Skip to main content
Epigenetics logoLink to Epigenetics
. 2025 May 6;20(1):2500949. doi: 10.1080/15592294.2025.2500949

Research progress on the mechanism of tumor cell ferroptosis regulation by epigenetics

Yuyang Xiao a,b, Mengyang He b, Xupeng Zhang b, Meng Yang b, Zhangchi Yuan b, Shanhu Yao c,d, Yuexiang Qin a,e,
PMCID: PMC12064064  PMID: 40327848

ABSTRACT

Cancer remains a significant barrier to human longevity and a leading cause of mortality worldwide. Despite advancements in cancer therapies, challenges such as cellular toxicity and drug resistance to chemotherapy persist. Regulated cell death (RCD), once regarded as a passive process, is now recognized as a programmed mechanism with distinct biochemical and morphological characteristics, thereby presenting new therapeutic opportunities. Ferroptosis, a novel form of RCD characterized by iron-dependent lipid peroxidation and unique mitochondrial damage, differs from apoptosis, autophagy, and necroptosis. It is driven by reactive oxygen species (ROS)-induced lipid peroxidation and is implicated in tumorigenesis, anti-tumor immunity, and resistance, particularly in tumors undergoing epithelial-mesenchymal transition. Moreover, ferroptosis is associated with ischemic organ damage, degenerative diseases, and aging, regulated by various cellular metabolic processes, including redox balance, iron metabolism, and amino acid, lipid, and glucose metabolism. This review focuses on the role of epigenetic factors in tumor ferroptosis, exploring their mechanisms and potential applications in cancer therapy. It synthesizes current knowledge to provide a comprehensive understanding of epigenetic regulation in tumor cell ferroptosis, offering insights for future research and clinical applications.

KEYWORDS: Ferroptosis, epigenetic regulation, cancer therapy, tumor development, regulated cell death

Introduction

Ferroptosis is a distinct form of regulated cell death (RCD), first introduced by B.R. Stockwell in 2012 [1]. It is characterized by the excessive intracellular accumulation of iron ions and reactive oxygen species (ROS), which trigger lipid peroxidation and ultimately lead to cell death [1,2] (Figure 1). The defense mechanisms for ferroptosis are cellular antioxidant systems that balance lipid peroxidation, each with a unique subcellular localization and complex regulatory mechanisms, including SLC7A11–GSH – GPX4 defense system, FSP1-CoQH2 defense system, GCH1-BH4 defense system, DHODH-CoQH2 defense system and Thioredoxin antioxidant system. Ferroptosis can be regulated by directly or indirectly inhibiting lipid peroxidation or by pharmacologically reducing intracellular iron levels through epigenetic mechanisms [1,3]. Additionally, it is influenced by various pathways, including redox homeostasis, iron metabolism, mitochondrial stability, and the metabolism of amino acids, lipids, and glucose [4], thereby playing a crucial role in cellular functions. Research indicates that ferroptosis is significantly involved in tumor development, anti-tumor immunity, tumor resistance, and other related processes [5–7], as well as in ischemic organ damage, degenerative diseases, and cellular senescence [8], highlighting its substantial research significance.

Figure 1.

Figure 1.

Schematic diagram of the feature comparisons between ferroptosis and other cell death modes.

Numerous studies have demonstrated that ferroptosis is precisely regulated by protein post-translational modifications (PTMs) and epigenetic modifications [3,9–12]. Epigenetic modifications refer to heritable changes in gene activity that occur through covalent modifications of nucleic acids and histones, collectively regulating gene function, expression, and chromatin structure [13,14] without altering the underlying DNA sequence [15]. These modifications are reversible and dynamically regulated, occurring at various levels of gene expression, including DNA methylation, histone PTMs, chromatin remodeling (the dynamic spatiotemporal localization of nucleosomes), and non-coding RNA (ncRNA) regulation [13,14]. Epigenetic mechanisms comprise various enzymes and protein domains [16] that fine-tune gene expression programs, thereby controlling essential biological processes such as cell differentiation and embryogenesis [17].

Cancer poses one of the greatest threats to global public health, significantly impacting human health and socio-economic development [18–20]. According to WHO estimates, approximately 20 million new cases of malignant tumors and 9.7 million related deaths occurred worldwide in 2022, accounting for two-thirds of new cases and deaths globally. It is projected that the number of new cases will reach 35 million by 2025, representing a 77% increase over 2022. Understanding the mechanisms underlying cancer occurrence and development, as well as researching cancer prevention, diagnosis, and treatment, remains a primary focus of medical research. Currently, cancer treatment primarily involves surgical resection, radiotherapy, and chemotherapy [21]; however, these conventional methods carry risks of recurrence and metastasis and often have significant side effects. Therefore, exploring novel cancer treatment strategies is urgent. Research has shown that uncontrolled ferroptosis plays a critical role in tumor development, closely linked to the regulation of epigenetics on ferroptosis in tumor cells, thus providing a new avenue for tumor treatment [3].

This review summarizes the molecular mechanisms of ferroptosis and then discusses the role of epigenetic regulation of ferroptosis in tumor development. Finally, we explore the potential applications of targeted ferroptosis epigenetic regulators in tumor therapy and anticipate future advancements in this field.

Molecular mechanisms of ferroptosis

Main characteristics of ferroptosis

Compared with other modes of cell death such as apoptosis, necrosis, and autophagy, ferroptosis is morphologically, biochemically, immunologically unique [1,22]. Each of these aspects is discussed below.

Morphological characteristics of ferroptosis

Cells undergoing ferroptosis show a series of specific morphological changes, including loss of cytoplasmic membrane integrity and swelling of the cytoplasm and organelles [23], but the nucleus remains intact, with no chromatin condensation or margination. Ferroptosis under certain conditions is accompanied by cell detachment and aggregation, with a corresponding increase in the number of autophagic vesicles [24]. More uniquely, cellular ferroptosis can rapidly propagate along intercellular contacts in the form of waves that spread to neighboring cells [25], but the mechanism remains unclear. At the ultrastructural level, iron-dead cells usually exhibit mitochondrial abnormalities, including mitochondrial condensation, decreased number, reduced volume, increased density of the bilayer membrane, decreased or absent mitochondrial cristae, and rupture of the outer membrane [26]. It has been shown that mitochondria-mediated ROS production, DNA stress and metabolic reprogramming are required for the induction of lipid peroxidation and ferroptosis [23,27].

Biochemical characterization of ferroptosis

Iron ion metabolism has a key role in ferroptosis, lipid peroxidation initiation can be hindered by iron chelators, and Fe2+ supplementation increases cellular sensitivity to ferroptosis inducers [1]. The accumulation of intracellular free iron ions directly triggers cellular ferroptosis, along with excess ferrous and nonferrous heme iron [24,28]. Overaccumulation of Fe2+ correlates significantly with ROS overload and lipid peroxidation [29], Fe2+ oxidizes lipids through the Fenton reaction and causes a cascade reaction occurs, leading to significant ROS accumulation, and when intracellular reduced glutathione (GSH) is depleted or glutathione peroxidase 4 (GPX4) activity is reduced, ROS are not scavenged in a timely manner, and the cell will undergo ferroptosis [30]. And then the cellular antioxidant defense system is suppressed, oxidative stress is exacerbated, and intracellular antioxidant enzymes suffer, further deteriorating the oxidative state of the cells [31].

Lipid metabolism during ferroptosis is also significantly characterized. During ferroptosis, the concentration of reactive aldehydes such as malondialdehyde (MDA) and 4-hydroxynonenal (4HNE), which are important products of lipid peroxidation, continues to rise [32]. Furthermore, peroxidation of polyunsaturated fatty acids (PUFAs) plays a key role in ferroptosis [32]. Notably, lipid peroxidation in ferroptosis is not an isolated event; it is closely related to other intracellular biochemical reactions, which together lead to impairment of cellular structure and function and drive the ferroptosis process [33].

Immunological characterization of ferroptosis

The effects of ferroptosis on immunity are manifold. Ferroptosis damages leukocytes and affects immune function; lipid peroxidation induces ferroptosis in T cells and exacerbates viral and parasitic infections [34]. In addition, ferroptosis alters the immune system’s handling of antigens released by dead cells and the corresponding antibody production [35]. In ferroptosis, 4HNE regulates the expression of a variety of genes associated with immune and inflammatory responses through the activation of transcription factors such as NF-κB [36], which affects cell signaling and gene expression and regulates immune and inflammatory responses [37]. Ferroptosis may also interfere with the clearance of dead cells, affect the release and activation of damage-associated molecular patterns (DAMP), and exacerbate immune, inflammatory, and autoimmune responses [34,38], affecting an individual’s health and disease state.

Ferroptosis inducers and inhibitors

Four classes of ferroptosis inducers have been identified, Erastin, RSL3 and its analogs ML162 and ML210, FIN56 and FINO2, which induce ferroptosis in different ways. Inhibitors of ferroptosis are the oxidizing inhibitor Ferrostatin-1 (Fer-1), Deferoxamine (DFO) and water soluble Vitamin E. The targets and mechanisms by which the above substances induce and inhibit ferroptosis are detailed in Table 1. With in-depth studies on the mechanisms of ferroptosis, their inducers and inhibitors will provide new ideas for the treatment of many diseases [1].

Table 1.

Ferroptosis inducers and inhibitors.

