IMR Press / FBL / Volume 28 / Issue 12 / DOI: 10.31083/j.fbl2812332
Open Access Review
Lipid Peroxidation in Ferroptosis and Association with Nonalcoholic Fatty Liver Disease
Show Less
1 School of Clinical Medicine, Changchun University of Chinese Medicine, 130117 Changchun, Jilin, China
2 State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022 Changchun, Jilin, China
*Correspondence: ccguoyan@163.com (Yan Guo); xzyin@ciac.ac.cn (Xunzhe Yin)
Front. Biosci. (Landmark Ed) 2023, 28(12), 332; https://doi.org/10.31083/j.fbl2812332
Submitted: 23 May 2023 | Revised: 28 July 2023 | Accepted: 21 August 2023 | Published: 6 December 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Nonalcoholic fatty liver disease (NAFLD) constitutes a commonly diagnosed liver pathology with perturbed lipid metabolism, which is mainly caused by excessive accumulation of fat in hepatocytes by various pathogenic factors. Currently, there are no effective drug treatments for NAFLD. Ferroptosis represents a novel form of programmed cell death depending on iron, which is driven by large cellular amounts of reactive oxygen species (ROS) and lipid peroxides. Ferroptosis plays critical regulatory roles in the pathogenesis of NAFLD, and overaccumulation of Fe2+ contributes to lipid peroxidation, which subsequently aggravates NAFLD. Therefore, ferroptosis suppression might constitute an important target for NAFLD treatment. This article reviews the discovery, production pathways, and defense mechanisms of ferroptosis, and explores its association with NAFLD. This may provide new reference targets and strategies for the development of NAFLD drugs from the perspective of ferroptosis.

Keywords
ferroptosis
lipid peroxidation
nonalcoholic fatty liver disease
ferritinophagy
1. Introduction

Nonalcoholic fatty liver disease (NAFLD) constitutes the commonest chronic liver disease, affecting about 25% of humans globally and projected to become the main reason for liver transplantation by 2030, therefore representing a worldwide public health issue [1, 2, 3]. Current evidence shows that NAFLD is a generic term designating a multisystem disease, including a range of liver diseases with metabolic risk factors [4, 5]. NAFL refers to hepatic steatosis, in which fat is present in more than 5% of hepatocytes. Subsequently, nonalcoholic steatohepatitis (NASH) occurs after NAFL aggravates different injuries. NAFL/NASH is associated with hepatocellular damage and usually has four phases: (1) fat overaccumulation in hepatocytes, also termed NAFL; (2) hepatocyte ballooning degeneration and diffuse lobular inflammation; (3) fibrosis, caused by inflammation and hepatocellular damage, which features an excessive extracellular matrix (ECM) accumulation that alters the normal hepatocyte features; (4) cirrhosis that sustainably progresses to the terminal stage of liver disease. Approximately 20% of NAFL cases progress to NASH, and more than 40% of NASH cases develop fibrosis; liver transplantation is considered in case of severe cirrhosis [6, 7].

Recently, ferroptosis has rapidly attracted attention among scientists studying chronic liver disease, as excessive accumulation of iron, which is the main feature of most liver diseases, can cause oxidative damage in the liver. Evidence suggests ferroptosis is tightly associated with lipid peroxidation (LPO) in NAFLD and can therefore be an important target for preventing and treating NAFLD [8]. This article reviews the generation and regulation of ferroptosis and elucidates the association of LPO in ferroptosis with NAFLD.

2. Ferroptosis
2.1 Development of Ferroptosis

Ferroptosis, a newly identified iron-dependent cell death, is characterized by iron accumulation and LPO, and has different morphological, biochemical and genetic characteristics from other forms of programmed cell death. Ultrastructural analysis showed the key morphological features distinguishing ferroptosis from apoptosis, necrosis and autophagy include cell membrane rupture and blister, decreased mitochondrial size relative to normal, mitochondrial ridge reduction or disappearance, and increased membrane density [9].

LPO is a key factor inducing ferroptosis, with polyunsaturated fatty acids (PUFAs) generally serving as reaction substrates. Carbon-carbon double bonds and carbon-hydrogen bonds in PUFAs are transformed to generate PUFAs-CoA by acyl-CoA synthetase long chain family member 4 (ACSL4), and PUFAs-coenzyme A (PUFAs-CoA) are then esterified by lysophosphatidylcholine acyltransferase 3 (LPCAT3) into substrate PUFAs-PL in membrane phospholipids. Therefore, ACSL4 is considered a ferroptosis marker [10]. Free radicals interact with the hydrogen of PUFAs-PL to form the phospholipid radical (PL), which then reacts with oxygen to generate phospholipid peroxyl radical (PL-OO). Hydrogen is extracted from other PUFAs to generate lipid peroxides (PL-OOH) and PL, and so on. However, excessive Fe2+ catalyzes the reduction of PL-OOH to PL-O through the Fenton reaction, which further promotes non-enzymatic free radical chain reaction, ultimately altering cell membrane integrity and inducing ferroptosis [11].

LPO occurs in many subcellular structures such as mitochondria, endoplasmic reticulum (ER) and lysosomes [12]. Reactive oxygen species (ROS) generated by the tricarboxylic acid (TCA) cycle and electron transport chain (ETC) in the mitochondria, can target PUFAs-double bonds to produce LPO and induce ferroptosis [13]. Moreover, ER stress has a definite correlation with ferroptosis, and lysosomal ROS may also participate in ferroptosis as ferrostatin was observed in lysosomes [14, 15]. However, the specific mechanism remains undefined. Reactive lipid species generated by LPO include 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), whose contents are the highest and associated with ferroptosis [16].

2.2 Defense Mechanisms against Ferroptosis

Currently, three pathways are known to inhibit LPO to prevent ferroptosis, including the glutathione pathway (GSH-GPX4), the ubiquinone pathway (NADPH-FSP1-CoQ10) and the tetrahydrobiopterin pathway (GCH1-BH4) [17]. Firstly, the glutathione peroxidase 4 (GPX4) pathway is one of the commonest defense systems, which mainly depends on the Xc- transporter system, glutathione (GSH) synthesis and GPX4 activity. The Xc- system transports equal amounts of extracellular cystine and intracellular glutamic acid. Cystine then undergoes reduction by GSH to generate cysteine, thereby interfering with LPO reactions and preventing ferroptosis. Nuclear factor erythroid 2-related factor 2 (Nrf2) represents a major upstream regulatory gene of defense systems that can target the subunit solute carrier family 7 member 11 (SLC7A11), the light chain subunit of the Xc- system [18]. Studies have suggested that drugs may activate Nrf2 to initiate the GPX4 defense mechanism to mediate ferroptosis and reduce the occurrence and development of diseases, especially NAFLD [19, 20, 21]. This provides a promising strategy for developing drug candidates targeting Nrf2 for the treatment of NAFLD.

Secondly, ferroptosis suppressor protein 1 (FSP1) is a newly reported strong and effective inhibitor of ferroptosis. The FSP1 pathway does not depend on GSH for its inhibitory effect, but instead functions by regulating coenzyme Q10 (CoQ10) [22]. Acetylated FSP1 is targeted to the plasma membrane and CoQ10 stimulates NAD(P)H to shuttle into the lipid bilayer with the help of FSP1; then, free radicals in the droplets are regulated to capture antioxidants, maintaining the quality of lipids and preventing LPO from inducing ferroptosis [23]. Currently, relatively few studies have examined the BH4 pathway. BH4 participates in the antioxidant system and the overexpression of its rate-limiting enzyme GTP cyclohydrolase 1 (GCH1) selectively inhibits the peroxidation of some PUFAs. Therefore, ferroptosis can be inhibited through the GCH1-BH4 pathway [24, 25]. The major regulators of ferroptosis and associated mechanisms are summarized (Fig. 1).

Fig. 1.

Pathways of ferroptosis. Ferroptosis can be triggered through inhibiting the system Xc-/GSH/GPX4 axis or the accumulation of Fe2+ and lipid peroxidation. Blocking or inhibiting the Xc-/GSH/GPX4 axis can result in decreased intracellular cysteine levels and suppressing the lipid repair function of GPX4. Consequently, disruption of the antioxidant capacity of cells contributes to the initiation and progression of ferroptosis. In lipid metabolism, the most important characteristics of ferroptosis are the increase of intracellular iron ion concentration and the abnormal accumulation of lipid ROS. FSP1 can negatively regulate ferroptosis through CoQ10, thereby inhibiting the delivery of lipid peroxides. Ferritinophagy plays a crucial role in the association between ferritinophagy and ferroptosis. Intracellular ferritin is transported to autophagy lysosomes for degradation, leading to the release of free iron and ultimately triggering ferroptosis. Abbreviations: GPX4, glutathione peroxidase 4; PUFAs, polyunsaturated fatty acids; ACSL4, acyl-CoA synthetase long chain family member 4; LPCAT3, lysophosphatidylcholine acyltransferase 3; PL-OO, phospholipid peroxyl radical; TCA, tricarboxylic acid; GSH, glutathione; ROS, reactive oxygen species; FSP1, ferroptosis suppressor protein 1; CoQ10, coenzyme Q10; GCH1, GTP cyclohydrolase 1. ATP, adenosine triphosphate.

3. Ferroptosis and Other Models of Programmed Cell Death
3.1 Ferroptosis and Necroptosis

Necroptosis is a programmed form of cell death that can regulate necrosis with passive and active proinflammatory functions [26]. Numerous studies have consistently shown that ferroptosis is invariably accompanied by necroptosis [27]. The key steps involved in necroptosis include tumor necrosis factor (TNF) signaling, deubiquitination of receptor-interacting protein (RIP) 1, phosphorylation of RIP1 and RIP3, inactivation of caspase-8, and phosphorylation of mixed lineage kinase domain-like protein (MLKL) [28]. Ferroptosis can induce mitochondrial damage, resulting in the opening of the mitochondrial permeability transition pore. Subsequently, the phosphorylation of RIP1/3 was further intensified, ultimately culminating in necroptosis [29]. These findings demonstrate that there is a crosstalk between ferroptosis and necroptosis, but the regulatory relationship between necroptosis and ferroptosis still requires further study.

3.2 Ferroptosis and Autophagy

Autophagy refers to the process where the detached double membrane of the ribosome-free attachment zone of the rough endoplasmic reticulum envelops cytoplasm, organelles, proteins, and other cellular components that require degradation to form autophagosomes [30]. Subsequently, the autophagosomes fuse with lysosomes to form autophagolysosomes, wherein the enclosed contents are degraded to fulfill the metabolic requirements and facilitate organelle renewal. A growing body of evidence suggests that ferroptosis relies on the autophagy machinery for its execution, making it a form of autophagy-dependent cell death [31]. Recent studies have discovered that autophagy plays a role in promoting ferroptosis by degrading the ferroptosis-related protein, ferritin, through the autophagy-related gene (ATG) 5/ATG7-nuclear receptor coactivator 4 (NCOA4) autophagic pathway [32]. Moreover, ras-related protein Rab-7a (RAB7A)-mediated lipophagy, BECN1-mediated system Xc- inhibition, signal transducer and activator of transcription 3 (STAT3)-mediated lysosomal cell death, and sequestosome 1 (SQSTM1)-mediated clockophagy are involved in the regulation of ferroptosis [33]. All of these studies highlight the regulatory role of autophagy in ferroptosis, presenting a promising new treatment hypothesis by targeting autophagic ferroptosis.

3.3 Ferroptosis and Apoptosis

Apoptosis is a well-established mode of programmed cell death involving both extracellular and intracellular pathways [34]. The extracellular pathway is triggered by receptors on the cell membrane, such as TNF-α, while the intracellular pathway is primarily influenced by the permeability of the mitochondrial membrane and the regulation of the Bcl-2 protein family. Indeed, several studies have reported an interrelationship between ferroptosis and apoptosis that apoptosis can be converted to ferroptosis [35]. In addition, the ferroptosis inducer, erastin, activates the p53-dependent CHOP/PUMA pathway, enhancing sensitivity to apoptosis [36]. These findings indicate the existence of close link between ferroptosis and apoptosis.

3.4 Ferroptosis and Pyroptosis

Pyroptosis, also known as cell inflammatory necrosis, is a form of programmed cell death that relies on inflammatory caspases [37]. Pyroptosis is characterized by nuclear pyknosis, cell swelling, the formation of lipid membrane vacuoles at the plasma membrane, and eventual cell rupture without DNA fragmentation. Both pyroptosis and ferroptosis are associated with damage to the cytoplasmic membrane. Notably, ROS-mediated damage to the cytoplasmic membrane may contribute to reciprocal regulation between pyroptosis and ferroptosis. Furthermore, research has revealed that both types of cell death can be triggered by elevated levels of intracellular iron and ROS [38]. The aforementioned study further demonstrates that a synergistic relationship and crosstalk is existed between pyroptosis and ferroptosis, but the underlying mechanisms still need to be thoroughly investigated.

4. Relationship between Ferroptosis and NAFLD

The liver, an important organ regulating lipid balance, is critical for the metabolism of lipids. Meanwhile, the liver also maintains iron homeostasis in the body and constitutes the main organ that stores excess iron and synthesizes hepcidin. In case of an imbalance between energy intake and consumption or abnormal energy storage function in adipocytes, the liver is most inclined to accumulate fat leading to hepatic steatosis, which subsequently causes systemic metabolic disorders [39, 40]. In this regard, the disruption of iron metabolism is correlated with obesity and insulin resistance (IR) in patients with NAFLD. Furthermore, disturbed iron metabolism has close correlations with the typical features of obesity and pancreatic resistance in NAFLD patients [41]. The “two-hit” and “multiple-hit” hypotheses are classic theoretical explanations for NAFLD pathogenesis [42]. Major risk factors for NAFLD are considered to include IR, altered lipid metabolism, ER stress, mitochondrial dysfunction, lipid toxicity, inflammatory cascade, genetic susceptibility, epigenetic changes and disturbed intestinal flora [43]. Thus, LPO may aggravate ferroptosis. Recently, ferroptosis was suggested to play a critical role by effectively promoting and inhibiting ferroptosis-related factors in NAFLD and NASH mice [44, 45]. In NAFLD, this is mainly associated with the LPO reaction triggered by iron overload and oxidative stress.

4.1 Iron Overload

Iron participates in multiple cellular processes, including oxygen storage and transport, mitochondrial respiration, DNA replication, and intercellular signaling in the body [46]. Intracellular iron balance considers iron absorption, output, use and storage. Excess iron is mostly stored in ferritin, as a redox inactive form. Generally, Fe3+ is bound to transferrin and transferrin receptor 1 (Tfr1) in cells and stored in the form of Fe3+-ferritin complex [47]. Subsequently, hepcidin synthesized in the liver is important in maintaining iron homeostasis by controlling ferritin synthesis. A small quantity of Fe2+ generate a labile iron pool [48]. Iron metabolism disorders are mainly divided into two categories, including iron overload and deficiency. In case of iron overload in cells, Fe3+ is reduced to Fe2+ in lysosomes and released into the labile iron pool through the divalent metal transporter 1. Free Fe2+, with high oxidative potential, easily undergoes the Fenton reaction with lipid peroxides, produces hydroxyl radicals, induces oxidative stress, enhances ROS accumulation and induces ferroptosis [49]. Fe2+ is thus also considered a cofactor increasing the activities of multiple metabolic enzymes, promoting lipid ROS synthesis and enhancing ferroptosis. In addition, nuclear receptor coactivator 4 (NCOA4) can mediate ferritinophagy, which dissociates the Fe3+-ferritin complex to release Fe2+ into LIP [50, 51]. Iron metabolism disorder may therefore induce the production of large amounts of Fe2+, which is involved in the conversion of PUFAs through the LPO process, aggravating NAFLD, diabetes, IR and other diseases. In conclusion, iron is a crucial factor in ferroptosis, which requires iron metabolism.

Hepatocytes are critical for liver iron homeostasis. Iron shortage in hepatocytes and excessive iron accumulation in hepatic stellate cells mediated by Kupffer cells-induced secretion of iron-rich extracellular vesicles (EVs), can lead to NAFLD and NASH. Meanwhile, blocking EV secretion or EV iron transport can restore iron balance in the liver and alleviate NAFLD/NASH-associated hepatic steatosis and fibrosis. A previous study revealed that the lack of hepcidin inhibitor matriptase-2 may alleviate obesity, accelerate lipolysis, improve glucose tolerance and insulin sensitivity, and prevent hepatic steatosis in high-fat diet mice [52]. This suggests that regulation of iron homeostasis in the liver has a significant correlation with anti-steatosis.

4.2 Oxidative Stress

Oxidative stress describes an imbalance between oxidative and antioxidant events, where the generation of free radicals exceeds the capacity of antioxidant defense systems. Iron overload promotes the production of ROS, thereby inducing oxidative stress. Furthermore, oxidative stress-related transcription factors downregulate or upregulate ferroptosis-related genes, which regulates ferroptosis [53, 54, 55]. The “multiple-hit” hypothesis considers oxidative stress one of the key mechanisms underlying NAFLD-related liver injury. When ROS are excessively produced through the external environment (e.g., physical and chemical factors) and the internal environment (e.g., mitochondrial dysfunction, ER stress and peroxidation), they target the double bonds of PUFAs, resulting in LPO and the formation of lipid peroxides, which induces ferroptosis and aggravates hepatocyte injury. Recent evidence suggests mitochondrial ROS play a role in NASH-related ferroptosis. Epigallocatechin gallate prevents liver lipotoxicity by suppressing mitochondrial ROS-related liver ferroptosis [41]. Quercetin reduces lipid accumulation, hepatic lipotoxicity and LPO in high-fat diet mice, ultimately inhibiting ferroptosis to alleviate NAFLD [56]. Besides, obeticholic acid reduced the amounts of triglyceride-containing PUFAs, while elevating free PUFAs and phosphatidylethanolamine-containing PUFAs, which are prone to oxidization to generate lipid peroxides, triggering ferroptosis in hepatocytes and causing hepatic stellate cell activation [57]. Taken together, both oxidative stress and iron overload can induce the LPO process, and even synergically worsen the degree of cell damage, resulting in NAFLD progression from simple hepatic steatosis to inflammation and then fibrosis.

5. Associations of Ferroptosis with the Main Regulatory Factors of NAFLD
5.1 Glutathione

GSH is the main antioxidant in mammalian cells, which is composed of glutamate, cysteine and glycine, and represents a cofactor of GPX4 [58]. However, GSH depletion disrupts the dynamic balance of intracellular redox activity, which leads to ROS accumulation and aggravates ferroptosis [59, 60]. Cysteine is the main component and thus the limiting amino acid in GSH biosynthesis; suppressing cysteine import via the cystine/glutamate antiporter SLC7A11 induces ferroptosis via GSH depletion [61]. Therefore, GSH is often used as an evaluation indicator of oxidative stress, especially in studies developing drug therapies for ferroptosis-induced NAFLD.

5.2 Glutathione Peroxidase 4

GPX4 was the first discovered selenium-containing protein inhibiting ferroptosis. It is mainly found in the cytoplasm and mitochondria, and plays a role in scavenging lipid hydrogen peroxide. GPX4 can convert toxic lipid hydroperoxides into corresponding nontoxic lipid alcohols, which forms a defense mechanism against LPO and plays an important regulatory role in ferroptosis [62]. In methionine and choline deficient L-amino acid diet (MCD)-fed NAFLD mice treated with the ferroptosis inducer (1S,3R)-RSL3, liver GPX4 was downregulated while it was upregulated by lipoxygenase and an apoptosis inducer. Subsequently, supplementation of the GPX4 activator sodium nitrite increased GPX4 expression and reduced the severity of NASH. These results indicated that GPX4 plays a significant role in ferroptosis-related changes in hepatocytes that cause NASH [63]. Therefore, GPX4 regulation may control the development of NAFLD inflammation before it progresses to NASH. NAFLD rats were treated with ferroptosis inducer (erastin) and inhibitor (ferrostatin 1 [Fer-1]), and steatosis in human liver cell 7702 (L02) was induced by palmitic acid (PA), respectively. Both in cultured cells and experimental animals, interfered GPX4 expression decreased the protective effects on rat liver and mitochondrial membrane integrity in L02 cells, and also regulated B-cell lymphoma-2 (Bcl-2), Bcl-2 associated x (Bax), Caspase-3 and superoxide dismutase 1 (SOD1), attenuating the treatment effect in NAFLD. This study revealed that suppressing GPX4-related ferroptosis may provide a novel approach for NAFLD treatment [64].

5.3 Xc-system

The Xc- system is known as the cystine/glutamate transporter on the cell membrane; its light chain subunit SLC7A11 is critical for the metabolism of lipid ROS. Recent data revealed p53 downregulates SLC7A11 to markedly affect the metabolism of lipid ROS in ferroptosis [65, 66]. High SLC7A11 expression promoted tumor growth partially by inhibiting ferroptosis. Nevertheless, malignant cells highly expressing SLC7A11 also endure the substantial burden imposed by SLC7A11-indcued metabolic reprogramming, resulting in enhanced dependence on glucose and glutamine, which represents possible metabolic vulnerabilities for target therapies in malignant diseases [67]. Sorafenib attenuates liver damage and extracellular matrix accumulation in CCl4-dependent liver fibrosis, with concomitant SLC7A11 and GPX4 downregulation at the protein level. SLC7A11 is upregulated by hypoxia inducible factor (HIF)-1α, and HIF-1α/SLC7A11 pathway suppression is essential in sorafenib-related ferroptosis in hepatic stellate cells, ultimately limiting antifibrotic activity [68]. Furthermore, BECN1 combined with SLC7A11 can directly block the activity of the Xc- system, indicating that SLC7A11 is a core component of the transporter [69]. As an inducer of Nrf2, SLC7A11 accelerates the transport of cystine and glutamate, promotes the synthesis of GSH, and thus achieves the purpose of suppressing ferroptosis [70, 71].

5.4 Nuclear Factor Erythroid 2-Related Factor 2

Nrf2 interacts with nuclear factors to activate downstream transcription, which regulates antioxidative events and detoxification [72]. Nrf2 regulates multiple biological events, e.g., the transcription of the antioxidant system, the detoxification of endogenous and exogenous substances, NADPH regeneration and heme metabolism, as well as many cell processes, including autophagy and unfolded protein response [73]. Mounting evidence suggests a regulatory role for Nrf2 in ferroptosis. Nrf2 constitutes an upstream effector of some essential ferroptosis-related genes such as GPX4 and Xc- system [74]. Consequently, Nrf2 negatively regulates ferroptosis and promotes ferroptosis resistance. After Kelch-like ECH-associated protein 1 (KEAP1) activates Nrf2 in oxidative stress, it recognizes antioxidant response elements and activates antioxidant genes, including SLC7A11. Furthermore, it upregulates antioxidant proteins associated with ferroptosis, e.g., heme oxygenase-1 (HO-1) and GPX4, and increases GSH levels [75, 76]. A recent study reported Nrf2 activation by iron-dependent, mitochondrial pro-oxidants, which upregulates bone morphogenetic protein 6 (Bmp6) in hepatic sinusoidal endothelial cells and increases the synthesis of hepcidin in adjacent hepatocytes. In Nrf2-deficient mice, impaired Bmp6-hepcidin axis was reported, causing iron accumulation and worsened liver damage. Meanwhile, pharmacological induction of Nrf2 resulted in activated Bmp6-hepcidin signaling, ameliorating iron homeostasis and countering Bmp6 suppression [77]. Overall, Nrf2 links cellular sensitivity to iron toxicity, systemic iron homeostasis and antioxidation, representing a potential target for treating iron-related diseases.

Besides, the Nrf2 contributes to reducing ROS overproduction to exert anti-inflammatory effects for NAFLD. Indeed, a small interfering RNA (siRNA) of Nrf2 or a glutathione peroxidase suppressor could abolish nuclear factor kappa-B (NF-κB) repression and decrease the secretion of inflammatory cytokines [78]. As shown in Fig. 2, downstream effectors of Nrf2, such as HO-1, GSH and GPX4, were upregulated to reduce MDA, inhibit ROS accumulation, LPO and reduce high-fat diet-induced NAFLD [79].

Fig. 2.

KEAP1-Nrf2 pathway. When cells experience oxidative stress, the KEAP1-Nrf2 interaction is altered, preventing KEAP1 from targeting Nrf2 for degradation. As a result, Nrf2 is stabilized and accumulates in the cell. One of the key mechanisms by which Nrf2 contributes to protecting cells from ferroptosis is through the upregulation of genes involved in iron metabolism. Nrf2 induces the expression of GPX4, HO-1, and SLC7A11, releasing iron in a controlled manner. By regulating iron levels, Nrf2 helps prevent the accumulation of toxic levels of labile iron that can catalyze lipid peroxidation, a hallmark of ferroptosis. Abbreviations: KEAP1, Kelch-like ECH-associated protein 1; GPX4, glutathione peroxidase 4; HO-1, heme oxygenase-1; SLC7A11, subunit solute carrier family 7 member 11; Nrf2, nuclear factor erythroid 2-related factor 2.

5.5 Ferroptosis Suppressor Protein 1

In NAFLD, liver inflammation is a critical component of disease progression. FSP1 has been associated with the regulation of inflammatory responses in various cell types. Its involvement in immune cell migration and activation may influence the extent of liver inflammation in NAFLD [18]. FSP1 was initially identified as the homologous regulation factor of the mitochondrial pro-apoptotic protein apoptosis-inducing factor (AIF)/AIF mitochondrion-associated 1 (AIFM1), termed AIFM2. However, it was later confirmed that FSP1 lacks the N-terminal sequence for mitochondrial targeting, is not localized in mitochondria and does not promote apoptosis. It was then renamed by Bersuker et al. [23] as FSP1 and its cellular function was described. FSP1 is present in lipid droplets and cell membranes, and it acts as a suppressor of ferroptosis by reducing lipid peroxidation and protecting cells from oxidative damage. Mechanically, CoQ10 was identified as a major substrate using a ferroptosis activator [80]. FSP1 expression shows a positive correlation with resistance to GPX4 suppressors, and plays a critical role in maintaining cancer cell proliferation in case of no GPX4 expression. Notably, similar to FSP1, GCH1 was recently described as a new protein suppressing ferroptosis based on whole-genome activation screening [81]. GCH1 overexpression occurs in presence of all three ferroptosis activators, including RSL3, erastin and GPX4. High GCH1 amounts enhance BH4 and CoQ10 levels, synergistically acting with the FSP1 pathway to hamper the LPO process. GCH1 expression and resistance to ferroptosis are overtly correlated, suggesting the GCH1-BH4-phospholipid pathway as a potential therapeutic target in iron-related diseases.

6. Iron Metabolism Regulatory Factors

Iron metabolism regulatory factors are essential proteins and molecules that play key roles in maintaining the balance of iron levels within cells and tissues. These factors are also known to have a close implications in NAFLD. Understanding its roles in NAFLD, such as Tfr1 and Ferritin, is crucial for elucidating the mechanisms underlying the disease and potentially identifying new therapeutic targets. They are tightly regulated by various iron metabolism regulatory factors that cells have access to adequate iron for their functions while preventing iron overload, which can lead to cellular damage and contribute to the development of iron-related NAFLD.

6.1 Transferrin Receptor 1

Transferrin receptor 1 (Tfr1), or cluster of differentiation 71 (CD71), interacts with transferrin and exerts an important effect on iron uptake by hepatocytes in a specific manner [82]. In palmitate-induced IR, palmitate simultaneously upregulates Tfr1 and induces intracellular ferroptosis. Knockdown of Tfr1 prevented palmitate-induced iron uptake and IR, and also translocated Tfr1 via enhancement of calcium influx. However, supplementation of a calcium chelator substantially decreased ferroptosis by suppressing Tfr1 translocation, and ultimately enhanced insulin sensitivity [83]. It was also found that the interaction between complexes was neutralized by antibodies against Tfr1, making Tfr1 a potential target for ferroptosis alleviation in NAFLD [84].

6.2 Ferritin

Excess iron in the body is stored in ferritin that plays a critical role in iron metabolism and storage in cells. It is usually involved in ferritinophagy which releases Fe3+ and is subsequently converted to Fe2+ to the labile iron pool, participating in the process of cell ferroptosis. Eventually, it sequesters excess iron in a non-toxic and bioavailable form to prevent the free iron from participating in harmful Fenton reactions that generate highly reactive and toxic hydroxyl radicals [85]. Therefore, suppressing ferritinophagy may provide a novel approach and target for treating NAFLD [86]. Elevated Ferritin levels are commonly observed in patients with NAFLD. It is important to note that the relationship between Ferritin and NAFLD is complex and multifactorial, involving iron metabolism, oxidative stress, and inflammation. It was reported that prominin-2, a pentapeptide protein associated with lipid homeostasis, is induced by ferroptosis response and induces the generation of ferritin-containing multivesicular bodies (MVBs), which transports iron out of cells to suppress ferroptosis. This suggests the prominin-2/MVBs-ferritin pathway has certain significance in iron homeostasis and intracellular transport [87]. Therefore, further investigation is needed to study how to inhibit ferroptosis-induced hepatic steatosis from the perspective of iron metabolism.

7. Ferritinophagy-Mediated Ferroptosis

Recently, ferroptosis has attracted increasing attention among investigators studying liver pathologies and was shown to involve the regulation of NCOA4-mediated ferritinophagy [88]. There is a feedback mechanism behind intracellular iron utilization in which NCOA4-mediated ferritinophagy degrades ferritin, and its activation increases the amounts of available iron within the cell (Fig. 3). In terms of mechanism, the selective autophagy receptor NCOA4 interacts with ferritin heavy chain 1 (FTH1) and mediates ferritin transfer to autolysosomes, eventually releasing free iron. Such regulatory effect is not unidirectional, as intracellular iron levels also affect the flux of ferritinophagy [89]. In case of high iron content, HECT and RLD domain containing E3 ubiquitin protein ligase 2 (HERC2), an enzyme 3 (E3) ubiquitin ligase, drives ubiquitin-dependent degradation of NCOA4. This process reduces the ferritinophagy flux, thereby maintaining iron homeostasis [90]. It is noteworthy that NCOA4 level is a central determinant of the ferritinophagy flux. In the non-disease state, ferritinophagy maintains cellular iron balance, but its overactivation can cause iron overload within cells. LPO, which is correlated with iron overload, is an important factor required for ferroptosis [89]. The above findings jointly suggest a close relation between ferritinophagy and ferroptosis. A study confirmed GPX4 suppresses ferroptosis and shows elevated expression in NCOA4-knockout mice, revealing an association of ferroptosis with ferritinophagy [91]. Additionally, several studies have suggested that ferritinophagy promotes ferroptosis, primarily because of iron overload resulting from NCOA4 upregulation. Sorafenib is an iron-dependent inducer of ferroptosis, whose potential mechanism involves increasing NCOA4 expression to accelerate iron overload and ROS generation [92]. Moreover, elevated iron content induced by NCOA4 overexpression increases sensitivity to ferroptosis. Conversely, ferritinophagy protects from ferroptosis. In case of NCOA4 deficiency or obstructed NCOA4 binding to FTH1, ferritinophagy is hindered, which may decrease sensitivity to ferroptosis within cells [32]. Furthermore, NCOA4 knockout blocks sideroflexin 1-mediated mitochondrial iron overload, thereby inhibiting ferroptosis [93]. Taken together, these data indicate iron overload represents the initial step of ferritinophagy-mediated ferroptosis. The major factor in ferritinophagy regulation is to influence its core molecule NCOA4 by controlling its expression or preventing its interaction with ferritin.

Fig. 3.

Mechanism of ferritinophagy. NCOA4-mediated ferritinophagy is a selective autophagic process. NCOA4 acts as a selective cargo receptor, specifically recognizing ferritin and targeting it to autophagosomes for degradation in lysosomes. Through ferritinophagy, intracellular ferritin is broken down, releasing free iron, which can contribute to the initiation of ferroptosis. Abbreviations: NCOA4, nuclear receptor coactivator 4.

Overall, NCOA4-dependent ferritinophagy exerts a crucial regulatory effect on ferroptosis by modulating iron homeostasis in cells and controlling ROS biosynthesis. Further research is necessary to investigate the pathways by which ferritinophagy modulates ferroptosis and to develop drugs that may target ferritinophagy as a therapeutic strategy to ameliorate the pathological processes associated with ferroptosis in various diseases.

8. Pharmacologic Regulation of Ferroptosis and Ferritinophagy in NAFLD and Other Diseases

Agents selectively regulating ferroptosis (inhibitors or inducers) are crucial for the mechanistic studies of ferroptosis-dependent NAFLD and other disorders. Table 1 (Ref. [23, 45, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105]) and Table 2 (Ref. [67, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126]) list the best-studied and most widely-applied inhibitors and inducers of ferroptosis, respectively.

Table 1.Major inhibitors of ferroptosis in NAFLD and other diseases.
Compound Possible mechanisms Functions and diseases
Deferiprone As an iron chelating agent, has affinity with Fe3+ Alleviates NASH and fibrosis [45]
Ferrostatin-1 Prevents lipid ROS accumulation and inhibits LPO, constituting a lipid ROS scavenger Alleviates NAFLD/NASH [94]
Ginkgolide B Promotes GPX4 and FTH1 expression, and reduces TFR1 expression, ROS levels, and intracellular iron content Improves fibrosis and diabetic nephropathy [95]
Trolox Damps lipoxygenase and abrogates hydroxyl group radicals, with potent antioxidant properties Revealed an association between blood redox status and the likelihood of NAFLD [96]
Liproxstatin-1 Functions as a radical-trapping antioxidant to inhibit lipid peroxidation Ameliorates NAFLD and acute renal failure [45, 97, 98]
Selenium Increases the expression of GPX4 Affects the progression of NASH [99]
CoQ10 Acts as a lipophilic radical-trapping antioxidant to prevent the generation of lipid peroxides Exerts hepatoprotective effects and is beneficial in NAFLD; sensitizes malignant cells to ferroptosis-activating chemotherapeutics [23, 100]
Baicalein Targets lipoxygenases and suppresses lipoxygenase-triggered LPO Exerts pharmacological effects in bladder cancer, vitiligo and ischemia-reperfusion injury [101, 102, 103]
XJB-5-131 Exerts antioxidant effects and suppresses LPO Improves ischemia-reperfusion injury and acute kidney injury [104, 105]

NASH, nonalcoholic steatohepatitis; NAFLD, nonalcoholic fatty liver disease; LPO, lipid peroxidation; CoQ10, coenzyme Q10; GPX4, glutathione peroxidase 4; FTH1, ferritin heavy chain 1.

Table 2.Major inducers of ferroptosis in NAFLD and other diseases.
Compound Possible mechanisms Functions and diseases
RSL-3 Suppresses GPX4 and induces the accumulation of lipid hydroperoxides Aggravates gastric cancer, hepatocellular carcinoma, triple-negative breast cancer, and colorectal cancer [106, 107, 108, 109]
High-fat diet Promotes iron overload and oxidative stress, and inhibits the expression of GPX4 and SLC7A11 Aggravates NASH [110]
Erastin Targets the Xc- system and decreases GSH Weakens therapeutic effects in NAFLD and enhances the growth of tumors, e.g., melanoma and triple negative breast cancer [111, 112]
Sorafenib Suppresses cystine uptake through the Xc- system and decreases GSH Ameliorates hepatocellular carcinoma and murine liver fibrosis [67, 113]
Sulfasalazine Suppresses cystine uptake through the Xc- system and decreases GSH Improves mesenchymal traits and disabled antioxidant program in head and neck cancer, hepatocellular carcinoma and pancreatic cancer [114, 115]
Glutamate High concentrations of glutamate inhibit cystine uptake by the Xc- system Improves non-small cell lung cancer [116]
ML-162 Suppresses GPX4 and causes disorders of lipid hydroperoxides Improves head and neck cancer [117]
FIN 56 Decreases GPX4 expression, activates squalene synthase and suppresses CoQ10 Exerts anticancer effects in human fibrosarcoma and head and neck cancer [117, 118]
Dihydro-artemisinin Decreases GPX4 expression Increases sensitivity to ferroptosis in cancer cells [119]
Ferric ammonium citrate Causes iron overload and LPO Protects against iron overload-associated knee osteoarthritis and osteoblast differentiation [120, 121]
Trigonelline Inhibits Nrf2 Reduces blood glucose and kidney injury in diabetes, and renders cancer cells more susceptible to apoptosis [122, 123]
FINO(2) Suppresses GPX4-related enzymatic activity and oxidizes iron, and causes LPO Exerts anticancer effects in human fibrosarcoma [124]
Brequinar Inhibits DHODH and thus decreases ubiquinone reduction to ubiquinol Suppresses renal, cervical and nasopharyngeal cancer growth [125, 126]

RSL-3, (1S,3R)-RSL3; SLC7A11, subunit solute carrier family 7 member 11; DHODH, dihydroorotate dehydrogenase.

Ferroptosis plays a crucial role in liver pathophysiology and is also a key factor in initiating the progression from NAFLD to NASH. Therefore, it holds great promise as a potential therapeutic target for preventing the development of NAFLD. While the existing data strongly suggest that targeted ferroptosis could be an excellent approach for treating NAFLD, it is essential to acknowledge the current gaps in research on this topic. To facilitate its understanding and potential applications, certain issues should be addressed in the future. These mainly include: (1) elucidating the mechanisms: further researches are needed to elucidate deeper and detailed molecular mechanism that are aiming to pave the way for more effective and targeted therapeutic interventions; (2) identifying biomarkers: in the context of NAFLD, the identification of reliable biomarkers associated with ferroptosis in NAFLD is essential for early detection and diagnosis; (3) addressing specific subtypes: investigating how ferroptosis contributes to the progression of different NAFLD subtypes will aid in tailoring treatments to specific patient populations; (4) investigating combination therapies: considering wide spectrum in NAFLD, exploring the combinations involving ferroptosis-targeting agents along with other existing treatments may offer a more comprehensive approach to management. By addressing the aforementioned gaps, we may have the opportunity to fully unleash the potential of targeted ferroptosis as a powerful strategy for effectively preventing and treating NAFLD.

9. Conclusions

Ferroptosis represents a newly discovered form of cell death associated with iron-dependent LPO. Currently, studies exploring ferroptosis mechanistically in human health are mainly focused on cancer, and based on reported mechanisms, new targets for developing anti-cancer drugs have been discovered. Summarizing recent studies on the pathways and defense mechanisms associated with ferroptosis, accumulating evidence indicates a certain correlation among the pathological processes of NAFLD such as lipid accumulation, hepatic steatosis, inflammatory cell infiltration, fibrosis, mitochondrial damage and ferroptosis. Iron overload and oxidative stress-induced LPO may mediate the progression of NAFLD, and the main antioxidant mechanism and regulatory factors controlling iron metabolism may become potential targets for NAFLD-related research. Among them, targeting Nrf2 activation can cause SLC7A11 to accelerate the transport of cysteine and glutamate through the Xc- system, promoting GSH synthesis. GSH, the most important endogenous antioxidant in the human body, is involved in the downregulation of GPX4, which suppresses LPO and prevents ferroptosis.

With multiple potential targets and overarching approaches for regulating ferroptosis emerging from recent reports, ferritinophagy intervention could pave a novel way for ferroptosis regulation. Ferritinophagy-mediated ferroptosis is a potential target for preventing the onset of NAFLD. Therefore, it is necessary to also take ferritinophagy into account in strategies for NAFLD therapy. The concept of ferroptosis has enormous research potential and clinical value in the field of NAFLD. This review provides new insights into ferroptosis relevant to the pathogenesis of NAFLD. However, more detailed mechanism of action for ferroptosis in NAFLD still needs further investigation.

Abbreviations

4-HNE, 4-hydroxynonenal; ACSL4, acyl-CoA synthetase long chain family member 4; Bmp6, bone morphogenetic protein 6; CD71, cluster of differentiation 71; CoQ10, coenzyme Q10; ECM, extracellular matrix; ER, endoplasmic reticulum; ETC, electron transport chain; EVs, extracellular vesicles; Fer-1, ferrostatin 1; FSP1, ferroptosis suppressor protein 1; FTH1, ferritin heavy chain 1; GCH1, GTP cyclohydrolase 1; HO-1, heme oxygenase-1; IR, insulin resistance; GSH, glutathione; KEAP1, Kelch-like ECH-associated protein 1; LPCAT3, lysophosphatidylcholine acyltransferase 3; LPO, lipid peroxidation; MDA, malondialdehyde; MVBs, multivesicular bodies; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; NCOA4, nuclear receptor coactivator 4; Nrf2, nuclear factor erythroid 2-related factor 2; PA, palmitic acid; PL, phospholipid radical; PUFAs, polyunsaturated fatty acids; ROS, reactive oxygen species; TCA; tricarboxylic acid; Tfr1, transferrin receptor 1.

Author Contributions

SNZ and XZY designed the study. SNZ, YG and XZY collected and analyzed the literatures. SNZ and XZY wrote the manuscript. YG and XZY discussed and critically revised the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This work was supported by the Science and Technology Development Plan of Jilin Province (20210204029YY and YDZJ202201ZYTS151).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Quek J, Chan KE, Wong ZY, Tan C, Tan B, Lim WH, et al. Global prevalence of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in the overweight and obese population: a systematic review and meta-analysis. The Lancet Gastroenterology & Hepatology. 2023; 8: 20–30.
[2]
Powell EE, Wong VWS, Rinella M. Non-alcoholic fatty liver disease. Lancet. 2021; 397: 2212–2224.
[3]
Estes C, Anstee QM, Arias-Loste MT, Bantel H, Bellentani S, Caballeria J, et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016-2030. Journal of Hepatology. 2018; 69: 896–904.
[4]
Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nature Medicine. 2018; 24: 908–922.
[5]
Yang L, Zhou MT, Lu XC, Zhu HY, Han HM. Research Progress on the Mechanism of Vitamin D3 in the Occurrence and Development of Nonalcoholic Fatty Liver Disease. Journal of Biological Regulators and Homeostatic Agents. 2022; 36: 797–805.
[6]
Sheka AC, Adeyi O, Thompson J, Hameed B, Crawford PA, Ikramuddin S. Nonalcoholic Steatohepatitis: A Review. JAMA. 2020; 323: 1175–1183.
[7]
Raza S, Rajak S, Upadhyay A, Tewari A, Sinha RA. Current treatment paradigms and emerging therapies for NAFLD/NASH. Frontiers in Bioscience (Landmark Edition). 2021; 26: 206–237.
[8]
Gautheron J, Gores GJ, Rodrigues CMP. Lytic cell death in metabolic liver disease. Journal of Hepatology. 2020; 73: 394–408.
[9]
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012; 149: 1060–1072.
[10]
Yan HF, Zou T, Tuo QZ, Xu S, Li H, Belaidi AA, et al. Ferroptosis: mechanisms and links with diseases. Signal Transduction and Targeted Therapy. 2021; 6: 49.
[11]
Wang S, Liu Z, Geng J, Li L, Feng X. An overview of ferroptosis in non-alcoholic fatty liver disease. Biomedicine & Pharmacotherapy. 2022; 153: 113374.
[12]
Yang WS, Stockwell BR. Ferroptosis: Death by Lipid Peroxidation. Trends in Cell Biology. 2016; 26: 165–176.
[13]
Wu ST, Chen JY, Martin V, Ng R, Zhang Y, Grover D, et al. Valence opponency in peripheral olfactory processing. Proceedings of the National Academy of Sciences of the United States of America. 2022; 119: e2120134119.
[14]
Zhao C, Yu D, He Z, Bao L, Feng L, Chen L, et al. Endoplasmic reticulum stress-mediated autophagy activation is involved in cadmium-induced ferroptosis of renal tubular epithelial cells. Free Radical Biology & Medicine. 2021; 175: 236–248.
[15]
Liu J, Liu Y, Wang Y, Li C, Xie Y, Klionsky DJ, et al. TMEM164 is a new determinant of autophagy-dependent ferroptosis. Autophagy. 2023; 19: 945–956.
[16]
Liu L, He J, Sun G, Huang N, Bian Z, Xu C, et al. The N6-methyladenosine modification enhances ferroptosis resistance through inhibiting SLC7A11 mRNA deadenylation in hepatoblastoma. Clinical and Translational Medicine. 2022; 12: e778.
[17]
Liang D, Minikes AM, Jiang X. Ferroptosis at the intersection of lipid metabolism and cellular signaling. Molecular Cell. 2022; 82: 2215–2227.
[18]
Zhang S, Sun Z, Jiang X, Lu Z, Ding L, Li C, et al. Ferroptosis increases obesity: Crosstalk between adipocytes and the neuroimmune system. Frontiers in Immunology. 2022; 13: 1049936.
[19]
Bathish B, Robertson H, Dillon JF, Dinkova-Kostova AT, Hayes JD. Nonalcoholic steatohepatitis and mechanisms by which it is ameliorated by activation of the CNC-bZIP transcription factor Nrf2. Free Radical Biology & Medicine. 2022; 188: 221–261.
[20]
Yang Y, Chen J, Gao Q, Shan X, Wang J, Lv Z. Study on the attenuated effect of Ginkgolide B on ferroptosis in high fat diet induced nonalcoholic fatty liver disease. Toxicology. 2020; 445: 152599.
[21]
Liu P, Anandhan A, Chen J, Shakya A, Dodson M, Ooi A, et al. Decreased autophagosome biogenesis, reduced NRF2, and enhanced ferroptotic cell death are underlying molecular mechanisms of non-alcoholic fatty liver disease. Redox Biology. 2023; 59: 102570.
[22]
Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019; 575: 693–698.
[23]
Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019; 575: 688–692.
[24]
Hu Q, Wei W, Wu D, Huang F, Li M, Li W, et al. Blockade of GCH1/BH4 Axis Activates Ferritinophagy to Mitigate the Resistance of Colorectal Cancer to Erastin-Induced Ferroptosis. Frontiers in Cell and Developmental Biology. 2022; 10: 810327.
[25]
Wang D, Liang W, Huo D, Wang H, Wang Y, Cong C, et al. SPY1 inhibits neuronal ferroptosis in amyotrophic lateral sclerosis by reducing lipid peroxidation through regulation of GCH1 and TFR1. Cell Death and Differentiation. 2023; 30: 369–382.
[26]
Bertheloot D, Latz E, Franklin BS. Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cellular & Molecular Immunology. 2021; 18: 1106–1121.
[27]
Zille M, Karuppagounder SS, Chen Y, Gough PJ, Bertin J, Finger J, et al. Neuronal Death After Hemorrhagic Stroke In Vitro and In Vivo Shares Features of Ferroptosis and Necroptosis. Stroke. 2017; 48: 1033–1043.
[28]
Li Q, Weiland A, Chen X, Lan X, Han X, Durham F, et al. Ultrastructural Characteristics of Neuronal Death and White Matter Injury in Mouse Brain Tissues After Intracerebral Hemorrhage: Coexistence of Ferroptosis, Autophagy, and Necrosis. Frontiers in Neurology. 2018; 9: 581.
[29]
Zhou Y, Liao J, Mei Z, Liu X, Ge J. Insight into Crosstalk between Ferroptosis and Necroptosis: Novel Therapeutics in Ischemic Stroke. Oxidative Medicine and Cellular Longevity. 2021; 2021: 9991001.
[30]
Luo C, Tao L. The Function and Mechanisms of Autophagy in Traumatic Brain Injury. Advances in Experimental Medicine and Biology. 2020; 1207: 635–648.
[31]
Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang D. Ferroptosis is a type of autophagy-dependent cell death. Seminars in Cancer Biology. 2020; 66: 89–100.
[32]
Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ 3rd, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016; 12: 1425–1428.
[33]
Liu J, Guo ZN, Yan XL, Huang S, Ren JX, Luo Y, et al. Crosstalk Between Autophagy and Ferroptosis and Its Putative Role in Ischemic Stroke. Frontiers in Cellular Neuroscience. 2020; 14: 577403.
[34]
Fricker M, Tolkovsky AM, Borutaite V, Coleman M, Brown GC. Neuronal Cell Death. Physiological Reviews. 2018; 98: 813–880.
[35]
Zheng DW, Lei Q, Zhu JY, Fan JX, Li CX, Li C, et al. Switching Apoptosis to Ferroptosis: Metal-Organic Network for High-Efficiency Anticancer Therapy. Nano Letters. 2017; 17: 284–291.
[36]
Hong SH, Lee DH, Lee YS, Jo MJ, Jeong YA, Kwon WT, et al. Molecular crosstalk between ferroptosis and apoptosis: emerging role of ER stress-induced p53-independent PUMA expression. Cancer Research. 2018; 78: 2321.
[37]
Chen KW, Demarco B, Heilig R, Shkarina K, Boettcher A, Farady CJ, et al. Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly. The EMBO Journal. 2019; 38: e101638.
[38]
Hu X, Chen H, Xu H, Wu Y, Wu C, Jia C, et al. Role of Pyroptosis in Traumatic Brain and Spinal Cord Injuries. International Journal of Biological Sciences. 2020; 16: 2042–2050.
[39]
Fromenty B, Roden M. Mitochondrial alterations in fatty liver diseases. Journal of Hepatology. 2023; 78: 415–429.
[40]
Scorletti E, Carr RM. A new perspective on NAFLD: Focusing on lipid droplets. Journal of Hepatology. 2022; 76: 934–945.
[41]
Gao H, Jin Z, Bandyopadhyay G, Wang G, Zhang D, Rocha KCE, et al. Aberrant iron distribution via hepatocyte-stellate cell axis drives liver lipogenesis and fibrosis. Cell Metabolism. 2022; 34: 1201–1213.e5.
[42]
Yin X, Guo X, Liu Z, Wang J. Advances in the Diagnosis and Treatment of Non-Alcoholic Fatty Liver Disease. International Journal of Molecular Sciences. 2023; 24: 2844.
[43]
Loomba R, Friedman SL, Shulman GI. Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell. 2021; 184: 2537–2564.
[44]
Tong J, Li D, Meng H, Sun D, Lan X, Ni M, et al. Targeting a novel inducible GPX4 alternative isoform to alleviate ferroptosis and treat metabolic-associated fatty liver disease. Acta Pharmaceutica Sinica. B. 2022; 12: 3650–3666.
[45]
Tong J, Lan XT, Zhang Z, Liu Y, Sun DY, Wang XJ, et al. Ferroptosis inhibitor liproxstatin-1 alleviates metabolic dysfunction-associated fatty liver disease in mice: potential involvement of PANoptosis. Acta Pharmacologica Sinica. 2023; 44: 1014–1028.
[46]
Chen X, Li J, Kang R, Klionsky DJ, Tang D. Ferroptosis: machinery and regulation. Autophagy. 2021; 17: 2054–2081.
[47]
Ganz T. Systemic iron homeostasis. Physiological Reviews. 2013; 93: 1721–1741.
[48]
Zanninelli G, Loréal O, Brissot P, Konijn AM, Slotki IN, Hider RC, et al. The labile iron pool of hepatocytes in chronic and acute iron overload and chelator-induced iron deprivation. Journal of Hepatology. 2002; 36: 39–46.
[49]
Shen Z, Liu T, Li Y, Lau J, Yang Z, Fan W, et al. Fenton-Reaction-Acceleratable Magnetic Nanoparticles for Ferroptosis Therapy of Orthotopic Brain Tumors. ACS Nano. 2018; 12: 11355–11365.
[50]
Ajoolabady A, Aslkhodapasandhokmabad H, Libby P, Tuomilehto J, Lip GYH, Penninger JM, et al. Ferritinophagy and ferroptosis in the management of metabolic diseases. Trends in Endocrinology and Metabolism. 2021; 32: 444–462.
[51]
Jia M, Zhang H, Qin Q, Hou Y, Zhang X, Chen D, et al. Ferroptosis as a new therapeutic opportunity for nonviral liver disease. European Journal of Pharmacology. 2021; 908: 174319.
[52]
Folgueras AR, Freitas-Rodríguez S, Ramsay AJ, Garabaya C, Rodríguez F, Velasco G, et al. Matriptase-2 deficiency protects from obesity by modulating iron homeostasis. Nature Communications. 2018; 9: 1350.
[53]
Chen GH, Song CC, Pantopoulos K, Wei XL, Zheng H, Luo Z. Mitochondrial oxidative stress mediated Fe-induced ferroptosis via the NRF2-ARE pathway. Free Radical Biology & Medicine. 2022; 180: 95–107.
[54]
Ding SB, Chu XL, Jin YX, Jiang JJ, Zhao X, Yu M. Epigallocatechin gallate alleviates high-fat diet-induced hepatic lipotoxicity by targeting mitochondrial ROS-mediated ferroptosis. Frontiers in Pharmacology. 2023; 14: 1148814.
[55]
Dong J, Du C, Xu C, Wang Q, Wang Z, Zhu Q, et al. Verbenalin attenuates hepatic damage and mitochondrial dysfunction in alcohol-associated steatohepatitis by regulating MDMX/PPARα-mediated ferroptosis. Journal of Ethnopharmacology. 2023; 307: 116227.
[56]
Jiang JJ, Zhang GF, Zheng JY, Sun JH, Ding SB. Targeting Mitochondrial ROS-Mediated Ferroptosis by Quercetin Alleviates High-Fat Diet-Induced Hepatic Lipotoxicity. Frontiers in Pharmacology. 2022; 13: 876550.
[57]
Zhuge A, Li S, Yuan Y, Han S, Xia J, Wang Q, et al. Microbiota-induced lipid peroxidation impairs obeticholic acid-mediated antifibrotic effect towards nonalcoholic steatohepatitis in mice. Redox Biology. 2023; 59: 102582.
[58]
Ursini F, Maiorino M. Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radical Biology & Medicine. 2020; 152: 175–185.
[59]
Mohs A, Otto T, Schneider KM, Peltzer M, Boekschoten M, Holland CH, et al. Hepatocyte-specific NRF2 activation controls fibrogenesis and carcinogenesis in steatohepatitis. Journal of Hepatology. 2021; 74: 638–648.
[60]
Liu Y, Xu W, Zhai T, You J, Chen Y. Silibinin ameliorates hepatic lipid accumulation and oxidative stress in mice with non-alcoholic steatohepatitis by regulating CFLAR-JNK pathway. Acta Pharmaceutica Sinica. B. 2019; 9: 745–757.
[61]
Chen J, Li X, Ge C, Min J, Wang F. The multifaceted role of ferroptosis in liver disease. Cell Death and Differentiation. 2022; 29: 467–480.
[62]
Maiorino M, Conrad M, Ursini F. GPx4, Lipid Peroxidation, and Cell Death: Discoveries, Rediscoveries, and Open Issues. Antioxidants & Redox Signaling. 2018; 29: 61–74.
[63]
Qi J, Kim JW, Zhou Z, Lim CW, Kim B. Ferroptosis Affects the Progression of Nonalcoholic Steatohepatitis via the Modulation of Lipid Peroxidation-Mediated Cell Death in Mice. The American Journal of Pathology. 2020; 190: 68–81.
[64]
Zhu Z, Zhang Y, Huang X, Can L, Zhao X, Wang Y, et al. Thymosin beta 4 alleviates non-alcoholic fatty liver by inhibiting ferroptosis via up-regulation of GPX4. European Journal of Pharmacology. 2021; 908: 174351.
[65]
Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015; 520: 57–62.
[66]
Zhang Z, Guo M, Li Y, Shen M, Kong D, Shao J, et al. RNA-binding protein ZFP36/TTP protects against ferroptosis by regulating autophagy signaling pathway in hepatic stellate cells. Autophagy. 2020; 16: 1482–1505.
[67]
Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein & Cell. 2021; 12: 599–620.
[68]
Yuan S, Wei C, Liu G, Zhang L, Li J, Li L, et al. Sorafenib attenuates liver fibrosis by triggering hepatic stellate cell ferroptosis via HIF-1α/SLC7A11 pathway. Cell Proliferation. 2022; 55: e13158.
[69]
Kang R, Zhu S, Zeh HJ, Klionsky DJ, Tang D. BECN1 is a new driver of ferroptosis. Autophagy. 2018; 14: 2173–2175.
[70]
Wang X, Chen X, Zhou W, Men H, Bao T, Sun Y, et al. Ferroptosis is essential for diabetic cardiomyopathy and is prevented by sulforaphane via AMPK/NRF2 pathways. Acta Pharmaceutica Sinica. B. 2022; 12: 708–722.
[71]
Fiore A, Zeitler L, Russier M, Groß A, Hiller MK, Parker JL, et al. Kynurenine importation by SLC7A11 propagates anti-ferroptotic signaling. Molecular Cell. 2022; 82: 920–932.e7.
[72]
Verma AK, Sharma A, Subramaniyam N, Gandhi CR. Augmenter of liver regeneration: Mitochondrial function and steatohepatitis. Journal of Hepatology. 2022; 77: 1410–1421.
[73]
Tonelli C, Chio IIC, Tuveson DA. Transcriptional Regulation by Nrf2. Antioxidants & Redox Signaling. 2018; 29: 1727–1745.
[74]
Fan Z, Wirth AK, Chen D, Wruck CJ, Rauh M, Buchfelder M, et al. Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 2017; 6: e371.
[75]
Stockwell BR, Jiang X, Gu W. Emerging Mechanisms and Disease Relevance of Ferroptosis. Trends in Cell Biology. 2020; 30: 478–490.
[76]
Wu J, Wang Y, Jiang R, Xue R, Yin X, Wu M, et al. Ferroptosis in liver disease: new insights into disease mechanisms. Cell Death Discovery. 2021; 7: 276.
[77]
Lim PJ, Duarte TL, Arezes J, Garcia-Santos D, Hamdi A, Pasricha SR, et al. Nrf2 controls iron homeostasis in haemochromatosis and thalassaemia via Bmp6 and hepcidin. Nature Metabolism. 2019; 1: 519–531.
[78]
Li J, Wang T, Liu P, Yang F, Wang X, Zheng W, et al. Hesperetin ameliorates hepatic oxidative stress and inflammation via the PI3K/AKT-Nrf2-ARE pathway in oleic acid-induced HepG2 cells and a rat model of high-fat diet-induced NAFLD. Food & Function. 2021; 12: 3898–3918.
[79]
Gao G, Xie Z, Li EW, Yuan Y, Fu Y, Wang P, 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.
[80]
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 Chemical Biology. 2016; 23: 225–235.
[81]
Kraft VAN, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, et al. GTP Cyclohydrolase 1/Tetrahydrobiopterin Counteract Ferroptosis through Lipid Remodeling. ACS Central Science. 2020; 6: 41–53.
[82]
Candelaria PV, Leoh LS, Penichet ML, Daniels-Wells TR. Antibodies Targeting the Transferrin Receptor 1 (TfR1) as Direct Anti-cancer Agents. Frontiers in Immunology. 2021; 12: 607692.
[83]
Cui R, Choi SE, Kim TH, Lee HJ, Lee SJ, Kang Y, et al. Iron overload by transferrin receptor protein 1 regulation plays an important role in palmitate-induced insulin resistance in human skeletal muscle cells. FASEB Journal. 2019; 33: 1771–1786.
[84]
Kawabata H. Transferrin and transferrin receptors update. Free Radical Biology & Medicine. 2019; 133: 46–54.
[85]
Chen X, Yu C, Kang R, Kroemer G, Tang D. Cellular degradation systems in ferroptosis. Cell Death and Differentiation. 2021; 28: 1135–1148.
[86]
Feng G, Byrne CD, Targher G, Wang F, Zheng MH. Ferroptosis and metabolic dysfunction-associated fatty liver disease: Is there a link? Liver International. 2022; 42: 1496–1502.
[87]
Brown CW, Amante JJ, Chhoy P, Elaimy AL, Liu H, Zhu LJ, et al. Prominin2 Drives Ferroptosis Resistance by Stimulating Iron Export. Developmental Cell. 2019; 51: 575–586.e4.
[88]
He J, Li Z, Xia P, Shi A, FuChen X, Zhang J, et al. Ferroptosis and ferritinophagy in diabetes complications. Molecular Metabolism. 2022; 60: 101470.
[89]
Mancias JD, Wang X, Gygi SP, Harper JW, Kimmelman AC. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 2014; 509: 105–109.
[90]
Mancias JD, Pontano Vaites L, Nissim S, Biancur DE, Kim AJ, Wang X, et al. Ferritinophagy via NCOA4 is required for erythropoiesis and is regulated by iron dependent HERC2-mediated proteolysis. eLife. 2015; 4: e10308.
[91]
Bellelli R, Federico G, Matte’ A, Colecchia D, Iolascon A, Chiariello M, et al. NCOA4 Deficiency Impairs Systemic Iron Homeostasis. Cell Reports. 2016; 14: 411–421.
[92]
Li H, Yu Y, Liu Y, Luo Z, Law BYK, Zheng Y, et al. Ursolic acid enhances the antitumor effects of sorafenib associated with Mcl-1-related apoptosis and SLC7A11-dependent ferroptosis in human cancer. Pharmacological Research. 2022; 182: 106306.
[93]
DeHart DN, Fang D, Heslop K, Li L, Lemasters JJ, Maldonado EN. Opening of voltage dependent anion channels promotes reactive oxygen species generation, mitochondrial dysfunction and cell death in cancer cells. Biochemical Pharmacology. 2018; 148: 155–162.
[94]
Li S, Zhuge A, Wang K, Xia J, Wang Q, Han S, et al. Obeticholic acid and ferrostatin-1 differentially ameliorate non-alcoholic steatohepatitis in AMLN diet-fed ob/ob mice. Frontiers in Pharmacology. 2022; 13: 1081553.
[95]
Chen J, Ou Z, Gao T, Yang Y, Shu A, Xu H, et al. Ginkgolide B alleviates oxidative stress and ferroptosis by inhibiting GPX4 ubiquitination to improve diabetic nephropathy. Biomedicine & Pharmacotherapy. 2022; 156: 113953.
[96]
Georgoulis M, Fragopoulou E, Kontogianni MD, Margariti A, Boulamatsi O, Detopoulou P, et al. Blood redox status is associated with the likelihood of nonalcoholic fatty liver disease irrespectively of diet’s total antioxidant capacity. Nutrition Research. 2015; 35: 41–48.
[97]
Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nature Cell Biology. 2014; 16: 1180–1191.
[98]
Zilka O, Shah R, Li B, Friedmann Angeli JP, Griesser M, Conrad M, et al. On the Mechanism of Cytoprotection by Ferrostatin-1 and Liproxstatin-1 and the Role of Lipid Peroxidation in Ferroptotic Cell Death. ACS Central Science. 2017; 3: 232–243.
[99]
Botham KM, Napolitano M, Bravo E. The Emerging Role of Disturbed CoQ Metabolism in Nonalcoholic Fatty Liver Disease Development and Progression. Nutrients. 2015; 7: 9834–9846.
[100]
Kong N, Chen X, Feng J, Duan T, Liu S, Sun X, et al. Baicalin induces ferroptosis in bladder cancer cells by downregulating FTH1. Acta Pharmaceutica Sinica. B. 2021; 11: 4045–4054.
[101]
Li M, Meng Z, Yu S, Li J, Wang Y, Yang W, et al. Baicalein ameliorates cerebral ischemia-reperfusion injury by inhibiting ferroptosis via regulating GPX4/ACSL4/ACSL3 axis. Chemico-Biological Interactions. 2022; 366: 110137.
[102]
Yang M, Li X, Li H, Zhang X, Liu X, Song Y. Baicalein inhibits RLS3-induced ferroptosis in melanocytes. Biochemical and Biophysical Research Communications. 2021; 561: 65–72.
[103]
Krainz T, Gaschler MM, Lim C, Sacher JR, Stockwell BR, Wipf P. A Mitochondrial-Targeted Nitroxide Is a Potent Inhibitor of Ferroptosis. ACS Central Science. 2016; 2: 653–659.
[104]
Zhao Z, Wu J, Xu H, Zhou C, Han B, Zhu H, et al. XJB-5-131 inhibited ferroptosis in tubular epithelial cells after ischemia-reperfusion injury. Cell Death & Disease. 2020; 11: 629.
[105]
Li P, Lin Q, Sun S, Yang N, Xia Y, Cao S, et al. Inhibition of cannabinoid receptor type 1 sensitizes triple-negative breast cancer cells to ferroptosis via regulating fatty acid metabolism. Cell Death & Disease. 2022; 13: 808.
[106]
Lee JY, Nam M, Son HY, Hyun K, Jang SY, Kim JW, et al. Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proceedings of the National Academy of Sciences of the United States of America. 2020; 117: 32433–32442.
[107]
Chen H, Qi Q, Wu N, Wang Y, Feng Q, Jin R, et al. Aspirin promotes RSL3-induced ferroptosis by suppressing mTOR/SREBP-1/SCD1-mediated lipogenesis in PIK3CA-mutatnt colorectal cancer. Redox Biology. 2022; 55: 102426.
[108]
Zhao Y, Li M, Yao X, Fei Y, Lin Z, Li Z, et al. HCAR1/MCT1 Regulates Tumor Ferroptosis through the Lactate-Mediated AMPK-SCD1 Activity and Its Therapeutic Implications. Cell Reports. 2020; 33: 108487.
[109]
He F, Zhang P, Liu J, Wang R, Kaufman RJ, Yaden BC, et al. ATF4 suppresses hepatocarcinogenesis by inducing SLC7A11 (xCT) to block stress-related ferroptosis. Journal of Hepatology. 2023; 79: 362–377.
[110]
Yang Y, Luo M, Zhang K, Zhang J, Gao T, Connell DO, et al. Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nature Communications. 2020; 11: 433.
[111]
Yu M, Gai C, Li Z, Ding D, Zheng J, Zhang W, et al. Targeted exosome-encapsulated erastin induced ferroptosis in triple negative breast cancer cells. Cancer Science. 2019; 110: 3173–3182.
[112]
Gao R, Kalathur RKR, Coto-Llerena M, Ercan C, Buechel D, Shuang S, et al. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Molecular Medicine. 2021; 13: e14351.
[113]
You JH, Lee J, Roh JL. Mitochondrial pyruvate carrier 1 regulates ferroptosis in drug-tolerant persister head and neck cancer cells via epithelial-mesenchymal transition. Cancer Letters. 2021; 507: 40–54.
[114]
Wang K, Zhang Z, Tsai HI, Liu Y, Gao J, Wang M, et al. Branched-chain amino acid aminotransferase 2 regulates ferroptotic cell death in cancer cells. Cell Death and Differentiation. 2021; 28: 1222–1236.
[115]
Kang YP, Mockabee-Macias A, Jiang C, Falzone A, Prieto-Farigua N, Stone E, et al. Non-canonical Glutamate-Cysteine Ligase Activity Protects against Ferroptosis. Cell Metabolism. 2021; 33: 174–189.e7.
[116]
Shin D, Kim EH, Lee J, Roh JL. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radical Biology & Medicine. 2018; 129: 454–462.
[117]
Zhu L, Hu S, Yan X, Zeng Q, Zhang B, Jiang L, et al. Ugi reaction-assisted assembly of covalent PROTACs against glutathione peroxidase 4. Bioorganic Chemistry. 2023; 134: 106461.
[118]
Chen GQ, Benthani FA, Wu J, Liang D, Bian ZX, Jiang X. Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death and Differentiation. 2020; 27: 242–254.
[119]
He Q, Yang J, Pan Z, Zhang G, Chen B, Li S, et al. Biochanin A protects against iron overload associated knee osteoarthritis via regulating iron levels and NRF2/System xc-/GPX4 axis. Biomedicine & Pharmacotherapy. 2023; 157: 113915.
[120]
Luo C, Xu W, Tang X, Liu X, Cheng Y, Wu Y, et al. Canonical Wnt signaling works downstream of iron overload to prevent ferroptosis from damaging osteoblast differentiation. Free Radical Biology & Medicine. 2022; 188: 337–350.
[121]
Zhao S, Lo CS, Miyata KN, Ghosh A, Zhao XP, Chenier I, et al. Overexpression of Nrf2 in Renal Proximal Tubular Cells Stimulates Sodium-Glucose Cotransporter 2 Expression and Exacerbates Dysglycemia and Kidney Injury in Diabetic Mice. Diabetes. 2021; 70: 1388–1403.
[122]
Arlt A, Sebens S, Krebs S, Geismann C, Grossmann M, Kruse ML, et al. Inhibition of the Nrf2 transcription factor by the alkaloid trigonelline renders pancreatic cancer cells more susceptible to apoptosis through decreased proteasomal gene expression and proteasome activity. Oncogene. 2013; 32: 4825–4835.
[123]
Gaschler MM, Andia AA, Liu H, Csuka JM, Hurlocker B, Vaiana CA, et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nature Chemical Biology. 2018; 14: 507–515.
[124]
Amos A, Jiang N, Zong D, Gu J, Zhou J, Yin L, et al. Depletion of SOD2 enhances nasopharyngeal carcinoma cell radiosensitivity via ferroptosis induction modulated by DHODH inhibition. BMC Cancer. 2023; 23: 117.
[125]
Jiang M, Song Y, Liu H, Jin Y, Li R, Zhu X. DHODH Inhibition Exerts Synergistic Therapeutic Effect with Cisplatin to Induce Ferroptosis in Cervical Cancer through Regulating mTOR Pathway. Cancers. 2023; 15: 546.
[126]
Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021; 593: 586–590.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top