M. Arumugam, J. Raes, E. Pelletier, L. Paslier, D. Yamada et al., Enterotypes of the human gut microbiome, Nature, vol.473, pp.174-80, 2011.
URL : https://hal.archives-ouvertes.fr/cea-00903625

J. Qin, R. Li, J. Raes, M. Arumugam, and K. S. Burgdorf, A human gut microbial gene catalogue established by metagenomic sequencing, Nature, vol.464, pp.59-65, 2010.
URL : https://hal.archives-ouvertes.fr/cea-00908974

T. Consortium, Structure, function and diversity of the healthy human microbiome, Nature, vol.486, pp.207-221, 2013.

H. J. Flint, K. P. Scott, P. Louis, and S. H. Duncan, The role of the gut microbiota in nutrition and health, Nat Rev Gastroenterol Hepatol, vol.9, pp.577-89, 2012.

J. R. Marchesi, D. H. Adams, F. Fava, G. D. Hermes, and G. M. Hirschfield, The gut microbiota and host health: a new clinical frontier, Gut, vol.65, pp.330-339, 2016.

N. Larsen, F. K. Vogensen, F. W. Van-den-berg, D. S. Nielsen, and A. S. Andreasen, Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults, PLoS One, vol.5, p.9085, 2010.

F. Backhed, H. Ding, T. Wang, L. V. Hooper, and G. Y. Koh, The gut microbiota as an environmental factor that regulates fat storage, Proc Natl Acad Sci U S A, vol.101, pp.15718-15741, 2004.

J. E. Koenig, A. Spor, N. Scalfone, A. D. Fricker, and J. Stombaugh, Succession of microbial consortia in the developing infant gut microbiome, Proc Natl Acad Sci U S A, vol.108, issue.1, pp.4578-85, 2011.

C. A. Thaiss, D. Zeevi, M. Levy, G. Zilberman-schapira, and J. Suez, Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis, Cell, vol.159, pp.514-543, 2014.

G. E. Flores, J. G. Caporaso, J. B. Henley, J. R. Rideout, and D. Domogala, Temporal variability is a personalized feature of the human microbiome, Genome Biol, vol.15, p.531, 2014.

S. A. Sarker, B. Berger, Y. Deng, S. Kieser, and F. Foata, Oral application of Escherichia coli bacteriophage: safety tests in healthy and diarrheal children from Bangladesh, Environ Microbiol, vol.19, pp.237-50, 2017.

A. Y. Voigt, P. I. Costea, J. R. Kultima, S. S. Li, and G. Zeller, Temporal and technical variability of human gut metagenomes, Genome Biol, vol.16, p.73, 2015.

P. Manrique, M. Dills, and M. J. Young, The Human Gut Phage Community and Its Implications for Health and Disease, Viruses, vol.9, 2017.

S. Mills, F. Shanahan, C. Stanton, C. Hill, A. Coffey et al., Movers and shakers: influence of bacteriophages in shaping the mammalian gut microbiota, Gut Microbes, vol.4, pp.4-16, 2013.

M. De-paepe, M. Leclerc, C. R. Tinsley, and M. A. Petit, Bacteriophages: an underestimated role in human and animal health?, Front Cell Infect Microbiol, vol.4, p.39, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204359

A. Reyes, M. Wu, N. P. Mcnulty, F. L. Rohwer, and J. I. Gordon, Gnotobiotic mouse model of phage-bacterial host dynamics in the human gut, Proc Natl Acad Sci U S A, vol.110, pp.20236-20277, 2013.

A. S. Waller, T. Yamada, D. M. Kristensen, J. R. Kultima, and S. Sunagawa, Classification and quantification of bacteriophage taxa in human gut metagenomes, ISME J, vol.8, pp.1391-402, 2014.

D. Maura, M. Galtier, L. Bouguenec, C. Debarbieux, and L. , Virulent bacteriophages can target O104:H4 enteroaggregative Escherichia coli in the mouse intestine, Antimicrob Agents Chemother, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-01538999

H. Brussow, Bacteriophage-host interaction: from splendid isolation into a messy reality, Curr Opin Microbiol, vol.16, pp.500-506, 2013.

C. B. Silveira and F. L. Rohwer, Piggyback-the-Winner in host-associated microbial communities, NPJ Biofilms Microbiomes, vol.2, p.16010, 2016.

M. De-paepe, L. Tournier, E. Moncaut, O. Son, P. Langella et al., Carriage of lambda Latent Virus Is Costly for Its Bacterial Host due to Frequent Reactivation in Monoxenic Mouse Intestine, PLoS Genet, vol.12, p.1005861, 2016.

J. R. Brum, B. L. Hurwitz, O. Schofield, H. W. Ducklow, and M. B. Sullivan, Seasonal time bombs: dominant temperate viruses affect Southern Ocean microbial dynamics, ISME J, vol.10, pp.437-486, 2016.

S. Houte, A. Buckling, and E. R. Westra, Evolutionary Ecology of Prokaryotic Immune Mechanisms. Microbiol Mol Biol Rev, vol.80, pp.745-63, 2016.

R. Barrangou, C. Fremaux, H. Deveau, M. Richards, and P. Boyaval, CRISPR provides acquired resistance against viruses in prokaryotes, Science, vol.315, pp.1709-1721, 2007.

K. S. Makarova, Y. I. Wolf, O. S. Alkhnbashi, F. Costa, and S. A. Shah, An updated evolutionary classification of CRISPR-Cas systems, Nat Rev Microbiol, vol.13, pp.722-758, 2015.

C. D. Yang, Y. H. Chen, H. Y. Huang, H. D. Huang, and C. P. Tseng, CRP represses the CRISPR/Cas system in Escherichia coli: evidence that endogenous CRISPR spacers impede phage P1 replication, Mol Microbiol, vol.92, pp.1072-91, 2014.

R. Edgar and U. Qimron, The Escherichia coli CRISPR system protects from lambda lysogenization, lysogens, and prophage induction, J Bacteriol, vol.192, pp.6291-6295, 2010.

W. Cenens, A. Makumi, S. K. Govers, R. Lavigne, and A. Aertsen, Viral Transmission Dynamics at Single-Cell Resolution Reveal Transiently Immune Subpopulations Caused by a Carrier State Association, PLoS Genet, vol.11, p.1005770, 2015.

J. M. Norman, S. A. Handley, M. T. Baldridge, L. Droit, and C. Y. Liu, Disease-specific alterations in the enteric virome in inflammatory bowel disease, Cell, vol.160, pp.447-60, 2015.

S. V. Owen, N. Wenner, R. Canals, A. Makumi, and D. L. Hammarlof, Characterization of the Prophage Repertoire of African Salmonella Typhimurium ST313 Reveals High Levels of Spontaneous Induction of Novel Phage BTP1, Front Microbiol, vol.8, p.235, 2017.

D. J. Mercanti, D. Carminati, J. A. Reinheimer, and A. Quiberoni, Widely distributed lysogeny in probiotic lactobacilli represents a potentially high risk for the fermentative dairy industry, Int J Food Microbiol, vol.144, pp.503-513, 2011.

M. L. Capra, J. A. Reinheimer, and A. L. Quiberoni, Characterisation of three temperate phages released from the same Lactobacillus paracasei commercial strain, International Journal of Dairy Technology, vol.63, pp.396-405, 2010.

A. L. Koch, Evolution of temperate pathogens: the bacteriophage/bacteria paradigm, Virol J, vol.4, p.121, 2007.

I. Sekirov, S. L. Russell, L. C. Antunes, and B. B. Finlay, Gut microbiota in health and disease, Physiol Rev, vol.90, pp.859-904, 2010.

H. Brussow, How stable is the human gut microbiota? And why this question matters, Environ Microbiol, vol.18, pp.2779-83, 2016.

S. Chibani-chennoufi, J. Sidoti, A. Bruttin, E. Kutter, S. Sarker et al., In vitro and in vivo bacteriolytic activities of Escherichia coli phages: implications for phage therapy, Antimicrob Agents Chemother, vol.48, pp.2558-69, 2004.

T. W. Berngruber, S. Lion, and S. Gandon, Spatial structure, transmission modes and the evolution of viral exploitation strategies, PLoS Pathog, vol.11, p.1004810, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02122352

S. Maslov and K. Sneppen, Well-temperate phage: optimal bet-hedging against local environmental collapses, Sci Rep, vol.5, p.10523, 2015.

H. T. Williams, Phage-induced diversification improves host evolvability, BMC Evol Biol, vol.13, p.17, 2013.

A. Nadeem and L. M. Wahl, Prophage as a genetic reservoir: Promoting diversity and driving innovation in the host community, Evolution, vol.71, pp.2080-2089, 2017.

J. Heinrich, M. Velleman, and H. Schuster, The tripartite immunity system of phages P1 and P7, FEMS Microbiol Rev, vol.17, pp.121-127, 1995.

A. Bailone and R. Devoret, Isolation of ultravirulent mutants of phage lambda, Virology, vol.84, pp.547-50, 1978.

J. W. Drake, Comparative rates of spontaneous mutation, Nature, vol.221, p.1132, 1969.

H. Lehnherr, E. Maguin, S. Jafri, and M. B. Yarmolinsky, Plasmid addiction genes of bacteriophage P1: doc, which causes cell death on curing of prophage, and phd, which prevents host death when prophage is retained, J Mol Biol, vol.233, pp.414-442, 1993.

, Références bibliographiques

H. Ackermann and M. S. , Virus of Prokaryotes, 1987.

E. M. Adriaenssens, R. Edwards, J. H. Nash, P. Mahadevan, and D. Seto, Integration of genomic and proteomic analyses in the classification of the Siphoviridae family, Virology, vol.477, pp.144-54, 2015.

T. Akerlund, K. Nordstrom, and R. Bernander, Analysis of cell size and DNA content in exponentially growing and stationary-phase batch cultures of Escherichia coli, J Bacteriol, vol.177, pp.6791-6798, 1995.

M. Alam, T. Midtvedt, and A. Uribe, Differential cell kinetics in the ileum and colon of germfree rats, Scand J Gastroenterol, vol.29, pp.445-51, 1994.

L. Albenberg, T. V. Esipova, C. P. Judge, K. Bittinger, and J. Chen, Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota, Gastroenterology, vol.147, p.8, 2014.

H. K. Allen, T. Looft, D. O. Bayles, S. Humphrey, and U. Y. Levine, Antibiotics in feed induce prophages in swine fecal microbiomes, MBio, vol.2, 2011.

A. Alves-de-matos, P. Timoteo, A. Roxo-rosa, and M. , Comparison of induction of B45 Helicobacter pylori prophage by acid and UV radiation, Microscopy and microanalysis, vol.19, pp.27-28, 2013.

R. I. Aminov, A. W. Walker, S. H. Duncan, H. J. Harmsen, G. W. Welling et al., Molecular diversity, cultivation, and improved detection by fluorescent in situ hybridization of a dominant group of human gut bacteria related to Roseburia spp. or Eubacterium rectale, Appl Environ Microbiol, vol.72, pp.6371-6377, 2006.

A. N. Ananthakrishnan, Epidemiology and risk factors for IBD, Nat Rev Gastroenterol Hepatol, vol.12, pp.205-222, 2015.

D. Arndt, J. R. Grant, A. Marcu, T. Sajed, and A. Pon, PHASTER: a better, faster version of the PHAST phage search tool, Nucleic Acids Res, vol.44, pp.16-21, 2016.

D. Arndt, A. Marcu, Y. Liang, D. S. Wishart, J. Raes et al., PHAST, PHASTER and PHASTEST: Tools for finding prophage in bacterial genomes, Brief Bioinform Arumugam M, vol.473, pp.174-80, 2011.

M. Asadulghani, Y. Ogura, T. Ooka, T. Itoh, and A. Sawaguchi, The defective prophage pool of Escherichia coli O157: prophage-prophage interactions potentiate horizontal transfer of virulence determinants, PLoS Pathog, vol.5, p.1000408, 2009.

O. T. Avery, C. M. Macleod, and M. Mccarty, Studies on the chemical nature of the substance inducing transformation of pneumococcal serotypes: inductions of transformation by a desoxyribonucleic acid fraction isolated from pneumococcus type III, The Journal of Experimental Medicine, vol.79, pp.137-57, 1944.

M. Avlund, S. Krishna, S. Semsey, I. B. Dodd, and K. Sneppen, Minimal gene regulatory circuits for a lysis-lysogeny choice in the presence of noise, PLoS One, vol.5, p.15037, 2010.

F. Backhed, H. Ding, T. Wang, L. V. Hooper, and G. Y. Koh, The gut microbiota as an environmental factor that regulates fat storage, Proc Natl Acad Sci U S A, vol.101, pp.15718-15741, 2004.

K. Baek, S. Svenningsen, H. Eisen, K. Sneppen, and S. Brown, Single-cell analysis of lambda immunity regulation, J Mol Biol, vol.334, pp.363-72, 2003.

I. Baess, Report on a pseudolysogenic mycobacterium and a review of the literature concerning pseudolysogeny, Acta Pathol Microbiol Scand B Microbiol Immunol, vol.79, pp.428-462, 1971.

L. Bai, J. Wang, D. Hurley, Z. Yu, and L. Wang, A novel disrupted mcr-1 gene and a lysogenized phage P1-like sequence detected from a large conjugative plasmid, cultured from a human atypical enteropathogenic Escherichia coli (aEPEC) recovered in China, J Antimicrob Chemother, vol.72, pp.1531-1564, 2017.

A. Bailone and R. Devoret, Isolation of ultravirulent mutants of phage lambda, Virology, vol.84, pp.547-50, 1978.

J. S. Baluch and R. , Correlation between UV dose requirement for lambda bacteriophage induction and lambda repressor concentration, J Virol, vol.26, pp.595-602, 1978.

D. J. Banks, B. Lei, and J. M. Musser, Prophage induction and expression of prophage-encoded virulence factors in group A Streptococcus serotype M3 strain MGAS315, Infect Immun, vol.71, pp.7079-86, 2003.

N. Barnich, F. A. Carvalho, A. L. Glasser, C. Darcha, and P. Jantscheff, CEACAM6 acts as a receptor for adherent-invasive E. coli, supporting ileal mucosa colonization in Crohn disease, J Clin Invest, vol.117, pp.1566-74, 2007.

J. J. Barondess and J. Beckwith, bor gene of phage lambda, involved in serum resistance, encodes a widely conserved outer membrane lipoprotein, J Bacteriol, vol.177, pp.1247-53, 1995.

J. J. Barr, A bacteriophages journey through the human body, Immunological Reviews, vol.279, pp.106-128, 2017.

J. J. Barr, R. Auro, M. Furlan, K. L. Whiteson, and M. L. Erb, Bacteriophage adhering to mucus provide a non-host-derived immunity, Proc Natl Acad Sci U S A, vol.110, pp.10771-10777, 2013.

