Abstract
Cardiovascular diseases (CVDs) remain the world’s leading cause of death. Cardiomyocyte autophagy helps maintain normal metabolism and functioning of the heart. Importantly, mounting evidence has revealed that autophagy plays a dual role in CVD pathology. Under physiological conditions, moderate autophagy maintains cell metabolic balance by degrading and recycling damaged organelles and proteins, and it promotes myocardial survival, but excessive or insufficient autophagy is equally deleterious and contributes to disease progression. Noncoding RNAs (ncRNAs) are a class of RNAs transcribed from the genome, but most ncRNAs do not code for functional proteins. In recent years, increasingly, various ncRNAs have been identified, and they play important regulatory roles in the physiological and pathological processes of organisms, as well as in autophagy. Thus, determining whether ncRNA-regulated autophagy plays a protective role in CVDs or promotes their progression can help us to develop ncRNAs as therapeutic targets in autophagy-related CVDs. In this review, we briefly summarize the regulatory roles of several important ncRNAs, including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), in the autophagy of various CVDs to provide a theoretical basis for the etiology and pathogenesis of CVDs and develop novel therapies to treat CVDs.
Keywords: autophagy, miRNA, lncRNA, circRNA, cardiovascular diseases
Graphical Abstract

Cardiovascular diseases (CVDs) remain the world’s leading cause of death. Mounting evidence has revealed that ncRNAs and autophagy play critical roles in CVD pathology. Determining whether ncRNA-regulated autophagy plays a protective role in CVDs or promotes their progression can help us to develop ncRNAs as therapeutic targets in autophagy-related CVDs.
Main Text
Autophagy is a dynamic equilibrium process that removes damaged cell inclusions or organelles in a lysosomal-dependent way and plays a critical role in maintaining cellular homeostasis.1 Autophagy processes can be classified as chaperone-mediated autophagy, microautophagy, and macroautophagy depending on their physiological functions and delivery routes.2 Among these processes, macroautophagy is the most common and deeply studied process. In this review, “autophagy” refers to macroautophagy, and this process involves the following steps: (1) initiation: after receiving external stimuli, phagophores are formed in the cytoplasm; (2) elongation: vesicles extend to wrap damaged organelles; (3) closure: vesicles are sealed into double-membrane autophagosomes; (4) maturation: autophagosomes combine with lysosomes to form autolysosomes; and (5) degradation: cytoplasmic materials are degraded by lysosomal acid hydrolases, and the products are transported to the cytoplasm for reuse.3
Autophagy is highly conserved in evolution. More than 30 autophagy-related genes (ATGs) have been identified in yeast, many of which have mammalian orthologs. ATGs regulate the occurrence of autophagy in a coordinated manner. Among them, beclin-1 (the mammalian ortholog of yeast ATG6) was first discovered as an important autophagosome initiation regulator that forms a class III phosphatidylinositol 3-kinase (PI3K) complex with hVsp34, p150 (Vps15), and ATG14, while the level of LC3-II (ATG8-II) is directly proportional to the autophagosome number. Thus, beclin-1 and the LC3-II/LC3-I ratio are the most widely used autophagy markers.4 Activation of the autophagy machinery is regulated by two central modulators, mTOR (mammalian target of rapamycin) and AMPK (adenosine 5′-monophosphate [AMP]-activated protein kinase). mTOR can integrate upstream signals, such as PI3K/Akt and mitogen-activated protein kinase (MAPK)/Erk1/2 signaling, and function as a metabolic sensor to negatively regulate autophagy. AMPK can be activated at a high cellular AMP/ATP ratio, and it is an important mediator for maintaining cellular energy homeostasis. AMPK inhibits mTOR kinase activity by phosphorylating the tuberous sclerosis 1/2 (TSC1/2) complex and activates autophagy. Moreover, AMPK can phosphorylate Unc-51-like autophagy-activating kinase 1 (ULK1) to activate the class III PI3K complex.5, 6, 7
Cardiovascular diseases (CVDs) remain the world’s leading cause of death, as more people die from CVDs each year than from any other cause. Cardiomyocyte autophagy helps maintain normal metabolism and functioning of the heart. Importantly, an increasing body of evidence supports the dual role of autophagy in the pathogenesis of CVDs.8,9 Under physiological conditions, moderate autophagy helps maintain metabolic balance in cells through the degradation and recycling of damaged organelles and proteins, in addition to promoting myocardial survival. However, excessive or insufficient autophagy may lead to disease. Under pathological conditions, autophagy strives to ensure cell survival, but when it fails to exert its protective effect, autophagic cell death occurs. Noncoding RNAs (ncRNAs) are a class of RNAs transcribed from the genome, but most ncRNAs do not code for functional proteins. In recent years, a large number of diverse ncRNAs have been identified, and they play important regulatory roles in the physiological and pathological processes of organisms, as well as in autophagy.10, 11, 12 In this review, we briefly summarize the regulatory roles of several important ncRNAs, including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), in the autophagy of various CVDs to provide a theoretical basis for the etiology and pathogenesis of CVDs and to facilitate the development of novel therapies for the treatment of CVDs.
miRNAs and autophagy
miRNAs constitute a class of single-stranded ncRNA molecules with lengths of 19–24 nt, and they negatively regulate gene expression at the post-translational level by completely or partially base pairing with the 3′ UTR region of the target gene, which leads to the cleavage/degradation of mRNA transcripts or protein translation repression (Figure 1). Subsequent studies have shown that a few miRNAs can also bind to the 5′ UTR or even the coding regions to exert their functions.13,14 Typically, each miRNA can have multiple target genes, and, conversely, several miRNAs can regulate the same gene. This complex regulatory network not only can regulate the expression of multiple genes through one miRNA, but it also can finely regulate the expression of a gene through the combination of several miRNAs. Recent studies have found that miRNAs participate in the regulation of autophagy by regulating the expression of ATGs and play an important role in the development of organisms and the occurrence of CVDs. Significantly, it has been found that autophagy not only has different characteristics in different physiological states but also has different effects on cardiomyocytes in different states. The first reported autophagy-related miRNA contributing to cardiac pathologies was miR-204, which plays a role during myocardial ischemia/reperfusion (I/R) injury.15 Since then, many dysregulated autophagic miRNAs (see Table 1 and Figure 1 for a complete list) have been implicated in various CVDs.
Figure 1.
Regulatory roles of miRNAs in cardiac autophagy and related CVDs
miRNAs negatively regulate gene expression by completely or partially base pairing with the 3′ UTR region of the target gene, leading to the cleavage/degradation of mRNA transcripts or protein translation repression. See text and Table 1 for detailed molecular mechanism explanations. Pink oval represents anti-autophagic miRNAs with protective effects in CVDs; yellow oval represents anti-autophagic miRNAs with deleterious effects; purple oval represents pro-autophagic miRNAs with protective effects in CVDs; green oval represents pro-autophagic miRNAs with deleterious effects in CVDs.
Table 1.
Dysregulated miRNAs and their roles in autophagy during the progression of CVDs
| miRNAs | Roles in autophagy | Target genes/signaling pathways | Outcomes | Refs. |
|---|---|---|---|---|
| Myocardial infarction/injury | ||||
| miR-20b-5p | anti- | ULK1 | ↓ H/R-induced injury | 16 |
| miR-21 | anti- | Akt/mTOR | ↓ myocardial apoptosis | 17 |
| miR-27a-5p | anti- | Atg7 | ↓ hypoxia-induced injury | 18 |
| miR-34a | anti- | TNF-α | ↓ myocardial I/R injury | 19 |
| miR-101 | anti- | DDIT4 | ↓ myocardial I/R injury | 20 |
| miR-103a-3p | anti- | Atg5 | ↓ H/R-induced apoptosis | 21 |
| miR-129-5p | anti- | ATG14 | ↓ H2O2-induced apoptosis | 22 |
| miR-188-3p | anti- | ATG7 | ↓ myocardial I/R injury | 23 |
| miR-204 | anti- | LC3-II | ↓ myocardial I/R injury | 15 |
| anti- | SIRT1 | ↓ H/R-induced injury | 24 | |
| miR-206/216b | anti- | Atg13 | ↓ myocardial I/R injury | 25 |
| miR-223 | anti- | PARP-1 | ↓ H/R-induced injury | 26 |
| miR-638 | anti- | ATG5 | ↓ myocardial apoptosis | 27 |
| miR-30a | anti- | Beclin1 | ↑ myocardial injury | 28 |
| miR-101 | anti- | RAB5A | ↑ myocardial apoptosis | 29 |
| miR-122 | anti- | PTEN/PI3K/AKT | ↑ myocardial apoptosis | 30 |
| miR-153 | anti- | Mcl-1 | ↑ myocardial apoptosis | 31 |
| miR-181b-5p | anti- | Hspa5 | ↑ myocardial apoptosis | 32 |
| miR-429 | anti- | MO25/LKB1/AMPK | ↑ myocardial I/R injury | 33 |
| miR-143 | anti- | Atg7 | ↓ CPC survival | 34 |
| miR-1 | pro- | PIK3CA | ↓ myocardial injury | 35 |
| miR-22 | pro- | p38α | ↓ myocardial apoptosis | 36 |
| miR-145 | pro- | Akt3/mTOR signaling | ↓ myocardial apoptosis | 37 |
| pro- | Angpt2 | ↓ myocardial I/R injury | 38 | |
| pro- | FRS2 | ↓ myocardial I/R injury | 39 | |
| miR-144 | pro- | mTOR | ↓ myocardial I/R injury | 40 |
| miR-99a | pro- | mTOR/P70/S6K | ↑ cardiac function | 41 |
| miR-325 | pro- | ARC | ↑ myocardial I/R injury | 42 |
| Cardiac hypertrophy | ||||
| miR-30a | anti- | Beclin1 | ↓ myocardial hypertrophy | 43 |
| miR-34a | anti- | ATG9A | ↓ myocardial hypertrophy | 44 |
| miR-181a | anti- | ATG5 | ↓ myocardial hypertrophy | 45 |
| miR-29a | anti- | PTEN/AKT/mTOR | ↑ cardiac hypertrophy | 46 |
| miR-199a | anti- | GSK3β/mTOR signaling | ↑ cardiac hypertrophy | 47 |
| miR-212/132 | anti- | FoxO3/calcineurin signaling | ↑ cardiac hypertrophy | 48 |
| miR302-367 | anti- | PTEN/PI3K/AKT/mTORC1 | ↑ cardiac hypertrophy | 49 |
| miR-365 | anti- | Skp2 | ↑ cardiac hypertrophy | 50 |
| miR-208-3p | pro- | PDCD4 | ↑ myocardial hypertrophy | 51 |
| Cardiac fibrosis | ||||
| miR-19a/b-3p | anti- | TGF-βRII | ↓ cardiac fibrosis | 52 |
| miR-26a-5p | anti- | ULK1 | ↓ cardiac fibrosis | 53 |
| miR-200b | anti- | – | ↓ cardiac fibrosis | 54 |
| Heart failure | ||||
| miR-29-3p | anti- | SPARC | ↓ cardiomyocytes apoptosis | 55 |
| miR-30e | anti- | Beclin1 | ↓ heart failure | 56 |
| miR-183-3p | anti- | BNIP3L | ↓ Chronic systolic heart failure | 57 |
| miR-221 | anti- | p27/CDK2/Mtor axis | ↑ heart failure | 58 |
| miR-222 | anti- | p27 | ↑ heart failure | 59 |
| Cardiomyopathy | ||||
| miR-451 | anti- | TSC1 | ↓ hypertrophic cardiomyopathy | 60 |
| miR-30c | anti- | BECN1 | ↓ diabetic cardiomyopathy | 61 |
| miR-371a-5p | anti- | BAG3 | ↓ takotsubo cardiomyopathy | 62 |
| miR-34a | anti- | – | ↑ diabetic cardiomyopathy | 63 |
| Atherosclerosis | ||||
| miR-33 | anti- | Abca1/Atg5/Lamp1/Prkaa1 | ↑ atherosclerosis | 64 |
| miR-129-5p | anti- | Beclin1 | ↑ atherosclerosis | 65 |
| miR-155 | anti- | ATG5 | ↑ oxidant-induced injury | 66 |
| miR-216a | anti- | Beclin1 | ↑ atherosclerosis | 67 |
| Sepsis cardiac dysfunction | ||||
| miR-21-3p | pro- | SORBS2 | ↑ cardiac dysfunction | 68 |
miRNAs, autophagy, myocardial infarction (MI), and myocardial I/R injury
Autophagy plays a key role in maintaining energy production and myocardial viability during myocardial ischemia.69 Reperfusion therapies, including thrombolysis, interventional treatment, and coronary artery bypass grafting can benefit patients with MI by restoring blood flow. However, subsequent I/R injury increases the risk of cardiac dysfunction, heart failure (HF), and death. In the early stage of myocardial ischemia, starvation occurs owing to the lack of cellular nutrients. In this stage, autophagy recovers amino acids to synthesize the proteins necessary for survival and degrades damaged organelles, thereby reducing myocardial damage and apoptosis and maintaining cardiac function.32 During reperfusion, because of the activation of the above autophagy pathways, increased levels of reactive oxygen species (ROS) induce mitochondrial damage, which in turn can promote autophagy. This vicious cycle leads to excessive activation of autophagy and accelerates damage to cardiomyocytes and enlargement of infarct size.22
Anti-autophagic miRNAs with beneficial impacts
