Next Article in Journal
MicroRNA Biogenesis, Gene Regulation Mechanisms, and Availability in Foods
Previous Article in Journal
Non-Coding RNA as a Biomarker in Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interplay of microRNAs and circRNAs in Epithelial Ovarian Cancer

by
Heidi Schwarzenbach
Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
Non-Coding RNA 2024, 10(5), 51; https://doi.org/10.3390/ncrna10050051
Submission received: 20 August 2024 / Revised: 23 September 2024 / Accepted: 28 September 2024 / Published: 30 September 2024

Abstract

:
Epithelial ovarian cancer (EOC) with its high death incidence rate is generally detected at advanced stages. During its progression, EOC often develops peritoneal metastasis aggravating the outcomes of EOC patients. Studies on non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and circular RNAs (circRNAs), have analyzed the impact of miRNAs and circRNAs, along with their interaction among each other, on cancer cells. MiRNAs can act as oncogenes or tumor suppressors modulating post-transcriptional gene expression. There is accumulating evidence that circRNAs apply their stable, covalently closed, continuous circular structures to competitively inhibit miRNA function, and so act as competing endogenous RNAs (ceRNAs). This interplay between both ncRNAs participates in the malignity of a variety of cancer types, including EOC. In the current review, I describe the characteristics of miRNAs and circRNAs, and discuss their interplay with each other in the development, progression, and drug resistance of EOC. Sponging of miRNAs by circRNAs may be used as a biomarker and therapeutic target in EOC.

1. Introduction

Epithelial ovarian cancer (EOC) is the predominant ovarian cancer subtype and accounts for more than 95% of cases. The other non-epithelial subtypes include germ cell and sex-cord stromal cancer types, rare, small cell carcinoma, and ovarian sarcoma. EOC is histologically grouped into the following five major subtypes: high-grade serous, low-grade serous, clear cell, endometrioid, and mucinous ovarian cancer [1]. In 1973, the International Federation of Gynecology and Obstetrics (FIGO) staging system was introduced and revised in 1988 and 2013. There are four stages: FIGO I and II involve the restriction of the tumor to the ovaries and the tumor spread in the pelvis, respectively. FIGO III leads to the tumor spread outside the pelvis and its subtype FIGO IIIC defines the metastatic involvement of retroperitoneal lymph nodes, while FIGO IV covers distant metastases [2]. The high death incidence rate of EOC is due to asymptomatic features leading to the late diagnosis of the majority of patients. About three-fourths of FIGO III/IV patients have relatively poor survival prospects. Currently, the relative 5-year survival is 44%. In contrast, early-stage disease is treatable in nearly 90% of women, even in patients with more aggressive histologic subtypes. In this respect, the American Cancer Society estimates the number of cases with a new diagnosis of ovarian cancer of about 19,680 women in the United States for 2024, while 12,740 women will die from ovarian cancer (URL: https://www.cancer.org/cancer/types/ovarian-cancer.html, accessed on 27 September 2024).
EOC progression is accompanied with epithelial–mesenchymal transition (EMT) in which epithelial cells acquire fibroblast-like mesenchymal features and cellular plasticity resulting in the loss of their cell polarity, and adhesion to adjacent cells and the basement membrane. In turn, these phenotypically changed cells migrate through the extracellular matrix and settle down in other organs. As multicellular spheroids, they invade either locally to adjacent tissues or to pelvic and distal organs within the peritoneal fluid or ascites [3].
Therapy decisions are based on disease stage, histology, and biology, and comprise surgery and chemotherapy. Chemotherapy usually applies the combination of two different types of drugs, and includes platinum compounds, such as cisplatin or carboplatin, and taxanes, such as paclitaxel or docetaxel. Moreover, the administration of inhibitors against poly (ADP-ribose) polymerase molecules (PARP), which share in the DNA damage-repair process, have led to treatment success in some recurrent patients [4].
So far, numerous investigations have shown that non-coding RNAs (ncRNAs) participate in different stages of cancer, namely initiation, progression, and metastasis. In particular, abundant studies on microRNAs (miRNAs) have been published and demonstrated the impact of miRNAs in EOC along with its treatment [5]. MiRNAs can modulate post-transcriptionally the gene expression, especially by their inhibitory activity through binding to the 3′ untranslational region (UTR) [6], leading to the repression of translation. However, they can also activate the transcription through binding to promoter or enhancer regions [7]. Their dissemination is ensured by the apoptotic and necrotic cells that release them into the blood circulation or other body fluids (called liquid biopsy) [8], where they exist either cell-free, or associated with Argonaut (AGO) proteins [9]. MiRNAs may also be actively secreted in exosomes, small extracellular vesicles, into the blood [10] which transfer them from cell to cell, to modulate the characteristics of the recipient cells. Hence, these exosomal miRNAs play an important role in cell-to-cell communication, to induce tumor progression and spread [11,12]. Circular RNAs (circRNAs) are also single-stranded ncRNAs but are created as a circular conformation via non-canonical splicing or back-splicing events [13]. They form an important crosstalk network with miRNAs and mRNAs, regulating a variety of cellular events including cell division, signaling pathways, cell mobility, and metastasis [14]. The majority of circRNAs have been detected in the cytoplasm where they act as competing endogenous RNAs (ceRNAs), leading to the inhibition of miRNA expression.
The current review article deals with the regulatory network of miRNAs and circRNAs and their interplay between each other. Their impact on EOC development, and progression and possible therapy approaches using these ncRNAs are discussed.

