Next Article in Journal
Combination of Radix Astragali and Safflower Promotes Angiogenesis in Rats with Ischemic Stroke via Silencing PTGS2
Previous Article in Journal
The Dynamic and Crucial Role of the Arginine Methylproteome in Myoblast Cell Differentiation
Previous Article in Special Issue
The Role of Circular RNAs in the Physiology and Pathology of the Mammalian Ovary
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circular RNAs as a Diagnostic and Therapeutic Target in Cardiovascular Diseases

by
Victor Hugo Antonio Joaquim
,
Noemy Pinto Pereira
,
Tiago Fernandes
and
Edilamar Menezes Oliveira
*
Laboratory of Biochemistry and Molecular Biology Applied to the Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2125; https://doi.org/10.3390/ijms24032125
Submission received: 18 August 2022 / Revised: 4 October 2022 / Accepted: 7 October 2022 / Published: 21 January 2023
(This article belongs to the Special Issue The Lesser Known World of RNA Circles)

Abstract

:
Circular RNAs (circRNAs) are a family of noncoding RNAs (ncRNAs) that are endogenous and widely distributed in different species, performing several functions, mainly their association with microRNAs (miRNAs) and RNA-binding proteins. CVDs remain the leading cause of death worldwide; therefore, the development of new therapies and strategies, such as gene therapies or nonpharmacological therapies, with low cost, such as physical exercise, to alleviate these diseases is of extreme importance for society. With increasing evidence of ncRNA participating in the progression of CVDs, several studies have reported these RNAs as promising targets for diagnosis and treatment. There are several studies of CVDs and the role of miRNAs and lncRNAs; however, little is known about the new class of RNAs, called circRNAs, and CVDs. In this mini review, we focus on the mechanisms of circRNAs and CVDs.

1. Biogenesis, Characteristics, and Function

The first circRNA molecule that is covalently closed was discovered by Sanger et al., (1976) [1] in plant viroids; since then, some studies associated with circRNAs have been published. However, only in 2010 with advances in RNA sequencing technology did several studies emerge linking circRNAs to diseases such as cardiovascular diseases (CVDs) and cancer [2,3,4,5,6,7,8,9].
CircRNAs are formed through pre-mRNA backsplicing; during backsplicing, a downstream 5′ splice site is linked to an upstream 3′ splice site in a reverse orientation, thus resulting in a circular RNA molecule with a 3′,5′ phosphodiester bond at the backsplicing junction site [10]. This process can occur both at the co- and posttranscriptional levels [10,11,12]. However, the circular RNA molecule can be formed in several ways, and until recent studies, are divided into five categories: exonic circular RNA (EcircRNA), where most are located in the cytoplasm, intronic circular RNA (ciRNA), exon–intron circular RNA (EIcircRNA), where the latter two are mostly located nucleus fusion circRNAs (f-circRNAs), and read-through circRNAs (rt-circRNAs) [10,11] (Figure 1).
Therefore, as circRNAs are generated from pre-mRNA backsplicing, as mentioned above, the literature shows that there are three types of models of biogenesis, including intron-pairing-triggered circularization, RNA binding protein (RBP)-driven circularization, and lariat-triggered circularization.
Circularization driven by intron pairing is considered the primary model for the formation of circRNA, and intronic complementary sequences (ICSs), such as Alu elements, which are known as transposable elements that flank backsplicing exons, are joined by base pairing, which triggers the formation of circRNA and exon-intron-containing circular RNA (EIcircRNA). In RNA-binding protein (RBP)-driven circularization, RBPs recognize unique motifs in the pre-mRNA to initiate circRNA formation. The docking of RBPs into ICSs promotes or inhibits circRNA formation. In lariat-driven circularization, intronic lariats that escape debranching and degradation can be processed into circular intronic RNA (ciRNA) during splicing [8,9,12].
CircRNAs have multiple functions, including [1] regulating transcription mediated by polymerase II (Pol II), [2] acting as miRNA sponges to block inhibitory effects on mRNAs, [3] sponging proteins to suppress their activity, or serving as scaffolds to bring in proteins to promote protein activity, [4] encode peptides, and proteins, and [5] serve as biomarker (Figure 1).
The role of circRNA as miRNA sponge is currently one of the most studied. When the miRNA performs partial base pairing with the 3′ UTR of its complementary mRNA, it promotes translational repression. Some research has reported the existence of miRNA binding sites on circRNA. It has been shown that circRNA can be a competing endogenous RNA (ceRNA) that binds to miRNA, thus weakening the inhibitory effect of miRNA on mRNA but promoting the expression of target genes [13]. This process is the “effect sponge” from circRNA. However, it will only work when the abundance of circRNA that has a sponge effect matches the abundance of miRNA. A single circRNA can bind many miRNAs at one or more sites and can have opposite effects on different diseases.

2. circRNA Expression in Cardiovascular Diseases

The study of circRNAs has gained strength with the advancement of transcriptomics analysis technologies and has become crucial to understand the molecular mechanisms of the pathophysiology of several health conditions. CVDs are the leading cause of death worldwide [14], making the cardiovascular system one of the main focuses of attention in circRNA studies, showing an emerging body of evidence about the ubiquitous role of this class of ncRNA at the posttranscriptional level [15].
CircRNAs can regulate gene expression in CVDs usually through a circRNA-miRNA-mRNA axis [16], acting as ceRNAs (“miRNA sponges”) or as protein antagonists, modulating several biological processes, such as cell proliferation, migration, differentiation, and apoptosis [17].
Using RNA sequencing and specific bioinformatics tools, Tan et al., [2] found 15,318 and 3017 circRNAs expressed in 12 human and 25 mouse hearts, respectively, across a 28-day differentiation time course. In human hearts, 82.32% of identified circRNAs were exonic and generated from their respective linear cognate gene. Among these, the most abundant cardiac-expressed circRNA was generated from the SLC8A1 gene (or NCX1 gene, from Na+/Ca++ exchanger), found in all 12 heart samples and formed by a single exon. The following top 10 highest circRNAs expression ranking found in all human heart samples also contains circHIPK3-2, circEXOC6B-14, circALPK2-2, and circMB-2. A comprehensive analysis of RNA-seq collected from two databases found 15,278 circRNAs in five human adult heart samples and 797 circRNAs in four mouse heart samples [18]. They also found 1608 circRNAs in three human fetus samples, reinforcing the evidence that the abundance of circRNAs seems to vary throughout biological development [19]. In Fischer 344 rats, Gong et al., [20] found that a significant number of circRNAs were differentially expressed depending on the organ (across 11 organs), sex (male and female), and age (juvenile, adolescence, adult, and aging). Additionally, when investigating the translational landscape of 80 human hearts, van Heesch et al., [21] found 8878 circRNAs in 3181 genes. The most interesting finding of this study was that 40 of these circRNAs had the potential to translate into proteins due to their ribosome association, a function considered rare but previously reported [22].

3. circRNAs in Arterial Hypertension

Arterial hypertension (AH) is characterized by a chronic and abnormal rise in blood pressure, with a multifactorial etiology involving genetic, environmental, and social determinants, and is the main contributor to the global burden of CVDs [23].
Liu et al., [24] found 485 differentially expressed circRNAs in aortic vascular tissue of spontaneously hypertensive rats (SHR) when compared to Wistar Kyoto (WKT) rats. Three circRNA-miRNA-mRNA axes were predicted and later confirmed in SHR aortas, regulating the NOTCH1, FOXO3, and STAT3 genes, all of which are involved in vascular diseases. The researchers highlighted that some of these differentially expressed circRNAs found in rats were highly similar to homologous sequences in human circRNAs. In another study, researchers used a model of angiotensin-II-induced vascular smooth muscle cell senescence and vascular samples from human hypertensive patients to demonstrate that the overexpression of circACTA2 decreased CDK4 mRNA stability and protein expression, leading to cell senescence [25]. Endothelial dysfunction, crucial for the development of arterial hypertension, is closely associated with several ncRNAs, especially lncRNAs, circRNAs, and miRNAs [26]. CircACTA2 may also be involved in vascular function and remodeling by regulating smooth muscle alpha actin (αSMA) expression [27,28].
To date, the human circRNAs_0037911, _0126991, and _0005870 seem to be the best candidates as hypertension biomarkers. CircRNA_0037911 and _0126991 were found to be highly regulated in the blood of hypertensive patients, while circRNA_0005870 was downregulated. Taken together, these circRNAs seem to be involved in vascular endothelial dysfunction and, consequently, the development of arterial hypertension (Table 1).

4. circRNAs in Myocardial Infarction

Myocardial infarction (MI) is a highly prevalent CVD with substantial global morbidity and mortality rates [37], making MI one of the main focuses of circRNA-related studies, with a large number of described circRNAs [38] (Table 2). Ischemia/reperfusion (I/R) injury is one methodology to study the mechanisms that lead to tissue injury in this disease, and we have had a huge increase in the understanding of how ncRNAs are involved in several underlying pathological mechanisms in IR injury, acting as biomarkers or as therapeutic strategies [39,40,41], especially those related to myocardial inflammation, apoptosis, angiogenesis, fibrosis, and oxidative damage.
With a known key role in fibrotic and tumoral diseases [42], circHIPK3, which is highly expressed in several human tissues, seems to be involved in even more pathophysiological processes. Si et al., [43] showed the cardiac regeneration effect of circHIPK3 by modulating cardiomyogenesis and angiogenesis after MI. When overexpressed, circHIPK3 increased the proliferation, migration, and tube formation of coronary vessel endothelial cells, attenuated cardiac dysfunction, and decreased the MI-related fibrotic area. Researchers conclude that circHIPK3 may be an ideal candidate as a therapeutic target to improve prognosis after MI. Another study also found that circHIPK3 regulates post-MI cardiac angiogenesis, but in cardiomyocyte-derived exosomes [44].
Myocardial-infarction-associated circular RNA (MICRA), from the ZNF604 locus, remains the main circRNA biomarker associated with MI. Salgado-Somoza et al., [45] used the blood levels of MICRA to stratify patients into groups depending on how preserved their left ventricular ejection fraction (LVEF) was four months after MI. MICRA was able to improve the risk classification and was proposed as a biomarker in the prognostication strategy. In another study, Vausort et al., [46] enrolled infarcted patients from two independent cohorts and found that MI patients had lower MICRA expression levels than healthy subjects. They also indicated MICRA as a strong predictor of left ventricle dysfunction.
Table 2. List of circRNAs associated with myocardial infarction and I/R injury.
Table 2. List of circRNAs associated with myocardial infarction and I/R injury.
IdHost GeneSpeciesSourceExpressionActionRef.
Cdr1as/CiRS-7CDR1MouseCardiomyocytesUpPromotes CM apoptosis by sponging miR-7a.[47]
PigMyocardiumUpIt is positively correlated with better left and right ventricle function and infarcted size decrease.[48]
circFndc3bFNDC3BMouse
Human
CardiomyocytesDownLimits ischemic injury via FUS/VEGF signaling.[49]
circNfixNfixMouseCardiomyocytesUpPromotes cardiac regenerative repair after MI by suppressing Ybx1 degradation and increasing miR-214 activity.[50]
circ-Ttc3TTC3RatCardiomyocytesUpPromotes cardioprotection via miR-15b/Arl2 axis.[51]
MICRAZNF609HumanBloodDownBiomarker.[45]
circFASTKD1FASTKD1HumanHUVECs, HCMECsUpIts downregulation improved cardiac function after MI by enhancing angiogenesis via miR-106a/LATS1/2/YAP pathway.[52]
circHelzHelzMouseMyocardium
NMVCs
UpPromotes pyroptosis resulting in myocardial injury via miR-133a-3p/NLRP3 axis.[53]
circJARID2Jarid2MouseCardiac tissuesUpApoptotic and inflammatory damage in CM promoted by miR-9-5p/BNIP3 axis.[54]
circ-100338SNX27HumanHCAECDownInduces angiogenesis in I/R injury by sponging miRNA-200a-3p.[55]
circDLGAP4HECTD1MouseHUVECsDownPlays a role in apoptosis and cell migration via miR-143/HECTD1 axis.[56]
circ-SWT1SWT1HumanHC AC16 cellsDownReduces apoptosis, oxidative stress, and endoplasmic reticulum stress via miR-192-5p/SOD2 axis.[57]
circROBO2Robo2MouseMyocardial tissuesUpcircROBO2 knockdown reduces apoptosis by sponging miR-1184 and enhancing TRADD expression levels.[58]
circRNA-101237CDK8MouseCardiomyocytesUpRegulates CM apoptosis via let-7a-5p/IGF2BP3 axis.[59]
circPAN3PAN3RatCardiac tissuesUpPromotes cardiac fibrosis after MI via miR-221/FoxO3/ATG7 axis.[60]
circNCX1NCX1MouseCardiomyocytesUpPromotes CM apoptosis via miR-133a-3p/CDIP1 regulatory pathway.[61]
circRNA1615Copb1MouseMyocardial tissueDownModulates autophagy by the miRNA152/3p/LRP6 molecular axis, reducing ferroptosis in MI mouse hearts.[62]
circ_0023461ARAP1HumanHC AC16 cellsUpReduces hypoxia-induced dysfunction in cardiomyocytes via miR-370-3p/PDE4D axis.[63]
circHipk3Hipk3MouseCardiomyocyteUpTriggers CM proliferation and angiogenesis by miR-133a/Notch1 signaling path.[43]
MouseCardiomyocyteUpInduces tube formation, cell proliferation, and migration via miR-29a/VEGFA axis, stimulating cardiac angiogenesis after MI.[44]
HumanHCMUpInduces CM apoptosis after I/R injury by sponging miRNA-124-3p.[64]
circTLK1TLK1MouseCardiomyocyteUpPromotes CM apoptosis via miR-214/RIPK1-mediated TNF signaling pathway.[65]
MFACRSmyd4MouseCardiomyocyteUpInduces CM apoptosis via miR-652-3p/MTP18 axis.[66]
HumanPlasma,
HC AC16 cells
UpPromotes CM apoptosis by downregulating miR-125b through methylation.[67]
circUbe3aUbe3aMouseCardiac tissueUpExacerbates MI-induced myocardial fibrosis via miR-138-5p/RhoC axis.[68]
circ_0060745Cse1lMouseCardiomyocyteUpIncreases infarct size and impaired cardiac function after MI.[69]
ACAP2ACAP2HumanPlasma,
HC AC16 cells
UpInduces CM apoptosis by promoting maturation of miR-532.[70]
circCDYLCDYLMouseCardiomyocyteDownPromotes CM proliferation through miR-4793-5p/APP pathway.[71]
circTRRAPTRRAPHumanHC AC16 cellsUpIncreases inflammatory, apoptotic, and oxidative damage in CM via miR-370-3p/PAWR axis.[72]
circMACF1Macf1MouseCardiomyocyteDownAttenuates CM apoptosis via miR-500b-5p/EMP1 axis.[73]
circACR-MouseCardiomyocyteDownPromotes cardioprotection, decreasing myocardial infarction size, autophagy, and cell death via Pink1/FAM65B axis.[74]
circMAT2BMAT2BRatH9c2 cellsUpcircMAT2B knockdown promotes anti-inflammatory and antiapoptotic role via miR-133/PI3K/AKT and Raf/MEK/ERK
pathways.
[75]
circNFIBNfibMouseCardiac fibroblastDownReduces cardiac fibrosis after MI by sponging miR-433.[76]
MI—myocardial infarction; CM—cardiomyocyte; I/R—ischemia/reperfusion; HUVECs—human umbilical vein endothelial cells; HCMECs—human cardiac microvascular endothelial cells; HCM—human-derived cardiomyocytes; NMVCs—neonatal mouse ventricular cardiomyocytes; HCAEC—human coronary endothelial cells; HC AC16 cells—human cardiomyocytes AC16 cells.

5. circRNAs in Coronary Artery Disease

Additionally, known as coronary heart disease or even ischemic heart disease, coronary artery disease (CAD) is characterized by atherosclerotic occlusions of the coronary arteries, possibly leading to myocardial infarction and sudden death, and it is the primary cause of death worldwide [77,78]. Conditions such as chronic inflammation and endothelial injuries are involved in the complex etiology of CAD, and some circRNAs have been linked with the development and progression of this disease [79] (Table 3).
In a mouse model study, Gong et al., [80] reported the role of circEsyt2 in vascular remodeling by enhancing cell proliferation and migration while inhibited apoptosis and differentiation in vascular smooth muscle cells (VSMC). The experimental findings were in accordance with the higher circEsyt2 expression found in severe-CAD patients when compared to mild-CAD patients, suggesting a proatherosclerotic role of this circRNA. Inversely, circANRIL was found to induce apoptosis and inhibit proliferation, conferring atheroprotection [81]. The most interesting finding here is that both linear and circular ANRIL were differentially expressed in CAD patients vs. controls, but linear ANRIL showed an atheros progressive function.
Several circRNAs have been proposed as CAD biomarkers thus far. In CAD vs. non-CAD subjects, the combined expression of circRNAs hsa_circ_0001879 and hsa_circ_0004104 showed equal diagnostic value compared to Holter monitoring, treadmill exercise tests, and coronary computed tomography angiography [82]. hsa_circ_0004104 may also be involved in CAD pathogenesis. hsa_circ_0124644 is also a promising CAD biomarker. In a cohort study, hsa_circ_0124644 was upregulated (2.2-fold change) in the CAD group (n = 137) vs. control group (n = 115). In the CAD vs. control scenario, hsa_circ_0124644 expression had a sensitivity and specificity of 0.86 and 0.62, respectively. The subjects from the CAD group were then classified into two different groups according to disease severity, and hsa_circ_0124644 retained its diagnostic value [83].
Table 3. List of circRNAs associated with CADs.
Table 3. List of circRNAs associated with CADs.
IdHost GeneSpeciesSourceExpressionActionRef.
hsa_circ_0001879NIPSNAP3AHumanPeripheral bloodUpBiomarker.[82]
hsa_circ_0004104SPARCHumanPeripheral bloodUpBiomarker. Its upregulation might contribute to the pathogenesis of atherosclerosis.
circ-YOD1YOD1HumanBlood/HASMCsUpBiomarker.[84]
hsa_circ_0124644ROBO2HumanPeripheral bloodUpBiomarker.[83]
circEsyt2Esyt2MouseAortae tissueUpEnhances cell migration and proliferation and inhibits apoptosis and differentiation in VSMC.[80]
circANRILANRILHumanPeripheral bloodDownPromotes atheroprotection by increasing apoptosis and inhibiting macrophages proliferation.[81]
hsa_circ_0001445SMARCA5HumanPlasmaDownBiomarker.[85]
hsa_circ_0005540MCTP1HumanPlasmaUpBiomarker.[86]
HASMCs—human aortic smooth muscle cells; VSMC—vascular smooth muscle cells.

