Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Distinguishing between resistance, tolerance and persistence to antibiotic treatment

Abstract

Antibiotic tolerance is associated with the failure of antibiotic treatment and the relapse of many bacterial infections. However, unlike resistance, which is commonly measured using the minimum inhibitory concentration (MIC) metric, tolerance is poorly characterized, owing to the lack of a similar quantitative indicator. This may lead to the misclassification of tolerant strains as resistant, or vice versa, and result in ineffective treatments. In this Opinion article, we describe recent studies of tolerance, resistance and persistence, outlining how a clear and distinct definition for each phenotype can be developed from these findings. We propose a framework for classifying the drug response of bacterial strains according to these definitions that is based on the measurement of the MIC together with a recently defined quantitative indicator of tolerance, the minimum duration for killing (MDK). Finally, we discuss genes that are associated with increased tolerance — the 'tolerome' — as targets for treating tolerant bacterial strains.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Characteristic drug responses of resistance, tolerance and persistence.
Figure 2: Tolerance arises from slow growth or lag phase.

Similar content being viewed by others

References

  1. McKeegan, K. S., Borges-Walmsley, M. I. & Walmsley, A. R. Microbial and viral drug resistance mechanisms. Trends Microbiol. 10, S8–S14 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Scholar, E. M. & Pratt, W. B. (eds) The Antimicrobial Drugs (Oxford Univ. Press, 2000).

    Google Scholar 

  3. D'Costa, V. M., McGrann, K. M., Hughes, D. W. & Wright, G. D. Sampling the antibiotic resistome. Science 311, 374–377 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Bigger, J. W. Treatment of staphylococcal infections with penicillin by intermittent sterilisation. Lancet 244, 497–500 (1944).

    Article  Google Scholar 

  5. Hobby, G. L., Meyer, K. & Chaffee, E. Observations on the mechanism of action of penicillin. Proc. Soc. Exp. Biol. Med. 50, 281–285 (1942).

    Article  CAS  Google Scholar 

  6. Horne, D. & Tomasz, A. Tolerant response of Streptococcus sanguis to β-lactams and other cell-wall inhibitors. Antimicrob. Agents Chemother. 11, 888–896 (1977).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Balaban, N. Q., Gerdes, K., Lewis, K. & McKinney, J. D. A problem of persistence: still more questions than answers? Nat. Rev. Microbiol. 11, 587–591 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Kester, J. C. & Fortune, S. M. Persisters and beyond: mechanisms of phenotypic drug resistance and drug tolerance in bacteria. Crit. Rev. Biochem. Mol. Biol. 49, 91–101 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Handwerger, S. & Tomasz, A. Antibiotic tolerance among clinical isolates of bacteria. Annu. Rev. Pharmacol. Toxicol. 25, 349–380 (1985).

    Article  CAS  PubMed  Google Scholar 

  10. Tuomanen, E., Cozens, R., Tosch, W., Zak, O. & Tomasz, A. The rate of killing of Escherichia coli by β-lactam antibiotics is strictly proportional to the rate of bacterial growth. J. Gen. Microbiol. 132, 1297–1304 (1986).

    CAS  PubMed  Google Scholar 

  11. McDermott, W. Microbial persistence. Yale J. Biol. Med. 30, 257–291 (1958).

    CAS  PubMed  Google Scholar 

  12. Lederberg, J. & Zinder, N. Concentration of biochemical mutants of bacteria with penicillin. J. Am. Chem. Soc. 70, 4267–4268 (1948).

