Non‑coding RNA‑mediated epigenetic modification of ferroptosis in non‑small cell lung cancer (Review)
- Authors:
- Published online on: December 9, 2024 https://doi.org/10.3892/ijo.2024.5714
- Article Number: 8
-
Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Cancer is the second leading cause of mortality worldwide after cardiovascular disease (1). Among men, the most common cancers include lung, liver, prostate, colorectal and gastric cancers (2). Lung cancer, broadly classified into small-cell (SC) and non-SC lung cancer (NSCLC), remains the leading cause of cancer-related mortality in males and the second highest cause in female patients, following breast cancer globally (3). NSCLC represents the major histological subtype of lung cancer (4-7), accounting for >80% of all cases. It comprises two primary subtypes: Lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD) (8). The global incidence of lung cancer is projected to increase from 1.7 million new cases in 2018 to 3.8 million/year by 2070 (9,10).
Although multiple approaches have decreased lung cancer mortality, the 5-year overall survival (OS) rate remains low, at 4-17%, depending on the disease stage and geographical factors (10). Surgery, targeted therapy, immunotherapy and radiotherapy are recommended therapeutic regimens for patients with NSCLC. For advanced NSCLC and earlier stages, the standard first-line treatment remains systemic platinum-based chemotherapy (11). Platinum-based chemotherapy is often combined with pemetrexed, vinorelbine/taxanes and gemcitabine as a first-line regimen for NSCLC. However, the effectiveness of platinum-based chemotherapy is limited by chemoresistance, which varies between individuals (12,13). Therefore, novel therapeutic mechanisms are needed (14,15).
Ferroptosis is a recently discovered type of regulated cell death (RCD) characterized by iron-dependent toxic accumulation of lipid peroxides on cellular membranes (16-18). Conventional therapies, including chemotherapy, radiotherapy, immunotherapy, and targeted cancer therapy, exert anti-tumor effects by inducing ferroptosis (16,19-22). Thus, targeting ferroptosis offers a promising therapeutic strategy, especially for cancer resistant to conventional treatments, including NSCLC (13,23). Understanding the molecular mechanisms that regulate ferroptosis in NSCLC may enable development of more effective therapeutic strategies for this disease.
Non-coding RNAs (ncRNAs) are a class of RNA molecules with no or limited protein-coding potential (24). ncRNAs, which include circular RNAs (circRNAs), long ncRNAs (lncRNAs) and microRNAs (miRNAs or miRs), are key epigenetic regulators of ferroptosis (25-31). ncRNAs regulate tumor malignancy by modulating ferroptosis in various types of cancers, including NSCLC (32,33). However, the mechanism underlying ncRNA-mediated epigenetic regulation of ferroptosis in NSCLC remains unclear. The present review summarizes the core molecular mechanisms of ferroptosis, how epigenetic mechanisms modify ferroptosis in NSCLC and research progress on ncRNA-mediated regulation of ferroptosis in NSCLC to clarify understanding of ncRNA-mediated epigenetic modulation of ferroptosis in NSCLC, highlighting the ncRNA-ferroptosis axis as a potential therapeutic target for NSCLC.
Core mechanisms of ferroptosis
Ferroptosis was first identified in 2012 as a form of RCD driven by iron-dependent lipid peroxidation (LPO) (34-39) (Fig. 1). This discovery stemmed from studying the mechanisms by which small molecules induce cancer cell death. Research has since identified specific small-molecule inhibitors of ferroptosis, which have revealed additional intermediate nodes in the ferroptosis signaling cascade (40). Three essential elements, reactive oxygen species (ROS), oxidizable lipids and LPO, drive the initiation and induction of ferroptosis (41) (Fig. 2). An imbalance between ferroptosis inhibitors and inducers enhances accumulation of lethal lipid peroxides (lipid hydroperoxides) on cell membranes, leading to membrane rupture and ferroptotic cell death (16,42-44).
Ferroptosis inducers
The synthesis and peroxidation of polyunsaturated fatty acid-containing phospholipids (PUFA-PLs), along with iron and mitochondrial metabolism, are the primary inducers that initiate ferroptosis (16,45-47).
LPO
Phospholipid peroxidation, a process that depends on transition metal iron, PUFA-PL and ROS, induces ferroptosis (34,48,49). Both LPO and the accumulation of peroxidized lipids trigger ferroptosis (44,50). Iron chelation studies have clarified the link between iron and ferroptosis, highlighting interplay between lipids and iron (48,49,51). PUFA-PLs, which are highly susceptible to peroxidation, serve as effective substrates for LPO via enzymatic and non-enzymatic mechanisms (43). Initiation, propagation and termination constitute the mechanism underlying PUFA-PL-mediated LPO (52,53). The incorporation and formation of peroxidized lipids in membrane phospholipids trigger ferroptosis (54,55). In the first step of PUFA-PL synthesis, acyl-CoA synthetase long-chain family member 4 (ACSL4) ligates coenzyme A (CoA) to fatty acids, with a high preference for arachidonic acid (AA) (54,55). Arachidonoyl-CoA is then esterified in membranes by lysophosphatidylcholine acyltransferase 3, which preferentially binds lysophosphatidylethanolamine and lysophosphatidylcholine to produce phospholipids in the endoplasmic reticulum (ER) (56).
Lipoxygenases (LOXs), NADPH oxidases (NOXs), oxidoreductase cytochrome P450 reductase (POR) and NADH-cytochrome b5 reductase (CYB5R1) are enzymes that generate oxidants to initiate and induce LPO (57-64). LOXs, including 15-, 12- and 5-LOX, are a class of enzymes that catalyze reactions through the involvement of iron and other metals. LOXs facilitate double oxidation of PUFAs, exacerbating cell membrane instability and triggering ferroptosis (64,65). Specifically, 15-LOX, which has a high selectivity for PUFA-PLs, oxidizes PUFA-PLs to PL-PUFA-OOH (PL-OOH). POR and CYB5R1 induce ferroptosis by transferring electrons to oxygen to produce hydrogen peroxide (H2O2), which damages cell membranes by oxidizing membrane phospholipids via Fenton reaction (57). NOXs directly produce ROS and cause LPO (66). Mitochondria produce substantial amounts of ROS, further contributing to initiation of LPO and promoting ferroptosis (47,67). The interaction of iron with lipids leads to LPO, producing lipid peroxides, PUFA-PL hydroperoxides, peroxidized PUFA-PLs (PUFA-PL-OOH) and derivatives such as 4-hydroxynonenal and malondialdehyde (MDA) (44).
Iron in ferroptosis
Iron induces ferroptosis by directly driving LPO via a non-enzymatic Fenton reaction and serving as an essential cofactor for iron-dependent peroxidases (the enzymatic LPO pathway) (16,50,53,68,69). Iron exists in two states: Ferric (Fe3+) and ferrous iron (Fe2+) (70). PUFA-PLs can react with ROS, such as lipid peroxyl (LO•) or hydroxyl radicals (HO•), via the Fenton reaction to produce PUFA-PL-OOH, triggering LPO (71-73). When PUFA-PL-OOH is not neutralized promptly, it can propagate peroxidation to neighboring PUFA-PLs in the presence of labile iron. Inhibiting the iron exporter ferroportin (74-76), promoting transferrin uptake (77) or inducing autophagic degradation of ferritin (78,79) increase the labile iron pool in cells, enhancing cellular sensitivity to ferroptosis (50). In the enzymatic LPO pathway, Fe2+ serves as a key cofactor for iron-dependent peroxidases, initiating dioxygenation of membrane PUFA-PLs (80,81). In this pathway, LOXs and POR use labile iron and O2 to peroxidize PUFA-PLs, forming PUFA-PL-OOH (43,58,62). Previous reviews provide further details on lipid resources involved in ferroptosis: ACSL4 that activates long-chain fatty acids by converting them into acyl-CoA esters, lysophosphatidylcholine acyltransferase 3 (LPCAT3) that incorporates PUFA into phosphatidylethanolamines (PEs), whereas the other activates sterol O-acyltransferase 1-producing PUFA-cholesteryl esters instead of PUFA-PEs), proxisomes that is involved in fatty acid breakdown, hydrogen peroxide production and PUFA plasmalogen biosynthesis, and lipophagy that selectively degrades lipid droplets, releasing lipids for peroxidation (41,53,82).
