Open Access

NETosis in myocardial ischemia‑reperfusion injury: From mechanisms to therapies (Review)

  • Authors:
    • Ziyang Zhang
    • Yanxin Wang
    • Tie Li
    • Hongfeng Wang
  • View Affiliations

  • Published online on: May 13, 2025     https://doi.org/10.3892/br.2025.1991
  • Article Number: 113
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The present review describes the mechanisms of NETosis and its role in myocardial ischemia‑reperfusion injury (MIRI), focusing on the release of neutrophil extracellular traps (NETs) by activated neutrophils. NETs, composed of depolymerized chromatin and granule proteins, are crucial for pathogen entrapment, infection control and immune regulation. However, NET formation, linked to neutrophil death (NETosis), exacerbates MIRI by promoting inflammation and tissue damage. To address therapeutic strategies for NETosis in MIRI, several potential clinically significant approaches were explored, including peptidylarginine deaminase 4 inhibition, DNase therapy, antioxidants, inflammation modulation, and antithrombotic treatments, which not only provide novel diagnostic biomarkers and therapeutic targets in MIRI, but are also expected to improve patient prognosis and advance the development of personalised medicine.

1. Introduction

Neutrophils are the first immune cells to arrive at the site of inflammation, where they contribute to the destruction and containment of pathogenic microorganisms through phagocytosis, the release of bactericidal substances and the production of pro-inflammatory cytokines (1). Neutrophil activation is initiated by both microbial and endogenous stimuli, resulting in the release of chromatin and granule proteins, including neutrophil elastase (NE), myeloperoxidase (MPO) and other components such as histone G and defensins. These elements collectively form extracellular structures termed neutrophil extracellular traps (NETs) (2). The process, termed NETosis, is integral to the pathophysiology of myocardial ischemia-reperfusion injury (MIRI) by modulating mechanisms such as reactive oxygen species (ROS) production, inflammation and energy metabolism, thereby contributing to the advancement of myocardial injury (3,4). NETosis plays an important role in the physiopathology of aseptic inflammatory conditions, such as MIRI, by affecting ROS production, inflammation, energy metabolism and other such mechanisms, but the specific mechanisms and directions of its action are not yet fully understood.

Despite the development of more efficient and effective reperfusion techniques, including percutaneous coronary intervention and novel antiplatelet and antithrombotic therapies, effective treatments to prevent MIRI remain limited (5,6). Recent data show that cardiovascular disease is responsible for ~20% of all deaths worldwide each year (7). MIRI refers to myocardial injury that occurs after rapid restoration of blood flow in the ischemic area and accounts for 50% of the final infarct size (8). These injuries can be devastating, leading to fatal myocardial damage, myocardial dysfunction, vascular injury and tissue edema (9). The pathological mechanisms underlying these injuries include apoptosis, autophagy, endoplasmic reticulum stress, ferroptosis, pyroptosis, and ultimately, exacerbated oxidative stress, calcium imbalance, activation of inflammatory cascades, endothelial dysfunction and microvascular injury. Recently, it has been shown that specific biomarkers (such as DNA, histones and NE) that are released during NETosis may provide novel biomarkers for the early diagnosis of MIRI (10). By regulating the NETosis process, the development of novel therapeutic strategies, such as inhibiting the formation of NETosis or promoting its clearance, may help to mitigate MIRI and improve the prognosis of patients.

The present review systematically compiles and analyzes the research progress on the role of NETosis in MIRI in recent years, focusing on the formation mechanism of NETosis, its dynamic changes during myocardial ischemia-reperfusion and the potential therapeutic mechanism of NETosis-targeted therapy for MIRI.

2. NETosis formation mechanisms

To date, the formation of NETosis has been classified into two forms: i) Suicidal NETosis; and ii) vital NETosis (11). Regardless of the stimulus, NETosis formation involves the cellular process of chromatin decondensation, which is essential for the release of NETs (12).

Suicidal NETosis

Suicidal NETosis denotes a mechanism of NET formation that is concomitant with neutrophil apoptosis. Upon activation by stimuli including phorbol ester, specific autoantibodies, immune complexes or conditions characterized by elevated calcium ion (Ca²+) concentrations, neutrophils initiate the NADPH oxidase complex (NOX), leading to the production of ROS such as superoxide anion, hydroxyl radical and hydrogen peroxide (13,14). These ROS are highly reactive and play a crucial role in facilitating NOX-dependent NET formation (15). The release of ROS activates peptidylarginine deaminase 4 (PAD4), the only nuclear-localized isoenzyme in the PAD family, which is expressed in both the nucleus and cytoplasmic granules of immune cells (16). PAD4 facilitates the citrullination of core and linker histones, which diminishes their affinity for DNA, thereby promoting chromatin decondensation and the unfolding of densely packed chromatin structures (17,18).

Activation of PAD4 also triggers the translocation of NE and MPO from azurophilic granules to the nucleus (19), which further enhances histone H3 citrullination (H3Cit) and chromatin decondensation, a pivotal process in the formation of NETs (20,21). Consequently, H3Cit is considered a specific and well-established marker for NETs (22). With chromatin decondensation and nuclear membrane rupture, the decondensed chromatin mixes with granule proteins and is released into the cytoplasm (23). Eventually, the cytoplasmic membrane leaks, and the modified chromatin is expelled from the neutrophil, forming extracellular NETs in a meshwork structure. NETs are released into the extracellular space 3-8 h after neutrophil activation, leading to cell death due to membrane disintegration (24). Plasma levels of MPO have been shown to be positively correlated with the risk of coronary artery disease (25), and MPO-DNA complexes are associated with severe adverse cardiovascular events (26).

Moreover, the serum concentrations of critical markers of NETs, such as double-stranded DNA (dsDNA), MPO and NE, are markedly elevated in patients with coronary atherosclerotic disease (27). Additionally, extracellular H3Cit exhibits significant cytotoxicity, contributing to tissue damage and thereby elevating the risk of mortality among patients with cardiovascular disease (28). These observations indicate that MPO, NE and H3Cit are integral to NET formation, further implicating NETosis in the pathophysiological processes underlying MIRI.

Vital NETosis

Vital NET formation represents a type of NETosis that transpires shortly after neutrophil activation, distinguished by the generation of NETs without compromising the structural integrity of the neutrophil (29). In contrast to suicidal NETosis, this process involves the encapsulation of modified chromatin within vesicles that are secreted by the nucleus and subsequently expelled from the cell, thereby preserving neutrophil integrity (30). Consequently, neutrophils remain intact and retain the ability to perform additional functions (Fig. 1).

3. Pathogenesis of NETosis in MIRI

Myocardial ischemia is primarily attributed to atherosclerosis, thrombosis, insufficient coronary blood supply and elevated myocardial oxygen demand, with atherosclerosis and luminal thrombosis identified as the predominant mechanisms.

Pathogenesis of NETosis during ischemia

NETosis is a critical factor in the pathophysiology of myocardial ischemia, particularly within pro-inflammatory and pro-thrombotic contexts. NETs are observed to form in regions of elevated cholesterol within atherosclerotic lesions. Elevated blood cholesterol levels result in the deposition of cholesterol crystals within the vessel walls, which are subsequently recognized and phagocytosed by macrophages (31). This process induces lysosomal damage and activates the NLR family pyrin domain containing 3 (NLRP3) inflammasome within macrophages (32). PAD4 plays a critical role in the pathogenesis of atherosclerosis by regulating apoptosis-associated speck-like proteins containing a caspase recruitment domain (ASC proteins) and NLRP3 protein levels, thereby facilitating the assembly of NLRP3 inflammasomes (33). Upon activation, these multiprotein complexes lead to the production of pro-inflammatory cytokines such as interleukin (IL)-1β and IL-18 (34,35).

These factors recruit more immune cells to the vascular endothelium, which exacerbates the inflammatory response and further activates neutrophils leading to NETosis (36). The release of NETs can exacerbate tissue damage and contribute to a pro-inflammatory milieu. Activated neutrophils secrete cytokines and chemokines, which further recruit additional immune cells and amplify the inflammatory response. This phenomenon is particularly evident in conditions such as acute myocardial infarction, where enhanced neutrophil-platelet interactions lead to increased thrombus formation (37). Thrombus formation plays a critical role in the pathogenesis of myocardial ischemia. For example, Zhou et al (38) identified elevated levels of NETs in coronary thrombosis among patients with ST-segment elevated myocardial infarction. Neutrophils retained within thrombi play a crucial role in initiating the endogenous coagulation cascade through the release of NETs, which facilitate the activation of coagulation factor XII. This process can subsequently lead to thrombin generation and platelet activation (39). The structural network of NETs further supports the deposition of platelet adhesion molecules and promotes thrombin-mediated fibrin formation (40).

Pathogenesis of NETosis in MIRI

Oxidative stress. Myocardial energy supply is primarily derived from fatty acid and glucose metabolism (41), and neutrophils primarily rely on glycolysis, rather than oxidative phosphorylation, to meet their energy demands (42). During ischemia-reperfusion, due to a lack of synthetic substrates, mitochondrial dysfunction and calcium overload, cardiomyocytes rely on an attenuated glycolytic pathway for energy production for 0.5-2 h (43). When endogenous cellular free radical scavenging enzymes, such as superoxide dismutase, catalase and glutathione peroxidase, are unable to neutralize ROS effectively, this leads to an imbalance between the oxidation system and antioxidant enzymes (44,45). This in turn disrupts cellular membrane integrity, altering its fluidity and permeability, leading to further membrane damage, cellular swelling and functional impairment (46). NETs play a significant role in the pathophysiological process of MIRI, particularly through NOX-dependent pathways. The restoration of blood flow following ischemia induces a severe inflammatory response, characterized by neutrophil activation. These activated neutrophils release NETs, which can trap pathogens, but also contribute to myocardial tissue damage and inflammation (47). Upon stimulation, neutrophils activate the NOX complex, initiating a pathway referred to as NOX-dependent NETosis. In this pathway, NOX catalyzes the production of ROS, which are crucial for the formation of NETs. This oxidative burst not only facilitates NET formation but also intensifies oxidative stress within myocardial tissues, thereby contributing to additional cellular damage and inflammation (48,49). The formation of NETs requires an increase in ATP availability, which further reduces the oxygen supply to cardiomyocytes, indirectly diminishing glycolytic efficiency (50). In addition, the excessive accumulation of NETs within the ischemic myocardium facilitates the formation of microthrombi, thereby obstructing blood flow and contributing to the ‘no-reflow’ phenomenon, wherein myocardial tissue remains insufficiently perfused despite the restoration of blood supply (51).

Calcium ions are crucial for the maintenance of normal cardiomyocyte function, with mitochondrial Ca2+ levels modulating the rate of oxidative metabolism to align with ATP depletion in the cytoplasm. This regulation ensures that cardiomyocytes receive adequate energy to sustain their normal systolic and diastolic functions (52). During reperfusion injury, cardiomyocytes exacerbate injury by reversing the Na+/Ca²+ exchanger, leading to sodium efflux and subsequent intracellular calcium overload. Excessive calcium efflux contributes to mitochondrial permeability transition pore opening and mitochondrial ROS production (53,54). In a high Ca²+ concentration environment, calcium ions can directly or indirectly activate NOX by altering its conformation or active state, triggering NOX activation and promoting NETosis (55). This pathway is referred to as NOX-independent NETosis. Research has shown that direct activation of small-conductance calcium-activated potassium (SK) channels can induce NETosis, with the involvement of SK3 (56,57).

Inflammation. During reperfusion, cells that have been compromised by ROS release endogenous damage-associated molecular patterns (DAMPs) and inflammatory cytokines, including tumor necrosis factor-α and IL-1β (58). The release of these cytokines further recruits and activates inflammatory cells, such as neutrophils and macrophages, thereby intensifying the inflammatory response and perpetuating a deleterious cycle of ROS production. Mitochondrial DNA (mtDNA) fragments are identified as DAMPs by pattern recognition receptors of the immune system, including Toll-like receptors (TLRs) and NOD-like receptors, thereby initiating inflammatory responses (59). mtDNA activates immune responses via CpG/TLR9 interactions, promoting mitogen-activated protein kinase (MAPK) signaling and thereby exacerbating MIRI (60,61). Additionally, the recognition of DNA within NETs by TLR9 activates plasmacytoid dendritic cells, which perpetuate vascular inflammation (62).

Extracellular histones, as critical DAMPs, contribute to myocardial injury during MIRI (63). Extracellular histones exacerbate the inflammatory response by activating TLR2 and TLR4, leading to the production of pro-inflammatory cytokines. Histone interactions with T cells promote Th17 cytokine production, further amplifying the inflammatory state (64-67). High mobility group protein B1, a critical DAMP released from necrotic cardiac tissue following prolonged ischemic injury, interacts with the receptor for advanced glycation end-products. This interaction facilitates the recruitment of a substantial number of neutrophils to the injured myocardium, thereby promoting the production of IL-6 through signaling pathways such as MAPK and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), along with the release of other inflammatory mediators, ultimately triggering NET formation (68). The inflammatory pathway factors involved in NETosis during reperfusion injury and potential treatments are described in Table I.

Table I

Inflammatory pathway agents involved in NETosis during reperfusion injury and potential therapeutic approaches.

Table I

Inflammatory pathway agents involved in NETosis during reperfusion injury and potential therapeutic approaches.

First author/s, yearDrug/moleculeType of experimentPathway of actionOutcome(Refs.)
Xu et al, 2019AdenosineClinical trialAdenosine exerts anti-inflammatory effects by binding to its receptors and inhibiting neutrophil activation and NETosis.HNE-DNA ELISA assay(113)
Xie et al, 2018Cpg-ODNAnimalCpG-ODN treatment acts on the TLR9-p38 MAPK signaling pathway, leading to increased cardiomyocyte damage and inflammatory response.Myocardial infarction(61)
Clark et al, 2007HistoneClinical trialExogenous chromatin acts as a key driver to amplify the inflammatory response in aseptic diseases and infections by enhancing TLR4 binding.Expression of IL-1β mRNA(67)
Sayegh et al, 2023Polyvinyl alcohol (PEG-4MAL) hydrogelAnimalDelivery of CD39 and CD73 via hydrogel can modulate the innate immune response and thus protect cardiac function. Echocardiography(116)
Yu et al, 2022S100A8/A9Clinical trialS100A8/A9 promotes granulopoiesis after myocardial infarction by inducing NETosis.NETosis marker(86)
Mangold et al, 2019Monocyte subpopulationClinical trialExogenous chromatin may promote the differentiation response of monocytes and reduce their CX3CR1 expression at the site of the lesion, thus affecting the migration of non-classical monocytes to ischemic tissue and myocardial healing.CX3CR1 expression level(78)
Savchenko et al, 2014DNase I and rhADAMTS13AnimalTargeting extracellular DNA and VWF may be a novel cardioprotective therapeutic strategy. Echocardiography(98)
Ge et al, 2015DNase and rt-PAAnimalCombination therapy with DNase I and rt-PA effectively reduces NET density, improves coronary microcirculation patency and limits the extent of myocardial infarction.Myocardial infarction(99)

[i] HNE, human neutrophil elastase; Cpg-ODN, CpG-oligodeoxynucleotide; TLR, Toll-like receptor; CX3CR1, CX3C motif chemokine receptor 1; ADAMTS13, recombinant ADAM metallopeptidase with thrombospondin type 1 motif 13; VWF, von Willebrand factor; rt-PA, recombinant tissue-type plasminogen activator; NET, neutrophil extracellular trap.

Endothelial dysfunction. During myocardial ischemia-reperfusion, endothelial cell damage occurs, accompanied by a reduction in antioxidant activity and a decrease in nitric oxide synthesis and release, culminating in impaired vasodilation (69,70). Myocardial injury is exacerbated by coronary artery constriction, neutrophil aggregation and abnormal platelet responses, resulting in microvascular spasm and restenosis (71,72).

Damaged endothelial cells release a variety of pro-inflammatory factors, such as IL-1β and IL-6, which further amplify the inflammatory response (73). NETs can induce NF-κB-dependent endothelial angiogenesis, altering the plaque microenvironment and promoting the development of unstable plaques (74,75). Angiotensin II has been demonstrated to induce NETosis through both the ROS/PAD4 pathway and autophagy-dependent mechanisms (76).

Involvement of NETosis in myocardial fibrosis after reperfusion injury

Subsequent to cardiomyocyte injury and necrosis, there is a pronounced accumulation of extracellular matrix components, accompanied by the differentiation of cardiac fibroblasts into myofibroblasts under stress conditions. These myofibroblasts exhibit enhanced proliferation and secretion activities, culminating in the development of cardiac fibrosis (77). NETs are intricately linked to inflammatory processes and may significantly influence the fibroblastic response during myocardial injury. Additionally, non-classical monocytes, integral components of the immune system, play a crucial role in myocardial healing. It has been observed that overproduction of NETs can reduce the expression of CX3C chemokine receptor 1 on non-classical monocytes at the site of injury. This reduction may impair the migration of non-classical monocytes to ischemic tissues, resulting in delayed or compromised myocardial healing (78).

The release of NETs also stimulates the proliferation and activation of cardiac fibroblasts, which secrete collagen and other fibrosis-associated proteins, thus contributing to the progression of myocardial fibrosis (79). NETs recognize and bind endocytosed dsDNA via TLR9, which activates the myeloid differentiation factor 88 and, in turn, triggers the NF-κB signaling pathway (80,81). Upon activation of the TLR-NF-κB signaling axis, myofibroblast differentiation is promoted by regulating the expression of genes such as transforming growth factor-β, collagen I (a major component of the cardiac extracellular matrix) and α-smooth muscle actin, a signature protein of myofibroblasts. These changes are accompanied by stress fiber formation and contribute to the progression of myocardial fibrosis (82).

4. NETosis predicts the prognosis in MIRI

During myocardial ischemia-reperfusion, excessive production of NETs can facilitate thrombosis and worsen microvascular obstruction, thereby elevating the risk of myocardial infarction. Chang et al (83) concluded that the combined use of circulating cell-free DNA (cfDNA) and creatine kinase-MB may enhance the diagnostic accuracy and sensitivity for acute myocardial ischemia, enabling a more comprehensive evaluation of the condition of patients. Recent research has demonstrated that in patients with acute myocardial ischemia, plasma cfDNA is predominantly released through NETosis, further substantiating its potential as a diagnostic marker for acute myocardial ischemia (84).

Additionally, markers such as platelet count, MPO-DNA complexes, NE-DNA complexes, H3Cit and soluble platelet selectin have been identified as independent predictors of major adverse cardiovascular events 1 year after myocardial infarction (85). A prospective clinical study demonstrated that plasma levels of bactericidal/permeability-increasing protein (BPI) in patients experiencing acute myocardial ischemia exhibited a positive correlation with IL-1β, high-sensitivity C-reactive protein, MPO-DNA and S100A8/A9. Consequently, BPI may serve as a novel biomarker for myocardial ischemia, with its primary mechanism potentially involving the mediation of NET formation through interactions between platelets and neutrophils (86,87).

Animal studies have found that colchicine alleviates inflammation and cardiac remodeling after acute myocardial infarction by inhibiting the formation of NETs, thereby improving cardiac function (88). Additionally, colchicine reduces microvascular obstruction by suppressing the proliferation of neutrophils in the bone marrow and their migration to the ischemia-reperfusion injury area (89). Building on these findings from animal studies, clinical research has further validated the potential application of colchicine in cardiovascular diseases. Clinical studies have shown that low-dose colchicine provides significant clinical benefits in post-myocardial infarction patients, reducing the incidence of cardiovascular events with a favorable safety profile, suggesting its potential applicability in MIRI (90,91).

5. Targeting MIRI with NETosis

NETs may exert a dual role in the management of MIRI. Modulating NETs represents a potentially significant therapeutic strategy for the treatment of MIRI. By inhibiting the formation of NETs or facilitating their degradation, it may be possible to mitigate myocardial tissue damage and enhance tissue repair. There are no clinical or randomized controlled trials on the mechanism of NETosis in MIRI, and more basic research is needed to support this area of study. However, clinical studies on NETosis in other diseases, such as pneumonia and cancer, offer valuale insights (92-95).

PAD4

Considering that NETosis is primarily reliant on PAD4, targeting PAD4 represents a promising therapeutic strategy. The inhibitor GSK484 has demonstrated significant attenuation of MIRI in an animal model (96). Additionally, BB-CLA, a potent PAD4 inhibitor, effectively prevented the formation and release of NETs induced by polyinosinic acid-polycytidylic acid, a synthetic analog of viral double-stranded RNA (97). Additionally, inhibition of von Willebrand factor (VWF) activity, which mediates leukocyte recruitment, also affects PAD4 activity and promotes NET formation, suggesting that drugs targeting both VWF and NETs may offer a novel therapeutic approach to reduce ischemia-related myocardial injury (98). Currently, studies on the combination therapy of NETosis in MIRI are limited and mainly focused on the animal experimental stage. Combination treatment of DNase I with rhADAMTS13 or rt-PA effectively improved myocardial infarct size and microcirculatory impairment in rats with MIRI (99). Combination of NETosis inhibitors with other anti-inflammatory or antithrombotic drugs may have a synergistic effect and significantly attenuate the pathological damage in MIRI (100). Based on the pathological mechanism of NETosis and related therapeutic targets, it is theorized that combination therapies, such as combining NETosis inhibitors with anti-inflammatory, antioxidant, or antithrombotic drugs may have greater therapeutic potential. The use of combination therapy in MIRI will be further explored in future studies.

The activity of DNase, an enzyme that degrades NETs, plays a crucial role in the clearance of NETs and subsequent modulation of inflammation and thrombosis. Alterations in DNase activity have been linked to ST-segment regression and the onset of myocardial infarction (101). DNase treatment can reduce the cytotoxicity of NETs by degrading their DNA backbone, thus offering potential for reducing thrombotic inflammation (102). However, DNase primarily targets DNA and has limited effects on other NET components, such as histones, which remain capable of causing vascular damage (103). Despite these limitations, dual-active DNase formulations, capable of targeting both DNA and histones, may offer further promise in modulating thrombotic inflammation (104). Recent studies have also suggested that DNase could serve as a prognostic biomarker in patients with atherosclerosis, offering new insights into recurrent ischemic events (105).

Antioxidants

The glutathione metabolic pathway plays a crucial antioxidant role in NETosis. Isocrystalline xanthophylls have been demonstrated to mitigate reperfusion-induced oxidative stress injury and ferroptosis in cardiomyocytes by upregulating glutathione peroxidase 4(106). Studies indicate that the application of leukotriene C4 (LTC4) receptor antagonists can obstruct the LTC4 signaling pathway, thereby inhibiting NETosis and reducing MIRI (107,108). The expression of the transcription factor HIF-1α is upregulated under hypoxic conditions; notably, in myocardial infarction, the expression level of HIF-1α is significantly increased in neutrophils (109). HIF-1α induction may help promote neutrophil survival, mitigate oxidative stress and inhibit NET formation.

Krüppel-like factor 2 (KLF2) is a prominent member of the zinc finger protein transcription factor family. Under homeostatic conditions, KLF2 acts to inhibit HIF-1 signaling. The regulation of the neutrophil KLF2-NETosis-thrombosis pathway may represent a novel therapeutic target for the treatment of reperfusion injury (110). SkQ1, a mitochondria-targeted antioxidant, effectively scavenges ROS within mitochondria during NETosis (111). Furthermore, colchicine treatment has been shown to inhibit the formation of NETs and reduce inflammation by decreasing NOX2/ROS production, which significantly enhances survival rates and improves left ventricular ejection fraction in murine models (88).

Regulation of inflammation

Chemokines such as chemokine (C-X-C motif) ligand 4 (CXCL4) and chemokine (C-C motif) ligand 5 (CCL5) play a pivotal role in the inflammatory response. A synthetic peptide, MKEY, has been shown to block the CCL5-CXCL4 interaction, preventing NET formation by neutrophils in vivo (112). Adenosine, an immunosuppressive molecule, inhibits neutrophil activation and NET release (113). Inhibition of ectonucleotidases CD39 and CD73, which play a role in purinergic signaling, has garnered attention as a potential immunotherapeutic strategy in cancer treatment (114). Previous studies indicate that hydrogel-delivered CD39 and CD73 can reduce NETosis and immune infiltration, improving MIRI outcomes (115,116).

Antithrombotic

Recent research has demonstrated that in the context of coronary thrombosis, NETs primarily activate neutrophils and erythrocytes as the predominant constituents of most coronary thrombi, as opposed to the traditional emphasis on platelet aggregation (117). The inhibition of NET formation through VWF-mediated leukocyte recruitment presents a potentially novel therapeutic approach to mitigating ischemia-related myocardial injury. Research utilizing Lnk gene deletion mice (Lnk-/-) and LNK(TT) [neutrophils derived from human induced pluripotent stem cells with LNK(TT) mutations] indicates that the associated pathologies primarily manifest as abnormalities in platelets and neutrophils. These abnormalities include conditions such as coronary artery disease, thrombocytosis and neutropenia. The Lnk-/- and LNK(TT) models may exhibit heightened sensitivity of neutrophils to external stimuli, such as oxidized phospholipids (OxPL), which subsequently increases their susceptibility to NETosis (118). Consequently, therapeutic interventions targeting OxPL could be advantageous for genetically predisposed human populations. Recombinant tissue plasminogen activator (rt-PA) is a recombinant tissue-type plasminogen activator that restores blood flow in large vessels by dissolving thrombi. rt-PA combined with DNase I can simultaneously solve the problems of microcirculation and obstruction of large vessels, improve reperfusion and alleviate the phenomenon of ‘no reflow’.

6. Conclusion

In the complex pathological process of MIRI, neutrophils exert multifaceted effects. They contribute to cardiomyocyte death, thereby exacerbating myocardial injury, and significantly impact normal vascular function. Additionally, neutrophils promote inflammation by releasing various cytokines and inflammatory mediators, which hinder the post-injury repair process, and their functions and phenotypes can vary depending on the inflammatory environment and stimuli present.

In detailed investigations of molecular mechanisms, PAD4 facilitates the formation of NETs through its interaction with granule proteins, thereby contributing to MIRI. Nonetheless, the excessive production and release of NETs in the context of MIRI present a dualistic effect. While they aid in the clearance of pathogens from the injury site, they also intensify cardiomyocyte apoptosis and the inflammatory response, potentially impairing cardiac function.

Recent therapeutic approaches for MIRI focus on targeting the NETosis process to attenuate or prevent the injury, including the use of PAD4 inhibitors, antioxidants, anti-inflammatory drugs and other related therapies. As research advances, the identification of different neutrophil subtypes with distinct functional properties in the inflammatory response has broadened the scope for potential treatments. Although these NETosis-based treatments show promise in laboratory and animal models, their clinical application requires further validation.

Therapeutic strategies targeting NETosis represent a promising novel approach for the treatment of MIRI. Although significant progress has been made in animal models, the translation of these strategies into clinical practice continues to face substantial challenges. Clinical trial outcomes have often been inconsistent, potentially due to individual variations in susceptibility to reperfusion injury and the limited therapeutic time window for intervention (119). Furthermore, there is a notable lack of high-quality clinical trials to rigorously validate the safety and efficacy of targeted therapies or to elucidate their precise mechanisms of action. Consequently, future research must focus on deepening the understanding of the specific mechanisms by which NETosis contributes to MIRI, as well as on developing more effective and clinically applicable interventions.

In conclusion, targeting NETosis in MIRI represents a promising therapeutic avenue that may significantly improve patient prognosis. By reducing myocardial damage and enhancing repair mechanisms, these therapies are expected to change the way MIRI is managed, providing patients with a better prognosis and quality of life. Future clinical trials should prioritize patient stratification, biomarker development, and evaluation of combination therapies to realise the full clinical potential of NETosis-targeted therapies.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the National Key Research and Development Program of China (grant no. 2022YFC3500705).

Availability of data and materials

Not applicable.

Authors' contributions

ZZ and YW conceived the review topic, conducted the literature search, and wrote the original draft. TL and HW reviewed and edited the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Brinkmann V and Zychlinsky A: Neutrophil extracellular traps: Is immunity the second function of chromatin? J Cell Biol. 198:773–783. 2012.PubMed/NCBI View Article : Google Scholar

2 

Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, Wahn V, Papayannopoulos V and Zychlinsky A: Myeloperoxidase is required for neutrophil extracellular trap formation: Implications for innate immunity. Blood. 117:953–959. 2011.PubMed/NCBI View Article : Google Scholar

3 

Remijsen Q, Kuijpers TW, Wirawan E, Lippens S, Vandenabeele P and Vanden Berghe T: Dying for a cause: NETosis, mechanisms behind an antimicrobial cell death modality. Cell Death Differ. 18:581–588. 2011.PubMed/NCBI View Article : Google Scholar

4 

Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: Recommendations of the Nomenclature committee on cell death 2018. Cell Death Differ. 25:486–541. 2018.PubMed/NCBI View Article : Google Scholar

5 

Reed GW, Rossi JE and Cannon CP: Acute myocardial infarction. Lancet. 389:197–210. 2017.PubMed/NCBI View Article : Google Scholar

6 

Welt FGP, Batchelor W, Spears JR, Penna C, Pagliaro P, Ibanez B, Drakos SG, Dangas G and Kapur NK: Reperfusion injury in patients with acute myocardial infarction: JACC scientific statement. J Am Coll Cardiol. 83:2196–2213. 2024.PubMed/NCBI View Article : Google Scholar

7 

Global Cardiovascular Risk Consortium. Magnussen C, Ojeda FM, Leong DP, Alegre-Diaz J, Amouyel P, Aviles-Santa L, De Bacquer D, Ballantyne CM, Bernabé-Ortiz A, et al: Global effect of modifiable risk factors on cardiovascular disease and mortality. N Engl J Med. 389:1273–1285. 2023.PubMed/NCBI View Article : Google Scholar

8 

Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, et al: Heart disease and stroke statistics-2022 update: A report from the American Heart Association. Circulation. 145:e153–e639. 2022.PubMed/NCBI View Article : Google Scholar

9 

Hausenloy DJ and Yellon DM: Myocardial ischemia-reperfusion injury: A neglected therapeutic target. J Clin Invest. 123:92–100. 2013.PubMed/NCBI View Article : Google Scholar

10 

Han T, Tang H, Lin C, Shen Y, Yan D, Tang X and Guo D: Extracellular traps and the role in thrombosis. Front Cardiovasc Med. 9(951670)2022.PubMed/NCBI View Article : Google Scholar

11 

Yipp BG and Kubes P: NETosis: how vital is it? Blood. 122:2784–2794. 2013.PubMed/NCBI View Article : Google Scholar

12 

Thiam HR, Wong SL, Qiu R, Kittisopikul M, Vahabikashi A, Goldman AE, Goldman RD, Wagner DD and Waterman CM: NETosis proceeds by cytoskeleton and endomembrane disassembly and PAD4-mediated chromatin decondensation and nuclear envelope rupture. Proc Natl Acad Sci USA. 117:7326–7337. 2020.PubMed/NCBI View Article : Google Scholar

13 

Hakkim A, Fuchs TA, Martinez NE, Hess S, Prinz H, Zychlinsky A and Waldmann H: Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat Chem Biol. 7:75–77. 2011.PubMed/NCBI View Article : Google Scholar

14 

Steinberg BE and Grinstein S: Unconventional roles of the NADPH oxidase: Signaling, ion homeostasis, and cell death. Sci STKE. 2007(pe11)2007.PubMed/NCBI View Article : Google Scholar

15 

Nishinaka Y, Arai T, Adachi S, Takaori-Kondo A and Yamashita K: Singlet oxygen is essential for neutrophil extracellular trap formation. Biochem Biophys Res Commun. 413:75–79. 2011.PubMed/NCBI View Article : Google Scholar

16 

Christophorou MA, Castelo-Branco G, Halley-Stott RP, Oliveira CS, Loos R, Radzisheuskaya A, Mowen KA, Bertone P, Silva JC, Zernicka-Goetz M, et al: Citrullination regulates pluripotency and histone H1 binding to chromatin. Nature. 507:104–108. 2014.PubMed/NCBI View Article : Google Scholar

17 

Ho JW, Quan C, Gauger MA, Alam HB and Li Y: Role of peptidylarginine deiminase and neutrophil extracellular traps in injuries: Future novel diagnostics and therapeutic targets. Shock. 59:247–255. 2023.PubMed/NCBI View Article : Google Scholar

18 

Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, Hayama R, Leonelli L, Han H, Grigoryev SA, et al: Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol. 184:205–213. 2009.PubMed/NCBI View Article : Google Scholar

