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.

  • Protocol
  • Published:

An LC-MS/MS assay and complementary web-based tool to quantify and predict compound accumulation in E. coli

Abstract

Novel classes of broad-spectrum antibiotics have been extremely difficult to discover, largely due to the impermeability of the Gram-negative membranes coupled with a poor understanding of the physicochemical properties a compound should possess to promote its accumulation inside the cell. To address this challenge, numerous methodologies for assessing intracellular compound accumulation in Gram-negative bacteria have been established, including classic radiometric and fluorescence-based methods. The recent development of accumulation assays that utilize liquid chromatography–tandem mass spectrometry (LC-MS/MS) have circumvented the requirement for labeled compounds, enabling assessment of a substantially broader range of small molecules. Our unbiased study of accumulation trends in Escherichia coli using an LC-MS/MS-based assay led to the development of the eNTRy rules, which stipulate that a compound is most likely to accumulate in E. coli if it has an ionizable Nitrogen, has low Three-dimensionality and is relatively Rigid. To aid in the implementation of the eNTRy rules, we developed a complementary web tool, eNTRyway, which calculates relevant properties and predicts compound accumulation. Here we provide a comprehensive protocol for analysis and prediction of intracellular accumulation of small molecules in E. coli using an LC-MS/MS-based assay (which takes ~2 d) and eNTRyway, a workflow that is readily adoptable by any microbiology, biochemistry or chemical biology laboratory.

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: Stages of the accumulation workflow.
Fig. 2: Accumulation assay workflow.
Fig. 3: Procedure for prediction of compound accumulation in bacteria using eNTRyway.
Fig. 4: Validation of compound accumulation in E. coli.
Fig. 5: Calibration curve and multiple reaction monitoring.

Similar content being viewed by others

Data availability

The main data discussed in this protocol are available in the supporting primary research papers (https://doi.org/10.1038/nature22308 and https://doi.org/10.1038/s41564-019-0604-5). Source data are provided with this paper. Additional requests should be addressed to the corresponding authors.

Code availability

Source code for eNTRyway for local use is available on GitHub (https://github.com/HergenrotherLab/entry-cli).

References

  1. Centers for Disease Control and Prevention. Antibiotic Resistance Threats in the United States, 2019. https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threats-report-508.pdf (2019).

  2. Tommasi, R., Brown, D. G., Walkup, G. K., Manchester, J. I. & Miller, A. A. ESKAPEing the labyrinth of antibacterial discovery. Nat. Rev. Drug. Discov. 14, 529–542 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Richter, M. F. & Hergenrother, P. J. The challenge of converting Gram-positive-only compounds into broad-spectrum antibiotics. Ann. NY Acad. Sci. 1435, 18–38 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Rice, L. B. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J. Infect. Dis. 197, 1079–1081 (2008).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  6. Gladki, A., Kaczanowski, S., Szczesny, P. & Zielenkiewicz, P. The evolutionary rate of antibacterial drug targets. BMC Bioinforma. 14, 36–36 (2013).

    Article  Google Scholar 

  7. Krishnamoorthy, G. et al. Synergy between active efflux and outer membrane diffusion defines rules of antibiotic permeation into Gram-negative bacteria. mBio 8, e01172–172017 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Krishnamoorthy, G. et al. Breaking the permeability barrier of Escherichia coli by controlled hyperporination of the outer membrane. Antimicrob. Agents Chemother. 60, 7372–7381 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bazile, S., Moreau, N., Bouzard, D. & Essiz, M. Relationships among antibacterial activity, inhibition of DNA gyrase, and intracellular accumulation of 11 fluoroquinolones. Antimicrob. Agents Chemother. 36, 2622–2627 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cai, H., Rose, K., Liang, L. H., Dunham, S. & Stover, C. Development of a liquid chromatography/mass spectrometry-based drug accumulation assay in Pseudomonas aeruginosa. Anal. Biochem. 385, 321–325 (2009).

    Article  CAS  PubMed  Google Scholar 

  11. Capobianco, J. O. & Goldman, R. C. Macrolide transport in Escherichia coli strains having normal and altered OmpC and/or OmpF porins. Int. J. Antimicrob. Agents 4, 183–189 (1994).

    Article  CAS  PubMed  Google Scholar 

  12. Chopra, I. Transport of tetracyclines into Escherichia coli requires a carboxamide group at the C2 position of the molecule. J. Antimicrob. Chemother. 18, 661–666 (1986).

