- Review
- Open access
- Published:
From mechanisms to medicine: Ferroptosis as a Therapeutic target in liver disorders
Cell Communication and Signaling volume 23, Article number: 125 (2025)
Abstract
In recent 10 years, ferroptosis has become a hot research direction in the scientific research community as a new way of cell death. Iron toxicity accumulation and lipotoxicity are unique features. Several studies have found that ferroptosis is involved in the regulation of the hepatic microenvironment and various hepatic metabolisms, thereby mediating the progression of related liver diseases. For example, NRF2 and FSP1, as important regulatory proteins of ferroptosis, are involved in the development of liver tumors and liver failure. In this manuscript, we present the mechanisms involved in ferroptosis, the concern of ferroptosis with the liver microenvironment and the progression of ferroptosis in various liver diseases. In addition, we summarize recent clinical advances in targeted ferroptosis therapy for related diseases. We expect that this manuscript can provide a new perspective for clinical treatment of related diseases.
Introduction
Over the past decade, the scientific community has witnessed a surge of interest in ferroptosis, a novel form of regulated cell death [1]. The term "ferroptosis" is derived from its characteristic features, primarily the accumulation of iron and the peroxidation of polyunsaturated fatty acids within the cell membrane, leading to cell death. The unique nature of ferroptosis, with its dependence on iron metabolism and vulnerability to lipid peroxidation, positions it as a pivotal player in the pathogenesis of liver diseases.
Liver diseases constitute a significant global public health challenge, with mechanisms of cellular death playing a pivotal role in the initiation and progression of a variety of hepatic disorders [2]. Ferroptosis, a distinct form of regulated cell death, has garnered attention for its involvement in a spectrum of liver diseases. Particularly in NAFLD, iron deposition and oxidative stress, which arise from metabolic disorders, may induce ferroptosis, thereby promoting the progression of liver disease. In ALD, chronic ethanol-induced ROS overproduction and oxidative stress may trigger ferroptosis in hepatocytes, promoting liver disease progression. In HCC, ferroptosis is implicated not only in tumorigenesis but also potentially influences the response to therapeutic agents such as sorafenib.
In recent years, the study of ferroptosis has advanced rapidly in hepatic medicine. Targeting ferroptosis in hepatology presents a promising therapeutic approach for liver diseases. Traditional drugs, including Deferoxamine, Deferiprone, Vitamin E and Selenium, have shown potential in reducing liver damage (Table 1). This article further reviews the latest progress in iron-targeting drug development, encompassing natural compounds (e.g., Diosgenin, Nuciferine, Xanthohumol), artificial extracts (e.g., sulfated polysaccharides), small molecule inhibitors (e.g., GLX351322), modifications of traditional drugs (e.g., Donafenib), and novel nanomaterials (e.g., GOA/miR-targeted nanocomposites, mitochondrial-targeted antioxidants like MitoTEMPO and MnO2Nfs), while also examining the potential limitations of these strategies.
This review aims to provide a comprehensive overview of the current understanding of ferroptosis, highlighting its significance in the context of liver health and disease. We will discuss the key regulatory proteins involved in ferroptosis, such as NRF2, FSP1, and GPX4, and their implications in the development of liver tumors and failure. Furthermore, we will explore the multifaceted interactions between ferroptosis and the hepatic microenvironment, including iron homeostasis, glucose and lipid metabolism, amino acid metabolism, gut-liver axis and the mitochondria. Delving into the intricate mechanisms of ferroptosis, its association with the liver microenvironment, and its role in the progression of various liver diseases. These discussions will be grounded in the latest research findings, providing a platform for understanding the complex interplay between ferroptosis and liver diseases.
Three pivotal features of ferroptosis in research foci
So far, it has been demonstrated that subcellular cells undergoing ferroptosis may evolve at least three different pivotal features. These primarily involve the iron toxicity accumulation from disrupted iron-homeostasis, phospholipid peroxidation caused by excessive intracellular lipid reactive oxygen species [32], and the occurrence of iron-induced apoptosis in cells characterized by abnormal mitochondrial morphology (Fig. 1). By investigating the profound alterations in iron metabolism, we can acquire a meticulous comprehension of the onset and progression of ferroptosis-related disorders.
Iron toxicity accumulation
Over ten years ago, the term " ferroptosis " was first introduced as an innovative form of cell death and remains an attractive field of investigation [1]. Ferroptosis, as the name indicates, is inexorably tied to poisonous iron accumulation. Iron homeostasis normally firmly regulates the systemic circulatory system. One aspect, dietary iron absorption ensures that the body has enough iron, while also preventing harmful iron sediment. Iron accumulation can directly cause varying degrees of toxicity in different organs, such as the kidney and pancreas [33, 34].
Phospholipid peroxidation
In healthy human tissues, the generation of ROS is a natural physiological occurrence. In pathological conditions, heightened metabolic activity and increased oxidative stress often leads to the catalysis of the Fenton reaction, resulting in the production of hydroxyl radicals (•OH). This can surpass the antioxidant capacity of cells, causing damage to biomolecules [35].In the absence of universal standards for diagnosing iron toxicity, oxidized phospholipids are often used as indicators [36].Research suggests that polyunsaturated fatty acids (PUFAs) are particularly prone to peroxidation and may contribute to ferroptosis [37].
Dysfunctional mitochondria
In ferroptotic cells, morphological assessments reveal notable changes such as cell rounding, mitochondrial cristae shrinkage, and cytoplasmic membrane disintegration, with the nucleus often remaining unaffected [38, 39]. Additionally, mitochondria exhibit structural anomalies like reduced size and increased membrane density, which are indicative of cellular injury in ferroptosis [40].However, the precise mechanisms by which aberrant mitochondria mediate ferroptosis remain an area of inquiry that warrants further exploration. In their physiological role, mitochondria primarily serve as the powerhouses of the cell, orchestrating adenosine triphosphate (ATP) production [40]. Mitochondrial dysfunction, potentially through impaired oxidative phosphorylation, may precipitate ferroptosis by escalating ROS production beyond the cell's antioxidant threshold [41]. Additionally, mitochondrial-associated molecules, structural proteins, and metabolic disruptions may also mediate the onset of ferroptosis. Aberrant mitochondrial GPx4, DHODH, and ACSL family expressions correlate with cellular vulnerability to iron-overload, representing a research focus [42]. CISD1, a CDGSH iron-sulfur domain protein, suppresses iron-catalyzed cell death by regulating mitochondrial iron deposition [43]. Ferroptotic cell death is closely linked to mitochondrial metabolic dysregulation, which includes impaired mitochondrial protein degradation, metabolic reconfiguration, and mitochondrial stress signaling [44].
Regulatory molecules involved in ferroptosis
GPX4
GPX4 is a peroxidase that relies on the presence of glutathione (GSH) and contains selenium [45]. It primarily occurs in the cytoplasm and mitochondria of mammals. It has the capacity to eliminate toxins such as GSH and lipid hydroperoxide by converting them into harmless lipid ethanol [45, 46]. Additionally, it acts as a key inhibitor of ferroptosis, displaying distinct properties. Multiple investigations have demonstrated that it serves as the primary regulator of ferroptosis. GPX4 depletion causes the disruption of the lipid peroxidation cascade, leading to the death of non-apoptotic cells [45,46,47,48]. Once GPX4 is increased, it can prevent iron-induced cell death and safeguard different cells and tissues from harm [47]. More precisely, the transcription factor or the protein itself regulates GPX4 expression. Compounds like sulfasalazine and sorafenib, known as ferroptosis inducers, have the ability to impede its function, conversely, the activity of this substance can be enhanced by intracellular selenium or glutathione supplementation [47].
GPX4 has been demonstrated to play an essential part in the development and prognosis of numerous disorders. It is worth mentioning that GPX4 deficiency in neurological diseases causes cognitive impairment and neurodegeneration, which can be alleviated by iron-death inhibitions [48]. GPX4 is essential for the liver, and its low level primarily ensures the integrity of PL-PUFA in a healthy liver, but this stable condition may be altered in several metabolic problem liver disorders [49]. Furthermore, GPX4 is widely recognized as a tumor suppressor, and the GSH/GPX4 axis plays a crucial role in restraining the metabolism of cancer cells when catalytically active iron is present (Fig. 2) [50].
Regulatory modulators and mechanism involved in ferroptosis. In particular, the GSH metabolism, iron sequestration and metabolism, GPX4 inactivation, PUFA synthesis, and the loss of membrane ionic homeostasis are all orchestrally engaged in the modulation of ferroptosis, with pathways such as the CoQ10-FSP1 axis, DHODH, and NRF2 contributing to this sophisticated regulatory mechanism. Abbreviations: Cys, cysteine; Glu, glutamate; Gln, glutamine; GLS2, glutaminase 2; HO-1, heme oxygenase-1; KGM, konjac glucomannan; GOT-1, glutamic oxaloacetic transaminase 1; MTORC 1, mechanistic target of rapamycin complex 1; Gly, glycine; miR-9, microRNA-9; DHFR, dihydrofolate reductase; GCH1, GTP cyclohydrolase 1; MTX, methotrexate; BH4, tetrahydrobiopterin; GPX4, glutathione peroxidase 4; FIN5B, FYVE and coiled-coil domain containing 2; FSP1, farnesyl pyrophosphate synthase 1; CoQ10, coenzyme Q10; ESCRT-III, endosomal sorting complexes required for transport III; DHODH, dihydroorotate dehydrogenase; ARE, antioxidant response element; Keap1, kelch-like ech-associated protein 1; ACSL4, acyl-coA synthetase long-chain family member 4; ALOXs, arachidonate lipoxygenases; BHA, butylated hydroxyanisole; DMT1, divalent metal transporter 1; PUFA-PL, phospholipid containing polyunsaturated fatty acid chain; STEAP3, six transmembrane epithelial antigen of the prostate 3; TCA, tricarboxylic acid cycle; SLC1A5, solute carrier family 1 (neutral amino acid transporter), member 5; SLC3A2, solute carrier family 3 (activatory amino acid transporter), member 2
System Xc −
System Xc− is an antiporter composed of two subunits: SLC7A11 and SLC3A2 [51].Cells utilize SLC7A11 and SLC3A2 to transport amino acids. SLC7A11 transports cystine, while SLC3A2 is a heavy chain component of amino acid transporters (HATs) that ferry important amino acids alongside SLC7 family members [51]. Cysteine availability is crucial for GSH synthesis, and System Xc− mostly absorbs extracellular cystine via exchanging glutamate in cells to preserve redox equilibrium [52, 53]. Blocking the system will decrease cysteine levels, disrupt glutathione (GSH) synthesis, impair the activity of the GPx4 enzyme, and ultimately result in ferroptosis due to iron accumulation [53]. To summarize, blocking antiporters can increase cell susceptibility to ferroptosis by decreasing the production of GSH [52].
SLC7A11 gene translation is finely transcriptionally directed, with oxygen exhaustion and amino acid depletion conditions stimulating factor, hence dependently inducing its production. In the SLC7A11 systemic knockout mouse model, depletion of GSH levels was observed, but did not cause spontaneous hepatic iron deficiency, but rather increased sensitivity to ferroptosis [52]. Further, SLC7A11 knock-out cell showed significant inhibition of tumor proliferation [52]. Remarkably, deletion of SLC7A11 in normal cells does not directly trigger iron-death, but may enhance cellular sensitivity to ferroptosis [54]. The iron-death inhibitory molecule p53 downregulates System Xc− expression, resulting in cystine deficiency and sensitivity to ferroptosis, favoring anticancer [55]. These findings emphasize SLC7A11 in controlling cellular reactions to iron-toxicity and could offer innovative approaches for treatment (Fig. 2).
NRF2
Nuclear factor erythroid 2-related factor 2 [56] is a fundamental local transcription element that promotes resistance to ferroptosis and is crucial for preserving the balance of antioxidants and metabolic stability in cells [56]. NRF2 possesses a conserved basic leucine zipper (bZIP) domain and encounters the capability to create heterodimers with other proteins, which then attach to anti-oxidative stress elements (ARE) [56]. Typically, NRF2 expression remains consistently low because it binds to a cytoplasmic protein called Keap1 and is then broken down through ubiquitination [57]. During stress-induced oxidation signaling, NRF2 dissociates from Keap1, avoids degradation, moves into the nucleus to begin the transcription of downstream genes. NRF2 is an essential upstream regulator that controls the expression of antioxidant genes, it controls the activities of GPX4 and the System Xc−, which are vital for glucose metabolism [58]. Also, the p62-Keap1-NRF2 signaling cascade is linked to the upregulation of genes entailed in ferroptosis and ROS metabolic processes, demonstrating the diverse impact of NRF2 [59].
NRF2 has a dual function, whereby reducing its expression can enhance the susceptibility to cancer cells to iron-death, while raising NRF2 levels can impede the ferroptosis process [15, 60]. In hepatocellular carcinoma (HCC), the activation of NRF2 leads to increased resistance to sorafenib by up-regulating Metallothionein-1G(MT-1G), which hinders lipid metabolism [61]. In vitro models, lowering NRF2 expression by molecular biological methods enhances the effectiveness of anticancer treatments [61]. Activated the p62-Keap1-NRF2 axis shields cancerous cells from iron-induced cell death by inhibiting the degradation of NRF2 [59]. NRF2 may mitigate iron-death in Acute Lung Injury(ALI) by upregulating the production of antioxidant enzymes and associated genes, protecting alveolar epithelial cells, and minimizing lung damage [62]. These findings show that NRF2 and its downstream signaling pathways could be an innovative therapy for triggering ferroptosis (Fig. 2).
FSP1
The recently discovered endogenous robust Ferroptosis-suppressor protein 1 (FSP1) may function in tandem with coenzyme Q10 to preserve mitochondrial function and shield them from oxidative damage [63, 64]. Its precursor is apoptosis-inducing factor mitochondria-2 (AIFM2), more precisely re-termed FSP1 due to its vital function in the distinct FSP1–Q10–NADPH mechanism, independent of the classical GSH-GPx4 anti-ferroptosis pathway [63]. Specifically, FSP1 can also function in a new electron transport mechanism that reduces coenzyme Q10 to prevent iron-induced cytotoxic damage when GSH is depleted.
Furthermore, FSP1 is postulated to operate in a parallel mechanism to GPX4, and its expression positively correlates with cellular resistance to GPX4, hence maintaining tumor cell proliferation [63]. FSP1 also impacts iron death-associated metabolic enzymes, sustaining the biological function of S-adenosylhomocysteine hydrolase (SAHH), whose continued presence is necessary to avert ferroptosis [65]. Research has revealed that FSP1 can effectively limit the iron-death by engaging in intricate interactions with regulatory pathways [65, 66]. Besides, ESCRT-III (endosomal sorting complex required for transport III)-dependent membrane repair mechanisms can hinder ferroptosis. The activation of these mechanisms processes generates a suitable environment for survived tumor cells and indirectly supports the role of FSP1 (Fig. 2) [66].
DHODH
The ferroptosis defense mechanism is typically classified into three types depending on intracellular localization: cytosolic and mitochondrial GPx4, plasma membrane-localized FSP1, and mitochondrial dihydroorotate dehydrogenase (DHODH) [67]. These three systems are independent and interact to provide antioxidant effects, the first two system parts were previously described in detail, and we will now focus on the mitochondrial inner membrane enzyme DHODH [68]. It catalyzes de novo pyrimidine ribonucleotide synthesis, and cancer cells proliferate and require more nucleotides, hence it has been investigated researched as cancer treatment [69].Metabolomic profiling studies have demonstrated the involvement of DHODH and mito-GPX4 in scavenging reactive oxygen species (ROS) and their unique roles in inducing apoptosis, irrespective of their usual function in nucleotide synthesis [18, 70].
Lowering the expression of DHODH promotes intracellular GSH levels, hence enhancing the antioxidant effectiveness of GPX4 and preventing iron-induced toxicity [71]. Utilizing its catalytic CoQ10H2 (ubiquinol) in the mitochondrial electron transport chain, performs antioxidant activities and reduces peroxidation of mitochondrial membrane phospholipids, protecting against iron-death [71]. Although there were initial efforts to manage tumor growth by employing DHODH inhibitors, the effectiveness of drugs in early clinical trials did not reach expectations [72, 73]. This underscores the necessity for thorough investigation into the impacts of DHODH inhibitors on the ferroptosis process in tumors, which in turn affects the effectiveness evaluation of drugs (Fig. 2).
GOT-1
Incorporating the miRNA-associated epigenetic modifications discussed earlier, we focus on an enzyme referred to as glutamic-oxaloacetic transaminase-1 (GOT-1). MiR-9 specifically targets GOT-1 by interacting in the 3'-untranslated region (3'-UTR), modifying GOT1 expression, whereas inhibiting miR-9 increases its expression [74]. More precisely, an excessive amount of miR-9 causes a decrease in both the mRNA and protein levels of GOT-1, consequently diminishing the intracellular Fe2 + accumulation induced by glutamate and sulfasalazine [74]. Specifically, GOT-1 is involved in the metabolic process of glutamine, where glutamine is transformed into glutamate by the enzyme glutaminase (GLS), and then GOT-1 changes glutamate into alpha-ketoglutarate (α-KG) [54, 75]. Variations in GOT-1 can affect the catabolism of glutamine, thereby influencing the balance of glutamate and cysteine, playing a pivotal role in ferroptosis (Fig. 2).
ACSL4
The enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4), pivotal in chemical process of lipids, facilitates iron overload and mitigates hepatic damage as well as fatty acid peroxidation [76]. It is the main function that it attaches coenzyme A to long-chain polyunsaturated fatty acids (PUFAs) and converts free fatty acids into fatty coenzyme A esters, a process that is mainly carried out on mitochondria and the outer membrane of the endoplasmic reticulum [77]. When embedded inside membranes, PUFAs initiate oxidation, subsequently triggering extensive lipid peroxidation and the onset of iron-death [78].
Molecular screenings have been conducted on several cell lines to assess the ACSL family members, which include ACSL1, ACSL3-6. Among them, ACSL4 has been identified as a significant activator in the ferroptosis-induced death [77, 79]. Reduced production of peroxided lipids and induction of iron overload were observed in an ACSL4 conditional deletion mouse, conversely, the phenotype was reversed upon overexpression of ACSL4. [77]. Likewise, the reduction of ACSL4 made cells resistant to iron overload, whereas its increased expression intensified susceptibility to RSL3-induced iron-death [80]. Particularly, ACSL4 levels are higher in cells susceptible to ferroptosis than in those exhibiting resistance. Additionally, individuals showing positive responses to sorafenib exhibit increased ACSL4 levels compared to those with inadequate responses to the drug [77, 81]. The findings indicate that targeting ACSL4 could be a new approach to trigger iron-death and could potentially be used as a predictive factor for overall long-term and illness-free survival in patients with advanced liver cancer following sorafenib treatment (Fig. 2) [81, 82].
BH4-GCH1
Tetrahydrobiopterin (BH4) is a lipophilic reductant that plays a vital part as a coenzyme for numerous enzymes that regulate key metabolic pathways [83]. BH4 is vital for the biological function of iron-death and serves as both a reactive antioxidant, protecting cells from lipid-induced peroxidation, but also for the biosynthesis of coenzyme Q10 (a powerful antioxidant) [83, 84]. Guanosine triphosphate cyclohydrolase-1 (GCH1), has been demonstrated to hinder ferric ptosis in laboratory conditions by its overexpression. This is achieved by avoiding the crucial hydrolysis of phosphatidylcholine and PUFA chains [85]. CRISPR-based genome assays detected GCH1, an important enzyme in BH4 production, being a strong inhibitor of ferroptosis, and this inhibition occurs regardless of the presence of GPX4 [83, 84]. Furthermore, the GCH1-BH4 axis could serve as a promising focus in cancer treatment, as its expression is strongly linked to cancer cell resistance against ferroptosis (Fig. 2).
Pharmacological and genetic modulators of ferroptosis
In the realms of pharmacology and genetics, drugs and compounds are typically classified as inducers and inhibitors based on their impact on biological processes. In the field of ferroptosis-related pharmacology, inducers are primarily categorized into three classes. Class I ferroptosis promoters are compounds that target the system Xc− and activate ferroptosis by inhibiting this system, leading to the depletion of intracellular GSH and the inactivation of GPX4, such compounds include Erastin [86], Sorafenib [87], and Glutamate [5]. Class II ferroptosis promoters are direct inhibitors of GPX4 that covalently bind to GPX4 to suppress its function, resulting in the accumulation of toxic lipid peroxides and the induction of ferroptosis. Examples include RAS-selective lethal protein 3(RSL3) [36]and ML-162 [12]. Other types may affect the abundance of GPX4 or other fundamental regulators of ferroptosis through alternative pathways. Classical pharmacological inhibitors include Fer-1 [19, 86], vitamin E [22], and Trolox [22],which possess lipophilic antioxidant properties,see Table 1.
At the genetic level, ferroptosis activators and inhibitors can be categorized based on their mechanisms of action, which involve a variety of iron-regulatory molecules. For a detailed summary, see Table 2 and Fig. 2.
Ferroptosis and other classic forms of regulated cell death
Cellular death is not merely a cessation of cell life, but rather an intricately orchestrated biological process designed to remove cells that are damaged, senescent, or no longer required. Cell death mechanisms are divided into two main types: non-lytic and immunologically silent, such as apoptosis, where cells emit the phagocytosis signals for phagocytic clearance without rupturing; and the lytic and pro-inflammatory type, including ferroptosis, necroptosis, and pyroptosis, which release cellular contents upon rupture, inducing inflammation [111].
Ferroptosis and apoptosis
Apoptosis is vital for development, immune tolerance, and tissue homeostasis, and is a key area of research in cell death [112]. Apoptosis is primarily initiated through the activation of caspase family cysteine proteases [113]. Extrinsic apoptosis is initiated through the activation of caspase-8, while intrinsic apoptosis is mediated by the mitochondrial release of cytochrome c and SMAC/DIABLO, leading to the activation of caspase-9, which in turn activates downstream effector caspases such as caspase-3 and caspase-7 [112, 114]. These effector caspases cleave a multitude of protein substrates, resulting in the orchestrated dismantling of the cell. Ferroptosis is an iron-dependent form of non-apoptotic cell death that occurs without caspase activation and is not reversible by caspase inhibitors [115]. As previously mentioned, NRF2, a key antioxidant transcription factor in ferroptosis, has been shown to play an opposite role in apoptosis [116]. BCL-2 family proteins are primarily associated with apoptosis, but the inactivation of certain members like BCL-2, BCL-xL, and MCL-1, which induce mitochondrial damage, can indirectly affect ferroptosis by altering mitochondrial function [114]. In technological applications, ferroptosis provides an alternative therapy for apoptosis-resistant tumor cells. Two-dimensional Ca2Mn8O16 nanosheets mimic enzyme activities, generate ROS, and induce Ca2 + overload, amplifying the synergistic effects of ferroptosis and apoptosis [117].
Ferroptosis and necroptosis
Scholars have proposed that apoptosis, necroptosis, and pyroptosis be collectively referred to as 'programmed cell death' [112]. Necroptosis was initially characterized by the suppression of non-caspase-mediated necrosis following treatment with necrostatin-1 [118]. In apoptosis-impaired conditions, the activation of specific receptors by their ligands can induce necroptosis, with RIPK1 being a pivotal mediator [119]. Necroptosis and ferroptosis are both forms of lytic cell death with membrane rupture. Morphologically, necroptosis typically presents as cellular swelling, whereas ferroptosis is characterized by reduced mitochondrial volume and decreased cristae [120]. Additionally, they differ in their mechanisms of executing cell death: necroptosis typically serves as an alternative pathway when apoptosis is inhibited, mediated by the activation of the pseudokinase MLKL, hereas ferroptosis is triggered by the accumulation of iron-dependent lipid peroxides [121].
Ferroptosis and autophagy
In the process of autophagy, lysosomal fusion merge with autophagosomes to form autolysosomes, which are crucial for sustaining cellular renewal and metabolic processes [122]. Autophagy is a normal physiological process, and the formation of autophagosomes occurs as a result of disruptions in intracellular metabolic activity, and its suppression can result in necroptosis [123]. Extensive research highlights the involvement of autophagy in ferroptosis [124]. Certain necroptosis inhibitors can mitigate ferroptosis by modulating the autophagy pathway. For instance, KW-2449 inhibits the activity of ULK1 kinase, thereby blocking the autophagy process and preventing ferroptosis. Similarly, Necrostatin-1 has been found to inhibit ferroptosis by modulating the autophagy pathway [125]. Moreover, ferroptosis is viewed as an autophagy-dependent cell death mechanism, with autophagy modulating iron metabolism through increased ferritin degradation [126]. Autophagy elevates free iron concentrations in subarachnoid hemorrhage animal models, resulting in iron toxicity accumulation [127].
Ferroptosis and pyroptosis
Cellular pyroptosis, mediated by caspases, is an inflammatory type of programmed cell death marked by membrane disruption and the release of pro-inflammatory cytokines, which amplifies local inflammation [128]. Dysregulation of pyroptosis can lead to diseases such as sepsis, autoimmune diseases, and neuroinflammation. It exhibits parallels with ferroptosis, characterized by membrane disruption, phospholipid peroxidation, and iron toxicity accumulation [129]. Plasma membrane rupture involves the permeation of pore-forming proteins including gasdermin D, and natriuretic peptide 1 [130]. Iron within cells catalyzes ROS production via oxidase enzymes and lipoxygenase, and excess ROS triggers oxidative stress, activating caspases that cleave ferritin, resulting in its degradation [131].
Ferroptosis and cellular senescence
Typically, moderate and sustained levels of ROS induce cellular senescence by causing DNA damage, oxidation of proteins and lipids, and subsequently activate cell cycle arrest-related signaling pathways, such as p53-p21 [132]. Excessive ROS target polyunsaturated fatty acids in the cell membrane, culminating in membrane rupture and ferroptosis [63]. The relationship between ferroptosis and cellular senescence is intricate, not simply additive. In vitro analyses of senescent cells have shown compromised ferritinophagy and enhanced resilience against ferroptosis [133]. Certain ferroptosis inducers can selectively eliminate senescent cells, offering potential for anti-aging therapies. Under Erastin treatment, the proportion of SA-β-gal positive cells, a classic marker of senescence, significantly decreased with the addition of the ferroptosis inhibitor Fer-1 [134]. Treatment with FIN56 significantly reduced the survival of primary and paracrine senescent cells, while TRX-CBI showed cytotoxicity against senescent cells at low concentrations, with minimal effects on non-senescent cells [135]. Senescent cells accumulate in tissues, disrupting iron metabolism and leading to iron accumulation, which increases the risk of ferroptosis, creating a vicious cycle that is regulated by ROS and autophagy. Autophagy delays senescence, but its dysfunction increases ROS accumulation, accelerating senescence [125]. DHA clears senescent cells via autophagy-dependent ferroptosis, modulating the AMPK/mTOR pathway to enhance antioxidant capacity and reduce ROS [136]. Chronic inflammation promotes the production of ROS, exacerbating oxidative damage, which further extends cellular senescence and ferroptosis, leading to age-related pathologies. In Alzheimer's disease, the iron toxicity accumulation and phospholipid peroxidation are linked to neuronal death [137], while in cardiovascular diseases, ferroptosis can result in endothelial dysfunction and atherosclerosis [138].
Ferroptosis in the liver microenvironment
Glucose metabolism: a dynamic regulator of iron homeostasis
Iron homeostasis is maintained by balancing iron supply with iron loss, erythropoiesis being a major part of iron demand, while iron supply arises primarily from intestinal absorption and iron cycling in macrophages [139, 140]. Hepcidin, a 25-residue peptide signaling molecule produced and released mainly by hepatocytes, disseminates through the bloodstream and is eliminated by renal excretion [141, 142]. Mutations in the gene regulating this molecule can lead to disorders characterized by iron-overload [143]. Research has demonstrated that the liver has an iron-sensing function; specifically, the liver can integrate signals to control the production of hepatic protein hormones when the iron level of plasma, tissue, or cells changes abruptly [144]. Hepatocytes obtain transferrin-bound iron (TBI) through TFR1, and the liver mainly maintains balance through these mechanisms [145]. The hepatic iron-sensing apparatus is a multifaceted regulatory hub that synergizes diverse mechanisms, including the IRP/IRE system, HIF-mediated responses, and the BMP-SMAD signaling pathway, to dynamically adjust hepcidin expression in accordance with cellular iron demands and systemic iron balance [146,147,148].
Although there are no set standards for identifying ferroptosis, lipid peroxidation and a decrease in nicotinamide adenine dinucleotide phosphate (NADPH) in its reduced form are the most frequently reported indicators [36, 46, 149]. Research indicates that NADPH functions in concert with FSP1 to convert CoQ10 into CoQ10-H2, a strong antioxidant that stops peroxidation of fatty acids from spreading across cell membranes(Fig. 3) [63]. The processes by which the liver maintains glucose homeostasis are bidirectionally regulated by iron homeostasis. In type 2 diabetes and NAFLD, increased hepatic gluconeogenesis contributes to hyperglycemia, while mitochondrial electron transport chain (ETC) dysfunction leads to ROS accumulation and disrupted iron homeostasis [150]. Mitochondrial complex defects (e.g., Cox10 deficiency) inhibit pyruvate-to-acetyl-CoA conversion, upregulate PDK4, and thereby block glucose metabolism and exacerbate lipid accumulation, forming a vicious cycle [151]. Current research predominantly focuses on the unidirectional impact of hyperglycemia on iron homeostasis, while the mechanisms by which iron overload feedback-regulates glucose metabolism (e.g., insulin resistance) remain unclear. Additionally, whether the mechanism by which metformin reduces hepatic gluconeogenesis through mitochondrial ETC inhibition is related to iron metabolism requires further validation.
The liver microenvironment orchestrates ferroptosis through its pivotal signaling pathways. The liver serves as a pivotal organ in regulating the metabolism of various nutrients, encompassing lipids, carbohydrates, and the synthesis of iron. After glycolysis, dietary components are converted into fatty acids under the regulation of lipid metabolic factors ACSL4 and LPCAT3. LOX catalyzes lipid peroxidation of ROS and PUFAs within cells, producing lipid ROS. When these overwhelm the liver's antioxidant defenses, ferroptosis in hepatocytes is triggered. Postprandially, glucose rapidly enters the liver, generating energy through glycolysis and producing NADPH via the pentose phosphate pathway. The liver is also the site of GSH synthesis, where SLC7A11-mediated cysteine uptake is crucial for GSH production, which can inhibit ferroptosis. mTORC2, acting as an amino acid sensor, suppresses ferroptosis by enhancing intracellular NADPH levels. Specifically, Hepatocytes synthesize and secrete hepcidin, which facilitates iron uptake through the endocytosis of Tf-Fe3+-TrR complex. Erythropoiesis is the major consumer of iron, supplied primarily by intestinal absorption and macrophage recycling. Gut microbiota activates the Nrf2/HO-1 pathway, promoting phospholipid peroxidation in the liver. Heterogeneous iron deposition is linked to ferroptosis and associated with various liver diseases. Abbreviations: GSH, glutathione; GSSH, glutathione persulfide; ACSL4, acyl-coenzyme synthetase long-chain family member 4; LPCAT3, lysophosphatidylcholine acyltransferase 3; SLC7A11, solute carrier family 7, member 11; MTORC2, mammalian target of rapamycin complex 2; Tf-Fe3+-TrR complex, Transferrin-Ferric Iron-Transferrin Receptor complex; TBI, Transferrin-bound iron
Aberrant lipid metabolism: a catalyst for ferroptosis
The interplay between hepatic lipid metabolism and ferroptosis is particularly pronounced [64]. Fatty acid β-oxidation primarily depends on the mitochondrial electron transport chain (ETC) to generate NADH and FADH₂. Dysfunction of the ETC, such as deficiencies in Complex III or V, leads to fatty acid accumulation and reduced histone acetylation levels, ultimately impeding hepatocyte proliferation [151]. ACSL4, a crucial component in lipid metabolism implicated in the fundamental regulatory molecules of ferroptosis, exhibits a protective role against hepatocellular toxicity and lipid accumulation upon its conditional deletion [152]. The investigation by Singh et al. [153] has elucidated that the downregulation of hepatic ACSL4 in hepatocytes within a murine model fed a hyperlipidemic diet results in a diminished accumulation of lipids and an exacerbation of insulin resistance. Another lipid metabolic modulator, lysophosphatidylcholine acyltransferase 3 (LPCAT3), has been identified through genetic analysis, which can induce iron-death by accumulating fatty acids [79]. Doll's team found that ACSL4 deficiency enhances cellular resistance to GPX4-deficient ferroptosis, whereas LPCAT3 knockout fails to afford robust protection, implying a potential alternative system between these enzymes [88]. Naturally occurring omega-3 PUFAs serve as principal peroxidation precursors in ferroptosis, exhibiting certain tumor suppressive mechanisms (Fig. 3) [154].
Lipid peroxidation, catalyzed by mitochondria-associated membranes through calcium transport and lipid remodeling, accelerates ferroptotic cell death by promoting the peroxidation of PUFAs [155]. In MDS patients, hepatocyte mitochondrial DNA depletion leads to ROS accumulation, which drives lysosomal ferritin degradation and iron release, triggering a lipid peroxidation-ferroptosis cascade [156]. Although peripheral lipolysis has been confirmed as a major source of lipids in the regenerating liver [151], the role of hepatic de novo lipogenesis in ferroptosis remains to be elucidated. Furthermore, it remains questionable whether lipid metabolites such as ketones have protective effects.
Amino acid metabolism: the double-edged sword of iron homeostasis
Amino acids indirectly affect hepatic metabolism by modulating glucagon secretion [157]. Dysregulation of these macronutrients can lead to oxidative damage, induce liver disease, and potentially result in organ failure [158]. Research indicates that certain amino acids are directly associated with the modulation of ferroptosis. GSH acts as a universal endogenous antioxidant and inhibition of its transport via the amino acid reverse transporter SLC7A11 is adequate to provoke iron toxicity via depleting GSH levels [159]. Gu et al.'s research has uncovered that mTORC2 is intricately involved in the regulation of cystine uptake and GSH metabolism, linking changes in growth factor receptor signaling, amino acid metabolism, and ROS regulation in cancer [160]. This finding also underscores the pivotal role of cystine/cysteine in mediating ferroptosis within cells, establishing a link to disease pathology (Fig. 3).
Recent studies have indicated that the absence of other amino acids, such as arginine, prevents cysteine depletion from triggering ferroptosis, highlighting the complementary roles of multiple amino acids in modulating iron-death [104]. In NAFLD, elevated plasma amino acids activate mTORC1 signaling, promoting hepatocyte proliferation while exacerbating iron toxicity [161]. In hereditary hemochromatosis, downregulation of the cystine-glutamate transporter directly exacerbates ferroptosis-driven liver injury, while N-acetylcysteine supplementation restores glutathione levels and inhibits ferroptotic cell death [54]. We hypothesize that the variable regulatory roles of amino acid metabolism in ferroptosis may be due to differences in the differing amino acid ratios. Future research should address:(I)the specific mechanisms by which BCAA/AAA ratios modulate iron metabolism; (II) the potential role of amino acid metabolites (e.g., α-ketoglutarate) in regulating iron-related gene expression through epigenetic modifications.
Gut-liver axis: the hidden regulator of ferroptosis
Ferroptosis within the tumor microenvironment is well-studied; however, the hepatic microenvironment remains understudied and represents a burgeoning area of inquiry. The gut-liver axis is indispensable in the study of the hepatic microenvironment. Specifically, intestinal barrier disruption leads to microbiota translocation and endotoxinemia, triggering hepatic immune cell activation and the secretion of inflammatory cytokines, which can further exacerbate hepatic injury [162]. A plethora of preclinical study models has substantiated that the dysregulation of the gut-liver axis and the impetus of inflammatory mediators exert a biphasic effect on the process of ferroptosis, with a mechanism that is intricate. For instance, certain metabolic byproducts of intestinal bacteria have the capacity to suppress the process of ferroptosis in the context of intestinal ischemia–reperfusion injury [163, 164]. Whereas certain metabolites of bile acids are found to promote ferroptosis [165].
In a hepatic sepsis model, elevated levels of iron-overload markers including ROS, MDA, and TNF-α indicate a possible link between ferroptosis and inflammatory responses [166]. The imbalance of the gut-liver axis, together with the impetus of ferroptosis, can sometimes have a complex and additive effect on the evolution of liver disease. In NAFLD, gut-liver axis imbalances reduce beneficial microbes and increase intestinal permeability, leading to heightened microbial metabolite levels that drive inflammation, which are key factors in NAFLD progression [167, 168]. Ferroptosis intensifies NAFLD progression by inducing insulin resistance, excessive ROS production, and hepatic fibrosis through the accumulation of toxic iron levels [169]. Research also indicates that hepatocyte iron burden promotes fibrosis progression by upregulating HO-1 expression and increasing FGF21 levels, thereby amplifying ferrous cell apoptosis [170]. Yu et al. [171] have identified the NRF2/HO-1 pathway as a crucial regulator of the hepatic microenvironment in ferroptosis, with its activation playing a protective role against oxidative stress in intestinal and hepatic cells. There is an anticipation for further investigation into the role of ferroptosis within the microenvironment of the gut-liver axis, potentially offering new treatment strategies for various liver diseases in the future (Fig. 3).
Mitochondria: the nexus of iron-dependent mechanisms
As previously discussed, mitochondrial morphological aberrations are the among pivotal features of ferroptosis. In probing the mechanisms of ferroptosis within the hepatic microenvironment, we question whether such mitochondrial anomalies are a driving force or simply a corollary of ferroptosis?
Mitochondria, pivotal in the TCA cycle, mediate the metabolism of carbohydrates, lipids, and GSH-related amino acids. Research indicates that the TCA cycle heightens cellular susceptibility to iron toxicity, particularly when cysteine is scarce [172]. Furthermore, a multitude of key anti-ferroptotic components are also localized in the mitochondria, such as the CoQ-FSP1 pathway, the BH4-GCH1 axis, and GPX4. Mitochondria function as the nexus for iron processing and metabolism, and they amplify the cell's vulnerability to ferroptosis by facilitating increased iron import via Mitoferrin 1 (MFRN1) [173]. Mitochondria contribute to ferroptosis via several mechanisms: (I)Energy Metabolism Imbalance: ETC dysfunction reduces ATP synthesis, activates AMPK signaling, and promotes lipid peroxidation [156]; (II)Organelle Interactions: MAM-mediated ER-mitochondrial calcium signaling and lipid exchange directly modulate ferroptosis sensitivity [155]; (III)Mitophagy: Impaired mitophagy of damaged mitochondria leads to ROS accumulation and ferroptosis induction. In DGUOK mutation-associated mitochondrial DNA depletion syndrome, abnormal mitochondrial-lysosomal interactions drive ferritin degradation and iron release, contributing to liver failure [156]. In summary, we propose that mitochondria serve dual roles as both "triggers" and "defenders" in the process of ferroptosis.
Pathogenic narratives of liver-related diseases in ferroptosis
NAFLD
Globally, the prevalence of nonalcoholic fatty liver disease (NAFLD), now increasingly referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is eacalating, impacting approximately one-quarter of the world's population and solidifying its position as a leading cause of chronic liver conditions and hepatic damage [174]. NAFLD includes two pathological conditions: nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH), and its main pathological manifestations are lipid accumulation in the liver, apoptosis of hepatocytes and fibrosis processes from mild to significant [175].
Twenty years ago, researchers noticed that over ninety percent of patients with NAFLD had substantially greater concentrations of plasma oxidation indicators, such as malondialdehyde [69] and 4-hydroxynonenal (4-HNE) [176]. Metabolic dysfunction-induced iron surplus may escalate the severity of NAFLD, as indicated by the presence of hepatic iron deposition in affected individuals, with the amelioration of hepatic injury following iron removal [177, 178]. Recent highlight research indicates that in a murine model of nonalcoholic steatohepatitis (NASH) induced by a choline-deficient and ethionine-supplemented [25] diet, the administration of Trolox, a vitamin E analogue, has significantly decreased the levels of serum hepatic injury markers, attenuated the count of necrotic cells staining positively for specific biomarkers, and mitigated inflammatory responses [179]. Nagita et al. [180]demonstrated that in MASLD, compromised GSH-GPX4 axis and decreased lipophilic antioxidants enhance ferroptosis, resulting in hepatic mitochondrial dysfunction. Metabolomic investigations show that hepatocellular GSH concentrations are lower in a specific group of patients with MASLD, which is accompanied by poor production of VLDL [181]. The gut microbiota modulates bile acid metabolism, with metabolites like lithocholic acid triggering ferroptotic cell death among MASLD patients [165]. Additionally, hepatocytes undergoing ferroptosis release molecules that can activate adjacent immune cells, such as Kupffer cells, thereby influencing hepatic inflammation and fibrotic processes [182]. Targeting this vulnerability with sorafenib or similar agents could represent a novel therapeutic approach, selectively inducing ferroptosis resistance in cancer cells [183, 184]. Understanding the mechanisms of ferroptosis in NAFLD is essential for devising innovative treatment approaches (Fig. 4).
Pathogenic narratives of liver-related diseases in ferroptosis. Ferroptosis is implicated in range of liver diseases, including non-alcoholic fatty liver disease, metabolic dysfunction-associated steatotic liver disease, alcoholic liver disease, Ischemia reperfusion injury and so on. Abbreviations: 4-HNE, 4-hydroxynonenal; MDA, malondialdehyde; VLDL, very Low Density Lipoprotein; Lip-1, liproxstatin-1; Fer-1, ferrostatin-1; α-KG, α-Ketoglutaric acid; CISD1, CDGSH iron-sulfur domain-containing protein 1; IDO1, indoleamine 2,3-dioxygenase 1; RNS, reactive nitrogen species; ART, artemether; HSC, hematopoietic stem cell; HO-1, heme oxygenase-1; HMGB1, high mobility group box 1; ACSL4, acyl-coenzyme synthetase long-chain family member 4; HH, hemochromatosis; HCC, hepatocellular carcinoma; DILI, drug-induced liver injury; ALF, acute liver failure
ALD
Within the spectrum of chronic liver diseases, in addition to NAFLD and MASLD, alcoholic liver disease (ALD) is also a prevalent condition. Alcohol abuse stands as a significant contributor to global morbidity and mortality, with ALD being a prevalent consequence of such misuse [185]. The condition arises from the toxic effects of excessive alcohol, whose metabolites disrupt cellular functions like protein homeostasis, lipid synthesis, redox equilibrium, culminating in hepatocyte death [186]. Iron-overload is a critical factor leading to chronic hepatic injury, and its inhibition can effectively reverse the detrimental effects on the liver [67]. In a murine model of ALD subjected to ethanol feeding for approximately two weeks, it has been demonstrated that the ferroptosis inhibitor, Ferrostatin-1, can ameliorate hepatic injury and toxicity [187]. Likewise, dimethyl fumarate protects against alcohol-induced oxidative stress by suppressing iron-death through the activation of the Nrf2 element, which enhances cellular resistance to this form of cell death [188].
Specifically, there is debate surrounding the mechanisms leading to ferroptosis in hepatocytes due to chronic ethanol accumulation. Luo et al. [189]indicated that merely inhibiting GPx4 does not suffice to trigger ferroptosis, implying that prolonged ethanol exposure might disrupt the methionine cycle, eventually leading to significant GSH depletion and the indirect inactivation of GPx4. Other researchers suggest that GPx4 remains intact at both transcriptional and translational levels, possibly due to a cysteine-consuming mechanism that hinders the System Xc− cystine-glutamate antiporter [190]. Briefly, ferroptosis is a downstream process caused by excessive ethanol buildup, which results in hepatocyte damage. Additionally, ferroptosis also participates in the genesis and progression of ALD through interactions with other organs within the hepatic microenvironment, primarily involving the liver-gut and the liver-adipose tissue axis [191]. Sirtuin1 (SIRT1), a well-established histone deacetylase enzyme with significant protective effects against alcoholic hepatitis [192]. Elevating the levels of SIRT1 in the gut counteracts the negative effects of alcohol on iron balance and stops the onset of ferroptosis, therefore promoting the healing process of liver damage [193]. In the context of liver-adipose tissue axis, elevated lipin-1 levels aggravate abnormal iron deposition and increases hepatic MDA levels in GPx4-independent manner, thereby aggravating alcoholic steatohepatitis and hepatobiliary injury [193]. In chronic liver disease, SIRT1 activation, intestinal enzyme modulation, and liver-adipose tissue axis are key in mitigating ALD-induced ferroptosis and liver injury (Fig. 4).
IRI
Ischemia reperfusion injury (IRI) is also a common type of chronic liver disease. Strictly speaking, IRI is a pathological state, with impacts that are not confined to the liver but also occur in other vital organs, including the heart and kidneys, leading to a series of adverse physiological reactions [194]. In the field of liver transplantation, IRI is a pivotal issue, triggering local inflammation and immune responses that could escalate into rejection, negatively impacting graft function and patient outcomes [195]. During surgery, the liver inevitably experiences an ischemic period followed by reperfusion, leading to IRI. This process induces cellular stress and triggers ferroptosis-associated death pathways.
Significantly, research has confirmed the pathophysiological link between ferroptosis and IRI in liver models [196]. The dysregulation of the GSH/GPX4 axis, precipitating ferroptosis, significantly promotes IRI and the inhibitor lip-1 effectively counteracts hepatic damage [196]. Yamada et al. [197]revealed that elevated serum ferritin, a biomarker of ferroptosis in donors, is an independent risk factor for liver damage post-pediatric living donor liver transplantation. Notably, ferroptosis inhibitors like fer-1 and alpha-tocopherol significantly reduce hepatic injury, lipid peroxidation, and inflammation, whereas the high-iron diet intensifies liver-IRI [197]. Hence, elucidating the mechanisms of ferroptosis in liver transplantation is crucial for devising new treatments to ameliorate IRI and enhance graft survival (Fig. 4).
HCC
Hepatocellular carcinoma (HCC) is a grave malignant neoplasm, ranking as the globe's sixth frequently occurring cancer and stands as the third primary source of cancer-related mortality [198]. The etiology of HCC is multifactorial, with early symptoms often being insidious [199]. Chronic liver diseases previously discussed, including NAFLD, MASLD, and ALD, may progress to cirrhosis and potentially escalate to hepatocellular carcinoma [198]. Following its approval by the FDA in 2007, Sorafenib, an oral multi-targeted tyrosine kinase inhibitor, has emerged as a foundational component of the primary therapeutic regimen for HCC [200]. Nevertheless, the emergence of drug resistance limits the therapeutic efficacy of sorafenib, posing an ongoing challenge in the management of HCC. An array of studies has demonstrated a significant correlation between sorafenib resistance and the onset of ferroptosis in hepatocytes, with multiple key molecules of the ferroptosis pathway implicated in the initiation, progression, and prognosis of HCC.
Iron, an essential trace element, has been demonstrated to accelerate the onset of HCC through high-iron diets, a phenomenon that may be intimately linked to the activation of the ferroptosis pathway [201]. HCC cells can be protected from ferroptosis by activating the p62-Keap1-Nrf2 path, which blocks the degradation of iron-death resistant components [87]. The retinoblastoma protein (Rb), a pivotal tumor suppressor, is commonly deactivated in the pathogenesis of various malignancies [202]. Interestingly, loss of Rb activity does not worsen HCC progression; rather, by exacerbating ferroptosis, it improves sorafenib's effectiveness to treat liver cancer [202].
According to this study, the mechanism would entail a disturbance of iron homeostasis, which would lead to a build-up of lipid ROS and subsequently improve sorafenib's therapeutic efficacy. CDGSH iron-sulfur domain 1 (CISD1) functions as a negative regulator of ferroptosis in HCC, with upregulation observed following erastin treatment [43]. Reducing iron toxicity accumulation and mitochondrial dysfunction through CISD1 gene inhibition enhances the process of erastin-induced iron ptosis, which does not involve GPx4 [43]. The aforementioned fundamental regulator of ferroptosis, ACSL4, also serves as a key predictor of cellular susceptibility to ferroptosis [88]. In liver tissues of patients who exhibit a positive response to sorafenib, ACSL4 is highly expressed, whereas the opposite is observed in patients with poor treatment outcomes [88]. In advanced HCC patients treated with sorafenib, ACSL4 potentially acts as a prognostic indicator for both overall and disease-free survival [203].
Exploring the mechanism of sorafenib-induced ferroptosis in hepatocellular carcinoma cells is a valuable research domain. Yao's research reveals that the leukemia inhibitory factor receptor (LIFR) is crucial in the context of sorafenib-induced ferroptosis [204]. The absence of LIFR expression in cells hinders the ability of sorafenib to initiate this form of cell death [204]. In addition, ferroptosis holds significant research value in HCC prognosis. The high-risk group had a much lower overall survival rate than the low-risk group in the latest study that used a machine-learning model to classify patients into risk groups and find a new genetic marker for HCC prognosis associated to iron deposition (Fig. 4) [205].
DILI
Drug-induced liver injury (DILI) refers to hepatic harm that occurs during the therapeutic use of medications, attributable to the drugs themselves or their metabolites. DILI is a rare adverse event that occurs in about 14 to 19 occurrences per 100,000 people and can manifest as either acute or chronic, with a spectrum of clinical presentations ranging from asymptomatic elevations in liver enzymes to severe hepatic dysfunction [206].
In the academic discourse, the prevailing research delineates DILI into two principal subclasses: intrinsic hepatotoxicity and idiosyncratic hepatotoxicity. The medications implicated in intrinsic DILI are typically lipophilic, enabling them to permeate freely through the lipid membrane that surrounds hepatocellular [207]. Acetaminophen (APAP)-induced intrinsic DILI is relatively common in clinical practice, constituting majority of the cases that progress from intrinsic DILI to acute liver failure [208, 209]. Idiosyncratic hepatotoxicity is an uncommon condition, affecting a select population and lacking a clear dose-dependent relationship [210]. It represents an unpredictable form of liver injury associated with a genetically susceptible individual's unique response to medication. Accordingly, the APAP-induced DILI model is the most prevalent experimental model in animal studies, effectively mimicking the intrinsic DILI toxicity mechanisms observed in humans [211].
Research indicates that in APAP-induced models, a reduction in GSH levels is a common occurrence [212]. Given the exhaustion of GSH is a pivotal mechanism in ferroptosis, these findings support the notion that ferroptosis may be a key pathological process in intrinsic DILI. Similarly, some scholars have found a decrease in GSH and an increase in MDA levels in APAP cellular models [213]. In animal models, APAP also induces a cascade of reactions associated with ferroptosis [76]. Through mass spectrometry analysis, investigators observed an elevation in the levels of lipid peroxides derived from n-6 PUFAs subsequent to APAP treatment, predominantly reflecting the oxidation of arachidonic acid (AA), with this oxidation primarily occurring via an autoxidation mechanism [76]. Upon treatment with ferroptosis inhibitors, a significant reduction in the pathophysiological manifestations was observed in the murine models, concomitant with a substantial increase in overall survival rates [76]. Furthermore, the elimination of ACSL4 significantly attenuates the iron accumulation toxicity induced by APAP, highlighting the potential therapeutic relevance of targeting ACSL4 in counteracting APAP-induced hepatotoxicity [76].
In a recent study incorporating methodologies from the field of biomaterials science, the EaV probe was utilized to monitor alterations within acetaminophen-induced liver injury models. The study unraveled the diminished esterase activity and heightened viscosity during DILI, which may be associated with the generation of ROS during ferroptosis [214]. Research into ferroptosis has uncovered novel therapeutic targets and shed light on the pathophysiology of DILI (Fig. 4).
AIH
Autoimmune Hepatitis (AIH) is a chronic liver condition characterized by aberrant immune system attacks on the liver, predominantly mediated by autoantibodies, and is typically associated with elevated levels of immunoglobulin G (IgG) [215]. AIH exhibits a spectrum of severity, with clinical manifestations ranging from mild dysfunction to severe hepatic impairment. Without effective treatment, AIH can progress to cirrhosis, hepatic failure, or HCC. Consequently, patients often require long-term immunosuppressive therapy to manage disease progression. The histological characteristics of AIH predominantly encompass extensive lymphocytic infiltration and the formation of rosette-like structures by hepatocytes, however, the precise mechanisms underlying these phenomena remain to be elucidated [216]. Concanavalin A (ConA) induces acute liver injury, mimicking AIH, and serves as a key model for studying its mechanisms and treatments [208]. The preliminary treatment with apoptosis inhibitors does not improve ConA-induced liver damage, indicating that this model is suitable for investigating the specific function of necroptotic cell death in the setting of autoimmune hepatitis [217].
Indoleamine 2,3-dioxygenase 1(IDO1), an intracellular heme enzyme, predominantly engages in the modulation of immune responses [218]. The activation of IDO1 enhances lipid peroxidation, which constitutes a pivotal event in the process of ferroptosis [219]. In the ConA-induced murine model of AIH, the upregulation of IDO1 is closely associated with liver injury, an increase in reactive nitrogen species (RNS), and the promotion of ferroptosis [220]. Conversely, the inhibition of IDO1 can mitigate hepatocyte ferroptosis by activating the expression of SLC7A1, also known as xCT [220]. Caveolin-1 (Cav-1), serving as a crucial plasma membrane signaling protein, plays a pivotal protective role in the process of ferroptosis during AIH [221]. Cav-1 exerts a protective effect in AIH by diminishing reactive nitrogen species (RNS), thereby alleviating hepatocyte ferroptosis and shielding the liver from immunologically driven damage [221]. The modulation of Cav-1 expression is intricately linked to the regulation of ferroptosis and RNS, highlighting its critical role in AIH pathogenesis (Fig. 4).
Fibrosis
Hepatic fibrosis represents an early pathological state of the liver, wherein the activation of hepatic stellate cells (HSCs) constitutes the cornerstone of the fibrogenic process [222, 223]. Sustained inflammation and oxidative stress can promote the activation and proliferation of HSCs, exacerbating the deposition of extracellular matrix and leading to the formation of fibrosis [224]. Research suggests that one possible therapeutic approach for healing fibrosis could be to regulate iron-death. Scholarly research has shown that improving hepatic fibrosis in mouse models can be achieved by increasing the iron-chelating action generated by sorafenib [225].Artemether (ART) prevents stimulated HSCs from causing fibrogenicity by restoring normal iron homeostasis and lipophilic peroxidation through increasing p53 transcription and nuclear binding [226]. HO-1, an intracellular enzyme implicated in anti-ferroptotic mechanisms, ameliorates fibrosis by inhibiting the expansion and stimulation of HSCs upon induction with magnesium isoglycyrrhizinate [227] (Fig. 4).
ALF
Acute liver failure [208] represents a rapidly progressive and severe hepatic condition, which typically afflicts individuals without a pre-existing history of chronic liver disease, culminating in profound hepatic impairment over a short period [228]. ALF is most clinically associated with APAP, but it can also result from hepatotoxicity due to a variety of other medications [229]. The initiation of ferroptosis in the acetaminophen-induced ALF experimental model is accompanied by a marked elevation in peroxides that manufactured from n-6 PUFAs [76]. Furthermore, by preserving mitochondrial functional homeostasis, it has been observed that the iron-overload inhibitors UAMC-3203 and VBIT-12 both mitigate ferroptosis in the model [230]. The other standard method for investigating ALF in mice entails administering lipopolysaccharide (LPS) and D-galactosamine (GalN) to induce hepatic injury [231]. Reducing iron-overload by by using glycyrrhizic acid, an inhibitor of HMGB1, has been demonstrated to reduce liver damage by lowering cellular oxidative damage [231]. Collectively, these investigations imply that ferroptosis could represent a promising therapeutic target for ALI, but additional data are essential to elucidate the intricate mechanisms involved(Fig. 4).
HH
Hemochromatosis [232] is a genetic disorder arising from mutations in genes associated with iron homeostasis [233]. The disorder enhances intestinal iron absorption, leading to iron deposition in parenchymal cells, which in turn induces liver injury and organ dysfunction, with other factors like alcohol abuse and hemorrhage also aggravating iron accumulation [233]. Although the precise role of ferroptosis in the pathogenesis of HH remains unclear, recent research suggests that it may be involved in the pathological processes of HH [234]. Investigators have observed the occurrence of ferroptosis in murine models of HH and have discerned a potentially significant role for ferroptosis in the pathogenic processes of HH [54]. Further gene expression analysis revealed that iron treatment upregulates the expression of solute carrier family 7 member 11 (Slc7a11), a gene previously implicated in ferroptosis [54]. Slc7a11 knockout mice exhibit increased susceptibility to ferroptosis under high-iron conditions, indicating a protective role for Slc7a11 in iron-overload-induced cell death [54]. Subsequent studies should further explore the link between ferroptosis and HH to better understand its pathology and inform new treatment approaches(Fig. 4).
Malaria
Ferroptosis plays a complex role in pathogen infection, with some pathogens potentially leveraging ferroptosis to facilitate their survival and dissemination, while host cells may induce ferroptosis to restrict pathogen infection. Malaria represents a grave public health challenge on a global scale, caused by infections with Plasmodium parasites, predominantly transmitted to humans through the bite of infected female Anopheles mosquitoes [235]. Cerebral malaria [198] is one of the most severe complications associated with malaria, and even with treatment and recovery, it can lead to significant neurological sequelae [236]. Liang et al. [237] have observed a significant elevation of markers indicative of iron-death, MDA and intracellular Fe2 + , in a murine model. In the CM model, neuronal damage is positively correlated with the downregulation of GPX4 expression, and Fer-1 can partially reverse this damage [237]. The iron toxicity and oxidative damage regulated by the System Xc−/GPX4 axis in hepatocytes may control malaria parasite infection, according to an experimental research [238]. Despite advancements, there is a paucity of research on ferroptosis in malaria, warranting further investigation into its mechanisms(Fig. 4).
Ferroptosis manifests several common features across various liver diseases, such as iron metabolism dysregulation, phospholipid peroxidation, and dysfunctional mitochondria. Nevertheless, distinct liver diseases exhibit unique characteristics in the triggers, sensitivity, and clinical treatment strategies of ferroptosis. Etiological factors of various liver diseases are detailed in the preceding text. Notably, these diseases show varying sensitivities to ferroptosis, requiring distinct pharmacological modulation strategies. Most inflammatory liver diseases require ferroptosis inhibitors to reduce the number of necrotic hepatocytes in order to reverse disease progression. Pharmacological modulation of ferroptosis in distinct hepatic microenvironments exhibits subtle differences in the treatment of drug-resistant hepatocellular carcinoma and other liver disorders. Several ferroptosis inducers have been proven to be effective in treating various types of cancer cells that exhibit insensitivity to apoptosis [239]. Notably, in 2019, researchers initiated exploration into the potential of combining immunotherapy with ferroptosis induction for cancer treatment. Anti-PD-L1 antibodies activate T cells to secrete IFN-γ, leading to downregulation of system xc⁻, enhancing cancer cells' susceptibility to ferroptosis and facilitating the cancer cell death process [240].
Clinical treatment strategies by targeting ferroptosis
In the realm of ferroptosis research, significant advancements have been made from elucidating mechanisms to medicine. This review encapsulates recent advancements in research, highlighting novel targets and agents, such as natural compounds, synthetic inhibitors, and clinical trial-stage candidates, all centered on ferroptosis strategies, to offer guidance for future therapeutic developments (Table 3).
In NAFLD, there is currently no standard clinical intervention. Many researchers are exploring new approaches based on mechanisms related to ferroptosis. Researchers have employed a high-fat diet to induce a NAFLD model in APOE−/− mice, revealing that testosterone deficiency can exacerbate hepatic iron deposition in hepatocytes by modulating BMAL1 [267]. Qiu et al. [241] have identified that Nuciferine can ameliorate the accumulation of ferrous iron by modulating the PPARα signaling pathway, suggesting their potential as therapeutic agents. In natural compounds, diosgenin and AP-derived polysaccharides show promise in mitigating ferroptosis and hepatic lipid accumulation, while the small molecule inhibitor GLX351322 combats oxidative damage by upregulating GPX4 [242, 243, 245]. Recent findings also indicate a notable negative correlation between lipophilic vitamins and MAFLD [246].
In the field of ALD research, scholars have constructed models of long-term ethanol exposure in cells, mice, and zebrafish to explore the efficacy of various drugs and targets. Research indicates that inhibiting the secretion of extracellular nicotinamide phosphoribosyltransferase (eNAMPT) from brown adipose tissue (BAT) induced by ethanol can reduce cellular ferroptosis, showing promise as a potential therapeutic target for ALD [268]. Additionally, artificially extracted sulfated polysaccharides have been shown to reduce iron toxicity accumulation and reverse liver damage [247]. Quercetin mitigates ethanol-induced liver damage by modulating PERK-dependent MAMs formation and inhibiting ferroptosis [248]. Poria cocos polysaccharides ameliorate ALD-induced liver damage by suppressing the NF-κB-mediated inflammatory pathway [249]. Selenium and ferrostatin-1 mitigate alcohol-induced oxidative damage by upregulating GPX4 and SLC7A11, with selenium exhibiting greater effectiveness [250].
IRI, a state of functional impairment post-organ transplantation, has seen recent advancements in targeted therapies for hepatocyte ferroptosis. Traditional Chinese medicine has demonstrated value in the study of liver IRI. Wogonin exerts hepatoprotective effects in IRI by inhibiting inducible nitric oxide synthase (iNOS) and mitigating phospholipid peroxidation [251]. Rehmannia glutinosa mitigates iron toxicity accumulation in liver IRI by inhibiting ZIP14 and promoting iron ion efflux mediated by transferrin, thereby alleviating ferroptotic damage [253]. Ticlopidine, primarily known for its antiplatelet aggregation effects, also demonstrates hepatoprotective properties in IRI by reducing the expression of the ferroptosis marker PTGS2 [254].
The role of ferroptosis in HCC differs from its role in other liver diseases, thus necessitating a distinct therapeutic approach based on ferroptosis. Sorafenib serves as the primary treatment for HCC, but its high resistance limits efficacy. Donafenib, a newer alternative, features enhanced tritium atom stability in its chemical structure [269]. Research findings indicate that Donafenib can downregulate the expression of ferroptosis resistance factors SLC7A11 and GPX4, enhancing ferroptosis and apoptosis in hepatic cells, thereby demonstrating its therapeutic potential [256]. In a parallel-controlled Phase II-III clinical trial, donafenib demonstrated significantly better overall survival than sorafenib, with a median OS of 12.1 months versus 10.3 months, and exhibited superior safety and tolerability profiles [270]. To address sorafenib resistance, researchers have investigated novel therapies using sorafenib-resistant hepatocellular carcinoma cell lines as experimental models. Inhibiting GSTA1 enzyme enhances sorafenib sensitivity and ferroptosis in hepatic cells, improving sorafenib efficacy [260]. And the use of the SOCE inhibitor SKF96365 in conjunction with sorafenib significantly enhances the responsiveness of SR cells to sorafenib [261]. In 2024, researchers developed GOA/miR-targeted nanocomposites that efficiently encapsulate miRNA inhibitors, inducing non-iron-dependent ferroptosis to significantly inhibit hepatic cell growth [259]. These nanocomposites exhibit excellent biocompatibility and immune activation potential, offering a promising biotherapeutic strategy for HCC treatment [259].
Similar to various other injurious liver diseases, DILI can also be mitigated by targeting and inhibiting the ferroptotic pathway to alleviate hepatic damage. Recent studies have unveiled innovative strategies, demonstrating that both the novel drug mifepristone (RU486) and the natural compound xanthohumol [151] can protect against DILI by activating ferroptosis-resistant molecules like Nrf2 [263, 264]. Oral fecal microbiota capsules have been shown to induce mitophagy and activate antioxidant molecules, mitigating DILI-induced ferroptotic hepatocyte apoptosis, offering potential for improved treatment efficacy [266]. MnO2Nfs, a novel nanomaterial, shows promise in biomedicine by significantly boosting intracellular glutathione and reducing oxidative damage, acting as a powerful ferroptosis inhibitor in the future [265].
Overall, within the field of hepatology, targeting ferroptosis offers a promising avenue for the treatment of liver diseases. Traditional ferroptosis-targeting drugs mainly include iron chelators like deferoxamine and deferiprone, antioxidants such as glutathione and vitamin E, and Nrf2 activators including sulforaphane and curcumin, all of which have shown promise in mitigating liver damage (see Table 1). Expanding on this, the review highlights recent progress in iron-targeting drug development (Table 3): (I) natural compounds like Diosgenin, Nuciferine, and xanthohumol; artificially derived sulfated polysaccharides [241, 263]; and small molecule inhibitors such as GLX351322 have demonstrated favorable outcomes in experimental assessments [243]; (II) improvements in traditional drugs: Donafenib offers enhanced overall survival compared to sorafenib [256], and Ticlopidine provides additional benefits [254]; (III) Development of novel nanomaterials: GOA/miR-targeted nanocomposites, mitochondrial-targeted antioxidant MitoTEMPO, and MnO2Nfs as potent inhibitors of ferroptosis [96, 259, 265].
While drugs targeting ferroptosis hold promise, they also present challenges: (I) lack of specificity: where iron chelators may disrupt normal cellular iron metabolism and cause iron-deficiency anemia; (II) delivery efficiency: as oral administration can lead to insufficient drug concentrations in the liver due to gastrointestinal factors, thus reducing efficacy; (III) individual variability: where genetic background, disease state, and comorbidities can affect drug response; (IV) potential hepatotoxicity, with some drugs causing liver function abnormalities; (V) the experimental nature of new targeted therapies, where unreported negative results may exist.
Conclusion
Ferroptosis, a form of regulated cell death distinct from apoptosis, autophagy, and necrosis, has emerged as a critical player in the pathogenesis of various liver diseases [271]. Characterized by iron toxicity accumulation, phospholipid peroxidation and dysfunctional mitochondria, ferroptosis is intricately linked to the hepatic microenvironment, influencing and being influenced by factors such as iron homeostasis, glucose metabolism, lipid metabolism, amino acid metabolism, and the gut-liver axis.
In this review, we have explored the multifaceted role of ferroptosis in liver diseases, emphasizing its significance in both the progression and potential treatment of conditions such as NAFLD, ALD, IRI, HCC, DILI and AIH. Our analysis reveals that regulatory proteins like NRF2, FSP1, and GPx4 are not only central to the execution of ferroptosis but also to the development of liver tumors and liver failure.
Furthermore, we highlight the importance of understanding the liver microenvironment in ferroptosis. Iron homeostasis, a key determinant of hepatic health, is disrupted in diseases such as hemochromatosis and fibrosis, leading to ferroptosis. Similarly, glucose and lipid metabolism disruptions, common in NAFLD, have been shown to induce ferroptosis, underscoring the metabolic nature of these conditions. The gut-liver axis also emerges as a significant factor in the modulation of ferroptosis. Disruptions in this axis can lead to increased susceptibility to ferroptosis, highlighting the importance of considering the holistic hepatic environment in disease pathogenesis.
Despite the burgeoning interest in ferroptosis and its implications in a spectrum of liver diseases, numerous gaps persist in the current understanding. The predominant research is confined to animal or cellular models, with a relative dearth of studies examining its role in human subjects [58, 272]. This disparity warrants further investigation. Moreover, while the upstream regulatory molecules of the ferroptotic process have been the focus of considerable attention, our knowledge of the downstream molecular players remains alarmingly scarce. The exploration of ferroptosis inhibitors is another area ripe for advancement; for instance, classic inhibitors such as fer-1 have been associated with issues of metabolic latency, indicating a need for more nuanced pharmacological strategies [273].
Building upon the preceding discourse, several queries and contemplations emerge: (I) At what juncture or stage should the ferroptosis process in specific pathological liver diseases be targeted for intervention? (II) Given the inherent heterogeneity in the efficacy of ferroptosis-related inhibitors and inducers across various liver diseases, can a unified assessment of intervention outcomes be achieved in the future? (III) In the context of liver cancer, is it possible to activate ferroptosis selectively in hepatocellular carcinoma cells without affecting healthy cells? (IV) Can combinatorial therapeutic strategies based on distinct ferroptosis regulatory molecules enhance treatment efficacy?
In conclusion, while ferroptosis presents a complex and nuanced mechanism in liver disease, it also offers a wealth of opportunities for therapeutic intervention. The development of targeted ferroptosis therapies, such as the use of small molecules to modulate key regulatory proteins, offers a promising frontier in the treatment of liver diseases. As our understanding of the molecular mechanisms of ferroptosis deepens, so does our potential to develop novel, effective treatments that can improve outcomes for patients with liver diseases.
Data availability
No datasets were generated or analysed during the current study.
References
Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486–541.
Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol. 2019;70:151–71.
Chen PH, Wu J, Ding CC, et al. Kinome screen of ferroptosis reveals a novel role of ATM in regulating iron metabolism. Cell Death Differ. 2020;27:1008–22.
Wu Y, Du L, Xu X, et al. Nano Self-Assemblies of Caffeic Acid-Fibronectin Mimic a Peptide Conjugate for the Treatment of Corneal Epithelial Injury. Mol Pharm. 2023;20:5937–46.
Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59:298–308.
Gout P, Buckley A, Simms C, Bruchovsky N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the xc− cystine transporter: a new action for an old drug. Leukemia. 2001;15:1633–40.
Song Q, Peng S, Sun Z, Heng X, Zhu X. Temozolomide Drives Ferroptosis via a DMT1-Dependent Pathway in Glioblastoma Cells. Yonsei Med J. 2021;62:843–9.
Li J, Lama R, Galster SL, et al. Small-Molecule MMRi62 Induces Ferroptosis and Inhibits Metastasis in Pancreatic Cancer via Degradation of Ferritin Heavy Chain and Mutant p53. Mol Cancer Ther. 2022;21:535–45.
Yoshioka H, Kawamura T, Muroi M, et al. Identification of a Small Molecule That Enhances Ferroptosis via Inhibition of Ferroptosis Suppressor Protein 1 (FSP1). ACS Chem Biol. 2022;17:483–91.
Yang J, Jia Z, Zhang J, et al. Metabolic Intervention Nanoparticles for Triple-Negative Breast Cancer Therapy via Overcoming FSP1-Mediated Ferroptosis Resistance. Adv Healthc Mater. 2022;11: e2102799.
Ning Y, Fang S, Zhang R, et al. Simvastatin induces ferroptosis and activates anti-tumor immunity to sensitize anti-PD-1 immunotherapy in microsatellite stable gastric cancer. Int Immunopharmacol. 2024;142: 113244.
Shin D, Kim EH, Lee J, Roh J-L. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radical Biol Med. 2018;129:454–62.
Lin R, Zhang Z, Chen L, et al. Dihydroartemisinin (DHA) induces ferroptosis and causes cell cycle arrest in head and neck carcinoma cells. Cancer Lett. 2016;381:165–75.
Huan H, Lyamzaev KG, Panteleeva AA, Chernyak BV. Mitochondrial lipid peroxidation is necessary but not sufficient for induction of ferroptosis. Front Cell Dev Biol. 2024;12:1452824.
Roh J-L, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 2017;11:254–62.
Nishizawa S, Araki H, Ishikawa Y, et al. Low tumor glutathione level as a sensitivity marker for glutamate-cysteine ligase inhibitors. Oncol Lett. 2018;15:8735–43.
Gaschler MM, Andia AA, Liu H, et al. FINO(2) initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat Chem Biol. 2018;14:507–15.
Mao C, Liu X, Zhang Y, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593:586–90.
Zilka O, Shah R, Li B, et al. On the mechanism of cytoprotection by ferrostatin-1 and liproxstatin-1 and the role of lipid peroxidation in ferroptotic cell death. ACS Cent Sci. 2017;3:232–43.
Xiang X, Xu M, Liu L, et al. Liproxstatin-1 attenuates acute hypertriglyceridemic pancreatitis through inhibiting ferroptosis in rats. Sci Rep. 2024;14:9548.
Liu T, Shu J, Liu Y, et al. Atorvastatin attenuates ferroptosis-dependent myocardial injury and inflammation following coronary microembolization via the Hif1a/Ptgs2 pathway. Front Pharmacol. 2022;13:1057583.
Feng H, Stockwell BR. Unsolved mysteries: How does lipid peroxidation cause ferroptosis? PLoS Biol. 2018;16: e2006203.
Sun Q, Liu D, Cui W, et al. Cholesterol mediated ferroptosis suppression reveals essential roles of Coenzyme Q and squalene. Commun Biol. 2023;6:1108.
Luo S, Zeng Y, Chen B, et al. Vitamin E and GPX4 cooperatively protect treg cells from ferroptosis and alleviate intestinal inflammatory damage in necrotizing enterocolitis. Redox Biol. 2024;75: 103303.
Eberhard Y, McDermott SP, Wang X, et al. Chelation of intracellular iron with the antifungal agent ciclopirox olamine induces cell death in leukemia and myeloma cells. Blood, The Journal of the American Society of Hematology. 2009;114:3064–73.
Zhang H, Wang Y, Wang S, et al. Tangeretin alleviates sepsis-induced acute lung injury by inhibiting ferroptosis of macrophage via Nrf2 signaling pathway. Chin Med. 2025;20:11.
Long Z, Yu X, Li S, et al. Sakuranetin Prevents Acetaminophen-Induced Liver Injury via Nrf2-Induced Inhibition of Hepatocyte Ferroptosis. Drug Des Devel Ther. 2025;19:159–71.
Zhou Y, Hu T, Zeng H, et al. Naringenin Inhibits Ferroptosis in Renal Tubular Epithelial Cells of Diabetic Nephropathy Through SIRT1/FOXO3a Signaling Pathway. Drug Dev Res. 2025;86: e70044.
Zhou X, Yang Y, Qiu X, et al. Antioxidant taurine inhibits chondrocyte ferroptosis through upregulation of OGT/Gpx4 signaling in osteoarthritis induced by anterior cruciate ligament transection. J Adv Res. 2025;S2090-1232:00029-3.
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;38:1234–9.
Tuo Q-Z, Masaldan S, Southon A, et al. Characterization of selenium compounds for anti-ferroptotic activity in neuronal cells and after cerebral ischemia–reperfusion injury. Neurotherapeutics. 2021;18:2682–91.
Allard JP, Aghdassi E, Mohammed S, et al. Nutritional assessment and hepatic fatty acid composition in non-alcoholic fatty liver disease (NAFLD): a cross-sectional study. J Hepatol. 2008;48:300–7.
Belavgeni A, Meyer C, Stumpf J, Hugo C, Linkermann A. Ferroptosis and necroptosis in the kidney. Cell Chem Biol. 2020;27:448–62.
Huang Z, Ma Y, Sun Z, Cheng L, Wang G. Ferroptosis: potential targets and emerging roles in pancreatic diseases. Arch Toxicol. 2024;98:75–94.
Dixon SJ, Stockwell BR. The role of iron and reactive oxygen species in cell death. Nat Chem Biol. 2014;10:9–17.
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. 2016;113:E4966–75.
Kagan VE, Mao G, Qu F, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81–90.
Abrams RP, Carroll WL, Woerpel K. Five-membered ring peroxide selectively initiates ferroptosis in cancer cells. ACS Chem Biol. 2016;11:1305–12.
Badgley MA, Kremer DM, Maurer HC, et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science. 2020;368:85–9.
Wang H, Liu C, Zhao Y, Gao G. Mitochondria regulation in ferroptosis. Eur J Cell Biol. 2020;99: 151058.
Wang H, Cheng Y, Mao C, et al. Emerging mechanisms and targeted therapy of ferroptosis in cancer. Mol Ther. 2021;29:2185–208.
Gao M, Yi J, Zhu J, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019;73(354–363): e353.
Yuan H, Li X, Zhang X, Kang R, Tang D. CISD1 inhibits ferroptosis by protection against mitochondrial lipid peroxidation. Biochem Biophys Res Commun. 2016;478:838–44.
Lee H-Y, Nga HT, Tian J, Yi H-S. Mitochondrial metabolic signatures in hepatocellular carcinoma. Cells. 1901;2021:10.
Carlson BA, Tobe R, Yefremova E, et al. Glutathione peroxidase 4 and vitamin E cooperatively prevent hepatocellular degeneration. Redox Biol. 2016;9:22–31.
Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–31.
Cui C, Yang F, Li Q. Post-translational modification of GPX4 is a promising target for treating ferroptosis-related diseases. Front Mol Biosci. 2022;9: 901565.
Hambright WS, Fonseca RS, Chen L, Na R, Ran Q. Ablation of ferroptosis regulator glutathione peroxidase 4 in forebrain neurons promotes cognitive impairment and neurodegeneration. Redox Biol. 2017;12:8–17.
Hassannia B, Van Coillie S, Vanden Berghe T. Ferroptosis: biological rust of lipid membranes. Antioxid Redox Signal. 2021;35:487–509.
Hangauer MJ, Viswanathan VS, Ryan MJ, et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551:247–50.
Sato H, Tamba M, Ishii T, Bannai S. Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J Biol Chem. 1999;274:11455–8.
Arensman MD, Yang XS, Leahy DM, et al. Cystine–glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc Natl Acad Sci. 2019;116:9533–42.
Conrad M, Sato H. The oxidative stress-inducible cystine/glutamate antiporter, system x c − : cystine supplier and beyond. Amino Acids. 2011;42:231–46.
Wang H, An P, Xie E, et al. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology. 2017;66:449–65.
Jiang L, Kon N, Li T, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57–62.
Dodson M, de la Vega MR, Cholanians AB, Schmidlin CJ, Chapman E, Zhang DD. Modulating NRF2 in Disease: Timing Is Everything. Annu Rev Pharmacol Toxicol. 2019;59:555–75.
Fan Z, Wirth AK, Chen D, et al. Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 2017;6:e371–e371.
Sun X, Niu X, Chen R, et al. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 2016;64:488–500.
Sun X, Ou Z, Chen R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2015;63:173–84.
Liu Q, Wang K. The induction of ferroptosis by impairing STAT3/Nrf2/GPx4 signaling enhances the sensitivity of osteosarcoma cells to cisplatin. Cell Biol Int. 2019;43:1245–56.
Yau T, Kang Y-K, Kim T-Y, et al. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients With Advanced Hepatocellular Carcinoma Previously Treated With Sorafenib. JAMA Oncol. 2020;6: e204564.
Qiu Y-b, Wan B-b, Liu G, et al: Nrf2 protects against seawater drowning-induced acute lung injury via inhibiting ferroptosis. Respiratory research 2020, 21:1–16.
Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575:688–92.
Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575:693–8.
Han Z, Ma K, Tao H, et al. A Deep Insight Into Regulatory T Cell Metabolism in Renal Disease: Facts and Perspectives. Front Immunol. 2022;13: 826732.
Dai E, Meng L, Kang R, Wang X, Tang D. ESCRT-III–dependent membrane repair blocks ferroptosis. Biochem Biophys Res Commun. 2020;522:415–21.
Wu J, Wang Y, Jiang R, et al. Ferroptosis in liver disease: new insights into disease mechanisms. Cell death discovery. 2021;7:276.
Madak JT, Bankhead A, Cuthbertson CR, Showalter HD, Neamati N. Revisiting the role of dihydroorotate dehydrogenase as a therapeutic target for cancer. Pharmacol Ther. 2019;195:111–31.
Li L, Ng SR, Colón CI, et al. Identification of DHODH as a therapeutic target in small cell lung cancer. Sci Transl Med. 2019;11:eaaw7852.
Chen J, Li X, Ge C, Min J, Wang F. The multifaceted role of ferroptosis in liver disease. Cell Death Differ. 2022;29:467–80.
Amos A, Amos A, Wu L, Xia H. The Warburg effect modulates DHODH role in ferroptosis: a review. Cell Communication and Signaling. 2023;21:100.
Peters GJ. Re-evaluation of Brequinar sodium, a dihydroorotate dehydrogenase inhibitor. Nucleosides, Nucleotides Nucleic Acids. 2018;37:666–78.
Carlos L. Arteaga TDBTDBJKHSLSTOrKBHJBD: Phase I clinical and pharmacokinetic trial of Brequinar sodium (DuP 785; NSC 368390). Cancer Res. 1989;49:4648–53.
Zhang K, Wu L, Zhang P, et al. miR-9 regulates ferroptosis by targeting glutamic-oxaloacetic transaminase GOT1 in melanoma. Mol Carcinog. 2018;57:1566–76.
Angeli JPF, Shah R, Pratt DA, Conrad M. Ferroptosis Inhibition: Mechanisms and Opportunities. Trends Pharmacol Sci. 2017;38:489–98.
Yamada N, Karasawa T, Kimura H, et al. Ferroptosis driven by radical oxidation of n-6 polyunsaturated fatty acids mediates acetaminophen-induced acute liver failure. Cell Death Dis. 2020;11:144.
Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2016;13:91–8.
Kagan VE, Mao G, Qu F, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2016;13:81–90.
Dixon SJ, Winter GE, Musavi LS, et al. Human Haploid Cell Genetics Reveals Roles for Lipid Metabolism Genes in Nonapoptotic Cell Death. ACS Chem Biol. 2015;10:1604–9.
Yuan H, Li X, Zhang X, Kang R, Tang D. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem Biophys Res Commun. 2016;478:1338–43.
Feng J, Lu P-z, Zhu G-z, et al: ACSL4 is a predictive biomarker of sorafenib sensitivity in hepatocellular carcinoma. Acta Pharmacologica Sinica 2020, 42:160–170.
Ndiaye H, Liu Jorlin Y, Hall A, Minogue S, Morgan Marsha Y, Waugh Mark G. Immunohistochemical staining reveals differential expression of ACSL3 and ACSL4 in hepatocellular carcinoma and hepatic gastrointestinal metastases. Biosci Rep. 2020;40:BSR20200219.
Soula M, Weber RA, Zilka O, et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol. 2020;16:1351–60.
Kraft VAN, Bezjian CT, Pfeiffer S, et al. GTP Cyclohydrolase 1/Tetrahydrobiopterin Counteract Ferroptosis through Lipid Remodeling. ACS Cent Sci. 2019;6:41–53.
Peleman C, Francque S, Berghe TV: Emerging role of ferroptosis in metabolic dysfunction-associated steatotic liver disease: revisiting hepatic lipid peroxidation. eBioMedicine 2024, 102:105088.
Dixon SJ, Lemberg KM, Lamprecht MR, et al: Ferroptosis: an iron-dependent form of nonapoptotic cell death. cell 2012, 149:1060–1072.
Sun X, Ou Z, Chen R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63:173–84.
Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13:91–8.
Wang F, Li J, Zhao Y, et al. miR-672-3p Promotes Functional Recovery in Rats with Contusive Spinal Cord Injury by Inhibiting Ferroptosis Suppressor Protein 1. Oxid Med Cell Longev. 2022;2022:6041612.
Wei Z, Hang S, Wiredu Ocansey DK, et al. Human umbilical cord mesenchymal stem cells derived exosome shuttling mir-129-5p attenuates inflammatory bowel disease by inhibiting ferroptosis. J Nanobiotechnology. 2023;21:188.
Wang X, Yang Y, Zhou X, et al. IFNγ regulates ferroptosis in KFs by inhibiting the expression of SPOCD1 through DNMT3A. Cell Death Discov. 2025;11:9.
Zhang Y, Shi J, Liu X, et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat Cell Biol. 2018;20:1181–92.
Yu Y, Jiang L, Wang H, et al. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood, The Journal of the American Society of Hematology. 2020;136:726–39.
Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X. Ferroptosis is an autophagic cell death process. Cell Res. 2016;26:1021–32.
Yan B, Ai Y, Sun Q, et al. Membrane Damage during Ferroptosis Is Caused by Oxidation of Phospholipids Catalyzed by the Oxidoreductases POR and CYB5R1. Mol Cell. 2021;81:355–369.e310.
Fang X, Wang H, Han D, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci U S A. 2019;116:2672–80.
Garcia-Bermudez J, Baudrier L, Bayraktar EC, et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature. 2019;567:118–22.
Mancias JD, Wang X, Gygi SP, Harper JW, Kimmelman AC. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 2014;509:105–9.
Kotschi S, Jung A, Willemsen N, et al. NFE2L1-mediated proteasome function protects from ferroptosis. Mol Metab. 2022;57: 101436.
Wu M, Zhang X, Zhang W, et al. Cancer stem cell regulated phenotypic plasticity protects metastasized cancer cells from ferroptosis. Nat Commun. 2022;13:1371.
Liu T, Jiang L, Tavana O, Gu W. The Deubiquitylase OTUB1 Mediates Ferroptosis via Stabilization of SLC7A11. Cancer Res. 2019;79:1913–24.
Song X, Xie Y, Kang R, et al. FANCD2 protects against bone marrow injury from ferroptosis. Biochem Biophys Res Commun. 2016;480:443–9.
Zhang Z, Zhang F, An P, et al. Ferroportin1 deficiency in mouse macrophages impairs iron homeostasis and inflammatory responses. Blood. 2011;118:1912–22.
Conlon M, Poltorack CD, Forcina GC, et al. A compendium of kinetic modulatory profiles identifies ferroptosis regulators. Nat Chem Biol. 2021;17:665–74.
Yao F, Deng Y, Zhao Y, et al. A targetable LIFR-NF-κB-LCN2 axis controls liver tumorigenesis and vulnerability to ferroptosis. Nat Commun. 2021;12:7333.
Gao G, Xie Z, Li E-w, et al: Dehydroabietic acid improves nonalcoholic fatty liver disease through activating the Keap1/Nrf2-ARE signaling pathway to reduce ferroptosis. Journal of Natural Medicines 2021, 75:540–552.
Yang L, Wang H, Yang X, et al. Auranofin mitigates systemic iron overload and induces ferroptosis via distinct mechanisms. Signal Transduct Target Ther. 2020;5:138.
Hasegawa M, Takahashi H, Rajabi H, et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget. 2016;7:11756.
Sun X, Ou Z, Xie M, et al. HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene. 2015;34:5617–25.
Fang X, Cai Z, Wang H, et al. Loss of cardiac ferritin H facilitates cardiomyopathy via Slc7a11-mediated ferroptosis. Circ Res. 2020;127:486–501.
Newton K, Strasser A, Kayagaki N, Dixit VM. Cell death. Cell. 2024;187:235–56.
Yuan J, Ofengeim D. A guide to cell death pathways. Nat Rev Mol Cell Biol. 2024;25:379–95.
Rajagopalan K, Selvan Christyraj JD, Chelladurai KS, et al. Understanding the molecular mechanism of regeneration through apoptosis-induced compensatory proliferation studies-updates and future aspects. Apoptosis. 2024;29:1–16.
Green DR. The Mitochondrial Pathway of Apoptosis Part II: The BCL-2 Protein Family. Cold Spring Harb Perspect Biol. 2022;14: a041046.
Wu P, Zhang X, Duan D, Zhao L. Organelle-Specific Mechanisms in Crosstalk between Apoptosis and Ferroptosis. Oxid Med Cell Longev. 2023;2023:1–14.
Perez MA, Magtanong L, Dixon SJ, Watts JL. Dietary Lipids Induce Ferroptosis in Caenorhabditiselegans and Human Cancer Cells. Dev Cell. 2020;54:447–454.e444.
Wang Z, Wang X, Dai X, et al. 2D Catalytic Nanozyme Enables Cascade Enzyodynamic Effect‐Boosted and Ca2+ Overload‐Induced Synergistic Ferroptosis/Apoptosis in Tumor. Adv Mater. 2024;36:e2312316.
Degterev A, Huang Z, Boyce M, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1:112–9.
Geng J, Ito Y, Shi L, et al. Regulation of RIPK1 activation by TAK1-mediated phosphorylation dictates apoptosis and necroptosis. Nat Commun. 2017;8:359.
Kayagaki N, Kornfeld OS, Lee BL, et al. NINJ1 mediates plasma membrane rupture during lytic cell death. Nature. 2021;591:131–6.
Sun L, Wang H, Wang Z, et al. Mixed Lineage Kinase Domain-like Protein Mediates Necrosis Signaling Downstream of RIP3 Kinase. Cell. 2012;148:213–27.
Luo C, Tao L. The Function and Mechanisms of Autophagy in Traumatic Brain Injury. Adv Exp Med Biol. 2020;1207:635–48.
Levine B. Autophagy in cell death: an innocent convict? J Clin Investig. 2005;115:2679–88.
Mizushima N, Levine B. Autophagy in Human Diseases. N Engl J Med. 2020;383:1564–76.
Lee S, Hwang N, Seok BG, Lee S, Lee S-J, Chung SW. Autophagy mediates an amplification loop during ferroptosis. Cell Death & Disease. 2023;14:464.
Liu J, Guo Z-N, Yan X-L, et al. Crosstalk Between Autophagy and Ferroptosis and Its Putative Role in Ischemic Stroke. Front Cell Neurosci. 2020;14:577403.
Patil N, Walsh P, Carrabre K, et al. Regionally Specific Human Pre-Oligodendrocyte Progenitor Cells Produce Both Oligodendrocytes and Neurons after Transplantation in a Chronically Injured Spinal Cord Rat Model after Glial Scar Ablation. J Neurotrauma. 2021;38:777–88.
Chen KW, Demarco B, Heilig R, et al. Extrinsic and intrinsic apoptosis activate pannexin‐1 to drive NLRP 3 inflammasome assembly. The EMBO J. 2019;38:e101638.
Xu R, Yang J, Qian Y, et al. Ferroptosis/pyroptosis dual-inductive combinational anti-cancer therapy achieved by transferrin decorated nanoMOF. Nanoscale Horizons. 2021;6:348–56.
Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature. 1988;335:440–2.
Zhou B, Zhang J-y, Liu X-s, et al: Tom20 senses iron-activated ROS signaling to promote melanoma cell pyroptosis Cell Research 2018, 28:1171–1185.
Almalki WH, Almujri SS. Aging, ROS, and cellular senescence: a trilogy in the progression of liver fibrosis. Biogerontology. 2025;26:10.
Wei X, Liu M, Zheng Z, et al. Defective NCOA4-dependent ferroptosis in senescent fibroblasts retards diabetic wound healing. Cell Death Discovery. 2023;9:138.
Nishizawa H, Matsumoto M, Chen G, et al. Lipid peroxidation and the subsequent cell death transmitting from ferroptotic cells to neighboring cells. Cell Death Dis. 2021;12:332.
Admasu TD, Kim K, Rae M, et al. Selective ablation of primary and paracrine senescent cells by targeting iron dyshomeostasis. Cell Reports. 2023;42:112058.
Wan X, Li C, Tan YH, et al. Dihydroartemisinin eliminates senescent cells by promoting autophagy-dependent ferroptosis via AMPK/mTOR signaling pathway. Cell Biol Int. 2024;48:726–36.
Ayton S, Portbury S, Kalinowski P, et al. Regional brain iron associated with deterioration in Alzheimer’s disease: A large cohort study and theoretical significance. Alzheimers Dement. 2021;17:1244–56.
Yuan W, Xia H, Xu Y, et al. The role of ferroptosis in endothelial cell dysfunction. Cell Cycle. 2022;21:1897–914.
Nicolas G, Bennoun M, Devaux I, et al. Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 ( <i>USF2</i> ) knockout mice. Proc Natl Acad Sci. 2001;98:8780–5.
Roetto A, Papanikolaou G, Politou M, et al. Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis. Nat Genet. 2002;33:21–2.
Park CH, Valore EV, Waring AJ, Ganz T. Hepcidin, a Urinary Antimicrobial Peptide Synthesized in the Liver. J Biol Chem. 2001;276:7806–10.
Pigeon C, Ilyin G, Courselaud B, et al. A New Mouse Liver-specific Gene, Encoding a Protein Homologous to Human Antimicrobial Peptide Hepcidin, Is Overexpressed during Iron Overload. J Biol Chem. 2001;276:7811–9.
Nicolas G, Chauvet C, Viatte L, et al. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin Investig. 2002;110:1037–44.
Ramos E, Kautz L, Rodriguez R, et al. Evidence for distinct pathways of hepcidin regulation by acute and chronic iron loading in mice. Hepatology. 2011;53:1333–41.
Yu Y, Jiang L, Wang H, et al. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood. 2020;136:726–39.
Muckenthaler MU, Rivella S, Hentze MW, Galy B. A red carpet for iron metabolism. Cell. 2017;168:344–61.
Anderson SA, Nizzi CP, Chang Y-I, et al. The IRP1-HIF-2α axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption. Cell Metab. 2013;17:282–90.
Wang R-H, Li C, Xu X, et al. A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression. Cell Metab. 2005;2:399–409.
Shimada K, Hayano M, Pagano NC, Stockwell BR. Cell-line selectivity improves the predictive power of pharmacogenomic analyses and helps identify NADPH as biomarker for ferroptosis sensitivity. Cell Chem Biol. 2016;23:225–35.
Jones JG. Hepatic glucose and lipid metabolism. Diabetologia. 2016;59:1098–103.
Wang X, Menezes CJ, Jia Y, et al: Metabolic inflexibility promotes mitochondrial health during liver regeneration. Science 2024, 384:eadj4301.
Yamada N, Karasawa T, Kimura H, et al. Ferroptosis driven by radical oxidation of n-6 polyunsaturated fatty acids mediates acetaminophen-induced acute liver failure. Cell Death Dis. 2020;11:144.
Singh AB, Kan CFK, Kraemer FB, Sobel RA, Liu J. Liver-specific knockdown of long-chain acyl-CoA synthetase 4 reveals its key role in VLDL-TG metabolism and phospholipid synthesis in mice fed a high-fat diet. American Journal of Physiology-Endocrinology And Metabolism. 2019;316:E880–94.
Han Y-M, Jeong M, Park J-M, et al. The ω-3 polyunsaturated fatty acids prevented colitis-associated carcinogenesis through blocking dissociation of β-catenin complex, inhibiting COX-2 through repressing NF-κB, and inducing 15-prostaglandin dehydrogenase. Oncotarget. 2016;7:63583.
Zhang Z, Zhou H, Gu W, et al. CGI1746 targets σ(1)R to modulate ferroptosis through mitochondria-associated membranes. Nat Chem Biol. 2024;20:699–709.
Guo J, Duan L, He X, et al. A Combined Model of Human iPSC-Derived Liver Organoids and Hepatocytes Reveals Ferroptosis in DGUOK Mutant mtDNA Depletion Syndrome. Adv Sci (Weinh). 2021;8:2004680.
Cusi K. Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology. 2012;142(711–725): e716.
Masoodi M, Gastaldelli A, Hyötyläinen T, et al. Metabolomics and lipidomics in NAFLD: biomarkers and non-invasive diagnostic tests. Nat Rev Gastroenterol Hepatol. 2021;18:835–56.
Sun Y, Zheng Y, Wang C, Liu Y. Glutathione depletion induces ferroptosis, autophagy, and premature cell senescence in retinal pigment epithelial cells. Cell Death Dis. 2018;9:753.
Gu Y, Albuquerque CP, Braas D, et al. mTORC2 regulates amino acid metabolism in cancer by phosphorylation of the cystine-glutamate antiporter xCT. Mol Cell. 2017;67(128–138): e127.
Winther-Sørensen M, Holst JJ, Wewer Albrechtsen NJ. The feedback cycles between glucose, amino acids and lipids and alpha cell secretion and their role in metabolic fatty liver disease. Curr Opin Lipidol. 2023;34:27–31.
Marshall JC. The gut as a potential trigger of exercise-induced inflammatory responses. Can J Physiol Pharmacol. 1998;76:479–84.
Wang F, Liu X, Huang F, et al: Gut microbiota-derived gamma-aminobutyric acid from metformin treatment reduces hepatic ischemia/reperfusion injury through inhibiting ferroptosis. Elife 2024, 12:RP89045.
Deng F, Zhao B-C, Yang X, et al. The gut microbiota metabolite capsiate promotes Gpx4 expression by activating TRPV1 to inhibit intestinal ischemia reperfusion-induced ferroptosis. Gut microbes. 2021;13:1902719.
Liu S, Gao Z, He W, et al. The gut microbiota metabolite glycochenodeoxycholate activates TFR-ACSL4-mediated ferroptosis to promote the development of environmental toxin–linked MAFLD. Free Radical Biol Med. 2022;193:213–26.
Wang J, Zhu Q, Li R, Zhang J, Ye X, Li X. YAP1 protects against septic liver injury via ferroptosis resistance. Cell Biosci. 2022;12:163.
Li Q, Liu W, Zhang H, et al. α-D-1, 3-glucan from Radix Puerariae thomsonii improves NAFLD by regulating the intestinal flora and metabolites. Carbohyd Polym. 2023;299: 120197.
Ji J, Wu L, Wei J, Wu J, Guo C. The gut microbiome and ferroptosis in MAFLD. J Clin Transl Hepatol. 2023;11:174.
Haber AL, Biton M, Rogel N, et al. A single-cell survey of the small intestinal epithelium. Nature. 2017;551:333–9.
Wu A, Feng B, Yu J, et al. Fibroblast growth factor 21 attenuates iron overload-induced liver injury and fibrosis by inhibiting ferroptosis. Redox Biol. 2021;46: 102131.
Yu X, Wang S, Ji Z, et al. Ferroptosis: An important mechanism of disease mediated by the gut-liver-brain axis. Life Sci. 2024;347:122650.
Fu C, Cao N, Zeng S, et al. Role of mitochondria in the regulation of ferroptosis and disease. Front Med. 2023;10:1301822.
Zhang T, Sun L, Hao Y, et al. ENO1 suppresses cancer cell ferroptosis by degrading the mRNA of iron regulatory protein 1. Nature Cancer. 2021;3:75–89.
Younossi Z, Anstee QM, Marietti M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. 2017;15:11–20.
Yatsuji S, Hashimoto E, Tobari M, Taniai M, Tokushige K, Shiratori K. Clinical features and outcomes of cirrhosis due to non-alcoholic steatohepatitis compared with cirrhosis caused by chronic hepatitis C. J Gastroenterol Hepatol. 2009;24:248–54.
Loguercio C, De Girolamo V, de Sio I, et al. Non-alcoholic fatty liver disease in an area of southern Italy: main clinical, histological, and pathophysiological aspects. J Hepatol. 2001;35:568–74.
Nelson JE, Wilson L, Brunt EM, et al. Relationship between the pattern of hepatic iron deposition and histological severity in nonalcoholic fatty liver disease. Hepatology. 2010;53:448–57.
Valenti L, Moscatiello S, Vanni E, et al. Venesection for non-alcoholic fatty liver disease unresponsive to lifestyle counselling–a propensity score-adjusted observational study. QJM. 2010;104:141–9.
Tsurusaki S, Tsuchiya Y, Koumura T, et al. Hepatic ferroptosis plays an important role as the trigger for initiating inflammation in nonalcoholic steatohepatitis. Cell Death & Disease. 2019;10:449.
Nagita A, Ando M. Assessment of hepatic vitamin E status in adult patients with liver disease. Hepatology. 1997;26:392–7.
Alonso C, Fernández-Ramos D, Varela-Rey M, et al. Metabolomic identification of subtypes of nonalcoholic steatohepatitis. Gastroenterology. 2017;152(1449–1461): e1447.
Tran S, Baba I, Poupel L, et al. Impaired Kupffer cell self-renewal alters the liver response to lipid overload during non-alcoholic steatohepatitis. Immunity. 2020;53(627–640): e625.
Pfister D, Núñez NG, Pinyol R, et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature. 2021;592:450–6.
Hassannia B, Vandenabeele P, Berghe TV. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.
Kourkoumpetis T, Sood G. Pathogenesis of alcoholic liver disease: an update. Clin Liver Dis. 2019;23:71–80.
Gao B, Bataller R. Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology. 2011;141:1572–85.
Liu C-Y, Wang M, Yu H-M, et al. Ferroptosis is involved in alcohol-induced cell death in vivo and in vitro. Biosci Biotechnol Biochem. 2020;84:1621–8.
Zhang Y, Zhao S, Fu Y, et al. Computational repositioning of dimethyl fumarate for treating alcoholic liver disease. Cell Death Dis. 2020;11:641.
Luo J, Song G, Chen N, et al. Ferroptosis contributes to ethanol-induced hepatic cell death via labile iron accumulation and GPx4 inactivation. Cell Death Discovery. 2023;9:311.
Mao L, Zhao T, Song Y, et al. The emerging role of ferroptosis in non-cancer liver diseases: hype or increasing hope? Cell Death Dis. 2020;11:518.
Poole LG, Dolin CE, Arteel GE. Organ–organ crosstalk and alcoholic liver disease. Biomolecules. 2017;7:62.
Yin H, Hu M, Liang X, et al. Deletion of SIRT1 from hepatocytes in mice disrupts lipin-1 signaling and aggravates alcoholic fatty liver. Gastroenterology. 2014;146:801–11.
Zhou Z, Ye TJ, Bonavita G, et al. Adipose-specific lipin-1 overexpression renders hepatic ferroptosis and exacerbates alcoholic steatohepatitis in mice. Hepatology Communications. 2019;3:656–69.
Li Y, Yang Y, Feng Y, et al. A review of melatonin in hepatic ischemia/reperfusion injury and clinical liver disease. Ann Med. 2014;46:503–11.
Ali JM, Davies SE, Brais RJ, et al. Analysis of ischemia/reperfusion injury in time-zero biopsies predicts liver allograft outcomes. Liver Transpl. 2015;21:487–99.
Friedmann Angeli JP, Schneider M, Proneth B, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 2014;16:1180–91.
Yamada N, Karasawa T, Wakiya T, et al. Iron overload as a risk factor for hepatic ischemia-reperfusion injury in liver transplantation: potential role of ferroptosis. Am J Transplant. 2020;20:1606–18.
Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma Nature reviews Disease primers. 2021;7:1–28.
Bruix J, Sherman M. Management of hepatocellular carcinoma. Hepatology. 2005;42:1208–36.
Dixon SJ, Patel DN, Welsch M, et al: Pharmacological inhibition of cystine–glutamate exchange induces endoplasmic reticulum stress and ferroptosis. elife 2014, 3:e02523.
Fonseca-Nunes A, Jakszyn P, Agudo A. Iron and cancer risk—a systematic review and meta-analysis of the epidemiological evidence. Cancer Epidemiol Biomark Prev. 2014;23:12–31.
Louandre C, Marcq I, Bouhlal H, et al. The retinoblastoma (Rb) protein regulates ferroptosis induced by sorafenib in human hepatocellular carcinoma cells. Cancer Lett. 2015;356:971–7.
Sun X-J, Xu G-L. Overexpression of Acyl-CoA Ligase 4 (ACSL4) in patients with hepatocellular carcinoma and its prognosis. Medical science monitor: international medical journal of experimental and clinical research. 2017;23:4343.
Yao F, Deng Y, Zhao Y, et al. A targetable LIFR− NF-κB− LCN2 axis controls liver tumorigenesis and vulnerability to ferroptosis. Nat Commun. 2021;12:7333.
Liang J-y, Wang D-s, Lin H-c, et al. A novel ferroptosis-related gene signature for overall survival prediction in patients with hepatocellular carcinoma. Int J Biol Sci. 2020;16:2430.
Sgro C, Clinard F, Ouazir K, et al. Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology. 2002;36:451–5.
Chen M, Borlak J, Tong W. High lipophilicity and high daily dose of oral medications are associated with significant risk for drug-induced liver injury. Hepatology. 2013;58:388–96.
Heymann F, Hamesch K, Weiskirchen R, Tacke F. The concanavalin A model of acute hepatitis in mice. Lab Anim. 2015;49:12–20.
Chalasani NP, Maddur H, Russo MW, Wong RJ, Reddy KR, Gastroenterology PPCotACo: ACG clinical guideline: diagnosis and management of idiosyncratic drug-induced liver injury. Official journal of the American College of Gastroenterology| ACG 2021, 116:878–898.
G. Aithal ESBNKGAK-UDL: EASL Clinical Practice Guidelines: Drug-induced liver injury. Null. 1970;70:1222–61.
Mossanen J, Tacke F. Acetaminophen-induced acute liver injury in mice. Lab Anim. 2015;49:30–6.
Du K, Ramachandran A, Jaeschke H. Oxidative stress during acetaminophen hepatotoxicity: Sources, pathophysiological role and therapeutic potential. Redox Biol. 2016;10:148–56.
Wang Z, Hao W, Hu J, et al. Maltol Improves APAP-Induced Hepatotoxicity by Inhibiting Oxidative Stress and Inflammation Response via NF-κB and PI3K/Akt Signal Pathways. Antioxidants. 2019;8:395.
Liu J, Xu X, Xie P, Yang X, Ye Y, Zhao Y. A novel activatable fluorescent probe for revealing the dynamic of esterase and viscosity during ferroptosis and DILI. Sens Actuators, B Chem. 2023;396: 134594.
Trivedi PJ, Hubscher SG, Heneghan M, Gleeson D, Hirschfield GM. Grand round: autoimmune hepatitis. J Hepatol. 2019;70:773–84.
Webb GJ, Hirschfield GM, Krawitt EL, Gershwin ME. Cellular and Molecular Mechanisms of Autoimmune Hepatitis. Annu Rev Pathol. 2018;13:247–92.
Künstle G, Hentze H, Germann PG, Tiegs G, Meergans T, Wendel A. Concanavalin A hepatotoxicity in mice: Tumor necrosis factor–mediated organ failure independent of caspase-3–like protease activation. Hepatology. 1999;30:1241–51.
Grohmann U. Tolerance, DCs and tryptophan: much ado about IDO. Trends Immunol. 2003;24:242–8.
Prendergast GC, Malachowski WJ, Mondal A, Scherle P, Muller AJ. Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer. Int Rev Cell Mol Biol. 2018;336:175–203.
Zeng T, Deng G, Zhong W, et al. Indoleamine 2, 3-dioxygenase 1enhanceshepatocytes ferroptosis in acute immune hepatitis associated with excess nitrative stress. Free Radical Biol Med. 2020;152:668–79.
Deng G, Li Y, Ma S, et al. Caveolin-1 dictates ferroptosis in the execution of acute immune-mediated hepatic damage by attenuating nitrogen stress. Free Radical Biol Med. 2020;148:151–61.
Karin D, Koyama Y, Brenner D, Kisseleva T. The characteristics of activated portal fibroblasts/myofibroblasts in liver fibrosis. Differentiation. 2016;92:84–92.
Tacke F, Trautwein C. Mechanisms of liver fibrosis resolution. J Hepatol. 2015;63:1038–9.
Paik YH, Kim JK, Lee JI, et al. Celecoxib induces hepatic stellate cell apoptosis through inhibition of Akt activation and suppresses hepatic fibrosis in rats. Gut. 2009;58:1517–27.
Zhang Z, Yao Z, Wang L, et al. Activation of ferritinophagy is required for the RNA-binding protein ELAVL1/HuR to regulate ferroptosis in hepatic stellate cells. Autophagy. 2018;14:2083–103.
Wang L, Zhang Z, Li M, et al. P53-dependent induction of ferroptosis is required for artemether to alleviate carbon tetrachloride-induced liver fibrosis and hepatic stellate cell activation. IUBMB Life. 2018;71:45–56.
Sui M, Jiang X, Chen J, Yang H, Zhu Y. Magnesium isoglycyrrhizinate ameliorates liver fibrosis and hepatic stellate cell activation by regulating ferroptosis signaling pathway. Biomed Pharmacother. 2018;106:125–33.
Stravitz RT, Lee WM. Acute liver failure. The Lancet. 2019;394:869–81.
Habib S, Shaikh OS. Drug-Induced Acute Liver Failure. Clin Liver Dis. 2017;21:151–62.
Niu B, Lei X, Xu Q, et al. Protecting mitochondria via inhibiting VDAC1 oligomerization alleviates ferroptosis in acetaminophen-induced acute liver injury. Cell Biol Toxicol. 2021;38:505–30.
Wang Y, Chen Q, Shi C, Jiao F, Gong Z. Mechanism of glycyrrhizin on ferroptosis during acute liver failure by inhibiting oxidative stress. Mol Med Reports. 2019;20:4081–90.
Muhammad A, Iftikhar N, Mashhood A, et al. Comparison of efficacy of platelet-rich plasma (PRP) with PRP microneedling in androgenetic alopecia. Cureus. 2022;14:e30418.
Powell LW, Seckington RC, Deugnier Y. Haemochromatosis. The Lancet. 2016;388:706–16.
Ong SY, Gurrin LC, Dolling L, et al. Reduction of body iron in HFE-related haemochromatosis and moderate iron overload (Mi-Iron): a multicentre, participant-blinded, randomised controlled trial. The Lancet Haematology. 2017;4:e607–14.
Cotter C, Sturrock HJW, Hsiang MS, et al. The changing epidemiology of malaria elimination: new strategies for new challenges. The Lancet. 2013;382:900–11.
Siddiqui AJ, Adnan M, Jahan S, Redman W, Saeed M, Patel M. Neurological disorder and psychosocial aspects of cerebral malaria: what is new on its pathogenesis and complications? A minireview. Folia Parasitologica. 2020;67:2020.015.
Liang J, Shen Y, Wang Y, et al. Ferroptosis participates in neuron damage in experimental cerebral malaria and is partially induced by activated CD8+ T cells. Mol Brain. 2022;15:57.
Kain HS, Glennon EKK, Vijayan K, et al. Liver stage malaria infection is controlled by host regulators of lipid peroxidation. Cell Death Differ. 2019;27:44–54.
Tsoi J, Robert L, Paraiso K, et al. Multi-stage Differentiation Defines Melanoma Subtypes with Differential Vulnerability to Drug-Induced Iron-Dependent Oxidative Stress. Cancer Cell. 2018;33:890–904.e895.
Wang W, Green M, Choi JE, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569:270–4.
Qiu J, Le Y, Liu N, et al. Nuciferine Alleviates High-Fat Diet- and ApoE(-/-)-Induced Hepatic Steatosis and Ferroptosis in NAFLD Mice via the PPARα Signaling Pathway. J Agric Food Chem. 2024;72:24417–31.
Zhang X, Yin G, Chen S, et al. Diosgenin ameliorating non-alcoholic fatty liver disease via Nrf2-mediated regulation of oxidative stress and ferroptosis. Diabetes Obes Metab. 2024;26:5745–56.
Wu S, Guo N, Xu H, et al. Caveolin-1 ameliorates hepatic injury in non-alcoholic fatty liver disease by inhibiting ferroptosis via the NOX4/ROS/GPX4 pathway. Biochem Pharmacol. 2024;230: 116594.
Wang H, Shan C, Guo G, Ning D, Miao F. Therapeutic potential of palmitoleic acid in non-alcoholic fatty liver disease: Targeting ferroptosis and lipid metabolism disorders. Int Immunopharmacol. 2024;142: 113025.
Pi D, Liang Z, Pan M, et al. Atractylodes lancea Rhizome Polysaccharide Alleviates MCD Diet-Induced NASH by Inhibiting the p53/mTOR Pathway. Int J Mol Sci. 2024;25:11112.
Miao Y, Jiang Z, Song H, et al. Vitamin D supplementation alleviates high fat diet-induced metabolic associated fatty liver disease by inhibiting ferroptosis pathway. Eur J Nutr. 2024;64:50.
Khazaei M, Ardeshir RA. Protective effects of sulfated polysaccharides from Enteromorpha intestinalis on oxidative stress, liver iron overload and Ferroptosis in Zebra fish exposed to ethanol. Biomed Pharmacother. 2024;181: 117715.
Lin H, Guo X, Liu J, et al. Ethanol-Induced Hepatic Ferroptosis Is Mediated by PERK-Dependent MAMs Formation: Preventive Role of Quercetin. Mol Nutr Food Res. 2024;68: e2300343.
Zhou X, Wang J, Zhou S. Poria cocos polysaccharides improve alcoholic liver disease by interfering with ferroptosis through NRF2 regulation. Aging (Albany NY). 2024;16:6147–62.
Chen F, Li Q, Xu X, Wang F. Selenium Attenuates Ethanol-induced Hepatocellular Injury by Regulating Ferroptosis and Apoptosis. Turk J Gastroenterol. 2024;35:778–86.
Jia D, Wu K, Luo J, et al. Wogonin Alleviates DCD Liver Ischemia/Reperfusion Injury by Regulating ALOX15/iNOS-mediated Ferroptosis. Transplant. 2024;108:2374–85.
Pi Y, Zuo H, Wang Y, et al. Oleanolic acid alleviating ischemia-reperfusion injury in rat severe steatotic liver via KEAP1/NRF2/ARE. Int Immunopharmacol. 2024;138:112617.
Zhang J, Ruiz M, Bergh PO, et al. Regulation of meiotic telomere dynamics through membrane fluidity promoted by AdipoR2-ELOVL2. Nat Commun. 2024;15:2315.
Ma Y, Yao X, Zou Y, et al. Ticlopidine protects hepatic ischemia-reperfusion injury via suppressing ferroptosis. Biochem Biophys Res Commun. 2024;733:150436.
Zhang ZY, Yang ZH, Wang S, Feng SL, Wang XL, Mao JY. Regulation of optimized new Shengmai powder on cardiomyocyte apoptosis and ferroptosis in ischemic heart failure rats: The mediating role of phosphatidylinositol-3-kinase/protein kinase B/tumor protein 53 signaling pathway. J Ethnopharmacol. 2024;330: 118264.
Liang J, Chen M, Yan G, et al. Donafenib activates the p53 signaling pathway in hepatocellular carcinoma, induces ferroptosis, and enhances cell apoptosis. Clin Exp Med. 2025;25:29.
Liu Z, Liu K, Shi S, et al. Alkali injury-induced pathological lymphangiogenesis in the iris facilitates the infiltration of T cells and ocular inflammation. JCI Insight. 2024;9:e175479.
Liu W, Liu C, Xiao J, et al. HTRA1 interacts with SLC7A11 to modulate colorectal cancer chemosensitivity by inhibiting ferroptosis. Cell Death Discov. 2024;10:228.
Qin Y, Ling X, Li Y, et al. Histidine phosphatase-ferroptosis crosstalk modulation for efficient hepatocellular carcinoma treatment. J Nanobiotechnology. 2024;22:622.
Ma S, Xie F, Wen X, et al. GSTA1/CTNNB1 axis facilitates sorafenib resistance via suppressing ferroptosis in hepatocellular carcinoma. Pharmacol Res. 2024;210:107490.
Ren R, Chen Y, Zhou Y, et al. STIM1 promotes acquired resistance to sorafenib by attenuating ferroptosis in hepatocellular carcinoma. Genes Dis. 2024;11:101281.
Jiang Y, Hui D, Pan Z, et al. Curcumin promotes ferroptosis in hepatocellular carcinoma via upregulation of ACSL4. J Cancer Res Clin Oncol. 2024;150:429.
Deng Y, Chu X, Li Q, et al. Xanthohumol ameliorates drug-induced hepatic ferroptosis via activating Nrf2/xCT/GPX4 signaling pathway. Phytomedicine. 2024;126:155458.
Chen J, Shi Z, Zhang C, Xiong K, Zhao W, Wang Y. Oroxin A alleviates early brain injury after subarachnoid hemorrhage by regulating ferroptosis and neuroinflammation. J Neuroinflammation. 2024;21:116.
Wu A, Li M, Chen Y, et al. Multienzyme Active Manganese Oxide Alleviates Acute Liver Injury by Mimicking Redox Regulatory System and Inhibiting Ferroptosis. Adv Healthc Mater. 2024;13:e2302556.
Yang CJ, Chang HC, Sung PC, et al. Oral fecal transplantation enriches Lachnospiraceae and butyrate to mitigate acute liver injury. Cell Rep. 2024;43:113591.
Fan Y, Ren Y, Deng L, et al. Testosterone deficiency aggravates diet-induced non-alcoholic fatty liver disease by inducing hepatocyte ferroptosis via targeting BMAL1 in mice. Int Immunopharmacol. 2025;144: 113641.
Zhou Y, Pang N, Li W, et al. Inhibition of ethanol-induced eNAMPT secretion attenuates liver ferroptosis through BAT-Liver communication. Redox Biol. 2024;75:103274.
Chen R, Ielasi L, di Carlo A, Tovoli F. Donafenib in hepatocellular carcinoma. Drugs of Today. 2023;59:83–90.
Qin S, Bi F, Gu S, et al. Donafenib Versus Sorafenib in First-Line Treatment of Unresectable or Metastatic Hepatocellular Carcinoma: A Randomized, Open-Label, Parallel-Controlled Phase II-III Trial. J Clin Oncol. 2021;39:3002–11.
Fortes GB, Alves LS, De Oliveira R, et al. Heme induces programmed necrosis on macrophages through autocrine TNF and ROS production. Blood, The Journal of the American Society of Hematology. 2012;119:2368–75.
Bai T, Wang S, Zhao Y, Zhu R, Wang W, Sun Y. Haloperidol, a sigma receptor 1 antagonist, promotes ferroptosis in hepatocellular carcinoma cells. Biochem Biophys Res Commun. 2017;491:919–25.
Devisscher L, Van Coillie S, Hofmans S, et al. Discovery of novel, drug-like ferroptosis inhibitors with in vivo efficacy. J Med Chem. 2018;61:10126–40.
Funding
The work was supported by by the Lu'an City Science and Technology Plan Project (Grant No. 2023kykt07).
Author information
Authors and Affiliations
Contributions
JC and ZH were involved in the conception of the study. YH, YL, JS, MS, XN, HS, CX were involved in writing the article. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
He, Y., Lin, Y., Song, J. et al. From mechanisms to medicine: Ferroptosis as a Therapeutic target in liver disorders. Cell Commun Signal 23, 125 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-025-02121-2
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-025-02121-2