    Article  CAS  PubMed  Google Scholar 

  13. Gefen, O. & Balaban, N. Q. The importance of being persistent: heterogeneity of bacterial populations under antibiotic stress. FEMS Microbiol. Rev. 33, 704–717 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Balaban, N. Q., Merrin, J., Chait, R., Kowalik, L. & Leibler, S. Bacterial persistence as a phenotypic switch. Science 305, 1622–1625 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Wakamoto, Y. et al. Dynamic persistence of antibiotic-stressed mycobacteria. Science 339, 91–95 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Depardieu, F., Podglajen, I., Leclercq, R., Collatz, E. & Courvalin, P. Modes and modulations of antibiotic resistance gene expression. Clin. Microbiol. Rev. 20, 79–114 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Blair, J. M., Webber, M. A., Baylay, A. J., Ogbolu, D. O. & Piddock, L. J. Molecular mechanisms of antibiotic resistance. Nat. Rev. Microbiol. 13, 42–51 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Chait, R., Craney, A. & Kishony, R. Antibiotic interactions that select against resistance. Nature 446, 668–671 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Wiegand, I., Hilpert, K. & Hancock, R. E. Agar and broth dilution methods to determine the minimal inhibitory concentration (MIC) of antimicrobial substances. Nat. Protoc. 3, 163–175 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Mattie, H. Antibiotic efficacy in vivo predicted by in vitro activity. Int. J. Antimicrob. Agents 14, 91–98 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Paterson, D. L. et al. Outcome of cephalosporin treatment for serious infections due to apparently susceptible organisms producing extended-spectrum β-lactamases: implications for the clinical microbiology laboratory. J. Clin. Microbiol. 39, 2206–2212 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ishida, K., Guze, P. A., Kalmanson, G. M., Albrandt, K. & Guze, L. B. Variables in demonstrating methicillin tolerance in Staphylococcus aureus strains. Antimicrob. Agents Chemother. 21, 688–690 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wolfson, J., Hooper, D., McHugh, G., Bozza, M. & Swartz, M. Mutants of Escherichia coli K-12 exhibiting reduced killing by both quinolone and β-lactam antimicrobial agents. Antimicrob. Agents Chemother. 34, 1938–1943 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mueller, M., de la Pena, A. & Derendorf, H. Issues in pharmacokinetics and pharmacodynamics of anti-infective agents: kill curves versus MIC. Antimicrob. Agents Chemother. 48, 369–377 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Barry, L. A. et al. Methods for determining bactericidal activity of antimicrobial agents; approved guideline. (National Committee for Clinical Laboratory Standards, 1999).

    Google Scholar 

  26. Keren, I., Kaldalu, N., Spoering, A., Wang, Y. P. & Lewis, K. Persister cells and tolerance to antimicrobials. Fems Microbiol. Lett. 230, 13–18 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Pasticci, M. B. 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. 10, 26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fridman, O., Goldberg, A., Ronin, I., Shoresh, N. & Balaban, N. Q. Optimization of lag time underlies antibiotic tolerance in evolved bacterial populations. Nature 513, 418–421 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Regoes, R. R. et al. Pharmacodynamic functions: a multiparameter approach to the design of antibiotic treatment regimens. Antimicrob. Agents Chemother. 48, 3670–3676 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gefen, O., Gabay, C., Mumcuoglu, M., Engel, G. & Balaban, N. Q. Single-cell protein induction dynamics reveals a period of vulnerability to antibiotics in persister bacteria. Proc. Natl Acad. Sci. USA 105, 6145–6149 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Helaine, S. et al. Dynamics of intracellular bacterial replication at the single cell level. Proc. Natl Acad. Sci. USA 107, 3746–3751 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Amato, S. M., Orman, M. A. & Brynildsen, M. P. Metabolic control of persister formation in Escherichia coli. Mol. Cell 50, 475–487 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Maisonneuve, E., Castro-Camargo, M. & Gerdes, K. (p)ppGpp controls bacterial persistence by stochastic induction of toxin–antitoxin activity. Cell 154, 1140–1150 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Chao, L. & Levin, B. R. Structured habitats and the evolution of anticompetitor toxins in bacteria. Proc. Natl Acad. Sci. USA 78, 6324–6328 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Rodionov, D. G. & Ishiguro, E. E. Effects of inhibitors of protein synthesis on lysis of Escherichia coli induced by β-lactam antibiotics. Antimicrob. Agents Chemother. 40, 899–903 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Orman, M. A. & Brynildsen, M. P. Dormancy is not necessary or sufficient for bacterial persistence. Antimicrob. Agents Chemother. 57, 3230–3239 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Johansen, H. K., Jensen, T. G., Dessau, R. B., Lundgren, B. & Frimodt-Moller, N. Antagonism between penicillin and erythromycin against Streptococcus pneumoniae in vitro and in vivo. J. Antimicrob. Chemother. 46, 973–980 (2000).

