Introduction
Acute respiratory distress syndrome (ARDS) is a severe clinical condition characterized by the rapid onset of respiratory failure due to widespread inflammation and damage to the alveolar‒capillary barrier.1 This syndrome manifests as severe hypoxemia, diminished lung compliance, and laboured breathing, which collectively result in substantial morbidity and mortality in affected individuals2. ARDS develops secondary to heterogeneous etiologies, including pneumonia, sepsis, trauma, and aspiration, and remains a critical clinical challenge in critical care medicine. According to global studies, ARDS accounts for approximately 10% of hospitalized patients and has a mortality rate ranging from 30% to 50%.3
Ferroptosis, an emerging non-apoptotic, iron-dependent cell death pathway distinct from necrosis, has emerged as a critical pathogenic mechanism in ARDS.4 This form of cell death is characterized by the accumulation of lethal levels of lipid peroxides (LPOs), driven by dysregulated iron homeostasis and redox imbalance.5 In ARDS, ferroptosis is strongly associated with the injury of alveolar epithelial cells (AECs),6 structural cells central to pulmonary homeostasis and gas exchange.7 The unique properties of ferroptosis make it a potential contributor to the pathophysiology of ARDS, particularly given the inflammatory microenvironment associated with this condition.8 As the understanding of the intricate relationships among cell death, inflammation, and oxidative stress deepens,9 new evidences suggest that ferroptosis may play dual roles—both as a facilitator of lung injury and as a target for therapeutic intervention. By examining the current research on ferroptosis in AECs within the context of ARDS, this review aims to elucidate its potential implications for diagnosis and treatment.
Pathophysiology of ARDS
ARDS is a complex and multifactorial condition that arising from diverse direct or indirect pulmonary insults, culminating in profound derangements of alveolar-capillary barrier function.10 Direct lung injuries refer to primary damage to the pulmonary parenchyma itself, exemplified by pneumonia,11 foreign body inhalation,12 inhalation of toxic smoke,13 and lung contusion caused by blunt chest trauma14. In contrast, indirect lung injuries originate from systemic processes that affect the lungs without initially damaging them directly. Examples of such indirect injuries include major trauma and sepsis, which can occur as a consequence of various infections.15 These systemic conditions can lead to inflammatory responses and changes in pulmonary vascular permeability, ultimately contributing to the development of ARDS.
ARDS is pathologically defined by an exaggerated inflammatory response within the lungs, which occurs in response to initial injury. Following an insult, such as bacterial infection, pneumonia, or trauma, a cascade of inflammatory events is triggered.16 Immune cells, such as alveolar macrophages, release proinflammatory cytokines, including tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β), which recruit neutrophils and other immune effector cells to the site of injury. As neutrophils infiltrate the lung tissue, they subsequently degranulate and release enzymes, reactive oxygen species (ROS), and additional proinflammatory mediators, thereby escalating the inflammatory response.17 This recruitment and activation results in damage to both epithelial and endothelial cells, compromising the integrity of the alveolar‒capillary membrane. Disruption of this barrier leads to increased permeability, allowing protein-rich fluid to escape into the alveoli. This fluid causes pulmonary oedema, which is a defining characteristic of ARDS.18 Moreover, this inflammatory milieu activates the coagulation cascade, contributing to microvascular thrombosis and further impairing gas exchange due to venous stasis and clot formation. The balance between proinflammatory and anti-inflammatory signals becomes dysregulated, resulting in a cycle of ongoing inflammation and lung injury.19
Mechanisms of Ferroptosis
Ferroptosis is a form of regulated cell death characterized by the accumulation of toxic levels of LPOs, which is primarily driven by iron metabolism and an imbalance between oxidative stress and antioxidant defences20 (Figure 1).
Figure 1 Mechanisms of Ferroptosis.
|
Key Characteristics of Ferroptosis
Ferroptosis is an iron-dependent regulated cell death pathway, which plays a crucial role in generating ROS.21 Elevated intracellular iron levels promote the Fenton reaction, where ferrous iron reacts with hydrogen peroxide to produce hydroxyl radicals. These radicals initiate lipid peroxidation, contributing to cellular damage.22 Additionally, central to ferroptosis is the accumulation of LPOs, generated through degradation of polyunsaturated fatty acids in cellular membranes. Unlike other forms of cell death, lipid peroxidation in ferroptosis reaches lethal levels, leading to membrane rupture and ultimately cell death. This process is influenced primarily by specific lipoxygenase enzymes that oxidize polyunsaturated fatty acids.23
Molecular Pathways Involved in Ferroptosis
Role of Glutathione and GPX4
Glutathione (GSH), a tripeptide composed of glutamic acid, cysteine, and glycine, serves as a central redox buffer in mammalian cells. It plays an essential role in protecting cells from oxidative stress by neutralizing ROS and detoxifying free radicals. High levels of GSH are necessary to maintain the cellular redox status and prevent excessive lipid peroxidation.24 In ferroptotic processes, GSH levels are critically altered. The depletion of intracellular glutathione (GSH) critically compromises cellular antioxidant defenses, significantly increasing susceptibility to ferroptotic cell death. When cellular GSH levels decrease, either because of increased ROS or inadequate synthesis, the capacity to detoxify LPOs is insufficient. The resulting failure in redox homeostasis leads to pathological accumulation of toxic lipid aldehydes, ultimately initiating the execution phase of ferroptotic cell death through iron-dependent phospholipid peroxidation.25
Glutathione peroxidase 4 (GPX4) is a key enzyme that uses GSH to reduce lipid hydroperoxides to their corresponding alcohols, effectively suppressing lipid peroxidation.26 When GPX4 activity is inhibited or its expression is downregulated, lethal LPOs accumulation results. Various regulatory factors influence GPX4 expression, including transcriptional regulators27 and posttranslational modifications, such as phosphorylation and ubiquitination.28 Furthermore, several studies have shown that lipids themselves regulate GPX4 activity through pathways such as cell membrane composition, LPOs production, and the regulation of bioactive lipids, linking lipid metabolism to the regulation of ferroptosis.29 Therefore, GPX4 not only mitigates lipid peroxidation but also serves as a critical checkpoint in the ferroptotic signalling pathway.30
System Xc− and the Cystine/Glutamate Antiporter
System Xc− is a vital cystine/glutamate antiporter that plays a crucial role in maintaining cellular redox homeostasis and influencing ferroptosis by regulating the levels of GSH.31 This cystine-glutamate antiporter, composed of two distinct subunits, mediates cystine uptake through solute carrier family 7 member 11 (SLC7A11) while utilizing solute carrier 3A2 (SLC3A2) to stabilize membrane expression and coordinate glutamate efflux. The transporter’s heteromeric structure enables simultaneous transmembrane movement of extracellular cystine into cellular compartments and cytosolic glutamate extrusion into the extracellular space.32 The entry of cystine is critical because it is converted into cysteine, a key precursor for GSH synthesis, which helps neutralize ROS and prevents oxidative damage. During oxidative stress or inflammatory episodes, cellular cysteine demand escalates;33 however, system Xc− dysfunction impairs cystine transport capacity, leading to diminished GSH biosynthesis. This depletion impedes the cell’s antioxidant capacity, increasing susceptibility to lipid peroxidation that ultimately triggers ferroptosis execution.34 Furthermore, the activity of system Xc− is influenced by various regulatory factors. Proinflammatory cytokines including TNF-α35 and IL-1β36 promote System Xc− expression, whereas anti-inflammatory cytokines such as IL-10 suppress the activity of System Xc−. Under oxidative stress, cytokines can regulate System Xc− by activating transcription factors such as nuclear factor E2 related factor 2 (Nrf2),37,38 highlighting their critical role not only in maintaining redox balance but also in modulating cell fate in response to stressors.
Interaction with Oxidative Stress
The interplay between ferroptosis and oxidative stress constitutes a pathological nexus in diverse disease manifestations.39 Ferroptosis, characterized by the iron-dependent accumulation of LPOs to lethal levels, strongly influences the state of oxidative stress within the cell.21,23,40 During iron overload or regulatory dysfunction, free iron can catalyse the Fenton reaction, producing highly reactive hydroxyl radicals (•OH). These radicals react with cellular components, leading to oxidative damage and further increasing the levels of ROS.41 Iron can also affect mitochondrial function, resulting in instability of the electron transport chain and excessive generation of ROS. As ferroptosis occurs, the loss of the mitochondrial membrane potential and depletion of adenosine triphosphate (ATP) contribute to mitochondrial dysfunction, which may further increase the production of ROS.42 Additionally, the intracellular components following ferroptosis, such as damage-associated molecular patterns (DAMPs), can trigger inflammatory responses and cause more immune cells to release oxidants, thereby worsening oxidative stress.43 Furthermore, during ferroptosis, the activity of antioxidant enzymes, such as GPX4, is often suppressed or reduced, leading to redox imbalance and diminished cellular capacity to clear ROS.44 This augmented ROS production leads to increased oxidative stress, which can deplete cellular antioxidants, particularly GSH, which are essential for protecting cells from oxidative damage.24,45 As GSH levels decrease, the detoxification of LPOs becomes impaired, increasing the susceptibility of cells to ferroptosis.46 Furthermore, excessive ROS can lead to peroxidation of polyunsaturated fatty acids (PUFAs) in cell membranes47 and cause direct damage to proteins and DNA, compounding cellular dysfunction and promoting further inflammation.48 The resulting cycle of oxidative damage not only facilitates the onset of ferroptosis but also exacerbates tissue injury.
Mechanisms of Ferroptosis in ARDS
In ARDS, ferroptosis is primarily driven by oxidative stress, which is exacerbated by inflammation and the dysregulation of antioxidant defense mechanisms, including GSH depletion and GPX4 inhibition.
Role of Molecular Pathways of Ferroptosis in ARDS
GPX4 Pathway Dysregulation in ARDS
In pulmonary physiology, GPX4 serves as a critical guardian against oxidative stress and lipid peroxidation in AECs, particularly within the inflammatory-oxidative stress microenvironment characteristic of ARDS. Under normal conditions, GPX4 helps maintain membrane integrity by preventing excessive lipid peroxidation, which is essential for preserving lung function. However, in ARDS, inflammatory cytokines and oxidative stress both abundant during the disease can downregulate or inhibit GPX4 expression and activity. This reduction impairs GPX4’s ability to counteract lipid peroxidation, leading to the unregulated accumulation of lipid hydroperoxides. As a result, this accumulation triggers ferroptosis in AECs and resident lung cells.49 Additionally, the inhibition or dysfunction of GPX4 contributes to the disruption of the alveolar-capillary barrier, exacerbating lung injury, impairing gas exchange, and potentially promoting fibrosis during the later stages of ARDS. Mechanistic studies reveal that pharmacological activation of the Keap1/Nrf2/GPX4 axis restores cellular antioxidant defenses and attenuates ferroptosis in AECs.50
System Xc- Dysfunction in ARDS
In ARDS, oxidative stress often impairs the function of system Xc-, leading to a reduction in the intracellular cystine pool and limiting the production of GSH. This redox imbalance reduces GPX4 activity, accelerating lipid hydroperoxides accumulation and triggering ferroptosis. Inflammatory signals and oxidative stress in ARDS result in the downregulation of system Xc- activity, thereby compromising GSH biosynthesis. This dysfunction further enhances lipid peroxidation and contributes to ferroptosis in lung cells, especially AECs, which are particularly vulnerable to oxidative damage. Additionally, dysfunctional system Xc- exacerbates the imbalance between pro- and anti-oxidant systems, creating a vicious cycle of inflammation, oxidative stress, and cell death. Ma et al collected bronchoalveolar lavage fluid of ARDS patients and found that SLC7A11 was significantly increased in patients with moderate ARDS compared with patients with mild ARDS. The level of neutrophils in peripheral blood of ARDS patients is positively correlated with the expression level of SLC7A11.51
Alternative Pathways of Ferroptosis in ARDS
One of the hallmark features of ferroptosis is iron homeostasis disruption. Elevated iron levels in the lungs, either through increased iron uptake or decreased iron sequestration, can promote lipid peroxidation and accelerate ferroptosis. In ARDS, inflammatory cytokines and hypoxia may alter iron metabolism, increasing iron deposition in lung tissues and driving ferroptosis. While GPX4 and system Xc- constitute principal regulatory nodes in ferroptosis execution, emerging evidence reveals that alternative biochemical pathways contribute to lipid peroxidation when canonical mechanisms become overwhelmed during pathological conditions like ARDS.
Acyl-CoA Synthetase Long-Chain Family Member 4 (ACSL4) is another key enzyme serving as a critical mediator in lipid peroxidation by catalyzing the esterification of long-chain fatty acids, particularly arachidonic acid, which is a precursor for the production of lipid hydroperoxides.52 In acute lung injury (ALI), the primary stage of ARDS, dysregulated upregulation of ACSL4 drives pathological accumulation of lipid peroxides that promotes ferroptosis.53 Targeting ACSL4 activity could help mitigate ferroptosis-induced lung damage, knockout of SHP2 can reduce ferroptosis by down-regulating the expression of ACSL4 in ALI,54 METTL3 promotes ferroptosis by altering the m6A modification of ACSL4.55
Beyond its canonical tumor-suppressive functions, p53 emerges as a central regulator of ferroptosis. Loss of SIRT1-mediated acetylation modification of p53 inhibits ferroptosis.56 In the context of ARDS, p53 can suppress the expression of SLC7A11, a subunit of the system Xc- antiporter, thus reducing cystine uptake and GSH synthesis, which promotes ferroptosis.57
Heme Oxygenase-1 (HO-1) is an enzyme involved in heme degradation, has been shown to play a dual role in ferroptosis. Under certain conditions, HO-1 activity may alleviate oxidative stress by degrading heme and producing biliverdin, an antioxidant. However, excessive HO-1 activity can lead to iron overload, which accelerates ferroptosis.58 In ARDS, the balance between protective and pathological effects of HO-1 activity may influence ferroptosis progression. Nrf2 inhibits ferroptosis and protects against ALI via regulating SLC7A11 and HO-1.59 IL-27 regulates macrophage ferroptosis by inhibiting the Nrf2/HO1 signaling pathway in sepsis-induced ARDS.60
Ferroptosis and Oxidative Stress in Early ARDS
Oxidative stress increases the generation of ROS by disrupting normal metabolic processes and cellular structures within AECs.61 The excessive accumulation of ROS can directly cause cellular damage by harming cell membranes, proteins, and DNA.62 Additionally, oxidative stress can activate signaling pathways such as the NF-κB and MAPK pathways, leading to the upregulation of enzymes that produce ROS, including NOX and cyclooxygenase-2 (COX-2), thereby promoting the release of inflammatory cytokines and chemokines.63 Elevated levels of ROS can oxidize ferritin proteins (such as ferritin and iron‒sulfur clusters), releasing more free iron, which increases substrates for the Fenton reaction and leads to iron overload, further promoting lipid peroxidation.64 Moreover, oxidative stress attacks PUFAs on cell membranes, triggering peroxidation reactions that produce LPOs and free radicals; these products can disrupt cell membranes and impair iron metabolism, thus promoting ferroptosis.65 Oxidative stress also inhibits the intracellular antioxidant system, diminishing the ability of the cell to defend against lipid peroxidation, as evidenced by GPX4 inactivation and GSH depletion, along with suppressed superoxide dismutase (SOD) and catalase activities.21,66 Furthermore, oxidative stress interferes with the function of lipid-metabolizing enzymes and alters the composition of lipids in cell membranes, increasing susceptibility to ferroptosis, such as through the upregulation of ACSL4 expression and activity via the Nrf2-ARE pathway.67 Similarly, LPOs formed from the oxidation of PUFAs during ferroptosis are a significant source of ROS, directly driving oxidative stress.68 Iron overload significantly accelerates the Fenton reaction, thereby increasing ROS generation and leading to a rapid increase in the level of intracellular oxidative stress. During ferroptosis, system Xc- consisting of SLC7A11 and SLC3A2, is inhibited, which decreases extracellular cysteine uptake and ultimately causes GSH depletion, exacerbating ROS accumulation and oxidative stress. Additionally, 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), which are generated during lipid peroxidation, covalently bind to proteins, DNA, and membrane lipids, which compromises cell membrane integrity, disrupts cellular signaling pathways, and further increases ROS production.69 This process ultimately creates a vicious cycle of “inflammation‒oxidative stress‒lipid peroxidation”, leading to ARDS (Figure 2).
![]() |
Figure 2 Ferroptosis, inflammation and oxidative stress in AECs. In early ARDS, oxidative stress, driven by reactive oxygen species (ROS), disrupts alveolar epithelial cell (AEC) integrity. Excessive ROS oxidizes polyunsaturated fatty acids (PUFAs), triggering lipid peroxidation (LPO) via ACSL4/LOX pathways, while depleting glutathione (GSH)/GPX4 antioxidant defenses. This results in iron overload, Fenton reaction activation, and ferroptosis, characterized by membrane lipid remodeling (4-HNE/MDA accumulation) and DAMPs release, which amplifies inflammation. In late ARDS, persistent ferroptosis reduces functional cell populations (AECs, endothelial cells, fibroblasts), impairing tissue repair. Ferroptosis sustains oxidative stress through TGF-β/SMAD signaling, promoting collagen deposition and myofibroblast differentiation.
|
Ferroptosis and Tissue Repair and Fibrosis in Late ARDS
In the later stages of ARDS, although inflammation gradually subsides, tissue repair and fibrosis become the primary pathological processes.70 However, ferroptosis persists as a critical pathogenic mechanism during this phase, exerting direct inhibitory effects on tissue repair processes that accelerate pulmonary fibrosis progression.71 Tissue repair in the later stages of ARDS mainly relies on normal cell regeneration. The ongoing occurrence of ferroptosis further damages cellular and tissue structures, reducing the number of functional cells available for repair. Key repair cells, such as AECs, endothelial cells, and fibroblasts, lose their function due to ferroptosis, which delays recovery by obstructing the repair process.72 Moreover, ferroptosis exacerbates the fibrosis process by inducing sustained oxidative stress. Proliferation of lipid peroxidation products (LPOs) and membrane integrity disruption synergistically drive fibroblast-myofibroblast hyperactivation, resulting in aberrant collagen deposition and extracellular matrix remodeling that exacerbates pulmonary fibrosis.73 Chronic ferroptosis and oxidative stress may ultimately result in lung tissue hardening, contributing to irreversible lung fibrosis, which is one of the leading causes of death in ARDS patients.74 Furthermore, persistent oxidative stress and ferroptosis create a feedback loop through the accumulation of ROS, further activating the expression of fibrosis-related factors such as transforming growth factor-beta (TGF-β).75 These factors not only promote fibroblast proliferation and collagen deposition but also inhibit normal cell function, accelerating the progression of fibrosis.76 Ferroptosis inducer erastin enhances TGF-β1 induced fibroblast-to-myofibroblast differentiation pulmonary fibrosis models in vitro.77
Pathological Mechanism of Ferroptosis in AECs
In the lungs, AECs, which include type I and type II cells, are crucial for maintaining the integrity of the alveolar barrier and facilitating gas exchange.78 Type I epithelial cells form the thin, gas-exchanging surface of the alveoli, whereas type II cells are responsible for surfactant production and alveolar repair.79 Both types of epithelial cells undergo significant damage during ARDS due to susceptibility to ferroptosis, oxidative stress, inflammation, lung injury repair dysfunction, and alveolar‒capillary barrier breakdown.
AECs are Susceptible to Ferroptosis
AECs are particularly susceptible to ferroptosis for several reasons. Firstly, AECs are constantly exposed to elevated levels of oxidative stress due to the continuous influx of oxygen into the lungs.80 Secondly, these cells contain a substantial amount of PUFAs within their membrane, which renders them more vulnerable to lipid peroxidation.81 Finally, the epithelial lining fluid contains a high concentration of iron, which can exacerbate oxidative stress and further promote ferroptosis.82 Together, these factors contribute to the increased susceptibility of AECs to iron-induced cell death, making it a significant pathway in lung injury.
Ferroptosis, Inflammation and Oxidative Stress in AECs
Ferroptosis, inflammation and oxidative stress are closely interconnected processes in AECs. When AECs undergo ferroptosis, cell membranes rupture and mitochondria damage result in DAMPs release, which then activates inflammasomes in nearby immune cells.83 This inflammasome activation triggers inflammatory cytokines secretion, such as TNF-α, IL-1β, and IL-6. These cytokines promote local inflammation by recruiting neutrophils to the injury site and stimulating the release of additional inflammatory mediators and ROS.84 Notably, IL-6 not only contributes to inflammation, but also induces lipid peroxidation and disrupts iron homeostasis, leading to an increase in iron accumulation within epithelial cells and further driving ferroptosis.85 Moreover, these cytokines activate multiple inflammatory signalling pathways, including the JAK-STAT, NF-κB, cGAS-STING, and MAPK pathways, impair the antioxidant defence systems of AECs and lead to the accumulation of ROS and LPOs.86 Additionally, both cytokines and mechanical stretching activate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), an enzyme embedded in the cell membrane, which directly generates ROS and exacerbates oxidative stress.87,88
Ferroptosis and Immune Cells in AECs
In ARDS, various immune cells, including neutrophils, macrophages, and lymphocytes, are significantly involved in the inflammatory response, which is crucial for combating pathogens and repairing tissues. However, an excessive or dysregulated immune response can exacerbate lung injury and worsen the severity of ARDS. Ferroptosis not only leads to a reduction in the number and function of immune cells, but also triggers inflammation or specific responses upon recognition by immune cells.89
Ferroptosis attracts neutrophils through DAMPs, such as high mobility group box 1 (HMGB1), which recruits and activates neutrophils via the Toll-like receptor 4 (TLR4) signaling pathway.90 The oxidative products released during ferroptosis (eg, 4-HNE and MDA) can further promote ROS generation or activate NF-κB signaling in neutrophils, leading to their enhanced participation in the inflammatory response.91 While ferroptosis triggers neutrophil activation, these immune cells reciprocally amplify ferroptotic processes through multifaceted mechanisms. Neutrophils promote ferroptosis by activating the immune response, generating ROS, and secreting proinflammatory factors, such as TNF-α, IL-1β, and chemokines.92 Research has demonstrated that neutrophil extracellular traps (NETs) increase ROS generation through cell-cell interactions and mediate m6A modification of GPX4, thereby promoting ferroptosis.93 In ALI, neutrophils regulate hallmark genes related to ferroptosis, including SLC7A11, which is involved in antioxidant stress responses and lipid metabolism.51 Additionally, neutrophils transfer myeloperoxidase (MPO)-containing granules into target cells, where MPO catalyses the generation of strong oxidants (eg, hypochlorous acid) from hydrogen peroxide (H2O2), accelerating lipid peroxidation. MPO also facilitates the release or accumulation of iron, and increased free iron can catalyse ROS production through the Fenton reaction, further promoting ferroptosis.94
The sensitivities of inflammatory macrophages and selectively activated macrophages to ferroptosis differ.95 When cells undergo ferroptosis, DAMPs and other inflammatory mediators can activate macrophage polarization towards a proinflammatory phenotype (M1). Secreted inflammatory mediators further influence the activity of enzymes involved in iron, lipid, and amino acid metabolism.96 For example, 4-HNE can bind to pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), triggering inflammatory signalling cascades (eg, the NF-κB and MAPK pathways) that lead to macrophage activation.97,98 Furthermore, iron-requiring cells can produce oxygenated phosphatidylethanolamines, activating TLR2 on macrophages and promoting their clearance.99 In addition, macrophages recycle stored iron by clearing aged erythrocytes, resulting in the release of free iron and ROS, inducing ferroptosis in macrophages themselves that functionally constrains their immune effector capabilities.100 M1 macrophages secrete cytokines, such as TNF-α and interferon-gamma (IFN-γ),101 which transcriptionally upregulate transferrin receptor 1 (TFR1) and ferritin heavy chain (FTH1) expression in neighboring cells. This process increases the cellular uptake and storage of iron and thus promotes ferroptosis.102,103 Moreover, macrophages can promote tissue repair through the production of anti-inflammatory cytokines, such as interleukin-10 (IL-10) and TGF-β, in the M2 phenotype, which also induce the expression of antioxidant proteins.104 This immunoregulatory mechanism maintains systemic iron homeostasis while attenuating oxidative stress, thereby limiting ferroptosis-driven tissue injury and promoting healing. For example, quercetin can enhance M2 macrophage polarization and the expression of endogenous antioxidants, thereby inhibiting ferroptosis and assisting in tissue repair.105 Uridine alleviates sepsis-induced ALI by inhibiting macrophage ferroptosis.106
Ferroptosis and the involvement of CD4+ and CD8+ T cells in AECs are critical areas of research, especially in ARDS. A bidirectional interaction exists between ferroptosis and T-cell activity. When CD4+ and CD8+ T cells are recruited to AECs, cytokines, such as interleukin-2 (IL-2) and IFN-γ secreted by T helper 1 (Th1) cells of CD4+ T cells, promote ferroptosis by enhancing the inflammatory response, downregulating SLC7A11 and depleting GSH.107,108 Among the CD4+ subsets, regulatory T cells (Tregs) and T helper 17 (Th17) cells are closely associated with the prognosis of ARDS. Th17 cells promote inflammation, and Tregs play a key role in maintaining immune homeostasis by secreting anti-inflammatory cytokines such as IL-10 and transforming growth factor-β (TGF-β).109 Alanyl-glutamine (Ala-Gln) can reduce lipopolysaccharide (LPS)-mediated ALI by increasing the number of Treg cells and decreasing the proportion of Th17 cells.110 Similarly, losartan treatment inhibited Th17 polarization after LPS-induced ALI.111 CD8+ T cells not only directly target and kill abnormal or infected cells but also release IFN-γ, which downregulates the expression of the two subunits SLC3A2 and SLC7A11 of system xc-. This reduction impairs the ability of cells to take up cysteine, leading to increased lipid peroxidation and ferroptosis.112 Due to its ability to inhibit the activity of IFN-γ, proanthocyanidin is considered a potential therapeutic agent for treating ALI induced by ferroptosis.113 In addition, GPX4 is crucial for maintaining peripheral T-cell homeostasis; the absence of GPX4 in CD8+ and CD4+ T cells leads to rapid accumulation of membrane LPOs and induces ferroptosis.114 Conversely, CD36-mediated ferroptosis exerts biphasic effects on T-cell immunity: it suppresses CD8+ T cell effector functions while simultaneously disrupting CD4+ T cell homeostasis.115,116 This regulatory axis becomes pathologically relevant during ferroptosis activation, where danger-associated molecular patterns (DAMPs) from dying cells enhance CD8+ T cell activation through co-stimulatory receptor signaling, creating a feedforward loop with IFN-γ-mediated amplification.117–119 However, when ferroptosis induces excessive levels of immunosuppressive factors or creates a strong oxidative stress environment, the ROS and 4-HNE produced can directly damage CD8+ T cells, reducing their proliferation and cytotoxic functions.120 Furthermore, in inflammatory or anti-infective environments, ferroptosis can promote the activity of CD4+ T cells (such as Th1/Th17 cells) by downregulating the xCT-GSH-GPX4 pathway.121 Conversely, chronic inflammation or immunosuppressive environments may suppress the effector functions of CD4+ T cells through the activation of Treg cells122 (Figure 3).
![]() |
Figure 3 Ferroptosis and immune cells in AECs.
|
Ferroptosis Leads to Impaired Repair Functions in Lung Injury
Type II AECs are responsible for the production of surfactant, a lipoprotein complex that reduces surface tension and prevents alveolar collapse.123 Ferroptosis in type II cells can impair surfactant production and disrupt normal alveolar fluid clearance, leading to further respiratory compromise.6,124 In type II AECs, cGAS promotes ferroptosis in lung injury by enhancing NCOA4-mediated ferritin phagocytosis.125 Additionally, type II cells are involved in the repair of damaged alveolar epithelium, and ferroptosis may hinder this regenerative process, delaying recovery and promoting chronic lung injury.124 Diminished surfactant production increases the risk of alveolar collapse, which contributes to atelectasis and further impedes gas exchange. This disruption of homeostasis within the alveolar environment not only prolongs the inflammatory state but also promotes the progression of ARDS.126 In addition to affecting surfactant production and tissue repair, ferroptosis may have broader implications in lung injury. For example, the secretory profile of ferroptotic type II AECs may influence the inflammatory landscape in the lungs, including the recruitment and activation of immune cells.127 This process could lead to a vicious cycle in which inflammation drives more pronounced ferroptosis, further inhibiting the reparative capacity of type II AECs.
Ferroptosis Leads to Disruption of the Alveolar‒Capillary Barrier
Ferroptosis disrupts alveolar-capillary barrier integrity through mechanisms that compromise pulmonary function and gas exchange. This critical barrier, composed of alveolar epithelial cells (AECs) and endothelial cells, prevents vascular leakage while maintaining efficient oxygenation.128 When AECs undergo ferroptosis, lipid peroxidation occurs, and excessive ROS accumulation can control renin‒angiotensin system (RAS) activation of matrix metalloproteinases (MMPs) to degrade the basement membrane, thereby impairing the integrity of the cell membrane.129 This process results in increased membrane tension in AECs, which activates Piezo1 and transient receptor potential (TRP) channels, thus increasing cation permeability.130 Concurrently, tight junction protein complexes become dysfunctional, leading to increased paracellular permeability. This barrier breakdown permits protein-rich edema fluid accumulation in alveolar spaces, exacerbating pulmonary edema and hypoxemia. Furthermore, AEC injury can trigger a cascade of inflammatory responses, inviting additional immune cells to the site of injury, which may further perpetuate the cycle of inflammation and oxidative damage. Consequently, the loss of functional AECs due to ferroptosis not only destabilizes the barrier but also hampers the repair processes necessary for recovery from lung injuries, contributing significantly to the progression of ARDS.
Targeting Ferroptosis as a Treatment Strategy for ARDS
Increasing research has indicated that dysregulated iron metabolism is reported in ARDS patients.131 Specifically, compared to healthy controls, the levels of total iron and non-heme iron in the plasma and bronchoalveolar lavage fluid of ARDS patients are elevated.132 It has been found that the levels of iron and iron-related proteins in bronchoalveolar lavage fluid (BALF) are associated with the severity of ARDS.133 Moreover, iron and iron-related proteins are found to excess in the lower respiratory tract of ARDS patients.132 Ferroptosis-related genes, such as Cp, Slc7a11 and Slc39a14, have been identified to as key regulators in patients to aggravate ARDS.51 Lipid hydroperoxides have also been detected in the pulmonary edema fluid of ARDS patients.134 In experimental models, the use of ferroptosis inhibitor Fer-1 has been shown to alleviate lung injury in ARDS.135 Furthermore, Panax ginseng activates the Keap1-Nrf2/HO-1 pathway,136 and adenosine diphosphate (ADP) (by modulating the SOD1/CREB1/HMOX1 pathway),115 reducing the generation of lipid peroxides, thus mitigating the effects of ferroptosis on LPS-induced ALI in mice. These studies suggest that ferroptosis-targeted therapeutic strategies for ARDS hold promise in reducing lung injury and improving patient prognosis. Because ferroptosis is characterized by iron-dependent oxidative cell death due to lethal lipid peroxidation, interventions aimed at modulating this process may help preserve cellular integrity and reduce inflammation in the lungs. These interventions mainly include the use of antioxidants, alone or in combination with anti-inflammatory drugs.
Antioxidants and Their Role in Preventing Oxidative Damage and Ferroptosis
Pharmacological Antioxidants
One of the primary strategies for targeting ferroptosis involves enhancing antioxidant defences within pulmonary cells, particularly by maintaining or restoring the levels of GSH. Previous study found that total GSH was deficient in the alveolar epithelial lining fluid of patients with ARDS compared to normal subjects.137 It has been reported that oral and intravenous GSH supplementation could relieve dyspnea in patients with ARDS caused by COVID-19.138 Studies using animal models have demonstrated that the ROS inhibitors GKT137831 and Apocynin can reduce lung ischaemia/reperfusion injury in mice by blocking Nox4-dependent H2O2 production.139 Mitochondrial-targeted antioxidants, such as the SOD mimetics MitoQ and MitoTempo,140 the SOD derivative phospholipated SOD2 (PC-SOD),141 and recombinant SOD1,142 have also been shown to significantly improve lung injury in mice. Recent research demonstrates that dipyridamole binds to and activates SOD1 to mitigate ferroptosis and pulmonary damage in ARDS mouse models. Furthermore, the clinical trial further corroborates that dipyridamole adjunctive therapy improves outcomes in ARDS patients.143 In recent ongoing clinical trials, the role of antioxidants in critical care is being more closely evaluated. For instance, the use of mitochondrial-targeted antioxidants like MitoQ is currently under investigation in ARDS patients, with preliminary findings suggesting improved clinical outcomes through reduction in oxidative stress and enhanced mitochondrial function.144 Clinical trials are exploring the efficacy of antioxidants like MitoQ in sepsis-induced ARDS, providing key insights into potential therapeutic avenues for modulating ferroptosis and oxidative damage in critically ill patients.145 Moreover, the organoselenium compound ebselen, which mimics the catalytic mechanism of glutathione peroxidase (GPx), reacts with hydroperoxides, including H2O2, thereby inactivating them and facilitating the regeneration of GSH.146 By regulating the TLR4/MyD88/NF-κB and Keap1/Nrf2/HO-1 pathway, bakuchiol reduces the levels of MDA and 4-HNE and the expression of proinflammatory cytokines in lung tissue, increases the activities of SOD and GPx, and alleviates LPS-induced ALI.147 In addition to increasing GSH levels, pharmacological agents that inhibit key enzymes involved in lipid peroxidation can serve as effective treatments. For example, lipoxygenase inhibitors can inhibit the oxidation of polyunsaturated fatty acids, thereby reducing the formation of toxic LPOs and preventing the initiation of ferroptosis.148 These inhibitors are currently being tested in clinical trials focused on reducing inflammatory responses and cellular damage during the acute phase of ARDS (Figure 4).
![]() |
Figure 4 Pharmacological antioxidants.
|
Nuclear Factor Erythroid 2-Related Factor 2
In addition to increasing GSH levels, the transcription factor Nrf2 regulates antioxidant gene expression.149 During oxidative stress, cysteines in Keap1 are oxidized, leading to the release of Nrf2 through the Cullin3/Rbx1 ubiquitination system.150 Nrf2 can activate the expression of various factors involved in the detoxification of ROS, triggering an antioxidant response, including glutamate-cysteine ligase catalytic subunit (GCLC), glutamate-cysteine ligase modifier subunit (GCLM), GPx, glutathione reductase (GR), haem oxygenase-1 (HO-1), peroxiredoxins (Prx), and SOD3.151 Animal models have shown that Nrf2 can bind to antioxidant response elements (AREs) in the promoter regions of target genes, with Nrf2-deficient mice exhibiting exacerbated intestinal ischemia/reperfusion-induced ALI.152 Additionally, Nrf2 possesses anti-inflammatory functions; it prevents macrophages from activating to the M1 phenotype and promotes alternative activation towards the M2 phenotype.153 Consequently, studies have shown that pharmacological agents such as tert-butylhydroquinone (tBHQ),154 bardoxolone (CDDO),155 and resveratrol156 can stabilize Nrf2 by altering its binding with Keap1. The Nrf2 activator RTA408 alleviates LPS-induced acute lung injury by inhibiting BACH1-mediated ferroptosis.157
Iron Homeostasis
Moreover, manipulating iron homeostasis represents another critical strategy for mitigating ferroptosis. Iron chelators, such as deferoxamine, are molecules that promote tissue regeneration and angiogenesis and possess antioxidant properties;158 they can be used to reduce excessive intracellular iron levels, thereby decreasing the overall susceptibility of cells to ferroptosis.159 In recent trials, iron chelation therapy has been shown to reduce iron accumulation in the lungs, with studies investigating the effects of deferoxamine on improving lung function and reducing inflammation in ARDS patients. Deferoxamine reduces iron accumulation, increases dopamine levels, and alters the expression of iron transport proteins, α-synuclein, and oxidative stress markers.160 By restricting free iron availability, these agents can prevent the catalysis of harmful Fenton reactions that generate ROS, thus protecting AECs from oxidative stress and toxicity.161 Studies have shown that iron chelators exhibit iron-chelating, antiviral, and immunomodulatory effects both in vitro and in vivo.162 They can reduce the activity of NF-κB, which inhibits the synthesis of IL-6 and helps alleviate inflammatory responses.163 By mediating the activation of triggering receptors expressed on myeloid cells-2 (TREM2) through the mTOR signaling pathway, they induce autophagy in microglia following traumatic brain injury, suppressing ferroptosis and reducing brain oedema and neuroinflammation.164,165 Furthermore, maintaining iron homeostasis helps mitigate cellular ageing and inflammation.166 Additionally, these agents can inhibit viral replication, decrease iron utilization, increase B-cell activity, and suppress inflammation, which collectively reduces iron accumulation in the lungs and improves lung function.167
Intracellular Nutrients Also Support Approaches Targeting Ferroptosis
Various intracellular nutrients, such as Coenzyme Q10 (CoQ10) and vitamins, are closely related to cellular antioxidant systems and participate in the regulation of ferroptosis.168 CoQ10 increases the phosphorylation of 5′-monophosphate (AMP)-activated protein kinase (AMPK), which can prevent the production of LPOs. Pharmacological CoQ10 supplementation demonstrates potent anti-ferroptotic effects through AMPK-mediated inhibition of lipid peroxidation.169 Similarly, the CoQ10 analogue idebenone can also block cellular lipid peroxidation by inhibiting excessive autophagy through the ROS-AMPK-mTOR pathway, thereby alleviating ferroptosis.170 Inhibitors of 3-hydroxyl-3-methylglutaryl coenzyme A (HMG-CoA) reductase, which has a clearance effect on CoQ10, can downregulate GPX4 to induce ferroptosis by inhibiting the mevalonate pathway.171 In addition, ferroptosis suppressor protein 1 (FSP1), an NAD(P)H-quinone oxidoreductase, inhibits ferroptosis by acting on the CoQ10-NADPH axis and the vitamin K reduction cycle,172 whereas curcumin induces ferroptosis by inhibiting the FSP1-CoQ10-NADPH pathway.173
Among vitamins, vitamin A synergizes with other ferroptosis-inhibitory vitamins (eg, E and K) through redox-modulating mechanisms. Additionally, the metabolites of vitamin A, all-trans-retinal (atRAL) and all-trans-retinoic acid (atRA), directly neutralize lipid-derived free radicals involved in ferroptosis.174 Similarly, vitamin B6 promotes the expression of antioxidant enzymes by mediating Nrf2 expression, thereby inhibiting LPS-induced ferroptosis.175 Animal studies have indicated that vitamin C is also an antioxidant with protective effects against ALI in mice.176 Vitamin D not only alleviates cognitive impairment and mitigates ferroptosis in ageing mice via the Nrf2 signaling pathway,177 but also reduces inflammation by activating the Klotho/p53 signaling pathway178 and mediating ACSL4.179 As a lipid-soluble antioxidant, vitamin E helps stabilize cell membranes, alleviating the impact of lipid peroxidation, and synergizes with GPX4 to protect Treg cells from ferroptosis, thus reducing inflammation.180 Mucin 1 demonstrates pleiotropic protective effects by simultaneously inhibiting ferroptosis and amplifying vitamin E efficacy through GSK3β/Keap1-Nrf2-GPX4 axis modulation, thereby attenuating sepsis-induced acute lung injury.181 In addition to its traditional function related to blood coagulation as a cofactor for γ-glutamyl carboxylase, vitamin K possesses strong ferroptosis-inhibitory properties.182 Vitamin K2 functions by activating GPX4 and suppressing ferroptosis and extracellular matrix degradation.183,184 FSP1 effectively reduces vitamin K to its hydroquinone form, acting as a potent free radical scavenger and inhibitor of lipid peroxidation, thus protecting cells from harmful lipid peroxidation and ferroptosis.185
Additionally, dietary intake of monounsaturated fatty acids (MUFAs) and PUFAs modulates ferroptosis by directly alteration of membrane lipid composition.186 Moreover, amino acids and glucose intersect with the ferroptosis pathway through nutrient-sensitive kinase mechanistic target of rapamycin complex 1 (mTORC1)187,188 and AMPK signaling. The inactivation of upstream regulatory factors of mTORC1 and the negative regulator AMPK can promote the expression of SREBP1/SCD1, a key transcriptional regulator of lipogenesis, thereby inhibiting ferroptosis through the production of MUFAs.189 Synergistic application of these pharmacological antioxidant strategies shows therapeutic potential for redox imbalance management in ARDS (Figure 5).
![]() |
Figure 5 Targeting ferroptosis as a treatment strategy for ARDS.
|
Summary
In conclusion, ARDS constitutes a life-threatening clinical entity arising from rapid respiratory failure, driven by pathogenic interactions between inflammatory cascades and alveolar-capillary barrier dysfunction. The multifactorial etiology of ARDS, exemplified by pneumonia and sepsis pathogenesis, underscores the imperative for mechanistic elucidation of its pathophysiological underpinnings. Emerging evidence positions ferroptosis—a regulated cell death modality characterized by iron-dependent LPO accumulation—as a central pathological mediator in ARDS. The increased susceptibility of AECs to ferroptosis underscores their potential role in exacerbating lung injury and compromising gas exchange—critical functions necessary for patient survival. Moreover, the inflammatory microenvironment in ARDS further facilitates this unique form of cell death, suggesting that ferroptosis not only contributes to lung damage but may also serve as a therapeutic target.
Importantly, ferroptosis represents a critical intersection between molecular pathology and clinical outcomes in ARDS. The ferroptosis of AECs under oxidative stress transcends mere cellular attrition, functioning as a pathological linchpin that drives disease progression—from acute inflammatory exacerbation to irreversible pulmonary fibrosis. This dual-phase role implies that targeting ferroptosis may not only mitigate epithelial injury in the early stage but also preserve tissue regenerative potential in the late fibrotic phase. These insights underscore the translational significance of ferroptosis, providing a molecular framework for the development of stage-specific diagnostic markers and treatment strategies.
Advancements in understanding the tripartite interplay between ferroptosis, oxidative stress, and inflammation in AECs reveal compelling therapeutic opportunities for targeted modulation of ferroptotic pathways. Strategies aimed at modulating ferroptosis through antioxidants and nutrient support represent an innovative approach to mitigate oxidative damage and improve lung repair processes. Future research should continue to explore the diverse roles of ferroptosis in ARDS, focusing on clarifying its dual nature as both a facilitator of injury and a potential target for intervention. Translational integration of these mechanistic insights could enhance clinical diagnostics while catalyzing development of phase-specific therapeutics to reduce ARDS-associated morbidity and mortality, ultimately improving patient prognoses.
Abbreviations
ARDS, Acute Respiratory Distress Syndrome; AECs, alveolar epithelial cells; TNF-α, tumor necrosis factor-alpha; IL-6, interleukin-6; IL-1β, interleukin-1β; ROS, reactive oxygen species; LPOs, lipid peroxides; GSH, Glutathione; GPX4, Glutathione peroxidase 4; SLC3A2, solute carrier 3A2; SLC7A11, solute carrier family 7 member 11; Nrf2, nuclear factor E2 related factor 2; ATP, adenosine triphosphate; DAMPs, damage-associated molecular patterns; PUFAs, polyunsaturated fatty acids; NOX, NADPH oxidase; NADPH, nicotinamide adenine dinucleotide phosphate; COX-2, cyclooxygenase-2; SOD, superoxide dismutase; 4-HNE, 4-hydroxynonenal; MDA, malondialdehyde; HMGB1, high mobility group box 1; TLR4, Toll-like receptor4; NETs, neutrophil extracellular traps; MPO, myeloperoxidase; TLRs, Toll-like receptors; PRRs, pattern recognition receptors; IFN-γ, interferon-gamma; TFR1, transferrin receptor1; FTH1, ferritin heavy chain; IL-10, interleukin-10; TGF-β, transforming growth factor-β; Ala-Gln, Alanyl-glutamine; LPS, Lipopolysaccharide; MMPs, matrix metalloproteinases; TRP, transient receptor potential; RAS, renin-angiotensin system; PC-SOD, phospholipatedSOD2; GPx, glutathione peroxidase; GCLC, glutamate-cysteine ligase catalytic subunit; HO-1, heme oxygenase-1; GR, glutathione reductase; GCLM, glutamate-cysteine ligase modifier subunit; Prx, peroxiredoxins; AREs, antioxidant response elements; tBHQ, tert-butylhydroquinone; CDDO, bardoxolone; TREM2, triggering receptors expressed on myeloid cells-2; CoQ10, CoenzymeQ10; AMPK, AMP-activated protein kinase; AMP, 5′-monophosphate; HMG-CoA, 3-hydroxyl-3-methylglutarylcoenzymeA; FSP1, ferroptosis suppressor protein 1; atRAL, all-trans-retinal; atRA, all-trans-retinoic acid; MUFA, monounsaturated fatty acids; mTORC1, mechanistic target of rapamycin complex 1; FPN, Ferroportin; Fe2+, ferrous iron; Fe3+, ferric iron; LIP, labile iron pool; TCA, mitochondrial tricarboxylic acid cycle; PUFA-PL, phospholipid with polyunsaturated fatty acid; ACSL4, acyl-CoA synthetase long-chain family member 4; LPCAT3, lysophosphatidylcholine acyltransferase 3; ALOXS, arachidonate lipoxygenase; GLS2, glutaminase 2; ACLY, ATP citrate lyase; IPP, intracisternal A particle-promoted polypeptide; Glu, glutamate; GSSG, L-Glutathione Oxidized; NFE2L2, nuclear factor, erythroid 2-like 2; CARS1, cysteinyl-tRNA synthetase; ARF, cyclin-dependent kinase inhibitor 2A; SCD1, stearoyl CoA desaturase; EGLN1, egl-9 family hypoxia-inducible factor 1; LSH, lymphoid tissue-specific helicase; BCAT2, branched chain amino-acid transaminase.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Gragossian A, Siuba MT. Acute Respiratory Distress Syndrome. Emergency Med Clin North Am. 2022;40(3):459–472. doi:10.1016/j.emc.2022.05.002
2. Grotberg JC, Reynolds D, Kraft BD. Management of severe acute respiratory distress syndrome: a primer. Crit Care. 2023;27(1):289. doi:10.1186/s13054-023-04572-w
3. Yıldırım F, Karaman İ, Kaya A. Current situation in ARDS in the light of recent studies: classification, epidemiology and pharmacotherapeutics. Güncel çalışmalar ışığında ARDS’de mevcut durum: sınıflandırma, epidemiyoloji ve farmakoterapötikler. Tuberk Toraks. 2021;69(4):535–546. doi:10.5578/tt.20219611
4. Liu X, Zhang J, Xie W. The role of ferroptosis in acute lung injury. Mol Cell Biochem. 2022;477(5):1453–1461. doi:10.1007/s11010-021-04327-7
5. Tu H, Tang L-J, Luo X-J, Ai K-L, Peng J. Insights into the novel function of system Xc- in regulated cell death. Eur Rev Med Pharmacol Sci. 2021;25(3):1650–1662. doi:10.26355/eurrev_202102_24876
6. Qi X, Luo Y, Xiao M, et al. Mechanisms of Alveolar Type 2 Epithelial Cell Death During Acute Lung Injury. Stem Cells. 2023;41(12):1113–1132. doi:10.1093/stmcls/sxad074
7. Frey A, Lunding LP, Wegmann M. The Dual Role of the Airway Epithelium in Asthma: active Barrier and Regulator of Inflammation. Cells. 2023;12(18):2208. doi:10.3390/cells12182208
8. Dou J, Liu X, Yang L, Huang D, Tan X. Ferroptosis interaction with inflammatory microenvironments: mechanism, biology, and treatment. Biomed Pharmacother. 2022;155:113711. doi:10.1016/j.biopha.2022.113711
9. Pokharel MD, Garcia-Flores A, Marciano D, et al. Mitochondrial network dynamics in pulmonary disease: bridging the gap between inflammation, oxidative stress, and bioenergetics. Redox Biol. 2024;70:103049. doi:10.1016/j.redox.2024.103049
10. Tang W, Tang R, Zhao Y, Peng J, Wang D. Comparison of Clinical Characteristics and Predictors of Mortality between Direct and Indirect ARDS. Medicina. 2022;58(11):1563. doi:10.3390/medicina58111563
11. Long ME, Mallampalli RK, Horowitz JC. Pathogenesis of pneumonia and acute lung injury. Clin Sci. 2022;136(10):747–769. doi:10.1042/CS20210879
12. White JJ, Cambron JD, Gottlieb M, Long B. Evaluation and Management of Airway Foreign Bodies in the Emergency Department Setting. J Emerg Med. 2023;64(2):145–155. doi:10.1016/j.jemermed.2022.12.008
13. Bittner E, Sheridan R. Acute Respiratory Distress Syndrome, Mechanical Ventilation, and Inhalation Injury in Burn Patients. Surg Clin North Am. 2023;103(3):439–451. doi:10.1016/j.suc.2023.01.006
14. Sayed MS, Elmeslmany KA, Elsawy AS, Mohamed NA. The Validity of Quantifying Pulmonary Contusion Extent by Lung Ultrasound Score for Predicting ARDS in Blunt Thoracic Trauma. Crit Care Res Pract. 2022;2022:3124966. doi:10.1155/2022/3124966
15. Wei S, Shen Z, Yin Y, et al. Advances of presepsin in sepsis-associated ARDS. Postgrad Med J. 2024;100(1182):209–218. doi:10.1093/postmj/qgad132
16. Bos LDJ, Ware LB. Acute respiratory distress syndrome: causes, pathophysiology, and phenotypes. Lancet. 2022;400(10358):1145–1156. doi:10.1016/S0140-6736(22)01485-4
17. Toya S, Struyf S, Huerta L, et al. A narrative review of chemokine receptors CXCR1 and CXCR2 and their role in acute respiratory distress syndrome. Eur Respir Rev. 2024;33(173):230172. doi:10.1183/16000617.0172-2023
18. Qiao X, Yin J, Zheng Z, Li L, Feng X. Endothelial cell dynamics in sepsis-induced acute lung injury and acute respiratory distress syndrome: pathogenesis and therapeutic implications. Cell Commun Signal. 2024;22(1):241. doi:10.1186/s12964-024-01620-y
19. Lupu L, Palmer A, Huber-Lang M. Inflammation, Thrombosis, and Destruction: the Three-Headed Cerberus of Trauma- and SARS-CoV-2-Induced ARDS. Front Immunol. 2020;11:584514. doi:10.3389/fimmu.2020.584514
20. Chen X, Comish PB, Tang D, Kang R. Characteristics and Biomarkers of Ferroptosis. Front Cell Dev Biol. 2021;9:637162. doi:10.3389/fcell.2021.637162
21. Chen Y, Guo X, Zeng Y, et al. Oxidative stress induces mitochondrial iron overload and ferroptotic cell death. Sci Rep. 2023;13(1):15515. doi:10.1038/s41598-023-42760-4
22. Wang C, Wang Z, Zeng B, et al. Fenton-like reaction of the iron(II)-histidine complex generates hydroxyl radicals: implications for oxidative stress and Alzheimer’s disease. Chem Commun. 2021;57(92):12293–12296. doi:10.1039/d1cc05000a
23. Pope LE, Dixon SJ. Regulation of ferroptosis by lipid metabolism. Trends Cell Biol. 2023;33(12):1077–1087. doi:10.1016/j.tcb.2023.05.003
24. Liu T, Sun L, Zhang Y, Wang Y, Zheng J. Imbalanced GSH/ROS and sequential cell death. J Biochem Mol Toxicol. 2022;36(1):e22942. doi:10.1002/jbt.22942
25. Zhang XD, Liu ZY, Wang MS, et al. Mechanisms and regulations of ferroptosis. Front Immunol. 2023;14:1269451. doi:10.3389/fimmu.2023.1269451
26. Ursini F, Maiorino M. Lipid peroxidation and ferroptosis: the role of GSH and GPx4. Free Radic Biol Med. 2020;152:175–185. doi:10.1016/j.freeradbiomed.2020.02.027
27. Li P, Jiang M, Li K, et al. Glutathione peroxidase 4-regulated neutrophil ferroptosis induces systemic autoimmunity. Nat Immunol. 2021;22(9):1107–1117. doi:10.1038/s41590-021-00993-3
28. Cui C, Yang F, Li Q. Post-Translational Modification of GPX4 is a Promising Target for Treating Ferroptosis-Related Diseases. Front Mol Biosci. 2022;9:901565. doi:10.3389/fmolb.2022.901565
29. Zhang W, Liu Y, Liao Y, Zhu C, Zou Z. GPX4, ferroptosis, and diseases. Biomed Pharmacother. 2024;174:116512. doi:10.1016/j.biopha.2024.116512
30. Liu Y, Wan Y, Jiang Y, Zhang L, Cheng W. GPX4: the hub of lipid oxidation, ferroptosis, disease and treatment. Biochim Biophys Acta Rev Cancer. 2023;1878(3):188890. doi:10.1016/j.bbcan.2023.188890
31. Liu MR, Zhu WT, Pei DS. System Xc-: a key regulatory target of ferroptosis in cancer. Invest New Drugs. 2021;39(4):1123–1131. doi:10.1007/s10637-021-01070-0
32. Rochette L, Dogon G, Rigal E, et al. Lipid Peroxidation and Iron Metabolism: two Corner Stones in the Homeostasis Control of Ferroptosis. Int J Mol Sci. 2022;24(1):449. doi:10.3390/ijms24010449
33. Raghu G, Berk M, Campochiaro PA, et al. The Multifaceted Therapeutic Role of N-Acetylcysteine (NAC) in Disorders Characterized by Oxidative Stress. Curr Neuropharmacol. 2021;19(8):1202–1224. doi:10.2174/1570159X19666201230144109
34. Parker JL, Deme JC, Kolokouris D, et al. Molecular basis for redox control by the human cystine/glutamate antiporter system xc. Nat Commun. 2021;12(1):7147. doi:10.1038/s41467-021-27414-1
35. Chen X, Liu C, Yu R, et al. Interaction between ferroptosis and TNF-α: impact in obesity-related osteoporosis. FASEB J. 2023;37(6):e22947. doi:10.1096/fj.202201958R
36. He Q, Liu M, Huang W, et al. IL-1β-Induced Elevation of Solute Carrier Family 7 Member 11 Promotes Hepatocellular Carcinoma Metastasis Through Up-regulating Programmed Death Ligand 1 and Colony-Stimulating Factor 1. Hepatology. 2021;74(6):3174–3193. doi:10.1002/hep.32062
37. Hou H, Qin X, Li G, et al. Nrf2-mediated redox balance alleviates LPS-induced vascular endothelial cell inflammation by inhibiting endothelial cell ferroptosis. Sci Rep. 2024;14(1):3335. doi:10.1038/s41598-024-53976-3
38. Dong H, Xia Y, Jin S, et al. Nrf2 attenuates ferroptosis-mediated IIR-ALI by modulating TERT and SLC7A11. Cell Death Dis. 2021;12(11):1027. doi:10.1038/s41419-021-04307-1
39. Li J, Zhou Y, Wang H, et al. Oxidative Stress-Induced Ferroptosis in Cardiovascular Diseases and Epigenetic Mechanisms. Front Cell Dev Biol. 2021;9:685775. doi:10.3389/fcell.2021.685775
40. Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life Sci. 2016;73(11–12):2195–2209. doi:10.1007/s00018-016-2194-1
41. Dickson KB, Zhou J. Role of reactive oxygen species and iron in host defense against infection. Front Biosci. 2020;25(8):1600–1616. doi:10.2741/4869
42. Duan G, Li J, Duan Y, et al. Mitochondrial Iron Metabolism: the Crucial Actors in Diseases. Molecules. 2022;28(1):29. doi:10.3390/molecules28010029
43. Kuang F, Liu J, Tang D, Kang R. Oxidative Damage and Antioxidant Defense in Ferroptosis. Front Cell Dev Biol. 2020;8:586578. doi:10.3389/fcell.2020.586578
44. Ali SS, Ahsan H, Zia MK, Siddiqui T, Khan FH. Understanding oxidants and antioxidants: classical team with new players. J Food Biochem. 2020;44(3):e13145. doi:10.1111/jfbc.13145
45. Park WH. Enhanced cell death effects of MAP kinase inhibitors in propyl gallate-treated lung cancer cells are related to increased ROS levels and GSH depletion. Toxicol In Vitro. 2021;74:105176. doi:10.1016/j.tiv.2021.105176
46. Tan M, Yin Y, Ma X, et al. Glutathione system enhancement for cardiac protection: pharmacological options against oxidative stress and ferroptosis. Cell Death Dis. 2023;14(2):131. doi:10.1038/s41419-023-05645-y
47. Wang B, Wang Y, Zhang J, et al. ROS-induced lipid peroxidation modulates cell death outcome: mechanisms behind apoptosis, autophagy, and ferroptosis. Arch Toxicol. 2023;97(6):1439–1451. doi:10.1007/s00204-023-03476-6
48. Jakubczyk K, Dec K, Kałduńska J, et al. Reactive oxygen species – sources, functions, oxidative damage. Pol Merkur Lekarski. 2020;48(284):124–127.
49. Wen Y, Liu Y, Liu W, et al. Ferroptosis: a potential target for acute lung injury. Inflamm Res. 2024;73(10):1615–1629. doi:10.1007/s00011-024-01919-z
50. Xu L, Zhang L, Xiang Y, Zhang X. Knockdown of lncRNA NEAT1 suppresses streptococcus pneumoniae-induced ferroptosis in alveolar epithelial cells by regulating the Nrf2-GPX4 pathway. Toxicon. 2024;243:107705. doi:10.1016/j.toxicon.2024.107705
51. Ma A, Feng Z, Li Y, et al. Ferroptosis-related signature and immune infiltration characterization in acute lung injury/acute respiratory distress syndrome. Respir Res. 2023;24(1):154. doi:10.1186/s12931-023-02429-y
52. Ding K, Liu C, Li L, et al. Acyl-CoA synthase ACSL4: an essential target in ferroptosis and fatty acid metabolism. Chin Med J. 2023;136(21):2521–2537. doi:10.1097/CM9.0000000000002533
53. Yu T, Sun S. Role and mechanism of ferroptosis in acute lung injury. Cell Cycle. 2023;22(19):2119–2129. doi:10.1080/15384101.2023.2278328
54. Li B, Wang Z, Yuan J, Liang D, Cheng Y, Wang Z. Knockdown of SHP2 attenuated LPS-induced ferroptosis via downregulating ACSL4 expression in acute lung injury. Allergol Immunopathol. 2023;51(3):143–152. doi:10.15586/aei.v51i3.856
55. Wu D, Spencer CB, Ortoga L, Zhang H, Miao C. Histone lactylation-regulated METTL3 promotes ferroptosis via m6A-modification on ACSL4 in sepsis-associated lung injury. Redox Biol. 2024;74:103194. doi:10.1016/j.redox.2024.103194
56. Chen H, Lin X, Yi X, et al. SIRT1-mediated p53 deacetylation inhibits ferroptosis and alleviates heat stress-induced lung epithelial cells injury. Int J Hyperthermia. 2022;39(1):977–986. doi:10.1080/02656736.2022.2094476
57. Yang Y, Ma Y, Li Q, et al. STAT6 inhibits ferroptosis and alleviates acute lung injury via regulating P53/SLC7A11 pathway. Cell Death Dis. 2022;13(6):530. doi:10.1038/s41419-022-04971-x
58. Ryter SW. Heme Oxygenase-1: an Anti-Inflammatory Effector in Cardiovascular, Lung, and Related Metabolic Disorders. Antioxidants. 2022;11(3):555. doi:10.3390/antiox11030555
59. Dong H, Qiang Z, Chai D, et al. Nrf2 inhibits ferroptosis and protects against acute lung injury due to intestinal ischemia reperfusion via regulating SLC7A11 and HO-1. Aging. 2020;12(13):12943–12959. doi:10.18632/aging.103378
60. Xiong M, Luo R, Zhang Z, et al. IL-27 regulates macrophage ferroptosis by inhibiting the Nrf2/HO1 signaling pathway in sepsis-induced ARDS. Inflamm Res. 2025;74(1):39. doi:10.1007/s00011-024-01986-2
61. Park HS, Kim SR, Lee YC. Impact of oxidative stress on lung diseases. Respirology. 2009;14(1):27–38. doi:10.1111/j.1440-1843.2008.01447
62. Ezraty B, Gennaris A, Barras F, Collet JF. Oxidative stress, protein damage and repair in bacteria. Nat Rev Microbiol. 2017;15(7):385–396. doi:10.1038/nrmicro.2017.26
63. Hanson RL, Batchelor E. Coordination of MAPK and p53 dynamics in the cellular responses to DNA damage and oxidative stress. Mol Syst Biol. 2022;18(12):e11401. doi:10.15252/msb.202211401
64. Gautam S, Alam F, Moin S, Noor N, Arif SH. Role of ferritin and oxidative stress index in gestational diabetes mellitus. J Diabetes Metab Disord. 2021;20(2):1615–1619. doi:10.1007/s40200-021-00911-2
65. Henning Y, Blind US, Larafa S, Matschke J, Fandrey J. Hypoxia aggravates ferroptosis in RPE cells by promoting the Fenton reaction. Cell Death Dis. 2022;13(7):662. doi:10.1038/s41419-022-05121-z
66. Guo Q, Li F, Duan Y, et al. Oxidative stress, nutritional antioxidants and beyond. Sci China Life Sci. 2020;63(6):866–874. doi:10.1007/s11427-019-9591-5
67. Chen GH, Song CC, Pantopoulos K, et al. Mitochondrial oxidative stress mediated Fe-induced ferroptosis via the NRF2-ARE pathway. Free Radic Biol Med. 2022;180:95–107. doi:10.1016/j.freeradbiomed.2022.01.012
68. Feng J, Wang J, Wang Y, et al. Oxidative Stress and Lipid Peroxidation: prospective Associations Between Ferroptosis and Delayed Wound Healing in Diabetic Ulcers. Front Cell Dev Biol. 2022;10:898657. doi:10.3389/fcell.2022.898657
69. Xiao M, Zhong H, Xia L, Tao Y, Yin H. Pathophysiology of mitochondrial lipid oxidation: role of 4-hydroxynonenal (4-HNE) and other bioactive lipids in mitochondria. Free Radic Biol Med. 2017;111:316–327. doi:10.1016/j.freeradbiomed.2017.04.363
70. Villar J, Zhang H, Slutsky AS. Lung Repair and Regeneration in ARDS: role of PECAM1 and Wnt Signaling. Chest. 2019;155(3):587–594. doi:10.1016/j.chest.2018.10.022
71. Zhang F, Xiang Y, Ma Q, Guo E, Zeng X. A deep insight into ferroptosis in lung disease: facts and perspectives. Front Oncol. 2024;14:1354859. doi:10.3389/fonc.2024.1354859
72. Yin J, Xu X, Guo Y, et al. Repair and regeneration: ferroptosis in the process of remodeling and fibrosis in impaired organs. Cell Death Discov. 2024;10(1):424. doi:10.1038/s41420-024-02181-2
73. Wang C, Hua S, Song L. Ferroptosis in pulmonary fibrosis: an emerging therapeutic target. Front Physiol. 2023;14:1205771. doi:10.3389/fphys.2023.1205771
74. Otoupalova E, Smith S, Cheng G, Thannickal VJ. Oxidative Stress in Pulmonary Fibrosis. Compr Physiol. 2020;10(2):509–547. doi:10.1002/cphy.c190017
75. Gonzalez-Gonzalez FJ, Chandel NS, Jain M, Budinger GRS. Reactive oxygen species as signaling molecules in the development of lung fibrosis. Transl Res. 2017;190:61–68. doi:10.1016/j.trsl.2017.09.005
76. Schramm F, Schaefer L, Wygrecka M. EGFR Signaling in Lung Fibrosis. Cells. 2022;11(6):986. doi:10.3390/cells11060986
77. Gong Y, Wang N, Liu N, Dong H. Lipid peroxidation and GPX4 inhibition are common causes for Myofibroblast differentiation and ferroptosis. DNA Cell Biol. 2019;38:725–733. doi:10.1089/dna.2018.4541
78. Sanches Santos Rizzo Zuttion M, Moore SKL, Chen P, Beppu AK, Hook JL. New Insights into the Alveolar Epithelium as a Driver of Acute Respiratory Distress Syndrome. Biomolecules. 2022;12(9):1273. doi:10.3390/biom12091273
79. Guillot L, Nathan N, Tabary O, et al. Alveolar epithelial cells: master regulators of lung homeostasis. Int J Biochem Cell Biol. 2013;45(11):2568–2573. doi:10.1016/j.biocel.2013.08.009
80. Lin CR, Bahmed K, Kosmider B. Impaired Alveolar Re-Epithelialization in Pulmonary Emphysema. Cells. 2022;11(13):2055. doi:10.3390/cells11132055
81. Roque W, Romero F. Cellular metabolomics of pulmonary fibrosis, from amino acids to lipids. Am J Physiol Cell Physiol. 2021;320(5):C689–C695. doi:10.1152/ajpcell.00586.2020
82. Shahpoury P, Zhang ZW, Arangio A, et al. The influence of chemical composition, aerosol acidity, and metal dissolution on the oxidative potential of fine particulate matter and redox potential of the lung lining fluid. Environ Int. 2021;148:106343. doi:10.1016/j.envint.2020.106343
83. Wickert A, Schwantes A, Fuhrmann DC. Brüne B.Inflammation in a ferroptotic environment. Front Pharmacol. 2024;15:1474285. doi:10.3389/fphar.2024.1474285
84. Hsieh PC, Wu YK, Yang MC, et al. Deciphering the role of damage-associated molecular patterns and inflammatory responses in acute lung injury. Life Sci. 2022;305:120782. doi:10.1016/j.lfs.2022.120782
85. Han F, Li S, Yang Y, Bai Z. Interleukin-6 promotes ferroptosis in bronchial epithelial cells by inducing reactive oxygen species-dependent lipid peroxidation and disrupting iron homeostasis. Bioengineered. 2021;12(1):5279–5288. doi:10.1080/21655979.2021.1964158
86. Michaeloudes C, Abubakar-Waziri H, Lakhdar R, et al. Molecular mechanisms of oxidative stress in asthma. Mol Aspects Med. 2022;85:101026. doi:10.1016/j.mam.2021.101026
87. Taylor JP, Tse HM. The role of NADPH oxidases in infectious and inflammatory diseases. Redox Biol. 2021;48:102159. doi:10.1016/j.redox.2021.102159
88. Tanaka T, Saito Y, Matsuda K, et al. Cyclic mechanical stretch-induced oxidative stress occurs via a NOX-dependent mechanism in type II alveolar epithelial cells. Respir Physiol Neurobiol. 2017;242:108–116. doi:10.1016/j.resp.2017.04.007
89. Chen X, Kang R, Kroemer G, Tang D. Ferroptosis in infection, inflammation, and immunity. J Exp Med. 2021;218(6):e20210518. doi:10.1084/jem.20210518
90. Yang H, Wang H, Andersson U. Targeting Inflammation Driven by HMGB1. Front Immunol. 2020;11:484. doi:10.3389/fimmu.2020.00484
91. Xu T, Liu S, Ma T, et al. Aldehyde dehydrogenase 2 protects against oxidative stress associated with pulmonary arterial hypertension. Redox Biol. 2017;11:286–296. doi:10.1016/j.redox.2016.12.019
92. Yee PP, Wei Y, Kim SY, et al. Neutrophil-induced ferroptosis promotes tumor necrosis in glioblastoma progression. Nat Commun. 2020;11(1):5424. doi:10.1038/s41467-020-19193-y
93. Zhang H, Liu J, Zhou Y, et al. Neutrophil extracellular traps mediate m6A modification and regulates sepsis-associated acute lung injury by activating ferroptosis in alveolar epithelial cells. Int J Biol Sci. 2022;18(8):3337–3357. doi:10.7150/ijbs.69141
94. Xiao X, Saha P, Yeoh BS, et al. Myeloperoxidase deficiency attenuates systemic and dietary iron-induced adverse effects. J Nutr Biochem. 2018;62:28–34. doi:10.1016/j.jnutbio.2018.08.003
95. Piattini F, Matsushita M, Muri J, et al. Differential sensitivity of inflammatory macrophages and alternatively activated macrophages to ferroptosis. Eur J Immunol. 2021;51(10):2417–2429. doi:10.1002/eji.202049114
96. Li B, Gong J, Sheng S, et al. Increased hepcidin in hemorrhagic plaques correlates with iron-stimulated IL-6/STAT3 pathway activation in macrophages. Biochem Biophys Res Commun. 2019;515:394–400. doi:10.1016/j.bbrc.2019.05.123
97. Liu Z, Ma Y, Cui Q, et al. Toll-like receptor 4 plays a key role in advanced glycation end products-induced M1 macrophage polarization. Biochem Biophys Res Commun. 2020;531:602–608. doi:10.1016/j.bbrc.2020.08.014
98. Fisch D, Zhang T, Sun H, et al. Molecular definition of the endogenous Toll-like receptor signalling pathways. Nature. 2024;631(8021):635–644. doi:10.1038/s41586-024-07614-7
99. Luo X, Gong HB, Gao HY, et al. Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death Differ. 2021;28(6):1971–1989. doi:10.1038/s41418-020-00719-2
100. Youssef LA, Rebbaa A, Pampou S, et al. Increased erythrophagocytosis induces ferroptosis in red pulp macrophages in a mouse model of transfusion. Blood. 2018;131(23):2581–2593. doi:10.1182/blood-2017-12-822619
101. Ng CT, Fong LY, Abdullah MNH. Interferon-gamma (IFN-γ): reviewing its mechanisms and signaling pathways on the regulation of endothelial barrier function. Cytokine. 2023;166:156208. doi:10.1016/j.cyto.2023.156208
102. Wang Z, Yao X, Wang K, Wang B. TFR1-Mediated Iron Metabolism Orchestrates Tumor Ferroptosis and Immunity in Non-Small Cell Lung Cancer. J Environ Pathol Toxicol Oncol. 2024;43(2):1–12. doi:10.1615/JEnvironPatholToxicolOncol.2023049084
103. Sun Y, Ren Y, Song LY, et al. Targeting iron-metabolism: a potential therapeutic strategy for pulmonary fibrosis. Biomed Pharmacother. 2024;172:116270. doi:10.1016/j.biopha.2024.116270
104. Griess B, Mir S, Datta K, Teoh-Fitzgerald M. Scavenging reactive oxygen species selectively inhibits M2 macrophage polarization and their pro-tumorigenic function in part, via Stat3 suppression. Free Radic Biol Med. 2020;147:48–60. doi:10.1016/j.freeradbiomed.2019.12.018
105. Tsai CF, Chen GW, Chen YC, et al. Regulatory Effects of Quercetin on M1/M2 Macrophage Polarization and Oxidative/Antioxidative Balance. Nutrients. 2021;14(1):67. doi:10.3390/nu14010067
106. Lai K, Song C, Gao M, et al. Uridine Alleviates Sepsis-Induced Acute Lung Injury by Inhibiting Ferroptosis of Macrophage. Int J Mol Sci. 2023;24(6):5093. doi:10.3390/ijms24065093
107. Wei TT, Zhang MY, Zheng XH, et al. Interferon-γ induces retinal pigment epithelial cell Ferroptosis by a JAK1-2/STAT1/SLC7A11 signaling pathway in Age-related Macular Degeneration. FEBS J. 2022;289:1968–1983. doi:10.1111/febs.16272
108. McKinstry KK, Alam F, Flores-Malavet V, et al. Memory CD4 T cell-derived IL-2 synergizes with viral infection to exacerbate lung inflammation. PLoS Pathog. 2019;15(8):e1007989. doi:10.1371/journal.ppat.1007989
109. Thomas R, Qiao S, Yang X. Th17/Treg Imbalance: implications in Lung Inflammatory Diseases. Int J Mol Sci. 2023;24(5):4865. doi:10.3390/ijms24054865
110. Hou YC, Pai MH, Liu JJ, Yeh SL. Alanyl-glutamine resolves lipopolysaccharide-induced lung injury in mice by modulating the polarization of regulatory T cells and T helper 17 cells. J Nutr Biochem. 2013;24(9):1555–1563. doi:10.1016/j.jnutbio.2013.01.004
111. Liu J, Zhang PS, Yu Q, et al. Losartan inhibits conventional dendritic cell maturation and Th1 and Th17 polarization responses: Νovel mechanisms of preventive effects on lipopolysaccharide-induced acute lung injury. Int J Mol Med. 2012;29(2):269–276. doi:10.3892/ijmm.2011.818
112. Wang W, Green M, Choi JE, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569(7755):270–274. doi:10.1038/s41586-019-1170-y
113. Lv YW, Du Y, Ma SS, et al. Proanthocyanidins attenuates ferroptosis against influenza-induced acute lung injury in mice by reducing IFN-γ. Life Sci. 2023;314:121279. doi:10.1016/j.lfs.2022.121279
114. Matsushita M, Freigang S, Schneider C, et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J Exp Med. 2015;212(4):555–568. doi:10.1084/jem.20140857
115. Ma X, Xiao L, Liu L, et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 2021;33(5):1001–1012.e5. doi:10.1016/j.cmet.2021.02.015
116. Li H, Wang PF, Luo W, et al. CD36-mediated ferroptosis destabilizes CD4+ T cell homeostasis in acute Stanford type-A aortic dissection. Cell Death Dis. 2024;15(9):669. doi:10.1038/s41419-024-07022-9
117. Lin Z, Zou S, Wen K. The crosstalk of CD8+ T cells and ferroptosis in cancer. Front Immunol. 2024;14:1255443. doi:10.3389/fimmu.2023.1255443
118. Hayashi K, Nikolos F, Chan KS. Inhibitory DAMPs in immunogenic cell death and its clinical implications. Cell Stress. 2021;5(4):52–54. doi:10.15698/cst2021.04.247
119. Liao P, Wang W, Wang W, et al. CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell. 2022;40(4):365–378.e6. doi:10.1016/j.ccell.2022.02.003
120. Chávez MD, Tse HM. Targeting Mitochondrial-Derived Reactive Oxygen Species in T Cell-Mediated Autoimmune Diseases. Front Immunol. 2021;12:703972. doi:10.3389/fimmu.2021.703972
121. Kong Z, Cai S, Xie W, et al. CD4 + T cells ferroptosis is associated with the development of sepsis in severe polytrauma patients. Int Immunopharmacol. 2024;127:111377. doi:10.1016/j.intimp.2023.111377
122. Moreno Ayala MA, Campbell TF, Zhang C, et al. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8+ T cell antitumor immunity. Immunity. 2023;56(7):1613–1630.e5. doi:10.1016/j.immuni.2023.06.003
123. King SD, Chen SY. Recent progress on surfactant protein A: cellular function in lung and kidney disease development. Am J Physiol Cell Physiol. 2020;319(2):C316–C320. doi:10.1152/ajpcell.00195.2020
124. Wang Y, Wang L, Ma S, Cheng L, Yu G. Repair and regeneration of the alveolar epithelium in lung injury. FASEB J. 2024;38(8):e23612. doi:10.1096/fj.202400088R
125. Liu H, Wang J, Xiong J, Hu Z. cGAS deficiency mitigated PM2.5-induced lung injury by inhibiting ferroptosis. Ecotoxicol Environ Saf. 2024;288:117321. doi:10.1016/j.ecoenv.2024.117321
126. Ruaro B, Salton F, Braga L, et al. The History and Mystery of Alveolar Epithelial Type II Cells: focus on Their Physiologic and Pathologic Role in Lung. Int J Mol Sci. 2021;22(5):2566. doi:10.3390/ijms22052566
127. Cheng H, Feng D, Li X, et al. Iron deposition-induced ferroptosis in alveolar type II cells promotes the development of pulmonary fibrosis. Biochim Biophys Acta Mol Basis Dis. 2021;1867(12):166204. doi:10.1016/j.bbadis.2021.166204
128. Huang Q, Le Y, Li S, Bian Y. Signaling pathways and potential therapeutic targets in acute respiratory distress syndrome (ARDS). Respir Res. 2024;25(1):30. doi:10.1186/s12931-024-02678-5
129. Soccal PM, Gasche Y, Pache JC, et al. Matrix metalloproteinases correlate with alveolar-capillary permeability alteration in lung ischemia-reperfusion injury. Transplantation. 2000;70(7):998–1005. doi:10.1097/00007890-200010150-00002
130. Hirata Y, Cai R, Volchuk A, et al. Lipid peroxidation increases membrane tension, Piezo1 gating, and cation permeability to execute ferroptosis. Curr Biol. 2023;33(7):1282–1294.e5. doi:10.1016/j.cub.2023.02.060
131. Sun S, Shen J, Jiang J, Wang F, Min J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther. 2023;8:372. doi:10.1038/s41392-023-01606-1
132. Ghio AJ, Carter JD, Richards JH, et al. Iron and iron-related proteins in the lower respiratory tract of patients with acute respiratory distress syndrome. Crit Care Med. 2003;31:395–400. doi:10.1097/01.CCM.0000050284.35609.97
133. Lee EH, Shin MH, Gi M, et al. Inhibition of Pendrin by a small molecule reduces Lipopolysaccharide-induced acute Lung Injury. Theranostics. 2020;10(22):9913–9922. doi:10.7150/thno.46417
134. Cross CE, Forte T, Stocker R, et al. Oxidative stress and abnormal cholesterol metabolism in patients with adult respiratory distress syndrome. J Lab Clin Med. 1990;115:396–404.
135. Liu P, Feng Y, Li H, et al. Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis. Cell Mol Biol Lett. 2020;25:10. doi:10.1186/s11658-020-00205-0
136. Li J, Lu K, Sun F, et al. Panaxydol attenuates ferroptosis against LPS-induced acute lung injury in mice by Keap1-Nrf2/HO-1 pathway. J Transl Med. 2021;19(1):96. doi:10.1186/s12967-021-02745-1
137. Pacht ER, Timerman AP, Lykens MG, Merola AJ. Deficiency of alveolar fluid glutathione in patients with sepsis and the adult respiratory distress syndrome. Chest. 1991;100(5):1397–1403. doi:10.1378/chest.100.5.1397
138. Horowitz RI, Freeman PR, Bruzzese J. Efficacy of glutathione therapy in relieving dyspnea associated with COVID-19 pneumonia: a report of 2 cases. Respir Med Case Rep. 2020;30:101063. doi:10.1016/j.rmcr.2020.101063
139. Cui Y, Wang Y, Li G, et al. The Nox1/Nox4 inhibitor attenuates acute lung injury induced by ischemia-reperfusion in mice. PLoS One. 2018;13(12):e0209444. doi:10.1371/journal.pone.0209444
140. Park WH. Tempol differently affects cellular redox changes and antioxidant enzymes in various lung-related cells. Sci Rep. 2021;11(1):14869. doi:10.1038/s41598-021-94340-z
141. Tanaka KI, Tamura F, Sugizaki T, et al. Evaluation of Lecithinized Superoxide Dismutase for the Prevention of Acute Respiratory Distress Syndrome in Animal Models. Am J Respir Cell Mol Biol. 2017;56(2):179–190. doi:10.1165/rcmb.2016-0158OC
142. Wu NC, Liao FT, Cheng HM, et al. Intravenous superoxide dismutase as a protective agent to prevent impairment of lung function induced by high tidal volume ventilation. BMC Pulm Med. 2017;17(1):105. doi:10.1186/s12890-017-0448-9
143. Chen X, Shen J, Jiang X, et al. Characterization of dipyridamole as a novel ferroptosis inhibitor and its therapeutic potential in acute respiratory distress syndrome management. Theranostics. 2024;14(18):6947–6968. doi:10.7150/thno.102318
144. Cen M, Ouyang W, Zhang W, et al. MitoQ protects against hyperpermeability of endothelium barrier in acute lung injury via a Nrf2-dependent mechanism. Redox Biol. 2021;41:101936. doi:10.1016/j.redox.2021.101936
145. Hou L, Zhang J, Liu Y, et al. MitoQ alleviates LPS-mediated acute lung injury through regulating Nrf2/Drp1 pathway. Free Radic Biol Med. 2021;165:219–228. doi:10.1016/j.freeradbiomed.2021.01.045
146. Sies H, Parnham MJ. Potential therapeutic use of ebselen for COVID-19 and other respiratory viral infections. Free Radic Biol Med. 2020;156:107–112. doi:10.1016/j.freeradbiomed.2020.06.032
147. Zhao L, Zhang Z, Li P, Gao Y, Shi Y. Bakuchiol regulates TLR4/MyD88/NF-κB and Keap1/Nrf2/HO-1 pathways to protect against LPS-induced acute lung injury in vitro and in vivo. Naunyn Schmiedebergs Arch Pharmacol. 2024;397(5):3301–3312. doi:10.1007/s00210-023-02813-x
148. Shaghaghi Z, Motieian S, Alvandi M, et al. Ferroptosis Inhibitors as Potential New Therapeutic Targets for Cardiovascular Disease. Mini Rev Med Chem. 2022;22(17):2271–2286. doi:10.2174/1389557522666220218123404
149. Audousset C, McGovern T, Martin JG. Role of Nrf2 in Disease: novel Molecular Mechanisms and Therapeutic Approaches – Pulmonary Disease/Asthma. Front Physiol. 2021;12:727806. doi:10.3389/fphys.2021.727806
150. Vomund S, Schäfer A, Parnham MJ, Brüne B, von Knethen A. Nrf2, the Master Regulator of Anti-Oxidative Responses. Int J Mol Sci. 2017;18(12):2772. doi:10.3390/ijms18122772
151. Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol. 2013;53:401–426. doi:10.1146/annurev-pharmtox-011112-140320
152. Liang W, Greven J, Fragoulis A, et al. Sulforaphane-Dependent Up-Regulation of NRF2 Activity Alleviates Both Systemic Inflammatory Response and Lung Injury After Hemorrhagic Shock/Resuscitation in Mice. Shock. 2022;57(2):221–229. doi:10.1097/SHK.0000000000001859
153. Wei J, Chen G, Shi X, et al. Nrf2 activation protects against intratracheal LPS induced mouse/murine acute respiratory distress syndrome by regulating macrophage polarization. Biochem Biophys Res Commun. 2018;500(3):790–796. doi:10.1016/j.bbrc.2018.04.161
154. Pei X, Zhang XJ, Chen HM. Bardoxolone treatment alleviates lipopolysaccharide (LPS)-induced acute lung injury through suppressing inflammation and oxidative stress regulated by Nrf2 signaling. Biochem Biophys Res Commun. 2019;516(1):270–277. doi:10.1016/j.bbrc.2019.06.006
155. Mathis BJ, Cui T. CDDO and Its Role in Chronic Diseases. Adv Exp Med Biol. 2016;929:291–314. doi:10.1007/978-3-319-41342-6_13. PMID: 27771930.
156. de Oliveira MTP, Coutinho DS, Guterres SS, et al. Resveratrol-Loaded Lipid-Core Nanocapsules Modulate Acute Lung Inflammation and Oxidative Imbalance Induced by LPS in Mice. Pharmaceutics. 2021;13(5):683. doi:10.3390/pharmaceutics13050683
157. Wu Y, Zhang Y, Ge L, et al. RTA408 alleviates lipopolysaccharide-induced acute lung injury via inhibiting Bach1-mediated ferroptosis. Int Immunopharmacol. 2024;142(Pt B):113250. doi:10.1016/j.intimp.2024.113250
158. Holden P, Nair LS. Deferoxamine: an Angiogenic and Antioxidant Molecule for Tissue Regeneration. Tissue Eng Part B Rev. 2019;25(6):461–470. doi:10.1089/ten.TEB.2019.0111
159. Guo Z, Lin J, Sun K, et al. Deferoxamine Alleviates Osteoarthritis by Inhibiting Chondrocyte Ferroptosis and Activating the Nrf2 Pathway. Front Pharmacol. 2022;13:791376. doi:10.3389/fphar.2022.791376
160. Sachan N, Srikrishna S, Patel DK, Singh MP. Deferoxamine Ameliorates Cypermethrin-Induced Iron Accumulation and Associated Alterations. Mol Neurobiol. 2024;61(7):4178–4187. doi:10.1007/s12035-023-03827-5
161. Farmakis D, Porter J, Taher A, et al. 2021 Thalassaemia International Federation Guidelines for the Management of Transfusion-dependent Thalassemia. Hemasphere. 2022;6(8):e732. doi:10.1097/HS9.0000000000000732
162. Chhabra R, Saha A, Chamani A, et al. Iron Pathways and Iron Chelation Approaches in Viral, Microbial, and Fungal Infections. Pharmaceuticals. 2020;13(10):275. doi:10.3390/ph13100275
163. Visseren F, Verkerk MS, van der Bruggen T, et al. Iron chelation and hydroxyl radical scavenging reduce the inflammatory response of endothelial cells after infection with Chlamydia pneumoniae or influenza A. Eur J Clin Invest. 2002;32(1):84–90. doi:10.1046/j.1365-2362.2002.0320s1084.x
164. Zhang C, Xu C, Jing Y, et al. Deferoxamine Induces Autophagy Following Traumatic Brain Injury via TREM2 on Microglia. Mol Neurobiol. 2024;61(7):4649–4662. doi:10.1007/s12035-023-03875-x
165. Jia H, Liu X, Cao Y, et al. Deferoxamine ameliorates neurological dysfunction by inhibiting ferroptosis and neuroinflammation after traumatic brain injury. Brain Res. 2023;1812:148383. doi:10.1016/j.brainres.2023.148383
166. Guo Z, Lin Y, Liu H, et al. Deferoxamine alleviates chondrocyte senescence and osteoarthritis progression by maintaining iron homeostasis. Int Immunopharmacol. 2024;139:112619. doi:10.1016/j.intimp.2024.112619
167. Dalamaga M, Karampela I, Mantzoros CS. Commentary: could iron chelators prove to be useful as an adjunct to COVID-19 Treatment Regimens? Metabolism. 2020;108:154260. doi:10.1016/j.metabol.2020.154260
168. Qi Y, Zhang X, Wu Z, et al. Ferroptosis Regulation by Nutrient Signalling. Nutr Res Rev. 2022;35(2):282–294. doi:10.1017/S0954422421000226
169. Sun Q, Liu D, Cui W, et al. Cholesterol mediated ferroptosis suppression reveals essential roles of Coenzyme Q and squalene. Commun Biol. 2023;6(1):1108. doi:10.1038/s42003-023-05477-8
170. Li D, Zhang G, Wang Z, et al. Idebenone attenuates ferroptosis by inhibiting excessive autophagy via the ROS-AMPK-mTOR pathway to preserve cardiac function after myocardial infarction. Eur J Pharmacol. 2023;943:175569. doi:10.1016/j.ejphar.2023.175569
171. Kitsugi K, Noritake H, Matsumoto M, et al. Simvastatin inhibits hepatic stellate cells activation by regulating the ferroptosis signaling pathway. Biochim Biophys Acta Mol Basis Dis. 2023;1869(7):166750. doi:10.1016/j.bbadis.2023.166750
172. Li W, Liang L, Liu S, Yi H, Zhou Y. FSP1: a key regulator of ferroptosis. Trends Mol Med. 2023;29(9):753–764. doi:10.1016/j.molmed.2023.05.013
173. Zhou J, Zhang L, Yan J, et al. Curcumin Induces Ferroptosis in A549 CD133+ Cells through the GSH-GPX4 and FSP1-CoQ10-NAPH Pathways. Discov Med. 2023;35(176):251–263. doi:10.24976/Discov.Med.202335176.26
174. Jakaria M, Belaidi AA, Bush AI, Ayton S. Vitamin A metabolites inhibit ferroptosis. Biomed Pharmacother. 2023;164:114930. doi:10.1016/j.biopha.2023.114930
175. Shan M, Yu X, Li Y, Fu C, Zhang C. Vitamin B6 Alleviates Lipopolysaccharide-induced Myocardial Injury by Ferroptosis and Apoptosis Regulation. Front Pharmacol. 2021;12:766820. doi:10.3389/fphar.2021.766820
176. Tyml K, Li F, Wilson JX. Septic impairment of capillary blood flow requires nicotinamide adenine dinucleotide phosphate oxidase but not nitric oxide synthase and is rapidly reversed by ascorbate through an endothelial nitric oxide synthase-dependent mechanism. Crit Care Med. 2008;36(8):2355–2362. doi:10.1097/CCM.0b013e31818024f6
177. Li J, Cao Y, Xu J, et al. Vitamin D Improves Cognitive Impairment and Alleviates Ferroptosis via the Nrf2 Signaling Pathway in Aging Mice. Int J Mol Sci. 2023;24(20):15315. doi:10.3390/ijms242015315
178. Chen H, Zhang Y, Miao Y, et al. Vitamin D inhibits ferroptosis and mitigates the kidney injury of prediabetic mice by activating the Klotho/p53 signaling pathway. Apoptosis. 2024;29(9–10):1780–1792. doi:10.1007/s10495-024-01955-4
179. Gao S, Sun C, Kong J. Vitamin D Attenuates Ulcerative Colitis by Inhibiting ACSL4-Mediated Ferroptosis. Nutrients. 2023;15(22):4845. doi:10.3390/nu15224845
180. Luo S, Zeng Y, Chen B, et al. Vitamin E and GPX4 cooperatively protect treg cells from ferroptosis and alleviate intestinal inflammatory damage in necrotizing enterocolitis. Redox Biol. 2024;75:103303. doi:10.1016/j.redox.2024.103303
181. Wang YM, Gong FC, Qi X, et al. Mucin 1 Inhibits Ferroptosis and Sensitizes Vitamin E to Alleviate Sepsis-Induced Acute Lung Injury through GSK3β/Keap1-Nrf2-GPX4 Pathway. Oxid Med Cell Longev. 2022;2022:2405943. doi:10.1155/2022/2405943
182. Mishima E, Wahida A, Seibt T, Conrad M. Diverse biological functions of vitamin K: from coagulation to ferroptosis. Nat Metab. 2023;5(6):924–932. doi:10.1038/s42255-023-00821-y
183. He Q, Lin Y, Chen B, et al. Vitamin K2 ameliorates osteoarthritis by suppressing ferroptosis and extracellular matrix degradation through activation GPX4’s dual functions. Biomed Pharmacother. 2024;175:116697. doi:10.1016/j.biopha.2024.116697
184. Wang Y, Yang W, Liu L, et al. Vitamin K2 (MK-7) attenuates LPS-induced acute lung injury via inhibiting inflammation, apoptosis, and ferroptosis. PLoS One. 2023;18(11):e0294763. doi:10.1371/journal.pone.0294763
185. Mishima E, Ito J, Wu Z, et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature. 2022;608(7924):778–783. doi:10.1038/s41586-022-05022-3
186. Magtanong L, Ko PJ, To M, et al. Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State. Cell Chem Biol. 2019;26(3):420–432.e9. doi:10.1016/j.chembiol.2018.11.016
187. Lei G, Zhuang L, Gan B. mTORC1 and ferroptosis: regulatory mechanisms and therapeutic potential. Bioessays. 2021;43(8):e2100093. doi:10.1002/bies.202100093
188. Zhang Y, Swanda RV, Nie L, et al. mTORC1 couples cyst(e)ine availability with GPX4 protein synthesis and ferroptosis regulation. Nat Commun. 2021;12(1):1589. doi:10.1038/s41467-021-21841-w
189. Zhao Y, Li M, Yao X, et al. HCAR1/MCT1 Regulates Tumor Ferroptosis through the Lactate-Mediated AMPK-SCD1 Activity and Its Therapeutic Implications. Cell Rep. 2020;33(10):108487. doi:10.1016/j.celrep.2020.108487