Identification of Flavonoids of Kalanchoe Pinnata as Candidate Drugs for COVID-19 Gamma-Variant Treatment

Authors

  • Yulia Aurora Biomedicine Department, School of Life Science, Indonesia International Institute for Life Sciences
  • Immanuella Putri N Tarigan Biomedicine Department, School of Life Science, Indonesia International Institute for Life Sciences
  • Nio Meinwen Metta Suryanto Biomedicine Department, School of Life Science, Indonesia International Institute for Life Sciences
  • Pramujinoto Santosa Biomedicine Department, School of Life Science, Indonesia International Institute for Life Sciences
  • Viona Pricillia Bioinformatics Department, School of Life Science, Indonesia International Institute for Life Sciences
  • Arli Aditya Parikesit Bioinformatics Department, School of Life Science, Indonesia International Institute for Life Sciences https://orcid.org/0000-0001-8716-3926

DOI:

https://doi.org/10.11113/mjfas.v18n6.2594

Keywords:

Flavonoids, SARS-CoV-2 gamma variant, Spike protein, ACE2 receptor

Abstract

Treatment of COVID-19 that is based on plants could be a more cost-effective therapy against the disease. Flavonoids, a group of compounds that have been observed to have various effects, including antiviral activity, were chosen as the candidate molecule for treatment of COVID-19. Kalanchoe Pinnata is one of the plants containing flavonoids that has been demonstrated to have antiviral activity. The structure of ACE2 and various flavonoids were retrieved and cleaned from unnecessary residues. The ACE2 structure was subjected to molecular docking in order to analyze the binding affinity. Following that, the ADME properties of each flavonoid were analyzed accordingly. The QSAR analysis was also performed for each type of flavonoid. Lastly, molecular dynamics simulation was conducted. All of the tested compounds were able to bind to human ACE2 and SARS-CoV-2 Spike protein, but were unable to compete with them as the binding affinity of the compounds to the protein were lower compared to ACE2-Spike interaction. The ADME and toxicity analysis showed that most of the ligands were able to be absorbed by the GI tract, but have low bioavailability. The compounds also cause no major toxicity effects and were able to be sufficiently distributed to the body. Molecular dynamics analysis also revealed that among the compounds, quercetin and rutin were able to interact with ACE2 and Spike protein stably. The QSAR analysis showed that friedelin, kaempferol, quercetin, and rutin are mostly non-toxic, but the high Cramer values indicate that there are no initial safety impressions for these molecules and could cause toxicity. In conclusion, quercetin and rutin have potential to be a candidate for COVID-19 drug development based on the in-silico predictions results obtained. Friedelin and Narcissin whose affinity to the proteins were relatively stronger but had unstable interactions from molecular dynamics simulation results, may also be a potential COVID-19 treatment with further investigation. However, further research is required to assess the effectiveness and also specially to measure the toxicity of the compounds.

References

Wu, Y. C., Chen, C. S., & Chan, Y. J. (2020). The outbreak of COVID-19: An overview. Journal of the Chinese Medical Association: JCMA, 83(3), 217-220. https://doi.org/10.1097/JCMA.0000000000000270.

Krishnan, A., Hamilton, J. P., Alqahtani, S. A., & Woreta, T. A. (2021). COVID-19: An overview and a clinical update. World Journal of Clinical Cases, 9(1), 8-23. https://doi.org/10.12998/wjcc.v9.i1.8.

Cucinotta, D., & Vanelli, M. (2020). WHO declares COVID-19 a pandemic. Acta Bio-medica: Atenei Parmensis, 91(1), 157-160. https://doi.org/10.23750/abm.v91i1.9397.

Liu, Y., Gayle, A. A., Wilder-Smith, A., & Rocklöv, J. (2020). The reproductive number of COVID-19 is higher compared to SARS coronavirus. Journal of Travel Medicine, 27(2), taaa021. https://doi.org/10.1093/jtm/taaa021.

