Next Article in Journal
A Double-Humanized Mouse Model for Studying Host Gut Microbiome–Immune Interactions in Gulf War Illness
Previous Article in Journal
GLI Transcriptional Targets S100A7 and KRT16 Show Upregulated Expression Patterns in Epidermis Overlying the Tumor Mass in Melanoma Samples
Previous Article in Special Issue
Poly (I:C)-Induced microRNA-30b-5p Negatively Regulates the JAK/STAT Signaling Pathway to Mediate the Antiviral Immune Response in Silver Carp (Hypophthalmichthys molitrix) via Targeting CRFB5
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circular RNAs in Cell Cycle Regulation of Cancers

by
Pannathon Thamjamrassri
1,2,3 and
Chaiyaboot Ariyachet
1,2,*
1
Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
2
Center of Excellence in Hepatitis and Liver Cancer, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
3
Medical Biochemistry Program, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 6094; https://doi.org/10.3390/ijms25116094
Submission received: 28 April 2024 / Revised: 26 May 2024 / Accepted: 29 May 2024 / Published: 31 May 2024
(This article belongs to the Special Issue Recent Progress in Molecular Biology of RNA 2.0)

Abstract

:
Cancer has been one of the most problematic health issues globally. Typically, all cancers share a common characteristic or cancer hallmark, such as sustaining cell proliferation, evading growth suppressors, and enabling replicative immortality. Indeed, cell cycle regulation in cancer is often found to be dysregulated, leading to an increase in aggressiveness. These dysregulations are partly due to the aberrant cellular signaling pathway. In recent years, circular RNAs (circRNAs) have been widely studied and classified as one of the regulators in various cancers. Numerous studies have reported that circRNAs antagonize or promote cancer progression through the modulation of cell cycle regulators or their associated signaling pathways, directly or indirectly. Mostly, circRNAs are known to act as microRNA (miRNA) sponges. However, they also hold additional mechanisms for regulating cellular activity, including protein binding, RNA-binding protein (RBP) recruitment, and protein translation. This review will discuss the current knowledge of how circRNAs regulate cell cycle-related proteins through the abovementioned mechanisms in different cancers.

1. Introduction

In 2023, the United States reported that the number of new cancer cases reached 1,958,310, while the number of casualties was estimated at 609,820. Lung and colorectum cancers contribute to the highest number of cancer-related deaths in both men and women [1]. Many internal biological factors have been identified to drive cancer progression, such as oncogenes, mutations in cell cycle-regulated genes, and aberrations in signaling pathways [2,3]. It was shown that mutations in p53, a cell cycle suppressor gene, occurred in 60% of cancer cases [2]. Furthermore, the mutated site in p53 usually occurs in the DNA-binding domain, thus disabling it from interacting with DNA [4]. Amplification of cell cycle-encoding genes, including cyclin D, cyclin E, and cyclin-dependent kinase 4 (CDK4), can also occur in certain types of cancer [5]. Additionally, several cancers were found to alter their signaling pathways that control the cell cycle [3,6]. Numerous proteins in the signaling pathway, such as PI3K/Akt and MAPK signaling, can be dysregulated due to pathway hyperactivation, allowing excessive cell proliferation [5].
Discovered several decades ago, circular RNAs (circRNAs) were once identified as junk [7]. However, high-throughput sequencing and bioinformatics analysis revealed that they are abundantly synthesized throughout all eukaryotes [8,9]. In fact, circRNAs play an important role in many cellular and physiological processes under normal conditions [10]. When dysregulated, several circRNAs are thus expected to participate in cancer development. Multiple studies also indicated that circRNAs are involved in abnormal signal transduction in cancer initiation, progression, metastasis, stemness, and resistance to therapy [11]. For example, overexpression of circZNF609 activates the Hedgehog signaling pathway by sponging miR-15a-5p and miR-15b-5p to enhance the stemness of hepatocellular carcinoma (HCC) cells [12]. CircPTK2 sponges miR-200a-3p to promote breast cancer tumorigenesis through the modulation of HIPPO/YAP signaling [13]. Interestingly, dysregulated circRNAs may indirectly influence the alteration of cell cycle regulator expression via controlling signaling pathways. To illustrate, circSHPRH sponges multiple miRNA targets, a process which results in the inhibition of JAK/STAT signaling and a reduction in cyclin-dependent kinase (CDK) 4, and -6 expression [14]. However, the signaling pathway that circRNAs mediate to control cell cycle regulator expression in many studies is still left unexplored and certainly requires more attention.
This review will focus on the influence of circRNAs on cell cycle-related protein expression in different types of cancer and associated signaling pathways.

2. Circular RNA and Biogenesis

CircRNA is a single-strand noncoding RNA with a covalently closed structure, lacking 3′ poly(A) tail modification and 5′ cap [15] (Figure 1). It is produced from a pre-mRNA splicing event called backsplicing, in which the downstream splice-donor site is closely brought to the upstream splice-acceptor site in a loop-like manner [16]. Invert repeat sequences, such as Alu, and dimerized trans-acting RNA-binding proteins (RBPs) are known to mediate this loop formation [17]. These RBPs include protein quaking (HQI), fused in sarcoma (FUS), and nuclear factor 90/110 (NF90/NF110) [18,19]. Moreover, NF90/110 was found to promote the stabilization of intronic RNA pairs, enhancing circRNA biogenesis [20]. The combination of multiple heterogeneous nuclear ribonucleoproteins (hnRNPs) and serine-arginine (SR)-rich proteins can also drive circRNA formation [21]. In contrast, the adenosine deaminase (ADAR) enzyme and ATP-dependent RNA helicase A (DHX9) interfere with circRNA biogenesis by blocking the complementary base pairing between invert repeat sequences [22,23]. However, backsplicing is less preferred than canonical splicing due to its low efficiency [24].
In addition to backsplicing, circRNAs can be generated from exon skipping in an alternative splicing event to form looped structures containing both exons and introns. This structure is called lariat, which can further undergo circularization and become exon–intron circRNA (EIcircRNA) or be processed by internal splicing to remove the intron, becoming exonic circRNA (ecircRNA) [25,26]. In addition, the canonical splicing byproduct, intronic lariat, can also escape from debranching and eventually circularize into circular intronic RNA (ciRNA) [27]. Interestingly, some studies have revealed that the fused gene in some cancers can give rise to circRNAs. For example, F-circBA1 is a product derived from a fusion of the BCR and ABL genes in chronic myeloid leukemia [28]. F-circEA1 in non-small-cell lung cancer (NSCLC) also results from the fusion of the EML4 and ALK1 genes [29].

3. Functions of circRNAs

The majority of circRNAs are proposed to act as miRNA sponges or decoys. Although miRNA was found to share a miRNA-binding element (MRE) not only with circRNA but also with other non-coding RNAs such as long-noncoding RNAs and pseudogenic RNAs. Competing endogenous RNAs (ceRNAs) were termed to describe the non-coding RNAs that are capable of binding to MRE. Therefore, it can be implied that the interaction between circRNA and its target miRNA might fall under the ‘multiple-to-multiple’ model, which is composed of a complex regulatory network involving ceRNAs [30,31]. Factors known to influence ceRNA activity are its abundance, subcellular localization of ceRNAs and miRNAs, miRNA-ceRNA affinity, and interaction with RNA-binding proteins (RBPs) [32,33,34]. However, more study is needed to completely clarify ceRNA activity, which might deeply affect the action of circRNAs on miRNAs.
In addition to being miRNA sponges, some circRNAs have been confirmed to encode a protein with biological function [35]. More evidence also revealed that they can regulate the expression of the parental gene by interacting directly with the promoter or forming a protein complex. Indeed, circRNA is thus capable of recruiting RBPs, binding to proteins, or acting as a protein sponge [26].
Recent studies showed that circRNA plays an important role in several cellular activities via post-transcriptional regulation (Figure 1). Therefore, dysregulation in their expression could lead to abnormalities in many biological processes and might eventually lead to the development of disease, not only cancer. To illustrate, an upregulated circCHD2 promotes hepatic leukemia factor (HLF) expression to stimulate the activation of hepatic stellate cells, an essential driver of liver fibrosis [36,37].

3.1. Regulating the Transcription Activity of the Parental Gene

Interestingly, parental gene expression was found to be repressed by its transcribed circRNAs as they bind to the synthesis locus, forming an RNA-DNA hybrid called R-loop [38]. The interaction halts the parental gene transcription activity [38]. CircSEP3 and circSMARCA5 are known to mediate such functions [38,39]. EIcircRNA establishes the complex with U1 small nuclear ribonucleoprotein (U1 snRNP) and binds to the promotor to upregulate the expression of the parental gene. Similar to EIcircRNA, ciRNAs accumulate at their synthesis site and elongate polymerase III activity to increase parental gene expression [27].

3.2. RNA Binding

Other than regulating its parental gene, circRNA has been mostly described as miRNA sponges or miRNA decoys as they contain many miRNA-binding sites, protecting mRNA targets from degradation. Moreover, recent studies also revealed that they can bind to mRNA to directly modulate post-transcriptional regulation. To illustrate, circPAN3 reinforces the stability of interleukin-13 (IL-13) mRNA through direct interaction [40].

3.3. Protein Translation

N6-methyladenosine (m6A) is a type of RNA modification that can be controlled by readers, writers, and erasers. The presence of m6A in circRNA can lead to various regulation mechanisms, including circRNA metabolism, localization, degradation, biogenesis, and protein encoding [41,42,43,44,45]. YTH N6-methyladenosine RNA-binding protein F3 (YTHDF3) recognizes the m6A-modified start codon and interacts with eIF4G2 to initiate protein translation. Mett13/14 was found to promote this activity, whereas fat mass and obesity-associated protein (FTO) inhibit the translation [45]. With the presence of an internal ribosome entry site (IRES) in some circRNA, ribosomes can be recruited and begin translation [46]. Moreover, circZNF609 utilizes the combination of the abovementioned processes to translate proteins, as m6A modification enhances the efficiency of IRES-mediated translation [47]. However, the protein encoded from circRNA can act against or be analogous to that of its linear counterpart [48,49].

3.4. Protein Binding

Numerous studies have indicated the protein-binding function of circRNAs [15]. However, bioinformatics analysis revealed that circRNA features a lower RBP binding density compared to linear RNA, suggesting that many circRNAs cannot interact with proteins [50]. The binding of circRNA to proteins may result in a different action. CircRNA binds to two different proteins to facilitate their activity, acting as a scaffold. To illustrate, circFOXO3 recruits both p53 and mouse double minute 2 (MDM2), resulting in MDM2-mediated p53 ubiquitination and degradation [51]. Another example, circNFIX, binds to Y-box binding protein 1 (Ybx1) and Nedd4I, a ubiquitin ligase, to promote ubiquitination of Ybx1 by Nedd4I and trigger degradation [52]. In other circumstances, circRNA binds to one protein and inhibits it from interacting with the other protein that does not bind directly to circRNA. For example, circGSK3β interacts with its parental gene product, GSK3β, which further phosphorylates β-catenin. By binding to circRNA, GSK3β is therefore prevented from exerting its function to induce β-catenin degradation [53]. Similar to circGSK3β, polypyrimidine track-binding protein 1 (PTBP1) is targeted by circH19, and this interaction impedes PTBP1-mediated SREBP1 cleavage [54]. CircRNA can also yield a similar result by attracting both proteins, which were originally combined in order to function, and preventing their association [15]. Naturally, CCNB1 and CDK1 form a complex to initiate mitosis progression, including condensation of chromosomes, spindle pole assembly, and nuclear envelope breakdown [55,56]. CircCCNB1 abolishes their activity by binding to both CCNB1 and CDK1, impairing complex formation [57]. In addition, circRNA is also capable of recruiting RBPs, which regulate mRNA splicing, stability, and translation, thus forming the circRNA–protein–mRNA ternary complex. CircNSUN2 binds to IGF2BP2 and enhances its ability to stabilize HMGA2 mRNA [42]. CircPOK was also shown to possess a similar mechanism by recruiting interleukin enhancer binding factor 2/3 (ILF2/3). The complex formation allows ILF2/3 to stabilize its target, IL-6, and VEGF mRNA [58].

4. Cell Cycle and Regulators

The cell cycle is a biological process involving cellular growth and proliferation. It can be divided into four separate phases: G1, S, G2, and M phase or mitotic phase. G1, S, and G2 phases are included in interphase, while M phase can be divided into mitosis and cytokinesis [59,60]. Although cells can exit the cell cycle into a quiescent state (G0) under specific conditions [61]. The progression to the next phase is mainly regulated by CDK and cyclin [62].
During the G1 phase, CDK4 or CDK6 associates with cyclin D to form cyclin D–CDK4/6 complex, which partially phosphorylates retinoblastoma protein (RB) [63,64]. The RB phosphorylation triggers detachment between RB and its target, E2F, a transcriptional factor that facilitates early cell cycle gene expression [65]. E2F upregulates the protein levels of cyclin A, cyclin E, and c-Myc [66]. With the activation of c-Myc, CDK2 can bind to cyclin E to establish cyclin E–CDK2 complex and then fully phosphorylate RB, proceeding to the S phase [64]. In the S phase, cyclin A joins CDK2 to form cyclin A–CDK2 complex, which is essential in stimulating the expression of proteins involved in DNA synthesis [67]. After entering the G2 phase, cyclin B–CDK1 complex gradually increases throughout the phase from the beginning to the end, promoting cells to the M phase. However, cyclin B–CDK1 complex begins to decline slowly in the M phase, which in turn drives the completion of mitosis [68].
In order to be tightly controlled, cyclin or CDK are regulated by several cell cycle regulators, such as inhibitors of the CDK4 (INK4) family, which impede cell cycle progression at the G1 phase by competitively binding to CDK4 or CDK6. This prevents the association with D-type cyclins. The INK4 family contains p15INK4b (Cdkn2b), p16INK4a (Cdkn2a), p18INK4c (Cdkn2c), and p19INK4d (Cdkn2d). WEE1 kinase can also phosphorylate CDK1 and lead to its inactivation. Another well-known cell cycle inhibitor is the CIP/KIP protein family. Cdkn1a (p21CIP1), Cdkn1b (p27KIP1), and Cdkn1c (p57KIP2) are included in this protein family [69]. With the activation of p53, CIP/KIP could inhibit the kinase activity of CDK1, CDK2, and other cyclin–CDK complexes. The cell division cycle 25 (CDC25) protein family also participates in cell cycle regulation by dephosphorylating CDK, resulting in CDK activation. For instance, CDC25A dephosphorylates CDK2 in the cyclin A–CDK2 complex, while CDC25C removes phosphate from CDK1 in the cyclin A/B–CDK1 complex [63]. In addition, there are several cellular signaling pathways indirectly affecting the cell cycle such as the PI3K/Akt, Wnt/β-catenin, JAK/STAT, Hedgehog, and MAPK signaling pathways.
Cell cycle regulators can also be post-transcriptionally controlled via miRNA-mediated suppression. For example, the miR-15 cluster promotes cell cycle arrest by targeting multiple key cell cycle regulators, including CDK1, CDK2, CDK6, cyclin D1, cyclin D3, and cyclin E1 [70,71]. Similarly, CDK6 expression was also found to be repressed by miR-107 [72].

5. CircRNA and Cell Cycle Regulators in Cancer

CircRNAs have become important mediators in the biology of cancer, deeply entwined with cell cycle regulation. Their carcinogenic potential is demonstrated in Figure 2, where specific circRNAs influence cell cycle regulators to cause abnormal cell proliferation. On the other hand, circRNAs with tumor-suppressive characteristics are shown in Figure 3, frequently through the inhibition of important cell cycle machinery components. Table 1 presents a thorough summary of the varied functions of circRNAs in different types of cancer, emphasizing their complex significance in carcinogenesis and their potential as targets.

5.1. Bladder Cancer

Numerous cellular signaling pathways contribute to the progression of the cell cycle, and certain circRNAs utilize these pathways to manipulate the behavior of cancer cells. For instance, it has been demonstrated that the mitogen-activated protein kinase (MAPK) signaling pathway positively regulates cyclin D1 expression and enhances the activation of the cyclin–CDK complex during the G1 phase [195]. CircLRBA, a circRNA that is naturally upregulated in bladder cancer tissue and cell lines, is known to aggravate the cell cycle of bladder cancer via MAPK signaling manipulation. Mechanistically, circLRBA sponges miR-19b-3p to enhance citron Rho-interacting serine/threonine kinase (CIT) expression. CIT suppresses the activation of phosphorylated ERK1/2, which is a part of the downstream signaling pathway of MAPK. This leads to an increase in the expression of CDK1 and cyclin D1, while the activity of p53 is suppressed [73]. In addition to the MAPK signaling pathway, the Wnt/β-catenin signaling pathway also plays an important role in driving the cell cycle. The evidence indicates that β-catenin induces the expression of c-Myc and cyclin D1, leading to the advancement of the G1 phase [196,197]. By utilizing Wnt/β-catenin signaling, circRIMS1 can reinforce cell cycle activity by sponging miR-443-3p [74]. By counteracting the inhibitory influence of miR-443-3p on its target, cell division cycle and apoptosis regulator 1 (CCAR1), which is recognized as a co-activator of β-catenin [198]. An increase in CCAR1 level thus activates the downstream cascade of the pathway and ultimately upregulates c-Myc, respectively [74].
Several circRNAs were known to directly promote cell cycle-related proteins, regardless of the cellular signaling pathway. For example, hsa_circ_0058063 (circATIC) positively regulates CDK6 by endogenously competing with miR-1485-5p, which inhibits CDK6 mRNA translation. As a result, CDK6 expression is then enhanced due to the downregulation of miR-1485-5p [75]. Hsa_circ_0000615 (circZNF609) also sponges miR-1200 to increase its downstream target expression, CDC25B [76]. According to the prior study, CDC25B is required to stimulate the G2-M phase transition, and it also regulates checkpoint proteins in response to DNA damage [199]. Another example is that circMYLK can upregulate cyclin D3 expression by specifically targeting miR-34a. Once sequestrated by circMYLK, the downstream target of miR-34a, CCND3, can be more readily translated into cyclin D3 [77]. Recent research revealed that circGLIS3 can also support cell cycle progression by sponging miR-1273f, which targets S-phase kinase-associated protein (SKP1). The inhibition of miR-1273f leads to the activation of SKP1, which in turn stimulates the production of cyclin D1 and facilitates the transition from the G0/G1 phase to the S phase in the cell cycle [79].
Despite the majority of circRNAs that promote cell cycle progression, a few studies have also shown that some circRNAs have anti-cell cycle properties. Naturally downregulated in bladder cancer cell lines and tissue, circBCRC-3 was found to inhibit cell cycle progression by sponging miR-182-5p, thus upregulating its target expression, p27. The rise in p27 levels hinders the shift from G0/G1 to S phase [80]. Another essential signaling pathway involving the cell cycle is PI3K/Akt signaling. Multiple studies have indicated that PI3K/Akt signaling promotes cell cycle activity. To illustrate, Akt promotes the G1 progression by phosphorylating p21, which normally forms complexes with proliferating cell nuclear antigen (PCNA), cyclin, and CDK [200]. Phosphorylation of p21 in T145 residue leads to the dissociation of PCNA from the quaternary complex and a reduction in the binding affinity of p21 [200,201]. Free PCNA binds to DNA polymerase δ holoenzyme to promote DNA synthesis, while p21 decreases its inhibitory function on CDK2 [202]. CircITCH utilizes this signaling network to suppress bladder cancer proliferation by regulating multiple miRNAs, including miR-17 and miR-224. Notably, miR-17 specifically targets p21, while miR-224 specifically targets PTEN mRNA. The overexpression of circITCH thus results in the upregulation of p21 and PTEN, which is considered a key inhibitor of PI3K/Akt signaling [81,203]. Hsa_circ_0006117 (circPTPRA) impairs the G0/G1-S phase transition by interacting with insulin-like growth factor 2 binding protein 1 (IGF2BP1), an N6-methyladenosine (m6A) reader [82]. It is shown that IGF2BP1 can identify m6A-modified mRNAs and influence the stability and translation of the mRNA it targets [204]. Therefore, the interaction with circPTPRA blocks IGF2BP1 from recognizing m6A-modified mRNAs, including c-Myc mRNA, eventually suppressing the cell cycle [82]. Employing an uncommon molecular mechanism, circNR3C1 targets miR-27a-3p, which further binds to the 5′-UTR of CCND1 to promote cyclin D1 expression. Therefore, the reduction in miR-27a-3p sequestered by circNR3C1 has a detrimental impact on CCND1 translation, decreasing the cyclin D1 level and impeding the cell cycle [83].

