Abstract
Ferroptosis is a form of nonapoptotic cell death that involves iron-dependent phospholipid peroxidation induced by accumulation of reactive oxygen species (ROS), and results in plasma membrane damage and the release of damage-associated molecular patterns. Ferroptosis has been implicated in aging and immunity, as well as disease states including intestinal and liver conditions and cancer. To date, several ferroptosis-associated genes and pathways have been implicated in liver disease. While ferroptotic cell death is associated with dysfunction of the intestinal epithelium, the underlying molecular basis is poorly understood. As the mechanisms regulating ferroptosis become further elucidated, there is clear potential to utilize ferroptosis to achieve therapeutic benefit.
Keywords: cancer, inflammation, tumor microenvironment, cell death
Ferroptosis is an iron-dependent mechanism of non-apoptotic cell death caused by phospholipid peroxidation. As a distinct form of regulated cell death (RCD),1–3 ferroptosis has gained much attention in recent years due to its involvement in pathological processes and immunity including cancer and its related therapy. Ferroptosis initiators have been broadly classified as extrinsic resulting from altering the activity of cell membrane amino acid transporters and iron transporters, or as intrinsic due to inhibition of intracellular antioxidant enzymes such as glutathione peroxidase GPX4.4 Ferroptosis is regulated not by a single universal mechanism, but by diverse and context-dependent pathways that converge at the accumulation of lipid peroxides in the plasma membrane.5 Unlike other forms of RCD such as apoptosis (caspases), necroptosis (mixed-lineage kinase domain like pseudokinase, aka MLKL) or pyroptosis (gasdermin D), specific and indispensable mediators for the execution of ferroptosis remain largely unknown, although proposed models exist.6, 7 Propagation of ferroptosis through the release of damage-associated molecular patterns (DAMPs) has been shown.8–10 DAMPs are immuno-modulatory and can connect ferroptosis with the tumor microenvironment (TME) with implications for immunotherapy as cancer treatment. Of much clinical relevance is how to therapeutically target ferroptosis in a disease context. In this review, we discuss mechanisms and regulation of ferroptosis including its relevance in diseases of the gut and cancer. Furthermore, regulatory axes of ferroptosis and their targeting represent a therapeutic opportunity in cancer cells that have developed acquired resistance to other forms of RCD.
Ferroptosis regulation
Key pathways that can trigger ferroptotic cell death converge in the peroxidation of membrane lipids. One such pathway involves the degradation of ferritin and the engagement of transferrin receptor (TFRC)-mediated iron import, both of which serve to increase the intracellular iron pool leading to lipid peroxidation. Through the Fenton reaction, this iron pool and lipid metabolizing enzymes (ACSL4 and LPCAT3) contribute to an increase in phospholipid peroxidation that leads to ferroptosis. Glutathione peroxidase 4 (GPX4) is the canonical ferroptosis-controlling pathway for the regulation of ferroptosis. The NRF2 transcription factor regulates GPX411 and SCL7A11,12 as well as other lipid peroxidation and ferroptosis-related genes.13 Initiators of ferroptosis that act through the GPX4-dependent pathway include depletion of glutathione (GSH) [the reducing substrate of GPX4 activity], inhibition of the cysteine importer known as system Xc- using erastin,14 or GPX4 inhibition via the small molecule RSL315. There are also GPX4-independent pathways that serve as alternative antioxidant defenses against ferroptosis, such as the ferroptosis suppressor protein 1 (FSP1), dihydroorotate dehydrogenase (DHODH) and GTP cyclohydrolase 1 (GCH1). These antioxidant enzymes result in generation of metabolites (such as CoQ10, BH4, etc.) with radical-trapping antioxidant activity (RTA) to inhibit lipid peroxidation (Figure 1). Most recently, a novel mechanism independent of both GPX4 and RTA has been discovered to suppress ferroptosis through MBOAT1/2-mediated cellular phospholipid remodeling.16
Figure 1.
Pathways that can trigger ferroptotic cell death converge in the peroxidation of membrane lipids. End executors of ferroptosis remain to be elucidated. As a receptor in the plasma membrane, cystine-glutamine antiporter (system Xc-) can be inhibited by erastin or glutamate to extrinsically induce ferroptosis, whereas inhibition of intracellular antioxidant enzymes (GPX4, FSP1, DHODH, GCH1 and DHFR) lead to intrinsically induced ferroptosis.14 Depletion of GSH by MRP1 export,32 upregulated cys dioxygenase 1 (CDO1),202 or treatment with cisplatin can also sensitize cells to ferroptosis.203, 204 The NRF2 pathway is involved in the expression of multiple proteins that can inhibit ferroptosis, including system Xc-, GPX4 and MT1G. The ferroptosis suppressor protein 1 (FSP1) catalyzes the regeneration of coenzyme Q10 (CoQ10)33, 34 whose reduced form ubiquinol, traps lipid peroxyl radicals that mediate lipid peroxidation and prevents ferroptosis induced by GPX4 depletion. GTP cyclohydroxylase-1 [GCH1] is the rate limiting enzyme of the tetrahydrobiopterin (BH4) biosynthetic pathway which is another inhibitor of lipid peroxidation.39 The lipid metabolism enzyme ACSL4, which preferentially acylates AA, and LPCAT3 which preferentially inserts acylated AA into membrane phospholipids, have critical roles in GPX4 inhibition-induced ferroptosis.86, 87 The first step in GSH synthesis is catalyzed by the glutamate-cysteine ligase catalytic subunit (GCLC) contributing to ferroptosis resistance.205 An increase in the intracellular iron pool through ferritinophagy and TFRC also leads to enhanced lipid peroxidation.43
GPX4 pathway in the regulation of ferroptosis
The intestinal mucosal epithelium is highly dependent on dietary cysteine whose deficiency can lead to impaired cell proliferation, defective barrier function, and cell death due to ferroptosis.17 A rate limiting step in cellular GSH synthesis is the availability of cysteine that plays an essential role in cellular redox homoeostasis. Cysteine is a key constituent of the GSH tripeptide that is required for GPX4 to inhibit lipid peroxidation and ferroptosis. Cysteine requires specialized transport systems for its import into the cell. System Xc− is a dedicated cystine transporter that imports cystine in exchange for intracellular glutamate. System Xc− is frequently upregulated in cancer cells to counteract elevated levels of ROS, making it an attractive target for anticancer treatment. Furthermore, system Xc- contains a light chain subunit, SLC7A11, that is targeted by inhibitors such as sulfasalazine or the multikinase inhibitor sorafenib to induce ferroptosis.18,19 SLC7A11 expression can be repressed by the BRCA1-associated protein 1 (BAP1) that inhibits cystine uptake leading to elevated lipid peroxidation and ferroptosis.20 Several non-coding RNAs21–23 have also been shown to modulate expression of SLC7A11 resulting in ferroptosis. Ionizing radiation (IR) can inhibit ferroptosis by inducing expression of both SLC7A11 and GPX4 as an adaptive response leading to radioresistance.24 Interestingly, ferroptosis induced by erastin or RSL3 was shown to be enhanced by sodium butyrate, a short-chain fatty acid (SCFA), by inducing lipid reactive oxygen species (ROS) production via downregulation of the expression of SLC7A11 and GPX425. The nuclear factor erythroid 2-related 2 (NRF2) transcription factor regulates GPX411 and SCL7A1112 as well as many other lipid peroxidation- and ferroptosis- related genes such as metallothionein-1 (MT-1G), an important regulator of lipid peroxidation13 (Figure 1). Inhibition of NRF2 (using all-trans retinoic acid, or brusatol) induces ferroptosis by mechanisms including sensitizing cells to erastin and sorafenib,12 reducing MT-1G expression,26 and inducing redox imbalance and apoptosis in cancer cells.27 There is also a synergistic effect of brusatol with lapatinib, an inhibitor of HER2/EGFR.28
Modulation of GPX4 activity regulates ferroptosis whereby inducers of ferroptosis, including statins, can indirectly downregulate GPX4 expression or activity in cancer cells.29 Conversely, upregulation of GPX4 expression by activating transcription factors (TFAP2c and SP1) resulting from selenium supplementation can inhibit ferroptosis.30 MRP1, a multidrug resistance (MDR) gene,31 functions to increase the cellular efflux of GSH,32 and high levels of MRP1 expression (MDR phenotype) sensitize cancer cells to pro-ferroptotic agents while conferring resistance to some pro-apoptotic anticancer drugs.32 Accordingly, induction of ferroptosis can be exploited to target treatment-resistant cancer cells.
GPX4-independent pathways
The FSP1-CoQ10-NAD(P)H axis represents a parallel system to the GPX4-GSH axis for inhibition of lipid peroxidation and ferroptosis.33, 34 Of note, a combined administration of FSP1 and GPX4 inhibitors showed a more potent induction of ferroptosis compared to individual drugs.35 The ferroptosis suppressor protein 1 (FSP1) can reduce vitamin K to its hydroquinone which acts as an inhibitor of lipid peroxidation, thereby protecting cells from ferroptosis.36 Other parallel systems that inhibit ferroptosis include DHODH,37 and vitamin D3 has been shown to reduce cisplatin-induced intestinal injury by reversing downregulation of both GPX4 and DHODH, explaining its anti-ferroptotic role.38 The tetrahydrobiopterin (BH4) biosynthesis pathway inhibits ferroptosis independent of cytosolic GPX4 or FSP1 (Figure 1). BH4, a radical-trapping antioxidant cofactor, prevents membrane lipid peroxidation and its availability alters iron metabolism and mitochondrial function in T cells. Methotrexate inhibition of dihydrofolate reductase, an enzyme that regenerates BH4, synergizes with GPX4 inhibition in the induction of ferroptosis.39
Iron regulation and iron-induced lipid peroxidation
Phospholipid (PL) peroxidation can be initiated in cells through enzymatic and nonenzymatic processes. If PL hydroperoxide (PLOOH) is formed and not neutralized promptly, it can undergo the iron-catalyzed Fenton reaction, resulting in generation of lipid hydroxyl and lipid peroxyl radicals. As the catalyst of the Fenton reaction, the cellular labile iron pool plays a crucial role in determining cellular susceptibility to ferroptosis. These lipid radicals propagate peroxidation to neighboring PL-containing polyunsaturated fatty acid (PUFA) chains which can then react with oxygen to form lipid peroxyl radicals. This chain reaction amplifies PL peroxidation and ultimately leads to ferroptosis.
Cellular iron homeostasis is exquisitely regulated by iron regulatory protein-1 (IRP1) and IRP2 that control the level and activity of a series of protein factors involved in iron import, export, storage, and release which coordinately maintain cellular iron homeostasis.40, 41 As expected, these protein factors can also modulate ferroptosis.3, 42 For example, a major mechanism for cellular iron uptake is mediated by transferrin and its receptor, TFRC, which mediates uptake of iron-bound transferrin that is required for ferroptosis induced by cysteine deprivation.43 Plasma membrane expression of TFRC was shown to be significantly increased during ferroptosis and might serve as a specific ferroptosis marker.44 Additionally, non-transferrin-bound iron can be imported via encoded transmembrane proteins SLC39A8/ZIP8 and SLC39A14/ZIP14. ZIP14-mediated iron uptake has been reported to promote ferroptotic liver injury.45 An excessive amount of cellular iron can be exported by iron exporters such as ferroportin and prominin, and downregulation of these exporters has been shown to promote ferroptosis.46, 47 Inside of cells, excess iron is mainly stored in a non-toxic form by the protein complex ferritin. As such, cytosolic ferritin and iron chaperone poly(rC)-binding protein 1 (PCBP1) confer resistance to ferroptosis by limiting iron availability.48, 49 Conversely, iron can be released from ferritin through nuclear receptor coactivator 4 (NCOA4)-mediated ferritin-selective autophagy, known as ferritinophagy, which can increase labile iron content and increase the susceptibility of the cell to ferroptosis.50, 51
The maintenance of mitochondrial iron homeostasis is also crucial in regulating ferroptosis. Mitoferrin 1 (SLC25A37) and mitoferrin 2 (SLC25A28) are essential mitochondrial iron importers involved in heme and Fe–S biogenesis. Mitoferrin 2 has been shown to promote ferroptosis likely through increased mitochondria iron content.52 Mitochondrial Fe–S proteins are also involved in ferroptosis regulation,53 and can prevent cancer cells from undergoing ferroptosis, probably through limiting mitochondrial iron level.54, 55 In contrast, heme oxygenase 1 (HO1), a mitochondrial enzyme that degrades heme to produce ferrous iron, leads to mitochondrial iron overload and sensitizes cancer cells to ferroptosis.56, 57 However, it was also reported that mild upregulation of HO1 protected cells from ferroptosis,58 suggesting complex and a likely context-dependent role of HO1 in ferroptosis.
Metabolic Pathways
Ferroptosis can be considered as a natural outcome of cellular metabolism. Phospholipid(PL)-PUFA, a product of lipid metabolism and essential component of cell membranes, is the substrate for PL peroxidation, arguably the executing step of ferroptosis. On the other hand, ROS and free radicals, which initiate PL peroxidation and provide the initial trigger for ferroptosis, are inevitable products of cellular metabolism, particularly energy metabolism. Accordingly, various metabolic processes have been demonstrated to be important in ferroptosis of which examples are provided below. Glucose starvation, which mimics an energy stress condition, inhibits lipid peroxidation and ferroptotic cell death of cancer cells. Inactivation of AMPK (AMP-activated protein kinase), serves to inhibit acetyl-CoA carboxylase (ACC) and PUFA biosynthesis, and largely abolishes the protective effects of energy stress on ferroptosis.59, 60 Importantly, cancer cells with loss of function mutations of LKB1, a tumor suppressor and upstream AMPK activator, display high basal AMPK activity and show increased resistance to ferroptosis.60 Conversely, glucose uptake mediated by SLC2A1 (GLUT1) promotes glycolysis, pyruvate oxidation, tricarboxylic acid cycle (TCA), and fatty acid synthesis which ultimately facilitates ferroptosis in cancer cells.61 Mammalian target of rapamycin complex 1 (mTORC1) is a nutrient sensor activated by amino acids, energy, and growth factors.62 Oncogenic activation of the PI3K-AKT-mTOR signaling axis was shown to suppress ferroptosis in cancer cells by increasing cellular monounsaturated fatty acid (MUFA) content. Conversely, inhibition of PI3K-AKT-mTOR signaling can sensitize cancer cells to ferroptosis induction.63 Specifically, mTORC1 inhibition was shown to sensitize cancer cells to ferroptosis through decreasing GPX4 expression.64 However, another study showed that ATP-competitive mTOR inhibitors suppressed ferroptosis triggered by system Xc− inhibition or direct cystine deprivation which may be due to upregulated macropinocytosis of albumin that is an alternative source of cellular cysteine.65, 66 Therefore, the effect of mTORC1 on ferroptosis can be context dependent.
PL de novo synthesis and its remodeling process play significant roles in regulating ferroptosis sensitivity by determining the composition of the plasma membrane.67 Various factors in these pathways have been shown to increase ferroptosis sensitivity including the uptake of essential PUFA by CD36,68, 69 the synthesis of long-chain PUFA by FADS1, FADS2, ELOVL5,70, 71 the activation of PUFA by ACSL4,72–74 as well as the incorporation of PUFA-CoA into lyso-PL by LPCAT3.72 Previous studies have demonstrated that exogenous MUFA75 or de novo synthesized MUFA inhibits ferroptosis.63 Membrane Bound O-Acyltransferase Domain Containing 1 and 2 (MBOAT1/2) selectively incorporates MUFA-CoA into lyso-phosphatidyl ethanolamine (PE), competitively inhibiting the synthesis of PE-PUFA. MBOAT1/2 suppresses ferroptosis in a GPX4-independent manner. Furthermore, the regulation of MBOAT1 and 2 is mediated by estrogen receptor and androgen receptor signaling, respectively, making them critical therapeutic targets in certain cancer types.16
NADPH levels play a crucial role in sustaining redox homeostasis and cell survival.76 Cellular NADPH abundance serves as a biomarker for predicting sensitivity to ferroptosis across different cancer cell lines.77 NADPH can be produced through various metabolic pathways, including the pentose phosphate pathway and NAD kinase (NADK)-mediated phosphorylation of NAD. Suppressing NADK reduces intracellular NADPH levels and increases cell sensitivity to ferroptosis.77 Conversely, overexpression of MESH1, a cytosolic NADPH phosphatase that degrades NADPH into NADH, depletes cellular NADPH and sensitizes cells to ferroptosis.78
Mitochondrial Metabolism
Mitochondria play a critical role in in cell homeostasis and cellular energy metabolism in eukaryotic cells. While mitochondria provide intracellular ATP via oxidative phosphorylation, they also function as key mediators of various forms of regulated cell death including apoptosis, pyroptosis, necroptosis, and ferroptosis. The involvement of mitochondria in ferroptosis was suggested based on the morphological changes observed in ferroptotic cancer cells and that events associated with mitochondrial energy metabolism promote ferroptosis.14, 43, 79 Accumulating evidence suggests that an impaired ferroptotic response is associated with changes in mitochondrial function. Mitochondrial ROS and the release of mitochondrial DAMPs contribute to inflammation in ulcerative colitis (UC).80 Furthermore, Paneth cells are highly susceptible to mitochondrial dysfunction in Crohn’s disease (CD).
To date, the role of mitochondria in regulating ferroptosis is poorly understood. Mitochondrial activity is crucial for cysteine-deprivation-induced (CDI) ferroptosis in contrast to RSL3-induced ferroptosis.43, 79 CDI ferroptosis fails to occur in the absence of glutamine since glutamine metabolism fuels the mitochondrial TCA cycle to enhance generation of mitochondria-derived ROS, which are necessary for lipid peroxidation during CDI ferroptosis. Metabolite intermediates of the TCA cycle such as α-ketoglutarate, fumarate, succinate, and malate can replace glutamine to induce CDI ferroptosis.79 Inhibiting the TCA cycle or the electron transfer chain mitigates CDI ferroptosis.79 As mitochondria play a central role in ferroptosis initiation, cells have developed potential ferroptosis suppressing mechanisms localized in the organelle, such as that mediated by DHODH and its product, reduced CoQ10.37
Mitochondria are the center of iron metabolism and energy production, leading to altered lipid peroxidation sensitivity. Mitochondria can modulate ferroptosis through mechanisms including the synthesis of Fe-S clusters. Additionally, mitochondrial Ca2+ plays a crucial role in triggering ferroptosis. Reducing Ca2+ influx can protect cells from ferroptosis induced by system Xc− inhibitors such as erastin and sulfasalazine.81 Recent studies have reported that FUNDC2 regulates ferroptosis by interacting with the mitochondrial glutathione transporter SLC25A11 to negatively regulate mitochondrial GSH levels.82 It has also been shown that FUNDC2 contributes to cardiomyopathy induced by the anthracycline doxorubicin through ferroptosis.82 Betaoxidation is generally believed to have a suppressive effect on ferroptosis by reducing the availability of unesterified PUFAs. The enzyme 2,4-dienoyl-CoA reductase 1 (DECR1), which is involved in PUFA beta-oxidation in the mitochondria, is overexpressed in prostate cancer. Knockout of DECR1 induces endoplasmic reticulum (ER) stress and sensitizes castration-resistant prostate cancer cells to ferroptosis both in vitro and in vivo. Furthermore, inhibiting beta-oxidation enhances ferroptosis in cancer cells.83, 84
As a major regulator of the antioxidant response, NRF2 plays a crucial role in regulating mitochondria respiration, controlling mitochondria-mediated ROS production, as well as the biosynthesis of glutathione and NADPH. It has been shown that NRF2 is a key determinant of the therapeutic response to ferroptosis-targeted therapies in HCC cells.12 Under oxidative stress, NRF2 is released from KEAP1-mediated ubiquitination. Stabilized NRF2 then accumulates in the nucleus, and transcriptionally activates a panel of genes that are involved in countering oxidative stress, including but not limited to SLC7A11 (cystine transporter); G6PD and PGD (NADPH regeneration); ferritin and ferroportin (iron regulation); FSP1 and DHFR (antioxidant generation), which serves as feedback regulation to maintain cellular redox homeostasis.85
Lipid peroxidation
Lipid metabolic enzymes ACSL4 and LPCAT3 have fundamental roles in GPX4-induced ferroptosis (Figure 1).86, 87 ACSL4 preferentially activates arachidonic acid (AA) to AA-CoA whereas LPCAT3 preferentially incorporates AA into membrane phospholipids.86, 87 Thus, inhibition of these enzymes prevents ferroptosis by reducing the pool of oxidation-sensitive fatty acids in cell membranes.86 PKCβII catalyzes the phosphorylation and activation of ACSL4, thus amplifying lipid peroxidation that leads to ferroptosis.88 Inhibition of PKCβII-ACSL4 blocks ferroptosis and limits ferroptosis-associated cancer immunotherapy.88 Ionizing radiation (IR) induces ferroptosis in cancer cells through increased levels of ROS and upregulation of ACSL4, leading to increased lipid peroxidation. Downregulation of ACSL4 blocks IR-induced ferroptosis and enhances radioresistance.24 In the context of immunotherapy, interferon gamma (IFN-γ) produced by CD8+ T cells and AA upregulate ACSL4 leading to lipid peroxidation and immunogenic tumor cell ferroptosis.89 Whereas high levels of AA promote tumor ferroptosis, downregulation of ACSL4 enhances tumor progression.89 Importantly, low-dose AA administration was shown to enhance the therapeutic efficacy of PD-L1 blockade and improve anti-tumor T cell responses through the IFN-γ signaling pathway.89
Ferroptosis in gastrointestinal disease
Common ferroptotic mechanisms in intestinal as well as other diseases include GPX4 inhibition, system Xc− suppression, lipid peroxide accumulation, and iron overload. Key regulators such as GPX4, SLC7A11, ACSL4, and p53 are also important for mediating ferroptosis-associated intestinal diseases. To date, the role of ferroptosis in gastrointestinal (GI) disease remains poorly characterized, and further research is needed to identify disease-specific ferroptotic mechanisms that may enable development of disease context-dependent therapeutic approaches. Studies have found correlations between ferroptosis and other forms of cell death in intestinal diseases that may share common pathways and key regulators, and may guide therapeutic interventions.
Iron overload is an important trigger of ferroptosis that can exacerbate intestinal inflammation. Regulation of iron absorption in enterocytes is dependent on the liver secreted hormone hepcidin that forms a complex with ferroportin. When this complex enters iron-absorptive enterocytes, ferroportin is degraded resulting in iron accumulation with reduced delivery to plasma.90 Excess intracellular iron that is unbound to ferritin, i.e., labile cellular iron, has the potential to increase inflammation and lipid peroxidation resulting in cell death, including ferroptosis. Levels of hepcidin are influenced by gut microbiota, thus affecting iron uptake.91 Iron overload can disrupt the gut microbiome to promote the growth of potentially pathogenic bacteria,90 and resultant dysbiosis can lead to enhanced ferroptosis in intestinal epithelial cells. While it has been reported that iron supplementation alters gut microbial homeostasis and exacerbates intestinal inflammation92, the specific mechanisms by which microbiota can affect ferroptosis await further study.
The intestinal epithelium is one of the fastest-renewing tissues in the human body. This rapid turnover requires significant metabolic activity and energy generation leading to increased ROS, iron metabolism,93 and increase susceptibility to ferroptosis. Ferroptosis has been linked to disease generation through intestinal epithelial cell death mediated by ER stress.92 Inflammatory responses triggered by ferroptosis can promote immunogenicity via activation of NF-kB by DAMPs.94 Furthermore, lipid peroxidation products stimulate phagocytosis (via TLR2 by oxidized phospholipid) and recruit macrophages by secreting chemokines.95 While these data suggest that inhibition of ferroptosis may be therapeutically beneficial, acquired resistance to ferroptosis with prevention of cell death has been shown to enhance neoplastic development and progression.96 Although beyond the scope of this review, other forms of cell death distinct from ferroptosis occur in GI diseases such as apoptosis, necrosis, necroptosis and pyroptosis. Molecular cross-talk has been described between these forms of cell death and ferroptosis, and multiple forms of cell death can be observed in the context of the same disease.97 The relative proportion each mechanism of cell death and its specific contribution to a disease entity awaits further research.98–100
Inflammatory Bowel Disease
Ferroptosis appears to play a role in inflammation-related conditions such as inflammatory bowel disease (IBD), and has been implicated in both experimental colitis and in human IBD. IBD is characterized by chronic and progressive inflammation of the GI mucosa which has been shown to manifest features of ferroptosis including iron deposition, lipid peroxidation, and GPX4 inactivation.92 Ferroptosis was shown to regulate colitis in the dextran sulphate sodium (DSS)-induced model of colitis which showed increased lipid peroxidation.92, 101 Furthermore, excess iron can increase mucosal ROS production which was shown to exacerbate intestinal inflammation in the DSS-induced colitis model.92 Inhibition of ferroptosis was shown to reduce disease activity scores in DSS-induced colitis.102
ER stress signaling can mediate cell death signaling including ferroptosis, that was significantly increased in intestinal epithelial cells (IECs) from patients with UC and in mice with experimentally-induced colitis.92, 102 Ferroptosis is recognized as an important contributor to IEC death in both humans and mice with UC.14, 92 In IEC-6 cells treated with H2O2, downregulation of phosphorylated STAT3 was observed which could be reactivated by the ferroptosis inhibitor, Fer-1.103 Phosphorylated NF-κBp65 can inhibit ER stress signaling by binding eukaryotic initiation factor 2α,92 and deletion of NF-κBp65 in IECs was shown to upregulate ferroptosis and to exacerbate colitis. Iron overload has also been observed to induce colitis, enhance ferroptosis, and significantly alter the microbiota in mouse models resulting in modulation of the expression of several ferroptosis-related genes such as SLC7A11 and GPX4.104 GPX4 plays an important role by protecting against lipid peroxidation and ferroptotic cell death102. Of note, IECs derived from patients with CD were shown to exhibit impaired GPX4 activity and signs of lipid peroxidation.105 PUFAs, and specifically AA, were shown to trigger a cytokine response in IECs which was restricted by GPX4.105 Furthermore, a PUFA-enriched Western diet was shown to induce a granuloma-like neutrophilic enteritis in mice that lacked one allele of Gpx4 in IECs, indicating that dietary PUFAs can serve as a trigger of GPX4-restricted mucosal inflammation with a phenotype of human CD. Together, these data indicate that ferroptosis may contribute to IBD via ER stress-mediated IEC cell death, and suggest that ferroptosis is a potential therapeutic target in IBD.92
The purported role of ferroptosis in IBD is believed to be linked to altered short-chain fatty acid (SCFA) metabolism.106 As previously stated, RSL3- and erastin-induced cell ferroptosis were shown to be enhanced by sodium butyrate. Specifically, sodium butyrate mediates the downregulation of anti-ferroptosis proteins, SLC7A11 and GPX4, thus acting as a pro-ferroptotic SCFA.25 Downregulation of these anti-ferroptotic proteins is mediated by the FFAR2-AKT-NRF2 and the FFAR2-mTORC1 axes.25 Butyrate is a naturally occurring SCFA in the gut produced as a result of the microbial metabolism from dietary fiber. Butyrate is known to play a role in intestinal homeostasis and immune and epithelial barrier function. Butyrate is the primary energy source of IECs107 and its mitochondrial oxidation accounts for over 70% of the oxygen consumed in IECs.106 In DSS-induced mice, iron chelation by deferasirox treatment strongly reversed the alterations caused by ferroptosis. Deferasirox was shown to significantly reduce DSS-induced UC in mice in association with a reduction in the level of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α and INF-γ)108. Furthermore, deferasirox treatment reshaped the composition of intestinal microbiota, and metabolomics analysis indicated the SCFAs production was enhanced in deferasirox-treated mice.
Targeting ferroptosis for therapeutic advantage has been demonstrated in animal models of colitis where inhibiting ferroptosis can attenuate intestinal injury in IBD. Suppression of ferroptosis was been shown to ameliorate DSS-induced colitis by blocking Nrf2/HO-1 signaling.101 Inhibition of ferroptosis by ferrostatin-1 was shown to alleviate colitis in the TNBS (trinitrobenzenesulfonic acid)-induced murine model.102 In addition, a potent ROS scavenger (pH-sensitive molybdenum-based polyoxometalate nanocluster) was shown to attenuate ferroptosis and reduce inflammatory indicators in DSS-induced mouse models of UC.109 Lipid peroxidation and elevated ferroptotic markers were also detected in the AOM/DSS (azoxymethane/DSS)-induced mouse model of colitis-associated carcinogenesis (CAC). In this model, treatment with ferrostatin-1 was shown to increase the incidence of CAC induced by a high fat diet, while induction of ferroptosis with RSL3 partially reversed this effect.110 It should be noted that while ferrostatin-1 has been shown to inhibit ferroptosis,14 its mechanism of action relies on preventing the accumulation of lipid peroxides (not a specific protein target) which hinders the potential to further improve the potency of the molecule.111 While ferrostatin has been used to inhibit ferroptosis, concerns exists regarding its efficacy as a ferroptosis inhibitor in vivo.
Ferrostatin-1 is unstable in plasma and has low blood-brain barrier permeability which underscores the need for alternative inhibitors with improved pharmacokinetic characteristics.111 In fact, there are already ferrostatin-1 analogs like liproxstatin-1 and UAMC-2418, that have better pharmacokinetic properties and outperform other inhibitors of ferroptosis in vivo.112
Ferroptosis in liver disease
Several forms of programmed cell death including apoptosis, necroptosis, and ferroptosis have been implicated in the pathogenesis of various liver diseases. Ferroptosis appears to be highly context dependent which is further supported by its involvement in conditions affecting the liver. With respect to liver injury and non-neoplastic conditions, normal liver cells are susceptible to ferroptosis whereas liver cancer cells display intrinsic or acquired resistance to ferroptosis. Accumulating evidence suggests that ferroptosis may serve as a promising target for the prevention and treatment of many forms of liver disease.
Ferroptosis has been linked to an expanding number of hepatic metabolic pathways113 including the NADPH pathway,114 the metabolism of fatty acids,115 and amino acids such as cysteine.116 Excessive iron regulates ferroptosis, and iron metabolism is primarily regulated by the liver which produces transferrin and hepcidin to regulate dietary iron absorption.117 Lower levels of these iron regulators have been found in patients with cirrhosis and liver fibrosis compared to healthy controls.45, 118 In animal models, hepatocyte-specific knockout of the transferrin gene Trf coupled with a high iron diet was shown to sensitize to ferroptosis-induced liver fibrosis that could be rescued with ferrostatin-1.45 Recently, the antioxidant vitamin E has been proposed to limit ferroptosis in the liver and to decrease hepcidin levels leading to iron depletion.119 However, it remains unknown as to whether abnormal levels of iron regulators such as hepcidin are a cause or a consequence of deregulated iron homoeostasis.117
Acute and chronic liver injury
Acute liver injury frequently results from hepatotoxic agents like drugs, alcohol, ischemic injury, or viral infections.120 Acetaminophen is a notable example, with studies indicating that ferroptosis inhibitors can moderately protect against acetaminophen-induced liver damage in mouse hepatocytes.121 Patients with alcoholic liver disease (ALD) show ethanol-induced upregulation of the transferrin receptor, TFRC, in hepatocytes leading to iron overload.122, 123 Chronic alcohol consumption is also associated with downregulation of hepcidin resulting in increased iron transport and absorption in the intestine.124 Iron overload leads to oxidative stress and hepatocyte ferroptosis, and both iron chelators and ferroptosis inhibitors can protect against ALD.123 Moreover, ethanol has been shown to inhibit mitochondrial frataxin expression, a key player in iron homeostasis whose downregulation leads to enhanced sensitivity to ethanol-induced ferroptosis, as do increased levels of ACSL4 and reduced system Xc- and GPX4.125 Restoring mitochondrial frataxin expression was shown to decrease sensitivity to ethanol-induced ferroptosis. Alcohol exposure also results in overexpression of fibronectin type III domain-containing protein 3B (FNDC3B) whose deletion in hepatocytes results in ethanol-induced steatosis through the inhibition of the AMPK pathway. Inhibition of AMPK downregulates transferrin expression leading to iron overload and predisposes to increased lipid peroxidation and ferroptosis. 126 Other forms of cell death are also involved in alcohol-induced hepatotoxicity, including apoptosis and autophagy.125
Lipin-1, a lipid metabolic enzyme expressed in adipose tissue, is overexpressed in animal models of alcoholic steatohepatitis and was shown to enhance hepatic ferroptosis through the inhibition of adiponectin signaling. 127 Aberrant liver sirtuin 1 (SIRT1) is also implicated in the pathogenesis of ALD. Animal models of ALD with intestine-specific knock-out of SIRT1 show reduced hepatic inflammation and liver injury, at least partially due to attenuated ferroptosis in the liver.128 In mouse models of hepatocyte injury and acute-on-chronic liver failure (ACLF) which has been associated with ferroptosis, Nrf2 activation ameliorates liver injury and protects against hepatotoxicity through inhibition of ferroptosis and inflammation. 129 Both ferroptosis and necroptosis have been found to contribute to ACLF through YAP signaling, and inhibition of the YAP pathway results in reduced liver fibrosis partially through inhibition of ferroptosis resulting from upregulation of ACSL4 and downregulation of SLC7A11.130 Of note, YAP levels in plasma in patients with ACLF are increased compared to healthy controls.130 Given the accumulated evidence linking ethanol consumption with ferroptosis in the liver, therapeutic strategies have the potential to reduce ethanol-related liver injury, although the precise molecular mechanisms await further study.
Viral hepatitis
Infection with hepatitis B and hepatitis C are associated with development of liver fibrosis and in the setting of cirrhosis, can lead to hepatocellular carcinoma (HCC).131 Exosomes derived from hepatocytes infected with hepatitis B virus were shown to activate hepatic stellate cells and to promote fibrosis through the miR-222/TFRC axis.132, 133 In HCC cells, inhibition of miR-142–3p or overexpression of SCL3A2 were shown to inhibit ferroptosis and to reduce cell proliferation, migration and invasion.133 Other studies have revealed that arginine methyltransferase 9 (PRMT9) expression is promoted by the HBV X protein (HBx), and targets the heat shock protein family A member 8 (HSPA8) for arginine methylation resulting in upregulation of CD44 which, in turn, inhibits ferroptosis and promotes HCC progression.134, 135 Low doses of selenium have been shown to inhibit the hepatotoxicity of HBx through GPX4-mediated ferroptosis inhibition in cell lines and in mouse models.136 The enzyme fatty acid desaturase 2 (FADS2), which promotes lipid peroxidation, has emerged as a rate-limiting factor in HCV replication, indicating a potential role of ferroptosis in hepatitis C infection. In hepatitis C-infected cells, overexpression of FADS2 was found to inhibit viral replication and sensitize cells to ferroptosis.137
Metabolic dysfunction-associated steatotic liver disease
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an increasingly prevalent condition that typically begins with steatosis and can progress to fibrosis and ultimately cirrhosis.138 Many forms of cell death are involved in MASLD, including ferroptosis.139 In patients with metabolic dysfunction-associated steatohepatitis (MASH), elevated end products of lipid peroxidation and iron overload are frequently observed, and iron overload correlates with the histological severity of the liver disease.139 Additionally, arachidonate 12-lipoxygenase (ALOX-12), particularly in its interaction with acetyl-CoA carboxylase 1 (ACC1), has been found to promote the progression of MASH.140 Murine models of MASH show that both a high fat diet and iron overload induce lipophagy and ferritinophagy, respectively, and synergize in promoting ferroptosis that results in lobular inflammation and increased fibrosis.141 Ferroptosis was shown to aggravate MASH progression whereas inhibition of ferroptosis decreased disease severity in a mouse model of MASLD induced by a high-fat diet.142 Treatment with melatonin, a potent antioxidant, was shown to reduce hepatocyte ferroptosis by inhibition of ER stress through the MT2/cAMP/PKA/IRE1 pathway.138 These studies suggest that inhibition of ferroptosis may be a potential therapeutic approach to reduce or prevent liver damage in patients with MASLD.
Hepatic fibrosis
A key step in development of liver fibrosis is the transdifferentiation of hepatic stellate cells (HSCs) into matrix-producing myofibroblasts.143 In this context, regulators of ferroptosis in HSCs include RNA-binding proteins ELAV-like protein 1 (ELAVL1)144 and zinc finger protein 36 (ZFP36).145 Upon exposure to ferroptosis-inducing compounds, ELAVL1 protein expression was increased through inhibition of the ubiquitin-proteasome pathway. While transfection with an ELAVL1 plasmid induced ferroptosis, ELAVL1 knockdown by siRNA led to ferroptosis resistance. Upregulated ELAVL1 expression also appeared to increase autophagosome formation and autophagic flux, which appeared to be the underlying mechanism for ELAVL1-enhanced ferroptosis.144 In mice, treatment with sorafenib attenuated murine liver fibrosis by inducing HSC ferroptosis which was impaired by HSC-specific knockdown of ELAVL1. Similarly and upon exposure to ferroptosis-inducing compounds, a ubiquitin ligase (FBXW7/CDC4) decreased expression of the RNA binding protein ZFP36 whereas the ZFP36 plasmid impaired FBXW7 plasmid-induced ferroptosis in HSCs and inhibited induction of autophagy.145 In mice, HSC-specific overexpression of ZFP36 impaired erastin- or sorafenib-induced ferroptosis. Together, these results identify ELAVL1 and ZFP36 in the induction of autophagy-dependent ferroptosis and as potential targets for the treatment of liver fibrosis, although selective induction of ferroptosis in HSCs is needed with sparing of hepatocytes.
Hemochromatosis
Hereditary hemochromatosis is an iron-overload disease caused by mutations in genes involved in iron absorption.146 Excessive iron is absorbed by the intestine and deposited in parenchymal cells leading to tissue damage and organ failure. Apart from genetic factors, environmental factors such as alcohol intake and blood loss can also influence iron accumulation. Excessive iron usually produces massive ROS through the Fenton reaction and subsequently leads to DNA damage and tissue injury. Treatment with iron was shown to induce ferroptosis in murine primary hepatocytes, and ferroptosis was also observed in mice fed a high-iron diet or in mouse models of hereditary hemochromatosis with severe iron overload, but not in mice with only mild iron overload.114 Importantly, iron overload–induced liver damage was rescued by inhibition of ferroptosis by ferrostatin-1. Genes found to be significantly up-regulated in iron-treated cells and hemochromatosis include SLC7A11, a known ferroptosis-related gene, whose genetic deletion was insufficient to induce ferroptosis unless SLC7A11−/− mice were fed a high iron diet. Data indicate that iron can upregulate SLC7A11 expression through the ROS–Nrf2–antioxidant response element axis which may be a potential compensatory mechanism to protect against iron overload-induced ferroptosis in hemochromatosis. In addition, SLC7A11 was shown to confer protection against ferroptosis during iron overload by uptaking cystine and reducing ROS production. In these models, iron-induced ferroptosis was not mediated by ER stress, the mitogen-activated protein kinase (MAPK) pathway, or autophagy.114 These results suggest that ferroptosis may be a target for treating hemochromatosis-related tissue damage.
Ferroptosis and cancer
Ferroptosis has multiple implications in tumor development. First, several cancer signaling pathways are involved in the regulation of ferroptotic cell death.147 Moreover, cancer cells with dysregulated metabolism, a high accumulation of ROS, and specific mutations render some tumor cells more vulnerable to ferroptosis, and thus, create a therapeutic window for ferroptosis inducers as a therapeutic strategy.147 Importantly, ferroptosis is triggered by several conventional cancer therapies such as chemotherapy, radiotherapy, immunotherapy, and targeted therapy.147
Cancer-related pathways in ferroptosis
Several alterations in lipid metabolism have been described in cancer cells148 including those that contribute to ferroptosis resistance such as activating mutations of PI3K or loss of PTEN function through downstream SREBP1/SCD1-mediated lipogenesis.63 Furthermore, activation of the p62-Keap1-NRF2 pathway confers resistance to ferroptosis in HCC cells.12 Genes regulated by NRF2, including NQO1, HO1, and FTH1, modify both iron metabolism and lipid peroxidation to confer ferroptosis resistance. Inhibition of the PI3K-AKT-mTOR signaling axis sensitizes cancer cells to ferroptosis induction with therapeutic potential through mTORC1 inhibition.63 SLC47A1 acts as a phospholipid transporter149 and can block ferroptosis through the inhibition of the ACSL4-SOART1 axis that leads to lipid peroxidation. Targeting SLC47A1 sensitizes tumor cells to ferroptosis induction and may be a strategy for tumor suppression and overcoming drug resistance.149 Studies indicate that SLC27A5/FATP5, an enzyme involved in the metabolism of fatty acids and bile acids, is downregulated in HCC cells that are resistant to the multikinase inhibitor, sorafenib. SLC27A5 deficiency facilitates this resistance by suppressing ferroptosis. Mechanistically, loss of SLC27A5 enhances glutathione reductase expression in a NRF2dependent manner, and renders HCC cells insensitive to sorafenib-induced ferroptosis in vitro and in vivo.150
TP53 acts as a metabolic regulator and inhibits cysteine uptake which sensitizes cells to ferroptosis through transcriptional repression of SLC7A11.151 SAT1 expression, which is transcriptionally regulated by p53, can trigger lipid peroxidation and sensitize cells to ROS-induced ferroptosis.152 Moreover, SAT1 increases expression of ALOX15 which catalyzes peroxidation of AA, and is a key metabolic regulator in turning oxidative stress into lipid peroxidation.153 SAT1 expression is downregulated in human tumors possibly contributing to resistance to ferroptosis, and inhibition of ALOX15 abrogates SAT1-induced ferroptosis.152 On the other hand, p53 has also been shown to promote ferroptosis resistance such as via transcriptionally upregulating p21 and thus, decreasing cellular metabolism154or via non-transcriptionally blocking the activity of dipeptidyl peptidase-4 (DPP4), which is involved in lipid peroxidation through unknown mechanisms .155
Intercellular interactions mediated by E-cadherin regulate ferroptosis through the intracellular NF2-Hippo signaling pathway. Inhibition of this axis leads to YAP signaling upregulation of ferroptosis modulators, such as ACSL4 and TFRC.156 As alterations in the cadherin-NF2-Hippo-YAP signaling axis are common in cancer and can sensitize to ferroptosis, this signaling axis may be therapeutically targeted.156 Furthermore, because of the role of YAP signaling in epithelial-mesenchymal transition (EMT), the function of YAP to promote ferroptosis provides a mechanism explaining why therapy-resistant mesenchymal tumor cells are often more susceptible to ferroptosis induction and are addicted to GPX4 activity.29 Moreover, dependency on GPX4 is correlated with high level expression of ZEB1 in therapy-resistant tumor cells,29 which links mesenchymal gene expression with lipid metabolism via direct regulation of the transcription of PPARγ and EMT-associated remodeling of the plasma membrane.29 Selective sensitivity of ZEB1 overexpressing cells to lipid-peroxide activity and ferroptosis can be achieved by targeted inhibition of GPX4 or GSH for therapeutic advantage.29 In addition, the RAS family of proto-oncogenes are among the more frequently mutated genes in cancers. Transcription of SLC7A11 is controlled by oncogenic RAS signaling through ETS-1 in a regulatory network that allows RASdriven tumors to evade ferroptosis.157 This creates an opportunity for targeting cystine uptake and GSH biosynthesis through pharmacological targeting of system Xc- in RAS-driven tumors, which has been shown to impair tumor growth in vivo.157
Ferroptosis and CRC
Colorectal cancer (CRC) is the third most common human cancer158 and ranks fourth as a cause of cancer-related death worldwide.158 Patients with metastatic disease are generally incurable, and drug resistance underlies treatment failure which underscores the need for new therapies to improve patient survival.
Regulation of ferroptosis in CRC
Tumor cell ferroptosis is a complex and context specific process. In CRC cells with intrinsic resistance to ferroptosis induction, the simultaneous inhibition of the GPX4-GSH axis and the FSP1-CoQ10 axis, still requires additional inhibitors of glycolysis or the Warburg effect to induce ferroptosis.159 In contrast to TP53 transcription-dependent induction of ferroptosis reported in other tumor cell types, TP53 can block erastin-induced ferroptosis in CRC cells through the inhibition of DPP4 activity in a transcription-independent manner by promoting its localization to the nucleus where it is inactive.155 Downregulation of TP53 enables DPP4-dependent lipid peroxidation that ultimately leads to ferroptosis.155 CRC cells can be protected from ferroptosis in a TP53-dependent manner by degradation of the RNA binding protein HNRNPC (heterogeneous nuclear ribonucleoprotein C) mediated by the E3 ubiquitin ligase, cullin-9.160 Downstream alternative splicing targets of HNRNPC include TP53 and SLC47A1 mRNA.160 The TP53-induced glycolysis and apoptosis regulator (TIGAR) is overexpressed in CRC, and its activity increases available NADPH to convert oxidized glutathione to GSH and to reduce ROS levels.161 Downregulation of TIGAR sensitizes CRC cells to erastin-induced ferroptosis via increased intracellular ROS, activation of AMPK, and downregulation of SCD1.161 Hypoxia-inducible factor 2α (HIF-2α) is involved in CRC progression, and its activation triggers proinflammatory responses in epithelial cells,162 and also renders CRC cells vulnerable to ferroptosis through upregulation of lipid and iron regulatory genes.162
Butyrate not only has important effects in the induction of ferroptosis but also in preventing the development of CRC through the inhibition of the activation of mTOR1.25 In CRC cells, butyrate induces ferroptosis through both the increased expression of ACSL4, which increases the susceptibility of cells to lipid peroxidation,163 and reduction of system Xc- expression which limits cysteine availability.164 It should be noted that in CRC, system Xc- expression is higher compared to tumor-adjacent tissue.164 Butyrate has been shown to enhance the pro-ferroptotic effects of the cytotoxic chemotherapy drug oxaliplatin in CRC cells in vitro and in vivo.164 The combined intake of highly fermentable dietary fiber to enhance butyrate production and of omega-3 polyunsaturated fatty acids has been proposed to reduce the risk of CRC. These treatments can promote ferroptosis through GPX4 and mitochondrial metabolism pathways.107 The “butyrate paradox”, where butyrate enhances proliferation of normal cell but not of cancer cells, also shows the context dependent nature of its metabolism.25
Several genes and RNA molecules with altered expression in CRC have been associated with ferroptosis regulation and tumor development.158 Furthermore, many ferroptosis-related gene signatures, deposited in public databases, have been proposed to prognosticate patients with CRC,158 however, limitations exist and there is the need for validation. These signatures have the potential to identify important regulatory pathways of ferroptosis in CRC that may lead to development of new therapeutic strategies.
Therapeutic strategies involving ferroptosis in CRC
Induction of ferroptosis is a potential approach for the treatment of CRC, and several drugs have been associated with this form of RCD.158 A subset of cancer cells may avoid therapy-induced cell death by switching to a slow, yet reversible proliferation state that is drug tolerant. Drug-tolerant persistent CRC models show increased GPX4 expression making them vulnerable to GPX4 inhibitors,165 but more potent and bioavailable compounds to achieve GPX4 inhibition are desirable.165 Overexpression of lipocalin 2 (LCN2), involved in the regulation of iron homeostasis, in CRC cells promotes chemoresistance to 5-fluorouracil (5-FU) through the inhibition of ferroptosis.166 Conversely, inhibition of LCN2 using monoclonal antibodies was shown to sensitize cells to chemotherapy and to inhibit tumor growth.166 However, LCN2 was also shown to inhibit the NF-κB/SNAIL pathway and to prevent EMT and metastasis of CRC. Conflicting data exist in that silencing of LCN2 was shown to promote tumor growth167 and thus, the role of LCN2 in CRC progression remains controversial. It is established that KRAS mutated CRC cell lines are resistant to therapy with EGFR inhibitors, including cetuximab.168 Inhibition of the Nrf2/HO-1 axis by cetuximab has been shown to promote ferroptosis induced by RSL3 in KRAS-mutated CRC cells.168
In the context of clinical application for cancer therapy, ferroptosis can be targeted by exploiting cancer cell regulatory alterations that result in enhanced metabolism to sustain increased requirement for energy and metabolite synthesis. These changes can make cancer cells more susceptible to ferroptosis (for example, by being more dependent on GPX4 activity or other ferroptosis surveillance mechanism) creating therapeutic windows for ferroptosis inducers in specific cancers. Cancer cells that have acquired resistance to ferroptosis can be re-sensitized by targeting the specific mechanisms that protect them. Furthermore, ferroptosis inducers can sensitize cancer cells to conventional anti-cancer therapies or overcome acquired resistance to thereby, improve efficacy.147
The roles of ferroptosis in the tumor microenvironment and immunotherapy
The pros of ferroptosis in immunotherapy
The release of DAMPs is one of the major features of immunogenic cell death which activates the immune system and synergizes with immune checkpoint blockade (ICB). HMGB1 is a DAMP that is released by ferroptotic cells in an autophagy-dependent manner. Of note, the ATG5/ATG7 autophagic axis is necessary for the release of acetylated HMGB1 during ferroptosis169. Other immunogenic DAMPs, including ATP, CRT, DCN were also confirmed to be released by ferroptotic cells.170
Moreover, recent studies suggested that CD8+T cells can kill cancer cells via ferroptosis. IFN-γ secreted by activated CD8+ T cells downregulates the expression of SLC7A11 in cancer cells by JAK-STAT1 pathway and sensitizes cancer cells to ferroptosis.171 IFN-γ in combination with AA can further promote ferroptosis in cancer cells. IFN-γ upregulates ACSL4 expression through the STAT1/IRF1 signaling pathway in cancer cells, promotes the incorporation of AA into phospholipids, and sensitizes cancer cells to ferroptosis. Combining AA and ICBs can synergistically inhibit tumor growth through IFN-γ.89 These studies have indicated a synergistic effect between ferroptosis induction and ICBs, representing a potential treatment strategy.
The cons of ferroptosis in immunotherapy
Chronic inflammation can lead to tumor initiation, growth and progression by providing a tumor-supportive microenvironment.172 Tumor cells release proinflammatory molecules that promote immune evasion, angiogenesis, and growth of cancer stem cells.8, 172 Several immunosuppressive cell types have been identified in the TME, including myeloid-derived suppressor cells (MDSC), tumor-associated macrophages (TAMs), and regulatory T cells (Tregs)173 which may participate in ferroptosis-related immunosuppression. Although induction of ferroptosis to target tumors resistant to conventional therapies is promising, the resulting release of proinflammatory signals can have a negative impact on ferroptosis-based therapies for cancer. Cancer cell immune evasion involves mechanisms including macrophage polarization, impaired immune cell cytotoxicity, and upregulation of immunosuppressive cells.8, 172 Importantly, ferroptosis plays a role in all of these mechanisms in the TME (Figure 2).
Figure 2.
Involvement of ferroptosis in the tumor microenvironment and modulation of immune responses. Tumor cell ferroptosis promotes the activation of immunosuppressive cells and impairs the activity of anti-tumor immune cells. Immunosuppressive Tregs and MDSCs are protected from ferroptosis through upregulation of GPX4 and ASAH2, respectively. Downregulation of Acyl CoA dehydrogenases (ACADS), and exposure to HMGB1, KRASG12D, PGE2 and 8-OHG resulting from tumor cell ferroptosis in the TME leads to macrophage polarization with a M2 phenotype. TAMs release of proinflammatory TNF is dependent on HMGB1 interaction with the advanced glycosylation end-product specific receptor (AGER) receptor. The inflammatory environment reduces NOS2 expression in M2 macrophages and makes them more vulnerable to ferroptosis. Active CD8+ T cells release IFN-γ that impairs tumor cancer cell expression of system Xc- that results in ferroptotic cell death. Ferroptotic cancer cells increase the expression of COX2 that elaborates PGE2 which serves to inhibit immune cell (CD8+ T cells, DCs and NKs) activities. CD8+ T cells are led to ferroptosis through both cysteine depletion as a consequence of their inactive system Xc- and accumulation inside MDSC and by increased lipids in the TME that are uptaken through the CD36 membrane receptor. In DC cells, downregulation of system Xc- and increased lipids in the TME (activating the PPAR-γ receptor pathway) enhance ferroptosis and impair their activation of CD8+ T cells and their release of proinflammatory IL-6 and TNF. L-KYN release from ferroptotic cancer cells in the TME leads to ferroptosis of NK cells.
The release of HMGB1 has implications for immune responses and cancer, including the proinflammatory TME.8, 174 The binding of HMGB1 to toll-like receptor 4 (TLR4) and AGER, mediates immune responses such as the release of the cytokine tumor necrosis factor (TNF).169 Mutated KRASG12D acts as a DAMP during ferroptosis whose release following autophagy-dependent ferroptosis activates AGER and leads to macrophage polarization through STAT3-dependent fatty acid oxidation.175 Alternatively activated or M2 macrophages is associated with tumor progression due to their angiogenic 176 and immunosuppressive 177 activities. In mutated KRASG12D pancreatic tumors, tumorigenesis is promoted by depletion of GPX4 or high-iron diets leading to ferroptosis.178 The release of 8-hydroxyguanosine during ferroptosis activates a DNA-sensing pathway that regulates inflammation and immune responses, causing macrophage migration and activation178. Polarization of macrophages is also promoted by downregulation of ACADS enzyme which can also enhance CRC tumor growth.179 Interestingly, M2 macrophages are more susceptible to ferroptotic cell death than M1 macrophages.180 Upregulation of NOS2 in M1 macrophages protects them from lipid peroxidation, whereas downregulation of NOS2 is promoted in pro-inflammatory conditions including those of the TME.180
Treg cells are protected from ferroptosis by GPX4, sustaining their activation and anti-tumor immune activity in the TME.181 Treg-specific deletion of GPX4 limits tumor growth and enhances antitumor immune responses, such as production of proinflammatory cytokines including IL-1β.181 MDSCs, which inhibit T-cell mediated anti-tumor immunity, are protected from ferroptosis through N-acylsphingosine amidohydrolase (ASAH2) overexpression. ASAH2 targets and suppresses the p53/Hmox1 axis reducing lipid peroxidation and preventing ferroptosis.182 One of the mechanisms of MDSCs for blocking T-cell activation is the depletion of cysteine. T-cells lack both cystathionase (that converts methionine to cysteine) and a complete system Xc- (to import cystine), whereas MDSCs do express a functional system Xc- but not the ASC neutral amino acid transporter that exports cysteine.183 MDSCs block T-cell activation by sequestering cystine and limiting its availability, which leads to T-cell ferroptotic vulnerability in the setting of a TME that is enriched in MDSCs. Mouse models with T cells lacking GPX4 have reduced peripheral CD8+ T cells and impaired response to infection, whereas both CD4+ and CD8+ T-cells lacking GPX4 underwent ferroptotic cell death in vitro.184 Also, CD8+ T-cells are more sensitive to ferroptosis induction using inhibitors of GPX4 activity than are cancer cell lines.185 Membrane glycoprotein CD36 mediates ferroptosis of CD8+ T-cells through its fatty acid uptake activity, impairing CD8+ T-cells anti-tumor activity.69 Mechanistically, the uptake of oxidized low-density lipoproteins by CD36 induces p38 phosphorylation, and this activation limits secretion of TNF and IFN-γ (effectors of CD8+ T-cell immune activity) while also inducing lipid peroxidation and ferroptosis.186 Knockout of ACSL4 protects these cells from ferroptosis but impairs their antitumor activity, whereas overexpression of GPX4 or FSP1 also protects from ferroptosis while retaining their functionality.185 In fact, overexpression of GPX4 not only protects CD8+ T-cells from ferroptosis, but also enhances their immune activity.186 IFN-γ released from immunotherapy-activated CD8+ T-cells downregulates the expression of system Xc- in tumor cells, promoting lipid peroxidation and ferroptosis 171 while also sensitizing to radiotherapy.187 Since IFN-γ induces expression of PD-L1 and promotes tumor progression,188 the addition of immune checkpoint blockade should be considered as a potential combination therapy. Apolipoprotein L3 (APOL3) promotes ferroptosis of CRC cells and inhibits tumor growth through L-lactate dehydrogenase A (LDHA) ubiquitination and degradation in the proteasome.189 This pathway enhances the anti-tumor activity of CD8+ T-cells increasing the levels of IFN-γ and reducing lactic acid concentration.189 The combination of overexpression of APOL3, ferroptosis induction with RSL3, and inhibition of PD-L1 has a synergistic effect in CRC models.189
Other immune cell types have impaired cytotoxic activity in the TME. In this regard, the activity of natural killer (NK) cells is downregulated in the TME190 whereas activation of the NRF2 signaling pathway restores their anti-tumor activity.190 Dendritic cells (DCs) are antigen-presenting cells that are important for the activation of cytotoxic T lymphocytes.191 Moreover, DC functions are impaired by lipid peroxidation byproducts that cause ER stress.192 DC induction of ferroptosis can be achieved with the RSL3 inhibitor of GPX4, but not with the SLC7A11 inhibitor erastin (due to the already low expression of SLC7A11 in DCs).193 Mechanistically, RSL3-induced ferroptosis in DCs is dependent on peroxisome proliferator activated receptor gamma (PPAR-γ), acting as a key transcription factor for lipid metabolism.193 DCs undergoing ferroptosis lose their immune functions such as production of proinflammatory cytokines (TNF and IL-6) and activation of CD8+ T-cells.193 Moreover, engulfment of ferroptotic dying cancer cells by DCs not only impedes antigen cross-presentation in DC cells, but also impedes the immunogenicity of chemotherapy-induced apoptotic cells, resulting in decreased proliferative capacity of CTLs.194
Polymorphonuclear MDSCs (PMN-MDSCs) in the TME are highly sensitive to ferroptosis. Interestingly, spontaneous ferroptosis of PMN-MDSCs does not enhance anti-tumor immunity by reducing the number of PMN-MDSCs. Instead, it confers immunosuppressive activity on PMN-MDSCs. Ferroptotic PMN-MDSCs release PGE2 and oxidized phospholipids, influencing the activity of CD8+ T cells and TAMs and rendering a more immunosuppressive TME. Inhibition of ferroptosis in immunocompetent mice abrogates the immunosuppressive activity of PMN-MDSCs, particularly when combined with ICBs, and reduces tumor progression. This indicates that PMN-MDSC ferroptosis may be a targetable immunosuppressive mechanism for cancer immunotherapy.195 Overexpression of cyclooxygenase-2 (COX2) in CRC and other cancer cell types196 has been reported in cancer cells undergoing ferroptosis.15 COX-2 enzymatic activity is the rate limiting step in prostaglandin E2 (PGE2) synthesis whose membrane receptors can inhibit anti-tumor immune responses.197, 198 The COX-2-PGE2-EP axis suppresses activation of myeloid cells197 and the activity of DCs, NK cells and T-cells, while promoting polarization of TAMs to a M2 phenotype198 thereby contributing to tumor immune escape. Future studies are needed to clarify the immunomodulatory effects of ferroptosis, including its association with diseases outcomes. In addition, further study of ferroptosis-related signal transduction by different immune cell subpopulations is awaited.
Targeting tumor ferroptosis to improve the efficacy of immunotherapy
Due to the heterogeneity and high complexity of TME, ferroptosis inducing agents (FINs) can cause either immune active or immunosuppressive function in a TME-dependent manner. Therefore, it demands the development of FINs that selectively induce tumor ferroptosis and improve the efficacy of immunotherapy. SLC7A11 had been shown to be dispensable for T cells proliferation in vivo and for their antitumor immune responses. Consequently, tumor cell SLC7A11 loss acts synergistically with the immunotherapeutic agent anti–CTLA4, laying the foundation for utilizing specific SLC7A11 inhibitors to expand the efficacy of existing anticancer immunotherapeutics.199 Recently, a small molecule compound, N6F11 had been shown to selectively trigger degradation of GPX4 in tumor cells but not immune cells. N6F11 caused ferroptotic cancer cell death, initiated HMGB1-dependent antitumor immunity, and sensitized immune checkpoint blockade in advanced cancer models.200 Another study showed that deubiquitinase inhibitor PR-619 degraded GPX4 and increased the efficacy of anti-PD1 in a colon cancer model.201 These findings may establish a safe and efficient strategy to boost ferroptosis-driven antitumor immunity by targeting GPX4 selectively in tumor cells.
Conclusions
Ferroptosis is a form of metabolically regulated cell death. Data continue to emerge regarding ferroptotic regulatory pathways and the relationship between ferroptosis and pathophysiology of diseases including those affecting the gastrointestinal tract. Iron overload, ROS accumulation, lipid peroxidation and impaired antioxidant systems are critical steps in pathways regulating ferroptosis that are involved in GI diseases. While studies have used the accumulation of lipid peroxidation as an indicator of ferroptosis, this process can also be part of other types of RCD including apoptosis, necroptosis, or pyroptosis. Although much progress has been made in understanding pathological roles of ferroptosis, critical questions remain to enable development of ferroptosis-targeted therapies. Ferroptosis appears to have dual roles whereby inhibition of ferroptosis can alleviate intestinal damage whereas induction of ferroptosis has been shown to inhibit CRC cell migration and proliferation. There is an unmet need to identify specific biomarkers of ferroptosis and further research is required to detect ferroptotic mechanisms that are specific for individual disease entities. Therapeutic opportunities related to ferroptosis hold promise for therapy of multiple disease states. Cellular susceptibility to ferroptosis is dependent on the in vivo microenvironment, and vulnerability to ferroptosis among cancer cells, immune cells and within the TME awaits further study. It remains to be determined if we can activate ferroptosis specifically in cancer cells without affecting healthy cells. Studies suggest that ferroptosis plays an important role in tumor suppression as well as resistance to cancer therapy which can provide novel therapeutic opportunities. In conclusion, ferroptosis is a mechanistically and morphologically distinct form of RCD that has provided insights into pathophysiology of multiple disease entities. Elucidating the regulatory pathways related to ferroptosis can provide new opportunities for understanding pathophysiology of disease inlcuding cancer, and identifying novel targets and strategies for exploitation of ferroptosis for therapeutic advantage.
Acknowledgements
JJER is affiliated with Instituto de Investigación Biomédica de A Coruña (A Coruña, Spain). The authors extend their appreciation to Bahar Saberzadeh-Ardestani, M.D. for her very capable assistance with the preparation of the manuscript.
Grant support:
JJER is supported by Xunta de Galicia (Spain) grant for postdoctoral formation (budget code 06.A2.561A.481.0, record code IN606B-2022/012) with additional Fulbright program support (US Department of State). JX is supported by NIH R01CA204232, NIH R01CA258622, and NIH R01CA166413, and an NCI cancer center core grant P30 CA008748 to MSKCC.
Abbreviations:
- ACLF
acute-on-chronic liver failure
- ALD
alcoholic liver disease
- AOM
azoxymethane
- CAC
colitis-associated carcinogenesis
- CD
Chron’s disease
- CDI
cysteine-deprivation-induced
- CRC
colorectal cancer
- DAMPs
damage-associated molecular patterns
- DCs
dendritic cells
- DSS
dextran sulphate sodium
- ER
endoplasmic reticulum
- FIN
ferroptosis inducing agent
- 5-FU
5-fluorouracil
- GSH
glutathione
- HCC
hepatocellular carcinoma
- HSCs
hepatic stellate cells
- IBD
inflammatory bowel disease
- ICB
immune checkpoint blockade
- IECs
intestinal epithelial cells
- IFN-γ
interferon gamma
- IR
ionizing radiation
- MDR
multidrug resistant
- MDSC
myeloid-derived suppressor cell
- MUFA
monounsaturated fatty acid
- MASLD
Metabolic dysfunction-associated steatotic liver disease
- MASH
metabolic dysfunction-associated steatohepatitis
- NK
natural killer
- PL
phospholipid
- PMN
Polymorphonuclear
- PUFA
polyunsaturated fatty acid
- RCD
regulated cell death
- ROS
reactive oxygen species
- RTA
radical-trapping antioxidant
- SCFA
short-chain fatty acid
- TAMs
tumor associated macrophages
- Tregs
regulatory T cells
- TME
tumor microenvironment
- UC
ulcerative colitis
Footnotes
Disclosure:
LD is an inventor on a patent related to autophagy. JX is an inventor on patents related to autophagy and cell death; and holds equity of and consults for Exarta Therapeutics and Lime Therapeutics. Other authors disclose no conflicts.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018;25:486–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mou Y, Wang J, Wu J, et al. Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol 2019;12:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 2021;22:266–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chen X, Kang R, Kroemer G, et al. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol 2021;18:280–296. [DOI] [PubMed] [Google Scholar]
- 5.Magtanong L, Mueller GD, Williams KJ, et al. Context-dependent regulation of ferroptosis sensitivity. Cell Chem Biol 2022;29:1409–1418 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tang D, Kroemer G. Ferroptosis. Current Biology 2020;30:R1292–R1297. [DOI] [PubMed] [Google Scholar]
- 7.Chen X, Kang R, Kroemer G, et al. Ferroptosis in infection, inflammation, and immunity. J Exp Med 2021;218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bi Q, Sun ZJ, Wu JY, et al. Ferroptosis-Mediated Formation of Tumor-Promoting Immune Microenvironment. Front Oncol 2022;12:868639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Exp Mol Med 2022;54:91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Xu H, Ye D, Ren M, et al. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends Mol Med 2021;27:856–867. [DOI] [PubMed] [Google Scholar]
- 11.Osburn WO, Wakabayashi N, Misra V, et al. Nrf2 regulates an adaptive response protecting against oxidative damage following diquat-mediated formation of superoxide anion. Arch Biochem Biophys 2006;454:7–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sun X, Ou Z, Chen R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology 2016;63:173–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol 2019;23:101107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012;149:1060–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014;156:317–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liang D, Feng Y, Zandkarimi F, et al. Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell 2023;186:2748–2764 e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Jiao N, Wang L, Wang Y, et al. Cysteine Exerts an Essential Role in Maintaining Intestinal Integrity and Function Independent of Glutathione. Mol Nutr Food Res 2022;66:e2100728. [DOI] [PubMed] [Google Scholar]
- 18.Dixon SJ, Patel DN, Welsch M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife 2014;3:e02523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gout PW, Buckley AR, Simms CR, et al. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the x(c)- cystine transporter: a new action for an old drug. Leukemia 2001;15:1633–40. [DOI] [PubMed] [Google Scholar]
- 20.Zhang Y, Shi J, Liu X, et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat Cell Biol 2018;20:1181–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wu Y, Sun X, Song B, et al. MiR-375/SLC7A11 axis regulates oral squamous cell carcinoma proliferation and invasion. Cancer Med 2017;6:1686–1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Drayton RM, Dudziec E, Peter S, et al. Reduced expression of miRNA-27a modulates cisplatin resistance in bladder cancer by targeting the cystine/glutamate exchanger SLC7A11. Clin Cancer Res 2014;20:1990–2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Liu XX, Li XJ, Zhang B, et al. MicroRNA-26b is underexpressed in human breast cancer and induces cell apoptosis by targeting SLC7A11. FEBS Lett 2011;585:1363–7. [DOI] [PubMed] [Google Scholar]
- 24.Lei G, Zhang Y, Koppula P, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res 2020;30:146–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wang G, Qin S, Chen L, et al. Butyrate dictates ferroptosis sensitivity through FFAR2-mTOR signaling. Cell Death Dis 2023;14:292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sun X, Niu X, Chen R, et al. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology 2016;64:488–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lee JH, Rangappa S, Mohan CD, et al. Brusatol, a Nrf2 Inhibitor Targets STAT3 Signaling Cascade in Head and Neck Squamous Cell Carcinoma. Biomolecules 2019;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tian Z, Yang Y, Wu H, et al. The Nrf2 inhibitor brusatol synergistically enhances the cytotoxic effect of lapatinib in HER2-positive cancers. Heliyon 2022;8:e10410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Viswanathan VS, Ryan MJ, Dhruv HD, et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 2017;547:453–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Alim I, Caulfield JT, Chen Y, et al. Selenium Drives a Transcriptional Adaptive Program to Block Ferroptosis and Treat Stroke. Cell 2019;177:1262–1279 e25. [DOI] [PubMed] [Google Scholar]
- 31.Cole SP. Multidrug resistance protein 1 (MRP1, ABCC1), a “multitasking” ATP-binding cassette (ABC) transporter. J Biol Chem 2014;289:30880–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cao JY, Poddar A, Magtanong L, et al. A Genome-wide Haploid Genetic Screen Identifies Regulators of Glutathione Abundance and Ferroptosis Sensitivity. Cell Rep 2019;26:1544–1556 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019;575:693–698. [DOI] [PubMed] [Google Scholar]
- 34.Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 2019;575:688–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Miyauchi W, Shishido Y, Matsumi Y, et al. Simultaneous regulation of ferroptosis suppressor protein 1 and glutathione peroxidase 4 as a new therapeutic strategy of ferroptosis for esophageal squamous cell carcinoma. Esophagus 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mishima E, Ito J, Wu Z, et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature 2022;608:778–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Mao C, Liu X, Zhang Y, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 2021;593:586–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu X, Wang S, Jin S, et al. Vitamin D(3) attenuates cisplatin-induced intestinal injury by inhibiting ferroptosis, oxidative stress, and ROS-mediated excessive mitochondrial fission. Food Funct 2022;13:10210–10224. [DOI] [PubMed] [Google Scholar]
- 39.Soula M, Weber RA, Zilka O, et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol 2020;16:1351–1360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rouault TA. The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nat Chem Biol 2006;2:406–14. [DOI] [PubMed] [Google Scholar]
- 41.Zhang DL, Ghosh MC, Rouault TA. The physiological functions of iron regulatory proteins in iron homeostasis - an update. Front Pharmacol 2014;5:124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Stockwell BR. Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell 2022;185:2401–2421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gao M, Monian P, Quadri N, et al. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol Cell 2015;59:298–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Feng H, Schorpp K, Jin J, et al. Transferrin Receptor Is a Specific Ferroptosis Marker. Cell Rep 2020;30:3411–3423 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yu Y, Jiang L, Wang H, et al. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood 2020;136:726–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Brown CW, Amante JJ, Chhoy P, et al. Prominin2 Drives Ferroptosis Resistance by Stimulating Iron Export. Dev Cell 2019;51:575–586 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Geng N, Shi BJ, Li SL, et al. Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharmacol Sci 2018;22:3826–3836. [DOI] [PubMed] [Google Scholar]
- 48.Fang X, Cai Z, Wang H, et al. Loss of Cardiac Ferritin H Facilitates Cardiomyopathy via Slc7a11Mediated Ferroptosis. Circ Res 2020;127:486–501. [DOI] [PubMed] [Google Scholar]
- 49.Protchenko O, Baratz E, Jadhav S, et al. Iron Chaperone Poly rC Binding Protein 1 Protects Mouse Liver From Lipid Peroxidation and Steatosis. Hepatology 2021;73:1176–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hou W, Xie Y, Song X, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016;12:1425–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gao M, Monian P, Pan Q, et al. Ferroptosis is an autophagic cell death process. Cell Res 2016;26:1021–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zhang Z, Guo M, Shen M, et al. The BRD7-P53-SLC25A28 axis regulates ferroptosis in hepatic stellate cells. Redox Biol 2020;36:101619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Alvarez SW, Sviderskiy VO, Terzi EM, et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature 2017;551:639–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yuan H, Li X, Zhang X, et al. CISD1 inhibits ferroptosis by protection against mitochondrial lipid peroxidation. Biochem Biophys Res Commun 2016;478:838–44. [DOI] [PubMed] [Google Scholar]
- 55.Kim EH, Shin D, Lee J, et al. CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer Lett 2018;432:180–190. [DOI] [PubMed] [Google Scholar]
- 56.Kwon MY, Park E, Lee SJ, et al. Heme oxygenase-1 accelerates erastin-induced ferroptotic cell death. Oncotarget 2015;6:24393–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chang LC, Chiang SK, Chen SE, et al. Heme oxygenase-1 mediates BAY 11–7085 induced ferroptosis. Cancer Lett 2018;416:124–137. [DOI] [PubMed] [Google Scholar]
- 58.Adedoyin O, Boddu R, Traylor A, et al. Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol Renal Physiol 2018;314:F702–F714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lee H, Zandkarimi F, Zhang Y, et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol 2020;22:225–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Li C, Dong X, Du W, et al. LKB1-AMPK axis negatively regulates ferroptosis by inhibiting fatty acid synthesis. Signal Transduct Target Ther 2020;5:187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Song X, Liu J, Kuang F, et al. PDK4 dictates metabolic resistance to ferroptosis by suppressing pyruvate oxidation and fatty acid synthesis. Cell Rep 2021;34:108767. [DOI] [PubMed] [Google Scholar]
- 62.Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol 2020;21:183–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Li J, Cao F, Yin HL, et al. Ferroptosis: past, present and future. Cell Death Dis 2020;11:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhang Y, Swanda RV, Nie L, et al. mTORC1 couples cyst(e)ine availability with GPX4 protein synthesis and ferroptosis regulation. Nat Commun 2021;12:1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Conlon M, Poltorack CD, Forcina GC, et al. A compendium of kinetic modulatory profiles identifies ferroptosis regulators. Nat Chem Biol 2021;17:665–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Armenta DA, Laqtom NN, Alchemy G, et al. Ferroptosis inhibition by lysosome-dependent catabolism of extracellular protein. Cell Chem Biol 2022;29:1588–1600 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Liang D, Minikes AM, Jiang X. Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell 2022;82:2215–2227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Muri J, Thut H, Bornkamm GW, et al. B1 and Marginal Zone B Cells but Not Follicular B2 Cells Require Gpx4 to Prevent Lipid Peroxidation and Ferroptosis. Cell Rep 2019;29:2731–2744 e4. [DOI] [PubMed] [Google Scholar]
- 69.Ma X, Xiao L, Liu L, et al. CD36-mediated ferroptosis dampens intratumoral CD8(+) T cell effector function and impairs their antitumor ability. Cell Metab 2021;33:1001–1012 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lee J-Y, Nam M, Son HY, et al. Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proceedings of the National Academy of Sciences of the United States of America 2020;117:32433–32442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yamane D, Hayashi Y, Matsumoto M, et al. FADS2-dependent fatty acid desaturation dictates cellular sensitivity to ferroptosis and permissiveness for hepatitis C virus replication. Cell Chemical Biology 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Dixon SJ, Winter GE, Musavi LS, et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chemical Biology 2015;10:1604–1609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nature Chemical Biology 2017;13:91–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kagan VE, Mao G, Qu F, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nature Chemical Biology 2017;13:81–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Magtanong L, Ko P-J, To M, et al. Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State. Cell Chemical Biology 2019;26:420–432.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ju HQ, Lin JF, Tian T, et al. NADPH homeostasis in cancer: functions, mechanisms and therapeutic implications. Signal Transduct Target Ther 2020;5:231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Shimada K, Hayano M, Pagano NC, et al. Cell-Line Selectivity Improves the Predictive Power of Pharmacogenomic Analyses and Helps Identify NADPH as Biomarker for Ferroptosis Sensitivity. Cell Chem Biol 2016;23:225–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ding CC, Rose J, Sun T, et al. MESH1 is a cytosolic NADPH phosphatase that regulates ferroptosis. Nat Metab 2020;2:270–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gao M, Yi J, Zhu J, et al. Role of Mitochondria in Ferroptosis. Mol Cell 2019;73:354–363 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ho GT, Theiss AL. Mitochondria and Inflammatory Bowel Diseases: Toward a Stratified Therapeutic Intervention. Annu Rev Physiol 2022;84:435–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Maher P, van Leyen K, Dey PN, et al. The role of Ca(2+) in cell death caused by oxidative glutamate toxicity and ferroptosis. Cell Calcium 2018;70:47–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ta N, Qu C, Wu H, et al. Mitochondrial outer membrane protein FUNDC2 promotes ferroptosis and contributes to doxorubicin-induced cardiomyopathy. Proc Natl Acad Sci U S A 2022;119:e2117396119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Blomme A, Ford CA, Mui E, et al. 2,4-dienoyl-CoA reductase regulates lipid homeostasis in treatment-resistant prostate cancer. Nat Commun 2020;11:2508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Nassar ZD, Mah CY, Dehairs J, et al. Human DECR1 is an androgen-repressed survival factor that regulates PUFA oxidation to protect prostate tumor cells from ferroptosis. Elife 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mishima E, Conrad M. Nutritional and Metabolic Control of Ferroptosis. Annu Rev Nutr 2022;42:275–309. [DOI] [PubMed] [Google Scholar]
- 86.Dixon SJ, Winter GE, Musavi LS, et al. Human Haploid Cell Genetics Reveals Roles for Lipid Metabolism Genes in Nonapoptotic Cell Death. ACS Chem Biol 2015;10:1604–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yang Y, Zhu T, Wang X, et al. ACSL3 and ACSL4, Distinct Roles in Ferroptosis and Cancers. Cancers (Basel) 2022;14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zhang HL, Hu BX, Li ZL, et al. PKCbetaII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nat Cell Biol 2022;24:88–98. [DOI] [PubMed] [Google Scholar]
- 89.Liao P, Wang W, Wang W, et al. CD8(+) T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell 2022;40:365–378 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Delesderrier E, Monteiro JDC, Freitas S, et al. Can Iron and Polyunsaturated Fatty Acid Supplementation Induce Ferroptosis? Cell Physiol Biochem 2023;57:24–41. [DOI] [PubMed] [Google Scholar]
- 91.Yao T, Li L. The influence of microbiota on ferroptosis in intestinal diseases. Gut Microbes 2023;15:2263210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Xu M, Tao J, Yang Y, et al. Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis. Cell Death Dis 2020;11:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Oates PS, West AR. Heme in intestinal epithelial cell turnover, differentiation, detoxification, inflammation, carcinogenesis, absorption and motility. World J Gastroenterol 2006;12:4281–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chen Y, Fang ZM, Yi X, et al. The interaction between ferroptosis and inflammatory signaling pathways. Cell Death Dis 2023;14:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhang X, Ma Y, Lv G, et al. Ferroptosis as a therapeutic target for inflammation-related intestinal diseases. Front Pharmacol 2023;14:1095366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gong D, Chen M, Wang Y, et al. Role of ferroptosis on tumor progression and immunotherapy. Cell Death Discovery 2022;8:427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Subramanian S, Geng H, Tan XD. Cell death of intestinal epithelial cells in intestinal diseases. Sheng Li Xue Bao 2020;72:308–324. [PMC free article] [PubMed] [Google Scholar]
- 98.Zhou P, Zhang S, Wang M, et al. The Induction Mechanism of Ferroptosis, Necroptosis, and Pyroptosis in Inflammatory Bowel Disease, Colorectal Cancer, and Intestinal Injury. Biomolecules 2023;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zhu X, Li S. Ferroptosis, Necroptosis, and Pyroptosis in Gastrointestinal Cancers: The Chief Culprits of Tumor Progression and Drug Resistance. Adv Sci (Weinh) 2023;10:e2300824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Macias-Rodriguez RU, Inzaugarat ME, Ruiz-Margain A, et al. Reclassifying Hepatic Cell Death during Liver Damage: Ferroptosis-A Novel Form of Non-Apoptotic Cell Death? Int J Mol Sci 2020;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chen Y, Zhang P, Chen W, et al. Ferroptosis mediated DSS-induced ulcerative colitis associated with Nrf2/HO-1 signaling pathway. Immunol Lett 2020;225:9–15. [DOI] [PubMed] [Google Scholar]
- 102.Xu J, Liu S, Cui Z, et al. Ferrostatin-1 alleviated TNBS induced colitis via the inhibition of ferroptosis. Biochem Biophys Res Commun 2021;573:48–54. [DOI] [PubMed] [Google Scholar]
- 103.Huang F, Zhang S, Li X, et al. STAT3-mediated ferroptosis is involved in ulcerative colitis. Free Radic Biol Med 2022;188:375–385. [DOI] [PubMed] [Google Scholar]
- 104.Gu K, Wu A, Yu B, et al. Iron overload induces colitis by modulating ferroptosis and interfering gut microbiota in mice. Sci Total Environ 2023;905:167043. [DOI] [PubMed] [Google Scholar]
- 105.Mayr L, Grabherr F, Schwarzler J, et al. Dietary lipids fuel GPX4-restricted enteritis resembling Crohn’s disease. Nat Commun 2020;11:1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Colgan SP, Wang RX, Hall CHT, et al. Revisiting the “starved gut” hypothesis in inflammatory bowel disease. Immunometabolism (Cobham) 2023;5:e0016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Chapkin RS, Navarro SL, Hullar MAJ, et al. Diet and Gut Microbes Act Coordinately to Enhance Programmed Cell Death and Reduce Colorectal Cancer Risk. Dig Dis Sci 2020;65:840–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wu Y, Ran L, Yang Y, et al. Deferasirox alleviates DSS-induced ulcerative colitis in mice by inhibiting ferroptosis and improving intestinal microbiota. Life Sci 2023;314:121312. [DOI] [PubMed] [Google Scholar]
- 109.Yang F, Chen Y, Xiao Y, et al. pH-sensitive molybdenum (Mo)-based polyoxometalate nanoclusters have therapeutic efficacy in inflammatory bowel disease by counteracting ferroptosis. Pharmacol Res 2023;188:106645. [DOI] [PubMed] [Google Scholar]
- 110.Zhang X, Li W, Ma Y, et al. High-fat diet aggravates colitis-associated carcinogenesis by evading ferroptosis in the ER stress-mediated pathway. Free Radic Biol Med 2021;177:156–166. [DOI] [PubMed] [Google Scholar]
- 111.Scarpellini C, Klejborowska G, Lanthier C, et al. Beyond ferrostatin-1: a comprehensive review of ferroptosis inhibitors. Trends Pharmacol Sci 2023. [DOI] [PubMed] [Google Scholar]
- 112.Van Coillie S, Van San E, Goetschalckx I, et al. Targeting ferroptosis protects against experimental (multi)organ dysfunction and death. Nat Commun 2022;13:1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Tsurusaki S, Tsuchiya Y, Koumura T, et al. Hepatic ferroptosis plays an important role as the trigger for initiating inflammation in nonalcoholic steatohepatitis. Cell Death Dis 2019;10:449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wang H, An P, Xie E, et al. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology 2017;66:449–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Yang WS, Stockwell BR. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol 2016;26:165176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Sun Y, Zheng Y, Wang C, et al. Glutathione depletion induces ferroptosis, autophagy, and premature cell senescence in retinal pigment epithelial cells. Cell Death Dis 2018;9:753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kouroumalis E, Tsomidis I, Voumvouraki A. Iron as a therapeutic target in chronic liver disease. World J Gastroenterol 2023;29:616–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Vela D. Low hepcidin in liver fibrosis and cirrhosis; a tale of progressive disorder and a case for a new biochemical marker. Mol Med 2018;24:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Baratz E, Protchenko O, Jadhav S, et al. Vitamin E Induces Liver Iron Depletion and Alters Iron Regulation in Mice. J Nutr 2023;153:1866–1876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Stravitz RT, Lee WM. Acute liver failure. Lancet 2019;394:869–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Lorincz T, Jemnitz K, Kardon T, et al. Ferroptosis is Involved in Acetaminophen Induced Cell Death. Pathol Oncol Res 2015;21:1115–21. [DOI] [PubMed] [Google Scholar]
- 122.Suzuki Y, Saito H, Suzuki M, et al. Up-regulation of transferrin receptor expression in hepatocytes by habitual alcohol drinking is implicated in hepatic iron overload in alcoholic liver disease. Alcohol Clin Exp Res 2002;26:26S–31S. [DOI] [PubMed] [Google Scholar]
- 123.Li LX, Guo FF, Liu H, et al. Iron overload in alcoholic liver disease: underlying mechanisms, detrimental effects, and potential therapeutic targets. Cell Mol Life Sci 2022;79:201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Dostalikova-Cimburova M, Balusikova K, Kratka K, et al. Role of duodenal iron transporters and hepcidin in patients with alcoholic liver disease. J Cell Mol Med 2014;18:1840–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Liu J, He H, Wang J, et al. Oxidative stress-dependent frataxin inhibition mediated alcoholic hepatocytotoxicity through ferroptosis. Toxicology 2020;445:152584. [DOI] [PubMed] [Google Scholar]
- 126.You Y, Liu C, Liu T, et al. FNDC3B protects steatosis and ferroptosis via the AMPK pathway in alcoholic fatty liver disease. Free Radic Biol Med 2022;193:808–819. [DOI] [PubMed] [Google Scholar]
- 127.Zhou Z, Ye TJ, Bonavita G, et al. Adipose-Specific Lipin-1 Overexpression Renders Hepatic Ferroptosis and Exacerbates Alcoholic Steatohepatitis in Mice. Hepatol Commun 2019;3:656–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Zhou Z, Ye TJ, DeCaro E, et al. Intestinal SIRT1 Deficiency Protects Mice from Ethanol-Induced Liver Injury by Mitigating Ferroptosis. Am J Pathol 2020;190:82–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Wu J, Xue R, Wu M, et al. Nrf2-Mediated Ferroptosis Inhibition Exerts a Protective Effect on Acute-on-Chronic Liver Failure . Oxid Med Cell Longev 2022;2022:4505513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zhao W, Lei M, Li J, et al. Yes-associated protein inhibition ameliorates liver fibrosis and acute and chronic liver failure by decreasing ferroptosis and necroptosis. Heliyon 2023;9:e15075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lu Y, Hu J, Chen L, et al. Ferroptosis as an emerging therapeutic target in liver diseases. Front Pharmacol 2023;14:1196287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zhang Q, Qu Y, Zhang Q, et al. Exosomes derived from hepatitis B virus-infected hepatocytes promote liver fibrosis via miR-222/TFRC axis. Cell Biol Toxicol 2023;39:467–481. [DOI] [PubMed] [Google Scholar]
- 133.Hu Z, Yin Y, Jiang J, et al. Exosomal miR-142–3p secreted by hepatitis B virus (HBV)-hepatocellular carcinoma (HCC) cells promotes ferroptosis of M1-type macrophages through SLC3A2 and the mechanism of HCC progression. J Gastrointest Oncol 2022;13:754–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Deng W, Ai J, Zhang W, et al. Arginine methylation of HSPA8 by PRMT9 inhibits ferroptosis to accelerate hepatitis B virus-associated hepatocellular carcinoma progression. J Transl Med 2023;21:625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wang Y, Zhao M, Zhao L, et al. HBx-Induced HSPA8 Stimulates HBV Replication and Suppresses Ferroptosis to Support Liver Cancer Progression. Cancer Res 2023;83:1048–1061. [DOI] [PubMed] [Google Scholar]
- 136.Shi J, Liu Z, Li W, et al. Selenium Donor Inhibited Hepatitis B Virus Associated Hepatotoxicity via the Apoptosis and Ferroptosis Pathways . Anal Cell Pathol (Amst) 2023;2023:6681065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Yamane D, Hayashi Y, Matsumoto M, et al. FADS2-dependent fatty acid desaturation dictates cellular sensitivity to ferroptosis and permissiveness for hepatitis C virus replication. Cell Chem Biol 2022;29:799–810 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Guan Q, Wang Z, Hu K, et al. Melatonin Ameliorates Hepatic Ferroptosis in NAFLD by Inhibiting ER Stress via the MT2/cAMP/PKA/IRE1 Signaling Pathway. Int J Biol Sci 2023;19:3937–3950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Zhao J, Hu Y, Peng J. Targeting programmed cell death in metabolic dysfunction-associated fatty liver disease (MAFLD): a promising new therapy. Cell Mol Biol Lett 2021;26:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Zhang XJ, Ji YX, Cheng X, et al. A small molecule targeting ALOX12-ACC1 ameliorates nonalcoholic steatohepatitis in mice and macaques. Sci Transl Med 2021;13:eabg8116. [DOI] [PubMed] [Google Scholar]
- 141.Honma K, Kirihara S, Nakayama H, et al. Selective autophagy associated with iron overload aggravates non-alcoholic steatohepatitis via ferroptosis. Exp Biol Med (Maywood) 2023:15353702231191197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Qi J, Kim JW, Zhou Z, et al. Ferroptosis Affects the Progression of Nonalcoholic Steatohepatitis via the Modulation of Lipid Peroxidation-Mediated Cell Death in Mice. Am J Pathol 2020;190:68–81. [DOI] [PubMed] [Google Scholar]
- 143.Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol 2017;14:397–411. [DOI] [PubMed] [Google Scholar]
- 144.Zhang Z, Yao Z, Wang L, et al. Activation of ferritinophagy is required for the RNA-binding protein ELAVL1/HuR to regulate ferroptosis in hepatic stellate cells. Autophagy 2018;14:2083–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Zhang Z, Guo M, Li Y, et al. RNA-binding protein ZFP36/TTP protects against ferroptosis by regulating autophagy signaling pathway in hepatic stellate cells. Autophagy 2020;16:1482–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Powell LW, Seckington RC, Deugnier Y. Haemochromatosis. Lancet 2016;388:706–16. [DOI] [PubMed] [Google Scholar]
- 147.Lei G, Zhuang L, Gan B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer 2022;22:381–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lee J, Shin D, Roh JL. Lipid metabolism alterations and ferroptosis in cancer: Paving the way for solving cancer resistance. Eur J Pharmacol 2023:175497. [DOI] [PubMed] [Google Scholar]
- 149.Lin Z, Liu J, Long F, et al. The lipid flippase SLC47A1 blocks metabolic vulnerability to ferroptosis. Nat Commun 2022;13:7965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Xu FL, Wu XH, Chen C, et al. SLC27A5 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by downregulating glutathione reductase. Cell Death Dis 2023;14:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Jiang L, Kon N, Li T, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 2015;520:57–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ou Y, Wang SJ, Li D, et al. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci U S A 2016;113:E6806–E6812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Seiler A, Schneider M, Forster H, et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab 2008;8:23748. [DOI] [PubMed] [Google Scholar]
- 154.Tarangelo A, Magtanong L, Bieging-Rolett KT, et al. p53 Suppresses Metabolic Stress-Induced Ferroptosis in Cancer Cells. Cell Rep 2018;22:569–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Xie Y, Zhu S, Song X, et al. The Tumor Suppressor p53 Limits Ferroptosis by Blocking DPP4 Activity. Cell Rep 2017;20:1692–1704. [DOI] [PubMed] [Google Scholar]
- 156.Wu J, Minikes AM, Gao M, et al. Intercellular interaction dictates cancer cell ferroptosis via NF2–YAP signalling. Nature 2019;572:402–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Lim JKM, Delaidelli A, Minaker SW, et al. Cystine/glutamate antiporter xCT (SLC7A11) facilitates oncogenic RAS transformation by preserving intracellular redox balance. Proc Natl Acad Sci U S A 2019;116:9433–9442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hu B, Yin Y, Li S, et al. Insights on Ferroptosis and Colorectal Cancer: Progress and Updates. Molecules 2022;28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gotorbe C, Durivault J, Meira W, et al. Metabolic Rewiring toward Oxidative Phosphorylation Disrupts Intrinsic Resistance to Ferroptosis of the Colon Adenocarcinoma Cells. Antioxidants (Basel) 2022;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Yang L, WenTao T, ZhiYuan Z, et al. Cullin-9/p53 mediates HNRNPC degradation to inhibit erastin-induced ferroptosis and is blocked by MDM2 inhibition in colorectal cancer. Oncogene 2022;41:3210–3221. [DOI] [PubMed] [Google Scholar]
- 161.Liu MY, Li HM, Wang XY, et al. TIGAR drives colorectal cancer ferroptosis resistance through ROS/AMPK/SCD1 pathway. Free Radic Biol Med 2022;182:219–231. [DOI] [PubMed] [Google Scholar]
- 162.Singhal R, Mitta SR, Das NK, et al. HIF-2alpha activation potentiates oxidative cell death in colorectal cancers by increasing cellular iron. J Clin Invest 2021;131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Zhao Y, Li J, Guo W, et al. Periodontitis-level butyrate-induced ferroptosis in periodontal ligament fibroblasts by activation of ferritinophagy. Cell Death Discov 2020;6:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.He Y, Ling Y, Zhang Z, et al. Butyrate reverses ferroptosis resistance in colorectal cancer by inducing c-Fos-dependent xCT suppression. Redox Biol 2023;65:102822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Zhang X, Ma Y, Ma J, et al. Glutathione Peroxidase 4 as a Therapeutic Target for Anti-Colorectal Cancer Drug-Tolerant Persister Cells. Front Oncol 2022;12:913669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Chaudhary N, Choudhary BS, Shah SG, et al. Lipocalin 2 expression promotes tumor progression and therapy resistance by inhibiting ferroptosis in colorectal cancer. Int J Cancer 2021;149:14951511. [DOI] [PubMed] [Google Scholar]
- 167.Feng M, Feng J, Chen W, et al. Lipocalin2 suppresses metastasis of colorectal cancer by attenuating NF-kappaB-dependent activation of snail and epithelial mesenchymal transition. Mol Cancer 2016;15:77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Yang J, Mo J, Dai J, et al. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis 2021;12:1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Wen Q, Liu J, Kang R, et al. The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun 2019;510:278–283. [DOI] [PubMed] [Google Scholar]
- 170.Zheng Y, Sun L, Guo J, et al. The crosstalk between ferroptosis and anti-tumor immunity in the tumor microenvironment: molecular mechanisms and therapeutic controversy. Cancer Commun (Lond) 2023;43:1071–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Wang W, Green M, Choi JE, et al. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019;569:270–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Wang D, DuBois RN. Immunosuppression associated with chronic inflammation in the tumor microenvironment. Carcinogenesis 2015;36:1085–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Liu Y, Cao X. Immunosuppressive cells in tumor immune escape and metastasis. J Mol Med (Berl) 2016;94:509–22. [DOI] [PubMed] [Google Scholar]
- 174.Ge Y, Huang M, Yao YM. The Effect and Regulatory Mechanism of High Mobility Group Box-1 Protein on Immune Cells in Inflammatory Diseases. Cells 2021;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Dai E, Han L, Liu J, et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy 2020;16:2069–2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Roszer T. Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediators Inflamm 2015;2015:816460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Pathria P, Louis TL, Varner JA. Targeting Tumor-Associated Macrophages in Cancer. Trends Immunol 2019;40:310–327. [DOI] [PubMed] [Google Scholar]
- 178.Dai E, Han L, Liu J, et al. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat Commun 2020;11:6339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Jiang M, Cui H, Liu Z, et al. The Role of Amino Acid Metabolism of Tumor Associated Macrophages in the Development of Colorectal Cancer. Cells 2022;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kapralov AA, Yang Q, Dar HH, et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat Chem Biol 2020;16:278–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Xu C, Sun S, Johnson T, et al. The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity. Cell Rep 2021;35:109235. [DOI] [PubMed] [Google Scholar]
- 182.Zhu H, Klement JD, Lu C, et al. Asah2 Represses the p53-Hmox1 Axis to Protect Myeloid-Derived Suppressor Cells from Ferroptosis. J Immunol 2021;206:1395–1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Srivastava MK, Sinha P, Clements VK, et al. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 2010;70:68–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Matsushita M, Freigang S, Schneider C, et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J Exp Med 2015;212:555–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Drijvers JM, Gillis JE, Muijlwijk T, et al. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8(+) T Cells. Cancer Immunol Res 2021;9:184–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Xu S, Chaudhary O, Rodriguez-Morales P, et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8(+) T cells in tumors. Immunity 2021;54:1561–1577 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Lang X, Green MD, Wang W, et al. Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11. Cancer Discov 2019;9:1673–1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Abiko K, Matsumura N, Hamanishi J, et al. IFN-gamma from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer. Br J Cancer 2015;112:1501–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Lv Y, Tang W, Xu Y, et al. Apolipoprotein L3 enhances CD8+ T cell antitumor immunity of colorectal cancer by promoting LDHA-mediated ferroptosis. Int J Biol Sci 2023;19:1284–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Poznanski SM, Singh K, Ritchie TM, et al. Metabolic flexibility determines human NK cell functional fate in the tumor microenvironment. Cell Metab 2021;33:1205–1220 e5. [DOI] [PubMed] [Google Scholar]
- 191.Herber DL, Cao W, Nefedova Y, et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat Med 2010;16:880–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, et al. ER Stress Sensor XBP1 Controls Anti-tumor Immunity by Disrupting Dendritic Cell Homeostasis. Cell 2015;161:1527–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Han L, Bai L, Qu C, et al. PPARG-mediated ferroptosis in dendritic cells limits antitumor immunity. Biochem Biophys Res Commun 2021;576:33–39. [DOI] [PubMed] [Google Scholar]
- 194.Wiernicki B, Maschalidi S, Pinney J, et al. Cancer cells dying from ferroptosis impede dendritic cell-mediated anti-tumor immunity. Nat Commun 2022;13:3676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Kim R, Hashimoto A, Markosyan N, et al. Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature 2022;612:338–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Eberhart CE, Coffey RJ, Radhika A, et al. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology 1994;107:1183–8. [DOI] [PubMed] [Google Scholar]
- 197.Zelenay S, van der Veen AG, Bottcher JP, et al. Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity. Cell 2015;162:1257–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Liu B, Qu L, Yan S. Cyclooxygenase-2 promotes tumor growth and suppresses tumor immunity. Cancer Cell Int 2015;15:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Arensman MD, Yang XS, Leahy DM, et al. Cystine-glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc Natl Acad Sci U S A 2019;116:9533–9542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Li J, Liu J, Zhou Z, et al. Tumor-specific GPX4 degradation enhances ferroptosis-initiated antitumor immune response in mouse models of pancreatic cancer. Sci Transl Med 2023;15:eadg3049. [DOI] [PubMed] [Google Scholar]
- 201.Wu J, Liu C, Wang T, et al. Deubiquitinase inhibitor PR-619 potentiates colon cancer immunotherapy by inducing ferroptosis. Immunology 2023;170:439–451. [DOI] [PubMed] [Google Scholar]
- 202.Hao S, Yu J, He W, et al. Cysteine Dioxygenase 1 Mediates Erastin-Induced Ferroptosis in Human Gastric Cancer Cells. Neoplasia 2017;19:1022–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Eling N, Reuter L, Hazin J, et al. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience 2015;2:517–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Roh JL, Kim EH, Jang H, et al. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol 2017;11:254–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Kang YP, Mockabee-Macias A, Jiang C, et al. Non-canonical Glutamate-Cysteine Ligase Activity Protects against Ferroptosis. Cell Metab 2021;33:174–189 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]


