| Peer-Reviewed

Molecular Docking and Molecular Dynamic Simulation of Potential Inhibitors of Integrase from Human Immunodeficiency Virus 1 (HIV-1) Using Phytochemicals

Received: 30 May 2022    Accepted: 22 June 2022    Published: 30 June 2022
Views:       Downloads:
Abstract

Background: In tremendously effective antiretroviral therapy for Human Immunodeficiency Virus 1 (HIV1) infections, integrase inhibitors are essential drugs. Resistance resulting from mutations, on the other hand, poses a threat to the medication’s long-term efficacy in HIV-1 infected people. Purpose: The current study utilized in silico techniques, we searched for phytochemicals or compounds that can inhibit the activity of the integrase enzyme. Material & Methods: Compounds were collected from databases, and potential candidates were screened using pharmacokinetics and structure-based virtual screening methodologies. The compounds were docked, and the binding affinity was evaluated to set the cut-off value for selecting compounds. When compared to standard drugs, some compounds had a higher binding affinity. Molecular dynamics simulation was then used to gain insight into the stability of the complexes, revealing two lead compounds, Withaferin and Isatin, indicating that these compounds have potency for drug development. These compounds were further investigated for their toxicity, indicating that Isatin was safe among the two. Conclusion: Thus, the study showcased that Isatin is a suitable drug candidate, and we hope that the findings of this study will be useful in the development of an antiviral drug against integrase enzyme.

Published in Computational Biology and Bioinformatics (Volume 10, Issue 1)
DOI 10.11648/j.cbb.20221001.16
Page(s) 34-48
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Human Immunodeficiency Virus 1 (HIV1), Integrase, Molecular Docking, Molecular Dynamic Simulation