2. ncRNAs

Approximately 80% of the genome is transcribed to ncRNAs [15]. They constitute subgroups of miRNAs, circRNAs, lncRNAs (long non-coding RNAs), siRNAs (small interfering RNAs), and piRNAs (PIWI-interacting RNAs). These ncRNAs mainly act as silencers by inhibiting translation through binding to their mRNA targets. They play an essential role in various stages of cancer, namely initiation, progression, and metastasis [16]. In particular, their roles in various stages of EOC and their potential use in therapies have been reported [17]. Their ability to interact with each other allows the sequestered ncRNAs to no longer bind to their mRNA target, leading to an undisturbed translation of the mRNAs performed by the ribosomes. The ceRNA activity results in the formation of a large-scale RNA regulatory network across the transcriptome, and is involved in numerous processes, such as DNA methylation, apoptosis, DNA damage repair, EMT, and multidrug resistance (MDR). It is dependent on the concentration and character of the ncRNAs; therefore, it is difficult to predict which interactions between ncRNAs take place and what impact they have on the signaling network and the cellular behavior. Thus, such a regulatory network may imply the formation of an altered cellular protein expression profile that should also be considered in pathological conditions [18].

3. Biogenesis and Characteristics of miRNAs

MiRNAs are a family of evolutionary conserved, small ncRNA molecules which, as mentioned above, post-transcriptionally inhibit the mRNA translation by binding to the 3′ UTR [6]. Remarkably, miRNAs can also activate mRNA translation by binding different target sites, including the DNA promoter [19].
Similar to protein-coding mRNAs, the 21–23 nucleotide-long miRNAs are typically generated by splicing, capping, and polyadenylating [20]. In the nucleus, RNA polymerase II transcribes the long primary miRNAs (pri-miRNAs) that are in turn cleaved by the endonuclease Drosha which associates with the RNA-binding protein DiGeorge syndrome critical region 8 (DGCR8), to generate the 60–70-nucleotide stem-loop precursor miRNAs (pre-miRNAs) [21]. In a Ran-GTPase dependent fashion, the pre-miRNAs are then transferred from the nucleus to the cytoplasm by exportin 5. The hairpin-like structure is further processed by DICER1 in association with the RNA-binding protein transactivation-responsive RNA-binding protein (TRBP) to produce the mature duplex miRNAs. One of the strands of the mature miRNAs is loaded into the miRNA-induced silencing complex (miRISC), containing DICER1 and AGO proteins [22], whilst the other (star) strand is usually degraded. However, occasionally, both strands can be integrated into RISC. Here, the interaction of miRNAs with mRNAs takes place by sequence complementary binding, inhibiting the translation into protein or degrading the mRNA. In addition, miRNAs may also be produced by alternative routes on which mirtrons bypass the Drosha processing step, whereby the premiRNA is produced by a splicing reaction. The subsequent steps within RISC are similar (Figure 1) [23].
In cancer, the expression of miRNAs is deregulated, either up- or downregulated, leading to aberrant signaling pathways. These modulated levels are associated with tumorigenesis, tumor progression, metastasis, and drug resistance. In this respect, miRNAs can act as oncogenes or tumor suppressors [24]. So far, numerous miRNAs have been reported in ovarian cancer [25,26]. For example, miR-21 inhibits PTEN expression, regulates PI3K/AKT signaling, reduces cell apoptosis, and increases cell proliferation in EOC [27]. The expression of miR-214 and miR-150 is abnormally upregulated in EOC and these miRNAs can also inhibit PTEN, downregulate the PTEN protein, and initiate the protein kinase B (AKT) pathway [28]. MiR-135a, miR-200c, miR-216a, and miR-340 regulate EMT and thus, modulate the invasiveness of EOC cells [29]. Elevated levels of miR-130a have been shown to circulate in the serum of early recurrence patients before elevated levels of serum marker CA125 are detected. The deregulation of miR-130a, which regulates the multidrug resistance 1 (MDR1) and PTEN gene expression, correlates with the development of cisplatin resistance [30]. The miR-200 family is associated with EMT and tumor angiogenesis [25,26]. The levels of exosomal miR-200b and miR-200c can distinguish between malignant and benign ovarian tumors and their increased levels are mainly observed in advanced EOC [31]. The exosomal levels of miR-200b correlate with patient overall survival, and influence cell proliferation and apoptosis [32]. In contrast, miR-340 and miR-377 act as tumor suppressors. They decrease EOC cell migration and invasion, downregulate the Wnt/β-catenin pathway, and inhibit EMT [33,34].

