Abstract
The most widespread type of liver cancer, hepatocellular carcinoma (HCC), is associated with disabled cellular death pathways. Despite therapeutic advancements, resistance to current systemic treatments (including sorafenib) compromises the prognosis of HCC patients, driving the search for agents that might target novel cell death pathways. Ferroptosis, a form of iron-mediated non-apoptotic cell death, has gained considerable attention as a potential target for cancer therapy, especially in HCC. The role of ferroptosis in HCC is complex and diverse. On one hand, ferroptosis can both contribute to the progression of HCC through its involvement in acute and chronic liver conditions. On the other hand, having ferroptosis affect HCC cells might be desirable. This review examines the role of ferroptosis in HCC from cellular, animal, and human perspectives, while examining its mechanisms, regulation, biomarkers, and clinical implications.
Keywords: ferroptosis, hepatocellular carcinoma, cell death, targeted therapy
Introduction
The most prevalent primary liver cancer in adults is hepatocellular carcinoma (HCC), a global health concern with a rising incidence and mortality rate (1). HCC is more prevalent in sub-Saharan Africa and Southeast Asia compared to Western countries, frequently resulting from chronic liver disease caused by hepatitis B/C infection, alcohol abuse, or metabolic syndrome (e.g., non-alcoholic fatty liver disease [NAFLD]) (2). Despite the increasing utilization of surgical and locoregional treatments, approximately 50%-60% of HCC patients will undergo systemic therapies. Targeted therapies have constituted the main treatment approach for advanced-stage HCC for over a decade. The oral multi-tyrosine kinase inhibitors sorafenib and lenvatinib (and donafenib in China) confer median overall survival benefits of 11-14 months, and regorafenib, cabozantinib (which inhibit vascular endothelial growth factor [VEGF] receptor 2 [VEGFR2] and other tyrosine kinases), and ramucirumab (a monoclonal antibody [mAb] specific for VEGFR2) yield benefits in the order of 8-11 months overall survival in second-line treatment (3). The new standard of care for HCC is the combination of the immune checkpoint inhibitor atezolizumab (an anti-CD274/PD-L1 antibody) and VEGF blockade with bevacizumab (4). However, the development of drug resistance, which at least is partly due to disabled apoptosis, remains a significant challenge in clinical oncology. The deconvolution of such resistance mechanisms as well as the development of drugs that induce non-apoptotic cell death could provide new opportunities for HCC therapy.
Cell death is a crucial process that plays both physiological and pathological roles in human health and disease, including cancer. Historically, cell death was categorized based on morphological criteria into apoptosis (characterized by cell shrinkage and fragmentation into membrane-enclosed “apoptotic bodies”), necrosis (characterized by the swelling of organelles, plasma membrane rupture, and cell lysis), and autophagy (characterized by the formation of autophagic vacuoles containing organelles). Currently, cell death is classified into accidental cell death (ACD) and regulated cell death (RCD) (5). In contrast to ACD, RCD is characterized by well-defined molecular and cellular mechanisms that can be manipulated through pharmacological or genetic means (6). Ferroptosis was initially identified as a type of iron-dependent, non-apoptotic RCD in RAS-mutant engineered cells that can be selectively induced by the chemical compound erastin (7). Further research has revealed that the core mechanism of ferroptosis involves lipid peroxidation caused by oxidative damage, which can be avoided by a complex antioxidant system (8). The study of ferroptosis and its implications for human diseases, including liver disease (9) and HCC (10), represent a cutting-edge area in biomedicine (11, 12).
Here, we will summarize the mechanisms of, and defenses against, ferroptosis, outline the regulators of ferroptosis, discuss biomarkers that may predict ferroptosis sensitivity in HCC, and analyze the potential use of ferroptosis-based scoring systems in clinical HCC practice. Finally, we will outline current strategies to pharmacologically target ferroptosis.
The activation mechanism of ferroptosis
Ferroptosis is a type of RCD caused by unrestricted lipid peroxidation and resultant membrane damage, dependent on iron accumulation (13). Ferroptosis differs in morphological, biochemical, and genetic aspects from other RCDs such as apoptosis, necroptosis, and pyroptosis (6). Ferroptosis can be triggered in different fashions, namely, (i) through the extrinsic pathway (e.g., the inhibition of amino acid antiporter system xc− or the activation of iron transporter transferrin [TF] and lactotransferrin on cell membrane) and (ii) the intrinsic pathway (mainly by blocking the expression or activity of intracellular glutathione peroxidase 4 [GPX4]) (14). Ferroptosis does not require the main executors of apoptosis (driven by a specific subset of caspases), necroptosis (mediated by mixed-lineage kinase domain-like pseudokinase [MLKL]), and pyroptosis (executed by gasdermin D [GSDMD]) (7). Upon modification or cleavage, MLKL or GSDMD assemble into multimeric structures that function as porin proteins, facilitating the permeabilization and damage of cellular membranes. Rather, ferroptosis is driven by lipid peroxidation products, among which 4-hydroxy 2-nonenal (4HNE) is particularly toxic and abundant (15). This highlights that ferroptosis does not rely on known pore protein for lethal plasma membrane rupture to occur. In the forthcoming paragraphs, we will summarize the three key steps that ignite ferroptosis in cancer, including in HCC (Fig. 1).
Figure 1. The core mechanism and regulation of ferroptosis in HCC.
Ferroptosis is a type of cell death triggered by lipid peroxidation, a process that causes damage to cellular membranes. This process occurs in three stages: the generation of reactive oxygen species (ROS), the accumulation of polyunsaturated fatty acids (PUFAs), and the activation of enzymes involved in lipid peroxidation. There are multiple sources of these molecules. To counter ferroptosis, cells employ different defense systems. For instance, the production of monounsaturated fatty acids (MUFAs) through the SREBP1-SCD pathway can effectively inhibit PUFAs and their derivatives produced by the ACSL4-LPCAT3, ACSL4-SOAT1, and SLC1A5-GLS2 metabolic pathways. Additionally, antioxidant pathways in the cytoplasm, plasma membrane, and mitochondria work to prevent lipid peroxidation in a context-dependent manner. Finally, any damage to the membrane can be repaired through the action of the flippase SLC47A1 or the ESCRT-III machinery.
Generation of free radicals
Free radicals, including reactive oxygen species (ROS) and reactive nitrogen species, are unstable molecules with an unpaired electron that can cause oxidative damage to cells, tissues, and DNA. ROS, which can be generated as a result of environmental factors (e.g., radiation, pollution, and tobacco smoke) and normal metabolic processes, favors the ignition of ferroptosis. The main ferroptosis-relevant sources of ROS are mitochondria, enzymes of the NADPH oxidase (NOX) family, and the iron-mediated Fenton reaction. Mitochondria are more abundant in hepatocytes than in any other cell type. Mitochondria generate the majority of cellular ROS as a side product of oxidative phosphorylation (OXPHOS). In the liver, NOX enzymes are expressed by immune cells endowed with the capacity of phagocytosis, as well as by non-phagocytic cells (16). NOX enzymes generate superoxide and other downstream forms of ROS in the plasma membrane. Finally, an excess of free reactive iron (II; Fe2+) can damage cells via the Fenton reaction with hydrogen peroxide (H2O2), yielding hydroxide (OH−) and hydroxyl (OH•) radicals.
The level of free iron in cells is regulated by an integrated system controlling iron absorption, storage, utilization, and outflow. In the liver, iron-induced oxidative stress can lead to fibrosis and cirrhosis, which increases the risk of HCC (17). Ferroptosis can be caused by an increase in TF-mediated iron absorption or by autophagy-dependent degradation of ferritin, which is a protein that stores and inactivates iron (18, 19). However, TF present in hepatocytes may attenuate high iron diet-induced liver fibrosis and cirrhosis (20), suggesting that it may play an organ-specific hepatoprotective role.
The release of Fe2+ into the extracellular space requires iron transporters, such as solute carrier family 40 member 1 (SLC40A1, also known as ferroportin-1) and another iron transporter, lipocalin 2 (LCN2) in HCC cells. Fe2+ transported out of the cell by SLC40A1 need to be oxidized by ferroxidase (e.g., ceruloplasmin [CP]) to facilitate Fe3+ loading on TF. The deletion of CP can promote erastin- or RSL3-induced ferroptosis in HCC cells by preventing iron release (21). In contrast, the upregulation of LCN2 increases iron release, thereby preventing ferroptotic cell death in HCC cells (22). The expression of CDGSH iron sulfur domain 1 (CISD1), a mitochondrial iron-sulfur protein, negatively impacts the survival rate of patients with HCC (23) (Table 1). Additionally, CISD1 can prevent iron-triggered intra-mitochondrial lipid peroxidation and ferroptosis in HCC cells (24). These findings elucidate (part of) the link between disturbances in iron metabolism and susceptibility to ferroptosis in HCC.
Table 1.
Regulators of ferroptosis in HCC
Name | Function in ferroptosis |
Expression in HCC patients |
Function in liver tumorigenesis |
References |
---|---|---|---|---|
ACSL4 | Promoter; increases PUFA | Upregulation | Promoter | (26, 31, 32) |
AIFM2 | Suppressor; increases CoQ10 and membrane repair | Upregulation | Suppressor | (53, 56) |
BECN1 | Promoter; increases autophagy | Upregulation | Suppressor | (144, 145) |
CISD1 | Suppressor; inhibits mitochondrial iron | Upregulation | Promoter | (23, 24) |
DHODH | Suppressor; increases CoQ10 | Upregulation | Promoter | (57) |
GLS2 | Promoter; increases glutamate production | Downregulation | Suppressor | (87) |
GPX4 | Suppressor; inhibits lipid peroxidation | Upregulation | N/A | (48) |
LCN2 | Suppressor; promotes iron release | Upregulation | Promoter | (22, 146) |
NCOA4 | Promoter; increases iron | Upregulation | N/A | (147) |
NFE2L2 | Suppressor; inhibits oxidative damage | Upregulation | Suppressor or promoter | (67, 69, 71) |
POR | Promoter; increases lipid peroxidation | Downregulation | Suppressor | (37, 38) |
SCD | Suppressor; increases MUFA | Upregulation | Promoter | (33, 34) |
SLC47A1 | Suppressor; promotes membrane repair | Upregulation | N/A | (60) |
SLC7A11 | Suppressor; increases cystine uptake | Upregulation | Promoter | (47) |
SREBP1 | Suppressor; increase MUFA production | Upregulation | Promoter | (35, 148) |
TF | Promoter; increases iron absorption | Upregulation | Suppressor | (20) |
TP53 | Suppressor or promoter; inhibits SLC7A11 or promotes PLTP/GLS2 expression | Upregulation or downregulation | Suppressor or promoter | (84, 85, 87, 90) |
YAP1 | Promoter; increases TFRC expression | Upregulation | Promoter | (96) |
ZSCAN25 | Suppressor; inhibits TP53 activity | Upregulation | Promoter | (86) |
Poly- and monounsaturated fatty acids
Polyunsaturated fatty acids (PUFAs) are a type of lipid found in cell membranes that help maintain the membranes’ fluidity and stability by controlling the movement of membrane components. PUFAs also act as precursors to signaling molecules, such as eicosanoids, which regulate various physiological processes, including inflammation, coagulation, and blood pressure (25). The incorporation of PUFAs, such as arachidonic acid (AA) and adrenic acid (AdA), into phospholipids present in the cell membrane requires two key enzymes, acyl-CoA synthetase long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3) (26). ACSL4 converts AA/AdA into AA/AdA-CoA derivatives and promotes their esterification into phospholipids, while LPCAT3 catalyzes the conjugation of AA/AdA-CoA and membrane phosphatidylethanolamine (PE) to form AA/AdA–PE. A deficiency in the lipid flippase solute carrier family 47 member 1 (SLC47A1) promotes the use the ACSL4-sterol O-acyltransferase 1 (SOAT1) pathway instead of the ACSL4-LPCAT3 pathway to produce PUFA-containing cholesterol esters and promote ferroptosis (27). Other pathways, such as glutaminolysis-mediated α-ketoglutarate production (28), lipophagy-mediated degradation of lipid droplets (29), and peroxisome-driven biosynthesis of plasmalogens, (30) can provide an additional supply of PUFAs to enhance ferroptosis sensitivity in cancer cells, including in HCC. In contrast, tumor protein D52 (TPD52)-dependent lipid storage inhibits RSL3-induced ferroptosis in HCC cells (29). It is important to note that ACSL4 is expressed at elevated levels in the liver of NAFLD patients and likely plays a role in promoting the development of HCC (31, 32).
