Document Type : Original Research Article

Authors

1 Department of Chemistry, Federal University Lokoja, P.M.B., 1154, Lokoja, Kogi State, Nigeria

2 Department of Chemistry, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State Nigeria

Abstract

Typhoid fever, a disease caused by a Gram-negative bacterium known as Salmonella typhi constitutes a significant cause of morbidity and mortality, especially in developing nations. The rising cases of resistance to existing antibiotics by this bacterium have necessitated the search for the novel drug candidates. In this study, a data set of some anti-Salmonella typhi pyridine-substituted coumarins were subjected to Molecular Docking-based Virtual Screening against the active sites of DNA gyrase of the bacterium using EasyDock Vina 2.0 of AutoDock Vina software. Prior to the molecular docking calculation, the structures of the compounds were optimized using the DFT method of Spartan 14 software to obtain their minimum energy conformations. The outcome of the Virtual Screening led to the selection of compounds 12, 13, and 15 as template molecules for the design of more potent analogues because they bind better to the active sites of DNA gyrase target with binding affinity values (ΔG) of -9.6 kcal/mol, -9.5 kcal/mol and -9.6 kcal/mol, respectively. Subsequently, the template molecules were subjected to structural modifications leading to the design of more potent analogues with ΔG values ranging from -9.9 kcal/mol to -10.6 kcal/mol against DNA gyrase target. Furthermore, insilico drug-likeness and ADMET evaluation of the designed ligands revealed that they possess good oral bioavailability and positive pharmacokinetic profiles. It is hoped that the findings of this research would provide an excellent template for the development of novel drugs that could curb the alarming rate of resistance to existing antibiotics by Salmonella typhi.

Graphical Abstract

Molecular Docking Study and Insilico Design of Novel Drug Candidates against Salmonella typhi

