Open Access

Potential intervention target of atherosclerosis: Ferroptosis (Review)

  • Authors:
    • Jia Li
    • Ling Xu
    • Yi Xuan Zuo
    • Xue Qin Chang
    • Hai Tao Chi
  • View Affiliations

  • Published online on: September 21, 2022     https://doi.org/10.3892/mmr.2022.12859
  • Article Number: 343
  • Copyright: © Li 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

Atherosclerosis (AS) is a chronic inflammatory disease of the blood vessels, which is mainly characterized by the form of atherosclerotic plaques and vascular endothelial injury. Its formation involves abnormal lipid metabolism, oxidative stress and inflammation, as well as other processes. AS is the direct cause of various acute cardiovascular and cerebrovascular diseases, such as acute myocardial infarction and acute ischemic stroke. Early intervention in the atherosclerotic inflammatory process and lesion progression is beneficial, and has been associated with the primary prevention of a range of related diseases. Ferroptosis is a non‑apoptotic form of cell death different from cell necrosis and autophagy, which has been shown to participate in atherogenesis and atherosclerotic progression through numerous signaling pathways. The main characteristic of ferroptosis is the formation of high levels of cellular iron catalytic free radicals, unsaturated fatty acid accumulation and iron‑induced lipid reactive oxygen species accumulation, which can cause oxidative stress, and subsequent DNA, protein and lipid damage. There are numerous hypotheses about the pathogenesis of AS. At present, it has been suggested that ferroptosis can accelerate the progression of AS and that inflammation is associated with the whole process of AS. The mechanisms and signaling pathways related to the involvement of neuroinflammation and ferroptosis in the progression of AS, and therapeutic targets associated with ferroptosis have not yet been elucidated. The present review article evaluated the involvement of ferroptosis in the progression of AS from the perspectives of ferroptotic cell death, the pathogenesis of AS and nervous system inflammation, with the aim of exploring new therapeutic targets for AS.

Introduction

Ferroptosis is an iron-dependent lipid reactive oxygen species (ROS)-induced form of non-apoptotic cell death, first proposed by Dixon et al in 2012 (1). This previous study revealed that the Ras-selective lethal molecule erastin can affect glutathione (GSH) peroxidase (GPX) activity, leading to redox imbalance, ROS accumulation, membrane lipid peroxidation and ultimately to the destruction of the integrity of the cell membrane (1). Ferroptosis induces morphological changes that manifest as smaller mitochondria, increased mitochondrial membrane density, decreased or lack of mitochondrial cristae, and outer membrane breaks (2). The main characteristic of ferroptosis is redox imbalance and the whole ferroptosis process, which involves iron metabolism, lipid metabolism, oxidative stress mechanisms, and the biosynthesis of nicotinamide adenine dinucleotide phosphate, GSH and coenzyme Q10 (3), participates in the development and progression of atherosclerosis (AS) (4). AS is characterized by the accumulation of lipids in the arteries and the formation of plaques. As the disease progresses, the blood vessels narrow, and resulting in blood flow restriction or even blockage (5). In 1999, Ross (6) proposed that AS is a chronic and progressive inflammatory disease, the mechanism of which involves endothelial cell dysfunction and lipid accumulation, and that AS is an important pathological cause of acute cardiovascular and cerebrovascular diseases.

Epidemiological studies have reported that the incidence rate of cardiovascular and cerebrovascular diseases caused by AS has increased year on year, and that it has become the primary cause of death in China and also worldwide (7). Notably, >75% of disabling and fatal cardiovascular and cerebrovascular diseases in China are caused by AS (8). Intracranial atherosclerotic stenosis (ICAS) is an important cause of ischemic stroke. A previous study on symptomatic intracranial arterial stenosis and occlusion in China reported that the incidence of intracranial AS in patients with ischemic stroke or transient ischemic attack was 46.6% (9), among which patients with ICAS had more severe symptoms, longer hospital stays and higher stroke recurrence rates; moreover, the recurrence rate increased with stenosis severity. Worldwide, ~2 billion individuals have carotid AS and the incidence of AS is higher among male individuals than female individuals (7). AS is closely related to blood pressure, blood sugar and blood lipid levels, smoking and obesity among other risk factors, which over a period of 20–30 years can gradually lead to vascular stenosis, causing a range of vascular diseases.

AS is an inflammatory lesion caused by numerous risk factors that damage vascular endothelial cells and is mainly characterized by disordered lipid metabolism. It is well known that lipid peroxidation, namely lipid oxidation degradation, generates lipid peroxide radicals and hydrogen peroxide that serve crucial roles in AS by causing inflammation and endothelial dysfunction, whereas lipid peroxidation is also a core feature of ferroptosis (10). Therefore, ferroptosis may be a key factor in the occurrence and development of AS. Ferroptosis is associated with various stages of AS development through numerous physiological mechanisms, such as iron ion metabolism, lipid metabolism and amino acid metabolism (11). Atherosclerotic stenosis is the root cause of vascular-related diseases, such as coronary heart disease, stroke and peripheral vascular disease. In particular, the stability of neck AS plaques has been reported to be closely related to the occurrence of ischemic stroke (12). According to global data (13,14), stroke currently remains the second most common cause of mortality and disability, with ischemic stroke accounting for 62.4% of all new stroke cases in 2019, thus placing a heavy burden on society and the families of patients. A previous meta-analysis on the subtypes of ischemic stroke demonstrated that ischemic stroke due to large artery atheromatous sclerosis accounted for 23% of cases and that the occurrence of cardiogenic stroke was also closely related to major AS in the Asian population (15). Therefore, a more precise understanding of the causes and mechanisms of AS progression may be beneficial for the treatment of patients.

Iron and ferroptosis

Regulation of iron homeostasis

Iron is the most abundant transition element in the brain; it participates in the oxidation reaction, myelination, neurotransmitter synthesis and metabolism, and it serves a key role in cell respiration and energy generation (16). The body ingests iron through the diet, and intestinal epithelial cells absorb iron and transport it to the blood (17). In addition, macrophages phagocytose aged red blood cells, which increases the iron content in the blood (18), which has been reported to be an independent predictor of vascular damage (19). Extracellular Fe3+ is transported into cells by transferrin receptor 1 (TfR1) and is converted into Fe2+, which participates in the synthesis of heme in mitochondria (20) or is exported from cells by ferroportin; iron is also stored in monocyte macrophages in the form of ferritin, which helps to regulate iron storage and balance in the body (Fig. 1). The intracellular iron content is in a dynamic equilibrium and reduction of the iron content in the body is protective (21). Nuclear receptor coactivator 4 (NCOA4) can recognize ferritin and promote its autophagic degradation, leading to the release of free iron (22). Therefore, knocking out the NCOA4 gene can block ferritin deposition and avoid the accumulation of free iron, thus protecting neurons from ferroptosis. Iron is transported from the cytosol to the mitochondria by mitochondrial ferritin (FTMT) and is used to synthesis iron porphyrin to reduce free iron levels and protect mitochondria from oxidative stress. Moreover, the upregulation of FTMT can increase iron consumption and inhibit ferroptosis (23). It has been reported that cyanidin-3-glucoside can inhibit the occurrence of ferroptosis, affecting ischemia-reperfusion injury in the myocardium by alleviating oxidative stress, reducing the free iron content and downregulating TfR1 expression in cells and tissues (24).

The imbalance of iron metabolism mediates the occurrence of ferroptosis, and promotes the formation and development of AS. Initially, a toxic iron reaction is associated with an excess of stored iron (25), which can accelerate cerebral tissue oxidation by increasing oxygen radical generation (26). This leads to super-oxidation damage in the inner wall of the cerebral artery, further aggravating the iron overload through the oxidized low-density lipoprotein (ox-LDL)-mediated Toll-like receptor 4 (TLR4)/nuclear factor κB (NF-κB) signaling pathway in macrophages (27). Moreover, the occurrence of ferroptosis activates the TLR4/NF-κB signaling pathway and can increase the expression of pro-inflammatory cytokine genes (28). When intracellular free iron levels increase, the intracellular labile iron pool also increases (3). Excess divalent iron may be involved in the Fenton reaction and other lipid peroxidation processes that induce ROS overproduction and may be involved in oxidative stress, with ROS promoting lipid peroxidation and inducing ferroptosis (29). ROS mainly include oxygen molecules, hydroxyl radicals, superoxide anions and hydrogen peroxide radicals. Furthermore, iron is a potent oxidant, which through the Haber-Weiss reaction catalyzed by a large number of ROS, promotes the intracellular lipid peroxidation reaction. The intracellular lipid peroxidation reaction causes damage to proteins and nucleic acids, promotes macrophage apoptosis and leads to the release of numerous components of the cell contents; these substances can further promote the infiltration of macrophages and enhance the lipid peroxidation reaction (30). LDL-C passes through damaged endothelial cells and enters macrophages through oxidation, resulting in the formation of foam cells. The aggregation of foam cells becomes the lipid core of atherosclerotic plaques, which aggravates cell damage and lipid peroxidation, increasing the production of ROS and lipid peroxide products, such as MDA (31). In addition, activated macrophages releases inflammatory factors, such as TNF-α, IL-6 and IL-1β, increase the inflammatory response, mediate the oxidation of lipoprotein, and further accelerate the occurrence of AS.

Lipid metabolism pathway

Lipid peroxidation is the core process of ferroptosis; specifically, the peroxidation of cell membrane phospholipids by free radical-driven arachidonate lipoxygenases (ALOXs) (32). Concurrently, iron is associated with various stages of lipid peroxide generation, including iron-catalyzed lipid oxidation and esterification, the oxidation of polyunsaturated fatty acids and lipid ROS generation via the Fenton reaction. Mitochondria are the main organelles where iron utilization occurs in catabolic and anabolic pathways, and they serve an important role in iron metabolism, as well as material and energy metabolism (33). The Fenton reaction and other peroxidation processes that incorporate iron (Fig. 2) can convert the mitochondrial oxidation respiration product, hydrogen peroxide, into hydroxyl radicals through the catalysis of ferrous ions. During the oxidative phosphorylation of the electron transport chain, which takes place on the inner mitochondrial membrane, electrons leak from the complex and oxygen forms ROS through a series of redox processes (34). Excessive ROS generation can cause DNA damage, protein degeneration, lipid peroxidation and can induce ferroptotic cell death. However, ROS derived from mitochondria can also activate NLRP3 inflammatory bodies and lead to the activation of the iron death signaling pathway (35). Therefore, as a mitochondrially derived antioxidant, the free radical scavenger MitoQ can reduce ROS production by inhibiting mitochondrial respiration and enhancing glycolytic function, thus protecting the mitochondria and preventing GPX4-dependent ferroptosis (36). XJB-5-131 has been reported to have a dual antioxidant effect, which can scavenge free radicals by targeting mitochondria and inhibit DNA damage, providing protection from ischemia-reperfusion-induced kidney injury and inflammation in mice (37). Recently published research has focused on the ROS produced by mitochondria, which may promote the occurrence of ferroptosis by enhancing lipid peroxidation, heme degradation and free iron overload, promoting ferroptosis through HMOX-1 in the mitochondria (2). However, this process involves the participation of multiple signaling pathways, and these factors involved may have cross or offset effects, and the clear participating mechanism requires further assessment in animal experiments. Mitochondria-targeting antioxidants have been reported to be effective in animal experiments (38); they can attenuate kidney injury and promote tubular epithelial cells repair after ischemia/reperfusion injury, and the overexpression of mitochondrial ferritin has been shown to inhibit ferroptosis by promoting the storage of mitochondrial iron.

GPX4 is a GPX that catalyzes the reduction of hydrogen peroxide, organic hydroperoxide and lipid hydroperoxides, thereby protecting cells from oxidative damage and ferroptosis (39). As a ferroptosis inducer, Ras-selective lethal small molecule 3 directly binds and inhibits GPX4 activity, and can cause lipid ROS accumulation, mitochondrial damage, disruption of ATP production, lipid peroxide accumulation in cells and the promotion of ferroptosis (40). Acyl-CoA synthetase long chain family member 4 (ACSL4) converts free arachidonic acid into arachidonic arachidonoyl-CoA (41) and promotes unsaturated phospholipid production, the main substrate for lipid peroxidation (42). GPX4 can reduce lipid peroxide, and following GPX4 inhibition (43), ACSL4 is considered to be required for the occurrence of ferroptosis. Furthermore, microRNA-17-92 can protect endothelial cells from ferroptosis in AS by mediating the A20-ACSL4 axis (44). Doll et al (45) reported that knockdown of the ACLS4 gene significantly inhibited ferroptotic cell death, thus suggesting that ACLS4 may participate in the ferroptosis cascade via the lipid oxidation pathway. ACSL4 may also aggravate neuronal damage through neuronal ferroptosis and promote the release of pro-inflammatory cytokines from microglia (46). Baicalin has been reported to inhibit ROS production, reduce ACSL4 expression, and mediate iron uptake and autophagic degradation of ferritin to reduce intracellular iron levels, which may participate in the prevention of myocardial ischemia/reperfusion injury through anti-ferroptotic mechanisms (47).

Ebselen (Ebs) is a small molecule organo-selenium compound, which simulates GPX activity. As a lipid-soluble compound, Ebs can easily enter the cell through the cell membrane to exert antioxidant effects (48). Ebs can also inhibit the activity of ALOX5 and ALOX15, and the synthesis of leukotriene; inhibition of lipoxygenase and factors such as leukotriene are important for preventing AS and inhibiting inflammation. It has been reported that promoting the phosphorylation of AKT can increase the expression of endothelial nitric oxide synthase in vascular endothelial cells, increase nitric oxide release, and protect against myocardial ischemia and reperfusion injury, while also reducing oxidative stress and protecting the myocardial mitochondria (49).

Amino acid metabolism pathway

Under normal physiological conditions (50), extracellular cystine is imported into cells in exchange for intracellular glutamate via the cystine/glutamate antiporter system xc, maintaining glutamate balance inside and outside the cell. Intracellular cystine is converted into cysteine by cysteine reductase, and GSH is generated by glutamate-cysteine ligase and GSH synthetase. With antioxidants, GPX4 prevents ferroptosis by reducing lipid peroxides to the alcohol form using GSH (Fig. 2). Furthermore, intracellular cysteine deficiency caused by cysteine uptake disorder can lead to the depletion of the antioxidant peptide, GSH, which is composed of glutamate, cysteine and glycine, thus also leading to GPX4 inactivation and peroxide accumulation at lethal levels (Fig. 2). The depletion of GSH also leads to glutamate-mediated iron and ROS generation, and triggers oxidative cell death; specifically, ferroptosis through the amino acid metabolic pathway (51). GSH levels have been reported to be reduced in a mouse model of middle cerebral artery occlusion where the infarct size was reduced following intervention with the ferroptosis-related inhibitors, liproxstatin-1 and ferrostatin-1 (46). Carvacrol has also been reported to reduce the level of lipid peroxide in the ischemic brain tissue of gerbils by increasing the expression of GPX4, inhibiting ferroptosis, reducing cell death, and conserving the memory and learning ability of gerbils following ischemia-reperfusion (52).

It has been reported that the deposition of iron at atherosclerotic plaques leads to the accumulation of ROS and the death of macrophages; therefore, the loss of the antioxidant capacity of macrophages directly leads to ferroptosis and plaque formation (3). GSH, as a tripeptide antioxidant and a cofactor of GPX4,is a key substrate for GPX4, which can reduce the production of lipid peroxide. The cell defense against lipid peroxidation is decreased due to the depletion of GSH; however, cells do not reduce the amount of ROS produced by Fenton reactions and other iron peroxidation reactions in response to GSH depletion and therefore they are more sensitive to ferroptosis (53). Erastin functions as a ferroptosis inducer (54) that restrains the activity of the xc system, inhibiting cystine uptake, which means that the cysteine in the cells cannot be used for GSH synthesis. Intracellular reduced-GSH and oxidized GSH depletion (55), the accumulation of peroxidized phospholipids, the accumulation of lipid ROS and protein or membrane damage can all trigger ferroptotic cell death. GPX4 is a peroxide inhibitor protein discovered in 1982, which belongs to the seleno-proteins, that produces water or alcohol and protects cells by catalyzing certain reducing reactions of hydrogen peroxide; therefore, a single dose of selenium delivered to the brain can promote antioxidant GPX4 expression, protect neurons and inhibit plaque growth in AS (56). Impaired GPX4 activity caused by GPX4 deficiency or GSH depletion can lead to the inactivation of T cells and promote ferroptosis; however, it can also promote the differentiation of peripheral blood monocytes into B-cells and natural killer cells (51).

Solute carrier family 7 member 11 (SLC7A11, also known as xCT) is a key component of the cystine/glutamate antiporter system xc, which transports extracellular cystine into cells in exchange for intracellular glutamate, maintaining the glutamate balance inside and outside the cell. The inhibition of cysteine-dependent GSH synthesis via SLC7A11 leads to the inactivation of GPX4 and ultimately causes ferroptosis in cells SLC7A11 has been reported as a well-validated target for the prevention of ferroptotic cell death. Furthermore, nuclear factor erythroid 2-related factor 2 (Nrf2) regulates the occurrence of ferroptosis at the transcriptional level (57). Nrf2 regulates phagocytosis following oxidative stress in macrophages by inhibiting cellular iron uptake, reducing ROS production and upregulating SLC7A11 expression, as demonstrated by a high Nrf2 expression in astrocytes, preventing neuronal cell death (58). Therefore, Nrf2 also functions as a key regulator of lipid peroxidation and ferroptosis. Kaempferol has been reported to protect cells from ferroptosis via activation of the Nrf2/SLC7A11/GPX4 signaling pathway (59).

Nrf2 is a transcription factor, whose activation promotes iron storage, reduces cellular iron uptake and limits ROS production (55). Nrf2 is one of the key regulators of the oxidative stress pathway (60), which negatively regulates ferroptosis. Under normal physiological conditions, Kelch-like ECH-associated protein 1 binds to Nrf2 via its C-terminal Kelch domain and Nrf2 expression remains low through the ubiquitination of the proteasome (61). When oxidative stress occurs, Nrf2 dissociates from KEAP1 and undergoes nuclear translocation, where it recognizes antioxidant response sites and activates antioxidant genes, including heme oxygenase-1 (HO-1) and SLC7A11 (62). The expression of SLC7A11, as a target of Nrf2, is upregulated when Nrf2 is activated, protecting neuronal cells against ferroptosis, and high Nrf2 expression in astrocytes protects against neuronal cell death (63). HO-1 is a stress-inducing enzyme encoded by the Hmox1 gene. The activation of Nrf2 can initiate the downstream signal of HO-1, thus preventing oxidative stress and scavenging free radicals (64). The knockout of Nrf2 can significantly reduce the protein expression levels of SLC7A11 and HO-1, and can promote the accumulation of lipid peroxides, trigger ferroptosis through iron overload, excess ROS generation and lipid peroxidation, and aggravate the progression of AS. Furthermore, transglutaminase 2 can lead to neuronal death during stroke by inducing GSH depletion, thereby promoting ROS accumulation and ferroptosis (65).

Ferroptosis is involved in the formation and progression of AS

Formation of AS

The pathogenesis of AS involves endothelial cell dysfunction, lipid accumulation, foam cell formation, vascular smooth muscle cell (SMC) migration and inflammatory factor infiltration. The occurrence of AS begins with abnormal lipid metabolism and a large amount of LDL being deposited on the intima of the vascular wall of endothelial cells where it is oxidized to ox-LDL during the process of oxidative stress (10). Free iron accelerates this modification process through the action of hydroxyl free radicals. Monocytes adhere to the vascular endothelium and are transferred to the subendothelium through chemotaxis to transform into macrophages (66). Macrophages serve a pivotal role in AS progression, recognizing and destroying endothelial cells. Macrophages phagocytize ox-LDL through protease and oxygen free radicals secreted by scavenger receptors and are then converted into foam cells, and excessive lipid deposition forms lipid streaks that progress into lipid-containing plaques; iron deposition is visible in the plaque formed by apolipoprotein E-deficient mice and can be seen by staining analysis or imaging (67). SMCs then migrate to the inner membrane and proliferate, forming a fibrous cap of plaque, which is the fibrous plaque phase of AS. Moreover, ox-LDL triggers the immune response, damaged endothelial cells are activated and express monocyte chemoattractant proteins (MCP)-1 and −8, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), E-selectin, P-selectin and other inflammatory factors (68). These factors attract lymphocytes and monocytes to bind to endothelial cells and infiltrate the arterial wall, which causes inflammation. For macrophages, high mobility group protein B1 (HMGB1) is considered necessary for the macrophage inflammatory response, as the ferroptosis inducer erastin promotes the release of damage-associated molecular patterns and HMGB1 (69). The plaque formation process promotes the cooperation of iron and lipid accumulation in macrophages, which lead to the development of AS (70). Moreover, AS plaques with macrophage-derived foam cells as the main components are less stable and more prone to rupture (71).

Immune responses are present throughout the development of AS and inflammation relies on mediators, such as immune cells and inflammatory factors, which are involved in AS development through signaling pathways, such as TLR4, NF-κB and JAK/STAT (72). As well as innate immune cells, such as macrophages, adaptive immune cells participate in the formation of AS by exerting pro-inflammatory or anti-inflammatory effects through the secretion of various cytokines or antibodies (73). Th1 cells promote the development of AS by the secretion of IFN-γ, TNF-α and IL-2. Th2 cells regulate the progression of AS inflammation by the secretion of the anti-inflammatory factor IL-13 and the pro-inflammatory factor IL-4. IL-13 stimulates macrophage polarization to the M2 isoform, releases IL-10 and TGF-β, and functions against AS development through the activation of STAT3; however, IL-4 increases CD36 expression, thereby enhancing macrophage phagocytosis and promoting AS progression (74). Th17 accelerates the progression of AS and Th17 cells produce IL-17, IL-22 and IL-23, recruit neutrophils and promote inflammation at the site of infection (75). Tregs inhibit the activity of multiple immune cells, exerting anti-AS effects by secreting TGF-β1 and IL-10 (76).

Effects of ferroptosis on AS

AS is mainly caused by vascular endothelial dysfunction and plaques that form on blood vessel walls. Atherosclerotic plaque progression is characterized by ox-LDL accumulation within macrophages, macrophage death, necrotic core formation, the rupture or shedding of unstable plaques, and entry into the secondary lesion stage of AS. The majority of acute ischemic events, such as acute coronary syndromes and stroke, are attributed to vulnerable plaques (77). Typically, the core characteristics of vulnerable plaques are active inflammation; in addition, vulnerable plaques are more prone to plaque rupture, and they include the following characteristics (78): i) A thin fibrous cap, large lipid core and micro-calcifications; ii) intraplaque hemorrhage; iii) fiber cap rupture or ulcer formation; and iv) numerous infiltrating macrophages and neo-angiogenesis. The stability of atherosclerotic plaques is closely related to the size of the lipid core, the thickness of the fiber cap and the number of inflammatory cells within the plaque (77). Generally, macrophage death is considered to be a major factor in necrotic core formation and plaque instability, and ferroptosis is mainly involved in oxidative stress and the breakdown of endothelial function (79). Macrophages secrete matrix metalloproteases that degrade collagen fibers in the plaque extracellular matrix, causing plaque rupture, hemorrhage and thrombosis, the release of peroxides and nitrogen radicals, and cause the death of surrounding cells (72). The lipid accumulation process produces a large number of oxygen radicals involved in the inflammatory reaction, promoting the formation of AS, while releasing cytokines and proteases to degrade the collagen matrix of the fiber cap, prompting the plaque to become brittle and rupture, affecting its stability (80). A previous study showed that GPX4 was highly expressed in the early stage of plaque formation. With the progression of plaques, the expression levels of NLRP3 and caspase-1 were increased, and in the late stage, the expression of NLRP3 was significantly upregulated and the expression of GPX4 was decreased. These vulnerable plaques are closely related to a large number of acute ischemic events, including acute coronary syndrome and ischemic stroke (77).

As well as the accumulation of LDL, numerous other factors, such as high uric acid and high homocysteine levels are considered independent risk factors for AS progression. High uric acid levels cause vascular endothelial cell dysfunction by inhibiting the protein expression of endothelial nitric oxide synthase; furthermore, high uric acid levels stimulate monocytes to produce IL-1, IL-6 and TNF-α, which is supported by homocysteine (HCY) via JAK2/STAT3 signaling. Reducing HCY-enhanced STAT3 phosphorylation can significantly reduce HCY-induced microglial activation, and IL-6 and TNF-α production (81). Moreover, endothelial cells generate chemokines and adhesion molecules, promote the migration and adhesion of SMCs, aggravate vascular stenosis and plaque instability, promote vascular calcification, promote the precipitation of uric acid crystallization, increase blood viscosity, induce 5-HT release, increase platelet number and induce internal platelet activation; thus promoting thrombosis. High HCY levels damage endothelial cells, further exacerbating oxidative injury and inflammatory processes, increasing fibrinogen production, abnormal coagulation and platelet dysfunction (82). Platelets release arachidonic acid to produce ROS, resulting in calcium and lipid deposits in the endothelium, which reduces the elasticity of the arterial wall and also causes oxidative stress by affecting cellular respiration, resulting in LDL oxidation. In addition, vascular endothelial cells are damaged, and the normal proportion of endothelin-1 and NO secreted by them to maintain the vasomotor function is broken; the impaired bioavailability of NO, and subsequent vasoconstriction and proliferation of smooth muscle can lead to the progression of AS (83).

Furthermore, gut dysbiosis exacerbates the progression of AS by regulating intestinal structure, intestinal barrier integrity, the inflammatory status and host metabolism (84). It has been reported that the inflammatory response caused by remote infection can aggravate plaque development or cause plaque rupture, and that the metabolism of cholesterol and lipids by intestinal microorganisms can affect the development of atherosclerotic plaques (85). Notably, ferroptosis may also be involved in this process. Chapkin et al observed that n-3 polyunsaturated fatty acids (PUFAs) and the short-chain fatty acid, butyrate, induced apoptosis via a mitochondrially-targeted antioxidant, mitoQ, which is a ubiquinone derivative, and overexpression of GPX4. This mirrored the phenomenon that fatty acid metabolism by the gut microbiota occurs by disrupting the energy metabolism balance of mitochondria, eliciting abnormalities in intracellular Ca2+ homeostasis systems and releasing ROS, which induce cell ferroptosis (86). Similar reports, such as that by Hayase and Jenq (87) reported that the gut flora metabolite, trimethylamine N-oxide (TMAO), enhanced M1 macrophage polarization through inflammasome formation and the activation of NLRP3 in endothelial cells. Furthermore, TMAO has been reported to cause the overexpression of TNF-α, IL-6 and C-reactive proteins, directly inducing vascular inflammation and endothelial dysfunction (88). This process mainly promotes the occurrence and development of AS through the NF-κB pathway. Concurrently, TMAO can increase platelet hyperreactivity and promote arterial thrombosis through Toll receptor signaling pathways (89). Therefore, it could be concluded that AS risk factors break the intestinal ecological balance and that gut microbial metabolites promote AS progression with the involvement of ferroptosis and inflammation.

Following ischemic stroke, the NLRP3 inflammasome has been reported to promote neuroinflammation, and to trigger the apoptosis of glial cells and neurons. NLRP3 activates caspase-1, triggers the release of inflammatory mediators, such as the cytokines IL-18 and IL-1, exacerbates oxidative stress and endoplasmic reticulum stress, and promotes brain edema and atherosclerotic processes (90).

Ferroptosis is involved in AS progression

The progression of AS is often inevitable and is generally considered to occur due to the accumulation of oxidative lipids in the intima, activation of the inflammatory process and the presence of two types of macrophages in its plaque (91). In the early stages, M2 macrophages are the main type of infiltrated cells, the plaque is relatively stable and the anti-inflammatory M2 macrophages produce IL-10 and IL-13, which inactivate Th1 lymphocytes, reduce inflammation and promote tissue damage repair (92). As the disease progresses, long-term iron overload causes the number of M1 macrophages to increase and dominate, further promoting the development of AS; M1 macrophages have a potent phagocytic activity and secrete proinflammatory factors, such as TNF-α (93) (Fig. 3). Blood vessels that are constantly infiltrated by inflammatory factors for a long time are prone to the aggregation and oxidation of LDL, and the deposition of atherosclerotic plaques, promoting AS and the ferroptosis of macrophages in the plaque (94).

In the early stage, AS appears in the form of hemosiderin deposition. In the highly oxidative environment of atherosclerotic lesions, red blood cells rapidly dissolve and release hemoglobin, which is easily oxidized and releases heme. The pro-oxidant and pro-inflammatory effects of heme affect the functions of endothelial cells and macrophages, and promote the oxidation of LDL. Therefore, cholesterol levels are closely related to iron deposition, and activation of the cellular heme oxygenase-1/ferritin system can slow down the progression of this process. Notably, iIncreased iron deposition has been reported to occur in the cerebral cortex of rabbits fed a high-cholesterol diet (95). In MRI T2-weighted images, intra-plaque hemorrhage and iron deposition have been observed (96). Therefore, macrophage ferroptosis may be considered to be involved in the progression of AS (97) and could serve as a potential target for intervention with AS progression. Vinchi et al (98) reported that AS was profoundly aggravated in iron-loaded mice. It has also been reported that iron can exacerbate AS by lipid profile alterations, vascular permeabilization, sustained endothelial activation, elevation of pro-atherogenic inflammatory mediators and reduced nitric oxide availability (99). Treatment with iron chelators in mice has been reported to restore vascular endothelial function, reduce the levels of IL-6, TNF-α and MCP-1, and suppress AS (100). Iron overload leads to endothelial cell dysfunction through pro-oxidative and proinflammatory effects, directly causing AS progression, whereas the occurrence of ferroptosis affects AS progression through increased ROS production and cytokine secretion (101). Consistent with this, it has been confirmed in human coronary artery endothelial cells in vitro that Tanshinone IIA can inhibit ferroptosis-induced endothelial lipid peroxidation and dysfunction by activating the Nrf2 pathway, thus ultimately alleviating AS (102). Furthermore, it has also been reported that ferroptosis is involved in the progression of AS mainly through oxidative stress and the exacerbated breakdown of endothelial function (67).

In a previous study, iron content was directly quantified by nuclear magnetic resonance spectroscopy and it was reported that the level of redox active iron was increased in carotid atherosclerotic lesions compared with in normal healthy human endothelium, and it was further increased in late AS, inducing oxidative stress and inflammatory responses (103). Notably, Bai et al (104) assessed ApoE−/− mice with high-fat diet-induced AS and evaluated the expression of ferroptosis-related factors. This previous study reported significant reductions in the mRNA and protein expression levels of SLC7A11, a decrease in endothelial cell and angiogenic markers, such as CD31, and mitochondrial damage in endothelial cells. Moreover, the opposite results were reported in the same model treated with an iron chelator; the expression levels of the pro-angiogenic factor VEGF, and the adhesion molecules ICAM-1 and VCAM-1 were decreased, and the lesion area in AS was reduced, which indicated that ferroptosis may be involved in the progression of AS (104).

Processes, such as lipid oxidation, inflammatory reactions and iron accumulation, can occur during the pathogenesis of AS. Currently, the applied targets of ferroptosis inhibitors focus on the imbalance of lipid peroxidation and ferrous ions. Iron chelators, antioxidants and free radical scavengers reduce the ferroptosis of endothelial cells by reducing the lipid peroxidation of the plaques in the AS. In vitro, vascular endothelial cells treated with ox-LDL have ferroptotic properties and the use of ferroptotic inhibitors can interfere with this process (Table I).

Table I.

Ferroptosis-related factors and possible targets.

Table I.

Ferroptosis-related factors and possible targets.

First author (year)Ferroptotic factorRegulatory mechanismPossible intervention target to inhibit ferroptosis(Refs.)
Lu et al (2020)TFR1lncRNA PVT1 regulates miR-214-mediated TFR1 expressionSilencing of lncRNA PVT1 and miR-214 overexpression markedly decrease PVT1 levels to suppress ferroptosis in vivo(125)
Li et al (2021)NCOA4Degradation of ferritin leads to free iron releaseKnockout of NCOA4 notably abrogates ferritinophagy and thus inhibits ferroptosis(126)
Chen et al (2021)ACSL4Enhancing lipid peroxidationROSI inhibits ACSL4 and blocks the lipid peroxidation process(127)
Lu et al (2018)ROSNrf2/NADPH/ROS pathwayArtesunate suppresses oxidative toxicity and inflammatory by activating Nrf2 and downregulating ROS(128)
Liu et al (2020)GPX4 xc/GSH/GPX4Sulforaphane alleviates the cytotoxicity of erastin by promoting the expression of genes related to GSH synthesis(129)
Dong et al (2020)Nrf2 Nrf2/SLC7A11/HO-1Nrf2 alleviates OGD/R-induced ferroptosis by upregulating SLC7A11 and HO-1(64)
Wang et al (2020)HMOX1Nrf2/HO-1The upregulation of Nrf2 iron-related target gene HMOX-1 exerts antioxidant and anti-inflammatory effects(2)
Ratan (2020)HIF-1αHIF-1α/HO-1Adaptaquin selectively inhibits HIF prolyl hydroxylases and stabilizes HIF-1 to protect neurons from ferroptosis(57)

[i] TFR1, transferrin receptor 1; NCOA4, nuclear receptor coactivator 4; ACSL4, acyl-CoA synthetase long-chain family member 4; ROS, reactive oxygen species; GSH, glutathione; GPX4, GSH peroxidase 4; Nrf2, nuclear factor erythroid 2-related factor 2; HMOX1, heme oxygenase 1; HIF-1α, hypoxia inducible factor-1α; lncRNA, long non-coding RNA; miR, microRNAs; xc, system xccystine/glutamate antiporter; SLC7A11, solute carrier family 7 member 11; HO-1, heme oxygenase-1; ROSI, rosiglitazone; OGD/R, oxygen-glucose deprivation and reoxygenation.

Ferroptosis accelerates the progression of AS, leading to ischemic stroke

Ferroptosis accelerates AS progression mainly through vascular endothelial disorder and the lipid peroxidation of vascular endothelial cells, endothelial function impairment, platelet adhesion aggregation and eventually, thrombosis (105). The causal association between iron and AS is as follows. Firstly, iron overload leads to the activation of lipoxygenase (106), the upregulation of ACSL4 and ALOX, and catalyzes the lipid peroxidation of phospholipids containing PUFAs in the lipid bilayer of the cell membrane, which are degraded by oxidation, resulting in damage to the cell membrane. Secondly, the interaction between iron and oxygen radicals leads to lipid peroxidation and neuronal death, promotes thrombosis by accelerating the progression of AS and intravascular platelet activation, and eventually, ischemic stroke (98). It has previously been reported that ALOX15 knockout can reduce iron deposition in the cerebral cortex, the level of ROS and the level of 4-hydroxynonenol, which is the final product of lipid peroxidation, and reduces nerve injury through the spermidine/spermine N1-acetyltransferase 1/ALOX15 axis (107).

During the process of an ischemic stroke, iron accumulation can exacerbate neuronal injury in patients or in animal models; iron accumulation is associated with AS and the occurrence of ferroptosis accelerates plaque formation (3). However, this process can be prevented by iron chelation therapy (108). A previous study reported a reduction in iron accumulation and attenuated neuronal degeneration in mice that had been treated with Fer1 (109). Clinical trials have reported that reducing the systemic iron content within 1–3 days of ischemic stroke may provide benefits for patients with acute ischemic stroke (110). Hypoxia-inducible factor (HIF)-1 prolyl hydroxylases (PHDs) may be a target of iron chelators to inhibit ferroptosis. In the brains of hypobaric hypoxic rats pre-treated with deferoxamine, hypoxia inactivated PHDs, causing the accumulation of HIF-1α and the level of HIF-1α protein to be significantly upregulated (111). Cells were more able to tolerate the hypoxic environment and hypoxic cells could recover faster, eventually reducing the volume of cerebral infarction. Moreover, HIF-1 may downregulate ACSL4 expression by binding the ACSL4 promoter to inhibit its transcription and alleviate ischemic brain injury (112).

Neuronal death and secondary inflammation due to cerebral ischemia are directly related to poor functional outcomes, with inflammation occurring throughout the entire phase of ischemia-reperfusion injury and ferroptosis interlinking with inflammation. Following cerebral ischemia, the release of inflammatory cytokines and neurotoxic mediators can be induced through multiple signaling pathways, such as NF-κB and STAT3, leading to neuronal damage and death (113). NF-κB activation following ischemia, and the expression of TNF-α, IL-1 and IL-6, is upregulated in cells to promote the inflammatory response (11). The high expression of IL-6 promotes the continuous phosphorylation of signal transducers and STAT3, and the transcription factor NF-κB enters the nucleus from the cytoplasm, regulating the expression of inflammatory cytokines, causing endothelial cell dysfunction and macrophage polarization, and promoting inflammation (114). IL-6/STAT3 is an important pathway for mediating intracellular inflammatory signaling, which can mediate the production of the proinflammatory cytokines, TNF-α and IL-1, and the anti-inflammatory cytokines, IL-4 and IL-10. Furthermore, the NF-κB/IL-6/STAT3 signaling pathway participates in the regulation of ferroptosis through the inflammatory response after cerebral ischemia and increases the expression of hepcidin (115). Notably, red wine polyphenol extract has been shown to efficiently suppress the inflammation of intestinal epithelial cells by inhibiting JAK/STAT and promoting Nrf2 pathways (116). Therefore, it may be hypothesized that enhancing the activity of Nrf2 and inhibiting the JAK/STAT signaling pathway, may reduce inflammation and monocyte differentiation to macrophages, and regulate SLC7A11 to inhibit ferroptosis. Artesunate is an antimalarial drug, and research has indicated that it also has antitumor and anti-inflammatory effects; it can inactivate the generation of pro-inflammatory mediators in microglia by affecting the NF-κB, p38/MAPK and Nrf2/ARE-dependent signaling pathways, thus inhibiting activation of the immune response following cerebral ischemia (117).

The presence of ferroptosis will continuously damage neural tissue for days to weeks; following ferroptosis, danger-associated molecular patterns (DAMPs) trigger neutrophil recruitment, neutrophil infiltration, proinflammatory cytokine expression (118), leukocyte death, and changes the immune status of the body (119). DAMPs associated with ferroptosis include HMGB1 and IL-33 (120). HMGB1 is released by ferroptotic cells and acts as an adjuvant to activate the recognition receptor of the NF-κB pathway (20); moreover, it triggers an inflammatory response in peripheral macrophages and exacerbates the poor prognosis of ischemic stroke, including cerebral edema and the risk of ischemia-reperfusion (121,122). Furthermore, it has been reported that ferroptosis is the main cause of neuronal death after ischemic stroke. Abnormal tau phosphorylation aggregation leads to neuronal winding, which is involved in the mechanism of ischemic and hemorrhagic stroke. Therefore, inhibiting tau protein expression can inhibit the excitatory cytotoxicity of cells, promote iron outflow to prevent the occurrence of ferroptosis and reduce damage to nerve cells (123). Moreover, the main role of tau protein in neurons is to promote the formation of neuronal microtubule structures and play a key role in axonal transport and cognitive function, but as mice grow older, tau has double effects (124); the tau protein is hyperphosphorylated and amyloid protein is formed, causing nerve fiber damage and nerve fiber degeneration, thus aggravating the neurotoxic iron accumulation.

Conclusion and future perspectives

The mechanisms of AS are complex and involve multiple processes in numerous cell types. Crucially, with the participation of foam cells and macrophages, ferroptosis drives the progression of AS through oxidative stress and inflammatory responses. The present review summarized that ferroptosis is involved in the entire period of atherogenesis and progression through numerous signaling pathways, including lipid pattern, atherosclerotic plaque, fiber plaque and plaque rupture, while interlinking with inflammation to exacerbate the poor prognosis of AS-related diseases. During the development of AS, lipids are deposited under the vascular endothelium forming fatty streak plaques. With the progression of the disease, vascular endothelial cells are damaged, ferroptosis and inflammation participate in the atherosclerotic plaque stage, smooth muscle cells gradually migrate, and the formation of fibrous caps on the surface of the plate indicates the progression of the fibrous plaque stage. The presence of ferroptosis and inflammation further damages endothelial cells, rupture the plaque and forms a series of ischemic events. Undoubtedly, advances in the study of ferroptosis-associated mechanisms will change the traditional concept of AS, and may improve the ability to manage AS risk and address the inevitable risks that remain following current interventions. In conclusion, ferroptosis serves a crucial role in the pathogenesis of AS, ischemic stroke and coronary heart disease, and with the exploration of clinical feasibility, the targeting of ferroptosis may provide novel insights into the treatment of vascular-related diseases.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Dalian High-level Talents Innovation Support Program (grant no. 2018RQ54).

Availability of data and materials

Not applicable.

Authors' contributions

JL, LX contributed to the conception and design of this study. JL prepared the tables and figures, and wrote the manuscript. YXZ, XQC revised the manuscript critically and and added relevant relevant literature. HTC was responsible for revising the manuscript and given final approval of the version to be published. 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 that they have no competing interests.

References

1 

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

2 

Wang H, Liu C, Zhao Y and Gao G: Mitochondria regulation in ferroptosis. Eur J Cell Biol. 99:1510582020. View Article : Google Scholar : PubMed/NCBI

3 

Qiu Y, Cao Y, Cao W, Jia Y and Lu N: The application of ferroptosis in diseases. Pharmacol Res. 159:1049192020. View Article : Google Scholar : PubMed/NCBI

4 

Lin L, Zhang MX, Zhang L, Zhang D, Li C and Li YL: Autophagy, pyroptosis, and ferroptosis: New regulatory mechanisms for atherosclerosis. Front Cell Dev Biol. 9:8099552022. View Article : Google Scholar : PubMed/NCBI

5 

Chen X, Li X, Xu X, Li L, Liang N, Zhang L, Lv J, Wu YC and Yin H: Ferroptosis and cardiovascular disease: Role of free radical-induced lipid peroxidation. Free Radic Res. 55:405–415. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Ross R: Atherosclerosis-an inflammatory disease. N Engl J Med. 340:115–126. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Barquera S, Pedroza-Tobias A, Medina C, Hernández-Barrera L, Bibbins-Domingo K, Lozano R and Moran AE: Global overview of the epidemiology of atherosclerotic cardiovascular disease. Arch Med Res. 46:328–338. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Herrington W, Lacey B, Sherliker P, Armitage J and Lewington S: Epidemiology of atherosclerosis and the potential to reduce the global Burden of atherothrombotic disease. Circ Res. 118:535–546. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Wang Y, Zhao X, Liu L, Soo YO, Pu Y, Pan Y, Wang Y, Zou X, Leung TW, Cai Y, et al: Prevalence and outcomes of symptomatic intracranial large artery stenoses and occlusions in China: The Chinese Intracranial Atherosclerosis (CICAS) study. Stroke. 45:663–669. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Falk E: Pathogenesis of atherosclerosis. J Am Coll Cardiol. 47 (Suppl 8):C7–C12. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Wang Y, Zhao Y, Ye T, Yang L, Shen Y and Li H: Ferroptosis signaling and regulators in atherosclerosis. Front Cell Dev Biol. 9:8094572021. View Article : Google Scholar : PubMed/NCBI

12 

Jiang C, Zhang J, Zhu J, Wang X, Wen Z, Zhao X and Yuan C; CARE-II Investigators, : Association between coexisting intracranial artery and extracranial carotid artery atherosclerotic diseases and ipsilateral cerebral infarction: A Chinese atherosclerosis risk evaluation (CARE-II) study. Stroke Vasc Neurol. 6:595–602. 2021. View Article : Google Scholar : PubMed/NCBI

13 

GBD 2019 Stroke Collaborators, . Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of disease study 2019. Lancet Neurol. 20:795–820. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Saini V, Guada L and Yavagal DR: Global epidemiology of stroke and access to acute ischemic stroke interventions. Neurology. 97 (Suppl 2):S6–S16. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Ornello R, Degan D, Tiseo C, Di Carmine C, Perciballi L, Pistoia F, Carolei A and Sacco S: Distribution and temporal trends from 1993 to 2015 of ischemic stroke subtypes: A systematic review and meta-analysis. Stroke. 49:814–819. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Gong L, Tian X, Zhou J, Dong Q, Tan Y, Lu Y, Wu J, Zhao Y and Liu X: Iron dyshomeostasis induces binding of APP to BACE1 for amyloid pathology, and impairs APP/Fpn1 complex in microglia: Implication in pathogenesis of cerebral microbleeds. Cell Transplant. 28:1009–1017. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Zhang C: Essential functions of iron-requiring proteins in DNA replication, repair and cell cycle control. Protein Cell. 5:750–760. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Pisano G, Lombardi R and Fracanzani AL: Vascular damage in patients with nonalcoholic fatty liver disease: Possible role of iron and ferritin. Int J Mol Sci. 17:6752016. View Article : Google Scholar : PubMed/NCBI

19 

Valenti L, Dongiovanni P, Motta BM, Swinkels DW, Bonara P, Rametta R, Burdick L, Frugoni C, Fracanzani AL and Fargion S: Serum hepcidin and macrophage iron correlate with MCP-1 release and vascular damage in patients with metabolic syndrome alterations. Arterioscler Thromb Vasc Biol. 31:683–690. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Liu J, Kuang F, Kroemer G, Klionsky DJ, Kang R and Tang D: Autophagy-dependent ferroptosis: Machinery and regulation. Cell Chem Biol. 27:420–435. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Ma J, Qian C, Bao Y, Liu MY, Ma HM, Shen MQ, Li W, Wang JJ, Bao YX, Liu Y, et al: Apolipoprotein E deficiency induces a progressive increase in tissue iron contents with age in mice. Redox Biol. 40:1018652021. View Article : Google Scholar : PubMed/NCBI

22 

Yuan H, Pratte J and Giardina C: Ferroptosis and its potential as a therapeutic target. Biochem Pharmacol. 186:1144862021. View Article : Google Scholar : PubMed/NCBI

23 

Fuhrmann DC, Mondorf A, Beifuß J, Jung M and Brune B: Hypoxia inhibits ferritinophagy, increases mitochondrial ferritin, and protects from ferroptosis. Redox Biol. 36:1016702020. View Article : Google Scholar : PubMed/NCBI

24 

Shan X, Lv ZY, Yin MJ, Chen J, Wang J and Wu QN: The protective effect of cyanidin-3-glucoside on myocardial ischemia-reperfusion injury through ferroptosis. Oxid Med Cell Longev. 2021:88801412021. View Article : Google Scholar : PubMed/NCBI

25 

Weinberg ED: The hazards of iron loading. Metallomics. 2:732–740. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Yang F, Bao Q, Wang Z, Ma M, Shen J, Ye F and Xie X: Sex-specific genetically predicted iron status in relation to 12 vascular diseases: A mendelian randomization study in the UK Biobank. Biomed Res Int. 2020:62460412020. View Article : Google Scholar : PubMed/NCBI

27 

Ouyang S, You J, Zhi C, Li P, Lin X, Tan X, Ma W, Li L and Xie W: Ferroptosis: The potential value target in atherosclerosis. Cell Death Dis. 12:7822021. View Article : Google Scholar : PubMed/NCBI

28 

Xiao Z, Kong B, Fang J, Qin T, Dai C, Shuai W and Huang H: Ferrostatin-1 alleviates lipopolysaccharide-induced cardiac dysfunction. Bioengineered. 12:9367–9376. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Naito Y, Masuyama T and Ishihara M: Iron and cardiovascular diseases. J Cardiol. 77:160–165. 2021. View Article : Google Scholar : PubMed/NCBI

30 

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

31 

Ellulu MS, Patimah I, Khaza'ai H, Rahmat A, Abed Y and Ali F: Atherosclerotic cardiovascular disease: A review of initiators and protective factors. Inflammopharmacology. 24:1–10. 2016. View Article : Google Scholar : PubMed/NCBI

32 

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

33 

Gan B: Mitochondrial regulation of ferroptosis. J Cell Biol. 220:e2021050432021. View Article : Google Scholar : PubMed/NCBI

34 

Liu J, Kang R and Tang D: Signaling pathways and defense mechanisms of ferroptosis. FEBS J. Jun 6–2021.(Epub ahead of print). View Article : Google Scholar

35 

Mishra SR, Mahapatra KK, Behera BP, Patra S, Bhol CS, Panigrahi DP, Praharaj PP, Singh A, Patil S, Dhiman R and Bhutia SK: Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 136:1060132021. View Article : Google Scholar : PubMed/NCBI

36 

Jelinek A, Heyder L, Daude M, Plessner M, Krippner S, Grosse R, Diederich WE and Culmsee C: Mitochondrial rescue prevents glutathione peroxidase-dependent ferroptosis. Free Radic Biol Med. 117:45–57. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Zhao Z, Wu J, Xu H, Zhou C, Han B, Zhu H, Hu Z, Ma Z, Ming Z, Yao Y, et al: XJB-5-131 inhibited ferroptosis in tubular epithelial cells after ischemia-reperfusion injury. Cell Death Dis. 11:6292020. View Article : Google Scholar : PubMed/NCBI

38 

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

39 

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

40 

Li N, Jiang W, Wang W, Xiong R, Wu X and Geng Q: Ferroptosis and its emerging roles in cardiovascular diseases. Pharmacol Res. 166:1054662021. View Article : Google Scholar : PubMed/NCBI

41 

Tuo QZ, Liu Y, Xiang Z, Yan HF, Zou T, Shu Y, Ding XL, Zou JJ, Xu S, Tang F, et al: Thrombin induces ACSL4-dependent ferroptosis during cerebral ischemia/reperfusion. Signal Transduct Target Ther. 7:592022. View Article : Google Scholar : PubMed/NCBI

42 

Oh BM, Lee SJ, Park GL, Hwang YS, Lim J, Park ES, Lee KH, Kim BY, Kwon YT, Cho HJ and Lee HG: Erastin inhibits septic shock and inflammatory gene expression via suppression of the NF-kappaB pathway. J Clin Med. 8:22102019. View Article : Google Scholar : PubMed/NCBI

43 

Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, Song S, Tavana O and Gu W: ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 21:579–591. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Xiao FJ, Zhang D, Wu Y, Jia QH, Zhang L, Li YX, Yang YF, Wang H, Wu CT and Wang LS: miRNA-17-92 protects endothelial cells from erastin-induced ferroptosis through targeting the A20-ACSL4 axis. Biochem Biophys Res Commun. 515:448–454. 2019. View Article : Google Scholar : PubMed/NCBI

45 

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

46 

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

47 

Fan Z, Cai L, Wang S, Wang J and Chen B: Baicalin prevents myocardial ischemia/reperfusion injury through inhibiting ACSL4 mediated ferroptosis. Front Pharmacol. 12:6289882021. View Article : Google Scholar : PubMed/NCBI

48 

Noguchi N: Ebselen, a useful tool for understanding cellular redox biology and a promising drug candidate for use in human diseases. Arch Biochem Biophys. 595:109–112. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Chu D and Zhang Z: Trichosanthis pericarpium aqueous extract protects H9c2 cardiomyocytes from Hypoxia/Reoxygenation injury by regulating PI3K/Akt/NO pathway. Molecules. 23:24092018. View Article : Google Scholar : PubMed/NCBI

50 

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

51 

Chen X, Yu C, Kang R, Kroemer G and Tang D: Cellular degradation systems in ferroptosis. Cell Death Differ. 28:1135–1148. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Guan X, Li X, Yang X, Yan J, Shi P, Ba L, Cao Y and Wang P: The neuroprotective effects of carvacrol on ischemia/reperfusioninduced hippocampal neuronal impairment by ferroptosis mitigation. Life Sci. 235:1167952019. View Article : Google Scholar : PubMed/NCBI

53 

Zhang Y, Lu X, Tai B, Li W and Li T: Ferroptosis and its multifaceted roles in cerebral stroke. Front Cell Neurosci. 15:6153722021. View Article : Google Scholar : PubMed/NCBI

54 

Wei X, Yi X, Zhu XH and Jiang DS: Posttranslational modifications in ferroptosis. Oxid Med Cell Longev. 2020:88320432020. View Article : Google Scholar : PubMed/NCBI

55 

Xu T, Ding W, Ji X, Ao X, Liu Y, Yu W and Wang J: Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med. 23:4900–4912. 2019. View Article : Google Scholar : PubMed/NCBI

56 

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. e252019. View Article : Google Scholar : PubMed/NCBI

57 

Ratan RR: The chemical biology of ferroptosis in the central nervous system. Cell Chem Biol. 27:479–498. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Song X and Long D: Nrf2 and Ferroptosis: A new research direction for neurodegenerative diseases. Front Neurosci. 14:2672020. View Article : Google Scholar : PubMed/NCBI

59 

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

60 

Anandhan A, Dodson M, Schmidlin CJ, Liu P and Zhang DD: Breakdown of an ironclad defense system: the critical role of NRF2 in mediating ferroptosis. Cell Chem Biol. 27:436–447. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Fan Z, Wirth AK, Chen D, Wruck CJ, Rauh M, Buchfelder M and Savaskan N: Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 6:e3712017. View Article : Google Scholar : PubMed/NCBI

62 

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

63 

Dodson M, Castro-Portuguez R and Zhang DD: NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 23:1011072019. View Article : Google Scholar : PubMed/NCBI

64 

Dong H, Qiang Z, Chai D, Peng J, Xia Y, Hu R and Jiang H: Nrf2 inhibits ferroptosis and protects against acute lung injury due to intestinal ischemia reperfusion via regulating SLC7A11 and HO-1. Aging (Albany NY). 12:12943–12959. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Colak G and Johnson GV: Complete transglutaminase 2 ablation results in reduced stroke volumes and astrocytes that exhibit increased survival in response to ischemia. Neurobiol Dis. 45:1042–1050. 2012. View Article : Google Scholar : PubMed/NCBI

66 

Yang Y, Wang Y, Guo L, Gao W, Tang TL and Yan M: Interaction between macrophages and ferroptosis. Cell Death Dis. 13:3552022. View Article : Google Scholar : PubMed/NCBI

67 

Marques VB, Leal MAS, Mageski JGA, Fidelis HG, Nogueira BV, Vasquez EC, Meyrelles SDS, Simões MR and Dos Santos L: Chronic iron overload intensifies atherosclerosis in apolipoprotein E deficient mice: Role of oxidative stress and endothelial dysfunction. Life Sci. 233:1167022019. View Article : Google Scholar : PubMed/NCBI

68 

Bosseboeuf E and Raimondi C: Signalling, metabolic pathways and iron homeostasis in endothelial cells in health, atherosclerosis and Alzheimer's disease. Cells. 9:20552020. View Article : Google Scholar : PubMed/NCBI

69 

Wen Q, Liu J, Kang R, Zhou B and Tang D: The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 510:278–283. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Xiao L, Luo G, Guo X, Jiang C, Zeng H, Zhou F, Li Y, Yu J and Yao P: Macrophage iron retention aggravates atherosclerosis: Evidence for the role of autocrine formation of hepcidin in plaque macrophages. Biochim Biophys Acta Mol Cell Biol Lipids. 1865:1585312020. View Article : Google Scholar : PubMed/NCBI

71 

Luo Y, Duan H, Qian Y, Feng L, Wu Z, Wang F, Feng J, Yang D, Qin Z and Yan X: Macrophagic CD146 promotes foam cell formation and retention during atherosclerosis. Cell Res. 27:352–372. 2017. View Article : Google Scholar : PubMed/NCBI

72 

Zhu Y, Xian X, Wang Z, Bi Y, Chen Q, Han X, Tang D and Chen R: Research progress on the relationship between atherosclerosis and inflammation. Biomolecules. 8:802018. View Article : Google Scholar : PubMed/NCBI

73 

Gao Z, Xu X, Li Y, Sun K, Yang M, Zhang Q, Wang S, Lin Y, Lou L, Wu A, et al: Mechanistic Insight into PPARү and Tregs in Atherosclerotic Immune Inflammation. Front Pharmacol. 12:7500782021. View Article : Google Scholar : PubMed/NCBI

74 

Gistera A and Hansson GK: The immunology of atherosclerosis. Nat Rev Nephrol. 13:368–380. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Lee GR: The balance of Th17 versus treg cells in autoimmunity. Int J Mol Sci. 19:7302018. View Article : Google Scholar : PubMed/NCBI

76 

Meng X, Yang J, Dong M, Zhang K, Tu E, Gao Q, Chen W, Zhang C and Zhang Y: Regulatory T cells in cardiovascular diseases. Nat Rev Cardiol. 13:167–179. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Libby P: The changing landscape of atherosclerosis. Nature. 592:524–533. 2021. View Article : Google Scholar : PubMed/NCBI

78 

Bentzon JF, Otsuka F, Virmani R and Falk E: Mechanisms of plaque formation and rupture. Circ Res. 114:1852–1866. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Kobayashi M, Suhara T, Baba Y, Kawasaki NK, Higa JK and Matsui T: Pathological roles of iron in cardiovascular disease. Curr Drug Targets. 19:1068–1076. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Martinet W, Coornaert I, Puylaert P and De Meyer GRY: Macrophage death as a pharmacological target in atherosclerosis. Front Pharmacol. 10:3062019. View Article : Google Scholar : PubMed/NCBI

81 

Chen S, Dong Z, Cheng M, Zhao Y, Wang M, Sai N, Wang X, Liu H, Huang G and Zhang X: Homocysteine exaggerates microglia activation and neuroinflammation through microglia localized STAT3 overactivation following ischemic stroke. J Neuroinflammation. 14:1872017. View Article : Google Scholar : PubMed/NCBI

82 

Zhang T, Jiang Y, Zhang S, Tie T, Cheng Y, Su X, Man Z, Hou J, Sun L, Tian M, et al: The association between homocysteine and ischemic stroke subtypes in Chinese: A meta-analysis. Medicine (Baltimore). 99:e194672020. View Article : Google Scholar : PubMed/NCBI

83 

Kumar A, Palfrey HA, Pathak R, Kadowitz PJ, Gettys TW and Murthy SN: The metabolism and significance of homocysteine in nutrition and health. Nutr Metab (Lond). 14:782017. View Article : Google Scholar : PubMed/NCBI

84 

Zhou W, Cheng Y, Zhu P, Nasser MI, Zhang X and Zhao M: Implication of gut microbiota in cardiovascular diseases. Oxid Med Cell Longev. 2020:53940962020. View Article : Google Scholar : PubMed/NCBI

85 

Jonsson AL and Backhed F: Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 14:79–87. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Chapkin RS, Navarro SL, Hullar MAJ and Lampe JW: Diet and gut microbes act coordinately to enhance programmed cell death and reduce colorectal cancer risk. Dig Dis Sci. 65:840–851. 2020. View Article : Google Scholar : PubMed/NCBI

87 

Hayase E and Jenq RR: Too much TMAO and GVHD. Blood. 136:383–385. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Janeiro MH, Ramirez MJ, Milagro FI, Martinez JA and Solas M: Implication of trimethylamine N-Oxide (TMAO) in disease: Potential biomarker or new therapeutic target. Nutrients. 10:13982018. View Article : Google Scholar : PubMed/NCBI

89 

Lassiger-Herfurth A, Pontarollo G, Grill A and Reinhardt C: The gut microbiota in cardiovascular disease and arterial thrombosis. Microorganisms. 7:6912019. View Article : Google Scholar : PubMed/NCBI

90 

Tuttolomondo A, Puleo MG, Velardo MC, Corpora F, Daidone M and Pinto A: Molecular biology of atherosclerotic ischemic strokes. Int J Mol Sci. 21:93722020. View Article : Google Scholar : PubMed/NCBI

91 

Cornelissen A, Guo L, Sakamoto A, Virmani R and Finn AV: New insights into the role of iron in inflammation and atherosclerosis. EBioMedicine. 47:598–606. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Tabas I and Bornfeldt KE: Macrophage phenotype and function in different stages of atherosclerosis. Circ Res. 118:653–667. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Chen X, Kang R, Kroemer G and Tang D: Ferroptosis in infection, inflammation, and immunity. J Exp Med. 218:e202105182021. View Article : Google Scholar : PubMed/NCBI

94 

Wolf D and Ley K: Immunity and inflammation in atherosclerosis. Circ Res. 124:315–327. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Jeney V, Balla G and Balla J: Red blood cell, hemoglobin and heme in the progression of atherosclerosis. Front Physiol. 5:3792014. View Article : Google Scholar : PubMed/NCBI

96 

Raman SV, Winner MW III, Tran T, Velayutham M, Simonetti OP, Baker PB, Olesik J, McCarthy B, Ferketich AK and Zweier JL: In vivo atherosclerotic plaque characterization using magnetic susceptibility distinguishes symptom-producing plaques. JACC Cardiovasc Imaging. 1:49–57. 2008. View Article : Google Scholar : PubMed/NCBI

97 

Hu H, Chen Y, Jing L, Zhai C and Shen L: The link between ferroptosis and cardiovascular diseases: A novel target for treatment. Front Cardiovasc Med. 8:7109632021. View Article : Google Scholar : PubMed/NCBI

98 

Vinchi F, Porto G, Simmelbauer A, Altamura S, Passos ST, Garbowski M, Silva AMN, Spaich S, Seide SE, Sparla R, et al: Atherosclerosis is aggravated by iron overload and ameliorated by dietary and pharmacological iron restriction. Eur Heart J. 41:2681–2695. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Neven E, De Schutter TM, Behets GJ, Gupta A and D'Haese PC: Iron and vascular calcification. Is there a link? Nephrol Dial Transplant. 26:1137–1145. 2011. View Article : Google Scholar : PubMed/NCBI

100 

Kempf T and Wollert KC: Iron and atherosclerosis: Too much of a good thing can be bad. Eur Heart J. 41:2696–2698. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Le Y, Zhang Z, Wang C and Lu D: Ferroptotic cell death: New regulatory mechanisms for metabolic diseases. Endocr Metab Immune Disord Drug Targets. 21:785–800. 2021. View Article : Google Scholar : PubMed/NCBI

102 

He L, Liu YY, Wang K, Li C, Zhang W, Li ZZ, Huang XZ and Xiong Y: Tanshinone IIA protects human coronary artery endothelial cells from ferroptosis by activating the NRF2 pathway. Biochem Biophys Res Commun. 575:1–7. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Stadler N, Lindner RA and Davies MJ: Direct detection and quantification of transition metal ions in human atherosclerotic plaques: Evidence for the presence of elevated levels of iron and copper. Arterioscler Thromb Vasc Biol. 24:949–954. 2004. View Article : Google Scholar : PubMed/NCBI

104 

Bai T, Li M, Liu Y, Qiao Z and Wang Z: Inhibition of ferroptosis alleviates atherosclerosis through attenuating lipid peroxidation and endothelial dysfunction in mouse aortic endothelial cell. Free Radic Biol Med. 160:92–102. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Chen Z, Yan Y, Qi C, Liu J, Li L and Wang J: The role of ferroptosis in cardiovascular disease and its therapeutic significance. Front Cardiovasc Med. 8:7332292021. View Article : Google Scholar : PubMed/NCBI

106 

Huang F, Yang R, Xiao Z, Xie Y, Lin X, Zhu P, Zhou P, Lu J and Zheng S: Targeting ferroptosis to treat cardiovascular diseases: A new continent to be explored. Front Cell Dev Biol. 9:7379712021. View Article : Google Scholar : PubMed/NCBI

107 

Zhao J, Wu Y, Liang S and Piao X: Activation of SSAT1/ALOX15 axis aggravates cerebral ischemia/reperfusion injury via triggering neuronal ferroptosis. Neuroscience. 485:78–90. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Selim M: Treatment with the iron chelator, deferoxamine mesylate, alters serum markers of oxidative stress in stroke patients. Transl Stroke Res. 1:35–39. 2010. View Article : Google Scholar : PubMed/NCBI

109 

Xie BS, Wang YQ, Lin Y, Mao Q, Feng JF, Gao GY and Jiang JY: 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

110 

Millan M, DeGregorio-Rocasolano N, Perez de la Ossa N, Reverté S, Costa J, Giner P, Silva Y, Sobrino T, Rodríguez-Yáñez M, Nombela F, et al: Targeting pro-oxidant iron with deferoxamine as a treatment for ischemic stroke: Safety and optimal dose selection in a randomized clinical trial. Antioxidants (Basel). 10:12702021. View Article : Google Scholar : PubMed/NCBI

111 

Li W, Xiang Z, Xing Y, Li S and Shi S: Mitochondria bridge HIF signaling and ferroptosis blockage in acute kidney injury. Cell Death Dis. 13:3082022. View Article : Google Scholar : PubMed/NCBI

112 

Bai YT, Xiao FJ, Wang H, Ge RL and Wang LS: Hypoxia protects H9c2 cells against Ferroptosis through SENP1-mediated protein DeSUMOylation. Int J Med Sci. 18:1618–1627. 2021. View Article : Google Scholar : PubMed/NCBI

113 

Liu H, Wu X, Luo J, Wang X, Guo H, Feng D, Zhao L, Bai H, Song M, Liu X, et al: Pterostilbene attenuates astrocytic inflammation and neuronal oxidative injury after ischemia-reperfusion by inhibiting NF-kB phosphorylation. Front Immunol. 10:24082019. View Article : Google Scholar : PubMed/NCBI

114 

Yang L, Wang H, Yang X, Wu Q, An P, Jin X, Liu W, Huang X, Li Y, Yan S, et al: Auranofin mitigates systemic iron overload and induces ferroptosis via distinct mechanisms. Signal Transduct Target Ther. 5:1382020. View Article : Google Scholar : PubMed/NCBI

115 

Erta M, Quintana A and Hidalgo J: Interleukin-6, a major cytokine in the central nervous system. Int J Biol Sci. 8:1254–1266. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Nunes C, Teixeira N, Serra D, Freitas V, Almeida L and Laranjinha J: Red wine polyphenol extract efficiently protects intestinal epithelial cells from inflammation via opposite modulation of JAK/STAT and Nrf2 pathways. Toxicol Res (Camb). 5:53–65. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Zuo S, Li Q, Liu X, Feng H and Chen Y: The potential therapeutic effects of artesunate on stroke and other central nervous system diseases. Biomed Res Int. 2016:14890502016. View Article : Google Scholar : PubMed/NCBI

118 

Khoshnam SE, Winlow W, Farzaneh M, Farbood Y and Moghaddam HF: Pathogenic mechanisms following ischemic stroke. Neurol Sci. 38:1167–1186. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Iadecola C, Buckwalter MS and Anrather J: Immune responses to stroke: Mechanisms, modulation, and therapeutic potential. J Clin Invest. 130:2777–2788. 2020. View Article : Google Scholar : PubMed/NCBI

120 

Zhang Y, Xin L, Xiang M, Shang C, Wang Y, Wang Y, Cui X and Lu Y: The molecular mechanisms of ferroptosis and its role in cardiovascular disease. Biomed Pharmacother. 145:1124232022. View Article : Google Scholar : PubMed/NCBI

121 

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

122 

Yan HF, Tuo QZ, Yin QZ and Lei P: The pathological role of ferroptosis in ischemia/reperfusion-related injury. Zool Res. 41:220–230. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Derry PJ, Hegde ML, Jackson GR, Kayed R, Tour JM, Tsai AL and Kent TA: Revisiting the intersection of amyloid, pathologically modified tau and iron in Alzheimer's disease from a ferroptosis perspective. Prog Neurobiol. 184:1017162020. View Article : Google Scholar : PubMed/NCBI

124 

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

125 

Lu J, Xu F and Lu H: LncRNA PVT1 regulates ferroptosis through miR-214-mediated TFR1 and p53. Life Sci. 260:1183052020. View Article : Google Scholar : PubMed/NCBI

126 

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

127 

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

128 

Lu H, Wang B, Cui N and Zhang Y: Artesunate suppresses oxidative and inflammatory processes by activating Nrf2 and ROSdependent p38 MAPK and protects against cerebral ischemia-reperfusion injury. Mol Med Rep. 17:6639–6646. 2018.PubMed/NCBI

129 

Liu Z, Lv X, Song E and Song Y: Fostered Nrf2 expression antagonizes iron overload and glutathione depletion to promote resistance of neuron-like cells to ferroptosis. Toxicol Appl Pharmacol. 407:1152412020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 26 Issue 5

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
Li J, Xu L, Zuo YX, Chang XQ and Chi HT: Potential intervention target of atherosclerosis: Ferroptosis (Review). Mol Med Rep 26: 343, 2022
APA
Li, J., Xu, L., Zuo, Y.X., Chang, X.Q., & Chi, H.T. (2022). Potential intervention target of atherosclerosis: Ferroptosis (Review). Molecular Medicine Reports, 26, 343. https://doi.org/10.3892/mmr.2022.12859
MLA
Li, J., Xu, L., Zuo, Y. X., Chang, X. Q., Chi, H. T."Potential intervention target of atherosclerosis: Ferroptosis (Review)". Molecular Medicine Reports 26.5 (2022): 343.
Chicago
Li, J., Xu, L., Zuo, Y. X., Chang, X. Q., Chi, H. T."Potential intervention target of atherosclerosis: Ferroptosis (Review)". Molecular Medicine Reports 26, no. 5 (2022): 343. https://doi.org/10.3892/mmr.2022.12859