World Health Organization. (2021). WHO Coronavirus (COVID-19) Dashboard. Retrieved 28 September 2021, from https://covid19.who.int.

Petersen, E., Koopmans, M., Go, U., Hamer, D. H., Petrosillo, N., Castelli, F., … Simonsen, L. (2020). Comparing SARS-CoV-2 with SARS-CoV and influenza pandemics. The Lancet Infectious Diseases. https://doi.org/10.1016/s1473-3099(20)30484-9.

World Health Organization. Naming the coronavirus disease (COVID-19) and the virus that causes it. Retrieved 28 September 2021, from https://www.who.int/emergencies/diseases/novel-coronavirus-2019/technical-guidance/naming-the-coronavirus-disease-(covid-2019)-and-the-virus-that-causes-it.

Shang, J., Wan, Y., Luo, C., Ye, G., Geng, Q., Auerbach, A., & Li, F. (2020). Cell entry mechanisms of SARS-CoV-2. Proceedings of the National Academy of Sciences, 117(21), 11727-11734. https://doi.org/10.1073/pnas.2003138117.

Cascella, M., Rajnik, M., Aleem, A., Dulebohn, S., & Napoli, R. (2021). Features, evaluation, and treatment of coronavirus (COVID-19). Retrieved 22 September 2021, from https://www.ncbi.nlm.nih.gov/books/NBK554776/.

Yuan H, Ma Q, Ye L, Piao G. (2016). The traditional medicine and modern medicine from natural products. Molecules. 21(5): 559. Doi: 10.3390/molecules21050559. PMID: 27136524; PMCID: PMC6273146.

Panche, A. N., Diwan, A. D., & Chandra, S. R. (2016). Flavonoids: an overview. Journal of Nutritional Science, 5, e47. https://doi.org/10.1017/jns.2016.4.

Lalani, S., & Poh, C. (2020). Flavonoids as antiviral agents for enterovirus A71 (EV-A71). Viruses, 12(2), 184. https://doi.org/10.3390/v12020184.

Ninfali, P., Antonelli, A., Magnani, M., & Scarpa, E. S. (2020). Antiviral properties of flavonoids and delivery strategies. Nutrients, 12(9), 2534. https://doi.org/10.3390/nu12092534.

Vickers, A., Zollman, C., & Lee, R. (2001). Herbal medicine. The Western Journal of Medicine, 175(2), 125-128. https://doi.org/10.1136/ewjm.175.2.125.

Aoki, C., Hartati, S., Santi, M. R., Lydwina, Firdaus, R., Hanafi, M., Kardono, L. B., Shimizu, Y. K., Sudarmono, P., & Hotta, H. (2014). Isolation and identification of substances with anti-hepatitis c activities from kalanchoe pinnata. International Journal of Pharmacy and Pharmaceutical Sciences, 6(2), 211-215.

Rajsekhar, P. B., Arvind Bharani, R. S., Ramachandran, M., Jini Angel, K., & Vardhini, R. S. P. (2016). A comparative study on the extracts of Kalanchoe Pinnata (Linn.) Pers. using chromatographic techniques. International Journal of Pharmaceutical Sciences and Research, 7(1), 345-348.

Trott, O., & Olson, J. (2010). AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. Journal of Computational Chemistry, 31, 455-461. https://doi.org/10.1002/jcc.21334.

BIOVIA, Dassault Systèmes, BIOVIA Workbook, 2020; BIOVIA Pipeline Pilot, Release 2020, San Diego: Dassault Systèmes, [2021].

Desta, I. T., Porter, K. A., Xia, B., Kozakov, D., Vajda, S. (2020). Performance and its limits in rigid body protein-protein docking. Structure, 28(9), 1071-1081. https://doi.org/10.1016/j.str.2020.06.006.