Keywords

Main Subjects

  1. S. Fauci, L.D. Kasper, L.D. Longo, E. Braunwald, L.S. Hauser, L.J. Jameson , J. Loscalzo. Harrison´s Principles of Internal Medicine, Internal Medicine Journal, 38 (2008) 932-932.
  2. World Health Organization, Typhoid. Retrieved from http://www.who.int/news-room/fact-sheets/detail/typhoid, (2018); January 31)
  3. A. Ernanto, R.J. Palimbongan, R.A. Manufandu, S. Darmawati. Identification of Salmonella typhi contamination by amplification fliC gene in grass-jelly from traditional markets and minimarket in Semarang city, Jurnal Teknologi Laboratorium, 9 (2020) 136–144.
  4. A. Dewan, R. Corner, M. Hashizume, T.E. Ongee. Typhoid Fever and its association with environmental factors in the Dhaka Metropolitan Area of Bangladesh: a spatial and time-series approach, PLOS Neglected Tropical Disease, 7 (2013) e1998.
  5. M. Khan, L.R. Ochiai, B.S. Soofi, L. Von-Seidlein, J.M. Khan, M.S. Sahito, et al, Risk factors associated with typhoid fever in children aged 2–16 years in Karachi, Pakistan. Epidemiol Infect, 140 (2012) 665–72.
  6. E. Klemm, S. Shakoor, J.A. Page, N.F. Qamar, K. Judge, K.D. Saeed, et al, Emergence of an extensively drugresistant Salmonella enterica serovar Typhi clone harboring a promiscuous plasmid encoding resistance to fluoroquinolones and third-generation cephalosporins. mBio 9 (2018) e00105-18.
  7. W. Mutai, T.W.A. Muigai, P. Waiyaki, S. Kariuki, Multi-drug resistant Salmonella entericaserovar Typhi isolates with reduced susceptibility to ciprofloxacin in Kenya. BMC Microbiology, 18 (2018) 187
  8. Haider, A. Chauhan, S. Tariq, D.P. Pathak, N. Siddiqui, S. Ali, et al. Application of In silico Methods in the Design of Drugs for Neurodegenerative Diseases. Current Topics in Medicinal Chemistry. 21 (2021) 995–1011.
  9. N. Namitha, V Velmurugan. Review of bioinformatic tools used in Computer Aided Drug Design (CADD). World Journal of Advanced Research and Reviews, 14(2022) 453–465
  10. Ray, A.K. Panigrahi, S. Dutta, M. Gochayat. Computer aided drug design: tools to develop drug for covid 19. Asian Journal of Advances in Medical Science. 3 (2021) 127–146.
  11. T. Talele, A.S. Khedkar, C.A. Rigby, Successful Applications of Computer Aided Drug Discovery: Moving Drugs from Concept to the Clinic. Current Topics in Medicinal Chemistry, 10 (2010) 127-141
  12. P. Kore, M.M. Mutha, V.R. Antre, J.R. Oswal, S.S. Kshirsagar, Computer-Aided Drug Design: An Innovative Tool for Modeling. Open Journal of Medicinal Chemistry, 2 (2012) 139-148.
  13. S. Padole, J.A. Asnani, D.R. Chaple, G.S. Katre, A review of approaches in computer-aided drug design in drug discovery, GSC Biological and Pharmaceutical Sciences, 19 (2022) 075–083.
  14. Hussain, N. Rasool, Y.D. Khan. Insights into Machine Learning-based approaches for Virtual Screening in Drug Discovery: Existing strategies and streamlining through FP-CADD. Current Drug Discovery Technologies. 18 (2021) 463–72
  15. Behl, I. Kaur, A. Sehgal, S. Singh, S. Bhatia, A. Al-Harrasi , et al. Bioinformatics Accelerates the Major Tetrad: A Real Boost for the Pharmaceutical Industry. IJMS. 22 (2021) 1-28
  16. Huai, H. Yang, X. Li, Z. Sun, SAMPL7 TrimerTrip host–guest binding affinities from extensive alchemical and end-point free energy calculations. J Comput Aided Mol Des, 35 (2021) 117–129
  17. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, druglikeness and medicinal chemistry friendliness of small molecules. Scientific Reports, 7:42717,
  18. E.K. Daoud, P.K. Deb, M. Alzweiri, P. Borah, K.N. Venugopala, W. Hourani, et al. ADMET Profiling in Drug Discovery and Development: Perspectives of In Silico, In Vitro and Integrated Approaches. Current Drug Metabolism. 22 (2021).
  19. Levine, H. Hiasa, K.J. Marians, DNA gyrase and topoisomerase IV: Biochemical activities, physiological roles during chromosome replication, and drug sensitivities Biochim. Biophys. Acta, 1400 (1998) 29– 43
  20. J. Champoux, DNA topoisomerases: Structure, function, and mechanism Annu. Rev. Biochem., 70 (2001) 369–413
  21. Drlica, H. Hiasa, R. Kerns,M. Malik, A. Mustaev, X. Zhao. Quinolones: Action and resistance updated. Curr. Top. Med. Chem., 9 (2009) 981–998
  22. Drlica, M. Malik, R.J. Kerns, X. Zhao, Quinolone-mediated bacterial death Antimicrob. Agents Chemother., 52 (2008) 385–392
