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.

  • Review Article
  • Published:

From the soil to the clinic: the impact of microbial secondary metabolites on antibiotic tolerance and resistance

Abstract

Secondary metabolites profoundly affect microbial physiology, metabolism and stress responses. Increasing evidence suggests that these molecules can modulate microbial susceptibility to commonly used antibiotics; however, secondary metabolites are typically excluded from standard antimicrobial susceptibility assays. This may in part account for why infections by diverse opportunistic bacteria that produce secondary metabolites often exhibit discrepancies between clinical antimicrobial susceptibility testing results and clinical treatment outcomes. In this Review, we explore which types of secondary metabolite alter antimicrobial susceptibility, as well as how and why this phenomenon occurs. We discuss examples of molecules that opportunistic and enteric pathogens either generate themselves or are exposed to from their neighbours, and the nuanced impacts these molecules can have on tolerance and resistance to certain antibiotics.

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

Access options

Buy this article

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

Fig. 1: Secondary metabolite-mediated regulation of multidrug resistance efflux pumps.
Fig. 2: Secondary metabolite interactions with oxidative stress.
Fig. 3: Secondary metabolites as interspecies modulators of antibiotic resilience.

Similar content being viewed by others

References

  1. Maplestone, R. A., Stone, M. J. & Williams, D. H. The evolutionary role of secondary metabolites — a review. Gene 115, 151–157 (1992).

    Article  CAS  PubMed  Google Scholar 

  2. Demain, A. L. & Fang, A. The natural functions of secondary metabolites. Adv. Biochem. Eng. Biotechnol. 69, 1–39 (2000).

    CAS  PubMed  Google Scholar 

  3. Keller, N. P., Turner, G. & Bennett, J. W. Fungal secondary metabolism–from biochemistry to genomics. Nat. Rev. Microbiol. 3, 937–947 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Tyc, O., Song, C., Dickschat, J. S., Vos, M. & Garbeva, P. The ecological role of volatile and soluble secondary metabolites produced by soil bacteria. Trends Microbiol. 25, 280–292 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Wang, S. & Lu, Z. in Biocommunication of Archaea (ed. Witzany, G.) 235–239 (Springer, 2017).

  6. Price-Whelan, A., Dietrich, L. E. P. & Newman, D. K. Rethinking “secondary” metabolism: physiological roles for phenazine antibiotics. Nat. Chem. Biol. 2, 71–78 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Davies, J. Specialized microbial metabolites: functions and origins. J. Antibiot. 66, 361–364 (2013).

    Article  CAS  Google Scholar 

  8. Haslam, E. Secondary metabolism — fact and fiction. Nat. Prod. Rep. 3, 217 (1986).

    Article  CAS  Google Scholar 

  9. Dietrich, L. E., Price-Whelan, A., Petersen, A., Whiteley, M. & Newman, D. K. The phenazine pyocyanin is a terminal signalling factor in the quorum sensing network of Pseudomonas aeruginosa. Mol. Microbiol. 61, 1308–1321 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Glasser, N. R., Kern, S. E. & Newman, D. K. Phenazine redox cycling enhances anaerobic survival in Pseudomonas aeruginosa by facilitating generation of ATP and a proton-motive force. Mol. Microbiol. 92, 399–412 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, Y. et al. Phenazine-1-carboxylic acid promotes bacterial biofilm development via ferrous iron acquisition. J. Bacteriol. 193, 3606–3617 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. McRose, D. L. & Newman, D. K. Redox-active antibiotics enhance phosphorus bioavailability. Science 371, 1033–1037 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ling, L. L. et al. A new antibiotic kills pathogens without detectable resistance. Nature 517, 455–459 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. Brauner, A., Fridman, O., Gefen, O. & Balaban, N. Q. Distinguishing between resistance, tolerance and persistence to antibiotic treatment. Nat. Rev. Microbiol. 14, 320–330 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Balaban, N. Q. et al. Definitions and guidelines for research on antibiotic persistence. Nat. Rev. Microbiol. 17, 441–448 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Piddock, L. J. V. Clinically relevant chromosomally encoded multidrug resistance efflux pumps in bacteria. Clin. Microbiol. Rev. 19, 382–402 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, X.-Z., Plésiat, P. & Nikaido, H. The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteria. Clin. Microbiol. Rev. 28, 337–418 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Piddock, L. J. V. Multidrug-resistance efflux pumps – not just for resistance. Nat. Rev. Microbiol. 4, 629–636 (2006).

    Article  CAS  PubMed  Google Scholar 

  20. Martinez, J. L. et al. Functional role of bacterial multidrug efflux pumps in microbial natural ecosystems. FEMS Microbiol. Rev. 33, 430–449 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Mousa, J. J. & Bruner, S. D. Structural and mechanistic diversity of multidrug transporters. Nat. Prod. Rep. 33, 1255–1267 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Du, D. et al. Multidrug efflux pumps: structure, function and regulation. Nat. Rev. Microbiol. 16, 523–539 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Anes, J., McCusker, M. P., Fanning, S. & Martins, M. The ins and outs of RND efflux pumps in Escherichia coli. Front. Microbiol. 6, 587 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Li, X.-Z. & Nikaido, H. in Efflux-Mediated Antimicrobial Resistance in Bacteria (eds Li, X.-Z., Elkins, C. A. & Zgurskaya, H. I.) 219–259 (Springer, 2016).

  25. Ruiz, C. & Levy, S. B. Regulation of acrAB expression by cellular metabolites in Escherichia coli. J. Antimicrob. Chemother. 69, 390–399 (2014). This work identifies endogenous cellular metabolites that induce expression of a major multidrug efflux pump in E. coli.

    Article  CAS  PubMed  Google Scholar 

  26. Chubiz, L. M. & Rao, C. V. Aromatic acid metabolites of Escherichia coli K-12 can induce the marRAB operon. J. Bacteriol. 192, 4786–4789 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hirakawa, H., Inazumi, Y., Masaki, T., Hirata, T. & Yamaguchi, A. Indole induces the expression of multidrug exporter genes in Escherichia coli. Mol. Microbiol. 55, 1113–1126 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Nishino, K., Honda, T. & Yamaguchi, A. Genome-wide analyses of Escherichia coli gene expression responsive to the BaeSR two-component regulatory system. J. Bacteriol. 187, 1763–1772 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nikaido, E. et al. Effects of indole on drug resistance and virulence of Salmonella enterica serovar Typhimurium revealed by genome-wide analyses. Gut Pathog. 4, 5 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nishino, K., Nikaido, E. & Yamaguchi, A. Regulation of multidrug efflux systems involved in multidrug and metal resistance of Salmonella enterica serovar Typhimurium. J. Bacteriol. 189, 9066–9075 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee, H. H., Molla, M. N., Cantor, C. R. & Collins, J. J. Bacterial charity work leads to population-wide resistance. Nature 467, 82–85 (2010). This work demonstrates that the production of indole by highly antibiotic-resistant mutants of E. coli increases the antibiotic tolerance and resistance of less-resistant strains, thus establishing a precedent for the role of a secondary metabolite in mediating the overall antibiotic susceptibility of a bacterial population.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Paździor, E., Pękala-Safińska, A. & Wasyl, D. Phenotypic diversity and potential virulence factors of the Shewanella putrefaciens group isolated from freshwater fish. J. Vet. Res. 63, 321–332 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Shyu, J. B. H., Lies, D. P. & Newman, D. K. Protective role of tolC in efflux of the electron shuttle anthraquinone-2,6-disulfonate. J. Bacteriol. 184, 1806–1810 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sakhtah, H. et al. The Pseudomonas aeruginosa efflux pump MexGHI–OpmD transports a natural phenazine that controls gene expression and biofilm development. Proc. Natl Acad. Sci. USA 113, E3538–E3547 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Meirelles, L. A. & Newman, D. K. Both toxic and beneficial effects of pyocyanin contribute to the lifecycle of Pseudomonas aeruginosa. Mol. Microbiol. 110, 995–1010 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Meirelles, L. A., Perry, E. K., Bergkessel, M. & Newman, D. K. Bacterial defenses against a natural antibiotic promote collateral resilience to clinical antibiotics. PLoS Biol. 19, e3001093 (2021). This work shows that a toxic secondary metabolite can increase tolerance to fluoroquinolones in strains of P. aeruginosa and other opportunistic pathogens, and can also promote the establishment of spontaneous antibiotic-resistant mutants in populations of these bacteria.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lau, G. W., Hassett, D. J., Ran, H. & Kong, F. The role of pyocyanin in Pseudomonas aeruginosa infection. Trends Mol. Med. 10, 599–606 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Mavrodi, D. V., Blankenfeldt, W. & Thomashow, L. S. Phenazine compounds in fluorescent Pseudomonas spp. biosynthesis and regulation. Annu. Rev. Phytopathol. 44, 417–445 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Llanes, C. et al. Role of the MexEF-OprN efflux system in low-level resistance of Pseudomonas aeruginosa to ciprofloxacin. Antimicrob. Agents Chemother. 55, 5676–5684 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Richardot, C. et al. Amino acid substitutions account for most MexS alterations in clinical nfxC mutants of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 60, 2302–2310 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lomovskaya, O. & Bostian, K. A. Practical applications and feasibility of efflux pump inhibitors in the clinic — a vision for applied use. Biochem. Pharmacol. 71, 910–918 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Jamshidi, S., Sutton, J. M. & Rahman, K. M. Computational study reveals the molecular mechanism of the interaction between the efflux inhibitor PAβN and the AdeB transporter from Acinetobacter baumannii. ACS Omega 2, 3002–3016 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wolloscheck, D., Krishnamoorthy, G., Nguyen, J. & Zgurskaya, H. I. Kinetic control of quorum sensing in Pseudomonas aeruginosa by multidrug efflux pumps. ACS Infect. Dis. 4, 185–195 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Schiessl, K. T. et al. Phenazine production promotes antibiotic tolerance and metabolic heterogeneity in Pseudomonas aeruginosa biofilms. Nat. Commun. 10, 762 (2019). This study reveals that phenazine production alters both the metabolic profile of biofilms and their tolerance to different classes of clinical antibiotics, suggesting that beyond induction of specific cellular defences, secondary metabolites can also affect antibiotic susceptibility via indirect mechanisms.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhu, K., Chen, S., Sysoeva, T. A. & You, L. Universal antibiotic tolerance arising from antibiotic-triggered accumulation of pyocyanin in Pseudomonas aeruginosa. PLoS Biol. 17, e3000573 (2019). In this work, the authors report that sublethal antibiotic treatment can trigger PYO production in P. aeruginosa, and that PYO enables multiple bacterial species to grow to higher cell densities in the presence of diverse clinical antibiotics.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lister, P. D., Wolter, D. J. & Hanson, N. D. Antibacterial-resistant Pseudomonas aeruginosa: clinical impact and complex regulation of chromosomally encoded resistance mechanisms. Clin. Microbiol. Rev. 22, 582–610 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yonehara, R., Yamashita, E. & Nakagawa, A. Crystal structures of OprN and OprJ, outer membrane factors of multidrug tripartite efflux pumps of Pseudomonas aeruginosa. Proteins 84, 759–769 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Glavier, M. et al. Antibiotic export by MexB multidrug efflux transporter is allosterically controlled by a MexA–OprM chaperone-like complex. Nat. Commun. 11, 4948 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Clarke-Pearson, M. F. & Brady, S. F. Paerucumarin, a new metabolite produced by the pvc gene cluster from Pseudomonas aeruginosa. J. Bacteriol. 190, 6927–6930 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Iftikhar, A. et al. Mutation in pvcABCD operon of Pseudomonas aeruginosa modulates MexEF–OprN efflux system and hence resistance to chloramphenicol and ciprofloxacin. Microb. Pathog. 149, 104491 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Sokol, P. A., Lewis, C. J. & Dennis, J. J. Isolation of a novel siderophore from Pseudomonas cepacia. J. Med. Microbiol. 36, 184–189 (1992).