Vajda, S., Yueh, C., Beglov, D., Bohnuud, T., Mottarella, S. E., Xia, B., Hall, D. R., Kozakov, D. (2017). New additions to the ClusPro server motivated by CAPRI. Proteins: Structure, Function, and Bioinformatics, 85(3), 435-444.

Kozakov, D., Beglov, D., Bohnuud, T., Mottarella, S., Xia, B., Hall, D. R., Vajda, S. (2013). How good is automated protein docking? Proteins: Structure, Function, and Bioinformatics, 81(12), 2159-66.

Kozakov, D., Hall, D. R., Xia, B., Porter, K. A., Padhorny, D., Yueh, C., Beglov, D., Vajda, S. (2017). The ClusPro web server for protein-protein docking. Nature Protocols, 12(2), 255-278.

Vangone, A. & Bonvin, A. M. J. J. (2015). Contact-based prediction of binding affinity in protein-protein complexes. eLife, 4, e07454.

Xue, L., Rodrigues, J., Kastritis, P., Bonvin, A., & Vangone, A. (2016). PRODIGY: a web server for predicting the binding affinity of protein–protein complexes. Bioinformatics, btw514. https://doi.org/10.1093/bioinformatics/btw514.

Xiong, G., Wu, Z., Yi, J., Fu, L., Yang, Z., Hsieh, C., Yin, M., Zeng, X., Wu, C., Lu, A., Chen, X., Hou, T., & Cao, D. (2021). ADMETlab 2.0: an integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Research, 49(W1), W5-W14. https://doi.org/10.1093/nar/gkab255.

Dong, J., Wang, N N., Yao, Z J. et al. (2018). ADMETlab: a platform for systematic ADMET evaluation based on a comprehensively collected ADMET database, J Cheminform. 10, 29. https://doi.org/10.1186/s13321-018-0283-x.

Lagunin, A., Stepanchikova, A., Filimonov, D., & Poroikov, V. (2000). PASS: prediction of activity spectra for biologically active substances. Bioinformatics (Oxford, England), 16(8), 747-748. https://doi.org/10.1093/bioinformatics/16.8.747.

Huang, T., Sun, G., Zhao, L., Zhang, N., Zhong, R., & Peng, Y. (2021). Quantitative structure-activity relationship (QSAR) studies on the toxic effects of nitroaromatic compounds (NACs): a systematic review. International Journal of Molecular Sciences, 22(16), 8557. https://doi.org/10.3390/ijms22168557.

Banerjee, P., Eckert, A. O., Schrey, A. K., & Preissner, R. (2018). ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Research, 46(W1), W257-W263. https://doi.org/10.1093/nar/gky318.

Wijaya, R. M., Hafidzhah, M. A., Kharisma, V. D., Ansori, A. N. M., & Parikesit, A. A. (2021). COVID-19 in silico drug with zingiber officinale natural product compound library targeting the mpro protein. Makara Journal of Science, 25(3). https://doi.org/10.7454/mss.v25i3.1244. Available at: https://scholarhub.ui.ac.id/science/vol25/iss3/5.

Kuriata, A., Gierut, A. M., Oleniecki, T., Ciemny, M. P., Kolinski, A., Kurcinski, M., & Kmiecik, S. (2018). CABS-flex 2.0: a web server for fast simulations of flexibility of protein structures. Nucleic Acids Research, 46(W1), W338-W343. https://doi.org/10.1093/nar/gky356.

Duru, C. E., Umar, H. I., Duru, I. A., Enenebeaku, U. E., Ngozi-Olehi, L. C., & Enyoh, C. E. (2021). Blocking the interactions between human ACE2 and coronavirus spike glycoprotein by selected drugs: A computational perspective. Environmental Analysis Health and Toxicology, 36(2). https://doi.org/10.5620/eaht.2021010.

