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:

Celastrol inhibits the DPYSL2-JAK/STAT pathway by targeting mito-IDHs mediated mitochondrial metabolism to exhaust breast cancer

Abstract

Targeting mitochondrial metabolism represents a novel approach in the discovery and development of anti-tumor drugs. Celastrol (Cel) is a naturally-derived small molecule from Tripterygium wilfordii with significant anticancer activities. In this study, we investigated the anti-tumor mechanisms involving mitochondrial metabolic reprogramming regulated by Cel in breast cancer (BRCA). We showed that Cel potently inhibited the proliferation of triple-negative breast cancer cells (MDA-MB-231) and ER+ breast cancer cells (MCF-7) with IC50 values of 2.15 μM and 2.29 μM, respectively. Administration of Cel (5, 2, 2 mg/kg, i.p. for three times after tumor formation) significantly suppressed the tumor growth in syngeneic allograft and CDX breast cancer mouse models. Using activity-based protein profiling (ABPP) technology, we identified mitochondrial isocitrate dehydrogenases (including IDH2 and IDH3A, collectively referred to as mito-IDHs) as direct targets of Cel. We demonstrated that Cel significantly inhibited mito-IDHs mediated mitochondrial metabolism to induce the accumulation of metabolites α-ketoglutaric acid, and that Cel enhanced the interaction between DPYSL2 with IDH3A while promoting the accumulation of DPYSL2 within mitochondria of BRCA cells resulting in inactivation of JAK/STAT pathway and ultimately induced ferroptosis and apoptosis in cancer cells. Collectively, this study elucidates a pharmacological mechanism by which Cel exerts its tumor-inhibiting effects through modulation of mitochondrial metabolism. Furthermore, it provides compelling evidence supporting Cel as a promising candidate for development as a small-molecule inhibitor targeting mitochondrial metabolism.

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: Cel effectively inhibits BRCA progression at both in vitro cell levels and in vivo mice levels.
Fig. 2: IDH3A is a direct target of Cel.
Fig. 3: Cel covalently targets Cys127 residues of IDH3A.
Fig. 4: Cel targets mito-IDHs to induce α-KG accumulation in BRCA cells and inhibit cell proliferation.
Fig. 5: Cel inhibits mito-IDHs-mediated mitochondrial metabolism.
Fig. 6: Cel promotes the interaction between DPYSL2 and IDH3A in mitochondria and reduces the cytoplasmic distribution.
Fig. 7: Cel targets mito-IDHs to promote ferroptosis and apoptosis of BRCA cells.

Similar content being viewed by others

References

  1. Abdel-Wahab AF, Mahmoud W, Al-Harizy RM. Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharmacol Res. 2019;150:104511.

    Article  PubMed  CAS  Google Scholar 

  2. Weinberg SE, Chandel NS. Targeting mitochondria metabolism for cancer therapy. Nat Chem Biol. 2015;11:9–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Moreno-Sanchez R, Rodriguez-Enriquez S, Marin-Hernandez A, Saavedra E. Energy metabolism in tumor cells. FEBS J. 2007;274:1393–418.

    Article  PubMed  CAS  Google Scholar 

  4. Momcilovic M, Jones A, Bailey ST, Waldmann CM, Li R, Lee JT, et al. Publisher Correction: In vivo imaging of mitochondrial membrane potential in non-small-cell lung cancer. Nature. 2020;577:E7.

    Article  PubMed  CAS  Google Scholar 

  5. May JL, Kouri FM, Hurley LA, Liu J, Tommasini-Ghelfi S, Ji Y, et al. IDH3alpha regulates one-carbon metabolism in glioblastoma. Sci Adv. 2019;5:eaat0456.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Du B, Sun T, Li X, Diao Y, Li Y. Effect of IDH3a on glucose uptake in lung adenocarcinoma: A pilot study based on [18F]FDG. Cancer Med. 2019;8:5341–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Liu X, Qiao Y, Ting X, Si W. Isocitrate dehydrogenase 3A, a rate-limiting enzyme of the TCA cycle, promotes hepatocellular carcinoma migration and invasion through regulation of MTA1, a core component of the NuRD complex. Am J Cancer Res. 2020;10:3212–29.

    PubMed  PubMed Central  CAS  Google Scholar 

  8. Li JJ, Yu T, Zeng P, Tian J, Liu P, Qiao S, et al. Wild-type IDH2 is a therapeutic target for triple-negative breast cancer. Nat Commun. 2024;15:3445.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Zeng L, Morinibu A, Kobayashi M, Zhu Y, Wang X, Goto Y, et al. Aberrant IDH3alpha expression promotes malignant tumor growth by inducing HIF-1-mediated metabolic reprogramming and angiogenesis. Oncogene. 2015;34:4758–66.