    Article  CAS  PubMed  Google Scholar 

  13. de Cristóbal, R. E., Vincent, P. A. & Salomón, R. A. Multidrug resistance pump AcrAB-TolC is required for high-level, Tet(A)-mediated tetracycline resistance in Escherichia coli. J. Antimicrob. Chemother. 58, 31–36 (2006).

    Article  PubMed  Google Scholar 

  14. Li, X. Z., Livermore, D. M. & Nikaido, H. Role of efflux pump(s) in intrinsic resistance of Pseudomonas aeruginosa: resistance to tetracycline, chloramphenicol, and norfloxacin. Antimicrob. Agents Chemother. 38, 1732–1741 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Piddock, L. J. V., Jin, Y.-F., Ricci, V. & Asuquo, A. E. Quinolone accumulation by Pseudomonas aeruginosa, Staphylococcus aureus and Escherichia coli. J. Antimicrob. Chemother. 43, 61–70 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Williams, K. J. & Piddock, L. J. Accumulation of rifampicin by Escherichia coli and Staphylococcus aureus. J. Antimicrob. Chemother. 42, 597–603 (1998).

    Article  CAS  PubMed  Google Scholar 

  17. Richter, M. F. et al. Predictive compound accumulation rules yield a broad-spectrum antibiotic. Nature 545, 299–304 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Motika, S. E. et al. A Gram-negative antibiotic active through inhibition of an essential riboswitch. J. Am. Chem. Soc. 142, 10856–10862 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Parker, E. N. et al. Implementation of permeation rules leads to a FabI inhibitor with activity against Gram-negative pathogens. Nat. Microbiol. 5, 67–75 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Li, Y. et al. First-generation structure-activity relationship studies of 2,3,4,9-tetrahydro-1H-carbazol-1-amines as CpxA phosphatase inhibitors. Bioorg. Med. Chem. Lett. 29, 1836–1841 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Lukežič, T. et al. Engineering atypical tetracycline formation in Amycolatopsis sulphurea for the production of modified chelocardin antibiotics. ACS Chem. Biol. 14, 468–477 (2019).

    Article  PubMed  CAS  Google Scholar 

  22. Masci, D. et al. Switching on the activity of 1,5-diaryl-pyrrole derivatives against drug-resistant ESKAPE bacteria: structure-activity relationships and mode of action studies. Eur. J. Med. Chem. 178, 500–514 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Hu, Y. et al. Discovery of pyrido[2,3-b]indole derivatives with gram-negative activity targeting both DNA gyrase and topoisomerase IV. J. Med. Chem. 63, 9623–9649 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Andrews, L. D. et al. Optimization and mechanistic characterization of pyridopyrimidine inhibitors of bacterial biotin carboxylase. J. Med. Chem. 62, 7489–7505 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cohen, F. et al. Optimization of LpxC inhibitors for antibacterial activity and cardiovascular safety. ChemMedChem 14, 1560–1572 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Perlmutter, S. J. et al. Compound uptake into E. coli can be facilitated by N-alkyl guanidiniums and pyridiniums. ACS Infect. Dis. 7, 162–173 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Munoz, K. A. & Hergenrother, P. J. Facilitating compound entry as a means to discover antibiotics for Gram-negative bacteria. Acc. Chem. Res. 54, 1322–1333 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. O’Shea, R. & Moser, H. E. Physicochemical properties of antibacterial compounds: implications for drug discovery. J. Med. Chem. 51, 2871–2878 (2008).

    Article  PubMed  CAS  Google Scholar 

  29. Thanassi, D. G., Suh, G. S. B. & Nikaido, H. Role of outer membrane barrier in efflux-mediated tetracycline resistance in Escherichia coli. J. Bacteriol. 177, 998–1007 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Leive, L., Telesetsky, S., Coleman, W. G. J. & Carr, D. Tetracyclines of various hydrophobicities as a probe for permeability of Escherichia coli outer membranes. Antimicrob. Agents Chemother. 25, 539–544 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lindley, E. V., Munske, G. R. & Magnuson, J. A. Kinetic analysis of tetracycline accumulation by Streptococcus faecalis. J. Bacteriol. 158, 334–336 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sigler, A., Schubert, P., Hillen, W. & Niederweis, M. Permeation of tetracyclines through membranes of liposomes and Escherichia coli. Eur. J. Biochem. 267, 527–534 (2000).

    Article  CAS  PubMed  Google Scholar 

  33. Vergalli, J. et al. Spectrofluorimetric quantification of antibiotic drug concentration in bacterial cells for the characterization of translocation across bacterial membranes. Nat. Protoc. 13, 1348–1361 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Cinquin, B. et al. Microspectrometric insights on the uptake of antibiotics at the single bacterial cell level. Sci. Rep. 5, 17968 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Masi, M. et al. Fluorescence enlightens RND pump activity and the intrabacterial concentration of antibiotics. Res. Microbiol. 169, 432–441 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Renggli, S., Keck, W., Jenal, U. & Ritz, D. Role of autofluorescence in flow cytometric analysis of Escherichia coli treated with bactericidal antibiotics. J. Bacteriol. 195, 4067–4073 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Six, D. A., Krucker, T. & Leeds, J. A. Advances and challenges in bacterial compound accumulation assays for drug discovery. Curr. Opin. Chem. Biol. 44, 9–15 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Davis, T. D., Gerry, C. J. & Tan, D. S. General platform for systematic quantitative evaluation of small-molecule permeability in bacteria. ACS Chem. Biol. 9, 2535–2544 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Volkmer, B. & Heinemann, M. Condition-dependent cell volume and concentration of Escherichia coli to facilitate data conversion for systems biology modeling. PLoS One 6, e23126 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Prochnow, H. et al. Subcellular quantification of uptake in Gram-negative bacteria. Anal. Chem. 91, 1863–1872 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Iyer, R. et al. Evaluating LC–MS/MS to measure accumulation of compounds within bacteria. ACS Infect. Dis. 4, 1336–1345 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Smith, P. A. et al. Optimized arylomycins are a new class of Gram-negative antibiotics. Nature 561, 189–194 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Rhomberg, P. R., Sader, H. S. & Jones, R. N. Reproducibility of daptomycin MIC results using dry-form commercial trays with appropriate supplemental calcium content. Int. J. Antimicrob. Agents 25, 274–275 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. O’Boyle, N. M. et al. Open Babel: an open chemical toolbox. J. Cheminformatics 3, 33 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Richter for optimization and development of the LC-MS/MS-based accumulation assay, and we thank B. Drown for the creation of the web tool eNTRyway. This work was supported by the University of Illinois and the NIH (R01AI136773).

Author information

Authors and Affiliations

Authors

Contributions

E.J.G. and Z.L. performed experiments. All authors wrote the manuscript and were involved in editing of the final manuscript.

Corresponding author

Correspondence to Paul J. Hergenrother.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Kim Lewis 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.

Related links

Key references using this protocol:

Richter, M. F. et al. Nature 545, 299–304 (2017): https://doi.org/10.1038/nature22308

Parker, E. N. et al. Nat. Microbiol. 5, 67–75 (2020): https://doi.org/10.1038/s41564-019-0604-5

Motika, S. E. et al. J. Am. Chem. Soc. 142, 10856–10862 (2020): https://doi.org/10.1021/jacs.0c04427

Extended data

Extended Data Fig. 1 Importance of amine steric accessibility and amphiphilic moment (vsurf_A).

a, Primary amines demonstrate higher accumulation than the mono-methyl amine, di-methyl amine, tri-methyl amine and amide comparisons. Primary amines on primary carbons also show improved accumulation over primary amines on secondary or tertiary carbons. b, Increasing amphiphilic moment trends with increasing accumulation. Accumulation is reported in nmol/1012 CFUs. Data are taken from Richter et al.17 with permission.

Extended Data Fig. 2 Screenshot of the ‘input’ box for SMILES strings.

SMILES strings are canonicalized using Open Babel GUI.

Extended Data Fig. 3 Screenshots of the process of predicting accumulation using the web tool eNTRyway.

SMILES strings are submitted to eNTRyway, and compounds are prioritized for evaluation based on how closely they meet the eNTRy rules. In the example here, both ampicillin and 6-DNM-NH3 meet all of the criteria and are predicted to accumulate, whereas penicillin and DNM are not. A portion of this figure is taken from Parker et al.19 with permission.

Supplementary information

Source data

Source Data Fig. 4

Raw accumulation data.

Source Data Fig. 5

Calibration curve and mass spectrum.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Geddes, E.J., Li, Z. & Hergenrother, P.J. An LC-MS/MS assay and complementary web-based tool to quantify and predict compound accumulation in E. coli. Nat Protoc 16, 4833–4854 (2021). https://doi.org/10.1038/s41596-021-00598-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-021-00598-y

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research