Skip to main content

Role of ferroptosis and ferroptosis-related long non'coding RNA in breast cancer

Abstract

Ferroptosis, a therapeutic strategy for tumours, is a regulated cell death characterised by the increased accumulation of iron-dependent lipid peroxides (LPO). Tumour-associated long non-coding RNAs (lncRNAs), when combined with traditional anti-cancer medicines or radiotherapy, can improve efficacy and decrease mortality in cancer. Investigating the role of ferroptosis-related lncRNAs may help strategise new therapeutic options for breast cancer (BC). Herein, we briefly discuss the genes and pathways of ferroptosis involved in iron and reactive oxygen species (ROS) metabolism, including the XC/GSH/GPX4 system, ACSL4/LPCAT3/15-LOX and FSP1/CoQ10/NAD(P)H pathways, and investigate the correlation between ferroptosis and LncRNA in BC to determine possible biomarkers related to ferroptosis.

Introduction

The GLOBOCAN 2020 Global Cancer Burden report states that the risk of breast cancer (BC) is higher in women, which is the fourth leading cause of cancer-related deaths worldwide [1]. BC, which can be classified as either invasive or non-invasive [2], is a condition in which various carcinogens act on the breast epithelial cells, resulting in abnormal or uncontrolled proliferation [3]. BC is typically characterised by the presence of breast lumps, nipple discharge and increased/enlarged lymph nodes in the axilla. In advanced stages, cancer cells can metastasise to distant locations, resulting in multi-organ damage and increasing the risk of mortality [4, 5].

Programmed cell death and accidental cell death (ACD) are the two main types of cell death. Accidental attacks and injuries can trigger ACD, outweighing any control mechanism, and are regulated by precise signalling cascades triggered by specific effector molecules with distinct biochemical, functional and immunological effects [6]. The different forms of regulated cell death include apoptosis, necrosis, autophagy and ferroptosis. This study explored ferroptosis as a novel regulatory mode of cell death dependent on iron and lipotoxicity [6, 7], and several genes and pathways are involved in the modulation of iron and reactive oxygen species (ROS) metabolism, including the XC/GSH/GPX4 system and ACSL4/LPCAT3/15-LOX and FSP1/CoQ10/NAD(P)H pathways [8,9,10]. Dysregulation of iron metabolism, one of the risk factors for tumours, and the overdependence of cancer cells on iron proliferation promote the growth of tumour cells [11]. Activating ferroptosis in cancer cells is a novel approach to mitigating cancer risk, particularly for those resistant to conventional chemotherapy [12, 13]. Cilamethicin and lapatinib can trigger ferroptosis in BC cells, suggesting that they could serve as viable treatments for BC patients [14]. Nevertheless, studies analysing the relationship between BC and ferroptosis are lacking, and the influence of ferroptosis on the prognosis of patients with BC remains uncertain.

Long non-coding RNAs (lncRNAs) are RNA molecules that are longer than 200 nucleotides and lack the ability to encode proteins [15, 16]. Because of their unique involvement in cancer, lncRNAs have gained considerable attention [17]. Peptides or proteins encoded by tumour-associated lncRNAs increase efficacy and reduce mortality in combination with conventional cancer drugs and radiotherapy [18]. LncRNA-encoded ASRPS contributes to the progression of triple-negative breast cancer (TNBC), whereas the lncRNA HOXB cluster anti-sense RNA 3 (HOXB-AS3) peptide inhibits the growth of colorectal cancer (CRC) [19, 20]. Moreover, lncRNAs regulate ferroptosis in BC [21]. However, existing studies on lncRNAs related to ferroptosis in BC are scarce, and only a few lncRNAs involved in regulating ferroptosis have been identified. However, the recognition of the repetitive molecular mechanisms of lncRNAs has been made possible by emerging technologies that have enhanced the ability of researchers to functionally annotate cancer-related lncRNAs, such as the identification of potential ferroptosis-associated lncRNAs, which can be achieved using high-speed sequencing technologies [22].

This review discusses lncRNAs associated with the activation or inhibition of cellular ferroptosis, which exert anti-cancer effects, thereby providing potential insights for strategising new cancer treatment regimens. However, studies on ferroptosis-related lncRNAs in BC remain limited. Therefore, we briefly discuss the role of ferroptosis in BC, the association between BC and lncRNAs,and identify potential ferroptosis-related lncRNAs in BC.

Ferroptosis: a brief overview

Ferroptosis was first discovered in 2003 and involved the use of erastin to selectively induce cell death in genetically engineered cells with oncogenic RAS mutations, but not in normal cells [23]. Brent Stockwell, in 2012, coined the term ferroptosis for the iron-dependent cell death mode of non-apoptotic RCD induced by erastin [24]. Ferroptosis is a distinct type of regulated cell death mediated by iron and lipotoxicity that has been recently identified. Ferroptosis inhibits the activity of the lipid repair enzyme glutathione peroxidase 4 (GPX4), leading to the accumulation of lipid ROS, particularly lipid hydroperoxides [7]. In terms of genetics, multiple genes regulate ferroptosis. In contrast to other morphological changes associated with cell death, ferroptosis occurs primarily within the cell. This results in smaller mitochondria, increased membrane density, decreased and disappeared cristae, fragmentation of the outer membrane without disruption of the cell membrane and minimal transformation in the morphology of the nucleus without chromatin concentration [24,25,26]. Biochemically, the phospholipid peroxidase GPX4 primarily contributes to the deficiency in peroxidation repair capacity, acquisition of reactive iron and oxidation of polyunsaturated fatty acids (PUFA)-containing phospholipids that induce ferroptosis [27]. The intracellular antioxidant capacity decreases and lipid ROS accumulates, ultimately leading to cellular ferroptosis. Glutathione peroxidase is affected by various pathways, such as the XC/GSH/GPX4 system, and the ACSL4/LPCAT3/15-LOX and FSP1/CoQ10/NAD(P)H pathways [8,9,10] (Fig. 1).

Fig. 1
figure 1

Ferroptosis pathways. Genes and pathways of ferroptosis involved in the regulation of iron and ROS metabolism, including the system XC/GSH/GPX4, and ACSL4/LPCAT3/15-LOX and FSP1/CoQ10/NAD(P)H pathways. Created with https://www.biorender.com/

System XC /GSH/GPX4 and ferroptosis

Both impaired elimination and overproduction of lipid peroxide (LPO) during ferroptosis can lead to its accumulation to lethal levels. Cystine availability, glutathione (GSH) biosynthesis and GPX4 function are required to maintain redox homeostasis and protect cells from ferroptosis [24, 28, 29].

System XC/GSH/GPX4 is the antioxidant system that is crucial for ferroptosis [30]. System XC functions as a glutamate–cysteine reverse transporter at the plasma membrane, importing cysteine into the cytosol to facilitate GSH biosynthesis [31]. The inhibition of system XC results in a reduction in the intracellular cysteine pool, which is a precursor for glutathione synthesis [32]. GPX4 is the primary enzyme involved in the reduction and detoxification of phospholipid hydroperoxides (PLOOHs) in mammalian cells [33]; therefore, a general mechanism for the induction of ferroptosis by erastin/RSL3 has been identified. GSH peroxidase 4 inhibitor (RSL3) directly inactivates GPX4, whereas erastin indirectly inactivates it by inhibiting cysteine input, thereby depriving the cells of cysteine, an essential cellular component of GSH. Therefore, the accumulation of PLOOHs may cause rapid and irreversible damage to cell membranes, resulting in cell death.

GPX4 converts GSH to oxidised glutathione disulphide (GSSG), reduces LPO and maintains cell redox homeostasis [28]. Moreover, GPX4 is the only enzyme that directly reduces hydrogen peroxide from biofilm lipids [34]. Suppressing the system XC/GSH/GPX4 axis results in the accumulation of LPO, thereby leading to ferroptosis; for example, system XC activity is directly inhibited by the ferroptosis inducer erastin, which disrupts redox homeostasis and increases LPO accumulation, leading to ferroptosis [24]. Intracellular and extracellular cysteine are needed to maintain glutathione biosynthesis and inhibit mammalian cell death, which can also be treated with iron sequestrants or hydrophilic antioxidants [35].

ACSL4/LPCAT3/15-LOX with ferroptosis

Clustered regularly interspaced palindromic repeats (CRISPR)–Cas9 and genome-wide haploid-based screening analyses have identified two membrane turnover enzymes: lysophosphatidylcholine acyltransferase 3 (LPCAT3) and acyl-coenzyme A synthase long chain family member 4 (ACSL4) [36, 37] as key drivers of ferroptosis. These enzymes are essential for endogenous iron chain activation through metabolic lipid reprogramming [38]. ACSL4 is a prominent isoenzyme involved in the biometabolism of PUFAs and determines their susceptibility to ferroptosis [39]. Lipid synthesis-mediated production of PUFAs increases the susceptibility to ferroptosis [40]. The entry of PUFAs into phospholipids, a crucial step in ferroptosis, requires ACSL4 [40], which links coenzyme A to long-chain PUFAs, which are then transesterified into phospholipids by several LPCATases, increasing the integration of long-chain PUFAs into lipids and membranes [7].

Elevated ACSL4 expression increases the sensitivity of cells to ferroptosis by optimising the catalysis of several PUFAs, with a strong affinity for arachidonic acid (AA) and adrenaline (AdA). ACSL4 catalyses the conversion of AA and AdA into AACoA and AdA-CoA, respectively, resulting in LPO production. The derivatives were first esterified with LPCAT3 to form phosphatidylethanolamines (AA-PE and AdA-PE), followed by the direct oxidation of their lipid hydrogen peroxide by 15-LOX (ALOX15), which acts as a signal for ferric ions and, ultimately, promotes ferroptosis [31, 36, 41, 42]. Additionally, this process affects the cellular lipid composition [8, 36]. Therefore, ACSL4/LPCAT3/15-LOX may play an important role in the generation of lethal LPOs during ferroptosis.

ACSL4 determines the susceptibility to ferroptosis by modifying cellular lipid composition [36, 43]. The lipoxygenase enzyme, which contains iron, promotes cell death by producing LPO via lipid biosynthesis in ACSL4 [40]. ACSL4, a target of miR-424-5P, is upregulated in ovarian cancer (OC) and inhibits OC cell ferroptosis [44]. The inhibition of ACSL4 expression may be the primary mechanism that renders cells insensitive to iron leaps.

FSP1/CoQ10/NAD(P)H with ferroptosis

Apoptosis-inducing factor mitochondria-associated 2 (AIFM2), a member of the apoptosis-inducing factor (AIF) family, is involved in oxidoreductase function and can induce programmed cell death [45]. Recently, AIFM2 was recognised as an anti-iron porphyrin gene and was later renamed ferrocyte apoptosis suppressor protein 1 (FSP1). FSP1 inhibits iron through ubiquitin ketone (CoQ10), a reductant that scavenges the lipid peroxyl radicals responsible for lipid peroxidation. The use of FSP1 as a pharmacological target in combination with GPX4 inhibitors induces ferroptosis in various tumour types [46].

CoQ10 plays key roles in the mevalonate (MVA) pathway; regulating the MVA pathway could be a possible strategy for controlling the course of ferroptosis [47]. Following cardamoylation, FSP1 is recruited to the plasma membrane, where it acts as an oxidoreductase to catalyse the generation of ubiquitin from CoQ10 via NADPH. As a lipophilic anti-radical catcher, NADPH decreases LPO levels [46, 48]. Therefore, FSP1/CoQ10/NAD(P)H acts synergistically with GPX4 and GSH to protect against phospholipid peroxidation and ferroptosis [46].

Other genes and pathways for ferroptosis

P53 mediates cell cycle pausing, senescence and apoptosis, and its inactivation is a key factor in the formation of most tumours; therefore, the p53 gene is considered a potential tumour suppressor gene. Additionally, P53 is involved in various metabolic activities [49]. P53 downregulates the expression of solute carrier family 7 member 11 (SLC7A11) and inhibits the systemic uptake of cystine through GPX4 activity. This leads to reduced cellular antioxidant capacity and the accumulation of lipid ROS, resulting in ferroptosis [49, 50].

In addition, autophagy contributes to ferroptosis. Although ferroptosis can lead to the lipid peroxidation of plasma membranes, the major membrane modulator proteins remain unclear [51, 52]. Autophagy removes a wide range of components by forming dynamic membrane structures, such as phagosomes, autophagic vesicles and autophagosomes [53]. However, excessive autophagic activity mediates ferroptosis [54]. Autophagy facilitates swift, non-apoptotic, non-necrotic cell death during amino acid starvation. This condition triggers potent autophagy, but only if sufficient serum is provided in the culture medium, as it requires iron-supporting transferrin and the amino acid glutamine in saline. Death is triggered by cysteine deficiency in the cell growth media, which could be attributed to ferroptosis [55]. Autophagy is implicated in cysteine deprivation and is sensitised to ferroptosis through the autophagic degradation of ferritin, which is also termed ferritin autophagy, leading to increased levels of unstable iron in cells [56, 57]. For instance, HPCAL1 (hippocampalin-like protein 1) is a novel autophagic receptor that selectively degrades CDH2 (calbindin 2) during ferroptosis. HPCAL1 facilitates ferroptosis through its non-canonical role in autophagy. The CDH2/N-calbindin protein is a straightforward substrate for HPCAL1-dependent autophagic degradation and triggers ferroptosis by compromising membrane tension [52, 58].

In addition, methods for inducing or inhibiting ferroptosis have been extensively studied. For instance, glucose starvation inhibits ferroptosis [59, 60], whereas arachidonic acid enhances RSL3-mediated ferroptosis in mouse foetal fibroblasts[38]. Other signalling pathways that regulate cellular ferroptosis, such as Keleh-like ECH-associated protein 1 (Keap1), nuclear factor red lineage-associated factor 2 (Nrf2) and lymphoid tissue-specific deconjugating enzyme (LSH), have also been identified. Furthermore, EgI nine homologue 1 (EGLN1)/cellular myeloid cell tumour proto-oncogene (c-Myc), sulphur transfer, mucin 1C-terminal (MUC1-C)/ systemic XC (xCT) and heat-shock factor-1 (HSF1)/heat-shock protein beta 1 (HSPB1) pathways mediate ferroptosis [61].

Association between ferroptosis and BC

Ferroptosis has been implicated in the pathological processes of several disorders, such as neurological disorders, blood disorders, kidney damage, ischaemia–reperfusion injury and tumours. However, the natural mechanisms underlying the induction of ferroptosis under these conditions remain unclear. Ferroptosis is an oxidative stress-induced cell death process that is closely associated with cellular metabolism. Cancer cells, which have a more active metabolism and a higher ROS load, may have a stronger tendency towards ferroptosis. Because cellular ferroptosis inhibits tumour growth, targeting ferroptosis pathways could be a promising anti-cancer strategy [27]. In addition, ferroptosis occurs during cancer treatment. For example, low-density lipoprotein (LDL)–docosahexaenoic acid (DHA) nanoparticles and sorafenib induce ferroptosis in hepatocellular carcinoma cells [9, 62]. Cysteine dioxygenase 1 (CDO1) modulates erastin in vitro in gastric cancer cells [63], whereas cisplatin and dipeptidyl peptidase-4 (DPP4) regulate erastin-induced ferroptosis in colorectal cancer [64, 65]. Piperonylamine (PL), cyclophosphamide (CNA), liuzasulfapyridine combination, cottonin A (CN-A), phenethyl isothiocyanate (PEITC) and artesunate (ART) induce ROS generation, activation and ferroptosis in pancreatic cancer cell lines to inhibit proliferation [66, 67]. Erastin upregulates and activates P53, inhibits the activity of SLC7A11 and induces ferroptosis in lung cancer cells [68].

Cellular ferroptosis is recognised as a key mechanism by which certain chemotherapeutic agents induce cell death in cancer cells [65, 69]. BC is a diverse tumour; based on the hormone receptors (ER and PR) and HER2 (ERBB2) signatures, BC is clinically classified into three main subtypes: TNBC, tubular ER+ and PR+, and HER2+ [3, 70, 71]. The National Comprehensive Cancer Network (NCCN) recommendations recommends endocrine therapy for ER+ BC and anti-HER2-targeted therapy for HER2+ BC. Targeted therapies for TNBC are currently lacking [72]. Although the potential benefits of inducing ferroptosis in tumour therapy have been suggested, the genes related to ferroptosis have not been extensively studied in BC patients [73].

Treatment of TNBC remains challenging, and identifying the coordinated role of pathways in regulating ferroptosis will provide a fresh impetus for a therapeutic strategy for TNBC. TNBC was more sensitive to ferroptosis than ER+ BC [36]. TNBC can be divided into four categories: mesenchymal-like subtype (MES), luminal androgen receptor (LAR) subtype, immunomodulatory subtype (IM) and basal-like immunosuppressive subtype (BLIS) subtypes. The LAR subtype can induce ferroptosis using GPX4 inhibitors. This subtype is characterised by the upregulation of the oxidised phosphatidylethanolamine and glutathione metabolism (in particular, GPX4). Furthermore, inhibition of GPX4 not only leads to tumour ferroptosis but also enhances anti-tumour immune function [74]. By signalling epithelial-to-mesenchymal transition, MES cells can promote the activity of iron-connected molecules (as transferrin receptor 1, ferritin) and enhance iron uptake, storage and utilisation [10, 75]. Notably, this process is not limited to cancer cells but can also occur in non-cancerous cells. MES is characterised by an MES state enriched in iron metabolic pathways but lacking fatty acid (FA) metabolism and ROS pathway activity, indicating that, compared with LAR, MES subtypes are more susceptible to ferroptosis [76, 77]. Both IM and BLIS are characterised by typical stromal-like tumours in the presence of ferroptotic area [74].

One finding demonstrated that GPX4 expression was lower in BC MCF7 and MDA-MB-231 cell lines than in non-BC MCF10A cell lines [78], and that GPX4 expression was positively correlated with ER and PR labelling [79]. GPX4 may exert anti-tumour activity and reflect an improved differentiation phenotype in BC [73]. Erastin targets MDA-MB-231 cells selectively and effectively induces ferroptosis in TNBC cells [80].

Notably, ACSL4 expression levels in a subpopulation of TNBC cell lines were correlated with their sensitivity to ferroptosis reagents. This correlation appears to be similar to that observed in the treatment of refractory mesenchymal carcinoma cells and clear cell renal carcinoma cells [7]. ACSL4 is elevated in BC tissues compared with in healthy tissues adjacent to the cancer, and ACSL4 expression is negatively correlated with ER [81, 82]. Clinically, radiotherapy upregulates the expression of ACSL4, resulting in increased lipid synthesis and, consequently, oxidative injury, leading to ferroptosis [83]. High expression of ACSL4 promotes BC aggressiveness, is a potential prognostic indicator and therapeutic target [82], and plays a substantial role in radiation resistance in BC by modulating the expression of transporter proteins implicated in cancer resistance via the mTOR pathway and regulating forkhead box protein M1 (FOXM1) [84].

These findings indicate that ferroptosis may be an essential adaptation process for eradicating cancer cells [85].

Role of lncRNAs in BC

LncRNAs are RNA molecules that are ˃ 200 nucleotides long and lack the ability to encode proteins [86]. They are widely present in humans and are critical in regulating human gene expression and physiological and pathological processes [87]. LncRNAs can be broadly classified into the following three types: direct linking to DNA or transcription factors at the transcription level; binding of mRNAs, miRNAs or proteins to modulate their activity and steady state in a post-transcriptional manner; and interference with the chromatin complex to activate or suppress gene expression in an epigenetic manner [86, 88,89,90].

The mammalian genome contains numerous lncRNAs. A small but increasing number of these lncRNAs have functional profiles in various processes and diseases, such as infection, innate immunity and acquired immunity [91,92,93,94,95,96]. Cancer is a genetic disease that involves an alteration in the flow of information within the cell to alter cellular homeostasis and promote growth [22]. Non-coding RNAs regulate inter- and intracellular signalling in BC [97]. LncRNA-encoded peptides affect BC cells [98]. For instance, the micropeptide CIP2A-binding peptide (CIP2A-BP) encoded by LINC0665 is highly correlated with the survival of BC recipients. Poor CIP2A-BP expression is associated with low survival in BC patients. In addition, CIP2A-BP levels in patients with metastatic BC were markedly lower than in those without metastasis. Both the introduction of the CIP2A-BP gene and direct infusion of the CIP2A-BP micropeptide markedly attenuated lung metastasis and improved overall survival, suggesting that the micropeptide CIP2A-BP suppressed the migration and invasion of TNBC cells [99]. LINC00908 encodes ASRPS, a potential anti-cancer micropeptide that is endogenously expressed and downregulated in TNBC and inhibits tumour angiogenesis in BC [19].

Several lncRNAs that promote BC development have been identified and their functions have been investigated. This information can aid in the diagnosis, prognostic judgement, pathogenesis prediction and therapeutic intervention for BC (Table 1).

Table 1 Role of various lncRNAs in breast cancer

LncRNAs that inhibit BC development

NKILA, NEF,GAS5, MT1JP, LET, LncKLHDC7B and TFAP2A-AS1 prevent BC cell invasion and migration; NLIPMT, XIXT, MALAT1 and MEG3 inhibit distant metastasis in BC cells [100].

LINC02273 knockdown inhibits BC metastasis [101]. LncRNA GAS5 is frequently downregulated in several cancers. In BC, GAS5 activates several proteins, including DKK2, PTEN, SUFU, PDCD4 and FOXO1, via various miRNA-mediated competing endogenouse RNA (ceRNA) mechanisms. These mechanisms involve miR-196a-5p, miR-21, miR-221-3p, miR-222 and miR-378a-5p, which bind to multiple microRNA response elements (MREs) in GAS5 to upregulate the expression of BC suppressor proteins. Furthermore, through epigenetic and other mechanisms, GAS5 may enhance the sensitivity to several drugs and improve prognosis [102].

LncRNAs that promote BC development

Several lncRNAs that affect the invasiveness, proliferation and apoptosis of BC cells have been identified. For instance, LINC00461, DANCR, H19, HOX transcriptional anti-toxic intergenic RNAs (HOTAIR), LINC00152, LINC01857 and NEAT1 facilitate BC cell invasion and migration; and HOTAIR, H19, MALAT1, RP1 and HIF1A-AS2 promote BC cell long-distant metastasis. LncRNAs, including H19 [103], PRNCR1, HOTAIR [104], LSINCT5 [105], SRA [106], Smad7 [107], NEAT1 [108], LINC01296 [109] AFAP1-AS1 [110], GHET1 [111], BRAF [112] and SNHG12 [113] promote cell proliferation and inhibit apoptosis in BC. Some lncRNAs can promote BC cell resistance, such as UCA1 [114], CRALA [115], lnc-ATB [116], LINC00518 [117] and DSCAM-AS1 [118].

H19 is located in the human genome downstream of IGF2, and its levels are elevated in a variety of cancers, notably BC, promoting BC cell proliferation [103, 119]. H19 expression is significantly upregulated in tamoxifen-refractory BC cell lines and tissues, and silencing of H19 in MCF7/TAMR cells is sensitive to tamoxifen therapy in vivo and in vitro [120]. Metformin may cause ferroptosis in BC by blocking autophagy in H19 [121]. UCA1 inhibits p27, which partially contributes to its oncogenic role in BC. Overexpression of UCA1 is overexpressed causes hnRNP I in the cytoplasm to be recruited to UCA1, reducing the access of p27 to hnRNP I, inducing a cell cycle pause in the G1 stage; therefore, UCA1 could be a potential biomarker for BC diagnosis [122].

HOTAIR, a new type of lncRNAs belonging to a subclass of intergenic lncRNAs tightly regulates genes related to mammalian embryonic development [123]. HOTAIR expression is highly upregulated in BC, and silencing HOTAIR induces apoptosis and prevents cell proliferation. The mechanism of action involves linking miRNA and post-transcriptional networks to promote BC development [123, 124]. For instance, HOTAIR acts as a mediator between frizzled homologue 7 (FZD7) and miR-129-5p, and promotes epithelial–mesenchymal transition and metastasis, leading to BC progression. HOTAIR knockdown inhibited tumour growth in a xenograft mode, whereas killing of miR-129-5p reversed the silencing function of HOTAIR and FZD7 restored the suppressive function of miR-129-5p, suggesting that HOTAIR controls the miR-129-5p/FZD7 axis [125]. HOTAIR further facilitates BC metabolism by targeting miR-601 via a sponge mechanism to control AKT signalling, which is dependent on zinc finger E-box binding homology box 1 (ZEB1) [126]. HOTAIR overexpression in surgically resected early stage BC is a strong predictor of metastatic disease progression and overall survival [127].

The significance of cell signalling pathways in tumourigenesis, tumour progression and metastasis cannot be overlooked. LncRNAs affect aspects of tumourigenesis by participating in or interfering with these pathways and, consequently, they exhibit either an oncogenic or a tumour-suppressive role [128].

Ferroptosis-associated lncRNAs in BC

Diverse physiological conditions and pathological stressors trigger ferroptosis in humans and animals [24]. Ferroptosis has been increasingly recognised as an adaptive feature in the elimination of malignant tumours. The immune system plays a crucial role in the suppression of tumourigenesis, by removing cells that have been damaged by infection, environmental stress or a lack of key nutrients [182]. The classical oxidative stress pathway is an important therapeutic element that may contribute to ferroptosis. Despite the delicate balance between thiols and catalytic iron in cancer cells under sustained oxidative stress, this process occurs infrequently during cancer progression. However, the underlying molecular mechanisms remain unclear [13].

Abnormally expressed lncRNAs typically affect disease progression by regulating transcription and translation and can also influence cancer progression through the regulation of ferroptosis. The cytoplasmic lncRNA P53RRA in lung adenocarcinoma cancer cells binds to the structural portion of the Ras GTPase-activating protein-binding protein 1 (G3BP1) RNA recognition motif (RRM), leading to nuclear segregation of P53 and retained P53 in the nucleus and accumulation of lipid ROS in the nucleus, subsequently leading to cellular ferroptosis [21]. LncRNA LINC00618-induced ferroptosis increases lipid ROS and iron levels, and lowers SLC7A11 expression [183]. LncRNA GABPB1-AS1 modulates erastin-induced GABPB1 ferroptosis in HepG2 hepatoma cells [184]. Similarly, the GSK3β/Nrf2 signalling pathway is implicated in BC, which increases Nrf2 expression to counteract ferroptosis [185]. In addition, prominence protein 2 (PROM2) could reduce ferroptosis in BC cells and facilitate tumour progression by encouraging iron transport [186]. Because only specific lncRNAs are associated with ferroptosis, existing studies on ferroptosis-associated lncRNAs in BC are scarce.

RNA-sequencing data and one-way COX regression analyses in BC patients have led to the identification of 231 lncRNAs which affects the prognosis; 293 genes associated with ferroptosis were also downloaded from the Ferroptosis Database [187]. Furthermore, 11 lncRNAs (AC092916.1, L133467.1, USP30-AS1, AC108474.1, LINC01235, AL365356.1, AC072039.2, AC012213.3, LIPE-AS1, MAPT-AS1 and TDRKHAS1) that were significantly different were identified. Among them, lncRNA USP30-AS1 was co-expressed with nine ferroptosis-linked genes (SOCS1, CAPG, IFNG, PML, TNFAIP3, NCF2, SLC2A6, GCH1 and CYBB), suggesting that overexpression of USP30-AS1 in BC is associated with prolonged overall survival. LncRNA LIPE-AS1 interacts with five ferroptosis genes (GPX4, PHKG2, EGLN2, MAPK14 and HRAS), and improves the prognosis of BC patients. AC108474.1 interacts with five ferroptosis-related genes (HIC1, ISCU, PLIN4, CAV1 and TAZ), suggesting that AC108474.1 is also a protective factor for BC patients [188].

LncRNA HCP5 regulates baculoviral IAP repeat-containing 3 (BIRC3) by sponging miR-219a-5p as a ceRNA and promotes TNBC progression [189]. Moreover, the amino acid encoded by HCP5, HCP5-132aa promotes the malignant progression of TNBC through its dependence on GPX4 and lipid ROS levels. RNA sequencing results showed that silencing of the HCP5-132 amino acid (aa) open reading frame (ORF) resulted in an enrichment of differentially expressed genes (DEGS) associated with the ferroptosis pathway (which had a positive impact on intracellular Fe homeostasis, progesterone metabolic processes and cell proliferation), suggesting that disturbances in ferroptosis, progesterone metabolism and cell proliferation may affect BC development [190]. This resulted in an increase in the mitochondrial membrane density and a reduction in the mitochondrial cristae, with effects similar to those of erastin. In contrast, overexpression of HCP5-132aa ORF inhibited erastin-induced changes in mitochondrial morphology. Moreover, silencing of HCP5-132aa, along with the elevation of ROS levels when cells were primed with the ferroptosis enhancers RSL3 and erastin, was also counteracted by ferroptosis inhibitors and upregulation of HCP5-132aa. Furthermore, excessive expression of HCP5-132aa was associated with a worse patient prognosis, suggesting that HCP5-132aa might be a prognostic factor in TNBC [191] (see Fig. 2).

Fig. 2
figure 2

Ferroptosis-related lncRNAs in breast cancer. Created with https://www.biorender.com/

Knockdown of RUNX1 intronic transcript 1 (RUNX1-IT1), a newly identified lncRNA that plays a key role in breast carcinogenesis, was significantly overexpressed in human BC tissues, inhibited BC cell survival and invasion, and suppressed tumour growth in an in situ transplantation model. Furthermore, RUNX1-IT1 inhibited ferroptosis by increasing GPX4 expression. RUNXI-IT1 specifically binds directly to the N6-methyladenosine M6A reader, IGF2BP1, and promotes the assembly of the IGF2BP1 liquid–liquid phase (LLP) biomolecule condensate site, resulting in IGF2BP1 greater occupancy of GPX4 mRNA and increased GPX4 mRNA stability. The elevated GPX4 expression prevent lipid peroxidation and ferroptosis, thereby promoting BC development, which indicates that the abnormal regulation of RUNX1-IT1/IGF2BP1/GPX4 is associated with BC development [192].

Ferroptosis-associated lncRNAs can serve as prognostic indicators for constructing a prognostic map of BC based on early warning signs, treatment goals and the anti-tumour immune microenvironment of BC to guide clinical therapy. A previous study screened 11 lncRNAs associated with ferroptosis from the TCGA dataset and built a prognostic map. Based on differences in the expression levels of ferroptosis-associated lncRNAs in tissues from BC patients and healthy tissues, patients were grouped into high- and low-risk clusters. In this study, three genes, lncRNAs YTHDF3-AS1, AC079298.3 and AC012213.3, which are overexpressed in at-risk populations, were screened, indicating that they may be at-risk oncogenes for BC. In addition, lncRNA NC012213.3, a downstream molecule of AC012213.3, and overexpressed LncRNA AC012213.3 promotes BC multiplication, invasion and invasiveness through the RAD54B/PI3K/AKT axis and is associated with poor patient outcomes [193]. However, studies on YTHDF3-AS1 and AC079298.3 are limited and require further investigation. To date, lncRNA-associated ferroptosis particularly its association with BC has not been documented, warranting further research to identify new therapeutic targets for BC [190].

Conclusions

Our study has comprehensively elucidated the underlying mechanism of ferroptosis in relation to BC, providing potential insights for strategising new approaches for anti-tumour therapy. Ferroptosis occurs when there is an imbalance between the detoxification and accumulation of lipid hydroperoxides [194]. A stressful environment can cause an imbalance in lipid ROS accumulation, which contributes to ferroptosis [195]. Cancer cells typically promote survival and metastasis by resisting ferroptosis. Certain drugs used in BC treatment [196], such as cyclophosphamide, tamoxifen, paclitaxel and anthracycline, may induce excessive ROS generation, resulting in cell death [197,198,199,200]. LncRNAs are common transcription products in human and mammalian genomes. The roles of certain lncRNAs in tumourigenesis and tumour development have gained considerable attention, with established functions elucidated in primary studies. The disclosure of the universal genetic code has enabled researchers to identify defects in functional proteins; however, understanding the impact of lncRNA biology on cellular function remains challenging using existing predictive frameworks [22]. Owing to their enhanced efficiency, tissue specificity and stability, lncRNAs have the potential to be initial diagnostic and therapeutic targets [190].

Therefore, understanding the relationship between lncRNAs and ferroptosis, as well as their regulatory mechanisms in BC, can be beneficial for the development and therapy of this disease. Our findings demonstrated that lncRNA HCP5 encodes a novel protein, HCP5-132aa, and promotes TNBC growth by controlling GPX4 and inhibiting ROS levels, ultimately inhibiting ferroptosis. Patients with TNBC who overexpress HCP5-132aa typically have worse disease characteristics and prognoses [191]. Ferroptosis-associated lncRNAs may play a prognostic role in BC, enabling the construction of a prognostic model for screening markers, therapeutic targets, evaluating the anti-tumour immune microenvironment and guiding clinical therapy [190]. Nevertheless, future studies investigating the correlation between lncRNAs and ferroptosis and their underlying mechanisms in BC are warranted.

Availability of data and materials

Not applicable.

Abbreviations

AA:

Arachidonic acid

ACD:

Accidental cell death

ACSL4:

Acyl-Coenzyme A synthase long chain family member 4

AdA:

Adrenaline

AIF:

Apoptosis-inducing factor

AIFM2:

Apoptosis-inducing factor mitochondria-associated 2

ALOX15:

AdA-PE to lipid hydroperoxides by 15-LOX

ART:

Artesunate

BC:

Breast cancer

BIRC3:

Baculoviral IAP repeat containing 3

BLIS:

Basal-like immunosuppressive subtype

CAN:

Cyclophosphamide

CCTs:

Circulating tumor cells

CDH2:

Calbindin 2

CDO1:

Cysteine dioxygenase 1

CRC:

Colorectal cancer

CIP2A-BP:

CIP2A-binding peptide

c-Myc:

Cellular myeloid cell tumor proto-oncogene

CNA:

Combination of cottonin A

CoQ10:

Ubiquitin ketone

DHA:

Docosahexaenoic acid

DPP4:

Dipeptidyl peptidase-4

EGLN1:

EgI nine homologue 1

FA:

Fatty acid

FINs:

Ferroptosis inducers

FOXM1:

Forkhead box protein M1

FSP1:

Ferrocyte apoptosis suppressor protein 1

FZD7:

Frizzled homolog 7

Gas5:

Growth inhibitory specific 5

GPX4:

Glutathione peroxidase 4

GSH:

Glutathione

GSSG:

Glutathione disulphide

G3BP1:

Ras GTPase-activating protein-binding protein 1

HOTAIR:

HOX transcriptional anti-sense intergenic RNAs

HPCAL1:

Hippocampalin-like protein 1

HSF-1:

Heat-shock factor-1

HSPB1:

Heat-shock protein beta 1

IM:

Immunomodulatory subtype

Keap1:

Keleh-like ECH-associated protein 1

LAR:

Luminal androgen receptor subtype

LDHA:

Lactate dehydrogenase a

LDL:

Low-density lipoprotein

LncRNA:

Long non-coding RNA

LLP:

Liquid–liquid phase

LOXs:

Lipoxygenases

LPO:

Lipid peroxide

LPCAT3:

Lysophosphatidylcholine acyltransferase 3

LSH:

Lymphoid tissue-specific deconjugating enzyme

MES:

Mesenchymal-like subtype

MEG3:

Maternal expression 3

MUC1-C:

Mucin 1C-terminal

MVA:

Mevalonate

NCCN:

National Comprehensive Cancer Network

Nrf2:

Nuclear factor red lineage-associated factor 2

OC:

Ovarian cancer

PEITC:

Phenethyl isothiocyanate

PL:

Piperonylamine

PLOOH:

Phospholipid hydroperoxides

PROM2:

Prominence protein 2

PTBP1:

Polypyrimidine tract-binding protein 1

PUFA:

Polyunsaturated fatty acids

RCD:

Programmed cell death

ROS:

Reactive oxygen species

RRM:

RNA recognition motif

RUNX1-IT1:

RUNX1 intronic transcript 1

RSL3:

GSH peroxidase 4 inhibitor

SLC7A11:

Solute carrier family 7 member 11

TGF:

Transforming growth factor

TNBC:

Triple-negative breast cancer

xCT:

Systemic XC

ZEB1:

Zinc finger E box-binding homology box 1

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.

    Article  PubMed  Google Scholar 

  2. Akram M, Iqbal M, Daniyal M, Khan AU. Awareness and current knowledge of breast cancer. Biol Res. 2017;50:33.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Loibl S, Poortmans P, Morrow M, Denkert C, Curigliano G. Breast cancer. Lancet (London, England). 2021;397:1750–69.

    Article  CAS  PubMed  Google Scholar 

  4. Alkabban FM, Ferguson T. Breast Cancer, StatPearls, StatPearls Publishing Copyright © 2023. Treasure Island: StatPearls Publishing LLC.; 2023.

    Google Scholar 

  5. Harbeck N, Penault-Llorca F, Cortes J, Gnant M, Houssami N, Poortmans P, Ruddy K, Tsang J, Cardoso F. Breast cancer. Nat Rev Dis Primers. 2019;5:66.

    Article  PubMed  Google Scholar 

  6. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res. 2019;29:347–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22:266–82.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Li D, Li Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct Target Ther. 2020;5:108.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Louandre C, Marcq I, Bouhlal H, Lachaier E, Godin C, Saidak Z, François C, Chatelain D, Debuysscher V, Barbare JC, Chauffert B, Galmiche A. The retinoblastoma (Rb) protein regulates ferroptosis induced by sorafenib in human hepatocellular carcinoma cells. Cancer Lett. 2015;356:971–7.

    Article  CAS  PubMed  Google Scholar 

  10. Wu J, Minikes AM, Gao M, Bian H, Li Y, Stockwell BR, Chen ZN, Jiang X. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature. 2019;572:402–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Manz DH, Blanchette NL, Paul BT, Torti FM, Torti SV. Iron and cancer: recent insights. Ann NY Acad Sci. 2016;1368:149–61.

    Article  CAS  PubMed  Google Scholar 

  12. Hassannia B, Vandenabeele P, Vanden Berghe T. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.

    Article  CAS  PubMed  Google Scholar 

  13. Mou Y, Wang J, Wu J, He D, Zhang C, Duan C, Li B. Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol. 2019;12:34.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Ma S, Henson ES, Chen Y, Gibson SB. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7: e2307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Guttman M, Rinn JL. Modular regulatory principles of large non-coding RNAs. Nature. 2012;482:339–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Safi A, Saberiyan M, Sanaei MJ, Adelian S, Davarani Asl F, Zeinaly M, Shamsi M, Ahmadi R. The role of noncoding RNAs in metabolic reprogramming of cancer cells. Cell Mol Biol Lett. 2023;28:37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, Cabili MN, Jaenisch R, Mikkelsen TS, Jacks T, Hacohen N, Bernstein BE, Kellis M, Regev A, Rinn JL, Lander ES. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wu P, Mo Y, Peng M, Tang T, Zhong Y, Deng X, Xiong F, Guo C, Wu X, Li Y, Li X, Li G, Zeng Z, Xiong W. Emerging role of tumor-related functional peptides encoded by lncRNA and circRNA. Mol Cancer. 2020;19:22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang Y, Wu S, Zhu X, Zhang L, Deng J, Li F, Guo B, Zhang S, Wu R, Zhang Z, Wang K, Lu J, Zhou Y. LncRNA-encoded polypeptide ASRPS inhibits triple-negative breast cancer angiogenesis. J Exp Med. 2020;217:e20190950.

    Article  Google Scholar 

  20. Huang JZ, Chen M, Chen D, Gao XC, Zhu S, Huang H, Hu M, Zhu H, Yan GR. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cell. 2017;68:171-184.e176.

    Article  CAS  PubMed  Google Scholar 

  21. Mao C, Wang X, Liu Y, Wang M, Yan B, Jiang Y, Shi Y, Shen Y, Liu X, Lai W, Yang R, Xiao D, Cheng Y, Liu S, Zhou H, Cao Y, Yu W, Muegge K, Yu H, Tao Y. A G3BP1-interacting lncRNA promotes ferroptosis and apoptosis in cancer via nuclear sequestration of p53. Can Res. 2018;78:3484–96.

    Article  CAS  Google Scholar 

  22. Schmitt AM, Chang HY. Long noncoding RNAs in cancer pathways. Cancer Cell. 2016;29:452–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3:285–96.

    Article  CAS  PubMed  Google Scholar 

  24. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B 3rd, Stockwell BR. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Abrams RP, Carroll WL, Woerpel KA. Five-membered ring peroxide selectively initiates ferroptosis in cancer cells. ACS Chem Biol. 2016;11:1305–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B, Wang G. Ferroptosis: past, present and future. Cell Death Dis. 2020;11:88.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Li Z, Chen L, Chen C, Zhou Y, Hu D, Yang J, Chen Y, Zhuo W, Mao M, Zhang X, Xu L, Wang L, Zhou J. Targeting ferroptosis in breast cancer. Biomark Res. 2020;8:58.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, Brown LM, Girotti AW, Cornish VW, Schreiber SL, Stockwell BR. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Seiler A, Schneider M, Förster H, Roth S, Wirth EK, Culmsee C, Plesnila N, Kremmer E, Rådmark O, Wurst W, Bornkamm GW, Schweizer U, Conrad M. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab. 2008;8:237–48.

    Article  CAS  PubMed  Google Scholar 

  30. Li FJ, Long HZ, Zhou ZW, Luo HY, Xu SG, Gao LC. System X(c) (-)/GSH/GPX4 axis: an important antioxidant system for the ferroptosis in drug-resistant solid tumor therapy. Front Pharmacol. 2022;13: 910292.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, Kapralov AA, Amoscato AA, Jiang J, Anthonymuthu T, Mohammadyani D, Yang Q, Proneth B, Klein-Seetharaman J, Watkins S, Bahar I, Greenberger J, Mallampalli RK, Stockwell BR, Tyurina YY, Conrad M, Bayır H. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81–90.

    Article  CAS  PubMed  Google Scholar 

  32. Yang WS, Stockwell BR. Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 2016;26:165–76.

    Article  CAS  PubMed  Google Scholar 

  33. Ursini F, Maiorino M, Valente M, Ferri L, 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. 1982;710:197–211.

    Article  CAS  PubMed  Google Scholar 

  34. Brigelius-Flohé R, Maiorino M. Glutathione peroxidases. Biochim Biophys Acta. 1830;2013:3289–303.

    Google Scholar 

  35. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascon S, Hatzios SK, Kagan VE, Noel K, Jiang X, Linkermann A, Murphy ME, Overholtzer M, Oyagi A, Pagnussat GC, Park J, Ran Q, Rosenfeld CS, Salnikow K, Tang D, Torti FM, Torti SV, Toyokuni S, Woerpel KA, Zhang DD. Ferroptosis: a regulated cell death nexus linking metabolism. Redox Biol Dis Cell. 2017;171:273–85.

    CAS  Google Scholar 

  36. Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, Prokisch H, Trümbach D, Mao G, Qu F, Bayir H, Füllekrug J, Scheel CH, Wurst W, Schick JA, Kagan VE, Angeli JP, Conrad M. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13:91–8.

    Article  CAS  PubMed  Google Scholar 

  37. Zou Y, Palte MJ, Deik AA, Li H, Eaton JK, Wang W, Tseng YY, Deasy R, Kost-Alimova M, Dančík V, Leshchiner ES, Viswanathan VS, Signoretti S, Choueiri TK, Boehm JS, Wagner BK, Doench JG, Clish CB, Clemons PA, Schreiber SL. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat Commun. 2019;10:1617.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Liao P, Wang W, Wang W, Kryczek I, Li X, Bian Y, Sell A, Wei S, Grove S, Johnson JK, Kennedy PD, Gijón M, Shah YM, Zou W. CD8(+) T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell. 2022;40:365-378.e366.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cui Y, Zhang Y, Zhao X, Shao L, Liu G, Sun C, Xu R, Zhang Z. ACSL4 exacerbates ischemic stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav Immun. 2021;93:312–21.

    Article  CAS  PubMed  Google Scholar 

  40. Chen X, Li J, Kang R, Klionsky DJ, Tang D. Ferroptosis: machinery and regulation. Autophagy. 2021;17:2054–81.

    Article  CAS  PubMed  Google Scholar 

  41. Küch EM, Vellaramkalayil R, Zhang I, Lehnen D, Brügger B, Sreemmel W, Ehehalt R, Poppelreuther M, Füllekrug J. Differentially localized acyl-CoA synthetase 4 isoenzymes mediate the metabolic channeling of fatty acids towards phosphatidylinositol. Biochim Biophys Acta. 1841;2014:227–39.

    Google Scholar 

  42. Soupene E, Kuypers FA. Mammalian long-chain acyl-CoA synthetases. Exp Biol Med (Maywood). 2008;233:507–21.

    Article  CAS  PubMed  Google Scholar 

  43. Yuan H, Li X, Zhang X, Kang R, Tang D. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem Biophys Res Commun. 2016;478:1338–43.

    Article  CAS  PubMed  Google Scholar 

  44. Ma LL, Liang L, Zhou D, Wang SW. Tumor suppressor miR-424-5p abrogates ferroptosis in ovarian cancer through targeting ACSL4. Neoplasma. 2021;68:165–73.

    Article  CAS  PubMed  Google Scholar 

  45. Novo N, Ferreira P, Medina M. The apoptosis-inducing factor family: Moonlighting proteins in the crosstalk between mitochondria and nuclei. IUBMB Life. 2021;73:568–81.

    Article  CAS  PubMed  Google Scholar 

  46. Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Goya Grocin A, Xavier da Silva TN, Panzilius E, Scheel CH, Mourão A, Buday K, Sato M, Wanninger J, Vignane T, Mohana V, Rehberg M, Flatley A, Schepers A, Kurz A, White D, Sauer M, Sattler M, Tate EW, Schmitz W, Schulze A, O’Donnell V, Proneth B, Popowicz GM, Pratt DA, Angeli JPF, Conrad M. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575:693–8.

    Article  CAS  PubMed  Google Scholar 

  47. Warner GJ, Berry MJ, Moustafa ME, Carlson BA, Hatfield DL, Faust JR. Inhibition of selenoprotein synthesis by selenocysteine tRNA[Ser]Sec lacking isopentenyladenosine. J Biol Chem. 2000;275:28110–9.

    Article  CAS  PubMed  Google Scholar 

  48. Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, Bassik MC, Nomura DK, Dixon SJ, Olzmann JA. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575:688–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R, Gu W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Jiang L, Hickman JH, Wang SJ, Gu W. Dynamic roles of p53-mediated metabolic activities in ROS-induced stress responses. Cell Cycle. 2015;14:2881–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lin Z, Liu J, Kang R, Yang M, Tang D. Lipid metabolism in ferroptosis. Adv Biol. 2021;5: e2100396.

    Article  Google Scholar 

  52. Chen X, Song X, Li J, Zhang R, Yu C, Zhou Z, Liu J, Liao S, Klionsky DJ, Kroemer G, Liu J, Tang D, Kang R. Identification of HPCAL1 as a specific autophagy receptor involved in ferroptosis. Autophagy. 2023;19:54–74.

    Article  CAS  PubMed  Google Scholar 

  53. Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 2018;19:349–64.

    Article  CAS  PubMed  Google Scholar 

  54. Liu J, Kuang F, Kroemer G, Klionsky DJ, Kang R, Tang D. Autophagy-dependent ferroptosis: machinery and regulation, cell. Chem Biol. 2020;27:420–35.

    Google Scholar 

  55. Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59:298–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ 3rd, Kang R, Tang D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12:1425–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X. Ferroptosis is an autophagic cell death process. Cell Res. 2016;26:1021–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Rebaud S, Wang CK, Sarkis J, Mason L, Simon A, Blum LJ, Hofmann A, Girard-Egrot AP. Specific interaction to PIP2 increases the kinetic rate of membrane binding of VILIPs, a subfamily of neuronal calcium sensors (NCS) proteins. Biochim Biophys Acta. 1838;2014:2698–707.

    Google Scholar 

  59. Lee H, Zandkarimi F, Zhang Y, Meena JK, Kim J, Zhuang L, Tyagi S, Ma L, Westbrook TF, Steinberg GR, Nakada D, Stockwell BR, Gan B. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol. 2020;22:225–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Li C, Dong X, Du W, Shi X, Chen K, Zhang W, Gao M. LKB1-AMPK axis negatively regulates ferroptosis by inhibiting fatty acid synthesis. Signal Transduct Target Ther. 2020;5:187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life Sci. 2016;73:2195–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ou W, Mulik RS, Anwar A, McDonald JG, He X, Corbin IR. Low-density lipoprotein docosahexaenoic acid nanoparticles induce ferroptotic cell death in hepatocellular carcinoma. Free Radic Biol Med. 2017;112:597–607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Hao S, Yu J, He W, Huang Q, Zhao Y, Liang B, Zhang S, Wen Z, Dong S, Rao J, Liao W, Shi M. Cysteine dioxygenase 1 mediates erastin-induced ferroptosis in human gastric cancer cells. Neoplasia. 2017;19:1022–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, Zhong M, Yuan H, Zhang L, Billiar TR, Lotze MT, Zeh HJ 3rd, Kang R, Kroemer G, Tang D. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20:1692–704.

    Article  CAS  PubMed  Google Scholar 

  65. Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, Dai X, Li Z, Wu G. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer Res Treat. 2018;50:445–60.

    Article  CAS  PubMed  Google Scholar 

  66. Eling N, Reuter L, Hazin J, Hamacher-Brady A, Brady NR. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience. 2015;2:517–32.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Yamaguchi Y, Kasukabe T, Kumakura S. Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int J Oncol. 2018;52:1011–22.

    CAS  PubMed  Google Scholar 

  68. Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, Sabatini DM, Birsoy K, Possemato R. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 2017;551:639–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhang X, Sui S, Wang L, Li H, Zhang L, Xu S, Zheng X. Inhibition of tumor propellant glutathione peroxidase 4 induces ferroptosis in cancer cells and enhances anticancer effect of cisplatin. J Cell Physiol. 2020;235:3425–37.

    Article  CAS  PubMed  Google Scholar 

  70. Yeo SK, Guan JL. Breast cancer: multiple subtypes within a tumor? Trends Cancer. 2017;3:753–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Denkert C, Liedtke C, Tutt A, von Minckwitz G. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet (London, England). 2017;389:2430–42.

    Article  CAS  PubMed  Google Scholar 

  72. Gradishar WJ, Moran MS, Abraham J, Aft R, Agnese D, Allison KH, Blair SL, Burstein HJ, Dang C, Elias AD, Giordano SH, Goetz MP, Goldstein LJ, Hurvitz SA, Isakoff SJ, Jankowitz RC, Javid SH, Krishnamurthy J, Leitch M, Lyons J, Matro J, Mayer IA, Mortimer J, O’Regan RM, Patel SA, Pierce LJ, Rugo HS, Sitapati A, Smith KL, Smith ML, Soliman H, Stringer-Reasor EM, Telli ML, Ward JH, Wisinski KB, Young JS, Burns JL, Kumar R. NCCN guidelines® insights: breast cancer, version 4.2021. J Natl Compr Canc Netw. 2021;19:484–93.

    Article  PubMed  Google Scholar 

  73. Sha R, Xu Y, Yuan C, Sheng X, Wu Z, Peng J, Wang Y, Lin Y, Zhou L, Xu S, Zhang J, Yin W, Lu J. Predictive and prognostic impact of ferroptosis-related genes ACSL4 and GPX4 on breast cancer treated with neoadjuvant chemotherapy. EBioMedicine. 2021;71: 103560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yang F, Xiao Y, Ding JH, Jin X, Ma D, Li DQ, Shi JX, Huang W, Wang YP, Jiang YZ, Shao ZM. Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab. 2023;35:84-100.e108.

    Article  CAS  PubMed  Google Scholar 

  75. Müller S, Sindikubwabo F, Cañeque T, Lafon A, Versini A, Lombard B, Loew D, Wu TD, Ginestier C, Charafe-Jauffret E, Durand A, Vallot C, Baulande S, Servant N, Rodriguez R. CD44 regulates epigenetic plasticity by mediating iron endocytosis. Nat Chem. 2020;12:929–38.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Gong Y, Ji P, Yang YS, Xie S, Yu TJ, Xiao Y, Jin ML, Ma D, Guo LW, Pei YC, Chai WJ, Li DQ, Bai F, Bertucci F, Hu X, Jiang YZ, Shao ZM. Metabolic-pathway-based subtyping of triple-negative breast cancer reveals potential therapeutic targets. Cell Metab. 2021;33:51-64.e59.

    Article  CAS  PubMed  Google Scholar 

  77. Xiao Y, Ma D, Yang YS, Yang F, Ding JH, Gong Y, Jiang L, Ge LP, Wu SY, Yu Q, Zhang Q, Bertucci F, Sun Q, Hu X, Li DQ, Shao ZM, Jiang YZ. Comprehensive metabolomics expands precision medicine for triple-negative breast cancer. Cell Res. 2022;32:477–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Rusolo F, Capone F, Pasquale R, Angiolillo A, Colonna G, Castello G, Costantini M, Costantini S. Comparison of the seleno-transcriptome expression between human non-cancerous mammary epithelial cells and two human breast cancer cell lines. Oncol Lett. 2017;13:2411–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Cejas P, García-Cabezas MA, Casado E, Belda-Iniesta C, De Castro J, Fresno JA, Sereno M, Barriuso J, Espinosa E, Zamora P, Feliu J, Redondo A, Hardisson DA, Renart J, González-Barón M. Phospholipid hydroperoxide glutathione peroxidase (PHGPx) expression is downregulated in poorly differentiated breast invasive ductal carcinoma. Free Radic Res. 2007;41:681–7.

    Article  CAS  PubMed  Google Scholar 

  80. Yu M, Gai C, Li Z, Ding D, Zheng J, Zhang W, Lv S, Li W. Targeted exosome-encapsulated erastin induced ferroptosis in triple negative breast cancer cells. Cancer Sci. 2019;110:3173–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Monaco ME, Creighton CJ, Lee P, Zou X, Topham MK, Stafforini DM. Expression of long-chain fatty acyl-coA synthetase 4 in breast and prostate cancers is associated with sex steroid hormone receptor negativity. Transl Oncol. 2010;3:91–8.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Dinarvand N, Khanahmad H, Hakimian SM, Sheikhi A, Rashidi B, Pourfarzam M. Evaluation of long-chain acyl-coenzyme A synthetase 4 (ACSL4) expression in human breast cancer. Res Pharm Sci. 2020;15:48–56.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, Ajani JA, Xiao Q, Liao Z, Wang H, Gan B. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30:146–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Kwon YS, Lee MG, Baek J, Kim NY, Jang H, Kim S. Acyl-CoA synthetase-4 mediates radioresistance of breast cancer cells by regulating FOXM1. Biochem Pharmacol. 2021;192: 114718.

    Article  CAS  PubMed  Google Scholar 

  85. Dixon SJ. Ferroptosis: bug or feature? Immunol Rev. 2017;277:150–7.

    Article  CAS  PubMed  Google Scholar 

  86. Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22:96–118.

    Article  CAS  PubMed  Google Scholar 

  87. Mei J, Hao L, Wang H, Xu R, Liu Y, Zhu Y, Liu C. Systematic characterization of non-coding RNAs in triple-negative breast cancer. Cell Prolif. 2020;53: e12801.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Marchese FP, Raimondi I, Huarte M. The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 2017;18:206.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Salviano-Silva A, Lobo-Alves SC, Almeida RC, Malheiros D, Petzl-Erler ML. Besides pathology: long non-coding RNA in cell and tissue homeostasis. Noncoding RNA. 2018;4:3.

    PubMed  PubMed Central  Google Scholar 

  90. Fernandes JCR, Acuña SM, Aoki JI, Floeter-Winter LM, Muxel SM. Long non-coding RNAs in the regulation of gene expression: physiology and disease. Noncoding RNA. 2019;5:17.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Wang P, Xu J, Wang Y, Cao X. An interferon-independent lncRNA promotes viral replication by modulating cellular metabolism. Science (New York, NY). 2017;358:1051–5.

    Article  CAS  PubMed  Google Scholar 

  92. Gomez JA, Wapinski OL, Yang YW, Bureau JF, Gopinath S, Monack DM, Chang HY, Brahic M, Kirkegaard K. The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus. Cell. 2013;152:743–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Fortes P, Morris KV. Long noncoding RNAs in viral infections. Virus Res. 2016;212:1–11.

    Article  CAS  PubMed  Google Scholar 

  94. Castellanos-Rubio A, Fernandez-Jimenez N, Kratchmarov R, Luo X, Bhagat G, Green PH, Schneider R, Kiledjian M, Bilbao JR, Ghosh S. A long noncoding RNA associated with susceptibility to celiac disease. Science (New York, NY). 2016;352:91–5.

    Article  CAS  PubMed  Google Scholar 

  95. Atianand MK, Hu W, Satpathy AT, Shen Y, Ricci EP, Alvarez-Dominguez JR, Bhatta A, Schattgen SA, McGowan JD, Blin J, Braun JE, Gandhi P, Moore MJ, Chang HY, Lodish HF, Caffrey DR, Fitzgerald KA. A long noncoding RNA lincRNA-EPS acts as a transcriptional brake to restrain inflammation. Cell. 2016;165:1672–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ranzani V, Rossetti G, Panzeri I, Arrigoni A, Bonnal RJ, Curti S, Gruarin P, Provasi E, Sugliano E, Marconi M, De Francesco R, Geginat J, Bodega B, Abrignani S, Pagani M. The long intergenic noncoding RNA landscape of human lymphocytes highlights the regulation of T cell differentiation by linc-MAF-4. Nat Immunol. 2015;16:318–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Klinge CM. Non-Coding RNAs in breast cancer: intracellular and intercellular communication. Noncoding RNA. 2018;4:40.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Jin H, Du W, Huang W, Yan J, Tang Q, Chen Y, Zou Z. lncRNA and breast cancer: progress from identifying mechanisms to challenges and opportunities of clinical treatment. Mol Ther Nucleic Acids. 2021;25:613–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Guo B, Wu S, Zhu X, Zhang L, Deng J, Li F, Wang Y, Zhang S, Wu R, Lu J, Zhou Y. Micropeptide CIP2A-BP encoded by LINC00665 inhibits triple-negative breast cancer progression. EMBO J. 2020;39: e102190.

    Article  CAS  PubMed  Google Scholar 

  100. Huang QY, Liu GF, Qian XL, Tang LB, Huang QY, Xiong LX. Long non-coding RNA: dual effects on breast cancer metastasis and clinical applications. Cancers. 2019;11:1802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Xiu B, Chi Y, Liu L, Chi W, Zhang Q, Chen J, Guo R, Si J, Li L, Xue J, Shao ZM, Wu ZH, Huang S, Wu J. LINC02273 drives breast cancer metastasis by epigenetically increasing AGR2 transcription. Mol Cancer. 2019;18:187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Zhang Z, Zhu Z, Watabe K, Zhang X, Bai C, Xu M, Wu F, Mo YY. Negative regulation of lncRNA GAS5 by miR-21. Cell Death Differ. 2013;20:1558–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Berteaux N, Lottin S, Monté D, Pinte S, Quatannens B, Coll J, Hondermarck H, Curgy JJ, Dugimont T, Adriaenssens E. H19 mRNA-like noncoding RNA promotes breast cancer cell proliferation through positive control by E2F1. J Biol Chem. 2005;280:29625–36.

    Article  CAS  PubMed  Google Scholar 

  104. Shi F, Chen X, Wang Y, Xie Y, Zhong J, Su K, Li M, Li Y, Lin Q, Zhou Y, Wang J, Xiong L. HOTAIR/miR-203/CAV1 crosstalk influences proliferation, migration, and invasion in the breast cancer cell. Int J Mol Sci. 2022;23:11755.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Silva JM, Boczek NJ, Berres MW, Ma X, Smith DI. LSINCT5 is over expressed in breast and ovarian cancer and affects cellular proliferation. RNA Biol. 2011;8:496–505.

    Article  CAS  PubMed  Google Scholar 

  106. Lanz RB, Chua SS, Barron N, Söder BM, DeMayo F, O’Malley BW. Steroid receptor RNA activator stimulates proliferation as well as apoptosis in vivo. Mol Cell Biol. 2003;23:7163–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Arase M, Horiguchi K, Ehata S, Morikawa M, Tsutsumi S, Aburatani H, Miyazono K, Koinuma D. Transforming growth factor-β-induced lncRNA-Smad7 inhibits apoptosis of mouse breast cancer JygMC(A) cells. Cancer Sci. 2014;105:974–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Lo PK, Zhang Y, Wolfson B, Gernapudi R, Yao Y, Duru N, Zhou Q. Dysregulation of the BRCA1/long non-coding RNA NEAT1 signaling axis contributes to breast tumorigenesis. Oncotarget. 2016;7:65067–89.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Jiang M, Xiao Y, Liu D, Luo N, Gao Q, Guan Y. Overexpression of long noncoding RNA LINC01296 indicates an unfavorable prognosis and promotes tumorigenesis in breast cancer. Gene. 2018;675:217–24.

    Article  CAS  PubMed  Google Scholar 

  110. Ma D, Chen C, Wu J, Wang H, Wu D. Up-regulated lncRNA AFAP1-AS1 indicates a poor prognosis and promotes carcinogenesis of breast cancer. Breast Cancer. 2019;26:74–83.

    Article  PubMed  Google Scholar 

  111. Song R, Zhang J, Huang J, Hai T. Long non-coding RNA GHET1 promotes human breast cancer cell proliferation, invasion and migration via affecting epithelial mesenchymal transition. Cancer Biomark. 2018;22:565–73.

    Article  CAS  PubMed  Google Scholar 

  112. Jiang J, Shi SH, Li XJ, Sun L, Ge QD, Li C, Zhang W. Long non-coding RNA BRAF-regulated lncRNA 1 promotes lymph node invasion, metastasis and proliferation, and predicts poor prognosis in breast cancer. Oncol Lett. 2018;15:9543–52.

    PubMed  PubMed Central  Google Scholar 

  113. Wang O, Yang F, Liu Y, Lv L, Ma R, Chen C, Wang J, Tan Q, Cheng Y, Xia E, Chen Y, Zhang X. C-MYC-induced upregulation of lncRNA SNHG12 regulates cell proliferation, apoptosis and migration in triple-negative breast cancer. Am J Transl Res. 2017;9:533–45.

    PubMed  PubMed Central  Google Scholar 

  114. Li X, Wu Y, Liu A, Tang X. Long non-coding RNA UCA1 enhances tamoxifen resistance in breast cancer cells through a miR-18a-HIF1α feedback regulatory loop. Tumour Biol. 2016;37:14733–43.

    Article  CAS  PubMed  Google Scholar 

  115. Li Y, Wang B, Lai H, Li S, You Q, Fang Y, Li Q, Liu Y. Long non-coding RNA CRALA is associated with poor response to chemotherapy in primary breast cancer. Thorac Cancer. 2017;8:582–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Shi SJ, Wang LJ, Yu B, Li YH, Jin Y, Bai XZ. LncRNA-ATB promotes trastuzumab resistance and invasion-metastasis cascade in breast cancer. Oncotarget. 2015;6:11652–63.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Chang L, Hu Z, Zhou Z, Zhang H. Linc00518 contributes to multidrug resistance through regulating the MiR-199a/MRP1 axis in breast cancer. Cell Physiol Biochem. 2018;48:16–28.

    Article  CAS  PubMed  Google Scholar 

  118. Ma Y, Bu D, Long J, Chai W, Dong J. LncRNA DSCAM-AS1 acts as a sponge of miR-137 to enhance Tamoxifen resistance in breast cancer. J Cell Physiol. 2019;234:2880–94.

    Article  CAS  PubMed  Google Scholar 

  119. Xu X, Ji S, Li W, Yi B, Li H, Zhang H, Ma W. LncRNA H19 promotes the differentiation of bovine skeletal muscle satellite cells by suppressing Sirt1/FoxO1. Cell Mol Biol Lett. 2017;22:10.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Wang J, Xie S, Yang J, Xiong H, Jia Y, Zhou Y, Chen Y, Ying X, Chen C, Ye C, Wang L, Zhou J. The long noncoding RNA H19 promotes tamoxifen resistance in breast cancer via autophagy. J Hematol Oncol. 2019;12:81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Chen J, Qin C, Zhou Y, Chen Y, Mao M, Yang J. Metformin may induce ferroptosis by inhibiting autophagy via lncRNA H19 in breast cancer. FEBS Open Bio. 2022;12:146–53.

    Article  PubMed  Google Scholar 

  122. Huang J, Zhou N, Watabe K, Lu Z, Wu F, Xu M, Mo YY. Long non-coding RNA UCA1 promotes breast tumor growth by suppression of p27 (Kip1). Cell Death Dis. 2014;5: e1008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Raju GSR, Pavitra E, Bandaru SS, Varaprasad GL, Nagaraju GP, Malla RR, Huh YS, Han YK. HOTAIR: a potential metastatic, drug-resistant and prognostic regulator of breast cancer. Mol Cancer. 2023;22:65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Müller V, Oliveira-Ferrer L, Steinbach B, Pantel K, Schwarzenbach H. Interplay of lncRNA H19/miR-675 and lncRNA NEAT1/miR-204 in breast cancer. Mol Oncol. 2019;13:1137–49.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Wu D, Zhu J, Fu Y, Li C, Wu B. LncRNA HOTAIR promotes breast cancer progression through regulating the miR-129-5p/FZD7 axis. Cancer Biomark. 2021;30:203–12.

    Article  CAS  PubMed  Google Scholar 

  126. Wang Y, Gong G, Xu J, Zhang Y, Wu S, Wang S. Long noncoding RNA HOTAIR promotes breast cancer development by targeting ZEB1 via sponging miR-601. Cancer Cell Int. 2020;20:320.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, Wang Y, Brzoska P, Kong B, Li R, West RB, van de Vijver MJ, Sukumar S, Chang HY. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464:1071–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Peng WX, Koirala P, Mo YY. LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 2017;36:5661–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Ding W, Ren J, Ren H, Wang D. Long noncoding RNA HOTAIR modulates MiR-206-mediated Bcl-w signaling to facilitate cell proliferation in breast cancer. Sci Rep. 2017;7:17261.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Liu LC, Wang YL, Lin PL, Zhang X, Cheng WC, Liu SH, Chen CJ, Hung Y, Jan CI, Chang LC, Qi X, Hsieh-Wilson LC, Wang SC. Long noncoding RNA HOTAIR promotes invasion of breast cancer cells through chondroitin sulfotransferase CHST15. Int J Cancer. 2019;145:2478–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Zhao W, Geng D, Li S, Chen Z, Sun M. LncRNA HOTAIR influences cell growth, migration, invasion, and apoptosis via the miR-20a-5p/HMGA2 axis in breast cancer. Cancer Med. 2018;7:842–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Zhao C, Ling X, Xia Y, Yan B, Guan Q. LncRNA UCA1 promotes SOX12 expression in breast cancer by regulating m(6)A modification of miR-375 by METTL14 through DNA methylation. Cancer Gene Ther. 2022;29:1043–55.

    Article  CAS  PubMed  Google Scholar 

  133. Giro-Perafita A, Luo L, Khodadadi-Jamayran A, Thompson M, Akgol Oksuz B, Tsirigos A, Dynlacht BD, Sánchez I, Esteva FJ. LncRNA RP11–19E11 is an E2F1 target required for proliferation and survival of basal breast cancer. NPJ Breast Cancer. 2020;6:1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Zhang N, Zeng X, Sun C, Guo H, Wang T, Wei L, Zhang Y, Zhao J, Ma X. LncRNA LINC00963 promotes tumorigenesis and radioresistance in breast cancer by sponging miR-324-3p and inducing ACK1 expression. Mol Ther Nucleic Acids. 2019;18:871–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Mondal P, Meeran SM. Long non-coding RNAs in breast cancer metastasis. Noncoding RNA Res. 2020;5:208–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Li Y, Song Y, Wang Z, Zhang Z, Lu M, Wang Y. Long non-coding RNA LINC01787 drives breast cancer progression via disrupting miR-125b generation. Front Oncol. 2019;9:1140.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, Lin L, Yao H, Su F, Li D, Zeng M, Song E. A cytoplasmic NF-κB interacting long noncoding RNA blocks IκB phosphorylation and suppresses breast cancer metastasis. Cancer Cell. 2015;27:370–81.

    Article  CAS  PubMed  Google Scholar 

  138. Ma Y, Yu L, Yan W, Qiu L, Zhang J, Jia X. lncRNA GAS5 sensitizes breast cancer cells to ionizing radiation by inhibiting DNA repair. Biomed Res Int. 2022;2022:1987519.

    PubMed  PubMed Central  Google Scholar 

  139. Shi P, Zhang J, Li X, Li W, Li H, Fu P. Long non-coding RNA NORAD inhibition upregulates microRNA-323a-3p to suppress tumorigenesis and development of breast cancer through the PUM1/eIF2 axis. Cell Cycle. 2021;20:1295–307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Zhou K, Ou Q, Wang G, Zhang W, Hao Y, Li W. High long non-coding RNA NORAD expression predicts poor prognosis and promotes breast cancer progression by regulating TGF-β pathway. Cancer Cell Int. 2019;19:63.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Tan BS, Yang MC, Singh S, Chou YC, Chen HY, Wang MY, Wang YC, Chen RH. LncRNA NORAD is repressed by the YAP pathway and suppresses lung and breast cancer metastasis by sequestering S100P. Oncogene. 2019;38:5612–26.

    Article  CAS  PubMed  Google Scholar 

  142. Liang Y, Song X, Li Y, Chen B, Zhao W, Wang L, Zhang H, Liu Y, Han D, Zhang N, Ma T, Wang Y, Ye F, Luo D, Li X, Yang Q. LncRNA BCRT1 promotes breast cancer progression by targeting miR-1303/PTBP3 axis. Mol Cancer. 2020;19:85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Hu J, Huang H, Xi Z, Ma S, Ming J, Dong F, Guo H, Zhang H, Zhao E, Yao G, Yang L, Zhang F, Zheng W, Chen H, Huang T, Li L. LncRNA SEMA3B-AS1 inhibits breast cancer progression by targeting miR-3940/KLLN axis. Cell Death Dis. 2022;13:800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Zhang Q, Li T, Wang Z, Kuang X, Shao N, Lin Y. lncRNA NR2F1-AS1 promotes breast cancer angiogenesis through activating IGF-1/IGF-1R/ERK pathway. J Cell Mol Med. 2020;24:8236–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhang Y, Dong X, Guo X, Li C, Fan Y, Liu P, Yuan D, Ma X, Wang J, Zheng J, Li H, Gao P. LncRNA-BC069792 suppresses tumor progression by targeting KCNQ4 in breast cancer. Mol Cancer. 2023;22:41.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Zong S, Dai W, Guo X, Wang K. LncRNA-SNHG1 promotes macrophage M2-like polarization and contributes to breast cancer growth and metastasis. Aging. 2021;13:23169–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Ouyang J, Liu Z, Yuan X, Long C, Chen X, Wang Y, Liu L, Liu S, Liang H. LncRNA PRNCR1 promotes breast cancer proliferation and inhibits apoptosis by modulating microRNA-377/CCND2/MEK/MAPK axis. Arch Med Res. 2021;52:471–82.

    Article  CAS  PubMed  Google Scholar 

  148. Li X, Deng S, Pang X, Song Y, Luo S, Jin L, Pan Y. LncRNA NEAT1 silenced miR-133b promotes migration and invasion of breast cancer cells. Int J Mol Sci. 2019;20:3616.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Song X, Liu Z, Yu Z. LncRNA NEF is downregulated in triple negative breast cancer and correlated with poor prognosis. Acta Biochim Biophys Sin (Shanghai). 2019;51:386–92.

    Article  CAS  PubMed  Google Scholar 

  150. Li Q, Mo W, Ding Y, Ding X. Study of lncRNA TPA in promoting invasion and metastasis of breast cancer mediated by TGF-β signaling pathway. Front Cell Dev Biol. 2021;9: 688751.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Fang Z, Wang Y, Wang Z, Xu M, Ren S, Yang D, Hong M, Xie W. ERINA is an estrogen-responsive LncRNA that drives breast cancer through the E2F1/RB1 pathway. Can Res. 2020;80:4399–413.

    Article  CAS  Google Scholar 

  152. Gong X, Dong T, Niu M, Liang X, Sun S, Zhang Y, Li Y, Li D. lncRNA LCPAT1 upregulation promotes breast cancer progression via enhancing MFAP2 transcription. Mol Ther Nucleic Acids. 2020;21:804–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Li Q, Gao H, Zhou S, Liao Y. LncRNA PlncRNA-1 overexpression inhibits the growth of breast cancer by upregulating TGF-β1 and downregulating PHGDH. Breast Cancer. 2018;25:619–25.

    Article  PubMed  Google Scholar 

  154. Liu M, Gou L, Xia J, Wan Q, Jiang Y, Sun S, Tang M, He T, Zhang Y. LncRNA ITGB2-AS1 could promote the migration and invasion of breast cancer cells through up-regulating ITGB2. Int J Mol Sci. 2018;19:1866.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Jia X, Shi L, Wang X, Luo L, Ling L, Yin J, Song Y, Zhang Z, Qiu N, Liu H, Deng M, He Z, Li H, Zheng G. KLF5 regulated lncRNA RP1 promotes the growth and metastasis of breast cancer via repressing p27kip1 translation. Cell Death Dis. 2019;10:373.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, Liu Y, Zhang N, Yang Q. LncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215. J Cell Physiol. 2019;234:9105–17.

    Article  CAS  PubMed  Google Scholar 

  157. Shi C, Ren S, Zhao X, Li Q. lncRNA MALAT1 regulates the resistance of breast cancer cells to paclitaxel via the miR-497-5p/SHOC2 axis. Pharmacogenomics. 2022;23:973–85.

    Article  CAS  PubMed  Google Scholar 

  158. Kim J, Piao HL, Kim BJ, Yao F, Han Z, Wang Y, Xiao Z, Siverly AN, Lawhon SE, Ton BN, Lee H, Zhou Z, Gan B, Nakagawa S, Ellis MJ, Liang H, Hung MC, You MJ, Sun Y, Ma L. Long noncoding RNA MALAT1 suppresses breast cancer metastasis. Nat Genet. 2018;50:1705–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Xu Y, Ren W, Li Q, Duan C, Lin X, Bi Z, You K, Hu Q, Xie N, Yu Y, Xu X, Hu H, Yao H. LncRNA Uc003xsl.1-mediated activation of the NFκB/IL8 axis promotes progression of triple-negative breast cancer. Cancer Res. 2022;82:556–70.

    Article  CAS  PubMed  Google Scholar 

  160. Sheng X, Dai H, Du Y, Peng J, Sha R, Yang F, Zhou L, Lin Y, Xu S, Wu Y, Yin W, Lu J. LncRNA CARMN overexpression promotes prognosis and chemosensitivity of triple negative breast cancer via acting as miR143-3p host gene and inhibiting DNA replication. J Exp Clin Cancer Res. 2021;40:205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Liu P, Feng Y, Li H, Chen X, Wang G, Xu S, Li Y, Zhao L. Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis. Cell Mol Biol Lett. 2020;25:10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Zhang Z, Lu YX, Liu F, Sang L, Shi C, Xie S, Bian W, Yang JC, Yang Z, Qu L, Chen SY, Li J, Yang L, Yan Q, Wang W, Fu P, Shao J, Li X, Lin A. lncRNA BREA2 promotes metastasis by disrupting the WWP2-mediated ubiquitination of Notch1. Proc Natl Acad Sci USA. 2023;120: e2206694120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Chen X, Luo R, Zhang Y, Ye S, Zeng X, Liu J, Huang D, Liu Y, Liu Q, Luo ML, Song E. Long noncoding RNA DIO3OS induces glycolytic-dominant metabolic reprogramming to promote aromatase inhibitor resistance in breast cancer. Nat Commun. 2022;13:7160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Zhu P, He F, Hou Y, Tu G, Li Q, Jin T, Zeng H, Qin Y, Wan X, Qiao Y, Qiu Y, Teng Y, Liu M. A novel hypoxic long noncoding RNA KB-1980E6.3 maintains breast cancer stem cell stemness via interacting with IGF2BP1 to facilitate c-Myc mRNA stability. Oncogene. 2021;40:1609–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Gooding AJ, Zhang B, Jahanbani FK, Gilmore HL, Chang JC, Valadkhan S, Schiemann WP. The lncRNA BORG drives breast cancer metastasis and disease recurrence. Sci Rep. 2017;7:12698.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Yang F, Lv S. LncRNA EPB41L4A-AS1 regulates cell proliferation, apoptosis and metastasis in breast cancer. Ann Clin Lab Sci. 2022;52:3–11.

    CAS  PubMed  Google Scholar 

  167. Zhang X, Zhao X, Chang L, Liu F, Li C, Ge P. LncRNA FOXD3-AS1 promotes breast cancer progression by mediating ARF6. Breast Cancer. 2022;29:908–20.

    Article  PubMed  Google Scholar 

  168. Zhai D, Li T, Ye R, Bi J, Kuang X, Shi Y, Shao N, Lin Y. LncRNA LGALS8-AS1 promotes breast cancer metastasis through miR-125b-5p/SOX12 feedback regulatory network. Front Oncol. 2021;11: 711684.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Shen P, Yu Y, Yan Y, Yu B, You W. LncRNA CASC15 regulates breast cancer cell stemness via the miR-654-5p/MEF2D axis. J Biochem Mol Toxicol. 2022;36: e23023.

    Article  CAS  PubMed  Google Scholar 

  170. Han M, Wang Y, Gu Y, Ge X, Seng J, Guo G, Zhang X, Zhao Y, Dou D. lncRNA GHET1 knockdown suppresses breast cancer activity in vitro and in vivo. Am J Transl Res. 2019;11:31–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Lei C, Li S, Fan Y, Hua L, Pan Q, Li Y, Long Z, Yang R. LncRNA DUXAP8 induces breast cancer radioresistance by modulating the PI3K/AKT/mTOR pathway and the EZH2-E-cadherin/RHOB pathway. Cancer Biol Ther. 2022;23:1–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Lv X, Zhang Q. LncRNA RP11–214F16.8 drives breast cancer tumorigenesis via a post-translational repression on NISCH expression. Cell Signal. 2022;92:110271.

    Article  CAS  PubMed  Google Scholar 

  173. Xu J, Hu M, Gao Y, Wang Y, Yuan X, Yang Y, Song W, Yin W, Gong P, Wei L, Zhang J. LncRNA MIR17HG suppresses breast cancer proliferation and migration as ceRNA to target FAM135A by sponging miR-454–3p. Mol Biotechnol. 2023. https://doi.org/10.1007/s12033-023-00706-1.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Qiu S, Chen G, Peng J, Liu J, Chen J, Wang J, Li L, Yang K. LncRNA EGOT decreases breast cancer cell viability and migration via inactivation of the Hedgehog pathway. FEBS Open Bio. 2020;10:817–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Li K, Ma YB, Tian YH, Xu XL, Gao Y, He YQ, Pan WT, Zhang JW, He CJ, Wei L. Silencing lncRNA SNHG6 suppresses proliferation and invasion of breast cancer cells through miR-26a/VASP axis. Pathol Res Pract. 2019;215: 152575.

    Article  CAS  PubMed  Google Scholar 

  176. Xu X, Xie Q, Xie M, Zeng Y, Liu Q. LncRNA SNHG8 serves as an oncogene in breast cancer through miR-634/ZBTB20 axis. Cancer Manag Res. 2021;13:3017–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Qu H, Li X, Chen F, Zhang M, Lu X, Gu Y, Lv M, Lu C. LncRNA PVT1 influences breast cancer cells glycolysis through sponging miR-145-5p, Genes. Genomics. 2023;45:581–92.

    CAS  Google Scholar 

  178. Lu Q, Wang L, Gao Y, Zhu P, Li L, Wang X, Jin Y, Zhi X, Yu J, Li X, Qin X, Zhou P. lncRNA APOC1P1-3 promoting anoikis-resistance of breast cancer cells. Cancer Cell Int. 2021;21:232.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Fang K, Xu ZJ, Jiang SX, Tang DS, Yan CS, Deng YY, Zhao FY. lncRNA FGD5-AS1 promotes breast cancer progression by regulating the hsa-miR-195-5p/NUAK2 axis. Mol Med Rep. 2021;23:1.

    Article  PubMed  Google Scholar 

  180. Ding Z, Ye P, Yang X, Cai H. LncRNA FBXL19-AS1 promotes breast cancer cells proliferation and invasion via acting as a molecular sponge to miR-718. Biosci Rep. 2019;39:BSR20182018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Wang N, Hou M, Zhan Y, Sheng X. LncRNA PTCSC3 inhibits triple-negative breast cancer cell proliferation by downregulating lncRNA H19. J Cell Biochem. 2019;120:15083–8.

    Article  CAS  PubMed  Google Scholar 

  182. Fearnhead HO, Vandenabeele P, Vanden Berghe T. How do we fit ferroptosis in the family of regulated cell death. Cell Death Differ. 2017;24:1991–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Wang Z, Chen X, Liu N, Shi Y, Liu Y, Ouyang L, Tam S, Xiao D, Liu S, Wen F, Tao Y. A nuclear long non-coding RNA LINC00618 accelerates ferroptosis in a manner dependent upon apoptosis. Mol Ther. 2021;29:263–74.

    Article  CAS  PubMed  Google Scholar 

  184. Qi W, Li Z, Xia L, Dai J, Zhang Q, Wu C, Xu S. LncRNA GABPB1-AS1 and GABPB1 regulate oxidative stress during erastin-induced ferroptosis in HepG2 hepatocellular carcinoma cells. Sci Rep. 2019;9:16185.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Wu X, Liu C, Li Z, Gai C, Ding D, Chen W, Hao F, Li W. Regulation of GSK3β/Nrf2 signaling pathway modulated erastin-induced ferroptosis in breast cancer. Mol Cell Biochem. 2020;473:217–28.

    Article  CAS  PubMed  Google Scholar 

  186. Brown CW, Amante JJ, Chhoy P, Elaimy AL, Liu H, Zhu LJ, Baer CE, Dixon SJ, Mercurio AM. Prominin2 drives ferroptosis resistance by stimulating iron export. Dev Cell. 2019;51:575-586.e574.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Zhou N, Bao J. FerrDb: a manually curated resource for regulators and markers of ferroptosis and ferroptosis-disease associations. Database (Oxford). 2020;2020:baaa021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Zhang K, Ping L, Du T, Liang G, Huang Y, Li Z, Deng R, Tang J. A ferroptosis-related lncRNAs signature predicts prognosis and immune microenvironment for breast cancer. Front Mol Biosci. 2021;8: 678877.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Wang L, Luan T, Zhou S, Lin J, Yang Y, Liu W, Tong X, Jiang W. LncRNA HCP5 promotes triple negative breast cancer progression as a ceRNA to regulate BIRC3 by sponging miR-219a-5p. Cancer Med. 2019;8:4389–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Jia CL, Yang F, Li R. Prognostic model construction and immune microenvironment analysis of breast cancer based on ferroptosis-related lncRNAs. Int J Gen Med. 2021;14:9817–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Tong X, Yu Z, Xing J, Liu H, Zhou S, Huang Y, Lin J, Jiang W, Wang L. LncRNA HCP5-encoded protein regulates ferroptosis to promote the progression of triple-negative breast cancer. Cancers. 2023;15:1880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Wang S, Wang Y, Li Q, Zeng K, Li X, Feng X. RUNX1-IT1 favors breast cancer carcinogenesis through regulation of IGF2BP1/GPX4 axis. Discov Oncol. 2023;14:42.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Zhong H, Zeng G, He L. Overexpression of the lncRNA AC012213.3 promotes proliferation, migration and invasion of breast cancer via RAD54B/PI3K/AKT axis and is associated with worse patient prognosis. Cancer Manag Res. 2021;13:7213–23.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Dixon SJ, Stockwell BR. The hallmarks of ferroptosis. Ann Rev Cancer Biol. 2019;3:35–54.

    Article  Google Scholar 

  195. Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, Liao P, Lang X, Kryczek I, Sell A, Xia H, Zhou J, Li G, Li J, Li W, Wei S, Vatan L, Zhang H, Szeliga W, Gu W, Liu R, Lawrence TS, Lamb C, Tanno Y, Cieslik M, Stone E, Georgiou G, Chan TA, Chinnaiyan A, Zou W. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569:270–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Lu B, Chen XB, Ying MD, He QJ, Cao J, Yang B. The role of ferroptosis in cancer development and treatment response. Front Pharmacol. 2017;8:992.

    Article  PubMed  Google Scholar 

  197. Stěrba M, Popelová O, Vávrová A, Jirkovský E, Kovaříková P, Geršl V, Simůnek T. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxid Redox Signal. 2013;18:899–929.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Roy SS, Chakraborty P, Bhattacharya S. Intervention in cyclophosphamide induced oxidative stress and DNA damage by a flavonyl-thiazolidinedione based organoselenocyanate and evaluation of its efficacy during adjuvant therapy in tumor bearing mice. Eur J Med Chem. 2014;73:195–209.

    Article  CAS  PubMed  Google Scholar 

  199. Jiang H, Zhang XW, Liao QL, Wu WT, Liu YL, Huang WH. Electrochemical monitoring of paclitaxel-induced ROS release from mitochondria inside single cells. Small. 2019;15: e1901787.

    Article  PubMed  Google Scholar 

  200. Vernier M, Dufour CR, McGuirk S, Scholtes C, Li X, Bourmeau G, Kuasne H, Park M, St-Pierre J, Audet-Walsh E, Giguère V. Estrogen-related receptors are targetable ROS sensors. Genes Dev. 2020;34:544–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by grants from Postgraduate Scientific Research Innovation Project of Hunan Province (CX20230955), the National Natural Science Foundation of China (no. 82103171, Jianbo Feng) and the Natural Science Foundation of Hunan Province (no. 2022JJ70121, Xing Ren) and the Clinical Medical Research “4310” Program of the University of South China (20224310NHYCG07).

Author information

Authors and Affiliations

Authors

Contributions

SX and WY collected relevant literature and received and drafted the manuscript. XR summarized the figures and tables. XZ and JF reviewed the manuscript and revised it. All authors contributed to the manuscript and approved its submission.

Corresponding authors

Correspondence to Jianbo Feng or Xuyu Zu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiang, S., Yan, W., Ren, X. et al. Role of ferroptosis and ferroptosis-related long non'coding RNA in breast cancer. Cell Mol Biol Lett 29, 40 (2024). https://doi.org/10.1186/s11658-024-00560-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s11658-024-00560-2

Keywords