Log in

Identification of inhibitors for Agr quorum sensing system of Staphylococcus aureus by machine learning, pharmacophore modeling, and molecular dynamics approaches

  • Original Paper
  • Published:
Journal of Molecular Modeling Aims and scope Submit manuscript

Abstract

Context

Staphylococcus aureus is a highly pathogenic organism that is the most common cause of postoperative complications as well as severe infections like bacteremia and infective endocarditis. By mediating the formation of biofilms and the expression of virulent genes, the quorum sensing (QS) mechanism is a major contributor to the development of these diseases. By hindering its QS network, an innovative approach to avoiding this bacterial infection is taken. Targeting the AgrA of the Agr system serves as beneficial in holding the top position in the QS system cascade.

Methods

Using known AgrA inhibitors, the machine learning algorithms (artificial neural network, naïve Bayes, random forest, and support vector machine) and pharmacophore model were developed. The potential lead compounds were screened against the Zinc and COCONUT databases using the best pharmacophore hypothesis. The hits were then subjected second screening process using the best machine learning model. The predicted active compounds were then reranked based on the docking score. The stability of AgrA-lead compounds was studied using molecular dynamics approaches, and an ADME profile was also carried out. Five lead compounds, namely, CNP02386963,4,5-trihydroxy-2-[({7,13,14-trihydroxy-3,10-dioxo-2,9-dioxatetracyclo[6.6.2.04,16.011,15]hexadeca-1(14),4,6,8(16),11(15),12-hexaen-6-yl}oxy)methyl]benzoic acid, CNP0129274 4-(dimethylamino)-1,5,6,10,12,12a-hexahydroxy-6-methyl-3,11-dioxo-3,4,4a,5,5a,6,11,12a-octahydrotetracene-2-carboxamide, CNP0242717 3-Hydroxyasebotin, CNP0361624 3,4,5-trihydroxy-6-[(2,4,5,6,7-pentahydroxy-1-oxooctan-3-yl)oxy]oxane-2-carboxylic acid, and CNP0285058 2-{[4,5-dihydroxy-6-(hydroxymethyl)-3-[(3,4,5-trihydroxy-6-methyloxan-2-yl)oxy]oxan-2-yl]oxy}-2-(4-hydroxyphenyl)acetonitrile were obtained using the two-step virtual screening process. The molecular dynamics study revealed that the CNP0238696 was found to be stable in the binding pocket of AgrA. ADME profiles show that this compound has two Lipinski violations and low bioavailability. Further studies should be performed to assess the anti-biofilm activity of the lead compound in vitro.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price includes VAT (Germany)

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

References

  1. Christaki E, Marcou M, Tofarides A (2020) Antimicrobial resistance in bacteria: mechanisms, evolution, and persistence. J Mol Evol 88:26–40

    Article  CAS  PubMed  Google Scholar 

  2. Lee JH (2019) Perspectives towards antibiotic resistance: from molecules to population. J Microbiol (Seoul, Korea) 57:181–184

    Google Scholar 

  3. Liu W, Yang C, Gao R, Zhang C, Ou-Yang W, Feng Z, Zhang C, Pan X, Huang P, Kong D, Wang W (2021) Polymer composite sponges with inherent antibacterial, hemostatic, inflammation-modulating and proregenerative performances for methicillin-resistant Staphylococcus aureus -infected wound healing. Adv Healthcare Mater 10:2101247

    Article  CAS  Google Scholar 

  4. GBD (2019) Antimicrobial Resistance Collaborators (2023). Global mortality associated with 33 bacterial pathogens in 2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet (London, England) 400:2221–2248

    Google Scholar 

  5. Idrees M, Sawant S, Karodia N, Rahman A (2021) Staphylococcus aureus biofilm: morphology, genetics, pathogenesis and treatment strategies. Int J Environ Res Public Health 18:7602

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ganesh PS, Veena K, Senthil R, Iswamy K, Ponmalar EM, Mariappan M, Girija ASS, Vadivelu V, Nagarajan S, Challabathula D, Shankar EM (2022) Biofilm-associated Agr and Sar quorum sensing systems of Staphylococcus aureus are inhibited by 3-hydroxybenzoic acid derived from Illicium verum. ACS Omega 7:14653–14665

