Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
June-2025 Volume 55 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
June-2025 Volume 55 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review)

  • Authors:
    • Jiaqi Yang
    • Yu Wang
    • Fangyuan Liu
    • Yizhong Zhang
    • Fengjuan Han
  • View Affiliations / Copyright

    Affiliations: Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China, Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
    Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 97
    |
    Published online on: April 28, 2025
       https://doi.org/10.3892/ijmm.2025.5538
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Ferroptosis is a unique mode of cell death driven by iron‑dependent phospholipid peroxidation, and its mechanism primarily involves disturbances in iron metabolism, imbalances in the lipid antioxidant system and accumulation of lipid peroxides. Protein processing, modification and folding in the endoplasmic reticulum (ER) are closely related regulatory processes that determine cell function, fate and survival. The uncontrolled proliferative capacity of malignant cells generates an unfavorable microenvironment characterized by high metabolic demand, hypoxia, nutrient deprivation and acidosis, which promotes the accumulation of misfolded or unfolded proteins in the ER, leading to ER stress (ERS). Ferroptosis and ERS share common pathways in several diseases, and the two interact to affect cell survival and death. Additionally, cell death pathways are not linear signaling cascades, and different pathways of cell death may be interrelated at multiple levels. Ferroptosis and ERS in ovarian cancer (OC) have attracted increasing research interest; however, both are discussed separately regarding OC. The present review aims to summarize the associations and potential links between ferroptosis and ERS, aiming to provide research references for the development of therapeutic approaches for the management of OC.

Introduction

Ovarian cancer (OC) ranks third among all gynecologic malignancies, is one of the three most diagnosed malignancies in the female reproductive system and has the highest mortality rate among gynecologic tumors (1). Due to the insidious nature of its onset during the early stages, the majority of patients are diagnosed with advanced-stage OC in the first instance, and at this stage, the primary treatment methods, such as surgery, radiotherapy, chemotherapy and immunotherapy, have well-established side effects, including recurrence, metastasis and chemotherapy resistance, and thus, OC is characterized by a high mortality rate (2). As the incidence of OC is increasing annually, with an increasing number of younger individuals developing OC (3,4), the need for methods and biomarkers to allow earlier detection and more effective treatment options after diagnosis is becoming ever more pertinent (5-8).

Ferroptosis, distinct from apoptosis, necrosis and autophagy, is an iron-dependent form of programmed cell death triggered by lipid peroxide (LPO) accumulation (9). While conventional cancer therapies usually eliminate tumor cells by inducing cell death to generate drug resistance, ferroptosis has become an increasingly researched topic in cancer therapy given its role in regulating cell death (10-12). Several studies have shown that ferroptosis is associated with resistance to cancer treatment, potentially allowing for the reversal of cancer treatment resistance (13-15). Current research has shown that neurological disorders, kidney damage, breast cancer, pancreatic cancer and several other diseases are associated with ferroptosis (16-19). Among these, ferroptosis is most closely related to malignant tumors, and tumor cells may be notably sensitive to ferroptosis (20). Ferroptosis can regulate the development of OC through different mechanisms or factors, thereby increasing the sensitivity of OC cells to ferroptosis-targeting therapeutics, and thus, combatting resistance to chemotherapy (21,22) and improving the efficacy of chemotherapeutic drugs in the management of OC (23). Additionally, a related study has demonstrated the pattern of immune infiltration and associated genetic features of ferroptosis, which can hopefully be applied to predict the prognosis of patients with OC (24). The combination of ferroptosis with chemotherapy, nanotechnology, X-ray therapy and photodynamic therapy has been demonstrated to improve therapeutic efficacy (25), providing potential targets and novel therapeutic directions for ferroptosis in the treatment of OC.

The endoplasmic reticulum (ER) is involved in lipid metabolism, Ca2+ regulation, and protein processing, folding and transport, and is an important organelle in eukaryotic cells (26). ER stress (ERS) is induced in cells by hypoxic conditions, genetic mutations, nutrient deficiencies and oxidative stress, which leads to the accumulation of misfolded and unfolded proteins in the lumen of the ER and the activation of the unfolded protein response (UPR) to counteract the unfavorable external environments (27). However, sustained high-intensity ERS, which results in cell death when the tolerance threshold of the UPR is exceeded (28), can lead to a range of diseases such as cancer, atherosclerosis, diabetic retinopathy and ischemic nephropathy (29,30). The adverse microenvironment in tumor cells due to high metabolic demands and oxidative stress, among others, interferes with ER homeostasis in immune cells, which can have an impact on protective anticancer immunity (31). A study has shown that targeting the relevant pathways in ERS can inhibit the proliferation of OC cells and reduce their resistance to chemotherapy (32). Thus, ERS serves a key role in the development of OC, as well as in its treatment.

Several studies have found that ferroptosis and ERS share a common regulatory pathway and that the two can alter the development of several diseases by interacting with each other (33-36). However, to the best of our knowledge, the link between them has not been investigated in detail in studies on the pathogenesis of OC or related therapies. Therefore, the present review discusses the mechanisms of ferroptosis and ERS in OC and the potential relevance of both to highlight novel strategies and potential targets for the treatment of OC.

Ferroptosis in OC

Ferroptosis is a mode of programmed cell death characterized by excessive accumulation of iron, lipid antioxidation and lipid peroxidation (37). Patients with OC frequently exhibit resistance to chemotherapy, and a study has shown that this is closely related to ferroptosis (38).

Iron metabolism in OC

Iron is one of the essential trace elements in the human body and serves an important role in human growth and development, energy metabolism, and immune system functions (39). Abnormalities in iron metabolism affect redox reactions, gene regulation, enzymatic reactions and DNA synthesis or repair (40). In the human body, iron has a complex and elaborate circulatory mechanism to ensure its proper distribution, utilization and storage to maintain appropriate and non-toxic cellular iron levels (41). Iron in the body exists in two forms, Fe2+ and Fe3+, and there are transport carriers and modes for the different forms of iron. Daily intake of dietary iron includes heme iron and non-heme iron (42). They arrive in the intestinal lumen as Fe3+ and are reduced to Fe2+ by duodenal cytochrome B to be absorbed; non-heme iron in the intestine is absorbed via divalent metal transporter protein 1 (43). Heme iron is then transported into the duodenal epithelium via heme carrier protein 1 and is absorbed, internalized, and broken down into Fe2+ and heme oxygenase-1 (HO-1) (44). Thereafter, iron can remain in the enterocytes or enter the bloodstream from the basolateral membrane of intestinal epithelial cells via membrane iron transport protein 1, while being oxidized by ferrous oxidase or ceruloplasmin to produce Fe3+ (45). Upon entering the bloodstream, plasma transferrin (TF) spreads the captured Fe3+ throughout the cells of the body and is used by various organs to synthesize specific iron-containing components through TF receptor (TFR)-mediated holo-TF endocytosis (46). For example, the liver synthesizes hemosiderin, muscle tissue produces myoglobin and the bone marrow produces red blood cells containing hemoglobin. Cells promote iron uptake primarily through the TF and TFR systems, and Fe3+ is reduced to Fe2+ by ferric oxide reductase, most of which binds to ferritin to form storage iron, and a small portion enters the cytoplasm to collectively form the labile iron pool (47). Due to the instability and high oxidizability of Fe2+, excess iron ions catalyze the production of reactive oxygen species (ROS), which promotes lipid peroxidation through the Fenton reaction (48), thereby causing oxidative damage to lipid membranes, proteins and DNA, and ultimately leading to cell death (49). Among the three main mechanisms of ferroptosis, excessive accumulation of Fe2+ in the labile iron pool increases the sensitivity of cells to ferroptosis and is the initial component responsible for ferroptosis (47).

To the best of our knowledge, the initial signal for ferroptosis has not yet been identified, but ferroptosis is closely related to the levels of intracellular free iron content (50). Iron metabolism serves a key role in the pathophysiology of OC, and the degree of intracellular iron accumulation serves a decisive role in the course of OC (51). Concurrently, abnormalities in iron metabolism, especially the acquisition and retention of excess iron, contribute to tumorigenesis and tumor growth (52,53). Iron accumulation increases the risk of diseases such as cancer and damage to tissues (54). Therefore, maintaining homeostasis of intracellular iron ions is critical. According to Basuli et al (55), OC-initiating cells show high iron dependence. Increased iron efflux also decreases the proliferation and invasion of OC-initiating cells, and conversely increased iron uptake increases OC cell proliferation and invasion. High-grade serous OC (HGSOC) typically originates from the fallopian tube and is diagnosed at a later stage, and it has been observed that the iron content of HGSOC is higher than that of normal ovarian tissues and low-grade serous OC, suggesting that there is a strong association between HGSOC and iron metabolism (56). In addition, the malignant transformation and metastasis of cancer cells are closely associated with changes in cellular redox status (57). The molecular damage caused by the excessive and harmful ROS catalyzed by free iron is often referred to as 'oxidative damage', and Bauckman et al (58) reported that ROS can transform normal ovarian tissue into cancerous tissue by promoting the MAPK pathway. In addition, ROS can hydroxylate DNA residues to generate the highly mutagenic 8-hydroxy-2′-deoxyguanosine, elevated levels of which have been found to be associated with a poor prognosis in patients with HGSOC (59,60). Iron polyporphyrin heme binds to p53, interfering with p53-DNA interactions, leading to nuclear export and cellular degradation of p53, and increased susceptibility to HGSOC (61,62). Basuli et al (55) demonstrated that excess iron simultaneously affects tumor cell proliferation, metabolism and metastasis. Increasing the expression of ferroportin on cell membranes (63), decreasing iron intake (64), or decreasing the concentration of TF (65) and TFR in vivo (66) can inhibit tumor growth. In addition, iron metabolism can contribute to the development of OC by regulating hypoxia-inducible factor (HIF). HIF-1α induces OC progression by inhibiting p53 function, promoting IL-6 expression or being regulated by long non-coding RNAs (67,68). Iron metabolism is also closely related to resistance to chemotherapy, and solute carrier family 40 member 1 (SLC40A1), an iron metabolism-related gene, which is the only known iron-exporting gene (69), serves a crucial role in transporting iron from the intracellular environment to the extracellular environment, and thus, the physiological expression of SLC40A serves a vital role in the regulation of iron homeostasis. SLC40A1-induced iron overload leads to cisplatin resistance in OC (70). Upregulation of SLC40A1 reduces cisplatin resistance by exporting iron, decreasing the intracellular iron concentration and oxidative stress. By contrast, an increase in the intracellular iron concentration and enhanced oxidative stress induced by the downregulation of SLC40A1 increase cisplatin resistance (70). Thus, modulating iron levels to influence the redox system may be a potential strategy to reverse drug resistance in OC therapy.

The iron-dependent nature of OC tumor-initiating cells also makes them sensitive to drugs that induce ferroptosis, as well as to iron chelators, providing a potential therapeutic approach for the management of OC (71). Desferrioxamine (72), a natural iron chelator used in the treatment of iron overload, has shown promising results in the treatment of OC. Wang et al (72) assessed the effects of desferrioxamine on OC cell lines and found that it not only inhibited OC stem cells but also enhanced cisplatin efficacy, improved resistance to chemotherapy and increased the length of survival. There are also drugs that regulate iron metabolism in several ways and may have effects on other biological processes. For example, artemisinin is known for its anti-malarial, anti-inflammatory and antitumor effects, and its compounds (artesunate) can reduce cell proliferation and induce ROS production in OC cells (73). In addition, artesunate can activate the function of lysosomes, promote the degradation of ferritin and lead to the release of iron in lysosomes, and thereby mediate cell death (74). Regulators, including iron uptake-related regulators (75), iron storage-related regulators (76) and iron transport-related regulators (77), influence the pathological process of OC through the regulation of iron metabolism levels.

Lipid peroxidation in OC

Ferroptosis is an iron-dependent non-apoptotic cell death mechanism caused by iron-dependent membrane lipid peroxidation and massive accumulation of cellular ROS (78). Membrane lipids serve an important role in the regulation of cell fate and lipid metabolism, which is fundamental in determining the fate of ferroptosis (79), and is similarly critical for ferroptosis execution.

Among various lipids, polyunsaturated fatty acids (PUFAs) associated with several phospholipids such as phosphatidylethanolamine and phosphatidylcholine are responsible for inducing lipid peroxidation in ferroptosis. Phospholipids containing PUFAs are susceptible to oxidation, but less oxidizable saturated/monounsaturated fatty acids protect cells from ferroptosis (80). Thus, the enzymes and pathways that regulate the metabolism of PUFAs and monounsaturated fatty acids, as well as the balance of PUFAs and monounsaturated fatty acids in membrane phospholipids, can influence cellular sensitivity to ferroptosis (81). The findings of the aforementioned study provide a novel strategy for lipid peroxidation treatment of OC ferroptosis. Since the membrane phospholipids of PUFAs that drive ROS production catalyzed by iron ions interacting with PUFAs are responsible for triggering lipid peroxidation leading to cellular ferroptosis, and not free PUFA itself, the enzymes that bind free PUFA to phospholipids serve a crucial role in ferroptosis (81). Acyl-CoA synthetase long-chain family member 4 is an essential component of ferroptosis execution as shown by microarray analysis of ferroptosis-resistant cell lines and using a genome-wide CRISPR-based genetic screening system (82). Under the catalytic action of Acyl-CoA synthetase long-chain family member 4, free long-chain fatty acids are converted to acyl-CoA by linking with CoA. Acyl-CoA inserts into membrane phospholipids and binds to phosphatidylethanolamine to form PUFA phospholipids catalyzed by lysophosphatidylcholine acyltransferase 3 (83). Lipid peroxidation, via two primary mechanisms, non-enzymatic spontaneous oxidation and enzyme-mediated lipid peroxidation (84), results in the production of phospholipid hydroperoxides (PLOOHs), and if antioxidants do not convert PLOOH to phospholipid hydroxides (PLOHs) in a timely manner, the resulting accumulation of PLOOH leads to extensive lipid peroxidation and activation of the antioxidant system, directly inducing damage to cell membranes and ultimately leading to cell dysfunction and ferroptosis (85). Non-enzymatic lipid peroxidation (also known as lipid autoxidation) is a free radical-driven chain reaction. Hydroxyl radicals (OH·) are generated through the reaction of H2O2 with Fe2+, which reacts with PUFAs in the plasma membrane in a Fenton reaction to generate LPO, leading to ferroptosis (86,87). Enhanced accumulation of LPO is therefore necessary to increase the efficiency of ferroptosis (88), and OH· is the most active ROS (89), and thus, it also serves as an emerging cancer treatment target for the management of OC via chemodynamic therapy. H2O2 nano-enzymes developed by Sun et al (90) using CoNi alloys encapsulated with nitrogen-doped carbon nanotubes exhibited glucose oxidase and lactate oxidase activities to effectively disrupt the antioxidant defense system by catalyzing the formation of OH·, increasing ROS content in the tumor microenvironment and damaging the tumor cells, while depleting glutathione (GSH) to induce ferroptosis of the cells. Liang et al (91) demonstrated that polydopamine-mediated Michael addition combined with Fe2+ depletion of GSH enhanced OH· accumulation and ultimately resulted in the intracellular release of the chemotherapeutic drug doxorubicin, thus inducing ferroptosis. In addition, lipid peroxidation is tightly regulated by various metabolic and signaling pathways, such as the cytochrome P450 oxidoreductase pathway, and iron-containing enzymes, including lipoxygenase, also contribute to the process of lipid peroxidation (92). Enzymatic lipid peroxidation, conversely, is a process of direct oxidation of free PUFAs into various types of lipid hydroperoxides catalyzed by lipoxygenase (93,94). Among them, the arachidonic acid lipoxygenase family regulates lipid peroxidation. It has been shown that the binding of 5-lipoxygenase to microsomal GSH S-transferase 1 led to a decrease in lipid peroxidation and mediated ferroptosis in cancer cells (95). 12-lipoxygenase, conversely, has been identified as a key factor in p53-mediated ferroptosis under conditions of ROS-induced stress (96), while the expression levels of 15-lipoxygenase are associated with the spermine/spermine N1-acetyltransferase 1 gene, a transcriptional target of p53 (97). Zhang et al (98) demonstrated that chemotherapeutic drugs for OC induced excessive lipid peroxidation through ROS and initiated ovarian cell ferroptosis, thus leading to ovarian cell death. A study (99) has demonstrated that p53 serves an important role in the ferroptosis process. p53 exhibits a bidirectional regulatory effect based on specific environmental conditions. When lipid peroxidation levels are low, p53 inhibits the occurrence of ferroptosis, promoting cell survival. However, when excess lipid peroxidation persists, ferroptosis is induced (99).

In summary, further studies on the effects of various lipid metabolic pathways on lipid peroxidation and ferroptosis from the perspective of chemotherapy-induced oxidative stress and ferroptosis may help control ovarian damage and improve the quality of life of patients with OC. All of them will provide a deeper understanding of ferroptosis and therapeutic strategies for OC.

Lipid antioxidation in OC

Oxidative damage occurs due to an imbalance between the cellular antioxidant system with the production of free radicals and/or the neutralization or elimination of their deleterious effects. ROS-mediated lipid peroxidation is a key step to drive cellular ferroptosis, and inactivation of the antioxidant system is the primary cause of ferroptosis (100). At present, it has been shown that the primary antioxidant systems regulating ferroptosis include the System Xc−-GSH-GSH peroxidase 4 (GPX4) pathway, the ferroptosis suppressor protein 1 (FSP1)-coenzyme Q10 (CoQ10) pathway, the guanosine triphosphate cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4) pathway and the dihydroorotate dehydrogenase (DHODH)-CoQ10H2 pathway. Among these, the System Xc−-GSH-GPX4 pathway is the most fundamental antioxidant system, which serves a key role in protecting cells from ferroptosis (101). An overview of the primary antioxidant systems regulating ferroptosis is shown in Fig. 1.

Figure 1

Primary antioxidant systems regulating ferroptosis. System Xc−-GSH-GPX4: Cystine is oxidized to cysteine through the System Xc-, which leads to the synthesis of GSH, and GPX4 reduces PLOOH to PLOH with the participation of GSH, which induces the onset of ferroptosis when GPX4 is inhibited. FSP1-CoQ10-NADPH: FSP1 promotes the transfer of CoQ10 from mitochondria to the cell membrane by myristoylation of the N-terminus with the participation of CoQ10 and its reduction to CoQ10H2 catalyzed by NADPH, which prevents cellular ferroptosis by trapping free radicals. GCH1-BH4-DHFR: GCH1 is the rate-limiting enzyme for the biosynthesis of BH4. BH4 acts as a free radical-trapping antioxidant, inhibiting ferroptosis. It is recycled by DHFR and subjected to redox cycling. DHODH-CoQ10H2: DHODH is located on the outer surface of the inner mitochondrial membrane and inhibits cellular ferroptosis by reducing lipid reactive oxygen species in mitochondria by reducing CoQ10 to CoQ10H2. Supplementation with DHODH substrates or products (DHO or OA) regulates cellular ferroptosis. BH2, dihydrobiopterin; BH4, tetrahydrobiopterin; CoQ10, CoQ10, coenzyme Q10; CoQ10H2, ubiquinol-10; DHFR, dihydrofolate reductase; DHO, dihydroorotate; DHODH, dihydroorotate dehydrogenase; FSP1, ferroptosis suppressor protein 1; GCH1, guanosine triphosphate cyclohydrolase 1; GSH, glutathione; GPX4, glutathione peroxidase 4; OA, orotate; PLOH, phospholipid hydroxide; PLOOH, phospholipid hydroperoxide; PUFA, polyunsaturated fatty acid; SLC3A2, solute carrier family 3 member 2; SLC7A11, solute carrier family 7 member 11.

System Xc−-GSH-GPX4 pathway

The System Xc—GSH-GPX4 pathway serves a key role in the antioxidant defense mechanism of ferroptosis. System Xc− is a cystine-glutamate reverse transporter protein composed of solute carrier family 7 member 11 (SLC7A11) and solute carrier family 3 member 2 (102). System Xc− oxidizes intracellular cystine to cysteine, which is further converted to GSH (103). By contrast, GPX4 protects cells against ferroptosis by reducing reactive PLOOH to nontoxic PLOH, which involves GSH (the reducing cofactor of GPX4) (104). Inhibition of GPX4 induces lipid ROS accumulation and induces the onset of ferroptosis, thereby inhibiting tumor cell proliferation (105).

Studies have shown that erastin (38), sorafenib (106), sulfasalazine (107) and p53 (108) could cause GSH production and induce ferroptosis by inhibiting System Xc−. Metallothionein-1G is a key factor and potential therapeutic target for regulating sorafenib resistance in human hepatocellular carcinoma. Downregulating metallothionein-1G increases lipid peroxidation and GSH depletion, leading to ferroptosis in hepatocellular carcinoma (106). Yuan et al (109) used an innovative approach, employing a combination of chemotherapy and chemokinetic therapy, which inhibited malignant cell proliferation by inactivating GPX4 by inducing GSH depletion, and this approach showed a high degree of biosafety. Luo et al (110) found that paired box 8 (PAX8) (a GPX4-dependent OC susceptibility gene) depletion sensitized OC cells to GPX4 inhibitors. The combination of PAX8 inhibitor and RSL3 inhibited proliferation and induced ferroptosis in OC cells (110). In addition, the System Xc−-GSH-GPX4 pathway is a key antioxidant system that prevents lipid peroxidation-mediated ferroptosis, and blocking this pathway promotes the onset of ferroptosis in tumor cells and prevents chemotherapeutic resistance (111). Okuno et al (112) reported that the System Xc− transporter, which is involved in the transport of cystine and glutamate, had a regulatory effect on intracellular GSH levels and cisplatin resistance in OC cell lines. The results revealed that cisplatin-resistant variant OC cells had an ~4.5-fold higher rate of cystine uptake and higher intracellular GSH levels than OC cell lines due to the acquisition of cystine transporter activity mediated by the System Xc−; however, the GSH levels were decreased following glutamate overdose. Cystine uptake was also inhibited. It is therefore suggested that System Xc− serves an important role in maintaining higher levels of GSH and can confer cisplatin resistance in OC cell lines. In addition, a study has shown that the release of GSH and cysteine in OC fibroblasts contributes to the reduction of nuclear accumulation of platinum (112). Additionally, CD8+ T cells can inhibit this resistance by regulating GSH and cysteine metabolism in fibroblasts (113).

In summary, inhibition of System Xc−, depletion of GSH and reduction of GPX4 activity all mediate metabolic processes involving amino acids that increase sensitivity to ferroptosis-inducing agents, and targeting this system may reverse chemotherapeutic resistance and reduce the rate of OC progression.

FSP1-CoQ10-NADPH pathway

Bersuker et al (114) identified FSP1 as a potent resistance factor to ferroptosis, arguing that the FSP1-CoQ10-NADPH pathway is independent of the classical System Xc−-GSH-GPX4 pathway, highlighting another pathway involved in the antioxidant regulation of ferroptosis and suggesting that its pharmacological inhibition may render cancer cells sensitive to ferroptosis-inducing chemotherapeutic agents. FSP1 is a key protein that prevents cells from undergoing ferroptosis, and knockdown of FSP1 increases the sensitivity of cell lines to ferroptosis-inducing agents, as well as being a positive regulator of mitochondrial apoptosis. FSP1 is recruited to the plasma membrane via myristoylation, thereby inhibiting ferroptosis (114,115). FSP1 is primarily associated with the outer mitochondrial membrane and undergoes myristoylation at the N-terminus to promote the transfer of CoQ10 from mitochondria to the cell membrane. The NADPH-catalyzed reduction of CoQ10 to ubiquinol-10 (CoQ10H2) traps free radicals mediating lipid peroxidation, and thus prevents ferroptosis of cells (114). In addition, it has been shown that downregulation of stearoyl-CoA desaturase-1 was followed by a reduction in the lipophilic antioxidant CoQ10, which induced the potential for ferroptosis by inhibiting intracellular synthesis of protective lipids. Inhibition of stearoyl-CoA desaturase-1 enhanced the antitumor effects of ferroptosis inducers in OC cell lines. Combining stearoyl-CoA desaturase-1 inhibitors with ferroptosis inducers may offer a novel treatment option for the management of OC (116). The small molecule compound FIN56 inhibits CoQ10 synthesis in the mevalonate pathway by binding to and activating squalene synthase, resulting in a decrease in CoQ10 levels, and thus, increasing sensitivity to ferroptosis (117).

Nanogels developed by Yang et al (118) enhanced cellular lipid peroxidation through inhibition of the FSP1-CoQ10-NADPH pathway, leading to ferroptosis of immunogenic cells, and resulting in effective tumor ablation and immune responses in a mouse breast cancer model. Furthermore, downregulation of FSP1 in hepatocellular carcinoma cells promoted sorafenib-induced ferroptosis (119).

Therefore, the FSP1-CoQ10-NADPH pathway may complement and work with the System Xc−-GSH-GPX4 pathway to inhibit lipid peroxidation in ferroptosis, providing a potential therapeutic strategy for the treatment of OC.

GCH1-BH4-dihydrofolate reductase (DHFR) pathway

A previous study has identified the GCH1-BH4-DHFR pathway as an alternative compensatory mechanism for the System Xc−-GSH-GPX4 pathway (120). GCH1 is the rate-limiting enzyme for BH4 biosynthesis, which promotes ferroptosis via the metabolites BH4 and dihydrobiopterin (BH2). BH4 acts as a free radical trapping antioxidant that can be recycled by DHFR for redox cycling, and BH4 has antioxidant degradation effects on phospholipids containing two PUFA tails and prevents lipid peroxidation and thus ferroptosis by inhibiting the production of specific LPOs (120,121).

Through the direct trapping of antioxidant free radicals and the production of CoQ10 (120), when GCH1 is upregulated, it promotes BH4 production and abrogates the deleterious effects of RSL3-induced cellular ferroptosis. Additionally, overexpression of GCH1 has been shown to decrease the sensitivity of drug-resistant cancer cells to ferroptosis, which in turn further ameliorated the progression of ferroptosis in cancer cells through the regulation of CoQ10 (122).

In addition, relevant studies have shown that BH4 is involved in the synthesis of dopamine, nitric oxide synthase and melatonin (123), whereas exogenous dopamine or melatonin can inhibit ferroptosis (124). Several studies have shown that nitric oxide can induce or inhibit ferroptosis in tumor cells depending on environmental conditions (125-127). DHFR reduces BH2 through the involvement of NADPH, thereby promoting BH4 synthesis. If DHFR is inhibited, ferroptosis of tumor cells is promoted via the synergistic action of GPX4 inhibitors (87).

Therefore, the GCH1-BH4-DHFR pathway serves a role in regulating the balance between oxidative damage and antioxidant defense during ferroptosis and interacts with the System Xc−-GSH-GPX4 pathway and the FSP1-CoQ10-NADPH pathway in a synergistic or complementary manner. Although other specific mechanisms and potential therapeutic targets remain to be determined, the identified pathways may serve as potential therapeutic targets for overcoming resistance to chemotherapy in OC.

Mitochondrial DHODH-CoQ10H2 pathway

The mitochondrial DHODH-CoQ10H2 pathway and the FSP1-CoQ10-NADPH pathway are the two major lipid antioxidant systems in mitochondria. If one system is inhibited, the cell becomes more dependent on the other antioxidant system, and if both systems are inhibited, mitochondrial lipid peroxidation occurs, resulting in ferroptosis (128).

CoQ10H2, a free radical trapping antioxidant with anti-ferroptotic activity. DHODH is located on the outer surface of the inner mitochondrial membrane and inhibits cellular ferroptosis by converting CoQ10 to CoQ10H2 to reduce lipid ROS in mitochondria. In addition, supplementation with dihydroorotate or orotate, substrates or products of DHODH, attenuates or enhances the inhibitory effect of GPX4, respectively, thereby modulating cellular ferroptosis (129).

The mitochondrial DHODH-CoQ10H2 pathway and the FSP1-CoQ10-NADPH pathway function independently of each other, but they both reduce CoQ10 to CoQ10H2 to enhance the mitochondrial defense mechanism against ferroptosis.

ERS in OC

When tumor cells are challenged by intrinsic factors such as oncogenic activation, altered chromosome numbers or enhanced secretory capacity, as well as by extrinsic factors such as hypoxia, nutrient deprivation and acidosis, protein homeostasis is altered, resulting in the accumulation of unfolded or misfolded proteins in the lumen of the ER, activating the ERS and the UPR, thus restoring homeostasis to the cell (32). However, when ERS is prolonged or the stimulus is too strong, exceeding the tolerance threshold of the UPR leads to cell death, which in turn causes the development of cancer (130).

UPR is initiated by three major ERS sensors located in the ER membrane, including inositol-requiring protein 1α (IRE1α), protein kinase RNA-like ER kinase (PERK) and activating transcription factor 6 (ATF6) (131). The ER-resident chaperone binding immunoglobulin protein (BiP) acts as a master regulator of the UPR by binding to and inactivating three ERS sensors, IRE1α, PERK and ATF6, negatively regulating them and keeping them in an inactive state. When misfolded proteins accumulate in the ER, they bind to the inhibitory chaperone BiP and sequester it, which activates the ERS sensors to initiate UPR signaling (132). An overview of the mechanisms of ERS is shown in Fig. 2.

Figure 2

Mechanisms of ERS. Accumulation of unfolded or misfolded proteins in the lumen of the ER activates three transmembrane proteins of the unfolded protein response (IRE1, PERK and ATF6) and thereby restores cellular homeostasis. IRE1: Activation of IRE1 kinase results in the excision of an intron in the mRNA encoding the XBP1 transcription factor, and ligase mediates the linking of two mRNA fragments to produce stably active XBP1s. Stable XBP1s activity is involved in subsequent ER biogenesis. PERK: GADD34 forms a loop by regulating eIF2α dephosphorylation, thereby modulating ATF4-mediated ER biogenesis. ATF6: ATF6 is translocated to the Golgi under conditions of ERS and is sequentially hydrolyzed by S1P and S2P proteins, thereby regulating ER biogenesis. ATF4, activating transcription factor 4; ASK, apoptosis signal-regulating kinase; ATF6, activating transcription factor 6; BiP, binding immunoglobulin protein; eIF2α, eukaryotic translation initiation factor 2α; ER, endoplasmic reticulum; ERS, ER stress; GADD34, growth arrest DNA-damage 34; IRE1, inositol-requiring protein 1α; P, phosphorylated; PERK, protein kinase RNA-like ER kinase; RIDD, regulated IRE1-dependent decay; S1P, serine protease site 1; S2P, metalloprotease site 2; TRAF2, tumor necrosis factor receptor-associated factor 2; XBP1, homeostasis transcription factor X-box protein 1.

IRE1 pathway

IRE1 consists of two isoforms, IRE1α and IRE1β; IRE1β is primarily expressed in the respiratory and gastrointestinal tracts, whereas IRE1α is more widely expressed (133). The cytoplasmic tail of IRE1α has two domains, a serine/threonine kinase structural domain and a ribonuclease (RNase) structural domain, which work together (134). The kinase of IRE1α is activated upon binding to misfolded proteins and undergoes dimerization/coupling and trans-autophosphorylation, leading to ectopic activation of the structural domain of RNase (135). Under the catalysis of active RNase, the intron containing 26 nucleotides is excised from the mRNA encoding the homeostasis transcription factor X-box protein 1 (XBP1). Then, two mRNA fragments are cleaved by RNA splicing ligase RNA 2′,3′-cyclic phosphate and 5′-OH ligase, thereby generating the stable active transcription factor XBP1s (spliced form) (136,137). XBP1s (spliced form) is involved in the expression of genes encoding ER membrane biogenesis, ER protein folding, ER-associated degradation and multiple UPR targets (138). C/EBP homologous protein (CHOP), a major regulator of ERS-induced apoptosis, is activated by activating transcription factor 4 (ATF4) through the PERK-ATF4-CHOP pathway (139). In addition, IRE1α promotes apoptosis by activating the apoptosis signal-regulating kinase 1/JNK pathway, and by binding to tumor necrosis factor receptor-associated factor 2 (140). In addition, regulated IRE1-dependent decay is a novel UPR-regulated pathway that has been identified to control cell fate under ERS. Activated RNase can target other mRNAs and microRNAs (miRs) by regulating this pathway (141).

Zundell et al (142) showed that pharmacological inhibition of the IRE1α/XBP1 pathway may serve as a novel therapeutic strategy for Arid1a-mutant cancers, and that knockdown of the XBP1 gene improved survival in mice harboring Arid1a-inactivated ovarian clear cell carcinomas. Song et al (143) demonstrated that controlling ERS or targeting IRE1α-XBP1 signaling modulated mitochondrial activity, and thus, may help control T-cell metabolic adaptations and anti-tumorigenic capacity in patients with OC. The mitochondria-associated ER membrane can also serve as an important link between the mitochondria and the ER (144). Cubillos-Ruiz et al (145) showed that the IRE1α-XBP1 pathway of dendritic cells in the OC microenvironment was continuously activated due to sustained ERS, which disrupted the antigen-presenting capacity and metabolic homeostasis of the dendritic cells and diminished their protective function in supporting of T cells against tumors, highlighting a unique immunotherapeutic approach for the management of OC. OC cells utilize ERS for survival through activation of the IRE1α/XBP1s pathway among other pathways, and coactivator associated arginine methyltransferase 1 (CARM1), which is typically upregulated in OC cells, has been revealed to regulate the expression of XBP1s target genes and to be selectively sensitive to the inhibition of the IRE1α/XBP1s pathway, providing a potential therapeutic strategy for the treatment of CARM1-expressing OC (146).

PERK pathway

PERK is a type I transmembrane protein with similarities to IRE1, which has an ER lumenal dimerization structural domain and a cytoplasmic kinase structural domain. The tubulin dimerization structural domain of PERK has less sequence similarity to IRE1 but is structurally and functionally similar to the structural domain of IRE1. The cytoplasmic kinase structural domain of PERK also undergoes trans-autophosphorylation in response to ERS, but it differs from IRE1 in that PERK also phosphorylates the eukaryotic translation initiation factor 2α (eIF2α) at serine 51, and phosphorylated eIF2α inhibits overall translation of proteins, and thus, reduces the amount of proteins entering the lumen of the ER (147,148). In addition, phosphorylation of eIF2α alters the utilization efficiency of the AUG start codon (149), which results in preferential translation of the ATF4 mRNA (148).

ATF4 is a transcription factor that activates downstream UPR target genes, such as the expression of growth arrest DNA-damage 34 (GADD34), which induces CHOP expression (148,150). CHOP promotes DNA damage, inhibits cell proliferation and activates apoptosis by upregulating the pro-apoptotic members of the B-cell lymphoma-2 family of proteins (151). Therefore, ATF4 serves an important role in ER-functional gene expression, ERS-mediated ROS production and ERS-induced apoptosis. ATF4 can also regulate the dephosphorylation of eIF2α through GADD34 to form an inhibitory feedback loop to reverse PERK-mediated translational decay (152). In addition, PERK phosphorylates nuclear factor erythroid 2-related factor 2 (Nrf2), thereby upregulating antioxidants to promote cellular antioxidation (153). Overall, the PERK-eIF2α pathway first mediates the promotion of cell survival during ERS but shifts to the promotion of apoptosis when ERS is sustained and maintains cellular homeostatic balance by activating ATF4 and NRF2. Therefore, the PERK pathway is an attractive therapeutic target in OC therapy.

ATF6 pathway

ATF6 is a type II transmembrane protein with a carboxy-terminal stress-sensing lumenal structural domain and an amino-terminal bZip transcription factor structural domain (154). ATF6 is transported to the Golgi under conditions of ERS, where it is sequentially hydrolyzed by serine protease site 1 and metalloprotease site 2 (S2P) proteins to release its amino-terminal transcription factor structural domain, which, synergistically with XBP1, upregulates genes involved in protein folding and ER amplification, as well as genes involved in ER-associated degradation pathway components (155). It has been shown that ATF6 expression in OC tumor tissues is higher than that in normal ovarian tissues (156), and under irreversible ERS, it downregulates the levels of anti-apoptotic proteins (157). In addition, by regulating ATF6, the sensitivity of OC cells to chemotherapeutic agents can be altered (158). However, the role of ATF6 in ER-induced cell death and co-targeting of chemotherapeutic agents to improve survival in OC still needs to be explored further.

Ferroptosis and ERS interactions in OC

With the gradual increase in interest in ferroptosis and ERS, an increasing number of studies have shown that ferroptosis and ERS have an important impact on OC and that there is a close relationship between the two (35,159-161).

Chen et al (162) found that regulating ferroptosis in OC cells increased the anti-proliferative effect of cannabidiol derivatives and induced ferroptosis in OC cells, both in vitro and in vivo, effectively inhibiting the development of OC (163). Liu et al (164) used organoids to show that ovarian tumorigenesis was effectively inhibited by targeting ferroptosis. In addition, ferroptosis-related mechanisms can also reverse cisplatin resistance in OC (14), affecting drug resistance and prognosis in patients with OC (165,166). Li et al (25) facilitated the possible clinical transition of targeting ferroptosis for OC diagnosis and synergistic treatment by combining the ferroptosis mechanism with nanotechnology, MRI and cisplatin-based chemotherapeutic treatment (167,168).

ERS also serves a crucial role in the development and prognosis of OC. Studies have shown that activation of UPR sensors, and thus, induction of ERS can lead to apoptosis of OC cells (169), and regulation of ERS-associated targets influences drug resistance in OC treatment (170,171), highlighting potentially novel targets for the management of OC. Zhang et al (172) developed promising medical aids for the prognostic assessment of patients with epithelial OC by establishing a risk classifier for differentially expressed genes associated with ERS. Ma et al (173) utilized nanotechnology to accurately and durably induce ERS through photodynamic responses, thus inducing an antitumor effect in a mouse model of OC.

A growing body of research has shown a link between ferroptosis and ERS, which share common pathways (174-176), and that ROS, a byproduct of ERS, may exacerbate ferroptosis, while the ER is a key site for lipid peroxidation during ferroptosis, and conversely ROS produced during ferroptosis further exacerbate ERS. However, ferroptosis and ERS have been discussed separately regarding OC, and, to the best of our knowledge, there are no studies assessing their interactions in OC. In the present review, the potential therapeutic targets for OC treatment are examined based on a potential association between ERS and ferroptosis in OC using the existing literature. An overview of interactions between ferroptosis and ERS is shown in Fig. 3 and an overview of mechanisms associated with ferroptosis and ERS is shown in Table I.

Figure 3

Interactions between ferroptosis and ERS. ERS induces ferroptosis. ERS activates the IRE1α-XBP1s-Gα12, PERK-eIF2α-ATF4-CHOP, PERK-Nrf2-HO-1, PERK-P53-System Xc− and ATF6-PLA2G4A-AA-PGE2 pathways to induce ferroptosis. ERS inhibits ferroptosis. ERS inhibits ferroptosis by activating the PERK-eIF2α-ATF4-HSPA5 and PERK-eIF2α-ATF4-CHAC1 pathways. AA, arachidonic acid; ALOX12, arachidonate 12-lipoxygenase, 12S type; ATF4, activating transcription factor 4; ATF6, activating transcription factor 6; CHAC1, cation transport regulator homolog 1; CHOP, C/EBP homologous protein; eIF2α, eukaryotic translation initiation factor 2α; ERS, endoplasmic reticulum stress; Gα12, G protein subunit α 12; GSH, glutathione; HO-1, heme oxygenase-1; Hrd1, E3 ligase; HSPA5, heat shock 70 kDa protein 5; IRE1α, inositol-requiring protein 1α; miR, microRNA; Nrf2, nuclear factor erythroid 2-related factor 2; PERK, protein kinase RNA-like ER kinase; PGE2, prostaglandin E2; PLA2G4A, phospholipase A2 group IVA; XBP1, homeostasis transcription factor X-box protein 1.

Table I

Mechanisms associated with ferroptosis and ERS.

Table I

Mechanisms associated with ferroptosis and ERS.

First author/s, yearDrug/active ingredientExperimental model
Role in ferroptosis and ERSPathways/targets affecting ferroptosis/ERSRole as a potential therapeutic target(Refs.)
In vitroIn vivo
Dixon et al, 2012ErastinHT-1080-Induction of ferroptosisSystem Xc−Leads to non-apoptotic destruction of tumor cells(38)
Sun et al, 2016SorafenibHepaG2, Hep3BNude miceInduction of ferroptosisSystem Xc−Modulating drug resistance in hepatocellular carcinoma cells(106)
Guan et al, 2009SulfasalazineNCI-H69, NCI-H82-Induction of ferroptosisSystem Xc−Inhibition of small cell lung cancer cell proliferation(107)
Jiang et al, 2015p53H1299, U2OS, MCF7Nude miceInduction of ferroptosisSystem Xc−Inhibition of tumor cell proliferation(108)
Luo et al, 2024PAX8 inhibitor-Xenograft mouse modelInduction of ferroptosisGPX4Inhibits the proliferation of OC cells(110)
Yang et al, 2024 Chitosan-ferrocenesodium alginate crosslinked nanogels-Mouse breast modelInduction of ferroptosis FSP1-CoQ10-NADPHProducing effective tumor ablation in a mouse model of breast cancer(118)
Liu et al, 2023SorafenibHepG2, Huh-7Nude miceInduction of ferroptosisFSP1Novel opportunities for the development of ferroptosis-associated cancer therapies(119)
Zundell et al, 2021IRE1α inhibitorOVCA429, OVTOKO, SKOV3, OVISE, TOV21GMouse modelInduction of ERSIRE1α-XBP1A novel therapeutic strategy for ARID1A-mutant OC(142)
Song et al, 2018-Stage III-IV human OC tumors and malignant ascitesTransgenic miceAdjustment of ERSIRE1α-XBP1Contributes to the restoration of antitumor capacity and metabolic adaptation of cancer host T cells(143)
Lin et al, 2021IRE1α inhibitorHGSOC, ID8, UPK10Six-week-old female immunod-eficiency gamma (NSG) miceAdjustment of ERSIRE1α-XBP1A novel therapeutic strategy for CARM1-expressing cancers(146)
Meng et al, 2020DNA binding inhibitorHEY, HEY A8, SKOV3, SKOV3 ip1, OVCA420, OVCA429, OVCA433, A2780Nude miceInduction of ERSATF6Improving chemotherapy resistance in cancer cells(158)
Zhong et al, 2024Liprox-statin-1 and the iron chela tor deferoxamine-CIH model in 4-week-old male miceInduction of ferroptosis, and thus, ERSNrf2, PERK-ATF4-CHOPAttenuating chronic intermittent hypoxia-induced neuronal damage and cognitive dysfunction(34)
Han et al, 2024Polydatin-Male C57BL/ 6 miceInhibition of ERS and ferroptosisSIRT1Improving early brain injury after subarachnoid hemorrhage(179)
Wang et al, 20244-Phenylbu tyric acidBEAS-2B-Inhibition of ERS, and thus, ferroptosisGSH, ACSL4, COX-2, FTH1Prevention of acute lung injury(180)
Tak et al, 2022Acetaminophen or other ERS inducers-Gna12 KO miceInduction of ERS, and thus, ferroptosisIRE1α-XBP1, Gα12, miR-15α, ALOX12A promising strategy for constituting acute liver injury(190)
Liu et al, 2023Ferrostatin-1-DN miceInduction of ERS, and thus, ferroptosisXBP1-Hrd1-Nrf2, SLC7A11, GSHProviding insights into potential mechanisms that delay epithelial-mesenchymal transition in renal tubular epithelial cells(35)
Ji et al, 2024EsculinHCT116Colorectal cancer mouse modelInduction of ERS, and thus, ferroptosisPERK, Nrf2/HO-1, eIF2α/CHOPInhibition of colon cancer development and progression(191)
Guo et al, 2024Tanshinone IIAHepG2H22-bearing miceInhibition of ERS, and thus, ferroptosis PERK-ATF4-HSPA5Inhibits tumor growth in mice(192)
Xu et al, 2023Salidroside-male C57BL/ 6 J miceInhibition of ERS, and thus, ferroptosisAMPK-SIRT1, ATF4-CHAC1Mitigation of acetaminophen-induced acute liver injury(193)
Chen et al, 2019 DihydroartemisininU251, U373-Induction of ERS, and thus, ferroptosisPERK, ATF4, GPX4Potentially novel anticancer mechanism in gliomas(194)

[i] ACSL4, acyl-CoA synthetase long chain family member 4; ALOX12, arachidonate 12-lipoxygenase, 12S type; ARID1A, AT-rich interaction domain 1A; ATF4, activating transcription factor 4; ATF6, activating transcription factor 6; CARM1, coactivator associated arginine methyltransferase 1; CHAC1, cation transport regulator homolog 1; CHOP, C/EBP homologous protein; CIH, chronic intermittent hypoxia; CoQ10, coenzyme Q10; COX-2, cyclooxygenase-2; eIF2α, eukaryotic translation initiation factor 2α; ERS, endoplasmic reticulum stress; FSP1, ferroptosis suppressor protein 1; FTH1, ferritin heavy chain 1; Gα12, G protein subunit alpha 12; GPX4, glutathione peroxidase 4; GSH, glutathione; HO-1, heme oxygenase-1; Hrd1, E3 ligase; HSPA5, heat shock 70 kDa protein 5; IRE1α, inositol-requiring protein 1α; miR, microRNA; Nrf2, nuclear factor erythroid 2-related factor 2; OC, ovarian cancer; PAX8, paired box 8; PERK, protein kinase RNA-like ER kinase; SIRT1, sirtuin 1; SLC7A11, solute carrier family 7 member 11; XBP1, homeostasis transcription factor X-box protein 1.

A study has shown that ERS is modulated by regulation of ferroptosis (34). Zhong et al (34) demonstrated that ferroptosis and ferroptosis-mediated ERS cause damage to prefrontal cortex neurons, and that ferroptosis in prefrontal cortex neurons activates the ERS-associated PERK-ATF4-CHOP pathway. The ferroptosis inhibitor liproxstatin-1 and the iron ion chelator deferoxamine reduced the expression levels of partially restored ferroptosis-related proteins, upregulated Nrf2 expression, downregulated phosphorylated-PERK, ATF4 and CHOP, and reduced ERS by inhibiting ferroptosis. This resulted in the attenuation of chronic intermittent hypoxia-induced neuronal damage and cognitive dysfunction, providing a potential therapeutic target for neurocognitive dysfunction induced by exposure to chronic intermittent hypoxia (34). ERS serves an important role in the etiology of obesity-associated myocardial malformations, and ERS markers are upregulated in persistent obesity. Taurine deoxycholic acid attenuates obesity-associated ERS-induced myocardial dysfunction, whereas ferroptosis induces the elimination of the beneficial effects provided by taurine deoxycholic acid and enhances the effects of ERS (177). Yang et al (176) found that activation of ferroptosis signaling in tumor cells promoted the formation and secretion of exosomes containing unfolded and misfolded proteins, inhibited ERS, and improved the survival rate of tumor cells. Additionally, it has been shown that the ERS-ferroptosis signaling-exosome pathway induced ERS drug resistance, highlighting a potentially key intracellular signaling mechanism that may be involved in ER signaling, ER homeostasis and drug resistance in cancer (176). Dihydroartemisinin (DHA) can induce ferroptosis of immunogenic cells in lung cancer by accumulating LPO, and at the same time induce cellular ERS. Further analysis showed that ferroptosis inhibitors eliminate the DHA-induced ERS, highlighting a potential novel therapeutic avenue for the management of cancer with traditional Chinese medicines in cancer immunotherapy (178).

Similarly, the regulation of ERS can also affect ferroptosis. Han et al (179) showed that polydatin ameliorated early brain injury after subarachnoid hemorrhage by inhibiting ERS through upregulation of sirtuin 1 (SIRT1) expression, and thus, inhibiting ferroptosis in neuronal cells. Wang et al (180) revealed that the ERS inhibitor 4-phenylbutyric acid inhibited ferroptosis in airway epithelial cells to prevent acute lung injury by downregulating ERS, reversing the lipopolysaccharide-induced decrease in GSH, and inhibiting the expression of ferroptosis-associated proteins, Acyl-CoA synthetase long-chain family member 4, cyclooxygenase-2 and ferritin heavy chain 1, thus highlighting a potential means of intervention for management of acute lung injury. In vitro experiments on ovarian granulosa cells revealed elevated ROS production, lipid peroxidation and intracellular iron content in testosterone-treated cells. The expression levels of SLC7A11, a key protein of System XC−, were also altered, resulting in reduced intracellular GSH synthesis and cystine deficiency. This led to a decrease in the activity of GPX4, the primary intracellular antioxidant, thereby inducing ferroptosis in granulosa cells. However, the testosterone-induced ferroptosis process was reduced by ERS inhibitors (181). Jiang et al (182) found that IRE1α, a regulatory protein that serves an important role in UPR, also determines susceptibility to ferroptosis by regulating GSH synthesis, suggesting that inhibition of IRE1α is a promising therapeutic strategy for ameliorating pathologies associated with ferroptosis. In addition, it has been demonstrated that exogenous melatonin, a drug candidate for the treatment of hepatocyte ferroptosis in non-alcoholic fatty liver disease, inhibits hepatocyte ferroptosis by reducing ERS through the regulation of the type 1B/cAMP/protein kinase A/IRE1 pathway (183). The high-risk heavy metal cadmium induces hepatocyte ferroptosis, causing liver injury, and the occurrence of ferroptosis is often accompanied by activation of the PERK-eIF2α-ATF4-CHOP signaling pathway, which can be countered by inhibition of ERS to reduce ferroptosis; therefore, cadmium-induced ferroptosis is dependent on ERS (184). Cadmium also regulates ferroptosis and induces nephrotoxicity in renal tubular epithelial cells through the same aforementioned mechanisms, resulting in kidney injury (185). Data from an ulcerative colitis study suggest that ERS is involved in the development of ferroptosis. eIF2α is a key component of the PERK branch of the ERS response, and phosphorylated NF-κBp65 inhibits ERS, and thus, protects the intestinal epithelium from ferroptosis in ulcerative colitis by directly interacting with eIF2α (186).

Colorectal cancer is a common digestive malignancy in which both surgery and chemotherapy exhibit limited therapeutic efficacy (187). Tagitinin C, a natural product, induces ERS production, which results in nuclear translocation of Nrf2 and upregulation of HO-1. HO-1 is a downstream effector of Nrf2, which leads to an increase in the pool of unstable iron, and thus, promotes lipid peroxidation. This increase in the pool of unstable iron together with erastin exerts synergistic antitumor effects in inducing ferroptosis in colorectal cancer cells. Thus, tagitinin C has been identified as a novel ferroptosis inducer and potent sensitizer (188). Furthermore, in a prostate cancer study, the mediation of arachidonic acid release and prostaglandin E2 production via ATF6α-phospholipase A2 group IVA was found to protect prostate cancer cells from ferroptosis (189). Upregulation of Gα12 through the IRE1α-XBP1 pathway after ERS in hepatocytes thereby promotes hepatic ferroptosis and exacerbates acute liver injury through Rho associated coiled-coil containing protein kinase 1-mediated dysregulation of 12-lipoxygenase and miR-15α (190). Furthermore, in a study related to diabetic nephropathy, ERS downregulated SLC7A11 expression through the XBP1-E3 ligase-Nrf2 pathway, which decreased GSH antioxidant levels and enhanced cellular sensitivity to ferroptosis, thereby inducing ferroptosis, providing insights into the potential mechanism of delaying epithelial-mesenchymal transition in renal tubular epithelial cells (35).

In addition, several studies have demonstrated that herbal components can also improve cellular ferroptosis by modulating ERS. Esculin, a compound extracted from the cortex of the willow bark, inhibits the development and progression of colon cancer by activating the ERS PERK signaling pathway, and inducing apoptosis and ferroptosis through the Nrf2/HO-1 and eIF2α/CHOP pathways (191). Tanshinone IIA, the primary active ingredient of the traditional Chinese medicine Danshen, has been shown to exert antitumor effects primarily in the ER-mediated ferroptosis signaling pathway, downregulating the ferroptosis of tumor cells through the PERK-ATF4-heat shock 70 kDa protein 5 pathway (192). Acetaminophen overdose is a major cause of drug-induced liver injury. Salidroside inhibits ER stress-mediated ferroptosis via the ATF4-cation transport regulator homolog 1 axis by activating AMPK-SIRT1 signaling, and serves an important role in alleviating acetaminophen-induced acute liver injury (193). In a study on glioma, DHA-induced ERS upregulated ATF4 via PERK, thereby increasing the expression and activity of GPX4, thus inhibiting DHA-induced lipid peroxidation and protecting glioma cells from ferroptosis, highlighting a novel anticancer mechanism in glioma (194).

In addition, ERS induces Ca2+ release, and TF transport is regulated by cytoplasmic Ca2+ levels, thus affecting intracellular iron content and inducing ferroptosis in colon cancer cells (195). Ferroptosis therapy, which is centered on increased intracellular ROS production and LPO accumulation, has emerged as a novel treatment modality for lung cancer. A ferroptosis nano-inducer consisting of DHA and pH-responsive calcium phosphate is delivered to the lungs by nebulization. The burst of Ca2+ can mediate ERS, thereby promoting the accumulation of ROS and leading to the intensification of ferroptosis, providing a novel research direction for the treatment of lung cancer (196).

It has been shown that ERS and ferroptosis are also related in terms of tumor angiogenesis. In a glioma-related study, elevated expression of ATF4, a transcription factor that activates downstream UPR target genes, promoted angiogenesis by facilitating tumor shaping of vascular structures in a System Xc−-dependent manner, and erastin, a ferroptosis inducer, and RSL3, a GPX4 inhibitor, could reduce ATF4-induced angiogenesis (197).

Drug development research on natural compounds and their derivatives is a popular research topic for cancer treatment. Research on novel drugs also takes into consideration the side effects and off-target effects, which can negatively affect the quality of life of patients. Drugs with high specificity and selectivity need to be developed to minimize off-target effects and their potential toxicities (198). In OC, reducing off-target effects during treatment can help improve patient outcomes and prognosis (199). Studies have shown that off-target effects are strongly linked to ferroptosis and ERS. Dahlmanns et al (200) found that adjusting the mechanism of ferroptosis improved tumor therapy, but that interfering with the induction of ferroptosis produced an off-target effect, and thus, a reduction in therapeutic efficacy. Similarly, it has been shown that ERS may modulate off-target effects in glioblastoma multiforme (201). Inhibitors of ROS, which are closely related to ferroptosis and ERS, likewise have some degree of influence on off-target effects (202). Recent advances in research on coupled drug delivery systems (203), liposomes (204) and nanotechnology (205) in combination with bioactive agents for the treatment of a wide range of diseases, including cancer, may lead to improved efficacy through the localized and precise delivery of drugs, thereby reducing off-target effects.

In summary, ERS interacts with ferroptosis in several diseases through various pathways, regulatory proteins and related factors to regulate disease development, highlighting potential therapeutic targets and strategies for disease treatment and prevention. However, the specific mechanisms by which ERS interacts with ferroptosis in the development, treatment and prevention of OC remain to be investigated.

Conclusions and future perspectives

Ferroptosis and ERS are both increasingly popular topics in relation to cancer research, and studies have shown that ferroptosis and ERS are related to the development and potential treatment of gynecological tumors (13,20,32,206,207). OC, as the gynecological malignant tumor with the highest mortality rate, has attracted attention; however, there is no relevant research on the potential mechanism of the interaction between ERS and ferroptosis in OC. In the present review, the pathogenesis of ERS and ferroptosis in OC, and the common pathways and related links between the two in liver, lung and colorectal cancer, are discussed, with the aim of providing novel strategies for the prevention, treatment and prognosis of OC in the future.

Numerous studies (208-212) have shown that ferroptosis and ERS serve as potential therapeutic targets for preventing the occurrence and development of OC, as well as in the treatment and prognostic prediction of patients with OC, while the combination with other drugs and novel technologies has also achieved favorable therapeutic effects, and has provided novel avenues for OC research. In particular, the proliferation and growth of OC cells can be inhibited by regulating the levels of iron in OC cells, and ferroptosis can thus be induced in the cells (105), which affects the occurrence and development of OC. Additionally, the induction of ferroptosis can also be achieved to counter chemoresistance (213). ERS can inhibit cell proliferation by regulating relevant pathways (214) and targeting chemoresistant cancer cells, and ERS-induced apoptosis has been shown to improve the sensitivity of OC cells to paclitaxel, thereby improving the prognosis of patients with OC (215).

There is also an association between the two, with ERS inducing ferroptosis by causing Fe2+ accumulation and lipid peroxidation through related pathways (195), and the ER, which contains more than half of all the lipid bilayers in any given cell, being the source of lipids for the majority of the cytosolic membranes, thus being critical for the initiation of ferroptosis (216). Ferroptosis can maintain ER homeostasis by signaling and regulating the degree of ERS. Ferroptosis can also be positively regulated by inducing ERS through various pathways.

However, ferroptosis and ERS have different and complex mechanistic pathways, and several novel mechanisms, pathways and potential targets are gradually being identified, although it is likely that more remain to be uncovered. For example, the detailed mechanisms of the four pathways associated with lipid antioxidation, particularly the GCH1-BH4-DHFR pathway and the DHODH-CoQ10H2 pathway in OC, still remain to be explored. Oxidative stress in mitochondria is closely associated with related processes in ferroptosis and ERS, and the mechanisms involved in mitochondria are not fully understood at present. Furthermore, the early onset of OC is insidious, and regarding diagnostic accuracy, there is still a lack of research on the difference in iron content between OC tissues of different stages and normal ovarian tissues (55). Interfering with iron metabolism during early OC may inhibit the further development of OC (104). The occurrence, progression and treatment of OC is a complex process, and there is a complex connection between ERS and ferroptosis. Whether the relevant pathways present in other diseases have the same role in OC, whether there are obvious differences in the roles in different cells at different stages of OC, and whether they have the same regulatory role in the different stages of OC occurrence, development, and recurrence remain to be determined. Furthermore, the prognostic prediction of OC still has major deficiencies and lacks the support of clinical data. Although current studies on various diseases have shown that the crosstalk between ferroptosis and ERS has a related improvement effect on the diseases (34,217-219), there is still a lack of relevant research regarding mechanisms for the crosstalk between ERS and ferroptosis in terms of OC.

Based on the clearer understanding of potential targets of ERS and ferroptosis, the role of the mechanistic pathways in the process of OC, the impact of the crosstalk between the two, and how to translate the results of research and experiments into clinical practice is an important and challenging aspect. In terms of clinical index detection, the aforementioned mechanism and modern scientific technologies such as nano-molecular materials, MRI and X-ray may be combined to detect the pathological factors of OC before the onset of the disease, and analyze the efficacy or assess prognosis during and following treatment of the disease. Assessment of iron metabolism levels during relevant clinical testing may allow for earlier detection of OC, potentially improving treatment options and patient outcomes. The direction of these studies, which await further translation to the clinic, may serve as a means of assessing patients to improve prevention, and assess efficacy and recurrence. In terms of drug development, for the commonly used chemotherapeutic regimens, there are obvious side effects alongside drug resistance. Thus, additional screening of safe and effective therapeutic drugs is required. Whether ferroptosis inhibitors and inducers of ERS can be used clinically or as references for future drug development remains to be explored. In addition, several studies (14,80,192,220) have found that traditional Chinese herbs are effective in regulating the crosstalk between ferroptosis and ERS-related mechanisms in relation to OC, thus the dosage and clinical safety of these options should be investigated. Whether the combination of various drugs, chemotherapeutic treatments and modern technologies will have a synergistic effect or inhibit resistance to chemotherapy and side effects also needs to be tested.

