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.

  • Letter
  • Published:

Enabling genetic analysis of diverse bacteria with Mobile-CRISPRi

Abstract

The vast majority of bacteria, including human pathogens and microbiome species, lack genetic tools needed to systematically associate genes with phenotypes. This is the major impediment to understanding the fundamental contributions of genes and gene networks to bacterial physiology and human health. Clustered regularly interspaced short palindromic repeats interference (CRISPRi), a versatile method of blocking gene expression using a catalytically inactive Cas9 protein (dCas9) and programmable single guide RNAs, has emerged as a powerful genetic tool to dissect the functions of essential and non-essential genes in species ranging from bacteria to humans1,2,3,4,5,6. However, the difficulty of establishing effective CRISPRi systems across bacteria is a major barrier to its widespread use to dissect bacterial gene function. Here, we establish ‘Mobile-CRISPRi’, a suite of CRISPRi systems that combines modularity, stable genomic integration and ease of transfer to diverse bacteria by conjugation. Focusing predominantly on human pathogens associated with antibiotic resistance, we demonstrate the efficacy of Mobile-CRISPRi in gammaproteobacteria and Bacillales Firmicutes at the individual gene scale, by examining drug–gene synergies, and at the library scale, by systematically phenotyping conditionally essential genes involved in amino acid biosynthesis. Mobile-CRISPRi enables genetic dissection of non-model bacteria, facilitating analyses of microbiome function, antibiotic resistances and sensitivities, and comprehensive screens for host–microorganism interactions.

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: Mobile-CRISPRi overview.
Fig. 2: Mobile-CRISPRi stability, transfer and knockdown efficiency.
Fig. 3: CRISPRi knockdown of folA increases sensitivity to trimethoprim in multiple species.
Fig. 4: A Mobile-CRISPRi library targeting auxotrophic genes in E.cloacae.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon request.

References

  1. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  Google Scholar 

  2. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  CAS  Google Scholar 

  3. Mimee, M., Tucker, A. C., Voigt, C. A. & Lu, T. K. Programming a human commensal bacterium, Bacteroides thetaiotaomicron, to sense and respond to stimuli in the murine gut microbiota. Cell Syst. 1, 62–71 (2015).

    Article  CAS  Google Scholar 

  4. Peters, J. M. et al. A comprehensive, CRISPR-based functional analysis of essential genes in bacteria. Cell 165, 1493–1506 (2016).

    Article  CAS  Google Scholar 

  5. Rock, J. M. et al. Programmable transcriptional repression in mycobacteria using an orthogonal CRISPR interference platform. Nat. Microbiol. 2, 16274 (2017).

    Article  CAS  Google Scholar 

  6. Tan, S. Z., Reisch, C. R. & Prather, K. L. J. A robust CRISPR interference gene repression system in Pseudomonas. J. Bacteriol. 200, e00575-17 (2018).

    Article  Google Scholar 

  7. Liu, X. et al. High‐throughput CRISPRi phenotyping identifies new essential genes in Streptococcus pneumoniae. Mol. Syst. Biol. 13, 931 (2017).

    Article  Google Scholar 

  8. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    Article  CAS  Google Scholar 

  9. Jost, M. et al. Combined CRISPRi/a-based chemical genetic screens reveal that rigosertib is a microtubule-destabilizing agent. Mol. Cell 68(1), 210–223 (2017).

    Article  CAS  Google Scholar 

  10. Vigouroux, A., Oldewurtel, E., Cui, L., Bikard, D. & Van Teeffelen, S. Tuning dCas9’s ability to block transcription enables robust, noiseless knockdown of bacterial genes. Mol. Syst. Biol. 14, e7899 (2018).

    Article  Google Scholar 

  11. Zhao, H. et al. Depletion of undecaprenyl pyrophosphate phosphatases disrupts cell envelope biogenesis in Bacillus subtilis. J. Bacteriol. 198, 2925–2935 (2016).

    Article  CAS  Google Scholar 

  12. van Opijnen, T., Bodi, K. L. & Camilli, A. Tn-seq: high-throughput parallel sequencing for fitness and genetic interaction studies in microorganisms. Nat. Methods 6, 767–772 (2009).

    Article  Google Scholar 

  13. Ji, W. et al. Specific gene repression by CRISPRi system transferred through bacterial conjugation. ACS Synth. Biol. 3, 929–931 (2014).

    Article  CAS  Google Scholar 

  14. Peters, J. E. Tn7. Microbiol. Spectr. 2, MDNA-30010-2014 (2014).

  15. Choi, K.-H. et al. A Tn7-based broad-range bacterial cloning and expression system. Nat. Methods 2, 443–448 (2005).

    Article  CAS  Google Scholar 

  16. Johnson, C. M. & Grossman, A. D. Integrative and conjugative elements (ICEs): what they do and how they work. Annu. Rev. Genet. 49, 577–601 (2015).

    Article  CAS  Google Scholar 

  17. Brophy, J. A. N. et al. Engineered integrative and conjugative elements for efficient and inducible DNA transfer to undomesticated bacteria. Nat. Microbiol. 3, 1043–1053 (2018).

    Article  CAS  Google Scholar 

  18. Bokulich, N. A. & Mills, D. A. Facility-specific ‘house’ microbiome drives microbial landscapes of artisan cheesemaking plants. Appl. Environ. Microbiol. 79, 5214–5223 (2013).

