| Peer-Reviewed

Research Progress on the Mechanism and Treatment of Ferroptosis in Brain Glioma

Received: 10 April 2023    Accepted: 2 May 2023    Published: 10 May 2023
Views:       Downloads:
Abstract

Ferroptosis is a new type of programmed cell death discovered in recent years. It is a regulatory cell death induced by iron dependent lipid peroxide injury. Ferroptosis plays a critical role in the development of glioma, affecting tumor proliferation, angiogenesis, tumor cell necrosis, and the formation of an immune-resistant tumor microenvironment. Glioma is the most common intracranial malignant tumor, with the characteristics of high incidence rate, high recurrence rate, high mortality rate and low cure rate. At present, the main standard treatment plan is tumor surgical resection, synchronous radiotherapy and chemotherapy, etc. Because of the extremely low 5-year survival rate and high recurrence rate of glioma patients, new effective treatment strategies are expected. With the extensive study of regulatory cell death in malignant tumors, there is increasing evidence that iron death is closely related to the development and outcome of glioma. Inducing iron death becomes an attractive strategy for glioma treatment. In this paper, we summarize the research on this aspect and summarize it in gelatin. The mechanism of action and therapeutic research value of tumor are expected to develop new therapeutic strategies and provide a certain theoretical basis for the in-depth research in this field.

Published in Cancer Research Journal (Volume 11, Issue 2)
DOI 10.11648/j.crj.20231102.12
Page(s) 44-48
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Ferroptosis, Glioma, Therapeutic Target, Molecular Mechanism

References
[1] Fang Y, Zhang Z. Arsenic trioxide as a novel anti-glioma drug: a review. Cell Mol Biol Lett. 2020 Sep 24; 25: 44.
[2] Ostrom QT, Gittleman H, Truitt G, et al. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011-2015. Neuro Oncol. 2018 Oct 1; 20 (suppl_4): iv1-iv86.
[3] Xu S, Tang L, Li X, et al. Immunotherapy for glioma: Current management and future application. Cancer Lett. 2020 Apr 28; 476: 1-12.
[4] Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016 Jun; 131 (6): 803-20.
[5] Smoll NR, Schaller K, Gautschi OP. Long-term survival of patients with glioblastoma multiforme (GBM). J Clin Neurosci. 2013 May; 20 (5): 670-5.
[6] Jin L, Guo S, Zhang X, et al. Optimal treatment strategy for adult patients with newly diagnosed glioblastoma: a systematic review and network meta-analysis. Neurosurg Rev. 2021 Aug; 44 (4): 1943-1955.
[7] Milano MT, Johnson MD, Sul J, et al. Primary spinal cord glioma: a Surveillance, Epidemiology, and End Results database study. J Neurooncol. 2010 May; 98 (1): 83-92.
[8] Miller JJ, Shih HA, Andronesi OC, et al. Isocitrate dehydrogenase-mutant glioma: Evolving clinical and therapeutic implications. Cancer. 2017 Dec 1; 123 (23): 4535-4546.
[9] Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012 May 25; 149 (5): 1060-72.
[10] Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018 Mar; 25 (3): 486-541.
[11] Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life Sci. 2016 Jun; 73 (11-12): 2195-209.
[12] Sun Y, Chen P, Zhai B, et al. The emerging role of ferroptosis in inflammation. Biomed Pharmacother. 2020 Jul; 127: 110108.
[13] Liu MR, Zhu WT, Pei DS. System xc (-): a key regulatory target of ferroptosis in cancer. Invest New Drugs (2021) 39 (4): 1123–31.
[14] Ingold I, Berndt C, Schmitt S, et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell. 2018 Jan 25; 172 (3): 409-422.e21.
[15] Gao M, Monian P, Quadri N, et al. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol Cell. 2015 Jul 16; 59 (2): 298-308.
[16] Muhoberac BB, Vidal R. Iron, Ferritin, Hereditary Ferritinopathy, and Neurodegeneration. Front Neurosci. 2019 Dec 11; 13: 1195.
[17] Du J, Wang T, Li Y, et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic Biol Med. 2019 Feb 1; 131: 356-369.
[18] Cheng J, Fan YQ, Liu BH, et al. Acsl4 suppresses glioma cells proliferation via activating ferroptosis. Oncol Rep (2020) 43 (1): 147–58.
[19] Lei P, Bai T, Sun Y. Mechanisms of Ferroptosis and Relations With Regulated Cell Death: A Review. Front Physiol. 2019 Feb 26; 10: 139.
[20] Gaschler MM, Stockwell BR. Lipid peroxidation in cell death. Biochem Biophys Res Commun. 2017 Jan 15; 482 (3): 419-425.
[21] Boonnoy P, Karttunen M, Wong-Ekkabut J. Alpha-tocopherol inhibits pore formation in oxidized bilayers. Phys Chem Chem Phys. 2017 Feb 22; 19 (8): 5699-5704.
[22] Hou W, Xie Y, Song X, et al. Autophagy Promotes Ferroptosis by Degradation Ferritin Autophagy (2016) 12 (8): 1425–8.
[23] Zhang L, Wang X, Cueto R, et al. Biochemical basis and metabolic interplay of redox regulation. Redox Biol. 2019 Sep; 26: 101284.
[24] Wenzel SE, Tyurina YY, Zhao J, et al. PEBP1 Wardens Ferroptosis by Enabling Lipoxygenase Generation of Lipid Death Signals. Cell. 2017 Oct 19; 171 (3): 628-641.e26.
[25] Feng H, Stockwell BR. Unsolved mysteries: How does lipid peroxidation cause ferroptosis? PLoS Biol. 2018 May 24; 16 (5): e2006203.
[26] Gout PW, Buckley AR, Simms CR, et al. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the x (c)- cystine transporter: a new action for an old drug. Leukemia. 2001 Oct; 15 (10): 1633-40.
[27] KOPPENOL WH, BOUNDS PL, DANG CV. Otto Warburg's contributions to current concepts of cancer metabolism [J]. Nat Rev Cancer, 2011, 11 (5): 325-337.
[28] Proneth B, Conrad M. Ferroptosis and necroinflammation, a yet poorly explored link. Cell Death Differ. 2019 Jan; 26 (1): 14-24.
[29] Tang D, Kepp O, Kroemer G. Ferroptosis becomes immunogenic: implications for anticancer treatments. Oncoimmunology. 2020 Dec 29; 10 (1): 1862949.
[30] Koike N, Kota R, Naito Y, et al. 2-Nitroimidazoles induce mitochondrial stress and ferroptosis in glioma stem cells residing in a hypoxic niche. Commun Biol. 2020 Aug 17; 3 (1): 450.
[31] Sehm T, Rauh M, Wiendieck K, et al. Temozolomide toxicity operates in a xCT/SLC7a11 dependent manner and is fostered by ferroptosis. Oncotarget. 2016 Nov 15; 7 (46): 74630-74647.
[32] Chen TC, Chuang JY, Ko CY, et al. AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide-resistant glioblastoma through disrupting GPX4-mediated redox homeostasis. Redox Biol (2020) 30: 101413.
[33] Chen LY, Li XX, Liu LB, et al. Erastin sensitizes glioblastoma cells to temozolomide by restraining xCT and cystathionine-gamma-lyase function. Oncol Rep (2015) 33 (3): 1465–74.
[34] Ye LF, Chaudhary KR, Zandkarimi F, et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem Biol (2020) 15 (2): 469–84.
[35] Zhou BR, Liu J, Kang R, et al. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol (2020) 66: 89–100.
[36] Chen D, Fan Z, Rauh M, et al. ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene (2017) 36 (40): 5593–608.
[37] Shen LS, Zhou YX, He HF, et al. Crosstalk between macrophages, T cells, and iron metabolism in tumor microenvironment. Oxid Med Cell Longevity (2021) 2021: 8865791.
[38] Yee PP, Wei Y, Kim SY, et al. Neutrophil-induced ferroptosis promotes tumor necrosis in glioblastoma progression. Nat Commun. 2020 Oct 27; 11 (1): 5424.
[39] Hu ZF, Mi YJ, Qian HM, et al. A potential mechanism of temozolomide resistance in glioma-ferroptosis. Front Oncol (2020) 10.
[40] Buccarelli M, Marconi M, Pacioni S, et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis (2018) 9 (8): 841.
[41] Yi RX, Wang HD, Deng CL, et al. Dihydroartemisinin initiates ferroptosis in glioblastoma through GPX4 inhibition. Biosci Rep (2020) 40: 841.
[42] Chen Y, Li N, Wang HJ, et al. Amentoflavone suppresses cell proliferation and induces cell death through triggering autophagy-dependent ferroptosis in human glioma. Life Sci (2020) 247: 299–312.
[43] Lu S, Wang XZ, He C, et al. ATF3 contributes to brucine-triggered glioma cell ferroptosis via promotion of hydrogen peroxide and iron. Acta Pharmacol Sin (2021) 42 (10): 1690–702.
[44] Gao X, Guo N, Xu H, et al. Ibuprofen induces ferroptosis of glioblastoma cells via downregulation of nuclear factor erythroid 2-related factor 2 signaling pathway. Anticancer Drugs (2020) 31 (1): 27–34.
[45] Jing Z, Li M, Wang H, et al. Gallic Acid-gold nanoparticles enhance radiation-induced cell death of human glioma U251 cells. IUBMB Life (2021) 73 (2): 398–407.
[46] Zhang Y, Xi K, Fu X, et al. Versatile metal-phenolic network nanoparticles for multitargeted combination therapy and magnetic resonance tracing in glioblastoma. Biomaterials (2021) 278: 121163.
[47] Xie B, Guo Y. Molecular mechanism of cell ferroptosis and research progress in regulation of ferroptosis by noncoding RNAs in tumor cells. Cell Death Discov. 2021 May 12; 7 (1): 101.
[48] Zhang X, Wang L, Li H, et al. Crosstalk between noncoding RNAs and ferroptosis: new dawn for overcoming cancer progression. Cell Death Dis. 2020 Jul 24; 11 (7): 580.
[49] Song Z, Jia G, Ma P, et al. Exosomal miR-4443 promotes cisplatin resistance in non-small cell lung carcinoma by regulating FSP1 m6A modification-mediated ferroptosis. Life Sci. 2021 Jul 1; 276: 119399.
[50] Bai T, Liang R, Zhu R, et al. MicroRNA-214-3p enhances erastin-induced ferroptosis by targeting ATF4 in hepatoma cells. J Cell Physiol. 2020 Jul; 235 (7-8): 5637-5648.
[51] Gomaa A, Peng D, Chen Z, et al. Epigenetic regulation of AURKA by miR-4715-3p in upper gastrointestinal cancers. Sci Rep. 2019 Nov 18; 9 (1): 16970.
Cite This Article
  • APA Style

    Xiaohui Han, Shihua Liu, Sen Yang, Aixia Sui. (2023). Research Progress on the Mechanism and Treatment of Ferroptosis in Brain Glioma. Cancer Research Journal, 11(2), 44-48. https://doi.org/10.11648/j.crj.20231102.12

    Copy | Download

    ACS Style

    Xiaohui Han; Shihua Liu; Sen Yang; Aixia Sui. Research Progress on the Mechanism and Treatment of Ferroptosis in Brain Glioma. Cancer Res. J. 2023, 11(2), 44-48. doi: 10.11648/j.crj.20231102.12

    Copy | Download

    AMA Style

    Xiaohui Han, Shihua Liu, Sen Yang, Aixia Sui. Research Progress on the Mechanism and Treatment of Ferroptosis in Brain Glioma. Cancer Res J. 2023;11(2):44-48. doi: 10.11648/j.crj.20231102.12

    Copy | Download

  • @article{10.11648/j.crj.20231102.12,
      author = {Xiaohui Han and Shihua Liu and Sen Yang and Aixia Sui},
      title = {Research Progress on the Mechanism and Treatment of Ferroptosis in Brain Glioma},
      journal = {Cancer Research Journal},
      volume = {11},
      number = {2},
      pages = {44-48},
      doi = {10.11648/j.crj.20231102.12},
      url = {https://doi.org/10.11648/j.crj.20231102.12},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.crj.20231102.12},
      abstract = {Ferroptosis is a new type of programmed cell death discovered in recent years. It is a regulatory cell death induced by iron dependent lipid peroxide injury. Ferroptosis plays a critical role in the development of glioma, affecting tumor proliferation, angiogenesis, tumor cell necrosis, and the formation of an immune-resistant tumor microenvironment. Glioma is the most common intracranial malignant tumor, with the characteristics of high incidence rate, high recurrence rate, high mortality rate and low cure rate. At present, the main standard treatment plan is tumor surgical resection, synchronous radiotherapy and chemotherapy, etc. Because of the extremely low 5-year survival rate and high recurrence rate of glioma patients, new effective treatment strategies are expected. With the extensive study of regulatory cell death in malignant tumors, there is increasing evidence that iron death is closely related to the development and outcome of glioma. Inducing iron death becomes an attractive strategy for glioma treatment. In this paper, we summarize the research on this aspect and summarize it in gelatin. The mechanism of action and therapeutic research value of tumor are expected to develop new therapeutic strategies and provide a certain theoretical basis for the in-depth research in this field.},
     year = {2023}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Research Progress on the Mechanism and Treatment of Ferroptosis in Brain Glioma
    AU  - Xiaohui Han
    AU  - Shihua Liu
    AU  - Sen Yang
    AU  - Aixia Sui
    Y1  - 2023/05/10
    PY  - 2023
    N1  - https://doi.org/10.11648/j.crj.20231102.12
    DO  - 10.11648/j.crj.20231102.12
    T2  - Cancer Research Journal
    JF  - Cancer Research Journal
    JO  - Cancer Research Journal
    SP  - 44
    EP  - 48
    PB  - Science Publishing Group
    SN  - 2330-8214
    UR  - https://doi.org/10.11648/j.crj.20231102.12
    AB  - Ferroptosis is a new type of programmed cell death discovered in recent years. It is a regulatory cell death induced by iron dependent lipid peroxide injury. Ferroptosis plays a critical role in the development of glioma, affecting tumor proliferation, angiogenesis, tumor cell necrosis, and the formation of an immune-resistant tumor microenvironment. Glioma is the most common intracranial malignant tumor, with the characteristics of high incidence rate, high recurrence rate, high mortality rate and low cure rate. At present, the main standard treatment plan is tumor surgical resection, synchronous radiotherapy and chemotherapy, etc. Because of the extremely low 5-year survival rate and high recurrence rate of glioma patients, new effective treatment strategies are expected. With the extensive study of regulatory cell death in malignant tumors, there is increasing evidence that iron death is closely related to the development and outcome of glioma. Inducing iron death becomes an attractive strategy for glioma treatment. In this paper, we summarize the research on this aspect and summarize it in gelatin. The mechanism of action and therapeutic research value of tumor are expected to develop new therapeutic strategies and provide a certain theoretical basis for the in-depth research in this field.
    VL  - 11
    IS  - 2
    ER  - 

    Copy | Download

Author Information
  • Department of Oncology, Hebei General Hospital, Shijiazhuang, China

  • Department of Oncology, Hebei General Hospital, Shijiazhuang, China

  • Department of Oncology, Hebei General Hospital, Shijiazhuang, China

  • Department of Oncology, Hebei General Hospital, Shijiazhuang, China

  • Sections