Ferroptosis in the pathogenesis of brain tumors

Cover Page

Cite item

Full Text

Abstract

The field of research on ferroptosis has seen an explosive growth in the past few years since the term was coined in 2012. This review highlights the current state of knowledge on the developmental mechanism of this unique mode of cell death, induced by iron-dependent phospholipid peroxidation, which is regulated by a variety of cellular metabolic events, including redox homeostasis. The xCT system, an amino acid antiporter that supports the synthesis of glutathione (GSH) and oxidation protection, is among these factors. The risk of iron accumulation in neurons, astrocytes, oligodendrocytes, microglia, and Schwann cells and the development of oxidative stress are discussed. Ferroptosis triggers a cascade of events including activation of inflammation, oxidation of neurotransmitters, impaired neuronal communication, myelin sheath degeneration, astrocyte dysregulation, dementia, and cell death. On the other hand, the exceptional vulnerability of cancer cells originating from the nervous tissue to ferroptosis is estimated. The evidence is given for the initiation of ferroptosis in tumor cells as a factor inhibiting the growth and promoting the death of these cells. Particular attention is paid to the pharmacological modulation of ferroptosis through its induction and inhibition for the treatment of drug-resistant cancers. The choice of targets for the induction of ferroptosis in cancer cells is discussed. Glutathione peroxidase 4 and xCT amino acid antiporter are recognized as the most preferred targets and the antitumor potential of their inhibition and side effects are evaluated.

About the authors

Alexandr A. Nikolaev

Astrakhan State Medical University

Author for correspondence.
Email: chimnik@mail.ru
ORCID iD: 0000-0001-6607-430X
SPIN-code: 8417-3876

MD, PhD, Professor

Russian Federation, 121, Bakinskaya street, Astrakhan, 414000

Vladimir V. Belopasov

Astrakhan State Medical University

Email: belopasov@yandex.ru
ORCID iD: 0000-0003-0458-0703
SPIN-code: 6098-1321

MD, PhD, Professor

Russian Federation, 121, Bakinskaya street, Astrakhan, 414000

References

  1. Louis DN, Perry A, Wesseling P, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021;23(8):1231–1251. doi: 10.1093/neuonc/noab106
  2. Wirsching HG, Galanis E, Weller M. Glioblastoma. Handb Clin Neurol. 2016;134:381–397. doi: 10.1016/B978-0-12-802997-8.00023-2
  3. Nakagawara A, Li Y, Izumi H, et al. Neuroblastoma. Jpn J Clin Oncol. 2018;48(3):214–241. doi: 10.1093/jjco/hyx176
  4. Klekner L, Szivos L, Virga J, et al. Significance of liquid biopsyin glioblastoma: A review. J Biotechnol. 2019;298:82–87. doi: 10.1016/j.jbiotec.2019.04.011
  5. Sathornsumetee S, Rich JN. New approaches to primary brain tumor treatment. Anticancer Drugs. 2006;17(9):1003–1016. doi: 10.1097/01.cad.0000231473.00030.1f
  6. Qiu Y. The relation between necessary trace element iron and various diseases. Biol Trace Elem Res. 1999;4:19–22. doi: 10.16755/j.cnki.issn.1006-446x.1997.10.006
  7. Agrawal KN. Iron Brain. J Univers Coll Med Sci. 2013;1:1–6. doi: 10.3126/jucms.v1i1.8425
  8. Mccann S, Amado M, Moore SE. The role of iron in brain development: A systematic review. Nutrients. 2020;12(7): 2001–2023. doi: 10.3390/nu12072001
  9. Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology, and role in disease. Nat Rev Mol Cell Biol. 2021; 22(4):266–282. doi: 10.1038/s41580-020-00324-8
  10. Rouault TA. The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nat Chem Biol. 2006;2(8): 406–414. doi: 10.1038/nchembio807
  11. Montalbetti N, Simonin A, Kovacs G, Hediger MA. Mammalian iron transporters: Families SLC11 and SLC40. Mol Aspects Med. 2013;34(2-3):270–287. doi: 10.1016/j.mam.2013.01.002
  12. DeGregorio-Rocasolano N, Martí-Sistac O, Gasull T. Deciphering the iron side of stroke: neurodegeneration at the crossroads between iron dyshomeostasis, excitotoxicity, and ferroptosis. Front Neurosci. 2019;13(1):85–96. doi: 10.3389/fnins.2019.00085
  13. Reichert CO, de Freitas FA, Sampaio-Silva J, et al. Ferroptosis mechanisms involvel in neurodegenerative diseases. Int J Mol Sci. 2020;21(22):8765–8783. doi: 10.3390/ijms21228765
  14. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–1072. doi: 10.1016/j.cell.2012.03.042
  15. Bayır H, Anthonymuthu TS, Tyurina YY, et al. Achieving life through death: redox biology of lipid peroxidation in ferroptosis. Cell Chem. Biol. 2020;27(4):387–408. doi: 10.1016/j.chembiol.2020.03.014
  16. Fricker M, Tolkovsky AM, Borutaite V, Coleman M. Neuronal cell death. Physiol Rev. 2018;98(2):813–880. doi: 10.1152/physrev.00011.2017
  17. Sun Y, Chen P, Zhai B, et al. The emerging role of ferroptosis in inflammation. Biomed Pharmacother. 2020;127:110108. doi: 10.1016/j.biopha.2020.110108
  18. Galaris D, Barbouti A, Pantopoulos K. Iron homeostasis and oxidative stress: an intimate relationship. Biochim Biophys Acta Mol Cell Res. 2019;1866(12):118535–118548 doi: 10.1016/j.bbamcr.2019.118535
  19. Mao H, Zhao Y, Li H, Lei L. Ferroptosis as an emerging target in inflammatory diseases. Prog Biophys Mol Biol. 2020;155:20–28. doi: 10.1016/j.pbiomolbio.2020.04.001
  20. Lachaier E, Louandre C, Godin C, et al. Sorafenib induces ferroptosis in human cancer cell lines originating from different solid tumors. Anticancer Res. 2014;34(11):6417–6422.
  21. Kabiraj P, Valenzuela CA, Marin JE, et al. The neuroprotective role of ferrostatin-1 under rotenoneinduced oxidative stress in dopaminergic neuroblastoma cells. Protein J. 2015;34(5): 349–358. doi: 10.1007/s10930-015-9629-7
  22. Wang YQ, Chang SY, Wu Q, et al. The protective role of mitochondrial ferritin on erastin-induced ferroptosis. Front Aging Neurosci. 2016;8:308. doi: 10.3389/fnagi.2016.00308
  23. Ivanov SD, Semenov AL, Mikhelson VM, et al. Effects of iron ion additional introduction in radiation therapy of tumor-bearing animals. Radiats Biol Radioecol. 2013;53(3):296–303 doi: 10.7868/s0869803113030065
  24. Ivanov SD, Semenov AL, Kovanko EG, Yamshanov VA. Effects of iron ions and iron chelation on the efficiency of experimental radiotherapy of animals with gliomas. Bull Exp Biol Med. 2015;158(6):800–803. doi: 10.1007/s10517-015-2865-1
  25. Chen D, Rauh M, Buchfelder M, Savaskan N. The oxido-metabolic driver ATF4 enhances temozolamide chemo-resistance in human gliomas. Oncotarget. 2017;8(31):51164–51176. doi: 10.18632/oncotarget.17737
  26. Sehm T, Rauh M, Wiendieck K, et al. Temozolomide toxicity operates in a xCT/SLC7a11 dependent manner and is fostered by ferroptosis. Oncotarget. 2017;7(46):74630–74647. doi: 10.18632/oncotarget.11858
  27. Chung WJ, Sontheimer H. Sulfasalazine inhibits the growth of primary brain tumorsindependent of nuclear factor-kappa B. J Neurochem. 2009;110(1):182–193. doi: 10.1111/j.1471-4159.2009.06129.x
  28. Sehm T, Fan Z, Ghoochani A, et al. Sulfasalazine impacts on ferroptotic cell deathand alleviates the tumor microenvironment and glioma-induced brain edema. Oncotarget. 2016;7(24): 36021–36033. doi: 10.18632/oncotarget.8651
  29. Hare D, Ayton S, Bush A, Lei P. A delicate balance: Iron metabolism and diseases of the brain. Front Aging Neurosci. 2013;5:34. doi: 10.3389/fnagi.2013.00034
  30. Dias V, Junn E, Mouradian MM. The role of oxidative stress in parkinsons disease. J Parkinson Dis. 2013;3(4):461–491. doi: 10.3233/JPD-130230
  31. Fan Z, Wirth AK, Chen D, et al. Nrf2-Keap1pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 2017; 6(8):371–385. doi: 10.1038/oncsis.2017.65
  32. Ye P, Mimura J, Okada T, et al. Nrf2- and ATF4-dependent upregulation of xCT modulates the sensitivity of T24 bladder carcinoma cells to proteasome inhibition. Mol Cell Biol. 2014;34(18):3421–3434. doi: 10.1128/MCB.00221-14
  33. Habib E, Linher-Melville K, Lin HX, Singh G. Expression of xCT and activity of systemxc(-) are regulated by NRF2 in human breast cancer cells in response to oxidative stress. Redox Biol. 2015;5:33–42. doi: 10.1016/j.redox.2015.03.003
  34. Hassannia B, Wiernicki B, Ingold I, et al. Nano-targeted induction of dual ferroptotic mechanismseradicates high-risk neuroblastoma. J Clin Invest. 2018;128(8):3341–3355. doi: 10.1172/JCI99032
  35. Abdalkader M, Lampinen R, Kanninen KM, Malm TM. Targeting Nrf2 to suppress ferroptosis and mitochondrial dysfunction in neurodegeneration. Front Neurosci. 2018;12:466–481. doi: 10.3389/fnins.2018.00466
  36. Hassannia B, Vandenabeele P, Vanden Berghe T. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35(6):830–849. doi: 10.1016/j.ccell.2019.04.002
  37. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1-2): 317–331. doi: 10.1016/j.cell.2013.12.010
  38. Chen Y, Liu Y T, et al. Quantitative profiling of protein carbonylations in ferroptosis by an aniline-derived probe. J Am Chem Soc. 2018;140(13):4712–4720. doi: 10.1021/jacs.8b01462
  39. Eaton JK, Ruberto RA, Kramm A, et al. Diacylfuroxans are masked nitrile oxides that inhibit GPX4 covalently. J Am Chem Soc. 2019;141(51):20407–20415. doi: 10.1021/jacs.9b10769
  40. Eaton JK, LR, Ruberto RA, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506. doi: 10.1038/s41589-020-0501-5
  41. Conrad M, Pratt DA. The chemical basis of ferroptosis. Nat Chem Biol. 2019;15(12):1137–1147. doi: 10.1038/s41589-019-0408-1
  42. Weiland A, Wang Y, Wu W, et al. J Mol Neurobiol. 2019;56(7): 4880–4893. doi: 10.1007/s12035-018-1403-3
  43. Sato H AM, Kimata M, et al. Redox imbalance in cystine/glutamate transporter-deficient mice. J Biol Chem. 2005;280(45): 37423–37429. doi: 10.1074/jbc.M506439200
  44. Robert MS, Buckingham S, Campbell S, et al. SLC7A11 expression is associated with seizures and predicts poor survival in patients with malignant glioma. Sci Transl Med. 2015;7(289):289ra286. doi: 10.1126/scitranslmed.aaa8103
  45. Zhang Y, Tan HJ, Daniels J, et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem Biol. 2019;26(5):623–633e629. doi: 10.1016/j.chembiol.2019.01.008
  46. Badgley MA, Kremer DM, Maurer HC, et al. Cysteine. Science. 2020;368(6486):85–89. doi: 10.1126/science.aaw 9872
  47. Sato M, Onuma K, Mio D, et al. Loss of the cystine/glutamate antiporter in melanoma abrogates tumor metastasis and markedly increases survival rates of mice. Int J Cancer. 2020; 147(11):3224–3235. doi: 10.1002/ijc.33262
  48. Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693–698. doi: 10.1038/s41586-019-1707-0
  49. Sano H, Futamura M, Gaowa S, et al. p53/Mieap-regulated mitochondrial quality control plays an important role as a tumor suppressor in gastric and esophageal cancers. Biochem Biophys Res Commun. 2020;529(3):582–589. doi: 10.1016/j.bbrc.2020.05.168
  50. Liu Y, Gu W. p53 in ferroptosis regulation: The new weapon for the old guardian. Cell Death Differ. 2022;29(5):895–910. doi: 10.1038/s41418-022-00943-y
  51. Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693–698. doi: 10.1038/s41586-019-1707-0 2019
  52. Yuan B, Zhao XD, Shen JD, et al. Activation of SIRT1 alleviates ferroptosis in the early brain injury after subarachnoid hemorrhage. Oxid Med Cell Longev. 2022;2022:9069825. doi: 10.1155/2022/9069825

Copyright (c) 2023 Eco-Vector

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies