Evaluation of novel Anti-SARS-CoV-2 compounds by targeting nucleoprotein and envelope protein through homology modeling, docking simulations, ADMET, and molecular dynamic simulations with the MM/GBSA calculation

Emmanuel Israel Edache , Adamu Uzairu , Paul Andrew Mamza , Gideon Adamu Shallangwa , Muhammad Tukur Ibrahim

Intelligent Pharmacy ›› 2024, Vol. 2 ›› Issue (3) : 346 -366.

PDF (7666KB)
Intelligent Pharmacy ›› 2024, Vol. 2 ›› Issue (3) : 346 -366. DOI: 10.1016/j.ipha.2024.02.008
Full length article

Evaluation of novel Anti-SARS-CoV-2 compounds by targeting nucleoprotein and envelope protein through homology modeling, docking simulations, ADMET, and molecular dynamic simulations with the MM/GBSA calculation

Author information +
History +
PDF (7666KB)

Abstract

The current prominent virus that induces severe acute respiratory syndrome is SARS-CoV-2. The incidence of COVID-19 cases is increasing, necessitating the immediate development of effective treatments. Our objective was to employ an in-silico approach to evaluate the effectiveness of conventional compounds against COVID-19’s nucleoprotein and envelope protein. A docking simulation was performed on 9 compounds as SARS-coronavirus inhibitors using AMDock software. Anti-covid-19 activities were further evaluated for the compounds. Based on docking results, the binding affinity of “N-(4-carbamoylphenyl)-8-cyclopropyl-7-(naphthalen-1-ylmethyl)-5-oxo-2,3- dihydro-5H-thiazolo[3,2-a]pyridine-3-carboxamide,” also called compound 36 in this research, was found to be -8.8 kcal/mol for the modeled envelope protein and -7.3 kcal/mol for the template envelope protein, while -10.1 kcal/mol for the modeled nucleocapsid proteins (NP) and -8.7 kcal/mol for the template nucleocapsid proteins (NP) of SARS-coronavirus, respectively. The ligand and control drug (ritonavir) with high docking scores were subjected to pharmacological screening, molecular dynamic simulations, and Molecular Mechanics-generalized Born Surface Area (MM/GBSA) calculations. Furthermore, the jobs of pharmacokinetics were assessed, and the outcomes acquired show that the proposed compound 36 includes great oral bioavailability and a capacity to diffuse through various organic boundaries. The protein-ligand complexes were subjected to dynamic simulation analyses with a re-enactment time of 10 ns, likewise, their free binding energy was inspected operating the MM/GBSA approach. The docking (MD simulation) results acquired emphasize the pivotal residues answerable for the proteinligand interaction, giving an understanding of the method of association. The MD simulation analysis verifies the structural stability of the selected complexes during the MD trajectory, with minor changes detected. The MM/GBSA data show that compound 36 has the lowest free energy of -12.498 kcal/mol for EP and -57.5185 kcal/mol for NP proteins of SARS-coronavirus, confirming the molecular docking result. As a result, the identified chemical can be used to develop a new family of antiviral medications against SARS-coronavirus-2.

Keywords

SARS-Coronavirus / Comparative modeling / Docking / Molecular dynamics simulation / And MM/GBSA calculation

Cite this article

Download citation ▾
Emmanuel Israel Edache, Adamu Uzairu, Paul Andrew Mamza, Gideon Adamu Shallangwa, Muhammad Tukur Ibrahim. Evaluation of novel Anti-SARS-CoV-2 compounds by targeting nucleoprotein and envelope protein through homology modeling, docking simulations, ADMET, and molecular dynamic simulations with the MM/GBSA calculation. Intelligent Pharmacy, 2024, 2(3): 346-366 DOI:10.1016/j.ipha.2024.02.008

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Bitam S, Hamadache M, Hanini S. 2D-QSAR, docking, molecular dynamics, studies of PF-07321332 analogues to identify alternative inhibitors against 3CLpro enzyme in SARS-CoV disease. J Biomol Struct Dyn.2022. https://doi.org/10.1080/07391102.2022.2113822.

[2]

Luo H, Tang Q-L. Shang Y-X, et al. Can Chinese medicine Be used for prevention of corona virus disease 2019 (COVID-19)? A review of historical classics, research evidence and current prevention programs. Chin J Integr Med. 2020;26:243–250.

[3]

Saddala MS, Adi PKJ, Rani UA. In silico drug design and molecular docking studies of potent inhibitors against cathepsin-L (Ctsl) for sars disease. J. Res. Development. 2016;4:2. https://doi.org/10.4172/2311-3278.1000145.

[4]

Obaidullah AJ, Alanazi MM, Alsaif NA, et al. Immunoinformatics-guided design of a multiepitope vaccine based on the structural proteins of severe acute respiratory syndrome coronavirus. RSC Adv. 2021;11:18103.

[5]

Edache EI, Uzairu A, Mamza PA, Shallangwa GA. A 2D-QSAR, homology modeling, docking, ADMET, and molecular dynamics simulations studies for assessment of a novel SARS-Cov-2 and Pseudomonas aeruginosa inhibitors. J. Virol. Viral. Dis. 2022;2.

[6]

Mou K, Abdalla M, Wei DQ, et al. Emerging mutations in the envelope protein of SARS-CoV-2 and their effect on thermodynamic properties. Inform Med Unlocked. 2021;26:100675.

[7]

Mahmud S, Paul GK, Biswas S, et al. Prospective role of peptide-based antiviral therapy against the main protease of SARS-CoV-2. Front Mol Biosci. 2021;8:628585.

[8]

Tariqa A, Mateen RM, Afzal MS, Saleem M. Paromomycin: a potential dual-targeted drug effectively inhibits both spike (S1) and main protease of COVID-19. Int J Infect Dis. 2020;98:166–175.

[9]

Velavan TP, Meyer CG. The COVID-19 epidemic. Trop Med Int Health. 2020;25:278–280.

[10]

Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273. https://doi.org/10.1038/S41586-020-2012-7.

[11]

V’Kovski P, Kratzel A, Steiner S, Stalder H, Thiel V. Coronavirus biology and replication: implications for SARS-CoV-2. Nat Rev Microbiol. 2021;19:155–170.

[12]

Satarker S, Nampoothiri M. Structural proteins in severe acute respiratory syndrome coronavirus-2. Arch Med Res. 2020;51:482–491.

[13]

McBride R, van Zyl M, Fielding BC. The coronavirus nucleocapsid is a multifunctional protein. Viruses. 2014;6:2991–3018.

[14]

Schoeman D, Fielding BC. Is there a link between the pathogenic human coronavirus envelope protein and immunopathology? A review of the literature. Front Microbiol. 2020;11:2086.

[15]

Shepley-McTaggart A, Sagum CA, Oliva I, et al. SARS-CoV-2 Envelope (E) protein interacts with PDZ-domain-2 of host tight junction protein ZO1. PLoS One. 2021;16: e0251955.

[16]

Ghosh S, Keretsu S, Cho SJ. Molecular modeling studies of N-phenylpyrimidine-4-amine derivatives for inhibiting FMS-like tyrosine kinase-3. Int J Mol Sci. 2021;22:12511.

[17]

Edache EI, Uzairu A, Mamza PA, Shallangwa GA. Theoretical investigation of the cooperation of iminoguanidine with the enzymes-binding domain of covid-19 and bacterial lysozyme inhibitors and their pharmacokinetic properties. J. Mex. Chem. Soc. 2022;66:513–542.

[18]

Zerroug A, Belaidi S, BenBrahim I, Sinha L, Chtita S. Virtual screening in druglikeness and structure/activity relationship of pyridazine derivatives as Anti-Alzheimer drugs. J King Saud Univ Sci. 2019;31:595–601.

[19]

Papaj K, Spychalska P, Hopko K, et al. Investigation of thiocarbamates as potential inhibitors of the SARS-CoV-2 mpro. Pharmaceuticals. 2021;14:1153.

[20]

Edache EI, Uzairu A, Mamza PA, Shallangwa GA. Investigation of salicylidene acylhydrazides derivatives: molecular Docking, ADMET, and Molecular Dynamic Simulations were used in conjunction towards the design of new Yersinia pseudotuberculosis inhibitors. Turkish Comp Theo Chem. 2022;6:9–31.

[21]

Eid RA, Elgendy MO, El-Gendy AO, et al. Efficacy of ceftazidime and cefepime in the management of COVID-19 patients: single center report from Egypt. Antibiotics. 2021;10:1278.

[22]

Uhomoibhi JO, Idowu KA, Shode FO, Sabiu S. Molecular modeling identification of potential drug candidates from selected African plants against SARS-CoV-2 key druggable proteins. Sci. Afr. 2022;17:e01279.

[23]

Khamouli S, Belaidi S, Almi Z, Medjahed S, Belaidi HJ. Property/activity relationships and drug likeness for pyrimidine derivatives as serine/threonine protein kinase B inhibitors. Bionanosci. 2017;11(4):301–309.

[24]

Sanner MF. Python: a programming language for software integration and development. J Mol Graph Model. 1999;17:57–61.

[25]

Waterhouse AM, Procter JB, Martin DMA, Clamp M, Barton GJ. Jalview Version 2 -a multiple sequence alignment editor and analysis workbench. Bioinformatics. 2009;25:1189–1191.

[26]

Henikoff S, Henikoff JG. Amino acid substitution matrices from protein blocks. Proc Natl Acad Sci USA. 1992;89:10915–10919.

[27]

Webb B, Sali A. Comparative protein structure modeling using MODELLER. Curr. Protoc. Bioinforma. 2016;54. 5.6.1–5.6.37.

[28]

Ye WL, Zhang LX, Guan YD, et al. Virtual screening and experimental validation of eEF2K inhibitors by combining homology modeling, QSAR, and molecular docking from FDA-approved drugs. New J Chem. 2019;43:19097–19106.

[29]

Daddam JR, Dowlathabad MR, Panthangi S, Jasti P. Molecular docking and P-glycoprotein inhibitory activity of flavonoids. Interdiscipl Sci Comput Life Sci. 2014;6:167–175.

[30]

Sippl MJ. Recognition of errors in three-dimensional structures of proteins. Proteins: Struct, Funct, Bioinf. 1993;17:355–362.

[31]

Khattak F, Haseeb M, Fazal S. Molecular homology modeling & docking studies to predict the 3D structure and drug determination for HBV core of Hepatitis B. J Chem Pharmaceut Res. 2015;7:1136–1146.

[32]

Suleman M, Yousafi Q, Ali J, et al. Bioinformatics analysis of the differences in the binding profile of the wild-type and mutants of the SARS-CoV-2 spike protein variants with the ACe2 receptor. Comput Biol Med. 2021;138:104936.

[33]

Edache EI, Uzairu A, Mamza PA, Shallangwa GA. QSAR, homology modeling, and docking simulation on SARS-CoV-2 and pseudomonas aeruginosa inhibitors, ADMET, and molecular dynamic simulations to find a possible oral lead candidate. J Genet Eng Biotechnol. 2022;20:88.

[34]

Valdés-Tresanco MS, Valdés-Tresanco ME, Valiente PA, Moreno E, Amdock. A versatile graphical tool for assisting molecular docking with Autodock Vina and Autodock4. Biol Direct. 2020;15:12.

[35]

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;31:455–461.

[36]

Harris R, Olson A, Goodsell D. Automated prediction of ligand-binding sites in proteins. Proteins. 2008;70:1505–1517.

[37]

Feinstein WP, Brylinski M. Calculating an optimal box size for ligand docking and virtual screening against experimental and predicted binding pockets. J Cheminf. 2015;7:18.

[38]

Ugbe FA, Shallangwa GA, Uzairu A, Abdulkadir I. Activity modeling, molecular docking, and pharmacokinetic studies of some boron-pleuromutilins as antiwolbachia agents with potential for the treatment of filarial diseases. Chem. Data Collect. 2021;36:100783.

[39]

Edache EI, Uzairu A, Shallangwa GA, Mamza PA. Virtual screening, pharmacokinetics, and molecular dynamics simulations studies to identify potent approved drugs for Chlamydia trachomatis treatment. Futur. J. Pharm. Sci. 2021;7:220.

[40]

Aziz M, Ejaz SA, Tamam N, et al. Identification of potent inhibitors of NEK7 protein using a comprehensive computational approach. Sci Rep. 2022;12:1–17.

[41]

Kar S, Roy K, Leszczynski J. Silico tools and software to predict ADMET of new drug candidates. In: silico methods for predicting drug toxicity. Methods Mol Biol. 2022;2425:85–115.

[42]

Phillips JC, H DJ, Maia JDC, et al. Scalable molecular dynamics on CPU and GPU architectures with NAMD. J Chem Phys. 2020;153:044130, 044110.041063/044135.0014475.

[43]

Humphrey W, Dalke A, Schulten K. VMD: visual molecular dynamics. J Mol Graph. 1996;14:33–38.

[44]

Wang E, Sun H, Wang J, et al. End-point binding free energy calculation with MM/PBSA and MM/GBSA: strategies and applications in drug design. Chem Rev. 2019;119:9478–9508.

[45]

Bai Q, Tan S, Xu T, Liu H, Huang J, Yao X. MolAICal: a soft tool for 3D drug design of protein targets by artificial intelligence and classical algorithm. Briefings Bioinf. 2021;22.

[46]

Polanski J, Duszkiewicz R, Pedrys A, Gasteiger J. Scoring ligand efficiency. Acta Pol Pharm. 2019;76:761–768.

[47]

Loving K, Alberts I, Sherman W. Computational approaches for fragment-based and de novo design. Curr Top Med Chem. 2012;10:14–32.

[48]

Dong J, Wang NN, Yao ZJ, et al. ADMETlab: a platform for systematic ADMET evaluation based on a comprehensively collected ADMET database. J Cheminf. 2018;10:29.

[49]

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. https://doi.org/10.1016/S0169-409x(00)00129-0.

[50]

Hughes JD, Blagg J, Price DA, et al. Physiochemical drug properties associated with in vivo toxicological outcomes. Bioorg Med Chem Lett. 2008;18:4872–4875.

[51]

Gleeson MP. Generation of a set of simple, interpretable ADMET rules of thumb. J Med Chem. 2008;51:817–834.

[52]

Mohamed EAR, Abdelwahab SF, Alqaisi AM, Nasr AMS, Hassan HA. Identification of promising anti-EBOV inhibitors: de novo drug design, molecular docking, and molecular dynamics studies. R Soc Open Sci. 2022;9:220369.

RIGHTS & PERMISSIONS

2024 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd.

AI Summary AI Mindmap
PDF (7666KB)

Supplementary files

Supporting Information

425

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/