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:

Deep learning-guided discovery of an antibiotic targeting Acinetobacter baumannii

Abstract

Acinetobacter baumannii is a nosocomial Gram-negative pathogen that often displays multidrug resistance. Discovering new antibiotics against A. baumannii has proven challenging through conventional screening approaches. Fortunately, machine learning methods allow for the rapid exploration of chemical space, increasing the probability of discovering new antibacterial molecules. Here we screened ~7,500 molecules for those that inhibited the growth of A. baumannii in vitro. We trained a neural network with this growth inhibition dataset and performed in silico predictions for structurally new molecules with activity against A. baumannii. Through this approach, we discovered abaucin, an antibacterial compound with narrow-spectrum activity against A. baumannii. Further investigations revealed that abaucin perturbs lipoprotein trafficking through a mechanism involving LolE. Moreover, abaucin could control an A. baumannii infection in a mouse wound model. This work highlights the utility of machine learning in antibiotic discovery and describes a promising lead with targeted activity against a challenging Gram-negative pathogen.

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: Machine learning-guided discovery of abaucin.
Fig. 2: Abaucin has a narrow phylogenetic spectrum of antibacterial activity.
Fig. 3: Abaucin inhibits lipoprotein trafficking in A. baumannii.
Fig. 4: Abaucin can suppress A. baumannii infection in a wound model.

Similar content being viewed by others

Data Availability

GenBank accession numbers for sequencing of abaucin-resistant mutants are BankIt2629921 – OP677864, OP677865, OP677866 and OP677867. GEO accession numbers for RNA sequencing datasets are GSE214305GSM6603484, GSM6603485, GSM6603486, GSM6603487, GSM6603488, GSM6603489 and GSM6603490. Source data are provided with this paper.

Code Availability

All custom code used for antibiotic prediction is open source and can be accessed without restriction at https://github.com/chemprop/chemprop. A cloned snapshot used for this paper is available at https://github.com/GaryLiu152/chemprop_abaucin. All commercial software used is described in Methods. Source data are provided with this paper.

References

  1. Antunes, L. C. S., Visca, P. & Towner, K. J. Acinetobacter baumannii: evolution of a global pathogen. Pathog. Dis. 71, 292–301 (2014).

    CAS  PubMed  Google Scholar 

  2. 2020 Antibacterial Agents in Clinical and Preclinical Development: An Overview and Analysis (World Health Organization, 2021); https://www.who.int/publications/i/item/9789240021303

  3. Walsh, C. Where will new antibiotics come from? Nat. Rev. Microbiol. 1, 65–70 (2003).

    CAS  PubMed  Google Scholar 

  4. 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).

    CAS  PubMed  Google Scholar 

  5. Stokes, J. M. et al. A deep learning approach to antibiotic discovery. Cell 180, 688–702 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Ma, Y. et al. Identification of antimicrobial peptides from the human gut microbiome using deep learning. Nat. Biotechnol. 40, 921–931 (2022).

    CAS  PubMed  Google Scholar 

  7. Lluka, T. & Stokes, J. M. Antibiotic discovery in the artificial intelligence era. Ann. N. Y. Acad. Sci. 1519, 74–93 (2023).

    CAS  PubMed  Google Scholar 

  8. Melander, R. J., Zurawski, D. V. & Melander, C. Narrow-spectrum antibacterial agents. MedChemComm 9, 12–21 (2018).

    CAS  PubMed  Google Scholar 

  9. Theriot, C. M. et al. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat. Commun. 5, 3114 (2014).

    PubMed  Google Scholar 

  10. Willing, B. P. et al. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology 139, 1844–1854 (2010).

    PubMed  Google Scholar 

  11. Turnbaugh, P. J. et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031 (2006).

    PubMed  Google Scholar 

  12. Kelly, J. R. et al. Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 82, 109–118 (2016).

    PubMed  Google Scholar 

  13. Wu, N. et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb. Ecol. 66, 462–470 (2013).

    CAS  PubMed  Google Scholar 

  14. Lee, H. S., Plechot, K., Gohil, S. & Le, J. Clostridium difficile: diagnosis and the consequence of over diagnosis. Infect. Dis. Ther. 10, 687–697 (2021).

    PubMed  PubMed Central  Google Scholar 

  15. Corsello, S. M. et al. The Drug Repurposing Hub: a next-generation drug library and information resource. Nat. Med. 23, 405–408 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Kaplan, E., Greene, N. P., Crow, A. & Koronakis, V. Insights into bacterial lipoprotein trafficking from a structure of LolA bound to the LolC periplasmic domain. Proc. Natl Acad. Sci. USA 115, E7389–E7397 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Tang, X. et al. Structural basis for bacterial lipoprotein relocation by the transporter LolCDE. Nat. Struct. Mol. Biol. 28, 347–355 (2021).

