A. Aertsen and C. Michiels, Mrr instigates the SOS response after high pressure stress in Escherichia coli, Molecular Microbiology, vol.99, issue.5, pp.1381-1391, 2005.
DOI : 10.1073/pnas.092269199

A. Aertsen, T. Mebrhatu, M. Michiels, and C. , Activation of the Salmonella Typhimurium Mrr protein, Biochemical and Biophysical Research Communications, vol.367, issue.2, pp.435-439, 2008.
DOI : 10.1016/j.bbrc.2007.12.151

A. Mamun, A. Gautam, S. Humayun, and M. , Hypermutagenesis in mutA cells is mediated by mistranslational corruption of polymerase, and is accompanied by replication fork collapse, Molecular Microbiology, vol.148, issue.6, pp.1752-1763, 2006.
DOI : 10.1128/JB.183.5.1796-1800.2001

K. Allen and M. Griffiths, Impact of hydroxyl- and superoxide anion-based oxidative stress on logarithmic and stationary phase Escherichia coli O157:H7 stress and virulence gene expression, Food Microbiology, vol.29, issue.1, pp.141-147, 2012.
DOI : 10.1016/j.fm.2011.09.014

J. Aranda, M. Poza, M. Shingu-vazquez, P. Cortes, J. Boyce et al., Identification of a DNA-Damage-Inducible Regulon in Acinetobacter baumannii, Journal of Bacteriology, vol.195, issue.24, 2013.
DOI : 10.1128/JB.00853-13

L. Aravind, S. Anand, and L. Iyer, Novel autoproteolytic and DNA-damage sensing components in the bacterial SOS response and oxidized methylcytosine-induced eukaryotic DNA demethylation systems, Biology Direct, vol.14, issue.10, p.20, 2013.
DOI : 10.1093/bioinformatics/14.10.892

M. Bagdasarian, A. Bailone, J. Angulo, P. Scholz, M. Bagdasarian et al., PsiB, an anti-SOS protein, is transiently expressed by the F sex factor during its transmission to an Escherichia coli K-12 recipient, Molecular Microbiology, vol.130, issue.7, pp.885-893, 1992.
DOI : 10.1016/0378-1119(84)90012-X

Z. Baharoglu and D. Mazel, ABSTRACT, Antimicrobial Agents and Chemotherapy, vol.55, issue.5, pp.2438-2441, 2011.
DOI : 10.1128/AAC.01549-10

Z. Baharoglu, D. Bikard, and D. Mazel, Conjugative DNA Transfer Induces the Bacterial SOS Response and Promotes Antibiotic Resistance Development through Integron Activation, PLoS Genetics, vol.310, issue.Pt 9, p.1001165, 2010.
DOI : 10.1371/journal.pgen.1001165.s006

Z. Baharoglu, E. Krin, and D. Mazel, Connecting Environment and Genome Plasticity in the Characterization of Transformation-Induced SOS Regulation and Carbon Catabolite Control of the Vibrio cholerae Integron Integrase, Journal of Bacteriology, vol.194, issue.7, 2012.
DOI : 10.1128/JB.05982-11

Z. Baharoglu, E. Krin, and D. Mazel, RpoS Plays a Central Role in the SOS Induction by Sub-Lethal Aminoglycoside Concentrations in Vibrio cholerae, PLoS Genetics, vol.336, issue.4, 2013.
DOI : 10.1371/journal.pgen.1003421.t002

URL : https://hal.archives-ouvertes.fr/pasteur-01691109

Z. Baharoglu, A. Babosan, and D. Mazel, Identification of genes involved in low aminoglycoside-induced SOS response in Vibrio cholerae: a role for transcription stalling and Mfd helicase, Nucleic Acids Research, vol.53, issue.4, 2013.
DOI : 10.1111/j.1365-2958.2004.04154.x

URL : https://hal.archives-ouvertes.fr/pasteur-01423602

Z. Baharoglu, R. Lestini, S. Duigou, and B. Michel, RNA polymerase mutations that facilitate replication progression in the rep uvrD recF mutant lacking two accessory replicative helicases, Molecular Microbiology, vol.95, issue.2, 2010.
DOI : 10.1111/j.1365-2958.2010.07208.x

URL : https://hal.archives-ouvertes.fr/hal-00552639

S. Balashov and M. Humayun, Mistranslation induced by streptomycin provokes a RecABC/RuvABC-dependent mutator phenotype in Escherichia coli cells 1 1Edited by M. Gottesman, Journal of Molecular Biology, vol.315, issue.4, pp.513-527, 2002.
DOI : 10.1006/jmbi.2001.5273

J. Beaber, B. Hochhut, and M. Waldor, SOS response promotes horizontal dissemination of antibiotic resistance genes, Nature, vol.53, issue.6969, pp.72-74, 2004.
DOI : 10.1073/pnas.120163297

O. Belov, O. Chuluunbaatar, M. &. Kapralov, and N. Sweilam, The role of the bacterial mismatch repair system in SOS-induced mutagenesis: A theoretical background, Journal of Theoretical Biology, vol.332, pp.30-41, 2013.
DOI : 10.1016/j.jtbi.2013.04.026

P. Bernard and M. Couturier, Cell killing by the F plasmid CcdB protein involves poisoning of DNA-topoisomerase II complexes, Journal of Molecular Biology, vol.226, issue.3, pp.735-745, 1992.
DOI : 10.1016/0022-2836(92)90629-X

B. Boles and P. Singh, Endogenous oxidative stress produces diversity and adaptability in biofilm communities, Proceedings of the National Academy of Sciences, vol.100, issue.14, pp.12503-12508, 2008.
DOI : 10.1073/pnas.1033133100

URL : http://www.pnas.org/content/105/34/12503.full.pdf

H. Boubakri, A. De-septenville, E. Viguera, and B. Michel, The helicases DinG, Rep and UvrD cooperate to promote replication across transcription units in vivo, The EMBO Journal, vol.178, issue.1, pp.145-157, 2010.
DOI : 10.1093/nar/20.24.6713

C. Boutry, B. Delplace, A. Clippe, L. Fontaine, and P. Hols, SOS Response Activation and Competence Development Are Antagonistic Mechanisms in Streptococcus thermophilus, Journal of Bacteriology, vol.195, issue.4, pp.696-707, 2013.
DOI : 10.1128/JB.01605-12

URL : http://jb.asm.org/content/195/4/696.full.pdf

M. Butala, D. Zgur-bertok, and S. Busby, The bacterial LexA transcriptional repressor, Cellular and Molecular Life Sciences, vol.66, issue.1, pp.82-93, 2009.
DOI : 10.1007/s00018-008-8378-6

M. Butala, D. Klose, V. Hodnik, A. Rems, Z. Podlesek et al., Interconversion between bound and free conformations of LexA orchestrates the bacterial SOS response, Nucleic Acids Research, vol.160, issue.15, pp.6546-6557, 2011.
DOI : 10.1016/j.resmic.2009.08.012

J. Barbe, D. Mazel, and I. Erill, Prevalence of SOS-mediated control of integron integrase expression as an adaptive trait of chromosomal and mobile integrons, Mob DNA, vol.2, p.6, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00598347

R. Centore, M. Leeson, and S. Sandler, UvrD303, a hyper-helicase mutant that antagonizes RecA-dependent SOS expression by a mechanism that depends on its C-terminus, 2008.