6. circRNAs in Abdominal Aortic Aneurysm

Although there is a lack of consensus, abdominal aortic aneurysm (AAA) could be defined as a greater than 1.5-fold increase in a localized region of the abdominal aorta diameter, with a slow expansion until its rupture [87]. It is estimated that AAA causes 150,000–200,000 deaths per year worldwide, with higher prevalence in the male, smoker, and older populations [88,89]. Rupture is the main AAA complication, with only ~20% chance of survival [90]; however, cardiovascular events are the most common cause of death in these patients, with increased cardiovascular death risk by approximately 3% per year after AAA diagnosis [91].
Receiving far less attention than other CVDs, probably due to the difficulty of diagnosis, AAA lacks accurate and reliable detection, risk stratification, and rupture prediction tools [92], while emerging evidence supports the potential use of molecular biology to act as biomarkers or therapeutic approach in this condition [93].
Zhou et al., [94] screened the circRNA expression profile in aortas from four AAA patients. RNA high-throughput sequencing detected 13,295 circRNAs, with 411 differentially expressed (145 upregulated and 266 downregulated) compared to control. The relative downregulated expression of hsa_circ_0005360 in these aortic tissues, confirmed by qRT-PCR, reveals the potential role of this circRNA in AAA pathogenesis since it is an alternatively transcript of AAA-associated LDLR gene [95]. In addition, circRNA/miRNA interaction analysis predicted one binding site between hsa_circ_0005360 and miR-181b, with miRNA associated with vascular inflammation, atherosclerosis, and aneurysms [96,97]. This same expression profile study was added to an integrated analysis [98] compiling four gene expression datasets, including 23 normal artery tissues and 97 AAA samples, and using different prediction tools (e.g., Gene Ontology, Kyoto Encyclopedia of Genes and Genomes analysis, Targetscan, and CircInteractome). The majority of differently expressed genes (DEGs) (263 upregulated and 177 downregulated) were associated with extracellular matrix, tumor necrosis factor-α (TNF-α), and transforming growth factor-β (TGF-β). Bioinformatic analysis, used to build an interaction network between circRNAs, miRNAs, and mRNAs, revealed that the circ_0005073 may modulate several miRNAs affecting the expression of important DEGs in AAA, such as CCL2 (macrophage infiltration), SPP1 (proinflammatory), and UBA52 (protein degradation).
AAA has a complex and multifactorial pathogenic process. Although the literature addressing circRNAs specifically in AAA is still scarce (Table 4) (Figure 2), the current evidence strongly supports the role of circRNAs in cellular processes that could be associated with the development of AAA, such as the modulation of endothelial cells, macrophages, and VSMCs [99].

7. Exercise-Related circRNAs in the Cardiovascular System

Previous studies reviewed the literature focusing on how CVS-related ncRNA expression responds to physical exercise. The conclusion is quite similar between them: although there is great interest in this topic, we lack studies aiming to describe these underlying molecular mechanisms. Aerobic exercise improved cardiac function and decreased cardiac apoptosis and fibrosis by modulating the expression of a few long noncoding RNAs (lncRNAs): growth arrest specific 5 (GAS5), myocardial-infarction-association transcript (MIAT), metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), and H19 (encoded by the H19 gene) [111]. Targeting another class of ncRNAs, Wang et al., [112] described exercised-regulated miRNAs that benefit the heart by promoting protective effects in myocardial fibrosis (miR-29a and miR-29c), ischemia-reperfusion injury (miR-17-3p and miR-222), myocardial infarction (miR-1, miR-214), and heart failure (miR-21-5p, miR-132-3p, miR-208b-3p, miR-212-3p, and miR-335). The majority of studies focus on running and swimming exercise.
With regard to physical-exercise-related circular RNA expression, the evidence is even scarcer. Guo et al., [113] compared the skeletal muscle circRNA profile in sedentary young versus aged mice and aged mice under aerobic training and identified circBBS9 as a potential biomarker of aging-associated muscle dysfunction regulated by exercise. Niu et al., [114] found reduced expression of circRIMS2 and brain-derived neurotrophic factor (BDNF) in the serum of patients with vascular cognitive impairment (VCI) and experimentally showed that aerobic exercise improved cognitive function by inhibiting neuronal apoptosis through the circRIMS2/miR-186/BDNF axis. In another study, aerobic exercise combined with glucosamine therapy improved knee osteoarthritis in a rabbit model by enhancing the expression of CircUNK and, consequently, regulating cartilage-differentiation- and apoptosis-related genes. Meinecke et al., [115] explored the circRNA expression profile in the plasma of marathon runners and proposed circMBOAT2 as a promising biomarker for cardiopulmonary adaptation.
Recently, Zhu et al., [116] showed the role of the circRNA Circ-Ddx60 in heart failure using physical exercise as preventive treatment. The rationale of this study is based on the report in which physical exercise provided an antihypertrophic memory to myocardial muscle even after the regression of exercise-induced physiological hypertrophy [117]. In this study, the term exercise hypertrophic preconditioning (EHP) was applied, as preventive intervention, and was followed by a transverse aortic constriction (TAC) or sham surgery. The EHP group showed an increased expression of Circ-Ddx60, and its silencing attenuated the antihypertrophic effect of exercise, worsening heart failure in animals that underwent TAC.
To the extent of our knowledge, this is the first research article to explore the potential of exercise-related circRNA in a cardiovascular disease, providing interesting insights in this vast research field.

8. Conclusions

Discovered in the 1970s, circRNAs were thought to be nothing more than splicing errors until the early 2010s. Recently, with proper technology and advances in molecular biology research, circRNAs have become a hot topic in health science. The study of circRNA function in physiological and pathological conditions seems to have great potential to improve the understanding of the underlying mechanisms in several aspects of the cardiovascular system, which could be applied in the diagnosis and treatment of cardiac-related diseases.
Some interesting characteristics, such as high stability and resistance, abundance, and sex-, age-, tissue-, and cell-specific expression, make circRNAs a promising tool for diagnostic and prognostic biomarkers. On the other hand, their involvement with some crucial cell processes, such as proliferation, migration, differentiation, apoptosis, and autophagy, suggests that circRNAs play important roles in the development and progression of diseases.
Ultimately, the current literature exploring the role of physical exercise/training as a nonpharmacological intervention, extensively evidenced in other health/performance scenarios, is still scarce when related to circRNAs.

9. Perspectives

Despite these advances, it is still difficult to translate circRNA findings into clinical practice. The lack of standardized procedures, small-sample-size cohort studies, and other minor issues challenge their use in current medicine.
As quoted before, circRNAs opened “a new era in the study of CVDs” [118], and we are just beginning explore and understand this vast field.

Funding

This research received no external funding.

Acknowledgments

The researchers were supported by the Sao Paulo Research Foundation 295 (FAPESP: # Proc No. #2015/22814-5; 2018/22579-4; # 2022/00531-5). National Council for Scientific 296 and Technological Development (CNPq: #313376/2021-2; #200723/2020-0; #422514/2021-7). Pro-rec- 297 tory of Research and Innovation at the USP (Proc No. #2021.1.1 0424.1.9) and Coordination for the 298 Improvement of Higher Education Personnel (CAPES-PROEX: #88887.484856/2020-00).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sanger, H.L.; Klotz, G.; Riesner, D.; Gross, H.J.; Kleinschmidt, A.K. Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc. Natl. Acad. Sci. USA 1976, 73, 3852–3856. [Google Scholar] [CrossRef] [Green Version]
  2. Tan, W.L.W.; Lim, B.T.S.; Anene-Nzelu, C.G.O.; Ackers-Johnson, M.; Dashi, A.; See, K.; Tiang, Z.; Lee, D.P.; Chua, W.W.; Luu, T.D.A.; et al. A Landscape of Circular RNA Expression in the Human Heart. Cardiovasc. Res. 2017, 113, 298–309. [Google Scholar] [CrossRef] [PubMed]
  3. Werfel, S.; Nothjunge, S.; Schwarzmayr, T.; Strom, T.-M.; Meitinger, T.; Engelhardt, S. Characterization of circular RNAs in human, mouse and rat hearts. J. Mol. Cell. Cardiol. 2016, 98, 103–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Li, Y.; Zhang, J.; Huo, C.; Ding, N.; Li, J.; Xiao, J.; Lin, X.; Cai, B.; Zhang, Y.; Xu, J. Dynamic Organization of lncRNA and Circular RNA Regulators Collectively Controlled Cardiac Differentiation in Humans. eBioMedicine 2017, 24, 137–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Fan, X.; Weng, X.; Zhao, Y.; Chen, W.; Gan, T.; Xu, D. Circular RNAs in Cardiovascular Disease: An Overview. BioMed Res. Int. 2017, 2017, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Li, Y.; Zheng, Q.; Bao, C.; Li, S.; Guo, W.; Zhao, J.; Chen, D.; Gu, J.; He, X.; Huang, S. Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 2015, 25, 981–984. [Google Scholar] [CrossRef] [Green Version]
  7. Vo, J.N.; Cieslik, M.; Zhang, Y.; Shukla, S.; Xiao, L.; Zhang, Y.; Wu, Y.-M.; Dhanasekaran, S.M.; Engelke, C.G.; Cao, X.; et al. The Landscape of Circular RNA in Cancer. Cell 2019, 176, 869–881.e813. [Google Scholar] [CrossRef] [Green Version]
  8. Zhang, Y.; Xue, W.; Li, X.; Zhang, J.; Chen, S.; Zhang, J.-L.; Yang, L.; Chen, L.-L. The Biogenesis of Nascent Circular RNAs. Cell Rep. 2016, 15, 611–624. [Google Scholar] [CrossRef] [Green Version]
  9. Liang, D.; Wilusz, J.E. Short intronic repeat sequences facilitate circular RNA production. Genes Dev. 2014, 28, 2233–2247. [Google Scholar] [CrossRef] [Green Version]
  10. LiLi, 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. 2015, 22, 256–264. [Google Scholar] [CrossRef]
  11. Zhang, Y.; Zhang, X.-O.; Chen, T.; Xiang, J.-F.; Yin, Q.-F.; Xing, Y.-H.; Zhu, S.; Yang, L.; Chen, L.-L. Circular Intronic Long Noncoding RNAs. Mol. Cell 2013, 51, 792–806. [Google Scholar] [CrossRef] [Green Version]
  12. Jeck, W.R.; Sorrentino, J.A.; Wang, K.; Slevin, M.K.; Burd, C.E.; Liu, J.; Marzluff, W.F.; Sharpless, N.E. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 2013, 19, 141–157. [Google Scholar] [CrossRef] [Green Version]
  13. Ebert, M.S.; Neilson, J.R.; Sharp, P.A. MicroRNA sponges: Competitive inhibitors of small RNAs in mammalian cells. Nat. Methods 2007, 4, 721–726. [Google Scholar] [CrossRef]
  14. World Health Organization. World Health Statistics 2021: Monitoring Health for the SDGs, Sustainable Development Goals; World Health Organization: Geneva, Switzerland, 2021; ISBN 9789240027053. [Google Scholar]
  15. Lim, T.B.; Lavenniah, A.; Foo, R.S.-Y. Circles in the heart and cardiovascular system. Cardiovasc. Res. 2020, 116, 269–278. [Google Scholar] [CrossRef]
  16. Su, Q.; Lv, X. Revealing New Landscape of Cardiovascular Disease through Circular RNA-MiRNA-MRNA Axis. Genomics 2020, 112, 1680–1685. [Google Scholar] [CrossRef]
  17. Li, B.; Li, Y.; Hu, L.; Liu, Y.; Zhou, Q.; Wang, M.; An, Y.; Li, P. Role of Circular RNAs in the Pathogenesis of Cardiovascular Disease. J. Cardiovasc. Transl. Res. 2020, 13, 572–583. [Google Scholar] [CrossRef]
  18. Xia, S.; Feng, J.; Lei, L.; Hu, J.; Xia, L.; Wang, J.; Xiang, Y.; Liu, L.; Zhong, S.; Han, L.; et al. Comprehensive characterization of tissue-specific circular RNAs in the human and mouse genomes. Brief. Bioinform. 2017, 18, 984–992. [Google Scholar] [CrossRef] [Green Version]
  19. Misir, S.; Wu, N.; Yang, B.B. Specific expression and functions of circular RNAs. Cell Death Differ. 2022, 29, 481–491. [Google Scholar] [CrossRef]
  20. Gong, B.; Xu, J.Z.; Tong, W. Landscape of circRNAs Across 11 Organs and 4 Ages in Fischer 344 Rats. Chem. Res. Toxicol. 2020, 34, 240–246. [Google Scholar] [CrossRef]
  21. van Heesch, S.; Witte, F.; Schneider-Lunitz, V.; Schulz, J.F.; Adami, E.; Faber, A.B.; Kirchner, M.; Maatz, H.; Blachut, S.; Sandmann, C.-L.; et al. The Translational Landscape of the Human Heart. Cell 2019, 178, 242–260.e29. [Google Scholar] [CrossRef]
  22. Pamudurti, N.R.; Bartok, O.; Jens, M.; Ashwal-Fluss, R.; Stottmeister, C.; Ruhe, L.; Hanan, M.; Wyler, E.; Perez-Hernandez, D.; Ramberger, E.; et al. Translation of CircRNAs. Mol. Cell 2017, 66, 9–21.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Brouwers, S.; Sudano, I.; Kokubo, Y.; Sulaica, E.M. Arterial hypertension. Lancet 2021, 398, 249–261. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, Y.; Dong, Y.; Dong, Z.; Song, J.; Zhang, Z.; Liang, L.; Liu, X.; Sun, L.; Li, X.; Zhang, M.; et al. Expression Profiles of Circular RNA in Aortic Vascular Tissues of Spontaneously Hypertensive Rats. Front. Cardiovasc. Med. 2021, 8, 814402. [Google Scholar] [CrossRef] [PubMed]
  25. Ma, Y.; Zheng, B.; Zhang, X.-H.; Nie, Z.-Y.; Yu, J.; Zhang, H.; Wang, D.-D.; Shi, B.; Bai, Y.; Yang, Z.; et al. circACTA2 mediates Ang II-induced VSMC senescence by modulation of the interaction of ILF3 with CDK4 mRNA. Aging 2021, 13, 11610–11628. [Google Scholar] [CrossRef]
  26. Zhang, J.-R.; Sun, H.-J. LncRNAs and circular RNAs as endothelial cell messengers in hypertension: Mechanism insights and therapeutic potential. Mol. Biol. Rep. 2020, 47, 5535–5547. [Google Scholar] [CrossRef] [PubMed]
  27. Weiser-Evans, M.C.M. Smooth Muscle Differentiation Control Comes Full Circle. Circ. Res. 2017, 121, 591–593. [Google Scholar] [CrossRef]
  28. Sun, Y.; Yang, Z.; Zheng, B.; Zhang, X.-H.; Zhang, M.-L.; Zhao, X.-S.; Zhao, H.-Y.; Suzuki, T.; Wen, J.K. A Novel Regulatory Mechanism of Smooth Muscle α-Actin Expression by NRG-1/circACTA2/miR-548f-5p AxisNovelty and Significance. Circ. Res. 2017, 121, 628–635. [Google Scholar] [CrossRef]
  29. Bao, X.; He, X.; Zheng, S.; Sun, J.; Luo, Y.; Tan, R.; Zhao, J.; Zhong, F.; Zhang, L. Up-regulation of circular RNA hsa_circ_0037909 promotes essential hypertension. J. Clin. Lab. Anal. 2019, 33, e22853. [Google Scholar] [CrossRef] [Green Version]
  30. Bao, X.; Zheng, S.; Mao, S.; Gu, T.; Liu, S.; Sun, J.; Zhang, L. A potential risk factor of essential hypertension in case-control study: Circular RNA hsa_circ_0037911. Biochem. Biophys. Res. Commun. 2018, 498, 789–794. [Google Scholar] [CrossRef]
  31. Cheng, X.; Joe, B. Circular RNAs in rat models of cardiovascular and renal diseases. Physiol. Genom. 2017, 49, 484–490. [Google Scholar] [CrossRef]
  32. He, X.; Bao, X.; Tao, Z.; Sun, J.; Zheng, S.; Zhong, F.; Zhang, L. The microarray identification circular RNA hsa_circ_0105015 up-regulated involving inflammation pathway in essential hypertension. J. Clin. Lab. Anal. 2021, 35, e23603. [Google Scholar] [CrossRef]
  33. Liu, L.; Gu, T.; Bao, X.; Zheng, S.; Zhao, J.; Zhang, L. Microarray Profiling of Circular RNA Identifies hsa_circ_0126991 as a Potential Risk Factor for Essential Hypertension. Cytogenet. Genome Res. 2019, 157, 203–212. [Google Scholar] [CrossRef]
  34. Tao, Z.; Zheng, S.; He, X.; Sun, J.; He, C.; Zhang, L. Hsa_circ_0037897 may be a risk factor for essential hypertension via hsa-miR-145-5p. Clin. Exp. Hypertens. 2021, 43, 281–286. [Google Scholar] [CrossRef]
  35. Zheng, S.; Gu, T.; Bao, X.; Sun, J.; Zhao, J.; Zhang, T.; Zhang, L. Circular RNA hsa_circ_0014243 may serve as a diagnostic biomarker for essential hypertension. Exp. Ther. Med. 2018, 17, 1728–1736. [Google Scholar] [CrossRef] [Green Version]
  36. Wu, N.; Jin, L.; Cai, J. Profiling and bioinformatics analyses reveal differential circular RNA expression in hypertensive patients. Clin. Exp. Hypertens. 2017, 39, 454–459. [Google Scholar] [CrossRef]
  37. Anderson, J.L.; Morrow, D.A. Acute myocardial infarction. New Engl. J. Med. 2017, 376, 2053–2064. [Google Scholar] [CrossRef] [Green Version]
  38. Wen, Z.-J.; Xin, H.; Wang, Y.-C.; Liu, H.-W.; Gao, Y.-Y.; Zhang, Y.-F. Emerging roles of circRNAs in the pathological process of myocardial infarction. Mol. Ther. Nucleic Acids 2021, 26, 828–848. [Google Scholar] [CrossRef]
  39. Gong, C.; Zhou, X.; Lai, S.; Wang, L.; Liu, J. Long Noncoding RNA/Circular RNA-MiRNA-MRNA Axes in Ischemia-Reperfusion Injury. Biomed. Res. Int. 2020, 2020, 8838524. [Google Scholar] [CrossRef]
  40. Marinescu, M.; Lazar, A.; Marta, M.M.; Cozma, A.; Catana, C. Non-Coding RNAs: Prevention, Diagnosis, and Treatment in Myocardial Ischemia—Reperfusion Injury. Int. J. Mol. Sci. 2022, 23, 2728. [Google Scholar] [CrossRef]
  41. Huang, F.; Mai, J.; Chen, J.; He, Y.; Chen, X. Non-coding RNAs modulate autophagy in myocardial ischemia-reperfusion injury: A systematic review. J. Cardiothorac. Surg. 2021, 16, 140. [Google Scholar] [CrossRef]
  42. Zhou, J.; Wang, B.; Bin, X.; Xie, C.; Li, B.; Liu, O.; Tang, Z. CircHIPK3: Key Player in Pathophysiology and Potential Diagnostic and Therapeutic Tool. Front. Med. 2021, 8, 615417. [Google Scholar] [CrossRef] [PubMed]
  43. Si, X.; Zheng, H.; Wei, G.; Li, M.; Li, W.; Wang, H.; Guo, H.; Sun, J.; Li, C.; Zhong, S.; et al. circRNA Hipk3 Induces Cardiac Regeneration after Myocardial Infarction in Mice by Binding to Notch1 and miR-133a. Mol. Ther. Nucleic Acids 2020, 21, 636–655. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, Y.; Zhao, R.; Shen, C.; Liu, W.; Yuan, J.; Li, C.; Deng, W.; Wang, Z.; Zhang, W.; Ge, J.; et al. Exosomal CircHIPK3 Released from Hypoxia-Induced Cardiomyocytes Regulates Cardiac Angiogenesis after Myocardial Infarction. Oxidative Med. Cell. Longev. 2020, 2020, 1–19. [Google Scholar] [CrossRef] [PubMed]
  45. Salgado-Somoza, A.; Zhang, L.; Vausort, M.; Devaux, Y. The circular RNA MICRA for risk stratification after myocardial infarction. IJC Heart Vasc. 2017, 17, 33–36. [Google Scholar] [CrossRef] [PubMed]
  46. Vausort, M.; Salgado-Somoza, A.; Zhang, L.; Leszek, P.; Scholz, M.; Teren, A.; Burkhardt, R.; Thiery, J.; Wagner, D.R.; Devaux, Y. Myocardial Infarction-Associated Circular RNA Predicting Left Ventricular Dysfunction. J. Am. Coll. Cardiol. 2016, 68, 1247–1248. [Google Scholar] [CrossRef]
  47. Geng, H.-H.; Li, R.; Su, Y.-M.; Xiao-Ping, J.; Pan, M.; Cai, X.-X.; Ji, X.-P. The Circular RNA Cdr1as Promotes Myocardial Infarction by Mediating the Regulation of miR-7a on Its Target Genes Expression. PLoS ONE 2016, 11, e0151753. [Google Scholar] [CrossRef]
  48. Mester-Tonczar, J.; Winkler, J.; Einzinger, P.; Hasimbegovic, E.; Kastner, N.; Lukovic, D.; Zlabinger, K.; Spannbauer, A.; Traxler, D.; Batkai, S.; et al. Association between Circular RNA CDR1as and Post-Infarction Cardiac Function in Pig Ischemic Heart Failure: Influence of the Anti-Fibrotic Natural Compounds Bufalin and Lycorine. Biomolecules 2020, 10, 1180. [Google Scholar] [CrossRef]
  49. Garikipati, V.N.S.; Verma, S.K.; Cheng, Z.; Liang, D.; Truongcao, M.M.; 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. 2019, 10, 4317. [Google Scholar] [CrossRef] [Green Version]
  50. 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 2019, 139, 2857–2876. [Google Scholar] [CrossRef]
  51. Cai, L.; Qi, B.; Wu, X.; Peng, S.; Zhou, G.; Wei, Y.; Xu, J.; Chen, S.; Liu, S. Circular RNA Ttc3 regulates cardiac function after myocardial infarction by sponging miR-15b. J. Mol. Cell. Cardiol. 2019, 130, 10–22. [Google Scholar] [CrossRef]
  52. Gao, W.-Q.; Hu, X.-M.; Zhang, Q.; Yang, L.; Lv, X.-Z.; Chen, S.; Wu, P.; Duan, D.-W.; Lang, Y.-H.; Ning, M.; et al. Downregulation of circFASTKD1 ameliorates myocardial infarction by promoting angiogenesis. Aging 2021, 13, 3588–3604. [Google Scholar] [CrossRef]
  53. Bian, Y.; Pang, P.; Li, X.; Yu, S.; Wang, X.; Liu, K.; Ju, J.; Wu, H.; Gao, Y.; Liu, Q.; et al. CircHelz activates NLRP3 inflammasome to promote myocardial injury by sponging miR-133a-3p in mouse ischemic heart. J. Mol. Cell. Cardiol. 2021, 158, 128–139. [Google Scholar] [CrossRef]
  54. Cai, X.; Li, B.; Wang, Y.; Zhu, H.; Zhang, P.; Jiang, P.; Yang, X.; Sun, J.; Hong, L.; Shao, L. CircJARID2 Regulates Hypoxia-Induced Injury in H9c2 Cells by Affecting miR-9-5p–Mediated BNIP3. J. Cardiovasc. Pharmacol. 2021, 78, e77–e85. [Google Scholar] [CrossRef]
  55. Chang, H.; Li, Z.-B.; Wu, J.-Y.; Zhang, L. Circ-100338 Induces Angiogenesis after Myocardial Ischemia-Reperfusion Injury by Sponging MiR-200a-3p. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 6323–6332. [Google Scholar] [CrossRef]
  56. Chen, L.; Luo, W.; Zhang, W.; Chu, H.; Wang, J.; Dai, X.; Cheng, Y.; Zhu, T.; Chao, J. circDLPAG4/HECTD1 mediates ischaemia/reperfusion injury in endothelial cells via ER stress. RNA Biol. 2020, 17, 240–253. [Google Scholar] [CrossRef]
  57. Chen, S.; Sun, L.; Hao, M.; Liu, X. Circ-SWT1 Ameliorates H2O2-Induced Apoptosis, Oxidative Stress and Endoplasmic Reticulum Stress in Cardiomyocytes via miR-192-5p/SOD2 Axis. Cardiovasc. Toxicol. 2022, 22, 378–389. [Google Scholar] [CrossRef]
  58. Chen, T.-P.; Zhang, N.-J.; Wang, H.-J.; Hu, S.-G.; Geng, X. Knockdown of circROBO2 attenuates acute myocardial infarction through regulating the miR-1184/TRADD axis. Mol. Med. 2021, 27, 21. [Google Scholar] [CrossRef]
  59. Gan, J.; Yuan, J.; Liu, Y.; Lu, Z.; Xue, Y.; Shi, L.; Zeng, H. Circular RNA_101237 mediates anoxia/reoxygenation injury by targeting let-7a-5p/IGF2BP3 in cardiomyocytes. Int. J. Mol. Med. 2020, 45, 451–460. [Google Scholar] [CrossRef] [Green Version]
  60. Li, F.; Long, T.-Y.; Bi, S.-S.; Sheikh, S.A.; Zhang, C.-L. circPAN3 exerts a profibrotic role via sponging miR-221 through FoxO3/ATG7-activated autophagy in a rat model of myocardial infarction. Life Sci. 2020, 257, 118015. [Google Scholar] [CrossRef]
  61. Li, M.; Ding, W.; Tariq, M.A.; Chang, W.; Zhang, X.; Xu, W.; Hou, L.; Wang, Y.; Wang, J. A circular transcript of ncx1 gene mediates ischemic myocardial injury by targeting miR-133a-3p. Theranostics 2018, 8, 5855–5869. [Google Scholar] [CrossRef]
  62. Li, R.-L.; Fan, C.-H.; Gong, S.-Y.; Kang, S. Effect and Mechanism of LRP6 on Cardiac Myocyte Ferroptosis in Myocardial Infarction. Oxidative Med. Cell. Longev. 2021, 2021, 1–12. [Google Scholar] [CrossRef] [PubMed]
  63. Ren, K.; Li, B.; Jiang, L.; Liu, Z.; Wu, F.; Zhang, Y.; Liu, J.; Duan, W. circ_0023461 Silencing Protects Cardiomyocytes from Hypoxia-Induced Dysfunction through Targeting miR-370-3p/PDE4D Signaling. Oxidative Med. Cell. Longev. 2021, 2021, 1–18. [Google Scholar] [CrossRef] [PubMed]
  64. Bai, M.; Pan, C.-L.; Jiang, G.-X.; Zhang, Y.-M.; Zhang, Z. CircHIPK3 aggravates myocardial ischemia-reperfusion injury by binding to miRNA-124-3p. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10107–10114. [Google Scholar] [CrossRef]
  65. Song, Y.-F.; Zhao, L.; Wang, B.-C.; Sun, J.-J.; Hu, J.-L.; Zhu, X.-L.; Zhao, J.; Zheng, D.-K.; Ge, Z.-W. The circular RNA TLK1 exacerbates myocardial ischemia/reperfusion injury via targeting miR-214/RIPK1 through TNF signaling pathway. Free Radic. Biol. Med. 2020, 155, 69–80. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, K.; Gan, T.-Y.; Li, N.; Liu, C.-Y.; Zhou, L.-Y.; Gao, J.-N.; Chen, C.; Yan, K.-W.; Ponnusamy, M.; Zhang, Y.-H.; et al. Circular RNA mediates cardiomyocyte death via miRNA-dependent upregulation of MTP18 expression. Cell Death Differ. 2017, 24, 1111–1120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Wang, S.; Li, L.; Deng, W.; Jiang, M. CircRNA MFACR Is Upregulated in Myocardial Infarction and Downregulates miR-125b to Promote Cardiomyocyte Apoptosis Induced by Hypoxia. J. Cardiovasc. Pharmacol. 2021, 78, 802–808. [Google Scholar] [CrossRef]
  68. Wang, Y.; Li, C.; Zhao, R.; Qiu, Z.; Shen, C.; Wang, Z.; Liu, W.; Zhang, W.; Ge, J.; Shi, B. CircUbe3a from M2 macrophage-derived small extracellular vesicles mediates myocardial fibrosis after acute myocardial infarction. Theranostics 2021, 11, 6315–6333. [Google Scholar] [CrossRef]
  69. Zhai, C.; Qian, G.; Wu, H.; Pan, H.; Xie, S.; Sun, Z.; Shao, P.; Tang, G.; Hu, H.; Zhang, S. Knockdown of circ_0060745 alleviates acute myocardial infarction by suppressing NF-κB activation. J. Cell. Mol. Med. 2020, 24, 12401–12410. [Google Scholar] [CrossRef]
  70. Zhang, J.; Tang, Y.; Zhang, J.; Wang, J.; He, J.; Zhang, Z.; Liu, F. CircRNA ACAP2 Is Overexpressed in Myocardial Infarction and Promotes the Maturation of miR-532 to Induce the Apoptosis of Cardiomyocyte. J. Cardiovasc. Pharmacol. 2021, 78, 247–252. [Google Scholar] [CrossRef]
  71. Zhang, M.; Wang, Z.; Cheng, Q.; Wang, Z.; Lv, X.; Wang, Z.; Li, N. Circular RNA (CircRNA) CDYL Induces Myocardial Regeneration by CeRNA after Myocardial Infarction. Med. Sci. Monit. 2020, 26, e923188-1–e923188-8. [Google Scholar] [CrossRef]
  72. Zhang, Y.; Li, Z.; Wang, J.; Chen, H.; He, R.; Wu, H. CircTRRAP Knockdown Has Cardioprotective Function in Cardiomyocytes via the Signal Regulation of miR-370-3p/PAWR Axis. Cardiovasc. Ther. 2022, 2022, 1–12. [Google Scholar] [CrossRef]
  73. Zhao, B.; Li, G.; Peng, J.; Ren, L.; Lei, L.; Ye, H.; Wang, Z.; Zhao, S. CircMACF1 Attenuates Acute Myocardial Infarction Through MiR-500b-5p-EMP1 Axis. J. Cardiovasc. Trans. Res. Cell 2020, 14, 161–172. [Google Scholar] [CrossRef]
  74. 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.; et al. 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. [Google Scholar] [CrossRef]
  75. Zhu, Y.; Zou, C.; Jia, Y.; Zhang, H.; Ma, X.; Zhang, J. Knockdown of circular RNA circMAT2B reduces oxygen-glucose deprivation-induced inflammatory injury in H9c2 cells through up-regulating miR-133. Cell Cycle 2020, 19, 2622–2630. [Google Scholar] [CrossRef]
  76. Zhu, Y.; Pan, W.; Yang, T.; Meng, X.; Jiang, Z.; Tao, L.; Wang, L. Upregulation of Circular RNA CircNFIB Attenuates Cardiac Fibrosis by Sponging miR-433. Front. Genet. 2019, 10, 564. [Google Scholar] [CrossRef] [Green Version]
  77. Nowbar, A.N.; Gitto, M.; Howard, J.P.; Francis, D.P.; Al-Lamee, R. Mortality from Ischemic Heart Disease: Analysis of Data from the World Health Organization and Coronary Artery Disease Risk Factors from NCD Risk Factor Collaboration. Circ. Cardiovasc. Qual. Outcomes 2019, 12, e005375. [Google Scholar] [CrossRef]
  78. Malakar, A.K.; Choudhury, D.; Halder, B.; Paul, P.; Uddin, A.; Chakraborty, S. A review on coronary artery disease, its risk factors, and therapeutics. J. Cell. Physiol. 2019, 234, 16812–16823. [Google Scholar] [CrossRef]
  79. Cao, Q.; Guo, Z.; Du, S.; Ling, H.; Song, C. Circular RNAs in the pathogenesis of atherosclerosis. Life Sci. 2020, 255, 117837. [Google Scholar] [CrossRef]
  80. Gong, X.; Tian, M.; Cao, N.; Yang, P.; Xu, Z.; Zheng, S.; Liao, Q.; Chen, C.; Zeng, C.; Jose, P.A.; et al. Circular RNA circEsyt2 regulates vascular smooth muscle cell remodeling via splicing regulation. J. Clin. Investig. 2021, 131, e147031. [Google Scholar] [CrossRef]
  81. Holdt, L.M.; Stahringer, A.; Sass, K.; Pichler, G.; Kulak, N.A.; Wilfert, W.; Kohlmaier, A.; Herbst, A.; Northoff, B.H.; Nicolaou, A.; et al. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat. Commun. 2016, 7, 12429. [Google Scholar] [CrossRef]
  82. 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 2019, 286, 88–96. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, Z.; Li, X.; Gao, C.; Jian, D.; Hao, P.; Rao, L.; Li, M. Peripheral blood circular RNA hsa_circ_0124644 can be used as a diagnostic biomarker of coronary artery disease. Sci. Rep. 2017, 7, 39918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Miao, L.; Yin, R.-X.; Zhang, Q.-H.; Liao, P.-J.; Wang, Y.; Nie, R.-J.; Li, H. A novel circRNA-miRNA-mRNA network identifies circ-YOD1 as a biomarker for coronary artery disease. Sci. Rep. 2019, 9, 18314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Vilades, D.; Martínez-Camblor, P.; Ferrero-Gregori, A.; Bär, C.; Lu, D.; Xiao, K.; Vea, À.; Nasarre, L.; Vega, J.S.; Leta, R.; et al. Plasma circular RNA hsa_circ_0001445 and coronary artery disease: Performance as a biomarker. FASEB J. 2020, 34, 4403–4414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Wu, W.-P.; Pan, Y.-H.; Cai, M.-Y.; Cen, J.-M.; Chen, C.; Zheng, L.; Liu, X.; Xiong, X.-D. Plasma-Derived Exosomal Circular RNA hsa_circ_0005540 as a Novel Diagnostic Biomarker for Coronary Artery Disease. Dis. Markers 2020, 2020, 1–7. [Google Scholar] [CrossRef]
  87. DeRoo, E.; Stranz, A.; Yang, H.; Hsieh, M.; Se, C.; Zhou, T. Endothelial Dysfunction in the Pathogenesis of Abdominal Aortic Aneurysm. Biomolecules 2022, 12, 509. [Google Scholar] [CrossRef]
  88. Sampson, U.K.A.; Norman, P.E.; Fowkes, F.G.R.; Aboyans, V.; Song, Y.; Harrell, F.E.H., Jr.; Forouzanfar, M.H.; Naghavi, M.; Denenberg, J.O.; McDermott, M.M.; et al. Global and Regional Burden of Aortic Dissection and Aneurysms: Mortality Trends in 21 World Regions, 1990 to 2010. Glob. Heart 2014, 9, 171–180.e10. [Google Scholar] [CrossRef]
  89. Kessler, V.; Klopf, J.; Eilenberg, W.; Neumayer, C.; Brostjan, C. AAA Revisited: A Comprehensive Review of Risk Factors, Management, and Hallmarks of Pathogenesis. Biomedicines 2022, 10, 94. [Google Scholar] [CrossRef]
  90. Wang, Z.; You, Y.; Yin, Z.; Bao, Q.; Lei, S.; Yu, J.; Xie, C.; Ye, F.; Xie, X. Burden of Aortic Aneurysm and Its Attributable Risk Factors from 1990 to 2019: An Analysis of the Global Burden of Disease Study 2019. Front. Cardiovasc. Med. 2022, 9, 901225. [Google Scholar] [CrossRef]
  91. Bath, M.F.; Gokani, V.J.; Sidloff, D.A.; Jones, L.R.; Choke, E.; Sayers, R.D.; Bown, M.J. Systematic review of cardiovascular disease and cardiovascular death in patients with a small abdominal aortic aneurysm. Br. J. Surg. 2015, 102, 866–872. [Google Scholar] [CrossRef]
  92. Haller, S.J.; Azarbal, A.F.; Rugonyi, S. Predictors of Abdominal Aortic Aneurysm Risks. Bioengineering 2020, 7, 79. [Google Scholar] [CrossRef]
  93. Miyake, T.; Miyake, T.; Kurashiki, T.; Morishita, R. Molecular Pharmacological Approaches for Treating Abdominal Aortic Aneurysm. Ann. Vasc. Dis. 2019, 12, 137–146. [Google Scholar] [CrossRef] [Green Version]
  94. Zhou, M.; Shi, Z.; Cai, L.; Li, X.; Ding, Y.; Xie, T.; Fu, W. Circular RNA expression profile and its potential regulative role in human abdominal aortic aneurysm. BMC Cardiovasc. Disord. 2020, 20, 70. [Google Scholar] [CrossRef] [Green Version]
  95. Kuivaniemi, H.; Ryer, E.J.; Elmore, J.R.; Tromp, G. Understanding the pathogenesis of abdominal aortic aneurysms. Expert Rev. Cardiovasc. Ther. 2015, 13, 975–987. [Google Scholar] [CrossRef] [Green Version]
  96. Di Gregoli, K.; Anuar, N.N.M.; Bianco, R.; White, S.J.; Newby, A.C.; George, S.J.; Johnson, J.L. MicroRNA-181b Controls Atherosclerosis and Aneurysms Through Regulation of TIMP-3 and Elastin. Circ. Res. 2017, 120, 49–65. [Google Scholar] [CrossRef] [Green Version]
  97. Sun, X.; He, S.; Wara, A.K.M.; Icli, B.; Shvartz, E.; Tesmenitsky, Y.; Belkin, N.; Li, D.; Blackwell, T.S.; Sukhova, G.K.; et al. Systemic Delivery of MicroRNA-181b Inhibits Nuclear Factor-κB Activation, Vascular Inflammation, and Atherosclerosis in Apolipoprotein E–Deficient Mice. Circ. Res. 2014, 114, 32–40. [Google Scholar] [CrossRef] [Green Version]
  98. Si, K.; Lu, D.; Tian, J. Integrated analysis and the identification of a circRNA-miRNA-mRNA network in the progression of abdominal aortic aneurysm. PeerJ 2021, 9, e12682. [Google Scholar] [CrossRef]
  99. Han, Y.; Zhang, H.; Bian, C.; Chen, C.; Tu, S.; Guo, J.; Wu, Y.; Böckler, D.; Zhang, J. Circular RNA Expression: Its Potential Regulation and Function in Abdominal Aortic Aneurysms. Oxidative Med. Cell. Longev. 2021, 2021, 1–21. [Google Scholar] [CrossRef]
  100. He, X.; Li, X.; Han, Y.; Chen, G.; Xu, T.; Cai, D.; Sun, Y.; Wang, S.; Lai, Y.; Teng, Z.; et al. CircRNA Chordc1 protects mice from abdominal aortic aneurysm by contributing to the phenotype and growth of vascular smooth muscle cells. Mol. Ther. Nucleic Acids 2022, 27, 81–98. [Google Scholar] [CrossRef]
  101. Liu, Y.; Zhong, Z.; Xiao, L.; Li, W.; Wang, Z.; Duan, Z.; Li, X. Identification of Circ-FNDC3B, an Overexpressed circRNA in Abdominal Aortic Aneurysm, as a Regulator of Vascular Smooth Muscle Cells. Int. Heart J. 2021, 62, 21–186. [Google Scholar] [CrossRef]
  102. Lv, P.; Yin, Y.-J.; Kong, P.; Cao, L.; Xi, H.; Wang, N.; Yang, H.-C.; Lv, Y.-H.; Chen, N.; Wang, R.; et al. SM22α Loss Contributes to Apoptosis of Vascular Smooth Muscle Cells via Macrophage-Derived circRasGEF1B. Oxidative Med. Cell. Longev. 2021, 2021, 1–20. [Google Scholar] [CrossRef] [PubMed]
  103. Ma, M.; Yang, X.; Han, F.; Wang, H. Circ_0092291 attenuates angiotensin II–induced cell damages in human aortic vascular smooth muscle cells via mediating the miR-626/COL4A1 signal axis. J. Physiol. Biochem. 2022, 78, 245–256. [Google Scholar] [CrossRef] [PubMed]
  104. Song, H.; Yang, Y.; Sun, Y.; Wei, G.; Zheng, H.; Chen, Y.; Cai, D.; Li, C.; Ma, Y.; Lin, Z.; et al. Circular RNA Cdyl promotes abdominal aortic aneurysm formation by inducing M1 macrophage polarization and M1-type inflammation. Mol. Ther. 2022, 30, 915–931. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, S.; Yuan, Q.; Zhao, W.; Zhou, W. Circular RNA RBM33 contributes to extracellular matrix degradation via miR-4268/EPHB2 axis in abdominal aortic aneurysm. PeerJ 2021, 9, e12232. [Google Scholar] [CrossRef] [PubMed]
  106. Yang, R.; Wang, Z.; Meng, G.; Hua, L. Circular RNA CCDC66 facilitates abdominal aortic aneurysm through the overexpression of CCDC66. Cell Biochem. Funct. 2020, 38, 830–838. [Google Scholar] [CrossRef]
  107. Yue, J.; Zhu, T.; Yang, J.; Si, Y.; Xu, X.; Fang, Y.; Fu, W. CircCBFB-mediated miR-28-5p facilitates abdominal aortic aneurysm via LYPD3 and GRIA4. Life Sci. 2020, 253, 117533. [Google Scholar] [CrossRef]
  108. Ma, X.; Xu, J.; Lu, Q.; Feng, X.; Liu, J.; Cui, C.; Song, C. Hsa_circ_0087352 promotes the inflammatory response of macrophages in abdominal aortic aneurysm by adsorbing hsa-miR-149-5p. Int. Immunopharmacol. 2022, 107, 108691. [Google Scholar] [CrossRef]
  109. Zhao, F.; Chen, T.; Jiang, N. CDR1as/miR-7/CKAP4 axis contributes to the pathogenesis of abdominal aortic aneurysm by regulating the proliferation and apoptosis of primary vascular smooth muscle cells. Exp. Ther. Med. 2020, 19, 3760–3766. [Google Scholar] [CrossRef] [Green Version]
  110. Zheng, C.; Niu, H.; Li, M.; Zhang, H.; Yang, Z.; Tian, L.; Wu, Z.; Li, D.; Chen, X. Cyclic RNA has-circ-000595 regulates apoptosis of aortic smooth muscle cells. Mol. Med. Rep. 2015, 12, 6656–6662. [Google Scholar] [CrossRef] [Green Version]
  111. Correia, C.C.M.; Rodrigues, L.F.; Pelozin, B.R.D.A.; Oliveira, E.M.; Fernandes, T. Long Non-Coding RNAs in Cardiovascular Diseases: Potential Function as Biomarkers and Therapeutic Targets of Exercise Training. Non-Coding RNA 2021, 7, 65. [Google Scholar] [CrossRef]
  112. Wang, L.; Lv, Y.; Li, G.; Xiao, J. MicroRNAs in heart and circulation during physical exercise. J. Sport Health Sci. 2018, 7, 433–441. [Google Scholar] [CrossRef]
  113. Guo, M.; Qiu, J.; Shen, F.; Wang, S.; Yu, J.; Zuo, H.; Yao, J.; Xu, S.; Hu, T.; Wang, D.; et al. Comprehensive analysis of circular RNA profiles in skeletal muscles of aging mice and after aerobic exercise intervention. Aging 2020, 12, 5071–5090. [Google Scholar] [CrossRef]
  114. Niu, Y.; Wan, C.; Zhang, J.; Zhang, S.; Zhao, Z.; Zhu, L.; Wang, X.; Ren, X.; Wang, J.; Lei, P. Aerobic exercise improves VCI through circRIMS2/miR-186/BDNF-mediated neuronal apoptosis. Mol. Med. 2021, 27, 4. [Google Scholar] [CrossRef]
  115. Meinecke, A.; Mitzka, S.; Just, A.; Cushman, S.; Stojanović, S.D.; Xiao, K.; Mooren, F.C.; Fiedler, J.; Thum, T. Cardiac endurance training alters plasma profiles of circular RNA MBOAT2. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H13–H21. [Google Scholar] [CrossRef]
  116. Zhu, Y.; Zheng, C.; Zhang, R.; Yan, J.; Li, M.; Ma, S.; Chen, K.; Chen, L.; Liu, J.; Xiu, J.; et al. Circ-Ddx60 contributes to the antihypertrophic memory of exercise hypertrophic preconditioning. J. Adv. Res. 2022, in press. [Google Scholar] [CrossRef]
  117. Lin, H.; Zhu, Y.; Zheng, C.; Hu, D.; Ma, S.; Chen, L.; Wang, Q.; Chen, Z.; Xie, J.; Yan, Y.; et al. Antihypertrophic Memory After Regression of Exercise-Induced Physiological Myocardial Hypertrophy Is Mediated by the Long Noncoding RNA Mhrt779. Circulation 2021, 143, 2277–2292. [Google Scholar] [CrossRef]
  118. Yu, Z.; Huang, Q.; Zhang, Q.; Wu, H.; Zhong, Z. CircRNAs open a new era in the study of cardiovascular disease (Review). Int. J. Mol. Med. 2021, 47, 49–64. [Google Scholar] [CrossRef]
Figure 1. Biogenesis, forms, functions, and biological roles of circRNAs. miRNA—microRNAs; RBP—RNA-binding proteins.
Figure 1. Biogenesis, forms, functions, and biological roles of circRNAs. miRNA—microRNAs; RBP—RNA-binding proteins.
Ijms 24 02125 g001
Figure 2. circRNAs associated with human CVDs. AH—arterial hypertension, MI—myocardial infarction, I/R—ischemia/reperfusion injury, CAD—coronary artery disease, AAA—abdominal aortic aneurysm. ↑—upregulated, ↓—downregulated.
Figure 2. circRNAs associated with human CVDs. AH—arterial hypertension, MI—myocardial infarction, I/R—ischemia/reperfusion injury, CAD—coronary artery disease, AAA—abdominal aortic aneurysm. ↑—upregulated, ↓—downregulated.
Ijms 24 02125 g002
Table 1. List of circRNAs associated with arterial hypertension.
Table 1. List of circRNAs associated with arterial hypertension.
IdHost GeneSpeciesSourceExpressionActionRef.
hsa_circ_0037909GSPT1HumanBlood, HAECs, HUVECsUpSponges hsa-miR-637, possibly impacting in LDL and serum creatinine concentrations.[29]
hsa_circ_0037911GSPT1HumanBloodUpBiomarker.[30]
rno_circ_006016Erc2RatKidneyDownControls blood pressure by multiple circRNA–miRNA–gene interactions.[31]
hsa_circ_0105015GSPT1HumanBlood
Endothelial cells
UpPromotes target hsa-miR-637 to activate the inflammatory pathway.[32]
hsa_circ_0126991SEPT11HumanBloodUpBiomarker.[33]
rno_circ_0009197Cdh23RatAortic vascular tissuesDownAortic circRNAs play potential roles in regulating hypertensive vascular remodeling and dysfunction.[24]
rno_circ_0005818Dnajc1Up
rno_circ_0005304-Up
rno_circ_0005506Ryr2Up
rno_circ_0009301-Up
circACTA2ACTA2HumanArtery tissuesUpPromotes vascular smooth muscle cell senescence by targeting circACTA2/ILF3/CDK4 axis.[25]
HumanHASMCsUpRegulates α-SMA expression.[28]
hsa_circ_0037897GSPT1HumanBloodUpMay be involved in hypertension by sponging hsa-miR-145-5p.[34]
hsa_circ_0014243CHTOPHumanBloodUpPlays a crucial role in genesis and development of hypertension. Could be used as biomarker.[35]
has_circ_0005870SETD2HumanPlasmaDownBiomarker.[36]
HAECs—human aortic endothelial cells; HUVECs—human umbilical vein endothelial cells; HASMCs—human aortic smooth muscle cells.
Table 4. List of circRNAs associated with AAA.
Table 4. List of circRNAs associated with AAA.
IdHost GeneSpeciesSourceExpressionActionRef.
circChordc1Chordc1MouseAbdominal aortaDownImproves VSMCs growth, suppressing aneurysm formation and reducing the risk of rupture by inducing vimentin degradation.[100]
circ-FNDC3BFNDC3BHumanAortic tissueUpPromotes VSMCs inflammation and oxidative stress through miR-143-3p/ADAM10 axis, leading to the development of AAA.[101]
circRasGEF1BRasgef1bMouseAbdominal aortaUpcircRasGEF1B-ZFP36 axis mediates macrophage-induced VSMC apoptosis in an Sm22α-/- mice AAA model.[102]
circ_0092291EIF2S2HumanBlood
HAVSMC
DownInhibits AAA-associated cell damage via circ_0092291/miR-626/COL4A1 axis.[103]
circCdylCdylMouseAbdominal aortaUpPromotes vascular inflammation through M1-type macrophages polarization, inducing AAA formation.[104]
circRBM33RBM33HumanAbdominal aortaUpPromotes ECM degradation in aorta-isolated VSMCs via the miR-4268/EPHB2 axis.[105]
circCCDC66CCDC6 6HumanVSMCsUpUpregulates its host gene, modulating VSMCs proliferation and apoptosis and facilitating AAA development.[106]
circCBFBCBFBHumanHAVSMC-Its suppression increased the expression of miR-28-5p, promoting apoptosis of VSMCs.[107]
hsa_circ_0087352UBQLN1HumanTHP-1 cells
HAVSMC
UpPromotes inflammatory response in macrophages, enhancing the expression and secretion of IL-6 and TNF-α by sponging hsa-miR-149-5p.[108]
CircCDR1asCDR1HumanAortic tissues
VSMCs
DownRegulates VSMC apoptosis and proliferation via circCDR1as/miR-7/CKAP4 axis.[109]
hsa_circ_000595-HumanAortic tissues
HAVSMC
UpIts knockdown decreased apoptosis in HAVSMC.[110]
VSMC—vascular smooth muscle cell; AAA—abdominal aorta aneurysm; Sm22α-/-—smooth muscle knockout; HAVSMC—human aortic vascular muscle cell; ECM—extracellular matrix; THP-1—human leukemia monocytic cell line; IL-6—interleukin 6; TNF-α—tumor necrosis factor alpha.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Joaquim, V.H.A.; Pereira, N.P.; Fernandes, T.; Oliveira, E.M. Circular RNAs as a Diagnostic and Therapeutic Target in Cardiovascular Diseases. Int. J. Mol. Sci. 2023, 24, 2125. https://doi.org/10.3390/ijms24032125

AMA Style

Joaquim VHA, Pereira NP, Fernandes T, Oliveira EM. Circular RNAs as a Diagnostic and Therapeutic Target in Cardiovascular Diseases. International Journal of Molecular Sciences. 2023; 24(3):2125. https://doi.org/10.3390/ijms24032125

Chicago/Turabian Style

Joaquim, Victor Hugo Antonio, Noemy Pinto Pereira, Tiago Fernandes, and Edilamar Menezes Oliveira. 2023. "Circular RNAs as a Diagnostic and Therapeutic Target in Cardiovascular Diseases" International Journal of Molecular Sciences 24, no. 3: 2125. https://doi.org/10.3390/ijms24032125

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop