Molecular mechanisms of autophagy in cardiac ischemia/reperfusion injury (Review)

  • Authors:
    • Xiao‑Long Lin
    • Wei‑Jin Xiao
    • Le‑Le Xiao
    • Mi‑Hua Liu
  • View Affiliations

  • Published online on: May 16, 2018     https://doi.org/10.3892/mmr.2018.9028
  • Pages: 675-683
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Autophagy is a maintenance process for recycling long-lived proteins and cytoplasmic organelles. The level of this process is enhanced during ischemia/reperfusion (I/R) injury. Autophagy can trigger survival signaling in myocardial ischemia, whereas defective autophagy during reperfusion is detrimental. Autophagy can be regulated through multiple signaling pathways in I/R, including Beclin‑1/class III phosphatidylinositol‑3 kinase (PI‑3K), adenosine monophosphate activated protein kinase/mammalian target of rapamycin (mTOR), and PI‑3K/protein kinase B/mTOR pathways, which consequently lead to different functions. Thus, autophagy has both protective and detrimental functions, which are determined by different signaling pathways and conditions. Targeting the activation of autophagy can be a promising new therapeutic strategy for treating cardiovascular disease.

Introduction

Myocardial ischemia was one of the main causes of sudden cardiac death in the past decades (1). Acute myocardial infarction is a leading cause of death worldwide (2). Strategies for reducing ischemia/reperfusion (I/R)-induced injury in cardiomyocytes are receiving considerable attention due to the failure of cardiomyocytes to regenerate (1). Coronary reperfusion is the most effective treatment for ischemic diseases. However, it may initially aggravate cellular damage during the ischemic period (3).

Cardiac ischemic preconditioning (IPC), which is achieved through repeated brief I/R periods, is one of the well-known protective strategies of the myocardium against I/R injury (4). Recently, autophagy has been linked to IPC-mediated cardioprotection (5). In addition, a number of pharmaceutical therapies targeting I/R injury have been developed to orchestrate multiple protein complexes and signaling pathways in autophagy (6,7). In this review, we aim to draw attention to the role of autophagy in cardioprotection.

Autophagy

Autophagy is a self-protective mechanism of living cells under various stress conditions (8). During autophagy, cellular cytoplasm constituents are delivered to lysosomes for degradation and recycling (9). Autophagy limits the production of reactive oxygen species and excessive protein aggregation to maintain intracellular or extracellular homeostasis (10). Autophagy has emerged as a potential drug target for numerous diseases including cancer, neurodegenerative diseases, and cardiovascular disease (11,12).

Autophagy plays multifaceted roles in heart and diseases (13). Under basal conditions, autophagy is a maintenance process for recycling long-lived proteins and cytoplasmic organelles in the heart (14). Furthermore, autophagy plays significant roles in starvation, aging, inflammation, and reverse cardiac remodeling by maintaining cellular homeostasis (15). Autophagy can be regarded as an end effector in hypoxic and ischemic conditions to eliminate superfluous, damaged, or aged cells or organelles (1618). However, this process will cause detrimental autophagic cell death when triggered by severe ischemia or in cardiovascular diseases (19).

Autophagy and cardioprotection

Myocardial I/R injury is a complex process that destroys proteins, DNA, and plasma membrane, thereby resulting in cell death and decreased cardiac output (20,21). Many studies have reported an increase in the number of autophagosomes in the heart during I/R in animal models (5,22). Autophagy induced by ischemia was subsequently enhanced by reperfusion in isolated rabbit hearts (23) and in mouse hearts (24). The activation of autophagy is reflected in the abundance of autophagy-related protein pathways, such as light chain 3 (LC3), Beclin-1, autophagy-related gene (ATG) 5–12 complex, and p62 (2527). Hu et al reported that approximately 20 min of aortic clamping with hyperkalemic cold blood cardioplegia to achieve total autophagy, which in accordance with previous evidence (28). The abundance of autophagic proteins will actually decrease with the progress of autophagy because of self-degradation (25,26). In particular, in biopsies from the right atrial appendage of patients undergoing valve surgery or coronary artery bypass grafting, the expression of autophagy-related proteins, including LC3-I, LC3-II, ATG5-12, Beclin-1, and p62, is reduced during reperfusion (26).

Until recently, the debate continues whether autophagy plays a protective or deleterious role in the I/R injury process. On the one hand, modest levels of autophagy triggered by mild to moderate hypoxia/ischemia are protective and seem to prevent the activation of apoptosis (23,29). On the other hand, high levels of autophagy induced by severe hypoxia or I/R may cause self-digestion and eventual cell death (30). Therefore, autophagic flux induced by ischemia during the early stage of I/R has been speculated to be beneficial; however, it is harmful during reperfusion at the later stage of I/R (15,19).

Autophagy may play an alternative role in I/R, which determines cell fate. The extent of autophagy in response to ischemia is considered based on the severity and duration of ischemic insults (31). Nutrient and oxygen deprivation in the heart threatens cellular survival during I/R, and increased autophagy may provide at least a temporary reprieve for a threatened myocardium by serving as a source of intracellular nutrients (32). Oxidative stress, calcium overload, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction maintain a high level of autophagy during reperfusion (33). However, high levels or long-term upregulation of autophagy can lead to excessive degradation of essential proteins and organelles (34). If intracellular energy sources become inadequate, then autophagic processes will be a particular form of cell death, called type II or autophagic cell death (35). In fact, aware that necrosis and apoptosis are not the only mechanisms of cell death is increasing (36). Autophagic cell death has been identified as a cell death phenotype via electron microscope observations; it has a morphological term characterized by abundant autophagic vacuoles in the cytoplasm (37,38).

Moreover, increased autophagy after I/R is not due to increased autophagosome formation, but instead, to impaired clearance of autophagosomes (39); this assumption is derived from the concept of autophagic flux (40). Furthermore, a rapid decline induced by reperfusion in LAMP2, which is a critical protein for autophagosome-lysosome fusion, can impair autophagosome processing and mitochondrial permeabilization, thereby increasing ROS generation and triggering cardiomyocyte death (41). In addition, when the engulfed targets or autophagosomes cannot fuse with lysosomes and digest their contents, a cell may eject the autophagosomes as a response, which induces an acute and significant inflammatory response (42).

Mechanism of autophagy in cardioprotection

Autophagy is a complex and dynamic multi-step process that depends on strict regulation and coordination through multiple signaling pathways (43). To date, several cellular signaling pathways are considered to trigger autophagy in I/R. In addition, autophagy has been shown to be regulated by several signaling pathways (44), including Beclin-1/class III phosphatidylinositol-3 kinase (PI-3K), AMPK/mammalian target of rapamycin (mTOR), and PI-3K/Akt/mTOR pathways.

Beclin-1/class III PI-3K pathway

Beclin-1, which is a phylogenetically conserved protein, the mammalian homologue of the yeast Atg6, and the interacting protein of the anti-apoptotic protein Bcl-2, is a key molecule involved in mediating autophagy (45,46). It plays a crucial role in engaging class III PI-3K to positively modulate autophagy in mammalian cells (47,48). Autophagy in mammalian cells is reported to be activated by the class III PI-3K complex, which contains Vps34 and Beclin-1 (29,49). Moreover, a coiled-coil domain (aa 140–268) is present in this 450 amino acid-long protein in Beclin-1; this domain can mediate binding to class III PI-3K Vps34 by interacting with an evolutionarily conserved domain (ECD; aa 244–337) (50). RNA interference of Beclin-1, which inhibits autophagy, will subsequently enhance cardiac cell survival (51).

Autophagy is involved in delayed cardioprotection induced by sevoflurane preconditioning (52). Sevoflurane preconditioning reduces the autophagy induced by H/R by decreasing the Beclin-1 expression (52). Accordingly, IPC protects the rat heart against MI/R injury by inhibiting Bcl-2 dissociation from Beclin-1 during the reperfusion phase in vivo, although IPC-induced autophagy reflects a compensatory pro-survival response to I/R injury (53). Bcl-2 is the prototype of a protein family, which contains at least one Bcl-2 homology (BH) region (54). Bcl-2 binding molecules have been recently shown to regulate autophagy activation (55). Transgenic mice with a cardiac-specific overexpression of Bcl-2 are protected from I/R injury (56,57). Autophagy is disrupted when Bcl-2 binds to Beclin-1 (58). In addition, when a mutant of Beclin-1 that lacks the Bcl-2 binding domain is overexpressed in cells, excessive autophagy and cell death are induced (47). Bcl-2 can also inhibit Beclin-1/Vsp34 PI-3K complex formation and the activity of Beclin-1-associated class III PI-3K (53). Furthermore, the class III PI-3K autophagic pathway is inhibited by combining the BH3 hydrophobic groove in Bcl-2 and the BH3-like amphipathic α-helix in Beclin-1 (59). However, the interaction with Bcl-2 (and Bcl-xL) in the ER, rather than in the mitochondria, inhibits the Beclin-1 activity in autophagy (60). The interaction between Bcl-2 and Beclin-1 maintains autophagy at levels (47). Blocking the interaction between the BH3 domains of Beclin-1 and Bcl-2 increases autophagic activity (53). Recent studies indicate that the increase in the interaction between Beclin-1 and Bcl-2 is caused by IPC (53). C-Jun N-terminal kinase (JNK), which is a member of an evolutionarily conserved subfamily of mitogen-activated protein kinases, is critical for the cellular responses of multiple environmental and cellular stimuli (61,62). I/R can trigger Bcl2-regulated autophagy by inducing a dominant increase in mitoJNK activation, which causes cell death (63). Xu et al reported that mitoJNK activation, and not JNK mitochondrial localization, induced autophagy, which further aggravates I/R injury (63). In addition, the mitoJNK phosphorylate Bcl2, which antagonizes Bcl2 anti-apoptotic and anti-autophagic activities, may contribute to the deleterious role of mitoJNK in I/R injury (64,65).

Heat shock protein (Hsp20) is the only member of the sHsps family that contains the consensus peptide motif RRAS for protein kinase A-/protein kinase G-dependent phosphorylation at Ser16 (66). Qian et al demonstrated that non-phosphorylated Hsp20S16A is detrimental in I/R injury because it suppresses autophagy and further increases cell death (36). Ischemic/hypoxic adaptation improves cardiac cell survival by suppressing the BAG-1 protein expression (67). BAG-1 can bind with both Bcl-2 and Hsc70 molecules (67). Autophagosomal membrane contains a significantly higher amount of Hsc70 proteins (68). BAG-1 has been shown exhibit numerous functions through its interaction with Hsc70 (69). The treatment of rats with wortmannin, an inhibitor of class III PI-3K, has been used to suppress autophagy in many studies (70,71), and attenuates both the LC3-II and BAG-1 protein expressions (67). Zheng et al (72) reported that the activated PI3K/Akt pathway contributes to the berbamine postconditioning-induced cardioprotection through modulating autophagy. The Beclin-1/class III PI-3K pathway-regulated autophagy and autophagy-mediated function in I/R injury are shown in Fig. 1.

AMPK/mTOR pathway

AMPK, which is activated in response to stress that exhaust cellular ATP supplies, such as ischemia and hypoxia, plays a crucial role as a master regulator of cellular energy homeostasis (73). AMPK is ubiquitously expressed in metabolically active tissues, such as cardiac muscles, and activated upon the depletion of energy stores by functioning as an intracellular fuel sensor (74). Ischemia has been proposed to stimulate autophagy via an AMPK-dependent mechanism (53), which is one of the most significant approaches in upregulating autophagy (75,76). During I/R injury, intracellular ATP stores are rapidly consumed and cannot be supplemented with decreasing glucose supply (77). AMPK signaling can positively regulate autophagy by activating Ulk1 via the phosphorylation of Ser 317 and Ser 777 or indirectly by inhibiting mTOR signaling (78,79). Moreover, AMPK functions as a master regulator of the autophagy pathway through inactivating mTOR (80).

High mTOR activity negatively regulates autophagy by inhibiting the activation of Ulk1, which is one of the mammalian autophagy-initiating kinases that is important for membrane nucleation via the phosphorylation of Ulk1 Ser 757 (1,81,82). The association between ATG1 and ATG13 is negatively regulated by mTOR, which inhibits autophagy (83). In addition, the activation of Ulk1 and its combination with other molecules, such as ATG13 and FIP200, initiate the nucleation of the autophagic membrane (1). Lekli et al proposed that tocotrienol could induce autophagy through the mTOR pathway, which would consequently lead to cell survival and cardioprotection (84). The overexpression of Bnip3, a hypoxia-inducible Bcl-2 homology 3 domain-containing protein (85) and the pro-apoptotic molecule present in the mitochondrial membrane; can upregulate autophagy and protect cardiac myocytes against I/R injury-related apoptosis (86). The high-mobility group box 1 protein (HMGB1)-mediated activation of mTOR inhibits hypoxia and reoxygenation injury in rat cardiomyocytes (87,88). Moreover, Bnip3 can inhibit the mTOR pathway and induce autophagy by directly binding to the Ras homolog that is enriched in the brain (Rheb), which is a Ras-related small guanosine triphosphatase (85,89).

