Skip to main content

Advertisement

Log in

Disruption of mitochondrial oxidative phosphorylation by chidamide eradicates leukemic cells in AML

  • RESEARCH ARTICLE
  • Published:
Clinical and Translational Oncology Aims and scope Submit manuscript

Abstract

Purpose

Nowadays, the oxidative phosphorylation (OXPHOS) correlated with leukemogenesis and treatment response is extensive. Thus, exploration of novel approaches in disrupting OXPHOS in AML is urgently needed.

Materials and methods

Bioinformatical analysis of TCGA AML dataset was performed to identify the molecular signaling of OXPHOS. The OXPHOS level was measured through a Seahorse XFe96 cell metabolic analyzer. Flow cytometry was applied to measure mitochondrial status. Real-time qPCR and western blot were used to analyze the expression of mitochondrial or inflammatory factors. MLL-AF9-induced leukemic mice were conducted to measure the anti-leukemia effect of chidamide.

Results

Here, we reported that AML patients with high OXPHOS level were in a poor prognosis, which was associated with high expression of HDAC1/3 (TCGA). Inhibition of HDAC1/3 by chidamide inhibited cell proliferation and induced apoptotic cell death in AML cells. Intriguingly, chidamide could disrupt mitochondrial OXPHOS as assessed by inducing mitochondrial superoxide and reducing oxygen consumption rate, as well as decreasing mitochondrial ATP production. We also observed that chidamide augmented HK1 expression, while glycolysis inhibitor 2-DG could reduce the elevation of HK1 and improve the sensitivity of AML cells exposed to chidamide. Furthermore, HDAC3 was correlated with hyperinflammatory status, while chidamide could downregulate the inflammatory signaling in AML. Notably, chidamide eradicated leukemic cells in vivo and prolonged the survival time of MLL-AF9-induced AML mice.

Conclusion

Chidamide disrupted mitochondrial OXPHOS, promoted cell apoptosis and reduced inflammation in AML cells. These findings exhibited a novel mechanism that targeting OXPHOS would be a novel strategy for AML treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

Data availability

The data presented in this study are available on request from the corresponding author.

References

  1. de Beauchamp L, Himonas E, Helgason GV. Mitochondrial metabolism as a potential therapeutic target in myeloid leukaemia. Leukemia. 2022;36(1):1–12.

    Article  PubMed  Google Scholar 

  2. Amaya ML, Inguva A, Pei S, Jones C, Krug A, Ye H, et al. The STAT3-MYC axis promotes survival of leukemia stem cells by regulating SLC1A5 and oxidative phosphorylation. Blood. 2022;139(4):584–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lagadinou ED, Sach A, Callahan K, Rossi RM, Neering SJ, Minhajuddin M, et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell. 2013;12(3):329–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Pei S, Minhajuddin M, Adane B, Khan N, Stevens BM, Mack SC, et al. AMPK/FIS1-mediated mitophagy Is required for self-renewal of human AML stem cells. Cell Stem Cell. 2018;23(1):86-100.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Khan DH, Mullokandov M, Wu Y, Voisin V, Gronda M, Hurren R, et al. Mitochondrial carrier homolog 2 is necessary for AML survival. Blood. 2020;136(1):81–92.

