A. Bird, Perceptions of epigenetics, Nature, vol.447, pp.396-398, 2007.

M. A. Sánchez-romero, I. Cota, and J. Casadesús, DNA methylation in bacteria: from the methyl group to the methylome, Curr. Opin. Microbiol, vol.25, pp.9-16, 2015.

J. Beaulaurier, E. E. Schadt, and G. Fang, Deciphering bacterial epigenomes using modern sequencing technologies, Nat. Rev. Genet, vol.20, p.157, 2019.

K. Vasu and V. Nagaraja, Diverse functions of restriction-modification systems in addition to cellular defense. Microbiol, Mol. Biol. Rev. MMBR, vol.77, pp.53-72, 2013.

A. S. Seshasayee, P. Singh, and S. Krishna, Context-dependent conservation of DNA methyltransferases in bacteria, Nucleic Acids Res, vol.40, pp.7066-7073, 2012.

J. Casadesús and D. Low, Epigenetic gene regulation in the bacterial world. Microbiol, Mol. Biol. Rev, vol.70, pp.830-856, 2006.

M. J. Blow, T. A. Clark, C. G. Daum, A. M. Deutschbauer, A. Fomenkov et al., The epigenomic landscape of prokaryotes, PLoS Genet, vol.12, p.1005854, 2016.

J. Krebes, R. D. Morgan, B. Bunk, C. Spröer, K. Luong et al., The complex methylome of the human gastric pathogen Helicobacter pylori, Nucleic Acids Res, vol.42, pp.2415-2432, 2014.

I. A. Murray, T. A. Clark, R. D. Morgan, M. Boitano, B. P. Anton et al., The methylomes of six bacteria, Nucleic Acids Research, vol.40, issue.22, pp.11450-11462, 2012.

M. L. Bendall, K. Luong, K. M. Wetmore, M. Blow, J. Korlach et al., Exploring the roles of DNA methylation in the metal-reducing bacterium Shewanella oneidensis MR-1, J. Bacteriol, vol.195, pp.4966-4974, 2013.

D. Gonzalez, J. B. Kozdon, H. H. Mcadams, L. Shapiro, and J. Collier, The functions of DNA methylation by CcrM in Caulobacter crescentus: a global approach, Nucleic Acids Res, vol.42, pp.3720-3735, 2014.

K. L. Seib, J. , F. E. Tan, A. Scott, A. L. Kumar et al., Specificity of the ModA11, ModA12 and ModD1 epigenetic regulator N6-adenine DNA methyltransferases of Neisseria meningitidis, Nucleic Acids Res, vol.43, pp.4150-4162, 2015.

I. Estibariz, A. Overmann, F. Ailloud, J. Krebes, C. Josenhans et al., The core genome m5C methyltransferase JHP1050 (M.Hpy99III) plays an important role in orchestrating gene expression in Helicobacter pylori, Nucleic Acids Res, vol.47, pp.2336-2348, 2019.

D. A. Low, N. J. Weyand, and M. J. Mahan, Roles of DNA adenine methylation in regulating bacterial gene expression and virulence, Infect. Immun, vol.69, pp.7197-7204, 2001.

S. Kumar, B. C. Karmakar, D. Nagarajan, A. K. Mukhopadhyay, R. D. Morgan et al., N4-cytosine DNA methylation regulates transcription and pathogenesis in Helicobacter pylori, Nucleic Acids Res, vol.46, pp.3429-3445, 2018.

P. H. Oliveira, J. W. Ribis, E. M. Garrett, D. Trzilova, A. Kim et al., Epigenomic characterization of Clostridioides difficile finds a conserved DNA methyltransferase that mediates sporulation and pathogenesis, Nat. Microbiol, vol.5, pp.166-180, 2020.
URL : https://hal.archives-ouvertes.fr/hal-02988691

F. Costa, J. E. Hagan, J. Calcagno, M. Kane, P. Torgerson et al., Global morbidity and mortality of leptospirosis: a systematic review, PLoS Negl. Trop. Dis, vol.9, p.3898, 2015.

M. Picardeau, Virulence of the zoonotic agent of leptospirosis: still terra incognita?, Nat. Rev. Microbiol, vol.15, pp.297-307, 2017.

A. T. Vincent, O. Schiettekatte, C. Goarant, V. K. Neela, E. Bernet et al., Revisiting the taxonomy and evolution of pathogenicity of the genus Leptospira through the prism of genomics, PLoS Negl. Trop. Dis, vol.13, p.7270, 2019.
URL : https://hal.archives-ouvertes.fr/pasteur-02275485

G. L. Murray, V. Morel, G. M. Cerqueira, J. Croda, A. Srikram et al., Genome-wide transposon mutagenesis in pathogenic Leptospira species, Infect. Immun, vol.77, pp.810-816, 2009.

H. C. Ellinghausen and W. G. Mccullough, Nutrition of Leptospira pomona and growth of 13 other serotypes: fractionation of oleic albumin complex and a medium of bovine albumin and polysorbate 80, Am. J. Vet. Res, vol.26, pp.45-51, 1965.

C. J. Pappas, N. Benaroudj, and M. Picardeau, A replicative plasmid vector allows efficient complementation of pathogenic Leptospira strains, Appl. Environ. Microbiol, vol.81, pp.3176-3181, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-02536538

J. Matsunaga and D. A. Haake, cis-acting determinant limiting expression of sphingomyelinase gene sph2 in Leptospira interrogans, identified with a gfp reporter plasmid, Appl. Environ. Microbiol, vol.84, pp.2068-2086, 2018.

M. Picardeau, Conjugative transfer between Escherichia coli and Leptospira spp. as a New Genetic Tool, Appl. Environ. Microbiol, vol.74, pp.319-322, 2008.

J. D. Dukes, P. Whitley, and A. D. Chalmers, The MDCK variety pack: choosing the right strain, BMC Cell Biol, vol.12, p.43, 2011.

Q. E. Stassen, F. M. Riemers, H. Reijmerink, P. A. Leegwater, and L. C. Penning, Reference genes for reverse transcription quantitative PCR in canine brain tissue, BMC Res. Notes, vol.8, p.761, 2015.

A. Eshghi, J. Henderson, M. S. Trent, and M. Picardeau, Leptospira interrogans lpxD homologue is required for thermal acclimatization and virulence, Infect. Immun, vol.83, pp.4314-4321, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-02539055

V. S. Lioy, A. Cournac, M. Marbouty, S. Duigou, J. Mozziconacci et al., Multiscale structuring of the E. coli chromosome by nucleoid-associated and condensin proteins, Cell, vol.172, pp.771-783, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02180787

A. Cournac, H. Marie-nelly, M. Marbouty, R. Koszul, and J. Mozziconacci, Normalization of a chromosomal contact map, BMC Genomics, vol.13, p.436, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00769663

B. Langmead and S. L. Salzberg, Fast gapped-read alignment with Bowtie 2, Nat. Methods, vol.9, pp.357-359, 2012.

R. R. Wick, L. M. Judd, C. L. Gorrie, and K. E. Holt, Unicycler: resolving bacterial genome assemblies from short and long sequencing reads, PLOS Comput. Biol, vol.13, p.1005595, 2017.

P. Rice, I. Longden, and A. Bleasby, EMBOSS: the european molecular biology open software suite, Trends Genet. TIG, vol.16, pp.276-277, 2000.

R. J. Roberts, T. Vincze, J. Posfai, and D. Macelis, REBASE--enzymes and genes for DNA restriction and modification, Nucleic Acids Res, vol.35, pp.269-270, 2007.

T. Cokelaer, D. Desvillechabrol, R. Legendre, and M. Cardon, Sequana': a set of snakemake NGS pipelines, J. Open Source Softw, vol.16, p.352, 2017.

M. Martin, Cutadapt removes adapter sequences from high-throughput sequencing reads, EMBnet.journal, vol.17, pp.10-12, 2011.

B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biol, vol.10, p.25, 2009.

Y. Liao, G. K. Smyth, and W. Shi, featureCounts: an efficient general purpose program for assigning sequence reads to genomic features, Bioinforma. Oxf. Engl, vol.30, pp.923-930, 2014.

R. Development-core and . Team, R: a language and environment for statistical computing, 2020.

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, p.550, 2014.

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate: a practical and powerful approach to multiple testing, J. R. Stat. Soc. Ser. B Methodol, vol.57, pp.289-300, 1995.

K. Satou, M. Shimoji, H. Tamotsu, A. Juan, N. Ashimine et al., Complete genome sequences of low-passage virulent and high-passage avirulent variants of pathogenic Leptospira interrogans serovar Manilae strain UP-MMC-NIID, originally isolated from a patient with severe leptospirosis, determined using PacBio single-molecule real-time technology, Genome Announc, vol.3, pp.882-897, 2015.

A. Eshghi, K. Lourdault, G. L. Murray, T. Bartpho, R. W. Sermswan et al., Leptospira interrogans catalase is required for resistance to H 2 O 2 and for virulence, Infect. Immun, vol.80, pp.3892-3899, 2012.

M. Gomes-solecki, I. Santecchia, and C. Werts, Animal models of leptospirosis: of mice and hamsters, Front. Immunol, vol.8, p.58, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-02337120

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, Infect. Immun, vol.80, pp.2019-2025, 2012.

A. Eshghi, J. Becam, A. Lambert, O. Sismeiro, M. A. Dillies et al., A putative regulatory genetic locus modulates virulence in the pathogen Leptospira interrogans, Infect. Immun, vol.82, pp.2542-2552, 2014.

J. Nölling and W. M. De-vos, Identification of the CTAG-recognizing restriction-modification systems MthZI and MthFI from Methanobacterium thermoformicicum and characterization of the plasmid-encoded mthZIM gene, Nucleic Acids Res, vol.20, pp.5047-5052, 1992.

A. Wasserfallen, J. Nölling, P. Pfister, J. Reeve, and E. Conway-de-macario, Phylogenetic analysis of 18 thermophilic Methanobacterium isolates supports the proposals to create a new genus, Methanothermobacter gen. nov., and to reclassify several isolates in three species, Methanothermobacter thermautotrophicus comb. nov., Methanothermobacter wolfeii comb. nov., and Methanothermobacter marburgensis sp. nov, vol.50, pp.43-53, 2000.

M. Ouellette, J. P. Gogarten, J. Lajoie, A. M. Makkay, and R. T. Papke, Characterizing the DNA methyltransferases of Haloferax volcanii via bioinformatics, gene deletion, and SMRT sequencing, Genes, vol.9, p.129, 2018.

M. Marbouty, A. Le-gall, D. I. Cattoni, A. Cournac, A. Koh et al., Condensin-and replication-mediated bacterial chromosome folding and origin condensation revealed by Hi-C and super-resolution imaging, Mol. Cell, vol.59, pp.588-602, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01419993

A. Zhukova, L. G. Fernandes, P. Hugon, C. J. Pappas, O. Sismeiro et al.,

, Genome-wide transcriptional start site mapping and sRNA identification in the pathogen Leptospira interrogans, Front. Cell. Infect. Microbiol, vol.7, p.10

G. E. Crooks, G. Hon, J. Chandonia, and S. E. Brenner, WebLogo: a sequence logo generator, Genome Res, vol.14, pp.1188-1190, 2004.

J. D. Helmann, The extracytoplasmic function (ECF) sigma factors, Adv. Microb. Physiol, vol.46, pp.47-110, 2002.

S. Falkow, Molecular Koch's postulates applied to microbial pathogenicity, Rev. Infect. Dis, vol.10, issue.2, pp.274-276, 1988.

C. Fontana, A. Lambert, N. Benaroudj, D. Gasparini, O. Gorgette et al., Analysis of a spontaneous non-motile and avirulent mutant shows that FliM is required for full endoflagella assembly in Leptospira interrogans, PLoS One, p.152916, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01435250

E. A. Wunder, C. P. Figueira, N. Benaroudj, B. Hu, B. A. Tong et al., A novel flagellar sheath protein, FcpA, determines filament coiling, translational motility and virulence for the Leptospira spirochete, Mol. Microbiol, vol.101, pp.457-470, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-02548535

M. C. Chao, S. Zhu, S. Kimura, B. M. Davis, E. E. Schadt et al., A cytosine methytransferase modulates the cell envelope stress response in the cholera pathogen, PLos Genet, vol.11, p.1005666, 2015.

N. Hofmann, R. Wurm, and R. Wagner, The E. coli anti-sigma factor Rsd: studies on the specificity and regulation of its expression, PLoS One, vol.6, p.19235, 2011.
URL : https://hal.archives-ouvertes.fr/in2p3-00005476

M. J. Kazmierczak, M. Wiedmann, and K. J. Boor, Alternative sigma factors and their roles in bacterial virulence. Microbiol, Mol. Biol. Rev, vol.69, pp.527-543, 2005.

M. A. Llamas, A. Van-der-sar, B. C. Chu, M. Sparrius, H. J. Vogel et al., A novel extracytoplasmic function (ECF) sigma factor regulates virulence in Pseudomonas aeruginosa, PLoS Pathog, vol.5, p.1000572, 2009.

P. Rattanama, J. R. Thompson, N. Kongkerd, K. Srinitiwarawong, V. Vuddhakul et al., Sigma E regulators control hemolytic activity and virulence in a shrimp pathogenic Vibrio harveyi, PLoS One, vol.7, p.32523, 2012.

M. D. Bashyam and S. E. Hasnain, The extracytoplasmic function sigma factors: role in bacterial pathogenesis, Infect. Genet. Evol, vol.4, pp.301-308, 2004.

D. E. Fouts, M. A. Matthias, H. Adhikarla, B. Adler, L. Amorim-santos et al., What makes a bacterial species pathogenic?: comparative genomic analysis of the genus Leptospira, PLoS Negl. Trop. Dis, vol.10, p.4403, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01436457

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, pp.2164-2172, 2004.

T. L. Raivio and T. J. Silhavy, Periplasmic stress and ECF sigma factors, Annu. Rev. Microbiol, vol.55, pp.591-624, 2001.

J. Qin, Y. Y. Sheng, Z. M. Zhang, Y. Z. Shi, P. He et al., Genome-wide transcriptional analysis of temperature shift in L. interrogans serovar lai strain 56601, BMC Microbiol, vol.6, p.51, 2006.

B. Lobb, B. J. Tremblay, .. Moreno-hagelsieb, G. Doxey, and A. C. , An assessment of genome annotation coverage across the bacterial tree of life, Microb. Genomics, vol.6, p.341, 2020.

T. Fraser and P. D. Brown, Temperature and oxidative stress as triggers for virulence gene expression in pathogenic Leptospira spp, Front. Microbiol, vol.8, p.783, 2017.

P. H. Oliveira and G. Fang, Conserved DNA methyltransferases: a window into fundamental mechanisms of epigenetic regulation in bacteria, Trends Microbiol, 2020.

S. Budroni, E. Siena, J. C. Dunning-hotopp, K. L. Seib, D. Serruto et al., Neisseria meningitidis is structured in clades associated with restriction modification systems that modulate homologous recombination, Proc. Natl. Acad. Sci, vol.108, pp.4494-4499, 2011.

T. Nandi, M. T. Holden, X. Didelot, K. Mehershahi, J. A. Boddey et al., Burkholderia pseudomallei sequencing identifies genomic clades with distinct recombination, accessory, and epigenetic profiles, Genome Res, vol.25, pp.129-141, 2015.

P. H. Oliveira, M. Touchon, and E. P. Rocha, The interplay of restriction-modification systems with mobile genetic elements and their prokaryotic hosts, Nucleic Acids Res, vol.42, pp.10618-10631, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01374960

M. S. Fullmer, M. Ouellette, A. S. Louyakis, R. T. Papke, and J. P. Gogarten, The patchy distribution of restriction-modification system genes and the conservation of orphan methyltransferases in Halobacteria, Genes, vol.10, p.233, 2019.

C. Goarant, M. Picardeau, S. Morand, and K. M. Mcintyre, Leptospirosis under the bibliometrics radar: evidence for a vicious circle of neglect, Institut Pasteur user on, vol.9, p.10302, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01965923