Open Access

CircRNAs open a new era in the study of cardiovascular disease (Review)

  • Authors:
    • Zhikang Yu
    • Qingyan Huang
    • Qunji Zhang
    • Heming Wu
    • Zhixiong Zhong
  • View Affiliations

  • Published online on: November 18, 2020     https://doi.org/10.3892/ijmm.2020.4792
  • Pages: 49-64
  • Copyright: © Yu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Circular RNAs (circRNAs) are non‑coding RNAs that are found in the cytoplasm or stored in the exosomes, where they are not affected by RNA exonucleases. CircRNAs are widely expressed in mammalian tissues and cells. A number of studies have suggested that circRNAs are associated with the physiology and pathology of cardiovascular diseases (CVDs). Therefore, circRNAs have been considered as effector molecules and biomarkers in the cardiovascular system. The present review article summarizes the biological origin and roles of circRNAs as well as the available databases and research methods for their identification. Furthermore, it describes their regulatory mechanisms in cardiovascular physiology and pathology, including the regulation of atherosclerosis, immunity, cell proliferation, apoptosis and autophagy. In addition, the current review discusses the unresolved problems in circRNA research and the application of circRNAs in the treatment of CVDs. Finally, the CVD‑associated circRNAs are also reviewed.

1. Introduction

It is estimated that only 20% of the nucleic acids in the human genome are encoded into proteins, while the rest, the so called non-coding nucleic acids, are considered as the 'dark matter' of the genome (1). However, several studies have shown that these non-coding RNAs are involved in important biological processes, such as cell proliferation, differentiation, apoptosis, metabolism, senescence, and especially in post-transcriptional regulation (1-3). Circular RNAs (circRNAs) were first discovered in the early 1970s (4,5), however, they have been poorly studied for nearly three decades due to the limited available technology. Recent advances in RNA sequencing technologies have promoted the identification of an increasing number of circRNAs (3,6-8). Several studies have shown that circRNAs can compete with mRNAs for the target binding sites on microRNAs (miRNAs) to affect gene expression. However, the function of the majority of circRNAs remains unclear. In 2013, Hansen et al (9) reported the overlapping co-expression of circRNA ciRS-7 and miR-7 in murine brain tissues. In addition, it was also demonstrated that the circRNA sex-determining region Y-9 (SRY-9) could act as a sponge for miR-138, thus providing the first solid evidence for the biological roles of circRNAs. To date, >183,000 circRNAs have been identified from human transcripts, to the best of our knowledge (10). The role of circRNAs in different tissues and different diseases have also been gradually revealed (11). The cardiovascular system is considered as one of the two major vascular systems in the human body. Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. A study found that the circRNAs solute carrier family 8 member A1 (SLC8A1), ataxin 10 (ATXN10), SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily A member 5 (SMARCA5), chromodomain Y (CDY) and myoblast determination protein 1 (MyoD) are associated with myocardial differentiation (12). In addition, several circRNAs are involved in the pathological process of atherosclerosis (AS) (13). Furthermore, stress stimuli such as hypoxia and high temperature have been associated with the expression of cardiovascular-related factors (14-16). Emerging evidence has shown that circRNAs are involved in the progression of CVD, and are therefore considered promising targets for diagnosis and treatment. The present review article aimed to summarize the current knowledge on the biological aspects of circRNAs and highlight the current research progress on the mechanisms underlying the function of circRNAs in the physiology and pathology of the cardiovascular system.

2. Formation and classification of circRNAs

It is considered that circRNAs are derived from the variable splicing of precursor mRNAs (pre-mRNA), mediated by RNA polymerase II (17-19). However, to the best of our knowledge, the mechanism underlying the formation of circRNAs remains to be elucidated.

CircRNAs can be cyclized by RNA binding protein (RBP) (20). For example, the RNA binding proteins, mannose-binding lectin (17), RBP quaking (21) and fused in sarcoma (22) are known to drive the folding and formation of precursor mRNAs of circRNAs. The aforementioned RBPs bind to introns near the splicing site to promote the formation of dimers upstream and downstream of the cyclic exons to aid their splicing, thus leading to the production of circRNAs.

CircRNAs may also be formed by complementary pairing of flanking introns located in dorsal exons, such as short interspersed nuclear elements or non-repetitive complementary sequences (23,24). Among them, Alu repetitive elements are widespread in the expressed RNA and in the non-coding parts (25,26). Alu repeats serve a key role in the formation of circRNAs, with >1 million copies in the genome (24). It is considered that the probability and intensity of the reverse intron folding, mediated by the repeats encompassed in the pre-mRNA, may also determine the frequency of reverse splicing (25,27).

Lariat-driven circularization is another important process of circRNA formation (28). Exon skipping occurs when the pre-mRNA is spliced, resulting in a lariat intermediate containing exons and introns. Subsequently, the internal splicing of the lariat leads to the removal or retention of introns, thus resulting in the formation of exon or exon-intron circRNAs (29-32).

Based on their biogenesis, circRNAs are divided into exon circRNAs, encompassing a single exon or multiple exons, exon-intron circRNAs, and intron circRNAs (Fig. 1). The majority of circRNAs belong to the first subtype and mainly exist in the cytoplasm (33). However, a few circRNAs are produced in the nucleus and can be secreted into the body fluids, such as the serum, saliva and cerebrospinal fluid (34-36). The exon-intron circRNAs are mainly located in the nucleus and interact with RNA polymerase II to promote the transcription of their parental genes (37). Finally, the intron circRNAs exist in the nucleus (38).

3. Biological functions of circRNAs

Gradually, circRNAs have been found to exert more specific biological functions, and their mechanism of action remains in the focus of attention (39,40). To date, several functions of circRNAs have been verified (Fig. 1).

MicroRNA sponges

Apart from a small number of circRNAs generated by intron circularization, which are located in the nucleus, the majority are mainly located in the cytoplasm (41). Several studies have shown that circRNAs can compete with mRNAs for binding to miRNAs located in the cytoplasm, thereby regulating the expression of mRNAs (3,9). ciRS-7/cerebellar degeneration-related protein 1 antisense RNA (CDR1as) sponging miR-7 and mouse SRY sponging miR-138 are considered as the most representative circRNAs supporting this biological function (3). Notably, only a few circRNAs have been identified to encompass multiple target binding sites for miRNAs (42). Furthermore, the binding of circRNAs by miRNAs may initiate circRNA decay (43). It has been also demonstrated that the CDR1as-miR-7 axis is modulated by Cyrano, a long non-coding RNA (lncRNA) (44). In the non-coding regulatory network, miR-671 shares a highly complementary sequence with CDR1as, which triggers the rare argonaute2-mediated RNA cleavage targeted by miRNAs in mammals and even vertebrates (43). The aforementioned findings indicate that circRNA-miRNA interactions not only mediate miRNA sequestration, but also exert other functions that are worth studying.

Competition with pre-mRNAs for cleavage and splicing during transcription

CircRNAs are formed as a result of the pre-mRNA atypical splicing, whereas mRNA is the final product of the pre-mRNA typical linear splicing (45). A study revealed that improving the efficiency of typical linear splicing could lead to a significant reduction in the number of generated circRNAs (17). When the length of the intron flanking the exon is longer, the efficiency of typical linear splicing is significantly decreased, and cyclization occurs (46). These findings indicate that circRNAs can compete with pre-mRNA during transcription.

CircRNAs regulate transcription and chromatin interactions, and act as protein sponges

CircRNAs located in the nucleus can bind RNA polymerase II or U1 small nuclear ribonucleoprotein to regulate the transcription of their parental genes (47,48). A study has shown that the expression of the parental genes is significantly attenuated when circRNAs are knocked out (47). Furthermore, back-splicing was observed in RNAs transcribed from centromeric retrotransposons in maize, while the resulting circular CRM1 RNAs could bind to maize centromeres through R-loops to promote the formation of chromatin loops. Previous studies have demonstrated that QKI attenuated doxorubicin (DOX)-induced cardiotoxicity via binding to titin (Ttn)-, formin homology 2 domain containing 3 (Fhod3)- and striatin 3 (Strn3)-derived circRNAs expressed in the heart (49). In addition, circRNAs contain binding sites for the RBPs of the host and can regulate their expression. For example, circRNA zinc finger protein 609 (ZNF609) can regulate the proportion of phosphorylated (p)-Rb/Rb and the levels of p-protein kinase B (Akt), thus affecting the G1/S phase progression in cells (50).

Translation of circRNAs

CircRNAs were originally defined as non-coding RNAs (51). However, it has been reported that translation may be carried out by the internal ribosome entry site (IRES) of circRNAs. For example, the untranslated region of endogenous circRNA ZNF609 can act as an IRES in a splicing-dependent manner, suggesting that circRNA ZNF609 can drive IRES-dependent translation (52). A study revealed that consensus N6-methyladenosine (m6A) motifs are enriched in circRNAs, while a single m6A site is sufficient to initiate translation. This m6A-dependent translation requires eukaryotic translation initiation factor 4F and the m6A reader, YTH N6-methyladenosine RNA binding protein 3 (53). Additionally, m6A-dependent translation can be enhanced by methyltransferase like 3/14, inhibited by demethylase obesity-associated protein and is upregulated in response to heat shock (53). m6A-driven circRNA translation commonly occurs, and hundreds of endogenous circRNAs exert translational capability (53). The translation capacity of circRNAs is not considered as their common function, however, further study on this field may provide revolutionary changes.

4. Databases and testing methods for circRNAs

Several laboratories and institutions have developed specialized databases and analytical tools to study circRNAs (10,54-65) (Table I). These databases provide the convenience to further investigate the already identified circRNAs. RNA sequencing analysis of rRNA-depleted total RNAs and microarrays are currently applied for the discovery of novel circRNAs (66). Nevertheless, the verification of the predicted circRNAs by in vitro/in vivo experiments is always necessary. Several widely used molecular biology techniques have been applied to detect and verify circRNAs, including northern blot analysis, which is considered as the gold standard for circRNAs verification (3,30), reverse transcription (RT)-PCR, which is the most commonly used and basic verification technique (17), quantitative PCR which is used to identify and quantitatively measure differentially expressed circRNAs, droplet digital PCR, which is performed to quantify the expression of circRNAs in challenging samples such as plasma (67,68), and fluorescence in situ hybridization, which is used to detect circRNA localization and interactions. Furthermore, overexpression and silencing techniques are performed to investigate the role of circRNAs in vitro or in vivo. For example, for the overexpression experiments, following amplification of a target DNA sequence encompassing the characteristic flanking Alu sequences, the sequence is digested with a restriction endonuclease and ligated to the pEGFP-C1 vector. Subsequently, the overexpression plasmid is transfected into the corresponding cell sample, thus resulting in the overexpression of the circRNAs (69,70). Additionally, for silencing experiments, small interfering RNA (siRNA) sequences complementary to the back-splice junction are synthesized and the RNA-induced silencing complex specifically targets circRNA, leading to its cleavage and degradation, thus silencing its expression (52,71). Notably, the effectiveness of siRNAs depends on transfection efficiency, and these RNAs only temporarily inhibit the expression of their corresponding targets (72). However, short hairpin RNAs (shRNAs) or Ago shRNAs exert fewer off-target effects, thus resulting in more stable target-circRNA knockdown (73). The progress on studies concerning the effects of circRNAs will promote the merging of specific versions of databases to expand research in the field of circRNAs.

Table I

List of the main circRNA databases.

Table I

List of the main circRNA databases.

First author, yearDatabaseFunctionURLLast update(Refs.)
Glažar et al, 2014CircBaseFacilitates the identification of circRNA in sequencing datahttp://www.circbase.org2017(54)
Panda et al, 2018 CircInteractomePredicts the interaction between circRNA and RNA binding proteins and miRNAhttp://circinteractome.nia.nih.gov/2018(55)
Liu et al, 2016CircNetProvides expression data of circRNA and regulatory network of circRNA-miRNA in different tissueshttp://circnet.mbc.nctu.edu.tw2016(56)
Zheng et al, 2016Deep BaseProvides small RNAs, long non-coding RNAs and circRNAs for deep sequencing of different species and tissueshttp://rna.sysu.edu.cn/deepBase2016(57)
Liu et al, 2019CircbankComprehensive database with circRNAs standard naminghttp://www.circbank.cn2019(58)
Xia et al, 2018CSCDDatabase of specific circRNAs associated with cancerhttp://gb.whu.edu.cn/CSCD2016(59)
Ghosal et al, 2013Circ2TraitsCircRNAs database related to human diseaseshttp://gyanxet-beta.com/circdb2013(60)
Li et al, 2014StarBaseLarge-scale CLIP-Seq decoding the Interaction Networkshttp://starbase.sysu.edu.cn/starbase2/index.php2019(61)
Dong et al, 2018CIRCpediaAllows comparison of circRNA expression in tissues and cell lines and classification of alternative reverse splicing and alternative splicinghttp://www.picb.ac.cn/rnomics/circpedia/2018(10)
Wang, 2018HppRNAMeets the needs of large sample RNA-Seq system analysis for circRNAs researchhttps://sourceforge.net/projects/hpprna/2018(62)
Wu et al, 2020CircAtlas2.0Provides functional annotation and prioritization of circRNAs for further verification and functional researchhttp://circatlas.biols.ac.cn/2020(63)
Cai et al, 2020VirusCircBaseThe first comprehensive database of circular RNA molecules in viruseshttp://www.computationalbiology.cn/ViruscircBase/home.html2019(64)
Chen et al, 2016CircRNADbProvides comments on chromosome location, source gene, genome length, protein codinghttp://reprod.njmu.edu.cn/CircRNAdb2016(65)

[i] circRNA, circular RNA.

5. Expression of circRNAs in the heart

Several studies have reported the comprehensive expression profiles of circRNAs in rat, murine and human hearts (74-76), including the first study on the development and expression of circRNAs in human induced pluripotent stem cell-derived cardiomyocytes (76). A study reported for the first time a catalog of 575 candidate circRNAs in murine hearts. Among them, several candidate genes were found to be associated with different diseases (74). Furthermore, another study extensively compared human, rat and murine circRNA conservation in a specific tissue (75). Only 10% of these circRNAs were conserved among the three species. In addition, this study demonstrated that the overall circRNA expression was significantly decreased in the heart of adult animals compared with neonatal rats. The expression of circRNAs in the human heart was approximately twice the baseline level of adult rats and mice. A study analyzed the characteristics of circRNA expression in 12 human hearts, 25 mouse hearts and across a 28-day differentiation time-course from human embryonic stem cell-derived cardiomyocytes (76). A total of 15,318 and 3,017 circRNAs were identified in human and murine myocardial tissue, respectively. The expression abundance of these circRNAs was basically associated with their cognate linear RNA. The parental genes of circRNAs with the highest content were also considered as key cardiac genes, such as ryanodine receptor 2 (Ryr2), Ttn and dystrophin. Among them, circRNA SLC8A1-1 was the most abundantly expressed circRNA in the myocardium, whereas 402 different circRNA isoforms were expressed from the Ttn gene locus. These circRNAs were dynamically and highly expressed during heart development. In addition, it has been reported that the circRNAs SLC8A1, circ-calcium voltage-gated channel subunit α1 D, SPHKAP and α kinase 2 are also specifically expressed in the heart (77). Another study showed that the circRNAs ATXN10, SMARCA5, CDY, MYOD, SLC8A1, ATXN7 and PHF2 were involved in cardiomyocyte differentiation (12). Furthermore, a notable study investigated the changes in the expression of lncRNAs, circRNAs and protein-coding genes via recording the RNA sequencing data at sequential stages of cardiomyocyte differentiation (78). The stages of differentiation studied were the following: Undifferentiated cardiomyocytes, mesoderm cardiomyocytes, cardiac progenitors and differentiated cardiomyocytes. This study described the dynamic expression pattern of non-coding RNAs during cardiogenesis, where the expression of circRNA SLC8A1 was gradually upregulated during cardiac differentiation. CircRNAs are derived from exons containing annotated start codons, which are also termed AUG circRNAs (79). Jakobi et al (80) described the AUG circRNAs in a heart model system for the first time, to the best of our knowledge. The authors demonstrated that m6A methylation was enriched in AUG circRNAs. They also reported the potential negative regulatory effect of AUG circRNAs on the translation of host genes. A recent study demonstrated that 40 ribosome-associated cardiac circRNAs could be translated into microproteins with different biological functions (81). Among them, the circRNAs CASP8 and FADD like apoptosis regulator, SLC8A1, myosin binding protein C3 and Ryr2 were reported for the first time. Therefore, the research on circRNAs will open a new chapter in heart biology.

6. Expression of circRNAs in cells of the cardiovascular system

It is understood that cardiomyocytes play an important role in the cardiovascular system (75,82). It has been reported that several circRNAs, such as hsa_circ_0001879, hsa_circ_0004104 and hsa_circTCF25, can be isolated from peripheral blood mononuclear cells, serving as biomarkers of coronary artery disease (CAD) (83,84). Another study revealed that the expression of circANRIL subtypes were inversely correlated with the risk and the severity of AS (85). Furthermore, a recent study documented for the first time the association between the expression of circRNAs and atrial fibrillation (86). Therefore, a total of five circRNAs were identified with notable biological significance, including chr9:15474007-15490122, chr16:75445723-75448593, hsa_circ_0007256, chr12:56563313-56563992 and hsa_ circ_0003533. Regarding vascular endothelial cells, circRNAs may directly or indirectly affect the occurrence of CVDs through functional changes, such as endothelial cell apoptosis, endothelial-mesenchymal transition and angiogenesis (87-90). For example, in vitro results demonstrated that the expression of circRNA ZNF292 in human umbilical vein endothelial cells (HUVECs) was significantly upregulated, suggesting that and circRNA ZNF292 could exert pro-angiogenic effects (71). Li et al (91) reported that the expression of hsa_circ_0003575 and hsa_circ_0003204 were significantly increased in oxidized low-density lipoprotein (ox-LDL)-induced HUVECs. Furthermore, hsa_circ_0003575 silencing significantly inhibited apoptosis and promoted proliferation and angiogenesis of ox-LDL-induced HUVECs. Yang et al (92) found that the circRNA HECT, C2 and WW domain containing E3 ubiquitin protein ligase 2 induced the expression of autophagy-related 5 (ATG5) in methamphetamine- or lipopolysaccharide (LPS)-induced human brain microvascular endothelial cells via binding to miR-30d, and ATG5 was shown to affect the endothelial-mesenchymal transition in an autophagy-independent manner. Liu et al (93) revealed the mechanism underlying the microvascular dysfunction mediated by diabetes. This mechanism also mediates the biological changes in human retinal vascular endothelial cells following circRNA PWWP domain-containing protein 2A transfer from vascular pericytes to HUVECs through exosomes (93). Vascular smooth muscle cells (VSMCs) are another notable type of cardiovascular cells. A recent study reported that the circRNA low-density lipoprotein receptor-related protein 6 (Lrp6) can promote the phenotypic transformation of VSMCs via inhibiting miR-145, while silencing of circRNA Lrp6, using shRNA technology, attenuated intimal hyperplasia in a mouse carotid artery injury model (94). Additionally, Zheng et al (95) demonstrated that hsa_circ_000595 is upregulated in human aortic aneurysm patients and hypoxic VSMCs. In addition, hsa_circ_000595 can directly target cyclooxygenase 2, hypoxia-inducible factor 1α (HIF-1α) and nuclear factor-κB to promote its biological function in regulating cell apoptosis via targeting miR-19a. By contrast, hsa-circ-000595 knockdown significantly attenuated apoptosis (95). Sun et al (96) reported that angiotensin II inhibits apoptosis of mouse aortic smooth muscle cells through regulating the circNRG-1/miR-193b-5p/NRG-1 axis. Several circRNAs have been identified to regulate the phenotypic transformation of VSMCs, including WD repeat domain 77, diaphanous related formin 3 and actin α 2 (97,98). Regarding cardiomyocytes and cardiac fibroblasts, Gupta et al (49) demonstrated that QKI could inhibit DOX-induced cardiotoxicity via regulating the expression of the circRNAs Ttn, Fhod3 and Strn3. Furthermore, QKI could regulate circRNA expression to protect cardiomyocytes against adriamycin-induced injury in primary rat, H9C2 and HL-1 cardiomyocytes in vitro. CircRNAs are also considered as potential targets for treating cardiac fibrosis (99-101). Therefore, a study showed that the expression of circRNA_000203 of the myosin I9A transcripts was increased in the myocardium of diabetic mice and in angiotensin II-induced cardiac fibroblasts (102). It has been also revealed that the circRNA_000203/miR-26b-5p/collagen type I α 2 and connective tissue growth factor axes regulate the anti-fibrotic effects of cardiac fibroblasts (102). Wang et al (82) demonstrated that the mitochondrial fission and apoptosis-related circRNA (MFACR) could mediate cardiomyocyte mitochondrial fission and apoptosis via regulating the MFACR/miR-652-3p/mitochondrial trifunctional protein 18 axis. These findings provided novel knowledge for understanding the molecular events associated with mitochondrial fission. The circRNAs involved in the cardiovascular system are listed in Table II.

