Open Access

Regulators of epigenetic change in ferroptosis‑associated cancer (Review)

  • Authors:
    • Jiaming Wu
    • Shuang Zhu
    • Peng Wang
    • Jinge Wang
    • Jingjing Huang
    • Tong Wang
    • Lingfeng Guo
    • Desen Liang
    • Qinghui Meng
    • Huayang Pan
  • View Affiliations

  • Published online on: October 20, 2022     https://doi.org/10.3892/or.2022.8430
  • Article Number: 215
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The occurrence of tumors is associated with the upregulation or downregulation of certain genes. The identification of novel tumor therapies has revealed that regulation of tumor cell death can either promote or suppress the occurrence and development of tumors. Iron‑dependent lipid free oxygen radical accumulation causes tumor cells to die by ferroptosis, a form of regulated cell death. Multiple mechanisms mediate this mode of cell death, including redox homeostasis, iron metabolism, mitochondrial activity, breakdown of amino acids, lipids and sugars and epigenetic regulatory and disease‑associated signaling pathways. The present review discussed epigenetic mechanism of ferroptosis with the aim of providing novel insight for optimization of the effects of antitumor therapy.

Introduction

Under physiological and pathological conditions, cell life cycle comes to an end with cell death. Necrosis and apoptosis are considered forms of cell death. Studies have reported that certain forms of programmed cell death, such as autophagy and necroptosis, exhibit unique pathophysiological features that are distinct from other forms of programmed cell death, such as necrosis and apoptosis (1,2). In 2012, the concept of ferroptosis was proposed (3). During ferroptosis, lipid reactive oxygen species (ROS) are produced as a result of iron-dependent, non-apoptotic cell death. In ferroptosis, iron reacts with hydrogen peroxide, releasing electrons and generating hydroxyl radicals. These reactions damage intracellular lipids and proteins and cause oxidative damage to DNA, which accelerates death of tumor cells (4). Ferroptosis is morphologically and functionally distinct from necrosis, autophagy and apoptosis; it does not show typical characteristics of necrosis or apoptosis, including cytoplasmic swelling, cell shrinkage and rupture, presence of apoptotic bodies or cytoskeletal disintegration. Additionally, unlike autophagy, it does not involve formation of closed lipid membrane bilayers, but a specific structure known as the autophagic vacuole (5). In terms of morphology, ferroptosis is marked by mitochondrial shrinkage and increased membrane density (6). Ferroptosis is associated with a range of diseases (eg:gastric cancer) (7). Ferroptosis is regulated by epigenetic mechanisms (8). Epigenetic modifications are alterations in gene expression or cell phenotype caused by heritable changes that affect DNA methylation, histone modifications and non-coding RNA (ncRNA) regulation. Epigenetic mechanisms also control gene transcription, cell proliferation, developmental processes and immune function (9). Furthermore, epigenetic regulation is implicated in ferroptosis and tumorigenesis via modulation of metabolic genes and intermediates, thereby regulating and altering lipid peroxidation (10). The epigenetic control of ferroptosis also provides novel directions for development of therapeutic interventions, which may overcome the current barriers in antitumor therapy (11). The present review aimed to discuss how epigenetic regulators regulate ferroptosis and how ferroptosis contributes to tumor biology.

Epigenetics, iron homeostasis and ROS

Iron is a key trace element in the human body. It participates in heme synthesis and serves an important role in various physiological metabolic processes as a co-factor of certain key enzymes (12). The human body maintains iron homeostasis by regulating intestinal circulation and absorption of iron in the reticuloendothelial system via coordination of iron absorption, utilization, storage and circulation (13). There are numerous genes involved in maintaining iron homeostasis. Cellular iron homeostasis is primarily controlled by iron regulatory protein (IRP)-iron response element (14). IRP is involved in regulation of iron absorption, transport and storage, alongside transferrin receptor 1 (TFR1), divalent metal ion transporter (DMT1) and ferritin (15). Iron metabolism in the human body is primarily regulated by hepcidin-ferroportin (HAMP-FPN), in which HAMP regulates iron uptake and release in tissue, as well as iron homeostasis in the body (16). Under iron overload, HAMP binds to FPN on target cell membranes and internalizes and degrades FPN in lysosomes, thereby inhibiting iron absorption by enterocytes and releasing iron from macrophages or hepatocytes into the serum, thus regulating iron absorption and distribution (17). At the molecular level, HAMP is regulated by the bone morphogenetic protein (BMP)/hemojuvelin (HJV)/SMAD signaling pathway (18). HJV is a co-receptor of BMP (19). By enhancing BMP expression, the SMAD signaling pathway is stimulated and HAMP expression is promoted (20). BMP stimulates HAMP expression; this process is also regulated by HFE protein (21). In a high-iron environment, transferrin competes to capture HFE-bound TFR1, resulting in HFE dissociating from TFR1 and binding to TFR2, thereby activating the SMAD signaling pathway and inducing HAMP expression. Membrane-type serine protease 2 inhibits HAMP expression by shear HJV negative feedback. Any gene abnormalities in the signal transduction pathway regulated by HAMP affect the expression of HAMP and cause disorders associated with iron metabolism (22). Recent studies (2325) on iron metabolism have revealed that epigenetic inheritance serves an important role in iron homeostasis metabolism. DNA methylation generally occurs in cytosine-guanine dinucleotide CpG-rich DNA regions, called CpG islands (CGIs), which are the most widely studied epigenetic modifications and serve an important role in the regulation of gene expression. Vertebrates have a higher methylation rate of CpG dinucleotides than mammals, with ~80% containing 5-methylcytosine (5mC), a gene silencing marker. There are certain exceptions to this hypermethylated state, namely CGIs, which are typically hypomethylated in comparison with the rest of the genome (26). CGIs in promoter regions overlap in the earliest vertebrates and humans, suggesting a coevolutionary CGI-compatible system. DNA sequence features are characteristic of CGI, including hypomethylation of DNA, high CpG and GC content and transcription factor binding (27). Transcription factors and chromatin-modifying enzymes are recruited along with transcriptional activators by these sequence features. CGIs represent a ubiquitous class of DNA sequences commonly associated with promoters of vertebrate genes whose sequence features make them transcriptionally active (28). DNA sequence and chromatin determinants allow identification of CGIs, including decreased DNA methylation (5mC), increased CpG and GC content and histone H3 trimethylation (H3K4me3) and transcription factor binding sites (29). Increased DNA methylation leads to gene silencing, while decreased DNA methylation activates gene expression. Iron metabolism is associated with DNA methylation and iron level and Oxidation state affect DNA methylation (30). Increased free iron content in the brain due to oxidative stress leads to excessive S-adenosine homocysteine in HFE mutant mice by inhibition of the activity of DNA methyltransferase (31). Treatment with the iron chelator deferoxamine improves activity of DNA methyltransferase and certain breast cancer cells are induced by methylation (32). 3-Hydroxybutyrate dehydrogenase 2 is a rate-limiting enzyme in mammalian ferriphilin synthesis. Inhibition of its expression causes iron overload in cells and mitochondria (33). In tumor cell proliferation, iron demand is increased and the expression of genes associated with iron metabolism is affected. Previous studies (34,35) have found that low expression of FPN in patients with breast cancer indicates poor survival. In vitro studies (36,37) also found that inhibition of FPN expression promotes tumor growth, while overexpression of FPN hinders tumor growth. Low expression of FPN in breast cancer is regulated by nuclear factor erythroid 2-related factor 2 (NRF2) and is inhibited by hypermethylation of CGIs in the FPN promoter region, indicating that breast cancer can be inhibited by targeting FPN or upstream regulatory factors (38). HAMP expression is inhibited in patients with hepatocellular carcinoma (HCC), which is accompanied by hypermethylation of the highly conserved CGI site in its promoter region (38). Demethylating drug treatment removes hypermethylation of the HAMP promoter and upregulates expression of HAMP in HCC cells (39). TFR2 in HCC cells is also regulated by hepcidin in hepatocellular carcinoma. Therefore, regulation of HAMP by hypermethylation is a recently (40) identified form of epigenetic regulation of iron metabolism, which provides a novel area of research on tumors and iron. Since iron can easily change its valence state and switch between Fe2+ and Fe3+, iron is a key catalyst to produce active free radicals within aerobic organisms. Rigid binding between iron and transferrin occurs in vivo. The activity of O2− and H2O2 is not sufficient for oxidation of certain macromolecules, such as nucleic acids and proteins. Iron overload is not always toxic and the excess iron can initially be chelated in ferritin or lysosomes in a safe manner. However, once the quantity of accumulated iron exceeds storage capacity, Iron not chelated with ferritin emerges, promoting oxidative stress responses (41). The reaction between Fe2+ and H2O2 produces hydroxyl radical (HO.), which is known as the Fenton reaction (42). Iron not chelated with ferritin is more conducive to Fenton reaction than iron chelated with ferritin and unstable iron present in the body passes through HO. with RO. generation, which enhances the effect of H2O2 and other organic peroxides in cells (43). The majority of O2 consumed by aerobic organisms is safely reduced to H2O in a four-electron transfer reaction catalyzed by the cytochrome oxidase complex IV of the mitochondrial inner membrane respiratory chain (44). However, a fraction of unused O2 is involved in other physiological reactions, such as phagocytosis and immune activation. The toxic intermediates that form are called ROS. Under normal conditions, specific enzymes rapidly metabolize ROS, such as glutathione peroxidase (GPX) (45). There are a variety of biochemical and physiological oxidative processes in the body that produce ROS; these processes are also linked to numerous physiological and pathological processes, eg:heart disease (46). By regulating intracellular signal transduction and homeostasis, ROS exert beneficial effects at low concentrations. At high concentrations, however, ROS primarily cause protein, lipid and DNA damage. Endogenous ROS are primarily derived from byproducts of subcellular organelles such as mitochondria. A normal cell can become malignant if it contains high levels of ROS. It has been shown that ROS influence epigenetic inheritance, including DNA methylation (47). In cancer cells, ROS enhance DNA methylation, leading to silencing of tumor suppressor and antioxidant genes, and cancer cell proliferation under oxidative stress. In colorectal cancer cells, ROS increase expression of caudal type homeobox 1 (CDX1) (48). Treatment with H2O2 increases expression and activity of CDX1, DNA methylated transferase 1 (DNMT1) and histone deacetylase (HDAC1) in cancer cells, which promotes cancer progression (49). In Parkinson's disease, peroxisome proliferators-activated receptors coactivator 1α (PGC-1α) downregulation is associated with mitochondrial dysfunction, oxidative stress and inflammation. PGC-1α is required by nerve cells to induce proteins that detoxify ROS, such as GPX (50). In PGC-1α knockout mice, glial cells are more susceptible compared with normol mice to neuroinflammation induced by Toll-like receptor 4 agonists. PGC-1α downregulation leads to increased ROS and neurological damage (51,52) Previous studies (53,54) have shown that proinflammatory fatty acid palmitate leads to atypical cytosine methylation in the PGC-1α gene promoter and decreases gene expression and mitochondrial content. Thus, PGC-1α promoter methylation is associated with dysfunctional inflammatory signaling (55). Cells in the body continuously produce and remove ROS in a dynamic equilibrium under physiological conditions. Once redox imbalance occurs in cells, redox signals are generated, which lead to adaptive changes in gene expression. Iron and iron derivatives bind to ROS-producing enzymes, such as the NADPH and cytochrome P450 enzyme systems. Lysosomes also contain redox-active iron pools from extracellular sources that catalyze the generation of damaging free radicals via the Fenton reaction (56).

Mechanisms of ferroptosis in cancer

Cell death mediated by ferroptosis occurs in numerous types of disease, including cancer. Unlike autophagy and apoptosis, ferroptosis is characterized by dependence on intracellular free iron ions and ROS (57). Activated circulating iron (Fe3+) enters cells and is reduced to Fe2+ by ferrooxidoreductase six transmembrane epithelial antigen 3 (58). Once endosomes release Fe2+ via DMT1, it is transported to the cytoplasm, where it couples with ROS, leading to lipid peroxidation and ferroptosis (59). ROS are molecules containing reduced oxygen, which include superoxide (O2−) and peroxides (H2O2 and ROOH), as well as free radicals (HO and RO). In cells, increased ROS production induces ferroptosis (60). In cases of lipid peroxidation and oxidative stress, the permeability of the cell membrane directly leads to reduction or disappearance of intracellular mitochondria and the rupture or aggregation of mitochondrial membranes (61). In the case of malignant tumors, ferroptosis serves a crucial role in eradicating tumor cells as an adaptive process (62). Tumor cells are highly sensitive to ferroptosis, which controls the abundance of iron by activating expression of transferrin receptor and ferritin, thus activating the RAS/MEK signaling pathway. When the RAS/MEK signaling pathway is activated, tumor cells are prevented from absorbing cystine and ROS are released through mitochondrial voltage-dependent anion channels (63). For example, activated RAS/MEK signaling induces expression of SOSC1, which controls expression of P53, decreases expression of cystine transporter and sensitizes cells to ferroptosis (64). In addition, various components (Beclin1) promote ferroptosis and indirectly decrease activity of system Xc (65). Low member 11 of the solute carrier family 7 (SLC7A11) expression prevents Cys2 transport and activation of the glutathione (GSH)-independent thioredoxin (TXN) system, leading to dysfunction of the GSH/GPX4 lipid peroxidation pathway (66). Mechanistically, this may be due to the inability to transfer a sufficient quantity of reducing equivalents from Cys2 to TXN (via TXN reductase) to maintain the endogenous antioxidant lipid α-tocopherol in a reduced state, thereby promoting ROS accumulation and ferroptosis (67). In cancer, the mechanism of ferroptosis primarily involves three major systems (Fig. 1).

Xc-/GSH/GPX4 system

The antioxidant system Xc- consists of transporters located on cell membranes. The primary system maintains homeostasis of glutamate and cystine on cell membranes, where it pumps out glutamate molecules and pumps in cystine molecules (68). The functional subunits of the system Xc- are SLC7A11 and SLC3A2 (68). GSH is synthesized by reduction of cystine to cysteine by system Xc-. Glutamyl-L-cysteine-L-glycine (GSH) is an endogenous antioxidant consisting of glutamic acid, cysteine and glycine that scavenges free radicals. Its production is dependent on activity of glutamate-cysteine ligase and inhibition of system Xc- attenuates its activity, resulting in GSH depletion and leading to ferroptosis (69). Trans-sulfuration is another mechanism by which methionine synthesizes cysteine. Cysteine is still synthesized when intracellular system Xc- is inhibited (70). Thus, ferroptotic inducers that negatively regulate system Xc- cannot effectively kill cells. The trans-sulfuration pathway is activated by interfering with RNA dynamics to decrease expression of cysteine transfer RNA synthetase. As a result, cells become less sensitive to ferroptosis-inducing agents (71). Cells that contain GPX4 decrease lipid peroxidation, which promotes their survival. Aberrant expression of GPX4 is linked to a range of diseases (breast cancer) in humans and depletion of GPX4 increases H2O2-containing phospholipids (72), promotes lipoxygenase-mediated lipid peroxidation and induces ferroptosis (73). GPX4 is highly expressed in tumor tissue, while histone H3 is trimethylated at lysine 4 (H3K4me3) and acetylated at lysine 27 (H3K27ac) at the GPX4 transcriptional start site. Patients with cancer have a poor prognosis when epigenetic modifications of GPX4 are present; thus, elevated expression of GPX4 in tumor tissue may be due to epigenetic modifications (74). In addition, the activation of autophagy degrades intracellular ferritin and directly induces ferroptosis in tumor cells. Autophagy is a lysosome-dependent process that promotes ferroptosis by generating lysosomal ROS. Promoting autophagy effectively enhances ferroptosis in tumor cells (75). The autophagy-mediated nuclear receptor coactivator (NCOA4) signaling pathway promotes ferritin degradation and enhances ferroptosis when NCOA4 is overexpressed (76), which demonstrates the association between ferroptosis and apoptosis.

NADPH/ferroptosis suppressor protein 1 (FSP1)/coenzyme Q10 (COQ10)

Although GPX4 is a key regulator of ferroptosis, inhibition of GPX4 does not induce ferroptosis in certain cell lines. A previous study identified potential factors that regulate ferroptosis independently of the GPX4 signaling pathway (72). Cells were treated with a ferroptosis initiator [RAS selective lethal 3 (RSL3)], and it was observed that Glutathione Independent Iron Death Inhibitor Protein (FSP1) induced apoptosis. The inhibition of GPX4 leads to ferroptosis when FSP1 is overexpressed. Therefore, FSP1 may act as a ferroptosis inhibitor (77). However, AIFM1, although homologous to FSP1, does not inhibit ferroptosis (78). FSP1 may inhibit ferroptosis because its N-terminal myristoylated mediates FSP1 localization to the plasma membrane. Furthermore, myristoylation of the N-terminus of FSP1 promotes binding of target proteins to the cell membrane. In the plasma membrane, COQ10 serves as a lipophilic radical scavenger (79), while FSP1 serves as a NADPH-dependent COQ oxidoreductase that regulates COQ10 in vitro. FSP1 is a component of the traditional mitochondrial respiratory chain that catalyzes the same reactions as complex I. Extra-mitochondrial ubiquinone is reduced from COQ10 by FSP1, which either directly captures lipid free radicals or indirectly serves as an antioxidant by recycling α-tocopherol (77). Idebenone, a soluble analog of COQ10, inhibits ferroptosis and lipid peroxidation by catalyzing the first step in the biosynthesis of COQ2 (80). The aforementioned findings showed that FSP1, like GPX4, inhibits ferroptosis by regulating the non-mitochondrial COQ10 antioxidant system (80).

GTP cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4)

The redox-active cofactor BH4 serves a role in production of neurotransmitters, carbon monoxide and aromatic amino acids. In vitro, BH4 exhibits antioxidant properties and GCH1 limits BH4 synthesis (82). In tumor cells, GCH1 overexpression clears lipid peroxidation and protects cells from ferroptosis. Overexpression of GCH1 protects against RSL3-induced ferroptosis when cells were treated with RSL3 and apoptosis-inducing agents. GCH1 protects cells from ferroptosis (83). Treatment of RSL3-exposed cells with BH4 completely prevents ferroptosis. It has been previously shown that GCH1 acts independently of the ferroptosis and GSH pathways to prevent ferroptosis (84). Notably, BH4 as a cofactor for biosynthetic enzymes does not serve a role in ferroptosis protection (85). In addition, BH4 converts phenylalanine to tyrosine to promote synthesis of COQ10, thereby exerting an antioxidant effect (86).

Epigenetic regulation of ferroptosis in cancer

Alterations in DNA methylation, histone modifications and ncRNA regulation are characteristic of epigenetic regulation, which determines gene transcription, cell proliferation, developmental processes and immune function (87). Epigenetic regulation is important for ferroptosis.

DNA methylation and ferroptosis

There are numerous roles for DNA methylation in cancer, including suppression of DNA methylation at transcriptional start sites of key gene regulatory elements, such as enhancers and promoters (88). In DNA methylation, a methyl group is typically inserted at the carbon 5 position of the cytosine base (5mC) via DNA methylation mediated by CpG transferase DNMTs (89). DNA methylation primarily regulates mitotic gene expression, centromere stability and chromatin segregation. In addition, 5-hydroxymethylcytosine (5hmC) has been demonstrated to be produced by 5mC oxidation. 5hmC has been widely studied in relation to its potential role in modifying methylation landscapes by ten-eleven translocation (TET) enzymes (90,91). The ability of TET proteins to oxidize 5hmC to 5-formylcytosine and 5-carboxycytosine indicates utilization of the base excision repair pathway, while thymine DNA glycosylase excises cytosine and replaces it with an unmodified cytosine (92). CpG sites are found at ~28 million sites in the human genome. The distribution of CpG sites in somatic cells is uneven and ~70% of CpG sites are oxidized in normal somatic cells (93). Clusters of CpG sites are known as CGIs; CpG sites in CGIs tend not to be methylated in somatic cells. Chromosomes are their primary medium. Unmethylated CpG sites are found in promoter CGIs, where transcription factors bind to control gene expression. The CpG coast is ~2 kb pairs away from the CGI and has a lower density of CpGs than the CGI (94). Normal cells also contain these unmethylated regions that regulate gene activity (95). During tumorigenesis, normal epigenetic processes such as DNA methylation are disrupted. Tumor development is primarily accompanied by genome-wide hypomethylation and DNA hypermethylation in the promoter regions of CGIs (96). Cell proliferation and tumor suppressor genes and downstream signaling pathways are primarily silenced by the hypermethylation of CGIs (97). Moreover, tumors also abnormally express DNMT enzymes, resulting in aberrant DNA methylation across the genome, potentially causing mutations in genomic sequences (98). Aneuploidy and genomic instability are associated with genome-wide hypomethylation in cancer (99). The abnormal expression of transposable elements and oncogenes is also associated with genome-wide DNA hypomethylation, which disrupts cellular pathways and changes chromatin structure, as chromatin structure and DNA methylation are associated and a nucleosome is required before 5mC can be obtained (100). To anchor the DNMT enzyme, abnormal DNA methylation must disrupt chromatin structure (101). The most studied (102) epigenetic modification is DNA methylation (103). However, the mechanism of this process in unclear and the number of cancer-associated genes that are silenced and targeted is unknown. A recent study on the association between DNA methylation, ferroptosis and cancer has led to novel targeted therapy for cancer (104). Hydricase, lymphoid specific (HELLS/LSH) is a chromatin remodeling enzyme that inhibits ferroptosis by activating metabolic genes such as stearoyl-coenzyme A desaturase (SCD). HELLS induces epigenetic silencing of the long non-coding RNA (lncRNA) LINC00472. LINC00472, as a tumor suppressor, is downregulated in tumors and can inhibit ferroptosis (105). Egl nine homolog 1 and c-Myc in tumor cells activate LSH expression by inhibiting hypoxia inducible factor 1. LSH inhibits iron dynamics by activating GLUT1, SCD1 and fatty acid desaturase 2, which are involved in lipid metabolism, by interacting with WD repeat domain 76. Several types of cancer tissue (pancreatic and lung cancer) express high levels of SCD1, an enzyme that catalyzes fatty acid synthesis at the rate-limiting step. Through inhibition of SCD1, COQ10 activity is decreased, resulting in lipid oxidation and cell death (106). GPX4 DNA methylation in the nucleus pulposus is induced by homocysteine treatment, resulting in ferroptosis (107). Furthermore, DNA hypermethylation of the cadherin 1 (CDH1) gene promoter increases ferroptosis by suppressing expression of E-cadherin, which is encoded by CDH1 (108). DNA methylation contributes to ferroptosis-associated gene silencing, as demonstrated by the aforementioned studies. There is, however, a need for further research to determine whether DNA methylation also affects other ferroptosis-associated genes. Notably, TET proteins also catalyze DNA demethylation by oxidizing 5mC (109).

ncRNAs and ferroptosis

RNA is a key biological macromolecule in cells and it can be classified into two categories according to its function, namely, coding RNA and ncRNA. RNA is responsible for gene regulation and information transmission and is involved in disease. For example, nc-RNAs encode tumor peptides or proteins (110,111). The following subsections review ncRNAs in the context of microRNAs (miRNAs or miRs), lncRNAs and circular RNAs (circRNAs) involved in ferroptosis.

miRNAs and ferroptosis

miRNAs are the most well-studied (112) ncRNAs in tumor biology (113). Endogenous genes encode a class of RNA molecules known as miRNAs, which are single-stranded ncRNA molecules with a length of 20–24 nucleotides. miRNAs regulate translational and transcriptional activities that control metabolic pathways, tumor growth and cell migration and invasion and induce resistance to chemotherapeutic agents (113). During post-transcriptional processing, miRNAs bind to the 3′ untranslated region of target mRNAs and regulate gene expression. miRNAs that bind to specific mRNAs directly inhibit translation by suppressing translation or decreasing mRNA stability post-transcriptionally (114). In addition to controlling cell differentiation, proliferation, drug resistance and lipid metabolism, miRNAs also participate in several other biological processes. For example, mirnas prevent translation by inactivating or inducing mRNA degradation, which is a key step in post-transcriptional gene regulation (115). Cancer cells are genetically or epigenetically altered as a result. Furthermore, miRNAs inhibit tumor cell proliferation by altering ferroptosis (116). For example, miR-9 interferes with the catalytic efficacy of glutamic-oxaloacetic transaminase 1 in melanoma cells, thus inhibiting its ability to transaminate α-ketoglutarate. By contrast, miR-9 inhibition causes lipid ROS accumulation, thereby increasing ferroptosis (117). Upregulation of miR-137 protects melanoma cells against ferroptosis by inhibiting the glutamine transporter SLC1A5 (118). By decreasing ferrous iron levels in the cytoplasm, miR-7-5p also decreases tumor cell migration and invasion (119). By decreasing tumor cell susceptibility to ferroptosis induced by treatment with RSL3 and erastin, activating transcription factor 4 knockout increases inhibition of tumor cell proliferation (120). In addition, GPX4 is inhibited by miR-4715-3p and cisplatin sensitivity is enhanced by miR-4715-3p. As a result of inhibition of the intracellular aurora kinase A, a key serine-threonine kinase in mitosis, the expression of miR-4715-3p is suppressed (121). In addition, inhibition of miR-4715-3p results in decreased levels of intracellular GSH, thereby preventing ferroptosis (122). Human colon adenocarcinoma cells are protected from oxidative damage by miR-185, which increases GPX2 levels in cells (123). Furthermore, specific transcriptional targets of miRNAs are involved in GSH metabolism, which is associated with ferroptosis and tumorigenesis (124,125). miRNAs serve both tumor-suppressive and -promotive functions in ferroptosis. Additionally, miRNAs modulate gene transcription at the mRNA level to decrease tumorigenesis, ferroptosis or degraded exon production (126). To determine the roles of miRNAs in tumor resistance mechanisms, it is imperative to understand the molecular mechanisms of miRNAs.

lncRNAs and ferroptosis

RNA molecules >200 nucleotides in length are known as lncRNAs and influence gene expression at different levels, including transcription and post-transcriptional translation. lncRNAs have also been reported to be key for a variety of biological processes, such as cell differentiation, regulation of the cell cycle and maintenance of stem cell pluripotency (127). Via downregulation of membrane receptors and inhibition of necroptosis-associated proteins, overexpression of lncRNAs inhibits the extrinsic apoptotic pathway in tumor cells (128). lncRNAs are important regulators of ferroptosis. A number of them affect miRNAs, while others bind to specific enzymes. Wang et al (129) reported that the lncRNAs LINC00336 and embryonic lethal, abnormal vision, Drosophila-like 1 (ELAVL1) inhibit ferroptosis by interacting with each other. In addition, ELAVL1 increases LINC00336 expression by stabilizing its post-transcriptional levels when RSL3 is used to activate ferroptosis in lung cancer cell lines. Overexpression of LINC00336 inhibits ferroptosis caused by RSL3 treatment, as well as protein kinase-induced ferroptosis. Furthermore, overexpression of LINC00336 decreases intracellular Fe2+ and mitochondrial superoxide concentrations, as well as ROS production, demonstrating that LINC00336 overexpression inhibits ferroptosis (129). Urothelial cancer associated 1 (UCA1) sponges miR-16 and increases expression of glutaminase 2 (GLS2) in bladder cancer cells. There are two mechanisms by which lncRNAs affect ferroptosis (130). GLS2 enzyme catalyzes conversion of glutamine to glutamate and subsequent synthesis of GSH. NADPH is produced when glutamine enters the tricarboxylic acid cycle. Glutathione is reduced to GSH in the presence of NADPH (131). Therefore, UCA1 regulates GSH and NADPH expression, thereby enhancing the antioxidant effects of GPX4 and inhibiting ferroptosis in tumor cells by decreasing ROS production (132). The transcription factor NRF2 also has antioxidant properties and can activate downstream antioxidant factors. NRF2 can promote the production of NADPH via the pentose phosphate pathway, thereby enhancing antioxidant activity. Additionally, NRF2 activates transcription of GSH and GPX family genes, while GPX4 is activated to exert antioxidant effects during ferroptosis (133). The balance of intracellular iron levels is key for cell survival. The co-function of transferrin and transferrin receptor is required for the absorption of Fe3+ by cells. Subsequently, Fe3+ becomes Fe2+ through a redox reaction and it is then stored in the iron pool (134). Ferroptosis occurs when Fe2+ donates electrons to oxygen in the Fenton reaction to form ROS, which catalyzes generation of lipid free oxygen radicals (135). In HCC, for example, expression of the ncRNA plasmacytoma variant translocation 1 (PVT1) is increased; PVT1 binds to miR-150 and regulates the target hypoxia-inducible gene (HIG2) (136). HIG2 is involved in ferroptosis and iron uptake via hypoxia-induced protein expression. Decreased transferrin receptor and increased ferritin light chain expression are observed when PVT1 is silenced, thereby decreasing iron uptake and affecting ferroptosis (137). The induction of ferroptosis by erastin results in upregulation of the lncRNA GA binding protein subunit β1 (GABPB1)-antisense RNA 1, blocking of the transcription and translation of GABPB1 and the inhibition of peroxidase 5 (PRDX5) expression. PRDX5 is less effective than non tumor tissue at preventing ROS production from H2O2, which ultimately impairs the antioxidant capacity of tumor cells, resulting in increased cell death (138). Melanoma glucose-6-phosphate dehydrogenase (G6PD) may stimulate NADPH production, thereby activating NADPH oxidase 4 (NOX4) (139). Superoxide and ROS are produced by NOX4, a transmembrane protein. When growth arrest specific 5 is silenced, G6PD and NOX4 activity is increased (140). Furthermore, ferroptosis is induced by G6PD and NOX4 in tumor cells (141). Inhibition of iron-induced cell death by targeting the miR-106B-5p/acyl-coenzyme A synthetase long chain family member 4 axis is a mechanism by which H19 promotes cancer progression (142). Iron-induced death is also regulated by PVT1 via activation of TFR1 and TP53 by miR-214. The PVT1/miR-214-3p/GPX4 signaling pathway activates ketamine and inhibits the proliferation of HCC cells both in vitro and in vivo, leading to iron-induced cell death (143). In addition, lncRNA ZNFX antisense RNA 1 acts as a competing endogenous RNA for iron-induced death via the miR-150-5p/SLC38A1 axis (144). Multiple other lncRNAs are involved in iron-induced tumor cell death. Further studies are needed to investigate the specific mechanism by which lncRNA regulates iron-related cell death.

circRNAs and ferroptosis

circRNAs are formed by the 3′ and 5′ ends of mRNAs. According to their structural features, they are primarily classified into three categories, namely, exonic, circular intronic and exonic intronic circRNAs, and they function as miRNA sponges and protein scaffolds (145). Compared with miRNAs, the abundance, conservation and tissue specificity of circRNAs make them better biomarkers for certain pathological states. For example, circrNA-002178 is abnormally expressed in lung cancer, which is of great help in the diagnosis of lung cancer (146). The majority of circRNAs are highly stable and specific. Cancer cells express abnormal levels of circRNAs, which are involved in cell proliferation, autophagy and apoptosis, among other aspects of programmed cell death (147). circRNAs affect the post-transcriptional stability of miRNAs by altering their target gene expression. In tumor cells, circRNAs serve as sponges and regulate ferroptosis (148). Furthermore, they enhance tumor cell proliferation and invasion by sponging miR-761 and activating integrin β8 (ITGB8) in glioma, inhibit ferroptosis by activating ITGB8 and inhibit apoptosis by targeting MET, while knockdown of circ-tubulin kinase 2 promotes erastin-induced ferroptosis (149). In addition, 526 circRNAs are dysregulated in cervical cancer cells according to a previous high-throughput microarray-based study (150). circRNAs are predominantly involved in GSH metabolism according to a previous bioinformatics analysis (151). However, associated downstream factors and miRNAs have not been screened (152). Therefore, further studies on regulation of circRNAs in ferroptosis are needed (Fig. 2).

Histone modifications and ferroptosis

Chemical modifications of the tails of four core histone proteins (H2A, H2B, H3 and H4) alter the interactions between histones and other nuclear proteins, according to a recent study (153). Consequently, histone modification modifies expression of target genes. For example, the ferroptosis-associated genes GPX4 and SLC7A11 are regulated by histone modifications in non-Hodgkin lymphoma (154,155). Therefore, regulation of ferroptosis is dependent on histone modification.

Histone methylation and ferroptosis

The histone H3 lysine 9 demethylase that regulates SLC7A11 expression in response to erastin-induced ferroptosis is lysine demethylase 3B (156). E1A binding protein p300-CREB binding protein (CREBBP), which is involved in transactivation of genes, acetylates nuclear factor erythroid-derived 2-like 2 (NFE2L2) (157). Mouse double minute 2 homolog is a proto-oncogene associated with TP53 stability and activation, while cyclin-dependent kinase inhibitor 2A/ARF promotes its degradation. Ferroptosis is promoted by ARF independently of TP53 (158). Cancer is frequently caused by TP53 inactivation or mutations. A number of genes controlled by TP53 serve key roles in various cellular processes such as cell proliferation, cell cycle progression, cell death and response to DNA damage. Cancer cells, when activated by TP53, not only promote tumor growth but also inhibit ferroptosis (159). ARF protein disrupts CREBBP-NFE2L2 interaction, resulting in attenuated acetylation of NFE2L2 and the decreased expression of SLC7A11 mediated by NFE2L2 (160). Furthermore, members of the bromodomain (BRD) family are epigenetic regulators that recognize acetylated lysine residues on histones. JQ1, an inhibitor of the BRD4 complex, induces ferroptosis in breast and lung cancer cells by decreasing expression of GPX4, SLC7A11 and SLC3A2. Other anti-ferroptotic genes are also regulated by BRD4 (161).

Histone ubiquitination and ferroptosis

Cancer cells overexpress deubiquitinase OUT deubiquitinase ubiquitin aldehyde binding 1 (OTUB1), which stabilizes cystine transporter SLC7A11, inhibits ferroptosis and promotes tumor growth (162). BRCA1-associated protein 1 (BAP1) decreases SLC7A11 promoter H2A ubiquitination via an H2A deubiquitinase that functions as a tumor suppressor. As a result, ferroptosis is regulated and SLC7A11 expression is inhibited. The H2A ubiquitin ligases BAP1 and protein regulator of cytokinesis 1 inhibit expression of SLC7A11. The tumor suppressor deubiquitinating enzymes (DUB) inactivates P53 and BAP1 by downregulating SLC7A11 expression or enhancing OTUB1 or CD44 expression to stabilize SLC7A11 (153). As a result, tumor growth is suppressed and proteasomal activity and apoptosis are inhibited. The DUB inhibitor ubiquitin-specific protease (USP7) enhances caspase-dependent apoptosis during ferroptosis while degrading GPX4, which results in ubiquitinated protein accumulation and cell death (163). The expression of H2Bub1, which binds to the regulatory region of SLC7A11, is decreased by P53, thereby inhibiting SLC7A11 expression (164). Furthermore, ferritin degradation occurs via lysosomal or proteasomal mechanisms. Via ferritin heavy chain 1, NCOA4 binds to iron-rich ferritin on autophagosomes, thus transporting it to lysosomes for iron release. The ubiquitin ligase HERC2, which affects stability of proteins, ubiquitinates and degrades NCOA4 in environments with high iron concentrations. Therefore, ferroptosis is suppressed by inhibition of NCOA4 (165).

Histone acetylation and ferroptosis

The four aforementioned histones are acetylated on specific lysine residues, which is associated not only with gene transcription but also with DNA replication and repair (166). During transcription, histone acetylation induces HAT to acetylate lysine residues in histones, while histone deacetylation inhibits HDAC (167). HAT and HDAC catalyze histone acetylation and deacetylation, respectively (168). Patients with cancer have a poor prognosis when GPX4 expression is higher in tumor than in normal tissue (169). In tumor cells, high GPX4 levels may be due to epigenetic regulation such as methylation of DNA and histones and acetylation of histones, as reported in a previous study that analyzed upstream regulation of GPX4 (170). Mutant P533KR causes lipid peroxidation and ferroptosis via decreased acetylation and cysteine uptake (171). As a result of hydrolysis of cystinase, GSH is synthesized and GSH metabolism is affected. Cysteinase depletion also cause ferroptosis (Table I) (172).

Table I.

Epigenetic regulatory mechanisms associated with ferroptosis in cancer.

Table I.

Epigenetic regulatory mechanisms associated with ferroptosis in cancer.

A, DNA modification

Molecular mechanismEffect on ferroptosisFirst authorYear of publicationCountry(Refs.)
HELLS induce epigenetic silencing of the lncRNA LINC00472InhibitionMao et al,2018China(105)
EGLN1 and c-Myc in tumor cells activate LSH expression by inhibiting HIF-1InhibitionJiang et al,2017China(106)
DNA methylation modifier LSH interacts with WDR76 by activating lipid metabolism-related genes GLUT1, SCD1, and FADS2InhibitionJiang et al,2017China(106)
Inhibition of SCD1 leads to a decrease in coenzyme Q10 activityPromotionJiang et al,2017China(106)
Homocysteine treatment inducesPromotionZhang et al,2020China(107)
DNAmethylation of GPX4 gene in the nucleus pulposus
DNA hypermethylation of the CDH1 gene promoter suppresses expression of E-cadherinPromotionVan,2014Belgium(108)

B, Non-coding RNA

Molecular mechanismEffect on ferroptosisFirst authorYear of publicationCountry(Refs.)

miR-9 inhibits GOT1 and prevents the transamination of a-ketoglutarate results in accumulation of lipid ROSPromotionLande et al,2020India(117)
miR-137 inhibits SLC1A5 expressionInhibitionLuo et al,2018China(118)
miR-7-5p decreases ferrous iron levelsInhibitionTomita et al,2021Japan(119)
miR-4715-3p inhibits GPX4 expressionPromotionGomaa et al,2019USA(121)
Inhibition of AURKA expression inhibiting miR-4715-3p expression reduces depletion of GSH levelsInhibitionGomaa et al,2019USA(121)
miR-185 upregulates GPX2 expressionInhibitionZhuang et al,2020China(123)
lncRNA LINC00336 relates toInhibitionWang et al,2019China(129)
ELAVI 1 to decrease the intracellular Fe2+, ROS level
lncRNA UCA1 upregulates the expression of GSH and NADPH, enabling GPX4 to exert an antioxidant effect and to reduce the production of ROSInhibitionChen et al,2019China(132)
lncRNA PVT1 regulates miR-150/HIG2 axisPromotionXu et al,2018China(136)
Circ-TTBK2 regulates miR-761/ITGB8 axisInhibitionZhang et al,2020China(149)

C, Histone modification

Molecular mechanismEffect on ferroptosisFirst authorYear of publicationCountry(Refs.)

KDM3B induces H3K9me3 demethylation at the promoter region of SLC7A11 and promotes transcriptionInhibitionWang et al,2020China(156)
Overexpression OTUB1 increase SLC7A11 expressionInhibitionLiu et al,2019USA(162)
BAP1 and PRC1 reduces H2A ubiquitination of the SLC7A11 promoter and suppresses its expressionPromotionZhang et al,2019USA(153)
USP7 reduces GPX4 expressionPromotionDai et al,2020China(163)
Ubiquitin ligase HERC2 ubiquitinates and degrades NCOA4PromotionLin et al,2021China(165)
mutant P533KR inhibits cysteine uptake and reduces GSH, leading to lipid peroxidationPromotionJiang et al,2015USA(171)

Conclusion

Iron is a key element and its absorption, transport, storage and excretion are tightly regulated in the human body. Iron promotes formation of free radicals; excessive free radicals can be harm to the body by promoting tumor formation and subsequent metastasis. Study of iron metabolism in tumors has demonstrated that the combined actions of iron transporters and iron efflux regulator hepcidin promote tumorigenesis (173). Ferroptosis is an iron-dependent mechanism involving production of ROS. Mechanisms controlling transcripts that encode iron-regulated proteins, known as epigenetic regulatory mechanisms, have been reported in tumor cells (57). Furthermore, epigenetic changes in gene expression levels are not induced by changes in genomic sequences but DNA methylation, chromatin histone modifications and ncRNA regulation. Thus, abnormal epigenetic mechanisms affect gene transcription, which promotes tumor occurrence and development with certain generality and tissue specificity (174). Epigenetic modifications are key for tumor cell death and ferroptosis. The aforementioned findings may provide new therapeutic insights for the treatment of cancer in the future.

The present review summarizes the research of epigenetic inheritance and ferroptosis. The three main regulatory mechanisms of ferroptosis and ROS were introduced, with emphasis on the association between ROS and ferroptosis and epigenetic inheritance. The process of iron homeostasis metabolism and the association between iron homeostasis and reactive oxygen species were also described, which has not performed in the other two papers (175,176). In addition, the epigenetic regulation mechanism of ferroptosis was introduced, especially the association between ncRNA, miRNA and circRNA in ferroptosis and histone ubiquitination, methylation and acetylation with ferroptosis. The aforementioned findings may provide novel options for the treatment of tumors in terms of epigenetic regulation of ferroptosis (177).

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

JWu, SZ and PW wrote the manuscript. JWa, JH, TW and LG constructed figures and tables. DL, QM and HP performed the literature review. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Subramanian S, Geng H and Tan XD: Cell death of intestinal epithelial cells in intestinal diseases. Sheng Li Xue Bao. 72:308–324. 2020.PubMed/NCBI

3 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Su LJ, Zhang JH, Gomez H, Murugan R, Hong X, Xu D, Jiang F and Peng ZY: Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid Med Cell Longev. 2019:50808432019. View Article : Google Scholar : PubMed/NCBI

5 

D'Arcy MS: Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 43:582–592. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H and Vandenabeele P: Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 15:135–147. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Hirschhorn T and Stockwell BR: The development of the concept of ferroptosis. Free Radic Biol Med. 133:130–143. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Zhang X, Sui S, Wang L, Li H, Zhang L, Xu S and Zheng X: Inhibition of tumor propellant glutathione peroxidase 4 induces ferroptosis in cancer cells and enhances anticancer effect of cisplatin. J Cell Physiol. 235:3425–3437. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Deans C and Maggert KA: What do you mean, ‘epigenetic’? Genetics. 199:887–896. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Zhu J, Xiong Y, Zhang Y, Wen J, Cai N, Cheng K, Liang H and Zhang W: The molecular mechanisms of regulating oxidative stress-induced ferroptosis and therapeutic strategy in tumors. Oxid Med Cell Longev. 2020:88107852020. View Article : Google Scholar : PubMed/NCBI

11 

Wei Y, Lv H, Shaikh AB, Han W, Hou H, Zhang Z, Wang S and Shang P: Directly targeting glutathione peroxidase 4 may be more effective than disrupting glutathione on ferroptosis-based cancer therapy. Biochim Biophys Acta Gen Subj. 1864:1295392020. View Article : Google Scholar : PubMed/NCBI

12 

Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, Zhu N, Yin X, et al: Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 116:2672–2680. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Bogdan AR, Miyazawa M, Hashimoto K and Tsuji Y: Regulators of iron homeostasis: New players in metabolism, cell death, and disease. Trends Biochem Sci. 41:274–286. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Anderson CP, Shen M, Eisenstein RS and Leibold EA: Mammalian iron metabolism and its control by iron regulatory proteins. Biochim Biophys Acta. 1823:1468–1483. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Kato J, Kobune M, Ohkubo S, Fujikawa K, Tanaka M, Takimoto R, Takada K, Takahari D, Kawano Y, Kohgo Y and Niitsu Y: Iron/IRP-1-dependent regulation of mRNA expression for transferrin receptor, DMT1 and ferritin during human erythroid differentiation. Exp Hematol. 35:879–887. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Bolotta A, Abruzzo PM, Baldassarro VA, Ghezzo A, Scotlandi K, Marini M and Zucchini C: New insights into the hepcidin-ferroportin axis and iron homeostasis in iPSC-derived cardiomyocytes from friedreich's ataxia patient. Oxid Med Cell Longev. 2019:76230232019. View Article : Google Scholar : PubMed/NCBI

17 

Gao G, Li J, Zhang Y and Chang YZ: Cellular iron metabolism and regulation. Adv Exp Med Biol. 1173:21–32. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Arruda SF, Ramos LV, Barbosa JLA, Hankins NAC, Rodrigues PAM and Cunha MSBD: The action of JAK/STAT3 and BMP/HJV/SMAD signaling pathways on hepcidin suppression by tucum-do-cerrado in a normal and iron-enriched diets. Nutrients. 12:15152020. View Article : Google Scholar : PubMed/NCBI

19 

Babitt JL, Huang FW, Wrighting DM, Xia Y, Sidis Y, Samad TA, Campagna JA, Chung RT, Schneyer AL, Woolf CJ, et al: Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression. Nat Genet. 38:531–539. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Yu PB, Hong CC, Sachidanandan C, Babitt JL, Deng DY, Hoyng SA, Lin HY, Bloch KD and Peterson RT: Dorsomorphin inhibits BMP signals required for embryogenesis and iron metabolism. Nat Chem Biol. 4:33–41. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Truksa J, Lee P and Beutler E: Two BMP responsive elements, STAT, and bZIP/HNF4/COUP motifs of the hepcidin promoter are critical for BMP, SMAD1, and HJV responsiveness. Blood. 113:688–695. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Srole DN and Ganz T: Erythroferrone structure, function, and physiology: Iron homeostasis and beyond. J Cell Physiol. 236:4888–4901. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Duan L, Yin X, Meng H, Fang X, Min J and Wang F: Progress on epigenetic regulation of iron homeostasis. Zhejiang Da Xue Xue Bao Yi Xue Ban. 49:58–70. 2020.(In Chinese). PubMed/NCBI

24 

Patnaik MM and Tefferi A: Myelodysplastic syndromes with ring sideroblasts (MDS-RS) and MDS/myeloproliferative neoplasm with RS and thrombocytosis (MDS/MPN-RS-T)-‘2021 update on diagnosis, risk-stratification, and management’. Am J Hematol. 96:379–394. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Rawat PS, Jaiswal A, Khurana A, Bhatti JS and Navik U: Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed Pharmacother. 139:1117082021. View Article : Google Scholar : PubMed/NCBI

26 

Blackledge NP and Klose R: CpG island chromatin: A platform for gene regulation. Epigenetics. 6:147–152. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Illingworth RS, Gruenewald-Schneider U, Webb S, Kerr AR, James KD, Turner DJ, Smith C, Harrison DJ, Andrews R and Bird AP: Orphan CpG islands identify numerous conserved promoters in the mammalian genome. PLoS Genet. 6:e10011342010. View Article : Google Scholar : PubMed/NCBI

28 

Grand RS, Burger L, Gräwe C, Michael AK, Isbel L, Hess D, Hoerner L, Iesmantavicius V, Durdu S, Pregnolato M, et al: BANP opens chromatin and activates CpG-island-regulated genes. Nature. 596:133–137. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Maunakea AK, Nagarajan RP, Bilenky M, Ballinger TJ, D'Souza C, Fouse SD, Johnson BE, Hong C, Nielsen C, Zhao Y, et al: Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 466:253–257. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Horvath S and Raj K: DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet. 19:371–384. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Ye Q, Trivedi M, Zhang Y, Böhlke M, Alsulimani H, Chang J, Maher T, Deth R and Kim J: Brain iron loading impairs DNA methylation and alters GABAergic function in mice. FASEB J. 33:2460–2471. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Macková E, Hrušková K, Bendová P, Vávrová A, Jansová H, Hašková P, Kovaříková P, Vávrová K and Simůnek T: Methyl and ethyl ketone analogs of salicylaldehyde isonicotinoyl hydrazone: Novel iron chelators with selective antiproliferative action. Chem Biol Interact. 197:69–79. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Yang WC, Lin SF, Wang SC, Tsai WC, Wu CC and Wu SC: The effects of human BDH2 on the cell cycle, differentiation, and apoptosis and associations with leukemia transformation in myelodysplastic syndrome. Int J Mol Sci. 21:30332020. View Article : Google Scholar : PubMed/NCBI

34 

Ma S, Henson ES, Chen Y and Gibson SB: Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 7:e23072016. View Article : Google Scholar : PubMed/NCBI

35 

Kajarabille N and Latunde-Dada GO: Programmed cell-death by ferroptosis: Antioxidants as mitigators. Int J Mol Sci. 20:49682019. View Article : Google Scholar : PubMed/NCBI

36 

Gao J, Luo T and Wang J: Gene interfered-ferroptosis therapy for cancers. Nat Commun. 12:53112021. View Article : Google Scholar : PubMed/NCBI

37 

Wang F, Wang J, Shen Y, Li H, Rausch WD and Huang X: Iron dyshomeostasis and ferroptosis: A new Alzheimer's disease hypothesis? Front Aging Neurosci. 14:8305692022. View Article : Google Scholar : PubMed/NCBI

38 

Chen Y, Zhang S, Wang X, Guo W, Wang L, Zhang D, Yuan L, Zhang Z, Xu Y and Liu S: Disordered signaling governing ferroportin transcription favors breast cancer growth. Cell Signal. 27:168–176. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Udali S, Castagna A, Corbella M, Ruzzenente A, Moruzzi S, Mazzi F, Campagnaro T, De Santis D, Franceschi A, Pattini P, et al: Hepcidin and DNA promoter methylation in hepatocellular carcinoma. Eur J Clin Invest. 48:e128702018. View Article : Google Scholar : PubMed/NCBI

40 

Al-Amer O and Alsharif KF: Frequency of the HAMP (c.-582 A>G) polymorphism in iron deficiency in Saudi Arabia. Pak J Biol Sci. 24:146–150. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Sies H: Oxidative stress: A concept in redox biology and medicine. Redox Biol. 4:180–183. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Tang Z, Liu Y, He M and Bu W: Chemodynamic therapy: Tumour microenvironment-mediated fenton and fenton-like reactions. Angew Chem Int Ed Engl. 58:946–956. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Miller CJ, Rose AL and Waite TD: Importance of iron complexation for fenton-mediated hydroxyl radical production at circumneutral pH. Front Mar Sci. 3:1342016. View Article : Google Scholar

44 

Yu L, Lin Z, Cheng X, Chu J, Li X, Chen C, Zhu T, Li W, Lin W and Tang W: Thorium inhibits human respiratory chain complex IV (cytochrome c oxidase). J Hazard Mater. 424:1275462022. View Article : Google Scholar : PubMed/NCBI

45 

Rattanawong K, Koiso N, Toda E, Kinoshita A, Tanaka M, Tsuji H and Okamoto T: Regulatory functions of ROS dynamics via glutathione metabolism and glutathione peroxidase activity in developing rice zygote. Plant J. 108:1097–1115. 2021. View Article : Google Scholar : PubMed/NCBI

46 

Peoples JN, Saraf A, Ghazal N, Pham TT and Kwong JQ: Mitochondrial dysfunction and oxidative stress in heart disease. Exp Mol Med. 51:1–13. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Prasad S, Gupta SC and Tyagi AK: Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 387:95–105. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Zhang R, Kang KA, Kim KC, Na SY, Chang WY, Kim GY, Kim HS and Hyun JW: Oxidative stress causes epigenetic alteration of CDX1 expression in colorectal cancer cells. Gene. 524:214–219. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Yang Y, Karakhanova S, Hartwig W, D'Haese JG, Philippov PP, Werner J and Bazhin AV: Mitochondria and mitochondrial ROS in cancer: Novel targets for anticancer therapy. J Cell Physiol. 231:2570–2581. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Islam MT: Oxidative stress and mitochondrial dysfunction-linked neurodegenerative disorders. Neurol Res. 39:73–82. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Zhao Y, Zhang J, Zheng Y, Zhang Y, Zhang XJ, Wang H, Du Y, Guan J, Wang X and Fu J: NAD+ improves cognitive function and reduces neuroinflammation by ameliorating mitochondrial damage and decreasing ROS production in chronic cerebral hypoperfusion models through Sirt1/PGC-1α pathway. J Neuroinflammation. 18:2072021. View Article : Google Scholar : PubMed/NCBI

52 

Uittenbogaard M, Baxter KK and Chiaramello A: The neurogenic basic helix-loop-helix transcription factor NeuroD6 confers tolerance to oxidative stress by triggering an antioxidant response and sustaining the mitochondrial biomass. ASN Neuro. 2:e000342010. View Article : Google Scholar : PubMed/NCBI

53 

Hou K, Chen Y, Zhu D, Chen G, Chen F, Xu N, Barakat K, Zheng J, Xie X and Chen R: Curcumin inhibits high glucose oxidative stress and apoptosis in pancreatic beta cells via CHOP/PCG-1a and pERK1/2. Front Biosci (Landmark Ed). 25:1974–1984. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Tang G, Li S, Zhang C, Chen H, Wang N and Feng Y: Clinical efficacies, underlying mechanisms and molecular targets of Chinese medicines for diabetic nephropathy treatment and management. Acta Pharm Sin B. 11:2749–2767. 2021. View Article : Google Scholar : PubMed/NCBI

55 

Su X, Chu Y, Kordower JH, Li B, Cao H, Huang L, Nishida M, Song L, Wang D and Federoff HJ: PGC-1α promoter methylation in Parkinson's disease. PLoS One. 10:e01340872015. View Article : Google Scholar : PubMed/NCBI

56 

Li Y, Chen M, Xu Y, Yu X, Xiong T, Du M, Sun J, Liu L, Tang Y and Yao P: Iron-mediated lysosomal membrane permeabilization in ethanol-induced hepatic oxidative damage and apoptosis: Protective effects of quercetin. Oxid Med Cell Longev. 2016:41476102016.PubMed/NCBI

57 

Park E and Chung SW: ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 10:8222019. View Article : Google Scholar : PubMed/NCBI

58 

Chen H, Xu C, Yu Q, Zhong C, Peng Y, Chen J and Chen G: Comprehensive landscape of STEAP family functions and prognostic prediction value in glioblastoma. J Cell Physiol. 236:2988–3000. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Song Q, Peng S, Sun Z, Heng X and Zhu X: Temozolomide drives ferroptosis via a DMT1-dependent pathway in glioblastoma cells. Yonsei Med J. 62:843–849. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Cheung EC and Vousden KH: The role of ROS in tumour development and progression. Nat Rev Cancer. 22:280–297. 2022. View Article : Google Scholar : PubMed/NCBI

61 

Wei S, Qiu T, Yao X, Wang N, Jiang L, Jia X, Tao Y, Wang Z, Pei P, Zhang J, et al: Arsenic induces pancreatic dysfunction and ferroptosis via mitochondrial ROS-autophagy-lysosomal pathway. J Hazard Mater. 384:1213902020. View Article : Google Scholar : PubMed/NCBI

62 

Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G and Tang D: Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol. 66:89–100. 2020. View Article : Google Scholar : PubMed/NCBI

63 

Cao JY and Dixon SJ: Mechanisms of ferroptosis. Cell Mol Life Sci. 73:2195–2209. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Zhuang Y, Li T, Xiao H, Wu J, Su S, Dong X, Hu X, Hua Q, Liu J, Shang W, et al: LncRNA-H19 drives cardiomyocyte senescence by targeting miR-19a/socs1/p53 axis. Front Pharmacol. 12:6318352021. View Article : Google Scholar : PubMed/NCBI

65 

Mou Y, Wang J, Wu J, He D, Zhang C, Duan C and Li B: Ferroptosis, a new form of cell death: Opportunities and challenges in cancer. J Hematol Oncol. 12:342019. View Article : Google Scholar : PubMed/NCBI

66 

Koppula P, Zhuang L and Gan B: Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 12:599–620. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Zhang H, He Y, Wang JX, Chen MH, Xu JJ, Jiang MH, Feng YL and Gu YF: miR-30-5p-mediated ferroptosis of trophoblasts is implicated in the pathogenesis of preeclampsia. Redox Biol. 29:1014022020. View Article : Google Scholar : PubMed/NCBI

68 

Liu MR, Zhu WT and Pei DS: System Xc: A key regulatory target of ferroptosis in cancer. Invest New Drugs. 39:1123–1131. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, Ding HF, Zhang J, Wang H, Chen X and Yan C: ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 27:662–675. 2020. View Article : Google Scholar : PubMed/NCBI

70 

Kong R, Wang N, Han W, Bao W and Lu J: IFNγ-mediated repression of system xc drives vulnerability to induced ferroptosis in hepatocellular carcinoma cells. J Leukoc Biol. 110:301–314. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Floros KV, Cai J, Jacob S, Kurupi R, Fairchild CK, Shende M, Coon CM, Powell KM, Belvin BR, Hu B, et al: MYCN-amplified neuroblastoma is addicted to iron and vulnerable to inhibition of the system Xc-/glutathione axis. Cancer Res. 81:1896–1908. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, Wang M, Chen Y and Zhang Q: Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 14:192021. View Article : Google Scholar : PubMed/NCBI

73 

Imai H, Matsuoka M, Kumagai T, Sakamoto T and Koumura T: Lipid peroxidation-dependent cell death regulated by GPx4 and ferroptosis. Curr Top Microbiol Immunol. 403:143–170. 2017.PubMed/NCBI

74 

Li H, Liu W, Zhang X, Wu F, Sun D and Wang Z: Ketamine suppresses proliferation and induces ferroptosis and apoptosis of breast cancer cells by targeting KAT5/GPX4 axis. Biochem Biophys Res Commun. 585:111–116. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Tang X, Ding H, Liang M, Chen X, Yan Y, Wan N, Chen Q, Zhang J and Cao J: Curcumin induces ferroptosis in non-small-cell lung cancer via activating autophagy. Thorac Cancer. 12:1219–1230. 2021. View Article : Google Scholar : PubMed/NCBI

76 

Zhang Y, Kong Y, Ma Y, Ni S, Wikerholmen T, Xi K, Zhao F, Zhao Z, Wang J, Huang B, et al: Loss of COPZ1 induces NCOA4 mediated autophagy and ferroptosis in glioblastoma cell lines. Oncogene. 40:1425–1439. 2021. View Article : Google Scholar : PubMed/NCBI

77 

Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al: The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 575:688–692. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Wang CX, Chen LH, Zhuang HB, Shi ZS, Chen ZC, Pan JP and Hong ZS: Auriculasin enhances ROS generation to regulate colorectal cancer cell apoptosis, ferroptosis, oxeiptosis, invasion and colony formation. Biochem Biophys Res Commun. 587:99–106. 2022. View Article : Google Scholar : PubMed/NCBI

79 

Santoro MM: The antioxidant role of non-mitochondrial CoQ10: mystery solved! Cell Metab. 31:13–15. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Goya Grocin A, Xavier da Silva TN, Panzilius E, Scheel CH, et al: FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 575:693–698. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Zheng J and Conrad M: The metabolic underpinnings of ferroptosis. Cell Metab. 32:920–937. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Hu Q, Wei W, Wu D, Huang F, Li M, Li W, Yin J, Peng Y, Lu Y, Zhao Q and Liu L: Blockade of GCH1/BH4 axis activates ferritinophagy to mitigate the resistance of colorectal cancer to erastin-induced ferroptosis. Front Cell Dev Biol. 10:8103272022. View Article : Google Scholar : PubMed/NCBI

83 

Qiu C, Liu T, Luo D, Luan D, Cheng L and Wang S: Novel therapeutic savior for osteosarcoma: The endorsement of ferroptosis. Front Oncol. 12:7460302022. View Article : Google Scholar : PubMed/NCBI

84 

Gao M, Fan K, Chen Y, Zhang G, Chen J and Zhang Y: Understanding the mechanistic regulation of ferroptosis in cancer: The gene matters. J Genet Genomics. S1673-8527(22)00160-6. 2022.(Epub ahead of print). View Article : Google Scholar

85 

Kraft VAN, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, Merl-Pham J, Bao X, Anastasov N, Kössl J, et al: GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 6:41–53. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Chew GT and Watts GF: Coenzyme Q10 and diabetic endotheliopathy: Oxidative stress and the ‘recoupling hypothesis’. QJM. 97:537–548. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Ilango S, Paital B, Jayachandran P, Padma PR and Nirmaladevi R: Epigenetic alterations in cancer. Front Biosci (Landmark Ed). 25:1058–1109. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Wu SC and Zhang Y: Active DNA demethylation: Many roads lead to Rome. Nat Rev Mol Cell Biol. 11:607–620. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Goll MG and Bestor TH: Eukaryotic cytosine methyltransferases. Annu Rev Biochem. 74:481–514. 2005. View Article : Google Scholar : PubMed/NCBI

90 

Lu F, Liu Y, Jiang L, Yamaguchi S and Zhang Y: Role of Tet proteins in enhancer activity and telomere elongation. Genes Dev. 28:2103–2119. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Jiang J, Yan T and Guo F: Global DNA 5hmC and CK195hmC+ contents: A promising biomarker for predicting prognosis in small hepatocellular carcinoma. Curr Oncol. 28:3758–3770. 2021. View Article : Google Scholar : PubMed/NCBI

92 

Ito S, Shen L, Dai Q, Wu SC, Collins LB, Swenberg JA, He C and Zhang Y: Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science. 333:1300–1303. 2011. View Article : Google Scholar : PubMed/NCBI

93 

Hashimoto H, Vertino PM and Cheng X: Molecular coupling of DNA methylation and histone methylation. Epigenomics. 2:657–669. 2010. View Article : Google Scholar : PubMed/NCBI

94 

Jara-Espejo M and Line SR: DNA G-quadruplex stability, position and chromatin accessibility are associated with CpG island methylation. FEBS J. 287:483–495. 2020. View Article : Google Scholar : PubMed/NCBI

95 

Horii T and Hatada I: Regulation of CpG methylation by Dnmt and Tet in pluripotent stem cells. J Reprod Dev. 62:331–335. 2016. View Article : Google Scholar : PubMed/NCBI

96 

Van Tongelen A, Loriot A and De Smet C: Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes. Cancer Lett. 396:130–137. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Klutstein M, Nejman D, Greenfield R and Cedar H: DNA methylation in cancer and aging. Cancer Res. 76:3446–3450. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Jair KW, Bachman KE, Suzuki H, Ting AH, Rhee I, Yen RW, Baylin SB and Schuebel KE: De novo CpG island methylation in human cancer cells. Cancer Res. 66:682–692. 2006. View Article : Google Scholar : PubMed/NCBI

99 

Mendoza-Pérez J, Gu J, Herrera LA, Tannir NM, Matin SF, Karam JA, Huang M, Chang DW, Wood CG and Wu X: Genomic DNA hypomethylation and risk of renal cell carcinoma: A case-control study. Clin Cancer Res. 22:2074–2082. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Zhang L, Xiao X and Xu ZC: iPromoter-5mC: A novel fusion decision predictor for the identification of 5-methylcytosine sites in genome-wide DNA promoters. Front Cell Dev Biol. 8:6142020. View Article : Google Scholar : PubMed/NCBI

101 

Robert MF, Morin S, Beaulieu N, Gauthier F, Chute IC, Barsalou A and MacLeod AR: DNMT1 is required to maintain CpG methylation and aberrant gene silencing in human cancer cells. Nat Genet. 33:61–65. 2003. View Article : Google Scholar : PubMed/NCBI

102 

Choi SJ, Jung SW, Huh S, Chung YS, Cho H and Kang H: Alteration of DNA methylation in gastric cancer with chemotherapy. J Microbiol Biotechnol. 27:1367–1378. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Logie E, Van Puyvelde B, Cuypers B, Schepers A, Berghmans H, Verdonck J, Laukens K, Godderis L, Dhaenens M, Deforce D and Vanden Berghe W: Ferroptosis induction in multiple myeloma cells triggers DNA methylation and histone modification changes associated with cellular senescence. Int J Mol Sci. 22:122342021. View Article : Google Scholar : PubMed/NCBI

104 

Xu Y, Hong M, Kong D, Deng J, Zhong Z and Liang J: Ferroptosis-associated DNA methylation signature predicts overall survival in patients with head and neck squamous cell carcinoma. BMC Genomics. 23:632022. View Article : Google Scholar : PubMed/NCBI

105 

Mao C, Wang X, Liu Y, Wang M, Yan B, Jiang Y, Shi Y, Shen Y, Liu X, Lai W, et al: A G3BP1-interacting lncRNA promotes ferroptosis and apoptosis in cancer via nuclear sequestration of p53. Cancer Res. 78:3484–3496. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Jiang Y, Mao C, Yang R, Yan B, Shi Y, Liu X, Lai W, Liu Y, Wang X, Xiao D, et al: EGLN1/c-Myc induced lymphoid-specific helicase inhibits ferroptosis through lipid metabolic gene expression changes. Theranostics. 7:3293–3305. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Zhang X, Huang Z, Xie Z, Chen Y, Zheng Z, Wei X, Huang B, Shan Z, Liu J, Fan S, et al: Homocysteine induces oxidative stress and ferroptosis of nucleus pulposus via enhancing methylation of GPX4. Free Radic Biol Med. 160:552–565. 2020. View Article : Google Scholar : PubMed/NCBI

108 

van Roy F: Beyond E-cadherin: Roles of other cadherin superfamily members in cancer. Nat Rev Cancer. 14:121–134. 2014. View Article : Google Scholar : PubMed/NCBI

109 

Tan L and Shi YG: Tet family proteins and 5-hydroxymethylcytosine in development and disease. Development. 139:1895–1902. 2012. View Article : Google Scholar : PubMed/NCBI

110 

Tang Y, Li C, Zhang YJ and Wu ZH: Ferroptosis-related long non-coding RNA signature predicts the prognosis of Head and neck squamous cell carcinoma. Int J Biol Sci. 17:702–711. 2021. View Article : Google Scholar : PubMed/NCBI

111 

Wang J, Zhu S, Meng N, He Y, Lu R and Yan GR: ncRNA-encoded peptides or proteins and cancer. Mol Ther. 27:1718–1725. 2019. View Article : Google Scholar : PubMed/NCBI

112 

Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, Zhang Q, Lin D, Ge S, Bai M, et al: CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 19:432020. View Article : Google Scholar : PubMed/NCBI

113 

Hill M and Tran N: miRNA interplay: Mechanisms and consequences in cancer. Dis Model Mech. 14:dmm0476622021. View Article : Google Scholar : PubMed/NCBI

114 

Mishra S, Yadav T and Rani V: Exploring miRNA based approaches in cancer diagnostics and therapeutics. Crit Rev Oncol Hematol. 98:12–23. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Sengupta D, Deb M, Kar S, Pradhan N, Parbin S, Kirtana R, Singh SP, Suma SG, Niharika, Roy A, et al: Dissecting miRNA facilitated physiology and function in human breast cancer for therapeutic intervention. Semin Cancer Biol. 72:46–64. 2021. View Article : Google Scholar : PubMed/NCBI

116 

Lee YS and Dutta A: MicroRNAs in cancer. Annu Rev Pathol. 4:199–227. 2009. View Article : Google Scholar : PubMed/NCBI

117 

Lande K, Gupta J, Ranjan R, Kiran M, Torres Solis LF, Solís Herrera A, Aliev G and Karnati R: Exosomes: Insights from retinoblastoma and other eye cancers. Int J Mol Sci. 21:70552020. View Article : Google Scholar : PubMed/NCBI

118 

Luo M, Wu L, Zhang K, Wang H, Zhang T, Gutierrez L, O'Connell D, Zhang P, Li Y, Gao T, et al: miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 25:1457–1472. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Tomita K, Nagasawa T, Kuwahara Y, Torii S, Igarashi K, Roudkenar MH, Roushandeh AM, Kurimasa A and Sato T: MiR-7-5p is involved in ferroptosis signaling and radioresistance thru the generation of ROS in radioresistant HeLa and SAS cell lines. Int J Mol Sci. 22:83002021. View Article : Google Scholar : PubMed/NCBI

120 

Chen D, Fan Z, Rauh M, Buchfelder M, Eyupoglu IY and Savaskan N: ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene. 36:5593–5608. 2017. View Article : Google Scholar : PubMed/NCBI

121 

Gomaa A, Peng D, Chen Z, Soutto M, Abouelezz K, Corvalan A and El-Rifai W: Epigenetic regulation of AURKA by miR-4715-3p in upper gastrointestinal cancers. Sci Rep. 9:169702019. View Article : Google Scholar : PubMed/NCBI

122 

Yu W, Yao J, Lyu P, Zhou J, Chen X, Liu X and Xiao S: XPG is modulated by miR-4715-3p and rs873601 genotypes in lung cancer. Cancer Manag Res. 13:3417–3427. 2021. View Article : Google Scholar : PubMed/NCBI

123 

Zhuang ST, Cai YJ, Liu HP, Qin Y and Wen JF: LncRNA NEAT1/miR-185-5p/IGF2 axis regulates the invasion and migration of colon cancer. Mol Genet Genomic Med. 8:e11252020. View Article : Google Scholar : PubMed/NCBI

124 

Marengo B, Pulliero A, Izzotti A and Domenicotti C: miRNA regulation of glutathione homeostasis in cancer initiation, progression and therapy resistance. Microrna. 9:187–197. 2020. View Article : Google Scholar : PubMed/NCBI

125 

Hu X, Miao J, Zhang M, Wang X, Wang Z, Han J, Tong D and Huang C: miRNA-103a-3p promotes human gastric cancer cell proliferation by targeting and suppressing ATF7 in vitro. Mol Cells. 41:390–400. 2018.PubMed/NCBI

126 

Zhi Y, Gao L, Wang B, Ren W, Liang KX and Zhi K: Ferroptosis holds novel promise in treatment of cancer mediated by non-coding RNAs. Front Cell Dev Biol. 9:6869062021. View Article : Google Scholar : PubMed/NCBI

127 

Bridges MC, Daulagala AC and Kourtidis A: LNCcation: lncRNA localization and function. J Cell Biol. 220:e2020090452021. View Article : Google Scholar : PubMed/NCBI

128 

Peng WX, Koirala P and Mo YY: LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar : PubMed/NCBI

129 

Wang M, Mao C, Ouyang L, Liu Y, Lai W, Liu N, Shi Y, Chen L, Xiao D, Yu F, et al: Long noncoding RNA LINC00336 inhibits ferroptosis in lung cancer by functioning as a competing endogenous RNA. Cell Death Differ. 26:2329–2343. 2019. View Article : Google Scholar : PubMed/NCBI

130 

Wang Y, Li X, Chen W and Wu W: The common region of lncRNAs UCA1 and UCA1α contributes to the bladder cancer tumorigenesis. Eur J Cancer Prev. 30:389–392. 2021. View Article : Google Scholar : PubMed/NCBI

131 

Kim S, Kim JE, Kim YH, Hwang T, Kim SK, Xu WJ, Shin JY, Kim JI, Choi H, Kim HC, et al: Glutaminase 2 expression is associated with regional heterogeneity of 5-aminolevulinic acid fluorescence in glioblastoma. Sci Rep. 7:122212017. View Article : Google Scholar : PubMed/NCBI

132 

Chen J, Hu Q, Zhang BF, Liu XP, Yang S and Jiang H: Long noncoding RNA UCA1 inhibits ischaemia/reperfusion injury induced cardiomyocytes apoptosis via suppression of endoplasmic reticulum stress. Genes Genomics. 41:803–810. 2019. View Article : Google Scholar : PubMed/NCBI

133 

Shin D, Kim EH, Lee J and Roh JL: Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med. 129:454–462. 2018. View Article : Google Scholar : PubMed/NCBI

134 

Yang Y, Tian Q, Wu S, Li Y, Yang K, Yan Y, Shang L, Li A and Zhang L: Blue light-triggered Fe2+-release from monodispersed ferrihydrite nanoparticles for cancer iron therapy. Biomaterials. 271:1207392021. View Article : Google Scholar : PubMed/NCBI

135 

Xie Z, Hou H, Luo D, An R, Zhao Y and Qiu C: ROS-dependent lipid peroxidation and reliant antioxidant ferroptosis-suppressor-protein 1 in rheumatoid arthritis: A covert clue for potential therapy. Inflammation. 44:35–47. 2021. View Article : Google Scholar : PubMed/NCBI

136 

Xu Y, Luo X, He W, Chen G, Li Y, Li W, Wang X, Lai Y and Ye Y: Long non-coding RNA PVT1/miR-150/HIG2 axis regulates the proliferation, invasion and the balance of iron metabolism of hepatocellular carcinoma. Cell Physiol Biochem. 49:1403–1419. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Zheng J, Zhou Z, Qiu Y, Wang M, Yu H, Wu Z, Wang X and Jiang X: A prognostic ferroptosis-related lncRNAs signature associated with immune landscape and radiotherapy response in glioma. Front Cell Dev Biol. 9:6755552021. View Article : Google Scholar : PubMed/NCBI

138 

Qi W, Li Z, Xia L, Dai J, Zhang Q, Wu C and Xu S: LncRNA GABPB1-AS1 and GABPB1 regulate oxidative stress during erastin-induced ferroptosis in HepG2 hepatocellular carcinoma cells. Sci Rep. 9:161852019. View Article : Google Scholar : PubMed/NCBI

139 

Hu T, Zhang C, Tang Q, Su Y, Li B, Chen L, Zhang Z, Cai T and Zhu Y: Variant G6PD levels promote tumor cell proliferation or apoptosis via the STAT3/5 pathway in the human melanoma xenograft mouse model. BMC Cancer. 13:2512013. View Article : Google Scholar : PubMed/NCBI

140 

Su X, Yang Y, Yang Q, Pang B, Sun S, Wang Y, Qiao Q, Guo C, Liu H and Pang Q: NOX4-derived ROS-induced overexpression of FOXM1 regulates aerobic glycolysis in glioblastoma. BMC Cancer. 21:11812021. View Article : Google Scholar : PubMed/NCBI

141 

Wang Z, Ding Y, Wang X, Lu S, Wang C, He C, Wang L, Piao M, Chi G, Luo Y and Ge P: Pseudolaric acid B triggers ferroptosis in glioma cells via activation of Nox4 and inhibition of xCT. Cancer Lett. 428:21–33. 2018. View Article : Google Scholar : PubMed/NCBI

142 

Chen B, Wang H, Lv C, Mao C and Cui Y: Long non-coding RNA H19 protects against intracerebral hemorrhage injuries via regulating microRNA-106b-5p/acyl-CoA synthetase long chain family member 4 axis. Bioengineered. 12:4004–4015. 2021. View Article : Google Scholar : PubMed/NCBI

143 

He GN, Bao NR, Wang S, Xi M, Zhang TH and Chen FS: Ketamine induces ferroptosis of liver cancer cells by targeting lncRNA PVT1/miR-214-3p/GPX4. Drug Des Devel Ther. 15:3965–3978. 2021. View Article : Google Scholar : PubMed/NCBI

144 

Yang Y, Tai W, Lu N, Li T, Liu Y, Wu W, Li Z, Pu L, Zhao X, Zhang T and Dong Z: lncRNA ZFAS1 promotes lung fibroblast-to-myofibroblast transition and ferroptosis via functioning as a ceRNA through miR-150-5p/SLC38A1 axis. Aging (Albany NY). 12:9085–9102. 2020. View Article : Google Scholar : PubMed/NCBI

145 

Chen L, Wang C, Sun H, Wang J, Liang Y, Wang Y and Wong G: The bioinformatics toolbox for circRNA discovery and analysis. Brief Bioinform. 22:1706–1728. 2021. View Article : Google Scholar : PubMed/NCBI

146 

Wang J, Zhao X, Wang Y, Ren F, Sun D, Yan Y, Kong X, Bu J, Liu M and Xu S: circRNA-002178 act as a ceRNA to promote PDL1/PD1 expression in lung adenocarcinoma. Cell Death Dis. 11:322020. View Article : Google Scholar : PubMed/NCBI

147 

Li R, Jiang J, Shi H, Qian H, Zhang X and Xu W: CircRNA: A rising star in gastric cancer. Cell Mol Life Sci. 77:1661–1680. 2020. View Article : Google Scholar : PubMed/NCBI

148 

Liu Z, Wang Q, Wang X, Xu Z, Wei X and Li J: Circular RNA cIARS regulates ferroptosis in HCC cells through interacting with RNA binding protein ALKBH5. Cell Death Discov. 6:722020. View Article : Google Scholar : PubMed/NCBI

149 

Zhang HY, Zhang BW, Zhang ZB and Deng QJ: Circular RNA TTBK2 regulates cell proliferation, invasion and ferroptosis via miR-761/ITGB8 axis in glioma. Eur Rev Med Pharmacol Sci. 24:2585–2600. 2020.PubMed/NCBI

150 

Zhang S, Sun J, Gu M, Wang G and Wang X: Circular RNA: A promising new star for the diagnosis and treatment of colorectal cancer. Cancer Med. 10:8725–8740. 2021. View Article : Google Scholar : PubMed/NCBI

151 

Qiao G, Zhang W and Dong K: Regulation of ferroptosis by noncoding RNAs: A novel promise treatment in esophageal squamous cell carcinoma. Mol Cell Biochem. 477:2193–2202. 2022. View Article : Google Scholar : PubMed/NCBI

152 

Kinoshita C and Aoyama K: The role of non-coding RNAs in the neuroprotective effects of glutathione. Int J Mol Sci. 22:42452021. View Article : Google Scholar : PubMed/NCBI

153 

Zhang Y, Koppula P and Gan B: Regulation of H2A ubiquitination and SLC7A11 expression by BAP1 and PRC1. Cell Cycle. 18:773–783. 2019. View Article : Google Scholar : PubMed/NCBI

154 

Wang X, Liu K, Gong H, Li D, Chu W, Zhao D, Wang X and Xu D: Death by histone deacetylase inhibitor quisinostat in tongue squamous cell carcinoma via apoptosis, pyroptosis, and ferroptosis. Toxicol Appl Pharmacol. 410:1153632021. View Article : Google Scholar : PubMed/NCBI

155 

He C, Wang C, Liu H and Shan B: Kayadiol exerted anticancer effects through p53-mediated ferroptosis in NKTCL cells. BMC Cancer. 22:7242022. View Article : Google Scholar : PubMed/NCBI

156 

Wang Y, Zhao Y, Wang H, Zhang C, Wang M, Yang Y, Xu X and Hu Z: Histone demethylase KDM3B protects against ferroptosis by upregulating SLC7A11. FEBS Open Bio. 10:637–643. 2020. View Article : Google Scholar : PubMed/NCBI

157 

Liu J, Kang R and Tang D: Signaling pathways and defense mechanisms of ferroptosis. FEBS J. Jun 6–2021.(Epub ahead of print). View Article : Google Scholar

158 

Chen X, Kang R, Kroemer G and Tang D: Broadening horizons: The role of ferroptosis in cancer. Nat Rev Clin Oncol. 18:280–296. 2021. View Article : Google Scholar : PubMed/NCBI

159 

Kang R, Kroemer G and Tang D: The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med. 133:162–168. 2019. View Article : Google Scholar : PubMed/NCBI

160 

Galván I, Inácio Â, Dañino M, Corbí-Llopis R, Monserrat MT and Bernabeu-Wittel J: High SLC7A11 expression in normal skin of melanoma patients. Cancer Epidemiol. 62:1015822019. View Article : Google Scholar : PubMed/NCBI

161 

Sui S, Zhang J, Xu S, Wang Q, Wang P and Pang D: Ferritinophagy is required for the induction of ferroptosis by the bromodomain protein BRD4 inhibitor (+)-JQ1 in cancer cells. Cell Death Dis. 10:3312019. View Article : Google Scholar : PubMed/NCBI

162 

Liu T, Jiang L, Tavana O and Gu W: The deubiquitylase OTUB1 mediates ferroptosis via stabilization of SLC7A11. Cancer Res. 79:1913–1924. 2019. View Article : Google Scholar : PubMed/NCBI

163 

Dai X, Lu L, Deng S, Meng J, Wan C, Huang J, Sun Y, Hu Y, Wu B, Wu G, et al: USP7 targeting modulates anti-tumor immune response by reprogramming tumor-associated macrophages in lung cancer. Theranostics. 10:9332–9347. 2020. View Article : Google Scholar : PubMed/NCBI

164 

Wang Y, Yang L, Zhang X, Cui W, Liu Y, Sun QR, He Q, Zhao S, Zhang GA, Wang Y and Chen S: Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. EMBO Rep. 20:e475632019. View Article : Google Scholar : PubMed/NCBI

165 

Lin PL, Tang HH, Wu SY, Shaw NS and Su CL: Saponin formosanin C-induced ferritinophagy and ferroptosis in human hepatocellular carcinoma cells. Antioxidants (Basel). 9:6822020. View Article : Google Scholar : PubMed/NCBI

166 

Xiao S, Liu X, Yuan L and Wang F: A ferroptosis-related lncRNAs signature predicts prognosis and therapeutic response of gastric cancer. Front Cell Dev Biol. 9:7366822021. View Article : Google Scholar : PubMed/NCBI

167 

Wang Z, Zhang X, Tian X, Yang Y, Ma L, Wang J and Yu Y: CREB stimulates GPX4 transcription to inhibit ferroptosis in lung adenocarcinoma. Oncol Rep. 45:882021. View Article : Google Scholar : PubMed/NCBI

168 

Zilka O, Poon JF and Pratt DA: Radical-trapping antioxidant activity of copper and nickel Bis(Thiosemicarbazone) complexes underlies their potency as inhibitors of ferroptotic cell death. J Am Chem Soc. 143:19043–19057. 2021. View Article : Google Scholar : PubMed/NCBI

169 

Hassannia B, Vandenabeele P and Vanden Berghe T: Targeting ferroptosis to iron out cancer. Cancer Cell. 35:830–849. 2019. View Article : Google Scholar : PubMed/NCBI

170 

Poon JF and Pratt DA: Recent insights on hydrogen atom transfer in the inhibition of hydrocarbon autoxidation. Acc Chem Res. 51:1996–2005. 2018. View Article : Google Scholar : PubMed/NCBI

171 

Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R and Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 520:57–62. 2015. View Article : Google Scholar : PubMed/NCBI

172 

Wang SJ, Li D, Ou Y, Jiang L, Chen Y, Zhao Y and Gu W: Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 17:366–373. 2016. View Article : Google Scholar : PubMed/NCBI

173 

Nakamura T, Naguro I and Ichijo H: Iron homeostasis and iron-regulated ROS in cell death, senescence and human diseases. Biochim Biophys Acta Gen Subj. 1863:1398–1409. 2019. View Article : Google Scholar : PubMed/NCBI

174 

Luo Y, Huang Q, He B, Liu Y, Huang S and Xiao J: Regulation of ferroptosis by non-coding RNAs in the development and treatment of cancer (Review). Oncol Rep. 45:29–48. 2021. View Article : Google Scholar : PubMed/NCBI

175 

Pei Y, Qian Y, Wang H and Tan L: Epigenetic regulation of ferroptosis-associated genes and its implication in cancer therapy. Front Oncol. 12:7718702022. View Article : Google Scholar : PubMed/NCBI

176 

Moloney JN and Cotter TG: ROS signalling in the biology of cancer. Semin Cell Dev Biol. 80:50–64. 2018. View Article : Google Scholar : PubMed/NCBI

177 

Wu Y, Zhang S, Gong X, Tam S, Xiao D, Liu S and Tao Y: The epigenetic regulators and metabolic changes in ferroptosis-associated cancer progression. Mol Cancer. 19:392020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2022
Volume 48 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu J, Zhu S, Wang P, Wang J, Huang J, Wang T, Guo L, Liang D, Meng Q, Pan H, Pan H, et al: Regulators of epigenetic change in ferroptosis‑associated cancer (Review). Oncol Rep 48: 215, 2022
APA
Wu, J., Zhu, S., Wang, P., Wang, J., Huang, J., Wang, T. ... Pan, H. (2022). Regulators of epigenetic change in ferroptosis‑associated cancer (Review). Oncology Reports, 48, 215. https://doi.org/10.3892/or.2022.8430
MLA
Wu, J., Zhu, S., Wang, P., Wang, J., Huang, J., Wang, T., Guo, L., Liang, D., Meng, Q., Pan, H."Regulators of epigenetic change in ferroptosis‑associated cancer (Review)". Oncology Reports 48.6 (2022): 215.
Chicago
Wu, J., Zhu, S., Wang, P., Wang, J., Huang, J., Wang, T., Guo, L., Liang, D., Meng, Q., Pan, H."Regulators of epigenetic change in ferroptosis‑associated cancer (Review)". Oncology Reports 48, no. 6 (2022): 215. https://doi.org/10.3892/or.2022.8430