Circular RNAs in cardiovascular diseases

https://doi.org/10.1016/j.pharmthera.2021.107991Get rights and content

Abstract

In eukaryotes, precursor mRNAs (pre-mRNAs) produce a unique class of biologically active molecules namely circular RNAs (circRNAs) with a covalently closed-loop structure via back-splicing. Because of this unconventional circular form, circRNAs exhibit much higher stability than linear RNAs due to the resistance to exonuclease degradation and thereby play exclusive cellular regulatory roles. Recent studies have shown that circRNAs are widely expressed in eukaryotes and display tissue- and disease-specific expression patterns, including in the cardiovascular system. Although numerous circRNAs are discovered by in silico methods, a limited number of circRNAs have been studied. This review intends to summarize the current understanding of the characteristics, biogenesis, and functions of circRNAs and delineate the practical approaches for circRNAs investigation. Moreover, we discuss the emerging roles of circRNAs in cardiovascular diseases.

Introduction

Transcriptomic analyses have revealed that human genome can transcribe ~90% of the genomic DNA, but only 1-2% of transcripts encode proteins, whereas a large amount of the transcripts are categorized as noncoding RNA (ncRNAs) (Consortium, 2004). Emerging evidence has shown that aberrant ncRNA expression is associated with a number of diseases, including developmental, oncological, neurological, cardiovascular, autoimmune, and cutaneous disorders (Amaral & Mattick, 2008; Esteller, 2011; Li, Baptista, & Kissinger, 2020; Mattick & Rinn, 2015; Mehler & Mattick, 2007; Tang et al., 2019; Tang et al., 2020; Tang, Zhang, Wang, Mei, & Chen, 2017; Uchida & Dimmeler, 2015; Wu et al., 2015). Recently, a novel class of ncRNA, circRNA, has drawn much attention from researchers in different fields, ascribed to its unusual structure and diverse biological features compared to other long noncoding RNAs. The major difference between circRNAs and long noncoding RNAs is their structure, in which circRNAs show a covalently closed continuous loop without 5′–3′ polarity, but long noncoding RNAs show linear form with or without the polyadenylated tail. With this unique form, circRNAs have longer half-life than long noncoding RNAs. Another difference is the cellular location. Compared with long noncoding RNAs, a large portion of circRNAs is in the cytoplasm.

The first circular form of RNA molecule was reported in 1976, due to the discovery of plant viroids that are covalently closed circular RNA molecules. Two ends of viroids were ligated by host cellular enzymes (Sanger, Klotz, Riesner, Gross, & Kleinschmidt, 1976). Since then, sporadic studies have identified a number of circRNAs in different species such as humans, mice, and rats, although most of them were considered as byproducts of RNA splicing or so-called “scrambled exons” (Capel et al., 1993; Cocquerelle, Daubersies, Majerus, Kerckaert, & Bailleul, 1992; Hsu & Coca-Prados, 1979; Nigro et al., 1991; Patop, Wust, & Kadener, 2019; Zaphiropoulos, 1996). In the 2000s, advances in next-generation sequencing (NGS) and computational pipelines led to an explosion of RNA-related researches and the scale of NGS datasets (Mardis, 2008). However, due to the lack of polyadenylation tail (poly-A tail), circRNAs have not caught the attention of scientists in the first wave of NGS, which has mainly focused on the polyadenylated transcriptomes. Starting in the early 2010s, new pre-sequencing approaches were developed to preserve non-polyadenylated RNAs, which revealed that circRNAs are expressed in many species, including mice, rats, pigs, monkeys, humans, zebrafish, and fruitflies, as well as plants, fungi, and protists, etc (Barrett, Wang, & Salzman, 2015; Broadbent et al., 2015; Dong, Ma, Chen, & Yang, 2017; Fan et al., 2015; Guo, Agarwal, Guo, & Bartel, 2014; Jeck et al., 2013; Lu et al., 2015; Luo et al., 2018; Memczak et al., 2013; Salzman, Gawad, Wang, Lacayo, & Brown, 2012; Shen, Guo, & Wang, 2017; Westholm et al., 2014; Yang, Duff, Graveley, Carmichael, & Chen, 2011; Zhang et al., 2014). So far, more than 183,000 circRNAs have been identified in human tissues/cells (Dong, Ma, Li, & Yang, 2018). Unlike linear mRNAs that are generated from precursor mRNAs (pre-mRNAs) and undergo normal splicing following RNA polymerase II (Pol II)-mediated transcription, circRNAs are formed via back-splicing, in which a downstream 5′ splice site is ligated to an upstream 3′ splice site across exon or exons (Black, 2003; Li, Yang, & Chen, 2018). Besides, intron lariats derived from the normal splicing process erratically escape from debranching at times and cause a circular formation and are referred as circular intronic RNAs (ciRNAs) (Zhang et al., 2013). Additionally, another type of circRNAs called exon-intronic circRNAs or exon-intron-containing-circular-RNAs (EIciRNAs) is predominantly localized in the nucleus and retrain intronic information. This type of circRNAs is largely caused by escaping of EIciRNAs from decompression of debranching enzymes following splicing based on the nucleotide information in these introns (Geng, Jiang and Wu, 2018, Li et al., 2015).

Given the unique characteristics, circRNAs exhibit excellent stability compared to the linear mRNA transcripts because they are resistant to exonucleolytic decay by the exosome ribonucleases (Szabo & Salzman, 2016). The average half-life of circRNAs ranges from 19 to 48 hours, whereas linear RNA transcripts only have a lifetime of 4-9 hours (Enuka et al., 2016; Jeck et al., 2013; Schwanhausser et al., 2011). Therefore, despite a relatively low expression level compared to the linear RNAs, circRNAs have shown disease- and tissue-specific expression patterns and extraordinary potential in modulating the pathogenesis of diseases, such as oncological, cardiovascular, neurodegenerative, and immune disorders (Aufiero, Reckman, Pinto, & Creemers, 2019; Chen et al., 2020; Kumar, Shamsuzzama Haque, Baghel, & Nazir, 2017; Li et al., 2019; Saaoud et al., 2021; Shang, Yang, Jia, & Ge, 2019). In this review, we provide an overview of the recent progress in circRNA biology, including characteristics, biogenesis, and functions of circRNAs, as well as strategies for investigating circRNAs and up-to-date available online resources. Moreover, the emerging roles of circRNAs in cardiovascular diseases are discussed.

Section snippets

Characteristics of circRNAs

CircRNAs are considered to be very diverse and referred as a heterogeneous class of RNAs. The co- or post-transcriptional back-splicing processes the circularization of RNA molecules (Zhang, Yang, & Chen, 2016). A circRNA typically consists of exons only, introns only, or a mixture of exons and introns. Using NGS, Zhang et al., 2014 revealed several key genomic features of circRNAs. Even though particular motifs are not required, single exon circularization has to meet the minimal length

Biogenesis of circRNAs

Back-splicing is the essential step of circRNA formation, which occurs at the co- and post-transcriptional level (Zhang, Yang, & Chen, 2016). As an atypical alternative splicing event, back-splicing of exons is coupled with Pol II-mediated transcription and regulated by spliceosome machinery, evidenced by a study identifying a mutual competition between splicing factor muscleblind (MBL) and circMbl that is derived from MBL linear transcripts (Ashwal-Fluss et al., 2014; Starke et al., 2015).

Functions, translocation, and degradation of circRNAs

Due to the relatively low abundance of circRNAs, circRNAs initially were thought to be the byproduct of splicing or “junk transcripts” (Liu, Liu, Wang, & He, 2017). However, recent blooming studies of circRNAs have completely reshaped the perspective and suggested that circRNAs not only feature unique characteristics but also play potent roles in regulating physiological processes (Aufiero et al., 2019; Chen, 2020; Li, Yanfang, et al., 2018).

Detection of circRNAs

Recent advances in NGS technologies and the booming of bioinformatics have given rise to an expansion of circRNA-related studies. In 2012, the first genome-wide profiling of circRNAs by RNA-Seq was exposed to the public (Salzman et al., 2012). A year later, a landmark study conducted by Memczak et al. (Memczak et al., 2013) not only demonstrated that circRNAs might act as miRNA sponge and associate with certain RBPs but also developed the first computational pipeline-find_circ, which allow ones

Myocardial infarction

Among different types of cardiovascular diseases, myocardial infarction (MI) is the leading cause of mortality and disability (Hausenloy & Yellon, 2013). MI is characterized by cardiac damage and remodeling on account of acute occlusion of the coronary artery, followed by prolonged myocardial ischemia that leads to myocardial cell death, cardiomyocyte loss, cardiac dysfunction, and ultimately heart failure (Yeap, Dehn, Adelman, Lipsitz, & Thorp, 2013). CircRNAs play important roles in MI and

CircRNA-related databases and online resources

Database and online resources proffer opportunities to get access to large-scale datasets and valuable analysis tools. Currently, there are more than 20 mammalian circRNA online database portals. Most of these databases store circRNA information from numerous NGS datasets based on literature, and others provide practical analysis pipelines to process NGS data (Table 4). Since the number of circRNA databases is still growing, the content and quality need to be updated or improved frequently (

Summary and perspective

Once regarded as scramble byproduct of splicing with no biological functions, circRNAs have become the research hotspot in recent years. Even though exciting progress has been achieved, the biological functions of the majority of circRNAs remain unclear. The exact mechanisms controlling circRNA biogenesis are also elusive. Since back-splicing is very different from canonical splicing, there must be novel factors specifically modulating back-splicing. Since strategies for dissecting the

Declaration of Competing Interest

Xiaohan Mei & Shi-You Chen declare that there are no conflicts of interest.

Acknowledgments

This work was supported by grants from National Institutes of Health (HL119053, HL135854, HL147313, and HL117247 to S.-Y.C.).

Reference (230)

  • A. Ivanov et al.

    Analysis of intron sequences reveals hallmarks of circular RNA biogenesis in animals

    Cell Reports

    (2015)
  • A. Lavenniah et al.

    Engineered circular RNA sponges act as miRNA inhibitors to attenuate pressure overload-induced cardiac hypertrophy

    Molecular Therapy

    (2020)
  • H. Li et al.

    riboCIRC: A comprehensive database of translatable circRNAs

    Genome Biology

    (2021)
  • X. Li et al.

    The biogenesis, functions, and challenges of circular RNAs

    Molecular Cell

    (2018)
  • Y. Li et al.

    Noncoding RNAs in apicomplexan parasites: An update

    Trends in Parasitology

    (2020)
  • Z. Li et al.

    Tumor-released exosomal circular RNA PDE8A promotes invasive growth via the miR-338/MACC1/MET pathway in pancreatic cancer

    Cancer Letters

    (2018)
  • W.C. Liang et al.

    Translation of the circular RNA circbeta-catenin promotes liver cancer cell growth through activation of the Wnt pathway

    Genome Biology

    (2019)
  • K. Abdelmohsen et al.

    Identification of HuR target circular RNAs uncovers suppression of PABPN1 translation by CircPABPN1

    RNA Biology

    (2017)
  • T. Aktas et al.

    DHX9 suppresses RNA processing defects originating from the Alu invasion of the human genome

    Nature

    (2017)
  • M.R. Alexander et al.

    Epigenetic control of smooth muscle cell differentiation and phenotypic switching in vascular development and disease

    Annual Review of Physiology

    (2012)
  • P.P. Amaral et al.

    Noncoding RNA in development

    Mammalian Genome

    (2008)
  • S. Aufiero et al.

    Circular RNAs open a new chapter in cardiovascular biology

    Nature Reviews. Cardiology

    (2019)
  • S. Aufiero et al.

    Cardiac circRNAs arise mainly from constitutive exons rather than alternatively spliced exons

    RNA

    (2018)
  • A. Bachmayr-Heyda et al.

    Correlation of circular RNA abundance with proliferation--exemplified with colorectal and ovarian cancer, idiopathic lung fibrosis, and normal human tissues

    Scientific Reports

    (2015)
  • Y. Bai et al.

    Circular RNA DLGAP4 ameliorates ischemic stroke outcomes by targeting miR-143 to regulate endothelial-mesenchymal transition associated with blood-brain barrier integrity

    The Journal of Neuroscience

    (2018)
  • S.P. Barrett et al.

    Circular RNA biogenesis can proceed through an exon-containing lariat precursor

    Elife

    (2015)
  • H.A. Bazan et al.

    Carotid plaque rupture is accompanied by an increase in the ratio of serum circR-284 to miR-221 levels

    Circulation. Cardiovascular Genetics

    (2017)
  • H.A. Bazan et al.

    Acute loss of miR-221 and miR-222 in the atherosclerotic plaque shoulder accompanies plaque rupture

    Stroke

    (2015)
  • D.L. Black

    Mechanisms of alternative pre-messenger RNA splicing

    Annual Review of Biochemistry

    (2003)
  • K.M. Broadbent et al.

    Strand-specific RNA sequencing in Plasmodium falciparum malaria identifies developmentally regulated long non-coding RNA and circular RNA

    BMC Genomics

    (2015)
  • C.Y. Chen et al.

    Initiation of protein synthesis by the eukaryotic translational apparatus on circular RNAs

    Science

    (1995)
  • L. Chen et al.

    The bioinformatics toolbox for circRNA discovery and analysis

    Briefings in Bioinformatics

    (2020)
  • L.L. Chen

    The expanding regulatory mechanisms and cellular functions of circular RNAs

    Nature Reviews. Molecular Cell Biology

    (2020)
  • N. Chen et al.

    A novel FLI1 exonic circular RNA promotes metastasis in breast cancer by coordinately regulating TET1 and DNMT1

    Genome Biology

    (2018)
  • S. Chen et al.

    Widespread and functional RNA circularization in localized prostate cancer

    Cell

    (2019)
  • X. Chen et al.

    circRNADb: A comprehensive database for human circular RNAs with protein-coding annotations

    Scientific Reports

    (2016)
  • C. Cocquerelle et al.

    Splicing with inverted order of exons occurs proximal to large introns

    The EMBO Journal

    (1992)
  • V.M. Conn et al.

    A circRNA from SEPALLATA3 regulates splicing of its cognate mRNA through R-loop formation

    Nat Plants

    (2017)
  • Consortium, E. P

    The ENCODE (ENCyclopedia Of DNA Elements) Project

    Science

    (2004)
  • W.F. Dietrich et al.

    A comprehensive genetic map of the mouse genome

    Nature

    (1996)
  • R. Dong et al.

    Increased complexity of circRNA expression during species evolution

    RNA Biology

    (2017)
  • Y. Dou et al.

    Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes

    Scientific Reports

    (2016)
  • W.W. Du et al.

    Foxo3 circular RNA promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses

    European Heart Journal

    (2017)
  • W.W. Du et al.

    Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2

    Nucleic Acids Research

    (2016)
  • D.B. Dudekula et al.

    CircInteractome: A web tool for exploring circular RNAs and their interacting proteins and microRNAs

    RNA Biology

    (2016)
  • C.E. Dunbar et al.

    Gene therapy comes of age

    Science

    (2018)
  • Y. Enuka et al.

    Circular RNAs are long-lived and display only minimal early alterations in response to a growth factor

    Nucleic Acids Research

    (2016)
  • L. Errichelli et al.

    FUS affects circular RNA expression in murine embryonic stem cell-derived motor neurons

    Nature Communications

    (2017)
  • M. Esteller

    Non-coding RNAs in human disease

    Nature Reviews. Genetics

    (2011)
  • C. Fan et al.

    CircR2Disease: A manually curated database for experimentally supported circular RNAs associated with various diseases

    Database: The Journal of Biological Databases and Curation

    (2018)
  • Cited by (17)

    • Non-coding RNAs in gynecologic cancer

      2023, Clinica Chimica Acta
    • Circular RNA in multiple myeloma: A new target for therapeutic intervention

      2022, Pathology Research and Practice
      Citation Excerpt :

      Circular RNAs (circRNAs) are universal and diversified endogenous non-coding RNAs found in all eukaryotic cells [1,2].

    View all citing articles on Scopus
    View full text