19 

Papayannopoulos V, Metzler KD, Hakkim A and Zychlinsky A: Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol. 191:677–691. 2010.PubMed/NCBI View Article : Google Scholar

20 

Zollet V, Arenas Hoyos I, Hirsiger S, Brahim BB, Petrucci MF, Casoni D, Wang J, Spirig R, Nettelbeck K, Garcia L, et al: Neutrophil extracellular traps and citrullinated fibrinogen contribute to injury in a porcine model of limb ischemia and reperfusion. Front Immunol. 15(1436926)2024.PubMed/NCBI View Article : Google Scholar

21 

Chen Z, Zhang H, Qu M, Nan K, Cao H, Cata JP, Chen W and Miao C: Review: The emerging role of neutrophil extracellular traps in sepsis and sepsis-associated thrombosis. Front Cell Infect Microbiol. 11(653228)2021.PubMed/NCBI View Article : Google Scholar

22 

Thålin C, Hisada Y, Lundström S, Mackman N and Wallén H: Neutrophil extracellular traps: Villains and targets in arterial, venous, and cancer-associated thrombosis. Arterioscler Thromb Vasc Biol. 39:1724–1738. 2019.PubMed/NCBI View Article : Google Scholar

23 

Leshner M, Wang S, Lewis C, Zheng H, Chen XA, Santy L and Wang Y: PAD4 mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front Immunol. 3(307)2012.PubMed/NCBI View Article : Google Scholar

24 

Papayannopoulos V: Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. 18:134–147. 2018.PubMed/NCBI View Article : Google Scholar

25 

Friedman GD, Klatsky AL and Siegelaub AB: The leukocyte count as a predictor of myocardial infarction. N Engl J Med. 290:1275–1278. 1974.PubMed/NCBI View Article : Google Scholar

26 

Borissoff JI, Joosen IA, Versteylen MO, Brill A, Fuchs TA, Savchenko AS, Gallant M, Martinod K, Ten Cate H, Hofstra L, et al: Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state. Arterioscler Thromb Vasc Biol. 33:2032–2040. 2013.PubMed/NCBI View Article : Google Scholar

27 

Wang Y, Yang M, Xu Y, Yan S, Jin E and Li X: Neutrophil extracellular trap burden correlates with the stenosis of coronary atherosclerosis. PeerJ. 11(e15471)2023.PubMed/NCBI View Article : Google Scholar

28 

Kawasaki H and Iwamuro S: Potential roles of histones in host defense as antimicrobial agents. Infect Disord Drug Targets. 8:195–205. 2008.PubMed/NCBI View Article : Google Scholar

29 

Yipp BG, Petri B, Salina D, Jenne CN, Scott BN, Zbytnuik LD, Pittman K, Asaduzzaman M, Wu K, Meijndert HC, et al: Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat Med. 18:1386–1393. 2012.PubMed/NCBI View Article : Google Scholar

30 

Pilsczek FH, Salina D, Poon KK, Fahey C, Yipp BG, Sibley CD, Robbins SM, Green FH, Surette MG, Sugai M, et al: A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol. 185:7413–7425. 2010.PubMed/NCBI View Article : Google Scholar

31 

Schmitt MMN, Megens RTA, Zernecke A, Bidzhekov K, van den Akker NM, Rademakers T, van Zandvoort MA, Hackeng TM, Koenen RR and Weber C: Endothelial junctional adhesion molecule-a guides monocytes into flow-dependent predilection sites of atherosclerosis. Circulation. 129:66–76. 2014.PubMed/NCBI View Article : Google Scholar

32 

Yalcinkaya M, Liu W, Xiao T, Abramowicz S, Wang R, Wang N, Westerterp M and Tall AR: Cholesterol trafficking to the ER leads to the activation of CaMKII/JNK/NLRP3 and promotes atherosclerosis. J Lipid Res. 65(100534)2024.PubMed/NCBI View Article : Google Scholar

33 

Münzer P, Negro R, Fukui S, di Meglio L, Aymonnier K, Chu L, Cherpokova D, Gutch S, Sorvillo N, Shi L, et al: NLRP3 Inflammasome assembly in neutrophils is supported by PAD4 and promotes NETosis under sterile conditions. Front Immunol. 12(683803)2021.PubMed/NCBI View Article : Google Scholar

34 

Warnatsch A, Ioannou M, Wang Q and Papayannopoulos V: Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 349:316–320. 2015.PubMed/NCBI View Article : Google Scholar

35 

Yalcinkaya M, Liu W, Thomas LA, Olszewska M, Xiao T, Abramowicz S, Papapetrou EP, Westerterp M, Wang N, Tabas I and Tall AR: BRCC3-Mediated NLRP3 deubiquitylation promotes inflammasome activation and atherosclerosis in Tet2 clonal hematopoiesis. Circulation. 148:1764–1777. 2023.PubMed/NCBI View Article : Google Scholar

36 

Westerterp M, Fotakis P, Ouimet M, Bochem AE, Zhang H, Molusky MM, Wang W, Abramowicz S, la Bastide-van Gemert S, Wang N, et al: Cholesterol efflux pathways suppress inflammasome activation, NETosis, and atherogenesis. Circulation. 138:898–912. 2018.PubMed/NCBI View Article : Google Scholar

37 

Li H, Tang C, Zhu X, Zhang W, Abudupataer M, Ding S, Duan C, Yang X and Ge J: Histamine deficiency facilitates coronary microthrombosis after myocardial infarction by increasing neutrophil-platelet interactions. J Cell Mol Med. 24:3504–3520. 2020.PubMed/NCBI View Article : Google Scholar

38 

Zhou J, Chen R, Liu C, Zhou P, Li J, Wang Y, Zhao X, Zhao H, Song L and Yan H: Associations of NETs with inflammatory risk and atherosclerotic severity in ST-segment elevation myocardial infarction. Thromb Res. 203:5–11. 2021.PubMed/NCBI View Article : Google Scholar

39 

Stakos DA, Kambas K, Konstantinidis T, Mitroulis I, Apostolidou E, Arelaki S, Tsironidou V, Giatromanolaki A, Skendros P, Konstantinides S and Ritis K: Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction. Eur Heart J. 36:1405–1414. 2015.PubMed/NCBI View Article : Google Scholar

40 

Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD Jr, Wrobleski SK, Wakefield TW, Hartwig JH and Wagner DD: Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA. 107:15880–15885. 2010.PubMed/NCBI View Article : Google Scholar

41 

Liang GY, Cai QY, Niu YM, Zheng H, Gao ZY, Liu DX and Xu G: Cardiac glucose uptake and suppressed expression/translocation of myocardium glucose transport-4 in dogs undergoing ischemia-reperfusion. Exp Biol Med (Maywood). 233:1142–1148. 2008.PubMed/NCBI View Article : Google Scholar

42 

Remijsen Q, Vanden Berghe T, Wirawan E, Asselbergh B, Parthoens E, De Rycke R, Noppen S, Delforge M, Willems J and Vandenabeele P: Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 21:290–304. 2011.PubMed/NCBI View Article : Google Scholar

43 

Tian H, Zhao X, Zhang Y and Xia Z: Abnormalities of glucose and lipid metabolism in myocardial ischemia-reperfusion injury. Biomed Pharmacother. 163(114827)2023.PubMed/NCBI View Article : Google Scholar

44 

Zamanian M, Hajizadeh M, Shamsizadeh A, Moemenzadeh M, Amirteimouri M, Elshiekh M and Allahtavakoli M: Effects of naringin on physical fatigue and serum MMP-9 concentration in female rats. Pharm Biol. 55:423–427. 2017.PubMed/NCBI View Article : Google Scholar

45 

Zamanian M, Shamsizadeh A, Esmaeili Nadimi A, Hajizadeh M, Allahtavakoli F, Rahmani M, Kaeidi A, Safari Khalegh H and Allahtavakoli M: Short-term effects of troxerutin (vitamin P4) on muscle fatigue and gene expression of Bcl-2 and Bax in the hepatic tissue of rats. Can J Physiol Pharmacol. 95:708–713. 2017.PubMed/NCBI View Article : Google Scholar

46 

Chouchani ET, Pell VR, Gaude E, Aksentijević D, Sundier SY, Robb EL, Logan A, Nadtochiy SM, Ord ENJ, Smith AC, et al: Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 515:431–435. 2014.PubMed/NCBI View Article : Google Scholar

47 

Awasthi D, Nagarkoti S, Sadaf S, Chandra T, Kumar S and Dikshit M: Glycolysis dependent lactate formation in neutrophils: A metabolic link between NOX-dependent and independent NETosis. Biochim Biophys Acta Mol Basis Dis. 1865(165542)2019.PubMed/NCBI View Article : Google Scholar

48 

Sun L, Wu Q, Nie Y, Cheng N, Wang R, Wang G, Zhang D, He H, Ye RD and Qian F: A role for MK2 in enhancing neutrophil-derived ROS production and aggravating liver ischemia/reperfusion injury. Front Immunol. 9(2610)2018.PubMed/NCBI View Article : Google Scholar

49 

Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C and Wen Z: Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol. 74(103231)2024.PubMed/NCBI View Article : Google Scholar

50 

Stojkov D, Gigon L, Peng S, Lukowski R, Ruth P, Karaulov A, Rizvanov A, Barlev NA, Yousefi S and Simon HU: Physiological and Pathophysiological Roles of Metabolic Pathways for NET Formation and Other Neutrophil Functions. Front Immunol. 13(826515)2022.PubMed/NCBI View Article : Google Scholar

51 

Kurian GA, Rajagopal R, Vedantham S and Rajesh M: The role of oxidative stress in myocardial ischemia and reperfusion injury and remodeling: Revisited. Oxid Med Cell Longev. 2016(1656450)2016.PubMed/NCBI View Article : Google Scholar

52 

Bolli R and Marbán E: Molecular and cellular mechanisms of myocardial stunning. Physiol Rev. 79:609–634. 1999.PubMed/NCBI View Article : Google Scholar

53 

Meng M, Jia R, Wei M, Meng X, Zhang X, Du R, Sun W, Wang L and Song L: Oxidative stress activates Ryr2-Ca2+ and apoptosis to promote PM2.5-induced heart injury of hyperlipidemia mice. Ecotoxicol Environ Saf. 232(113228)2022.PubMed/NCBI View Article : Google Scholar

54 

Gorski PA, Jang SP, Jeong D, Lee A, Lee P, Oh JG, Chepurko V, Yang DK, Kwak TH, Eom SH, et al: Role of SIRT1 in modulating acetylation of the sarco-endoplasmic reticulum Ca2+-ATPase in heart failure. Circ Res. 124:e63–e80. 2019.PubMed/NCBI View Article : Google Scholar

55 

Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V and Zychlinsky A: Novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 176:231–241. 2007.PubMed/NCBI View Article : Google Scholar

56 

Ellson CD, Davidson K, Ferguson GJ, O'Connor R, Stephens LR and Hawkins PT: Neutrophils from p40phox-/- mice exhibit severe defects in NADPH oxidase regulation and oxidant-dependent bacterial killing. J Exp Med. 203:1927–1937. 2006.PubMed/NCBI View Article : Google Scholar

57 

Douda DN, Khan MA, Grasemann H and Palaniyar N: SK3 channel and mitochondrial ROS mediate NADPH oxidase-independent NETosis induced by calcium influx. Proc Natl Acad Sci USA. 112:2817–2822. 2015.PubMed/NCBI View Article : Google Scholar

58 

Liao X, Song X, Li J, Li L, Fan X, Qin Q, Zhong C, Yang P, Zhan J and Cai Y: An injectable co-assembled hydrogel blocks reactive oxygen species and inflammation cycle resisting myocardial ischemia-reperfusion injury. Acta Biomater. 149:82–95. 2022.PubMed/NCBI View Article : Google Scholar

59 

Andreadou I, Cabrera-Fuentes HA, Devaux Y, Frangogiannis NG, Frantz S, Guzik T, Liehn EA, Gomes CPC, Schulz R and Hausenloy DJ: Immune cells as targets for cardioprotection: New players and novel therapeutic opportunities. Cardiovasc Res. 115:1117–1130. 2019.PubMed/NCBI View Article : Google Scholar

60 

Zhang L, Deng S, Zhao S, Ai Y, Zhang L, Pan P, Su X, Tan H and Wu D: Intra-peritoneal administration of mitochondrial DNA provokes acute lung injury and systemic inflammation via toll-like receptor 9. Int J Mol Sci. 17(1425)2016.PubMed/NCBI View Article : Google Scholar

61 

Xie L, He S, Kong N, Zhu Y, Tang Y, Li J, Liu Z, Liu J and Gong J: Cpg-ODN, a TLR9 agonist, aggravates myocardial ischemia/reperfusion injury by activation of TLR9-P38 MAPK signaling. Cell Physiol Biochem. 47:1389–1398. 2018.PubMed/NCBI View Article : Google Scholar

62 

Hidalgo A, Libby P, Soehnlein O, Aramburu IV, Papayannopoulos V and Silvestre-Roig C: Neutrophil extracellular traps: From physiology to pathology. Cardiovasc Res. 118:2737–2753. 2022.PubMed/NCBI View Article : Google Scholar

63 

Shah M, He Z, Rauf A, Beikoghli Kalkhoran S, Heiestad CM, Stensløkken KO, Parish CR, Soehnlein O, Arjun S, Davidson SM and Yellon D: Extracellular histones are a target in myocardial ischaemia-reperfusion injury. Cardiovasc Res. 118:1115–1125. 2022.PubMed/NCBI View Article : Google Scholar

64 

Wilson AS, Randall KL, Pettitt JA, Ellyard JI, Blumenthal A, Enders A, Quah BJ, Bopp T, Parish CR and Brüstle A: Neutrophil extracellular traps and their histones promote Th17 cell differentiation directly via TLR2. Nat Commun. 13(528)2022.PubMed/NCBI View Article : Google Scholar

65 

Tsourouktsoglou TD, Warnatsch A, Ioannou M, Hoving D, Wang Q and Papayannopoulos V: Histones, DNA, and citrullination promote neutrophil extracellular trap inflammation by regulating the localization and activation of TLR4. Cell Rep. 31(107602)2020.PubMed/NCBI View Article : Google Scholar

66 

Liu S, Su X, Pan P, Zhang L, Hu Y, Tan H, Wu D, Liu B, Li H, Li H, et al: Neutrophil extracellular traps are indirectly triggered by lipopolysaccharide and contribute to acute lung injury. Sci Rep. 6(37252)2016.PubMed/NCBI View Article : Google Scholar

67 

Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, et al: Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 13:463–469. 2007.PubMed/NCBI View Article : Google Scholar

68 

Maugeri N, Campana L, Gavina M, Covino C, De Metrio M, Panciroli C, Maiuri L, Maseri A, D'Angelo A, Bianchi ME, et al: Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps. J Thromb Haemost. 12:2074–2088. 2014.PubMed/NCBI View Article : Google Scholar

69 

Kawashima S and Yokoyama M: Dysfunction of endothelial nitric oxide synthase and atherosclerosis. Arterioscler Thromb Vasc Biol. 24:998–1005. 2004.PubMed/NCBI View Article : Google Scholar

70 

Carlstrom M, Weitzberg E and Lundberg JO: Nitric oxide signaling and regulation in the cardiovascular system: Recent advances. Pharmacol Rev. 76:1038–1062. 2024.PubMed/NCBI View Article : Google Scholar

71 

Godo S, Takahashi J, Shiroto T, Yasuda S and Shimokawa H: Coronary microvascular spasm: Clinical presentation and diagnosis. Eur Cardiol. 18(e07)2023.PubMed/NCBI View Article : Google Scholar

72 

Zhou Q, Cao J and Chen L: Apelin/APJ system: A novel therapeutic target for oxidative stress-related inflammatory diseases (Review). Int J Mol Med. 37:1159–1169. 2016.PubMed/NCBI View Article : Google Scholar

73 

Wu X, Xu M, Liu Z, Zhang Z, Liu Y, Luo S, Zheng X, Little PJ, Xu S and Weng J: Pharmacological inhibition of IRAK1 and IRAK4 prevents endothelial inflammation and atherosclerosis in ApoE-/- mice. Pharmacol Res. 175(106043)2022.PubMed/NCBI View Article : Google Scholar