References
[1] P. M. Sharp and B. H. Hahn, “Origins of HIV and the AIDS Pandemic,” Cold Spring Harb. Perspect. Med., vol. 1, no. 1, Sep. 2011, doi: 10.1101/CSHPERSPECT.A006841.
[2] M. Peeters, M. Jung, and A. Ayouba, “The origin and molecular epidemiology of HIV,” Expert Rev. Anti. Infect. Ther., vol. 11, no. 9, pp. 885–896, Jan. 2013, doi: 10.1586/14787210.2013.825443.
[3] F. E. McCutchan, “Global epidemiology of HIV,” J. Med. Virol., vol. 78, no. SUPPL. 1, Jul. 2006, doi: 10.1002/JMV.20599.
[4] J. S. Olesen et al., “HIV-2 continues to decrease, whereas HIV-1 is stabilizing in Guinea-Bissau,” AIDS, vol. 32, no. 9, pp. 1193–1198, Jun. 2018, doi: 10.1097/QAD.0000000000001827.
[5] Ö. Dağlı and P. T. Dergisi, “Screening of hepatitis and HIV infections in an alcohol and drug addiction treatment center,” Pamukkale Med. J., vol. 13, no. 1, pp. 177–186, Jan. 2020, doi: 10.31362/PATD.644886.
[6] D. Cattaneo and C. Gervasoni, “Pharmacokinetics and Pharmacodynamics of Cabotegravir, a Long-Acting HIV Integrase Strand Transfer Inhibitor,” Eur. J. Drug Metab. Pharmacokinet., vol. 44, no. 3, pp. 319–327, Jun. 2019, doi: 10.1007/S13318-018-0526-2.
[7] P. Cid-Silva et al., “Clinical Experience with the Integrase Inhibitors Dolutegravir and Elvitegravir in HIV-infected Patients: Efficacy, Safety and Tolerance,” Basic Clin. Pharmacol. Toxicol., vol. 121, no. 5, pp. 442–446, Nov. 2017, doi: 10.1111/BCPT.12828.
[8] A. L. Perryman et al., “A Dynamic Model of HIV Integrase Inhibition and Drug Resistance,” J. Mol. Biol., vol. 397, no. 2, p. 600, Mar. 2010, doi: 10.1016/J.JMB.2010.01.033.
[9] P. Gupta, P. Garg, and N. Roy, “Identification of Novel HIV-1 Integrase Inhibitors Using Shape-Based Screening, QSAR, and Docking Approach,” Chem. Biol. Drug Des., vol. 79, no. 5, pp. 835–849, May 2012, doi: 10.1111/J.1747-0285.2012.01326.X.
[10] V. Dewi Prasasty, R. Anthony Hutagalung, K. Grazzolie, F. Xaverius Ivan, and C. Vivitri Dewi Prasasty, “Selective docking of promising retroviral integrase inhibitors towards prototype foamy virus integrase,” ~ 265 ~ J. Med. Plants Stud., vol. 6, no. 2, pp. 265–272, 2018.
[11] A. Kolakowska, A. F. Maresca, I. J. Collins, and J. Cailhol, “Update on Adverse Effects of HIV Integrase Inhibitors,” Curr. Treat. options Infect. Dis., vol. 11, no. 4, pp. 372–387, Dec. 2019, doi: 10.1007/S40506-019-00203-7.
[12] Y. Zheng et al., “HPLC-MS/MS method for the simultaneous quantification of dolutegravir, elvitegravir, rilpivirine, darunavir, ritonavir, raltegravir and raltegravir-β-d-glucuronide in human plasma,” J. Pharm. Biomed. Anal., vol. 182, Apr. 2020, doi: 10.1016/J.JPBA.2020.113119.
[13] P. Gupta, P. Garg, and N. Roy, “In silico screening for identification of novel HIV-1 integrase inhibitors using QSAR and docking methodologies,” Med. Chem. Res. 2013 2210, vol. 22, no. 10, pp. 5014–5028, Feb. 2013, doi: 10.1007/S00044-013-0490-Y.
[14] J. Vora et al., “Molecular docking, QSAR and ADMET based mining of natural compounds against prime targets of HIV,” https://doi.org/10.1080/07391102.2017.1420489, vol. 37, no. 1, pp. 131–146, Jan. 2018, doi: 10.1080/07391102.2017.1420489.
[15] S. K. Tripathi, C. Selvaraj, S. K. Singh, and K. K. Reddy, “Molecular docking, qpld, and adme prediction studies on hiv-1 integrase leads,” Med. Chem. Res., vol. 21, no. 12, pp. 4239–4251, Dec. 2012, doi: 10.1007/S00044-011-9940-6.
[16] R. Singh, A. Nath, and B. Sharma, “Docking Studies of HIV-1 Reverse Transcriptase and HIV-1 Integrase with Phytocompounds of Carissa Carandas L.,” J. Clin. Res. HIV AIDS Prev., vol. 3, no. 4, pp. 10–19, May 2019, doi: 10.14302/ISSN.2324-7339.JCRHAP-19-2847.
[17] V. Jha et al., “Human immunodeficiency virus type 1: Role of proteins in the context of viral life cycle,” J Adv Biotechnol Exp Ther, vol. 5, no. 2, pp. 307–319, 2022, doi: 10.5455/jabet.2022.d117.
[18] S. K. Burley et al., “RCSB Protein Data Bank: Powerful new tools for exploring 3D structures of biological macromolecules for basic and applied research and education in fundamental biology, biomedicine, biotechnology, bioengineering and energy sciences,” Nucleic Acids Res., vol. 49, no. 1, pp. D437–D451, 2021, doi: 10.1093/NAR/GKAA1038.
[19] J. A. Duke, “Database of Biologically Active Phytochemicals & Their Activity,” Boca Raton, Fla CRC Press., p. 183, 1992.
[20] K. Mohanraj et al., “IMPPAT: A curated database of Indian Medicinal Plants, Phytochemistry And Therapeutics OPEN,” Sci. REPoRTS |, vol. 8, p. 4329, 2018, doi: 10.1038/s41598-018-22631-z.
[21] S. Kim et al., “PubChem in 2021: New data content and improved web interfaces,” Nucleic Acids Res., vol. 49, no. D1, pp. D1388–D1395, Jan. 2021, doi: 10.1093/nar/gkaa971.
[22] A. Daina, O. Michielin, and V. Zoete, “SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules,” Sci. Reports 2017 71, vol. 7, no. 1, pp. 1–13, Mar. 2017, doi: 10.1038/srep42717.