    CAS  PubMed  Google Scholar 

  18. Yang, K. et al. Analyzing learned molecular representations for property prediction. J. Chem. Inf. Model. 59, 3370–3388 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Landrum, G. RDKit: a software suite for cheminformatics, computational chemistry, and predictive modeling. https://www.rdkit.org/RDKit_Overview.pdf

  20. Seok, S. J. et al. Blockade of CCL2/CCR2 signalling ameliorates diabetic nephropathy in db/db mice. Nephrol. Dial. Transplant. 28, 1700–1710 (2013).

    CAS  PubMed  Google Scholar 

  21. Cerri, C. et al. The chemokine CCL2 mediates the seizure-enhancing effects of systemic inflammation. J. Neurosci. 36, 3777–3788 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Chargari, C. et al. Preclinical assessment of JNJ-26854165 (Serdemetan), a novel tryptamine compound with radiosensitizing activity in vitro and in tumor xenografts. Cancer Lett. 312, 209–218 (2011).

    CAS  PubMed  Google Scholar 

  23. Lehman, J. A. et al. Serdemetan antagonizes the Mdm2-HIF1α axis leading to decreased levels of glycolytic enzymes. PLoS ONE 8, e74741 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Stokes, J. M., Lopatkin, A. J., Lobritz, M. A. & Collins, J. J. Bacterial metabolism and antibiotic efficacy. Cell Metab. 30, 251–259 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Zheng, E. J., Stokes, J. M. & Collins, J. J. Eradicating bacterial persisters with combinations of strongly and weakly metabolism-dependent antibiotics. Cell Chem. Biol. 27, 1544–1552 (2020).

    CAS  PubMed  Google Scholar 

  26. Francino, M. P. Antibiotics and the human gut microbiome: dysbioses and accumulation of resistances. Front. Microbiol. 6, 1543 (2016).

    PubMed  PubMed Central  Google Scholar 

  27. Smits, W. K., Lyras, D., Lacy, D. B., Wilcox, M. H. & Kuijper, E. J. Clostridium difficile infection. Nat. Rev. Dis. Prim. 2, 16020 (2016).

    PubMed  Google Scholar 

  28. Sharma, S. et al. Mechanism of LolCDE as a molecular extruder of bacterial triacylated lipoproteins. Nat. Commun. 12, 4687 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Nicholson, W. L. & Maughan, H. The spectrum of spontaneous rifampin resistance mutations in the rpoB gene of Bacillus subtilis 168 spores differs from that of vegetative cells and resembles that of Mycobacterium tuberculosis. J. Bacteriol. 184, 4936–4940 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Baek, M. et al. Accurate prediction of protein structures and interactions using a three-track neural network. Science 373, 871–876 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Buel, G. R. & Walters, K. J. Can AlphaFold2 predict the impact of missense mutations on structure? Nat. Struct. Mol. Biol. 29, 1–2 (2022).

    CAS  PubMed  Google Scholar 

  32. Raivio, T. L., Leblanc, S. K. D. & Price, N. L. The Escherichia coli Cpx envelope stress response regulates genes of diverse function that impact antibiotic resistance and membrane integrity. J. Bacteriol. 195, 2755–2767 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Guest, R. L., Wang, J., Wong, J. L. & Raivio, T. L. A bacterial stress response regulates respiratory protein complexes to control envelope stress adaptation. J. Bacteriol. 199, e00153-17 (2017).

    PubMed  PubMed Central  Google Scholar 

  34. Delhaye, A., Laloux, G. & Collet, J.-F. The lipoprotein NlpE is a Cpx sensor that serves as a sentinel for protein sorting and folding defects in the Escherichia coli envelope. J. Bacteriol. 201, e00611-18 (2019).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Pathania, R. et al. Chemical genomics in Escherichia coli identifies an inhibitor of bacterial lipoprotein targeting. Nat. Chem. Biol. 5, 849–856 (2009).

    CAS  PubMed  Google Scholar 

  37. McLeod, S. M. et al. Small-molecule inhibitors of Gram-negative lipoprotein trafficking discovered by phenotypic screening. J. Bacteriol. 197, 1075–1082 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Manchanda, V., Sanchaita, S. & Singh, N. Multidrug resistant acinetobacter. J. Glob. Infect. Dis. 2, 291–304 (2010).

    PubMed  PubMed Central  Google Scholar 

  39. Davis, K. A., Moran, K. A., McAllister, C. K. & Gray, P. J. Multidrug-resistant Acinetobacter extremity infections in soldiers. Emerg. Infect. Dis. 11, 1218–1224 (2005).

    PubMed  PubMed Central  Google Scholar 

  40. Jin, W. et al. Deep learning identifies synergistic drug combinations for treating COVID-19. Proc. Natl Acad. Sci. USA 118, e2105070118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Preuer, K. et al. DeepSynergy: predicting anti-cancer drug synergy with Deep Learning. Bioinformatics 34, 1538–1546 (2018).

    CAS  PubMed  Google Scholar 

  42. Jin, W., Barzilay, R. & Jaakkola, T. Junction tree variational autoencoder for molecular graph generation. In Proc. 35th International Conference on Machine Learning (eds Dy, J. & Krause, A.) 2323–2332 (PMLR, 2018).

  43. Pedregosa, F. et al. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830 (2011).

    Google Scholar 

  44. Deatherage, D. E. & Barrick, J. E. Identification of mutations in laboratory-evolved microbes from next-generation sequencing data using breseq. Methods Mol. Biol. 1151, 165–188 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    CAS  PubMed  Google Scholar 

  46. Ihaka, R. & Gentleman, R. R: a language for data analysis and graphics. J. Comput. Graph. Stat. 5, 299–314 (1996).

    Google Scholar 

  47. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  48. Karp, P. D. Pathway databases: a case study in computational symbolic theories. Science 293, 2040–2044 (2001).

    CAS  PubMed  Google Scholar 

  49. Keseler, I. M. et al. EcoCyc: fusing model organism databases with systems biology. Nucleic Acids Res. 41, D605–D612 (2013).

    CAS  PubMed  Google Scholar 

  50. Karp, P. D. et al. Pathway tools version 19.0 update: software for pathway/genome informatics and systems biology. Brief. Bioinform. 17, 877–890 (2016).

    CAS  PubMed  Google Scholar 

  51. Calvo-Villamañán, A. et al. On-target activity predictions enable improved CRISPR–dCas9 screens in bacteria. Nucleic Acids Res. 48, e64 (2020).

    PubMed  PubMed Central  Google Scholar 

  52. Depardieu, F. & Bikard, D. Gene silencing with CRISPRi in bacteria and optimization of dCas9 expression levels. Methods 172, 61–75 (2020).

    CAS  PubMed  Google Scholar 

  53. UniProt Consortium. UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Res. 49, D480–D489 (2021).

    Google Scholar 

  54. Altschul, S. F. et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25, 3389–3402 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Altschul, S. F. et al. Protein database searches using compositionally adjusted substitution matrices. FEBS J. 272, 5101–5109 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    CAS  PubMed  Google Scholar 

  57. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 30, 70–82 (2021).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. French from McMaster University for technical assistance with fluorescence microscopy experiments. This work was supported by the David Braley Centre for Antibiotic Discovery (to J.M.S.); the Weston Family Foundation (POP and Catalyst to J.M.S.); the Audacious Project (to J.J.C. and J.M.S.); the C3.ai Digital Transformation Institute (to R.B.); the Abdul Latif Jameel Clinic for Machine Learning in Health (to R.B.); the DTRA Discovery of Medical Countermeasures Against New and Emerging (DOMANE) threats program (to R.B.); the DARPA Accelerated Molecular Discovery program (to R.B.); the Canadian Institutes of Health Research (FRN-156361 to B.K.C.); Genome Canada GAPP (OGI-146 to M.G.S.); the Canadian Institutes of Health Research (FRN-148713 to M.G.S.); the Faculty of Health Sciences of McMaster University (to J.M.); the Boris Family (to J.M.); a Marshall Scholarship (to K.S.); and the DOE BER (DE-FG02-02ER63445 to A.C-P.).

Author information

Authors and Affiliations

Authors

Contributions

J.M.S. and J.J.C. conceptualized the study; J.M.S., G.L., K.S. and W.J. performed model building and training; J.M.S., D.B.C., K.R. and A.C-P. performed mechanistic investigations; J.M.S., K.R. and S.A.S. performed spectrum of activity experiments; J.C.M. conducted mouse model experiments; M.F. performed chemical synthesis; J.M.S. and J.J.C. wrote the paper; J.M.S., J.J.C., R.B., T.J., M.G.S., B.K.C. and J.M. supervised the research.

Corresponding authors

Correspondence to James J. Collins or Jonathan M. Stokes.

