Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Translational Therapeutics

Targeting NQO1/GPX4-mediated ferroptosis by plumbagin suppresses in vitro and in vivo glioma growth

Abstract

Background

Ferroptosis has attracted increasing interest in cancer therapy. Emerging evidences suggest that naturally occurring naphthoquinones exhibit potent anti-glioma effects via various mechanisms.

Methods

The anti-glioma effects of plumbagin were evaluated by in vitro and in vivo experiments. Anti-glioma mechanism of plumbagin was studied by proteomics, flow cytometry, MDA assay, western blot, and RT-PCR. Gene knockdown/overexpression, molecular docking, PharmMappper database, and coimmunoprecipitation were used to study the targets of plumbagin.

Results

Plumbagin showed higher blood–brain barrier penetration ability than that of lapachol and shikonin and elicited significant growth inhibitory effects in vitro and in vivo. Ferroptosis was the main mechanism of plumbagin-induced cell death. Mechanistically, plumbagin significantly downregulated the protein and mRNA levels of xCT and decreased GPX4 protein levels. NAD(P)H quinone dehydrogenase 1 (NQO1) was revealed as a plumbagin predictive target using PharmMappper database and molecular docking. Plumbagin enhanced NQO1 activity and decreased xCT expression, resulting in NQO1-dependent cell death. It also induced GPX4 degradation via the lysosome pathway and caused GPX4-dependent cell death.

Conclusions

Plumbagin inhibited in vitro and in vivo glioma growth via targeting NQO1/GPX4-mediated ferroptosis, which might be developed as a novel ferroptosis inducer or anti-glioma candidate.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: PLB significantly inhibited glioma growth in vitro and in vivo.
Fig. 2: PLB induced ferroptosis in glioma cells.
Fig. 3: PLB induced ferroptosis-dependent cell death.
Fig. 4: xCT and GPX4 expression levels decreased in PLB-induced ferroptosis.
Fig. 5: PLB induced GPX4-dependent cell death by causing GPX4 degradation.
Fig. 6: PLB induced ferroptosis by targeting NQO1.

Similar content being viewed by others

Data availability

Data are also available as Supplementary Information.

References

  1. Herrlinger U, Tzaridis T, Mack F, Steinbach JP, Schlegel U, Sabel M, et al. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, phase 3 trial. Lancet. 2019;393:678–88.

    Article  CAS  Google Scholar 

  2. Hu Z, Mi Y, Qian H, Guo N, Yan A, Zhang Y, et al. A potential mechanism of temozolomide resistance in glioma–ferroptosis. Front Oncol. 2020;10:897.

    Article  Google Scholar 

  3. Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 2016;113:E4966–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q, et al. Ferroptosis and its role in diverse brain diseases. Mol Neurobiol. 2019;56:4880–93.

    Article  CAS  Google Scholar 

  5. Qu Y, Wang J, Ray PS, Guo H, Huang J, Shin-Sim M, et al. Thioredoxin-like 2 regulates human cancer cell growth and metastasis via redox homeostasis and NF-κB signaling. J Clin Investig. 2011;121:212–25.

    Article  CAS  Google Scholar 

  6. Yuzbasioglu Baran M, Guvenalp Z, Saracoglu I, Kazaz C, Salih B, Demirezer LO, et al. Cytotoxic naphthoquinones from Arnebia densiflora (Nordm.) Ledeb and determining new apoptosis inducers. Nat Prod Res. 2020;34:1669–77.

    Article  CAS  Google Scholar 

  7. Aminin D, Polonik S. 1,4-Naphthoquinones: some biological properties and application. Chem Pharm Bull. 2020;68:46–57.

    Article  Google Scholar 

  8. Redaelli M, Mucignat-Caretta C, Isse AA, Gennaro A, Pezzani R, Pasquale R, et al. New naphthoquinone derivatives against glioma cells. Eur J Med Chem. 2015;96:458–66.

    Article  CAS  Google Scholar 

  9. Xu H, Chen Q, Wang H, Xu P, Yuan R, Li X, et al. Inhibitory effects of lapachol on rat C6 glioma in vitro and in vivo by targeting DNA topoisomerase I and topoisomerase II. J Exp Clin Cancer Res. 2016;35:178.

    Article  Google Scholar 

  10. Yin Z, Zhang J, Chen L, Guo Q, Yang B, Zhang W, et al. Anticancer effects and mechanisms of action of plumbagin: review of research advances. Biomed Res Int. 2020;2020:6940953.

    PubMed  PubMed Central  Google Scholar 

  11. Cao YY, Yu J, Liu TT, Yang K, Yang L, Chen Q, et al. Plumbagin inhibits the proliferation and survival of esophageal cancer cells by blocking STAT3-PLK1-AKT signaling. Cell Death Dis. 2018;9:17.

    Article  Google Scholar 

  12. Liang Y, Zhou R, Liang X, Kong X, Yang B. Pharmacological targets and molecular mechanisms of plumbagin to treat colorectal cancer: a systematic pharmacology study. Eur J Pharmacol. 2020;881:173227.

    Article  CAS  Google Scholar 

  13. Li N, Ou J, Bao N, Chen C, Shi Z, Chen L, et al. Design, synthesis and biological evaluation of novel plumbagin derivatives as potent antitumor agents with STAT3 inhibition. Bioorg Chem. 2020;2020:104208.

    Article  Google Scholar 

  14. Yang T, Zang D, Shan W, Guo A, Wu J, Wang YJ, et al. Synthesis and evaluations of novel apocynin derivatives as anti-glioma agents. Front Pharmacol. 2019;10:951.

    Article  CAS  Google Scholar 

  15. Lu L, Zhan S, Liu X, Zhao X, Lin X, Xu H. Antitumor effects and the compatibility mechanisms of herb pair Scleromitrion diffusum (Willd.) R. J. Wang–Sculellaria barbata D. Don. Front Pharmacol. 2020;11:292.

    Article  CAS  Google Scholar 

  16. Li FD, Yang YC, Li Y, Yang H, Wang H. Quantitative analysis of the global proteome in peripheral blood mononuclear cells from patients with new-onset psoriasis. Proteomics. 2018;18:e1800003.

    Article  Google Scholar 

  17. Wang X, Shen Y, Wang S, Li S, Zhang W, Liu X, et al. PharmMapper 2017 update: a web server for potential drug target identification with a comprehensive target pharmacophore database. Nucleic Acids Res. 2017;45:W356–360.

    Article  CAS  Google Scholar 

  18. Wu Z, Geng Y, Lu X, Shi Y, Wu G, Zhang M, et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci USA. 2019;116:2996–3005.

    Article  CAS  Google Scholar 

  19. Song X, Long D. Nrf2 and ferroptosis: a new research direction for neurodegenerative diseases. Front Neurosci. 2020;14:267.

    Article  Google Scholar 

  20. Yu K, Wang Z, Wu Z, Tan H, Mishra A, Peng J. High-throughput profiling of proteome and posttranslational modifications by 16-Plex TMT labeling and mass spectrometry. Methods Mol Biol. 2021;2228:205–24.

    Article  CAS  Google Scholar 

  21. Battaglia AM, Chirillo R, Aversa I, Sacco A, Costanzo F, Biamonte F. Ferroptosis and cancer: mitochondria meet the “iron maiden” cell death. Cells. 2020;9:1505.

    Article  CAS  Google Scholar 

  22. Conrad M, Pratt DA. The chemical basis of ferroptosis. Nat Chem Biol. 2019;15:1137–47.

    Article  CAS  Google Scholar 

  23. Zou Y, Palte MJ, Deik AA, Li H, Eaton K, Wang W, et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat Commun. 2019;10:1617.

    Article  Google Scholar 

  24. Alexiou GA, Gerogianni P, Vartholomatos E, Kyritsis AP. Deferiprone enhances temozolomide cytotoxicity in glioma cells. Cancer Invest. 2016;34:489–95.

    Article  CAS  Google Scholar 

  25. Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575:693–8.

    Article  CAS  Google Scholar 

  26. Gong Q, Hu J, Wang P, Li X, Zhang X. A comprehensive review on beta-lapachone: mechanisms, structural modifications, and therapeutic potentials. Eur J Med Chem. 2021;210:112962.

    Article  CAS  Google Scholar 

  27. Li X, Liu Z, Zhang A, Han C, Shen A, Jiang L, et al. NQO1 targeting prodrug triggers innate sensing to overcome checkpoint blockade resistance. Nat Commun. 2019;10:3251.

    Article  Google Scholar 

  28. Zhang K, Chen D, Ma K, Wu X, Hao H, Jiang S. NAD(P)H:Quinone oxidoreductase 1 (NQO1) as a therapeutic and diagnostic target in cancer. J Med Chem. 2018;61:6983–7003.

    Article  CAS  Google Scholar 

  29. Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–31.

    Article  CAS  Google Scholar 

  30. Chiang HL, Terlecky SR, Plant CP, Dice JF. A role for a 70-kilodalton heat shock protein in lysosomal degradation of intracellular proteins. Science. 1989;246:382–5.

    Article  CAS  Google Scholar 

  31. Cuervo AM, Dice JF. A receptor for the selective uptake and degradation of proteins by lysosomes. Science. 1996;273:501–3.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Mrs. Yang Hui in core facility centre of Capital Medical University for their technique assistance in transmission electron microscopy analysis.

Funding

This study was supported by grants from the National Natural Science Foundation of China (Nos. 81774191 and 82174265), the International Collaborative of Ministry of Science and Technology (No. 2017YEE0915000), and National Science Foundation of Shandong Province in China (Nos. ZR201911020118 and ZR201911110299).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualisation, H-lX, X-kL, MX; methodology, SZ, LL, R-rM, X-qW, S-sP; formal analysis, SZ, H-lX; writing—original draft preparation, SZ, LL; writing—review and editing, H-lX, X-kL, MX; funding acquisition, H-lX, X-kL. All authors have approved the final article.

Corresponding author

Correspondence to Huan-li Xu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

All animal experiments were approved by the Committee of Animal Experiments and Experimental Animal Welfare of Capital Medical University in Beijing, China (Nos. AEEI-2019078 and 37363) and performed in accordance with the National Institutes of Health guide for the care and use of laboratory animals (NIH Publications No. 8023, revised 1978).

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhan, S., Lu, L., Pan, Ss. et al. Targeting NQO1/GPX4-mediated ferroptosis by plumbagin suppresses in vitro and in vivo glioma growth. Br J Cancer 127, 364–376 (2022). https://doi.org/10.1038/s41416-022-01800-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-022-01800-y

This article is cited by

Search

Quick links