The cardioprotection effect of resveratrol has been shown to induce autophagy by facilitating AMPK activation (90,91). In addition, AMPK expression is elevated with ACh during H/R (92). ACh activates cytoprotective autophagy through the AMPK-mTOR-dependent pathway that is activated by a muscarinic receptor (92). Xie et al found that the post-reperfusion AMPK activation induced by a slow-releasing organic H2S donor that could restore I/R impaired autophagic flux; is critical to H2S cardioprotection (93). Accordingly, studies have proven that autophagy activation through the AMPK/mTOR pathway plays a cardioprotection role (94). Recently, ultraviolet B-induced autophagy has been found to activate AMPK by inhibiting the phosphorylation of GSK3β (95), which is a critical mediator of cardioprotection via anesthetic preconditioning (96). Sevoflurane provides cardioprotection against I/R injury via the ROS-mediated upregulation of autophagy (97). Hariharan et al reported that oxidative stress triggers autophagic flux during MI/R injury (98). In addition, trimetazidine (99) and thioredoxin-2 (100) protect against hypoxia/reoxygenation injury by promoting the AMPK-dependent autophagic flux in H9c2 cardiomyocytes. The AMPK/mTOR pathway-regulated autophagy and autophagy-mediated function in I/R injury are illustrated in Fig. 2.

PI-3K/Akt/mTOR pathway

PI-3K/Akt/mTOR signaling may also provide an additional benefit against I/R injury (101). In addition, class III PI-3K focuses on the formation of autophagosomes, whereas class I PI-3K will inhibit the induction of autophagy through the phosphorylation of Akt and mTOR (70). Thus, the interaction of Akt with mTOR is multifaceted and bidirectional (101). Moreover, the self-regulation of autophagy has been postulated to be regulated by the autophagy-induced inhibition of mTOR (102). Furthermore, rapamycin provided a strong beneficial effect against cardiomyocyte anoxia/reoxygenation injury, which would mediate cardioprotection via autophagy that probably depended on the PI-3K/Akt signaling pathway (103). During prolonged ischemia and I/R, the differential effects of GSK-3β, which is the downstream of PI-3K/Akt, on myocardial injury has been suggested to be determined by changes in autophagy (104). GSK-3β inhibition modulates mTOR-dependent attenuation of autophagy, thereby causing the death of cardiomyocytes during prolonged ischemia while mediating their survival during reperfusion (104). In addition, mTOR activation via GSK-3β has been suggested to provide cardioprotection via autophagy (104). The PI-3K/Akt/mTOR pathway-regulated autophagy and autophagy-mediated function in I/R injury are illustrated in Fig. 3.

Others

The p53 transcription factor is a major regulator of cellular response to acute stress (105). Knockdown of p53 can activate autophagy in cardiomyocytes, thereby protecting the myocardium against ischemic injury (106). Autophagy is inhibited with STAT1 to modulate stress response to I/R in STAT1−/− null hearts (32). In I/R, STAT1 can interact directly with p53 and regulate its functional activity (107), thereby suggesting that STAT1 can act with p53 to modulate autophagy (32). The mitochondrial permeability transition pore (MPTP) plays an important role in myocardial I/R injury, and the opening of MPTP has also been shown to trigger autophagy (108). Making the heart more tolerant to subsequent I/R injury is a crucial step in transient MPTP opening before prolonged ischemia (109,110). Moreover, PKC has been reported to trigger the phosphorylation of a regulatory sub-unit of VPATPase, which subsequently induces autophagy (111113).

Autophagy as a therapeutic target for I/R injury

Microarray analysis showed that autophagy-associated genes and the unfolded protein response were upregulated under the condition of repetitive coronary occlusion achieved during chronic local ischemic conditioning in mice (114,115). In another study, inhibiting mTOR decreased infarct size in mice (116). Rapamycin (116), caloric restriction (117), exercise (118), nitric oxide (119), and lipopolysaccharide (120) has been identified as cardioprotective interventions for triggering autophagy. Gurusamy et al used isolated rat heart models and demonstrated that the induction of IPC via repeated I/R cycles immediately enhanced the expression of LC3-II and Beclin-1 (67). In vivo swine models, infarct size was limited after chloramphenicol succinate was used before ischemia (121). Han et al found that cardioprotection induced by remote limb ischemic postconditioning was associated with elevated autophagy 3 h post-reperfusion (2). A similar phenomenon was observed by Hamacher-Brady et al in HL-1 myocytes; they found that simulated I/R-mediated cell death was prevented by strengthening autophagy, whereas its inhibition caused cell death (40). Other researchers have observed that blocking autophagy via cell-permeable Tat-Atg5K130R concurrently increased infarct size in hearts when treated with SUL (39). Moreover, Tibetan patients with coronary heart disease resist I/R injury during cardiac surgery better than patients living at sea level, which is possibly correlated with the upregulation of basal autophagy resulting from chronic hypoxia (28).

Autophagy has been determined as a significant element of the endogenous defense mechanisms activated by various preconditioning types. Induction of autophagy may represent a novel therapeutic approach to myocardial protection in humans (39). The identification of agents that can rapidly induce autophagy can contribute to the discovery of new cardioprotective drugs (122). In addition, induction of autophagy can preserve heart function during I/R injury (91,121,123). Other studies have suggested that autophagy is detrimental because it contributes to cell death (15,124). The beneficial or detrimental role of autophagy may be a consequence of balance, depending on the extent of autophagy (7). Thus, for autophagy to be effective, searching for a candidate cardioprotective drug that can induce autophagy in a target population is important, together with the appropriate timing and response magnitude (16). Various autophagy modulators on cardiac ischemia/injury are summarized in Table I.

Table I.

Summary of various autophagy modulators on cardiac ischemia/reperfusion injury.

Table I.

Summary of various autophagy modulators on cardiac ischemia/reperfusion injury.

Author, yearDrugsMechanism of actionEffect on cardiac ischemia/reperfusion injury(Refs.)
Huang et al, 2010SulfaphenazoleActivate protein kinase CProtects against myocardial I/R and reduces infarct size(39)
Xie et al, 2015SevofluraneInhibition Beclin 1-mediated autophagic cell deathDelayed cardioprotection(52)
Zheng et al, 2017BerbamineActivate PI3K/Akt pathwayImproved post-ischemic myocardial function and attenuated cell death(72)
Lekli et al, 2010TocotrienolActivate the mTOR pathwayReduces cardiomyocyte apoptosis(84)
Gurusamy et al, 2010ResveratrolActivate mTOR-Rictor survival pathwayAttenuates myocardial I/R injury and reduces infarct size(91)
Zhao et al, 2013AcetylcholineActivate AMPK-mTOR pathwayReduces cardiomyocyte death(92)
Xie et al, 2015H2SActivate AMP-activated protein kinaseProtects against myocardial I/R injury(93)
Zhong et al, 2017TrimetazidineActivate AMPK-mTOR pathwayReduces hypoxia/reoxygenation injury(99)
Wang et al, 2015RapamycinActivate PI3k/Akt pathwayAttenuates anoxia/reoxygenation injury(103)

[i] mTOR, mammalian target of rapamycin; I/R, ischemia/reperfusion.

Conclusions

Recent studies have shown that autophagy plays an important role in I/R injury. Moreover, evidence has emerged that autophagy plays various roles in I/R through multiple mechanisms. Autophagy can trigger a survival signal in the case of myocardial ischemia, whereas defective autophagy during reperfusion is detrimental. Although we have obtained substantial knowledge about the function of autophagy in I/R injury, the autophagy pathway is highly complex and remains far from being understood completely. Additional studies are necessary to identify the molecular components of the autophagy pathway, characterize the role of autophagy in I/R injury, elucidate the diverse processes that regulate autophagy expression and activity, and determine the contribution of autophagy to myocardial infarction protection in humans. Such studies will provide additional insights into the role of autophagy in I/R injury and potentially discover novel therapeutic strategies for treating the diseases.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the National natural science foundation of china (grant no. 81600342) and Graduate student research innovation project of Hunan province (grant no. CX2013B397).

Availability of data and materials

Not applicable.

Authors' contributions

X-LL and M-HL conceived and designed this review. X-LL, L-LX and W-JX contributed the central idea, analyzed most of the data, and wrote the initial draft of the paper. L-LX and M-HL revised the manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yang K, Xu C, Li X and Jiang H: Combination of D942 with curcumin protects cardiomyocytes from ischemic damage through promoting autophagy. J Cardiovasc Pharmacol Therap. 18:570–581. 2013. View Article : Google Scholar

2 

Han Z, Cao J, Song D, Tian L, Chen K, Wang Y, Gao L, Yin Z, Fan Y and Wang C: Autophagy is involved in the cardioprotection effect of remote limb ischemic postconditioning on myocardial ischemia/reperfusion injury in normal mice, but not diabetic mice. PLoS One. 9:e868382014. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Das DK and Maulik N: Preconditioning potentiates redox signaling and converts death signal into survival signal. Arch Biochem Biophys. 420:305–311. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Aoyagi T, Kusakari Y, Xiao CY, Inouye BT, Takahashi M, Scherrer-Crosbie M, Rosenzweig A, Hara K and Matsui T: Cardiac mTOR protects the heart against ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 303:H75–H85. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Li Y, Xiang Y, Zhang S, Wang Y, Yang J, Liu W and Xue F: Intramyocardial injection of thioredoxin 2-expressing lentivirus alleviates myocardial ischemia-reperfusion injury in rats. Am J Transl Res. 9:4428–4439. 2017.PubMed/NCBI

7 

Sasaki Y, Ikeda Y, Iwabayashi M, Akasaki Y and Ohishi M: The impact of autophagy on cardiovascular senescence and diseases. Int Heart J. 58:666–673. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Yang L, Wang H, Shen Q, Feng L and Jin H: Long non-coding RNAs involved in autophagy regulation. Cell Death Dis. 8:e30732017. View Article : Google Scholar : PubMed/NCBI

9 

Green DR, Galluzzi L and Kroemer G: Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science. 333:1109–1112. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Sica V, Galluzzi L, Pedro Bravo-San JM, Izzo V, Maiuri MC and Kroemer G: Organelle-specific initiation of autophagy. Mol Cell. 59:522–539. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Mowers EE, Sharifi MN and MacLeod KF: Functions of autophagy in the tumor microenvironment and cancer metastasis. FEBS J. 2018.doi: 10.1111/febs.14388. View Article : Google Scholar : PubMed/NCBI

12 

Pellacani C and Costa LG: Role of autophagy in environmental neurotoxicity. Environ Pollut. 235:791–805. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Yang X, Cohen MV and Downey JM: Mechanism of cardioprotection by early ischemic preconditioning. Cardiovasc Drugs Ther. 24:225–234. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Levine B and Klionsky DJ: Development by self-digestion: Molecular mechanisms and biological functions of autophagy. Dev Cell. 6:463–477. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Ma H, Guo R, Yu L, Zhang Y and Ren J: Aldehyde dehydrogenase 2 (ALDH2) rescues myocardial ischaemia/reperfusion injury: Role of autophagy paradox and toxic aldehyde. Eur Heart J. 32:1025–1038. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Huang C, Yitzhaki S, Perry CN, Liu W, Giricz Z, Mentzer RM Jr and Gottlieb RA: Autophagy induced by ischemic preconditioning is essential for cardioprotection. J Cardiovasc Transl Res. 3:365–373. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Shintani T and Klionsky DJ: Autophagy in health and disease: A double-edged sword. Science. 306:990–995. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Rubinsztein DC, Gestwicki JE, Murphy LO and Klionsky DJ: Potential therapeutic applications of autophagy. Nat Rev Drug Discov. 6:304–312. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Sciarretta S, Hariharan N, Monden Y, Zablocki D and Sadoshima J: Is autophagy in response to ischemia and reperfusion protective or detrimental for the heart? Pediatr Cardiol. 32:275–281. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES, Fox CS, Franco S, et al: Executive summary: Heart disease and stroke statistics-2014 update: A report from the American Heart Association. Circulation. 129:399–410. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Bulluck H, Yellon DM and Hausenloy DJ: Reducing myocardial infarct size: Challenges and future opportunities. Heart. 102:341–348. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Hamacher-Brady A, Brady NR, Logue SE, Sayen MR, Jinno M, Kirshenbaum LA, Gottlieb RA and Gustafsson AB: Response to myocardial ischemia/reperfusion injury involves Bnip3 and autophagy. Cell Death Differ. 14:146–157. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Decker RS and Wildenthal K: Lysosomal alterations in hypoxic and reoxygenated hearts. I. Ultrastructural and cytochemical changes. Am J Pathol. 98:425–444. 1980.PubMed/NCBI

