Involvement of non‑coding RNAs in the pathogenesis of myocardial ischemia/reperfusion injury (Review)

  • Authors:
    • Qi Li
    • Zhuqing Li
    • Zhixing Fan
    • Ying Yang
    • Chengzhi Lu
  • View Affiliations

  • Published online on: February 5, 2021     https://doi.org/10.3892/ijmm.2021.4875
  • Article Number: 42
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Myocardial ischemia/reperfusion injury (MIRI) may cause myocardial stunning, reperfusion arrhythmia, no‑reflow phenomenon and lethal reperfusion injury, which has a significant effect on the prognosis of patients undergoing thrombolytic agent therapy and percutaneous coronary intervention. Increasing evidence suggests that apoptosis, innate inflammation, oxidative stress, calcium overload and autophagy are involved in the pathogenesis of MIRI. Recent advancements in RNA sequencing technologies and genome‑wide analyses led to the finding of small non‑coding RNAs (ncRNAs). ncRNAs modulate cellular processes such as signal transduction, transcription, chromatin remodeling and post‑transcriptional modification. The effects of ncRNAs on cellular biology is more considerable than initially expected, and thus ncRNAs have gained increasing attention and focus in modern medical research. There are several types of ncRNAs, such as microRNAs (miRNAs), long non‑coding RNAs (lncRNAs) and circular RNAs (circRNAs), which have been shown to regulate gene expression at the transcription, post‑transcription and epigenetic levels. Dysregulation of ncRNAs, including miRNAs, lncRNAs and circRNAs, may participate in the molecular mechanisms of MIRI. The present review summarizes the characteristics and biological roles of miRNAs, lncRNAs and circRNAs, with particular emphasis on their role in MIRI, which show the novel complexity of ischemic hearts and may offer valuable insights into the pathogenesis of MIRI.

1. Introduction

Myocardial infarction (MI) is one of the main causes of morbidity and mortality in the United States, where approximately every 40 sec, an individual will suffer from MI (1). Recovering reperfusion using thrombolytic agents and percutaneous coronary intervention may rapidly reverse myocardial ischemia and preserve cardiac systolic function (2). However, the recanalization of blood flow can also result in irreversible detrimental effects known as myocardial ischemia/reperfusion injury (MIRI), which may cause myocardial stunning, reperfusion arrhythmia, or the no-reflow phenomenon and lethal reperfusion injury. Over the past two decades, a growing number of studies have demonstrated that apoptosis, innate inflammation, oxidative stress, calcium overload and autophagy are involved in the pathogenesis of MIRI (3-5). However, there is still no effective strategy for limiting or preventing MIRI. Thus, developing novel therapies for MIRI is crucial and remains an opportunity that will provide significant clinical benefits.

The human genome contains approximately 20,000 protein-coding genes. Only 2% of the human genome is transcribed into RNA transcripts, which are translated into protein, with the vast majority of transcripts comprising non-coding RNAs (ncRNAs) (6,7). There are numerous types of ncRNAs, which are generally divided into two categories according to their nucleotide length: Long ncRNAs (lncRNAs), which are ≥200 nucleotides in length, and small ncRNAs, which are ≤200 nucleotides in length, such as microRNAs (miRNAs/miRs) (8,9). The newly defined classes of lncRNAs and miRNAs have been shown to regulate gene expression at the transcription, post-transcription and epigenetic levels. In addition, circRNAs have been identified as part of the lncRNA family, which also play important roles in the regulation of gene expression (10,11). These RNAs are covalently closed, endogenous biomolecules with no 5′ end caps or 3′ poly(A) tails, are not easily degraded by ribonuclease R and are more stable than other lncRNAs (10). The biogenesis of ncRNAs is complex, and the specific process of each ncRNA is shown in Fig. 1. Recent findings have shown that several novel ncRNAs can participate in a variety of important biological control processes and the development of multiple cardiovascular diseases, including MIRI (9,12-14). Such leads, not only enhance the understanding of disease pathogenesis, but also identify non-coding transcripts that potentially serve as MIRI diagnosis biomarkers or therapeutic targets.

The present review summarizes the characteristics and biological roles of miRNAs, lncRNAs and circRNAs, with particular emphasis on their role in MIRI. A more comprehensive understanding of these ncRNAs may bridge a major gap in the knowledge of the molecular mechanisms in the pathogenesis of MIRI.

2. Biological functions of miRNA

miRNAs, which are typically 20-22 nucleotides in length, are highly conserved and single-stranded ncRNAs that are encoded by the genome of several DNA viruses and all eukaryotic organisms (15). Their primary function is to negatively modulate gene expression by binding to the target mRNA and subsequently inhibiting its translation or promoting degradation in a sequence-dependent manner (16). Only mature miRNAs are incorporated into the RNA-induced silencing complex to direct mRNA target silencing. Canonically, this complex binds a target gene via partial sequence complementarity between the miRNA and a conserved site within the 3′-untranslated region of their target mRNAs. There are differences in the complementary degree between miRNAs: A single gene can be modulated by several miRNAs, whereas mRNA duplexes enable a single miRNA to target multiple mRNAs (17). miRNAs can regulate the translation of >60% of protein-coding genes. Furthermore, miRNAs can be sequestered by pseudogenes or other lncRNAs, introducing a new layer of regulatory complexity (18).

miRNAs that are present in cardiac tissue have important functional implications in cardiovascular biology, such as proliferation, smooth muscle maturation, cardiogenesis and endothelial function (19-22). All metabolic activities of an organism in the cardiovascular system are considered to be influenced by miRNA regulatory processes. Increasing evidence has shown that miRNAs are involved in the pathogenesis of MIRI, including inflammation, oxidative stress and apoptosis. Table I shows a summary of some important miRNAs (23-25).

Table I

Summary of miRNAs in the pathogenesis of MIRI.

Table I

Summary of miRNAs in the pathogenesis of MIRI.

miRNAExpression with MIRITargeted genesPathological mechanism(Refs.)
miR-1IncreaseMAPK3Pro-apoptotic(13)
miR-15b-5pIncreaseKCNJ2Pro-apoptotic(31)
miR-22DecreaseCBP Anti-inflammatory(38)
miR-29aIncreaseSIRT1pro-oxidative stress(41)
miR-30eDecreaseNotch1 Anti-apoptotic
Anti-oxidative stress
(4)
miR-128-1-5pDecreaseGadd45gAnti-apoptotic(28)
miR-129-5pDecreaseHMGB1Anti-apoptotic(29)
miR-135aDecreasePTP1BAnti-apoptotic(26)
miR-138DecreaseHIF1-αAnti-apoptotic(27)
miR-140DecreaseYES1Anti-apoptotic(30)
miR-145DecreaseCaMKII
NF-κB
Anti-apoptotic
Anti-inflammatory
(31)
miR-181aDecreasec-Fos Anti-inflammatory(24)
miR-193bDecreaseMAML1Anti-apoptotic(23)
miR-202-3pDecreaseTRPM6 Anti-inflammatory
Anti-oxidative stress
(36)
miR-327IncreaseARCPro-apoptotic(34)
RP105 Pro-inflammatory(39)
miR-330IncreaseSRYVentricular r;emodeling(42)
miR-421IncreaseSirt3 Pro-apoptotic
Pro-oxidative stress
(25)
miR-483-3pIncreaseMDM4Anti-apoptotic(35)
miR-489IncreaseSPIN1Pro-apoptotic(33)
miR-590-3pDecreaseRIPK1 Anti-apoptotic
Anti-inflammatory
Anti-oxidative stress
(40)

[i] miRNA, microRNA; MIRI, myocardial ischemia/reperfusion injury.

3. Role of miRNAs in MIRI

miRNAs as mediators of cardiomyocyte apoptosis in MIRI

Cardiomyocyte apoptosis has been proven to be the main cause responsible for the secondary injury of the myocardium during MIRI. In addition, several miRNAs have been identified to be involved in the pathogenesis of MIRI through regulating cardiomyocyte apoptosis. It has been documented that the increased expression of miR-30e, miR-193b, miR-135a, miR-138, miR-140, miR-145, miR-15b-5p, miR-483 and miR-590-3p could suppress cardiomyocyte apoptosis induced by MIRI. Similar findings were observed following the decreased expression of miR-1, miR-327, miR-128-1-5p, miR-129-5p, miR-489 and miR-421 (4,13,23,25-35). Zhang et al (23) indicated that miR-193b overexpression may reduce apoptosis after MIRI and alleviate IR-induced myocardial injury by targeting mastermind-like 1, which is a transcriptional coactivator in the Notch signaling pathway. Wang et al (26) revealed that miR-135a targeted and negatively regulated protein tyrosine phosphatase 1B (PTP1B), and the expression of proapoptotic-related genes was reduced in association with PTP1B downregulation. In addition, miR-135a improved MIRI by reducing cardiomyocyte apoptosis both in vitro and in vivo. Liu et al (27) found that miR-138 upregulation inhibited the expression of proteins related to mitochondrial morphology and apoptosis by targeting hypoxia inducible factor-α (HIF1-α), thus exerting protective functions against MIRI.

Additionally, miR-128-1-5p was found to play a cardioprotective role in both H9c2 cardiomyocytes after hypoxia/reoxygenation (H/R) and H2O2-treated neonatal rat cardiomyocytes. This may have occurred by the regulation of miR-128-1-5p on growth arrest DNA damage-inducible gene 45 γ-mediated cardiomyocyte apoptosis in MIRI (28). Moreover, miR-129-5p overexpression could also attenuate cardiomyocyte apoptosis both in H9c2 cardiomyocytes after H/R and rats after I/R injury by targeting high mobility group box-1 (HMGB1) (29). Yang et al (30) demonstrated that miR-140 overexpression attenuated myocardial infarct size and myocardial enzymes via inhibition of mitochondrial-mediated apoptosis by targeting Yes-associated protein 1 (YES-1) in the progression of MIRI. Liu et al (31) reported that miR-145 exerted protective effects against MIRI by reducing cardiomyocyte apoptosis and the protective effects of miR-145 were possibly attributed to the inhibition of the CaMKII-mediated apoptosis signal-regulating kinase 1 anti-apoptotic signaling pathway.

Furthermore, Hao et al (13) found that inhibition of miR-1 alleviated MIRI by inhibiting MAPK3 via negatively regulating the PI3K/Akt signaling pathway, which is associated with reduced apoptosis. Niu et al (32) reported that miR-15b-5p was upregulated in MIRI rat models and could promote myocardial apoptosis via suppressing potassium voltage-gated channel subfamily J member 2 expression. Similarly, inhibition of miR-489 was found to promote activation of the PI3K/Akt signaling pathway by negatively targeting spindlin-1 and inhibiting apoptosis in H/R H9c2 cells (33). Our latest study reported that miR-327 downregulation improved I/R-induced myocardial injury and suppressed both extrinsic and intrinsic apoptotic cascades via targeting apoptosis repressor with caspase recruitment domain both in vitro and in vivo (34). Recently, inhibition of miR-483-3p was also demonstrated to ameliorate H/R injury and inhibit apoptosis in H9c2 cells via targeting murine double minute 4 via the p53 signaling pathway (35).

miRNAs as mediators of inflammation in MIRI

Innate inflammation is an important pathological characteristic of MIRI, which can lead to the activation of diverse inflammatory factors. This effect can be regulated by miR-145, miR-181a, miR-202-3p, miR-22, miR-327 and miR-590-3p (24,29,32,34-36). Wei et al (24) found that miR-181a could efficiently attenuate MIRI in rats by inhibiting the inflammatory response and increasing the regulatory T-cell ratio. The mechanism of action may involve the inhibition of c-Fos protein, which is considered a key immunoactivator that contributes to dendritic cell-related immune functions from all links and processions. Liu et al (31) also demonstrated that upregulated miR-145 alleviated I/R-induced myocardial electrophysiological instability and inflammatory response, which may predominantly be due to inhibition of the NF-κB p65 anti-inflammatory signaling pathway. At the same time, Wu et al (36) indicated that miR-202-3p upregulation alleviated inflammatory response and oxidative stress by activating the TGF-β1/Smads signaling pathway by targeting transient receptor potential cation channel, subfamily M, member 6 (TRPM6) expression. The TRPM6 gene is a member of the transient receptor potential channel gene family and has been found to play an important role in the regulation of extracellular divalent cations in cardiomyocytes (37). Previous findings showed that miR-22 had a protective effect on MIRI and this protective mechanism, at least in part, was due to its anti-inflammatory function via the suppression of the p38 MAPK/CBP/c-Jun-activator protein-1 (AP-1) signaling pathway (38). In addition to this, our latest findings suggested that inhibition of miR-327 improved MIRI by negatively targeting radioprotective 105 kDa protein by suppressing inflammation in rat models (39).

miRNAs regulate other pathological characteristics of MIRI

In addition to cardiomyocyte apoptosis and innate inflammation, miRNAs can also actively participate in the modulation of oxidative stress and other pathological characteristics of MIRI such as autophagy, pyroptosis and ventricular remodeling. Liu et al (25) reported that miR-421 promotes JNK/AP-1 pathway activation by directly targeting sirtuin-3 and further promotes H/R-induced oxidative stress and caspase-9/3-dependent cardiomyocyte apoptosis. Zheng et al (4) suggested that miR-30e serves a significant role in suppressing MIRI-induced oxidative stress and apoptosis via the Notch1/Hes1/Akt signaling pathway. In H/R H9c2 cells, overexpression of miR-590-3p inhibited activation of the NF-κB signaling pathway by targeting the inhibition of the receptor-interacting protein kinase 1 (RIPK1) gene, thereby alleviating oxidative stress, apoptosis and inflammatory response (40). In another H/R model and MIRI rat models, downregulated miR-29a could improve myocardial injury by negatively targeting sirtuin-1 by suppressing oxidative stress and NOD-like receptor pyrin domain-containing-3 (NLRP3)-mediated pyroptosis (41). Additionally, our unpublished data show that miR-327 knockdown can improve cardiac function after MIRI, partly through the suppression of oxidative stress. Recently, Liu et al (42) demonstrated that inhibition of miR-330 inhibited TGF-β1/Smad3 signaling pathway activation by negatively targeting SRY and thus prevented left ventricular remodeling after MIRI.

Moreover, recent findings indicated that vitamin C attenuated the inflammation, apoptosis and oxidative damage of H2O2-induced human umbilical vein endothelial cells via miRNA signaling networks, including miR-3928-5p and miR-323a-5p (43). Given the important role of inflammation, apoptosis and oxidative stress in MIRI, miR-3928-5p and miR-323a-5p constitute potential therapeutic targets for treating MIRI. Future studies are needed to determine whether these miRNAs that are modified by anti-inflammation, anti-apoptotic and antioxidant effects affect MIRI. Generally, the critical contribution of miRNAs in apoptosis, inflammation, oxidative stress as well as other pathological characteristics via different signaling pathways has opened up new avenues to exploit their role in the pathogenesis of MIRI. Future research should pay attention to the interaction between miRNAs with the regulatory network in view of the contribution to the potential molecular mechanisms of MIRI.

4. Biological functions of lncRNAs

lncRNAs, a class of transcripts without protein-coding capacity, are expressed at low levels with 10-fold lower median expression abundance than their protein-coding counterparts (44). Although most lncRNAs are localized in the nucleus, several lncRNAs also exert functions in the cytoplasm (45). lncRNAs manifest specific and diverse subcellular localizations in various cell types depending on their molecular function. In the nucleus, lncRNAs can interact with DNA to form RNA-DNA complexes to regulate gene expression and act as molecular scaffolds to suppress or activate transcription. In addition, other lncRNAs are enriched in the cytoplasm, where they can also interact with proteins and modulate mRNA translation (46,47). In fact, it was shown that lncRNAs may participate in a wide range of pathophysiological processes and biological events. They regulate target gene expression mainly through cis- or trans-regulation (48).

Increasing evidence has suggested that lncRNAs act as regulators of almost every cellular process, and the expression of these molecules seems to be strictly regulated in physiological conditions and several cardiovascular diseases, including MIRI (14,49). Since the discovery of lncRNAs, their functional characterization makes these molecules a potential biomarker and therapeutic target for MIRI as shown in Table II, which provides a summary of the major lncRNAs.

Table II

Summary of lncRNAs in the pathogenesis of MIRI.

Table II

Summary of lncRNAs in the pathogenesis of MIRI.

lncRNAExpression with MIRITargeted genesPathological mechanism(Refs.)
FOXD3-AS1IncreaseNFκB/iNOS/COX2 Pro-apoptotic
Pro-autophagy
(48)
LINC00652IncreaseGLP-1RPro-apoptotic(49)
AK139128IncreasemiR-499 Pro-apoptotic
Pro-autophagy
(56)
H19Decrease miR-877-3p
Bcl-2
miR-103
Anti-apoptotic
Anti-apoptotic
Anti-apoptotic
(57)
miR-107Anti-necrosis(71)
Oprm1DecreasemiR-30b-5pAnti-apoptotic(54)
TUG1IncreasemiR-142-3p Pro-apoptotic
Pro-autophagy
(58)
Neat1IncreasemiR-193aPro-apoptotic(59)
AK088388IncreasemiR-30a Pro-apoptotic
Pro-autophagy
(60)
MEG3IncreasemiR-7-5pPro-apoptotic(61)
KCNQ1OT1IncreasemiR-204-5pPro-apoptotic(62)
HIF1A-AS1IncreasemiR-204 Pro-apoptotic
ventricular remodeling
(63)
NRFIncreasemiR-873Pro-necrosis(67)
RORIncreasep38/MAPKPro-oxidative stress(73)
Gpr19IncreasemiR-324-5p Pro-apoptotic
Pro-oxidative stress
(74)
GAS5IncreasemiR-532-5pPro-apoptotic(64)
HULC
Decrease
miR-377-5p
Anti-apoptotic
Anti-inflammatory
(70)
Gm4419IncreasemiR-682 Pro-inflammatory
Pro-apoptotic
(71)
MALAT1IncreasemiR-144-3pPro-apoptotic(77)
miR-26b(78)

[i] MIRI, myocardial ischemia/reperfusion injury; lncRNA, long non-coding RNAs.

5. Role of lncRNAs in MIRI

LncRNAs as mediators of cardiomyocyte autophagy and apoptosis in MIRI

To date, a variety of lncRNAs have been revealed to participate in the regulation of cardiomyocyte autophagy and apoptosis during the process of MIRI. Ma et al (14) showed that lncRNA nuclear enriched abundant transcript 1 (Neat1) expression was increased after MIRI in diabetic rats, and increased Neat1 expression aggravated autophagy and apoptosis response via regulating forkhead box protein O1 expression during MIRI. LncRNA FOXD3 antisense RNA 1 (FOXD3-AS1) was also found to be upregulated during MIRI. Furthermore, FOXD3-AS1 overexpression promoted cardiomyocyte autophagy and apoptosis via activating the NF-κB/inducible nitric oxide synthase/cyclooxygenase 2 signaling pathway, which led to aggravated MIRI (49). During sevoflurane-induced cardioprotection against MIRI, lncRNA LINC00652 overexpression was identified to reduce this protective effect and increase infarct size and cell apoptosis via inactivating the cAMP/protein kinase A pathway by negatively targeting glucagon-like peptide-1 receptor (GLP-1R) (50). GLP-1R is a gut incretin hormone that exerts an anti-oxidative protective effect on various tissues. It has been identified as a vital physiological regulator of insulin secretion and a major therapeutic target for the treatment of ischemic stroke and diabetes (51,52). As a signaling molecule, hydrogen sulfide (H2S) has been recognized as an endogenous critical gas that participates in the physiological and pathological progression of the cardiovascular system (53). Endogenous H2S is produced by cystathionine-γ-lyase (CSE), which can be inhibited by the miR-30 family (54). Hu et al (55) suggested that lncRNA Mu-type opioid receptor (Oprm1) was competitively combined with miR-30b-5p, which inhibited CSE expression. In-depth investigation showed that Oprm1 overexpression increased endogenous H2S and reduced I/R-induced myocardial injury and apoptosis via activating the PI3K/Akt pathway by targeting the miR-30b-5p/CSE axis (55).

Competing endogenous RNA (ceRNA) is a novel pattern of lncRNA expression, which can indirectly modulate the expression of target genes by competing for binding with miRNAs (56). Zhu and Zhao (57) revealed that lncRNA AK139128 could be used as a ceRNA to adsorb miR-499 to suppress miR-499 expression, thereby promoting cardiomyocyte apoptosis and autophagy in H/R injury. H19, a well-known conserved lncRNA, is also abundantly expressed in murine hearts and can function as a ceRNA to regulate the availability of miR-877-3p. Upregulated H19 attenuated mitochondrial apoptosis and cardiomyocyte injury induced by MIRI via suppressing the miR-877-3p/Bcl-2 pathway (58). Su et al (59) reported that lncRNA taurine-upregulated gene 1 increased the expression of HMGB1 and Rac1 by sponging miR-142-3p, which critically contributed to autophagy and apoptosis under H/R conditions. A recent study by Ren et al (60) also suggested that lncRNA Neat1 played a promotive role in MIRI, and inhibition of lncRNA Neat1 enhanced cell proliferation and inhibited cell apoptosis, which may be the mechanisms through which lncRNA Neat1 negatively targets miR-193a in H/R injury H9c2 cells. Similarly, Wang et al (61) demonstrated that lncRNA AK088388 could increase the expression of LC3-II and Beclin-1 and eventually promote cardiomyocyte apoptosis and autophagy. The mechanism of action was attributed to competitive binding of miR-30a, which derepresses the miR-30a to regulate cardiomyocyte injury in an H/R model. Subsequently, Zou et al (62) reported that lncRNA maternally expressed gene 3 (MEG3) inhibition protected cardiomyocytes against I/R-induced apoptosis both in vitro and in vivo. The potential mechanisms were investigated, and it was found that lncRNA MEG3 negatively targeted miR-7-5p and its downstream target gene poly (ADP-ribose) polymerase (PARP)1 as well as the caspase-3 signaling pathway. In the same year, another study by Rong et al (63) revealed that lncRNA KCNQ1OT1 could regulate galectin-3 (LGALS3) expression by binding to miR-204-5p, which promoted cardiomyocyte apoptosis both in H/R and MIRI mouse models. Thus, the KCNQ1OT1/miR-204-5p/LGALS3 axis may provide a new therapeutic target for MIRI treatment.

It was also shown that lncRNA HIF1A-AS1 could function as a ceRNA of miR-204 to regulate the expression of suppressors of cytokine signaling 2, thus playing a key role in promoting ventricular remodeling and aggravating cardiac function after MIRI in mice (64). Han et al (65) revealed that lncRNA growth arrest specific 5 (GAS5) served a function in MIRI via the PI3K/Akt pathway by sponging miR-532-5p, and silencing lncRNA GAS5 could improve cell survival, decrease the occurrence of apoptosis and ultimately attenuate MIRI. Interestingly, lncRNA GAS5 was also found to function as a ceRNA of miR-137 to suppress miR-137 expression (66). Furthermore, previous findings have shown that miR-137 negatively regulated H2O2-induced cardiomyocyte apoptosis by targeting CDC42 (67). This indicated that the lncRNA GAS5/miR-137/CDC42 axis may also be involved in the regulation of I/R-induced myocardial injury and apoptosis. However, this speculation requires further verification by experimental studies.

LncRNAs as mediators of cell necrosis and inflammation in MIRI

In addition to apoptosis, several lncRNAs can also modulate cell necrosis and inflammation, which is one of the pathological mechanisms of MIRI. Wang et al (68) showed that lncRNA necrosis-related factor contributed to H2O2-induced cardiomyocyte necrosis and I/R-induced myocardial injury by targeting miR-873 and regulating RIPK1/RIPK3. It is well known that RIPK1 is necessary in TRAIL, TNF-α and CD95 ligand-induced necrotic cell death and RIPK3 phosphorylates the mixed lineage kinase domain-like protein, which is also a key determinant in mediating the RIPK1 necrotic signaling pathway (69,70). Liang et al (71) reported that lncRNA HULC was downregulated and miR-377-5p was upregulated during MIRI, and HULC overexpression inhibited the inflammation and apoptosis of H/R-induced H9c2 cells. Mechanistic experiments revealed that lncRNA HULC sponged miR-377-5p to inactivate the NLRP3/caspase-1/IL-1β pathway to exert a protective effect. Furthermore, lncRNA Gm4419 was demonstrated to regulate MIRI by targeting the miR-682/TNF receptor-associated factor 3 axis, and Gm4419 knockdown reduced the myocardial infarction area, inflammatory cytokine levels and apoptosis (72). Another study by Wang et al (73) also revealed a novel function of H19 in modulating H9c2 cell necrosis induced by H2O2 and demonstrated that H19 acts as a ceRNA to repress miR-103/107, thereby regulating the expression of FADD, which acted as a negative regulator of necrosis by preventing the formation of the RIPK1/RIPK3 complex.

lncRNAs regulate other pathological characteristics of MIRI

Blood reflow during MIRI can result in reactive oxygen species (ROS) production, which causes cytotoxic and oxidative stress damage. LncRNA regulator of reprogramming was found to promote ROS production, NADPH oxidase (NOX) activity and NOX2 protein levels by regulating the p38/MAPK signaling pathway, thereby increasing oxidative damage and cardiomyocyte apoptosis as well as aggravating MIRI (74). Huang et al (75) showed that lncRNA Gpr19 was upregulated in an oxygen glucose deprivation/recovery system of neonatal rat ventricular cardiomyocytes or MIRI in mice, and inhibition of Gpr19 attenuated oxidative stress and apoptosis by regulation of the miR-324-5p/mitochondrial fission regulator 1 axis. Additionally, another lncRNA, metastasis associated lung adenocarcinoma transcript 1 (MALAT1), which also acts as a ceRNA for numerous miRNAs, was upregulated in cardiac tissue, and its inhibition reduced cardiomyocyte apoptosis and improved left ventricular function in diabetic rats (76). Wang et al (77) inferred that MALAT1 may modulate cardiomyocyte inflammation and myocardial injury induced by I/R via negatively targeting miR-203 expression. Interestingly, Gong et al (78) recently demonstrated that MALAT1 promoted cardiomyocyte apoptosis following MIRI via targeting miR-144-3p, but not miR-203. However, the effects of MALAT1 in regulating MIRI-induced inflammatory response have not been completely elucidated. It is known that MALAT1 can also function as a ceRNA to suppress miR-26b expression (79). In addition, Ge et al (80) showed that miR-26b reduced the inflammatory response and improved myocardial remodeling during myocardial infarction via binding to prostaglandin-endoperoxide synthase 2 (PTGS2) and inactivating the MAPK pathway. Based on these data, we speculated that the MALAT1-miR-26b-PTGs2 axis may participate in MIRI and aggravate the inflammatory response induced by MIRI.

The exact molecular mechanism of lncRNAs in the pathogenesis of inflammatory response remains to be determined. Overall, the lncRNA-miRNA modulatory network is a potential axis that affects the progression of MIRI by modulating apoptosis, autophagy and oxidative stress. Although our understanding of lncRNAs is still developing, these examples provided valuable insights on how lncRNAs may ultimately be used in the clinic as new therapeutic targets for MIRI in the future.

6. Biological functions of circRNAs

circRNAs, a novel type of large endogenous transcripts, are typically produced by back-splicing events on maturing pre-mRNAs that join a donor site with an upstream acceptor site (81). These circRNAs differ from linear RNAs in that they are circularized RNA molecules with covalently closed loop structures without 5′-3′ polarity or a polyadenylated tail. Most of them are stable, abundant, endogenous and exhibit cell type-, tissue- and developmental stage-specific expression patterns in eukaryotic cells (82). Compared to miRNAs and lncRNAs, the understanding of circRNA functions are still limited. At present, four main roles have been described and are shared by a subset of circRNAs. CircRNAs can function as miRNA sponges to inhibit the translation of mRNAs and interact with RNA-binding proteins to regulate protein levels. In addition, they can also act as regulators of splicing and transcription to alter gene expression and dynamic protein scaffolds to facilitate contact and assembly of proteins (11,83,84).

While the main function of circRNAs is exerted through their activity as miRNA sponges, their second most important function is exerted via circRNA-protein interactions, including as protein sponges, scaffolds, decoys and recruiters (84). In addition, several circRNAs were also reported to encode proteins through the translational machinery in a cap-independent manner (85). Thus, circRNA-protein interactions can also influence protein expression, biogenesis and pathophysiological processes, which requires further in-depth studies. Deep sequencing of cardiac tissues revealed the abundance, conservation, evolution and developmental stage-specific differentiation of circRNAs expressed in the heart (86). Since this has been identified in a number of other investigation areas, circRNA regulation and function in the cardiovascular system have also attracted widespread attention. Increasing evidence has demonstrated that circRNAs are involved in a wide variety of biological processes, such as cell proliferation, differentiation and apoptosis, and may play significant roles in MIRI, as shown in Table III which includes a summary of the major circRNAs (12).

Table III

Summary of circRNAs in the pathogenesis of MIRI.

Table III

Summary of circRNAs in the pathogenesis of MIRI.

circRNAExpression with MIRITargeted genesPathological mechanism(Refs.)
circNCX1IncreasemiR-133a-3pPro-apoptotic(87)
ACRDecreaseDnmt3BAnti-autophagy(93)
CircDLGAP4DecreasemiR-143Anti-ER stress(96)
Anti-apoptotic(98)
CircANXA2IncreasemiRNA-133Pro-apoptotic(88)
circTLK1IncreasemiR-214Pro-apoptotic(89)
circRNA-0068566DecreasemiR-6322 Anti-apoptotic
Anti-oxidative stress
(99)
Circ-100338DecreasemiRNA-200a-3pPro-angiogenic(100)

[i] MIRI, myocardial ischemia/reperfusion injury; circRNA, circular RNAs.

7. Role of circRNAs in MIRI

CircRNAs as mediators of MIRI

More recently, deep sequencing studies revealed that >15,318 and 3,017 cardiac circRNAs were identified in human and mouse heart, respectively, some of which are correlated with apoptosis, autophagy and endoplasmic reticulum (ER) stress during MIRI (12,87). CircNCX1, a circRNA transcribed from the 2nd exon of the sodium/calcium exchanger 1 (ncx1) gene, is widely expressed in cardiac tissue and showed differential expression levels during cardiomyopathy. Li et al (88) revealed that circNCX1 was upregulated in response to MIRI. Furthermore, suppression of circNCX1 reduced apoptosis and attenuated I/R-induced myocardial injury by binding to miR-133a-3p and inhibiting miR-133a-3p regulation on pro-apoptotic gene cell death-inducing protein (CDIP1) activity. Thus, the circNCX1/miR-133a-3p/CDIP1 axis plays a critical regulatory role in regulating cardiomyocyte apoptosis, thus contributing to MIRI. Similarly, Zong and Wang (89) showed that circANXA2 promoted myocardial apoptosis during MIRI via suppressing miRNA-133 expression and aggravated myocardial injury.

Moreover, circTLK1 is an upregulated circRNA found in MIRI mouse models and could exacerbate myocardial injury and cardiomyocyte apoptosis during MIRI by targeting the miR-214/RIPK1-mediated TNF signaling pathway (90). As a conserved mitochondrial serine/threonine protein kinase, PTEN-induced putative kinase 1 (PINK1) can be imported into the mitochondrial matrix and targets the outer membrane of the mitochondria, which ensures quality control of the mitochondria (91,92). Previous findings suggested that PINK1 is downregulated during human end-stage heart failure, which showed that PINK1 is essential for normal heart function (93). Recently, Zhou et al (94) reported that a circRNA known as autophagy-related circular RNA (ACR) attenuated cardiomyocyte autophagy and protected the heart from I/R injury by positively targeting Pink1. Moreover, they identified ACR-activated PINK1 expression by directly binding to Dnmt3B and blocking Dnmt3B-mediated DNA methylation of Pink1 promoter and regulated its downstream gene FAM65B, which inhibits autophagy and cell death in the heart.

Vascular endothelial cell dysfunction plays a significant role in the progression of MIRI, which is involved in angiogenesis via various mechanisms, such as ER stress, oxidative stress, autophagy and ubiquitination (95,96). CircDLGAP4 was found to be decreased in endothelial cells after MIRI. Moreover, miR-143 was the main sponge target of circDLGAP4, which was involved in endothelial cell dysfunction caused by I/R exposure via regulating miR-143 expression (97). One of the potential targets of miR-143 is E3 ubiquitin-protein ligase HECTD1 (HECTD1), a novel protein that participates in cell migration and apoptosis in endothelial cells (98). It was suggested that circDLGAP4 can regulate HECTD1 expression by sponging miR-143 and mediate I/R-induced myocardial injury in endothelial cells through ER stress (97). Another interesting phenomenon was that miR-143 could also act as a sponge for Bcl-2, thereby inhibiting the apoptotic response (99). Consequently, the circDLGAP4/miR-143/Bcl-2 pathway may also play a critical regulatory role in cardiomyocyte apoptosis, thereby attenuating MIRI. Zhou et al (100) reported that circRNA-0068566 had a protective effect on MIRI and may significantly suppress I/R-induced cardiac dysfunction, apoptosis and oxidative stress by activating the miR-6322/PARP2 signaling pathway. In addition, Chang et al (101) revealed that Circ-100338 induced angiogenesis and regulated MIRI metastasis by combining with miRNA-200a-3p.

Other circRNAs identified in MIRI

At present, the understanding of circRNAs in the molecular mechanisms of MIRI remains limited. It is important to further investigate the pathological roles of circRNAs in the development of MIRI or other cardiovascular diseases. Numerous other circRNAs have been implicated in I/R injury. Ge et al (102) recently showed that 119 circRNAs were downregulated and 66 circRNAs were upregulated during MIRI, and 42 of them were newly identified circRNAs. Furthermore, they constructed a circRNA-miRNA network with five circRNAs, including mmu_circ_001007, mmu_circ_008351, mmu_circ_0001336, mmu_circ_008228 and mmu-circ007845, which could modulate miRNA expression. Moreover, circRNA-miRNA network analysis revealed that one miRNA could combine with multiple circRNAs and one circRNA can also function on different miRNAs. The identified circRNAs may participate in the pathology of MIRI. Compared with lncRNAs and miRNAs, the understanding of circRNAs in the molecular mechanisms of diabetic cardiomyopathy is still limited. Future studies are required to verify the exact mechanisms through which circRNAs interact with proteins and other ncRNAs to mediate the pathogenesis of MIRI.

8. Conclusion

ncRNAs, first discovered in 1993 by Lee and colleagues, modulate cellular processes such as signal transduction, transcription, chromatin remodeling and post-transcriptional modification (103). The effects of ncRNAs on cellular biology are more considerable than initially expected and thus makes ncRNAs the focus of modern medical research. Dysregulation of ncRNAs, including miRNAs, lncRNAs and circRNAs, can have a significant impact on myocardial function, thereby aggravating, maintaining and promoting MIRI processes. Another class of ncRNAs known as PIWI-interacting RNAs (piRNAs) was identified, and their regulatory roles in gene expression is increasingly studied (104). Evidence has demonstrated that piRNAs influence the Akt signaling pathway, a crucial network in cardiac physiopathology (105). Moreover, it was recently highlighted that anti-inflammatory treatment with vitamin C to attenuate H2O2-mediated endothelial cell senescence may be associated with changes in the expression of piRNAs that are linked to the cell cycle (106). Thus, piRNAs could play a functional role in inflammation and endothelial dysfunction-related cardiovascular regulatory pathways. Future mechanistic studies are required to determine whether the expression of piRNAs is crucial to MIRI.

The present review summarized the recent progress in the involvement of ncRNAs in the pathogenesis of MIRI, such as apoptosis, autophagy, inflammation and oxidative stress. In fact, a number of miRNAs, more recently circRNAs and lncRNAs, have been shown to be potential therapeutic targets in the treatment of MIRI. The main function of ncRNAs is to regulate the expression of target genes by binding to the target mRNA or sponging miRNAs. The therapeutic targeting of ncRNAs in MIRI has only been recently identified and more information remains to be studied, particularly on lncRNAs and circRNAs. In order to keep up the pace and identify more novel ncRNAs, a highly organized network of ncRNA research institutes is essential. Finally, further studies are required to clarify the molecular mechanisms of ncRNAs in the development of MIRI and determine whether there are other novel molecules that regulate lncRNAs and circRNAs, thus forming novel therapeutic targets for MIRI and other cardiovascular diseases.

In conclusion, the pathological roles of ncRNAs in the progression of MIRI shows the complexity of the ischemic heart and may provide valuable insights into the pathogenesis of ischemic heart disease.

Funding

This study was supported by research grants from the National Natural Science Foundation of China (grant no. 81970303).

Availability of data and materials

Not applicable.

Authors' contributions

QL, ZL, ZF, YY and CL conceived and designed the article. QL and YY prepared the figures. QL and ZL drafted the manuscript. CL and ZF critically revised the article for important intellectual content. All authors read and approved the final manuscript, and agree to be accountable for all aspects of the work.

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

Not applicable.

References

1 

Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, et al: Heart Disease and Stroke Statistics-2019 Update: A report from the American Heart Association. Circulation. 139:e56–e528. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Ibanez B, Heusch G, Ovize M and Van de Werf F: Evolving therapies for myocardial ischemia/reperfusion injury. J Am Coll Cardiol. 65:1454–1471. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Wang EW, Han YY and Jia XS: PAFR-deficiency alleviates myocardial ischemia/reperfusion injury in mice via suppressing inflammation, oxidative stress and apoptosis. Biochem Biophys Res Commun. 495:2475–2481. 2018. View Article : Google Scholar

4 

Zheng J, Li J, Kou B, Yi Q and Shi T: MicroRNA-30e protects the heart against ischemia and reperfusion injury through autophagy and the Notch1/Hes1/Akt signaling pathway. Int J Mol Med. 41:3221–3230. 2018.

5 

Xu T, Ding W, Ao X, Chu X, Wan Q, Wang Y, Xiao D, Yu W, Li M, Yu F and Wang J: ARC regulates programmed necrosis and myocardial ischemia/reperfusion injury through the inhibition of mPTP opening. Redox Biol. 20:414–426. 2019. View Article : Google Scholar

6 

Stein LD: Human genome: End of the beginning. Nature. 431:915–916. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Lorenzen JM and Thum T: Long noncoding RNAs in kidney and cardiovascular diseases. Nat Rev Nephrol. 12:360–373. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Eddy SR: Non-coding RNA genes and the modern RNA world. Nat Rev Genet. 2:919–929. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Beermann J, Piccoli MT, Viereck J and Thum T: Non-coding RNAs in development and disease: Background, mechanisms, and therapeutic approaches. Physiol Rev. 96:1297–1325. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, et al: Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 22:256–264. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK and Kjems J: Natural RNA circles function as efficient microRNA sponges. Nature. 495:384–388. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Altesha MA, Ni T, Khan A, Liu K and Zheng X: Circular RNA in cardiovascular disease. J Cell Physiol. 234:5588–5600. 2019. View Article : Google Scholar

13 

Hao YL, Fang HC, Zhao HL, Li XL, Luo Y, Wu BQ, Fu MJ, Liu W, Liang JJ and Chen XH: The role of microRNA-1 targeting of MAPK3 in myocardial ischemia-reperfusion injury in rats undergoing sevoflurane preconditioning via the PI3K/Akt pathway. Am J Physiol Cell Physiol. 315:C380–C388. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Ma M, Hui J, Zhang QY, Zhu Y, He Y and Liu XJ: Long non-coding RNA nuclear-enriched abundant transcript 1 inhibition blunts myocardial ischemia reperfusion injury via autophagic flux arrest and apoptosis in streptozotocin-induced diabetic rats. Atherosclerosis. 277:113–122. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S and Kim VN: The nuclear RNase III Drosha initiates microRNA processing. Nature. 425:415–419. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Meister G and Tuschl T: Mechanisms of gene silencing by double-stranded RNA. Nature. 431:343–349. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Ebert MS, Neilson JR and Sharp PA: MicroRNA sponges: Competitive inhibitors of small RNAs in mammalian cells. Nat Methods. 4:721–726. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Xu P, Vernooy SY, Guo M and Hay BA: The Drosophila microRNA Mir-14 suppresses cell death and is required for normal fat metabolism. Curr Biol. 13:790–795. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Kwon C, Han Z, Olson EN and Srivastava D: MicroRNA1 influences cardiac differentiation in Drosophila and regulates Notch signaling. Proc Natl Acad Sci USA. 102:18986–18991. 2005. View Article : Google Scholar

21 

Katz MG, Fargnoli AS, Kendle AP, Hajjar RJ and Bridges CR: The role of microRNAs in cardiac development and regenerative capacity. Am J Physiol Heart Circ Physiol. 310:H528–H541. 2016. View Article : Google Scholar :

22 

Cai W, Zhang J and Yang J, Fan Z, Liu X, Gao W, Zeng P, Xiong M, Ma C and Yang J: MicroRNA-24 attenuates vascular remodeling in diabetic rats through PI3K/Akt signaling pathway. Nutr Metab Cardiovasc Dis. 29:621–632. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Zhang J, Niu J, Tian B and Zhao M: MicroRNA-193b protects against myocardial ischemia-reperfusion injury in mouse by targeting mastermind-like 1. J Cell Biochem. 120:14088–14094. 2019. View Article : Google Scholar

24 

Wei Z, Qiao S, Zhao J, Liu Y, Li Q, Wei Z, Dai Q, Kang L and Xu B: MiRNA-181a over-expression in mesenchymal stem cell-derived exosomes influenced inflammatory response after myocardial ischemia-reperfusion injury. Life Sci. 232:1166322019. View Article : Google Scholar : PubMed/NCBI

25 

Liu Y, Qian XM, He QC and Weng JK: MiR-421 inhibition protects H9c2 cells against hypoxia/reoxygenation-induced oxidative stress and apoptosis by targeting Sirt3. Perfusion. 35:255–262. 2020. View Article : Google Scholar

26 

Wang S, Cheng Z, Chen X and Xue H: MicroRNA-135a protects against myocardial ischemia-reperfusion injury in rats by targeting protein tyrosine phosphatase 1B. J Cell Biochem. 120:10421–10433. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Liu Y, Zou J, Liu X and Zhang Q: MicroRNA-138 attenuates myocardial ischemia reperfusion injury through inhibiting mitochondria-mediated apoptosis by targeting HIF1-α. Exp Ther Med. 18:3325–3332. 2019.PubMed/NCBI

28 

Wan X, Yao B, Ma Y, Liu Y, Tang Y, Hu J, Li M, Fu S, Zheng X and Yin D: MicroRNA-128-1-5p attenuates myocardial ischemia/reperfusion injury by suppressing Gadd45g-mediated apoptotic signaling. Biochem Biophys Res Commun. 530:314–321. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Chen ZX, He D, Mo QW, Xie LP, Liang JR, Liu L and Fu WJ: MiR-129-5p protects against myocardial ischemia-reperfusion injury via targeting HMGB1. Eur Rev Med Pharmacol Sci. 24:4440–4450. 2020.PubMed/NCBI

30 

Yang S, Li H and Chen L: MicroRNA-140 attenuates myocardial ischemia-reperfusion injury through suppressing mitochondria-mediated apoptosis by targeting YES1. J Cell Biochem. 120:3813–3821. 2019. View Article : Google Scholar

31 

Liu Z, Tao B, Fan S, Pu Y, Xia H and Xu L: MicroRNA-145 protects against myocardial ischemia reperfusion injury via CaMKII-Mediated antiapoptotic and anti-inflammatory pathways. Oxid Med Cell Longev. 2019:89486572019. View Article : Google Scholar : PubMed/NCBI

32 

Niu S, Xu L, Yuan Y, Yang S, Ning H, Qin X, Xin P, Yuan D, Jiao J and Zhao Y: Effect of down-regulated miR-15b-5p expression on arrhythmia and myocardial apoptosis after myocardial ischemia reperfusion injury in mice. Biochem Biophys Res Commun. 530:54–59. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Chen Y, Wu S, Zhang XS, Wang DM and Qian CY: MicroRNA-489 promotes cardiomyocyte apoptosis induced by myocardial ischemia-reperfusion injury through inhibiting SPIN1. Eur Rev Med Pharmacol Sci. 23:6683–6690. 2019.PubMed/NCBI

34 

Li Q and Yang J, Zhang J, Liu XW, Yang CJ, Fan ZX, Wang HB, Yang Y, Zheng T and Yang J: Inhibition of microRNA-327 ameliorates ischemia/reperfusion injury-induced cardiomyocytes apoptosis through targeting apoptosis repressor with caspase recruitment domain. J Cell Physiol. 235:3753–3767. 2020. View Article : Google Scholar

35 

Zhang H, Wang J, Du A and Li Y: MiR-483-3p inhibition ameliorates myocardial ischemia/reperfusion injury by targeting the MDM4/p53 pathway. Mol Immunol. 125:9–14. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Wu HY, Wu JL and Ni ZL: Overexpression of microRNA-202-3p protects against myocardial ischemia-reperfusion injury through activation of TGF-β1/Smads signaling pathway by targeting TRPM6. Cell Cycle. 18:621–637. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Gwanyanya A, Amuzescu B, Zakharov SI, Macianskiene R, Sipido KR, Bolotina VM, Vereecke J and Mubagwa K: Magnesium-inhibited, TRPM6/7-like channel in cardiac myocytes: Permeation of divalent cations and pH-mediated regulation. J Physiol. 559:761–776. 2004. View Article : Google Scholar : PubMed/NCBI

38 

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

39 

Yang Y, Yang J, Liu XW, Ding JW, Li S, Guo X, Yang CJ, Fan ZX, Wang HB, Li Q, et al: Down-regulation of miR-327 alleviates ischemia/reperfusion-induced myocardial damage by targeting RP105. Cell Physiol Biochem. 49:1049–1063. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Zhao C, Jiang J, Wang YL and Wu YQ: Overexpression of microRNA-590-3p promotes the proliferation of and inhibits the apoptosis of myocardial cells through inhibition of the NF-κB signaling pathway by binding to RIPK1. J Cell Biochem. 120:3559–3573. 2019. View Article : Google Scholar

41 

Ding S, Liu D, Wang L, Wang G and Zhu Y: Inhibiting microRNA-29a protects myocardial ischemia-reperfusion injury by targeting SIRT1 and suppressing oxidative stress and NLRP3-mediated pyroptosis pathway. J Pharmacol Exp Ther. 372:128–135. 2020. View Article : Google Scholar

42 

Liu ZY, Pan HW, Cao Y, Zheng J, Zhang Y, Tang Y, He J, Hu YJ, Wang CL, Zou QC, et al: Downregulated microRNA-330 suppresses left ventricular remodeling via the TGF-β1/Smad3 signaling pathway by targeting SRY in mice with myocardial ischemia-reperfusion injury. J Cell Physiol. 234:11440–11450. 2019. View Article : Google Scholar

43 

Wu J, Liang J, Li M, Lin M, Mai L, Huang X, Liang J, Hu Y and Huang Y: Modulation of miRNAs by vitamin C in H2O2-exposed human umbilical vein endothelial cells. Int J Mol Med. 46:2150–2160. 2020. View Article : Google Scholar :

44 

Zhang Y, Du W and Yang B: Long non-coding RNAs as new regulators of cardiac electrophysiology and arrhythmias: Molecular mechanisms, therapeutic implications and challenges. Pharmacol Ther. 203:1073892019. View Article : Google Scholar : PubMed/NCBI

45 

Cabili MN, Dunagin MC, McClanahan PD, Biaesch A, Padovan-Merhar O, Regev A, Rinn JL and Raj A: Localization and abundance analysis of human lncRNAs at single-cell and single-molecule resolution. Genome Biol. 16:202015. View Article : Google Scholar :

46 

Kuo CC, Hanzelmann S, Sentürk Cetin N, Frank S, Zajzon B, Derks JP, Akhade VS, Ahuja G, Kanduri C, Grummt I, et al: Detection of RNA-DNA binding sites in long noncoding RNAs. Nucleic Acids Res. 47:e322019. View Article : Google Scholar : PubMed/NCBI

47 

Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, et al: Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 458:223–227. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al: The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 47:199–208. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Tong G, Wang Y, Xu C, Xu Y, Ye X, Zhou L, Zhu G, Zhou Z and Huang J: Long non-coding RNA FOXD3-AS1 aggravates ischemia/reperfusion injury of cardiomyocytes through promoting autophagy. Am J Transl Res. 11:5634–5644. 2019.PubMed/NCBI

50 

Zhang SB, Liu TJ, Pu GH, Li BY, Gao XZ and Han XL: Suppression of Long Non-Coding RNA LINC00652 restores sevoflurane-induced cardioprotection against myocardial ischemia-reperfusion injury by targeting GLP-1R through the cAMP/PKA pathway in mice. Cell Physiol Biochem. 49:1476–1491. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Fujita H, Morii T, Fujishima H, Sato T, Shimizu T, Hosoba M, Tsukiyama K, Narita T, Takahashi T, Drucker DJ, et al: The protective roles of GLP-1R signaling in diabetic nephropathy: Possible mechanism and therapeutic potential. Kidney Int. 85:579–589. 2014. View Article : Google Scholar

52 

Zhang H, Liu Y, Guan S, Qu D, Wang L, Wang X, Li X, Zhou S, Zhou Y, Wang N, et al: An orally active allosteric GLP-1 receptor agonist is neuroprotective in cellular and rodent models of stroke. PLoS One. 11:e01488272016. View Article : Google Scholar : PubMed/NCBI

53 

van Goor H, van den Born JC, Hillebrands JL and Joles JA: Hydrogen sulfide in hypertension. Curr Opin Nephrol Hypertens. 25:107–113. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Shen Y, Shen Z, Miao L, Xin X, Lin S, Zhu Y, Guo W and Zhu YZ: MiRNA-30 family inhibition protects against cardiac ischemic injury by regulating cystathionine-Ƴ-lyase expression. Antioxid Redox Signal. 22:224–240. 2015. View Article : Google Scholar :

55 

Hu X, Liu B, Wu P, Lang Y and Li T: LncRNA Oprm1 overexpression attenuates myocardial ischemia/reperfusion injury by increasing endogenous hydrogen sulfide via Oprm1/miR-30b-5p/CSE axis. Life Sci. 254:1176992020. View Article : Google Scholar : PubMed/NCBI

56 

Tay Y, Rinn J and Pandolfi PP: The multilayered complexity of ceRNA crosstalk and competition. Nature. 505:344–352. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Zhu Z and Zhao C: LncRNA AK 139128 promotes cardiomyocyte autophagy and apoptosis in myocardial hypoxia-reoxygenation injury. Life Sci. 1167052019.Epub ahead of print. View Article : Google Scholar

58 

Li X, Luo S, Zhang J, Yuan Y, Jiang W, Zhu H, Ding X, Zhan L, Wu H, Xie Y, et al: lncRNA H19 alleviated myocardial I/RI via suppressing miR-877-3p/Bcl-2-mediated mitochondrial apoptosis. Mol Ther Nucleic Acids. 17:297–309. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Su Q, Liu Y, Lv XW, Ye ZL, Sun YH, Kong BH and Qin ZB: Inhibition of lncRNA TUG1 upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion via targeting HMGB1- and Rac1-induced autophagy. J Mol Cell Cardiol. 133:12–25. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Ren L, Chen S, Liu W, Hou P, Sun W and Yan H: Downregulation of long non-coding RNA nuclear enriched abundant transcript 1 promotes cell proliferation and inhibits cell apoptosis by targeting miR-193a in myocardial ischemia/reperfusion injury. BMC Cardiovasc Disord. 19:1922019. View Article : Google Scholar

61 

Wang JJ, Bie ZD and Sun CF: Long noncoding RNA AK088388 regulates autophagy through miR-30a to affect cardiomyocyte injury. J Cell Biochem. 120:10155–10163. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Zou L, Ma X, Lin S, Wu B, Chen Y and Peng C: Long noncoding RNA-MEG3 contributes to myocardial ischemia-reperfusion injury through suppression of miR-7-5p expression. Biosci Rep. 39:BSR201902102019. View Article : Google Scholar

63 

Rong J, Pan H, He J, Zhang Y, Hu Y, Wang C, Fu Q, Fan W, Zou Q, Zhang L, et al: Long non-coding RNA KCNQ1OT1/microRNA-204-5p/LGALS3 axis regulates myocardial ischemia/reperfusion injury in mice. Cell Signal. 66:1094412020. View Article : Google Scholar

64 

Xue X and Luo L: LncRNA HIF1A-AS1 contributes to ventricular remodeling after myocardial ischemia/reperfusion injury by adsorption of microRNA-204 to regulating SOCS2 expression. Cell Cycle. 18:2465–2480. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Han Y, Wu N, Xia F, Liu S and Jia D: Long non-coding RNA GAS5 regulates myocardial ischemia-reperfusion injury through the PI3K/AKT apoptosis pathway by sponging miR-532-5p. Int J Mol Med. 45:858–872. 2020.

66 

Chen F, Zhang L, Wang E, Zhang C and Li X: LncRNA GAS5 regulates ischemic stroke as a competing endogenous RNA for miR-137 to regulate the Notch1 signaling pathway. Biochem Biophys Res Commun. 496:184–190. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Wang J, Xu R, Wu J and Li Z: MicroRNA-137 negatively regulates H2O2-induced cardiomyocyte apoptosis through CDC42. Med Sci Monit. 21:3498–3504. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Wang K, Liu F, Liu CY, An T, Zhang J, Zhou LY, Wang M, Dong YH, Li N, Gao JN, et al: The long noncoding RNA NRF regulates programmed necrosis and myocardial injury during ischemia and reperfusion by targeting miR-873. Cell Death Differ. 23:1394–1405. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M and Chan FK: Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell. 137:1112–1123. 2009. View Article : Google Scholar : PubMed/NCBI

70 

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

71 

Liang H, Li F, Li H, Wang R and Du M: Overexpression of lncRNA HULC attenuates myocardial Ischemia/reperfusion injury in rat models and apoptosis of Hypoxia/reoxygenation cardiomyocytes via targeting miR-377-5p through NLRP3/Caspase1/IL1β signaling pathway inhibition. Immunol Invest. Jul 17–2020.Epub ahead of print. View Article : Google Scholar

72 

Zhao G, Hailati J, Ma X, Bao Z, Bakeyi M and Liu Z: LncRNA Gm4419 regulates myocardial ischemia/reperfusion injury through targeting the miR-682/TRAF3 Axis. J Cardiovasc Pharmacol. 76:305–312. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Wang JX, Zhang XJ, Li Q, Wang K, Wang Y, Jiao JQ, Feng C, Teng S, Zhou LY, Gong Y, et al: MicroRNA-103/107 regulate programmed necrosis and myocardial ischemia/reperfusion injury through targeting FADD. Circ Res. 117:352–363. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Zhang W, Li Y and Wang P: Long non-coding RNA-ROR aggravates myocardial ischemia/reperfusion injury. Braz J Med Biol Res. 51:e65552018. View Article : Google Scholar : PubMed/NCBI

75 

Huang L, Guo B, Liu S, Miao C and Li Y: Inhibition of the LncRNA Gpr19 attenuates ischemia-reperfusion injury after acute myocardial infarction by inhibiting apoptosis and oxidative stress via the miR-324-5p/Mtfr1 axis. IUBMB Life. 72:373–383. 2020. View Article : Google Scholar

76 

Zhang M, Gu H, Xu W and Zhou X: Down-regulation of lncRNA MALAT1 reduces cardiomyocyte apoptosis and improves left ventricular function in diabetic rats. Int J Cardiol. 203:214–216. 2016. View Article : Google Scholar

77 

Wang S, Yu W, Chen J, Yao T and Deng F: LncRNA MALAT1 sponges miR-203 to promote inflammation in myocardial ischemia-reperfusion injury. Int J Cardiol. 268:2452018. View Article : Google Scholar : PubMed/NCBI

78 

Gong X, Zhu Y, Chang H, Li Y and Ma F: Long noncoding RNA MALAT1 promotes cardiomyocyte apoptosis after myocardial infarction via targeting miR-144-3p. Biosci Rep. 39:BSR201911032019. View Article : Google Scholar : PubMed/NCBI

79 

Li Z, Li J and Tang N: Long noncoding RNA Malat1 is a potent autophagy inducer protecting brain microvascular endothelial cells against oxygen-glucose deprivation/reoxygenation-induced injury by sponging miR-26b and upregulating ULK2 expression. Neuroscience. 354:1–10. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Ge ZW, Zhu XL, Wang BC, Hu JL, Sun JJ, Wang S, Chen XJ, Meng SP, Liu L and Cheng ZY: MicroRNA-26b relieves inflammatory response and myocardial remodeling of mice with myocardial infarction by suppression of MAPK pathway through binding to PTGS2. Int J Cardiol. 280:152–159. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al: Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 495:333–338. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF and Sharpless NE: Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA. 19:141–157. 2013. View Article : Google Scholar :

83 

Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan M, Evantal N, Memczak S, Rajewsky N and Kadener S: CircRNA biogenesis competes with pre-mRNA splicing. Mol Cell. 56:55–66. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Huang A, Zheng H, Wu Z, Chen M and Huang Y: Circular RNA-protein interactions: Functions, mechanisms, and identification. Theranostics. 10:3503–3517. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, Fatica A, Santini T, Andronache A, Wade M, et al: Circ-ZNF609 is a circular RNA that can be translated and functions in Myogenesis. Mol Cell. 66:22–37. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Werfel S, Nothjunge S, Schwarzmayr T, Strom TM, Meitinger T and Engelhardt S: Characterization of circular RNAs in human, mouse and rat hearts. J Mol Cell Cardiol. 98:103–107. 2016. View Article : Google Scholar : PubMed/NCBI

87 

Tan WL, Lim BT, Anene-Nzelu CG, Ackers-Johnson M, Dashi A, See K, Tiang Z, Lee DP, Chua WW, Luu TD, et al: A landscape of circular RNA expression in the human heart. Cardiovasc Res. 113:298–309. 2017.PubMed/NCBI

88 

Li M, Ding W, Tariq MA, Chang W, Zhang X, Xu W, Hou L, Wang Y and Wang J: A circular transcript of ncx1 gene mediates ischemic myocardial injury by targeting miR-133a-3p. Theranostics. 8:5855–5869. 2018. View Article : Google Scholar

89 

Zong L and Wang W: CircANXA2 promotes myocardial apoptosis in myocardial ischemia-reperfusion injury via inhibiting miRNA-133 expression. Biomed Res Int. 2020:85908612020. View Article : Google Scholar : PubMed/NCBI

90 

Song YF, Zhao L, Wang BC, Sun JJ, Hu JL, Zhu XL, Zhao J, Zheng DK and Ge ZW: The circular RNA TLK1 exacerbates myocardial ischemia/reperfusion injury via targeting miR-214/RIPK1 through TNF signaling pathway. Free Radic Biol Med. 155:69–80. 2020. View Article : Google Scholar : PubMed/NCBI

91 

Cherra SR III, Dagda RK, Tandon A and Chu CT: Mitochondrial autophagy as a compensatory response to PINK1 deficiency. Autophagy. 5:1213–1214. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Clark IE, Dodson MW, Jiang C, Cao JH, Huh JR, Seol JH, Yoo SJ, Hay BA and Guo M: Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin. Nature. 441:1162–1166. 2006. View Article : Google Scholar : PubMed/NCBI

93 

Billia F, Hauck L, Konecny F, Rao V, Shen J and Mak TW: PTEN-inducible kinase 1 (PINK1)/Park6 is indispensable for normal heart function. Proc Natl Acad Sci USA. 108:9572–9577. 2011. View Article : Google Scholar : PubMed/NCBI

94 

Zhou LY, Zhai M, Huang Y, Xu S, An T, Wang YH, Zhang RC, Liu CY, Dong YH, Wang M, et al: The circular RNA ACR attenuates myocardial ischemia/reperfusion injury by suppressing autophagy via modulation of the Pink1/FAM65B pathway. Cell Death Differ. 26:1299–1315. 2019. View Article : Google Scholar

95 

Zweier JL and Talukder MA: The role of oxidants and free radicals in reperfusion injury. Cardiovasc Res. 70:181–190. 2006. View Article : Google Scholar : PubMed/NCBI

96 

Zhu T, Yao Q, Wang W, Yao H and Chao J: iNOS Induces vascular endothelial cell migration and apoptosis via autophagy in Ischemia/Reperfusion injury. Cell Physiol Biochem. 38:1575–1588. 2016. View Article : Google Scholar : PubMed/NCBI

97 

Chen L, Luo W, Zhang W, Chu H, Wang J, Dai X, Cheng Y, Zhu T and Chao J: circDLPAG4/HECTD1 mediates ischaemia/reperfusion injury in endothelial cells via ER stress. RNA Biol. 17:240–253. 2020. View Article : Google Scholar :

98 

Fang S, Guo H, Cheng Y, Zhou Z, Zhang W, Han B, Luo W, Wang J, Xie W and Chao J: circHECTD1 promotes the silica-induced pulmonary endothelial-mesenchymal transition via HECTD1. Cell Death Dis. 9:3962018. View Article : Google Scholar : PubMed/NCBI

99 

Liu M, Jia J, Wang X, Liu Y, Wang C and Fan R: Long non-coding RNA HOTAIR promotes cervical cancer progression through regulating BCL2 via targeting miR-143-3p. Cancer Biol Ther. 19:391–399. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Zhou HF, Xu LL, Xie B, Ding HG, Fang F and Fang Q: Hsa-circ-0068566 inhibited the development of myocardial ischemia reperfusion injury by regulating hsa-miR-6322/PARP2 signal pathway. Eur Rev Med Pharmacol Sci. 24:6980–6993. 2020.PubMed/NCBI

101 

Chang H, Li ZB, Wu JY and Zhang L: Circ-100338 induces angiogenesis after myocardial ischemia-reperfusion injury by sponging miR-200a-3p. Eur Rev Med Pharmacol Sci. 24:6323–6332. 2020.PubMed/NCBI

102 

Ge X, Meng Q, Zhuang R, Yuan D, Liu J, Lin F, Fan H and Zhou X: Circular RNA expression alterations in extracellular vesicles isolated from murine heart post ischemia/reperfusion injury. Int J Cardiol. 296:136–140. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993. View Article : Google Scholar : PubMed/NCBI

104 

Iwasaki YW, Siomi MC and Siomi H: PIWI-Interacting RNA: Its biogenesis and functions. Annu Rev Biochem. 84:405–433. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Vella S, Gallo A, Lo Nigro A, Galvagno D, Raffa GM, Pilato M and Conaldi PG: PIWI-interacting RNA (piRNA) signatures in human cardiac progenitor cells. Int J Biochem Cell Biol. 76:1–11. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Zheng S, Zheng H, Huang A, Mai L, Huang X, Hu Y and Huang Y: Piwi-interacting RNAs play a role in vitamin C-mediated effects on endothelial aging. Int J Med Sci. 17:946–952. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2021
Volume 47 Issue 4

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
Li Q, Li Z, Fan Z, Yang Y and Lu C: Involvement of non‑coding RNAs in the pathogenesis of myocardial ischemia/reperfusion injury (Review). Int J Mol Med 47: 42, 2021
APA
Li, Q., Li, Z., Fan, Z., Yang, Y., & Lu, C. (2021). Involvement of non‑coding RNAs in the pathogenesis of myocardial ischemia/reperfusion injury (Review). International Journal of Molecular Medicine, 47, 42. https://doi.org/10.3892/ijmm.2021.4875
MLA
Li, Q., Li, Z., Fan, Z., Yang, Y., Lu, C."Involvement of non‑coding RNAs in the pathogenesis of myocardial ischemia/reperfusion injury (Review)". International Journal of Molecular Medicine 47.4 (2021): 42.
Chicago
Li, Q., Li, Z., Fan, Z., Yang, Y., Lu, C."Involvement of non‑coding RNAs in the pathogenesis of myocardial ischemia/reperfusion injury (Review)". International Journal of Molecular Medicine 47, no. 4 (2021): 42. https://doi.org/10.3892/ijmm.2021.4875