[23] L. Z. Benet, C. M. Hosey, O. Ursu, and T. I. Oprea, “BDDCS, the Rule of 5 and Drugability Graphical abstract HHS Public Access,” Adv Drug Deliv Rev, vol. 101, pp. 89–98, 2016, doi: 10.1016/j.addr.2016.05.007.
[24] E. F. Pettersen et al., “UCSF Chimera--a visualization system for exploratory research and analysis,” J. Comput. Chem., vol. 25, no. 13, pp. 1605–1612, Oct. 2004, doi: 10.1002/JCC.20084.
[25] S. Dallakyan and A. J. Olson, “Small-molecule library screening by docking with PyRx,” Methods Mol. Biol., vol. 1263, pp. 243–250, 2015, doi: 10.1007/978-1-4939-2269-7_19.
[26] V. Jha et al., “Screening of Phytochemicals as Potential Inhibitors of Breast Cancer using Structure Based Multitargeted Molecular Docking Analysis,” Phytomedicine Plus, vol. 2, no. 2, May 2022, doi: 10.1016/j.phyplu.2022.100227.
[27] E. Bursal et al., “Determination of Phenolic Content, Biological Activity, and Enzyme Inhibitory Properties with Molecular Docking Studies of Rumex nepalensis, an Endemic Medicinal Plant,” J. Food Nutr. Res., vol. 9, no. 3, pp. 114–123, Mar. 2021, doi: 10.12691/JFNR-9-3-3.
[28] Lindahl, Abraham, Hess, and van der Spoel, “GROMACS 2021 Source code,” Jan. 2021, doi: 10.5281/ZENODO.4457626.
[29] P. Banerjee, A. O. Eckert, A. K. Schrey, and R. Preissner, “ProTox-II: a webserver for the prediction of toxicity of chemicals,” Nucleic Acids Res., vol. 46, pp. 257–263, 2018, doi: 10.1093/nar/gky318.
[30] G. Xiong et al., “ADMETlab 2.0: an integrated online platform for accurate and comprehensive predictions of ADMET properties,” Nucleic Acids Res., vol. 49, no. W1, pp. W5–W14, Jul. 2021, doi: 10.1093/NAR/GKAB255.
[31] R. Akbar and W. K. Yam, “Interaction of ganoderic acid on HIV related target: molecular docking studies,” Bioinformation, vol. 7, no. 8, pp. 413–417, Dec. 2011, doi: 10.6026/97320630007413.
[32] R. G. Maroun et al., “Self-association and Domains of Interactions of an Amphipathic Helix Peptide Inhibitor of HIV-1 Integrase Assessed by Analytical Ultracentrifugation and NMR Experiments in Trifluoroethanol/H2O Mixtures *,” J. Biol. Chem., vol. 274, no. 48, pp. 34174–34185, Nov. 1999, doi: 10.1074/JBC.274.48.34174.
[33] C. A. Lipinski, F. Lombardo, B. W. Dominy, and P. J. Feeney, “Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings,” Adv. Drug Deliv. Rev., vol. 46, no. 1–3, pp. 3–26, Mar. 2001, doi: 10.1016/S0169-409X(00)00129-0.
[34] D. F. Veber, S. R. Johnson, H. Y. Cheng, B. R. Smith, K. W. Ward, and K. D. Kopple, “Molecular properties that influence the oral bioavailability of drug candidates,” J. Med. Chem., vol. 45, no. 12, pp. 2615–2623, Jun. 2002, doi: 10.1021/JM020017N.
[35] D. Dahlgren, M. Sjöblom, and H. Lennernäs, “Intestinal absorption-modifying excipients: A current update on preclinical in vivo evaluations,” Eur. J. Pharm. Biopharm., vol. 142, pp. 411–420, Sep. 2019, doi: 10.1016/J.EJPB.2019.07.013.
[36] P. Tripathi, S. Ghosh, and S. Nath Talapatra, “Bioavailability prediction of phytochemicals present in Calotropis procera (Aiton) R. Br. by using Swiss-ADME tool,” World Sci. News, vol. 131, pp. 147–163, 2019, Accessed: Jun. 17, 2022. [Online]. Available: www.worldscientificnews.com
[37] Y. S. Krishnaiah, “Pharmaceutical Technologies for Enhanc-ing Oral Bioavailability of Poorly Soluble Drugs,” J Bioequiv Availab, vol. 2, no. 2, pp. 28–036, 2010, doi: 10.4172/jbb.1000027.
[38] P. Baghel, A. Roy, S. Verma, T. Satapathy, and S. Bahadur, “Amelioration of lipophilic compounds in regards to bioavailability as self-emulsifying drug delivery system (SEDDS),” Futur. J. Pharm. Sci. 2020 61, vol. 6, no. 1, pp. 1–11, Jun. 2020, doi: 10.1186/S43094-020-00042-0.
[39] V. Jha et al., “Computational screening of phytochemicals to discover potent inhibitors against chinkungunya virus,” Vegetos, vol. 34, no. 3, pp. 515–527, Sep. 2021, doi: 10.1007/S42535-021-00227-9.
[40] M. J. Waring, “Lipophilicity in drug discovery.,” Expert Opin. Drug Discov., vol. 5, no. 3, pp. 235–248, Mar. 2010, doi: 10.1517/17460441003605098.
[41] M. D. Segall and C. Barber, “Addressing toxicity risk when designing and selecting compounds in early drug discovery,” Drug Discov. Today, vol. 19, no. 5, pp. 688–693, 2014, doi: 10.1016/J.DRUDIS.2014.01.006.
[42] D. B. Kitchen, H. Decornez, J. R. Furr, and J. Bajorath, “Docking and scoring in virtual screening for drug discovery: methods and applications,” Nat. Rev. Drug Discov., vol. 3, no. 11, pp. 935–949, Nov. 2004, doi: 10.1038/NRD1549.
[43] L. Pinzi and G. Rastelli, “Molecular Docking: Shifting Paradigms in Drug Discovery,” Int. J. Mol. Sci., vol. 20, no. 18, Sep. 2019, doi: 10.3390/IJMS20184331.
[44] L. Martínez, “Automatic identification of mobile and rigid substructures in molecular dynamics simulations and fractional structural fluctuation analysis,” PLoS One, vol. 10, no. 3, Mar. 2015, doi: 10.1371/JOURNAL.PONE.0119264.
Cite This Article
  • APA Style