J. J. Barr, R. Auro, N. Sam-soon, S. Kassegne, and G. Peters, Subdiffusive motion of bacteriophage in mucosal surfaces increases the frequency of bacterial encounters, Proc Natl Acad Sci U S A, vol.112, pp.13675-80, 2015.

R. Barrangou, C. Fremaux, H. Deveau, M. Richards, and P. Boyaval, CRISPR provides acquired resistance against viruses in prokaryotes, Science, vol.315, pp.1709-1721, 2007.

M. W. Belfort and D. , The roles of the lambda c3 gene and the Escherichia coli catabolite gene activation system in the establishment of lysogeny by bacteriophage lambda, Proc Natl Acad Sci U S A, vol.71, pp.779-82, 1974.

L. Benevides, S. Burman, R. Martin, V. Robert, and M. Thomas, New Insights into the Diversity of the Genus Faecalibacterium. Front Microbiol, vol.8, p.1790, 2017.

B. A. Bensing, I. R. Siboo, and P. M. Sullam, Proteins PblA and PblB of Streptococcus mitis, which promote binding to human platelets, are encoded within a lysogenic bacteriophage, Infect Immun, vol.69, pp.6186-92, 2001.

L. Berkowitz, B. M. Schultz, G. A. Salazar, C. Pardo-roa, and V. P. Sebastian, Impact of Cigarette Smoking on the Gastrointestinal Tract Inflammation: Opposing Effects in Crohn's Disease and Ulcerative Colitis, Front Immunol, vol.9, p.74, 2018.

G. W. Bertani and J. J. , Host controlled variation in bacterial viruses, J Bacteriol, vol.65, pp.113-134, 1953.

R. Bibiloni, M. Mangold, K. L. Madsen, R. N. Fedorak, and G. W. Tannock, The bacteriology of biopsies differs between newly diagnosed, untreated, Crohn's disease and ulcerative colitis patients, J Med Microbiol, vol.55, pp.1141-1150, 2006.

T. Billard-pomares, S. Fouteau, M. E. Jacquet, D. Roche, and V. Barbe, Characterization of a P1-like bacteriophage carrying an SHV-2 extended-spectrum beta-lactamase from an Escherichia coli strain, Antimicrob Agents Chemother, vol.58, pp.6550-6557, 2014.

E. Bille, J. Meyer, A. Jamet, D. Euphrasie, and J. P. Barnier, A virulence-associated filamentous bacteriophage of Neisseria meningitidis increases host-cell colonisation, PLoS Pathog, vol.13, p.1006495, 2017.

L. M. Bobay, E. P. Rocha, and M. Touchon, The adaptation of temperate bacteriophages to their host genomes, Mol Biol Evol, vol.30, pp.737-51, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01374945

L. M. Bobay, M. Touchon, and E. P. Rocha, Pervasive domestication of defective prophages by bacteria, Proc Natl Acad Sci U S A, vol.111, pp.12127-12159, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01374961

B. Bohannan and R. E. , Effect of resource enrichment on a chemostat community of bacteria and bacteriophage, Ecology society of america, vol.78, pp.2303-2318, 1997.

N. A. Bokulich, J. Chung, T. Battaglia, N. Henderson, and M. Jay, Antibiotics, birth mode, and diet shape microbiome maturation during early life, Sci Transl Med, vol.8, pp.343-82, 2016.

A. Bolotin, B. Quinquis, A. Sorokin, and S. D. Ehrlich, Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin, Microbiology, vol.151, pp.2551-61, 2005.

J. Bondy-denomy, A. Pawluk, K. L. Maxwell, and A. R. Davidson, Bacteriophage genes that inactivate the CRISPR/Cas bacterial immune system, Nature, vol.493, pp.429-461, 2013.

J. D. Bondy-denomy and A. R. , When a virus is not a parasite: the beneficial effects of prophages on bacterial fitness, J Microbiol, vol.52, pp.235-277, 2014.

G. Bonemann, A. Pietrosiuk, and A. Mogk, Tubules and donuts: a type VI secretion story, Mol Microbiol, vol.76, pp.815-836, 2010.

L. Bossi, J. A. Fuentes, G. Mora, and N. Figueroa-bossi, Prophage contribution to bacterial population dynamics, J Bacteriol, vol.185, pp.6467-71, 2003.

J. Boudeau, A. L. Glasser, E. Masseret, B. Joly, and A. Darfeuille-michaud, Invasive ability of an Escherichia coli strain isolated from the ileal mucosa of a patient with Crohn's disease, Infect Immun, vol.67, pp.4499-509, 1999.

T. Bouvier and P. A. , Key role of selective viral-induced mortality in determining marine bacterial community composition, Environ Microbiol, vol.9, pp.287-97, 2007.

E. F. Boyd, Bacteriophage-encoded bacterial virulence factors and phage-pathogenicity island interactions, Adv Virus Res, vol.82, pp.91-118, 2012.

F. Boyer, G. Fichant, J. Berthod, Y. Vandenbrouck, and I. Attree, Dissecting the bacterial type VI secretion system by a genome wide in silico analysis: what can be learned from available microbial genomic resources?, BMC Genomics, vol.10, p.104, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00377407

M. Breitbart, M. Haynes, S. Kelley, F. Angly, and R. A. Edwards, Viral diversity and dynamics in an infant gut, Res Microbiol, vol.159, pp.367-73, 2008.

M. R. Breitbart and F. , Here a virus, there a virus, everywhere the same virus?, Trends Microbiol, vol.13, pp.278-84, 2005.

J. Breton, N. Tennoune, N. Lucas, M. Francois, and R. Legrand, Gut Commensal E. coli Proteins Activate Host Satiety Pathways following Nutrient-Induced Bacterial Growth. Cell Metab, vol.23, pp.324-358, 2016.

M. A. Brockhurst, A. D. Morgan, A. Fenton, and A. Buckling, Experimental coevolution with bacteria and phage. The Pseudomonas fluorescens--Phi2 model system, Infect Genet Evol, vol.7, pp.547-52, 2007.

M. A. Brockhurst, P. B. Rainey, and A. Buckling, The effect of spatial heterogeneity and parasites on the evolution of host diversity, Proc Biol Sci, vol.271, pp.107-118, 2004.

F. Broecker, J. Klumpp, M. Schuppler, G. Russo, and L. Biedermann, Long-term changes of bacterial and viral compositions in the intestine of a recovered Clostridium difficile patient after fecal microbiota transplantation, Cold Spring Harb Mol Case Stud, vol.2, p.448, 2016.

F. Broecker, M. Kube, J. Klumpp, M. Schuppler, and L. Biedermann, Analysis of the intestinal microbiome of a recovered Clostridium difficile patient after fecal transplantation, Digestion, vol.88, pp.243-51, 2013.

F. Broecker, G. Russo, J. Klumpp, and K. Moelling, Stable core virome despite variable microbiome after fecal transfer, Gut Microbes, vol.8, pp.214-234, 2017.

G. W. Broussard, L. M. Oldfield, V. M. Villanueva, B. L. Lunt, E. E. Shine et al., Integrationdependent bacteriophage immunity provides insights into the evolution of genetic switches, Mol Cell, vol.49, pp.237-285, 2013.

S. P. Brown, L. Chat, L. , D. Paepe, M. Taddei et al., Ecology of microbial invasions: amplification allows virus carriers to invade more rapidly when rare, Curr Biol, vol.16, pp.2048-52, 2006.

J. R. Brum, B. L. Hurwitz, O. Schofield, H. W. Ducklow, and M. B. Sullivan, Seasonal time bombs: dominant temperate viruses affect Southern Ocean microbial dynamics, ISME J, vol.10, pp.437-486, 2016.

H. Brussow, Bacteriophage-host interaction: from splendid isolation into a messy reality, Curr Opin Microbiol, vol.16, pp.500-506, 2013.

H. Brussow, How stable is the human gut microbiota? And why this question matters, Environ Microbiol, vol.18, pp.2779-83, 2016.

H. Brussow, C. Canchaya, and W. D. Hardt, Phages and the evolution of bacterial pathogens: from genomic rearrangements to lysogenic conversion, Microbiol Mol Biol Rev, vol.68, pp.45-54, 2004.

A. R. Buckling and P. B. , Antagonistic coevolution between a bacterium and a bacteriophage, Proc Biol Sci, vol.269, pp.931-937, 2002.

F. Bushman, Lateral DNA transfer: mechanisms and consequences. Cold spring Harbor, 2002.

C. Canchaya, G. Fournous, and H. Brussow, The impact of prophages on bacterial chromosomes, Mol Microbiol, vol.53, pp.9-18, 2004.

M. Carrolo, M. J. Frias, F. R. Pinto, J. Melo-cristino, and M. Ramirez, Prophage spontaneous activation promotes DNA release enhancing biofilm formation in Streptococcus pneumoniae, PLoS One, vol.5, p.15678, 2010.

F. A. Carvalho, N. Barnich, A. Sivignon, C. Darcha, and C. H. Chan, Crohn's disease adherentinvasive Escherichia coli colonize and induce strong gut inflammation in transgenic mice expressing human CEACAM, J Exp Med, vol.206, pp.2179-89, 2009.

S. Casjens, Prophages and bacterial genomics: what have we learned so far?, Mol Microbiol, vol.49, pp.277-300, 2003.

S. H. Casjens and R. W. , Bacteriophages and the bacterial genome, pp.39-53, 2005.

J. L. Castro-mejia, M. K. Muhammed, W. Kot, H. Neve, and C. M. Franz, Optimizing protocols for extraction of bacteriophages prior to metagenomic analyses of phage communities in the human gut, vol.3, p.64, 2015.

E. P. Cato, W. Carolyn, W. E. Salmon, and . Moore, Fusobacterium prausnitzii (Hauduroy et al.) Moore and Holdeman: Emended Description and Designation of, Neotype Strain International Journal of Systematic Bacteriology, vol.24, pp.225-254, 1974.

W. Cenens, A. Makumi, S. K. Govers, R. Lavigne, and A. Aertsen, Viral Transmission Dynamics at Single-Cell Resolution Reveal Transiently Immune Subpopulations Caused by a Carrier State Association, PLoS Genet, vol.11, p.1005770, 2015.

W. Cenens, M. T. Mebrhatu, A. Makumi, P. J. Ceyssens, and R. Lavigne, Expression of a novel P22 ORFan gene reveals the phage carrier state in Salmonella typhimurium, PLoS Genet, vol.9, p.1003269, 2013.

J. Y. Chang, S. M. Shin, J. Chun, J. H. Lee, and J. K. Seo, Pyrosequencing-based molecular monitoring of the intestinal bacterial colonization in preterm infants, J Pediatr Gastroenterol Nutr, vol.53, pp.512-521, 2011.

C. Chehoud, A. Dryga, Y. Hwang, D. Nagy-szakal, and E. B. Hollister, Transfer of Viral Communities between Human Individuals during Fecal Microbiota Transplantation, MBio, vol.7, p.322, 2016.

T. Chen, C. Y. Kim, A. Kaur, L. Lamothe, and M. Shaikh, Dietary fibre-based SCFA mixtures promote both protection and repair of intestinal epithelial barrier function in a Caco-2 cell model, Food Funct, vol.8, pp.1166-73, 2017.

C. Cherbuy, E. Honvo-houeto, A. Bruneau, C. Bridonneau, and C. Mayeur, Microbiota matures colonic epithelium through a coordinated induction of cell cycle-related proteins in gnotobiotic rat, Am J Physiol Gastrointest Liver Physiol, vol.299, pp.348-57, 2010.

S. Chibani-chennoufi, J. Sidoti, A. Bruttin, E. Kutter, S. Sarker et al., In vitro and in vivo bacteriolytic activities of Escherichia coli phages: implications for phage therapy, Antimicrob Agents Chemother, vol.48, pp.2558-69, 2004.

J. Choi, S. M. Kotay, and R. Goel, Various physico-chemical stress factors cause prophage induction in Nitrosospira multiformis 25196--an ammonia oxidizing bacteria, Water Res, vol.44, pp.4550-4558, 2010.

A. Chopin, A. Bolotin, A. Sorokin, S. D. Ehrlich, and M. Chopin, Analysis of six prophages in Lactococcus lactis IL1403: different genetic structure of temperate and virulent phage populations, Nucleic Acids Res, vol.29, pp.644-51, 2001.

M. C. Chopin, A. Chopin, and E. Bidnenko, Phage abortive infection in lactococci: variations on a theme, Curr Opin Microbiol, vol.8, pp.473-482, 2005.

G. Christie and R. , Interactions between satellite bacteriophage P4 and its helpers, Annu Rev Genet, vol.24, pp.465-90, 1990.

H. Chu, A. Khosravi, I. P. Kusumawardhani, A. H. Kwon, and A. C. Vasconcelos, Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease, Science, vol.352, pp.1116-1136, 2016.

N. D. Chu, S. A. Clarke, S. Timberlake, M. F. Polz, A. D. Grossman et al., A Mobile Element in mutS Drives Hypermutation in a Marine Vibrio, MBio, vol.8, 2017.

I. Y. Chung, H. W. Bae, H. J. Jang, B. O. Kim, and Y. H. Cho, Superinfection exclusion reveals heteroimmunity between Pseudomonas aeruginosa temperate phages, J Microbiol, vol.52, pp.515-535, 2014.

I. Y. Chung, H. J. Jang, H. W. Bae, and Y. H. Cho, A phage protein that inhibits the bacterial ATPase required for type IV pilus assembly, Proc Natl Acad Sci U S A, vol.111, pp.11503-11511, 2014.

M. J. Claesson, I. B. Jeffery, S. Conde, S. E. Power, and E. M. O'connor, Gut microbiota composition correlates with diet and health in the elderly, Nature, vol.488, pp.178-84, 2012.

M. J. Claesson, O. O'sullivan, Q. Wang, J. Nikkila, and J. R. Marchesi, Comparative analysis of pyrosequencing and a phylogenetic microarray for exploring microbial community structures in the human distal intestine, PLoS One, vol.4, p.6669, 2009.

M. R. Clokie, A. D. Millard, A. V. Letarov, and S. Heaphy, Phages in nature. Bacteriophage, vol.1, pp.31-45, 2011.

C. Guemes, A. G. Youle, M. Cantu, V. A. Felts, B. Nulton et al., Viruses as Winners in the Game of Life, Annu Rev Virol, vol.3, pp.197-214, 2016.

H. L. Coetzee, D. Klerk, H. C. Coetzee, J. N. Smit, and J. A. , Bacteriophage-tail-like particles associated with intra-species killing of Proteus vulgaris, J Gen Virol, vol.2, pp.29-36, 1968.

