J. F. Guespin-michel, Phenotypic reversion in some early blocked sporulation mutants of Bacillus subtilis. Genetic study of polymyxin resistant partial revertants, Mol Gen Genet, vol.112, issue.3, pp.243-54, 1971.

B. R. Levin, J. Concepción-acevedo, and K. I. Udekwu, Persistence: a copacetic and parsimonious hypothesis for the existence of non-inherited resistance to antibiotics. Current Opinion in Microbiology, vol.21, pp.18-21, 2014.

J. W. Bigger, The bactericidal action of penicillin on staphylococcus pyogenes, Irish Journal of Medical Science. déc, vol.19, issue.12, pp.585-95, 1944.

G. L. Hobby, K. Meyer, and E. Chaffee, Observations on the Mechanism of Action of Penicillin, Proceedings of the Society for Experimental Biology and Medicine. 1 juin, vol.50, issue.2, pp.281-286, 1942.

A. Brauner, O. Fridman, O. Gefen, and N. Q. Balaban, Distinguishing between resistance, tolerance and persistence to antibiotic treatment, Nat Rev Microbiol, vol.14, issue.5, pp.320-350, 2016.

N. Q. Balaban, J. Merrin, R. Chait, L. Kowalik, and S. Leibler, Bacterial Persistence as a Phenotypic Switch, Science. 10 sept, vol.305, issue.5690, pp.1622-1627, 2004.

S. Handwerger and A. Tomasz, Antibiotic tolerance among clinical isolates of bacteria, Annu Rev Pharmacol Toxicol, vol.25, pp.349-80, 1985.

L. A. Barry, Methods for determining bactericidal activity of antimicrobial agents; approved guideline, 1999.

K. Ishida, P. A. Guze, G. M. Kalmanson, K. Albrandt, and L. B. Guze, Variables in demonstrating methicillin tolerance in Staphylococcus aureus strains, Antimicrob Agents Chemother. avr, vol.21, issue.4, pp.688-90, 1982.

J. S. Wolfson, D. C. Hooper, G. L. Mchugh, M. A. Bozza, and M. N. Swartz, Mutants of Escherichia coli K-12 exhibiting reduced killing by both quinolone and beta-lactam antimicrobial agents, Antimicrob Agents Chemother. oct, vol.34, issue.10, pp.1938-1981, 1990.

I. Keren, N. Kaldalu, A. Spoering, Y. Wang, and K. Lewis, Persister cells and tolerance to antimicrobials, FEMS Microbiol Lett. 15 janv, vol.230, issue.1, pp.13-21, 2004.

M. B. Pasticci, A. Moretti, G. Stagni, V. Ravasio, L. Soavi et al., Bactericidal activity of oxacillin and glycopeptides against Staphylococcus aureus in patients with endocarditis: looking for a relationship between tolerance and outcome, Ann Clin Microbiol Antimicrob. 9 juin, vol.10, p.26, 2011.

O. Fridman, A. Goldberg, I. Ronin, N. Shoresh, and N. Q. Balaban, Optimization of lag time underlies antibiotic tolerance in evolved bacterial populations, Nature. sept, vol.513, issue.7518, pp.418-439, 2014.

E. Tuomanen, R. Cozens, W. Tosch, O. Zak, and A. Tomasz, The rate of killing of Escherichia coli by beta-lactam antibiotics is strictly proportional to the rate of bacterial growth, J Gen Microbiol. mai, vol.132, issue.5, pp.1297-304, 1986.

R. R. Regoes, C. Wiuff, R. M. Zappala, K. N. Garner, F. Baquero et al., Pharmacodynamic functions: a multiparameter approach to the design of antibiotic treatment regimens, Antimicrob Agents Chemother, vol.48, issue.10, pp.3670-3676, 2004.

H. K. Johansen, T. G. Jensen, R. B. Dessau, B. Lundgren, and N. Frimodt-moller, Antagonism between penicillin and erythromycin against Streptococcus pneumoniae in vitro and in vivo, J Antimicrob Chemother. déc, vol.46, issue.6, pp.973-80, 2000.

J. De-steenwinkel, G. J. De-knegt, M. T. Ten-kate, A. Van-belkum, H. A. Verbrugh et al., Time-kill kinetics of anti-tuberculosis drugs, and emergence of resistance, in relation to metabolic activity of Mycobacterium tuberculosis, J Antimicrob Chemother. déc, vol.65, issue.12, pp.2582-2591, 2010.

A. Jõers, N. Kaldalu, and T. Tenson, The Frequency of Persisters in Escherichia coli Reflects the Kinetics of Awakening from Dormancy, Journal of Bacteriology. 1 juill, vol.192, issue.13, pp.3379-84, 2010.

O. Gefen, C. Gabay, M. Mumcuoglu, G. Engel, and N. Q. Balaban, Single-cell protein induction dynamics reveals a period of vulnerability to antibiotics in persister bacteria, Proc Natl Acad Sci, vol.105, issue.16, pp.6145-6154, 2008.

N. M. Vega, K. R. Allison, A. S. Khalil, and J. J. Collins, Signaling-mediated bacterial persister formation, Nat Chem Biol. 18 mars, vol.8, issue.5, pp.431-434, 2012.

R. Hengge and . Stationary-phase, Gene Regulation in Escherichia coli §. EcoSal Plus. déc, vol.4, issue.2, 2011.

P. Landini, T. Egli, J. Wolf, and S. Lacour, sigmaS, a major player in the response to environmental stresses in Escherichia coli: role, regulation and mechanisms of promoter recognition, Environ Microbiol Rep. févr, vol.6, issue.1, pp.1-13, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00926634

K. Lewis, Persister cells, Annu Rev Microbiol, vol.64, pp.357-72, 2010.

E. Maisonneuve and K. Gerdes, Molecular Mechanisms Underlying Bacterial Persisters, Cell. avr, vol.157, issue.3, pp.539-587, 2014.

