PDF
Abstract
Background: Indole phytoalexins, plant-derived compounds present in cruciferous vegetables, have demonstrated anticancer properties. Brassinin (BSN), derived from Brassica campestris L. var. campestris, is known for its potent antitumor effects on various cancers. However, the role of ferroptosis in regulating the antitumor effects of BSN has not been fully elucidated.
Methods: The components of B. campestris L. against colorectal cancer (CRC) were analyzed by network pharmacology. CCK-8 assay and colony formation assay detected cell viability induced by BSN. Molecular docking verified the binding of BSN to the target protein. Western blot and reverse transcription–quantitative polymerase chain reaction (RT-qPCR) assay revealed whether BSN can inactivate the NRF2 signaling and inhibit the expression of p62 and HO-1. The RKO-xenograft tumor models were established and then were treated by 75 or 150 mg/kg BSN to verify the antitumor efficacy and side effects of BSN.
Results: Network pharmacology suggested that BSN is the most important component of B. campestris L. against CRC. BSN inhibits CRC cell viability in a dose- and time-dependent manner. Furthermore, this inhibitory effect is associated with the induction of ferroptosis, as BSN suppresses the cell viability of CRC by inducing GPX4-regulated ferroptosis. BSN may bind to NRF2 protein to inactivate the NRF2 signaling, inhibiting the expression of p62 and HO-1. Importantly, a low dose or a high dose of BSN significantly reduced the tumor growth in vivo.
Conclusions: Our findings reveal that BSN blocks CRC growth by inducing p62/NRF2/GPX4-regulated ferroptosis, which may be a novel lead compound for tumor treatment.
Keywords
Brassinin
/
colorectal cancer
/
ferroptosis
/
GPX4
/
NRF2
Cite this article
Download citation ▾
Shi-Yuan Wen, Rui-Rui Gao, Yan-Yan Chen, Yi-Jie Wang, Xin-Tong Wang, Hai-Xin Liu.
Brassinin from Brassica campestris L. inhibits colorectal cancer by inducing p62/NRF2/GPX4-regulated ferroptosis.
Animal Models and Experimental Medicine, 2025, 8(7): 1155-1165 DOI:10.1002/ame2.12521
| [1] |
Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019; 394(10207): 1467-1480.
|
| [2] |
Tang R, Sansonetti PJ, Gao Y-Z. Stem cell retrograde: a new reason why colorectal cancer is more common than small intestinal cancer? Innovation (Camb). 2023; 4(2): 100387.
|
| [3] |
Chai C, Tang X, Chi X, Li X, Zhang H, Wu L. Therapeutic effects of coptisine derivative EHLJ7 on colorectal cancer by inhibiting PI3K/AKT pathway. Cell Signal. 2024; 116: 111053.
|
| [4] |
Tjader NP, Toland AE. Immunotherapy for colorectal cancer: insight from inherited genetics. Trends Cancer. 2024; 10: 444-456.
|
| [5] |
Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021; 22(4): 266-282.
|
| [6] |
Liu X, Yan C, Chang C, et al. FOXA2 suppression by TRIM36 exerts anti-tumor role in colorectal cancer via inducing NRF2/GPX4-regulated Ferroptosis. Adv Sci (Weinh). 2023; 10(35): e2304521.
|
| [7] |
Zhu J-F, Liu Y, Li W-T, et al. Ibrutinib facilitates the sensitivity of colorectal cancer cells to ferroptosis through BTK/NRF2 pathway. Cell Death Dis. 2023; 14(2): 151.
|
| [8] |
Sadeghi MR, Jeddi F, Soozangar N, Somi MH, Samadi N. The role of Nrf2-Keap1 axis in colorectal cancer, progression, and chemoresistance. Tumour Biol. 2017; 39(6): 1010428317705510.
|
| [9] |
Kopacz A, Kloska D, Forman HJ, Jozkowicz A, Grochot-Przeczek A. Beyond repression of Nrf2: an update on Keap1. Free Radic Biol Med. 2020; 157: 63-74.
|
| [10] |
Huang Y, Yang W, Yang L, et al. Nrf2 inhibition increases sensitivity to chemotherapy of colorectal cancer by promoting ferroptosis and pyroptosis. Sci Rep. 2023; 13(1): 14359.
|
| [11] |
Pandurangan AK, Divya T, Kumar K, Dineshbabu V, Velavan B, Sudhandiran G. Colorectal carcinogenesis: insights into the cell death and signal transduction pathways: a review. World J Gastrointest Oncol. 2018; 10(9): 244-259.
|
| [12] |
Donadio JLS, Prado SBRD, Soares CG, et al. Ripe papaya pectins inhibit the proliferation of colon cancer spheroids and the formation of chemically induced aberrant crypts in rats colons. Carbohydr Polym. 2024; 331: 121878.
|
| [13] |
Chripkova M, Zigo F, Mojzis J. Antiproliferative effect of indole Phytoalexins. Molecules. 2016; 21(12): 1626.
