Fact-checked by Grok 2 weeks ago

Circular RNA

Circular RNAs (circRNAs) are a class of endogenous, single-stranded molecules that form a covalently closed, continuous loop structure without free 5′ or 3′ ends, generated primarily through back-splicing of precursor mRNA (pre-mRNA) exons. This unique topology confers exceptional stability, with half-lives of 19–24 hours compared to 4–7 hours for linear mRNAs, rendering them resistant to degradation. CircRNAs are ubiquitously expressed across , from viruses to mammals, and represent a substantial portion of the , with over 100,000 distinct circRNAs identified in neurons alone. The biogenesis of circRNAs involves canonical splicing machinery but proceeds in a reverse direction, where a downstream splice donor joins an upstream splice acceptor, often facilitated by complementary intronic sequences or RNA-binding proteins such as Quaking (QKI) or Muscleblind (MBL). They are classified into major types, including exonic circRNAs (ecircRNAs), which consist primarily of spliced exons; exonic-intronic circRNAs (EIciRNAs), retaining introns; circular intronic RNAs (ciRNAs), derived from lariat introns; and tRNA intronic circRNAs (tricRNAs) from pre-tRNA splicing. Post-transcriptional modifications, such as N6-methyladenosine (m6A), further regulate their export, localization, and degradation via pathways involving YTH domain-containing proteins and RNases. Functionally, circRNAs exert diverse regulatory roles, including acting as (miRNA) sponges to sequester and inhibit miRNA activity—exemplified by CDR1as, which harbors over 70 binding sites for miR-7—thereby modulating . They also interact with proteins to form complexes that influence transcription, such as circPAIP2 enhancing transcription of its parental , or , where select circRNAs like circFBXW7 encode functional peptides via internal entry sites (IRES) or m6A-driven mechanisms. In the mammalian , circRNAs are highly enriched in neuronal synapses and dynamically upregulated during , suggesting roles in neural and plasticity. Dysregulation of circRNAs is implicated in numerous diseases, particularly cancers, where they promote tumorigenesis through miRNA sponging or protein modulation—such as circFoxo3 enhancing anti-apoptotic effects via p53-MDM2 interactions—and cardiovascular or neurological disorders. Their tissue-specific expression, stability, and detectability in biofluids position circRNAs as promising biomarkers and therapeutic targets, with ongoing research exploring their translational potential in precision medicine.

Discovery and Historical Context

Early Observations of Circular RNAs

The first reports of circular RNAs emerged in 1976, when H. L. Sanger and colleagues identified viroid-like, single-stranded covalently closed circular RNA molecules in infected plant cells, which they characterized as highly base-paired rod-like structures using electron microscopy and enzymatic assays. These findings were initially met with , as the circular forms were dismissed by some as experimental artifacts or contaminants rather than biologically relevant entities in eukaryotic cells. In 1979, M. T. Hsu and M. Coca-Prados provided direct visual evidence of circular RNAs in the cytoplasm of eukaryotic cells, including cells, through electron microscopy, revealing lariat-like structures indicative of circularization and describing them as containing "scrambled exons" resulting from non-canonical splicing. This observation extended the presence of circular RNAs beyond viral contexts to host cellular environments, though they were still viewed as rare anomalies possibly arising from splicing errors in plant mitochondria and other systems. A significant advancement came in 1991, when J. M. Nigro and colleagues reported exon scrambling in transcripts from the tumor suppressor gene in colorectal tumors, identifying abnormally spliced RNAs where s were joined in non-collinear , suggesting a mechanism of back-splicing that produces circular forms. These discoveries highlighted circular RNAs as potential products of in mammalian cells, yet they remained underappreciated due to prevailing biases toward linear detection. Early challenges in recognizing circular RNAs stemmed from detection methods optimized for linear transcripts, such as poly-A selection, which systematically underrepresented non-polyadenylated circular species, leading to their perception as infrequent splicing byproducts rather than distinct entities. Key experiments confirming their circular nature included electron microscopy for visualizing closed-loop structures, as demonstrated in the study, and later RNase R resistance assays, which selectively degrade linear RNAs while leaving circular forms intact, thereby verifying covalent closure.

Genome-Wide Identification and Abundance Revelation

The advent of high-throughput technologies in the early 2010s revolutionized the detection of circular RNAs (circRNAs), shifting from sporadic observations to genome-wide identification and revealing their unexpected abundance across species. In , Salzman et al. employed deep on total with depletion from diverse cell lines, including normal fibroblasts and cells, identifying circular isoforms from hundreds of genes where these circRNAs often predominated over linear counterparts in some genes, estimated at about 10% of transcripts from 1–2% of expressed genes. Their analysis underscored a previously underappreciated splicing outcome in . Building on this, 2013 marked pivotal advancements with independent studies that cataloged thousands of circRNAs using specialized computational tools. Memczak et al. analyzed rRNA-depleted data from , , and samples, developing a to detect back-splice junctions and identifying over 2,700 conserved circRNAs in humans, many exhibiting tissue-specific expression. Concurrently, Jeck et al. introduced the find_circ , which filters for paired-end reads mapping to back-splice sites, applied to RNase R-enriched from human fibroblasts and tissues, revealing approximately 27,000 circRNA candidates that were abundant, evolutionarily conserved, and frequently associated with Alu repeats in flanking introns. These tools enabled systematic annotation, demonstrating circRNAs as a pervasive feature of eukaryotic transcriptomes rather than rare anomalies. By 2014, analyses of large-scale datasets further illuminated circRNA distribution, with the project's rRNA-depleted (Ribozero) from multiple human tissues highlighting their enrichment in neural samples compared to other organs. This was corroborated in 2015 by deep sequencing of tissues, which showed circRNAs comprising up to 10-20% of back-spliced reads in neuronal contexts, far exceeding levels in non-neural tissues. Tissue-specific patterns emerged prominently in neural systems, where circRNAs were upregulated during development and synaptic activity; for instance, the circRNA CDR1as (also known as ciRS-7), derived from the CDR1 locus, exhibited exceptionally high expression in the , particularly in granule cells, acting as a potent sponge. Advancements from 2015 to 2018 refined detection precision and explored pathological contexts. The upgraded CIRCexplorer2 pipeline, introduced in 2016, improved back-splice junction identification by integrating TopHat-Fusion for chimeric read alignment and enabling comprehensive annotation of alternative back-splicing events across diverse datasets. During this period, studies in models revealed dysregulated circRNA profiles; for example, hippocampal RNA-seq from 5xFAD transgenic mice identified 143 differentially expressed circRNAs, including those modulating amyloid-beta pathways, suggesting roles in neurodegeneration. Overall quantification across these efforts indicated that circRNAs can represent up to 20% of detected transcripts in specific conditions like brain tissue, with their closed-loop conferring greater —often with half-lives exceeding 24 hours—compared to linear mRNAs, which degrade within hours. This , validated through actinomycin D chase assays, positions circRNAs as durable regulatory elements in cellular . Subsequent tools like CIRI2 (2015) and updates through the have further improved detection accuracy across species and conditions.

Biogenesis Mechanisms

Back-Splicing and Splicing Variants

Back-splicing represents the core mechanism of circular RNA (circRNA) biogenesis, in which a downstream 5′ donor site is covalently ligated to an upstream 3′ acceptor site on the pre-mRNA transcript, generating a continuous without free ends, 5′ caps, or poly(A) tails. This process contrasts with canonical , where exons are joined in sequential 5′ to 3′ order to form linear mRNAs; back-splicing instead proceeds in reverse orientation using the same spliceosomal machinery, often yielding both a circRNA and a linear byproduct with skipped exons. Although initially viewed as aberrant errors, back-splicing is now recognized as a regulated, non-canonical pathway that produces diverse circles across eukaryotes. The biogenesis of circRNAs via back-splicing involves several key steps, starting with the spatial proximity of splice sites achieved through pre-mRNA secondary structure formation, such as base-pairing between reverse complementary motifs in flanking introns. This brings the donor and acceptor into close contact, enabling assembly and formation of a intermediate, akin to canonical intron excision but resolved to circularize the exons or introns. In the exon-skipping model, first generates a containing the prospective circRNA sequence, followed by heads-to-tails ; alternatively, direct back-splicing can occur without prior skipping. Such events can produce scrambled exon arrangements, where non-sequential s are joined, reflecting variations in splicing patterns rather than random errors. Back-splicing yields multiple circRNA variants based on genomic origin and composition. Exonic circRNAs (ecircRNAs), the most prevalent type accounting for 80–90% of detected circRNAs, are predominantly composed of one or more spliced exons lacking intronic sequences. Intronic circRNAs (ciRNAs) arise from back-spliced introns that resist debranching and degradation, while exonic-intronic hybrids (EIciRNAs) retain both exons and introns in the circle. Intergenic circRNAs form from sequences between genes, and full-intron circRNAs represent cases where entire introns are looped without exonic inclusion. Early experimental validation of back-splicing relied on , which resolved circular RNAs as distinct spots with altered electrophoretic mobility compared to linear isoforms, confirming their topological closure and distinguishing them from linear contaminants. Complementary elements like Alu repeats can briefly enhance splice site juxtaposition during this process.

Regulatory Factors in CircRNA Formation

The formation of circular RNAs (circRNAs) through back-splicing is tightly regulated by cis-acting sequence elements that promote pre-mRNA looping. Complementary repeats, such as Alu elements within flanking introns, facilitate base-pairing across introns, thereby stabilizing the looped structure necessary for the to join upstream exons to downstream ones. In , reverse complementary Alu sequences (rcAlu) are particularly enriched in circRNA-generating loci, driving higher circularization efficiency compared to linear splicing; for instance, in humans, approximately 88% of circRNAs are flanked by such intronic Alu elements. These elements create a competitive dynamic where intra-intronic pairing favors linear RNA production, while inter-intronic pairing slows linear splicing progression and boosts circRNA yield by up to several-fold in reporter assays. Trans-acting splicing factors further modulate circRNA biogenesis by binding these intronic repeats to enhance loop stability. The Quaking (QKI) recognizes QKI-binding motifs in introns and dimerizes to bridge flanking regions, promoting back-splicing and increasing circRNA levels during processes like epithelial-mesenchymal transition; depletion of QKI reduces circRNA output from multiple loci by over 50%. Similarly, Muscleblind (MBL/MBNL1) binds intronic repeats adjacent to circRNA exons, stabilizing loops and inhibiting splicing, as seen in its autoregulation where MBL overexpression elevates its own circMBL isoform. These factors collectively shift the splicing equilibrium toward circularization without altering the core spliceosomal machinery. RNA editing by adenosine deaminases acting on RNA (ADAR) enzymes introduces A-to-I modifications that disrupt or, in some cases, fine-tune base-pairing for circularization. ADAR1 primarily suppresses circRNA formation by editing dsRNA structures formed by Alu repeats, destabilizing loops and reducing back-splicing efficiency; ADAR1 knockdown significantly elevates circRNA abundance genome-wide, with many showing >2-fold increases. However, site-specific editing can enhance circularization if it strengthens complementary pairing elsewhere, as exemplified in circNFIX where ADAR-mediated edits in flanking introns modulate loop formation and circRNA expression in neural contexts. Epigenetic modifications influence circRNA biogenesis by altering accessibility at splice sites. marks such as H3K36me3 and H3K79me2 negatively correlate with circRNA production across types, likely by recruiting factors that favor linear splicing over back-splicing through enhanced definition. at promoter CpG islands represses host gene transcription, indirectly limiting pre-mRNA availability for circularization, while hypomethylation promotes open conducive to events yielding circRNAs. More recently, additional RNA-binding proteins such as FUS and NOVA2 have been shown to promote back-splicing, while the DExH-box DHX9 suppresses it by unwinding base-pairing. These layers integrate with sequence and protein regulators to fine-tune circRNA output in a context-dependent manner.

Structural Characteristics

Classification by Origin and Type

Circular RNAs (circRNAs) are classified primarily by their genomic origin, which determines their composition and potential regulatory roles. Exonic circRNAs (ecircRNAs) are derived exclusively from one or more exons of protein-coding genes and represent the most abundant class, comprising over 80% of detected circRNAs in eukaryotic transcriptomes. Intronic circRNAs (ciRNAs) originate from introns that evade typical splicing and pathways, while exonic-intronic circRNAs (EIciRNAs) incorporate both exons and retained introns, often preserving regulatory elements like intronic repeats. Intergenic circRNAs arise from non-coding intergenic regions or read-through transcripts spanning adjacent genes, forming a less common but diverse category. Another class, tRNA intronic circRNAs (tricRNAs), are generated from the introns of precursor tRNAs (pre-tRNAs) during tRNA splicing. Structurally, circRNAs vary in their exon composition and processing state. Single-exon circRNAs consist of a solitary looped via back-splicing, often from unusually long exons, whereas multi-exon circRNAs, which are more prevalent, include two or more exons joined covalently, typically ranging from 2 to 15 exons per circle. Lariat circRNAs retain the branched 2',5'- from splicing intermediates, distinguishing them from fully processed circRNAs, which form linear-like circles after debranching and exhibit higher stability. For instance, ciRNAs like ci-ankrd52 exemplify lariat-derived structures, predominantly localized in the . Biogenesis of circRNAs involves alternative splicing events such as back-splicing, where a downstream splice donor joins an upstream splice acceptor. This can occur via direct back-splicing facilitated by complementary sequences in flanking (e.g., Alu repeats), often yielding ecircRNAs in the . Other pathways include lariat debranching, where exon-skipped lariat intermediates resist degradation by the DBR1 , leading to stable circular introns or exon-inclusive forms like ciRNAs and EIciRNAs. Intron excision circularization, driven by specific motifs (e.g., GU-rich and C-rich elements), allows intronic lariats to circularize without exonic involvement, typically producing nuclear ciRNAs. EcircRNAs dominate cytoplasmic abundance, while ciRNAs and EIciRNAs are primarily nuclear, reflecting their biogenesis constraints. Standardized nomenclature facilitates circRNA identification and comparison across studies. The circBase database assigns unique identifiers prefixed by species (e.g., "hsa" for Homo sapiens), followed by "circ_" and a numerical ID, such as hsa_circ_000001, linking to genomic coordinates and supporting datasets from high-throughput sequencing. This system unifies annotations from diverse origins, enabling precise referencing in research.

Size, Stability, and Cellular Localization

Circular RNAs (circRNAs) exhibit a characteristic size profile that distinguishes them from linear RNAs. Exonic circRNAs (ecircRNAs), derived primarily from back-spliced exons, typically range from 200 to 500 () in length, though some can extend up to 4 (). In contrast, circular intronic RNAs (ciRNAs) vary in size, often spanning hundreds to over 1000 depending on intron lengths. Unlike most linear messenger RNAs (mRNAs), circRNAs lack a poly(A) tail, which contributes to their resistance to poly(A)-dependent degradation pathways. The closed-loop structure of circRNAs confers exceptional stability compared to linear RNAs. This circular conformation protects them from degradation, as demonstrated by their enrichment following treatment with RNase R, an enzyme that preferentially digests linear RNAs; circRNAs are typically enriched 10- to 100-fold relative to linear counterparts under these conditions. The of ecircRNAs often exceeds 48 hours, in stark contrast to the average 10-hour of mRNAs. analyses further confirm this durability, showing circRNAs migrating more slowly than linear RNAs of equivalent length due to their covalently closed structure, which resists denaturation and enzymatic processing. These properties enable circRNAs to accumulate to higher levels in cells, facilitating sustained regulatory functions over extended periods. Subcellular localization of circRNAs varies by type and influences their functional roles. EcircRNAs predominantly reside in the cytoplasm, often associating with ribonucleoprotein (RNP) granules such as stress granules or processing bodies, where they can interact with microRNAs or proteins. Conversely, ciRNAs and exon-intron circRNAs (EIciRNAs) are primarily nuclear, where they associate with RNA polymerase II (Pol II) machinery, potentially modulating transcription. CircRNAs also display tissue-specific enrichment, with notable abundance in the brain and heart, reflecting their roles in neural and cardiac processes.

Molecular Functions

MicroRNA Sponging and Competing Endogenous RNA Activity

Circular RNAs (circRNAs) function prominently as (miRNA) sponges within the competing endogenous RNA (ceRNA) framework, where they harbor multiple miRNA response elements (MREs) to sequester miRNAs, thereby preventing their binding to target mRNAs and allowing derepression of those transcripts. This sponging activity is facilitated by the circRNAs' covalently closed , which confers resistance to exonuclease-mediated degradation and enables the stable accumulation of dense MRE clusters without the typical 5' cap or 3' poly-A tail vulnerabilities of linear . The paradigm was established with CDR1as (also termed ciRS-7), a brain-enriched circRNA derived from the CDR1 locus, which contains more than 70 conserved binding sites for miR-7, one of the most abundant miRNAs in neural tissues. By competitively binding miR-7, CDR1as inhibits its repressive effects on targets such as those involved in neuronal signaling, with the interaction validated through (AGO) sequencing (CLIP-seq) showing dense AGO-miR-7 occupancy on CDR1as. In vivo functional evidence comes from embryos, where injection of synthetic CDR1as RNA mimics miR-7 knockdown phenotypes, resulting in sensorineural defects like impaired development due to miR-7 dysregulation and consequent target derepression. Additional examples illustrate the versatility of this mechanism across tissues and contexts. CircSRY, originating from the sex-determining region Y gene, acts as a sponge for miR-138 through multiple binding sites, reducing miR-138-mediated repression in reporter assays and altering pathways. Similarly, circHIPK3, derived from the HIPK3 gene, sponges a repertoire of miRNAs—including miR-124, miR-152, and miR-29a—in proliferating cells, with CLIP-seq confirming direct interactions and assays demonstrating miRNA-dependent modulation of target expression in cancer models. The circular architecture uniquely supports this ceRNA role by permitting high-affinity, multivalent miRNA interactions without RNA turnover, as evidenced by pull-down experiments where circRNAs retain bound miRNAs longer than linear counterparts. Such sponging can lead to significant upregulation of target mRNA levels upon circRNA overexpression or miRNA , underscoring the regulatory scale in physiological networks.

Transcriptional and Post-Transcriptional Regulation

Circular RNAs (circRNAs) play significant roles in transcriptional regulation primarily through nuclear-localized forms such as circular intronic RNAs (ciRNAs) and exon-intron circRNAs (EIciRNAs). CiRNAs, derived solely from introns, associate with RNA polymerase II (Pol II) elongation machinery and promote cis-regulatory enhancement of parental gene transcription. For instance, ci-ankrd52, originating from the second intron of the ANKRD52 gene, accumulates at its transcription site and boosts ANKRD52 expression by interacting with Pol II, as demonstrated by RNA immunoprecipitation (RIP) experiments showing direct Pol II binding. Similarly, EIciRNAs, which retain unspliced introns, interact with U1 small nuclear ribonucleoprotein (snRNP) to form a complex that recruits Pol II to parental gene promoters, thereby enhancing transcription initiation and elongation. Depletion of specific EIciRNAs via RNA interference results in 20-50% reductions in parental gene expression levels, underscoring their regulatory impact. Beyond transcription, circRNAs influence post-transcriptional processes such as by serving as scaffolds for splicing factors. The muscleblind (MBL) protein binds to intronic sequences flanking its own pre-mRNA, promoting back-splicing to generate circMBL, which in turn sequesters excess MBL and fine-tunes its availability for linear splicing events. This autoregulatory loop maintains balanced MBL protein levels essential for neuronal and muscle development. N6-methyladenosine (m6A) modifications on circRNAs further modulate their post-transcriptional fate, affecting and subcellular localization. The m6A reader protein YTHDC1 recognizes these modifications and facilitates the nuclear export of m6A-modified circRNAs, enabling their cytoplasmic functions while preventing nuclear retention. This process influences circRNA , with m6A promoting via endonuclease recruitment in some contexts or enhancing through reader interactions. CircRNAs also regulate post-transcriptional events by sponging RNA-binding proteins (RBPs), distinct from their miRNA sponging activity. For example, circPABPN1 binds and sequesters the RBP HuR (also known as ELAVL1), preventing HuR from stabilizing and translating PABPN1 mRNA, thereby repressing polyadenylate-binding protein nuclear 1 (PABPN1) expression. Databases like CIRCpedia catalog numerous such circRNA-RBP interactions, revealing widespread RBP sequestration as a mechanism for fine-tuning across types. Certain circRNAs exhibit protein-coding potential through cap-independent mechanisms, such as internal ribosome entry sites (IRES) or m6A-modified cap-independent (IRES- or m6A-driven). For instance, circFBXW7, derived from the , translates a 21-kDa protein (FBXW7-185aa) that inhibits cell by enhancing ubiquitination of c-Myc. This function has been validated in cancer models, highlighting circRNAs' role in producing functional peptides that modulate cellular processes. As of 2024, advances in and have identified hundreds of translatable circRNAs across , expanding their regulatory repertoire beyond non-coding roles.

Biological Roles

Involvement in Immune Responses

Circular RNAs (circRNAs) play significant roles in modulating innate immune responses, particularly in antiviral defense mechanisms. In type 1 (HSV-1) infections, interferon-induced circRNAs, such as circRELL1, escape viral host shutoff and restrict viral replication by enhancing innate antiviral signaling. These circRNAs activate the (RLR) pathway, where RIG-I senses foreign circRNAs lacking N6-methyladenosine modifications, triggering type I production and downstream antiviral . This activation promotes innate immunity without excessive , as demonstrated in studies showing that engineered circRNAs potently stimulate RIG-I-dependent responses to inhibit viral spread. Additionally, circRNAs can act as miRNA sponges to derepress immune genes; for instance, by sequestering miRNAs that suppress innate immunity activators, they upregulate interferon-stimulated genes during viral challenges. In cardiac injury, circCdr1as promotes anti-inflammatory M2 macrophage polarization via miR-7 and miR-671, as shown in 2025 studies. In inflammatory processes, circRNAs regulate polarization and production through key signaling pathways. The circRNA circPPM1F promotes activation by sponging miR-134, thereby suppressing PPM1F translation and facilitating pathway activation, which enhances pro-inflammatory release such as TNF-α and IL-6. Conversely, circCdr1as (also known as ciRS-7) modulates phenotypes toward an anti-inflammatory M2 state by interacting with miR-7 and miR-671, reducing excessive in contexts like cardiac injury. Knockdown of circCdr1as in leads to increased pro-inflammatory production, highlighting its regulatory role in balancing inflammatory responses. These mechanisms underscore circRNAs' involvement in fine-tuning signaling to prevent chronic . CircRNAs also contribute to adaptive immunity, particularly in T and B cell functions. In B cells, circRNAs are enriched and influence activation and differentiation; for example, RNA-seq analyses of hematopoietic cells have identified over 100 immune-specific circRNAs differentially expressed in lymphoid lineages, supporting production and immune memory. A notable example is circPTPN22, derived from the PTPN22 locus (a risk factor for and other autoimmune diseases), which sponges miR-4689 to upregulate S1PR1 expression, thereby modulating T-cell activation and proliferation. This circRNA's overexpression enhances signaling, promoting adaptive responses but also contributing to autoimmune susceptibility when dysregulated. Studies from 2018 to 2023 using on immune cells have revealed hundreds of such circRNAs, with knockout models demonstrating reduced responses (e.g., IL-2 and IFN-γ) upon circRNA depletion, confirming their essential role in T-cell mediated immunity. Overall, through miRNA sponging of immune repressors and RIG-I enhancement, circRNAs integrate innate and adaptive arms of immunity for robust host defense.

Regulation of Development and Differentiation

Circular RNAs (circRNAs) play pivotal roles in embryonic , where their expression often peaks during early stages to orchestrate key processes such as tissue specification and . In pre-implantation embryos, circRNA abundance dynamically varies from 28,774 to 190,008 copies per embryo across developmental stages, highlighting their temporal regulation during early embryogenesis. Similarly, in porcine models, circRNAs exhibit peak expression at embryonic day 60 (E60) in the , coinciding with rapid neural , while in embryos, circRNA profiles reveal stage-specific peaks distributed across embryogenesis, suggesting involvement in sequential patterning events like somitogenesis. For instance, comprehensive profiling in identifies thousands of circRNAs with differential expression at various embryonic time points, implicating them in mesodermal segmentation and axial . In cell , circRNAs contribute to lineage commitment and tissue specification, particularly in maintaining pluripotency and guiding trajectories. Single-cell RNA sequencing analyses across and tissues demonstrate that circRNAs display cell-type and developmental stage-specific expression patterns, with higher abundance in cells transitioning to differentiated states. In hematopoiesis, circRNAs such as those derived from antisense transcripts regulate maintenance by modulating interactions and competing endogenous networks, ensuring balanced self-renewal and . Depletion studies further underscore their necessity, as /Cas9-mediated knockdown of specific circRNAs, like circQKI in myogenic lineages, impairs efficiency, leading to prolonged states. During , circZNF609 exemplifies circRNA function by promoting myoblast essential for muscle . Expressed in both murine and myoblasts, circZNF609 knockdown via siRNA reduces rates without affecting the linear ZNF609 mRNA, indicating a circRNA-specific . This occurs through cap-independent of circZNF609 into a protein that drives G1-S phase progression, as evidenced by delayed entry in knockdown models. Additionally, circZNF609 associates with IGF2BP1 to enhance its own stability, amplifying proliferative signals during early myogenic commitment. In , circRNAs fine-tune neuronal and circuit formation, with prominent examples including circHOMER1 and CDR1as. CircHOMER1 regulates synaptic development and experience-dependent plasticity in the mouse visual cortex, where its depletion disrupts dendritic morphogenesis and excitatory synapse formation, as shown in 2025 studies. Single-cell reveals circHOMER1's enrichment in excitatory neurons, supporting its role in branching and connectivity during cortical specification. Likewise, CDR1as, highly expressed in tissues, modulates by sponging miR-7, and its knockout in mice impairs excitatory transmission, delaying neuronal maturation and integration. Adipogenesis involves circRNAs that interact with epigenetic regulators to promote preadipocyte into mature adipocytes. CircFIRRE, derived from the FIRRE locus, emerges as a highly abundant circRNA during human embryonic stem cell toward adipogenic lineages, correlating with enhanced lipid accumulation and metabolic reprogramming. Relatedly, circSAMD4A acts as a miR-138-5p sponge to upregulate , a that represses anti-adipogenic genes, thereby accelerating in obesity-associated models. Experimental evidence from CRISPR-based knockdowns confirms these roles; for example, targeting circZNF609 in myoblasts or CDR1as in delays by 2-3 days, as measured by marker gene expression and morphological assays. Complementary single-cell datasets across developmental stages further validate stage-specific circRNA upregulation, such as in dopaminergic progenitors, where circRNAs like those regulating size and peak during specification.

