peruvian Amazon, PLoS Negl Trop Dis, vol.2, issue.4, p.2271056, 2008.

A. L. Delcher, A. Phillippy, J. Carlton, and S. L. Salzberg, Fast algorithms for large-scale genome alignment and comparison, Nucleic acids research, vol.30, issue.11, pp.2478-83, 2002.

P. Central and P. , , p.117189

D. E. Fouts, E. F. Mongodin, R. E. Mandrell, W. G. Miller, D. A. Rasko et al., Major Structural Differences and Novel Potential Virulence Mechanisms from the Genomes of Multiple Campylobacter Species, PLoS Biol, vol.3, issue.1, p.539331, 2005.

D. E. Fouts, H. L. Tyler, R. T. Deboy, S. Daugherty, Q. Ren et al., Complete Genome Sequence of the N2-Fixing Broad Host Range Endophyte Klebsiella pneumoniae 342 and Virulence Predictions Verified in Mice, PLoS Genet, vol.4, issue.7, 2008.

P. Central and P. , , p.2453333

T. Davidsen, E. Beck, A. Ganapathy, R. Montgomery, N. Zafar et al., The comprehensive microbial resource, Nucleic acids research, vol.38, p.2808947, 2010.

Y. Chen, O. C. Stine, J. H. Badger, A. I. Gil, G. B. Nair et al., Comparative Genomic Analysis of Vibrio parahaemolyticus: Serotype Conversion and Virulence, BMC Genomics, vol.12, p.3130711, 2011.

R. D. Finn, J. Clements, and S. R. Eddy, HMMER web server: interactive sequence similarity searching, Nucleic acids research, vol.39, p.3125773, 2011.
DOI : 10.1093/nar/gkr367

URL : https://academic.oup.com/nar/article-pdf/39/suppl_2/W29/7628106/gkr367.pdf

J. R. Cole, Q. Wang, E. Cardenas, J. Fish, B. Chai et al., The Ribosomal Database Project: improved alignments and new tools for rRNA analysis, Nucleic acids research, vol.37, p.2686447, 2009.

E. L. Sonnhammer and V. Hollich, Scoredist: a simple and robust protein sequence distance estimator, BMC Bioinformatics, vol.6, p.1131889, 2005.

, phylipFasta-Wrapper for the Phylip Package Written in Ruby

J. Felsenstein, PHYLIP-Phylogeny Inference Package (Version 3.2), Cladistics, vol.5, pp.164-170, 1989.

J. Felsenstein and . Phylip, Phylogeny Inference Package) version 3.69. Distributed by the author, 2009.

F. Sievers, A. Wilm, D. Dineen, T. J. Gibson, K. Karplus et al., Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega, Mol Syst Biol, vol.7, p.3261699, 2011.

S. Boonsilp, J. Thaipadungpanit, P. Amornchai, V. Wuthiekanun, M. S. Bailey et al., A single multilocus sequence typing (MLST) scheme for seven pathogenic Leptospira species, PLoS Negl Trop Dis, vol.7, issue.1, p.23359622, 2013.

P. Central and P. , , p.3554523

K. Tamura, D. Peterson, N. Peterson, G. Stecher, M. Nei et al., MEGA5: molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods. Molecular biology and evolution, vol.28, p.3203626, 2011.
DOI : 10.1093/molbev/msr121

URL : https://academic.oup.com/mbe/article-pdf/28/10/2731/2954190/msr121.pdf

S. Guindon and O. Gascuel, A simple, fast, and accurate algorithm to estimate large phylogenies by maximum likelihood, Systematic biology, vol.52, issue.5, pp.696-704, 2003.

J. R. Brown, C. J. Douady, M. J. Italia, W. E. Marshall, and M. J. Stanhope, Universal trees based on large combined protein sequence data sets, Nat Genet, vol.28, issue.3, pp.281-286, 2001.
DOI : 10.1038/90129

S. R. Santos and H. Ochman, Identification and phylogenetic sorting of bacterial lineages with universally conserved genes and proteins, Environ Microbiol, vol.6, issue.7, pp.754-763, 2004.

F. D. Ciccarelli, T. Doerks, C. Von-mering, C. J. Creevey, B. Snel et al., Toward automatic reconstruction of a highly resolved tree of life, Science, vol.311, issue.5765, pp.1283-1290, 2006.

J. D. Thompson, D. G. Higgins, and T. J. Gibson, CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice, Nucleic acids research, vol.22, issue.22, p.308517, 1994.

S. Capella-gutierrez, J. M. Silla-martinez, and T. Gabaldon, trimAl: a tool for automated alignment trimming in large-scale phylogenetic analyses, Bioinformatics, vol.25, issue.15, p.2712344, 2009.

A. Stamatakis, RAxML-VI-HPC: maximum likelihood-based phylogenetic analyses with thousands of taxa and mixed models, Bioinformatics, vol.22, issue.21, 2006.

D. E. Fouts and . Phage_finder, Automated Identification and Classification of Prophage Regions in Complete Bacterial Genome Sequences, Nucleic Acids Res, vol.34, issue.20, p.17062630, 2006.

P. Central and P. , , p.1635311

J. P. Meier-kolthoff, A. F. Auch, H. P. Klenk, and M. Göker, Genome sequence-based species delimitation with confidence intervals and improved distance functions, BMC Bioinformatics, vol.14, p.60, 2012.

D. E. Fouts, L. Brinkac, E. Beck, J. Inman, and S. G. Panoct, Automated Clustering of Orthologs Using Conserved Gene Neighborhood for Pan-Genomic Analysis of Bacterial Strains and Closely Related Species, Nucleic Acids Res, vol.40, issue.22, p.3526259, 2012.

H. Tettelin, V. Masignani, M. J. Cieslewicz, C. Donati, D. Medini et al., Genome analysis of multiple pathogenic isolates of Streptococcus agalactiae: implications for the microbial

, Proc Natl Acad Sci U S A, vol.102, issue.39, p.1216834, 2005.