    Vikas Jha, Navdeep Kaur, Kabir Thakur, Vrushali Dhamapurkar, Prakruti Kapadia, et al. (2022). Molecular Docking and Molecular Dynamic Simulation of Potential Inhibitors of Integrase from Human Immunodeficiency Virus 1 (HIV-1) Using Phytochemicals. Computational Biology and Bioinformatics, 10(1), 34-48. https://doi.org/10.11648/j.cbb.20221001.16

    Copy | Download

    ACS Style

    Vikas Jha; Navdeep Kaur; Kabir Thakur; Vrushali Dhamapurkar; Prakruti Kapadia, et al. Molecular Docking and Molecular Dynamic Simulation of Potential Inhibitors of Integrase from Human Immunodeficiency Virus 1 (HIV-1) Using Phytochemicals. Comput. Biol. Bioinform. 2022, 10(1), 34-48. doi: 10.11648/j.cbb.20221001.16

    Copy | Download

    AMA Style

    Vikas Jha, Navdeep Kaur, Kabir Thakur, Vrushali Dhamapurkar, Prakruti Kapadia, et al. Molecular Docking and Molecular Dynamic Simulation of Potential Inhibitors of Integrase from Human Immunodeficiency Virus 1 (HIV-1) Using Phytochemicals. Comput Biol Bioinform. 2022;10(1):34-48. doi: 10.11648/j.cbb.20221001.16