H. S. Moyed and K. P. Bertrand, hipA, a newly recognized gene of Escherichia coli K-12 that affects frequency of persistence after inhibition of murein synthesis, Journal of bacteriology, vol.155, issue.2, pp.768-775, 1983.

S. M. Amato and M. P. Brynildsen, Persister Heterogeneity Arising from a Single Metabolic Stress, Curr Biol. 17 août, vol.25, issue.16, pp.2090-2098, 2015.

. Bergh-bv-den, J. E. Michiels, T. Wenseleers, E. M. Windels, P. V. Boer et al., Frequency of antibiotic application drives rapid evolutionary adaptation of Escherichia coli persistence, Nature Microbiology. mai, vol.1, issue.5, p.16020, 2016.

K. Stepanyan, T. Wenseleers, E. A. Duéñez-guzmán, F. Muratori, B. Van-den-bergh et al., Fitness trade-offs explain low levels of persister cells in the opportunistic pathogen Pseudomonas aeruginosa, Mol Ecol. avr, vol.24, issue.7, pp.1572-83, 2015.

S. Helaine, A. M. Cheverton, K. G. Watson, L. M. Faure, S. A. Matthews et al., Internalization of Salmonella by Macrophages Induces Formation of Nonreplicating Persisters, Science. 10 janv, vol.343, issue.6167, pp.204-212, 2014.

B. Stewart and D. E. Rozen, Genetic variation for antibiotic persistence in Escherichia coli, Evolution. mars, vol.66, issue.3, pp.933-942, 2012.

N. Hofsteenge, E. Van-nimwegen, and O. K. Silander, Quantitative analysis of persister fractions suggests different mechanisms of formation among environmental isolates of E. coli, BMC Microbiology, vol.13, issue.1, p.25, 2013.

N. Wu, L. He, P. Cui, W. Wang, Y. Yuan et al., Ranking of persister genes in the same Escherichia coli genetic background demonstrates varying importance of individual persister genes in tolerance to different antibiotics, Front Microbiol, vol.6, p.1003, 2015.

M. Arnoldini, R. Mostowy, S. Bonhoeffer, and M. Ackermann, Evolution of stress response in the face of unreliable environmental signals, PLoS Comput Biol, vol.8, issue.8, p.1002627, 2012.

I. G. De-jong, P. Haccou, and O. P. Kuipers, Bet hedging or not? A guide to proper classification of microbial survival strategies, Bioessays. mars, vol.33, issue.3, pp.215-238, 2011.

E. Kussell, R. Kishony, N. Q. Balaban, and S. Leibler, Bacterial persistence: a model of survival in changing environments, Genetics. avr, vol.169, issue.4, pp.1807-1821, 2005.

E. Maisonneuve, M. Castro-camargo, and K. Gerdes, controls bacterial persistence by stochastic induction of toxin-antitoxin activity, Cell. 29 août, vol.154, issue.5, pp.1140-50, 2013.

D. Shah, Z. Zhang, A. Khodursky, N. Kaldalu, K. Kurg et al., Persisters: a distinct physiological state of E. coli, BMC Microbiol. 12 juin, vol.6, p.53, 2006.

, Stochastic induction of persister cells by HipA through (p)ppGpp-mediated activation of mRNA endonucleases

S. H. Hong, X. Wang, H. F. O'connor, M. J. Benedik, and T. K. Wood, Bacterial persistence increases as environmental fitness decreases, Microb Biotechnol. juill, vol.5, issue.4, pp.509-531, 2012.

L. W. Goneau, N. S. Yeoh, K. W. Macdonald, P. A. Cadieux, J. P. Burton et al., Selective target inactivation rather than global metabolic dormancy causes antibiotic tolerance in uropathogens, Antimicrob Agents Chemother, vol.58, issue.4, pp.2089-97, 2014.

Y. Wu, M. Vuli?, K. I. Lewis, and K. , Role of oxidative stress in persister tolerance, Antimicrob Agents Chemother. sept, vol.56, issue.9, pp.4922-4928, 2012.

V. Defraine, M. Fauvart, and J. Michiels, Fighting bacterial persistence: Current and emerging anti-persister strategies and therapeutics, Drug Resist Updat. mai, vol.38, pp.12-26, 2018.

I. Keren, D. Shah, A. Spoering, N. Kaldalu, and K. Lewis, Specialized persister cells and the mechanism of multidrug tolerance in Escherichia coli, J Bacteriol. déc, vol.186, issue.24, pp.8172-80, 2004.

K. Sato, Y. Inoue, T. Fujii, H. Aoyama, and S. Mitsuhashi, Antibacterial activity of ofloxacin and its mode of action, Infection. 1 juill, vol.14, issue.4, pp.226-256, 1986.

T. Dörr, K. Lewis, and M. Vuli?, SOS Response Induces Persistence to Fluoroquinolones in Escherichia coli, PLoS Genet [Internet]. 11 déc, vol.5, issue.12, 2009.

T. Dörr, M. Vuli?, and K. Lewis, Ciprofloxacin causes persister formation by inducing the TisB toxin in Escherichia coli, PLoS Biol. 23 févr, vol.8, issue.2, p.1000317, 2010.

Y. Pu, Z. Zhao, Y. Li, J. Zou, Q. Ma et al., Enhanced Efflux Activity Facilitates Drug Tolerance in Dormant Bacterial Cells, Mol Cell, vol.21, issue.2, pp.284-94, 2016.

K. N. Adams, K. Takaki, L. E. Connolly, H. Wiedenhoft, K. Winglee et al., Drug Tolerance in Replicating Mycobacteria Mediated by a Macrophage-Induced Efflux Mechanism, Cell. 1 avr, vol.145, issue.1, pp.39-53, 2011.

N. R. Cohen, M. A. Lobritz, and J. J. Collins, Microbial Persistence and the Road to Drug Resistance, Cell Host & Microbe. 12 juin, vol.13, issue.6, pp.632-674, 2013.

A. Mamun, A. Lombardo, M. Shee, C. Lisewski, A. M. Gonzalez et al., Identity and function of a large gene network underlying mutagenic repair of DNA breaks, Science. 7 déc, vol.338, issue.6112, pp.1344-1352, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-02173971

A. Harms, E. Maisonneuve, and K. Gerdes, Mechanisms of bacterial persistence during stress and antibiotic exposure, Science. 16 déc, vol.354, issue.6318, p.4268, 2016.

M. Cashel and J. Gallant, Two Compounds implicated in the Function of the RC Gene of Escherichia coli, Nature. mars, vol.221, issue.5183, pp.838-879, 1969.

M. F. Traxler, V. M. Zacharia, S. Marquardt, S. M. Summers, H. Nguyen et al., Discretely calibrated regulatory loops controlled by ppGpp partition gene induction across the « feast to famine » gradient in Escherichia coli, Mol Microbiol. févr, vol.79, issue.4, pp.830-875, 2011.

S. M. Amato, M. A. Orman, and M. P. Brynildsen, Metabolic control of persister formation in Escherichia coli, Mol Cell. 23 mai, vol.50, issue.4, pp.475-87, 2013.

W. Gao, K. Chua, J. K. Davies, H. J. Newton, T. Seemann et al., Two novel point mutations in clinical Staphylococcus aureus reduce linezolid susceptibility and switch on the stringent response to promote persistent infection, PLoS Pathog. 10 juin, vol.6, issue.6, p.1000944, 2010.

K. Yamanaka and M. Inouye, Growth-phase-dependent expression of cspD, encoding a member of the CspA family in Escherichia coli, J Bacteriol. août, vol.179, issue.16, pp.5126-5156, 1997.

Y. Kim and T. K. Wood, Toxins Hha and CspD and small RNA regulator Hfq are involved in persister cell formation through MqsR in Escherichia coli, Biochem Biophys Res Commun. 1 janv, vol.391, issue.1, pp.209-222, 2010.

, Cyclic AMP Receptor Protein Regulates cspD, a Bacterial Toxin Gene, in Escherichia coli

A. Nakagawa, T. Oshima, and H. Mori, Identification and characterization of a second, inducible promoter of relA in Escherichia coli, Genes Genet Syst, vol.81, issue.5, pp.299-310, 2006.

A. L. Spoering and K. Lewis, Biofilms and planktonic cells of Pseudomonas aeruginosa have similar resistance to killing by antimicrobials, J Bacteriol. déc, vol.183, issue.23, pp.6746-51, 2001.

I. Keren, S. Minami, E. Rubin, and K. Lewis, Characterization and transcriptome analysis of Mycobacterium tuberculosis persisters, MBio, vol.2, issue.3, pp.100-00111, 2011.

M. Lago, V. Monteil, T. Douche, J. Guglielmini, A. Criscuolo et al., Proteome remodelling by the stress sigma factor RpoS/?S in Salmonella: identification of small proteins and evidence for post-transcriptional regulation. Sci Rep, vol.7, p.2127, 2017.

C. Lévi-meyrueis, V. Monteil, O. Sismeiro, M. Dillies, M. Monot et al., Expanding the RpoS/?S-network by RNA sequencing and identification of ?S-controlled small RNAs in Salmonella, PLoS ONE, vol.9, issue.5, p.96918, 2014.

K. Murakami, T. Ono, D. Viducic, S. Kayama, M. Mori et al., Role for rpoS gene of Pseudomonas aeruginosa in antibiotic tolerance, FEMS Microbiol Lett. 1 janv, vol.242, issue.1, pp.161-168, 2005.

S. Hansen, K. Lewis, and M. Vuli?, Role of global regulators and nucleotide metabolism in antibiotic tolerance in Escherichia coli, Antimicrob Agents Chemother. août, vol.52, issue.8, pp.2718-2744, 2008.

P. S. Stewart, M. J. Franklin, K. S. Williamson, J. P. Folsom, L. Boegli et al., Contribution of Stress Responses to Antibiotic Tolerance in Pseudomonas aeruginosa Biofilms, Antimicrobial Agents and Chemotherapy. juill, vol.59, issue.7, pp.3838-3885, 2015.

K. G. Völzing and M. P. Brynildsen, Stationary-Phase Persisters to Ofloxacin Sustain DNA Damage and Require Repair Systems Only during Recovery, MBio. 1 sept, vol.6, issue.5, pp.731-00715, 2015.

S. P. Bernier, D. Lebeaux, A. S. Defrancesco, A. Valomon, G. Soubigou et al., Starvation, together with the SOS response, mediates high biofilm-specific tolerance to the fluoroquinolone ofloxacin, PLoS Genet, vol.9, issue.1, p.1003144, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01385427

R. Czajkowski and S. Jafra, Quenching of acyl-homoserine lactone-dependent quorum sensing by enzymatic disruption of signal molecules, Acta Biochim Pol, vol.56, issue.1, pp.1-16, 2009.

N. Möker, C. R. Dean, and J. Tao, Pseudomonas aeruginosa Increases Formation of MultidrugTolerant Persister Cells in Response to Quorum-Sensing Signaling Molecules, J Bacteriol. avr, vol.192, issue.7, pp.1946-55, 2010.

N. M. Høyland-kroghsbo, Interspecies quorum sensing as a stress-anticipation signal in E. coli, 2014.

A. Battesti, N. Majdalani, and S. Gottesman, The RpoS-mediated general stress response in Escherichia coli, Annu Rev Microbiol, vol.65, pp.189-213, 2011.