Various miRNAs have been found to exert a protective effect against myocardial injury by inhibiting excessive autophagy. It has been reported that miR-34a and miR-101 can attenuate myocardial I/R injury through suppressing autophagy by targeting tumor necrosis factor α (TNF-α) and DNA damage-inducible transcript 4 (DDIT4), respectively.19,20 mTOR plays a vital role in autophagy regulation, in that it negatively regulates autophagy by inhibiting ULK1, and the mTOR-dependent pathway is regulated by PI3K/Akt and MAPK/Erk1/2 signaling. Overexpression of miR-21 activates the Akt/mTOR pathway, inhibits the autophagic activity, and decreases myocardial apoptosis, thus protecting H9c2 cells against hypoxia/reoxygenation (H/R) injury.17 In human umbilical vein endothelial cells (HUVECs) with H/R injury, propofol treatment inhibits autophagic cell death through the upregulation of miR-20b-5p and downregulation of ULK1. In contrast, in propofol-preconditioned HUVECs with H/R injury, ULK1 overexpression promotes cell autophagy and apoptosis and restrains the protective effect of miR-20b-5p.16 In neonatal rat cardiomyocytes and H9c2 cells, it is thought that miR-223 represses hypoxia-induced apoptosis and overactivated autophagy through the Akt/mTOR pathway by targeting PARP-1.26 ATG proteins are essential for autophagic flux. ATG13 is vital for the activation and stability of ULK1 together with the scaffold protein FIP200, and this complex is important for membrane nucleation. Ding et al.25 reported that histamine increases miR-206 and miR-216b expression, and these molecules work in combination to downregulate ATG13, leading to the reduction of autophagy. Moreover, they demonstrated that ATG13 can interact with Fas-associated protein with death domain (FADD) to activate the caspase-8 apoptosis pathway. Thus, the miR-206-216b/ATG13 axis mediates the prohibitive role of histamine in MI-induced autophagy activation and myocardial apoptosis.25 ATG14 is required for the membrane targeting of the PI3KC3-C1 complex and the induction of autophagy. Zhang et al.22 revealed that ATG14 is a direct target of miR-129-5p, and ATG14 inhibition due to miR-129-5p overexpression suppresses H2O2-induced autophagy and apoptosis in H9c2 cells through the activation of the PI3K/Akt/mTOR pathway. Among the 41 ATG proteins, ATG5 and ATG7 are indispensable for autophagic vesicle formation.70 miR-103-3p and miR-638 both suppress cardiomyocyte autophagy and apoptosis by targeting ATG5.21,27 miR-27a-5p and miR-188-3p attenuate hypoxia- or A/R-induced cardiomyocyte injury by inhibiting ATG7.18,23 Furthermore, SIRT1 and LC3-II have been recognized as potential targets of miR-204 to mediate the cardioprotective role of miR-204 on I/R-induced cardiomyocytes apoptosis through the repression of autophagy.15,24
Anti-autophagic miRNAs with deleterious impacts
A few miRNAs contribute to myocardial I/R injury through the suppression of autophagy initiation and autophagic flux. Beclin-1, the mammalian homolog of yeast Atg6, was the first identified mammalian autophagy protein that promotes autophagosome formation. miR-30a is highly enriched in exosomes in acute MI patients.28 Either inhibition of exosome release or miR-30a increases the levels of beclin-1, Atg12, as well as the LC3-II/LC3-I ratio, thus maintaining the autophagic response and attenuated apoptosis in cardiomyocytes after hypoxia.28 In hypoxic H9c2 cells, miR-122 is significantly upregulated, and overexpression of miR-122 aggravates the effects of hypoxia on increased cell apoptosis through inhibition of the PTEN/PI3K/Akt pathway.30 Hspa5 is an endoplasmic reticulum (ER) stress chaperone synchronously with LC3-II and is cardioprotective.32 miR-181b-5p contributes to starvation-induced cardiomyocyte autophagy and apoptosis by directly targeting Hspa5 through the PI3K/Akt/mTOR pathway.32 In addition, the inhibition of miR-429 by antagomir transfection enhances autophagy and suppresses apoptosis through the MO25/LKB1/AMPK signaling pathway, whereas the overexpression of miR-429 by mimic transfection has the opposite effects.33 miR-101 can target Ras-related protein Rab-5A (RAB5A), a small GTPase that can interact with hVPS34 and beclin-1 to induce autophagosome formation, while the inhibition of miR-101 ameliorates myocardial ischemic injury though the induction of cardiomyocyte autophagy.29 Oxidative stress can activate autophagy to clear damaged cells, and such autophagy may delay cell death and maintain cellular homeostasis. Zou et al.31 reported that miR-153 suppresses cardiomyocyte autophagy and reduces cell survival upon oxidative stimuli by attenuating Mcl-1 expression. Moreover, they found that the silencing of miR-143 upregulates Atg7 expression and promotes the autophagy of c-kit+ cardiac progenitor cells (CPCs) exposed to H2O2.34 Moreover, the transplantation of c-kit+ CPCs with miR-143 inhibitor improves cell survival and enhanced therapeutic efficiency in MI.34
Pro-autophagic miRNAs with beneficial impacts
Many miRNAs activate cardiomyocyte autophagy through upregulation of ATGs, such as beclin-1, and LC3, and by promoting the formation of autophagosomes, thus improving the cardiac function. For instance, miR-99a overexpression prevents cardiomyocyte apoptosis and enhances autophagy through the mTOR/P70/S6K signaling pathway, thus exhibiting a protective role against MI and ameliorating the subsequent cardiac remodeling.41 Similarly, miR-22 can protect cardiomyocytes from starvation-induced injury through the promotion of autophagy and inhibition of apoptosis by targeting p38α.36 The upregulation of miR-1 upon paeonol treatment suppresses the expression of PIK3CA, enhances autophagy, and mitigates the cardiac dysfunction induced by epirubicin.35 By targeting mTOR, miR-144 mediates the cardioprotective role of remote ischemic preconditioning in I/R injury.71 Intravenous administration of miR-145 could accelerate the transition of LC3-I to LC3-II and enhance cardiomyocyte autophagy to reduce myocardial I/R injury.39 In this study, fibroblast growth factor receptor substrate 2 (FRS2) was determined as a target of miR-145. In addition, angiopoietin-2 (Angpt2) and the Akt3/mTOR pathway also function as downstream targets of miR-145 to mediate its protective role in MI through the induction of autophagy.37,38
Pro-autophagic miRNAs with deleterious impacts
Moreover, miRNA can promote autophagy through the downregulation of anti-autophagic proteins, leading to increased MI size and even the development of cardiac failure. Bo et al.42 reported that the E2F1-miR-325-ARC axis play an important role in the regulation of autophagy during H/R and I/R. In stress-induced HF, ARC transgenic (Tg) mice exhibited reduced autophagy and MI area. E2F1 facilitates the expression of miR-325 to inhibit ARC. Consequently, ARC is unable to repress the autophagic program, thus enhancing autophagy and aggravating myocardial I/R injury.42
miRNAs, autophagy, and cardiac hypertrophy
Cardiac hypertrophy is a compensatory response to pressure overload, and it manifests as thickening of the left ventricular (LV) wall, enlarged cardiomyocyte size accompanied by cytoskeletal remodeling, and upregulated fetal gene expression. Sustained hypertrophic growth often causes HF and even sudden death, leading to a poor prognosis. An excessively activated autophagy level may be involved in the pathophysiological process of cardiac hypertrophy. Moreover, deficient autophagy has also been observed in the process.
Anti-autophagic miRNAs with beneficial impacts
Ang II is an important factor leading to hypertension and cardiac hypertrophy. Pan et al.43 found that Ang II induces excessive cardiomyocyte autophagy through the downregulation of miR-30 expression and upregulation of its target gene beclin-1, and this excessive autophagy mediates the development of cardiac hypertrophy. ATG9 is a key transmembrane autophagic protein involved in the regulation of autophagosome formation. Huang et al.44 reported that miR-34a can suppress ATG9A expression by directly binding to its 3′ UTR. miR-34a inhibition or ATG9A overexpression enhances the autophagic activity and aggravates Ang II-induced myocardial hypertrophy.44 Similarly, miR-181a mediates Ang II-induced myocardial hypertrophy by enhancing ATG5-stimulated autophagy.45
Pro-autophagic miRNAs with deleterious impacts
However, Wang et al.51 reported increased levels of miR-208a-3p in Ang-II induced H9c2 cardiomyocytes and aggravated autophagy through the inhibition of programmed cell death protein 4 (PDCD4) and upregulation of ATG5. Thus, miRNA (miR-208-3a) or its downregulation (miR-30a/miR-34a/miR-181a) can trigger the autophagy cascade in a direct or indirect manner, and the excessive degradation of organelles and proteins eventually leads to cardiac hypertrophy.
Anti-autophagic miRNAs with deleterious impacts
Both in transverse aortic constriction (TAC) surgery mice and Ang II-stimulated H9c2 cells, miR-29a has been found to be upregulated, and the inhibition of miR-29a can partially mitigate Ang II-induced hypertrophy.46 The overexpression of miR-29a promotes cardiac hypertrophy through the suppression of PTEN, thus activating the AKT/mTOR pathway and inhibiting autophagy.46 Interestingly, miR302-367 has been identified as another upstream regulator of PTEN and PI3K/AKT/mTORC1 signaling to inhibit autophagy and promote cardiac hypertrophy.49 Skp2 is sufficient to promote autophagy through mTORC1 inhibition, and miR-365 negatively modulates autophagy in hypertrophic cardiomyocytes by targeting Skp2.50 miR-199a Tg mice had significantly larger hearts and lower autophagy levels.47 When these mice were treated with rapamycin, an autophagy inducer, the autophagic activity was enhanced, and cardiac hypertrophy improved. Furthermore, when stimulated with the lysosomal inhibitor chloroquine, the LC3 ratio and the p62 of the Tg and control mice were upregulated, indicating that miR-199 did not inhibit lysosomal degradation but the formation of autophagic vesicles. Mechanically, miR-199 targets the pro-autophagic and antihypertrophic GSK3β to activate mTOR signaling and inhibit autophagy.47 The overexpression of GSK3β enhanced the autophagy level and attenuated the hypertrophic responses in the miR-199 Tg mice.47 Similarly, miR-212/132 directly targeted the anti-hypertrophic and pro-autophagic FoxO3 transcription factor to impair autophagic response and activate calcineurin/NFAT signaling, leading to pathological cardiac hypertrophy.48 In summary, the above miRNAs can inhibit autophagy, and lower levels of autophagy activity block their protective effect, thereby promoting the occurrence and development of cardiac hypertrophy.
miRNAs, autophagy, and cardiac fibrosis
Cardiac fibrosis refers to the proliferation of fibroblasts in the heart and their transformation into myofibroblasts, which leads to excessive deposition of collagen fibers in the extracellular matrix of the myocardium, accompanied by dysregulation of collagen types and disordered arrangement. The occurrence of various CVDs, such as atrial fibrillation (AF), ischemic cardiomyopathy, chronic HF, and hypertensive heart disease, is inseparable from myocardial fibrosis.
Anti-autophagic miRNAs with beneficial impacts
TGF-β1 plays an important role in fibrogenesis, which promotes COL1A2 and fibronectin synthesis and activates autophagy in human cardiac fibroblasts.52 Low expression levels of miR-19a-3p/19b-3p were observed in HF patients’ plasma. miR-19a-3p/19b-3p overexpression attenuates autophagy and ameliorates the fibrosis induced by TGF-β1. Further functional studies have demonstrated that miR-19a-3p/19b-3p suppresses autophagy-mediated fibrogenesis by targeting TGF-βRII mRNA.52 miR-26a-5p directly interferes with ULK1 and reduces the transformation of LC3-I to LC3-II, which simultaneously inhibits the collagen expression in cardiac fibroblasts.53 miR-200b is negatively regulated by DNA (cytosine-5-)-methyltransferase 3 alpha (DNMT3A) and represses cardiac fibroblast autophagy during cardiac fibrosis.54 Collectively, the above miRNAs can inhibit autophagy and exhibit a cardioprotective effect during fibrogenesis in the heart.
miRNAs, autophagy, and HF
HF is the end stage of many CVDs caused by ventricular filling or impaired ejection capacity due to abnormal heart structure and function, which seriously affects the quality of life and prognosis of patients.
Anti-autophagic miRNAs with beneficial impacts
The clinical application of doxorubicin for treating human tumor diseases is limited by its cardiotoxicity effect, which can cause cardiomyopathy and congestive HF. Angiotensin converting enzyme 2 (ACE2) can metabolize a variety of vasoactive peptide substrates for HF therapy.72,73 Lai et al.56 reported that exogenous administration of recombinant human ACE2 suppressed the harmful side effects of doxorubicin and improved LV function by upregulating miR-30e expression, inhibiting the expression of its target gene beclin-1 and reducing the LC3-II/LC3-I ratio. Therefore, ACE2 attenuates doxorubicin-induced cardiac dysfunction by preventing autophagy-induced cardiomyocyte apoptosis in a miR-30e/beclin-1 signaling pathway.56 Likewise, vagus nerve stimulation, an emerging method for chronic systolic HF (CSHF) treatment, improves the cardiac function in CSHF through the upregulation of miR-183-3p, which represses the BNIP3L-mediated autophagy.57 The cardioprotective miR-29b-3p targeting secreted protein acidic and rich in cysteine (SPARC) is downregulated in HF patients and hypoxia-induced H9c2 cells, and it attenuates the autophagy and apoptosis of cardiomyocytes by inhibiting TGF-β1/Smad3 activation.55
Anti-autophagic miRNAs with deleterious impacts
Conversely, both cardiac-specific overexpression of miR-221 and miR-222 provoke cardiac dysfunction and HF.58,59 Mechanically, direct suppression of the cyclin-dependent kinase inhibitor p27 by miR-221/222 leads to mTOR activation and autophagic flux impairment, resulting in pathological cardiac remodeling.58,59 Hence, the basal autophagy is extremely important for maintaining normal cardiac function and structure. The modulation of miR-221/222 levels may provide new targets and strategies for HF treatment.
miRNAs, autophagy, and cardiomyopathy
Cardiomyopathies are a group of heterogeneous heart diseases that lead to abnormal mechanical and electrical activities of the heart owing to different pathogenies, including genetic, infection, metabolism, and endocrine, and they manifest as improper ventricular hypertrophy or dilation.74 Severe cardiomyopathy can cause cardiovascular death or progressive HF.
Anti-autophagic miRNAs with beneficial impacts
Hypertrophic cardiomyopathy (HCM), a common inherited disease caused by mutations in the MYH7 and MYBPC3 genes, is defined by unexplained myocardial hypertrophy and a high risk of sudden cardiac death. In the hearts of HCM, miR-451 is downregulated, and the level of autophagy is increased.60 Forced expression of miR-451 in vitro significantly inhibits the formation of autophagosomes and decreases the surface area of cardiomyocytes. miR-451 can directly bind to the 3′ UTR of TSC1, which is a negative regulator upstream of mTORC1, suggesting that TSC1 may be a potential target for miR-451 to regulate autophagy in HCM.60
Diabetic cardiomyopathy (DCM) is one of the common complications of diabetes, and it is characterized by myocardial hypertrophy and contractile dysfunction. Nandi et al.75 found that miR-133a was less expressed in the cardiomyocytes of patients with diabetic HF than in healthy cardiomyocytes. miR-133a can inhibit cardiomyocyte autophagy by targeting beclin-1 and LC3B. In addition, the mTOR activity in patients with diabetic HF was found to be significantly lower than that in healthy people. These findings demonstrate that the attenuation of miR-133a and mTOR inhibition may be the mechanisms of autophagy induction and hypertrophy in DCM.75 In diabetic (db/db) hearts, miR-30c expression is attenuated, and autophagy is induced.61 Overexpression of miR-30c results in reduced BECN1 levels and autophagy, thus improving cardiac function and structure. Similarly, miR-30 inhibition raises the levels of BECN1 and autophagy and aggravates cardiac abnormalities. Furthermore, SP1, an important transcription factor involved in the regulation of energy metabolism, has been revealed to be an upstream activator of miR-30c. Taken together, this study demonstrates that in db/db hearts, elevated free fatty acids downregulate SP1, leading to lower levels of transcribed miR-30c and, subsequently, increased levels of BECN1 and autophagic flux, which contribute to the pathological process of DCM.61
Takotsubo cardiomyopathy (TCM) is an acute heart syndrome first described by Sato et al. in 199076. It is characterized by transient reversible LV wall motion dysfunction but without coronary artery abnormality.77 This disease, also called broken heart syndrome, usually occurs in people with emotional changes or excessive stress. d’Avenia et al.62 found that excessive epinephrine induced ERK phosphorylation-dependent β-catenin nuclear translocation in cardiomyocytes, resulting in increased miR-371-5p expression. Although miRNA can suppress the target protein level by binding to the 3′ UTR of the target mRNA, a growing body of evidence has indicated that miRNA can also stimulate gene expression at the post-translational level through multiple mechanisms.62 As revealed in this study, miR-371a-5p binds to the 3′ UTR of Bcl2-asscociated athanogene 3 (BAG3), leading to increased BAG3 protein expression. BAG3 is a member of the BAG family of auxiliary chaperones that interact with ATPase domain of HSP70. BAG is strongly expressed in cardiomyocytes and participates in molecular chaperone-mediated autophagy. The absence of BAG in mice can cause fatal cardiomyopathy. TCM patients often exhibit the g2252c mutation in BAG3 3′ UTR, which results in the inability of miR-371a-5p to bind to the 3′ UTR of BAG3 and, thus, induces an altered response to epinephrine stimulation, which may be a new mechanism of TCM pathogenesis.62
Anti-autophagic miRNAs with deleterious impacts
In contrast, Ni et al.63 indicated that miR-34a was upregulated and autophagy was impaired in high-glucose-induced cardiomyocytes and in diabetic hearts. Dihydromyricetin can restore impaired autophagy by reducing miR-34a levels, thus decelerating the development of DCM.63
miRNAs, autophagy, and atherosclerosis (AS)
AS is a chronic inflammatory process characterized by lipid accumulation and inflammatory cell infiltration. The stability of atherosclerotic plaque is the main risk factor for coronary heart disease and stroke. Recent studies have found that autophagy plays an important role in regulating the formation of AS and the stabilization of atherosclerotic plaques.78,79 When stimulated by AS risk factors, such as oxidized low-density lipoprotein (ox-LDL) and ROS, endothelial cells (ECs) undergo a stress-repairing autophagic process, which can degrade the damaged organelles in the cell and protect ECs from inflammation and oxidative stress. However, defective autophagy can cause the apoptosis of ECs, resulting in a loss of endothelial integrity to facilitate local lipid deposition, plaque instability, and even acute coronary occlusion and sudden death.
Anti-autophagic miRNAs with deleterious impacts
miR-129-5p is increased both in ApoE−/− mice supplied with a high-fat diet and human aortic ECs treated with ox-LDL, which impairs the protective effects of EC autophagy against AS by suppressing the translation of beclin-1.65 miR-216a may be a regulator linker between endothelial dysfunction and autophagy, and it can induce ox-LDL aggregation and monocyte adhesion in HUVECs through suppression of the expression of beclin-1 and ATG5.67 In senescent HUVECs, miR-216a downregulation enhances the autophagic activity and reduces ox-LDL uptake and THP-1 adhesion, thereby exerting a protective anti-atherogenic role against ox-LDL treatment.67
In ox-LDL-exposed HUVECs, miR-155 expression is significantly increased, and inhibition of miR-155 inhibits autophagic activity. miR-155 can promote the ox-LDL-induced autophagy of HUVECs by enhancing LC3 levels and reducing p62 levels.80 Moreover, miR-155 directly targets FADD to inhibit the apoptosis of RAW264.7 macrophage cells and the formation of atherosclerotic plaque.81 However, in an H2O2-induced oxidative stress EC model, miR-155 downregulation promoted the proliferation of ECs and decreased intracellular ROS injury by modulating the expression of ATG5.66
Impaired cholesterol metabolism and defective efferocytosis contribute to AS. In macrophages, miR-33 targets key autophagic regulators to reduce the clearances of lipid droplets and apoptotic cells.64 Furthermore, miR-33 regulation of autophagy acts upstream of ATP-binding cassette transporter A1 (ABCA1) to control cholesterol efflux, whereas miR-33 inhibition restores defective autophagy and promotes efferocytosis, lysosomal biogenesis, and apoptotic degradation to reduce plaque necrosis in atherosclerotic Ldlr−/− mice.64 Collectively, these data reveal that miR-33 regulates cellular cholesterol homeostasis and AS through autophagy.
miRNAs, autophagy, and sepsis cardiac dysfunction
Sepsis is a systemic inflammatory response syndrome caused by infection, and its further development is among the main causes of death in intensive care units. Sepsis cardiac dysfunction is a severe complication of sepsis, and it is one of the main factors that determine the outcome and prognosis of sepsis. Recent studies have shown that inflammation, EC damage, oxidative stress, mitochondrial dysfunction, cardiomyocyte autophagy, and apoptosis play important roles in sepsis cardiac insufficiency.82,83
Pro-autophagic miRNA with deleterious impact
Wang et al.68 established a murine model of sepsis cardiac dysfunction exposed to lipopolysaccharide (LPS) and identified a remarkable increase in miR-21 in the myocardium of LPS-treated mice through microarray analysis. Inhibition of miR-21-3p with antagomiR prevents mitochondrial ultrastructural damage and autophagy, attenuates LPS-induced cardiac dysfunction, and improves the survival of mice treated with LPS in vivo. However, forced expression of miR-21-3p by using agomiR exacerbates sepsis-associated cardiac dysfunction through the suppression of SORBS2. Additionally, the receiver operating characteristic curve (0.939) suggests that miR-21-3p might be a specific predictor of the development of cardiac dysfunction in septic patients, thus providing a new strategy for disease treatment.68
lncRNAs and autophagy
lncRNAs are a class of RNA molecules with lengths greater than 200 nt, and they can regulate gene expression at the epigenetic, transcriptional, and post-transcriptional levels, in addition to directly modulating protein activity.84 The mechanisms of lncRNA action are extremely complex and have not been fully elucidated yet. According to existing studies, the mechanisms of lncRNAs can be classified into multiple types, including the following ones: (1) transcriptional interference, (2) induced chromatin remodeling and histone modification, (3) binding to specific protein to modulate their activity or alter their localization, and (4) miRNA sponge (Figure 2).85 Among these, serving as a competing endogenous RNA (ceRNA) is the most well-described mechanism through which lncRNAs can attenuate the activity of miRNA via sequestration, leading to elevated expression of the miRNA target gene. In a microarray study, 1,249 lncRNAs were identified as being differentially expressed in Ang II-induced cardiomyocyte autophagy, thus pointing to the existence of a correlation between abnormal lncRNA expression and autophagy in the heart.86 In a microarray analysis of H9c2 myocytes exposed to H/R, the lncRNA E230034O05Rik (HRIM) was found to play a pro-autophagic role to reduce the viability of myocytes.87 Nevertheless, H19 demonstrably protected mice against acute MI by activating autophagy.88 Thus far, numerous lncRNAs (see Table 2 and Figure 2 for a complete list) have been found to exert different effects in CVD occurrence either through pro- or anti-autophagy.
Figure 2.
Regulatory roles of lncRNAs in cardiac autophagy and related CVDs
The molecular mechanisms of lncRNAs are shown. lncRNAs can function as a sponge for miRNAs or as a scaffold by interacting with proteins such as transcription factors or components of chromatin-modifying complexes to activate or repress gene expression. lncRNAs can also bind to specific protein to modulate their activity or alter their localization. See text and Table 2 for detailed molecular mechanism explanations. Pink oval represents anti-autophagic lncRNAs with protective effects in CVDs; yellow oval represents anti-autophagic lncRNAs with deleterious effects; purple oval represents pro-autophagic lncRNAs with protective effects in CVDs; green oval represents pro-autophagic lncRNAs with deleterious effects in CVDs.
Table 2.
Dysregulated lncRNAs and their roles in autophagy and in the progression of CVDs
| LncRNAs | Roles in autophagy | Binding partners/downstream targets | Action modes | Outcomes | Refs |
|---|---|---|---|---|---|
| Myocardial infarction | |||||
| APF | pro- | miR-188/ATG7 | miRNA sponge | ↑ cell death | 23 |
| Galont | pro- | miR-338/ATG5 | miRNA sponge | ↑ cell death | 89 |
| RMRP | pro- | miR-206/ATG3 PI3K/Akt/mTOR pathway | miRNA sponge | ↑ myocardial I/R injury | 90 |
| TUG1 | pro- | miR-142-3p/HMGB1, Rac1 | miRNA sponge | ↑ myocardial I/R injury | 91 |
| XIST | pro- | miR-133a/SOCS2 | miRNA sponge | ↑ myocardial I/R injury | 92 |
| AK088388 | pro- | miR-30a/Beclin1 | miRNA sponge | ↑ cardiomyocyte damage | 93 |
| Mirf | anti- | miR-26a/USP15 | miRNA sponge | ↑ ischemic myocardial injury | 94 |
| MALAT1 | pro- | miR-20b/Beclin1 | miRNA sponge | ↑ cardiomyocyte injury | 95 |
| pro- | miR-558/ULK1 | miRNA sponge | ↓ cardiomyocyte apoptosis | 96 | |
| anti- | EZH2 protein TSC2 promoter | transcriptional suppression | ↑ cardiomyocyte apoptosis | 97 | |
| CAIF | anti- | p53 protein/myocardin | transcriptional suppression | ↓ myocardial infarction | 98 |
| EGOT | anti- | cyclin D1 protein PI3K/Akt/mTOR pathway | modulate protein activity | ↓ hypoxia-induced injury | 99 |
| FOXD3-AS1 | pro- | NF-κB/COX-2/iNOS pathway | modulate protein activity | ↑ myocardial I/R injury | 100 |
| HRIM | pro- | – | – | ↓ myocytes viability | 87 |
| H19 | pro- | – | – | ↓ acute myocardial infarction | 88 |
| Myocardial hypertrophy | |||||
| Chast | anti- | Plekhm1 | transcriptional suppression | ↑ cardiac remodeling | 101 |
| MIAT | pro- | mTOR, AMPK | modulate protein activity | ↑ myocardial hypertrophy | 102 |
| Diabetic cardiomyopathy | |||||
| DCRF | pro- | miR-551b-5p/PCDH17 | miRNA sponge | ↓ cardiac function | 103 |
| AK139328 | pro- | miR-204-3p | miRNA sponge | ↑ myocardial I/R injury | 104 |
| Neat1 | pro- | Foxo1 | modulate protein activity | ↑ myocardial I/R injury | 105 |
| Coronary atherosclerotic heart disease | |||||
| MALAT1 | anti- | miR-15b-5p/MAPK1 mTOR pathway | miRNA sponge | ↑ atherosclerosis | 106 |
lncRNAs, autophagy, MI, and myocardial I/R injury
lncRNA-miRNA sponging
Pro-autophagic lncRNAs with deleterious impacts
The first reported lncRNA that regulates autophagy and cell death in the heart was AK079427 in 2015, and it was named autophagy promoting factor (APF). Through in vitro and in vivo analyses, APF has been demonstrated to play a pro-autophagic role during I/R-induced MI by targeting miR-188-3p and the autophagy promoting gene ATG7.23 lncRNAs can be positive regulators of mRNA targets by sponging miRNAs and preventing them from completing their regulatory roles. miR-188-3p has been found to be involved in the regulation of ATG7 expression, and APF directly binds to miR-188-3p to block its ability to suppress autophagy and cell death.23 Similar to APF, other lncRNAs have also been confirmed to promote autophagic cell death in myocardial I/R injury through the lncRNA-miRNA-mRNA axis. GATA1-activated lncRNA (Galont) can sponge miR-338 to promote the expression of its downstream target ATG5 and autophagic cell death after anoxia/reoxygenation (A/R) stimuli.89 The enforced expression of the lncRNA component of mitochondrial RNA processing endoribonuclease (RMRP) aggravates hypoxia-induced injury by regulating the activity of miR-206 and the expression of its target ATG3.90 Recently, the miR-133a/SOCS2 axis has been demonstrated to convey the autophagic signal of lncRNA XIST. miR-133a directly targets SOCS2 to suppress apoptosis and autophagy and promote cell viability.92 XIST knockdown inhibits autophagy and cell apoptosis through the downregulation of miR-133a and attenuation of the MI area after I/R injury.92
HMGB1 and Rac1 are important autophagy-inducing proteins associated with the mTOR signaling pathway.91,107,108 Interestingly, miR-142-3p regulates both HMGB1 and Rac1; therefore, both of them are upregulated by lncRNA taurine upregulated gene 1 (TUG1) when it interacts with miR-142-3p, thus stimulating autophagic cell death in response to I/R.91 In addition, beclin-1 has been determined to be a direct target of both miR-30a and miR-20a-5p, as suggested by their opposite expression change trends after treatment and dual-luciferase assay verification.93,95 Simultaneously, lncRNA AK088388 and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) have been revealed to positively regulate the de-repression of beclin-1 by sponging miR-30a and miR-20a-5p, separately.93,95 Thus, the axes of AK088388/miR-30a and MALAT1/miR-20b contribute to beclin-1-mediated autophagy in cardiomyocyte injury.
Pro-autophagic lncRNAs with beneficial impacts
In response to stress, either extremely low autophagy levels or overactivation of autophagy is harmful to cells. Significantly, a certain level of basic autophagy is necessary for cell survival. In contrast to the previously described function of driving autophagic cell death,95 according to Guo et al.,96 MALAT1 promoted ULK1-mediated protective autophagy and facilitate myocardial protection in isoproterenol (ISO)-infused MI hearts by sponging miR-558. Initially, autophagy inhibitors 3-methyladenine (3M) and chloroquine were used to verify the protective role of autophagy in ISO-treated H9c2 cells; that is, autophagy inhibition enhanced the expression levels of the death substrate PARP and increased cell apoptosis. Silencing of MALAT1 increased miR-558 expression and decreased the level of the miR-558 target ULK1, leading to the suppression of autophagy and increased cell death. However, overexpression of MALAT1 sponged miR-558 and upregulated ULK1 to promote the protective autophagy.96 Taken together, these experiments revealed the protective role of the MALAT1/miR-558/ULK1 pathway against ISO-induced cardiomyocyte injury in a mitochondrial-dependent way.96
Anti-autophagic lncRNAs with deleterious impacts
According to Liang et al.,94 the lncRNA 2810403D21Rik/Mirf may be a potential therapeutic target for autophagy-related heart diseases. Mirf was upregulated, and Mir26a was significantly downregulated in the ischemic heart and cardiomyocytes. Moreover, in vivo silencing of Mir26a reduced autophagy levels and aggravated the impairment of cardiac function. However, overexpression of Mir26a could activate cardiomyocyte autophagy by targeting Usp15, thereby reducing ischemic stress-induced cardiomyocyte death.94 Mirf exerted its ceRNA mechanism to target and regulate the activity of Mir26a, thus inhibiting cardiomyocyte autophagy. In contrast, Mirf loss significantly upregulated the expression of Mir26a, activated autophagy, and ultimately reduced the ischemia-induced cardiac injury and improved mice cardiac function.94 Taken together, Mirf acts as an anti-autophagy molecule through the Mir26a/Usp15 axis to exacerbate ischemic cardiac injury.
lncRNA-protein interaction and regulation
Anti-autophagic lncRNAs with beneficial impacts
Wang and colleagues109 identified a lncRNA called CAIF that can inhibit cardiomyocyte autophagy and protect mouse heart against IR injury. Myocardin is a transcriptional coactivator of the serum response factor and is especially expressed in cardiac and smooth muscle cells. Through in vitro and in vivo studies, Wang and colleagues109 first demonstrated that myocardin positively regulates the induction of the autophagic process and cardiomyocyte death and promotes MI. Moreover, beclin-1 was revealed as the potential target of myocardin. Second, the transcription factor p53 can bind to myocardin promoter and activate its transcription. In contrast, CAIF can directly bind to p53 and block the p53-mediated transcription of myocardin, resulting in the downregulation of myocardin expression, which in turn inhibits cardiomyocyte autophagy. Wang and colleagues revealed the important role of the CAIF-p53-myocardin regulatory axis in cardiomyocyte autophagy, and they provided a potential therapeutic target for autophagy-related CVDs.
The expression of lncRNA EGOT was found to significantly decrease in H9c2 cells suffering from hypoxia.99 Moreover, EGOT was found to mediate the protective effects of the recombinant human brain natriuretic on hypoxia-induced injury in H9c2 cells by regulating the expression of cyclin D1 and activating the PI3K/AKT/mTOR pathway with reduced apoptosis and autophagic levels.99
Pro-autophagic lncRNAs with deleterious impacts
In oxygen glucose deprivation (OGD)/R-induced H9c2 cells, lncRNA FOXD3-AS1 was found to increase, and its overexpression aggravated myocardial IR injury by promoting autophagy through the activation of the nuclear factor κB (NF-κB)/COX-2/inducible NO (nitric oxide) synthase (iNOS) signaling pathway.100 However, the administration of 3M downregulated the associated proteins and reversed the aggravation of cardiac myocyte damage.100
lncRNA-DNA binding and chromatin remodeling
lncRNAs can regulate epigenetic modifiers, such as DNMTs and EZH2, to induce chromatin remodeling and histone modification. Epigenetic changes to certain genes are involved in the progression of CVDs. For instance, lncRNA MALAT1 can recruit EZH2 to induce H3K27me3 modification in the TSC2 promoter region, thus repressing its transcription, while TSC2 overexpression inhibits mTOR signaling and activates autophagy. Therefore, the increased MALAT1 expression induced by H/R injury inhibits cardiomyocyte proliferation and enhances apoptosis through autophagy inhibition by regulating TSC2-mTOR signaling activity.97
lncRNAs, autophagy, and cardiac hypertrophy
lncRNA-transcription regulation
Viereck et al.101 observed a significant upregulation of Chast (cardiac hypertrophy-associated transcript) in cardiomyocytes from TAC mice as well as in hypertrophic heart tissue from aortic stenosis patients. They identified the roles of Chast in impeding autophagy and promoting cardiac hypertrophy by conducting overexpression and silencing experiments. Mechanistically, Chast is activated by the well-defined pro-hypertrophic transcription factor NFAT. In addition, Chast is partially transcribed from the opposite strand of the protein-coding gene Plekhm1, which can intensely repress the transcription of Plekhm1. The silencing of Plekhm1 results in reduced autophagic capacity and hypertrophic responses. This reveals a transcriptional regulation circuit of the lncRNA Chast in autophagy and cardiac hypertrophy.
lncRNA-protein interaction
The lncRNA MIAT (MI-associated transcript) has been initially reported to play a role in MI, and its over-expanding role in other diseases has been recently revealed.110 MIAT mediates the inhibitory effect of berberine on cardiac hypertrophy by regulating autophagy.102 MIAT knockdown alleviates the hypertrophic responses and the relative protein levels of phosphorylated (p-)mTOR, p-AMPK, and LC3 in H9c2 cells treated with Ang II.
lncRNAs, autophagy, and DCM
lncRNA-miRNA sponging
The lncRNA DCRF (DCM-related factor) was first described as being upregulated in DCM in a microarray-based screen for dysregulated lncRNAs in diabetic hearts with pathological remodeling versus normal hearts.103 DCRF knockdown significantly relieved high glucose-induced cardiomyocyte autophagy and improved histological abnormalities and cardiac function in diabetic rats. When functioning as a ceRNA, DCRF can increase the expression of PCDH17 through miR-155b-5p sponging and consequently impel autophagy.103 Thus, it can be thought that the DCRF/miR-155b-5p/PCDH17 axis contributes to the progression of DCM.
Similarly, by using a microarray approach to compare the lncRNA transcriptome of I/R injury in diabetic mice and normal mice, Yu et al.104 identified the overexpression of AK139328 in diabetic mice. A subsequent molecular mechanism investigation revealed that AK139328 knockdown blunted cardiomyocyte autophagy and attenuated myocardial I/R injury through direct sponging of miR-204-3p.104 Nevertheless, the target protein-coding gene of miR-204-3p has not been studied.
lncRNA-protein interaction
Through ex vivo and in vivo analyses, Ma et al.105 demonstrated the participation of the lncRNA nuclear-enriched abundant transcript 1 (Neat1) in diabetic rats with I/R injury. Neat1 was found to exacerbate cardiomyocyte injuries through the activation of myocardial apoptosis and autophagy, as indicated by the elevated fluorescent LC3 puncta and increased levels of autophagy-related proteins. Furthermore, Neat1 promoted the autophagy level by upregulating the expression of Foxo1, a transcription factor that modulates physiological functions through the transcription and transmission of various growth factors and cytokine signals.105
lncRNAs, autophagy, and coronary atherosclerotic heart disease
The MALAT1/miR-15b-5p/MAPK1 axis has been described in coronary atherosclerotic heart disease, and according to this description, MALAT1 directly targets miR-15b-5p to amplify MAPK1 expression.106 Subsequently, this signal axis triggers the mTOR signaling pathway to enhance the progression of coronary atherosclerotic heart disease. In contrast, MALAT1 inhibition promotes the autophagy of endothelial progenitor cells and increases cell viability, thereby protecting mice against AS.106
circRNAs and autophagy
circRNAs are covalently closed cyclic RNAs with high frequencies of eukaryotic transcripts, and they can act as miRNA sponges to regulate splicing or transcription and the expression of parental genes. Recent studies have demonstrated their critical roles in neurological disorders, infectious diseases, cancers, and CVDs.111 As an important process of cellular physiological activity, the relationship between autophagy and circRNAs has been studied. In 2017, Yao and colleagues112 first reported that circHIPK2 can modulate astrocyte activation through cooperation between autophagy and ER stress by targeting the MIR124-2HG/SIGMAR1 axis. Subsequently, many circRNAs have been identified to regulate autophagy through the sponging of miRNAs in various cancers.113, 114, 115, 116
Thus far, only two studies have examined the autophagy-related circRNAs in cardiovascular pathologies. Different from their most common regulatory mechanism of miRNA sponges, these two circRNAs exert their functions through interactions with proteins. Wang and colleagues117 reported that an autophagy-related circRNA called ACR participated in regulating autophagy in a myocardial I/R injury model (Figure 3). ACR expression decreased significantly in cardiomyocytes after A/R treatment. ACR overexpression reduced the accumulation of GFP-LC3 puncta, expression levels of LC-II, and numbers of autophagic vesicles in A/R-treated cells, indicating that ACR inhibited A/R-induced autophagy and mitophagy in cardiomyocytes. Through transcriptome microarray analysis, they revealed that ACR can inhibit PINK1 methylation and promote PINK1 expression by directly binding to Dnmt3B. Furthermore, Wang and colleagues117 demonstrated that PINK1 regulates the autophagy pathway in vitro and in vivo. To identify the mechanism through which PINK1 regulates autophagy, the interaction between PINK1 and FAM65B was tested with western identification after co-immunoprecipitation. Based on mass spectrometry analysis and antibody verification, S46 was proved to be the PINK1 site for targeting FAM65B. PINK1 can facilitate the phosphorylation of FAM65B, thus inhibiting cardiomyocyte autophagy and protecting cells during myocardial I/R injury. The other one is circRNA ZNF 292, which can alleviate OGD-induced ischemic injury, including apoptosis and autophagy, in H9c2 cells by targeting BNIP3 through Wnt/β-catenin and mTOR activation.118 Overall, these findings reveal novel roles of circRNA ARC and ZNF292 in regulating autophagy and their potential applications as therapeutic targets in the treatment of CVDs.
Figure 3.
Mechanism diagram of circRNA ACR in myocardial I/R injury
Though directly binding to Dnmt3B, ACR inhibits PINK1 methylation and promotes PINK1 expression, which facilitates the phosphorylation of FAM65B at Ser46, resulting in reduced autophagy and myocardial I/R injury.
Clinical relevance of ncRNAs in CVDs
Due to the lack of effective therapeutic targets, the global morbidity and mortality of CVDs are increasing year by year. Nevertheless, it is inspiring that the research on ncRNAs, which play a critical regulatory role in gene expression in the physiology and pathophysiology of cardiovascular system, has developed rapidly, and the potential of ncRNAs for the diagnosis and treatment of CVDs is supported by multiple studies. We searched the https://clinicaltrials.gov database for publicly available studies by the keywords associated with ncRNAs, and then the clinical trials details were downloaded. At the time of our search, there were 939 studies found for miRNA, 100 of which were CVDs related; 48 studies were found for lncRNAs, 7 of which were CVDs related; and 7 studies were found for circRNAs, 2 of which were CVDs related. Given the high tissue specificity and stability of ncRNAs, as well as their easy access for monitoring, it is reasonable that most of the above trials are diagnostic, where ncRNAs are used as novel clinical biomarkers for CVDs diagnosis. Furthermore, with the development of gene-based technologies including RNA interference (RNAi) and antisense oligonucleotides, a lot of therapeutic clinical trials using ncRNAs as treatment targets and therapeutics have begun.119,120 Thus, ncRNAs are worth looking forward to for their use in elucidating the pathogenesis of certain CVDs and becoming novel diagnostic biomarkers and intriguing targeted therapeutic tools.
ncRNAs as CVD biomarkers
Although cardiac troponin I (cTnl), creatine kinase isoenzyme (CK-MB), and brain natriuretic peptide (BNP) have been widely used in the diagnosis of myocardial ischemia, MI, and HF, more stable and specific disease diagnostic markers still need to be explored widely. At present, a number of studies focus on mining ncRNAs in body fluids as CVD biomarkers and are carried out for clinical research in a population.
ncRNAs can be used to screen and distinguish biomarkers in the diagnosis of CVDs. Screening of differentially expressed miRNAs in the blood of healthy people and patients with HCM identified that 12 miRNAs were significantly elevated in the plasma of patients. Among them, only circulating miR-29a was significantly correlated with both hypertrophy and fibrosis, indicating it as a potential diagnosis biomarker for cardiac remodeling assessment in HCM.121 miR-423-5p was tested as a specific biomarker for clinical HF, the levels of which are related to N-terminal pro-BNP (NT-proBNP) and New York Heart Association (NYHA) classification.122 As acute MI is a cardiovascular event with high mortality, a rapid diagnostic method with high sensitivity and specificity is urgently needed. Of the six acute MI-relevant miRNAs, circulating miR-22-5p and miR-122-5p are promising diagnostic biomarkers, and the joint detection of them increased the distinguished sensitivity (98.6%) between acute MI patients and healthy controls.123 Moreover, a full-featured microwell detector has been developed to rapidly detect miRNAs in plasma for CVD diagnosis, which can simplify the detection steps and eliminate human errors.124 Analysis of miRNAs in the plasma of patients with HF with reduced or preserved LV ejection fraction (HFrEF versus HFpEF) revealed that miR-125a-5p, miR-190a, miR-550a-5p, and miR-638 can be used to distinguish these two subtypes of HF.125 BNP and NT-proBNP are the gold standards for clinical diagnosis of HF, with high sensitivity but poor specificity. In addition, multi-miRNA panels in combination with NT-proBNP can significantly improve the specificity and accuracy in identifying nonacute HF with HFpEF.126
Mitochondrial lncRNA LIPCAR is differentially expressed between patients with or without LV remodeling after MI, and it is also independent of other risk markers associated with future cardiovascular mortality, suggesting that it may be used as a biomarker of cardiac remodeling and predict survival in patients with HF.127 Other lncRNAs such as CoroMarker and PPARδ are useful biomarkers for coronary artery disease (CAD) diagnosis.128 As for disease classification, lncRNA uc004cov.4 and uc022bqu.1 can distinguish the obstructive HCM from the nonobstructive subtype.129 de Gonzalo-Calvo et al.130 reported for the first time that lncRNA SENCR can be used as a potential candidate molecular marker for evaluating the efficacy of individualized treatment of type 2 diabetes mellitus (T2DM) patients with pioglitazone, a thiazolidinedione insulin sensitizer that improves LV diastolic function in T2DM patients. To date, lncRNA prostate cancer antigen-3 (PCA3) is the first and only ncRNA that has been approved by the US Food and Drug Administration (FDA) for routine clinical diagnosis of prostate cancer.131 Thus, whether lncRNA can be used in clinical diagnosis of CVDs remains to be further verified.
Distinct from miRNAs and lncRNAs, the closed ring structure makes circRNAs more stable and have more advantages in the application as clinical diagnostic markers.132 Salgado-Somoza et al.133 reported that circRNA MICRA is differentially low in patients with HF after acute MI (EF of ≤40%), and a bootstrap internal validation revealed that MICRA was selected in 86% of the models to be an optimal predictive marker, which is completely comparable to the traditional NT-proBNP and has high diagnostic value. So far, lots of circRNAs have been identified and verified as potential biomarkers to diagnose CVDs, such as hsa_circ_0124644 and hsa_circ_ 11783-2 for CAD, hsa_circ_ 081881 for acute MI, hsa_circ_ 025016 for postoperative AF, and so on. Nevertheless, only with multi-center clinical trials with larger sample size can we more convincingly confirm the role of ncRNAs in the diagnosis of CVDs. Additionally, more basic studies are needed to clarify the biological role of ncRNAs in the occurrence and development of CVDs.
Therapeutic targeting of ncRNAs in CVD preclinical studies and clinical trials
In addition to their roles as potential biomarkers, ncRNAs also represent targets for new therapeutic approaches and attractive tools.134,135 In 2018, the first RNAi drug patisiran (reduces TTR levels) was approved by the FDA for the treatment of neurodegenerative disease hereditary transthyretin amyloidosis, marking the dawn of a new era of clinical application of RNA as a target and tool.136 Recently, cobomarsen (also termed MRG-106; miRagen Therapeutics), a locked nucleic acid (LNA)-based oligonucleotide against miR-155, has entered a phase II clinical trial in patients with cutaneous T cell lymphoma, mycosis fungoides subtype (ClinicalTrials.gov: NCT03837457), and a phase I clinical trial in patients with different leukemia/lymphoma (ClinicalTrials.gov: NCT02580552). Among other miRNA candidates, MRG-201 (remlarsen; miRagen Therapeutics), a miR-29 mimic, is currently undergoing a phase II clinical trial in participants with cutaneous fibrosis (ClinicalTrials.gov: NCT03601052). Moreover, MRG-201 has the potential clinical application in a number of pathological fibrotic conditions, including cardiac fibrosis.
At present, a larger number of preclinical studies on ncRNA-based therapeutics for the treatment of CVDs are under way, and some of them have entered clinical trials. For example, miR-33a/b is involved in the regulation of lipid metabolism.137,138 Preclinical studies have shown that silencing miR-33 with anti-miR-33 in non-human primates can effectively reduce the plasma very-low-density lipoprotein (VLDL) and raise the high-density lipoprotein (HDL) level without significant side effects, indicating that miR-33 has the potential to become a therapeutic target for the treatment of AS and dyslipidemia.139,140 Other therapeutic candidates such as LNA antimiR-21 for cardiac fibrosis treatment, antimiR-208 for cardiac remodeling treatment, and antimiR-15 for MI treatment are expected to become a hotspot in clinical research.134 Recently, MRG-110 (LNA antimir-92) aimed for HF indication has just completed the phase I clinical trial (ClinicalTrials.gov: NCT03603431) by intradermal injection in heathy volunteers. The small interfering RNA (siRNA) drug inclisiran (ALN-PCS) is directed against proprotein convertase subtilisin-kexin type 9 (PCSK9) mRNA to reduce LDL cholesterol (LDL-C) level, which is high cardiovascular risk of AS and CAD.141 In the phase II clinical trial (ClinicalTrials.gov: NCT02597127 and NCT03060577) of inclisiran injection in participants with atherosclerotic CVDs, the serum LDL-C levels were decreased, and the tolerance was acceptable, and now a phase III clinical trial (ClinicalTrials.gov: NCT03705234) is under way to assess its effect on clinical outcomes among people with CVDs. In future studies, the combination of high-throughput sequencing and CRISPR-Cas9 gene editing technologies will help to quickly screen ncRNAs that play critical roles in CVDs. However, although having good prospects, there are still some issues to be solved in clinical trials: (1) for lncRNA and circRNA with poor conservation among species, it is necessary to find appropriate preclinical animal models; (2) ncRNAs are widely distributed, and thus drug delivery systems that specifically target heart tissue are in need of development; and (3) the in vivo safety of the currently used ncRNA mimics or inhibitors, such as anti-miR, antagomir, LNA oligonucleotides, and viral vectors, should be verified.
Conclusions
The normal progress of material synthesis and catabolism maintains the human body’s homeostasis. Autophagy is one of the important metabolic pathways that removes dysfunctional organelles, misfolded proteins, and macromolecules in eukaryotic cells, thus realizing the efficient and stable operation of the intracellular environment. Autophagy is critical for cardiovascular system homeostasis and closely related to CVDs, including myocardial I/R injury, cardiac hypertrophy, HF, cardiomyopathy, and AS. This systematic review, despite focusing on the ncRNA regulation field, indicates that autophagy plays a dual role in CVD pathology. Moderate levels of autophagy can protect cells from apoptosis and help maintain normal cardiovascular metabolism by degrading and recycling damaged organelles and aging proteins. However, excessive autophagy can destroy the essential cell components necessary for cell survival and cardiac contraction, and insufficient autophagy can lead to the accumulation of damaged organelles and redundant proteins, which are detrimental to myocardial cells and contribute to disease progression. Thus, regulators of autophagy, such as mTOR, AMPK, and others, can be used as targets to prevent or treat CVDs. So far, drugs such as rapamycin (an mTOR inhibitor), histone deacetylase inhibitors, and spermidine have been demonstrated to be useful for improving cardiovascular functions and decelerating the progression of CVD in animal models through modulation of autophagy.142, 143, 144, 145 However, the clinical evaluation of autophagy-targeting drugs within the cardiovascular system is limited owing to the lack of drug compounds that specifically regulate autophagy in heart and non-invasive biomarkers that reflect their effects on autophagy.146
In recent years, various pieces evidence have indicated that ncRNA-mediated post-transcriptional regulation of gene expression represents an integral part of the autophagy regulatory network and has a substantial effect on autophagy-related diseases.147 Therefore, determining whether ncRNA-regulated autophagy plays a protective role against CVDs or promotes the progression of CVDs can facilitate the development of ncRNAs as therapeutic targets in the treatment of autophagy-related CVDs. For instance, CAIF is a novel lncRNA with protective effects on MI through the inhibition of autophagy, and patients with end-stage cardiomyopathy exhibit downregulated CAIF levels.98 Additionally, ATG5-associated lncRNA Chast has been tested to verify the relationship between autophagy dysregulation and CAD.148,149 These results suggest that autophagy-regulating ncRNAs can potentially serve as novel targets in the development of diagnostic and therapeutic agents. Moreover, it is reasonable to think that the mechanism research on autophagy and ncRNAs can provide new strategies for the prevention and treatment of CVDs.
Acknowledgments
This work was supported by the National Natural Science Foundation of China (no. 81800264); the Major Research Program of the National Natural Science Foundation of China (no. 91849209); the Applied Basic Research Source Innovation Plan of Qingdao, China (no. 19-6-2-78-cg); the Launch Fund of Qingdao University (DC1900003122); and by the Program of Qilu Hospital of Shandong University (Qingdao) (QDKY2017QN05).
Author contributions
J.G. and K.S. conceived this article. X.C. and C.S. collected the related papers. J.G. wrote the manuscript. K.S. drew the figures. Y.W. and P.L. helped to revise the manuscript and do the final editing.
Declarations of interests
The authors declare no competing interests.
Contributor Information
Jinning Gao, Email: gjn.1127@163.com.
Kai Shao, Email: qlshaokai@163.com.
References
- 1.Sanchez P., De Carcer G., Sandoval I.V., Moscat J., Diaz-Meco M.T. Localization of atypical protein kinase C isoforms into lysosome-targeted endosomes through interaction with p62. Mol. Cell. Biol. 1998;18:3069–3080. doi: 10.1128/mcb.18.5.3069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Yun H.R., Jo Y.H., Kim J., Shin Y., Kim S.S., Choi T.G. Roles of autophagy in oxidative stress. Int. J. Mol. Sci. 2020;21:3289. doi: 10.3390/ijms21093289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Klionsky D.J., Emr S.D. Autophagy as a regulated pathway of cellular degradation. Science. 2000;290:1717–1721. doi: 10.1126/science.290.5497.1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Liao S.X., Sun P.P., Gu Y.H., Rao X.M., Zhang L.Y., Ou-Yang Y. Autophagy and pulmonary disease. Ther. Adv. Respir. Dis. 2019;13 doi: 10.1177/1753466619890538. 1753466619890538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Russell R.C., Tian Y., Yuan H., Park H.W., Chang Y.Y., Kim J., Kim H., Neufeld T.P., Dillin A., Guan K.L. ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat. Cell Biol. 2013;15:741–750. doi: 10.1038/ncb2757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kim J., Kim Y.C., Fang C., Russell R.C., Kim J.H., Fan W., Liu R., Zhong Q., Guan K.L. Differential regulation of distinct Vps34 complexes by AMPK in nutrient stress and autophagy. Cell. 2013;152:290–303. doi: 10.1016/j.cell.2012.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gwinn D.M., Shackelford D.B., Egan D.F., Mihaylova M.M., Mery A., Vasquez D.S., Turk B.E., Shaw R.J. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell. 2008;30:214–226. doi: 10.1016/j.molcel.2008.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Terman A., Brunk U.T. Autophagy in cardiac myocyte homeostasis, aging, and pathology. Cardiovasc. Res. 2005;68:355–365. doi: 10.1016/j.cardiores.2005.08.014. [DOI] [PubMed] [Google Scholar]
- 9.Zhang C., Syed T.W., Liu R., Yu J. Role of endoplasmic reticulum stress, autophagy, and inflammation in cardiovascular disease. Front. Cardiovasc. Med. 2017;4:29. doi: 10.3389/fcvm.2017.00029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sun T., Li M.Y., Li P.F., Cao J.M. MicroRNAs in cardiac autophagy: small molecules and big role. Cells. 2018;7:104. doi: 10.3390/cells7080104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Turkieh A., Charrier H., Dubois-Deruy E., Porouchani S., Bouvet M., Pinet F. Noncoding RNAs in cardiac autophagy following myocardial infarction. Oxid. Med. Cell. Longev. 2019;2019:8438650. doi: 10.1155/2019/8438650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yao H., Han B., Zhang Y., Shen L., Huang R. Non-coding RNAs and autophagy. Adv. Exp. Med. Biol. 2019;1206:199–220. doi: 10.1007/978-981-15-0602-4_10. [DOI] [PubMed] [Google Scholar]
- 13.Tay Y., Zhang J., Thomson A.M., Lim B., Rigoutsos I. MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation. Nature. 2008;455:1124–1128. doi: 10.1038/nature07299. [DOI] [PubMed] [Google Scholar]
- 14.Miranda K.C., Huynh T., Tay Y., Ang Y.S., Tam W.L., Thomson A.M., Lim B., Rigoutsos I. A pattern-based method for the identification of microRNA binding sites and their corresponding heteroduplexes. Cell. 2006;126:1203–1217. doi: 10.1016/j.cell.2006.07.031. [DOI] [PubMed] [Google Scholar]
- 15.Xiao J., Zhu X., He B., Zhang Y., Kang B., Wang Z., Ni X. miR-204 regulates cardiomyocyte autophagy induced by ischemia-reperfusion through LC3-II. J. Biomed. Sci. 2011;18:35. doi: 10.1186/1423-0127-18-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhen W., Hui D., Wenying S., Yulong S. MicroRNA-20b-5p regulates propofol-preconditioning-induced inhibition of autophagy in hypoxia-and-reoxygenation-stimulated endothelial cells. J. Biosci. 2020;45:35. [PubMed] [Google Scholar]
- 17.Huang Z., Wu S., Kong F., Cai X., Ye B., Shan P., Huang W. MicroRNA-21 protects against cardiac hypoxia/reoxygenation injury by inhibiting excessive autophagy in H9c2 cells via the Akt/mTOR pathway. J. Cell. Mol. Med. 2017;21:467–474. doi: 10.1111/jcmm.12990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhang J., Qiu W., Ma J., Wang Y., Hu Z., Long K., Wang X., Jin L., Tang Q., Tang G. miR-27a-5p attenuates hypoxia-induced rat cardiomyocyte injury by inhibiting Atg7. Int. J. Mol. Sci. 2019;20:2418. doi: 10.3390/ijms20102418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shao H., Yang L., Wang L., Tang B., Wang J., Li Q. MicroRNA-34a protects myocardial cells against ischemia-reperfusion injury through inhibiting autophagy via regulating TNFα expression. Biochem. Cell Biol. 2018;96:349–354. doi: 10.1139/bcb-2016-0158. [DOI] [PubMed] [Google Scholar]
- 20.Li Q., Gao Y., Zhu J., Jia Q. miR-101 attenuates myocardial infarction-induced Injury by targeting DDIT4 to regulate autophagy. Curr. Neurovasc. Res. 2020;17:123–130. doi: 10.2174/1567202617666200211113016. [DOI] [PubMed] [Google Scholar]
- 21.Zhang C., Zhang C., Wang H., Qi Y., Kan Y., Ge Z. Effects of miR-103a-3p on the autophagy and apoptosis of cardiomyocytes by regulating Atg5. Int. J. Mol. Med. 2019;43:1951–1960. doi: 10.3892/ijmm.2019.4128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zhang H., Zhang X., Zhang J. miR-129-5p inhibits autophagy and apoptosis of H9c2 cells induced by hydrogen peroxide via the PI3K/AKT/mTOR signaling pathway by targeting ATG14. Biochem. Biophys. Res. Commun. 2018;506:272–277. doi: 10.1016/j.bbrc.2018.10.085. [DOI] [PubMed] [Google Scholar]
- 23.Wang K., Liu C.Y., Zhou L.Y., Wang J.X., Wang M., Zhao B., Zhao W.K., Xu S.J., Fan L.H., Zhang X.J. APF lncRNA regulates autophagy and myocardial infarction by targeting miR-188-3p. Nat. Commun. 2015;6:6779. doi: 10.1038/ncomms7779. [DOI] [PubMed] [Google Scholar]
- 24.Qiu R., Li W., Liu Y. MicroRNA-204 protects H9C2 cells against hypoxia/reoxygenation-induced injury through regulating SIRT1-mediated autophagy. Biomed. Pharmacother. 2018;100:15–19. doi: 10.1016/j.biopha.2018.01.165. [DOI] [PubMed] [Google Scholar]
- 25.Ding S., Abudupataer M., Zhou Z., Chen J., Li H., Xu L., Zhang W., Zhang S., Zou Y., Hong T. Histamine deficiency aggravates cardiac injury through miR-206/216b-Atg13 axis-mediated autophagic-dependant apoptosis. Cell Death Dis. 2018;9:694. doi: 10.1038/s41419-018-0723-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Liu X., Deng Y., Xu Y., Jin W., Li H. MicroRNA-223 protects neonatal rat cardiomyocytes and H9c2 cells from hypoxia-induced apoptosis and excessive autophagy via the Akt/mTOR pathway by targeting PARP-1. J. Mol. Cell. Cardiol. 2018;118:133–146. doi: 10.1016/j.yjmcc.2018.03.018. [DOI] [PubMed] [Google Scholar]
- 27.Zhao P., Zhang B.L., Liu K., Qin B., Li Z.H. Overexpression of miR-638 attenuated the effects of hypoxia/reoxygenation treatment on cell viability, cell apoptosis and autophagy by targeting ATG5 in the human cardiomyocytes. Eur. Rev. Med. Pharmacol. Sci. 2018;22:8462–8471. doi: 10.26355/eurrev_201812_16546. [DOI] [PubMed] [Google Scholar]
- 28.Yang Y., Li Y., Chen X., Cheng X., Liao Y., Yu X. Exosomal transfer of miR-30a between cardiomyocytes regulates autophagy after hypoxia. J. Mol. Med. (Berl.) 2016;94:711–724. doi: 10.1007/s00109-016-1387-2. [DOI] [PubMed] [Google Scholar]
- 29.Wu D., Jiang H., Chen S., Zhang H. Inhibition of microRNA-101 attenuates hypoxia/reoxygenation-induced apoptosis through induction of autophagy in H9c2 cardiomyocytes. Mol. Med. Rep. 2015;11:3988–3994. doi: 10.3892/mmr.2015.3215. [DOI] [PubMed] [Google Scholar]
- 30.Zhang Z., Li H., Chen S., Li Y., Cui Z., Ma J. Knockdown of microRNA-122 protects H9c2 cardiomyocytes from hypoxia-induced apoptosis and promotes autophagy. Med. Sci. Monit. 2017;23:4284–4290. doi: 10.12659/MSM.902936. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 31.Zou Y., Liu W., Zhang J., Xiang D. miR-153 regulates apoptosis and autophagy of cardiomyocytes by targeting Mcl-1. Mol. Med. Rep. 2016;14:1033–1039. doi: 10.3892/mmr.2016.5309. [DOI] [PubMed] [Google Scholar]
- 32.Chang L., Chai X., Chen P., Cao J., Xie H., Zhu J. miR-181b-5p suppresses starvation-induced cardiomyocyte autophagy by targeting Hspa5. Int. J. Mol. Med. 2019;43:143–154. doi: 10.3892/ijmm.2018.3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhu Q., Hu F. Antagonism of miR-429 ameliorates anoxia/reoxygenation injury in cardiomyocytes by enhancing MO25/LKB1/AMPK mediated autophagy. Life Sci. 2019;235:116842. doi: 10.1016/j.lfs.2019.116842. [DOI] [PubMed] [Google Scholar]
- 34.Ma W., Ding F., Wang X., Huang Q., Zhang L., Bi C., Hua B., Yuan Y., Han Z., Jin M. By targeting Atg7 MicroRNA-143 mediates oxidative stress-induced autophagy of c-Kit+ mouse cardiac progenitor cells. EBioMedicine. 2018;32:182–191. doi: 10.1016/j.ebiom.2018.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wu J., Sun C., Wang R., Li J., Zhou M., Yan M., Xue X., Wang C. Cardioprotective effect of paeonol against epirubicin-induced heart injury via regulating miR-1 and PI3K/AKT pathway. Chem. Biol. Interact. 2018;286:17–25. doi: 10.1016/j.cbi.2018.02.035. [DOI] [PubMed] [Google Scholar]
- 36.Li G., Wang G., Ma L., Guo J., Song J., Ma L., Zhao X. miR-22 regulates starvation-induced autophagy and apoptosis in cardiomyocytes by targeting p38α. Biochem. Biophys. Res. Commun. 2016;478:1165–1172. doi: 10.1016/j.bbrc.2016.08.086. [DOI] [PubMed] [Google Scholar]
- 37.Yan L., Guo N., Cao Y., Zeng S., Wang J., Lv F., Wang Y., Cao X. miRNA-145 inhibits myocardial infarction-induced apoptosis through autophagy via Akt3/mTOR signaling pathway in vitro and in vivo. Int. J. Mol. Med. 2018;42:1537–1547. doi: 10.3892/ijmm.2018.3748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hu S., Cao S., Tong Z., Liu J. FGF21 protects myocardial ischemia-reperfusion injury through reduction of miR-145-mediated autophagy. Am. J. Transl. Res. 2018;10:3677–3688. [PMC free article] [PubMed] [Google Scholar]
- 39.Higashi K., Yamada Y., Minatoguchi S., Baba S., Iwasa M., Kanamori H., Kawasaki M., Nishigaki K., Takemura G., Kumazaki M. MicroRNA-145 repairs infarcted myocardium by accelerating cardiomyocyte autophagy. Am. J. Physiol. Heart Circ. Physiol. 2015;309:H1813–H1826. doi: 10.1152/ajpheart.00709.2014. [DOI] [PubMed] [Google Scholar]
- 40.Li J., Cai S.X., He Q., Zhang H., Friedberg D., Wang F., Redington A.N. Intravenous miR-144 reduces left ventricular remodeling after myocardial infarction. Basic Res. Cardiol. 2018;113:36. doi: 10.1007/s00395-018-0694-x. [DOI] [PubMed] [Google Scholar]
- 41.Li Q., Xie J., Li R., Shi J., Sun J., Gu R., Ding L., Wang L., Xu B. Overexpression of microRNA-99a attenuates heart remodelling and improves cardiac performance after myocardial infarction. J. Cell. Mol. Med. 2014;18:919–928. doi: 10.1111/jcmm.12242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bo L., Su-Ling D., Fang L., Lu-Yu Z., Tao A., Stefan D., Kun W., Pei-Feng L. Autophagic program is regulated by miR-325. Cell Death Differ. 2014;21:967–977. doi: 10.1038/cdd.2014.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Pan W., Zhong Y., Cheng C., Liu B., Wang L., Li A., Xiong L., Liu S. miR-30-regulated autophagy mediates angiotensin II-induced myocardial hypertrophy. PLoS ONE. 2013;8:e53950. doi: 10.1371/journal.pone.0053950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Huang J., Sun W., Huang H., Ye J., Pan W., Zhong Y., Cheng C., You X., Liu B., Xiong L., Liu S. miR-34a modulates angiotensin II-induced myocardial hypertrophy by direct inhibition of ATG9A expression and autophagic activity. PLoS ONE. 2014;9:e94382. doi: 10.1371/journal.pone.0094382. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 45.Li A.L., Lv J.B., Gao L. miR-181a mediates Ang II-induced myocardial hypertrophy by mediating autophagy. Eur. Rev. Med. Pharmacol. Sci. 2017;21:5462–5470. doi: 10.26355/eurrev_201712_13936. [DOI] [PubMed] [Google Scholar]
- 46.Shi J.Y., Chen C., Xu X., Lu Q. miR-29a promotes pathological cardiac hypertrophy by targeting the PTEN/AKT/mTOR signalling pathway and suppressing autophagy. Acta Physiol. (Oxf.) 2019;227:e13323. doi: 10.1111/apha.13323. [DOI] [PubMed] [Google Scholar]
- 47.Li Z., Song Y., Liu L., Hou N., An X., Zhan D., Li Y., Zhou L., Li P., Yu L. miR-199a impairs autophagy and induces cardiac hypertrophy through mTOR activation. Cell Death Differ. 2017;24:1205–1213. doi: 10.1038/cdd.2015.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ucar A., Gupta S.K., Fiedler J., Erikci E., Kardasinski M., Batkai S., Dangwal S., Kumarswamy R., Bang C., Holzmann A. The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy. Nat. Commun. 2012;3:1078. doi: 10.1038/ncomms2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jin L., Zhou Y., Han L., Piao J. MicroRNA302-367-PI3K-PTEN-AKT-mTORC1 pathway promotes the development of cardiac hypertrophy through controlling autophagy. In Vitro Cell. Dev. Biol. Anim. 2020;56:112–119. doi: 10.1007/s11626-019-00417-5. [DOI] [PubMed] [Google Scholar]
- 50.Wu H., Wang Y., Wang X., Li R., Yin D. MicroRNA-365 accelerates cardiac hypertrophy by inhibiting autophagy via the modulation of Skp2 expression. Biochem. Biophys. Res. Commun. 2017;484:304–310. doi: 10.1016/j.bbrc.2017.01.108. [DOI] [PubMed] [Google Scholar]
- 51.Wang L., Ye N., Lian X., Peng F., Zhang H., Gong H. miR-208a-3p aggravates autophagy through the PDCD4-ATG5 pathway in Ang II-induced H9c2 cardiomyoblasts. Biomed. Pharmacother. 2018;98:1–8. doi: 10.1016/j.biopha.2017.12.019. [DOI] [PubMed] [Google Scholar]
- 52.Zou M., Wang F., Gao R., Wu J., Ou Y., Chen X., Wang T., Zhou X., Zhu W., Li P. Autophagy inhibition of hsa-miR-19a-3p/19b-3p by targeting TGF-β R II during TGF-β1-induced fibrogenesis in human cardiac fibroblasts. Sci. Rep. 2016;6:24747. doi: 10.1038/srep24747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zheng L., Lin S., Lv C. miR-26a-5p regulates cardiac fibroblasts collagen expression by targeting ULK1. Sci. Rep. 2018;8:2104. doi: 10.1038/s41598-018-20561-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhao X.D., Qin R.H., Yang J.J., Xu S.S., Tao H., Ding X.S., Shi K.H. DNMT3A controls miR-200b in cardiac fibroblast autophagy and cardiac fibrosis. Inflamm. Res. 2018;67:681–690. doi: 10.1007/s00011-018-1159-2. [DOI] [PubMed] [Google Scholar]
- 55.Zhou S., Lei D., Bu F., Han H., Zhao S., Wang Y. MicroRNA-29b-3p targets SPARC gene to protect cardiocytes against autophagy and apoptosis in hypoxic-induced H9c2 cells. J. Cardiovasc. Transl. Res. 2019;12:358–365. doi: 10.1007/s12265-018-9858-1. [DOI] [PubMed] [Google Scholar]
- 56.Lai L., Chen J., Wang N., Zhu G., Duan X., Ling F. miRNA-30e mediated cardioprotection of ACE2 in rats with doxorubicin-induced heart failure through inhibiting cardiomyocytes autophagy. Life Sci. 2017;169:69–75. doi: 10.1016/j.lfs.2016.09.006. [DOI] [PubMed] [Google Scholar]
- 57.Ouyang S., Chen W., Zeng G., Lei C., Tian G., Zhu M., Liu Y., Yang M. MicroRNA-183-3p up-regulated by vagus nerve stimulation mitigates chronic systolic heart failure via the reduction of BNIP3L-mediated autophagy. Gene. 2020;726:144136. doi: 10.1016/j.gene.2019.144136. [DOI] [PubMed] [Google Scholar]
- 58.Su M., Wang J., Wang C., Wang X., Dong W., Qiu W., Wang Y., Zhao X., Zou Y., Song L. MicroRNA-221 inhibits autophagy and promotes heart failure by modulating the p27/CDK2/mTOR axis. Cell Death Differ. 2015;22:986–999. doi: 10.1038/cdd.2014.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Su M., Chen Z., Wang C., Song L., Zou Y., Zhang L., Hui R., Wang J. Cardiac-specific overexpression of miR-222 induces heart failure and inhibits autophagy in mice. Cell. Physiol. Biochem. 2016;39:1503–1511. doi: 10.1159/000447853. [DOI] [PubMed] [Google Scholar]
- 60.Song L., Su M., Wang S., Zou Y., Wang X., Wang Y., Cui H., Zhao P., Hui R., Wang J. miR-451 is decreased in hypertrophic cardiomyopathy and regulates autophagy by targeting TSC1. J. Cell. Mol. Med. 2014;18:2266–2274. doi: 10.1111/jcmm.12380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chen C., Yang S., Li H., Yin Z., Fan J., Zhao Y., Gong W., Yan M., Wang D.W. Mir30c is involved in diabetic cardiomyopathy through regulation of cardiac autophagy via BECN1. Mol. Ther. Nucleic Acids. 2017;7:127–139. doi: 10.1016/j.omtn.2017.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.d’Avenia M., Citro R., De Marco M., Veronese A., Rosati A., Visone R., Leptidis S., Philippen L., Vitale G., Cavallo A. A novel miR-371a-5p-mediated pathway, leading to BAG3 upregulation in cardiomyocytes in response to epinephrine, is lost in Takotsubo cardiomyopathy. Cell Death Dis. 2015;6:e1948. doi: 10.1038/cddis.2015.280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ni T., Lin N., Lu W., Sun Z., Lin H., Chi J., Guo H. Dihydromyricetin prevents diabetic cardiomyopathy via miR-34a suppression by activating autophagy. Cardiovasc. Drugs Ther. 2020;34:291–301. doi: 10.1007/s10557-020-06968-0. [DOI] [PubMed] [Google Scholar]
- 64.Ouimet M., Ediriweera H., Afonso M.S., Ramkhelawon B., Singaravelu R., Liao X., Bandler R.C., Rahman K., Fisher E.A., Rayner K.J. microRNA-33 regulates macrophage autophagy in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2017;37:1058–1067. doi: 10.1161/ATVBAHA.116.308916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Geng Z., Xu F., Zhang Y. miR-129-5p-mediated Beclin-1 suppression inhibits endothelial cell autophagy in atherosclerosis. Am. J. Transl. Res. 2016;8:1886–1894. [PMC free article] [PubMed] [Google Scholar]
- 66.Chen H., Liu Gao M.Y., Zhang L., He F.L., Shi Y.K., Pan X.H., Wang H. MicroRNA-155 affects oxidative damage through regulating autophagy in endothelial cells. Oncol. Lett. 2019;17:2237–2243. doi: 10.3892/ol.2018.9860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Menghini R., Casagrande V., Marino A., Marchetti V., Cardellini M., Stoehr R., Rizza S., Martelli E., Greco S., Mauriello A. miR-216a: a link between endothelial dysfunction and autophagy. Cell Death Dis. 2014;5:e1029. doi: 10.1038/cddis.2013.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang H., Bei Y., Shen S., Huang P., Shi J., Zhang J., Sun Q., Chen Y., Yang Y., Xu T. miR-21-3p controls sepsis-associated cardiac dysfunction via regulating SORBS2. J. Mol. Cell. Cardiol. 2016;94:43–53. doi: 10.1016/j.yjmcc.2016.03.014. [DOI] [PubMed] [Google Scholar]
- 69.Sciarretta S., Yee D., Nagarajan N., Bianchi F., Saito T., Valenti V., Tong M., Del Re D.P., Vecchione C., Schirone L. Trehalose-induced activation of autophagy improves cardiac remodeling after myocardial infarction. J. Am. Coll. Cardiol. 2018;71:1999–2010. doi: 10.1016/j.jacc.2018.02.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ye X., Zhou X.J., Zhang H. Exploring the role of autophagy-related gene 5 (ATG5) yields important insights into autophagy in autoimmune/autoinflammatory diseases. Front. Immunol. 2018;9:2334. doi: 10.3389/fimmu.2018.02334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Li J., Rohailla S., Gelber N., Rutka J., Sabah N., Gladstone R.A., Wei C., Hu P., Kharbanda R.K., Redington A.N. MicroRNA-144 is a circulating effector of remote ischemic preconditioning. Basic Res. Cardiol. 2014;109:423. doi: 10.1007/s00395-014-0423-z. [DOI] [PubMed] [Google Scholar]
- 72.Zhong J., Basu R., Guo D., Chow F.L., Byrns S., Schuster M., Loibner H., Wang X.H., Penninger J.M., Kassiri Z. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation. 2010;122:717–728, 718 pp. following 728. doi: 10.1161/CIRCULATIONAHA.110.955369. [DOI] [PubMed] [Google Scholar]
- 73.Oudit G.Y., Penninger J.M. Recombinant human angiotensin-converting enzyme 2 as a new renin-angiotensin system peptidase for heart failure therapy. Curr. Heart Fail. Rep. 2011;8:176–183. doi: 10.1007/s11897-011-0063-7. [DOI] [PubMed] [Google Scholar]
- 74.Maron B.J., Towbin J.A., Thiene G., Antzelevitch C., Corrado D., Arnett D., Moss A.J., Seidman C.E., Young J.B., American Heart Association. Council on Clinical Cardiology, Heart Failure and Transplantation Committee. Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups. Council on Epidemiology and Prevention Contemporary definitions and classification of the cardiomyopathies: an American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention. Circulation. 2006;113:1807–1816. doi: 10.1161/CIRCULATIONAHA.106.174287. [DOI] [PubMed] [Google Scholar]
- 75.Nandi S.S., Duryee M.J., Shahshahan H.R., Thiele G.M., Anderson D.R., Mishra P.K. Induction of autophagy markers is associated with attenuation of miR-133a in diabetic heart failure patients undergoing mechanical unloading. Am. J. Transl. Res. 2015;7:683–696. [PMC free article] [PubMed] [Google Scholar]
- 76.Sinning Ch., Keller T., Abegunewardene N., Kreitner K.-F., Münzel T., Blankenberg S. Blankenberg S. Tako-Tsubo syndrome: dying of a broken heart? Clin. Res. Cardiol. 2010;99:771–780. doi: 10.1007/s00392-010-0224-9. [DOI] [PubMed] [Google Scholar]
- 77.Kurisu S., Sato H., Kawagoe T., Ishihara M., Shimatani Y., Nishioka K., Kono Y., Umemura T., Nakamura S. Tako-tsubo-like left ventricular dysfunction with ST-segment elevation: a novel cardiac syndrome mimicking acute myocardial infarction. Am. Heart J. 2002;143:448–455. doi: 10.1067/mhj.2002.120403. [DOI] [PubMed] [Google Scholar]
- 78.Schrijvers D.M., De Meyer G.R., Martinet W. Autophagy in atherosclerosis: a potential drug target for plaque stabilization. Arterioscler. Thromb. Vasc. Biol. 2011;31:2787–2791. doi: 10.1161/ATVBAHA.111.224899. [DOI] [PubMed] [Google Scholar]
- 79.Martinet W., De Meyer G.R. Autophagy in atherosclerosis: a cell survival and death phenomenon with therapeutic potential. Circ. Res. 2009;104:304–317. doi: 10.1161/CIRCRESAHA.108.188318. [DOI] [PubMed] [Google Scholar]
- 80.Zhang Z., Pan X., Yang S., Ma A., Wang K., Wang Y., Li T., Liu S. miR-155 promotes ox-LDL-induced autophagy in human umbilical vein endothelial cells. Mediators Inflamm. 2017;2017:9174801. doi: 10.1155/2017/9174801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zhu G.F., Yang L.X., Guo R.W., Liu H., Shi Y.K., Wang H., Ye J.S., Yang Z.H., Liang X. miR-155 inhibits oxidized low-density lipoprotein-induced apoptosis of RAW264.7 cells. Mol. Cell. Biochem. 2013;382:253–261. doi: 10.1007/s11010-013-1741-4. [DOI] [PubMed] [Google Scholar]
- 82.Zhang E., Zhao X., Zhang L., Li N., Yan J., Tu K., Yan R., Hu J., Zhang M., Sun D., Hou L. Minocycline promotes cardiomyocyte mitochondrial autophagy and cardiomyocyte autophagy to prevent sepsis-induced cardiac dysfunction by Akt/mTOR signaling. Apoptosis. 2019;24:369–381. doi: 10.1007/s10495-019-01521-3. [DOI] [PubMed] [Google Scholar]
- 83.Rudiger A., Singer M. Mechanisms of sepsis-induced cardiac dysfunction. Crit. Care Med. 2007;35:1599–1608. doi: 10.1097/01.CCM.0000266683.64081.02. [DOI] [PubMed] [Google Scholar]
- 84.Gao J., Xu W., Wang J., Wang K., Li P. The role and molecular mechanism of non-coding RNAs in pathological cardiac remodeling. Int. J. Mol. Sci. 2017;18:608. doi: 10.3390/ijms18030608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wilusz J.E., Sunwoo H., Spector D.L. Long noncoding RNAs: functional surprises from the RNA world. Genes Dev. 2009;23:1494–1504. doi: 10.1101/gad.1800909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gu Y., Yang F., Xu R.M., Zhang Y.Y., Li Y., Liu S.X., Zhang G.X., Wang G.K., Ma L.P. Differential expression profile of long non-coding RNA in cardiomyocytes autophagy induced by angiotensin II. Cell Biol. Int. 2017;41:1076–1082. doi: 10.1002/cbin.10809. [DOI] [PubMed] [Google Scholar]
- 87.Huang Z., Ye B., Wang Z., Han J., Lin L., Shan P., Cai X., Huang W. Inhibition of lncRNA-HRIM increases cell viability by regulating autophagy levels during hypoxia/reoxygenation in myocytes. Cell. Physiol. Biochem. 2018;46:1341–1351. doi: 10.1159/000489149. [DOI] [PubMed] [Google Scholar]
- 88.Zhou M., Zou Y.G., Xue Y.Z., Wang X.H., Gao H., Dong H.W., Zhang Q. Long non-coding RNA H19 protects acute myocardial infarction through activating autophagy in mice. Eur. Rev. Med. Pharmacol. Sci. 2018;22:5647–5651. doi: 10.26355/eurrev_201809_15831. [DOI] [PubMed] [Google Scholar]
- 89.Yin G., Yang X., Li Q., Guo Z. GATA1 activated lncRNA (Galont) promotes anoxia/reoxygenation-induced autophagy and cell death in cardiomyocytes by sponging miR-338. J. Cell. Biochem. 2018;119:4161–4169. doi: 10.1002/jcb.26623. [DOI] [PubMed] [Google Scholar]
- 90.Kong F., Jin J., Lv X., Han Y., Liang X., Gao Y., Duan X. Long noncoding RNA RMRP upregulation aggravates myocardial ischemia-reperfusion injury by sponging miR-206 to target ATG3 expression. Biomed. Pharmacother. 2019;109:716–725. doi: 10.1016/j.biopha.2018.10.079. [DOI] [PubMed] [Google Scholar]
- 91.Su Q., Liu Y., Lv X.W., Ye Z.L., Sun Y.H., Kong B.H., Qin Z.B. 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. 2019;133:12–25. doi: 10.1016/j.yjmcc.2019.05.021. [DOI] [PubMed] [Google Scholar]
- 92.Li Z., Zhang Y., Ding N., Zhao Y., Ye Z., Shen L., Yi H., Zhu Y. Inhibition of lncRNA XIST improves myocardial I/R injury by targeting miR-133a through inhibition of autophagy and regulation of SOCS2. Mol. Ther. Nucleic Acids. 2019;18:764–773. doi: 10.1016/j.omtn.2019.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wang J.J., Bie Z.D., Sun C.F. Long noncoding RNA AK088388 regulates autophagy through miR-30a to affect cardiomyocyte injury. J. Cell. Biochem. 2019;120:10155–10163. doi: 10.1002/jcb.28300. [DOI] [PubMed] [Google Scholar]
- 94.Liang H., Su X., Wu Q., Shan H., Lv L., Yu T., Zhao X., Sun J., Yang R., Zhang L. lncRNA 2810403D21Rik/Mirf promotes ischemic myocardial injury by regulating autophagy through targeting Mir26a. Autophagy. 2020;16:1077–1091. doi: 10.1080/15548627.2019.1659610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wang S., Yao T., Deng F., Yu W., Song Y., Chen J., Ruan Z. lncRNA MALAT1 promotes oxygen-glucose deprivation and reoxygenation induced cardiomyocytes injury through sponging miR-20b to enhance beclin1-mediated autophagy. Cardiovasc. Drugs Ther. 2019;33:675–686. doi: 10.1007/s10557-019-06902-z. [DOI] [PubMed] [Google Scholar]
- 96.Guo X., Wu X., Han Y., Tian E., Cheng J. lncRNA MALAT1 protects cardiomyocytes from isoproterenol-induced apoptosis through sponging miR-558 to enhance ULK1-mediated protective autophagy. J. Cell. Physiol. 2019;234:10842–10854. doi: 10.1002/jcp.27925. [DOI] [PubMed] [Google Scholar]
- 97.Hu H., Wu J., Yu X., Zhou J., Yu H., Ma L. Long non-coding RNA MALAT1 enhances the apoptosis of cardiomyocytes through autophagy inhibition by regulating TSC2-mTOR signaling. Biol. Res. 2019;52:58. doi: 10.1186/s40659-019-0265-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wu D., Zhou Y., Fan Y., Zhang Q., Gu F., Mao W., Zhang M. lncRNA CAIF was downregulated in end-stage cardiomyopathy and is a promising diagnostic and prognostic marker for this disease. Biomarkers. 2019;24:735–738. doi: 10.1080/1354750X.2019.1677778. [DOI] [PubMed] [Google Scholar]
- 99.Zhang C., Pan S., Aisha A., Abudoukelimu M., Tang L., Ling Y. Recombinant human brain natriuretic peptide regulates PI3K/AKT/mTOR pathway through lncRNA EGOT to attenuate hypoxia-induced injury in H9c2 cardiomyocytes. Biochem. Biophys. Res. Commun. 2018;503:1186–1193. doi: 10.1016/j.bbrc.2018.07.023. [DOI] [PubMed] [Google Scholar]
- 100.Tong G., Wang Y., Xu C., Xu Y., Ye X., Zhou L., Zhu G., Zhou Z., Huang J. Long non-coding RNA FOXD3-AS1 aggravates ischemia/reperfusion injury of cardiomyocytes through promoting autophagy. Am. J. Transl. Res. 2019;11:5634–5644. [PMC free article] [PubMed] [Google Scholar]
- 101.Viereck J., Kumarswamy R., Foinquinos A., Xiao K., Avramopoulos P., Kunz M., Dittrich M., Maetzig T., Zimmer K., Remke J. Long noncoding RNA Chast promotes cardiac remodeling. Sci. Transl. Med. 2016;8:326ra22. doi: 10.1126/scitranslmed.aaf1475. [DOI] [PubMed] [Google Scholar]
- 102.Zeng Z., Pan Y., Wu W., Li L., Wu Z., Zhang Y., Deng B., Wei S., Zhang W., Lin F., Song Y. Myocardial hypertrophy is improved with berberine treatment via long non-coding RNA MIAT-mediated autophagy. J. Pharm. Pharmacol. 2019;71:1822–1831. doi: 10.1111/jphp.13170. [DOI] [PubMed] [Google Scholar]
- 103.Feng Y., Xu W., Zhang W., Wang W., Liu T., Zhou X. lncRNA DCRF regulates cardiomyocyte autophagy by targeting miR-551b-5p in diabetic cardiomyopathy. Theranostics. 2019;9:4558–4566. doi: 10.7150/thno.31052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Yu S.Y., Dong B., Fang Z.F., Hu X.Q., Tang L., Zhou S.H. Knockdown of lncRNA AK139328 alleviates myocardial ischaemia/reperfusion injury in diabetic mice via modulating miR-204-3p and inhibiting autophagy. J. Cell. Mol. Med. 2018;22:4886–4898. doi: 10.1111/jcmm.13754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ma M., Hui J., Zhang Q.Y., Zhu Y., He Y., Liu X.J. 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. 2018;277:113–122. doi: 10.1016/j.atherosclerosis.2018.08.031. [DOI] [PubMed] [Google Scholar]
- 106.Zhu Y., Yang T., Duan J., Mu N., Zhang T. MALAT1/miR-15b-5p/MAPK1 mediates endothelial progenitor cells autophagy and affects coronary atherosclerotic heart disease via mTOR signaling pathway. Aging (Albany NY) 2019;11:1089–1109. doi: 10.18632/aging.101766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Foglio E., Puddighinu G., Germani A., Russo M.A., Limana F. HMGB1 inhibits apoptosis following MI and induces autophagy via mTORC1 inhibition. J. Cell. Physiol. 2017;232:1135–1143. doi: 10.1002/jcp.25576. [DOI] [PubMed] [Google Scholar]
- 108.Wei Y.M., Li X., Xu M., Abais J.M., Chen Y., Riebling C.R., Boini K.M., Li P.L., Zhang Y. Enhancement of autophagy by simvastatin through inhibition of Rac1-mTOR signaling pathway in coronary arterial myocytes. Cell. Physiol. Biochem. 2013;31:925–937. doi: 10.1159/000350111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Liu C.Y., Zhang Y.H., Li R.B., Zhou L.Y., An T., Zhang R.C., Zhai M., Huang Y., Yan K.W., Dong Y.H. lncRNA CAIF inhibits autophagy and attenuates myocardial infarction by blocking p53-mediated myocardin transcription. Nat. Commun. 2018;9:29. doi: 10.1038/s41467-017-02280-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ishii N., Ozaki K., Sato H., Mizuno H., Susumu Saito, Takahashi A., Miyamoto Y., Ikegawa S., Kamatani N., Hori M. Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction. J. Hum. Genet. 2006;51:1087–1099. doi: 10.1007/s10038-006-0070-9. [DOI] [PubMed] [Google Scholar]
- 111.Holdt L.M., Kohlmaier A., Teupser D. Molecular roles and function of circular RNAs in eukaryotic cells. Cell. Mol. Life Sci. 2018;75:1071–1098. doi: 10.1007/s00018-017-2688-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Huang R., Zhang Y., Han B., Bai Y., Zhou R., Gan G., Chao J., Hu G., Yao H. Circular RNA HIPK2 regulates astrocyte activation via cooperation of autophagy and ER stress by targeting MIR124-2HG. Autophagy. 2017;13:1722–1741. doi: 10.1080/15548627.2017.1356975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yang C., Wu S., Wu X., Zhou X., Jin S., Jiang H. Silencing circular RNA UVRAG inhibits bladder cancer growth and metastasis by targeting the microRNA-223/fibroblast growth factor receptor 2 axis. Cancer Sci. 2019;110:99–106. doi: 10.1111/cas.13857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhang X., Wang S., Wang H., Cao J., Huang X., Chen Z., Xu P., Sun G., Xu J., Lv J., Xu Z. Circular RNA circNRIP1 acts as a microRNA-149-5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol. Cancer. 2019;18:20. doi: 10.1186/s12943-018-0935-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Zhang Y., Li C., Liu X., Wang Y., Zhao R., Yang Y., Zheng X., Zhang Y., Zhang X. circHIPK3 promotes oxaliplatin-resistance in colorectal cancer through autophagy by sponging miR-637. EBioMedicine. 2019;48:277–288. doi: 10.1016/j.ebiom.2019.09.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Meng L., Liu S., Ding P., Chang S., Sang M. Circular RNA ciRS-7 inhibits autophagy of ESCC cells by functioning as miR-1299 sponge to target EGFR signaling. J. Cell. Biochem. 2020;121:1039–1049. doi: 10.1002/jcb.29339. [DOI] [PubMed] [Google Scholar]
- 117.Zhou L.Y., Zhai M., Huang Y., Xu S., An T., Wang Y.H., Zhang R.C., Liu C.Y., Dong Y.H., Wang M. The circular RNA ACR attenuates myocardial ischemia/reperfusion injury by suppressing autophagy via modulation of the Pink1/ FAM65B pathway. Cell Death Differ. 2019;26:1299–1315. doi: 10.1038/s41418-018-0206-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ren Q., Li H., Wang X. The circular RNA ZNF292 alleviates OGD-induced injury in H9c2 cells via targeting BNIP3. Cell Cycle. 2019;18:3365–3377. doi: 10.1080/15384101.2019.1676585. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 119.Poller W., Tank J., Skurk C., Gast M. Cardiovascular RNA interference therapy: the broadening tool and target spectrum. Circ. Res. 2013;113:588–602. doi: 10.1161/CIRCRESAHA.113.301056. [DOI] [PubMed] [Google Scholar]
- 120.Roberts T.C., Wood M.J. Therapeutic targeting of non-coding RNAs. Essays Biochem. 2013;54:127–145. doi: 10.1042/bse0540127. [DOI] [PubMed] [Google Scholar]
- 121.Roncarati R., Viviani Anselmi C., Losi M.A., Papa L., Cavarretta E., Da Costa Martins P., Contaldi C., Saccani Jotti G., Franzone A., Galastri L. Circulating miR-29a, among other up-regulated microRNAs, is the only biomarker for both hypertrophy and fibrosis in patients with hypertrophic cardiomyopathy. J. Am. Coll. Cardiol. 2014;63:920–927. doi: 10.1016/j.jacc.2013.09.041. [DOI] [PubMed] [Google Scholar]
- 122.Tijsen A.J., Creemers E.E., Moerland P.D., de Windt L.J., van der Wal A.C., Kok W.E., Pinto Y.M. miR423-5p as a circulating biomarker for heart failure. Circ. Res. 2010;106:1035–1039. doi: 10.1161/CIRCRESAHA.110.218297. [DOI] [PubMed] [Google Scholar]
- 123.Wang Y., Chang W., Zhang Y., Zhang L., Ding H., Qi H., Xue S., Yu H., Hu L., Liu D. Circulating miR-22-5p and miR-122-5p are promising novel biomarkers for diagnosis of acute myocardial infarction. J. Cell. Physiol. 2019;234:4778–4786. doi: 10.1002/jcp.27274. [DOI] [PubMed] [Google Scholar]
- 124.Ding H., An Y., Zhao T., Liu B., Wang Y., Zhang L., Wang Y., Zhang Y., Wang M., Dong Y. Large-scale rapid detection of circulating microRNAs in plasma for diagnosis and screening of specific diseases. Nanoscale. 2019;11:16879–16885. doi: 10.1039/c9nr04407h. [DOI] [PubMed] [Google Scholar]
- 125.Wong L.L., Armugam A., Sepramaniam S., Karolina D.S., Lim K.Y., Lim J.Y., Chong J.P., Ng J.Y., Chen Y.T., Chan M.M. Circulating microRNAs in heart failure with reduced and preserved left ventricular ejection fraction. Eur. J. Heart Fail. 2015;17:393–404. doi: 10.1002/ejhf.223. [DOI] [PubMed] [Google Scholar]
- 126.Wong L.L., Zou R., Zhou L., Lim J.Y., Phua D.C.Y., Liu C., Chong J.P.C., Ng J.Y.X., Liew O.W., Chan S.P. Combining circulating microRNA and NT-proBNP to detect and categorize heart failure subtypes. J. Am. Coll. Cardiol. 2019;73:1300–1313. doi: 10.1016/j.jacc.2018.11.060. [DOI] [PubMed] [Google Scholar]
- 127.Kumarswamy R., Bauters C., Volkmann I., Maury F., Fetisch J., Holzmann A., Lemesle G., de Groote P., Pinet F., Thum T. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circ. Res. 2014;114:1569–1575. doi: 10.1161/CIRCRESAHA.114.303915. [DOI] [PubMed] [Google Scholar]
- 128.Zhang Y., Zhang L., Wang Y., Ding H., Xue S., Qi H., Li P. MicroRNAs or long noncoding RNAs in diagnosis and prognosis of coronary artery disease. Aging Dis. 2019;10:353–366. doi: 10.14336/AD.2018.0617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kitow J., Derda A.A., Beermann J., Kumarswarmy R., Pfanne A., Fendrich J., Lorenzen J.M., Xiao K., Bavendiek U., Bauersachs J., Thum T. Mitochondrial long noncoding RNAs as blood based biomarkers for cardiac remodeling in patients with hypertrophic cardiomyopathy. Am. J. Physiol. Heart Circ. Physiol. 2016;311:H707–H712. doi: 10.1152/ajpheart.00194.2016. [DOI] [PubMed] [Google Scholar]
- 130.de Gonzalo-Calvo D., Kenneweg F., Bang C., Toro R., van der Meer R.W., Rijzewijk L.J., Smit J.W., Lamb H.J., Llorente-Cortes V., Thum T. Circulating long noncoding RNAs in personalized medicine: response to pioglitazone therapy in type 2 diabetes. J. Am. Coll. Cardiol. 2016;68:2914–2916. doi: 10.1016/j.jacc.2016.10.014. [DOI] [PubMed] [Google Scholar]
- 131.Hessels D., Klein Gunnewiek J.M., van Oort I., Karthaus H.F., van Leenders G.J., van Balken B., Kiemeney L.A., Witjes J.A., Schalken J.A. DD3PCA3-based molecular urine analysis for the diagnosis of prostate cancer. Eur. Urol. 2003;44:8–15, discussion 15–16. doi: 10.1016/s0302-2838(03)00201-x. [DOI] [PubMed] [Google Scholar]
- 132.Wang W., Wang Y., Piao H., Li B., Huang M., Zhu Z., Li D., Wang T., Xu R., Liu K. Circular RNAs as potential biomarkers and therapeutics for cardiovascular disease. PeerJ. 2019;7:e6831. doi: 10.7717/peerj.6831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Salgado-Somoza A., Zhang L., Vausort M., Devaux Y. The circular RNA MICRA for risk stratification after myocardial infarction. Int. J. Cardiol. Heart Vasc. 2017;17:33–36. doi: 10.1016/j.ijcha.2017.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Rupaimoole R., Slack F.J. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017;16:203–222. doi: 10.1038/nrd.2016.246. [DOI] [PubMed] [Google Scholar]
- 135.Setten R.L., Rossi J.J., Han S.P. The current state and future directions of RNAi-based therapeutics. Nat. Rev. Drug Discov. 2019;18:421–446. doi: 10.1038/s41573-019-0017-4. [DOI] [PubMed] [Google Scholar]
- 136.Adams D., Gonzalez-Duarte A., O’Riordan W.D., Yang C.C., Ueda M., Kristen A.V., Tournev I., Schmidt H.H., Coelho T., Berk J.L. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N. Engl. J. Med. 2018;379:11–21. doi: 10.1056/NEJMoa1716153. [DOI] [PubMed] [Google Scholar]
- 137.Rayner K.J., Suárez Y., Dávalos A., Parathath S., Fitzgerald M.L., Tamehiro N., Fisher E.A., Moore K.J., Fernández-Hernando C. miR-33 contributes to the regulation of cholesterol homeostasis. Science. 2010;328:1570–1573. doi: 10.1126/science.1189862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Dávalos A., Goedeke L., Smibert P., Ramírez C.M., Warrier N.P., Andreo U., Cirera-Salinas D., Rayner K., Suresh U., Pastor-Pareja J.C. miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc. Natl. Acad. Sci. USA. 2011;108:9232–9237. doi: 10.1073/pnas.1102281108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Rayner K.J., Esau C.C., Hussain F.N., McDaniel A.L., Marshall S.M., van Gils J.M., Ray T.D., Sheedy F.J., Goedeke L., Liu X. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature. 2011;478:404–407. doi: 10.1038/nature10486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Goedeke L., Salerno A., Ramírez C.M., Guo L., Allen R.M., Yin X., Langley S.R., Esau C., Wanschel A., Fisher E.A. Long-term therapeutic silencing of miR-33 increases circulating triglyceride levels and hepatic lipid accumulation in mice. EMBO Mol. Med. 2014;6:1133–1141. doi: 10.15252/emmm.201404046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Ray K.K., Landmesser U., Leiter L.A., Kallend D., Dufour R., Karakas M., Hall T., Troquay R.P., Turner T., Visseren F.L. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N. Engl. J. Med. 2017;376:1430–1440. doi: 10.1056/NEJMoa1615758. [DOI] [PubMed] [Google Scholar]
- 142.Das A., Durrant D., Koka S., Salloum F.N., Xi L., Kukreja R.C. Mammalian target of rapamycin (mTOR) inhibition with rapamycin improves cardiac function in type 2 diabetic mice: potential role of attenuated oxidative stress and altered contractile protein expression. J. Biol. Chem. 2014;289:4145–4160. doi: 10.1074/jbc.M113.521062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Jung H., Lee E., Kim I., Song J.H., Kim G.J. Histone deacetylase inhibition has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy. Physiol. Res. 2019;68:727–737. doi: 10.33549/physiolres.934110. [DOI] [PubMed] [Google Scholar]
- 144.Eisenberg T., Abdellatif M., Schroeder S., Primessnig U., Stekovic S., Pendl T., Harger A., Schipke J., Zimmermann A., Schmidt A. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat. Med. 2016;22:1428–1438. doi: 10.1038/nm.4222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Xu X., Lu Z., Fassett J., Zhang P., Hu X., Liu X., Kwak D., Li J., Zhu G., Tao Y. Metformin protects against systolic overload-induced heart failure independent of AMP-activated protein kinase α2. Hypertension. 2014;63:723–728. doi: 10.1161/HYPERTENSIONAHA.113.02619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Salabei J.K., Conklin D.J. Cardiovascular autophagy: crossroads of pathology, pharmacology and toxicology. Cardiovasc. Toxicol. 2013;13:220–229. doi: 10.1007/s12012-013-9200-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Yang Y., Liang C. MicroRNAs: an emerging player in autophagy. ScienceOpen Res. 2015;2015:2015. doi: 10.14293/S2199-1006.1.SOR-LIFE.A181CU.v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Ebadi N., Ghafouri-Fard S., Taheri M., Arsang-Jang S., Parsa S.A., Omrani M.D. Dysregulation of autophagy-related lncRNAs in peripheral blood of coronary artery disease patients. Eur. J. Pharmacol. 2020;867:172852. doi: 10.1016/j.ejphar.2019.172852. [DOI] [PubMed] [Google Scholar]
- 149.Wang X., Wang L., Ma Z., Liang W., Li J., Li Y., Gui Y., Ai S. Early expressed circulating long noncoding RNA CHAST is associated with cardiac contractile function in patients with acute myocardial infarction. Int. J. Cardiol. 2020;302:15–20. doi: 10.1016/j.ijcard.2019.12.058. [DOI] [PubMed] [Google Scholar]



