In silico studies on potential MCF-7 inhibitors of alkaloid and phenolic compounds isolated from Zanthoxylum nitidum: A combination of molecular docking and admet analysis

Tran Thi Tuyen, Pham Cao Bach, Ha Viet Hai, Pham Thi Hong Minh, Nguyen Thi Hong Van, Pham Minh Quan
Author affiliations

Authors

  • Tran Thi Tuyen Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Ha Noi, Viet Nam
  • Pham Cao Bach Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Ha Noi, Viet Nam
  • Ha Viet Hai Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Ha Noi, Viet Nam
  • Pham Thi Hong Minh Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Ha Noi, Viet Nam
  • Nguyen Thi Hong Van Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Ha Noi, Viet Nam
  • Pham Minh Quan Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Ha Noi, Viet Nam https://orcid.org/0000-0001-6922-1627

DOI:

https://doi.org/10.15625/2525-2518/16945

Keywords:

Zanthoxylum nitidum, MCF-7, tubulin, anticancer, Molecular Docking, ADMET analysis

Abstract

Cancer is one of the leading cause of death worldwide that cause a burden for the health system and economic of both developed and developing countries. Tubulin is a wellknown biochemical target in the field of cancer drug discovery. The inhibition of tubulin-microtubule dynamics would result in the mitosis during cell division, thus, killing the cancerous cells. To date, there have been some tubulin targeting drugs have been developed including paclitaxel and the vinca alkaloids, however, treatments are not high ineffective and efforts to discover new molecules are still in urgent needed. In this study, computational molecular simulation and assessment of drug-like properties were used to gain insights into the binding ability of 16 alkaloid and phenolic compounds isolated from Zanthoxylum nitidum on tubulin protein. Among studied candidates, compounds 8 and 10 were identified as potential candidates for inhibiting the function of tubulin at the active site regarding binding affinity, dock pose and ADMET properties analysis. These findings shed light on the anti-cancer potential of compounds isolated from Zanthoxylum nitidum.

Downloads

Download data is not yet available.

References

Minatel I. O., Borges C. V., Ferreira M. I., Gomez H. A. G., Chen C. Y. O. and Lima G. P. P. - Phenolic Compounds: Functional Properties, Impact of Processing and Bioavailability, In: Phenolic Compounds - Biological Activity, Chapter Chapter 1, 2017.

Thawabteh A., Juma S., Bader M., Karaman D., Scrano L., Bufo S. and Karaman R. - The Biological Activity of Natural Alkaloids against Herbivores, Cancerous Cells and Pathogens, Toxins (Basel) 11 (11) (2019). https://doi.org/10.3390/toxins11110656.

Nguyen T. H. V., Tran T. T., Cam T. I., Pham M. Q., Pham Q. L., Vu D. H., Nguyen X. N., Chau V. M. and Van K. P. - Alkaloids From Zanthoxylum nitidum and Their Cytotoxic Activity, Nat. Prod. Commun. 14 (5) (2019). https://doi.org/10.1177/ 1934578x19844133.

González-Abuín N., Martínez-Micaelo N., Margalef M., Blay M., Arola-Arnal A., Muguerza B., Ardévol A. and Pinent M. - A grape seed extract increases active glucagon-like peptide-1 levels after an oral glucose load in rats, Food Funct. 5 (9) (2014). https://doi.org/10.1039/c4fo00447g.

Doan L. P., Nguyen T. T., Pham M. Q., Tran Q. T., Pham Q. L., Tran D. Q., Than V. T. and Bach L. G. - Extraction Process, Identification of Fatty Acids, Tocopherols, Sterols and Phenolic Constituents, and Antioxidant Evaluation of Seed Oils from Five Fabaceae Species, Processes 7 (7) (2019). https://doi.org/10.3390/pr7070456.

Otshudi A. L., Apers S., Pieters L., Claeys M., Pannecouque C., De Clercq E., Van Zeebroeck A., Lauwers S., Frédérich M. and Foriers A. - Biologically active bisbenzylisoquinoline alkaloids from the root bark of Epinetrum villosum, J. Ethnopharmacol 102 (1) (2005) 89-94. https://doi.org/10.1016/j.jep.2005.05.021.

Seo Y. S., Yim M. J., Kim B. H., Kang K. R., Lee S. Y., Oh J. S., You J. S., Kim S. G., Yu S. J., Lee G. J., Kim D. K., Kim C. S., Kim J. S., and Kim J. S. - Berberine-induced anticancer activities in FaDu head and neck squamous cell carcinoma cells, Oncol. Rep. 34 (6) (2015) 3025-3034. https://doi.org/10.3892/or.2015.4312.