H. Cooper, S. N. Shah, R. S. Sedergran, and D. J. , Clinicopathologic study of dextran sulfate sodium experimental murine colitis, Laboratory investigation, vol.69, pp.238-287, 1999.

C. Cordonnier, L. Bihan, G. Emond-rheault, J. G. Garrivier, A. Harel et al., Vitamin B12 Uptake by the Gut Commensal Bacteria Bacteroides thetaiotaomicron Limits the Production of Shiga Toxin by Enterohemorrhagic Escherichia coli, Toxins, 2016.

D. Court, L. Green, and H. Echols, Positive and negative regulation by the cII and cIII gene products of bacteriophage lambda, Virology, vol.63, pp.484-91, 1975.

J. M. Cowlishaw and M. , Coevolution of a virus-alga system, Appl Microbiol, vol.29, pp.234-243, 1975.

E. H. Crost, E. H. Ajandouz, C. Villard, P. A. Geraert, A. Puigserver et al., Ruminococcin C, a new anti-Clostridium perfringens bacteriocin produced in the gut by the commensal bacterium Ruminococcus gnavus E1, Biochimie, vol.93, pp.1487-94, 2011.

F. D'herelle, On an invisible microbe antagonistic toward dysenteric bacilli: brief note by Mr. F. D'Herelle, presented by Mr. Roux, Res Microbiol, vol.158, pp.553-557, 1917.

A. Darfeuille-michaud, J. Boudeau, P. Bulois, C. Neut, and A. L. Glasser, High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn's disease, Gastroenterology, vol.127, pp.412-433, 2004.

A. Darfeuille-michaud, C. Neut, N. Barnich, E. Lederman, D. Martino et al., Presence of adherent Escherichia coli strains in ileal mucosa of patients with Crohn's disease, Gastroenterology, vol.115, pp.1405-1418, 1998.

K. A. Datsenko and B. L. Wanner, One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products, Proc Natl Acad Sci U S A, vol.97, pp.6640-6645, 2000.

E. V. Davies, C. Winstanley, J. L. Fothergill, and C. E. James, The role of temperate bacteriophages in bacterial infection, FEMS Microbiol Lett, vol.363, p.15, 2016.

J. Davies and D. Davies, Origins and evolution of antibiotic resistance, Microbiol Mol Biol Rev, vol.74, pp.417-450, 2010.

R. K. Dawkins and J. R. , Arms races between and within species, Proc R Soc Lond B Biol Sci, vol.205, pp.489-511, 1979.

C. De-filippo, D. Cavalieri, D. Paola, M. Ramazzotti, M. Poullet et al., Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa, Proc Natl Acad Sci U S A, vol.107, pp.14691-14697, 2010.

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 Genet, vol.10, p.1004181, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01134590

M. De-paepe, M. Leclerc, C. R. Tinsley, and M. A. Petit, Bacteriophages: an underestimated role in human and animal health?, Front Cell Infect Microbiol, vol.4, p.39, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204359

M. De-paepe, L. Tournier, E. Moncaut, O. Son, P. Langella et al., Carriage of lambda Latent Virus Is Costly for Its Bacterial Host due to Frequent Reactivation in Monoxenic Mouse Intestine, PLoS Genet, vol.12, p.1005861, 2016.

D. Sordi, L. Khanna, V. Debarbieux, and L. , The Gut Microbiota Facilitates Drifts in the Genetic Diversity and Infectivity of Bacterial Viruses, Cell Host Microbe, vol.22, p.3, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01827316

H. Delattre, O. Souiai, K. Fagoonee, R. Guerois, and M. A. Petit, Phagonaute: A web-based interface for phage synteny browsing and protein function prediction, Virology, vol.496, pp.42-50, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01357336

J. J. Dennehy, What Can Phages Tell Us about Host-Pathogen Coevolution?, Int J Evol Biol, vol.2012, p.396165, 2012.

F. D. Depardieu, J. P. Bernheim, A. Sherlock, A. Molina, H. Duclos et al., A Eukaryotic-like Serine/Threonine Kinase Protects Staphylococci against Phages, Cell Host Microbe, vol.20, pp.471-81, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01402430

L. Dethlefsen, S. Huse, M. L. Sogin, and D. A. Relman, The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing, PLoS Biol, vol.6, p.280, 2008.

L. Dethlefsen and D. A. Relman, Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation, Proc Natl Acad Sci U S A, vol.108, issue.1, pp.4554-61, 2011.

M. Diard, E. Bakkeren, J. K. Cornuault, K. Moor, and A. Hausmann, Inflammation boosts bacteriophage transfer between Salmonella spp, Science, vol.355, pp.1211-1226, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01607431

I. B. Dodd, K. E. Shearwin, and J. B. Egan, Revisited gene regulation in bacteriophage lambda, Curr Opin Genet Dev, vol.15, pp.145-52, 2005.

I. B. Dodd, K. E. Shearwin, A. J. Perkins, T. Burr, A. Hochschild et al., Cooperativity in long-range gene regulation by the lambda CI repressor, Genes Dev, vol.18, pp.344-54, 2004.

G. P. Donaldson, M. S. Ladinsky, K. B. Yu, J. G. Sanders, and B. B. Yoo, Gut microbiota utilize immunoglobulin A for mucosal colonization, Science, vol.360, pp.795-800, 2018.

G. P. Donaldson, S. M. Lee, and S. K. Mazmanian, Gut biogeography of the bacterial microbiota, Nat Rev Microbiol, vol.14, pp.20-32, 2016.

S. Doron, S. Melamed, G. Ofir, A. Leavitt, and A. Lopatina, Systematic discovery of antiphage defense systems in the microbial pangenome, Science, vol.359, 2018.

S. Doulatov, A. Hodes, L. Dai, N. Mandhana, and M. Liu, Tropism switching in Bordetella bacteriophage defines a family of diversity-generating retroelements, Nature, vol.431, pp.476-81, 2004.

J. W. Drake, A constant rate of spontaneous mutation in DNA-based microbes, Proc Natl Acad Sci U S A, vol.88, pp.7160-7164, 1991.

A. Dri and P. L. , Control of the LexA regulon by pH: evidence for a reversible inactivation of the LexA repressor during the growth cycle of Escherichia coli, Mol Microbiol, vol.12, pp.621-630, 1994.

B. A. Duerkop, C. V. Clements, D. Rollins, J. L. Rodrigues, and L. V. Hooper, A composite bacteriophage alters colonization by an intestinal commensal bacterium, Proc Natl Acad Sci U S A, vol.109, pp.17621-17627, 2012.

S. H. Duncan, R. I. Aminov, K. P. Scott, P. Louis, T. B. Stanton et al., Proposal of Roseburia faecis sp. nov., Roseburia hominis sp. nov. and Roseburia inulinivorans sp. nov., based on isolates from human faeces, Int J Syst Evol Microbiol, vol.56, pp.2437-2478, 2006.

S. H. Duncan, G. L. Hold, A. Barcenilla, C. S. Stewart, and H. J. Flint, Roseburia intestinalis sp. nov., a novel saccharolytic, butyrate-producing bacterium from human faeces, Int J Syst Evol Microbiol, vol.52, pp.1615-1635, 2002.

S. H. Duncan, G. L. Hold, H. J. Harmsen, C. S. Stewart, and H. J. Flint, Growth requirements and fermentation products of Fusobacterium prausnitzii, and a proposal to reclassify it as Faecalibacterium prausnitzii gen. nov., comb. nov, Int J Syst Evol Microbiol, vol.52, pp.2141-2147, 2002.

S. H. Duncan, P. Louis, J. M. Thomson, and H. J. Flint, The role of pH in determining the species composition of the human colonic microbiota, Environ Microbiol, vol.11, pp.2112-2134, 2009.

D. Ehrlich, S. , and M. Hitc, , 2010.

, Gastroenterol Clin Biol, vol.34, pp.23-31

B. E. Dutilh, N. Cassman, K. Mcnair, S. E. Sanchez, and G. G. Silva, A highly abundant bacteriophage discovered in the unknown sequences of human faecal metagenomes, Nat Commun, vol.5, p.4498, 2014.

A. Dydecka, S. Bloch, A. Rizvi, S. Perez, and B. Nejman-falenczyk, Bad Phages in Good Bacteria: Role of the Mysterious orf63 of lambda and Shiga Toxin-Converting Phi24B Bacteriophages, Front Microbiol, vol.8, p.1618, 2017.

P. B. Eckburg, E. M. Bik, C. N. Bernstein, E. Purdom, and L. Dethlefsen, Diversity of the human intestinal microbial flora, Science, vol.308, pp.1635-1643, 2005.

R. A. Edwards, K. Mcnair, K. Faust, J. Raes, and B. E. Dutilh, Computational approaches to predict bacteriophage-host relationships, FEMS Microbiol Rev, vol.40, pp.258-72, 2016.

C. Eggers and D. S. , Molecular evidence for a new bacteriophage of Borrelia burgdorferi, J Bacteriol, vol.181, pp.7308-7321, 1999.

J. Eid, A. Fehr, J. Gray, K. Luong, and J. Lyle, Real-time DNA sequencing from single polymerase molecules, Science, vol.323, pp.133-141, 2009.

F. Enault, A. Briet, L. Bouteille, S. Roux, M. B. Sullivan et al., Phages rarely encode antibiotic resistance genes: a cautionary tale for virome analyses, ISME J, vol.11, pp.237-284, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01557484

P. Enck, K. Zimmermann, K. Rusch, A. Schwiertz, S. Klosterhalfen et al., The effects of ageing on the colonic bacterial microflora in adults, Z Gastroenterol, vol.47, pp.653-661, 2009.

T. Engelhardt, J. Kallmeyer, H. Cypionka, and B. Engelen, High virus-to-cell ratios indicate ongoing production of viruses in deep subsurface sediments, ISME J, vol.8, pp.1503-1512, 2014.

Z. Erez, I. Steinberger-levy, M. Shamir, S. Doron, and A. Stokar-avihail, Communication between viruses guides lysis-lysogeny decisions, Nature, vol.541, pp.488-93, 2017.

L. Falgenhauer, Y. Yao, M. Fritzenwanker, J. Schmiedel, C. Imirzalioglu et al., Complete Genome Sequence of Phage-Like Plasmid pECOH89, Encoding CTX-M-15, Genome Announc, vol.2, 2014.

G. Falony, M. Joossens, S. Vieira-silva, J. Wang, and Y. Darzi, Population-level analysis of gut microbiome variation, Science, vol.352, pp.560-564, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01518384

Y. Fang, R. G. Mercer, L. M. Mcmullen, and M. G. Ganzle, Induction of Shiga Toxin-Encoding Prophage by Abiotic Environmental Stress in Food, Appl Environ Microbiol, vol.83, 2017.

R. Feiner, T. Argov, L. Rabinovich, N. Sigal, I. Borovok et al., A new perspective on lysogeny: prophages as active regulatory switches of bacteria, Nat Rev Microbiol, vol.13, pp.641-50, 2015.

S. Finegold, V. L. Sutter, and G. E. Mathisen, Human Intestinal Microflora in Health and Disease, Normal Indigenous Intestinal Flora, p.3, 1983.

V. A. Fischetti, In vivo acquisition of prophage in Streptococcus pyogenes, Trends Microbiol, vol.15, pp.297-300, 2007.

H. J. Flint, Microbiology: Antibiotics and adiposity, Nature, vol.488, pp.601-603, 2012.

C. O. Flores, J. R. Meyer, S. Valverde, L. Farr, and J. S. Weitz, Statistical structure of host-phage interactions, Proc Natl Acad Sci U S A, vol.108, pp.288-97, 2011.

C. O. Flores, S. Valverde, and J. S. Weitz, Multi-scale structure and geographic drivers of crossinfection within marine bacteria and phages, ISME J, vol.7, pp.520-552, 2013.

A. Folkmanis, W. Maltzman, P. Mellon, A. Skalka, and H. Echols, The essential role of the cro gene in lytic development by bacteriophage lambda, Virology, vol.81, pp.352-62, 1977.

M. E. Ford, G. J. Sarkis, A. E. Belanger, R. W. Hendrix, and G. F. Hatfull, Genome structure of mycobacteriophage D29: implications for phage evolution, J Mol Biol, vol.279, pp.143-64, 1998.

J. L. Fothergill, E. Mowat, M. J. Walshaw, M. J. Ledson, C. E. James et al., Effect of antibiotic treatment on bacteriophage production by a cystic fibrosis epidemic strain of Pseudomonas aeruginosa, Antimicrob Agents Chemother, vol.55, pp.426-434, 2011.

D. Friedman and D. L. , Transcription antitermination: the lambda paradigm updated, Mol Microbiol, vol.18, pp.191-200, 1995.

J. A. Fuhrman, Marine viruses and their biogeochemical and ecological effects, Nature, vol.399, pp.541-549, 1999.

C. Galiez, M. Siebert, F. Enault, J. Vincent, and J. Soding, WIsH: who is the host? Predicting prokaryotic hosts from metagenomic phage contigs, Bioinformatics, vol.33, pp.3113-3127, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01982199

J. A. Gama, A. M. Reis, I. Domingues, H. Mendes-soares, A. M. Matos et al., Temperate bacterial viruses as double-edged swords in bacterial warfare, PLoS One, vol.8, p.59043, 2013.

S. Gandon, A. Buckling, E. Decaestecker, and T. Day, Host-parasite coevolution and patterns of adaptation across time and space, J Evol Biol, vol.21, pp.1861-1867, 2008.

E. Garcia-gutierrez, M. J. Mayer, P. D. Cotter, and A. Narbad, Gut microbiota as a source of novel antimicrobials, Gut Microbes, pp.1-57, 2018.

D. Gebhart, S. R. Williams, K. A. Bishop-lilly, G. R. Govoni, and K. M. Willner, Novel high-molecularweight, R-type bacteriocins of Clostridium difficile, J Bacteriol, vol.194, pp.6240-6247, 2012.

B. L. Geller, R. G. Ivey, J. E. Trempy, and B. Hettinger-smith, Cloning of a chromosomal gene required for phage infection of Lactococcus lactis subsp. lactis C2, J Bacteriol, vol.175, pp.5510-5519, 1993.

P. Gemski, L. S. Baron, and N. Yamamoto, Formation of hybrids between coliphage lambda and Salmonella phage P22 with a Salmonella typhimurium hybrid sensitive to these phages, Proc Natl Acad Sci U S A, vol.69, pp.3110-3114, 1972.

P. Gibson and O. Rosella, Interleukin 8 secretion by colonic crypt cells in vitro: response to injury suppressed by butyrate and enhanced in inflammatory bowel disease, Gut, vol.37, pp.536-579, 1995.

E. B. Gilcrease and S. R. Casjens, The genome sequence of Escherichia coli tailed phage D6 and the diversity of Enterobacteriales circular plasmid prophages, Virology, vol.515, pp.203-217, 2018.