Table II

List of circRNAs associated with cardiovascular diseases.

Table II

List of circRNAs associated with cardiovascular diseases.

First author/s, yearType of diseaseCircRNAHost geneCardiovascular cell typesRegulation (in disease)FunctionRegulatory mechanism(Refs.)
Wang et al, 2016Cardiac hypertrophyCircHRCRPWWP2AMouse cardiomyocytesDownregulatedOverexpression attenuates cardiac hypertrophyHRCR sponges miR-223 to regulate ARC expression to prevent cardiac hypertrophy(69)
Lim et al, 2019CircSLc8a1SLc8a1Mouse cardiomyocytesUpregulatedKnockdown reduces myocardial hypertrophy caused by pressure overloadCircSLc8a1 acts as a sponge of miR-133a, which is related to cardiac hypertrophy(108)
Tang et al, 2017Circ_000203Myo9aNeonatal mouse ventricular cardiomyocytesUpregulatedOverexpression aggravates cardiac hypertrophyCircRNA_000203 can sponge miR-26b-5p and miR-140-3p, exacerbating cardiac hypertrophy(102)
Huang et al, 2019Myocardial infarctionCircNfixNFIXCardiomyocytesDownregulatedDownregulated expression promotes cardiomyocyte proliferation and angiogenesisDual function as a miR-214 sponge and interacts with YBX1 and NEDD4L to induce YBX1 degradation(107)
Garikipati et al, 2019CircFndc3bFndc3bMouse cardiac endothelial cells and human umbilical vein endothelial cellsDownregulatedOverexpression enhances endothelial cell function and reduces cardiomyocyte apoptosisOverexpression of circFndc3b can significantly reduce FUS levels and upregulate VEGF-A levels to promote cardiac neovascularization and improve remodeling after myocardial infarction(131)
Cai et al, 2019CircTtc3Ttc3Rat cardiac fibroblasts and cardiomyocytesUpregulatedOverexpression inhibits apoptosis of cardiomyocytesCirc-Ttc3 as a miR-15b-5p sponge can increase Arl2 expression and inhibit myocardial cell apoptosis induced by myocardial infarction(115)
Geng et al, 2016Circ-CDR1asCdr1Rat cardiomyocytesUpregulatedOverexpression in MCM cells promotes cell apoptosisCirc-CDR1as can bind to miR-7a, resulting in increased expression of PARP and SP1, which increases myocardial infarct size(104)
Zhou et al, 2019CircMFACRSmyd4Mouse cardiomyocytesUpregulatedOverexpression attenuates autophagy and cell death in cardiomyocytescircMFACR represses autophagy and myocardial infarction by targeting Pink1-mediated phosphorylation of FAM65B(128)
Zhu et al, 2019Myocardial fibrosisCircNFIBNfibMouse cardiomyocytesDownregulatedOverexpression inhibits the proliferation and differentiation of mouse myocardial fibroblastsCircNFIB as a sponge for miR-433 inhibits myocardial fibroblast proliferation induced by TGF-β(99)
Ni et al, 2019CircHIPK3Hipk3Cardiac fibroblastsUpregulatedOverexpression attenuates the proliferation and migration of cardiac fibrosisCircHIPK-3 as a sponge for miR-29b-3p changes the expression of α-SMA, COL1A1 and COL3A1 to regulate cardiac fibrosis(111)
Sun et al, 2020Circ_Las1lLas1lHuman cardiac fibroblastsDownregulatedOverexpression inhibits cardiac fibroblast proliferation and migration, and promotes apoptosisCirc_Las1l as a sponge of miR-125b promotes the expression of SFRP5, and inhibits the activation, proliferation and migration of cardiac fibroblasts(100)
Zhou and Yu, 2017CircRNA_ 010567ZswimDiabetic mouse cardiomyocytesUpregulatedSilencing inhibits the translation of fibrosiscircRNA_010567 silencing can upregulate miR-141 and downregulate TGF-β1 expression(101)
Holdt et al, 2016 AtherosclerosisCircANRILINK4HumanDownregulated monocyticOverexpression increases apoptosis and decreases proliferationCircANRIL governs pre-rRNA maturation and nucleolar stress through interaction with multiple RBPs(85)
Zhang et al, 2020Circ_0003204USP36Human aorta endothelial cellsUpregulatedEctopic expression inhibits proliferation, migration and tube formation of ox- LDL-induced human aorta endothelial cellsCirc_0003204 as a sponge of miR-370 promotes protein expression of TGFβR2 and its downstream phosphorylated-SMAD3 and exacerbate deactivation of HAECs in response to ox-LDL(109)
Chen et al, 2017CircWDR77WDR77Human vascular smooth muscle cellsUpregulatedSilencing inhibits vascular smooth muscle cells proliferation and migrationCircWDR77 regulates vascular smooth muscle cells proliferation and migration by targeting miR-124/FGF2(97)
Li et al, 2017Circ_0003575CHMP5Human umbilical vein endothelial cellsUnchangedSilencing enhances human umbilical vein endothelial cells proliferation and angiogenesisUnknown(91)
Zeng et al, 2017Cardiac senescenceCircAmotl1Amotl1Mouse cardiomyocytesDownregulated during agingEctopic expression induces primary cardiomyocyte proliferationCircAmotl1 induces Akt phosphorylation and phosphorylated-Akt nuclear transport by binding Akt and PDK1, and activates the Akt signaling pathway to promote cell survival and proliferation(130)
Du et al, 2017CircFOXO3FOXO3Mouse cardiomyocytesUpregulatedEctopic expression induces senescenceCircFOXO3 interacts with id-1, E2F1 and FAK to inhibit their effects and accelerate cell senescence(15)
Liu et al, 2017Coronary heart diseaseCircZNF609ZNF609Human umbilical vein endothelial cellsUpregulatedSilencing increases endothelial cell viability, proliferation, migration, and tube formationCircZNF609 sponges miR-615-5p leading to increased MEF2A expression to reduce migration, proliferation, and apoptosis of endothelial cells(90)
Hall et al, 2019CircLrp6Lrp6Mouse vascular smooth muscle cellsUnchangedSilencing prevents intimal hyperplasia in mouse carotids.Circ_LRP6 sponges miR-145 to inhibit its mediated regulation of vascular smooth muscle cell migration, proliferation, and differentiation(94)
Sun et al, 2019CircNrg1Nrg1MASMCsDownregulatedKnockdown inhibits the apoptosis of MASMCsAngiotensin II downregulates the expression of circNrg-1 and sponges miR-193b-5p to regulate the expression of Nrg-1 to inhibit the apoptosis of MASMCs(96)
Zou et al, 2018CircTCF25TCF25Human umbilical vein endothelial cellsDownregulatedDownregulated expression in coronary heart diseaseCircTCF25 is a miR-103 sponge. Coronary artery disease risk factors inhibit the expression of circTCF25 in human umbilical vein endothelial cells(84)

[i] circRNA, circular RNA; MASMCs, mouse aortic smooth muscle cells; miR, microRNA; ox-LDL, oxidized low-density lipoprotein; FOXO3, forkhead box O3.

7. Regulatory mechanisms of circRNAs on CVD

It is understood that circRNAs are involved in numerous pathological mechanisms underlying CVDs, such as AS, immune response, apoptosis, cell proliferation, autophagy, cell senescence and myocardial repair (103-109). Emerging evidence has suggested that circRNAs play an important role in the occurrence and development of CVD (110). The documented regulatory mechanisms of circRNAs on CVDs are listed in Fig. 2.

Role of circRNAs in cell proliferation

The proliferation of cells involved in the cardiovascular system plays an important role in the development of CVDs, such as hypertension, AS and restenosis (110). CircRNAs serve an important role in the development of CVDs (103,111). Shen et al (103) demonstrated that circRNA-0044073 is highly expressed in patients with AS. In addition, it was shown that circRNA-0044073 promotes the proliferation and invasion of human umbilical vein smooth muscle cells and HUVECs, and accelerates the development of AS via targeting miR-107 and activating the Janus kinase/signal transducer and activator of transcription. Wang et al (112) found that the circRNA homeodomain inter-acting protein kinase 3 could act as a sponge to inhibit miR-29a activity, resulting in increased insulin-like growth factor 1 expression, which in turn could attenuate the dysfunction of oxidative stress-induced cerebral microvascular endothelial cells (CMVECs). Additionally, the circRNA HIPK3 promoted the proliferation and survival of CMVECs under hypoxic conditions. Another study demonstrated that HIPK3 could sponge miR-29b-3p to regulate the proliferation, migration and development of cardiac fibroblasts in angiotensin II-induced myocardial fibrosis (111).

Role of circRNAs in cardiomyocyte apoptosis

Cardiomyocyte apoptosis is an important factor affecting the occurrence and development of CVDs (104,113-115). Geng et al (104) investigated the effect of the co-expression of circ-Cdr1as with miR-7a in mouse myocardial infarction injury model. Cdr1as over-expression upregulated the expression of poly (ADP-ribose) polymerase and specific protein 1 to promote cardiomyocyte apoptosis, but was then reversed by miR-7a overexpression. Li et al (113) suggested that reactive oxygen species could upregulate circ-NCX1, which in turn could promote cardiomyocyte apoptosis. Mechanistically, circ-NCX1 could competitively bind to miR-133a-3p to increase the expression levels of the pro-apoptotic protein gene cell death-inducing protein 1 (CDIP1). Therefore, circNCX1 knockdown reduced the expression of CDIP1 in murine cardiomyocytes and heart tissues, thereby improving the condition of myocardial ischemia-reperfusion injury. Several other circRNAs have been identified to play a role in promoting cardiomyocyte apoptosis, including circ-MFACR and circRNA_0010729 (82,114).

Role of circRNAs in the regulation of AS

As early as 2010, a study revealed that the INK4/ADP ribosylation factor (ARF) gene locus is adjacent to the chromosome 9p21, which is associated with CAD (116). Circ-ANRIL, the antisense transcript of the INK4/ARF gene, was shown to be regulated by a single nucleotide polymorphism in the 9p21 chromosome and is associated with the risk of AS. Song et al (117) demonstrated that circ-ANRIL downregulation could prevent coronary AS via attenuating vascular endothelial cell apoptosis and the expression of inflammatory factors. However, another study suggested that circ-ANRIL exerts a protective effect against AS (85). The proportion of circ-ANRIL/lncRNA ANRIL in patients with coronary heart disease is higher, and patients with increased expression of circ-ANRIL rarely develop coronary heart disease. Furthermore, circ-ANRIL could induce nucleolar stress and p53 activation in VSMCs and macrophages, thus inducing and inhibiting apoptosis and proliferation, respectively. Therefore, the role of circ-ANRIL in regulating AS should be further investigated. In addition, a study revealed that circ-DLG associated protein 4 (DLGAP4) is negatively associated with the risk of coronary heart disease (118). Therefore, it was demonstrated that circ-DLGAP4 was down-regulated in ox-LDL-induced foam cells in a time-dependent manner. In addition, circ-DLGAP4 could sponge endogenous miR-143 to reduce the size of the atherosclerotic plaque and macrophage infiltration in mice (119,120). This finding indicated that circ-DLGAP4 could be considered as a protective factor against monocyte-macrophage foaming, and as a potential inhibitory factor of AS plaque forming.

Role of circRNAs in the regulation of immune responses

CircRNAs are involved in innate immunity (121). A study showed that circ_0124644 could affect the progression of CAD through regulating the immune responses such as cell adhesion, therefore, it is considered as a biomarker for CAD (105). Another study investigated the expression of circRNAs in bone marrow-derived macrophages under two distinct polarization conditions, namely M1 macrophages stimulated with interferon-γ and LPS, and M2 macrophages induced by interleukin-4 (122). The results revealed an interaction between circRNA-010231 and miR-141-5p, miR-145a-5p, miR-1964-5p, miR-19b-2-5p and miR-6950-5p. circRNA-010231 regulated the expression of the aforementioned miRNAs via participating in the differentiation and polarization of macrophages, thereby regulating the immune responses. The circRNA Ras-GEF domain-containing family member 1B (RasGEF1B) predominantly exists in the cytoplasm of murine macrophages (123). Ng et al (123) demonstrated that the expression of intercellular adhesion molecule-1 (ICAM-1) is downregulated following circ-RasGEF1B knockout in LPS-induced macro-phages, indicating that ICAM-1 is a downstream regulator of circ-RasGEF1B-associated immune responses. In addition, circ-RasGEF1B mediates the immune responses via regulating the stability of mature ICAM-1 mRNAs. Additional studies have also reported the role of different circRNAs on the regulation of the immune responses (121,124,125). Therefore, the effects of circRNAs on the immune function in CVDs are worth further investigation.

Role of circRNAs in the regulation of autophagy

CircRNAs are involved in autophagy by regulating the transcription and post-transcriptional modifications of autophagy-related genes (126). Cell autophagy is considered as a double-edged sword in health and disease, while it exerts protective effects on restoring homeostasis, it has also been reported that excessive autophagy mediates cardiomyocyte death (127). Zhou et al (128) demonstrated that circacrosin (ACR) attenuates autophagy and cell death in cardiomyocytes. In addition, ACR protects against ischemia-reperfusion injury and reduces myocardial infarct sizes in a myocardial ischemia/reperfusion injury model. The same study revealed that circ-ACR could induce the expression of putative kinase 1 (PINK-1) via directly binding to DNA meth-yltransferase 3B (Dnmt3b), thus blocking the Dnmt3b-mediated DNA methylation of the PINK-1 promoter. Furthermore, PINK-1 could phosphorylate its downstream target, Rho family-interacting cell polarization regulator 2 on serine 46 and inhibit cardiac autophagy and cell death. Another study on autophagy demonstrated that circRNA_101237 acts as a sponge of let-7a-5p to regulate insulin like growth factor 2 mRNA binding protein 3 (IGF2BP3)-dependent autophagy (106). Therefore, circRNA_101237 downregulation decreases the expression of IGF2BP3, which in turn attenuates the apoptosis of primary cardiomyocytes and inhibits hypoxia/reoxygenation-induced autophagy. The aforementioned studies indicated that autophagy could serve an important role in the regulation of CVDs.

Role of circRNAs in the regulation of cell senescence and myocardial repair

The senescence and repair of tissues or cells is closely associated with their function (107,129-131). A study showed that the circRNA forkhead box O3 (FOXO3) is significantly upregulated in heart samples of elderly patients and mice and is associated with cell senescence (15). Therefore, circ-FOXO3 is mainly distributed in the cytoplasm, where it can interact with the anti-senescence protein inhibitor of DNA binding-1, the transcription factor E2F1 and the anti-stress proteins focal adhesion kinase and HIF-1α, thereby promoting cardiomyocyte senescence. Additional mechanistic studies suggested that the pro-senescent effect of circ-FOXO3 could be associated with the regulation of the cell cycle. Therefore, in this study, the ectopically expressed circ-FOXO3 could form a ternary complex by binding with cyclin kinase 2 (CDK2) and cycle dependent kinase inhibitor 1 (p21). The formed circ-FOXO3-p21-CDK2 ternary complex blocks the function of CDK2, thereby inhibiting the progression of the cell cycle (129). Zeng et al (130) investigated the role of circRNA in myocardial repair by screening the myocardial tissues of patients of different ages. The results revealed that circRNA angiomotin Like 1 (Amotl1) is significantly upregulated in myocardial tissues, thus enhancing the myocardial function in the neonatal heart. Furthermore, circ-Amotl1 induces the phosphorylation of Akt and its nuclear transportation via binding Akt and phosphoinositide dependent kinase 1. This process promotes the activation of the Akt signaling pathway, resulting in enhanced cell survival and proliferation. In addition, circ-Amotl1 could play a protective role in the DOX-induced cardiomyopathy (130). In another study, the authors investigated the positive and negative regulatory effects of the signaling pathways Meis1/circ-nuclear factor 1 X-type (Nfix)/miR-214/Gsk3β/β-catenin and circ-Nfix/Ybx1/cyclin A2 and cyclin B1 by functional verification experiments (107). The results revealed a close association between the expression of circ-Nfix and heart regeneration and repair. The aforementioned studies indicated that circRNAs could serve an important role in regulating cell senescence and cell regeneration, and could be used as targets for treating heart failure and myocardial infarction in the future.

8. Perspectives and conclusions

Nowadays, the interest of the scientific community regarding circRNAs is increasing with their comprehensive exploration, and the mystery of circRNAs has been slowly revealed. Although several functions of circRNAs have been identified, numerous remain to be elucidated. Due to their high stability and resistance to RNases, circRNAs may accumulate in quiescent or post-mitotic cells, such as neurons (132). Several studies revealed that the expression of circRNAs in proliferative cells and tissues, including cancer cells, is lower compared with terminally differentiated cells, indicating a negative association between circRNA abundance and cell proliferation (133). Previous studies have shown that circRNAs are enriched in extracellular vesicles and are secreted by cells in the form of vesicles (133-137). Notably, smaller circRNAs are more prone to vesicle-mediated transport (35). In addition, circRNAs maintain their circular structure in vesicles and perform their activities in recipient cells, suggesting that circRNA transport may be important for cell-to-cell communication (136). In cardiac metabolic diseases, serum markers in peripheral blood, such as cardiac troponin and creatine kinase, serve a significant role in the diagnosis of several diseases. It has been reported that certain linear non-coding RNAs can be used as biomarkers in the diagnosis of CAD (137,138). Therefore, circRNAs could be considered as potential biomarkers for the diagnosis and treatment of CVDs. However, whether circRNAs selectively enter into exosomes, and the mechanism and significance of exosomal circRNA formation should be verified first. Certain circRNAs accumulate in the nucleus, while the majority are effectively transported to the cytoplasm (41). The accumulation of circRNAs in the nucleus is mediated by the ATP-dependent RNA helicase DexH/D-box, and their nuclear export depends on the length of the mature circRNA (139). m6A modification widely occurs on circRNAs (140). In view of the influence of m6A-binding protein YTH domain-containing 1 (YTHDC1) on the nuclear export of methylated mRNA (141), the effect of m6A on the export of circRNAs remains to be elucidated, to the best of our knowledge. Several circRNA-mediated degradation mechanisms have been reported (121,142,143). Therefore, the binding of circRNAs by miRNAs can initiate circRNA decay (121). Furthermore, circRNA can also be degraded by the cytoplasmic endonuclease RNase L, which is activated in the presence of pathogenic double-strand RNA during viral infection (142). In addition, it has been reported that the YTHDF2-HRSP12-RNase P/MRP and UPF1-G3BP1 complexes mediate m6A-modified and high-structured circRNAs, respectively (143). However, to the best of our knowledge, how the degradation mechanisms of circRNAs affect the disease course remains to be investigated. It is understood that circRNAs are conserved among various species (75), and are specifically expressed in different tissues and different developmental stages (12,76). Nevertheless, the reasons for the conservatism and specificity of circRNAs and how they affect the functional development of tissues and organs have not been reported to date. Although it has been documented that circRNAs exert a folded secondary structure (144), to the best of our knowledge, the association between their secondary structure and their function remains unclear, and it is worthy of further investigation. There are a number of limited reports regarding the translational function of circRNAs, including the IRES and m6A modification pathways (52,53,81,145). Recently, researchers have combined the stability of circRNAs and their potential for continuous translation to develop a new expression system that could produce a protein in a sustainable and stable manner (146). This finding indicated that the translational function of circRNAs could be applied in the future for biological therapy.

A number of circRNAs have been identified, discovered and applied in the CAD (84,105,118). It has been suggested that active pharmaceutical ingredients can interfere with the differential expression of circRNAs in CAD (147). Several networks of circRNA-miRNA signaling pathways in the CAD have been also reported (103,148). These networks are systematic and three-dimensional and can truly and dynamically reflect the occurrence and development of CAD. A study revealed that circRNAs could be involved in chemoresistance, thus providing a novel approach for circRNAs in drug intervention and targeted therapy for CVDs (149). In addition, a research team used RNA polymers and poly (lactic-co-glycolic acid) (PLGA) coated stents containing and releasing therapeutic factors to treat local lesions in AS (149). The results demonstrated that these stents could promote endothelial cell regeneration and reduce the risk of coronary artery restenosis. Another study suggested that modern transfection methods could be used to transfer circRNAs during cardiovascular operations (150). Recently, a study introduced the circmiRs technology, opening a new chapter in the in vivo treatment of CAD using circRNAs (151). In this study, a circmiR sponge was constructed targeting the known cardiac pro-hypertrophic miRs-132 and miRs-212. Therefore, the hypertrophic characteristics of the disease were attenuated, and the cardiac function was preserved in transverse aortic constriction-treated mice, thus supporting the potential of circmiRs as novel therapeutic tools. By exploiting the characteristics of circRNAs in CVDs through drug intervention, artificial overexpression or knockout, and circRNA sequencing, it is possible to achieve a more accurate treatment strategy for CVDs.

Funding

The present study was supported by the Key Scientific and Technological Project of Meizhou People's Hospital (grant no. MPHKSTP-20180101 to Dr ZZ), the Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translation Research of Hakka Population (grant no. 2018B030322003) and the Science and Technology Program of Meizhou (grant no. 2019B0202001).

Availability of data and materials

Not applicable.

Authors' contributions

ZY, HW and ZZ contributed to the conception of the study, performed the literature search and wrote the manuscript. QH, QZ and HW edited the manuscript, assisted in the literature search and critically revised the article for important intellectual content. All the authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Pennisi E: Shining a light on the genome's 'dark matter'. Science. 330:16142010. View Article : Google Scholar : PubMed/NCBI

2 

Mattick JS and Makunin IV: Non-coding RNA. Hum Mol Genet. 15(Suppl 1): R17–R29. 2006. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Hsu MT and Coca-Prados M: Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature. 280:339–340. 1979. View Article : Google Scholar : PubMed/NCBI

5 

Sanger HL, Klotz G, Riesner D, Gross HJ and Kleinschmidt AK: Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci USA. 73:3852–3856. 1976. View Article : Google Scholar : PubMed/NCBI

6 

Salzman J, Chen RE, Olsen MN, Wang PL and Brown PO: Cell-type specific features of circular RNA expression. PLoS Genet. 9:e10037772013. View Article : Google Scholar : PubMed/NCBI

7 

Wang PL, Bao Y, Yee MC, Barrett SP, Hogan GJ, Olsen MN, Dinneny JR, Brown PO and Salzman J: Circular RNA is expressed across the eukaryotic tree of life. PLoS One. 9:e908592014. View Article : Google Scholar : PubMed/NCBI

8 

Jeck WR and Sharpless NE: Detecting and characterizing circular RNAs. Nat Biotechnol. 32:453–461. 2014. View Article : Google Scholar : PubMed/NCBI

9 

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

10 

Dong R, Ma XK, Li GW and Yang L: CIRCpedia v2: An updated database for comprehensive circular RNA annotation and expression comparison. Genomics Proteomics Bioinformatics. 16:226–233. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Han B, Chao J and Yao H: Circular RNA and its mechanisms in disease: From the bench to the clinic. Pharmacol Ther. 187:31–44. 2018. View Article : Google Scholar

12 

Siede D, Rapti K, Gorska AA, Katus HA, Altmüller J, Boeckel JN, Meder B, Maack C, Völkers M, Müller OJ, et al: Identification of circular RNAs with host gene-independent expression in human model systems for cardiac differentiation and disease. J Mol Cell Cardiol. 109:48–56. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Zhang F, Zhang R, Zhang X, Wu Y, Li X, Zhang S, Hou W, Ding Y, Tian J, Sun L and Kong X: Comprehensive analysis of circRNA expression pattern and circRNA-miRNA-mRNA network in the pathogenesis of atherosclerosis in rabbits. Aging (Albany NY). 10:2266–2283. 2018. View Article : Google Scholar

14 

Santos-Ribeiro D, Godinas L, Pilette C and Perros F: The inte-grated stress response system in cardiovascular disease. Drug Discov Today. 23:920–929. 2018. View Article : Google Scholar

15 

Du WW, Yang W, Chen Y, Wu ZK, Foster FS, Yang Z, Li X and Yang BB: Foxo3 circular RNA promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses. Eur Heart J. 38:1402–1412. 2017.

16 

Pan T, Sun X, Liu Y, Li H, Deng G, Lin H and Wang S: Heat stress alters genome-wide profiles of circular RNAs in Arabidopsis. Plant Mol Biol. 96:217–229. 2018. View Article : Google Scholar

17 

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

18 

Fu XD and Ares M Jr: Context-dependent control of alternative splicing by RNA-binding proteins. Nat Rev Genet. 15:689–701. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Starke S, Jost I, Rossbach O, Schneider T, Schreiner S, Hung LH and Bindereif A: Exon circularization requires canonical splice signals. Cell Rep. 10:103–111. 2015. View Article : Google Scholar

20 

Kramer MC, Liang D, Tatomer DC, Gold B, March ZM, Cherry S and Wilusz JE: Combinatorial control of drosophila circular RNA expression by intronic repeats, hnRNPs, and SR proteins. Genes Dev. 29:2168–2182. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Conn SJ, Pillman KA, Toubia J, Conn VM, Salmanidis M, Phillips CA, Roslan S, Schreiber AW, Gregory PA and Goodall GJ: The RNA binding protein quaking regulates formation of circRNAs. Cell. 160:1125–1134. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Errichelli L, Dini Modigliani S, Laneve P, Colantoni A, Legnini I, Capauto D, Rosa A, De Santis R, Scarfò R, Peruzzi G, et al: FUS affects circular RNA expression in murine embryonic stem cell-derived motor neurons. Nat Commun. 8:147412017. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Ivanov A, Memczak S, Wyler E, Torti F, Porath HT, Orejuela MR, Piechotta M, Levanon EY, Landthaler M, Dieterich C and Rajewsky N: Analysis of intron sequences reveals hallmarks of circular RNA biogenesis in animals. Cell Rep. 10:170–177. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, et al: Initial sequencing and analysis of the human genome. Nature. 409:860–921. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Sela N, Mersch B, Gal-Mark N, Lev-Maor G, Hotz-Wagenblatt A and Ast G: Comparative analysis of transposed element insertion within human and mouse genomes reveals Alu's unique role in shaping the human transcriptome. Genome Biol. 8:R1272007. View Article : Google Scholar : PubMed/NCBI

27 

Liang D and Wilusz JE: Short intronic repeat sequences facilitate circular RNA production. Genes Dev. 28:2233–2247. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Ruskin B and Green MR: An RNA processing activity that debranches RNA lariats. Science. 229:135–140. 1985. View Article : Google Scholar : PubMed/NCBI

29 

Aufiero S, van den Hoogenhof MMG, Reckman YJ, Beqqali A, van der Made I, Kluin J, Khan MAF, Pinto YM and Creemers EE: Cardiac circRNAs arise mainly from constitutive exons rather than alternatively spliced exons. RNA. 24:815–827. 2018. View Article : Google Scholar :

30 

Salzman J, Gawad C, Wang PL, Lacayo N and Brown PO: Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS One. 7:e307332012. View Article : Google Scholar :

31 

Talhouarne GJS and Gall JG: Lariat intronic RNAs in the cytoplasm of vertebrate cells. Proc Natl Acad Sci USA. 115:E7970–E7977. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Noto JJ, Schmidt CA and Matera AG: Engineering and expressing circular RNAs via tRNA splicing. RNA Biol. 14:978–984. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Pasman Z, Been MD and Garcia-Blanco MA: Exon circularization in mammalian nuclear extracts. RNA. 2:603–610. 1996.PubMed/NCBI

34 

Lyu D and Huang S: The emerging role and clinical implication of human exonic circular RNA. RNA Biol. 14:1000–1006. 2017. View Article : Google Scholar :

35 

Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, Chen D, Gu J, He X and Huang S: Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 25:981–984. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Zhao RT, Zhou J, Dong XL, Bi CW, Jiang RC, Dong JF, Tian Y, Yuan HJ and Zhang JN: Circular ribonucleic acid expression alteration in exosomes from the brain extracellular space after traumatic brain injury in mice. J Neurotrauma. 35:2056–2066. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Hu Q and Zhou T: EIciRNA-mediated gene expression: Tunability and bimodality. FEBS Lett. 592:3460–3471. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Panda AC, De S, Grammatikakis I, Munk R, Yang X, Piao Y, Dudekula DB, Abdelmohsen K and Gorospe M: High-purity circular RNA isolation method (RPAD) reveals vast collection of intronic circRNAs. Nucleic Acids Res. 45:e1162017. View Article : Google Scholar : PubMed/NCBI

39 

Piwecka M, Glažar P, Hernandez-Miranda LR, Memczak S, Wolf SA, Rybak-Wolf A, Filipchyk A, Klironomos F, Cerda Jara CA, Fenske P, et al: Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science. 357:eaam85262017. View Article : Google Scholar

40 

Ji P, Wu W, Chen S, Zheng Y, Zhou L, Zhang J, Cheng H, Yan J, Zhang S, Yang P and Zhao F: Expanded expression landscape and prioritization of circular RNAs in mammals. Cell Rep. 26:3444–3460.e5. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Kristensen LS, Andersen MS, Stagsted LVW, Ebbesen KK, Hansen TB and Kjems J: The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 20:675–691. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Guo JU, Agarwal V, Guo H and Bartel DP: Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 15:4092014. View Article : Google Scholar : PubMed/NCBI

43 

Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ and Kjems J: miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular anti-sense RNA. EMBO J. 30:4414–4422. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Kleaveland B, Shi CY, Stefano J and Bartel DP: A network of noncoding regulatory RNAs acts in the mammalian brain. Cell. 174:350–362.e17. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Chen LL: The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 17:205–211. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Liang D, Tatomer DC, Luo Z, Wu H, Yang L, Chen LL, Cherry S and Wilusz JE: The output of protein-coding genes shifts to circular RNAs when the pre-mRNA processing machinery is limiting. Mol Cell. 68:940–954.e3. 2017. View Article : Google Scholar : PubMed/NCBI

47 

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

48 

Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, Zhu S, Yang L and Chen LL: Circular intronic long noncoding RNAs. Mol Cell. 51:792–806. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Gupta SK, Garg A, Bär C, Chatterjee S, Foinquinos A, Milting H, Streckfuß-Bömeke K, Fiedler J and Thum T: Quaking inhibits doxorubicin-mediated cardiotoxicity through regulation of cardiac circular RNA expression. Circ Res. 122:246–254. 2018. View Article : Google Scholar :

50 

Rossi F, Legnini I, Megiorni F, Colantoni A, Santini T, Morlando M, Di Timoteo G, Dattilo D, Dominici C and Bozzoni I: Circ-ZNF609 regulates G1-S progression in rhabdomyosarcoma. Oncogene. 38:3843–3854. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Cardona-Monzonís A, García-Giménez JL, Mena-Mollá S, Pareja-Galeano H, de la Guía-Galipienso F, Lippi G, Pallardó FV and Sanchis-Gomar F: Non-coding RNAs and coronary artery disease. Adv Exp Med Biol. 1229:273–285. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, Fatica A, Santini T, Andronache A, Wade M, et al: Circ-ZNF609 is a circular RNA that can be translated and functions in myogenesis. Mol Cell. 66:22–37.e9. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, Jin Y, Yang Y, Chen LL, Wang Y, et al: Extensive translation of circular RNAs driven by N6-methyladenosine. Cell Res. 27:626–641. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Glažar P, Papavasileiou P and Rajewsky N: CircBase: A database for circular RNAs. RNA. 20:1666–1670. 2014. View Article : Google Scholar

55 

Panda AC, Dudekula DB, Abdelmohsen K and Gorospe M: Analysis of circular RNAs using the web tool CircInteractome. Methods Mol Biol. 1724:43–56. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Liu YC, Li JR, Sun CH, Andrews E, Chao RF, Lin FM, Weng SL, Hsu SD, Huang CC, Cheng C, et al: CircNet: A database of circular RNAs derived from transcriptome sequencing data. Nucleic Acids Res. 44(D1): D209–D215. 2016. View Article : Google Scholar :

57 

Zheng LL, Li JH, Wu J, Sun WJ, Liu S, Wang ZL, Zhou H, Yang JH and Qu LH: DeepBase v2.0: Identification, expression, evolution and function of small RNAs, LncRNAs and circular RNAs from deep-sequencing data. Nucleic Acids Res. 44(D1): D196–D202. 2016. View Article : Google Scholar :

58 

Liu M, Wang Q, Shen J, Yang BB and Ding X: Circbank: A comprehensive database for circRNA with standard nomenclature. RNA Biol. 16:899–905. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Xia S, Feng J, Chen K, Ma Y, Gong J, Cai F, Jin Y, Gao Y, Xia L, Chang H, et al: CSCD: A database for cancer-specific circular RNAs. Nucleic Acids Res. 46(D1): D925–D929. 2018. View Article : Google Scholar :

60 

Ghosal S, Das S, Sen R, Basak P and Chakrabarti J: Circ2Traits: A comprehensive database for circular RNA potentially associated with disease and traits. Front Genet. 4:2832013. View Article : Google Scholar : PubMed/NCBI

61 

Li JH, Liu S, Zhou H, Qu LH and Yang JH: starBase v2.0: Decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 42(Database Issue): D92–D97. 2014. View Article : Google Scholar

62 

Wang D: hppRNA-a snakemake-based handy parameter-free pipeline for RNA-Seq analysis of numerous samples. Brief Bioinform. 19:622–626. 2018.

63 

Wu W, Ji P and Zhao F: CircAtlas: An integrated resource of one million highly accurate circular RNAs from 1070 vertebrate transcriptomes. Genome Biol. 21:1012020. View Article : Google Scholar : PubMed/NCBI

64 

Cai Z, Fan Y, Zhang Z, Lu C, Zhu Z, Jiang T, Shan T and Peng Y: VirusCircBase: A database of virus circular RNAs. Brief Bioinform. Apr 29–2020.Epub ahead of print. View Article : Google Scholar

65 

Chen X, Han P, Zhou T, Guo X, Song X and Li Y: circRNADb: A comprehensive database for human circular RNAs with protein-coding annotations. Sci Rep. 6:349852016. View Article : Google Scholar : PubMed/NCBI

66 

Chen X, Chen RX, Wei WS, Li YH, Feng ZH, Tan L, Chen JW, Yuan GJ, Chen SL, Guo SJ, et al: PRMT5 circular RNA promotes metastasis of urothelial carcinoma of the bladder through sponging miR-30c to induce epithelial-mesenchymal transition. Clin Cancer Res. 24:6319–6330. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Li T, Shao Y, Fu L, Xie Y, Zhu L, Sun W, Yu R, Xiao B and Guo J: Plasma circular RNA profiling of patients with gastric cancer and their droplet digital RT-PCR detection. J Mol Med (Berl). 96:85–96. 2018. View Article : Google Scholar

68 

Chen DF, Zhang LJ, Tan K and Jing Q: Application of droplet digital PCR in quantitative detection of the cell-free circulating circRNAs. Biotechnol Biotechnol Equip. 32:116–123. 2018. View Article : Google Scholar

69 

Wang K, Long B, Liu F, Wang JX, Liu CY, Zhao B, Zhou LY, Sun T, Wang M, Yu T, et al: A circular RNA protects the heart from pathological hypertrophy and heart failure by targeting miR-223. Eur Heart J. 37:2602–2611. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Kamens J: The addgene repository: An international nonprofit plasmid and data resource. Nucleic Acids Res. 43(Database Issue): D1152–D1157. 2015. View Article : Google Scholar :

71 

Boeckel JN, Jaé N, Heumüller AW, Chen W, Boon RA, Stellos K, Zeiher AM, John D, Uchida S and Dimmeler S: Identification and characterization of hypoxia-regulated endothelial circular RNA. Circ Res. 117:884–890. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Bramsen JB, Laursen MB, Nielsen AF, Hansen TB, Bus C, Langkjaer N, Babu BR, Højland T, Abramov M, Van Aerschot A, et al: A large-scale chemical modification screen identifies design rules to generate siRNAs with high activity, high stability and low toxicity. Nucleic Acids Res. 37:2867–2881. 2009. View Article : Google Scholar : PubMed/NCBI

73 

de Bruyns A, Geiling B and Dankort D: Construction of modular lentiviral vectors for effective gene expression and knockdown. Methods Mol Biol. 1448:3–21. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Jakobi T, Czaja-Hasse LF, Reinhardt R and Dieterich C: Profiling and validation of the circular RNA repertoire in adult murine hearts. Genomics Proteomics Bioinformatics. 14:216–223. 2016. View Article : Google Scholar : PubMed/NCBI

75 

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

76 

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

77 

Lei W, Feng T, Fang X, Yu Y, Yang J, Zhao ZA, Liu J, Shen Z, Deng W and Hu S: Signature of circular RNAs in human induced pluripotent stem cells and derived cardiomyocytes. Stem Cell Res Ther. 9:562018. View Article : Google Scholar : PubMed/NCBI

78 

Li Y, Zhang J, Huo C, Ding N, Li J, Xiao J, Lin X, Cai B, Zhang Y and Xu J: Dynamic organization of lncRNA and circular RNA regulators collectively controlled cardiac differentiation in humans. EBioMedicine. 24:137–146. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Stagsted LV, Nielsen KM, Daugaard I and Hansen TB: Noncoding AUG circRNAs constitute an abundant and conserved subclass of circles. Life Sci Alliance. 2:e2019003982019. View Article : Google Scholar : PubMed/NCBI

80 

Jakobi T, Siede D, Eschenbach J, Heumüller AW, Busch M, Nietsch R, Meder B, Most P, Dimmeler S, Backs J, et al: Deep characterization of circular RNAs from human cardiovascular cell models and cardiac tissue. Cells. 9:16162020. View Article : Google Scholar

81 

van Heesch S, Witte F, Schneider-Lunitz V, Schulz JF, Adami E, Faber AB, Kirchner M, Maatz H, Blachut S, Sandmann CL, et al: The translational landscape of the human heart. Cell. 178:242–260.e29. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Wang K, Gan TY, Li N, Liu CY, Zhou LY, Gao JN, Chen C, Yan KW, Ponnusamy M, Zhang YH and Li PF: Circular RNA mediates cardiomyocyte death via miRNA-dependent upregulation of MTP18 expression. Cell Death Differ. 24:1111–1120. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Wang L, Shen C, Wang Y, Zou T, Zhu H, Lu X, Li L, Yang B, Chen J, Chen S, et al: Identification of circular RNA Hsa_ circ_0001879 and Hsa_circ_0004104 as novel biomarkers for coronary artery disease. Atherosclerosis. 286:88–96. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Zou TY, Li L, Huang JF, Yang B and Wang LY: Expression and clinical significance of circular RNA circTCF25 in patients with coronary artery disease. Mol Cardiol China. 3:27–31. 2018.

85 

Holdt LM, Stahringer A, Sass K, Pichler G, Kulak NA, Wilfert W, Kohlmaier A, Herbst A, Northoff BH, Nicolaou A, et al: Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat Commun. 7:124292016. View Article : Google Scholar : PubMed/NCBI

86 

Hu X, Chen L, Wu S, Xu K, Jiang W, Qin M, Zhang Y and Liu X: Integrative analysis reveals key circular RNA in atrial fibrillation. Front Genet. 10:1082019. View Article : Google Scholar : PubMed/NCBI

87 

Ludmer PL, Selwyn AP, Shook TL, Wayne RR, Mudge GH, Alexander RW and Ganz P: Paradoxical vasoconstriction induced by acetylcholine in atherosclerotic coronary arteries. N Engl J Med. 315:1046–1051. 1986. View Article : Google Scholar : PubMed/NCBI

88 

Simionescu M: Implications of early structural-functional changes in the endothelium for vascular disease. Arterioscler Thromb Vasc Biol. 27:266–274. 2007. View Article : Google Scholar

89 

Chen PY, Qin L, Baeyens N, Li G, Afolabi T, Budatha M, Tellides G, Schwartz MA and Simons M: Endothelial-to-mesenchymal transition drives atherosclerosis progression. J Clin Invest. 125:4514–4528. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Liu C, Yao MD, Li CP, Shan K, Yang H, Wang JJ, Liu B, Li XM, Yao J, Jiang Q and Yan B: Silencing Of Circular RNA-ZNF609 ameliorates vascular endothelial dysfunction. Theranostics. 7:2863–2877. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Li CY, Ma L and Yu B: Circular RNA hsa_circ_0003575 regulates oxLDL induced vascular endothelial cells proliferation and angiogenesis. Biomed Pharmacother. 95:1514–1519. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Yang L, Han B, Zhang Y, Bai Y, Chao J, Hu G and Yao H: Engagement of circular RNA HECW2 in the nonautophagic role of ATG5 implicated in the endothelial-mesenchymal transition. Autophagy. 14:404–418. 2018. View Article : Google Scholar :

93 

Liu C, Ge HM, Liu BH, Dong R, Shan K, Chen X, Yao MD, Li XM, Yao J, Zhou RM, et al: Targeting pericyte-endothelial cell crosstalk by circular RNA-cPWWP2A inhibition aggravates diabetes-induced microvascular dysfunction. Proc Natl Acad Sci USA. 116:7455–7464. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Hall IF, Climent M, Quintavalle M, Farina FM, Schorn T, Zani S, Carullo P, Kunderfranco P, Civilini E, Condorelli G and Elia L: Circ_Lrp6, a Circular RNA enriched in vascular smooth muscle cells, acts as a sponge regulating miRNA-145 function. Circ Res. 124:498–510. 2019. View Article : Google Scholar

95 

Zheng C, Niu H, Li M, Zhang H, Yang Z, Tian L, Wu Z, Li D and Chen X: Cyclic RNA has-circ-000595 regulates apoptosis of aortic smooth muscle cells. Mol Med Rep. 12:6656–6662. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Sun Y, Zhang S, Yue M, Li Y, Bi J and Liu H: Angiotensin II inhibits apoptosis of mouse aortic smooth muscle cells through regulating the circNRG-1/miR-193b-5p/NRG-1 axis. Cell Death Dis. 10:3622019. View Article : Google Scholar : PubMed/NCBI

97 

Chen J, Cui L, Yuan J, Zhang Y and Sang H: Circular RNA WDR77 target FGF-2 to regulate vascular smooth muscle cells proliferation and migration by sponging miR-124. Biochem Biophys Res Commun. 494:126–132. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Sun Y, Yang Z, Zheng B, Zhang XH, Zhang ML, Zhao XS, Zhao HY, Suzuki T and Wen JK: A novel regulatory mechanism of smooth muscle α-actin expression by NRG-1/circACTA2/miR-548f-5p Axis. Circ Res. 121:628–635. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Zhu Y, Pan W, Yang T, Meng X, Jiang Z, Tao L and Wang L: Upregulation of circular RNA circNFIB attenuates cardiac fibrosis by sponging miR-433. Front Genet. 10:5642019. View Article : Google Scholar : PubMed/NCBI

100 

Sun LY, Zhao JC, Ge XM, Zhang H, Wang CM and Bie ZD: Circ_LAS1L regulates cardiac fibroblast activation, growth, and migration through miR-125b/SFRP5 pathway. Cell Biochem Funct. 38:443–450. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Zhou B and Yu JW: A novel identified circular RNA, circRNA_010567, promotes myocardial fibrosis via suppressing miR-141 by targeting TGF-β1. Biochem Biophys Res Commun. 487:769–775. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Tang CM, Zhang M, Huang L, Hu ZQ, Zhu JN, Xiao Z, Zhang Z, Lin QX, Zheng XL, Yang M, et al: CircRNA_000203 enhances the expression of fibrosis-associated genes by derepressing targets of miR-26b-5p Col1a2 and CTGF, in cardiac fibroblasts. Sci Rep. 7:403422017. View Article : Google Scholar

103 

Shen L, Hu Y, Lou J, Yin S, Wang W, Wang Y, Xia Y and Wu W: CircRNA-0044073 is upregulated in atherosclerosis and increases the proliferation and invasion of cells by targeting miR-107. Mol Med Rep. 19:3923–3932. 2019.PubMed/NCBI

104 

Geng HH, Li R, Su YM, Xiao J, Pan M, Cai XX and Ji XP: The circular RNA Cdr1as promotes myocardial infarction by mediating the regulation of miR-7a on its target genes expression. PLoS One. 11:e01517532016. View Article : Google Scholar : PubMed/NCBI

105 

Zhao Z, Li X, Gao C, Jian D, Hao P, Rao L and Li M: Peripheral blood circular RNA hsa_circ_0124644 can be used as a diagnostic biomarker of coronary artery disease. Sci Rep. 7:399182017. View Article : Google Scholar : PubMed/NCBI

106 

Gan J, Yuan J, Liu Y, Lu Z, Xue Y, Shi L and Zeng H: Circular RNA_101237 mediates anoxia/reoxygenation injury by targeting let-7a-5p/IGF2BP3 in cardiomyocytes. Int J Mol Med. 45:451–460. 2020.PubMed/NCBI

107 

Huang S, Li X, Zheng H, Si X, Li B, Wei G, Li C, Chen Y, Chen Y, Liao W, et al: Loss of super-enhancer-regulated circRNA Nfix induces cardiac regeneration after myocardial infarction in adult mice. Circulation. 139:2857–2876. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Lim TB, Aliwarga E, Luu TDA, Li YP, Ng SL, Annadoray L, Sian S, Ackers-Johnson MA and Foo RS: Targeting the highly abundant circular RNA circSlc8a1 in cardiomyocytes attenuates pressure overload induced hypertrophy. Cardiovasc Res. 115:1998–2007. 2019. View Article : Google Scholar : PubMed/NCBI

109 

Zhang S, Song G, Yuan J, Qiao S, Xu S, Si Z, Yang Y, Xu X and Wang A: Circular RNA circ_0003204 inhibits proliferation, migration and tube formation of endothelial cell in atherosclerosis via miR-370-3p/TGFβR2/phosph-SMAD3 axis. J Biomed Sci. 27:112020. View Article : Google Scholar

110 

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

111 

Ni H, Li W, Zhuge Y, Xu S, Wang Y, Chen Y, Shen G and Wang F: Inhibition of circHIPK3 prevents angiotensin II-induced cardiac fibrosis by sponging miR-29b-3p. Int J Cardiol. 292:188–196. 2019. View Article : Google Scholar : PubMed/NCBI

112 

Wang Y, Zhao R, Liu W, Wang Z, Rong J, Long X, Liu Z, Ge J and Shi B: Exosomal circHIPK3 released from hypoxia-pretreated cardiomyocytes regulates oxidative damage in cardiac micro-vascular endothelial cells via the miR-29a/IGF-1 pathway. Oxid Med Cell Longev. 2019:79546572019. View Article : Google Scholar

113 

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

114 

Jin Q and Chen Y: Silencing circular RNA circ_0010729 protects human cardiomyocytes from oxygen-glucose deprivation-induced injury by up-regulating microRNA-145-5p. Mol Cell Biochem. 462:185–194. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Cai L, Qi B, Wu X, Peng S, Zhou G, Wei Y, Xu J, Chen S and Liu S: Circular RNA Ttc3 regulates cardiac function after myocardial infarction by sponging miR-15b. J Mol Cell Cardiol. 130:10–22. 2019. View Article : Google Scholar : PubMed/NCBI

116 

Holdt LM, Beutner F, Scholz M, Gielen S, Gäbel G, Bergert H, Schuler G, Thiery J and Teupser D: ANRIL expression is associated with atherosclerosis risk at chromosome 9p21. Arterioscler Thromb Vasc Biol. 30:620–627. 2010. View Article : Google Scholar : PubMed/NCBI

117 

Song CL, Wang JP, Xue X, Liu N, Zhang XH, Zhao Z, Liu JG, Zhang CP, Piao ZH, Liu Y and Yang YB: Effect of Circular ANRIL on the inflammatory response of vascular endothelial cells in a rat model of coronary atherosclerosis. Cell Physiol Biochem. 42:1202–1212. 2017. View Article : Google Scholar : PubMed/NCBI

118 

Menglan LI, Siying HE, Jialing R, Bin L, Xiaokang Z and Fang Z: The possible protective role of circDLGAP4 from peripheral blood in coronary heart disease. Chin J Clin Lab Sci. 37:109–112. 2019.

119 

Parahuleva MS, Euler G, Mardini A, Parviz B, Schieffer B, Schulz R and Aslam M: Identification of microRNAs as potential cellular monocytic biomarkers in the early phase of myocardial infarction: A pilot study. Sci Rep. 7:159742017. View Article : Google Scholar : PubMed/NCBI

120 

Sala F, Aranda JF, Rotllan N, Ramírez CM, Aryal B, Elia L, Condorelli G, Catapano AL, Fernández-Hernando C and Norata GD: MiR-143/145 deficiency attenuates the progression of atherosclerosis in Ldlr-/-mice. Thromb Haemost. 112:796–802. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Liu CX, Li X, Nan F, Jiang S, Gao X, Guo SK, Xue W, Cui Y, Dong K, Ding H, et al: Structure and degradation of circular RNAs regulate PKR activation in innate immunity. Cell. 177:865–880.e21. 2019. View Article : Google Scholar : PubMed/NCBI

122 

Zhang Y, Zhang Y, Li X, Zhang M and Lv K: Microarray analysis of circular RNA expression patterns in polarized macrophages. Int J Mol Med. 39:373–379. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Ng WL, Marinov GK, Liau ES, Lam YL, Lim YY and Ea CK: Inducible RasGEF1B circular RNA is a positive regulator of ICAM-1 in the TLR4/LPS pathway. RNA Biol. 13:861–871. 2016. View Article : Google Scholar : PubMed/NCBI

124 

Chen YG, Kim MV, Chen X, Batista PJ, Aoyama S, Wilusz JE, Iwasaki A and Chang HY: Sensing self and foreign circular RNAs by intron identity. Mol Cell. 67:228–238.e5. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Li X, Liu CX, Xue W, Zhang Y, Jiang S, Yin QF, Wei J, Yao RW, Yang L and Chen LL: Coordinated circRNA biogenesis and function with NF90/NF110 in viral infection. Mol Cell. 67:214–227.e7. 2017. View Article : Google Scholar : PubMed/NCBI

126 

Zhang J, Wang P, Wan L, Xu S and Pang D: The emergence of noncoding RNAs as heracles in autophagy. Autophagy. 13:1004–1024. 2017. View Article : Google Scholar : PubMed/NCBI

127 

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

128 

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

129 

Du WW, Yang W, Liu E, Yang Z, Dhaliwal P and Yang BB: Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Res. 44:2846–2858. 2016. View Article : Google Scholar : PubMed/NCBI

130 

Zeng Y, Du WW, Wu Y, Yang Z, Awan FM, Li X, Yang W, Zhang C, Yang Q, Yee A, et al: A circular RNA binds to and activates AKT phosphorylation and nuclear localization reducing apoptosis and enhancing cardiac repair. Theranostics. 7:3842–3855. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Garikipati VNS, Verma SK, Cheng Z, Liang D, Truongcao MM, Cimini M, Yue Y, Huang G, Wang C, Benedict C, et al: Circular RNA CircFndc3b modulates cardiac repair after myocardial infarction via FUS/VEGF-A axis. Nat Commun. 10:43172019. View Article : Google Scholar : PubMed/NCBI

132 

Westholm JO, Miura P, Olson S, Shenker S, Joseph B, Sanfilippo P, Celniker SE, Graveley BR and Lai EC: Genome-wide analysis of drosophila circular RNAs reveals their structural and sequence properties and age-dependent neural accumulation. Cell Rep. 9:1966–1980. 2014. View Article : Google Scholar : PubMed/NCBI

133 

Bachmayr-Heyda A, Reiner AT, Auer K, Sukhbaatar N, Aust S, Bachleitner-Hofmann T, Mesteri I, Grunt TW, Zeillinger R and Pils D: Correlation of circular RNA abundance with proliferation-exemplified with colorectal and ovarian cancer, idiopathic lung fibrosis, and normal human tissues. Sci Rep. 5:80572015. View Article : Google Scholar

134 

Lasda E and Parker R: Circular RNAs co-precipitate with extra-cellular vesicles: A possible mechanism for circRNA clearance. PLoS One. 11:e01484072016. View Article : Google Scholar

135 

Dou Y, Cha DJ, Franklin JL, Higginbotham JN, Jeppesen DK, Weaver AM, Prasad N, Levy S, Coffey RJ, Patton JG and Zhang B: Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes. Sci Rep. 6:379822016. View Article : Google Scholar : PubMed/NCBI

136 

Preußer C, Hung LH, Schneider T, Schreiner S, Hardt M, Moebus A, Santoso S and Bindereif A: Selective release of circRNAs in platelet-derived extracellular vesicles. J Extracell Vesicles. 7:14244732018. View Article : Google Scholar

137 

Viereck J and Thum T: Circulating noncoding RNAs as biomarkers of cardiovascular disease and injury. Circ Res. 120:381–399. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Memczak S, Papavasileiou P, Peters O and Rajewsky N: Identification and characterization of circular RNAs as a new class of putative biomarkers in human blood. PLoS One. 10:e01412142015. View Article : Google Scholar : PubMed/NCBI

139 

Huang C, Liang D, Tatomer DC and Wilusz JE: A length-dependent evolutionarily conserved pathway controls nuclear export of circular RNAs. Genes Dev. 32:639–644. 2018. View Article : Google Scholar : PubMed/NCBI

140 

Zhou C, Molinie B, Daneshvar K, Pondick JV, Wang J, Van Wittenberghe N, Xing Y, Giallourakis CC and Mullen AC: Genome-wide maps of m6A circRNAs identify widespread and cell-type-specific methylation patterns that are distinct from mRNAs. Cell Rep. 20:2262–2276. 2017. View Article : Google Scholar : PubMed/NCBI

141 

Roundtree IA, Luo GZ, Zhang Z, Wang X, Zhou T, Cui Y, Sha J, Huang X, Guerrero L, Xie P, et al: YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. Elife. 6:e313112017. View Article : Google Scholar

142 

Han Y, Donovan J, Rath S, Whitney G, Chitrakar A and Korennykh A: Structure of human RNase L reveals the basis for regulated RNA decay in the IFN response. Science. 343:1244–1248. 2014. View Article : Google Scholar : PubMed/NCBI

143 

Park OH, Ha H, Lee Y, Boo SH, Kwon DH, Song HK and Kim YK: Endoribonucleolytic cleavage of m6A-containing RNAs by RNase P/MRP complex. Mol Cell. 74:494–507.e8. 2019. View Article : Google Scholar

144 

Tapsin S, Sun M, Shen Y, Zhang H, Lim XN, Susanto TT, Yang SL, Zeng GS, Lee J, Lezhava A, et al: Genome-wide identification of natural RNA aptamers in prokaryotes and eukaryotes. Nat Commun. 9:12892018. View Article : Google Scholar : PubMed/NCBI

145 

Yang Y, Gao X, Zhang M, Yan S, Sun C, Xiao F, Huang N, Yang X, Zhao K, Zhou H, et al: Novel role of FBXW7 Circular RNA in repressing glioma tumorigenesis. J Natl Cancer Inst. 110:304–315. 2018. View Article : Google Scholar :

146 

Costello A, Lao NT, Barron N and Clynes M: Continuous translation of circularized mRNA improves recombinant protein titer. Metab Eng. 52:284–292. 2019. View Article : Google Scholar : PubMed/NCBI

147 

Xie YZ, Yang F, Tan W, Li X, Jiao C, Huang R and Yang BB: The anti-cancer components of Ganoderma lucidum possesses cardiovascular protective effect by regulating circular RNA expression. Oncoscience. 3:203–207. 2016. View Article : Google Scholar :

148 

Dang RY, Liu FL and Li Y: Circular RNA hsa_circ_0010729 regulates vascular endothelial cell proliferation and apoptosis by targeting the miR-186/HIF-1α axis. Biochem Biophys Res Commun. 490:104–110. 2017. View Article : Google Scholar : PubMed/NCBI

149 

Wei L, Sun J, Zhang N, Zheng Y, Wang X, Lv L, Liu J, Xu Y, Shen Y and Yang M: Noncoding RNAs in gastric cancer: Implications for drug resistance. Mol Cancer. 19:622020. View Article : Google Scholar : PubMed/NCBI

150 

Zhang Y, Yu J, Kahkoska AR and Gu Z: Photoacoustic drug delivery. Sensors (Basel). 17. pp. 14002017, View Article : Google Scholar

151 

Lavenniah A, Luu TDA, Li YP, Lim TB, Jiang J, Ackers-Johnson M and Foo RS: Engineered circular RNA sponges act as miRNA inhibitors to attenuate pressure overload-induced cardiac hyper-trophy. Mol Ther. 28:1506–1517. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 47 Issue 1

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu Z, Huang Q, Zhang Q, Wu H and Zhong Z: CircRNAs open a new era in the study of cardiovascular disease (Review). Int J Mol Med 47: 49-64, 2021
APA
Yu, Z., Huang, Q., Zhang, Q., Wu, H., & Zhong, Z. (2021). CircRNAs open a new era in the study of cardiovascular disease (Review). International Journal of Molecular Medicine, 47, 49-64. https://doi.org/10.3892/ijmm.2020.4792
MLA
Yu, Z., Huang, Q., Zhang, Q., Wu, H., Zhong, Z."CircRNAs open a new era in the study of cardiovascular disease (Review)". International Journal of Molecular Medicine 47.1 (2021): 49-64.
Chicago
Yu, Z., Huang, Q., Zhang, Q., Wu, H., Zhong, Z."CircRNAs open a new era in the study of cardiovascular disease (Review)". International Journal of Molecular Medicine 47, no. 1 (2021): 49-64. https://doi.org/10.3892/ijmm.2020.4792