X. Charpentier, P. Polard, and J. Claverys, Induction of competence for genetic transformation by antibiotics: convergent evolution of stress responses in distant bacterial species lacking SOS?, Current Opinion in Microbiology, vol.15, issue.5, pp.570-576, 2012.
DOI : 10.1016/j.mib.2012.08.001

URL : https://hal.archives-ouvertes.fr/hal-00787606

R. Cirz and F. Romesberg, Induction and Inhibition of Ciprofloxacin Resistance-Conferring Mutations in Hypermutator Bacteria, Antimicrobial Agents and Chemotherapy, vol.50, issue.1, pp.220-225, 2006.
DOI : 10.1128/AAC.50.1.220-225.2006

J. Claverys, B. Martin, and P. Polard, The genetic transformation machinery: composition, localization, and mechanism, FEMS Microbiology Reviews, vol.140, issue.3, pp.643-656, 2009.
DOI : 10.1073/pnas.90.9.3875

URL : https://hal.archives-ouvertes.fr/hal-00407190

S. Cohen and G. Walker, The Transcription Elongation Factor NusA Is Required for Stress-Induced Mutagenesis in Escherichia coli, Current Biology, vol.20, issue.1, pp.80-85, 2010.
DOI : 10.1016/j.cub.2009.11.039

J. Courcelle, A. Khodursky, B. Peter, P. Brown, and P. Hanawalt, Comparative gene expression profiles following UV exposure in wild-type and SOS-deficient Escherichia coli, Genetics, vol.158, pp.41-64, 2001.

A. Czyz, B. Wrobel, and G. Wegrzyn, Vibrio harveyi bioluminescence plays a role in stimulation of DNA repair, Microbiology, vol.146, issue.2, pp.283-288, 2000.
DOI : 10.1099/00221287-146-2-283

A. Czyz, K. Plata, and G. Wegrzyn, Stimulation of DNA repair as an evolutionary drive for bacterial luminescence, Luminescence, vol.44, issue.3, pp.140-144, 2003.
DOI : 10.1126/science.190.4209.74

S. Da-re, F. Garnier, E. Guerin, S. Campoy, F. Denis et al., The SOS response promotes qnrB quinolone-resistance determinant expression, EMBO reports, vol.112, issue.8, pp.929-933, 2009.
DOI : 10.1128/AAC.00339-07

URL : https://hal.archives-ouvertes.fr/inserm-00533078

M. Daly, A new perspective on radiation resistance based on Deinococcus radiodurans, Nature Reviews Microbiology, vol.26, issue.3, pp.237-245, 2009.
DOI : 10.1099/00221287-147-7-1709

E. Davis, E. &. Dullaghan, and L. Rand, Definition of the Mycobacterial SOS Box and Use To Identify LexA-Regulated Genes in Mycobacterium tuberculosis, Journal of Bacteriology, vol.184, issue.12, pp.3287-3295, 2002.
DOI : 10.1128/JB.184.12.3287-3295.2002

D. Septenville, A. Duigou, S. Boubakri, H. Michel, and B. , Replication Fork Reversal after Replication???Transcription Collision, PLoS Genetics, vol.97, issue.4, p.1002622, 2012.
DOI : 10.1371/journal.pgen.1002622.s005

A. Deaconescu, A. Chambers, A. Smith, B. Nickels, A. Hochschild et al., Structural Basis for Bacterial Transcription-Coupled DNA Repair, Cell, vol.124, issue.3, pp.507-520, 2006.
DOI : 10.1016/j.cell.2005.11.045

URL : https://doi.org/10.1016/j.cell.2005.11.045

S. Delmas and I. Matic, Cellular response to horizontally transferred DNA in Escherichia coli is tuned by DNA repair systems, DNA Repair, vol.4, issue.2, pp.221-229, 2005.
DOI : 10.1016/j.dnarep.2004.09.008

E. Denamur, O. Tenaillon, C. Deschamps, D. Skurnik, E. Ronco et al., Intermediate Mutation Frequencies Favor Evolution of Multidrug Resistance in Escherichia coli, Genetics, vol.171, issue.2, pp.825-827, 2005.
DOI : 10.1534/genetics.105.045526

J. Didier, R. Villet, E. Huggler, D. Lew, D. Hooper et al., ABSTRACT, Antimicrobial Agents and Chemotherapy, vol.55, issue.5, pp.1946-1952, 2011.
DOI : 10.1128/AAC.01407-10

T. Dorr, K. Lewis, and M. Vulic, SOS Response Induces Persistence to Fluoroquinolones in Escherichia coli, PLoS Genetics, vol.178, issue.12, p.1000760, 2009.
DOI : 10.1371/journal.pgen.1000760.t001

T. Dorr, M. Vulic, and K. Lewis, Ciprofloxacin Causes Persister Formation by Inducing the TisB toxin in Escherichia coli, PLoS Biology, vol.183, issue.2, p.1000317, 2010.
DOI : 10.1371/journal.pbio.1000317.s001

D. Dubnau, DNA Uptake in Bacteria, Annual Review of Microbiology, vol.53, issue.1, pp.217-244, 1999.
DOI : 10.1146/annurev.micro.53.1.217

S. Dukan, A. Farewell, M. Ballesteros, F. Taddei, M. Radman et al., Protein oxidation in response to increased transcriptional or translational errors, Proceedings of the National Academy of Sciences, vol.440, issue.3, pp.5746-5749, 2000.
DOI : 10.1016/S0014-5793(98)01495-1

D. Dutta, K. Shatalin, V. Epshtein, M. Gottesman, and E. Nudler, Linking RNA Polymerase Backtracking to Genome Instability in E.??coli, Cell, vol.146, issue.4, pp.533-543, 2011.
DOI : 10.1016/j.cell.2011.07.034

D. Dwyer, M. Kohanski, and J. Collins, Role of reactive oxygen species in antibiotic action and resistance, Current Opinion in Microbiology, vol.12, issue.5, pp.482-489, 2009.
DOI : 10.1016/j.mib.2009.06.018

D. Dwyer, M. Kohanski, B. Hayete, and J. Collins, Gyrase inhibitors induce an oxidative damage cellular death pathway in Escherichia coli, Molecular Systems Biology, vol.273, issue.1, p.91, 2007.
DOI : 10.1016/0968-0004(96)20008-8

URL : http://msb.embopress.org/content/msb/3/1/91.full.pdf

I. Erill, S. Campoy, and J. Barbe, Aeons of distress: an evolutionary perspective on the bacterial SOS response, FEMS Microbiology Reviews, vol.31, issue.6, pp.637-656, 2007.
DOI : 10.1016/S0723-2020(99)80063-0

I. Erill, M. Escribano, S. Campoy, and J. Barbe, In silico analysis reveals substantial variability in the gene contents of the gamma proteobacteria LexA-regulon, Bioinformatics, vol.19, issue.17, pp.2225-2236, 2003.
DOI : 10.1093/bioinformatics/btg303

B. Ezraty, A. Vergnes, and M. Banzhaf, Fe-S Cluster Biosynthesis Controls Uptake of Aminoglycosides in a ROS-Less Death Pathway, Science, vol.74, issue.6, pp.1583-1587, 2013.
DOI : 10.1111/j.1365-2958.2009.06954.x

S. Farr and T. Kogoma, Oxidative Stress Responses in Escherichia-Coli and Salmonella- Typhimurium, Microbiological Reviews, vol.55, pp.561-585, 1991.

A. Fernandez-de-henestrosa, E. Rivera, A. Tapias, and J. Barbe, Identification of the Rhodobacter sphaeroides SOS box, Molecular Microbiology, vol.6, issue.5, pp.991-1003, 1998.
DOI : 10.1111/j.1365-2958.1992.tb00847.x

A. Fernandez-de-henestrosa, J. Cune, I. Erill, J. Magnuson, and J. Barbe, A Green Nonsulfur Bacterium, Dehalococcoides ethenogenes, with the LexA Binding Sequence Found in Gram-Positive Organisms, Journal of Bacteriology, vol.184, issue.21, pp.6073-6080, 2002.
DOI : 10.1128/JB.184.21.6073-6080.2002

A. Fernandez-de-henestrosa, T. Ogi, S. Aoyagi, D. Chafin, J. Hayes et al., Identification of additional genes belonging to the LexA regulon in Escherichia coli, Molecular Microbiology, vol.180, issue.6, pp.1560-1572, 2000.
DOI : 10.1073/pnas.83.15.5469

J. Fick, H. Soderstrom, R. Lindberg, C. Phan, M. Tysklind et al., CONTAMINATION OF SURFACE, GROUND, AND DRINKING WATER FROM PHARMACEUTICAL PRODUCTION, Environmental Toxicology and Chemistry, vol.28, issue.12, pp.2522-2527, 2009.
DOI : 10.1897/09-073.S1

M. Flores, N. Sanchez, and B. Michel, A fork-clearing role for UvrD, Molecular Microbiology, vol.73, issue.6, pp.1664-1675, 2005.
DOI : 10.1016/0300-9084(91)90196-8

L. Fonseca, J. Da-silva, J. Milanez, C. Monteiro-vitorello, L. Momo et al., Leptospira interrogans serovar Copenhageni Harbors Two lexA Genes Involved in SOS Response, PLoS ONE, vol.33, issue.10, p.76419, 2013.
DOI : 10.1371/journal.pone.0076419.s007

URL : http://doi.org/10.1371/journal.pone.0076419

J. Foti, B. Devadoss, J. Winkler, J. Collins, and G. Walker, Oxidation of the Guanine Nucleotide Pool Underlies Cell Death by Bactericidal Antibiotics, Science, vol.2, issue.7073, pp.315-319, 2012.
DOI : 10.1016/j.molcel.2005.01.012

E. Friedberg, R. Wagner, and M. Radman, Specialized DNA Polymerases, Cellular Survival, and the Genesis of Mutations, Science, vol.296, issue.5573, pp.1627-1630, 2002.
DOI : 10.1126/science.1070236

S. Fujii, A. Isogawa, and R. Fuchs, RecFOR proteins are essential for Pol V-mediated translesion synthesis and mutagenesis, The EMBO Journal, vol.11, issue.24, pp.5754-5763, 2006.
DOI : 10.1016/0921-8777(93)90024-B

URL : http://emboj.embopress.org/content/embojnl/25/24/5754.full.pdf

W. Gan, Z. Guan, J. Liu, T. Gui, K. Shen et al., R-loop-mediated genomic instability is caused by impairment of replication fork progression, Genes & Development, vol.25, issue.19, pp.2041-2056, 2011.
DOI : 10.1101/gad.17010011

J. Gibson, M. Lombardo, and P. Thornton, The ??E stress response is required for stress-induced mutation and amplification in Escherichia coli, Molecular Microbiology, vol.53, issue.2, pp.415-430, 2010.
DOI : 10.1101/SQB.1979.043.01.095

H. Gotoh, N. Kasaraneni, N. Devineni, S. Dallo, and T. Weitao, SOS involvement in stress-inducible biofilm formation, Biofouling, vol.48, issue.5, pp.603-611, 2010.
DOI : 10.1038/35101627

E. Guerin, G. Cambray, N. Sanchez-alberola, S. Campoy, I. Erill et al., The SOS Response Controls Integron Recombination, Science, vol.9, issue.5, p.1034, 2009.
DOI : 10.1016/j.mib.2006.07.002

URL : https://hal.archives-ouvertes.fr/hal-00409031

E. Gullberg, S. Cao, O. Berg, C. Ilback, L. Sandegren et al., Selection of Resistant Bacteria at Very Low Antibiotic Concentrations, PLoS Pathogens, vol.21, issue.7, p.1002158, 2011.
DOI : 10.1371/journal.ppat.1002158.s003

I. Gustafsson, M. Sjolund, E. Torell, M. Johannesson, L. Engstrand et al., Bacteria with increased mutation frequency and antibioticresistance are enriched in the commensal flora of patients with high antibiotic usage, Journal of Antimicrobial Chemotherapy, vol.52, issue.4, pp.645-650, 2003.
DOI : 10.1093/jac/dkg427

A. Gutierrez, L. Laureti, and S. Crussard, ??-lactam antibiotics promote bacterial mutagenesis via an RpoS-mediated reduction in replication fidelity, Nature Communications, vol.22, pp.1610-1640
DOI : 10.1111/j.1365-2958.1996.tb02664.x

B. Haggard and L. Bartsch, Net Changes in Antibiotic Concentrations Downstream from an Effluent Discharge, Journal of Environment Quality, vol.38, issue.1, pp.343-352, 2009.
DOI : 10.2134/jeq2007.0540

B. Hammer and B. Bassler, Quorum sensing controls biofilm formation in Vibrio cholerae, Molecular Microbiology, vol.99, issue.1, pp.101-104, 2003.
DOI : 10.1128/jb.173.13.4039-4048.1991

J. Han, O. Sahin, Y. Barton, and Q. Zhang, Key Role of Mfd in the Development of Fluoroquinolone Resistance in Campylobacter jejuni, PLoS Pathogens, vol.37, issue.6, p.1000083, 2008.
DOI : 10.1371/journal.ppat.1000083.t002

N. Handel, J. Schuurmans, S. Brul, and B. Kuile, ABSTRACT, Antimicrobial Agents and Chemotherapy, vol.57, issue.8, pp.3752-3762, 2013.
DOI : 10.1128/AAC.02096-12

A. Helmrich, M. Ballarino, and L. Tora, Collisions between Replication and Transcription Complexes Cause Common Fragile Site Instability at the Longest Human Genes, Molecular Cell, vol.44, issue.6, pp.966-977, 2011.
DOI : 10.1016/j.molcel.2011.10.013

A. Helmrich, M. Ballarino, E. Nudler, and L. Tora, Transcription-replication encounters, consequences and genomic instability, Nature Structural & Molecular Biology, vol.269, issue.4, pp.412-418, 2013.
DOI : 10.1126/science.1280856

E. Henle and S. Linn, Formation, Prevention, and Repair of DNA Damage by Iron/Hydrogen Peroxide, Journal of Biological Chemistry, vol.263, issue.6 suppl., pp.19095-19098, 1997.
DOI : 10.1074/jbc.271.44.27536

E. Henle, Z. Han, N. Tang, P. Rai, Y. Luo et al., -mediated Fenton Reactions Has Possible Biological Implications, Journal of Biological Chemistry, vol.266, issue.2, pp.962-971, 1999.
DOI : 10.1074/jbc.273.24.14683

URL : http://www.jbc.org/content/274/2/962.full.pdf

D. Hocquet and X. Bertrand, Metronidazole increases the emergence of ciprofloxacin- and amikacin-resistant Pseudomonas aeruginosa by inducing the SOS response, Journal of Antimicrobial Chemotherapy, vol.3, issue.6, 2013.
DOI : 10.1371/journal.pbio.0030176

URL : https://hal.archives-ouvertes.fr/hal-00926039

D. Hocquet, C. Llanes, M. Thouverez, H. Kulasekara, X. Bertrand et al., Evidence for Induction of Integron-Based Antibiotic Resistance by the SOS Response in a Clinical Setting, PLoS Pathogens, vol.47, issue.6, p.1002778, 2012.
DOI : 10.1371/journal.ppat.1002778.s008

URL : https://hal.archives-ouvertes.fr/hal-00802793

L. Hoffman, D. Argenio, D. Maccoss, M. Zhang, Z. Jones et al., Aminoglycoside antibiotics induce bacterial biofilm formation, Nature, vol.171, issue.7054, pp.1171-1175, 2005.
DOI : 10.1128/jb.171.12.6649-6655.1989

D. Hughes and D. Andersson, Selection of resistance at lethal and non-lethal antibiotic concentrations, Current Opinion in Microbiology, vol.15, issue.5, pp.555-560, 2012.
DOI : 10.1016/j.mib.2012.07.005

I. Husain, S. Chaney, and A. Sancar, Repair of cis-platinum-DNA adducts by ABC excinuclease in vivo and in vitro, J Bacteriol, vol.163, pp.817-823, 1985.

J. Imlay, Pathways of Oxidative Damage, Annual Review of Microbiology, vol.57, issue.1, pp.395-418, 2003.
DOI : 10.1146/annurev.micro.57.030502.090938

N. Jena, DNA damage by reactive species: Mechanisms, mutation and repair, Journal of Biosciences, vol.439, issue.3, pp.503-517, 2012.
DOI : 10.1038/nature04561

E. Jolivet, F. Lecointe, G. Coste, K. Satoh, I. Narumi et al., Limited concentration of RecA delays DNA double-strand break repair in Deinococcus radiodurans R1, Molecular Microbiology, vol.131, issue.1, pp.338-349, 2006.
DOI : 10.1186/1471-2180-5-17

URL : https://hal.archives-ouvertes.fr/hal-00079723

T. Jove, S. Da-re, F. Denis, D. Mazel, and M. Ploy, Inverse Correlation between Promoter Strength and Excision Activity in Class 1 Integrons, PLoS Genetics, vol.314, issue.1, p.1000793, 2010.
DOI : 10.1371/journal.pgen.1000793.s004

URL : https://hal.archives-ouvertes.fr/inserm-00533193

J. Kaplan, Antibiotic-induced biofilm formation, The International Journal of Artificial Organs, vol.34, issue.9, pp.737-751, 2011.
DOI : 10.5301/ijao.5000027

J. Kaplan, S. Jabbouri, and I. Sadovskaya, Extracellular DNA-dependent biofilm formation by Staphylococcus epidermidis RP62A in response to subminimal inhibitory concentrations of antibiotics, Research in Microbiology, vol.162, issue.5, pp.535-541, 2011.
DOI : 10.1016/j.resmic.2011.03.008

H. Karoui, F. Bex, P. Dreze, and M. Couturier, Ham22, a mini-F mutation which is lethal to host cell and promotes recA-dependent induction of lambdoid prophage, EMBO J, vol.2, pp.1863-1868, 1983.

M. Keaton, R. Rosato, K. Plata, C. Singh, and A. Rosato, Exposure of Clinical MRSA Heterogeneous Strains to ??-Lactams Redirects Metabolism to Optimize Energy Production through the TCA Cycle, PLoS ONE, vol.183, issue.8, p.71025, 2013.
DOI : 10.1371/journal.pone.0071025.s005

W. Kelley, Lex marks the spot: the virulent side of SOS and a closer look at the LexA regulon, Molecular Microbiology, vol.9, issue.5, pp.1228-1238, 2006.
DOI : 10.1086/315239

I. Keren, Y. Wu, J. Inocencio, L. Mulcahy, and K. Lewis, Killing by Bactericidal Antibiotics Does Not Depend on Reactive Oxygen Species, Science, vol.485, issue.7399, pp.1213-1216, 2013.
DOI : 10.1038/485439a

K. Keyamura, C. Sakaguchi, Y. Kubota, H. Niki, and T. Hishida, RecA recruits SMC-like RecN to DNA double-strand breaks, J Biol Chem, 2013.

K. Keyer and J. Imlay, Superoxide accelerates DNA damage by elevating free-iron levels, Proceedings of the National Academy of Sciences, vol.295, issue.2, pp.13635-13640, 1996.
DOI : 10.1016/0003-9861(92)90513-V

URL : http://www.pnas.org/content/93/24/13635.full.pdf

F. Khan, S. Singh, and B. Mishra, Summary, Journal of Integrative Bioinformatics, vol.5, issue.1, 2008.
DOI : 10.1515/jib-2008-86

S. Kim, K. Matsui, M. Yamada, P. Gruz, and T. Nohmi, Roles of chromosomal and episomal dinB genes encoding DNA pol IV in targeted and untargeted mutagenesis in Escherichia coli, Mol Genet Genomics, vol.266, pp.207-215, 2001.

H. Kimsey and M. Waldor, Vibrio cholerae LexA Coordinates CTX Prophage Gene Expression, Journal of Bacteriology, vol.191, issue.22, pp.6788-6795, 2009.
DOI : 10.1128/JB.00682-09

URL : http://jb.asm.org/content/191/22/6788.full.pdf

K. Ko, P. Tai, and C. Derby, ABSTRACT, Antimicrobial Agents and Chemotherapy, vol.56, issue.4, pp.1725-1734, 2012.
DOI : 10.1128/AAC.05874-11

M. Kohanski, D. Dwyer, and J. Collins, How antibiotics kill bacteria: from targets to networks, Nature Reviews Microbiology, vol.106, issue.6, pp.423-435, 2010.
DOI : 10.1038/nrmicro1949

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2896384/pdf

M. Kohanski, M. Depristo, and J. Collins, Sublethal Antibiotic Treatment Leads to Multidrug Resistance via Radical-Induced Mutagenesis, Molecular Cell, vol.37, issue.3, pp.311-320, 2010.
DOI : 10.1016/j.molcel.2010.01.003

URL : https://doi.org/10.1016/j.molcel.2010.01.003

M. Kohanski, D. Dwyer, B. Hayete, C. Lawrence, and J. Collins, A Common Mechanism of Cellular Death Induced by Bactericidal Antibiotics, Cell, vol.130, issue.5, pp.797-810, 2007.
DOI : 10.1016/j.cell.2007.06.049

M. Kohiyama, V. Contremoulins, and X. Baudin, Trashing of Single-Stranded DNA Generated during Processing of Arrested Replication Fork in E. coli, Journal of Molecular Biology, vol.425, issue.23, 2013.
DOI : 10.1016/j.jmb.2013.06.027

URL : https://hal.archives-ouvertes.fr/hal-00879896

L. Kovacic, N. Paulic, A. Leonardi, V. Hodnik, G. Anderluh et al., Structural insight into LexA-RecA* interaction, Nucleic acids research, 2013.

A. Krisko and M. Radman, Protein damage and death by radiation in Escherichia coli and Deinococcus radiodurans, Proceedings of the National Academy of Sciences, vol.193, issue.1, pp.14373-14377, 2010.
DOI : 10.1038/ismej.2007.116

URL : http://www.pnas.org/content/107/32/14373.full.pdf

K. Kummerer, Antibiotics in the aquatic environment ??? A review ??? Part II, Chemosphere, vol.75, issue.4, pp.435-441, 2009.
DOI : 10.1016/j.chemosphere.2008.12.006

K. Kummerer, Antibiotics in the aquatic environment ??? A review ??? Part I, Chemosphere, vol.75, issue.4, pp.417-434, 2009.
DOI : 10.1016/j.chemosphere.2008.11.086

K. Kumura, M. Sekiguchi, A. &. Steinum, and E. Seeberg, Stimulation of the UvrABC enzyme-catalyzed repair reactions by the UvrD protein (DNA helicase II), Nucleic Acids Research, vol.13, issue.5, pp.1483-1492, 1985.
DOI : 10.1093/nar/13.5.1483

J. Layton and P. Foster, Error-prone DNA polymerase IV is controlled by the stress-response sigma factor, RpoS, in Escherichia coli, Molecular Microbiology, vol.99, issue.2, pp.549-561, 2003.
DOI : 10.1073/pnas.092269199

J. Kim, The Helicobacter pylori Mfd protein is important for antibiotic resistance and DNA repair, Diagn Microbiol Infect Dis, vol.65, pp.454-456, 2009.

R. Leplae, D. Geeraerts, R. Hallez, J. Guglielmini, P. Dreze et al., Diversity of bacterial type II toxin???antitoxin systems: a comprehensive search and functional analysis of novel families, Nucleic Acids Research, vol.293, issue.13, pp.5513-5525, 2011.
DOI : 10.1371/journal.pbio.0030248

R. Lestini and B. Michel, UvrD controls the access of recombination proteins to blocked replication forks, The EMBO Journal, vol.134, issue.16, pp.3804-3814, 2007.
DOI : 10.1038/sj.emboj.7601804

C. Levesque, S. Brassard, J. Lapointe, and P. Roy, Diversity and relative strength of tandem promoters for the antibiotic-resistance genes of several integron, Gene, vol.142, issue.1, pp.49-54, 1994.
DOI : 10.1016/0378-1119(94)90353-0

C. Lewin and S. Amyes, The role of the SOS response in bacteria exposed to zidovudine or trimethoprim, Journal of Medical Microbiology, vol.34, issue.6, pp.329-332, 1991.
DOI : 10.1099/00222615-34-6-329

K. Lewis, Persister Cells, Annual Review of Microbiology, vol.64, issue.1, 2010.
DOI : 10.1146/annurev.micro.112408.134306

Y. Liu and J. Imlay, Cell Death from Antibiotics Without the Involvement of Reactive Oxygen Species, Science, vol.266, issue.3, pp.1210-1213, 2013.
DOI : 10.1111/j.1365-2958.2010.07418.x

Y. Liu, W. Huang, T. Huang, and C. Kunin, Extent of antibiotic use in Taiwan shown by antimicrobial activity in urine, The Lancet, vol.354, issue.9187, p.1360, 1999.
DOI : 10.1016/S0140-6736(99)07446-2

Y. Liu and W. Huang, Detection of Antimicrobial Activity in Urine for Epidemiologic Studies of Antibiotic Use, Journal of Clinical Epidemiology, vol.52, issue.6, pp.539-545, 1999.
DOI : 10.1016/S0895-4356(99)00027-X

E. Lopez and J. Blazquez, Effect of Subinhibitory Concentrations of Antibiotics on Intrachromosomal Homologous Recombination in Escherichia coli, Antimicrobial Agents and Chemotherapy, vol.53, issue.8, pp.3411-3415, 2009.
DOI : 10.1128/AAC.00358-09

A. Lu, X. Li, Y. Gu, P. Wright, and D. Chang, Repair of Oxidative DNA Damage: Mechanisms and Functions, Cell Biochemistry and Biophysics, vol.35, issue.2, pp.141-170, 2001.
DOI : 10.1385/CBB:35:2:141

T. Lu and J. Collins, Engineered bacteriophage targeting gene networks as adjuvants for antibiotic therapy, Proceedings of the National Academy of Sciences, vol.6, issue.1, pp.4629-4634, 2009.
DOI : 10.1371/journal.pbio.0060010

URL : http://www.pnas.org/content/106/12/4629.full.pdf

S. Lusetti, J. Drees, E. Stohl, H. Seifert, and M. Cox, The DinI and RecX Proteins Are Competing Modulators of RecA Function, Journal of Biological Chemistry, vol.261, issue.53, pp.55073-55079, 2004.
DOI : 10.1073/pnas.131007498