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lade, Chung SH, Lee Y, Kumbhar BV, Joo H-S, Kim Y-G, Yang Y-H, Kim J-S (2022) Thymol reduces agr-mediated virulence factor phenol-soluble modulin production in staphylococcus aureus. BioMed Research International 8221622

  8. Rajasree K, Fasim A, Gopal B (2016) Conformational features of the Staphylococcus aureus AgrA-promoter interactions rationalize quorum-sensing triggered gene expression. Biochem Biophys Reports 6:124–134

    Article  Google Scholar 

  9. Iobbi V, Brun P, Bernabé G, DouguéKentsop RA, Donadio G, Ruffoni B, Fossa P, Bisio A, De Tommasi N (2021) Labdane Diterpenoids from Salvia tingitana Etl. Synergize with Clindamycin against methicillin-resistant Staphylococcus aureus. Molecules (Basel, Switzerland) 26:6681

    Article  CAS  PubMed  Google Scholar 

  10. Leonard PG, Bezar IF, Sidote DJ, Stock AM (2012) Identification of a hydrophobic cleft in the LytTR domain of AgrA as a locus for small molecule interactions that inhibit DNA binding. Biochemistry 51:10035–10043

    Article  CAS  PubMed  Google Scholar 

  11. Wang B, Muir TW (2016) Regulation of virulence in Staphylococcus aureus: molecular mechanisms and remaining puzzles. Cell Chem Biol 23:214–224

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bernabè G, Dal Pra M, Ronca V, Pauletto A, Marzaro G, Saluzzo F, Stefani A, Artusi I, De Filippis V, Ferlin MG, Brun P, Castagliuolo I (2021) A novel Aza-derivative inhibits agr quorum sensing signaling and synergizes methicillin-resistant Staphylococcus aureus to clindamycin. Front Microbiol 12:610859

    Article  PubMed  PubMed Central  Google Scholar 

  13. Greenberg M, Kuo D, Jankowsky E, Long L, Hager C, Bandi K, Ma D, Manoharan D, Shoham Y, Harte W, Ghannoum MA, Shoham M (2018) Small-molecule AgrA inhibitors F12 and F19 act as antivirulence agents against Gram-positive pathogens. Sci Rep 8:14578

    Article  PubMed  PubMed Central  Google Scholar 

  14. Palaniappan B, Solomon AP, C DR, (2021) Targeting AgrA quorum sensing regulator by bumetanide attenuates virulence in Staphylococcus aureus - a drug repurposing approach. Life Sci 273:119306

    Article  CAS  PubMed  Google Scholar 

  15. Mahdally NH, George RF, Kashef MT, Al-Ghobashy M, Murad FE, Attia AS (2021) Staquorsin: a novel Staphylococcus aureus Agr-mediated quorum sensing inhibitor impairing virulence in vivo without notable resistance development. Front Microbiol 12:700494

    Article  PubMed  PubMed Central  Google Scholar 

  16. Sully EK, Malachowa N, Elmore BO, Alexander SM, Femling JK, Gray BM, DeLeo FR, Otto M, Cheung AL, Edwards BS, Sklar LA, Horswill AR, Hall PR, Gresham HD (2014) Selective chemical inhibition of agr quorum sensing in Staphylococcus aureus promotes host defense with minimal impact on resistance. PLoS Pathog 10:e1004174

    Article  PubMed  PubMed Central  Google Scholar 

  17. Kumari M, Tiwari N, Chandra S, Subbarao N (2018) Comparative analysis of machine learning based QSAR models and molecular docking studies to screen potential anti-tubercular inhibitors against InhA of mycobacterium tuberculosis. Int J Comput Biol Drug Des 11:209

    Article  Google Scholar 

  18. Liang JW, Wang MY, Wang S, Li SL, Li WQ, Meng FH (2020) Identification of novel CDK2 inhibitors by a multistage virtual screening method based on SVM, pharmacophore and docking model. J Enzyme Inhib Med Chem 35:235–244

