DOI QR코드

DOI QR Code

Computer-aided drug design of Azadirachta indica compounds against nervous necrosis virus by targeting grouper heat shock cognate protein 70 (GHSC70): quantum mechanics calculations and molecular dynamic simulation approaches

  • Islam, Sk Injamamul (Department of Fisheries and Marine Bioscience, Faculty of Biological Science, Jashore University of Science and Technology) ;
  • Saloa, Saloa (Department of Environmental Science and Technology, Faculty of Applied Science and Technology, Jashore University of Science and Technology) ;
  • Mahfuj, Sarower (Department of Fisheries and Marine Bioscience, Faculty of Biological Science, Jashore University of Science and Technology) ;
  • Islam, Md Jakiul (Faculty of Fisheries, Sylhet Agricultural University) ;
  • Jahan Mou, Moslema (Department of Genetic Engineering and Biotechnology, Faculty of Life and Earth Science, University of Rajshahi)
  • 투고 : 2021.10.28
  • 심사 : 2022.08.17
  • 발행 : 2022.09.30

초록

Nervous necrosis virus (NNV) is a deadly infectious disease that affects several fish species. It has been found that the NNV utilizes grouper heat shock cognate protein 70 (GHSC70) to enter the host cell. Thus, blocking the virus entry by targeting the responsible protein can protect the fishes from disease. The main objective of the study was to evaluate the inhibitory potentiality of 70 compounds of Azadirachta indica (Neem plant) which has been reported to show potential antiviral activity against various pathogens, but activity against the NNV has not yet been reported. The binding affinity of 70 compounds was calculated against the GHSC70 with the docking and molecular dynamics (MD) simulation approaches. Both the docking and MD methods predict 4 (PubChem CID: 14492795, 10134, 5280863, and 11119228) inhibitory compounds that bind strongly with the GHSC70 protein with a binding affinity of -9.7, -9.5, -9.1, and -9.0 kcal/mol, respectively. Also, the ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties of the compounds confirmed the drug-likeness properties. As a result of the investigation, it may be inferred that Neem plant compounds may act as significant inhibitors of viral entry into the host cell. More in-vitro testing is needed to establish their effectiveness.

키워드

과제정보

The author thanks Dr. Foysal Ahmed Sagore and Dr. Kazi Abdus Samad for helpful comments.

참고문헌

  1. Bandin I, Souto S. Betanodavirus and VER disease: a 30-year research review. Pathogens 2020;9:106. https://doi.org/10.3390/pathogens9020106
  2. Chi SC, Shieh JR, Lin SJ. Genetic and antigenic analysis of betanodaviruses isolated from aquatic organisms in Taiwan. Dis Aquat Organ 2003;55:221-228. https://doi.org/10.3354/dao055221
  3. Munday BL, Kwang J, Moody N. Betanodavirus infections of teleost fish: a review. J Fish Dis 2002;25:127-142. https://doi.org/10.1046/j.1365-2761.2002.00350.x
  4. Chi SC, Lo CF, Kou GH, Chang PS, Peng SE, Chen SN. Mass mortalities associated with viral nervous necrosis (VNN) disease in two species of hatchery-reared grouper, Epinephelus fuscogutatus and Epinephelus akaara (Temminck & Schlegel). J Fish Dis 1997;20:185-193. https://doi.org/10.1046/j.1365-2761.1997.00291.x
  5. Yoshikoshi K, Inoue K. Viral nervous necrosis in hatchery-reared larvae and juveniles of Japanese parrotfish, Oplegnathus fasciatus (Temminck & Schlegel). J Fish Dis 1990;13:69-77. https://doi.org/10.1111/j.1365-2761.1990.tb00758.x
  6. Kim JO, Kim WS, Cho JK, Kim KM, Son MH, Oh MJ. Complete genome sequence of nervous necrosis virus isolated from sevenband grouper (Epinephelus septemfasciatus) in South Korea. Genome Announc 2014;2:e01264-01214.
  7. Mori K, Nakai T, Muroga K, Arimoto M, Mushiake K, Furusawa I. Properties of a new virus belonging to nodaviridae found in larval striped jack (Pseudocaranx dentex) with nervous necrosis. Virology 1992;187:368-371. https://doi.org/10.1016/0042-6822(92)90329-N
  8. Sommerset I, Nerland AH. Complete sequence of RNA1 and subgenomic RNA3 of Atlantic halibut nodavirus (AHNV). Dis Aquat Organ 2004;58:117-125. https://doi.org/10.3354/dao058117
  9. Chen LJ, Su YC, Hong JR. Betanodavirus non-structural protein B1: A novel anti-necrotic death factor that modulates cell death in early replication cycle in fish cells. Virology 2009;385:444-454. https://doi.org/10.1016/j.virol.2008.11.048
  10. Fenner BJ, Thiagarajan R, Chua HK, Kwang J. Betanodavirus B2 is an RNA interference antagonist that facilitates intracellular viral RNA accumulation. J Virol 2006;80:85-94. https://doi.org/10.1128/JVI.80.1.85-94.2006
  11. Iwamoto T, Okinaka Y, Mise K, Mori K, Arimoto M, Okuno T, et al. Identification of host-specificity determinants in betanodaviruses by using reassortants between striped jack nervous necrosis virus and sevenband grouper nervous necrosis virus. J Virol 2004;78:1256-1262. https://doi.org/10.1128/JVI.78.3.1256-1262.2004
  12. Shieh JR, Chi SC. Production of monoclonal antibodies against grouper nervous necrosis virus (GNNV) and development of an antigen capture ELISA. Dis Aquat Organ 2005;63:53-60. https://doi.org/10.3354/dao063053
  13. Liu W, Hsu CH, Hong YR, Wu SC, Wang CH, Wu YM, et al. Early endocytosis pathways in SSN-1 cells infected by dragon grouper nervous necrosis virus. J Gen Virol 2005;86:2553-2561. https://doi.org/10.1099/vir.0.81021-0
  14. Chang JS, Chi SC. GHSC70 is involved in the cellular entry of nervous necrosis virus. J Virol 2015;89:61-70. https://doi.org/10.1128/JVI.02523-14
  15. Jalil A, Ashfaq UA, Shahzadi S, Rasul I, Rehman SU, Shah M, et al. Screening and design of anti-diabetic compounds sourced from the leaves of neem (Azadirachta indica). Bioinformation 2013;9:1031-1035. https://doi.org/10.6026/97320630091031
  16. Srivastava AK, Maurya R. Antihyperglycemic activity of compounds isolated from Indian medicinal plants. Indian J Exp Biol 2010;48:294-298.
  17. Subapriya R, Nagini S. Medicinal properties of neem leaves: a review. Curr Med Chem Anticancer Agents 2005;5:149-146. https://doi.org/10.2174/1568011053174828
  18. Lim SM, Xie T, Westover KD, Ficarro SB, Tae HS, Gurbani D, et al. Development of small molecules targeting the pseudokinase Her3. Bioorg Med Chem Lett 2015;25:3382-3389. https://doi.org/10.1016/j.bmcl.2015.04.103
  19. Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early drug discovery. Br J Pharmacol 2011;162:1239-1249. https://doi.org/10.1111/j.1476-5381.2010.01127.x
  20. Szymanski P, Markowicz M, Mikiciuk-Olasik E. Adaptation of high-throughput screening in drug discovery-toxicological screening tests. Int J Mol Sci 2012;13:427-452.
  21. Liang PH, Cheng WC, Lee YL, Yu HP, Wu YT, Lin YL, et al. Novel five-membered iminocyclitol derivatives as selective and potent glycosidase inhibitors: new structures for antivirals and osteoarthritis. Chembiochem 2006;7:165-173. https://doi.org/10.1002/cbic.200500321
  22. Wichapong K, Nueangaudom A, Pianwanit S, Sippl W, Kokpol S. Identification of potential hit compounds for Dengue virus NS2B/NS3 protease inhibitors by combining virtual screening and binding free energy calculations. Trop Biomed 2013;30:388-408.
  23. Wu J, Hu B, Sun X, Wang H, Huang Y, Zhang Y, Liu M, et al. In silico study reveals existing drugs as α-glucosidase inhibitors: structure-based virtual screening validated by experimental investigation. J Mol Struct 2020;1218:128532. https://doi.org/10.1016/j.molstruc.2020.128532
  24. Wu C, Liu Y, Yang Y, Zhang P, Zhong W, Wang Y, et al. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm Sin B 2020;10: 766-788. https://doi.org/10.1016/j.apsb.2020.02.008
  25. Combet C, Blanchet C, Geourjon C, Deleage G. NPS@: network protein sequence analysis. Trends Biochem Sci 2000;25:147-150. https://doi.org/10.1016/S0968-0004(99)01540-6
  26. Xu J, McPartlon M, Li J. Improved protein structure prediction by deep learning irrespective of co-evolution information. Nat Mach Intell 2021;3:601-609. https://doi.org/10.1038/s42256-021-00348-5
  27. Heo L, Park H, Seok C. GalaxyRefine: protein structure refinement driven by side-chain repacking. Nucleic Acids Res 2013;41:W384-W388. https://doi.org/10.1093/nar/gkt458
  28. Wiederstein M, Sippl MJ. ProSA-web: interactive web service for the recognition of errors in three-dimensional structures of proteins. Nucleic Acids Res 2007;35:W407-W410. https://doi.org/10.1093/nar/gkm290
  29. Ramachandran GN, Ramakrishnan C, Sasisekharan V. Stereochemistry of polypeptide chain configurations. J Mol Biol 1963;7:95-99. https://doi.org/10.1016/S0022-2836(63)80023-6
  30. Morris GM, Huey R, Lindstrom W, Sanner MF, Belew RK, Goodsell DS, et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J Comput Chem 2009;30:2785-2791. https://doi.org/10.1002/jcc.21256
  31. Mohanraj K, Karthikeyan BS, Vivek-Ananth RP, Chand RP, Aparna SR, Mangalapandi P, et al. IMPPAT: a curated database of Indian medicinal plants, phytochemistry and therapeutics. Sci Rep 2018;8:4329. https://doi.org/10.1038/s41598-018-22631-z
  32. Dallakyan S, Olson AJ. Small-molecule library screening by docking with PyRx. Methods Mol Biol 2015;1263:243-250. https://doi.org/10.1007/978-1-4939-2269-7_19
  33. Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, et al. Current trends in drug metabolism and pharmacokinetics. Acta Pharm Sin B 2019;9:1113-1144. https://doi.org/10.1016/j.apsb.2019.10.001
  34. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep 2017;7:42717. https://doi.org/10.1038/srep42717
  35. Li C, Wang J, Wang Y, Gao H, Wei G, Huang Y, et al. Recent progress in drug delivery. Acta Pharm Sin B 2019;9:1145-1162. https://doi.org/10.1016/j.apsb.2019.08.003
  36. Banerjee P, Eckert AO, Schrey AK, Preissner R. ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Res 2018;46:W257-W263. https://doi.org/10.1093/nar/gky318
  37. Lu Z, Zhang Y. Interfacing ab initio quantum mechanical method with classical drude osillator polarizable model for molecular dynamics simulation of chemical reactions. J Chem Theory Comput 2008;4:1237-1248. https://doi.org/10.1021/ct800116e
  38. Maity A, Samanta S, Biswas D, Chakravorty D. Studies on nanoconfinement effect of NiO-SiO2 spin glass within mesoporous Al2O3 template. J Alloys Compd 2021;887:161447. https://doi.org/10.1016/j.jallcom.2021.161447
  39. Hanwell MD, Curtis DE, Lonie DC, Vandermeersch T, Zurek E, Hutchison GR. Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. J Cheminform 2012;4:17. https://doi.org/10.1186/1758-2946-4-17
  40. Li Y, Evans JN. The Fukui function: a key concept linking frontier molecular orbital theory and the hard-soft-acid-base principle. J Am Chem Soc 1995;117:7756-7759. https://doi.org/10.1021/ja00134a021
  41. Opo F, Rahman MM, Ahammad F, Ahmed I, Bhuiyan MA, Asiri AM. Structure based pharmacophore modeling, virtual screening, molecular docking and ADMET approaches for identification of natural anti-cancer agents targeting XIAP protein. Sci Rep 2021;11:4049. https://doi.org/10.1038/s41598-021-83626-x
  42. Bharadwaj S, Dubey A, Yadava U, Mishra SK, Kang SG, Dwivedi VD. Exploration of natural compounds with anti-SARS-CoV-2 activity via inhibition of SARS-CoV-2 Mpro. Brief Bioinform 2021;22:1361-1377. https://doi.org/10.1093/bib/bbaa382
  43. Ahammad F, Alam R, Mahmud R, Akhter S, Talukder EK, Tonmoy AM, et al. Pharmacoinformatics and molecular dynamics simulation-based phytochemical screening of neem plant (Azadiractha indica) against human cancer by targeting MCM7 protein. Brief Bioinform 2021;22:bbab098. https://doi.org/10.1093/bib/bbab098
  44. Doan QK, Vandeputte M, Chatain B, Morin T, Allal F. Viral encephalopathy and retinopathy in aquaculture: a review. J Fish Dis 2017;40:717-742. https://doi.org/10.1111/jfd.12541
  45. Zorriehzahra MJ. Chapter 30: viral nervous necrosis disease. In: Emerging and Reemerging Viral Pathogens (Ennaji MM, ed.). London: Academic Press, 2020. pp. 673-703.
  46. Gomez-Casado E, Estepa A, Coll JM. A comparative review on European-farmed finfish RNA viruses and their vaccines. Vaccine 2011;29:2657-2671. https://doi.org/10.1016/j.vaccine.2011.01.097
  47. Terada K, Ueda I, Ohtsuka K, Oda T, Ichiyama A, Mori M. The requirement of heat shock cognate 70 protein for mitochondrial import varies among precursor proteins and depends on precursor length. Mol Cell Biol 1996;16:6103-6109. https://doi.org/10.1128/MCB.16.11.6103
  48. Ahn SG, Kim SA, Yoon JH, Vacratsis P. Heat-shock cognate 70 is required for the activation of heat-shock factor 1 in mammalian cells. Biochem J 2005;392:145-152. https://doi.org/10.1042/BJ20050412
  49. Almas K. The antimicrobial effects of extracts of Azadirachta indica (Neem) and Salvadora persica (Arak) chewing sticks. Indian J Dent Res 1999;10:23-26.
  50. Alzohairy MA. Therapeutics role of Azadirachta indica (Neem) and their active constituents in diseases prevention and treatment. Evid Based Complement Alternat Med 2016;2016: 7382506. https://doi.org/10.1155/2016/7382506
  51. Dutta A, Kundabala M. Antimicrobial efficacy of endodontic irrigants from Azadirachta indica: an in vitro study. Acta Odontol Scand 2013;71:1594-1598. https://doi.org/10.3109/00016357.2013.780290
  52. Chakraborty SB, Horn P, Hancz C. Application of phytochemicals as growth-promoters and endocrine modulators in fish culture. Rev Aquac 2014;6:1-19. https://doi.org/10.1111/raq.12021
  53. Sastry GM, Adzhigirey M, Day T, Annabhimoju R, Sherman W. Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichments. J Comput Aided Mol Des 2013;27:221-234. https://doi.org/10.1007/s10822-013-9644-8
  54. Lipinski CA. Lead- and drug-like compounds: the rule-of-five revolution. Drug Discov Today Technol 2004;1:337-341. https://doi.org/10.1016/j.ddtec.2004.11.007
  55. Pollastri MP. Overview on the rule of five. Curr Protoc Pharmacol 2010;Chapter 9:Unit 9.12.
  56. Aljahdali MO, Molla MH, Ahammad F. Compounds identified from marine mangrove plant (Avicennia alba) as potential antiviral drug candidates against WDSV, an in-silico approach. Mar Drugs 2021;19:253. https://doi.org/10.3390/md19050253
  57. Krupanidhi S, Abraham Peele K, Venkateswarulu TC, Ayyagari VS, Nazneen Bobby M, John Babu D, et al. Screening of phytochemical compounds of Tinospora cordifolia for their inhibitory activity on SARS-CoV-2: an in silico study. J Biomol Struct Dyn 2021;39:5799-5803. https://doi.org/10.1080/07391102.2020.1787226