The present review highlights the potential therapeutic promise of crosstalk between ferroptosis and ERS in OC, suggesting that the intricate interactions merit deeper exploration. In addition, several potential treatments have shown therapeutic potential and efficacy in combination with conventional chemotherapy for the treatment of OC in preclinical testing, and future studies are required to determine the detailed molecular mechanisms of these compounds and to assess their efficacy in the clinic. To address these unknown challenges, it is necessary to draw on the results of existing research to help understand the focus and direction of the research. After clarifying the mechanism of each target and pathway, it is necessary to screen for important links or representative factors and more precise biomarkers. For drug development, a large amount of clinical data is required.

Overall, understanding ferroptosis and ERS, and the potential crosstalk between them, may provide novel therapeutic approaches for the management of OC.

Availability of data and materials

Not applicable.

Authors' contributions

JY wrote and revised the manuscript. YW revised the manuscript. FL revised the manuscript. YZ and FH conceived the topic of study. Data authentication is not applicable. All authors have read and approved the final version of the 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.

Abbreviations:

OC

ovarian cancer

ER

endoplasmic reticulum

ERS

endoplasmic reticulum stress

UPR

unfolded protein response

TF

transferrin

TFR

transferrin receptor

ROS

reactive oxygen species

HGSOC

high-grade serous ovarian cancer

HIF

hypoxia-inducible factor

SLC40A1

solute carrier family 40 member 1

PUFA

polyunsaturated fatty acid

PLOOH

phospholipid hydroperoxide

PLOH

phospholipid hydroxide

OH·

hydroxyl radical

LPO

lipid peroxide

GSH

glutathione

GPX4

glutathione peroxidase 4

FSP1

ferroptosis suppressor protein 1

CoQ10

coenzyme Q10

CoQ10H2

ubiquinol-10

GCH1

guanosine triphosphate cyclohydrolase 1

BH4

tetrahydrobiopterin

BH2

dihydrobiopterin

DHFR

dihydrofolate reductase

DHODH

dihydroorotate dehydrogenase

PAX8

paired box 8

SLC7A11

solute carrier family 7 member 11

IRE1α

inositol-requiring protein 1α

PERK

protein kinase RNA-like ER kinase

ATF6

activating transcription factor 6

BiP

binding immunoglobulin protein

RNase

ribonuclease

XBP1

homeostasis transcription factor X-box protein 1

CHOP

C/EBP homologous protein

ATF4

activating transcription factor 4

miRs

microRNAs

CARM1

coactivator associated arginine methyltransferase 1

eIF2α

eukaryotic translation initiation factor 2α

GADD34

growth arrest DNA-damage 34

Nrf2

nuclear factor erythroid 2-related factor 2

S2P

metalloprotease site 2

DHA

dihydroartemisinin

HO-1

heme oxygenase-1

SIRT1

sirtuin 1

Acknowledgments

Not applicable.

Funding

The present review was supported by funding from the National Natural Science Foundation of China (grant no. 82274566).

References

1 

Jiang C, Shen C, Ni M, Huang L, Hu H, Dai Q, Zhao H and Zhu Z: Molecular mechanisms of cisplatin resistance in ovarian cancer. Genes Dis. 11:1010632023. View Article : Google Scholar : PubMed/NCBI

2 

Maioru OV, Radoi VE, Coman MC, Hotinceanu IA, Dan A, Eftenoiu AE, Burtavel LM, Bohiltea LC and Severin EM: Developments in genetics: better management of ovarian cancer patients. Int J Mol Sci. 24:159872023. View Article : Google Scholar : PubMed/NCBI

3 

Ekmann-Gade AW, Høgdall CK, Seibæk L, Noer MC, Fagö-Olsen CL and Schnack TH: Incidence, treatment, and survival trends in older versus younger women with epithelial ovarian cancer from 2005 to 2018: A nationwide Danish study. Gynecol Oncol. 164:120–128. 2022. View Article : Google Scholar

4 

Mallen A, Todd S, Robertson SE, Kim J, Sehovic M, Wenham RM, Extermann M and Chon HS: Impact of age, comorbidity, and treatment characteristics on survival in older women with advanced high grade epithelial ovarian cancer. Gynecol Oncol. 161:693–699. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Lumish MA, Kohn EC and Tew WP: Top advances of the year: Ovarian cancer. Cancer. 130:837–845. 2024. View Article : Google Scholar

6 

Wang A, Wang Y, Du C, Yang H, Wang Z, Jin C and Hamblin MR: Pyroptosis and the tumor immune microenvironment: A new battlefield in ovarian cancer treatment. Biochim Biophys Acta Rev Cancer. 1879:1890582024. View Article : Google Scholar

7 

Wong RW and Cheung ANY: Predictive and prognostic biomarkers in female genital tract tumours: An update highlighting their clinical relevance and practical issues. Pathology. 56:214–227. 2024. View Article : Google Scholar : PubMed/NCBI

8 

Wang Y, Liu L, Jin X and Yu Y: Efficacy and safety of mirvetuximab soravtansine in recurrent ovarian cancer with FRa positive expression: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 194:1042302024. View Article : Google Scholar

9 

Zhang C, Liu X, Jin S, Chen Y and Guo R: Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol Cancer. 21:472022. View Article : Google Scholar : PubMed/NCBI

10 

Wang N, Chen M, Wu M, Liao Y, Xia Q, Cai Z, He C, Tang Q, Zhou Y, Zhao L, et al: High-adhesion ovarian cancer cell resistance to ferroptosis: The activation of NRF2/FSP1 pathway by junctional adhesion molecule JAM3. Free Radic Biol Med. 228:1–13. 2025. View Article : Google Scholar

11 

Teng K, Ma H, Gai P, Zhao X and Qi G: SPHK1 enhances olaparib resistance in ovarian cancer through the NFκB/NRF2/ferroptosis pathway. Cell Death Discov. 11:292025. View Article : Google Scholar

12 

Yao Y, Wang B, Jiang Y, Guo H and Li Y: The mechanisms crosstalk and therapeutic opportunities between ferroptosis and ovary diseases. Front Endocrinol (Lausanne). 14:11940892023. View Article : Google Scholar : PubMed/NCBI

13 

Kapper C, Oppelt P, Arbeithuber B, Gyunesh AA, Vilusic I, Stelzl P and Rezk-Füreder M: Targeting ferroptosis in ovarian cancer: Novel strategies to overcome chemotherapy resistance. Life Sci. 349:1227202024. View Article : Google Scholar : PubMed/NCBI

14 

Guo W, Wang W, Lei F, Zheng R, Zhao X, Gu Y, Yang M, Tong Y and Wang Y: Angelica sinensis polysaccharide combined with cisplatin reverses cisplatin resistance of ovarian cancer by inducing ferroptosis via regulating GPX4. Biomed Pharmacother. 175:1166802024. View Article : Google Scholar : PubMed/NCBI

15 

Ni M, Zhou J, Zhu Z, Xu Q, Yin Z, Wang Y, Zheng Z and Zhao H: Shikonin and cisplatin synergistically overcome cisplatin resistance of ovarian cancer by inducing ferroptosis via upregulation of HMOX1 to promote Fe2+ accumulation. Phytomedicine. 112:1547012023. View Article : Google Scholar

16 

Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11:882020. View Article : Google Scholar : PubMed/NCBI

17 

Li Y, Liu J, Wu S, Xiao J and Zhang Z: Ferroptosis: Opening up potential targets for gastric cancer treatment. Mol Cell Biochem. 479:2863–2874. 2024. View Article : Google Scholar

18 

Tang D, Kroemer G and Kang R: Ferroptosis in hepatocellular carcinoma: From bench to bedside. Hepatology. 80:721–739. 2024. View Article : Google Scholar

19 

Qiu X, Bi Q, Wu J, Sun Z and Wang W: Role of ferroptosis in fibrosis: From mechanism to potential therapy. Chin Med J (Engl). 137:806–817. 2024. View Article : Google Scholar

20 

Hushmandi K, Klionsky DJ, Aref AR, Bonyadi M, Reiter RJ, Nabavi N, Salimimoghadam S and Saadat SH: Ferroptosis contributes to the progression of female-specific neoplasms, from breast cancer to gynecological malignancies in a manner regulated by non-coding RNAs: Mechanistic implications. Noncoding RNA Res. 9:1159–1177. 2024. View Article : Google Scholar : PubMed/NCBI

21 

Wu Y, Jia C, Liu W, Zhan W, Chen Y, Lu J, Bao Y, Wang S, Yu C, Zheng L, et al: Sodium citrate targeting Ca2+/CAMKK2 pathway exhibits anti-tumor activity through inducing apoptosis and ferroptosis in ovarian cancer. J Adv Res. 65:89–104. 2024. View Article : Google Scholar : PubMed/NCBI

22 

Zhao L, Yang H, Wang Y, Yang S, Jiang Q, Tan J, Zhao X and Zi D: STUB1 suppresses paclitaxel resistance in ovarian cancer through mediating HOXB3 ubiquitination to inhibit PARK7 expression. Commun Biol. 7:14392024. View Article : Google Scholar : PubMed/NCBI

23 

Li J, Chen M, Huang D, Li Z, Chen Y, Huang J, Chen Y, Zhou Z and Yu Z: Inhibition of Selenoprotein I promotes ferroptosis and reverses resistance to platinum chemotherapy by impairing Akt phosphorylation in ovarian cancer. MedComm (2020). 5:e700332024. View Article : Google Scholar : PubMed/NCBI

24 

Zhang B, Guo B, Kong H, Yang L, Yan H, Liu J, Zhou Y, An R and Wang F: Decoding the ferroptosis-related gene signatures and immune infiltration patterns in ovarian cancer: Bioinformatic prediction integrated with experimental validation. J Inflamm Res. 17:10333–10346. 2024. View Article : Google Scholar : PubMed/NCBI

25 

Li G, Shi S, Tan J, He L, Liu Q, Fang F, Fu H, Zhong M, Mai Z, Sun R, et al: Highly efficient synergistic chemotherapy and magnetic resonance imaging for targeted ovarian cancer therapy using hyaluronic acid-coated coordination polymer nanoparticles. Adv Sci (Weinh). 11:e23094642024. View Article : Google Scholar : PubMed/NCBI

26 

Hetz C, Chevet E and Oakes SA: Proteostasis control by the unfolded protein response. Nat Cell Biol. 17:829–838. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Sims SG, Cisney RN, Lipscomb MM and Meares GP: The role of endoplasmic reticulum stress in astrocytes. Glia. 70:5–19. 2022. View Article : Google Scholar :

28 

Hu H, Tian M, Ding C and Yu S: The C/EBP homologous protein (CHOP) transcription factor functions in endoplasmic reticulum stress-induced apoptosis and microbial infection. Front Immunol. 9:30832019. View Article : Google Scholar : PubMed/NCBI

29 

Ren J, Bi Y, Sowers JR, Hetz C and Zhang Y: Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases. Nat Rev Cardiol. 18:499–521. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Oakes SA and Papa FR: The role of endoplasmic reticulum stress in human pathology. Annu Rev Pathol. 10:173–194. 2015. View Article : Google Scholar

31 

Song M and Cubillos-Ruiz JR: Endoplasmic reticulum stress responses in intratumoral immune cells: Implications for cancer immunotherapy. Trends Immunol. 40:128–141. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Yan T, Ma X, Guo L and Lu R: Targeting endoplasmic reticulum stress signaling in ovarian cancer therapy. Cancer Biol Med. 20:748–764. 2023.PubMed/NCBI

33 

Wang L, Ma X, Zhou L, Luo M, Lu Y, Wang Y, Zheng P, Liu H, Liu X, Liu W and Wei S: Dual-targeting TrxR-EGFR Alkynyl-Au(I) gefitinib complex induces ferroptosis in gefitinib-resistant lung cancer via degradation of GPX4. J Med Chem. 68:5275–5291. 2025. View Article : Google Scholar : PubMed/NCBI

34 

Zhong P, Li L, Feng X, Teng C, Cai W, Zheng W, Wei J, Li X, He Y, Chen B, et al: Neuronal ferroptosis and ferroptosismediated endoplasmic reticulum stress: Implications in cognitive dysfunction induced by chronic intermittent hypoxia in mice. Int Immunopharmacol. 138:1125792024. View Article : Google Scholar

35 

Liu Z, Nan P, Gong Y, Tian L, Zheng Y and Wu Z: Endoplasmic reticulum stress-triggered ferroptosis via the XBP1-Hrd1-Nrf2 pathway induces EMT progression in diabetic nephropathy. Biomed Pharmacother. 164:1148972023. View Article : Google Scholar : PubMed/NCBI

36 

Song W, Zhang W, Yue L and Lin W: Revealing the effects of endoplasmic reticulum stress on ferroptosis by two-channel real-time imaging of pH and viscosity. Anal Chem. 94:6557–6565. 2022. View Article : Google Scholar : PubMed/NCBI

37 

Ye R, Mao YM, Fei YR, Shang Y, Zhang T, Zhang ZZ, Liu YL, Li JY, Chen SL and He YB: Targeting ferroptosis for the treatment of female reproductive system disorders. J Mol Med (Berl). 103:381–402. 2025. View Article : Google Scholar : PubMed/NCBI

38 

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

39 

Muckenthaler MU, Rivella S, Hentze MW and Galy B: A red carpet for iron metabolism. Cell. 168:344–361. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Galy B, Conrad M and Muckenthaler M: Mechanisms controlling cellular and systemic iron homeostasis. Nat Rev Mol Cell Biol. 25:133–155. 2024. View Article : Google Scholar

41 

Anderson GJ and Frazer DM: Current understanding of iron homeostasis. Am J Clin Nutr. 106(Suppl 6): 1559S–1566S. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Gulec S, Anderson GJ and Collins JF: Mechanistic and regulatory aspects of intestinal iron absorption. Am J Physiol Gastrointest Liver Physiol. 307:G397–G409. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Montalbetti N, Simonin A, Kovacs G and Hediger MA: Mammalian iron transporters: Families SLC11 and SLC40. Mol Aspects Med. 34:270–287. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Li H, Wang D, Wu H, Shen H, Lv D, Zhang Y, Lu H, Yang J, Tang Y and Li M: SLC46A1 contributes to hepatic iron metabolism by importing heme in hepatocytes. Metabolism. 110:1543062020. View Article : Google Scholar : PubMed/NCBI

45 

Mayneris-Perxachs J, Moreno-Navarrete JM and Fernández Real JM: The role of iron in host-microbiota crosstalk and its effects on systemic glucose metabolism. Nat Rev Endocrinol. 18:683–698. 2022. View Article : Google Scholar : PubMed/NCBI

46 

Seyoum Y, Baye K and Humblot C: Iron homeostasis in host and gut bacteria-a complex interrelationship. Gut Microbes. 13:1–19. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Zhao H, Tang C, Wang M, Zhao H and Zhu Y: Ferroptosis as an emerging target in rheumatoid arthritis. Front Immunol. 14:12608392023. View Article : Google Scholar : PubMed/NCBI

48 

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

49 

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

50 

Zhang L, Rao J, Liu X, Wang X, Wang C, Fu S and Xiao J: Attenuation of sepsis-induced acute kidney injury by exogenous H2S via inhibition of ferroptosis. Molecules. 28:47702023. View Article : Google Scholar

51 

Deng M, Tang F, Chang X, Zhang Y, Liu P, Ji X, Zhang Y, Yang R, Jiang J, He J and Miao J: A targetable OSGIN1-AMPK-SLC2A3 axis controls the vulnerability of ovarian cancer to ferroptosis. NPJ Precis Oncol. 9:152025. View Article : Google Scholar

52 

Torti SV and Torti FM: Iron and cancer: more ore to be mined. Nat Rev Cancer. 13:342–355. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Torti SV and Torti FM: Ironing out cancer. Cancer Res. 71:1511–1514. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Toyokuni S: The origin and future of oxidative stress pathology: From the recognition of carcinogenesis as an iron addiction with ferroptosis-resistance to non-thermal plasma therapy. Pathol Int. 66:245–259. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Basuli D, Tesfay L, Deng Z, Paul B, Yamamoto Y, Ning G, Xian W, McKeon F, Lynch M, Crum CP, et al: Iron addiction: A novel therapeutic target in ovarian cancer. Oncogene. 36:4089–4099. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Chen Y, Liao X, Jing P, Hu L, Yang Z, Yao Y, Liao C and Zhang S: Linoleic acid-glucosamine hybrid for endogenous iron-activated ferroptosis therapy in high-grade serous ovarian cancer. Mol Pharm. 19:3187–3198. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Piskounova E, Agathocleous M, Murphy MM, Hu Z, Huddlestun SE, Zhao Z, Leitch AM, Johnson TM, DeBerardinis RJ and Morrison SJ: Oxidative stress inhibits distant metastasis by human melanoma cells. Nature. 527:186–191. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Bauckman K, Haller E, Taran N, Rockfield S, Ruiz-Rivera A and Nanjundan M: Iron alters cell survival in a mitochondria-dependent pathway in ovarian cancer cells. Biochem J. 466:401–413. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Xu X, Wang Y, Guo W, Zhou Y, Lv C, Chen X and Liu K: The significance of the alteration of 8-OHdG in serous ovarian carcinoma. J Ovarian Res. 6:742013. View Article : Google Scholar : PubMed/NCBI

60 

Valavanidis A, Vlachogianni T and Fiotakis C: 8-Hydroxy-2′-deoxyguanosine (8-OHdG): A critical biomarker of oxidative stress and carcinogenesis. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 27:120–139. 2009.PubMed/NCBI

61 

Prat J, D'Angelo E and Espinosa I: Ovarian carcinomas: At least five different diseases with distinct histological features and molecular genetics. Hum Pathol. 80:11–27. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Shen J, Sheng X, Chang Z, Wu Q, Wang S, Xuan Z, Li D, Wu Y, Shang Y, Kong X, et al: Iron metabolism regulates p53 signaling through direct heme-p53 interaction and modulation of p53 localization, stability, and function. Cell Rep. 7:180–193. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Deng Z, Manz DH, Torti SV and Torti FM: Effects of ferroportin-mediated iron depletion in cells representative of different histological subtypes of prostate cancer. Antioxid Redox Signal. 30:1043–1061. 2019. View Article : Google Scholar :

64 

Hann HW, Stahlhut MW and Blumberg BS: Iron nutrition and tumor growth: Decreased tumor growth in iron-deficient mice. Cancer Res. 48:4168–4170. 1988.PubMed/NCBI

65 

White S, Taetle R, Seligman PA, Rutherford M and Trowbridge IS: Combinations of anti-transferrin receptor monoclonal antibodies inhibit human tumor cell growth in vitro and in vivo: Evidence for synergistic antiproliferative effects. Cancer Res. 50:6295–6301. 1990.PubMed/NCBI

66 

Sandoval-Acuña C, Torrealba N, Tomkova V, Jadhav SB, Blazkova K, Merta L, Lettlova S, Adamcová MK, Rosel D, Brábek J, et al: Targeting mitochondrial iron metabolism suppresses tumor growth and metastasis by inducing mitochondrial dysfunction and mitophagy. Cancer Res. 81:2289–2303. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Moon EJ, Mello SS, Li CG, Chi JT, Thakkar K, Kirkland JG, Lagory EL, Lee IJ, Diep AN, Miao Y, et al: The HIF target MAFF promotes tumor invasion and metastasis through IL11 and STAT3 signaling. Nat Commun. 12:43082021. View Article : Google Scholar : PubMed/NCBI

68 

Wang X, Du ZW, Xu TM, Wang XJ, Li W, Gao JL, Li J and Zhu H: HIF-1α is a rational target for future ovarian cancer therapies. Front Oncol. 11:7851112021. View Article : Google Scholar

69 

Benyamin B, Esko T, Ried JS, Radhakrishnan A, Vermeulen SH, Traglia M, Gögele M, Anderson D, Broer L, Podmore C, et al: Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis. Nat Commun. 5:49262014. View Article : Google Scholar : PubMed/NCBI

70 

Wu J, Bao L, Zhang Z and Yi X: Nrf2 induces cisplatin resistance via suppressing the iron export related gene SLC40A1 in ovarian cancer cells. Oncotarget. 8:93502–93515. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Szymonik J, Wala K, Górnicki T, Saczko J, Pencakowski B and Kulbacka J: The impact of iron chelators on the biology of cancer stem cells. Int J Mol Sci. 23:892021. View Article : Google Scholar

72 

Wang L, Li X, Mu Y, Lu C, Tang S, Lu K, Qiu X, Wei A, Cheng Y and Wei W: The iron chelator desferrioxamine synergizes with chemotherapy for cancer treatment. J Trace Elem Med Biol. 56:131–138. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Greenshields AL, Shepherd TG and Hoskin DW: Contribution of reactive oxygen species to ovarian cancer cell growth arrest and killing by the anti-malarial drug artesunate. Mol Carcinog. 56:75–93. 2017. View Article : Google Scholar

74 

Yang ND, Tan SH, Ng S, Shi Y, Zhou J, Tan KS, Wong WS and Shen HM: Artesunate induces cell death in human cancer cells via enhancing lysosomal function and lysosomal degradation of ferritin. J Biol Chem. 289:33425–33441. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Cheng Y, Qu W, Li J, Jia B, Song Y, Wang L, Rui T, Li Q and Luo C: Ferristatin II, an iron uptake inhibitor, exerts neuroprotection against traumatic brain injury via suppressing ferroptosis. ACS Chem Neurosci. 13:664–675. 2022. View Article : Google Scholar : PubMed/NCBI

76 

Zhang Y, He F, Hu W, Sun J, Zhao H, Cheng Y, Tang Z, He J, Wang X, Liu T, et al: Bortezomib elevates intracellular free Fe2+ by enhancing NCOA4-mediated ferritinophagy and synergizes with RSL-3 to inhibit multiple myeloma cells. Ann Hematol. 103:3627–3637. 2024. View Article : Google Scholar : PubMed/NCBI

77 

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

78 

Stockwell BR, Friedmann 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

79 

Mortensen MS, Ruiz J and Watts JL: Polyunsaturated fatty acids drive lipid peroxidation during ferroptosis. Cells. 12:8042023. View Article : Google Scholar : PubMed/NCBI

80 

Liu S, Yang X, Zheng S, Chen C, Qi L, Xu X and Zhang D: Research progress on the use of traditional Chinese medicine to treat diseases by regulating ferroptosis. Genes Dis. 12:1014512024. View Article : Google Scholar

81 

Rodencal J and Dixon SJ: A tale of two lipids: Lipid unsaturation commands ferroptosis sensitivity. Proteomics. 23:e21003082023. View Article : Google Scholar

82 

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

83 

Forcina GC and Dixon SJ: GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 19:e18003112019. View Article : Google Scholar : PubMed/NCBI

84 

Yin H, Xu L and Porter NA: Free radical lipid peroxidation: Mechanisms and analysis. Chem Rev. 111:5944–5972. 2011. View Article : Google Scholar : PubMed/NCBI

85 

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

86 

Murphy MP: How mitochondria produce reactive oxygen species. Biochem J. 417:1–13. 2009. View Article : Google Scholar

87 

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

88 

Chen J, Duan Z, Deng L, Li L, Li Q, Qu J, Li X and Liu R: Cell membrane-targeting type I/II photodynamic therapy combination with FSP1 inhibition for ferroptosis-enhanced photodynamic immunotherapy. Adv Healthc Mater. 13:e23044362024. View Article : Google Scholar : PubMed/NCBI

89 

Wang Q, Ji H, Hao Y, Jia D, Ma H, Song C, Qi H, Li Z and Zhang C: Illumination of hydroxyl radical generated in cells during ferroptosis, Arabidopsis thaliana, and mice using a new turn-on near-infrared fluorescence probe. Anal Chem. 96:20189–20196. 2024. View Article : Google Scholar : PubMed/NCBI

90 

Sun M, Chen Q, Ren Y, Zhuo Y, Xu S, Rao H, Wu D, Feng B and Wang Y: CoNiCoNC tumor therapy by two-ways producing H2O2 ferroptosis. J to aggravate energy metabolism, chemokinetics, and Colloid Interface Sci. 678:925–937. 2025. View Article : Google Scholar

91 

Liang KA, Chih HY, Liu IJ, Yeh NT, Hsu TC, Chin HY, Tzang BS and Chiang WH: Tumor-targeted delivery of hyaluronic acid/polydopamine-coated Fe2+-doped nano-scaled metal-organic frameworks with doxorubicin payload for glutathione depletion-amplified chemodynamic-chemo cancer therapy. J Colloid Interface Sci. 677:400–415. 2025. View Article : Google Scholar

92 

Stockwell BR: Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell. 185:2401–2421. 2022. View Article : Google Scholar : PubMed/NCBI

93 

Foret MK, Lincoln R, Do Carmo S, Cuello AC and Cosa G: Connecting the 'Dots': From free radical lipid autoxidation to cell pathology and disease. Chem Rev. 120:12757–12787. 2020. View Article : Google Scholar : PubMed/NCBI

94 

Stoyanovsky DA, Tyurina YY, Shrivastava I, Bahar I, Tyurin VA, Protchenko O, Jadhav S, Bolevich SB, Kozlov AV, Vladimirov YA, et al: Iron catalysis of lipid peroxidation in ferroptosis: Regulated enzymatic or random free radical reaction? Free Radic Biol Med. 133:153–161. 2019. View Article : Google Scholar :

95 

Kuang F, Liu J, Xie Y, Tang D and Kang R: MGST1 is a redox-sensitive repressor of ferroptosis in pancreatic cancer cells. Cell Chem Biol. 28:765–775.e5. 2021. View Article : Google Scholar : PubMed/NCBI

96 

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

97 

Ou Y, Wang SJ, Li D, Chu B and Gu W: Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci USA. 113:E6806–E6812. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Zhang S, Liu Q, Chang M, Pan Y, Yahaya BH, Liu Y and Lin J: Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis. Cell Death Dis. 14:3402023. View Article : Google Scholar : PubMed/NCBI

99 

Xu R, Wang W and Zhang W: Ferroptosis and the bidirectional regulatory factor p53. Cell Death Discov. 9:1972023. View Article : Google Scholar : PubMed/NCBI

100 

Kuang F, Liu J, Tang D and Kang R: Oxidative damage and antioxidant defense in ferroptosis. Front Cell Dev Biol. 8:5865782020. View Article : Google Scholar : PubMed/NCBI

101 

Liu Y, Lu S, Wu LL, Yang L, Yang L and Wang J: The diversified role of mitochondria in ferroptosis in cancer. Cell Death Dis. 14:5192023. View Article : Google Scholar : PubMed/NCBI

102 

Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, et al: The cystine/glutamate antiporter system x(c)(-) in health and disease: From molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 18:522–555. 2013. View Article : Google Scholar :

103 

Hu S, Chu Y, Zhou X and Wang X: Recent advances of ferroptosis in tumor: From biological function to clinical application. Biomed Pharmacother. 166:1154192023. View Article : Google Scholar : PubMed/NCBI

104 

Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI

105 

Li D, Zhang M and Chao H: Significance of glutathione peroxidase 4 and intracellular iron level in ovarian cancer cells-'utilization' of ferroptosis mechanism. Inflamm Res. 70:1177–1189. 2021. View Article : Google Scholar : PubMed/NCBI

106 

Sun X, Niu X, Chen R, He W, Chen D, Kang R and Tang D: Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 64:488–500. 2016. View Article : Google Scholar : PubMed/NCBI

107 

Guan J, Lo M, Dockery P, Mahon S, Karp CM, Buckley AR, Lam S, Gout PW and Wang YZ: The xc-cystine/glutamate antiporter as a potential therapeutic target for small-cell lung cancer: Use of sulfasalazine. Cancer Chemother Pharmacol. 64:463–472. 2009. View Article : Google Scholar

108 

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

109 

Yuan J, Wang J, Song M, Zhao Y, Shi Y and Zhao L: Brain-targeting biomimetic disguised manganese dioxide nanoparticles via hybridization of tumor cell membrane and bacteria vesicles for synergistic chemotherapy/chemodynamic therapy of glioma. J Colloid Interface Sci. 676:378–395. 2024. View Article : Google Scholar : PubMed/NCBI

110 

Luo Y, Liu X, Chen Y, Tang Q, He C, Ding X, Hu J, Cai Z, Li X, Qiao H and Zou Z: Targeting PAX8 sensitizes ovarian cancer cells to ferroptosis by inhibiting glutathione synthesis. Apoptosis. 29:1499–1514. 2024. View Article : Google Scholar : PubMed/NCBI

111 

Li FJ, Long HZ, Zhou ZW, Luo HY, Xu SG and Gao LC: System Xc -/GSH/GPX4 axis: An important antioxidant system for the ferroptosis in drug-resistant solid tumor therapy. Front Pharmacol. 13:9102922022. View Article : Google Scholar

112 

Okuno S, Sato H, Kuriyama-Matsumura K, Tamba M, Wang H, Sohda S, Hamada H, Yoshikawa H, Kondo T and Bannai S: Role of cystine transport in intracellular glutathione level and cisplatin resistance in human ovarian cancer cell lines. Br J Cancer. 88:951–956. 2003. View Article : Google Scholar : PubMed/NCBI

113 

Wang W, Kryczek I, Dostál L, Lin H, Tan L, Zhao L, Lu F, Wei S, Maj T, Peng D, et al: Effector T cells abrogate stroma-mediated chemoresistance in ovarian cancer. Cell. 165:1092–1105. 2016. View Article : Google Scholar : PubMed/NCBI

114 

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

115 

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

116 

Tesfay L, Paul BT, Konstorum A, Deng Z, Cox AO, Lee J, Furdui CM, Hegde P, Torti FM and Torti SV: Stearoyl-CoA desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer Res. 79:5355–5366. 2019. View Article : Google Scholar : PubMed/NCBI

117 

Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, Brown LM, Valenzuela CA, Wolpaw AJ and Stockwell BR: Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 12:497–503. 2016. View Article : Google Scholar : PubMed/NCBI

118 

Yang N, Pan X, Zhou X, Liu Z, Yang J, Zhang J, Jia Z and Shen Q: Biomimetic nanoarchitectonics with chitosan nanogels for collaborative induction of ferroptosis and anticancer immunity for cancer therapy. Adv Healthc Mater. 13:e23027522024. View Article : Google Scholar

119 

Liu MR, Shi C, Song QY, Kang MJ, Jiang X, Liu H and Pei DS: Sorafenib induces ferroptosis by promoting TRIM54-mediated FSP1 ubiquitination and degradation in hepatocellular carcinoma. Hepatol Commun. 7:e02462023. View Article : Google Scholar : PubMed/NCBI

120 

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

121 

Ge A, Xiang W, Li Y, Zhao D, Chen J, Daga P, Dai CC, Yang K, Yan Y, Hao M, et al: Broadening horizons: The multifaceted role of ferroptosis in breast cancer. Front Immunol. 15:14557412024. View Article : Google Scholar : PubMed/NCBI

122 

Wang M, Liu J, Yu W, Shao J, Bao Y, Jin M, Huang Q and Huang G: Gambogenic acid suppresses malignant progression of non-small cell lung cancer via GCH1-mediated ferroptosis. Pharmaceuticals (Basel). 18:3742025. View Article : Google Scholar : PubMed/NCBI

123 

Werner ER, Blau N and Thöny B: Tetrahydrobiopterin: Biochemistry and pathophysiology. Biochem J. 438:397–414. 2011. View Article : Google Scholar : PubMed/NCBI

124 

NaveenKumar SK, Hemshekhar M, Kemparaju K and Girish KS: Hemin-induced platelet activation and ferroptosis is mediated through ROS-driven proteasomal activity and inflammasome activation: Protection by melatonin. Biochim Biophys Acta Mol Basis Dis. 1865:2303–2316. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Homma T, Kobayashi S, Conrad M, Konno H, Yokoyama C and Fujii J: Nitric oxide protects against ferroptosis by aborting the lipid peroxidation chain reaction. Nitric Oxide. 115:34–43. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Jiang L, Zheng H, Lyu Q, Hayashi S, Sato K, Sekido Y, Nakamura K, Tanaka H, Ishikawa K, Kajiyama H, et al: Lysosomal nitric oxide determines transition from autophagy to ferroptosis after exposure to plasma-activated Ringer's lactate. Redox Biol. 43:1019892021. View Article : Google Scholar : PubMed/NCBI

127 

Deng G, Li Y, Ma S, Gao Z, Zeng T, Chen L, Ye H, Yang M, Shi H, Yao X, et al: Caveolin-1 dictates ferroptosis in the execution of acute immune-mediated hepatic damage by attenuating nitrogen stress. Free Radic Biol Med. 148:151–161. 2020. View Article : Google Scholar

128 

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

129 

Vasan K, Werner M and Chandel NS: Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 32:341–352. 2020. View Article : Google Scholar : PubMed/NCBI

130 

Wang Y, Wang K, Jin Y and Sheng X: Endoplasmic reticulum proteostasis control and gastric cancer. Cancer Lett. 449:263–271. 2019. View Article : Google Scholar : PubMed/NCBI

131 

Hetz C: The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 13:89–102. 2012. View Article : Google Scholar : PubMed/NCBI

132 

Kopp MC, Larburu N, Durairaj V, Adams CJ and Ali MMU: UPR proteins IRE1 and PERK switch BiP from chaperone to ER stress sensor. Nat Struct Mol Biol. 26:1053–1062. 2019. View Article : Google Scholar : PubMed/NCBI

133 

Tabas I and Ron D: Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol. 13:184–190. 2011. View Article : Google Scholar : PubMed/NCBI

134 

Lu Y, Liang FX and Wang X: A synthetic biology approach identifies the mammalian UPR RNA ligase RtcB. Mol Cell. 55:758–770. 2014. View Article : Google Scholar : PubMed/NCBI

135 

Belyy V, Zuazo-Gaztelu I, Alamban A, Ashkenazi A and Walter P: Endoplasmic reticulum stress activates human IRE1α through reversible assembly of inactive dimers into small oligomers. Elife. 11:e743422022. View Article : Google Scholar

136 

Grandjean JMD, Madhavan A, Cech L, Seguinot BO, Paxman RJ, Smith E, Scampavia L, Powers ET, Cooley CB, Plate L, et al: Pharmacologic IRE1/XBP1s activation confers targeted ER proteostasis reprogramming. Nat Chem Biol. 16:1052–1061. 2020. View Article : Google Scholar : PubMed/NCBI

137 

Mahdizadeh SJ, Stier M, Carlesso A, Lamy A, Thomas M and Eriksson LA: Multiscale in silico study of the mechanism of activation of the RtcB ligase by the PTP1B phosphatase. J Chem Inf Model. 64:905–917. 2024. View Article : Google Scholar : PubMed/NCBI

138 

Walter P and Ron D: The unfolded protein response: From stress pathway to homeostatic regulation. Science. 334:1081–1086. 2011. View Article : Google Scholar : PubMed/NCBI

139 

Choy KW, Murugan D and Mustafa MR: Natural products targeting ER stress pathway for the treatment of cardiovascular diseases. Pharmacol Res. 132:119–129. 2018. View Article : Google Scholar : PubMed/NCBI

140 

Yang J and Yao S: JNK-Bcl-2/Bcl-xL-Bax/Bak pathway mediates the crosstalk between matrine-induced autophagy and apoptosis via interplay with beclin 1. Int J Mol Sci. 16:25744–25758. 2015. View Article : Google Scholar : PubMed/NCBI

141 

Maurel M, Chevet E, Tavernier J and Gerlo S: Getting RIDD of RNA: IRE1 in cell fate regulation. Trends Biochem Sci. 39:245–254. 2014. View Article : Google Scholar : PubMed/NCBI

142 

Zundell JA, Fukumoto T, Lin J, Fatkhudinov N, Nacarelli T, Kossenkov AV, Liu Q, Cassel J, Hu CA, Wu S and Zhang R: Targeting the IRE1α/XBP1 endoplasmic reticulum stress response pathway in ARID1A-mutant ovarian cancers. Cancer Res. 81:5325–5335. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Song M, Sandoval TA, Chae CS, Chopra S, Tan C, Rutkowski MR, Raundhal M, Chaurio RA, Payne KK, Konrad C, et al: IRE1α-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity. Nature. 562:423–428. 2018. View Article : Google Scholar : PubMed/NCBI

144 

Dong YF, Zhang J, Zhou JH, Xiao YL, Pei WJ and Liu HP: Mitochondrial-associated endoplasmic reticulum membrane interference in ovarian cancer (Review). Oncol Rep. 52:1122024. View Article : Google Scholar

145 

Cubillos-Ruiz JR, Bettigole SE and Glimcher LH: Molecular pathways: Immunosuppressive roles of IRE1α-XBP1 signaling in dendritic cells of the tumor microenvironment. Clin Cancer Res. 22:2121–2126. 2016. View Article : Google Scholar : PubMed/NCBI

146 

Lin J, Liu H, Fukumoto T, Zundell J, Yan Q, Tang CA, Wu S, Zhou W, Guo D, Karakashev S, et al: Targeting the IRE1α/XBP1s pathway suppresses CARM1-expressing ovarian cancer. Nat Commun. 12:53212021. View Article : Google Scholar

147 

Harding HP, Zhang Y and Ron D: Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature. 397:271–274. 1999. View Article : Google Scholar : PubMed/NCBI

148 

Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M and Ron D: Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell. 6:1099–1108. 2000. View Article : Google Scholar : PubMed/NCBI

149 

Jackson RJ, Hellen CUT and Pestova TV: The mechanism of eukaryotic translation initiation and principles of its regulation. Nat Rev Mol Cell Biol. 11:113–127. 2010. View Article : Google Scholar : PubMed/NCBI

150 

Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, Saunders T, Bonner-Weir S and Kaufman RJ: Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell. 7:1165–1176. 2001. View Article : Google Scholar : PubMed/NCBI

151 

Giampietri C, Petrungaro S, Conti S, Facchiano A, Filippini A and Ziparo E: Cancer microenvironment and endoplasmic reticulum stress response. Mediators Inflamm. 2015:4172812015. View Article : Google Scholar : PubMed/NCBI

152 

Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, Sadri N, Yun C, Popko B, Paules R, et al: An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 11:619–633. 2003. View Article : Google Scholar : PubMed/NCBI

153 

Cullinan SB, Zhang D, Hannink M, Arvisais E, Kaufman RJ and Diehl JA: Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival. Mol Cell Biol. 23:7198–7209. 2003. View Article : Google Scholar : PubMed/NCBI

154 

Dan W, Fan Y, Wang Y, Hou T, Wei Y, Liu B, Li M, Chen J, Fang Q, Que T, et al: The tumor suppressor TPD52-governed endoplasmic reticulum stress is modulated by APCCdc20. Adv Sci (Weinh). 11:e24054412024. View Article : Google Scholar

155 

Wiseman RL, Mesgarzadeh JS and Hendershot LM: Reshaping endoplasmic reticulum quality control through the unfolded protein response. Mol Cell. 82:1477–1491. 2022. View Article : Google Scholar : PubMed/NCBI

156 

Park SJ, Yoon BH, Kim SK and Kim SY: GENT2: An updated gene expression database for normal and tumor tissues. BMC Med Genomics. 12(Suppl 5): S1012019. View Article : Google Scholar

157 

Cai Y, Arikkath J, Yang L, Guo ML, Periyasamy P and Buch S: Interplay of endoplasmic reticulum stress and autophagy in neurodegenerative disorders. Autophagy. 12:225–244. 2016. View Article : Google Scholar : PubMed/NCBI

158 

Meng J, Liu K, Shao Y, Feng X, Ji Z, Chang B, Wang Y, Xu L and Yang G: ID1 confers cancer cell chemoresistance through STAT3/ATF6-mediated induction of autophagy. Cell Death Dis. 11:1372020. View Article : Google Scholar : PubMed/NCBI

159 

Qiu L, Liu H, Chen S, Wu Y and Yan J: Ferroptosis contributed to endoplasmic reticulum stress in preterm birth by targeting LHX1 and IRE-1. Cell Signal. 132:1117772025. View Article : Google Scholar : PubMed/NCBI

160 

Song T, Li J, Xia Y, Hou S, Zhang X and Wang Y: 1,25-D3 ameliorates ischemic brain injury by alleviating endoplasmic reticulum stress and ferroptosis: Involvement of vitamin D receptor and p53 signaling. Cell Signal. 122:1113312024. View Article : Google Scholar : PubMed/NCBI

161 

Zeng T, Zhou Y, Yu Y, Wang JW, Wu Y, Wang X, Zhu L, Zhou LM and Wan LH: rmMANF prevents sepsis-associated lung injury via inhibiting endoplasmic reticulum stressinduced ferroptosis in mice. Int Immunopharmacol. 114:1096082023. View Article : Google Scholar

162 

Chen Y, Li H, Liu J, Ni J, Deng Q, He H, Wu P, Wan Y, Seeram NP, Liu C, et al: Cytotoxicity of natural and synthetic cannabinoids and their synergistic antiproliferative effects with cisplatin in human ovarian cancer cells. Front Pharmacol. 15:14961312024. View Article : Google Scholar : PubMed/NCBI

163 

Wang Z, Liu M, Li GX, Zhang L, Ding KY, Li SQ, Gao BQ, Chen P, Choe HC, Xia LY, et al: A herbal pair of Scutellaria barbata D. Don and Scleromitrion diffusum (Willd.) R.J. Wang induced ferroptosis in ovarian cancer A2780 cells via inducing heme catabolism and ferritinophagy. J Integr Med. 22:665–682. 2024. View Article : Google Scholar : PubMed/NCBI

164 

Liu Y, Li J, Xu J, Long Y, Wang Y, Liu X, Hu J, Wei Q, Luo Q, Luo F, et al: m6A-driven NAT10 translation facilitates fatty acid metabolic rewiring to suppress ferroptosis and promote ovarian tumorigenesis through enhancing ACOT7 mRNA acetylation. Oncogene. 43:3498–3516. 2024. View Article : Google Scholar : PubMed/NCBI

165 

Furutake Y, Yamaguchi K, Yamanoi K, Kitamura S, Takamatsu S, Taki M, Ukita M, Hosoe Y, Murakami R, Abiko K, et al: YAP1 Suppression by ZDHHC7 is associated with ferroptosis resistance and poor prognosis in ovarian clear cell carcinoma. Mol Cancer Ther. 23:1652–1665. 2024. View Article : Google Scholar : PubMed/NCBI

166 

Han Y, Fu L, Kong Y, Jiang C, Huang L and Zhang H: STEAP3 affects ovarian cancer progression by regulating ferroptosis through the p53/SLC7A11 pathway. Mediators Inflamm. 2024:40485272024. View Article : Google Scholar : PubMed/NCBI

167 

Nevins S, McLoughlin CD, Oliveros A, Stein JB, Rashid MA, Hou Y, Jang MH and Lee KB: Nanotechnology approaches for prevention and treatment of chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy in breast and ovarian cancer survivors. Small. 20:e23007442024.

168 

Luo L, Zhou H, Wang S, Pang M, Zhang J, Hu Y and You J: The application of nanoparticle-based imaging and phototherapy for female reproductive organs diseases. Small. 20:e22076942024.

169 

Lee J, Jang S, Im J, Han Y, Kim S, Jo H, Wang W, Cho U, Kim SI, Seol A, et al: Stearoyl-CoA desaturase 1 inhibition induces ER stress-mediated apoptosis in ovarian cancer cells. J Ovarian Res. 17:732024. View Article : Google Scholar : PubMed/NCBI

170 

Yan T, Ma X, Zhou K, Cao J, Tian Y, Zheng H, Tong Y, Xie S, Wang Y, Guo L and Lu R: A novel CSN5/CRT O-GlcNAc/ER stress regulatory axis in platinum resistance of epithelial ovarian cancer. Int J Biol Sci. 20:1279–1296. 2024. View Article : Google Scholar : PubMed/NCBI

171 

Kim TW and Lee HG: 6-Shogaol overcomes gefitinib resistance via ER stress in ovarian cancer cells. Int J Mol Sci. 24:26392023. View Article : Google Scholar : PubMed/NCBI

172 

Zhang M and Wang Y, Xu S, Huang S, Wu M, Chen G and Wang Y: Endoplasmic reticulum stress-related ten-biomarker risk classifier for survival evaluation in epithelial ovarian cancer and TRPM2: A potential therapeutic target of ovarian cancer. Int J Mol Sci. 24:140102023. View Article : Google Scholar : PubMed/NCBI

173 

Ma X, Zhou W, Zhang R, Zhang C, Yan J, Feng J, Rosenholm JM, Shi T, Shen X and Zhang H: Minimally invasive injection of biomimetic Nano@Microgel for in situ ovarian cancer treatment through enhanced photodynamic reactions and photothermal combined therapy. Mater Today Bio. 20:1006632023. View Article : Google Scholar : PubMed/NCBI

174 

Zhang J, Guo J, Yang N, Huang Y, Hu T and Rao C: Endoplasmic reticulum stress-mediated cell death in liver injury. Cell Death Dis. 13:10512022. View Article : Google Scholar : PubMed/NCBI

175 

Cheng C, Yuan Y, Yuan F and Li X: Acute kidney injury: Exploring endoplasmic reticulum stress-mediated cell death. Front Pharmacol. 15:13087332024. View Article : Google Scholar : PubMed/NCBI

176 

Yang J, Xu H, Wu W, Huang H, Zhang C, Tang W, Tang Q and Bi F: Ferroptosis signaling promotes the release of misfolded proteins via exosomes to rescue ER stress in hepatocellular carcinoma. Free Radic Biol Med. 202:110–120. 2023. View Article : Google Scholar : PubMed/NCBI

177 

Li FJ, Abudureyimu M, Zhang ZH, Tao J, Ceylan AF, Lin J, Yu W, Reiter RJ, Ashrafizadeh M, Guo J and Ren J: Inhibition of ER stress using tauroursodeoxycholic acid rescues obesity-evoked cardiac remodeling and contractile anomalies through regulation of ferroptosis. Chem Biol Interact. 398:1111042024. View Article : Google Scholar : PubMed/NCBI

178 

Han N, Yang ZY, Xie ZX, Xu HZ, Yu TT, Li QR, Li LG, Peng XC, Yang XX, Hu J, et al: Dihydroartemisinin elicits immunogenic death through ferroptosis-triggered ER stress and DNA damage for lung cancer immunotherapy. Phytomedicine. 112:1546822023. View Article : Google Scholar : PubMed/NCBI

179 

Han Y, Hao G, Han S, Zhu T, Dong Y, Chen L, Yang X, Li X, Jin H, Liang G, et al: Polydatin ameliorates early brain injury after subarachnoid hemorrhage through up-regulating SIRT1 to suppress endoplasmic reticulum stress. Front Pharmacol. 15:14502382024. View Article : Google Scholar : PubMed/NCBI

180 

Wang S, Xu F, Liu H, Shen Y, Zhang J, Hu L and Zhu L: Suppressing endoplasmic reticulum stress alleviates LPS-induced acute lung injury via inhibiting inflammation and ferroptosis. Inflammation. 47:1067–1082. 2024. View Article : Google Scholar : PubMed/NCBI

181 

Ding H, Xiang Y, Zhu Q, Wu H, Xu T, Huang Z and Ge H: Endoplasmic reticulum stress-mediated ferroptosis in granulosa cells contributes to follicular dysfunction of polycystic ovary syndrome driven by hyperandrogenism. Reprod Biomed Online. 49:1040782024. View Article : Google Scholar : PubMed/NCBI

182 

Jiang D, Guo Y, Wang T, Wang L, Yan Y, Xia L, Bam R, Yang Z, Lee H, Iwawaki T, et al: IRE1α determines ferroptosis sensitivity through regulation of glutathione synthesis. Nat Commun. 15:41142024. View Article : Google Scholar

183 

Guan Q, Wang Z, Hu K, Cao J, Dong Y and Chen Y: Melatonin ameliorates hepatic ferroptosis in NAFLD by inhibiting ER stress via the MT2/cAMP/PKA/IRE1 signaling pathway. Int J Biol Sci. 19:3937–3950. 2023. View Article : Google Scholar : PubMed/NCBI

184 

He Z, Shen P, Feng L, Hao H, He Y, Fan G, Liu Z, Zhu K, Wang Y, Zhang N, et al: Cadmium induces liver dysfunction and ferroptosis through the endoplasmic stress-ferritinophagy axis. Ecotoxicol Environ Saf. 245:1141232022. View Article : Google Scholar : PubMed/NCBI

185 

Zhao C, Yu D, He Z, Bao L, Feng L, Chen L, Liu Z, Hu X, Zhang N, Wang T and Fu Y: Endoplasmic reticulum stress-mediated autophagy activation is involved in cadmium-induced ferroptosis of renal tubular epithelial cells. Free Radic Biol Med. 175:236–248. 2021. View Article : Google Scholar : PubMed/NCBI

186 

Xu M, Tao J, Yang Y, Tan S, Liu H, Jiang J, Zheng F and Wu B: Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis. Cell Death Dis. 11:862020. View Article : Google Scholar : PubMed/NCBI

187 

Thulasinathan B, Suvilesh KN, Maram S, Grossmann E, Ghouri Y, Teixeiro EP, Chan J, Kaif JT and Rachagani S: The impact of gut microbial short-chain fatty acids on colorectal cancer development and prevention. Gut Microbes. 17:24837802025. View Article : Google Scholar : PubMed/NCBI

188 

Wei R, Zhao Y, Wang J, Yang X, Li S, Wang Y, Yang X, Fei J, Hao X, Zhao Y, et al: Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. Int J Biol Sci. 17:2703–2717. 2021. View Article : Google Scholar : PubMed/NCBI

189 

Zhao R, Lv Y, Feng T, Zhang R, Ge L, Pan J, Han B, Song G and Wang L: ATF6α promotes prostate cancer progression by enhancing PLA2G4A-mediated arachidonic acid metabolism and protecting tumor cells against ferroptosis. Prostate. 82:617–629. 2022. View Article : Google Scholar : PubMed/NCBI

190 

Tak J, Kim YS, Kim TH, Park GC, Hwang S and Kim SG: Gα12 overexpression in hepatocytes by ER stress exacerbates acute liver injury via ROCK1-mediated miR-15a and ALOX12 dysregulation. Theranostics. 12:1570–1588. 2022. View Article : Google Scholar :

191 

Ji X, Chen Z, Lin W, Wu Q, Wu Y, Hong Y, Tong H, Wang C and Zhang Y: Esculin induces endoplasmic reticulum stress and drives apoptosis and ferroptosis in colorectal cancer via PERK regulating eIF2α/CHOP and Nrf2/HO-1 cascades. J Ethnopharmacol. 328:1181392024. View Article : Google Scholar

192 

Guo C, Zhao W, Wang W, Yao Z, Chen W and Feng X: Study on the antitumor mechanism of tanshinone IIA in vivo and in vitro through the regulation of PERK-ATF4-HSPA5 pathway-mediated ferroptosis. Molecules. 29:15572024. View Article : Google Scholar : PubMed/NCBI

193 

Xu J, Zhao L, Zhang X, Ying K, Zhou R, Cai W, Wu X, Jiang H, Xu Q, Miao D, et al: Salidroside ameliorates acetaminophen-induced acute liver injury through the inhibition of endoplasmic reticulum stress-mediated ferroptosis by activating the AMPK/SIRT1 pathway. Ecotoxicol Environ Saf. 262:1153312023.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

194 

Chen Y, Mi Y, Zhang X, Ma Q, Song Y, Zhang L, Wang D, Xing J, Hou B, Li H, et al: Dihydroartemisinin-induced unfolded protein response feedback attenuates ferroptosis via PERK/ATF4/HSPA5 pathway in glioma cells. J Exp Clin Cancer Res. 38:4022019. View Article : Google Scholar : PubMed/NCBI

195 

Kuang H, Sun X, Liu Y, Tang M, Wei Y, Shi Y, Li R, Xiao G, Kang J, Wang F, et al: Palmitic acid-induced ferroptosis via CD36 activates ER stress to break calcium-iron balance in colon cancer cells. FEBS J. 290:3664–3687. 2023. View Article : Google Scholar : PubMed/NCBI

196 

Fu F, Wang W, Wu L, Wang W, Huang Z, Huang Y, Wu C and Pan X: Inhalable biomineralized liposomes for cyclic Ca2+-burst-centered endoplasmic reticulum stress enhanced lung cancer ferroptosis therapy. ACS Nano. 17:5486–5502. 2023. View Article : Google Scholar : PubMed/NCBI

197 

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

198 

Li Y, Gao Y, Zhang X, Guo H and Gao H: Nanoparticles in precision medicine for ovarian cancer: From chemotherapy to immunotherapy. Int J Pharm. 591:1199862020. View Article : Google Scholar : PubMed/NCBI

199 

Wu M and Zhou S: Harnessing tumor immunogenomics: Tumor neoantigens in ovarian cancer and beyond. Biochim Biophys Acta Rev Cancer. 1878:1890172023. View Article : Google Scholar : PubMed/NCBI

200 

Dahlmanns M, Yakubov E and Dahlmanns JK: Genetic profiles of ferroptosis in malignant brain tumors and off-target effects of ferroptosis induction. Front Oncol. 11:7830672021. View Article : Google Scholar : PubMed/NCBI

201 

He Y, Su J, Lan B, Gao Y and Zhao J: Targeting off-target effects: Endoplasmic reticulum stress and autophagy as effective strategies to enhance temozolomide treatment. Onco Targets Ther. 12:1857–1865. 2019. View Article : Google Scholar : PubMed/NCBI

202 

Sassetti E, Clausen MH and Laraia L: Small-molecule inhibitors of reactive oxygen species production. J Med Chem. 64:5252–5275. 2021. View Article : Google Scholar : PubMed/NCBI

203 

Zhu YS, Wu J and Zhi F: Advances in conjugate drug delivery system: Opportunities and challenges. Int J Pharm. 667:1248672024. View Article : Google Scholar : PubMed/NCBI

204 

Agrawal SS, Baliga V and Londhe VY: Liposomal formulations: A recent update. Pharmaceutics. 17:362024. View Article : Google Scholar

205 

Liu Z, Xiang C, Zhao X, Aizawa T, Niu R, Zhao J, Guo F, Li Y, Luo W, Liu W and Gu R: Regulation of dynamic spatiotemporal inflammation by nanomaterials in spinal cord injury. J Nanobiotechnology. 22:7672024. View Article : Google Scholar : PubMed/NCBI

206 

Tang S and Chen L: The recent advancements of ferroptosis of gynecological cancer. Cancer Cell Int. 24:3512024. View Article : Google Scholar : PubMed/NCBI

207 

Ying Y, Zhang J, Ren D, Zhao P, Zhang W and Lu X: ERP29 regulates the proliferation of endometrial carcinoma via M6A modification. Life Sci. 354:1229762024. View Article : Google Scholar : PubMed/NCBI

208 

Zhao Y, Lu L, Chen X and Yin Q: Natural compounds targeting ferroptosis in ovarian cancer: Research progress and application potential. Pharmacol Res. 215:1077292025. View Article : Google Scholar : PubMed/NCBI

209 

Jin N, Qian YY, Jiao XF, Wang Z, Li X, Pan W, Jiang JK, Huang P, Wang SY, Jin P, et al: Niraparib restricts intraperitoneal metastases of ovarian cancer by eliciting CD36-dependent ferroptosis. Redox Biol. 80:1035282025. View Article : Google Scholar : PubMed/NCBI

210 

Miglietta S, Sollazzo M, Gherardi I, Milioni S, Cavina B, Marchio L, De Luise M, Coada CA, Fiorillo M, Perrone AM, et al: Mitochondrial chaperonin DNAJC15 promotes vulnerability to ferroptosis of chemoresistant ovarian cancer cells. Open Biol. 15:2401512025. View Article : Google Scholar : PubMed/NCBI

211 

Yin ZY, He SM, Zhang XY, Yu XC, Sheng KX, Fu T, Jiang YX, Xu L, Hu BX, Zhang JB, et al: Apolipoprotein B100 acts as a tumor suppressor in ovarian cancer via lipid/ER stress axis-induced blockade of autophagy. Acta Pharmacol Sin. Jan 29–2025.Epub ahead of print. View Article : Google Scholar

212 

Li D, Geng D and Wang M: Advances in natural products modulating autophagy influenced by cellular stress conditions and their anticancer roles in the treatment of ovarian cancer. FASEB J. 38:e700752024. View Article : Google Scholar : PubMed/NCBI

213 

Chan DW, Yung MM, Chan YS, Xuan Y, Yang H, Xu D, Zhan JB, Chan KK, Ng TB and Ngan HY: MAP30 protein from Momordica charantia is therapeutic and has synergic activity with cisplatin against ovarian cancer in vivo by altering metabolism and inducing ferroptosis. Pharmacol Res. 161:1051572020. View Article : Google Scholar : PubMed/NCBI

214 

Zhang GH, Kai JY, Chen MM, Ma Q, Zhong AL, Xie SH, Zheng H, Wang YC, Tong Y, Tian Y, et al: Downregulation of XBP1 decreases serous ovarian cancer cell viability and enhances sensitivity to oxidative stress by increasing intracellular ROS levels. Oncol Lett. 18:4194–4202. 2019.PubMed/NCBI

215 

Janczar S, Nautiyal J, Xiao Y, Curry E, Sun M, Zanini E, Paige AJ and Gabra H: WWOX sensitises ovarian cancer cells to paclitaxel via modulation of the ER stress response. Cell Death Dis. 8:e29552017. View Article : Google Scholar : PubMed/NCBI

216 

Chen Q, Wang Y, Yue T, Wei H, Li S and Dong B: Fluorescence imaging of intracellular glutathione levels in the endoplasmic reticulum to reveal the inhibition effect of rutin on ferroptosis. Anal Chem. January 9–2023.Epub ahead of print.

217 

Huang M, Wang Y, Wu X and Li W: Crosstalk between endoplasmic reticulum stress and ferroptosis in liver diseases. Front Biosci (Landmark Ed). 29:2212024. View Article : Google Scholar : PubMed/NCBI

218 

Ao Q, Hu H and Huang Y: Ferroptosis and endoplasmic reticulum stress in rheumatoid arthritis. Front Immunol. 15:14388032024. View Article : Google Scholar : PubMed/NCBI

219 

Li Y, Li M, Feng S, Xu Q, Zhang X, Xiong X and Gu L: Ferroptosis and endoplasmic reticulum stress in ischemic stroke. Neural Regen Res. 19:611–618. 2024. View Article : Google Scholar

220 

Zhang YG, Yan XF, Liu F, Hao WZ, Cai Y, Liu Y, Liu LL and Li XJ: Astragalus polysaccharides induces ferroptosis in ovarian adenocarcinoma cells through Nrf2/SLC7A11/GPX4 signaling pathway. Zhongguo Zhong Yao Za Zhi. 49:6459–6467. 2024.In Chinese.

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Yang J, Wang Y, Liu F, Zhang Y and Han F: Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review). Int J Mol Med 55: 97, 2025.
APA
Yang, J., Wang, Y., Liu, F., Zhang, Y., & Han, F. (2025). Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review). International Journal of Molecular Medicine, 55, 97. https://doi.org/10.3892/ijmm.2025.5538
MLA
Yang, J., Wang, Y., Liu, F., Zhang, Y., Han, F."Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review)". International Journal of Molecular Medicine 55.6 (2025): 97.
Chicago
Yang, J., Wang, Y., Liu, F., Zhang, Y., Han, F."Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review)". International Journal of Molecular Medicine 55, no. 6 (2025): 97. https://doi.org/10.3892/ijmm.2025.5538
Copy and paste a formatted citation
x
Spandidos Publications style
Yang J, Wang Y, Liu F, Zhang Y and Han F: Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review). Int J Mol Med 55: 97, 2025.
APA
Yang, J., Wang, Y., Liu, F., Zhang, Y., & Han, F. (2025). Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review). International Journal of Molecular Medicine, 55, 97. https://doi.org/10.3892/ijmm.2025.5538
MLA
Yang, J., Wang, Y., Liu, F., Zhang, Y., Han, F."Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review)". International Journal of Molecular Medicine 55.6 (2025): 97.
Chicago
Yang, J., Wang, Y., Liu, F., Zhang, Y., Han, F."Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review)". International Journal of Molecular Medicine 55, no. 6 (2025): 97. https://doi.org/10.3892/ijmm.2025.5538
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team