5.2. Breast Cancer

Hsa_circ_0091074 (circTCONS_00016926) absorbs miR-1297, which is a repressor of transcriptional coactivator with PDZ-binding motif (TAZ). Given that TAZ is a downstream effector of the Hippo signaling pathway. Without the inhibition from miR-1297, TAZ is relieved from repression and induces the protein levels of cyclin D1, CDK4, and CDK6. However, circTCONS_00016926 is usually downregulated in breast cancer tissue and cell lines and does not exhibit cell cycle supportive activity in normal condition [84]. Naturally upregulated by the induction of estrogen, circPGR aggravates breast cancer progression by suppressing its target, miR-301a-5p, which partially binds to CDK1, CDK6, and checkpoint kinase 2 (CHEK2) mRNA. Thus, the decrease in miR-301a-5p level mediated by circPGR can lead to the upregulation of the translated products of the abovementioned mRNAs [85]. Interestingly, circHMCU was found to hold potential for binding miR-let7 family, which comprises of let-7a-5p, let-7c-5p, and let7-7f-5p. Therefore, their downstream target mRNAs: c-Myc; HMGA2; and cyclin D1, are then prevented from degradation. Furthermore, the expression of other cell cycle regulators, including cyclin B1, cyclin D3, CDK4, and CDK6, is also increased, despite that they are not the direct targets of these miRNAs [86]. CircUBE2D2 also contributes to cell cycle progression via sponging miR-512-3p to upregulate the expression of cell division cycle-associated 3 (CDCA3) [87]. Similarly, hsa_circ_0136666 (circPRKDC) stimulates the cell cycle by increasing the expression of CDK6. Without the suppression of miR-1299 that is sponged by circPRKDC, CDK6 abundance can flourish [88]. Using the same mechanism, hsa_circ_0004676 (circDHTKD1) sequesters miR-377-3p, which targets E2F6 mRNA, thus allowing E2F6 to be translated and further binds to partner of NOB1 homolog (PNO1) promotor to stimulate its expression. PNO1 then induces p53 ubiquitination, impairing its inhibitory effects on the cell cycle [90]. CircUBR1 is overexpressed in breast cancer, and it sponges miR-1299 that targets CCND1 mRNA. MiR-1299 is subsequently downregulated, while cyclin D1 is upregulated in the presence of circUBR1 [91]. Lastly, hsa_circ_0008039 (circPRKAR1B) supports E2F3 expression via suppressing its inhibitor, miR-432-5p [92].
There are the other circRNAs that mediate the cell cycle through downstream indirect effectors. To illustrate, the high abundance of hsa_circ_0000515 (circRPPH1) decreases the expression of p27, while increases the expression of cyclin D1, and CDK4. Mechanistically, circRPPH1 sponges miR-296-5p, abating the repressive effect on C-X-C motif chemokine ligand 10 (CXCL10) which positively affects the abovementioned cell cycle regulators [93]. CircPLK1 competes with miR-4500 and allows insulin-like growth factor (IGF1) mRNA, a target of miR-4500, to be translated. IGF1 later stimulates the upregulation of CDK4 and CDK6, supporting cell cycle activity [98].
Hsa_circ_0001785 (circELP3) decreases PCNA expression and intervenes in cell cycle progression by targeting miR-942. Subsequently, SOCS3 is overexpressed, inhibiting the JAK/STAT signaling pathway [99,205]. Surprisingly, circFBXW7 utilizes both miRNA sponging and protein translation mechanisms to inhibit breast cancer. It was shown that the protein encoded from circFBXW7 called FBXW7-185aa can bind to USP28 and suppresses c-Myc activity. Moreover, circFBXW7 itself also sponges miR-197-3p to upregulate the expression of its parental gene, F-box/WD repeat-containing protein 7 (FBXW7), which in turn synergizes the inhibitory effect in repressing c-Myc [100]. Another cell cycle-suppressive circRNA, circSETD2, lowers the expression of miR-155-5p by absorbing. This interaction relieves miR-155-5p target, signal peptide CUB domain and EGF like domain containing 2 (SCUBE2), from suppression. SCUBE2 then exerts its inhibitory function on CDK4 and cyclin D1, forestalling the G0/G1 to S phase transition [101].

5.3. Cervical Cancer

Most of the circRNAs reported in cervical cancer progression are cell cycle supporters such as circZFR, which upregulates many cell cycle regulators via binding to single-stranded DNA-binding protein 1 (SSBP1). This interaction results in the association of cyclin E1–CDK2 complex, which further phosphorylates Rb, releasing E2F1 and CDK2 expression, respectively [102]. Another example, hsa_circ_0000326 (circTCONS_l2_00004572), which sponges miR-338-3p to upregulate CDK4. Moreover, cyclin D1 expression is increased, whereas p21 and p27 are decreased [105]. Similarly, miR-1296 directly targets CDK2 mRNA to promote its degradation unless hsa_circ_0000520 (circRPPH1) abolishes the activity by sponging miR-1296. In addition to the upregulation of CDK2 mediated by circRPPH1, the expression of cyclin D1 is also increased, while that of p21 and p27 is decreased [94]. Found abundantly expressed in cervical cancer cell lines, hsa_circ_ 0000263 (circTCONS_00017720) suppresses p53 activity by sequestrating miR-150-5p. MDM4, a negative regulator of p53, is a target of miR-150-5p. Therefore, circTCONS_00017720-mediated miR-150-5p depletion leads to an increase in MDM4 and a decrease in p53, respectively [106]. Hsa_circ_0009035 (circRACGAP1) is also overexpressed in cervical cancer, and it aggravates cell cycle progression by sponging miR-889-3p. Therefore, homeobox B7 (HOXB7), a target of miR-889-3p, can now escape from miRNA-mediated degradation and stimulate PCNA expression [107]. Additionally, hsa_circ_0003221 (circPTK2) increases cytoplasmic polyadenylation element binding protein 4 (CPEB4) by sponging to miR-758-3p, leading to the upregulation of PCNA and cyclin D1 [108]. CircESRP1 supports cyclin D1 expression via suppressing the inhibitory effect of miR-634 and miR-142-3p to increase tumor protein D52 (TPD52) and ADP-ribosylation factor-like protein (ARL2), respectively. The overexpression of TPD52 or ARL2 leads to the upregulation of cyclin D1 and PCNA in the latter [109,110]. As well as hsa_circ_0000285 (circHIPK3) and circASAP1, which sponge miR-197-3p and miR-338-3p, respectively. MiR-197-3p represses ELK1 mRNA, while miR-338-3p interferes with RPP25 mRNA translation. The increased expression of PCNA and cyclin D1 can be observed when overexpressing ELK1 or RPP25 [111,112].

5.4. Colon Cancer

Hsa_circ_0008231 (circNBPF11) enhances colon cancer proliferation via PI3K/Akt signaling by endogenously competing with miR-338-3p. Without miR-338-3p-mediated repression, ETS proto-oncogene 1 (ETS1) expression is then increased and triggers phosphorylation of the upstream players of PI3K/Akt signaling, such as PI3K and Akt. Therefore, the upregulation of cyclin D1 is then followed [114]. Another study reported that circCTIC1 employs a different approach to modulate tumor-initiating cells in colon cancer. CircCTIC1 interacts with bromodomain PHD finger transcription factor (BPTF), one of the crucial partners of nuclear remodeling factor (NURF) complex, to mediate the localization into the nucleus. NURF complex binds to c-Myc promotor and thus increases c-Myc expression, promoting self-renewal capacity [115]. In the other way, circCSPP1 positively regulates Rho-associated coiled-coil containing protein kinase 1 (ROCK1) abundance through miR-431 inhibition. ROCK1 further promotes cyclin D1 and CDK4 expression, which, in turn, phosphorylate Rb to allow the G0/G1-S phase transition [116].

5.5. Colorectal Cancer

Utilizing Wnt/β-catenin signaling, hsa_circ_0005615 (circNFATC3) reinforces colorectal cancer cell behavior through increasing the expression of tankyrase (TNKS) by sponging miR-149-5p [117]. In the absence of Wnt, β-catenin is captured by the protein complex containing AXIN, adenomatous polyposis coli (APC), casein kinase 1 (CK1), and glycogen synthase kinase 3 (GSK-3), preventing β-catenin from functioning [206]. However, TNKS abolishes the suppressive effects of the complex by inhibiting AXIN to release β-catenin. Consequently, cyclin D1 expression is then mediated [117]. Unlike circNFATC3, circMETTL9 directly supports CDK6 expression through the inhibition of miR-551b-5p [118]. Similarly, the suppressive effect of miR-377 on E2F3 mRNA can be reversed by circPRMT5, that acts as a competing endogenous RNA [119]. Another example, hsa_circ_0007142 (circDOCK1) also sponges miR-122-5p, allowing its target CDC25A to express [120].
Nonetheless, there are circRNAs that can promote the cell cycle through indirect effectors. To illustrate, circCTNNA1 absorbs miR-149-5p to upregulate forkhead box protein M1 (FOXM1), a transcriptional activator of cyclin B1 and cyclin D1. This results in the rise of cyclin B1 and cyclin D1, along with the significant downregulation of p21 and p27 [121]. CircPRKDC and hsa_circ_0000512 (circRPPH1) promote cyclin D1 expression via different miRNAs and their downstream targets. CircPRKDC sponges miR-198 to increase discoidin domain receptor tyrosine kinase 1 (DDR1), whereas circRPPH1 binds to miR-296-5p to upregulate RUN family transcription factor 1 (RUNX1). Both DDR1 and RUNX1 further positively induce cyclin D1 levels [89,95]. Employing a similar approach, circZFR represses miR-147a to abrogate the inhibitory effect on CDK2-associated cullin domain 1 (CACUL1). Consequently, the increased expression of CDK2, CDK4, CDK6, and cyclin D1 is then conveyed by CACUL [103].

5.6. Endometrial Cancer

Hsa_circ_0002577 (circWDR26) was found to be upregulated in endometrial cancer tissue and cell lines. Moreover, circWDR26 modulates cell cycle progression by controlling the Wnt/β-catenin signaling pathway. As miR-197 interferes with the translation of catenin delta 1 (CTNND1), circWDR26 absorbs miR-197 to restore CTNND1 expression. CTNND1 thereby activates Wnt/β-catenin signaling, resulting in a surge of β-catenin, cyclin D1, and c-Myc [122]. In contrast to circWDR26, hsa_circ_0001610 (circTNFRSF21) uses a straightforward approach to promote cyclin B1 by repressing its suppressor, miR-139-5p [123].

5.7. Esophageal Cancer

Normally found upregulated, circFIG4 promotes cell cycle activity through sponging miR-493-5p, releasing E2F3 from being repressed [124]. Interestingly, circFNDC3B and hsa_circ_00014879 (circDCAF8) positively influence the same downstream target, CDC25A. CircFNDC3B mediates the action through inhibiting miR-214-3p, whereas circDCAF8 suppresses miR-519-3p [125,126]. Hsa_circ_0003340 (circOGDH) was reported to absorb miR-940 and modulate its downstream target expression, protein kinase AMP-activated catalytic subunit alpha 1 (PRKAA1). PRKAA1 can induce cyclin D1 and PCNA expression to positively regulate the cell cycle [127]. Similarly, hsa_circ_0120816 (circGFPT1) participates in cell cycle progression by sequestering miR-1305, which inhibits thioredoxin reductase 1 (TXNRD1) expression. Consequently, the upregulation of TXNRD1 is then mediated, followed by the increase in cyclin B1 induced by TXNRD1 [128].

5.8. Gastric Cancer

CircPGPEP1 was revealed to stimulate the G0/G1 to S phase transition as it targets miR-1297 to directly increase E2F3 abundance [129]. Likewise, hsa_circ_104433 competes with miR-497-5p, leading to the significant expression of its target, CDC25A. Interestingly, cyclin B1 was also found to be positively correlated to hsa_circ_104433 level [130]. Unlike the previous example, circRNF111 sponges miR-876-3p to restore Krüppel-like Factor 12 (KLF12). KLF12 subsequently mediates the upregulation of cyclin D1, thus supporting cell cycle [131].
On the other hand, circRNAs that act against cell cycle are, indeed, found to be downregulated in gastric cancer tissue and cell lines. To begin with, hsa_circ_0006470 (circMFN2) promotes p53 inducible protein 11 (TP53I11) by sponging miR-1234. The upregulation of TP53I11 prevents Akt phosphorylation and leads to the inhibition of the PI3K/Akt signaling pathway. Furthermore, TP53I11 also stimulates FOXO1 synthesis to suppress the activity of CDK2 [133]. As with circEVI5, it sequesters miR-4793-3p to relieve FOXO1 expression, followed by the encoding of p21 and p27 [134].

5.9. Glioma

Hsa_circ_0001495 (circCCNB1) can be induced by a transcription factor, eukaryotic translation initiation factor 4A3 (EIF4A3). CircCCNB1 employs multiple mechanisms for modulating glioma aggressiveness. It recruits HuR, an RNA-binding protein that functions as a stabilizer of CCND1 mRNA. Moreover, circCCNB1 simultaneously sponges miR-516b-5p to prevent miRNA-mediated degradation of CCND1, resulting in an increased cyclin D1 protein level [135]. Utilizing only miRNA sponge capability, hsa_circ_0001982 (circRNF111) sequesters miR-1205, which is an inhibitor of E2F1 mRNA. Due to the lack of miR-1205 presence, E2F1 is freed from repression and participates in the S phase transition [132]. Nonetheless, hsa_circ_0012129 (circB4GALT2) and hsa_circ_0030018 (circPOSTN) can also promote the progression of glioma without the direct control of cell cycle-related proteins. CircB4GALT2 upregulates TGIF2 expression through sponging miR-761. Subsequently, TGIF2 boosts the abundance of CDK2 and CDK4 to proceed to the S phase [136]. In a similar manner, circPOSTN absorbs miR-1297 to increase RAB21 level, which then facilitates the elevation of cyclin D1 and PCNA [137].
DNA damage in gliomas can induce circCDR1as, which serves as a protein sponge to attract p53. The interaction prevents p53 from establishing a complex with MDM2, its negative regulator [138]. MDM2 blocks transcriptional activation and subjects p53 for degradation mediated by ubiquitination [207]. Consequently, p53 and its downstream effector p21 are then increased, forestalling the cell cycle of glioma cells [138].

5.10. Hepatocellular Carcinoma

The majority of circRNA involving cell cycle activity in hepatocellular carcinoma is upregulated in both tumor tissue and cell lines. In addition, most of them are classified as cell cycle supporters. To begin with, hsa_circ_0036412 (circETFA) manipulates the Hedgehog signaling network through binding to ELAV like RNA-binding protein 1 (ELAVL1), an RNA-binding protein. ELAVL1 stabilizes GLI2 mRNA and increases protein levels [139]. GLI2 is an essential regulator in the Hedgehog signaling pathway. A previous study showed that downregulation of GLI2 negatively affects hepatocellular carcinoma proliferation due to the increase in p21 and p27 expression [208]. High GLI2 abundance mediated by circETFA then stimulates cyclin D1. In addition, circETFA utilizes miRNA sponge capability to synergistically enhance GLI2 production by sponging its suppressor, miR-579-3p [139].
Hsa_circ_0091581 (circGPC3) efficiently promotes hepatocellular carcinoma proliferation by establishing a positive feedback loop. To illustrate, miR-526b, which is a repressor of c-Myc mRNA, is sponged by circGPC3, thus upregulating c-Myc translation. The increase in c-Myc further stimulates circGPC3 production and cell cycle progression [140]. Hsa_circ_0000519 (circRPPH1) facilitates the G0/G1 to S phase transition through sponging miR-1296. Identified as one of the targets of miR-1296, E2F7 becomes upregulated when overexpressed with cirRPPH1. Interestingly, cyclin D1 and cyclin E1 were also found to be elevated, even though they are not direct targets to miR-1296 [96]. Another example, circROBO1 absorbs miR-130a-5p, which inhibits CCNT2 mRNA translation. This results in the reduction of miR-130a-5p, followed by the elevation of cyclin T2 level and allows the cell cycle to proceed to the M phase [142]. CircMMP9 acts via the miR-149/CCND2 axis to support cell cycle. Upon the downregulation of miR-149 by circMMP9, cyclin D2 expression is then increased [143]. Hsa_circ_101555 elevates CDCA3 expression by sponging its inhibitor, miR-145-5p. In the absence of miR-145-5p, CDCA3 abundance is thus more evident [144]. CircZNF83 also participates in cell proliferation via modulating indirect downstream targets. MiR-324-5p, a suppressor of CDK16 and a target of circZNF83, is then downregulated, allowing the expression of CDK16 to thrive. Moreover, circZNF83 relies on the JAK/STAT pathway to simultaneously aggravate cancer behavior, as the phosphorylated JAK2 and STAT3 were found to be increased. Nonetheless, the expression of the other cell cycle regulators has not been examined [145]. Lastly, circSLC7A11 endogenously silences miR-330-3p, which targets CDK1 mRNA, resulting in the downregulation of miR-330-3p and the upregulation of cyclin D1 expression, respectively. In addition, an increase in cyclin B1 level can be observed in circSLC7A11-enriched condition [146].
As previously mentioned, circGPC3 greatly supports cell cycle activity. A recent study showed that the other circular transcript of circGPC3, hsa_circ_0091579, can also contribute to cell cycle progression. Mechanistically, hsa_circ_0091579 sponges miR-1287 to inhibit miR-1287-mediated mRNA degradation on pyruvate dehydrogenase kinase 2 (PDK2) mRNA. An elevated PDK2 level positively influences cyclin D1 expression [141]. Similarly, hsa_circ_0005397 (circRHOT1), which is originally overexpressed in hepatocellular carcinoma, also sequesters miR-326 to upregulate PDK2. Indeed, an increase in cyclin D1 abundance can be detected, along with a decrease in p21 expression [147]. Hsa_circ_0000517, the other circular transcript of circRPPH1, promotes the transition of the cell cycle to the S phase by sponging miR-326. After circRNA-mediated sequestration, miR-326 level is then declined, which leads to the upregulation of its target, SMAD6. Cyclin D1 expression is subsequently upregulated under the influence of SMAD6 [97].
CircVAMP3 participates in anti-cell cycle activity as it features protein binding capability and acts against cell proliferation. Mechanistically, circVAMP3 directly binds to cell cycle-associated protein 1 (CAPRIN1) to block the interaction with G3BP stress granule assembly factor 1 (G3BP1). Without the complex resulting from the association between CAPRIN1 and G3PB1, the activation of c-Myc is restrained [148]. Another example, circLARP4, absorbs miR-761 to restore RUNX3. Thus, RUNX3 expression is elevated during miR-761 depletion and then positively influences p53 and p21 expression, inhibiting the cell cycle [149]. As well as hsa_circ_ 0000204 (circWDR37), which acts as cell cycle-suppressive circRNA. Through sponging miR-191, circWDR37 can upregulate KLF6 expression, which further inhibits cyclin D1, cyclin D2, and c-Myc [150].

5.11. Leukemia

As previously described, some circRNAs can be synthesized from fused genes in cancer, such as F-circBA1, which resulted from the fusion of BCR and ABL gene. A study indicated the cell cycle supportive role of F-circBA1 as it aggravates the transition of the G2 to M phase in chronic myeloid leukemia. F-circBA1 binds to miR-148b-3p which suppresses CDC25B mRNA translation, thus upregulating CDC25B abundance [28]. Hsa_circ_0005774 (circCDK1) sponges miR-192-5p to increase the expression of uncoordinated 51-like kinase 1 (ULK1). The report showed that ULK1 is a positive regulator of cyclin D1 and PCNA because these proteins are increased in the presence of ULK1 [151]. As well as hsa_circ_0094100 (circSFMBT2), it increases the upregulation of cyclin D1 and PCNA through the miR-217/ATP1B1 axis. The decline of miR-217 mediated by circSFMBT2 results in the elevation of ATPase Na+/K+-transporting subunit beta 1 (ATP1B1), which further stimulates cyclin D1 and PCNA [152]. In addition, circNFIX was found to upregulate the expression of cyclin D1. By sponging miR-876-3p, circNFIX can increase the level of tripartite motif containing 31 (TRIM31), which then activates the synthesis of cyclin D1 [153].

