Open Access

Myocardial ischemia/reperfusion injury: Mechanisms of injury and implications for management (Review)

  • Authors:
    • Jianfeng He
    • Danyong Liu
    • Lixia Zhao
    • Dongcheng Zhou
    • Jianhui Rong
    • Liangqing Zhang
    • Zhengyuan Xia
  • View Affiliations

  • Published online on: May 6, 2022     https://doi.org/10.3892/etm.2022.11357
  • Article Number: 430
  • Copyright: © He 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

Myocardial infarction is one of the primary causes of mortality in patients with coronary heart disease worldwide. Early treatment of acute myocardial infarction restores blood supply of ischemic myocardium and decreases the mortality risk. However, when the interrupted myocardial blood supply is recovered within a certain period of time, it causes more serious damage to the original ischemic myocardium; this is known as myocardial ischemia/reperfusion injury (MIRI). The pathophysiological mechanisms leading to MIRI are associated with oxidative stress, intracellular calcium overload, energy metabolism disorder, apoptosis, endoplasmic reticulum stress, autophagy, pyroptosis, necroptosis and ferroptosis. These interplay with one another and directly or indirectly lead to aggravation of the effect. In the past, apoptosis and autophagy have attracted more attention but necroptosis and ferroptosis also serve key roles. However, the mechanism of MIRI has not been fully elucidated. The present study reviews the mechanisms underlying MIRI. Based on current understanding of the pathophysiological mechanisms of MIRI, the association between cell death‑associated signaling pathways were elaborated, providing direction for investigation of novel targets in clinical treatment.

1. Introduction

Coronary heart disease (CHD) is one of the primary causes of mortality and disability worldwide. For instance, according to the data obtained through the Unified Health System database from the Ministry of Health of Brazil, the average mortality rate for CHD increased to 78.75 during 2016-2018 in Brazil (1). Timely restoration of blood flow to ischemic myocardium in the early stage effectively limits infarct size and is therefore standard treatment to prevent death of cardiomyocytes at risk (2,3). Evidence has indicated that during myocardial ischemia/reperfusion (MIR), both ischemia and reperfusion cause injury to the ischemic myocardium (4,5). Therefore, myocardial reperfusion may further aggravate death of ischemic cardiomyocytes in patients with myocardial infarction (MI); this is known as MIR injury (MIRI) (3,4).

MIRI, first reported by Jennings et al in 1960(6), causes four types of injury including myocardial stunning (7), the no-reflow phenomenon (8), reperfusion arrhythmia (9) and lethal reperfusion injury (10). The first two forms are reversible while the others are irreversible (11). Clinically, percutaneous coronary intervention (PCI) (12), antiplatelet (13) and anticoagulant (14) therapy are primarily used for patients exposed to the latter two types of injury to maintain coronary artery patency. During myocardial reperfusion, timely and effective restoration of blood flow cannot effectively prevent MIRI.

Numerous studies have researched mechanisms underlying MIRI; the pathophysiological mechanisms of MIRI are associated with oxidative stress, intracellular calcium overload, energy metabolism disorder, apoptosis, endoplasmic reticulum stress (ERS), autophagy, pyroptosis, ferroptosis and necroptosis (15,16). Furthermore, these mechanisms are interrelated and may directly or indirectly lead to the aggravation of cell death. The present review summarizes research on the pathophysiological mechanisms underlying MIRI.

2. Pathophysiological mechanisms of MIRI

Oxidative stress

Reactive oxygen species (ROS), including superoxide, hydrogen peroxide (H2O2), singlet oxygen, lipid peroxides, peroxynitrite, hypochlorite, ozone and hydroxyl, hydrogen peroxide and alkoxy radicals (17), are small reactive molecules involved in regulation of various cellular functions and biological processes. ROS at low-to-moderate levels serve as signaling molecules, while uncontrolled high levels of ROS lead to free radical damage associated with structural and functional changes in proteins, lipids and DNA (18,19). For example, ROS at moderate levels may activate cell signaling, augmenting production and release of proinflammatory cytokines, thereby perpetuating the well-controlled inflammatory responses, which are part of our body's innate immunity, protecting against invading pathogens (20). As ROS induces various types of cell death (such as apoptosis, necrosis and pyroptosis), ROS are considered to serve a key role in IRI in a number of organs (20). Furthermore, vascular endothelial dysfunction induced by ROS also contributes to the occurrence and development of IRI in different organs, such as the heart, cerebrum and liver (21,22). When overproduction of ROS exceeds the ability to clear them, oxidative stress occurs, leading to imbalance of oxidation and antioxidant systems and cell and tissue damage (23).

During IR, especially reperfusion, ROS levels increase due to multiple mechanisms such as increased xanthine oxidase formation, neutrophil respiratory burst and damage of the mitochondrial electron transport chain (24). The excessive ROS decrease membrane fluidity, increase calcium permeability, aggravate intracellular calcium overload and mitochondrial damage and contribute to release of pro-apoptotic factors, such as cytochrome C (CytC) (25). In addition, oxidative reactions between ROS and proteins cause loss of original protein structure and function and damage of nucleic acids and chromosomes (19). In addition, ROS may trigger the inflammatory cascade reaction and expression of adhesion molecules that result in leukocyte aggregation, endothelial cell swelling and the no-reflow phenomenon that refers to the incomplete and uneven reperfusion at the microvascular level even though the proximal artery has been re-opened after a period of ischemia (26). Oxidative stress induced by reperfusion after ischemia is considered to be the primary mechanism of IRI (20). Oxidative stress is involved in ventricular remodeling by causing endothelial dysfunction, myocardial cell injury, apoptosis and other pathological changes that promote formation and development of cardiomyopathy (27) and cause cardiac dysfunction. Among potential sources of ROS, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, xanthine oxidase, mitochondria and uncoupled nitric oxide synthase (NOS) are key sources of reperfusion-induced oxidative stress (28). When ROS levels exceed the ability of the cellular endogenous radical scavenging system to remove them (29), uncontrolled ROS burst damages the membrane and proteins and indirectly causes damage by opening mitochondria permeability transition pore (mPTP) and promoting activation of the apoptosis pathway. Thus, ROS participate in myocardial injury and death through multiple mechanisms (30).

NO, synthesized by endothelial (e)NOS, is a key defense mechanism against MIRI (31-33). Studies by our group indicated that activation of PI3K/AKT and Janus kinase 2/STAT3 pathways attenuates MIRI (34-36) and the underlying mechanism is associated with activation of eNOS and increased levels of NO (32,37,38).

Intracellular calcium overload

Contraction and relaxation of myocytes are controlled by balance of calcium. In the steady-state, calcium influx and efflux are maintained in balance. Any increase in calcium influx (for example, during β-adrenergic stimulation) is met by a corresponding increase in calcium efflux (39,40). Under normal conditions, calcium influx and efflux balance ensures cells do not become overloaded with calcium. Sarcoplasmic reticulum calcium content is increased when calcium influx exceeds efflux until a new steady-state of influx and efflux is achieved (39).

After a certain period of hypoxia in cardiomyocytes, the intracellular anaerobic metabolism may result in accumulation of H+, decreased intracellular pH and increased levels of intracellular Na+ via Na+/H+ exchange (NHE) (41). The aggregation of Na+ in cardiomyocytes increases the activity of Na+/Ca2+ exchange (NCX) proteins, which decreases aggregation of intracellular Na+ and transports extracellular Ca2+ into the cells, thus increasing the concentration of Ca2+ in the cytoplasm and causing calcium overload (42). Rapid reoxygenation increases the pH of extracellular fluid and H+ gradient across the cell membrane, so that the activity of NHE and NCX increases, causing intracellular calcium overload (43). In addition to the aforementioned NHE and NCX mechanisms, the H+/Ca2+ exchange mechanism is also a key cause of intracellular calcium overload (44).

Calcium processing and further inflow or outflow are regulated by protein channels (45). Strict regulation of intracellular calcium homeostasis is key for maintaining normal function and growth of myocardial cells and disruption of intracellular calcium homeostasis leads to ERS, aggravating ischemic injury of myocardial cells (19). Chen et al (46) indicated that diltiazem, a calcium antagonist, has protective effects on ischemic myocardium, which decreases oxidative stress, restores normal energy metabolism, improves endothelial function and decreases apoptosis. In addition, clinical trials have found that diltiazem exerts a cardioprotective effect in patients who exhibit post-transplant hypertension and vasospastic angina (47,48). As reported in the aforementioned studies, diltiazem improves left ventricular systolic function and coronary hemodynamics in patients with myocardial ischemia and prevents reperfusion arrhythmia in patients with ST segment elevation MI following treatment with PCI (46).

Energy metabolism disorder

Mitochondria, a key organelle responsible for production of adenosine triphosphate (ATP) and energy metabolism in cells, also contribute to normal physiological function of the heart (49). Mitochondrial integrity and loss of function in cardiomyocytes are considered to be pathological factors underlying changes in cardiac structure and function (49). By forming a dynamic network, mitochondria continuously change shape by division and fusion to meet functional needs of cardiomyocytes (50). Since cardiomyocytes consume lots of energy, the density of mitochondria is relatively higher compared with other types of cell (51).

The function of mitochondria is regulated by numerous factors. Physiologically, mitochondrial Ca2+ is an active effector, which triggers activation of mitochondrial metabolic mechanisms that increase production of ATP. However, during IR, cytoplasmic calcium overload leads to mitochondrial calcium overload (52). Ca2+ enter mitochondria primarily via the mitochondrial calcium uniporter, a type of small conductance Ca2+ selective channel that regulates intracellular Ca2+, production of ATP and cell death (52). The movement of Ca2+ across the mitochondrial membrane leads to amplification of mitochondrial Ca2+ overload, causing mitochondrial dysfunction (53). Overload of mitochondrial Ca2+ dissipates membrane potential, promotes opening of mPTP, a non-selective channel in the inner mitochondrial membrane that is considered to be one of the key participants in IRI, and leads to impaired ATP synthesis (54). The inner mitochondrial membrane is normally impermeable to ions and proteins and is responsible for maintaining formation of mitochondrial transmembrane potential pores, creating non-selective channels and causing potential to dissipate across the membrane. Our previous study found that the degree of mPTP opening degree corresponds to change of mitochondrial morphology, which indicates that mitochondrial function is associated with its morphology during MIRI (50).

Disturbance of mitochondrial homeostasis contributes to acute organ failure and cell damage following ischemia (22). The opening and closing of mPTP is the primary mechanism for maintaining mitochondrial membrane potential; ischemia induces mPTP closure whereas reperfusion promotes mPTP opening (54). During acute IR, mPTP remains closed during ischemia and opens due to mitochondrial calcium and phosphate overload, oxidative stress and rapid pH correction during reperfusion (55,56). ROS levels increase in a short period of time due to IRI, resulting in excessive cytoplasmic and mitochondrial Ca2+, which induces opening of mPTP and depolarizes the mitochondrial membrane potential (54). The opening of mPTP may contribute to loss of ATP, mitochondrial swelling and CytC release, leading to apoptosis (57). Our previous study demonstrated that regulation of mitochondrial morphology, inhibition of mPTP opening and maintenance of mitochondrial membrane potential protect the myocardium against MIRI; the underlying mechanism involves regulation of mitofusin 2 (Mfn2), optic atrophy 1 (Opa1) and dynamin-related protein 1 (Drp1) proteins, which provides a theoretical basis for targeted therapy (50).

Apoptosis

Apoptosis is a form of regulated cell death that leads to cell contraction, condensation of cytoplasm and nucleus and formation of apoptotic bodies (58). Cytoskeletal and nuclear proteins are degraded and cleaved by caspases during the execution phase of apoptosis (59). Finally, apoptotic bodies are phagocytosed by macrophages that recognize externalized phosphatidylserine on the outer leaflet of the bilayer (60). Because apoptotic bodies are surrounded by cell membranes and are swallowed and digested by phagocytes such as neutrophils, macrophages, and dendritic cells (DCs), they eventually degrade without causing inflammation (61). However, part of the cell structures (e.g. cell membrane) can be preserved till the late stage of apoptosis (62).

There are three classic apoptosis signaling pathways: Extrinsic (death receptor), intrinsic (mitochondrial) and ERS pathway (63).

Extrinsic pathway-induced apoptosis is triggered by transmembrane death receptors, which are members of tumor necrosis factor receptor (TNFR) family containing the ‘death domain’. Through the death domain, certain ligands and associated death receptors, including apoptosis-stimulating fragment ligand (FasL)/FasR, TNF-α/TNFR1, TNF-related apoptosis-inducing ligand (TRAIL)/death receptors and TRAIL/DR5, facilitate transmission of death signals from cell surface to intracellular pathways (63). For example, binding of FasL to FasR or TNF-α to TNFR1 causes recruitment of Fas-associated death domain (FADD) and binding to the ligand-receptor complex (64). FADD activates pro-caspase-8 consecutively and forms a death-inducing signaling complex (DISC) (65). DISC-activated caspase-8 leads to activation of caspase-3 and -7 to induce the apoptosis cascade response, which results in the execution phase of apoptosis (66,67). During of MIRI, ischemia and reperfusion increases levels of ROS, cellular injury and cardiovascular dysfunction, which leads to cardiomyocyte apoptosis. Upregulation of cardiac-specific caspase-3 increases post-ischemic infarct size and risk of mortality following MIRI (68). Furthermore, the Fas pathway serves as a critical mediator of the cardiomyocyte apoptosis caused by MIRI (69). In addition, TNF-α and TRAIL levels increase during onset of reperfusion in mouse models of MIRI (70).

The intrinsic pathway, which is activated by hypoxia, hyperthermia and low levels of growth factors (e.g. nerve growth factor), is triggered by mitochondria and contributes to apoptosis. The aforementioned stimuli promote opening of mPTP and block mitochondrial transmembrane potential, thereby increasing release of pro-apoptotic proteins (e.g. Bax) involved in CytC and apoptosis from the intermembrane to the cytoplasm (71). By binding and activating caspase-9, CytC promotes formation of caspase-3 and -7. These events induced by mitochondria are regulated by B-cell lymphoma 2 (Bcl-2) family proteins located in the mitochondrial outer membrane, which control the permeability of the mitochondrial membrane and regulate release of CytC. Bcl-2 family proteins are functionally divided into pro- and anti-apoptotic proteins (67). Bcl-2 family proteins lead to downregulation of apoptosis mediated by upregulation of tumor suppressor protein p53 in the nucleus or mitochondria. When expression of p53 is inhibited, Bax and Caspase-3 are downregulated in heart tissue in CHD (72-74). There is also an interaction between the extrinsic and intrinsic pathways (75). In the intrinsic apoptosis pathway, Bax is overexpressed in ischemic myocardial tissue and inhibition of Bax activation decreases apoptosis and improves MIRI (76). Furthermore, cardiac-specific overexpression of Bcl-2 markedly relieves cardiomyocyte apoptosis and infarct size following MIRI (77).

Previous studies have indicated that apoptotic cell death is one of the primary forms of cardiomyocyte death during MIRI (78-84).

ERS

ER is a key site for protein synthesis, modification and processing and its normal function serves an important role in maintaining cellular homeostasis (85). However, adverse stimuli such as ischemia and hypoxia lead to accumulation or misfolding of proteins in the ER. The accumulation of unfolded protein simultaneously activates three transmembrane stress sensors, inositol-requiring enzyme 1 (IRE1), activating transcription factor (ATF) 6 and PERK, known as the unfolded protein response (UPR) (86). ERS is a relatively novel pathway regulating apoptosis that is involved in multiple physiological functions and pathological injury, including protein folding, intracellular Ca2+ storage, oxidative stress, hypoxia, ischemia and lipid metabolism disorder (87). Although ERS is key for cell survival, chronic ERS leads to apoptosis.

IRI may be associated with ERS (87). Unfolded proteins in ERS induce myocardial injury, which further induces ERS, thereby affecting the metabolic state of cardiomyocytes and causing greater injury (86). During MIRI, ERS is increased and inhibition of ERS has been shown to attenuate MIRI (88-90). However, it has also been reported that not all ERS is harmful. For instance, ERS transcription factor ATF6 induces protective effects against IRI in cardiomyocytes (91,92). However, sustained ERS causes apoptosis (93).

ER and mitochondria are physically connected to form dedicated structural domains known as mitochondria-associated ER membranes (MAMs), which participate in key biological processes, including lipid and Ca2+ homeostasis, mitochondrial dynamics and associated cellular behavior, such as autophagy, ERS and apoptosis. Gao et al (94) demonstrated the role of MAMs in maintaining normal function of both ER and mitochondria, which are associated with occurrence of cardiovascular disease. Another study also suggested that GSK3β serves an important role in controlling Ca2+ flow from the sarcoplasmic reticulum to mitochondria via MAMs during MIRI (95).

Changes in ER oxidation lead to aberrant formation of disulfide bonds and accumulation of peptides, thus activating intracellular reactions called unfolded protein response (96), a form of acute response in cardiovascular disease (97). Stimulation of UPR leads to three primary response mechanisms, IRE1α, PERK and ATF6, which regulate the protein folding ability of ER (98). In the absence of PERK, endogenous apoptosis induced by ERS is weakened due to decreased MAM formation and inhibited ROS signal transmission to adjacent mitochondria (99). IRE1 in MAMs promotes effectiveness of inositol 1,4,5-trisphosphate receptor, which is responsible for transfer of Ca2+ to mitochondria (100). These mechanisms connect ERS and mitochondrial function, thus affecting the fate of cells.

Autophagy in MIRI

Autophagy is a phenomenon in which cells digest cytoplasm in lysosomes and is key to maintaining normal structure and function of the heart. Autophagy, the primary function of which is to remove and recover misfolded or damaged proteins and organelles, is not only associated with cell survival but also with cell death (101-103). Apoptosis and autophagy are both adaptive responses that are key for cell growth, survival and homeostasis (104). Specifically, apoptosis is type 1 programmed cell death, which involves early degradation of cytoskeleton but preservation of organelles until the late phase, whereas autophagy is type 2 cell death and involves early degradation of organelles but preservation of cytoskeleton until the late stage (105). However, it has been reported that activation of autophagy does not contribute to cell death in MIRI and may serve a protective role (106). A study has indicated that activation of autophagy via chloramphenicol therapy decreases infarct size in a pig model of MIRI (106).

During myocardial ischemia, autophagy degrades non-functional cytoplasmic proteins and provides key nutrients for cell growth and survival, thereby inhibiting apoptosis and necrosis. A recent study suggested that autophagy is necessary to decrease myocardial damage following acute myocardial ischemia and that autophagy limits activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome by removing damaged mitochondria (43). However, it is also reported that excessive autophagy during reperfusion may aggravate the injury of the heart (107,108).

Currently, three forms of autophagy are known: Macroautophagy, microautophagy and chaperone-mediated autophagy (CMA) (109). In macroautophagy, the primary sources of membranes are ER and Golgi bodies. Autophagosomes are formed by completely including the object to be degraded and fusing it with the lysosomal membrane before degradation by lysosomal enzymes (110). In microautophagy, degradation products are directly encapsulated in lysosome membranes for degradation and digestion in lysosomes (111). In CMA, chaperone proteins are bind to the protein to be degraded to guide transport of the substance to lysosomes, which are digested and broken down by enzymes. CMA pathway is a lysosomal process with obvious selectivity, which is different from the aforementioned processes (112).

The molecular mechanism of autophagy is primarily composed of four pathways that involve mTOR complex 1, AMP-activated protein kinase (AMPK), ERS and p53. mTOR and Beclin1 are autophagy-associated molecules that serve key roles in different stages of MIRI. During the ischemic phase, mTOR acts via AMPK/mTOR and PI3K/AKT/mTOR pathways (78), while Beclin1 is upregulated during reperfusion (43,101,102). Beclin1 is a key autophagy protein that regulates formation and processing of autophagosomes. Upregulation of Beclin1 is responsible for autophagy activation during reperfusion (113). However, it is unclear how MIRI activates Beclin1. One potential mechanism is its association with Bcl-2. In vitro study have shown that Beclin1-mediated autophagy to nutritional deficiency (e.g. a lack of amino acids and serum) of cardiomyocytes is regulated by Bcl-2 protein (114). Moreover, ROS may also induce Beclin1-mediated autophagy during reperfusion. During reperfusion, high levels of ROS are not an energy crisis, but a key promoter of autophagy. Reperfusion results in increased oxidative stress with overexpression of Beclin1 (115,116). In addition to regulating expression of Beclin1, ROS may also oxidize and decrease activity of autophagy-associated proteins, leading to lipidation of light chain 3 (LC3) as well as initiation of autophagy. As Beclin1 is primary located in ER (117), it remains to be determined whether reperfusion-induced ERS also participates in upregulation of Beclin1.