Reagents Classification Targets Mechanism of action References
Ferroptosis Inducers Erastin SLC7A11 Reduces GSH levels by inhibiting SLC7A11 thereby inducing cellular ferroptosis. Dixon et al. [1]
Erastin VDAC2, VDAC3 Targeting VDAC2 and VDAC3 leads to cell death by inducing mitochondrial dysfunction. Dixon et al. [1]
Erastin Erastin accumulation in the endoplasmic reticulum suggests that the endoplasmic reticulum may also be one of the key target sites for Erastin-induced ferroptosis to occur. Gaschler et al. [39]
It has been found that lipid hydroperoxides are mainly concentrated in the endoplasmic reticulum, which corroborates the existence of a complex mechanism for Erastin-induced ferroptosis. Kagan et al. [40]
RSL3, ML162, ML210 GPX4 Inhibition of GPX4 activity in cells and mitochondria increases lipid ROS and lipid peroxidation. Yang and Stockwell [41], Weïwer et al. [42]
FIN56 Directly promotes the degradation of GPX4. Shimada et al. [43]
FIN56 Binding to squalene synthase leads to depletion of endogenous antioxidant coenzyme Q10 and enhances cellular sensitivity to FIN56-induced ferroptosis. Shimada et al.[43]
FINO2 Induces mitochondrial lipid peroxidation and oxidized Fe2+ production, which leads to ferroptosis due to the combined effects of direct oxidation of unstable iron and GPX4 inactivation. Abrams et al. [44], Gaschler et al. [45]
FAC Promotes iron overload via Fe3+ accumulation, enhancing Fenton reactions and lipid peroxidation. Battaglia et al. [46]
Ferlixit Provides exogenous Fe3+, increases intracellular LIP, and induces ferroptosis in cells. Battaglia et al.[46]
Ferroptosis Inhibitors Fer-1 ROS Remove oxides. Dixon et al. [1]
  DFO ROS Remove oxides. Dixon et al. [1]
  Vitamin E ROS Remove oxides. Dixon et al. [1]

Mechanisms of non-epigenetic regulation of ferroptosis

Four major mechanisms of non-epigenetic regulation of ferroptosis play an important role in ferroptosis, including iron ion metabolism, lipid metabolism, oxygen-containing radicals, and the degradation system.The four mechanisms of non-epigenetic regulation of ferroptosis are listed in Table 2. Exploring the intricate mechanisms underlying ferroptosis reveals novel therapeutic perspectives for the treatment of a spectrum of diseases.

Table 2.

Mechanisms of non-epigenetic regulation of ferroptosis.

Methods of regulation Interacting molecule Target of action Effect of action References
Ferrous ion metabolism Ferrous ion chelator Increase in intracellular unstable ferrous ions during the process, effectively inhibiting the onset of ferroptosis. Dixon et al. [1], Holliday [15]
Ferrous ion Electron transport chain, redox metabolism, and DNA synthesis Affects cellular sensitivity to ferroptosis. Abnormalities in iron intake, storage, utilization, and metabolism may lead to pathological accumulation of iron and enhance cellular lipid peroxidation damage due to the Fenton reaction. Dixon et al. [1], Friedmann Angeli and Conrad [47]
TF Non-heme iron Uptake of exogenous iron ions. TF dissociates Fe3+ at low pH in vesicles. Iron ion uptake and transport are inhibited in the absence of TF function, as is ferroptosis. Alim et al. [48], Bersuker et al. [49]
TFRC TF incorporating non-heme iron TFs that bind nonheme iron are recognized in the cell membrane and cytosolized into the cell, forming specialized vesicles. Iron ion uptake and transport are inhibited in the absence of TFRC function, as is ferroptosis. Alim et al. [48], Bersuker et al. [49]
STEAP3 Fe3+ Fe3+ is reduced to Fe2+ by the iron reductase STEAP3. Alim et al. [48]
SLC11A2/DMT1 Fe2+ Transport of Fe2+ into the cytoplasm is involved in ferroptosis. Bersuker et al. [49]
LIP Fe2+ Plays a transit role in the storage, transport and utilization of iron ions. Enriched for Fe2+, which is normally translocated into mitochondria for the synthesis of ferrous heme and iron-sulfur clusters. Doll et al. [50], Stefely and Pagliarini [51]
SLC25, Mitochondrial ferritin Iron in the cytoplasm Translocated into the mitochondrial membrane. Fanet et al.[52], Kraft et al. [53]
PINK1/PRKN/PARK2 SLC25 Degradation of SLC25, limiting iron ion accumulation and inhibiting mitochondrial pathway ferroptosis. Fanet et al. [52], Kraft et al. [53]
SLC40A1 Fe2+ Translocates Fe2+, a process that requires extracellular oxidation of Fe2+ to Fe3+. Soula et al. [54]
CP Iron in SLC40A1 Oxidation of iron in SLC40A1, negatively regulates hepatocyte ferroptosis, and CP deficiency can increase intracellular Fe2+ and lipid peroxidation. Soula et al. [54]
LOX, POR Ferrous ion Iron ions are important cofactors for LOX and POR, both of which play key roles in ferroptosis in ferroptosis.In essence, disruptions in intracellular iron homeostasis, particularly Fe2+ accumulation, can provoke ferroptosis. Lowering intracellular Fe2+ concentrations effectively thwarts ferroptosis. Mao et al. [55]
Lipid metabolism LOX, POR Produces lipid peroxides, and its characteristic diffusion pattern relies on LIP-mediated diffusion of peroxides across the membrane. Ding et al. [56]
ACSL4, LPCAT3 PUFAs ACSL4 catalyzes the biochemical reaction of AA and AdA adrenate with CoA to generate AA/AdA-CoA derivatives esterified to phospholipids. LPCAT3 catalyzes the biosynthesis of AA/AdA-CoA with membrane PE to generate AA/AdA-PEPUFA.PUFA promotes lipid peroxidation. Lee et al. [57], Ma et al. [58]
FSP1 CoQ10 Catalyses the regeneration of CoQ10 using NAD(P)H, reduces CoQ10 to CoQH2. CoQH2 can trap lipid peroxyl radicals, thereby preventing the accumulation of lipid peroxides in the cell membrane and inhibiting ferroptosis. Doll et al. [50]
FAO Fatty acids Consume most of the fatty acids, resulting in a reduced rate of lipid peroxidation. Gao et al. [59]
ACLY CoA Catalyzes the production of citric acid. Kakhlon and Cabantchik [60]
ACSF2, CS Mitochondrial fatty acid metabolism Required for mitochondrial fatty acid metabolism. Knockdown reversed ferroptosis, suggesting a role for mitochondrial fatty acid metabolism in promoting ferroptosis. Hogg et al. [14], Philpott et al. [61]
TOFA PUFA Inhibition of ferroptosis, demonstrating the key role of PUFA as a substrate for ferroptosis lipid peroxidation. Wu et al. [3], Paradkar et al. [62]
MUFAs It is not susceptible to peroxidation and inhibits ferroptosis. Li et al. [63]
ACSL3, SCD1 MUFA Catalyzes MUFA synthesis. Also has an inhibitory effect on ferroptosis. Shang et al. [64]
Oxygen-containing radicals NOX ROS ROS generation primarily depends on mitochondrial metabolism and the activity of NOX located in the cell membrane. Hogg et al. [14]
AMPK Maintains ATP homeostasis and regulates ROS production through protein phosphorylation. Wu et al. [65]
VDAC Controls the exchange of substances between mitochondria and other organelles during oxidative stress. Zou et al. [66]
RSL3 VDAC2 VDAC2 is directly targeted for carbonylation during RSL3-induced ferroptosis. Geng et al. [67]
Erastin VDAC2, VDAC3 Erastin can induce the degradation of VDAC2 and VDAC3 via an E3 ubiquitin ligase-dependent mechanism, thereby promoting ferroptosis. Geng et al. [67]
NOXs Complexes composed of cell membranes and enzymes Transport of electrons through the plasma membrane generates superoxide and other ROS, which contribute to lipid peroxidation in ferroptosis. Kagan et al. [40], Stockwell et al. [68], Doll et al. [69], Liang et al. [70], Tesfay et al. [71]
Degradation system Oxidative stress and lipid peroxidation products A powerful inducer of autophagy, excessive autophagy enhances ferroptosis. [32]
Rapamycin mTOR Leading to the formation of autophagic vesicles. [72,73]
Autophagy effector Inhibition of cancer cell ferroptosis. Holliday [15], Lee et al. [72], Yang et al. [74]
SLC7A11, GPX4 SLC7A11 and GPX4 deletion impairs the autophagic process and resists ferroptosis triggered by Golgi stress, revealing a complex interplay between autophagy, ferroptosis, and organelle stress. Holliday [15], Lee et al. [72], Yang et al. [74]
OTU deubiquitinases, OTUB1 SLC7A11 Stabilizes SLC7A11. Inhibition of OTUB1 enhances SLC7A11 degradation and sensitivity of cancer cells to ferroptosis. Li et al. [75]
ACSL4, VDAC2, VDAC3 Regulation of protein stability at the ferroptosis promoter. Shoshan-Barmatz et al.[76]
RNF113A Plays a key role in DNA damage-induced ferroptosis. deletion of RNF113A stimulates an ferroptosis process that is tightly linked to DNA damage. Yang et al. [77]
LONP1 Mediates mitochondrial degradation, leading to mitochondrial DNA damage and activation of STING1/TMEM173, which triggers an autophagic response that leads to ferroptosis in pancreatic cancer cells. Wu et al. [65]
NEDD4 VDAC2, VDAC3 Inhibition of ferroptosis. Bedard and Krause [78]
NEDD4L LTF Slow down the accumulation of iron in cancer cells and inhibit ferroptosis triggered by lipid peroxidation. Chen et al. [79]

Epigenetic regulation of ferroptosis

Epigenetic modifications encompass histone posttranslational modifications, DNA and RNA methylation, and ncRNA regulation, etc. The roles of these modifications in regulating tumor ferroptosis are outlined below.

Posttranslational modification of histones regulates ferroptosis

The nucleosome is the fundamental unit of chromatin, comprising a segment of DNA and an octamer of four core histones H3, H4, H2A, and H2B. These histones serve as scaffolds that encapsulate and concentrate the DNA, playing a critical role in the compaction and disassembly of chromatin [80,81]. Histone post-translational modifications (PTMs) are a group of multifunctional epigenetic marks that regulate the conformation of chromatin and the accessibility of transcription factors, co-activators and co-inhibitors as well as involve in transcription, DNA damage, apoptosis, and cell cycle regulation [80,82]. The miswriting, misinterpretation and misclearance of histone modifications are closely related to the development of tumors, and the disturbance of histone coding leads to the disorder of gene expression and cell characteristics [83,84].