Ethics declarations

Competing interests

J.M.S. is cofounder and scientific director of Phare Bio. J.J.C. is cofounder and scientific advisory board chair of Phare Bio. J.J.C. is cofounder and scientific advisory board chair of Enbiotix. The other authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Jean Francois Collet and the other, anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Model training data and prediction.

(a) Replicate plot showing primary screening data of 7,684 small molecules for those that inhibited the growth of A. baumannii ATCC 17978 in LB medium at 50 µM. (b) Rank-ordered growth inhibition data of the prioritized 240 molecules from our prediction set that were selected for empirical validation (top); rank-ordered growth inhibition data of the 240 predicted molecules with the lowest prediction score (middle); rank-ordered growth inhibition data of the 240 predicted molecules with the highest prediction score that were not found in the training dataset (bottom). Experiments were conducted in biological duplicate. Individual replicates with means connected are plotted. Dashed horizontal line represents the stringent hit cut-off of >80% growth inhibition at 50 µM. (c) Growth inhibition of A. baumannii by abaucin (blue) and serdemetan (red) in LB medium. Experiments were conducted in biological duplicate. Individual replicates with means connected are plotted. The structure of serdemetan is shown. (d) Growth kinetics of A. baumannii cells after treatment with abaucin at varying concentrations for 6 hours. Experiments were conducted in biological duplicate. Individual replicates with means connected are plotted.

Extended Data Fig. 2 Antibacterial activity of abaucin against human commensal species.

(a) Growth inhibition of A. baumannii ATCC 17978 by ampicillin (blue) and ciprofloxacin (red) in LB medium. Experiments were conducted in biological duplicate. Individual replicates with means connected are plotted. (b) Growth inhibition of B. breve by abaucin. Experiments were conducted in biological duplicate. (c) Growth inhibition of B. longum by abaucin. Experiments were conducted in biological duplicate. Individual replicates with means connected are plotted. (d) Non-validated (see Fig. 2e) growth inhibition of E. lenta by abaucin. Experiments were conducted in biological duplicate. Individual replicates with means connected are plotted.

Extended Data Fig. 3 Abaucin mechanism of action.

(a–h) Growth inhibition of wildtype A. baumannii (WT) and the four independent abaucin-resistant mutants by a collection of diverse antibiotics. From left to right for each plot, the mutants are: A362T variant 1, Y394F, intergenic, and A362T variant 2. Experiments were conducted in biological duplicate. Note that the abaucin-resistant mutants do not display cross-resistance to other antibiotics. (i) Structural prediction of wildtype A. baumannii LolE using RoseTTAFold (bottom), with the structural error estimate of each amino acid (top). Position 362 is highlighted orange and resides in a disordered region of the protein. (j) same as (i), except with the Y362T abaucin-resistant mutant of LolE. (k) RNA sequencing of wildtype A. baumannii treated with 5x MIC abaucin for 4.5 hr (top) or 6 hr (bottom). Data are the mean of biological duplicates. Transcript abundance is normalized to no-drug control cultures grown in identical conditions. Vertical black lines show statistical significance cut-off values. Note the highly significant downregulation of genes involved in the electron transport chain and transmembrane ion transport. (l) Growth inhibition of A. baumannii harboring an empty CRISPRi vector (red), or three distinct sgRNAs targeting lolE (blue, teal, and green). All strains were grown in LB medium without induction. Experiments were conducted in biological duplicate. Individual replicates with means connected are plotted. (m) qPCR quantifying the expression of lolE relative to the housekeeping gene gltA (left) and gyrB (right) in all four abaucin resistant mutants, normalized to wildtype A. baumannii. Experiments were conducted in biological duplicate with technical triplicates. Bar height represents mean expression.

Supplementary information

Supplementary Information

Supplementary Tables 1–7 and Note.

Reporting Summary

Supplementary Data

Supplementary Data 1: Growth inhibition data against A. baumannii for model training. Supplementary Data 2: Model prediction scores of compounds in the Drug Repurposing Hub. Supplementary Data 3: Experimental validation of (prioritized/poorest/top 240) predictions from the Drug Repurposing Hub. Supplementary Data 4: GO enrichment for up- and down-regulated transcripts in A. baumannii treated with 5x MIC abaucin.

Source data

Source Data Fig. 4a

Raw data of measured bacterial load from mouse wound infection models.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, G., Catacutan, D.B., Rathod, K. et al. Deep learning-guided discovery of an antibiotic targeting Acinetobacter baumannii. Nat Chem Biol 19, 1342–1350 (2023). https://doi.org/10.1038/s41589-023-01349-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01349-8

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology