International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Programmed cell death (PCD) is defined as an intrinsic component of physiological developmental programs or tissue renewal, occurring independently of exogenous environmental factors (1). Importantly, accumulating evidence indicates that PCD has strong effects on a number of lesions including chronic inflammation and cancer (2-4). Ferroptosis, a term introduced by Brent Stockwell in 2012, is a distinct form of iron-dependent PCD characterized by the accumulation of lipid peroxides and subsequent disruption of the cell membrane, ultimately leading to cell death (5). Recent advances in molecular biology have revealed the critical involvement of ferroptosis in the pathophysiology of gastrointestinal diseases, establishing it as a rapidly evolving research frontier in gastroenterology (6).
According to the global cancer statistics in 2022 released by GLOBOCAN, gastric cancer (GC) ranks fifth in both incidence and mortality worldwide, with >950,000 new cases and >650,000 mortalities in the whole year (7). Helicobacter pylori (H. pylori) infection, the most prevalent chronic bacterial infection worldwide, is a major etiological factor for GC (8). Studies have demonstrated that H. pylori mediates chronic inflammation by upregulating proinflammatory cytokines, including interleukin (IL)-8, IL-1β and tumor necrosis factor (TNF) (9-11). H. pylori-associated chronic gastritis, also referred to as type B gastritis, can progress to GC through a sequence of histopathological changes, a process first described in 1975 and termed as Correa's cascade (12). Despite significant advancements in the prevention and treatment of GC based on Correa's cascade, several critical challenges persist. In the precancerous lesions of GC (PLGC), traditional therapies such as H. pylori eradication and symptomatic treatment often fail to effectively suppress chronic inflammation or halt the progression of Correa's cascade (13,14). Moreover, the early detection of GC is substantially hampered by the absence of specific clinical manifestations in the initial stages, the suboptimal sensitivity of current screening biomarkers, and the low popularity rate of endoscopic screening (15,16). Consequently, a substantial proportion of patients are diagnosed at advanced stages of disease progression. In the management of advanced GC, several therapeutic limitations remain unresolved, including the development of chemoresistance, the paucity of novel molecular targets for targeted therapies, and the low response rate of immunotherapy, all of which represent pressing unmet needs in contemporary oncology practice (17-20). Targeted regulation of ferroptosis-related strategies, characterized by precision targeting, antioxidant properties, anti-inflammatory effects and potential anticancer activity, represents a promising approach to overcoming these limitations.
This comprehensive review systematically investigated the intricate interplay between H. pylori infection and ferroptosis, while proposing novel promising strategies for GC prevention and treatment through targeted regulation of ferroptosis-related pathways across several different stages of the Correa's cascade.
Oxygen serves as the terminal electron acceptor in the majority of metabolic oxidation-reduction reactions for most organisms, highlighting the necessity of oxidative stress. Oxidative stress induces oxidative modifications of the cell's bilayer membrane, particularly lipid oxidation, which impacts various cellular physiological processes such as developmental regulation, immune response, tumor suppression, metabolic balance and aging. Ferroptosis, a form of PCD, is characterized by extensive lipid peroxidation (21). Since its introduction in 2012, ferroptosis research has centered around several fundamental components: i) The systemic xc−-glutathione (GSH)-glutathione peroxidase (GPX)4 ferroptosis suppression pathway; ii) phospholipid hydroperoxides (PLOOHs); iii) iron regulation; iv) GPX4-independent regulatory pathways; and v) other important regulators such as tumor suppressor p53 and related signaling pathways.
GSH is a crucial intracellular reductant, and also functions as a cofactor for enzymes such as GPXs and GSH-S-transferases. GSH biosynthesis relies on cysteine, which can be imported from the environment via neutral amino acid transporters, taken up in its oxidized form (cystine) through the xc−-cystine/glutamate antiporter system [comprising solute carrier family 7 member 11 and solute carrier family 3 member 2 (SLC3A2 subunits)], or synthesized through the trans-sulfuration pathway utilizing methionine and glucose (21-23). The transporter protein in the xc− system is a disulfide-linked heterodimer consisting of a light chain (xCT) and a heavy chain (4F2hc) (24). GPX4 plays a pivotal role in the ferroptosis process, serving as the primary enzyme that catalyzes the reduction and detoxification of PLOOHs in mammalian cells (25).
As a type of lipid-derived reactive oxygen species (ROS), PLOOHs mark the beginning of lipid peroxidation. This process begins with the abstraction of a bisallylic hydrogen atom from the polyunsaturated fatty acid (PUFA) acyl chain of phospholipids within the lipid bilayer, generating a carbon-centered phospholipid radical (PL•). This radical subsequently reacts with molecular oxygen to form a phospholipid peroxyl radical (PLOO•) (26), which abstracts hydrogen from another PUFA, resulting in PLOOH formation. In the absence of timely reduction of PLOOHs to their corresponding alcohols (PLOH) by GPX4, the chain reaction products, including lipid peroxide breakdown products [e.g., 4-hydroxynonenal (4-HNE) and malondialdehyde] and oxidized/modified proteins, disrupt membrane integrity and ultimately lead to organelle and/or membrane breakdown (21).
The regulation of ferroptosis has emerged as a critical focus in disease mechanism research. In the context of ferroptosis modulation, two distinct mechanisms have been identified: Erastin exerts its effect through indirect inhibition of GPX4 by targeting system xc, thereby disrupting cystine uptake, while RSL3 demonstrates direct GPX4 inhibition. These compounds represent two fundamental classes of ferroptosis inducers, as indicated in previous studies (27,28). Furthermore, PUFAs and PUFA-containing lipids within biofilms are susceptible to direct oxidation by certain lipoxygenases (LOXs), suggesting that LOXs may also constitute a target for ferroptosis induction (29). Additionally, iron is also crucial for the regulation of ferroptosis. Inhibition of GPX4 triggers the Fenton reaction, leading to a rapid accumulation of PLOOHs, a characteristic of iron toxicity (26). Moreover, it has been shown that cytochrome P450 oxidoreductase can directly or indirectly trigger lipid peroxidation by removing hydrogen from PUFAs or by reducing ferric iron (Fe3+) to its ferrous form (Fe2+) after its downstream electron acceptor is reduced (30).
Apart from the GSH-GPX4 inhibitory pathway, which is recognized as the predominant ferroptosis regulatory system (31), one such mechanism involves ferroptosis suppressor protein 1 (FSP1, also known as AIFM2), which inhibits lipid peroxidation and ferroptosis by synthesizing panthenol and rejuvenating oxidized α-tocopherol radicals (vitamin E) (32,33). Another mechanism entails guanosine triphosphate cyclohydrolase 1 protecting against ferroptosis through its metabolites tetrahydrobiopterin and dihydrobiopterin (34).
There is compelling evidence linking ferroptosis to a spectrum of pathologies involving tissue damage, encompassing cancer, neurodegeneration, inflammation and infection (35). Targeting ferroptosis mostly may offer a therapeutic avenue for related disorders. However, a number of cancer cells exhibit heightened vulnerability to ferroptosis, suggesting its potential as an anticancer strategy. Ferroptosis has been closely implicated in several cancer-associated signaling pathways. A study on the interplay between energy stress and ferroptosis has revealed that energy stress can inhibit ferroptosis through AMP-activated protein kinase pathway (36). Furthermore, lactate produced by cancer cells under energy stress may inhibit ferroapoptosis of tumor cells and promote their metastatic spread (37). The phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) signaling axis has also been shown to shield cancer cells from oxidative stress and ferroptosis through sterol regulatory element-binding protein 1/stearoyl-CoA desaturase 1-mediated lipid synthesis (38).
In addition, the regulation of ferroptosis is also related to cell density (39). Specifically, E-cadherin, a key regulator of epithelial cell-cell adhesion, is positively associated with cell density and functions upstream to repress Yes-associated protein activity in the nucleus (40,41). Components of the Hippo pathway, directly involved in this regulatory network, are frequently mutated in cancer. Secondly, a variety of tumor suppressants, including p53, have been shown to increase the sensitivity to ferroptosis. Specifically, p53 has been shown to enhance ferroptosis by inhibiting the transcription of the systemic xc−-subunit SLC7A11 (42). Conversely, p53 has also been reported to inhibit ferroptosis in cancer cells under cysteine deprivation by regulating the transcriptional target cyclin-dependent kinase inhibitor 1A and limiting erastin-induced ferroptosis by blocking dipeptidyl peptidase-4 activity in a transcription-independent manner (43,44). Therefore, the interaction between these tumor suppressors and ferroptosis appears to be influenced by a multitude of complex factors. Finally, some chemotherapeutic agents and targeted agents, such as cisplatin and sorafenib, have previously been shown to be able to achieve antitumor effects by inducing ferroptosis (45,46).
At present, cancer management research targeting ferroptosis has achieved breakthroughs in a number of aspects (Fig. 1). First, the identification of ferroptosis-related biomarkers is one of the promising strategies in cancer management (47,48). Secondly, in addition to being transformed in chemoradiotherapy and immunotherapy, therapeutic strategies to induce ferroptosis have also been applied in the emerging field of nanotherapy (49). Additionally, some external factors that can lead to the dysregulation of ferroptosis are also worthy of attention. It has been documented that some infectious pathogens such as H. pylori can cause dysregulation of ferroptosis (50). Furthermore, N-3 PUFA peroxidation has been shown to selectively induce ferroptosis in cancer cells. Consequently, N-3 long-chain PUFA-rich foods may be a dietary strategy for patients with cancer (51). In general, targeting ferroptosis is a promising strategy for cancer treatment in the future.
Autophagy and apoptosis represent the most prominent forms of PCD in GC, with their dysregulation being closely linked to specific virulence factors of H. pylori infection (52,53). Vacuolar cell toxin (VacA), a key virulence factor, induces autophagic cell death through endoplasmic reticulum (ER) stress in gastric epithelial cells (54), while simultaneously promoting apoptosis via the p38/MAPK pathway-mediated downregulation of TNF receptor-associated protein 1 (55). Another critical virulence determinant, cytotoxin-associated gene A (CagA), triggers mitochondrial membrane depolarization by elevating hydrogen peroxide (H2O2) levels through spermine oxidase activation, subsequently initiating caspase-dependent apoptosis (56). The oxidative stress induced by H. pylori infection, characterized by excessive production of ROS and reactive nitrogen species (RNS) from neutrophils, leads to DNA damage (57,58), which may be mitigated through apoptosis induction to prevent oncogenic mutations (59). The mitochondrial apoptotic pathway is further regulated by H. pylori through modulation of the B-cell lymphoma-2 (Bcl-2)-associated X/Bcl-2 ratio by outer inflammatory protein A and VacA (60,61).
Notably, H. pylori exhibits bidirectional regulation of apoptosis, as evidenced by its ability to attenuate caspase-8-dependent apoptosis through the type IV secretion system-mediated formation of pAbIT735 (62). Beyond autophagy and apoptosis, emerging evidence implicates other PCD pathways, including pyroptosis, necroptosis and ferroptosis, in H. pylori-associated pathogenesis (Fig. 2) (63-65).
Accumulating evidence indicates that bacterial infection can promote ferroptosis following tissue damage. Mycobacterium tuberculosis (Mtb), for instance, secretes protein tyrosine phosphatase A, which inhibits GPX4 expression, thereby inducing ferroptosis and enhancing Mtb pathogenicity and transmission (66). Similarly, Pseudomonas aeruginosa utilizes host polyunsaturated phosphatidylethanolamine to induce lipid peroxidation and ferroptosis in bronchial epithelial cells (67). H. pylori infection elicits a robust inflammatory response in the gastric mucosa, leading to the generation of ROS and RNS, which in turn facilitates lipid peroxidation (68). In H. pylori infection, the release of virulence factors also affects ferroptosis (Fig. 3). Inhibition of GPX4 by RSL3 renders cells unable to eliminate accumulated lipid hydroperoxides, ultimately leading to ferroptosis. A study has corroborated that phosphorylase kinase G2 promotes RSL3-induced ferroptosis in GC cells by enhancing arachidonate 5-lipoxygenase expression in CagA-positive H. pylori infections, but the mechanism of action of CagA in this process requires further investigation (50). Another study clarified that CagA could promote the synthesis of polyunsaturated ether phospholipids through the MEK/ERK/serum response factor pathway, leading to the susceptibility to ferroptosis (69). An investigation into the iron toxicity-associated gene tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon (YWHAE) demonstrated significantly elevated YWHAE expression levels in H. pylori-induced GC, which was positively correlated with ferroptosis in GC (70).
Beyond CagA and YWHA, H. pylori outer membrane vesicles (OMVs) also contribute to aberrant ferroptosis regulation. H. pylori and its OMVs modulate ferroptosis through three primary mechanisms: i) Reducing cellular iron uptake and lipid peroxide production by downregulating transferrin receptor/transferrin receptor protein 1 (TFRC/TFR1) and the endosomal metal reductase six-transmembrane epithelial antigen of the prostate 3; ii) upregulating expression of the cystine/glutamate antiporter subunit SLC3A2 and GSH synthesis genes; and iii) inhibiting ferroptosis by decreasing substrate availability for arachidonic acid-associated lipid peroxidation through downregulation of lysophosphatidylcholine acyltransferase 3 (71). Concurrently, H. pylori can epigenetically influence cell ferroptosis, such as by triggering demethylation and upregulating glucocerebrosidase, thereby inhibiting ferroptosis in GC cells (72).
Notably, ferroptosis interacts with other types of PCD in the process of GC induced by H. pylori infection. Ferroptosis-driven lipid peroxidation activates pro-apoptotic signals, while apoptosis-related proteins (e.g., caspases) may conversely enhance ferroptosis by degrading inhibitors such as GPX4 (42,73). Autophagy further promotes ferroptosis sensitivity through ferritin degradation (releasing free iron) or depletion of antioxidants such as GPX4 (74,75). Additionally, ferroptosis-induced oxidative stress in the GC microenvironment activates the nucleotide-binding oligomerization domain-like receptor protein 3 inflammasome, triggering IL-1β release and pyroptosis (76). These findings indicate that H. pylori infection induces dysregulation of multiple PCD pathways, which functionally interact during tumorigenesis. Critically, H. pylori-driven PCD dysregulation-including aberrant ferroptosis-significantly accelerates Correa's cascade of GC, underscoring H. pylori eradication as a foundational strategy for GC prevention (77). Nevertheless, chronic inflammatory responses and pathological progression frequently persist during intermediate to advanced precancerous stages despite successful H. pylori eradication therapy (78,79). This persistent pathological progression highlights the potential therapeutic value of strategies targeting various types of PCD, including ferroptosis, as adjunctive interventions to complement conventional eradication therapy in PLGC.
Studies on the regulation of ferroptosis not only provide a new perspective on the pathogenesis of H. pylori but also provide a direction for exploring new therapeutic targets in Correa's cascade. The systematic review of ferroptosis in the three key stages of chronic atrophic gastritis (CAG), intestinal metaplasia (IM) and GC is conducive to bringing new breakthroughs in the prevention and treatment of GC.
CAG is the initial stage in the 'inflammation-cancer transformation model'. Knowing how to treat CAG timely and accurately, and block or reverse the development of CAG to GC is crucial for the prevention of GC. Conventional therapeutic approaches for CAG primarily encompass H. pylori eradication therapy, gastric mucosal protection and gastrointestinal function enhancement. However, clinical evidence indicates that these interventions demonstrate limited efficacy in reversing gastric mucosal damage, particularly in patients presenting with extensive or moderate-to-severe mucosal atrophy (80). Notably, accumulating evidence from multiple studies has demonstrated that pharmacological inhibition of ferroptosis can directly modulate the core pathological processes of CAG through dual mechanisms: Attenuating gastric mucosal injury and suppressing inflammatory responses (81,82).
A number of studies have focused on differentially expressed genes associated with ferroptosis in CAG (83,84). Mechanistically, previous clinical studies have revealed that patients with CAG frequently exhibit abnormal iron metabolism or iron deficiency anemia, which is associated with hepcidin, an antimicrobial polypeptide secreted by gastric parietal cells (Fig. 4) (85,86). Hepcidin inhibits iron efflux by directly binding to ferroportin to cause conformational change and trigger endocytosis and lysosomal degradation, which plays an important role in regulating iron balance (87). Furthermore, CAG has been shown to mediate hepcidin expression via the IL-6/signal transducer and activator of transcription 3 (STAT3) signaling pathway, with increased IL-6 expression being intimately linked to H. pylori infection (84,88). Elevated hepcidin levels decrease the expression of divalent metal transporter 1 and ferroportin 1 proteins, inhibiting duodenal iron absorption and leading to disrupted iron metabolism and gastric cell ferroptosis (84).
The current clinical drug mechanisms for the treatment of CAG mainly include regulation of gastric acid secretion, eradication of H. pylori, protection of gastric mucosa and inhibition of inflammatory factors (89). However, due to the limitations and adverse effects associated with the long-term use of conventional medications, recent studies have explored the therapeutic efficacy and underlying mechanisms of traditional Chinese medicine (TCM) drugs and natural molecular compounds in CAG, particularly focusing on their modulation of inflammation and ferroptosis (81,82). One notable study demonstrated that Xianglianhuazhuo can regulate the Yin Yang 1/miR-320a/TFRC axis, effectively inhibiting gastric epithelial cell proliferation, promoting apoptosis, suppressing ferroptosis, ameliorating gastric mucosa pathology and alleviating CAG symptoms (81). These beneficial effects are postulated to be related to the anti-inflammatory, anticancer and antioxidant properties of components like berberine (Fig. 4) (81). Similar studies have also reported the potential value of Galangin targeting ferroptosis in the treatment of CAG (82). Therefore, the therapeutic strategy of inhibiting ferroptosis in CAG through low-toxicity TCM and natural molecular compounds has potential.
The most controversial issue regarding the stage of IM is whether its progression can be reversed by therapeutic strategies such as eradication of H. pylori (90,91). A previous study has revealed an important role of apoptosis in the transformation of IM to GC (92). However, to the best of our knowledge, there are relatively few studies related to other types of PCD in IM. Notably, several studies have identified ferroptosis-related genes in IM as potential biomarkers for IM diagnosis and novel therapeutic targets such as GOT1, ACSF2, SESN2, HMOX1 and FTL (93-95). Furthermore, a recent study reported that ranolrazole could attenuate IM by inhibiting GPX4 expression to enhance ferroptosis (Fig. 4) (96). However, in general, the regulatory mechanism of ferroptosis in IM remains to be elucidated.
Although progress has been made in exploring ferroptosis as a therapeutic target for PLGC, significant limitations remain. First, systematic studies are lacking to definitively establish whether dysregulated ferroptosis constitutes a key mechanism driving lesion progression or regression. Due to the well-documented relationship between ferroptosis and chronic inflammation, research focusing on the inflammation-cancer transformation axis may offer an insight in addressing this fundamental question (97,98). Secondly, suitable experimental models are still deficient for elucidating the temporal dynamics and spatial heterogeneity of ferroptosis regulation within PLGC, particularly in IM. Emerging technologies such as single-cell sequencing and spatial transcriptomics, alongside models such as spasmolytic polypeptide expressing metaplasia, hold promise for providing novel insights and guiding future experimental designs (99,100). Finally, the related research on targeting ferroptosis in PLGC is still at the basic theoretical stage and lacks evidence to achieve clinical translation. The establishment of gastric organoids derived from CAG/IM patient tissues may be a key model to highly mimic the in vivo environment in the future (101).
At present, there are notable issues in the first-line conventional treatment regimens and experimental novel treatment regimens for GC. Current therapeutic strategies for GC face significant challenges in both conventional first-line treatments and emerging experimental regimens. Persistent issues including acquired drug resistance, restricted patient eligibility for targeted therapies and dose-limiting toxicities associated with combination therapies necessitate urgent optimization (102,103). Furthermore, the clinical application of innovative approaches such as CAR-T cell therapy is constrained by suboptimal efficacy and the absence of well-defined molecular targets, which substantially impedes the advancement of novel treatment paradigms (104,105).
Emerging evidence has demonstrated that oxidative stress plays a pivotal role in the initial phases of inflammation-associated carcinogenesis. Mechanistic studies have revealed that reduced iron uptake and diminished intracellular iron reserves may significantly contribute to GC pathogenesis. These findings provide a compelling rationale for developing targeted therapeutic strategies against GC through selective induction of ferroptosis in malignant cells (Table I) (106-108). The systemic xc−-GSH-GPX4 pathway plays a pivotal role in ferroptosis inhibition, thereby promoting the development of GC. Specifically, the transcription factor megakaryocytic leukemia factor 1 binds to CArG box sites in the promoters of SLC3A2 and SLC7A11, enhancing their transcription and subsequently increasing GSH levels, which inhibits ferroptosis in GC cells (109). Glutamate-cysteine ligase, the rate-limiting enzyme for GSH synthesis, is crucial for this process (110). Furthermore, Aldo-keto reductase 1 member B1 participates in lipid metabolism regulation by removing the aldehyde group from GSH. It specifically modulates GPX4 by decreasing ROS accumulation and lipid peroxidation, lowering intracellular ferrous ion and malondialdehyde levels, and increasing GSH expression, thereby inhibiting RSL3-induced ferroptosis in GC.
Table IPromising strategies for gastric cancer through induction of ferroptosis and relevant mechanisms. |
Previous evidence has increasingly highlighted the pivotal role of ferroptosis in the metastasis and invasion of GC. Epithelial-mesenchymal transition (EMT) is well recognized as a critical mechanism driving tumor metastasis. Specifically, 2,2'-dipyridinone hydrazide dithiocarbamate butyrate demonstrates anticancer efficacy in gastric and esophageal cancer cells. It inhibits transforming growth factor-β1 in GC cells by inducing ferritinophagy and activating the p53 and prolyl hydroxylase domain protein 2/hypoxia-inducible factor 1α (HIF-1α) pathways, ultimately suppressing EMT (111,112). Additionally, its homolog, 2,2'-dipyridyl ketone hydrazine-thiocarbamate, also exhibits inhibitory effects on EMT in GC cells through the induction of ferritinophagy and activation of the p53/AKT/mTOR pathway (113). Furthermore, ferroptosis triggered by ferritin autophagy, coupled with the generation of excessive ROS, further mediates the suppression of EMT (112). Moreover, A previous study revealed that the cystatin inhibitor, cystatin SN, regulates GPX4 protein stability by recruiting OTU domain-containing ubiquitin aldehyde-binding protein 1 to inhibit ferroptosis, thereby promoting GC metastasis (114). Collectively, these findings suggest that targets associated with ferroptosis may offer promising avenues for inhibiting tumor metastasis and progression.
Epigenetic modulation of ferroptosis also constitutes a pivotal mechanism in the development and progression of GC. A recent study has demonstrated that mesenchymal GC cells exhibit upregulated expression of very long chain fatty acid elongation protein 5 and fatty acid desaturase 1, sensitizing them to ferroptosis. Conversely, intestinal-type GC cells display resistance to ferroptosis due to the silencing of these enzymes via DNA methylation (115). Additionally, non-coding RNAs are linked to ferroptosis regulation. Furthermore, research on long non-coding RNA (lncRNA) PMAN has revealed that HIF-1α inhibits ferroptosis in peritoneal metastasis of GC by upregulating lncRNA-PMAN, which is highly expressed in peritoneal metastases and is associated with poor prognosis (116).
The induction of ferroptosis as a novel strategy for the treatment of GC has made some achievements in recent years. On the one hand, emerging studies indicate that novel molecular compounds exert antitumor effects in GC through ferroptosis induction, offering a promising therapeutic alternative for patients with compromised tolerance to conventional chemoradiotherapy-associated systemic toxicity (Fig. 4) (117-120). On the other hand, inducing ferroptosis to improve the chemoresistance of GC has been shown to be an indirect way to inhibit the development of GC. Related studies have further explored and developed substances that can regulate ferroptosis-related genes (121,122) (Fig. 4). Ferroptosis negative regulation-related genes (GPX4, SLC7A11 and ferritin heavy chain 1) and STAT3 have been reported to be upregulated in 5-FU-resistant cells and xenografts (121). W1131 can alleviate chemoresistance in GC by inducing ferroptosis as a novel STAT3 inhibitor, which makes it combine with chemotherapeutic drugs for the treatment of chemotherapy-resistant GC (121). In addition to the aforementioned strategies, there are some innovative studies that provide novel perspectives for the treatment of GC. One study has proposed that atanorin driven by nanomaterials superparamagnetic iron oxide nanoparticles can be used to induce ferroptosis of GC stem cells (122).
In summary, ferroptosis-targeting strategies hold significant therapeutic promise for GC. However, several key challenges require further elucidation. The current mechanistic understanding remains insufficient. Critical unresolved questions include the differential regulation of ferroptosis across molecular GC subtypes and the influence of the tumor microenvironment on ferroptosis sensitivity (123,124). Future investigations should prioritize applying single-cell multi-omics analyses and GC organoid/immune cell co-culture models to address these gaps (124). In addition, the clinical translation of ferroptosis induction faces substantial limitations. Specifically, existing ferroptosis inducers lack tumor-specific targeting, and the synergistic potential of ferroptosis induction combined with immunotherapy or targeted therapy lacks robust theoretical and experimental validation. Consequently, future research efforts should focus on integrating advanced drug delivery technologies (e.g., responsive nanocarriers) and rigorously exploring novel combination therapeutic strategies (125,126).
The high diagnosis rate of advanced GC indicates that the prevention and treatment of GC remain to be improved. The prognostic markers related to ferroptosis screened by relevant studies have important clinical significance in guiding the treatment of GC (Table II). A study from Japan investigated the relationship between GPX4, FSP1 and 4-HNE in tissues of patients with GC and their prognosis (127). In this study, by combining 163 pT3 or pT4 GC tissue samples and OS analysis, it was found that patients with high GPX4 expression and low 4-HNE accumulation had a poor prognosis (P=0.023), while patients with low FSP1 expression and high 4-HNE accumulation had an improved prognosis (P=0.033) (127). The results also suggest that GPX4 and FSP1 may be potential therapeutic targets for patients with GC with poor prognosis. SLC2A3 is another ferroptosis marker. Univariate and multivariate Cox regression analysis revealed that high expression of SLC2A3 was associated with poor prognosis of patients with GC. Functional enrichment analysis showed that SLC2A3 was related to cytokine-cytokine receptor interaction, epithelial-mesenchymal transition, T cell receptor signaling pathway, B cell receptor signaling pathway, immune checkpoints and tumor microenvironment regulation. SLC2A3 and related miRNAs are potential prognostic biomarkers and therapeutics (128).
In addition, lncRNAs can regulate ferroptosis on the epigenetic mechanism of GC, and the use of a variety of lncRNAs to construct GC risk models has shown great advantages. A relative study developed a novel ferroptosis-related prognostic model incorporating 2 mRNAs and 15 lncRNAs to predict outcomes in patients with GC. The model combined clinical features and key factors, showed good predictive ability, and performed well in external patient data validation, which is expected to improve the clinical treatment effect of patients with GC (129). Another study identified 26 ferroptosis-related lncRNAs with independent prognostic value and constructed a risk score model based on four high-risk lncRNAs associated with poor prognosis of gastric adenocarcinoma (130).
Ferroptosis, a newly identified form of regulated cell death, plays a key role in numerous physiological and pathological processes. While significant progress has been made in elucidating the molecular mechanisms of ferroptosis through basic research, its precise role in diseases-particularly H. pylori-associated GC-remains incompletely understood. In the context of limited effective treatments for GC, systematic investigations into ferroptosis dysregulation during H. pylori pathogenesis and the identification of ferroptosis-related therapeutic targets within Correa's cascade are critical for developing novel and effective strategies. By integrating multidisciplinary approaches, including systems biology, nanotechnology and computational drug design, innovative drug platforms can be developed to precisely modulate ferroptosis pathways. These advancements could pave the way for novel strategies to halt or even reverse the progression of Correa's cascade. In conclusion, targeting ferroptosis represents a promising strategy with significant potential for the timely intervention of PLGC, as well as the early diagnosis and precision treatment of GC.
The data generated in the present study may be requested from the corresponding author.
CW wrote the manuscript. MW and CX revised the manuscript. CY and MW contributed to the manuscript equally. All authors have read and approved the final read manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
These authors declare that they have no competing interests.
|
GC |
gastric cancer |
|
PLGC |
precancerous lesions of gastric cancer |
|
H. pylori |
Helicobacter pylori; PCD, programmed cell death |
|
IL |
interleukin |
|
TNF |
tumor necrosis factor |
|
GSH |
glutathione |
|
GPX |
glutathione peroxidase |
|
PLOOHs |
phospholipid hydroperoxides |
|
SLC3A2 |
solute carrier family 3 member 2 |
|
ROS |
reactive oxygen species |
|
PUFA |
polyunsaturated fatty acid |
|
4-HNE |
4-hydroxynonenal |
|
LOX |
lipoxygenase |
|
FSP1 |
ferroptosis suppressor protein 1 |
|
PI3K |
phosphatidylinositol-3-kinase |
|
AKT |
protein kinase B |
|
mTOR |
mammalian target of rapamycin |
|
mTORC1 |
mechanistic target of rapamycin complex 1 |
|
VacA |
vacuolar cell toxin |
|
CagA |
cytotoxin-associated gene A |
|
RNS |
reactive nitrogen species |
|
Bcl-2 |
B-cell lymphoma-2 |
|
Mtb |
Mycobacterium tuberculosis |
|
YWHAE |
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon |
|
OMVs |
outer membrane vesicles |
|
TFRC/TFR1 |
transferrin receptor/transferrin receptor protein 1 |
|
CAG |
chronic atrophic gastritis |
|
IM |
intestinal metaplasia |
|
STAT3 |
signal transducer and activator of transcription 3 |
|
EMT |
epithelial-mesenchymal transition |
|
HIF-1α |
hypoxia-inducible factor 1α |
The authors would like to thank Dr. Huan Wang of the First Affiliated Hospital of Nanchang University (Nanchang, China) for her guidance in the development of the framework for the article as well as her help in writing this paper.
This work was supported by the National Natural Science Foundation of China (grant nos. 82100599 and 82560121); the Jiangxi Provincial Department of Science and Technology (grant no. 20242BAB26122); the Science and Technology Plan of Jiangxi Provincial Administration of Traditional Chinese Medicine (grant no. 2023Z021); the Project of Jiangxi Provincial Academic and Technical Leaders Training Program for Major Disciplines (grant no. 20243BCE51001); and the Ganpo Talent Program - Innovative High end Talents (grant no. gpyc20240212).
|
Yuan J and Ofengeim D: A guide to cell death pathways. Nat Rev Mol Cell Biol. 25:379–395. 2024. View Article : Google Scholar | |
|
Koren E and Fuchs Y: Modes of regulated cell death in cancer. Cancer Discov. 11:245–265. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kolb JP, Oguin TH III, Oberst A and Martinez J: Programmed Cell Death and Inflammation: Winter Is Coming. Trends Immunol. 38:705–718. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X and Shi S: Targeting cell death pathways for cancer therapy: Recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 15:1742022. View Article : Google Scholar : PubMed/NCBI | |
|
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 | |
|
Escuder-Rodríguez JJ, Liang D, Jiang X and Sinicrope FA: Ferroptosis: Biology and Role in Gastrointestinal Disease. Gastroenterology. 167:231–249. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Moss SF, Shah SC, Tan MC and El-Serag HB: Evolving Concepts in Helicobacter pylori Management. Gastroenterology. 166:267–283. 2024. View Article : Google Scholar | |
|
Lei CQ, Wu X, Zhong X, Jiang L, Zhong B and Shu HB: USP19 Inhibits TNF-α- and IL-1β-Triggered NF-κB Activation by Deubiquitinating TAK1. J Immunol. 203:259–268. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Bartchewsky W Jr, Martini MR, Masiero M, Squassoni AC, Alvarez MC, Ladeira MS, Salvatore D, Trevisan M, Pedrazzoli J Jr and Ribeiro ML: Effect of Helicobacter pylori infection on IL-8, IL-1beta and COX-2 expression in patients with chronic gastritis and gastric cancer. Scand J Gastroenterol. 44:153–161. 2009. View Article : Google Scholar | |
|
El Filaly H, Desterke C, Outlioua A, Badre W, Rabhi M, Karkouri M, Riyad M, Khalil A, Arnoult D and Akarid K: CXCL-8 as a signature of severe Helicobacter pylori infection and a stimulator of stomach region-dependent immune response. Clin Immunol. 252:1096482023. View Article : Google Scholar : PubMed/NCBI | |
|
Correa P, Haenszel W, Cuello C, Tannenbaum S and Archer M: A model for gastric cancer epidemiology. Lancet. 2:58–60. 1975. View Article : Google Scholar : PubMed/NCBI | |
|
Tahara S, Tahara T, Horiguchi N, Kato T, Shinkai Y, Yamashita H, Yamada H, Kawamura T, Terada T, Okubo M, et al: DNA methylation accumulation in gastric mucosa adjacent to cancer after Helicobacter pylori eradication. Int J Cancer. 144:80–88. 2019. View Article : Google Scholar | |
|
Qu X and Shi Y: Bile reflux and bile acids in the progression of gastric intestinal metaplasia. Chin Med J (Engl). 135:1664–1672. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Matsuoka T and Yashiro M: Novel biomarkers for early detection of gastric cancer. World J Gastroenterol. 29:2515–2533. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bae S, Lee H, Her EY, Lee K, Kim JS, Ahn J, Choi IJ, Jun JK, Choi KS and Suh M: Cost Utility analysis of National cancer screening program for gastric cancer in Korea: A markov model analysis. J Korean Med Sci. 40:e432025. View Article : Google Scholar : PubMed/NCBI | |
|
Sugimoto N, Kawada J, Oka Y, Ueda S, Murakami K, Nishikawa K, Kurokawa Y, Fujitani K, Kawakami H, Endo S, et al: Salvage-line of capecitabine plus oxaliplatin therapy (XELOX) for patients with inoperable/advanced gastric cancer resistant/intolerant to cisplatin (OGSG1403). Anticancer Res. 45:307–313. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Nakamura Y, Kawazoe A, Lordick F, Janjigian YY and Shitara K: Biomarker-targeted therapies for advanced-stage gastric and gastro-oesophageal junction cancers: An emerging paradigm. Nat Rev Clin Oncol. 18:473–487. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Shitara K, Özgüroğlu M, Bang YJ, Di Bartolomeo M, Mandalà M, Ryu MH, Fornaro L, Olesiński T, Caglevic C, Chung HC, et al: Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): A randomised, open-label, controlled, phase 3 trial. Lancet. 392:123–133. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, Wyrwicz L, Yamaguchi K, Skoczylas T, Campos Bragagnoli A, et al: First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): A randomised, open-label, phase 3 trial. Lancet. 398:27–40. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Bannai S and Kitamura E: Transport interaction of L-cystine and L-glutamate in human diploid fibroblasts in culture. J Biol Chem. 255:2372–2376. 1980. View Article : Google Scholar : PubMed/NCBI | |
|
Sato H, Tamba M, Ishii T and Bannai S: Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J Biol Chem. 274:11455–11458. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Sato H, Tamba M, Kuriyama-Matsumura K, Okuno S and Bannai S: Molecular cloning and expression of human xCT, the light chain of amino acid transport system xc. Antioxid Redox Signal. 2:665–671. 2000. View Article : Google Scholar | |
|
Ursini F, Maiorino M, Valente M, Ferri L and Gregolin C: Purification from pig liver of a protein which protects liposomes and biomembranes from peroxidative degradation and exhibits glutathione peroxidase activity on phosphatidylcholine hydroperoxides. Biochim Biophys Acta. 710:197–211. 1982. View Article : Google Scholar : PubMed/NCBI | |
|
Conrad M and Pratt DA: The chemical basis of ferroptosis. Nat Chem Biol. 15:1137–1147. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WS and Stockwell BR: Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 15:234–245. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Dolma S, Lessnick SL, Hahn WC and Stockwell BR: Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 3:285–296. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Cao Z, Liu X, Zhang W, Zhang K, Pan L, Zhu M, Qin H, Zou C, Wang W, Zhang C, et al: Biomimetic macrophage membrane-camouflaged nanoparticles induce ferroptosis by promoting mitochondrial damage in glioblastoma. ACS Nano. 17:23746–23760. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zou Y, Li H, Graham ET, Deik AA, Eaton JK, Wang W, Sandoval-Gomez G, Clish CB, Doench JG and Schreiber SL: Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 16:302–309. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Wan Y, Jiang Y, Zhang L and Cheng W: GPX4: The hub of lipid oxidation, ferroptosis, disease and treatment. Biochim Biophys Acta Rev Cancer. 1878:1888902023. View Article : Google Scholar : PubMed/NCBI | |
|
Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al: The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 575:688–692. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, 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 | |
|
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 | |
|
Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : | |
|
Lee H, Zandkarimi F, Zhang Y, Meena JK, Kim J, Zhuang L, Tyagi S, Ma L, Westbrook TF, Steinberg GR, et al: Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol. 22:225–234. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Li M, Yao X, Fei Y, Lin Z, Li Z, Cai K, Zhao Y and Luo Z: HCAR1/MCT1 regulates tumor ferroptosis through the lactate-mediated AMPK-SCD1 activity and its therapeutic implications. Cell Rep. 33:1084872020. View Article : Google Scholar : PubMed/NCBI | |
|
Yi J, Zhu J, Wu J, Thompson CB and Jiang X: Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci USA. 117:31189–31197. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Minikes AM, Gao M, Bian H, Li Y, Stockwell BR, Chen ZN and Jiang X: Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature. 572:402–406. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
van Roy F and Berx G: The cell-cell adhesion molecule E-cadherin. Cell Mol Life Sci. 65:3756–3788. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Kim NG, Koh E, Chen X and Gumbiner BM: E-cadherin mediates contact inhibition of proliferation through Hippo signaling-pathway components. Proc Natl Acad Sci USA. 108:11930–11935. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R and Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 520:57–62. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Tarangelo A, Magtanong L, Bieging-Rolett KT, Li Y, Ye J, Attardi LD and Dixon SJ: p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 22:569–575. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, Zhong M, Yuan H, Zhang L, Billiar TR, et al: The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 20:1692–1704. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, Dai X, Li Z and Wu G: Ferroptosis: A novel anti-tumor action for cisplatin. Cancer Res Treat. 50:445–460. 2018. View Article : Google Scholar : | |
|
Lachaier E, Louandre C, Godin C, Saidak Z, Baert M, Diouf M, Chauffert B and Galmiche A: Sorafenib induces ferroptosis in human cancer cell lines originating from different solid tumors. Anticancer Res. 34:6417–6422. 2014.PubMed/NCBI | |
|
Zhang X, Hong B, Li H, Sun Z, Zhao J, Li M, Wei D, Wang Y and Zhang N: Disulfidptosis and ferroptosis related genes define the immune microenvironment and NUBPL serves as a potential biomarker for predicting prognosis and immunotherapy response in bladder cancer. Heliyon. 10:e376382024. View Article : Google Scholar : PubMed/NCBI | |
|
Kuang Y, Yang K, Meng L, Mao Y, Xu F and Liu H: Identification and validation of ferroptosis-related biomarkers and the related pathogenesis in precancerous lesions of gastric cancer. Sci Rep. 13:160742023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Kang R, Kroemer G and Tang D: Broadening horizons: The role of ferroptosis in cancer. Nat Rev Clin Oncol. 18:280–296. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu W, Liu D, Lu Y, Sun J, Zhu J, Xing Y, Ma X, Wang Y, Ji M and Jia Y: PHKG2 regulates RSL3-induced ferroptosis in Helicobacter pylori related gastric cancer. Arch Biochem Biophys. 740:1095602023. View Article : Google Scholar : PubMed/NCBI | |
|
Dierge E, Debock E, Guilbaud C, Corbet C, Mignolet E, Mignard L, Bastien E, Dessy C, Larondelle Y and Feron O: Peroxidation of n-3 and n-6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis-mediated anticancer effects. Cell Metab. 33:1701–1715.e5. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Greenfield LK and Jones NL: Modulation of autophagy by Helicobacter pylori and its role in gastric carcinogenesis. Trends Microbiol. 21:602–612. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Genta RM: Helicobacter pylori, inflammation, mucosal damage, and apoptosis: Pathogenesis and definition of gastric atrophy. Gastroenterology. 113(6 Suppl): S51–S55. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu P, Xue J, Zhang ZJ, Jia YP, Tong YN, Han D, Li Q, Xiang Y, Mao XH and Tang B: Helicobacter pylori VacA induces autophagic cell death in gastric epithelial cells via the endoplasmic reticulum stress pathway. Cell Death Dis. 8:32072017. View Article : Google Scholar : PubMed/NCBI | |
|
Teng Y, Liu X, Han B, Ma Q, Liu Y, Kong H, Lv Y, Mao F, Cheng P, Hao C, et al: Helicobacter pylori-downregulated tumor necrosis factor receptor-associated protein 1 mediates apoptosis of human gastric epithelial cells. J Cell Physiol. 234:15698–15707. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chaturvedi R, Asim M, Romero-Gallo J, Barry DP, Hoge S, de Sablet T, Delgado AG, Wroblewski LE, Piazuelo MB, Yan F, et al: Spermine oxidase mediates the gastric cancer risk associated with Helicobacter pylori CagA. Gastroenterology. 141:1696-1708.e1–e2. 2011. View Article : Google Scholar | |
|
Wu S, Chen Y, Chen Z, Wei F, Zhou Q, Li P and Gu Q: Reactive oxygen species and gastric carcinogenesis: The complex interaction between Helicobacter pylori and host. Helicobacter. 28:e130242023. View Article : Google Scholar : PubMed/NCBI | |
|
Salvatori S, Marafini I, Laudisi F, Monteleone G and Stolfi C: Helicobacter pylori and gastric cancer: Pathogenetic mechanisms. Int J Mol Sci. 24:28952023. View Article : Google Scholar : PubMed/NCBI | |
|
Srinivas US, Tan BWQ, Vellayappan BA and Jeyasekharan AD: ROS and the DNA damage response in cancer. Redox Biol. 25:1010842019. View Article : Google Scholar : PubMed/NCBI | |
|
Teymournejad O, Mobarez AM, Hassan ZM and Talebi Bezmin Abadi A: Binding of the Helicobacter pylori OipA causes apoptosis of host cells via modulation of Bax/Bcl-2 levels. Sci Rep. 7:80362017. View Article : Google Scholar : PubMed/NCBI | |
|
Jain P, Luo ZQ and Blanke SR: Helicobacter pylori vacuolating cytotoxin A (VacA) engages the mitochondrial fission machinery to induce host cell death. Proc Natl Acad Sci USA. 108:16032–16037. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Posselt G, Wiesauer M, Chichirau BE, Engler D, Krisch LM, Gadermaier G, Briza P, Schneider S, Boccellato F, Meyer TF, et al: Helicobacter pylori-controlled c-Abl localization promotes cell migration and limits apoptosis. Cell Commun Signal. 17:102019. View Article : Google Scholar : PubMed/NCBI | |
|
Lin Y, Liu K, Lu F, Zhai C and Cheng F: Programmed cell death in Helicobacter pylori infection and related gastric cancer. Front Cell Infect Microbiol. 14:14168192024. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar S and Dhiman M: Inflammasome activation and regulation during Helicobacter pylori pathogenesis. Microb Pathog. 125:468–474. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Cui G, Yuan A and Li Z: Occurrences and phenotypes of RIPK3-positive gastric cells in Helicobacter pylori infected gastritis and atrophic lesions. Dig Liver Dis. 54:1342–1349. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Qiang L, Zhang Y, Lei Z, Lu Z, Tan S, Ge P, Chai Q, Zhao M, Zhang X, Li B, et al: A mycobacterial effector promotes ferroptosis-dependent pathogenicity and dissemination. Nat Commun. 14:14302023. View Article : Google Scholar : PubMed/NCBI | |
|
Dar HH, Tyurina YY, Mikulska-Ruminska K, Shrivastava I, Ting HC, Tyurin VA, Krieger J, St Croix CM, Watkins S, Bayir E, et al: Pseudomonas aeruginosa utilizes host polyunsaturated phosphatidylethanolamines to trigger theft-ferroptosis in bronchial epithelium. J Clin Invest. 128:4639–4653. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Drake IM, Mapstone NP, Schorah CJ, White KL, Chalmers DM, Dixon MF and Axon AT: Reactive oxygen species activity and lipid peroxidation in Helicobacter pylori associated gastritis: Relation to gastric mucosal ascorbic acid concentrations and effect of H pylori eradication. Gut. 42:768–771. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Peng Y, Lei X, Yang Q, Zhang G, He S, Wang M, Ling R, Zheng B, He J, Chen X, et al: Helicobacter pylori CagA-mediated ether lipid biosynthesis promotes ferroptosis susceptibility in gastric cancer. Exp Mol Med. 56:441–452. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu D, Peng J, Xie J and Xie Y: Comprehensive analysis of the function of helicobacter-associated ferroptosis gene YWHAE in gastric cancer through multi-omics integration, molecular docking, and machine learning. Apoptosis. 29:439–456. 2024. View Article : Google Scholar | |
|
Melo J, Cavadas B, Pereira L, Figueiredo C and Leite M: Transcriptomic remodeling of gastric cells by Helicobacter pylori outer membrane vesicles. Helicobacter. 29:e130312024. View Article : Google Scholar | |
|
Shen C, Liu H, Chen Y, Liu M, Wang Q and Liu J and Liu J: Helicobacter pylori induces GBA1 demethylation to inhibit ferroptosis in gastric cancer. Mol Cell Biochem. 480:1845–1863. 2025. View Article : Google Scholar | |
|
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 | |
|
Gryzik M, Asperti M, Denardo A, Arosio P and Poli M: NCOA4-mediated ferritinophagy promotes ferroptosis induced by erastin, but not by RSL3 in HeLa cells. Biochim Biophys Acta Mol Cell Res. 1868:1189132021. View Article : Google Scholar | |
|
Chen X, Yu C, Kang R, Kroemer G and Tang D: Cellular degradation systems in ferroptosis. Cell Death Differ. 28:1135–1148. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Y, Xu W and Zhou R: NLRP3 inflammasome activation and cell death. Cell Mol Immunol. 18:2114–2127. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Miao R, Xia J, Zhou Y, Yao J and Shao S: Infection of Helicobacter pylori contributes to the progression of gastric cancer through ferroptosis. Cell Death Discov. 10:4852024. View Article : Google Scholar : PubMed/NCBI | |
|
Piscione M, Mazzone M, Di Marcantonio MC, Muraro R and Mincione G: Eradication of helicobacter pylori and gastric cancer: A controversial relationship. Front Microbiol. 12:6308522021. View Article : Google Scholar : PubMed/NCBI | |
|
White JR, Winter JA and Robinson K: Differential inflammatory response to Helicobacter pylori infection: Etiology and clinical outcomes. J Inflamm Res. 8:137–147. 2015.PubMed/NCBI | |
|
Shah SC, Piazuelo MB, Kuipers EJ and Li D: AGA clinical practice update on the diagnosis and management of atrophic gastritis: Expert review. Gastroenterology. 161:1325–1332.e7. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Y, Jia X, Du P, Wang J, Du Y, Li B, Xue Y, Jiang J, Cai Y and Yang Q: Mechanistic insights into the ameliorative effects of Xianglianhuazhuo formula on chronic atrophic gastritis through ferroptosis mediated by YY1/miR-320a/TFRC signal pathway. J Ethnopharmacol. 323:1176082024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang T, Lu M, Jiang W, Jin D, Sun M, Mao H and Han H: Galangin alleviates gastric mucosal injury in rats with chronic atrophic gastritis by reducing ferroptosis. Histol Histopathol. January 24–2025.Epub ahead of print. | |
|
Pan W, Liu C, Ren T, Chen X, Liang C, Wang J and Yang J: Exploration of lncRNA/circRNA-miRNA-mRNA network in patients with chronic atrophic gastritis in Tibetan plateau areas based on DNBSEQ-G99 RNA sequencing. Sci Rep. 14:92122024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Zhao J, Ma H, Han Y, Xu W, Wang J, Cai Y, Jia X, Jia Q and Yang Q: High hepcidin levels promote abnormal iron metabolism and ferroptosis in chronic atrophic gastritis. Biomedicines. 11:23382023. View Article : Google Scholar : PubMed/NCBI | |
|
Lanser L, Fuchs D, Kurz K and Weiss G: Physiology and inflammation driven pathophysiology of iron homeostasis-mechanistic insights into anemia of inflammation and its treatment. Nutrients. 13:37322021. View Article : Google Scholar : PubMed/NCBI | |
|
Schwarz P, Kübler JA, Strnad P, Müller K, Barth TF, Gerloff A, Feick P, Peyssonnaux C, Vaulont S, Adler G and Kulaksiz H: Hepcidin is localised in gastric parietal cells, regulates acid secretion and is induced by Helicobacter pylori infection. Gut. 61:193–201. 2012. View Article : Google Scholar | |
|
Ganz T and Nemeth E: Hepcidin and disorders of iron metabolism. Annu Rev Med. 62:347–360. 2011. View Article : Google Scholar | |
|
Santos MP, Pereira JN, Delabio RW, Smith MAC, Payão SLM, Carneiro LC, Barbosa MS and Rasmussen LT: Increased expression of interleukin-6 gene in gastritis and gastric cancer. Braz J Med Biol Res. 54:e106872021. View Article : Google Scholar : PubMed/NCBI | |
|
Jia J, Zhao H, Li F, Zheng Q, Wang G, Li D and Liu Y: Research on drug treatment and the novel signaling pathway of chronic atrophic gastritis. Biomed Pharmacother. 176:1169122024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu F, Zhang X, Li P and Zhu Y: Effect of Helicobacter pylori eradication on gastric precancerous lesions: A systematic review and meta-analysis. Helicobacter. 28:e130132023. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Y, Yang Y, Nong R, Huang H, Chen X, Deng Y, Huang Z, Huang J, Cheng C, Ji M, et al: Do atrophic gastritis and intestinal metaplasia reverse after Helicobacter pylori eradication? Helicobacter. 29:e130422024. View Article : Google Scholar | |
|
Bir F, Calli-Demirkan N, Tufan AC, Akbulut M and Satiroglu-Tufan NL: Apoptotic cell death and its relationship to gastric carcinogenesis. World J Gastroenterol. 13:3183–3188. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Li T, Yang Q, Liu Y, Jin Y, Song B, Sun Q, Wei S, Wu J and Li X: Machine learning identify ferroptosis-related genes as potential diagnostic biomarkers for gastric intestinal metaplasia. Technol Cancer Res Treat. 23:153303382412720362024. View Article : Google Scholar : PubMed/NCBI | |
|
Song B, Li T, Zhang Y, Yang Q, Pei B, Liu Y, Wang J, Dong G, Sun Q, Fan S and Li X: Identification and verification of ferroptosis-related genes in gastric intestinal metaplasia. Front Genet. 14:11524142023. View Article : Google Scholar : PubMed/NCBI | |
|
Hamedi Asl D, Naserpour Farivar T, Rahmani B, Hajmanoochehri F, Emami Razavi AN, Jahanbin B, Soleimani Dodaran M and Peymani A: The role of transferrin receptor in the Helicobacter pylori pathogenesis; L-ferritin as a novel marker for intestinal metaplasia. Microb Pathog. 126:157–164. 2019. View Article : Google Scholar | |
|
Xie J, Liang X, Xie F, Huang C, Lin Z, Xie S, Yang F, Zheng F, Geng L, Xu W, et al: Rabeprazole suppressed gastric intestinal metaplasia through activation of GPX4-mediated ferroptosis. Front Pharmacol. 15:14090012024. View Article : Google Scholar : PubMed/NCBI | |
|
Ebrahimi N, Adelian S, Shakerian S, Afshinpour M, Chaleshtori SR, Rostami N, Rezaei-Tazangi F, Beiranvand S, Hamblin MR and Aref AR: Crosstalk between ferroptosis and the epithelial-mesenchymal transition: Implications for inflammation and cancer therapy. Cytokine Growth Factor Rev. 64:33–45. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M, Zhong J, Song Z, Xu Q, Chen Y and Zhang Z: Regulatory mechanisms and potential therapeutic targets in precancerous lesions of gastric cancer: A comprehensive review. Biomed Pharmacother. 177:1170682024. View Article : Google Scholar : PubMed/NCBI | |
|
Gu L, Chen H, Geng R, Sun M, Shi Q, Chen Y, Chang J, Wei J, Ma W, Xiao J, et al: Single-cell and Spatial transcriptomics reveals ferroptosis as the most enriched programmed cell death process in hemorrhage stroke-induced oligodendrocyte-mediated white matter injury. Int J Biol Sci. 20:3842–3862. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Miao ZF, Sun JX, Adkins-Threats M, Pang MJ, Zhao JH, Wang X, Tang KW, Wang ZN and Mills JC: DDIT4 licenses only healthy cells to proliferate during injury-induced metaplasia. Gastroenterology. 160:260–271.e10. 2021. View Article : Google Scholar | |
|
Pang MJ, Burclaff JR, Jin R, Adkins-Threats M, Osaki LH, Han Y, Mills JC, Miao ZF and Wang ZN: Gastric organoids: Progress and remaining challenges. Cell Mol Gastroenterol Hepatol. 13:19–33. 2022. View Article : Google Scholar | |
|
Hu X, Ma Z, Xu B, Li S, Yao Z, Liang B, Wang J, Liao W, Lin L, Wang C, et al: Glutamine metabolic microenvironment drives M2 macrophage polarization to mediate trastuzumab resistance in HER2-positive gastric cancer. Cancer Commun (Lond). 43:909–937. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang F, Li A, Liu H and Zhang H: Gastric cancer combination therapy: Synthesis of a hyaluronic acid and cisplatin containing lipid prodrug coloaded with sorafenib in a nanoparticulate system to exhibit enhanced anticancer efficacy and reduced toxicity. Drug Des Devel Ther. 12:3321–3333. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Cao Y, et al: Claudin18.2-specific CAR T cells in gastrointestinal cancers: Phase 1 trial interim results. Nat Med. 28:1189–1198. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yasuda T and Wang YA: Gastric cancer immunosuppressive microenvironment heterogeneity: Implications for therapy development. Trends Cancer. 10:627–642. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gorrini C, Harris IS and Mak TW: Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov. 12:931–947. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Fonseca-Nunes A, Agudo A, Aranda N, Arija V, Cross AJ, Molina E, Sanchez MJ, Bueno-de-Mesquita HB, Siersema P, Weiderpass E, et al: Body iron status and gastric cancer risk in the EURGAST study. Int J Cancer. 137:2904–2914. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Noto JM, Piazuelo MB, Shah SC, Romero-Gallo J, Hart JL, Di C, Carmichael JD, Delgado AG, Halvorson AE, Greevy RA, et al: Iron deficiency linked to altered bile acid metabolism promotes Helicobacter pylori-induced inflammation-driven gastric carcinogenesis. J Clin Invest. 132:e1478222022. View Article : Google Scholar : PubMed/NCBI | |
|
Dai ZT, Wu YL, Li XR and Liao XH: MKL-1 suppresses ferroptosis by activating system Xc- and increasing glutathione synthesis. Int J Biol Sci. 19:4457–4475. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lu SC: Regulation of glutathione synthesis. Mol Aspects Med. 30:42–59. 2009. View Article : Google Scholar : | |
|
Guan D and Li C, Li Y, Li Y, Wang G, Gao F and Li C: The DpdtbA induced EMT inhibition in gastric cancer cell lines was through ferritinophagy-mediated activation of p53 and PHD2/hif-1α pathway. J Inorg Biochem. 218:1114132021. View Article : Google Scholar | |
|
Guan D, Zhou W, Wei H, Wang T, Zheng K, Yang C, Feng R, Xu R, Fu Y, Li C, et al: Ferritinophagy-mediated ferroptosis and activation of Keap1/Nrf2/HO-1 pathway were conducive to EMT inhibition of gastric cancer cells in action of 2,2'-Di-pyridineketone hydrazone dithiocarbamate butyric acid ester. Oxid Med Cell Longev. 2022:39206642022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Z, Feng J, Li Y, Guan D, Chen H, Zhai X, Zhang L and Li C and Li C: The vicious cycle between ferritinophagy and ROS production triggered EMT inhibition of gastric cancer cells was through p53/AKT/mTor pathway. Chem Biol Interact. 328:1091962020. View Article : Google Scholar : PubMed/NCBI | |
|
Li D, Wang Y, Dong C, Chen T, Dong A, Ren J, Li W, Shu G, Yang J, Shen W, et al: CST1 inhibits ferroptosis and promotes gastric cancer metastasis by regulating GPX4 protein stability via OTUB1. Oncogene. 42:83–98. 2023. View Article : Google Scholar : | |
|
Lee JY, Nam M, Son HY, Hyun K, Jang SY, Kim JW, Kim MW, Jung Y, Jang E, Yoon SJ, et al: Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proc Natl Acad Sci USA. 117:32433–32442. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lin Z, Song J, Gao Y, Huang S, Dou R, Zhong P, Huang G, Han L, Zheng J, Zhang X, et al: Hypoxia-induced HIF-1α/lncRNA-PMAN inhibits ferroptosis by promoting the cytoplasmic translocation of ELAVL1 in peritoneal dissemination from gastric cancer. Redox Biol. 52:1023122022. View Article : Google Scholar | |
|
Liu Y, Song Z, Liu Y, Ma X, Wang W, Ke Y, Xu Y, Yu D and Liu H: Identification of ferroptosis as a novel mechanism for antitumor activity of natural product derivative a2 in gastric cancer. Acta Pharm Sin B. 11:1513–1525. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hu C, Zu D, Xu J, Xu H, Yuan L, Chen J, Wei Q, Zhang Y, Han J, Lu T, et al: Polyphyllin B suppresses gastric tumor growth by modulating iron metabolism and inducing ferroptosis. Int J Biol Sci. 19:1063–1079. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ding L, Dang S, Sun M, Zhou D, Sun Y, Li E, Peng S, Li J and Li G: Quercetin induces ferroptosis in gastric cancer cells by targeting SLC1A5 and regulating the p-Camk2/p-DRP1 and NRF2/GPX4 Axes. Free Radic Biol Med. 213:150–163. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Lu C, Zhou H, Zhao X, Huang C, Cheng Z, Liu G and You X: Synergistic effects of dihydroartemisinin and cisplatin on inducing ferroptosis in gastric cancer through GPX4 inhibition. Gastric Cancer. 28:187–210. 2025. View Article : Google Scholar | |
|
Ouyang S, Li H, Lou L, Huang Q, Zhang Z, Mo J, Li M, Lu J, Zhu K, Chu Y, et al: Inhibition of STAT3-ferroptosis negative regulatory axis suppresses tumor growth and alleviates chemoresistance in gastric cancer. Redox Biol. 52:1023172022. View Article : Google Scholar : PubMed/NCBI | |
|
Ni Z, Nie X, Zhang H, Wang L, Geng Z, Du X, Qian H, Liu W and Liu T: Atranorin driven by nano materials SPION lead to ferroptosis of gastric cancer stem cells by weakening the mRNA 5-hydroxymethylcytidine modification of the Xc-/GPX4 axis and its expression. Int J Med Sci. 19:1680–1694. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cui JX, Xu XH, He T, Liu JJ, Xie TY, Tian W and Liu JY: L-kynurenine induces NK cell loss in gastric cancer microenvironment via promoting ferroptosis. J Exp Clin Cancer Res. 42:522023. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar V, Ramnarayanan K, Sundar R, Padmanabhan N, Srivastava S, Koiwa M, Yasuda T, Koh V, Huang KK, Tay ST, et al: Single-cell atlas of lineage states, tumor microenvironment, and subtype-specific expression programs in gastric cancer. Cancer Discov. 12:670–691. 2022. View Article : Google Scholar | |
|
Zhang Q, Kuang G, Li W, Wang J, Ren H and Zhao Y: Stimuli-responsive gene delivery nanocarriers for cancer therapy. Nanomicro Lett. 15:442023.PubMed/NCBI | |
|
Cheng X, Dai E, Wu J, Flores NM, Chu Y, Wang R, Dang M, Xu Z, Han G, Liu Y, et al: Atlas of metastatic gastric cancer links ferroptosis to disease progression and immunotherapy response. Gastroenterology. 167:1345–1357. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tamura K, Tomita Y, Kanazawa T, Shinohara H, Sakano M, Ishibashi S, Ikeda M, Kinoshita M, Minami J, Yamamoto K, et al: Lipid peroxidation regulators GPX4 and FSP1 as prognostic markers and therapeutic targets in advanced gastric cancer. Int J Mol Sci. 25:92032024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin L, Que R, Wang J, Zhu Y, Liu X and Xu R: Prognostic value of the ferroptosis-related gene SLC2A3 in gastric cancer and related immune mechanisms. Front Genet. 13:9193132022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Liu Y, Ye S, Feng H and Ma L: A new ferroptosis-related signature model including messenger RNAs and long non-coding RNAs predicts the prognosis of gastric cancer patients. J Transl Int Med. 11:145–155. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cai Y, Wu S, Jia Y, Pan X and Li C: Potential key markers for predicting the prognosis of gastric adenocarcinoma based on the expression of ferroptosis-related lncRNA. J Immunol Res. 2022:12492902022. View Article : Google Scholar : PubMed/NCBI | |
|
Ryu MH, Lee KH, Shen L, Yeh KH, Yoo C, Hong YS, Park YI, Yang SH, Shin DB, Zang DY, et al: Randomized phase II study of capecitabine plus cisplatin with or without sorafenib in patients with metastatic gastric cancer (STARGATE). Cancer Med. 12:7784–7794. 2023. View Article : Google Scholar : | |
|
Xu X and Li Y, Wu Y, Wang M, Lu Y, Fang Z, Wang H and Li Y: Increased ATF2 expression predicts poor prognosis and inhibits sorafenib-induced ferroptosis in gastric cancer. Redox Biol. 59:1025642023. View Article : Google Scholar | |
|
Zhuang J, Liu X, Yang Y, Zhang Y and Guan G: Sulfasalazine, a potent suppressor of gastric cancer proliferation and metastasis by inhibition of xCT: Conventional drug in new use. J Cell Mol Med. 25:5372–5380. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Shitara K, Doi T, Nagano O, Fukutani M, Hasegawa H, Nomura S, Sato A, Kuwata T, Asai K, Einaga Y, et al: Phase 1 study of sulfasalazine and cisplatin for patients with CD44v-positive gastric cancer refractory to cisplatin (EPOC1407). Gastric Cancer. 20:1004–1009. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Zheng L, Shang W, Yang Z, Li T, Liu F, Shao W, Lv L, Chai L, Qu L, et al: Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ. 29:2190–2202. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu C, Wang S, Huang T, Xi X, Xu L, Wang J, Hou Y, Xia Y, Xu L, Wang L and Huang X: NPR1 promotes cisplatin resistance by inhibiting PARL-mediated mitophagy-dependent ferroptosis in gastric cancer. Cell Biol Toxicol. 40:932024. View Article : Google Scholar : PubMed/NCBI | |
|
Fu D, Wang C, Yu L and Yu R: Induction of ferroptosis by ATF3 elevation alleviates cisplatin resistance in gastric cancer by restraining Nrf2/Keap1/xCT signaling. Cell Mol Biol Lett. 26:262021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Q, Liu T, Qian W, Ji J, Cai Q, Jin Y, Jiang J and Zhang J: HNF4A-BAP31-VDAC1 axis synchronously regulates cell proliferation and ferroptosis in gastric cancer. Cell Death Dis. 14:3562023. View Article : Google Scholar : PubMed/NCBI | |
|
Qu X, Liu B, Wang L, Liu L, Zhao W, Liu C, Ding J, Zhao S, Xu B, Yu H, et al: Loss of cancer-associated fibroblast-derived exosomal DACT3-AS1 promotes malignant transformation and ferroptosis-mediated oxaliplatin resistance in gastric cancer. Drug Resist Updat. 68:1009362023. View Article : Google Scholar : PubMed/NCBI | |
|
Yao L, Hou J, Wu X, Lu Y, Jin Z, Yu Z, Yu B, Li J, Yang Z, Li C, et al: Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation. Redox Biol. 67:1029232023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun J, Li J, Pantopoulos K, Liu Y, He Y, Kang W and Ye X: The clustering status of detached gastric cancer cells inhibits anoikis-induced ferroptosis to promote metastatic colonization. Cancer Cell Int. 24:772024. View Article : Google Scholar : PubMed/NCBI | |
|
Yao X, He Z, Qin C, Deng X, Bai L, Li G and Shi J: SLC2A3 promotes macrophage infiltration by glycolysis reprogramming in gastric cancer. Cancer Cell Int. 20:5032020. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Xu X, Wang X, Zhang C, Hu A and Li Y: MGST1 expression is associated with poor prognosis, enhancing the Wnt/β-catenin pathway via regulating AKT and inhibiting ferroptosis in gastric cancer. ACS Omega. 8:23683–23694. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Shi M, Ji J, Cai Q, Zhao Q, Jiang J, Liu J, Zhang H, Zhu Z and Zhang J: Stearoyl-CoA desaturase 1 (SCD1) facilitates the growth and anti-ferroptosis of gastric cancer cells and predicts poor prognosis of gastric cancer. Aging (Albany NY). 12:15374–15391. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Yang S, Feng X, Zheng Y, Zhou J, Wang H, Zhang Y, Sun H and He C: The modification of ferroptosis and abnormal lipometabolism through overexpression and knockdown of potential prognostic biomarker perilipin2 in gastric carcinoma. Gastric Cancer. 23:241–259. 2020. View Article : Google Scholar | |
|
Zang J, Cui M, Xiao L, Zhang J and Jing R: Overexpression of ferroptosis-related genes FSP1 and CISD1 is related to prognosis and tumor immune infiltration in gastric cancer. Clin Transl Oncol. 25:2532–2544. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Qian J, Xu J, Bai H, Yang J and Chen L: NRF2 inhibits RSL3 induced ferroptosis in gastric cancer through regulation of AKR1B1. Exp Cell Res. 442:1142102024. View Article : Google Scholar : PubMed/NCBI | |
|
Tu RH, Wu SZ, Huang ZN, Zhong Q, Ye YH, Zheng CH, Xie JW, Wang JB, Lin JX, Chen QY, et al: Neurotransmitter receptor HTR2B regulates lipid metabolism to inhibit ferroptosis in gastric cancer. Cancer Res. 83:3868–3885. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Qu H, Liang Y, Guo Q, Lu L, Yang Y, Xu W, Zhang Y and Qin Y: Identifying CTH and MAP1LC3B as ferroptosis biomarkers for prognostic indication in gastric cancer decoding. Sci Rep. 14:43522024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Zhang S, Li X, Leng C, Li X, Lv J, Zhao S, Qiu W and Guo J: Development of oxidative stress- and ferroptosis-related prognostic signature in gastric cancer and identification of CDH19 as a novel biomarker. Hum Genomics. 18:1212024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y, Li L, Li W, Liu K, Wu Y and Wang Z: NFS1 inhibits ferroptosis in gastric cancer by regulating the STAT3 pathway. J Bioenerg Biomembr. 56:573–587. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y, Li W, Zhang J, Liu K, Wu Y and Wang Z: NFS1 as a candidate prognostic biomarker for gastric cancer correlated with immune infiltrates. Int J Gen Med. 17:3855–3868. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu W, Zhang F, Yang K and Yan Y: Comprehensive analysis regarding the prognostic significance of downregulated ferroptosis-related gene AKR1C2 in gastric cancer and its underlying roles in immune response. PLoS One. 18:e02809892023. View Article : Google Scholar : PubMed/NCBI |