    Article  CAS  PubMed  Google Scholar 

  38. Thonus, I. P., Fontijne, P. & Michel, M. F. Ampicillin susceptibility and ampicillin-induced killing rate of Escherichia coli. Antimicrob. Agents Chemother. 22, 386–390 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mascio, C. T., Alder, J. D. & Silverman, J. A. Bactericidal action of daptomycin against stationary-phase and nondividing Staphylococcus aureus cells. Antimicrob. Agents Chemother. 51, 4255–4260 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. de Steenwinkel, J. E. et al. Time–kill kinetics of anti-tuberculosis drugs, and emergence of resistance, in relation to metabolic activity of Mycobacterium tuberculosis. J. Antimicrob. Chemother. 65, 2582–2589 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Evans, D. J., Allison, D. G., Brown, M. R. & Gilbert, P. Susceptibility of Pseudomonas aeruginosa and Escherichia coli biofilms towards ciproflaxin: effect of specific growth rate. J. Antimicrob. Chemother. 27, 177–184 (1991).

    Article  CAS  PubMed  Google Scholar 

  42. Manina, G., Dhar, N. & McKinney, J. D. Stress and host immunity amplify Mycobacterium tuberculosis phenotypic heterogeneity and induce nongrowing metabolically active forms. Cell Host Microbe 17, 32–46 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Kitano, K. & Tomasz, A. Escherichia coli mutants tolerant to β-lactam antibiotics. J. Bacteriol. 140, 955–963 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bernier, S. P. et al. Starvation, together with the SOS response, mediates high biofilm-specific tolerance to the fluoroquinolone ofloxacin. PloS Genet. 9, e1003144 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sandberg, A. et al. Intra- and extracellular activities of dicloxacillin against Staphylococcus aureus in vivo and in vitro. Antimicrob. Agents Chemother. 54, 2391–2400 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dorr, T., Davis, B. M. & Waldor, M. K. Endopeptidase-mediated β-lactam tolerance. PloS Pathog. 11, e1004850 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Dorr, T., Vulic, M. & Lewis, K. Ciprofloxacin causes persister formation by inducing the TisB toxin in Escherichia coli. PloS Biol. 8, e1000317 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Wiuff, C. & Andersson, D. I. Antibiotic treatment in vitro of phenotypically tolerant bacterial populations. J. Antimicrob. Chemother. 59, 254–263 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Johnson, P. J. T. & Levin, B. R. Pharmacodynamics, population dynamics, and the evolution of persistence in Staphylococcus aureus. PloS Genet. 9, e1003123 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Gefen, O., Fridman, O., Ronin, I. & Balaban, N. Q. Direct observation of single stationary-phase bacteria reveals a surprisingly long period of constant protein production activity. Proc. Natl Acad. Sci. USA 111, 556–561 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Lewis, K. Persister cells, dormancy and infectious disease. Nat. Rev. Microbiol. 5, 48–56 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Nguyen, D. et al. Active starvation responses mediate antibiotic tolerance in biofilms and nutrient-limited bacteria. Science 334, 982–986 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Levin-Reisman, I. et al. Automated imaging with ScanLag reveals previously undetectable bacterial growth phenotypes. Nat. Methods 7, 737–739 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Luidalepp, H., Joers, A., Kaldalu, N. & Tenson, T. Age of inoculum strongly influences persister frequency and can mask effects of mutations implicated in altered persistence. J. Bacteriol. 193, 3598–3605 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Madar, D. et al. Promoter activity dynamics in the lag phase of Escherichia coli. BMC Syst. Biol. 7, 136 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Joers, A., Kaldalu, N. & Tenson, T. The frequency of persisters in Escherichia coli reflects the kinetics of awakening from dormancy. J. Bacteriol. 192, 3379–3384 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Putrinš, M., Kogermann, K., Lukk, E. & Lippus, M. Phenotypic heterogeneity enables uropathogenic Escherichia coli to evade killing by antibiotics and serum complement. Infect. Immun. 83, 1056–1067 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Pearl, S., Gabay, C., Kishony, R., Oppenheim, A. & Balaban, N. Q. Nongenetic individuality in the host–phage interaction. PloS Biol. 6, 957–964 (2008).