Epigenetic modifications and PTMs regulate gene expression during transcription and post-transcription respectively, and regulate protein activity, function, and degradation after transcription [85]. Their dysregulation results in abnormal gene and protein expression, contributing to the transformation into malignant phenotypes and facilitating tumor initiation and progression [85–87].

Histone methylation regulates ferroptosis

Histone methylation modifications associated with ferroptosis mainly involve the methylation of H3K4 and H3K9, catalyzed by histone methyltransferase (HMTs) [88]. Research indicates that GPX4 is more abundantly expressed in tumor cells than in normal cells, which correlates with increased levels of H3K4me3 on the GPX4 promoter [89]. Methionine adenosine transferase 2A (MAT2A) promotes the production of methylated donor SAM, which upregulates ACSL3 by increasing the abundance of H3K4me3 on the promoter, thereby inhibiting ferroptosis [90]. JQ1 inhibits HMT expression called G9a, reducing H3K4me3 abundance of BRD4, and finally induces ferroptosis in cancer cells [91]. In breast cancer, mucin 1-C binds to the CD44 variant to stabilize the SLC7A11 molecule, while H3K9me2/3 on the Mucin 1-C promoter suppresses its transcription, thereby affecting GPX4’s capacity to induce ferroptosis [92]. Branched-chain amino acid transaminase-1 (BCAT1) can regulate cellular ferroptosis by modulating H3K9me3. The absence of H3K9me3 at the EGR1 promoter site leads to rapid upregulation of EGR1. EGR1 directly binds to the GPX4 promoter region, thereby inhibiting GPX4 transcription. As a result, mesenchymal stromal cells become more sensitive to ferroptosis, which provides new insights for the treatment of liver diseases [93].

Histone acetylation regulates ferroptosis

Histone acetylation disrupts histone binding to DNA, leading to nucleosome deaggregation and gene transcriptional activation. Histone acetylation depends on Bromodomain-containing protein, BRD family, Histone acetyltransferases (HATs) and Histone deacetylases (HDACs) [94]. The BRD family can recognize acetylation markers, and the BRD4 inhibitor JQ1 can enhance the expression of HDAC called SIRT1, inducing ferroptosis by reducing H3K27ac levels upstream of BRD4 and affecting the recognition of acetylation sites on GPX4 and SLC7A11 genomic proteins [95]. Studies have shown that aberrations in HDAC3 can lead to the suppression of GPX4, thereby driving renal ferroptosis and the progression of acute kidney injury and chronic kidney disease. This inhibitory effect on GPX4 May be related to the action of HDAC3 in combination with Kruppel-like factor 5 [96].

Histone acetylation by HATs is linked to transcriptional activation [97]. NRF2 recruits P300, CBP and P300/CBP associated factors (PCAF) to increase H3K9ac levels of NRF2 and regulate ferroptosis in renal tubulointerstitial fibrosis [98]. The lysine acetyltransferase 5 (KAT5) inhibitor ketamine reduces H3K27ac levels in the GPX4 promoter region, which boosts ferroptosis from breast cancer [99]. Hepatocyte nuclear factor 4 alpha (HNF4A) and HIC ZBTB transcriptional repressor 1 (HIC ZBTB transcriptional repressor 1) in liver cancer HIC1) binds competitively to KAT2B, inhibiting the production of GSH and promoting ferroptosis [100].

Similar to HAT inhibitors, HDACS-mediated histone deacetylation also exhibits transcriptional inhibition, which triggers the induction of ferroptosis by inhibiting EMT markers in cancer cells [101]. The effect of HDAC in the tumor microenvironment is reversed by an inhibitor, and the HDAC inhibitor BEBT-908 acetylates p53 to promote ferroptosis signaling [102]. However, the regulation of ferroptosis by different cells after the same HDAC inhibitor intervention may be opposite [103]. In neurons and cancer cells with a similar mechanism of Erastin induced ferroptosis, Class I HDAC inhibitors enhance the ferroptosis of cancer cells while protecting neurons from ferroptosis [104]. This specific effect may be attributed to variations in the expression of HDAC between cancer cells and primary neurons [105].

Histone ubiquitination regulates ferroptosis

Histone ubiquitination related to ferroptosis in cancer cells, particularly involving Histone 2A ubiquitination (H2Aub) and Histone 2B ubiquitination (H2Bub). Both are associated with SLC7A11 expression [106]. The reduction of the tumor suppressor protein BAP1 and the ubiquitin ligase PRC1 of H2Aub can increase H2Aub uptake of SLC7A11 promoter [107]. The regulation of H2Aub by PRC1 and BAP1 leads to contrasting outcomes, yet both suppress the expression of SLC7A11 [108]. Furthermore, p53 promotes the recruitment of ubiquitin specific peptidase 7 (USP7), resulting in diminished H2Bub levels at the regulatory region of SLC7A11, thus lowering SLC7A11 expression independently of p53 as a transcription factor [106].

Histone modification can regulate cell susceptibility to ferroptosis by affecting the expression of associated genes and metabolic pathways of ferroptosis. However, the influence of histone modification on ferroptosis is context-dependent; different modification patterns may either counteract or synergistically affect iron-dead cells [109]. Further exploration is essential to comprehensively understand how histone modification regulates ferroptosis (Figure 2).

Figure 2.

Figure 2.

The multifaceted role of histone modifications in regulating cellular ferroptosis.

DNA methylation regulates ferroptosis

DNA methylation is a prevalent epigenetic modification in eukaryotic cells, typically utilizing S-adenosyl methionine (SAM) as a methyl donor, predominantly catalyzed by DNA methyltransferases (DNMTs) such as DNMT1, DNMT3A, and DNMT3B. This process is often inversely linked to gene expression levels [110,111]. In the context of ferroptosis regulation, DNA methylation influences lipid metabolism. In mesenchymal gastric cancer cells (GCs), Elongated long-chain fatty acid protein 5 (ELOVL5) and fatty acid desaturase 1 (FADS1) promote unsaturated fatty acid synthesis, but in enteric GC, these genes are suppressed by DNA methylation, rendering the cells resistant to ferroptosis [112]. Lymphospecific helicase (LSH) in lung cancer activates metabolic genes by modifying DNA methylation through WD repeat domain 76 (WDR76) to reduce lipid ROS levels and inhibit ferroptosis [113], where LSH action is antagonized by DDB1- and CUL4-associated factor 8 (DCAF8) [114].

DNA methylation can directly regulate the expression of ferroptosis related molecules such as GPX4 and SLC7A11. Glycine enhances SAM-mediated Gpx4 promoter methylation catalyzed by DNMT1, DNMT3A and DNMT3B to induce ferroptosis [115,116]. The DNMT1 inhibitor 6-thioguanine is an inducer of ferroptosis in gastric cancer, which may be related to its indirect inactivation system Xc−. In addition, DNA methylation involves in the regulation of intercellular interactions in ferroptosis. DNMT1 inhibitor 5-azaCdR decreases cadherin-1 (CDH1) methylation levels, increases e-cadherin expression, and decreases ferroptosis sensitivity in head and neck cancers. Thus, DNA methylation actively regulates ferroptosis in tumor [117]. Notably, oxidative stress and iron metabolism directly affect DNA methylation levels [118]. Chronic iron exposure increases LIP levels in colon cells and promotes ferroptosis while triggering demethylation of NRF2 targets such as NOQ1 and GPX2, avoiding the occurrence of ferroptosis [119,120]. In essence, DNA methylation plays a pivotal role in ferroptosis regulation through various mechanisms, including lipid metabolism and cellular interactions [121] (Figure 3).

Figure 3.

Figure 3.

The key role of DNA methylation in the regulation of ferroptosis.

RNA methylation regulates ferroptosis

RNA methylation stands as a prominent area in epigenetic investigations, constituting over 60% of all RNA modifications, with RNA m6A emerging as the most prevalent mRNA post-transcriptional alteration [122]. The biological roles of RNA m6A modification are guided by ‘readers,’ ‘writers,’ and ‘erasers.’ Writers such as METTL3, METTL14, WTAP, and KIAA1429 primarily orchestrate RNA methylation, while erasers like ALKBH5 and FTO play pivotal roles in m6A demethylation. Readers encompass RNA m6A binding proteins, notably the YTH domain protein family and the HNRNP family, which identify mRNAs featuring m6A markings [123]. Research indicates that the YTH domain of m6A (YTHDC2) can bind to SLC7A11 mRNA, facilitating its degradation [124]. In non-small cell lung cancer, FSP1 (ferroptosis resistance gene) has an mRNA carrying five m6A sites and is upregulated by ῠ3 binding. Notably, in type A AD patients, the METTL3 protein level inversely correlates with FSP1 expression, hindering SLC7A11 expression and fostering ferroptosis in human aortic smooth muscle cells. Conversely, in hepatoblastoma and lung adenocarcinoma, METTL3 bolsters SLC7A11 mRNA stability, impeding ferroptosis [125]. This may depend on how readers express it.

YTHDF1 incentivizes mRNA translation. In liver cancer, YTHDF1 recognizes m6A markers on SLC7A11 mRNA, intensifying ferroptosis inhibition, while in liver stellate cells, YTHDF1 identifies m6A markers on BECN1 mRNA, amplifying ferritin phagocytosis and triggering ferroptosis [126]. In contrast, YTHDF2 induces mRNA degradation. Studies coupling YTHDF2 with METTL14 unveil that YTHDF2-mediated degradation of SLC7A11 mRNA necessitates METTL14-mediated RNA m6A modification in hepatocellular carcinoma [125,127,128]. In lung adenocarcinoma, YTHDC2 targets SLC3A2 and SLC7A11 in an RNA m6A-dependent manner, acting as an endogenous ferroptosis inducer [129].

