Open Access

Recent advances in potential therapeutic targets of ferroptosis‑associated pathways for the treatment of stroke (Review)

  • Authors:
    • Hao Dong
    • Ya-Ping Ma
    • Mei-Mei Cui
    • Zheng-Hao Qiu
    • Mao-Tao He
    • Bao-Gang Zhang
  • View Affiliations

  • Published online on: May 22, 2024     https://doi.org/10.3892/mmr.2024.13252
  • Article Number: 128
  • Copyright: © Dong et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Stroke is a severe neurological disease that is associated with high rates of morbidity and mortality, and the underlying pathological processes are complex. Ferroptosis fulfills a significant role in the progression and treatment of stroke. It is well established that ferroptosis is a type of programmed cell death that is distinct from other forms or types of cell death. The process of ferroptosis involves multiple signaling pathways and regulatory mechanisms that interact with mechanisms inherent to stroke development. Inducers and inhibitors of ferroptosis have been shown to exert a role in the onset of this cell death process. Furthermore, it has been shown that interfering with ferroptosis affects the occurrence of stroke, indicating that targeting ferroptosis may offer a promising therapeutic approach for treating patients of stroke. Hence, the present review aimed to summarize the latest progress that has been made in terms of using therapeutic interventions for ferroptosis as treatment targets in cases of stroke. It provides an overview of the relevant pathways and molecular mechanisms that have been investigated in recent years, highlighting the roles of inducers and inhibitors of ferroptosis in stroke. Additionally, the intervention potential of various types of Traditional Chinese Medicine is also summarized. In conclusion, the present review provides a comprehensive overview of the potential therapeutic targets afforded by ferroptosis‑associated pathways in stroke, offering new insights into how ferroptosis may be exploited in the treatment of stroke.

Introduction

Globally, stroke ranks as the second leading cause of death, and the primary cause of disability (1). It imposes a significant burden on patients, their families and society as a whole, with ~3 million new cases occurring each year (2,3). Stroke is usually referred to as an acute cerebrovascular disease, the main mechanism of which is the sudden rupture or blockage of blood vessels in the brain, resulting in an inability of blood to enter the brain, leading to a lack of blood sugar (4) and oxygen (5), resulting in metabolic changes (6), cell death (7) and brain damage. Stroke is usually divided into two major categories; namely, ischemic stroke (including cerebral infarction) and hemorrhagic stroke (including cerebral hemorrhage and subarachnoid hemorrhage) (8). Concurrently, ischemia-reperfusion injury often occurs during the treatment of stroke, especially when reperfusion therapies, such as vascular recanalization procedures, are employed (9). A close association exists among stroke, ischemia-reperfusion injury and hemorrhagic stroke, as abnormalities in cerebral blood supply are a common feature for all of these conditions, which may lead to either damage or death of neural cells (9). When treating stroke, it is essential to consider these distinct types of injury mechanisms and their corresponding therapeutic approaches. The incidence of stroke is increasing, due to the increasing population and aging (8). Long-term disability and cognitive impairment are considered to be major causes of stroke, which is characterized by high morbidity and disability rates; thereby, stroke generally requires the support of the healthcare system (10). Therefore, exploring different means of intervention therapy for the purposes of the treatment of stroke remains a current international concern.

Ferroptosis is rapidly becoming understood to be one of the key cell death mechanisms associated with stroke (11). As is well known, ferroptosis is a type of programmed cell death that is distinct from other forms or types of cell death; it is characterized by an increase in the level of lipid peroxides, which are lethal substances that ultimately lead to oxidative stress and cell death (12). Ferroptosis differs from other forms of cell death in that morphologically, it typically manifests as increased cell volume, mitochondrial swelling and endoplasmic reticulum expansion (13); physiologically, ferroptosis involves processes such as excessive accumulation of iron ions and oxidative stress (14); and genetically, mutations or genetic variations in genes related to iron metabolism (15) or oxidative stress (16) may affect the occurrence and progression of ferroptosis (17). In addition, another characteristic of ferroptosis is that it is induced by abnormal oxidation in the intracellular microenvironment, primarily under the influence of glutathione peroxidase 4 (GPX4) activity (18). Decreased activity of GPX4 prevents the metabolism of lipid peroxides via GPX4-catalyzed glutathione (GSH) reduction reactions, resulting in the oxidation of divalent iron ions and the generation of reactive oxygen species (ROS) in lipids (19). As the cellular antioxidant capacity weakens and lipid ROS accumulate, the redox balance within cells is thereby disrupted, which induces cell death (20). Moreover, this process has also been shown to affect both upstream- and downstream-related proteins (or genes), thereby exerting different effects on the intracellular microenvironment, which ultimately influences the outcome of ferroptosis (21). The accumulation of iron ions is also one of the hallmark features of ferroptosis, accompanied by an accumulation of lethal levels of lipid peroxidation, which occurs in response to the Fenton reaction (22). The untimely increase or decrease in the intraorganismal level of iron, as the expression of iron in ferroptosis is a crucial process, will have a marked impact on the organism in question, eventually leading to the development of various diseases such as hemochromatosis or Parkinson's disease (15). In other words, the majority of the changes that occur with respect to the level of iron within organisms are primarily associated with iron metabolism (23). The interconversion between Fe3+ and Fe2+ generates toxic ROS that are often detrimental to cells, and hence iron metabolism is strictly regulated within the body (24). When the expression of proteins associated with iron metabolism is affected, this can influence either the intake or loss of iron, thereby impacting ferroptosis (25). Although the presence of ferroptosis was first demonstrated in cancer, given the increasing number of associated studies, ferroptosis has been shown to fulfill an important role within the nervous system (11,2630), and common neurological disorders, including ischemic stroke (31), Alzheimer's disease (32) and Parkinson's disease (33), have been found to be closely associated with ferroptosis, the latter two being common neurodegenerative disorders wherein the molecular mechanism is mainly concerned with an aggregation of iron in the hippocampal region of the brain or in dense areas of the substantia nigra, and these responses have been shown to be inhibited by the ferroptosis inhibitor, ferrostatin-1 (Fer-1) (3436).

In conclusion, a growing number of studies confirm the link between ferroptosis and stroke. The inherent pathological changes of stroke have been shown to be closely related to the characteristics of ferroptosis, including iron metabolism disorders, lipid peroxidation, and elevated ROS levels. Ferroptosis may provide a promising therapeutic approach for treating stroke patients. Therefore, the present review provides a comprehensive overview of potential therapeutic targets provided by ferroptosis-related pathways in stroke, providing new insights into how ferroptosis can be exploited to treat stroke.

Ferroptosis-associated pathways in stroke

Over previous years, ferroptosis has become an area of interest for researchers (12). Ferroptosis is a relatively recently discovered form of regulated cell death that is characterized by the accumulation of iron-dependent lipid peroxides (37). It has been implicated in various pathological conditions, including stroke (30). When the concept of ferroptosis was initially proposed, researchers identified key features of ferroptosis in HT-1080 cells using H2DCFDA and C11-BODIPY fluorescent probes (38). These features included abnormal accumulation of lipid peroxides and ROS (38). Subsequently, molecular experimental techniques such as western blotting and polymerase chain reaction were used to investigate the changes in ferroptosis-related proteins and genes in stroke. The results revealed the involvement of various pathways, including iron metabolism, lipid peroxidation and oxidative stress in ferroptosis (39). Understanding the ferroptosis-associated pathways in stroke can guide the development of novel therapeutic strategies for stroke treatment (40). Targeting these pathways may help to mitigate oxidative stress (41), lipid peroxidation (30) and subsequent neuronal damage (42), ultimately improving stroke outcomes. However, further research is needed to fully elucidate the intricate molecular mechanisms involved in ferroptosis and their potential as therapeutic targets in stroke.

Iron metabolism

Iron metabolism is a crucial system in organisms, and it has been shown to be closely associated with ferroptosis (15). Ferroptosis not only influences the organism, but also has an impact on various other physiological processes. The overload of iron ions exacerbates the occurrence of cerebral hemorrhage, inducing the onset of ferroptosis (11). In the event of cerebral hemorrhage, blood vessels rupture, leading to the release of a substantial amount of blood and hemoglobin (43). Subsequently, microglia rapidly engulf the released hemoglobin and metabolize Fe2+/Fe3+ (30). The accumulation of Fe2+ and Fe3+ signifies an iron overload due to excessive iron build-up, and this is a key factor in ferroptosis (15). Released Fe3+ ions enter cells by binding with transferrin receptors on the cell membrane (44). Once inside the cell, Fe3+ can be reduced to Fe2+ by ferric reductase, a process that is facilitated by hydroxyl radicals. Accumulated ROS resulting from this process lead to the peroxidization of membrane lipids, subsequently leading to a loss of cellular function and cell death. This phenomenon represents one of the characteristic features of ferroptosis (44). Alternatively, excess Fe3+ can enter the unstable iron pool through solute carrier family 39 member 14 (45), further promoting ferroptosis. Excess Fe2+ can be re-oxidized to Fe3+, which is subsequently moved to the extracellular compartment, contributing to a series of iron metabolism processes associated with ferroptosis. Moreover, the presence of this free iron stimulates the production of lipid ROS via participating in the inflammatory response (46), inducing oxidative stress (47), and the Fenton reaction (22). Consequently, there is an accumulation of lipid ROS in vivo, which leads to DNA (48), protein (49) and lipid damage (50), ultimately resulting in cell death. Studies have also demonstrated that neurons require both the ferroptosis inhibitory factor, GPX4, and genes involved in the GPX4-synthesis pathway to survive under conditions of oxidative stress (21,51,52). GPX4 utilizes GSH to reduce peroxidized lipids, thereby preventing lipoatrophy (53). This suggests that neurons are prone to undergoing ferroptosis when exposed to oxidative stress (54). Additionally, the excess iron ions metabolized by microglia are expelled through the transferrin receptor system, leading to a significant accumulation of iron in neurons (55). Subsequently, the neurons undergo the classical reaction of ferroptosis (the Fenton reaction) (56). This reaction catalyzes the generation of ROS, further promoting lipid peroxidation and leading to lipid peroxide accumulation, ultimately inducing ferroptosis (57) (Fig. 1).

Lipid peroxidation

Lipid peroxidation is a process in which lipids lose hydrogen atoms due to the activity of free radicals or lipid peroxidases (58). This leads to oxidation, fragmentation and the shortening of lipid carbon chains, resulting in the generation of lipid free radicals, lipid hydroperoxides and reactive aldehydes (such as malondialdehyde and 4-hydroxynonenal) (59). Ultimately, this process causes the oxidative degradation of lipids, thereby damaging the cells. The end product of the Fenton reaction, -OH, fulfills a crucial role in ferroptosis (60). The increase in -OH radicals induces oxidative damage, leading to ferroptosis, and an exacerbation of the edema response at the site of cerebral hemorrhage (61,62). Building upon this, in the event of a cerebral hemorrhage, ferroptosis occurring in the affected region triggers the release of iron ions from blood cells (63). These iron ions instigate oxidative stress reactions, leading to the production of -OH radicals that subsequently target DNA, proteins, and lipid membranes. The resulting damage to these components often aligns with the manifestation of ferroptosis in the brain. Furthermore, the regions affected by ferroptosis are subjected to significant iron accumulation. This accumulation of iron has two subsequent effects. First, it stimulates microglia to continue engulfing the hemoglobin that is released from blood cells, leading to the secretion of yet more iron ions, creating a positive feedback loop (64). Secondly, the accumulating iron participates in various redox reactions in the brain, resulting in an increase in the production of -OH radicals (65) (Fig. 2). In other words, when cerebral hemorrhage occurs, there can be a positive feedback system loop promoting ferroptosis through Fe/-OH/DNA damage/Fe and Fe/microglia/Fe interactions. Consequently, the brain is subjected to further oxidative damage. This oxidative damage, in the presence of interleukin and nitric oxide, further compromises the integrity of the blood-brain barrier, ultimately leading to the development of cerebral edema at the site of hemorrhage (66).

Lipoxygenases (LOXs), a key component in the process of ferroptosis, have gained significant attention in recent years (67). One study demonstrated that conducting diphenyl-1-pyrenylphosphine experiments using HEK-293 cells revealed the formation of lipid hydroperoxides catalyzed by LOX (68). Based on the findings of this study, it is hypothesized that LOX activity may enhance the accumulation of lipid hydroperoxides within cells, thereby promoting ferroptosis. This may be associated with a specific ferroptotic pathway, such as the hypoxia-inducible factor-prolyl hydroxylase domain pathway (69). Therefore, measuring LOX activity and the levels of lipid hydroperoxides may serve as valuable assays for assessing the occurrence of ferroptosis (70), given that LOX activation may drive this process (71).

System Xc−

It is widely recognized that elevated levels of glutamate can exert neurotoxic effects on the brain during disease conditions (72). In response to this neurotoxic effect, system Xc fulfills a crucial function. System Xc is a heterodimeric cystine/glutamate antiporter protein that consists of two core components: Solute carrier family 7 member 11 (SLC7A11), serving as the catalytic subunit, and solute carrier family 3 member 2, which acts as an anchoring protein (73). In the context of stroke and its relevance to ferroptosis, particular attention has been focused on the SLC7A11 gene, which encodes a sodium-independent member of the anionic amino acid transport system (51). Its highly specific role in transporting cysteine (74) and glutamate (75) has been shown to be of great importance. In general, system Xc facilitates the extracellular transport of high intracellular concentrations of glutamate, whereas the anionic form of cysteine is transported in exchange for glutamate (76). The intracellularly transported cysteine is subsequently utilized for the synthesis of cysteine and GSH (77). GSH, a tripeptide composed of glutamate, cysteine and glycine, fulfills critical roles in various physiological functions, including the scavenging of free radicals, acting as an antioxidant (78) and maintaining cellular redox balance (79). GSH also activates various enzymes that influence cellular metabolic processes, and serves as an essential intracellular antioxidant in brain diseases (such as Parkinson's disease), contributing to the scavenging of free radicals and preserving redox balance both inside and outside of cells (80).

System Xc functions within the central nervous system, where microglia, the representative immune cells, are activated from a resting state and migrate to the injured brain (81). Activated microglia can exert anti-neuroinflammatory effects (82). However, it will inhibit the release of glutamate (83), which has the consequence of inhibiting the protective effects mediated by GSH in stroke. Following a stroke, two main areas are primarily affected: i) The infarct zone (where cell death occurs due to ischemia) (84); and ii) the surrounding ischemic penumbra (85). Alterations in glutamate are known to induce the occurrence of ferroptosis during a stroke, with a significant increase in extracellular glutamate levels released by neurons in these areas (86). Consequently, the balance between system Xc and glutamate transport is disrupted, leading to both a reduced intracellular uptake of glutamate and an inhibition of GSH synthesis due to the lack of necessary raw materials (87). Ultimately, this impairment in the cellular antioxidant defense results in an accumulation of lipid ROS, with the subsequent induction of ferroptosis.

GPX4, a downstream target of GSH action, functions as a unique intracellular antioxidant enzyme acting on membrane lipid repair, which is able to directly reduce peroxidized phospholipids produced in cell membranes (88,89), and it converts toxic lipid ROS into non-toxic lipid alcohols with the aid of GSH, which thereby reduces the level of oxidative damage to cells (90). However, the generation of large amounts of lipid ROS during stroke disrupts this oxidative balance, resulting in both a large accumulation of lipid ROS and the development of peroxidation, which is the main feature of ferroptosis (42). In addition, lipid ROS have also been shown to directly react with polyunsaturated fatty acids (PUFAs) on lipid membranes via oxidation, which directly leads to the occurrence of ferroptosis in cells (91).

AMP-activated protein kinase (AMPK) signaling pathway

AMPK has a critical role in ferroptosis following cerebral hemorrhage. AMPK is an energy sensor widely present in various tissues and cells. For instance, when it is present in brain tissue and microglial cells, it can alleviate secondary damage from cerebral haemorrhage (92). It regulates cellular energy metabolism balance (93), and participates in the regulation of multiple biological processes, including glycogen synthesis (94), fatty acid synthesis (95) and mitochondrial biogenesis (96). Studies have found that Spatholobi Caulis (SC) has the ability to activate AMPK (97). Subsequent 2,7-dichlorodihydrofluorescein diacetate experiments using HepG2 cells revealed that the combined effect of arachidonic acid (AA) and iron led to increased intracellular ROS production (97). However, pretreatment with SC suppressed the production of ROS when combined with AA and iron. This suggests that AMPK may possess antioxidant capacity (97). In in vivo experiments, using a mouse liver injury model, it was shown that oral administration of SC (which activates AMPK) could alleviate liver damage mediated by acute acetaminophen (by enhancing oxidative stress and increasing cell injury), exerting antioxidant effects (97). Therefore, both in vivo and in vitro experiments indicate that AMPK can play a role in protecting against oxidative damage, possibly by inhibiting ferroptosis to provide neuroprotection (97). Furthermore, in vitro and in vivo models of intracerebral hemorrhage were created by treating BV2 cells with hemoglobin and intraventricular injection of type IV collagenase into Sprague-Dawley rats, respectively. In vitro experiments showed a phosphorylation reaction of AMPK after initial intervention with AMPK inhibitors in BV2 cells. Subsequently, pharmacological intervention led to upregulation of ROS and lipid peroxidation levels (positive regulators of ferroptosis) in BV2 cells, and silenced GPX4 (a key negative regulator of ferroptosis) levels through the AMPK signaling pathway (92). In in vivo experiments, iron deposition at the site of brain injury was observed through Perl's staining (92). So, both in vitro and in vivo experiments have demonstrated the occurrence of ferroptosis during cerebral hemorrhage. Additionally, the AMPK signaling pathway, targeting GPX4 (a negative regulator of ferroptosis), was found to be involved in neuroprotection (92). Moreover, in the field of tumor research, it has been shown that AMPK can reduce the occurrence of ferroptosis by regulating intracellular lipid synthesis metabolism (98). When ferroptosis occurs, the AMPK-mediated phosphorylation of acetyl-coenzyme A carboxylase is considered to inhibit ferroptosis by limiting the production of PUFAs (99). However, the specific role and mechanism of AMPK in the field of cerebral hemorrhage and ferroptosis have yet to be fully elucidated. When AMPK activation protects neurons from damage caused by cerebral hemorrhage through a series of antioxidant, anti-inflammatory and anti-apoptotic pathways, this may result in a reduction in the occurrence of ferroptosis (98). However, following cerebral hemorrhage, AMPK activation may increase the levels of intracellular free iron ions, which, in turn, may exacerbate oxidative stress and cell damage, potentially contributing to ferroptosis (92). Therefore, the mechanism of action of AMPK in cerebral hemorrhage may potentially exert an impact on ferroptosis. However, the current research findings are both inconsistent and limited. Further research is needed to address this issue.

Sirtuin 2 (SIRT2)-P53 signaling pathway

The SIRT2-p53 pathway exerts neuroprotective effects by modulating GPX4 and SCL7A11, inhibiting the occurrence of ferroptosis (100). SIRT2 is a NAD+-dependent deacetylase predominantly localized in the cytoplasm and has been demonstrated to be involved in the mechanisms of neuroinflammation- and neuroimmunology-related diseases (101). Additionally, P53, a tumor suppressor protein, has also been implicated in ferroptosis. In the context of ferroptosis, P53 has been shown to regulate the expression of key genes involved in iron metabolism, lipid peroxidation and antioxidant defense. P53 inhibits cystine uptake and sensitizes cells to ferroptosis by inhibiting the expression of SLC7A11, a key component of cystine/glutamate retrotransporters. Previously, the traumatic brain injury model using the controlled cortical impact (CCI) injury method found that knockdown of P53 could significantly block ferroptosis after CCI. In addition, inhibition of SIRT2 led to upregulation of acetylation and expression of P53, exacerbating ferroptosis after CCI. In other words, P53-mediated ferroptosis is involved in the pathogenesis of TBI, and SIRT2 exerts a neuroprotective effect on TBI by inhibiting P53-mediated ferroptosis (100).

Nuclear receptor coactivator 4 (NCOA4) signaling pathway

The overall role of NCOA4 in cerebral stroke has been controversial, and its specific effects on ferroptosis, contributing to this process, have yet to be fully elucidated. Currently, studies on NCOA4 have revealed its potential dual role in either promoting (102) or inhibiting ferroptosis (103). Moreover, investigations on ovarian cancer cell models with manipulated NCOA4 expression have implicated NCOA4 in the processes of both formation and degradation of intracellular iron storage autophagosomes (104). Furthermore, in ovarian cancer cells, it has been observed that up-regulation of C-MYC (a gene regulating tumor proliferation) leads to a significant reduction in ROS content (104). On the other hand, overexpression of NCOA4 reverses these changes. These findings suggest that C-MYC may exert an inhibitory effect on ferroptosis in ovarian cancer cells through NCOA4-mediated ferritin autophagy (104). The process of NCOA4-mediated ferritin autophagy appears to play a role in suppressing ferroptosis in ovarian cancer cells (104). Autophagy-associated pathways induce an excessive degradation of ferritin, leading to an increase in the amount of free iron in neurons. Therefore, when cerebral ischemia occurs, NCOA4 may promote the release of iron ions and trigger ferroptosis in cells (105). Due to the large impact of NCOA4 in cerebral ischemia-reperfusion injury, it has been shown that the autophagy-related 5 (ATG5)-ATG7-NCOA4 pathway also has an important role in ferroptosis (106). Notably, NCOA4 is a selective autophagy receptor that is essential for mediating ferritin phagocytosis in certain tissues (for example, brain tissue) and cells (for example, red blood cells) (107,108). However, other studies have found that, under conditions such as hypoxic-ischemic brain injury, NCOA4 may protect cells from excessive damage caused by free iron ions via regulating intracellular iron metabolism. In this case, NCOA4 is considered as a factor that inhibits ferroptosis (109,110).

Ferroptosis inducers and stroke

In animal models of ischemic stroke and associated clinical specimens, it was found that, when cerebral ischemia-reperfusion occurs, an increase in the level of iron ions exacerbates neuronal damage (111). However, subsequent research has indicated that there may be a close link between the increase in the level of iron ions and ferroptosis. The increase in iron undoubtedly exacerbates the occurrence of ferroptosis, and so, iron may act as a catalyst for the occurrence of ferroptosis, or serve a role as an inducer (112). Therefore, when a stroke occurs, is it possible that some of the inducers associated with ferroptosis may accelerate progression of the stroke, thereby leading to more serious consequences (Table I) (113120).

Table I.

Common ferroptosis inducers and their potential targets and mechanisms of action.

Table I.

Common ferroptosis inducers and their potential targets and mechanisms of action.

Ferroptosis inducersPotential targetsMechanisms of action(Refs.)
ErastinInhibit system XcInhibit the uptake of system Xc cystine(113)
Sulfasalazine Inhibit system Xc(114)
Glutamate Reduce the activity of system Xc(115)
RSL3Inhibit GPX4 pathwayCombined with GPX4 to reduce GPX4(116)
ML162 Combined with GPX4 to reduce GPX4(117)
iFSP1OthersaInhibit FSP1 activity and inactivate GPX4(118)
Hemin Increase unstable iron in cells(119)
Lapatinib Increase the expression of transferrin(120)

a Used to display categories other than those already listed. RSL3, RAS-selective lethal 3; ML162, 2-chloro-N-(3-chloro-4-methoxyphenyl)-N-(2-oxo-2-(phenethylamino)-1-(thiophen-2-yl)ethyl)acetamide; iFSP1, FSP1 inhibitor; Xc, cystine/glutamate antiporter; GPX4, glutathione peroxidase 4.

Inducers of ferroptosis may generally be grouped into categories, based on the effects of targeted interventions at different sites. In general, class I ferroptosis inducers that are typically used are erastin (113) (which inhibits Xc cystine uptake), glutamate (121) (which inhibits glutamate transfer to reduce Xc activity) and sulfasalazine (122) (which inhibits Xc in the cell membrane). However, the most common class I ferroptosis inducer is erastin and was first discovered before the concept of ferroptosis came into existence (38). It was then shown that the newly discovered compound erastin had no effect on either the apoptosis or necrosis of cells, but it was found that lipophilic ROS were involved in the process of ferroptosis of cells (113). After the concept of ferroptosis had been confirmed, further studies identified that erastin inhibited Xc cystine uptake, and therefore it was categorized as a class I inducer of ferroptosis for medical research (123,124). Previous studies have revealed that erastin-induced ferroptosis is a significant feature of intracerebral hemorrhage. In a mouse model of intracerebral hemorrhage (collagenase model), it was observed that the mRNA expression of GPX4, a crucial regulator of ferroptosis, was increased. This finding suggests that ferroptosis is regulated in the context of intracerebral hemorrhage (125). Moreover, cell experiments were conducted using rat PC12 cells, and the results indicated a distinct decrease in the survival rate of these cells when treated with erastin. This decrease in cell survival strongly suggests the occurrence of ferroptosis in response to erastin treatment (125). In various investigations examining the link between stroke and ferroptosis, erastin has been utilized to trigger ferroptosis, revealing its potential neuroprotective effect in alleviating stroke symptoms (126). However, further experimental investigations are needed to fully understand the precise mechanism by which erastin induces ferroptosis during stroke. It is hypothesized that erastin may play a valuable role in providing neuroprotection during strokes in the future.

The commonly used class II ferroptosis inducers are RAS-selective lethal 3 (RSL3) (127), 2-chloro-N-(3-chloro-4-methoxyphenyl)-N-(2-oxo-2-(phenethylamino)-1-(thiophen-2-yl)ethyl)acetamide (128) and cisplatin (which binds to GSH and inactivates GPX4) (129). However, the most commonly used class II ferroptosis inducer is RSL3, predominantly since RSL3 is an inducer that can either indirectly or directly induce the occurrence of ferroptosis (130). The main effect of RSL3 is that it can directly bind to GPX4 protein, thereby inactivating GPX4, at which point the production of lipid ROS is increased, leading to the occurrence of ferroptosis (131). Furthermore, it has been shown in other studies that the protective effects against cerebral ischemia-reperfusion injury, achieved by inhibiting ferroptosis, are partially diminished upon induction by RSL3 (125,132). In the context of cerebral hemorrhage ischemia-reperfusion injury, the action of RSL3 has been found to worsen the incidence of ferroptosis (133). However, the precise underlying mechanisms of this action have not yet been fully elucidated, necessitating further experimental and theoretical investigations.

There are also class III and class IV ferroptosis inducers, which similarly intervene in the process of ferroptosis through a number of different pathways. The more commonly used class III ferroptosis inducers are FSP1 inhibitor (which inhibits FSP1 activity and reduces coenzyme Q10 production) (134), statins (which inhibit the mevalonate pathway) (135,136), and class IV ferroptosis inducers, such as heme (which increases the amount of intracellular iron in the unstable state) (137) and artemisinin (which induces ferritin autophagy, causing the release of iron in the unstable state) (138). These ferroptosis inducers are representative in research related to cerebral stroke and can mitigate the occurrence of cerebral stroke by inducing ferroptosis (125,126).

Ferroptosis inhibitors and stroke

Following a stroke, various forms of cell death occur in the body, the main ones of which are apoptosis (139), necrosis (140) and autophagy (141). It has been shown that the use of different inhibitors to inhibit apoptosis, necrosis and autophagy is more effective than the use of any of these inhibitors in isolation, as in the case of cerebral hemorrhage, where caspase inhibitors have been shown to be unsuccessful in terms of inhibiting hemoglobin-induced neuronal death (142). Therefore, when ferroptosis was initially discovered, its inhibitors were also investigated within the field of stroke research (143). Currently in the medical field, a number of ferroptosis inhibitors have been identified, for which specific information for commonly used inhibitors such as Fer-1 is shown in Table II (144152). In addition, with respect to the ferroptosis inhibitors summarized in the present review and those that are similar to them in terms of their function, their specific target sites have been identified, as shown in Table III (146,147,153155). Both in vitro and in vivo experiments have shown that the levels of molecular markers of ferroptosis are increased when cerebral hemorrhage occurs. Experiments performed in vivo have shown that the mortality rate may be reduced by ~80% with the use of ferroptosis inhibitors (156,157). In vitro experiments performed in a previously published study (140) have confirmed the occurrence of ferroptosis without autophagy or apoptosis. Therefore, as a novel form of cell death that is distinct from apoptosis, necrosis, autophagy and other types of cell death, the use of certain inhibitors targeting ferroptosis may have more pronounced therapeutic effects with regard to the treatment of stroke (40). The present review provided detailed information on several different inhibitors of ferroptosis. Deferoxamine (DFO) is an iron chelator that can act on iron ions to inhibit the occurrence of ferroptosis (158). GPX4, as an important negative regulator of ferroptosis (159), may also effectively suppress the occurrence of ferroptosis (150). Fer-1, a commonly used inhibitor of ferroptosis, has been shown to have a significant role in terms of inhibiting ferroptosis (146). Additionally, when combined with inhibitors of other types of programmed cell death, Fer-1 has also been shown to exert inhibitory effects on ferroptosis (160). These effects can alleviate adverse reactions caused by stroke, thereby demonstrating that is has some therapeutic potential in terms of the treatment of stroke (161).

Table II.

Common ferroptosis inhibitors including their potential targets and mechanisms of action.

Table II.

Common ferroptosis inhibitors including their potential targets and mechanisms of action.

Ferroptosis inhibitorsPotential targetsMechanisms of action(Refs.)
Deferoxamine mesylateInhibition of Fenton reactionIron chelating agents(144)
Deferiprone Reduce unstable iron in cells(145)
Ferrostain-1AntioxidantInhibition of lipid peroxidation(146)
Liproxstatin-1 Impact Nrf2/GPX4 pathway(147)
VKH2 Inhibition of lipid oxidation(148)
XJB-5-131 Clear ROS(149)
Inhibition GPX4Inhibit classical ferroptosis pathway proteinKey proteins of the ferroptosis pathway(150)
Troglitazone Inhibiting ACSL4 function and reducing the production of lipid peroxidation raw materials(151)
Zileuton Inhibiting 5-LOX(152)

[i] VKH2, vitamin K hydroquinone; ACSL4, acyl-CoA synthetase long-chain family member 4; 5-LOX, 5-lipoxygenase; Nrf2, nuclear factor erythroid 2-related factor 2; ROS, reactive oxygen species; GPX4, glutathione peroxidase 4.

Table III.

Summary of the target sites of some ferroptosis inhibitors, including their specific target sites and specific targeting properties.

Table III.

Summary of the target sites of some ferroptosis inhibitors, including their specific target sites and specific targeting properties.

Ferroptosis inhibitorsTargets(Refs.)
DFO Fe2+(153)
CPX (154)
Ferrostain-1Iron and trace lipid(146)
Liproxstain-1hydroperoxides in liposomes(147)
GPX4GSH to GSSG(155)
L-OOH to L-OH

[i] DFO, deferoxamine; GPX4, glutathione peroxidase 4; GSH, glutathione; GSSG, oxidized glutathione; L-OOH, lipid hydroperoxide; L-OH, lipid hydroxide; CPX, ciclopirox.

DFO can relieve stroke

DFO induces ferroptosis during a stroke; DFO is an iron chelator that is able to reduce the accumulation and precipitation of iron in cells or tissues by binding to ferric (Fe3+) ions to form a stable complex, thereby allowing the removal of excess iron from cells (158). In the general field of cancer research, DFO has been shown to have good antioxidant activity (162); it acts as an anti-proliferative agent, and can induce apoptosis in cancer cells (163). At present, extensive research is being performed on the use of DFO in various iron overload-associated brain diseases (representing a class of neurological disorders caused by excessive accumulation of iron ions in the body, such as thalassemia). DFO has been shown to help regulate iron balance in the body, and to reduce neurological damage and functional impairments associated with iron overload (164). Similarly, significant research has also been performed on the role of DFO in neurodegenerative diseases (such as diseases that are characterized by neuronal cell death and functional impairments, including Alzheimer's disease, Parkinson's disease and Huntington's disease, which are associated with oxidative stress, abnormal neurodevelopment and protein aggregation) (153). Upon reviewing the literature, DFO has been shown to reduce the level of cell death through reducing free radicals (165), and to promote wound healing and healing in diabetic patients (166), although due to the potential toxicity of DFO itself (167), the research remains only at an early stage (160). In addition, animal experiments were used to demonstrate that the treatment of mice with DFO, wherein the aging process was simulated, led to a marked alleviation of the occurrence of ferroptosis, with the consequent inhibition of the increase of age spots in mice due to iron overload, thereby achieving the desired protective effect of delaying aging (168). In investigating the neurological and cognitive functions of aged mice in a study focused on the auditory cortex, the effect of alleviating ferroptosis in the brain was achieved by treatment with DFO, suggesting that DFO may be used to treat auditory and cognitive impairment resulting from age-associated problems (169). It is noteworthy that, although DFO primarily functions as an iron chelator in ferroptosis (158), investigations utilizing rodent models propose that DFO could impact stroke through gene-mediated mechanisms (69,170). Therefore, the role of DFO in ferroptosis warrants further investigation.

Fer-1 alleviates stroke

Fer-1 has been shown to inhibit ferroptosis during stroke (161). Traditionally, the most direct way to inhibit ferroptosis has been to utilize appropriate ferroptosis inhibitors. In the field of stroke-related research, the most commonly used ferroptosis inhibitor is Fer-1, whose main effects are to scavenge ROS and to inhibit erastin, the aforementioned class I inducer of ferroptosis (113,171). The effect of Fer-1 was verified with experimentally administered ventricular Fer-1 in experiments with rats, where it was found to inhibit erastin-induced accumulation of ROS in the cytoplasm and lipids, resulting in a decrease in ROS and lower levels of ferroptosis, demonstrating that Fer-1 could appreciably slow down the onset of ferroptosis in the brain (171). Notably, when Fer-1 intervention was present in the ventricles, the levels of iron deposition and neuronal degeneration were both significantly reduced, reducing the level of cellular damage, while also improving long-term motor and cognitive function (172). After a stroke, iron tends to accumulate in the damaged area, where it participates in oxidative stress reactions, leading to more cell damage and inflammatory responses (173). In addition, free iron is able to promote abnormal protein aggregation and neurodevelopmental abnormalities (174). The inhibitor Fer-1 may reduce oxidative stress reactions by reducing levels of free iron ions, thereby reducing cell damage and inflammatory responses following a stroke (166). Furthermore, inhibiting ferroptosis may also lead to the aggregation of abnormal proteins and abnormalities in neurodevelopment (175). Fer-1 exerts protective effects on cerebral ischemia-reperfusion injury by activating the Akt/GSK-3β pathway, indicating that ferroptosis may become a novel target in the treatment of ischemic stroke in the future (176). However, at present, few studies have reported on the specific effects and associated pathways of Fer-1 following a stroke, and therefore our knowledge is currently relatively limited; further experimental and clinical studies are required to explore its underlying mechanisms of action, and the potential therapeutic effects.

Effects of Traditional Chinese Medicine (TCM) on ferroptosis

Through clinical trial studies, TCM combined with the influencing factors associated with ferroptosis has been shown to be more effective than single-drug treatment for stroke (177,178). It is well established that TCM herbs have antioxidant, anti-inflammatory and blood-brain barrier-protective effects, and that they can prevent stroke in advance by various means (179181). For example, Danhong injection, a standardized injection comprising danshen (Salvia miltiorrhiza) and saffron, has been shown in studies to improve ferroptosis in ischemic stroke (182,183). In addition, moxibustion (a form of therapy that entails the burning of mugwort leaves), as one of the more frequently used treatments, has an important role in the treatment of cerebral infarction. It has been found that moxibustion can reduce neurological damage and neuronal death, reduce the accumulation of ROS and inhibit ferroptosis (184). In conclusion, the effects of certain Chinese medicines and their active ingredients on stroke both involve multiple pathways and are multi-targeted. In addition, the intervention of Chinese medicines on ferroptosis has been shown to be more stable and safer to use compared with small-molecule inducers or inhibitors of ferroptosis (185). For example, astragaloside IV can alleviate brain injury by inhibiting the ferroptosis-associated sequestosome-1/kelch-like ECH associated protein 1/nuclear factor erythroid 2-related factor 2 pathway (177,186). In addition, there are studies reporting that TCM treatment can reduce the side effects of drug toxicity in patients via targeting ferroptosis, leading to significant improvements in patient safety and quality of life (187,188). The impact that specific Chinese medicines and their active constituents have on stroke involves multiple pathways and targets, and these are summarized in Table IV (125,182,189191).

Table IV.

Summary of how certain TCM utilize ferroptosis to treat stroke, including an overview of specific pathways and target points through which these Chinese medicines engage with ferroptosis to address stroke.

Table IV.

Summary of how certain TCM utilize ferroptosis to treat stroke, including an overview of specific pathways and target points through which these Chinese medicines engage with ferroptosis to address stroke.

TCMFerroptosis pathwayTherapeutic targetClinical trial stagesRegulatory bodies(Refs.)
Salvia miltiorrhizaReduced expression of Lip ROSNRF2Pharmaceutical research stageNO(189)
Angong NiuhuangActivate PPARγ/GPX4Listed for useNational Medical(190)
WanAKT/GPX4 pathway Products Administration
Scutellaria baicalensisReduce iron depositionFePreclinical biological research stageNO(125)
DanhongSATB1/SLC7A11/HO-1 axisSLC7A11Preclinical biological research stageNO(182)
Astragaloside IVACSL4-related pathwaysACSL4Pharmaceutical research stageNO(191)

[i] TCM, Traditional Chinese Medicine; ROS, reactive oxygen species; GPX4, glutathione peroxidase 4; SLC7A11, solute carrier family 7 member 11; Lip, lipid; NRF2, nuclear factor erythroid 2-related factor 2; PPARγ, peroxisome proliferator-activated receptor γ; AKT, protein kinase B; SATB1, special AT-rich sequence-binding protein 1; HO-1, heme oxygenase-1; ACSL4, acyl-CoA synthetase long-chain family member 4; NO, records have not yet been approved.

Discussion

The present review provides a comprehensive overview of the potential therapeutic targets of ferroptosis-associated pathways in stroke, providing novel insights into the application of ferroptosis in the treatment of stroke. Ferroptosis is a relatively recently discovered form of cell death that was first proposed by the laboratory of Brent R. Stockwell in 2012 (38). It is characterized by an excessive accumulation of lipid peroxides, and this accumulation is dependent on iron ions (192). Ferroptosis distinguishes itself from other forms of cell death, such as apoptosis, necrosis, autophagy (193,194) and pyroptosis, in terms of its morphological and biological features, and its underlying mechanistic regulation (44). It is also associated with inflammation and oxidative stress, along with other pathological processes (195). The characteristic morphological changes observed in ferroptosis primarily include mitochondrial atrophy, a ruptured outer membrane, reduced cristae, a compressed inner membrane and intact nuclei (196). By contrast, apoptosis and necrosis typically exhibit swollen mitochondria and fragmented nuclei (197). In recent years, the role of ferroptosis in various pathological processes has gained significant attention. Although ferroptosis was initially identified in studies that were associated with cancer, it has been demonstrated to fulfill a crucial role in the progression and toxicity of numerous neurological diseases, including stroke (198), Parkinson's disease (199) and Alzheimer's disease (32). Multiple reviews have highlighted ferroptosis as a promising target for various neurological disorders, and these reviews have also summarized the major regulators and associated studies in this field (178,200,201). The present review focused on summarizing the ferroptosis-associated pathways in stroke and discussed the potential therapeutic interventions using inhibitors and inducers of ferroptosis, with the ultimate goal of alleviating the impact of stroke.

The collection of studies published on ferroptosis in stroke to date have provided a comprehensive and informative overview of the field. By summarizing the findings from these studies, specific research goals have been identified that should guide targeted and precise investigations in the area of ferroptosis in stroke. This approach allows results to be achieved faster and more efficiently, avoiding unnecessary detours. By summarizing this article, important signaling pathways of iron death in stroke, and inhibitors and inductors of iron death can be refined. Intervening in the signaling pathways of iron death and applying inductors can slow down the occurrence of stroke. This improved understanding will enable more accurate interventions for stroke management, specifically targeting ferroptosis. In conclusion, several preclinical studies (202,203) have confirmed the protective effect of ferroptosis inhibitors in stroke, and have highlighted the potential of these inhibitors as novel therapeutic drugs for stroke treatment.

Although ferroptosis-associated pathways are potential targets for the treatment of stroke, there remain certain limitations that need to be addressed. First, although iron overload is a key factor in ferroptosis, it remains unclear whether other metal ions also serve a role in this process, or whether alternative forms of cell death involving different metal ions also have a participatory role (204). Further investigations are needed to explore these possibilities. Additionally, despite the numerous studies that have been published on stroke and ferroptosis, very few of these have translated into clinical applications. Although it has been established that neuronal cells in the brain are particularly susceptible to ferroptosis, the effects of ferroptosis on other cell types in the brain have yet to be fully elucidated (51). Further studies exploring the effects of ferroptosis on various brain cell types are necessary to guide future research directions. In summary, targeting ferroptosis-associated pathways represents a promising approach for stroke treatment. However, there is a need for further research to improve understanding of the roles and potential targets of ferroptosis-associated pathways in stroke, which will provide valuable insights for the prevention and treatment of stroke.

The present review focused on the impact of ferroptosis on stroke and studied whether intervening in the ferroptosis pathway can prevent and treat stroke. Inhibiting ferroptosis pathways has shown promise in reducing neuronal cell death, protecting brain tissue, and improving functional outcomes in stroke models. This therapeutic approach not only provides new therapeutic avenues beyond traditional approaches, but also highlights the importance of understanding the mechanisms of ferroptosis in developing more effective stroke therapies. Therefore, the exploration of ferroptosis inhibitors or inducers represents a major advance in stroke treatment and provides new strategies for the prevention and treatment of cerebral stroke.

Acknowledgements

Not applicable.

Funding

This study was supported by The National Natural Science Foundation of China (grant no. 82101410), The Medicine and Health Science and Technology Development Plan Project of Shandong (grant no. 202101040805) and Research and Innovation Plan Project of Weifang Medical University (grant no. 2021BKQ).

Availability of data and materials

Not applicable.

Authors' contributions

BGZ and MTH conceptualized the study; HD was responsible for methodology and visualization; HD, YPM, MMC and ZHQ were responsible for the writing, reviewing and editing of the manuscript; and MTH and BGZ provided supervision and corrections. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare they have no competing interests.

References

1 

Shehjar F, Maktabi B, Rahman ZA, Bahader GA, James AW, Naqvi A, Mahajan R and Shah ZA: Stroke: Molecular mechanisms and therapies: Update on recent developments. Neurochem Int. 162:1054582023. View Article : Google Scholar : PubMed/NCBI

2 

Wu S, Wu B, Liu M, Chen Z, Wang W, Anderson CS, Sandercock P, Wang Y, Huang Y, Cui L, et al: Stroke in China: Advances and challenges in epidemiology, prevention, and management. Lancet Neurol. 18:394–405. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Barthels D and Das H: Current advances in ischemic stroke research and therapies. Biochim Biophys Acta Mol Basis Dis. 1866:1652602020. View Article : Google Scholar : PubMed/NCBI

4 

Martin S: Stroke: Does intensive blood sugar control improve prognosis? Dtsch med Wochenschr. 137:26282012.(In German). PubMed/NCBI

5 

Wu X, You J, Chen X, Zhou M, Ma H, Zhang T and Huang C: An overview of hyperbaric oxygen preconditioning against ischemic stroke. Metab Brain Dis. 38:855–872. 2023. View Article : Google Scholar : PubMed/NCBI

6 

Shin TH, Lee DY, Basith S, Manavalan B, Paik MJ, Rybinnik I, Mouradian MM, Ahn JH and Lee G: Metabolome changes in cerebral ischemia. Cells. 9:16302020. View Article : Google Scholar : PubMed/NCBI

7 

Tuo QZ, Zhang ST and Lei P: Mechanisms of neuronal cell death in ischemic stroke and their therapeutic implications. Med Res Rev. 42:259–305. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Delling FN, et al: Heart disease and stroke statistics-2020 update: A report from the american heart association. Circulation. 141:e139–e596. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Przykaza Ł: Understanding the connection between common stroke comorbidities, their associated inflammation, and the course of the cerebral ischemia/reperfusion cascade. Front Immunol. 12:7825692021. View Article : Google Scholar : PubMed/NCBI

10 

Rothwell PM, Algra A and Amarenco P: Medical treatment in acute and long-term secondary prevention after transient ischaemic attack and ischaemic stroke. Lancet. 377:1681–1692. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Guo J, Tuo Q and Lei P: Iron, ferroptosis, and ischemic stroke. J Neurochem. 165:487–520. 2023. View Article : Google Scholar : PubMed/NCBI

12 

Li J, Cao F, Yin H, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11:882020. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Xu L, Liu Y, Chen X, Zhong H and Wang Y: Ferroptosis in life: To be or not to be. Biomed Pharmacother. 159:1142412023. View Article : Google Scholar : PubMed/NCBI

15 

Sun Y, Li Q, Guo H and He Q: Ferroptosis and iron metabolism after intracerebral hemorrhage. Cells. 12:902022. View Article : Google Scholar : PubMed/NCBI

16 

Chen GH, Song CC, Pantopoulos K, Wei XL, Zheng H and Luo Z: Mitochondrial oxidative stress mediated Fe-induced ferroptosis via the NRF2-ARE pathway. Free Radic Biol Med. 180:95–107. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Sun Y, Chen P, Zhai B, Zhang M, Xiang Y, Fang J, Xu S, Gao Y, Chen X, Sui X and Li G: The emerging role of ferroptosis in inflammation. Biomed Pharmacother. 127:1101082020. View Article : Google Scholar : PubMed/NCBI

18 

Liu Y, Wan Y, Jiang Y, Zhang L and Cheng W: GPX4: The hub of lipid oxidation, ferroptosis, disease and treatment. Biochim Biophys Acta Rev Cancer. 1878:1888902023. View Article : Google Scholar : PubMed/NCBI

19 

Wang X, Shen T, Lian J, Deng K, Qu C, Li E, Li G, Ren Y, Wang Z, Jiang Z, et al: Resveratrol reduces ROS-induced ferroptosis by activating SIRT3 and compensating the GSH/GPX4 pathway. Mol Med. 29:1372023. View Article : Google Scholar : PubMed/NCBI

20 

Snezhkina AV, Kudryavtseva AV, Kardymon OL, Savvateeva MV, Melnikova NV, Krasnov GS and Dmitriev AA: ROS generation and antioxidant defense systems in normal and malignant cells. Oxid Med Cell Longev. 2019:61758042019. View Article : Google Scholar : PubMed/NCBI

21 

Fu C, Wu Y, Liu S, Luo C, Lu Y, Liu M, Wang L, Zhang Y and Liu X: Rehmannioside A improves cognitive impairment and alleviates ferroptosis via activating PI3K/AKT/Nrf2 and SLC7A11/GPX4 signaling pathway after ischemia. J Ethnopharmacol. 289:1150212022. View Article : Google Scholar : PubMed/NCBI

22 

Henning Y, Blind US, Larafa S, Matschke J and Fandrey J: Hypoxia aggravates ferroptosis in RPE cells by promoting the Fenton reaction. Cell Death Dis. 13:6622022. View Article : Google Scholar : PubMed/NCBI

23 

Kosman DJ: Redox cycling in iron uptake, efflux, and trafficking. J Biol Chem. 285:26729–26735. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Lee J and Hyun DH: The interplay between intracellular iron homeostasis and neuroinflammation in neurodegenerative diseases. Antioxidants (Basel). 12:9182023. View Article : Google Scholar : PubMed/NCBI

25 

Recalcati S, Gammella E and Cairo G: Dysregulation of iron metabolism in cancer stem cells. Free Radic Biol Med. 133:216–220. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Pan F, Xu W, Ding J and Wang C: Elucidating the progress and impact of ferroptosis in hemorrhagic stroke. Front Cell Neurosci. 16:10675702023. View Article : Google Scholar : PubMed/NCBI

27 

Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q and Wang J: Ferroptosis and its role in diverse brain diseases. Mol Neurobiol. 56:4880–4893. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Li Q, Han X, Lan X, Gao Y, Wan J, Durham F, Cheng T, Yang J, Wang Z, Jiang C, et al: Inhibition of neuronal ferroptosis protects hemorrhagic brain. JCI Insight. 2:e907772017. View Article : Google Scholar : PubMed/NCBI

29 

Speer RE, Karuppagounder SS, Basso M, Sleiman SF, Kumar A, Brand D, Smirnova N, Gazaryan I, Khim SJ and Ratan RR: Hypoxia-inducible factor prolyl hydroxylases as targets for neuroprotection by ‘antioxidant’ metal chelators: From ferroptosis to stroke. Free Radic Biol Med. 62:26–36. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Xu Y, Li K, Zhao Y, Zhou L, Liu Y and Zhao J: Role of ferroptosis in stroke. Cell Mol Neurobiol. 43:205–222. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Liu J, Guo ZN, Yan XL, Huang S, Ren JX, Luo Y and Yang Y: Crosstalk between autophagy and ferroptosis and its putative role in ischemic stroke. Front Cell Neurosci. 14:5774032020. View Article : Google Scholar : PubMed/NCBI

32 

Bao WD, Pang P, Zhou XT, Hu F, Xiong W, Chen K, Wang J, Wang F, Xie D, Hu YZ, et al: Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer's disease. Cell Death Differ. 28:1548–1562. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Mahoney-Sánchez L, Bouchaoui H, Ayton S, Devos D, Duce JA and Devedjian JC: Ferroptosis and its potential role in the physiopathology of Parkinson's disease. Prog Neurobiol. 196:1018902021. View Article : Google Scholar : PubMed/NCBI

34 

Wang Y, Chen G and Shao W: Identification of ferroptosis-related genes in Alzheimer's disease based on bioinformatic analysis. Front Neurosci. 16:8237412022. View Article : Google Scholar : PubMed/NCBI

35 

Wang C, Chen S, Guo H, Jiang H, Liu H, Fu H and Wang D: Forsythoside A mitigates Alzheimer's-like pathology by inhibiting ferroptosis-mediated neuroinflammation via Nrf2/GPX4 axis activation. Int J Biol Sci. 18:2075–2090. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Jakaria M, Belaidi AA, Bush AI and Ayton S: Ferroptosis as a mechanism of neurodegeneration in Alzheimer's disease. J Neurochem. 159:804–825. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI

38 

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

39 

Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al: DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 593:586–590. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Alim I, Caulfield JT, Chen Y, Swarup V, Geschwind DH, Ivanova E, Seravalli J, Ai Y, Sansing LH, Ste Marie EJ, et al: Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell. 177:1262–1279.e25. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Ren JX, Li C, Yan XL, Qu Y, Yang Y and Guo ZN: Crosstalk between oxidative stress and ferroptosis/oxytosis in ischemic stroke: Possible targets and molecular mechanisms. Oxid Med Cell Longev. 2021:66433822021. View Article : Google Scholar : PubMed/NCBI

42 

Si W, Sun B, Luo J, Li Z, Dou Y and Wang Q: Snap25 attenuates neuronal injury via reducing ferroptosis in acute ischemic stroke. Exp Neurol. 367:1144762023. View Article : Google Scholar : PubMed/NCBI

43 

Kuriakose D and Xiao Z: Pathophysiology and treatment of stroke: Present status and future perspectives. Int J Mol Sci. 21:76092020. View Article : Google Scholar : PubMed/NCBI

44 

Liu Y, Fang Y, Zhang Z, Luo Y, Zhang A, Lenahan C and Chen S: Ferroptosis: An emerging therapeutic target in stroke. J Neurochem. 160:64–73. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Prajapati M, Conboy HL, Hojyo S, Fukada T, Budnik B and Bartnikas TB: Biliary excretion of excess iron in mice requires hepatocyte iron import by Slc39a14. J Biol Chem. 297:1008352021. View Article : Google Scholar : PubMed/NCBI

46 

Chen Y, Fang ZM, Yi X, Wei X and Jiang DS: The interaction between ferroptosis and inflammatory signaling pathways. Cell Death Dis. 14:2052023. View Article : Google Scholar : PubMed/NCBI

47 

Wang W, Jing X, Du T, Ren J, Liu X, Chen F, Shao Y, Sun S, Yang G and Cui X: Iron overload promotes intervertebral disc degeneration via inducing oxidative stress and ferroptosis in endplate chondrocytes. Free Radic Biol Med. 190:234–246. 2022. View Article : Google Scholar : PubMed/NCBI

48 

Shi F, Zhang Z, Cui H, Wang J, Wang Y, Tang Y, Yang W, Zou P, Ling X, Han F, et al: Analysis by transcriptomics and metabolomics for the proliferation inhibition and dysfunction through redox imbalance-mediated DNA damage response and ferroptosis in male reproduction of mice and TM4 Sertoli cells exposed to PM2.5. Ecotoxicol Environ Saf. 238:1135692022. View Article : Google Scholar : PubMed/NCBI

49 

Lin Q, Li S, Jin H, Cai H, Zhu X, Yang Y, Wu J, Qi C, Shao X, Li J, et al: Mitophagy alleviates cisplatin-induced renal tubular epithelial cell ferroptosis through ROS/HO-1/GPX4 axis. Int J Biol Sci. 19:1192–1210. 2023. View Article : Google Scholar : PubMed/NCBI

50 

Su LJ, Zhang JH, Gomez H, Murugan R, Hong X, Xu D, Jiang F and Peng ZY: Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid Med Cell Longev. 2019:50808432019. View Article : Google Scholar : PubMed/NCBI

51 

Yuan Y, Zhai Y, Chen J, Xu X and Wang H: Kaempferol ameliorates oxygen-glucose deprivation/reoxygenation-induced neuronal ferroptosis by activating Nrf2/SLC7A11/GPX4 axis. Biomolecules. 11:9232021. View Article : Google Scholar : PubMed/NCBI

52 

Liu H, Zhang T, Zhang WY, Huang SR, Hu Y and Sun J: Rhein attenuates cerebral ischemia-reperfusion injury via inhibition of ferroptosis through NRF2/SLC7A11/GPX4 pathway. Exp Neurol. 369:1145412023. View Article : Google Scholar : PubMed/NCBI

53 

Ursini F and Maiorino M: Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radic Biol Med. 152:175–185. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Ralhan I, Chang J, Moulton MJ, Goodman LD, Lee NYJ, Plummer G, Pasolli HA, Matthies D, Bellen HJ and Ioannou MS: Autolysosomal exocytosis of lipids protect neurons from ferroptosis. J Cell Biol. 222:e2022071302023. View Article : Google Scholar : PubMed/NCBI

55 

Mamais A, Kluss JH, Bonet-Ponce L, Landeck N, Langston RG, Smith N, Beilina A, Kaganovich A, Ghosh MC, Pellegrini L, et al: Correction: Mutations in LRRK2 linked to Parkinson disease sequester Rab8a to damaged lysosomes and regulate transferrin-mediated iron uptake in microglia. PLoS Biol. 20:e30016212022. View Article : Google Scholar : PubMed/NCBI

56 

Reyhani A, McKenzie TG, Fu Q and Qiao GG: Fenton-chemistry-mediated radical polymerization. Macromol Rapid Commun. 40:19002202019. View Article : Google Scholar : PubMed/NCBI

57 

Chen Y, Yang Z, Wang S, Ma Q, Li L, Wu X, Guo Q, Tao L and Shen X: Boosting ROS-mediated lysosomal membrane permeabilization for cancer ferroptosis therapy. Adv Healthc Mater. 12:22021502023. View Article : Google Scholar

58 

Von Krusenstiern AN, Robson RN, Qian N, Qiu B, Hu F, Reznik E, Smith N, Zandkarimi F, Estes VM, Dupont M, et al: Identification of essential sites of lipid peroxidation in ferroptosis. Nat Chem Biol. 19:719–730. 2023. View Article : Google Scholar : PubMed/NCBI

59 

Ayala A, Muñoz MF and Argüelles S: Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014:3604382014. View Article : Google Scholar : PubMed/NCBI

60 

Zhu G, Chi H, Liu M, Yin Y, Diao H, Liu Z, Guo Z, Xu W, Xu J, Cui C, et al: Multifunctional ‘ball-rod’ Janus nanoparticles boosting Fenton reaction for ferroptosis therapy of non-small cell lung cancer. J Colloid Interface Sci. 621:12–23. 2022. View Article : Google Scholar : PubMed/NCBI

61 

Kajarabille N and Latunde-Dada GO: Programmed cell-death by ferroptosis: Antioxidants as mitigators. Int J Mol Sci. 20:49682019. View Article : Google Scholar : PubMed/NCBI

62 

Li J, Jia B, Cheng Y, Song Y, Li Q and Luo C: Targeting Molecular mediators of ferroptosis and oxidative stress for neurological disorders. Oxid Med Cell Longev. 2022:39990832022.PubMed/NCBI

63 

Wan J, Ren H and Wang J: Iron toxicity, lipid peroxidation and ferroptosis after intracerebral haemorrhage. Stroke Vasc Neurol. 4:93–95. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Garton T, Keep RF, Hua Y and Xi G: CD163, a hemoglobin/haptoglobin scavenger receptor, after intracerebral hemorrhage: Functions in microglia/macrophages versus neurons. Transl Stroke Res. 8:612–616. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Hare D, Ayton S, Bush A and Lei P: A delicate balance: Iron metabolism and diseases of the brain. Front Aging Neurosci. 5:342013. View Article : Google Scholar : PubMed/NCBI

66 

Yang C, Hawkins KE, Doré S and Candelario-Jalil E: Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol. 316:C135–C153. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Bouchaoui H, Mahoney-Sanchez L, Garçon G, Berdeaux O, Alleman LY, Devos D, Duce JA and Devedjian JC: ACSL4 and the lipoxygenases 15/15B are pivotal for ferroptosis induced by iron and PUFA dyshomeostasis in dopaminergic neurons. Free Radic Biol Med. 195:145–157. 2023. View Article : Google Scholar : PubMed/NCBI

68 

Rock C and Moos PJ: Selenoprotein P protects cells from lipid hydroperoxides generated by 15-LOX-1. Prostaglandins Leukot Essent Fatty Acids. 83:203–210. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Karuppagounder SS, Alim I, Khim SJ, Bourassa MW, Sleiman SF, John R, Thinnes CC, Yeh TL, Demetriades M, Neitemeier S, et al: Therapeutic targeting of oxygen-sensing prolyl hydroxylases abrogates ATF4-dependent neuronal death and improves outcomes after brain hemorrhage in several rodent models. Sci Transl Med. 8:328ra292016. View Article : Google Scholar : PubMed/NCBI

70 

Wang H, Xu L, Tang X, Jiang Z and Feng X: Lipid peroxidation-induced ferroptosis as a therapeutic target for mitigating neuronal injury and inflammation in sepsis-associated encephalopathy: Insights into the hippocampal PEBP-1/15-LOX/GPX4 pathway. Lipids Health Dis. 23:1282024. View Article : Google Scholar : PubMed/NCBI

71 

Shah R, Shchepinov MS and Pratt DA: Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci. 4:387–396. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Martami F and Holton KF: Targeting glutamate neurotoxicity through dietary manipulation: Potential treatment for migraine. Nutrients. 15:39522023. View Article : Google Scholar : PubMed/NCBI

73 

Saini KK, Chaturvedi P, Sinha A, Singh MP, Khan MA, Verma A, Nengroo MA, Satrusal SR, Meena S, Singh A, et al: Loss of PERK function promotes ferroptosis by downregulating SLC7A11 (System Xc-) in colorectal cancer. Redox Biol. 65:1028332023. View Article : Google Scholar : PubMed/NCBI

74 

Koppula P, Zhuang L and Gan B: Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 12:599–620. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Dahlmanns M, Dahlmanns JK, Savaskan N, Steiner HH and Yakubov E: Glial glutamate transporter-mediated plasticity: System xc-/xCT/SLC7A11 and EAAT1/2 in brain diseases. Front Biosci (Landmark Ed). 28:572023. View Article : Google Scholar : PubMed/NCBI

76 

Albrecht P, Lewerenz J, Dittmer S, Noack R, Maher P and Methner A: Mechanisms of oxidative glutamate toxicity: The glutamate/cystine antiporter system xc-as a neuroprotective drug target. CNS Neurol Disord Drug Targets. 9:373–382. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Puka-Sundvall M, Eriksson P, Nilsson M, Sandberg M and Lehmann A: Neurotoxicity of cysteine: interaction with glutamate. Brain Res. 705:65–70. 1995. View Article : Google Scholar : PubMed/NCBI

78 

Yuan Y, Yucai L, Lu L, Hui L, Yong P and Haiyang Y: Acrylamide induces ferroptosis in HSC-T6 cells by causing antioxidant imbalance of the XCT-GSH-GPX4 signaling and mitochondrial dysfunction. Toxicol Lett. 368:24–32. 2022. View Article : Google Scholar : PubMed/NCBI

79 

Zhang W, Niu C, Liu Y and Chen B: Glutathione redox balance in hibernating Chinese soft-shelled turtle Pelodiscus sinensis hatchlings. Comp Biochem Physiol B Biochem Mol Biol. 207:9–14. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Iskusnykh IY, Zakharova AA and Pathak D: Glutathione in brain disorders and aging. Molecules. 27:3242022. View Article : Google Scholar : PubMed/NCBI

81 

Miladinovic T and Singh G: Spinal microglia contribute to cancer-induced pain through system xC-mediated glutamate release. Pain Rep. 4:e7382019. View Article : Google Scholar : PubMed/NCBI

82 

Shen SY, Yu R, Li W, Liang LF, Han QQ, Huang HJ, Li B, Xu SF, Wu GC, Zhang YQ and Yu J: The neuroprotective effects of GPR55 against hippocampal neuroinflammation and impaired adult neurogenesis in CSDS mice. Neurobiol Dis. 169:1057432022. View Article : Google Scholar : PubMed/NCBI

83 

Chen Z and Trapp BD: Microglia and neuroprotection. J Neurochem. 136 (Suppl 1):S10–S17. 2016. View Article : Google Scholar

84 

Frank D, Gruenbaum BF, Grinshpun J, Melamed I, Severynovska O, Kuts R, Semyonov M, Brotfain E, Zlotnik A and Boyko M: Measuring post-stroke cerebral edema, infarct zone and blood-brain barrier breakdown in a single set of rodent brain samples. J Vis Exp. 2020:e613092020.

85 

Raper DMS and Abla AA: Commentary: Encephalodu-roarteriosynangiosis averts stroke in atherosclerotic patients with border-zone infarct: Post hoc analysis from a performance criterion phase II trial. Neurosurgery. 88:E319–E320. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Fan G, Liu M, Liu J and Huang Y: The initiator of neuroexcitotoxicity and ferroptosis in ischemic stroke: Glutamate accumulation. Front Mol Neurosci. 16:11130812023. View Article : Google Scholar : PubMed/NCBI

87 

Bridges R, Lutgen V, Lobner D and Baker DA: Thinking outside the cleft to understand synaptic activity: Contribution of the cystine-glutamate antiporter (System xc-) to normal and pathological glutamatergic signaling. Pharmacol Rev. 64:780–802. 2012. View Article : Google Scholar : PubMed/NCBI

88 

Seibt TM, Proneth B and Conrad M: Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 133:144–152. 2019. View Article : Google Scholar : PubMed/NCBI

89 

Liu J, Yang G and Zhang H: Glyphosate-triggered hepatocyte ferroptosis via suppressing Nrf2/GSH/GPX4 axis exacerbates hepatotoxicity. Sci Total Environ. 862:1608392023. View Article : Google Scholar : PubMed/NCBI

90 

Jin M, Shi C, Li T, Wu Y, Hu C and Huang G: Solasonine promotes ferroptosis of hepatoma carcinoma cells via glutathione peroxidase 4-induced destruction of the glutathione redox system. Biomed Pharmacother. 129:1102822020. View Article : Google Scholar : PubMed/NCBI

91 

Delesderrier E, Monteiro JDC, Freitas S, Pinheiro IC, Batista MS and Citelli M: Can iron and polyunsaturated fatty acid supplementation induce ferroptosis? Cell Physiol Biochem. 57:24–41. 2023. View Article : Google Scholar : PubMed/NCBI

92 

Xie G, Liang Y, Gao W, Wu L, Zhang Y, Ye Z and Qin C: Artesunate alleviates intracerebral haemorrhage secondary injury by inducing ferroptosis in M1-polarized microglia and suppressing inflammation through AMPK/mTORC1/GPX4 pathway. Basic Clin Pharmacol Toxicol. 132:369–383. 2023. View Article : Google Scholar : PubMed/NCBI

93 

Steinberg GR and Hardie DG: New insights into activation and function of the AMPK. Nat Rev Mol Cell Biol. 24:255–272. 2023. View Article : Google Scholar : PubMed/NCBI

94 

Muraleedharan R and Dasgupta B: AMPK in the brain: Its roles in glucose and neural metabolism. FEBS J. 289:2247–2262. 2022. View Article : Google Scholar : PubMed/NCBI

95 

Taghiyar S, Pourrajab F and Aarabi MH: Astaxanthin improves fatty acid dysregulation in diabetes by controlling the AMPK-SIRT1 pathway. EXCLI J. 22:502–515. 2023.PubMed/NCBI

96 

Malik N, Ferreira BI, Hollstein PE, Curtis SD, Trefts E, Weiser Novak S, Yu J, Gilson R, Hellberg K, Fang L, et al: Induction of lysosomal and mitochondrial biogenesis by AMPK phosphorylation of FNIP1. Science. 380:eabj55592023. View Article : Google Scholar : PubMed/NCBI

97 

Bae SJ, Bak SB and Kim YW: Coordination of AMPK and YAP by Spatholobi caulis and procyanidin B2 provides antioxidant effects in vitro and in vivo. Int J Mol Sci. 23:137302022. View Article : Google Scholar : PubMed/NCBI

98 

Lee H, Zandkarimi F, Zhang Y, Meena JK, Kim J, Zhuang L, Tyagi S, Ma L, Westbrook TF, Steinberg GR, et al: Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol. 22:225–234. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Chen X, Kang R, Kroemer G and Tang D: Broadening horizons: The role of ferroptosis in cancer. Nat Rev Clin Oncol. 18:280–296. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Gao J, Li Y and Song R: SIRT2 inhibition exacerbates p53-mediated ferroptosis in mice following experimental traumatic brain injury. Neuroreport. 32:1001–1008. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Lu W, Ji H and Wu D: SIRT2 plays complex roles in neuroinflammation neuroimmunology-associated disorders. Front Immunol. 14:11741802023. View Article : Google Scholar : PubMed/NCBI

102 

Wu H, Liu Q, Shan X, Gao W and Chen Q: ATM orchestrates ferritinophagy and ferroptosis by phosphorylating NCOA4. Autophagy. 19:2062–2077. 2023. View Article : Google Scholar : PubMed/NCBI

103 

Mi Y, Wei C, Sun L, Liu H, Zhang J, Luo J, Yu X, He J, Ge H and Liu P: Melatonin inhibits ferroptosis and delays age-related cataract by regulating SIRT6/p-Nrf2/GPX4 and SIRT6/NCOA4/FTH1 pathways. Biomed Pharmacother. 157:1140482023. View Article : Google Scholar : PubMed/NCBI

104 

Jin Y, Qiu J, Lu X and Li G: C-MYC inhibited ferroptosis and promoted immune evasion in ovarian cancer cells through NCOA4 mediated ferritin autophagy. Cells. 11:41272022. View Article : Google Scholar : PubMed/NCBI

105 

Li C, Sun G, Chen B, Xu L, Ye Y, He J, Bao Z, Zhao P, Miao Z, Zhao L, et al: Nuclear receptor coactivator 4-mediated ferritinophagy contributes to cerebral ischemia-induced ferroptosis in ischemic stroke. Pharmacol Res. 174:1059332021. View Article : Google Scholar : PubMed/NCBI

106 

Santana-Codina N, Gikandi A and Mancias JD: The role of NCOA4-mediated ferritinophagy in ferroptosis. Ferroptosis: Mechanism and Diseases. Vol. 1301. Florez AF and Alborzinia H: Springer International Publishing; Cham: pp. 41–57. 2021, PubMed/NCBI

107 

Fang Y, Chen X, Tan Q, Zhou H, Xu J and Gu Q: Inhibiting ferroptosis through disrupting the NCOA4-FTH1 interaction: A new mechanism of action. ACS Cent Sci. 7:980–989. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Santana-Codina N, Gableske S, Quiles del Rey M, Małachowska B, Jedrychowski MP, Biancur DE, Schmidt PJ, Fleming MD, Fendler W, Harper JW, et al: NCOA4 maintains murine erythropoiesis via cell autonomous and non-autonomous mechanisms. Haematologica. 104:1342–1354. 2019. View Article : Google Scholar : PubMed/NCBI

109 

Bellelli R, Federico G, Matte' A, Colecchia D, Iolascon A, Chiariello M, Santoro M, De Franceschi L and Carlomagno F: NCOA4 deficiency impairs systemic iron homeostasis. Cell Rep. 14:411–421. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Nai A, Lidonnici MR, Federico G, Pettinato M, Olivari V, Carrillo F, Geninatti Crich S, Ferrari G, Camaschella C, Silvestri L and Carlomagno F: NCOA4-mediated ferritinophagy in macrophages is crucial to sustain erythropoiesis in mice. Haematologica. 106:795–805. 2021.PubMed/NCBI

111 

Xu W, Guo ZN and Shao A: Editorial: Ferroptosis in stroke, neurotrauma and neurodegeneration, volume II. Front Cell Neurosci. 17:12384252023. View Article : Google Scholar : PubMed/NCBI

112 

Rochette L, Dogon G, Rigal E, Zeller M, Cottin Y and Vergely C: Lipid peroxidation and iron metabolism: Two corner stones in the homeostasis control of ferroptosis. Int J Mol Sci. 24:4492022. View Article : Google Scholar : PubMed/NCBI

113 

Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, Ding HF, Zhang J, Wang H, Chen X and Yan C: ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 27:662–675. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Verbruggen L, Sprimont L, Bentea E, Janssen P, Gharib A, Deneyer L, De Pauw L, Lara O, Sato H, Nicaise C and Massie A: Chronic sulfasalazine treatment in mice induces system xc-independent adverse effects. Front Pharmacol. 12:6256992021. View Article : Google Scholar : PubMed/NCBI

115 

de Baat A, Meier DT, Fontana A, Böni-Schnetzler M and Donath MY: Cystine/Glutamate antiporter system xc- deficiency impairs macrophage glutathione metabolism and cytokine production. PLoS One. 18:e02919502023. View Article : Google Scholar : PubMed/NCBI

116 

Sui X, Zhang R, Liu S, Duan T, Zhai L, Zhang M, Han X, Xiang Y, Huang X, Lin H and Xie T: RSL3 drives ferroptosis through GPX4 inactivation and ROS production in colorectal cancer. Front Pharmacol. 9:13712018. View Article : Google Scholar : PubMed/NCBI

117 

Cui C, Yang F and Li Q: Post-translational modification of GPX4 is a promising target for treating ferroptosis-related diseases. Front Mol Biosci. 9:9015652022. View Article : Google Scholar : PubMed/NCBI

118 

Zhou J, Zhang L, Yan J, Hou A, Sui W and Sun M: Curcumin induces ferroptosis in A549 CD133+ cells through the GSH-GPX4 and FSP1-CoQ10-NAPH pathways. Discov Med. 35:251–263. 2023. View Article : Google Scholar : PubMed/NCBI

119 

Almahi WA, Yu KN, Mohammed F, Kong P and Han W: Hemin enhances radiosensitivity of lung cancer cells through ferroptosis. Exp Cell Res. 410:1129462022. View Article : Google Scholar : PubMed/NCBI

120 

Ma S, Henson ES, Chen Y and Gibson SB: Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 7:e23072016. View Article : Google Scholar : PubMed/NCBI

121 

Zhang X, Yu K, Ma L, Qian Z, Tian X, Miao Y, Niu Y, Xu X, Guo S, Yang Y, et al: Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Theranostics. 11:5650–5674. 2021. View Article : Google Scholar : PubMed/NCBI

122 

Sun S, Guo C, Gao T, Ma D, Su X, Pang Q and Zhang R: Hypoxia enhances glioma resistance to sulfasalazine-induced ferroptosis by upregulating SLC7A11 via PI3K/AKT/HIF-1α axis. Oxid Med Cell Longev. 2022:78624302022. View Article : Google Scholar : PubMed/NCBI

123 

Zhao Y, Li Y, Zhang R, Wang F, Wang T and Jiao Y: The role of erastin in ferroptosis and its prospects in cancer therapy. Onco Targets Ther. 13:5429–5441. 2020. View Article : Google Scholar : PubMed/NCBI

124 

Zhang Y, Tan H, Daniels JD, Zandkarimi F, Liu H, Brown LM, Uchida K, O'Connor OA and Stockwell BR: Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem Biol. 26:623–633.e9. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Duan L, Zhang Y, Yang Y, Su S, Zhou L, Lo PC, Cai J, Qiao Y, Li M, Huang S, et al: Baicalin inhibits ferroptosis in intracerebral hemorrhage. Front Pharmacol. 12:6293792021. View Article : Google Scholar : PubMed/NCBI

126 

Li J, Wei G, Song Z, Chen Z, Gu J, Zhang L and Wang Z: SIRT5 regulates ferroptosis through the Nrf2/HO-1 signaling axis to participate in ischemia-reperfusion injury in ischemic stroke. Neurochem Res. 49:998–1007. 2024. View Article : Google Scholar : PubMed/NCBI

127 

Li Y, Bao Y, Li Y, Duan X, Dong S, Lin J, Chang X, Tan Y, Zhang H and Shan H: RSL3 inhibits porcine epidemic diarrhea virus replication by activating ferroptosis. Viruses. 15:20802023. View Article : Google Scholar : PubMed/NCBI

128 

Cheff DM, Huang C, Scholzen KC, Gencheva R, Ronzetti MH, Cheng Q, Hall MD and Arnér ESJ: The ferroptosis inducing compounds RSL3 and ML162 are not direct inhibitors of GPX4 but of TXNRD1. Redox Biol. 62:1027032023. View Article : Google Scholar : PubMed/NCBI

129 

Hu J, Gu W, Ma N, Fan X and Ci X: Leonurine alleviates ferroptosis in cisplatin-induced acute kidney injury by activating the Nrf2 signalling pathway. Br J Pharmacol. 179:3991–4009. 2022. View Article : Google Scholar : PubMed/NCBI

130 

Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, Jiang L and Ye L: Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 12:10792021. View Article : Google Scholar : PubMed/NCBI

131 

Li S, He Y, Chen K, Sun J, Zhang L, He Y, Yu H and Li Q: RSL3 drives ferroptosis through NF-κB pathway activation and GPX4 depletion in glioblastoma. Oxid Med Cell Longev. 2021:29150192021. View Article : Google Scholar : PubMed/NCBI

132 

Sun X, Huang N, Li P, Dong X, Yang J, Zhang X, Zong WX, Gao S and Xin H: TRIM21 ubiquitylates GPX4 and promotes ferroptosis to aggravate ischemia/reperfusion-induced acute kidney injury. Life Sci. 321:1216082023. View Article : Google Scholar : PubMed/NCBI

133 

Li M, Meng Z, Yu S, Li J, Wang Y, Yang W and Wu H: Baicalein ameliorates cerebral ischemia-reperfusion injury by inhibiting ferroptosis via regulating GPX4/ACSL4/ACSL3 axis. Chem Biol Interact. 366:1101372022. View Article : Google Scholar : PubMed/NCBI

134 

Nakamura T, Hipp C, Santos Dias Mourão A, Borggräfe J, Aldrovandi M, Henkelmann B, Wanninger J, Mishima E, Lytton E, Emler D, et al: Phase separation of FSP1 promotes ferroptosis. Nature. 619:371–377. 2023. View Article : Google Scholar : PubMed/NCBI

135 

Yao X, Xie R, Cao Y, Tang J, Men Y, Peng H and Yang W: Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J Nanobiotechnology. 19:3112021. View Article : Google Scholar : PubMed/NCBI

136 

Shu X and Wu J, Zhang T, Ma X, Du Z, Xu J, You J, Wang L, Chen N, Luo M and Wu J: Statin-induced geranylgeranyl pyrophosphate depletion promotes ferroptosis-related senescence in adipose tissue. Nutrients. 14:43652022. View Article : Google Scholar : PubMed/NCBI

137 

Miyamoto HD, Ikeda M, Ide T, Tadokoro T, Furusawa S, Abe K, Ishimaru K, Enzan N, Sada M, Yamamoto T, et al: Iron overload via heme degradation in the endoplasmic reticulum triggers ferroptosis in myocardial ischemia-reperfusion injury. JACC Basic Transl Sci. 7:800–819. 2022. View Article : Google Scholar : PubMed/NCBI

138 

Chen GQ, Benthani FA, Wu J, Liang D, Bian ZX and Jiang X: Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 27:242–254. 2020. View Article : Google Scholar : PubMed/NCBI

139 

Pei X, Tian M, Wang Y, Xin Y, Jiang J, Wang Y and Gong Y: Advances in the knowledge on the role of apoptosis repressor with caspase recruitment domain in hemorrhagic stroke. J Intensive Med. 3:138–143. 2023. View Article : Google Scholar : PubMed/NCBI

140 

Zille M, Karuppagounder SS, Chen Y, Gough PJ, Bertin J, Finger J, Milner TA, Jonas EA and Ratan RR: Neuronal death after hemorrhagic stroke in vitro and in vivo shares features of ferroptosis and necroptosis. Stroke. 48:1033–1043. 2017. View Article : Google Scholar : PubMed/NCBI

141 

Fu K, Xu W, Lenahan C, Mo Y, Wen J, Deng T, Huang Q, Guo F, Mo L and Yan J: Autophagy regulates inflammation in intracerebral hemorrhage: Enemy or friend? Front Cell Neurosci. 16:10363132023. View Article : Google Scholar : PubMed/NCBI

142 

Wang X, Mori T, Sumii T and Lo EH: Hemoglobin-induced cytotoxicity in rat cerebral cortical neurons: Caspase activation and oxidative stress. Stroke. 33:1882–1888. 2002. View Article : Google Scholar : PubMed/NCBI

143 

Xu Y, Liu Y, Li K, Yuan D, Yang S, Zhou L, Zhao Y, Miao S, Lv C and Zhao J: COX-2/PGE2 pathway inhibits the ferroptosis induced by cerebral ischemia reperfusion. Mol Neurobiol. 59:1619–1631. 2022. View Article : Google Scholar : PubMed/NCBI

144 

Dendorfer A, Heidbreder M, Hellwig-Bürgel T, Jöhren O, Qadri F and Dominiak P: Deferoxamine induces prolonged cardiac preconditioning via accumulation of oxygen radicals. Free Radic Biol Med. 38:117–124. 2005. View Article : Google Scholar : PubMed/NCBI

145 

Wang C, Xie L, Xing Y, Liu M, Yang J, Gao N and Cai Y: Iron-overload-induced ferroptosis in mouse cerebral toxoplasmosis promotes brain injury and could be inhibited by Deferiprone. PLoS Negl Trop Dis. 17:e00116072023. View Article : Google Scholar : PubMed/NCBI

146 

Miotto G, Rossetto M, Di Paolo ML, Orian L, Venerando R, Roveri A, Vučković AM, Bosello Travain V, 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

147 

Gao Z, Zhang Z, Gu D, Li Y, Zhang K, Dong X, Liu L, Zhang J, Chen J, Wu D and Zeng M: Hemin mitigates contrast-induced nephropathy by inhibiting ferroptosis via HO-1/Nrf2/GPX4 pathway. Clin Exp Pharma Physio. 49:858–870. 2022. View Article : Google Scholar : PubMed/NCBI

148 

Mishima E, Ito J, Wu Z, Nakamura T, Wahida A, Doll S, Tonnus W, Nepachalovich P, Eggenhofer E, Aldrovandi M, et al: A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature. 608:778–783. 2022. View Article : Google Scholar : PubMed/NCBI

149 

Sun W, Lv Z, Li W, Lu J, Xie Y, Wang P, Jiang R, Dong J, Guo H, Liu Z, et al: XJB-5-131 protects chondrocytes from ferroptosis to alleviate osteoarthritis progression via restoring Pebp1 expression. J Orthop Translat. 44:114–124. 2024. View Article : Google Scholar : PubMed/NCBI

150 

Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al: The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 575:688–692. 2019. View Article : Google Scholar : PubMed/NCBI

151 

Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al: ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 13:91–98. 2017. View Article : Google Scholar : PubMed/NCBI

152 

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

153 

Kosyakovsky J, Fine JM, Frey WH II and Hanson LR: Mechanisms of intranasal deferoxamine in neurodegenerative and neurovascular disease. Pharmaceuticals (Basel). 14:952021. View Article : Google Scholar : PubMed/NCBI

154 

Huang Z and Huang S: Reposition of the fungicide ciclopirox for cancer treatment. Recent Pat Anticancer Drug Discov. 16:122–135. 2021. View Article : Google Scholar : PubMed/NCBI

155 

Averill-Bates DA: The antioxidant glutathione. Vitam Horm. 121:109–141. 2023. View Article : Google Scholar : PubMed/NCBI

156 

Chen J, Yang L, Geng L, He J, Chen L, Sun Q, Zhao J and Wang X: Inhibition of Acyl-CoA synthetase long-chain family member 4 facilitates neurological recovery after stroke by regulation ferroptosis. Front Cell Neurosci. 15:6323542021. View Article : Google Scholar : PubMed/NCBI

157 

Shen L, Lin D, Li X, Wu H, Lenahan C, Pan Y, Xu W, Chen Y, Shao A and Zhang J: Ferroptosis in acute central nervous system injuries: The future direction? Front Cell Dev Biol. 8:5942020. View Article : Google Scholar : PubMed/NCBI

158 

Guo Z, Lin J, Sun K, Guo J, Yao X, Wang G, Hou L, Xu J, Guo J and Guo F: Deferoxamine alleviates osteoarthritis by inhibiting chondrocyte ferroptosis and activating the Nrf2 pathway. Front Pharmacol. 13:7913762022. View Article : Google Scholar : PubMed/NCBI

159 

Xue Q, Yan D, Chen X, Li X, Kang R, Klionsky DJ, Kroemer G, Chen X, Tang D and Liu J: Copper-dependent autophagic degradation of GPX4 drives ferroptosis. Autophagy. 19:1982–1996. 2023. View Article : Google Scholar : PubMed/NCBI

160 

Wang L, Zhang X, Xu M, Zheng G, Chen J, Li S, Cui J and Zhang S: Implication of ferroptosis in hepatic toxicity upon single or combined exposure to polystyrene microplastics and cadmium. Environ Pollut. 334:1222502023. View Article : Google Scholar : PubMed/NCBI

161 

Chen Y, He W, Wei H, Chang C, Yang L, Meng J, Long T, Xu Q and Zhang C: Srs11-92, a ferrostatin-1 analog, improves oxidative stress and neuroinflammation via Nrf2 signal following cerebral ischemia/reperfusion injury. CNS Neurosci Ther. 29:1667–1677. 2023. View Article : Google Scholar : PubMed/NCBI

162 

Holden P and Nair LS: Deferoxamine: An angiogenic and antioxidant molecule for tissue regeneration. Tissue Eng Part B Rev. 25:461–470. 2019. View Article : Google Scholar : PubMed/NCBI

163 

Kim JL, Lee DH, Na YJ, Kim BR, Jeong YA, Lee SI, Kang S, Joung SY, Lee SY, Oh SC and Min BW: Iron chelator-induced apoptosis via the ER stress pathway in gastric cancer cells. Tumor Biol. 37:9709–9719. 2016. View Article : Google Scholar : PubMed/NCBI

164 

Lin S, Gao W, Zhu C, Lou Q, Ye C, Ren Y, Mehmood R, Huang B and Nan K: Efficiently suppress of ferroptosis using deferoxamine nanoparticles as a new method for retinal ganglion cell protection after traumatic optic neuropathy. Biomater Adv. 138:2129362022. View Article : Google Scholar : PubMed/NCBI

165 

You H, Wang D, Wei L, Chen J, Li H and Liu Y: Deferoxamine inhibits acute lymphoblastic leukemia progression through repression of ROS/HIF-1α, Wnt/β-catenin, and p38MAPK/ERK pathways. J Oncol. 2022:82812672022. View Article : Google Scholar : PubMed/NCBI

166 

Abdul Y, Li W, Ward R, Abdelsaid M, Hafez S, Dong G, Jamil S, Wolf V, Johnson MH, Fagan SC and Ergul A: Deferoxamine treatment prevents post-stroke vasoregression and neurovascular unit remodeling leading to improved functional outcomes in type 2 male diabetic rats: Role of endothelial ferroptosis. Transl Stroke Res. 12:615–630. 2021. View Article : Google Scholar : PubMed/NCBI

167 

Jones G, Zeng L and Kim J: Mechanism-based pharmacokinetic modeling of absorption and disposition of a deferoxamine-based nanochelator in rats. Mol Pharm. 20:481–490. 2023. View Article : Google Scholar : PubMed/NCBI

168 

Chen X, Li D, Sun H, Wang W, Wu H and Kong W and Kong W: Relieving ferroptosis may partially reverse neurodegeneration of the auditory cortex. FEBS J. 287:4747–4766. 2020. View Article : Google Scholar : PubMed/NCBI

169 

Tuo Q, 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

170 

Kannan M, Sil S, Oladapo A, Thangaraj A, Periyasamy P and Buch S: HIV-1 Tat-mediated microglial ferroptosis involves the miR-204-ACSL4 signaling axis. Redox Biol. 62:1026892023. View Article : Google Scholar : PubMed/NCBI

171 

Li Y, Zeng X, Lu D, Yin M, Shan M and Gao Y: Erastin induces ferroptosis via ferroportin-mediated iron accumulation in endometriosis. Hum Reprod. 36:951–964. 2021. View Article : Google Scholar : PubMed/NCBI

172 

Xie B, Wang Y, Lin Y, Mao Q, Feng J, Gao G and Jiang J: Inhibition of ferroptosis attenuates tissue damage and improves long-term outcomes after traumatic brain injury in mice. CNS Neurosci Ther. 25:465–475. 2019. View Article : Google Scholar : PubMed/NCBI

173 

Carbonell T and Rama R: Iron, oxidative stress and early neurological deterioration in ischemic stroke. Curr Med Chem. 14:857–874. 2007. View Article : Google Scholar : PubMed/NCBI

174 

Silver MK, Lozoff B and Meeker JD: Blood cadmium is elevated in iron deficient U.S. children: A cross-sectional study. Environ Health. 12:1172013. View Article : Google Scholar : PubMed/NCBI

175 

Zhang Q, Luo C, Li Z, Huang W, Zheng S, Liu C, Shi X, Ma Y, Ni Q, Tan W, et al: Astaxanthin activates the Nrf2/Keap1/HO-1 pathway to inhibit oxidative stress and ferroptosis, reducing triphenyl phosphate (TPhP)-induced neurodevelopmental toxicity. Ecotoxicol Environ Saf. 271:1159602024. View Article : Google Scholar : PubMed/NCBI

176 

Liu X, Du Y, Liu J, Cheng L, He W and Zhang W: Ferrostatin-1 alleviates cerebral ischemia/reperfusion injury through activation of the AKT/GSK3β signaling pathway. Brain Res Bull. 193:146–157. 2023. View Article : Google Scholar : PubMed/NCBI

177 

Chen B and Jin W: A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci. 17:12000612023. View Article : Google Scholar : PubMed/NCBI

178 

Lou Y, Ma M, Jiang Y, Xu H, Gao Z, Gao L and Wang Y: Ferroptosis: A new strategy for traditional Chinese medicine treatment of stroke. Biomed Pharmacother. 156:1138062022. View Article : Google Scholar : PubMed/NCBI

179 

Li J, Zhao X, Zhang Y, Wan H, He Y, Li X, Yu L and Jin W: Comparison of traditional Chinese medicine in the long-term secondary prevention for patients with ischemic stroke: A systematical analysis. Front Pharmacol. 12:7229752021. View Article : Google Scholar : PubMed/NCBI

180 

Zhu T, Wang L, Wang L and Wan Q: Therapeutic targets of neuroprotection and neurorestoration in ischemic stroke: Applications for natural compounds from medicinal herbs. Biomed Pharmacother. 148:1127192022. View Article : Google Scholar : PubMed/NCBI

181 

Wang J, Hu J, Chen X, Lei X, Feng H, Wan F and Tan L: Traditional Chinese medicine monomers: Novel strategy for endogenous neural stem cells activation after stroke. Front Cell Neurosci. 15:6281152021. View Article : Google Scholar : PubMed/NCBI

182 

Zhan S, Liang J, Lin H, Cai J, Yang X, Wu H, Wei J, Wang S and Xian M: SATB1/SLC7A11/HO-1 axis ameliorates ferroptosis in neuron cells after ischemic stroke by danhong injection. Mol Neurobiol. 60:413–427. 2023. View Article : Google Scholar : PubMed/NCBI

183 

Ko G, Kim J, Jeon YJ, Lee D, Baek HM and Chang KA: Salvia miltiorrhiza alleviates memory deficit induced by ischemic brain injury in a transient MCAO mouse model by inhibiting ferroptosis. Antioxidants (Basel). 12:7852023. View Article : Google Scholar : PubMed/NCBI

184 

Zhang J, Cai W, Wei X, Shi Y, Zhang K, Hu C, Wan J, Luo K and Shen W: Moxibustion ameliorates cerebral ischemia-reperfusion injury by regulating ferroptosis in rats. Clin Exp Pharmacol Physiol. 50:779–788. 2023. View Article : Google Scholar : PubMed/NCBI

185 

Liu J, Jiang G, He P, Du X, Hu Z and Li F: Mechanism of ferroptosis in traditional chinese medicine for clinical treatment: A review. Front Pharmacol. 13:11088362023. View Article : Google Scholar : PubMed/NCBI

186 

Wang L, Liu C, Wang L and Tang B: Astragaloside IV mitigates cerebral ischaemia-reperfusion injury via inhibition of P62/Keap1/Nrf2 pathway-mediated ferroptosis. Eur J Pharmacol. 944:1755162023. View Article : Google Scholar : PubMed/NCBI

187 

Jiang Y, Zhao S, Zhou Y and Wei Z: Research progress of traditional Chinese medicine in ferroptosis-related diseases. Med Nov Technol Devices. 16:1001932022. View Article : Google Scholar : PubMed/NCBI

188 

Yang K, Zeng L, Zeng J, Deng Y, Wang S, Xu H, He Q, Yuan M, Luo Y, Ge A and Ge J: Research progress in the molecular mechanism of ferroptosis in Parkinson's disease and regulation by natural plant products. Ageing Res Rev. 91:1020632023. View Article : Google Scholar : PubMed/NCBI

189 

Wu C, Duan F, Yang R, Dai Y, Chen X and Li S: 15, 16-Dihydrotanshinone I protects against ischemic stroke by inhibiting ferroptosis via the activation of nuclear factor erythroid 2-related factor 2. Phytomedicine. 114:1547902023. View Article : Google Scholar : PubMed/NCBI

190 

Bai X, Zheng E, Tong L, Liu Y, Li X and Yang H, Jiang J, Chang Z and Yang H: Angong Niuhuang Wan inhibit ferroptosis on ischemic and hemorrhagic stroke by activating PPARγ/AKT/GPX4 pathway. J Ethnopharmacol. 321:1174382024. View Article : Google Scholar : PubMed/NCBI

191 

Jin Z, Gao W, Guo F, Liao S, Hu M, Yu T, Yu S and Shi Q: Astragaloside IV alleviates neuronal ferroptosis in ischemic stroke by regulating fat mass and obesity-associated-N6-methyladenosine-acyl-CoA synthetase long-chain family member 4 axis. J Neurochem. 166:328–345. 2023. View Article : Google Scholar : PubMed/NCBI

192 

Hirata Y, Cai R, Volchuk A, Steinberg BE, Saito Y, Matsuzawa A, Grinstein S and Freeman SA: Lipid peroxidation increases membrane tension, Piezo1 gating, and cation permeability to execute ferroptosis. Curr Biol. 33:1282–1294.e5. 2023. View Article : Google Scholar : PubMed/NCBI

193 

Gao W, Wang X, Zhou Y, Wang X and Yu Y: Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Sig Transduct Target Ther. 7:1962022. View Article : Google Scholar : PubMed/NCBI

194 

Zhou Y, Liao J, Mei Z, Liu X and Ge J: Insight into crosstalk between ferroptosis and necroptosis: Novel therapeutics in ischemic stroke. Oxid Med Cell Longev. 2021:99910012021. View Article : Google Scholar : PubMed/NCBI

195 

Yu Y, Yan Y, Niu F, Wang Y, Chen X, Su G, Liu Y, Zhao X, Qian L, Liu P and Xiong Y: Ferroptosis: A cell death connecting oxidative stress, inflammation and cardiovascular diseases. Cell Death Discov. 7:1932021. View Article : Google Scholar : PubMed/NCBI

196 

Battaglia AM, Chirillo R, Aversa I, Sacco A, Costanzo F and Biamonte F: Ferroptosis and cancer: Mitochondria meet the ‘iron maiden’ cell death. Cells. 9:15052020. View Article : Google Scholar : PubMed/NCBI

197 

Jeong SY and Seol DW: The role of mitochondria in apoptosis. BMB Rep. 41:11–22. 2008. View Article : Google Scholar : PubMed/NCBI

198 

Cui Y, Zhang Y, Zhao X, Shao L, Liu G, Sun C, Xu R and Zhang Z: ACSL4 exacerbates ischemic stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav Immun. 93:312–321. 2021. View Article : Google Scholar : PubMed/NCBI

199 

Wang ZL, Yuan L, Li W and Li JY: Ferroptosis in Parkinson's disease: Glia-neuron crosstalk. Trends Mol Med. 28:258–269. 2022. View Article : Google Scholar : PubMed/NCBI

200 

Ren JX, Sun X, Yan XL, Guo ZN and Yang Y: Ferroptosis in neurological diseases. Front Cell Neurosci. 14:2182020. View Article : Google Scholar : PubMed/NCBI

201 

Chen Y, Long T, Xu Q and Zhang C: Bibliometric analysis of ferroptosis in stroke from 2013 to 2021. Front Pharmacol. 12:8173642022. View Article : Google Scholar : PubMed/NCBI

202 

Jin Y, Zhuang Y, Liu M, Che J and Dong X: Inhibiting ferroptosis: A novel approach for stroke therapeutics. Drug Discov Today. 26:916–930. 2021. View Article : Google Scholar : PubMed/NCBI

203 

Yang K, Zeng L, Yuan X, Wang S, Ge A, Xu H, Zeng J and Ge J: The mechanism of ferroptosis regulating oxidative stress in ischemic stroke and the regulation mechanism of natural pharmacological active components. Biomed Pharmacother. 154:1136112022. View Article : Google Scholar : PubMed/NCBI

204 

Xing G, Meng L, Cao S, Liu S, Wu J, Li Q, Huang W and Zhang L: PPARα alleviates iron overload-induced ferroptosis in mouse liver. EMBO Rep. 23:e522802022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2024
Volume 30 Issue 1

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
Dong H, Ma Y, Cui M, Qiu Z, He M and Zhang B: Recent advances in potential therapeutic targets of ferroptosis‑associated pathways for the treatment of stroke (Review). Mol Med Rep 30: 128, 2024
APA
Dong, H., Ma, Y., Cui, M., Qiu, Z., He, M., & Zhang, B. (2024). Recent advances in potential therapeutic targets of ferroptosis‑associated pathways for the treatment of stroke (Review). Molecular Medicine Reports, 30, 128. https://doi.org/10.3892/mmr.2024.13252
MLA
Dong, H., Ma, Y., Cui, M., Qiu, Z., He, M., Zhang, B."Recent advances in potential therapeutic targets of ferroptosis‑associated pathways for the treatment of stroke (Review)". Molecular Medicine Reports 30.1 (2024): 128.
Chicago
Dong, H., Ma, Y., Cui, M., Qiu, Z., He, M., Zhang, B."Recent advances in potential therapeutic targets of ferroptosis‑associated pathways for the treatment of stroke (Review)". Molecular Medicine Reports 30, no. 1 (2024): 128. https://doi.org/10.3892/mmr.2024.13252