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.

Sequence-specific antimicrobials using efficiently delivered RNA-guided nucleases

Abstract

Current antibiotics tend to be broad spectrum, leading to indiscriminate killing of commensal bacteria and accelerated evolution of drug resistance. Here, we use CRISPR-Cas technology to create antimicrobials whose spectrum of activity is chosen by design. RNA-guided nucleases (RGNs) targeting specific DNA sequences are delivered efficiently to microbial populations using bacteriophage or bacteria carrying plasmids transmissible by conjugation. The DNA targets of RGNs can be undesirable genes or polymorphisms, including antibiotic resistance and virulence determinants in carbapenem-resistant Enterobacteriaceae and enterohemorrhagic Escherichia coli. Delivery of RGNs significantly improves survival in a Galleria mellonella infection model. We also show that RGNs enable modulation of complex bacterial populations by selective knockdown of targeted strains based on genetic signatures. RGNs constitute a class of highly discriminatory, customizable antimicrobials that enact selective pressure at the DNA level to reduce the prevalence of undesired genes, minimize off-target effects and enable programmable remodeling of microbiota.

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: RGN constructs delivered by bacteriophage particles (ΦRGN) exhibit efficient and specific antimicrobial effects against strains harboring plasmid or chromosomal target sequences.
Figure 2: Characterization of ΦRGN-mediated killing of antibiotic-resistant bacteria.
Figure 3: ΦRGN particles elicit sequence-specific toxicity against enterohemorrhagic E. coli in vitro and in vivo.
Figure 4: Programmable remodeling of a synthetic microbial consortium.

References

  1. Centers for Disease Control and Prevention. Antibiotic Resistance Threats in the United States (2013).

  2. Nordmann, P., Dortet, L. & Poirel, L. Carbapenem resistance in Enterobacteriaceae: here is the storm!. Trends Mol. Med. 18, 263–272 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Barrangou, R. et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709–1712 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Garneau, J.E. et al. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 468, 67–71 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Mali, P., Esvelt, K.M. & Church, G.M. Cas9 as a versatile tool for engineering biology. Nat. Methods 10, 957–963 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Deltcheva, E. et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 471, 602–607 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rasheed, J.K. et al. Characterization of the extended-spectrum beta-lactamase reference strain, Klebsiella pneumoniae K6 (ATCC 700603), which produces the novel enzyme SHV-18. Antimicrob. Agents Chemother. 44, 2382–2388 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rasheed, J.K. et al. New Delhi metallo-β-lactamase-producing Enterobacteriaceae, United States. Emerg. Infect. Dis. 19, 870–878 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bikard, D., Hatoum-Aslan, A., Mucida, D. & Marraffini, L.A. CRISPR interference can prevent natural transformation and virulence acquisition during in vivo bacterial infection. Cell Host Microbe 12, 177–186 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Gomaa, A.A. et al. Programmable removal of bacterial strains by use of genome-targeting CRISPR-Cas systems. MBio 5, e00928–13 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Lutz, R. & Bujard, H. Independent and tight regulation of transcriptional units in Escherichia coli via the LacR/O, the TetR/O and AraC/I1–I2 regulatory elements. Nucleic Acids Res. 25, 1203–1210 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pérez-Mendoza, D. & de la Cruz, F. Escherichia coli genes affecting recipient ability in plasmid conjugation: are there any? BMC Genomics 10, 71 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Jacoby, G.A. Mechanisms of resistance to quinolones. Clin. Infect. Dis. 41 (suppl. 2), S120–S126 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Pennington, J.M. & Rosenberg, S.M. Spontaneous DNA breakage in single living Escherichia coli cells. Nat. Genet. 39, 797–802 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hayes, F. Toxins-antitoxins: plasmid maintenance, programmed cell death, and cell cycle arrest. Science 301, 1496–1499 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Mnif, B. et al. Molecular characterization of addiction systems of plasmids encoding extended-spectrum beta-lactamases in Escherichia coli. J. Antimicrob. Chemother. 65, 1599–1603 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Kaper, J.B., Nataro, J.P. & Mobley, H.L. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2, 123–140 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Desbois, A.P. & Coote, P.J. Utility of greater wax moth larva (Galleria mellonella) for evaluating the toxicity and efficacy of new antimicrobial agents. Adv. Appl. Microbiol. 78, 25–53 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Sonnenburg, J.L. & Fischbach, M.A. Community health care: therapeutic opportunities in the human microbiome. Sci. Transl. Med. 3, 78ps12 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Paddon, C.J. et al. High-level semi-synthetic production of the potent antimalarial artemisinin. Nature 496, 528–532 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Duan, F. & March, J.C. Engineered bacterial communication prevents Vibrio cholerae virulence in an infant mouse model. Proc. Natl. Acad. Sci. USA 107, 11260–11264 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lu, T.K. & Collins, J.J. Dispersing biofilms with engineered enzymatic bacteriophage. Proc. Natl. Acad. Sci. USA 104, 11197–11202 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lu, T.K. & Collins, J.J. Engineered bacteriophage targeting gene networks as adjuvants for antibiotic therapy. Proc. Natl. Acad. Sci. USA 106, 4629–4634 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Edgar, R., Friedman, N., Molshanski-Mor, S. & Qimron, U. Reversing bacterial resistance to antibiotics by phage-mediated delivery of dominant sensitive genes. Appl. Environ. Microbiol. 78, 744–751 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Seed, K.D., Lazinski, D.W., Calderwood, S.B. & Camilli, A. A bacteriophage encodes its own CRISPR/Cas adaptive response to evade host innate immunity. Nature 494, 489–491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jiang, W., Bikard, D., Cox, D., Zhang, F. & Marraffini, L.A. RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat. Biotechnol. 31, 233–239 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Vercoe, R.B. et al. Cytotoxic chromosomal targeting by CRISPR/Cas systems can reshape bacterial genomes and expel or remodel pathogenicity islands. PLoS Genet. 9, e1003454 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Williams, J.J. & Hergenrother, P.J. Artificial activation of toxin-antitoxin systems as an antibacterial strategy. Trends Microbiol. 20, 291–298 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Esvelt, K.M., Smidler, A.L., Catteruccia, F. & Church, G.M. Concerning RNA-guided gene drives for the alteration of wild populations. Elife e03401 (2014).

  31. Datta, S., Costantino, N. & Court, D.L. A set of recombineering plasmids for Gram-negative bacteria. Gene 379, 109–115 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Gibson, D.G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Sikorski, R.S. & Hieter, P. A system of shuttle vectors and yeast host strains designed for efficient manipulation of DNA in Saccharomyces cerevisiae. Genetics 122, 19–27 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Dwyer, D.J., Kohanski, M.A., Hayete, B. & Collins, J.J. Gyrase inhibitors induce an oxidative damage cellular death pathway in Escherichia coli. Mol. Syst. Biol. 3, 91 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Clinical and Laboratory Standards Institute. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically; Approved Standard. Seventh Edition. (Clinical and Laboratory Standards Institute, Wayne, Pennsylvania, USA, 2006).

  36. Chung, C.T., Niemela, S.L. & Miller, R.H. One-step preparation of competent Escherichia coli: transformation and storage of bacterial cells in the same solution. Proc. Natl. Acad. Sci. USA 86, 2172–2175 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chasteen, L., Ayriss, J., Pavlik, P. & Bradbury, A.R.M. Eliminating helper phage from phage display. Nucleic Acids Res. 34, e145 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Westwater, C. et al. Use of genetically engineered phage to deliver antimicrobial agents to bacteria: an alternative therapy for treatment of bacterial infections. Antimicrob. Agents Chemother. 47, 1301–1307 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dong, D., Sutaria, S., Hwangbo, J.Y. & Chen, P. A simple and rapid method to isolate purer M13 phage by isoelectric precipitation. Appl. Microbiol. Biotechnol. 97, 8023–8029 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Ramarao, N., Nielsen-Leroux, C. & Lereclus, D. The insect Galleria mellonella as a powerful infection model to investigate bacterial pathogenesis. J. Vis. Exp. 4392, 10.3791/4392 (2012).

Download references

Acknowledgements

We would like to thank R. Meyer (Institute for Cell and Molecular Biology, University of Texas Austin) for R1162, D.L. Court (Center for Cancer Research, National Cancer Institute at Frederick) for pSIM9, and A.R.M. Bradbury (Los Alamos National Laboratory) for M13cp. The authors thank J. Rubens for assistance with flow cytometry experiments and H. Gancz and D. Zurawski for assistance with the Galleria infection model. T.K.L. acknowledges support from the US National Institutes of Health (NIH) New Innovator Award (1DP2OD008435), an NIH National Centers for Systems Biology grant (1P50GM098792), the Defense Threat Reduction Agency (HDTRA1-14-1-0007), the US Army Research Laboratory and the US Army Research Office through the Institute for Soldier Nanotechnologies (W911NF13D0001) and the Henry L. and Grace Doherty Professorship in Ocean Utilization. R.J.C. is supported by funding from the NIH/National Institute of General Medical Sciences Interdepartmental Biotechnology Training Program (5T32 GM008334), and M.M. is a Howard Hughes Medical Institute International Student Research fellow and a recipient of a Fonds de recherche Santé Québec Master's Training Award.

Author information

Authors and Affiliations

Authors

Contributions

R.J.C. and M.M. designed and performed experiments. R.J.C., M.M. and T.K.L. conceived this study, analyzed the data, discussed results and wrote the manuscript.

Corresponding author

Correspondence to Timothy K Lu.

Ethics declarations

Competing interests

R.J.C., M.M. and T.K.L. have filed a provisional application with the US Patent and Trademark Office on this work.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8, Supplementary Tables 1–4 and Supplementary Discussion (PDF 1560 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Citorik, R., Mimee, M. & Lu, T. Sequence-specific antimicrobials using efficiently delivered RNA-guided nucleases. Nat Biotechnol 32, 1141–1145 (2014). https://doi.org/10.1038/nbt.3011

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3011

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