24 

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

25 

Kassiotis C, Ballal K, Wellnitz K, Vela D, Gong M, Salazar R, Frazier OH and Taegtmeyer H: Markers of autophagy are downregulated in failing human heart after mechanical unloading. Circulation. 120 11 Suppl:S191–S197. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Jahania SM, Sengstock D, Vaitkevicius P, Andres A, Ito BR, Gottlieb RA and Mentzer RM Jr: Activation of the homeostatic intracellular repair response during cardiac surgery. J Am Coll Surg. 216:719–729. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Schiattarella GG and Hill JA: Therapeutic targeting of autophagy in cardiovascular disease. J Mol Cell Cardiol. 95:86–93. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Hu Y, Sun Q, Li Z, Chen J, Shen C, Song Y and Zhong Q: High basal level of autophagy in high-altitude residents attenuates myocardial ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 148:1674–1680. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Hamacher-Brady A, Brady NR and Gottlieb RA: The interplay between pro-death and pro-survival signaling pathways in myocardial ischemia/reperfusion injury: Apoptosis meets autophagy. Cardiovasc Drugs Ther. 20:445–462. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Gustafsson AB and Gottlieb RA: Eat your heart out: Role of autophagy in myocardial ischemia/reperfusion. Autophagy. 4:416–421. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Song X, Kusakari Y, Xiao CY, Kinsella SD, Rosenberg MA, Scherrer-Crosbie M, Hara K, Rosenzweig A and Matsui T: mTOR attenuates the inflammatory response in cardiomyocytes and prevents cardiac dysfunction in pathological hypertrophy. Am J Physiol Cell Physiol. 299:C1256–C1266. 2010. View Article : Google Scholar : PubMed/NCBI

32 

McCormick J, Suleman N, Scarabelli TM, Knight RA, Latchman DS and Stephanou A: STAT1 deficiency in the heart protects against myocardial infarction by enhancing autophagy. J Cell Mol Med. 16:386–393. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Gustafsson AB and Gottlieb RA: Autophagy in ischemic heart disease. Circ Res. 104:150–158. 2009. View Article : Google Scholar : PubMed/NCBI

34 

House SL, Branch K, Newman G, Doetschman T and Jel Schultz J: Cardioprotection induced by cardiac-specific overexpression of fibroblast growth factor-2 is mediated by the MAPK cascade. Am J Physiol Heart Circ Physiol. 289:H2167–H2175. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Baehrecke EH: Autophagy: Dual roles in life and death? Nat Rev Mol Cell Biol. 6:505–510. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Qian J, Ren X, Wang X, Zhang P, Jones WK, Molkentin JD, Fan GC and Kranias EG: Blockade of Hsp20 phosphorylation exacerbates cardiac ischemia/reperfusion injury by suppressed autophagy and increased cell death. Circ Res. 105:1223–1231. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Tsujimoto Y and Shimizu S: Another way to die: Autophagic programmed cell death. Cell Death Differ. 12 Suppl 2:S1528–S1534. 2005. View Article : Google Scholar

38 

Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L and Kroemer G: Cell death modalities: Classification and pathophysiological implications. Cell Death Differ. 14:1237–1243. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Huang C, Liu W, Perry CN, Yitzhaki S, Lee Y, Yuan H, Tsukada YT, Hamacher-Brady A, Mentzer RM Jr and Gottlieb RA: Autophagy and protein kinase C are required for cardioprotection by sulfaphenazole. Am J Physiol Heart Circ Physiol. 298:H570–H579. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Hamacher-Brady A, Brady NR and Gottlieb RA: Enhancing macroautophagy protects against ischemia/reperfusion injury in cardiac myocytes. J Biol Chem. 281:29776–29787. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Huber SM, Misovic M, Mayer C, Rodemann HP and Dittmann K: EGFR-mediated stimulation of sodium/glucose cotransport promotes survival of irradiated human A549 lung adenocarcinoma cells. Radiother Oncol. 103:373–379. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Gottlieb RA and Mentzer RM: Autophagy during cardiac stress: Joys and frustrations of autophagy. Annu Rev Physiol. 72:45–59. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Sciarretta S, Zhai P, Shao D, Zablocki D, Nagarajan N, Terada LS, Volpe M and Sadoshima J: Activation of NADPH oxidase 4 in the endoplasmic reticulum promotes cardiomyocyte autophagy and survival during energy stress through the protein kinase RNA-activated-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/activating transcription factor 4 pathway. Circ Res. 113:1253–1264. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Wei L, Wu RB, Yang CM, Zheng SY and Yu XY: Cardioprotective effect of a hemoglobin-based oxygen carrier on cold ischemia/reperfusion injury. Cardiology. 120:73–83. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Boya P and Kroemer G: Beclin 1: A BH3-only protein that fails to induce apoptosis. Oncogene. 28:2125–2127. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Zalckvar E, Berissi H, Eisenstein M and Kimchi A: Phosphorylation of Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL. Autophagy. 5:720–722. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD and Levine B: Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 122:927–939. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Itakura E, Kishi C, Inoue K and Mizushima N: Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG. Mol Biol Cell. 19:5360–5372. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Martinet W, Knaapen MW, Kockx MM and De Meyer GR: Autophagy in cardiovascular disease. Trends Mol Med. 13:482–491. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Furuya N, Yu J, Byfield M, Pattingre S and Levine B: The evolutionarily conserved domain of Beclin 1 is required for Vps34 binding, autophagy and tumor suppressor function. Autophagy. 1:46–52. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Valentim L, Laurence KM, Townsend PA, Carroll CJ, Soond S, Scarabelli TM, Knight RA, Latchman DS and Stephanou A: Urocortin inhibits Beclin1-mediated autophagic cell death in cardiac myocytes exposed to ischaemia/reperfusion injury. J Mol Cell Cardiol. 40:846–852. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Xie H, Liu Q, Qiao S, Jiang X and Wang C: Delayed cardioprotection by sevoflurane preconditioning: A novel mechanism via inhibiting Beclin 1-mediated autophagic cell death in cardiac myocytes exposed to hypoxia/reoxygenation injury. Int J Clin Exp Pathol. 8:217–226. 2015.PubMed/NCBI

53 

Peng W, Liu Y, Xu WJ and Xia QH: Role of Beclin 1-dependent autophagy in cardioprotection of ischemic preconditioning. J Huazhong Univ Sci Technolog Med Sci. 33:51–56. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Levine B, Sinha S and Kroemer G: Bcl-2 family members: Dual regulators of apoptosis and autophagy. Autophagy. 4:600–606. 2008. View Article : Google Scholar :

55 

Shimizu S, Kanaseki T, Mizushima N, Mizuta T, Arakawa-Kobayashi S, Thompson CB and Tsujimoto Y: Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat Cell Biol. 6:1221–1228. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Brocheriou V, Hagege AA, Oubenaissa A, Lambert M, Mallet VO, Duriez M, Wassef M, Kahn A, Menasché P and Gilgenkrantz H: Cardiac functional improvement by a human Bcl-2 transgene in a mouse model of ischemia/reperfusion injury. J Gene Med. 2:326–333. 2000. View Article : Google Scholar : PubMed/NCBI

57 

Imahashi K, Schneider MD, Steenbergen C and Murphy E: Transgenic expression of Bcl-2 modulates energy metabolism, prevents cytosolic acidification during ischemia and reduces ischemia/reperfusion injury. Circ Res. 95:734–741. 2004. View Article : Google Scholar : PubMed/NCBI

58 

Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman JE, Berry G, Herman B and Levine B: Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol. 72:8586–8596. 1998.PubMed/NCBI

59 

Ke J, Yao B, Li T, Cui S and Ding H: A2 Adenosine receptor-mediated cardioprotection against reperfusion injury in rat hearts is associated with autophagy downregulation. J Cardiovasc Pharmacol. 66:25–34. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Maiuri MC, Le Toumelin G, Criollo A, Rain JC, Gautier F, Juin P, Tasdemir E, Pierron G, Troulinaki K, Tavernarakis N, et al: Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J. 26:2527–2539. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Weston CR and Davis RJ: The JNK signal transduction pathway. Curr Opin Cell Biol. 19:142–149. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Zhao Y and Herdegen T: Cerebral ischemia provokes a profound exchange of activated JNK isoforms in brain mitochondria. Mol Cell Neurosci. 41:186–195. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Xu J, Qin X, Cai X, Yang L, Xing Y, Li J, Zhang L, Tang Y, Liu J, Zhang X and Gao F: Mitochondrial JNK activation triggers autophagy and apoptosis and aggravates myocardial injury following ischemia/reperfusion. Biochim Biophys Acta. 1852:262–270. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Madesh M, Antonsson B, Srinivasula SM, Alnemri ES and Hajnoczky G: Rapid kinetics of tBid-induced cytochrome c and Smac/DIABLO release and mitochondrial depolarization. J Biol Chem. 277:5651–5659. 2002. View Article : Google Scholar : PubMed/NCBI

65 

Yang J, Liu X, Bhalla K, Kim CN, Ibrado AM, Cai J, Peng TI, Jones DP and Wang X: Prevention of apoptosis by Bcl-2: Release of cytochrome c from mitochondria blocked. Science. 275:1129–1132. 1997. View Article : Google Scholar : PubMed/NCBI

66 

Chu G, Egnaczyk GF, Zhao W, Jo SH, Fan GC, Maggio JE, Xiao RP and Kranias EG: Phosphoproteome analysis of cardiomyocytes subjected to beta-adrenergic stimulation: Identification and characterization of a cardiac heat shock protein p20. Circ Res. 94:184–193. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Gurusamy N, Lekli I, Gorbunov NV, Gherghiceanu M, Popescu LM and Das DK: Cardioprotection by adaptation to ischaemia augments autophagy in association with BAG-1 protein. J Cell Mol Med. 13:373–387. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Overbye A, Fengsrud M and Seglen PO: Proteomic analysis of membrane-associated proteins from rat liver autophagosomes. Autophagy. 3:300–322. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Townsend PA, Stephanou A, Packham G and Latchman DS: BAG-1: A multi-functional pro-survival molecule. Int J Biochem Cell Biol. 37:251–259. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Petiot A, Ogier-Denis E, Blommaart EF, Meijer AJ and Codogno P: Distinct classes of phosphatidylinositol 3′-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J Biol Chem. 275:992–998. 2000. View Article : Google Scholar : PubMed/NCBI

71 

Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI and Deretic V: Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell. 119:753–766. 2004. View Article : Google Scholar : PubMed/NCBI

72 

Zheng Y, Gu S, Li X, Tan J, Liu S, Jiang Y, Zhang C, Gao L and Yang HT: Berbamine postconditioning protects the heart from ischemia/reperfusion injury through modulation of autophagy. Cell Death Dis. 8:e25772017. View Article : Google Scholar : PubMed/NCBI

73 

Herzig S and Shaw RJ: AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 19:121–135. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Hardie DG and Sakamoto K: AMPK: A key sensor of fuel and energy status in skeletal muscle. Physiology (Bethesda). 21:48–60. 2006.PubMed/NCBI

75 

Meley D, Bauvy C, Houben-Weerts JH, Dubbelhuis PF, Helmond MT, Codogno P and Meijer AJ: AMP-activated protein kinase and the regulation of autophagic proteolysis. J Biol Chem. 281:34870–34879. 2006. View Article : Google Scholar : PubMed/NCBI

76 

Samari HR and Seglen PO: Inhibition of hepatocytic autophagy by adenosine, aminoimidazole-4-carboxamide riboside and N6-mercaptopurine riboside. Evidence for involvement of amp-activated protein kinase. J Biol Chem. 273:23758–23763. 1998. View Article : Google Scholar : PubMed/NCBI

77 

Rohailla S, Clarizia N, Sourour M, Sourour W, Gelber N, Wei C, Li J and Redington AN: Acute, delayed and chronic remote ischemic conditioning is associated with downregulation of mTOR and enhanced autophagy signaling. PLoS One. 9:e1112912014. View Article : Google Scholar : PubMed/NCBI

78 

Kandadi MR, Hu N and Ren J: ULK1 plays a critical role in AMPK-mediated myocardial autophagy and contractile dysfunction following acute alcohol challenge. Curr Pharm Des. 19:4874–4887. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Park CW, Hong SM, Kim ES, Kwon JH, Kim KT, Nam HG and Choi KY: BNIP3 is degraded by ULK1-dependent autophagy via MTORC1 and AMPK. Autophagy. 9:345–360. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Przyklenk K, Undyala VV, Wider J, Sala-Mercado JA, Gottlieb RA and Mentzer RM Jr: Acute induction of autophagy as a novel strategy for cardioprotection: Getting to the heart of the matter. Autophagy. 7:432–433. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Dunlop EA, Hunt DK, Acosta-Jaquez HA, Fingar DC and Tee AR: ULK1 inhibits mTORC1 signaling, promotes multisite Raptor phosphorylation and hinders substrate binding. Autophagy. 7:737–747. 2011. View Article : Google Scholar : PubMed/NCBI

82 

Kim J, Kundu M, Viollet B and Guan KL: AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 13:132–141. 2011. View Article : Google Scholar : PubMed/NCBI

83 

Fiordaliso F, Li B, Latini R, Sonnenblick EH, Anversa P, Leri A and Kajstura J: Myocyte death in streptozotocin-induced diabetes in rats in angiotensin II-dependent. Lab Invest. 80:513–527. 2000. View Article : Google Scholar : PubMed/NCBI

84 

Lekli I, Ray D, Mukherjee S, Gurusamy N, Ahsan MK, Juhasz B, Bak I, Tosaki A, Gherghiceanu M, Popescu LM and Das DK: Co-ordinated autophagy with resveratrol and gamma-tocotrienol confers synergetic cardioprotection. J Cell Mol Med. 14:2506–2518. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Li Y, Wang Y, Kim E, Beemiller P, Wang CY, Swanson J, You M and Guan KL: Bnip3 mediates the hypoxia-induced inhibition on mammalian target of rapamycin by interacting with Rheb. J Biol Chem. 282:35803–35813. 2007. View Article : Google Scholar : PubMed/NCBI

86 

Hamacher-Brady A, Brady NR, Gottlieb RA and Gustafsson AB: Autophagy as a protective response to Bnip3-mediated apoptotic signaling in the heart. Autophagy. 2:307–309. 2006. View Article : Google Scholar : PubMed/NCBI

87 

Xu W, Jiang H, Hu X and Fu W: Effects of high-mobility group box 1 on the expression of Beclin-1 and LC3 proteins following hypoxia and reoxygenation injury in rat cardiomyocytes. Int J Clin Exp Med. 7:5353–5357. 2014.PubMed/NCBI

88 

Ouyang F, Huang H, Zhang M, Chen M, Huang F and Zhou S: HMGB1 induces apoptosis and EMT in association with increased autophagy following H/R injury in cardiomyocytes. Int J Mol Med. 37:679–689. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Sciarretta S, Zhai P, Shao D, Maejima Y, Robbins J, Volpe M, Condorelli G and Sadoshima J: Rheb is a critical regulator of autophagy during myocardial ischemia: Pathophysiological implications in obesity and metabolic syndrome. Circulation. 125:1134–1146. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Shi WY, Xiao D, Wang L, Dong LH, Yan ZX, Shen ZX, Chen SJ, Chen Y and Zhao WL: Therapeutic metformin/AMPK activation blocked lymphoma cell growth via inhibition of mTOR pathway and induction of autophagy. Cell Death Dis. 3:e2752012. View Article : Google Scholar : PubMed/NCBI

91 

Gurusamy N, Lekli I, Mukherjee S, Ray D, Ahsan MK, Gherghiceanu M, Popescu LM and Das DK: Cardioprotection by resveratrol: A novel mechanism via autophagy involving the mTORC2 pathway. Cardiovasc Res. 86:103–112. 2010. View Article : Google Scholar : PubMed/NCBI

92 

Zhao M, Sun L, Yu XJ, Miao Y, Liu JJ, Wang H, Ren J and Zang WJ: Acetylcholine mediates AMPK-dependent autophagic cytoprotection in H9c2 cells during hypoxia/reoxygenation injury. Cell Physiol Biochem. 32:601–613. 2013. View Article : Google Scholar : PubMed/NCBI

93 

Xie H, Xu Q, Jia J, Ao G, Sun Y, Hu L, Alkayed NJ, Wang C and Cheng J: Hydrogen sulfide protects against myocardial ischemia and reperfusion injury by activating AMP-activated protein kinase to restore autophagic flux. Biochem Biophys Res Commun. 458:632–638. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Takagi H, Matsui Y, Hirotani S, Sakoda H, Asano T and Sadoshima J: AMPK mediates autophagy during myocardial ischemia in vivo. Autophagy. 3:405–407. 2007. View Article : Google Scholar : PubMed/NCBI

95 

Yang Y, Wang H, Wang S, Xu M, Liu M, Liao M, Frank JA, Adhikari S, Bower KA, Shi X, et al: GSK3β signaling is involved in ultraviolet B-induced activation of autophagy in epidermal cells. Int J Oncol. 41:1782–1788. 2012. View Article : Google Scholar : PubMed/NCBI

96 

Onishi A, Miyamae M, Kaneda K, Kotani J and Figueredo VM: Direct evidence for inhibition of mitochondrial permeability transition pore opening by sevoflurane preconditioning in cardiomyocytes: Comparison with cyclosporine A. Eur J Pharmacol. 675:40–46. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Shiomi M, Miyamae M, Takemura G, Kaneda K, Inamura Y, Onishi A, Koshinuma S, Momota Y, Minami T and Figueredo VM: Sevoflurane induces cardioprotection through reactive oxygen species-mediated upregulation of autophagy in isolated guinea pig hearts. J Anesth. 28:593–600. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Hariharan N, Zhai P and Sadoshima J: Oxidative stress stimulates autophagic flux during ischemia/reperfusion. Antioxid Redox Signal. 14:2179–2190. 2011. View Article : Google Scholar : PubMed/NCBI

99 

Zhong Y, Zhong P, He S, Zhang Y, Tang L, Ling Y, Fu S, Tang Y, Yang P, Luo T, et al: Trimetazidine protects cardiomyocytes against hypoxia/reoxygenation injury by promoting AMP-activated protein kinase-dependent autophagic flux. J Cardiovasc Pharmacol. 69:389–397. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Li YY, Xiang Y, Zhang S, Wang Y, Yang J, Liu W and Xue FT: Thioredoxin-2 protects against oxygen-glucose deprivation/reperfusion injury by inhibiting autophagy and apoptosis in H9c2 cardiomyocytes. Am J Transl Res. 9:1471–1482. 2017.PubMed/NCBI

101 

Shiomi M, Miyamae M, Takemura G, Kaneda K, Inamura Y, Onishi A, Koshinuma S, Momota Y, Minami T and Figueredo VM: Induction of autophagy restores the loss of sevoflurane cardiac preconditioning seen with prolonged ischemic insult. Eur J Pharmacol. 724:58–66. 2014. View Article : Google Scholar : PubMed/NCBI

102 

He C and Klionsky DJ: Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 43:67–93. 2009. View Article : Google Scholar : PubMed/NCBI

103 

Wang LQ, Cheng XS, Huang CH, Huang B and Liang Q: Rapamycin protects cardiomyocytes against anoxia/reoxygenation injury by inducing autophagy through the PI3k/Akt pathway. J Huazhong Univ Sci Technolog Med Sci. 35:10–15. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Zhai P, Sciarretta S, Galeotti J, Volpe M and Sadoshima J: Differential roles of GSK-3β during myocardial ischemia and ischemia/reperfusion. Circ Res. 109:502–511. 2011. View Article : Google Scholar : PubMed/NCBI

105 

Horn HF and Vousden KH: Coping with stress: Multiple ways to activate p53. Oncogene. 26:1306–1316. 2007. View Article : Google Scholar : PubMed/NCBI

106 

Hoshino A, Matoba S, Iwai-Kanai E, Nakamura H, Kimata M, Nakaoka M, Katamura M, Okawa Y, Ariyoshi M, Mita Y, et al: p53-TIGAR axis attenuates mitophagy to exacerbate cardiac damage after ischemia. J Mol Cell Cardiol. 52:175–184. 2012. View Article : Google Scholar : PubMed/NCBI

107 

Townsend PA, Scarabelli TM, Davidson SM, Knight RA, Latchman DS and Stephanou A: STAT-1 interacts with p53 to enhance DNA damage-induced apoptosis. J Biol Chem. 279:5811–5820. 2004. View Article : Google Scholar : PubMed/NCBI

108 

Halestrap AP, Clarke SJ and Javadov SA: Mitochondrial permeability transition pore opening during myocardial reperfusion-a target for cardioprotection. Cardiovasc Res. 61:372–385. 2004. View Article : Google Scholar : PubMed/NCBI

109 

Hausenloy D, Wynne A, Duchen M and Yellon D: Transient mitochondrial permeability transition pore opening mediates preconditioning-induced protection. Circulation. 109:1714–1717. 2004. View Article : Google Scholar : PubMed/NCBI

110 

Saotome M, Katoh H, Yaguchi Y, Tanaka T, Urushida T, Satoh H and Hayashi H: Transient opening of mitochondrial permeability transition pore by reactive oxygen species protects myocardium from ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 296:H1125–H1132. 2009. View Article : Google Scholar : PubMed/NCBI

111 

Nanda A, Gukovskaya A, Tseng J and Grinstein S: Activation of vacuolar-type proton pumps by protein kinase C. Role in neutrophil pH regulation. J Biol Chem. 267:22740–22746. 1992.PubMed/NCBI

112 

Nordstrom T, Grinstein S, Brisseau GF, Manolson MF and Rotstein OD: Protein kinase C activation accelerates proton extrusion by vacuolar-type H(+)-ATPases in murine peritoneal macrophages. FEBS Lett. 350:82–86. 1994. View Article : Google Scholar : PubMed/NCBI

113 

Voss M, Vitavska O, Walz B, Wieczorek H and Baumann O: Stimulus-induced phosphorylation of vacuolar H (+)-ATPase by protein kinase A. J Biol Chem. 282:33735–33742. 2007. View Article : Google Scholar : PubMed/NCBI

114 

Shen YT, Depre C, Yan L, Park JY, Tian B, Jain K, Chen L, Zhang Y, Kudej RK, Zhao X, et al: Repetitive ischemia by coronary stenosis induces a novel window of ischemic preconditioning. Circulation. 118:1961–1969. 2008. View Article : Google Scholar : PubMed/NCBI

115 

Depre C, Park JY, Shen YT, Zhao X, Qiu H, Yan L, Tian B, Vatner SF and Vatner DE: Molecular mechanisms mediating preconditioning following chronic ischemia differ from those in classical second window. Am J Physiol Heart Circ Physiol. 299:H752–H762. 2010. View Article : Google Scholar : PubMed/NCBI

116 

Khan S, Salloum F, Das A, Xi L, Vetrovec GW and Kukreja RC: Rapamycin confers preconditioning-like protection against ischemia-reperfusion injury in isolated mouse heart and cardiomyocytes. J Mol Cell Cardiol. 41:256–264. 2006. View Article : Google Scholar : PubMed/NCBI

117 

Marzetti E, Wohlgemuth SE, Anton SD, Bernabei R, Carter CS and Leeuwenburgh C: Cellular mechanisms of cardioprotection by calorie restriction: State of the science and future perspectives. Clin Geriatr Med. 25:715–732. 2009. View Article : Google Scholar : PubMed/NCBI

118 

Kavazis AN, Alvarez S, Talbert E, Lee Y and Powers SK: Exercise training induces a cardioprotective phenotype and alterations in cardiac subsarcolemmal and intermyofibrillar mitochondrial proteins. Am J Physiol Heart Circ Physiol. 297:H144–H152. 2009. View Article : Google Scholar : PubMed/NCBI

119 

Jones SP and Bolli R: The ubiquitous role of nitric oxide in cardioprotection. J Mol Cell Cardiol. 40:16–23. 2006. View Article : Google Scholar : PubMed/NCBI

120 

Ha T, Hua F, Liu X, Ma J, McMullen JR, Shioi T, Izumo S, Kelley J, Gao X, Browder W, et al: Lipopolysaccharide-induced myocardial protection against ischaemia/reperfusion injury is mediated through a PI3K/Akt-dependent mechanism. Cardiovasc Res. 78:546–553. 2008. View Article : Google Scholar : PubMed/NCBI

121 

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

122 

Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK and Sadoshima J: Aging and autophagy in the heart. Circ Res. 118:1563–1576. 2016. View Article : Google Scholar : PubMed/NCBI

123 

Gottlieb RA and Mentzer RM Jr: Cardioprotection through autophagy: Ready for clinical trial? Autophagy. 7:434–435. 2011. View Article : Google Scholar : PubMed/NCBI

124 

Kanamori H, Takemura G, Goto K, Maruyama R, Tsujimoto A, Ogino A, Takeyama T, Kawaguchi T, Watanabe T, Fujiwara T, et al: The role of autophagy emerging in postinfarction cardiac remodelling. Cardiovasc Res. 91:330–339. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 18 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin XL, Xiao WJ, Xiao LL and Liu MH: Molecular mechanisms of autophagy in cardiac ischemia/reperfusion injury (Review). Mol Med Rep 18: 675-683, 2018
APA
Lin, X., Xiao, W., Xiao, L., & Liu, M. (2018). Molecular mechanisms of autophagy in cardiac ischemia/reperfusion injury (Review). Molecular Medicine Reports, 18, 675-683. https://doi.org/10.3892/mmr.2018.9028
MLA
Lin, X., Xiao, W., Xiao, L., Liu, M."Molecular mechanisms of autophagy in cardiac ischemia/reperfusion injury (Review)". Molecular Medicine Reports 18.1 (2018): 675-683.
Chicago
Lin, X., Xiao, W., Xiao, L., Liu, M."Molecular mechanisms of autophagy in cardiac ischemia/reperfusion injury (Review)". Molecular Medicine Reports 18, no. 1 (2018): 675-683. https://doi.org/10.3892/mmr.2018.9028