|
| [14] |
Tischlerova V, Kello M, Budovska M, Mojzis J. Indole phytoalexin derivatives induce mitochondrial-mediated apoptosis in human colorectal carcinoma cells. World J Gastroenterol. 2017; 23(24): 4341-4353.
|
| [15] |
Yamashita N, Taga C, Ozawa M, Kanno Y, Sanada N, Kizu R. Camalexin, an indole phytoalexin, inhibits cell proliferation, migration, and mammosphere formation in breast cancer cells via the aryl hydrocarbon receptor. J Nat Med. 2022; 76(1): 110-118.
|
| [16] |
Takasugi M, Katsui N, Shirata A. Isolation of three novel sulphur-containing phytoalexins from the chinese cabbage Brassica campestris L. ssp. pekinensis. J Chem Soc Chem Commun. 1986; 14: 1077-1078.
|
| [17] |
Yang MH, Lee JH, Ko J-H, Jung SH, Sethi G, Ahn KS. Brassinin represses invasive potential of lung carcinoma cells through deactivation of PI3K/Akt/mTOR signaling Cascade. Molecules. 2019; 24(8): 1584.
|
| [18] |
Kim S-M, Oh EY, Lee JH, et al. Brassinin combined with capsaicin enhances apoptotic and anti-metastatic effects in PC-3 human prostate cancer cells. Phytother Res. 2015; 29(11): 1828-1836.
|
| [19] |
Hong T, Ham J, Song J, Song G, Lim W. Brassinin inhibits proliferation in human liver cancer cells via mitochondrial dysfunction. Cells. 2021; 10(2): 332.
|
| [20] |
Wen S-Y, Chen Y-Y, Deng C-M, Zhang C-Q, Jiang M-M. Nerigoside suppresses colorectal cancer cell growth and metastatic potential. Phytomedicine. 2019; 57: 352-363.
|
| [21] |
Wen SY, Liu YT, Wei BY, Ma JQ, Chen YY. PDCD6 promotes hepatocellular carcinoma cell proliferation and metastasis through the AKT/GSK3β/β-catenin pathway. Biomed Environ Sci. 2023; 36(3): 241-252.
|
| [22] |
Gu Y, Becker V, Qiu M, et al. Brassinin promotes the degradation of Tie2 and FGFR1 in endothelial cells and inhibits triple-negative breast cancer angiogenesis. Cancers (Basel). 2022; 14(14): 4530.
|
| [23] |
Izutani Y, Yogosawa S, Sowa Y, Sakai T. Brassinin induces G1 phase arrest through increase of p21 and p27 by inhibition of the phosphatidylinositol 3-kinase signaling pathway in human colon cancer cells. Int J Oncol. 2012; 40(3): 816-824.
|
| [24] |
Kello M, Drutovic D, Chripkova M, et al. ROS-dependent antiproliferative effect of brassinin derivative homobrassinin in human colorectal cancer Caco2 cells. Molecules. 2014; 19(8): 10877-10897.
|
| [25] |
Yang MH, Ha IJ, Lee S-G, et al. Brassinin induces apoptosis, autophagy, and Paraptosis via MAPK signaling pathway activation in chronic Myelogenous leukemia cells. Biology (Basel). 2023; 12(2): 307.
|
| [26] |
Wang T, Zhou Z, Wang C, et al. LTBP2 knockdown promotes Ferroptosis in gastric cancer cells through p62-Keap1-Nrf2 pathway. Biomed Res Int. 2022; 2022: 6532253.
|
| [27] |
Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019; 23: 101107.
|
| [28] |
Saigusa D, Motoike IN, Saito S, et al. Impacts of NRF2 activation in non-small-cell lung cancer cell lines on extracellular metabolites. Cancer Sci. 2020; 111(2): 667-678.
|
| [29] |
Gonzalez-Donquiles C, Alonso-Molero J, Fernandez-Villa T, Vilorio-Marqués L, Molina AJ, Martín V. The NRF2 transcription factor plays a dual role in colorectal cancer: a systematic review. PLoS One. 2017; 12(5): e0177549.
|
| [30] |
Cordaro M, D'Amico R, Morabito R, et al. Physiological and biochemical changes in NRF2 pathway in aged animals subjected to brain injury. Cell Physiol Biochem. 2021; 55(2): 160-179.
|
| [31] |
Hirschhorn T, Stockwell BR. The development of the concept of ferroptosis. Free Radic Biol Med. 2019; 133: 130-143.
|
| [32] |
Zhang J, Yang S, Xu B, et al. p62 functions as an oncogene in colorectal cancer through inhibiting apoptosis and promoting cell proliferation by interacting with the vitamin D receptor. Cell Prolif. 2019; 52(3): e12585.
|
| [33] |
Umemura A, He F, Taniguchi K, et al. p62, upregulated during Preneoplasia, Induces Hepatocellular Carcinogenesis by Maintaining Survival of Stressed HCC-Initiating Cells. Cancer Cell. 2016; 29(6): 935-948.
|
RIGHTS & PERMISSIONS
2025 The Author(s). Animal Models and Experimental Medicine published by John Wiley & Sons Australia, Ltd on behalf of The Chinese Association for Laboratory Animal Sciences.