Cancer Letters

Cancer Letters

Volume 404, 28 September 2017, Pages 8-18
Cancer Letters

Mini-review
Recent progress in circular RNAs in human cancers

https://doi.org/10.1016/j.canlet.2017.07.002Get rights and content

Highlights

  • CircRNAs are no longer regarded as “non-coding” RNA, since they can be translated to protein driven by m6A modification.

  • For most circRNAs rarely contain miRNA target sites, miRNA sponge might be not the predominant mode of circRNA function.

  • CircRNAs were found to be dysregulated in cancer cell lines, tumor tissues, and even plasma samples from patients.

  • CircRNAs were deemed to be promising biomarkers for the early diagnosis and prognosis prediction of cancer.

Abstract

Circular RNAs (circRNAs) are a large class of endogenous RNAs, formed by exon skipping or back-splicing events as covalently closed loops, which are expressed abundantly in mammalian cells. Although their biological functions remain largely unknown, recent studies show that circRNAs have three main functions in mammalian cells. First, circRNAs can regulate transcription and RNA splicing. Second, circRNAs function as microRNA (miRNA) sponges. Third, they can be translated into protein driven by N6-methyladenosine modification. Taking advantage of RNA sequencing (RNA-seq) technology, the expressions of circRNAs were found to be dysregulated in all types of cancer cell lines, tumor tissues, and even plasma samples from patients, which correlated with certain clinical characteristics, suggesting the potential roles of circRNAs in tumor progression. Considering their conserved sequences and stable structures, circRNAs were deemed to be promising biomarkers for the early diagnosis and prognosis prediction of cancer. In this review, we describe briefly the formation and properties of circRNAs, and focus mainly on recent progress in research into their function, regulation, and clinical relevance in different cancers.

Introduction

Circular RNAs (circRNAs) are a large class of endogenous RNAs that are formed by exon skipping or back-splicing events; however, they attracted little attention until their function in post-transcriptional regulation of gene expression was discovered. Potato spindle tuber viroid (PSTVd) was the first identified circRNA in 1976 when researchers studying potato spindle tuber disease observed that the viroid could infect plants and cause death. Different from viruses, the viroid lacks a protein envelope and the genome is a closed, single-stranded RNA molecule [1]. In 1979, Hsu and Coca-Prados observed the presence of a circular form of RNA in the cytoplasm of several eukaryotic cells using electron microscopy [2].

In the early 1990s, circRNAs in higher eukaryotes were discovered. The first clue to the mechanism of endogenous circRNA generation emerged from studies of the transcripts of the tumor suppressor gene DCC. Very low levels of transcripts of the DCC gene with exons joined accurately at consensus splice sites were found in normal and neoplastic cells, primarily in the nonpolyadenylated component of cytoplasmic RNA. These results demonstrated that the splicing process does not always pair sequential exons in the order predicted from their positions in the genome, which was called “exon scrambling” [3]. Cocquerelle et al. identified a scrambled transcript of the human c-ets-1 gene, which is non-polyadenylated and is expressed at much lower levels than the normal transcript [4]. They further determined the structure of these transcripts as circular RNA molecules containing only exons in the genomic order [5]. In adult mouse testis, circular transcripts of the testis-determining gene sex-determining region Y (Sry) were detected. These circular RNAs, which represent the most abundant transcript in the testis, are located in the cytoplasm, but are not bound substantially to polysomes [6]. Later, atypical RNA molecules containing an incomplete exon tandem repetition, or having exons with a different order compared with the corresponding genomic DNA, were identified from the Drosophila melanogaster muscleblind (mbl) locus. Considering its lack of polyadenylation and downstream splicing events, its small size, and polyacrylamide gel electrophoresis (PAGE) behavior, the non-canonical transcript mblE2E2′ was deemed to be the first identified circular RNA in invertebrates [7].

Recently, taking advantage of RNA-seq technology and bioinformatic tools, more circRNAs have been discovered and characterized. In 2012, Salzman et al. developed an algorithm to detect scrambled exons in RNA-seq datasets of five bone marrow samples from pediatric acute lymphoblastic leukemia (ALL). They identified more than 1232 genes with evidence of exon scrambling, which were further validated by reverse transcription polymerase chain reaction (RT-PCR). Intriguingly, all examples of exon scrambling were also detected in peripheral blood collected from the same ALL patients and H9 ES cells, providing strong support for the view that a circular RNA isoform resulting from a non-canonical mode of RNA splicing is actually a general feature of the gene expression program in diverse human cells [8]. This group further developed a new bioinformatic approach and investigated circRNA expression using published RNA-Seq data from Drosophila brains and a series of cancer and non-cancer cell lines. They showed that circRNA expression was conserved evolutionarily across model organisms. In addition, the expression profiles, the ratio of circular to linear transcripts, and the pattern of splice isoforms of circRNAs of individual genes were cell-type specific. They also estimated that circular RNAs might account for about 1% of poly(A) RNA in humans [9]. Jeck et al. identified over 25,000 distinct RNAs containing non-linear exons from 14.4% of actively transcribed genes in human fibroblasts. Surprisingly, the abundance of certain circRNAs was 10-fold more than that of associated linear mRNAs. Bioinformatic analysis revealed that these circularized exons were always flanked by long introns that contained complementary ALU repeats. Moreover, they found that circRNAs could be degraded by siRNAs, suggesting their potential role as competing endogenous RNAs [10], [11]. Combining ribominus sequencing data for HEK293 cells and human leukocyte data, Memczak et al. identified 1950 kinds of human circRNAs from at least two independent junction-spanning reads. They also identified 1903 mouse circRNAs (81 of these mapped to human circRNAs) and 724 circRNAs from various C. elegans developmental stages. Further studies focused on a human circRNA, which is antisense to the cerebellar degeneration-related protein 1 transcript (CDR1as), and identified the regulatory potential of circRNA as an miRNA antagonist [12]. Thereafter, they sequenced RNA in human peripheral whole blood and detected thousands of circRNAs reproducibly. Hundreds of circRNAs were expressed at much higher levels than the corresponding linear mRNAs, which were not accessible by classical mRNA specific assays, demonstrating the potential of circRNAs as biomarkers for human disease in an easily accessible body fluid [13].

Section snippets

The biogenesis of CircRNAs

Most eukaryotic genes are split genes, in which exons are interrupted by sections of introns, so the precursor mRNA (pre-mRNA) transcripts must be modified such that non-coding introns are removed and protein coding exons are joined together. In rare cases, the splicing machinery fails to join the 3′ end of one exon to the 5′ end of the next and instead, appears to mis-splice by, for example, connecting the downstream 5′ splice site (5′ ss) to an upstream 3′ splice site (3′ ss), thereby

The biological function of CircRNAs

Although thousands of circRNAs have been identified using RNA-seq in diverse cell types from several model organisms, and circular transcripts are the predominant isoforms of hundreds of human genes, the biological functions of most circRNAs remain unknown. Recent studies have demonstrated that circRNAs have important physiological functions via binding to RBPs or other proteins, neutralizing endogenous miRNAs, or even translation into proteins, implying that circRNAs may regulate gene

CircRNAs in human cancers

Unlike conventional linear RNAs that contain 5′ and 3′ ends, circRNA molecules with closed ring structure are resistant to degradation by cellular RNA decay machineries, which recognize the ends of linear RNAs [8], [38]. CircRNAs are also enriched and stable in exosomes [39]. Thus, because of their extensive distribution, stability, cell type-specific and tissue-specific expression, circRNAs might serve as novel and promising biomarkers for the diagnosis and prognosis of human diseases,

Perspectives

Currently, researchers recognize that circular RNAs are abundant and conserved endogenous RNAs with extensive distribution, cell type-specificity, tissue-specific expression, and multiple functions. Using high-throughput sequencing, many dysregulated circRNAs have been identified in various types of cancer. However, little is known about their function and molecular mechanisms during cancer initiation, progression, and metastasis. Currently, most available or potential cancer biomarkers are not

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (81372149, 81401894, 81402289), the National Science Foundation Projects of Guangdong Province (2014A030313547), the Outstanding Young Teachers Project in Colleges and Universities of Guangdong Province (YQ2015144), and the Shenzhen Municipal Government of China (JCYJ20160427105140594, KQCX20140519104925300, JCYJ20140418091413510, JCYJ20160307155641741, JCYJ20160422091914681, GJHZ20160301164637011).

References (91)

  • M. Zhu et al.

    Circular BANP, an upregulated circular RNA that modulates cell proliferation in colorectal cancer

    Biomed. Pharmacother.

    (2017)
  • N.Y. Chia et al.

    Molecular classification of gastric cancer

    Ann. Oncol.

    (2016)
  • P.F. Li et al.

    Using circular RNA as a novel type of biomarker in the screening of gastric cancer

    Clin. Chim. Acta

    (2015)
  • S. Chen et al.

    Using circular RNA hsa_circ_0000190 as a new biomarker in the diagnosis of gastric cancer

    Clin. Chim. Acta

    (2017)
  • J. Chen et al.

    Circular RNA profile identifies circPVT1 as a proliferative factor and prognostic marker in gastric cancer

    Cancer Lett.

    (2017)
  • S. Qu et al.

    Microarray expression profile of circular RNAs in human pancreatic ductal adenocarcinoma

    Genom. Data

    (2015)
  • M. Sand et al.

    Circular RNA expression in cutaneous squamous cell carcinoma

    J. Dermatol. Sci.

    (2016)
  • H.L. Sanger et al.

    Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures

    Proc. Natl. Acad. Sci. U. S. A.

    (1976)
  • M.T. Hsu et al.

    Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells

    Nature

    (1979)
  • C. Cocquerelle et al.

    Splicing with inverted order of exons occurs proximal to large introns

    EMBO J.

    (1992)
  • C. Cocquerelle et al.

    Mis-splicing yields circular RNA molecules

    FASEB J.

    (1993)
  • J.M. Houseley et al.

    Noncanonical RNAs from transcripts of the Drosophila muscleblind gene

    J. Hered.

    (2006)
  • J. Salzman et al.

    Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types

    PLoS One

    (2012)
  • J. Salzman et al.

    Cell-type specific features of circular RNA expression

    PLoS Genet.

    (2013)
  • W.R. Jeck et al.

    Circular RNAs are abundant, conserved, and associated with ALU repeats

    RNA

    (2013)
  • W.R. Jeck et al.

    Detecting and characterizing circular RNAs

    Nat. Biotechnol.

    (2014)
  • S. Memczak et al.

    Circular RNAs are a large class of animal RNAs with regulatory potency

    Nature

    (2013)
  • S. Memczak et al.

    Identification and characterization of circular RNAs as a new class of putative biomarkers in human blood

    PLoS One

    (2015)
  • J.U. Guo et al.

    Expanded identification and characterization of mammalian circular RNAs

    Genome Biol.

    (2014)
  • Z. Li et al.

    Exon-intron circular RNAs regulate transcription in the nucleus

    Nat. Struct. Mol. Biol.

    (2015)
  • S.R. Salgia et al.

    Two reactions of Haloferax volcanii RNA splicing enzymes: joining of exons and circularization of introns

    RNA

    (2003)
  • Z. Lu et al.

    Metazoan tRNA introns generate stable circular RNAs in vivo

    RNA

    (2015)
  • U. Braunschweig et al.

    Widespread intron retention in mammals functionally tunes transcriptomes

    Genome Res.

    (2014)
  • C.W. Chao et al.

    The mouse formin (Fmn) gene: abundant circular RNA transcripts and gene-targeted deletion analysis

    Mol. Med.

    (1998)
  • W.W. Du et al.

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

    Nucleic Acids Res.

    (2016)
  • T.B. Hansen et al.

    Natural RNA circles function as efficient microRNA sponges

    Nature

    (2013)
  • Q.P. Zheng et al.

    Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging multiple miRNAs

    Nat. Commun.

    (2016)
  • X. You et al.

    Neural circular RNAs are derived from synaptic genes and regulated by development and plasticity

    Nat. Neurosci.

    (2015)
  • A. Kos et al.

    The hepatitis delta (delta) virus possesses a circular RNA

    Nature

    (1986)
  • M.G. AbouHaidar et al.

    Novel coding, translation, and gene expression of a replicating covalently closed circular RNA of 220 nt

    Proc. Natl. Acad. Sci. U. S. A.

    (2014)
  • C.Y. Chen et al.

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

    Science

    (1995)
  • R. Perriman et al.

    Circular mRNA can direct translation of extremely long repeating-sequence proteins in vivo

    RNA Publ. RNA Soc.

    (1998)
  • Y. Wang et al.

    Efficient backsplicing produces translatable circular mRNAs

    RNA

    (2015)
  • Y. Yang et al.

    Extensive translation of circular RNAs driven by N6-methyladenosine

    Cell Res.

    (2017)
  • J.E. Wilusz et al.

    Molecular biology. A circuitous route to noncoding RNA

    Science

    (2013)
  • Cited by (96)

    • Dynamic m6A-ncRNAs association and their impact on cancer pathogenesis, immune regulation and therapeutic response

      2023, Genes and Diseases
      Citation Excerpt :

      Consequently, the increased circNSUN2 in the cytoplasm specifically interacted with IGF2BP2, thus stabilizing its downstream target HMGA2 and promoting CRC cell aggressiveness.116 Altered m6A regulators may contribute to the aberrant expression profiles of circRNAs seen in disease pathogenesis and progression.117,118 The majority of circRNAs are synthesized by a co-transcriptional back-splicing pattern.119,120

    • CircRNA: An emerging star in the progression of glioma

      2022, Biomedicine and Pharmacotherapy
    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text