    Article  PubMed  Google Scholar 

  6. Ma J, Liu B, Yu D, Zuo Y, Cai R, Yang J, et al. SIRT3 deacetylase activity confers chemoresistance in AML via regulation of mitochondrial oxidative phosphorylation. Br J Haematol. 2019;187(1):49–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, et al. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther. 2019;4:62.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Sun Y, Hong JH, Ning Z, Pan D, Fu X, Lu X, et al. Therapeutic potential of tucidinostat, a subtype-selective HDAC inhibitor, in cancer treatment. Front Pharmacol. 2022;13: 932914.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gui L, Cao J, Ji D, Zhang H, Fan Q, Zhu J, et al. Chidamide combined with cyclophosphamide, doxorubicin, vincristine and prednisone in previously untreated patients with peripheral T-cell lymphoma. Chin J Cancer Res. 2021;33(5):616–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhong M, Lin F, Jiang Y, Pan G, Tan J, Zhou H, et al. Therapeutic interaction of Apatinib and Chidamide in T-Cell acute lymphoblastic Leukemia through interference with mitochondria associated biogenesis and intrinsic apoptosis. J Pers Med. 2021;11(10):977.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Chen K, Yang Q, Zha J, Deng M, Zhou Y, Fu G, et al. Preclinical evaluation of a regimen combining chidamide and ABT-199 in acute myeloid leukemia. Cell Death Dis. 2020;11(9):778.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang H, Liu YC, Zhu CY, Yan F, Wang MZ, Chen XS, et al. Chidamide increases the sensitivity of refractory or relapsed acute myeloid leukemia cells to anthracyclines via regulation of the HDAC3 -AKT-P21-CDK2 signaling pathway. J Exp Clin Cancer Res. 2020;39(1):278.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Li G, Li D, Yuan F, Cheng C, Chen L, Wei X. Synergistic effect of chidamide and venetoclax on apoptosis in acute myeloid leukemia cells and its mechanism. Ann Transl Med. 2021;9(20):1575.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16(5):284–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18(1):220.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Mills EL, Kelly B, O’Neill LAJ. Mitochondria are the powerhouses of immunity. Nat Immunol. 2017;18(5):488–98.

    Article  CAS  PubMed  Google Scholar 

  17. Molina JR, Sun Y, Protopopova M, Gera S, Bandi M, Bristow C, et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat Med. 2018;24(7):1036–46.

    Article  CAS  PubMed  Google Scholar 

  18. Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer. 2006;6(1):38–51.

    Article  CAS  PubMed  Google Scholar 

  19. Bhaskara S, Knutson SK, Jiang G, Chandrasekharan MB, Wilson AJ, Zheng S, et al. Hdac3 is essential for the maintenance of chromatin structure and genome stability. Cancer Cell. 2010;18(5):436–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Long J, Fang WY, Chang L, Gao WH, Shen Y, Jia MY, et al. Targeting HDAC3, a new partner protein of AKT in the reversal of chemoresistance in acute myeloid leukemia via DNA damage response. Leukemia. 2017;31(12):2761–70.

    Article  CAS  PubMed  Google Scholar 

  21. Guo C, Li J, Steinauer N, Wong M, Wu B, Dickson A, et al. Histone deacetylase 3 preferentially binds and collaborates with the transcription factor RUNX1 to repress AML1-ETO-dependent transcription in t(8;21) AML. J Biol Chem. 2020;295(13):4212–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Beyer M, Romanski A, Mustafa AM, Pons M, Buchler I, Vogel A, et al. HDAC3 Activity is essential for human leukemic cell growth and the expression of beta-catenin, MYC, and WT1. Cancers (Basel). 2019;11(10):1436.

    Article  CAS  PubMed  Google Scholar 

  23. Chi Z, Chen S, Xu T, Zhen W, Yu W, Jiang D, et al. Histone deacetylase 3 couples mitochondria to drive IL-1beta-dependent inflammation by configuring fatty acid oxidation. Mol Cell. 2020;80(1):43-58.e7.

    Article  CAS  PubMed  Google Scholar 

  24. Weinberg SE, Chandel NS. Targeting mitochondria metabolism for cancer therapy. Nat Chem Biol. 2015;11(1):9–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sriskanthadevan S, Jeyaraju DV, Chung TE, Prabha S, Xu W, Skrtic M, et al. AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress. Blood. 2015;125(13):2120–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xiong Y, Guan KL. Mechanistic insights into the regulation of metabolic enzymes by acetylation. J Cell Biol. 2012;198(2):155–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang C, Cui G, Wang D, Wang M, Chen Q, Wang Y, et al. Crosstalk of oxidative phosphorylation-related subtypes, establishment of a prognostic signature and immune infiltration characteristics in colorectal adenocarcinoma. Cancers (Basel). 2022;14(18):4503.

    Article  CAS  PubMed  Google Scholar 

  28. Chen W, Yang Z, Chen Y. A novel oxidative phosphorylation-associated gene signature for prognosis prediction in patients with hepatocellular carcinoma. Dis Markers. 2022;2022:3594901.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Luna-Yolba R, Marmoiton J, Gigo V, Marechal X, Boet E, Sahal A, et al. Disrupting mitochondrial electron transfer chain complex i decreases immune checkpoints in murine and human acute myeloid leukemic cells. Cancers (Basel). 2021;13(14):3499.

    Article  CAS  PubMed  Google Scholar 

  30. Wang J, Fang Y, Ma S, Su N, Zhang Y, Huang H, et al. Comparison of chidamide-contained treatment modalities versus chemotherapy in the second-line treatment for relapsed or refractory peripheral T-cell lymphoma. Leuk Res. 2021;111: 106705.

    Article  CAS  PubMed  Google Scholar 

  31. Li Y, Wang Y, Zhou Y, Li J, Chen K, Zhang L, et al. Cooperative effect of chidamide and chemotherapeutic drugs induce apoptosis by DNA damage accumulation and repair defects in acute myeloid leukemia stem and progenitor cells. Clin Epigenetics. 2017;9:83.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Wang L, Luo J, Chen G, Fang M, Wei X, Li Y, et al. Chidamide, decitabine, cytarabine, aclarubicin, and granulocyte colony-stimulating factor (CDCAG) in patients with relapsed/refractory acute myeloid leukemia: a single-arm, phase 1/2 study. Clin Epigenetics. 2020;12(1):132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Dr. Shou-Jie Wang from Center for Precision Medicine, SYSU Platform of Metabolomics for the assistance with Seahorse XF Cell Mito Stress experiment.

Funding

This study was supported by Talent Training Project of The Third Affiliated Hospital of Sun Yat-sen University.

Author information

Authors and Affiliations

Authors

Contributions

JYW and ZJL: conceived and designed the experiments. JDW and JQX: performed the experiments. JDW analyzed the data and wrote the article. All authors revised the manuscript.

Corresponding authors

Correspondence to Zi-Jie Long or Jian-Yu Weng.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Informed consent

Not applicable.

Ethical approval and Research involving human participants and/or animals

The animal study was conducted with the approval of Institutional Animal Care and Use Committee.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

12094_2023_3079_MOESM1_ESM.tif

Supplementary file1 Effects of different metabolic inhibitors with chidamide. AML cells were treated chidamide (1 μM) or/and FCCP (1 µM/2 µM) (A, E), Oligomycin (1 µM/2 µM) (B, F), Rotenone (0.5 µM/1 µM) (C, G) or Metfoemin (5 mM/10 mM) (D, H) for 24 h and CCK-8 assay was applied to detect cell viability. Data were presented as mean ± SD, *p<0.05, **p<0.01, ***p<0.001 (TIF 9851 KB)

12094_2023_3079_MOESM2_ESM.tif

Supplementary file2 Relation between HDAC3 and immune cell infiltration is performed. (A) Immune cell enrichment was performed in HDAC3 high and low AML patients by ssGSEA. (B) Unsupervised clustering of immune checkpoint genes and infiltrating cells were analyzed in HDAC3 high and low groups. (C) Correlation between each infiltrating cell signature was assessed by spearman analysis. (D) Infiltrating cell infiltration abundance analysis in HDAC3 high and low groups was shown (TIF 32323 KB)

Supplementary file3 (DOCX 15 KB)

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

Wang, JD., Xu, JQ., Long, ZJ. et al. Disruption of mitochondrial oxidative phosphorylation by chidamide eradicates leukemic cells in AML. Clin Transl Oncol 25, 1805–1820 (2023). https://doi.org/10.1007/s12094-023-03079-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12094-023-03079-8

Keywords

Navigation