  23. Collin F, Karkare S, Maxwell A. Exploiting bacterial DNA gyrase as a drug target: current state and perspectives, Appl Microbiol Biotechnol., 92 (2011) 479–497.
  24. I. Anebi, M.T. Ibrahim, A.G. Shallangwa, S. Isyaku, S. Abdulsalam, M.A. Danmallam, Molecular docking study, drug-likeness and pharmacokinetic properties (ADMET) prediction of some novel thiophene derivatives as salmonella typhi inhibitors, Bayero Journal of Pure and Applied Sciences, 14 (2020).
  25. Kumar, S. Yadav, A. Singh, S. Kakkar, B. Narasimhan, A. Sharma, Benzoxazole Derivatives: QSAR and Molecular Docking Studies. Preprint. 2022.
  26. A. Coba-Males, J. Santamaría-Aguirre, C.D. Alcívar-León, In Silico Evaluation of New Fluoroquinolones as Possible Inhibitors of Bacterial Gyrases in Resistant Gram-Negative Pathogens. Chem. Proc., 8 (2022) 43.
  27. S. Elseginy, M.M. Anwar MM, Pharmacophore-Based Virtual Screening and Molecular Dynamics Simulation for Identification of a Novel DNA Gyrase B Inhibitor with Benzoxazine Acetamide Scaffold. ACS Omega, 7 (2022) 1150−1164.
  28. H. Lad, R.R. Giri, L.Y. Chovatiya, I.D. Brahmbhatt, Synthesis of modified pyridine and bipyridine substituted coumarins as potent antimicrobial agents. J. Serb. Chem. Soc., 80 (2015), 739–747.
  29. Trott, A.J. Olson, AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading, Journal of Computational Chemistry, 31 (2010) 455-61.
  30. Ejeh, A. Uzairu, G.A. Shallangwa, S.E. Abechi, M.T. Ibrahim In silico design and pharmacokinetics investigation of some novel hepatitis C virus NS5B inhibitors: pharmacoinformatics approach, Bulletin of the National Research Centre 46 (2022) 109
  31. E. Adeniji, A. Ajala, D.E. Arthur, M. Abdullahi, O.I. Areguamen, Chemometric Study, Homology Modeling of G Protein-Coupled Bile Acids Receptor (GPBAR_HUMAN) of Type-2 Diabetes Mellitus, Virtual Screening Evaluation, Drug-Likeness and ADME Prediction for Newly Designed Compounds, Macromolecular research, (2022) 1-19.
  32. Perumal, V.P. Pandey, P. Parasuraman, Docking studies on some novel piperidine analogues against DNA gyrase enzyme. Inventi Rapid Mol Model, 1 (2014) 1–4
  33. Maxwell, D.M. Lawson, The ATP binding site of type II topoisomerases as a target for antibacterial drugs. Curr Top Med Chem., 3 (2003) 283–303
  34. F. Verber, S.R. Johnson, H.Y. Cheng, B.R. Smith, K.W. Ward, K.D. Kopple, Molecular properties that influence the oral bioavailability of drug candidates. J. med.chem., 45 (2002) 2615-2623.
  35. Hossain, B. Sarkar, M.N.I. Prottoy, Y. Araf, M.A. Taniya, M.A. Ullah, Thrombolytic activity, drug likeness property and ADME/T analysis of isolated phytochemicals from ginger (Zingiber officinale) using in silico approaches. Mod. Res. Inflamm., 8 (2019) 29–43.
  36. Yu, A. Adedoyin, ADME–Tox in drug discovery: Integration of experimental and computational technologies. Drug Discov. Today, 8 (2003) 852–861.
  37. Ziraldo, A. Hanke, S.D. Levene, Kinetic pathways of topology simplification by Type-II topoisomerases in knotted supercoiled DNA. Nucleic Acids Research, 47 (2019) 69–84
  38. C. Dorman, J.M. Dorman, DNA supercoiling is a fundamental regulatory principle in the control of bacterial gene expression. Biophysical Reviews, 8 (2016) 209–220.
  39. A. Schoeffler, M.J. Berger, DNA topoisomerases: harnessing and constraining energy to govern chromosome topology. Quarterly Reviews of Biophysics, 41 (2008) 41–101
  40. Jakopin, J. Ilas, M. Barancokova, et al., Discovery of substituted oxadiazoles as a novel scaffold for DNA gyrase inhibitors, European Journal of Medicinal Chemistry, 130 (2017) 171–184.
  41. Tian, J. Wang, Y. Li, D. Li, L. Xu, T. Hou, The application of in silico drug-likeness predictions in pharmaceutical research. Adv. Drug Deliv. Rev. 86 (2015) 1–10.
  42. Wang, J. Xing, Y. Xu, N. Zhou, J. Peng, Z. Xiong, X. Liu, X. Luo, C. Luo, K. Chen, In silico ADME/T modelling for rational drug design. Q. Rev. Biophys., 48 (2015) 488–515
  43. Anzenbacher, E. Anzenbacherova, Cytochromes P450 and metabolism of xenobiotics, Cell. Mol. Life Sci. CMLS, 58 (2001) 737–747.
  44. De Graaf, N.P. Vermeulen, K.A. Feenstra, Cytochrome P450 in silico: An integrative modeling approach. J. Med. Chem., 48 (2005) 2725–2755
  45. C. Lamb, M.R. Waterman, S.L. Kelly, F.P. Guengerich, Cytochromes P450 and drug discovery. Curr. Opin. Biotechnol., 18 (2007) 504–51