Monounsaturated fatty acids (MUFAs), such as oleic acid and palmitoleic acid, produced through the partial hydrogenation of unsaturated fatty acids during desaturation, can inhibit PUFA-related ferroptosis (33). MUFAs can also be obtained from dietary sources like olive oil, avocados, and nuts, or produced by the body through the action of stearoyl-CoA desaturase (SCD). SCD is overexpressed in surgically resected HCC tissues and acts as a major component in lipogenesis pathways (34). The inhibition of SCD reduces endogenous MUFA production and increases ferroptosis in Hep3B and Huh-7 cells (35). This process can be controlled by mitochondrial metabolism. Specifically, lactate uptake mediated by solute carrier family 16 member 1 (SLC16A1/MCT1, a monocarboxylate transporter) induces the upregulation of SCD in HCC cells, thereby inhibiting the activation of AMP-activated protein kinase (AMPK) and promoting subsequent expression of sterol regulatory element binding transcription factor 1 (SREBP1), a transcription factor for SCD mRNA expression (35). Of note, endogenously synthesized MUFAs probably contribute more to membrane lipids than MUFAs absorbed through nutrition. Thus, SCD is central for providing ferroptosis-inhibiting MUFAs. A major unresolved challenge is the lack of understanding regarding the intricate and fluctuating interactions between lipid metabolism and other metabolic processes in HCC.
Activation of lipid peroxidation
Lipid peroxidation can be initiated through both enzymatic and non-enzymatic mechanisms (25). Iron can increase the likelihood of ferroptosis by enhancing the activity of lipoxygenases (ALOXs), which are a class of non-heme iron-containing enzymes that catalyze the oxidation of PUFAs. This oxidation results in the formation of hydroperoxyl derivatives, such as lipid hydroperoxides (LOOHs) and reactive aldehydes, such as malondialdehyde and 4HNE. These aldehydes not only act as mediators of inflammation, but can also lead to the formation of adducts and crosslinks in proteins or DNA, eventually perturbing proteostasis and genomic stability (25). The mammalian ALOX family includes six members (ALOXE3, ALOX5, ALOX12, ALOX12B, ALOX15, and ALOX15B), each of which can play a context-dependent role in lipid peroxidation and ferroptosis (36).
Cytochrome P450 oxidoreductase (POR) is yet another enzyme that promotes PUFA peroxidation during ferroptosis (37). Unlike the ALOXs, which are expressed in a tissue-selective fashion, POR appears to be widely expressed in various tissues and has a broad-spectrum role in promoting lipid peroxidation. The POR-dependent lipid peroxidation process is initiated by the auto-oxidation of POR, which generates ROS in a reaction that is dependent on two co-factors: flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) (37). These co-factors enhance the peroxidation of PUFAs during ferroptosis (37). Specifically, within HCC, the expression of POR is enhanced in tumor tissues, and this overexpression is associated with a poor prognosis (38). POR is also involved in regulating the expression of cytochrome P450 genes, as well as in controlling the localization and stability of cytochrome P450 enzymes within the cell (39). Future studies should investigate the impact of POR on the metabolism of ferroptosis-inducing drugs in the liver.
Of note, although lipid peroxidation is considered a key hallmark of ferroptosis, it is not an effector molecule of ferroptosis (40). Indeed, increases in oxidative stress-related iron and/or lipid metabolites are observed in many pathological conditions, which may not be related to ferroptosis (41). The point of no return for ferroptotic cells is unclear, one hypothesis may be irreversible damage to normal ion exchange pumps and membrane tension at the plasma membrane, leading to cellular edema and swelling (42, 43).
Defense mechanisms against ferroptosis
Defense against ferroptosis is mediated by antioxidant systems and membrane repair machinery that suppress lipid peroxidation and membrane rupture, respectively (Fig. 1).
The antioxidant system
The system xc−-glutathione (GSH)-GPX4 pathway plays a major role in antioxidant defense (44). Indeed, much of our understanding of the role and regulation of ferroptosis comes from the study of chemical inhibitors of system xc− (e.g., erastin) or GPX4 (e.g., RSL3). System xc−, also known as the xCT antiporter, is a cell membrane transporter composed of solute carrier family 7 member 11 (SLC7A11) and solute carrier family 3 member 2 (SLC3A2). It facilitates the transport of cystine into the cell, which is then converted into cysteine and used in the synthesis of the antioxidant GSH (45). GSH is crucial for the activity of GPX4, a selenoenzyme that acts as a key repressor of ferroptosis by reducing toxic phospholipid hydroperoxides (PLOOHs) to non-toxic phospholipid alcohols (PLOHs). Through alternative splicing, the single GPX4 gene yields three GPX4 isoforms, among which the cytosolic and mitochondrial (but not the nuclear) isoforms have been documented to inhibit ferroptosis (44, 46). In humans, GPX4 and SLC7A11 are expressed at higher levels in HCC tumor tissue compared to normal tissues (47, 48). Mice with a liver-specific deficiency of Gpx4 died shortly after birth due to extensive hepatocyte degeneration caused by ferroptosis, emphasizing the importance of GPX4 in liver development (49). However, in other mouse tissues, Gpx4 deficiency does not necessarily cause ferroptosis. For example, Gpx4 deficiency in myeloid cells enhances pyroptosis (rather than ferroptosis) during bacterial infection (50). Gpx4 deficiency in mouse erythroid precursors leads to functional inactivation of caspase 8 (CASP8) by glutathionylation, resulting in necroptosis (51). Similarly, a low expression of GPX4 in cancer cells does not necessarily result in increased sensitivity to ferroptosis (52), suggesting that other antioxidant systems may protect against ferroptosis.
Two antioxidant systems have been found to play a role similar to that of GPX4 in protecting against ferroptosis. The first is the apoptosis-inducing factor mitochondria-associated 2 (AIFM2; also known as FSP1)-coenzyme Q (CoQ10) axis in the plasma membrane, which was discovered through CRISPR screening in Gpx4-deficient cells (53). CoQ10, also known as ubiquinone, is a coenzyme that helps protect cells from damage caused by free radicals. The role of CoQ10 in apoptosis and ferroptosis is determined by AIFM2. After AIFM2 transfers from mitochondria to the plasma membrane, it loses its pro-apoptotic activity and acquires an anti-ferroptotic effect due to its ability to reduce non-mitochondrial CoQ10 to ubiquinol using NADPH (53). AIFM2 can also reduce vitamin K to promote coagulation (54) or mediate cell membrane repair (55), suggesting that the inhibition of ferroptosis by AIFM2 may involve several mechanisms. Interestingly, increased expression of AIFM2 is positively associated with both overall survival and disease-specific survival in HCC (56), suggesting that AIFM2 may act as a tumor suppressor.
The second antioxidant system that backs up GPX4 in ferroptosis suppression is the dihydroorotate dehydrogenase (DHODH)-CoQ10 system in mitochondria, identified in cancer cells with low GPX4 expression (52). The enzyme DHODH converts CoQ10 to ubiquinol, which combats lipid peroxidation and protects cancer cells from ferroptosis (52). Among solid cancers, HCC has the highest levels of DHODH mRNA expression, and DHODH overexpression is correlated with tumor progression and metastasis (57). StAR-related lipid transfer domain containing 7 (STARD7), which is dual-localized to the intermembrane space of mitochondria and the cytosol after cleavage by the rhomboid protease presenilin-associated rhomboid-like (PARL), ensures the synthesis and transport of CoQ10 to the plasma membrane (58). Thus, STARD7 cleavage links the DHODH-CoQ10 system and the activation of the AIFM2-CoQ10 axis.
Recent advances in the study of ferroptosis have shown that stress proteins, such as nuclear protein 1 transcriptional regulator (NUPR1), can reduce the susceptibility to ferroptosis in HCC cells (59). Gaining a deeper understanding of the intricate relationships between various defense systems will shed light on the mechanisms regulating ferroptosis and could spur the development of targeted therapeutic strategies.
Membrane repair system
The membrane repair system is crucial for preserving the integrity and function of the plasma membrane, which is vital for cell survival and effective cellular communication. The system encompasses various pathways, such as those for the regrouping of lipids, the formation of lipid rafts, and the involvement of membrane-repair proteins like phospholipases, lipases, phosphatidylinositol transfer proteins, phosphatidylserine decarboxylases, and phospholipid flippases. SLC47A1, a lipid flippase, can prevent ferroptosis in cancer cells by facilitating lipid restructuring without affecting the transport of ferroptosis-inducing drugs such as erastin or RSL3 (27). An elevated expression of SLC47A1 has been observed in various cancers (including HCC), which may contribute to resistance to chemotherapy (60). As such, targeting SLC47A1 may be a promising approach to overcome resistance to ferroptosis in HCC cells.
The endosomal sorting complexes required for transport (ESCRTs) are multi-protein complexes involved in membrane trafficking and intracellular cargo sorting. They play a role in the formation and regulation of intraluminal vesicles within multivesicular endosomes, leading to the selective sorting and degradation of transmembrane proteins and lipids. The ESCRTs consist of four distinct complexes (ESCRT-0, -I, -II, and -III), each of which operates in a sequential and hierarchical manner to sort, package, and expel membrane cargos into intraluminal vesicles. The ESCRT-III machinery plays a crucial role in repairing the plasma membrane and preventing various forms of regulated necrosis, including necroptosis, pyroptosis, and ferroptosis (61, 62). The inhibition of charged multivesicular body protein 5 (CHMP5) or CHMP6, key components of ESCRT-III, increases ferroptosis susceptibility in HCC cells (63). The activation of ESCRT-III on cell membranes is dependent on calcium signaling, implying the role of calcium homeostasis and endoplasmic reticulum stress in the regulation of ferroptosis.
Ferroptosis inhibitors
Iron chelators, free radical-scavenging antioxidants, and ALOX inhibitors are the most commonly used inhibitors of ferroptosis (Fig. 1) (13). Two iron chelators are currently available and approved by the United States Food and Drug Administration: deferoxamine and deferasirox. These iron chelators effectively prevent ferroptosis by inhibiting iron-induced Fenton reaction and ROS generation. Ferrostatins (e.g., ferrostatin-1) and liproxstatins (e.g., liproxstatin-1) are radical-trapping antioxidants that slow down the rate of autoxidation of hydrocarbons (lipids) by reacting with chain-carrying peroxyl radicals (64). Most lipid peroxidation inhibitors, such as baicalein (65) and zileuton (66), target ALOX with variable activity in vitro and in vivo. Ferrostatins and liproxstatins may be more specific in preventing ferroptosis, and iron chelators and ALOX inhibitors have been previously reported to inhibit apoptosis and other cell deaths. Overall, the identification of ferroptosis requires the combination of different cell death inhibitors in vitro and in vivo.
Regulation of ferroptosis in HCC
In addition to the core regulators of ferroptosis summarized above, the susceptibility of HCC to ferroptosis can be regulated on a number of different levels, such as gene transcription, epigenetics, protein degradation, and solute macromolecule uptake.
Transcriptional mechanism
Ferroptosis involves the activation of multiple transcription factors. Below we summarize three relatively well-studied transcription factors involved in the regulation of ferroptosis in HCC (Fig. 2).
Figure 2. Master transcriptional regulation of ferroptosis in HCC.
(A) NFE2L2 is a crucial anti-ferroptotic transcription factor in HCC cells, as it regulates multiple genes involved in enhancing antioxidant capacity, reducing iron toxicity, and facilitating drug efflux. The expression of NFE2L2 is primarily controlled by the KEAP1-mediated degradation pathway through the proteasomal system. The level of KEAP1 is, in turn, regulated by autophagic degradation via SQSTM1. Multiple upstream kinases or enzymes regulate the sensitivity to ferroptosis by controlling the SQSTM1-KEAP1-NFE2L2 pathway in HCC cells. (B) TP53 plays a dual role in ferroptosis, depending on the expression of its target genes. Under normal circumstances, TP53 is degraded by MDM2. If MDM2 is absent, the stability of TP53 increases and its transcriptional function is further amplified through Ser 46 phosphorylation and the presence of the P47S variant. In addition to boosting PLTP expression, TP53-mediated inhibition of DPP4 leads to the suppression of ferroptosis. (C) YAP1 promotes ferroptosis by inducing the expression of TFRC, which increases iron uptake. The O-GlcNAcylation of YAP1, which is mediated by O-GlcNAc, enhances the activity of YAP1 and its ability to induce TFRC expression in HCC cells.
NFE2L2
The transcription factor NFE2-like BZIP transcription factor 2 (NFE2L2), also known as NRF2, plays a key role in maintaining redox balance and controlling antioxidant genes by binding to antioxidant response elements (AREs). The expression of NFE2L2 is elevated in HCC and has been implicated in promoting cell proliferation and advancing tumor progression, making it an independent predictor of outcomes in HCC patients (67). Furthermore, mutations in NFE2L2 (occurring in 5.1% of patients with HCC) have been implicated in HCC pathology (68). However, in Nfe2l2-deficient mice, chronic exposure to non-genotoxic hepatocarcinogens, such as pentachlorophenol and piperonyl butoxide, triggers oxidative stress and stimulates the development of pre-neoplastic lesions as well as their progression to HCC (69). These findings support the notion that oxidative stress has a dual role in liver tumorigenesis.
The protein NFE2L2 is involved in conferring cellular resistance against ferroptosis therapy in HCC (Fig. 2A). Ferroptosis activators, such as erastin and sorafenib, can increase NFE2L2 protein stability in HCC cells. This process is regulated by the autophagy receptor sequestosome 1 (SQSTM1/p62) that increases NFE2L2 expression through the inactivation of kelch-like ECH-associated protein 1 (KEAP1) (70). NFE2L2-mediated ferroptosis resistance in HCC cells is achieved by the upregulation of multiple target genes, including well-known NFE2L2-targeted genes like NAD(P)H quinone dehydrogenase 1 (NQO1), GPX4, SLC7A11, and ferritin heavy-chain 1 (FTH1), as well as the newly identified NFE2L2-targeted gene metallothionein 1G (MT1G) (71). MT1G has also been found to be a positive predictor of sorafenib responses in patients with HCC (72). In the context of sorafenib treatment, the upregulation of mRNAs coding for ATP binding cassette subfamily C member 5 (ABCC5) or sigma non-opioid intracellular receptor 1 (SIGMAR1) by NFE2L2 promotes ferroptosis resistance by promoting drug efflux or activating the system xc−-GSH-GPX4 pathway in HCC cells (73, 74). Further research is needed to determine whether the expression of ABCC5 or SIGMAR1 is related to the effectiveness of sorafenib treatment in a clinical setting.
Radiotherapy has been used in the treatment of HCC for many years. It not only triggers apoptosis but also induces ferroptosis. In HCC cells, the expression of adiponectin receptor 1 (ADIPOR1) is upregulated and actually constitutes a negative prognostic biomarker after stereotactic body radiation therapy (75). ADIPOR1 is a mediator of resistance to radiotherapy-induced ferroptosis by activating NFE2L2-dependent SLC7A11 expression (75). However, it is not yet clear if adiponectin, the known ligand of ADIPOR1, has a similar effect in inhibiting ferroptosis. ADIPOR1-mediated changes in cell membrane composition may also play a role in impairing lipid metabolism during ferroptosis, independent of adiponectin (76).
Multiple mechanisms have been identified that regulate the expression or activity of NFE2L2, which in turn controls the sensitivity of HCC cells to ferroptosis. As a first example, the long non-coding RNA LINC01134 increases the expression of GPX4 by enhancing NFE2L2 binding to the GPX4 promoter, which in turn inhibits oxaliplatin-induced ferroptosis in HCC cells (77). The activation of the LINC01134-NFE2L2-GPX4 pathway is also associated with oxaliplatin resistance in HCC patients (77). As a second example, the enzyme glutathione S-transferase zeta 1 (GSTZ1), which is involved in phenylalanine catabolism, suppresses NFE2L2 expression and enhances sorafenib-induced ferroptosis in HepG2 and SNU449 cells or in xenograft models (78). The GPX4 inhibitor RSL3 can restore the sensitivity of GSTZ1−/− HCC cells or Gstz1−/− mice to sorafenib, suggesting that NFE2L2-mediated GPX4 expression contributes to sorafenib resistance (78). As a third example, quiescin-sulfhydryl oxidase 1 (QSOX1), a disulfide catalyst, enhances sorafenib-induced ferroptosis by inhibiting epidermal growth factor (EGF)/epidermal growth factor receptor (EGFR)-dependent NFE2L2 activation in HCC cells (79). The negative correlation between QSOX1 and NFE2L2 expression in HCC patient tumor tissues further supports the idea that QSOX1 is a negative regulator of antioxidant defense (79). Given that anti-EGFR antibodies like cetuximab can increase the susceptibility of KRAS-mutant colorectal cancer cells to ferroptosis (80), it may be worth testing the combination of cetuximab and sorafenib in inducing ferroptosis in HCC. Finally, the inhibition of several upstream kinases (e.g., TANK binding kinase 1 [TBK1]) or enzymes (e.g., pro-protein convertase subtilisin/kexin type 9 [PCSK9]) can cause NFE2L2 degradation through the SQSTM1-KEAP1/NFE2L2 pathway, thereby promoting ferroptosis in HCC cells (81, 82).
TP53
Tumor protein p53 (TP53) is one of the most eminent tumor suppressors. Approximately 30% of HCC cases are associated with TP53 gene mutations, making it the second-most frequent gene mutation in HCC (83). TP53 is activated in the hepatocytes of individuals with chronic liver disease, which can be caused by viruses, alcohol use, and fat accumulation. Interestingly, in mice lacking the Mdm2 proto-oncogene, causing TP53 accumulation in hepatocytes, TP53 paradoxically promoted HCC development by inducing excessive cell death, chronic inflammation, and a compensatory expansion of hepatic progenitor cells (84). This suggests that TP53 may play an unrecognized role in liver tumorigenesis by selectively sustaining cancer stem cell growth in a cell death-initiated inflammatory tumor microenvironment.
The role of TP53 in ferroptosis is a subject of ongoing research, but most studies suggest that TP53 plays a pro-ferroptotic role in HCC cells by inducing protocadherin beta 14 (PCDHB14) to inhibit the expression of SLC7A11 that is mediated by nuclear factor kappa B (NF-κB) (85) (Fig. 2B). This process is also regulated through the phosphorylation of TP53 on serine 46, which is activated by cell death signals to increase TP53 transcriptional activity. Zinc finger and SCAN domain containing 25 (ZSCAN25, also known as ZNF498) is a transcriptional regulator that can suppress apoptosis and ferroptosis in HepG2 cells by inhibiting serine 46 phosphorylation of TP53 by the complex formed by protein kinase C delta (PRKCD/PKCδ) and tumor protein p53-inducible nuclear protein 1 (TP53INP1) (86). In addition to promoting diethylnitrosamine-induced liver tumorigenesis in mice, higher levels of ZSCAN25 in tumors have been linked to a lower survival rate in HCC patients, suggesting that ZSCAN25 may have an oncogenic effect due to its anti-TP53 activity in apoptosis and ferroptosis (86). Whether targeting ZSCAN25-mediated TP53 inhibition is a potential strategy to restore TP53 tumor suppressor function will require further validation in transgenic animal models.
Glutaminase 2 (GLS2) is a mitochondrial enzyme that is highly expressed in the liver and is responsible for the hydrolysis of glutamine to glutamate in mitochondria. A reduced expression of GLS2 is common in HCC patients, and reducing its expression promotes HCC growth by activating the phosphoinositide 3-kinase (PI3K)-AKT-mammalian target of rapamycin (MTOR) pathway, which is a negative regulator of ferroptosis (87). These findings also suggest that TP53 may increase GLS2 expression to promote ferroptosis in HCC cells (87). However, the induced expression of GLS2 not only promotes ferroptosis but also mediates oxidative defense (88). More work is needed to define the exact role of GLS2 in modulating oxidative protection and damage.
Finally, TP53 has been shown to promote the upregulation of phospholipid transfer protein (PLTP), which transfers phospholipids from triglyceride-rich lipoproteins into high density lipoprotein. Mice carrying the P47S variant of TP53 (which cannot undergo phosphorylation of the adjacent serine 46) are highly susceptible to HCC (89). TP53-mediated PLTP upregulation promotes lipid droplet formation and confers resistance to ferroptosis in HepG2 cells with the P47S variant (90). Of note, TP53 does not only act as a transcription factor but also acts outside of the nucleus (91). Thus, TP53 suppresses ferroptosis in HCC by directly inhibiting dipeptidyl peptidase 4 (DPP4)-mediated NOX activation (92). Taken together with previous studies in colorectal cancer (93), these findings suggest that TP53 may exert an anti-ferroptotic role in HCC. It will be important to understand which gene-transactivating and extranuclear effects of TP53 contribute to a probably context-dependent modulation of ferroptosis.
YAP1
Yes1-associated transcriptional regulator (YAP1) is the mammalian equivalent of the Drosophila gene Yorkie (Yki) and plays an important role in the regulation of cell development, growth, repair, and homeostasis. YAP1 acts as a transcriptional coactivator and is regulated by the Hippo signaling pathway. YAP1 dysfunction has been linked to the development and progression of various cancers, including HCC, where it is expressed in approximately 62% of cases (94). The expression of YAP1 in HCC is correlated with a higher serum level of alpha-fetoprotein and poor tumor differentiation (95).
In HCC cells, YAP1 promotes ferroptosis by increasing the expression of TF receptor (TFRC, also known TFR1 or CD71), leading to the accumulation of intracellular free iron and ROS production (Fig. 2C). The O-GlcNAcylation of YAP1, which is a posttranscriptional modification catalyzed by O-linked N-acetylglucosamine (O-GlcNAc), can further enhance the activation of TFRC expression and promote ferroptosis in Bel-7402 and SMMC-7721 HCC cells (96). In contrast, O-GlcNAcylation of the transcription factor c-JUN on serine 73 can suppress ferroptosis by stimulating the synthesis of GSH without affecting phospholipids and Fe2+ (97). Thus, targeting O-GlcNAcylation to modulate ferroptosis sensitivity requires consideration of its protein substrates.
Epigenetic mechanism
The modification of RNA by N6-methyladenosine (m6A) is prevalent in eukaryotic messenger RNAs (mRNAs) and influences ferroptosis in HCC cells. The translation of SLC7A11 RNA into protein is facilitated by m6A modification, leading to resistance against ferroptosis (98). The expression of SLC7A11 is suppressed by the core component of the m6A methyltransferase complex, methyltransferase 14, N6-adenosine-methyltransferase subunit (METTL14), which has a tumor-suppressive effect under hypoxia in HCC (99). Conversely, DAZ-associated protein 1 (DAZAP1), which is oncogenic in HCC, can increase resistance to ferroptosis by directly binding to the 3’UTR of SLC7A11 mRNA, thus promoting SLC7A11 protein expression (100). Furthermore, various RNA molecules, including circular RNAs (e.g., CircIL4R), miRNAs (e.g., miR-541-3p), and lncRNAs (e.g., LINC01134, lncFAL, HEPFAL, and GABPB1-AS1), can regulate the sensitivity of HCC cells to ferroptosis by targeting the mRNAs of GPX4, AIFM2, or peroxiredoxin 5 (PRDX5), thus impacting cellular antioxidant capacity (101-104). Collectively, these studies may provide information for the development of ferroptosis-associated RNA drugs.
Protein degradation mechanism
The susceptibility of HCC cells to ferroptosis is influenced by two major intracellular degradation mechanisms: the ubiquitin–proteasome system and autophagy. Tribbles pseudokinase 2 (TRIB2) is a player in liver tumorigenesis, functioning as a molecular adapter that facilitates protein ubiquitination and destabilization. An increase in TRIB2 levels in Bel-7404 and SK-Hep1 HCC cells suppresses ferroptosis by promoting TFRC ubiquitination and reducing labile iron levels through the beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC/βTrCP) pathway (105). The downregulation of centrosomal protein 290 (CEP290) and ubiquitin-like modifier activating enzyme 1 (UBA1) reduces the level of NFE2L2 protein by the ubiquitin–proteasome pathway, leading to ferroptosis in HCC cells (106, 107).
Autophagy, especially selective autophagy, may also contribute to the ferroptotic demise of HCC cells (Fig. 3). As previously mentioned, RAB7A-mediated lipophagy increases lipid availability, thereby enhancing the sensitivity of HCC cells to ferroptosis (29). Activating nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy by erastin, sorafenib, or sulfasalazine can suppress SREBP1-mediated BCAT2 expression in HCC cells, and BCAT2 acts as a suppressor of ferroptosis by increasing intracellular glutamate and subsequent GSH production (108). These findings also establish a connection between protein degradation and the activation of transcription factors that affect lipid homeostasis and antioxidant production. Another example of this process is provided by artesunate-induced ferritinophagy-dependent ferroptosis in HCC cells (109), although the proximal target of artesunate remains elusive.
Figure 3. The role of autophagy in ferroptosis in HCC.
Autophagy is a degradation process that occurs within lysosomes and involves the formation of multiple membrane structures to engulf cellular components such as organelles or proteins. Nonselective autophagy promotes cell survival by recycling and reusing degraded materials to produce energy. Conversely, selective autophagy, including ferritinophagy and lipophagy, can trigger ferroptosis by increasing the availability of free iron and PUFAs. The protein BECN1 can bind to SLC7A11, limiting the production of GSH and the activation of GPX4. On the other hand, PNO1-mediated autophagy can prevent ferroptosis in HCC cells by increasing the synthesis of GSH.
Beclin 1 (BECN1) is a crucial regulator of autophagy that can bind SLC7A11 and inhibit GSH production, thereby suppressing sorafenib-induced ferroptosis (110). HCC cells with a high expression of GABARAPL1 (a member of the LC3 superfamily of proteins required for autophagosome formation) are particularly vulnerable to ferroptosis caused by erastin and sorafenib (111). However, recent research shows that partner of NOB1 homolog (PNO1)-induced autophagy can inhibit sorafenib-induced ferroptosis by promoting GSH biosynthesis in Hep3B and HLE HCC cells (112), highlighting the dual role of autophagy in cell survival and death in the context of ferroptosis stimulation. Further investigation is required to clarify the specific targets degraded by autophagy and how they impact ferroptosis in positive or negative terms.
Cellular uptake mechanism
The process of micropinocytosis, through which molecules dissolved in extracellular fluid cross the cell membrane in small vesicles, has been found to play a significant role in preventing sorafenib-induced ferroptosis in HCC cells (Fig. 4). The activation of the PI3K-Rac family small GTPase 1 (RAC1)-ribonuclease A family member 9 (RNASE9/PAK1) signaling pathway, triggered by mitochondrial damage, is responsible for sorafenib-driven macropinocytosis in HCC cells (113). Amiloride, an inhibitor of micropinocytosis, enhances the capacity of sorafenib in killing HCC cells (113). Indeed, sorafenib-resistant HCC cells are characterized by high levels of macropinocytosis, and suppressing micropinocytosis makes these resistant tumors more responsive to sorafenib treatment (113). Targeting micropinocytosis therefore holds a potential to reduce the development of sorafenib resistance and to improve therapeutic outcomes.
Figure 4. The role of micropinocytosis in ferroptosis in HCC.
Sorafenib can suppress the activity of system xc−, reducing cystine uptake and the subsequent synthesis of cysteine and GSH in HCC cells. This is essential for the functioning of GPX4, which helps prevent lipid peroxidation and ferroptosis. However, sorafenib's impact on the mitochondria can trigger a negative feedback mechanism, activating the PI3K-RAC1-RNASE9 pathway. This results in micropinocytosis-mediated uptake of extracellular protein, producing more cysteine to counteract the effects of ferroptosis.
Ferroptosis in HCC tumorigenesis
Inflammation-associated immune suppression is often linked to the development and progression of cancer, and a ferroptosis-induced macrophage polarization has been shown to promote pancreatic cancer tumorigenesis through the use of Pdx1-Cre;KrasG12D/+;Gpx4−/− mice (114). Similarly, the loss of hepatocyte-restricted GPX4 in mice does not suppress hepatocellular tumorigenesis (115). Instead, Gpx4 depletion induces ferroptotic death of hepatocytes, which triggers a tumor-suppressive microenvironment characterized by the upregulation of immune checkpoints and the infiltration of myeloid-derived suppressor cells (MDSCs) mediated by high-mobility group box 1 (HMGB1) (115). Thus, ferroptotic damage-mediated inflammation-associated immune suppression may promote the tumorigenesis of HCC. This is also supported by several studies that have shown that iron overload in HCC can drive tumorigenesis, proliferation, and growth (116). Additionally, ferroptosis is related to both acute liver diseases (e.g., carbon tetrachloride-induced liver injury (117)) and chronic liver diseases (e.g., hepatitis B/C (118, 119), non-alcoholic fatty liver disease (120-123), and hereditary hemochromatosis (124)), which can progress to HCC (Fig. 5). The overexpression of the ferroptosis core promoter ACSL4 in HCC patients has been linked to poor patient outcomes (31). The depletion of ACSL4 in HCC cells can effectively inhibit cell proliferation by disrupting the c-Myc-SREBP1 pathway (31). Furthermore, lipid peroxidation-derived DNA adducts, such as γ-OHPdG, have been shown to promote hepatocarcinogenesis in mice (125). Therefore, reducing exposure to risk factors that cause ferroptotic damage could help prevent HCC.
Figure 5. The dual roles of ferroptosis in HCC.
On the one hand, ferroptotic damage has been linked to a number of acute or chronic liver diseases that can lead to HCC. On the other hand, inducing ferroptosis through chemotherapy, immunotherapy (anti-PDCD/PD-1 or anti-CD274/PD-L1 antibodies) or radiotherapy can be effective in suppressing tumor growth in established HCC.
Ferroptosis in HCC therapy
The induction of ferroptosis may constitute a strategy for combating established HCC (Fig. 5). There are several compounds and clinical drugs that have been used to trigger ferroptosis in HCC (Table 2). As described above, classical inducers, such as erastin and RSL3, inhibit the system xc−-GSH-GPX4 pathway. Among ferroptosis-inducing drugs, sorafenib is most commonly used to treat HCC. Sorafenib is an oral multi-kinase inhibitor used in the treatment of advanced renal cell carcinoma, liver cancer, and thyroid cancer. Sorafenib was initially characterized as an apoptosis inducer, and this activity was attributed to its action on oncogenic kinases (126). The mechanism of ferroptosis induced by sorafenib is not mediated by kinase inhibition (127). Indeed, high doses of sorafenib (10 μM) may induce ferroptosis by directly inhibiting SLC7A11, although this is controversial (128). Regardless, cancer cells display varying degrees of sensitivity to ferroptosis induction upon the inhibition of system xc−, as the transsulfuration pathway can bypass system xc− by synthesizing cysteine from methionine to maintain GSH synthesis (129).
Table 2.
Examples of ferroptosis inducers and inhibitors
Name | Mode of action | Application | References |
---|---|---|---|
Ferroptosis inducers | |||
Abemaciclib | Inhibits CDK4/6 | Preclinical and clinical | (130) |
Alkaloid trigonelline | Inhibits NFE2L2 | Preclinical | (70) |
Anthracyclins | Increases PUFA | Preclinical | (149) |
Artesunate | Induces ferritinophagy | Preclinical and clinical | (109) |
Disulfiram | Induces mitochondrial damage | Preclinical and clinical | (150) |
Erastin | Inhibits SLC7A11 | Preclinical | (70) |
Haloperidol | Inhibits SIGMAR1 | Preclinical | (74) |
Oxaliplatin | Induces ROS production | Preclinical and clinical | (77) |
RSL3 | Inhibits GPX4 | Preclinical | (21) |
Saponin formosanin C | Induces ferritinophagy | Preclinical | (18) |
Solasonine | Inhibits GPX4 | Preclinical | (151) |
Sorafenib | Inhibits SLC7A11 | Preclinical and clinical | (128) |
Tiliroside | Inhibits TBK1 and induces NFE2L2 degradation | Preclinical | (82) |
Vanadium -iIron -oOxide | Inhibits GPX4 | Preclinical | (152) |
ZZW-115 | Inhibits NUPR1 | Preclinical | (59) |
Ferroptosis inhibitors | |||
Deferoxamine | Iron chelator | Preclinical and clinical | (153) |
Deferasirox | Iron chelator | Preclinical and clinical | (154) |
Ferrostatin-1 | Free radical-scavenging antioxidant | Preclinical | (153) |
Liproxstatin-1 | Free radical-scavenging antioxidant | Preclinical | (153) |
Baicalein | Inhibits ALOX | Preclinical and clinical | (65) |
Zileuton | Inhibits ALOX | Preclinical and clinical | (66) |
Abemaciclib, a cyclin-dependent kinase (CDK) inhibitor selective for CDK4 and CDK6, has been clinically approved for the treatment of advanced metastatic breast cancer. Theoretically, the combination of abemaciclib and sorafenib has the potential to improve patient outcomes in HCC by inducing synthetic lethality. However, phosphoseryl-TRNA kinase (PSTK) has been identified as a mediator of resistance to abemaciclib and sorafenib treatment, inhibiting ferroptosis induction in HCC through the activation of GPX4 (130).