Pyroptosis

Pyroptosis is a novel type of proinflammatory programmed cell death that is caused by activation of the NLRP3/apoptosis-associated speck-like protein (ASC)/caspase-1 pathway and high levels of interleukin-1β (IL-1β) (118). The primary biological features of pyroptosis are dependence on caspase-1 and the accompanying inflammatory cascade reaction. Under endogenous and exogenous stimuli, ASC acts on pro-caspase-1 to form inflammasome and activate pro-caspase-1(119). The activated caspase-1 promotes activation and expression of downstream cytokines such as IL-1β and IL-18, resulting in cell and tissue damage (119). It is also a key host mechanism in response to pathogens and endogenous damage (119,120).

Compared with other modes of cell death, pyroptosis has unique features, including activation by intracellular inflammatory caspase, formation of membrane pores and DNA fragmentation/destruction (67). Pyroptosis leads to release of inflammatory cytokines and exhibits certain characteristics of apoptosis (121) (including DNA fragmentation and nuclear concentration) and necrosis (122) (such as loss of plasma membrane integrity and release of intracellular content such as lactate dehydrogenase).

Cell membrane rupture during pyroptosis is mediated by Gasdermin D protein (GSDMD) (67). In healthy cells, GSDMD stays functionally inactive via intramolecular interactions between its COOH-(inhibitory) and NH2-terminal (pro-death) domains. Hydrolysis of inflammatory caspase-1 to GSDMD produces NH2-terminal lysate (GSDMD-NT) (123). GSDMD-NT monomers oligomerize to form pores in the plasma membrane, leading to release of intracellular material and contributing to cell death; however, the underlying mechanism is unknown. GSDMD is also released in pyroptosis without damaging adjacent cells (124). As a novel form of cell death, pyroptosis is associated with development of the mouse model of MIRI (125,126).

ROS produced by mitochondria are hypothesized to serve as a signal for activation of NLRP3 inflammation downstream of MIRI-associated mitochondrial dysfunction (127). In addition, inflammasomes are activated by ROS production and potassium efflux with myocardial ischemia and hypoxia, resulting in activation of caspase-1 and cleavage of GSDMD, which aggravates MIRI (128). A study has indicated that silencing of calpain attenuates myocardial dysfunction caused by MIRI by suppressing activation of the NLRP3/ASC/caspase-1 axis (125). Furthermore, inhibition of microRNA-29a improves MIRI by targeting silent information regulator factor 2-related enzyme 1 via suppressing oxidative stress and NLRP3-mediated pyroptosis (129). Numerous studies have demonstrated that pyroptosis is a key mechanism in exacerbating MIRI (130,131).

Ferroptosis

Ferroptosis, a novel and unique iron-dependent non-apoptosis-regulated form of cell death; it is also called iron-dependent programmed cell death due to the destruction of glutathione-dependent antioxidant defense mechanism and accumulation of lipid peroxides, which are caused by the increased ROS level related to Fe2+ (132,133). Ferroptosis is characterized by the depletion of plasma membrane polyunsaturated fatty acids, condensed mitochondrial membrane densities, and vanishing of mitochondria crista (126). Studies have shown that ferroptosis is involved in regulation of tumor (133), liver cancer (134), Alzheimer's disease (135), cerebral ischemic (136) and acute kidney injury (137) and other types of disease.

Ferroptosis is dependent on intracellular iron and is morphologically, biochemically and genetically distinct from apoptosis, necrosis and autophagy (138). Iron participates in oxidative phosphorylation of mitochondria, thereby increasing ROS levels and ATP production (139). ROS levels that exceed cell antioxidant capacity lead to oxidative stress, which directly or indirectly damages large molecules such as proteins, nucleic acids and lipids, leading to cell damage and death (139-141). The ferric carrier protein transferrin and amino acid glutamine are hypothesized to be responsible for ferroptosis (142). The cell surface transferrin receptor and glutamine-mediated intracellular metabolic pathway glutamine breakdown serve critical roles in cell death (143). Almost all genes involved in ferroptosis are regulated by nuclear factor erythroid 2-related factor 2 transcription, including genes that regulate glutathione and NADPH regeneration, which are key for glutathione peroxidase 4 activity, lipid peroxidation and iron regulation (144).

It is hypothesized that during ischemia and early reperfusion, cellular acidosis, internal environmental instability and other factors promote release of ferrivalent or ferrous ions from enzymes containing iron and sulfur clusters and activate the iron-mediated Fenton reaction, resulting in increased generation of ROS, which leads to oxidative stress injury and ferroptosis of cardiomyocytes (145,146). In addition, deposition of iron in the periinfarct and non-infarct areas has been observed in a mouse model of MI (147). Moreover, Baba et al (148) observed positive iron staining in non-cardiac cells surrounding cardiac scar.

A recent study by our group indicated that myocardial ferroptosis (as well as apoptosis pyroptosis) is increased during MIRI in diabetes and is attenuated by effective treatment with antioxidant N-acetylcysteine (130). However, the interaction and underlying mechanism of apoptosis, pyroptosis and ferroptosis in MIRI have yet to be elucidated.

Necroptosis

In the death receptor pathway, necrosis is induced by activation of receptor-interacting protein kinase 3 (RIPK3), a serine/threonine kinase. RIPK3 is typically activated by phosphorylation of homologous RIPK1(149). RIPK3 phosphorylates and activates pseudokinase mixed lineage kinase domain-like protein (MLKL), which oligomyelizes and permeates the plasma membrane, leading to necrosis (149). Necrosis is mediated by complex IIb including RIPK1, RIPK3, and MLKL. One of the key targets of RIPK3-induced necrosis is pseudokinase MLKL. RIPK3-mediated phosphorylation of MLKL exposes a four-helix bundle in MLKL that promotes cysteine-dependent tetramylation, progression to amyloid filaments and transfer and osmosis to the plasma membrane (150). MLKL phosphorylation is a key for programmed necrosis. Studies have shown that MLKL is the executor of the necrotic effect (150,151). MLKL contains an N-terminal four-helix bundle domain connected to a C-terminal pseudokinase; the helical bundle is the functional domain of MLKL. Under normal conditions, RIPK3 phosphorylates MLKL, resulting in conformational changes that counteract the autophagy effect of the kinase homologous C-terminal region in MLKL (152). MLKL is considered to be the primary target of necrosis but its downstream signaling pathway requires further investigation (153).

Expression levels of necroptosis-associated proteins, including RIPK1, RIPK3 and MLKL, have been shown to be increased in an in vivo MIRI model (154-160). However, reperfusion duration and species-specific susceptibility to cell death should be considered when evaluating necroptosis (161). A study has shown that RIPK3 is also involved in ERS and intracellular Ca2+ overload (162), which aggravates MIRI.

Our previous studies showed that RIP3-mediated necroptosis is a key mechanism of enhanced inflammation and lung tissue injury in high dose lipopolysaccharide-induced severe acute respiratory distress syndrome in mice (153) and in cardiomyocytes with H2O2-induced necroptotic and apoptotic cell death (163). To the best of our knowledge, studies regarding the role and mechanism of RIP3-mediated necroptosis and its interaction with apoptosis and other forms of cell death in MIRI are rare (157,164). Therefore, further studies in this area are required.

3. Conclusion

The present review summarizes research on the pathophysiology of MIRI, including the role of ROS in cell death during MIRI, ischemia- and hypoxia-induced metabolic and functional dysfunction of the electron transport chain in cardiomyocyte mitochondria, mitochondrial disorder caused by decreased ATP production and oxidative stress promoted by increased ROS production caused by mitochondrial damage and electrolyte imbalance during reperfusion. Increased ROS levels lead to cell damage and death via autophagy, apoptosis, programmed cell death inflammation, ferroptosis and necrosis. These interplay with one another, which directly or indirectly leads to aggravation of the effect (Fig. 1).

Figure 1

During ischemia, cardiomyocytes undergo anaerobic metabolism, which results in decreased ATP levels and pH, which increase levels of intracellular Na+ and Ca2+. Reperfusion leads to further increases in intracellular Ca2+ and ROS, which cause MIRI. The primary apoptosis signaling pathways are the intrinsic (mitochondrial) and extrinsic (death receptor) pathway. The intrinsic pathway is induced by release of CytC, which is regulated by Bcl-2 family proteins. The extrinsic pathway is triggered by death receptors (such as TNFR) that activate caspase-8. Programmed necroptosis is mediated by the complex RIP1-RIP3 and MLKL when caspase-8 is inhibited, but the downstream pathway of MLKL remains unclear. Necroptosis and apoptosis have certain commonalities. For instance, caspase-8 decreases MIRI via inhibiting necroptosis by binding to an anti-apoptotic protein (cFLIP). Three forms of autophagy are macroautophagy, microautophagy and chaperone-mediated autophagy. Macroautophagy is the primary pathway, which is promoted by PI3K, AMPK and Beclin1, while AKT/mTOR pathways inhibit this type of autophagy. Beclin1 is abundant in the ER. However, it remains uncertain whether ERS regulates expression of Beclin1 during ischemia or reperfusion. During ischemia and early reperfusion, cellular acidosis, internal environmental instability and other factors promote the release of ferrivalent or ferrous ions from enzymes, thereby activating the Fenton reaction, resulting in increased ROS, which leads to ferroptosis of cardiomyocytes. ROS, reactive oxygen species; MIRI, myocardial ischemia/reperfusion injury; CytC, cytochrome c; TNFR, tumor necrosis factor receptor; RIPK, receptor-interacting protein kinase; MLKL, mixed lineage kinase domain-like protein; cFLIP, CASP8 and FADD-like apoptosis regulator; AMPK, AMP-activated protein kinase; ERS, endoplasmic reticulum stress; JAK, Janus kinase; GFR, growth factor receptor; FADD, Fas-associated death domain; TRADD, tumor necrosis factor receptor type 1-associated death domain protein; MCU, mitochondrial calcium uniporter; mPTP, mitochondrial permeability transition pore; EPO, erythropoietin; ?, unknown mechanism.

To the best of our knowledge, the underlying mechanism of MIRI has not been fully clarified. Among the known mechanisms, explanations for Ca2+ overload, energy metabolism disorder, oxidative stress and autophagy are relatively complete, whereas the role of pyroptosis, necrosis and ferroptosis, as well as downstream molecules of known signaling pathways, need to be further investigated. Clinical trials have shown that infarct size can be limited by non-pharmacological strategies such as ischemic postconditioning and remote ischemic conditioning, drugs, such as cyclosporine, insulin, glucagon-like peptide-1 agonists and β-blockers, or stimulation of cyclic guanosine monophosphate synthesis (165). Our clinical studies have shown that Captopril pretreatment (166) and intraoperative use of antioxidant therapy, such as propofol (167) and Salvia miltiorrhiza (168) alleviate MIRI and improve prognosis in humans. In addition, clinical treatments such as remote ischemic preconditioning (RIPC) attenuate MIRI and improve short-term prognosis. A key cardioprotective mechanism of RIPC is associated with decreased opening of mPTP in the heart (36,169).

MIRI is a complex process involving multilevel and multifactor interactions between genes, molecules, cells and tissue. A full understanding of the pathogenesis and mechanisms underlying development of pathophysiology may provide novel therapeutic targets for improving the prognosis of MIRI and decreasing mortality associated with cardiovascular disease.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by The National Natural Science Foundation of China (grant no. 81970247) and the Basic and Applied Basic research of Guangdong Province (grant no. 2019A1515110732).

Availability of data and materials

Not applicable.

Authors' contributions

JH and DL drafted the manuscript. LXZ, JR and DZ collated and checked references. DL, JH and DZ prepared the figure. LQZ and ZX edited the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

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 

Moreira PVL, de Arruda Neta ADCP, Ferreira SS, Ferreira FELL, de Lima RLFC, de Toledo Vianna RP, de Araújo JM, de Alencar Rodrigues RE, da Silva Neto JM and O'Flaherty M: Coronary heart disease and stroke mortality trends in Brazil 2000-2018. PLoS One. 16(e253639)2021.PubMed/NCBI View Article : Google Scholar

2 

Roger VL, Go AS, Lloyd-Jones DM, Adams RJ, Berry JD, Brown TM, Carnethon MR, Dai S, de Simone G, Ford ES, et al: Heart disease and stroke statistics-2011 update: A report from the American Heart Association. Circulation. 123:e18–e209. 2011.PubMed/NCBI View Article : Google Scholar

3 

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

4 

Ogura Y, Ouchi N, Ohashi K, Shibata R, Kataoka Y, Kambara T, Kito T, Maruyama S, Yuasa D, Matsuo K, et al: Therapeutic impact of follistatin-like 1 on myocardial ischemic injury in preclinical models. Circulation. 126:1728–1738. 2012.PubMed/NCBI View Article : Google Scholar

5 

Verma S, Fedak PW, Weisel RD, Butany J, Rao V, Maitland A, Li RK, Dhillon B and Yau TM: Fundamentals of reperfusion injury for the clinical cardiologist. Circulation. 105:2332–2336. 2002.PubMed/NCBI View Article : Google Scholar

6 

Jennings RB, Sommers HM, Smyth GA, Flack HA and Linn H: Myocardial necrosis induced by temporary occlusion of a coronary artery in the dog. Arch Pathol. 70:68–78. 1960.PubMed/NCBI

7 

Heusch G: Myocardial stunning and hibernation revisited. Nat Rev Cardiol. 18:522–536. 2021.PubMed/NCBI View Article : Google Scholar

8 

Caiazzo G, Musci RL, Frediani L, Umińska J, Wanha W, Filipiak KJ, Kubica J and Navarese EP: State of the art: No-Reflow phenomenon. Cardiol Clin. 38:563–573. 2020.PubMed/NCBI View Article : Google Scholar

9 

Nagao K, Ooiwa K and Kanmatsuse K: Reperfusion arrhythmia. Ryoikibetsu Shokogun Shirizu. 277–281. 1996.PubMed/NCBI(In Japanese).

10 

Yellon DM and Hausenloy DJ: Myocardial reperfusion injury. N Engl J Med. 357:1121–1135. 2007.PubMed/NCBI View Article : Google Scholar

11 

Yang CF: Clinical manifestations and basic mechanisms of myocardial ischemia/reperfusion injury. Ci Ji Yi Xue Za Zhi. 30:209–215. 2018.PubMed/NCBI View Article : Google Scholar

12 

Javat D, Heal C, Banks J, Buchholz S and Zhang Z: Regional to tertiary inter-hospital transfer versus in-house percutaneous coronary intervention in acute coronary syndrome. PLoS One. 13(e198272)2018.PubMed/NCBI View Article : Google Scholar

13 

Li Y, Li Y, Li B, Liu Y, Zhang J, Kuang W, Lu J, Cao Z, Cui J, Fan Z, et al: Antiplatelet therapy with integrated traditional Chinese and western medicine for use in myocardial Ischemia-Reperfusion injury: A review of clinical applications and mechanisms. Evid Based Complement Alternat Med. 2021(7409094)2021.PubMed/NCBI View Article : Google Scholar

14 

Li Q, Shen L, Wang Z, Jiang HP and Liu LX: Tanshinone IIA protects against myocardial ischemia reperfusion injury by activating the PI3K/Akt/mTOR signaling pathway. Biomed Pharmacother. 84:106–114. 2016.PubMed/NCBI View Article : Google Scholar

15 

Hotchkiss RS, Strasser A, McDunn JE and Swanson PE: Cell death. N Engl J Med. 361:1570–1583. 2009.PubMed/NCBI View Article : Google Scholar

16 

Kalogeris T, Baines CP, Krenz M and Korthuis RJ: Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 298:229–317. 2012.PubMed/NCBI View Article : Google Scholar

17 

Li R, Jia Z and Trush MA: Defining ROS in biology and medicine. React Oxyg Species (Apex). 1:9–21. 2016.PubMed/NCBI View Article : Google Scholar

18 

González-Montero J, Brito R, Gajardo AI and Rodrigo R: Myocardial reperfusion injury and oxidative stress: Therapeutic opportunities. World J Cardiol. 10:74–86. 2018.PubMed/NCBI View Article : Google Scholar

19 

Kandula V, Kosuru R, Li H, Yan D, Zhu Q, Lian Q, Ge RS, Xia Z and Irwin MG: Forkhead box transcription factor 1: Role in the pathogenesis of diabetic cardiomyopathy. Cardiovasc Diabetol. 15(44)2016.PubMed/NCBI View Article : Google Scholar

20 

Li H, Xia Z, Chen Y, Qi D and Zheng H: Mechanism and therapies of oxidative Stress-Mediated cell death in ischemia reperfusion injury. Oxid Med Cell Longev. 2018(2910643)2018.PubMed/NCBI View Article : Google Scholar

21 

Xia Z, Chen Y, Fan Q and Xue M: Oxidative stress-mediated reperfusion injury: Mechanism and therapies. Oxid Med Cell Longev. 2014(373081)2014.PubMed/NCBI View Article : Google Scholar

22 

Xia Z, Chen Y, Fan Q, Xue M and Liu KX: Oxidative stress-mediated reperfusion injury 2014. Oxid Med Cell Longev. 2015(689416)2015.PubMed/NCBI View Article : Google Scholar

23 

García N, Zazueta C and Aguilera-Aguirre L: Oxidative stress and inflammation in cardiovascular disease. Oxid Med Cell Longev. 2017(5853238)2017.PubMed/NCBI View Article : Google Scholar

24 

Goldhaber JI and Weiss JN: Oxygen free radicals and cardiac reperfusion abnormalities. Hypertension. 20:118–1127. 1992.PubMed/NCBI View Article : Google Scholar

25 

Brookes PS, Yoon Y, Robotham JL, Anders MW and Sheu SS: Calcium, ATP, and ROS: A mitochondrial love-hate triangle. Am J Physiol Cell Physiol. 287:C817–C833. 2004.PubMed/NCBI View Article : Google Scholar

26 

Chen Y, Liu C, Zhou P, Li J, Zhao X, Wang Y, Chen R, Song L, Zhao H and Yan H: Coronary endothelium No-Reflow injury is associated with ROS-Modified mitochondrial fission through the JNK-Drp1 signaling pathway. Oxid Med Cell Longev. 2021(6699516)2021.PubMed/NCBI View Article : Google Scholar

27 

Pena E, Brito J, El Alam S and Siques P: Oxidative stress, kinase activity and inflammatory implications in right ventricular hypertrophy and heart failure under hypobaric hypoxia. Int J Mol Sci. 21(6421)2020.PubMed/NCBI View Article : Google Scholar

28 

Cadenas S: ROS and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection. Free Radic Biol Med. 117:76–89. 2018.PubMed/NCBI View Article : Google Scholar

29 

Kura B, Szeiffova BB, Kalocayova B, Sykora M and Slezak J: Oxidative stress-responsive MicroRNAs in heart injury. Int J Mol Sci. 21(358)2020.PubMed/NCBI View Article : Google Scholar

30 

Wu MY, Yiang GT, Liao WT, Tsai AP, Cheng YL, Cheng PW, Li CY and Li CJ: Current mechanistic concepts in ischemia and reperfusion injury. Cell Physiol Biochem. 46:1650–1667. 2018.PubMed/NCBI View Article : Google Scholar

31 

Li H, Yao W, Liu Z, Xu A, Huang Y, Ma XL, Irwin MG and Xia Z: Hyperglycemia abrogates ischemic postconditioning cardioprotection by impairing AdipoR1/Caveolin-3/STAT3 signaling in diabetic rats. Diabetes. 65:942–955. 2016.PubMed/NCBI View Article : Google Scholar

32 

Su W, Zhang Y, Zhang Q, Xu J, Zhan L, Zhu Q, Lian Q, Liu H, Xia ZY, Xia Z and Lei S: N-acetylcysteine attenuates myocardial dysfunction and postischemic injury by restoring caveolin-3/eNOS signaling in diabetic rats. Cardiovasc Diabetol. 15(146)2016.PubMed/NCBI View Article : Google Scholar

33 

Jeddi S, Gheibi S, Kashfi K, Carlström M and Ghasemi A: Protective effect of intermediate doses of hydrogen sulfide against myocardial ischemia-reperfusion injury in obese type 2 diabetic rats. Life Sci. 256(117855)2020.PubMed/NCBI View Article : Google Scholar

34 

Wang T, Mao X, Li H, Qiao S, Xu A, Wang J, Lei S, Liu Z, Ng KF, Wong GT, et al: N-Acetylcysteine and allopurinol up-regulated the Jak/STAT3 and PI3K/Akt pathways via adiponectin and attenuated myocardial postischemic injury in diabetes. Free Radic Biol Med. 63:291–303. 2013.PubMed/NCBI View Article : Google Scholar

35 

Xue R, Lei S, Xia ZY, Wu Y, Meng Q, Zhan L, Su W, Liu H, Xu J, Liu Z, et al: Selective inhibition of PTEN preserves ischaemic post-conditioning cardioprotection in STZ-induced Type 1 diabetic rats: Role of the PI3K/Akt and JAK2/STAT3 pathways. Clin Sci (Lond). 130:377–392. 2016.PubMed/NCBI View Article : Google Scholar

36 

Wu Q, Wang T, Chen S, Zhou Q, Li H, Hu N, Feng Y, Dong N, Yao S and Xia Z: Cardiac protective effects of remote ischaemic preconditioning in children undergoing tetralogy of fallot repair surgery: A randomized controlled trial. Eur Heart J. 39:1028–1037. 2018.PubMed/NCBI View Article : Google Scholar

37 

Wang TT, Shi MM, Liao XL, Li YQ, Yuan HX, Li Y, Liu X, Ning DS, Peng YM, Yang F, et al: Overexpression of inducible nitric oxide synthase in the diabetic heart compromises ischemic postconditioning. J Mol Cell Cardiol. 129:144–153. 2019.PubMed/NCBI View Article : Google Scholar

38 

Liu Y, Paterson M, Baumgardt SL, Irwin MG, Xia Z, Bosnjak ZJ and Ge ZD: Vascular endothelial growth factor regulation of endothelial nitric oxide synthase phosphorylation is involved in isoflurane cardiac preconditioning. Cardiovasc Res. 115:168–178. 2019.PubMed/NCBI View Article : Google Scholar

39 

Trafford AW, Díaz ME, Negretti N and Eisner DA: Enhanced Ca2+ current and decreased Ca2+ efflux restore sarcoplasmic reticulum Ca2+ content after depletion. Circ Res. 81:477–4784. 1997.PubMed/NCBI View Article : Google Scholar

40 

Eisner D, Bode E, Venetucci L and Trafford A: Calcium flux balance in the heart. J Mol Cell Cardiol. 58:110–117. 2013.PubMed/NCBI View Article : Google Scholar

41 

Chen S and Li S: The Na+/Ca²+ exchanger in cardiac ischemia/reperfusion injury. Med Sci Monit. 18:RA161–RA165. 2012.PubMed/NCBI View Article : Google Scholar

42 

Murphy E and Steenbergen C: Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol Rev. 88:581–609. 2008.PubMed/NCBI View Article : Google Scholar

43 

Aghaei M, Motallebnezhad M, Ghorghanlu S, Jabbari A, Enayati A, Rajaei M, Pourabouk M, Moradi A, Alizadeh AM and Khori V: Targeting autophagy in cardiac ischemia/reperfusion injury: A novel therapeutic strategy. J Cell Physiol. 234:16768–16778. 2019.PubMed/NCBI View Article : Google Scholar

44 

Hotta Y, Ishikawa N, Ohashi N and Matsui K: Effects of SM-20550, a selective Na+-H+ exchange inhibitor, on the ion transport of myocardial mitochondria. Mol Cell Biochem. 219:83–90. 2001.PubMed/NCBI View Article : Google Scholar

45 

Talukder MA, Zweier JL and Periasamy M: Targeting calcium transport in ischaemic heart disease. Cardiovasc Res. 84:345–352. 2009.PubMed/NCBI View Article : Google Scholar

46 

Chen C, Lu W, Wu G, Lv L, Chen W, Huang L, Wu X, Xu N and Wu Y: Cardioprotective effects of combined therapy with diltiazem and superoxide dismutase on myocardial ischemia-reperfusion injury in rats. Life Sci. 183:50–59. 2017.PubMed/NCBI View Article : Google Scholar

47 

Kook H, Hong SJ, Yang KS, Lee S, Kim JS and Park CG: Comparison of nebivolol versus diltiazem in improving coronary artery spasm and quality of life in patients with hypertension and vasospastic angina: A prospective, randomized, double-blind pilot study. PLoS One. 15(e239039)2020.PubMed/NCBI View Article : Google Scholar

48 

Lodha AR, Pillai A, Sheth K and Hiremath J: A retrospective cohort study exploring diltiazem as a pharmaco-enhancer for tacrolimus, in a post-heart transplant setting. Clin Transplant. 34(e14100)2020.PubMed/NCBI View Article : Google Scholar

49 

Bou-Teen D, Kaludercic N, Weissman D, Turan B, Maack C, Di Lisa F and Ruiz-Meana M: Mitochondrial ROS and mitochondria-targeted antioxidants in the aged heart. Free Radic Biol Med. 167:109–124. 2021.PubMed/NCBI View Article : Google Scholar

50 

Yu J, Wu J, Xie P, Maimaitili Y, Wang J, Xia Z, Gao F, Zhang X and Zheng H: Sevoflurane postconditioning attenuates cardiomyocyte hypoxia/reoxygenation injury via restoring mitochondrial morphology. Peerj. 4(e2659)2016.PubMed/NCBI View Article : Google Scholar

51 

Boengler K, Kosiol M, Mayr M, Schulz R and Rohrbach S: Mitochondria and ageing: Role in heart, skeletal muscle and adipose tissue. J Cachexia Sarcopenia Muscle. 8:349–369. 2017.PubMed/NCBI View Article : Google Scholar

52 

Fieni F, Johnson DE, Hudmon A and Kirichok Y: Mitochondrial Ca2+ uniporter and CaMKII in heart. Nature. 513:E1–E2. 2014.PubMed/NCBI View Article : Google Scholar

53 

Santulli G, Xie W, Reiken SR and Marks AR: Mitochondrial calcium overload is a key determinant in heart failure. Proc Natl Acad Sci USA. 112:11389–11394. 2015.PubMed/NCBI View Article : Google Scholar

54 

Cheng Y, Xia Z, Han Y and Rong J: Plant natural product formononetin protects rat cardiomyocyte h9c2 cells against oxygen glucose deprivation and reoxygenation via inhibiting ROS formation and promoting GSK-3β phosphorylation. Oxid Med Cell Longev. 2016(2060874)2016.PubMed/NCBI View Article : Google Scholar

55 

Shintani-Ishida K, Inui M and Yoshida K: Ischemia-reperfusion induces myocardial infarction through mitochondrial Ca²+ overload. J Mol Cell Cardiol. 53:233–239. 2012.PubMed/NCBI View Article : Google Scholar

56 

Kulek AR, Anzell A, Wider JM, Sanderson TH and Przyklenk K: Mitochondrial quality control: Role in cardiac models of lethal ischemia-reperfusion injury. Cells. 9(214)2020.PubMed/NCBI View Article : Google Scholar

57 

Chang JC, Lien CF, Lee WS, Chang HR, Hsu YC, Luo YP, Jeng JR, Hsieh JC and Yang KT: Intermittent hypoxia prevents myocardial mitochondrial Ca 2+ Overload and cell death during ischemia/reperfusion: The role of reactive oxygen species. Cells-Basel. 8(564)2019.PubMed/NCBI View Article : Google Scholar

58 

Obeng E: Apoptosis (programmed cell death) and its signals-A review. Braz J Biol. 81:1133–1143. 2021.PubMed/NCBI View Article : Google Scholar

59 

Slee EA, Adrain C and Martin SJ: Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J Biol Chem. 276:7320–7326. 2001.PubMed/NCBI View Article : Google Scholar

60 

Schlegel RA and Williamson P: Phosphatidylserine, a death knell. Cell Death Differ. 8:551–563. 2001.PubMed/NCBI View Article : Google Scholar

61 

Xu X, Lai Y and Hua ZC: Apoptosis and apoptotic body: Disease message and therapeutic target potentials. Biosci Rep. 39(BSR20180992)2019.PubMed/NCBI View Article : Google Scholar

62 

D'Arcy MS: Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 43:582–592. 2019.PubMed/NCBI View Article : Google Scholar

63 

Dong Y, Chen H, Gao J, Liu Y, Li J and Wang J: Molecular machinery and interplay of apoptosis and autophagy in coronary heart disease. J Mol Cell Cardiol. 136:27–41. 2019.PubMed/NCBI View Article : Google Scholar

64 

Wajant H: The Fas signaling pathway: More than a paradigm. Science. 296:1635–1636. 2002.PubMed/NCBI View Article : Google Scholar

65 

Kischkel FC, Hellbardt S, Behrmann I, Germer M, Pawlita M, Krammer PH and Peter ME: Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 14:5579–5588. 1995.PubMed/NCBI View Article : Google Scholar

66 

Teringova E and Tousek P: Apoptosis in ischemic heart disease. J Transl Med. 15(87)2017.PubMed/NCBI View Article : Google Scholar

67 

Del Re DP, Amgalan D, Linkermann A, Liu Q and Kitsis RN: Fundamental mechanisms of regulated cell death and implications for heart disease. Physiol Rev. 99:1765–1817. 2019.PubMed/NCBI View Article : Google Scholar

68 

Condorelli G, Roncarati R, Ross JJ Jr, Pisani A, Stassi G, Todaro M, Trocha S, Drusco A, Gu Y, Russo MA, et al: Heart-targeted overexpression of caspase3 in mice increases infarct size and depresses cardiac function. Proc Natl Acad Sci USA. 98:9977–9982. 2001.PubMed/NCBI View Article : Google Scholar

69 

Lee P, Sata M, Lefer DJ, Factor SM, Walsh K and Kitsis RN: Fas pathway is a critical mediator of cardiac myocyte death and MI during ischemia-reperfusion in vivo. Am J Physiol Heart Circ Physiol. 284:H456–H463. 2003.PubMed/NCBI View Article : Google Scholar

70 

Jeremias I, Kupatt C, Martin-Villalba A, Habazettl H, Schenkel J, Boekstegers P and Debatin KM: Involvement of CD95/Apo1/Fas in cell death after myocardial ischemia. Circulation. 102:915–920. 2000.PubMed/NCBI View Article : Google Scholar

71 

Kosuru R, Cai Y, Kandula V, Yan D, Wang C, Zheng H, Li Y, Irwin MG, Singh S and Xia Z: AMPK contributes to cardioprotective effects of pterostilbene against myocardial ischemia-reperfusion injury in diabetic rats by suppressing cardiac oxidative stress and apoptosis. Cell Physiol Biochem. 46:1381–1397. 2018.PubMed/NCBI View Article : Google Scholar

72 

Forini F, Kusmic C, Nicolini G, Mariani L, Zucchi R, Matteucci M, Iervasi G and Pitto L: Triiodothyronine prevents cardiac ischemia/reperfusion mitochondrial impairment and cell loss by regulating miR30a/p53 axis. Endocrinology. 155:4581–4590. 2014.PubMed/NCBI View Article : Google Scholar

73 

Lin CL, Tseng HC, Chen RF, Chen WP, Su MJ, Fang KM and Wu ML: Intracellular zinc release-activated ERK-dependent GSK-3β-p53 and Noxa-Mcl-1 signaling are both involved in cardiac ischemic-reperfusion injury. Cell Death Differ. 18:1651–1663. 2011.PubMed/NCBI View Article : Google Scholar

74 

Zhang C, Shi J, Qian L, Zhang C, Wu K, Yang C, Yan D, Wu X and Liu X: Nucleostemin exerts anti-apoptotic function via p53 signaling pathway in cardiomyocytes. In Vitro Cell Dev Biol Anim. 51:1064–1071. 2015.PubMed/NCBI View Article : Google Scholar

75 

Li M, Wang D, He J, Chen L and Li H: Bcl-XL: A multifunctional anti-apoptotic protein. Pharmacol Res. 151(104547)2020.PubMed/NCBI View Article : Google Scholar

76 

Hochhauser E, Kivity S, Offen D, Maulik N, Otani H, Barhum Y, Pannet H, Shneyvays V, Shainberg A, Goldshtaub V, et al: Bax ablation protects against myocardial ischemia-reperfusion injury in transgenic mice. Am J Physiol Heart Circ Physiol. 284:H2351–H2359. 2003.PubMed/NCBI View Article : Google Scholar

77 

Chen Z, Chua CC, Ho YS, Hamdy RC and Chua BH: Overexpression of Bcl-2 attenuates apoptosis and protects against myocardial I/R injury in transgenic mice. Am J Physiol Heart Circ Physiol. 280:H2313–H2320. 2001.PubMed/NCBI View Article : Google Scholar

78 

Zhang D, He Y, Ye X, Cai Y, Xu J, Zhang L, Li M, Liu H, Wang S and Xia Z: Activation of autophagy inhibits nucleotide-binding oligomerization domain-like receptor protein 3 inflammasome activation and attenuates myocardial ischemia-reperfusion injury in diabetic rats. J Diabetes Investig. 11:1126–1136. 2020.PubMed/NCBI View Article : Google Scholar

79 

Cai Y, Ying F, Liu H, Ge L, Song E, Wang L, Zhang D, Hoi CTE, Xia Z and Irwin MG: Deletion of Rap1 protects against myocardial ischemia/reperfusion injury through suppressing cell apoptosis via activation of STAT3 signaling. FASEB J. 34:4482–4496. 2020.PubMed/NCBI View Article : Google Scholar

80 

Peng K, Chen WR, Xia F, Liu H, Meng XW, Zhang J, Liu HY, Xia ZY and Ji FH: Dexmedetomidine post-treatment attenuates cardiac ischaemia/reperfusion injury by inhibiting apoptosis through HIF-1α signalling. J Cell Mol Med. 24:850–861. 2020.PubMed/NCBI View Article : Google Scholar

81 

Gao S, Wang R, Dong S, Wu J, Perek B, Xia Z, Yao S and Wang T: Inactivation of TOPK caused by hyperglycemia blocks diabetic heart sensitivity to sevoflurane postconditioning by impairing the PTEN/PI3K/Akt signaling. Oxid Med Cell Longev. 2021(6657529)2021.PubMed/NCBI View Article : Google Scholar

82 

Pang L, Cai Y, Tang EH, Yan D, Kosuru R, Li H, Irwin MG, Ma H and Xia Z: Cox-2 inhibition protects against hypoxia/reoxygenation-induced cardiomyocyte apoptosis via Akt-dependent enhancement of iNOS expression. Oxid Med Cell Longev. 2016(3453059)2016.PubMed/NCBI View Article : Google Scholar

83 

Korshunova AY, Blagonravov ML, Neborak EV, Syatkin SP, Sklifasovskaya AP, Semyatov SM and Agostinelli E: BCL2-regulated apoptotic process in myocardial ischemia-reperfusion injury (Review). Int J Mol Med. 47:23–36. 2021.PubMed/NCBI View Article : Google Scholar

84 

Liu J, Liu M and Chen L: Novel pathogenesis: Regulation of apoptosis by Apelin/APJ system. Acta Biochim Biophys Sin (Shanghai). 49:471–478. 2017.PubMed/NCBI View Article : Google Scholar

85 

Qi Z and Chen L: Endoplasmic reticulum stress and autophagy. Adv Exp Med Biol. 1206:167–177. 2019.PubMed/NCBI View Article : Google Scholar

86 

Sanderson TH, Gallaway M and Kumar R: Unfolding the unfolded protein response: Unique insights into brain ischemia. Int J Mol Sci. 16:7133–7142. 2015.PubMed/NCBI View Article : Google Scholar

87 

Chen X, Wang Y, Xie X, Chen H, Zhu Q, Ge Z, Wei H, Deng J, Xia Z and Lian Q: Heme oxygenase-1 reduces Sepsis-Induced endoplasmic reticulum stress and acute lung injury. Mediators Inflamm. 2018(9413876)2018.PubMed/NCBI View Article : Google Scholar

88 

Zhang YM, Wang CY, Zheng FC, Gao FF, Chen YC, Huang ZQ, Xia ZY, Irwin MG, Li WQ, Liu XP, et al: Effects of N-n-butyl haloperidol iodide on the rat myocardial sarcoplasmic reticulum Ca(2+)-ATPase during ischemia/reperfusion. Biochem Biophys Res Commun. 425:426–430. 2012.PubMed/NCBI View Article : Google Scholar

89 

Liu Y, Baumgardt SL, Fang J, Shi Y, Qiao S, Bosnjak ZJ, Vásquez-Vivar J, Xia Z, Warltier DC, Kersten JR and Ge ZD: Transgenic overexpression of GTP cyclohydrolase 1 in cardiomyocytes ameliorates post-infarction cardiac remodeling. Sci Rep. 7(3093)2017.PubMed/NCBI View Article : Google Scholar

90 

Su RY, Geng XY, Yang Y and Yin HS: Nesfatin-1 inhibits myocardial ischaemia/reperfusion injury through activating Akt/ERK pathway-dependent attenuation of endoplasmic reticulum stress. J Cell Mol Med. 25:5050–5059. 2021.PubMed/NCBI View Article : Google Scholar

91 

Vekich JA, Belmont PJ, Thuerauf DJ and Glembotski CC: Protein disulfide isomerase-associated 6 is an ATF6-inducible ER stress response protein that protects cardiac myocytes from ischemia/reperfusion-mediated cell death. J Mol Cell Cardiol. 53:259–267. 2012.PubMed/NCBI View Article : Google Scholar

92 

Glembotski CC: Roles for ATF6 and the sarco/endoplasmic reticulum protein quality control system in the heart. J Mol Cell Cardiol. 71:11–15. 2014.PubMed/NCBI View Article : Google Scholar

93 

Dai B, Qiao L, Zhang M, Cheng L, Zhang L, Geng L, Shi C, Zhang M, Sui C, Shen W, et al: LncRNA AK054386 functions as a ceRNA to sequester miR-199 and induce sustained endoplasmic reticulum stress in hepatic reperfusion injury. Oxid Med Cell Longev. 2019(8189079)2019.PubMed/NCBI View Article : Google Scholar

94 

Gao P, Yan Z and Zhu Z: Mitochondria-Associated endoplasmic reticulum membranes in cardiovascular diseases. Front Cell Dev Biol. 8(604240)2020.PubMed/NCBI View Article : Google Scholar

95 

Zhou J, Ahmad F, Parikh S, Hoffman NE, Rajan S, Verma VK, Song J, Yuan A, Shanmughapriya S, Guo Y, et al: Loss of adult cardiac myocyte GSK-3 leads to mitotic catastrophe resulting in fatal dilated cardiomyopathy. Circ Res. 118:1208–1222. 2016.PubMed/NCBI View Article : Google Scholar

96 

Zeeshan HM, Lee GH, Kim HR and Chae HJ: Endoplasmic reticulum stress and associated ROS. Int J Mol Sci. 17(327)2016.PubMed/NCBI View Article : Google Scholar

97 

Zhang G, Wang X, Gillette TG, Deng Y and Wang ZV: Unfolded protein response as a therapeutic target in cardiovascular disease. Curr Top Med Chem. 19:1902–1917. 2019.PubMed/NCBI View Article : Google Scholar

98 

Song S, Tan J, Miao Y and Zhang Q: Crosstalk of ER stress-mediated autophagy and ER-phagy: Involvement of UPR and the core autophagy machinery. J Cell Physiol. 233:3867–3874. 2018.PubMed/NCBI View Article : Google Scholar