Erasers also take part in the regulation of ferroptosis. In hypopharyngeal squamous cell carcinoma, ALKBH5 targets RNA m6A residues in the NRF2 transcript 3 ‘UTR to inhibit transcription and promote ferroptosis [130]. The downregulation of ALKBH5 by black phosphorus quantum dots heightens overall RNA m6A levels in lung cells, leading to lipid peroxidation, iron accumulation, and mitochondrial dysfunction. FTO down-regulates SLC7A11 expression and induces ferroptosis in thyroid carcinomates [131]. Presently, RNA m6A modification research primarily focuses on readers and writers, with less attention on erasers, warranting further exploration by the academic community. Moreover, RNA modifications extend beyond m6A labeling, with common methods like 2’-O-methylation (Nm) and pseudouracil (ψ) holding promise as future research focal points (Figure 4).

Figure 4.

Figure 4.

RNA methylation and its role in the regulation of ferroptosis.

Non-coding RNA regulates ferroptosis

In recent years, an increasing number of Noncoding RNAs (ncRNAs) with biological functions have been discovered, which can be divided into constitutive ncRNAs and regulatory ncRNAs. Constitutive ncRNAs, reminiscent of housekeeping genes, play roles in translation and splicing, including ribosomal RNA (rRNA), transfer RNA (tRNA), and small nuclear RNA (snRNA). In contrast, regulatory ncRNAs are primarily involved in transcription and post-transcriptional modifications.

MicroRNAs (miRNAs) are a class of non-coding single-stranded RNA molecules with a length of about 22 nucleotides, encoded by endogenous genes, regulating gene expression at the mRNA level [132]. They participate in epigenetic regulation mainly by binding to the target sequence 3 ‘UTR to inhibit mRNA translation or promote mRNA degradation [133]. In the classical signaling pathway, the down-regulation of SLC7A11 expression by miR-5096, miR-375 and miR-378a-3p induces ferroptosis, while the inhibition of GPX4 expression by miR-15a-5p, miR-324-3p, miR-182-5p and miR-541-3p promotes ferroptosis [134–139]. Studies have shown that miR-302a-3p and miR-335 target transferrin and ferritin respectively and promote ferroptosis by regulating iron metabolism [140]; miR-30e-5p targets specific protein 1 (SP1) and inhibits AMPK pathway to induce cell ferroptosis; miR-214-3p promotes ferroptosis by targeting ATF4. ATF4 is a key gene in ER stress [141]. Endoplasmic reticulum stress plays a dual role in ferroptosis, with ATF4 inhibiting ferroptosis by upregulating SLC7A11 in human glioma cells, while ATF4 upregulating ChaC glutathione-specific glutamylcyclotransferase 1 (CHAC1) expression in breast cancer, promoting ferroptosis induced by cystine starvation [142]. Studies have suggested that miRNA regulating genes related to ferroptosis not only acts on one gene: miR-7-5p can simultaneously up-regulate the expression of ferritin, down-regulate the expression of ALOX12, and coordinate to reduce the level of LiperFluo to inhibit ferroptosis. miRNAs may also coordinate with other epigenetic mechanisms to dynamically regulate cellular ferroptosis.

Long noncoding RNAs (lncRNAs) are ncRNAs with a length greater than 200 nucleotides [143]. Its abnormal expression is closely related to tumors, degenerative diseases, ischemic injury, etc., and is widely involved in the regulation of ferroptosis [144,145]. LncRNAs predominantly regulate ferroptosis through post-transcriptional processes. For instance, lncRNA PVT1 regulates SLC7A11 by activating p53 expression via miR-214 [146]. OIP5-AS1 and SLC16A1-AS1 inhibit ferroptosis by targeting miR-128-3p and up-regulating SLC7A11 expression [147]. lncRNA ZFAS1 inhibits the expression of miR-150-5p and targets the glutamine absorption metabolic regulator SLC38A1 to induce ferroptosis [148]. LncRNAs NEAT1 and PR11–89 modulate cellular iron concentrations to regulate ferroptosis; the former increases and the latter decreases iron levels, with NEAT1 upregulating TFR and GOT1 via miR-9-5p, while PR11–89 upregulates PROM2 via miR-129-5p [149]. LncRNA MT1DP targets miR-365a-3p to downregulate NRF2 expression, increasing cellular sensitivity to ferroptosis [150].

lncRNA can also bind to mRNA to regulate the translation process. For example, lncRNA GABPB1-AS1 directly inhibits GABPB1 mRNA translation, down-regulating peroxidoredoxin 5 expression to induce ferroptosis [151]. Some lncRNAs regulate mRNA translation through lncRNA-protein complexes. LncRNA 00925 binds to Pumilio RNA binding family member 2 (Pum2) protein, resulting in Prdx6mRNA degradation, while lncRNA ASMTcl-AS1 recruits U2AF2, stabilizing SAT1 mRNA structure to induce ferroptosis. Beyond post-transcriptional regulation, lncRNAs also modulate gene transcription directly or indirectly [152]. lncRNA Meg3 binds directly to p53 to induce ferroptosis through the p53-GPX4 axis, while cytoplasmic lncRNA P53RRA collaborates with rasGTpase activating protein-binding protein 1 (GABP1) to activate p53 gene [153]. lncRNA LINC00618 promotes ferroptosis in an apoptosis-dependent manner, inhibiting SLC7A11 expression by attenuating LSH, so as to induce the transcription of SLC7A11 after recruitment to the SLC7A11 promoter.

Circular noncoding RNAs (circRNAs) are continuous rings of single-stranded RNA without A 5 ‘cap and 3’ end poly (A) tail, which eliminate inhibition of target genes by binding miRNA [154]. They are the focus of research in the field of ncRNA [155]. Depending on the targeted miRNA, the regulations of circRNA on ferroptosis vary. Several tumor-associated circRNAs influence ferroptosis through a molecular regulatory network involving circular RNA/miRNA interactions [156]. CircRHOT1, circ -0,008,035, circRNA1615 and circPSEN1 pass miR-106a-5/STAT3, miR-599/EIF4A1, miR152/LPR6 and miR-200b-3p/cofilin-2 axis respectively to inhibit ferroptosis [157]. In lipid metabolism, circPtpn14 inhibits ferroptosis by targeting miR-351-5p. circDTL, circIL4R, circKIF4A and cicr0000309 bind to inhibit miR-1287-5p, miR-541-3p, miR-1231 and miR-188-3p respectively to increase GPX4 expression [158]. A single circular RNA can carry multiple miRNA binding sites, for circEPSTI1 inhibits ferroptosis by simultaneously binding miR-375, miR-409-3p, and miR-51 (Figure 5).

Figure 5.

Figure 5.

The role of noncoding RNAs in the regulation of ferroptosis.

The application prospect of targeted ferroptosis epigenetic regulators in tumor therapy

Epigenetic alterations in cancer are reversible, presenting an appealing avenue for cancer therapy. Targeting ferroptosis through epigenetic modulation emerges as a promising strategy in cancer treatment, with relevant research integrated into anti-cancer therapeutics.

The BRD4 inhibitor JQ1 can trigger ferroptosis in apoptosis-resistant cancer cells, augmenting the anticancer efficacy with ferroptosis inducers. Mechanistically, JQ1 suppresses the expression of histone methyltransferase G9a by inhibiting BRD4, downregulating key ferroptosis-associated genes SLC7A11, SLC3A2, and GPX4, while enhancing histone deacetylase SIRT1 expression. In an IVDD mouse model, homocysteine, involved in homomethylation, boosts GPX4 methylation, instigating ferroptosis in nucleus pulposus cells [159]. The class I HDAC inhibitor vorinostat can enhance ferroptosis in small cell lung cancer by regulating histone or non-histone acetylation [160]. Controlling m6A-based transcriptomics can destroy the redox balance and induce ferroptosis in leukemia cells. GNPa-CSP12 functionalized gold nanorods can eliminate endogenous Fe2-dependent m6A demethylase activity, and its down-regulation is closely related to glycolysis, hypoxia and immune checkpoint pathway-related genes [161]. Tiliroside is a potential natural anti-cancer product that can target TBK1 to reduce the phosphorylation level of serine 349 on sequestosome-1 (p62), thereby decreasing the affinity of p62 for Kelch-like ECH-associated protein 1 (Keap1) and promoting Keap1-mediated ubiquitination and degradation of NRF2, which induces ferroptosis in hepatocellular carcinoma [162]. Corosolic acid can reduce the ubiquitination level of the E3 ubiquitin ligase MDM2 associated with GSS through HERPUD1, thereby promoting the ubiquitination of GSS and inhibiting GSH synthesis, which in turn enhances the sensitivity of liver cancer cells to ferroptosis [163]. QD394 is a quinazolinedione ROS inducer which can induce ferroptosis by inhibiting STAT3 phosphorylation, thereby inhibiting GPX4 expression in pancreatic cancer [164]. Eriodictyol induces ferroptosis by downregulating NRF2 phosphorylation, thereby decreasing the protein levels of SLC7A11 and GPX4 in ovarian cancer [165]. The nexus of epigenetic drug-mediated ferroptosis regulation with conventional chemotherapy, targeted therapy, immunotherapy, and radiotherapy presents substantial research potential and application prospects in cancer care (Table 3). However, the application of epigenetic regulation of ferroptosis in cancer treatment still requires a great deal of further research. The safety of regulating ferroptosis to achieve cancer treatment effects needs further investigation, as ferroptosis occurs in both normal and tumor cells [2]. Moreover, whether epigenetic mechanisms affect multiple ferroptosis-related genes and how these different epigenetic mechanisms interact with diverse signaling pathways to determine the cellular response to ferroptosis stimuli remain unclear. Further research is needed to elucidate these questions [167].

Table 3.

Targeting ferroptosis epigenetic regulator drugs in tumor therapy.

Name of drug Target of action Mechanism of action Types of diseases addressed References
JQ1 BRD4 Reduced G9a expression by inhibiting BRD4, down-regulated the expression of ferroptosis-related genes SLC7A11, SLC3A2 and GPX4, and enhanced the expression of SIRT1. Cancer Alborzinia et al.[166]
Homocysteine GPX4 Enhanced GPX4 methylation leads to ferroptosis in myeloid cells. Cancer Cai et al. [144]
Vorinostat HDAC Regulation of histone or non-histone acetylation enhances ferroptosis in small cell lung cancer. Small-cell lung cancer Yao et al. [145]
Gold nanorod m6A Elimination of endogenous Fe2+-dependent m6A demethylase activity, which is downregulated in close association with genes related to glycolysis, hypoxia, and the immune checkpoint pathway. Leukaemia Lu et al. [146]
Tiliroside TBK1 Targeted TBK1 to induce ferroptosis in cancer cells. Hepatocellular carcinoma Yang et al. [162]
Corosolic acid HERPUD1 Decreased the cellular GSH level and caused liver cancer cells to become more sensitive to ferroptosis. Hepatocellular carcinoma Peng et al. [163]
QD394 STAT3 Inhibited STAT3 phosphorylation, thereby inhibited GPX4 expression. Pancreatic cancer Hu et al. [164]
Eriodictyol NRF2 Downregulated NRF2 phosphorylation, thereby decreased the protein levels of SLC7A11 and GPX4. Ovarian cancer Wang et al. [165]

Conclusion and prospect

Epigenetics intricately influences ferroptosis in tumor cells, modulating ferroptosis-related genes and pathways without altering tumor gene expression levels. Current investigations on the epigenetic control of ferroptosis in tumor cells are extensive. Given the reversible nature of epigenetics, its modulation of tumor cell ferroptosis stands as a promising therapeutic target with bright application potential. However, we still need more research to confirm the efficacy of epigenetic drugs in ferroptosis regulation therapy. Hence, further exploration of the interplay between diverse epigenetic regulations and tumor ferroptosis, alongside the development of tumor-targeted drugs for distinct targets, in conjunction with conventional treatments, offers novel insights for cancer treatment.

Funding Statement

This research was supported by the National Natural Science Foundation of China [No. 82303133]; China Postdoctoral Science Foundation [No. 2022M723559]; Natural Science Foundation of Hunan Province [No. 2021JJ40951]; Health Research project of Hunan Provincial Health Commission [No. B202307017799, No. B202309018525].

Authors’ contributions

YX, SY and YQ designed this study. YX, MH, XZ, MY, ZY, SY and YQ reviewed and revised the manuscript. YX, MY and ZY drafted the original manuscript. YX, MH, XZ and YQ designed and completed the figures and tables. All authors approved the final manuscript submission and agreed to be responsible for all aspects of the work.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Availability of data and materials

No data was used for the research described in the review.

Consent for publication

All authors agree to publish this review in your esteemed journal.

References

  • [1].Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–20. doi: 10.1016/j.cell.2012.03.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171(2):273–285. doi: 10.1016/j.cell.2017.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Wu Y, Zhang S, Gong X, et al. The epigenetic regulators and metabolic changes in ferroptosis-associated cancer progression. Mol Cancer. 2020;19(1):39. doi: 10.1186/s12943-020-01157-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Jiang X, Stockwell BR, Conrad M.. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22(4):266–282. doi: 10.1038/s41580-020-00324-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Lei G, Zhuang L, Gan B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 2022;22(7):381–396. doi: 10.1038/s41568-022-00459-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Xu G, Wang H, Li X, et al. Recent progress on targeting ferroptosis for cancer therapy. Biochem Pharmacol. 2021;190:114584. doi: 10.1016/j.bcp.2021.114584 [DOI] [PubMed] [Google Scholar]
  • [7].Xu T, Ding W, Ji X, et al. Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med. 2019;23:4900–4912. doi: 10.1111/jcmm.14511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Tang D, Chen X, Kang R, et al. Ferroptosis: molecular mechanisms and health implications. Cell Res. 2021;31(2):107–125. doi: 10.1038/s41422-020-00441-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Zhang Y, Li M, Guo Y, et al. The organelle-specific regulations and epigenetic regulators in ferroptosis. Front Pharmacol. 2022;13:905501. doi: 10.3389/fphar.2022.905501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Tang D, Kroemer G. Ferroptosis. Curr Biol. 2020;30(21):R1292–r1297. doi: 10.1016/j.cub.2020.09.068 [DOI] [PubMed] [Google Scholar]
  • [11].Xu Y, Xing Z, Abdalla Ibrahim Suliman R, et al. Ferroptosis in liver cancer: a key role of post-translational modifications. Front Immunol. 2024;15:1375589. doi: 10.3389/fimmu.2024.1375589 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Wang Y, Yan D, Liu J, et al. Protein modification and degradation in ferroptosis. Redox Biol. 2024;75:103259. doi: 10.1016/j.redox.2024.103259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Wu YL, Lin ZJ, Li CC, et al. Epigenetic regulation in metabolic diseases: mechanisms and advances in clinical study. Signal Transduct Target Ther. 2023;8(1):98. doi: 10.1038/s41392-023-01333-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Hogg SJ, Beavis PA, Dawson MA, et al. Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov. 2020;19(11):776–800. doi: 10.1038/s41573-020-0077-5 [DOI] [PubMed] [Google Scholar]
  • [15].Holliday R. The inheritance of epigenetic defects. Science. 1987;238(4824):163–170. doi: 10.1126/science.3310230 [DOI] [PubMed] [Google Scholar]
  • [16].Biswas S, Rao CM. Epigenetic tools (The writers, the readers and the erasers) and their implications in cancer therapy. Eur J Pharmacol. 2018;837:8–24. doi: 10.1016/j.ejphar.2018.08.021 [DOI] [PubMed] [Google Scholar]
  • [17].Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12:31–46. doi: 10.1158/2159-8290.Cd-21-1059 [DOI] [PubMed] [Google Scholar]
  • [18].Van Heerden J, Van Heerden A. Attempted treatment of canine ehrlichiosis with imidocarb dipropionate. J S Afr Vet Assoc. 1981;52(3):173–175. [PubMed] [Google Scholar]
  • [19].Luengo-Fernandez R, Leal J, Gray A, et al. Economic burden of cancer across the European union: a population-based cost analysis. Lancet Oncol. 2013;14:1165–1174. doi: 10.1016/s1470-2045(13)70442-x [DOI] [PubMed] [Google Scholar]
  • [20].Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
  • [21].Kaur R, Bhardwaj A, Gupta S. Cancer treatment therapies: traditional to modern approaches to combat cancers. Mol Biol Rep. 2023;50(11):9663–9676. doi: 10.1007/s11033-023-08809-3 [DOI] [PubMed] [Google Scholar]
  • [22].Dolma S, Lessnick SL, Hahn WC, et al. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3(3):285–296. doi: 10.1016/s1535-6108(03)00050-3 [DOI] [PubMed] [Google Scholar]
  • [23].Gao M, Yi J, Zhu J, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73(2):354–363.e353. doi: 10.1016/j.molcel.2018.10.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Hou W, Xie Y, Song X, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12(8):1425–1428. doi: 10.1080/15548627.2016.1187366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Riegman M, Sagie L, Galed C, et al. Ferroptosis occurs through an osmotic mechanism and propagates independently of cell rupture. Nat Cell Biol. 2020;22(9):1042–1048. doi: 10.1038/s41556-020-0565-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Wang H, Liu C, Zhao Y, et al. Mitochondria regulation in ferroptosis. Eur J Cell Biol. 2020;99(1):151058. doi: 10.1016/j.ejcb.2019.151058 [DOI] [PubMed] [Google Scholar]
  • [27].Liu Y, Lu S, Wu LL, et al. The diversified role of mitochondria in ferroptosis in cancer. Cell Death Dis. 2023;14(8):519. doi: 10.1038/s41419-023-06045-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Li Q, Han X, Lan X, et al. Inhibition of neuronal ferroptosis protects hemorrhagic brain. JCI Insight. 2017;2(7):e90777. doi: 10.1172/jci.insight.90777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417:1–13. doi: 10.1042/bj20081386 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Forcina GC, Dixon SJ. GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 2019;19(18):e1800311. doi: 10.1002/pmic.201800311 [DOI] [PubMed] [Google Scholar]
  • [31].Eaton JK, Furst L, Ruberto RA, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506. doi: 10.1038/s41589-020-0501-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Gaschler MM, Stockwell BR. Lipid peroxidation in cell death. Biochem Biophys Res Commun. 2017;482(3):419–425. doi: 10.1016/j.bbrc.2016.10.086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Yang WS, Stockwell BR. Ferroptosis: death by Lipid Peroxidation. Trends Cell Biol. 2016;26:165–176. doi: 10.1016/j.tcb.2015.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Chen X, Kang R, Kroemer G, et al. Ferroptosis in infection, inflammation, and immunity. J Exp Med. 2021;218(6). doi: 10.1084/jem.20210518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Wang W, Green M, Choi JE, et al. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569(7755):270–274. doi: 10.1038/s41586-019-1170-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Ma X, Xiao L, Liu L, et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 2021;33(5):1001–1012.e1005. doi: 10.1016/j.cmet.2021.02.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Yatim N, Jusforgues-Saklani H, Orozco S, et al. RIPK1 and NF-κB signaling in dying cells determines cross-priming of CD8+ T cells. Science. 2015;350(6258):328–334. doi: 10.1126/science.aad0395 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Lang X, Green MD, Wang W, et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 2019;9:1673–1685. doi: 10.1158/2159-8290.Cd-19-0338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Gaschler MM, Hu F, Feng H, et al. Determination of the subcellular localization and mechanism of action of ferrostatins in suppressing ferroptosis. ACS Chem Biol. 2018;13(4):1013–1020. doi: 10.1021/acschembio.8b00199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Kagan VE, Mao G, Qu F, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13(1):81–90. doi: 10.1038/nchembio.2238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 2008;15(3):234–245. doi: 10.1016/j.chembiol.2008.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Weïwer M, Bittker JA, Lewis TA, et al. Development of small-molecule probes that selectively kill cells induced to express mutant RAS. Bioorg Med Chem Lett. 2012;22(4):1822–1826. doi: 10.1016/j.bmcl.2011.09.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Shimada K, Skouta R, Kaplan A, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12(7):497–503. doi: 10.1038/nchembio.2079 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Abrams RP, Carroll WL, Woerpel KA. Five-membered ring peroxide selectively initiates ferroptosis in cancer cells. ACS Chem Biol. 2016;11(5):1305–1312. doi: 10.1021/acschembio.5b00900 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Gaschler MM, Andia AA, Liu H, et al. FINO(2) initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat Chem Biol. 2018;14(5):507–515. doi: 10.1038/s41589-018-0031-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Battaglia AM, Sacco A, Vecchio E, et al. Iron affects the sphere-forming ability of ovarian cancer cells in non-adherent culture conditions. Front Cell Dev Biol. 2023;11:1272667. doi: 10.3389/fcell.2023.1272667 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Friedmann Angeli JP, Conrad M. Selenium and GPX4, a vital symbiosis. Free Radic Biol Med. 2018;127:153–159. doi: 10.1016/j.freeradbiomed.2018.03.001 [DOI] [PubMed] [Google Scholar]
  • [48].Alim I, Caulfield JT, Chen Y, et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell. 2019;177(5):1262–1279.e1225. doi: 10.1016/j.cell.2019.03.032 [DOI] [PubMed] [Google Scholar]
  • [49].Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575(7784):688–692. doi: 10.1038/s41586-019-1705-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693–698. doi: 10.1038/s41586-019-1707-0 [DOI] [PubMed] [Google Scholar]
  • [51].Stefely JA, Pagliarini DJ. Biochemistry of mitochondrial coenzyme Q biosynthesis. Trends Biochem Sci. 2017;42(10):824–843. doi: 10.1016/j.tibs.2017.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Fanet H, Capuron L, Castanon N, et al. Tetrahydrobioterin (BH4) pathway: from metabolism to neuropsychiatry. Curr Neuropharmacol. 2021;19(5):591–609. doi: 10.2174/1570159x18666200729103529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Kraft VAN, Bezjian CT, Pfeiffer S, et al. GTP cyclohydrolase 1/Tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 2020;6(1):41–53. doi: 10.1021/acscentsci.9b01063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Soula M, Weber RA, Zilka O, et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol. 2020;16(12):1351–1360. doi: 10.1038/s41589-020-0613-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Mao C, Liu X, Zhang Y, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593(7860):586–590. doi: 10.1038/s41586-021-03539-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Ding CC, Rose J, Sun T, et al. MESH1 is a cytosolic NADPH phosphatase that regulates ferroptosis. Nat Metab. 2020;2(3):270–277. doi: 10.1038/s42255-020-0181-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Lee S, Hwang N, Seok BG, et al. Autophagy mediates an amplification loop during ferroptosis. Cell Death Dis. 2023;14(7):464. doi: 10.1038/s41419-023-05978-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Ma S, Henson ES, Chen Y, et al. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7(7):e2307. doi: 10.1038/cddis.2016.208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Gao M, Monian P, Quadri N, et al. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59(2):298–308. doi: 10.1016/j.molcel.2015.06.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Kakhlon O, Cabantchik ZI. The labile iron pool: characterization, measurement, and participation in cellular processes1 1This article is part of a series of reviews on “Iron and cellular redox status.” the full list of papers may be found on the homepage of the journal. Free Radic Biol Med. 2002;33(8):1037–1046. doi: 10.1016/s0891-5849(02)01006-7 [DOI] [PubMed] [Google Scholar]
  • [61].Philpott CC, Protchenko O, Wang Y, et al. Iron-tracking strategies: chaperones capture iron in the cytosolic labile iron pool. Front Mol Biosci. 2023;10:1127690. doi: 10.3389/fmolb.2023.1127690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Paradkar PN, Zumbrennen KB, Paw BH, et al. Regulation of mitochondrial iron import through differential turnover of mitoferrin 1 and mitoferrin 2. Mol Cell Biol. 2009;29(4):1007–1016. doi: 10.1128/mcb.01685-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Li C, Zhang Y, Cheng X, et al. PINK1 and PARK2 suppress pancreatic tumorigenesis through control of mitochondrial iron-mediated immunometabolism. Dev Cell. 2018;46(4):441–455.e448. doi: 10.1016/j.devcel.2018.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Shang Y, Luo M, Yao F, et al. Ceruloplasmin suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma cells. Cell Signal. 2020;72:109633. doi: 10.1016/j.cellsig.2020.109633 [DOI] [PubMed] [Google Scholar]