    Article  CAS  Google Scholar 

  59. Baranyi, J. Stochastic modelling of bacterial lag phase. Int. J. Food Microbiol. 73, 203–206 (2002).

    Article  PubMed  Google Scholar 

  60. Akerlund, T., Nordstrom, K. & Bernander, R. Analysis of cell size and DNA content in exponentially growing and stationary-phase batch cultures of Escherichia coli. J. Bacteriol. 177, 6791–6797 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hartman, B. J. & Tomasz, A. Expression of methicillin resistance in heterogeneous strains of Staphylococcus aureus. Antimicrob. Agents Chemother. 29, 85–92 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Levin, B. R. & Rozen, D. E. Non-inherited antibiotic resistance. Nat. Rev. Microbiol. 4, 556–562 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Nataro, J. P., Blaser, M. J. & Cunningham-Rundles, S. (eds) in Persistent Bacterial Infections. 3–10 (ASM Press, 2000).

    Book  Google Scholar 

  64. Rotem, E. et al. Regulation of phenotypic variability by a threshold-based mechanism underlies bacterial persistence. Proc. Natl Acad. Sci. USA 107, 12541–12546 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Korch, S. B. & Hill, T. M. Ectopic overexpression of wild-type and mutant hipA genes in Escherichia coli: effects on macromolecular synthesis and persister formation. J. Bacteriol. 188, 3826–3836 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Moyed, H. S. & Bertrand, K. P. hipA, a newly recognized gene of Escherichia coli K-12 that affects frequency of persistence after inhibition of murein synthesis. J. Bacteriol. 155, 768–775 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Levin-Reisman, I. & Balaban, N. Q. in Bacterial Persistence: Methods and Protocols (eds Michiels, J. & Fauvart, M.) 75–81 (Humana Press, 2015).

    Google Scholar 

  68. El Meouche, I., Siu, Y. & Dunlop, M. J. Stochastic expression of a multiple antibiotic resistance activator confers transient resistance in single cells. Sci. Rep. 6, 19538 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Adams, K. N. et al. Drug tolerance in replicating mycobacteria mediated by a macrophage-induced efflux mechanism. Cell 145, 39–53 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kayser, F. H., Benner, E. J. & Hoeprich, P. D. Acquired and native resistance of Staphylococcus aureus to cephalexin and other β-lactam antibiotics. Appl. Microbiol. 20, 1–5 (1970).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. El-Halfawy, O. M. & Valvano, M. A. Antimicrobial heteroresistance: an emerging field in need of clarity. Clin. Microbiol. Rev. 28, 191–207 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Adams, K. N., Szumowski, J. D. & Ramakrishnan, L. Verapamil, and its metabolite norverapamil, inhibit macrophage-induced, bacterial efflux pump-mediated tolerance to multiple anti-tubercular drugs. J. Infect. Dis. 210, 456–466 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Mattie, H., Sekh, B. A., van Ogtrop, M. L. & van Strijen, E. Comparison of the antibacterial effects of cefepime and ceftazidime against Escherichia coli in vitro and in vivo. Antimicrob. Agents Chemother. 36, 2439–2443 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Coates, A. R. & Hu, Y. Targeting non-multiplying organisms as a way to develop novel antimicrobials. Trends Pharmacol. Sci. 29, 143–150 (2008).

    Article  CAS  PubMed  Google Scholar 

  75. Feng, J. et al. Identification of novel activity against Borrelia burgdorferi persisters using an FDA approved drug library. Emerg. Microbes Infect. 3, e49 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kim, J. S. et al. Selective killing of bacterial persisters by a single chemical compound without affecting normal antibiotic-sensitive cells. Antimicrob. Agents Chemother. 55, 5380–5383 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Fleck, L. E. et al. A screen for and validation of prodrug antimicrobials. Antimicrob. Agents Chemother. 58, 1410–1419 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Conlon, B. P. et al. Activated ClpP kills persisters and eradicates a chronic biofilm infection. Nature 503, 365–370 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Roostalu, J., Jõers, A., Luidalepp, H., Kaldalu, N. & Tenson, T. Cell division in Escherichia coli cultures monitored at single cell resolution. BMC Microbiol. 8, 68 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Claudi, B. et al. Phenotypic variation of Salmonella in host tissues delays eradication by antimicrobial chemotherapy. Cell 158, 722–733 (2014).