    Article  CAS  PubMed  Google Scholar 

  52. Darling, P., Chan, M., Cox, A. D. & Sokol, P. A. Siderophore production by cystic fibrosis isolates of Burkholderia cepacia. Infect. Immun. 66, 874–877 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Visca, P., Ciervo, A., Sanfilippo, V. & Orsi, N. Iron-regulated salicylate synthesis by Pseudomonas spp. J. Gen. Microbiol. 139, 1995–2001 (1993).

    Article  CAS  PubMed  Google Scholar 

  54. Bakker, P. A. H. M., Ran, L. & Mercado-Blanco, J. Rhizobacterial salicylate production provokes headaches! Plant. Soil. 382, 1–16 (2014).

    Article  CAS  Google Scholar 

  55. Nair, B. M., Cheung, K.-J., Griffith, A. & Burns, J. L. Salicylate induces an antibiotic efflux pump in Burkholderia cepacia complex genomovar III (B. cenocepacia). J. Clin. Invest. 113, 464–473 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Cohen, S. P., Levy, S. B., Foulds, J. & Rosner, J. L. Salicylate induction of antibiotic resistance in Escherichia coli: activation of the mar operon and a mar-independent pathway. J. Bacteriol. 175, 7856–7862 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Brochado, A. R. et al. Species-specific activity of antibacterial drug combinations. Nature 559, 259–263 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Burkhead, K. D., Schisler, D. A. & Slininger, P. J. Pyrrolnitrin production by biological control agent Pseudomonas cepacia B37w in culture and in colonized wounds of potatoes. Appl. Environ. Microbiol. 60, 2031–2039 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Jeong, Y. et al. Toxoflavin produced by Burkholderia glumae causing rice grain rot is responsible for inducing bacterial wilt in many field crops. Plant. Dis. 87, 890–895 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Depoorter, E. et al. Burkholderia: an update on taxonomy and biotechnological potential as antibiotic producers. Appl. Microbiol. Biotechnol. 100, 5215–5229 (2016). This review catalogues the toxic secondary metabolites known to be produced by Burkholderia species and describes what is known about their regulation, thus serving as a useful resource for identifying endogenous compounds that might affect antibiotic susceptibility in this family of opportunistic pathogens.

    Article  CAS  PubMed  Google Scholar 

  61. Depoorter, E., De Canck, E., Coenye, T. & Vandamme, P. Burkholderia bacteria produce multiple potentially novel molecules that inhibit carbapenem-resistant Gram-negative bacterial pathogens. Antibiotics (Basel) 10, 147 (2021).

    Article  CAS  Google Scholar 

  62. Lipuma, J. J. The changing microbial epidemiology in cystic fibrosis. Clin. Microbiol. Rev. 23, 299–323 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Jones, C. et al. Kill and cure: genomic phylogeny and bioactivity of Burkholderia gladioli bacteria capable of pathogenic and beneficial lifestyles. Microb. Genom. 7, mgen.0.000515 (2021).

    Google Scholar 

  64. Stern, K. G. Oxidation–reduction potentials of toxoflavin. Biochem. J. 29, 500–508 (1935).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Latuasan, H. E. & Berends, W. On the origin of the toxicity of toxoflavin. Biochim. Biophys. Acta 52, 502–508 (1961).

    Article  CAS  PubMed  Google Scholar 

  66. Gencheva, R., Cheng, Q. & Arnér, E. S. J. Efficient selenocysteine-dependent reduction of toxoflavin by mammalian thioredoxin reductase. Biochim. Biophys. Acta Gen. Subj. 1862, 2511–2517 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Li, X., Li, Y., Wang, R., Wang, Q. & Lu, L. Toxoflavin produced by Burkholderia gladioli from Lycoris aurea is a new broad-spectrum fungicide. Appl. Environ. Microbiol. 85, e00106-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Kim, J. et al. Quorum sensing and the LysR-type transcriptional activator ToxR regulate toxoflavin biosynthesis and transport in Burkholderia glumae. Mol. Microbiol. 54, 921–934 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Tahlan, K. et al. Initiation of actinorhodin export in Streptomyces coelicolor. Mol. Microbiol. 63, 951–961 (2007).

    Article  CAS  PubMed  Google Scholar 

  70. Willems, A. R. et al. Crystal structures of the Streptomyces coelicolor TetR-like protein ActR alone and in complex with actinorhodin or the actinorhodin biosynthetic precursor (S)-DNPA. J. Mol. Biol. 376, 1377–1387 (2008).

    Article  CAS  PubMed  Google Scholar 

  71. Kohanski, M. A., Dwyer, D. J., Hayete, B., Lawrence, C. A. & Collins, J. J. A common mechanism of cellular death induced by bactericidal antibiotics. Cell 130, 797–810 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Keren, I., Wu, Y., Inocencio, J., Mulcahy, L. R. & Lewis, K. Killing by bactericidal antibiotics does not depend on reactive oxygen species. Science 339, 1213–1216 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Liu, Y. & Imlay, J. A. Cell death from antibiotics without the involvement of reactive oxygen species. Science 339, 1210–1213 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Dwyer, D. J., Collins, J. J. & Walker, G. C. Unraveling the physiological complexities of antibiotic lethality. Annu. Rev. Pharmacol. Toxicol. 55, 313–332 (2015). This work comprehensively discusses the evidence that oxidative stress contributes to antibiotic lethality.

    Article  CAS  PubMed  Google Scholar 

  75. Zuccato, E. et al. Role of bile acids and metabolic activity of colonic bacteria in increased risk of colon cancer after cholecystectomy. Dig. Dis. Sci. 38, 514–519 (1993).

    Article  CAS  PubMed  Google Scholar 

  76. Karlin, D. A., Mastromarino, A. J., Jones, R. D., Stroehlein, J. R. & Lorentz, O. Fecal skatole and indole and breath methane and hydrogen in patients with large bowel polyps or cancer. J. Cancer Res. Clin. Oncol. 109, 135–141 (1985).

    Article  CAS  PubMed  Google Scholar 

  77. Vega, N. M., Allison, K. R., Khalil, A. S. & Collins, J. J. Signaling-mediated bacterial persister formation. Nat. Chem. Biol. 8, 431–433 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Shen, X., Lind, J. & Merenyi, G. One-electron oxidation of indoles and acid-base properties of the indolyl radicals. J. Phys. Chem. 91, 4403–4406 (1987).

    Article  Google Scholar 

  79. Garbe, T. R., Kobayashi, M. & Yukawa, H. Indole-inducible proteins in bacteria suggest membrane and oxidant toxicity. Arch. Microbiol. 173, 78–82 (2000).

    Article  CAS  PubMed  Google Scholar 

  80. Perry, E. K. & Newman, D. K. The transcription factors ActR and SoxR differentially affect the phenazine tolerance of Agrobacterium tumefaciens. Mol. Microbiol. 112, 199–218 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Voggu, L. et al. Microevolution of cytochrome bd oxidase in staphylococci and its implication in resistance to respiratory toxins released by Pseudomonas. J. Bacteriol. 188, 8079–8086 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Hassett, D. J., Charniga, L., Bean, K., Ohman, D. E. & Cohen, M. S. Response of Pseudomonas aeruginosa to pyocyanin: mechanisms of resistance, antioxidant defenses, and demonstration of a manganese-cofactored superoxide dismutase. Infect. Immun. 60, 328–336 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Priuska, E. M. & Schacht, J. Formation of free radicals by gentamicin and iron and evidence for an iron/gentamicin complex. Biochem. Pharmacol. 50, 1749–1752 (1995).

    Article  CAS  PubMed  Google Scholar 

  84. Prayle, A., Watson, A., Fortnum, H. & Smyth, A. Side effects of aminoglycosides on the kidney, ear and balance in cystic fibrosis. Thorax 65, 654–658 (2010).

    Article  PubMed  Google Scholar 

  85. Mosel, M., Li, L., Drlica, K. & Zhao, X. Superoxide-mediated protection of Escherichia coli from antimicrobials. Antimicrob. Agents Chemother. 57, 5755–5759 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Alexander, H. K. & MacLean, R. C. Stochastic bacterial population dynamics restrict the establishment of antibiotic resistance from single cells. Proc. Natl Acad. Sci. USA 117, 19455–19464 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Dwyer, D. J. et al. Antibiotics induce redox-related physiological alterations as part of their lethality. Proc. Natl Acad. Sci. USA 111, E2100–E2109 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Belenky, P. et al. Bactericidal antibiotics induce toxic metabolic perturbations that lead to cellular damage. Cell Rep. 13, 968–980 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Saini, V. et al. Ergothioneine maintains redox and bioenergetic homeostasis essential for drug susceptibility and virulence of Mycobacterium tuberculosis. Cell Rep. 14, 572–585 (2016). This work indicates that a metabolite involved in redox homeostasis has a key role in mediating antibiotic resistance and tolerance in M. tuberculosis, suggesting that such metabolites might also contribute to these phenotypes in other bacteria.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Hall, J. W., Yang, J., Guo, H. & Ji, Y. The Staphylococcus aureus AirSR two-component system mediates reactive oxygen species resistance via transcriptional regulation of staphyloxanthin production. Infect. Immun. 85, e00838-16 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Clauditz, A., Resch, A., Wieland, K.-P., Peschel, A. & Götz, F. Staphyloxanthin plays a role in the fitness of Staphylococcus aureus and its ability to cope with oxidative stress. Infect. Immun. 74, 4950–4953 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Liu, G. Y. et al. Sword and shield: linked group B streptococcal beta-hemolysin/cytolysin and carotenoid pigment function to subvert host phagocyte defense. Proc. Natl Acad. Sci. USA 101, 14491–14496 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Wu, C. et al. Genomic island TnSmu2 of Streptococcus mutans harbors a nonribosomal peptide synthetase-polyketide synthase gene cluster responsible for the biosynthesis of pigments involved in oxygen and H2O2 tolerance. Appl. Environ. Microbiol. 76, 5815–5826 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Edge, R. & Truscott, T. G. Singlet oxygen and free radical reactions of retinoids and carotenoids–a review. Antioxidants (Basel) 7, 5 (2018).

    Article  Google Scholar 

  95. Young, A. J. & Lowe, G. L. Carotenoids–antioxidant properties. Antioxidants (Basel) 7, 28 (2018).

    Article  Google Scholar 

  96. Hong, Y., Zeng, J., Wang, X., Drlica, K. & Zhao, X. Post-stress bacterial cell death mediated by reactive oxygen species. Proc. Natl Acad. Sci. USA 116, 10064–10071 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Shatalin, K., Shatalina, E., Mironov, A. & Nudler, E. H2S: a universal defense against antibiotics in bacteria. Science 334, 986–990 (2011).

    Article  CAS  PubMed  Google Scholar 

  98. Tkachenko, A. G. & Fedotova, M. V. Dependence of protective functions of Escherichia coli polyamines on strength of stress caused by superoxide radicals. Biochemistry. 72, 109–116 (2007).

    CAS  PubMed  Google Scholar 

  99. El-Halfawy, O. M. & Valvano, M. A. Putrescine reduces antibiotic-induced oxidative stress as a mechanism of modulation of antibiotic resistance in Burkholderia cenocepacia. Antimicrob. Agents Chemother. 58, 4162–4171 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  100. El-Halfawy, O. M. & Valvano, M. A. Chemical communication of antibiotic resistance by a highly resistant subpopulation of bacterial cells. PLoS ONE 8, e68874 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Tkachenko, A. G., Akhova, A. V., Shumkov, M. S. & Nesterova, L. Y. Polyamines reduce oxidative stress in Escherichia coli cells exposed to bactericidal antibiotics. Res. Microbiol. 163, 83–91 (2012).

    Article  CAS  PubMed  Google Scholar 

  102. Kreamer, N. N., Costa, F. & Newman, D. K. The ferrous iron-responsive BqsRS two-component system activates genes that promote cationic stress tolerance. mBio 6, e02549 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Häussler, S. & Becker, T. The pseudomonas quinolone signal (PQS) balances life and death in Pseudomonas aeruginosa populations. PLoS Pathog. 4, e1000166 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  104. Bredenbruch, F., Geffers, R., Nimtz, M., Buer, J. & Häussler, S. The Pseudomonas aeruginosa quinolone signal (PQS) has an iron-chelating activity. Environ. Microbiol. 8, 1318–1329 (2006).

    Article  CAS  PubMed  Google Scholar 

  105. 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 

  106. Han, M.-L. et al. Comparative metabolomics and transcriptomics reveal multiple pathways associated with polymyxin killing in Pseudomonas aeruginosa. mSystems 4, e00149-18 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  107. Sampson, T. R. et al. Rapid killing of Acinetobacter baumannii by polymyxins is mediated by a hydroxyl radical death pathway. Antimicrob. Agents Chemother. 56, 5642–5649 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Trimble, M. J., Mlynárčik, P., Kolář, M. & Hancock, R. E. W. Polymyxin: alternative mechanisms of action and resistance. Cold Spring Harb. Perspect. Med. 6, a025288 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  109. Bottery, M. J., Pitchford, J. W. & Friman, V.-P. Ecology and evolution of antimicrobial resistance in bacterial communities. ISME J. 15, 939–948 (2021).

    Article  PubMed  Google Scholar 

  110. Welp, A. L. & Bomberger, J. M. Bacterial community interactions during chronic respiratory disease. Front. Cell Infect. Microbiol. 10, 213 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Vega, N. M., Allison, K. R., Samuels, A. N., Klempner, M. S. & Collins, J. J. Salmonella typhimurium intercepts Escherichia coli signaling to enhance antibiotic tolerance. Proc. Natl Acad. Sci. USA 110, 14420–14425 (2013). This work reveals how a secondary metabolite that induces antibiotic tolerance can work across bacterial species.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Kim, J., Shin, B., Park, C. & Park, W. Indole-induced activities of β-lactamase and efflux pump confer ampicillin resistance in Pseudomonas putida KT2440. Front. Microbiol. 8, 433 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Bhargava, N., Sharma, P. & Capalash, N. Pyocyanin stimulates quorum sensing-mediated tolerance to oxidative stress and increases persister cell populations in Acinetobacter baumannii. Infect. Immun. 82, 3417–3425 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Heindorf, M., Kadari, M., Heider, C., Skiebe, E. & Wilharm, G. Impact of Acinetobacter baumannii superoxide dismutase on motility, virulence, oxidative stress resistance and susceptibility to antibiotics. PLoS ONE 9, e101033 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  115. Bandara, H. M. H. N. et al. Fluconazole resistance in Candida albicans is induced by Pseudomonas aeruginosa quorum sensing. Sci. Rep. 10, 7769 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Chmiel, J. F. et al. Antibiotic management of lung infections in cystic fibrosis. I. The microbiome, methicillin-resistant Staphylococcus aureus, Gram-negative bacteria, and multiple infections. Ann. Am. Thorac. Soc. 11, 1120–1129 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Schwab, U. et al. Localization of Burkholderia cepacia complex bacteria in cystic fibrosis lungs and interactions with Pseudomonas aeruginosa in hypoxic mucus. Infect. Immun. 82, 4729–4745 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Wood, K. B. & Cluzel, P. Trade-offs between drug toxicity and benefit in the multi-antibiotic resistance system underlie optimal growth of E. coli. BMC Syst. Biol. 6, 48 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Yung, D. B. Y., Sircombe, K. J. & Pletzer, D. Friends or enemies? The complicated relationship between Pseudomonas aeruginosa and Staphylococcus aureus. Mol. Microbiol. 116, 1–15 (2021).