4. Biogenesis and Characteristics of circRNA

More than 100,000 circRNAs have been identified in human transcriptomes. They form covalently closed loop structures with no exposed 3′ and 5′ ends. Their stable ring structure prevents exonuclease-mediated degradation, so that their half-life of circRNAs of more than 48 h is much longer than that of linear RNAs with 10 h [35].
CircRNAs are classified in three subtypes, exonic circRNAs (ecRNAs), exon-intron circRNAs (EIciRNAs), and circular intronic RNAs (ciRNAs) [13]. Four classical models describe the formation of circRNAs [36], as follows: (1) In the lariat-driven circularization model, the pre-mRNA is folded in such a way that nonadjacent exons are in close proximity to each other, initiating exon-skipping and back-splicing. Additional splicing by removing introns of the formed EIciRNAs leads to ecRNAs [37]. (2) In the intron-pairing-driven circularization model, the base pairing of long flanking complementary introns, such as Alu elements, induces back-splicing in which a downstream 5′ splice site is joined and ligated with an upstream 3′ splice site by a 3′-5′ phosphodiester bond at the junction site. (3) In the RNA-binding protein (RBP)-driven circularization model, RBPs bind to flanking introns on both sides of an exon and unite them for back-splicing. (4) In the ciRNA biogenesis, intron lariats are degraded by debranching enzymes, but a particular pre-mRNA structure, consisting of the GU- and C-rich elements in the 5′ and 3′-end, allows introns to escape debranching and become ciRNAs [37] (Figure 1).
CircRNAs are usually expressed at low levels. However, one gene can produce multiple circRNAs. Because of their circle formation, they are more resistant to exonucleases than linear RNAs. As miRNAs, they circulate in the bloodstream and can be transported by exosomes [38]. They either promote gene transcription by binding to DNA polymerase II or inhibit mRNA translation. However, the majority of circRNAs regulate the gene expression by sponging miRNAs, serving as ceRNAs. Thus, circRNAs inhibit miRNA activity by competing binding sites [14,39]. They can also directly interact with circRNA-binding proteins; hence, they can regulate the translocation of certain proteins [40].
In the following paragraphs, circRNAs are described as a sponge for miRNAs in ovarian cancer.

5. circRNAs Serve as Sponge for miRNAs

To date, many research articles on the sponging of miRNAs by circRNAs have been published (Table 1). The interactions of circRNAs with miRNAs regulate cellular functions and control the development of EOC through their involvement with different signaling pathways. The competition of circRNAs with miRNAs allows the activation or repression of the downstream components of the signaling pathways (Figure 2) [41,42,43,44].
Thus, circRNAs sponge miRNAs; therefore, they abrogate the binding of miRNAs to their targets in important cancer-associated signaling pathways leading to tumor progression.
In the following, circRNAs which sponge one or several miRNAs, as listed in Table 1, have been arbitrarily selected and some of them have been considered and described in more detail with a focus on circRNAs which have been reported to interact with several miRNAs.
Cisplatin, a cytostatic drug which inhibits DNA replication, is usually applied in the first-line treatment of EOC. In their study, Gao et al. [123] investigated the interactions among circ_000784, miR-532-5p, and NFIB by a dual-luciferase reporter assay and determined the effect of circ_0007841 on cisplatin in a xenograft mouse model. They showed that circ_0007841 conferred cisplatin resistance through the miR-532-5p/NFIB axis. In order to promote cisplatin resistance, circ_0007841 acted as a sponge for miR-532, leading to the upregulation of NFIB, a member of nuclear factor I (NFI) family which stimulated DNA replication [143]. Huang et al. [118] demonstrated that circ_0007841 also acted as a ceRNA for another miRNA, namely miR-151-3p, to stimulate the expression of the RNA-binding protein MEX3C, leading to cell proliferation, migration, and invasion of EOC cells.
In EOC, plexin B2 (PLXNB2) expression is usually upregulated, and the silencing of PLXNB2 inhibits cell proliferation and invasion. As demonstrated by Liang et al. [96], circ_0013958 acted as a sponge for miR-637 to regulate the expression of PLXNB2. In EOC tissues and cells, upregulated circ_0013958 downregulated miR-637 resulting in the activation of PLXNB2. The knockdown of circ_0013958 impeded EOC development through modulating the miR-637/PLXNB2 axis.
So far, circ_0025033 has been found to be a sponge for miR-370, miR-184, and miR-532. Ma et al. [119] reported that circ_0025033 was upregulated in EOC. Its knockdown blocked tumor growth in vivo. Circ_0025033 affected the solute carrier family member SLC1A5, a mitochondrial glutamine transporter for metabolic reprogramming in cancer cells [144], via sponging miR-370-3p. In this process, SLC1A5 abolished the anti-ovarian cancer part of miR-370-3p and reduced glutamine metabolism. Xenograft models were established by Hou et al. [57] to determine the role of circ_0025033 in vivo. The knockdown of circ_0025033 or the U6 snRNA-associated Sm-like protein LSM4 blocked the ability of colony formation, migration, invasion, and glycolysis metabolism in EOC cells. In this context, circ_0025033 promoted the progression of EOC cancer by activating the expression of LSM4 via targeting miR-184. The interaction between miR-532-3p and circ_0025033 or forkhead box protein M1 (FOXM1), a critical proliferation-associated transcription factor [145], was examined using a pull-down assay and a dual-luciferase reporter assay by Huang et al. [131]. They detected that circ_0025033 upregulated the expression of FOXM1 by sponging miR-532-3p. Knockdown of exosomes containing circ_0025033 and derived from paclitaxel-resistant cells impaired the resistance of this cytostatic drug in recipient EOC cells.
Circ_0061140 has been shown to sponge miR-136, miR-361, and miR-761. Zhu et al. [132] investigated the effects of circ_0061140 on tumor formation and paclitaxel sensitivity in vivo by a tumor formation assay. Knockdown of circ_0061140 inhibited cell proliferation, migration, and invasion, and promoted cell apoptosis and paclitaxel sensitivity via sponging miR-136. An in vivo model was also conducted by Zhang et al. [58] using a xenograft mouse. Circ_0061140 facilitated tumorigenesis in vivo through inhibiting miR-361 to upregulate the expression of RAB1A, a small GTPase known for its role in vesicular trafficking [146]. In addition, Ma et al. [59] found that circ_0061140 expression was upregulated in EOC tissues and cell lines. Knockdown of circ_0061140 significantly suppressed the proliferation, migration, invasion, and angiogenesis. The oncogenic behavior of circ_0061140 referred to its ability to upregulate the expression of a mitochondrial inner membrane protein LETM1 by sponging miR-761.
Circ_0078607 has been shown to be a ceRNA for miR-32, miR-196, and miR-518a and to usually act as a tumor suppressor gene. In their study, Jin et al. [56] detected that circ_0078607 and salt inducible kinase 1 (SIK1) were downregulated in EOC tissues and cells. Overexpressed circ_0078607 suppressed EOC cell proliferation, migration, invasion, and promoted apoptosis by sponging miR-32-5p leading to the upregulation of SIK1, a target of miR-32-5p. The inhibitory effect of circ_0078607 on EOC progression could be reversed by the silencing of SIK1, a protein kinase which targets major plasma membrane transporters, such as the Na(+)/K(+)-ATPase and Na(+)/H(+) exchangers [147]. Furthermore, Dai et al. [122] showed that the overexpression of circ_0078607 inhibited cisplatin resistance in nude mice by sequestering miR-196b-5p to upregulate GAS7, while Zhang et al. [60] confirmed that circ_0078607 was downregulated in EOC. Bioinformatics and luciferase reporter analysis identified miR-518a-5p as a target of circ_0078607, while Fas is a target of miR-518a-5p. Thus, circ_0078607 suppressed EOC progression by sponging oncogenic miR-518a-5p to induce expression of the cell death molecule Fas which triggered apoptosis [148].
Circ_CELSR1 has been shown to interact with miR-149, miR-598, and miR-1252. Using paclitaxel-resistant EOC cells and tissues, Wei et al. [134] revealed that, the levels of circ_CELSR1 were upregulated, and its knockdown increased paclitaxel sensitivity and cell apoptosis, whereas inhibiting cell viability, colony formation and cell cycle process of resistant EOC cells. In a murine xenograft model assay, circ_CELSR1 silencing impeded paclitaxel resistance by regulating the miR-149-5p/SIK2 axis. Accordingly, circ_CELSR1 positively modulated SIK2 expression via sponging miR-149-5p. The increased SIK2 expression resulted in EOC progression, and might permit a treatment approach through regulating cellular metabolism, comprising glucose and lipid metabolism [149]. In addition, Zhang et al. [135] found that the paclitaxel resistance of EOC by circ_CELSR1 can also occur by regulating the expression of the protooncogene FOXR2 via miR-1252. Furthermore, Zeng et al. [107] used an abdominal cavity metastasis nude mouse model to analyze the in vivo function of circ_CELSR1. They showed that knockdown of circ_CELSR1 suppressed proliferation, migration, invasion, and EMT, but stimulated apoptosis in EOC cells, and also suppressed EOC growth and metastasis in nude mice. These effects were reversed by the inhibition of miR-598 or overexpression of BRD4. BRD4 is a member of the bromodomain and extra-terminal (BET) protein family and plays a role in super-enhancer organization and oncogene expression regulation [40].
Finally, in EOC tissues, circ_MYLK levels are significantly higher than those in adjacent tissues and its expression is remarkably associated with pathological staging and poor prognosis in EOC patients. Accordingly, circ_MYLK may promote the malignant progression of EOC through the downregulation of miR-652 [86].

6. Sponging of a miRNA by Several circRNAs

As described above, circRNAs can sponge several miRNAs, whereas conversely, several circRNAs can interact with one miRNA. In the following, single miRNAs which are sponged by several circRNAs, as listed in Table 2, are arbitrarily selected and some of them considered and described in more detail.
MiR-145 has been exposed to be a target for circ_0009910, circ_0015756, circ_ITCH, circ_VPS13C, and circ_WHSC1. In their study, Li et al. [52] confirmed that miR-145 was bound by circ_0009910 which negatively regulated miR-145. MiR-145 reversed the biological function of circ_0009910 in proliferative and motile phenotypes, and active status of the proinflammatory signaling pathway of NF-κB [150] and the pathway of Notch which is involved in angiogenesis, stem cell maintenance, and EMT [151]. Using nude mice, Pan et al. [53] showed that circ_0015756 which was highly expressed in EOC cells and promoted the tumor growth via the miR-145-5p/PSAT1 axis. In mechanical analysis, circ_0015756 directly bound to miR-145-5p which targeted the phosphoserine aminotransferase PSAT1, accelerating EOC tumorigenesis. Moreover, Lu et al. [101] demonstrated that the treatment of EOC cells with the anesthetic substance propofol suppressed the cell viability, cycle, and motility whereas elevating the apoptosis rate, and upregulated miR-145 in a dose-dependent manner. The anti-tumor role of propofol is partly owed to the upregulation of miR-145 which is a direct target of circ_VPS13C. Thus, propofol suppressed the progression of EOC cancer through the upregulation of miR-145 via suppressing circ_VPS13C. Zong et al. [82] showed that circ_WHSC1 was upregulated in EOC tissues, and increased cell proliferation, migration, and invasion, whereas it inhibited cell apoptosis. It sponged miR-145, leading to the upregulation of the expression of downstream targets, namely the transmembrane glycoprotein mucin 1 and the telomerase reverse transcriptase hTERT. Exosomes transferred circ_WHSC1 to peritoneal mesothelial cells, promoting peritoneal dissemination. Conversely, Hu et al. [71] reported the suppressive role of circ_ITCH function in the malignant progression of EOC in vitro and in vivo. Circ_ITCH acted as a ceRNA to sponge miR-145, increasing the level of the Ras p21 protein activator RASA1, a regulator of Ras GDP and GTP, which are involved in angiogenesis, cell proliferation, and apoptosis [152].
MiR-370 serves as a sponge for circ_0025033, circ_0070203, and circ_0000714. Ma et al. [119] reported that circ_0025033 affected the mitochondrial glutamine transporter for metabolic reprogramming SLC1A5 in EOC cells [144], via sponging miR-370-3p, resulting in the abolition of its anti-ovarian cancer role. In serous ovarian cystadenocarcinoma (HGSOC) FIGO stages III-IV, Tang et al. showed that circ_0070203 could upregulate the expression of the TGFβ receptor 2 via sponging miR-370-3p using cell lines and tissues. Overexpression enhanced the migrative, invasive abilities of EOC cells though the expression of EMT-related proteins [3]. Using a microarray, Guo et al. [130] detected that RAB GTPase family proteins were significantly overexpressed in paclitaxel-resistant EOC cells. Circ_0000714 acted as a sponge for miR-370-3p, and increased the expression of the Ras-related protein RAB17 through the CDK6/RB signaling pathway, which plays a role in the malignant progression of paclitaxel-resistant EOC cells.
MiR-532 has been shown to be inhibited by circ_0007841, circ_0025033, and circ_ATP2B4. In a xenograft mouse model, Gao et al. [123] showed that circ_0007841 conferred cisplatin resistance through the miR-532-5p/NFIB axis. In an interrelated manner, Huang et al. [131] detected that circ_0025033 upregulated the expression of the proliferation-associated transcription factor FOXM1 by sponging miR-532-3p, playing a role in paclitaxel resistance. Wang et al. [109] reported the positive correlation of upregulated circ_ATP2B4 with EOC progression. Exosomes transmitted circ_ATP2B4, which acted as the ceRNA of miR-532-3p, to infiltrated macrophages, to relieve the repressive effect of miR-532-3p on its target of the sterol regulatory element-binding factor SREBF1. In addition, circ_ATP2B4 induced macrophage M2 polarization by regulating the miR-532-3p/SREBF1/PI3Kα/AKT axis, thereby resulting in immunosuppression and EOC metastasis in vitro and in vivo.
Finally, circ_0010467, circ_0013958, and circ_0051240 have been revealed to be ceRNAs for miR-637. Wu et al. [121] observed an increased expression of circ_0010467 in platinum-resistant EOC cells, tissues, and serum exosomes, and a positive correlation with advanced tumor stages and the poor prognosis of EOC patients. The AU-rich element RNA-binding protein AUF1 stimulated circ_0010467 to promote platinum resistance through inducing tumor cell stemness, while circ_0010467 acted as a miR-637 sponge to activate the leukemia inhibitory factor (LIF) in the miR-637/LIF/STAT3 axis. Liang et al. [96] found that upregulated circ_0013958 downregulated miR-637 resulting in the activation of the surface receptor PLXNB2 in EOC cells. Knockdown of circ_0013958 impeded EOC development through modulating the miR-637/PLXNB2 axis. Zhang et al. [103] observed significantly increased levels of circ_0051240 in EOC tissues. Circ_0051240 acted as a sponge for miR-637 which targeted the kallikrein-related peptidase 4 KLK4 mRNAs in EOC cells. It promoted EOC cell proliferation, migration, and invasion in vitro, while it stimulated tumor formation in vivo.

7. EOC Therapies

The current clinical standard treatment strategy usually includes tumor cytoreductive surgery followed by platinum and paclitaxel chemotherapy. Platinum, such as cisplatin and carboplatin, covalently binds to purine bases and so introduces DNA damage, including monoadducts or inter- and intra-strand crosslinks. This leads to the interference of the replication machinery, G2/M cell arrest, and cell death by apoptosis or necrosis [153]. Paclitaxel additionally disrupts cell division by impeding the breakdown of the spindle apparatus during mitosis. As a result, the mitosis remains incomplete with no reproduction of the cells by the non-distributed DNA [154].
However, frequently, patients succumb to chemotherapeutic resistance and recurrence, even within several years after the initial treatment with these drugs. Chemotherapy resistance is one of the main obstacles in cancer treatment. Despite the improved chemotherapy regimens, such as intraperitoneal delivery and target therapies including poly (ADP-ribose) polymerase (PARP) inhibitors and antiangiogenic agents, these methods have only somewhat contributed to extend the 5-year survival rate in advanced EOC. To address the problem of drug resistance and extend the 5-year survival rate, PARP inhibitors have been coupled with ferroptosis, a type of programmed cell death dependent on iron which accumulates lipid peroxides [155]. Likewise, the response rates to immunotherapy with immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR)-, and T cell receptor (TCR)-engineered T cells among EOC patients remain modest. Newly developed therapeutic targets that utilize nanomedicine technology provide new chances for the treatment of EOC patients, and might have the potential to prolong patient survival. However, the efficacy of such drugs may be accompanied with hyper-progressive disease and the toxicity of the treatments [156].
It has been reported that changes in the canonical Wnt/β-catenin and the Notch signaling pathways are relevant in EOC development, progression, and resistance. In the Wnt/β-catenin signaling pathway, Wnt proteins bind to receptors of the Frizzled and low-density lipoprotein receptor-related protein families on the cell surface. The generated signal is transferred through several cytoplasmic components until β-catenin, which in turn enters the nucleus to form a complex with the transcription factor TCF and activates the transcription of the Wnt target genes. Modulations of the Wnt/β-catenin signaling pathway include mutations in β-catenin or other key pathway members, as well as hypermethylation and silencing of gatekeeper antagonists, or overexpression of Wnt ligands or receptors. They lead to increased cancer cell proliferation and migration [157]. The Notch signaling pathway is activated by a receptor-ligand binding between two neighboring cells, leading to a conformational change in the Notch receptor. Following two cleavages, the Notch intracellular domain is transferred into the nucleus, where it binds to ubiquitous transcription factor CSL and converts a large co-repressor complex into a transcription activating complex. As a result, the transcription of Notch target genes is stimulated, among others p21, cyclin D1 and 3, c-myc, and members of the NF-κB family, which regulate proliferation, differentiation and apoptosis [151].
In particular, the modulation of these signaling pathways has led to the development of therapeutics that target these pathways. Accordingly, the development of new therapies using ncRNAs, such as miRNAs or circRNAs, may be of strategical interest.

8. circRNAs as Predictive Biomarkers

Due to the lack of an effective early detection screening test, the majority of EOC patients are initially diagnosed with advanced disease. CircRNAs play important roles in cancer tumorigenesis and progression, and represent prognostic biomarkers. Therefore, they could be eligible for a screening test. To date, there are numerous studies on circRNAs that describe circRNAs as biomarkers in tissues, plasma, serum and exosomes for EOC (Table 3).

9. circRNAs as Therapeutic Agents and Targets

So far, most studies have focused on the development of vaccines and nucleic acid-derived drugs considering miRNAs as therapeutic markers. In this respect, numerous investigations have taken advantage of the inhibitory effects of miRNAs on translation. MiRNAs can have oncogenic or/and tumor suppressive behavior. Restoring the miRNA tumor-suppressive function and inhibiting the oncogenic function have been considered to improve cancer treatment. Hence, assays, such as synthetic tumor suppressive miRNA mimics and anti-oncogenic agomiRs that bind miRNAs to agonist their oncogenic potential, have been developed [172,173,174]. However, circRNAs mainly act as a sponge for miRNAs, and so may serve as agomiRs, disturbing the function of miRNAs. Therefore, the interplay between miRNAs and circRNAs should be considered.
To date, the application of circRNAs in potential therapy approaches has been reviewed by Holdt et al. [175]. The chemistry, manufacturing, and controls (CMCs) process for circRNAs is comprised of the following four steps: (1) plasmid construction and proof of concept; (2) in vitro synthesis and purification; (3) circularization (spliceosomes and ligases) and purification; and (4) encapsulation and partitioning. Different methods to either overexpress or knockdown circRNAs in vitro and in vivo have been established [176]. Overexpression of circRNAs can be achieved by providing plasmids containing circRNA-producing exons. They can also be synthesized as miRNA mimics by applying RNA ligases or ribozymes and introducing photolabile linkers. In addition, synthetic circRNAs can be engineered to contain several miRNA-binding sites. They are transcribed and cyclized in vitro and can be delivered to specific tissue with adeno-associated virus (AAV) [177,178]. Chemical modifications of circRNAs improve stability and binding affinity, while coatings of circRNAs with proteins facilitates their recognition by cancer cells. In contrast, silencing of circRNAs can be achieved by siRNA, short hairpin RNAs (shRNA), CRISPR/Cas9, and RNA-targeting Cas13 system [179,180]. Typically, shRNAs and siRNAs that are 20–25 base pairs, double stranded RNA molecules operate via the RNA interference, and have been widely applied in functional studies to inhibit oncogenic circRNAs in vitro and in vivo [179]. It is worth mentioning that the CRISPR/Cas9 system established the first circRNA knockout mice model to knockdown circRNAs without apparent off-target effects. In addition, the CRISPR/Cas13 system successfully achieved the knockdown of circRNA expression [180].
In order to efficiently deliver circRNAs or their inhibitors to the target organs, various techniques have been employed including nanoparticles, adenoviruses, and plasmids [17,181,182,183,184]. Nanoparticles which are frequently used can be produced by organic materials, such as liposomes, polymers, and dendrimers or inorganic materials, such as gold and metal oxides [185].

10. Conclusions

Aberrant signal transduction pathways lead to tumor progression and metastasis and are excellent targets for therapeutic approaches that permit the inhibition of oncogenic signals. Therefore, the eligibility of ncRNAs that modulate these pathways should be considered for the use of therapeutic agents as well as biomarkers since they may reflect the dynamic of the disease course, including recurrence and resistance to therapy. The interplay between circRNAs and miRNAs provides new information that may contribute to understand the complex biological network of ncRNAs in the regulation of the mechanisms underlying EOC pathogenesis. The establishment of biological network models in which ncRNAs, e.g., circRNAs and miRNAs as well as lncRNAs, are involved have to be created to design EOC-associated signatures that allow their application for screening or treatment decisions by the physician. However, the multiple interactions between circRNAs and miRNAs should be taken into account. One circRNA can sponge several miRNAs and several circRNAs can sponge one miRNA. In addition, miRNAs can also be inhibited by other ncRNAs, such as lncRNAs. In addition, one signaling pathway can be disturbed by several ncRNAs. Conversely, one ncRNA can affect multiple signal pathways that may even cross-talk among each other. This complex interplay among ncRNAs and signaling pathways makes it particularly difficult to exactly define the effectiveness of a treatment with these molecules in a larger context. Therefore, before developing a targeted therapy using circRNAs to suppress miRNAs, their manifold behavior and interaction in both time and cellular location should be analyzed. Consequently, this should also include a circRNA as a therapeutic agent, which is considered to inhibit an oncogenic miRNA, and may also potentially inhibit tumor suppressive miRNAs.
Furthermore, researchers have identified numerous targets of miRNAs across different pathways within the same tumor types, including EOC. One miRNA is able to bind to a broad range of mRNA targets, leading to the inhibition and modulation of the expression of a variety of proteins, involved in both oncogenic and tumor suppressive functions and in different signaling pathways. These off-target effects mediated by miRNAs may result in potential non-specific toxicity and elicit unintended cellular and harmful immune responses that may accompany the treatment outcome. MiR-34 is an example for restoring tumor-suppressive functions along with non-specific toxicity. This miRNA is significantly downregulated in different cancer types including EOC and inhibits multiple oncogenic pathways [186]. A first-in-human phase I clinical trial (NCT01829971) was conducted to investigate the safety, pharmacokinetics, and clinical activity of a liposomal formulation of an miR-34 mimic (known as MRX34). The clinical study was stopped by the sponsoring company (Mirna Therapeutic, Inc.) since the patients dosed with MRX34 displayed multiple immune-related severe adverse effects after initial good tolerability [187]. Thus, designing tumor suppressive miRNA mimics or inhibitors for oncogenic miRNAs with a better specificity can help mitigate off-target effects. In addition, the technical platforms and the selection of an appropriate delivery system have to be advanced. New possibilities provide exosomes manipulated with specific surface marker to direct then to the cancer cells. To address these issues and assure that patients do not suffer from substantial adverse side effects, the development of an assay using a circRNA as a sponge for miRNAs may be a laborious endeavor.
In summary, the decoding of the regulatory network of ncRNAs remains an important task for the future to develop effective therapeutic agents to inhibit tumor progression and to overcome drug resistance.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

