Ferroptosis in Cancer and Inflammatory Diseases: Mechanisms and Therapeutic Implications

Guangyi Shen , Jiachen Liu , Yinhuai Wang , Zebin Deng , Fei Deng

MedComm ›› 2025, Vol. 6 ›› Issue (9) : e70349

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (9) : e70349 DOI: 10.1002/mco2.70349
REVIEW

Ferroptosis in Cancer and Inflammatory Diseases: Mechanisms and Therapeutic Implications

Author information +
History +
PDF

Abstract

Ferroptosis, an iron-dependent cell death pathway driven by lipid peroxidation, has emerged as a critical pathophysiological mechanism linking cancer and inflammatory diseases. The seemingly distinct pathologies exhibit shared microenvironmental hallmarks—oxidative stress, immune dysregulation, and metabolic reprogramming—that converge on ferroptosis regulation. This review synthesizes how ferroptosis operates at the intersection of these diseases, acting as both a tumor-suppressive mechanism and a driver of inflammatory tissue damage. In cancer, ferroptosis eliminates therapy-resistant cells but paradoxically facilitates metastasis through lipid peroxidation byproducts that remodel the tumor microenvironment and suppress antitumor immunity. In chronic inflammatory diseases—from atherosclerosis to rheumatoid arthritis—ferroptosis amplifies neuroinflammatory cascades while simultaneously exposing vulnerabilities for therapeutic targeting. Central to this duality are shared regulatory nodes, including nuclear factor kappa B-driven inflammation, NOD-like receptor family pyrin domain-containing 3 inflammasome activation, and GPX4 dysfunction. Therapeutically, ferroptosis induction shows promise against therapy-resistant cancers but risks exacerbating inflammatory damage, underscoring the need for precision modulation. Emerging strategies—nanoparticle-based inducers, immunotherapy combinations, and biomarker-guided patient stratification—aim to balance prodeath efficacy against off-target toxicity. By dissecting the ferroptosis–inflammation–cancer axis, this review provides a unified framework for understanding disease pathogenesis and advancing therapies for conditions resistant to conventional treatments. Future research must prioritize spatial mapping of ferroptosis dynamics, mechanistic crosstalk with immune checkpoints, and combinatorial regimens that exploit ferroptosis vulnerabilities while mitigating its inflammatory consequences.

Keywords

ferroptosis / lipid peroxidation / cancer immunotherapy / inflammatory signaling / tumor microenvironment

Cite this article

Download citation ▾
Guangyi Shen, Jiachen Liu, Yinhuai Wang, Zebin Deng, Fei Deng. Ferroptosis in Cancer and Inflammatory Diseases: Mechanisms and Therapeutic Implications. MedComm, 2025, 6(9): e70349 DOI:10.1002/mco2.70349

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

X. Jin, J. Tang, X. Qiu, et al., “Ferroptosis: Emerging mechanisms, biological function, and therapeutic potential in cancer and inflammation,” Cell Death Discov 10 (2024): 45.

[2]

S. J. Dixon, K. M. Lemberg, M. R. Lamprecht, et al., “Ferroptosis: An iron-dependent form of nonapoptotic cell death,” Cell 149, no. 5 (2012): 1060-1072.

[3]

Q. Zhou, Y. Meng, D. Li, et al., “Ferroptosis in cancer: From molecular mechanisms to therapeutic strategies,” Signal Transduct Target Ther 9, no. 1 (2024): 55.

[4]

X. Chen, R. Kang, G. Kroemer, and D. Tang, “Ferroptosis in infection, inflammation, and immunity,” Journal of Experimental Medicine 218, no. 6 (2021): e20210518.

[5]

Q. Dang, Z. Sun, Y. Wang, L. Wang, Z. Liu, and X. Han, “Ferroptosis: A double-edged sword mediating immune tolerance of cancer,” Cell death & disease 13, no. 11 (2022): 925.

[6]

H. Zhao, C. Tang, M. Wang, H. Zhao, and Y. Zhu, “Ferroptosis as an emerging target in rheumatoid arthritis,” Frontiers in immunology 14 (2023): 1260839.

[7]

T. Zhao, Q. Yang, Y. Xi, et al., “Ferroptosis in Rheumatoid Arthritis: A Potential Therapeutic Strategy,” Frontiers in immunology 13 (2022): 779585.

[8]

X. Jiang, B. R. Stockwell, and M. Conrad, “Ferroptosis: Mechanisms, biology and role in disease,” Nature Reviews Molecular Cell Biology 22, no. 4 (2021): 266-282.

[9]

A. M. Pisoschi and A. Pop, “The role of antioxidants in the chemistry of oxidative stress: A review,” European Journal of Medicinal Chemistry 97 (2015): 55-74.

[10]

X. Chen, R. Kang, G. Kroemer, and D. Tang, “Broadening horizons: The role of ferroptosis in cancer,” Nature reviews Clinical oncology 18, no. 5 (2021): 280-296.

[11]

Y. Mou, J. Wang, J. Wu, et al., “Ferroptosis, a new form of cell death: Opportunities and challenges in cancer,” J Hematol OncolJ Hematol Oncol 12, no. 1 (2019): 34.

[12]

M. P. Horowitz and J. T. Greenamyre, “Mitochondrial Iron Metabolism and Its Role in Neurodegeneration,” J Alzheimers Dis JAD 20, no. Suppl 2 (2010): S551-S568.

[13]

A. Pefanis, F. L. Ierino, J. M. Murphy, and P. J. Cowan, “Regulated necrosis in kidney ischemia-reperfusion injury,” Kidney International 96, no. 2 (2019): 291-301.

[14]

A. M. Pisoschi, A. Pop, F. Iordache, L. Stanca, G. Predoi, and A. I. Serban, “Oxidative stress mitigation by antioxidants—An overview on their chemistry and influences on health status,” European Journal of Medicinal Chemistry 209 (2021): 112891.

[15]

B. R. Stockwell, J. P. F. Angeli, H. Bayir, et al., “Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease,” Cell 171, no. 2 (2017): 273-285.

[16]

W. Wang, M. Green, J. E. Choi, et al., “CD8+ T cells regulate tumour ferroptosis During cancer immunotherapy,” Nature 569, no. 7755 (2019): 270-274.

[17]

J. P. Friedmann Angeli, D. V. Krysko, and M. Conrad, “Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion,” Nature Reviews Cancer 19, no. 7 (2019): 405-414.

[18]

D. Liang, A. M. Minikes, and X. Jiang, “Ferroptosis at the intersection of lipid metabolism and cellular signaling,” Molecular Cell 82, no. 12 (2022): 2215-2227.

[19]

H. Eagle, “Nutrition needs of mammalian cells in tissue culture,” Science 122, no. 3168 (1955): 501-514.

[20]

H. Eagle, “The Specific Amino Acid Requirements of a Human Carcinoma Cell (Strain Hela) in Tissue Culture,” Journal of Experimental Medicine 102, no. 1 (1955): 37-48.

[21]

J. R. Mitchell, D. J. Jollow, W. Z. Potter, J. R. Gillette, and B. B. Brodie, “Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione,” Journal of Pharmacology and Experimental Therapeutics 187, no. 1 (1973): 211-217.

[22]

S. Bannai, H. Tsukeda, and H. Okumura, “Effect of antioxidants on cultured human diploid fibroblasts exposed to cystine-free medium,” Biochemical and Biophysical Research Communications 74, no. 4 (1977): 1582-1588.

[23]

M. Yonezawa, S. A. Back, X. Gan, P. A. Rosenberg, and J. J. Volpe, “Cystine deprivation induces oligodendroglial death: Rescue by free radical scavengers and by a diffusible glial factor,” Journal of Neurochemistry 67, no. 2 (1996): 566-573.

[24]

S. Tan, D. Schubert, and P. Maher, “Oxytosis: A novel form of programmed cell death,” Current Topics in Medicinal Chemistry 1, no. 6 (2001): 497-506.

[25]

S. Mercille and B. Massie, “Induction of apoptosis in nutrient-deprived cultures of hybridoma and myeloma cells,” Biotechnology and Bioengineering 44, no. 9 (1994): 1140-1154.

[26]

S. Christgen, R. E. Tweedell, and T. D. Kanneganti, “Programming inflammatory cell death for therapy,” Pharmacology & Therapeutics 232 (2022): 108010.

[27]

L. Galluzzi, I. Vitale, S. A. Aaronson, et al., “Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018,” Cell Death and Differentiation 25, no. 3 (2018): 486-541.

[28]

D. R. Green and B. Victor, “The pantheon of the fallen: Why are there so many forms of cell death?,” Trends in Cell Biology 22, no. 11 (2012): 555-556.

[29]

H. O. Fearnhead, P. Vandenabeele, and T. Vanden Berghe, “How do we fit ferroptosis in the family of regulated cell death?,” Cell Death and Differentiation 24, no. 12 (2017): 1991-1998.

[30]

S. J. Dixon, “Ferroptosis: Bug or feature?,” Immunological Reviews 277, no. 1 (2017): 150-157.

[31]

S. J. Dixon and J. A. Olzmann, “The cell biology of ferroptosis,” Nature Reviews Molecular Cell Biology 25, no. 6 (2024): 424-442.

[32]

D. Tang, X. Chen, R. Kang, and G. Kroemer, “Ferroptosis: Molecular mechanisms and health implications,” Cell Research 31, no. 2 (2021): 107-125.

[33]

N. KC and F. Ka, “Apoptosis, Pyroptosis, and Necroptosis-Oh My! The Many Ways a Cell Can Die,” Journal of Molecular Biology 434, no. 4 (2022): 167378.

[34]

M. S. D'Arcy, “Cell death: A review of the major forms of apoptosis, necrosis and autophagy,” Cell Biology International 43, no. 6 (2019): 582-592.

[35]

W. Gao, X. Wang, Y. Zhou, X. Wang, and Y. Yu, “Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy,” Signal Transduct Target Ther 7, no. 1 (2022): 196.

[36]

J. Yan, P. Wan, S. Choksi, and Z. G. Liu, “Necroptosis and tumor progression,” Trends in cancer 8, no. 1 (2022): 21-27.

[37]

D. Bertheloot, E. Latz, and B. S. Franklin, “Necroptosis, pyroptosis and apoptosis: An intricate game of cell death,” Cell Mol Immunol 18, no. 5 (2021): 1106-1121.

[38]

H. Han, G. Zhang, X. Zhang, and Q. Zhao, “Nrf2-mediated ferroptosis inhibition: A novel approach for managing inflammatory diseases,” Inflammopharmacology 32, no. 5 (2024): 2961-2986.

[39]

S. O. Vasudevan, B. Behl, and V. A. Rathinam, “Pyroptosis-induced inflammation and tissue damage,” Seminars in Immunology 69 (2023): 101781.

[40]

P. Yu, X. Zhang, N. Liu, L. Tang, C. Peng, and X. Chen, “Pyroptosis: Mechanisms and diseases,” Signal Transduct Target Ther 6, no. 1 (2021): 128.

[41]

S. Liu, S. Yao, H. Yang, S. Liu, and Y. Wang, “Autophagy: Regulator of cell death,” Cell death & disease 14, no. 10 (2023): 648.

[42]

J. Debnath, N. Gammoh, and K. M. Ryan, “Autophagy and autophagy-related pathways in cancer,” Nature Reviews Molecular Cell Biology 24, no. 8 (2023): 560-575.

[43]

N. Mizushima and M. Komatsu, “Autophagy: Renovation of cells and tissues,” Cell 147, no. 4 (2011): 728-741.

[44]

D. Glick, S. Barth, and K. F. Macleod, “Autophagy: Cellular and molecular mechanisms,” Journal of Pathology 221, no. 1 (2010): 3-12.

[45]

T. V. Berghe, A. Linkermann, S. Jouan-Lanhouet, H. Walczak, and P. Vandenabeele, “Regulated necrosis: The expanding network of non-apoptotic cell death pathways,” Nature Reviews Molecular Cell Biology 15, no. 2 (2014): 135-147.

[46]

H. Wang, C. Liu, Y. Zhao, and G. Gao, “Mitochondria regulation in ferroptosis,” European Journal of Cell Biology 99, no. 1 (2020): 151058.

[47]

J. P. F. Angeli, M. Schneider, B. Proneth, et al., “Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice,” Nature Cell Biology 16, no. 12 (2014): 1180-1191.

[48]

L. J. Su, J. H. Zhang, H. Gomez, et al., “Reactive Oxygen Species-Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis,” Oxid Med Cell Longev 2019 (2019): 5080843.

[49]

G. C. Forcina and S. J. Dixon, “GPX4 at the Crossroads of Lipid Homeostasis and Ferroptosis,” Proteomics 19, no. 18 (2019): e1800311.

[50]

M. Matsushita, S. Freigang, C. Schneider, M. Conrad, G. W. Bornkamm, and M. Kopf, “T cell lipid peroxidation induces ferroptosis and prevents immunity to infection,” Journal of Experimental Medicine 212, no. 4 (2015): 555-568.

[51]

E. J. Jang, D. H. Kim, B. Lee, et al., “Activation of proinflammatory signaling by 4-hydroxynonenal-Src adducts in aged kidneys,” Oncotarget 7, no. 32 (2016): 50864-50874.

[52]

R. Kang, R. Chen, Q. Zhang, et al., “HMGB1 in health and disease,” Molecular Aspects of Medicine 40 (2014): 1-116.

[53]

Q. Wen, J. Liu, R. Kang, B. Zhou, and D. Tang, “The release and activity of HMGB1 in ferroptosis,” Biochemical and Biophysical Research Communications 510, no. 2 (2019): 278-283.

[54]

R. A. DeBose-Boyd, “Significance and regulation of lipid metabolism,” Seminars in cell & developmental biology 81 (2018): 97.

[55]

W. S. Yang, K. J. Kim, M. M. Gaschler, M. Patel, M. S. Shchepinov, and B. R. Stockwell, “Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis,” PNAS 113, no. 34 (2016): E4966-E4975.

[56]

S. Doll, B. Proneth, Y. Y. Tyurina, et al., “Acsl4 Dictates Ferroptosis Sensitivity by Shaping Cellular Lipid Composition,” Nature Chemical Biology 13, no. 1 (2017): 91-98.

[57]

V. E. Kagan, G. Mao, F. Qu, et al., “Oxidized Arachidonic/Adrenic Phosphatidylethanolamines Navigate Cells to Ferroptosis,” Nature Chemical Biology 13, no. 1 (2017): 81-90.

[58]

S. E. Wenzel, Y. Y. Tyurina, J. Zhao, et al., “PEBP1 Wardens Ferroptosis by Enabling Lipoxygenase Generation of Lipid Death Signals,” Cell 171, no. 3 (2017): 628-641. e26.

[59]

M. Conrad and D. A. Pratt, “The chemical basis of ferroptosis,” Nature Chemical Biology 15, no. 12 (2019): 1137-1147.

[60]

L. Magtanong, P. J. Ko, M. To, et al., “Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State,” Cell Chem Biol 26, no. 3 (2019): 420-432. e9.

[61]

R. Shah, M. S. Shchepinov, and D. A. Pratt, “Resolving the Role of Lipoxygenases in the Initiation and Execution of Ferroptosis,” ACS Cent Sci 4, no. 3 (2018): 387-396.

[62]

S. Khanna, S. Roy, H. Ryu, et al., “Molecular basis of vitamin E action: Tocotrienol modulates 12-lipoxygenase, a key mediator of glutamate-induced neurodegeneration,” Journal of Biological Chemistry 278, no. 44 (2003): 43508-43515.

[63]

J. Liu, R. Kang, and D. Tang, “Signaling pathways and defense mechanisms of ferroptosis,” Febs Journal 289, no. 22 (2022): 7038-7050.

[64]

R. P. L. van Swelm, J. F. M. Wetzels, and D. W. Swinkels, “The multifaceted role of iron in renal health and disease,” Nature Reviews Nephrology 16, no. 2 (2020): 77-98.

[65]

M. Gao, P. Monian, N. Quadri, R. Ramasamy, and X. Jiang, “Glutaminolysis and Transferrin Regulate Ferroptosis,” Molecular Cell 59, no. 2 (2015): 298-308.

[66]

Z. Wang, Y. Ding, X. Wang, et al., “Pseudolaric acid B triggers ferroptosis in glioma cells via activation of Nox4 and inhibition of xCT,” Cancer Letters 428 (2018): 21-33.

[67]

W. Luo, J. Wang, W. Xu, et al., “LncRNA RP11-89 facilitates tumorigenesis and ferroptosis resistance Through PROM2-activated iron export by sponging miR-129-5p in bladder cancer,” Cell death & disease 12, no. 11 (2021): 1043.

[68]

D. L. Zhang, M. C. Ghosh, and T. A. Rouault, “The physiological functions of iron regulatory proteins in iron homeostasis—an update,” Frontiers in pharmacology 5 (2014): 124.

[69]

H. Kawabata, “Transferrin and transferrin receptors update,” Free Radic Biol Med 133 (2019): 46-54.

[70]

W. Hou, Y. Xie, X. Song, et al., “Autophagy promotes ferroptosis by degradation of ferritin,” Autophagy 12, no. 8 (2016): 1425-1428.

[71]

M. Gao, P. Monian, Q. Pan, W. Zhang, J. Xiang, and X. Jiang, “Ferroptosis is an autophagic cell death process,” Cell Research 26, no. 9 (2016): 1021-1032.

[72]

Y. Bai, L. Meng, L. Han, et al., “Lipid storage and lipophagy regulates ferroptosis,” Biochemical and Biophysical Research Communications 508, no. 4 (2019): 997-1003.

[73]

B. Zhou, J. Liu, R. Kang, D. J. Klionsky, G. Kroemer, and D. Tang, “Ferroptosis is a type of autophagy-dependent cell death,” Seminars in Cancer Biology 66 (2020): 89-100.

[74]

J. D. Mancias, X. Wang, S. P. Gygi, J. W. Harper, and A. C. Kimmelman, “Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy,” Nature 509, no. 7498 (2014): 105-109.

[75]

M. Gao, J. Yi, J. Zhu, et al., “Role of Mitochondria in Ferroptosis,” Molecular Cell 73, no. 2 (2019): 354-363. e3.

[76]

S. M. Parikh, Y. Yang, L. He, C. Tang, M. Zhan, and Z. Dong, “Mitochondrial Function and Disturbances in the Septic Kidney,” Seminars in Nephrology 35, no. 1 (2015): 108-119.

[77]

P. J. Urrutia, N. P. Mena, and M. T. Núñez, “The interplay Between iron accumulation, mitochondrial dysfunction, and inflammation During the execution step of neurodegenerative disorders,” Frontiers in pharmacology 5 (2014): 38.

[78]

A. Campanella, E. Rovelli, P. Santambrogio, A. Cozzi, F. Taroni, and S. Levi, “Mitochondrial ferritin limits oxidative damage regulating mitochondrial iron availability: Hypothesis for a protective role in Friedreich ataxia,” Human Molecular Genetics 18, no. 1 (2009): 1-11.

[79]

F. Missirlis, S. Holmberg, T. Georgieva, B. C. Dunkov, T. A. Rouault, and J. H. Law, “Characterization of mitochondrial ferritin in Drosophila,” PNAS 103, no. 15 (2006): 5893-5898.

[80]

G. Gao, N. Zhang, Y. Q. Wang, et al., “Mitochondrial Ferritin Protects Hydrogen Peroxide-Induced Neuronal Cell Damage,” Aging Dis 8, no. 4 (2017): 458-470.

[81]

A. A. Starkov, “The role of mitochondria in reactive oxygen species metabolism and signaling,” Ann N Y Acad Sci 1147 (2008): 37-52.

[82]

Y. Q. Wang, S. Y. Chang, Q. Wu, et al., “The Protective Role of Mitochondrial Ferritin on Erastin-Induced Ferroptosis,” Frontiers in aging neuroscience 8 (2016): 308.

[83]

S. Toyokuni, F. Ito, K. Yamashita, Y. Okazaki, and S. Akatsuka, “Iron and thiol redox signaling in cancer: An exquisite balance to escape ferroptosis,” Free Radic Biol Med 108 (2017): 610-626.

[84]

S. Agrawal, J. Fox, B. Thyagarajan, and J. H. Fox, “Brain mitochondrial iron accumulates in Huntington's disease, mediates mitochondrial dysfunction, and can be removed pharmacologically,” Free Radic Biol Med 120 (2018): 317-329.

[85]

O. Edenharter, J. Clement, S. Schneuwly, and J. A. Navarro, “Overexpression of Drosophila frataxin triggers cell death in an iron-dependent manner,” Journal of Neurogenetics 31, no. 4 (2017): 189-202.

[86]

K. Chen, G. Lin, N. A. Haelterman, et al., “Loss of Frataxin induces iron toxicity, sphingolipid synthesis, and Pdk1/Mef2 activation, leading to neurodegeneration,” Elife 5 (2016): e16043.

[87]

S. J. Oh, M. Ikeda, T. Ide, K. Y. Hur, and M. S. Lee, “Mitochondrial event as an ultimate step in ferroptosis,” Cell Death Discov 8 (2022): 414.

