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.

  • Article
  • Published:

Activated ClpP kills persisters and eradicates a chronic biofilm infection

Subjects

Abstract

Chronic infections are difficult to treat with antibiotics but are caused primarily by drug-sensitive pathogens. Dormant persister cells that are tolerant to killing by antibiotics are responsible for this apparent paradox. Persisters are phenotypic variants of normal cells and pathways leading to dormancy are redundant, making it challenging to develop anti-persister compounds. Biofilms shield persisters from the immune system, suggesting that an antibiotic for treating a chronic infection should be able to eradicate the infection on its own. We reasoned that a compound capable of corrupting a target in dormant cells will kill persisters. The acyldepsipeptide antibiotic (ADEP4) has been shown to activate the ClpP protease, resulting in death of growing cells. Here we show that ADEP4-activated ClpP becomes a fairly nonspecific protease and kills persisters by degrading over 400 proteins, forcing cells to self-digest. Null mutants of clpP arise with high probability, but combining ADEP4 with rifampicin produced complete eradication of Staphylococcus aureus biofilms in vitro and in a mouse model of a chronic infection. Our findings indicate a general principle for killing dormant cells—activation and corruption of a target, rather than conventional inhibition. Eradication of a biofilm in an animal model by activating a protease suggests a realistic path towards developing therapies to treat chronic infections.

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

Figure 1
Figure 2: Quantitative proteomic analysis of S. aureus cells treated with ADEP4 reveals extensive protein degradation.
Figure 3: ADEP4 kills persisters.
Figure 4: ADEP4 with rifampicin eradicates a variety of S. aureus strains.
Figure 5: ADEP4 kills a S. aureus biofilm and in combination with rifampicin eradicates the population.
Figure 6: ADEP4 in combination with rifampicin eradicates a deep-seated mouse biofilm infection.

Similar content being viewed by others

References

  1. Alekshun, M. N. & Levy, S. B. Molecular mechanisms of antibacterial multidrug resistance. Cell 128, 1037–1050 (2007)

    Article  CAS  PubMed  Google Scholar 

  2. Lewis, K. Persister cells. Annu. Rev. Microbiol. 64, 357–372 (2010)

    Article  CAS  PubMed  Google Scholar 

  3. Keren, I., Shah, D., Spoering, A., Kaldalu, N. & Lewis, K. Specialized persister cells and the mechanism of multidrug tolerance in Escherichia coli. J. Bacteriol. 186, 8172–8180 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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  ADS  Google Scholar 

  5. Bigger, J. W. Treatment of staphylococcal infections with penicillin. Lancet 244, 497–500 (1944)

    Article  Google Scholar 

  6. Spoering, A. L. & Lewis, K. Biofilms and planktonic cells of Pseudomonas aeruginosa have similar resistance to killing by antimicrobials. J. Bacteriol. 183, 6746–6751 (2001)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Costerton, J. W., Stewart, P. S. & Greenberg, E. P. Bacterial biofilms: A common cause of persistent infections. Science 284, 1318–1322 (1999)

    Article  CAS  PubMed  ADS  Google Scholar 

  8. Kostakioti, M., Hadjifrangiskou, M. & Hultgren, S. J. Bacterial biofilms: development, dispersal, and therapeutic strategies in the dawn of the postantibiotic era. Cold Spring Harb. Perspect. Med. 3, a010306 (2013)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. 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  ADS  Google Scholar 

  10. 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)

    CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. Correia, F. F. et al. Kinase activity of overexpressed HipA is required for growth arrest and multidrug tolerance in Escherichia coli. J. Bacteriol. 188, 8360–8367 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Harrison, J. J. et al. The chromosomal toxin gene yafQ is a determinant of multidrug tolerance for Escherichia coli growing in a biofilm. Antimicrob. Agents Chemother. 53, 2253–2258 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. Dörr, T., Vulić, 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 

  16. Unoson, C. & Wagner, E. A small SOS-induced toxin is targeted against the inner membrane in Escherichia coli. Mol. Microbiol. 70, 258–270 (2008)

    Article  CAS  PubMed  Google Scholar 

  17. Gurnev, P. A., Ortenberg, R., Dorr, T., Lewis, K. & Bezrukov, S. M. Persister-promoting bacterial toxin TisB produces anion-selective pores in planar lipid bilayers. FEBS Lett. 586, 2529–2534 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lewis, K. Platforms for antibiotic discovery. Nature Rev. Drug Discov. 12, 371–387 (2013)

    Article  CAS  Google Scholar 

  19. Brötz-Oesterhelt, H. et al. Dysregulation of bacterial proteolytic machinery by a new class of antibiotics. Nature Med. 11, 1082–1087 (2005)

    Article  PubMed  CAS  Google Scholar 

  20. Gottesman, S., Roche, E., Zhou, Y. & Sauer, R. T. The ClpXP and ClpAP proteases degrade proteins with carboxy-terminal peptide tails added by the SsrA-tagging system. Genes Dev. 12, 1338–1347 (1998)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Li, D. H. et al. Acyldepsipeptide antibiotics induce the formation of a structured axial channel in ClpP: A model for the ClpX/ClpA-bound state of ClpP. Chem. Biol. 17, 959–969 (2010)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  22. Lee, B. G. et al. Structures of ClpP in complex with acyldepsipeptide antibiotics reveal its activation mechanism. Nature Struct. Mol. Biol. 17, 471–478 (2010)

    Article  CAS  Google Scholar 

  23. Kirstein, J. et al. The antibiotic ADEP reprogrammes ClpP, switching it from a regulated to an uncontrolled protease. EMBO Mol. Med. 1, 37–49 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Michel, K. H. & Kastner, R. E. A54556 antibiotics and process for production thereof. US patent 4,492. 650 (1985)

  25. Sass, P. et al. Antibiotic acyldepsipeptides activate ClpP peptidase to degrade the cell division protein FtsZ. Proc. Natl Acad. Sci. USA 108, 17474–17479 (2011)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  26. Michalik, S. et al. Proteolysis during long-term glucose starvation in Staphylococcus aureus COL. Proteomics 9, 4468–4477 (2009)

    Article  CAS  PubMed  Google Scholar 

  27. Buske, P. J. & Levin, P. A. A flexible C-terminal linker is required for proper FtsZ assembly in vitro and cytokinetic ring formation in vivo. Mol. Microbiol. 89, 249–263 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  29. Johnson, P. J. & 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 

  30. Frees, D., Qazi, S. N., Hill, P. J. & Ingmer, H. Alternative roles of ClpX and ClpP in Staphylococcus aureus stress tolerance and virulence. Mol. Microbiol. 48, 1565–1578 (2003)

    Article  CAS  PubMed  Google Scholar 

  31. Hussain, M., Hastings, J. G. & White, P. J. A chemically defined medium for slime production by coagulase-negative staphylococci. J. Med. Microbiol. 34, 143–147 (1991)

    Article  CAS  PubMed  Google Scholar 

  32. Stryjewski, M. E. & Chambers, H. F. Skin and soft-tissue infections caused by community-acquired methicillin-resistant Staphylococcus aureus. Clin. Infect. Dis. 46 (Suppl 5). S368–S377 (2008)

    Article  CAS  PubMed  Google Scholar 

  33. Gillaspy, A. F. et al. Role of the accessory gene regulator (agr) in pathogenesis of staphylococcal osteomyelitis. Infect. Immun. 63, 3373–3380 (1995)

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Miyazaki, M. et al. Vancomycin bactericidal activity as a predictor of 30-day mortality in patients with methicillin-resistant Staphylococcus aureus bacteremia. Antimicrob. Agents Chemother. 55, 1819–1820 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lewis, K. Antibiotics: recover the lost art of drug discovery. Nature 485, 439–440 (2012)

    Article  CAS  PubMed  ADS  Google Scholar 

  36. Hung, C. et al. Escherichia coli biofilms have an organized and complex extracellular matrix structure. mBio. 4, e00645–13 (2013)

  37. Vlamakis, H., Chai, Y., Beauregard, P., Losick, R. & Kolter, R. Sticking together: building a biofilm the Bacillus subtilis way. Nature Rev. Microbiol. 11, 157–168 (2013)

    Article  CAS  Google Scholar 

  38. LaFleur, M. D., Qi, Q. & Lewis, K. Patients with long-term oral carriage harbor high-persister mutants of Candida albicans. Antimicrob. Agents Chemother. 54, 39–44 (2010)

    Article  CAS  PubMed  Google Scholar 

  39. Mulcahy, L. R., Burns, J. L., Lory, S. & Lewis, K. Emergence of Pseudomonas aeruginosa strains producing high levels of persister cells in patients with cystic fibrosis. J. Bacteriol. 192, 6191–6199 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mwangi, M. M. et al. Tracking the in vivo evolution of multidrug resistance in Staphylococcus aureus by whole-genome sequencing. Proc. Natl Acad. Sci. USA 104, 9451–9456 (2007)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  42. Howden, B. P. et al. Evolution of multidrug resistance during Staphylococcus aureus infection involves mutation of the essential two component regulator WalKR. PLoS Pathog. 7, e1002359 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Leung, E. et al. Activators of cylindrical proteases as antimicrobials: identification and development of small molecule activators of ClpP protease. Chem. Biol. 18, 1167–1178 (2011)

    Article  CAS  PubMed  Google Scholar 

  44. Wang, Y. et al. Reversed-phase chromatography with multiple fraction concatenation strategy for proteome profiling of human MCF10A cells. Proteomics 11, 2019–2026 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Livesay, E. A. et al. Fully automated four-column capillary LC–MS system for maximizing throughput in proteomic analyses. Anal. Chem. 80, 294–302 (2008)

    Article  CAS  PubMed  Google Scholar 

  46. Mayampurath, A. M. et al. DeconMSn: a software tool for accurate parent ion monoisotopic mass determination for tandem mass spectra. Bioinformatics 24, 1021–1023 (2008)

    Article  CAS  PubMed  Google Scholar 

  47. Kim, S. et al. The generating function of CID, ETD, and CID/ETD pairs of tandem mass spectra: applications to database search. Mol. Cell Proteomics 9, 2840–2852 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Monroe, M. E., Shaw, J. L., Daly, D. S., Adkins, J. N. & Smith, R. D. MASIC: a software program for fast quantitation and flexible visualization of chromatographic profiles from detected LC-MS(/MS) features. Comput. Biol. Chem. 32, 215–217 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zuluaga, A. F. et al. Neutropenia induced in outbred mice by a simplified low-dose cyclophosphamide regimen: characterization and applicability to diverse experimental models of infectious diseases. BMC Infect. Dis. 6, 55 (2006)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Blaser, J. In-vitro model for simultaneous simulation of the serum kinetics of two drugs with different half-lives. J. Antimicrob. Chemother. 15. (Suppl A). 125–130 (1985)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank B. Wright and C. Blinn of AstraZeneca for assisting with the establishment of the mouse infection model, R. E. Lee, M. Pollastri and Z. Maglika for critical discussions and advice, I. Keren and S. Rowe for reading of the manuscript, H. Brewer, V. Petyuk and D. Camp II for assistance with proteomics, and Z. Zheng for assistance with ChemDraw. This work was supported by NIH award T-RO1 AI085585 to K.L., by Arietis Corporation to M.D.L and K.C., by the NIH-NIAID IAA Y1-AI-8401 to J.N.A. and P41 GM103493-11 to R.D.S. Proteomic analysis was performed in the EMSL, a DOE-BER national scientific user facility at Pacific Northwest National Laboratory (PNNL). PNNL is a multi-program national laboratory operated by Battelle Memorial Institute for the DOE under contract DE-AC05-76RLO 1830.

Author information

Authors and Affiliations

Authors

Contributions

B.P.C., M.D.L., K.C. and K.L. designed the study, analysed results and wrote the manuscript. B.P.C. performed in vitro antibiotic susceptibility assays, collected samples for proteomics, and performed biofilm susceptibility studies and mouse infection models. V.M.I. assisted with in vitro susceptibility assays. E.S.N. and J.N.A. performed i-TRAQ proteomics and analysed results. L.E.F. participated in mouse infection model experiments. S.N.L performed hollow-fibre experiments. M.D.L. was responsible for histopathology. R.D.S. provided the proteomics measurement capabilities.

Corresponding author

Correspondence to K. Lewis.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Combinations of conventional antibiotics against stationary phase S. aureus.

Data are representative of 3 independent experiments. Error bars represent s.d.

Extended Data Figure 2 ADEP4 resistant strains are heat sensitive.

Wild-type S. aureus ATCC 33591 and 9 ADEP4 resistant isolates with mutations in clpP were grown for 20 h in MHB at 44 °C in 96-well polystyrene plates. Data are representative of 3 independent experiments. Error bars represent s.d.

Extended Data Figure 3 ADEP4 with rifampicin eradicates S. aureus in a hollow-fibre infection model.

Antibiotics were delivered at concentrations mimicking human dosing, while the concentration of ADEP was varied over time to match the pharmacokinetics in the mouse model. Data are representative of 3 independent experiments. Error bars represent s.d.

Extended Data Figure 4 Conventional bactericidal antibiotics target active processes in bacterial cells (green) resulting in death.

In a dormant persister (blue), the antibiotic binds an inactive target, producing no effect. ADEP4 activates and dysregulates ClpP in growing cells and in dormant persisters, resulting in eradication of the bacterial population.

Related audio

41586_2013_BFnature12790_MOESM98_ESM.mp3

Researcher Kim Lewis discusses how a compound could help treat the chronic infections that antibiotics fail to fight off.

Supplementary information

Supplementary Data

This file contains Supplementary Tables 1-2. (XLSX 686 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Conlon, B., Nakayasu, E., Fleck, L. et al. Activated ClpP kills persisters and eradicates a chronic biofilm infection. Nature 503, 365–370 (2013). https://doi.org/10.1038/nature12790

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12790

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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