- 1Department of Orthopaedics, Frist Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- 2Gannan Medical University, 1 Harmony Avenue, Rongjiang New District, Ganzhou, Jiangxi Province, China
- 3Department of Spine Surgery, Yingtan People’s Hospital, Yingtan, Jiangxi, China
- 4Hospital 908, Joint Logistics Support Force, Nanchang, Jiangxi, China
Spinal cord ischemia/reperfusion injury (SCIRI) is a serious disease that leads to the loss of sensory and motor functions and is a common complication after spinal cord injury, spinal cord degeneration or thoracic and abdominal aortic surgery. At present, the spinal cord is mainly protected from ischemic injury through treatment strategies such as hypothermia, surgery and drug assistance, but these intervention measures cannot effectively improve these conditions. SCIRI is a complex process that leads to cell damage and death. Among them, oxidative stress is an important pathological event of ischemia/reperfusion injury. Oxidative stress can initiate multiple inflammatory and apoptotic pathways, triggering a series of destructive events such as inflammatory responses and cell death, further deteriorating the microenvironment at the injured site, and leading to neurological dysfunction. Based on the important role of oxidative stress in SCIRI, we believe that targeted inhibition of oxidative stress responses can effectively reduce secondary injuries caused by trauma, which has a certain positive effect on the rehabilitation and prognosis of patients with SCIRI. This review systematically expounds the spatiotemporal dynamic characteristics of oxidative stress during the SCIRI process and its molecular regulatory network, with a focus on analyzing the multivariate generation mechanism of ROS. To deeply explore the regulatory effects of ROS on pathological processes such as neuronal death, inflammatory response and blood-spinal barrier disruption under SCIRI conditions, as well as its interaction patterns with signaling pathways. In order to form a systematic treatment for SCIRI caused by oxidative stress and promote the recovery of neurological function after injury. This review is helpful for us to understand the effect of oxidative stress on SCIRI and provides a theoretical basis for the treatment of SCIRI based on oxidative stress.
1 Introduction
Spinal cord ischemia/reperfusion injury (SCIRI) is characterized by the interruption of spinal cord blood flow, tissue hypoxia, and a severe impact on the normal metabolism and function of cells. However, after restoring blood perfusion, instead of restoring the spinal cord tissue and function, it triggers a series of complex and harmful pathophysiological reactions, leading to a highly destructive clinical condition that further aggravates spinal cord injury. It often occurs after acute spinal vascular events such as aortic dissection, thoracic and abdominal aortic aneurysm surgery, spinal surgery, and spinal vascular obstruction (Bell et al., 2013; Rong et al., 2022). After SCIRI, it can lead to severe sensory and motor dysfunction, including paralysis and paraplegia, causing patients to have limited movement, loss of autonomous activity ability, and even death (Ling et al., 2021; Smith et al., 2012). This not only causes great difficulties for patients to take care of themselves in daily life but also brings a heavy burden to families and society (O'shea et al., 2017). Spinal cord injury caused by ischemia–reperfusion injury can be divided into ischemic injury and reperfusion injury. Ischemic injury leads to hypoxia and nutrient deprivation, causing insufficient cellular energy and accumulation of metabolic by-products. During reperfusion, oxidative stress, inflammatory responses, and increased production of reactive oxygen species (ROS) may occur, thereby exacerbating secondary injury (Duan et al., 2024). At present, the treatment methods for SCIRI mainly focus on prevention, early identification, and intervention, including intraoperative neurological function monitoring, reducing spinal cord ischemia time, increasing spinal cord perfusion pressure, and enhancing the tolerance of the spinal cord to ischemia through hypothermia, etc., attempting to protect the spinal cord from ischemic injury, reduce the degree of injury, and promote neurological function recovery (Wynn and Acher, 2014; Hnath et al., 2008; Khachatryan et al., 2022). However, the effects of these intervention measures are often unsatisfactory, and it is difficult to fundamentally solve the problem of nerve injury (Lemaire et al., 2012).Therefore, in-depth exploration of the pathogenesis of SCIRI and the search for new treatment strategies have tremendous clinical significance. The pathological cascade of SCIRI involves the synergistic effect of multi-dimensional pathological mechanisms. Its development process covers key pathological links such as inflammatory cascade activation, programmed cell death (including apoptosis and autophagy), disruption of the integrity of the blood-spinal barrier (BSCB), imbalance of redox homeostasis, intracellular calcium ion homeostasis disorder, and excitatory amino acid neurotoxicity (Liu et al., 2018; Zhao et al., 2018).Particularly worthy of attention is that oxidative stress, as the core regulatory hub of this pathological network, plays a key driving role in SCIRI by mediating the imbalance of free radical metabolism (Xu et al., 2023). Specifically, the outbreak of ROS triggered by ischemia–reperfusion can induce a lipid peroxidation chain reaction. Through multiple mechanisms such as activating calcium-dependent proteases, promoting the release of pro-inflammatory factors, and enhancing the sensitivity of the mitochondrial apoptotic pathway, it eventually leads to the disintegration of neuronal structure and decompensation of spinal cord conduction function, resulting in irreversible neurological deficits (Yu et al., 2016; Al Mamun et al., 2021; He et al., 2021). Based on this, the microenvironment regulation of key nodes of oxidative stress (such as eliminating excessive ROS and repairing the antioxidant defense system) has been established as an important neuroprotective strategy for improving the prognosis of SCIRI, and its intervention effect directly affects the repair process and neurological functional outcome of the injured tissue (Sun et al., 2021).
Oxidative stress refers to the imbalance between the oxidation and antioxidant systems in the body when the body is exposed to various harmful stimuli, resulting in the excessive generation of ROS, such as superoxide anions, hydroxyl radicals, and hydrogen peroxide, which exceeds the body’s own antioxidant defense capacity (Pizzino et al., 2017; Boyle et al., 1999). Under physiological conditions, ROS as an inevitable by-product of cellular aerobic metabolism, maintains a precise dynamic balance with the endogenous antioxidant system (Farías et al., 2017). However, during ischemia–reperfusion, bleeding and fluid resuscitation can promote excessive production of ROS, leading to the decompensation of the antioxidant defense system and ultimately inducing a cascade of oxidative stress responses (Yu et al., 2019). The pathological process of SCIRI shows significant biphasic characteristics: During the acute ischemic period, neuronal overexcitation and glutaminotropic excitotoxicity activate the inflammatory cascade reaction, trigger the initial oxidative stress event, and promote the excessive generation of ROS. In the subsequent reperfusion stage, with the sudden increase in oxygen partial pressure, the dysfunction of the mitochondrial electron transport chain will directly induce the explosive generation of oxygen free radicals (Goldhaber and Weiss, 1992). This persistent oxidative damage can trigger various cellular structural damages, including but not limited to lipid peroxidation chain reactions, DNA oxidative damage, and pro-inflammatory factor storms, ultimately leading to cell death and irreversible neuronal damage (Li et al., 2019; Yu et al., 2023; Zhang et al., 2024). Recent studies have confirmed that oxygen free radicals produced during the acute phase of SCIRI are a key driver of secondary neuronal necrosis. They can aggravate the injury of spinal cord tissue through pathways such as activating inflammatory responses and inducing apoptosis, forming a vicious cycle that continuously amplifies the injury effect (Hall, 2011). Preclinical studies have shown that antioxidant therapy has demonstrated in animal models the preliminary evidence of effectively improving spinal cord nerve function by neutralizing excessive ROS, inhibiting neuronal apoptosis, regulating calcium homeostasis imbalance and alleviating inflammatory cascade reactions (Hall, 2011; Zhou et al., 2018; Han et al., 2020; Feng et al., 2021). However, it should be emphasized that these findings are limited to laboratory research. Current clinical data show that antioxidant therapy has not yet confirmed a clear neurorepair effect in human trials. Furthermore, the formation of oxidative stress after ischemia–reperfusion and its impact on spinal cord tissue injury involve multiple complex mechanisms. In view of the complexity of this mechanism, targeted regulation of oxidative stress after SCIRI has potential research value.
This review systematically expounds the spatiotemporal dynamic characteristics of oxidative stress during the SCIRI process and its molecular regulatory network, and analyzes the multiple generation mechanisms of ROS mediated by SCIRI (including mitochondrial electron transport chain leakage, Phosphoramide adenine dinucleotide (NADPH) oxidase activation, xanthine oxidase system imbalance, etc.), and explores in depth the interaction mode between ROS and Nrf2/ARE, NF-κB, MAPK and other signaling pathways under SCIRI pathological conditions, as well as their regulatory effects on pathological processes such as neuronal death, inflammatory response, and blood–brain barrier disruption. By integrating various intervention measures targeting the regulation of redox balance in recent years, it aims to form a systematic treatment for SCIRI caused by oxidative stress and promote neurological function recovery after injury. This review is helpful for us to understand the effect of oxidative stress on SCIRI and provides a theoretical basis for the treatment of SCIRI based on oxidative stress.
2 Literature retrieval strategy
On January 20, 2025, the first author initiated a search of multiple databases, covering literature published from March 1998 to January 2025. The accessed databases include China National Knowledge Infrastructure, Wanfang Database and PubMed. The search strategy uses keywords such as “spinal cord ischemia–reperfusion injury,” “mitochondria,” “activated oxygen,” “oxidative stress” and “inflammation” to establish selection criteria. Various combinations of these keywords were used during the retrieval process. The retrieved literature mainly includes original research papers, review papers, comments, case reports and meta-analyses. The inclusion criteria include studies related to oxidative stress and SCIRI, which are highly original, have clear conclusions, and have been published in well-known journals within the past 20 years. Exclusion criteria include studies that are irrelevant to the topic of this review or studies with repetitive content. After the initial search, the relevance to this study was determined by reviewing the title and abstract, and then the full text was read comprehensively to evaluate the central topic of the selected study.
3 Sources of ROS in SCIRI
Under normal physiological conditions, ROS is constantly produced in the body, and there is also a complete antioxidant defense system to maintain the dynamic balance of ROS. ROS is a type of oxygen-containing molecules with high chemical reactivity, mainly including singlet oxygen, superoxide anions (O2 -), hydrogen peroxide (H2O2), and hydroxyl radicals (OH−) (Rahal et al., 2014; Giorgio et al., 2007). Oxidative stress is a series of adaptive responses caused by the imbalance between ROS levels and the antioxidant system. These responses lead to the accumulation of excessive free radicals and subsequent cytotoxic effects (Sies and Jones, 2020). ROS plays a crucial role in the occurrence and development of ischemia–reperfusion (I/R) injury (Halladin, 2015). After ischemia–reperfusion, ROS in the spinal cord is mainly produced by enzymatic reactions such as mitochondrial respiratory chain, xanthine oxidase, NADPH oxidase.
Mitochondria are the centers of intracellular energy metabolism and are also regarded as the main sites where ROS is produced (Quinlan et al., 2013). Under physiological conditions, the mitochondrial respiratory chain achieves energy conversion through a precisely coupled oxidative phosphorylation process. Specifically, the reduction equivalent released by the substrate dehydrogenation reaction is gradually transferred through the electron transport system composed of complex I, complex III and complex IV. Eventually, under the catalysis of complex IV, molecular oxygen undergoes electron reduction to form water molecules. In this cascade reaction process, the proton gradient across the inner mitochondrial membrane is gradually formed through the proton pump function established by complexes I, III, and IV, and drives the ATP synthase (complex V) to catalyze the phosphorylation of ADP to generate ATP, achieving an accurate coupling of redox reactions and energy storage (Zotta et al., 2024). However, during this process, some electrons escape from the respiratory chain, resulting in incomplete reduction of molecular oxygen, namely the generation of ROS (Li et al., 2022). The creation of ROS happens through oxidative phosphorylation, which is part of aerobic respiration in the mitochondrial electron transport chain (ETC). Mitochondrial ETC generates superoxides and H₂O₂ by regulating the proton gradient within the mitochondria (Cadenas, 2018). Electrons from complex 1 can directly react with oxygen to generate O₂−, rather than being reduced to H₂O (Brand, 2010). O2− can be spontaneously or catalyzed by superoxide dismutase (SOD) to be converted into H₂O₂ and O₂ (Breton-Romero and Lamas, 2014). And H₂O₂ oxidizes Fe2+ to Fe3+ through the Fenton reaction to generate hydroxyl radicals (Kevin et al., 2005). However, under ischemic and hypoxic conditions, the electron transport in the mitochondrial respiratory chain is blocked. Some electron carriers in the electron transport chain, such as coenzyme Q, will directly transfer electrons to oxygen, causing oxygen molecules to undergo single-electron reduction and thereby generating a large amount of O2−. In addition, during the ischemic period, intracellular ATP production is significantly reduced, and the functions of ATP-dependent ion pumps are impaired, such as sodium-potassium pumps and calcium pumps, which cannot work normally. This leads to a large accumulation of Na- and Ca2+ within cells and an imbalance in the intracellular environment. The increase of intracellular calcium ion concentration will activate a series of enzymes, such as phospholipase A2, protease, etc. The activation of phospholipase A2 can decompose phospholipids on the cell membrane, generating metabolic products such as arachidonic acid, which further participate in inflammatory responses and the generation of ROS. The activation of proteases can degrade intracellular proteins, disrupt the normal structure and function of cells, and may also indirectly promote the production of ROS (Alizadeh et al., 2019).
During the reperfusion period, with the re-supply of oxygen, the xanthine oxidase system is activated, which is an important reason for the further intensification of oxidative stress (Neri et al., 2015). The xanthine oxidase system consists of two interconvertible forms: xanthine dehydrogenase (XDH) preferentially uses NAD+ as the electron acceptor, while xanthine oxidase (XO) uses O2 as the terminal electron acceptor (Granger and Kvietys, 2015). During the ischemic period, due to the reduced production of ATP, a large amount of hypoxanthine, an energy metabolite within cells, accumulates. During reperfusion, the supply of oxygen enables xanthine dehydrogenase to be rapidly converted into xanthine oxidase. Xanthine oxidase uses hypoxanthine and xanthine as substrates. With the participation of oxygen, it oxidizes them to uric acid and simultaneously generates a large amount of superoxide anions (Mccord, 1985; Reimer and Jennings, 1985). This process leads to a sharp increase in the generation of ROS, exceeding the antioxidant defense capacity of the organism itself.
Phosphoramide adenine dinucleotide oxidase is a transmembrane protein complex and one of the important ROS-producing enzymes in the body. During ischemia–reperfusion, inflammatory cells such as neutrophils and macrophages are activated and, attracted by chemokines, gather at the site of ischemia–reperfusion injury (Taoka et al., 1997; Trivedi et al., 2006). Meanwhile, after being activated, these cells can activate NADPH oxidase. In the activated state, it uses NADPH as an electron donor to reduce oxygen to ROS, such as O₂−. These ROS can attack biological macromolecules such as cell membranes, proteins, and nucleic acids, triggering lipid peroxidation, protein oxidative modification, and DNA damage. This further leads to the destruction of cell structure and function (Khayrullina et al., 2015; Busquets-Cortés et al., 2018; Figure 1).

Figure 1. Schematic diagram of the generation mechanism of ROS during the SCIRI process. ROS is mainly produced through three pathways: mitochondria, xanthine oxidase, and NADPH oxidase. Oxidative stress injury occurs when the release of ROS exceeds the clearance capacity. Elevated ROS levels can induce lipid peroxidation, protein and DNA damage, and cross-interference in inflammatory responses, leading to further deterioration of SCIRI.
4 The impact of oxidative stress on SCIRI
After SCIRI, a large amount of ROS can be produced, causing disorders in the body’s oxidation/antioxidant system, thereby leading to oxidative stress and oxidative damage (Gu et al., 2020; Khattab et al., 2022). During the spinal cord ischemia stage, a large amount of ROS is induced to be produced, activating macrophages to release pro-inflammatory mediators such as TNF-α and IL-1β, triggering neutrophil infiltration and inflammasome activation. Inflammatory factors induce the dissociation of ZO-1 protein from the tight junction complex, simultaneously up-regulate the expression of MMPs, directly degrade the collagen components of the basement membrane, and disrupt the integrity of BSCB. The disruption of BSCB triggers tissue edema caused by plasma extravasation, compressing microcirculation, and secondary infiltration of peripheral immune cells. This forms a cascade amplification effect of “inflammation-BSCB leakage-reinflammation” (Qu et al., 2009). Furthermore, ROS also activates various enzymes, including phospholipase A, thereby destroying the integrity and permeability of the cell membrane of the nervous system, leading to cell death and reperfusion injury (Chen et al., 2011; Hall et al., 2016). This three-dimensional pathological network spanning oxidative stress, inflammatory response, and barrier disruption provides a molecular basis for the irreversible nerve injury of SCIRI. In this section, we mainly discussed and described the negative effects of ROS during ischemia–reperfusion by regulating inflammatory responses, the blood-ridge barrier, mitochondrial damage, and programmed cell death (Figure 2).

Figure 2. The effects of oxidative stress on inflammatory response, blood-spinal barrier, mitochondrial damage and cell death during SCIRI.
4.1 Promote inflammatory responses
The inflammatory response is a defense response against exogenous and/or endogenous signals, but persistent inflammation has adverse effects on the body (Iantomasi et al., 2023). It is known that the inflammatory response plays an important role after ischemia and leads to spinal cord injury (Matsumoto et al., 2003). It was found through single-cell sequencing technology that inflammation-related immune cells infiltrated in SCIRI, including macrophages and neutrophils (Kim et al., 2019). These inflammatory cells produce many pro-inflammatory mediators, such as IL-1β, TNF-α, and IL-6, etc. These cytokines are important mediators of ischemic inflammatory responses in the spinal cord. They recruit peripheral inflammatory cells into the spinal cord and cause delayed neuronal death (Altinors, 2009; Ilhan et al., 2004). Oxidative stress is involved in the occurrence and development process of SCIRI and is also one of the important factors inducing inflammation (Fakhri et al., 2022). Inflammation in SCIRI can lead to apoptosis of neuronal cells and dysfunction of the blood–brain barrier, thereby accelerating the pathological process of SCIRI (Fu et al., 2018). The increase in ROS production can lead to protein oxidation, and protein oxidation is converted into the release of inflammatory signaling molecules, such as peroxidase 2 (PRDX2) (Berlett and Stadtman, 1997; Salzano et al., 2014). PRDX2 can also act as a redox dependent inflammatory mediator to activate macrophages to produce and release TNF- α, thereby altering the REDOX state of cell surface receptors and inducing inflammatory responses (Salzano et al., 2014). It is notable that the inflammatory response caused by oxidative stress can, in turn, enhance oxidative stress (Li et al., 2019). Mitochondria stimulated by TNF can increase ROS, leading to apoptosis and necrotic apoptosis, while ROS can promote the generation of TNF. A positive feedback loop is formed between the two (Blaser et al., 2016). The increase in ROS production at the injury site leads to the release of inflammatory factors and adhesion molecules, and mediates the inflammatory response by activating the NF-κB signaling pathway (Morgan and Liu, 2011). This circuit was also found during the process of spinal cord ischemia–reperfusion (Gökce et al., 2016). This indicates that during the process of spinal cord injury, inflammation and oxidative stress promote each other, eventually leading to a vicious cycle.
4.2 Damage the blood-spinal barrier
The blood-spinal cord barrier (BSCB) is usually regarded as an extension of the blood–brain barrier. The BSCB is composed of capillary endothelial cells and their accompanying basement membrane, pericytes, and astrocyte endopods. Its function is to regulate blood flow in cerebrospinal fluid and control the inflow and outflow of nutrients, energy storage, metabolites, and toxins within the neurovascular unit (Mcconnell et al., 2017; Li et al., 2017). Some studies reported that increased levels of superoxide contribute to BSCB endothelial dysfunction (Lochhead et al., 2010). Oxidative stress causes the release of several pro-inflammatory cytokines, which break down the extracellular matrix and damage proteins that help keep the blood–brain barrier intact, leading to its failure and changes in blood flow (Allen and Bayraktutan, 2009; Janyou et al., 2017). Furthermore, oxidative stress can also activate various redox sensitive transcription factors and participate in and accelerate the inflammatory response, leading to BSCB damage (Kim et al., 2022). Inflammatory cytokines are one of the most pivotal factors for BSCB disruption in SCIRI. They cause ZO-1 to separate from the cytoskeleton and increase the levels of MMPs and TNFα, which are often linked to higher BSCB permeability. Nevertheless, BSCB disruption in turn exacerbates inflammatory reactions, resulting in irreversible spinal cord damage (Ding et al., 2010).
4.3 Mitochondrial damage
Mitochondria, as the main target of intracellular oxidative stress, are highly susceptible to it in terms of their structure and function. First of all, ROS can directly attack the mitochondrial inner membrane system. Excessive accumulation of ROS can induce structural disorders (swelling or rupture) of the mitochondrial cristae and promote abnormal opening of the mitochondrial permeability transition pore (mPTP). The continuous opening of mPTP can cause swelling of the mitochondrial matrix, which in turn leads to mechanical rupture of the outer membrane and causes membrane potential (ΔΨm) collapse. Meanwhile, pro-apoptotic factors such as cytochrome C (Cyt C) are released into the cytoplasm, activating the Caspase-9/3 cascade reaction and ultimately initiating programmed cell death (Holzerová and Prokisch, 2015; Yang et al., 2015; Skulachev, 1998). Secondly, ROS interferes with the mitochondrial dynamic balance. ROS disrupts the homeostasis regulation of mitochondrial division and fusion by oxidizing and modifying fissile proteins or fusion proteins, resulting in excessive fragmentation of the mitochondrial network. Fragmented mitochondria, due to the increase in surface area and the disorder of electron transport chains, further intensify the abnormal generation of ROS, thereby forming a vicious cycle and amplifying oxidative stress (Bernardi et al., 2015). Furthermore, damaged mitochondria amplify apoptotic signals through multiple pathways. In addition to releasing Cyt C, the rupture of its outer membrane can also lead to the outflow of pro-apoptotic proteins. By inhibiting apoptotic inhibitory proteins or directly cutting DNA, it synergistically enhances the caspase-dependent and non-dependent cell death pathways (Anne Stetler et al., 2013).
4.4 Cell death
Cell death is a highly conserved biological process, which not only participates in the morphogenesis and developmental regulation of organisms, but also plays an important role in various pathophysiological processes. Different cell death patterns exhibit unique morphological characteristics and molecular mechanisms, and mediate the pathogenesis of diseases through different pathways (Galluzzi et al., 2018). Apoptosis is a process of cell death that occurs through the activation of caspase cascade apoptosis under the strict regulation of apoptosis genes. It is characterized by cell contraction, nuclear condensation and chromatin condensation, and eventually forms apoptotic bodies (Tang et al., 2019; Maiuri et al., 2007). Pyroptosis is a type of pro-inflammatory cell death that depends on the activation of caspase-1 and the formation of plasma membrane pores by gasdermin-D (GSDMD) (Bergsbaken et al., 2009). Ferroptosis is a unique non-apoptotic regulated cell death, whose main characteristics are abnormal iron metabolism and lipid peroxidation within cells (Dixon et al., 2012). Necroptosis is a programmed form of cell death that does not rely on caspase (Zhu et al., 2022). In addition, other cell death patterns also exhibit unique mechanisms: Necrosis, as a non-programmed passive death, is often triggered by acute injury, accompanied by swelling of organelles and rupture of the plasma membrane (D'arcy, 2019). Parthanatos is a form of death caused by the excessive activation of PARP-1 activated by DNA damage, characterized by the release of mitochondrial apoptosis-inducing factors and chromatin dispersion (Huang et al., 2022). Autophagia mediates through excessive formation of autophagosomes and can induce cellular degradation under specific conditions (Sepehr et al., 2020). Entosis is a form of invasive cell death. Through cytoskeletal remodeling, target cells are phagocytosed by adjacent cells, and this process has special pathological significance in the tumor microenvironment (Mlynarczuk-Bialy et al., 2020). These diverse pathways of death constitute a complex regulatory network, which interact to jointly maintain tissue homeostasis and affect disease outcomes.
A large number of studies have confirmed that ROS plays an important role in the regulation of cell death, and it participates in the molecular cascade reactions of different death patterns (apoptosis, necrotizing apoptosis, pyroptosis, ferroptosis, and autophagy) through multiple pathways (Orrenius et al., 2007; Ryter et al., 2007; Figure 3). As a classic caspase-dependent programmed death, the initiation and execution of apoptosis are mainly regulated by caspase family proteases (Green, 1998). This process can be activated through two main pathways: exogenous and endogenous, and ROS, as a key regulatory hub, has an activation effect on both pathways (Mirzaei et al., 2021). In the endogenous apoptotic pathway, ROS induces the opening of mitochondrial mPTP by directly oxidizing the phospholipid components of the mitochondrial inner membrane (such as cardiolipin), promoting the release of Cyt C into the cytoplasm. Cyt C in the cytoplasm, together with apoptotic protease activator factor-1 (Apaf-1) and procaspase-9, jointly forms apoptotic body complexes. Through cascade activation of caspase-9 and its downstream effector proteins (such as caspase-3/7), it ultimately triggers apoptosis (Kalkavan and Green, 2018). In the exogenous apoptotic pathway, ROS promotes the formation of a death-inducing signal complex between adaptor protein FADD and procaspase-8/10 by activating death receptors such as TNF-R1, TRAIL-R1/2 and Fas. This complex activates caspase-8/10 and further activates downstream effector caspases (such as caspase-3/6/7), thereby initiating the apoptotic program and triggering apoptosis (Li et al., 1997). Necrotic apoptosis is a form of programmed necrosis regulated by genetic programming, and its core executive mechanism relies on the activation of the receptor-interacting protein kinase 1 (RIPK1) -RIPK3-MLKL signaling axis (Degterev et al., 2005; Zhou et al., 2022). This process can be initiated by various exogenous stimuli, including interferon, tumor necrosis factor and Toll-like ligands, etc. These stimuli eventually trigger the necrotic apoptosis cascade by activating the downstream death domain signal transduction network (Zhang et al., 2017). It is notable that ROS has been proven to mediate the execution of cell necrosis. Under oxidative stress conditions, the accumulated ROS in cells activates the RIPK1/RIPK3 complex, promotes the phosphorylation and oligomerization of mixed lineage kinase domain-like proteins, and enables their translocation to the cell membrane to form ion channels, thereby initiating the classic RIPK1-RIPK3-MLKL pathin-dependent necrotic apoptosis. Furthermore, under specific pathological conditions, ROS can also promote the release of apoptosis-inducing factors from mitochondria by activating polyadenosine diphosphate ribose polymerase, thereby triggering necrotizing apoptosis mediated by the non-classical RIPK1/RIPK3-PARP-AIF signaling axis (Mármol et al., 2017; Han et al., 2018). Pyroptosis is a caspase-1-dependent type of pro-inflammatory cell death, which differs from other forms of programmed cell death in terms of morphology and biochemical characteristics (Liu and Sun, 2019; Galluzzi et al., 2015; Fink and Cookson, 2006). Pyroptosis is divided into the classical inflammasome pathway mediated by caspase-1 and the non-classical pathways mediated by caspase 4, 5, and 11 (Lacey et al., 2018). Mitochondrial reactive oxygen species (mtROS) regulate pyroptosis by influencing lipid metabolism and inflammation. mtDNA oxidized by mtROS is released into the cytoplasm along with mitochondrial membrane damage, binding and triggering the activation of NLRP3 inflammatosomes, thereby inducing pyroptosis (Svensson et al., 2022; Zhong et al., 2018). It is reported that ROS is a common upstream mechanism in the activation process of NLRP3 inflammasome. ROS activates NLRP3 through the redox sensor thioredoxin interacting protein and triggers pyroptosis through the ROS/NLRP3-mediated caspase-1 pathway (Qiu et al., 2019; He et al., 2016). Ferroptosis is a form of cell death caused by the accumulation of iron-dependent lipid peroxides, and its main characteristics are abnormal iron metabolism and lipid peroxidation within cells (Dixon et al., 2012). During the SCIRI process, the red blood cells at the bleeding site rupture and release a large amount of free iron. Catalyzed by heme oxygenase-1 (HO-1), active iron ions are generated, resulting in local iron overload. Meanwhile, the activation of hypoxia-inducible factor (HIF-1α) promotes the outbreak of ROS and intensifies the excitotoxicity of glutamate by inhibiting the function of the glutamate-cystine reverse transporter. Multiple factors synergistically drive the occurrence of ferroptosis (Su et al., 2019). ROS weakens the cellular antioxidant defense system by consuming glutathione (GSH) and inhibiting the activity of glutathione peroxidase 4 (GPX4), leading to irreversible accumulation of lipid peroxides and ultimately causing ferroptosis (Yang et al., 2020). Autophagy is a highly conserved cellular self-degradation mechanism in eukaryotes. It degrades damaged or redundant intracellular components (such as abnormal proteins and senescent organelles) through lysosome-dependent pathways to maintain intracellular homeostasis and respond to stressful environments (Liu et al., 2023). In SCIRI, there exists a dynamic and bidirectional regulatory relationship between oxidative stress and autophagy, jointly influencing the fate choices of nerve cells (Yuan et al., 2023). Hypoxia during the ischemic period leads to dysfunction of the mitochondrial electron transport chain, triggering explosive accumulation of ROS. The recovery of oxygen supply during the reperfusion period further aggravates oxidative stress through pathways such as xanthine oxidase (Klionsky and Emr, 2000). Excessive ROS oxidizes and modifies key autophagy proteins (such as thiol oxidation of Beclin-1) and lysosomal membrane lipids, leading to autophagic flow arrest (such as autophagosome-lysosomal fusion obstruction), promoting abnormal accumulation of autophagic substrates and inducing autophagy-related death (Xie et al., 2017). The above studies indicate that ROS plays an important regulatory role in the cell death of SCIRI.
5 Signaling pathways involved in oxidative stress related SCIRI
Oxidative stress is an important pathological process in SCIRI and is regulated by multiple pathways, including signaling pathways such as Nrf2, NF-κB, and MAPKs. This chapter will introduce the role of these signaling pathways in oxidative stress in SCIRI (Figure 4).

Figure 4. The oxidative stress process in SCIRI may involve signaling pathways and interaction mechanisms. After the MAPK pathway is activated, it promotes the dissociation of Nrf2 and Keap1. Nrf2 translocates to the nucleus, heterodimerizes with sMaf in the nucleus, and binds to an enhancer sequence called ARE. This, in turn, stimulates the expression of HO-1, NQO1, CAT, and SOD, thereby inhibiting oxidative stress through feedback. Nrf2 and NF-κB can influence each other and jointly regulate the pathological process.
5.1 Nrf2 pathway
Nuclear factor erythrocyte 2-related factor 2 (Nrf2) is a redox sensitive transcriptional regulatory factor, and its core function is to regulate the transcriptional activation of downstream genes by binding to antioxidant response elements (ARE) (Ma, 2013). Under normal physiological conditions, Kelch-like epichlorohydrin-1 (Keap1) continuously promotes the proteasome degradation of Nrf2 through the ubiquitination pathway and maintains its low expression level. When the imbalance of the body’s oxidation-antioxidant homeostasis leads to abnormal accumulation of ROS, the negative regulatory effect of Keap1 is released, thereby inhibiting the degradation process of Nrf2 (Lu et al., 2016). Under oxidative stress, Nrf2 dissociates from Keap1 and undergoes nuclear translocation. By forming heterodimer complexes with the small Maf protein in the nucleus, it specifically recognizes the enhancer region regulated by ARE. This further initiates the transcriptional activation of antioxidant/drug metabolism enzyme genes including HO-1, SOD, quinone oxidoreductase (NQO1), and glutathione peroxidase (GPX), etc. (Krajka-KuŹniak et al., 2017; Hu et al., 2016). Through the above cascade reaction, cells were able to enhance the activity of the antioxidant enzyme system, eliminate excessive ROS, and antagonize oxidative damage (Wang et al., 2012). Through the above cascade reaction, cells are able to enhance the activity of the antioxidant enzyme system, eliminate excessive ROS and antagonize oxidative damage. It is notable that HO-1, as an endogenous protective enzyme with consistently high expression in spinal cord injury, can persist in SCIRI and significantly alleviate oxidative stress injury by stabilizing mitochondrial function (Lin et al., 2016). Furthermore, after activation of Nrf2, it can also up-regulate the expression of the downstream target gene HO-1, and counteract oxidative stress by removing the strong oxidant heme and increasing the levels of endogenous antioxidants carbon monoxide and bilirubin (Prestera et al., 1995). Zhang et al. treated the rat SCIRI model with ginsenoside Rg1. They found that Nrf2 was activated. The activation of Nrf2 increased the enzymatic activities of SOD and GPx by inducing the expression of HO-1, and simultaneously inhibited the generation of lipid peroxidation marker MDA, thereby exerting an antioxidant effect (Zhang et al., 2022). It is particularly worth noting that NQO1 regulated by Nrf2, specifically catalyzes the conversion of quinone substances to hydroquinone through reduction reactions, effectively blocking the process of ROS generation by quinone compounds in the redox cycle. This mechanism plays a key role in alleviating secondary oxidative damage (Jung and Kwak, 2010). In addition, Nrf2 can also exert antioxidant effects by promoting the regeneration of oxidative cofactors and proteins. In the SCIRI rat model, melatonin promotes the reduction of glutathione disulfide by glutathione reductase by activating Nrf2 (Kryl'skii et al., 2019). Meanwhile, Nrf2 regulates the production of mitochondrial ROS by enhancing the biosynthesis of glutathione and the activity of glutathione reductase, up-regulating the enzymes in the pentose phosphate pathway, and regulating the expression level of mitochondrial antioxidant enzymes, thereby protecting mitochondria from oxidative damage (Ebrahimy et al., 2022; Chen et al., 2022). Furthermore, Nrf2 can also regulate the expression of HO-1 and NADPH quinone oxidoreductase-1 genes by directly acting on the ARE element, thereby inducing the expression of stress response proteins (Li et al., 2019; Figure 4a).
5.2 NF-kB pathway
Nuclear factor-κB (NF-κB) is a complex protein system composed of transcription factors and is an important nuclear transcription factor within cells. The NF- κB family consists of five members: RelA (p65), RelB, c-Rel, precursor proteins NF- κB1 (p105) and NF- κB2 (p100) (Oeckinghaus et al., 2011). Studies have confirmed that NF-κB signal transduction is activated after SCIRI (Jin et al., 2021). Oxidative stress is a powerful stimulating factor for the activation of NF-κB. The activated NF-κB is involved in the regulation of various pathological processes, including inflammatory responses, oxidative stress and apoptosis, etc. The research found that after SCIRI, increased levels of ROS and malondialdehyde (MDA), decreased activity of SOD, and accumulation of inflammatory factors (IL-1β and IL-6) and TNF-α were observed (Bao et al., 2016). The excessive ROS generated can directly activate NF-κB or indirectly activate NF-κB through redox factor-1 or other pathways, such as regulating the activity of IκB kinase or affecting the affinity between NF-κB and IκB, thereby further promoting the release of inflammatory factors and ROS (Nguyen et al., 2019; Angkeow et al., 2002; El Assar et al., 2013; Figure 4b). Furthermore, NF-κB is also a redox regulated transcription factor (Karin et al., 2004). Wang et al. used stearic acid chitosan nanoparticles loaded with sesamol to intervene in LPS-induced spinal motor neuron cells and found that NF-κB was significantly inhibited, leading to increased SOD and GPx activity and reduced release of ROS and inflammatory factors, thereby weakening oxidative stress and inflammatory damage (Wang et al., 2021).
5.3 MAPK pathway
Mitogen-activated protein kinase (MAPK) family is a conserved serine/threonine protein kinase family, consisting of extracellular signal-regulated kinase (ERK), p38 and c-Jun NH 2-terminal kinase (JNK) (Liu et al., 2020; Kanno et al., 2009). Each MAPK signal contains at least three parts: MAPK kinase kinase (MAP3K), MAPK kinase (MAP2K), and MAPK (Kim and Choi, 2010). The MAPK pathway is an important transmembrane signal transduction pathway, including MAPKs, ERK1/2, JNK, P38, etc. These pathways are relatively sensitive to ROS stimulation (Son et al., 2011). It is reported that the activation of MAPK is crucial for the production of various inflammatory cytokines and the regulation of oxidative stress (Bachstetter et al., 2011). During the pathological process of SCIRI, the level of ROS significantly increased (Ahuja et al., 2017). Excessive production of ROS can trigger oxidative stress, which then activates the p38 MAPK signaling pathway, thereby inducing oxidative stress and inflammatory responses, and leading to cell death (Bode et al., 2012; Huang et al., 2018). Under the stimulation of oxidative stress or other damage effects, the body produces cytokines such as interleukin and TNF-α. These cytokines can activate the MAPK signaling pathway and promote the nuclear translocation of Nrf2, thereby initiating antioxidant effects (Han et al., 2016; Zhao et al., 2016; Chi et al., 2015; Figure 4c).
6 Remote ischemic preconditioning after SCIRI
It is reported that remote ischemic preconditioning (RIPC) is an effective method for alleviating ischemia–reperfusion injury (Liang et al., 2019). RIPC is a method to protect distant target tissues/organs from subsequent fatal ischemic injury through intermittent ischemia–reperfusion of tissues/organs (Hausenloy and Yellon, 2011; Yu et al., 2016). In clinical practice, RIPC is usually achieved by applying periodic inflation-deflation to the upper or lower limbs of patients through a standard blood pressure cuff (Lee et al., 2023). The specific operation is to wrap the blood pressure cuff around one or both limbs (such as the upper arm or thigh), inflate until the pressure is higher than the systolic pressure, continuously block the blood flow for 5 min, then release the air for 5 min to resume perfusion. This process is repeated for 3 to 4 cycles (Lee et al., 2023). Preclinical data and clinical trials have shown that RIPC therapy can alleviate myocardial injury and improve the short-term prognosis of patients undergoing coronary artery bypass grafting (Kleinbongard et al., 2018). Meanwhile, RIPC can promote the release of molecules with damage-related molecular patterns, thereby activating the natural defense system and protecting the kidneys from subsequent inflammation and chemical damage (Gassanov et al., 2014). Some studies have shown that RIPC can protect the spinal cord from I/R injury (Yunoki et al., 2017). In the rat brain I/R injury model, researchers found that distal ischemic postconditioning could reduce the permeability of the BSCB and brain edema after cerebral ischemia, thereby reducing the infarct area and improving neurological outcomes (Li et al., 2015; Ren et al., 2011). This method has the characteristics of non-invasiveness, simplicity, convenience, repeatability and cost-effectiveness. Animal experiments have shown that RIPC can protect the spinal cord from I/R injury. Dong et al. found in a rabbit experiment that achieving RIPC through bilateral femoral artery occlusion (10 min of ischemia/10 min of reperfusion, 2 cycles) before aortic occlusion of spinal cord ischemia in rabbits could significantly improve neurological prognosis and alleviate spinal cord tissue damage. They also found that in the rat model, limb RIPC-induced spinal cord ischemic tolerance was achieved through tertiary circulation of the right femoral artery and 3-min ischemia/3-min reperfusion (Dong et al., 2010; Su et al., 2009). Previous studies have shown that RIPC can reduce oxidative stress during cardiopulmonary bypass, thereby alleviating nerve injury (Arvola et al., 2016; Bystrom et al., 2017). In the oxygen–glucose deprivation experiment simulating cerebral ischemia, RIPC exerts anti-apoptotic and antioxidant effects in neural stem cells, thereby protecting human neural stem cells from cell death (Motomura et al., 2017). Meanwhile, Gu et al. also found that RIPC could promote the recovery of neurological function in SCIRI mice, inhibit spinal cord oxidative stress, and reduce neuronal damage (Gu et al., 2023). These studies show that RIPC can protect the spinal cord from I/R injury.
7 Therapy target for oxidative stress related SCIRI
During I/R injury, a large amount of ROS is generated around the injury site, which can cause excessive oxidative stress, leading to neuronal death and accelerating the development of SCIRI. It is notable that reducing oxidative stress may be a promising approach for the treatment of SCIRI (Yu et al., 2019) (Table 1).
7.1 Native compound
In recent years, the treatment of spinal cord injury using monomer compounds of traditional Chinese medicine has received extensive attention. Curcumin is a known active component in turmeric. Due to its role in scavenging free radicals, it has been widely studied in I/R traumatic diseases (Mokhtari-Zaer et al., 2019; Machado et al., 2022). For example, renal ischemia/reperfusion injury (Machado et al., 2022), cardiac I/R injury (Mokhtari-Zaer et al., 2019). In studies related to SCIRI, it was found that curcumin can alleviate neuronal apoptosis and oxidative stress induced by OGD/R by inhibiting oxidative markers LDH and MDA and increasing the levels of antioxidant stress SOD and GSH-Px, thereby slowing down the progression of SCIRI (Wu et al., 2024). Salidroside (Sal) is an effective component extracted from Cistanche deserticola and has multiple biological properties such as antioxidation, anti-apoptosis and neuroprotection (Cui et al., 2015). Gu research team confirmed in both in vivo and in vitro experiments of SCIRI that Sal can reduce the abnormal generation of ROS through antioxidant stress, lower the abnormal opening of mPTP, alleviate mitochondrial structural damage, and inhibit the release of Cyt C, blocking the activation of the mitochondrial-dependent apoptotic pathway (Gu et al., 2020). Nrf2 is a protective transcription factor that can regulate the oxidative stress response in cells, while HO-1 is an antioxidant protease that can enhance the resistance of neuronal cells to oxidative stress and protect neurological function (Abdel-Wahab et al., 2020). Hummus A (BA) is a methoxy isoflavone with antioxidant, anti-inflammatory and anti-apoptotic effects (Guo et al., 2019). A study has shown that BA alleviates cerebral ischemia/reperfusion injury by inhibiting apoptosis, endoplasmic reticulum stress and mitogen-activated protein kinase 14 pathway, thereby serving as an effective neuroprotective agent (Guo et al., 2021). In the SCIRI model, experimental studies have shown that BA intervention can specifically activate the Nrf2-mediated antioxidant defense system, significantly up-regulate the expression level of HO-1, and thereby effectively antagonize the oxidative stress cascade reaction induced by I/R, thereby significantly delaying I/R damage (Li et al., 2024). Astaxanthin (AST) is a powerful antioxidant and anti-inflammatory agent with multi-target neuroprotective effects (Fakhri et al., 2019). In the rat model of SCIRI, AST can alleviate pathological tissue damage and improve neurological function recovery after SCIRI by reducing oxidative stress (Fu et al., 2020). Ginsenoside Rb1 (GRb1) belongs to ginsenoside diol and has antioxidant and anti-apoptotic effects (Wynn et al., 2014). Previous studies have shown that GRb1 can significantly alleviate ischemia–reperfusion injury of the kidneys, brain, etc. (Schroeder et al., 2014). In the SCIRI study, GRb1 pretreatment of SCIRI rats can reduce neuronal apoptosis by increasing SOD activity in serum and spinal cord tissues and reducing the increase in MDA content. This indicates that GRb1 can alleviate SCIRI -induced apoptosis by inhibiting oxidative stress (Ye et al., 2019). In addition, monomer compounds such as Perilla aldehyde (Zheng et al., 2021), luteolin (Fu et al., 2018), and syringaldehyde (Malçok et al., 2020) can delay spinal cord I/R injury by reducing oxidative stress in ischemic spinal cord.
However, although these monomer compounds of traditional Chinese medicine have shown potential in basic research, their clinical transformation still faces multiple challenges. Firstly, the pharmacokinetic data of most compounds are limited, including that the absorption, distribution, metabolism and excretion characteristics have not been fully clarified, resulting in the lack of an optimization basis for the dosing regimen. Secondly, the high selectivity of the blood-spinal cord barrier severely limits the enrichment of drugs at the injury site. How to improve the targeted permeability remains a technical difficulty. Furthermore, most of the existing studies are based on animal models. The equivalence between the experimental dose and the human therapeutic dose is not yet clear, and the dose standards are inconsistent in different studies, which may affect the repeatability of therapeutic effects. More importantly, the vast majority of current achievements are still at the preclinical stage, lacking systematic clinical trials on human safety and efficacy, making it difficult to assess their actual medical value. In the future, strategies such as nano-delivery systems and structural modifications need to be adopted to improve bioavailability, and a standardized treatment system should be established in combination with multi-center clinical trials to promote the true clinical application of these natural active ingredients.
7.2 Pharmaceuticals
The recovery of neurological function after injury is a long process accompanying the entire pathological process. It has been found that many drugs can delay injury and promote the recovery of neurological function by inhibiting oxidative stress. For example, dexmedetomidine as a selective α 2-adrenergic receptor agonist, due to its extensive physiological functions and biological activities, multiple studies have shown that dexmedetomidine has a beneficial effect on I/R injury (Bunte et al., 2020). In the SCIRI rabbit model, dexmedetomidine can reduce the MDA content in spinal cord tissue, increase the SOD content, and improve the oxidative stress state, thereby alleviating SCIRI in rabbits (Liu et al., 2023). Lamotrigine is an anticonvulsant drug with multi-ion channel inhibitory properties, which has been proven to alleviate secondary neuronal injury by influencing different pathways. In studies related to SCIRI, Kahveci FO et al. found that lamotrigine treatment could protect the spinal cord from experimental ischemia–reperfusion injury by preventing lipid peroxidation and increased oxidative stress, as well as the consumption of antioxidant enzyme activity (Kahveci et al., 2021). It is reported that statins can reduce ischemia–reperfusion injury in many organs, including the spinal cord (Devarajan, 2006; Sironi et al., 2003). The antioxidant effect is one of the main protective mechanisms by which statins prevent neuronal death and cytotoxicity (Hwang et al., 2013). In studies related to spinal cord reperfusion injury, it has been found that statins such as atorvastatin (Kardes et al., 2017), rosuvastatin (Yavuz et al., 2013), and simvastatin (Sohn et al., 2017) can alleviate damage to spinal motor neurons caused by ischemia and reperfusion through their antioxidant effects. However, there are still significant limitations in the clinical application of the above-mentioned drugs. Firstly, the pharmacokinetic characteristics (such as half-life and tissue distribution) of dexmedetomidine and statins in the context of spinal cord injury have not been fully studied, which may affect the timing of administration and the stability of therapeutic effect. Secondly, the existence of the blood-spinal cord barrier severely limits the permeability of many drugs (such as lamotrigine) at the injury site, resulting in the actual action concentration being lower than the theoretical effective threshold. Furthermore, the dosage regimens in different studies vary greatly. For instance, there is no unified standard for the anti-inflammatory and antioxidant doses of statins, which may cause fluctuations in therapeutic efficacy or potential toxicity risks. More crucially, the existing evidence is mainly based on animal experiments, lacking large-scale clinical trials to verify the safety of these drugs in humans, their improvement effects on neurological function, and their long-term prognostic impact. In the future, spinal cord targeting needs to be enhanced by improving drug delivery systems (such as nanocarriers or targeted modifications), optimizing dosage strategies in combination with pharmacokinetic-pharmacodynamics models, and promoting multi-stage clinical trials to clarify its clinical translational value.
7.3 Other treatments
In addition to the above-mentioned treatments, the research also found that physical therapies such as hyperbaric oxygen and acupuncture can delay SCIRI by inhibiting oxidative damage. Studies have confirmed that hyperbaric oxygen pretreatment can induce ischemic tolerance in the brain and spinal cord (Xiong et al., 2000; Dong et al., 2002). In the rabbit SCIRI model, after pretreatment with hyperbaric oxygen, it was found that hyperbaric oxygen could activate the SOD activity in the rabbit spinal cord tissue, thereby eliminating ROS generated during ischemia–reperfusion and protecting the spinal cord from ROS-mediated injury (Nie et al., 2006). Furthermore, the research also found that acupuncture can inhibit oxidative stress to suppress the apoptosis of nerve cells, thereby delaying SCIIR (Cai and Shen, 2018). However, the clinical application of this type of physical therapy still faces many challenges. Firstly, the mechanism of action of hyperbaric oxygen and acupuncture has not been fully clarified, especially the insufficient systematic pharmacological research on their regulation of oxidative stress, making it difficult to precisely optimize the treatment plan. For example, the intervention timing, pressure parameters and treatment course standards of hyperbaric oxygen vary significantly in different studies, while the acupoint selection and stimulation intensity of acupuncture lack unified norms, which may affect the repeatability of therapeutic effects. Secondly, the targeting of physical therapy depends on local action, but the heterogeneity of the microenvironment in the spinal cord injury area may weaken its antioxidant effect. Furthermore, the existing research is mainly based on animal models and small-sample clinical trials, lacking large-scale multicenter randomized controlled trials to verify its long-term safety and the improvement effect on neurological function. In the future, it is necessary to clarify the dynamic mechanism of physical therapy through molecular imaging technology, establish individualized treatment parameters in combination with artificial intelligence algorithms, and promote evidence-based medical research to support clinical transformation.
8 Discussion and future outlook
The pathophysiological process after spinal cord ischemia–reperfusion injury is very complex. The imbalance of oxidants and antioxidant systems leads to an abnormal increase in ROS production. Excessive ROS can cause irreversible oxidative damage to macromolecules (such as nucleic acids, lipids and proteins), and interact with inflammatory responses, autophagy, and apoptosis. These processes can lead to complex secondary injury cascade reactions. This complexity may explain why many treatment strategies and experimental methods have not been successful in SCRI treatment. Oxidative stress plays an important role in SCIRI, and the levels of various free radicals can be used as indicators of damage. The ROS level can be regulated through the Nrf2 signaling pathway, and this regulation helps prevent or reduce spinal cord injury.
Although certain research results have been achieved on the mechanism of action and treatment strategies of oxidative stress in SCIRI at present, there are still many deficiencies. In terms of mechanism research, although it has been clearly identified that oxidative stress plays a key role in SCIRI, and certain understandings have been gained regarding the lipid peroxidation, protein oxidation, DNA damage it triggers, as well as its interaction with inflammatory responses, the potential molecular regulatory network between oxidative stress and other pathophysiological processes has not yet been fully clarified. For instance, there are relatively few studies on the interrelationships between oxidative stress and emerging research fields such as autophagy and ferroptosis in SCIRI. These complex interactions and potential molecular regulatory mechanisms still await in-depth exploration. Among the signaling pathways related to oxidative stress, although signaling pathways such as Nrf2, NF-kB and MAPK have received attention, there are still many downstream signaling molecules and the synergistic mechanisms among them that have not been fully clarified. In terms of treatment strategies, although the existing treatment methods based on oxidative stress have alleviated SCIRI to a certain extent, they still have limitations. The therapeutic effect of antioxidants is limited by factors such as the rapid generation rate of free radicals, drug stability and low bioavailability. It is difficult to completely eliminate excessive ROS in the body, and the optimal dosage, time window and combined use regimens of different antioxidants still need to be further optimized. Drugs that regulate oxidative stress signaling pathways face safety and side effect issues in clinical applications, and their long-term efficacy and potential risks also need to be verified by more large-scale clinical trials. The mechanisms of action of hyperbaric oxygen therapy and acupuncture therapy have not been fully clarified yet. There is a lack of unified treatment standards and norms, and the individual differences in treatment effects are relatively large.
In the future, in-depth research on the molecular mechanism of oxidative stress in SCIRI remains key. On the one hand, research on the interaction between oxidative stress and other pathophysiological processes should be strengthened. For instance, the relationship between oxidative stress and autophagy, ferroptosis, neuroplasticity, etc. should be deeply explored to reveal its potential molecular regulatory network and provide a theoretical basis for finding new therapeutic targets. Using multi-omics techniques, such as transcriptomics, proteomics, and metabolomics, etc., the changes of genes, proteins and metabolites during the SCIRI process were comprehensively analyzed to screen out the key molecules and signaling pathways closely related to oxidative stress. On the other hand, in terms of treatment strategies, efforts should be made to develop more effective treatment methods. Develop new types of antioxidants to enhance their stability, bioavailability and antioxidant activity, explore the application of emerging technologies such as nanotechnology and gene therapy in antioxidant treatment, and achieve precise delivery and targeted therapy of antioxidants. For example, through gene editing technology, the expression of antioxidant enzymes in spinal cord tissue is upregulated to enhance the body’s own antioxidant capacity. Using nanocarriers to precisely deliver antioxidants or drugs that regulate oxidative stress signaling pathways to damaged spinal cord tissue, improving therapeutic efficacy and reducing side effects. Further optimizing existing treatment methods and developing personalized treatment plans are also crucial. Based on the individual differences of patients, such as age, underlying diseases, and the degree of injury, optimize the regimens of antioxidant therapy, hyperbaric oxygen therapy, and acupuncture treatment to enhance the pertinence and effectiveness of the treatment. Strengthen research on combination therapy strategies, explore the synergistic mechanisms between different treatment methods, such as the combined application of antioxidants and stem cell therapy, neurotrophic factor therapy, etc., to provide more effective options for the clinical treatment of SCIRI. Strengthen the transformation between basic research and clinical practice, apply laboratory research results to clinical practice as soon as possible, carry out more large-scale and multi-center clinical trials, verify the safety and effectiveness of treatment methods, and bring better therapeutic effects and quality of life improvements to patients with SCIRI.
Author contributions
KZ: Writing – original draft, Writing – review & editing, Resources. Y-zX: Writing – original draft. MZ: Methodology, Writing – review & editing. J-jZ: Formal Analysis, Writing – review & editing. YX: Writing – original draft, Writing – review & editing. C-lX: Writing – original draft, Writing – review & editing.
Funding
The author(s) declare that no financial support was received for the research and/or publication of this article.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The authors declare that no Gen AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
Abdel-Wahab, B. A., Alkahtani, S. A., and Elagab, E. A. (2020). Tadalafil alleviates cisplatin-induced reproductive toxicity through the activation of the Nrf2/ho-1 pathway and the inhibition of oxidative stress and apoptosis in male rats. Reprod. Toxicol. 96, 165–174. doi: 10.1016/j.reprotox.2020.06.015
Ahuja, C. S., Wilson, J. R., Nori, S., Kotter, M. R. N., Druschel, C., Curt, A., et al. (2017). Traumatic spinal cord injury. Nat. Rev. Dis. Prim. 3:17018. doi: 10.1038/nrdp.2017.18
Al Mamun, A., Wu, Y., Monalisa, I., Jia, C., Zhou, K., Munir, F., et al. (2021). Role of pyroptosis in spinal cord injury and its therapeutic implications. J. Adv. Res. 28:97. doi: 10.1016/j.jare.2020.08.004
Alizadeh, A., Dyck, S. M., and Karimi-Abdolrezaee, S. (2019). Traumatic spinal cord injury: an overview of pathophysiology, models and acute injury mechanisms. Front. Neurol. 10:282. doi: 10.3389/fneur.2019.00282
Allen, C. L., and Bayraktutan, U. J. I. J. O. S. (2009). Oxidative stress and its role in the pathogenesis of ischaemic stroke. Int. J. Stroke 4, 461–470. doi: 10.1111/j.1747-4949.2009.00387.x
Altinors, N. J. (2009). Analysis of serum pro-inflammatory cytokine levels after rat spinal cord ischemia/reperfusion injury and correlation with tissue damage. Turk. Neurosurg. 19, 353–359.
Angkeow, P., Deshpande, S., Qi, B., Liu, Y.-X., Park, Y. C., Jeon, B. H., et al. (2002). Redox factor-1: an extra-nuclear role in the regulation of endothelial oxidative stress and apoptosis. Cell Death Differ. 9, 717–725. doi: 10.1038/sj.cdd.4401025
Anne Stetler, R., Leak, R. K., Gao, Y., and Chen, J. (2013). The dynamics of the mitochondrial organelle as a potential therapeutic target. J. Cereb. Blood Flow Metabol. 33, 22–32. doi: 10.1038/jcbfm.2012.158
Arvola, O., Haapanen, H., Herajärvi, J., Anttila, T., Puistola, U., Karihtala, P., et al. (2016). Remote ischemic preconditioning attenuates oxidative stress during cardiopulmonary bypass. Heart Surg. Forum 19, 192–197. doi: 10.1532/hsf.1590
Bachstetter, A. D., Xing, B., De Almeida, L., Dimayuga, E. R., Watterson, D. M., and Van Eldik, L. J. (2011). Microglial p38α Mapk is a key regulator of proinflammatory cytokine up-regulation induced by toll-like receptor (Tlr) ligands or beta-amyloid (Aβ). J. Neuroinflammation 8:79. doi: 10.1186/1742-2094-8-79
Bao, G., Li, C., Qi, L., Wang, N., and He, B. (2016). Tetrandrine protects against oxygen-glucose-serum deprivation/reoxygenation-induced injury via Pi3K/Akt/Nf-κB signaling pathway in rat spinal cord astrocytes. Biomed. Pharmacother. 84:925. doi: 10.1016/j.biopha.2016.10.007
Bell, M. T., Puskas, F., Agoston, V. A., Cleveland, J. C., Freeman, K. A., Gamboni, F., et al. (2013). Toll-like receptor 4-dependent microglial activation mediates spinal cord ischemia-reperfusion injury. Circulation 128, S152–S156. doi: 10.1161/CIRCULATIONAHA.112.000024
Bergsbaken, T., Fink, S. L., and Cookson, B. T. (2009). Pyroptosis: host cell death and inflammation. Nat. Rev. Microbiol. 7, 99–109. doi: 10.1038/nrmicro2070
Berlett, B. S., and Stadtman, E. R. J. J. O. B. C. (1997). Protein oxidation in aging, disease, and oxidative stress 272, 20313–20316. doi: 10.1074/jbc.272.33.20313
Bernardi, P., Rasola, A., Forte, M., and Lippe, G. (2015). The mitochondrial permeability transition pore: channel formation by F-Atp synthase, integration in signal transduction, and role in pathophysiology. Physiol. Rev. 95, 1111–1155. doi: 10.1152/physrev.00001.2015
Blaser, H., Dostert, C., Mak, T. W., and Brenner, D. (2016). TNF and ROS crosstalk in inflammation. Trends Cell Biol. 26, 249–261. doi: 10.1016/j.tcb.2015.12.002
Bode, J. G., Ehlting, C., and Häussinger, D. (2012). The macrophage response towards Lps and its control through the p38(Mapk)-Stat3 axis. Cell. Signal. 24, 1185–1194. doi: 10.1016/j.cellsig.2012.01.018
Boyle, E. M., Canty, T. G. Jr., Morgan, E. N., Morgan, E. N., Yun, W., Pohlman, T. H., et al. (1999). Treating myocardial ischemia-reperfusion injury by targeting endothelial cell transcription. Ann. Thorac. Surg. 68, 1949–1953. doi: 10.1016/S0003-4975(99)01033-4
Brand, M. D. (2010). The sites and topology of mitochondrial superoxide production. Exp. Gerontol. 45, 466–472. doi: 10.1016/j.exger.2010.01.003
Breton-Romero, R., and Lamas, S. J. (2014). Hydrogen peroxide signaling in vascular endothelial cells. Redox. Biol. 2, 529–534. doi: 10.1016/j.redox.2014.02.005
Bunte, S., Behmenburg, F., Majewski, N., Stroethoff, M., Raupach, A., Mathes, A., et al. (2020). Characteristics of Dexmedetomidine Postconditioning in the field of myocardial ischemia-reperfusion injury. Anesth. Analg. 130, 90–98. doi: 10.1213/ANE.0000000000004417
Busquets-Cortés, C., Capó, X., Argelich, E., Ferrer, M., Mateos, D., Bouzas, C., et al. (2018). Effects of Millimolar steady-state hydrogen peroxide exposure on inflammatory and redox gene expression in immune cells from humans with metabolic syndrome. Nutrients 10:1920. doi: 10.3390/nu10121920
Bystrom, P., Foley, N., Toledo-Pereyra, L., and Quesnelle, K. (2017). Ischemic preconditioning modulates Ros to confer protection in liver ischemia and reperfusion. EXCLI J. 16, 483–496.
Cadenas, S. (2018). Ros and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection. Free Radic. Biol. Med. 117, 76–89. doi: 10.1016/j.freeradbiomed.2018.01.024
Cai, W., and Shen, W. D. (2018). Anti-apoptotic mechanisms of acupuncture in neurological diseases: a review. Am. J. Chin. Med. 46, 515–535. doi: 10.1142/S0192415X1850026X
Chen, G. H., Song, C. C., Pantopoulos, K., Wei, X. L., Zheng, H., and Luo, Z. (2022). Mitochondrial oxidative stress mediated Fe-induced ferroptosis via the Nrf2-are pathway. Free Radic. Biol. Med. 180:95. doi: 10.1016/j.freeradbiomed.2022.01.012
Chen, S. D., Yang, D. I., Lin, T. K., Shaw, F. Z., Liou, C. W., and Chuang, Y. C. (2011). Roles of oxidative stress, apoptosis, Pgc-1α and mitochondrial biogenesis in cerebral ischemia. Int. J. Mol. Sci. 12, 7199–7215. doi: 10.3390/ijms12107199
Chi, P. L., Lin, C. C., Chen, Y. W., Hsiao, L. D., and Yang, C. M. (2015). Co induces Nrf2-dependent Heme Oxygenase-1 transcription by cooperating with Sp1 and c-Jun in rat brain astrocytes. Mol. Neurobiol. 52, 277–292. doi: 10.1007/s12035-014-8869-4
Cui, J.-L., Guo, T.-T., Ren, Z.-X., Zhang, N. S., and Wang, M. L. (2015). Diversity and antioxidant activity of culturable endophytic fungi from alpine plants of Rhodiola crenulata, R. angusta, and R. sachalinensis. PLoS One 10:e0118204. doi: 10.1371/journal.pone.0118204
D'arcy, M. S. (2019). Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 43, 582–592. doi: 10.1002/cbin.11137
Degterev, A., Huang, Z., Boyce, M., Li, Y., Jagtap, P., Mizushima, N., et al. (2005). Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 1, 112–119. doi: 10.1038/nchembio711
Devarajan, P. (2006). Update on mechanisms of ischemic acute kidney injury. J. Am. Soc. Nephrol. 17, 1503–1520. doi: 10.1681/ASN.2006010017
Ding, G. R., Qiu, L. B., Wang, X. W., Li, K. C., Zhou, Y. C., Zhou, Y., et al. (2010). Emp-induced alterations of tight junction protein expression and disruption of the blood-brain barrier. Toxicol. Lett. 196, 154–160. doi: 10.1016/j.toxlet.2010.04.011
Dixon, S. J., Lemberg, K. M., Lamprecht, M. R., Skouta, R., Zaitsev, E. M., Gleason, C. E., et al. (2012). Ferroptosis: an Iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072. doi: 10.1016/j.cell.2012.03.042
Dong, H., Xiong, L., Zhu, Z., Chen, S., Hou, L., and Sakabe, T. (2002). Preconditioning with hyperbaric oxygen and Hyperoxia induces tolerance against spinal cord ischemia in rabbits. Anesthesiology 96, 907–912. doi: 10.1097/00000542-200204000-00018
Dong, H. L., Zhang, Y., Su, B. X., Zhu, Z. H., Gu, Q. H., Sang, H. F., et al. (2010). Limb remote ischemic preconditioning protects the spinal cord from ischemia-reperfusion injury: a newly identified nonneuronal but reactive oxygen species-dependent pathway. Anesthesiology 112, 881–891. doi: 10.1097/ALN.0b013e3181d0486d
Duan, Y., Yang, F., Zhang, Y., Zhang, M., Shi, Y., Lang, Y., et al. (2024). Role of mitophagy in spinal cord ischemia-reperfusion injury. Neural Regen. Res. 21, 598–611. doi: 10.4103/NRR.NRR-D-24-00668
Ebrahimy, N., Gasterich, N., Behrens, V., Amini, J., Fragoulis, A., Beyer, C., et al. (2022). Neuroprotective effect of the Nrf2/are/mirna145-5p signaling pathway in the early phase of spinal cord injury. Life Sci. 304:120726. doi: 10.1016/j.lfs.2022.120726
El Assar, M., Angulo, J., and Rodríguez-Mañas, L. (2013). Oxidative stress and vascular inflammation in aging. Free Radic. Biol. Med. 65, 380–401. doi: 10.1016/j.freeradbiomed.2013.07.003
Fakhri, S., Abbaszadeh, F., Moradi, S. Z., Cao, H., Khan, H., and Xiao, J. (2022). Effects of polyphenols on oxidative stress, inflammation, and interconnected pathways during spinal cord injury. Oxidative Med. Cell. Longev. 2022, 1–34. doi: 10.1155/2022/8100195
Fakhri, S., Aneva, Y. I, Farzaei, M. H., and Sobarzo-Sánchez, E. (2019). The neuroprotective effects of Astaxanthin: therapeutic targets and clinical perspective. Molecules 24:2640. doi: 10.3390/molecules24142640
Farías, J. G., Molina, V. M., Carrasco, R. A., Zepeda, A. B., Figueroa, E., Letelier, P., et al. (2017). Antioxidant therapeutic strategies for cardiovascular conditions associated with oxidative stress. Nutrients 9:966. doi: 10.3390/nu9090966
Feng, Z., Min, L., Chen, H., Deng, W., Tan, M., Liu, H., et al. (2021). Iron overload in the motor cortex induces neuronal ferroptosis following spinal cord injury. Redox Biol. 43:101984. doi: 10.1016/j.redox.2021.101984
Fink, S. L., and Cookson, B. T. (2006). Caspase-1-dependent pore formation during pyroptosis leads to osmotic lysis of infected host macrophages. Cell. Microbiol. 8, 1812–1825. doi: 10.1111/j.1462-5822.2006.00751.x
Fu, J., Sun, H., Wei, H., Dong, M., Zhang, Y., Xu, W., et al. (2020). Astaxanthin alleviates spinal cord ischemia-reperfusion injury via activation of Pi3K/Akt/Gsk-3β pathway in rats. J. Orthop. Surg. Res. 15:275. doi: 10.1186/s13018-020-01790-8
Fu, J., Sun, H., Zhang, Y., Xu, W., Wang, C., Fang, Y., et al. (2018). Neuroprotective effects of Luteolin against spinal cord ischemia-reperfusion injury by attenuation of oxidative stress, inflammation, and apoptosis. J. Med. Food 21, 13–20. doi: 10.1089/jmf.2017.4021
Galluzzi, L., Bravo-San Pedro, J. M., Vitale, I., Aaronson, S. A., Abrams, J. M., Adam, D., et al. (2015). Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ. 22, 58–73. doi: 10.1038/cdd.2014.137
Galluzzi, L., Vitale, I., Aaronson, S. A., Abrams, J. M., Adam, D., Agostinis, P., et al. (2018). Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 25, 486–541. doi: 10.1038/s41418-017-0012-4
Gassanov, N., Nia, A. M., Caglayan, E., and Er, F. (2014). Remote ischemic preconditioning and renoprotection: from myth to a novel therapeutic option? J. Am. Soc. Nephrol. 25, 216–224. doi: 10.1681/ASN.2013070708
Giorgio, M., Trinei, M., Migliaccio, E., and Pelicci, P. G. (2007). Hydrogen peroxide: a metabolic by-product or a common mediator of ageing signals? Nat. Rev. Mol. Cell Biol. 8, 722–728. doi: 10.1038/nrm2240
Gökce, E. C., Kahveci, R., Gökce, A., Cemil, B., Aksoy, N., Sargon, M. F., et al. (2016). Neuroprotective effects of thymoquinone against spinal cord ischemia-reperfusion injury by attenuation of inflammation, oxidative stress, and apoptosis. J. Neurosurg. Spine 24, 949–959. doi: 10.3171/2015.10.SPINE15612
Goldhaber, J. I., and Weiss, J. N. J. (1992). Oxygen free radicals and cardiac reperfusion abnormalities. Hypertension 20, 118–127. doi: 10.1161/01.HYP.20.1.118
Granger, D. N., and Kvietys, P. R. (2015). Reperfusion injury and reactive oxygen species: the evolution of a concept. Redox. Biol. 6, 524–551. doi: 10.1016/j.redox.2015.08.020
Green, D. R. (1998). Apoptotic pathways: the roads to ruin. Cell 94, 695–698. doi: 10.1016/S0092-8674(00)81728-6
Gu, C., Kong, F., Zeng, J., Geng, X., Sun, Y., and Chen, X. (2023). Remote ischemic preconditioning protects against spinal cord ischemia-reperfusion injury in mice by activating Nmdar/Ampk/Pgc-1α/Sirt3 signaling. Cell Biosci. 13:57. doi: 10.1186/s13578-023-00999-4
Gu, C., Li, L., Huang, Y., Qian, D., Liu, W., Zhang, C., et al. (2020). Salidroside ameliorates mitochondria-dependent neuronal apoptosis after spinal cord ischemia-reperfusion injury partially through inhibiting oxidative stress and promoting Mitophagy. Oxidative Med. Cell. Longev. 2020, 1–22. doi: 10.1155/2020/3549704
Guo, M., Lu, H., Qin, J., Qu, S., Wang, W., Guo, Y., et al. (2019). Biochanin a provides neuroprotection against cerebral ischemia/reperfusion injury by Nrf2-mediated inhibition of oxidative stress and inflammation signaling pathway in rats. Med. Sci. Monit. 25, 8975–8983. doi: 10.12659/MSM.918665
Guo, M.-M., Qu, S.-B., Lu, H.-L., Wang, W. B., He, M. L., Su, J. L., et al. (2021). Biochanin a alleviates cerebral ischemia/reperfusion injury by suppressing endoplasmic reticulum stress-induced apoptosis and p38MAPK signaling pathway in vivo and in vitro. Front. Endocrinol. 12, 12:646720. doi: 10.3389/fendo.2021.646720
Hall, E. D. (2011). Antioxidant therapies for acute spinal cord injury. Neurotherapeutics 8, 152–167. doi: 10.1007/s13311-011-0026-4
Hall, E. D., Wang, J. A., Bosken, J. M., and Singh, I. N. (2016). Lipid peroxidation in brain or spinal cord mitochondria after injury. J. Bioenerg. Biomembr. 48, 169–174. doi: 10.1007/s10863-015-9600-5
Halladin, N. L. (2015). Oxidative and inflammatory biomarkers of ischemia and reperfusion injuries. Dan. Med. J. 62:B5054
Han, M. S., Barrett, T., Brehm, M. A., and Davis, R. J. (2016). Inflammation mediated by Jnk in myeloid cells promotes the development of hepatitis and hepatocellular carcinoma. Cell Rep. 15, 19–26. doi: 10.1016/j.celrep.2016.03.008
Han, C. H., Guan, Z. B., Zhang, P. X., Fang, H. L., Li, L., Zhang, H. M., et al. (2018). Oxidative stress induced necroptosis activation is involved in the pathogenesis of hyperoxic acute lung injury. Biochem. Biophys. Res. Commun. 495, 2178–2183. doi: 10.1016/j.bbrc.2017.12.100
Han, B., Jiang, W., Liu, H., Wang, J., Zheng, K., Cui, P., et al. (2020). Upregulation of neuronal Pgc-1α ameliorates cognitive impairment induced by chronic cerebral hypoperfusion. Theranostics 10, 2832–2848. doi: 10.7150/thno.37119
Hausenloy, D. J., and Yellon, D. M. (2011). The therapeutic potential of ischemic conditioning: an update. Nat. Rev. Cardiol. 8, 619–629. doi: 10.1038/nrcardio.2011.85
He, Y., Hara, H., and Núñez, G. (2016). Mechanism and regulation of Nlrp3 inflammasome activation. Trends Biochem. Sci. 41, 1012–1021. doi: 10.1016/j.tibs.2016.09.002
He, J., Ritzel, R. M., and Wu, J. (2021). Functions and mechanisms of the voltage-gated Proton Channel Hv1 in brain and spinal cord injury. Front. Cell. Neurosci. 15:662971. doi: 10.3389/fncel.2021.662971
Hnath, J. C., Mehta, M., Taggert, J. B., Sternbach, Y., Roddy, S. P., Kreienberg, P. B., et al. (2008). Strategies to improve spinal cord ischemia in endovascular thoracic aortic repair: outcomes of a prospective cerebrospinal fluid drainage protocol. J. Vasc. Surg. 48, 836–840. doi: 10.1016/j.jvs.2008.05.073
Holzerová, E., and Prokisch, H. (2015). Mitochondria: much ado about nothing? How dangerous is reactive oxygen species production? Int. J. Biochem. Cell Biol. 63:16. doi: 10.1016/j.biocel.2015.01.021
Hu, L., Wang, Y., Ren, R., Huo, H., Sun, J., Li, H., et al. (2016). Anti-oxidative stress actions and regulation mechanisms of Keap1-Nrf2/are signal pathway 43, 146–166. doi: 10.13220/j.cnki.jipr.2016.01.022
Huang, Y., Cai, G.-Q., Peng, J.-P., and Shen, C. (2018). Glucocorticoids induce apoptosis and matrix metalloproteinase-13 expression in chondrocytes through the Nox4/Ros/p38 Mapk pathway. J. Steroid Biochem. Mol. Biol. 181:52. doi: 10.1016/j.jsbmb.2018.03.001
Huang, P., Chen, G., Jin, W., Mao, K., Wan, H., and He, Y. (2022). Molecular mechanisms of Parthanatos and its role in diverse diseases. Int. J. Mol. Sci. 23:7292. doi: 10.3390/ijms23137292
Hwang, J., Han, J. I., Han, S. J. J. O. C., and Han, S. (2013). Effect of pretreatment with simvastatin on spinal cord ischemia-reperfusion injury in rats. J. Cardiothorac. Vasc. Anesth. 27, 79–85. doi: 10.1053/j.jvca.2012.01.025
Iantomasi, T., Romagnoli, C., Palmini, G., Donati, S., Falsetti, I., Miglietta, F., et al. (2023). Oxidative stress and inflammation in osteoporosis: molecular mechanisms involved and the relationship with micrornas. Int. J. Mol. Sci. 24:3772. doi: 10.3390/ijms24043772
Ilhan, A., Yilmaz, H. R., Armutcu, F., Gurel, A., and Akyol, O. (2004). The protective effect of nebivolol on ischemia/reperfusion injury in rabbit spinal cord. Prog. Neuropsychopharmacol. Biol. Psychiatry 28, 1153–1160. doi: 10.1016/j.pnpbp.2004.06.023
Janyou, A., Wicha, P., Jittiwat, J., Suksamrarn, A., Tocharus, C., and Tocharus, J. (2017). Dihydrocapsaicin attenuates blood brain barrier and cerebral damage in focal cerebral ischemia/reperfusion via oxidative stress and inflammatory. Sci. Rep. 7:10556. doi: 10.1038/s41598-017-11181-5
Jin, H., Ge, X., Huan, Z., Yao, H., Xu, C., and Cai, J. (2021). Stress-induced phosphoprotein 1 restrains spinal cord ischaemia-reperfusion injury by modulating Nf-κB signalling. J. Cell. Mol. Med. 25, 11075–11084. doi: 10.1111/jcmm.17030
Jung, K. A., and Kwak, M. K. (2010). The Nrf2 system as a potential target for the development of indirect antioxidants. Molecules 15, 7266–7291. doi: 10.3390/molecules15107266
Kahveci, F. O., Kahveci, R., Gokce, E. C., Gokce, A., Kısa, Ü., Sargon, M. F., et al. (2021). Biochemical, pathological and ultrastructural investigation of whether lamotrigine has neuroprotective efficacy against spinal cord ischemia reperfusion injury. Injury 52, 2803–2812. doi: 10.1016/j.injury.2021.08.004
Kalkavan, H., and Green, D. R. (2018). Cell suicide as a Bcl-2 family business. Cell Death Differ. 25, 46–55. doi: 10.1038/cdd.2017.179
Kanno, A., Ozawa, T., and Umezawa, Y. (2009). Bioluminescent imaging of Mapk function with intein-mediated reporter gene assay. Methods Mol. Biol. 574, 185–192. doi: 10.1007/978-1-60327-321-3_15
Kardes, O., Civi, S., Tufan, K., Oyar, E. O., Omeroglu, S., and Aykol, S. (2017). Effects of atorvastatin on experimental spinal cord ischemia-reperfusion injury in rabbits. Turk. Neurosurg. 27, 594–602. doi: 10.5137/1019-5149.JTN.16627-15.2
Karin, M., Yamamoto, Y., and Wang, Q. M. (2004). The Ikk Nf-kappa B system: a treasure trove for drug development. Nat. Rev. Drug Discov. 3, 17–26. doi: 10.1038/nrd1279
Kevin, L. G., Novalija, E., Stowe, D. F. J. A., and Stowe, D. F. (2005). Reactive oxygen species as mediators of cardiac injury and protection: the relevance to Anesthesia practice. Anesth Analg 101, 1275–1287. doi: 10.1213/01.ANE.0000180999.81013.D0
Khachatryan, Z., Haunschild, J., Von Aspern, K., Borger, M. A., and Etz, C. D. (2022). Ischemic spinal cord injury-experimental evidence and evolution of protective measures. Ann. Thorac. Surg. 113, 1692–1702. doi: 10.1016/j.athoracsur.2020.12.028
Khattab, M. G., Sayed, Z. S., Altaf, R. A., Yasen, N. S., Al Aiashy, A., Alfeqy, H. M., et al. (2022). The prophylactic roles of dietary antioxidants for medical radiology workers: a mini-review. Nrfhh 2, 391–407. doi: 10.53365/nrfhh/146248
Khayrullina, G., Bermudez, S., and Byrnes, K. R. (2015). Inhibition of Nox2 reduces locomotor impairment, inflammation, and oxidative stress after spinal cord injury. J. Neuroinflammation 12:172. doi: 10.1186/s12974-015-0391-8
Kim, W., Cho, S. B., Jung, H. Y., Yoo, D. Y., Oh, J. K., Choi, G. M., et al. (2019). Phosphatidylethanolamine-binding protein 1 ameliorates ischemia-induced inflammation and neuronal damage in the rabbit spinal cord. Cells 8:1370. doi: 10.3390/cells8111370
Kim, Y., Cho, A. Y., Kim, H. C., Ryu, D., Jo, S. A., and Jung, Y.-S. (2022). Effects of natural polyphenols on oxidative stress-mediated blood-brain barrier dysfunction. Antioxidants 11:197. doi: 10.3390/antiox11020197
Kim, E. K., and Choi, E.-J. (2010). Pathological roles of Mapk signaling pathways in human diseases. Biochim. Biophys Acta 1802, 396–405. doi: 10.1016/j.bbadis.2009.12.009
Kleinbongard, P., Peters, J., Jakob, H., Heusch, G., and Thielmann, M. (2018). Persistent survival benefit from remote ischemic pre-conditioning in patients undergoing coronary artery bypass surgery. J. Am. Coll Cardiol. 71, 252–254. doi: 10.1016/j.jacc.2017.10.083
Klionsky, D. J., and Emr, S. D. (2000). Autophagy as a regulated pathway of cellular degradation. Science 290, 1717–1721. doi: 10.1126/science.290.5497.1717
Krajka-KuŹniak, V., Paluszczak, J., and Baer-Dubowska, W. (2017). The Nrf2-are signaling pathway: an update on its regulation and possible role in cancer prevention and treatment. Pharmacol. Rep. 69, 393–402. doi: 10.1016/j.pharep.2016.12.011
Kryl'skii, E. D., Popova, T. N., Safonova, O. A., Stolyarova, A. O., Razuvaev, G. A., and de Carvalho, M. A. P. (2019). Transcriptional regulation of antioxidant enzymes activity and modulation of oxidative stress by melatonin in rats under cerebral ischemia/reperfusion conditions. Neuroscience 406:653. doi: 10.1016/j.neuroscience.2019.01.046
Lacey, C. A., Mitchell, W. J., Dadelahi, A. S., and Skyberg, J. A. (2018). Caspase-1 and Caspase-11 mediate Pyroptosis, inflammation, and control of Brucella joint infection. Infect. Immun. 86:e00361-18. doi: 10.1128/IAI.00361-18
Lee, K. E., Kim, J., Park, K. H., Ma, J., and Zhu, H. (2023). Remote ischemic pre-conditioning (Ripc). Methods Mol. Biol. 2597, 11–18.
Lemaire, S. A., Price, M. D., Green, S. Y., Zarda, S., and Coselli, J. S. (2012). Results of open thoracoabdominal aortic aneurysm repair. Ann. Cardiothor. Surg. 1, 286–292.
Li, Y., Chen, L., Gao, Y., Zou, X., and Wei, F. (2022). Oxidative stress and intervertebral disc degeneration: pathophysiology, signaling pathway, and therapy. Oxidative Med. Cell. Longev. 2022, 1–14. doi: 10.1155/2022/1984742
Li, X., Fu, J., Guan, M., Shi, H., Pan, W., and Lou, X. (2024). Biochanin a attenuates spinal cord injury in rats during early stages by inhibiting oxidative stress and inflammasome activation. Neural Regen. Res. 19, 2050–2056. doi: 10.4103/1673-5374.390953
Li, S., Hu, X., Zhang, M., Zhou, F., Lin, N., Xia, Q., et al. (2015). Remote ischemic post-conditioning improves neurological function by Aqp4 down-regulation in astrocytes. Behav. Brain Res. 289:1. doi: 10.1016/j.bbr.2015.04.024
Li, Y., Lucas-Osma, A. M., Black, S., Bandet, M. V., Stephens, M. J., and Vavrek, R. (2017). Pericytes impair capillary blood flow and motor function after chronic spinal cord injury. Nat. Med. 23, 733–741. doi: 10.1038/nm.4331
Li, P., Nijhawan, D., Budihardjo, I., Srinivasula, S. M., Ahmad, M., Alnemri, E. S., et al. (1997). Cytochrome c and dATP-dependent formation of Apaf-1/Caspase-9 complex initiates an apoptotic protease Cascade. Cell 91, 479–489. doi: 10.1016/S0092-8674(00)80434-1
Li, Z., Wu, F., Xu, D., Zhi, Z., and Xu, G. (2019). Inhibition of Trem1 reduces inflammation and oxidative stress after spinal cord injury (Sci) associated with ho-1 expressions. Biomed. Pharmacother. 109:2014. doi: 10.1016/j.biopha.2018.08.159
Liang, W., Lin, C., Yuan, L., Chen, L., Guo, P., Li, P., et al. (2019). Preactivation of Notch1 in remote ischemic preconditioning reduces cerebral ischemia-reperfusion injury through crosstalk with the Nf-κB pathway. J. Neuroinflammation 16:181. doi: 10.1186/s12974-019-1570-9
Lin, W. P., Xiong, G. P., Lin, Q., Chen, X. W., Zhang, L. Q., Shi, J. X., et al. (2016). Heme oxygenase-1 promotes neuron survival through down-regulation of neuronal Nlrp1 expression after spinal cord injury. J. Neuroinflammation 13:52. doi: 10.1186/s12974-016-0521-y
Ling, X., Lu, J., Yang, J., Qin, H., Zhao, X., Zhou, P., et al. (2021). Non-coding Rnas: emerging therapeutic targets in spinal cord ischemia-reperfusion injury. Front. Neurol. 12:680210. doi: 10.3389/fneur.2021.680210
Liu, B., Liang, Y., Huang, W., Zhang, H., Zhou, D., and Xiao, X. (2023). Dexmedetomidine attenuates spinal cord ischemia-reperfusion injury in rabbits by decreasing oxidation and apoptosis. Curr. Mol. Med. 23, 569–577. doi: 10.2174/1566524022666220525142954
Liu, Y., Pan, L., Jiang, A., and Yin, M. (2018). Hydrogen sulfide upregulated lncrna CasC7 to reduce neuronal cell apoptosis in spinal cord ischemia-reperfusion injury rat. Biomed. Pharmacother. 98:856. doi: 10.1016/j.biopha.2017.12.079
Liu, L., and Sun, B. (2019). Neutrophil pyroptosis: new perspectives on sepsis. Cell Mol. Life Sci. 76, 2031–2042. doi: 10.1007/s00018-019-03060-1
Liu, Z., Yao, X., Jiang, W., Li, W., Zhu, S., Liao, C., et al. (2020). Advanced oxidation protein products induce microglia-mediated neuroinflammation via Mapks-Nf-κB signaling pathway and pyroptosis after secondary spinal cord injury. J. Neuroinflammation 17:90. doi: 10.1186/s12974-020-01751-2
Liu, S., Yao, S., Yang, H., Liu, S. J., and Wang, Y. J. (2023). Autophagy: regulator of cell death. Cell Death Dis. 14:648. doi: 10.1038/s41419-023-06154-8
Lochhead, J. J., Mccaffrey, G., Quigley, C. E., Finch, J., DeMarco, K. M., Nametz, N., et al. (2010). Oxidative stress increases blood-brain barrier permeability and induces alterations in occludin during hypoxia-reoxygenation. J. Cerebr. Blood Flow Metab. 30, 1625–1636. doi: 10.1038/jcbfm.2010.29
Lu, M. C., Ji, J. A., Jiang, Z. Y., and You, Q. D. (2016). The Keap1–Nrf2–are pathway as a potential preventive and therapeutic target: an update. Med. Res. Rev. 36, 924–963. doi: 10.1002/med.21396
Ma, Q. (2013). Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 53:401. doi: 10.1146/annurev-pharmtox-011112-140320
Machado, D. I., De Oliveira, S. E., Ventura, S., and Vattimo, M. F. F. (2022). The effect of curcumin on renal ischemia/reperfusion injury in diabetic rats. Nutrients 14:2798. doi: 10.3390/nu14142798
Maiuri, M. C., Zalckvar, E., Kimchi, A., and Kroemer, G. (2007). Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 8, 741–752. doi: 10.1038/nrm2239
Malçok, Ü. A., Aras, A. B., Şehitoğlu, M. H., Akman, T., and Yüksel, Y. (2020). Therapeutic effects of syringaldehyde on spinal cord ischemia in rabbits. Saudi Med. J. 41, 341–350. doi: 10.15537/smj.2020.4.24993
Mármol, I., Virumbrales-Muñoz, M., Quero, J., Sánchez-de-Diego, C., Fernández, L., Ochoa, I., et al. (2017). Alkynyl gold(I) complex triggers necroptosis via Ros generation in colorectal carcinoma cells. J. Inorg. Biochem. 176:123. doi: 10.1016/j.jinorgbio.2017.08.020
Matsumoto, S., Matsumoto, M., Yamashita, A., Ohtake, K., Ishida, K., Morimoto, Y., et al. (2003). The temporal profile of the reaction of microglia, astrocytes, and macrophages in the delayed onset paraplegia after transient spinal cord ischemia in rabbits. Anesth. Analg. 96, 1777–1784. doi: 10.1213/01.ANE.0000064204.67561.73
Mcconnell, H. L., Kersch, C. N., Woltjer, R. L., and Neuwelt, E. A. (2017). The translational significance of the neurovascular unit. J. Biol. Chem. 292, 762–770. doi: 10.1074/jbc.R116.760215
Mccord, J. M. (1985). Oxygen-derived free radicals in Postischemic tissue injury. N. Engl. J. Med. 312, 159–163. doi: 10.1056/NEJM198501173120305
Mirzaei, S., Hushmandi, K., Zabolian, A., Saleki, H., Torabi, S. M. R., Ranjbar, A., et al. (2021). Elucidating role of reactive oxygen species (ROS) in cisplatin chemotherapy: a focus on molecular pathways and possible therapeutic strategies. Molecules 26:2382. doi: 10.3390/molecules26082382
Mlynarczuk-Bialy, I., Dziuba, I., Sarnecka, A., Platos, E., Kowalczyk, M., Pels, K. K., et al. (2020). Entosis: from cell biology to clinical Cancer pathology. Cancers 12:2481. doi: 10.3390/cancers12092481
Mokhtari-Zaer, A., Marefati, N., Atkin, S. L., Butler, A. E., and Sahebkar, A. (2019). The protective role of curcumin in myocardial ischemia–reperfusion injury. J. Cell Physiol. 234, 214–222. doi: 10.1002/jcp.26848
Morgan, M. J., and Liu, Z. G. (2011). Crosstalk of reactive oxygen species and Nf-κB signaling. Cell Res. 21, 103–115. doi: 10.1038/cr.2010.178
Motomura, A., Shimizu, M., Kato, A., Motomura, K., Yamamichi, A., Koyama, H., et al. (2017). Remote ischemic preconditioning protects human neural stem cells from oxidative stress. Apoptosis 22, 1353–1361. doi: 10.1007/s10495-017-1425-8
Neri, M., Fineschi, V., Di Paolo, M., Paolo, M., Pomara, C., Riezzo, I., et al. (2015). Cardiac oxidative stress and inflammatory cytokines response after myocardial infarction. Curr. Vasc. Pharmacol. 13, 26–36. doi: 10.2174/15701611113119990003
Nguyen, T. T., Ung, T. T., Li, S., Lian, S., Xia, Y., Park, S. Y., et al. (2019). Metformin inhibits lithocholic acid-induced interleukin 8 upregulation in colorectal cancer cells by suppressing Ros production and Nf-kB activity. Sci. Rep. 9:2003. doi: 10.1038/s41598-019-38778-2
Nie, H., Xiong, L., Lao, N., Chen, S., Xu, N., and Zhu, Z. (2006). Hyperbaric oxygen preconditioning induces tolerance against spinal cord ischemia by upregulation of antioxidant enzymes in rabbits. J. Cerebr. Blood Flow Metabol. 26, 666–674. doi: 10.1038/sj.jcbfm.9600221
Oeckinghaus, A., Hayden, M. S., and Ghosh, S. J. (2011). Crosstalk in NF-κB signaling pathways. Nat. Immunol. 12, 695–708. doi: 10.1038/ni.2065
Orrenius, S., Gogvadze, V., and Zhivotovsky, B. (2007). Mitochondrial oxidative stress: implications for cell death. Annu. Rev. Pharmacol. Toxicol. 47, 143–183. doi: 10.1146/annurev.pharmtox.47.120505.105122
O'shea, T. M., Burda, J. E., and Sofroniew, M. V. (2017). Cell biology of spinal cord injury and repair. J. Clin. Invest. 127, 3259–3270. doi: 10.1172/JCI90608
Pizzino, G., Irrera, N., Cucinotta, M., Pallio, G., Mannino, F., Arcoraci, V., et al. (2017). Oxidative stress: harms and benefits for human health. Oxidative Med. Cell. Longev. 2017:8416763. doi: 10.1155/2017/8416763
Prestera, T., Talalay, P., Alam, J., Ahn, Y. I., Lee, P. J., and Choi, A. M. K. (1995). Parallel induction of heme oxygenase-1 and chemoprotective phase 2 enzymes by electrophiles and antioxidants: regulation by upstream antioxidant-responsive elements (are). Mol. Med. 1, 827–837. doi: 10.1007/BF03401897
Qiu, Z., He, Y., Ming, H., Lei, S., Leng, Y., and Xia, Z. Y. (2019). Lipopolysaccharide (Lps) aggravates high glucose- and hypoxia/Reoxygenation-induced injury through activating Ros-dependent Nlrp3 Inflammasome-mediated Pyroptosis in H9C2 cardiomyocytes. J. Diabetes Res. 2019, 1–12. doi: 10.1155/2019/8151836
Qu, X. X., Cai, J., Li, M. J., Chi, Y. N., Liao, F. F., Liu, F. Y., et al. (2009). Role of the spinal cord Nr2B-containing Nmda receptors in the development of neuropathic pain. Exp. Neurol. 215, 298–307. doi: 10.1016/j.expneurol.2008.10.018
Quinlan, C. L., Perevoshchikova, I. V., Hey-Mogensen, M., Orr, A. L., and Brand, M. D. (2013). Sites of reactive oxygen species generation by mitochondria oxidizing different substrates. Redox Biol. 1, 304–312. doi: 10.1016/j.redox.2013.04.005
Rahal, A., Kumar, A., Singh, V., Yadav, B., Tiwari, R., Chakraborty, S., et al. (2014). Oxidative stress, prooxidants, and antioxidants: the interplay. Biomed. Res. Int. 2014:761264. doi: 10.1155/2014/761264
Reimer, K. A., and Jennings, R. J. C. (1985). Failure of the xanthine oxidase inhibitor allopurinol to limit infarct size after ischemia and reperfusion in dogs 71, 1069–1075. doi: 10.1161/01.CIR.71.5.1069
Ren, C., Gao, M., Dornbos, D. 3rd, Ding, Y., Zeng, X., Luo, Y., et al. (2011). Remote ischemic post-conditioning reduced brain damage in experimental ischemia/reperfusion injury. Neurol. Res. 33, 514–519. doi: 10.1179/016164111X13007856084241
Rong, Y., Fan, J., Ji, C., Wang, Z., Ge, X., Wang, J., et al. (2022). Usp11 regulates autophagy-dependent ferroptosis after spinal cord ischemia-reperfusion injury by deubiquitinating Beclin 1. Cell Death Differ. 29, 1164–1175. doi: 10.1038/s41418-021-00907-8
Ryter, S. W., Kim, H. P., Hoetzel, A., Park, J. W., Nakahira, K., Wang, X., et al. (2007). Mechanisms of cell death in oxidative stress. Antioxid. Redox. Signal 9, 49–89. doi: 10.1089/ars.2007.9.49
Salzano, S., Checconi, P., Hanschmann, E.-M., Lillig, C. H., Bowler, L. D., Chan, P., et al. (2014). Linkage of inflammation and oxidative stress via release of glutathionylated peroxiredoxin-2, which acts as a danger signal. Proc. Natl. Acad. Sci. U.S.A. 111, 12157–12162. doi: 10.1073/pnas.1401712111
Schroeder, G. D., Kwon, B. K., Eck, J. C., Eck, J. C., Savage, J. W., Hsu, W. K., et al. (2014). Survey of Cervical Spine Research Society members on the use of high-dose steroids for acute spinal cord injuries. Spine 39, 971–977. doi: 10.1097/BRS.0000000000000297
Sepehr, A., Motaghinejad, M., Heysieattalab, S., and Safari, S. (2020). Minocycline may be useful to prevent or treat methamphetamine-induced neural cell death: hypothetic role of Autophagia and apoptosis signaling pathway. Adv. Biomed. Res. 9:7. doi: 10.4103/abr.abr_258_19
Sies, H., and Jones, D. P. (2020). Reactive oxygen species (Ros) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 21, 363–383. doi: 10.1038/s41580-020-0230-3
Sironi, L., Cimino, M., Guerrini, U., Calvio, A. M., Lodetti, B., Asdente, M., et al. (2003). Treatment with statins after induction of focal ischemia in rats reduces the extent of brain damage. Arterioscler. Thromb. Vasc. Biol. 23, 322–327. doi: 10.1161/01.atv.0000044458.23905.3b
Skulachev, V. P. (1998). Cytochrome c in the apoptotic and antioxidant cascades. FEBS Lett. 423, 275–280. doi: 10.1016/S0014-5793(98)00061-1
Smith, P. D., Puskas, F., Meng, X., Lee, J. H., Cleveland, J. C., Weyant, M. J., et al. (2012). The evolution of chemokine release supports a bimodal mechanism of spinal cord ischemia and reperfusion injury. Circulation 126, S110–S117. doi: 10.1161/CIRCULATIONAHA.111.080275
Sohn, H. M., Hwang, J. Y., Ryu, J. H., Kim, J., Park, S., Park, J. W., et al. (2017). Simvastatin protects ischemic spinal cord injury from cell death and cytotoxicity through decreasing oxidative stress: in vitro primary cultured rat spinal cord model under oxygen and glucose deprivation-reoxygenation conditions. J. Orthop. Surg. Res. 12:36. doi: 10.1186/s13018-017-0536-9
Son, Y., Cheong, Y. K., Kim, N. H., Chung, H. T., Kang, D. G., and Pae, H. O. (2011). Mitogen-activated protein kinases and reactive oxygen species: how can Ros activate Mapk pathways? J. Signal Transd. 2011, 1–6. doi: 10.1155/2011/792639
Su, B., Dong, H., Ma, R., Zhang, X., Ding, Q., and Xiong, L. (2009). Cannabinoid 1 receptor mediation of spinal cord ischemic tolerance induced by limb remote ischemia preconditioning in rats. J. Thorac. Cardiovasc. Surg. 138, 1409–1416. doi: 10.1016/j.jtcvs.2009.07.014
Su, L. J., Zhang, J. H., Gomez, H., Murugan, R., Hong, X., Xu, D., et al. (2019). Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid. Med. Cell. Longev. 2019, 1–13. doi: 10.1155/2019/5080843
Sun, F., Zhang, H., Shi, J., Huang, T., and Wang, Y. (2021). Astragalin protects against spinal cord ischemia reperfusion injury through attenuating oxidative stress-induced necroptosis. Biomed. Res. Int. 2021:7254708. doi: 10.1155/2021/7254708
Svensson, E. C., Madar, A., Campbell, C. D., He, Y., Sultan, M., Healey, M. L., et al. (2022). Tet2-driven clonal hematopoiesis and response to canakinumab: an exploratory analysis of the cantos randomized clinical trial 7, 521–528. doi: 10.1001/jamacardio.2022.0386
Tang, D., Kang, R., Berghe, T. V., Vandenabeele, P., and Kroemer, G. (2019). The molecular machinery of regulated cell death. Cell Res. 29, 347–364. doi: 10.1038/s41422-019-0164-5
Taoka, Y., Okajima, K., Uchiba, M., Murakami, K., Kushimoto, S., Johno, M., et al. (1997). Gabexate mesilate, a synthetic protease inhibitor, prevents compression-induced spinal cord injury by inhibiting activation of leukocytes in rats. Crit. Care Med. 25, 874–879. doi: 10.1097/00003246-199705000-00026
Trivedi, A., Olivas, A. D., and Noble-Haeusslein, L. J. (2006). Inflammation and spinal cord injury: infiltrating leukocytes as determinants of injury and repair processes. Clin. Neurosci. Res. 6, 283–292. doi: 10.1016/j.cnr.2006.09.007
Wang, X., De Rivero, V. J. P., Wang, H., Diaz, P., German, R., Marcillo, A. E., et al. (2012). Activation of the nuclear factor E2-related factor 2/antioxidant response element pathway is neuroprotective after spinal cord injury. J. Neurotrauma 29, 936–945. doi: 10.1089/neu.2011.1922
Wang, N., Yu, H., Song, Q., Mao, P., Li, K., and Bao, G. (2021). Sesamol-loaded stearic acid-chitosan nanomicelles mitigate the oxidative stress-stimulated apoptosis and induction of pro-inflammatory cytokines in motor neuronal of the spinal cord through Nf-ĸb signaling pathway. Int. J. Biol. Macromol. 186:23. doi: 10.1016/j.ijbiomac.2021.06.171
Wu, F., Lin, Y., Xiao, L., Chen, Q., Lin, F., and Li, R. (2024). Administration with curcumin alleviates spinal cord ischemia-reperfusion injury by regulating anti-oxidative stress and microglia activation-mediated neuroinflammation via Nrf2/Nf-κB axis. In Vitro Cell. Dev. Biol. Anim. 60, 172–182. doi: 10.1007/s11626-023-00846-3
Wynn, M. M., and Acher, C. W. (2014). A modern theory of spinal cord ischemia/injury in thoracoabdominal aortic surgery and its implications for prevention of paralysis. J. Cardiothorac. Vasc. Anesth. 28, 1088–1099. doi: 10.1053/j.jvca.2013.12.015
Xie, L., Yu, S., Yang, K., Li, C., and Liang, Y. (2017). Hydrogen sulfide inhibits Autophagic neuronal cell death by reducing oxidative stress in spinal cord ischemia reperfusion injury. Oxidative Med. Cell. Longev. 2017:8640284. doi: 10.1155/2017/8640284
Xiong, L., Zhu, Z., Dong, H., Hu, W., Hou, L., and Chen, S. (2000). Hyperbaric oxygen preconditioning induces neuroprotection against ischemia in transient not permanent middle cerebral artery occlusion rat model. Chin. Med. J. 113, 836–839
Xu, T., Gao, P., Huang, Y., Wu, M., Yi, J., Zhou, Z., et al. (2023). Git1-Pgk1 interaction achieves self-protection against spinal cord ischemia-reperfusion injury by modulating Keap1/Nrf2 signaling. Redox Biol. 62:102682. doi: 10.1016/j.redox.2023.102682
Yang, S., Li, H., Tang, L., Ge, G., Ma, J., Qiao, Z., et al. (2015). Apelin-13 protects the heart against ischemia-reperfusion injury through the risk-Gsk-3β-mptp pathway. Arch. Med. Sci. 11, 1065–1073. doi: 10.5114/aoms.2015.54863
Yang, S., Lian, G. J. M., and Biochemistry, C. (2020). Ros and diseases: role in metabolism and energy supply. Mol. Cell Biochem. 467, 1–12. doi: 10.1007/s11010-019-03667-9
Yavuz, C., Demirtas, S., Guclu, O., Karahan, O., Caliskan, A., Yazici, S., et al. (2013). Rosuvastatin may have neuroprotective effect on spinal cord ischemia reperfusion injury. CNS Neurol. Disord. Drug Targets 12, 1011–1016. doi: 10.2174/18715273113129990085
Ye, J. T., Li, F. T., Huang, S. L., Xue, J. L., Aihaiti, Y., Wu, H., et al. (2019). Effects of ginsenoside Rb1 on spinal cord ischemia-reperfusion injury in rats. J. Orthop. Surg. Res. 14:259. doi: 10.1186/s13018-019-1299-2
Yu, Q., Huang, J., Hu, J., and Zhu, H. (2016). Advance in spinal cord ischemia reperfusion injury: blood-spinal cord barrier and remote ischemic preconditioning. Life Sci. 154:34. doi: 10.1016/j.lfs.2016.03.046
Yu, H., Shi, L., Zhao, S., Sun, Y., Gao, Y., Sun, Y., et al. (2016). Triptolide attenuates myocardial ischemia/reperfusion injuries in rats by inducing the activation of Nrf2/ho-1 defense pathway. Cardiovasc. Toxicol. 16, 325–335. doi: 10.1007/s12012-015-9342-y
Yu, M., Wang, Z., Wang, D., Aierxi, M., Ma, Z., and Wang, Y. (2023). Oxidative stress following spinal cord injury: from molecular mechanisms to therapeutic targets. J. Neurosci. Res. 101, 1538–1554. doi: 10.1002/jnr.25221
Yu, S., Xie, L., Liu, Z., Li, C., and Liang, Y. (2019). Mln4924 exerts a neuroprotective effect against oxidative stress via Sirt1 in spinal cord ischemia-reperfusion injury. Oxidative Med. Cell. Longev. 2019:7283639. doi: 10.1155/2019/7283639
Yuan, L., Xu, J., Qin, F., Yi, X., Zhong, Z., Gu, X., et al. (2023). Usp18 overexpression protects against spinal cord ischemia/reperfusion injury via regulating autophagy. Neurosci. Lett. 810:137359. doi: 10.1016/j.neulet.2023.137359
Yunoki, M., Kanda, T., Suzuki, K., Uneda, A., Hirashita, K., and Yoshino, K. (2017). Ischemic tolerance of the brain and spinal cord: a review. Neurol. Med. Chir. 57, 590–600. doi: 10.2176/nmc.ra.2017-0062
Zhang, Y., Deng, Q., Hong, H., Qian, Z., Wan, B., and Xia, M. (2024). Caffeic acid phenethyl ester inhibits neuro-inflammation and oxidative stress following spinal cord injury by mitigating mitochondrial dysfunction via the Sirt1/Pgc1α/Drp1 signaling pathway. J. Transl. Med. 22:304. doi: 10.1186/s12967-024-05089-8
Zhang, S., Tang, M.-B., Luo, H.-Y., Shi, C. H., and Xu, Y. M. (2017). Necroptosis in neurodegenerative diseases: a potential therapeutic target. Cell Death Dis. 8:e2905-e. doi: 10.1038/cddis.2017.286
Zhang, Z., Yang, K., Mao, R., Zhong, D., Xu, Z., Xu, J., et al. (2022). Ginsenoside Rg1 inhibits oxidative stress and inflammation in rats with spinal cord injury via Nrf2/ho-1 signaling pathway. Neuroreport 33, 81–89. doi: 10.1097/WNR.0000000000001757
Zhao, J., Cheng, W., He, X., Liu, Y., Li, J., Sun, J., et al. (2018). Chronic obstructive pulmonary disease molecular subtyping and pathway deviation-based candidate gene identification. Cell J. 20, 326–332. doi: 10.22074/cellj.2018.5412
Zhao, M., Zhu, P., Fujino, M., Nishio, Y., Chen, J., Ito, H., et al. (2016). 5-Aminolevulinic acid with sodium ferrous citrate induces autophagy and protects cardiomyocytes from hypoxia-induced cellular injury through Mapk-Nrf-2-ho-1 signaling cascade. Biochem. Biophys. Res. Commun. 479, 663–669. doi: 10.1016/j.bbrc.2016.09.156
Zheng, W., Liu, B., and Shi, E. (2021). Perillaldehyde alleviates spinal cord ischemia-reperfusion injury via activating the Nrf2 pathway. J. Surg. Res. 268, 308–317. doi: 10.1016/j.jss.2021.06.055
Zhong, Z., Liang, S., Sanchez-Lopez, E., He, F., Shalapour, S., Lin, X. J., et al. (2018). New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 560, 198–203. doi: 10.1038/s41586-018-0372-z
Zhou, L., Ouyang, L., Lin, S., Chen, S., Liu, Y. J., Zhou, W., et al. (2018). Protective role of β-carotene against oxidative stress and neuroinflammation in a rat model of spinal cord injury. Int. Immunopharmacol. 61:92. doi: 10.1016/j.intimp.2018.05.022
Zhou, Y., Wu, R., Wang, X., Bao, X., and Lu, C. (2022). Roles of necroptosis in alcoholic liver disease and hepatic pathogenesis. Cell Prolif. 55:e13193. doi: 10.1111/cpr.13193
Zhu, Y. T., Wan, C., Lin, J. H., Hammes, H. P., and Zhang, C. (2022). Mitochondrial oxidative stress and cell death in Podocytopathies. Biomol. Ther. 12:403. doi: 10.3390/biom12030403
Zotta, A., O'neill, L. A. J., and Yin, M. (2024). Unlocking potential: the role of the electron transport chain in immunometabolism. Trends Immunol. 45, 259–273. doi: 10.1016/j.it.2024.02.002
Glossary
SCIRI - Spinal cord ischemia/reperfusion injury
ROS - Reactive oxygen
BSCB - Blood-spinal cord barrier
O2 - - Superoxide anion
H2O2 - Hydrogen peroxide
OH− - hydroxyl radical
I/R - Ischemia reperfusion
ETC - Electron transport chain
SOD - superoxide dismutase
XDH - xanthine dehydrogenase
XO - Xanthine oxidase
NADPH - Phosphamide adenine dinucleotide
PRDX2 - Peroxidase 2
mPTP - Endometrial permeability transition pore
ΔΨm - membrane potential
Cyt C - Cytochrome C
GSDMD - gasdermin-D
Apaf-1 - Apoptotic protease activator-1
mtROS - Mitochondrial reactive oxygen species
HIF-1α - Hypoxia-inducible factor-1α
GSH - Glutathione
GPX4 - Glutathione peroxidase 4
Nrf-2 - Nuclear factor erythrocyte 2-related factor 2
ARE - Antioxidant response element
Keap1 - Kelch-like epichlorohydrin-associated protein-1
HO-1 - Heme oxygenase-1
NQO1 - Quinone oxidoreductase-1
GPX - glutathion peroxidase
NF-κB - Nuclear factor-κB
MDA - Malonaldehyde
RIPC - Remote ischemic preconditioning
MAPK - mitogen activated protein kinase
MMPs - Matrix metalloproteinase
ERK - Extracellular signal-regulated kinase
JNK - c-Jun NH 2-terminal kinase
Sal - salidroside
BA - Biochanin
AST - astaxanthin
GRb1 - Ginsenoside Rb1
MAP3K - APK kinase kinases
Keywords: spinal cord ischemia/reperfusion injury (SCIRI), oxidative stress, inflammation, cell death, mechanism
Citation: Xing Y, Xiao Y-z, Zhao M, Zhou J-j, Zhao K and Xiao C-l (2025) The role of oxidative stress in spinal cord ischemia reperfusion injury: mechanisms and therapeutic implications. Front. Cell. Neurosci. 19:1590493. doi: 10.3389/fncel.2025.1590493
Edited by:
Elzbieta Salinska, Polish Academy of Sciences, PolandReviewed by:
Mingzi Zhang, University of Southern California, United StatesElle Scheijen, KU Leuven, Belgium
Copyright © 2025 Xing, Xiao, Zhao, Zhou, Zhao and Xiao. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Kai Zhao, Y3Jhenlfa2FpQDE2My5jb20=; Chun-lin Xiao, MTIzNjYzMzQwQHFxLmNvbQ==
†These authors share first authorship