  • [65].Wu H, Wang F, Ta N, et al. The multifaceted regulation of mitochondria in ferroptosis. Life (Basel). 2021;11(3):222. doi: 10.3390/life11030222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Zou Y, Li H, Graham ET, et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 2020;16(3):302–309. doi: 10.1038/s41589-020-0472-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Geng N, Shi BJ, Li SL, et al. Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharmacol Sci. 2018;22(12):3826–3836. doi: 10.26355/eurrev_201806_15267 [DOI] [PubMed] [Google Scholar]
  • [68].Stockwell BR, Jiang X, Gu W. Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol. 2020;30(6):478–490. doi: 10.1016/j.tcb.2020.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13(1):91–98. doi: 10.1038/nchembio.2239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Liang D, Minikes AM, Jiang X. Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell. 2022;82(12):2215–2227. doi: 10.1016/j.molcel.2022.03.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Tesfay L, Paul BT, Konstorum A, et al. Stearoyl-CoA desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer Res. 2019;79:5355–5366. doi: 10.1158/0008-5472.Can-19-0369 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Lee H, Zandkarimi F, Zhang Y, et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol. 2020;22(2):225–234. doi: 10.1038/s41556-020-0461-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Yang WS, Kim KJ, Gaschler MM, et al. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U S A. 2016;113:E4966–4975. doi: 10.1073/pnas.1603244113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Yang Y, Zhu T, Wang X, et al. ACSL3 and ACSL4, distinct roles in ferroptosis and cancers. Cancers (Basel). 2022;14(23):5896. doi: 10.3390/cancers14235896 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Li C, Zhang Y, Liu J, et al. Mitochondrial DNA stress triggers autophagy-dependent ferroptotic death. Autophagy. 2021;17(4):948–960. doi: 10.1080/15548627.2020.1739447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Shoshan-Barmatz V, De Pinto V, Zweckstetter M, et al. VDAC, a multi-functional mitochondrial protein regulating cell life and death. Mol Aspects Med. 2010;31(3):227–285. doi: 10.1016/j.mam.2010.03.002 [DOI] [PubMed] [Google Scholar]
  • [77].Yang Y, Luo M, Zhang K, et al. Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nat Commun. 2020;11(1):433. doi: 10.1038/s41467-020-14324-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87:245–313. doi: 10.1152/physrev.00044.2005 [DOI] [PubMed] [Google Scholar]
  • [79].Chen X, Huang J, Yu C, et al. A noncanonical function of EIF4E limits ALDH1B1 activity and increases susceptibility to ferroptosis. Nat Commun. 2022;13(1):6318. doi: 10.1038/s41467-022-34096-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Zhao S, Allis CD, Wang GG. The language of chromatin modification in human cancers. Nat Rev Cancer. 2021;21(7):413–430. doi: 10.1038/s41568-021-00357-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Zhang Y, Zhang Q, Zhang Y, et al. The role of histone modification in DNA replication-coupled nucleosome assembly and cancer. Int J Mol Sci. 2023;24(5):4939. doi: 10.3390/ijms24054939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Davalos V, Esteller M. Cancer epigenetics in clinical practice. CA Cancer J Clin. 2023;73(4):376–424. doi: 10.3322/caac.21765 [DOI] [PubMed] [Google Scholar]
  • [83].Audia JE, Campbell RM. Histone modifications and cancer. Cold Spring Harb Perspect Biol. 2016;8(4):a019521. doi: 10.1101/cshperspect.a019521 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Sawan C, Herceg Z. Histone modifications and cancer. Adv Genet. 2010;70:57–85. doi: 10.1016/b978-0-12-380866-0.60003-4 [DOI] [PubMed] [Google Scholar]
  • [85].Shu F, Xiao H, Li QN, et al. Epigenetic and post-translational modifications in autophagy: biological functions and therapeutic targets. Signal Transduct Target Ther. 2023;8(1):32. doi: 10.1038/s41392-022-01300-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Garcia-Martinez L, Zhang Y, Nakata Y, et al. Epigenetic mechanisms in breast cancer therapy and resistance. Nat Commun. 2021;12(1):1786. doi: 10.1038/s41467-021-22024-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell Metab. 2019;29:1028–1044. doi: 10.1016/j.cmet.2019.03.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Li R, Wei X, Jiang DS. Protein methylation functions as the posttranslational modification switch to regulate autophagy. Cell Mol Life Sci. 2019;76:3711–3722. doi: 10.1007/s00018-019-03161-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Zhang X, Sui S, Wang L, et al. Inhibition of tumor propellant glutathione peroxidase 4 induces ferroptosis in cancer cells and enhances anticancer effect of cisplatin. J Cell Physiol. 2020;235:3425–3437. doi: 10.1002/jcp.29232 [DOI] [PubMed] [Google Scholar]
  • [90].Ma M, Kong P, Huang Y, et al. Activation of MAT2A-ACSL3 pathway protects cells from ferroptosis in gastric cancer. Free Radic Biol Med. 2022;181:288–299. doi: 10.1016/j.freeradbiomed.2022.02.015 [DOI] [PubMed] [Google Scholar]
  • [91].Sui S, Zhang J, Xu S, et al. Ferritinophagy is required for the induction of ferroptosis by the bromodomain protein BRD4 inhibitor (+)-JQ1 in cancer cells. Cell Death Dis. 2019;10(5):331. doi: 10.1038/s41419-019-1564-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Hasegawa M, Takahashi H, Rajabi H, et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget. 2016;7(11):11756–11769. doi: 10.18632/oncotarget.7598 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Hu G, Cui Z, Chen X, et al. Suppressing mesenchymal stromal cell ferroptosis via targeting a metabolism-epigenetics axis corrects their poor retention and insufficient healing benefits in the injured liver milieu. Adv Sci (Weinh). 2023;10:e2206439. doi: 10.1002/advs.202206439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Sabari BR, Zhang D, Allis CD, et al. Metabolic regulation of gene expression through histone acylations. Nat Rev Mol Cell Biol. 2017;18(2):90–101. doi: 10.1038/nrm.2016.140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Guo X, Fang ZM, Wei X, et al. HDAC6 is associated with the formation of aortic dissection in human. Mol Med. 2019;25(1):10. doi: 10.1186/s10020-019-0080-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Zhang L, Chen F, Dong J, et al. HDAC3 aberration-incurred GPX4 suppression drives renal ferroptosis and AKI-CKD progression. Redox Biol. 2023;68:102939. doi: 10.1016/j.redox.2023.102939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Zhang Z, Guo M, Shen M, et al. The BRD7-P53-SLC25A28 axis regulates ferroptosis in hepatic stellate cells. Redox Biol. 2020;36:101619. doi: 10.1016/j.redox.2020.101619 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Chung S, Kim S, Son M, et al. Inhibition of p300/CBP-Associated factor attenuates renal tubulointerstitial fibrosis through modulation of NF-kB and Nrf2. Int J Mol Sci. 2019;20(7):1554. doi: 10.3390/ijms20071554 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Li H, Liu W, Zhang X, et al. Ketamine suppresses proliferation and induces ferroptosis and apoptosis of breast cancer cells by targeting KAT5/GPX4 axis. Biochem Biophys Res Commun. 2021;585:111–116. doi: 10.1016/j.bbrc.2021.11.029 [DOI] [PubMed] [Google Scholar]
  • [100].Zhang X, Du L, Qiao Y, et al. Ferroptosis is governed by differential regulation of transcription in liver cancer. Redox Biol. 2019;24:101211. doi: 10.1016/j.redox.2019.101211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Liu L, Li Y, Cao D, et al. SIRT3 inhibits gallbladder cancer by induction of AKT-dependent ferroptosis and blockade of epithelial-mesenchymal transition. Cancer Lett. 2021;510:93–104. doi: 10.1016/j.canlet.2021.04.007 [DOI] [PubMed] [Google Scholar]
  • [102].Fan F, Liu P, Bao R, et al. A dual PI3K/HDAC inhibitor induces immunogenic ferroptosis to potentiate cancer immune checkpoint therapy. Cancer Res. 2021;81:6233–6245. doi: 10.1158/0008-5472.Can-21-1547 [DOI] [PubMed] [Google Scholar]
  • [103].Wang X, Liu K, Gong H, et al. Death by histone deacetylase inhibitor quisinostat in tongue squamous cell carcinoma via apoptosis, pyroptosis, and ferroptosis. Toxicol Appl Pharmacol. 2021;410:115363. doi: 10.1016/j.taap.2020.115363 [DOI] [PubMed] [Google Scholar]
  • [104].Zhang T, Sun B, Zhong C, et al. Targeting histone deacetylase enhances the therapeutic effect of erastin-induced ferroptosis in EGFR-activating mutant lung adenocarcinoma. Transl Lung Cancer Res. 2021;10(4):1857–1872. doi: 10.21037/tlcr-21-303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Zille M, Kumar A, Kundu N, et al. Ferroptosis in neurons and cancer cells is similar but differentially regulated by histone deacetylase inhibitors. eNeuro. 2019;6(1). doi: 10.1523/eneuro.0263-18.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Wang Y, Yang L, Zhang X, et al. Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. EMBO Rep. 2019;20:e47563. doi: 10.15252/embr.201847563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Zhang Y, Shi J, Liu X, et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat Cell Biol. 2018;20(10):1181–1192. doi: 10.1038/s41556-018-0178-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Zhang Y, Koppula P, Gan B. Regulation of H2A ubiquitination and SLC7A11 expression by BAP1 and PRC1. Cell Cycle. 2019;18(8):773–783. doi: 10.1080/15384101.2019.1597506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Tang J, Long G, Xiao L, et al. USP8 positively regulates hepatocellular carcinoma tumorigenesis and confers ferroptosis resistance through β-catenin stabilization. Cell Death Dis. 2023;14(6):360. doi: 10.1038/s41419-023-05747-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Zhang J, Yang C, Wu C, et al. DNA methyltransferases in cancer: biology, paradox, aberrations, and targeted therapy. Cancers (Basel). 2020;12(8):2123. doi: 10.3390/cancers12082123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Yi X, Jiang X, Li X, et al. Histone lysine methylation and congenital heart disease: from bench to bedside (Review). Int J Mol Med. 2017;40:953–964. doi: 10.3892/ijmm.2017.3115 [DOI] [PubMed] [Google Scholar]
  • [112].Lee JY, Nam M, Son HY, et al. Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proc Natl Acad Sci USA. 2020;117:32433–32442. doi: 10.1073/pnas.2006828117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Jiang Y, Mao C, Yang R, et al. EGLN1/c-myc induced lymphoid-specific helicase inhibits ferroptosis through lipid metabolic gene expression changes. Theranostics. 2017;7(13):3293–3305. doi: 10.7150/thno.19988 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Huang D, Li Q, Sun X, et al. CRL4(DCAF8) dependent opposing stability control over the chromatin remodeler LSH orchestrates epigenetic dynamics in ferroptosis. Cell Death Differ. 2021;28(5):1593–1609. doi: 10.1038/s41418-020-00689-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Ling H, Li M, Yang C, et al. Glycine increased ferroptosis via SAM-mediated GPX4 promoter methylation in rheumatoid arthritis. Rheumatology (Oxford). 2022;61(11):4521–4534. doi: 10.1093/rheumatology/keac069 [DOI] [PubMed] [Google Scholar]
  • [116].Zhang J, Gao M, Niu Y, et al. From DNMT1 degrader to ferroptosis promoter: drug repositioning of 6-thioguanine as a ferroptosis inducer in gastric cancer. Biochem Biophys Res Commun. 2022;603:75–81. doi: 10.1016/j.bbrc.2022.03.026 [DOI] [PubMed] [Google Scholar]
  • [117].Pogribny IP, Tryndyak VP, Pogribna M, et al. Modulation of intracellular iron metabolism by iron chelation affects chromatin remodeling proteins and corresponding epigenetic modifications in breast cancer cells and increases their sensitivity to chemotherapeutic agents. Int J Oncol. 2013;42:1822–1832. doi: 10.3892/ijo.2013.1855 [DOI] [PubMed] [Google Scholar]
  • [118].Cao LL, Liu H, Yue Z, et al. Iron chelation inhibits cancer cell growth and modulates global histone methylation status in colorectal cancer. Biometals. 2018;31(5):797–805. doi: 10.1007/s10534-018-0123-5 [DOI] [PubMed] [Google Scholar]
  • [119].Kawai K, Li YS, Song MF, et al. DNA methylation by dimethyl sulfoxide and methionine sulfoxide triggered by hydroxyl radical and implications for epigenetic modifications. Bioorg Med Chem Lett. 2010;20:260–265. doi: 10.1016/j.bmcl.2009.10.124 [DOI] [PubMed] [Google Scholar]
  • [120].Horniblow RD, Pathak P, Balacco DL, et al. Iron-mediated epigenetic activation of NRF2 targets. J Nutr Biochem. 2022;101:108929. doi: 10.1016/j.jnutbio.2021.108929 [DOI] [PubMed] [Google Scholar]
  • [121].Logie E, Van Puyvelde B, Cuypers B, et al. Ferroptosis induction in multiple myeloma cells triggers DNA methylation and histone modification changes associated with cellular senescence. Int J Mol Sci. 2021;22(22):12234. doi: 10.3390/ijms222212234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Niu Y, Zhao X, Wu YS, et al. N6-methyl-adenosine (m6A) in RNA: an old modification with a novel epigenetic function. Genomics Proteomics Bioinformatics. 2013;11(1):8–17. doi: 10.1016/j.gpb.2012.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Chen J, Wei X, Yi X, et al. RNA modification by m 6 a methylation in cardiovascular disease. Oxid Med Cell Longev. 2021;2021(1):8813909. doi: 10.1155/2021/8813909 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Ma L, Chen T, Zhang X, et al. The m(6)A reader YTHDC2 inhibits lung adenocarcinoma tumorigenesis by suppressing SLC7A11-dependent antioxidant function. Redox Biol. 2021;38:101801. doi: 10.1016/j.redox.2020.101801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Xu Y, Lv D, Yan C, et al. METTL3 promotes lung adenocarcinoma tumor growth and inhibits ferroptosis by stabilizing SLC7A11 m(6)A modification. Cancer Cell Int. 2022;22(1):11. doi: 10.1186/s12935-021-02433-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Shi H, Zhang X, Weng YL, et al. m(6)A facilitates hippocampus-dependent learning and memory through YTHDF1. Nature. 2018;563(7730):249–253. doi: 10.1038/s41586-018-0666-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Li Q, Ni Y, Zhang L, et al. HIF-1α-induced expression of m6A reader YTHDF1 drives hypoxia-induced autophagy and malignancy of hepatocellular carcinoma by promoting ATG2A and ATG14 translation. Signal Transduct Target Ther. 2021;6(1):76. doi: 10.1038/s41392-020-00453-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Ruan F, Zeng J, Yin H, et al. RNA m6A modification alteration by black phosphorus quantum dots regulates cell ferroptosis: implications for nanotoxicological assessment. Small Methods. 2021;5(3):e2001045. doi: 10.1002/smtd.202001045 [DOI] [PubMed] [Google Scholar]
  • [129].Ma L, Zhang X, Yu K, et al. Targeting SLC3A2 subunit of system X(C)(-) is essential for m(6)A reader YTHDC2 to be an endogenous ferroptosis inducer in lung adenocarcinoma. Free Radic Biol Med. 2021;168:25–43. doi: 10.1016/j.freeradbiomed.2021.03.023 [DOI] [PubMed] [Google Scholar]
  • [130].Ye J, Chen X, Jiang X, et al. RNA demethylase ALKBH5 regulates hypopharyngeal squamous cell carcinoma ferroptosis by posttranscriptionally activating NFE2L2 / NRF2 in an m 6 A-IGF2BP2 -dependent manner. J Clin Lab Anal. 2022;36(7):e24514. doi: 10.1002/jcla.24514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Ji FH, Fu XH, Li GQ, et al. FTO prevents thyroid cancer progression by SLC7A11 m6A methylation in a ferroptosis-dependent manner. Front Endocrinol (Lausanne). 2022;13:857765. doi: 10.3389/fendo.2022.857765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Pasquinelli AE, Ruvkun G. Control of developmental timing by microRNAs and their targets. Annu Rev Cell Dev Biol. 2002;18:495–513. doi: 10.1146/annurev.cellbio.18.012502.105832 [DOI] [PubMed] [Google Scholar]
  • [133].McManus MT. MicroRNAs and cancer. Semin Cancer Biol. 2003;13:253–258. doi: 10.1016/s1044-579x(03)00038-5 [DOI] [PubMed] [Google Scholar]
  • [134].Yadav P, Sharma P, Sundaram S, et al. SLC7A11/xCT is a target of miR-5096 and its restoration partially rescues miR-5096-mediated ferroptosis and anti-tumor effects in human breast cancer cells. Cancer Lett. 2021;522:211–224. doi: 10.1016/j.canlet.2021.09.033 [DOI] [PubMed] [Google Scholar]
  • [135].Li X, Si W, Li Z, et al. miR‑335 promotes ferroptosis by targeting ferritin heavy chain 1 in in vivo and in vitro models of Parkinson’s disease. Int J Mol Med. 2021;47(4). doi: 10.3892/ijmm.2021.4894 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Ding C, Ding X, Zheng J, et al. miR-182-5p and miR-378a-3p regulate ferroptosis in I/R-induced renal injury. Cell Death Dis. 2020;11(10):929. doi: 10.1038/s41419-020-03135-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Fan K, Huang W, Qi H, et al. The Egr-1/miR-15a-5p/GPX4 axis regulates ferroptosis in acute myocardial infarction. Eur J Pharmacol. 2021;909:174403. doi: 10.1016/j.ejphar.2021.174403 [DOI] [PubMed] [Google Scholar]
  • [138].Xiao FJ, Zhang D, Wu Y, et al. miRNA-17-92 protects endothelial cells from erastin-induced ferroptosis through targeting the A20-ACSL4 axis. Biochem Biophys Res Commun. 2019;515(3):448–454. doi: 10.1016/j.bbrc.2019.05.147 [DOI] [PubMed] [Google Scholar]
  • [139].Ni H, Qin H, Sun C, et al. MiR-375 reduces the stemness of gastric cancer cells through triggering ferroptosis. Stem Cell Res Ther. 2021;12:325. doi: 10.1186/s13287-021-02394-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Wei D, Ke YQ, Duan P, et al. MicroRNA-302a-3p induces ferroptosis of non-small cell lung cancer cells via targeting ferroportin. Free Radic Res. 2021;55(7):821–830. doi: 10.1080/10715762.2021.1947503 [DOI] [PubMed] [Google Scholar]
  • [141].Bai T, Liang R, Zhu R, et al. MicroRNA-214-3p enhances erastin-induced ferroptosis by targeting ATF4 in hepatoma cells. J Cell Physiol. 2020;235:5637–5648. doi: 10.1002/jcp.29496 [DOI] [PubMed] [Google Scholar]
  • [142].Chen D, Fan Z, Rauh M, et al. ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene. 2017;36(40):5593–5608. doi: 10.1038/onc.2017.146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Bhan A, Soleimani M, Mandal SS. Long noncoding RNA and Cancer: a new paradigm. Cancer Res. 2017;77:3965–3981. doi: 10.1158/0008-5472.Can-16-2634 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Cai HJ, Zhuang ZC, Wu Y, et al. Development and validation of a ferroptosis-related lncRNAs prognosis signature in colon cancer. Bosn J Basic Med Sci. 2021;21:569–576. doi: 10.17305/bjbms.2020.5617 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Yao J, Chen X, Liu X, et al. Characterization of a ferroptosis and iron-metabolism related lncRNA signature in lung adenocarcinoma. Cancer Cell Int. 2021;21:340. doi: 10.1186/s12935-021-02027-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Lu J, Xu F, Lu H. LncRNA PVT1 regulates ferroptosis through miR-214-mediated TFR1 and p53. Life Sci. 2020;260:118305. doi: 10.1016/j.lfs.2020.118305 [DOI] [PubMed] [Google Scholar]
  • [147].Zhang Y, Guo S, Wang S, et al. LncRNA OIP5-AS1 inhibits ferroptosis in prostate cancer with long-term cadmium exposure through miR-128-3p/SLC7A11 signaling. Ecotoxicol Environ Saf. 2021;220:112376. doi: 10.1016/j.ecoenv.2021.112376 [DOI] [PubMed] [Google Scholar]
  • [148].Yang Y, Tai W, Lu N, et al. lncRNA ZFAS1 promotes lung fibroblast-to-myofibroblast transition and ferroptosis via functioning as a ceRNA through miR-150-5p/SLC38A1 axis. Aging (Albany NY). 2020;12(10):9085–9102. doi: 10.18632/aging.103176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Luo W, Wang J, Xu W, 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 Dis. 2021;12(11):1043. doi: 10.1038/s41419-021-04296-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Gao M, Li C, Xu M, et al. LncRNA MT1DP aggravates cadmium-induced oxidative stress by repressing the function of Nrf2 and is dependent on interaction with miR-365. Adv Sci (Weinh). 2018;5:1800087. doi: 10.1002/advs.201800087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Qi W, Li Z, Xia L, et al. LncRNA GABPB1-AS1 and GABPB1 regulate oxidative stress during erastin-induced ferroptosis in HepG2 hepatocellular carcinoma cells. Sci Rep. 2019;9(1):16185. doi: 10.1038/s41598-019-52837-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Sui X, Hu N, Zhang Z, et al. ASMTL-AS1 impedes the malignant progression of lung adenocarcinoma by regulating SAT1 to promote ferroptosis. Pathol Int. 2021;71:741–751. doi: 10.1111/pin.13158 [DOI] [PubMed] [Google Scholar]
  • [153].Mao C, Wang X, Liu Y, et al. A G3BP1-interacting lncRNA promotes ferroptosis and apoptosis in cancer via nuclear sequestration of p53. Cancer Res. 2018;78:3484–3496. doi: 10.1158/0008-5472.Can-17-3454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Goodall GJ, Wickramasinghe VO. RNA in cancer. Nat Rev Cancer. 2021;21(1):22–36. doi: 10.1038/s41568-020-00306-0 [DOI] [PubMed] [Google Scholar]
  • [155].Wang Y, Mo Y, Gong Z, et al. Circular RNAs in human cancer. Mol Cancer. 2017;16(1):25. doi: 10.1186/s12943-017-0598-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Zuo YB, Zhang YF, Zhang R, et al. Ferroptosis in cancer progression: role of noncoding RNAs. Int J Biol Sci. 2022;18:1829–1843. doi: 10.7150/ijbs.66917 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Zhang H, Ge Z, Wang Z, et al. Circular RNA RHOT1 promotes progression and inhibits ferroptosis via mir-106a-5p/STAT3 axis in breast cancer. Aging (Albany NY). 2021;13(6):8115–8126. doi: 10.18632/aging.202608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Chen W, Fu J, Chen Y, et al. Circular RNA circKIF4A facilitates the malignant progression and suppresses ferroptosis by sponging miR-1231 and upregulating GPX4 in papillary thyroid cancer. Aging (Albany NY). 2021;13(12):16500–16512. doi: 10.18632/aging.203172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Zhang X, Huang Z, Xie Z, et al. Homocysteine induces oxidative stress and ferroptosis of nucleus pulposus via enhancing methylation of GPX4. Free Radic Biol Med. 2020;160:552–565. doi: 10.1016/j.freeradbiomed.2020.08.029 [DOI] [PubMed] [Google Scholar]
  • [160].Yang H, Zhao L, Gao Y, et al. Pharmacotranscriptomic analysis reveals novel drugs and gene networks regulating ferroptosis in cancer. Cancers (Basel). 2020;12(11):3273. doi: 10.3390/cancers12113273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Du Y, Han M, Cao K, et al. Gold nanorods exhibit intrinsic therapeutic activity via controlling N6-methyladenosine-based epitranscriptomics in acute myeloid leukemia. ACS Nano. 2021;15(11):17689–17704. doi: 10.1021/acsnano.1c05547 [DOI] [PubMed] [Google Scholar]
  • [162].Yang C, Lu T, Liu M, et al. Tiliroside targets TBK1 to induce ferroptosis and sensitize hepatocellular carcinoma to sorafenib. Phytomedicine. 2023;111:154668. doi: 10.1016/j.phymed.2023.154668 [DOI] [PubMed] [Google Scholar]
  • [163].Peng Y, Li N, Tang F, et al. Corosolic acid sensitizes ferroptosis by upregulating HERPUD1 in liver cancer cells. Cell Death Discov. 2022;8:376. doi: 10.1038/s41420-022-01169-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Hu S, Sechi M, Singh PK, et al. A novel redox modulator induces a GPX4-mediated cell Death that is dependent on iron and reactive oxygen species. J Med Chem. 2020;63:9838–9855. doi: 10.1021/acs.jmedchem.0c01016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Wang X, Chen J, Tie H, et al. Eriodictyol regulated ferroptosis, mitochondrial dysfunction, and cell viability via Nrf2/HO-1/NQO1 signaling pathway in ovarian cancer cells. J Biochem Mol Toxicol. 2023;37(7):e23368. doi: 10.1002/jbt.23368 [DOI] [PubMed] [Google Scholar]
  • [166].Alborzinia H, Ignashkova TI, Dejure FR, et al. Golgi stress mediates redox imbalance and ferroptosis in human cells. Commun Biol. 2018;1(1):210. doi: 10.1038/s42003-018-0212-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Pei Y, Qian Y, Wang H, et al. Epigenetic regulation of ferroptosis-associated genes and its implication in cancer therapy. Front Oncol. 2022;12:771870. doi: 10.3389/fonc.2022.771870 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data was used for the research described in the review.


Articles from Epigenetics are provided here courtesy of Taylor & Francis

RESOURCES