Oxaliplatin, a third-generation platinum drug used as first-line chemotherapy for colorectal cancer, can mediate resistance to ferroptosis induction in HCC through the expression of NFE2L2-mediated GPX4 (77). Radiotherapy has also been reported to induce ferroptosis in HCC cells by inhibiting SLC7A11 (131) or SLC1A5 (132). The use of engineered Man@pSiNPs-erastin has the potential to boost the effectiveness of immune checkpoint inhibitors (anti-PDCD/PD-1 or anti-CD274/PD-L1 antibodies) in an HCC model by suppressing pro-tumor polarization of macrophages (133). In addition, inhibitors of NFE2L2, such as alkaloid trigonelline, are expected to enhance the sensitivity of HCC cells to ferroptosis in cell cultures and in mouse models (70).
There are also natural products, metal-containing drugs, and nanoparticles that have been identified as potential ferroptosis inducers in HCC cells (examples are provided in Table 2). These findings have provided valuable pharmacological tools for investigating the signals and mechanisms of ferroptosis. However, they have also raised concerns about the specificity of ferroptosis-inducing agents.
Monitoring ferroptosis sensitivity
Non-invasive methods that measure the mechanisms and dynamics of ferroptosis would be extremely useful for understanding the diversity of HCC patients with respect to ferroptosis-inducing treatments, including drug therapy with sorafenib, alone or in combination with other drugs.
Effective monitoring of the response to drug-induced ferroptosis in cancer patients requires the use of sensitive and specific biomarkers, preferentially in serum or plasma. Mass spectrometry can be used to analyze changes in serum iron or lipid metabolites, while enzyme-linked immunosorbent assays (ELISAs) of damage-associated molecular patterns (DAMPs) can offer a quick, simple, and cost-effective way to assess ferroptosis. DAMPs, such as HMGB1 (134), are molecules released by dead or dying cells, irrespective of the specific death modality. Ferroptotic cells specifically release decorin (DCN) (135), a proteoglycan from the small leucine-rich proteoglycan family. In healthy livers, small amounts of decorin are found around central veins and in portal tracts, but during fibrogenesis, its levels significantly increase in connective tissue septa. Studies in mice have shown that decorin can inhibit liver carcinogenesis (136, 137). Nonetheless, pending further investigation, an elevation in serum DCN concentrations might indicate a therapeutically relevant sorafenib-induced ferroptosis response. Ferroptotic cells also release prostaglandin E2 (PGE2), which may inhibit the antitumor activity of immune cells (138). Whether the quantitation of circulating PGE2 provides information on the efficacy of sorafenib has not been determined.
Iron probes are valuable tools for measuring iron levels in different biological samples. Chromogenic methods using iron probes provide a convenient method for the simultaneous measurement of Fe2+ and Fe3+ (139). Fluorescent iron probes such as FerroFarRed, Calcein-AM, and FRET Iron Probe 1 are also widely used to analyze the dynamic changes of iron levels during ferroptosis. Lipid hydroperoxides are the major product of peroxidation reactions and can be measured directly by high-performance liquid chromatography (HPLC) with chemiluminescent detection. Furthermore, combining C11-BODIPY(591/581) staining with flow cytometry measurements can provide multiparametric information on intracellular oxidative stress damage in single cells during ferroptosis. High-throughput lipidomics can comprehensively assess abnormal lipid metabolism in ferroptosis.
Finally, immunohistochemistry analyses of key ferroptosis regulators, such as ACSL4, GPX4, SLC7A11, and TFRC, in surgically resected liver tumor samples can provide valuable insights into treatment response. In particular, ACSL4 has been shown to predict the prognosis of sorafenib-treated HCC patients, especially those experiencing sorafenib-mediated ferroptosis (140).
Clinical application of a ferroptosis scoring system in HCC
Bioinformatics has revealed a potential clinical connection between the ferroptosis-related genes and HCC patients. A prognostic scoring system has been established based on the expression of 25 ferroptosis-related genes in public TCGA, CGGA, and GEO databases. A consensus clustering analysis of 371 HCC cancer samples resulted in 3 independent clusters (141). Cluster 3 was found to have higher expression levels, an unfavorable prognosis, and higher histological tumor stage and grade compared to clusters 1 and 2. The expression of HSPA5, EMC2, SLC7A11, HSPB1, GPX4, FANCD2, CISD1, FDFT1, SLC1A5, TFRC, RPL8, DPP4, CS, CARS1, ATP5MC3, ALOX15, ACSL4, and ATL1 was significantly increased in HCC tissue compared to normal liver tissue (141). Conversely, the expression of NFE2L2, MT1G, SAT1, and GLS2 was decreased in HCC tissue compared to normal liver tissue (141). Among these genes, the increased expression of SLC7A11, SLC1A5, TFRC, RPL8, and CARS1 was associated with unfavorable overall survival in HCC patients (141). Another study assessed 374 HCC tumor samples using 214 ferroptosis-related genes from the FerrDb database (142). This study identified a four-gene signature for overall survival in HCC, consisting of GPX2, MT3, PRDX1, and SRXN1. PRDX1 was found to be the hub gene of the prognosis model and had high expression in HCC tumor tissue (142). On the other hand, researchers assessed 104 ferroptosis- and iron metabolism-related genes and found that a prognosis model constructed with four genes (ABCB6, FLVCR1, SLC48A1, and SLC7A11) independently predicted the prognosis of HCC patients with superior accuracy (143). The median value of the ferroptosis score was used to classify HCC patients into high- and low-ferroptosis score groups. HCC patients with high-ferroptosis scores exhibited a stronger immunosuppressive tumor microenvironment (143). More recently, 10 ferroptosis-related genes (UGT1A6, ATP6V1C1, MAFG, NUDCD1, PPP1R1A, TSKU, CTSB, AIFM2, CTSA, and CTNND2) were found to be posttranscriptionally regulated by RNA modifications in HCC (56).
Collectively, ever more sophisticated in silico analyses of big data could lead to the development of a predictive and prognostic ferroptosis scoring system applicable to HCC patients. To achieve this goal, it will be crucial to increase sample size, to gather more comprehensive clinical information on each HCC case, and to validate all results in independent cohorts. Additionally, it will be interesting to monitor ferroptosis-related molecular changes over time to understand their prognostic and predictive value for treatment responses and the subsequent resistance-associated relapse.
Conclusion and perspective
The discovery of ferroptosis as a type of non-apoptotic cell death has sparked numerous preclinical studies examining its contribution to chemotherapy, radiotherapy, or immunotherapy responses in HCC. Ferroptosis exhibits different in vitro and in vivo features than the well-studied apoptosis (Table 3). The effect of ferroptosis on liver cancer progression is complex and can vary at different stages of the tumor. Further research into the factors that influence the antitumor immune response in ferroptosis is crucial for developing new treatments and combination therapies. Of note, HCC patients often exhibit abnormal iron absorption and lipid metabolism disorders, which play a significant role in regulating ferroptosis. An aberrant expression of several ferroptosis regulators is also associated with poor HCC prognosis (Table 1).
Table 3.
Differences between ferroptosis and apoptosis
Ferroptosis | Apoptosis | |
---|---|---|
Morphological features | Enlarged and swollen cells with reduced mitochondrial volume and plasma membrane rupture | Cell shrinkage, membrane blebbing, chromatin condensation, and nuclear fragmentation |
Biochemical properties | Iron accumulation and lipid peroxidation | Caspases activation in many cases |
Immunological features | Pro-inflammatory in many cases | Anti-inflammatory in many cases |
Key positive regulators | ACSL4, POR, and ALOX | CASP3, CASP7, CASP8, and CASP9 |
Key negative regulators | SLC7A11, GPX4, AIFM2, DHODH, and NFE2L2 | BCL2, BCL2L1/BCLXL, and MCL1 |
Autophagy response | Increased autophagy promotes ferroptosis | Increased autophagy inhibits apoptosis |
In vivo intervention | Ferrostatin-1 or liproxstatin-1 inhibits ferroptotic damage in various tissues | Caspase inhibitor Z-VAD-FMK alleviates apoptotic damage in various tissues |
Biomarkers | ACSL4, DCN, PTGS2/COX2, and TFRC | Cleaved-CASP3, Cleaved-CAPS8, and Cleaved-PARP |
Despite progress in the area of ferroptosis, translating ferroptosis research into effective treatments for patients with HCC remains a challenge. One obstacle is that key regulators of ferroptosis, such as GPX4 and SLC7A11, are widely expressed throughout the body, making it difficult to target them without causing side effects. Additionally, multiple antioxidant systems can inhibit ferroptosis, meaning that targeting one single pathway may not be sufficient for a lasting response. Although the mechanism of ferroptosis is highly context-dependent, in many cases the deletion of the key ferroptosis promoters ACSL4 or POR can demonstrate ferroptosis in vivo and in vitro. A further challenge is the absence of confirmed biomarkers that can differentiate ferroptotic from non-ferroptotic cell death, making it difficult to determine the effectiveness of potential treatments. Combined analysis of specific lipid peroxide products and DAMPs (e.g., DCN) in serum may be a relatively sensitive and specific method for detecting ferroptosis in vivo.
To address these obstacles, gaining a deeper understanding of the signals, mechanisms, and modulations of ferroptosis in response to specific agents or combinations is crucial. Understanding the sensitivity and resistance of ferroptosis is a key objective. This requires the development of drugs specifically designed to induce ferroptosis in cancer cells while avoiding normal cells. Furthermore, the identification of accurate and easily accessible biomarkers will be instrumental in developing personalized ferroptotic therapies.
Acknowledgments
We thank Dave Primm (Department of Surgery, University of Texas Southwestern Medical Center) for his critical reading of the manuscript. Research by D.T. and R.K. was supported by grants from the National Institutes of Health (R01CA160417, R01CA229275, and R01CA211070).
Footnotes
Conflict of interest
Guido Kroemer has received grants from Daiichi Sankyo, Eleor, Kaleido, Lytix Pharma, PharmaMar, Osasuna Therapeutics, Samsara Therapeutics, Sanofi, Tollys, and Vascage. He has consulted for Reithera, and is on the board of directors of the Bristol Myers Squibb Foundation, France. He is a scientific co-founder of everImmune, Osasuna Therapeutics, Samsara Therapeutics, and Therafast Bio. He is on the scientific advisory boards of Hevolution, Institut Servier, and Longevity Vision Funds. He is the holder of patents covering therapeutic targeting of aging, cancer, cystic fibrosis, and metabolic disorders. His wife, Laurence Zitvogel, has received grants from GlaxoSmithKline, Incyte, Lytix, Kaleido, Innovate Pharma, Daiichi Sankyo, Pilege, Merus, Transgene, 9 m, Tusk, and Roche, was on the board of directors of Transgene, is a co-founder of everImmune, and holds patents covering the treatment of cancer and the therapeutic manipulation of the microbiota. His brother, Romano Kroemer, was employed by Sanofi and consults for Boehringer-Ingelheim. The funders had no role in the writing of the manuscript, or in the decision to publish the results. The remaining authors have no conflicts to report.
References
- 1.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209–249. [DOI] [PubMed] [Google Scholar]
- 2.Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:6. [DOI] [PubMed] [Google Scholar]
- 3.Llovet JM, Montal R, Sia D, Finn RS. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol 2018;15:599–616. [DOI] [PubMed] [Google Scholar]
- 4.Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med 2020;382:1894–1905. [DOI] [PubMed] [Google Scholar]
- 5.Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, 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]
- 6.Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res 2019;29:347–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012;149:1060–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Liu J, Kang R, Tang D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J 2022;289:7038–7050. [DOI] [PubMed] [Google Scholar]
- 9.Chen J, Li X, Ge C, Min J, Wang F. The multifaceted role of ferroptosis in liver disease. Cell Death Differ 2022;29:467–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ajoolabady A, Tang D, Kroemer G, Ren J. Ferroptosis in hepatocellular carcinoma: mechanisms and targeted therapy. Br J Cancer 2023;128:190–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol 2021;18:280–296. [DOI] [PubMed] [Google Scholar]
- 12.Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Chen X, Li J, Kang R, Klionsky DJ, Tang D. Ferroptosis: machinery and regulation. Autophagy 2021;17:2054–2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tang D, Chen X, Kang R, Kroemer G. Ferroptosis: molecular mechanisms and health implications. Cell Res 2021;31:107–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Chen X, Huang J, Yu C, Liu J, Gao W, Li J, Song X, et al. A noncanonical function of EIF4E limits ALDH1B1 activity and increases susceptibility to ferroptosis. Nat Commun 2022;13:6318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.De Minicis S, Brenner DA. NOX in liver fibrosis. Arch Biochem Biophys 2007;462:266–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Olynyk JK, St Pierre TG, Britton RS, Brunt EM, Bacon BR. Duration of hepatic iron exposure increases the risk of significant fibrosis in hereditary hemochromatosis: a new role for magnetic resonance imaging. Am J Gastroenterol 2005;100:837–841. [DOI] [PubMed] [Google Scholar]
- 18.Lin PL, Tang HH, Wu SY, Shaw NS, Su CL. Saponin Formosanin C-induced Ferritinophagy and Ferroptosis in Human Hepatocellular Carcinoma Cells. Antioxidants (Basel) 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhang Z, Yao Z, Wang L, Ding H, Shao J, Chen A, Zhang F, 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]
- 20.Yu Y, Jiang L, Wang H, Shen Z, Cheng Q, Zhang P, Wang J, 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]
- 21.Shang Y, Luo M, Yao F, Wang S, Yuan Z, Yang Y. Ceruloplasmin suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma cells. Cell Signal 2020;72:109633. [DOI] [PubMed] [Google Scholar]
- 22.Yao F, Deng Y, Zhao Y, Mei Y, Zhang Y, Liu X, Martinez C, et al. A targetable LIFR-NF-kappaB-LCN2 axis controls liver tumorigenesis and vulnerability to ferroptosis. Nat Commun 2021;12:7333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lu T, Li C, Xiang C, Gong Y, Peng W, Chen C. Overexpression of CISD1 Predicts Worse Survival in Hepatocarcinoma Patients. Biomed Res Int 2022;2022:7823191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Yuan H, Li X, Zhang X, Kang R, Tang D. CISD1 inhibits ferroptosis by protection against mitochondrial lipid peroxidation. Biochem Biophys Res Commun 2016;478:838–844. [DOI] [PubMed] [Google Scholar]
- 25.Lin Z, Liu J, Kang R, Yang M, Tang D. Lipid Metabolism in Ferroptosis. Adv Biol (Weinh) 2021;5:e2100396. [DOI] [PubMed] [Google Scholar]
- 26.Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol 2017;13:81–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lin Z, Liu J, Long F, Kang R, Kroemer G, Tang D, Yang M. The lipid flippase SLC47A1 blocks metabolic vulnerability to ferroptosis. Nat Commun 2022;13:7965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol Cell 2015;59:298–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bai Y, Meng L, Han L, Jia Y, Zhao Y, Gao H, Kang R, et al. Lipid storage and lipophagy regulates ferroptosis. Biochem Biophys Res Commun 2019;508:997–1003. [DOI] [PubMed] [Google Scholar]
- 30.Zou Y, Henry WS, Ricq EL, Graham ET, Phadnis VV, Maretich P, Paradkar S, et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 2020;585:603–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chen J, Ding C, Chen Y, Hu W, Yu C, Peng C, Feng X, et al. ACSL4 reprograms fatty acid metabolism in hepatocellular carcinoma via c-Myc/SREBP1 pathway. Cancer Lett 2021;502:154–165. [DOI] [PubMed] [Google Scholar]
- 32.Duan J, Wang Z, Duan R, Yang C, Zhao R, Feng Q, Qin Y, et al. Therapeutic targeting of hepatic ACSL4 ameliorates NASH in mice. Hepatology 2022;75:140–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Magtanong L, Ko PJ, To M, Cao JY, Forcina GC, Tarangelo A, Ward CC, et al. Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State. Cell Chem Biol 2019;26:420–432 e429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bansal S, Berk M, Alkhouri N, Partrick DA, Fung JJ, Feldstein A. Stearoyl-CoA desaturase plays an important role in proliferation and chemoresistance in human hepatocellular carcinoma. J Surg Res 2014;186:29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhao Y, Li M, Yao X, Fei Y, Lin Z, Li Z, Cai K, et al. HCAR1/MCT1 Regulates Tumor Ferroptosis through the Lactate-Mediated AMPK-SCD1 Activity and Its Therapeutic Implications. Cell Rep 2020;33:108487. [DOI] [PubMed] [Google Scholar]
- 36.Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U S A 2016;113:E4966–4975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zou Y, Li H, Graham ET, Deik AA, Eaton JK, Wang W, Sandoval-Gomez G, et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol 2020;16:302–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cao F, Luo A, Yang C. G6PD inhibits ferroptosis in hepatocellular carcinoma by targeting cytochrome P450 oxidoreductase. Cell Signal 2021;87:110098. [DOI] [PubMed] [Google Scholar]
- 39.Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 2013;138:103–141. [DOI] [PubMed] [Google Scholar]
- 40.Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, Kang R, et al. Ferroptosis: process and function. Cell Death Differ 2016;23:369–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Su LJ, Zhang JH, Gomez H, Murugan R, Hong X, Xu D, Jiang F, et al. Reactive Oxygen Species-Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis. Oxid Med Cell Longev 2019;2019:5080843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hirata Y, Cai R, Volchuk A, Steinberg BE, Saito Y, Matsuzawa A, Grinstein S, et al. Lipid peroxidation increases membrane tension, Piezo1 gating, and cation permeability to execute ferroptosis. Curr Biol 2023. [DOI] [PubMed] [Google Scholar]
- 43.Chen X, Song X, Li J, Zhang R, Yu C, Zhou Z, Liu J, et al. Identification of HPCAL1 as a specific autophagy receptor involved in ferroptosis. Autophagy 2022:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014;156:317–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lee J, Roh JL. SLC7A11 as a Gateway of Metabolic Perturbation and Ferroptosis Vulnerability in Cancer. Antioxidants (Basel) 2022;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tadokoro T, Ikeda M, Ide T, Deguchi H, Ikeda S, Okabe K, Ishikita A, et al. Mitochondria-dependent ferroptosis plays a pivotal role in doxorubicin cardiotoxicity. JCI Insight 2020;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Guo W, Zhao Y, Zhang Z, Tan N, Zhao F, Ge C, Liang L, et al. Disruption of xCT inhibits cell growth via the ROS/autophagy pathway in hepatocellular carcinoma. Cancer Lett 2011;312:55–61. [DOI] [PubMed] [Google Scholar]
- 48.Guerriero E, Capone F, Accardo M, Sorice A, Costantini M, Colonna G, Castello G, et al. GPX4 and GPX7 over-expression in human hepatocellular carcinoma tissues. Eur J Histochem 2015;59:2540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Carlson BA, Tobe R, Yefremova E, Tsuji PA, Hoffmann VJ, Schweizer U, Gladyshev VN, et al. Glutathione peroxidase 4 and vitamin E cooperatively prevent hepatocellular degeneration. Redox Biol 2016;9:22–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kang R, Zeng L, Zhu S, Xie Y, Liu J, Wen Q, Cao L, et al. Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis. Cell Host Microbe 2018;24:97–108 e104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Canli O, Alankus YB, Grootjans S, Vegi N, Hultner L, Hoppe PS, Schroeder T, et al. Glutathione peroxidase 4 prevents necroptosis in mouse erythroid precursors. Blood 2016;127:139–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Grocin AG, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019. [DOI] [PubMed] [Google Scholar]
- 54.Mishima E, Ito J, Wu Z, Nakamura T, Wahida A, Doll S, Tonnus W, 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]
- 55.Dai E, Zhang W, Cong D, Kang R, Wang J, Tang D. AIFM2 blocks ferroptosis independent of ubiquinol metabolism. Biochem Biophys Res Commun 2020;523:966–971. [DOI] [PubMed] [Google Scholar]
- 56.Zhu R, Gao C, Feng Q, Guan H, Wu J, Samant H, Yang F, et al. Ferroptosis-related genes with post-transcriptional regulation mechanisms in hepatocellular carcinoma determined by bioinformatics and experimental validation. Ann Transl Med 2022;10:1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhou Y, Tao L, Zhou X, Zuo Z, Gong J, Liu X, Zhou Y, et al. DHODH and cancer: promising prospects to be explored. Cancer Metab 2021;9:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Deshwal S, Onishi M, Tatsuta T, Bartsch T, Cors E, Ried K, Lemke K, et al. Mitochondria regulate intracellular coenzyme Q transport and ferroptotic resistance via STARD7. Nat Cell Biol 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Huang C, Santofimia-Castano P, Liu X, Xia Y, Peng L, Gotorbe C, Neira JL, et al. NUPR1 inhibitor ZZW-115 induces ferroptosis in a mitochondria-dependent manner. Cell Death Discov 2021;7:269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wittwer MB, Zur AA, Khuri N, Kido Y, Kosaka A, Zhang X, Morrissey KM, et al. Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling. J Med Chem 2013;56:781–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Liu J, Kang R, Tang D. ESCRT-III-mediated membrane repair in cell death and tumor resistance. Cancer Gene Ther 2021;28:1–4. [DOI] [PubMed] [Google Scholar]
- 62.Pedrera L, Espiritu RA, Ros U, Weber J, Schmitt A, Stroh J, Hailfinger S, et al. Ferroptotic pores induce Ca(2+) fluxes and ESCRT-III activation to modulate cell death kinetics. Cell Death Differ 2021;28:1644–1657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Dai E, Meng L, Kang R, Wang X, Tang D. ESCRT-III-dependent membrane repair blocks ferroptosis. Biochem Biophys Res Commun 2020;522:415–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Conrad M, Pratt DA. The chemical basis of ferroptosis. Nat Chem Biol 2019;15:1137–1147. [DOI] [PubMed] [Google Scholar]
- 65.Xie Y, Song X, Sun X, Huang J, Zhong M, Lotze MT, Zeh HJR, et al. Identification of baicalein as a ferroptosis inhibitor by natural product library screening. Biochem Biophys Res Commun 2016;473:775–780. [DOI] [PubMed] [Google Scholar]
- 66.Liu Y, Wang W, Li Y, Xiao Y, Cheng J, Jia J. The 5-Lipoxygenase Inhibitor Zileuton Confers Neuroprotection against Glutamate Oxidative Damage by Inhibiting Ferroptosis. Biol Pharm Bull 2015;38:1234–1239. [DOI] [PubMed] [Google Scholar]
- 67.Zhang M, Zhang C, Zhang L, Yang Q, Zhou S, Wen Q, Wang J. Nrf2 is a potential prognostic marker and promotes proliferation and invasion in human hepatocellular carcinoma. BMC Cancer 2015;15:531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Schulze K, Imbeaud S, Letouze E, Alexandrov LB, Calderaro J, Rebouissou S, Couchy G, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet 2015;47:505–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tasaki M, Kuroiwa Y, Inoue T, Hibi D, Matsushita K, Kijima A, Maruyama S, et al. Lack of nrf2 results in progression of proliferative lesions to neoplasms induced by long-term exposure to non-genotoxic hepatocarcinogens involving oxidative stress. Exp Toxicol Pathol 2014;66:19–26. [DOI] [PubMed] [Google Scholar]
- 70.Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R, Tang D. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology 2016;63:173–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sun X, Niu X, Chen R, He W, Chen D, Kang R, Tang D. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology 2016;64:488–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Houessinon A, Francois C, Sauzay C, Louandre C, Mongelard G, Godin C, Bodeau S, et al. Metallothionein-1 as a biomarker of altered redox metabolism in hepatocellular carcinoma cells exposed to sorafenib. Mol Cancer 2016;15:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Huang W, Chen K, Lu Y, Zhang D, Cheng Y, Li L, Huang W, et al. ABCC5 facilitates the acquired resistance of sorafenib through the inhibition of SLC7A11-induced ferroptosis in hepatocellular carcinoma. Neoplasia 2021;23:1227–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bai T, Lei P, Zhou H, Liang R, Zhu R, Wang W, Zhou L, et al. Sigma-1 receptor protects against ferroptosis in hepatocellular carcinoma cells. J Cell Mol Med 2019;23:7349–7359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Feng H, Liu Y, Gan Y, Li M, Liu R, Liang Z, Liu L, et al. AdipoR1 Regulates Ionizing Radiation-Induced Ferroptosis in HCC cells through Nrf2/xCT Pathway. Oxid Med Cell Longev 2022;2022:8091464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ruiz M, Stahlman M, Boren J, Pilon M. AdipoR1 and AdipoR2 maintain membrane fluidity in most human cell types and independently of adiponectin. J Lipid Res 2019;60:995–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kang X, Huo Y, Jia S, He F, Li H, Zhou Q, Chang N, et al. Silenced LINC01134 Enhances Oxaliplatin Sensitivity by Facilitating Ferroptosis Through GPX4 in Hepatocarcinoma. Front Oncol 2022;12:939605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wang Q, Bin C, Xue Q, Gao Q, Huang A, Wang K, Tang N. GSTZ1 sensitizes hepatocellular carcinoma cells to sorafenib-induced ferroptosis via inhibition of NRF2/GPX4 axis. Cell Death Dis 2021;12:426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sun J, Zhou C, Zhao Y, Zhang X, Chen W, Zhou Q, Hu B, et al. Quiescin sulfhydryl oxidase 1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by driving EGFR endosomal trafficking and inhibiting NRF2 activation. Redox Biol 2021;41:101942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chen P, Li X, Zhang R, Liu S, Xiang Y, Zhang M, Chen X, et al. Combinative treatment of beta-elemene and cetuximab is sensitive to KRAS mutant colorectal cancer cells by inducing ferroptosis and inhibiting epithelial-mesenchymal transformation. Theranostics 2020;10:5107–5119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Alannan M, Fatrouni H, Trezeguet V, Dittrich-Domergue F, Moreau P, Siegfried G, Liet B, et al. Targeting PCSK9 in Liver Cancer Cells Triggers Metabolic Exhaustion and Cell Death by Ferroptosis. Cells 2022;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yang C, Lu T, Liu M, Yuan X, Li D, Zhang J, Zhou L, et al. Tiliroside targets TBK1 to induce ferroptosis and sensitize hepatocellular carcinoma to sorafenib. Phytomedicine 2023;111:154668. [DOI] [PubMed] [Google Scholar]
- 83.Totoki Y, Tatsuno K, Covington KR, Ueda H, Creighton CJ, Kato M, Tsuji S, et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat Genet 2014;46:1267–1273. [DOI] [PubMed] [Google Scholar]
- 84.Makino Y, Hikita H, Fukumoto K, Sung JH, Sakano Y, Murai K, Sakane S, et al. Constitutive Activation of the Tumor Suppressor p53 in Hepatocytes Paradoxically Promotes Non-Cell Autonomous Liver Carcinogenesis. Cancer Res 2022;82:2860–2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Liu Y, Ouyang L, Mao C, Chen Y, Li T, Liu N, Wang Z, et al. PCDHB14 promotes ferroptosis and is a novel tumor suppressor in hepatocellular carcinoma. Oncogene 2022;41:3570–3583. [DOI] [PubMed] [Google Scholar]
- 86.Zhang X, Zheng Q, Yue X, Yuan Z, Ling J, Yuan Y, Liang Y, et al. ZNF498 promotes hepatocellular carcinogenesis by suppressing p53-mediated apoptosis and ferroptosis via the attenuation of p53 Ser46 phosphorylation. J Exp Clin Cancer Res 2022;41:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu J, Zhang C, Lin M, Zhu W, Liang Y, Hong X, Zhao Y, et al. Glutaminase 2 negatively regulates the PI3K/AKT signaling and shows tumor suppression activity in human hepatocellular carcinoma. Oncotarget 2014;5:2635–2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci U S A 2010;107:7455–7460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Jennis M, Kung CP, Basu S, Budina-Kolomets A, Leu JI, Khaku S, Scott JP, et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev 2016;30:918–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Gnanapradeepan K, Indeglia A, Stieg DC, Clarke N, Shao C, Dougherty JF, Murali N, et al. PLTP is a p53 target gene with roles in cancer growth suppression and ferroptosis. J Biol Chem 2022;298:102637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Green DR, Kroemer G. Cytoplasmic functions of the tumour suppressor p53. Nature 2009;458:1127–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Cui X, Yun X, Sun M, Li R, Lyu X, Lao Y, Qin X, et al. HMGCL-induced beta-hydroxybutyrate production attenuates hepatocellular carcinoma via DPP4-mediated ferroptosis susceptibility. Hepatol Int 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, Zhong M, et al. The Tumor Suppressor p53 Limits Ferroptosis by Blocking DPP4 Activity. Cell Rep 2017;20:1692–1704. [DOI] [PubMed] [Google Scholar]
- 94.Xu MZ, Yao TJ, Lee NP, Ng IO, Chan YT, Zender L, Lowe SW, et al. Yes-associated protein is an independent prognostic marker in hepatocellular carcinoma. Cancer 2009;115:4576–4585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Park H, Lee Y, Lee K, Lee H, Yoo JE, Ahn S, Park YN, et al. The Clinicopathological Significance of YAP/TAZ Expression in Hepatocellular Carcinoma with Relation to Hypoxia and Stemness. Pathol Oncol Res 2021;27:604600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Zhu G, Murshed A, Li H, Ma J, Zhen N, Ding M, Zhu J, et al. O-GlcNAcylation enhances sensitivity to RSL3-induced ferroptosis via the YAP/TFRC pathway in liver cancer. Cell Death Discov 2021;7:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chen Y, Zhu G, Liu Y, Wu Q, Zhang X, Bian Z, Zhang Y, et al. O-GlcNAcylated c-Jun antagonizes ferroptosis via inhibiting GSH synthesis in liver cancer. Cell Signal 2019;63:109384. [DOI] [PubMed] [Google Scholar]
- 98.Hill RA, Liu YY. N(6) -methyladenosine-RNA methylation promotes expression of solute carrier family 7 member 11, an uptake transporter of cystine for lipid reactive oxygen species scavenger glutathione synthesis, leading to hepatoblastoma ferroptosis resistance. Clin Transl Med 2022;12:e889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Fan Z, Yang G, Zhang W, Liu Q, Liu G, Liu P, Xu L, et al. Hypoxia blocks ferroptosis of hepatocellular carcinoma via suppression of METTL14 triggered YTHDF2-dependent silencing of SLC7A11. J Cell Mol Med 2021;25:10197–10212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wang Q, Guo Y, Wang W, Liu B, Yang G, Xu Z, Li J, et al. RNA binding protein DAZAP1 promotes HCC progression and regulates ferroptosis by interacting with SLC7A11 mRNA. Exp Cell Res 2021;399:112453. [DOI] [PubMed] [Google Scholar]
- 101.Xu Q, Zhou L, Yang G, Meng F, Wan Y, Wang L, Zhang L. CircIL4R facilitates the tumorigenesis and inhibits ferroptosis in hepatocellular carcinoma by regulating the miR-541-3p/GPX4 axis. Cell Biol Int 2020;44:2344–2356. [DOI] [PubMed] [Google Scholar]
- 102.Yuan J, Lv T, Yang J, Wu Z, Yan L, Yang J, Shi Y. HDLBP-stabilized lncFAL inhibits ferroptosis vulnerability by diminishing Trim69-dependent FSP1 degradation in hepatocellular carcinoma. Redox Biol 2022;58:102546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Zhang B, Bao W, Zhang S, Chen B, Zhou X, Zhao J, Shi Z, et al. LncRNA HEPFAL accelerates ferroptosis in hepatocellular carcinoma by regulating SLC7A11 ubiquitination. Cell Death Dis 2022;13:734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Qi W, Li Z, Xia L, Dai J, Zhang Q, Wu C, Xu S. LncRNA GABPB1-AS1 and GABPB1 regulate oxidative stress during erastin-induced ferroptosis in HepG2 hepatocellular carcinoma cells. Sci Rep 2019;9:16185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Guo S, Chen Y, Xue X, Yang Y, Wang Y, Qiu S, Cui J, et al. TRIB2 desensitizes ferroptosis via betaTrCP-mediated TFRC ubiquitiantion in liver cancer cells. Cell Death Discov 2021;7:196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Shan Y, Yang G, Lu Q, Hu X, Qi D, Zhou Y, Xiao Y, et al. Centrosomal protein 290 is a novel prognostic indicator that modulates liver cancer cell ferroptosis via the Nrf2 pathway. Aging (Albany NY) 2022;14:2367–2382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Shan Y, Yang G, Huang H, Zhou Y, Hu X, Lu Q, Guo P, et al. Ubiquitin-Like Modifier Activating Enzyme 1 as a Novel Diagnostic and Prognostic Indicator That Correlates With Ferroptosis and the Malignant Phenotypes of Liver Cancer Cells. Front Oncol 2020;10:592413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wang K, Zhang Z, Tsai HI, Liu Y, Gao J, Wang M, Song L, et al. Branched-chain amino acid aminotransferase 2 regulates ferroptotic cell death in cancer cells. Cell Death Differ 2021;28:1222–1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Jiang Z, Wang Z, Chen L, Zhang C, Liao F, Wang Y, Wang Y, et al. Artesunate induces ER-derived-ROS-mediated cell death by disrupting labile iron pool and iron redistribution in hepatocellular carcinoma cells. Am J Cancer Res 2021;11:691–711. [PMC free article] [PubMed] [Google Scholar]
- 110.Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, Xie Y, et al. AMPK-Mediated BECN1 Phosphorylation Promotes Ferroptosis by Directly Blocking System Xc(−) Activity. Curr Biol 2018;28:2388–2399 e2385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Du X, Qi Z, Xu J, Guo M, Zhang X, Yu Z, Cao X, et al. Loss of GABARAPL1 confers ferroptosis resistance to cancer stem-like cells in hepatocellular carcinoma. Mol Oncol 2022;16:3703–3719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Hu X, He Y, Han Z, Liu W, Liu D, Zhang X, Chen L, et al. PNO1 inhibits autophagy-mediated ferroptosis by GSH metabolic reprogramming in hepatocellular carcinoma. Cell Death Dis 2022;13:1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Byun JK, Lee S, Kang GW, Lee YR, Park SY, Song IS, Yun JW, et al. Macropinocytosis is an alternative pathway of cysteine acquisition and mitigates sorafenib-induced ferroptosis in hepatocellular carcinoma. J Exp Clin Cancer Res 2022;41:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Dai E, Han L, Liu J, Xie Y, Zeh HJ, Kang R, Bai L, 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]
- 115.Conche C, Finkelmeier F, Pesic M, Nicolas AM, Bottger TW, Kennel KB, Denk D, et al. Combining ferroptosis induction with MDSC blockade renders primary tumours and metastases in liver sensitive to immune checkpoint blockade. Gut 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Furutani T, Hino K, Okuda M, Gondo T, Nishina S, Kitase A, Korenaga M, et al. Hepatic iron overload induces hepatocellular carcinoma in transgenic mice expressing the hepatitis C virus polyprotein. Gastroenterology 2006;130:2087–2098. [DOI] [PubMed] [Google Scholar]
- 117.Li Y, Yu P, Fu W, Wang S, Zhao W, Ma Y, Wu Y, et al. Ginsenoside Rd Inhibited Ferroptosis to Alleviate CCl(4)-Induced Acute Liver Injury in Mice via cGAS/STING Pathway. Am J Chin Med 2023;51:91–105. [DOI] [PubMed] [Google Scholar]
- 118.Yamane D, Hayashi Y, Matsumoto M, Nakanishi H, Imagawa H, Kohara M, Lemon SM, 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 e794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Komissarov AA, Karaseva MA, Roschina MP, Shubin AV, Lunina NA, Kostrov SV, Demidyuk IV. Individual Expression of Hepatitis A Virus 3C Protease Induces Ferroptosis in Human Cells In Vitro. Int J Mol Sci 2021;22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Qi J, Kim JW, Zhou Z, Lim CW, Kim B. 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]
- 121.Lu D, Xia Q, Yang Z, Gao S, Sun S, Luo X, Li Z, et al. ENO3 promoted the progression of NASH by negatively regulating ferroptosis via elevation of GPX4 expression and lipid accumulation. Ann Transl Med 2021;9:661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Li X, Wang TX, Huang X, Li Y, Sun T, Zang S, Guan KL, et al. Targeting ferroptosis alleviates methionine-choline deficient (MCD)-diet induced NASH by suppressing liver lipotoxicity. Liver Int 2020;40:1378–1394. [DOI] [PubMed] [Google Scholar]
- 123.Shu YY, Gao WK, Chu HK, Yang L, Pan XL, Ye J. Attenuation by Time-Restricted Feeding of High-Fat and High-Fructose Diet-Induced NASH in Mice Is Related to Per2 and Ferroptosis. Oxid Med Cell Longev 2022;2022:8063897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Wang H, An P, Xie E, Wu Q, Fang X, Gao H, Zhang Z, et al. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology 2017;66:449–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Fu Y, Silverstein S, McCutcheon JN, Dyba M, Nath RG, Aggarwal M, Coia H, et al. An endogenous DNA adduct as a prognostic biomarker for hepatocarcinogenesis and its prevention by Theaphenon E in mice. Hepatology 2018;67:159–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, Wilhelm S, et al. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res 2006;66:11851–11858. [DOI] [PubMed] [Google Scholar]
- 127.Lachaier E, Louandre C, Godin C, Saidak Z, Baert M, Diouf M, Chauffert B, et al. Sorafenib induces ferroptosis in human cancer cell lines originating from different solid tumors. Anticancer Res 2014;34:6417–6422. [PubMed] [Google Scholar]
- 128.Zheng J, Sato M, Mishima E, Sato H, Proneth B, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis 2021;12:698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zhu J, Berisa M, Schworer S, Qin W, Cross JR, Thompson CB. Transsulfuration Activity Can Support Cell Growth upon Extracellular Cysteine Limitation. Cell Metab 2019;30:865–876 e865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Chen Y, Li L, Lan J, Cui Y, Rao X, Zhao J, Xing T, et al. CRISPR screens uncover protective effect of PSTK as a regulator of chemotherapy-induced ferroptosis in hepatocellular carcinoma. Mol Cancer 2022;21:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Chen Q, Zheng W, Guan J, Liu H, Dan Y, Zhu L, Song Y, et al. SOCS2-enhanced ubiquitination of SLC7A11 promotes ferroptosis and radiosensitization in hepatocellular carcinoma. Cell Death Differ 2023;30:137–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yuan Z, Liu T, Huo X, Wang H, Wang J, Xue L. Glutamine Transporter SLC1A5 Regulates Ionizing Radiation-Derived Oxidative Damage and Ferroptosis. Oxid Med Cell Longev 2022;2022:3403009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Tang B, Zhu J, Wang Y, Chen W, Fang S, Mao W, Xu Z, et al. Targeted xCT-mediated Ferroptosis and Protumoral Polarization of Macrophages Is Effective against HCC and Enhances the Efficacy of the Anti-PD-1/L1 Response. Adv Sci (Weinh) 2023;10:e2203973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.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]
- 135.Liu J, Zhu S, Zeng L, Li J, Klionsky DJ, Kroemer G, Jiang J, et al. DCN released from ferroptotic cells ignites AGER-dependent immune responses. Autophagy 2021:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Horvath Z, Kovalszky I, Fullar A, Kiss K, Schaff Z, Iozzo RV, Baghy K. Decorin deficiency promotes hepatic carcinogenesis. Matrix Biol 2014;35:194–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Reszegi A, Horvath Z, Feher H, Wichmann B, Tatrai P, Kovalszky I, Baghy K. Protective Role of Decorin in Primary Hepatocellular Carcinoma. Front Oncol 2020;10:645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Finetti F, Travelli C, Ercoli J, Colombo G, Buoso E, Trabalzini L. Prostaglandin E2 and Cancer: Insight into Tumor Progression and Immunity. Biology (Basel) 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Hirayama T, Nagasawa H. Chemical tools for detecting Fe ions. J Clin Biochem Nutr 2017;60:39–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Feng J, Lu PZ, Zhu GZ, Hooi SC, Wu Y, Huang XW, Dai HQ, et al. ACSL4 is a predictive biomarker of sorafenib sensitivity in hepatocellular carcinoma. Acta Pharmacol Sin 2021;42:160–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Zhang B, Zhao J, Liu B, Shang Y, Chen F, Zhang S, He J, et al. Development and Validation of a Novel Ferroptosis-Related Gene Signature for Prognosis and Immunotherapy in Hepatocellular Carcinoma. Front Mol Biosci 2022;9:940575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Luo L, Yao X, Xiang J, Huang F, Luo H. Identification of ferroptosis-related genes for overall survival prediction in hepatocellular carcinoma. Sci Rep 2022;12:10007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Tang B, Zhu J, Li J, Fan K, Gao Y, Cheng S, Kong C, et al. The ferroptosis and iron-metabolism signature robustly predicts clinical diagnosis, prognosis and immune microenvironment for hepatocellular carcinoma. Cell Commun Signal 2020;18:174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Huang CY, Chen LJ, Chen G, Chao TI, Wang CY. SHP-1/STAT3-Signaling-Axis-Regulated Coupling between BECN1 and SLC7A11 Contributes to Sorafenib-Induced Ferroptosis in Hepatocellular Carcinoma. Int J Mol Sci 2022;23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Liang C, Li W, Ge H, Zhang K, Li G, Wu J. Role of Beclin1 expression in patients with hepatocellular carcinoma: a meta-analysis. Onco Targets Ther 2018;11:2387–2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Chen J, Argemi J, Odena G, Xu MJ, Cai Y, Massey V, Parrish A, et al. Hepatic lipocalin 2 promotes liver fibrosis and portal hypertension. Sci Rep 2020;10:15558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Yang H, Sun W, Bi T, Wang Q, Wang W, Xu Y, Liu Z, et al. The PTBP1-NCOA4 axis promotes ferroptosis in liver cancer cells. Oncol Rep 2023;49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Meng H, Shen M, Li J, Zhang R, Li X, Zhao L, Huang G, et al. Novel SREBP1 inhibitor cinobufotalin suppresses proliferation of hepatocellular carcinoma by targeting lipogenesis. Eur J Pharmacol 2021;906:174280. [DOI] [PubMed] [Google Scholar]
- 149.Balgoma D, Kullenberg F, Calitz C, Kopsida M, Heindryckx F, Lennernas H, Hedeland M. Anthracyclins Increase PUFAs: Potential Implications in ER Stress and Cell Death. Cells 2021;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Ren X, Li Y, Zhou Y, Hu W, Yang C, Jing Q, Zhou C, et al. Overcoming the compensatory elevation of NRF2 renders hepatocellular carcinoma cells more vulnerable to disulfiram/copper-induced ferroptosis. Redox Biol 2021;46:102122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Jin M, Shi C, Li T, Wu Y, Hu C, Huang G. Solasonine promotes ferroptosis of hepatoma carcinoma cells via glutathione peroxidase 4-induced destruction of the glutathione redox system. Biomed Pharmacother 2020;129:110282. [DOI] [PubMed] [Google Scholar]
- 152.Yang X, Xiao J, Jiang L, Ran L, Fan Y, Zhang M, Xu Y, et al. A Multifunctional Vanadium-Iron-Oxide Nanoparticle Eradicates Hepatocellular Carcinoma via Targeting Tumor and Endothelial Cells. ACS Appl Mater Interfaces 2022;14:28514–28526. [DOI] [PubMed] [Google Scholar]
- 153.Li ZJ, Dai HQ, Huang XW, Feng J, Deng JH, Wang ZX, Yang XM, et al. Artesunate synergizes with sorafenib to induce ferroptosis in hepatocellular carcinoma. Acta Pharmacol Sin 2021;42:301–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Wu Y, Ran L, Yang Y, Gao X, Peng M, Liu S, Sun L, 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]