In Silico Prediction of Sulforaphane and Iberin from Brassica oleracea as Potential Anticancer Agent

Article Preview

Abstract:

Sulforaphane and iberin, the isothiocyanate compounds found mainly in Brassica oleracea, are well-reported as anticancer agents. This study examined the potential of sulforaphane and iberin to inhibit proteins implicated in cancer-signaling pathways, such as NF-κB, TGF-β Receptor, Wnt/β-catenin, and MAPK. The presence of sulforaphane and iberin in Brassica oleracea was previously analyzed using LCMS. Subsequently, molecular docking simulation using PyRx Virtual Screening Tool were conducted to assess their binding affinities and interactions with protein targets (3RZF, 5EBZ, 5E8V, 3TZM, 1H8F, 1M17, and 3I81). LCMS analysis revealed the presence of sulforaphane and iberin compounds in Brassica oleracea, identified by their respective abundance peaks in the chromatogram at molecular weights of 177 and 164 g/mol. Furthermore, our results consistently demonstrated that sulforaphane exhibits a higher binding affinity for multiple target proteins than iberin, as evidenced by its lower binding energy values and greater number of amino acid interactions. In conclusion, these findings suggest that sulforaphane and iberin may serve as promising inhibitor ligands for cancer treatment.

You might also be interested in these eBooks

Info:

Periodical:

Engineering Headway (Volume 24)

Pages:

109-120

Citation:

Online since:

July 2025

Export:

Price:

Permissions CCC:

Permissions PLS:

Сopyright:

© 2025 Trans Tech Publications Ltd. All Rights Reserved

Share:

Citation:

* - Corresponding Author

[1] B. M. Twaij and M. N. Hasan, Bioactive Secondary Metabolites from Plant Sources: Types, Synthesis, and Their Therapeutic Uses, Int. J. Plant Biol. 13(1) (2022) 4–14

DOI: 10.3390/ijpb13010003

Google Scholar

[2] M. Mitsiogianni, D. T. Trafalis, R. Franco, V. Zoumpourlis, A. Pappa, and M. I. Panayiotidis, Sulforaphane and iberin are potent epigenetic modulators of histone acetylation and methylation in malignant melanoma, Eur. J. Nutr. 60(1) (2021) 147–158

DOI: 10.1007/s00394-020-02227-y

Google Scholar

[3] F. J. Barba, N. Nikmaram, S. Roohinejad, A. Khelfa, Z. Zhu, and M. Koubaa, Bioavailability of Glucosinolates and Their Breakdown Products: Impact of Processing, Front. Nutr. 3 (2016) 1–12

DOI: 10.3389/fnut.2016.00024

Google Scholar

[4] K. Palani et al., Influence of fermentation on glucosinolates and glucobrassicin degradation products in sauerkraut, Food Chem. 190 (2016) 755–762

DOI: 10.1016/j.foodchem.2015.06.012

Google Scholar

[5] J. Sun et al., The effect of processing and cooking on glucoraphanin and sulforaphane in brassica vegetables, Food Chem. 360 130007 (2021)

DOI: 10.1016/j.foodchem.2021.130007

Google Scholar

[6] J. A. Bouranis et al., Article composition of the gut microbiome influences production of sulforaphane-nitrile and iberin-nitrile from glucosinolates in broccoli sprouts, Nutrients. 13(9) (2021)

DOI: 10.3390/nu13093013

Google Scholar

[7] S. A. Marshall et al., The broccoli-derived antioxidant sulforaphane changes the growth of gastrointestinal microbiota, allowing for the production of anti-inflammatory metabolites, J. Funct. Foods. 107 105645 (2023).

DOI: 10.1016/j.jff.2023.105645

Google Scholar

[8] M. Parasakthi and K. Sanjeev, Sulforaphane as a preventive agent in Oral Cancer- A Systematic Review, Oral Oncol. Reports. 10 100429 (2024)

DOI: 10.1016/j.oor.2024.100429

Google Scholar

[9] M. A. Amer, T. R. Mohamed, R. A. Abdel Rahman, M. Ali, and A. Badr, Studies on exogenous elicitors promotion of sulforaphane content in broccoli sprouts and its effect on the MDA-MB-231 breast cancer cell line, Ann. Agric. Sci. 66(1) (2021) 46–52.

DOI: 10.1016/j.aoas.2021.02.001

Google Scholar

[10] T. T. Gong et al., Isothiocyanate Iberin inhibits cell proliferation and induces cell apoptosis in the progression of ovarian cancer by mediating ROS accumulation and GPX1 expression, Biomed. Pharmacother., 142 111533 (2021)

DOI: 10.1016/j.biopha.2021.111533

Google Scholar

[11] P. Pocasap, N. Weerapreeyakul, and K. Thumanu, Alyssin and iberin in cruciferous vegetables exert anticancer activity in HepG2 by increasing intracellular reactive oxygen species and tubulin depolymerization, Biomol. Ther. 27(6) (2019) 540–552

DOI: 10.4062/biomolther.2019.027

Google Scholar

[12] Y. Hosokawa et al., "The Anti-Inflammatory Effects of Iberin on TNF- α -Stimulated," Biomedicines, 10 3155 (2022)

Google Scholar

[13] A. Das, B. Bhattacharya, and S. Roy, Decrypting a path based approach for identifying the interplay between PI3K and GSK3 signaling cascade from the perspective of cancer, Genes Dis. 9(4) (2022) 868–888

DOI: 10.1016/j.gendis.2021.12.025

Google Scholar

[14] B. Moser et al., The inflammatory kinase IKKα phosphorylates and stabilizes c-Myc and enhances its activity, Mol. Cancer, 20(1) (2021) 1–17

DOI: 10.1186/s12943-021-01308-8

Google Scholar

[15] L. Xia et al., "Role of the NFκB-signaling pathway in cancer," Onco. Targets. Ther. 11 (2018) 2063–(2073)

Google Scholar

[16] J. Kang, Z. Guo, H. Zhang, R. Guo, X. Zhu, and X. Guo, Dual Inhibition of EGFR and IGF-1R Signaling Leads to Enhanced Antitumor Efficacy against Esophageal Squamous Cancer, Int. J. Mol. Sci. 23(18) (2022) 1–18

DOI: 10.3390/ijms231810382

Google Scholar

[17] A. B. Baba et al., Transforming Growth Factor-Beta (TGF-β) Signaling in Cancer-A Betrayal Within, Front. Pharmacol. 13 (2022) 1–16

Google Scholar

[18] T. Zhan, N. Rindtorff, and M. Boutros, Wnt signaling in cancer, Oncogene. 36(11) (2017) 1461–1473

DOI: 10.1038/onc.2016.304

Google Scholar

[19] F. Bianconi, E. Baldelli, V. Ludovini, L. Crinò, A. Flacco, and P. Valigi, Computational model of EGFR and IGF1R pathways in lung cancer: A Systems Biology approach for Translational Oncology, Biotechnol. Adv. 30(1) (2012) 142–153

DOI: 10.1016/j.biotechadv.2011.05.010

Google Scholar

[20] P. F. Ayodele et al., Illustrated Procedure to Perform Molecular Docking Using PyRx and Biovia Discovery Studio Visualizer: A Case Study of 10kt With Atropine, Prog. Drug Discov. Biomed. Sci. 6(1) (2023)

DOI: 10.36877/pddbs.a0000424

Google Scholar

[21] X.-Y. Meng, H.-X. Zhang, M. Mezel, and Meng Cui, Molecular Docking: A Powerful Approach for Structure Based Drug Discovery, Curr Comput Aided Drug Des. 7(2) (2011). 146–157

DOI: 10.2174/157340911795677602

Google Scholar

[22] I. Asiamah, S. A. Obiri, W. Tamekloe, F. A. Armah, and L. S. Borquaye, Applications of molecular docking in natural products-based drug discovery, Sci. African. 20 (2023) 1–8

DOI: 10.1016/j.sciaf.2023.e01593

Google Scholar

[23] T. I. Adelusi et al., Molecular modeling in drug discovery, Informatics Med. Unlocked, 29 100880 (2022)

