β-elemene promotes ferroptosis to improve the sensitivity of imatinib in gastrointestinal stromal tumours by targeting N6AMT1

Jin Lan , Weili Zhang , Kaixuan Zeng , Cong Li , Jiahua He , Xinyue Li , Rong Yang , Jun Chi , Zhigang Hong , Weifeng Wang , Chi Zhou , Binyi Xiao , Wenhua Fan , Junzhong Lin , Qingjian Ou , Yujing Fang , Zhizhong Pan , Jianhong Peng , Weihao Li , Xiaojun Wu

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (9) : e70438

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (9) : e70438 DOI: 10.1002/ctm2.70438
RESEARCH ARTICLE

β-elemene promotes ferroptosis to improve the sensitivity of imatinib in gastrointestinal stromal tumours by targeting N6AMT1

Author information +
History +
PDF

Abstract

Background: Imatinib has been widely used in gastrointestinal stromal tumours and significantly improved the prognosis of GIST patients, but approximately half of patients develop acquired treatment resistance, highlighting the urgency for novel therapeutic strategies.

Methods: A variety of bioinformatic tools and laboratory experiments, RNA sequencing, animal models and the thermal proteome profiling assay were employed to validate our findings and investigate the antitumour effects of β-elemene.

Results: We found that imatinib-resistant GIST was associated with negative regulation of ferroptosis activity, and inducing ferroptosis can enhance the sensitivity of resistant cells to imatinib. Furthermore, we found that β-elemene enhances imatinib sensitivity in imatinib-resistant GIST cells through inducing ferroptosis. Moreover, the combination treatment of β-elemene and imatinib showed significantly increased antitumour efficacy, compared to each monotherapy, both in vitro and in vivo. Mechanistically, β-elemene specifically targets N6AMT1, inhibiting its transcriptional repression function and activating the nuclear factor erythroid 2-related factor 2 (NRF2)-HMOX1 signalling pathway to induce ferroptosis.

Conclusion: Β-elemene can target N6AMT1 and promote ferroptosis by increasing the expression of NRF2 and HMOX1. These findings suggest β-elemene as a prospective therapeutic strategy to improve the sensitivity of imatinib in gastrointestinal stromal tumours.

Keywords

ferroptosis / gastrointestinal stromal tumours / imatinib resistance / ROS / β-elemene

Cite this article

Download citation ▾
Jin Lan, Weili Zhang, Kaixuan Zeng, Cong Li, Jiahua He, Xinyue Li, Rong Yang, Jun Chi, Zhigang Hong, Weifeng Wang, Chi Zhou, Binyi Xiao, Wenhua Fan, Junzhong Lin, Qingjian Ou, Yujing Fang, Zhizhong Pan, Jianhong Peng, Weihao Li, Xiaojun Wu. β-elemene promotes ferroptosis to improve the sensitivity of imatinib in gastrointestinal stromal tumours by targeting N6AMT1. Clinical and Translational Medicine, 2025, 15(9): e70438 DOI:10.1002/ctm2.70438

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Demetri GD, von Mehren M, Antonescu CR, et al. NCCN Task Force report: update on the management of patients with gastrointestinal stromal tumors. J Natl Compr Canc Netw. 2010; 8(suppl 2)(0 2): S1-41.

[2]

de Pinieux G, Karanian M, Le Loarer F, et al. Nationwide incidence of sarcomas and connective tissue tumors of intermediate malignancy over four years using an expert pathology review network. PLoS One. 2021; 16(2): e0246958.

[3]

Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science. 2003; 299(5607): 708-710.

[4]

Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med. 2002; 347(7): 472-480.

[5]

Kee D, Zalcberg JR. Current and emerging strategies for the management of imatinib-refractory advanced gastrointestinal stromal tumors. Ther Adv Med Oncol. 2012; 4(5): 255-270.

[6]

Gramza AW, Corless CL, Heinrich MC. Resistance to tyrosine kinase inhibitors in gastrointestinal stromal tumors. Clin Cancer Res. 2009; 15(24): 7510-7518.

[7]

Wardelmann E, Thomas N, Merkelbach-Bruse S, et al. Acquired resistance to imatinib in gastrointestinal stromal tumours caused by multiple KIT mutations. Lancet Oncol. 2005; 6(4): 249-251.

[8]

Klug LR, Khosroyani HM, Kent JD, Heinrich MC. New treatment strategies for advanced-stage gastrointestinal stromal tumours. Nat Rev Clin Oncol. 2022; 19(5): 328-341.

[9]

Debiec-Rychter M, Cools J, Dumez H, et al. Mechanisms of resistance to imatinib mesylate in gastrointestinal stromal tumors and activity of the PKC412 inhibitor against imatinib-resistant mutants. Gastroenterology. 2005; 128(2): 270-279.

[10]

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144(5): 646-674.

[11]

Yu H, Guo P, Xie X, Wang Y, Chen G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J Cell Mol Med. 2017; 21(4): 648-657.

[12]

Xu T, Ding W, Ji X, et al. Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med. 2019; 23(8): 4900-4912.

[13]

Zhu T, Shi L, Yu C, et al. Ferroptosis promotes photodynamic therapy: supramolecular photosensitizer-inducer nanodrug for enhanced cancer treatment. Theranostics. 2019; 9(11): 3293-3307.

[14]

Ye Z, Hu Q, Zhuo Q, et al. Abrogation of ARF6 promotes RSL3-induced ferroptosis and mitigates gemcitabine resistance in pancreatic cancer cells. Am J Cancer Res. 2020; 10(4): 1182-1193.

[15]

Hong T, Lei G, Chen X, et al. PARP inhibition promotes ferroptosis via repressing SLC7A11 and synergizes with ferroptosis inducers in BRCA-proficient ovarian cancer. Redox Biol. 2021; 42: 101928.

[16]

Zhai B, Zhang N, Han X, et al. Molecular targets of beta-elemene, a herbal extract used in traditional Chinese medicine, and its potential role in cancer therapy: a review. Biomed Pharmacother. 2019; 114: 108812.

[17]

Gong M, Liu Y, Zhang J, et al. Beta-elemene inhibits cell proliferation by regulating the expression and activity of topoisomerases I and IIalpha in human hepatocarcinoma HepG-2 cells. Biomed Res Int. 2015; 2015: 153987.

[18]

Yao C, Jiang J, Tu Y, Ye S, Du H, Zhang Y. Beta-elemene reverses the drug resistance of A549/DDP lung cancer cells by activating intracellular redox system, decreasing mitochondrial membrane potential and P-glycoprotein expression, and inducing apoptosis. Thorac Cancer. 2014; 5(4): 304-312.

[19]

Pan Y, Wang W, Huang S, et al. Beta-elemene inhibits breast cancer metastasis through blocking pyruvate kinase M2 dimerization and nuclear translocation. J Cell Mol Med. 2019; 23(10): 6846-6858.

[20]

Liu S, Li Q, Li G, et al. The mechanism of m(6)A methyltransferase METTL3-mediated autophagy in reversing gefitinib resistance in NSCLC cells by beta-elemene. Cell Death Dis. 2020; 11(11): 969.

[21]

Mu L, Wang T, Chen Y, Tang X, Yuan Y, Zhao Y. Beta-elemene enhances the efficacy of gefitinib on glioblastoma multiforme cells through the inhibition of the EGFR signaling pathway. Int J Oncol. 2016; 49(4): 1427-1436.

[22]

Chen P, Li X, Zhang R, et al. Combinative treatment of beta-elemene and cetuximab is sensitive to KRAS mutant colorectal cancer cells by inducing ferroptosis and inhibiting epithelial-mesenchymal transformation. Theranostics. 2020; 10(11): 5107-5119.

[23]

Savitski MM, Reinhard FB, Franken H, et al. Tracking cancer drugs in living cells by thermal profiling of the proteome. Science. 2014; 346(6205): 1255784.

[24]

Seeliger D, de Groot BL. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J Comput Aided Mol Des. 2010; 24(5): 417-422.

[25]

Eberhardt J, Santos-Martins D, Tillack AF, Forli S. AutoDock Vina 1.2.0: new docking methods, expanded force field, and Python bindings. J Chem Inf Model. 2021; 61(8): 3891-3898.

[26]

Cheng X, Yan J, Liu Y, Wang J, Taubert S. eVITTA: a web-based visualization and inference toolbox for transcriptome analysis. Nucleic Acids Res. 2021; 49(W1): W207-W215.

[27]

Lu L, Katsaros D, de la Longrais IA, Sochirca O, Yu H. Hypermethylation of let-7a-3 in epithelial ovarian cancer is associated with low insulin-like growth factor-II expression and favorable prognosis. Cancer Res. 2007; 67(21): 10117-10122.

[28]

Delvaux M, Hague P, Craciun L, et al. Ferroptosis induction and YAP inhibition as new therapeutic targets in gastrointestinal stromal tumors (GISTs). Cancers (Basel). 2022; 14(20): 5050.

[29]

Ishida T, Takahashi T, Kurokawa Y, et al. Targeted therapy for drug-tolerant persister cells after imatinib treatment for gastrointestinal stromal tumours. Br J Cancer. 2021; 125(11): 1511-1522.

[30]

Cui Z, Sun H, Gao Z, et al. TRIM21/USP15 balances ACSL4 stability and the imatinib resistance of gastrointestinal stromal tumors. Br J Cancer. 2024; 130(4): 526-541.

[31]

Sun X, Zhang Q, Lin X, Shu P, Gao X, Shen K. Imatinib induces ferroptosis in gastrointestinal stromal tumors by promoting STUB1-mediated GPX4 ubiquitination. Cell Death Dis. 2023; 14(12): 839.

[32]

Zhang C, Liu X, Jin S, Chen Y, Guo R. Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer. 2022; 21(1): 47.

[33]

Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012; 149(5): 1060-1072.

[34]

Chang LC, Chiang SK, Chen SE, Yu YL, Chou RH, Chang WC. Heme oxygenase-1 mediates BAY 11–7085 induced ferroptosis. Cancer Lett. 2018; 416: 124-137.

[35]

Guo Z, Zhang W, Gao H, et al. High expression levels of haem oxygenase-1 promote ferroptosis in macrophage-derived foam cells and exacerbate plaque instability. Redox Biol. 2024; 76: 103345.

[36]

Zheng J, Conrad M. The metabolic underpinnings of ferroptosis. Cell Metab. 2020; 32(6): 920-937.

[37]

Kong J, Lyu H, Ouyang Q, et al. Insights into the roles of epigenetic modifications in ferroptosis. Biology (Basel). 2024; 13(2).

[38]

Zhang C, Yang T, Chen H, et al. METTL3 inhibition promotes radiosensitivity in hepatocellular carcinoma through regulation of SLC7A11 expression. Cell Death Dis. 2025; 16(1): 9.

[39]

Tang D, Chen X, Kang R, Kroemer G. Ferroptosis: molecular mechanisms and health implications. Cell Res. 2021; 31(2): 107-125.

[40]

Yang C, Wang T, Zhao Y, et al. Flavonoid 4,4'-dimethoxychalcone induced ferroptosis in cancer cells by synergistically activating Keap1/Nrf2/HMOX1 pathway and inhibiting FECH. Free Radic Biol Med. 2022; 188: 14-23.

[41]

Li W, Shi Y, Zhang T, Ye J, Ding J. Structural insight into human N6amt1-Trm112 complex functioning as a protein methyltransferase. Cell Discov. 2019; 5: 51.

[42]

Sun Y, Qiao Y, Liu Y, et al. ent-Kaurane diterpenoids induce apoptosis and ferroptosis through targeting redox resetting to overcome cisplatin resistance. Redox Biol. 2021; 43: 101977.

[43]

Fu D, Wang C, Yu L, Yu R. Induction of ferroptosis by ATF3 elevation alleviates cisplatin resistance in gastric cancer by restraining Nrf2/Keap1/xCT signaling. Cell Mol Biol Lett. 2021; 26(1): 26.

[44]

Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019; 19(7): 405-414.

[45]

Adedoyin O, Boddu R, Traylor A, et al. Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol Renal Physiol. 2018; 314(5): F702-F714.

[46]

Stockwell BR, Jiang X, Gu W. Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol. 2020; 30(6): 478-490.

[47]

Totsuka K, Ueta T, Uchida T, et al. Oxidative stress induces ferroptotic cell death in retinal pigment epithelial cells. Exp Eye Res. 2019; 181: 316-324.

[48]

Pei Y, Qian Y, Wang H, Tan L. Epigenetic regulation of ferroptosis-associated genes and its implication in cancer therapy. Front Oncol. 2022; 12: 771870.

[49]

Yang H, Hu Y, Weng M, et al. Hypoxia inducible lncRNA-CBSLR modulates ferroptosis through m6A-YTHDF2-dependent modulation of CBS in gastric cancer. J Adv Res. 2022; 37: 91-106.

[50]

Lee J, You JH, Kim MS, Roh JL. Epigenetic reprogramming of epithelial-mesenchymal transition promotes ferroptosis of head and neck cancer. Redox Biol. 2020; 37: 101697.

[51]

Chen J, Zhuang Y, Wang P, et al. Reducing N6AMT1-mediated 6mA DNA modification promotes breast tumor progression via transcriptional repressing cell cycle inhibitors. Cell Death Dis. 2022; 13(3): 216.

[52]

Baumert HM, Metzger E, Fahrner M, et al. Depletion of histone methyltransferase KMT9 inhibits lung cancer cell proliferation by inducing non-apoptotic cell death. Cancer Cell Int. 2020; 20: 52.

[53]

Kusevic D, Kudithipudi S, Jeltsch A. Substrate specificity of the HEMK2 protein glutamine methyltransferase and identification of novel substrates. J Biol Chem. 2016; 291(12): 6124-6133.

[54]

Diaz MO, Ziemin S, Le Beau MM, et al. Homozygous deletion of the alpha- and beta 1-interferon genes in human leukemia and derived cell lines. Proc Natl Acad Sci U S A. 1988; 85(14): 5259-5263.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

6

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/