5.12. Lung Cancer

EGFR stimulation can lead to the synthesis of hsa_circ_0000190 (circCNIH4), an upregulated circRNA in non-small-cell lung cancer (NSCLC). CircCNIH4 inhibits miR-142-5p to further restore CDK6 expression. In addition, circCNIH4 also takes advantage of the MAPK signaling pathway by increasing the phosphorylation of MEK1/2 and ERK1/2. Therefore, the downstream targets of the pathway, such as CDK1, CDK4, and phosphorylated Rb, are upregulated [154]. Another circRNA that modulates cell signaling to facilitate cell cycle progression is hsa_circ_0014235 (circS100A2). MiR-146b-5p naturally interferes with the translation of Yes-associated protein (YAP) mRNA, which is classified as one of the key regulators in Hippo signaling [155]. Previous studies reported that YAP is involved in the activation of E2F, CDK2, CDK4, and CDK6 [209]. The increase in YAP mediated by the circS100A2/miR-146b-5p axis thus facilitates the production of CDK4 [155]. Moreover, circS100A2 can also utilize the miRNA sponge function to bind miR-520a-5p, which is a suppressor of CDK4 [156]. Taken together, the evidence indicates that circS100A2 can effectively upregulate CDK4 expression.
To upregulate cyclin E1, circDENND2A and hsa_circ_0062389 (circPI4KA) sponge miR-34a and miR-103a-3p, respectively. Coincidentally, miR-34a and miR-103a-3p repress CCNE1 mRNA translation. Furthermore, circDENND2A and circPI4KA are naturally overexpressed in NSCLC, thus leading to the efficient upregulation of cyclin E1 [157,158]. Hsa_circ_0012384 (circCMPK1) sponges miR-302e to relieve cyclin D1 from suppression [159]. As well as circABCB10, it sequesters miR-584-5p, which inhibits the translation of E2F5 mRNA. As a result, E2F5 is indeed upregulated in the presence of circABCB10 [160]. The same mechanism can be found in hsa_circ_0001421 (circSEC31A). CDCA3 mRNA is repressed by miR-4677-3p, but circSEC31A abolishes this effect by sponging miR-4677-3p and elevating CDCA3, respectively [161]. Another example, circSATB2, is overexpressed in NSCLC, and it sponges miR-33a-5p to support cancer proliferation. Without miRNA-mediated inhibition, E2F7, which is one of the targets of miR-33a-5p, can thrive [162]. Lastly, circPITX1 endogenously competes with miR-1248 to upregulate cyclin D2 expression, while hsa_circ_0006692 (circTCONS_00025531) binds to miR-205-5p to increase CDK19, along with some regulators that are non-miRNA targets, such as cyclin D1 and PCNA [163,164].
Many circRNAs also modulate lung cancer through the production of indirect effectors. For example, circTP63 sponges miR-873-3p to stimulate the expression of FOXM1, thus further activating the activity and expression of cyclin B1 [165]. Another example, circFOXM1, upregulates cyclin D1 and cyclin E1 levels through the miR-614/FAM83D axis. Upon suppression by circFOXM1, miR-614 is then declined, which facilitates the expression of FAM83D, a positive regulator of cyclin D1 and cyclin E1 [166]. A similar path of action can also be found in hsa_circ_0046263 (circP4HB), which binds to miR-940 to prevent repressive function in NOVA alternative splicing regulator 2 (NOVA2). Next, NOVA2 triggers the abundance of cyclin D1 and PCNA [167]. As well as hsa_circ_0072088 (circZFR), which facilitates the expression of NOVA2 through sponging miR-377-5p. An increase in cyclin D1 and PCNA is then followed [104].
Hsa_circ_0065214 (circSCAP) recruits splicing factor 3a subunit 3 (SF3A3) and induces ubiquitin-mediated degradation via proteasome. The decline in SF3A3 allows its suppressed target, MDM4 exon 6 skipping transcript (MDM4-S), to upregulate and eventually activate p53. In addition, the expression of CDK2, CDK4, CDK6, and cyclin D1 is depressed under circSCAP influence [168]. Likewise, circNOL10 directly binds to sex comb on midleg-like 1 (SCML1), a transcription factor, to prevent ubiquitination and facilitate transcription. Consequently, members of humanin (HN) polypeptide family, HN2 and HN8, are then transcribed and translated. HN2 and HN8 trigger the phosphorylation of checkpoint CHEK2, CDC25A, and E2F1 and activate p53. Moreover, it also decreases cyclin D1, CDK2, and phosphorylated Rb, thus impeding the cell cycle [169]. Employing a less complicated approach, circFBXO7 sequesters miR-296-3p, leading to the expression of KLF15. Acting as a transcription factor, KLF15 translocates to the nucleus and binds to p21 promotor to initiate its transcription [170]. Similarly, circKIF20B sponges miR-615-3p to upregulate myocyte enhancer factor 2A (MEF2A), which later suppresses CDK4 expression [210].

5.13. Melanoma

Hsa_circ_0082835 (circEZH2) sponges miR-429, which inhibits its parental gene expression, EZH2. With a lower miR-429 level, EZH2 is relieved from repression and further activates Wnt5a and β-catenin, which are important components in the Wnt/β-catenin signaling network. Indeed, downstream cell cycle regulators, including cyclin D1 and cyclin E1, are then upregulated [171]. Similarly, hsa_circ_0062270 (circCDC45) mediates melanoma phenotype by modulating its parental gene product, CDC45, and functions as a protein sponge as well. EIF4A3, an RNA-binding protein, is recruited to bind to circCDC45 and assist CDC45 mRNA stabilization. This results in an increase in CDC45 protein levels and promotes cell cycle [172].

5.14. Multiple Myeloma

CircPSAP promotes the cell cycle through the miR-331-3p/histone deacetylase 4 (HDAC4) axis. The suppression of miR-331-3p mediated by circPSAP leads to the upregulation of HDAC4, which further stimulates cyclin D1 expression to drive the transition to the S phase [173].
As it is downregulated in multiple myeloma, circMYBL2 exerts its anti-cancer effect by facilitating the complex formation of cyclin F and its parental gene product, MYBL2. This prevents the phosphorylation of MYBL2, a transcriptional regulator of several oncogenes, thus inactivating MYBL2 and impeding cell cycle progression [174].

5.15. Oral Squamous Cell Carcinoma

Hsa_circ_0000745 (circSPECC1) combines both protein binding and miRNA sponge function to upregulate cyclin D1. To illustrate, circSPECC1 recruits HuR to assist CCND1 mRNA stabilization. Moreover, it absorbs miR-488, which suppresses CCND1 translation. Therefore, circSPECC1 can efficiently promote cyclin D1 abundance [175]. Another example is circOSBPL10, a downregulated circRNA that also acts against cell cycle by downregulating miR-299-3p. Without the repression from miR-299-3p, its target, CDK6, can be overexpressed. Due to its downregulated status in oral squamous cell carcinoma tissue and cell lines, cell cycle supportive function of circOSBPL10 is usually disabled [176].
Utilizing the same approach as circSPECC1, circYAP also features protein binding capability to ameliorate oral squamous cell carcinoma aggression. CDK4 is recruited to the binding site located within circYAP. This interaction prevents complex formation between CDK4 and cyclin D1 and impairs nucleus translocation. Thus, cyclin D1 expression in the nucleus is diminished, including other regulators such as PCNA, c-Myc, and Rb [177].

5.16. Osteosarcoma

Through the circARF3/miR-1299/CDK6 axis, the naturally overexpressed circARF3 can readily sponge miR-1299 in osteosarcoma cell lines. The downregulation of miR-1299 results in the elevation of CDK6, thus promoting the G0/G1 to S phase transition [178]. Hsa_circ_0008934 (circASAP1) is also known to support E2F3 expression by sponging its suppressor, miR-145-5p. Consequently, E2F3 abundance becomes more pronounced in the absence of miR-145-5p [113]. Similarly, hsa_circ_0000073 (circOMA1) mediates cyclin E2 and MDM2 expression via miR-1252-5p. Both cyclin E2 and MDM2 have been proven to be targets of miR-1252-5p. Therefore, circOMA1-mediated miR-1252-5p repression leads to the upregulation of cyclin E2 and MDM2 [179]. Hsa_circ_0084582 (circCHD7) modulates the cell cycle through sponging miR-485-5p. Identified as a target of miR-485-5p, jagged 1 (JAG1) can be upregulated upon the suppression of miR-485-5p by circCHD7. JAG1 next stimulates c-Myc expression and aggravates osteosarcoma phenotype [180]. Another example, circSAMD4A, was found to be significantly overexpressed in doxorubicin-resistant osteosarcoma tissue and cell lines. It competes with miR-218-5p that targets KLF8, thus leading to the reduction in miR-218-5p and the promotion of KFL8, respectively. Under KLF8 influence, cyclin D1 is then upregulated, whereas p21 is downregulated [181].

5.17. Ovarian Cancer

Using a similar mode of action to circCNNB1 in glioma, circE2F2 serves as a protein sponge to recruit HuR to the binding site. HuR subsequently promotes E2F2 mRNA stabilization and therefore increases E2F2 protein level [182]. The other circRNA that supports the cell cycle in ovarian cancer is hsa_circ_0000714 (circAARS). The study reported that circAARS was found to be upregulated in paclitaxel-resistant ovarian tissue and cell lines. Through absorbing miR-370-3p, circAARS can mediate RAB17 expression, which further stimulates CDK6 and Rb, aggravating cancer behavior [183].

5.18. Pancreatic Ductal Adenocarcinoma

CircNEIL3 exerts its cell cycle supportive role by sponging miR-432-5p which, in turn, targets adenosine deaminase acting on RNA 1 (ADAR1). ADAR1 induces adenosine-to-inosine (A-to-I) editing of GLI family zinc finger 1 (GLI1) and performs a positive feedback loop to stimulate the production of circNEIL3 [184]. As with GLI2, which was previously mentioned, GLI1 is also considered a downstream important key in the Hedgehog pathway [211]. The evidence reported that GLI1 depletion led to the downregulation of many cell cycle regulators [212]. Therefore, the upregulation of ADAR1-induced GLI1 results in a surge of cyclin D1, cyclin E1, CDK2, CDK4, and CDK6 [184].
CircPVRL3 represses several cell cycle regulators by relying on the transactivation between the JAK/STAT and PI3K/Akt pathways [185]. To illustrate, the expression of Akt can be abolished by silencing STAT3 or JAK2 [213]. Another study showed that trans-signaling of the JAK/STAT pathway mediated by IL-6 also activates both the PI3K/AKT and MAPK networks, indicating the importance of JAK/STAT signaling to the PI3K/Akt pathway [214]. CircPVRL3 absorbs miR-194-5p, which is a repressor of suppressor of cytokine signaling 2 (SOCS2), an inhibitor of JAK/STAT. The high expression of SOCS2 is then mediated to inactivate the pathway, negatively affecting PI3K/Akt signaling in the process. Thus, many regulators driving the cell cycle, such as cyclin D1, cyclin E1, CDK2, and CDK4, are declined, whereas p21 is increased instead [185].

5.19. Prostate Cancer

Few studies were conducted to examine the role of circRNA and cell cycle-related proteins. For instance, hsa_circ_0057558 (circSLC39A10) was demonstrated to boost c-Myc, cyclin B1, and cyclin D1 expression via inhibiting miR-206 activity. Ubiquitin specific peptidase (USP33) is identified as a target of miR-206, and its function is to deubiquitinate c-Myc, thus allowing c-Myc to escape degradation and to upregulate the abovementioned proteins [186]. However, circHMGCS1 employs different mechanisms for enhancing cell cycle. By sponging miR-205-5p, its downstream target, Erb-B2 receptor tyrosine kinase 3 (ErBB3), is then upregulated. ErBB3 promotes phosphorylation of Akt and PI3K to activate the PI3K/Akt signaling pathway, resulting in an increase in cyclin D1 and CDK4 [187]. Directly bound to CCND1 mRNA, miR-195 can impede its translation into cyclin D1. However, miR-195 is usually repressed by circDPP4 in prostate cancer cell lines, thus facilitating cyclin D1 expression [188]. Lastly, circDPP4 functions as a sponge to attract miR-195, a repressor of CCND1. The reduction in miR-195 level is then mediated, followed by the elevation of cyclin D1, respectively [188].

5.20. Thyroid Cancer

Functioning as a protein recruiter, circEIF3I attracts AU-rich element RNA-binding factor 1 (AUF1) to the binding site. AUF1 is then occupied and cannot be used to trigger CCND1 mRNA degradation. Subsequently, cyclin D1 is significantly increased [189]. CircPRKCI and hsa_circ_0000644 (circKIAA1199) were identified to share the same target E2F3, yet they mediate through different miRNAs. CircPRKCI binds to miR-335 while circKIAA1199 sponges miR-1205 [190,191].
CircHACE1 exhibits an anti-cell cycle in thyroid cancer cells through the miR-346/TFC2L1 axis. The overexpression of circHACE1 leads to a reduction in miR-346 levels and an increase in TFC2L1, respectively. TFC2L1 then downregulated cyclin B1 and PCNA expression, therefore impeding the cell cycle [192].

5.21. Other Cancers

In laryngeal carcinoma, a naturally overexpressed circMYLK aggravates cancer cell proliferation by sponging miR-195. Consequently, CCND1 mRNA, a target of miR-195, is free from its repressor and can be translated into cyclin D1 [78]. As found upregulated in nasopharyngeal carcinoma, circCAMSAP1 manages to reinforce the cancer phenotype through bizarre function. Rather than recruiting to miRNA or protein, circCAMSAP1 directly binds and manipulates serpin family H member 1 (SERPINH1) mRNA, enhancing stabilization. High expression of SERPINH1 prevents the ubiquitination and degradation of c-Myc. Moreover, the positive feedback loop can also be stimulated by the complex formation of c-Myc and splicing factor 10 (SRSF10), resulting in the upregulation of circCAMSAP1 [193]. CircFAM114A2 acts against cell cycle activity in urothelial carcinoma by sponging miR-222-3p and miR-146a-5p. Both miRNAs target negative cell cycle regulators. MiR-222-3p suppresses p27 translation, whereas miR-146a-5p targets p21. Thus, the overexpression of circFAM114A2 leads to the expression of p27 and p21 [194].

6. Conclusions and Future Perspectives

Cancer is a global health issue and is considered life threatening. An aberrant expression of cell cycle regulators combined with a modulation of cellular signaling is one of the key driving factors that aggravates cancer behavior. Emerging research indicated that circRNAs might play an important role in regulating cancer aggressiveness since they are most likely to be dysregulated in cancer tissues and cell lines. CircRNAs can serve as either cell cycle supporters or cell cycle inhibitors, depending on the expression status, miRNA targets, mRNA targets, or protein targets. Most circRNAs have been reported to serve as miRNA sponges. The mounting evidence suggests that they might also hold other molecular mechanisms such as protein sponge, protein translation, mRNA sponge, and guiding A-to-I editing [215]. However, many studies have only identified a brief correlation between circRNAs and cell cycle activity, without examining whether circRNAs indeed cause the altered expression of cell cycle regulators.
Due to the advancement of bioinformatics tools and high-throughput RNA sequencing that have been rapidly developing, the identification of novel modes of circRNA-mediated gene regulation has thus gained more attention. A recent study demonstrated that circRNA–miRNA interactions can be predicted using a combination of deep learning and algorithms called KGDCMI. RNA characteristics in the form of digitalized data can be retrieved from algorithms before constructing a possible interaction network. Lastly, the calculation of the final predicted score will be made. The prediction accuracy was considerably high compared to that of the existing model, indicating the effectiveness of KGDCMI [216]. Other than predicting circRNA–miRNA interactions, many bioinformatics tools have also been recently developed for the prediction of circRNA–protein interactions in recent years. Recently, circRNA regulator identification tool (CRIT) has utilized multiple datasets such as gene alteration, clinical significance, gene ontology (GO) similarity, and transcriptome data collected from twelve cancer types. All the data are subjected to being characterized and clustered using non-negative matrix factorization (NMF), resulting in 73 predicted candidates and 17 known candidates [217]. Therefore, CRIT might be another efficient tool for predicting circRNA–protein interactions, as it revealed both novel and known regulators. These recent bioinformatics tools are undoubtedly useful for the future discovery of circRNA modulating cell cycle regulation in cancers.
In addition to the growing trend in establishing prediction tools, many molecular techniques have also been developed, especially for the study of circRNA–protein interaction. Employing RNA immunoprecipitation (RIP) might be effective in revealing RBPs, but it would require a suitable antibody. Thus, electrophoretic mobility shift assays (EMSAs) are more appropriate for determining the interaction between circRNAs and candidate proteins. Basically, both circRNA and RBPs are incubated in the mixture before being subjected to electrophoresis. Free circRNA tends to migrate faster than circRNA-bearing RBPs [15]. Nonetheless, disadvantages persist because binding circRNAs to RBPs necessitates a stable condition during gel run, and large circRNAs may not be suitable for this approach [218]. In addition, these interactions may not be physiologically present in the cells. Although these methods have their limitations, they may help us learn more about how circRNAs and their RBPs play a role in cell cycle dysregulation. Moreover, it could reveal the possibility of discovering biomarkers and therapeutic targets.
Given that circRNAs are abnormally expressed in each organ or cancer tissue, measuring their quantity would be a promising diagnosis. Reports suggest that the aggressiveness of the tumor influences the expression of some circRNAs. However, there are still some limitations, as it is common to find circRNAs in chunks and in extremely low abundance, which can complicate the process of identifying the specific one. Moreover, investigating the significance of circRNAs from biological samples, especially blood and plasma, is also considered a long procedure and may lead to incomplete identification [219,220]. Therefore, huge developments and research are required before utilizing circRNAs as a biomarker for disease diagnosis in clinical practice.

Author Contributions

Conceptualization, P.T. and C.A.; writing—original draft preparation, P.T.; writing—review and editing, C.A.; visualization, P.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by Ratchadaphiseksomphot Fund, Graduate Affairs, Faculty of Medicine, Chulalongkorn University (GA66/076) and Center of Excellence in Hepatitis and Liver Cancer, Faculty of Medicine, Chulalongkorn University.

Acknowledgments

We would like to thank all members in Center of Excellence in Hepatitis and Liver Cancer, Faculty of Medicine, Chulalongkorn University for their assistance in writing this review.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Hassanpour, S.H.; Dehghani, M. Review of cancer from perspective of molecular. J. Cancer Res. Pract. 2017, 4, 127–129. [Google Scholar] [CrossRef]
  3. Yip, H.Y.K.; Papa, A. Signaling Pathways in Cancer: Therapeutic Targets, Combinatorial Treatments, and New Developments. Cells 2021, 10, 659. [Google Scholar] [CrossRef] [PubMed]
  4. Taylor, W.R.; Stark, G.R. Regulation of the G2/M transition by p53. Oncogene 2001, 20, 1803–1815. [Google Scholar] [CrossRef] [PubMed]
  5. Sever, R.; Brugge, J.S. Signal transduction in cancer. Cold Spring Harb. Perspect. Med. 2015, 5, 006098. [Google Scholar] [CrossRef]
  6. Pan-cancer analysis of whole genomes. Nature 2020, 578, 82–93. [CrossRef] [PubMed]
  7. Hsu, M.T.; Coca-Prados, M. Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature 1979, 280, 339–340. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, P.L.; Bao, Y.; Yee, M.C.; Barrett, S.P.; Hogan, G.J.; Olsen, M.N.; Dinneny, J.R.; Brown, P.O.; Salzman, J. Circular RNA is expressed across the eukaryotic tree of life. PLoS ONE 2014, 9, e90859. [Google Scholar] [CrossRef] [PubMed]
  9. 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]
  10. Chen, L.L. The expanding regulatory mechanisms and cellular functions of circular RNAs. Nat. Rev. Mol. Cell Biol. 2020, 21, 475–490. [Google Scholar] [CrossRef]
  11. Papatsirou, M.; Artemaki, P.I.; Scorilas, A.; Kontos, C.K. The role of circular RNAs in therapy resistance of patients with solid tumors. Pers. Med. 2020, 17, 469–490. [Google Scholar] [CrossRef] [PubMed]
  12. He, Y.; Huang, H.; Jin, L.; Zhang, F.; Zeng, M.; Wei, L.; Tang, S.; Chen, D.; Wang, W. CircZNF609 enhances hepatocellular carcinoma cell proliferation, metastasis, and stemness by activating the Hedgehog pathway through the regulation of miR-15a-5p/15b-5p and GLI2 expressions. Cell Death Dis. 2020, 11, 358. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, X.; Ji, C.; Hu, J.; Deng, X.; Zheng, W.; Yu, Y.; Hua, K.; Zhou, X.; Fang, L. Hsa_circ_0005273 facilitates breast cancer tumorigenesis by regulating YAP1-hippo signaling pathway. J. Exp. Clin. Cancer Res. 2021, 40, 29. [Google Scholar] [CrossRef] [PubMed]
  14. Sun, D.; Zhu, D. Circular RNA hsa_circ_0001649 suppresses the growth of osteosarcoma cells via sponging multiple miRNAs. Cell Cycle 2020, 19, 2631–2643. [Google Scholar] [CrossRef] [PubMed]
  15. Huang, A.; Zheng, H.; Wu, Z.; Chen, M.; Huang, Y. Circular RNA-protein interactions: Functions, mechanisms, and identification. Theranostics 2020, 10, 3503–3517. [Google Scholar] [CrossRef]
  16. Kelly, S.; Greenman, C.; Cook, P.R.; Papantonis, A. Exon Skipping Is Correlated with Exon Circularization. J. Mol. Biol. 2015, 427, 2414–2417. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, X.O.; Wang, H.B.; Zhang, Y.; Lu, X.; Chen, L.L.; Yang, L. Complementary sequence-mediated exon circularization. Cell 2014, 159, 134–147. [Google Scholar] [CrossRef] [PubMed]
  18. Conn, S.J.; Pillman, K.A.; Toubia, J.; Conn, V.M.; Salmanidis, M.; Phillips, C.A.; Roslan, S.; Schreiber, A.W.; Gregory, P.A.; Goodall, G.J. The RNA binding protein quaking regulates formation of circRNAs. Cell 2015, 160, 1125–1134. [Google Scholar] [CrossRef] [PubMed]
  19. Errichelli, L.; Dini Modigliani, S.; Laneve, P.; Colantoni, A.; Legnini, I.; Capauto, D.; Rosa, A.; De Santis, R.; Scarfò, R.; Peruzzi, G.; et al. FUS affects circular RNA expression in murine embryonic stem cell-derived motor neurons. Nat. Commun. 2017, 8, 14741. [Google Scholar] [CrossRef]
  20. Li, X.; Liu, C.X.; Xue, W.; Zhang, Y.; Jiang, S.; Yin, Q.F.; Wei, J.; Yao, R.W.; Yang, L.; Chen, L.L. Coordinated circRNA Biogenesis and Function with NF90/NF110 in Viral Infection. Mol. Cell 2017, 67, 214–227.e217. [Google Scholar] [CrossRef]
  21. Kramer, M.C.; Liang, D.; Tatomer, D.C.; Gold, B.; March, Z.M.; Cherry, S.; Wilusz, J.E. Combinatorial control of Drosophila circular RNA expression by intronic repeats, hnRNPs, and SR proteins. Genes Dev. 2015, 29, 2168–2182. [Google Scholar] [CrossRef] [PubMed]
  22. Eisenberg, E.; Levanon, E.Y. A-to-I RNA editing—Immune protector and transcriptome diversifier. Nat. Rev. Genet. 2018, 19, 473–490. [Google Scholar] [CrossRef] [PubMed]
  23. Koh, H.R.; Xing, L.; Kleiman, L.; Myong, S. Repetitive RNA unwinding by RNA helicase A facilitates RNA annealing. Nucleic Acids Res. 2014, 42, 8556–8564. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, L.L.; Yang, L. Regulation of circRNA biogenesis. RNA Biol. 2015, 12, 381–388. [Google Scholar] [CrossRef] [PubMed]
  25. Li, Z.; Huang, C.; Bao, C.; Chen, L.; Lin, M.; Wang, X.; Zhong, G.; Yu, B.; Hu, W.; Dai, L.; et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat. Struct. Mol. Biol. 2015, 22, 256–264. [Google Scholar] [CrossRef] [PubMed]
  26. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.W.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The biogenesis, biology and characterization of circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef] [PubMed]
  27. 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] [PubMed]
  28. Tan, Y.; Huang, Z.; Wang, X.; Dai, H.; Jiang, G.; Feng, W. A novel fusion circular RNA F-circBA1 derived from the BCR-ABL fusion gene displayed an oncogenic role in chronic myeloid leukemia cells. Bioengineered 2021, 12, 4816–4827. [Google Scholar] [CrossRef] [PubMed]
  29. Huo, Y.; Lv, T.; Ye, M.; Zhu, S.; Liu, J.; Liu, H.; Song, Y. F-circEA1 regulates cell proliferation and apoptosis through ALK downstream signaling pathway in non-small cell lung cancer. Hum. Cell 2022, 35, 260–270. [Google Scholar] [CrossRef]
  30. Arancio, W.; Pizzolanti, G.; Genovese, S.I.; Baiamonte, C.; Giordano, C. Competing endogenous RNA and interactome bioinformatic analyses on human telomerase. Rejuvenation Res. 2014, 17, 161–167. [Google Scholar] [CrossRef]
  31. Qi, X.; Zhang, D.H.; Wu, N.; Xiao, J.H.; Wang, X.; Ma, W. ceRNA in cancer: Possible functions and clinical implications. J. Med. Genet. 2015, 52, 710–718. [Google Scholar] [CrossRef] [PubMed]
  32. Yuan, Y.; Liu, B.; Xie, P.; Zhang, M.Q.; Li, Y.; Xie, Z.; Wang, X. Model-guided quantitative analysis of microRNA-mediated regulation on competing endogenous RNAs using a synthetic gene circuit. Proc. Natl. Acad. Sci. USA 2015, 112, 3158–3163. [Google Scholar] [CrossRef] [PubMed]
  33. Loinger, A.; Shemla, Y.; Simon, I.; Margalit, H.; Biham, O. Competition between small RNAs: A quantitative view. Biophys. J. 2012, 102, 1712–1721. [Google Scholar] [CrossRef] [PubMed]
  34. Denzler, R.; Agarwal, V.; Stefano, J.; Bartel, D.P.; Stoffel, M. Assessing the ceRNA hypothesis with quantitative measurements of miRNA and target abundance. Mol. Cell 2014, 54, 766–776. [Google Scholar] [CrossRef] [PubMed]
  35. 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.e27. [Google Scholar] [CrossRef] [PubMed]
  36. Hu, P.; Guo, J.; Zhao, B.; Zhang, Z.; Zhu, J.; Liu, F. CircCHD2/miR-200b-3p/HLF Axis Promotes Liver Cirrhosis. J. Environ. Pathol. Toxicol. Oncol. 2022, 41, 1–10. [Google Scholar] [CrossRef] [PubMed]
  37. Nokkeaw, A.; Thamjamrassri, P.; Tangkijvanich, P.; Ariyachet, C. Regulatory Functions and Mechanisms of Circular RNAs in Hepatic Stellate Cell Activation and Liver Fibrosis. Cells 2023, 12, 378. [Google Scholar] [CrossRef] [PubMed]
  38. Xu, X.; Zhang, J.; Tian, Y.; Gao, Y.; Dong, X.; Chen, W.; Yuan, X.; Yin, W.; Xu, J.; Chen, K.; et al. CircRNA inhibits DNA damage repair by interacting with host gene. Mol. Cancer 2020, 19, 128. [Google Scholar] [CrossRef] [PubMed]
  39. Conn, V.M.; Hugouvieux, V.; Nayak, A.; Conos, S.A.; Capovilla, G.; Cildir, G.; Jourdain, A.; Tergaonkar, V.; Schmid, M.; Zubieta, C.; et al. A circRNA from SEPALLATA3 regulates splicing of its cognate mRNA through R-loop formation. Nat. Plants 2017, 3, 17053. [Google Scholar] [CrossRef] [PubMed]
  40. Zhu, P.; Zhu, X.; Wu, J.; He, L.; Lu, T.; Wang, Y.; Liu, B.; Ye, B.; Sun, L.; Fan, D.; et al. IL-13 secreted by ILC2s promotes the self-renewal of intestinal stem cells through circular RNA circPan3. Nat. Immunol. 2019, 20, 183–194. [Google Scholar] [CrossRef]
  41. Di Timoteo, G.; Dattilo, D.; Centrón-Broco, A.; Colantoni, A.; Guarnacci, M.; Rossi, F.; Incarnato, D.; Oliviero, S.; Fatica, A.; Morlando, M.; et al. Modulation of circRNA Metabolism by m(6)A Modification. Cell Rep. 2020, 31, 107641. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, R.X.; Chen, X.; Xia, L.P.; Zhang, J.X.; Pan, Z.Z.; Ma, X.D.; Han, K.; Chen, J.W.; Judde, J.G.; Deas, O.; et al. N(6)-methyladenosine modification of circNSUN2 facilitates cytoplasmic export and stabilizes HMGA2 to promote colorectal liver metastasis. Nat. Commun. 2019, 10, 4695. [Google Scholar] [CrossRef] [PubMed]
  43. Park, O.H.; Ha, H.; Lee, Y.; Boo, S.H.; Kwon, D.H.; Song, H.K.; Kim, Y.K. Endoribonucleolytic Cleavage of m(6)A-Containing RNAs by RNase P/MRP Complex. Mol. Cell 2019, 74, 494–507.e498. [Google Scholar] [CrossRef] [PubMed]
  44. Tang, C.; Xie, Y.; Yu, T.; Liu, N.; Wang, Z.; Woolsey, R.J.; Tang, Y.; Zhang, X.; Qin, W.; Zhang, Y.; et al. m(6)A-dependent biogenesis of circular RNAs in male germ cells. Cell Res. 2020, 30, 211–228. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, Y.; Fan, X.; Mao, M.; Song, X.; Wu, P.; Zhang, Y.; Jin, Y.; Yang, Y.; Chen, L.L.; Wang, Y.; et al. Extensive translation of circular RNAs driven by N(6)-methyladenosine. Cell Res. 2017, 27, 626–641. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, L.; Hou, C.; Chen, C.; Guo, Y.; Yuan, W.; Yin, D.; Liu, J.; Sun, Z. The role of N(6)-methyladenosine (m(6)A) modification in the regulation of circRNAs. Mol. Cancer 2020, 19, 105. [Google Scholar] [CrossRef] [PubMed]
  47. Legnini, I.; Di Timoteo, G.; Rossi, F.; Morlando, M.; Briganti, F.; Sthandier, O.; Fatica, A.; Santini, T.; Andronache, A.; Wade, M.; et al. Circ-ZNF609 Is a Circular RNA that Can Be Translated and Functions in Myogenesis. Mol. Cell 2017, 66, 22–37.e29. [Google Scholar] [CrossRef] [PubMed]
  48. Pan, Z.; Cai, J.; Lin, J.; Zhou, H.; Peng, J.; Liang, J.; Xia, L.; Yin, Q.; Zou, B.; Zheng, J.; et al. A novel protein encoded by circFNDC3B inhibits tumor progression and EMT through regulating Snail in colon cancer. Mol. Cancer 2020, 19, 71. [Google Scholar] [CrossRef]
  49. Yang, Y.; Gao, X.; Zhang, M.; Yan, S.; Sun, C.; Xiao, F.; Huang, N.; Yang, X.; Zhao, K.; Zhou, H.; et al. Novel Role of FBXW7 Circular RNA in Repressing Glioma Tumorigenesis. J. Natl. Cancer Inst. 2018, 110, 304–315. [Google Scholar] [CrossRef]
  50. You, X.; Vlatkovic, I.; Babic, A.; Will, T.; Epstein, I.; Tushev, G.; Akbalik, G.; Wang, M.; Glock, C.; Quedenau, C.; et al. Neural circular RNAs are derived from synaptic genes and regulated by development and plasticity. Nat. Neurosci. 2015, 18, 603–610. [Google Scholar] [CrossRef]
  51. Du, W.W.; Fang, L.; Yang, W.; Wu, N.; Awan, F.M.; Yang, Z.; Yang, B.B. Induction of tumor apoptosis through a circular RNA enhancing Foxo3 activity. Cell Death Differ. 2017, 24, 357–370. [Google Scholar] [CrossRef]
  52. 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] [PubMed]
  53. Hu, X.; Wu, D.; He, X.; Zhao, H.; He, Z.; Lin, J.; Wang, K.; Wang, W.; Pan, Z.; Lin, H.; et al. circGSK3β promotes metastasis in esophageal squamous cell carcinoma by augmenting β-catenin signaling. Mol. Cancer 2019, 18, 160. [Google Scholar] [CrossRef] [PubMed]
  54. Zhu, Y.; Gui, W.; Lin, X.; Li, H. Knock-down of circular RNA H19 induces human adipose-derived stem cells adipogenic differentiation via a mechanism involving the polypyrimidine tract-binding protein 1. Exp. Cell Res. 2020, 387, 111753. [Google Scholar] [CrossRef] [PubMed]
  55. Yuan, J.; Krämer, A.; Matthess, Y.; Yan, R.; Spänkuch, B.; Gätje, R.; Knecht, R.; Kaufmann, M.; Strebhardt, K. Stable gene silencing of cyclin B1 in tumor cells increases susceptibility to taxol and leads to growth arrest in vivo. Oncogene 2006, 25, 1753–1762. [Google Scholar] [CrossRef] [PubMed]
  56. Mashal, R.D.; Lester, S.; Corless, C.; Richie, J.P.; Chandra, R.; Propert, K.J.; Dutta, A. Expression of cell cycle-regulated proteins in prostate cancer. Cancer Res. 1996, 56, 4159–4163. [Google Scholar] [PubMed]
  57. Fang, L.; Du, W.W.; Awan, F.M.; Dong, J.; Yang, B.B. The circular RNA circ-Ccnb1 dissociates Ccnb1/Cdk1 complex suppressing cell invasion and tumorigenesis. Cancer Lett. 2019, 459, 216–226. [Google Scholar] [CrossRef] [PubMed]
  58. Guarnerio, J.; Zhang, Y.; Cheloni, G.; Panella, R.; Mae Katon, J.; Simpson, M.; Matsumoto, A.; Papa, A.; Loretelli, C.; Petri, A.; et al. Intragenic antagonistic roles of protein and circRNA in tumorigenesis. Cell Res. 2019, 29, 628–640. [Google Scholar] [CrossRef] [PubMed]
  59. Wang, Z. Regulation of Cell Cycle Progression by Growth Factor-Induced Cell Signaling. Cells 2021, 10, 3327. [Google Scholar] [CrossRef]
  60. Panagopoulos, A.; Altmeyer, M. The Hammer and the Dance of Cell Cycle Control. Trends Biochem. Sci. 2021, 46, 301–314. [Google Scholar] [CrossRef]
  61. Davis, P.K.; Ho, A.; Dowdy, S.F. Biological methods for cell-cycle synchronization of mammalian cells. BioTechniques 2001, 30, 1322–1326, 1328, 1330–1331. [Google Scholar] [CrossRef] [PubMed]
  62. Schafer, K.A. The cell cycle: A review. Vet. Pathol. 1998, 35, 461–478. [Google Scholar] [CrossRef]
  63. Sherr, C.J.; Roberts, J.M. CDK inhibitors: Positive and negative regulators of G1-phase progression. Genes Dev. 1999, 13, 1501–1512. [Google Scholar] [CrossRef]
  64. Lundberg, A.S.; Weinberg, R.A. Functional inactivation of the retinoblastoma protein requires sequential modification by at least two distinct cyclin-cdk complexes. Mol. Cell. Biol. 1998, 18, 753–761. [Google Scholar] [CrossRef] [PubMed]
  65. Arroyo, M.; Raychaudhuri, P. Retinoblastoma-repression of E2F-dependent transcription depends on the ability of the retinoblastoma protein to interact with E2F and is abrogated by the adenovirus E1A oncoprotein. Nucleic Acids Res. 1992, 20, 5947–5954. [Google Scholar] [CrossRef]
  66. Otto, T.; Sicinski, P. Cell cycle proteins as promising targets in cancer therapy. Nat. Rev. Cancer 2017, 17, 93–115. [Google Scholar] [CrossRef]
  67. Petersen, B.O.; Lukas, J.; Sørensen, C.S.; Bartek, J.; Helin, K. Phosphorylation of mammalian CDC6 by cyclin A/CDK2 regulates its subcellular localization. EMBO J. 1999, 18, 396–410. [Google Scholar] [CrossRef]
  68. Riabowol, K.; Draetta, G.; Brizuela, L.; Vandre, D.; Beach, D. The cdc2 kinase is a nuclear protein that is essential for mitosis in mammalian cells. Cell 1989, 57, 393–401. [Google Scholar] [CrossRef] [PubMed]
  69. Hydbring, P.; Malumbres, M.; Sicinski, P. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat. Rev. Mol. Cell Biol. 2016, 17, 280–292. [Google Scholar] [CrossRef]
  70. Bueno, M.J.; Malumbres, M. MicroRNAs and the cell cycle. Biochim. Biophys. Acta 2011, 1812, 592–601. [Google Scholar] [CrossRef]
  71. Aqeilan, R.I.; Calin, G.A.; Croce, C.M. miR-15a and miR-16-1 in cancer: Discovery, function and future perspectives. Cell Death Differ. 2010, 17, 215–220. [Google Scholar] [CrossRef] [PubMed]
  72. Nokkeaw, A.; Thamjamrassri, P.; Chantaravisoot, N.; Tangkijvanich, P.; Ariyachet, C. Long non-coding RNA H19 promotes proliferation in hepatocellular carcinoma cells via H19/miR-107/CDK6 axis. Oncol. Res. 2023, 31, 989–1005. [Google Scholar] [CrossRef]
  73. Liu, Z.; Yang, Y.; Yang, Z.; Xia, S.; Lin, D.; Xiao, B.; Xiu, Y. Novel circRNA_0071196/miRNA-19b-3p/CIT axis is associated with proliferation and migration of bladder cancer. Int. J. Oncol. 2020, 57, 767–779. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, F.; Fan, M.; Cai, Y.; Zhou, X.; Tai, S.; Yu, Y.; Wu, H.; Zhang, Y.; Liu, J.; Huang, S.; et al. Circular RNA circRIMS1 Acts as a Sponge of miR-433-3p to Promote Bladder Cancer Progression by Regulating CCAR1 Expression. Mol. Ther. Nucleic Acids 2020, 22, 815–831. [Google Scholar] [CrossRef] [PubMed]
  75. Sun, M.; Zhao, W.; Chen, Z.; Li, M.; Li, S.; Wu, B.; Bu, R. Circ_0058063 regulates CDK6 to promote bladder cancer progression by sponging miR-145-5p. J. Cell. Physiol. 2019, 234, 4812–4824. [Google Scholar] [CrossRef] [PubMed]
  76. Feng, D.; Lv, J.; Li, K.; Cao, Q.; Han, J.; Yu, H.; Cheng, Y.; Zhuang, J.; Cai, L.; Yang, H.; et al. CircZNF609 promotes bladder cancer progression and inhibits cisplatin sensitivity via miR-1200/CDC25B pathway. Cell Biol. Toxicol. 2023, 39, 1–18. [Google Scholar] [CrossRef] [PubMed]
  77. Ye, W.; Chen, L.; Feng, C.; Liang, T. CircMYLK promotes the growth, migration, invasion, and survival of bladder cancer cells by upregulating CCND3 level via competitively binding to miR-34a. Drug Dev. Res. 2021, 82, 1206–1216. [Google Scholar] [CrossRef] [PubMed]
  78. Duan, X.; Shen, N.; Chen, J.; Wang, J.; Zhu, Q.; Zhai, Z. Circular RNA MYLK serves as an oncogene to promote cancer progression via microRNA-195/cyclin D1 axis in laryngeal squamous cell carcinoma. Biosci. Rep. 2019, 39, BSR20190227. [Google Scholar] [CrossRef] [PubMed]
  79. Wu, S.; Yang, J.; Xu, H.; Wang, X.; Zhang, R.; Lu, W.; Yang, J.; Li, X.; Chen, S.; Zou, Y.; et al. Circular RNA circGLIS3 promotes bladder cancer proliferation via the miR-1273f/SKP1/Cyclin D1 axis. Cell Biol. Toxicol. 2022, 38, 129–146. [Google Scholar] [CrossRef]
  80. Xie, F.; Li, Y.; Wang, M.; Huang, C.; Tao, D.; Zheng, F.; Zhang, H.; Zeng, F.; Xiao, X.; Jiang, G. Circular RNA BCRC-3 suppresses bladder cancer proliferation through miR-182-5p/p27 axis. Mol. Cancer 2018, 17, 144. [Google Scholar] [CrossRef]
  81. Yang, C.; Yuan, W.; Yang, X.; Li, P.; Wang, J.; Han, J.; Tao, J.; Li, P.; Yang, H.; Lv, Q.; et al. Circular RNA circ-ITCH inhibits bladder cancer progression by sponging miR-17/miR-224 and regulating p21, PTEN expression. Mol. Cancer 2018, 17, 19. [Google Scholar] [CrossRef]
  82. Xie, F.; Huang, C.; Liu, F.; Zhang, H.; Xiao, X.; Sun, J.; Zhang, X.; Jiang, G. CircPTPRA blocks the recognition of RNA N(6)-methyladenosine through interacting with IGF2BP1 to suppress bladder cancer progression. Mol. Cancer 2021, 20, 68. [Google Scholar] [CrossRef]
  83. Zheng, F.; Wang, M.; Li, Y.; Huang, C.; Tao, D.; Xie, F.; Zhang, H.; Sun, J.; Zhang, C.; Gu, C.; et al. CircNR3C1 inhibits proliferation of bladder cancer cells by sponging miR-27a-3p and downregulating cyclin D1 expression. Cancer Lett. 2019, 460, 139–151. [Google Scholar] [CrossRef]
  84. Hu, J.; Ji, C.; Hua, K.; Wang, X.; Deng, X.; Li, J.; Graham, D.; Fang, L. Hsa_circ_0091074 regulates TAZ expression via microRNA-1297 in triple negative breast cancer cells. Int. J. Oncol. 2020, 56, 1314–1326. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, L.; Yi, J.; Lu, L.Y.; Zhang, Y.Y.; Wang, L.; Hu, G.S.; Liu, Y.C.; Ding, J.C.; Shen, H.F.; Zhao, F.Q.; et al. Estrogen-induced circRNA, circPGR, functions as a ceRNA to promote estrogen receptor-positive breast cancer cell growth by regulating cell cycle-related genes. Theranostics 2021, 11, 1732–1752. [Google Scholar] [CrossRef]
  86. Song, X.; Liang, Y.; Sang, Y.; Li, Y.; Zhang, H.; Chen, B.; Du, L.; Liu, Y.; Wang, L.; Zhao, W.; et al. circHMCU Promotes Proliferation and Metastasis of Breast Cancer by Sponging the let-7 Family. Mol. Ther. Nucleic Acids 2020, 20, 518–533. [Google Scholar] [CrossRef] [PubMed]
  87. Dou, D.; Ren, X.; Han, M.; Xu, X.; Ge, X.; Gu, Y.; Wang, X.; Zhao, S. CircUBE2D2 (hsa_circ_0005728) promotes cell proliferation, metastasis and chemoresistance in triple-negative breast cancer by regulating miR-512-3p/CDCA3 axis. Cancer Cell Int. 2020, 20, 454. [Google Scholar] [CrossRef]
  88. Liu, L.H.; Tian, Q.Q.; Liu, J.; Zhou, Y.; Yong, H. Upregulation of hsa_circ_0136666 contributes to breast cancer progression by sponging miR-1299 and targeting CDK6. J. Cell. Biochem. 2019, 120, 12684–12693. [Google Scholar] [CrossRef] [PubMed]
  89. Wang, G.; Li, Y.; Zhu, H.; Huo, G.; Bai, J.; Gao, Z. Circ-PRKDC Facilitates the Progression of Colorectal Cancer through miR-198/DDR1 Regulatory Axis. Cancer Manag. Res. 2020, 12, 12853–12865. [Google Scholar] [CrossRef]
  90. Shao, G.; Fan, X.; Zhang, P.; Liu, X.; Huang, L.; Ji, S. Circ_0004676 exacerbates triple-negative breast cancer progression through regulation of the miR-377-3p/E2F6/PNO1 axis. Cell Biol. Toxicol. 2023, 39, 2183–2205. [Google Scholar] [CrossRef]
  91. Zhang, L.; Sun, D.; Zhang, J.; Tian, Y. Circ-UBR1 facilitates proliferation, metastasis, and inhibits apoptosis in breast cancer by regulating the miR-1299/CCND1 axis. Life Sci. 2021, 266, 118829. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, Y.; Lu, C.; Zhou, Y.; Zhang, Z.; Sun, L. Circular RNA hsa_circ_0008039 promotes breast cancer cell proliferation and migration by regulating miR-432-5p/E2F3 axis. Biochem. Biophys. Res. Commun. 2018, 502, 358–363. [Google Scholar] [CrossRef] [PubMed]
  93. Cai, F.; Fu, W.; Tang, L.; Tang, J.; Sun, J.; Fu, G.; Ye, G. Hsa_circ_0000515 is a novel circular RNA implicated in the development of breast cancer through its regulation of the microRNA-296-5p/CXCL10 axis. FEBS J. 2021, 288, 861–883. [Google Scholar] [CrossRef]
  94. Zheng, Q.; Zhang, J.; Zhang, T.; Liu, Y.; Du, X.; Dai, X.; Gu, D. Hsa_circ_0000520 overexpression increases CDK2 expression via miR-1296 to facilitate cervical cancer cell proliferation. J. Transl. Med. 2021, 19, 314. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, L.; Wu, H.; Chu, F.; Zhang, L.; Xiao, X. Knockdown of circ_0000512 Inhibits Cell Proliferation and Promotes Apoptosis in Colorectal Cancer by Regulating miR-296-5p/RUNX1 Axis. Onco Targets Ther. 2020, 13, 7357–7368. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, Y.; Tang, H.; Zhang, Y.; Wang, Q.; Li, S.; Wang, Z.; Shi, X. Circular RNA hsa_circ_0000519 contributes to angiogenesis and tumor progression in hepatocellular carcinoma through the miR-1296/E2F7 axis. Hum. Cell 2023, 36, 738–751. [Google Scholar] [CrossRef] [PubMed]
  97. He, S.; Guo, Z.; Kang, Q.; Wang, X.; Han, X. Circular RNA hsa_circ_0000517 modulates hepatocellular carcinoma advancement via the miR-326/SMAD6 axis. Cancer Cell Int. 2020, 20, 360. [Google Scholar] [CrossRef] [PubMed]
  98. Lin, G.; Wang, S.; Zhang, X.; Wang, D. Circular RNA circPLK1 promotes breast cancer cell proliferation, migration and invasion by regulating miR-4500/IGF1 axis. Cancer Cell Int. 2020, 20, 593. [Google Scholar] [CrossRef]
  99. Li, Z.; Zheng, J.; Lin, W.; Weng, J.; Hong, W.; Zou, J.; Zhang, T.; Ye, C.; Chen, Y. Circular RNA hsa_circ_0001785 inhibits the proliferation, migration and invasion of breast cancer cells in vitro and in vivo by sponging miR-942 to upregulate SOCS3. Cell Cycle 2020, 19, 2811–2825. [Google Scholar] [CrossRef]
  100. Ye, F.; Gao, G.; Zou, Y.; Zheng, S.; Zhang, L.; Ou, X.; Xie, X.; Tang, H. circFBXW7 Inhibits Malignant Progression by Sponging miR-197-3p and Encoding a 185-aa Protein in Triple-Negative Breast Cancer. Mol. Ther. Nucleic Acids 2019, 18, 88–98. [Google Scholar] [CrossRef]
  101. Shen, Y.; Zhang, M.; Da, L.; Huang, W.; Zhang, C. Circular RNA circ_SETD2 represses breast cancer progression via modulating the miR-155-5p/SCUBE2 axis. Open Med. 2020, 15, 940–953. [Google Scholar] [CrossRef]
  102. Zhou, M.; Yang, Z.; Wang, D.; Chen, P.; Zhang, Y. The circular RNA circZFR phosphorylates Rb promoting cervical cancer progression by regulating the SSBP1/CDK2/cyclin E1 complex. J. Exp. Clin. Cancer Res. 2021, 40, 48. [Google Scholar] [CrossRef] [PubMed]
  103. Tan, Y.; Wang, K.; Kong, Y. Circular RNA ZFR promotes cell cycle arrest and apoptosis of colorectal cancer cells via the miR-147a/CACUL1 axis. J. Gastrointest. Oncol. 2022, 13, 1793–1804. [Google Scholar] [CrossRef] [PubMed]
  104. Tan, Z.; Cao, F.; Jia, B.; Xia, L. Circ_0072088 promotes the development of non-small cell lung cancer via the miR-377-5p/NOVA2 axis. Thorac. Cancer 2020, 11, 2224–2236. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, Z.; Ren, C.; Yang, L.; Zhang, X.; Liu, J.; Zhu, Y.; Jiang, D. Silencing of circular RNA_0000326 inhibits cervical cancer cell proliferation, migration and invasion by boosting microRNA-338-3p-dependent down-regulation of CDK4. Aging 2021, 13, 9119–9134. [Google Scholar] [CrossRef]
  106. Cai, H.; Zhang, P.; Xu, M.; Yan, L.; Liu, N.; Wu, X. Circular RNA hsa_circ_0000263 participates in cervical cancer development by regulating target gene of miR-150-5p. J. Cell. Physiol. 2019, 234, 11391–11400. [Google Scholar] [CrossRef] [PubMed]
  107. Zhao, X.; Dong, W.; Luo, G.; Xie, J.; Liu, J.; Yu, F. Silencing of hsa_circ_0009035 Suppresses Cervical Cancer Progression and Enhances Radiosensitivity through MicroRNA 889-3p-Dependent Regulation of HOXB7. Mol. Cell Biol. 2021, 41, e0063120. [Google Scholar] [CrossRef] [PubMed]
  108. Xie, H.; Wang, J.; Wang, B. Circular RNA Circ_0003221 Promotes Cervical Cancer Progression by Regulating miR-758-3p/CPEB4 Axis. Cancer Manag. Res. 2021, 13, 5337–5350. [Google Scholar] [CrossRef] [PubMed]
  109. Shi, P.; Zhang, X.; Lou, C.; Xue, Y.; Guo, R.; Chen, S. Hsa_circ_0084927 Regulates Cervical Cancer Advancement via Regulation of the miR-634/TPD52 Axis. Cancer Manag. Res. 2020, 12, 9435–9448. [Google Scholar] [CrossRef]
  110. Chen, L.; Zhang, X.; Wang, S.; Lin, X.; Xu, L. Circ_0084927 Facilitates Cervical Cancer Development via Sponging miR-142-3p and Upregulating ARL2. Cancer Manag. Res. 2020, 12, 9271–9283. [Google Scholar] [CrossRef]
  111. Zhang, W.; Zhang, S. Downregulation of circRNA_0000285 Suppresses Cervical Cancer Development by Regulating miR197-3p-ELK1 Axis. Cancer Manag. Res. 2020, 12, 8663–8674. [Google Scholar] [CrossRef] [PubMed]
  112. Feng, L.; Zhao, M.; Wu, A. CircASAP1 promotes the development of cervical cancer through sponging miR-338-3p to upregulate RPP25. Anti-Cancer Drugs 2022, 33, e155–e165. [Google Scholar] [CrossRef] [PubMed]
  113. Li, S.; Zeng, M.; Yang, L.; Tan, J.; Yang, J.; Guan, H.; Kuang, M.; Li, J. Hsa_circ_0008934 promotes the proliferation and migration of osteosarcoma cells by targeting miR-145-5p to enhance E2F3 expression. Int. J. Biochem. Cell Biol. 2020, 127, 105826. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, D.; Li, Y.; Xu, A.; Tang, W.; Yu, B. CircRNA RNA hsa_circ_0008234 Promotes Colon Cancer Progression by Regulating the miR-338-3p/ETS1 Axis and PI3K/AKT/mTOR Signaling. Cancers 2023, 15, 2068. [Google Scholar] [CrossRef] [PubMed]
  115. Zhan, W.; Liao, X.; Wang, Y.; Li, L.; Li, J.; Chen, Z.; Tian, T.; He, J. circCTIC1 promotes the self-renewal of colon TICs through BPTF-dependent c-Myc expression. Carcinogenesis 2019, 40, 560–568. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, J.; Zhou, L.; Chen, B.; Yu, Z.; Zhang, J.; Zhang, Z.; Hu, C.; Bai, Y.; Ruan, X.; Wang, S.; et al. Circular RNA circCSPP1 promotes the occurrence and development of colon cancer by sponging miR-431 and regulating ROCK1 and ZEB1. J. Transl. Med. 2022, 20, 58. [Google Scholar] [CrossRef] [PubMed]
  117. Ma, Z.; Han, C.; Xia, W.; Wang, S.; Li, X.; Fang, P.; Yin, R.; Xu, L.; Yang, L. circ5615 functions as a ceRNA to promote colorectal cancer progression by upregulating TNKS. Cell Death Dis. 2020, 11, 356. [Google Scholar] [CrossRef] [PubMed]
  118. Li, M.; Zhi, Z.; Jiang, X.; Duan, G.C.; Zhu, W.N.; Pang, Z.; Wang, L.; Ge, R.; Dai, X.; Liu, J.M.; et al. METTL9 derived circular RNA circ-METTL9 sponges miR-551b-5p to accelerate colorectal cancer progression by upregulating CDK6. Carcinogenesis 2023, 44, 463–475. [Google Scholar] [CrossRef]
  119. Yang, B.; Du, K.; Yang, C.; Xiang, L.; Xu, Y.; Cao, C.; Zhang, J.; Liu, W. CircPRMT5 circular RNA promotes proliferation of colorectal cancer through sponging miR-377 to induce E2F3 expression. J. Cell. Mol. Med. 2020, 24, 3431–3437. [Google Scholar] [CrossRef]
  120. Yin, W.; Xu, J.; Li, C.; Dai, X.; Wu, T.; Wen, J. Circular RNA circ_0007142 Facilitates Colorectal Cancer Progression by Modulating CDC25A Expression via miR-122-5p. Onco Targets Ther. 2020, 13, 3689–3701. [Google Scholar] [CrossRef]
  121. Chen, P.; Yao, Y.; Yang, N.; Gong, L.; Kong, Y.; Wu, A. Circular RNA circCTNNA1 promotes colorectal cancer progression by sponging miR-149-5p and regulating FOXM1 expression. Cell Death Dis. 2020, 11, 557. [Google Scholar] [CrossRef] [PubMed]
  122. Shen, Q.; He, T.; Yuan, H. Hsa_circ_0002577 promotes endometrial carcinoma progression via regulating miR-197/CTNND1 axis and activating Wnt/β-catenin pathway. Cell Cycle 2019, 18, 1229–1240. [Google Scholar] [CrossRef]
  123. Gu, X.; Shi, Y.; Dong, M.; Jiang, L.; Yang, J.; Liu, Z. Exosomal transfer of tumor-associated macrophage-derived hsa_circ_0001610 reduces radiosensitivity in endometrial cancer. Cell Death Dis. 2021, 12, 818. [Google Scholar] [CrossRef]
  124. Huang, Z.; Wang, C.; Zhao, X. circFIG 4 drives the carcinogenesis and metastasis of esophagus cancer via the miR-493-5p/E2F3 axis. Thorac. Cancer 2022, 13, 783–794. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, J.; Li, X.; Duan, C.; Jia, Y. CircFNDC3B knockdown restrains the progression of oesophageal squamous cell carcinoma through miR-214-3p/CDC25A axis. Clin. Exp. Pharmacol. Physiol. 2022, 49, 1209–1220. [Google Scholar] [CrossRef] [PubMed]
  126. Liu, Z.; Lu, X.; Wen, L.; You, C.; Jin, X.; Liu, J. Hsa_circ_0014879 regulates the radiosensitivity of esophageal squamous cell carcinoma through miR-519-3p/CDC25A axis. Anticancer. Drugs 2022, 33, e349–e361. [Google Scholar] [CrossRef] [PubMed]
  127. Guan, X.; Guan, X.; Wang, Y.; Lan, T.; Cheng, T.; Cui, Y.; Xu, H. Circ_0003340 downregulation mitigates esophageal squamous cell carcinoma progression by targeting miR-940/PRKAA1 axis. Thorac. Cancer 2022, 13, 1164–1175. [Google Scholar] [CrossRef]
  128. Li, X.; Song, L.; Wang, B.; Tao, C.; Shi, L.; Xu, M. Circ0120816 acts as an oncogene of esophageal squamous cell carcinoma by inhibiting miR-1305 and releasing TXNRD1. Cancer Cell Int. 2020, 20, 526. [Google Scholar] [CrossRef]
  129. Wang, Y.; Liu, X.; Wang, L.; Zhang, Z.; Li, Z.; Li, M. Circ_PGPEP1 Serves as a Sponge of miR-1297 to Promote Gastric Cancer Progression via Regulating E2F3. Dig. Dis. Sci. 2021, 66, 4302–4313. [Google Scholar] [CrossRef]
  130. Wei, W.; Mo, X.; Yan, L.; Huang, M.; Yang, Y.; Jin, Q.; Zhong, H.; Cao, W.; Wu, K.; Wu, L.; et al. Circular RNA Profiling Reveals That circRNA_104433 Regulates Cell Growth by Targeting miR-497-5p in Gastric Cancer. Cancer Manag. Res. 2020, 12, 15–30. [Google Scholar] [CrossRef]
  131. Wu, G.; Zhang, A.; Yang, Y.; Wu, D. Circ-RNF111 aggravates the malignancy of gastric cancer through miR-876-3p-dependent regulation of KLF12. World J. Surg. Oncol. 2021, 19, 259. [Google Scholar] [CrossRef] [PubMed]
  132. Ma, Z.; Ma, J.; Lang, B.; Xu, F.; Zhang, B.; Wang, X. Circ_0001982 Up-regulates the Expression of E2F1 by Adsorbing miR-1205 to Facilitate the Progression of Glioma. Mol. Biotechnol. 2023, 65, 466–476. [Google Scholar] [CrossRef] [PubMed]
  133. Xie, J.; Ning, Y.; Zhang, L.; Lin, Y.; Guo, R.; Wang, S. Overexpression of hsa_circ_0006470 inhibits the malignant behavior of gastric cancer cells via regulation of miR-1234/TP53I11 axis. Eur. J. Histochem. 2022, 66, 3477. [Google Scholar] [CrossRef] [PubMed]
  134. Yan, M.; Niu, L.; Liu, J.; Yao, Y.; Li, H. circEVI5 acts as a miR-4793-3p sponge to suppress the proliferation of gastric cancer. Cell Death Dis. 2021, 12, 774. [Google Scholar] [CrossRef] [PubMed]
  135. Li, X.; Wang, C.; Chen, G.; Zou, W.; Deng, Y.; Zhou, F. EIF4A3-induced circCCNB1 (hsa_circ_0001495) promotes glioma progression by elevating CCND1 through interacting miR-516b-5p and HuR. Metab. Brain Dis. 2022, 37, 819–833. [Google Scholar] [CrossRef] [PubMed]
  136. Xu, W.; Xue, R.; Xia, R.; Liu, W.W.; Zheng, J.W.; Tang, L.; Kang, L.Y.; Wang, W.; Wei, W.T. Sevoflurane impedes the progression of glioma through modulating the circular RNA has_circ_0012129/miR-761/TGIF2 axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 5534–5548. [Google Scholar] [CrossRef] [PubMed]
  137. Song, D.; Ye, L.; Xu, Z.; Jin, Y.; Zhang, L. CircRNA hsa_circ_0030018 regulates the development of glioma via regulating the miR-1297/RAB21 axis. Neoplasma 2021, 68, 391–403. [Google Scholar] [CrossRef] [PubMed]
  138. Lou, J.; Hao, Y.; Lin, K.; Lyu, Y.; Chen, M.; Wang, H.; Zou, D.; Jiang, X.; Wang, R.; Jin, D.; et al. Circular RNA CDR1as disrupts the p53/MDM2 complex to inhibit Gliomagenesis. Mol. Cancer 2020, 19, 138. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, L.; Li, B.; Yi, X.; Xiao, X.; Zheng, Q.; Ma, L. Circ_0036412 affects the proliferation and cell cycle of hepatocellular carcinoma via hedgehog signaling pathway. J. Transl. Med. 2022, 20, 154. [Google Scholar] [CrossRef]
  140. Wei, X.; Zheng, W.; Tian, P.; He, Y.; Liu, H.; Peng, M.; Li, X.; Liu, X. Oncogenic hsa_circ_0091581 promotes the malignancy of HCC cell through blocking miR-526b from degrading c-MYC mRNA. Cell Cycle 2020, 19, 817–824. [Google Scholar] [CrossRef]
  141. Shu, J.; Du, J.; Wang, F.; Cheng, Y.; Chen, G.; Xu, B.; Zhang, D.; Chen, S. Circ_0091579 enhances the malignancy of hepatocellular carcinoma via miR-1287/PDK2 axis. Open Life Sci. 2021, 16, 69–83. [Google Scholar] [CrossRef] [PubMed]
  142. Meng, H.; Li, R.; Xie, Y.; Mo, Z.; Zhai, H.; Zhang, G.; Liang, G.; Shi, X.; Zhou, B. Nanoparticles Mediated circROBO1 Silencing to Inhibit Hepatocellular Carcinoma Progression by Modulating miR-130a-5p/CCNT2 Axis. Int. J. Nanomed. 2023, 18, 1677–1693. [Google Scholar] [CrossRef] [PubMed]
  143. Li, X.; Fang, J.; Wei, G.; Chen, Y.; Li, D. CircMMP9 accelerates the progression of hepatocellular carcinoma through the miR-149/CCND2 axis. J. Gastrointest. Oncol. 2022, 13, 1875–1888. [Google Scholar] [CrossRef] [PubMed]
  144. Gu, X.; Zhang, J.; Ran, Y.; Pan, H.; Jia, J.; Zhao, Y.; Zhao, X.; Li, W.; Song, S.; Yu, X. Circular RNA hsa_circ_101555 promotes hepatocellular carcinoma cell proliferation and migration by sponging miR-145-5p and regulating CDCA3 expression. Cell Death Dis. 2021, 12, 356. [Google Scholar] [CrossRef]
  145. Zhao, J.A.; Nie, W.; Dong, L.; Liu, W.; Wei, W. A curcumin analog GL63 inhibits the malignant behaviors of hepatocellular carcinoma by inactivating the JAK2/STAT3 signaling pathway via the circular RNA zinc finger protein 83/microRNA-324-5p/cyclin-dependent kinase 16 axis. J. Gastroenterol. Hepatol. 2021, 36, 2967–2977. [Google Scholar] [CrossRef] [PubMed]
  146. Huang, Y.; Ge, W.; Ding, Y.; Zhang, L.; Zhou, J.; Kong, Y.; Cui, B.; Gao, B.; Qian, X.; Wang, W. The circular RNA circSLC7A11 functions as a mir-330-3p sponge to accelerate hepatocellular carcinoma progression by regulating cyclin-dependent kinase 1 expression. Cancer Cell Int. 2021, 21, 636. [Google Scholar] [CrossRef]
  147. Gong, J.; Du, C.; Sun, N.; Xiao, X.; Wu, H. Circular RNA hsa_circ_0005397 promotes hepatocellular carcinoma progression by regulating the miR-326/PDK2 axis. J. Gene Med. 2021, 23, e3332. [Google Scholar] [CrossRef] [PubMed]
  148. Chen, S.; Cao, X.; Zhang, J.; Wu, W.; Zhang, B.; Zhao, F. circVAMP3 Drives CAPRIN1 Phase Separation and Inhibits Hepatocellular Carcinoma by Suppressing c-Myc Translation. Adv. Sci. 2022, 9, e2103817. [Google Scholar] [CrossRef] [PubMed]
  149. Chen, Z.; Zuo, X.; Pu, L.; Zhang, Y.; Han, G.; Zhang, L.; Wu, J.; Wang, X. circLARP4 induces cellular senescence through regulating miR-761/RUNX3/p53/p21 signaling in hepatocellular carcinoma. Cancer Sci. 2019, 110, 568–581. [Google Scholar] [CrossRef]
  150. Tian, F.; Yu, C.; Wu, M.; Wu, X.; Wan, L.; Zhu, X. MicroRNA-191 promotes hepatocellular carcinoma cell proliferation by has_circ_0000204/miR-191/KLF6 axis. Cell Prolif. 2019, 52, e12635. [Google Scholar] [CrossRef]
  151. Li, Q.; Luan, Q.; Zhu, H.; Zhao, Y.; Ji, J.; Wu, F.; Yan, J. Circular RNA circ_0005774 contributes to proliferation and suppresses apoptosis of acute myeloid leukemia cells via circ_0005774/miR-192-5p/ULK1 ceRNA pathway. Biochem. Biophys. Res. Commun. 2021, 551, 78–85. [Google Scholar] [CrossRef] [PubMed]
  152. Cao, J.; Huang, S.; Li, X. Rapamycin inhibits the progression of human acute myeloid leukemia by regulating the circ_0094100/miR-217/ATP1B1 axis. Exp. Hematol. 2022, 112–113, 60–69.e62. [Google Scholar] [CrossRef] [PubMed]
  153. Huang, L.; Huang, L.; Ming, X.; Wu, J.; Liu, W.; Xiao, Y. CircNFIX knockdown inhibited AML tumorigenicity by the miR-876-3p/TRIM31 axis. Hematology 2022, 27, 1046–1055. [Google Scholar] [CrossRef] [PubMed]
  154. Ishola, A.A.; Chien, C.S.; Yang, Y.P.; Chien, Y.; Yarmishyn, A.A.; Tsai, P.H.; Chen, J.C.; Hsu, P.K.; Luo, Y.H.; Chen, Y.M.; et al. Oncogenic circRNA C190 Promotes Non-Small Cell Lung Cancer via Modulation of the EGFR/ERK Pathway. Cancer Res. 2022, 82, 75–89. [Google Scholar] [CrossRef] [PubMed]
  155. Niu, R.; Li, D.; Chen, J.; Zhao, W. Circ_0014235 confers Gefitinib resistance and malignant behaviors in non-small cell lung cancer resistant to Gefitinib by governing the miR-146b-5p/YAP/PD-L1 pathway. Cell Cycle 2022, 21, 86–100. [Google Scholar] [CrossRef] [PubMed]
  156. Xu, X.; Tao, R.; Sun, L.; Ji, X. Exosome-transferred hsa_circ_0014235 promotes DDP chemoresistance and deteriorates the development of non-small cell lung cancer by mediating the miR-520a-5p/CDK4 pathway. Cancer Cell Int. 2020, 20, 552. [Google Scholar] [CrossRef]
  157. Zhang, Y.; Shan, C.; Chen, Y.; Sun, S.; Liu, D.; Zhang, X.; Zhang, S. CircDENND2A Promotes Non-small Cell Lung Cancer Progression via Regulating MiR-34a/CCNE1 Signaling. Front. Genet. 2020, 11, 987. [Google Scholar] [CrossRef] [PubMed]
  158. She, Y.; Han, Y.; Zhou, G.; Jia, F.; Yang, T.; Shen, Z. hsa_circ_0062389 promotes the progression of non-small cell lung cancer by sponging miR-103a-3p to mediate CCNE1 expression. Cancer Genet. 2020, 241, 12–19. [Google Scholar] [CrossRef]
  159. Cui, D.; Qian, R.; Li, Y. Circular RNA circ-CMPK1 contributes to cell proliferation of non-small cell lung cancer by elevating cyclin D1 via sponging miR-302e. Mol. Genet. Genom. Med. 2020, 8, e999. [Google Scholar] [CrossRef]
  160. Ma, D.; Qin, Y.; Huang, C.; Chen, Y.; Han, Z.; Zhou, X.; Liu, H. Circular RNA ABCB10 promotes non-small cell lung cancer progression by increasing E2F5 expression through sponging miR-584-5p. Cell Cycle 2020, 19, 1611–1620. [Google Scholar] [CrossRef]
  161. Zhang, K.; Hu, H.; Xu, J.; Qiu, L.; Chen, H.; Jiang, X.; Jiang, Y. Circ_0001421 facilitates glycolysis and lung cancer development by regulating miR-4677-3p/CDCA3. Diagn. Pathol. 2020, 15, 133. [Google Scholar] [CrossRef] [PubMed]
  162. Liu, P.; Wang, M.; Tang, W.; Li, G.; Gong, N. Circ_SATB2 Attenuates the Anti-Tumor Role of Celastrol in Non-Small-Cell Lung Carcinoma Through Targeting miR-33a-5p/E2F7 Axis. OncoTargets Ther. 2020, 13, 11899–11912. [Google Scholar] [CrossRef] [PubMed]
  163. Yue, Q.; Xu, Y.; Deng, X.; Wang, S.; Qiu, J.; Qian, B.; Zhang, Y. Circ-PITX1 Promotes the Progression of Non-Small Cell Lung Cancer Through Regulating the miR-1248/CCND2 Axis. OncoTargets Ther. 2021, 14, 1807–1819. [Google Scholar] [CrossRef] [PubMed]
  164. Liao, J.; Chen, Z.; Luo, X.; Su, Y.; Huang, T.; Xu, H.; Lin, K.; Zheng, Q.; Zhang, L.; Lin, G.; et al. Hsa_circ_0006692 Promotes Lung Cancer Progression via miR-205-5p/CDK19 Axis. Genes 2022, 13, 846. [Google Scholar] [CrossRef] [PubMed]
  165. Cheng, Z.; Yu, C.; Cui, S.; Wang, H.; Jin, H.; Wang, C.; Li, B.; Qin, M.; Yang, C.; He, J.; et al. circTP63 functions as a ceRNA to promote lung squamous cell carcinoma progression by upregulating FOXM1. Nat. Commun. 2019, 10, 3200. [Google Scholar] [CrossRef] [PubMed]
  166. Yu, C.; Cheng, Z.; Cui, S.; Mao, X.; Li, B.; Fu, Y.; Wang, H.; Jin, H.; Ye, Q.; Zhao, X.; et al. circFOXM1 promotes proliferation of non-small cell lung carcinoma cells by acting as a ceRNA to upregulate FAM83D. J. Exp. Clin. Cancer Res. CR 2020, 39, 55. [Google Scholar] [CrossRef] [PubMed]
  167. Li, G.; Zhao, C.; Zhang, H.; Yu, J.; Sun, Y.; Zhang, Y. Hsa_circ_0046263 Drives the Carcinogenesis and Metastasis of Non-Small Cell Lung Cancer Through the Promotion of NOVA2 by Absorbing Mir-940 as a Molecular Sponge. Cancer Manag. Res. 2020, 12, 12779–12790. [Google Scholar] [CrossRef] [PubMed]
  168. Chen, D.; Zhou, H.; Cai, Z.; Cai, K.; Liu, J.; Wang, W.; Miao, H.; Li, H.; Li, R.; Li, X.; et al. CircSCAP interacts with SF3A3 to inhibit the malignance of non-small cell lung cancer by activating p53 signaling. J. Exp. Clin. Cancer Res. 2022, 41, 120. [Google Scholar] [CrossRef] [PubMed]
  169. Nan, A.; Chen, L.; Zhang, N.; Jia, Y.; Li, X.; Zhou, H.; Ling, Y.; Wang, Z.; Yang, C.; Liu, S.; et al. Circular RNA circNOL10 Inhibits Lung Cancer Development by Promoting SCLM1-Mediated Transcriptional Regulation of the Humanin Polypeptide Family. Adv. Sci. 2019, 6, 1800654. [Google Scholar] [CrossRef]
  170. Wang, Z.H.; Ye, L.L.; Xiang, X.; Wei, X.S.; Niu, Y.R.; Peng, W.B.; Zhang, S.Y.; Zhang, P.; Xue, Q.Q.; Wang, H.L.; et al. Circular RNA circFBXO7 attenuates non-small cell lung cancer tumorigenesis by sponging miR-296-3p to facilitate KLF15-mediated transcriptional activation of CDKN1A. Transl. Oncol. 2023, 30, 101635. [Google Scholar] [CrossRef]
  171. Sun, Y.; Hou, Z.; Luo, B.; Li, C.; Liu, J.; Liu, J.; Tang, J.; Yao, G. Circular RNA circRNA_0082835 promotes progression and lymphatic metastasis of primary melanoma by sponging microRNA miRNA-429. Bioengineered 2021, 12, 4159–4173. [Google Scholar] [CrossRef] [PubMed]
  172. Hao, T.; Yang, Y.; He, J.; Bai, J.; Zheng, Y.; Luo, Z. Knockdown of circular RNA hsa_circ_0062270 suppresses the progression of melanoma via downregulation of CDC45. Histol. Histopathol. 2022, 37, 373–383. [Google Scholar] [CrossRef] [PubMed]
  173. Ma, H.; Shen, L.; Yang, H.; Gong, H.; Du, X. Circular RNA circPSAP functions as an efficient miR-331-3p sponge to regulate proliferation, apoptosis and bortezomib sensitivity of human multiple myeloma cells by upregulating HDAC4. J. Pharmacol. Sci. 2022, 149, 27–36. [Google Scholar] [CrossRef] [PubMed]
  174. Sun, Y.M.; Wang, W.T.; Zeng, Z.C.; Chen, T.Q.; Han, C.; Pan, Q.; Huang, W.; Fang, K.; Sun, L.Y.; Zhou, Y.F.; et al. circMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood 2019, 134, 1533–1546. [Google Scholar] [CrossRef] [PubMed]
  175. Li, K.; Fan, X.; Yan, Z.; Zhan, J.; Cao, F.; Jiang, Y. Circ_0000745 strengthens the expression of CCND1 by functioning as miR-488 sponge and interacting with HuR binding protein to facilitate the development of oral squamous cell carcinoma. Cancer Cell Int. 2021, 21, 271. [Google Scholar] [CrossRef] [PubMed]
  176. Tai, Y.; Li, Y.; Zhang, M. Silencing of circ_OSBPL10 affects the functional behaviors of oral squamous cell carcinoma cells by the miR-299-3p/CDK6 axis. Arch. Oral Biol. 2022, 136, 105363. [Google Scholar] [CrossRef] [PubMed]
  177. Zhang, X.Y.; Tang, H.; Liu, Y.; Du, N.; Tian, S.; Dou, Y.Q. circYap inhibits oral squamous cell carcinoma by arresting cell cycle. Acta Odontol. Scand. 2022, 80, 117–124. [Google Scholar] [CrossRef] [PubMed]
  178. Gao, A.M.; Yuan, C.; Hu, A.X.; Liu, X.S. circ_ARF3 regulates the pathogenesis of osteosarcoma by sponging miR-1299 to maintain CDK6 expression. Cell Signal 2020, 72, 109622. [Google Scholar] [CrossRef] [PubMed]
  179. Ren, Z.; Yang, Q.; Guo, J.; Huang, H.; Li, B.; Yang, Z.; Tian, X. Circular RNA hsa_circ_0000073 Enhances Osteosarcoma Cells Malignant Behavior by Sponging miR-1252-5p and Modulating CCNE2 and MDM2. Front. Cell Dev. Biol. 2021, 9, 714601. [Google Scholar] [CrossRef]
  180. Gao, P.; Zhao, X.; Yu, K.; Zhu, Z. Circ_0084582 Facilitates Cell Growth, Migration, Invasion, and Angiopoiesis in Osteosarcoma via Mediating the miR-485-3p/JAG1 Axis. Front. Genet. 2021, 12, 690956. [Google Scholar] [CrossRef]
  181. Wei, W.; Ji, L.; Duan, W.; Zhu, J. CircSAMD4A contributes to cell doxorubicin resistance in osteosarcoma by regulating the miR-218-5p/KLF8 axis. Open Life Sci. 2020, 15, 848–859. [Google Scholar] [CrossRef] [PubMed]
  182. Zhang, M.; Xu, Y.; Zhang, Y.; Li, B.; Lou, G. Circular RNA circE2F2 promotes malignant progression of ovarian cancer cells by upregulating the expression of E2F2 protein via binding to HuR protein. Cell. Signal. 2021, 84, 110014. [Google Scholar] [CrossRef] [PubMed]
  183. Guo, M.; Li, S.; Zhao, X.; Yuan, Y.; Zhang, B.; Guan, Y. Knockdown of Circular RNA Hsa_circ_0000714 Can Regulate RAB17 by Sponging miR-370-3p to Reduce Paclitaxel Resistance of Ovarian Cancer Through CDK6/RB Pathway. OncoTargets Ther. 2020, 13, 13211–13224. [Google Scholar] [CrossRef] [PubMed]
  184. Shen, P.; Yang, T.; Chen, Q.; Yuan, H.; Wu, P.; Cai, B.; Meng, L.; Huang, X.; Liu, J.; Zhang, Y.; et al. CircNEIL3 regulatory loop promotes pancreatic ductal adenocarcinoma progression via miRNA sponging and A-to-I RNA-editing. Mol. Cancer 2021, 20, 51. [Google Scholar] [CrossRef] [PubMed]
  185. Chi, B.; Zheng, Y.; Xie, F.; Fu, W.; Wang, X.; Gu, J.; Yang, J.; Yin, J.; Cai, L.; Tang, P.; et al. Increased expression of miR-194-5p through the circPVRL3/miR-194-5p/SOCS2 axis promotes proliferation and metastasis in pancreatic ductal adenocarcinoma by activating the PI3K/AKT signaling pathway. Cancer Cell Int. 2022, 22, 415. [Google Scholar] [CrossRef]
  186. Ding, T.; Zhu, Y.; Jin, H.; Zhang, P.; Guo, J.; Zheng, J. Circular RNA circ_0057558 Controls Prostate Cancer Cell Proliferation Through Regulating miR-206/USP33/c-Myc Axis. Front. Cell Dev. Biol. 2021, 9, 644397. [Google Scholar] [CrossRef] [PubMed]
  187. Li, J.; Cao, X.; Chu, T.; Lin, K.; Chen, L.; Lv, J.; Tan, Y.; Chen, M.; Li, M.; Wang, K.; et al. The circHMGCS1-miR-205-5p-ErBB3 axis mediated the Sanggenon C-induced anti-proliferation effects on human prostate cancer. Pharmacol. Res. 2023, 187, 106584. [Google Scholar] [CrossRef] [PubMed]
  188. Yang, D.; Yang, B.; Zhu, Y.; Xia, Q.; Zhang, Y.; Zhu, X.; Guo, J.; Ding, T.; Zheng, J. Circular RNA-DPP4 serves an oncogenic role in prostate cancer progression through regulating miR-195/cyclin D1 axis. Cancer Cell Int. 2021, 21, 379. [Google Scholar] [CrossRef] [PubMed]
  189. Yao, X.; Liu, H.; Wang, Z.; Lu, F.; Chen, W.; Feng, Q.; Miao, Y.; Zhang, J.; Wang, Y.; Chen, Y.; et al. Circular RNA EIF3I promotes papillary thyroid cancer progression by interacting with AUF1 to increase Cyclin D1 production. Oncogene 2023, 42, 3206–3218. [Google Scholar] [CrossRef]
  190. Liu, Y.; Chen, G.; Wang, B.; Wu, H.; Zhang, Y.; Ye, H. Silencing circRNA protein kinase C iota (circ-PRKCI) suppresses cell progression and glycolysis of human papillary thyroid cancer through circ-PRKCI/miR-335/E2F3 ceRNA axis. Endocr. J. 2021, 68, 713–727. [Google Scholar] [CrossRef]
  191. Nie, C.; Han, J.; Bi, W.; Qiu, Z.; Chen, L.; Yu, J.; Pang, R.; Liu, B.; Sheng, R.; Zhang, J. Circular RNA circ_0000644 promotes papillary thyroid cancer progression via sponging miR-1205 and regulating E2F3 expression. Cell Cycle 2022, 21, 126–139. [Google Scholar] [CrossRef] [PubMed]
  192. Li, X.; Yang, S.; Zhao, C.; Yang, J.; Li, C.; Shen, W.; Hu, H.; Zhang, W.; Yang, S. CircHACE1 functions as a competitive endogenous RNA to curb differentiated thyroid cancer progression by upregulating Tfcp2L1 through adsorbing miR-346. Endocr. J. 2021, 68, 1011–1025. [Google Scholar] [CrossRef] [PubMed]
  193. Wang, Y.; Yan, Q.; Mo, Y.; Liu, Y.; Wang, Y.; Zhang, S.; Guo, C.; Wang, F.; Li, G.; Zeng, Z.; et al. Splicing factor derived circular RNA circCAMSAP1 accelerates nasopharyngeal carcinoma tumorigenesis via a SERPINH1/c-Myc positive feedback loop. Mol. Cancer 2022, 21, 62. [Google Scholar] [CrossRef] [PubMed]
  194. Lv, J.; Zhou, Z.; Wang, J.; Yang, X.; Yu, H.; Han, J.; Feng, D.; Yuan, B.; Wu, Q.; Li, P.; et al. CircFAM114A2 Promotes Cisplatin Sensitivity via miR-222-3p/P27 and miR-146a-5p/P21 Cascades in Urothelial Carcinoma. Front. Oncol. 2021, 11, 659166. [Google Scholar] [CrossRef] [PubMed]
  195. Pagès, G.; Lenormand, P.; L’Allemain, G.; Chambard, J.C.; Meloche, S.; Pouysségur, J. Mitogen-activated protein kinases p42mapk and p44mapk are required for fibroblast proliferation. Proc. Natl. Acad. Sci. USA 1993, 90, 8319–8323. [Google Scholar] [CrossRef]
  196. Shtutman, M.; Zhurinsky, J.; Simcha, I.; Albanese, C.; D’Amico, M.; Pestell, R.; Ben-Ze’ev, A. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc. Natl. Acad. Sci. USA 1999, 96, 5522–5527. [Google Scholar] [CrossRef] [PubMed]
  197. Massagué, J. G1 cell-cycle control and cancer. Nature 2004, 432, 298–306. [Google Scholar] [CrossRef]
  198. Chang, T.S.; Wei, K.L.; Lu, C.K.; Chen, Y.H.; Cheng, Y.T.; Tung, S.Y.; Wu, C.S.; Chiang, M.K. Inhibition of CCAR1, a Coactivator of β-Catenin, Suppresses the Proliferation and Migration of Gastric Cancer Cells. Int. J. Mol. Sci. 2017, 18, 460. [Google Scholar] [CrossRef]
  199. Liu, K.; Zheng, M.; Lu, R.; Du, J.; Zhao, Q.; Li, Z.; Li, Y.; Zhang, S. The role of CDC25C in cell cycle regulation and clinical cancer therapy: A systematic review. Cancer Cell Int. 2020, 20, 213. [Google Scholar] [CrossRef]
  200. Waga, S.; Hannon, G.J.; Beach, D.; Stillman, B. The p21 inhibitor of cyclin-dependent kinases controls DNA replication by interaction with PCNA. Nature 1994, 369, 574–578. [Google Scholar] [CrossRef]
  201. Rössig, L.; Jadidi, A.S.; Urbich, C.; Badorff, C.; Zeiher, A.M.; Dimmeler, S. Akt-dependent phosphorylation of p21(Cip1) regulates PCNA binding and proliferation of endothelial cells. Mol. Cell. Biol. 2001, 21, 5644–5657. [Google Scholar] [CrossRef] [PubMed]
  202. Flores-Rozas, H.; Kelman, Z.; Dean, F.B.; Pan, Z.Q.; Harper, J.W.; Elledge, S.J.; O’Donnell, M.; Hurwitz, J. Cdk-interacting protein 1 directly binds with proliferating cell nuclear antigen and inhibits DNA replication catalyzed by the DNA polymerase delta holoenzyme. Proc. Natl. Acad. Sci. USA 1994, 91, 8655–8659. [Google Scholar] [CrossRef] [PubMed]
  203. Vazquez, F.; Sellers, W.R. The PTEN tumor suppressor protein: An antagonist of phosphoinositide 3-kinase signaling. Biochim. Biophys. Acta 2000, 1470, M21–M35. [Google Scholar] [CrossRef] [PubMed]
  204. Huang, H.; Weng, H.; Sun, W.; Qin, X.; Shi, H.; Wu, H.; Zhao, B.S.; Mesquita, A.; Liu, C.; Yuan, C.L.; et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 2018, 20, 285–295. [Google Scholar] [CrossRef] [PubMed]
  205. Yoshimura, A.; Ohkubo, T.; Kiguchi, T.; Jenkins, N.A.; Gilbert, D.J.; Copeland, N.G.; Hara, T.; Miyajima, A. A novel cytokine-inducible gene CIS encodes an SH2-containing protein that binds to tyrosine-phosphorylated interleukin 3 and erythropoietin receptors. EMBO J. 1995, 14, 2816–2826. [Google Scholar] [CrossRef] [PubMed]
  206. Reyes, M.; Flores, T.; Betancur, D.; Peña-Oyarzún, D.; Torres, V.A. Wnt/β-Catenin Signaling in Oral Carcinogenesis. Int. J. Mol. Sci. 2020, 21, 4682. [Google Scholar] [CrossRef] [PubMed]
  207. Wang, S.; Zhao, Y.; Aguilar, A.; Bernard, D.; Yang, C.Y. Targeting the MDM2-p53 Protein-Protein Interaction for New Cancer Therapy: Progress and Challenges. Cold Spring Harb. Perspect. Med. 2017, 7, 026245. [Google Scholar] [CrossRef] [PubMed]
  208. Kim, Y.; Yoon, J.W.; Xiao, X.; Dean, N.M.; Monia, B.P.; Marcusson, E.G. Selective down-regulation of glioma-associated oncogene 2 inhibits the proliferation of hepatocellular carcinoma cells. Cancer Res. 2007, 67, 3583–3593. [Google Scholar] [CrossRef] [PubMed]
  209. Xiao, Y.; Dong, J. The Hippo Signaling Pathway in Cancer: A Cell Cycle Perspective. Cancers 2021, 13, 6214. [Google Scholar] [CrossRef]
  210. Wei, S.L.; Ye, J.J.; Sun, L.; Hu, L.; Wei, Y.Y.; Zhang, D.W.; Xu, M.M.; Fei, G.H. Exosome-derived circKIF20B suppresses gefitinib resistance and cell proliferation in non-small cell lung cancer. Cancer Cell Int. 2023, 23, 129. [Google Scholar] [CrossRef]
  211. Avery, J.T.; Zhang, R.; Boohaker, R.J. GLI1: A Therapeutic Target for Cancer. Front. Oncol. 2021, 11, 673154. [Google Scholar] [CrossRef] [PubMed]
  212. Zhang, R.; Wu, J.; Ferrandon, S.; Glowacki, K.J.; Houghton, J.A. Targeting GLI by GANT61 involves mechanisms dependent on inhibition of both transcription and DNA licensing. Oncotarget 2016, 7, 80190–80207. [Google Scholar] [CrossRef] [PubMed]
  213. Lu, Y.; Zhou, J.; Xu, C.; Lin, H.; Xiao, J.; Wang, Z.; Yang, B. JAK/STAT and PI3K/AKT pathways form a mutual transactivation loop and afford resistance to oxidative stress-induced apoptosis in cardiomyocytes. Cell. Physiol. Biochem. 2008, 21, 305–314. [Google Scholar] [CrossRef] [PubMed]
  214. Zegeye, M.M.; Lindkvist, M.; Fälker, K.; Kumawat, A.K.; Paramel, G.; Grenegård, M.; Sirsjö, A.; Ljungberg, L.U. Activation of the JAK/STAT3 and PI3K/AKT pathways are crucial for IL-6 trans-signaling-mediated pro-inflammatory response in human vascular endothelial cells. Cell Commun. Signal. CCS 2018, 16, 55. [Google Scholar] [CrossRef] [PubMed]
  215. Feng, X.Y.; Zhu, S.X.; Pu, K.J.; Huang, H.J.; Chen, Y.Q.; Wang, W.T. New insight into circRNAs: Characterization, strategies, and biomedical applications. Exp. Hematol. Oncol. 2023, 12, 91. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, X.F.; Yu, C.Q.; Li, L.P.; You, Z.H.; Huang, W.Z.; Li, Y.C.; Ren, Z.H.; Guan, Y.J. KGDCMI: A New Approach for Predicting circRNA-miRNA Interactions From Multi-Source Information Extraction and Deep Learning. Front. Genet. 2022, 13, 958096. [Google Scholar] [CrossRef] [PubMed]
  217. Shao, M.; Hao, S.; Jiang, L.; Cai, Y.; Zhao, X.; Chen, Q.; Gao, X.; Xu, J. CRIT: Identifying RNA-binding protein regulator in circRNA life cycle via non-negative matrix factorization. Mol. Ther. Nucleic Acids 2022, 30, 398–406. [Google Scholar] [CrossRef] [PubMed]
  218. Ulshöfer, C.J.; Pfafenrot, C.; Bindereif, A.; Schneider, T. Methods to study circRNA-protein interactions. Methods 2021, 196, 36–46. [Google Scholar] [CrossRef]
  219. Zhou, Q.; Ju, L.L.; Ji, X.; Cao, Y.L.; Shao, J.G.; Chen, L. Plasma circRNAs as Biomarkers in Cancer. Cancer Manag. Res. 2021, 13, 7325–7337. [Google Scholar] [CrossRef] [PubMed]
  220. Allegra, A.; Cicero, N.; Tonacci, A.; Musolino, C.; Gangemi, S. Circular RNA as a Novel Biomarker for Diagnosis and Prognosis and Potential Therapeutic Targets in Multiple Myeloma. Cancers 2022, 14, 1700. [Google Scholar] [CrossRef]
Figure 1. CircRNA biogenesis and its functions. CircRNA is a product of backsplicing, which begins with the joining of the downstream splice-donor site and the upstream splice-acceptor site, forming a circular loop-like structure. Alu or RNA-binding proteins (RBPs) can stimulate this interaction. The current circular loop-like structure, EIcircRNA, can also be generated via an exon-skipping event, and it might proceed through an internal splicing event to eliminate the intron, resulting in an ecircRNA. Moreover, canonical splicing can generate the other type of circRNA, ciRNA, which is entirely composed of introns. CircRNAs might be imported to the cytoplasm, where they primarily function. Mostly, circRNAs possess the ability to bind to miRNAs to regulate gene expression. CircRNA was recently found to be a template for protein translation or to establish an interaction with proteins. It can act like a protein sponge, which might later facilitate or inhibit the interaction between two different proteins. Utilizing protein sponge activity, some proteins avoid ubiquitin-mediated degradation by binding to circRNAs. Remarkably, some circRNAs have also been discovered to regulate their parental genes through various mechanisms, such as directly binding to the promotor or recruiting proteins to mediate gene expression.
Figure 1. CircRNA biogenesis and its functions. CircRNA is a product of backsplicing, which begins with the joining of the downstream splice-donor site and the upstream splice-acceptor site, forming a circular loop-like structure. Alu or RNA-binding proteins (RBPs) can stimulate this interaction. The current circular loop-like structure, EIcircRNA, can also be generated via an exon-skipping event, and it might proceed through an internal splicing event to eliminate the intron, resulting in an ecircRNA. Moreover, canonical splicing can generate the other type of circRNA, ciRNA, which is entirely composed of introns. CircRNAs might be imported to the cytoplasm, where they primarily function. Mostly, circRNAs possess the ability to bind to miRNAs to regulate gene expression. CircRNA was recently found to be a template for protein translation or to establish an interaction with proteins. It can act like a protein sponge, which might later facilitate or inhibit the interaction between two different proteins. Utilizing protein sponge activity, some proteins avoid ubiquitin-mediated degradation by binding to circRNAs. Remarkably, some circRNAs have also been discovered to regulate their parental genes through various mechanisms, such as directly binding to the promotor or recruiting proteins to mediate gene expression.
Ijms 25 06094 g001
Figure 2. Oncogenic circRNAs and their path of action through various cell cycle regulators in bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, endometrial cancer, esophageal cancer, gastric cancer, glioma, hepatocellular carcinoma, laryngeal cancer, leukemia, lung cancer, melanoma, multiple myeloma, nasopharyngeal carcinoma, oral squamous cell carcinoma, osteosarcoma, ovarian cancer, pancreatic ductal adenocarcinoma, and prostate cancer.
Figure 2. Oncogenic circRNAs and their path of action through various cell cycle regulators in bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, endometrial cancer, esophageal cancer, gastric cancer, glioma, hepatocellular carcinoma, laryngeal cancer, leukemia, lung cancer, melanoma, multiple myeloma, nasopharyngeal carcinoma, oral squamous cell carcinoma, osteosarcoma, ovarian cancer, pancreatic ductal adenocarcinoma, and prostate cancer.
Ijms 25 06094 g002
Figure 3. Tumor-suppressing circRNAs and their path of action through various cell cycle regulators in bladder cancer, breast cancer, gastric cancer, glioma, hepatocellular carcinoma, lung cancer, multiple myeloma, oral squamous cell carcinoma, pancreatic ductal adenocarcinoma, thyroid cancer, and urothelial carcinoma.
Figure 3. Tumor-suppressing circRNAs and their path of action through various cell cycle regulators in bladder cancer, breast cancer, gastric cancer, glioma, hepatocellular carcinoma, lung cancer, multiple myeloma, oral squamous cell carcinoma, pancreatic ductal adenocarcinoma, thyroid cancer, and urothelial carcinoma.
Ijms 25 06094 g003
Table 1. CircRNAs and their functions in modulating cell cycle regulators throughout different cancers.
Table 1. CircRNAs and their functions in modulating cell cycle regulators throughout different cancers.
CircRNAsFunctionTarget miRNATarget mRNATarget ProteinMolecular MechanismDiseaseRole and
Expression Status
Ref.
Hsa_circ_0071196
(circLRBA)
miRNA spongemiR-19b-3pCITCITInhibit miR-19b-3p to upregulate CIT, a positive regulator of the MAPK pathway, leading to an increase in CDK1 and cyclin D1 expressionBladder cancerOncogenic
&
Upregulated
[73]
Hsa_circ_0132246
(circRIMS1)
miRNA spongemiR-433-3pCCAR1CCAR1Inhibit miR-433-3p to upregulate CCAR1, a coactivator of β-catenin, leading to an increase in c-Myc expressionBladder cancerOncogenic
&
Upregulated
[74]
Hsa_circ_0058063
(circATIC)
miRNA spongemiR-1485-5pCDK6CDK6Upregulate CDK6 by inhibiting its repressor, miR-1485-5pBladder cancerOncogenic
&
Upregulated
[75]
Hsa_circ_0000615
(circZNF609)
miRNA spongemiR-1200CDC25BCDC25BUpregulate CDC25B by inhibiting its repressor, miR-1200Bladder cancerOncogenic
&
Upregulated
[76]
CircMYLKmiRNA spongemiR-34aCCND3Cyclin D3Upregulate CCND3 by inhibiting its repressor, miR-34aBladder cancerOncogenic
&
Upregulated
[77]
miRNA spongemiR-195CCND1Cyclin D1Upregulate cyclin D1 by inhibiting its repressor, miR-195Laryngeal carcinomaOncogenic
&
Upregulated
[78]
CircGLIS3miRNA spongemiR-1273fSKP1SKP1- Promote SKP1 expression by inhibiting its repressor, miR-1273f
- SKP1 positively regulates cyclin D1
Bladder cancerOncogenic
&
Upregulated
[79]
CircBCRC-3miRNA spongemiR-182-5pP27P27Upregulate P27 by inhibiting its repressor, miR-182-5pBladder cancerAnti-oncogenic
& Downregulated
[80]
CircITCHmiRNA spongemiR-17P21P21Upregulate P21 by inhibiting its repressor, miR-17Bladder cancerAnti-oncogenic
& Downregulated
[81]
miR-24PTENPTENInhibit miR-24 to upregulate PTEN, a negative regulator of the PI3K/Akt pathway
Hsa_circ_0006117
(circPTPRA)
Protein sponge--IGF2BP1Block IGF2BP1 from recognizing m6A-modified mRNAs, such as c-Myc, and promoting stability and translationBladder cancerAnti-oncogenic
& Downregulated
[82]
CircNR3C1miRNA spongemiR-27a-3pCCND1Cyclin D1Downregulate cyclin D1 by inhibiting its activator, miR-27a-3pBladder cancerAnti-oncogenic
& Downregulated
[83]
Hsa_circ_0091074 (circTCONS_00016926)miRNA spongemiR-1297TAZTAZInhibit miR-1297 to upregulate TAZ, a downstream effector of the Hippo pathway, leading to an increase in CDK4, CDK6, and cyclin D1 expressionBreast cancerOncogenic
& Downregulated
[84]
CircPGRmiRNA spongemiR-301a-5pCDK1,
CDK6,
CHEK2
CDK1,
CDK6,
CHEK2
Upregulate CDK1, CDK6, and CHEK2 by inhibiting their repressor, miR-301a-5pBreast cancerOncogenic
&
Upregulated
[85]
CircHMCUmiRNA spongeLet-7 familyc-Myc,
CCND1
c-Myc,
Cyclin D1
Upregulate c-Myc and cyclin D1 by inhibiting their repressor, let-7 familyBreast cancerOncogenic
&
Upregulated
[86]
CircUBE2D2miRNA spongemiR-512-3pCDCA3CDCA3Upregulate CDCA3 by inhibiting its repressor, miR-512-3pBreast cancerOncogenic
&
Upregulated
[87]
Hsa_circ_0136666
(circPRKDC)
miRNA spongemiR-1299CDK6CDK6Upregulate CDK6 by inhibiting its repressor, miR-1299Breast cancerOncogenic
&
Upregulated
[88]
miRNA spongemiR-198DDR1DDR1- Upregulate DDR1 by inhibiting its repressor, miR-198
- DDR1 positively regulates cyclin D1
Colorectal cancerOncogenic
&
Upregulated
[89]
Hsa_circ_0004676
(circDHTKD1)
miRNA spongemiR-377-3pE2F6E2F6- Upregulate E2F6 by inhibiting its repressor, miR-377-3p
- E2F6 stimulates PNO1 to further induces p53 ubiquitination
Breast cancerOncogenic
&
Upregulated
[90]
CircUBR1miRNA spongemiR-1299CCND1Cyclin D1Upregulate cyclin D1 by inhibiting its repressor, miR-1299Breast cancerOncogenic
&
Upregulated
[91]
Hsa_circ_0008039
(circPRKAR1B)
miRNA spongemiR-432-5pE2F3E2F3Upregulate E2F3 by inhibiting its repressor, miR-432-5pBreast cancerOncogenic
&
Upregulated
[92]
Hsa_circ_0000515
(circRPPH1)
miRNA spongemiR-296-5pCXCL10CXCL10- Upregulate CXCL10 by inhibiting its repressor, miR-296-5p
- CXCL10 positively regulates CDK4, and cyclin D1 and negatively regulates p27
Breast cancerOncogenic
&
Upregulated
[93]
Hsa_circ_0000520
(circRPPH1)
miRNA spongemiR-1296CDK2CDK2Upregulate CDK2 by inhibiting its repressor, miR-1296Cervical cancerOncogenic
&
Upregulated
[94]
Hsa_circ_0000512
(circRPPH1)
miRNA spongemiR-296-5pRUNX1RUNX1- Upregulate RUNX1 by inhibiting its repressor, miR-296-5p
- RUNX1 positively regulates cyclin D1
Colorectal cancerOncogenic
&
Upregulated
[95]
Hsa_circ_0000519
(circRPPH1)
miRNA spongemiR-1296E2F7E2F7Upregulate E2F7 by inhibiting its repressor, miR-1296Hepatocellular carcinomaOncogenic
&
Upregulated
[96]
Hsa_circ_0000517
(circRPPH1)
miRNA spongemiR-326SMAD6SMAD6- Upregulate SMAD6 by inhibiting its repressor, miR-326
- SMAD6 positively regulates cyclin D1
Hepatocellular carcinomaOncogenic
&
Upregulated
[97]
CircPLK1miRNA spongemiR-4500IGF1IGF1- Upregulate IGF1 by inhibiting its repressor, miR-4500
- IGF1 positively regulates CDK4, and CDK6
Breast cancerOncogenic
&
Upregulated
[98]
Hsa_circ_0001785
(circELP3)
miRNA spongemiR-942SOCS3SOCS3Inhibit miR-942 to upregulate SOCS3, a negative regulator of the JAK/STAT pathway, leading to a decrease in PCNA expressionBreast cancerAnti-oncogenic
& Downregulated
[99]
CircFBXW7Protein encoding--FBXW7-185aaBind to USP28 and inhibit function in activating c-MycBreast cancerAnti-oncogenic
& Downregulated
[100]
miRNA spongemiR-197-3pFBXW7FBXW7- Upregulate FBXW7 by inhibiting its repressor, miR-197-3p
- FBXW7 negatively regulates c-Myc
CircSETD2miRNA spongemiR-155-5pSCUBE2SCUBE2- Upregulate SCUBE2 by inhibiting its repressor, miR-155-5p
- SCUBE2 negatively regulates CDK4, and cyclin D1
Breast cancerAnti-oncogenic
& Downregulated
[101]
CircZFRProtein sponge--SSBP1Promote the formation of CDK2/cyclin E1 complex which later induces p-Rb and releases E2F1Cervical cancerOncogenic
&
Upregulated
[102]
miRNA spongemiR-147aCACUL1CACUL1- Upregulate CACUL1 by inhibiting its repressor, miR-147a
- CACUL1 positively regulates CDK2, CDK4, CDK6, and cyclin D1
Colorectal cancerOncogenic
&
Upregulated
[103]
Hsa_circ_0072088
(circZFR)
miRNA spongemiR-377-5pNOVA2NOVA2- Upregulate NOVA2 by inhibiting its repressor, miR-377-5p
- NOVA2 positively regulates cyclin D1, and PCNA
Lung cancerOncogenic
&
Upregulated
[104]
Hsa_circ_0000326 (circTCONS_l2_00004572)miRNA spongemiR-338-3pCDK4CDK4Upregulate CDK4 by inhibiting its repressor, miR-338-3pCervical cancerOncogenic
&
Upregulated
[105]
Hsa_circ_0000263 (circTCONS_00017720)miRNA spongemiR-150-5pMDM4MDM4- Upregulate MDM4 by inhibiting its repressor, miR-150-5p
- MDM4 negatively regulates p53
Cervical cancerOncogenic
&
Upregulated
[106]
Hsa_circ_0009035
(circRACGAP1)
miRNA spongemiR-889-3pHOXB7HOXB7- Upregulate HOXB7 by inhibiting its repressor, miR-889-3p
- HOXB7 positively regulates PCNA
Cervical cancerOncogenic
&
Upregulated
[107]
Hsa_circ_0003221
(circPTK2)
miRNA spongemiR-758-3pCPEB4CPEB4- Upregulate CPEB4 by inhibiting its repressor, miR-758-3p
- CPEB4 positively regulates cyclin D1 and PCNA
Cervical cancerOncogenic
&
Upregulated
[108]
CircESRP1miRNA spongemiR-634TPD52TPD52- Upregulate TPD52 by inhibiting its repressor, miR-634
- TPD52 positively regulates cyclin D1
Cervical cancerOncogenic
&
Upregulated
[109]
miR-142-3pARL2ARL2- Upregulate ARL2 by inhibiting its repressor, miR-142-3p
- ARL2 positively regulates cyclin D1 and PCNA
[110]
Hsa_circ_0000285
(circHIPK3)
miRNA spongemiR-197-3pELK1ELK1- Upregulate ELK1 by inhibiting its repressor, miR-197-3p
- ELK1 positively regulates cyclin D1 and PCNA
Cervical cancerOncogenic
&
Upregulated
[111]
CircASAP1miRNA spongemiR-338-3pRPP25RPP25- Upregulate RPP25 by inhibiting its repressor, miR-338-3p
- RPP25 positively regulates cyclin D1 and PCNA
Cervical cancerOncogenic
&
Upregulated
[112]
Hsa_circ_0008934
(circASAP1)
miRNA spongemiR-145-5pE2F3E2F3Upregulate E2F3 by inhibiting its repressor, miR-145-5pOsteosarcomaOncogenic
&
Upregulated
[113]
Hsa_circ_0008231
(circNBPF11)
miRNA spongemiR-338-3pETS1ETS1Inhibit miR-338-3p to upregulate ETS1, a positive regulator of the JAK/STAT pathway, leading to an increase in cyclin D1 expressionColon cancerOncogenic
&
Upregulated
[114]
CircCTIC1Protein sponge--NURFPromote nuclear translocation of NURF to bind to c-Myc promotor and increase c-Myc expressiomColon cancerOncogenic
&
Upregulated
[115]
CircCSPP1miRNA spongemiR-431ROCK1ROCK1- Upregulate ROCK1 by inhibiting its repressor, miR-431
- ROCK1 positively regulates CDK4, cyclin D1, and p-Rb
Colon cancerOncogenic
&
Upregulated
[116]
Hsa_circ_0005615
(circNFATC3)
miRNA spongemiR-149-5pTNKSTNKSInhibit miR-149-5p to upregulate TNKS, a positive regulator of the Wnt/β-catenin pathway, leading to an increase in cyclin D1 expressionColorectal cancerOncogenic
&
Upregulated
[117]
CircMETTL9miRNA spongemiR-551b-5pCDK6CDK6Upregulate CDK6 by inhibiting its repressor, miR-551b-5pColorectal cancerOncogenic
&
Upregulated
[118]
CircPRMT5miRNA spongemiR-377E2F3E2F3Upregulate E2F3 by inhibiting its repressor, miR-377Colorectal cancerOncogenic
&
Upregulated
[119]
Hsa_circ_0007142
(circDOCK1)
miRNA spongemiR-122-5pCDC25ACDC25AUpregulate CDC25A by inhibiting its repressor, miR-122-5pColorectal cancerOncogenic
&
Upregulated
[120]
CircCTNNA1miRNA spongemiR-149-5pFOXM1FOXM1- Upregulate FOXM1 by inhibiting its repressor, miR-149-5p
- FOXM1 is a transcription factor of cyclin B1, and cyclin D1
Colorectal cancerOncogenic
&
Upregulated
[121]
Hsa_circ_0002577
(circWDR26)
miRNA spongemiR-197CTNND1CTNND1Inhibit miR-197 to upregulate CTNND1, a positive regulator of the Wnt/β-catenin pathway, leading to an increase in cyclin D1 and c-Myc expressionEndometrial cancerOncogenic
&
Upregulated
[122]
Hsa_circ_0001610
(circTNFRSF21)
miRNA spongemiR-139-5pCCNB1Cyclin B1Upregulate cyclin B1 by inhibiting its repressor, miR-139-5pEndometrial cancerOncogenic
&
Upregulated
[123]
CircFIG4miRNA spongemiR-493-5pE2F3E2F3Upregulate E2F3 by inhibiting its repressor, miR-493-5pEsophageal cancerOncogenic
&
Upregulated
[124]
CircFNDC3BmiRNA spongemiR-214-3pCDC25ACDC25AUpregulate CDC25A by inhibiting its repressor, miR-214-3pEsophageal cancerOncogenic
&
Upregulated
[125]
Hsa_circ_00014879
(circDCAF8)
miRNA spongemiR-519-3pCDC25ACDC25AUpregulate CDC25A by inhibiting its repressor, miR-519-3pEsophageal cancerOncogenic
&
Upregulated
[126]
Hsa_circ_0003340
(circOGDH)
miRNA spongemiR-940PRKAA1PRKAA1- Upregulate PRKAA1 by inhibiting its repressor, miR-940
- PRKAA1 positively regulates cyclin D1, and PCNA
Esophageal cancerOncogenic
&
Upregulated
[127]
Hsa_circ_0120816
(circGFPT1)
miRNA spongemiR-1305TXNRD1TXNRD1- Upregulate TXNRD1 by inhibiting its repressor, miR-1305
- TXNRD1 positively regulates cyclin B1
Esophageal cancerOncogenic
&
Upregulated
[128]
CircPGPEP1miRNA spongemiR-1297E2F7E2F7Upregulate E2F7 by inhibiting its repressor, miR-1297Gastric cancerOncogenic
&
Upregulated
[129]
Hsa_circ_104433miRNA spongemiR-497-5pCDC25ACDC25AUpregulate CDC25A by inhibiting its repressor, miR-497-5pGastric cancerOncogenic
&
Upregulated
[130]
CircRNF111miRNA spongemiR-876-3pKLF12KLF12- Upregulate KLF2 by inhibiting its repressor, miR-876-3p
- KLF2 positively regulates cyclin D1
Gastric cancerOncogenic
&
Upregulated
[131]
Hsa_circ_0001982
(circRNF111)
miRNA spongemiR-1205E2F1E2F1Upregulate E2F1 by inhibiting its repressor, miR-1205GliomaOncogenic
&
Upregulated
[132]
Hsa_circ_0006470
(circMFN2)
miRNA spongemiR-1234TP53I11TP53I11Inhibit miR-1234 to upregulate TP53I11, a negative regulator of the PI3K/Akt pathway, leading to an increase in CDK2 expressionGastric cancerAnti-oncogenic and Downregulated[133]
CircEVI5miRNA spongemiR-4793-3pFOXO1FOXO1- Upregulate FOXO1 by inhibiting its repressor, miR-4793-3p
- FOXO1 positively regulates p21 and p27
Gastric canerAnti-oncogenic and Downregulated[134]
Hsa_circ_0001495
(circCCNB1)
Protein sponge--HuRPromote stabilization of CCND1 mRNAGliomaOncogenic
&
Upregulated
[135]
miRNA spongemiR-516b-5pCCND1Cyclin D1Upregulate cyclin D1 by inhibiting its repressor, miR-516b-5p
Hsa_circ_0012129
(circB4GALT2)
miRNA spongemiR-761TGIF2TGIF2- Upregulate TGIF2 by inhibiting its repressor, miR-761
- TGIF2 positively regulates CDK2, and CDK4
GliomaOncogenic
&
Upregulated
[136]
Hsa_circ_0030018
(circPOSTN)
miRNA spongemiR-1297RAB21RAB21- Upregulate RAB21 by inhibiting its repressor, miR-1297
- RAB21 positively regulates cyclin D1, and PCNA
GliomaOncogenic
&
Upregulated
[137]
CircCDR1asProtein sponge--p53Prevent p53 from interacting with its negative regulator, MDM2GliomaAnti-oncogenic
&
No expression status
[138]
Hsa_circ_0036412
(circETFA)
Protein sponge--ELAVL1Promote stabilization of GLI2, a Hedgehog regulator, to increase cyclin D1Hepatocellular carcinomaOncogenic
&
Upregulated
[139]
Hsa_circ_0091581
(circGPC3)
miRNA spongemiR-526bc-Mycc-MycUpregulate c-Myc by inhibiting its repressor, miR-526bHepatocellular carcinomaOncogenic
&
Upregulated
[140]
Hsa_circ_0091579
(circGPC3)
miRNA spongemiR-1287PDK2PDK2- Upregulate PDK2 by inhibiting its repressor, miR-1287
- PDK2 positively regulates cyclin D1
Hepatocellular carcinomaOncogenic
&
Upregulated
[141]
CircROBO1miRNA spongemiR-130a-5pCCNT2Cyclin T2Upregulate cyclin T2 by inhibiting its repressor, miR-130a-5pHepatocellular carcinomaOncogenic
&
Upregulated
[142]
CircMMP9miRNA spongemiR-149CCND2Cyclin D2Upregulate cyclin D2 by inhibiting its repressor, miR-149Hepatocellular carcinomaOncogenic
&
Upregulated
[143]
Hsa_circ_101555miRNA spongemiR-145-5pCDCA3CDCA3Upregulate CDCA3 by inhibiting its repressor, miR-145-5pHepatocellular carcinomaOncogenic
&
Upregulated
[144]
CircZNF83miRNA spongemiR-324-5pCDK16CDK16Upregulate CDK16 by inhibiting its repressor, miR-324-5pHepatocellular carcinomaOncogenic
&
Upregulated
[145]
CircSLC7A11miRNA spongemiR-330-3pCDK1CDK1Upregulate CDK1 by inhibiting its repressor, miR-330-3pHepatocellular carcinomaOncogenic
&
Upregulated
[146]
Hsa_circ_0005397
(circRHOT1)
miRNA spongemiR-326PDK2PDK2- Upregulate PDK2 by inhibiting its repressor, miR-326
- PDK2 positively regulates cyclin D1 and negatively regulates p21
Hepatocellular carcinomaOncogenic
&
Upregulated
[147]
CircVAMP3Protein sponge--CAPRIN1Block CAPRIN1 from interacting with G3BP1 to decrease c-MycHepatocellular carcinomaAnti-oncogenic
& Downregulated (in tissue)
[148]
CircLARP4miRNA spongemiR-761RUNX3RUNX3- Upregulate RUNX3 by inhibiting its repressor, miR-761
- RUNX3 positively regulates p53, and p21
Hepatocellular carcinomaAnti-oncogenic
& Downregulated
[149]
Hsa_circ_ 0000204
(circWDR37)
miRNA spongemiR-191KLF6KLF6- Upregulate KLF6 by inhibiting its repressor, miR-761
- KLF6 negatively regulates cyclin D1, cyclin D2, and c-Myc
Hepatocellular carcinomaAnti-oncogenic
& Downregulated (in tissue)
[150]
F-circBA1miRNA spongemiR-148b-3pCDC25BCDC25BUpregulate CDC25B by inhibiting its repressor, miR-148b-3pLeukemiaOncogenic
&
No Expression Status
[28]
Hsa_circ_0005774
(circCDK1)
miRNA spongemiR-192-5pULK1ULK1- Upregulate ULK1 by inhibiting its repressor, miR-192-5p
- ULK1 positively regulates cyclin D1, and PCNA
LeukemiaOncogenic
&
Upregulated
[151]
Hsa_circ_0094100
(circSFMBT2)
miRNA spongemiR-217ATP1B1ATP1B1- Upregulate ATP1B1 by inhibiting its repressor, miR-217
- ATP1B1 positively regulates cyclin D1, and PCNA
LeukemiaOncogenic
&
Upregulated
[152]
CircNFIXmiRNA spongemiR-876-3pTRIM31TRIM31- Upregulate TRIM31 by inhibiting its repressor, miR-876-3p
- TRIM31 positively regulates cyclin D1
LeukemiaOncogenic
&
Upregulated
[153]
Hsa_circ_0000190
(circCNIH4)
miRNA spongemiR-142-5pCDK6CDK6- Upregulate CDK6 by inhibiting its repressor, miR-142-5p
- Promote MAP K signaling to increase CDK1, CDK4, and p-Rb
Lung cancerOncogenic
&
Upregulated
[154]
Hsa_circ_0014235
(circS100A2)
miRNA spongemiR-146b-5pYAPYAPInhibit miR-146b-5p to upregulate YAP, a downstream effector of the Hippo pathway, leading an increase in CDK4 expressionLung cancerOncogenic
&
Upregulated
[155]
miRNA spongemiR-520a-5pCDK4CDK4Upregulate CDK4 by inhibiting its repressor, miR-520a-5p[156]
CircDENND2AmiRNA spongemiR-34aCCNE1Cyclin E1Upregulate cyclin E1 by inhibiting its repressor, miR-34aLung cancerOncogenic
&
Upregulated (in tissue)
[157]
Hsa_circ_0062389
(circPI4KA)
miRNA spongemiR-103a-3pCCNE1Cyclin E1Upregulate cyclin E1 by inhibiting its repressor, miR-103a-3pLung cancerOncogenic
&
Upregulated
[158]
Hsa_circ_0012384
(circCMPK1)
miRNA spongemiR-302eCCND1Cyclin D1Upregulate cyclin D1 by inhibiting its repressor, miR-302eLung cancerOncogenic
&
Upregulated
[159]
CircABCB10miRNA spongemiR-584-5pE2F5E2F5Upregulate E2F5 by inhibiting its repressor, miR-584-5pLung cancerOncogenic
&
Upregulated
[160]
Hsa_circ_0001421
(circSEC31A)
miRNA spongemiR-4677-3pCDCA3CDCA3Upregulate CDCA3 by inhibiting its repressor, miR-4677-3pLung cancerOncogenic
&
Upregulated
[161]
CircSATB2miRNA spongemiR-33a-5pE2F7E2F7Upregulate E2F7 by inhibiting its repressor, miR-33a-5pLung cancerOncogenic
&
Upregulated
[162]
CircPITX1miRNA spongemiR-1248CCND2Cyclin D2Upregulate cyclin D2 by inhibiting its repressor, miR-1248Lung cancerOncogenic
&
Upregulated
[163]
Hsa_circ_0006692 (circTCONS_00025531)miRNA spongemiR-205-5pCDK19CDK19Upregulate cyclin D19 by inhibiting its repressor, miR-205-5pLung cancerOncogenic
&
Upregulated
[164]
CircTP63miRNA spongemiR-873-3pFOXM1FOXM1- Upregulate FOXM1 by inhibiting its repressor, miR-873-3p
- FOXM1 positively regulates cyclin B1
Lung cancerOncogenic
&
Upregulated (in tissue)
[165]
CircFOXM1miRNA spongemiR-614FAM83DFAM83D- Upregulate FAM83D by inhibiting its repressor, miR-614
- FAM83D positively regulates cyclin D1, and cyclin E1
Lung cancerOncogenic
&
Upregulated
[166]
Hsa_circ_0046263
(circP4HB)
miRNA spongemiR-940NOVA2NOVA2- Upregulate NOVA2 by inhibiting its repressor, miR-940
- NOVA2 positively regulates cyclin D1, and PCNA
Lung cancerOncogenic
&
Upregulated
[167]
Hsa_circ_0065214
(circSCAP)
Protein sponge--SF3A3- Induce SF3A3 degradation to increase MDM4-S
- MDM4-S positively regulates p53
Lung cancerAnti-oncogenic and Downregulated[168]
CircNOL10Protein sponge--SCML1- Prevent SCML1, a transcription factor, from ubiquitination to positively regulates HN2, and HN8
- HN2 and HN8 negatively regulate CHEK2, CDC25A, and E2F1 and positively regulate p53
Lung cancerAnti-oncogenic and Downregulated[169]
CircFBXO7miRNA spongemiR-296-3pKLF15KLF15- Upregulate KLF15 by inhibiting its repressor, miR-296-3p
- KLF15 positively regulates p21
Lung cancerAnti-oncogenic and Downregulated[170]
Hsa_circ_0082835
(circEZH2)
miRNA spongemiR-429EZH2EZH2Inhibit miR-429 to upregulate EZH2, a positive regulator of the Wnt/β-catenin pathway, leading to an increase in cyclin D1 and cyclin E1 expressionMelanomaOncogenic
&
No expression status
[171]
Hsa_circ_0062270
(circCDC45)
Protein sponge--EIF4A3Promote CDC45 mRNA stabilizationMelanomaOncogenic
&
Upregulated
[172]
CircPSAPmiRNA spongemiR-331-3pHDAC4HDAC4- Upregulate HDAC4 by inhibiting its repressor, miR-331-3p
- HDAC4 positively regulates cyclin D1
Multiple myelomaOncogenic
&
Upregulated
[173]
CircMYBL2Protein sponge--Cyclin F,
MYBL2
- Facilitate the binding between cyclin F and MYBL2
- Block MYBL2 from activating oncogenes
Multiple myelomaAnti-oncogenic and Downregulated[174]
Hsa_circ_0000745
(circSPECC1)
Protein sponge--HuRPromote CCND1 mRNA stabilizationOral squamous cell carcinomaOncogenic
&
Upregulated
[175]
miRNA spongemiR-488CCND1Cyclin D1Upregulate cyclin D1 by inhibiting its repressor, miR-488
CircOSBPL10miRNA spongemiR-299-3pCDK6CDK6Upregulate CDK6 by inhibiting its repressor, miR-299-3pOral squamous cell carcinomaOncogenic
& Downregulated
[176]
CircYAPProtein sponge--CDK4Block CDK4 from establishing cyclin D1–CDK4 complexOral squamous cell carcinomaAnti-oncogenic and Downregulated[177]
CircARF3miRNA spongemiR-1299CDK6CDK6Upregulate CDK6 by inhibiting its repressor, miR-1299OsteosarcomaOncogenic
&
Upregulated
[178]
Hsa_circ_0000073
(circOMA1)
miRNA spongemiR-1252-5pCCNE2Cyclin E2Upregulate cyclin E2 by inhibiting its repressor, miR-1252-5pOsteosarcomaOncogenic
&
Upregulated
[179]
Hsa_circ_0084582
(circCHD7)
miRNA spongemiR-485-5pJAG1JAG1- Upregulate JAG1 by inhibiting its repressor, miR-485-5p
- JAG1 positively regulates c-Myc
OsteosarcomaOncogenic
&
Upregulated
[180]
CircSAMD4AmiRNA spongemiR-218-5pKLF8KLF8- Upregulate KLF8 by inhibiting its repressor, miR-218-5p
- KLF8 positively regulates cyclin D1 and negatively regulates p21
OsteosarcomaOncogenic
&
Upregulated
[181]
CircE2F2Protein sponge--HuRPromote E2F2 mRNA stabilizationOvarian cancerOncogenic
&
Upregulated
[182]
Hsa_circ_0000714
(circAARS)
miRNA spongemiR-370-3pRAB17RAB17- Upregulate RAB17 by inhibiting its repressor, miR-370-3p
- RAB17 positively regulates CDK6, and Rb
Ovarian cancerOncogenic
&
Upregulated (in paclitaxel resistance)
[183]
CircNEIL3miRNA spongemiR-432-5pADAR1ADAR1- Upregulate ADAR1 by inhibiting its repressor, miR-432-5p
- ADAR1 positively regulates GLI1, a downstream regulator in the Hedgehog pathway
- GLI1 positively regulates CDK2, CDK4, CDK6, cyclin D1, and cyclin E1
Pancreatic ductal adenocarcinomaOncogenic
&
Upregulated
[184]
CircPVRL3miRNA spongemiR-194-5pSOCS2SOCS2Inhibit miR-194-5p to upregulate SOCS2, a negative regulator of the PI3K/Akt pathway, leading to a decrease in CDK2, CDK4, cyclin D1, and cyclin E1 expression and an increase in p21 expressionPancreatic ductal adenocarcinomaAnti-oncogenic and Downregulated[185]
Hsa_circ_0057558
(circSLC39A10)
miR-206USP33USP33- Upregulate USP33 by inhibiting its repressor, miR-206
- USP33 positively regulates cyclin B1, cyclin D1, and c-Myc
Prostate cancerOncogenic
&
Upregulated
[186]
CircHMGCS1miRNA spongemiR-205-5pErBB3ErBB3Inhibit miR-205-5p to upregulate ErBB3, a positive regulator of the PI3K/Akt pathway, leading to an increase in cyclin D1 and CDK4 expressionProstate cancerOncogenic
&
Upregulated
[187]
CircDPP4miRNA spongemiR-195CCND1Cyclin D1Upregulate cyclin D1 by inhibiting its repressor, miR-195Prostate cancerOncogenic
&
Upregulated
[188]
CircEIF3IProtein sponge--AUF1Prevent AUF1 from inducing CCND1 mRNA degradationThyroid cancerOncogenic
&
Upregulated
[189]
CircPRKCImiRNA spongemiR-335E2F3E2F3Upregulate E2F3 by inhibiting its repressor, miR-335Thyroid cancerOncogenic
&
Upregulated
[190]
Hsa_circ_0000644
(circKIAA1199)
miRNA spongemiR-1205E2F3E2F3Upregulate E2F3 by inhibiting its repressor, miR-1205Thyroid cancerOncogenic
&
Upregulated
[191]
CircHACE1miRNA spongemiR-346TFC2L1TFC2L1- Upregulate TFC2L1 by inhibiting its repressor, miR-346
- TFC2L1 negatively regulates cyclin B1, and PCNA
Thyroid cancerAnti-oncogenic and Downregulated[192]
CircCAMSAP1mRNA sponge-SERPINH1-Promote SERPINH1 stabilization to further increase c-Myc translationNasopharyngeal carcinomaOncogenic
&
Upregulated
[193]
CircFAM114A2miRNA spongemiR-222-3pP27P27Upregulate p27 by inhibiting its repressor, miR-222-3pUrothelial carcinomaAnti-oncogenic and Downregulated[194]
miR-146a-5pP21P21Upregulate p21 by inhibiting its repressor, miR-146a-5p
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

Thamjamrassri, P.; Ariyachet, C. Circular RNAs in Cell Cycle Regulation of Cancers. Int. J. Mol. Sci. 2024, 25, 6094. https://doi.org/10.3390/ijms25116094

AMA Style

Thamjamrassri P, Ariyachet C. Circular RNAs in Cell Cycle Regulation of Cancers. International Journal of Molecular Sciences. 2024; 25(11):6094. https://doi.org/10.3390/ijms25116094

Chicago/Turabian Style

Thamjamrassri, Pannathon, and Chaiyaboot Ariyachet. 2024. "Circular RNAs in Cell Cycle Regulation of Cancers" International Journal of Molecular Sciences 25, no. 11: 6094. https://doi.org/10.3390/ijms25116094

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