Ferroptosis inhibitors
Specific ferroptosis inhibitors typically suppress LPO to prevent unwanted ferroptosis (69). Ferroptosis inhibitors or defense systems that directly neutralize lipid peroxides comprise GPX4-dependent or -independent surveillance mechanisms with specific subcellular localization (35).
Solute carrier family 7 member 11-glutathione peroxidase 4 (SLC7A11-GPX4) axis
SLC7A11-GPX axis is a ferroptosis inhibitors (16,83). As a lipid repair enzyme (84,85), GPX4 converts reactive PUFA-PL-OOH to non-reactive PUFA-PL alcohols while concurrently oxidizing two reduced GSH molecules to oxidized glutathione (GSSG) (86,87). GPX4 has distinctive subcellular localizations, including mitochondrial, nuclear and cytosolic isoforms, and functions as a key ferroptosis inhibitor (34,88-91). Both mitochondrial and cytosolic GPX4 are key for ferroptosis (40). GPX4 works with the cystine/glutamate antiporter system Xc−, which consists of SLC7A11 and SLC3A2 (49). SLC7A11 exports intracellular glutamate and imports extracellular cysteine to biosynthesize reduced GSH (92,93).
Ferroptosis suppressor protein 1 (FSP1)-ubiquinol (CoQH2) system
In the FSP1-CoQH2 system, plasma membrane-localized FSP1 catalyzes the reduction of ubiquinone (CoQ10) to its reduced form, CoQH2, which functions as a lipid-soluble antioxidant to trap LO•, thereby preventing LPO and suppressing ferroptosis in cellular membranes (94-96). FSP1 inhibits ferroptosis by activating the endosomal sorting complex required for transport III complex, which causes plasma membrane damage (97,98).
GTP cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4) axis
GCH1-BH4 system is an inhibitor of ferroptosis independent of GPX4 (99,100). GCH1 inhibits ferroptosis by generating BH4, an endogenous metabolite and radical-trapping antioxidant. BH4 serves as a cofactor for aromatic amino acid hydroxylases and is analogous to CoQ10 in preventing LPO (99,100). Alternatively, GCH1 inhibits ferroptosis by remodeling the lipid membrane environment by increasing the abundance of CoQH2 and decreasing PUFA-PL levels (37).
Dihydroorotate dehydrogenase (DHODH)-dihydroubiquione (CoQH2) axis
The DHODH-CoQH2 pathway is a ferroptosis inhibitor independent of GPX4, with mitochondrial localization that inhibits LPO (101). DHODH is a mitochondrial enzyme in the inner mitochondrial membrane that enhances pyrimidine biosynthesis and converts CoQ10 to CoQH2, thereby decreasing mitochondrial CoQ10, analogous to FSP1, which functions in extra-mitochondrial membranes.
Membrane-bound O-acyltransferase domain-containing 1/2 (MBOAT1/2)-monounsaturated fatty acid (MUFA) system
MBOAT1/2-MUFA system is a newly identified ferroptosis inhibitor independent of GPX4 and FSP1 (102). MBOAT1/2 inhibits ferroptosis in this system (102). Phosphatidylethanolamine (PE)-PUFAs are the preferred substrates for LPO and dictate the ferroptosis sensitivity (103,104). MBOAT2 is a lyso-PL acyltransferase that selectively transfers MUFAs to lyso-PE, resulting in increased cellular PE-MUFA levels and decreased cellular PE-PUFAs, thereby suppressing ferroptosis induction (102).
Lathosterol oxidase (SC5D)-7-dehydrocholesterol (7-DHC) axis
The SC5D-7-DHC axis is a recently identified ferroptosis inhibitor, with 7-DHC serving as a natural inhibitor of ferroptosis (105,106). 7-DHC, generated in the ER, is found in mitochondria and cell membranes along the cholesterol synthesis pathway. It diverts the LPO pathway from phospholipids and traps radicals, suppressing LPO and subsequent ferroptosis in both mitochondria and the plasma membrane.
ncRNA-mediated epigenetic modification of ferroptosis in NSCLC
Increasing evidence indicates that dysregulated epigenetic modifications contribute to cancer initiation and progression through aberrant gene expression, protein alteration and malignant transformation (107-109). ncRNAs have recently emerged as key regulators of ferroptosis (28). Studies suggest that epigenetic modifications modulate ferroptosis at transcriptional, post-transcriptional and post-translational levels (39,110). Targeting epigenetic and post-translational modifications that modulate ferroptosis may offer new directions for cancer treatment (39,110). Recent research has shown that ncRNAs modulate ferroptosis by regulating glutathione metabolism, mitochondria-associated proteins, LPO and iron metabolism, thereby impacting cancer biology (25-31). In cancer, ncRNAs influence ferroptosis by modulating genes associated with ferroptosis inducers or inhibitors, affecting pathways such as the SLC7A11-GPX4 axis, lipid, glutamine and iron metabolism and Nrf2 signaling axis (28).
miRNAs modulate ferroptosis in NSCLC
Decreased miR-324-3p expression is observed in cisplatin-resistant A549 human lung cancer cells(DDP-A549) (111) (Fig. 3; Table I). Overexpression of miR-324-3p overcomes DDP resistance by targeting GPX4, where increased GPX4 expression reverses the miR-324-3p-induced sensitivity of DDP-A549 cells to DDP (111). miR-324-3p enhances DDP-triggered ferroptosis in DDP-A549 cells and GPX4 inhibitor RSL3 mimics miR-324-3p by increasing DDP-A549 cell sensitivity to DDP (111). Thus, miR-324-3p overcomes DDP resistance by promoting ferroptosis and suppressing GPX4 expression in NSCLC cells. Upregulation of miR-4443 has also been observed in exosomes derived from DDP-resistant NSCLC tumor tissues (112). Exosomal miR-4443 promotes resistance in recipient cells by inhibiting DDP-mediated ferroptosis through upregulation of FSP1 via m6A modification mediated by m6A-methylase methyltransferase-like protein 3 (METLL3) in vitro and enhancing tumor growth in vivo (112). Furthermore, lncRNA HCP5 promotes ferroptosis and limits brain metastasis by upregulating homeobox A7 (HOXA7) through competitive binding to miR-17-5p (113). Overexpression of miR-27a-3p, however, inhibits erastin-induced ferroptosis by suppressing SLC7A11 (114). miR-6077 prevents cisplatin/pemetrexed (CDDP/PEM)-mediated NSCLC cell death through cyclin-dependent kinase inhibitor 1A (CDKN1A)/cyclin-dependent kinase 1 (CDK1)-mediated cell cycle arrest and ferroptosis suppression by inactivating Kelch-like ECH-associated protein 1 (KEAP1), which subsequently increases Nrf2-SLC7A11/NQO1 expression and leads to chemoresistance in vitro and in vivo (115). Decreased expression of miR-302a-3p has also been observed in human NSCLC cells and tissue. miR-302a-3p inhibits cell proliferation and colony formation by inducing ferroptosis in NSCLC cells (116). Conversely, silencing miR-302a-3p inhibits erastin- or RSL3-induced ferroptosis and tumor suppression (116). miR-302a-3p binds ferroportin and decreases its protein expression; ferroportin overexpression, in turn, prevents miR-302a-3p-triggered ferroptosis and tumor suppression (116). Additionally, miR-302a-3p increases NSCLC cell sensitivity to DDP and paclitaxel. These results indicate that miR-302a-3p serves as a tumor suppressor by inducing ferroptosis through ferroportin targeting in NSCLC (116). miR-139 overexpression enhances the radiosensitivity of NSCLC cells in vitro and in vivo by targeting cJUN and Karyopherin-2 α (KPNA2), which inhibits Nrf2 signaling and promotes ionizing radiation-induced ferroptosis (117). Ionizing radiation induces miR-139 expression via early growth response 1, a transcription factor that binds to the promoter region to activate miR-139. These results suggest that miR-139 serves as a radiosensitizer by inhibiting Nrf2 expression in NSCLC (117).
lncRNAs modulate ferroptosis in NSCLC
Targeting classical ferroptosis inhibitors
Increased expression of lncRNA p53 upregulated regulator of P53 levels (PURPL) has been observed in lung cancer cells and patient samples (118) (Fig. 4; Table II). Overexpression of lncRNA PURPL promotes M2 macrophage polarization and suppresses ferroptosis by stabilizing SLC7A11 mRNA through regulation of RNA-binding motif 4 (118). Silencing SLC7A11 reverses the inhibitory effects of lncRNA PURPL on M2 macrophage polarization by inducing macrophage ferroptosis (118). Upregulation of lncRNA tyrosine protein kinase transmembrane receptor 1 antisense RNA 1 (ROR1-AS1), insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) and SLC7A11 is associated with poor prognosis in lung cancer. ROR1-AS1 stabilizes SLC7A11 mRNA through interaction with IGF2BP1 (119). Cancer-associated fibroblast (CAF)-derived exosomal ROR1-AS1 inhibits ferroptosis in lung cancer cells both in vitro and in vivo (119). Ferroptosis induced by overexpression of lncRNA ROR1-AS1 or IGF2BP1 is partially reversed by silencing IGF2BP1 or inhibiting SLC7A11 in lung cancer cells. These results suggest that CAF-secreted exosomal ROR1-AS1 inhibits ferroptosis by enhancing SLC7A11 expression or interacting with IGF2BP1 (119). Elevated levels of lncRNA-X-inactive specific transcript (XIST) mRNA and protein expression are also observed in clinical tissues and NSCLC cells (120). Silencing lncRNA-XIST reduces NSCLC cell viability by inducing ferroptosis, evidenced by increased ROS, MDA and Fe2+ levels and decreased expression of SLC7A11 and FANCD2. Additionally. silencing XIST downregulates GPX4 in NSCLC cells (120). Overexpression of GPX4 reverses ferroptosis and decreases cell viability caused by lncRNA-XIST knockdown (120). Increased expression of lncRNA Uc.339 has been observed in patients with NSCLC (121). Uc.339 inhibits the maturation of miR-339 by competitively binding to pri-miR-339. As miR-339 targets and downregulates SLC7A11 expression, Uc.339 promotes cell proliferation, migration and invasion by inhibiting ferroptosis through upregulation of SLC7A11 via miR-339 downregulation (121). Silencing lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) inhibits proliferation and induces ferroptosis by upregulating ACSL4 and downregulating SLC7A11/GPX4 (122). Silencing lncRNA H19 promotes curcumenol-induced ferroptosis (123). Increased expression of lncRNA lung cancer associated transcript 1 (LUCAT1) is observed in LUAD and LUSC tissue (124). Elevated lncRNA LUCAT1 expression is found in RSL3-treated A549 cells; this increase is reversed by the ferroptosis inhibitor Fer-1. Overexpression of LUCAT1 enhances cell proliferation and suppresses RSL3- and erastin-induced ferroptosis while silencing LUCAT1 decreases cell proliferation and facilitates ferroptosis (124). Mechanistic study have shown that silencing LUCAT1 downregulates FSP1 and GCH1 and upregulates miR-34a-5p, thereby downregulating GCH1 (124). Together, these results suggest that inhibition of LUCAT1 promotes ferroptosis by downregulating GCH1 via upregulation of miR-34a-5p (124).
Targeting iron regulators
lncRNA γ-butyrobetaine hydroxylase 1-antisense 1 (BBOX1-AS1) promotes cell proliferation, migration and invasion by inhibiting ferroptosis through post-transcriptional upregulation of prominin 2 (PROM2) expression by miR-326 sponging (125). lncRNA repulsive guidance molecule b-antisense 1 (RGMB-AS1) enhances ferroptosis by interacting with HO-1, preventing its ubiquitination by E3 ligase TRC8, which leads to increased HO-1 stability. Additionally, RGMB-AS1 binds to and promotes acetyltransferase activity of N-alpha-acetyltransferase 10, further contributing to ferroptosis (126).
Targeting transcription factors
The upregulated lncRNA ITGB2-AS1 is found in DDP-resistant NSCLC cells and cancer tissue from patients with NSCLC. Silencing ITGB2-AS1 inhibits cell proliferation and enhances apoptosis and ferroptosis in DDP-resistant NSCLC cells (137). ITGB2-AS1 promotes nicotinamide phosphoribosyltransferase (NAMPT) expression by binding to FOS-like antigen 2 (FOSL2), thereby suppressing p53 expression. FOSL2, a member of the activator protein 1 transcription factor family, enhances NSCLC malignancy (137). p53 increases cell sensitivity to ferroptosis by transcriptionally suppressing SLC7A11 expression (138). The cytosolic lncRNA P53RRA functions as a tumor suppressor by inducing ferroptosis via p53 activation (127). Downregulation of P53RRA inhibits cancer progression by acting as a tumor suppressor. Chromatin remodeling proteins CxxC finger protein 1 (Cfp1) and lymphoid-specific helicase (LSH) regulate P53RRA expression. P53RRA binds to Ras GTPase-activating protein-binding protein 1 (G3BP1) to form a P53RRA-G3BP1 cytosolic complex, displacing p53 from a G3BP1 complex and promoting p53 retention in the nucleus, which leads to cell cycle arrest, apoptosis and ferroptosis (127). The lncRNA metallothionein 1D pseudogene (MT1DP) increases sensitivity of NSCLC cells to erastin-induced ferroptosis by downregulating Nrf2 via stabilization of miR-365a-3p (128).
Targeting other factors
Increased expression of the ferroptosis suppressor aldo-keto reductase family 1 member C1 (AKR1C1) is found in gefitinib-resistant LUAD cells and associated with poor prognosis in patients with LUAD receiving first-generation epidermal growth factor receptor-tyrosine kinase inhibitor therapy (129). Silencing AKR1C1 increases the sensitivity of LUAD cells to gefitinib-induced ferroptosis (129). Reduced miR-338-3p expression leads to upregulation of AKR1C1 in gefitinib-resistant LUAD cells (129). lncRNA NEAT1_1 activates AKR1C1 by sponging miR-338-3p, thereby reducing miR-338-3p-mediated inhibition of AKR1C1. These results suggest that lncRNA NEAT1_1 promotes gefitinib resistance by suppressing ferroptosis via the upregulation of AKR1C1 via miR-338-3p sponging (129). Silencing lncRNA ITGB2-AS1 overcomes NSCLC resistance to DDP in vivo by inhibiting p53-mediated ferroptosis through activation of the FOSL2-NAMPT axis (130). Increased lncRNA HOXC-AS3 expression due to methylation promotes its binding to EP300, thereby suppressing ferroptosis in NSCLC cells (131). Upregulation of lncRNA ASMTL-AS1 inhibits tumor growth in vitro and in vivo by stabilizing spermidine/spermine N1-acetyltransferase 1, thereby stimulating ferroptosis (132). LINC00336 promotes malignancy by inhibiting ferroptosis via the absorption of miR6852, acting as a competing endogenous RNA (ceRNA) to increase the mRNA levels of cystathionine-β-synthase (133). The lncRNA syndecan-binding protein 2-antisense RNA 1 (SDCBP2-AS1) promotes cancer by inhibiting ferroptosis via sponging miR-656-3p, directly targeting cysteine-rich transmembrane BMP regulator 1 (CRIM1). SDCBP2-AS1 inhibits ferroptosis via miR-656-3p, while CRIM1 overexpression counteracts the effects of miR-656-3p on ferroptosis (134). lncRNA opioid growth factor receptor pseudogene 1 promotes NSCLC cell proliferation by inhibiting ferroptosis and suppressing miR-299-3p, leading to upregulation of SLC38A1 expression (135). β-elemene, a primary bioactive compound derived from Curcuma wenyujin, enhances sensitivity of cancer cells to erlotinib by inducing ferroptosis via upregulation of lncRNA H19 (136).
circRNAs modulate ferroptosis in NSCLC
Targeting SLC7A11
Increased circ_0070440 expression is observed in LUAD cells. Silencing circ_0070440 inhibits cell proliferation and enhances apoptosis and ferroptosis in LUAD cells (139). circ_0070440 promotes malignant progression by suppressing ferroptosis through upregulation of SLC7A11 expression via miR-485-5p sponging (139) (Fig. 5; Table III). Increased expression of circP4HB is observed in LUAD. circP4HB inhibits erastin-induced ferroptosis by promoting GSH synthesis (140). Mechanistic study indicates that circP4HB upregulates SLC7A11 by serving as a competing endogenous RNA and modulating miR-1184 expression (140). circPDSS1 expression is elevated in NSCLC cells (141). Silencing circPDSS1 inhibits NSCLC cell viability and induces ferroptosis (141). Mechanistic study suggest that circPDSS1 functions as a competing endogenous RNA 'sponge' to negatively regulate miR-137 expression (141). Silencing circPDSS1 also downregulates GPX4 and the glutamate-cysteine ligase catalytic subunit (141).
Targeting GPX4
circ_0082374 promotes tumorigenesis in NSCLC by suppressing ferroptosis via upregulation of GPX4 via miR-491-5p sponging (142). Increased circ_0082374 levels are observed in NSCLC tissue and cells. Silencing circ_0082374 inhibits cell proliferation and tumor metastasis while facilitating ferroptosis by decreasing iron and LPO levels in NSCLC (142). Mechanistic study show that circ_0082374 enhances GPX4 expression by sequestering miR-491-5p (142). In LUAD samples and cell lines, decreased circRNA signal peptide-CUB-EGF domain-containing protein 3 (circSCUBE3) expression is noted (143). Overexpression of circSCUBE3 decreases GSH levels and GSH/GSSG ratio in LUAD cells, while loss of circSCUBE3 reverses erastin-induced ferroptosis (143). circSCUBE3 downregulates GPX4 expression in LUAD cells, and silencing GPX4 counteracts the ferroptosis induced by circSCUBE3 deficiency (143). circSCUBE3 downregulates GPX4 expression by competitively interacting with CREB, which activates GPX4 transcription. These results suggest that circSCUBE3 inhibits LUAD development by promoting ferroptosis via the CREB/GPX4/GSH axis (143). Increased circDTL expression has been detected in NSCLC cells (144). Silencing circDTL promotes apoptosis and ferroptosis in NSCLC cells (144) and enhances their sensitivity to chemotherapy, thus inhibiting tumor growth in vivo. CircDTL acts as an oncogene by suppressing ferroptosis and increasing GPX4 expression via miR-1287-5p sponging (144).
Targeting ACSL4
Downregulated circSCN8A expression has been observed in NSCLC tissue and cells. Decreased circSCN8A expression is positively associated with aggressive clinicopathological characteristics and predicts poor prognosis in patients with NSCLC (145). circSCN8A suppresses tumor growth in vivo and inhibits cell proliferation, migration, invasion and epithelial-mesenchymal transition in vitro. circSCN8A enhances ferroptosis in NSCLC cells. Mechanistic studies revealed that circSCN8A functions as a ceRNA by sponging miR-1290 to upregulate ACSL4 (145). Silencing ACSL4 or overexpressing miR-1290 reverses circSCN8A-induced ferroptosis and inhibits cell proliferation and tumor metastasis (145). These results indicate that circSCN8A suppresses malignant progression by inducing ferroptosis through miR-1290 sponging to enhance ACSL4 expression (145).
Targeting other factors
Plasma exosomes from patients with LUAD show decreased LPO and reduced sensitivity of LUAD cells to ferroptosis (146). Exosomal circRNA_101093 maintains elevated intracellular circRNA_101093 levels in LUAD cells to modulate AA (146). circRNA_101093 binds to and upregulates fatty acid-binding protein 3 (FABP3), which transports AA and enhances its reaction with taurine (146). These results indicate that circRNA_101093 inhibits ferroptosis by upregulating FABP3 and suggest that blocking exosomes may be a novel therapeutic approach for LUAD (146).
Conclusion
The present review summarizes the role of ncRNA-mediated epigenetic modulation of ferroptosis in NSCLC and the roles of miRNAs, lncRNA and circRNA in regulating ferroptosis in NSCLC cells. However, studies on ncRNA-induced epigenetic modifications that modulate ferroptosis in NSCLC are in their early stages and the present study has limitations. First, other ncRNAs that modulate ferroptosis require further exploration. Second, while ncRNA-mediated epigenetic modifications of ferroptosis in NSCLC have been identified, it remains unclear whether small-molecule compounds effectively target these ncRNA-mediated mechanisms. Third, ncRNAs modulate the crosstalk between ferroptosis and regulated cell death mechanisms in cancer (147). However, the role of ncRNAs in the interplay between ferroptosis and novel RCD mechanisms, such as cuproptosis, in NSCLC remains largely unknown (148). The aforementioned studies primarily focused on classical pathways of ferroptosis, such as the ACSL4-dependent LPO or SLC7A11-GPX4 system. However, the effects of other ferroptosis inhibitors, including DHODH-CoQH2, MBOAT1/2-MUFA, GCH1-BH4 and SC5D-7-DHC, are still poorly understood. Fifth, many ncRNAs directly modulate ferroptosis by regulating ferroptosis-associated genes involved in antioxidant defense and iron and lipid metabolism or indirectly target ferroptosis modulators, such as activated transcription factor 4 in NSCLC (149). However, whether ncRNAs modulate other transcription factors, including Nrf2, remain poorly understood.
The clinical application of RNA-based therapeutics has made progress, primarily using antisense oligonucleotides and small interfering RNA (150). However, no lncRNA-based therapeutics or circRNA-targeted treatments have entered clinical practice, although miRNA mimics and anti-miRNA therapeutics are under clinical development (150-152). Emerging evidence suggests that small molecule compounds can target ncRNAs for cancer treatment (153). Small molecules targeting miRNA (154,155) and lncRNA (136,156) that regulate ferroptosis may offer new opportunities for NSCLC therapy. However, no small molecules targeting miRNAs and lncRNAs involved in regulating ferroptosis have yet been introduced for clinical use in NSCLC treatment.
Collectively, ncRNAs modulate the tumor burden in NSCLC by regulating ferroptosis through proteins or genes associated with pro- and anti-ferroptotic factors. The present review summarizes advances in understanding the ncRNA-mediated mechanisms affecting ferroptosis in NSCLC, highlighting a novel therapeutic avenue for NSCLC through the ncRNA-ferroptosis axis.
Availability of data and materials
Not applicable.
Authors' contributions
YW, JC, and MD designed and conceived the review. ZZ, HW and JSF edited the manuscript. YW, YL and YC wrote the manuscript. YW and JSF generated the figures. Data authentication is not applicable. All authors have read and approved the final manuscript.
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.
Acknowledgements
No applicable.
Funding
The present study was supported by the Wu Jieping Medical Foundation (320.6750.2024-13-59), Science Foundation of Aerospace Center hospital (grant nos. YN202402 and YN202423), Science Foundation of AMHT (grant nos. 2022YK01 and 2022YK27) and Grant of Chinese Medicine Education Association (grant no. 2022KTZ019).
References
Adhikari S, Bhattacharya A, Adhikary S, Singh V, Gadad SS, Roy S and Da C: The paradigm of drug resistance in cancer: An epigenetic perspective. Biosci Rep. 42:BSR202118122022. View Article : Google Scholar : PubMed/NCBI | |
Bukowski K, Kciuk M and Kontek R: Mechanisms of multi-drug resistance in cancer chemotherapy. Int J Mol Sci. 21:32332020. View Article : Google Scholar | |
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI | |
Leiter A, Veluswamy RR and Wisnivesky JP: The global burden of lung cancer: Current status and future trends. Nat Rev Clin Oncol. 20:624–639. 2023. View Article : Google Scholar : PubMed/NCBI | |
Rotow J and Bivona TG: Understanding and targeting resistance mechanisms in NSCLC. Nat Rev Cancer. 17:637–658. 2017. View Article : Google Scholar : PubMed/NCBI | |
LoPiccolo J, Gusev A, Christiani DC and Jänne PA: Lung cancer in patients who have never smoked-an emerging disease. Nat Rev Clin Oncol. 21:121–146. 2024. View Article : Google Scholar : PubMed/NCBI | |
Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023. View Article : Google Scholar : PubMed/NCBI | |
Otano I, Ucero AC, Zugazagoitia J and Paz-Ares L: At the crossroads of immunotherapy for oncogene-addicted subsets of NSCLC. Nat Rev Clin Oncol. 20:143–159. 2023. View Article : Google Scholar : PubMed/NCBI | |
Soerjomataram I and Bray F: Planning for tomorrow: Global cancer incidence and the role of prevention 2020-2070. Nat Rev Clin Oncol. 18:663–672. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL and Paz-Ares L: Lung cancer: Current therapies and new targeted treatments. Lancet. 389:299–311. 2017. View Article : Google Scholar | |
Nagasaka M and Gadgeel SM: Role of chemotherapy and targeted therapy in early-stage non-small cell lung cancer. Expert Rev Anticancer Ther. 18:63–70. 2018. View Article : Google Scholar | |
Yin JY, Li X, Zhou HH and Liu ZQ: Pharmacogenomics of platinum-based chemotherapy sensitivity in NSCLC: Toward precision medicine. Pharmacogenomics. 17:1365–1378. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Hu J, Fleishman JS, Li Y, Ren Z, Wang J, Feng Y, Chen J and Wang H: Inducing ferroptosis by traditional medicines: A novel approach to reverse chemoresistance in lung cancer. Front Pharmacol. 15:12901832024. View Article : Google Scholar : PubMed/NCBI | |
Herbst RS, Morgensztern D and Boshoff C: The biology and management of non-small cell lung cancer. Nature. 553:446–454. 2018. View Article : Google Scholar : PubMed/NCBI | |
Lim ZF and Ma PC: Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J Hematol Oncol. 12:1342019. View Article : Google Scholar : PubMed/NCBI | |
Lei G, Zhuang L and Gan B: Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 22:381–396. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kobayashi H, Yoshimoto C, Matsubara S, Shigetomi H and Imanaka S: A comprehensive overview of recent developments on the mechanisms and pathways of ferroptosis in cancer: The potential implications for therapeutic strategies in ovarian cancer. Cancer Drug Resist. 6:547–566. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yunchu Y, Miyanaga A and Seike M: Integrative analysis of ferroptosis-related genes in small cell lung cancer for the identification of biomarkers and therapeutic targets. Front Biosci (Landmark Ed). 28:1252023. View Article : Google Scholar : PubMed/NCBI | |
Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, Dai X, Li Z and Wu G: Ferroptosis: A novel anti-tumor action for cisplatin. Cancer Res Treat. 50:445–460. 2018. View Article : Google Scholar : | |
Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, Ajani JA, Xiao Q, Liao Z, Wang H and Gan B: The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30:146–162. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R and Tang D: Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 63:173–184. 2016. View Article : Google Scholar | |
Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, Liao P, Lang X, Kryczek I, Sell A, et al: CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 569:270–274. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Wu X, Ren Z, Li Y, Zou W, Chen J and Wang H: Overcoming cancer chemotherapy resistance by the induction of ferroptosis. Drug Resist Updat. 66:1009162023. View Article : Google Scholar : PubMed/NCBI | |
Lin X, Wu Z, Hu H, Luo ML and Song E: Non-coding RNAs rewire cancer metabolism networks. Semin Cancer Biol. 75:116–126. 2021. View Article : Google Scholar : PubMed/NCBI | |
Balihodzic A, Prinz F, Dengler MA, Calin GA, Jost PJ and Pichler M: Non-coding RNAs and ferroptosis: Potential implications for cancer therapy. Cell Death Differ. 29:1094–1106. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ensoy M, Bumin ZS, Jama HA and Cansaran-Duman D: The regulation role of ferroptosis mechanism of anti-cancer drugs and noncoding RNAs. Curr Med Chem. 30:1638–1656. 2023. View Article : Google Scholar | |
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 | |
Valashedi MR, Bamshad C, Najafi-Ghalehlou N, Nikoo A, Tomita K, Kuwahara Y, Sato T, Roushandeh AM and Roudkenar MH: Non-coding RNAs in ferroptotic cancer cell death pathway: Meet the new masters. Hum Cell. 35:972–994. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Tang L, Zhang Y, Ge G, Jiang X, Mo Y, Wu P, Deng X, Li L, Zuo S, et al: Regulatory pathways and drugs associated with ferroptosis in tumors. Cell Death Dis. 13:5442022. View Article : Google Scholar : PubMed/NCBI | |
Xie B and Guo Y: Molecular mechanism of cell ferroptosis and research progress in regulation of ferroptosis by noncoding RNAs in tumor cells. Cell Death Discov. 7:1012021. View Article : Google Scholar : PubMed/NCBI | |
Zuo YB, Zhang YF, Zhang R, Tian JW, Lv XB, Li R, Li SP, Cheng MD, Shan J, Zhao Z and Xin H: Ferroptosis in cancer progression: Role of noncoding RNAs. Int J Biol Sci. 18:1829–1843. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Fleishman JS, Cheng S, Wang W, Wu F and Wang Y and Wang Y: Epigenetic modification of ferroptosis by non-coding RNAs in cancer drug resistance. Mol Cancer. 23:1772024. View Article : Google Scholar : PubMed/NCBI | |
Sun L, Cao H, Wang Y and Wang H: Regulating ferroptosis by noncoding RNAs in hepatocellular carcinoma. Biol Direct. 19:802024. View Article : Google Scholar | |
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 | |
Gu Y, Li Y, Wang J, Zhang L, Zhang J and Wang Y: Targeting ferroptosis: Paving new roads for drug design and discovery. Eur J Med Chem. 247:1150152023. View Article : Google Scholar | |
Huo L, Liu C, Yuan Y, Liu X and Cao Q: Pharmacological inhibition of ferroptosis as a therapeutic target for sepsis-associated organ damage. Eur J Med Chem. 257:1154382023. View Article : Google Scholar : PubMed/NCBI | |
Stockwell BR: Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell. 185:2401–2421. 2022. View Article : Google Scholar : PubMed/NCBI | |
Yin L, Liu P, Jin Y, Ning Z, Yang Y and Gao H: Ferroptosis-related small-molecule compounds in cancer therapy: Strategies and applications. Eur J Med Chem. 244:1148612022. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J and Wang H: Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther. 8:4492023. View Article : Google Scholar : PubMed/NCBI | |
Dixon SJ and Olzmann JA: The cell biology of ferroptosis. Nat Rev Mol Cell Biol. 25:424–442. 2024. View Article : Google Scholar : PubMed/NCBI | |
Dai E, Chen X, Linkermann A, Jiang X, Kang R, Kagan VE, Bayir H, Yang WS, Garcia-Saez AJ, Ioannou MS, et al: A guideline on the molecular ecosystem regulating ferroptosis. Nat Cell Biol. 26:1447–1457. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Kang R, Kroemer G and Tang D: Ferroptosis in infection, inflammation, and immunity. J Exp Med. 218:e202105182021. View Article : Google Scholar : PubMed/NCBI | |
Hadian K and Stockwell BR: SnapShot: Ferroptosis. Cell. 181:1188–1188.e1. 2020. View Article : Google Scholar : PubMed/NCBI | |
Pope LE and Dixon SJ: Regulation of ferroptosis by lipid metabolism. Trends Cell Biol. 33:1077–1087. 2023. View Article : Google Scholar : PubMed/NCBI | |
Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gan B: Mitochondrial regulation of ferroptosis. J Cell Biol. 220:e2021050432021. View Article : Google Scholar : PubMed/NCBI | |
Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB and Jiang X: Role of mitochondria in ferroptosis. Mol Cell. 73:354–363.e3. 2019. View Article : Google Scholar : | |
Stockwell BR, Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang DD: Ironing out the details of ferroptosis. Nat Cell Biol. 26:1386–1393. 2024. View Article : Google Scholar : PubMed/NCBI | |
Liang D, Minikes AM and Jiang X: Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell. 82:2215–2227. 2022. View Article : Google Scholar : PubMed/NCBI | |
Doll S and Conrad M: Iron and ferroptosis: A still ill-defined liaison. IUBMB Life. 69:423–434. 2017. View Article : Google Scholar : PubMed/NCBI | |
Helberg J and Pratt DA: Autoxidation vs. antioxidants-the fight for forever. Chem Soc Rev. 50:7343–7358. 2021. View Article : Google Scholar : PubMed/NCBI | |
Conrad M and Pratt DA: The chemical basis of ferroptosis. Nat Chem Biol. 15:1137–1147. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kang MJ, Fujino T, Sasano H, Minekura H, Yabuki N, Nagura H, Iijima H and Yamamoto TT: A novel arachidonate-preferring acyl-CoA synthetase is present in steroidogenic cells of the rat adrenal, ovary, and testis. Proc Natl Acad Sci USA. 94:2880–2884. 1997. View Article : Google Scholar : PubMed/NCBI | |
Kim JW, Lee JY, Oh M and Lee EW: An integrated view of lipid metabolism in ferroptosis revisited via lipidomic analysis. Exp Mol Med. 55:1620–1631. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hishikawa D, Shindou H, Kobayashi S, Nakanishi H, Taguchi R and Shimizu T: Discovery of a lysophospholipid acyltransferase family essential for membrane asymmetry and diversity. Proc Natl Acad Sci USA. 105:2830–2835. 2008. View Article : Google Scholar : PubMed/NCBI | |
Yan B, Ai Y, Sun Q, Ma Y, Cao Y, Wang J, Zhang Z and Wang X: Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol Cell. 81:355–369.e10. 2021. View Article : Google Scholar | |
Zou Y, Li H, Graham ET, Deik AA, Eaton JK, Wang W, Sandoval-Gomez G, Clish CB, Doench JG and Schreiber SL: Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 16:302–309. 2020. View Article : Google Scholar : PubMed/NCBI | |
Poursaitidis I, Wang X, Crighton T, Labuschagne C, Mason D, Cramer SL, Triplett K, Roy R, Pardo OE, Seckl MJ, et al: Oncogene-selective sensitivity to synchronous cell death following modulation of the amino acid nutrient cystine. Cell Rep. 18:2547–2556. 2017. View Article : Google Scholar : PubMed/NCBI | |
Yang WH, Ding CC, Sun T, Rupprecht G, Lin CC, Hsu D and Chi JT: The hippo pathway effector TAZ regulates ferroptosis in renal cell carcinoma. Cell Rep. 28:2501–2508.e4. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, Song S, Tavana O and Gu W: ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 21:579–591. 2019. View Article : Google Scholar : PubMed/NCBI | |
Shah R, Shchepinov MS and Pratt DA: Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci. 4:387–396. 2018. View Article : Google Scholar : PubMed/NCBI | |
Anthonymuthu TS, Tyurina YY, Sun WY, Mikulska-Ruminska K, Shrivastava IH, Tyurin VA, Cinemre FB, Dar HH, VanDemark AP, Holman TR, et al: Resolving the paradox of ferroptotic cell death: Ferrostatin-1 binds to 15LOX/PEBP1 complex, suppresses generation of peroxidized ETE-PE, and protects against ferroptosis. Redox Biol. 38:1017442021. View Article : Google Scholar | |
Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS and Stockwell BR: Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 113:E4966–E4975. 2016. View Article : Google Scholar : PubMed/NCBI | |
Haeggström JZ and Funk CD: Lipoxygenase and leukotriene pathways: Biochemistry, biology, and roles in disease. Chem Rev. 111:5866–5898. 2011. View Article : Google Scholar : PubMed/NCBI | |
Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, Sabatini DM, Birsoy K and Possemato R: NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 551:639–643. 2017. View Article : Google Scholar : PubMed/NCBI | |
Gaschler MM, Hu F, Feng H, Linkermann A, Min W and Stockwell BR: Determination of the subcellular localization and mechanism of action of ferrostatins in suppressing ferroptosis. ACS Chem Biol. 13:1013–1020. 2018. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Li J, Kang R, Klionsky DJ and Tang D: Ferroptosis: Machinery and regulation. Autophagy. 17:2054–2081. 2021. View Article : Google Scholar : | |
Hassannia B, Vandenabeele P and Berghe TV: Targeting ferroptosis to iron out cancer. Cancer Cell. 35:830–849. 2019. View Article : Google Scholar : PubMed/NCBI | |
Jacquemyn J, Ralhan I and Ioannou MS: Driving factors of neuronal ferroptosis. Trends Cell Biol. 34:535–546. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ayala A, Muñoz MF and Argüelles S: Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014:3604382014. View Article : Google Scholar : PubMed/NCBI | |
Dos Santos AF, Fazeli G, da Silva TN and Angeli JP: Ferroptosis: Mechanisms and implications for cancer development and therapy response. Trends Cell Biol. 33:1062–1076. 2023. View Article : Google Scholar : PubMed/NCBI | |
Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X and Choi AM: Mechanisms of cell death in oxidative stress. Antioxid Redox Signal. 9:49–89. 2007. View Article : Google Scholar | |
Bao WD, Pang P, Zhou XT, Hu F, Xiong W, Chen K, Wang J, Wang F, Xie D, Hu YZ, et al: Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer's disease. Cell Death Differ. 28:1548–1562. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen PH, Wu J, Ding CC, Lin CC, Pan S, Bossa N, Xu Y, Yang WH, Mathey-Prevot B and Chi J: Kinome screen of ferroptosis reveals a novel role of ATM in regulating iron metabolism. Cell Death Differ. 27:1008–1022. 2020. View Article : Google Scholar : | |
Geng N, Shi BJ, Li SL, Zhong ZY, Li YC, Xua WL, Zhou H and Cai JH: Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharmacol Sci. 22:3826–3836. 2018.PubMed/NCBI | |
Gao M, Monian P, Quadri N, Ramasamy R and Jiang X: Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 59:298–308. 2015. View Article : Google Scholar : PubMed/NCBI | |
Gao M, Monian P, Pan Q, Zhang W, Xiang J and Jiang X: Ferroptosis is an autophagic cell death process. Cell Res. 26:1021–1032. 2016. View Article : Google Scholar : PubMed/NCBI | |
Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ III, Kang R and Tang D: Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 12:1425–1428. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Yu C, Kang R and Tang D: Iron metabolism in ferroptosis. Front Cell Dev Biol. 8:5902262020. View Article : Google Scholar : PubMed/NCBI | |
David S, Jhelum P, Ryan F, Jeong SY and Kroner A: Dysregulation of iron homeostasis in the central nervous system and the role of ferroptosis in neurodegenerative disorders. Antioxid Redox Signal. 37:150–170. 2022. View Article : Google Scholar | |
Stockwell BR and Jiang X: The chemistry and biology of ferroptosis. Cell Chem Biol. 27:365–375. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sun Y, Xia X, Basnet D, Zheng JC, Huang J and Liu J: Mechanisms of ferroptosis and emerging links to the pathology of neurodegenerative diseases. Front Aging Neurosci. 14:9041522022. View Article : Google Scholar : PubMed/NCBI | |
Brigelius-Flohé R and Flohé L: Regulatory phenomena in the glutathione peroxidase superfamily. Antioxid Redox Signal. 33:498–516. 2020. View Article : Google Scholar | |
Brigelius-Flohé R and Maiorino M: Glutathione peroxidases. Biochim Biophys Acta. 1830:3289–3303. 2013. View Article : Google Scholar | |
Seibt TM, Proneth B and Conrad M: Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 133:144–152. 2019. View Article : Google Scholar | |
Ursini F, Maiorino M, Valente M, Ferri L and Gregolin C: Purification from pig liver of a protein which protects liposomes and biomembranes from peroxidative degradation and exhibits glutathione peroxidase activity on phosphatidylcholine hydroperoxides. Biochim Biophys Acta. 710:197–211. 1982. View Article : Google Scholar : PubMed/NCBI | |
Forcina GC and Dixon SJ: GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 19:e18003112019. View Article : Google Scholar : PubMed/NCBI | |
Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, et al: Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 16:1180–1191. 2014. View Article : Google Scholar | |
Ingold I, Berndt C, Schmitt S, Doll S, Poschmann G, Buday K, Roveri A, Peng X, Freitas FP, Seibt T, et al: Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell. 172:409–422.e21. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish C, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI | |
Koppula P, Zhang Y, Zhuang L and Gan B: Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun (Lond). 38:122018.PubMed/NCBI | |
Sato H, Tamba M, Ishii T and Bannai S: Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J Biol Chem. 274:11455–11458. 1999. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Grocin AG, 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 | |
Nakamura T, Hipp C, Mourão ASD, Borggräfe J, Aldrovandi M, Henkelmann B, Wanninger J, Mishima E, Lytton E, Emler D, et al: Phase separation of FSP1 promotes ferroptosis. Nature. 619:371–377. 2023. View Article : Google Scholar : PubMed/NCBI | |
Dai E, Zhang W, Cong D, Kang R, Wang J and Tang D: AIFM2 blocks ferroptosis independent of ubiquinol metabolism. Biochem Biophys Res Commun. 523:966–971. 2020. View Article : Google Scholar : PubMed/NCBI | |
Pedrera L, Espiritu RA, Ros U, Weber J, Schmitt A, Stroh J, Hailfinger S, von Karstedt S and García-Sáez AJ: Ferroptotic pores induce Ca(2+) fluxes and ESCRT-III activation to modulate cell death kinetics. Cell Death Differ. 28:1644–1657. 2021. View Article : Google Scholar | |
Kraft V, 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 | |
Soula M, Weber RA, Zilka O, Alwaseem H, La K, Yen F, Molina H, Garcia-Bermudez J, Pratt DA and Birso K: Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol. 16:1351–1360. 2020. View Article : Google Scholar : PubMed/NCBI | |
Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al: DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 593:586–590. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liang D, Feng Y, Zandkarimi F, Wang H, Zhang Z, Kim J, Cai Y, Gu W, Stockwell BR and Jiang X: Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell. 186:2748–2764. 2023. View Article : Google Scholar : PubMed/NCBI | |
Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al: ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 13:91–98. 2017. View Article : Google Scholar : | |
Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, et al: Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 13:81–90. 2017. View Article : Google Scholar | |
Freitas FP, Alborzinia H, Dos Santos AF, Nepachalovich P, Pedrera L, Zilka O, Inague A, Klein C, Aroua N, Kaushal K, et al: 7-Dehydrocholesterol is an endogenous suppressor of ferroptosis. Nature. 626:401–410. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Ran Q, Duan Q, Jin J, Wang Y, Yu L, Wang C, Zhu Z, Chen X, Weng L, et al: 7-Dehydrocholesterol dictates ferroptosis sensitivity. Nature. 626:411–418. 2024. View Article : Google Scholar : PubMed/NCBI | |
Garcia-Martinez L, Zhang Y, Nakata Y, Chan HL and Morey L: Epigenetic mechanisms in breast cancer therapy and resistance. Nat Commun. 12:17862021. View Article : Google Scholar : PubMed/NCBI | |
Ling C and Rönn T: Epigenetics in human obesity and type 2 diabetes. Cell Metab. 29:1028–1044. 2019. View Article : Google Scholar : PubMed/NCBI | |
Shu F, Xiao H, Li QN, Ren XS, Liu ZG, Hu BW, Wang HS, Wang H and Jiang GM: Epigenetic and post-translational modifications in autophagy: Biological functions and therapeutic targets. Signal Transduct Target Ther. 8:322023. View Article : Google Scholar : PubMed/NCBI | |
Yang M, Luo H, Yi X, Wei X and Jiang DS: The epigenetic regulatory mechanisms of ferroptosis and its implications for biological processes and diseases. MedComm (2020). 4:e2672023. View Article : Google Scholar : PubMed/NCBI | |
Deng SH, Wu DM, Li L, Liu T, Zhang T, Li J, Yu Y, He M, Zhao YY, Han R and Xu Y: miR-324-3p reverses cisplatin resistance by inducing GPX4-mediated ferroptosis in lung adenocarcinoma cell line A549. Biochem Biophys Res Commun. 549:54–60. 2021. View Article : Google Scholar : PubMed/NCBI | |
Song Z, Jia G, Ma P and Cang S: Exosomal miR-4443 promotes cisplatin resistance in non-small cell lung carcinoma by regulating FSP1 m6A modification-mediated ferroptosis. Life Sci. 276:1193992021. View Article : Google Scholar : PubMed/NCBI | |
Chen Q, Pan Q, Gao H, Wang Y and Zhong X: miR-17-5p/HOXA7 is a potential driver for brain metastasis of lung adenocarcinoma related to ferroptosis revealed by bioinformatic analysis. Front Neurol. 13:8789472022. View Article : Google Scholar : PubMed/NCBI | |
Lu X, Kang N, Ling X, Pan M, Du W and Gao S: MiR-27a-3p promotes non-small cell lung cancer through SLC7A11-mediated-ferroptosis. Front Oncol. 11:7593462021. View Article : Google Scholar : PubMed/NCBI | |
Bi G, Liang J, Zhao M, Zhang H, Jin X, Lu T, Zheng Y, Bian Y, Chen Z, Huang Y, et al: miR-6077 promotes cisplatin/pemetrexed resistance in lung adenocarcinoma via CDKN1A/cell cycle arrest and KEAP1/ferroptosis pathways. Mol Ther Nucleic Acids. 28:366–386. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wei D, Ke YQ, Duan P, Zhou L, Wang CY and Cao P: MicroRNA-302a-3p induces ferroptosis of non-small cell lung cancer cells via targeting ferroportin. Free Radic Res. 55:821–830. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Xu Y, Cheng Z, Zhao J, Wang M, Sun Y, Mi Z, Yuan Z and Wu Z: The EGR1/miR-139/NRF2 axis orchestrates radiosensitivity of non-small-cell lung cancer via ferroptosis. Cancer Lett. 595:2170002024. View Article : Google Scholar : PubMed/NCBI | |
Guo J, Gong C and Wang H: PURPL promotes M2 macrophage polarization in lung cancer by regulating RBM4/xCT signaling. Crit Rev Eukaryot Gene Expr. 34:59–68. 2024. View Article : Google Scholar : PubMed/NCBI | |
Yao F, Zhao Y, Wang G, Zhao M, Hong X, Ye Z, Dong F, Li W and Deng Q: Exosomal lncRNA ROR1-AS1 from cancer-associated fibroblasts inhibits ferroptosis of lung cancer cells through the IGF2BP1/SLC7A11 signal axis. Cell Signal. 120:1112212024. View Article : Google Scholar : PubMed/NCBI | |
Lu CL, Liu J and Yang JF: LncRNA-XIST promotes lung adenocarcinoma growth and inhibits ferroptosis by regulating GPX4. Mol Biotechnol. 28: View Article : Google Scholar : 2023. | |
Zhang N, Huang J, Xu M and Wang Y: LncRNA T-UCR Uc.339/miR-339/SLC7A11 axis regulates the metastasis of ferroptosis-induced lung adenocarcinoma. J Cancer. 13:1945–1957. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wu H and Liu A: Long non-coding RNA NEAT1 regulates ferroptosis sensitivity in non-small-cell lung cancer. J Int Med Res. 49:3000605219961832021. View Article : Google Scholar : PubMed/NCBI | |
Zhang R, Pan T, Xiang Y, Zhang M, Xie H, Liang Z, Chen B, Xu C, Wang J, Huan Q, et al: Curcumenol triggered ferroptosis in lung cancer cells via lncRNA H19/miR-19b-3p/FTH1 axis. Bioact Mater. 13:23–36. 2022.PubMed/NCBI | |
Tai F, Zhai R, Ding K, Zhang Y, Yang H, Li H, Wang Q, Cao Z, Ge C, Fu H, et al: Long non-coding RNA lung cancer-associated transcript 1 regulates ferroptosis via microRNA-34a-5p-mediated GTP cyclohydrolase 1 downregulation in lung cancer cells. Int J Oncol. 64: View Article : Google Scholar : 2024. | |
An J, Shi J, Yang C, Luo J, Li Y, Ren J, Lv Y and Zhang Y: Regulation of tumorigenesis and ferroptosis in non-small cell lung cancer by a novel BBOX1-AS1/miR-326/PROM2 axis. Mol Cell Biochem. 479:2143–2155. 2023. View Article : Google Scholar : PubMed/NCBI | |
Gao GB, Chen L, Pan JF, Lei T, Cai X, Hao Z, Wang Q, Shan G and Li J: LncRNA RGMB-AS1 inhibits HMOX1 ubiquitination and NAA10 activation to induce ferroptosis in non-small cell lung cancer. Cancer Lett. 590:2168262024. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Gai C, Liu C, Wu X, Yu M, Zheng J, Zhang W, Lv S and Li W: MT1DP loaded by folate-modified liposomes sensitizes erastin-induced ferroptosis via regulating miR-365a-3p/NRF2 axis in non-small cell lung cancer cells. Cell Death Dis. 11:7512020. View Article : Google Scholar : PubMed/NCBI | |
Zhen S, Jia Y, Zhao Y, Wang J, Zheng B, Liu T, Duan Y, Lv W, Wang J, Xu F, et al: NEAT1_1 confers gefitinib resistance in lung adenocarcinoma through promoting AKR1C1-mediated ferroptosis defence. Cell Death Discov. 10:1312024. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Wang L, Liu J, Wan Z, Zhou L, Liao H and Wan R: LncRNA ITGB2-AS1 promotes cisplatin resistance of non-small cell lung cancer by inhibiting ferroptosis via activating the FOSL2/NAMPT axis. Cancer Biol Ther. 24:22233772023. View Article : Google Scholar : PubMed/NCBI | |
Shi Z, Zhang H, Shen Y, Zhang S, Zhang X, Xu Y and Sun D: SETD1A-mediated H3K4me3 methylation upregulates lncRNA HOXC-AS3 and the binding of HOXC-AS3 to EP300 and increases EP300 stability to suppress the ferroptosis of NSCLC cells. Thorac Cancer. 14:2579–2590. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sui X, Hu N, Zhang Z, Wang Y, Wang P and Xiu G: ASMTL-AS1 impedes the malignant progression of lung adenocarcinoma by regulating SAT1 to promote ferroptosis. Pathol Int. 71:741–751. 2021. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Dai N, Ma H and Feng Y: Silencing of long non-coding RNA SDCBP2-AS1/microRNA-656-3p/CRIM1 axis promotes ferroptosis of lung cancer cells. Cell Mol Biol (Noisy-le-grand). 69:189–194. 2023. View Article : Google Scholar : PubMed/NCBI | |
Liu L, Su S, Ye D, Yu Z, Lu W and Li X: Long non-coding RNA OGFRP1 regulates cell proliferation and ferroptosis by miR-299-3p/SLC38A1 axis in lung cancer. Anticancer Drugs. 33:826–839. 2022.PubMed/NCBI | |
Xu C, Jiang ZB, Shao L, Zhao ZM, Fan XX, Sui X, Yu LL, Wang XR, Zhang RN, Wang WJ, et al: β-Elemene enhances erlotinib sensitivity through induction of ferroptosis by upregulating lncRNA H19 in EGFR-mutant non-small cell lung cancer. Pharmacol Res. 191:1067392023. View Article : Google Scholar | |
Xu P, Wang L, Xie X, Hu F, Yang Q, Hu R, Jiang L, Ding F, Mei J, Liu J and Xiao H: Hsa_circ_0001869 promotes NSCLC progression via sponging miR-638 and enhancing FOSL2 expression. Aging (Albany NY). 12:23836–23848. 2020. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Zhao Y, Cui Q, Shen J, Shen W and Weng Y: Hsa_ circ_0070440 promotes lung adenocarcinoma progression by SLC7A11-mediated-ferroptosis. Histol Histopathol. 38:1429–1441. 2023.PubMed/NCBI | |
Pan CF, Wei K, Ma ZJ, He YZ, Huang JJ, Guo ZZ, Chen ZP, Barr MP, Shackelford RE, Xia Y and Wang J: CircP4HB regulates ferroptosis via SLC7A11-mediated glutathione synthesis in lung adenocarcinoma. Transl Lung Cancer Res. 11:366–380. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wu L, Li N, Zhu L and Shao G: CircPDSS1 (hsa_circ_0017998) silencing induces ferroptosis in non-small-cell lung cancer cells by modulating the miR-137/SLC7A11/GPX4/GCLC axis. Toxicol In Vitro. 99:1058872024. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Fan M, Zhou Z and Sang X: Circ_0082374 promotes the tumorigenesis and suppresses ferroptosis in non-small cell lung cancer by up-regulating GPX4 through sequestering miR-491-5p. Mol Biotechnol. 4: View Article : Google Scholar : 2024. | |
Fu H and Zhao Q: CircSCUBE3 promoted ferroptosis to inhibit lung adenocarcinoma progression. Cell Mol Biol (Noisy-le-grand). 70:161–168. 2024. View Article : Google Scholar : PubMed/NCBI | |
Shanshan W, Hongying M, Jingjing F, Yiming Y, Yu R and Rui Y: CircDTL functions as an oncogene and regulates both apoptosis and ferroptosis in non-small cell lung cancer cells. Front Genet. 12:7435052021. View Article : Google Scholar : PubMed/NCBI | |
Liu B, Ma H, Liu X and Xing W: CircSCN8A suppresses malignant progression and induces ferroptosis in non-small cell lung cancer by regulating miR-1290/ACSL4 axis. Cell Cycle. 22:758–776. 2023. View Article : Google Scholar : | |
Zhang X, Xu Y, Ma L, Yu K, Niu Y, Xu X, Shi Y, Guo S, Xue X, Wang Y, et al: Essential roles of exosome and circRNA_101093 on ferroptosis desensitization in lung adenocarcinoma. Cancer Commun (Lond). 42:287–313. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhang Q, Fan X, Zhang X and Ju S: Ferroptosis in tumors and its relationship to other programmed cell death: Role of non-coding RNAs. J Transl Med. 21:5142023. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Chen Y, Zhang J, Yang Y, Fleishman JS, Wang Y, Wang J, Chen J, Li Y and Wang H: Cuproptosis: A novel therapeutic target for overcoming cancer drug resistance. Drug Resist Updat. 72:1010182024. View Article : Google Scholar | |
Tang D and Kang R: NFE2L2 and ferroptosis resistance in cancer therapy. Cancer Drug Resist. 7:412024.PubMed/NCBI | |
Winkle M, El-Daly SM, Fabbri M and Calin GA: Noncoding RNA therapeutics-challenges and potential solutions. Nat Rev Drug Discov. 20:629–651. 2021. View Article : Google Scholar : PubMed/NCBI | |
Nappi F: Non-coding RNA-targeted therapy: A state-of-the-art review. Int J Mol Sci. 25:36302024. View Article : Google Scholar : PubMed/NCBI | |
He AT, Liu J, Li F and Yang BB: Targeting circular RNAs as a therapeutic approach: Current strategies and challenges. Signal Transduct Target Ther. 6:1852021. View Article : Google Scholar : PubMed/NCBI | |
Zhao R, Fu J, Zhu L, Chen Y and Liu B: Designing strategies of small-molecule compounds for modulating non-coding RNAs in cancer therapy. J Hematol Oncol. 15:142022. View Article : Google Scholar : PubMed/NCBI | |
Han B, Liu Y, Zhang Q and Liang L: Propofol decreases cisplatin resistance of non-small cell lung cancer by inducing GPX4-mediated ferroptosis through the miR-744-5p/miR-615-3p axis. J Proteomics. 274:1047772023. View Article : Google Scholar | |
Fu R, You Y, Wang Y, Wang J, Lu Y, Gao R, Pang M, Yang P and Wang H: Sanggenol L induces ferroptosis in non-small cell lung cancer cells via regulating the miR-26a-1-3p/MDM2/p53 signaling pathway. Biochem Pharmacol. 226:1163452024. View Article : Google Scholar : PubMed/NCBI | |
Huang J, Deng C, Guo T, Chen X, Chen P, Du S and Lu M: Cinobufotalin induces ferroptosis to suppress lung cancer cell growth by lncRNA LINC00597/hsa-miR-367-3p/TFRC pathway via resibufogenin. Anticancer Agents Med Chem. 23:717–725. 2023. View Article : Google Scholar |