J. Godeke, K. Paul, J. Lassak, and K. M. Thormann, Phage-induced lysis enhances biofilm formation in Shewanella oneidensis MR-1, ISME J, vol.5, pp.613-639, 2011.

C. Goerke, J. Koller, and C. Wolz, Ciprofloxacin and trimethoprim cause phage induction and virulence modulation in Staphylococcus aureus, Antimicrob Agents Chemother, vol.50, pp.171-178, 2006.

O. Goerlich, P. Quillardet, and M. Hofnung, Induction of the SOS response by hydrogen peroxide in various Escherichia coli mutants with altered protection against oxidative DNA damage, J Bacteriol, vol.171, pp.6141-6148, 1989.

G. W. Goldberg, W. Jiang, D. Bikard, and L. A. Marraffini, Conditional tolerance of temperate phages via transcription-dependent CRISPR-Cas targeting, Nature, vol.514, pp.633-640, 2014.

G. W. Goldberg, E. A. Mcmillan, A. Varble, J. W. Modell, and P. Samai, Incomplete prophage tolerance by type III-A CRISPR-Cas systems reduces the fitness of lysogenic hosts, Nat Commun, vol.9, p.61, 2018.

T. Goldfarb, H. Sberro, E. Weinstock, O. Cohen, and S. Doron, BREX is a novel phage resistance system widespread in microbial genomes, EMBO J, vol.34, pp.169-83, 2015.

A. Golomidova, E. Kulikov, A. Isaeva, A. Manykin, and A. Letarov, The diversity of coliphages and coliforms in horse feces reveals a complex pattern of ecological interactions, Appl Environ Microbiol, vol.73, pp.5975-81, 2007.

J. K. Goodrich, J. L. Waters, A. C. Poole, J. L. Sutter, and O. Koren, Human genetics shape the gut microbiome, Cell, vol.159, pp.789-99, 2014.

U. Gophna, K. Sommerfeld, S. Gophna, W. F. Doolittle, and S. J. Veldhuyzen-van-zanten, Differences between tissue-associated intestinal microfloras of patients with Crohn's disease and ulcerative colitis, J Clin Microbiol, vol.44, pp.4136-4177, 2006.

H. A. Gordon and E. Bruckner-kardoss, Effect of normal microbial flora on intestinal surface area, Am J Physiol, vol.201, pp.175-183, 1961.

H. Guo, L. V. Tse, R. Barbalat, S. Sivaamnuaiphorn, and M. Xu, Diversity-generating retroelement homing regenerates target sequences for repeated rounds of codon rewriting and protein diversification, Mol Cell, vol.31, pp.813-836, 2008.

B. E. Gustafsson, T. Midtvedt, and K. Strandberg, Effects of microbial contamination on the cecum enlargement of germfree rats, Scand J Gastroenterol, vol.5, pp.309-323, 1970.

J. Haaber, J. J. Leisner, M. T. Cohn, A. Catalan-moreno, and J. B. Nielsen, Bacterial viruses enable their host to acquire antibiotic resistance genes from neighbouring cells, Nat Commun, vol.7, p.13333, 2016.

A. R. Hall, P. D. Scanlan, and A. Buckling, Bacteria-phage coevolution and the emergence of generalist pathogens, Am Nat, vol.177, pp.44-53, 2011.

A. R. Hall, P. D. Scanlan, A. D. Morgan, and A. Buckling, Host-parasite coevolutionary arms races give way to fluctuating selection, Ecol Lett, vol.14, pp.635-677, 2011.

W. D. Hamilton, The genetical evolution of social behaviour. I, J Theor Biol, vol.7, pp.1-16, 1964.

W. D. Hamilton, The genetical evolution of social behaviour. II, J Theor Biol, vol.7, pp.17-52, 1964.

D. Hatziioanou, M. J. Mayer, S. H. Duncan, H. J. Flint, and A. Narbad, A representative of the dominant human colonic Firmicutes, Roseburia faecis M72/1, forms a novel bacteriocin-like substance, Anaerobe, vol.23, pp.5-8, 2013.

J. Heinrich, M. Velleman, and H. Schuster, The tripartite immunity system of phages P1 and P7, FEMS Microbiol Rev, vol.17, pp.121-127, 1995.

R. Hendrix, J. G. Hatfull, G. F. Casjens, and S. , Lambda II. Cold spring Harbor, 1983.

K. Hennes and M. , Significance of bacteriophages for controlling bacterioplankton growth in a mesotrophic lake, Appl Environ Microbiol, vol.61, pp.333-373, 1995.

C. Herman, D. Thevenet, D. 'ari, R. Bouloc, and P. , The HflB protease of Escherichia coli degrades its inhibitor lambda cIII, J Bacteriol, vol.179, pp.358-63, 1997.

A. Hershey and M. , Independent functions of viral protein and nucleic acid in growth of bacteriophage, J Gen Physiol, vol.36, pp.39-56, 1952.

Z. A. Hobbs and S. T. , Diversity of phage infection types and associated terminology: the problem with 'Lytic or lysogenic, FEMS Microbiol Lett, vol.363, 2016.

T. W. Hoffmann, H. P. Pham, C. Bridonneau, A. C. Lamas, and B. , Microorganisms linked to inflammatory bowel disease-associated dysbiosis differentially impact host physiology in gnotobiotic mice, ISME J, vol.10, pp.460-77, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01303788

P. Y. Hong, J. A. Croix, E. Greenberg, H. R. Gaskins, and R. I. Mackie, Pyrosequencing-based analysis of the mucosal microbiota in healthy individuals reveals ubiquitous bacterial groups and microheterogeneity, PLoS One, vol.6, p.25042, 2011.

L. V. Hooper, J. Xu, P. G. Falk, T. Midtvedt, and J. I. Gordon, A molecular sensor that allows a gut commensal to control its nutrient foundation in a competitive ecosystem, Proc Natl Acad Sci U S A, vol.96, pp.9833-9841, 1999.

M. J. Hopkins, G. T. Macfarlane, E. Furrie, A. Fite, and S. Macfarlane, Characterisation of intestinal bacteria in infant stools using real-time PCR and northern hybridisation analyses, FEMS Microbiol Ecol, vol.54, pp.77-85, 2005.

C. Howard-varona, K. R. Hargreaves, N. E. Solonenko, L. M. Markillie, and R. A. White, Multiple mechanisms drive phage infection efficiency in nearly identical hosts, Molecular biology of bacteriophage mu, vol.190, pp.624-656, 1975.

L. Hoyles, A. L. Mccartney, H. Neve, G. R. Gibson, and J. D. Sanderson, Characterization of viruslike particles associated with the human faecal and caecal microbiota, Res Microbiol, vol.165, pp.803-815, 2014.

G. Hutinet, A. Besle, O. Son, S. Mcgovern, and R. Guerois, Sak4 of Phage HK620 Is a RecA Remote Homolog With Single-Strand Annealing Activity Stimulated by Its Cognate SSB Protein, Front Microbiol, vol.9, p.743, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02179070

F. Jacob and J. Monod, Genetic regulatory mechanisms in the synthesis of proteins, J Mol Biol, vol.3, pp.318-56, 1961.

F. C. Jaomanjaka, O. Blanche-barbat, M. Petrel, M. Ballestra, P. Le-marrec et al., Characterization of a new virulent phage infecting the lactic acid bacterium Oenococcus oeni, Food Microbiology, vol.54, pp.167-77, 2016.

I. B. Jeffery, D. B. Lynch, and P. W. O'toole, Composition and temporal stability of the gut microbiota in older persons, ISME J, vol.10, pp.170-82, 2016.

S. Jiang and J. H. , Seasonal and diel abundance of viruses and occurrence of lysogeny/bacteriocinogeny in the marine environment, Marine Ecology Progress Series, vol.104, pp.163-72, 1994.

S. Jiang and J. H. , The abundance of lysogenic bacteria in marine microbial communities as determined by prophage induction, Marine Ecology Progress Series, vol.142, pp.27-38, 1996.

M. E. Johansson, J. K. Gustafsson, K. E. Sjoberg, J. Petersson, and L. Holm, Bacteria penetrate the inner mucus layer before inflammation in the dextran sulfate colitis model, PLoS One, vol.5, p.12238, 2010.

J. Joo, M. Gunny, M. Cases, P. Hudson, R. Albert et al., Bacteriophage-mediated competition in Bordetella bacteria, Proc Biol Sci, vol.273, pp.1843-1851, 2006.

M. Joossens, G. Huys, M. Cnockaert, D. Preter, V. Verbeke et al., Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives, Gut, vol.60, pp.631-638, 2011.

T. Jost, C. Lacroix, C. P. Braegger, F. Rochat, and C. Chassard, Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding, Environ Microbiol, vol.16, pp.2891-904, 2014.

R. Kahmann, F. Rudt, C. Koch, and G. Mertens, G inversion in bacteriophage Mu DNA is stimulated by a site within the invertase gene and a host factor, Cell, vol.41, pp.771-80, 1985.

A. D. Kaiser, Mutations in a temperate bacteriophage affecting its ability to lysogenize Escherichia coli, Virology, vol.3, pp.42-61, 1957.

N. Kamada, S. U. Seo, G. Y. Chen, and G. Nunez, Role of the gut microbiota in immunity and inflammatory disease, Nat Rev Immunol, vol.13, pp.321-356, 2013.

C. S. Kao and L. , The lit gene product which blocks bacteriophage T4 late gene expression is a membrane protein encoded by a cryptic DNA element, e14, J Bacteriol, vol.170, pp.2056-62, 1988.

K. M. Kauffman, F. A. Hussain, J. Yang, P. Arevalo, and J. M. Brown, A major lineage of non-tailed dsDNA viruses as unrecognized killers of marine bacteria, Nature, vol.554, pp.118-140, 2018.

L. A. Kelley, S. Mezulis, C. M. Yates, M. N. Wass, and M. J. Sternberg, The Phyre2 web portal for protein modeling, prediction and analysis, Nat Protoc, vol.10, pp.845-58, 2015.

K. J. Khan, T. A. Ullman, A. C. Ford, M. T. Abreu, and A. Abadir, Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis, Am J Gastroenterol, vol.106, pp.661-73, 2011.

M. S. Kim and J. W. Bae, Spatial disturbances in altered mucosal and luminal gut viromes of dietinduced obese mice, Environ Microbiol, vol.18, pp.1498-510, 2016.

M. Kim and J. W. , Lysogeny is prevalent and widely distributed in the murine gut microbiota, ISME J, vol.12, pp.1127-1168, 2018.

Y. Kim, S. H. Kim, K. Y. Whang, Y. J. Kim, and S. Oh, Inhibition of Escherichia coli O157:H7 attachment by interactions between lactic acid bacteria and intestinal epithelial cells, J Microbiol Biotechnol, vol.18, pp.1278-85, 2008.

B. Knowles, C. B. Silveira, B. A. Bailey, K. Barott, and V. A. Cantu, Lytic to temperate switching of viral communities, Nature, vol.531, pp.466-70, 2016.

O. Kobiler, S. Koby, D. Teff, D. Court, and A. B. Oppenheim, The phage lambda CII transcriptional activator carries a C-terminal domain signaling for rapid proteolysis, Proc Natl Acad Sci U S A, vol.99, pp.14964-14973, 2002.

O. Kobiler, A. Rokney, N. Friedman, D. L. Court, J. Stavans et al., Quantitative kinetic analysis of the bacteriophage lambda genetic network, Proc Natl Acad Sci U S A, vol.102, pp.4470-4475, 2005.

O. Kobiler, A. Rokney, and A. B. Oppenheim, Phage lambda CIII: a protease inhibitor regulating the lysis-lysogeny decision, PLoS One, vol.2, p.363, 2007.

J. E. Koenig, A. Spor, N. Scalfone, A. D. Fricker, and J. Stombaugh, Succession of microbial consortia in the developing infant gut microbiome, Proc Natl Acad Sci U S A, vol.108, issue.1, pp.4578-85, 2011.

B. B. Koskella and M. A. , Bacteria-phage coevolution as a driver of ecological and evolutionary processes in microbial communities, FEMS Microbiol Rev, vol.38, pp.916-947, 2014.

R. Kotlowski, C. N. Bernstein, S. Sepehri, and D. O. Krause, High prevalence of Escherichia coli belonging to the B2+D phylogenetic group in inflammatory bowel disease, Gut, vol.56, pp.669-75, 2007.

M. Krupovic and P. Forterre, Microviridae goes temperate: microvirus-related proviruses reside in the genomes of Bacteroidetes, PLoS One, vol.6, p.19893, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00593769

C. Kuo and H. , The extinction dynamics of bacterial pseudogenes, PLoS Genet, vol.6, 2010.

E. M. Kutter, S. J. Kuhl, and A. St, Re-establishing a place for phage therapy in western medicine, Future Microbiol, vol.10, pp.685-693, 2015.

J. M. Labonte, B. K. Swan, B. Poulos, H. Luo, and S. Koren, Single-cell genomics-based analysis of virus-host interactions in marine surface bacterioplankton, ISME J, vol.9, pp.2386-99, 2015.

S. J. Labrie, J. E. Samson, and S. Moineau, Bacteriophage resistance mechanisms, Nat Rev Microbiol, vol.8, pp.317-344, 2010.

J. E. Lambert, J. P. Myslicki, M. R. Bomhof, D. D. Belke, J. Shearer et al., Exercise training modifies gut microbiota in normal and diabetic mice, Appl Physiol Nutr Metab, vol.40, pp.749-52, 2015.

A. S. Lang and J. T. Beatty, Importance of widespread gene transfer agent genes in alphaproteobacteria, Trends Microbiol, vol.15, pp.54-62, 2007.

A. S. Lang, O. Zhaxybayeva, and J. T. Beatty, Gene transfer agents: phage-like elements of genetic exchange, Nat Rev Microbiol, vol.10, pp.472-82, 2012.

P. Lapaquette, A. L. Glasser, A. Huett, R. J. Xavier, and A. Darfeuille-michaud, Crohn's diseaseassociated adherent-invasive E. coli are selectively favoured by impaired autophagy to replicate intracellularly, Cell Microbiol, vol.12, pp.99-113, 2010.

N. Larsen, F. K. Vogensen, F. W. Van-den-berg, D. S. Nielsen, and A. S. Andreasen, Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults, PLoS One, vol.5, p.9085, 2010.

L. Latino, C. Midoux, Y. Hauck, G. Vergnaud, and C. Pourcel, Pseudolysogeny and sequential mutations build multiresistance to virulent bacteriophages in Pseudomonas aeruginosa, Microbiology, vol.162, pp.748-63, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01280758

M. F. Laursen, R. P. Laursen, A. Larnkjaer, C. Molgaard, and K. F. Michaelsen, Faecalibacterium Gut Colonization Is Accelerated by Presence of Older Siblings, 2017.

R. Lavigne, D. P. Summer, E. J. Seto, D. Mahadevan, and P. , Classification of Myoviridae bacteriophages using protein sequence similarity, BMC Microbiol, vol.9, p.224, 2009.

R. Lavigne, D. Seto, P. Mahadevan, H. W. Ackermann, and A. M. Kropinski, Unifying classical and molecular taxonomic classification: analysis of the Podoviridae using BLASTP-based tools, Res Microbiol, vol.159, pp.406-420, 2008.

T. D. Lawley and A. W. Walker, Intestinal colonization resistance, Immunology, vol.138, pp.1-11, 2013.

Y. K. Lee, K. Y. Puong, A. C. Ouwehand, and S. Salminen, Displacement of bacterial pathogens from mucus and Caco-2 cell surface by lactobacilli, J Med Microbiol, vol.52, pp.925-955, 2003.

I. Lehman and E. A. , On the structure of the glucosylated hydroxymethylcytosine nucleotides of coliphages T2, T4, and T6, J Biol Chem, vol.235, pp.3254-3263, 1960.

H. Lehnherr, E. Maguin, S. Jafri, and M. B. Yarmolinsky, Plasmid addiction genes of bacteriophage P1: doc, which causes cell death on curing of prophage, and phd, which prevents host death when prophage is retained, J Mol Biol, vol.233, pp.414-442, 1993.

J. P. Lemaitre, A. Duroux, R. Pimpie, J. M. Duez, and M. L. Milat, Listeria phage and phage tail induction triggered by components of bacterial growth media (phosphate, LiCl, nalidixic acid, and acriflavine), Appl Environ Microbiol, vol.81, pp.2117-2141, 2015.

R. L. Lenski and . Br, Constraints on the coevolution of bacteria and virulent phage -a model, some experiments, and predictions for natural communities, Am Nat, vol.125, pp.585-602, 1985.

P. Lepage, J. Colombet, P. Marteau, T. Sime-ngando, J. Dore et al., Dysbiosis in inflammatory bowel disease: a role for bacteriophages, vol.57, pp.424-429, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00526557

P. Lepage, P. Seksik, M. Sutren, M. F. De-la-cochetiere, and R. Jian, Biodiversity of the mucosaassociated microbiota is stable along the distal digestive tract in healthy individuals and patients with IBD, Inflamm Bowel Dis, vol.11, pp.473-80, 2005.

R. E. Ley, C. A. Lozupone, M. Hamady, R. Knight, and J. I. Gordon, Worlds within worlds: evolution of the vertebrate gut microbiota, Nat Rev Microbiol, vol.6, pp.776-88, 2008.

R. E. Ley, P. J. Turnbaugh, S. Klein, and J. I. Gordon, Microbial ecology: human gut microbes associated with obesity, Nature, vol.444, pp.1022-1025, 2006.

K. Licznerska, A. Dydecka, S. Bloch, G. Topka, and B. Nejman-falenczyk, The Role of the Exo-Xis Region in Oxidative Stress-Mediated Induction of Shiga Toxin-Converting Prophages, Oxid Med Cell Longev, vol.2016, p.8453135, 2016.

M. Lieb, The establishment of lysogenicity in Escherichia coli, J Bacteriol, vol.65, pp.642-51, 1953.

E. S. Lim, Y. Zhou, G. Zhao, I. K. Bauer, and L. Droit, Early life dynamics of the human gut virome and bacterial microbiome in infants, Nat Med, vol.21, pp.1228-1262, 2015.

G. Lindahl, G. Sironi, H. Bialy, and R. Calendar, Bacteriophage lambda; abortive infection of bacteria lysogenic for phage P2, Proc Natl Acad Sci U S A, vol.66, pp.587-94, 1970.

M. Liu, R. Deora, S. R. Doulatov, M. Gingery, and F. A. Eiserling, Reverse transcriptase-mediated tropism switching in Bordetella bacteriophage, Science, vol.295, pp.2091-2095, 2002.

Y. Y. Liu, Y. Wang, T. R. Walsh, L. X. Yi, and R. Zhang, Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: a microbiological and molecular biological study, Lancet Infect Dis, vol.16, pp.161-169, 2016.

M. B. Lobocka, D. J. Rose, G. Plunkett, M. Rusin, and A. Samojedny, Genome of bacteriophage P1, J Bacteriol, vol.186, pp.7032-68, 2004.

M. J. Loessner, R. B. Inman, P. Lauer, and R. Calendar, Complete nucleotide sequence, molecular analysis and genome structure of bacteriophage A118 of Listeria monocytogenes: implications for phage evolution, Mol Microbiol, vol.35, pp.324-364, 2000.

A. Lokta, Analytical note on certain rythmic relations in organic systems, Proc. Natl. Acad. Sci, vol.6, p.410, 1920.

A. Lopes, J. Amarir-bouhram, G. Faure, M. A. Petit, and R. Guerois, Detection of novel recombinases in bacteriophage genomes unveils Rad52, Rad51 and Gp2.5 remote homologs, Nucleic Acids Res, vol.38, pp.3952-62, 2010.

A. Lopes, P. Tavares, M. A. Petit, R. Guerois, and S. Zinn-justin, Automated classification of tailed bacteriophages according to their neck organization, BMC Genomics, vol.15, p.1027, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204418

M. Los, G. Wegrzyn, and P. Neubauer, A role for bacteriophage T4 rI gene function in the control of phage development during pseudolysogeny and in slowly growing host cells, Res Microbiol, vol.154, pp.547-52, 2003.

M. W. Los and G. , Pseudolysogeny. Advances in Virus Research, vol.82, pp.339-387, 2012.

M. J. Lu and U. Henning, Superinfection exclusion by T-even-type coliphages, Trends Microbiol, vol.2, pp.137-146, 1994.

S. Lucchini, F. Desiere, and H. Brussow, Comparative genomics of Streptococcus thermophilus phage species supports a modular evolution theory, J Virol, vol.73, pp.8647-56, 1999.

S. Lucchini, F. Desiere, and H. Brussow, Similarly organized lysogeny modules in temperate Siphoviridae from low GC content gram-positive bacteria, Virology, vol.263, pp.427-462, 1999.

G. A. Lugli, C. Milani, F. Turroni, D. Tremblay, and C. Ferrario, Prophages of the genus Bifidobacterium as modulating agents of the infant gut microbiota, Environ Microbiol, vol.18, pp.2196-213, 2016.

M. Lunde, A. H. Aastveit, J. M. Blatny, and I. F. Nes, Effects of diverse environmental conditions on {phi}LC3 prophage stability in Lactococcus lactis, Appl Environ Microbiol, vol.71, pp.721-728, 2005.

S. Luria and M. Delbrück, Mutations of Bacteria from Virus Sensitivity to Virus Resistance, Genetics, vol.28, pp.491-511, 1943.

A. Lwoff, Lysogeny. Bacteriol Rev, vol.17, pp.269-337, 1953.

M. Ly, M. B. Jones, S. R. Abeles, T. M. Santiago-rodriguez, and J. Gao, Transmission of viruses via our microbiomes, vol.4, p.64, 2016.

G. Macfarlane and G. R. , Carbohydrate fermentation, energy transduction and gas metabolism in the human large intestine, Gastrointestinal Microbiology, pp.269-318, 1997.

K. Machiels, M. Joossens, J. Sabino, D. Preter, V. Arijs et al., A decrease of the butyrateproducing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis, Gut, vol.63, pp.1275-83, 2014.

J. Mahony, S. Mcgrath, G. F. Fitzgerald, and D. Van-sinderen, Identification and characterization of lactococcal-prophage-carried superinfection exclusion genes, Appl Environ Microbiol, vol.74, pp.6206-6221, 2008.

M. A. Mahowald, F. E. Rey, H. Seedorf, P. J. Turnbaugh, and R. S. Fulton, Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla, Proc Natl Acad Sci U S A, vol.106, pp.5859-64, 2009.

E. Maiques, C. Ubeda, S. Campoy, N. Salvador, and I. Lasa, beta-lactam antibiotics induce the SOS response and horizontal transfer of virulence factors in Staphylococcus aureus, J Bacteriol, vol.188, pp.2726-2735, 2006.

K. S. Makarova, Y. I. Wolf, O. S. Alkhnbashi, F. Costa, and S. A. Shah, An updated evolutionary classification of CRISPR-Cas systems, Nat Rev Microbiol, vol.13, pp.722-758, 2015.

K. Malki, A. Kula, K. Bruder, E. Sible, and T. Hatzopoulos, Bacteriophages isolated from Lake Michigan demonstrate broad host-range across several bacterial phyla, Virol J, vol.12, p.164, 2015.

C. Manichanh, L. Rigottier-gois, E. Bonnaud, K. Gloux, and E. Pelletier, Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach, Gut, vol.55, pp.205-216, 2006.

P. Manrique, B. Bolduc, S. T. Walk, J. Van-der-oost, W. M. De-vos et al., Healthy human gut phageome, Proc Natl Acad Sci U S A, vol.113, pp.10400-10405, 2016.

P. Manrique, M. Dills, and M. J. Young, The Human Gut Phage Community and Its Implications for Health and Disease, Viruses, vol.9, 2017.

M. Marbouty, L. Baudry, A. Cournac, and R. Koszul, Scaffolding bacterial genomes and probing host-virus interactions in gut microbiome by proximity ligation (chromosome capture) assay, Sci Adv, vol.3, p.1602105, 2017.

M. Marbouty and R. Koszul, Generation and Analysis of Chromosomal Contact Maps of Bacteria, Methods Mol Biol, vol.1624, pp.75-84, 2017.

J. R. Marchesi, D. H. Adams, F. Fava, G. D. Hermes, and G. M. Hirschfield, The gut microbiota and host health: a new clinical frontier, Gut, vol.65, pp.330-339, 2016.

M. L. Capra, J. A. Reinheimer, and A. L. Quiberoni, Characterisation of three temperate phages released from the same Lactobacillus paracasei commercial strain, International Journal of Dairy Technology, vol.63, pp.396-405, 2010.

D. Mariat, O. Firmesse, F. Levenez, V. Guimaraes, and H. Sokol, The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age, BMC Microbiol, vol.9, p.123, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00657910

M. T. Marr, S. A. Datwyler, C. F. Meares, and J. W. Roberts, Restructuring of an RNA polymerase holoenzyme elongation complex by lambdoid phage Q proteins, Proc Natl Acad Sci U S A, vol.98, pp.8972-8980, 2001.

L. A. Marraffini, CRISPR-Cas immunity in prokaryotes, Nature, vol.526, pp.55-61, 2015.

M. F. Marston, F. J. Pierciey, J. Shepard, A. Gearin, G. Qi et al., Rapid diversification of coevolving marine Synechococcus and a virus, Proc Natl Acad Sci U S A, vol.109, pp.4544-4553, 2012.

P. Marteau, P. Pochart, J. Dore, C. Bera-maillet, A. Bernalier et al., Comparative study of bacterial groups within the human cecal and fecal microbiota, Appl Environ Microbiol, vol.67, pp.4939-4981, 2001.

H. M. Martin, B. J. Campbell, C. A. Hart, C. Mpofu, and M. Nayar, Enhanced Escherichia coli adherence and invasion in Crohn's disease and colon cancer, Gastroenterology, vol.127, pp.80-93, 2004.

R. Martin, F. Chain, M. S. Lu, J. Gratadoux, and J. J. , The commensal bacterium Faecalibacterium prausnitzii is protective in DNBS-induced chronic moderate and severe colitis models, Inflamm Bowel Dis, vol.20, pp.417-447, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00931932

M. Martinez-medina, X. Aldeguer, M. Lopez-siles, F. Gonzalez-huix, and C. Lopez-oliu, Molecular diversity of Escherichia coli in the human gut: new ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohn's disease, Inflamm Bowel Dis, vol.15, pp.872-82, 2009.

R. Martinez-rubio, N. Quiles-puchalt, M. Marti, S. Humphrey, and G. Ram, Phage-inducible islands in the Gram-positive cocci, ISME J, vol.11, pp.1029-1071, 2017.

J. T. Martinsohn, M. Radman, and M. A. Petit, The lambda red proteins promote efficient recombination between diverged sequences: implications for bacteriophage genome mosaicism, PLoS Genet, vol.4, p.1000065, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00496049

J. Martiny, L. Marston, M. F. Middelboe, and M. , Antagonistic coevolution of marine planktonic viruses and their hosts, Ann Rev Mar Sci, vol.6, pp.393-414, 2014.

R. C. Matos, N. Lapaque, L. Rigottier-gois, L. Debarbieux, and T. Meylheuc, Enterococcus faecalis prophage dynamics and contributions to pathogenic traits, PLoS Genet, vol.9, p.1003539, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01190746

D. Maura and L. Debarbieux, On the interactions between virulent bacteriophages and bacteria in the gut, Bacteriophage, vol.2, pp.229-262, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-01539087

D. Maura, M. Galtier, L. Bouguenec, C. Debarbieux, and L. , Virulent bacteriophages can target O104:H4 enteroaggregative Escherichia coli in the mouse intestine, Antimicrob Agents Chemother, vol.56, pp.6235-6277, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-01538999

T. Mavrich and G. F. , Bacteriophage evolution differs by host, lifestyle and genome, Nat Microbiol, vol.2, p.17112, 2017.

E. A. Mayer, K. Tillisch, and A. Gupta, Gut/brain axis and the microbiota, J Clin Invest, vol.125, pp.926-964, 2015.

C. L. Maynard, C. O. Elson, R. D. Hatton, and C. T. Weaver, Reciprocal interactions of the intestinal microbiota and immune system, Nature, vol.489, pp.231-272, 2012.

R. Mcmacken, N. Mantei, B. Butler, A. Joyner, and H. Echols, Effect of mutations in the c2 and c3 genes of bacteriophage lambda on macromolecular synthesis in infected cells, J Mol Biol, vol.49, pp.639-55, 1970.

M. Meessen-pinard, O. Sekulovic, and L. C. Fortier, Evidence of in vivo prophage induction during Clostridium difficile infection, Appl Environ Microbiol, vol.78, pp.7662-70, 2012.

R. Menouni, S. Champ, L. Espinosa, M. Boudvillain, and M. Ansaldi, Transcription termination controls prophage maintenance in Escherichia coli genomes, Proc Natl Acad Sci U S A, vol.110, pp.14414-14423, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00874882

D. J. Mercanti, D. Carminati, J. A. Reinheimer, and A. Quiberoni, Widely distributed lysogeny in probiotic lactobacilli represents a potentially high risk for the fermentative dairy industry, Int J Food Microbiol, vol.144, pp.503-513, 2011.

J. R. Meyer, D. T. Dobias, J. S. Weitz, J. E. Barrick, R. T. Quick et al., Repeatability and contingency in the evolution of a key innovation in phage lambda, Science, vol.335, pp.428-460, 2012.

Y. Michel-briand and C. Baysse, The pyocins of Pseudomonas aeruginosa, Biochimie, vol.84, pp.499-510, 2002.

M. Middelboe, A. Hagstrom, N. Blackburn, B. Sinn, and U. Fischer, Effects of Bacteriophages on the Population Dynamics of Four Strains of Pelagic Marine Bacteria, Microb Ecol, vol.42, pp.395-406, 2001.

M. H. Middelboe, K. Riemann, L. Nybroe, O. Haaber, and J. , Bacteriophages drive strain diversification in a marine Flavobacterium: implications for phage resistance and physiological properties, Environ Microbiol, vol.11, pp.1971-82, 2009.

M. Mikkonen, L. Dupont, T. Alatossava, and P. Ritzenthaler, Defective site-specific integration elements are present in the genome of virulent bacteriophage LL-H of Lactobacillus delbrueckii, Appl Environ Microbiol, vol.62, pp.1847-51, 1996.

S. Mills, F. Shanahan, C. Stanton, C. Hill, A. Coffey et al., Movers and shakers: influence of bacteriophages in shaping the mammalian gut microbiota, Gut Microbes, vol.4, pp.4-16, 2013.

I. Milne, G. Stephen, M. Bayer, P. J. Cock, and L. Pritchard, Using Tablet for visual exploration of second-generation sequencing data, Brief Bioinform, vol.14, pp.193-202, 2013.

S. Minot, A. Bryson, C. Chehoud, G. D. Wu, J. D. Lewis et al., Rapid evolution of the human gut virome, Proc Natl Acad Sci U S A, vol.110, pp.12450-12455, 2013.

S. Minot, S. Grunberg, G. D. Wu, J. D. Lewis, and F. D. Bushman, Hypervariable loci in the human gut virome, Proc Natl Acad Sci U S A, vol.109, pp.3962-3968, 2012.

S. Minot, R. Sinha, J. Chen, H. Li, and S. A. Keilbaugh, The human gut virome: inter-individual variation and dynamic response to diet, Genome Res, vol.21, pp.1616-1641, 2011.

S. Miquel, E. Peyretaillade, L. Claret, A. De-vallee, and C. Dossat, Complete genome sequence of Crohn's disease-associated adherent-invasive E. coli strain LF82, PLoS One, vol.5, 2010.

S. Mirold, W. Rabsch, M. Rohde, S. Stender, and H. Tschape, Isolation of a temperate bacteriophage encoding the type III effector protein SopE from an epidemic Salmonella typhimurium strain, Proc Natl Acad Sci U S A, vol.96, pp.9845-50, 1999.

M. Mirzaei and C. F. , Menage a trois in the human gut: interactions between host, bacteria and phages, Nat Rev Microbiol, vol.15, pp.397-408, 2017.

J. Mitchell, I. R. Siboo, D. Takamatsu, H. F. Chambers, and P. M. Sullam, Mechanism of cell surface expression of the Streptococcus mitis platelet binding proteins PblA and PblB, Mol Microbiol, vol.64, pp.844-57, 2007.

K. Mizoguchi, M. Morita, C. R. Fischer, M. Yoichi, Y. Tanji et al., Coevolution of bacteriophage PP01 and Escherichia coli O157:H7 in continuous culture, Appl Environ Microbiol, vol.69, pp.170-176, 2003.

S. R. Modi, H. H. Lee, C. S. Spina, and J. J. Collins, Antibiotic treatment expands the resistance reservoir and ecological network of the phage metagenome, Nature, vol.499, pp.219-241, 2013.

K. N. Moebus and H. , Bacteriophage sensitivity patterns among bacteria isolated from marine waters, HELGOLÄNDER MEERESUNTERSUCHUNGEN, vol.34, pp.375-85, 1981.

I. J. Molineux, Host-parasite interactions: recent developments in the genetics of abortive phage infections, New Biol, vol.3, pp.230-236, 1991.

S. Mondot, P. Lepage, P. Seksik, M. Allez, and X. Treton, Structural robustness of the gut mucosal microbiota is associated with Crohn's disease remission after surgery, Gut, vol.65, pp.954-62, 2016.

M. J. Morowitz, V. J. Denef, E. K. Costello, B. C. Thomas, and V. Poroyko, Strain-resolved community genomic analysis of gut microbial colonization in a premature infant, Proc Natl Acad Sci U S A, vol.108, pp.1128-1161, 2011.

K. C. Mountzouris, A. L. Mccartney, and G. R. Gibson, Intestinal microflora of human infants and current trends for its nutritional modulation, Br J Nutr, vol.87, pp.405-425, 2002.

M. G. Muller, J. Y. Ing, M. K. Cheng, B. A. Flitter, and G. R. Moe, Identification of a phage-encoded Igbinding protein from invasive Neisseria meningitidis, J Immunol, vol.191, pp.3287-96, 2013.

A. M. Nanda, A. Heyer, C. Kramer, A. Grunberger, D. Kohlheyer et al., Analysis of SOSinduced spontaneous prophage induction in Corynebacterium glutamicum at the singlecell level, J Bacteriol, vol.196, pp.180-188, 2014.

A. M. Nanda, K. Thormann, and J. Frunzke, Impact of spontaneous prophage induction on the fitness of bacterial populations and host-microbe interactions, J Bacteriol, vol.197, pp.410-419, 2015.

M. N. Neely and D. I. Friedman, Functional and genetic analysis of regulatory regions of coliphage H-19B: location of shiga-like toxin and lysis genes suggest a role for phage functions in toxin release, Mol Microbiol, vol.28, pp.1255-67, 1998.

J. Ni, G. D. Wu, L. Albenberg, and V. T. Tomov, Gut microbiota and IBD: causation or correlation?, Nat Rev Gastroenterol Hepatol, vol.14, pp.573-84, 2017.

P. Nicaise, A. Gleizes, F. Forestier, A. M. Quero, and C. Labarre, Influence of intestinal bacterial flora on cytokine (IL-1, IL-6 and TNF-alpha) production by mouse peritoneal macrophages, Eur Cytokine Netw, vol.4, pp.133-141, 1993.

B. E. Nickels, S. L. Dove, K. S. Murakami, S. A. Darst, and A. Hochschild, Protein-protein and protein-DNA interactions of sigma70 region 4 involved in transcription activation by lambdacI, J Mol Biol, vol.324, pp.17-34, 2002.

J. M. Norman, S. A. Handley, M. T. Baldridge, L. Droit, and C. Y. Liu, Disease-specific alterations in the enteric virome in inflammatory bowel disease, Cell, vol.160, pp.447-60, 2015.

M. Obuchowski, Y. Shotland, S. Koby, H. Giladi, and M. Gabig, Stability of CII is a key element in the cold stress response of bacteriophage lambda infection, J Bacteriol, vol.179, pp.5987-91, 1997.

G. Ofir, S. Melamed, H. Sberro, Z. Mukamel, and S. Silverman, DISARM is a widespread bacterial defence system with broad anti-phage activities, Nat Microbiol, vol.3, pp.90-98, 2018.

G. S. Ofir and R. , Contemporary Phage Biology: From Classic Models to New Insights, Cell, vol.172, pp.1260-70, 2018.

L. A. Ogilvie, L. D. Bowler, J. Caplin, C. Dedi, and D. Diston, Genome signature-based dissection of human gut metagenomes to extract subliminal viral sequences, Nat Commun, vol.4, p.2420, 2013.

L. A. Ogilvie and B. V. Jones, The human gut virome: a multifaceted majority, Front Microbiol, vol.6, p.918, 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.10618-10649, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01374960

A. B. Oppenheim, O. Kobiler, J. Stavans, D. L. Court, and S. Adhya, Switches in bacteriophage lambda development, Annu Rev Genet, vol.39, pp.409-438, 2005.

N. Otsuji, M. Sekiguchi, T. Iijima, and Y. Takagi, Induction of phage formation in the lysogenic Escherichia coli K-12 by mitomycin C, Nature, vol.184, pp.1079-80, 1959.

S. J. Ott, M. Musfeldt, D. F. Wenderoth, J. Hampe, and O. Brant, Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease, Gut, vol.53, pp.685-93, 2004.

S. V. Owen, N. Wenner, R. Canals, A. Makumi, and D. L. Hammarlof, Characterization of the Prophage Repertoire of African Salmonella Typhimurium ST313 Reveals High Levels of Spontaneous Induction of Novel Phage BTP1, Front Microbiol, vol.8, p.235, 2017.

D. Paez-espino, E. A. Eloe-fadrosh, G. A. Pavlopoulos, A. D. Thomas, and M. Huntemann, Uncovering Earth's virome, Nature, vol.536, pp.425-455, 2016.

C. Pal, M. D. Macia, A. Oliver, I. Schachar, and A. Buckling, Coevolution with viruses drives the evolution of bacterial mutation rates, Nature, vol.450, pp.1079-81, 2007.

C. Palmer, E. M. Bik, D. B. Digiulio, D. A. Relman, and P. O. Brown, Development of the human infant intestinal microbiota, PLoS Biol, vol.5, p.177, 2007.

R. J. Parsons, M. Breitbart, M. W. Lomas, and C. A. Carlson, Ocean time-series reveals recurring seasonal patterns of virioplankton dynamics in the northwestern Sargasso Sea, ISME J, vol.6, pp.273-84, 2012.

A. M. Patterson, I. E. Mulder, A. J. Travis, A. Lan, and N. Cerf-bensussan, Human Gut Symbiont Roseburia hominis Promotes and Regulates Innate Immunity, Front Immunol, vol.8, p.1166, 2017.

J. H. Paul, Prophages in marine bacteria: dangerous molecular time bombs or the key to survival in the seas?, ISME J, vol.2, pp.579-89, 2008.

S. I. Pavlova and L. Tao, Induction of vaginal Lactobacillus phages by the cigarette smoke chemical benzo[a]pyrene diol epoxide, Mutat Res, vol.466, pp.57-62, 2000.

A. Pawluk, J. Bondy-denomy, V. H. Cheung, K. L. Maxwell, and A. R. Davidson, A new group of phage anti-CRISPR genes inhibits the type I-E CRISPR-Cas system of Pseudomonas aeruginosa, MBio, vol.5, p.896, 2014.

A. Pawluk, A. R. Davidson, and K. L. Maxwell, Anti-CRISPR: discovery, mechanism and function, Nat Rev Microbiol, vol.16, pp.12-17, 2018.

A. Pawluk, R. H. Staals, C. Taylor, B. N. Watson, and S. Saha, Inactivation of CRISPR-Cas systems by anti-CRISPR proteins in diverse bacterial species, Nat Microbiol, vol.1, p.16085, 2016.

M. B. Paynter and . Hr, Dynamics of coliphage infections. Fermentation Advances, pp.323-359, 1969.

K. Pedersen, A. V. Zavialov, M. Y. Pavlov, J. Elf, K. Gerdes et al., The bacterial toxin RelE displays codon-specific cleavage of mRNAs in the ribosomal A site, Cell, vol.112, pp.131-171, 2003.

J. R. Penades, J. Chen, N. Quiles-puchalt, N. Carpena, and R. P. Novick, Bacteriophage-mediated spread of bacterial virulence genes, Curr Opin Microbiol, vol.23, pp.171-179, 2015.

J. R. Penades and G. E. Christie, The Phage-Inducible Chromosomal Islands: A Family of Highly Evolved Molecular Parasites, Annu Rev Virol, vol.2, pp.181-201, 2015.

J. Penders, C. Thijs, C. Vink, F. F. Stelma, and B. Snijders, Factors influencing the composition of the intestinal microbiota in early infancy, Pediatrics, vol.118, pp.511-532, 2006.

L. Peng, Z. R. Li, R. S. Green, I. R. Holzman, and J. Lin, Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers, J Nutr, vol.139, pp.1619-1644, 2009.

N. T. Perna, G. F. Mayhew, G. Posfai, S. Elliott, and M. S. Donnenberg, Molecular evolution of a pathogenicity island from enterohemorrhagic Escherichia coli O157:H7, Infect Immun, vol.66, pp.3810-3817, 1998.

N. T. Perna, G. Plunkett, V. Burland, B. Mau, and J. D. Glasner, Genome sequence of enterohaemorrhagic Escherichia coli O157:H7, Nature, vol.409, pp.529-562, 2001.

E. Pfeifer, M. Hunnefeld, O. Popa, T. Polen, and D. Kohlheyer, Silencing of cryptic prophages in Corynebacterium glutamicum, Nucleic Acids Res, vol.44, pp.10117-10148, 2016.

L. Philipson, P. A. Albertsson, and G. Frick, The purification and concentration of viruses by aqueous polymerphase systems, Virology, vol.11, pp.553-71, 1960.

M. Pleska, M. Lang, D. Refardt, B. R. Levin, and C. C. Guet, Phage-host population dynamics promotes prophage acquisition in bacteria with innate immunity, Nat Ecol Evol, vol.2, pp.359-66, 2018.

T. Poisot and . Lmhm, The structure of natural microbial enemy-victim networks, Ecol Process, vol.2, pp.1-9, 2013.

V. Poullain, S. Gandon, M. A. Brockhurst, A. Buckling, and M. E. Hochberg, The evolution of specificity in evolving and coevolving antagonistic interactions between a bacteria and its phage, Evolution, vol.62, pp.1-11, 2008.

U. Qimron, B. Marintcheva, S. Tabor, and C. C. Richardson, Genomewide screens for Escherichia coli genes affecting growth of T7 bacteriophage, Proc Natl Acad Sci U S A, vol.103, pp.19039-19083, 2006.

J. Qin, R. Li, J. Raes, M. Arumugam, and K. S. Burgdorf, A human gut microbial gene catalogue established by metagenomic sequencing, Nature, vol.464, pp.59-65, 2010.
URL : https://hal.archives-ouvertes.fr/cea-00908974

J. Qin, Y. Li, Z. Cai, S. Li, and J. Zhu, A metagenome-wide association study of gut microbiota in type 2 diabetes, Nature, vol.490, pp.55-60, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01204262

E. Quevrain, M. A. Maubert, C. Michon, F. Chain, and R. Marquant, Identification of an antiinflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn's disease, Gut, vol.65, pp.415-440, 2016.

E. Quevrain, M. A. Maubert, H. Sokol, B. Devreese, and P. Seksik, The presence of the antiinflammatory protein MAM, from Faecalibacterium prausnitzii, in the intestinal ecosystem, Gut, vol.65, p.882, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01301613

L. Rabinovich, N. Sigal, I. Borovok, R. Nir-paz, and A. A. Herskovits, Prophage excision activates Listeria competence genes that promote phagosomal escape and virulence, Cell, vol.150, pp.792-802, 2012.

D. V. Rakitina, A. I. Manolov, A. V. Kanygina, S. K. Garushyants, and J. P. Baikova, Genome analysis of E. coli isolated from Crohn's disease patients, BMC Genomics, vol.18, p.544, 2017.

J. Rakonjac, N. J. Bennett, J. Spagnuolo, D. Gagic, and M. Russel, Filamentous bacteriophage: biology, phage display and nanotechnology applications, Curr Issues Mol Biol, vol.13, pp.51-76, 2011.

B. J. Rauch, M. R. Silvis, J. F. Hultquist, C. S. Waters, and M. J. Mcgregor, Inhibition of CRISPR-Cas9 with Bacteriophage Proteins, Cell, vol.168, p.10, 2017.

N. V. Ravin, N15: the linear phage-plasmid, Plasmid, vol.65, pp.102-111, 2011.

A. T. Reese, E. H. Cho, B. Klitzman, S. P. Nichols, and N. A. Wisniewski, Antibiotic-induced changes in the microbiota disrupt redox dynamics in the gut, Elife, vol.7, 2018.

A. E. Reeves, M. J. Koenigsknecht, I. L. Bergin, and V. B. Young, Suppression of Clostridium difficile in the gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family Lachnospiraceae, Infect Immun, vol.80, pp.3786-94, 2012.

D. Refardt, Within-host competition determines reproductive success of temperate bacteriophages, ISME J, vol.5, pp.1451-60, 2011.

D. Refardt, Real-time quantitative PCR to discriminate and quantify lambdoid bacteriophages of Escherichia coli K-12, Bacteriophage, vol.2, pp.98-104, 2012.

S. C. Resta, Effects of probiotics and commensals on intestinal epithelial physiology: implications for nutrient handling, J Physiol, vol.587, pp.4169-74, 2009.

B. Revet, B. Von-wilcken-bergmann, H. Bessert, A. Barker, and B. Muller-hill, Four dimers of lambda repressor bound to two suitably spaced pairs of lambda operators form octamers and DNA loops over large distances, Curr Biol, vol.9, pp.151-155, 1999.

A. Reyes, L. V. Blanton, S. Cao, G. Zhao, and M. Manary, Gut DNA viromes of Malawian twins discordant for severe acute malnutrition, Proc Natl Acad Sci U S A, vol.112, pp.11941-11947, 2015.

A. Reyes, M. Haynes, N. Hanson, F. E. Angly, and A. C. Heath, Viruses in the faecal microbiota of monozygotic twins and their mothers, Nature, vol.466, pp.334-342, 2010.

A. Reyes, M. Wu, N. P. Mcnulty, F. L. Rohwer, and J. I. Gordon, Gnotobiotic mouse model of phagebacterial host dynamics in the human gut, Proc Natl Acad Sci U S A, vol.110, pp.20236-20277, 2013.

S. A. Rice, C. H. Tan, P. J. Mikkelsen, V. Kung, and J. Woo, The biofilm life cycle and virulence of Pseudomonas aeruginosa are dependent on a filamentous prophage, ISME J, vol.3, pp.271-82, 2009.

L. Rigottier-gois, Dysbiosis in inflammatory bowel diseases: the oxygen hypothesis, ISME J, vol.7, pp.1256-61, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01003298

S. M. Ripp and R. V. , The role of pseudolysogeny in bacteriophage-host interactions in a natural freshwater environment, Microbiology, vol.143, pp.2065-70, 1997.

S. M. Ripp and R. V. , Dynamics of the pseudolysogenic response in slowly growing cells of Pseudomonas aeruginosa, Microbiology, vol.144, pp.2225-2257, 1998.

J. W. Roberts, C. W. Roberts, N. L. Craig, and E. M. Phizicky, Activity of the Escherichia coli recA-gene product, Cold Spring Harb Symp Quant Biol, vol.43, pp.917-937, 1979.

R. Robles-sikisaka, M. Ly, T. Boehm, M. Naidu, J. Salzman et al., Association between living environment and human oral viral ecology, ISME J, vol.7, pp.1710-1734, 2013.

B. Rodriguez-brito, L. Li, L. Wegley, M. Furlan, and F. Angly, Viral and microbial community dynamics in four aquatic environments, ISME J, vol.4, pp.739-51, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02008659

F. Rohwer, Global phage diversity, Cell, vol.113, p.141, 2003.

N. Rolhion, F. A. Carvalho, and A. Darfeuille-michaud, OmpC and the sigma(E) regulatory pathway are involved in adhesion and invasion of the Crohn's disease-associated Escherichia coli strain LF82, Mol Microbiol, vol.63, pp.1684-700, 2007.

M. G. Rooks and W. S. Garrett, Gut microbiota, metabolites and host immunity, Nat Rev Immunol, vol.16, pp.341-52, 2016.

J. L. Rosner, Formation, induction, and curing of bacteriophage P1 lysogens, Virology, vol.48, pp.679-89, 1972.

A. Ross, S. Ward, and P. Hyman, More Is Better: Selecting for Broad Host Range Bacteriophages, Front Microbiol, vol.7, p.1352, 2016.

D. Rothschild, O. Weissbrod, E. Barkan, A. Kurilshikov, and T. Korem, Environment dominates over host genetics in shaping human gut microbiota, Nature, vol.555, pp.210-225, 2018.

S. Roux, F. Enault, B. L. Hurwitz, and M. B. Sullivan, VirSorter: mining viral signal from microbial genomic data, PeerJ, vol.3, p.985, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01557667

D. V. Rozanov, D. 'ari, R. Sineoky, and S. P. , RecA-independent pathways of lambdoid prophage induction in Escherichia coli, J Bacteriol, vol.180, pp.6306-6321, 1998.

H. Ruska, Die Sichtbarmachung der bakteriophagen Lyse im Uebermikroskop, Naturewissenschaften, vol.28, pp.45-46, 1940.

G. P. Salmond and P. C. Fineran, A century of the phage: past, present and future, Nat Rev Microbiol, vol.13, pp.777-86, 2015.

A. D. Samuel, T. P. Pitta, W. S. Ryu, P. N. Danese, E. C. Leung et al., Flagellar determinants of bacterial sensitivity to chi-phage, Proc Natl Acad Sci U S A, vol.96, pp.9863-9869, 1999.

E. L. Sandvik, C. H. Fazen, T. C. Henry, W. W. Mok, and M. P. Brynildsen, Non-Monotonic Survival of Staphylococcus aureus with Respect to Ciprofloxacin Concentration Arises from ProphageDependent Killing of Persisters, Pharmaceuticals (Basel), vol.8, pp.778-92, 2015.

R. Sanjuan, M. R. Nebot, N. Chirico, L. M. Mansky, and R. Belshaw, Viral mutation rates, J Virol, vol.84, pp.9733-9781, 2010.

T. M. Santiago-rodriguez, M. Ly, M. C. Daigneault, I. H. Brown, and J. A. Mcdonald, Chemostat culture systems support diverse bacteriophage communities from human feces, vol.3, p.58, 2015.

C. Sao-jose, C. Baptista, and M. A. Santos, Bacillus subtilis operon encoding a membrane receptor for bacteriophage SPP1, J Bacteriol, vol.186, pp.8337-8383, 2004.

R. B. Sartor, Microbial influences in inflammatory bowel diseases, Gastroenterology, vol.134, pp.577-94, 2008.

M. Sasaki, S. V. Sitaraman, B. A. Babbin, P. Gerner-smidt, and E. M. Ribot, Invasive Escherichia coli are a feature of Crohn's disease, Lab Invest, vol.87, pp.1042-54, 2007.

B. Sauer, Functional expression of the cre-lox site-specific recombination system in the yeast Saccharomyces cerevisiae, Mol Cell Biol, vol.7, pp.2087-96, 1987.

D. C. Savage, J. E. Siegel, J. E. Snellen, and D. D. Whitt, Transit time of epithelial cells in the small intestines of germfree mice and ex-germfree mice associated with indigenous microorganisms, Appl Environ Microbiol, vol.42, pp.996-1001, 1981.

P. D. Scanlan, A. R. Hall, G. Blackshields, V. P. Friman, M. R. Davis et al., Coevolution with bacteriophages drives genome-wide host evolution and constrains the acquisition of abiotic-beneficial mutations, Mol Biol Evol, vol.32, pp.1425-1460, 2015.

T. Z. Schillinger and N. , The low incidence of diversity-generating retroelements in sequenced genomes, Mob Genet Elements, vol.2, pp.287-91, 2012.

S. J. Schrag and J. E. Mittler, Host-Parasite Coexistence: The Role of Spatial Refuges in Stabilizing Bacteria-Phage Interactions, The american Naturalist, vol.148, pp.348-77, 1996.

R. Schuch and V. A. Fischetti, Detailed genomic analysis of the Wbeta and gamma phages infecting Bacillus anthracis: implications for evolution of environmental fitness and antibiotic resistance, J Bacteriol, vol.188, pp.3037-51, 2006.

S. Schuldiner, V. Agmon, J. Brandsma, A. Cohen, E. Friedman et al., Induction of SOS functions by alkaline intracellular pH in Escherichia coli, J Bacteriol, vol.168, pp.936-945, 1986.

J. Scott, S. V. Nguyen, C. J. King, C. Hendrickson, and W. M. Mcshan, Phage-Like Streptococcus pyogenes Chromosomal Islands (SpyCI) and Mutator Phenotypes: Control by Growth State and Rescue by a SpyCI-Encoded Promoter, Front Microbiol, vol.3, p.317, 2012.

J. Scott, P. Thompson-mayberry, S. Lahmamsi, C. J. King, and W. M. Mcshan, Phage-associated mutator phenotype in group A streptococcus, J Bacteriol, vol.190, pp.6290-301, 2008.

K. D. Seed, M. Yen, B. J. Shapiro, I. J. Hilaire, and R. C. Charles, Evolutionary consequences of intrapatient phage predation on microbial populations, vol.3, p.3497, 2014.

J. P. Segain, R. De-la-bletiere, D. Bourreille, A. Leray, V. Gervois et al., Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn's disease, Gut, vol.47, pp.397-403, 2000.

I. Sekirov, S. L. Russell, L. C. Antunes, and B. B. Finlay, Gut microbiota in health and disease, Physiol Rev, vol.90, pp.859-904, 2010.

P. Seksik, H. Sokol, P. Lepage, N. Vasquez, and C. Manichanh, Review article: the role of bacteria in onset and perpetuation of inflammatory bowel disease, Aliment Pharmacol Ther, vol.24, pp.11-19, 2006.

L. Selva, D. Viana, G. Regev-yochay, K. Trzcinski, and J. M. Corpa, Killing niche competitors by remote-control bacteriophage induction, Proc Natl Acad Sci U S A, vol.106, pp.1234-1242, 2009.

G. Sevrin, S. Massier, B. Chassaing, A. Agus, and J. Delmas, Adaptation of adherent-invasive E. coli to gut environment: Impact on flagellum expression and bacterial colonization ability, Gut Microbes, pp.1-17, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02196749

Q. Shao, J. T. Trinh, C. S. Mcintosh, B. Christenson, G. Balazsi et al., Lysis-lysogeny coexistence: prophage integration during lytic development, 2017.

R. Sharma, U. Schumacher, V. Ronaasen, and M. Coates, Rat intestinal mucosal responses to a microbial flora and different diets, Gut, vol.36, pp.209-223, 1995.

I. Sharon, M. J. Morowitz, B. C. Thomas, E. K. Costello, D. A. Relman et al., Time series community genomics analysis reveals rapid shifts in bacterial species, strains, and phage during infant gut colonization, Genome Res, vol.23, pp.111-131, 2013.

P. M. Sharp, Molecular evolution of bacteriophages: evidence of selection against the recognition sites of host restriction enzymes, Mol Biol Evol, vol.3, pp.75-83, 1986.

M. Shimizu-kadota, M. Kiwaki, S. Sawaki, Y. Shirasawa, H. Shibahara-sone et al., Insertion of bacteriophage phiFSW into the chromosome of Lactobacillus casei strain Shirota (S-1): characterization of the attachment sites and the integrase gene, Gene, vol.249, pp.127-161, 2000.

J. Shin and K. S. Ko, Effect of plasmids harbouring blaCTX-M on the virulence and fitness of Escherichia coli ST131 isolates, Int J Antimicrob Agents, vol.46, pp.214-222, 2015.

A. Shkoporov, E. V. Khokhlova, C. B. Fitzgerald, S. R. Stockdale, and L. A. Draper, ?CrAss001, a member of the most abundant bacteriophage family in the human gut, 2018.

P. Siringan, P. L. Connerton, N. J. Cummings, and I. F. Connerton, Alternative bacteriophage life cycles: the carrier state of Campylobacter jejuni, Open Biol, vol.4, p.130200, 2014.

E. Six and C. A. , Bacteriophage P4: a satellite virus depending on a helper such as prophage P2, Virology, vol.51, pp.327-371, 1973.

M. Slominska, P. Neubauer, and G. Wegrzyn, Regulation of bacteriophage lambda development by guanosine 5'-diphosphate-3'-diphosphate, Virology, vol.262, pp.431-472, 1999.

G. P. Smith, Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface, Science, vol.228, pp.1315-1322, 1985.

L. Snyder, Phage-exclusion enzymes: a bonanza of biochemical and cell biology reagents?, Mol Microbiol, vol.15, pp.415-435, 1995.

H. Sokol, C. Lay, P. Seksik, and G. W. Tannock, Analysis of bacterial bowel communities of IBD patients: what has it revealed?, Inflamm Bowel Dis, vol.14, pp.858-67, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00652970

H. Sokol, P. Seksik, J. P. Furet, O. Firmesse, and I. Nion-larmurier, Low counts of Faecalibacterium prausnitzii in colitis microbiota, Inflamm Bowel Dis, vol.15, pp.1183-1192, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00657435

H. Sokol, P. Seksik, L. Rigottier-gois, C. Lay, and P. Lepage, Specificities of the fecal microbiota in inflammatory bowel disease, Inflamm Bowel Dis, vol.12, pp.106-117, 2006.

R. Soret, J. Chevalier, D. Coppet, P. Poupeau, G. Derkinderen et al., Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats, Gastroenterology, vol.138, pp.1772-82, 2010.

F. E. St-pierre and D. , Determination of cell fate selection during phage lambda infection, Proc Natl Acad Sci U S A, vol.105, pp.20705-20715, 2008.

T. S. Stappenbeck, L. V. Hooper, and J. I. Gordon, Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells, Proc Natl Acad Sci U S A, vol.99, pp.15451-15456, 2002.

A. Stern, L. Keren, O. Wurtzel, G. Amitai, and R. Sorek, Self-targeting by CRISPR: gene regulation or autoimmunity?, Trends Genet, vol.26, pp.335-375, 2010.

A. Stern, E. Mick, I. Tirosh, O. Sagy, and R. Sorek, CRISPR targeting reveals a reservoir of common phages associated with the human gut microbiome, Genome Res, vol.22, pp.1985-94, 2012.

A. S. Stern and R. , The phage-host arms race: shaping the evolution of microbes, Bioessays, vol.33, pp.43-51, 2011.

F. M. Stewart and B. R. Levin, The population biology of bacterial viruses: why be temperate, Theor Popul Biol, vol.26, pp.93-117, 1984.

J. Stokholm, M. J. Blaser, J. Thorsen, M. A. Rasmussen, and J. Waage, Maturation of the gut microbiome and risk of asthma in childhood, Nat Commun, vol.9, p.141, 2018.

E. Strauch, H. Kaspar, C. Schaudinn, P. Dersch, and K. Madela, Characterization of enterocoliticin, a phage tail-like bacteriocin, and its effect on pathogenic Yersinia enterocolitica strains, Appl Environ Microbiol, vol.67, pp.5634-5676, 2001.

F. Studier and B. A. , Use of bacteriophage T7 RNA polymerase to direct selective high-level expression of cloned genes, J Mol Biol, vol.189, pp.113-143, 1986.

I. C. Sutcliffe, L. Tao, J. J. Ferretti, and R. R. Russell, MsmE, a lipoprotein involved in sugar transport in Streptococcus mutans, J Bacteriol, vol.175, pp.1853-1858, 1993.

C. Suttle and F. , Mechanisms and rates of decay of marine viruses in seawater, Appl Environ Microbiol, vol.58, pp.3721-3730, 1992.

Z. Tamanai-shacoori, I. Smida, L. Bousarghin, O. Loreal, and V. Meuric, Roseburia spp.: a marker of health, Future Microbiol, vol.12, pp.157-70, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01451661

J. Tap, S. Mondot, F. Levenez, E. Pelletier, and C. Caron, Towards the human intestinal microbiota phylogenetic core, Environ Microbiol, vol.11, pp.2574-84, 2009.

T. B. Stantonr and D. , Motility as a Factor in Bowel Colonization by Roseburia cecicola, an Obligately Anaerobic Bacterium from the Mouse Caecum, Microbiology, vol.130, pp.173-83, 1984.

R. Thomas, L. Berdjeb, T. Sime-ngando, and S. Jacquet, Viral abundance, production, decay rates and life strategies (lysogeny versus lysis) in Lake Bourget (France), Environ Microbiol, vol.13, pp.616-646, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00820093

N. E. Thompson and P. A. Pattee, Genetic transformation in Staphylococcus aureus: demonstration of a competence-conferring factor of bacteriophage origin in bacteriophage 80 alpha lysates, J Bacteriol, vol.148, pp.294-300, 1981.

M. Tock and D. T. , The biology of restriction and anti-restriction, Curr Opin Microbiol, vol.8, pp.466-72, 2005.

M. Touchon, A. Bernheim, and E. P. Rocha, Genetic and life-history traits associated with the distribution of prophages in bacteria, ISME J, vol.10, pp.2744-54, 2016.

M. H. Tschop, P. Hugenholtz, and C. L. Karp, Getting to the core of the gut microbiome, Nat Biotechnol, vol.27, pp.344-350, 2009.

G. F. Tu, G. E. Reid, J. G. Zhang, R. L. Moritz, and R. J. Simpson, C-terminal extension of truncated recombinant proteins in Escherichia coli with a 10Sa RNA decapeptide, J Biol Chem, vol.270, pp.9322-9328, 1995.

P. J. Turnbaugh, M. Hamady, T. Yatsunenko, B. L. Cantarel, and A. Duncan, A core gut microbiome in obese and lean twins, Nature, vol.457, pp.480-484, 2009.

F. W. Twort, An investigation on the nature of ultra-microscopic viruses, The Lancet, pp.1241-1284, 1915.

J. S. Tyler, M. J. Mills, and D. I. Friedman, The operator and early promoter region of the Shiga toxin type 2-encoding bacteriophage 933W and control of toxin expression, J Bacteriol, vol.186, pp.7670-7679, 2004.

P. Van-den-abbeele, C. Belzer, M. Goossens, M. Kleerebezem, D. Vos et al., Butyrateproducing Clostridium cluster XIVa species specifically colonize mucins in an in vitro gut model, ISME J, vol.7, pp.949-61, 2013.

P. Van-den-abbeele, T. Van-de-wiele, W. Verstraete, and S. Possemiers, The host selects mucosal and luminal associations of coevolved gut microorganisms: a novel concept, FEMS Microbiol Rev, vol.35, pp.681-704, 2011.

M. W. Van-der-woude, Phase variation: how to create and coordinate population diversity, Curr Opin Microbiol, vol.14, pp.205-216, 2011.

S. Van-houte, A. Buckling, and E. R. Westra, Evolutionary Ecology of Prokaryotic Immune Mechanisms. Microbiol Mol Biol Rev, vol.80, pp.745-63, 2016.

S. Van-houte, A. K. Ekroth, J. M. Broniewski, H. Chabas, and B. Ashby, The diversity-generating benefits of a prokaryotic adaptive immune system, Nature, vol.532, pp.385-393, 2016.

L. Van-valen, A new evolutionary law, Evol. Theory, pp.1-30, 1973.

D. Vandeputte, G. Kathagen, D. 'hoe, K. Vieira-silva, S. Valles-colomer et al., Quantitative microbiome profiling links gut community variation to microbial load, Nature, vol.551, pp.507-518, 2017.

T. Vanhoutte, G. Huys, E. Brandt, and J. Swings, Temporal stability analysis of the microbiota in human feces by denaturing gradient gel electrophoresis using universal and group-specific 16S rRNA gene primers, FEMS Microbiol Ecol, vol.48, pp.437-483, 2004.

E. Varela, C. Manichanh, M. Gallart, A. Torrejon, and N. Borruel, Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis, Aliment Pharmacol Ther, vol.38, pp.151-61, 2013.

R. B. Vercoe, J. T. Chang, R. L. Dy, C. Taylor, and T. Gristwood, Cytotoxic chromosomal targeting by CRISPR/Cas systems can reshape bacterial genomes and expel or remodel pathogenicity islands, PLoS Genet, vol.9, p.1003454, 2013.

S. Vermeire, G. Van-assche, and P. Rutgeerts, Classification of inflammatory bowel disease: the old and the new, Curr Opin Gastroenterol, vol.28, pp.321-327, 2012.

J. Vermeiren, P. Van-den-abbeele, D. Laukens, L. K. Vigsnaes, D. Vos et al., Decreased colonization of fecal Clostridium coccoides/Eubacterium rectale species from ulcerative colitis patients in an in vitro dynamic gut model with mucin environment, FEMS Microbiol Ecol, vol.79, pp.685-96, 2012.

V. Pacheco, S. , G. Gonzalez, O. , P. Contreras et al., The lom gene of bacteriophage lambda is involved in Escherichia coli K12 adhesion to human buccal epithelial cells, FEMS Microbiol Lett, vol.156, pp.129-161, 1997.

J. Villarroel, K. A. Kleinheinz, V. I. Jurtz, H. Zschach, and O. Lund, HostPhinder: A Phage Host Prediction Tool, Viruses, vol.8, 2016.

V. Volterra, Variations and fluctuations of the numbers of individuals in coexsting animal populations, Mem. R. Comitato talassogr. ital, p.131, 1927.

A. A. Vostrov, O. A. Vostrukhina, A. N. Svarchevsky, and V. N. Rybchin, Proteins responsible for lysogenic conversion caused by coliphages N15 and phi80 are highly homologous, J Bacteriol, vol.178, pp.1484-1490, 1996.

M. K. Waldor and J. J. Mekalanos, Lysogenic conversion by a filamentous phage encoding cholera toxin, Science, vol.272, pp.1910-1914, 1996.

A. W. Walker, J. D. Sanderson, C. Churcher, G. C. Parkes, and B. N. Hudspith, High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease, BMC Microbiol, vol.11, p.7, 2011.

S. L. Wang, Z. R. Wang, and C. Q. Yang, Meta-analysis of broad-spectrum antibiotic therapy in patients with active inflammatory bowel disease, Exp Ther Med, vol.4, pp.1051-56, 2012.

X. Wang, Y. Kim, Q. Ma, S. H. Hong, and K. Pokusaeva, Cryptic prophages help bacteria cope with adverse environments, Nat Commun, vol.1, p.147, 2010.

J. S. Webb, L. S. Thompson, S. James, T. Charlton, and T. Tolker-nielsen, Cell death in Pseudomonas aeruginosa biofilm development, J Bacteriol, vol.185, pp.4585-92, 2003.

Y. Wei, A. Kirby, and B. R. Levin, The population and evolutionary dynamics of Vibrio cholerae and its bacteriophage: conditions for maintaining phage-limited communities, Am Nat, vol.178, pp.715-740, 2011.

Y. Wei, P. Ocampo, and B. R. Levin, An experimental study of the population and evolutionary dynamics of Vibrio cholerae O1 and the bacteriophage JSF4, Proc Biol Sci, vol.277, pp.3247-54, 2010.

M. G. Weinbauer, Ecology of prokaryotic viruses, FEMS Microbiol Rev, vol.28, pp.127-81, 2004.

M. G. Weinbauer, K. Hornak, J. Jezbera, J. Nedoma, J. R. Dolan et al., Synergistic and antagonistic effects of viral lysis and protistan grazing on bacterial biomass, production and diversity, Environ Microbiol, vol.9, pp.777-88, 2007.

M. Weiss, E. Denou, A. Bruttin, R. Serra-moreno, M. L. Dillmann et al., In vivo replication of T4 and T7 bacteriophages in germ-free mice colonized with Escherichia coli, Virology, vol.393, pp.16-23, 2009.

J. L. Weissman, R. Holmes, R. Barrangou, S. Moineau, and W. F. Fagan, Immune loss as a driver of coexistence during host-phage coevolution, ISME J, vol.12, pp.585-97, 2018.

J. Weitz, T. Meyer, J. R. Flores, C. O. Valverde, S. Sullivan et al., Phagebacteria infection networks, Trends Microbiol, vol.21, pp.82-91, 2013.

B. Wiggins and M. , Minimum bacterial density for bacteriophage replication: implications for significance of bacteriophages in natural ecosystems, Appl Environ Microbiol, vol.49, pp.19-23, 1985.

K. Williamson, C. L. Drissi, J. M. Buckingham, C. P. Thompson, and R. R. Helton, Estimates of viral abundance in soils are strongly influenced by extraction and enumeration methods, Biol Fertil Soils, vol.49, pp.857-69, 2013.

B. P. Willing, J. Dicksved, J. Halfvarson, A. F. Andersson, and M. Lucio, A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes, Gastroenterology, vol.139, p.1, 2010.

G. Wilson and N. E. , Restriction and modification systems, Annu Rev Genet, vol.25, pp.585-627, 1991.

M. E. Woolhouse, J. P. Webster, E. Domingo, B. Charlesworth, and B. R. Levin, Biological and biomedical implications of the co-evolution of pathogens and their hosts, Nat Genet, vol.32, pp.569-77, 2002.

C. H. Wu, I. J. Liu, R. M. Lu, and H. C. Wu, Advancement and applications of peptide phage display technology in biomedical science, J Biomed Sci, vol.23, p.8, 2016.

L. Wu, M. Gingery, M. Abebe, D. Arambula, and E. Czornyj, Diversity-generating retroelements: natural variation, classification and evolution inferred from a large-scale genomic survey, Nucleic Acids Res, vol.46, pp.11-24, 2018.

Z. Xu, J. Xie, L. Yang, D. Chen, B. M. Peters et al., Complete Sequence of pCY-CTX, a Plasmid Carrying a Phage-Like Region and an ISEcp1-Mediated Tn2 Element from Enterobacter cloacae, Microb Drug Resist, vol.24, pp.307-320, 2018.

L. Yang, W. Li, G. Z. Jiang, W. H. Zhang, and H. Z. Ding, Characterization of a P1-like bacteriophage carrying CTX-M-27 in Salmonella spp. resistant to third generation cephalosporins isolated from pork in China, Sci Rep, vol.7, p.40710, 2017.

M. Yassour, T. Vatanen, H. Siljander, A. M. Hamalainen, and T. Harkonen, Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability, Sci Transl Med, vol.8, pp.343-81, 2016.

T. Yatsunenko, F. E. Rey, M. J. Manary, I. Trehan, and M. G. Dominguez-bello, Human gut microbiome viewed across age and geography, Nature, vol.486, pp.222-229, 2012.

Y. Ye, Identification of diversity-generating retroelements in human microbiomes, Int J Mol Sci, vol.15, pp.14234-14280, 2014.

S. H. Yoon, M. J. Han, H. Jeong, C. H. Lee, and X. X. Xia, Comparative multi-omics systems analysis of Escherichia coli strains B and K-12, Genome Biol, vol.13, p.37, 2012.

Z. Zeng, X. Liu, J. Yao, Y. Guo, and B. Li, Cold adaptation regulated by cryptic prophage excision in Shewanella oneidensis, ISME J, vol.10, pp.2787-800, 2016.

H. Zhang, D. E. Fouts, J. Depew, and R. H. Stevens, Genetic modifications to temperate Enterococcus faecalis phage Ef11 that abolish the establishment of lysogeny and sensitivity to repressor, and increase host range and productivity of lytic infection, Microbiology, vol.159, pp.1023-1058, 2013.

X. Zhang, A. D. Mcdaniel, L. E. Wolf, G. T. Keusch, M. K. Waldor et al., Quinolone antibiotics induce Shiga toxin-encoding bacteriophages, toxin production, and death in mice, J Infect Dis, vol.181, pp.664-70, 2000.

N. Zinder and J. , Genetic exchange in Salmonella, J Bacteriol, vol.64, pp.679-99, 1952.

R. Zink, M. J. Loessner, and S. Scherer, Characterization of cryptic prophages (monocins) in Listeria and sequence analysis of a holin/endolysin gene, Microbiology, vol.141, pp.2577-84, 1995.

E. G. Zoetendal, A. Wright, T. Vilpponen-salmela, K. Ben-amor, A. D. Akkermans et al., Mucosa-associated bacteria in the human gastrointestinal tract are uniformly distributed along the colon and differ from the community recovered from feces, Appl Environ Microbiol, vol.68, pp.3401-3408, 2002.

L. F. Gally, 5'-GTCTATTGTTCGAGCAATTC Jekyll JC 58: 5'-AAAGAAAGCCGACATTGTTG JC 59: 5'-CCGGTTATTGTAATGATTATTTCTC Prophage 1 JC 32: 5'-ACTGAAGTGCATAACTATGG JC 35: 5'-GACTTGATTGATCGTGATAC Prophage