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:

Identification of novel inhibitors for mycobacterial polyketide synthase 13 via in silico drug screening assisted by the parallel compound screening with genetic algorithm-based programs

Abstract

Identifying small compounds capable of inhibiting Mycobacterium tuberculosis polyketide synthase 13 (Pks13), in charge of final step of mycolic acid biosynthesis, could lead to the development of a novel antituberculosis drug. This study screened for lead compounds capable of targeting M. tuberculosis Pks13 from a chemical library comprising 154,118 compounds through multiple in silico docking simulations. The parallel compound screening (PCS), conducted via two genetic algorithm-based programs was applied in the screening strategy. Out of seven experimentally validated compounds, four compounds showed inhibitory effects on the growth of the model mycobacteria (Mycobacterium smegmatis). Subsequent docking simulation of analogs of the promising leads with the assistance of PCS resulted in the identification of three additional compounds with potent antimycobacterial effects (compounds A1, A2, and A5). Further, molecular dynamics simulation predicted stable interaction between M. tuberculosis Pks13 active site and compound A2, which showed potent antimycobacterial activity comparable to that of isoniazid. The present study demonstrated the efficacy of in silico structure-based drug screening through PCS in antituberculosis drug discovery.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Lönnroth K, Castro KG, Chakaya JM, Chauhan LS, Floyd K, Glaziou P, et al. Tuberculosis control and elimination 2010-50: cure, care, and social development. Lancet. 2010;375:1814–29.

    Article  Google Scholar 

  2. Dye C, Williams BG. The population dynamics and control of tuberculosis. Science. 2010;328:856–61.

    Article  CAS  Google Scholar 

  3. Torres JN, Paul LV, Rodwell TC, Victor TC, Amallraja AM, Elghraoui A, et al. Novel katG mutations causing isoniazid resistance in clinical M. tuberculosis isolates. Emerg Microbes Infect. 2015;4:e42

    Article  CAS  Google Scholar 

  4. Shaku M, Ealand C, Kana BD. Cell surface biosynthesis and remodeling pathways in mycobacteria reveal new drug targets. Front Cell Infect Microbiol. 2020;10:603382.

    Article  CAS  Google Scholar 

  5. Takayama K, Wang C, Besra GS. Pathway to synthesis and processing of mycolic acids in Mycobacterium tuberculosis. Clin Microbiol Rev. 2005;18:81–101.

    Article  CAS  Google Scholar 

  6. Portevin D, De sousa-D'auria C, Houssin C, Grimaldi C, Chami M, Daffe M, et al. A polyketide synthase catalyzes the last condensation step of mycolic acid biosynthesis in mycobacteria and related organisms. Proc Natl Acad Sci USA. 2004;101:314–9.

    Article  CAS  Google Scholar 

  7. Gavalda S, Leger M, van der Rest B, Stella A, Bardou F, Montrozier H, et al. The Pks13/FadD32 crosstalk for the biosynthesis of mycolic acids in Mycobacterium tuberculosis. J Biol Chem. 2009;284:19255–64.

    Article  CAS  Google Scholar 

  8. Gavalda S, Bardou F, Laval F, Bon C, Malaga W, Chalut C, et al. The polyketide synthase Pks13 catalyzes a novel mechanism of lipid transfer in mycobacteria. Chem Biol. 2014;21:1660–9.

    Article  CAS  Google Scholar 

  9. Lun S, Xiao S, Zhang W, Wang S, Gunosewoyo H, Yu LF, et al. Therapeutic potential of coumestan Pks13 inhibitors for tuberculosis. Antimicrob Agents Chemother. 2021;65:e02190–20.

    Article  CAS  Google Scholar 

  10. Ioerger TR, O’Malley T, Liao R, Guinn KM, Hickey MJ, Mohaideen N, et al. Identification of new drug targets and resistance mechanisms in Mycobacterium tuberculosis. PLoS ONE. 2013;8:e75245

    Article  CAS  Google Scholar 

  11. Wilson R, Kumar P, Parashar V, Vilcheze C, Veyron-Churlet R, Freundlich JS, et al. Antituberculosis thiophenes define a requirement for Pks13 in mycolic acid biosynthesis. Nat Chem Biol. 2013;9:499–506.

    Article  CAS  Google Scholar 

  12. North EJ, Jackson M, Lee RE. New approaches to target the mycolic acid biosynthesis pathway for the development of tuberculosis therapeutics. Curr Pharm Des. 2014;20:4357–78.

    Article  CAS  Google Scholar 

  13. Aggarwal A, Parai MK, Shetty N, Wallis D, Woolhiser L, Hastings C, et al. Development of a novel lead that targets M. tuberculosis polyketide synthase 13. Cell. 2017;170:249–59.e225.

    Article  CAS  Google Scholar 

  14. Wilson C, Ray P, Zuccotto F, Hernandez J, Aggarwal A, Mackenzie C, et al. Optimization of TAM16, a benzofuran that inhibits the thioesterase activity of Pks13; evaluation toward a preclinical candidate for a novel antituberculosis clinical target. J Med Chem. 2022;65:409–23.

    Article  CAS  Google Scholar 

  15. Koseki Y, Aoki S. Computational medicinal chemistry for rational drug design: Identification of novel chemical structures with potential anti-tuberculosis activity. Curr Top Med Chem. 2014;14:176–88.

    Article  CAS  Google Scholar 

  16. Kuriki K, Taira J, Kuroki M, Sakamoto H, Aoki S. Computer-assisted screening of mycobacterial growth inhibitors: Exclusion of frequent hitters with the assistance of the multiple target screening method. Int J Mycobacteriol. 2021;10:307–11.

    CAS  PubMed  Google Scholar 

  17. Izumizono Y, Arevalo S, Koseki Y, Kuroki M, Aoki S. Identification of novel potential antibiotics for tuberculosis by in silico structure-based drug screening. Eur J Med Chem. 2011;46:1849–56.

    Article  CAS  Google Scholar 

  18. Kanetaka H, Koseki Y, Taira J, Umei T, Komatsu H, Sakamoto H, et al. Discovery of InhA inhibitors with anti-mycobacterial activity through a matched molecular pair approach. Eur J Med Chem. 2015;94:378–85.

    Article  CAS  Google Scholar 

  19. Taira J, Umei T, Inoue K, Kitamura M, Berenger F, Sacchettini JC, et al. Improvement of the novel inhibitor for Mycobacterium enoyl-acyl carrier protein reductase (InhA): a structure-activity relationship study of KES4 assisted by in silico structure-based drug screening. J Antibiot. 2020;73:372–81.

    Article  CAS  Google Scholar 

  20. Taira J, Ito T, Nakatani H, Umei T, Baba H, Kawashima S, et al. In silico structure-based drug screening of novel antimycobacterial pharmacophores by DOCK-GOLD tandem screening. Int J Mycobacteriol. 2017;6:142–8.

    Article  CAS  Google Scholar 

  21. Taira J, Morita K, Kawashima S, Umei T, Baba H, Maruoka T, et al. Identification of a novel class of small compounds with anti-tuberculosis activity by in silico structure-based drug screening. J Antibiot. 2017;70:1057–64.

    Article  CAS  Google Scholar 

  22. Nakashima J, Takeuchi M, Kawamoto S, Monobe K, Taira J, Aoki S. Establishing parallel compound screening and identification of novel antimicrobial compounds targeting Staphylococcus aureus dihydrofolate reductase. J Appl Pharm Sci. 2022, in press.

  23. Ewing TJ, Makino S, Skillman AG, Kuntz ID. DOCK 4.0: search strategies for automated molecular docking of flexible molecule databases. J Comput Aided Mol Des. 2001;15:411–28.

    Article  CAS  Google Scholar 

  24. Verdonk ML, Cole JC, Hartshorn MJ, Murray CW, Taylor RD. Improved protein-ligand docking using GOLD. Proteins. 2003;52:609–23.

    Article  CAS  Google Scholar 

  25. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010;31:455–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46:3–26.

    Article  CAS  Google Scholar 

  27. Durant JL, Leland BA, Henry DR, Nourse JG. Reoptimization of MDL keys for use in drug discovery. J Chem Inf Comput Sci. 2002;42:1273–80.

    Article  CAS  Google Scholar 

  28. T JAS, J R, Rajan A, Shankar V. Features of the biochemistry of Mycobacterium smegmatis, as a possible model for Mycobacterium tuberculosis. J Infect Public Health. 2020;13:1255–64.

    Article  Google Scholar 

  29. Bloemberg GV, Keller PM, Stucki D, Trauner A, Borrell S, Latshang T, et al. Acquired resistance to bedaquiline and delamanid in therapy for tuberculosis. N Engl J Med. 2015;373:1986–8.

    Article  Google Scholar 

  30. Olayanju O, Limberis J, Esmail A, Oelofse S, Gina P, Pietersen E, et al. Long-term bedaquiline-related treatment outcomes in patients with extensively drug-resistant tuberculosis from South Africa. Eur Respir J. 2018;51.

  31. Marrakchi H, Laneelle MA, Daffe M. Mycolic acids: structures, biosynthesis, and beyond. Chem Biol. 2014;21:67–85.

    Article  CAS  Google Scholar 

  32. Cruz JN, Costa JFS, Khayat AS, Kuca K, Barros CAL, Neto A. Molecular dynamics simulation and binding free energy studies of novel leads belonging to the benzofuran class inhibitors of Mycobacterium tuberculosis Polyketide Synthase 13. J Biomol Struct Dyn. 2019;37:1616–27.

    Article  CAS  Google Scholar 

Download references

Funding

This work was supported in part by a grant from Takeda Science Foundation to JT and a Grant-in-Aid for Scientific Research (C) (26460145) to SA and a Grant-in-Aid for Transformative Research Areas (A) (21H05887) to JT from Japan Society for the Promotion of Science.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shunsuke Aoki.

Ethics declarations

Conflict of interest

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

Rights and permissions

Springer Nature or its licensor 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

Taira, J., Murakami, K., Monobe, K. et al. Identification of novel inhibitors for mycobacterial polyketide synthase 13 via in silico drug screening assisted by the parallel compound screening with genetic algorithm-based programs. J Antibiot 75, 552–558 (2022). https://doi.org/10.1038/s41429-022-00549-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41429-022-00549-z

Search

Quick links