74 

Aldabbous L, Abdul-Salam V, McKinnon T, Duluc L, Pepke-Zaba J, Southwood M, Ainscough AJ, Hadinnapola C, Wilkins MR, Toshner M and Wojciak-Stothard B: Neutrophil extracellular traps promote angiogenesis: Evidence from vascular pathology in pulmonary hypertension. Arterioscler Thromb Vasc Biol. 36:2078–2087. 2016.PubMed/NCBI View Article : Google Scholar

75 

Cao Y, Chen M, Jiao X, Li S, Wang D, Zhan Y, Li J, Hao Z, Li Q, Liu Y, et al: Neutrophil extracellular traps mediate the crosstalk between plaque microenvironment and unstable carotid plaque formation. Exp Mol Med. 56:1717–1735. 2024.PubMed/NCBI View Article : Google Scholar

76 

Chrysanthopoulou A, Gkaliagkousi E, Lazaridis A, Arelaki S, Pateinakis P, Ntinopoulou M, Mitsios A, Antoniadou C, Argyriou C, Georgiadis GS, et al: Angiotensin II triggers release of neutrophil extracellular traps, linking thromboinflammation with essential hypertension. JCI Insight. 6(e148668)2021.PubMed/NCBI View Article : Google Scholar

77 

Cohn JN, Ferrari R and Sharpe N: Cardiac remodeling-concepts and clinical implications: A consensus paper from an international forum on cardiac remodeling. Behalf of an International Forum on Cardiac Remodeling. J Am Coll Cardiol. 35:569–582. 2000.PubMed/NCBI View Article : Google Scholar

78 

Mangold A, Hofbauer TM, Ondracek AS, Artner T, Scherz T, Speidl WS, Krychtiuk KA, Sadushi-Kolici R, Jakowitsch J and Lang IM: Neutrophil extracellular traps and monocyte subsets at the culprit lesion site of myocardial infarction patients. Sci Rep. 9(16304)2019.PubMed/NCBI View Article : Google Scholar

79 

Jorch SK and Kubes P: An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med. 23:279–287. 2017.PubMed/NCBI View Article : Google Scholar

80 

Li C, Gao P, Zhuang F, Wang T, Wang Z, Wu G, Zhou Z, Xie H, Xie D, Zhao D, et al: Inhibition of ALOX12-12-HETE alleviates lung ischemia-reperfusion injury by reducing endothelial ferroptosis-mediated neutrophil extracellular trap formation. Research (Wash D C). 7(0473)2024.PubMed/NCBI View Article : Google Scholar

81 

Chen J, Wang T, Li X, Gao L, Wang K, Cheng M, Zeng Z, Chen L, Shen Y and Wen F: DNA of neutrophil extracellular traps promote NF-κB-dependent autoimmunity via cGAS/TLR9 in chronic obstructive pulmonary disease. Signal Transduct Target Ther. 9(163)2024.PubMed/NCBI View Article : Google Scholar

82 

He L, Liu R, Yue H, Zhu G, Fu L, Chen H, Guo Y and Qin C: NETs promote pathogenic cardiac fibrosis and participate in ventricular aneurysm formation after ischemia injury through the facilitation of perivascular fibrosis. Biochem Biophys Res Commun. 583:154–161. 2021.PubMed/NCBI View Article : Google Scholar

83 

Chang CP, Chia RH, Wu TL, Tsao KC, Sun CF and Wu JT: Elevated cell-free serum DNA detected in patients with myocardial infarction. Clin Chim Acta. 327:95–101. 2003.PubMed/NCBI View Article : Google Scholar

84 

Cao J, Roth S, Zhang S, Kopczak A, Mami S, Asare Y, Georgakis MK, Messerer D, Horn A, Shemer R, et al: DNA-sensing inflammasomes cause recurrent atherosclerotic stroke. Nature. 633:433–441. 2024.PubMed/NCBI View Article : Google Scholar

85 

Hally KE, Parker OM, Brunton-O'Sullivan MM, Harding SA and Larsen PD: Linking neutrophil extracellular traps and platelet activation: A composite biomarker score for predicting outcomes after acute myocardial infarction. Thromb Haemost. 121:1637–1649. 2021.PubMed/NCBI View Article : Google Scholar

86 

Yu S, Li M, Li Z, Xu P, Yao Z, Qian S, Qian F, Gao D and Wang H: Positive correlations between plasma BPI level and MPO-DNA and S100A8/A9 in myocardial infarction. Platelets. 33:603–611. 2022.PubMed/NCBI View Article : Google Scholar

87 

Nagareddy PR, Sreejit G, Abo-Aly M, Jaggers RM, Chelvarajan L, Johnson J, Pernes G, Athmanathan B, Abdel-Latif A and Murphy AJ: NETosis Is Required for S100A8/A9-induced granulopoiesis after myocardial infarction. Arterioscler Thromb Vasc Biol. 40:2805–2807. 2020.PubMed/NCBI View Article : Google Scholar

88 

Li YW, Chen SX, Yang Y, Zhang ZH, Zhou WB, Huang YN, Huang ZQ, He JQ, Chen TF, Wang JF, et al: Colchicine inhibits NETs and alleviates cardiac remodeling after acute myocardial infarction. Cardiovasc Drugs Ther. 38:31–41. 2024.PubMed/NCBI View Article : Google Scholar

89 

Tan Y, Bao X, Li Y, Song G, Lu H, Sun X, Gu R, Kang L and Xu B: Colchicine attenuates microvascular obstruction after myocardial ischemia-reperfusion injury by inhibiting the proliferation of neutrophil in bone marrow. Cardiovasc Drugs Ther. 39:259–273. 2025.PubMed/NCBI View Article : Google Scholar

90 

Banach M and Penson PE: Colchicine and cardiovascular outcomes: A critical appraisal of recent studies. Curr Atheroscler Rep. 23(32)2021.PubMed/NCBI View Article : Google Scholar

91 

Tardif JC, Kouz S, Waters DD, Bertrand OF, Diaz R, Maggioni AP, Pinto FJ, Ibrahim R, Gamra H, Kiwan GS, et al: Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med. 381:2497–2505. 2019.PubMed/NCBI View Article : Google Scholar

92 

Zhang W, Liu J, Li X, Bai Z, Sun Y and Chen X: Lidocaine effects on neutrophil extracellular trapping and angiogenesis biomarkers in postoperative breast cancer patients with different anesthesia methods: A prospective, randomized trial. BMC Anesthesiol. 24(162)2024.PubMed/NCBI View Article : Google Scholar

93 

Sapey E, Patel JM, Greenwood H, Walton GM, Grudzinska F, Parekh D, Mahida RY, Dancer RCA, Lugg ST, Howells PA, et al: Simvastatin improves neutrophil function and clinical outcomes in pneumonia. A pilot randomized controlled clinical trial. Am J Respir Crit Care Med. 200:1282–1293. 2019.PubMed/NCBI View Article : Google Scholar

94 

Ebrahimi F, Giaglis S, Hahn S, Blum CA, Baumgartner C, Kutz A, van Breda SV, Mueller B, Schuetz P, Christ-Crain M and Hasler P: Markers of neutrophil extracellular traps predict adverse outcome in community-acquired pneumonia: Secondary analysis of a randomised controlled trial. Eur Respir J. 51(1701389)2018.PubMed/NCBI View Article : Google Scholar

95 

Shin D, Kim J, Lee S and Chae MS: Impact of perioperative lidocaine on neutrophil extracellular trapping and serum cytokines in robot-assisted radical prostatectomy: Randomized controlled study. Medicina (Kaunas). 60(1452)2024.PubMed/NCBI View Article : Google Scholar

96 

Hu Z, Hua X, Mo X, Chang Y, Chen X, Xu Z, Tao M, Hu G and Song J: Inhibition of NETosis via PAD4 alleviated inflammation in giant cell myocarditis. IScience. 26(107162)2023.PubMed/NCBI View Article : Google Scholar

97 

Ai P, Pan H, Chen K, Zheng J, Gao Z and Jin G: Viral mimetic poly(I:C) induces neutrophil extracellular traps via PAD4 to promote inflammation and thrombosis. Biochem Biophys Res Commun. 565:64–71. 2021.PubMed/NCBI View Article : Google Scholar

98 

Savchenko AS, Borissoff JI, Martinod K, De Meyer SF, Gallant M, Erpenbeck L, Brill A, Wang Y and Wagner DD: VWF-mediated leukocyte recruitment with chromatin decondensation by PAD4 increases myocardial ischemia/reperfusion injury in mice. Blood. 123:141–148. 2014.PubMed/NCBI View Article : Google Scholar

99 

Ge L, Zhou X, Ji WJ, Lu RY, Zhang Y, Zhang YD, Ma YQ, Zhao JH and Li YM: Neutrophil extracellular traps in ischemia-reperfusion injury-induced myocardial no-reflow: Therapeutic potential of DNase-based reperfusion strategy. Am J Physiol Heart Circ Physiol. 308:H500–H509. 2015.PubMed/NCBI View Article : Google Scholar

100 

Di G, Vázquez-Reyes S, Díaz B, Peña-Martinez C, García-Culebras A, Cuartero MI, Moraga A, Pradillo JM, Esposito E, Lo EH, et al: Daytime DNase-I administration protects mice from ischemic stroke without inducing bleeding or tPA-induced hemorrhagic transformation, even with aspirin pretreatment. Stroke. 56:527–532. 2025.PubMed/NCBI View Article : Google Scholar

101 

Carminita E, Crescence L, Brouilly N, Altié A, Panicot-Dubois L and Dubois C: DNAse-dependent, NET-independent pathway of thrombus formation in vivo. Proc Natl Acad Sci USA. 118(e2100561118)2021.PubMed/NCBI View Article : Google Scholar

102 

Mangold A, Alias S, Scherz T, Hofbauer M, Jakowitsch J, Panzenböck A, Simon D, Laimer D, Bangert C, Kammerlander A, et al: Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ Res. 116:1182–1192. 2015.PubMed/NCBI View Article : Google Scholar

103 

Kolaczkowska E, Jenne CN, Surewaard BG, Thanabalasuriar A, Lee WY, Sanz MJ, Mowen K, Opdenakker G and Kubes P: Molecular mechanisms of NET formation and degradation revealed by intravital imaging in the liver vasculature. Nat Commun. 6(6673)2015.PubMed/NCBI View Article : Google Scholar

104 

Englert H, Göbel J, Khong D, Omidi M, Wolska N, Konrath S, Frye M, Mailer RK, Beerens M, Gerwers JC, et al: Targeting NETs using dual-active DNase1 variants. Front Immunol. 14(1181761)2023.PubMed/NCBI View Article : Google Scholar

105 

Liu T, Lv X, Xu Q, Qi X, Qiu S, Luan Y, Shen N, Cheng J, Jin L, Tian T, et al: Stroke-homing peptide-DNase1 alleviates intestinal ischemia reperfusion injury by selectively degrading neutrophil extracellular traps. Cell Prolif. (e70010)2025.PubMed/NCBI View Article : Google Scholar : (Epub ahead of print).

106 

Yao D, Bao L, Wang S, Tan M, Xu Y, Wu T, Zhang Z and Gong K: Isoliquiritigenin alleviates myocardial ischemia-reperfusion injury by regulating the Nrf2/HO-1/SLC7a11/GPX4 axis in mice. Free Radic Biol Med. 221:1–12. 2024.PubMed/NCBI View Article : Google Scholar

107 

Yang K, Gao R, Chen H, Hu J, Zhang P, Wei X, Shi J, Chen Y, Zhang L, Chen J, et al: Myocardial reperfusion injury exacerbation due to ALDH2 deficiency is mediated by neutrophil extracellular traps and prevented by leukotriene C4 inhibition. Eur Heart J. 45:1662–1680. 2024.PubMed/NCBI View Article : Google Scholar

108 

Lin K, Fang S, Cai B, Huang X, Zhang X, Lu Y, Zhang W and Wei E: ERK/Egr-1 signaling pathway is involved in CysLT2 receptor-mediated IL-8 production in HEK293 cells. Eur J Cell Biol. 93:278–288. 2014.PubMed/NCBI View Article : Google Scholar

109 

Dölling M, Eckstein M, Singh J, Schauer C, Schoen J, Shan X, Bozec A, Knopf J, Schett G, Muñoz LE and Herrmann M: Hypoxia promotes neutrophil survival after acute myocardial infarction. Front Immunol. 13(726153)2022.PubMed/NCBI View Article : Google Scholar

110 

Tang X, Wang P, Zhang R, Watanabe I, Chang E, Vinayachandran V, Nayak L, Lapping S, Liao S, Madera A, et al: KLF2 regulates neutrophil activation and thrombosis in cardiac hypertrophy and heart failure progression. J Clin Invest. 132(e147191)2022.PubMed/NCBI View Article : Google Scholar

111 

Vorobjeva N, Galkin I, Pletjushkina O, Golyshev S, Zinovkin R, Prikhodko A, Pinegin V, Kondratenko I, Pinegin B and Chernyak B: Mitochondrial permeability transition pore is involved in oxidative burst and NETosis of human neutrophils. Biochim Biophys Acta Mol Basis Dis. 1866(165664)2020.PubMed/NCBI View Article : Google Scholar

112 

Vajen T, Koenen RR, Werner I, Staudt M, Projahn D, Curaj A, Sönmez TT, Simsekyilmaz S, Schumacher D, Möllmann J, et al: Blocking CCL5-CXCL4 heteromerization preserves heart function after myocardial infarction by attenuating leukocyte recruitment and NETosis. Sci Rep. 8(10647)2018.PubMed/NCBI View Article : Google Scholar

113 

Xu K, Cooney KA, Shin EY, Wang L, Deppen JN, Ginn SC and Levit RD: Adenosine from a biologic source regulates neutrophil extracellular traps (NETs). J Leukoc Biol. 105:1225–1234. 2019.PubMed/NCBI View Article : Google Scholar

114 

Allard B, Longhi MS, Robson SC and Stagg J: The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev. 276:121–144. 2017.PubMed/NCBI View Article : Google Scholar

115 

Borg N, Alter C, Görldt N, Jacoby C, Ding Z, Steckel B, Quast C, Bönner F, Friebe D, Temme S, et al: CD73 on T cells orchestrates cardiac wound healing after myocardial infarction by purinergic metabolic reprogramming. Circulation. 136:297–313. 2017.PubMed/NCBI View Article : Google Scholar

116 

Sayegh MN, Cooney KA, Han WM, Cicka M, Strobel F, Wang L, García AJ and Levit RD: Hydrogel delivery of purinergic enzymes improves cardiac ischemia/reperfusion injury. J Mol Cell Cardiol. 176:98–109. 2023.PubMed/NCBI View Article : Google Scholar

117 

Chilingaryan Z, Deshmukh T, Leung HHL, Perdomo J, Emerson P, Kurup R, Chong BH and Chong JJH: Erythrocyte interaction with neutrophil extracellular traps in coronary artery thrombosis following myocardial infarction. Pathology. 54:87–94. 2022.PubMed/NCBI View Article : Google Scholar

118 

Dou H, Kotini A, Liu W, Fidler T, Endo-Umeda K, Sun X, Olszewska M, Xiao T, Abramowicz S, Yalcinkaya M, et al: Oxidized phospholipids promote NETosis and arterial thrombosis in LNK(SH2B3) deficiency. Circulation. 144:1940–1954. 2021.PubMed/NCBI View Article : Google Scholar

119 

Fordyce CB, Gersh BJ, Stone GW and Granger CB: Novel therapeutics in myocardial infarction: Targeting microvascular dysfunction and reperfusion injury. Trends Pharmacol Sci. 36:605–616. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2025
Volume 23 Issue 1

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Z, Wang Y, Li T and Wang H: NETosis in myocardial ischemia‑reperfusion injury: From mechanisms to therapies (Review). Biomed Rep 23: 113, 2025.
APA
Zhang, Z., Wang, Y., Li, T., & Wang, H. (2025). NETosis in myocardial ischemia‑reperfusion injury: From mechanisms to therapies (Review). Biomedical Reports, 23, 113. https://doi.org/10.3892/br.2025.1991
MLA
Zhang, Z., Wang, Y., Li, T., Wang, H."NETosis in myocardial ischemia‑reperfusion injury: From mechanisms to therapies (Review)". Biomedical Reports 23.1 (2025): 113.
Chicago
Zhang, Z., Wang, Y., Li, T., Wang, H."NETosis in myocardial ischemia‑reperfusion injury: From mechanisms to therapies (Review)". Biomedical Reports 23, no. 1 (2025): 113. https://doi.org/10.3892/br.2025.1991