D. Macphee and M. Ambrose, Catabolite repression of SOS-dependent and SOS-independent spontaneous mutagenesis in stationary-phase Escherichia coli, Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, vol.686, issue.1-2, pp.84-89, 2010.
DOI : 10.1016/j.mrfmmm.2010.01.022

E. Maiques, C. Ubeda, S. Campoy, N. Salvador, I. Lasa et al., ??-Lactam Antibiotics Induce the SOS Response and Horizontal Transfer of Virulence Factors in Staphylococcus aureus, Journal of Bacteriology, vol.188, issue.7, pp.2726-2729, 2006.
DOI : 10.1128/JB.188.7.2726-2729.2006

R. Manasherob, C. Miller, K. Kim, and S. Cohen, Ribonuclease E Modulation of the Bacterial SOS Response, PLoS ONE, vol.276, issue.6, p.38426, 2012.
DOI : 10.1371/journal.pone.0038426.s003

E. Masse and M. Drolet, DNA Topoisomerase I Inhibits R-loop Formation by Relaxing Transcription-induced Negative Supercoiling, Journal of Biological Chemistry, vol.175, issue.23, pp.16659-16664, 1999.
DOI : 10.1021/bi00167a044

S. Massoni, M. Leeson, J. Long, K. Gemme, A. Mui et al., Factors Limiting SOS Expression in Log-Phase Cells of Escherichia coli, Journal of Bacteriology, vol.194, issue.19, pp.5325-5333, 2012.
DOI : 10.1128/JB.00674-12

D. Mazel, Integrons: agents of bacterial evolution, Nature Reviews Microbiology, vol.91, issue.8, pp.608-620, 2006.
DOI : 10.1073/pnas.91.20.9218

G. Mazon, S. Campoy, I. Erill, and J. Barbe, Identification of the Acidobacterium capsulatum LexA box reveals a lateral acquisition of the Alphaproteobacteria lexA gene, Microbiology, vol.152, issue.4, pp.1109-1118, 2006.
DOI : 10.1099/mic.0.28376-0

G. Mazon, J. Lucena, S. Campoy, A. Fernandez-de-henestrosa, P. Candau et al., LexA-binding sequences in Gram-positive and cyanobacteria are closely related, Molecular Genetics and Genomics, vol.271, issue.1, pp.40-49, 2004.
DOI : 10.1007/s00438-003-0952-x

G. Mazon, I. Erill, S. Campoy, P. Cortes, E. Forano et al., Reconstruction of the evolutionary history of the LexA-binding sequence, Microbiology, vol.150, issue.11, pp.3783-3795, 2004.
DOI : 10.1099/mic.0.27315-0

T. Mcbride, B. Preston, and L. Loeb, Mutagenic spectrum resulting from DNA damage by oxygen radicals, Biochemistry, vol.30, issue.1, pp.207-213, 1991.
DOI : 10.1021/bi00215a030

P. Mcglynn, N. Savery, and M. Dillingham, The conflict between DNA replication and transcription, Molecular Microbiology, vol.14, issue.1, pp.12-20, 2012.
DOI : 10.1016/0092-8674(88)90163-8

R. Mcveigh and R. Yasbin, Phenotypic differentiation of "smart" versus "naive" bacteriophages of Bacillus subtilis., Journal of Bacteriology, vol.178, issue.11, pp.3399-3401, 1996.
DOI : 10.1128/jb.178.11.3399-3401.1996

H. Merrikh, A. Ferrazzoli, and S. Lovett, Growth Phase and (p)ppGpp Control of IraD, a Regulator of RpoS Stability, in Escherichia coli, Journal of Bacteriology, vol.191, issue.24, pp.7436-7446, 2009.
DOI : 10.1128/JB.00412-09

H. Merrikh, Y. Zhang, A. Grossman, and J. Wang, Replication???transcription conflicts in bacteria, Nature Reviews Microbiology, vol.396, issue.7, pp.449-458, 2012.
DOI : 10.1016/j.bbrc.2010.04.119

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3467967/pdf

H. Merrikh, A. Ferrazzoli, A. Bougdour, A. Olivier-mason, and S. Lovett, A DNA damage response in Escherichia coli involving the alternative sigma factor, RpoS, Proceedings of the National Academy of Sciences, vol.60, issue.1, pp.611-616, 2009.
DOI : 10.1111/j.1365-2958.2006.05093.x

L. Mesak and J. Davies, Phenotypic changes in ciprofloxacin-resistant Staphylococcus aureus, Research in Microbiology, vol.160, issue.10, pp.785-791, 2009.
DOI : 10.1016/j.resmic.2009.09.013

L. Mesak, V. Miao, and J. Davies, Effects of Subinhibitory Concentrations of Antibiotics on SOS and DNA Repair Gene Expression in Staphylococcus aureus, Antimicrobial Agents and Chemotherapy, vol.52, issue.9, pp.3394-3397, 2008.
DOI : 10.1128/AAC.01599-07

B. Michel, After 30 Years of Study, the Bacterial SOS Response Still Surprises Us, PLoS Biology, vol.305, issue.7, p.255, 2005.
DOI : 10.1371/journal.pbio.0030255.g002

F. Mika, S. Busse, A. Possling, J. Berkholz, N. Tschowri et al., Targeting of csgD by the small regulatory RNA RprA links stationary phase, biofilm formation and cell envelope stress in Escherichia coli, Molecular Microbiology, vol.31, issue.1, 2012.
DOI : 10.1093/nar/gkg595

C. Miller, H. Ingmer, L. Thomsen, K. Skarstad, and S. Cohen, DpiA Binding to the Replication Origin of Escherichia coli Plasmids and Chromosomes Destabilizes Plasmid Inheritance and Induces the Bacterial SOS Response, Journal of Bacteriology, vol.185, issue.20, pp.6025-6031, 2003.
DOI : 10.1128/JB.185.20.6025-6031.2003

C. Miller, L. Thomsen, C. Gaggero, R. Mosseri, H. Ingmer et al., SOS Response Induction by ??-Lactams and Bacterial Defense Against Antibiotic Lethality, Science, vol.305, issue.5690, pp.1629-1631, 2004.
DOI : 10.1126/science.1101630

T. Mori, T. Nakamura, N. Okazaki, A. Furukohri, H. Maki et al., Escherichia coli DinB inhibits replication fork progression without significantly inducing the SOS response, Genes ^|^ Genetic Systems, vol.87, issue.2, pp.75-87, 2012.
DOI : 10.1266/ggs.87.75

R. Napolitano, R. Janel-bintz, J. Wagner, and R. Fuchs, All three SOS-inducible DNA polymerases (Pol II, Pol IV and Pol V) are involved in induced mutagenesis, The EMBO Journal, vol.19, issue.22, pp.6259-6265, 2000.
DOI : 10.1093/emboj/19.22.6259

URL : http://embojnl.embopress.org/content/embojnl/19/22/6259.full.pdf

I. Narumi, K. Satoh, M. Kikuchi, T. Funayama, T. Yanagisawa et al., The LexA Protein from Deinococcus radiodurans Is Not Involved in RecA Induction following gamma Irradiation, Journal of Bacteriology, vol.183, issue.23, pp.6951-6956, 2001.
DOI : 10.1128/JB.183.23.6951-6956.2001

F. Nassar, E. Rahal, A. Sabra, and G. Matar, Effects of Subinhibitory Concentrations of Antimicrobial Agents on Escherichia coli O157:H7 Shiga Toxin Release and Role of the SOS Response, Foodborne pathogens and disease, 2013.

M. Ni, L. Yang, X. Liu, and O. Qi, Fluence-Response Dynamics of the UV-Induced SOS Response in Escherichia coli, Current Microbiology, vol.15, issue.6, pp.521-526, 2008.
DOI : 10.1016/S0076-6879(06)08020-7

T. Nohmi, J. Battista, L. Dodson, and G. Walker, RecA-mediated cleavage activates UmuD for mutagenesis: mechanistic relationship between transcriptional derepression and posttranslational activation., Proceedings of the National Academy of Sciences, vol.85, issue.6, pp.1816-1820, 1988.
DOI : 10.1073/pnas.85.6.1816

URL : http://www.pnas.org/content/85/6/1816.full.pdf

M. Norton, A. Spilkia, and V. Godoy, Antibiotic Resistance Acquired through a DNA Damage-Inducible Response in Acinetobacter baumannii, Journal of Bacteriology, vol.195, issue.6, pp.1335-1345, 2013.
DOI : 10.1128/JB.02176-12

P. Paez, M. Becerra, and I. Albesa, Comparison of Macromolecular Oxidation by Reactive Oxygen Species in Three Bacterial Genera Exposed to Different Antibiotics, Cell Biochemistry and Biophysics, vol.4, issue.3, pp.467-472, 2011.
DOI : 10.1038/sj.embor.embor799

V. Pages and R. Fuchs, Uncoupling of Leading- and Lagging-Strand DNA Replication During Lesion Bypass in Vivo, Science, vol.300, issue.5623, pp.1300-1303, 2003.
DOI : 10.1126/science.1083964

V. Pages, N. Koffel-schwartz, and R. Fuchs, recX, a new SOS gene that is co-transcribed with the recA gene in Escherichia coli, DNA Repair, vol.2, issue.3, pp.273-284, 2003.
DOI : 10.1016/S1568-7864(02)00217-3

D. Pandey and K. Gerdes, Toxin-antitoxin loci are highly abundant in free-living but lost from host-associated prokaryotes, Nucleic Acids Research, vol.33, issue.3, pp.966-976, 2005.
DOI : 10.1093/nar/gki201

S. Partridge, G. Tsafnat, E. Coiera, and J. Iredell, Gene cassettes and cassette arrays in mobile resistance integrons, FEMS Microbiology Reviews, vol.16, issue.4, pp.757-784, 2009.
DOI : 10.1128/AAC.01313-08

URL : https://academic.oup.com/femsre/article-pdf/33/4/757/18141700/33-4-757.pdf

J. Pennington and S. Rosenberg, Spontaneous DNA breakage in single living Escherichia coli cells, Nature Genetics, vol.64, issue.6, pp.797-802, 2007.
DOI : 10.1073/pnas.0606499103

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2856310/pdf

V. Petrova, S. Chitteni-pattu, J. Drees, R. Inman, and M. Cox, An SOS Inhibitor that Binds to Free RecA Protein: The PsiB Protein, Molecular Cell, vol.36, issue.1, pp.121-130, 2009.
DOI : 10.1016/j.molcel.2009.07.026

K. Plata, S. Riosa, C. Singh, R. Rosato, and A. Rosato, Targeting of PBP1 by ??-lactams Determines recA/SOS Response Activation in Heterogeneous MRSA Clinical Strains, PLoS ONE, vol.175, issue.4, p.61083, 2013.
DOI : 10.1371/journal.pone.0061083.t004

C. Potenski and H. Klein, R We There Yet? R-Loop Hazards to Finishing the Journey, Molecular Cell, vol.44, issue.6, pp.848-850, 2011.
DOI : 10.1016/j.molcel.2011.12.003

S. Proshkin, A. Rahmouni, A. Mironov, and E. Nudler, Cooperation Between Translating Ribosomes and RNA Polymerase in Transcription Elongation, Science, vol.180, issue.3, pp.504-508, 2010.
DOI : 10.1016/0022-2836(84)90027-5

M. Quinones, H. Kimsey, and M. Waldor, LexA Cleavage Is Required for CTX Prophage Induction, Molecular Cell, vol.17, issue.2, pp.291-300, 2005.
DOI : 10.1016/j.molcel.2004.11.046

URL : https://doi.org/10.1016/j.molcel.2004.11.046

M. Radman, SOS Repair Hypothesis: Phenomenology of an Inducible DNA Repair Which is Accompanied by Mutagenesis, Basic life sciences, vol.5, pp.355-367, 1975.
DOI : 10.1007/978-1-4684-2895-7_48

P. Rai, T. Cole, D. Wemmer, and S. Linn, Localization of Fe2+ at an RTGR sequence within a DNA duplex explains preferential cleavage by Fe2+ and H2O211Edited by M. F. Summers, Journal of Molecular Biology, vol.312, issue.5, pp.1089-1101, 2001.
DOI : 10.1006/jmbi.2001.5010

R. Rastogi, . Richa, A. Kumar, M. Tyagi, and R. Sinha, Molecular Mechanisms of Ultraviolet Radiation-Induced DNA Damage and Repair, Journal of Nucleic Acids, vol.8, issue.12, p.592980, 2010.
DOI : 10.1126/science.283.5404.1001

J. Roberts and J. Park, Mfd, the bacterial transcription repair coupling factor: translocation, repair and termination, Current Opinion in Microbiology, vol.7, issue.2, pp.120-125, 2004.
DOI : 10.1016/j.mib.2004.02.014

J. Rodriguez-beltran, A. Rodriguez-rojas, J. Guelfo, A. Couce, and J. Blazquez, The Escherichia coli SOS Gene dinF Protects against Oxidative Stress and Bile Salts, PLoS ONE, vol.24, issue.4, p.34791, 2012.
DOI : 10.1371/journal.pone.0034791.t001

M. Ronen, R. Rosenberg, B. Shraiman, and U. Alon, Assigning numbers to the arrows: Parameterizing a gene regulation network by using accurate expression kinetics, Proceedings of the National Academy of Sciences, vol.11, issue.1, pp.10555-10560, 2002.
DOI : 10.1063/1.1349892

K. Satoh, H. Ohba, H. Sghaier, and I. Narumi, Down-regulation of radioresistance by LexA2 in Deinococcus radiodurans, Microbiology, vol.152, issue.11, pp.3217-3226, 2006.
DOI : 10.1099/mic.0.29139-0

K. Satoh, M. Kikuchi, A. Ishaque, H. Ohba, M. Yamada et al., The role of Deinococcus radiodurans RecFOR proteins in homologous recombination, DNA Repair, vol.11, issue.4, pp.410-418, 2012.
DOI : 10.1016/j.dnarep.2012.01.008

N. Savery, The molecular mechanism of transcription-coupled DNA repair, Trends in Microbiology, vol.15, issue.7, pp.326-333, 2007.
DOI : 10.1016/j.tim.2007.05.005

K. Schaich and D. Borg, Fenton reactions in lipid phases, Lipids, vol.59, issue.6, pp.570-579, 1988.
DOI : 10.1002/ijch.198300064

W. Schroder, C. Goerke, and C. Wolz, Opposing effects of aminocoumarins and fluoroquinolones on the SOS response and adaptability in Staphylococcus aureus, Journal of Antimicrobial Chemotherapy, vol.78, issue.6, pp.529-538, 2013.
DOI : 10.1128/IAI.00088-10

V. Sharma, Y. Sakai, K. Smythe, and Y. Yokobayashi, Knockdown of recA gene expression by artificial small RNAs in Escherichia coli, Biochemical and Biophysical Research Communications, vol.430, issue.1, pp.256-259, 2013.
DOI : 10.1016/j.bbrc.2012.10.141

N. Shechter, L. Zaltzman, A. Weiner, V. Brumfeld, E. Shimoni et al., Stress-induced Condensation of Bacterial Genomes Results in Re-pairing of Sister Chromosomes, Journal of Biological Chemistry, vol.510, issue.35, 2013.
DOI : 10.1126/science.1077865

C. Shee, R. Ponder, J. Gibson, and S. Rosenberg, What Limits the Efficiency of Double-Strand Break-Dependent Stress-Induced Mutation in <i>Escherichia coli</i>?, Journal of Molecular Microbiology and Biotechnology, vol.4, issue.1-2, pp.8-19, 2011.
DOI : 10.1371/journal.pgen.1000264

L. Singletary, J. Gibson, E. Tanner, G. Mckenzie, P. Lee et al., An SOS-Regulated Type 2 Toxin-Antitoxin System, Journal of Bacteriology, vol.191, issue.24, pp.7456-7465, 2009.
DOI : 10.1128/JB.00963-09

URL : http://jb.asm.org/content/191/24/7456.full.pdf

J. Sussenbach and W. Berends, Photodynamic degradation of guanine, Biochemical and Biophysical Research Communications, vol.16, issue.3, pp.263-266, 1964.
DOI : 10.1016/0006-291X(64)90337-7

F. Taddei, I. Matic, and M. Radman, cAMP-dependent SOS induction and mutagenesis in resting bacterial populations., Proceedings of the National Academy of Sciences, vol.92, issue.25, pp.11736-11740, 1995.
DOI : 10.1073/pnas.92.25.11736

URL : http://europepmc.org/articles/pmc40477?pdf=render

A. Tapias and J. Barbe, Mutational analysis of the Rhizobium etli recA operator, J Bacteriol, vol.180, pp.6325-6331, 1998.

C. Ubeda, E. Maiques, E. Knecht, I. Lasa, R. Novick et al., Antibiotic-induced SOS response promotes horizontal dissemination of pathogenicity island-encoded virulence factors in staphylococci, Molecular Microbiology, vol.181, issue.3, pp.836-844, 2005.
DOI : 10.1086/315239

M. Val, S. Kennedy, A. Soler-bistue, V. Barbe, C. Bouchier et al., Fuse or die: how to survive the loss of Dam in Vibrio cholerae Bacterial SOS response: a food safety perspective, Mol Microbiol. van der Veen S & Abee T Curr Opin Biotechnol, vol.22, pp.136-142, 2011.

J. Wade, N. Reppas, G. Church, and K. Struhl, Genomic analysis of LexA binding reveals the permissive nature of the Escherichia coli genome and identifies unconventional target sites, Genes & Development, vol.19, issue.21, pp.2619-2630, 2005.
DOI : 10.1101/gad.1355605

S. Wadhawan, S. Gautam, and A. Sharma, A component of gamma-radiation-induced cell death in E. coli is programmed and interlinked with activation of caspase-3 and SOS response, Archives of Microbiology, vol.183, issue.12, 2013.
DOI : 10.1128/JB.183.15.4562-4570.2001

G. Walker, The SOS Response of Escherichia coli. Escherichia coli and Salmonella, American Society of Microbiology, vol.1, pp.1400-1416, 1996.

H. Wimberly, C. Shee, P. Thornton, P. Sivaramakrishnan, S. Rosenberg et al., R-loops and nicks initiate DNA breakage and genome instability in non-growing Escherichia coli, Nature communications, vol.4, p.2115, 2013.
DOI : 10.1038/ncomms3115

URL : http://www.nature.com/articles/ncomms3115.pdf

K. Winterling, D. Chafin, J. Hayes, J. Sun, A. Levine et al., The Bacillus subtilis DinR binding site: redefinition of the consensus sequence, J Bacteriol, vol.180, pp.2201-2211, 1998.

R. Yasbin, G. Wilson, and F. Young, Transformation and transfection in lysogenic strains of Bacillus subtilis: evidence for selective induction of prophage in competent cells, J Bacteriol, vol.121, pp.296-304, 1975.

T. Yasuda, K. Morimatsu, T. Horii, T. Nagata, and H. Ohmori, Inhibition of Escherichia coli RecA coprotease activities by DinI, The EMBO Journal, vol.17, issue.11, pp.3207-3216, 1998.
DOI : 10.1093/emboj/17.11.3207

G. Yim, J. Mcclure, M. Surette, and J. Davies, Modulation of Salmonella gene expression by subinhibitory concentrations of quinolones, The Journal of Antibiotics, vol.8, issue.1, pp.73-78, 2011.
DOI : 10.1371/journal.pbio.0060280

P. Ysern, B. Clerch, M. Castano, I. Gibert, J. Barbe et al., by fluoroquinolones, Mutagenesis, vol.5, issue.1, pp.63-66, 1990.
DOI : 10.1093/mutage/5.1.63

A. Zhang, Y. Pigli, and P. Rice, Structure of the LexA???DNA complex and implications for SOS box measurement, Nature, vol.35, issue.7308, pp.883-886, 2010.
DOI : 10.1038/nature09200

J. Zhu and J. Mekalanos, Quorum Sensing-Dependent Biofilms Enhance Colonization in Vibrio cholerae, Developmental Cell, vol.5, issue.4, pp.647-656, 2003.
DOI : 10.1016/S1534-5807(03)00295-8

URL : https://doi.org/10.1016/s1534-5807(03)00295-8