    Article  CAS  PubMed  Google Scholar 

  19. Hall M, Frank E, Holmes G, Pfahringer B, Reutemann P, Witten IH (2009) The WEKA Data Mining Software: an update. ACM SIGKDD Explorations Newsl 11:10

    Article  Google Scholar 

  20. Zupan J, Gasteiger J (1993) Neural networks for chemists; an introduction. VCH Publishers, Weinheim (Germany)

  21. Friedman N, Geiger D, Goldszmidt M (1997) Bayesian network classifiers. Mach Learn 29:131–163. https://doi.org/10.1023/A:1007465528199

    Article  Google Scholar 

  22. Breiman L (2001) Random forest. Mach Learn 45:5–32

    Article  Google Scholar 

  23. Vapnik VN (1998) Statistical Learning Theory. Wiley

    Google Scholar 

  24. Schrödinger Release 2022-3 (2022) Schrödinger Release 2022-3: Phase, Schrödinger, LLC, New York

  25. Misra S, Saini M, Ojha H, Sharma D, Sharma K (2017) Pharmacophore modelling, atom-based 3D-QSAR generation and virtual screening of molecules projected for mPGES-1 inhibitory activity. SAR QSAR Environ Res 28:17–39

    Article  CAS  PubMed  Google Scholar 

  26. Dixon SL, Smondyrev AM, Knoll EH, Rao SN, Shaw DE, Friesner RA (2006) PHASE: a new engine for pharmacophore perception, 3D QSAR model development, and 3D database screening: 1. Methodology and preliminary results. J Comput Aided Mol Des 20:647–671

    Article  CAS  PubMed  Google Scholar 

  27. Schrödinger Release 2022-3 (2022) Schrödinger Release 2022-3: Glide, Schrödinger, LLC, New York, NY

  28. Schrödinger Release 2022-3 (2022) Schrödinger Release 2022-3: LigPrep, Schrödinger, LLC, New York, NY

  29. Schrödinger Release 2022-3 (2022) Schrödinger Release 2022-3: Protein Preparation Wizard; Epik, Schrödinger, LLC, New York, NY

  30. Schrödinger Release 2022-3 (2022) Schrödinger Release 2022-3: SiteMap, Schrödinger, LLC, New York, NY

  31. Schrödinger Release 2022-3 (2022) Schrödinger Release 2022-3: Prime, Schrödinger, LLC, New York, NY

  32. Schrödinger Release 2020-4 (2020) Schrödinger Release 2020-4: Desmond Molecular Dynamics System. D. E. Shaw Research. Maestro-Desmond Interoperability Tools, Schrödinger, New York NY

  33. Schrödinger Release 2022-3 (2022) Schrödinger Release 2022-3: QikProp, Schrödinger, LLC, New York, NY

  34. Opo FADM, Rahman MM, Ahammad F, Ahmed I, Bhuiyan MA, Asiri AM (2021) Structure based pharmacophore modeling, virtual screening, molecular docking and ADMET approaches for identification of natural anti-cancer agents targeting XIAP protein. Sci Rep 11:4049

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Jamal S, Grover A, Grover S (2019) Machine learning from molecular dynamics trajectories to predict caspase-8 inhibitors against Alzheimer’s disease. Front Pharmacol 10:780

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mishra S, Mallick PK, Tripathy HK, Bhoi AK, González-Briones A (2020) Performance evaluation of a proposed machine learning model for chronic disease datasets using an integrated attribute evaluator and an improved decision tree classifier. Appl Sci 10(22):8137

  37. Sarker IH, Kayes ASM, Watters P (2019) Effectiveness analysis of machine learning classification models for predicting personalized context-aware smartphone usage. J Big Data 6:57

    Article  Google Scholar 

  38. Chicco D, Jurman G (2020) The advantages of the Matthews correlation coefficient (MCC) over F1 score and accuracy in binary classification evaluation. BMC Genomics 21:6

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hdoufane L, Stoycheva J, Tadjer A, Villemin D, Najdoska-Bogdanov M, Bogdanov J, Cherqaoui D (2019) QSAR and molecular docking studies of indole-based analogs as HIV-1 attachment inhibitors. J Mol Struct 1193:429–443

    Article  CAS  Google Scholar 

  40. Giordano D, Biancaniello C, Argenio MA, Facchiano A (2022) Drug design by pharmacophore and virtual screening approach. Pharmaceuticals (Basel, Switzerland) 15:646

    Article  CAS  PubMed  Google Scholar 

  41. Srivastava SK, Rajasree K, Fasim A, Arakere G, Gopal B (2014) Influence of the AgrC-AgrA complex on the response time of Staphylococcus aureus quorum sensing. J Bacteriol 196:2876–2888

    Article  PubMed  PubMed Central  Google Scholar 

  42. Daly SM, Elmore BO, Kavanaugh JS, Triplett KD, Figueroa M, Raja HA, El-Elimat T, Crosby HA, Femling JK, Cech NB, Horswill AR, Oberlies NH, Hall PR (2015) ω-Hydroxyemodin limits staphylococcus aureus quorum sensing-mediated pathogenesis and inflammation. Antimicrob Agents Chemother 59:2223–2235

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Paul P, Chakraborty P, Sarker RK, Chatterjee A, Maiti D, Das A, Mandal S, Bhattacharjee S, Dastidar DG, Tribedi P (2021) Tryptophan interferes with the quorum sensing and cell surface hydrophobicity of Staphylococcus aureus: a promising approach to inhibit the biofilm development. 3 Biotech 11:376

    Article  PubMed  PubMed Central  Google Scholar 

  44. Nazar A, Abbas G, Azam SS (2020) Deciphering the inhibition mechanism of under trialHsp90 inhibitors and their analogues: a comparative molecular dynamics simulation. J Chem Inf Model 60:3812–3830

    Article  PubMed  Google Scholar 

  45. Pal S, Kumar V, Kundu B, Bhattacharya S, Preethy N, Reddy MP, Taulkdar A (2019) Ligand-based pharmacophore modelling, virtual screening and molecular docking studies for discovery of potential topoisomerase I inhibitors. Comput Struct Biotechnol J 17:291–310

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ (1997) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 23:3–25

    Article  CAS  Google Scholar 

  47. Shahbazi S, Kuanar A, Gade DR, Kar D, Shrivastava A, Kunala P, Mahto MK (2016) Semiemperical investigation of the postmenopausal breast cancer treatment potential of xanthone derivatives. Nat Prod Chem Res 4:206

  48. Arora R, Issar U, Kakkar R (2019) Identification of novel urease inhibitors: pharmacophore modeling, virtual screening and molecular docking studies. J Biomol Struct Dyn 37:4312–4326

    Article  CAS  PubMed  Google Scholar 

  49. Kumar A, Sahoo SK, Padhee K, Kochar P, Satapathy A, Pathak N (2011) Review on solubility enhancement techniques for hydrophobic drugs. Pharm Glob: Int J Compr Pharm 2:001–007

    Google Scholar 

Download references

Acknowledgements

The authors are grateful to the Centre for Machine Learning and Intelligence (CMLI) at Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, India, for their continuous rendering of and support during the work.

Funding

This work was supported by the DST-CURIE-AI-PhaseII (Project No. 16), Centre for Machine Learning and Intelligence (CMLI) at Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, India.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: Rajeswari Murugesan. Development of methodology and acquisition of data: Monica Ramasamy and Rajeswari Murugesan, Analysis; interpretation of data: Monica Ramasamy, Aishwarya Vetrivel, and Rajeswari Murugesan; writing, review, and/or revision of the manuscript: Monica Ramasamy, Aishwarya Vetrivel, Sharulatha Venugopal, and Rajeswari Murugesan.

Corresponding author

Correspondence to Rajeswari Murugesan.

Ethics declarations

Competing interests

The authors declare no competing interests.

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

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 0.97 MB)

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

Ramasamy, M., Vetrivel, A., Venugopal, S. et al. Identification of inhibitors for Agr quorum sensing system of Staphylococcus aureus by machine learning, pharmacophore modeling, and molecular dynamics approaches. J Mol Model 29, 258 (2023). https://doi.org/10.1007/s00894-023-05647-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00894-023-05647-9

Keywords

Navigation