    Article  CAS  PubMed  Google Scholar 

  81. Mattie, H., Zhang, L. C., van Strijen, E., Sekh, B. R. & Douwes-Idema, A. E. Pharmacokinetic and pharmacodynamic models of the antistaphylococcal effects of meropenem and cloxacillin in vitro and in experimental infection. Antimicrob. Agents Chemother. 41, 2083–2088 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Arnoldini, M. et al. Bistable expression of virulence genes in Salmonella leads to the formation of an antibiotic-tolerant subpopulation. PLoS Biol. 12, e1001928 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Nickel, J. C., Ruseska, I., Wright, J. B. & Costerton, J. W. Tobramycin resistance of Pseudomonas aeruginosa cells growing as a biofilm on urinary catheter material. Antimicrob. Agents Chemother. 27, 619–624 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Hayes, C. S. & Low, D. A. Signals of growth regulation in bacteria. Curr. Opin. Microbiol. 12, 667–673 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Bhuyan, B. K., Fraser, T. J. & Day, K. J. Cell proliferation kinetics and drug sensitivity of exponential and stationary populations of cultured L1210 cells. Cancer Res. 37, 1057–1063 (1977).

    CAS  PubMed  Google Scholar 

  86. Sharma, S. V. et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Jayaraman, R. Bacterial persistence: some new insights into an old phenomenon. J. Biosci. 33, 795–805 (2008).

    Article  CAS  PubMed  Google Scholar 

  88. Cohen, N. R., Lobritz, M. A. & Collins, J. J. Microbial persistence and the road to drug resistance. Cell Host Microbe 13, 632–642 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Potrykus, K. & Cashel, M. (p)ppGpp: still magical? Annu. Rev. Microbiol. 62, 35–51 (2008).

    Article  CAS  PubMed  Google Scholar 

  90. Kaspy, I. et al. HipA-mediated antibiotic persistence via phosphorylation of the glutamyl-tRNA-synthetase. Nat. Commun. 4, 3001 (2013).

    Article  PubMed  CAS  Google Scholar 

  91. Germain, E., Castro-Roa, D., Zenkin, N. & Gerdes, K. Molecular mechanism of bacterial persistence by HipA. Mol. Cell 52, 248–254 (2013).

    Article  CAS  PubMed  Google Scholar 

  92. Hahn, J., Tanner, A. W., Carabetta, V. J., Cristea, I. M. & Dubnau, D. ComGA–RelA interaction and persistence in the Bacillus subtilis K-state. Mol. Microbiol. 97, 454–471 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Gerdes, K. & Maisonneuve, E. Bacterial persistence and toxin–antitoxin loci. Annu. Rev. Microbiol. 66, 103–123 (2012).

    Article  CAS  PubMed  Google Scholar 

  94. Girgis, H. S., Harris, K. & Tavazoie, S. Large mutational target size for rapid emergence of bacterial persistence. Proc. Natl Acad. Sci. USA 109, 12740–12745 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Hansen, S., Lewis, K. & Vulic, M. Role of global regulators and nucleotide metabolism in antibiotic tolerance in Escherichia coli. Antimicrob. Agents Chemother. 52, 2718–2726 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Spoering, A. L., Vulic, M. & Lewis, K. GlpD and PlsB participate in persister cell formation in Escherichia coli. J. Bacteriol. 188, 5136–5144 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Vazquez-Laslop, N., Lee, H. & Neyfakh, A. A. Increased persistence in Escherichia coli caused by controlled expression of toxins or other unrelated proteins. J. Bacteriol. 188, 3494–3497 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Shan, Y., Lazinski, D., Rowe, S. E., Camili, A. & Lewis, K. Genetic basis of persister tolerance to aminoglycosides in Escherichia coli. mBio 6, e00078-15 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Balaban, N. Q. Persistence: mechanisms for triggering and enhancing phenotypic variability. Curr. Opin. Genet. Dev. 21, 768–775 (2011).

    Article  CAS  PubMed  Google Scholar 

  100. Casadesus, J. & Low, D. A. Programmed heterogeneity: epigenetic mechanisms in bacteria. J. Biol. Chem. 288, 13929–13935 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Huh, D. & Paulsson, J. Non-genetic heterogeneity from stochastic partitioning at cell division. Nat. Genet. 43, 95–100 (2011).

    Article  CAS  PubMed  Google Scholar 

  102. Tsimring, L. S. Noise in biology. Rep. Progress Phys. 77, 026601 (2014).

    Article  CAS  Google Scholar 

  103. Gelens, L., Hill, L., Vandervelde, A., Danckaert, J. & Loris, R. A general model for toxin–antitoxin module dynamics can explain persister cell formation in E. coli. PLoS Comput. Biol. 9, e1003190 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Maisonneuve, E., Shakespeare, L. J., Jorgensen, M. G. & Gerdes, K. Bacterial persistence by RNA endonucleases. Proc. Natl Acad. Sci. USA 108, 13206–13211 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Helaine, S. et al. Internalization of Salmonella by macrophages induces formation of nonreplicating persisters. Science 343, 204–208 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Aldridge, B. B. et al. Asymmetry and aging of mycobacterial cells lead to variable growth and antibiotic susceptibility. Science 335, 100–104 (2012).

    Article  CAS  PubMed  Google Scholar 

  107. Kieser, K. J. & Rubin, E. J. How sisters grow apart: mycobacterial growth and division. Nat. Rev. Microbiol. 12, 550–562 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Vaubourgeix, J. et al. Stressed mycobacteria use the chaperone ClpB to sequester irreversibly oxidized proteins asymmetrically within and between cells. Cell Host Microbe 17, 178–190 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Wu, Y. X., Vulic, M., Keren, I. & Lewis, K. Role of oxidative stress in persister tolerance. Antimicrob. Agents Chemother. 56, 4922–4926 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Song, Y., Rubio, A., Jayaswal, R. K., Silverman, J. A. & Wilkinson, B. J. Additional routes to Staphylococcus aureus daptomycin resistance as revealed by comparative genome sequencing, transcriptional profiling, and phenotypic studies. PLoS ONE 8, e58469 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Brehm-Stecher, B. F. & Johnson, E. A. Single-cell microbiology: tools, technologies, and applications. Microbiol. Mol. Biol. Rev. 68, 538–559 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Ping, W. et al. Robust growth of Escherichia coli. Curr. Biol. 20, 1099–1103 (2010).

    Article  CAS  Google Scholar 

  113. Iino, R., Matsumoto, Y., Nishino, K., Yamaguchi, A. & Noji, H. Design of a large-scale femtoliter droplet array for single-cell analysis of drug-tolerant and drug-resistant bacteria. Front. Microbiol. 4, 300 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Shah, D. et al. Persisters: a distinct physiological state of E. coli. BMC Microbiol. 6, 53 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Orman, M. A. & Brynildsen, M. P. Establishment of a method to rapidly assay bacterial persister metabolism. Antimicrob. Agents Chemother. 57, 4398–4409 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Jarzembowski, T., Wisniewska, K., Jozwik, A. & Witkowski, J. Heterogeneity of methicillin-resistant Staphylococcus aureus strains (MRSA) characterized by flow cytometry. Curr. Microbiol. 59, 78–80 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank N. Shoresh for illuminating discussions regarding this manuscript, and the members of the Balaban laboratory, I. Kaspy and G. Glaser for comments and suggestions. This work is supported by the Minerva Center for Stochastic Decision Making in Microorganisms, a European Research Council (ERC) Starting Grant (260871) and the Israel Science Foundation (492/15).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nathalie Q. Balaban.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brauner, A., Fridman, O., Gefen, O. et al. Distinguishing between resistance, tolerance and persistence to antibiotic treatment. Nat Rev Microbiol 14, 320–330 (2016). https://doi.org/10.1038/nrmicro.2016.34

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro.2016.34

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing