Ferroptosis in hepatic ischemia‑reperfusion injury: Regulatory mechanisms and new methods for therapy (Review)

  • Authors:
    • Linfeng Luo
    • Guoheng Mo
    • Deqiang Huang
  • View Affiliations

  • Published online on: January 24, 2021     https://doi.org/10.3892/mmr.2021.11864
  • Article Number: 225
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ischemia‑reperfusion injury (IRI), also called reoxygenation injury, is the outcome of inflammatory processes and oxidative damage through the induction of oxidative stress. In the clinical setting, IRI contributes to severe hepatic injury, including liver cell death by apoptosis and ferroptosis. Ferroptosis is a novel type of cell death in hepatic IRI that involves small molecules that inhibit glutathione biosynthesis or glutathione peroxidase 4 (GPX4), which is a glutathione‑dependent antioxidant enzyme, causing mitochondrial damage. Currently, ferroptosis has been systematically described in neurological settings, kidney diseases and different types of cancer, while few studies have analysed the presence of ferroptosis and the regulatory mechanism of ferroptosis in hepatic IRI. Exploring the exact role played by ferroptosis in the liver following hepatic IRI in accordance with existing evidence and mechanisms could guide potential therapeutic interventions and provide a novel research avenue.

Introduction

Ferroptosis has emerged as a novel type of regulated cell death (RCD) in various diseases, particularly in hepatic or renal ischemia-reperfusion injury (IRI). The occurrence of ferroptosis is based on iron overload, which generates reactive oxygen species (ROS) and lipid peroxides, primarily phosphatidylethanolamine-OOH (PE-OOH) in vivo (13). Ferroptosis was identified in 2012 and was originally reported to be associated with mutant RAS cancer cells 1 (4,5). Although the mechanism of ferroptosis has a relatively specific description, primarily including iron-dependent accumulation of lipid ROS and the consumption of plasma membrane polyunsaturated fatty acids (PUFAs) (5), the role of ferroptosis in cancer, heart, liver or kidney injury, and neurotoxicity remains unclear (6). Severe hepatic IRI may lead to serious impairment of liver function or even acute liver failure (7,8). Therefore, it is critical to prevent hepatic IRI, especially in liver transplantation, due to the high risk of urgent re-transplantation (9). Distinct from other types of RCD (apoptosis, necroptosis and autophagy), ferroptosis is characterized by resulting oxidative damage in the mitochondria, which exerts harmful effects on hepatic ischemia-reperfusion (6,8). Furthermore, these injuries can be prevented by the ferroptosis-specific inhibitor ferrostatin-1 (Fer-1) and by iron chelators (6,8). Therefore, ferroptosis is a potential target for preventing and treating hepatic IRI. Thus, exploration of the exact mechanisms of ferroptosis in liver cell death is required.

The present review firstly introduces the types of hepatic IRI and subsequently describes the molecular mechanisms of liver IRI. Thirdly, the general mechanisms of ferroptosis and the role of ferroptosis in hepatic IRI are discussed; primarily including the inflammatory response and oxidative stress. In the final part of this review, several therapeutic strategies associated with ferroptosis are described in detail.

Hepatic IRI

Types of liver IRI

Hepatic IRI is still a long-standing problem in clinical conditions that occurs in hepatic resection surgery, liver transplantation and during states of shock. Two main types of hepatic IRI exist, including warm and cold IRI. Warm IRI, initiated by hepatocellular injury, occurs ischemia at routine temperature and is generally present in liver transplantation surgery or different forms of trauma or shock, and might lead to liver failure, or even bring the outcome of multiorgan failure (10). Cold IRI starts at the injury of endothelial cells in hepatic sinusoidal and microcirculation disorders with the temperature of liver decreasing rapidly and uniformly, which develops during in vitro preservation and is usually accompanied by warm IRI in the process of liver transplantation surgery (10,11). Although the two IRI types might possess distinct initial cellular targets, they do share similar pathophysiological processes, including local inflammatory innate immune activation (10,12,13), and expression of fibronectin (FN) in endothelial cells is a prominent feature of the liver injury response (14). At present, there is no evidence that hot or cold IRI causes different types of cell death.

In addition, IRI can be divided into two phases, ischemia and reperfusion, which are primarily the result of oxidative stress accompanied by nutritional deficiency, loss of blood flow, inflammation and other conditions (15). Such trauma primarily causes autophagy in liver cells, including apoptosis and necrosis (16). There is evidence that various markers of autophagy are elevated throughout the entire IRI process (17). Among them, iron-mediated death is primarily believed to be associated with oxidative stress from ROS, especially during blood reperfusion (18).

Ferroptosis is a type of iron-dependent oxidative cell death characterized by accumulation of intracellular ROS, which will be discussed in more detail. Furthermore, iron-mediated cell death is an important form of autophagy (19,20).

Molecular mechanisms of liver IRI

ROS (such as OH- and HOO-), chemically reactive species containing oxygen, are an important cause of initial liver injury (21) and are originally produced in Kupffer cells, which kill hepatocytes through lipid peroxidation, DNA oxidation and enzymatic degeneration (22). The pathway regulated by ROS that promotes apoptosis contains different molecules and transporters. Initially, ROS activates apoptosis signal-regulating kinase 1 (ASRK1) through TNF-receptor-associated factor 2 (TRAF-2) that leads to c-Jun-N-terminal kinase (JNK), which directly regulates the activities of pro- and anti-apoptotic mitochondrial proteins through different phosphorylation events or via upregulating pro-apoptotic genes through the trans-activation of specific transcription factors (23). In addition, tumour necrosis factor-α (TNF-α), subsequently released by ROS, can increase the damage after IRI by promoting extra release of inflammatory cytokines and creating positive feedback circuits, which leads to organ damage. Furthermore, TNF-α regulates the production of gangliochemical genes and adhesion molecules that are absorbed into the liver, and these neutrophils are eventually responsible for the subsequent stage of injury. In addition to TFN-α, other proinflammatory cytokines, such as IL-1β (24), IL-12 (25), IL-18 and IL-6 (10), are critical for the hepatic inflammatory response. Furthermore, IL-12 is indispensable for fully producing TNF-α in the liver and the ensuing inflammatory response, which was confirmed using neutralizing antibodies or IL-12 knockout mice to eliminate IL-12 (10). These injuries eventually lead to biliary microcirculatory disorders and apoptosis of biliary epithelial cells.

Relevant proinflammatory signalling pathways include nuclear factor κB (NF-κB), which is activated by proinflammatory cytokines, such as IL-1 and TNF-α.

Of note, ROS can also stimulate NF-κB to promote hepatic IRI (26,27). Hence, antioxidants decrease the expression of pro-inflammatory genes by inhibiting the activation of NF-κB (2831). On the other hand, superoxide formation in endothelial cells (32) and hepatocytes (33) was recently shown to originate from a phagocyte-type nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Therefore, inhibiting Rac1, a member of the Rho family of small GTPases that can regulate this oxidase, attenuates intracellular oxidant stress and protects against hepatocyte injury during the early reperfusion phase (33).

Brief overview of ferroptosis

Ferroptosis is a form of RCD that is dependent on iron and ROS, and is initiated by the failure of glutathione biosynthesis or the inactivation of glutathione peroxidase 4 (GPX4), an antioxidant enzyme that depends on glutathione, thus resulting in lipid peroxidation, the consumption of PUFAs, and eventual cell death (4). Ferroptosis has distinct features at the morphological, biochemical, and genetic level compared with other forms of RCD, including necroptosis, apoptosis and autophagy. Small molecules, such as erastin, ras-selective lethal small molecule (RSL3), high concentrations of glutamate, and sulfasalazine are known to reduce ferroptosis, while α-tocopherol, ferrostatin-1, liproxstatin-1, glutathione, zileuton and iron chelators (such as deferasirox, deferiprone, chelation with deferoxamine and 1,10-phenanthroline) are inhibitors involved in relevant mechanisms of ferroptosis that contribute to hepatic IRI (6,34,35). However, how ferroptosis plays an essential role in cell injury is not well understood. Moreover, the detailed signalling pathway that lie between IRI induction and ferroptosis activation remains unknown. Validation is still required to understand all the molecules on this pathway, and the known details are summarized, which is still the inevitable limitation of this review.

The ferroptotic signalling pathway

The activation of mitogen-activated protein kinase (MAPK), iron metabolism and lipid peroxidation signalling pathways are currently known to contribute to ferroptotic cell death (36). However, it has been reported that the MAPK pathway was associated to to cancer cell death, which inhibits ferroptosis induced by erastin by blocking the Ras/Raf/MEK/ERK pathway in Ras-mutated cancer cells (8). Hence, iron and ROS signalling pathways are primarily described in the present review.

Iron and ferroptosis

The principal role of iron is to transport oxygen in the haematological system. Iron possesses two forms in cells: Fe2+ and Fe3+. The Fe2+ absorbed into the blood is oxidized to Fe3+ by ceruloplasmin, and Fe3+ is transported to the tissues after binding with transferrin or is absorbed into the cell through the membrane transferrin receptor (TfR), then localised to the internal body, and Fe3+ is reduced to Fe2+ by the ferrireductase activity of six-transmembrane epithelial antigen of prostate 3 (STEAP3). Finally, release of Fe2+ from the endosome to a labile iron pool is mediated by divalent metal transporter 1 (DMT1) in the cytoplasm. Excess iron is kept in the monocyte-macrophage system of the liver, spleen, bone marrow and other organs in the form of ferritin and hemosiderin. Membrane protein ferroportin, an iron efflux pump that oxidizes Fe2+ into Fe3+, transmits signals to mediate iron output (Fig. 1). Fe2+ has the feature of a catalyst, which can transfer electrons and participate in various oxidation-reduction reactions, while Fe3+ primarily exists in the process of transportation and storage.

However, iron overload is recognized as poisonous to cells, since the transferred electrons are given to O2 and H2O2 to produce superoxide anions and hydroxyl radicals, which exert harmful influences on biological macromolecules, such as nucleic acids, proteins and lipids (37). Moreover, Fe2+ can oxidize organics combined with H2O2 to generate ROS by the Fenton reaction (3,37). As Wang et al (38) stated, hepatocytes and macrophages are sensitive to extracellular iron levels, and a high-iron diet in mice could trigger ferroptotic cell death. Additionally, shock protein family B member 1 (HSPB1) inhibits ferroptosis through decreasing intracellular iron levels and upholding glutathione (GSH) in its reduced form. Furthermore, TfR1-mediated iron uptake is inhibited by HSPB1, which blocks the endocytosis and recycling of transferrin to decrease intracellular iron levels (3941). These studies suggest that iron plays a critical role during ferroptosis, although the role of iron in the signalling pathway of ferroptosis remains poorly understood. To date, in addition to the Fenton reaction by Fe2+, there is an additional source involved in the iron-dependent accumulation of lipid ROS in ferroptosis: Lipid peroxidation controlled by iron-containing lipoxygenases (LOXs) (Fig. 1) (42).

LOXs are a family of non-haem, iron-containing enzymes, and most of them catalyse the deoxygenation of PUFAs, such as arachidonic acid (AA) and linolenic acid, in lipids containing a cis, cis−1,4-pentadiene into cell signalling agents (4345). As one type of PUFA, AA is converted to adrenoyl (AdA) under the action of elongase (2), and then on the endoplasmic reticulum or mitochondrial outer membrane, AA and AdA are catalysed by acyl-CoA synthetase long-chain family 4 (ACSL4) to form AdA-CoA/AA-CoA, which is next esterified to AA-PE under the action of lysophosphatidylcholine acyltransferase 3 (LPCAT3), finally forming AA-OOH-PE, a cell death signal of ferroptosis, under the oxidation of iron-containing LOXs (46,47). However, the role of iron in regulating LOXs relies on phosphorylase kinase G2 (PHKG2), which activates glycogen phosphorylase (GP) to release glucose-1-phosphate from glycogen, promoting the phosphorylation of LOXs to synthesise lipid peroxides. Furthermore, glycogen primarily exists in liver and muscle tissues; therefore, glycogen breakdown might be an important factor in ferroptosis in liver or muscle injury, although no current studies have confirmed this. In conclusion, iron-containing LOXs are required for ferroptosis in the reaction of lipid peroxidation, and inhibition of ACSL4 and LPCAT3 may decrease oxidation of some sensitive fatty acids in the membrane. However, more studies are required to further explain the detailed role of iron in mediating LOX activity.

ROS and ferroptosis

ROS are primarily located in the mitochondria during electron transport (48), and iron-dependent lipid ROS produced by the two aforementioned sources mediate lipid peroxidation that further promotes the accumulation of lipid peroxides (Fig. 1) (6). Furthermore, peroxides lead to fundamental changes in lipids, especially phospholipids, which are essential for maintaining the integrity of the mitochondrial membrane architecture. In addition, peroxides can affect the fluidity of lipids, thus blocking receptor clustering and propagating inflammatory signalling (49). The peroxidation of phospholipids also inactivates membrane-bound proteins, ultimately causing destruction of the membrane (50). In addition, the occurrence of ROS accompanies single electron leakage of oxidative phosphorylation in the mitochondria, thus decreasing the yield of ATP and inhibiting cell survival (49). Thus, increased mitochondrial ROS can destroy the integrity of the electron transport chain, causing respiratory chain dysfunction (51). Moreover, lipid peroxidation products lead to mtDNA damage, further contributing to mitochondrial mutations, whereas mutations in the mitochondria further increase levels of ROS with toxic effects (49,51), resulting in a vicious circle. On the one hand, stable aldehyde peroxidation products from PUFAs, such as 4-hydroxynonenal and malondialdehydes, are involved in mtDNA mutations or deletions (52). Although aldehydes have significant toxic effects, some enzymes in vivo, such as cytochrome P450 (CYP), aldehyde dehydrogenase and aldo-keto reductases, can metabolize them to less toxic compounds (53). Evidence shows that members of CYP3A and CYP4A oxidize 4-hydroxynonenal (38). However, when mitochondrial dysfunction and increased ROS occur, oxidation and antioxidant systems lose balance in vivo, and the mitochondrial crest decreases or disappears. The outer wall of the mitochondrial membrane ruptures, which is a typical feature of ferroptosis that differentiates it from apoptosis, necroptosis and autophagy (6). In conclusion, it is postulated that ROS-induced ferroptosis depends on changes in the mitochondria.

Ferroptosis in hepatic IRI

Recent evidence has shown that ferroptosis is associated with the pathogenesis of various diseases, such as neoplastic diseases and ischemic injury to the brain, heart, liver, kidney and intestine (68,5456). Furthermore, several studies have indicated that inhibitors of ferroptosis, such as ferrostatins-1 and liproxstatins-1, protect against cell death in the liver, kidney, brain and heart ischemic injury in mouse models (5557). In a study by Yamada et al (8), the role of ferroptosis in hepatic IRI was explored, which established a murine model of hepatic ischemia-reperfusion injury and found that upregulation of the ferroptosis marker Ptgs2, lipid peroxidation and liver damage were induced by hepatic ischemia reperfusion. Moreover, all of these liver cell injuries can be prevented when super-inducing the ferroptosis-specific inhibitor Fer-1 and by iron chelation (8,54). Thus, it seems that iron overload is a critical factor for hepatic IRI, and the pathogenesis of hepatic IRI is partly attributed to ferroptosis (8).

Role of iron in inflammation in hepatic IRI

At present, it is widely accepted that hepatic IRI is characterized by an excessive inflammatory response, release of inflammatory cytokines and chemokines, as well as neutrophil and macrophages infiltration. In addition, IL-1β is the decisive factor that drives many sterile inflammatory diseases (58), especially in hepatic IRI (Fig. 2). However, there are two stages in the release process of IL-1β: The synthesis of pro-IL-1β and the maturation of IL-1β. Regarding the priming process, Toll-like receptor 4 (TLR4) binds to the ligands (such as heat shock proteins, fibronectin, fibrinogen, high mobility group box 1, hyaluronan and heparin sulphate) to produce signal transduction (59) and then through NF-κB activation, induces pro-IL-1β synthesis. Concerning the maturation of IL-1β, it is reported that receptor family pyrin domain containing 3 (NLRP3) inflammasomes play an important role (6062). Firstly, NLRP3 inflammasomes, containing adaptor molecule apoptosis-associated speck-like protein, containing a caspase recruitment domain, cysteine protease caspase-1 and NLRP3, contribute to the activation of caspase-1. Secondly, pro-IL-1β is processed into its mature form by caspase-1 as an IL-1β converting enzyme, and caspase-1 induces the release of IL-1β. Finally, IL-1β induces expression of IL-6, TNF-α, Ccl2, Cxcl1 and Cxcl2, leading to tissue injury in the ischemia-reperfusion liver (63).

By performing real-time RT-PCR analysis, a previous investigation evaluated the expression of inflammatory cytokines and cell markers to confirm the association between iron overload and inflammatory response in the liver (8). The results showed that inflammatory cytokines and cell markers were significantly inhibited by Fer-1. Furthermore, the infiltration of neutrophils and macrophages was also apparently inhibited (8). This implies that ferroptosis in liver cell death might be closely associated with the inflammatory reaction in hepatic IRI. Further evidence demonstrated that iron is central to many aspects of the innate immune response, including ROS generation and host inflammatory regulation (1). Iron overload causes metabolic disturbance, leading to an increased susceptibility to infection and triggering the inflammatory response as the oversaturation of host transferrin leads to defective nutritional immunity (64). In a healthy individual, in vivo iron is a stable condition, and excess iron accumulation can lead to the production of ROS. Regarding the role of iron in inflammation in hepatic IRI, several studies have demonstrated that the TLR4-activated inflammatory response is modulated by iron, as well as increasing oxidative stress through the generation of reactive oxygen and nitrogen species (65,66). As previously stated, induction of pro-IL-1β synthesis is required for NF-κB activation, while ROS can activate the transcription factor NF-κB (67). Furthermore, systemic iron homeostasis is regulated in the liver, and the hepatic hormone hepcidin is the central regulator (68). There are two pathways to increase the expression of hepcidin at the transcriptional level, including inflammatory cytokines, such as IL-1β and IL-6 via the JAK/STAT3 pathway, and iron via the BMP/Smad signalling pathway (Fig. 3) (69). Increased hepcidin downregulates the level of ferroportin, the sole known iron exporter on the cell surface of hepatocytes, so that intracellular iron levels increase due to suppression of iron export (69). Hence, it is conjectured that the inflammatory response triggered in hepatic IRI induces iron overload in hepatocytes. In other words, ferroptosis greatly contributes to the pathogenesis of hepatic IRI.

Role of iron in oxidative damage in hepatic IRI

In order to continue exploring the role of iron in hepatic IRI, oxidative damage in liver cell injury is described in detail in the present review. In the mechanism of ferroptosis occurrence, iron-dependent accumulation of lipid ROS can be produced from GSH depletion and NADPH-dependent lipid peroxidation (5,70). Low levels of ROS, including hydrogen peroxide (H2O2), superoxide anions (O2−) and hydroxyl radicals (-OH) (7173), play an indispensable role in various molecular biological processes, such as intracellular messaging and molecular pathways in cellular progression (cell growth, differentiation and death) or immunity (74), the arrest of growth, and defence against microorganisms and apoptosis (75,76). In contrast, high or and/or inadequate removal of lipid ROS from Kupffer cells is the cardinal factor in vascular and parenchymal cell oxidative damage during reperfusion, which occurs by inducing oxidant stress (77). According to current studies, there are distinct factors participating oxidant stress. First, complement and polymorphonuclear neutrophils (PMNs) participate in the process of oxidant stress induced by Kupffer cells (Fig. 1) (78). Kupffer cells release intracellular proteins and ROS during hepatic ischemia, inducing the activation of complement and leading to slight initial injury. Complement, on the one hand, activates and further stimulates Kupffer cells to produce ROS, and on the other hand, directly or indirectly causes activation and generation of PMNs in the liver through aggravating the initial injury induced by Kupffer cells. Moreover, the mechanism of complement-induced activation of neutrophils has been described in other studies, which demonstrated that complement factors, such as C5a, can recruit neutrophils into sinusoids by upregulating the Mac-1 receptor on circulating neutrophils (79,80). However, the generation of PMNs in the liver also causes hepatocyte injury, and this damage further promotes the activation of complement and PMNs, stimulating the Kupffer cells to produce reactive oxygen that contributes to the oxidative damage in hepatic IRI (81). An investigation using cobra venom factor (CVF) that effectively inhibits complement activation through the classical and alternate pathway (82,83), induced the depletion of complement and a novel soluble complement receptor type 1, significantly attenuating the increase of plasma alanine aminotransferase (ALT) activities (81). This experiment confirms that complement exerts an indispensable role in Kupffer cell-induced oxidative injury. Secondly, a current view suggested that Kupffer cells could be potentially activated during the ischemic period, resulting in the Kupffer cells generating superoxides, such as the superoxide anion radical (O2−), when subjected to reoxygenation (84). This process is likely to be activated by nitric oxide (NOX), while the oxidase also stimulates the activation of ferroptosis by inhibiting glutathione biosynthesis or GPX4 (4). Moreover, NOX induces lipid peroxidation, producing many complex products, such as epoxides, hydroperoxides, and carbonyl compounds (85). Lipid peroxidation primarily targets cellular membranes, then peroxide PUFAs of membrane phospholipids, and finally causes structural and functional tissue damage due to the disintegration of the cellular membrane (86). In hepatic IRI, this mechanism exacerbates erythrocyte functions, impairing membrane integrity (87) and significantly altering erythrocyte deformability (86). Diminished erythrocyte deformability not only attenuates oxygen transport capacity of the erythrocytes but also affects the survival of circulating erythrocytes (87,88). Furthermore, another product of lipid peroxidation is malondialdehyde, which can react with DNA and as a result is toxic and mutagenic (88). Ultimately, malondialdehyde is substantially generated in the liver and results in the death of hepatic parenchyma cells. An antioxidant defence system containing glutathione peroxidase (GPX), ascorbic acid (vitamin C), superoxide dismutase (SOD), a-tocopherol (vitamin E), catalase (CAT), and GSH also exists in the body to fight against the generation of free radicals by eliminating superoxide anions and hydrogen peroxides (8992). When the balance between oxidation and antioxidant systems is disrupted, increased lipid peroxidation can induce oxidative stress (89). As previously reported, iron promotes ferroptosis by lipid peroxidation in hepatic IRI (6,8,36). To affirm this mechanism, a mouse ischemic model was given iron chelation with deferoxamine treatment, which decreased the liver iron content and serum ferritin levels (8).

Other types of regulated cell death

In addition to ferroptosis, there are other types of regulated cell death, such as apoptosis, necrosis and autophagy. Apoptosis, a form of programmed cell death, leads to characteristic cell changes including blebbing, cell shrinkage, nuclear fragmentation, chromatin condensation, chromosomal DNA fragmentation, and global mRNA decay, which finally leads to the formation of apoptotic bodies and phagocytosis of the apoptotic bodies by adjacent parenchymal cells, neoplastic cells or macrophages (93). The pathways that initiate apoptosis are categorized as intrinsic or extrinsic, which are initiated by different types of stimuli, and finally through pro-apoptotic proteins to activate caspase-9 and caspase-8, respectively (9496). Bcl-2 family members and cell death receptor/ligand (FasL/FasR and TNF-α/TNFR1) are the main molecules of the main apoptosis signal pathway (95,97,98). In contrast to apoptosis, necrosis, a passive type of RCD is initiated by external physical or chemical factors, and mainly characterized by swelling of cytoplasm and mitochondria, loss of plasma membrane integrity, resulting in the release of pro-inflammatory factors and the inflammation in the surrounding tissue (99). Similar to the extrinsic signaling pathway of apoptosis, necrosis also is initiated by cell death receptor/ligand (FasL/FasR and TNF-α/TNFR1), which forms a death-inducing signaling complex (DISC) with procaspase-8 by recruiting Fas-associated death-domain and receptor-interacting serine/threonine-protein kinase 1 (RIPK1) (94). Differentially, apoptosis originates from the activation of caspase-8 by the complex, while necrosis is caused by deubiquitinated RIPK1 recruiting RIPK3 through the RIP homotypic interaction motif interaction and phosphorylation of mixed-lineage kinase domain-like (MLKL) protein when caspase-8 activity is inhibited. The oligomerization of phosphorylated MLKL seems to bind to high-order inositol phosphate (IP), which is then transferred to the plasma membrane to induce cytolysis, resulting in the release of pro-inflammatory damage-associated molecular proteins. Besides, it also activates NLRP3, then leads to the secretion of interleukin (IL)-1β and IL-18 (100). It is common that the inflammatory responses activated by ferroptosis and necrosis both involve the participation of molecules such as IL-1β and TNF-α. However, ferroptosis accounts more for lipid peroxidation caused by iron overload, and the accumulation of ROS leads to the hepatocyte mitochondrial membrane permeability.

As aforementioned, hepatic IRI is divided into two processes: Ischemia and reperfusion. In the process of liver ischemia, it mainly causes hypoxia and energy depletion, and the reperfusion process causes oxidative stress and inflammatory reaction. Both of these processes will lead to apoptosis and necrosis, and finally result in autophagy. Autophagy is another type of RCD regarding to a process that cytoplasmic substances are transported to lysosomes, autophagy-related protein forms autophagosomes, and finally the components contained are degraded. More importantly, autophagy plays a critical role in regulating liver metabolism, energy production and quality control checkpoints as organelles such as mitochondria (101). However, a study shows that autophagy is also associated with hepatocyte death (102). Autophagy can be divided into three types: i) autophagy-associated cell death; ii) autophagy-mediated cell death, and iii) autophagy-dependent cell death (103). For the first two types, autophagy plays a minor role in the mechanism of cell death, thus should depend on other types of cell death, such as apoptosis, necrosis and ferroptosis. The third type of autophagy can independently mediate the mechanism of cell death, so in the process of hepatocyte death, apoptosis, necrosis, autophagy and ferroptosis can be either interdependent or independently mediated. Interestingly, studies have shown that there is a link between the activation of autophagy and the development of ferroptosis, a process known as ‘ferritinophagy’, which is characterized by autophagy degradation of ferritin. In this process, the nuclear receptor coactivator 4, a selective cargo receptor for the turnover of ferritin, enables the maintenance of iron homeostasis, then results in iron overload and promotes the development of ferroptosis through the degradation of ferritin (104,105).

Current therapeutic strategies in hepatic IRI

Currently, some potential therapeutic strategies have been reported for ferroptosis regulation in liver ischemia-reperfusion, primarily including antioxidants and iron-removing molecules (such as desferoxamine, ferrostatin-1, liproxstatin-1, α-tocopherol, ascorbic acid, GSH, alpha lipoic acid, gadolinium chloride, zileuton and gadolinium chloride).

Desferoxamine

As aforementioned, iron overload promotes lipid peroxidation and is involved in the inflammatory response of hepatic IRI, leading to ferroptosis in liver cells. Desferoxamine is an iron chelator that can decrease the levels of intracellular iron. Experimental studies on hepatic ischemia models using desferoxamine pretreatment have shown beneficial effects, such as decreasing the liver iron content, decreasing serum ferritin levels, and restoring total GSH levels, in response to warm or cold hepatic ischemia (8,106,107).

Ferrostatin-1

Ferrostatin-1 is a first-generation ferrostatin that inhibits ferroptosis by interfering with ROS accumulation from lipid peroxidation (7,8). Mechanistically, to fight against ferroptosis, a previous study (108) demonstrated that anti-ferroptotic activity of fer-1 primarily depends on the scavenging of initiating alkoxyl radicals produced by ferrous iron from lipid hydroperoxides, and moreover, when fer-1 attenuates lipid peroxidation, its levels are not significantly consumed. The mechanism underlying this effect is not currently understood, and more molecular studies are needed to explain it. In addition to ferrostatin-1, there exits second- and third-generation ferrostatins that are more stable, exhibiting increased metabolic stability in the plasma. All of the third-generation ferrostatins are significantly protective against tissue injury, including acute kidney injury and IRI, in vivo (6).

Liproxstatin-1

Liproxstatin-1 is a potent ferroptosis inhibitor in Gpx4−/− cells that acts by preventing ROS accumulation. Liproxstatin-1 also inhibits ferroptosis in a mouse model of liver tissue injury induced by ischemia-reperfusion (57). A previous study reported that liproxstatin-1 decreases voltage-dependent anion channel 1 levels and restores GPX4 levels to protect against ischemia-reperfusion (109). Furthermore, post-treatment with liproxstatin-1 protects mitochondrial structural integrity (109). Although liproxstatin-1 decreases ROS levels, it does not affect Ca2+-induced mitochondrial permeability transition pore opening. Moreover, compared to fer-1, liproxstatin-1 has relatively stronger potency. Liproxstatin-1 also suppresses ferroptosis-inducing agents (FINs), comprising RSL3, erastin, and BODIPY 581/591 C11 oxidation (57). Hence, liproxstatin-1 may represent an extremely promising therapeutic drug in hepatic IRI.

α-Tocopherol

α-Tocopherol is a type of membrane and extracellular antioxidant, also called vitamin E. It helps to prevent free radicals from damaging hepatic cells and serves as an inhibitor of protein kinase C and lipid peroxidation that increases GSH levels (110,111). The efficacy of protecting liver cells during ischemia-reperfusion was shown in an animal experiment, which indicated that the group treated with α-tocopherol exhibited a significantly higher survival rate (110). Another study showed that pretreatment with high doses of α-tocopherol (30 and 300 mg/kg of body weight administered intramuscularly) enhanced ATP levels, attenuated lipid peroxidation, and prevented the loss of hepatic glutathione (111113). Furthermore, α-tocopherol has shown beneficial effects in both cold and warm IRI, decreases mitochondrial damage induced by oxidative stress (113,114). Low doses of α-tocopherol can also protect against liver cell death if combined with gadolinium chloride (GdCl3) or ischemic preconditioning (IPC) (110,115).

Ascorbic acid

Ascorbic acid, also known as vitamin C, is a vital antioxidant with strong inhibition of lipid peroxidation and ROS scavenging ability (116). Ascorbic acid conveys an electron(s) to ROS, providing site-specific protection against oxidative stress (117). The clotting factors can be used to access acute liver cell damage, and after treatment with ascorbic acid, the activity of clotting factors I, II, V, VII, and X showed significant improvement (116). Furthermore, ascorbic acid avoids the oxidative degradation of vitamin E (a type of antioxidant) by reacting directly with intermediates of tocopherol oxidation, as well as free radicals (118). Another study treated rats with ascorbic acid (100 mg/kg, i.v.) 5 min before sustained ischemia, and IPC and ascorbic acid synergistically attenuated mitochondrial damage during reperfusion due to decreased oxidant stress (119). During the process of ferroptosis in hepatic IRI, iron reacts with hydrogen peroxide to form hydroxyl-like radicals, hydroxyl and ferric ions, and these products are reduced by ascorbic acid, which inhibits iron-dependent Fenton reactions (120). In accordance with another study, it was clearly demonstrated that serum aminotransferase levels, lipid peroxidation, the loss of bile flow and cholate output were inhibited by ascorbic acid doses of 30 and 100 mg/kg but were promoted by a dose of 1,000 mg/kg (120). Therefore, low doses of ascorbic acid (30 and 100 mg/kg) have antioxidant effects, while high doses (1,000 mg/kg) have pro-oxidant effects; thus, the dose should be adjusted when ascorbic acid is applied. Moreover, the therapeutic window might be appropriate over a short time prior to or just at the beginning of reperfusion (121).

GSH

GSH is a thiol-containing compound, oxidizing sulfhydryl group of cysteine that exerts antioxidant effects (122). It is a substrate of GPX4, and GSH depletion results in inactivation of GPX4, contributing to ferroptosis by accumulation of ROS from lipid peroxidation (70). Therefore, pretreatment with GSH directly scavenges ROS (123). The GSH/GSSG redox system majorly regulates intracellular redox status (112) and giving GSH in advance may promote intracellular reduction response to prevent oxidative damage. However, there exit some administrative limitations. For instance, whether intracellular GSH is simply available and its ability to decrease GSSG and to what extent are not completely understood (124). A previous analysis demonstrated significant protection for hepatocytes in both warm and cold liver ischemia by intravenous glutathione administration (doses over 100 mol/h/kg) (125). It is assumed that GSH will become an additional therapeutic approach for ferroptosis during hepatic IRI.

α lipoic acid (ALA)

ALA is a natural compound that occurs in vivo. ALA is an antioxidant that provides protection against damage to the body's cells. Because of its antioxidant and oxidant-scavenging properties, ALA may protect the liver against oxidative injury (126), and this function has been corroborated in rat liver that underwent 90 min of warm ischemia (127). ALA was shown to significantly decrease levels of AST, increase ATP content, and lower apoptotic hepatocyte injury by improving expression of anti-apoptotic proteins to decrease hepatic injury (128). Moreover, ALA also protects against IRI caused by cirrhosis or steatosis due to improving cholinesterase activity in the serum (128). Furthermore, another study reported additional findings for ALA in the treatment of hepatic IRI, including decreasing levels of TNF-α and IL-1β, reversing myeloperoxidase activity (indicating increased neutrophil infiltration to the tissue), and maintaining regular morphology of the central vein and hepatocytes (126). As a powerful direct chain-breaking antioxidant, ALA strengthens the antioxidant potency of both ascorbate and vitamin E (129). Currently, ALA is a potential strategy to protect against hepatic IRI.

CVF

CVF has been affirmed to be beneficial for the protection of hepatocytes during ischemia-reperfusion in clinical settings (81,130). CVF is a stable complement inhibitor from cobra venom that binds to factor B as a structural and functional analogue of the complement component C3b, forming the bimolecular complex CVF/Bb through the cleavage of factor D (130). CVF/Bb is a C3/C5 convertase that simultaneously cleaves complement C3 and C5 (131133). Thus, continuous activation of C3 and C5 leads to the depletion of complement components and inhibits their activation. In a final analysis, through subsequently suppressing the release of inflammatory mediators, such as TNF-α and IL-1β, oxidant stress induced by Kupffer cells and hepatic cell apoptosis was decreased to attenuate hepatic injury (78,130). However, the window of time available for therapeutic intervention to block complement-mediated inflammatory responses and oxidant stress in hepatic cells should be reviewed due to the recovery of complement activity and regeneration in hepatocytes (78,130). As an anticomplement protein, CVF may represent a novel therapy to improve multiple organ injury induced by ischemia-reperfusion.

Zileuton

Zileuton inhibits the biosynthesis of leukotrienes (LTB4, LTC4, LTD4 and LTE4) because it is an active inhibitor of 5-lipoxygenase. Zileuton decreases glutamate-induced ROS accumulation, significantly inhibiting glutamate- and erastin-induced ferroptosis (134).

Gadolinium chloride

GdCl3, a rare earth metal, is a protective intervention in a rat hepatic reperfusion injury model that inhibits Kupffer cell activation (115,135). It has been shown that pretreatment with GdCl3 for hepatic IRI enhances the survival rate (115), decreases neutrophil infiltration and myeloperoxidase activity (136), and decreases platelet aggregation in cold-perfusion liver (137). Additional experiments have concluded that GdCl3 promotes recovery of hepatic function (135,138), prevents sinusoid endothelial cell apoptosis (137), inhibits the formation of free radicals, and attenuates lipid peroxidation (139,140). However, the use of GdCl3 may induce side effects (115), including significant loss of bile flow, altered hepatocellular integrity (increased serum enzyme activities), and inhibition of phagocytic activity in Kupffer cells. In addition, inhibition of Kupffer cells damages host defences (141,142) because the ability to clear bacterial lipopolysaccharides from the blood is deranged. Hence, the dose of GdCl3 should be kept as low as possible due to potential adverse effects, and it is necessary to monitor hepatic function when using GdCl3 for treatment (115).

Conclusions and future perspectives

Hepatic IRI is a complex process that involves various pathways and is complicated by a range of factors. Therefore, it is important to understand the pathophysiological pathways involved in liver damage during ischemia-reperfusion. This review discussed the detailed mechanism of ferroptosis, a novel and determinant type of regulated cell death, and concluded that it involves differential activation of various signal transduction pathways. Iron primarily participates in the relevant inflammatory response, stimulating the release of inflammatory and cytokines, and induces iron-dependent lipid peroxidation that generates oxidative damage to hepatic cells. Currently, ferroptosis still comprises some challenges, including the lack of a specific marker for animal studies and clinical settings (7). Additional studies about ferroptosis in hepatic IRI are required to better understand the presence of ferroptosis in the liver. This novel type of cell death in hepatic IRI will provide more precise therapeutic targets and will be advantageous for developing new clinical therapeutic methods.

Acknowledgements

Not applicable.

Funding

The present work was supported by the National Nature Science Foundation of China (grant no. 82060450), and the Nature Science Foundation of Jiangxi province of China (grant nos. 20181BAB205050, 20192BAB205072 and 20171BCB23086).

Availability of data and materials

Not applicable.

Authors' contributions

LL, GM and DH conceived and designed the study; LL prepared the original draft of the manuscript; LL and GM reviewed and edited the manuscript; and DH supervised the study and acquired the funding. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AA

arachidonic acid

ACSL4

acyl-CoA synthetase long-chain family 4

AdA

adrenoyl

ALA

alpha-lipoic acid

ALT

alanine aminotransferase

ASRK1

apoptosis signal-regulating kinase 1

ATP

adenosine triphosphate

BMP

bone morphogenetic protein

CAT

catalase, Ccl2, chemokine (C-C motif) ligand 2

Cxcl1

chemokine (C-X-C motif) ligand 1

Cxcl2

chemokine (C-X-C motif) ligand 2

CYP

cytochrome P450

CVF

cobra venom factor

DISC

death-inducing signalling complex

DMT1

divalent metal transporter 1

ERK

extracellular signal-regulated kinase

Fas

Fas cell surface death receptor

Fer-1

ferrostatin-1

FN

fibronectin

FINs

ferroptosis-inducing agents

GdCl3

gadolinium chloride

GP

glycogen phosphorylase

GPX4

glutathione peroxidase 4

GSH

glutathione

GSSG

oxidized glutathione

HSPB1

shock protein family B member 1

IL

interleukin

INOS

inducible nitric oxide synthase

IP

inositol phosphate

IPC

ischemic preconditioning

IRI

ischemia-reperfusion injury

JAK

Janus kinase

JNK

c-Jun N-terminal kinases

LOOH

alkyl radical

LOXs

lipoxygenases

LPCAT3

lysophosphatidylcholine acyltransferase 3

LT

leukotrienes

MAPK

mitogen-activated protein kinase

MEK

mitogen-activated protein kinase

MLKL

mixed-lineage kinase domain-like

NADPH

nicotinamide adenine dinucleotide phosphate

NF-κB

transcription factors nuclear factor κB

NLRP3

pyrin domain containing 3

NOX

nitric oxide

PE-OOH

phosphatidylethanolamine-OOH

PHKG2

phosphorylase kinase G2

PMNs

polymorphonuclear neutrophils

PUFAs

polyunsaturated fatty acids

RCD

regulated cell death

RIPK1

receptor-interacting serine/threonine-protein kinase 1

ROS

reactive oxygen species

RSL3

Ras-selective lethal small molecule

SOD

superoxide dismutase

STAT3

signal transducer and activator of transcription 3

STEAP3

six-transmembrane epithelial antigen of prostate 3

TfR

transferrin receptor

TLR4

Toll-like receptor 4

TNF-α

tumor necrosis factor-α

TRAF-2

TNF-receptor-associated factor 2

References

1 

Dickson KB and Zhou J: Role of reactive oxygen species and iron in host defence against infection. Front Biosci (Landmark Ed). 25:1600–1616. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, et al: Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 13:81–90. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Valko M, Jomova K, Rhodes CJ, Kuča K and Musílek K: Redox- and non-redox-metal-induced formation of free radicals and their role in human disease. Arch Toxicol. 90:1–37. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Cao JY and Dixon SJ: Mechanisms of ferroptosis. Cell Mol Life Sci. 73:2195–2209. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, Kang R and Tang D: Ferroptosis: Process and function. Cell Death Differ. 23:369–379. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Macías-Rodríguez RU, Inzaugarat ME, Ruiz-Margáin A, Nelson LJ, Trautwein C and Cubero FJ: Reclassifying hepatic cell death during liver damage: Ferroptosis-A novel form of non-apoptotic cell death? Int J Mol Sci. 21:16512020. View Article : Google Scholar

8 

Yamada N, Karasawa T, Wakiya T, Sadatomo A, Ito H, Kamata R, Watanabe S, Komada T, Kimura H, Sanada Y, et al: Iron overload as a risk factor for hepatic ischemia-reperfusion injury in liver transplantation: Potential role of ferroptosis. Am J Transplant. 20:1606–1618. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Ploeg RJ, D'Alessandro AM, Knechtle SJ, Stegall MD, Pirsch JD, Hoffmann RM, Sasaki T, Sollinger HW, Belzer FO and Kalayoglu M: Risk factors for primary dysfunction after liver transplantation-a multivariate analysis. Transplantation. 55:807–813. 1993. View Article : Google Scholar : PubMed/NCBI

10 

Zhai Y, Petrowsky H, Hong JC, Busuttil RW and Kupiec-Weglinski JW: Ischaemia-reperfusion injury in liver transplantation-from bench to bedside. Nat Rev Gastroenterol Hepatol. 10:79–89. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Ikeda T, Yanaga K, Kishikawa K, Kakizoe S, Shimada M and Sugimachi K: Ischemic injury in liver transplantation: Difference in injury sites between warm and cold ischemia in rats. Hepatology. 16:454–461. 1992. View Article : Google Scholar : PubMed/NCBI

12 

Cannistra M, Ruggiero M, Zullo A, Gallelli G, Serafini S, Maria M, Naso A, Grande R, Serra R and Nardo B: Hepatic ischemia reperfusion injury: A systematic review of literature and the role of current drugs and biomarkers. Int J Surg. 33 (Suppl 1):S57–S70. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Zhai Y, Busuttil RW and Kupiec-Weglinski JW: Liver ischemia and reperfusion injury: New insights into mechanisms of innate-adaptive immune-mediated tissue inflammation. Am J Transplant. 11:1563–1569. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Duarte S, Shen XD, Fondevila C, Busuttil RW and Coito AJ: Fibronectin-α4β1 interactions in hepatic cold ischemia and reperfusion injury: Regulation of MMP-9 and MT1-MMP via the p38 MAPK pathway. Am J Transplant. 12:2689–2699. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Decuypere JP, Ceulemans LJ, Agostinis P, Monbaliu D, Naesens M, Pirenne J and Jochmans I: Autophagy and the Kidney: Implications for ischemia-reperfusion injury and therapy. Am J Kidney Dis. 66:699–709. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Pu T, Liao XH, Sun H, Guo H, Jiang X, Peng JB, Zhang L and Liu Q: Augmenter of liver regeneration regulates autophagy in renal ischemia-reperfusion injury via the AMPK/mTOR pathway. Apoptosis. 22:955–969. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Zhong X, Xiao Q, Liu Z, Wang W, Lai CH, Yang W, Yue P, Ye Q and Xiao J: TAK242 suppresses the TLR4 signaling pathway and ameliorates DCD liver IRI in rats. Mol Med Rep. 20:2101–2110. 2019.PubMed/NCBI

18 

Oliveira THC, Marques PE, Proost P and Teixeira MMM: Neutrophils: A cornerstone of liver ischemia and reperfusion injury. Lab Invest. 98:51–62. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Wang L, Zhang Z, Li M, Wang F, Jia Y, Zhang F, Shao J, Chen A and Zheng S: P53-dependent induction of ferroptosis is required for artemether to alleviate carbon tetrachloride-induced liver fibrosis and hepatic stellate cell activation. Iubmb Life. 71:45–56. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Li L, Tan J, Miao Y, Lei P and Zhang Q: ROS and autophagy: Interactions and molecular regulatory mechanisms. Cell Mol Neurobiol. 35:615–621. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Datta G, Fuller BJ and Davidson BR: Molecular mechanisms of liver ischemia reperfusion injury: Insights from transgenic knockout models. World J Gastroenterol. 19:1683–1698. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Papadopoulos D, Siempis T, Theodorakou E and Tsoulfas G: Hepatic ischemia and reperfusion injury and trauma: Current concepts. Arch Trauma Res. 2:63–70. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Dhanasekaran DN and Reddy EP: JNK signaling in apoptosis. Oncogene. 27:6245–6251. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Wanner GA, Ertel W, Muller P, Hofer Y, Leiderer R, Menger MD and Messmer K: Liver ischemia and reperfusion induces a systemic inflammatory response through Kupffer cell activation. Shock. 5:34–40. 1996. View Article : Google Scholar : PubMed/NCBI

25 

Lentsch AB, Yoshidome H, Kato A, Warner RL, Cheadle WG, Ward PA and Edwards MJ: Requirement for interleukin-12 in the pathogenesis of warm hepatic ischemia/reperfusion injury in mice. Hepatology. 30:1448–1453. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Jaeschke H: Reactive oxygen and mechanisms of inflammatory liver injury. J Gastroenterol Hepatol. 15:718–724. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Fan C, Zwacka RM and Engelhardt JF: Therapeutic approaches for ischemia/reperfusion injury in the liver. J Mol Med (Berl). 77:577–592. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Bauer M and Bauer I: Heme oxygenase-1: Redox regulation and role in the hepatic response to oxidative stress. Antioxid Redox Signal. 4:749–758. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Rensing H, Jaeschke H, Bauer I, Patau C, Datene V, Pannen BH and Bauer M: Differential activation pattern of redox-sensitive transcription factors and stress-inducible dilator systems heme oxygenase-1 and inducible nitric oxide synthase in hemorrhagic and endotoxic shock. Crit Care Med. 29:1962–1971. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Jaeschke H, Ho YS, Fisher MA, Lawson JA and Farhood A: Glutathione peroxidase-deficient mice are more susceptible to neutrophil-mediated hepatic parenchymal cell injury during endotoxemia: Importance of an intracellular oxidant stress. Hepatology. 29:443–450. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Essani NA, Fisher MA and Jaeschke H: Inhibition of NF-kappa B activation by dimethyl sulfoxide correlates with suppression of TNF-alpha formation, reduced ICAM-1 gene transcription, and protection against endotoxin-induced liver injury. Shock. 7:90–96. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Li JM and Shah AM: Differential NADPH-versus NADH-dependent superoxide production by phagocyte-type endothelial cell NADPH oxidase. Cardiovasc Res. 52:477–486. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Ozaki M, Deshpande SS, Angkeow P, Bellan J, Lowenstein CJ, Dinauer MC, Goldschmidt-Clermont PJ and Irani K: Inhibition of the Rac1 GTPase protects against nonlethal ischemia/reperfusion-induced necrosis and apoptosis in vivo. FASEB J. 14:418–429. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Li Y, Qian L and Yuan J: Small molecule probes for cellular death machines. Curr Opin Chem Biol. 39:74–82. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Degterev A and Linkermann A: Generation of small molecules to interfere with regulated necrosis. Cell Mol Life Sci. 73:2251–2267. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Dixon SJ and Stockwell BR: The role of iron and reactive oxygen species in cell death. Nat Chem Biol. 10:9–17. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Pignatello JJ, Oliveros E and MacKay A: Advanced oxidation processes for organic contaminant destruction based on the Fenton reaction and related chemistry. J Crit Rev Environmental Sci Technol. 36:1–84. 2006. View Article : Google Scholar

38 

Wang H, An P, Xie E, Wu Q, Fang X, Gao H, Zhang Z, Li Y, Wang X, Zhang J, et al: Characterization of ferroptosis in murine models of hemochromatosis. Hepatology. 66:449–465. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Sun X, Ou Z, Xie M, Kang R, Fan Y, Niu X, Wang H, Cao L and Tang D: HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene. 34:5617–5625. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Chen H, Zheng C, Zhang Y, Chang YZ, Qian ZM and Shen X: Heat shock protein 27 downregulates the transferrin receptor 1-mediated iron uptake. Int J Biochem Cell Biol. 38:1402–1416. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Arrigo AP, Virot S, Chaufour S, Firdaus W, Kretz-Remy C and Diaz-Latoud C: Hsp27 consolidates intracellular redox homeostasis by upholding glutathione in its reduced form and by decreasing iron intracellular levels. Antioxid Redox Signal. 7:414–422. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Lei P, Bai T and Sun Y: Mechanisms of ferroptosis and relations with regulated cell death: A review. Front Physiol. 10:1392019. View Article : Google Scholar : PubMed/NCBI

43 

Krieg P and Furstenberger G: The role of lipoxygenases in epidermis. Biochim Biophys Acta. 1841:390–400. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Haeggstrom JZ and Funk CD: Lipoxygenase and leukotriene pathways: Biochemistry, biology, and roles in disease. Chem Rev. 111:5866–5898. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Choi J, Chon JK, Kim S and Shin W: Conformational flexibility in mammalian 15S-lipoxygenase: Reinterpretation of the crystallographic data. Proteins. 70:1023–1032. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Shintoku R, Takigawa Y, Yamada K, Kubota C, Yoshimoto Y, Takeuchi T, Koshiishi I and Torii S: Lipoxygenase-mediated generation of lipid peroxides enhances ferroptosis induced by erastin and RSL3. Cancer Sci. 108:2187–2194. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS and Stockwell BR: Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 113:E4966–E4975. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Reddy PH and Beal MF: Are mitochondria critical in the pathogenesis of Alzheimer's disease? Brain Res Brain Res Rev. 49:618–632. 2005. View Article : Google Scholar : PubMed/NCBI

49 

Ademowo OS, Dias HKI, Burton DGA and Griffiths HR: Lipid (per) oxidation in mitochondria: An emerging target in the ageing process? Biogerontology. 18:859–879. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Wong-Ekkabut J, Xu Z, Triampo W, Tang IM, Tieleman DP and Monticelli L: Effect of lipid peroxidation on the properties of lipid bilayers: A molecular dynamics study. Biophys J. 93:4225–4236. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Simoncini C, Orsucci D, Caldarazzo Ienco E, Siciliano G, Bonuccelli U and Mancuso M: Alzheimer's pathogenesis and its link to the mitochondrion. Oxid Med Cell Longev. 2015:8039422015. View Article : Google Scholar : PubMed/NCBI

52 

Eckl PM, Ortner A and Esterbauer H: Genotoxic properties of 4-hydroxyalkenals and analogous aldehydes. Mutat Res. 290:183–192. 1993. View Article : Google Scholar : PubMed/NCBI

53 

Siems W and Grune T: Intracellular metabolism of 4-hydroxynonenal. Mol Aspects Med. 24:167–175. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Stockwell BR, Friedmann AJ, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascon S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Tuo QZ, Lei P, Jackman KA, Li XL, Xiong H, Li XL, Liuyang ZY, Roisman L, Zhang ST, Ayton S, et al: Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol Psychiatry. 22:1520–1530. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Skouta R, Dixon SJ, Wang J, Dunn DE, Orman M, Shimada K, Rosenberg PA, Lo DC, Weinberg JM, Linkermann A and Stockwell BR: Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J Am Chem Soc. 136:4551–4556. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Friedmann AJ, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, et al: Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 16:1180–1191. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Netea MG, van de Veerdonk FL, van der Meer JW, Dinarello CA and Joosten LA: Inflammasome-independent regulation of IL-1-family cytokines. Annu Rev Immunol. 33:49–77. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Kiziltas S: Toll-like receptors in pathophysiology of liver diseases. World J Hepatol. 8:1354–1369. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Usui F, Shirasuna K, Kimura H, Tatsumi K, Kawashima A, Karasawa T, Yoshimura K, Aoki H, Tsutsui H, Noda T, et al: Inflammasome activation by mitochondrial oxidative stress in macrophages leads to the development of angiotensin II-induced aortic aneurysm. Arterioscler Thromb Vasc Biol. 35:127–136. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Usui F, Shirasuna K, Kimura H, Tatsumi K, Kawashima A, Karasawa T, Hida S, Sagara J, Taniguchi S and Takahashi M: Critical role of caspase-1 in vascular inflammation and development of atherosclerosis in Western diet-fed apolipoprotein E-deficient mice. Biochem Biophys Res Commun. 425:162–168. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, et al: Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 123:594–604. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Sadatomo A, Inoue Y, Ito H, Karasawa T, Kimura H, Watanabe S, Mizushina Y, Nakamura J, Kamata R, Kasahara T, et al: Interaction of neutrophils with macrophages promotes IL-1β maturation and contributes to hepatic ischemia-reperfusion injury. J Immunol. 199:3306–3315. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Skaar EP: The battle for iron between bacterial pathogens and their vertebrate hosts. PLoS Pathog. 6:e10009492010. View Article : Google Scholar : PubMed/NCBI

65 

Wang L, Harrington L, Trebicka E, Shi HN, Kagan JC, Hong CC, Lin HY, Babitt JL and Cherayil BJ: Selective modulation of TLR4-activated inflammatory responses by altered iron homeostasis in mice. J Clin Invest. 119:3322–3328. 2009.PubMed/NCBI

66 

Weiss G, Werner-Felmayer G, Werner ER, Grunewald K, Wachter H and Hentze MW: Iron regulates nitric oxide synthase activity by controlling nuclear transcription. J Exp Med. 180:969–976. 1994. View Article : Google Scholar : PubMed/NCBI

67 

Bubici C, Papa S, Dean K and Franzoso G: Mutual cross-talk between reactive oxygen species and nuclear factor-kappa B: Molecular basis and biological significance. Oncogene. 25:6731–6748. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Ganz T and Nemeth E: Hepcidin and iron homeostasis. Biochim Biophys Acta. 1823:1434–1443. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Wunderer F, Traeger L, Sigurslid HH, Meybohm P, Bloch DB and Malhotra R: The role of hepcidin and iron homeostasis in atherosclerosis. Pharmacol Res. 153:1046642020. View Article : Google Scholar : PubMed/NCBI

70 

Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Jornot L, Petersen H and Junod AF: Hydrogen peroxide-induced DNA damage is independent of nuclear calcium but dependent on redox-active ions. Biochem J. 335:85–94. 1998. View Article : Google Scholar : PubMed/NCBI

72 

Mills EM, Takeda K, Yu ZX, Ferrans V, Katagiri Y, Jiang H, Lavigne MC, Leto TL and Guroff G: Nerve growth factor treatment prevents the increase in superoxide produced by epidermal growth factor in PC12 cells. J Biol Chem. 273:22165–22168. 1998. View Article : Google Scholar : PubMed/NCBI

73 

Hurst R, Bao Y, Jemth P, Mannervik B and Williamson G: Phospholipid hydroperoxide glutathione peroxidase activity of rat class theta glutathione transferase T2-2. Biochem Soc Trans. 25:S5591997. View Article : Google Scholar : PubMed/NCBI

74 

Yin GY, Yin YF and He XF: Effect of Zhuchun pill on immunity and endocrine function of elderly with kidney-yang deficiency. Zhongguo Zhong Xi Yi Jie He Za Zhi. 15:601–603. 1995.(In Chinese). PubMed/NCBI

75 

Lee YJ, Galoforo SS, Berns CM, Chen JC, Davis BH, Sim JE, Corry PM and Spitz DR: Glucose deprivation-induced cytotoxicity and alterations in mitogen-activated protein kinase activation are mediated by oxidative stress in multidrug-resistant human breast carcinoma cells. J Biol Chem. 273:5294–5299. 1998. View Article : Google Scholar : PubMed/NCBI

76 

Bae YS, Kang SW, Seo MS, Baines IC, Tekle E, Chock PB and Rhee SG: Epidermal growth factor (EGF)-induced generation of hydrogen peroxide. Role in EGF receptor-mediated tyrosine phosphorylation. J Biol Chem. 272:217–221. 1997. View Article : Google Scholar : PubMed/NCBI

77 

Jaeschke H and Farhood A: Neutrophil and Kupffer cell-induced oxidant stress and ischemia-reperfusion injury in rat liver. Am J Physiol. 260:G355–G362. 1991.PubMed/NCBI

78 

Straatsburg IH, Boermeester MA, Wolbink GJ, van Gulik TM, Gouma DJ, Frederiks WM and Hack CE: Complement activation induced by ischemia-reperfusion in humans: A study in patients undergoing partial hepatectomy. J Hepatol. 32:783–791. 2000. View Article : Google Scholar : PubMed/NCBI

79 

Bajt ML, Farhood A and Jaeschke H: Effects of CXC chemokines on neutrophil activation and sequestration in hepatic vasculature. Am J Physiol Gastrointest Liver Physiol. 281:G1188–G1195. 2001. View Article : Google Scholar : PubMed/NCBI

80 

Witthaut R, Farhood A, Smith CW and Jaeschke H: Complement and tumor necrosis factor-alpha contribute to Mac-1 (CD11b/CD18) up-regulation and systemic neutrophil activation during endotoxemia in vivo. J Leukoc Biol. 55:105–111. 1994. View Article : Google Scholar : PubMed/NCBI

81 

Jaeschke H, Farhood A, Bautista AP, Spolarics Z and Spitzer JJ: Complement activates Kupffer cells and neutrophils during reperfusion after hepatic ischemia. Am J Physiol. 264:G801–G809. 1993.PubMed/NCBI

82 

Yeh CG, Marsh HJ Jr, Carson GR, Berman L, Concino MF, Scesney SM, Kuestner RE, Skibbens R, Donahue KA and Ip SH: Recombinant soluble human complement receptor type 1 inhibits inflammation in the reversed passive arthus reaction in rats. J Immunol. 146:250–256. 1991.PubMed/NCBI

83 

Weisman HF, Bartow T, Leppo MK, Marsh HJ, Carson GR, Concino MF, Boyle MP, Roux KH, Weisfeldt ML and Fearon DT: Soluble human complement receptor type 1: In vivo inhibitor of complement suppressing post-ischemic myocardial inflammation and necrosis. Science. 249:146–151. 1990. View Article : Google Scholar : PubMed/NCBI

84 

Rymsa B, Wang JF and de Groot H: O2−. Release by activated Kupffer cells upon hypoxia-reoxygenation. Am J Physiol. 261:G602–G607. 1991.PubMed/NCBI

85 

Marnett LJ: Lipid peroxidation-DNA damage by malondialdehyde. Mutat Res. 424:83–95. 1999. View Article : Google Scholar : PubMed/NCBI

86 

Arslan M, Metin CF, Kucuk A, Ozturk L and Yaylak F: Dexmedetomidine protects against lipid peroxidation and erythrocyte deformability alterations in experimental hepatic ischemia reperfusion injury. Libyan J Med. 7:2012.doi: 10.3402/ljm.v7i0.18185. View Article : Google Scholar : PubMed/NCBI

87 

Kuypers FA: Red cell membrane damage. J Heart Valve Dis. 7:387–395. 1998.PubMed/NCBI

88 

Sivilotti ML: Oxidant stress and haemolysis of the human erythrocyte. Toxicol Rev. 23:169–188. 2004. View Article : Google Scholar : PubMed/NCBI

89 

Therond P, Bonnefont-Rousselot D, Davit-Spraul A, Conti M and Legrand A: Biomarkers of oxidative stress: An analytical approach. Curr Opin Clin Nutr Metab Care. 3:373–384. 2000. View Article : Google Scholar : PubMed/NCBI

90 

Stahl W, Junghans A, de Boer B, Driomina ES, Briviba K and Sies H: Carotenoid mixtures protect multilamellar liposomes against oxidative damage: Synergistic effects of lycopene and lutein. FEBS Lett. 427:305–308. 1998. View Article : Google Scholar : PubMed/NCBI

91 

Beaudeux JL, Gardes-Albert M, Delattre J, Legrand A, Rousselet F and Peynet J: Resistance of lipoprotein(a) to lipid peroxidation induced by oxygenated free radicals produced by gamma radiolysis: A comparison with low-density lipoprotein. Biochem J. 314:277–284. 1996. View Article : Google Scholar : PubMed/NCBI

92 

Jankowska R, Passowicz-Muszynska E, Banas T, Marcinkowska A and Medrala W: The influence of vitamin A on production of oxygen free radicals and activity of granulocyte catalase in patients with chronic bronchitis. Pneumonol Alergol Pol. 62:628–633. 1994.(In Polish). PubMed/NCBI

93 

Malhi H, Guicciardi ME and Gores GJ: Hepatocyte death: A clear and present danger. Physiol Rev. 90:1165–1194. 2010. View Article : Google Scholar : PubMed/NCBI

94 

Kischkel FC, Hellbardt S, Behrmann I, Germer M, Pawlita M, Krammer PH and Peter ME: Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO. 14:5579–5588. 1995. View Article : Google Scholar

95 

Stefan JR and Guy SS: The apoptosome: Signalling platform of cell death. Nat Rev Mol Cell Bio. 8:405–413. 2007. View Article : Google Scholar

96 

Yong-Ling PO, Douglas RG, Zhenyue H and Tak WM: Cytochrome c: Functions beyond respiration. Nat Rev Mol Cell Bio. 9:532–542. 2008. View Article : Google Scholar

97 

Nicholas SW, Vishva D and Avi A: Death receptor signal transducers: Nodes of coordination in immune signaling networks. Nat Immunol. 10:348–355. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Susan E: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

99 

Krysko DV, Vanden BT, D'Herde K and Vandenabeele P: Apoptosis and necrosis: Detection, discrimination and phagocytosis. Methods. 44:205–221. 2008. View Article : Google Scholar : PubMed/NCBI

100 

Ricardo W, Andrew O, Helen MB and Douglas RG: Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 18:127–136. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Ueno T and Komatsu M: Autophagy in the liver: Functions in health and disease. Nat Rev Gastroenterol Hepatol. 14:170–184. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Pierre ER, Dominique CH, Richard M, Claire F, Gérard F, Didier L, Éric OD, Pierre B, Dominique V and Fran OD: Acute liver cell damage in patients with anorexia nervosa: A possible role of starvation-induced hepatocyte autophagy. Gastroenterology. 135:840–848.e1-e3. 2008. View Article : Google Scholar : PubMed/NCBI

103 

Donna D and Sharad K: Autophagy-dependent cell death. Cell Death Differ. 26:605–616. 2019. View Article : Google Scholar : PubMed/NCBI

104 

Santana-Codina N and Mancias JD: The role of NCOA4-mediated ferritinophagy in health and disease. Pharmaceuticals (Basel). 11:1142018. View Article : Google Scholar

105 

Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ, Kang R and Tang D: Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 12:1425–1428. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Veitch K, Maisin L and Hue L: Trimetazidine effects on the damage to mitochondrial functions caused by ischemia and reperfusion. Am J Cardiol. 76:B25–B30. 1995. View Article : Google Scholar

107 

Guarnieri C and Muscari C: Effect of trimetazidine on mitochondrial function and oxidative damage during reperfusion of ischemic hypertrophied rat myocardium. Pharmacology. 46:324–331. 1993. View Article : Google Scholar : PubMed/NCBI

108 

Miotto G, Rossetto M, Di Paolo ML, Orian L, Venerando R, Roveri A, Vuckovic AM, Bosello TV, Zaccarin M, Zennaro L, et al: Insight into the mechanism of ferroptosis inhibition by ferrostatin-1. Redox Biol. 28:1013282020. View Article : Google Scholar : PubMed/NCBI

109 

Feng Y, Madungwe NB, Imam AA, Tombo N and Bopassa JC: Liproxstatin-1 protects the mouse myocardium against ischemia/reperfusion injury by decreasing VDAC1 levels and restoring GPX4 levels. Biochem Biophys Res Commun. 520:606–611. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Giakoustidis D, Papageorgiou G, Iliadis S, Giakoustidis A, Kostopoulou E, Kontos N, Botsoglou E, Tsantilas D, Papanikolaou V and Takoudas D: The protective effect of alpha-tocopherol and GdCl3 against hepatic ischemia/reperfusion injury. Surg Today. 36:450–456. 2006. View Article : Google Scholar : PubMed/NCBI

111 

Masaki H, Okano Y, Ochiai Y, Obayashi K, Akamatsu H and Sakurai H: Alpha-tocopherol increases the intracellular glutathione level in HaCaT keratinocytes. Free Radic Res. 36:705–709. 2002. View Article : Google Scholar : PubMed/NCBI

112 

Lee WY and Lee SM: Protective effects of alpha-tocopherol and ischemic preconditioning on hepatic reperfusion injury. Arch Pharm Res. 28:1392–1399. 2005. View Article : Google Scholar : PubMed/NCBI

113 

Giakoustidis D, Papageorgiou G, Iliadis S, Kontos N, Kostopoulou E, Papachrestou A, Tsantilas D, Spyridis C, Takoudas D, Botsoglou N, et al: Intramuscular administration of very high dose of alpha-tocopherol protects liver from severe ischemia/reperfusion injury. World J Surg. 26:872–877. 2002. View Article : Google Scholar : PubMed/NCBI

114 

Gondolesi GE, Lausada N, Schinella G, Semplici AM, Vidal MS, Luna GC, Toledo J, de Buschiazzo PM and Raimondi JC: Reduction of ischemia-reperfusion injury in parenchymal and nonparenchymal liver cells by donor treatment with DL-alpha-tocopherol prior to organ harvest. Transplant Proc. 34:1086–1091. 2002. View Article : Google Scholar : PubMed/NCBI

115 

Ruttinger D, Vollmar B, Wanner GA and Messmer K: In vivo assessment of hepatic alterations following gadolinium chloride-induced Kupffer cell blockade. J Hepatol. 25:960–967. 1996. View Article : Google Scholar : PubMed/NCBI

116 

Cerwenka H, Khoschsorur G, Bacher H, Werkgartner G, El-Shabrawi A, Quehenberger F, Rabl H and Mischinger HJ: Normothermic liver ischemia and antioxidant treatment during hepatic resections. Free Radic Res. 30:463–469. 1999. View Article : Google Scholar : PubMed/NCBI

117 

Kohen R and Nyska A: Oxidation of biological systems: Oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol. 30:620–650. 2002. View Article : Google Scholar : PubMed/NCBI

118 

Packer JE, Slater TF and Willson RL: Direct observation of a free radical interaction between vitamin E and vitamin C. Nature. 278:737–738. 1979. View Article : Google Scholar : PubMed/NCBI

119 

Lee WY, Lee JS and Lee SM: Protective effects of combined ischemic preconditioning and ascorbic acid on mitochondrial injury in hepatic ischemia/reperfusion. J Surg Res. 142:45–52. 2007. View Article : Google Scholar : PubMed/NCBI

120 

Seo MY and Lee SM: Protective effect of low dose of ascorbic acid on hepatobiliary function in hepatic ischemia/reperfusion in rats. J Hepatol. 36:72–77. 2002. View Article : Google Scholar : PubMed/NCBI

121 

Rabl H, Khoschsorur G and Petek W: Antioxidative vitamin treatment: Effect on lipid peroxidation and limb swelling after revascularization operations. World J Surg. 19:738–744. 1995. View Article : Google Scholar : PubMed/NCBI

122 

Bilzer M and Lauterburg BH: Effects of hypochlorous acid and chloramines on vascular resistance, cell integrity, and biliary glutathione disulfide in the perfused rat liver: Modulation by glutathione. J Hepatol. 13:84–89. 1991. View Article : Google Scholar : PubMed/NCBI

123 

Winterbourn CC and Metodiewa D: The reaction of superoxide with reduced glutathione. Arch Biochem Biophys. 314:284–290. 1994. View Article : Google Scholar : PubMed/NCBI

124 

Schauer RJ, Gerbes AL, Vonier D, Meissner H, Michl P, Leiderer R, Schildberg FW, Messmer K and Bilzer M: Glutathione protects the rat liver against reperfusion injury after prolonged warm ischemia. Ann Surg. 239:220–231. 2004. View Article : Google Scholar : PubMed/NCBI

125 

Cotgreave IA: N-acetylcysteine: Pharmacological considerations and experimental and clinical applications. Adv Pharmacol. 38:205–227. 1997. View Article : Google Scholar : PubMed/NCBI

126 

Dulundu E, Ozel Y, Topaloglu U, Sehirli O, Ercan F, Gedik N and Sener G: Alpha-lipoic acid protects against hepatic ischemia-reperfusion injury in rats. Pharmacology. 79:163–170. 2007. View Article : Google Scholar : PubMed/NCBI

127 

Müller C, Dunschede F, Koch E, Vollmar AM and Kiemer AK: Alpha-lipoic acid preconditioning reduces ischemia-reperfusion injury of the rat liver via the PI3-kinase/Akt pathway. Am J Physiol Gastrointest Liver Physiol. 285:G769–G778. 2003. View Article : Google Scholar : PubMed/NCBI

128 

Dunschede F, Erbes K, Kircher A, Westermann S, Seifert J, Schad A, Oliver K, Kiemer AK and Theodor J: Reduction of ischemia reperfusion injury after liver resection and hepatic inflow occlusion by alpha-lipoic acid in humans. World J Gastroenterol. 12:6812–6817. 2006. View Article : Google Scholar : PubMed/NCBI

129 

Kagan VE, Shvedova A, Serbinova E, Khan S, Swanson C, Powell R and Packer L: Dihydrolipoic acid-a universal antioxidant both in the membrane and in the aqueous phase. Reduction of peroxyl, ascorbyl and chromanoxyl radicals. Biochem Pharmacol. 44:1637–1649. 1992. View Article : Google Scholar : PubMed/NCBI

130 

Wang B, Xu H, Li J, Gao HM, Xing YH, Lin Z, Li HJ, Wang YQ and Cao SH: Complement depletion with cobra venom factor alleviates acute hepatic injury induced by ischemiareperfusion. Mol Med Rep. 18:4523–4529. 2018.PubMed/NCBI

131 

Mao YF, Yu QH, Zheng XF, Liu K, Liang WQ, Wang YW, Deng XM and Jiang L: Pre-treatment with Cobra venom factor alleviates acute lung injury induced by intestinal ischemia-reperfusion in rats. Eur Rev Med Pharmacol Sci. 17:2207–2217. 2013.PubMed/NCBI

132 

Vogel CW, Finnegan PW and Fritzinger DC: Humanized cobra venom factor: Structure, activity, and therapeutic efficacy in preclinical disease models. Mol Immunol. 61:191–203. 2014. View Article : Google Scholar : PubMed/NCBI

133 

Vogel CW and Fritzinger DC: Cobra venom factor: Structure, function, and humanization for therapeutic complement depletion. Toxicon. 56:1198–1222. 2010. View Article : Google Scholar : PubMed/NCBI

134 

Liu Y, Wang W, Li Y, Xiao Y, Cheng J and Jia J: The 5-lipoxygenase inhibitor zileuton confers neuroprotection against glutamate oxidative damage by inhibiting ferroptosis. Biol Pharm Bull. 38:1234–1239. 2015. View Article : Google Scholar : PubMed/NCBI

135 

Kukan M, Vajdova K, Horecky J, Nagyova A, Mehendale HM and Trnovec T: Effects of blockade of Kupffer cells by gadolinium chloride on hepatobiliary function in cold ischemia-reperfusion injury of rat liver. Hepatology. 26:1250–1257. 1997. View Article : Google Scholar : PubMed/NCBI

136 

Cutrin JC, Boveris A, Zingaro B, Corvetti G and Poli G: In situ determination by surface chemiluminescence of temporal relationships between evolving warm ischemia-reperfusion injury in rat liver and phagocyte activation and recruitment. Hepatology. 31:622–632. 2000. View Article : Google Scholar : PubMed/NCBI

137 

Sindram D, Porte RJ, Hoffman MR, Bentley RC and Clavien PA: Synergism between platelets and leukocytes in inducing endothelial cell apoptosis in the cold ischemic rat liver: A Kupffer cell-mediated injury. FASEB J. 15:1230–1232. 2001. View Article : Google Scholar : PubMed/NCBI

138 

Vajdova K, Smrekova R, Kukan M, Jakubovsky J, van Rooijen N, Horecky J, Lutterova M and Wsolova L: Endotoxin-induced aggravation of preservation-reperfusion injury of rat liver and its modulation. J Hepatol. 32:112–120. 2000. View Article : Google Scholar : PubMed/NCBI

139 

Giakoustidis DE, Iliadis S, Tsantilas D, Papageorgiou G, Kontos N, Kostopoulou E, Botsoglou NA, Gerasimidis T and Dimitriadou A: Blockade of Kupffer cells by gadolinium chloride reduces lipid peroxidation and protects liver from ischemia/reperfusion injury. Hepatogastroenterology. 50:1587–1592. 2003.PubMed/NCBIPubMed/NCBI

140 

Bremer C, Bradford BU, Hunt KJ, Knecht KT, Connor HD, Mason RP and Thurman RG: Role of Kupffer cells in the pathogenesis of hepatic reperfusion injury. Am J Physiol. 267:G630–G636. 1994.PubMed/NCBI

141 

Dixon LJ, Barnes M, Tang H, Pritchard MT and Nagy LE: Kupffer cells in the liver. Compr Physiol. 3:785–797. 2013.PubMed/NCBI

142 

Callery MP, Kamei T and Flye MW: Kupffer cell blockade increases mortality during intra-abdominal sepsis despite improving systemic immunity. Arch Surg. 125:36–41. 1990. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 23 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luo L, Mo G and Huang D: Ferroptosis in hepatic ischemia‑reperfusion injury: Regulatory mechanisms and new methods for therapy (Review). Mol Med Rep 23: 225, 2021
APA
Luo, L., Mo, G., & Huang, D. (2021). Ferroptosis in hepatic ischemia‑reperfusion injury: Regulatory mechanisms and new methods for therapy (Review). Molecular Medicine Reports, 23, 225. https://doi.org/10.3892/mmr.2021.11864
MLA
Luo, L., Mo, G., Huang, D."Ferroptosis in hepatic ischemia‑reperfusion injury: Regulatory mechanisms and new methods for therapy (Review)". Molecular Medicine Reports 23.3 (2021): 225.
Chicago
Luo, L., Mo, G., Huang, D."Ferroptosis in hepatic ischemia‑reperfusion injury: Regulatory mechanisms and new methods for therapy (Review)". Molecular Medicine Reports 23, no. 3 (2021): 225. https://doi.org/10.3892/mmr.2021.11864