Open Access

BCL2‑regulated apoptotic process in myocardial ischemia‑reperfusion injury (Review)

  • Authors:
    • Anna Yu. Korshunova
    • Mikhail L. Blagonravov
    • Ekaterina V. Neborak
    • Sergey P. Syatkin
    • Anastasia P. Sklifasovskaya
    • Said M. Semyatov
    • Enzo Agostinelli
  • View Affiliations

  • Published online on: November 4, 2020     https://doi.org/10.3892/ijmm.2020.4781
  • Pages: 23-36
  • Copyright: © Korshunova 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 leading cause of death in developed countries is cardiovascular disease, where coronary heart disease is the main cause of death. Myocardial reperfusion is the most significant method to prevent cell death after ischemia. However, restoration of blood flow may paradoxically lead to myocardial ischemia‑reperfusion injury (MI/RI) accompanied by metabolic disturbances and cardiomyocyte death. As the myocardium has an extremely limited ability to regenerate, the mechanisms of regulated cell death, including apoptosis, are the most significant for contemporary research due to their reversibility. BCL2 is a key anti‑apoptotic protein. There are several signaling pathways and compounds regulating BCL2, including PI3K/AKT and MEK1/ERK1/2, JAK2/STAT3, endothelial nitric oxide synthase, PTEN, cardiac ankyrin repeat protein and microRNA, which can serve as targets for modern methods of cardioprotective therapy inhibiting intrinsic apoptosis and saving viable cardiomyocytes after MI/RI. The present review considers the mechanisms of Bcl2‑regulated apoptosis in the development and treatment of MI/RI.

1. Introduction

In 1972, the Austrian pathologist J.F. Kerr, in cooperation with his Scottish colleagues A.H. Wyllie and A.R. Currie, introduced the concept of 'apoptosis' (after the ancient Greek ἀπόπτωσις-leaf fall) to describe a morphologically stereotypical form of cell death characterized by cytoplasmic volume depletion, chromatin condensation and margination, shrinkage of the nucleus (pyknosis), fragmentation of the nucleus (karyorhexis), blebbing of the membranes and formation of discrete apoptotic bodies with an undamaged cell membrane (1,2).

According to the contemporary biochemical classification of Nomenclature Committee on Cell Death, apoptosis is considered to be one of the morphological signs typical for different types of regulated cell death (RCD) (3). One of the forms of RCD is intrinsic apoptosis. Intrinsic apoptosis, initiated by the cell itself in response to intracellular damage, is also known as mitochondrial apoptosis, as the mitochondria performs the key role in this process (4). The trigger event is the increase in mitochondrial outer membrane permeabilization (MOMP) and release of proteins that are normally sequestered between the two mitochondrial membranes (5,6). The MOMP and thus the entire process of intrinsic apoptosis is regulated by members of the BCL2 protein family that are embedded in the outer membrane (6,7).

BCL2 is an acronym for B-cell lymphoma/leukemia-2. As its name suggests, the gene expressing BCL2 was for the first time found in B-cell malignant neoplasms. This acronym is also used for the designation of the entire family of homological proteins (8). Different proteins of this family contain BCL2 homology domains (BH: BH1, BH2, BH3 and BH4) (Fig. 1) (9) and can be divided into two groups: Pro-apoptotic and anti-apoptotic. Pro-apoptotic proteins include BCL2-associated X protein (BAX), (BCL2 antagonist/killer (BAK), BCL2-related ovarian killer, BH3 interacting domain death agonist, BCL2-associated agonist of cell death, BCL2-interacting killer, BCL2-interacting mediator of cell death), BCL2-modifying factor, activator of apoptosis harakiri, BCL2-interacting protein 3 (ANIP3), NIX (BNIP3-like), phorbol-12-myristate-13-acetate-induced protein 1 (NOXA) and p53 upregulated modulator of apoptosis (PUMA). Meanwhile, anti-apoptotic proteins include BCL2, BCL2 X-linked protein (BCL-XL), myeloid cell leukemia 1 and BCL-w and A1/BFL-1 (10-12). The BCL2 family proteins are capable of interacting with each other, whereby their different partnerships result in different outcome of the cell fate (9). In response to apoptosis stimulation, BAX and BAK proteins are exposed to oligomerization on the mitochondrial outer membrane (13,14). This process is blocked by BCL2 protein, which inhibits mitochondrial permeabilization and cell death by interacting with BAX and BAK (9,15). Enhanced expression of Bcl2 may increase cell resistance to apoptosis in cells, such as tumor cells. The BCL2/BAX ratio is a type of 'rheostat' regulating cell death depending on the balance between BCL2 and BAX in cells (16).

Cardiomyocyte apoptosis is a well-known key process during the development of ischemia (17) During apoptosis inhibition, the BCL2/BAX ratio is increased, which contributes to cardiomyocyte survival in the peri-infarct area (18). Previous investigations revealed a significant role of abnormal Bcl2 expression in cardiomyocyte apoptosis modulation in MI/RI, as its expression rate has a direct effect on cardiomyocyte apoptosis and cardiac function (19,20).

The key object in the clinical treatment of MI/RI based on molecular mechanisms of the injury progression is decreasing the rate of cardiomyocyte apoptosis. BCL2 is the key protein in the entire BCL2 family that is responsible for the anti-apoptotic process and promotion of cell survival. Therefore, the present review focused mainly on this protein and aimed to investigate how it is regulated during MI/RI and how this can be exploited for clinical use. The present review also assessed possible ways of using BCL2 as a target for pharmacological correction.

2. Pathogenesis of myocardial ischemia-reperfusion injury (MI/RI)

Cardiovascular diseases are the main cause of death world-wide. In 2016, 85% of cases resulted from myocardial infarction or cerebral stroke (21,22). Coronary heart disease is the main cause of death and disabilities (23). Myocardial infarction is tissue necrosis following acute ischemia, which is characterized by absolute insufficiency of coronary blood circulation (24).

Ischemia is a complex pathological mechanism resulting from a decrease in the local blood flow in a tissue or organ (25). Ischemia occurs commonly in the myocardium due to occlusion of the coronary arteries responsible for myocardial perfusion (25). The heart is a constantly contracting organ, requiring a high rate of metabolic activity, which makes it extremely susceptible to any disorders of oxygen supply. Under normal conditions, mitochondria consume oxygen and generate ATP. A decrease in oxygen supply leads to the inhibition of mitochondrial oxidative phosphorylation and, consequentially, the switch from aerobic to anaerobic metabolism (26). Anaerobic glycolysis causes a reduction of intracellular pH (26). The combination of enhanced sodium and calcium influx into cells, due to Na+-H+ and Na+-Ca2+ exchange, correspondingly increases acidity and intracellular calcium levels (27). Moreover, a rapid elevation in intracellular Ca2+ leads to a pathological increase in mitochondrial permeability transition; however, a reduction of intracellular pH inhibits this process (27). Disordered ion homeostasis is followed by osmotic gradient formation, which is accompanied by water inflow into the cell with a subsequent swelling and disturbance in intracellular ion balance (28). If blood supply is not properly restored after ischemia, the absence of sufficient ATP levels and high levels of Ca2+ lead to myocyte atrophy and eventually apoptosis and necrosis (28). The activation of caspase-3 and maximal activity of pro-apoptotic proteins BAX, Noxa and PUMA are observed on the 1st day post-coronary artery occlusion; however, anti-apoptotic proteins BCL2 and BCL-XL remain relatively unchanged, which indicated that the pro-apoptotic pathways are activated rapidly in MI/RI while cell protective pathways remain inactive (29).

Reperfusion of the stunned myocardium during percutaneous coronary intervention is necessary to minimize myocardial damage. For patients with myocardial infarction accompanied by elevation of ST-segment, the timely reperfusion of the myocardium using either thrombolytic therapy or primary percutaneous coronary intervention, is the most effective method of treatment to restrict the size of infarction area, support systolic function and reduce manifestations of heart failure (30). Reperfusion therapy of coronary insufficiency after myocardial infarction is also the most effective method to save cardiomyocytes suffering from hypoxia, support cardiac function and save patients' lives (31). Reperfusion is the most significant method to prevent tissue death after ischemia. However, restoration of blood flow can paradoxically lead to MI/RI, characterized by metabolic disturbances, local inflammatory response, cell death and a consequent cardiac remodeling and dysfunction, contributing to adverse cardiac events after myocardial ischemia (25,32,33). Although reperfusion is necessary for the restoration of oxygen and nutrient influx, which supports cellular metabolism, it may paradoxically cause consequent pathological processes aggravating tissue damage (34,35). MI/RI may exacerbate structural and functional disturbances of the myocardium and cause a strong effect on the restoration of cardiac function after recurrent reperfusion (35-37).

The phenomenon of paradoxical aggravation after oxygen flux restoration was described for the first time >50 years ago when it was shown that reperfusion caused several pathological changes in heart exposed to coronary occlusion (26). MI/RI is associated with different pathophysiological mechanisms, including calcium overload, production of oxygen free radicals, endothelial dysfunction, immune response, mitochondrial dysfunction, cardiomyocyte apoptosis and autophagy and platelet aggregation (38-41). During this process, apoptosis is the main pathological mechanism, which plays a critical role in cardiac remodeling after myocardial infarction (42).

Cardiomyocyte apoptosis and necrosis caused by MI/RI are the most critical pathological processes in cases of cardiac dysfunction after previous myocardial infarction (43). Myocardial necrosis is predominantly observed at the late stages of MI/RI while cell apoptosis is observed throughout the whole process (43). Apoptosis is one of the most important mechanisms of MI/RI and it has a considerable effect on the degree of damage and consequently on the prognosis of heart failure development (42). Thus, effective inhibition of apoptosis caused by MI/RI is one of the important lines of research and it is of great importance for cardiac function improvement after myocardial infarction and for preventing myocardial remodeling.

Apoptosis plays a critical role in the pathogenesis of MI/RI (44). Inhibition of apoptosis may decrease the degree of myocardial damage and prevent injury caused by MI/RI (45). Mitochondrial injury accompanying hypoxia contributes to a decrease in BCL2 content and opening the mitochondrial permeability transition pore (MPTP) (46,47). Reverse blockade of electron transport in ischemia supports high levels of BCL2 accompanied by a decrease in susceptibility to MPTP opening after ischemia (5). Functional inhibition of BCL2 using its low-molecular antagonist HA14-1 sensitizes MPTP opening in mitochondria under normal physiological conditions (46). These results indicated a potential link between decreased or inhibited function of BCL2 and MPTP opening in MI/RI. It can be hypothesized that the BCL2 protein family governs cells undergoing apoptosis. In this context, investigating the regulation of BCL2 during MI/RI may be beneficial in revealing pathways with a potential for possible clinical application.

3. The main pathways of BCL2 regulation in MI/RI

The BCL2 protein family regulates cardiomyocyte death in MI/RI (48,49). The synthesis of BAX and caspase-3 is significantly enhanced and production of BCL2 is inhibited during MI/RI (50). Studies have shown that the key role in apoptotic initiation is due to oxidative stress (51,52).

Two main types of protein activity regulation are known: Fast regulation via post-translational modification (usually phosphorylation/dephosphorylation) and slow regulation via gene expression regulation. BCL2 was shown to have three basic sites of phosphorylation (T69, S70 and S87) which results in changes in its anti-apoptotic activity (11). The modulating role of BCL2 phosphorylation remains to be fully elucidated, moreover, there are contradicting facts described in literature which can derive from the feasibility of single phosphorylation of different amino acids or triple phosphorylation of all three amino acids in the structure of BCL2 (10,46,53). Moreover, BCL2 phosphorylation of the same type in normal and cancer cells can lead to different effects. An attempt to clarify these contradictions was undertaken by Song et al (53), who managed to build a mathematical model for BCL2 phosphorylation in different types of cancer cells and revealed that the turning point was 50% triple phosphorylation (T69, S70 and S87) that switched BCL2 from apoptotic to anti-apoptotic action.

The limitation of this conclusion is that it can only be reliably applied to cancer cells.

Several kinases that have BCL2 as a target for phosphorylation are well described in literature: Protein kinase C α, JNK, p38/MAPK, ERK and pyruvate kinase isoform M2 (PKM2). Dephosphorylation of phosphorylated (p)-BCL2 is performed by protein phosphatase A2. BCL2 phosphorylation mediated by JNK, p38/MAPK and PKM2 was shown to occur in cardiomyocytes. JNK and p38/MAPK inactivate BCL2 by phosphorylating and inducing apoptosis, causing cardiomyocyte injury after ischemia and during oxidative stress (54,55). By contrast, PKM2 phosphorylates BCL2 with the aid of heat shock protein 90 to prevent its degradation, thus enhancing its stability and promoting its anti-apoptotic properties (56). Several publications link the degree between BCL2 triple phosphorylation with the crosstalk between autophagy and apoptosis (57-59). This switch point is feasible due to different affinities of BCL2 and p-BCL2 to beclin-1 as the main autophagy inducer (57). Thus, phosphorylation of BCL2 leads to the dissociation of beclin-1 from the BCL2-beclin-1 complex with consequent phosphorylation of beclin-1 and the formation of an active PI3K III complex and autophagy induction (57). The lower degree of BCL2 phosphorylation resulted in autophagy induction, while more extensive BCL2 phosphorylation reduced its affinity to BAX, causing its dissociation and thus resulting in apoptosis induction (58).

Other mechanisms of BCL2 regulation involve gene expression and result in changes in BCL2 intracellular levels. Several signaling pathways are known to regulate the rate of intrinsic apoptosis including PI3K/AKT and MEK1-ERK1/2, endothelial nitric oxide synthase (eNOS), PTEN and JAK2/STAT3 (59-65) (Fig. 2).

The reperfusion injury salvage kinase (RISK) pathway was described for the first time by Schulman et al (59) in 2002, while they were studying the mechanisms underlying the cardioprotective effect caused by urocortin. The RISK pathway is a combination of two parallel cascades: PI3K/AKT and MEK1/ERK1/2. The pathways were analyzed in detail in a series of subsequent pharmacological experiments in which the protective effect of several interventions was blocked by a simultaneous administration of PI3K and ERK inhibitors at different times (60). In the broadest term, RISK refers to the group of pro-survival protein kinases responsible for cardio-protection via specific activation during reperfusion.

The PI3K/AKT/mTOR signaling pathway is an important regulatory mechanism for protein synthesis and is closely associated with intracellular oxidation and reduction in the mitochondria (61). It was found that stress in vitro and in vivo may lead to an increase in the rate of tyrosine receptor phosphorylation which activates PI3K, indirectly stimulating AKT phosphorylation, increasing the rate of p-mTOR and activating the expression of the anti-apoptotic factor Bcl2 (61,62). It was also shown that the levels of PI3K, p-AKT and p-mTOR in rat myocardial cells after MI/RI were significantly lower compared with the controls (62). Following MI/RI, expression levels of caspase-3 and Bax were significantly increased in myocardial cells whereas Bcl2 expression significantly decreased (63).

The conformation of PI3K can be changed and activated by the action of growth factors and mitogens, which convert phosphatidylinositoldiphospate 2 (PIP2) into phosphatidylino-sitoltriphospate 3 (PIP3) (63). Several studies demonstrated that the PI3K/AKT signaling pathway may facilitate cell apoptosis in case of MI/RI by influencing the BCL2/BAX ratio (64,65). Zhang et al (66) showed that the PI3K/AKT/mTOR signaling pathway is inhibited in the cardiomyocytes of rats with myocardial infarction, which leads to significant activation of cardiomyocyte apoptosis.

A significant role in the regulation of the PI3K/AKT signaling pathway in MI/RI belongs to PTEN which dephosphorylates PIP3 back into PIP2, thus inhibiting the PI3K/AKT signaling pathway (Fig. 2). This protein plays an important role in apoptosis (67). Nevertheless, only a few studies evaluated the role of PTEN in MI/RI experimental models. In particular, it was shown that PTEN inhibition protected the myocardium from MI/RI by activating the PI3K/AKT/eNOS/ERK pathway, which is one of the variants of pro-apoptotic pathway induction (67). An increase of PTEN levels may suppress the activity of the PI3K/AKT signaling pathway, which may cause myocardial cell apoptosis during MI/RI (68). It was also shown that expression of PTEN and BAX levels in myocardial cells in the MI/RI group were markedly higher compared with sham-operated animals, but phosphorylation of AKT and BCL2 levels were significantly lower (69).

ERK1/2 plays a key role in the transduction of extracellular stimuli (70). ERK1/2 acts as an important protein kinase in reperfusion damage (71). Mitogen-activated protein kinase (MEK1 or MAP2K) was shown to hyperactivate the ERK1/2 signaling pathway (72). The ERK1/2 signaling cascade acts as the main regulator of intracellular apoptosis (73). Although the function of ERK1/2 in apoptosis is controversial (74), inhibition of this pathway is associated with a reduction in the number of apoptotic cells and the BAX/BCL2 ratio as well as a decrease in mitochondrial membrane potential and cell viability in MI/RI (75,76).

The anti-apoptotic effects of nitrogen oxide (NO) mediated-cGMP/protein kinase G (PKG) signaling can be associated with increased synthesis of anti-apoptotic BCL2 and inhibition of MPTP formation (77,78). Moreover, NO and natriuretic peptides may prevent cardiomyocyte apoptosis via cGMP/PKG-dependent inhibition of intracellular calcium overload (79).

The JAK/STAT signaling pathway is a key component of the survivor activating factor enhancement (SAFE) pathway, which can transmit cell signals from the plasmalemma to the nucleus, providing regulation of gene expression (80-85). The JAK/STAT pathway plays an important role in different mechanisms in the myocardium, including apoptosis (81,86), MI/RI (87,88), preconditioning (89) and postconditioning (90,91). In 2009, Lecour (92) showed that in addition to the RISK pathway, SAFE can be an alternative pathway mediating signaling activated by post-conditioning. The JAK/STAT pathway consists of the family of receptor-associated cytosol tyrosine kinases, which phosphorylate tyrosine (93). Phosphorylation and activation of signal transducer and activator of transcription (STAT) in response to ischemic preconditioning (IPC) contribute to cardioprotection by means of signaling cascades and inhibition of pro-apoptotic factors (94). STAT3 is a central component of cardioprotection (95,96). Subsequent studies showed that the JAK2/STAT3 signaling pathway takes part in the anti-apoptotic effect of preconditioning, which is realized by increasing the synthesis of anti-apoptotic BCL2 and suppressing the pro-apoptotic protein BAX (90,97).

The inhibition of pathways that increase the BCL2/Bax ratio and enhancement of pathways leading to its lowering is typically observed in MI/RI, which is associated with hypoxic conditions (84). In vitro MI/RI modeling in cardiac myoblasts revealed an increase in BCL2 protein levels accompanied by an increase in p-PI3K and p-AKT levels after antioxidant treatment (94). Cell survival was also increased while the expression of pro-apoptotic BAX was downregulated (98). These results supported the idea that hypoxia-induced oxidative stress acts as a main downregulatory factor for BCL2 and BCL2-family controlled intrinsic apoptosis.

Koeppen et al (99) found that expression of serum pro-inflammatory substances is significantly higher in patients with myocardial infarction compared with healthy people. This is an important factor contributing to disease progression due to apoptosis activation. It was also revealed that the toll-like receptor 4 (TLR4)/NF-κB signaling pathway was a potential therapeutic target for MI/RI treatment (100,101). Several studies showed that the TLR4/NF-κB signaling pathway plays a critical role in the regulation of the inflammatory response and cardiomyocyte apoptosis during MI/RI (102,103).

Cardiac ankyrin repeat protein (CARP), a transcription co-factor regulating gene expression in cardiomyocytes, inhibits apoptosis induced by MI/RI increasing Bcl2 gene expression (104). CARP is linked with the promotor site of the gene Bcl2 through formation of a complex with transcription factor GATA-4 which regulates transcription and enhances cardioprotection (104).

Hyperlipidemia can stimulate the activation of cardio-myocyte apoptosis in MI/RI. Immunocytochemical analysis revealed an increase in the expression of pro-apoptotic Bax and inhibition anti-apoptotic Bcl2 expression in the myocardium of rats exposed to a hypercholesterol diet (105). These results are in agreement with the data obtained by Guo et al (106) and Kuo et al (107). In this model, the levels of pro-apoptotic proteins BAK and BAX are significantly increased, which is a sign of induction of intrinsic apoptosis (108). Hypercholesterinemia is associated with an increase in the BCL2/BAX ratio in the myocardium which leads to the aggravation of myocardial damage after its reperfusion due to the activation of cardiomyocyte apoptosis rate (107). It was also shown in the experiments in Oryctolagus (rabbits) that Bcl2 expression is increased in the myocardium during hypercholesterolemia by 50% compared with the controls (109). In Oryctolagus with hypercholesterolemia and myocardial ischemia, a marked reduction of Bcl2 expression and similar degree of the increase in Bax expression were observed (109).

MicroRNAs (miRs) are one of the most important epigenetic regulators (110). In recent years, several studies revealed the role of miRs in the process of MI/RI (111-119). miRs change the key signaling mechanisms which makes them potential therapeutic targets (111,112). miRs act as transcription regulators in a wide range of biological processes underlying the response to stress, cell proliferation and cell death (113,114). miRs may bind to the 3′-untranslated region of the mRNA of a target gene, hence destroying mRNA or preventing mRNA translation and negatively regulating the expression of the target gene at the post-transcriptional level (115). Disturbances in miR expression or function are closely associated with cardiovascular diseases; miRNAs take part in different pathophysiological processes including myocardial infarction (116), MI/RI (37,117) or cardiac remodeling (118) with a possible role as aggravating (20) or neutralizing agents (37).

For example, miR-1 is predominantly expressed in cardiac myocytes and closely associated with MI/RI in rats as its levels inversely correlate with BCL2 protein synthesis in cardiomyocytes in MI/RI (119). Mice studies also showed that enhancement of miR-135b-5p expression in MI/RI leads to activation of the JAK2/STAT3 signaling pathway, Bax expression and Bcl2 inhibition (120). Hullinger et al (121) demonstrated that miR-15b, a member of the miR-15 family, aggravated myocardial damage caused by MI/RI via affecting BCL2. miR-16 expression is activated during MI/RI and has an inhibiting effect on Bcl2 expression, which contributed to the enhancement of cardiomyocyte apoptosis after MI/RI (122). Inhibition of miR-16 expression may suppress cardiomyocyte apoptosis after MI/RI, resulting in a reduction of infarction area (122). miR-221 is involved in the pathogenesis of MI/RI by regulating the PTEN/AKT signaling pathway, along with Bax and Bcl2 expression (123-125). Expression of Bcl2 and microtubule-associated proteins 1A/1B light chain 3B II in cardiomyocytes of newly born rats is significantly decreased, which is accompanied by enhanced expression of miR-497 in anoxia-reoxygenation (126). Another study revealed the cardioprotective role of mir-21 in MI/RI via the activation of the PTEN/AKT signaling pathway and BCL2 (127). miRNA-22 may inhibit cardiomyocyte apoptosis by inhibiting p53 acetylation and decreasing the levels of pro-apoptotic genes Bax and p21 by affecting one of its targets-cAMP response element-binding protein (128-130). miR-214 reduced myocardial damage caused by MI/RI via the PI3K/AKT signaling pathway, accompanied by a decrease in BAX levels and an increase in BCL2 levels (131). miR-34a, activated in rats with MI/RI, repressed Bcl2 in vivo and in vitro (132).

The regulation of BCL2-dependent apoptosis in MI/RI is quite versatile and depends on a large number of factors, including activation of emergency genetic programs, changes in metabolic processes and the involvement of additional signaling pathways protecting the myocardium from the negative effects of hypoxia. The ability to influence these mechanisms makes it possible to reduce cardiomyocyte damage, also via induction of BCL2.

4. Therapy of MI/RI

Various forms of cell death may occur during acute MI/RI including necrosis, apoptosis, autophagy, necroptosis and pyroptosis, which may influence the terminal size of the myocardial infarction area after MI/RI (3). This may be used as a new target for cardioprotection, which may include the activation of endogenous cardioprotective signaling pathways: Cascade NO/cGMP/PKG, RISK and SAFE pathways, mitochondrial morphology, cardiomyocyte apoptosis and others (77-79).

Cardiomyocytes of adult humans are characterized by an extremely limited regeneration capacity (133). As a result, there is a continuous process of renewal and reparation of cells mediated by different mechanisms, including apoptosis (134).

In the 1990s, studies focused on the role of different types of cell death in cardioprotection after MI/RI (135). Pro-apoptotic proteins were the main subjects of research at the time, where they were considered to be new targets in MI/RI (135). This was based on a hypothesis suggesting a possibility of saving viable cardiomyocytes when the signaling pathway of regulated cell death was potentially interrupted (135). For example, caspase inhibition during reperfusion restricted the size of the myocardial infarction area in animal models (136). Besides preventing cell death by inhibition of pro-apoptotic caspases, the focus was also given to the use of growth factors that prevented apoptotic processes via activation of proteins contributing to cell survival, such as kinases responsible for the survival associated with PI3K and ERK1/2 activation. This method was suggested to be protective against MI/RI (137,138).

However, there are still no effective methods for prevention of MI/RI in patients with myocardial infarction (139). Previous attempts to perform cardioprotective treatment of MI/RI (antioxidants, calcium blockers and anti-inflammatory drugs) were not successful (140). The advantages of growth factors (137,138) was restricted because the signaling pathways they were involved in lead simultaneously to activation of apoptosis and induction of fibrosis (141).

Oxidative stress, Ca2+ overload, pH changes and inflammation during early reperfusion are the main mediators of tissue alteration, which emphasizes the importance of this period for the pathogenesis of MI/RI (142, 143). In canine experiments, the size of the infarction area significantly increased on the 6th to 24th h after reperfusion. However, Argaud et al (144) revealed no difference in the size of the myocardial infarction area between the 4th and 72nd h after reperfusion in Oryctolagus cuniculus. Species differences and particular methods of MI/RI modeling can be referred to as the reasons for such different results (143,144).

Regardless of the success in the research of cardioprotective methods on animals, their use in clinical practice still present with severe difficulties (145-147). Some pharmaceutical approaches faced just little success, and although the suggested methods of ischemic conditioning seem promising, their effects may be minor and, in some cases, even controversial (148). Differences between preclinical models of transient myocardial ischemia and coronary heart disease with specific characteristics in patients including age, concomitant diseases and drug therapy may help explain the difficulties in introducing the potential cardioprotective techniques into clinical practice (149).

Numerous different methods of cardioprotective therapy of MI/RI have been suggested in the past three decades (150). These approaches are commonly based on the controlled use of short-term ischemia and reperfusion (ischemic conditioning), pharmacotherapy or physiotherapy including hypothermia or electric stimulation of nerve terminals (30,140).

Therapeutic methods of MI/RI based on ischemic conditioning include local IPC and ischemic post-conditioning (IPostC) as well as remote ischemic conditioning (140), which delays pH restoration, prevent NOS decomposition and consequent formation of reactive forms of oxygen and nitrogen and also increase the content of PKG, a component of the RISK pathway, and cause enhancement of the SAFE pathway in reperfused cardiomyocytes (151-153). As aforementioned, all these factors regulate BCL2 in MI/RI, indicating cardio-protective effects of ischemic conditioning due to inhibition of BCL2-family dependent apoptosis (Fig. 3). The following part of the review details the exploration of the mechanisms underlying these strategies of MI/RI therapy.

Effects of IPC on BCL2 regulated apoptosis in MI/RI

Murry et al (154) published an original study showing that IPC (several short-term cycles of ischemia and reperfusion) protected tissues from subsequent ischemic stroke. This discovery, described in Canis experiments, was afterwards reproduced in numerous preclinical studies in other animals and other organs besides the heart (155,156) and then in humans (157). The concept of IPC was then transformed into 'ischemic conditioning'-a wide term including a number of associated cardioprotective methods used either directly towards the heart (IPC or IPostC) or distantly (remote ischemic pre-, per- or postconditioning) (157). Thus, effective methods providing the reduction of MI/RI have become an important field of research.

The potential of IPC is inevitably restricted by the necessity to use it before ischemia, which is of great difficulty for patients with myocardial infarction (158). However, this method initiated a number of subsequent studies, which have brought considerable success in understanding the mechanisms underlying MI/RI and IPC as a result of the potential development of cardioprotective therapy (159).

The cardioprotective effect of IPC is evidenced by a decrease in the size of the myocardial infarction area and a reduction in the number of apoptotic cardiomyocytes (157). Activation of the JAK2/STAT3 signaling pathway in response to IPC contributed to cardioprotection via signaling cascades responsible for the inhibition of pro-apoptotic factors (160). Early phase of IPC enhanced JAK/STAT signal transduction by activation of STAT3, which is nearly neutralized by AG490, a JAK2 inhibitor (161). Constitutive deletion of STAT3 stimulated apoptosis, increased the size of infarction area and caused a reduction in cardioprotective effects after pharmacological preconditioning (162).

Studies showed that IPC increased the activity of cyclo-oxygenase-2 and inducible NOS 24 h after intervention, which depends on transcriptional regulation via the JAK/STAT signaling pathway (163,164). Taken together, these observations lead to the conclusion that IPC activated the SAFE pathway (Figs. 2 and 3).

Chen et al (165) investigated the cardioprotective action of exercise preconditioning on periodic cardiomyocyte apoptosis caused by hypoxia in rats. The results of this study showed that 5 days of exercise on a treadmill may decrease the apoptotic index of the myocardium and caspase-3 expression and increase the BCL2/BAX ratio, which indicated cardioprotective effects based on suppression of hypoxia-induced cardiomyocyte apoptosis. Based on previous studies, exercise preconditioning significantly reduced myocardial damage caused by physical load during ischemia, which is associated with lower levels of cardiac troponin I (cTnI) in the serum, a decrease in the size of the myocardial infarction area, suppression of cardiomyocyte apoptosis, an increase in the levels of anti-apoptotic protein BCL2 and a decrease in the activity of caspase-3 (165). These results are evidence of the cardioprotective action of preconditioning from MI/RI and accompanying apoptosis (160).

Effects of IPostC on BCL2 regulated apoptosis in MI/RI

IPostC was first described Zhao et al (166). IPostC, which is induced by short-term episodes of ischemia-reperfusion at the beginning of reperfusion can restrict MI/RI by the activation of intrinsic signaling cascade reactions.

Restoration of myocardial blood circulation caused by postconditioning improved the contractile function of the myocardium and also restricts the size of infarction area, which is confirmed by a lower serum concentration of creatine kinase (CK) and the activity of lactate dehydrogenase compared with the data obtained after MI/RI without previous postconditioning (167,168).

The effectiveness of IPostC as a method of myocardial protection from MI/RI was also confirmed in several other studies. IPostC does not only decrease the size of the infarction area (143,167) but also limits cardiomyocyte apoptosis after reperfusion. Budhram-Mahadeo et al (29) showed that IPostC stimulated BCL2 synthesis and inhibited BAX production. Another study demonstrated the ability of IPostC, similar to IPC, to restrict cardiomyocyte apoptosis after reperfusion via the SAFE pathway (169). IPostC activated STAT3 after reperfusion, and a JAK2 inhibitor (AG490) suppressed the anti-apoptotic effects of IPostC (170). The anti-apoptotic effects of the JAK2-STAT3 signaling pathway were demonstrated in several studies performed on tumors (171). Several genes encoding proteins mediating apoptosis, such as Bcl2 and Bcl-xl, were identified as target genes for STAT3 (170,171). Notably, an increase in BCL2 levels is typical for the period between the 2nd and 24th h after reperfusion in IPostC (166). IPostC might inhibit cardiomyocyte apoptosis during long-term reperfusion via regulation of anti-apoptotic factors such as BCL2 (167). A long-term anti-apoptotic effect of IPostC may be associated with an increase in BCL2 levels 24 h after reperfusion, which is controlled by JAK2/STAT3 (167). Moreover, the PI3K/AKT signaling pathway, regulated by JAK2 signaling, is necessary for cardioprotection of IPostC (169).

An increase in the expression of AKT and BCL2 proteins is accompanied by inhibition of BAX synthesis, which is a sign of activation of the PI3K/AKT signaling pathway and inhibition of cardiomyocyte apoptosis (44). Activation of this pathway, as the main component of the RISK pathway, prevented cardiomyocyte apoptosis, protected the myocardium from MI/RI and plays a critical role in IPostC effects (172-174). Goodman et al (175) demonstrated that JAK/STAT signaling may contribute to the initiation of RISK signal transduction via activation of PI3K/AKT, and JAK/STAT signaling alone, without subsequent activation of RISK, is not sufficient for cardioprotection after IPostC. Other studies showed that JAK2 signaling regulated the activation of the PI3K/AKT pathway after IPostC (169). Blocking the PI3K/AKT pathway decreased the cardioprotective effects of IPostC at every timepoint (169). Activation of the JAK2/STAT3/BCL2 pathway without activation of the PI3K/AKT pathway may be insufficient for apoptosis limitation (169).

The positive effects of IPostC may be inhibited by a high-cholesterol diet (109). Moreover, hypercholesterolemia inhibited the phosphorylation of AKT and ERK1/2, which were activated by IPostC in the myocardium and also caused excessive apoptosis due to inhibition of BCL2, increased levels of cytochrome c and enhanced activities of caspases 9 and 3 (176).

Effects of pharmacotherapy on BCL2-regulated apoptosis in MI/RI

In recent years, there is an increasing interest in studying the pharmacological methods of cardioprotection (150). The ultimate objectives of cardioprotection strategies include molecular targets mainly involved in signaling pathways of regulated cell death such as ion channels, proteases, reactive oxygen species, contractile elements or components of MPTP (141). As a rule, these strategies are based on existing medicines and they rarely undergo pre-clinical trials (140). The only exclusion is cyclosporine A, which is targeted at MPTP. However, cyclosporine A showed controversial results and failed in clinical trials (140).

Although pharmacotherapy is not commonly included in cardioprotective strategies, several investigations have shown that a number of medicines are capable of cushioning the effects of MI/RI (176,177,179-182,186-190). The present review briefly reviews those that promote cell survival and reduce apoptosis by affecting the BCL2/Bax ratio through specific signaling pathways. Most of the agents provide pleiotropic effects and activate several pathways simultaneously, leading to an increase of BCL2 expression (179-182,186-190). The comparative data on these medicines is summarized in Table I.

Table I

Effects of several pharmaceuticals on apoptosis via affecting BCL2 expression.

Table I

Effects of several pharmaceuticals on apoptosis via affecting BCL2 expression.

Name of compoundMolecular targetReferences
MetforminSTEAP4 AMPK activation176,177
BerberineJAK/STAT activation179
EscitalopramND180
Ilexsaponin AND181
Salvianolic acidCaspase-3 inhibition50
SevofluraneMicroRNA-135b-5p suppression119
Dexmedetomidineα2-adrenoreceptors, PI3K/AKT/GSK-3β182
RapamycinMAPK, JAK2/STAT3 activation183-186
MelatoninAMPK/PGC-1α/SIRT3, Notch1/Hes1 activation187-190,193

[i] STEAP4, metalloreductase STEAP4; ND, not determined; GSK-3β, glycogen synthase kinase-3β; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator-1α; SIRT3, NAD-dependent protein deacetylase sirtuin-3.

Metformin, which is widely used for the treatment of carbohydrate metabolism disorders, inhibits apoptosis in culture (H9c2 cells) and rat cardiomyocytes following injury caused by hypoxia-reoxygenation or ischemia-reoxygenation by increasing the BCL2/BAX ratio with the involvement of metalloreductase STEAP4 (177). These results in vitro and in vivo affirmed the hypothetical effects of metformin on MI/RI produced by cellular apoptosis inhibition. The molecular mechanisms of this anti-apoptotic function of metformin are still poorly understood, though it was earlier reported that they include activation of AMPK (178). AMPK is considered to be a key molecule for cardioprotection based on the modulation of several signaling pathways involved in glucose metabolism and energy homeostasis (179). AMPKs are proteins that promote cell protection in ischemic conditions as the AMP/ATP ratio indicates intercellular energetic status and is increased in ischemic tissues (191).

A considerable cardioprotective effect of berberine, an alkaloid from Berberis vulgaris, was revealed. This plant alkaloid was shown to reduce serum levels of heart injury markers such as CK-MB, LDG and cTnI with simultaneous upregulation of BCL2 expression and mitochondrial cytochrome c and downregulation of BAX (180).

The antidepressant escitalopram was shown to suppress cardiomyocyte apoptosis in patients with previous myocardial infarction compared with the controls, which was accompanied by a decrease in the BAX/BCL2 ratio (181).

Preliminary administration of Ilexsaponin A increased the levels of anti-apoptotic protein BCL2 and decreased pro-apoptotic protein BAX. These results confirmed that Ilexsaponin could suppress cardiomyocyte apoptosis in MI/RI, being a new potential cardioprotective agent which may be used for MI/RI treatment (182).

Preliminary introduction of Salvianolic acid (10, 20 or 30 mg/kg/day) effectively decreased myocardial synthesis of BAX and caspase-3 and increased BCL2 levels (50).

Inhaled administration of sevoflurane (halogenated anesthetic) inhibited BAX expression and enhanced Bcl2 expression in mice, which was mediated by suppression of miRNA-135b-5p, whereby drug prevented MI/RI by activating the JAK2/STAT3 signaling pathway (120).

Postconditioning with dexmedetomidine (high-selective agonist of α2-adrenoreceptors), which is widely used in anesthesiology and resuscitation, significantly increased the BCL2/BAX ratio in the rat myocardium with modeled diabetes mellitus and MI/RI via the PI3K/AKT/GSK-3β signaling pathway (183).

Rapamycin (an inhibitor of mTOR) is used for coating coronary stents containing special drugs to prevent in-stent restenosis after coronary angioplasty (184,185). Rapamycin induces unique cardioprotective signal transduction that includes phosphorylation of ERK, STAT3, eNOS and GSK-3β in association with increased BCL2/BAX ratio (184). JAK2/STAT3 signal transduction plays a critical role in cardioprotection induced by rapamycin, which is associated with an increase in BCL2/BAX (185). BCL2 expression was enhanced after STAT3 activation via ERK-dependent phosphorylation caused by rapamycin administration (186). Introduction of rapamycin before reperfusion is a promising method that might be capable of considerable restriction of the myocardial infarction area and inhibition of cardiomyocyte apoptosis after MI/RI via signaling pathways involving MAP kinases and PI3K/AKT (187).

Interestingly, a study showed an evident role of melatonin in cardioprotection through the enhancement of Bcl-xl and Bcl2 expression and inhibition of Bax gene expression by reduction of oxidative stress via the activation of the NAD-dependent protein deacetylase sirtuin-3 (SIRT3) signaling pathway (188-190). SIRT3 is localized in the mitochondria and regulates several mitochondrial metabolic pathways (192). Moreover, during MI/RI and type 1 diabetes, melatonin significantly inhibited apoptosis by suppression of caspase-3 and BAX production, cleavage of caspase-3 and an increase in BCL2 levels (192). These effects were also inhibited by a specific blocker of AMPK signal transduction (compound C) which determines that this signaling pathway plays a key role in the cardioprotective action of melatonin (192). Firstly, it was demonstrated that melatonin treatment is a potential strategy for prevention of MI/RI injury in cases of type 1 diabetes mellitus as it could enhance mitochondrial biogenesis and support normal functions of the mitochondria (192). Secondly, it was also shown that the AMPK/peroxisome proliferator-activated receptor γ coactivator 1α/SIRT3 signaling pathway played a key role in the cardioprotective action of melatonin (193). Melatonin also showed a strong protective effect via Notch1/Hes1 signal transduction in a receptor-dependent manner (193). The PTEN/AKT signaling pathway is a key consequent mediator of BCL2 expression enhancement in rats (in vivo) and cultivated H9C2 cardiomyocytes (in vitro) (194).

It is important to note that the mechanisms of metabolic cardioprotection of most preparations have been poorly investigated to date (177,180-184). The data of different randomized controlled trials often do not prove the effectiveness of the suggested methods (180-184). Clinical data is available for metformin, rapamycin, dexmedetomidine, berberine and sevoflurane, but sufficient evidence of effective cardioprotection is still missing (195). Considerable appending of new theoretical data is required that would include information concerning molecular and cellular mechanisms which this therapy would be targeted at.

5. Conclusions and perspectives

In recent years, focus on apoptosis has become a promising direction in the research of cardiovascular pathology since there is an opportunity to control this process and to protect the functional reserve of the myocardium. The studies mentioned in this review have demonstrated a number of effective methods for inhibiting cell apoptosis. Conclusions based on these results, unfortunately, did not lead to a final solution to the problem of prevention and treatment of MI/RI. There is still a lack of data to recommend or to introduce these results into clinical practice. This is predominantly explained by the fact that there is no consensus for common biological and pathogenetic significance of BCL2 associated processes: Is it cardioprotective or only a pathological mechanism leading to cardiomyocyte death and aggravation of myocardial degradation?

Proteins of the BCL2 family play main roles in intrinsic apoptosis, and regulation of their activity allows significantly reduced cell death. In addition to the influence of BCL2 protein on apoptosis development, it is worth paying attention to its non-apoptotic functions in MI/RI development. For example, BCL2 regulation features mitochondrial, nuclear and endoplasmic reticulum processes (including calcium homeostasis) and glucose and lipid metabolism (196-198).

The preservation of functionally active cardiomyocytes is a priority in the development of new algorithms for MI/RI treatment. A wider research of BCL2 integration into cellular processes in MI/RI is likely to result in building a more complete signaling network that can be targeted at for preventing reperfusion injury of cardiomyocytes.

Funding

The publication has been prepared with the support of the 'RUDN University Program 5-100'.

Availability of data and materials

Not applicable.

Authors' contributions

AYK and MLB conceptualized the study; AYK and MLB wrote the original draft; AYK, MLB, EVN, SPS and EA participated in writing and edited the review; EVN and APS prepared the figures; SMS edited the manuscript. All authors 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.

Acknowledgments

We thank Dr Bianca K. Verlinden (Department of Biochemistry, Centre for Sustainable Malaria Control, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa) for assisting in English language text editing.

References

1 

Kerr JF, Wyllie AH and Currie AR: Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 26:239–257. 1972. View Article : Google Scholar : PubMed/NCBI

2 

Rogalińska M: Alterations in cell nuclei during apoptosis. Cell Mol Biol Lett. 7:995–1018. 2002.

3 

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. View Article : Google Scholar : PubMed/NCBI

4 

Tait SW and Green DR: Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 11:621–632. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Kalkavan H and Green DR: MOMP, cell suicide as a BCL-2 family business. Cell Death Differ. 25:46–55. 2018. View Article : Google Scholar

6 

Czabotar PE, Lessene G, Strasser A and Adams JM: Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat Rev Mol Cell Biol. 15:49–63. 2014. View Article : Google Scholar

7 

Galluzzi L, Kepp O and Kroemer G: Mitochondrial regulation of cell death: A phylogenetically conserved control. Microb Cell. 3:101–108. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Moldoveanu T, Follis AV, Kriwacki RW and Green DR: Many players in BCL-2 family affairs. Trends Biochem Sci. 39:101–111. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Shamas-Din A, Kale J, Leber B and Andrews DW: Mechanisms of action of Bcl-2 family proteins. Cold Spring Harb Perspect Biol. 5:a0087142013. View Article : Google Scholar : PubMed/NCBI

10 

Hardwick JM, Chen Y and Jonas EA: Multipolar functions of BCL-2 proteins link energetics to apoptosis. Trends Cell Biol. 22:318–328. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Pihán P, Carreras-Sureda A and Hetz C: BCL-2 family: Integrating stress responses at the ER to control cell demise. Cell Death Differ. 24:1478–1487. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Zhang J and Ney PA: Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ. 16:939–946. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Zamorano S, Rojas-Rivera D, Lisbona F, Parra V, Court FA, Villegas R, Cheng EH, Korsmeyer SJ, Lavandero S and Hetz C: A BAX/BAK and cyclophilin D-independent intrinsic apoptosis pathway. PLoS One. 7:e377822012. View Article : Google Scholar : PubMed/NCBI

14 

Kilbride SM and Prehn JH: Central roles of apoptotic proteins in mitochondrial function. Oncogene. 32:2703–2711. 2013. View Article : Google Scholar

15 

Parsons M and Green D: Mitochondria in cell death. Essays Biochem. 47:99–114. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Korsmeyer SJ, Shutter JR, Veis DJ, Merry DE and Oltvai ZN: BCL2/Bax: A rheostat that regulates an anti-oxidant pathway and cell death. Semin Cancer Biol. 4:327–332. 1993.PubMed/NCBI

17 

Abbate A, Bussani R, Amin MS, Vetrovec GW and Baldi A: Acute myocardial infarction and heart failure: Role of apoptosis. Int J Biochem Cell Biol. 38:1834–1840. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Ahmad F, Lal H, Zhou J, Vagnozzi RJ, Yu JE, Shang X, Woodgett JR, Gao E and Force T: Cardiomyocyte-specific deletion of Gsk3α mitigates post-myocardial infarction remodeling, contractile dysfunction, and heart failure. J Am Coll Cardiol. 64:696–706. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Chinda K, Sanit J, Chattipakorn S and Chattipakorn N: Dipeptidyl peptidase-4 inhibitor reduces infarct size and preserves cardiac function via mitochondrial protection in ischaemia-reperfusion rat heart. Diab Vasc Dis Res. 11:75–83. 2014. View Article : Google Scholar

20 

Gao CK, Liu H, Cui CJ, Liang ZG, Yao H and Tian Y: Roles of MicroRNA-195 in cardiomyocyte apoptosis induced by myocardial ischemia-reperfusion injury. J Genet. 95:99–108. 2016. View Article : Google Scholar : PubMed/NCBI

21 

World Health Organization (WHO): World Health Statistics 2019: Monitoring health for the SDGs. WHO; Geneva: 2019

22 

Rajaleid K, Janszky I and Hallqvist J: Small birth size, adult over-weight, and risk of acute myocardial infraction. Epidemiology. 22:138–147. 2011. View Article : Google Scholar

23 

Minamino T: Cardioprotection from ischemia/reperfusion injury: Basic and translational research. Circ J. 76:1074–1082. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Li Z, Lu J, Luo Y, Li S and Chen M: High association between human circulating microRNA-497 and acute myocardial infarction. ScientificWorldJournal. 2014:9318452014.PubMed/NCBI

25 

Eltzschig HK and Eckle T: Ischemia and reperfusion-from mechanism to translation. Nat Med. 17:1391–1401. 2011. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Bak MI and Ingwall JS: Contribution of Na+/H+ exchange to Na+ overload in the ischemic hypertrophied hyperthyroid rat heart. Cardiovasc Res. 57:1004–1014. 2003. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Budhram-Mahadeo V, Fujita R, Bitsi S, Sicard P and Heads R: Co-expression of POU4F2/Brn-3b with p53 may be important for controlling expression of pro-apoptotic genes in cardiomyocytes following ischaemic/hypoxic insults. Cell Death Dis. 5:e15032014. View Article : Google Scholar : PubMed/NCBI

30 

Fröhlich GM, Meier P, White SK, Yellon DM and Hausenloy DJ: Myocardial reperfusion injury: Looking beyond primary PCI. Eur Heart J. 34:1714–1722. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Chen S, Hua F, Lu J, Jiang Y, Tang Y, Tao L, Zou B and Wu Q: Effect of dexmedetomidine on myocardial ischemia-reperfusion injury. Int J Clin Exp Med. 8:21166–21172. 2015.

32 

Moens AL, Claeys MJ, Timmermans JP and Vrints CJ: Myocardial ischemia/reperfusion-injury, a clinical view on a complex pathophysiological process. Int J Cardiol. 100:179–190. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Liu LF, Liang Z, Lv ZR, Liu XH, Bai J, Chen J, Chen C and Wang Y: MicroRNA-15a/b are up-regulated in response to myocardial ischemia/reperfusion injury. J Geriatr Cardiol. 9:28–32. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Brown DI and Griendling KK: Regulation of signal transduction by reactive oxygen species in the cardiovascular system. Circ Res. 116:531–549. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Kalogeris T, Baines CP, Krenz M and Korthuis RJ: Ischemia/reperfusion. Compr Physiol. 7:113–170. 2016. View Article : Google Scholar

36 

Neri M, Riezzo I, Pascale N, Pomara C and Turillazzi E: Ischemia/reperfusion injury following acute myocardial infarction: A critical issue for clinicians and forensic pathologists. Mediators Inflamm. 2017:70183932017. View Article : Google Scholar : PubMed/NCBI

37 

Wang X, Ha T, Zou J, Ren D, Liu L, Zhang X, Kalbfleisch J, Gao X, Williams D and Li C: MicroRNA-125b protects against myocardial ischaemia/reperfusion injury via targeting p53-medi-ated apoptotic signalling and TRAF6. Cardiovasc Res. 102:385–395. 2014. View Article : Google Scholar : PubMed/NCBI

38 

LeBaron TW, Kura B, Kalocayova B, Tribulova N and Slezak J: A new approach for the prevention and treatment of cardiovascular disorders. Molecular hydrogen significantly reduces the effects of oxidative stress. Molecules. 24:20762019. View Article : Google Scholar :

39 

Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B and Sadoshima J: Distinct roles of autophagy in the heart during ischemia and reperfusion: Roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ Res. 100:914–922. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Radomski MW, Palmer RM and Moncada S: Endogenous nitric oxide inhibits human platelet adhesion to vascular endothelium. Lancet. 2:1057–1058. 1987. View Article : Google Scholar : PubMed/NCBI

41 

Xia Y and Zweier JL: Substrate control of free radical generation from xanthine oxidase in the postischemic heart. J Biol Chem. 270:18797–18803. 1995. View Article : Google Scholar : PubMed/NCBI

42 

Zhang WP, Zong QF, Gao Q, Yu Y, Gu XY, Wang Y, Li ZH and Ge M: Effects of endomorphin-1 postconditioning on myocardial ischemia/reperfusion injury and myocardial cell apoptosis in a rat model. Mol Med Rep. 14:3992–3998. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Liu L, Zhang G, Liang Z, Liu X, Li T, Fan J, Bai J and Wang Y: MicroRNA-15b enhances hypoxia/reoxygenation-induced apoptosis of cardiomyocytes via a mitochondrial apoptotic pathway. Apoptosis. 19:19–29. 2014. View Article : Google Scholar

44 

Li CM, Shen SW, Wang T and Zhang XH: Myocardial ischemic post-conditioning attenuates ischemia reperfusion injury via PTEN/Akt signal pathway. Int J Clin Exp Med. 8:15801–15807. 2015.PubMed/NCBI

45 

Liou SF, Ke HJ, Hsu JH, Liang JC, Lin HH, Chen IJ and Yeh JL: San-Huang-Xie-Xin-Tang prevents rat hearts from ischemia/reperfusion-induced apoptosis through eNOS and MAPK pathways. Evid Based Complement Alternat Med. 2011:9150512011. View Article : Google Scholar : PubMed/NCBI

46 

Chen Q and Lesnefsky EJ: Blockade of electron transport during ischemia preserves bcl-2 and inhibits opening of the mitochondrial permeability transition pore. FEBS Lett. 585:921–926. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Chen Q, Xu H, Xu A, Ross T, Bowler E, Hu Y and Lesnefsky EJ: Inhibition of Bcl-2 sensitizes mitochondrial permeability transition pore (MPTP) opening in ischemia-damaged mitochondria. PLoS One. 10:e01188342015. View Article : Google Scholar : PubMed/NCBI

48 

Gustafsson AB and Gottlieb RA: Bcl-2 family members and apoptosis, taken to heart. Am J Physiol Cell Physiol. 292:C45–C51. 2007. View Article : Google Scholar

49 

Murphy E, Imahashi K and Steenbergen C: Bcl-2 regulation of mitochondrial energetics. Trends Cardiovasc Med. 15:283–290. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Qiao Z and Xu Y: Salvianolic acid b alleviating myocardium injury in ischemia reperfusion rats. Afr J Tradit Complement Altern Med. 13:157–161. 2016. View Article : Google Scholar

51 

Zhang HY, McPherson BC, Liu H, Baman TS, Rock P and Yao Z: H(2)O(2) opens mitochondrial K(ATP) channels and inhibits GABA receptors via protein kinase C-epsilon in cardiomyocytes. Am J Physiol Heart Circ Physiol. 282:H1395–H1403. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Meng G, Wang J, Xiao Y, Bai W, Xie L, Shan L, Moore PK and Ji Y: GYY4137 protects against myocardial ischemia and reperfusion injury by attenuating oxidative stress and apoptosis in rats. J Biomed Res. 29:203–213. 2015.PubMed/NCBI

53 

Song T, Wang P, Yu X, Wang A, Chai G, Fan Y and Zhang Z: Systems analysis of phosphorylation-regulated Bcl-2 interactions establishes a model to reconcile the controversy over the significance of Bcl-2 phosphorylation. Br J Pharmacol. 176:491–504. 2019. View Article : Google Scholar

54 

Markou T, Dowling AA, Kelly T and Lazou A: Regulation of Bcl-2 phosphorylation in response to oxidative stress in cardiac myocytes. Free Radic Res. 43:809–816. 2009. View Article : Google Scholar : PubMed/NCBI

55 

Syeda MZ, Fasae MB, Yue E, Ishimwe AP, Jiang Y, Du Z, Yang B and Bai Y: Anthocyanidin attenuates myocardial ischemia induced injury via inhibition of ROS-JNK-BCL2 pathway: New mechanism of anthocyanidin action. Phytother Res. 33:3129–3139. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Zhang Z, Deng X, Liu Y, Liu Y, Sun L and Chen F: PKM2, function and expression and regulation. Cell Biosci. 9:522019. View Article : Google Scholar : PubMed/NCBI

57 

Menon MB and Dhamija S: Beclin 1 Phosphorylation-at the center of autophagy regulation. Front Cell Dev Biol. 6:1372018. View Article : Google Scholar

58 

Wei Y, Sinha S and Levine B: Dual role of JNK1-mediated phosphorylation of Bcl-2 in autophagy and apoptosis regulation. Autophagy. 4:949–951. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Schulman D, Latchman DS and Yellon DM: Urocortin protects the heart from reperfusion injury via upregulation of p42/p44 MAPK signaling pathway. Am J Physiol Heart Circ Physiol. 283:H1481–H1488. 2002. View Article : Google Scholar : PubMed/NCBI

60 

Hausenloy DJ, Tsang A, Mocanu MM and Yellon DM: Ischemic preconditioning protects by activating prosurvival kinases at reperfusion. Am J Physiol Heart Circ Physiol. 288:H971–H976. 2005. View Article : Google Scholar

61 

Carter AN, Born HA, Levine AT, Dao AT, Zhao AJ, Lee WL and Anderson AE: Wortmannin attenuates seizure-induced hyperactive PI3K/Akt/mTOR signaling, impaired memory, and spine dysmorphology in rats. eNeuro. 4. pp. ENEURO.0354–16.2017. 2017, View Article : Google Scholar

62 

Very N, Vercoutter-Edouart AS, Lefebvre T, Hardivillé S and El Yazidi-Belkoura I: Cross-dysregulation of O-GlcNAcylation and PI3K/AKT/mTOR axis in human chronic diseases. Front Endocrinol (Lausanne). 9:6022018. View Article : Google Scholar

63 

Chi Y, Ma Q, Ding XQ, Qin X, Wang C and Zhang J: Research on protective mechanism of ibuprofen in myocardial ischemia-reperfusion injury in rats through the PI3K/Akt/mTOR signaling pathway. Eur Rev Med Pharmacol Sci. 23:4465–4473. 2019.PubMed/NCBI

64 

Liang K, Ye Y, Wang Y, Zhang J and Li C: Formononetin mediates neuroprotection against cerebral ischemia/reperfusion in rats via downregulation of the Bax/BCL2 ratio and upregulation PI3K/Akt signaling pathway. J Neurol Sci. 344:100–104. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Yu LN, Yu J, Zhang FJ, Yang MJ, Ding TT, Wang JK, He W, Fang T, Chen G and Yan M: Sevoflurane postconditioning reduces myocardial reperfusion injury in rat isolated hearts via activation of PI3K/Akt signaling and modulation of Bcl-2 family proteins. J Zhejiang Univ Sci B. 11:661–672. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Zhang J, Wang C, Yu S, Luo Z, Chen Y, Liu Q, Hua F, Xu G and Yu P: Sevoflurane postconditioning protects rat hearts against ischemia-reperfusion injury via the activation of PI3K/AKT/mTOR signaling. Sci Rep. 4:73172014. View Article : Google Scholar : PubMed/NCBI

67 

Keyes KT, Xu J, Long B, Zhang C, Hu Z and Ye Y: Pharmacological inhibition of PTEN limits myocardial infarct size and improves left ventricular function postinfarction. Am J Physiol Heart Circ Physiol. 298:H1198–H1208. 2010. View Article : Google Scholar : PubMed/NCBI

68 

Zu L, Zheng X, Wang B, Parajuli N, Steenbergen C, Becker LC and Cai ZP: Ischemic preconditioning attenuates mitochondrial localization of PTEN induced by ischemia-reperfusion. Am J Physiol Heart Circ Physiol. 300:H2177–H2186. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Zhu YB, Ding N, Yi HL and Li ZQ: The expression of overexpressed PTEN enhanced IR-induced apoptosis of myocardial cells. Eur Rev Med Pharmacol Sci. 23:4406–4413. 2019.PubMed/NCBI

70 

Robinson MJ and Cobb MH: Mitogen-activated protein kinase pathways. Curr Opin Cell Biol. 9:180–186. 1997. View Article : Google Scholar : PubMed/NCBI

71 

Hernández-Reséndiz S, Roldán FJ, Correa F, Martínez-Abundis E, Osorio-Valencia G, Ruíz-de-Jesús O, Alexánderson-Rosas E, Vigueras RM, Franco M and Zazueta C: Postconditioning protects against reperfusion injury in hypertensive dilated cardiomyopathy by activating MEK/ERK1/2 signaling. J Card Fail. 19:135–146. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Holderfield M, Deuker MM, McCormick F and McMahon M: Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat Rev Cancer. 14:455–467. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Balmanno K and Cook SJ: Tumour cell survival signalling by the ERK1/2 pathway. Cell Death Differ. 16:368–377. 2009. View Article : Google Scholar

74 

Zhou QL, Teng F, Zhang YS, Sun Q, Cao YX and Meng GW: FPR1 gene silencing suppresses cardiomyocyte apoptosis and ventricular remodeling in rats with ischemia/reperfusion injury through the inhibition of MAPK signaling pathway. Exp Cell Res. 370:506–518. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Sun G, Ye N, Dai D, Chen Y, Li C and Sun Y: The protective role of the TOPK/PBK pathway in myocardial ischemia/reperfusion and H2O2-induced injury in H9C2 cardiomyocytes. Int J Mol Sci. 17:2672016. View Article : Google Scholar

76 

Sun MH, Chen XC, Han M, Yang YN, Gao XM, Ma X, Huang Y, Li XM, Gai MT, Liu F, et al: Cardioprotective effects of constitutively active MEK1 against H2O2-induced apoptosis and autophagy in cardiomyocytes via the ERK1/2 signaling pathway. Biochem Biophys Res Commun. 512:125–130. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Lee ML, Sulistyowati E, Hsu JH, Huang BY, Dai ZK, Wu BN, Chao YY and Yeh JL: KMUP-1 ameliorates ischemia-induced cardiomyocyte apoptosis through the NOcGMPMAPK signaling pathways. Molecules. 24:13762019. View Article : Google Scholar

78 

Razavi HM, Hamilton JA and Feng Q: Modulation of apoptosis by nitric oxide: Implications in myocardial ischemia and heart failure. Pharmacol Ther. 106:147–162. 2005. View Article : Google Scholar : PubMed/NCBI

79 

Burley DS, Ferdinandy P and Baxter GF: Cyclic GMP and protein kinase-G in myocardial ischaemia-reperfusion: Opportunities and obstacles for survival signaling. Br J Pharmacol. 152:855–869. 2007. View Article : Google Scholar : PubMed/NCBI

80 

Shvedova M, Anfinogenova Y, Atochina-Vasserman EN, Schepetkin IA and Atochin DN: c-Jun N-Terminal Kinases (JNKs) in myocardial and cerebral ischemia/reperfusion injury. Front Pharmacol. 9:7152018. View Article : Google Scholar : PubMed/NCBI

81 

Zhang W, Zhang Y, Ding K, Zhang H, Zhao Q, Liu Z and Xu Y: Involvement of JNK1/2-NF-κBp65 in the regulation of HMGB2 in myocardial ischemia/reperfusion-induced apoptosis in human AC16 cardiomyocytes. Biomed Pharmacother. 106:1063–1071. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Wang Z, Huang H, He W, Kong B, Hu H, Fan Y, Liao J, Wang L, Mei Y, Liu W, et al: Regulator of G-protein signaling 5 protects cardiomyocytes against apoptosis during in vitro cardiac ischemia-reperfusion in mice by inhibiting both JNK1/2 and P38 signaling pathways. Biochem Biophys Res Commun. 473:551–557. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Chen Q, Xu T, Li D, Pan D, Wu P, Luo Y, Ma Y and Liu Y: JNK/PI3K/Akt signaling pathway is involved in myocardial ischemia/reperfusion injury in diabetic rats: Effects of salvianolic acid A intervention. Am J Transl Res. 8:2534–2548. 2016.PubMed/NCBI

84 

Frias MA and Montessuit C: JAK-STAT signaling and myocardial glucose metabolism. JAKSTAT. 2:e264582013.

85 

Harhous Z, Booz GW, Ovize M, Bidaux G and Kurdi M: An update on the multifaceted roles of STAT3 in the heart. Front Cardiovasc Med. 6:1502019. View Article : Google Scholar : PubMed/NCBI

86 

Zhang WY, Zhang QL and Xu MJ: Effects of propofol on myocardial ischemia reperfusion injury through inhibiting the JAK/STAT pathway. Eur Rev Med Pharmacol Sci. 23:6339–6345. 2019.PubMed/NCBI

87 

Bolli R, Dawn B and Xuan YT: Emerging role of the JAK-STAT pathway as a mechanism of protection against ischemia/reperfusion injury. J Mol Cell Cardiol. 33:1893–1896. 2001. View Article : Google Scholar : PubMed/NCBI

88 

Liu Y, Che G, Di Z, Sun W, Tian J and Ren M: Calycosin-7-O- β-D-glucoside attenuates myocardial ischemia-reperfusion injury by activating JAK2/STAT3 signaling pathway via the regulation of IL-10 secretion in mice. Mol Cell Biochem. 463:175–187. 2020. View Article : Google Scholar

89 

Bolli R, Dawn B and Xuan YT: Role of the JAK-STAT pathway in protection against myocardial ischemia/reperfusion injury. Trends Cardiovasc Med. 13:72–79. 2003. View Article : Google Scholar : PubMed/NCBI

90 

Luan HF, Zhao ZB, Zhao QH, Zhu P, Xiu MY and Ji Y: Hydrogen sulfide postconditioning protects isolated rat hearts against ischemia and reperfusion injury mediated by the JAK2/STAT3 survival pathway. Braz J Med Biol Res. 45:898–905. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Boengler K, Buechert A, Heinen Y, Roeskes C, Hilfiker-Kleiner D, Heusch G and Schulz R: Cardioprotection by ischemic post-conditioning is lost in aged and STAT3-deficient mice. Circ Res. 102:131–135. 2008. View Article : Google Scholar

92 

Lecour S: Activation of the protective Survivor Activating Factor Enhancement (SAFE) pathway against reperfusion injury: Does it go beyond the RISK pathway? J Mol Cell Cardiol. 47:32–40. 2009. View Article : Google Scholar : PubMed/NCBI

93 

Myers MG Jr: Cell biology. Moonlighting in mitochondria. Science. 323:723–724. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Suleman N, Somers S, Smith R, Opie LH and Lecour SC: Dual activation of STAT-3 and Akt is required during the trigger phase of ischaemic preconditioning. Cardiovasc Res. 79:127–133. 2008. View Article : Google Scholar : PubMed/NCBI

95 

Boengler K, Hilfiker-Kleiner D, Drexler H, Heusch G and Schulz R: The myocardial JAK/STAT pathway: From protection to failure. Pharmacol Ther. 120:172–185. 2008. View Article : Google Scholar : PubMed/NCBI

96 

Bolli R, Stein AB, Guo Y, Wang OL, Rokosh G, Dawn B, Molkentin JD, Sanganalmath SK, Zhu Y and Xuan YT: A murine model of inducible, cardiac-specific deletion of STAT3: Its use to determine the role of STAT3 in the upregulation of cardioprotective proteins by ischemic preconditioning. J Mol Cell Cardiol. 50:589–597. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Hattori R, Maulik N, Otani H, Zhu L, Cordis G, Engelman RM, Siddiqui MA and Das DK: Role of STAT3 in ischemic preconditioning. J Mol Cell Cardiol. 33:1929–1936. 2001. View Article : Google Scholar : PubMed/NCBI

98 

Shen P, Chen J and Pan M: The protective effects of total paeony glycoside on ischemia/reperfusion injury in H9C2 cells via inhibition of the PI3K/Akt signaling pathway. Mol Med Rep. 18:3332–3340. 2018.PubMed/NCBI

99 

Koeppen M, Lee JW, Seo SW, Brodsky KS, Kreth S, Yang IV, Buttrick PM, Eckle T and Eltzschig HK: Hypoxia-inducible factor 2-alpha-dependent induction of amphiregulin dampens myocardial ischemia-reperfusion injury. Nat Commun. 9:8162018. View Article : Google Scholar : PubMed/NCBI

100 

Li T, Yu J, Chen R, Wu J, Fei J, Bo Q, Xue L and Li D: Mycophenolate mofetil attenuates myocardial ischemia-reperfusion injury via regulation of the TLR4/NF-κB signaling pathway. Pharmazie. 69:850–855. 2014.

101 

Lin J, Wang H, Li J, Wang Q, Zhang S, Feng N, Fan R and Pei J: κ-Opioid receptor stimulation modulates TLR4/NF-κB signaling in the rat heart subjected to ischemia-reperfusion. Cytokine. 61:842–848. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Li J, Xie C, Zhuang J, Li H, Yao Y, Shao C and Wang H: Resveratrol attenuates inflammation in the rat heart subjected to ischemia-reperfusion: Role of the TLR4/NF-κB signaling pathway. Mol Med Rep. 11:1120–1126. 2015.

103 

Gao Y, Song G, Cao YJ, Yan KP, Li B, Zhu XF, Wang YP, Xing ZY, Cui L, Wang XX and Zhu MJ: The Guizhi Gancao Decoction attenuates myocardial ischemia-reperfusion injury by suppressing inflammation and cardiomyocyte apoptosis. Evid Based Complement Alternat Med. 2019:19474652019. View Article : Google Scholar : PubMed/NCBI

104 

Zhang N, Ye F, Zhu W, Hu D, Xiao C, Nan J, Su S, Wang Y, Liu M, Gao K, et al: Cardiac ankyrin repeat protein attenuates cardiomyocyte apoptosis by upregulation of Bcl-2 expression. Biochim Biophys Acta. 1863:3040–3049. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Ibrahim A: Inhibition of α-SMA, Bax and increase of BCL2 expression in myocardiocytes as response to chitosan administration to hypercholesterolemic rats. World J Pharm Pharmac Sci. 5:164–176. 2016.

106 

Guo J, Li HZ, Wang LC, Zhang WH, Li GW, Xing WJ, Wang R and Xu CQ: Increased expression of calcium-sensing receptors in atherosclerosis confers hypersensitivity to acute myocardial infarction in rats. Mol Cell Biochem. 366:345–354. 2012. View Article : Google Scholar : PubMed/NCBI

107 

Kuo WW, Hsu TC, Chain MH, Lai CH, Wang WH, Tsai FJ, Tsai CH, Wu CH, Huang CY and Tzang BS: Attenuated cardiac mitochondrial-dependent apoptotic effects by li-fu formula in hamsters fed with a hypercholesterol diet. Evid Based Complement Alternat Med. 2011:5303452011. View Article : Google Scholar :

108 

Latif N, Khan MA, Birks E, O'Farrell A, Westbrook J, Dunn MJ and Yacoub MH: Upregulation of the Bcl-2 family of proteins in end stage heart failure. J Am Coll Cardiol. 35:1769–1777. 2000. View Article : Google Scholar : PubMed/NCBI

109 

Wang TD, Chen WJ, Su SS, Lo SC, Lin WW and Lee YT: Increased cardiomyocyte apoptosis following ischemia and reperfusion in diet-induced hypercholesterolemia: Relation to Bcl-2 and Bax proteins and caspase-3 activity. Lipids. 37:385–394. 2002. View Article : Google Scholar : PubMed/NCBI

110 

Ye Y, Perez-Polo JR, Qian J and Birnbaum Y: The role of microRNA in modulating myocardial ischemia-reperfusion injury. Physiol Genomics. 43:534–542. 2011. View Article : Google Scholar

111 

Tang R, Long T, Lui KO, Chen Y and Huang ZP: A roadmap for fixing the heart: RNA regulatory networks in cardiac disease. Mol Ther Nucleic Acids. 20:673–686. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Kukreja RC, Yin C and Salloum FN: MicroRNAs: New players in cardiac injury and protection. Mol Pharmacol. 80:558–564. 2011. View Article : Google Scholar : PubMed/NCBI

113 

Çakmak HA and Demir M: MicroRNA and cardiovascular diseases. Balkan Med J. 37:60–71. 2020.PubMed/NCBI

114 

Peterson SM, Thompson JA, Ufkin ML, Sathyanarayana P, Liaw L and Congdon CB: Common features of microRNA target prediction tools. Front Genet. 5:232014. View Article : Google Scholar : PubMed/NCBI

115 

Cao L, Wang J and Wang PQ: MiR-326 is a diagnostic biomarker and regulates cell survival and apoptosis by targeting Bcl-2 in osteosarcoma. Biomed Pharmacother. 84:828–835. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Cheng Y, Tan N, Yang J, Liu X, Cao X, He P, Dong X, Qin S and Zhang C: A translational study of circulating cell-free microRNA-1 in acute myocardial infarction. Clin Sci (Lond). 119:87–95. 2010. View Article : Google Scholar

117 

Dehaini H, Awada H, El-Yazbi A, Zouein FA, Issa K, Eid AA, Ibrahim M, Badran A, Baydoun E, Pintus G and Eid AH: MicroRNAs as potential pharmaco-targets in ischemia-reperfusion injury compounded by diabetes. Cells. 8:1522019. View Article : Google Scholar :

118 

Yan H, Li Y, Wang C, Zhang Y, Liu C, Zhou K and Hua Y: Contrary microRNA expression pattern between fetal and adult cardiac remodeling: Therapeutic value for heart failure. Cardiovasc Toxicol. 17:267–276. 2017. View Article : Google Scholar

119 

Tang Y, Zheng J, Sun Y, Wu Z, Liu Z and Huang G: MicroRNA-1 regulates cardiomyocyte apoptosis by targeting Bcl-2. Int Heart J. 50:377–387. 2009. View Article : Google Scholar : PubMed/NCBI

120 

Xie XJ, Fan DM, Xi K, Chen YW, Qi PW, Li QH, Fang L and Ma LG: Suppression of microRNA-135b-5p protects against myocardial ischemia/reperfusion injury by activating JAK2/STAT3 signaling pathway in mice during sevoflurane anesthesia. Biosci Rep. 37:BSR201701862017. View Article : Google Scholar : PubMed/NCBI

121 

Hullinger TG, Montgomery RL, Seto AG, Dickinson BA, Semus HM, Lynch JM, Dalby CM, Robinson K, Stack C, Latimer PA, et al: Inhibition of miR-15 protects against cardiac ischemic injury. Circ Res. 110:71–81. 2012. View Article : Google Scholar :

122 

Liu X, Nie J and Li C: Targeted regulation of Bcl 2 by miR-16 for cardiomyocyte apoptosis after cardiac infarction. Int J Clin Exp Pathol. 10:4626–4632. 2017.

123 

Yang W, Yang Y, Xia L, Yang Y, Wang F, Song M, Chen X, Liu J, Song Y, Zhao Y and Yang C: MiR-221 promotes Capan-2 pancreatic ductal adenocarcinoma cells proliferation by targeting PTEN-Akt. Cell Physiol Biochem. 38:2366–2374. 2016. View Article : Google Scholar : PubMed/NCBI

124 

Ye Z, Hao R, Cai Y, Wang X and Huang G: Knockdown of miR-221 promotes the cisplatin-inducing apoptosis by targeting the BIM-Bax/Bak axis in breast cancer. Tumour Biol. 37:4509–4515. 2016. View Article : Google Scholar

125 

Kong QR, Ji DM, Li FR, Sun HY and Wang QX: MicroRNA-221 promotes myocardial apoptosis caused by myocardial ischemia-reperfusion by down-regulating PTEN. Eur Rev Med Pharmacol Sci. 23:3967–3975. 2019.PubMed/NCBI

126 

Li X, Zeng Z, Li Q, Xu Q, Xie J, Hao H, Luo G, Liao W, Bin J, Huang X and Liao Y: Inhibition of microRNA-497 ameliorates anoxia/reoxygenation injury in cardiomyocytes by suppressing cell apoptosis and enhancing autophagy. Oncotarget. 6:18829–18844. 2015. View Article : Google Scholar : PubMed/NCBI

127 

Yang Q, Yang K and Li A: microRNA-21 protects against ischemia-reperfusion and hypoxia-reperfusion-induced cardiocyte apoptosis via the phosphatase and tensin homolog/Akt-dependent mechanism. Mol Med Rep. 9:2213–2220. 2014. View Article : Google Scholar : PubMed/NCBI

128 

Fan ZX and Yang J: The role of microRNAs in regulating myocardial ischemia reperfusion injury. Saudi Med J. 36:787–793. 2015. View Article : Google Scholar : PubMed/NCBI

129 

Yang J, Chen L, Yang J, Ding J, Li S, Wu H, Zhang J, Fan Z, Dong W and Li X: MicroRNA-22 targeting CBP protects against myocardial ischemia-reperfusion injury through anti-apoptosis in rats. Mol Biol Rep. 41:555–561. 2014. View Article : Google Scholar

130 

Yang J, Fan Z, Yang J, Ding J, Yang C and Chen L: microRNA-22 attenuates myocardial ischemia-reperfusion injury via an anti-inflammatory mechanism in rats. Exp Ther Med. 12:3249–3255. 2016. View Article : Google Scholar : PubMed/NCBI

131 

Liu S, Yang Y, Song YQ, Geng J and Chen QL: Protective effects of N(2)-L-alanyl-L-glutamine mediated by the JAK2/STAT3 signaling pathway on myocardial ischemia reperfusion. Mol Med Rep. 17:5102–5108. 2018.PubMed/NCBI

132 

Xu D, Li H, Zhao Y and Wang C: Downregulation of miR-34 a attenuates myocardial ischemia/reperfusion injury by inhibiting cardiomyocyte apoptosis. Int J Clin Exp Pathol. 10:3865–3875. 2017.

133 

Kikuchi K and Poss KD: Cardiac regenerative capacity and mechanisms. Annu Rev Cell Dev Biol. 28:719–741. 2012. View Article : Google Scholar : PubMed/NCBI

134 

Biala AK and Kirshenbaum LA: The interplay between cell death signaling pathways in the heart. Trends Cardiovasc Med. 24:325–331. 2014. View Article : Google Scholar : PubMed/NCBI

135 

McCully JD, Wakiyama H, Hsieh YJ, Jones M and Levitsky S: Differential contribution of necrosis and apoptosis in myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 286:H1923–H1935. 2004. View Article : Google Scholar : PubMed/NCBI

136 

Mocanu MM, Baxter GF and Yellon DM: Caspase inhibition and limitation of myocardial infarct size: Protection against lethal reperfusion injury. Br J Pharmacol. 130:197–200. 2000. View Article : Google Scholar : PubMed/NCBI

137 

Baxter G, Mocanu M, Brar B, Latchman D and Yellon D: Cardioprotective effects of transforming growth factor-beta1 during early reoxygenation or reperfusion are mediated by p42/p44 MAPK. J Cardiovasc Pharmacol. 38:930–939. 2001. View Article : Google Scholar : PubMed/NCBI

138 

Yellon DM and Baxter GF: Reperfusion injury revisited: Is there a role for growth factor signaling in limiting lethal reperfusion injury? Trends Cardiovasc Med. 9:245–249. 1999. View Article : Google Scholar

139 

Davidson SM, Ferdinandy P, Andreadou I, Bøtker HE, Heusch G, Ibáñez B, Ovize M, Schulz R, Yellon DM, Hausenloy DJ, et al: Multitarget strategies to reduce myocardial ischemia/reperfusion injury: JACC review topic of the week. J Am Coll Cardiol. 73:89–99. 2019. View Article : Google Scholar : PubMed/NCBI

140 

Soares ROS, Losada DM, Jordani MC, Évora P and Castro-E-Silva O: Ischemia/reperfusion injury revisited: An overview of the latest pharmacological strategies. Int J Mol Sci. 20:50342019. View Article : Google Scholar :

141 

Euler G: Good and bad sides of TGFβ-signaling in myocardial infarction. Front Physiol. 6:662015. View Article : Google Scholar

142 

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

143 

Mykytenko J, Kerendi F, Reeves JG, Kin H, Zatta AJ, Jiang R, Guyton RA, Vinten-Johansen J and Zhao ZQ: Long-term inhibition of myocardial infarction by postconditioning during reperfusion. Basic Res Cardiol. 102:90–100. 2007. View Article : Google Scholar

144 

Argaud L, Gateau-Roesch O, Raisky O, Loufouat J, Robert D and Ovize M: Postconditioning inhibits mitochondrial permeability transition. Circulation. 111:194–197. 2005. View Article : Google Scholar : PubMed/NCBI

145 

Pagliaro P, Femminò S, Popara J and Penna C: Mitochondria in cardiac postconditioning. Front Physiol. 9:2872018. View Article : Google Scholar : PubMed/NCBI

146 

Heusch G: Critical issues for the translation of cardioprotection. Circ Res. 120:1477–1486. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Heusch G: Cardioprotection research must leave its comfort zone. Eur Heart J. 39:3393–3395. 2018. View Article : Google Scholar : PubMed/NCBI

148 

Hausenloy DJ, Botker HE, Engstrom T, Erlinge D, Heusch G, Ibanez B, Kloner RA, Ovize M, Yellon DM and Garcia-Dorado D: Targeting reperfusion injury in patients with ST-segment elevation myocardial infarction: Trials and tribulations. Eur Heart J. 38:935–941. 2017.

149 

Ferdinandy P, Hausenloy DJ, Heusch G, Baxter GF and Schulz R: Interaction of risk factors, comorbidities, and comedications with ischemia/reperfusion injury and cardioprotection by preconditioning, postconditioning, and remote conditioning. Pharmacol Rev. 66:1142–1174. 2014. View Article : Google Scholar : PubMed/NCBI

150 

Hausenloy DJ, Garcia-Dorado D, Bøtker HE, Davidson SM, Downey J, Engel FB, Jennings R, Lecour S, Leor J, Madonna R, et al: Novel targets and future strategies for acute cardioprotection: Position paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res. 113:564–585. 2017. View Article : Google Scholar : PubMed/NCBI

151 

Inserte J, Hernando V, Vilardosa Ú, Abad E, Poncelas-Nozal M and Garcia/Dorado D: Activation of cGMP/protein kinase G pathway in postconditioned myocardium depends on reduced oxidative Stress and preserved endothelial nitric oxide synthase coupling. J Am Heart Assoc. 2:e0059752013. View Article : Google Scholar : PubMed/NCBI

152 

Kleinbongard P, Skyschally A and Heusch G: Cardioprotection by remote ischemic conditioning and its signal transduction. Pflugers Arch. 469:159–181. 2017. View Article : Google Scholar

153 

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

154 

Murry CE, Jennings RB and Reimer KA: Preconditioning with ischemia: A delay of lethal cell injury in ischemic myocardium. Circulation. 74:1124–1136. 1986. View Article : Google Scholar : PubMed/NCBI

155 

Wever KE, Hooijmans CR, Riksen NP, Sterenborg TB, Sena ES, Ritskes-Hoitinga M and Warlé MC: Determinants of the efficacy of cardiac ischemic preconditioning: A systematic review and meta-analysis of animal studies. PLoS One. 10:e01420212015. View Article : Google Scholar : PubMed/NCBI

156 

Wever KE, Menting TP, Rovers M, van der Vliet JA, Rongen GA, Masereeuw R, Ritskes-Hoitinga M, Hooijmans CR and Warlé M: Ischemic preconditioning in the animal kidney, a systematic review and meta-analysis. PLoS One. 7:e322962012. View Article : Google Scholar : PubMed/NCBI

157 

Yellon DM, Alkhulaifi AM and Pugsley WB: Preconditioning the human myocardium. Lancet. 342:276–277. 1993. View Article : Google Scholar : PubMed/NCBI

158 

Rossello X and Yellon DM: The RISK pathway and beyond. Basic Res Cardiol. 113:22017. View Article : Google Scholar : PubMed/NCBI

159 

Hausenloy DJ, Barrabes JA, Bøtker HE, Davidson SM, Di Lisa F, Downey J, Engstrom T, Ferdinandy P, Carbrera-Fuentes HA, Heusch G, et al: Ischaemic conditioning and targeting reperfusion injury: A 30 year voyage of discovery. Basic Res Cardiol. 111:702016. View Article : Google Scholar

160 

Sun XJ and Mao JR: Role of Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway in cardioprotection of exercise preconditioning. Eur Rev Med Pharmacol Sci. 22:4975–4986. 2018.PubMed/NCBI

161 

Duan W, Yang Y, Yan J, Yu S, Liu J, Zhou J, Zhang J, Jin Z and Yi D: The effects of curcumin post-treatment against myocardial ischemia and reperfusion by activation of the JAK2/STAT3 signaling pathway. Basic Res Cardiol. 107:2632012. View Article : Google Scholar : PubMed/NCBI

162 

Goodman MD, Koch SE, Afzal MR and Butler KL: STAT subtype specificity and ischemic preconditioning in mice: Is STAT-3 enough? Am J Physiol Heart Circ Physiol. 300:H522–H526. 2011. View Article : Google Scholar :

163 

Dawn B, Xuan YT, Guo Y, Rezazadeh A, Stein AB, Hunt G, Wu WJ, Tan W and Bolli R: IL-6 plays an obligatory role in late preconditioning via JAK-STAT signaling and upregulation of iNOS and COX-2. Cardiovasc Res. 64:61–71. 2004. View Article : Google Scholar : PubMed/NCBI

164 

Xuan YT, Guo Y, Zhu Y, Wang OL, Rokosh G, Messing RO and Bolli R: Role of the protein kinase C-epsilon-Raf-1-MEK-1/2-p44/42 MAPK signaling cascade in the activation of signal transducers and activators of transcription 1 and 3 and induction of cyclooxygenase-2 after ischemic preconditioning. Circulation. 112:1971–1978. 2005. View Article : Google Scholar : PubMed/NCBI

165 

Chen TI, Shen YJ, Wang IC and Yang KT: Short-term exercise provides left ventricular myocardial protection against intermit-tent hypoxia-induced apoptosis in rats. Eur J Appl Physiol. 111:1939–1950. 2011. View Article : Google Scholar : PubMed/NCBI

166 

Zhao ZQ, Corvera JS, Halkos ME, Kerendi F, Wang NP, Guyton RA and Vinten-Johansen J: Inhibition of myocardial injury by ischemic postconditioning during reperfusion: Comparison with ischemic preconditioning. Am J Physiol Heart Circ Physiol. 285:H579–H588. 2003. View Article : Google Scholar : PubMed/NCBI

167 

Thibault H, Piot C, Staat P, Bontemps L, Sportouch C, Rioufol G, Cung TT, Bonnefoy E, Angoulvant D, Aupetit JF, et al: Long-term benefit of postconditioning. Circulation. 117:1037–1044. 2008. View Article : Google Scholar : PubMed/NCBI

168 

Zhao CM, Yang XJ, Yang JH, Cheng XJ, Zhao X, Zhou BY, Xu SD and Wang HF: Effect of ischaemic postconditioning on recovery of left ventricular contractile function after acute myocardial infarction. J Int Med Res. 40:1082–1088. 2012. View Article : Google Scholar : PubMed/NCBI

169 

Tian Y, Zhang W, Xia D, Modi P, Liang D and Wei M: Postconditioning inhibits myocardial apoptosis during prolonged reperfusion via a AK2-STAT3-BCL2 pathway. J Biomed Sci. 18:532011. View Article : Google Scholar

170 

Shyu WC, Lin SZ, Chiang MF, Chen DC, Su CY, Wang HJ, Liu RS, Tsai CH and Li H: Secretoneurin promotes neuroprotection and neuronal plasticity via the Jak2/Stat3 pathway in murine models of stroke. J Clin Invest. 118:133–148. 2008. View Article : Google Scholar

171 

Siddiquee K, Zhang S, Guida WC, Blaskovich MA, Greedy B, Lawrence HR, Yip ML, Jove R, McLaughlin MM, Lawrence NJ, et al: Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc Natl Acad Sci USA. 104:7391–7396. 2007. View Article : Google Scholar : PubMed/NCBI

172 

Hausenloy DJ, Tsang A and Yellon DM: The reperfusion injury salvage kinase pathway: A common target for both ischemic preconditioning and postconditioning. Trends Cardiovasc Med. 15:69–75. 2005. View Article : Google Scholar : PubMed/NCBI

173 

Jin YC, Lee YS, Kim YM, Seo HG, Lee JH, Kim HJ, Yun-Choi HS and Chang KC: (S)-1-(alpha-naphthylmethyl)-6,7-di hydroxy-1,2,3,4-tetrahydroisoquinoline (CKD712) reduces rat myocardial apoptosis against ischemia and reperfusion injury by activation of phosphatidylinositol 3-kinase/Akt signaling and anti-inflammatory action in vivo. J Pharmacol Exp Ther. 330:440–448. 2009. View Article : Google Scholar : PubMed/NCBI

174 

Takahama H, Minamino T, Hirata A, Ogai A, Asanuma H, Fujita M, Wa keno M, Tsukamoto O, Okada K, Komamura K, et al: Granulocyte colony-stimulating factor mediates cardioprotection against ischemia/reperfusion injury via phosphatidylinositol-3-kinase/Akt pathway in canine hearts. Cardiovasc Drugs Ther. 20:159–165. 2006. View Article : Google Scholar : PubMed/NCBI

175 

Goodman MD, Koch SE, Fuller-Bicer GA and Butler KL: Regulating RISK: A role for JAK-STAT signaling in postconditioning? Am J Physiol Heart Circ Physiol. 295:H1649–H1656. 2008. View Article : Google Scholar : PubMed/NCBI

176 

Wu N, Zhang X, Jia P and Jia D: Hypercholesterolemia aggravates myocardial ischemia reperfusion injury via activating endoplasmic reticulum stress-mediated apoptosis. Exp Mol Pathol. 99:449–454. 2015. View Article : Google Scholar : PubMed/NCBI

177 

Luo T, Zeng X, Yang W and Zhang Y: Treatment with metformin prevents myocardial ischemia-reperfusion injury via STEAP4 signaling pathway. Anatol J Cardiol. 21:261–271. 2019.PubMed/NCBI

178 

Hu M, Ye P, Liao H, Chen M and Yang F: Metformin protects H9C2 cardiomyocytes from high-glucose and hypoxia/reoxygenation injury via inhibition of reactive oxygen species generation and inflammatory responses: Role of AMPK and JNK. J Diabetes Res. 2016:29619542016. View Article : Google Scholar : PubMed/NCBI

179 

Kahn BB, Alquier T, Carling D and Hardie DG: AMP-activated protein kinase: Ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 1:15–25. 2005. View Article : Google Scholar : PubMed/NCBI

180 

Wang Y, Liu J, Ma A and Chen Y: Cardioprotective effect of berberine against myocardial ischemia/reperfusion injury via attenuating mitochondrial dysfunction and apoptosis. Int J Clin Exp Med. 8:14513–14519. 2015.PubMed/NCBI

181 

Wang Y, Zhang H, Chai F, Liu X and Berk M: The effects of escitalopram on myocardial apoptosis and the expression of Bax and BCL2 during myocardial ischemia/reperfusion in a model of rats with depression. BMC Psychiatry. 14:3492014. View Article : Google Scholar

182 

Zhang SW, Liu Y, Wang F, Qiang J, Liu P, Zhang J and Xu JW: Ilexsaponin A attenuates ischemia-reperfusion-induced myocardial injury through anti-apoptotic pathway. PLoS One. 12:e01709842017. View Article : Google Scholar : PubMed/NCBI

183 

Cheng X, Hu J, Wang Y, Ye H, Li X, Gao Q and Li Z: Effects of dexmedetomidine postconditioning on myocardial ischemia/reperfusion injury in diabetic rats: Role of the PI3K/Akt-dependent signaling pathway. J Diabetes Res. 2018:30719592018. View Article : Google Scholar : PubMed/NCBI

184 

Morris RE: Prevention and treatment of allograft rejection in vivo by rapamycin: Molecular and cellular mechanisms of action. Ann N Y Acad Sci. 685:68–72. 1993. View Article : Google Scholar : PubMed/NCBI

185 

Menown IBA, Mamas MA, Cotton JM, Hildick-Smith D, Eberli FR, Leibundgut G, Tresukosol D, Macaya C, Copt S, Sadozai Slama S and Stoll HP: First clinical evidence characterizing safety and efficacy of the new CoCr Biolimus-A9 eluting stent: The Biomatrix Alpha™ registry. Int J Cardiol Heart Vasc. 26:1004722020.

186 

Das A, Salloum FN, Durrant D, Ockaili R and Kukreja RC: Rapamycin protects against myocardial ischemia-reperfusion injury through JAK2-STAT3 signaling pathway. J Mol Cell Cardiol. 53:858–869. 2012. View Article : Google Scholar : PubMed/NCBI

187 

Filippone SM, Samidurai A, Roh SK, Cain CK, He J, Salloum FN, Kukreja RC and Das A: Reperfusion therapy with rapamycin attenuates myocardial infarction through activation of AKT and ERK. Oxid Med Cell Longev. 2017:46197202017. View Article : Google Scholar : PubMed/NCBI

188 

Zhai M, Li B, Duan W, Jing L, Zhang B, Zhang M, Yu L, Liu Z, Yu B, Ren K, et al: Melatonin ameliorates myocardial ischemia reperfusion injury through SIRT3-dependent regulation of oxidative stress and apoptosis. J Pineal Res. 63:2017. View Article : Google Scholar

189 

Hsu CP, Zhai P, Yamamoto T, Maejima Y, Matsushima S, Hariharan N, Shao D, Takagi H, Oka S and Sadoshima J: Silent information regulator 1 protects the heart from ischemia/reperfusion. Circulation. 122:2170–2182. 2010. View Article : Google Scholar : PubMed/NCBI

190 

Yu L, Sun Y, Cheng L, Jin Z, Yang Y, Zhai M, Pei H, Wang X, Zhang H, Meng Q, et al: Melatonin receptor-mediated protection against myocardial ischemia/reperfusion injury: Role of SIRT1. J Pineal Res. 57:228–238. 2014. View Article : Google Scholar : PubMed/NCBI

191 

Hardi DG: Keeping the home fires burning: AMP-activated protein kinase. J R Soc Interface. 15:201707742018. View Article : Google Scholar

192 

Zhang M, Zhao Z, Shen M, Zhang Y, Duan J, Guo Y, Zhang D, Hu J, Lin J, Man W, et al: Polydatin protects cardiomyocytes against myocardial infarction injury by activating Sirt3. Biochim Biophys Acta Mol Basis Dis. 1863:1962–1972. 2017. View Article : Google Scholar

193 

Yu L, Gong B, Duan W, Fan C, Zhang J, Li Z, Xue X, Xu Y, Meng D, Li B, et al: Melatonin ameliorates myocardial ischemia/reperfusion injury in type 1 diabetic rats by preserving mitochondrial function: Role of AMPK-PGC-1α-SIRT3 signaling. Sci Rep. 7:413372017. View Article : Google Scholar

194 

Yu L, Liang H, Lu Z, Zhao G, Zhai M, Yang Y, Yang J, Yi D, Chen W, Wang X, et al: Membrane receptor-dependent Notch1/Hes1 activation by melatonin protects against myocardial ischemia-reperfusion injury: In vivo and in vitro studies. J Pineal Res. 59:420–433. 2015. View Article : Google Scholar : PubMed/NCBI

195 

Heusch G: Myocardial ischaemia-reperfusion injury and cardio-protection in perspective. Nat Rev Cardiol. Jul 3–2020.Epub ahead of print. View Article : Google Scholar

196 

Gross A and Katz SG: Non-apoptotic functions of BCL-2 family proteins. Cell Death Differ. 24:1348–1358. 2017. View Article : Google Scholar : PubMed/NCBI

197 

Chiu WT, Chang HA, Lin YH, Lin YS, Chang HT, Lin HH, Huang SC, Tang MJ and Shen MR: Bcl-2 regulates store-operated Ca2+ entry to modulate ER stress-induced apoptosis. Cell Death Discov. 4:372018. View Article : Google Scholar

198 

Bonneau B, Prudent J, Popgeorgiev N and Gillet G: Non-apoptotic roles of Bcl-2 family: The calcium connection. Biochim Biophys Acta. 1833:1755–176. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 47 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
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
APA
Korshunova, A.Y., Blagonravov, M.L., Neborak, E.V., Syatkin, S.P., Sklifasovskaya, A.P., Semyatov, S.M., & Agostinelli, E. (2021). BCL2‑regulated apoptotic process in myocardial ischemia‑reperfusion injury (Review). International Journal of Molecular Medicine, 47, 23-36. https://doi.org/10.3892/ijmm.2020.4781
MLA
Korshunova, A. Y., Blagonravov, M. L., Neborak, E. V., Syatkin, S. P., Sklifasovskaya, A. P., Semyatov, S. M., Agostinelli, E."BCL2‑regulated apoptotic process in myocardial ischemia‑reperfusion injury (Review)". International Journal of Molecular Medicine 47.1 (2021): 23-36.
Chicago
Korshunova, A. Y., Blagonravov, M. L., Neborak, E. V., Syatkin, S. P., Sklifasovskaya, A. P., Semyatov, S. M., Agostinelli, E."BCL2‑regulated apoptotic process in myocardial ischemia‑reperfusion injury (Review)". International Journal of Molecular Medicine 47, no. 1 (2021): 23-36. https://doi.org/10.3892/ijmm.2020.4781