, A map of human genome variation from population-scale sequencing, Genomes Project Consortium, vol.467, p.1061, 2010.

H. Akaike, Information theory and an extension of the maximum likelihood principle, Selected papers of Hirotugu Akaike, p.199213, 1998.

M. T. Alam, R. A. Petit, E. K. Crispell, T. A. Thornton, K. N. Conneely et al., Dissecting vancomycinintermediate resistance in Staphylococcus aureus using genome-wide association, Genome biology and evolution, vol.6, issue.5, p.1185, 2014.

M. N. Alekshun and S. B. Levy, Molecular mechanisms of antibacterial multidrug resistance, Cell, vol.128, issue.6, p.10371050, 2007.

A. Ali-ahmad, F. Fadel, C. Sebban-kreuzer, M. Ba, G. D. Pélissier et al., Structural and functional insights into the periplasmic detector domain of the GacS histidine kinase controlling biolm formation in Pseudomonas aeruginosa, Scientic reports, vol.7, issue.1, p.11262, 2017.

K. Amsler, C. Santoro, B. Foleno, K. Bush, and R. Flamm, Comparison of broth microdilution, agar dilution, and Etest for susceptibility testing of doripenem against Gram-negative and Gram-positive pathogens, Journal of clinical microbiology, vol.48, issue.9, p.33533357, 2010.

D. I. Andersson and D. Hughes, Persistence of antibiotic resistance in bacterial populations, FEMS microbiology reviews, vol.35, issue.5, p.901911, 2011.

J. M. Andrews, Determination of minimum inhibitory concentrations, Journal of antimicrobial Chemotherapy, vol.48, issue.suppl_1, p.516, 2001.

D. Aubert, L. Poirel, J. Chevalier, S. Leotard, J. Pages et al., Oxacillinase-mediated resistance to cefepime and susceptibility to ceftazidime in Pseudomonas aeruginosa, Antimicrobial agents and chemotherapy, vol.45, issue.6, p.16151620, 2001.

J. A. Baaijens, A. Z. El-aabidine, E. Rivals, and A. Schönhuth, De novo assembly of viral quasispecies using overlap graphs, Genome research, vol.27, issue.5, p.835848, 2017.
URL : https://hal.archives-ouvertes.fr/lirmm-01693168

D. J. Balding, A tutorial on statistical methods for population association studies, Nature reviews genetics, vol.7, issue.10, p.781791, 2006.

J. A. Banas, S. Biswas, and M. Zhu, DNA methylation aects virulence gene expression in Streptococcus mutans, Applied and environmental microbiology, p.543, 2011.

A. K. Barczak, J. E. Gomez, B. B. Kaufmann, E. R. Hinson, L. Cosimi et al., RNA signatures allow rapid identication of pathogens and antibiotic susceptibilities, Proceedings of the national academy of sciences, p.201119540, 2012.

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate: a practical and powerful approach to multiple testing, Journal of the royal statistical society. Series B, p.289300, 1995.

Y. Benjamini and D. Yekutieli, The control of the false discovery rate in multiple testing under dependency, Annals of statistics, p.11651188, 2001.

D. A. Benson, M. Cavanaugh, K. Clark, I. Karsch-mizrachi, D. J. Lipman et al., Genbank. Nucleic acids research, vol.41, issue.D1, pp.36-42, 2012.

D. R. Bentley, S. Balasubramanian, H. P. Swerdlow, G. P. Smith, J. Milton et al., Accurate whole human genome sequencing using reversible terminator chemistry, Nature, vol.456, issue.7218, p.53, 2008.

R. Berk, L. Brown, A. Buja, K. Zhang, and L. Zhao, Valid post-selection inference. The annals of statistics, vol.41, p.802837, 2013.

E. Bernard, L. Jacob, J. Mairal, and J. Vert, , 2014.

, Ecient RNA isoform identication and quantication from RNA-Seq data with network ows, Bioinformatics, vol.30, issue.17, p.24472455

D. Bi, Y. Xie, C. Tai, X. Jiang, J. Zhang et al., A site-specic integrative plasmid found in Pseudomonas aeruginosa clinical isolate HS87 along with a plasmid carrying an aminoglycoside-resistant gene, PloS one, vol.11, issue.2, p.148367, 2016.

I. Birol, S. D. Jackman, C. B. Nielsen, J. Q. Qian, R. Varhol et al., De novo transcriptome assembly with ABySS, Bioinformatics, vol.25, issue.21, p.28722877, 2009.

B. Bischl, M. Lang, L. Kottho, J. Schiner, J. Richter et al., mlr: machine learning in R, Journal of machine learning research, vol.17, issue.170, p.15, 2016.

C. M. Bishop, Pattern recognition and machine learning, vol.1, 2006.

J. M. Blair, M. A. Webber, A. J. Baylay, D. O. Ogbolu, and L. J. Piddock, Molecular mechanisms of antibiotic resistance, Nature reviews microbiology, vol.13, issue.1, p.4251, 2015.

G. Blanchard, P. Neuvial, R. , and E. , Post hoc inference via joint family-wise error rate control, 2017.

J. M. Bland and D. G. Altman, Multiple signicance tests: the Bonferroni method, Bmj, issue.6973, p.170, 1995.

K. M. Borgwardt and H. Kriegel, Shortest-path kernels on graphs, Fifth IEEE international conference on data mining, p.8, 2005.

A. Boulesteix, R. De-bin, X. Jiang, and M. Fuchs, Ipf-lasso: Integrative l1-penalized regression with penalty factors for prediction based on multi-omics data. Computational and mathematical methods in medicine, 2017.

P. Bradley, N. C. Gordon, T. M. Walker, L. Dunn, S. Heys et al., Rapid antibiotic-resistance predictions from genome sequence data for Staphylococcus aureus and Mycobacterium tuberculosis, Nature communications, vol.6, p.10063, 2015.

F. J. Brandenburg, Drawing decorated graphs, 1999.

D. Branton, D. W. Deamer, A. Marziali, H. Bayley, S. A. Benner et al., The potential and challenges of nanopore sequencing, Nanoscience and technology: a collection of reviews from Nature journals, p.261268, 2010.

L. Breiman, Random forests. Machine learning, vol.45, p.532, 2001.

W. S. Bush and J. H. Moore, , 2012.

, Genome-wide association studies, PLoS computational biology, vol.8, issue.12, p.1002822

J. Butler, I. Maccallum, M. Kleber, I. A. Shlyakhter, M. K. Belmonte et al., ALLPATHS: de novo assembly of whole-genome shotgun microreads, Genome research, vol.18, issue.5, p.810820, 2008.

L. Buzdugan, M. Kalisch, A. Navarro, D. Schunk, E. Fehr et al., Assessing statistical signicance in multivariable genome wide association analysis, Bioinformatics, vol.32, issue.13, 2016.

G. Cabot, A. A. Ocampo-sosa, M. A. Domínguez, J. F. Gago, C. Juan et al., Genetic markers of widespread extensively drug-resistant (XDR) Pseudomonas aeruginosa high-risk clones. Antimicrobial agents and chemotherapy, p.1388, 2012.

G. Cabot, C. López-causapé, A. A. Ocampo-sosa, L. M. Sommer, M. Á. Domínguez et al., Deciphering the resistome of the widespread Pseudomonas aeruginosa sequence type 175 international high-risk clone through whole-genome sequencing, Antimicrobial agents and chemotherapy, vol.60, issue.12, p.74157423, 2016.

J. A. Cain, N. Solis, and S. J. Cordwell, Beyond gene expression: the impact of protein post-translational modications in bacteria, Journal of proteomics, vol.97, p.265286, 2014.

C. Camacho, G. Coulouris, V. Avagyan, N. Ma, J. Papadopoulos et al., BLAST+: architecture and applications, BMC bioinformatics, vol.10, issue.1, p.421, 2009.

A. Carattoli, E. Zankari, A. García-fernández, M. V. Larsen, O. Lund et al., In silico detection and typing of plasmids using PlasmidFinder and plasmid multilocus sequence typing, Antimicrobial agents and chemotherapy, vol.58, issue.7, p.38953903, 2014.

J. Casadesús and D. A. Low, Programmed heterogeneity: epigenetic mechanisms in bacteria, Journal of biological chemistry, vol.288, issue.20, p.1392913935, 2013.

C. C. Chang, C. C. Chow, L. C. Tellier, S. Vattikuti, S. M. Purcell et al., Second-generation PLINK: rising to the challenge of larger and richer datasets, Gigascience, vol.4, issue.1, p.7, 2015.

C. Chewapreecha, P. Marttinen, N. J. Croucher, S. J. Salter, S. R. Harris et al., Comprehensive identication of single nucleotide polymorphisms associated with beta-lactam resistance within pneumococcal mosaic genes, PLoS genetics, vol.10, issue.8, p.1004547, 2014.

R. Chikhi, A. Limasset, and P. Medvedev, Compacting de Bruijn graphs from sequencing data quickly and in low memory, Bioinformatics, vol.32, issue.12, p.208, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01395704

H. H. Cho, K. C. Kwon, S. Kim, K. , and S. H. , Correlation between virulence genotype and uoroquinolone resistance in carbapenem-resistant Pseudomonas aeruginosa, Annals of laboratory medicine, vol.34, issue.4, p.286292, 2014.

P. R. Chowdhury, M. Scott, P. Worden, P. Huntington, B. Hudson et al., Genomic islands 1 and 2 play key roles in the evolution of extensively drug-resistant ST235 isolates of Pseudomonas aeruginosa, Open biology, vol.6, issue.3, 2016.

F. Coll, R. Mcnerney, M. D. Preston, J. A. Guerra-assunção, A. Warry et al., Rapid determination of anti-tuberculosis drug resistance from whole-genome sequences, Genome medicine, vol.7, issue.1, p.51, 2015.

C. Collins and X. Didelot, A phylogenetic method to perform genome-wide association studies in microbes that accounts for population structure and recombination, PLOS Computational Biology, vol.14, issue.2, p.121, 2018.

D. R. Cox, The regression analysis of binary sequences, Journal of the royal statistical society. Series B, p.215242, 1958.

E. Cule, P. Vineis, D. Iorio, and M. , Signicance testing in ridge regression for genetic data, BMC bioinformatics, vol.12, issue.1, p.372, 2011.

J. J. Davis, S. Boisvert, T. Brettin, R. W. Kenyon, C. Mao et al., Antimicrobial resistance prediction in PATRIC and RAST, Scientic reports, vol.6, p.27930, 2016.

N. De-bruijn, A combinatorial problem, Proceedings of the koninklijke nederlandse akademie van wetenschappen. Series A, vol.49, p.758, 1946.

E. De-jong, J. A. Van-oers, A. Beishuizen, P. Vos, W. J. Vermeijden et al., Ecacy and safety of procalcitonin guidance in reducing the duration of antibiotic treatment in critically ill patients: a randomised, controlled, open-label trial, The Lancet Infectious Diseases, vol.16, issue.7, p.819827, 2016.

A. Dehman, C. Ambroise, and P. Neuvial, Performance of a blockwise approach in variable selection using linkage disequilibrium information, BMC bioinformatics, vol.16, issue.1, p.148, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01193074

B. Devlin and K. Roeder, Genomic control for association studies, Biometrics, vol.55, issue.4, p.9971004, 1999.

X. Didelot, R. Bowden, D. J. Wilson, T. E. Peto, and D. W. Crook, Transforming clinical microbiology with bacterial genome sequencing, Nature reviews genetics, vol.13, issue.9, p.601, 2012.

E. Drezen, G. Rizk, R. Chikhi, C. Deltel, C. Lemaitre et al., GATB: genome assembly & analysis tool box, Bioinformatics, vol.30, issue.20, p.29592961, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01088571

A. Drouin, S. Giguère, M. Déraspe, F. Laviolette, M. Marchand et al., Greedy biomarker discovery in the genome with applications to antimicrobial resistance, 2015.

W. M. Dunne, M. Jaillard, O. Rochas, and A. Van-belkum, Microbial genomics and antimicrobial susceptibility testing, Expert review of molecular diagnostics, vol.17, issue.3, p.257269, 2017.

I. Durieux, C. Ginevra, K. Picq, L. Attaiech, P. Juan et al., Widespread natural transformation in Legionella pneumophila clinical isolates, Proceedings of the 5th ESCMID study group for Legionella infections conference, p.22, 2018.

S. G. Earle, C. Wu, J. Charlesworth, N. Stoesser, N. C. Gordon et al., Identifying lineage eects when controlling for population structure improves power in bacterial association studies, Nature microbiology, p.16041, 2016.

D. W. Eyre, D. De-silva, K. Cole, J. Peters, M. J. Cole et al., WGS to predict antibiotic mics for Neisseria gonorrhoeae, Journal of antimicrobial chemotherapy, vol.72, issue.7, 2017.

D. Falush and R. Bowden, , 2006.

, Genome-wide association mapping in bacteria?, Trends in microbiology, vol.14, issue.8, p.353355

M. R. Farhat, B. J. Shapiro, K. J. Kieser, R. Sultana, K. R. Jacobson et al., Genomic analysis identies targets of convergent positive selection in drug-resistant Mycobacterium tuberculosis, Nature genetics, vol.45, issue.10, p.1189, 2013.

M. R. Farhat, R. Sultana, O. Iartchouk, S. Bozeman, J. Galagan et al., Genetic determinants of drug resistance in Mycobacterium tuberculosis and their diagnostic value. American journal of respiratory and critical care medicine, vol.194, p.621630, 2016.

J. Flandrois, G. Lina, and O. Dumitrescu, MUBII-TB-DB: a database of mutations associated with antibiotic resistance in Mycobacterium tuberculosis, BMC bioinformatics, vol.15, issue.1, p.107, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-00982132

M. Franz, C. T. Lopes, G. Huck, Y. Dong, O. Sumer et al., Cytoscape.js: a graph theory library for visualisation and analysis, Bioinformatics, vol.32, issue.2, p.309311, 2015.

L. Freschi, J. Jeukens, I. Kukavica-ibrulj, B. Boyle, M. Dupont et al., Clinical utilization of genomics data produced by the international Pseudomonas aeruginosa consortium, Frontiers in microbiology, vol.6, p.1036, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01352145

J. Friedman, T. Hastie, and R. Tibshirani, The elements of statistical learning, Springer series in statistics, vol.1, 2001.

J. Friedman, T. Hastie, and R. Tibshirani, Regularization paths for generalized linear models via coordinate descent, Journal of statistical software, vol.33, p.122, 2010.

P. Fuchs, A. Barry, and S. Brown, Susceptibility testing quality control studies with fosfomycin tromethamine, European journal of clinical microbiology and infectious diseases, vol.16, issue.7, p.538540, 1997.

M. Galardini, E. G. Biondi, M. Bazzicalupo, and A. Mengoni, Contiguator: a bacterial genomes nishing tool for structural insights on draft genomes, Source code for biology and medicine, vol.6, issue.1, p.11, 2011.

S. N. Gardner, T. Slezak, and B. G. Hall, kSNP 3.0: SNP detection and phylogenetic analysis of genomes without genome alignment or reference genome, Bioinformatics, issue.17, p.28772878, 2015.

E. Garrison, J. Sirén, A. M. Novak, G. Hickey, J. M. Eizenga et al., Variation graph toolkit improves read mapping by representing genetic variation in the reference, Nature biotechnology, 2018.

T. Gärtner, P. Flach, and S. Wrobel, On graph kernels: Hardness results and ecient alternatives, Learning theory and kernel machines, p.129143, 2003.

M. R. Gillings, Integrons: past, present, and future. Microbiology and molecular biology reviews, vol.78, p.257277, 2014.

J. J. Goeman, l1 penalized estimation in the Cox proportional hazards model, Biometrical journal, vol.52, issue.1, p.7084, 2010.

J. J. Goeman and A. Solari, Multiple hypothesis testing in genomics, Statistics in medicine, vol.33, issue.11, 2014.

N. Gordon, J. Price, K. Cole, R. Everitt, M. Morgan et al., Prediction of Staphylococcus aureus antimicrobial resistance by whole-genome sequencing, Journal of clinical microbiology, vol.52, issue.4, p.11821191, 2014.

C. Grangeasse, J. Stülke, and I. Mijakovic, Regulatory potential of post-translational modications in bacteria, Frontiers in microbiology, vol.6, p.500, 2015.

S. K. Gupta, B. R. Padmanabhan, S. M. Diene, R. Lopez-rojas, M. Kempf et al., ARG-ANNOT, a new bioinformatic tool to discover antibiotic resistance genes in bacterial genomes, Antimicrobial agents and chemotherapy, vol.58, issue.1, p.212220, 2014.

S. M. Gygli, S. Borrell, A. Trauner, and S. Gagneux, Antimicrobial resistance in Mycobacterium tuberculosis: mechanistic and evolutionary perspectives, FEMS microbiology reviews, vol.41, issue.3, p.354373, 2017.

D. H. Haft, M. Dicuccio, A. Badretdin, V. Brover, V. Chetvernin et al., RefSeq: an update on prokaryotic genome annotation and curation, Nucleic acids research, vol.46, issue.D1, pp.851-860, 2017.

S. Hernando-amado, F. Sanz-garcía, P. Blanco, and J. L. Martínez, Fitness costs associated with the acquisition of antibiotic resistance, Essays in biochemistry, vol.61, issue.1, p.3748, 2017.

J. A. Hindler and R. M. Humphries, Colistin MIC variability by method for contemporary clinical isolates of multidrug resistant Gram-negative bacilli, Journal of clinical microbiology, p.3385, 2013.

R. R. Hocking, The analysis and selection of variables in linear regression, Biometrics, vol.32, issue.1, p.149, 1976.

A. E. Hoerl and R. W. Kennard, Ridge regression: biased estimation for nonorthogonal problems, Technometrics, vol.12, issue.1, p.5567, 1970.

G. E. Homan, Correcting for population structure and kinship using the linear mixed model: theory and extensions, PLoS One, vol.8, issue.10, p.75707, 2013.

A. H. Holmes, L. S. Moore, A. Sundsfjord, M. Steinbakk, S. Regmi et al., Understanding the mechanisms and drivers of antimicrobial resistance, The Lancet, vol.387, p.176187, 2016.

D. C. Hooper and G. A. Jacoby, Mechanisms of drug resistance: quinolone resistance, Annals of the New York academy of sciences, vol.1354, issue.1, p.1231, 2015.

D. Illakkiam, M. Shankar, P. Ponraj, J. Rajendhran, G. et al., Genome sequencing of a mung bean plant growth promoting strain of P. aeruginosa with biocontrol ability, International journal of genomics, 2014.

. International-hapmap-consortium, The international HapMap project, Nature, vol.426, issue.6968, p.789, 2003.

Z. Iqbal, M. Caccamo, I. Turner, P. Flicek, and G. Mcvean, De novo assembly and genotyping of variants using colored de Bruijn graphs, Nature genetics, vol.44, issue.2, p.226232, 2012.

I. Consortium, Classication of staphylococcal cassette chromosome mec (SCCmec): guidelines for reporting novel SCCmec elements, Antimicrobial agents and chemotherapy, vol.53, issue.12, p.49614967, 2009.

S. D. Jackman, B. P. Vandervalk, H. Mohamadi, J. Chu, S. Yeo et al., ABySS 2.0: resourceecient assembly of large genomes using a bloom lter, Genome research, vol.27, issue.5, p.768777, 2017.

M. Jaillard, Fine mapping of antibiotic resistance determinants, 2018.
URL : https://hal.archives-ouvertes.fr/tel-02044150

M. Jaillard, S. Schicklin, A. Larue-triolet, and J. Veyrieras, A comprehensive microbial knowledge base to support the development of in vitro diagnostic solutions in infectious diseases, p.5559, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01887873

M. Jaillard, A. Van-belkum, K. C. Cady, D. Creely, D. Shortridge et al., Correlation between phenotypic antibiotic susceptibility and the resistome in Pseudomonas aeruginosa, International journal of antimicrobial agents, vol.50, issue.2, p.210218, 2017.

M. Jaillard, L. Lima, M. Tournoud, P. Mahé, A. Van-belkum et al., A fast and agnostic method for bacterial genome-wide association studies: bridging the gap between k-mers and genetic events. bioRxiv, pp.297754-137, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01920359

H. M. Kang, N. A. Zaitlen, C. M. Wade, A. Kirby, D. Heckerman et al., Ecient control of population structure in model organism association mapping, Genetics, vol.178, issue.3, p.17091723, 2008.

K. Katoh, K. Misawa, K. Kuma, and T. Miyata, MAFFT: a novel method for rapid multiple sequence alignment based on fast Fourier transform, Nucleic acids research, vol.30, issue.14, p.30593066, 2002.

A. Khaledi, M. Schniederjans, S. Pohl, R. Rainer, U. Bodenhofer et al., Transcriptome proling of antimicrobial resistance in Pseudomonas aeruginosa, Antimicrobial agents and chemotherapy, p.75, 2016.

E. Y. Klein, T. P. Van-boeckel, E. M. Martinez, S. Pant, S. Gandra et al., Global increase and geographic convergence in antibiotic consumption between, Proceedings of the national academy of sciences, p.201717295, 2000.

S. Koren and A. M. Phillippy, One chromosome, one contig: complete microbial genomes from long-read sequencing and assembly, Current opinion in microbiology, vol.23, p.110120, 2015.

V. N. Kos, M. Déraspe, R. E. Mclaughlin, J. D. Whiteaker, P. H. Roy et al., The resistome of Pseudomonas aeruginosa in relationship to phenotypic susceptibility, Antimicrobial agents and chemotherapy, 2014.

V. N. Kos, R. E. Mclaughlin, and H. A. Gardner, The elucidation of mechanisms of ceftazidime resistance among clinical isolates of Pseudomonas aeruginosa using genomic data, Antimicrobial agents and chemotherapy, p.3113, 2016.

C. U. Köser, M. T. Holden, M. J. Ellington, E. J. Cartwright, N. M. Brown et al., Rapid whole-genome sequencing for investigation of a neonatal MRSA outbreak, New England journal of medicine, vol.366, issue.24, p.2275, 2012.

P. S. Krawczyk, L. Lipinski, and A. Dziembowski, PlasFlow: predicting plasmid sequences in metagenomic data using genome signatures, Nucleic acids research, vol.46, issue.6, pp.35-35, 2018.

V. L. Kung, E. A. Ozer, and A. R. Hauser, The accessory genome of Pseudomonas aeruginosa, Microbiology and molecular biology reviews, vol.74, issue.4, p.621641, 2010.

M. Laabei, M. Recker, J. K. Rudkin, M. Aldeljawi, Z. Gulay et al., Predicting the virulence of MRSA from its genome sequence, Genome research, vol.24, issue.5, p.839849, 2014.

S. M. Lakin, C. Dean, N. R. Noyes, A. Dettenwanger, A. S. Ross et al., MEGARes: an antimicrobial resistance database for high throughput sequencing, Nucleic acids research, vol.45, issue.D1, pp.574-580, 2017.

P. Lambert, Mechanisms of antibiotic resistance in Pseudomonas aeruginosa, Journal of the royal society of medicine, vol.95, p.22, 2002.

T. Lambert, M. Ploy, C. , and P. , A spontaneous point mutation in the aac(6')-Ib' gene results in altered substrate specicity of aminoglycoside 6'-N-acetyltransferase of a Pseudomonas uorescens strain, FEMS microbiology letters, vol.115, p.297304, 1994.

B. Langmead and S. L. Salzberg, Fast gapped-read alignment with Bowtie 2, Nature methods, vol.9, issue.4, p.357359, 2012.

L. Bras, Y. Collin, O. Monjeaud, C. Lacroix, V. Rivals et al., Colib'read on galaxy: a tools suite dedicated to biological information extraction from raw NGS reads, vol.5, p.1, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01280238

H. Lee, S. Cho, H. Bang, J. Lee, G. Bai et al., Exclusive mutations related to isoniazid and ethionamide resistance among Mycobacterium tuberculosis isolates from Korea. The international journal of tuberculosis and lung disease, vol.4, p.441447, 2000.

J. A. Lees, M. Vehkala, N. Välimäki, S. R. Harris, C. Chewapreecha et al., Sequence element enrichment analysis to determine the genetic basis of bacterial phenotypes, Nature communications, vol.7, p.12797, 2016.

J. A. Lees, M. Galardini, S. D. Bentley, J. N. Weiser, C. et al., pyseer: a comprehensive tool for microbial pangenome-wide association studies, Bioinformatics, p.539, 2018.

C. Li and H. Li, Network-constrained regularization and variable selection for analysis of genomic data, Bioinformatics, vol.24, issue.9, p.11751182, 2008.

J. Li, K. Das, G. Fu, R. Li, and R. Wu, The bayesian lasso for genome-wide association studies, Bioinformatics, vol.27, issue.4, p.516523, 2010.

A. Liaw and M. Wiener, Classication and regression by randomForest, R News, vol.2, issue.3, p.1822, 2002.

C. A. Liebert, R. M. Hall, and A. O. Summers, Transposon Tn21, agship of the oating genome, Microbiology and Molecular Biology Reviews, vol.63, issue.3, p.507522, 1999.

A. Limasset, G. Rizk, R. Chikhi, P. , and P. , Fast and scalable minimal perfect hashing for massive key sets, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01566246

P. D. Lister, D. J. Wolter, H. , and N. D. , Antibacterial-resistant Pseudomonas aeruginosa: clinical impact and complex regulation of chromosomally encoded resistance mechanisms, Clinical microbiology reviews, vol.22, issue.4, 2009.

J. Liu, K. Wang, S. Ma, and J. Huang, Accounting for linkage disequilibrium in genome-wide association studies: a penalized regression method, Statistics and its interface, vol.6, issue.1, p.99, 2013.

Y. Liu, Y. Wang, T. R. Walsh, L. Yi, R. Zhang et al., Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: a microbiological and molecular biological study. The Lancet infectious diseases, vol.16, p.161168, 2016.

F. Llinares-lópez, D. G. Grimm, D. A. Bodenham, U. Gieraths, M. Sugiyama et al., Genome-wide detection of intervals of genetic heterogeneity associated with complex traits, Bioinformatics, issue.12, pp.240-249, 2015.

R. Lockhart, J. Taylor, R. J. Tibshirani, and R. Tibshirani, A signicance test for the lasso, Annals of statistics, vol.42, issue.2, p.413, 2014.

N. J. Loman, C. Constantinidou, J. Z. Chan, M. Halachev, M. Sergeant et al., High-throughput bacterial genome sequencing: an embarrassment of choice, a world of opportunity, Nature reviews microbiology, vol.10, issue.9, p.599, 2012.

F. D. Lowy, Antimicrobial resistance: the example of Staphylococcus aureus, Journal of clinical investigation, vol.111, issue.9, p.1265, 2003.

A. Macpherson, S. P. Otto, and S. L. Nuismer, Keeping pace with the red queen: Identifying the genetic basis of susceptibility to infectious disease, Genetics, vol.208, issue.2, p.779789, 2018.

T. C. Mah and G. A. O'toole, Mechanisms of biolm resistance to antimicrobial agents, Trends in microbiology, vol.9, issue.1, p.3439, 2001.

J. Mairal and B. Yu, Supervised feature selection in graphs with path coding penalties and network ows. The journal of machine learning research, vol.14, p.24492485, 2013.

T. A. Manolio, F. S. Collins, N. J. Cox, D. B. Goldstein, L. A. Hindor et al., Finding the missing heritability of complex diseases, Nature, issue.7265, p.747, 2009.

G. Marçais and C. Kingsford, A fast, lock-free approach for ecient parallel counting of occurrences of k-mers, Bioinformatics, vol.27, issue.6, p.764770, 2011.

T. Marschall, M. Marz, T. Abeel, L. Dijkstra, B. E. Dutilh et al., Computational pan-genomics: status, promises and challenges, Briengs in bioinformatics, p.89, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01390478

J. L. Martínez and F. Rojo, Metabolic regulation of antibiotic resistance, FEMS microbiology reviews, vol.35, issue.5, p.768789, 2011.

J. L. Martínez, A. Fajardo, L. Garmendia, A. Hernandez, J. F. Linares et al., A global view of antibiotic resistance, FEMS microbiology reviews, vol.33, issue.1, p.4465, 2008.

N. Masuda, E. Sakagawa, S. Ohya, N. Gotoh, H. Tsujimoto et al., Substrate specicities of MexAB-OprM, MexCD-OprJ, and MexXY-oprM eux pumps in Pseudomonas aeruginosa, Antimicrobial agents and chemotherapy, vol.44, issue.12, p.33223327, 2000.

D. Mazel, Integrons: agents of bacterial evolution, Nature Reviews Microbiology, vol.4, issue.8, p.608, 2006.

A. G. Mcarthur, N. Waglechner, F. Nizam, A. Yan, M. A. Azad et al., The comprehensive antibiotic resistance database, Antimicrobial agents and chemotherapy, p.419, 2013.

P. Mccullagh, Regression models for ordinal data, Journal of the royal statistical society. Series B (Methodological), p.109142, 1980.

B. Mckinney and N. Pajewski, Six degrees of epistasis: statistical network models for GWAS, Frontiers in genetics, vol.2, p.109, 2012.

N. Meinshausen, L. Meier, and P. Bühlmann, P-values for high-dimensional regression, Journal of the american statistical association, vol.104, issue.488, p.16711681, 2009.

M. Meyerson, S. Gabriel, and G. Getz, Advances in understanding cancer genomes through second-generation sequencing, Nature reviews genetics, vol.11, issue.10, p.685, 2010.

S. T. Micek, R. G. Wunderink, M. H. Kollef, C. Chen, J. Rello et al., An international multicenter retrospective study of Pseudomonas aeruginosa nosocomial pneumonia: impact of multidrug resistance, Crit Care, vol.19, issue.219, p.1186, 2015.

A. Mlynarczyk, G. Mlynarczyk, J. , and J. , The genome of Staphylococcus aureus: a review, Zentralblatt für Bakteriologie, vol.287, issue.4, p.277314, 1998.

D. Moradigaravand, M. Palm, A. Farewell, V. Mustonen, J. Warringer et al., Precise prediction of antibiotic resistance in Escherichia coli from full genome sequences. bioRxiv, p.338194, 2018.

J. L. Murray, T. Kwon, E. M. Marcotte, and M. Whiteley, Intrinsic antimicrobial resistance determinants in the superbug Pseudomonas aeruginosa, Mbio, vol.6, issue.6, p.160315, 2015.

W. S. Noble, How does multiple testing correction work?, Nature biotechnology, vol.27, issue.12, 2009.

E. I. Olekhnovich, A. T. Vasilyev, V. I. Ulyantsev, E. S. Kostryukova, and A. V. Tyakht, MetaCherchant: analyzing genomic context of antibiotic resistance genes in gut microbiota, Bioinformatics, vol.34, issue.3, p.434444, 2017.

A. Oliver, X. Mulet, C. López-causapé, J. , and C. , The increasing threat of Pseudomonas aeruginosa high-risk clones, Drug resistance updates, vol.21, p.4159, 2015.

B. D. Ondov, T. J. Treangen, P. Melsted, A. B. Mallonee, N. H. Bergman et al., Mash: fast genome and metagenome distance estimation using MinHash, Genome biology, vol.17, issue.1, p.132, 2016.

J. O'neill, Tackling drug-resistant infections globally: nal report and recommendations. Review on antimicrobial resistance, 2016.

R. Overbeek, R. Olson, G. D. Pusch, G. J. Olsen, J. J. Davis et al., The SEED and the rapid annotation of microbial genomes using subsystems technology (RAST), Nucleic acids research, vol.42, issue.D1, pp.206-214, 2013.

A. J. Page, C. A. Cummins, M. Hunt, V. K. Wong, S. Reuter et al., Roary: rapid large-scale prokaryote pan genome analysis, Bioinformatics, issue.22, p.36913693, 2015.

M. J. Pallen, N. J. Loman, P. , and C. W. , High-throughput sequencing and clinical microbiology: progress, opportunities and challenges, Current opinion in microbiology, vol.13, issue.5, p.625631, 2010.

J. C. Palomino and A. Martin, Drug resistance mechanisms in Mycobacterium tuberculosis, Antibiotics, vol.3, issue.3, p.317340, 2014.

B. Paten, A. M. Novak, J. M. Eizenga, and E. Garrison, Genome graphs and the evolution of genome inference, Genome research, vol.27, issue.5, p.665676, 2017.

Y. Peng, H. C. Leung, S. Yiu, C. , and F. Y. , IDBAa practical iterative de Bruijn graph de novo assembler, Annual international conference on research in computational molecular biology, p.426440, 2010.

Y. Peng, H. C. Leung, S. Yiu, C. , and F. Y. , IDBA-UD: a de novo assembler for single-cell and metagenomic sequencing data with highly uneven depth, Bioinformatics, vol.28, issue.11, p.14201428, 2012.

P. Peterlongo, N. Schnel, N. Pisanti, M. Sagot, and V. Lacroix, Identifying SNPs without a reference genome by comparing raw reads, International symposium on string processing and information retrieval, p.147158, 2010.
URL : https://hal.archives-ouvertes.fr/inria-00514887

P. A. Pevzner, H. Tang, and M. S. Waterman, An Eulerian path approach to DNA fragment assembly, Proceedings of the national academy of sciences, vol.98, p.97489753, 2001.

J. Pirnay, F. Bilocq, B. Pot, P. Cornelis, M. Zizi et al., Pseudomonas aeruginosa population structure revisited, PLoS one, vol.4, issue.11, p.7740, 2009.

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, vol.5, issue.8, p.12245, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-01126607

A. Popescu, A. L. Harper, M. Trick, I. Bancroft, and K. T. Huber, A novel and fast approach for population structure inference using kernel-PCA and optimization (PSIKO), Genetics, p.114, 2014.

R. A. Power, J. Parkhill, and T. Oliveira, Microbial genome-wide association studies: lessons from human GWAS, Nature reviews genetics, vol.18, issue.1, p.4150, 2017.

A. L. Price, N. J. Patterson, R. M. Plenge, M. E. Weinblatt, N. A. Shadick et al., Principal components analysis corrects for stratication in genome-wide association studies, Nature genetics, vol.38, issue.8, p.904, 2006.

J. K. Pritchard, M. Stephens, N. A. Rosenberg, and P. Donnelly, Association mapping in structured populations. The american journal of human genetics, vol.67, p.170181, 2000.

S. Purcell, B. Neale, K. Todd-brown, L. Thomas, M. A. Ferreira et al., PLINK: a tool set for whole-genome association and population-based linkage analyses, The american journal of human genetics, vol.81, issue.3, p.559575, 2007.

L. Qi, H. Li, C. Zhang, B. Liang, J. Li et al., Relationship between antibiotic resistance, biolm formation, and biolm-specic resistance in Acinetobacter baumannii, Frontiers in microbiology, vol.7, p.483, 2016.

M. A. Quail, M. Smith, P. Coupland, T. D. Otto, S. R. Harris et al., A tale of three next generation sequencing platforms: comparison of Ion Torrent, Pacic Biosciences and Illumina MiSeq sequencers, BMC genomics, vol.13, issue.1, p.341, 2012.

A. Rahman, I. Hallgrímsdóttir, M. B. Eisen, and L. Pachter, Association mapping from sequencing reads using k-mers, 2017.

A. Rahman, I. Hallgrímsdóttir, M. Eisen, and L. Pachter, Association mapping from sequencing reads using k-mers. eLife, vol.7, p.32920, 2018.

T. D. Read and R. C. Massey, Characterizing the genetic basis of bacterial phenotypes using genome-wide association studies: a new direction for bacteriology, Genome medicine, vol.6, issue.11, p.109, 2014.

K. Reinert, T. H. Dadi, M. Ehrhardt, H. Hauswedell, S. Mehringer et al., The SeqAn C++ template library for ecient sequence analysis: a resource for programmers, Journal of biotechnology, vol.261, p.157168, 2017.

L. B. Reller, M. Weinstein, J. H. Jorgensen, and M. J. Ferraro, Antimicrobial susceptibility testing: a review of general principles and contemporary practices, Clinical infectious diseases, vol.49, issue.11, p.17491755, 2009.

J. M. Rothberg, W. Hinz, T. M. Rearick, J. Schultz, W. Mileski et al., An integrated semiconductor device enabling non-optical genome sequencing, Nature, vol.475, issue.7356, p.348, 2011.

W. Rowe, K. S. Baker, D. Verner-jereys, C. Baker-austin, J. J. Ryan et al., Search engine for antimicrobial resistance: a cloud compatible pipeline and web interface for rapidly detecting antimicrobial resistance genes directly from sequence data, PLoS one, vol.10, issue.7, p.133492, 2015.

G. A. Sacomoto, J. Kielbassa, R. Chikhi, R. Uricaru, P. Antoniou et al., KISSPLICE: de novo calling alternative splicing events from RNA-seq data, BMC bioinformatics, vol.13, p.5, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00784407

T. Seemann, Prokka: rapid prokaryotic genome annotation, Bioinformatics, vol.30, issue.14, p.20682069, 2014.

E. Setakis, H. Stirnadel, and D. J. Balding, Logistic regression protects against population structure in genetic association studies, Genome research, vol.16, issue.2, p.290296, 2006.

S. K. Sheppard, X. Didelot, G. Meric, A. Torralbo, K. A. Jolley et al., , 2013.

, Genome-wide association study identies vitamin B5 biosynthesis as a host specicity factor in Campylobacter, Proceedings of the national academy of sciences, vol.110, p.1192311927

M. Slatkin, Linkage disequilibriumunderstanding the evolutionary past and mapping the medical future, Nature reviews genetics, vol.9, issue.6, p.477, 2008.

J. M. Stanton, Galton, Pearson, and the peas: a brief history of linear regression for statistics instructors, Journal of statistics education, vol.9, issue.3, 2001.

M. Stitson, J. Weston, A. Gammerman, V. Vovk, and V. Vapnik, Theory of support vector machines, vol.117, p.188191, 1996.

N. Stoesser, E. Batty, D. Eyre, M. Morgan, D. Wyllie et al., Predicting antimicrobial susceptibilities for Escherichia coli and Klebsiella pneumoniae isolates using whole genomic sequence data, Journal of antimicrobial chemotherapy, vol.68, issue.10, p.22342244, 2013.

S. Tasoulis, L. Cheng, N. Välimäki, N. J. Croucher, S. R. Harris et al., Random projection based clustering for population genomics, 2014 IEEE International Conference on, p.675682, 2014.

C. M. Thomas and K. M. Nielsen, Mechanisms of, and barriers to, horizontal gene transfer between bacteria, Nature reviews microbiology, vol.3, issue.9, p.711, 2005.

T. Thornton and M. S. Mcpeek, ROADTRIPS: case-control association testing with partially or completely unknown population and pedigree structure, The american journal of human genetics, vol.86, issue.2, p.172184, 2010.

R. Tibshirani, Regression shrinkage and selection via the lasso, Journal of the Royal Statistical Society. Series B (Methodological), p.267288, 1996.

E. Toprak, A. Veres, J. Michel, R. Chait, D. L. Hartl et al., Evolutionary paths to antibiotic resistance under dynamically sustained drug selection, Nature genetics, vol.44, issue.1, p.101, 2012.

M. Tournoud and P. Mahé, Predicting bacterial resistance from whole-genome sequences using k-mers and stability selection, BMC Bioinformatics, 2018.

F. Tran and J. Q. Boedicker, Genetic cargo and bacterial species set the rate of vesicle-mediated horizontal gene transfer, Scientic reports, vol.7, issue.1, p.8813, 2017.

H. Traore, K. Fissette, I. Bastian, M. Devleeschouwer, P. et al., Detection of rifampicin resistance in Mycobacterium tuberculosis isolates from diverse countries by a commercial line probe assay as an initial indicator of multidrug resistance. The international journal of tuberculosis and lung disease, vol.4, p.481484, 2000.

N. Tuite, K. Reddington, T. Barry, A. Zumla, E. et al., Rapid nucleic acid diagnostics for the detection of antimicrobial resistance in Gram-negative bacteria: is it time for a paradigm shift, Journal of antimicrobial chemotherapy, vol.69, issue.7, p.17291733, 2014.

, UniProt: the universal protein knowledgebase, Nucleic acids research, vol.45, issue.D1, pp.158-169, 2017.

R. Uricaru, G. Rizk, V. Lacroix, E. Quillery, O. Plantard et al., Reference-free detection of isolated SNPs, Nucleic acids research, vol.43, issue.2, pp.11-11, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01083715

A. Van-belkum and W. M. Dunne, Next generation antimicrobial susceptibility testing, Journal of clinical microbiology, p.313, 2013.

A. Van-belkum, L. B. Soriaga, M. C. Lafave, S. Akella, J. Veyrieras et al., Phylogenetic distribution of CRISPR-Cas systems in antibiotic-resistant Pseudomonas aeruginosa, mBio, vol.6, issue.6, p.179615, 2015.

J. Vert, K. Tsuda, and B. Schölkopf, A primer on kernel methods. Kernel methods in computational biology, vol.47, pp.3570-141, 2004.

T. M. Walker, T. A. Kohl, S. V. Omar, J. Hedge, C. D. Elias et al., Whole-genome sequencing for prediction of Mycobacterium tuberculosis drug susceptibility and resistance: a retrospective cohort study. The Lancet infectious diseases, vol.15, p.11931202, 2015.

C. Walsh, Antibiotics: actions, origins, resistance, 2003.

A. R. Wattam, J. J. Davis, R. Assaf, S. Boisvert, T. Brettin et al., Improvements to PATRIC, the all-bacterial bioinformatics database and analysis resource center, Nucleic acids research, vol.45, issue.D1, pp.535-542, 2016.

Q. Wei, S. Tarighi, A. Dötsch, S. Häussler, M. Müsken et al., Phenotypic and genome-wide analysis of an antibiotic-resistant small colony variant (SCV) of Pseudomonas aeruginosa, PloS one, vol.6, issue.12, p.29276, 2011.

H. Westh, D. Hougaard, J. Vuust, R. , and V. , Prevalence of erm gene classes in erythromycin-resistant Staphylococcus aureus strains isolated between 1959 and 1988, Antimicrobial agents and chemotherapy, vol.39, issue.2, p.369373, 1995.

C. Widmer, C. Lippert, O. Weissbrod, N. Fusi, C. Kadie et al., Further improvements to linear mixed models for genome-wide association studies, 2014.

L. Wiehlmann, G. Wagner, N. Cramer, B. Siebert, P. Gudowius et al., Population structure of Pseudomonas aeruginosa, Proceedings of the national academy of sciences, vol.104, p.81018106, 2007.

S. S. Wilks, The large-sample distribution of the likelihood ratio for testing composite hypotheses. The annals of mathematical statistics, vol.9, p.6062, 1938.

A. Witney, K. Gould, C. Pope, F. Bolt, N. Stoker et al., Genome sequencing and characterization of an extensively drug-resistant sequence type 111 serotype O12 hospital outbreak strain of Pseudomonas aeruginosa, Clinical Microbiology and Infection, vol.20, issue.10, pp.609-618, 2014.

P. Woerther, C. Angebault, H. Jacquier, O. Clermont, A. El-mniai et al., Characterization of fecal ESBL-producing Escherichia coli in a remote community during a long term period, Antimicrobial agents and chemotherapy, p.848, 2013.

, Antimicrobial resistance: global report on surveillance, World Health Organization, 2014.

, Global tuberculosis report, 2017.

L. L. Wright, J. F. Turton, K. L. Hopkins, D. M. Livermore, and N. Woodford, Genetic environment of metallo-?lactamase genes in Pseudomonas aeruginosa isolates from the UK, Journal of antimicrobial chemotherapy, vol.70, issue.12, p.32503258, 2015.

T. T. Wu, Y. F. Chen, T. Hastie, E. Sobel, and K. Lange, , 2009.

, Genome-wide association analysis by lasso penalized logistic regression, Bioinformatics, vol.25, issue.6, p.714721

P. Yanardag and S. Vishwanathan, Deep graph kernels, Proceedings of the 21th ACM SIGKDD international conference on knowledge discovery and data mining, p.13651374, 2015.
DOI : 10.1145/2783258.2783417

S. Yoon, Z. Xuan, V. Makarov, K. Ye, and J. Sebat, Sensitive and accurate detection of copy number variants using read depth of coverage, 2009.

J. Yu, G. Pressoir, W. H. Briggs, I. V. Bi, M. Yamasaki et al., A unied mixed-model method for association mapping that accounts for multiple levels of relatedness, Nature genetics, vol.38, issue.2, p.203, 2006.

E. Zankari, H. Hasman, R. S. Kaas, A. M. Seyfarth, Y. Agersø et al., Genotyping using wholegenome sequencing is a realistic alternative to surveillance based on phenotypic antimicrobial susceptibility testing, Journal of antimicrobial chemotherapy, vol.68, issue.4, p.771777, 2012.

E. Zankari, H. Hasman, S. Cosentino, M. Vestergaard, S. Rasmussen et al., Identication of acquired antimicrobial resistance genes, Journal of antimicrobial chemotherapy, vol.67, issue.11, p.26402644, 2012.

D. Zerbino and E. Birney, Velvet: algorithms for de novo short read assembly using de Bruijn graphs, Genome research, p.74492, 2008.

H. Zhang, D. Li, L. Zhao, J. Fleming, N. Lin et al., Genome sequencing of 161 Mycobacterium tuberculosis isolates from China identies genes and intergenic regions associated with drug resistance, Nature genetics, vol.45, issue.10, p.12551260, 2013.

W. Zhang, J. Chen, Y. Yang, Y. Tang, J. Shang et al., A practical comparison of de novo genome assembly software tools for next-generation sequencing technologies, PloS one, vol.6, issue.3, p.17915, 2011.

S. Zhao, G. Tyson, Y. Chen, C. Li, S. Mukherjee et al., Whole genome sequencing analysis accurately predicts antimicrobial resistance phenotypes in Campylobacter, Applied and environmental microbiology, p.2873, 2015.

X. Zhou and M. Stephens, , 2012.

, Genome-wide ecient mixed-model analysis for association studies, Nature genetics, vol.44, issue.7, p.821824

X. Zhou and M. Stephens, Ecient multivariate linear mixed-model algorithms for genome-wide association studies, Nature methods, vol.11, issue.4, p.407, 2014.
DOI : 10.1038/nmeth.2848

URL : http://europepmc.org/articles/pmc4211878?pdf=render

H. Zou and T. Hastie, Regularization and variable selection via the elastic net, Journal of the Royal Statistical Society: Series B (Statistical Methodology), vol.67, issue.2, p.301320, 2005.