V. Leung and C. M. Lévesque, A Stress-Inducible Quorum-Sensing Peptide Mediates the Formation of Persister Cells with Noninherited Multidrug Tolerance, J Bacteriol. mai, vol.194, issue.9, pp.2265-74, 2012.

K. Gerdes, P. B. Rasmussen, and S. Molin, Unique type of plasmid maintenance function: postsegregational killing of plasmid-free cells, Proc Natl Acad Sci U S A. mai, vol.83, issue.10, pp.3116-3136, 1986.

L. Van-melderen, S. De-bast, and M. , Bacterial toxin-antitoxin systems: more than selfish entities?, PLoS Genet. mars, vol.5, issue.3, p.1000437, 2009.

P. A. Gurnev, R. Ortenberg, T. Dörr, K. Lewis, and S. M. Bezrukov, Persister-promoting bacterial toxin TisB produces anion-selective pores in planar lipid bilayers, FEBS Lett. 30 juill, vol.586, issue.16, pp.2529-2563, 2012.

D. Wilmaerts, M. Bayoumi, L. Dewachter, W. Knapen, J. T. Mika et al., The Persistence-Inducing Toxin HokB Forms Dynamic Pores That Cause ATP Leakage, MBio. 14 août, vol.9, issue.4, 2018.

R. Leplae, D. Geeraerts, R. Hallez, J. Guglielmini, P. Drèze et al., Diversity of bacterial type II toxin-antitoxin systems: a comprehensive search and functional analysis of novel families, Nucleic Acids Res. juill, vol.39, issue.13, pp.5513-5538, 2011.

W. T. Chan, M. Espinosa, and C. C. Yeo, Keeping the Wolves at Bay: Antitoxins of Prokaryotic Type II Toxin-Antitoxin Systems, Front Mol Biosci, vol.3, p.9, 2016.

M. A. Schureck, T. Maehigashi, S. J. Miles, J. Marquez, S. E. Cho et al., Structure of the Proteus vulgaris HigB-(HigA)2-HigB toxin-antitoxin complex, J Biol Chem. 10 janv, vol.289, issue.2, pp.1060-70, 2014.

A. Bøggild, N. Sofos, K. R. Andersen, A. Feddersen, A. D. Easter et al., The crystal structure of the intact E. coli RelBE toxin-antitoxin complex provides the structural basis for conditional cooperativity, Structure, vol.20, issue.10, pp.1641-1649, 2012.

G. Li, Y. Zhang, M. Inouye, and M. Ikura, Inhibitory mechanism of Escherichia coli RelERelB toxin-antitoxin module involves a helix displacement near an mRNA interferase active site, J Biol Chem. 22 mai, vol.284, issue.21, pp.14628-14664, 2009.

M. A. Schumacher, P. Balani, J. Min, N. B. Chinnam, S. Hansen et al., HipBApromoter structures reveal the basis of heritable multidrug tolerance, Nature. 6 août, vol.524, issue.7563, pp.59-64, 2015.

F. Hayes and B. K?dzierska, Regulating toxin-antitoxin expression: controlled detonation of intracellular molecular timebombs, Toxins (Basel). 15 janv, vol.6, issue.1, pp.337-58, 2014.

T. R. Blower, G. Salmond, and B. F. Luisi, Balancing at survival's edge: the structure and adaptive benefits of prokaryotic toxin-antitoxin partners, Curr Opin Struct Biol. févr, vol.21, issue.1, pp.109-127, 2011.

F. Hayes and L. Van-melderen, Toxins-antitoxins: diversity, evolution and function, Crit Rev Biochem Mol Biol, vol.46, issue.5, pp.386-408, 2011.

B. K?dzierska and F. Hayes, Emerging Roles of Toxin-Antitoxin Modules in Bacterial Pathogenesis, Molecules. 17 juin, vol.21, issue.6, 2016.

Y. Yamaguchi, J. Park, and M. Inouye, Toxin-antitoxin systems in bacteria and archaea, Annu Rev Genet, vol.45, pp.61-79, 2011.

Y. Zhang, J. Zhang, K. P. Hoeflich, M. Ikura, G. Qing et al., MazF cleaves cellular mRNAs specifically at ACA to block protein synthesis in Escherichia coli, Mol Cell, vol.12, issue.4, pp.913-936, 2003.

P. Bernard and M. Couturier, Cell killing by the F plasmid CcdB protein involves poisoning of DNA-topoisomerase II complexes, J Mol Biol. 5 août, vol.226, issue.3, pp.735-780, 1992.

J. Yuan, Y. Yamaichi, and M. K. Waldor, The three vibrio cholerae chromosome II-encoded ParE toxins degrade chromosome I following loss of chromosome II, J Bacteriol. févr, vol.193, issue.3, pp.611-620, 2011.

H. Mutschler, M. Gebhardt, R. L. Shoeman, and A. Meinhart, A Novel Mechanism of Programmed Cell Death in Bacteria by Toxin-Antitoxin Systems Corrupts Peptidoglycan Synthesis, PLoS Biol [Internet]. 22 mars, vol.9, issue.3, 2011.

N. Goeders, R. Chai, B. Chen, A. Day, and G. Salmond, Structure, Evolution, and Functions of Bacterial Type III Toxin-Antitoxin Systems, Toxins (Basel). 28 sept, vol.8, issue.10, 2016.

M. Bélanger and S. Moineau, Mutational Analysis of the Antitoxin in the Lactococcal Type III Toxin-Antitoxin System AbiQ, Appl Environ Microbiol. juin, vol.81, issue.11, pp.3848-55, 2015.

P. C. Fineran, T. R. Blower, I. J. Foulds, D. P. Humphreys, K. S. Lilley et al., The phage abortive infection system, ToxIN, functions as a protein-RNA toxin-antitoxin pair, Proc Natl Acad Sci, vol.106, issue.3, pp.894-903, 2009.

J. E. Samson, M. Bélanger, and S. Moineau, Effect of the abortive infection mechanism and type III toxin/antitoxin system AbiQ on the lytic cycle of Lactococcus lactis phages, J Bacteriol. sept, vol.195, issue.17, pp.3947-56, 2013.

H. Masuda, Q. Tan, N. Awano, K. Wu, and M. Inouye, YeeU enhances the bundling of cytoskeletal polymers of MreB and FtsZ, antagonizing the CbtA (YeeV) toxicity in Escherichia coli, Mol Microbiol. juin, vol.84, issue.5, pp.979-89, 2012.

X. Wang, D. M. Lord, H. Cheng, O. Do, S. H. Hong et al., A Novel Type V TA System Where mRNA for Toxin GhoT is Cleaved by Antitoxin GhoS, Nat Chem Biol, vol.8, issue.10, pp.855-61, 2012.

C. D. Aakre, T. N. Phung, D. Huang, and M. T. Laub, A bacterial toxin inhibits DNA replication elongation through a direct interaction with the ? sliding clamp, Mol Cell. 12 déc, vol.52, issue.5, pp.617-645, 2013.

K. Gerdes and E. Maisonneuve, Bacterial persistence and toxin-antitoxin loci, Annu Rev Microbiol, vol.66, pp.103-126, 2012.

C. F. Schuster and R. Bertram, Toxin-Antitoxin Systems of Staphylococcus aureus, Toxins (Basel), vol.05, issue.5, 2016.

J. Albrethsen, J. Agner, S. R. Piersma, P. Højrup, T. V. Pham et al., Proteomic profiling of Mycobacterium tuberculosis identifies nutrient-starvation-responsive toxinantitoxin systems, Mol Cell Proteomics. mai, vol.12, issue.5, pp.1180-91, 2013.

X. Wang and T. K. Wood, Toxin-Antitoxin Systems Influence Biofilm and Persister Cell Formation and the General Stress Response, Appl Environ Microbiol. 15 août, vol.77, issue.16, pp.5577-83, 2011.

N. Verstraeten, W. J. Knapen, C. I. Kint, V. Liebens, B. Van-den-bergh et al., Obg and Membrane Depolarization Are Part of a Microbial Bet-Hedging Strategy that Leads to Antibiotic Tolerance, Mol Cell. 2 juill, vol.59, issue.1, pp.9-21, 2015.

A. M. Cheverton, B. Gollan, M. Przydacz, C. T. Wong, A. Mylona et al., A Salmonella Toxin Promotes Persister Formation through Acetylation of tRNA, Mol Cell, vol.07, issue.1, pp.86-96, 2016.

N. Q. Balaban, K. Gerdes, K. Lewis, and J. D. Mckinney, A problem of persistence: still more questions than answers?, Nature Reviews Microbiology. août, vol.11, issue.8, pp.587-91, 2013.

F. F. Correia, D. 'onofrio, A. Rejtar, T. Li, L. Karger et al., Kinase activity of overexpressed HipA is required for growth arrest and multidrug tolerance in Escherichia coli, J Bacteriol. déc, vol.188, issue.24, pp.8360-8367, 2006.

H. S. Moyed and S. H. Broderick, Molecular cloning and expression of hipA, a gene of Escherichia coli K-12 that affects frequency of persistence after inhibition of murein synthesis, J Bacteriol. mai, vol.166, issue.2, pp.399-403, 1986.

S. B. Korch, T. A. Henderson, and T. M. Hill, Characterization of the hipA7 allele of Escherichia coli and evidence that high persistence is governed by (p)ppGpp synthesis, Mol Microbiol, vol.50, issue.4, pp.1199-213, 2003.

Q. E. Yang and T. R. Walsh, Toxin-antitoxin systems and their role in disseminating and maintaining antimicrobial resistance, FEMS Microbiol Rev. 1 mai, vol.41, issue.3, pp.343-53, 2017.

E. Germain, M. Roghanian, K. Gerdes, and E. Maisonneuve, Stochastic induction of persister cells by HipA through (p)ppGpp-mediated activation of mRNA endonucleases, Proc Natl Acad Sci, vol.112, issue.16, pp.5171-5177, 2015.

S. B. Korch and T. M. Hill, Ectopic overexpression of wild-type and mutant hipA genes in Escherichia coli: effects on macromolecular synthesis and persister formation, J Bacteriol. juin, vol.188, issue.11, pp.3826-3862, 2006.

E. Germain, D. Castro-roa, N. Zenkin, and K. Gerdes, Molecular mechanism of bacterial persistence by HipA, Mol Cell, vol.52, issue.2, pp.248-54, 2013.

D. O. Osbourne, V. Soo, I. Konieczny, and T. K. Wood, Polyphosphate, cyclic AMP, guanosine tetraphosphate, and c-di-GMP reduce in vitro Lon activity, Bioengineered. août, vol.5, issue.4, pp.264-272, 2014.

Y. Shan, D. Lazinski, S. Rowe, A. Camilli, and K. Lewis, Genetic Basis of Persister Tolerance to Aminoglycosides in Escherichia coli, avr, vol.6, issue.7, 2015.

N. Chowdhury, B. W. Kwan, L. C. Mcgibbon, P. Babitzke, and T. K. Wood, Toxin MqsR cleaves single-stranded mRNA with various 5, ends. Microbiologyopen, vol.5, issue.3, pp.370-377, 2016.

Z. Baharoglu and D. Mazel, SOS, the formidable strategy of bacteria against aggressions, FEMS Microbiol Rev. nov, vol.38, issue.6, pp.1126-1171, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01423593

E. Wagner and C. Unoson, The toxin-antitoxin system tisB-istR1: Expression, regulation, and biological role in persister phenotypes, RNA Biol. déc, vol.9, issue.12, pp.1513-1522, 2012.

J. Vogel, L. Argaman, E. Wagner, and S. Altuvia, The small RNA IstR inhibits synthesis of an SOS-induced toxic peptide, Curr Biol. 29 déc, vol.14, issue.24, pp.2271-2277, 2004.

C. Unoson and E. Wagner, A small SOS-induced toxin is targeted against the inner membrane in Escherichia coli, Mol Microbiol, vol.70, issue.1, pp.258-70, 2008.

F. Darfeuille, C. Unoson, J. Vogel, and E. Wagner, An antisense RNA inhibits translation by competing with standby ribosomes, Mol Cell. 11 mai, vol.26, issue.3, pp.381-92, 2007.

S. Brantl and N. Jahn, sRNAs in bacterial type I and type III toxin-antitoxin systems, FEMS Microbiol Rev. mai, vol.39, issue.3, pp.413-440, 2015.

R. Page and W. Peti, Toxin-antitoxin systems in bacterial growth arrest and persistence, Nature Chemical Biology. 18 mars, vol.12, issue.4, pp.208-222, 2016.

D. Nguyen, A. Joshi-datar, F. Lepine, E. Bauerle, O. Olakanmi et al., Active starvation responses mediate antibiotic tolerance in biofilms and nutrient-limited bacteria, Science, vol.334, issue.6058, pp.982-988, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-00722468

B. Pascoe, L. Dams, T. S. Wilkinson, L. G. Harris, O. Bodger et al., Dormant Cells of Staphylococcus aureus Are Resuscitated by Spent Culture Supernatant, PLoS One [Internet]. 11 févr, vol.9, issue.2, 2014.

L. Laureti, I. Matic, and A. Gutierrez, Bacterial Responses and Genome Instability Induced by Subinhibitory Concentrations of Antibiotics. Antibiotics (Basel). 14 mars, vol.2, pp.100-114, 2013.

Z. Baharoglu and D. Mazel, Vibrio cholerae Triggers SOS and Mutagenesis in Response to a Wide Range of Antibiotics: a Route towards Multiresistance?, Antimicrob Agents Chemother. mai, vol.55, issue.5, pp.2438-2479, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-01423615

Z. Baharoglu, E. Krin, and D. Mazel, RpoS plays a central role in the SOS induction by sublethal aminoglycoside concentrations in Vibrio cholerae, PLoS Genet, vol.9, issue.4, p.1003421, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01691109

S. Helaine, J. A. Thompson, K. G. Watson, M. Liu, C. Boyle et al., Dynamics of intracellular bacterial replication at the single cell level, Proc Natl Acad Sci USA. 23 févr, vol.107, issue.8, pp.3746-51, 2010.

J. L. Blanchard, W. Wholey, E. M. Conlon, and P. J. Pomposiello, Rapid changes in gene expression dynamics in response to superoxide reveal SoxRS-dependent and independent transcriptional networks, PLoS ONE, vol.2, issue.11, p.1186, 2007.

D. Touati, Sensing and protecting against superoxide stress in Escherichia coli--how many ways are there to trigger soxRS response?, Redox Rep, vol.5, issue.5, pp.287-93, 2000.

C. Migdal and M. Serres, Espèces réactives de l'oxygène et stress oxydant. médecine/sciences, avr, vol.27, issue.4, pp.405-417, 2011.

K. Pedersen, S. K. Christensen, and K. Gerdes, Rapid induction and reversal of a bacteriostatic condition by controlled expression of toxins and antitoxins, Mol Microbiol. juill, vol.45, issue.2, pp.501-511, 2002.

P. J. Pomposiello and B. Demple, Global adjustment of microbial physiology during free radical stress, Adv Microb Physiol, vol.46, pp.319-360, 2002.

M. Khakimova, H. G. Ahlgren, J. J. Harrison, A. M. English, and D. Nguyen, The stringent response controls catalases in Pseudomonas aeruginosa and is required for hydrogen peroxide and antibiotic tolerance, J Bacteriol. mai, vol.195, issue.9, pp.2011-2031, 2013.

T. Bjarnsholt, The role of bacterial biofilms in chronic infections, APMIS Suppl. mai, issue.136, pp.1-51, 2013.

D. Lebeaux, J. Ghigo, and C. Beloin, Biofilm-related infections: bridging the gap between clinical management and fundamental aspects of recalcitrance toward antibiotics, Microbiol Mol Biol Rev. sept, vol.78, issue.3, pp.510-553, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01370744

T. Bjarnsholt, K. Buhlin, Y. F. Dufrêne, M. Gomelsky, A. Moroni et al., Biofilm formation -what we can learn from recent developments, J Intern Med. 1 juin, 2018.

P. S. Stewart, Antimicrobial Tolerance in Biofilms. Microbiol Spectr. juin, vol.3, issue.3, 2015.

H. Liu, Y. Xiao, H. Nie, Q. Huang, and W. Chen, Influence of (p)ppGpp on biofilm regulation in Pseudomonas putida KT2440. Microbiol Res, vol.204, pp.1-8, 2017.

G. Brackman and T. Coenye, Quorum sensing inhibitors as anti-biofilm agents, Curr Pharm Des, vol.21, issue.1, pp.5-11, 2015.

M. Fauvart, V. N. De-groote, and J. Michiels, Role of persister cells in chronic infections: clinical relevance and perspectives on anti-persister therapies, Journal of Medical Microbiology, vol.60, issue.6, pp.699-709, 2011.

S. M. Amato and M. P. Brynildsen, Nutrient Transitions Are a Source of Persisters in Escherichia coli Biofilms, PLoS One [Internet]. 25 mars, vol.9, issue.3, 2014.

J. E. Michiels, B. Van-den-bergh, N. Verstraeten, and J. Michiels, Molecular mechanisms and clinical implications of bacterial persistence, Drug Resist Updat, vol.29, pp.76-89, 2016.

A. Gutierrez, S. Jain, P. Bhargava, M. Hamblin, M. A. Lobritz et al., Understanding and Sensitizing Density-Dependent Persistence to Quinolone Antibiotics, Mol Cell. 21 déc, vol.68, issue.6, pp.1147-1154, 2017.

D. Fung, E. Chan, M. L. Chin, and R. Chan, Delineation of a bacterial starvation stress response network which can mediate antibiotic tolerance development, Antimicrob Agents Chemother. mars, vol.54, issue.3, pp.1082-93, 2010.

J. C. Betts, P. T. Lukey, L. C. Robb, R. A. Mcadam, and K. Duncan, Evaluation of a nutrient starvation model of Mycobacterium tuberculosis persistence by gene and protein expression profiling, Mol Microbiol. févr, vol.43, issue.3, pp.717-748, 2002.

B. P. Conlon, S. E. Rowe, A. B. Gandt, A. S. Nuxoll, N. P. Donegan et al., Persister formation in Staphylococcus aureus is associated with ATP depletion, vol.1, 2016.

Y. Li and Y. Zhang, PhoU Is a Persistence Switch Involved in Persister Formation and Tolerance to Multiple Antibiotics and Stresses in Escherichia coli, Antimicrob Agents Chemother. juin, vol.51, issue.6, pp.2092-2101, 2007.

R. A. Proctor, C. Von-eiff, B. C. Kahl, K. Becker, P. Mcnamara et al., Small colony variants: a pathogenic form of bacteria that facilitates persistent and recurrent infections, Nat Rev Microbiol. avr, vol.4, issue.4, pp.295-305, 2006.

R. A. Proctor, A. Kriegeskorte, B. C. Kahl, K. Becker, B. Löffler et al., Staphylococcus aureus Small Colony Variants (SCVs): a road map for the metabolic pathways involved in persistent infections. Front Cell Infect Microbiol

S. M. Amato, C. H. Fazen, T. C. Henry, W. Mok, M. A. Orman et al., The role of metabolism in bacterial persistence, Front Microbiol, vol.5, p.70, 2014.

W. Van-schaik and T. Abee, The role of sigmaB in the stress response of Gram-positive bacteria --targets for food preservation and safety, Curr Opin Biotechnol. avr, vol.16, issue.2, pp.218-242, 2005.

L. A. Onyango, H. Dunstan, R. Roberts, T. K. Macdonald, M. M. Gottfries et al., Phenotypic variants of staphylococci and their underlying population distributions following exposure to stress, PLoS ONE, vol.8, issue.10, p.77614, 2013.

P. Sendi and R. A. Proctor, Staphylococcus aureus as an intracellular pathogen: the role of small colony variants, Trends Microbiol. févr, vol.17, issue.2, pp.54-62, 2009.

T. K. Lu and J. J. Collins, Engineered bacteriophage targeting gene networks as adjuvants for antibiotic therapy, Proc Natl Acad Sci U S A. 24 mars, vol.106, issue.12, pp.4629-4663, 2009.

B. C. Kahl, K. Becker, and B. Löffler, Clinical Significance and Pathogenesis of Staphylococcal Small Colony Variants in Persistent Infections, Clin Microbiol Rev. avr, vol.29, issue.2, pp.401-428, 2016.

M. D. Lafleur, Q. Qi, and K. Lewis, Patients with long-term oral carriage harbor high-persister mutants of Candida albicans, Antimicrob Agents Chemother. janv, vol.54, issue.1, pp.39-44, 2010.

L. R. Mulcahy, J. L. Burns, S. Lory, and K. Lewis, Emergence of Pseudomonas aeruginosa Strains Producing High Levels of Persister Cells in Patients with Cystic Fibrosis, J Bacteriol. 1 déc, vol.192, issue.23, pp.6191-6200, 2010.

N. Dhar and J. D. Mckinney, Microbial phenotypic heterogeneity and antibiotic tolerance, Curr Opin Microbiol. févr, vol.10, issue.1, pp.30-38, 2007.

E. Wexselblatt, Y. Oppenheimer-shaanan, I. Kaspy, N. London, O. Schueler-furman et al., Novel Antibacterial Agent Targeting the Stringent Response, PLoS Pathog, vol.8, issue.9, 2012.

M. A. Orman and M. P. Brynildsen, Dormancy is not necessary or sufficient for bacterial persistence, Antimicrob Agents Chemother. juill, vol.57, issue.7, pp.3230-3239, 2013.

M. A. Orman and M. P. Brynildsen, Persister formation in Escherichia coli can be inhibited by treatment with nitric oxide, Free Radic Biol Med. avr, vol.93, pp.145-54, 2016.

Y. Hu, A. Shamaei-tousi, Y. Liu, and A. Coates, A New Approach for the Discovery of Antibiotics by Targeting Non-Multiplying Bacteria: A Novel Topical Antibiotic for Staphylococcal Infections, PLoS One [Internet]. 27 juill, vol.5, issue.7, 2010.

Y. Hu and A. Coates, Enhancement by novel anti-methicillin-resistant Staphylococcus aureus compound HT61 of the activity of neomycin, gentamicin, mupirocin and chlorhexidine: in vitro and in vivo studies, J Antimicrob Chemother. févr, vol.68, issue.2, pp.374-84, 2013.

V. Liebens, V. Defraine, W. Knapen, T. Swings, S. Beullens et al., 4-Dichlorophenethyl)Amino)-3-Phenoxypropan-2-ol, a Novel Antibacterial Compound Active against Persisters of Pseudomonas aeruginosa, Antimicrob Agents Chemother [Internet]. 24 août, vol.1, issue.2, 2017.

M. Y. Cruz-muñiz, L. E. López-jacome, M. Hernández-durán, R. Franco-cendejas, P. Liconalimón et al., Repurposing the anticancer drug mitomycin C for the treatment of persistent Acinetobacter baumannii infections, Int J Antimicrob Agents. janv, vol.49, issue.1, pp.88-92, 2017.

R. Schuch, H. M. Lee, B. C. Schneider, K. L. Sauve, C. Law et al., Combination therapy with lysin CF-301 and antibiotic is superior to antibiotic alone for treating methicillin-resistant Staphylococcus aureus-induced murine bacteremia, J Infect Dis. 1 mai, vol.209, issue.9, pp.1469-78, 2014.

R. Schuch, B. K. Khan, A. Raz, J. A. Rotolo, and M. Wittekind, Bacteriophage Lysin CF-301, a Potent Antistaphylococcal Biofilm Agent, Antimicrob Agents Chemother, issue.7, p.61, 2017.

N. Ooi, K. Miller, J. Hobbs, W. Rhys-williams, W. Love et al., XF-73, a novel antistaphylococcal membrane-active agent with rapid bactericidal activity, J Antimicrob Chemother, vol.64, issue.4, pp.735-775, 2009.

A. J. Lenaerts, V. Gruppo, K. S. Marietta, C. M. Johnson, D. K. Driscoll et al., Preclinical testing of the nitroimidazopyran PA-824 for activity against Mycobacterium tuberculosis in a series of in vitro and in vivo models, Antimicrob Agents Chemother. juin, vol.49, issue.6, pp.2294-301, 2005.

R. Singh, U. Manjunatha, H. Boshoff, Y. H. Ha, P. Niyomrattanakit et al., PA-824 kills nonreplicating Mycobacterium tuberculosis by intracellular NO release, Science, vol.322, issue.5906, pp.1392-1397, 2008.

R. Dawson, A. H. Diacon, D. Everitt, C. Van-niekerk, P. R. Donald et al., Efficiency and safety of the combination of moxifloxacin, pretomanid (PA-824), and pyrazinamide during the first 8 weeks of antituberculosis treatment: a phase 2b, openlabel, partly randomised trial in patients with drug-susceptible or drug-resistant pulmonary tuberculosis, Lancet. 2 mai, vol.385, issue.9979, pp.1738-1785, 2015.

J. Kim, P. Heo, T. Yang, K. Lee, D. Cho et al., Selective Killing of Bacterial Persisters by a Single Chemical Compound without Affecting Normal Antibiotic-Sensitive Cells?, Antimicrob Agents Chemother, vol.55, issue.11, pp.5380-5383, 2011.

C. Marques, A. Morozov, P. Planzos, and H. M. Zelaya, The Fatty Acid Signaling Molecule cis-2-Decenoic Acid Increases Metabolic Activity and Reverts Persister Cells to an Antimicrobial-Susceptible State, Appl Environ Microbiol, vol.80, issue.22, pp.6976-91, 2014.

K. R. Allison, M. P. Brynildsen, and J. J. Collins, Metabolite-Enabled Eradication of Bacterial Persisters by Aminoglycosides, Nature. 12 mai, vol.473, issue.7346, pp.216-236, 2011.

J. R. Morones-ramirez, J. A. Winkler, C. S. Spina, and J. J. Collins, Silver Enhances Antibiotic Activity Against Gram-negative Bacteria, Sci Transl Med. 19 juin, vol.5, pp.190-81, 0190.

J. Feng, S. Zhang, W. Shi, and Y. Zhang, Ceftriaxone Pulse Dosing Fails to Eradicate BiofilmLike Microcolony B. burgdorferi Persisters Which Are Sterilized by Daptomycin/ Doxycycline/Cefuroxime without Pulse Dosing, Front Microbiol, vol.7, p.1744, 2016.

J. Feng, W. Shi, S. Zhang, and Y. Zhang, Persister mechanisms in Borrelia burgdorferi: implications for improved intervention, Emerg Microbes Infect. août, vol.4, issue.8, p.51, 2015.

P. Cui, T. Xu, W. Zhang, and Y. Zhang, Molecular mechanisms of bacterial persistence and phenotypic antibiotic resistance, Yi Chuan, vol.20, issue.10, pp.859-71, 2016.

S. L. Chua, J. Yam, P. Hao, S. S. Adav, M. M. Salido et al., Selective labelling and eradication of antibiotic-tolerant bacterial populations in Pseudomonas aeruginosa biofilms, Nat Commun. 19 févr, vol.7, p.10750, 2016.

H. L. Torrey, K. I. Via, L. E. Lee, J. S. Lewis, and K. , High Persister Mutants in Mycobacterium tuberculosis, Kaufmann GF, éditeur. PLOS ONE. 13 mai, vol.11, issue.5, p.155127, 2016.

Y. Wakamoto, N. Dhar, R. Chait, K. Schneider, F. Signorino-gelo et al., Dynamic persistence of antibiotic-stressed mycobacteria, Science. 4 janv, vol.339, issue.6115, pp.91-96, 2013.

B. Van-den-bergh, M. Fauvart, and J. Michiels, Formation, physiology, ecology, evolution and clinical importance of bacterial persisters, FEMS Microbiology Reviews. 1 mai, vol.41, issue.3, pp.219-51, 2017.

P. Kaiser, R. R. Regoes, T. Dolowschiak, S. Y. Wotzka, J. Lengefeld et al., Cecum Lymph Node Dendritic Cells Harbor Slow-Growing Bacteria Phenotypically Tolerant to Antibiotic Treatment, PLOS Biology. févr, vol.12, issue.2, p.1001793, 2014.

B. Claudi, P. Spröte, A. Chirkova, N. Personnic, J. Zankl et al., Phenotypic Variation of Salmonella in Host Tissues Delays Eradication by Antimicrobial Chemotherapy, Cell. 14 août, vol.158, issue.4, pp.722-755, 2014.

N. Dhar and G. Manina, Single-cell analysis of mycobacteria using microfluidics and timelapse microscopy, Methods Mol Biol, vol.1285, pp.241-56, 2015.