J. S. Hogg, F. Z. Hu, B. Janto, R. Boissy, J. Hayes et al., Characterization and modeling of the Haemophilus influenzae core and supragenomes based on the complete genomic sequences of Rd and 12 clinical nontypeable strains, Genome Biol, vol.8, issue.6, p.2394751, 2007.

D. A. Rasko, M. J. Rosovitz, G. S. Myers, E. F. Mongodin, W. F. Fricke et al., The pangenome structure of Escherichia coli: comparative genomic analysis of E. coli commensal and pathogenic isolates, J Bacteriol, vol.190, issue.20, pp.6881-93, 2008.

P. Central and P. , , p.2566221

J. J. Davie, J. Earl, S. P. De-vries, A. A. Hu, F. Z. Bootsma et al., Comparative analysis and supragenome modeling of twelve Moraxella catarrhalis clinical isolates, BMC Genomics, p.3045334, 2011.

J. Park, Y. Zhang, A. M. Buboltz, X. Zhang, S. C. Schuster et al., Comparative genomics of the classical Bordetella subspecies: the evolution and exchange of virulence-associated diversity amongst closely related pathogens, BMC Genomics, vol.13, p.3533505, 2012.

E. N. Gordienko, M. D. Kazanov, and M. S. Gelfand, Evolution of pan-genomes of Escherichia coli, Shigella spp., and Salmonella enterica, J Bacteriol, vol.195, issue.12, p.3697250, 2013.

A. Jacobsen, R. S. Hendriksen, F. M. Aaresturp, D. W. Ussery, and C. Friis, The Salmonella enterica pangenome, Microb Ecol, vol.62, issue.3, p.3175032, 2011.

P. Bourhy, H. Storck, C. Theodose, R. Olive, C. Nicolas et al., Serovar diversity of pathogenic Leptospira circulating in the French West Indies, PLoS Negl Trop Dis, vol.7, issue.3, p.3597474, 2013.

P. Hochedez, M. Escher, H. Decoussy, L. Pasgrimaud, R. Martinez et al., Outbreak of leptospirosis among canyoning participants, Euro Surveill, vol.18, issue.18, p.23725775, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-01064951

C. S. Henry, M. Dejongh, A. A. Best, P. M. Frybarger, B. Linsay et al., High-throughput generation, optimization and analysis of genome-scale metabolic models, Nat Biotechnol, vol.28, issue.9, 2010.

A. Ebrahim, J. A. Lerman, B. O. Palsson, and D. R. Hyduke, COBRApy: COnstraints-Based Reconstruction and Analysis for Python, BMC Syst Biol, vol.7, p.3751080, 2013.

J. D. Orth, I. Thiele, and B. O. Palsson, What is flux balance analysis?, Nat Biotechnol, vol.28, issue.3, p.3108565, 2010.

, Gurobi Optimization I. Gurobi Optimizer Reference Manual, 2014.

V. S. Kumar and C. D. Maranas, GrowMatch: an automated method for reconciling in silico/in vivo growth predictions, PLoS Comput Biol, vol.5, issue.3, p.2645679, 2009.

C. Fong, L. Rohmer, M. Radey, M. Wasnick, and M. J. Brittnacher, PSAT: a web tool to compare genomic neighborhoods of multiple prokaryotic genomes, BMC Bioinformatics, vol.9, p.2358893, 2008.

P. S. Chain, D. V. Grafham, R. S. Fulton, M. G. Fitzgerald, J. Hostetler et al., Genome project standards in a new era of sequencing, Science, vol.326, issue.5950, p.3854948, 2009.

K. E. Nelson, G. M. Weinstock, S. K. Highlander, K. C. Worley, H. H. Creasy et al., A catalog of reference genomes from the human microbiome, Science, vol.328, issue.5981, p.2940224, 2010.

M. Saito, S. Y. Villanueva, Y. Kawamura, K. Iida, J. Tomida et al., Leptospira idonii sp. nov., isolated from environmental water, Int J Syst Evol Microbiol, vol.63, 2013.

P. Bourhy, L. Collet, S. Brisse, and M. Picardeau, Leptospira mayottensis sp. nov., a pathogenic species of the genus Leptospira isolated from humans, Int J Syst Evol Microbiol, vol.64, pp.4061-4068, 2014.

G. Vernikos, D. Medini, D. R. Riley, and H. Tettelin, Ten years of pan-genome analyses, Current opinion in microbiology, vol.23, pp.148-54, 2014.

H. Tettelin, D. Riley, C. Cattuto, and D. Medini, Comparative genomics: the bacterial pan-genome. Current opinion in microbiology, vol.11, pp.472-479, 2008.

J. E. Nally, J. P. Whitelegge, R. Aguilera, M. M. Pereira, D. R. Blanco et al., Purification and proteomic analysis of outer membrane vesicles from a clinical isolate of Leptospira interrogans serovar Copenhageni, Proteomics, vol.5, issue.1, p.15672460, 2005.

D. A. Haake, C. Martinich, T. A. Summers, E. S. Shang, J. D. Pruetz et al., Characterization of leptospiral outer membrane lipoprotein LipL36: downregulation associated with late-log-phase growth and mammalian infection, Infection and immunity, vol.66, issue.4, p.9529084, 1998.

J. K. Barnett, D. Barnett, C. A. Bolin, T. A. Summers, E. A. Wagar et al., Expression and distribution of leptospiral outer membrane components during renal infection of hamsters, Infection and immunity, vol.67, issue.2, p.9916100, 1999.

D. A. Haake, M. K. Mazel, A. M. Mccoy, F. Milward, G. Chao et al., Leptospiral outer membrane proteins OmpL1 and LipL41 exhibit synergistic immunoprotection, Infection and immunity, vol.67, issue.12, p.10569777, 1999.

D. A. Haake, G. Chao, R. L. Zuerner, J. K. Barnett, D. Barnett et al., The leptospiral major outer membrane protein LipL32 is a lipoprotein expressed during mammalian infection, Infection and immunity, vol.68, issue.4, p.10722630, 2000.

H. Guerreiro, J. Croda, B. Flannery, M. Mazel, J. Matsunaga et al., Leptospiral proteins recognized during the humoral immune response to leptospirosis in humans, Infection and immunity, vol.69, issue.8, p.11447174, 2001.

J. Matsunaga, Y. Sanchez, X. Xu, and D. A. Haake, Osmolarity, a key environmental signal controlling expression of leptospiral proteins LigA and LigB and the extracellular release of LigA, Infection and immunity, vol.73, issue.1, p.15618142, 2005.

J. Matsunaga, M. Lo, D. M. Bulach, R. L. Zuerner, B. Adler et al., Response of Leptospira interrogans to physiologic osmolarity: relevance in signaling the environment-to-host transition, Infection and immunity, vol.75, issue.6, p.1932867, 2007.

M. Pinne, J. Matsunaga, and D. A. Haake, Leptospiral outer membrane protein microarray, a novel approach to identification of host ligand-binding proteins, J Bacteriol, vol.194, issue.22, p.3486348, 2012.

P. Ristow, P. Bourhy, F. W. Da-cruz-mcbride, C. P. Figueira, M. Huerre et al., The OmpA-like protein Loa22 is essential for leptospiral virulence, PLoS Pathog, vol.3, issue.7, 2007.

C. L. Dupont, D. B. Rusch, S. Yooseph, M. J. Lombardo, R. A. Richter et al., Genomic insights to SAR86, an abundant and uncultivated marine bacterial lineage, Isme J, vol.6, issue.6, p.3358033, 2012.

R. W. Rose, T. Bruser, J. C. Kissinger, and M. Pohlschroder, Adaptation of protein secretion to extremely highsalt conditions by extensive use of the twin-arginine translocation pathway, Molecular microbiology, vol.45, issue.4, pp.943-50, 2002.

D. H. Haft, J. D. Selengut, R. A. Richter, D. Harkins, M. K. Basu et al., TIGRFAMs and Genome Properties in 2013, Nucleic acids research, vol.41, p.3531188, 2013.

G. E. Crooks, G. Hon, J. M. Chandonia, and S. E. Brenner, WebLogo: a sequence logo generator, Genome Res, vol.14, issue.6, p.419797, 2004.

M. Drozd, D. Gangaiah, Z. Liu, and G. Rajashekara, Contribution of TAT system translocated PhoX to Campylobacter jejuni phosphate metabolism and resilience to environmental stresses, PLoS One, vol.6, issue.10, p.22028859, 2011.

P. Central and P. , , p.3197622

P. Gonnet, K. E. Rudd, and F. Lisacek, Fine-tuning the prediction of sequences cleaved by signal peptidase II: a curated set of proven and predicted lipoproteins of Escherichia coli K-12, Proteomics, vol.4, issue.6, 2004.

J. C. Setubal, M. Reis, J. Matsunaga, and D. A. Haake, Lipoprotein computational prediction in spirochaetal genomes, Microbiology, vol.152, p.2667199, 2006.

M. Paetzel, R. E. Dalbey, and N. C. Strynadka, Crystal structure of a bacterial signal peptidase apoenzyme: implications for signal peptide binding and the Ser-Lys dyad mechanism, J Biol Chem, vol.277, issue.11, pp.9512-9521, 2001.

C. J. Sigrist, E. De-castro, L. Cerutti, B. A. Cuche, N. Hulo et al., New and continuing developments at PROSITE. Nucleic acids research, vol.41, p.3531220, 2013.

F. J. Munoa, K. W. Miller, R. Beers, M. Graham, and H. C. Wu, Membrane topology of Escherichia coli prolipoprotein signal peptidase (signal peptidase II), J Biol Chem, vol.266, issue.26, pp.17667-72, 1991.

A. Bordbar, J. M. Monk, Z. A. King, and B. O. Palsson, Constraint-based models predict metabolic and associated cellular functions, Nat Rev Genet, vol.15, issue.2, 2014.

J. M. Monk, P. Charusanti, R. K. Aziz, J. A. Lerman, N. Premyodhin et al., Genome-scale metabolic reconstructions of multiple Escherichia coli strains highlight strain-specific adaptations to nutritional environments, Proc Natl Acad Sci U S A, vol.110, issue.50, p.3864276, 2013.

J. D. Woodson, J. C. Escalante-semerena, and . Cbiz, an amidohydrolase enzyme required for salvaging the coenzyme B12 precursor cobinamide in archaea, Proc Natl Acad Sci U S A, vol.101, issue.10, p.373507, 2004.

B. Adler, History of leptospirosis and leptospira. Current topics in microbiology and immunology, vol.387, pp.1-9, 2015.

B. Adler, Vaccines against leptospirosis. Current topics in microbiology and immunology, vol.387, pp.251-72, 2015.

M. Faine, B. Adler, C. Bolin, P. Perolat, and L. Leptospira, , 1999.

D. M. Bulach, T. Kalambaheti, A. De-la-pena-moctezuma, and B. Adler, Lipopolysaccharide biosynthesis in Leptospira, J Mol Microbiol Biotechnol, vol.2, issue.4, p.11075908, 2000.

A. De-la-pena-moctezuma, D. M. Bulach, and B. Adler, Genetic differences among the LPS biosynthetic loci of serovars of Leptospira interrogans and Leptospira borgpetersenii, FEMS Immunol Med Microbiol, vol.31, issue.1, 2001.

A. De-la-pena-moctezuma, D. M. Bulach, T. Kalambaheti, and B. Adler, Comparative analysis of the LPS biosynthetic loci of the genetic subtypes of serovar Hardjo: Leptospira interrogans subtype Hardjoprajitno and Leptospira borgpetersenii subtype Hardjobovis. FEMS microbiology letters, vol.177, p.10474199, 1999.

P. N. Levett, R. E. Morey, R. L. Galloway, and A. G. Steigerwalt, Leptospira broomii sp. nov., isolated from humans with leptospirosis, Int J Syst Evol Microbiol, vol.56, pp.671-674, 2006.

P. Perolat, R. J. Chappel, B. Adler, G. Baranton, D. M. Bulach et al., Leptospira fainei sp. nov., isolated from pigs in Australia, Int J Syst Bacteriol, vol.48, p.9734039, 1998.

A. M. Petersen, K. Boye, J. Blom, P. Schlichting, and K. A. Krogfelt, First isolation of Leptospira fainei serovar Hurstbridge from two human patients with Weil's syndrome, J Med Microbiol, vol.50, issue.1, p.11192512, 2001.

R. J. Chappel, D. A. Khalik, B. Adler, D. M. Bulach, S. Faine et al., Serological titres to Leptospira fainei serovar hurstbridge in human sera in Australia, Epidemiol Infect, vol.121, issue.2, p.9825801, 1998.

A. De-la-pena-moctezuma, D. M. Bulach, T. Kalambaheti, and B. Adler, Comparative analysis of the LPS biosynthetic loci of the genetic subtypes of serovar Hardjo: Leptospira interrogans subtype Hardjoprajitno and Leptospira borgpetersenii subtype Hardjobovis. FEMS microbiology letters, vol.177, pp.319-345, 1999.

D. M. Bulach, T. Kalambaheti, A. De-la-pena-moctezuma, and B. Adler, Functional analysis of genes in the rfb locus of Leptospira borgpetersenii serovar Hardjo subtype Hardjobovis, Infection and immunity, vol.68, issue.7, p.101650, 2000.

C. R. Raetz, C. Whitfield, and . Lipopolysaccharide, Annual review of biochemistry, vol.71, p.2569852, 2002.

L. Cuthbertson, I. L. Mainprize, J. H. Naismith, and C. Whitfield, Pivotal roles of the outer membrane polysaccharide export and polysaccharide copolymerase protein families in export of extracellular polysaccharides in gram-negative bacteria, Microbiology and molecular biology reviews: MMBR, vol.73, issue.1, p.2650888, 2009.

W. J. Keenleyside, M. Perry, L. Maclean, C. Poppe, and C. Whitfield, A plasmid-encoded rfbO:54 gene cluster is required for biosynthesis of the O:54 antigen in Salmonella enterica serovar Borreze, Molecular microbiology, vol.11, issue.3, pp.437-485, 1994.

J. N. Ricaldi, D. E. Fouts, J. D. Selengut, D. M. Harkins, A. Moreno et al., Whole genome analysis of Leptospira licerasiae provides insight into Leptospiral evolution and pathogenicity, PLoS Negl Trop Dis, vol.6, p.23145189, 2012.

N. L. Que-gewirth, A. A. Ribeiro, S. R. Kalb, R. J. Cotter, D. M. Bulach et al., A methylated phosphate group and four amide-linked acyl chains in Leptospira interrogans lipid A. The membrane anchor of an unusual lipopolysaccharide that activates TLR2, J Biol Chem, vol.279, issue.24, p.2556802, 2004.

J. N. Ricaldi, M. A. Matthias, J. M. Vinetz, and A. L. Lewis, Expression of sialic acids and other nonulosonic acids in Leptospira, BMC Microbiol, vol.12, p.3438082, 2012.

P. Ristow, P. Bourhy, F. W. Da-cruz-mcbride, C. P. Figueira, M. Huerre et al., The OmpA-like protein Loa22 is essential for leptospiral virulence, PLoS pathogens, vol.3, issue.7, p.1914066, 2007.

H. Brussow and R. W. Hendrix, Phage genomics: small is beautiful, Cell, vol.108, p.11792317, 2002.

F. Rohwer, Global phage diversity, Cell, vol.113, p.12705861, 2003.
DOI : 10.1016/s0092-8674(03)00276-9

URL : https://doi.org/10.1016/s0092-8674(03)00276-9

I. Saint-girons, M. D. Amouriaux, P. Baranton, and G. , First isolation of bacteriophages for a spirochaete: potential genetic tools for Leptospira, Res Microbiol, vol.141, p.2092364, 1990.

S. Girons, I. Bourhy, P. Ottone, C. Picardeau, M. Yelton et al., The LE1 bacteriophage replicates as a plasmid within Leptospira biflexa: construction of an L. biflexa-Escherichia coli shuttle vector, J Bacteriol, vol.182, p.11004167, 2000.

P. Bourhy, L. Frangeul, E. Couve, P. Glaser, S. Girons et al., Complete nucleotide sequence of the LE1 prophage from the spirochete Leptospira biflexa and characterization of its replication and partition functions, J Bacteriol, vol.187, p.15937155, 2005.

U. Dobrindt, B. Hochhut, U. Hentschel, and J. Hacker, Genomic islands in pathogenic and environmental microorganisms, Nat Rev Microbiol, vol.2, p.15100694, 2004.

J. H. Qin, Q. Zhang, Z. M. Zhang, Y. Zhong, Y. Yang et al., Identification of a novel prophage-like gene cluster actively expressed in both virulent and avirulent strains of Leptospira interrogans serovar Lai, Infect Immun, vol.76, pp.2411-2420, 2008.

P. Bourhy, L. Salaün, A. Lajus, C. Médigue, C. Boursaux-eude et al., A genomic island of the pathogen Leptospira interrogans serovar Lai can excise from its chromosome, Infect Immun, vol.75, p.17118975, 2007.

H. Ackermann, 5500 Phages examined in the electron microscope, Arch Virol, vol.152, p.17051420, 2006.

S. Girons, I. Bourhy, P. Ottone, C. Picardeau, M. Yelton et al., The LE1 bacteriophage replicates as a plasmid within leptospira biflexa: construction of an L. biflexa-escherichia coli shuttle vecto, J Bacteriol, vol.182, issue.20, p.11004167, 2000.

D. H. Haft, J. Selengut, E. F. Mongodin, and K. E. Nelson, A guild of 45 CRISPR-associated (Cas) protein families and multiple CRISPR/Cas subtypes exist in prokaryotic genomes, PLoS Comput Biol, vol.1, issue.6, p.1282333, 2005.

K. S. Makarova, D. H. Haft, R. Barrangou, S. J. Brouns, E. Charpentier et al., Evolution and classification of the CRISPR-Cas systems, Nature reviews Microbiology, vol.9, issue.6, p.3380444, 2011.

C. Camacho, G. Coulouris, V. Avagyan, N. Ma, J. Papadopoulos et al., BLAST+: architecture and applications, BMC Bioinformatics, vol.10, p.2803857, 2009.

N. N. Nickerson, V. Joag, and M. J. Mcgavin, Rapid autocatalytic activation of the M4 metalloprotease aureolysin is controlled by a conserved N-terminal fungalysin-thermolysin-propeptide domain, Mol Microbiol, vol.69, issue.6, p.18673454, 2008.

K. Kassegne, W. Hu, D. M. Ojcius, D. Sun, Y. Ge et al., Identification of collagenase as a critical virulence factor for invasiveness and transmission of pathogenic Leptospira species, J Infect Dis, vol.209, issue.7, pp.1105-1120, 2014.

W. Janwitthayanan, S. Keelawat, S. Payungporn, A. Lowanitchapat, D. Suwancharoen et al., In vivo gene expression and immunoreactivity of Leptospira collagenase, Microbiol Res, vol.168, issue.5, pp.268-72, 2013.

J. Matsunaga, M. A. Barocchi, J. Croda, T. A. Young, Y. Sanchez et al., Pathogenic Leptospira species express surface-exposed proteins belonging to the bacterial immunoglobulin superfamily, Molecular microbiology, vol.49, issue.4, p.1237129, 2003.

Y. F. Chang, C. S. Chen, R. U. Palaniappan, H. He, S. P. Mcdonough et al., Immunogenicity of the recombinant leptospiral putative outer membrane proteins as vaccine candidates, Vaccine, vol.25, issue.48, pp.8190-8197, 2007.

Z. A. Hamburger, M. S. Brown, R. R. Isberg, and P. J. Bjorkman, Crystal structure of invasin: a bacterial integrinbinding protein, Science, vol.286, issue.5438, pp.291-296, 1999.

Y. Luo, E. A. Frey, R. A. Pfuetzner, A. L. Creagh, D. G. Knoechel et al., Crystal structure of enteropathogenic Escherichia coli intimin-receptor complex, Nature, vol.405, issue.6790, 2000.

R. U. Palaniappan, Y. F. Chang, S. S. Jusuf, S. Artiushin, J. F. Timoney et al., Cloning and molecular characterization of an immunogenic LigA protein of Leptospira interrogans, Infection and immunity, vol.70, issue.11, p.130282, 2002.

A. J. Mcbride, G. M. Cerqueira, M. A. Suchard, A. N. Moreira, R. L. Zuerner et al., Genetic diversity of the Leptospiral immunoglobulin-like (Lig) genes in pathogenic Leptospira spp. Infection, genetics and evolution: journal of molecular epidemiology and evolutionary genetics in infectious diseases, vol.9, pp.196-205, 2008.

P. Central and P. , , p.2812920

G. M. Cerqueira, A. J. Mcbride, M. Picardeau, S. G. Ribeiro, A. N. Moreira et al., Distribution of the leptospiral immunoglobulin-like (lig) genes in pathogenic Leptospira species and application of ligB to typing leptospiral isolates, J Med Microbiol, vol.58, p.2887549, 2009.

J. S. Lehmann, D. E. Fouts, D. H. Haft, A. P. Cannella, J. N. Ricaldi et al., Pathogenomic Inference of Virulence-Associated Genes in Leptospira interrogans, PLoS neglected tropical diseases, vol.7, issue.10, p.2468, 2013.

A. Lambert, M. Picardeau, D. A. Haake, R. W. Sermswan, A. Srikram et al., FlaA proteins in Leptospira interrogans are essential for motility and virulence but are not required for formation of the flagellum sheath, Infection and immunity, vol.80, issue.6, p.3370569, 2012.

S. Liao, A. Sun, D. M. Ojcius, S. Wu, J. Zhao et al., Inactivation of the fliY gene encoding a flagellar motor switch protein attenuates mobility and virulence of Leptospira interrogans strain Lai, BMC Microbiol, vol.9, p.3224694, 2009.

J. P. Keener, A molecular ruler mechanism for length control of extended protein structures in bacteria, Journal of theoretical biology, vol.263, issue.4, pp.481-490, 2010.

M. J. Pallen, C. W. Penn, and R. R. Chaudhuri, Bacterial flagellar diversity in the post-genomic era, Trends Microbiol, vol.13, issue.4, pp.143-152, 2005.

C. J. Jones, M. Homma, R. M. Macnab, P. L-, and M. , proteins of the flagellar basal body of Salmonella typhimurium: gene sequences and deduced protein sequences, J Bacteriol, vol.171, issue.7, p.210140, 1989.

N. W. Charon, A. Cockburn, C. Li, J. Liu, K. A. Miller et al., The unique paradigm of spirochete motility and chemotaxis. Annual review of microbiology, vol.66, p.3771095, 2012.

J. S. Lehmann, M. A. Matthias, J. M. Vinetz, and D. E. Fouts, Leptospiral Pathogenomics. Pathogens, vol.3, issue.2, pp.280-308, 2014.

N. Ahmed, S. M. Devi, V. Mde, L. Vijayachari, P. Machang et al., Multilocus sequence typing method for identification and genotypic classification of pathogenic Leptospira species, Ann Clin Microbiol Antimicrob, vol.5, p.17121682, 2006.

J. Thaipadungpanit, V. Wuthiekanun, W. Chierakul, L. D. Smythe, W. Petkanchanapong et al., A dominant clone of Leptospira interrogans associated with an outbreak of human leptospirosis in Thailand, PLoS Negl Trop Dis, vol.1, issue.1, 2007.

M. Richter and R. Rosselló-móra, Shifting the genomic gold standard for the prokaryotic species definition, Proc Natl Acad Sci USA, vol.106, pp.19126-19157, 2009.

B. J. Tindall, R. Rosselló-móra, H. J. Busse, W. Ludwig, and P. Kämpfer, Notes on the characterization of prokaryote strains for taxonomic purposes, Int J Syst Evol Microbiol, vol.60, pp.249-66, 2010.

C. Werts, R. I. Tapping, J. C. Mathison, T. H. Chuang, V. Kravchenko et al., Leptospiral lipopolysaccharide activates cells through a TLR2-dependent mechanism, Nat Immunol, vol.2, issue.4, p.11276206, 2001.
DOI : 10.1038/86354

M. A. Nahori, E. Fournie-amazouz, N. S. Que-gewirth, V. Balloy, M. Chignard et al., Differential TLR recognition of leptospiral lipid A and lipopolysaccharide in murine and human cells, J Immunol, vol.175, issue.9, pp.6022-6053, 2005.

N. Que-gewirth, A. A. Ribeiro, S. R. Kalb, R. J. Cotter, D. M. Bulach et al., A methylated phosphate group and four amide-linked acyl chains in Leptospira interrogans lipid A, J Biol Chem, vol.279, p.15044492, 2004.

S. Viriyakosol, J. Fierer, G. D. Brown, and T. N. Kirkland, Innate immunity to the pathogenic fungus Coccidioides posadasii is dependent on TLR2 and Dectin-1. Infection and immunity, 2005.

S. Faine and L. Leptospira, , 1994.

N. A. Feasey, G. Dougan, R. A. Kingsley, R. S. Heyderman, and M. A. Gordon, Invasive non-typhoidal salmonella disease: an emerging and neglected tropical disease in Africa, Lancet, vol.379, issue.9835, p.3402672, 2012.

H. Wildschutte, D. M. Wolfe, A. Tamewitz, and J. G. Lawrence, Protozoan predation, diversifying selection, and the evolution of antigenic diversity in Salmonella, Proc Natl Acad Sci, vol.101, issue.29, p.489988, 2004.

M. Fondi and P. Lio, Genome-scale metabolic network reconstruction, Methods in molecular biology, vol.1231, pp.233-56, 2015.
DOI : 10.1007/978-1-4939-1720-4_15

M. Fondi and P. Lio, Multi -omics and metabolic modelling pipelines: Challenges and tools for systems microbiology, Microbiol Res, vol.171, pp.52-64, 2015.
DOI : 10.1016/j.micres.2015.01.003

URL : https://doi.org/10.1016/j.micres.2015.01.003

O. H. Stalheim and J. B. Wilson, Cultivation of Leptospirae. I. Nutrition of Leptospira Canicola, J Bacteriol, vol.88, p.277255, 1964.

E. Shenberg, Growth of pathogenic Leptospira in chemically defined media, J Bacteriol, vol.93, issue.5, p.276655, 1967.

T. Murachi and M. Tabata, Use of a bioreactor consisting of sequentially aligned L-glutamate dehydrogenase and L-glutamate oxidase for the determination of ammonia by chemiluminescence, Biotechnol Appl Biochem, vol.9, issue.4, p.3663333, 1987.

A. Bohmer, A. Muller, M. Passarge, P. Liebs, H. Honeck et al., A novel L-glutamate oxidase from Streptomyces endus. Purification and properties, Eur J Biochem, vol.182, issue.2, p.2737205, 1989.

J. Monk, J. Nogales, and B. O. Palsson, Optimizing genome-scale network reconstructions, Nat Biotechnol, vol.32, issue.5, p.24811519, 2014.

J. R. Roth, J. G. Lawrence, and T. A. Bobik, Cobalamin (coenzyme B12): synthesis and biological significance, vol.50, pp.137-81, 1996.

M. J. Nielsen, M. R. Rasmussen, C. B. Andersen, E. Nexo, and S. K. Moestrup, Vitamin B12 transport from food to the body's cells-a sophisticated, multistep pathway, Nature reviews Gastroenterology & hepatology, vol.9, issue.6, pp.345-54, 2012.

N. Zhao, A. S. Zhang, and C. A. Enns, Iron regulation by hepcidin, J Clin Invest, vol.123, issue.6, p.3668831, 2013.

N. C. Andrews, Disorders of iron metabolism, N Engl J Med, vol.341, issue.26, pp.1986-95, 1999.

F. E. Austin, J. T. Barbieri, R. E. Corin, K. E. Grigas, and C. D. Cox, Distribution of superoxide dismutase, catalase, and peroxidase activities among Treponema pallidum and other spirochetes, Infection and immunity, vol.33, issue.2, p.350708, 1981.

S. Li, D. M. Ojcius, S. Liao, L. Li, F. Xue et al., Replication or death: distinct fates of pathogenic Leptospira strain Lai within macrophages of human or mouse origin, Innate Immun, vol.16, issue.2, pp.80-92, 2010.

C. Toma, N. Okura, C. Takayama, and T. Suzuki, Characteristic features of intracellular pathogenic Leptospira in infected murine macrophages, Cell Microbiol, vol.13, issue.11, p.21819516, 2011.

K. V. Evangelista and J. Coburn, Leptospira as an emerging pathogen: a review of its biology, pathogenesis and host immune responses, Future Microbiology, vol.5, issue.9, p.3037011, 2010.

K. V. Evangelista, B. Hahn, E. A. Wunder, A. I. Ko, D. A. Haake et al., Identification of Cell-Binding Adhesins of Leptospira interrogans, PLoS Negl Trop Dis, vol.8, issue.10, p.4183468, 2014.

K. Evangelista, R. Franco, A. Schwab, and J. Coburn, Leptospira interrogans binds to cadherins, PLoS Negl Trop Dis, vol.8, issue.1, p.3907533, 2014.

L. G. Fernandes, M. L. Vieira, I. J. Alves, Z. M. De-morais, S. A. Vasconcellos et al., Functional and immunological evaluation of two novel proteins of Leptospira spp, Microbiology, vol.160, 2014.

G. H. Siqueira, M. V. Atzingen, I. J. Alves, Z. M. De-morais, S. A. Vasconcellos et al., Characterization of three novel adhesins of Leptospira interrogans. The American journal of tropical medicine and hygiene, vol.89, p.3854887, 2013.

R. Oliveira, R. F. Domingos, G. H. Siqueira, L. G. Fernandes, N. M. Souza et al., Adhesins of Leptospira interrogans mediate the interaction to fibrinogen and inhibit fibrin clot formation in vitro, PLoS neglected tropical diseases, vol.7, issue.8, p.24009788, 2013.

P. Central and P. , , p.3757074

N. M. Souza, M. L. Vieira, I. J. Alves, Z. M. De-morais, S. A. Vasconcellos et al., Lsa30, a novel adhesin of Leptospira interrogans binds human plasminogen and the complement regulator C4bp. Microbial pathogenesis, vol.53, p.22732096, 2012.

R. F. Domingos, M. L. Vieira, E. C. Romero, A. P. Goncales, Z. M. De-morais et al., Features of two proteins of Leptospira interrogans with potential role in host-pathogen interactions. BMC microbiology, vol.12, p.3444417, 2012.

R. Oliveira, Z. M. De-morais, A. P. Goncales, E. C. Romero, S. A. Vasconcellos et al., Characterization of novel OmpA-like protein of Leptospira interrogans that binds extracellular matrix molecules and plasminogen, PLoS One, vol.6, issue.7, p.3130794, 2011.

R. S. Mendes, V. Atzingen, M. De-morais, Z. M. Goncales, A. P. Serrano et al., The novel leptospiral surface adhesin Lsa20 binds laminin and human plasminogen and is probably expressed during infection, Infection and immunity, vol.79, issue.11, p.3257903, 2011.

M. L. Vieira, Z. M. De-morais, A. P. Goncales, E. C. Romero, S. A. Vasconcellos et al., Lsa63, a newly identified surface protein of Leptospira interrogans binds laminin and collagen IV, The Journal of infection, vol.60, issue.1, pp.52-64, 2010.

T. R. Oliveira, M. T. Longhi, A. P. Goncales, Z. M. De-morais, S. A. Vasconcellos et al., LipL53, a temperature regulated protein from Leptospira interrogans that binds to extracellular matrix molecules. Microbes and infection / Institut Pasteur, vol.12, pp.207-224, 2010.

M. T. Longhi, T. R. Oliveira, E. C. Romero, A. P. Goncales, Z. M. De-morais et al., A newly identified protein of Leptospira interrogans mediates binding to laminin, Journal of medical microbiology, vol.58, pp.1275-82, 2009.

E. A. Maciel, A. L. De-carvalho, S. F. Nascimento, R. B. De-matos, E. L. Gouveia et al., Household transmission of Leptospira infection in urban slum communities. PLoS neglected tropical diseases, vol.2, p.2270796, 2008.

R. Domingos, L. Fernandes, E. Romero, Z. De-morais, S. Vasconcellos et al., The novel Leptospira interrogans protein Lsa32 is expressed during infection and binds laminin and plasminogen. Microbiology, 2015.
DOI : 10.1099/mic.0.000041

URL : http://mic.microbiologyresearch.org/deliver/fulltext/micro/161/4/851_mic000041.pdf?itemId=/content/journal/micro/10.1099/mic.0.000041&mimeType=pdf&isFastTrackArticle=

M. L. Vieira, L. G. Fernandes, R. F. Domingos, R. Oliveira, G. H. Siqueira et al., Leptospiral extracellular matrix adhesins as mediators of pathogen-host interactions, FEMS Microbiol Lett, vol.352, issue.2, pp.129-168, 2014.

H. A. Choy, M. M. Kelley, J. Croda, J. Matsunaga, J. T. Babbitt et al., The multifunctional LigB adhesin binds homeostatic proteins with potential roles in cutaneous infection by pathogenic Leptospira interrogans, PLoS One, vol.6, issue.2, p.3036719, 2011.

C. P. Figueira, J. Croda, H. A. Choy, D. A. Haake, M. G. Reis et al., Heterologous expression of pathogen-specific genes ligA and ligB in the saprophyte Leptospira biflexa confers enhanced adhesion to cultured cells and fibronectin, BMC microbiology, vol.11, p.3133549, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-00629083

L. G. Fernandes, M. L. Vieira, K. Kirchgatter, I. J. Alves, Z. M. De-morais et al., OmpL1 is an extracellular matrix-and plasminogen-interacting protein of Leptospira spp, Infection and immunity, vol.80, issue.10, pp.3679-92, 2012.

P. Central and P. , , p.3457549

M. L. Vieira, M. V. Atzingen, R. Oliveira, R. S. Mendes, R. F. Domingos et al., Plasminogen binding proteins and plasmin generation on the surface of Leptospira spp.: the contribution to the bacteria-host interactions, J Biomed Biotechnol, p.3481863, 2012.

M. Potempa and J. Potempa, Protease-dependent mechanisms of complement evasion by bacterial pathogens, Biol Chem, vol.393, issue.9, p.3488274, 2012.

T. Meri, R. Murgia, P. Stefanel, S. Meri, and M. Cinco, Regulation of complement activation at the C3-level by serum resistant leptospires, Microb Pathog, vol.39, issue.4, pp.139-186, 2005.

A. S. Barbosa, P. A. Abreu, S. A. Vasconcellos, Z. M. Morais, A. P. Goncales et al., Immune evasion of Leptospira species by acquisition of human complement regulator C4BP, Infection and immunity, vol.77, issue.3, 2009.

P. Central and P. , , p.2643629

A. Verma, J. Hellwage, S. Artiushin, P. F. Zipfel, P. Kraiczy et al., LfhA, a novel factor H-binding protein of Leptospira interrogans, Infection and immunity, vol.74, issue.5, p.1459737, 2006.

A. S. Barbosa, D. Monaris, L. B. Silva, Z. M. Morais, S. A. Vasconcellos et al., Functional characterization of LcpA, a surface-exposed protein of Leptospira spp. that binds the human complement regulator C4BP, Infection and immunity, vol.78, issue.7, p.2897400, 2010.

H. A. Choy, Multiple activities of LigB potentiate virulence of Leptospira interrogans: inhibition of alternative and classical pathways of complement, PLoS One, vol.7, issue.7, p.3402383, 2012.

R. F. Domingos, M. L. Vieira, E. C. Romero, A. P. Goncales, Z. M. De-morais et al., Features of two proteins of Leptospira interrogans with potential role in host-pathogen interactions, BMC Microbiol, vol.12, 2012.

P. Central and P. , , p.3444417

N. M. Souza, M. L. Vieira, I. J. Alves, Z. M. De-morais, S. A. Vasconcellos et al., Lsa30, a novel adhesin of Leptospira interrogans binds human plasminogen and the complement regulator C4bp

, Microb Pathog, vol.53, issue.3-4, pp.125-159, 2012.

M. M. Castiblanco-valencia, T. R. Fraga, L. B. Silva, D. Monaris, P. A. Abreu et al., Leptospiral immunoglobulin-like proteins interact with human complement regulators factor H, FHL-1, FHR-1, and C4BP, J Infect Dis, vol.205, issue.6, pp.995-1004, 2012.

G. H. Siqueira, M. V. Atzingen, I. J. Alves, Z. M. De-morais, S. A. Vasconcellos et al., Characterization of three novel adhesins of Leptospira interrogans, Am J Trop Med Hyg, vol.89, issue.6, p.3854887, 2013.

O. A. Adekoya and I. Sylte, The thermolysin family (M4) of enzymes: therapeutic and biotechnological potential, Chem Biol Drug Des, vol.73, issue.1, pp.7-16, 2009.

A. J. Laarman, M. Ruyken, C. L. Malone, J. A. Van-strijp, A. R. Horswill et al., Staphylococcus aureus metalloprotease aureolysin cleaves complement C3 to mediate immune evasion, J Immunol, vol.186, issue.11, pp.6445-53, 2011.
DOI : 10.4049/jimmunol.1002948

URL : http://www.jimmunol.org/content/186/11/6445.full.pdf

T. R. Fraga, C. Ddos, S. Castiblanco-valencia, M. M. Hirata, I. Y. Vasconcellos et al., Immune evasion by pathogenic Leptospira strains: the secretion of proteases that directly cleave complement proteins, J Infect Dis, vol.209, issue.6, pp.876-86, 2013.

J. N. Ricaldi, M. A. Matthias, J. M. Vinetz, and A. L. Lewis, Expression of sialic acids and other nonulosonic acids in Leptospira, BMC microbiology, vol.12, p.161, 2012.

P. Central and P. , , p.3438082

D. J. Mcnally, I. C. Schoenhofen, R. S. Houliston, N. H. Khieu, D. M. Whitfield et al., CMP-pseudaminic acid is a natural potent inhibitor of PseB, the first enzyme of the pseudaminic acid pathway in Campylobacter jejuni and Helicobacter pylori, ChemMedChem, vol.3, issue.1, pp.55-64, 2007.

G. Raddi, D. R. Morado, J. Yan, D. A. Haake, X. F. Yang et al., Three-dimensional structures of pathogenic and saprophytic Leptospira species revealed by cryo-electron tomography, J Bacteriol, vol.194, issue.6, p.3294836, 2012.

I. C. Schoenhofen, E. Vinogradov, D. M. Whitfield, J. R. Brisson, and S. M. Logan, The CMP-legionaminic acid pathway in Campylobacter: biosynthesis involving novel GDP-linked precursors, Glycobiology, vol.19, issue.7, pp.715-740, 2009.

J. S. Lehmann, D. E. Fouts, D. H. Haft, A. P. Cannella, J. N. Ricaldi et al., Pathogenomic inference of virulence-associated genes in Leptospira interrogans, PLoS Negl Trop Dis, vol.7, issue.10, p.3789758, 2013.

A. Lambert, N. Takahashi, N. W. Charon, and M. Picardeau, Chemotactic behavior of pathogenic and nonpathogenic Leptospira species, Applied and environmental microbiology, vol.78, issue.23, p.3497369, 2012.

M. S. Islam, K. Takabe, S. Kudo, and S. Nakamura, Analysis of the chemotactic behaviour of Leptospira using microscopic agar-drop assay, FEMS microbiology letters, vol.356, issue.1, p.24894019, 2014.

J. Malmstrom, M. Beck, A. Schmidt, V. Lange, E. W. Deutsch et al., Proteome-wide cellular protein concentrations of the human pathogen Leptospira interrogans, Nature, vol.460, issue.7256, p.2723184, 2009.

M. Buck, M. T. Gallegos, D. J. Studholme, Y. Guo, and J. D. Gralla, The bacterial enhancer-dependent sigma 54 (sigma N) transcription factor, J Bacteriol, vol.182, issue.15, p.10894718, 2000.

A. Hubner, X. Yang, D. M. Nolen, T. G. Popova, F. C. Cabello et al., Expression of Borrelia burgdorferi OspC and DbpA is controlled by a RpoN-RpoS regulatory pathway, Proc Natl Acad Sci, vol.98, issue.22, p.60121, 2001.

X. F. Yang, S. M. Alani, and M. V. Norgard, The response regulator Rrp2 is essential for the expression of major membrane lipoproteins in Borrelia burgdorferi, Proc Natl Acad Sci U S A, vol.100, p.12949258, 2003.

A. Staro?, H. J. Sofia, S. Dietrich, L. E. Ulrich, H. Liesegang et al., The third pillar of bacterial signal transduction: classification of the extracytoplasmic function (ECF) ? factor protein family, Molecular microbiology, vol.74, issue.3, pp.557-81, 2009.

J. D. Helmann, The extracytoplasmic function (ECF) sigma factors, Adv Microb Physiol, vol.46, p.12073657, 2002.

Y. Hu, S. Kendall, N. G. Stoker, and A. Coates, The Mycobacterium tuberculosis sigJ gene controls sensitivity of the bacterium to hydrogen peroxide2004

T. D. Ho and C. D. Ellermeier, Extra cytoplasmic function ? factor activation, Current Opinion in Microbiology, vol.15, issue.2, pp.182-190, 2012.
DOI : 10.1016/j.mib.2012.01.001

URL : http://europepmc.org/articles/pmc3320685?pdf=render

C. M. Fraser, S. Casjens, W. M. Huang, G. G. Sutton, R. Clayton et al., Genomic sequence of a Lyme disease spirochaete, Borrelia burgdorferi, Nature, vol.390, p.9403685, 1997.

C. M. Fraser, S. J. Norris, G. M. Weinstock, O. White, G. G. Sutton et al., Complete genome sequence of Treponema pallidum, the syphilis spirochete, Science, vol.281, p.9665876, 1998.