Implications in Disease

Neurological and Neurodegenerative Disorders

Circular RNAs (circRNAs) have emerged as key regulators in the pathogenesis of Alzheimer's disease (AD), with dysregulation observed in post-mortem brain tissues and biofluids. Genome-wide RNA sequencing studies have identified 147 differentially expressed circRNAs in AD brains across regions like the superior temporal lobe and parahippocampal gyrus, with fold changes ≥1.15-fold compared to healthy controls. For instance, circHDAC9 expression is significantly decreased in the sera and brains of AD patients and mouse models, leading to reduced sponging of miR-138. This downregulation allows miR-138 to inhibit SIRT1, thereby promoting amyloid-β (Aβ) accumulation and mitochondrial dysfunction; overexpression of circHDAC9 in vitro suppresses excessive Aβ production by enhancing SIRT1 activity. In APP/PS1 mouse models of AD, knockdown of circCwc27—a circRNA upregulated in AD—reduces Aβ plaque load by approximately 40% in the hippocampus and improves spatial learning and memory performance. In (), circRNAs contribute to α-synuclein pathology through sponging mechanisms. circSNCA, derived from the SNCA , is upregulated in PD models and acts as a sponge for miR-7, which normally binds the 3' of SNCA mRNA to suppress its . This stabilizes SNCA mRNA, leading to increased α-synuclein protein levels and enhanced neuronal ; treatment with downregulates circSNCA, thereby restoring miR-7 activity, reducing cell death, and promoting in PD cell models. Post-mortem analyses of PD brains confirm elevated circSNCA expression, correlating with disease severity. Beyond major neurodegenerative disorders, circRNAs influence other neurological conditions, including epilepsy and schizophrenia. Transcriptomic studies indicate circRNA dysregulation in these disorders, potentially affecting neuronal excitability and synaptic plasticity. circRNAs also play roles in neuronal repair mechanisms, such as axon regeneration following injury. Neuronal circRNAs are enriched and stable in exosomes, facilitating intercellular communication in the central nervous system; for example, circ-Spidr promotes axon regrowth in dorsal root ganglion neurons after peripheral nerve injury by modulating regenerative pathways. In AD contexts, circRNAs from brain tissue exhibit disease-specific profiles, highlighting their potential as biomarkers for monitoring neurodegeneration. These findings underscore the therapeutic promise of targeting circRNAs to mitigate synaptic loss and protein aggregation in neurological disorders.

Cardiovascular, Metabolic, and Oncological Conditions

Circular RNAs (circRNAs) play significant roles in cardiovascular diseases, particularly in and (MI). Studies have identified differentially expressed circRNAs in MI models, suggesting their involvement in , , and following ischemia. For instance, circFndc3b has been shown to modulate cardiac repair post-MI by regulating endothelial and . In renal and metabolic disorders, circRNAs contribute to and dysregulation. circACTR2 promotes renal by sponging miR-561 to activate the in cells. In , a complication of , circRNAs serve as potential diagnostic markers, with expression changes correlating to disease severity and renal function decline. Oncological conditions feature prominent circRNA dysregulation across multiple cancers. In hepatocellular carcinoma (HCC), circRNAs drive tumor progression through competing endogenous RNA activity. Similarly, in , circHIPK3 enhances and invasion by sponging miR-7. Pan-cancer analyses from the , integrating data from sources like (TCGA), have cataloged over 1,000 oncogenic circRNAs, revealing conserved networks where circRNAs sponge tumor-suppressive miRNAs to promote and across tumor types. CircRNAs hold diagnostic promise as circulating biomarkers in these conditions. In MI, plasma levels of specific circRNAs, such as circ_0049271, achieve high discriminatory accuracy with an area under the curve () greater than 0.85 for early detection, outperforming some traditional markers. Therapeutic targeting of circRNAs also shows efficacy; for example, siRNA-mediated knockdown of oncogenic circRNAs like circRNA-SORE in sorafenib-resistant HCC xenografts restores sensitivity and reduces tumor growth, demonstrating potential for precision . TCGA-integrated studies further validate these networks, linking circRNA expression to clinical outcomes in metabolic and oncological diseases.

Evolutionary Perspectives

Conservation Across Species

Circular RNAs (circRNAs) exhibit varying degrees of conservation across species, reflecting the evolutionary persistence of their biogenesis mechanisms and functional elements. The back-splicing process that generates most eukaryotic circRNAs relies on the , a conserved splicing machinery universal among eukaryotes, which enables the covalent joining of splice donor and acceptor sites in reverse order. In mammals, this is often facilitated by Alu repeats providing intron complementarity, while analogous repetitive elements and base-pairing motifs support back-splicing in non-mammalian species such as , where circRNAs are highly enriched and conserved across fly species. This shared dependence on canonical splicing components underscores the ancient origins of circRNA formation, predating major eukaryotic divergences. At the sequence level, certain circRNAs display strong , particularly in vertebrates. For instance, the circRNA CDR1as (also known as ciRS-7), a well-studied miRNA , is highly conserved across mammals, with its core sequence and multiple miR-7 binding sites preserved in , , and other vertebrates, though the back-splice junctions themselves show less conservation than the miRNA target sites. Comparative analyses reveal that while global circRNA repertoires evolve rapidly, a subset of circRNAs—often those with regulatory functions—exhibits appreciable sequence between closely related like and , with orthologous circRNAs comprising approximately 10-20% of identified repertoires in shared tissues. Back-spliced circRNAs are abundant across eukaryotic kingdoms beyond , including and fungi, but are absent in prokaryotes due to the lack of spliceosomes; however, other circular RNAs, such as circular 23S rRNAs, are abundant in archaeal ribosomes across diverse lineages, suggesting ancient circular RNA structures in prokaryotes. In , thousands of circRNAs have been identified in species like , often derived from exonic regions involved in responses and , demonstrating back-splicing via intron pairing without reliance on mammalian-specific repeats. Similarly, in fungi such as Magnaporthe oryzae, over 8,000 circRNAs arise from back-splicing during mycelial growth, indicating conserved biogenesis signals in this kingdom. Evolutionary studies highlight the persistence of intron pair complementarity as a key biogenesis signal, with motifs enabling circularization conserved over hundreds of millions of years in eukaryotic lineages, and circRNA exons showing elevated sequence conservation compared to non-circRNA exons from the same genes, suggestive of functional constraints. Recent from 2016 to 2023, integrating datasets across vertebrates, further confirm that 10-20% of circRNAs are orthologous between and , with higher rates in and heart tissues, emphasizing species-specific yet functionally preserved roles.

Role in RNA World Hypothesis and Viroids

Circular RNAs (circRNAs) have been proposed as potential relics from the precellular , where RNA molecules served dual roles as genetic material and catalysts prior to the emergence of DNA and proteins. Their covalently closed structure confers exceptional stability against exonucleases, a property that would have been advantageous in primitive, error-prone self-replicating systems, enabling the formation of self-sustaining RNA networks capable of Darwinian . This stability, combined with the absence of free ends, mirrors the requirements for efficient replication in an RNA-dominated era, positioning circRNAs as plausible survivors of early life forms. Viroids, the smallest known infectious agents, exemplify non-coding circular RNAs and reinforce these connections to ancient RNA-based systems. These plant pathogens consist of single-stranded, circular RNAs typically 250-400 nucleotides in length that lack protein-coding capacity and replicate using host RNA polymerases. The (PSTVd), the first identified , was discovered in 1971 by Theodor O. Diener through studies on , revealing its unconventional nature as a naked RNA replicator. Parallels between viroids and cellular circRNAs highlight shared mechanisms for , particularly in circularization and durability. Viroids in the family Avsunviroidae employ hammerhead ribozymes—self-cleaving motifs—for processing linear precursors into mature circular forms during replication, a process that enhances their resistance to degradation. Cellular circRNAs, while formed via backsplicing rather than ribozymes, exhibit analogous resistance due to their closed loops, suggesting they may mimic viroid-like strategies for long-term in modern cells. Some hypotheses suggest that viroid-like circular RNAs may represent relics from the , potentially protecting early RNA genomes from degradation. Recent 2020s studies on synthetic circRNAs have explored their properties through assembly and testing, demonstrating enhanced durability and protein expression that echo prebiotic RNA dynamics. These experiments support the idea that circular forms could have facilitated genome protection and replication in early progenitors. Supporting evidence includes the in vitro demonstration of viroid replication via a rolling-circle , where circular templates generate multimeric intermediates that are cleaved and ligated to produce new circles, a process reliant on host enzymes but initiated by self-processing. This underscores the viroid's durability, akin to circRNA resistance to RNases, which allows both to evade typical RNA turnover pathways and persist in diverse conditions.

References

  1. [1]
    Molecular mechanisms of circular RNA translation - Nature
    Jun 14, 2024 · Circular RNAs (circRNAs) are covalently closed single-stranded RNAs without a 5′ cap structure and a 3′ poly(A) tail typically present in ...
  2. [2]
  3. [3]
    Circular RNA: metabolism, functions and interactions with proteins
    Dec 14, 2020 · Circular RNAs (CircRNAs) are single-stranded, covalently closed RNA molecules that are ubiquitous across species ranging from viruses to mammals.
  4. [4]
    Viroids are single-stranded covalently closed circular RNA ...
    Viroids are uncoated infectious RNA molecules pathogenic to certain higher plants. Four different highly purified viroids were studied.Missing: eukaryotic cells
  5. [5]
    Electron microscopic evidence for the circular form of RNA ... - PubMed
    Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature. 1979 Jul 26;280(5720):339-40. ... Authors. M T Hsu, M ...
  6. [6]
    Early history of circular RNAs, children of splicing - PMC - NIH
    In this commentary we briefly summarize early work on circular RNAs derived from spliceosome mediated circularization.
  7. [7]
    Scrambled exons - PubMed - NIH
    Feb 8, 1991 · We identified several abnormally spliced transcripts in which exons from a candidate tumor suppressor gene (DCC) were scrambled during the splicing process in ...
  8. [8]
    Full article: Early history of circular RNAs, children of splicing
    In this commentary we briefly summarize early work on circular RNAs derived from spliceosome mediated circularization. We highlight how this early work inspired ...
  9. [9]
    Characterization of RNase R-digested cellular RNA source that ...
    This result shows that the circular RNAs, even in extremely low amounts (16), are resistant to RNase R digestion, while the enormously abundant linear RNAs are ...
  10. [10]
    Circular RNAs Are the Predominant Transcript Isoform from ...
    Our results suggest that a non-canonical mode of RNA splicing, resulting in a circular RNA isoform, is a general feature of the gene expression program in human ...
  11. [11]
    Circular RNAs are a large class of animal RNAs with ... - Nature
    Feb 27, 2013 · Circular RNAs (circRNAs) in animals are an enigmatic class of RNA with unknown function. To explore circRNAs systematically, we sequenced and computationally ...
  12. [12]
    Loss of a mammalian circular RNA locus causes miRNA ... - Science
    Taken together, these data show that loss of Cdr1as does not affect miR-7 and miR-671 in tissues outside the brain that normally exhibit very low levels of ...
  13. [13]
    Identifying circRNA-associated-ceRNA networks in the ... - Aging-US
    A total of 555 circRNAs, 183 miRNAs and 319 mRNAs were identified to be significantly dysregulated (fold-change ≥ 2.0 and p-value < 0.05) in the hippocampus of ...<|separator|>
  14. [14]
  15. [15]
    Regulation of circRNA biogenesis - PMC - NIH
    Top, RNA pairing formed across flanking introns promotes back-splicing, leading to the formation of a circRNA and a linear RNA with exon exclusion. Bottom, the ...
  16. [16]
    Circular RNA biogenesis can proceed through an exon-containing ...
    Jun 9, 2015 · The basic mechanism responsible for the biogenesis of circular RNA involves a 'backsplicing' reaction in which a branch point upstream of a ...Missing: seminal | Show results with:seminal
  17. [17]
  18. [18]
  19. [19]
  20. [20]
  21. [21]
  22. [22]
    Deep Insights in Circular RNAs: from biogenesis to therapeutics
    May 15, 2020 · So far, three types of circRNAs have been identified: exonic circRNAs (ecircRNAs), intronic RNAs (ciRNAs) [22] and exon-intron circRNAs ( ...
  23. [23]
    Mechanisms and therapeutic implications of gene expression ...
    Jan 17, 2025 · Based on their origin, circRNAs can be categorized into four distinct types: exon circRNAs (EcircRNAs), composed solely of exons; intronic ...
  24. [24]
    Circular RNAs: history, metabolism, mechanisms of function, and ...
    Jan 9, 2025 · EcircRNAs, consisting of one or more exons, make up the majority (over 80%) of circRNAs detected. Exons of single-exon circRNAs are much longer ...
  25. [25]
    Circular RNAs: diversity of form and function
    ### Summary of Structural Types of circRNAs
  26. [26]
  27. [27]
    circBase
    Here you can explore public circRNA datasets and download the custom python scripts needed to discover circRNAs in your own (ribominus) RNA-seq data.Downloads · List search · Help · Web BLATMissing: nomenclature hsa_circ_000001
  28. [28]
    circBase: a database for circular RNAs - PMC - NIH
    We developed a database and website, “circBase,” where merged and unified data sets of circRNAs and the evidence supporting their expression can be accessed.Missing: hsa_circ_000001 | Show results with:hsa_circ_000001
  29. [29]
    Circular RNAs: diversity of form and function - PMC - NIH
    They can comprise a single exon or multiple exons. The size of the spliced circle molecule can range from under 100 nt to over 4 kb. A gene that gives rise ...
  30. [30]
    CircRNA: functions and properties of a novel potential biomarker for ...
    May 23, 2017 · Circular RNAs may function as potential molecular markers for disease diagnosis and treatment and play an important role in the initiation and progression of ...
  31. [31]
    Circular RNAs: Expression, localization, and therapeutic potentials
    Cytoplasm localization of proteins can be facilitated by circRNAs. circFoxo3, for instance, a circRNA that is highly expressed in the heart tissues of aged ...
  32. [32]
    landscape of circular RNA expression in the human heart
    The most abundant cardiac-expressed circRNA is a cytoplasmic localized single-exon circSLC8A1-1. The longest human transcript TTN alone generates up to 415 ...
  33. [33]
    Natural RNA circles function as efficient microRNA sponges - Nature
    Feb 27, 2013 · Natural RNA circles function as efficient microRNA sponges. Nature 495, 384–388 (2013). https://doi.org/10.1038/nature11993. Download ...
  34. [34]
    Circular RNA profiling reveals an abundant circHIPK3 that regulates ...
    Apr 6, 2016 · Circular RNAs (circRNAs) represent a class of widespread and diverse endogenous RNAs that may regulate gene expression in eukaryotes.
  35. [35]
    YTHDC1 mediates nuclear export of N6-methyladenosine ... - eLife
    Oct 6, 2017 · Here, we show that the m 6 A-binding protein YTHDC1 mediates export of methylated mRNA from the nucleus to the cytoplasm in HeLa cells.Missing: circular | Show results with:circular
  36. [36]
    Identification of HuR target circular RNAs uncovers suppression of ...
    Jan 12, 2017 · We propose that the extensive binding of CircPABPN1 to HuR prevents HuR binding to PABPN1 mRNA and lowers PABPN1 translation.
  37. [37]
    Linking the association between circRNAs and Alzheimer's disease ...
    Jul 3, 2020 · In conclusion, our findings indicate that circRNAs in parahippocampal gyrus are possible biomarkers and regulators of AD as well as potential ...
  38. [38]
    Exploring the regulatory roles of circular RNAs in Alzheimer's disease
    Sep 21, 2020 · When present at an excessive concentration, MBL binds to the mbl pre-mRNA and causes its back-splicing to form circMbl. However, when the MBL ...
  39. [39]
    Circular RNA Cwc27 contributes to Alzheimer's disease ...
    Sep 9, 2021 · Circular RNA Cwc27 contributes to Alzheimer's disease pathogenesis by repressing Pur-α activity ... 2017;357:1254. doi: 10.1126/science ...Rna Pull Down And Mass... · Results · Pur-α Reduces Aβ Load And...
  40. [40]
    CircSNCA downregulation by pramipexole treatment mediates cell ...
    The downregulation of circSNCA by PPX treatment reduced cell apoptosis and promoted cell autophagy in PD via a mechanism that served as a miR-7 sponge to ...
  41. [41]
    Insights into the multifaceted role of circular RNAs - Nature
    Jan 10, 2022 · CDR1as is highly expressed in excitatory neurons, and mice lacking CDR1as exhibit neuropsychiatric disorders with dysfunctional synaptic ...Mirna And Protein Decoys · Circrnas In Pd Pathogenetic... · Snca And Mir-7<|separator|>
  42. [42]
    Identification and characterization of circular RNA in the model of ...
    Based on the results in Figure 4, circ-Mbd5, circ-Birc6, circ-Ttc3, circ-613766, circ-632694, and circ-623036 were significantly downregulated, while circ-Banp, ...
  43. [43]
    Circular RNA in Schizophrenia and Depression - Frontiers
    Current research indicates that non-coding RNA (ncRNA) may play a role in the occurrence and development of SZ and DEP.Missing: DLGAP1 | Show results with:DLGAP1
  44. [44]
    Circ-Spidr enhances axon regeneration after peripheral nerve injury
    Oct 17, 2019 · Through in vitro and in vivo experiments, we determined that down-regulation of circ-Spidr could suppress axon regeneration of DRG neurons after ...Missing: circNFIX | Show results with:circNFIX
  45. [45]
    Identification of circRNAs linked to Alzheimer's disease and related ...
    Feb 16, 2023 · We identified 48 circRNAs that were significantly associated with AD. We observed that circRNA expression differed by dementia subtype.
  46. [46]
    Circular RNA in cardiovascular disease - PubMed
    In this paper, we summarize current understanding of circRNA biogenesis, properties, expression profiles, detection methods, functions, and their implication ...
  47. [47]
    The emerging landscape of circular RNA in cardiovascular diseases
    Here, we review the biogenesis, properties and biological function of circRNAs, and summarize their roles in cardiovascular disorders.
  48. [48]
    Circular RNA CircFndc3b modulates cardiac repair after myocardial ...
    Sep 20, 2019 · Here we report identification of circRNA transcripts that are differentially expressed in post myocardial infarction (MI) mouse hearts ...
  49. [49]
    Circular RNAs in Diabetic Nephropathy: Updates and Perspectives
    CircRNAs are key in DN and therefore important for understanding DN pathophysiology and developing new therapeutic strategies.
  50. [50]
    The role of circular RNA in Diabetic Nephropathy
    May 20, 2022 · Studies have shown that CircRNA plays an important role in diabetic inflammation and renal tubular fibrosis, and the degree of inflammation is ...
  51. [51]
    Circular RNAs in metabolism and metabolic disorders - Zeng - 2021
    Feb 12, 2021 · Dysregulation of circRNAs can lead to metabolic disorders, indicating that circRNAs represent plausible potential targets to alleviate metabolic ...Missing: renal | Show results with:renal
  52. [52]
    Comprehensive analysis of the competing endogenous circRNA ...
    May 28, 2021 · A prognostic circRNA-lncRNA-miRNA-mRNA ceRNA network for HCC was constructed for the first time, and a seven-gene signature was identified and ...
  53. [53]
    Circular RNAs in cancer: roles, mechanisms, and therapeutic ... - NIH
    Jan 4, 2025 · This review delves into the role of circRNAs in four key cancer types: colorectal cancer (CRC), gastric cancer (GC), liver cancer (HCC), and lung cancer (LUAD).
  54. [54]
    Pan-cancer proteogenomic analysis reveals long and circular ...
    Aug 11, 2020 · Recent studies show that annotated long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) encode for stable, functional peptides that contribute to human ...
  55. [55]
    Circ_0049271: A Novel Circular RNA Biomarker for Diagnosis and ...
    Aug 20, 2025 · ... (AUC) = 0.9099, 95 ... Circ_0049271: A Novel Circular RNA Biomarker for Diagnosis and Prognostic Assessment in Acute Myocardial Infarction.
  56. [56]
    Going circular: history, present, and future of circRNAs in cancer
    Aug 16, 2023 · We review the current knowledge of circRNA biogenesis and function, and of their involvement in tumorigenesis and potential utility in cancer-targeted therapy.Circrnas As Regulators Of... · Circrnas As Powerful... · Circrnas As Predictive...
  57. [57]
    Phylogenetic comparison and splice site conservation of eukaryotic ...
    Jun 17, 2021 · Although splicing machinery is thought to be preserved among eukaryotes, distinct mechanisms have been observed in humans, yeast and Arabidopsis ...
  58. [58]
    The circular RNA Cdr1as, via miR-7 and its targets, regulates insulin ...
    Jul 27, 2015 · The Cdr1as molecule was recently demonstrated to reduce miR-7 activity in embryonic zebrafish brain. We hypothesized that Cdr1as may also affect ...Missing: dysregulation | Show results with:dysregulation
  59. [59]
    Insights into circular RNA biology - PMC - PubMed Central
    Circular RNAs (circRNAs) are non-coding RNA with a covalently closed-loop structure, formed by backsplicing, and are involved in modulating gene expression.Missing: definition | Show results with:definition
  60. [60]
    Characterization and Function of Circular RNAs in Plants - Frontiers
    May 18, 2020 · Emerging evidence has shown that circRNAs are involved in the regulation of gene expression at transcriptional and post-transcriptional levels.Missing: ELF18 receptor
  61. [61]
    Genome-wide Identification and characterization of circular RNAs in ...
    Apr 30, 2018 · Here, we identified a total of 8,848 circRNAs from the model plant pathogenic fungus M. oryzae. 5,840 circRNAs were identified from mycelium, ...Missing: prokaryotes | Show results with:prokaryotes
  62. [62]
    Circular RNA repertoires are associated with evolutionarily young ...
    Sep 20, 2021 · First described more than 40 years ago, circular RNAs (circRNAs) were originally perceived as a curiosity of gene expression, yet they have ...<|separator|>
  63. [63]
    CircAtlas: an integrated resource of one million highly accurate ...
    Apr 28, 2020 · CIRCpedia v2: an updated database for comprehensive circular RNA annotation and expression comparison. Genomics Proteomics Bioinformatics ...
  64. [64]
    Circular RNAs: relics of precellular evolution? - PNAS
    Recent models of such preceflular RNA systems are patterned after the properties of intron-derived ribozymes. On the basis of a phylogenetic analysis and known.
  65. [65]
    Circular at the very beginning: on the initial genomes in the RNA world
    Jul 17, 2024 · Here, we explore the scenario of a circular RNA genome plus linear ribozyme(s) at the precellular stage of the RNA world through computer modelling.
  66. [66]
    Potato spindle tuber viroid: the simplicity paradox resolved? - 2007
    Jul 20, 2007 · The 1971 discovery of potato spindle tuber viroid (PSTVd) by T. O. Diener was a landmark event in modern biology. Viroids are the smallest ...
  67. [67]
    Viroid Replication: Rolling-Circles, Enzymes and Ribozymes - PMC
    Viroid replication proceeds through an RNA-based rolling-circle mechanism with three steps that, with some variations, operate in both polarity strands.
  68. [68]
    Avsunviroidae family: Viroids containing hammerhead ribozymes
    Avsunviroidae, or hammerhead viroids, are viroids that can adopt hammerhead structures and self-cleave, and lack messenger activity.
  69. [69]
    Engineering circular RNA for enhanced protein production - Nature
    Jul 18, 2022 · Circular RNAs (circRNAs) are stable and prevalent RNAs in eukaryotic cells that arise from back-splicing. Synthetic circRNAs and some ...