Rizzuti, B., Grande, F., Conforti, F., Jimenez-Alesanco, A., Ceballos-Laita, L., Ortega-Alarcon, D., … Velazquez-Campoy, A. (2021). Rutin is a low micromolar inhibitor of SARS-CoV-2 main protease 3CLpro: Implications for drug design of quercetin analogs. Biomedicines, 9(4), 375. https://doi.org/10.3390/biomedicines9040375.

Agrawal, P. K., Agrawal, C., & Blunden, G. (2021). Rutin: a potential antiviral for repurposing as a SARS-CoV-2 main protease (Mpro) Inhibitor. Natural Product Communications, 16(4), 1934578X2199172. https://doi.org/10.1177/1934578x2199172.

Owona, V., Galani, B., & Moundipa, P. (2021). In silico identification of apigenin and narcissin (food-flavonoids) as potential targets against SARS-CoV-2 viral proteins: comparison with the effect of remdesivir. Journal Of Clinical Anesthesia and Pain Management, 5(1). https://doi.org/ 10.36959/377/356.

Adejoro, I. A., Babatunde, D. D., & Tolufashe, G. F. (2020). Molecular docking and dynamic simulations of some medicinal plants compounds against SARS-CoV-2: an in silico study. Journal of Taibah University for Science, 14(1), 1563-1570. https://doi.org/10.1080/16583655.2020.1848049.

Kar, P., Sharma, N. R., Singh, B., Sen, A., & Roy, A. (2021). Natural compounds from Clerodendrum spp. as possible therapeutic candidates against SARS-CoV-2: An in silico investigation. Journal of Biomolecular Structure & Dynamics, 39(13), 4774-4785. https://doi.org/10.1080/07391102.2020.178094.

Pan, B., Fang, S., Zhang, J., Pan, Y., Liu, H., & Wang, Y. et al. (2020). Chinese herbal compounds against SARS-CoV-2: Puerarin and quercetin impair the binding of viral S-protein to ACE2 receptor. Computational And Structural Biotechnology Journal, 18, 3518-3527. https://doi.org/10.1016/j.csbj.2020.11.010.

Tallei, T. E., Tumilaar, S. G., Niode, N. J., Fatimawali, Kepel, B.J., Idroes, R., Effendi, Y., Sakib, S.A., & Emran, T. B. (2020). Potential of plant bioactive compounds as SARS-CoV-2 main protease (Mpro) and Spike (S) glycoprotein inhibitors: a molecular docking study. Scientifica, 2020, 6307457. https://doi.org/10.1155/2020/6307457.

Nguyen, T., Jung, J. H., Kim, M. K., Lim, S., Choi, J. M., Chung, B., Kim, D. W., & Kim, D. (2021). The inhibitory effects of plant derivate polyphenols on the main protease of SARS Coronavirus 2 and their structure-activity relationship. Molecules (Basel, Switzerland), 26(7), 1924. https://doi.org/10.3390/molecules26071924.

Shawan, M., Halder, S. K., & Hasan, M. A. (2021). Luteolin and abyssinone II as potential inhibitors of SARS-CoV-2: an in silico molecular modeling approach in battling the COVID-19 outbreak. Bulletin of the National Research Centre, 45(1), 27. https://doi.org/10.1186/s42269-020-00479-6.

Shadrack, D. M., Deogratias, G., Kiruri, L. W., Onoka, I., Vianney, J.-M., Swai, H., & Nyandoro, S. S. (2021). Luteolin: a blocker of SARS-CoV-2 cell entry based on relaxed complex scheme, molecular dynamics simulation, and metadynamics. Journal of Molecular Modeling, 27(8). https://doi.org/10.1007/s00894-021-04833-x.

Bernatova, I., & Liskova, S. (2021). Mechanisms Modified by (−)-Epicatechin and Taxifolin Relevant for the Treatment of Hypertension and Viral Infection: Knowledge from Preclinical Studies. Antioxidants, 10(3), 467. https://doi.org/10.3390/antiox10030467.

Downloads

Published

29-12-2022