    Article  CAS  PubMed  Google Scholar 

  120. Camus, L., Briaud, P., Vandenesch, F. & Moreau, K. How bacterial adaptation to cystic fibrosis environment shapes interactions between Pseudomonas aeruginosa and Staphylococcus aureus. Front. Microbiol. 12, 617784 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  121. Briaud, P. et al. Impact of coexistence phenotype between Staphylococcus aureus and Pseudomonas aeruginosa isolates on clinical outcomes among cystic fibrosis patients. Front. Cell Infect. Microbiol. 10, 266 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Radlinski, L. et al. Pseudomonas aeruginosa exoproducts determine antibiotic efficacy against Staphylococcus aureus. PLoS Biol. 15, e2003981 (2017). In this work, the authors screen clinical isolates of P. aeruginosa for effects on the antibiotic susceptibility of S. aureus and find that the interactions are strain specific and complex, highlighting the challenges of understanding how secondary metabolites affect polymicrobial infections.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Noto, M. J., Burns, W. J., Beavers, W. N. & Skaar, E. P. Mechanisms of pyocyanin toxicity and genetic determinants of resistance in Staphylococcus aureus. J. Bacteriol. 199, e00221-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  124. Orazi, G., Ruoff, K. L. & O’Toole, G. A. Pseudomonas aeruginosa increases the sensitivity of biofilm-grown Staphylococcus aureus to membrane-targeting antiseptics and antibiotics. mBio 10, e01501-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  125. Orazi, G. & O’Toole, G. A. Pseudomonas aeruginosa alters Staphylococcus aureus sensitivity to vancomycin in a biofilm model of cystic fibrosis infection. mBio 8, e00873-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  126. Biswas, L., Biswas, R., Schlag, M., Bertram, R. & Götz, F. Small-colony variant selection as a survival strategy for Staphylococcus aureus in the presence of Pseudomonas aeruginosa. Appl. Environ. Microbiol. 75, 6910–6912 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. McNamara, P. J. & Proctor, R. A. Staphylococcus aureus small colony variants, electron transport and persistent infections. Int. J. Antimicrob. Agents 14, 117–122 (2000).

    Article  CAS  PubMed  Google Scholar 

  128. Wilson, R. et al. Measurement of Pseudomonas aeruginosa phenazine pigments in sputum and assessment of their contribution to sputum sol toxicity for respiratory epithelium. Infect. Immun. 56, 2515–2517 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Cruickshank, C. N. & Lowbury, E. J. The effect of pyocyanin on human skin cells and leucocytes. Br. J. Exp. Pathol. 34, 583–587 (1953).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Levin-Reisman, I., Brauner, A., Ronin, I. & Balaban, N. Q. Epistasis between antibiotic tolerance, persistence, and resistance mutations. Proc. Natl Acad. Sci. USA 116, 14734–14739 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Santi, I., Manfredi, P., Maffei, E., Egli, A. & Jenal, U. Evolution of antibiotic tolerance shapes resistance development in chronic Pseudomonas aeruginosa infections. mBio 12, e03482-20 (2021). In this work, the authors analyse the microevolution of P. aeruginosa within patients with chronic infection and find that antibiotic tolerance promotes the evolution of antibiotic resistance, validating earlier in vitro studies that showed a link between tolerance and resistance.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Windels, E. M. et al. Bacterial persistence promotes the evolution of antibiotic resistance by increasing survival and mutation rates. ISME J. 13, 1239–1251 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Levin-Reisman, I. et al. Antibiotic tolerance facilitates the evolution of resistance. Science 355, 826–830 (2017).

    Article  CAS  PubMed  Google Scholar 

  134. Liu, J., Gefen, O., Ronin, I., Bar-Meir, M. & Balaban, N. Q. Effect of tolerance on the evolution of antibiotic resistance under drug combinations. Science 367, 200–204 (2020).

    Article  CAS  PubMed  Google Scholar 

  135. Martina, P. et al. Hypermutation in Burkholderia cepacia complex is mediated by DNA mismatch repair inactivation and is highly prevalent in cystic fibrosis chronic respiratory infection. Int. J. Med. Microbiol. 304, 1182–1191 (2014).

    Article  CAS  PubMed  Google Scholar 

  136. Ryan, R. P. et al. The versatility and adaptation of bacteria from the genus Stenotrophomonas. Nat. Rev. Microbiol. 7, 514–525 (2009).

    Article  CAS  PubMed  Google Scholar 

  137. Walterson, A. M. & Stavrinides, J. Pantoea: insights into a highly versatile and diverse genus within the Enterobacteriaceae. FEMS Microbiol. Rev. 39, 968–984 (2015).

    Article  CAS  PubMed  Google Scholar 

  138. Smith, D. D. N., Kirzinger, M. W. B. & Stavrinides, J. Draft genome sequence of the antibiotic-producing cystic fibrosis isolate Pantoea agglomerans Tx10. Genome Announc. 1, e00904-13 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  139. König, S., Vogel, H.-J., Harms, H. & Worrich, A. Physical, chemical and biological effects on soil bacterial dynamics in microscale models. Front. Ecol. Evol. 8, 53 (2020).

    Article  Google Scholar 

  140. Severi, E. & Thomas, G. H. Antibiotic export: transporters involved in the final step of natural product production. Microbiology 165, 805–818 (2019).

    Article  CAS  PubMed  Google Scholar 

  141. Martín, J. F., Casqueiro, J. & Liras, P. Secretion systems for secondary metabolites: how producer cells send out messages of intercellular communication. Curr. Opin. Microbiol. 8, 282–293 (2005).

    Article  PubMed  Google Scholar 

  142. Crits-Christoph, A., Bhattacharya, N., Olm, M. R., Song, Y. S. & Banfield, J. F. Transporter genes in biosynthetic gene clusters predict metabolite characteristics and siderophore activity. Genome Res. 31, 239–250 (2020).

    Article  Google Scholar 

  143. Glasser, N. R., Saunders, S. H. & Newman, D. K. The colorful world of extracellular electron shuttles. Annu. Rev. Microbiol. 71, 731–751 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Yan, J. & Bassler, B. L. Surviving as a community: antibiotic tolerance and persistence in bacterial biofilms. Cell Host Microbe 26, 15–21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Ersoy, S. C. et al. Correcting a fundamental flaw in the paradigm for antimicrobial susceptibility testing. EBioMedicine 20, 173–181 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  146. Song, Y. et al. Inhibition of staphyloxanthin virulence factor biosynthesis in Staphylococcus aureus: in vitro, in vivo, and crystallographic results. J. Med. Chem. 52, 3869–3880 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Liu, C.-I. et al. A cholesterol biosynthesis inhibitor blocks Staphylococcus aureus virulence. Science 319, 1391–1394 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Costa, K. C., Glasser, N. R., Conway, S. J. & Newman, D. K. Pyocyanin degradation by a tautomerizing demethylase inhibits Pseudomonas aeruginosa biofilms. Science 355, 170–173 (2017).

    Article  CAS  PubMed  Google Scholar 

  149. VanDrisse, C. M., Lipsh-Sokolik, R., Khersonsky, O., Fleishman, S. J. & Newman, D. K. Computationally designed pyocyanin demethylase acts synergistically with tobramycin to kill recalcitrant Pseudomonas aeruginosa biofilms. Proc. Natl Acad. Sci. USA 118, e2022012118 (2021). This works reveals that degradation of a secondary metabolite that promotes antibiotic tolerance in biofilms can increase antibiotic lethality, suggesting that targeting such secondary metabolites may be a viable approach to potentiating exisiting clinical antibiotics.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Liu, G. Y. & Nizet, V. Color me bad: microbial pigments as virulence factors. Trends Microbiol. 17, 406–413 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Shatalin, K. et al. Inhibitors of bacterial H2S biogenesis targeting antibiotic resistance and tolerance. Science 372, 1169–1175 (2021). In this work, the authors demonstrate that antibiotic efficacy is increased both in vitro and in a mouse infection model by inhibiting the production of a bacterial metabolite previously implicated in intrinsic antibiotic tolerance and resistance.

    Article  CAS  PubMed  Google Scholar 

  152. Abbott, I. J. & Peleg, A. Y. Stenotrophomonas, Achromobacter, and nonmelioid Burkholderia species: antimicrobial resistance and therapeutic strategies. Semin. Respir. Crit. Care Med. 36, 99–110 (2015).

    Article  PubMed  Google Scholar 

  153. Hazan, R. et al. Auto poisoning of the respiratory chain by a quorum-sensing-regulated molecule favors biofilm formation and antibiotic tolerance. Curr. Biol. 26, 195–206 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Shukla, P. et al. “On demand” redox buffering by H2S contributes to antibiotic resistance revealed by a bacteria-specific H2S donor. Chem. Sci. 8, 4967–4972 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Anderson, R. J. & Newman, M. S. The chemistry of the lipids of tubercle bacilli. J. Bio. Chem. 103, 405–412 (1933).

    Article  CAS  Google Scholar 

  156. Gardner, P. R. Superoxide production by the mycobacterial and pseudomonad quinoid pigments phthiocol and pyocyanine in human lung cells. Arch. Biochem. Biophys. 333, 267–274 (1996).

    Article  CAS  PubMed  Google Scholar 

  157. Giddens, S. R., Feng, Y. & Mahanty, H. K. Characterization of a novel phenazine antibiotic gene cluster in Erwinia herbicola Eh1087. Mol. Microbiol. 45, 769–783 (2002).

    Article  CAS  PubMed  Google Scholar 

  158. Giddens, S. R. & Bean, D. C. Investigations into the in vitro antimicrobial activity and mode of action of the phenazine antibiotic d-alanylgriseoluteic acid. Int. J. Antimicrob. Agents 29, 93–97 (2007).

    Article  CAS  PubMed  Google Scholar 

  159. Krishnamurthi, V. S., Buckley, P. J. & Duerre, J. A. Pigment formation from l-tryptophan by a particulate fraction from an Achromobacter species. Arch. Biochem. Biophys. 130, 636–645 (1969).

    Article  CAS  PubMed  Google Scholar 

  160. Wells, J. M., Cole, R. J. & Kirksey, J. W. Emodin, a toxic metabolite of Aspergillus wentii isolated from weevil-damaged chestnuts. Appl. Microbiol. 30, 26–28 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Lim, F. Y. et al. Genome-based cluster deletion reveals an endocrocin biosynthetic pathway in Aspergillus fumigatus. Appl. Environ. Microbiol. 78, 4117–4125 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Imlay, J. A. The molecular mechanisms and physiological consequences of oxidative stress: lessons from a model bacterium. Nat. Rev. Microbiol. 11, 443–454 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Dietrich, L. E. P., Teal, T. K., Price-Whelan, A. & Newman, D. K. Redox-active antibiotics control gene expression and community behavior in divergent bacteria. Science 321, 1203–1206 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Gu, M. & Imlay, J. A. The SoxRS response of Escherichia coli is directly activated by redox-cycling drugs rather than by superoxide. Mol. Microbiol. 79, 1136–1150 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Singh, A. K., Shin, J.-H., Lee, K.-L., Imlay, J. A. & Roe, J.-H. Comparative study of SoxR activation by redox-active compounds. Mol. Microbiol. 90, 983–996 (2013).

    Article  CAS  PubMed  Google Scholar 

  166. Tanabe, M. et al. The multidrug resistance efflux complex, EmrAB from Escherichia coli forms a dimer in vitro. Biochem. Biophys. Res. Commun. 380, 338–342 (2009).

    Article  CAS  PubMed  Google Scholar 

  167. Lu, S. & Zgurskaya, H. I. Role of ATP binding and hydrolysis in assembly of MacAB-TolC macrolide transporter. Mol. Microbiol. 86, 1132–1143 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Wei, Q. et al. Global regulation of gene expression by OxyR in an important human opportunistic pathogen. Nucleic Acids Res. 40, 4320–4333 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Ochsner, U. A., Vasil, M. L., Alsabbagh, E., Parvatiyar, K. & Hassett, D. J. Role of the Pseudomonas aeruginosa oxyR–recG operon in oxidative stress defense and DNA repair: OxyR-dependent regulation of katB–ankB, ahpB, and ahpC–ahpF. J. Bacteriol. 182, 4533–4544 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Romsang, A., Dubbs, J. M. & Mongkolsuk, S. in Stress and Environmental Regulation Of Gene Expression and Adaptation in Bacteria (ed. de Bruijn, F. J.) 1090–1102 (John Wiley & Sons, 2016).

  171. Blin, K. et al. antiSMASH 5.0: updates to the secondary metabolite genome mining pipeline. Nucleic Acids Res. 47, W81–W87 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Liu, J. et al. Metabolic co-dependence gives rise to collective oscillations within biofilms. Nature 523, 550–554 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Liu, J. et al. Coupling between distant biofilms and emergence of nutrient time-sharing. Science 356, 638–642 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Spero, M. A. & Newman, D. K. Chlorate specifically targets oxidant-starved, antibiotic-tolerant populations of Pseudomonas aeruginosa biofilms. mBio 9, e01400-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Saunders, S. H. et al. Extracellular DNA promotes efficient extracellular electron transfer by pyocyanin in Pseudomonas aeruginosa biofilms. Cell 182, 919–932.e19 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Rosche, W. A. & Foster, P. L. Determining mutation rates in bacterial populations. Methods 20, 4–17 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Zheng, Q. A new practical guide to the Luria–Delbrück protocol. Mutat. Res. 781, 7–13 (2015).

    Article  CAS  PubMed  Google Scholar 

  178. Somayaji, R. et al. Antimicrobial susceptibility testing (AST) and associated clinical outcomes in individuals with cystic fibrosis: a systematic review. J. Cyst. Fibros. 18, 236–243 (2019).

    Article  CAS  PubMed  Google Scholar 

  179. Hurley, M. N., Ariff, A. H. A., Bertenshaw, C., Bhatt, J. & Smyth, A. R. Results of antibiotic susceptibility testing do not influence clinical outcome in children with cystic fibrosis. J. Cyst. Fibros. 11, 288–292 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Jorgensen, J. H. & Ferraro, M. J. Antimicrobial susceptibility testing: a review of general principles and contemporary practices. Clin. Infect. Dis. 49, 1749–1755 (2009).

    Article  CAS  PubMed  Google Scholar 

  181. Brown, M. R. Nutrient depletion and antibiotic susceptibility. J. Antimicrob. Chemother. 3, 198–201 (1977).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Work in the corresponding author’s laboratory was supported by grants to D.K.N. from the NIH (1R01AI127850-01A1, 1R01HL152190-01) and the Doren Family Foundation. E.K.P. was supported by a National Science Foundation Graduate Research Fellowship under Grant No. DGE-1745301.

Author information

Authors and Affiliations

Authors

Contributions

E.K.P., L.A.M. and D.K.N. conceived the idea. E.K.P. and L.A.M. wrote the paper. D.K.N. edited the paper.

Corresponding author

Correspondence to Dianne K. Newman.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Microbiology thanks B. Luisi, who co-reviewed with Y. Ntsogo and E. Petsolari; and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Tolerance

The ability to survive transient antibiotic exposure.

Resistance

The ability to grow in the presence of antibiotics at a given concentration.

Antibiotic resilience

The ability of a bacterial population to be refractory to antibiotic treatment via tolerance and/or resistance.

Efflux pumps

Membrane-associated transport proteins that are responsible for the extrusion of various compounds out of the cell.

Persisters

A subpopulation of bacteria that is killed by a given antibiotic at a much slower rate than the rest of the population, in a manner that is non-heritable.

Antioxidant activity

The ability to neutralize highly reactive free radicals.

Pro-oxidant activity

The ability to induce oxidative stress.

Polymicrobial infection

An infection that is caused by more than one species of microorganism.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Perry, E.K., Meirelles, L.A. & Newman, D.K. From the soil to the clinic: the impact of microbial secondary metabolites on antibiotic tolerance and resistance. Nat Rev Microbiol 20, 129–142 (2022). https://doi.org/10.1038/s41579-021-00620-w

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41579-021-00620-w

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