    Article  PubMed  CAS  Google Scholar 

  10. Lim HY, Ong PS, Wang L, Goel A, Ding L, Li-Ann Wong A, et al. Celastrol in cancer therapy: recent developments, challenges and prospects. Cancer Lett. 2021;521:252–67.

    Article  PubMed  CAS  Google Scholar 

  11. Wang J, Zhang CJ, Chia WN, Loh CC, Li Z, Lee YM, et al. Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum. Nat Commun. 2015;6:10111.

    Article  PubMed  CAS  Google Scholar 

  12. Luo P, Liu D, Zhang Q, Yang F, Wong YK, Xia F, et al. Celastrol induces ferroptosis in activated HSCs to ameliorate hepatic fibrosis via targeting peroxiredoxins and HO-1. Acta Pharm Sin B. 2022;12:2300–14.

    Article  PubMed  CAS  Google Scholar 

  13. Zhang Q, Luo P, Xia F, Tang H, Chen J, Zhang J, et al. Capsaicin ameliorates inflammation in a TRPV1-independent mechanism by inhibiting PKM2-LDHA-mediated Warburg effect in sepsis. Cell Chem Biol. 2022;29:1248–59.e6.

    Article  PubMed  CAS  Google Scholar 

  14. Huang L, Zhang J, Wei B, Chen S, Zhu S, Qi W, et al. Small-molecule MHC-II inducers promote immune detection and anti-cancer immunity via editing cancer metabolism. Cell Chem Biol. 2023;30:1076–89.e11.

    Article  PubMed  CAS  Google Scholar 

  15. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.

    Article  PubMed  CAS  Google Scholar 

  16. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Shin DH, Nam JH, Lee ES, Zhang Y, Kim SJ. Inhibition of Ca2+ release-activated Ca2+ channel (CRAC) by curcumin and caffeic acid phenethyl ester (CAPE) via electrophilic addition to a cysteine residue of Orai1. Biochem Biophys Res Commun. 2012;428:56–61.

    Article  PubMed  CAS  Google Scholar 

  18. Huang L, Wei B, Zhao Y, Gong X, Chen L. DYNLT1 promotes mitochondrial metabolism to fuel breast cancer development by inhibiting ubiquitination degradation of VDAC1. Mol Med. 2023;29:72.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Topf U, Suppanz I, Samluk L, Wrobel L, Boser A, Sakowska P, et al. Quantitative proteomics identifies redox switches for global translation modulation by mitochondrially produced reactive oxygen species. Nat Commun. 2018;9:324.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Chasara RS, Ajayi TO, Leshilo DM, Poka MS, Witika BA. Exploring novel strategies to improve anti-tumour efficiency: The potential for targeting reactive oxygen species. Heliyon. 2023;9:e19896.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Lin B, Li Y, Wang T, Qiu Y, Chen Z, Zhao K, et al. CRMP2 is a therapeutic target that suppresses the aggressiveness of breast cancer cells by stabilizing RECK. Oncogene. 2020;39:6024–40.

    Article  PubMed  CAS  Google Scholar 

  22. Abu Rmaileh A, Solaimuthu B, Khatib A, Lavi S, Tanna M, Hayashi A, et al. DPYSL2 interacts with JAK1 to mediate breast cancer cell migration. J Cell Biol. 2022;221:e202106078.

  23. Vasan K, Werner M, Chandel NS. Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 2020;32:341–52.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Porporato PE, Filigheddu N, Pedro JMB, Kroemer G, Galluzzi L. Mitochondrial metabolism and cancer. Cell Res. 2018;28:265–80.

    Article  PubMed  CAS  Google Scholar 

  25. Zou J, Huang R, Chen Y, Huang X, Li H, Liang P, et al. Dihydropyrimidinase like 2 promotes bladder cancer progression via pyruvate kinase M2-induced aerobic glycolysis and epithelial-mesenchymal transition. Front Cell Dev Biol. 2021;9:641432.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Zu L, He J, Zhou N, Tang Q, Liang M, Xu S. Identification of multiple organ metastasis-associated hub mRNA/miRNA signatures in non-small cell lung cancer. Cell Death Dis. 2023;14:798.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Li F, Ling Q, Lian J, Chen Y, Hu C, Yang M, et al. Dihydropyrimidinase-like 2 can serve as a novel therapeutic target and prognostic biomarker in acute myeloid leukemia. Cancer Med. 2023;12:8319–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Soleymani F, Paquet E, Viktor H, Michalowski W, Spinello D. Protein-protein interaction prediction with deep learning: a comprehensive review. Comput Struct Biotechnol J. 2022;20:5316–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Meyer LK, Verbist KC, Albeituni S, Scull BP, Bassett RC, Stroh AN, et al. JAK/STAT pathway inhibition sensitizes CD8 T cells to dexamethasone-induced apoptosis in hyperinflammation. Blood. 2020;136:657–68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Escher TE, Lui AJ, Geanes ES, Walter KR, Tawfik O, Hagan CR, et al. Interaction between MUC1 and STAT1 drives IFITM1 overexpression in aromatase inhibitor-resistant breast cancer cells and mediates estrogen-induced apoptosis. Mol Cancer Res. 2019;17:1180–94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Wang C, Dai S, Zhao X, Zhang Y, Gong L, Fu K, et al. Celastrol as an emerging anticancer agent: current status, challenges and therapeutic strategies. Biomed Pharmacother. 2023;163:114882.

    Article  PubMed  CAS  Google Scholar 

  32. Yu X, Zhu D, Luo B, Kou W, Cheng Y, Zhu Y. IFNgamma enhances ferroptosis by increasing JAK‑STAT pathway activation to suppress SLCA711 expression in adrenocortical carcinoma. Oncol Rep. 2022;47:97.

  33. Kong R, Wang N, Han W, Bao W, Lu J. IFNgamma-mediated repression of system xc(-) drives vulnerability to induced ferroptosis in hepatocellular carcinoma cells. J Leukoc Biol. 2021;110:301–14.

    Article  PubMed  CAS  Google Scholar 

  34. Liu T, Li Q, Xu X, Li G, Tian C, Zhang T. Molecular mechanisms of anti-cancer bioactivities of seaweed polysaccharides. Chin Herb Med. 2022;14:528–34.

    PubMed  PubMed Central  CAS  Google Scholar 

  35. Kong F, Wang C, Zhang J, Wang X, Sun B, Xiao X, et al. Chinese herbal medicines for prostate cancer therapy: From experimental research to clinical practice. Chin Herb Med. 2023;15:485–95.

    PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Artemisinin Research Center, China Academy of Chinese Medical Sciences for assistance with the generation of confocal microscopy data. The schematic diagram was drawn by Figdraw. This work was supported by National Natural Science Foundation of China (82404657, U24A20798); the Natural Science Foundation of Shenzhen for Basic Research (JCYJ20240813104209012) and Guangdong Provincial Enterprise Joint Fund for Basic and Applied Research Foundation (2024A1515220163); Guangdong Basic and Applied Basic Research Foundation (2023A1515110710); the National Key Research and Development Program of China (2020YFA0908000, 2022YFC2303603); the Scientific and Technological Innovation Project of China Academy of Chinese Medical Sciences (CI2023D003, CI2023E005TS05, CI2023E005TS09); the Shenzhen Medical Research Funds (B2302051); the Establishment of Sino-Austria “Belt and Road” Joint Laboratory on Traditional Chinese Medicine for Severe Infectious Diseases and Joint Research (2020YFE0205100); the CACMS Innovation Fund (CI2023E002, CI2021A05101, ZG2024001-05); the Fundamental Research Funds for the Central Public Welfare Research Institutes (ZZ13-ZD-07, ZZ14-YQ-050, ZZ14-FL-010, ZZ14-ND-010, ZZ15-ND-10, ZZ16-ND-10-23, ZZ17-ND-10, ZZ18-ND-10).

Author information

Authors and Affiliations

Authors

Contributions

LH contributed to the conception. LH, QLS, CRF, HYL, and PLW designed the project. All authors contributed to data acquisition and analysis. QLS, CRF, HYL, PLW, WHK, and GJL conducted the research, data analysis, figure production, and contributed to draft the manuscript. SJQ performed bioinformatics analysis of the clinical data. PC, XW, RXC, RL, and JZZ performed the mice experiment and LC-MS/MS assay. LH and QLS wrote the original draft of the paper. LH, JGW, PS, and YY provided funding support. All authors reviewed and edited the paper.

Corresponding authors

Correspondence to Ping Song, Yuan Yuan, Ji-gang Wang or Ling Huang.

Ethics declarations

Competing interests

The authors declare no competing interests.

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

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shi, Ql., Feng, Cr., Li, Hy. et al. Celastrol inhibits the DPYSL2-JAK/STAT pathway by targeting mito-IDHs mediated mitochondrial metabolism to exhaust breast cancer. Acta Pharmacol Sin 46, 2765–2778 (2025). https://doi.org/10.1038/s41401-025-01548-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41401-025-01548-0

Keywords

Search

Quick links