    Article  CAS  Google Scholar 

  19. Cardona, S. T., Selin, C. & Gislason, A. S. Genomic tools to profile antibiotic mode of action. Crit. Rev. Microbiol. 41, 465–472 (2015).

    Article  CAS  Google Scholar 

  20. Baccanari, D., Phillips, A., Smith, S., Sinski, D. & Burchall, J. Purification and properties of Escherichia coli dihydrofolate reductase. Biochemistry 14, 5267–5273 (1975).

    Article  CAS  Google Scholar 

  21. McMahon, S. A. et al. Extensive DNA mimicry by the ArdA anti-restriction protein and its role in the spread of antibiotic resistance. Nucleic Acids Res. 37, 4887–4897 (2009).

    Article  CAS  Google Scholar 

  22. Peters, J. E., Makarova, K. S., Shmakov, S. & Koonin, E. V. Recruitment of CRISPR–Cas systems by Tn7-like transposons. Proc. Natl Acad. Sci. USA 114, E7358–E7366 (2017).

    Article  CAS  Google Scholar 

  23. Choi, K.-H. & Schweizer, H. P. Mini-Tn7 insertion in bacteria with single attTn7 sites: example Pseudomonas aeruginosa. Nat. Protoc. 1, 153–161 (2006).

    Article  CAS  Google Scholar 

  24. Ferrières, L. et al. Silent mischief: bacteriophage Mu insertions contaminate products of Escherichia coli random mutagenesis performed using suicidal transposon delivery plasmids mobilized by broad-host-range RP4 conjugative machinery. J. Bacteriol. 192, 6418–6427 (2010).

    Article  Google Scholar 

  25. Choi, K.-H. et al. Genetic tools for select-agent-compliant manipulation of Burkholderia pseudomallei. Appl. Environ. Microbiol. 74, 1064–1075 (2008).

    Article  CAS  Google Scholar 

  26. Auchtung, J. M., Lee, C. A., Monson, R. E., Lehman, A. P. & Grossman, A. D. Regulation of a Bacillus subtilis mobile genetic element by intercellular signaling and the global DNA damage response. Proc. Natl Acad. Sci. USA 102, 12554–12559 (2005).

    Article  CAS  Google Scholar 

  27. Auchtung, J. M., Lee, C. A., Garrison, K. L. & Grossman, A. D. Identification and characterization of the immunity repressor (ImmR) that controls the mobile genetic element ICEBs1 of Bacillus subtilis. Mol. Microbiol. 64, 1515–1528 (2007).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. Koo, B.-M. et al. Construction and analysis of two genome-scale deletion libraries for Bacillus subtilis. Cell Syst. 4, 291–305 (2017).

    Article  CAS  Google Scholar 

  30. Kritikos, G. et al. A tool named Iris for versatile high-throughput phenotyping in microorganisms. Nat. Microbiol. 2, 17014 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Goldberg (Emory University) and H. Schweizer (University of Florida) for Tn7 plasmids, L. (Stanley) Qi (Stanford University) for a plasmid encoding human codon-optimized dCas9, the American Type Culture Collection, H. Mobley (University of Michigan), B. DeGrado (University of California, San Francisco), K. C. Huang (Stanford University), A. Banta (Stanford University) and P. Welander (Stanford University) for strains, J. Garbarino (University of California, San Francisco) and M. Jost (University of California, San Francisco) for help with flow cytometry, and the C.A.G. and O.S.R. labs for helpful comments. This work was supported by the NIH F32 GM108222 (to J.M.P.), the US Department of Agriculture National Institute of Food and Agriculture Hatch Project NYC-189438 (to J.E.P.), NIH R35 GM118061 and Innovative Genomics Institute, UC Berkeley (to C.A.G.), and NIAID R01 AI128214, Chan-Zuckerberg Biohub, CF Foundation Research Development Program, and Gilead Sciences Research Scholars Program in Cystic Fibrosis (to O.S.R).

Author information

Authors and Affiliations

Authors

Contributions

J.M.P., B.-M.K., M.M.H., A.D.G., J.E.P., J.N.E., R.J.D., C.A.G. and O.S.R. designed the study. J.M.P., B.-M.K., R.P., G.E.H., C.C.H., Y.F.I., C.H.S.L., J.Q. and M.R.S. performed the experiments. J.M.P., B.-M.K., R.P., G.E.H., Y.F.I. and J.S.H. analysed the data. J.M.P., B.-M.K., H.O., C.A.G. and O.S.R. wrote the manuscript.

Corresponding authors

Correspondence to Jason M. Peters, Carol A. Gross or Oren S. Rosenberg.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figures 1–11.

Reporting Summary

Supplementary Table 1

Primers and oligos used in this study.

Supplementary Table 2

Growth phenotypes for E. cloacae CRISPRi strains in minimal media (pooled screen).

Supplementary Table 3

Growth phenotypes for E. cloacae CRISPRi strains in minimal media (arrayed screen).

Supplementary Table 4

Plasmids used in this study.

Supplementary Table 5

Strains used in this study.

Supplementary Table 6

Next generation sequencing oligos used in this study.

Supplementary Table 7

MIC values for folA knockdown strains.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Peters, J.M., Koo, BM., Patino, R. et al. Enabling genetic analysis of diverse bacteria with Mobile-CRISPRi. Nat Microbiol 4, 244–250 (2019). https://doi.org/10.1038/s41564-018-0327-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-018-0327-z

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