[88]

K. G. Lyamzaev, A. A. Panteleeva, R. A. Simonyan, A. V. Avetisyan, and B. V. Chernyak, “Mitochondrial Lipid Peroxidation Is Responsible for Ferroptosis,” Cells 12, no. 4 (2023): 611.

[89]

M. Colombini, “VDAC: The channel at the interface Between mitochondria and the cytosol,” Molecular and Cellular Biochemistry 256-257 (2004): 107-115. 1-2.

[90]

N. Yagoda, M. von Rechenberg, E. Zaganjor, et al., “RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels,” Nature 447, no. 7146 (2007): 864-868.

[91]

R. Guo, J. Duan, S. Pan, et al., “The Road From AKI to CKD: Molecular Mechanisms and Therapeutic Targets of Ferroptosis,” Cell death & disease 14, no. 7 (2023): 426.

[92]

Y. Chen, Y. Liu, T. Lan, et al., “Quantitative Profiling of Protein Carbonylations in Ferroptosis by an Aniline-Derived Probe,” Journal of the American Chemical Society 140, no. 13 (2018): 4712-4720.

[93]

Y. Yang, M. Luo, K. Zhang, et al., “Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma,” Nature Communications 11, no. 1 (2020): 433.

[94]

M. A. Badgley, D. M. Kremer, H. C. Maurer, et al., “Cysteine depletion induces pancreatic tumor ferroptosis in mice,” Science 368, no. 6486 (2020): 85-89.

[95]

X. Lang, M. D. Green, W. Wang, et al., “Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11,” Cancer discovery 9, no. 12 (2019): 1673-1685.

[96]

T. Himi, M. Ikeda, T. Yasuhara, and S. I. Murota, “Oxidative neuronal death caused by glutamate uptake inhibition in cultured hippocampal neurons,” Journal of Neuroscience Research 71, no. 5 (2003): 679-688.

[97]

L. Wang, Y. Liu, T. Du, et al., “ATF3 promotes erastin-induced ferroptosis by suppressing system Xc,” Cell Death and Differentiation 27, no. 2 (2020): 662-675.

[98]

Y. Xu, Y. Li, J. Li, and W. Chen, “Ethyl carbamate triggers ferroptosis in liver Through inhibiting GSH synthesis and suppressing Nrf2 activation,” Redox Biology 53 (2022): 102349.

[99]

J. Wang, Y. Liu, Y. Wang, and L. Sun, “The Cross-Link Between Ferroptosis and Kidney Diseases,” Oxid Med Cell Longev 2021 (2021): 6654887.

[100]

F. Ursini, M. Maiorino, M. Valente, L. Ferri, and C. Gregolin, “Purification From pig liver of a protein which protects liposomes and biomembranes From peroxidative degradation and exhibits glutathione peroxidase activity on phosphatidylcholine hydroperoxides,” Biochimica Et Biophysica Acta 710, no. 2 (1982): 197-211.

[101]

V. E. Kagan, G. Mao, F. Qu, et al., “Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis,” Nature Chemical Biology 13, no. 1 (2017): 81-90.

[102]

F. Ursini and M. Maiorino, “Lipid peroxidation and ferroptosis: The role of GSH and GPx4,” Free Radic Biol Med 152 (2020): 175-185.

[103]

K. Hosohata, T. Harnsirikarn, and S. Chokesuwattanaskul, “Ferroptosis: A Potential Therapeutic Target in Acute Kidney Injury,” International Journal of Molecular Sciences 23, no. 12 (2022): 6583.

[104]

T. M. Seibt, B. Proneth, and M. Conrad, “Role of GPX4 in ferroptosis and its pharmacological implication,” Free Radic Biol Med 133 (2019): 144-152.

[105]

C. Chen, D. Wang, Y. Yu, et al., “Legumain promotes tubular ferroptosis by facilitating chaperone-mediated autophagy of GPX4 in AKI,” Cell death & disease 12, no. 1 (2021): 65.

[106]

K. Wu, M. Yan, T. Liu, et al., “Creatine kinase B suppresses ferroptosis by phosphorylating GPX4 Through a moonlighting function,” Nature Cell Biology 25, no. 5 (2023): 714-725.

[107]

Q. Xue, D. Yan, X. Chen, et al., “Copper-dependent autophagic degradation of GPX4 drives ferroptosis,” Autophagy 19, no. 7 (2023): 1982-1996.

[108]

D. M. Cheff, C. Huang, K. C. Scholzen, et al., “The ferroptosis inducing compounds RSL3 and ML162 are not direct inhibitors of GPX4 but of TXNRD1,” Redox Biology 62 (2023): 102703.

[109]

P. Sabatier, C. M. Beusch, R. Gencheva, Q. Cheng, R. Zubarev, and E. S. J. Arnér, “Comprehensive chemical proteomics analyses reveal that the new TRi-1 and TRi-2 compounds are more specific thioredoxin reductase 1 inhibitors Than auranofin,” Redox Biology 48 (2021): 102184.

[110]

W. Li, L. Liang, S. Liu, H. Yi, and Y. Zhou, “FSP1: A key regulator of ferroptosis,” Trends in Molecular Medicine 29, no. 9 (2023): 753-764.

[111]

F. Zeng, X. Chen, and G. Deng, “The anti-ferroptotic role of FSP1: Current molecular mechanism and therapeutic approach,” Mol Biomed 3, no. 1 (2022): 37.

[112]

K. Bersuker, J. M. Hendricks, Z. Li, et al., “The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis,” Nature 575, no. 7784 (2019): 688-692.

[113]

K. Shimada, R. Skouta, A. Kaplan, et al., “Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis,” Nature Chemical Biology 12, no. 7 (2016): 497-503.

[114]

P. Mladěnka, K. Macáková, L. Kujovská Krčmová, et al., “Vitamin K—sources, physiological role, kinetics, deficiency, detection, therapeutic use, and toxicity,” Nutrition Reviews 80, no. 4 (2022): 677-698.

[115]

X. Lin, Q. Zhang, Q. Li, et al., “Upregulation of CoQ shifts ferroptosis dependence From GPX4 to FSP1 in acquired radioresistance,” Drug Resist Updat Rev Comment Antimicrob Anticancer Chemother 73 (2023): 101032.

[116]

Y. Liu, D. Cheng, Y. Wang, et al., “UHRF1-mediated ferroptosis promotes pulmonary fibrosis via epigenetic repression of GPX4 and FSP1 genes,” Cell death & disease 13, no. 12 (2022): 1070.

[117]

J. Zheng and M. Conrad, “The Metabolic Underpinnings of Ferroptosis,” Cell metabolism 32, no. 6 (2020): 920-937.

[118]

H. Yuan, X. Li, X. Zhang, R. Kang, and D. Tang, “Identification of ACSL4 as a biomarker and contributor of ferroptosis,” Biochemical and Biophysical Research Communications 478, no. 3 (2016): 1338-1343.

[119]

I. Ingold, C. Berndt, S. Schmitt, et al., “Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis,” Cell 172, no. 3 (2018): 409-422. e21.

[120]

H. L. Zhang, B. X. Hu, Z. L. Li, et al., “PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis,” Nature Cell Biology 24, no. 1 (2022): 88-98.

[121]

P. Liao, W. Wang, and W. Wang, “CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4,” Cancer Cell 40, no. 4 (2022): 365-378. e6.

[122]

Y. Yang, T. Zhu, X. Wang, et al., “ACSL3 and ACSL4, Distinct Roles in Ferroptosis and Cancers,” Cancers 14, no. 23 (2022): 5896.

[123]

M. Li, Z. Meng, S. Yu, et al., “Baicalein ameliorates cerebral ischemia-reperfusion injury by inhibiting ferroptosis via regulating GPX4/ACSL4/ACSL3 axis,” Chemico-Biological Interactions 366 (2022): 110137.

[124]

C. Louandre, I. Marcq, H. Bouhlal, et al., “The retinoblastoma (Rb) protein regulates ferroptosis induced by sorafenib in human hepatocellular carcinoma cells,” Cancer Letters 356, no. 2 Pt B (2015): 971-977.

[125]

S. M. Wilhelm, L. Adnane, P. Newell, A. Villanueva, J. M. Llovet, and M. Lynch, “Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling,” Molecular Cancer Therapeutics 7, no. 10 (2008): 3129-3140.

[126]

G. M. Keating and A. Santoro, “Sorafenib: A review of its use in advanced hepatocellular carcinoma,” Drugs 69, no. 2 (2009): 223-240.

[127]

Y. Zhang, D. Xue, X. Wang, M. Lu, B. Gao, and X. Qiao, “Screening of kinase inhibitors targeting BRAF for regulating autophagy based on kinase pathways,” Mol Med Rep 9, no. 1 (2014): 83-90.

[128]

S. Dolma, S. L. Lessnick, W. C. Hahn, and B. R. Stockwell, “Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells,” Cancer Cell 3, no. 3 (2003): 285-296.

[129]

W. S. Yang, R. SriRamaratnam, M. E. Welsch, et al., “Regulation of ferroptotic cancer cell death by GPX4,” Cell 156, no. 1-2 (2014): 317-331.

[130]

D. Moosmayer, A. Hilpmann, J. Hoffmann, et al., “Crystal structures of the selenoprotein glutathione peroxidase 4 in its apo form and in complex With the covalently bound inhibitor ML162,” Acta Crystallogr Sect Struct Biol 77, no. Pt 2 (2021): 237-248.

[131]

Y. Wu, S. Zhang, X. Gong, et al., “The epigenetic regulators and metabolic changes in ferroptosis-associated cancer progression,” Molecular cancer 19 (2020): 39.

[132]

H. Yamaguchi, J. L. Hsu, C. T. Chen, et al., “Caspase-independent cell death is involved in the negative effect of EGF receptor inhibitors on cisplatin in non-small cell lung cancer cells,” Clin Cancer Res Off J Am Assoc Cancer Res 19, no. 4 (2013): 845-854.

[133]

M. Y. Kwon, E. Park, S. J. Lee, and S. W. Chung, “Heme oxygenase-1 accelerates erastin-induced ferroptotic cell death,” Oncotarget 6, no. 27 (2015): 24393-24403.

[134]

P. W. Gout, A. R. Buckley, C. R. Simms, and N. Bruchovsky, “Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the x(c)- cystine transporter: A new action for an old drug,” Leukemia 15, no. 10 (2001): 1633-1640.

[135]

R. Skouta, S. J. Dixon, J. Wang, et al., “Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models,” Journal of the American Chemical Society 136, no. 12 (2014): 4551-4556.

[136]

A. Hinman, C. R. Holst, J. C. Latham, et al., “Vitamin E hydroquinone is an endogenous regulator of ferroptosis via redox control of 15-lipoxygenase,” PLoS ONE 13, no. 8 (2018): e0201369.

[137]

R. Kang, L. Zeng, S. Zhu, et al., “Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis,” Cell Host & Microbe 24, no. 1 (2018): 97-108. e4.

[138]

D. Wallach, T. B. Kang, and A. Kovalenko, “Concepts of tissue injury and cell death in inflammation: A historical perspective,” Nature Reviews Immunology 14, no. 1 (2014): 51-59.

[139]

B. Proneth and M. Conrad, “Ferroptosis and necroinflammation, a yet poorly explored link,” Cell Death and Differentiation 26, no. 1 (2019): 14-24.

[140]

Y. Qiu, Y. Cao, W. Cao, Y. Jia, and N. Lu, “The Application of Ferroptosis in Diseases,” Pharmacological Research 159 (2020): 104919.

[141]

Y. Sun, P. Chen, B. Zhai, et al., “The emerging role of ferroptosis in inflammation,” Biomedicine & Pharmacotherapy 127 (2020): 110108.

[142]

H. Mao, Y. Zhao, H. Li, and L. Lei, “Ferroptosis as an emerging target in inflammatory diseases,” Progress in Biophysics and Molecular Biology 155 (2020): 20-28.

[143]

W. S. Yang, K. J. Kim, M. M. Gaschler, M. Patel, M. S. Shchepinov, and B. R. Stockwell, “Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis,” PNAS 113, no. 34 (2016): E4966-E4975.

[144]

J. Y. Lee, W. K. Kim, K. H. Bae, S. C. Lee, and E. W. Lee, “Lipid Metabolism and Ferroptosis,” Biology 10, no. 3 (2021): 184.

[145]

H. Mao, Y. Zhao, H. Li, and L. Lei, “Ferroptosis as an emerging target in inflammatory diseases,” Progress in Biophysics and Molecular Biology 155 (2020): 20-28.

[146]

Y. Sun, P. Chen, B. Zhai, et al., “The emerging role of ferroptosis in inflammation,” Biomedicine & Pharmacotherapy 127 (2020): 110108.

[147]

J. Loscalzo, “Membrane Redox State and Apoptosis: Death by Peroxide,” Cell metabolism 8, no. 3 (2008): 182-183.

[148]

G. Folco and R. C. Murphy, “Eicosanoid transcellular biosynthesis: From cell-cell interactions to in vivo tissue responses,” Pharmacological Reviews 58, no. 3 (2006): 375-388.

[149]

B. Proneth and M. Conrad, “Ferroptosis and necroinflammation, a yet poorly explored link,” Cell Death and Differentiation 26, no. 1 (2019): 14-24.

[150]

H. H. Dar, Y. Y. Tyurina, K. Mikulska-Ruminska, et al., “Pseudomonas aeruginosa utilizes host polyunsaturated phosphatidylethanolamines to trigger theft-ferroptosis in bronchial epithelium,” Journal of Clinical Investigation 128, no. 10 (2019): 4639-4653.

[151]

I. Splichal, S. M. Donovan, V. Jenistova, et al., “High Mobility Group Box 1 and TLR4 Signaling Pathway in Gnotobiotic Piglets Colonized/Infected With L. amylovorus, L. mucosae, E. coli Nissle 1917 and S. Typhimurium,” International Journal of Molecular Sciences 20, no. 24 (2019): 6294.

[152]

Q. Wen, J. Liu, R. Kang, B. Zhou, and D. Tang, “The release and activity of HMGB1 in ferroptosis,” Biochemical and Biophysical Research Communications 510, no. 2 (2019): 278-283.

[153]

Y. Sun, P. Chen, B. Zhai, et al., “The emerging role of ferroptosis in inflammation,” Biomedicine & Pharmacotherapy 127 (2020): 110108.

[154]

S. J. Dixon, K. M. Lemberg, M. R. Lamprecht, et al., “Ferroptosis: An iron-dependent form of nonapoptotic cell death,” Cell 149, no. 5 (2012): 1060-1072.

[155]

E. M. Kenny, E. Fidan, Q. Yang, et al., “Ferroptosis Contributes to Neuronal Death and Functional Outcome After Traumatic Brain Injury∗,” Critical Care Medicine 47, no. 3 (2019): 410-418.

[156]

Z. Zhang, Y. Wu, S. Yuan, et al., “Glutathione peroxidase 4 participates in secondary brain injury Through mediating ferroptosis in a rat model of intracerebral hemorrhage,” Brain Research 1701 (2018): 112-125.

[157]

D. Martin-Sanchez, O. Ruiz-Andres, J. Poveda, et al., “Ferroptosis, but not necroptosis, is important in nephrotoxic folic acid-induced AKI,” Journal of the American Society of Nephrology 28, no. 1 (2017): 218-229.

[158]

S. Masaldan, A. A. Belaidi, S. Ayton, and A. I. Bush, “Cellular senescence and iron dyshomeostasis in alzheimer's disease,” Pharmaceuticals 12, no. 2 (2019): 93.

[159]

M. Sarhan, W. G. Land, W. Tonnus, C. P. Hugo, and A. Linkermann, “Origin and Consequences of Necroinflammation,” Physiological Reviews 98, no. 2 (2018): 727-780.

[160]

A. Linkermann, “Death and fire-the concept of necroinflammation,” Cell Death and Differentiation 26, no. 1 (2019): 1-3.

[161]

T. Gong, L. Liu, W. Jiang, and R. Zhou, “DAMP-sensing receptors in sterile inflammation and inflammatory diseases,” Nature Reviews Immunology 20, no. 2 (2020): 95-112.

[162]

F. Pandolfi, S. Altamura, S. Frosali, and P. Conti, “Key Role of DAMP in Inflammation, Cancer, and Tissue Repair,” Clinical Therapeutics 38, no. 5 (2016): 1017-1028.

[163]

S. Reuter, S. C. Gupta, M. M. Chaturvedi, and B. B. Aggarwal, “Oxidative stress, inflammation, and cancer: How are they linked?,” Free Radic Biol Med 49, no. 11 (2010): 1603-1616.

[164]

T. W. Mak, M. Grusdat, G. S. Duncan, et al., “Glutathione Primes T Cell Metabolism for Inflammation,” Immunity 46, no. 4 (2017): 675-689.

[165]

C. J. Chen, H. S. Huang, and W. C. Chang, “Depletion of phospholipid hydroperoxide glutathione peroxidase up-regulates arachidonate metabolism by 12S-lipoxygenase and cyclooxygenase 1 in human epidermoid carcinoma A431 cells,” FASEB J Off Publ Fed Am Soc Exp Biol 17, no. 12 (2003): 1694-1696.

[166]

H. Sakamoto, H. Imai, and Y. Nakagawa, “Involvement of phospholipid hydroperoxide glutathione peroxidase in the modulation of prostaglandin D2 synthesis,” Journal of Biological Chemistry 275, no. 51 (2000): 40028-40035.

[167]

T. W. Mak, M. Grusdat, G. S. Duncan, et al., “Glutathione Primes T Cell Metabolism for Inflammation,” Immunity 46, no. 4 (2017): 675-689.

[168]

C. Li, X. Deng, X. Xie, Y. Liu, J. P. Friedmann Angeli, and L. Lai, “Activation of Glutathione Peroxidase 4 as a Novel Anti-inflammatory Strategy,” Frontiers in pharmacology 9 (2018): 1120.

[169]

H. Suzuki, Y. Kayama, M. Sakamoto, et al., “Arachidonate 12/15-lipoxygenase-induced inflammation and oxidative stress are involved in the development of diabetic cardiomyopathy,” Diabetes 64, no. 2 (2015): 618-630.

[170]

Y. Zhou, K. T. Que, Z. Zhang, et al., “Iron overloaded polarizes macrophage to proinflammation phenotype Through ROS/acetyl-p53 pathway,” Cancer medicine 7, no. 8 (2018): 4012-4022.

[171]

P. Handa, S. Thomas, V. Morgan-Stevenson, et al., “Iron alters macrophage polarization status and leads to steatohepatitis and fibrogenesis,” J Leukoc Biol 105, no. 5 (2019): 1015-1026.

[172]

S. Islam, S. Jarosch, J. Zhou, et al., “Anti-inflammatory and anti-bacterial effects of iron chelation in experimental sepsis,” Journal of Surgical Research 200, no. 1 (2016): 266-273.

[173]

T. Thorburn, M. Aali, L. Kostek, et al., “Anti-inflammatory effects of a novel iron chelator, DIBI, in experimental sepsis,” Clinical Hemorheology and Microcirculation 67, no. 3-4 (2017): 241-250.

[174]

X. Hu, J. Li, M. Fu, X. Zhao, and W. Wang, “The JAK/STAT signaling pathway: From bench to clinic,” Signal Transduct Target Ther 6, no. 1 (2021): 402.

[175]

Y. Luo, M. Alexander, M. Gadina, J. J. O'Shea, F. Meylan, and D. M. Schwartz, “JAK-STAT signaling in human disease: From genetic syndromes to clinical inhibition,” Journal of Allergy and Clinical Immunology 148, no. 4 (2021): 911-925.

[176]

N. Awasthi, C. Liongue, and A. C. Ward, “STAT proteins: A kaleidoscope of canonical and non-canonical functions in immunity and cancer,” J Hematol OncolJ Hematol Oncol 14, no. 1 (2021): 198.

[177]

P. Xin, X. Xu, C. Deng, et al., “The role of JAK/STAT signaling pathway and its inhibitors in diseases,” International Immunopharmacology 80 (2020): 106210.

[178]

R. Kong, N. Wang, W. Han, W. Bao, and J. Lu, “IFNγ-mediated repression of system xc- drives vulnerability to induced ferroptosis in hepatocellular carcinoma cells,” J Leukoc Biol 110, no. 2 (2021): 301-314.

[179]

W. Wang, M. Green, J. E. Choi, et al., “CD8+ T cells regulate tumour ferroptosis During cancer immunotherapy,” Nature 569, no. 7755 (2019): 270-274.

[180]

Y. Zhang, Y. Zuo, B. Li, et al., “Propofol prevents oxidative stress and apoptosis by regulating iron homeostasis and targeting JAK/STAT3 signaling in SH-SY5Y cells,” Brain Research Bulletin 153 (2019): 191-201.

[181]

Y. P. Liu, Z. Z. Qiu, X. H. Li, and E. Y. Li, “Propofol induces ferroptosis and inhibits malignant phenotypes of gastric cancer cells by regulating miR-125b-5p/STAT3 axis,” World journal of gastrointestinal oncology 13, no. 12 (2021): 2114-2128.

[182]

M. Y. Hsu, E. Mina, A. Roetto, and P. E. Porporato, “Iron: An Essential Element of Cancer Metabolism,” Cells 9, no. 12 (2020): 2591.

[183]

Y. Chen, F. Wang, P. Wu, et al., “Artesunate induces apoptosis, autophagy and ferroptosis in diffuse large B cell lymphoma cells by impairing STAT3 signaling,” Cell. Signalling 88 (2021): 110167.

[184]

K. V. Kowdley, E. M. Gochanour, V. Sundaram, R. A. Shah, and P. Handa, “Hepcidin Signaling in Health and Disease: Ironing Out the Details,” Hepatol Commun 5, no. 5 (2021): 723-735.

[185]

L. Yang, H. Wang, X. Yang, et al., “Auranofin mitigates systemic iron overload and induces ferroptosis via distinct mechanisms,” Signal Transduct Target Ther 5, no. 1 (2020): 138.

[186]

E. Saint-Germain, L. Mignacca, M. Vernier, D. Bobbala, S. Ilangumaran, and G. Ferbeyre, “SOCS1 regulates senescence and ferroptosis by modulating the expression of p53 target genes,” Aging 9, no. 10 (2017): 2137-2162.

[187]

L. Opazo-Ríos, Y. Sanchez Matus, R. R. Rodrigues-Díez, et al., “Anti-inflammatory, antioxidant and renoprotective effects of SOCS1 mimetic peptide in the BTBR ob/ob mouse model of type 2 diabetes,” BMJ Open Diabetes Res Care 8, no. 1 (2020): e001242.

[188]

A. Tefferi, “Primary myelofibrosis: 2021 update on diagnosis, risk-stratification and management,” American Journal of Hematology 96, no. 1 (2021): 145-162.

[189]

Q. Zhang, M. J. Lenardo, and D. Baltimore, “30 Years of NF-κB: A Blossoming of Relevance to Human Pathobiology,” Cell 168, no. 1-2 (2017): 37-57.

[190]

J. P. Mitchell and R. J. Carmody, “NF-κB and the Transcriptional Control of Inflammation,” Int Rev Cell Mol Biol 335 (2018): 41-84.

[191]

H. Yu, L. Lin, Z. Zhang, H. Zhang, and H. Hu, “Targeting NF-κB pathway for the therapy of diseases: Mechanism and clinical study,” Signal Transduct Target Ther 5, no. 1 (2020): 209.

[192]

X. Zhong, Z. Zhang, H. Shen, et al., “Hepatic NF-κB-Inducing Kinase and Inhibitor of NF-κB Kinase Subunit α Promote Liver Oxidative Stress, Ferroptosis, and Liver Injury,” Hepatol Commun 5, no. 10 (2021): 1704-1720.

[193]

F. Yao, Y. Deng, Y. Zhao, et al., “A targetable LIFR-NF-κB-LCN2 axis controls liver tumorigenesis and vulnerability to ferroptosis,” Nature Communications 12, no. 1 (2021): 7333.

[194]

N. Yan, Z. Xu, C. Qu, and J. Zhang, “Dimethyl fumarate improves cognitive deficits in chronic cerebral hypoperfusion rats by alleviating inflammation, oxidative stress, and ferroptosis via NRF2/ARE/NF-κB signal pathway,” International Immunopharmacology 98 (2021): 107844.

[195]

S. Li, Y. He, K. Chen, et al., “RSL3 Drives Ferroptosis Through NF-κB Pathway Activation and GPX4 Depletion in Glioblastoma,” Oxid Med Cell Longev 2021 (2021): 2915019.

[196]

B. M. Oh, S. J. Lee, G. L. Park, et al., “Erastin Inhibits Septic Shock and Inflammatory Gene Expression via Suppression of the NF-κB Pathway,” Journal of Clinical Medicine 8, no. 12 (2019): 2210.

[197]

K. H. Pulkkinen, S. Ylä-Herttuala, and A. L. Levonen, “Heme oxygenase 1 is induced by miR-155 via reduced BACH1 translation in endothelial cells,” Free Radic Biol Med 51, no. 11 (2011): 2124-2131.

[198]

M. P. Seldon, G. Silva, N. Pejanovic, et al., “Heme oxygenase-1 inhibits the expression of adhesion molecules associated With endothelial cell activation via inhibition of NF-kappaB RelA phosphorylation at serine 276,” J Immunol Baltim Md 1950 179, no. 11 (2007): 7840-7851.

[199]

Y. Guan, X. Zhao, W. Liu, and Y. Wang, “Galuteolin suppresses proliferation and inflammation in TNF-α-induced RA-FLS cells by activating HMOX1 to regulate IKKβ/NF-κB pathway,” J Orthop Surg 15, no. 1 (2020): 484.

[200]

X. Kou, Y. Jing, W. Deng, et al., “Tumor necrosis factor-α attenuates starvation-induced apoptosis Through upregulation of ferritin heavy chain in hepatocellular carcinoma cells,” BMC cancer 13 (2013): 438.

[201]

A. Zarjou, L. M. Black, K. R. McCullough, et al., “Ferritin Light Chain Confers Protection Against Sepsis-Induced Inflammation and Organ Injury,” Frontiers in immunology 10 (2019): 131.

[202]

V. A. K. Rathinam and K. A. Fitzgerald, “Inflammasome Complexes: Emerging Mechanisms and Effector Functions,” Cell 165, no. 4 (2016): 792-800.

[203]

O. Schnappauf, J. J. Chae, D. L. Kastner, and I. Aksentijevich, “The Pyrin Inflammasome in Health and Disease,” Frontiers in immunology 10 (2019): 1745.

[204]

D. Chauhan, L. Vande Walle, and M. Lamkanfi, “Therapeutic modulation of inflammasome pathways,” Immunological Reviews 297, no. 1 (2020): 123-138.

[205]

U. Gupta, S. Ghosh, C. T. Wallace, et al., “Increased LCN2 (lipocalin 2) in the RPE decreases autophagy and activates inflammasome-ferroptosis processes in a mouse model of dry AMD,” Autophagy 19, no. 1 (2023): 92-111.

[206]

S. S. Xie, Y. Deng, S. L. Guo, et al., “Endothelial cell ferroptosis mediates monocrotaline-induced pulmonary hypertension in rats by modulating NLRP3 inflammasome activation,” Scientific Reports 12, no. 1 (2022): 3056.

[207]

L. Meihe, G. Shan, K. Minchao, et al., “The Ferroptosis-NLRP1 Inflammasome: The Vicious Cycle of an Adverse Pregnancy,” Frontiers in Cell and Developmental Biology 9 (2021): 707959.

[208]

M. Orecchioni, K. Kobiyama, H. Winkels, et al., “Olfactory receptor 2 in vascular macrophages drives atherosclerosis by NLRP3-dependent IL-1 production,” Science 375, no. 6577 (2022): 214-221.

[209]

S. Chokchaiwong, Y. T. Kuo, S. H. Lin, et al., “Coenzyme Q10 serves to couple mitochondrial oxidative phosphorylation and fatty acid β-oxidation, and attenuates NLRP3 inflammasome activation,” Free Radic Res 52, no. 11-12 (2018): 1445-1455.

[210]

C. G. Hsu, C. L. Chávez, C. Zhang, M. Sowden, C. Yan, and B. C. Berk, “The lipid peroxidation product 4-hydroxynonenal inhibits NLRP3 inflammasome activation and macrophage pyroptosis,” Cell Death and Differentiation 29, no. 9 (2022): 1790-1803.

[211]

Y. Zhou, H. Zhou, L. Hua, et al., “Verification of ferroptosis and pyroptosis and identification of PTGS2 as the hub gene in human coronary artery atherosclerosis,” Free Radic Biol Med 171 (2021): 55-68.

[212]

R. Kang, L. Zeng, S. Zhu, et al., “Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis,” Cell Host & Microbe 24, no. 1 (2018): 97-108.

[213]

R. Fan, J. Sui, X. Dong, B. Jing, and Z. Gao, “Wedelolactone alleviates acute pancreatitis and associated lung injury via GPX4 mediated suppression of pyroptosis and ferroptosis,” Free Radic Biol Med 173 (2021): 29-40.

[214]

J. S. C. Arthur and S. C. Ley, “Mitogen-activated protein kinases in innate immunity,” Nature Reviews Immunology 13, no. 9 (2013): 679-692.

[215]

M. Gaestel, “MAPK-Activated Protein Kinases (MKs): Novel Insights and Challenges,” Frontiers in Cell and Developmental Biology 3 (2015): 88.

[216]

A. Martínez-Limón, M. Joaquin, M. Caballero, F. Posas, and E. de Nadal, “The p38 Pathway: From Biology to Cancer Therapy,” International Journal of Molecular Sciences 21, no. 6 (2020): 1913.

[217]

K. Zhu, X. Zhu, S. Sun, et al., “Inhibition of TLR4 prevents hippocampal hypoxic-ischemic injury by regulating ferroptosis in neonatal rats,” Experimental Neurology 345 (2021): 113828.

[218]

K. Zhu, X. Zhu, S. Sun, et al., “Inhibition of TLR4 prevents hippocampal hypoxic-ischemic injury by regulating ferroptosis in neonatal rats,” Experimental Neurology 345 (2021): 113828.

[219]

D. U. Kim, D. G. Kim, J. W. Choi, et al., “Loganin Attenuates the Severity of Acute Kidney Injury Induced by Cisplatin Through the Inhibition of ERK Activation in Mice,” International Journal of Molecular Sciences 22, no. 3 (2021): 1421.

[220]

Y. Ikeda, A. Satoh, Y. Horinouchi, et al., “Iron accumulation causes impaired myogenesis correlated With MAPK signaling pathway inhibition by oxidative stress,” FASEB J Off Publ Fed Am Soc Exp Biol 33, no. 8 (2019): 9551-9564.

[221]

S. A. Salama and A. M. Kabel, “Taxifolin ameliorates iron overload-induced hepatocellular injury: Modulating PI3K/AKT and p38 MAPK signaling, inflammatory response, and hepatocellular regeneration,” Chemico-Biological Interactions 330 (2020): 109230.

[222]

Z. Cavdar, M. A. Oktan, C. Ural, et al., “Renoprotective Effects of Alpha Lipoic Acid on Iron Overload-Induced Kidney Injury in Rats by Suppressing NADPH Oxidase 4 and p38 MAPK Signaling,” Biological Trace Element Research 193, no. 2 (2020): 483-493.

[223]

S. Fu, R. Lv, L. Wang, H. Hou, H. Liu, and S. Shao, “Resveratrol, an antioxidant, protects spinal cord injury in rats by suppressing MAPK pathway,” Saudi J Biol Sci 25, no. 2 (2018): 259-266.

[224]

S. Doll, F. P. Freitas, R. Shah, et al., “FSP1 is a glutathione-independent ferroptosis suppressor,” Nature 575, no. 7784 (2019): 693-698.

[225]

E. H. Sidhom, C. Kim, M. Kost-Alimova, et al., “Targeting a Braf/Mapk pathway rescues podocyte lipid peroxidation in CoQ-deficiency kidney disease,” Journal of Clinical Investigation 131, no. 5 (2021): e141380. 141380.

[226]

F. Huang, S. Zhang, X. Li, Y. Huang, S. He, and L. Luo, “STAT3-mediated ferroptosis is involved in ulcerative colitis,” Free Radic Biol Med 188 (2022): 375-385.

[227]

Y. Chen, P. Zhang, W. Chen, and G. Chen, “Ferroptosis mediated DSS-induced ulcerative colitis associated With Nrf2/HO-1 signaling pathway,” Immunology Letters 225 (2020): 9-15.

[228]

M. Xu, J. Tao, Y. Yang, et al., “Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis,” Cell death & disease 11, no. 2 (2020): 86.

[229]

J. V. Patankar and C. Becker, “Cell death in the gut epithelium and implications for chronic inflammation,” Nature reviews Gastroenterology & hepatology 17, no. 9 (2020): 543-556.

[230]

J. Huang, J. Zhang, J. Ma, et al., “Inhibiting Ferroptosis: A Novel Approach for Ulcerative Colitis Therapeutics,” Oxid Med Cell Longev 2022 (2022): 9678625.

[231]

Y. Chen, P. Zhang, W. Chen, and G. Chen, “Ferroptosis mediated DSS-induced ulcerative colitis associated With Nrf2/HO-1 signaling pathway,” Immunology Letters 225 (2020): 9-15.

[232]

L. Luo, S. Zhang, N. Guo, H. Li, and S. He, “ACSF2-mediated ferroptosis is involved in ulcerative colitis,” Life Sciences 313 (2023): 121272.

[233]

D. J. Cui, C. Chen, W. Q. Yuan, Y. H. Yang, and L. Han, “Integrative analysis of ferroptosis-related genes in ulcerative colitis,” Journal of International Medical Research 49, no. 9 (2021): 3000605211042975.

[234]

Y. Chen, P. Zhang, W. Chen, and G. Chen, “Ferroptosis mediated DSS-induced ulcerative colitis associated With Nrf2/HO-1 signaling pathway,” Immunology Letters 225 (2020): 9-15.

[235]

H. Ma, Q. Shu, D. Li, et al., “Accumulation of Intracellular Ferrous Iron in Inflammatory-Activated Macrophages,” Biological Trace Element Research 201, no. 5 (2023): 2303-2310.

[236]

M. Gao, P. Monian, Q. Pan, W. Zhang, J. Xiang, and X. Jiang, “Ferroptosis is an autophagic cell death process,” Cell Research 26, no. 9 (2016): 1021-1032.

[237]

M. Xu, J. Tao, Y. Yang, et al., “Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis,” Cell death & disease 11, no. 2 (2020): 86.

[238]

S. K. Panda, V. Peng, R. Sudan, et al., “Repression of the aryl-hydrocarbon receptor prevents oxidative stress and ferroptosis of intestinal intraepithelial lymphocytes,” Immunity 56, no. 4 (2023): 797-812.

[239]

M. Xu, J. Tao, Y. Yang, et al., “Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis,” Cell death & disease 11, no. 2 (2020): 86.

[240]

J. H. Lin, P. Walter, and T. S. B. Yen, “Endoplasmic reticulum stress in disease pathogenesis,” Annu Rev Pathol 3 (2008): 399-425.

[241]

Y. Hu, S. Sun, and H. Li. Identification and functional exploration of Ferroptosis and Immune Related Long Non-Coding RNA in Inflammatory bowel diseas. Published online 2022. https://doi.org/10.21203/rs.3.rs-1857506/v1

[242]

J. M. O'Brien, N. A. Ali, S. K. Aberegg, and E. Abraham, “Sepsis,” American Journal of Medicine 120, no. 12 (2007): 1012-1022.

[243]

D. Ma, P. Jiang, Y. Jiang, H. Li, and D. Zhang, “Effects of Lipid Peroxidation-Mediated Ferroptosis on Severe Acute Pancreatitis-Induced Intestinal Barrier Injury and Bacterial Translocation,” Oxid Med Cell Longev 2021 (2021): 6644576.

[244]

R. Ma, L. Fang, L. Chen, X. Wang, J. Jiang, and L. Gao, “Ferroptotic stress promotes macrophages Against intracellular bacteria,” Theranostics 12, no. 5 (2022): 2266-2289.

[245]

M. Matsushita, S. Freigang, C. Schneider, M. Conrad, G. W. Bornkamm, and M. Kopf, “T cell lipid peroxidation induces ferroptosis and prevents immunity to infection,” Journal of Experimental Medicine 212, no. 4 (2015): 555-568.

[246]

N. Li, W. Wang, H. Zhou, et al., “Ferritinophagy-mediated ferroptosis is involved in sepsis-induced cardiac injury,” Free Radic Biol Med 160 (2020): 303-318.

[247]

C. Wang, W. Yuan, A. Hu, et al., “Dexmedetomidine alleviated sepsis‑induced myocardial ferroptosis and septic heart injury,” Mol Med Rep 22, no. 1 (2020): 175-184.

[248]

J. Fang, B. Kong, W. Shuai, et al., “Ferroportin-mediated ferroptosis involved in new-onset atrial fibrillation With LPS-induced endotoxemia,” European Journal of Pharmacology 913 (2021): 174622.

[249]

Y. Zhang, L. Zheng, H. Deng, et al., “Electroacupuncture Alleviates LPS-Induced ARDS Through α7 Nicotinic Acetylcholine Receptor-Mediated Inhibition of Ferroptosis,” Frontiers in immunology 13 (2022): 832432.

[250]

N. N. Liang, Y. Zhao, Y. Y. Guo, et al., “Mitochondria-derived reactive oxygen species are involved in renal cell ferroptosis During lipopolysaccharide-induced acute kidney injury,” International Immunopharmacology 107 (2022): 108687.

[251]

Z. Xie, M. Xu, J. Xie, et al., “Inhibition of Ferroptosis Attenuates Glutamate Excitotoxicity and Nuclear Autophagy in a CLP Septic Mouse Model,” Shock Augusta Ga 57, no. 5 (2022): 694-702.

[252]

J. Xiao, Q. Yang, Y. Zhang, et al., “Maresin conjugates in tissue regeneration-1 suppresses ferroptosis in septic acute kidney injury,” Cell Biosci 11, no. 1 (2021): 221.

[253]

Z. Xie, M. Xu, J. Xie, et al., “Inhibition of Ferroptosis Attenuates Glutamate Excitotoxicity and Nuclear Autophagy in a CLP Septic Mouse Model,” Shock Augusta Ga 57, no. 5 (2022): 694-702.

[254]

S. Wei, J. Bi, L. Yang, et al., “Serum irisin levels are decreased in patients With sepsis, and exogenous irisin suppresses ferroptosis in the liver of septic mice,” Clinical and translational medicine 10, no. 5 (2020): e173.

[255]

X. B. Wei, W. Q. Jiang, J. H. Zeng, et al., “Exosome-Derived lncRNA NEAT1 Exacerbates Sepsis-Associated Encephalopathy by Promoting Ferroptosis Through Regulating miR-9-5p/TFRC and GOT1 Axis,” Molecular Neurobiology 59, no. 3 (2022): 1954-1969.

[256]

J. A. Kellum, P. Romagnani, G. Ashuntantang, C. Ronco, A. Zarbock, and H. J. Anders, “Acute kidney injury,” Nat Rev Dis Primer 7, no. 1 (2021): 1-17.

[257]

K. Lee, H. R. Jang, J. Jeon, et al., “Repair phase modeling of ischemic acute kidney injury: Recovery vs. transition to chronic kidney disease,” American journal of translational research 14, no. 1 (2022): 554.

[258]

Q. Wei, H. Sun, S. Song, et al., “MicroRNA-668 represses MTP18 to preserve mitochondrial dynamics in ischemic acute kidney injury,” Journal of Clinical Investigation 128, no. 12: 5448-5464.

[259]

S. R. Mulay, M. M. Honarpisheh, O. Foresto-Neto, et al., “Mitochondria Permeability Transition versus Necroptosis in Oxalate-Induced AKI,” J Am Soc Nephrol JASN 30, no. 10 (2019): 1857-1869.

[260]

G. P. Kaushal, “Autophagy protects proximal tubular cells From injury and apoptosis,” Kidney International 82, no. 12 (2012): 1250-1253.

[261]

A. B. Sanz, M. D. Sanchez-Niño, A. M. Ramos, and A. Ortiz, “Regulated cell death pathways in kidney disease,” Nature Reviews Nephrology 19, no. 5 (2023): 281-299.

[262]

J. Guerrero-Mauvecin, N. Villar-Gómez, S. Rayego-Mateos, et al., “Regulated necrosis role in inflammation and repair in acute kidney injury,” Frontiers in immunology 14 (2023): 1324996.

[263]

D. Martin-Sanchez, M. Fontecha-Barriuso, J. M. Martinez-Moreno, et al., “Ferroptosis and kidney disease,” Nefrologia: Publicacion Oficial De La Sociedad Espanola Nefrologia 40, no. 4 (2020): 384-394.

[264]

D. Martin-Sanchez, O. Ruiz-Andres, J. Poveda, et al., “Ferroptosis, but Not Necroptosis, Is Important in Nephrotoxic Folic Acid-Induced AKI,” J Am Soc Nephrol JASN 28, no. 1 (2017): 218-229.

[265]

Z. Hu, H. Zhang, S. K. Yang, et al., “Emerging Role of Ferroptosis in Acute Kidney Injury,” Oxid Med Cell Longev 2019 (2019): 8010614.

[266]

J. Li, S. Zheng, Y. Fan, and K. Tan, “Emerging significance and therapeutic targets of ferroptosis: A potential avenue for human kidney diseases,” Cell death & disease 14, no. 9 (2023): 628.

[267]

Y. Wang, M. Zhang, R. Bi, et al., “ACSL4 deficiency confers protection Against ferroptosis-mediated acute kidney injury,” Redox Biology 51 (2022): 102262.

[268]

W. H. Tao, X. S. Shan, J. X. Zhang, et al., “Dexmedetomidine Attenuates Ferroptosis-Mediated Renal Ischemia/Reperfusion Injury and Inflammation by Inhibiting ACSL4 via α2-AR,” Frontiers in pharmacology 13 (2022): 782466.

[269]

Z. Sun, J. Wu, Q. Bi, and W. Wang, “Exosomal lncRNA TUG1 derived From human urine-derived stem cells attenuates renal ischemia/reperfusion injury by interacting With SRSF1 to regulate ASCL4-mediated ferroptosis,” Stem Cell Res Ther 13, no. 1 (2022): 297.

[270]

L. Huang, L. Zhang, Z. Zhang, et al., “Loss of nephric augmenter of liver regeneration facilitates acute kidney injury via ACSL4-mediated ferroptosis,” Journal of Cellular and Molecular Medicine (2023), https://doi.org/10.1111/jcmm.18076. Published online December 13.

[271]

L. L. Huang, R. T. Long, G. P. Jiang, et al., “Augmenter of liver regeneration promotes mitochondrial biogenesis in renal ischemia-reperfusion injury,” Apoptosis Int J Program Cell Death 23, no. 11-12 (2018): 695-706.

[272]

D. J. Zhu, X. H. Liao, W. Q. Huang, H. Sun, L. Zhang, and Q. Liu, “Augmenter of Liver Regeneration Protects Renal Tubular Epithelial Cells From Ischemia-Reperfusion Injury by Promoting PINK1/Parkin-Mediated Mitophagy,” Front Physiol 11 (2020): 178.

[273]

L. L. Huang, X. H. Liao, H. Sun, X. Jiang, Q. Liu, and L. Zhang, “Augmenter of liver regeneration protects the kidney From ischaemia-reperfusion injury in ferroptosis,” Journal of Cellular and Molecular Medicine 23, no. 6 (2019): 4153-4164.

[274]

X. Sun, N. Huang, P. Li, et al., “TRIM21 ubiquitylates GPX4 and promotes ferroptosis to aggravate ischemia/reperfusion-induced acute kidney injury,” Life Sciences 321 (2023): 121608.

[275]

R. Feng, Y. Xiong, Y. Lei, et al., “Lysine-specific demethylase 1 aggravated oxidative stress and ferroptosis induced by renal ischemia and reperfusion injury Through activation of TLR4/NOX4 pathway in mice,” Journal of Cellular and Molecular Medicine 26, no. 15 (2022): 4254-4267.

[276]

A. Ozkok and C. L. Edelstein, “Pathophysiology of cisplatin-induced acute kidney injury,” BioMed research international 2014 (2014): 967826.

[277]

F. Deng, I. Sharma, Y. Dai, M. Yang, and Y. S. Kanwar, “Myo-inositol oxygenase expression profile modulates pathogenic ferroptosis in the renal proximal tubule,” Journal of Clinical Investigation 129, no. 11 (2019): 5033-5049.

[278]

J. H. Baek, A. Yalamanoglu, R. P. Brown, D. M. Saylor, R. A. Malinauskas, and P. W. Buehler, “Renal Toxicodynamic Effects of Extracellular Hemoglobin After Acute Exposure,” Toxicol Sci Off J Soc Toxicol 166, no. 1 (2018): 180-191.

[279]

L. Zhou, P. Yu, T. T. Wang, et al., “Polydatin Attenuates Cisplatin-Induced Acute Kidney Injury by Inhibiting Ferroptosis,” Oxid Med Cell Longev 2022 (2022): 9947191.

[280]

Y. Ma, L. Huang, Z. Zhang, et al., “CD36 promotes tubular ferroptosis by regulating the ubiquitination of FSP1 in acute kidney injury,” Genes Dis 11, no. 1 (2023): 449-463.

[281]

L. J. Yan, “Folic acid-induced animal model of kidney disease,” Anim Models Exp Med 4, no. 4 (2021): 329-342.

[282]

R. Kandel and K. P. Singh, “Higher Concentrations of Folic Acid Cause Oxidative Stress, Acute Cytotoxicity, and Long-Term Fibrogenic Changes in Kidney Epithelial Cells,” Chem. Res. Toxicol. 35, no. 11 (2022): 2168-2179.

[283]

D. Li, B. Liu, Y. Fan, et al., “Nuciferine protects Against folic acid-induced acute kidney injury by inhibiting ferroptosis,” British Journal of Pharmacology 178, no. 5 (2021): 1182-1199.

[284]

X. Yang, S. Dong, Y. Fan, et al., “Krüppel-Like Factor 15 Suppresses Ferroptosis by Activating an NRF2/GPX4 Signal to Protect Against Folic Acid-Induced Acute Kidney Injury,” International Journal of Molecular Sciences 24, no. 19 (2023): 14530.

[285]

L. Zhang, F. Chen, J. Dong, et al., “HDAC3 aberration-incurred GPX4 suppression drives renal ferroptosis and AKI-CKD progression,” Redox Biology 68 (2023): 102939.

[286]

R. Vanholder, M. Sükrü Sever, and N. Lameire, “Kidney problems in disaster situations,” Nephrol Ther 17S (2021): S27-S36.

[287]

G. A. Herrera, “Myoglobin and the kidney: An overview,” Ultrastructural Pathology 18, no. 1-2 (1994): 113-117.

[288]

R. A. Zager and K. Burkhart, “Myoglobin toxicity in proximal human kidney cells: Roles of Fe, Ca2+, H2O2, and terminal mitochondrial electron transport,” Kidney International 51, no. 3 (1997): 728-738.

[289]

X. Bosch, E. Poch, and J. M. Grau, “Rhabdomyolysis and acute kidney injury,” New England Journal of Medicine 361, no. 1 (2009): 62-72.

[290]

H. Zhu, J. Cen, C. Hong, et al., “Targeting Labile Iron-Mediated Ferroptosis Provides a Potential Therapeutic Strategy for Rhabdomyolysis-Induced Acute Kidney Injury,” Acs Chemical Biology 18, no. 6 (2023): 1294-1304.

[291]

S. Zhao, X. Wang, X. Zheng, et al., “Iron deficiency exacerbates cisplatin- or rhabdomyolysis-induced acute kidney injury Through promoting iron-catalyzed oxidative damage,” Free Radic Biol Med 173 (2021): 81-96.

[292]

J. M. O'Brien, N. A. Ali, S. K. Aberegg, and E. Abraham, “Sepsis,” American Journal of Medicine 120, no. 12 (2007): 1012-1022.

[293]

S. Peerapornratana, C. L. Manrique-Caballero, H. Gómez, and J. A. Kellum, “Acute kidney injury From sepsis: Current concepts, epidemiology, pathophysiology, prevention and treatment,” Kidney International 96, no. 5 (2019): 1083-1099.

[294]

J. T. Poston and J. L. Koyner, “Sepsis associated acute kidney injury,” The BMJ 364 (2019): k4891.

[295]

S. Buchanan, E. Combet, P. Stenvinkel, and P. G. Shiels, “Klotho, Aging, and the Failing Kidney,” Front Endocrinol 11 (2020): 560.

[296]

T. Landry, D. Shookster, and H. Huang, “Circulating α-klotho regulates metabolism via distinct central and peripheral mechanisms,” Metabolism 121 (2021): 154819.

[297]

P. Zhou, C. Zhao, Y. Chen, X. Liu, C. Wu, and Z. Hu, “Klotho activation of Nrf2 inhibits the ferroptosis signaling pathway to ameliorate sepsis-associated acute kidney injury,” Transl Androl Urol 12, no. 12 (2023): 1871-1884.

[298]

W. Q. Zhuo, Y. Wen, H. J. Luo, Z. L. Luo, and L. Wang, “Mechanisms of ferroptosis in chronic kidney disease,” Frontiers in Molecular Biosciences 9 (2022): 975582.

[299]

B. J. Nankivell, R. A. Boadle, and D. C. Harris, “Iron accumulation in human chronic renal disease,” Am J Kidney Dis Off J Natl Kidney Found 20, no. 6 (1992): 580-584.

[300]

Y. Liu and J. Wang, “Ferroptosis, a Rising Force Against Renal Fibrosis,” Oxid Med Cell Longev 2022 (2022): 7686956.

[301]

M. Maus, V. López-Polo, L. Mateo, et al., “Iron accumulation drives fibrosis, senescence and the senescence-associated secretory phenotype,” Nat Metab 5, no. 12 (2023): 2111-2130.

[302]

Y. Sato and M. Yanagita, “Immune cells and inflammation in AKI to CKD progression,” American Journal of Physiology. Renal Physiology 315, no. 6 (2018): F1501-F1512.

[303]

K. T. K. Giuliani, A. Grivei, P. Nag, et al., “Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c+ dendritic cells,” Cell death & disease 13, no. 8 (2022): 739.

[304]

P. Bhargava and R. G. Schnellmann, “Mitochondrial energetics in the kidney,” Nature Reviews Nephrology 13, no. 10 (2017): 629-646.

[305]

J. Wang, Y. Wang, Y. Liu, et al., “Ferroptosis, a new target for treatment of renal injury and fibrosis in a 5/6 nephrectomy-induced CKD rat model,” Cell Death Discov 8, no. 1 (2022): 127.

[306]

J. Zhou, Y. Tan, R. Wang, and X. Li, “Role of Ferroptosis in Fibrotic Diseases,” J Inflamm Res 15 (2022): 3689-3708.

[307]

L. Li, H. Fu, and Y. Liu, “The fibrogenic niche in kidney fibrosis: Components and mechanisms,” Nature Reviews Nephrology 18, no. 9 (2022): 545-557.

[308]

Y. Zhang, Y. Mou, J. Zhang, et al., “Therapeutic Implications of Ferroptosis in Renal Fibrosis,” Frontiers in Molecular Biosciences 9 (2022): 890766.

[309]

L. Zhou, X. Xue, Q. Hou, and C. Dai, “Targeting Ferroptosis Attenuates Interstitial Inflammation and Kidney Fibrosis,” Kidney Dis Basel Switz 8, no. 1 (2022): 57-71.

[310]

B. Zhang, X. Chen, F. Ru, et al., “Liproxstatin-1 attenuates unilateral ureteral obstruction-induced renal fibrosis by inhibiting renal tubular epithelial cells ferroptosis,” Cell death & disease 12, no. 9 (2021): 843.

[311]

Y. Dai, Y. Chen, D. Mo, et al., “Inhibition of ACSL4 ameliorates tubular ferroptotic cell death and protects Against fibrotic kidney disease,” Communications Biology 6, no. 1 (2023): 907.

[312]

W. Lai, R. Huang, B. Wang, et al., “Novel aspect of neprilysin in kidney fibrosis via ACSL4-mediated ferroptosis of tubular epithelial cells,” MedComm 4, no. 4 (2023): e330.

[313]

B. Wang, L. N. Yang, L. T. Yang, et al., “Fisetin ameliorates fibrotic kidney disease in mice via inhibiting ACSL4-mediated tubular ferroptosis,” Acta Pharmacologica Sinica (2023), https://doi.org/10.1038/s41401-023-01156-w. Published online September 11.

[314]

M. S. Balzer, T. Doke, Y. W. Yang, et al., “Single-cell analysis highlights differences in druggable pathways underlying adaptive or fibrotic kidney regeneration,” Nature Communications 13 (2022): 4018.

[315]

A. Perrone, A. Giovino, J. Benny, and F. Martinelli, “Advanced Glycation End Products (AGEs): Biochemistry, Signaling, Analytical Methods, and Epigenetic Effects,” Oxid Med Cell Longev 2020 (2020): 3818196.

[316]

Y. Wang, R. Bi, F. Quan, et al., “Ferroptosis involves in renal tubular cell death in diabetic nephropathy,” European Journal of Pharmacology 888 (2020): 173574.

[317]

S. Kim, S. W. Kang, J. Joo, et al., “Characterization of ferroptosis in kidney tubular cell death Under diabetic conditions,” Cell death & disease 12, no. 2 (2021): 160.

[318]

S. Li, L. Zheng, J. Zhang, X. Liu, and Z. Wu, “Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy,” Free Radic Biol Med 162 (2021): 435-449.

[319]

X. Feng, S. Wang, Z. Sun, et al., “Ferroptosis Enhanced Diabetic Renal Tubular Injury via HIF-1α/HO-1 Pathway in db/db Mice,” Front Endocrinol 12 (2021): 626390.

[320]

T. Robert, L. Berthelot, A. Cambier, E. Rondeau, and R. C. Monteiro, “Molecular Insights Into the Pathogenesis of IgA Nephropathy,” Trends in Molecular Medicine 21, no. 12 (2015): 762-775.

[321]

H. Wang, K. Nishiya, H. Ito, T. Hosokawa, K. Hashimoto, and T. Moriki, “Iron deposition in renal biopsy specimens From patients With kidney diseases,” Am J Kidney Dis Off J Natl Kidney Found 38, no. 5 (2001): 1038-1044.

[322]

Z. Y. Tian, Z. Li, L. Chu, et al., “Iron metabolism and chronic inflammation in IgA nephropathy,” Renal Failure 45, no. 1 (2023): 2195012.

[323]

H. Lu, X. Sun, M. Jia, et al., “Rosiglitazone Suppresses Renal Crystal Deposition by Ameliorating Tubular Injury Resulted From Oxidative Stress and Inflammatory Response via Promoting the Nrf2/HO-1 Pathway and Shifting Macrophage Polarization,” Oxid Med Cell Longev 2021 (2021): 5527137.

[324]

Z. Ye, Y. Xia, L. Li, et al., “p53 deacetylation alleviates calcium oxalate deposition-induced renal fibrosis by inhibiting ferroptosis,” Biomed Pharmacother Biomedecine Pharmacother 164 (2023): 114925.

[325]

M. Tsujihata, “Mechanism of calcium oxalate renal stone formation and renal tubular cell injury,” Int J Urol Off J Jpn Urol Assoc 15, no. 2 (2008): 115-120.

[326]

S. R. Khan, “Reactive oxygen species as the molecular modulators of calcium oxalate kidney stone formation: Evidence From clinical and experimental investigations,” Journal of Urology 189, no. 3 (2013): 803-811.

[327]

J. Zhou, L. Meng, Z. He, et al., “Melatonin exerts a protective effect in ameliorating nephrolithiasis via targeting AMPK/PINK1-Parkin mediated mitophagy and inhibiting ferroptosis in vivo and in vitro,” International Immunopharmacology 124 (2023): 110801.

[328]

Z. He, W. Liao, Q. Song, et al., “Role of ferroptosis induced by a high concentration of calcium oxalate in the formation and development of urolithiasis,” International Journal of Molecular Medicine 47, no. 1 (2021): 289-301.

[329]

L. Li, Z. Ye, Y. Xia, et al., “YAP/ACSL4 Pathway-Mediated Ferroptosis Promotes Renal Fibrosis in the Presence of Kidney Stones,” Biomedicines 11, no. 10 (2023): 2692.

[330]

Z. Mao, K. Zhong, X. Liu, and X. Zeng, “Ferroptosis contributes to cyclophosphamide-induced hemorrhagic cystitis,” Chemico-Biological Interactions 384 (2023): 110701.

[331]

X. Wang, T. Ji, Z. Jiang, J. Wang, X. Su, and L. Shan, “Tolterodine ameliorates inflammatory response and ferroptosis Against LPS in human bladder epithelial cells,” Journal of Biochemical and Molecular Toxicology 38, no. 1 (2024): e23517.

[332]

D. Lin, M. Zhang, C. Luo, P. Wei, K. Cui, and Z. Chen, “Targeting Ferroptosis Attenuates Inflammation, Fibrosis, and Mast Cell Activation in Chronic Prostatitis,” Journal of Immunology Research 2022 (2022): 6833867.

[333]

A. Ghoochani, E. C. Hsu, M. Aslan, et al., “Ferroptosis Inducers Are a Novel Therapeutic Approach for Advanced Prostate Cancer,” Cancer Research 81, no. 6 (2021): 1583-1594.

[334]

J. Wang, L. Zeng, N. Wu, et al., “Inhibition of phosphoglycerate dehydrogenase induces ferroptosis and overcomes enzalutamide resistance in castration-resistant prostate cancer cells,” Drug Resist Updat Rev Comment Antimicrob Anticancer Chemother 70 (2023): 100985.

[335]

H. Wang, X. Yu, D. Liu, et al., “VDR Activation Attenuates Renal Tubular Epithelial Cell Ferroptosis by Regulating Nrf2/HO-1 Signaling Pathway in Diabetic Nephropathy,” Adv Sci Weinh Baden-Wurtt Ger 11, no. 10 (2024): e2305563.

[336]

Z. Liu, P. Nan, Y. Gong, L. Tian, Y. Zheng, and Z. Wu, “Endoplasmic reticulum stress-triggered ferroptosis via the XBP1-Hrd1-Nrf2 pathway induces EMT progression in diabetic nephropathy,” Biomed Pharmacother Biomedecine Pharmacother 164 (2023): 114897.

[337]

H. Xiao, X. Du, Z. Tao, et al., “Taurine Inhibits Ferroptosis Mediated by the Crosstalk Between Tumor Cells and Tumor-Associated Macrophages in Prostate Cancer,” Adv Sci Weinh Baden-Wurtt Ger 11, no. 3 (2024): e2303894.

[338]

W. Wang, J. Zhang, Y. Wang, Y. Xu, and S. Zhang, “Identifies microtubule-binding protein CSPP1 as a novel cancer biomarker associated With ferroptosis and tumor microenvironment,” Computational and Structural Biotechnology Journal 20 (2022): 3322-3335.

[339]

D. Liang, Y. Feng, F. Zandkarimi, et al., “Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones,” Cell 186, no. 13 (2023): 2748-2764. e22.

[340]

S. Chang, M. Tang, B. Zhang, D. Xiang, and F. Li, “Ferroptosis in inflammatory arthritis: A promising future,” Frontiers in immunology 13 (2022): 955069.

[341]

B. Lai, C. H. Wu, C. Y. Wu, S. F. Luo, and J. H. Lai, “Ferroptosis and Autoimmune Diseases,” Frontiers in immunology 13 (2022): 916664.

[342]

J. Hu, R. Zhang, Q. Chang, et al., “p53: A Regulator of Ferroptosis Induced by Galectin-1 Derived Peptide 3 in MH7A Cells,” Frontiers in Genetics 13 (2022): 920273.

[343]

G. Nygaard and G. S. Firestein, “Restoring synovial homeostasis in rheumatoid arthritis by targeting fibroblast-Like synoviocytes,” Nat Rev Rheumatol 16, no. 6 (2020): 316-333.

[344]

M. Taghadosi, M. Adib, A. Jamshidi, M. Mahmoudi, and E. Farhadi, “The p53 status in rheumatoid arthritis With focus on fibroblast-Like synoviocytes,” Immunologic Research 69, no. 3 (2021): 225-238.

[345]

L. Jiang, J. H. Hickman, S. J. Wang, and W. Gu, “Dynamic roles of p53-mediated metabolic activities in ROS-induced stress responses,” Cell Cycle Georget Tex 14, no. 18 (2015): 2881-2885.

[346]

Y. Xie, S. Zhu, X. Song, et al., “The Tumor Suppressor p53 Limits Ferroptosis by Blocking DPP4 Activity,” Cell reports 20, no. 7 (2017): 1692-1704.

[347]

Y. Su, B. Zhao, L. Zhou, et al., “Ferroptosis, a novel pharmacological mechanism of anti-cancer drugs,” Cancer Letters 483 (2020): 127-136.

[348]

J. Jhun, J. Moon, J. Ryu, et al., “Liposome/gold hybrid nanoparticle encoded With CoQ10 (LGNP-CoQ10) suppressed rheumatoid arthritis via STAT3/Th17 targeting,” PLoS ONE 15, no. 11 (2020): e0241080.

[349]

S. Ravalli, M. A. Szychlinska, R. M. Leonardi, and G. Musumeci, “Recently highlighted nutraceuticals for preventive management of osteoarthritis,” World J Orthop 9, no. 11 (2018): 255-261.

[350]

D. Jean-Gilles, L. Li, V. G. Vaidyanathan, et al., “Inhibitory effects of polyphenol punicalagin on type-II collagen degradation in vitro and inflammation in vivo,” Chemico-Biological Interactions 205, no. 2 (2013): 90-99.

[351]

H. Luo and R. Zhang, “Icariin enhances cell survival in lipopolysaccharide-induced synoviocytes by suppressing ferroptosis via the Xc-/GPX4 axis,” Exp Ther Med 21, no. 1 (2021): 72.

[352]

R. Zhou, Y. Chen, S. Li, et al., “TRPM7 channel inhibition attenuates rheumatoid arthritis articular chondrocyte ferroptosis by suppression of the PKCα-NOX4 axis,” Redox Biology 55 (2022): 102411.

[353]

G. Ma, Y. Yang, Y. Chen, et al., “Blockade of TRPM7 Alleviates Chondrocyte Apoptosis and Articular Cartilage Damage in the Adjuvant Arthritis Rat Model Through Regulation of the Indian Hedgehog Signaling Pathway,” Frontiers in pharmacology 12 (2021): 655551.

[354]

K. F. Zhai, H. Duan, C. Y. Cui, et al., “Liquiritin From Glycyrrhiza uralensis Attenuating Rheumatoid Arthritis via Reducing Inflammation, Suppressing Angiogenesis, and Inhibiting MAPK Signaling Pathway,” Journal of Agricultural and Food Chemistry 67, no. 10 (2019): 2856-2864.

[355]

H. Liu, H. Xue, Q. Guo, et al., “Ferroptosis meets inflammation: A new frontier in cancer therapy,” Cancer Letters 620 (2025): 217696.

[356]

Y. Dong, M. Zheng, W. Ding, H. Guan, J. Xiao, and F. Li, “Nrf2 activators for the treatment of rare iron overload diseases: From bench to bedside,” Redox Biology 81 (2025): 103551.

[357]

M. T. Nuñez, and P. Chana-Cuevas, “New Perspectives in Iron Chelation Therapy for the Treatment of Neurodegenerative Diseases,” Pharmaceuticals 11, no. 4 (2018): 109.

[358]

Y. Dong, M. Zheng, W. Ding, H. Guan, J. Xiao, and F. Li, “Nrf2 activators for the treatment of rare iron overload diseases: From bench to bedside,” Redox Biology 81 (2025): 103551.

[359]

T. Ramadoss, D. S. Weimer, and H. N. Mayrovitz, “Topical Iron Chelator Therapy: Current Status and Future Prospects,” Cureus 15, no. 10: e47720.

[360]

Y. Wang, Y. Song, L. Xu, et al., “A Membrane-Targeting Aggregation-Induced Emission Probe for Monitoring Lipid Droplet Dynamics in Ischemia/Reperfusion-Induced Cardiomyocyte Ferroptosis,” Adv Sci Weinh Baden-Wurtt Ger 11, no. 26 (2024): e2309907.

[361]

M. Tan, W. Li, H. He, et al., “Targeted mitochondrial fluorescence probe With large stokes shift for detecting viscosity changes in vivo and in ferroptosis process,” Spectrochimica Acta. Part A, Molecular and Biomolecular Spectroscopy 315 (2024): 124246.

[362]

J. Yin, X. Zheng, Y. Zhao, et al., “Investigating the Therapeutic Effects of Ferroptosis on Myocardial Ischemia-Reperfusion Injury Using a Dual-Locking Mitochondrial Targeting Strategy,” Angewandte Chemie (International ed in English) 63, no. 21 (2024): e202402537.

[363]

X. Luo, C. Zhang, C. Yue, Y. Jiang, F. Yang, and Y. Xian, “A near-infrared light-activated nanoprobe for simultaneous detection of hydrogen polysulfide and sulfur dioxide in myocardial ischemia-reperfusion injury,” Chemical Science 14, no. 48 (2023): 14290-14301.

[364]

W. Yang, Y. Wang, C. Fu, et al., “Quantitative visualization of myocardial ischemia-reperfusion-induced cardiac lesions via ferroptosis magnetic particle imaging,” Theranostics 14, no. 3 (2024): 1081-1097.

[365]

H. L. Zhang, B. X. Hu, Z. P. Ye, et al., “TRPML1 triggers ferroptosis defense and is a potential therapeutic target in AKT-hyperactivated cancer,” Science Translational Medicine 16, no. 753 (2024): eadk0330.

[366]

N. Lorito, A. Subbiani, A. Smiriglia, et al., “FADS1/2 control lipid metabolism and ferroptosis susceptibility in triple-negative breast cancer,” EMBO Molecular Medicine 16, no. 7 (2024): 1533-1559.

[367]

D. Chen, C. Zhao, J. Zhang, et al., “Small Molecule MIF Modulation Enhances Ferroptosis by Impairing DNA Repair Mechanisms,” Adv Sci Weinh Baden-Wurtt Ger 11, no. 32 (2024): e2403963.

[368]

H. Meng, Y. Yu, E. Xie, et al., “Hepatic HDAC3 Regulates Systemic Iron Homeostasis and Ferroptosis via the Hippo Signaling Pathway,” Res Wash DC 6 (2023): 0281.

[369]

Z. Jiang, H. Yang, W. Ni, et al., “Attenuation of neuronal ferroptosis in intracerebral hemorrhage by inhibiting HDAC1/2: Microglial heterogenization via the Nrf2/HO1 pathway,” CNS neuroscience & therapeutics 30, no. 3 (2024): e14646.

[370]

C. Zhong, J. Yang, Y. Zhang, et al., “TRPM2 Mediates Hepatic Ischemia-Reperfusion Injury via Ca2+-Induced Mitochondrial Lipid Peroxidation Through Increasing ALOX12 Expression,” Res Wash DC 6 (2023): 0159.

[371]

T. Li, L. Zhao, Y. Li, et al., “PPM1K mediates metabolic disorder of branched-chain amino acid and regulates cerebral ischemia-reperfusion injury by activating ferroptosis in neurons,” Cell death & disease 14, no. 9 (2023): 634.

[372]

P. Xu, L. Kong, C. Tao, et al., “Elabela-APJ axis attenuates cerebral ischemia/reperfusion injury by inhibiting neuronal ferroptosis,” Free Radic Biol Med 196 (2023): 171-186.

[373]

T. Wu, G. Shi, Z. Ji, S. Wang, L. Geng, and Z. Guo, “Circulating small extracellular vesicle-encapsulated SEMA5A-IT1 attenuates myocardial ischemia-reperfusion injury After cardiac surgery With cardiopulmonary bypass,” Cellular & Molecular Biology Letters 27, no. 1 (2022): 95.

[374]

X. Li, X. Peng, X. Zhou, et al., “Small extracellular vesicles delivering lncRNA WAC-AS1 aggravate renal allograft ischemia‒reperfusion injury by inducing ferroptosis propagation,” Cell Death and Differentiation 30, no. 9 (2023): 2167-2186.

[375]

J. Vadolas, G. Z. Ng, K. Kysenius, et al., “SLN124, a GalNac-siRNA targeting transmembrane serine protease 6, in combination With deferiprone therapy reduces ineffective erythropoiesis and hepatic iron-overload in a mouse model of β-thalassaemia,” British Journal of Haematology 194, no. 1 (2021): 200-210.

[376]

Y. Liu, D. Zhao, F. Yang, et al., “Situ Self-Assembled Phytopolyphenol-Coordinated Intelligent Nanotherapeutics for Multipronged Management of Ferroptosis-Driven Alzheimer's Disease,” ACS Nano 18, no. 11 (2024): 7890-7906.

[377]

Y. Ying, Z. Huang, Y. Tu, et al., “A shear-thinning, ROS-scavenging hydrogel combined With dental pulp stem cells promotes spinal cord repair by inhibiting ferroptosis,” Bioact Mater 22 (2023): 274-290.

[378]

C. Li, Y. Wu, Q. Chen, et al., “Pleiotropic Microenvironment Remodeling Micelles for Cerebral Ischemia-Reperfusion Injury Therapy by Inhibiting Neuronal Ferroptosis and Glial Overactivation,” ACS Nano 17, no. 18 (2023): 18164-18177.

[379]

Y. Zhang, Z. Zou, S. Liu, et al., “Edaravone-loaded poly(amino acid) nanogel inhibits ferroptosis for neuroprotection in cerebral ischemia injury,” Asian Journal of Pharmaceutical Sciences 19, no. 2 (2024): 100886.

[380]

Y. Q. Geng, L. N. Qiu, Y. Q. Cheng, et al., “Alleviating Recombinant Tissue Plasminogen Activator-induced Hemorrhagic Transformation in Ischemic Stroke via Targeted Delivery of a Ferroptosis Inhibitor,” Adv Sci Weinh Baden-Wurtt Ger 11, no. 24 (2024): e2309517.

[381]

Y. Wang, Z. Liu, L. Li, et al., “Anti-ferroptosis exosomes engineered for targeting M2 microglia to improve neurological function in ischemic stroke,” J Nanobiotechnology 22, no. 1 (2024): 291.

[382]

C. Ding, B. Wang, J. Zheng, et al., “Neutrophil Membrane-Inspired Nanorobots Act as Antioxidants Ameliorate Ischemia Reperfusion-Induced Acute Kidney Injury,” ACS Appl Mater Interfaces 15, no. 34 (2023): 40292-40303.

[383]

W. Feng, N. Zhu, Y. Xia, et al., “Melanin-Like nanoparticles alleviate ischemia-reperfusion injury in the kidney by scavenging reactive oxygen species and inhibiting ferroptosis,” Iscience 27, no. 4 (2024): 109504.

[384]

X. Zhao, Z. Wang, G. Wu, et al., “Apigenin-7-glucoside-loaded nanoparticle alleviates intestinal ischemia-reperfusion by ATF3/SLC7A11-mediated ferroptosis,” J Control Release Off J Control Release Soc 366 (2024): 182-193.

[385]

H. Yu, Z. Song, J. Yu, et al., “Supramolecular self-assembly of EGCG-selenomethionine nanodrug for treating osteoarthritis,” Bioact Mater 32 (2024): 164-176.

[386]

W. Li, Z. Lv, P. Wang, et al., “Near Infrared Responsive Gold Nanorods Attenuate Osteoarthritis Progression by Targeting TRPV1,” Adv Sci Weinh Baden-Wurtt Ger 11, no. 16 (2024): e2307683.

[387]

Y. Li, Z. Cai, W. Ma, L. Bai, E. Luo, and Y. Lin, “A DNA tetrahedron-based ferroptosis-suppressing nanoparticle: Superior delivery of curcumin and alleviation of diabetic osteoporosis,” Bone Res 12, no. 1 (2024): 14.

[388]

C. Wu and K. Shemisa, “Sorafenib-Associated Heart Failure Complicated by Cardiogenic Shock After Treatment of Advanced Stage Hepatocellular Carcinoma: A Clinical Case Discussion,” Case Rep Cardiol 2017 (2017): 7065759.

[389]

Y. Xie, W. Hou, X. Song, et al., “Ferroptosis: Process and function,” Cell Death and Differentiation 23, no. 3 (2016): 369-379.

[390]

X. Fang, Z. Cai, H. Wang, et al., “Loss of Cardiac Ferritin H Facilitates Cardiomyopathy via Slc7a11-Mediated Ferroptosis,” Circulation Research 127, no. 4 (2020): 486-501.

[391]

X. Fang, H. Wang, D. Han, et al., “Ferroptosis as a target for protection Against cardiomyopathy,” Pnas 116, no. 7 (2019): 2672-2680.

[392]

W. Li, G. Feng, J. M. Gauthier, et al., “Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment After heart transplantation,” Journal of Clinical Investigation 129, no. 6 (2019): 2293-2304.

[393]

Y. Yu, L. Jiang, H. Wang, et al., “Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis,” Blood 136, no. 6 (2020): 726-739.

[394]

H. Wang, P. An, E. Xie, et al., “Characterization of ferroptosis in murine models of hemochromatosis,” Hepatology 66, no. 2 (2017): 449.

[395]

H. S. Kain, E. K. K. Glennon, K. Vijayan, et al., “Liver stage malaria infection is controlled by host regulators of lipid peroxidation,” Cell Death and Differentiation 27, no. 1 (2020): 44-54.

[396]

E. Dai, W. Zhang, D. Cong, R. Kang, J. Wang, and D. Tang, “AIFM2 blocks ferroptosis independent of ubiquinol metabolism,” Biochemical and Biophysical Research Communications 523, no. 4 (2020): 966-971.

[397]

S. Y. Xu, S. S. Yin, L. Wang, H. Zhong, H. Wang, and H. Y. Yu, “Insights Into emerging mechanisms of ferroptosis: New regulators for cancer therapeutics,” Cell Biology and Toxicology 41, no. 1 (2025): 63.

[398]

D. Jiao, Y. Yang, K. Wang, and Y. Wang, “Ferroptosis: A novel pathogenesis and therapeutic strategies for Parkinson disease: A review,” Medicine 104, no. 3 (2025): e41218.

[399]

F. Alves, D. Lane, T. P. M. Nguyen, A. I. Bush, and S. Ayton, “In defence of ferroptosis,” Signal Transduct Target Ther 10, no. 1 (2025): 1-29.

[400]

Y. Wang, L. Yang, X. Zhang, et al., “Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53,” Embo Reports 20, no. 7 (2019): e47563.

[401]

L. Jiang, N. Kon, T. Li, et al., “Ferroptosis as a p53-mediated activity During tumour suppression,” Nature 520, no. 7545 (2015): 57-62.

[402]

B. Chu, N. Kon, D. Chen, et al., “ALOX12 is required for p53-mediated tumour suppression Through a distinct ferroptosis pathway,” Nature Cell Biology 21, no. 5 (2019): 579-591.

[403]

D. E. Jensen, M. Proctor, S. T. Marquis, et al., “BAP1: A novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression,” Oncogene 16, no. 9 (1998): 1097-1112.

[404]

Y. Zhang, J. Shi, X. Liu, et al., “BAP1 links metabolic regulation of ferroptosis to tumour suppression,” Nature Cell Biology 20, no. 10 (2018): 1181-1192.

[405]

N. A. Alam, S. Olpin, A. Rowan, et al., “Missense mutations in fumarate hydratase in multiple cutaneous and uterine leiomyomatosis and renal cell cancer,” J Mol Diagn JMD 7, no. 4 (2005): 437-443.

[406]

G. S. Chuang, A. Martinez-Mir, A. Geyer, et al., “Germline fumarate hydratase mutations and evidence for a founder mutation underlying multiple cutaneous and uterine leiomyomata,” Journal of the American Academy of Dermatology 52 (2005): 410-416. 3 Pt 1.

[407]

M. Gao, J. Yi, J. Zhu, et al., “Role of Mitochondria in Ferroptosis,” Molecular Cell 73, no. 2 (2019): 354-363. e3.

[408]

P. S. Hammerman, M. S. Lawrence, D. Voet, et al., “Comprehensive genomic characterization of squamous cell lung cancers,” Nature 489, no. 7417 (2012): 519-525.

[409]

S. Scalera, M. Mazzotta, C. Cortile, et al., “KEAP1-Mutant NSCLC: The Catastrophic Failure of a Cell-Protecting Hub,” Journal of thoracic oncology 17, no. 6 (2022): 751-757.

[410]

P. Koppula, G. Lei, Y. Zhang, et al., “A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers,” Nature Communications 13, no. 1 (2022): 2206.

[411]

Z. Fan, A. K. Wirth, D. Chen, et al., “Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis,” Oncogenesis 6, no. 8 (2017): e371-e371.

[412]

S. Egolf, J. Zou, A. Anderson, et al., “MLL4 mediates differentiation and tumor suppression Through ferroptosis,” Science Advances 7, no. 50 (2021): eabj9141.

[413]

L. Kuroki and S. R. Guntupalli, “Treatment of epithelial ovarian cancer,” Bmj 371 (2020): m3773.

[414]

Y. Liu, X. Liu, H. Wang, P. Ding, and C. Wang, “Agrimonolide inhibits cancer progression and induces ferroptosis and apoptosis by targeting SCD1 in ovarian cancer cells,” Phytomedicine Int J Phytother Phytopharm 101 (2022): 154102.

[415]

H. Wang, M. G. Klein, H. Zou, et al., “Crystal structure of human stearoyl-coenzyme A desaturase in complex With substrate,” Nature structural & molecular biology 22, no. 7 (2015): 581-585.

[416]

M. T. Bohnsack and K. E. Sloan, “The mitochondrial epitranscriptome: The roles of RNA modifications in mitochondrial translation and human disease,” Cellular and molecular life sciences CMLS 75, no. 2 (2018): 241-260.

[417]

S. Delaunay and M. Frye, “RNA modifications regulating cell fate in cancer,” Nature Cell Biology 21, no. 5 (2019): 552-559.

[418]

Z. Shi, S. Xu, S. Xing, et al., “Mettl17, a regulator of mitochondrial ribosomal RNA modifications, is required for the translation of mitochondrial coding genes,” FASEB J Off Publ Fed Am Soc Exp Biol 33, no. 11 (2019): 13040-13050.

[419]

H. Li, K. Yu, H. Hu, et al., “METTL17 coordinates ferroptosis and tumorigenesis by regulating mitochondrial translation in colorectal cancer,” Redox Biology 71 (2024): 103087.

[420]

R. Wei, Y. Zhao, J. Wang, et al., “Tagitinin C induces ferroptosis Through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells,” Int J Biol Sci 17, no. 11 (2021): 2703-2717.

[421]

X. Miao and N. Zhang, “Role of RBM3 in the regulation of cell proliferation in hepatocellular carcinoma,” Experimental and Molecular Pathology 117 (2020): 104546.

[422]

N. Melling, K. Bachmann, B. Hofmann, et al., “Prevalence and clinical significance of RBM3 immunostaining in non-small cell lung cancers,” Journal of Cancer Research and Clinical Oncology 145, no. 4 (2019): 873-879.

[423]

Z. Wang, W. Shu, R. Zhao, Y. Liu, and H. Wang, “Sodium butyrate induces ferroptosis in endometrial cancer cells via the RBM3/SLC7A11 axis,” Apoptosis Int J Program Cell Death 28, no. 7-8 (2023): 1168-1183.

[424]

A. E. Whiteley, T. T. Price, G. Cantelli, and D. A. Sipkins, “Leukaemia: A model metastatic disease,” Nature Reviews Cancer 21, no. 7 (2021): 461-475.

[425]

J. Du, T. Wang, Y. Li, et al., “DHA inhibits proliferation and induces ferroptosis of leukemia cells Through autophagy dependent degradation of ferritin,” Free Radic Biol Med 131 (2019): 356-369.

[426]

J. Jiang, H. L. Liu, L. Tao, et al., “Let‑7d inhibits colorectal cancer cell proliferation Through the CST1/p65 pathway,” International Journal of Oncology 53, no. 2 (2018): 781-790.

[427]

D. Li, Y. Wang, C. Dong, et al., “CST1 inhibits ferroptosis and promotes gastric cancer metastasis by regulating GPX4 protein stability via OTUB1,” Oncogene 42, no. 2 (2023): 83-98.

[428]

X. Guan, Y. Wang, W. Yu, et al., “Blocking Ubiquitin-Specific Protease 7 Induces Ferroptosis in Gastric Cancer via Targeting Stearoyl-CoA Desaturase,” Adv Sci Weinh Baden-Wurtt Ger 11, no. 18 (2024): e2307899.

[429]

J. Gu, R. Sun, D. Tang, F. Liu, X. Chang, and Q. Wang, “Astragalus mongholicus Bunge-Curcuma aromatica Salisb. suppresses growth and metastasis of colorectal cancer cells by inhibiting M2 macrophage polarization via a Sp1/ZFAS1/miR-153-3p/CCR5 regulatory axis,” Cell Biology and Toxicology 38, no. 4 (2022): 679-697.

[430]

C. Yi, S. Wu, Q. Duan, et al., “Ferroptosis-dependent breast cancer cell-derived exosomes inhibit migration and invasion of breast cancer cells by suppressing M2 macrophage polarization,” PeerJ 11 (2023): e15060.

[431]

C. Q. Wang, C. H. Tang, Y. Wang, et al., “Upregulated WTAP expression appears to both promote breast cancer growth and inhibit lymph node metastasis,” Scientific Reports 12, no. 1 (2022): 1023.

[432]

J. Liu, X. Song, F. Kuang, et al., “NUPR1 is a critical repressor of ferroptosis,” Nature Communications 12, no. 1 (2021): 647.

[433]

M. Tan, Y. He, J. Yi, et al., “WTAP Mediates NUPR1 Regulation of LCN2 Through m6A Modification to Influence Ferroptosis, Thereby Promoting Breast Cancer Proliferation, Migration and Invasion,” Biochemical Genetics 62, no. 2 (2024): 876-891.

[434]

M. Jiang, Y. Jike, K. Liu, et al., “Exosome-mediated miR-144-3p promotes ferroptosis to inhibit osteosarcoma proliferation, migration, and invasion Through regulating ZEB1,” Molecular cancer 22, no. 1 (2023): 113.

[435]

M. J. Rane, Y. Zhao, and L. Cai, “Krϋppel-Like factors (KLFs) in renal physiology and disease,” EBioMedicine 40 (2019): 743-750.

[436]

Y. Lu, H. Qin, B. Jiang, et al., “KLF2 inhibits cancer cell migration and invasion by regulating ferroptosis Through GPX4 in clear cell renal cell carcinoma,” Cancer Letters 522 (2021): 1-13.

[437]

R. L. Siegel, K. D. Miller, H. E. Fuchs, and A. Jemal, “Cancer Statistics, 2021,” CA: A Cancer Journal for Clinicians 71, no. 1 (2021): 7-33.

[438]

X. Zhang, X. Zheng, X. Ying, W. Xie, Y. Yin, and X. Wang, “CEBPG suppresses ferroptosis Through transcriptional control of SLC7A11 in ovarian cancer,” Journal of translational medicine 21, no. 1 (2023): 334.

[439]

D. P. Modest, I. Ricard, and V. Heinemann, “Outcome according to KRAS-, NRAS- and BRAF-mutation as well as KRAS mutation variants: Pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO colorectal cancer study group,” Ann Oncol Off J Eur Soc Med Oncol 27, no. 9 (2016): 1746-1753.

[440]

Q. Miao, W. Q. Deng, W. Y. Lyu, et al., “Erianin inhibits the growth and metastasis Through autophagy-dependent ferroptosis in KRASG13D colorectal cancer,” Free Radic Biol Med 204 (2023): 301-312.

[441]

J. M. Baughman, F. Perocchi, H. S. Girgis, et al., “Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter,” Nature 476, no. 7360 (2011): 341-345.

[442]

X. Wang, Y. Li, Z. Li, et al., “Mitochondrial Calcium Uniporter Drives Metastasis and Confers a Targetable Cystine Dependency in Pancreatic Cancer,” Cancer Research 82, no. 12 (2022): 2254-2268.

[443]

T. W. Lager, C. Conner, C. R. Keating, J. N. Warshaw, and A. D. Panopoulos, “Cell surface GRP78 and Dermcidin cooperate to regulate breast cancer cell migration Through Wnt signaling,” Oncogene 40, no. 23 (2021): 4050-4059.

[444]

S. Samanta, S. Yang, B. Debnath, et al., “The Hydroxyquinoline Analogue YUM70 Inhibits GRP78 to Induce ER Stress-Mediated Apoptosis in Pancreatic Cancer,” Cancer Research 81, no. 7 (2021): 1883-1895.

[445]

Q. Wang, S. Ke, Z. Liu, H. Shao, M. He, and J. Guo, “HSPA5 Promotes the Proliferation, Metastasis and Regulates Ferroptosis of Bladder Cancer,” International Journal of Molecular Sciences 24, no. 6 (2023): 5144.

[446]

J. Ketteler and D. Klein, “Caveolin-1, cancer and therapy resistance,” International Journal of Cancer 143, no. 9 (2018): 2092-2104.

[447]

T. Lu, Z. Zhang, X. Pan, et al., “Caveolin-1 promotes cancer progression via inhibiting ferroptosis in head and neck squamous cell carcinoma,” J Oral Pathol Med Off Publ Int Assoc Oral Pathol Am Acad Oral Pathol 51, no. 1 (2022): 52-62.

[448]

J. Paris, C. Wilhelm, C. Lebbé, et al., “PROM2 overexpression induces metastatic potential Through epithelial-to-mesenchymal transition and ferroptosis resistance in human cancers,” Clinical and translational medicine 14, no. 3 (2024): e1632.

[449]

S. N. Dowland, R. J. Madawala, C. E. Poon, L. A. Lindsay, and C. R. Murphy, “Prominin-2 Prevents the Formation of Caveolae in Normal and Ovarian Hyperstimulated Pregnancy,” Reprod Sci Thousand Oaks Calif 25, no. 8 (2018): 1231-1242.

[450]

V. J. N. Bykov, S. E. Eriksson, J. Bianchi, and K. G. Wiman, “Targeting mutant p53 for efficient cancer therapy,” Nature Reviews Cancer 18, no. 2 (2018): 89-102.

[451]

K. T. Bieging, S. S. Mello, and L. D. Attardi, “Unravelling mechanisms of p53-mediated tumour suppression,” Nature Reviews Cancer 14, no. 5 (2014): 359-370.

[452]

G. Lei, Y. Zhang, T. Hong, et al., “Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity,” Oncogene 40, no. 20 (2021): 3533-3547.

[453]

Y. Wang, L. Yang, X. Zhang, et al., “Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53,” Embo Reports 20, no. 7 (2019): e47563.

[454]

Y. Ou, S. J. Wang, D. Li, B. Chu, and W. Gu, “Activation of SAT1 engages polyamine metabolism With p53-mediated ferroptotic responses,” PNAS 113, no. 44 (2016): E6806-E6812.

[455]

M. Jennis, C. P. Kung, S. Basu, et al., “An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model,” Genes & development 30, no. 8 (2016): 918-930.

[456]

L. Jiang, N. Kon, T. Li, et al., “Ferroptosis as a p53-mediated activity During tumour suppression,” Nature 520, no. 7545 (2015): 57-62.

[457]

A. Tarangelo, L. Magtanong, K. T. Bieging-Rolett, et al., “p53 Suppresses Metabolic Stress-Induced Ferroptosis in Cancer Cells,” Cell reports 22, no. 3 (2018): 569-575.

[458]

M. Carbone, H. Yang, H. I. Pass, T. Krausz, J. R. Testa, and G. Gaudino, “BAP1 and cancer,” Nature Reviews Cancer 13, no. 3 (2013): 153-159.

[459]

K. H. Ventii, N. S. Devi, K. L. Friedrich, et al., “BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization,” Cancer Research 68, no. 17 (2008): 6953-6962.

[460]

M. Carbone, J. W. Harbour, J. Brugarolas, et al., “Biological Mechanisms and Clinical Significance of BAP1 Mutations in Human Cancer,” Cancer discovery 10, no. 8 (2020): 1103-1120.

[461]

Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012; 489(7417): 519-525.

[462]

Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014; 511(7511): 543-550.

[463]

L. Baird and M. Yamamoto, “The Molecular Mechanisms Regulating the KEAP1-NRF2 Pathway,” Molecular and Cellular Biology 40, no. 13 (2020): e00099-20.

[464]

T. W. Kensler, N. Wakabayashi, and S. Biswal, “Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway,” Annual Review of Pharmacology and Toxicology 47 (2007): 89-116.

[465]

D. Chen, O. Tavana, B. Chu, et al., “NRF2 Is a Major Target of ARF in p53-Independent Tumor Suppression,” Molecular Cell 68, no. 1 (2017): 224-232. e4.

[466]

L. Jiang, N. Kon, T. Li, et al., “Ferroptosis as a p53-mediated activity During tumour suppression,” Nature 520, no. 7545 (2015): 57-62.

[467]

E. Kastenhuber and S. Lowe, “Putting p53 in context,” Cell 170, no. 6 (2017): 1062-1078.

[468]

T. Li, N. Kon, L. Jiang, et al., “Tumor suppression in the absence of p53-mediated cell cycle arrest, apoptosis, and senescence,” Cell 149, no. 6 (2012): 1269-1283.

[469]

Y. Liu and W. Gu, “p53 in ferroptosis regulation: The new weapon for the old guardian,” Cell Death and Differentiation 29, no. 5 (2022): 895-910.

[470]

Y. Ou, S. J. Wang, L. Jiang, B. Zheng, and W. Gu, “p53 Protein-mediated Regulation of Phosphoglycerate Dehydrogenase (PHGDH) Is Crucial for the Apoptotic Response upon Serine Starvation,” Journal of Biological Chemistry 290, no. 1 (2015): 457-466.

[471]

M. Wang, C. Mao, L. Ouyang, et al., “Long noncoding RNA LINC00336 inhibits ferroptosis in lung cancer by functioning as a competing endogenous RNA,” Cell Death and Differentiation 26, no. 11 (2019): 2329-2343.

[472]

S. J. Wang, D. Li, Y. Ou, et al., “Acetylation Is Crucial for p53-mediated Ferroptosis and Tumor Suppression,” Cell reports 17, no. 2 (2016): 366-373.

[473]

T. Thomas and T. J. Thomas, “Polyamine metabolism and cancer,” Journal of Cellular and Molecular Medicine 7, no. 2 (2003): 113-126.

[474]

Y. Ou, S. J. Wang, D. Li, B. Chu, and W. Gu, “Activation of SAT1 engages polyamine metabolism With p53-mediated ferroptotic responses,” PNAS 113, no. 44 (2016): E6806-E6812.

[475]

B. Chu, N. Kon, D. Chen, et al., “ALOX12 is required for p53-mediated tumor suppression Through a distinct ferroptosis pathway,” Nature Cell Biology 21, no. 5 (2019): 579-591.

[476]

X. Yang, J. Liu, C. Wang, et al., “miR-18a promotes glioblastoma development by Down-regulating ALOXE3-mediated ferroptotic and anti-migration activities,” Oncogenesis 10, no. 2 (2021): 15.

[477]

W. Hu, C. Zhang, R. Wu, Y. Sun, A. Levine, and Z. Feng, “Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function,” PNAS 107, no. 16 (2010): 7455-7460.

[478]

M. Kobayashi and M. Yamamoto, “Nrf2-Keap1 regulation of cellular defense mechanisms Against electrophiles and reactive oxygen species,” Advances in Enzyme Regulation 46 (2006): 113-140.

[479]

D. D. Zhang, S. C. Lo, J. V. Cross, D. J. Templeton, and M. Hannink, “Keap1 is a redox-regulated substrate adaptor protein for a Cul3-dependent ubiquitin ligase complex,” Molecular and Cellular Biology 24, no. 24 (2004): 10941-10953.

[480]

J. W. Kaspar, S. K. Niture, and A. K. Jaiswal, “Nrf2:INrf2 (Keap1) signaling in oxidative stress,” Free Radic Biol Med 47, no. 9 (2009): 1304-1309.

[481]

M. K. Kwak, J. M. Cho, B. Huang, S. Shin, and T. W. Kensler, “Role of increased expression of the proteasome in the protective effects of sulforaphane Against hydrogen peroxide-mediated cytotoxicity in murine neuroblastoma cells,” Free Radic Biol Med 43, no. 5 (2007): 809-817.

[482]

C. Tonelli, I. I. C. Chio, and D. A. Tuveson, “Transcriptional Regulation by Nrf2,” Antioxid Redox Signaling 29, no. 17 (2018): 1727-1745.

[483]

M. Nezu, N. Suzuki, and M. Yamamoto, “Targeting the KEAP1-NRF2 System to Prevent Kidney Disease Progression,” American Journal of Nephrology 45, no. 6 (2017): 473-483.

[484]

P. Koppula, G. Lei, Y. Zhang, et al., “A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers,” Nature Communications 13, no. 1 (2022): 2206.

[485]

D. A. Guertin and D. M. Sabatini, “Defining the role of mTOR in cancer,” Cancer Cell 12, no. 1 (2007): 9-22.

[486]

T. Porstmann, C. R. Santos, B. Griffiths, et al., “SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth,” Cell metabolism 8, no. 3 (2008): 224-236.

[487]

J. Yi, J. Zhu, J. Wu, C. B. Thompson, and X. Jiang, “Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis,” PNAS 117, no. 49 (2020): 31189-31197.

[488]

Y. Zhang, R. V. Swanda, L. Nie, et al., “mTORC1 couples cyst(e)ine availability With GPX4 protein synthesis and ferroptosis regulation,” Nature Communications 12, no. 1 (2021): 1589.

[489]

R. Shen, A. Martin, A. Ni, et al., “Harnessing Clinical Sequencing Data for Survival Stratification of Patients With Metastatic Lung Adenocarcinomas,” JCO Precis Oncol 3 (2019): PO.18.00307.

[490]

C. A. Wohlhieter, A. L. Richards, F. Uddin, et al., “Concurrent Mutations in STK11 and KEAP1 Promote Ferroptosis Protection and SCD1 Dependence in Lung Cancer,” Cell reports 33, no. 9 (2020): 108444.

[491]

B. Keith, R. S. Johnson, and M. C. Simon, “HIF1α and HIF2α: Sibling rivalry in hypoxic tumour growth and progression,” Nature Reviews Cancer 12, no. 1 (2012): 9-22.

[492]

V. Infantino, A. Santarsiero, P. Convertini, S. Todisco, and V. Iacobazzi, “Cancer Cell Metabolism in Hypoxia: Role of HIF-1 as Key Regulator and Therapeutic Target,” International Journal of Molecular Sciences 22, no. 11 (2021): 5703.

[493]

S. Sun, C. Guo, T. Gao, et al., “Hypoxia Enhances Glioma Resistance to Sulfasalazine-Induced Ferroptosis by Upregulating SLC7A11 via PI3K/AKT/HIF-1 α Axis,” Oxid Med Cell Longev 2022 (2022): 7862430.

[494]

Z. Lin, J. Song, Y. Gao, et al., “Hypoxia-induced HIF-1α/lncRNA-PMAN inhibits ferroptosis by promoting the cytoplasmic translocation of ELAVL1 in peritoneal dissemination From gastric cancer,” Redox Biology 52 (2022): 102312.

[495]

H. Choudhry and A. L. Harris, “Advances in Hypoxia-Inducible Factor Biology,” Cell metabolism 27, no. 2 (2018): 281-298.

[496]

S. Cai, Z. Ding, X. Liu, and J. Zeng, “Trabectedin induces ferroptosis via regulation of HIF-1α/IRP1/TFR1 and Keap1/Nrf2/GPX4 axis in non-small cell lung cancer cells,” Chemico-Biological Interactions 369 (2023): 110262.

[497]

Y. S. Green, M. C. Ferreira dos Santos, D. G. Fuja, et al., “ISCA2 inhibition decreases HIF and induces ferroptosis in clear cell renal carcinoma,” Oncogene 41, no. 42 (2022): 4709-4723.

[498]

B. Zhao, Q. Y. Lei, and K. L. Guan, “The Hippo-YAP pathway: New connections Between regulation of organ size and cancer,” Current Opinion in Cell Biology 20, no. 6 (2008): 638-646.

[499]

F. van Roy and G. Berx, “The cell-cell adhesion molecule E-cadherin,” Cellular and molecular life sciences CMLS 65, no. 23 (2008): 3756-3788.

[500]

N. G. Kim, E. Koh, X. Chen, and B. M. Gumbiner, “E-cadherin mediates contact inhibition of proliferation Through Hippo signaling-pathway components,” PNAS 108, no. 29 (2011): 11930-11935.

[501]

J. Wu, A. M. Minikes, M. Gao, et al., “Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling,” Nature 572, no. 7769 (2019): 402-406.

[502]

W. Gan, X. Dai, X. Dai, et al., “LATS suppresses mTORC1 activity to directly coordinate Hippo and mTORC1 pathways in growth control,” Nature Cell Biology 22, no. 2 (2020): 246-256.

[503]

J. M. Drijvers, J. E. Gillis, T. Muijlwijk, et al., “Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells,” Cancer immunology research 9, no. 2 (2021): 184-199.

[504]

M. Matsushita, S. Freigang, C. Schneider, M. Conrad, G. W. Bornkamm, and M. Kopf, “T cell lipid peroxidation induces ferroptosis and prevents immunity to infection,” Journal of Experimental Medicine 212, no. 4 (2015): 555-568.

[505]

J. M. Drijvers, J. E. Gillis, T. Muijlwijk, et al., “Pharmacologic screening identifies metabolic vulnerabilities of CD8+ T cells,” Cancer immunology research 9, no. 2 (2021): 184-199.

[506]

S. D. Jeong, B. K. Jung, D. Lee, et al., “Enhanced Immunogenic Cell Death by Apoptosis/Ferroptosis Hybrid Pathway Potentiates PD-L1 Blockade Cancer Immunotherapy,” ACS Biomater Sci Eng 8, no. 12 (2022): 5188-5198.

[507]

C. Xu, S. Sun, T. Johnson, et al., “The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity,” Cell reports 35, no. 11 (2021): 109235.

[508]

A. A. Kapralov, Q. Yang, H. H. Dar, et al., “Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death,” Nature Chemical Biology 16, no. 3 (2020): 278-290.

[509]

X. Luo, H. B. Gong, H. Y. Gao, et al., “Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting With TLR2,” Cell Death and Differentiation 28, no. 6 (2021): 1971-1989.

[510]

Z. Shi, J. Zheng, W. Tang, et al., “Multifunctional Nanomaterials for Ferroptotic Cancer Therapy,” Front Chem 10 (2022): 868630.

[511]

Z. Gu, T. Liu, C. Liu, et al., “Ferroptosis-Strengthened Metabolic and Inflammatory Regulation of Tumor-Associated Macrophages Provokes Potent Tumoricidal Activities,” Nano Letters 21, no. 15 (2021): 6471-6479.

[512]

K. Chen, H. Li, A. Zhou, et al., “Cell Membrane Camouflaged Metal Oxide-Black Phosphorus Biomimetic Nanocomplex Enhances Photo-chemo-dynamic Ferroptosis,” ACS Appl Mater Interfaces 14, no. 23 (2022): 26557-26570.

[513]

R. Kim, A. Hashimoto, N. Markosyan, et al., “Ferroptosis of tumour neutrophils causes immune suppression in cancer,” Nature 612, no. 7939 (2022): 338-346.

[514]

F. Veglia, V. A. Tyurin, M. Blasi, et al., “Fatty acid transport protein 2 reprograms neutrophils in cancer,” Nature 569, no. 7754 (2019): 73-78.

[515]

A. Ugolini, V. A. Tyurin, Y. Y. Tyurina, et al., “Polymorphonuclear myeloid-derived suppressor cells limit antigen cross-presentation by dendritic cells in cancer,” JCI Insight 5, no. 15 (2020): e138581. 138581.

[516]

M. K. Srivastava, P. Sinha, V. K. Clements, P. Rodriguez, and S. Ostrand-Rosenberg, “Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine,” Cancer Research 70, no. 1 (2010): 68-77.

[517]

J. P. Friedmann Angeli, D. V. Krysko, and M. Conrad, “Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion,” Nature Reviews Cancer 19, no. 7 (2019): 405-414.

[518]

X. Luo, H. B. Gong, H. Y. Gao, et al., “Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting With TLR2,” Cell Death and Differentiation 28, no. 6 (2021): 1971-1989.

[519]

J. P. Friedmann Angeli, M. Schneider, B. Proneth, et al., “Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice,” Nature Cell Biology 16, no. 12 (2014): 1180-1191.

[520]

D. Wang and R. N. Dubois, “Eicosanoids and cancer,” Nature Reviews Cancer 10, no. 3 (2010): 181-193.

[521]

W. Wang, M. Green, J. E. Choi, et al., “CD8+ T cells regulate tumour ferroptosis During cancer immunotherapy,” Nature 569, no. 7755 (2019): 270-274.

[522]

D. H. Kim, W. D. Kim, S. K. Kim, D. H. Moon, and S. J. Lee, “TGF-β1-mediated repression of SLC7A11 drives vulnerability to GPX4 inhibition in hepatocellular carcinoma cells,” Cell death & disease 11, no. 5 (2020): 406.

[523]

J. Guo, B. Xu, Q. Han, et al., “Ferroptosis: A Novel Anti-tumor Action for Cisplatin,” Cancer research and treatment: official journal of Korean Cancer Association 50, no. 2 (2018): 445-460.

[524]

S. Ma, E. S. Henson, Y. Chen, and S. B. Gibson, “Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells,” Cell death & disease 7, no. 7 (2016): e2307.

[525]

Y. Yamaguchi, T. Kasukabe, and S. Kumakura, “Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly Through the induction of ferroptosis,” International Journal of Oncology 52, no. 3 (2018): 1011-1022.

[526]

C. Liang, X. Zhang, M. Yang, and X. Dong, “Recent Progress in Ferroptosis Inducers for Cancer Therapy,” Adv Mater Deerfield Beach Fla 31, no. 51 (2019): e1904197.

[527]

H. Yamaguchi, J. L. Hsu, C. T. Chen, et al., “Caspase-independent cell death is involved in the negative effect of EGF receptor inhibitors on cisplatin in non-small cell lung cancer cells,” Clin Cancer Res Off J Am Assoc Cancer Res 19, no. 4 (2013): 845-854.

[528]

L. Chen, X. Li, L. Liu, B. Yu, Y. Xue, and Y. Liu, “Erastin sensitizes glioblastoma cells to temozolomide by restraining xCT and cystathionine-γ-lyase function,” Oncology Reports 33, no. 3 (2015): 1465-1474.

[529]

X. Tong, R. Tang, M. Xiao, et al., “Targeting cell death pathways for cancer therapy: Recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research,” J Hematol OncolJ Hematol Oncol 15 (2022): 174.

[530]

S. J. Dixon, D. N. Patel, M. Welsch, et al., “Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis,” Elife 3 (2014): e02523.

[531]

W. S. Yang, R. SriRamaratnam, M. E. Welsch, et al., “Regulation of ferroptotic cancer cell death by GPX4,” Cell 156, no. 1-2 (2014): 317-331.

[532]

Y. Zhang, H. Tan, J. D. Daniels, et al., “Imidazole Ketone Erastin Induces Ferroptosis and Slows Tumor Growth in a Mouse Lymphoma Model,” Cell Chem Biol 26, no. 5 (2019): 623-633. e9.

[533]

G. K. Balendiran, R. Dabur, and D. Fraser, “The role of glutathione in cancer,” Cell Biochemistry and Function 22, no. 6 (2004): 343-352.

[534]

C. Liang, X. Zhang, M. Yang, and X. Dong, “Recent Progress in Ferroptosis Inducers for Cancer Therapy,” Adv Mater Deerfield Beach Fla 31, no. 51 (2019): e1904197.

[535]

A. Recasens and L. Munoz, “Targeting Cancer Cell Dormancy,” Trends in Pharmacological Sciences 40, no. 2 (2019): 128-141.

[536]

H. L. Zhang, B. X. Hu, Z. P. Ye, et al., “TRPML1 triggers ferroptosis defense and is a potential therapeutic target in AKT-hyperactivated cancer,” Science Translational Medicine 16, no. 753 (2024): eadk0330.

[537]

Z. Li, Z. M. Xu, W. P. Chen, et al., “Tumor-repopulating cells evade ferroptosis via PCK2-dependent phospholipid remodeling,” Nature Chemical Biology 20, no. 10 (2024): 1341-1352.

[538]

L. Ma, C. Chen, C. Zhao, et al., “Targeting carnitine palmitoyl transferase 1A (CPT1A) induces ferroptosis and synergizes With immunotherapy in lung cancer,” Signal Transduct Target Ther 9, no. 1 (2024): 64.

[539]

W. Wang, K. Lu, X. Jiang, et al., “Ferroptosis inducers enhanced cuproptosis induced by copper ionophores in primary liver cancer,” J Exp Clin Cancer Res CR 42, no. 1 (2023): 142.

[540]

Y. Li, H. Yan, X. Xu, H. Liu, C. Wu, and L. Zhao, “Erastin/sorafenib induces cisplatin-resistant non-small cell lung cancer cell ferroptosis Through inhibition of the Nrf2/xCT pathway,” Oncology letters 19, no. 1 (2020): 323-333.

[541]

Q. Cheng, L. Bao, M. Li, K. Chang, and X. Yi, “Erastin synergizes With cisplatin via ferroptosis to inhibit ovarian cancer growth in vitro and in vivo,” Journal of Obstetrics and Gynaecology Research 47, no. 7 (2021): 2481-2491.

[542]

Y. Shibata, H. Yasui, K. Higashikawa, N. Miyamoto, and Y. Kuge, “Erastin, a ferroptosis-inducing agent, sensitized cancer cells to X-ray irradiation via glutathione starvation in vitro and in vivo,” PLoS ONE 14, no. 12 (2019): e0225931.

[543]

B. Tang, J. Zhu, Y. Wang, et al., “Targeted xCT-mediated Ferroptosis and Protumoral Polarization of Macrophages Is Effective Against HCC and Enhances the Efficacy of the Anti-PD-1/L1 Response,” Adv Sci Weinh Baden-Wurtt Ger 10, no. 2 (2023): e2203973.

[544]

Z. Yang, R. Huang, Y. Wang, et al., “SIRT6 drives sensitivity to ferroptosis in anaplastic thyroid cancer Through NCOA4-dependent autophagy,” Am J Cancer Res 13, no. 2 (2023): 464-474.

[545]

W. S. Yang, R. SriRamaratnam, M. E. Welsch, et al., “Regulation of ferroptotic cancer cell death by GPX4,” Cell 156, no. 1-2 (2014): 317-331.

[546]

L. F. Ye, K. R. Chaudhary, F. Zandkarimi, et al., “Radiation-Induced Lipid Peroxidation Triggers Ferroptosis and Synergizes With Ferroptosis Inducers,” Acs Chemical Biology 15, no. 2 (2020): 469-484.

[547]

L. Ferrada, M. J. Barahona, M. Vera, B. R. Stockwell, and F. Nualart, “Dehydroascorbic acid sensitizes cancer cells to system xc- inhibition-induced ferroptosis by promoting lipid droplet peroxidation,” Cell death & disease 14, no. 9 (2023): 637.

[548]

G. Shan, G. Bi, G. Zhao, et al., “Inhibition of PKA/CREB1 pathway confers sensitivity to ferroptosis in non-small cell lung cancer,” Respiratory Research 24, no. 1 (2023): 277.

[549]

S. L. Cramer, A. Saha, J. Liu, et al., “Systemic depletion of L-cyst(e)ine With cyst(e)inase increases reactive oxygen species and suppresses tumor growth,” Nature Medicine 23, no. 1 (2017): 120-127.

[550]

S. W. Alvarez, V. O. Sviderskiy, E. M. Terzi, et al., “NFS1 undergoes positive selection in lung tumours and protects cells From ferroptosis,” Nature 551, no. 7682 (2017): 639-643.

[551]

W. Wang, M. Green, J. E. Choi, et al., “CD8+ T cells regulate tumour ferroptosis During cancer immunotherapy,” Nature 569, no. 7755 (2019): 270-274.

[552]

Y. Bai, L. Meng, L. Han, et al., “Lipid storage and lipophagy regulates ferroptosis,” Biochemical and Biophysical Research Communications 508, no. 4 (2019): 997-1003.

[553]

F. Yang, Y. Xiao, J. H. Ding, et al., “Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy,” Cell metabolism 35, no. 1 (2023): 84-100. e8.

[554]

S. Y. Park, K. J. Jeong, A. Poire, et al., “Irreversible HER2 inhibitors overcome resistance to the RSL3 ferroptosis inducer in non-HER2 amplified luminal breast cancer,” Cell death & disease 14, no. 8 (2023): 532.

[555]

X. Zhang, S. Sui, L. Wang, et al., “Inhibition of tumor propellant glutathione peroxidase 4 induces ferroptosis in cancer cells and enhances anticancer effect of cisplatin,” Journal of Cellular Physiology 235, no. 4 (2020): 3425-3437.

[556]

J. H. You, J. Lee, and J. L. Roh, “Mitochondrial pyruvate carrier 1 regulates ferroptosis in drug-tolerant persister head and neck cancer cells via epithelial-mesenchymal transition,” Cancer Letters 507 (2021): 40-54.

[557]

L. Zhou, J. Chen, R. Li, et al., “Metal-Polyphenol-Network Coated Prussian Blue Nanoparticles for Synergistic Ferroptosis and Apoptosis via Triggered GPX4 Inhibition and Concurrent In Situ Bleomycin Toxification,” Small Weinh Bergstr Ger 17, no. 47 (2021): e2103919.

[558]

Q. Hu, W. Zhu, J. Du, et al., “A GPX4-targeted photosensitizer to reverse hypoxia-induced inhibition of ferroptosis for non-small cell lung cancer therapy,” Chemical Science 14, no. 34 (2023): 9095-9100.

[559]

X. Zhang, Y. Guo, H. Li, and L. Han, “FIN56, a novel ferroptosis inducer, triggers lysosomal membrane permeabilization in a TFEB-dependent manner in glioblastoma,” Journal of Cancer 12, no. 22 (2021): 6610-6619.

[560]

G. Lei, Y. Zhang, P. Koppula, et al., “The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression,” Cell Research 30, no. 2 (2020): 146-162.

[561]

M. M. Gaschler, A. A. Andia, H. Liu, et al., “FINO2 initiates ferroptosis Through GPX4 inactivation and iron oxidation,” Nature Chemical Biology 14, no. 5 (2018): 507-515.

[562]

Z. He, H. Zhou, Y. Zhang, et al., “Oxygen-boosted biomimetic nanoplatform for synergetic phototherapy/ferroptosis activation and reversal of immune-suppressed tumor microenvironment,” Biomaterials 290 (2022): 121832.

[563]

T. Nakamura, C. Hipp, A. Santos Dias Mourão, et al., “Phase separation of FSP1 promotes ferroptosis,” Nature 619, no. 7969 (2023): 371-377.

[564]

Y. Zhou, K. Chen, W. K. Lin, et al., “Photo-Enhanced Synergistic Induction of Ferroptosis for Anti-Cancer Immunotherapy,” Adv Healthc Mater 12, no. 27 (2023): e2300994.

[565]

J. W. S. Cheu, D. Lee, Q. Li, et al., “Ferroptosis Suppressor Protein 1 Inhibition Promotes Tumor Ferroptosis and Anti-tumor Immune Responses in Liver Cancer,” Cell Mol Gastroenterol Hepatol 16, no. 1 (2023): 133-159.

[566]

M. Mu, Y. Wang, S. Zhao, et al., “Engineering a pH/Glutathione-Responsive Tea Polyphenol Nanodevice as an Apoptosis/Ferroptosis-Inducing Agent,” ACS Appl Bio Mater 3, no. 7 (2020): 4128-4138.

[567]

X. Wang, M. Wu, X. Zhang, et al., “Hypoxia-responsive nanoreactors based on self-enhanced photodynamic sensitization and triggered ferroptosis for cancer synergistic therapy,” J Nanobiotechnology 19, no. 1 (2021): 204.

[568]

C. Zhang, Z. Liu, Y. Zhang, L. Ma, E. Song, and Y. Song, “Iron free‘’ zinc oxide nanoparticles With ion-leaking properties disrupt intracellular ROS and iron homeostasis to induce ferroptosis,” Cell death & disease 11, no. 3 (2020): 183.

[569]

Z. Shen, J. Song, B. C. Yung, Z. Zhou, A. Wu, and X. Chen, “Emerging Strategies of Cancer Therapy Based on Ferroptosis,” Adv Mater Deerfield Beach Fla 30, no. 12 (2018): e1704007.

[570]

M. Gao, J. Deng, F. Liu, et al., “Triggered ferroptotic polymer micelles for reversing multidrug resistance to chemotherapy,” Biomaterials 223 (2019): 119486.

[571]

W. Wang, M. Green, J. E. Choi, et al., “CD8+ T cells regulate tumour ferroptosis During cancer immunotherapy,” Nature 569, no. 7755 (2019): 270-274.

[572]

P. Liao, W. Wang, W. Wang, et al., “CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4,” Cancer Cell 40, no. 4 (2022): 365-378. e6.

[573]

X. Lang, M. D. Green, W. Wang, et al., “Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11,” Cancer discovery 9, no. 12 (2019): 1673-1685.

[574]

Y. Xue, F. Lu, Z. Chang, et al., “Intermittent dietary methionine deprivation facilitates tumoral ferroptosis and synergizes With checkpoint blockade,” Nature Communications 14, no. 1 (2023): 4758.

[575]

H. L. Zhang, B. X. Hu, Z. L. Li, et al., “PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis,” Nature Cell Biology 24, no. 1 (2022): 88-98.

[576]

E. Dai, L. Han, J. Liu, et al., “Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein,” Autophagy 16, no. 11 (2020): 2069-2083.

[577]

Q. Wen, J. Liu, R. Kang, B. Zhou, and D. Tang, “The release and activity of HMGB1 in ferroptosis,” Biochemical and Biophysical Research Communications 510, no. 2 (2019): 278-283.

[578]

E. Dai, L. Han, J. Liu, et al., “Ferroptotic damage promotes pancreatic tumorigenesis Through a TMEM173/STING-dependent DNA sensor pathway,” Nature Communications 11, no. 1 (2020): 6339.

[579]

J. P. Friedmann Angeli, D. V. Krysko, and M. Conrad, “Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion,” Nature Reviews Cancer 19, no. 7 (2019): 405-414.

[580]

D. Wang and R. N. DuBois, “The Role of Prostaglandin E(2) in Tumor-Associated Immunosuppression,” Trends in Molecular Medicine 22, no. 1 (2016): 1-3.

[581]

A. M. Johnson, E. K. Kleczko, and R. A. Nemenoff, “Eicosanoids in Cancer: New Roles in Immunoregulation,” Frontiers in pharmacology 11 (2020): 595498.

[582]

S. Zelenay, A. G. van der Veen, J. P. Böttcher, et al., “Cyclooxygenase-Dependent Tumor Growth Through Evasion of Immunity,” Cell 162, no. 6 (2015): 1257-1270.

[583]

J. P. Böttcher, E. Bonavita, P. Chakravarty, et al., “NK Cells Stimulate Recruitment of cDC1 Into the Tumor Microenvironment Promoting Cancer Immune Control,” Cell 172, no. 5 (2018): 1022-1037. e14.

[584]

D. Wang and R. N. DuBois, “Immunosuppression associated With chronic inflammation in the tumor microenvironment,” Carcinogenesis 36, no. 10 (2015): 1085-1093.

[585]

S. E. Weinberg, L. A. Sena, and N. S. Chandel, “Mitochondria in the regulation of innate and adaptive immunity,” Immunity 42, no. 3 (2015): 406-417.

[586]

M. A. Ligtenberg, D. Mougiakakos, M. Mukhopadhyay, et al., “Coexpressed Catalase Protects Chimeric Antigen Receptor-Redirected T Cells as well as Bystander Cells From Oxidative Stress-Induced Loss of Antitumor Activity,” J Immunol Baltim Md 1950 196, no. 2 (2016): 759-766.

[587]

N. E. Scharping, A. V. Menk, R. D. Whetstone, X. Zeng, and G. M. Delgoffe, “Efficacy of PD-1 Blockade Is Potentiated by Metformin-Induced Reduction of Tumor Hypoxia,” Cancer immunology research 5, no. 1 (2017): 9-16.

[588]

S. E. Weinberg, B. D. Singer, E. M. Steinert, et al., “Mitochondrial complex III is essential for suppressive function of regulatory T cells,” Nature 565, no. 7740 (2019): 495-499.

[589]

J. Wei, M. Zhang, and J. Zhou, “Myeloid-derived suppressor cells in major depression patients suppress T-cell responses Through the production of reactive oxygen species,” Psychiatry Research 228, no. 3 (2015): 695-701.

[590]

X. Ma, L. Xiao, L. Liu, et al., “CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability,” Cell metabolism 33, no. 5 (2021): 1001-1012. e5.

[591]

S. Xu, O. Chaudhary, P. Rodríguez-Morales, et al., “Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors,” Immunity 54, no. 7 (2021): 1561-1577. e7.

[592]

S. M. Poznanski, K. Singh, T. M. Ritchie, et al., “Metabolic flexibility determines human NK cell functional fate in the tumor microenvironment,” Cell metabolism 33, no. 6 (2021): 1205-1220. e5.

[593]

T. Rothe, F. Gruber, S. Uderhardt, et al., “12/15-Lipoxygenase-mediated enzymatic lipid oxidation regulates DC maturation and function,” Journal of Clinical Investigation 125, no. 5 (2015): 1944-1954.

[594]

Y. Gao, F. Souza-Fonseca-Guimaraes, T. Bald, et al., “Tumor immunoevasion by the conversion of effector NK cells Into type 1 innate lymphoid cells,” Nature Immunology 18, no. 9 (2017): 1004-1015.

[595]

D. N. Khalil, E. L. Smith, R. J. Brentjens, and J. D. Wolchok, “The future of cancer treatment: Immunomodulation, CARs and combination immunotherapy,” Nature reviews Clinical oncology 13, no. 6 (2016): 394.

[596]

W. Zou, J. D. Wolchok, and L. Chen, “PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations,” Science Translational Medicine 8, no. 328 (2016): 328rv4.

[597]

P. Jiang, S. Gu, D. Pan, et al., “Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response,” Nature Medicine 24, no. 10 (2018): 1550-1558.

[598]

J. L. Benci, L. R. Johnson, R. Choa, et al., “Opposing Functions of Interferon Coordinate Adaptive and Innate Immune Responses to Cancer Immune Checkpoint Blockade,” Cell 178, no. 4 (2019): 933-948. e14.

[599]

M. B. Dong, G. Wang, R. D. Chow, et al., “Systematic Immunotherapy Target Discovery Using Genome-Scale In Vivo CRISPR Screens in CD8 T Cells,” Cell 178, no. 5 (2019): 1189-1204. e23.

[600]

Q. Jiang, K. Wang, X. Zhang, et al., “Platelet Membrane-Camouflaged Magnetic Nanoparticles for Ferroptosis-Enhanced Cancer Immunotherapy,” Small Weinh Bergstr Ger 16, no. 22 (2020): e2001704.

[601]

R. Song, T. Li, J. Ye, et al., “Acidity-Activatable Dynamic Nanoparticles Boosting Ferroptotic Cell Death for Immunotherapy of Cancer,” Adv Mater Deerfield Beach Fla 33, no. 31 (2021): e2101155.

[602]

W. B. Coley, “The Treatment of Inoperable Sarcoma by Bacterial Toxins (the Mixed Toxins of the Streptococcus erysipelas and the Bacillus prodigiosus),” Proceedings of the Royal Society of Medicine 3 (1910): 1-48. Surg Sect.

[603]

L. T. Nguyen and P. S. Ohashi, “Clinical blockade of PD1 and LAG3-potential mechanisms of action,” Nature Reviews Immunology 15, no. 1 (2015): 45-56.

[604]

L. T. Nguyen and P. S. Ohashi, “Clinical blockade of PD1 and LAG3-potential mechanisms of action,” Nature Reviews Immunology 15, no. 1 (2015): 45-56.

[605]

W. Wang, I. Kryczek, L. Dostál, et al., “Effector T Cells Abrogate Stroma-Mediated Chemoresistance in Ovarian Cancer,” Cell 165, no. 5 (2016): 1092-1105.

[606]

W. Wang, M. Green, J. E. Choi, et al., “CD8+ T cells regulate tumour ferroptosis During cancer immunotherapy,” Nature 569, no. 7755 (2019): 270-274.

[607]

G. Delaney, S. Jacob, C. Featherstone, and M. Barton, “The role of radiotherapy in cancer treatment: Estimating optimal utilization From a review of evidence-based clinical guidelines,” Cancer 104, no. 6 (2005): 1129-1137.

[608]

K. E. Baidoo, K. Yong, and M. W. Brechbiel, “Molecular pathways: Targeted α-particle radiation therapy,” Clin Cancer Res Off J Am Assoc Cancer Res 19, no. 3 (2013): 530-537.

[609]

G. Lei, Y. Zhang, P. Koppula, et al., “The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression,” Cell Research 30, no. 2 (2020): 146-162.

[610]

L. F. Ye, K. R. Chaudhary, F. Zandkarimi, et al., “Radiation-Induced Lipid Peroxidation Triggers Ferroptosis and Synergizes With Ferroptosis Inducers,” Acs Chemical Biology 15, no. 2 (2020): 469-484.

[611]

X. Lang, M. D. Green, W. Wang, et al., “Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11,” Cancer discovery 9, no. 12 (2019): 1673-1685.

[612]

X. Lang, M. D. Green, W. Wang, et al., “Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11,” Cancer discovery 9, no. 12 (2019): 1673-1685.

[613]

G. Lei, Y. Zhang, P. Koppula, et al., “The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression,” Cell Research 30, no. 2 (2020): 146-162.

[614]

X. Li, L. Duan, S. Yuan, X. Zhuang, T. Qiao, and J. He, “Ferroptosis inhibitor alleviates Radiation-induced lung fibrosis (RILF) via Down-regulation of TGF-β1,” J Inflamm Lond Engl 16 (2019): 11.

[615]

X. Zhang, X. Xing, H. Liu, et al., “Ionizing radiation induces ferroptosis in granulocyte-macrophage hematopoietic progenitor cells of murine bone marrow,” International Journal of Radiation Biology 96, no. 5 (2020): 584-595.

[616]

O. Warburg and S. Minami, “Versuche an Überlebendem Carcinom-gewebe,” Klinische Wochenschrift 2, no. 17 (1923): 776-777.

[617]

M. V. Gwangwa, A. M. Joubert, and M. H. Visagie, “Crosstalk Between the Warburg effect, redox regulation and autophagy induction in tumourigenesis,” Cellular & Molecular Biology Letters 23 (2018): 20.

[618]

N. E. Scharping, D. B. Rivadeneira, A. V. Menk, et al., “Mitochondrial stress induced by continuous stimulation Under hypoxia rapidly drives T cell exhaustion,” Nature Immunology 22, no. 2 (2021): 205-215.

[619]

C. M. Bebber, F. Müller, L. Prieto Clemente, J. Weber, and S. von Karstedt, “Ferroptosis in Cancer Cell Biology,” Cancers 12, no. 1 (2020): 164.

[620]

S. K. Watkins, N. K. Egilmez, J. Suttles, and R. D. Stout, “IL-12 rapidly alters the functional profile of tumor-associated and tumor-infiltrating macrophages in vitro and in vivo,” J Immunol Baltim Md 1950 178, no. 3 (2007): 1357-1362.

[621]

S. Nucera, D. Biziato, and M. De Palma, “The interplay Between macrophages and angiogenesis in development, tissue injury and regeneration,” International Journal of Developmental Biology 55, no. 4-5 (2011): 495-503.

[622]

E. Dai, L. Han, J. Liu, et al., “Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein,” Autophagy 16, no. 11 (2020): 2069-2083.

[623]

Q. Jiang, K. Wang, X. Zhang, et al., “Platelet Membrane-Camouflaged Magnetic Nanoparticles for Ferroptosis-Enhanced Cancer Immunotherapy,” Small Weinh Bergstr Ger 16, no. 22 (2020): e2001704.

[624]

C. Wan, Y. Sun, Y. Tian, et al., “Irradiated tumor cell-derived microparticles mediate tumor eradication via cell killing and immune reprogramming,” Science Advances 6, no. 13 (2020): eaay9789.

[625]

T. Xu, Y. Ma, Q. Yuan, et al., “Enhanced Ferroptosis by Oxygen-Boosted Phototherapy Based on a 2-in-1 Nanoplatform of Ferrous Hemoglobin for Tumor Synergistic Therapy,” ACS Nano 14, no. 3 (2020): 3414-3425.

[626]

V. D. Turubanova, I. V. Balalaeva, T. A. Mishchenko, et al., “Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine,” Journal for ImmunoTherapy of Cancer 7, no. 1 (2019): 350.

[627]

C. D. Mills, K. Kincaid, J. M. Alt, M. J. Heilman, and A. M. Hill, “M-1/M-2 macrophages and the Th1/Th2 paradigm,” J Immunol Baltim Md 1950 164, no. 12 (2000): 6166-6173.

[628]

F. Zhang, F. Li, G. H. Lu, et al., “Engineering Magnetosomes for Ferroptosis/Immunomodulation Synergism in Cancer,” ACS Nano 13, no. 5 (2019): 5662-5673.

[629]

A. Linkermann, R. Skouta, N. Himmerkus, et al., “Synchronized renal tubular cell death involves ferroptosis,” PNAS 111, no. 47 (2014): 16836-16841.

[630]

R. Tang, J. Xu, B. Zhang, et al., “Ferroptosis, necroptosis, and pyroptosis in anticancer immunity,” J Hematol OncolJ Hematol Oncol 13, no. 1 (2020): 110.

[631]

T. L. Aaes, A. Kaczmarek, T. Delvaeye, et al., “Vaccination With Necroptotic Cancer Cells Induces Efficient Anti-tumor Immunity,” Cell reports 15, no. 2 (2016): 274-287.

[632]

W. Li, G. Feng, J. M. Gauthier, et al., “Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment After heart transplantation,” Journal of Clinical Investigation 129, no. 6 (2019): 2293-2304.

[633]

V. D. Turubanova, I. V. Balalaeva, T. A. Mishchenko, et al., “Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine,” Journal for ImmunoTherapy of Cancer 7, no. 1 (2019): 350.

[634]

B. Proneth and M. Conrad, “Ferroptosis and necroinflammation, a yet poorly explored link,” Cell Death and Differentiation 26, no. 1 (2019): 14-24.

[635]

H. Wang, O. Bloom, M. Zhang, et al., “HMG-1 as a late mediator of endotoxin lethality in mice,” Science 285, no. 5425 (1999): 248-251.

[636]

Q. Wen, J. Liu, R. Kang, B. Zhou, and D. Tang, “The release and activity of HMGB1 in ferroptosis,” Biochemical and Biophysical Research Communications 510, no. 2 (2019): 278-283.

[637]

A. D. Garg, D. V. Krysko, P. Vandenabeele, and P. Agostinis, “Hypericin-based photodynamic therapy induces surface exposure of damage-associated molecular patterns Like HSP70 and calreticulin,” Cancer Immunol Immunother CII 61, no. 2 (2012): 215-221.

[638]

J. Tsoi, L. Robert, K. Paraiso, et al., “Multi-stage differentiation defines melanoma subtypes With differential vulnerability to drug-induced iron-dependent oxidative stress,” Cancer Cell 33, no. 5 (2018): 890-904. e5.

[639]

V. S. Viswanathan, M. J. Ryan, H. D. Dhruv, et al., “Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway,” Nature 547, no. 7664 (2017): 453-457.

[640]

X. Chen, R. Kang, G. Kroemer, and D. Tang, “Broadening horizons: The role of ferroptosis in cancer,” Nature reviews Clinical oncology 18, no. 5 (2021): 280-296.

[641]

H. Wang, Y. Cheng, C. Mao, et al., “Emerging mechanisms and targeted therapy of ferroptosis in cancer,” Mol Ther J Am Soc Gene Ther 29, no. 7 (2021): 2185-2208.

[642]

G. Delaney, S. Jacob, C. Featherstone, and M. Barton, “The role of radiotherapy in cancer treatment: Estimating optimal utilization From a review of evidence-based clinical guidelines,” Cancer 104, no. 6 (2005): 1129-1137.

[643]

K. E. Baidoo, K. Yong, and M. W. Brechbiel, “Molecular pathways: Targeted α-particle radiation therapy,” Clin Cancer Res Off J Am Assoc Cancer Res 19, no. 3 (2013): 530-537.

[644]

A. Balihodzic, F. Prinz, M. A. Dengler, G. A. Calin, P. J. Jost, and M. Pichler, “Non-coding RNAs and ferroptosis: Potential implications for cancer therapy,” Cell Death and Differentiation 29, no. 6 (2022): 1094-1106.

[645]

Y. Luo, G. Niu, H. Yi, et al., “Nanomedicine promotes ferroptosis to inhibit tumour proliferation in vivo,” Redox Biology 42 (2021): 101908.

[646]

S. H. Deng, D. M. Wu, L. Li, et al., “miR-324-3p reverses cisplatin resistance by inducing GPX4-mediated ferroptosis in lung adenocarcinoma cell line A549,” Biochemical and Biophysical Research Communications 549 (2021): 54-60.

[647]

Z. Song, G. Jia, P. Ma, and S. Cang, “Exosomal miR-4443 promotes cisplatin resistance in non-small cell lung carcinoma by regulating FSP1 m6A modification-mediated ferroptosis,” Life Sciences 276 (2021): 119399.

[648]

X. Deng, W. Xiong, X. Jiang, et al., “LncRNA LINC00472 regulates cell stiffness and inhibits the migration and invasion of lung adenocarcinoma by binding to YBX1,” Cell death & disease 11, no. 11 (2020): 945.

[649]

C. Gai, C. Liu, X. Wu, et al., “MT1DP loaded by folate-modified liposomes sensitizes erastin-induced ferroptosis via regulating miR-365a-3p/NRF2 axis in non-small cell lung cancer cells,” Cell death & disease 11, no. 9 (2020): 751.

[650]

L. H. Dong, J. J. Huang, P. Zu, et al., “CircKDM4C upregulates P53 by sponging hsa-let-7b-5p to induce ferroptosis in acute myeloid leukemia,” Environmental Toxicology 36, no. 7 (2021): 1288-1302.

[651]

P. Wu, C. Li, D. M. Ye, et al., “Circular RNA circEPSTI1 accelerates cervical cancer progression via miR-375/409-3P/515-5p-SLC7A11 axis,” Aging 13, no. 3 (2021): 4663-4673.

[652]

Z. Liu, Q. Wang, X. Wang, Z. Xu, X. Wei, and J. Li, “Circular RNA cIARS regulates ferroptosis in HCC cells Through interacting With RNA binding protein ALKBH5,” Cell Death Discov 6 (2020): 72.

[653]

Y. Tang, C. Li, Y. J. Zhang, and Z. H. Wu, “Ferroptosis-Related Long Non-Coding RNA signature predicts the prognosis of Head and neck squamous cell carcinoma,” Int J Biol Sci 17, no. 3 (2021): 702-711.

[654]

G. Lei, L. Zhuang, and B. Gan, “Targeting ferroptosis as a vulnerability in cancer,” Nature Reviews Cancer 22, no. 7 (2022): 381-396.

[655]

M. J. Hangauer, V. S. Viswanathan, M. J. Ryan, et al., “Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition,” Nature 551, no. 7679 (2017): 247-250.

[656]

V. S. Viswanathan, M. J. Ryan, H. D. Dhruv, et al., “Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway,” Nature 547, no. 7664 (2017): 453-457.

[657]

J. Tsoi, L. Robert, K. Paraiso, et al., “Multi-stage Differentiation Defines Melanoma Subtypes With Differential Vulnerability to Drug-Induced Iron-Dependent Oxidative Stress,” Cancer Cell 33, no. 5 (2018): 890-904. e5.

[658]

Y. Zou, W. S. Henry, E. L. Ricq, et al., “Plasticity of ether lipids promotes ferroptosis susceptibility and evasion,” Nature 585, no. 7826 (2020): 603-608.

[659]

C. Mao, X. Liu, Y. Zhang, et al., “DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer,” Nature 593, no. 7860 (2021): 586-590.

[660]

S. Zhang, L. Kang, X. Dai, et al., “Manganese induces tumor cell ferroptosis Through type-I IFN dependent inhibition of mitochondrial dihydroorotate dehydrogenase,” Free Radic Biol Med 193 (2022): 202-212. Pt 1.

[661]

W. Zhang, J. Wang, Z. Liu, et al., “Iron-dependent ferroptosis participated in benzene-induced anemia of inflammation Through IRP1-DHODH-ALOX12 axis,” Free Radic Biol Med 193 (2022): 122-133. Pt 1.

[662]

J. A. Stefely and D. J. Pagliarini, “Biochemistry of Mitochondrial Coenzyme Q Biosynthesis,” Trends in Biochemical Sciences 42, no. 10 (2017): 824-843.

[663]

N. Rizzardi, I. Liparulo, G. Antonelli, et al., “Coenzyme Q10 Phytosome Formulation Improves CoQ10 Bioavailability and Mitochondrial Functionality in Cultured Cells,” Antioxid Basel Switz 10, no. 6 (2021): 927.

[664]

H. Lee, F. Zandkarimi, Y. Zhang, et al., “Energy-stress-mediated AMPK activation inhibits ferroptosis,” Nature Cell Biology 22, no. 2 (2020): 225-234.

[665]

B. Gan, “Mitochondrial regulation of ferroptosis,” Journal of Cell Biology 220, no. 9 (2021): e202105043.

[666]

W. S. Yang, R. SriRamaratnam, M. E. Welsch, et al., “Regulation of ferroptotic cancer cell death by GPX4,” Cell 156, no. 1-2 (2014): 317-331.

[667]

R. Brigelius-Flohé, and M. Maiorino, “Glutathione peroxidases,” Biochimica Et Biophysica Acta 1830, no. 5 (2013): 3289-3303.

[668]

T. C. Chen, J. Y. Chuang, C. Y. Ko, et al., “AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide-resistant glioblastoma Through disrupting GPX4-Mediated redox homeostasis,” Redox Biology 30 (2020): 101413.

[669]

C. Yang, Y. Zhang, S. Lin, Y. Liu, and W. Li, “Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin,” Aging 13, no. 10 (2021): 13515-13534.

[670]

J. Du, X. Wang, Y. Li, et al., “DHA exhibits synergistic therapeutic efficacy With cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via modulation of iron metabolism,” Cell death & disease 12, no. 7 (2021): 705.

[671]

N. Chaudhary, B. S. Choudhary, S. G. Shah, et al., “Lipocalin 2 expression promotes tumor progression and therapy resistance by inhibiting ferroptosis in colorectal cancer,” International Journal of Cancer 149, no. 7 (2021): 1495-1511.

[672]

A. L. Turcu, A. Versini, N. Khene, et al., “DMT1 Inhibitors Kill Cancer Stem Cells by Blocking Lysosomal Iron Translocation,” Chem - Eur J 26, no. 33 (2020): 7369-7373.

[673]

J. L. Roh, E. H. Kim, H. J. Jang, J. Y. Park, and D. Shin, “Induction of ferroptotic cell death for overcoming cisplatin resistance of head and neck cancer,” Cancer Letters 381, no. 1 (2016): 96-103.

[674]

D. Fu, C. Wang, L. Yu, and R. Yu, “Induction of ferroptosis by ATF3 elevation alleviates cisplatin resistance in gastric cancer by restraining Nrf2/Keap1/xCT signaling,” Cellular & Molecular Biology Letters 26, no. 1 (2021): 26.

[675]

D. Tang, O. Kepp, and G. Kroemer, “Ferroptosis becomes immunogenic: Implications for anticancer treatments,” Oncoimmunology 10, no. 1 (2020): 1862949.

[676]

X. Luo, H. B. Gong, H. Y. Gao, et al., “Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting With TLR2,” Cell Death and Differentiation 28, no. 6 (2021): 1971-1989.

[677]

C. Xu, S. Sun, T. Johnson, et al., “The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity,” Cell reports 35, no. 11 (2021): 109235.

[678]

A. A. Kapralov, Q. Yang, H. H. Dar, et al., “Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death,” Nature Chemical Biology 16, no. 3 (2020): 278-290.

[679]

Z. Jiang, S. O. Lim, M. Yan, et al., “TYRO3 induces anti-PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis,” Journal of Clinical Investigation 131, no. 8 (2021): e139434. 139434.

[680]

G. Lyu, H. Liao, and R. Li, “Ferroptosis and renal fibrosis: Mechanistic insights and emerging therapeutic targets,” Renal Failure 47, no. 1 (2025): 2498629.

[681]

M. Zhang, C. Liu, J. Tu, et al., “Advances in cancer immunotherapy: Historical perspectives, current developments, and future directions,” Molecular cancer 24, no. 1 (2025): 136.

[682]

X. Zhang, Y. Duan, S. Li, et al., “CRISPR screening identifies PRMT1 as a key pro-ferroptotic gene via a two-layer regulatory mechanism,” Cell reports 43, no. 9 (2024): 114662.

[683]

X. Wu, Z. Bai, H. Wang, et al., “CRISPR-Cas9 gene editing strengthens cuproptosis/chemodynamic/ferroptosis synergistic cancer therapy,” Acta Pharm Sin B 14, no. 9 (2024): 4059-4072.

[684]

Y. Xiao, M. He, X. Zhang, et al., “Research progress on the mechanism of tumor cell ferroptosis regulation by epigenetics,” Epigenetics 20, no. 1 (2025): 2500949.

[685]

W. S. Yang, R. SriRamaratnam, M. E. Welsch, et al., “Regulation of ferroptotic cancer cell death by GPX4,” Cell 156, no. 1-2 (2014): 317-331.

[686]

A. C. Araújo, C. E. Wheelock, and J. Z. Haeggström, “The Eicosanoids, Redox-Regulated Lipid Mediators in Immunometabolic Disorders,” Antioxid Redox Signaling 29, no. 3 (2018): 275-296.

[687]

C. Li, X. Deng, X. Xie, Y. Liu, J. P. Friedmann Angeli, and L. Lai, “Activation of Glutathione Peroxidase 4 as a Novel Anti-inflammatory Strategy,” Frontiers in pharmacology 9 (2018): 1120.

[688]

A. J. Marrogi, W. D. Travis, J. A. Welsh, et al., “Nitric oxide synthase, cyclooxygenase 2, and vascular endothelial growth factor in the angiogenesis of non-small cell lung carcinoma,” Clin Cancer Res Off J Am Assoc Cancer Res 6, no. 12 (2000): 4739-4744.

[689]

X. Sun, Z. Ou, M. Xie, et al., “HSPB1 as a novel regulator of ferroptotic cancer cell death,” Oncogene 34, no. 45 (2015): 5617-5625.

[690]

L. Jiang, N. Kon, T. Li, et al., “Ferroptosis as a p53-mediated activity During tumour suppression,” Nature 520, no. 7545 (2015): 57-62.

[691]

Y. Ou, S. J. Wang, D. Li, B. Chu, and W. Gu, “Activation of SAT1 engages polyamine metabolism With p53-mediated ferroptotic responses,” PNAS 113, no. 44 (2016): E6806-E6812.

[692]

Y. Zhang, J. Shi, X. Liu, et al., “BAP1 links metabolic regulation of ferroptosis to tumour suppression,” Nature Cell Biology 20, no. 10 (2018): 1181-1192.

[693]

S. J. Wang, D. Li, Y. Ou, et al., “Acetylation Is Crucial for p53-Mediated Ferroptosis and Tumor Suppression,” Cell reports 17, no. 2 (2016): 366-373.

[694]

G. Kroemer, C. Galassi, L. Zitvogel, and L. Galluzzi, “Immunogenic cell stress and death,” Nature Immunology 23, no. 4 (2022): 487-500.

[695]

D. Shen, J. Luo, L. Chen, et al., “PARPi treatment enhances radiotherapy-induced ferroptosis and antitumor immune responses via the cGAS signaling pathway in colorectal cancer,” Cancer Letters 550 (2022): 215919.

[696]

J. L. Liang, X. K. Jin, S. M. Zhang, et al., “Specific activation of cGAS-STING pathway by nanotherapeutics-mediated ferroptosis evoked endogenous signaling for boosting systemic tumor immunotherapy,” Sci Bull 68, no. 6 (2023): 622-636.

[697]

Q. Ding, W. Tang, X. Li, et al., “Mitochondrial-targeted brequinar liposome boosted mitochondrial-related ferroptosis for promoting checkpoint blockade immunotherapy in bladder cancer,” Journal of Controlled Release 363 (2023): 221-234.

[698]

E. Dai, L. Han, J. Liu, et al., “Ferroptotic damage promotes pancreatic tumorigenesis Through a TMEM173/STING-dependent DNA sensor pathway,” Nature Communications 11, no. 1 (2020): 6339.

[699]

C. Fang, F. Mo, L. Liu, et al., “Oxidized mitochondrial DNA sensing by STING signaling promotes the antitumor effect of an irradiated immunogenic cancer cell vaccine,” Cell Mol Immunol 18, no. 9 (2021): 2211-2223.

[700]

A. M. Johnson, E. K. Kleczko, and R. A. Nemenoff, “Eicosanoids in Cancer: New Roles in Immunoregulation,” Frontiers in pharmacology 11 (2020): 595498.

[701]

P. Ni, J. Xie, C. Chen, Y. Jiang, Y. Lu, and X. Hu, “Fluorometric determination of the activity of alkaline phosphatase and its inhibitors based on ascorbic acid-induced aggregation of carbon dots,” Microchimica Acta 186, no. 3 (2019): 202.

[702]

Y. Hu, D. Chen, M. Hong, et al., “Apoptosis, Pyroptosis, and Ferroptosis Conspiringly Induce Immunosuppressive Hepatocellular Carcinoma Microenvironment and γδ T-Cell Imbalance,” Frontiers in immunology 13 (2022): 845974.

[703]

E. Dai, L. Han, J. Liu, et al., “Ferroptotic damage promotes pancreatic tumorigenesis Through a TMEM173/STING-dependent DNA sensor pathway,” Nature Communications 11, no. 1 (2020): 6339.

[704]

I. Efimova, E. Catanzaro, L. Van der Meeren, et al., “Vaccination With early ferroptotic cancer cells induces efficient antitumor immunity,” Journal for ImmunoTherapy of Cancer 8, no. 2 (2020): e001369.

[705]

C. Conche, F. Finkelmeier, M. Pešić, et al., “Combining ferroptosis induction With MDSC blockade renders primary tumours and metastases in liver sensitive to immune checkpoint blockade,” Gut 72, no. 9 (2023): 1774-1782.

[706]

N. Chande, D. J. Tsoulis, and J. K. MacDonald, “Azathioprine or 6-mercaptopurine for induction of remission in Crohn's disease,” Cochrane Database of Systematic Reviews (Online), no. 4 (2013): CD000545, https://doi.org/10.1002/14651858.CD000545.pub4.

[707]

W. Wang, M. Green, J. E. Choi, et al., “CD8+ T cells regulate tumour ferroptosis During cancer immunotherapy,” Nature 569, no. 7755 (2019): 270-274.

[708]

V. S. Viswanathan, M. J. Ryan, H. D. Dhruv, et al., “Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway,” Nature 547, no. 7664 (2017): 453-457.

[709]

A. C. Araújo, C. E. Wheelock, and J. Z. Haeggström, “The Eicosanoids, Redox-Regulated Lipid Mediators in Immunometabolic Disorders,” Antioxid Redox Signaling 29, no. 3 (2018): 275-296.

[710]

X. Liu, T. Wang, W. Wang, et al., “Emerging Potential Therapeutic Targets of Ferroptosis in Skeletal Diseases,” Oxid Med Cell Longev 2022 (2022): 3112388.

[711]

M. J. Hangauer, V. S. Viswanathan, M. J. Ryan, et al., “Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition,” Nature 551, no. 7679 (2017): 247-250.

[712]

X. Chen, R. Kang, G. Kroemer, and D. Tang, “Broadening horizons: The role of ferroptosis in cancer,” Nature reviews Clinical oncology 18, no. 5 (2021): 280-296.

[713]

T. Liu, W. Liu, M. Zhang, et al., “Ferrous-Supply-Regeneration Nanoengineering for Cancer-Cell-Specific Ferroptosis in Combination With Imaging-Guided Photodynamic Therapy,” ACS Nano 12, no. 12 (2018): 12181-12192.

[714]

D. Shin, E. H. Kim, J. Lee, and J. L. Roh, “Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer,” Free Radic Biol Med 129 (2018): 454-462.

[715]

F. Ursini, M. Maiorino, M. Valente, L. Ferri, and C. Gregolin, “Purification From pig liver of a protein which protects liposomes and biomembranes From peroxidative degradation and exhibits glutathione peroxidase activity on phosphatidylcholine hydroperoxides,” Biochimica Et Biophysica Acta 710, no. 2 (1982): 197-211.

[716]

K. Shimada, R. Skouta, A. Kaplan, et al., “Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis,” Nature Chemical Biology 12, no. 7 (2016): 497-503.

[717]

H. Fan, G. Yan, Z. Zhao, et al., “A Smart Photosensitizer-Manganese Dioxide Nanosystem for Enhanced Photodynamic Therapy by Reducing Glutathione Levels in Cancer Cells,” Angewandte Chemie (International ed in English) 55, no. 18 (2016): 5477-5482.

[718]

Z. Cong, F. Yuan, H. Wang, et al., “BTB domain and CNC homolog 1 promotes glioma invasion mainly Through regulating extracellular matrix and increases ferroptosis sensitivity,” Biochim Biophys Acta Mol Basis Dis 1868, no. 12 (2022): 166554.

[719]

E. Dierge, E. Debock, C. Guilbaud, et al., “Peroxidation of n-3 and n-6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis-mediated anticancer effects,” Cell metabolism 33, no. 8 (2021): 1701-1715. e5.

[720]

G. Lei, L. Zhuang, and B. Gan, “The roles of ferroptosis in cancer: Tumor suppression, tumor microenvironment, and therapeutic interventions,” Cancer Cell 42, no. 4 (2024): 513-534.

[721]

X. Chen, R. Kang, G. Kroemer, and D. Tang, “Broadening horizons: The role of ferroptosis in cancer,” Nature reviews Clinical oncology 18, no. 5 (2021): 280-296.

[722]

N. Ebrahimi, S. Adelian, S. Shakerian, et al., “Crosstalk Between ferroptosis and the epithelial-mesenchymal transition: Implications for inflammation and cancer therapy,” Cytokine & Growth Factor Reviews 64 (2022): 33-45.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

16

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/