D. Rios-covian, P. Ruas-madiedo, A. Margolles, M. Gueimonde, C. De-los-reyes-gavilan et al., Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health, Frontiers in Microbiology, vol.24, p.185, 2016.
DOI : 10.1016/j.anaerobe.2013.09.012

R. Canani, M. Costanzo, L. Leone, M. Pedata, R. Meli et al., Potential beneficial effects of butyrate in intestinal and extraintestinal diseases, World Journal of Gastroenterology, vol.17, issue.12, pp.1519-1547, 2011.
DOI : 10.3748/wjg.v17.i12.1519

E. Quevrain, M. Maubert, C. Michon, F. Chain, R. Marquant et al., , a commensal bacterium deficient in Crohn???s disease, Gut, vol.107, issue.3, pp.415-440, 2016.
DOI : 10.1073/pnas.0904055107

H. Sokol, B. Pigneur, L. Watterlot, O. Lakhdari, L. Bermudez-humaran et al., Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients, Proceedings of the National Academy of Sciences, vol.41, issue.4, pp.16731-16737, 2008.
DOI : 10.1136/gut.41.4.487

URL : https://hal.archives-ouvertes.fr/hal-00652961

S. Miquel, R. Martin, A. Lashermes, M. Gillet, M. Meleine et al., Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models. Sci Rep, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01263911

O. Rossi, L. Van-berkel, F. Chain, T. Khan, M. Taverne et al., Faecalibacterium prausnitzii A2-165 has a high capacity to induce IL-10 in human and murine dendritic cells and modulates T cell responses, Scientific Reports, vol.70, issue.1, p.18507, 2016.
DOI : 10.1128/AEM.70.10.5810-5817.2004

URL : https://hal.archives-ouvertes.fr/hal-01306730

Y. Cao, J. Shen, and Z. Ran, Association between Faecalibacterium prausnitzii reduction and inflammatory bowel disease: a meta-analysis and systematic review of the literature, Gastroenterol Res Pract, vol.2014, p.872725, 2014.

H. Sokol, P. Seksik, J. Furet, O. Firmesse, I. Nion-larmurier et al., Low counts of Faecalibacterium prausnitzii in colitis microbiota, Inflammatory Bowel Diseases, vol.40, issue.8, pp.1183-1192, 2009.
DOI : 10.1128/JCM.40.12.4423-4427.2002

URL : https://hal.archives-ouvertes.fr/hal-00657435

S. Mills, F. Shanahan, C. Stanton, C. Hill, A. Coffey et al., Movers and shakers, Gut Microbes, vol.64, issue.1, pp.4-16, 2013.
DOI : 10.1371/journal.pone.0017447

S. Minot, R. Sinha, J. Chen, H. Li, S. Keilbaugh et al., The human gut virome: Inter-individual variation and dynamic response to diet, Genome Research, vol.21, issue.10, pp.1616-1641, 2011.
DOI : 10.1101/gr.122705.111

J. Bondy-denomy and A. Davidson, When a virus is not a parasite: the beneficial effects of prophages on bacterial fitness, Journal of Microbiology, vol.191, issue.3, pp.235-277, 2014.
DOI : 10.1128/JB.00797-08

H. Brussow, C. Canchaya, W. K. Hardt, and J. Frunzke, Phages and the evolution of bacterial pathogens: from genomic rearrangements to lysogenic conversion Impact of spontaneous prophage induction on the fitness of bacterial populations and host-microbe interactions, Microbiol Mol Biol Rev. J Bacteriol, vol.68197, issue.33, pp.560-602410, 2004.

M. De-paepe, M. Leclerc, C. Tinsley, and M. Petit, Bacteriophages: an underestimated role in human and animal health?, Frontiers in Cellular and Infection Microbiology, vol.181, issue.9, p.39, 2014.
DOI : 10.1086/315239

URL : https://hal.archives-ouvertes.fr/hal-01204359

P. Manrique, M. Dills, and M. Young, The Human Gut Phage Community and Its Implications for Health and Disease, Viruses, vol.159, issue.6, 2017.
DOI : 10.1186/2049-2618-1-3

M. Kim, E. Park, S. Roh, and J. Bae, ABSTRACT, Applied and Environmental Microbiology, vol.77, issue.22, pp.8062-70, 2011.
DOI : 10.1128/AEM.06331-11

URL : https://hal.archives-ouvertes.fr/hal-01058407

P. Manrique, B. Bolduc, S. Walk, J. Van-der-oost, W. De-vos et al., Healthy human gut phageome, Proceedings of the National Academy of Sciences, vol.5, issue.37, pp.10400-10405, 2016.
DOI : 10.1371/journal.pone.0035053

J. Norman, S. Handley, M. Baldridge, L. Droit, C. Liu et al., Disease-Specific Alterations in the Enteric Virome in Inflammatory Bowel Disease, Cell, vol.160, issue.3, pp.447-60, 2015.
DOI : 10.1016/j.cell.2015.01.002

J. Ren, N. Ahlgren, Y. Lu, J. Fuhrman, and F. Sun, VirFinder: a novel k-mer based tool for identifying viral sequences from assembled metagenomic data, Microbiome, vol.11, issue.1, p.69, 2017.
DOI : 10.1186/1471-2105-11-119

L. Benevides, S. Burman, R. Martin, V. Robert, M. Thomas et al., New Insights into the Diversity of the Genus Faecalibacterium, Frontiers in Microbiology, vol.46, p.1790, 2017.
DOI : 10.1099/00207713-46-1-341

URL : https://hal.archives-ouvertes.fr/hal-01617957

Y. Zhou, Y. Liang, K. Lynch, J. Dennis, and D. Wishart, PHAST: A Fast Phage Search Tool, Nucleic Acids Research, vol.113, issue.2, pp.347-52, 2011.
DOI : 10.1016/S0092-8674(03)00276-9

D. Arndt, J. Grant, A. Marcu, T. Sajed, A. Pon et al., PHASTER: a better, faster version of the PHAST phage search tool, Nucleic Acids Research, vol.6, issue.W1, pp.16-21, 2016.
DOI : 10.1093/bioinformatics/bts565

J. Kiss, M. Szabo, and F. Olasz, Site-specific recombination by the DDE family member mobile element IS30 transposase, Proceedings of the National Academy of Sciences, vol.271, issue.5255, pp.15000-15005, 2003.
DOI : 10.1126/science.271.5255.1592

R. Lavigne, D. Seto, P. Mahadevan, H. Ackermann, and A. Kropinski, Unifying classical and molecular taxonomic classification: analysis of the Podoviridae using BLASTP-based tools, Research in Microbiology, vol.159, issue.5, pp.406-420, 2008.
DOI : 10.1016/j.resmic.2008.03.005

E. Adriaenssens and J. Brister, How to name and classify your phage: an informal guide, Viruses, vol.9, issue.4, 2017.

S. Minot, S. Grunberg, G. Wu, J. Lewis, and F. Bushman, Hypervariable loci in the human gut virome, Proceedings of the National Academy of Sciences, vol.334, issue.6052, pp.3962-3968, 2012.
DOI : 10.1126/science.1208344

R. Heler, L. Marraffini, and D. Bikard, Adapting to new threats: the generation of memory by CRISPR-Cas immune systems, Molecular Microbiology, vol.50, issue.1, pp.1-9, 2014.
DOI : 10.1016/j.molcel.2013.05.001

A. Lopes, P. Tavares, M. Petit, R. Guerois, and S. Zinn-justin, Automated classification of tailed bacteriophages according to their neck organization, BMC Genomics, vol.15, issue.1, p.1027, 2014.
DOI : 10.1016/j.jmb.2013.02.012

URL : https://hal.archives-ouvertes.fr/hal-01204418

C. Hulo, P. Masson, L. Mercier, P. Toussaint, and A. , A structured annotation frame for the transposable phages: A new proposed family ???Saltoviridae??? within the Caudovirales, Virology, vol.477, pp.155-63, 2015.
DOI : 10.1016/j.virol.2014.10.009

A. Toussaint, Transposable Mu-like phages in Firmicutes: new instances of divergence generating retroelements, Research in Microbiology, vol.164, issue.4, pp.281-288, 2013.
DOI : 10.1016/j.resmic.2013.01.008

H. Delattre, O. Souiai, K. Fagoonee, R. Guerois, and M. Petit, Phagonaute: A web-based interface for phage synteny browsing and protein function prediction, Virology, vol.496, pp.42-50, 2016.
DOI : 10.1016/j.virol.2016.05.007

URL : https://hal.archives-ouvertes.fr/hal-01357336

H. Guo, D. Arambula, P. Ghosh, and J. Miller, Diversity-generating Retroelements in Phage and Bacterial Genomes, Microbiology Spectrum, vol.2, issue.6, 2014.
DOI : 10.1128/microbiolspec.MDNA3-0029-2014

M. Liu, R. Deora, S. Doulatov, M. Gingery, F. Eiserling et al., Reverse Transcriptase-Mediated Tropism Switching in Bordetella Bacteriophage, Science, vol.295, issue.5562, pp.2091-2095, 2002.
DOI : 10.1126/science.1067467

B. Paul, D. Burstein, C. Castelle, S. Handa, D. Arambula et al., Retroelement-guided protein diversification abounds in vast lineages of Bacteria and Archaea, Nature Microbiology, vol.2, p.17045, 2017.
DOI : 10.1093/sysbio/syq010

T. Schillinger and N. Zingler, The low incidence of diversity-generating retroelements in sequenced genomes, Mobile Genetic Elements, vol.2, issue.6, pp.287-91, 2012.
DOI : 10.1073/pnas.1105613108

J. Barr, R. Auro, M. Furlan, K. Whiteson, M. Erb et al., Bacteriophage adhering to mucus provide a non-host-derived immunity, Proceedings of the National Academy of Sciences, vol.66, issue.12, pp.10771-10777, 2013.
DOI : 10.1016/0022-2836(89)90464-6

B. Allet, Mu insertion duplicates a 5 base pair sequence at the host inserted site, Cell, vol.16, issue.1, pp.123-132, 1979.
DOI : 10.1016/0092-8674(79)90193-4

J. Garneau, F. Depardieu, L. Fortier, D. Bikard, and M. Monot, PhageTerm: a tool for fast and accurate determination of phage termini and packaging mechanism using next-generation sequencing data, Scientific Reports, vol.18, issue.1, p.8292, 2017.
DOI : 10.1186/s12864-017-3744-0

URL : https://hal.archives-ouvertes.fr/pasteur-01613364

J. Weitz, T. Poisot, J. Meyer, C. Flores, S. Valverde et al., Phage???bacteria infection networks, Trends in Microbiology, vol.21, issue.2, pp.82-91, 2013.
DOI : 10.1016/j.tim.2012.11.003

M. De-leeuw, M. Baron, A. Brenner, and A. Kushmaro, Genome Analysis of a Novel Broad Host Range Proteobacteria Phage Isolated from a Bioreactor Treating Industrial Wastewater, Genes, vol.32, issue.1, 2017.
DOI : 10.3389/fmicb.2014.00599

K. Malki, A. Kula, K. Bruder, E. Sible, T. Hatzopoulos et al., Bacteriophages isolated from Lake Michigan demonstrate broad host-range across several bacterial phyla, Virology Journal, vol.11, issue.1, p.164, 2015.
DOI : 10.1111/j.1462-2920.2009.02030.x

S. Kim, K. Ryu, D. Biswas, and J. Ahn, Survival, prophage induction, and invasive properties of lysogenic Salmonella Typhimurium exposed to simulated gastrointestinal conditions, Archives of Microbiology, vol.47, issue.9, pp.655-664, 2014.
DOI : 10.1111/j.1472-765X.2008.02429.x

M. De-paepe, L. Tournier, E. Moncaut, O. Son, P. Langella et al., Carriage of ?? Latent Virus Is Costly for Its Bacterial Host due to Frequent Reactivation in Monoxenic Mouse Intestine, PLOS Genetics, vol.77, issue.2, p.1005861, 2016.
DOI : 10.1371/journal.pgen.1005861.s006

M. Diard, E. Bakkeren, J. Cornuault, K. Moor, A. Hausmann et al., spp., Science, vol.66, issue.6330, pp.1211-1216, 2017.
DOI : 10.1016/j.cub.2014.07.028

URL : https://hal.archives-ouvertes.fr/hal-01607431

L. Bobay, M. Touchon, and E. Rocha, Manipulating or Superseding Host Recombination Functions: A Dilemma That Shapes Phage Evolvability, PLoS Genetics, vol.18, issue.1, p.1003825, 2013.
DOI : 10.1371/journal.pgen.1003825.s010

URL : https://hal.archives-ouvertes.fr/pasteur-01374955

M. De-paepe, G. Hutinet, O. Son, J. Amarir-bouhram, S. Schbath et al., Temperate Phages Acquire DNA from Defective Prophages by Relaxed Homologous Recombination: The Role of Rad52-Like Recombinases, PLoS Genetics, vol.57, issue.5, p.1004181, 2014.
DOI : 10.1371/journal.pgen.1004181.s012

URL : https://hal.archives-ouvertes.fr/hal-01134590

M. Touchon, A. Bernheim, and E. Rocha, Genetic and life-history traits associated with the distribution of prophages in bacteria, The ISME Journal, vol.41, issue.11, pp.2744-54, 2016.
DOI : 10.1371/journal.pcbi.0030005

F. Martinez-hernandez, O. Fornas, L. Gomez, M. Bolduc, B. et al., Single-virus genomics reveals hidden cosmopolitan and abundant viruses, Nature Communications, vol.5, p.15892, 2017.
DOI : 10.1038/ismej.2010.168

J. Krumsiek, R. Arnold, and T. Rattei, Gepard: a rapid and sensitive tool for creating dotplots on genome scale, Bioinformatics, vol.22, issue.15, pp.1026-1034, 2007.
DOI : 10.1093/bioinformatics/btl277

A. Biswas, R. Staals, S. Morales, P. Fineran, and C. Brown, CRISPRDetect: A flexible algorithm to define CRISPR arrays, BMC Genomics, vol.5, issue.1, p.356, 2016.
DOI : 10.1371/journal.pone.0008926

A. Bolger, M. Lohse, and B. Usadel, Trimmomatic: a flexible trimmer for Illumina sequence data, Bioinformatics, vol.18, issue.15, pp.2114-2134, 2014.
DOI : 10.1101/gr.074492.107

A. Reyes, L. Blanton, S. Cao, G. Zhao, M. Manary et al., Gut DNA viromes of Malawian twins discordant for severe acute malnutrition, Proceedings of the National Academy of Sciences, vol.27, issue.3, pp.11941-11947, 2015.
DOI : 10.1128/AEM.00062-07

A. Bankevich, S. Nurk, D. Antipov, A. Gurevich, M. Dvorkin et al., SPAdes: A New Genome Assembly Algorithm and Its Applications to Single-Cell Sequencing, Journal of Computational Biology, vol.19, issue.5, pp.455-77, 2012.
DOI : 10.1089/cmb.2012.0021

I. Milne, G. Stephen, M. Bayer, P. Cock, L. Pritchard et al., Using Tablet for visual exploration of second-generation sequencing data, Briefings in Bioinformatics, vol.38, issue.4, pp.193-202, 2013.
DOI : 10.1093/nar/gkp874

B. Chassaing, G. Srinivasan, M. Delgado, A. Young, A. Gewirtz et al., Fecal lipocalin 2, a sensitive and broadly dynamic non-invasive biomarker for intestinal inflammation):e44328. ? We accept pre-submission inquiries ? Our selector tool helps you to find the most relevant journal ? We provide round the clock customer support ? Convenient online submission ? Thorough peer review ? Inclusion in PubMed and all major indexing services ? Maximum visibility for your research Submit your manuscript at www.biomedcentral, com/submit Submit your next manuscript to BioMed Central and we will help you at every step, 2012.