99 

Verfaillie T, Rubio N, Garg AD, Bultynck G, Rizzuto R, Decuypere JP, Piette J, Linehan C, Gupta S, Samali A and Agostinis P: PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 19:1880–1891. 2012.PubMed/NCBI View Article : Google Scholar

100 

Carreras-Sureda A, Jaña F, Urra H, Durand S, Mortenson DE, Sagredo A, Bustos G, Hazari Y, Ramos-Fernández E, Sassano ML, et al: Non-canonical function of IRE1α determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat Cell Biol. 21:755–767. 2019.PubMed/NCBI View Article : Google Scholar

101 

Shi B, Ma M, Zheng Y, Pan Y and Lin X: MTOR and Beclin1: Two key autophagy-related molecules and their roles in myocardial ischemia/reperfusion injury. J Cell Physiol. 234:12562–12568. 2019.PubMed/NCBI View Article : Google Scholar

102 

Wang S, Wang C, Yan F, Wang T, He Y, Li H, Xia Z and Zhang Z: N-Acetylcysteine attenuates diabetic myocardial ischemia reperfusion injury through inhibiting excessive autophagy. Mediators Inflamm. 2017(9257291)2017.PubMed/NCBI View Article : Google Scholar

103 

Klionsky DJ, Abdel-Aziz AK, Abdelfatah S, Abdellatif M, Abdoli A, Abel S, Abeliovich H, Abildgaard MH, Abudu YP, Acevedo-Arozena A, et al: Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition)1. Autophagy. 17:1–382. 2021.PubMed/NCBI View Article : Google Scholar

104 

Ouyang C, You J and Xie Z: The interplay between autophagy and apoptosis in the diabetic heart. J Mol Cell Cardiol. 71:71–80. 2014.PubMed/NCBI View Article : Google Scholar

105 

Levine B and Yuan J: Autophagy in cell death: An innocent convict? J Clin Invest. 115:2679–2688. 2005.PubMed/NCBI View Article : Google Scholar

106 

Sala-Mercado JA, Wider J, Undyala VV, Jahania S, Yoo W, Mentzer RJ, Gottlieb RA and Przyklenk K: Profound cardioprotection with chloramphenicol succinate in the swine model of myocardial ischemia-reperfusion injury. Circulation. 122 (11 Suppl):S179–S184. 2010.PubMed/NCBI View Article : Google Scholar

107 

Wang Y, Zhou L, Su W, Huang F, Zhang Y, Xia ZY, Xia Z and Lei S: Selective inhibition of PKCβ2 restores ischemic Postconditioning-Mediated cardioprotection by modulating autophagy in diabetic rats. J Diabetes Res. 2020(2408240)2020.PubMed/NCBI View Article : Google Scholar

108 

Chen Z, Hu Z, Lu Z, Cai S, Gu X, Zhuang H, Ruan Z, Xia Z, Irwin MG, Feng D and Zhang L: Differential microRNA profiling in a cellular hypoxia reoxygenation model upon posthypoxic propofol treatment reveals alterations in autophagy signaling network. Oxid Med Cell Longev. 2013(378484)2013.PubMed/NCBI View Article : Google Scholar

109 

Abdrakhmanov A, Gogvadze V and Zhivotovsky B: To Eat or to Die: Deciphering selective forms of autophagy. Trends Biochem Sci. 45:347–364. 2020.PubMed/NCBI View Article : Google Scholar

110 

Cong Y, Dinesh KN, Mauthe M, Verlhac P and Reggiori F: Manipulation of selective macroautophagy by pathogens at a glance. J Cell Sci. 133(jcs240440)2020.PubMed/NCBI View Article : Google Scholar

111 

Schuck S: Microautophagy-distinct molecular mechanisms handle cargoes of many sizes. J Cell Sci. 133(jcs246322)2020.PubMed/NCBI View Article : Google Scholar

112 

Yang Q, Wang R and Zhu L: Chaperone-Mediated autophagy. Adv Exp Med Biol. 1206:435–452. 2019.PubMed/NCBI View Article : Google Scholar

113 

Zhu H and He L: Beclin 1 biology and its role in heart disease. Curr Cardiol Rev. 11:229–237. 2015.PubMed/NCBI View Article : Google Scholar

114 

Brady NR, Hamacher-Brady A, Yuan H and Gottlieb RA: The autophagic response to nutrient deprivation in the hl-1 cardiac myocyte is modulated by Bcl-2 and sarco/endoplasmic reticulum calcium stores. FEBS J. 274:3184–3197. 2007.PubMed/NCBI View Article : Google Scholar

115 

Ma X, Liu H, Foyil SR, Godar RJ, Weinheimer CJ, Hill JA and Diwan A: Impaired autophagosome clearance contributes to cardiomyocyte death in ischemia/reperfusion injury. Circulation. 125:3170–3181. 2012.PubMed/NCBI View Article : Google Scholar

116 

Ma X, Liu H, Foyil SR, Godar RJ, Weinheimer CJ and Diwan A: Autophagy is impaired in cardiac ischemia-reperfusion injury. Autophagy. 8:1394–1396. 2012.PubMed/NCBI View Article : Google Scholar

117 

Manganelli V, Matarrese P, Antonioli M, Gambardella L, Vescovo T, Gretzmeier C, Longo A, Capozzi A, Recalchi S, Riitano G, et al: Raft-like lipid microdomains drive autophagy initiation via AMBRA1-ERLIN1 molecular association within MAMs. Autophagy. 17:2528–2548. 2021.PubMed/NCBI View Article : Google Scholar

118 

Sun L, Ma W, Gao W, Xing Y, Chen L, Xia Z, Zhang Z and Dai Z: Propofol directly induces caspase-1-dependent macrophage pyroptosis through the NLRP3-ASC inflammasome. Cell Death Dis. 10(542)2019.PubMed/NCBI View Article : Google Scholar

119 

Ball DP, Taabazuing CY, Griswold AR, Orth EL, Rao SD, Kotliar IB, Vostal LE, Johnson DC and Bachovchin DA: Caspase-1 interdomain linker cleavage is required for pyroptosis. Life Sci Alliance. 3(e202000664)2020.PubMed/NCBI View Article : Google Scholar

120 

Toldo S, Mauro AG, Cutter Z and Abbate A: Inflammasome, pyroptosis, and cytokines in myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 315:H1553–H1568. 2018.PubMed/NCBI View Article : Google Scholar

121 

Tsuchiya K: Inflammasome-associated cell death: Pyroptosis, apoptosis, and physiological implications. Microbiol Immunol. 64:252–269. 2020.PubMed/NCBI View Article : Google Scholar

122 

Frank D and Vince JE: Pyroptosis versus necroptosis: Similarities, differences, and crosstalk. Cell Death Differ. 26:99–114. 2019.PubMed/NCBI View Article : Google Scholar

123 

Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H and Lieberman J: Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 535:153–158. 2016.PubMed/NCBI View Article : Google Scholar

124 

He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, Yang ZH, Zhong CQ and Han J: Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res. 25:1285–1298. 2015.PubMed/NCBI View Article : Google Scholar

125 

Yue RC, Lu SZ, Luo Y, Wang T, Liang H, Zeng J, Liu J and Hu HX: Calpain silencing alleviates myocardial ischemia-reperfusion injury through the NLRP3/ASC/Caspase-1 axis in mice. Life Sci. 233(116631)2019.PubMed/NCBI View Article : Google Scholar

126 

Djulbegovic MB and Uversky VN: Ferroptosis-an iron- and disorder-dependent programmed cell death. Int J Biol Macromol. 135:1052–1069. 2019.PubMed/NCBI View Article : Google Scholar

127 

Liu Q, Zhang D, Hu D, Zhou X and Zhou Y: The role of mitochondria in NLRP3 inflammasome activation. Mol Immunol. 103:115–124. 2018.PubMed/NCBI View Article : Google Scholar

128 

Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, et al: Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 123:594–604. 2011.PubMed/NCBI View Article : Google Scholar

129 

Ding S, Liu D, Wang L, Wang G and Zhu Y: Inhibiting MicroRNA-29a protects myocardial Ischemia-Reperfusion injury by targeting SIRT1 and suppressing oxidative stress and NLRP3-Mediated pyroptosis pathway. J Pharmacol Exp Ther. 372:128–135. 2020.PubMed/NCBI View Article : Google Scholar

130 

Wang C, Zhu L, Yuan W, Sun L, Xia Z, Zhang Z and Yao W: Diabetes aggravates myocardial ischaemia reperfusion injury via activating Nox2-related programmed cell death in an AMPK-dependent manner. J Cell Mol Med. 24:6670–6679. 2020.PubMed/NCBI View Article : Google Scholar

131 

Popov SV, Maslov LN, Naryzhnaya NV, Mukhomezyanov AV, Krylatov AV, Tsibulnikov SY, Ryabov VV, Cohen MV and Downey JM: The role of pyroptosis in ischemic and reperfusion injury of the heart. J Cardiovasc Pharmacol Ther. 26:562–574. 2021.PubMed/NCBI View Article : Google Scholar

132 

Yang WS and Stockwell BR: Ferroptosis: Death by lipid peroxidation. Trends Cell Biol. 26:165–176. 2016.PubMed/NCBI View Article : Google Scholar

133 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012.PubMed/NCBI View Article : Google Scholar

134 

Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R and Tang D: Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 63:173–184. 2016.PubMed/NCBI View Article : Google Scholar

135 

Yan N and Zhang J: Iron metabolism, ferroptosis, and the links with alzheimer's disease. Front Neurosci. 13(1443)2019.PubMed/NCBI View Article : Google Scholar

136 

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

137 

Hu Z, Zhang H, Yang SK, Wu X, He D, Cao K and Zhang W: Emerging role of ferroptosis in acute kidney injury. Oxid Med Cell Longev. 2019(8010614)2019.PubMed/NCBI View Article : Google Scholar

138 

Yu H, Guo P, Xie X, Wang Y and Chen G: Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J Cell Mol Med. 21:648–657. 2017.PubMed/NCBI View Article : Google Scholar

139 

Su LJ, Zhang JH, Gomez H, Murugan R, Hong X, Xu D, Jiang F and Peng ZY: Reactive oxygen Species-Induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid Med Cell Longev. 2019(5080843)2019.PubMed/NCBI View Article : Google Scholar

140 

Stoyanovsky DA, Tyurina YY, Shrivastava I, Bahar I, Tyurin VA, Protchenko O, Jadhav S, Bolevich SB, Kozlov AV, Vladimirov YA, et al: Iron catalysis of lipid peroxidation in ferroptosis: Regulated enzymatic or random free radical reaction? Free Radic Biol Med. 133:153–1561. 2019.PubMed/NCBI View Article : Google Scholar

141 

Zhao WK, Zhou Y, Xu TT and Wu Q: Ferroptosis: Opportunities and challenges in myocardial Ischemia-Reperfusion injury. Oxid Med Cell Longev. 2021(9929687)2021.PubMed/NCBI View Article : Google Scholar

142 

Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, Zhu N, Yin X, et al: Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 116:2672–2680. 2019.PubMed/NCBI View Article : Google Scholar

143 

Gao M, Monian P, Quadri N, Ramasamy R and Jiang X: Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 59:298–308. 2015.PubMed/NCBI View Article : Google Scholar

144 

Ge ZD, Lian Q, Mao X and Xia Z: Current status and challenges of NRF2 as a potential therapeutic target for diabetic cardiomyopathy. Int Heart J. 60:512–520. 2019.PubMed/NCBI View Article : Google Scholar

145 

Williams RE, Zweier JL and Flaherty JT: Treatment with deferoxamine during ischemia improves functional and metabolic recovery and reduces reperfusion-induced oxygen radical generation in rabbit hearts. Circulation. 83:1006–1014. 1991.PubMed/NCBI View Article : Google Scholar

146 

Drossos G, Lazou A, Panagopoulos P and Westaby S: Deferoxamine cardioplegia reduces superoxide radical production in human myocardium. Ann Thorac Surg. 59:169–172. 1995.PubMed/NCBI View Article : Google Scholar

147 

Omiya S, Hikoso S, Imanishi Y, Saito A, Yamaguchi O, Takeda T, Mizote I, Oka T, Taneike M, Nakano Y, et al: Downregulation of ferritin heavy chain increases labile iron pool, oxidative stress and cell death in cardiomyocytes. J Mol Cell Cardiol. 46:59–66. 2009.PubMed/NCBI View Article : Google Scholar

148 

Baba Y, Higa JK, Shimada BK, Horiuchi KM, Suhara T, Kobayashi M, Woo JD, Aoyagi H, Marh KS, Kitaoka H and Matsui T: Protective effects of the mechanistic target of rapamycin against excess iron and ferroptosis in cardiomyocytes. Am J Physiol Heart Circ Physiol. 314:H659–H668. 2018.PubMed/NCBI View Article : Google Scholar

149 

Murphy JM, Czabotar PE, Hildebrand JM, Lucet IS, Zhang JG, Alvarez-Diaz S, Lewis R, Lalaoui N, Metcalf D, Webb AI, et al: The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity. 39:443–453. 2013.PubMed/NCBI View Article : Google Scholar

150 

Wu J, Huang Z, Ren J, Zhang Z, He P, Li Y, Ma J, Chen W, Zhang Y, Zhou X, et al: Mlkl knockout mice demonstrate the indispensable role of Mlkl in necroptosis. Cell Res. 23:994–1006. 2013.PubMed/NCBI View Article : Google Scholar

151 

Sun L, Wang H, Wang Z, He S, Chen S, Liao D, Wang L, Yan J, Liu W, Lei X and Wang X: Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell. 148:213–227. 2012.PubMed/NCBI View Article : Google Scholar

152 

Li L, Tong A, Zhang Q, Wei Y and Wei X: The molecular mechanisms of MLKL-dependent and MLKL-independent necrosis. J Mol Cell Biol. 13:3–14. 2021.PubMed/NCBI View Article : Google Scholar

153 

Wang L, Wang T, Li H, Liu Q, Zhang Z, Xie W, Feng Y, Socorburam T, Wu G, Xia Z and Wu Q: Receptor interacting protein 3-Mediated necroptosis promotes Lipopolysaccharide-Induced inflammation and acute respiratory distress syndrome in mice. PLoS One. 11(e155723)2016.PubMed/NCBI View Article : Google Scholar

154 

Oberst A, Dillon CP, Weinlich R, McCormick LL, Fitzgerald P, Pop C, Hakem R, Salvesen GS and Green DR: Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature. 471:363–367. 2011.PubMed/NCBI View Article : Google Scholar

155 

Dillon CP, Weinlich R, Rodriguez DA, Cripps JG, Quarato G, Gurung P, Verbist KC, Brewer TL, Llambi F, Gong YN, et al: RIPK1 blocks early postnatal lethality mediated by caspase-8 and RIPK3. Cell. 157:1189–1202. 2014.PubMed/NCBI View Article : Google Scholar

156 

Dannappel M, Vlantis K, Kumari S, Polykratis A, Kim C, Wachsmuth L, Eftychi C, Lin J, Corona T, Hermance N, et al: RIPK1 maintains epithelial homeostasis by inhibiting apoptosis and necroptosis. Nature. 513:90–94. 2014.PubMed/NCBI View Article : Google Scholar

157 

Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, Liu Y, Zheng W, Shang H, Zhang J, et al: CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 22:175–182. 2016.PubMed/NCBI View Article : Google Scholar

158 

Bai J, Wang Q, Qi J, Yu H, Wang C, Wang X, Ren Y and Yang F: Promoting effect of baicalin on nitric oxide production in CMECs via activating the PI3K-AKT-eNOS pathway attenuates myocardial ischemia-reperfusion injury. Phytomedicine. 63(153035)2019.PubMed/NCBI View Article : Google Scholar

159 

Zhou H, Li D, Zhu P, Ma Q, Toan S, Wang J, Hu S, Chen Y and Zhang Y: Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemia-reperfusion injury. J Pineal Res. 65(e12503)2018.PubMed/NCBI View Article : Google Scholar

160 

Yang J, Zhang F, Shi H, Gao Y, Dong Z, Ma L, Sun X, Li X, Chang S, Wang Z, et al: Neutrophil-derived advanced glycation end products-Nε-(carboxymethyl) lysine promotes RIP3-mediated myocardial necroptosis via RAGE and exacerbates myocardial ischemia/reperfusion injury. FASEB J. 33:14410–14422. 2019.PubMed/NCBI View Article : Google Scholar

161 

Ying L, Benjanuwattra J, Chattipakorn SC and Chattipakorn N: The role of RIPK3-regulated cell death pathways and necroptosis in the pathogenesis of cardiac ischaemia-reperfusion injury. Acta Physiol (Oxf). 231(e13541)2021.PubMed/NCBI View Article : Google Scholar

162 

Zhu P, Hu S, Jin Q, Li D, Tian F, Toan S, Li Y, Zhou H and Chen Y: Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol. 16:157–168. 2018.PubMed/NCBI View Article : Google Scholar

163 

Yin W, Wang C, Peng Y, Yuan W, Zhang Z, Liu H, Xia Z, Ren C and Qian J: Dexmedetomidine alleviates H2O2-induced oxidative stress and cell necroptosis through activating of α2-adrenoceptor in H9C2 cells. Mol Biol Rep. 47:3629–3639. 2020.PubMed/NCBI View Article : Google Scholar

164 

Liu X, Zhang D, Dong X, Zhu R, Ye Y, Li L and Jiang Y: Pharmacological activation of CB2 receptor protects against ethanol-induced myocardial injury related to RIP1/RIP3/MLKL-mediated necroptosis. Mol Cell Biochem. 474:1–14. 2020.PubMed/NCBI View Article : Google Scholar

165 

Garcia-Dorado D, Rodríguez-Sinovas A, Ruiz-Meana M and Inserte J: Protection against myocardial ischemia-reperfusion injury in clinical practice. Rev Esp Cardiol (Engl Ed). 67:394–404. 2014.PubMed/NCBI View Article : Google Scholar

166 

Tian Y, Li H, Liu P, Xu JM, Irwin MG, Xia Z and Tian G: Captopril pretreatment produces an additive cardioprotection to isoflurane preconditioning in attenuating myocardial ischemia reperfusion injury in rabbits and in humans. Mediators Inflamm. 2015(819232)2015.PubMed/NCBI View Article : Google Scholar

167 

Huang Z, Zhong X, Irwin MG, Ji S, Wong GT, Liu Y, Xia ZY, Finegan BA and Xia Z: Synergy of isoflurane preconditioning and propofol postconditioning reduces myocardial reperfusion injury in patients. Clin Sci (Lond). 121:57–69. 2011.PubMed/NCBI View Article : Google Scholar

168 

Xia Z, Gu J, Ansley DM, Xia F and Yu J: Antioxidant therapy with Salvia miltiorrhiza decreases plasma endothelin-1 and thromboxane B2 after cardiopulmonary bypass in patients with congenital heart disease. J Thorac Cardiovasc Surg. 126:1404–1410. 2003.PubMed/NCBI View Article : Google Scholar

169 

Heusch G: Molecular basis of cardioprotection: Signal transduction in ischemic pre-, post-, and remote conditioning. Circ Res. 116:674–699. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2022
Volume 23 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He J, Liu D, Zhao L, Zhou D, Rong J, Zhang L and Xia Z: Myocardial ischemia/reperfusion injury: Mechanisms of injury and implications for management (Review). Exp Ther Med 23: 430, 2022
APA
He, J., Liu, D., Zhao, L., Zhou, D., Rong, J., Zhang, L., & Xia, Z. (2022). Myocardial ischemia/reperfusion injury: Mechanisms of injury and implications for management (Review). Experimental and Therapeutic Medicine, 23, 430. https://doi.org/10.3892/etm.2022.11357
MLA
He, J., Liu, D., Zhao, L., Zhou, D., Rong, J., Zhang, L., Xia, Z."Myocardial ischemia/reperfusion injury: Mechanisms of injury and implications for management (Review)". Experimental and Therapeutic Medicine 23.6 (2022): 430.
Chicago
He, J., Liu, D., Zhao, L., Zhou, D., Rong, J., Zhang, L., Xia, Z."Myocardial ischemia/reperfusion injury: Mechanisms of injury and implications for management (Review)". Experimental and Therapeutic Medicine 23, no. 6 (2022): 430. https://doi.org/10.3892/etm.2022.11357