    Copy | Download

  • @article{10.11648/j.cbb.20221001.16,
      author = {Vikas Jha and Navdeep Kaur and Kabir Thakur and Vrushali Dhamapurkar and Prakruti Kapadia and Shraddha Tiwari and Aparna Sahu and Divya Nikumb and Abhishek Kumar and Shruti Narvekar},
      title = {Molecular Docking and Molecular Dynamic Simulation of Potential Inhibitors of Integrase from Human Immunodeficiency Virus 1 (HIV-1) Using Phytochemicals},
      journal = {Computational Biology and Bioinformatics},
      volume = {10},
      number = {1},
      pages = {34-48},
      doi = {10.11648/j.cbb.20221001.16},
      url = {https://doi.org/10.11648/j.cbb.20221001.16},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.cbb.20221001.16},
      abstract = {Background: In tremendously effective antiretroviral therapy for Human Immunodeficiency Virus 1 (HIV1) infections, integrase inhibitors are essential drugs. Resistance resulting from mutations, on the other hand, poses a threat to the medication’s long-term efficacy in HIV-1 infected people. Purpose: The current study utilized in silico techniques, we searched for phytochemicals or compounds that can inhibit the activity of the integrase enzyme. Material & Methods: Compounds were collected from databases, and potential candidates were screened using pharmacokinetics and structure-based virtual screening methodologies. The compounds were docked, and the binding affinity was evaluated to set the cut-off value for selecting compounds. When compared to standard drugs, some compounds had a higher binding affinity. Molecular dynamics simulation was then used to gain insight into the stability of the complexes, revealing two lead compounds, Withaferin and Isatin, indicating that these compounds have potency for drug development. These compounds were further investigated for their toxicity, indicating that Isatin was safe among the two. Conclusion: Thus, the study showcased that Isatin is a suitable drug candidate, and we hope that the findings of this study will be useful in the development of an antiviral drug against integrase enzyme.},
     year = {2022}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Molecular Docking and Molecular Dynamic Simulation of Potential Inhibitors of Integrase from Human Immunodeficiency Virus 1 (HIV-1) Using Phytochemicals
    AU  - Vikas Jha
    AU  - Navdeep Kaur
    AU  - Kabir Thakur
    AU  - Vrushali Dhamapurkar
    AU  - Prakruti Kapadia
    AU  - Shraddha Tiwari
    AU  - Aparna Sahu
    AU  - Divya Nikumb
    AU  - Abhishek Kumar
    AU  - Shruti Narvekar
    Y1  - 2022/06/30
    PY  - 2022
    N1  - https://doi.org/10.11648/j.cbb.20221001.16
    DO  - 10.11648/j.cbb.20221001.16
    T2  - Computational Biology and Bioinformatics
    JF  - Computational Biology and Bioinformatics
    JO  - Computational Biology and Bioinformatics
    SP  - 34
    EP  - 48
    PB  - Science Publishing Group
    SN  - 2330-8281
    UR  - https://doi.org/10.11648/j.cbb.20221001.16
    AB  - Background: In tremendously effective antiretroviral therapy for Human Immunodeficiency Virus 1 (HIV1) infections, integrase inhibitors are essential drugs. Resistance resulting from mutations, on the other hand, poses a threat to the medication’s long-term efficacy in HIV-1 infected people. Purpose: The current study utilized in silico techniques, we searched for phytochemicals or compounds that can inhibit the activity of the integrase enzyme. Material & Methods: Compounds were collected from databases, and potential candidates were screened using pharmacokinetics and structure-based virtual screening methodologies. The compounds were docked, and the binding affinity was evaluated to set the cut-off value for selecting compounds. When compared to standard drugs, some compounds had a higher binding affinity. Molecular dynamics simulation was then used to gain insight into the stability of the complexes, revealing two lead compounds, Withaferin and Isatin, indicating that these compounds have potency for drug development. These compounds were further investigated for their toxicity, indicating that Isatin was safe among the two. Conclusion: Thus, the study showcased that Isatin is a suitable drug candidate, and we hope that the findings of this study will be useful in the development of an antiviral drug against integrase enzyme.
    VL  - 10
    IS  - 1
    ER  - 

    Copy | Download

Author Information
  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • Department of Biotechnology, B. K. Birla College, Kalyan, India

  • Department of Biotechnology, B. K. Birla College, Kalyan, India

  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • National Facility for Biopharmaceuticals, Guru Nanak Khalsa College of Arts, Science & Commerce, Mumbai, India

  • Sections