Lou C., Yokoyama S., Saiki I., and Hayakawa Y. - Selective anticancer activity of hirsutine against HER2-positive breast cancer cells by inducing DNA damage, Oncol. Rep. 33 (4) (2015) 2072-2076. https://doi.org/10.3892/or.2015.3796.

Zhang Y., Wu L. J., Tashiro S. I., Onodera S., and Ikejima T. - Intracellular Regulation of Evodiamine-Induced A375-S2 Cell Death, Biol. Pharm. Bull. 26 (11) (2003) 1543-1547. https://doi.org/10.1248/bpb.26.1543.

Yixiang H., Shenghui Z., Jianbo W., Kang Y., Yu Z., Lihui Y. and Laixi B. - Matrine induces apoptosis of human multiple myeloma cells via activation of the mitochondrial pathway, Leuk. Lymphoma 51 (7) (2010) 1337-1346. https://doi.org/10.3109/ 10428194.2010.488708.

Jabbarzadeh Kaboli P., Rahmat A., Ismail P. and Ling K. H. - Targets and mechanisms of berberine, a natural drug with potential to treat cancer with special focus on breast cancer, Eur. J. Pharmacol 740 (2014) 584-595. https://doi.org/10.1016/j.ejphar.2014.06.025.

Wang C., Li S. and Wang M. W. - Evodiamine-induced human melanoma A375-S2 cell death was mediated by PI3K/Akt/caspase and Fas-L/NF-κB signaling pathways and augmented by ubiquitin–proteasome inhibition, Toxicol. In Vitro 24 (3) (2010) 898-904. https://doi.org/10.1016/j.tiv.2009.11.019.

Dai Z. J., Gao J., Ji Z. Z., Wang X. J., Ren H. T., Liu X. X., Wu W. Y., Kang H. F., and Guan H. T. - Matrine induces apoptosis in gastric carcinoma cells via alteration of Fas/FasL and activation of caspase-3, J. Ethnopharmacol 123 (1) (2009) 91-96. https:// doi.org/10.1016/j.jep.2009.02.022.

Bhardwaj R. K., Glaeser H., Becquemont L., Klotz U., Gupta S. K., and Fromm M. F. - Piperine, a Major Constituent of Black Pepper, Inhibits Human P-glycoprotein and CYP3A4, J. Pharmacol. Exp. Ther. 302 (2) (2002) 645-650. https://doi.org/ 10.1124/jpet.102.034728.

Hussain A. R., Al-Jomah N. A., Siraj A. K., Manogaran P., Al-Hussein K., Abubaker J., Platanias L. C., Al-Kuraya K. S. and Uddin S. - Sanguinarine-Dependent Induction of Apoptosis in Primary Effusion Lymphoma Cells, Cancer Res. 67 (8) (2007) 3888-3897. https://doi.org/10.1158/0008-5472.Can-06-3764.

Stefely J. A., Palchaudhuri R., Miller P. A., Peterson R. J., Moraski G. C., Hergenrother P. J. and Miller M. J. - N-((1-Benzyl-1H-1,2,3-triazol-4-yl)methyl)arylamide as a New Scaffold that Provides Rapid Access to Antimicrotubule Agents: Synthesis and Evaluation of Antiproliferative Activity Against Select Cancer Cell Lines, J. Med. Chem. 53 (8) (2010) 3389-3395. https://doi.org/10.1021/jm1000979.

Parveen I., Khan P., Ali S., Hassan M. I. and Ahmed N. - Synthesis, molecular docking and inhibition studies of novel 3-N-aryl substituted-2-heteroarylchromones targeting microtubule affinity regulating kinase 4 inhibitors, Eur. J. Med. Chem. 159 (2018) 166-177. https://doi.org/10.1016/j.ejmech.2018.09.030.

Shi J. and Mitchison T. J. - Cell death response to anti-mitotic drug treatment in cell culture, mouse tumor model and the clinic, Endocr. Relat. Cancer 24 (9) (2017) T83-T96. https://doi.org/10.1530/erc-17-0003.

van Vuuren R. J., Visagie M. H., Theron A. E. and Joubert A. M. - Antimitotic drugs in the treatment of cancer, Cancer Chemother. Pharmacol. 76 (6) (2015) 1101-1112. https://doi.org/10.1007/s00280-015-2903-8.

Santos-Garcia L., Assis L. C., Silva D. R., Ramalho T. C. and da Cunha E. F. F. - QSAR analysis of nicotinamidic compounds and design of potential Bruton’s tyrosine kinase (Btk) inhibitors, J. Biomol. Struct. Dyn. 34 (7) (2016) 1421-1440. https://doi.org/ 10.1080/07391102.2015.1070750.

Wang F. F., Yang W., Shi Y. H. and Le G. W. - In silico study on β-aminoketone derivatives as thyroid hormone receptor inhibitors: a combined 3D-QSAR and molecular docking study, J. Biomol. Struct. Dyn. (2016) 1-13. https://doi.org/10.1080/ 07391102.2015.1124806.

Tuyen T. T., Quan P. M., Thu Le V. T., Toan T. Q., Nghi D. H., Bach P. C., Inh C. T., Hanh N. P., Vien T. A., Hong Minh P. T., Long P. Q., Khoi Nguyen N. H., Thuy Dung P. N. and Hong Van N. T. - Chemical Composition, Antimicrobial, and Cytotoxic Activities of Leaf, Fruit, and Branch Essential Oils Obtained From Zanthoxylum nitidum Grown in Vietnam, Nat. Prod. Commun. 16 (1) (2021). https://doi.org/10.1177/1934578x20985649.

Van N. T. H., Tuyen T. T., Quan P. M., Long P. Q., Nhiem N. X., Tai B. H., Inh C. T., Hoang V. D., Van Kiem P., Van Minh C., Park S. and Kim S. H. - Alkaloid glycosides and their cytotoxic constituents from Zanthoxylum nitidum, Phytochemistry Letters 32 (2019) 47-51. https://doi.org/10.1016/j.phytol.2019.04.022.

Schrodinger, LLC, The PyMOL Molecular Graphics System, Version 1.3r1. 2010.

Allouche A. R. - Gabedit-A graphical user interface for computational chemistry softwares, J. Comput. Chem. 32 (1) (2011) 174-182. https://doi.org/10.1002/jcc.21600.

Gigant B., Wang C., Ravelli R. B. G., Roussi F., Steinmetz M. O., Curmi P. A., Sobel A. and Knossow M. - Structural basis for the regulation of tubulin by vinblastine, Nature 435 (7041) (2005) 519-522. https://doi.org/10.1038/nature03566.

Morris G. M., Huey R., Lindstrom W., Sanner M. F., Belew R. K., Goodsell D. S. and Olson A. J. - AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility, J. Comput. Chem. 30 (16) (2009) 2785-2791. https://doi.org/10.1002/jcc.21256.

Pineda O., Farràs J., Maccari L., Manetti F., Botta M. and Vilarrasa J. - Computational comparison of microtubule-stabilising agents laulimalide and peloruside with taxol and colchicine, Bioorg. Med. Chem. Lett. 14 (19) (2004) 4825-4829. https://doi.org/ 10.1016/j.bmcl.2004.07.053.

Dassault Systèmes BIOVIA, Discovery Studio Visualizer, v20.1.0.19295, San Diego: Dassault Systèmes, (2020).

Cheng F., Li W., Zhou Y., Shen J., Wu Z., Liu G., Lee P. W. and Tang Y. - AdmetSAR: A Comprehensive Source and Free Tool for Assessment of Chemical ADMET Properties, J. Chem. Inf. Model. 52 (11) (2012) 3099-3105. https://doi.org/10.1021/ci300367a.

Quan P. M., Huong L. T. T., Toan T. Q., Hung N. P., Nam P. H., Kiet N. T., Ha N. X., Le D. T. T., An T. N. T., Show P. L. and Thi H. H. P. - Cannabis sativa L. chemical compositions as potential plasmodium falciparum dihydrofolate reductase-thymidinesynthase enzyme inhibitors: An in silico study for drug development, Open Chemistry 19 (1) (2021) 1235-1241. https://doi.org/10.1515/chem-2021-0102.

Wang G., Liu W., Gong Z., Huang Y., Li Y. and Peng Z. - Synthesis, biological evaluation, and molecular modelling of new naphthalene-chalcone derivatives as potential anticancer agents on MCF-7 breast cancer cells by targeting tubulin colchicine binding site, J. Enzyme Inhib. Med. Chem. 35 (1) (2019) 139-144. https://doi.org/10.1080/14756366.2019.1690479.

Cheng H. C. - The power issue: determination of KB or Ki from IC50, J. Pharmacol. Toxicol. Methods 46 (2) (2001) 61-71. https://doi.org/10.1016/s1056-8719(02)00166-1.

Downloads

Published

05-04-2023

How to Cite

[1]
T. Thi Tuyen, P. Cao Bach, H. Viet Hai, P. Thi Hong Minh, N. Thi Hong Van, and P. Minh Quan, “In silico studies on potential MCF-7 inhibitors of alkaloid and phenolic compounds isolated from Zanthoxylum nitidum: A combination of molecular docking and admet analysis”, Vietnam J. Sci. Technol., vol. 61, no. 3, pp. 355–364, Apr. 2023.

Issue

Section

Natural Products

Most read articles by the same author(s)