Google Scholar

[24] I. V. Ogungbe and W. N. Setzer, The Potential of secondary metabolites from plants as drugs or leads against protozoan neglected diseases-Part III: In-Silico molecular docking investigations, Molecules. 21(10) (2016) 25] M. Zulfat et al., Identification of novel NLRP3 inhibitors as therapeutic options for epilepsy by machine learning-based virtual screening, molecular docking and biomolecular simulation studies, Heliyon. 10(15) (2024)

DOI: 10.3390/molecules21101389

Google Scholar

[26] D. A. Skoog, D. M. West, F. J. Holler, and S. R. Crouch, Fundamentals of Analytical Chemistry, 6(2) (2014).

Google Scholar

[27] R. Parasuraman, S. Balamurugan, S. Muralidharan, K. Jayaraj, V. Ijayan, and S. Anish, An Overview of Liquid Chromatography-Mass Spectroscopy Instrumentation, Pharm. Methods. 5(2) (2014) 47–55

Google Scholar

[28] C. Guan, X. Zhou, H. Li, X. Ma, and J. Zhuang, NF-κB inhibitors gifted by nature: The anticancer promise of polyphenol compounds, Biomed. Pharmacother. 156 113951 (2022)

DOI: 10.1016/j.biopha.2022.113951

Google Scholar

[29] A. Chauhan, A. U. Islam, H. Prakash, and S. Singh, Phytochemicals targeting NF-κB signaling: Potential anti-cancer interventions, J. Pharm. Anal. 12(3) (2022) 394–405

DOI: 10.1016/j.jpha.2021.07.002

Google Scholar

[30] I. Piotrowski, K. Kulcenty, and W. Suchorska, Interplay between inflammation and cancer, Reports Pract. Oncol. Radiother. 25(3) (2020), 422–427

DOI: 10.1016/j.rpor.2020.04.004

Google Scholar

[31] T. Zhang, C. Ma, Z. Zhang, H. Zhang, and H. Hu, NF-κB signaling in inflammation and cancer, MedComm. 2(4) (2021) 618–653

DOI: 10.1002/mco2.104

Google Scholar

[32] S. Shin, N. C. Ha, B. C. Oh, T. K. Oh, and B. H. Oh, Enzyme mechanism and catalytic property of β propeller phytase, Structure, 9(9) (2001) 851–858

DOI: 10.1016/s0969-2126(01)00637-2

Google Scholar

[33] Z. J. Chia, Y. N. Cao, P. J. Little, and D. Kamato, Transforming growth factor-β receptors: versatile mechanisms of ligand activation, Acta Pharmacol. Sin. 45(7) (2024) 1337–1348

DOI: 10.1038/s41401-024-01235-6

Google Scholar

[34] J. Massague, TGFβ in Cancer, Cell, 134(2) 2008 215–230

Google Scholar

[35] Y. Yang et al., The Role of TGF- β Signaling Pathways in Cancer and Its Potential as a Therapeutic Target, Evidence-based Complement. Altern. Med. (2021)

Google Scholar

[36] H. Zhao et al., Wnt signaling in colorectal cancer: pathogenic role and therapeutic target, Mol. Cancer. 21(1) (2022) 1–34

Google Scholar

[37] R. Mancinelli et al., Multifaceted roles of GSK-3 in cancer and autophagy-related diseases, Oxid. Med. Cell. Longev. (2017)

Google Scholar

[38] Q. Liu, S. Yu, W. Zhao, S. Qin, Q. Chu, and K. Wu, EGFR-TKIs resistance via EGFR-independent signaling pathways, Mol. Cancer. 17(1) (2018) 1–9

DOI: 10.1186/s12943-018-0793-1

Google Scholar

[39] E. Alfaro-Arnedo et al., IGF1R acts as a cancer-promoting factor in the tumor microenvironment facilitating lung metastasis implantation and progression, Oncogene, 41(28) (2022) 3625–3639.

DOI: 10.1038/s41388-022-02376-w

Google Scholar