ABCCATP-Binding Cassette Subfamily C Member
ANXAAnnexin A
ATGAutophagy-related
BRD4Bromodomain
CapnCalpain
CBXChromobox
CDC5L/PEAK1Cell Division Cycle 5 Like
CEBPGCCAAT Enhancer-Binding Protein Gamma
CLUClusterin
CULCullin
DUSPDual Specificity Phosphatase
EIFEukaryotic Translation Initiation Factor
ELKETS Transcription Factor
EZHEnhancer of Zeste Homolog
FBXF-box protein
FMNLFormin-like
FOXForkhead-box
FZDFrizzled Class Receptor
GASGrowth Arrest-Specific
hTERTHuman Telomerase Reverse Transcriptase
IGFBPInsulin-like growth factor binding protein
JAK1Janus Kinase
KLFKrüppel-like factor
KLKKallikrein-Related Peptidase
LARPLa Ribonucleoprotein
LETM1Leucine Zipper-EF-Hand-Containing Transmembrane Protein
LIFLeukemia Inhibitory Factor
LSMU6 snRNA-Associated Sm-like Protein
MAP3KMitogen-Activated Protein Kinase
MDMMouse double minute
MTMMyotubular Myopathy
MTSSMetastasis suppressor
MUC1Mucin
NFNuclear Factor
p-GPP-Glycoprotein
PIK3RPhosphoinositide-3-Kinase Regulatory Subunit
PLXNPlexin
PPAInorganic Pyrophosphatase
PSATPhosphoserine Aminotransferase
PTENPhosphatase and Tensin homolog
RABRas-related protein
RACGAPRac GTPase Activating Protein
RASSFRas Association Domain Family Member
ROCKRho-Kinase
S100BCalcium Binding Protein B
SFRPSecreted Frizzled Related Protein
SIKSalt Inducible Kinase
SLCSolute Carrier Family
SMGSerine/threonine-protein kinase
SOCSSuppressor of Cytokine Signaling
SOXSRY-Box Transcription Factor
SREBFSterol regulatory element-binding transcription factor
STATSignal transducer and activator of transcription
TGFβRTransforming Growth Factor β receptor
TUBBTubulin Beta
VAMPVesicle-Associated Membrane Protein
VEGFVascular Endothelial Growth Factor

References

  1. Sambasivan, S. Epithelial Ovarian Cancer: Review Article. Cancer Treat Res. Commun. 2022, 33, 100629. [Google Scholar] [CrossRef] [PubMed]
  2. Kandukuri, S.R.; Rao, J. FIGO 2013 Staging System for Ovarian Cancer: What Is New in Comparison to the 1988 Staging System? Curr. Opin. Obstet. Gynecol. 2015, 27, 48–52. [Google Scholar] [CrossRef] [PubMed]
  3. Jonckheere, S.; Adams, J.; De Groote, D.; Campbell, K.; Berx, G.; Goossens, S. Epithelial-Mesenchymal Transition (EMT) as a Therapeutic Target. Cells Tissues Organs 2022, 211, 157–182. [Google Scholar] [CrossRef] [PubMed]
  4. Morand, S.; Devanaboyina, M.; Staats, H.; Stanbery, L.; Nemunaitis, J. Ovarian Cancer Immunotherapy and Personalized Medicine. Int. J. Mol. Sci. 2021, 22, 6532. [Google Scholar] [CrossRef] [PubMed]
  5. Zhao, L.; Liang, X.; Wang, L.; Zhang, X. The Role of MiRNA in Ovarian Cancer: An Overview. Reprod. Sci. 2022, 29, 561–575. [Google Scholar] [CrossRef]
  6. Bartel, D.P. MicroRNAs: Target Recognition and Regulatory Functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef]
  7. Zhang, Y.; Fan, M.; Zhang, X.; Huang, F.; Wu, K.; Zhang, J.; Liu, J.; Huang, Z.; Luo, H.; Tao, L.; et al. Cellular MicroRNAs Up-Regulate Transcription via Interaction with Promoter TATA-Box Motifs. RNA 2014, 20, 1878–1889. [Google Scholar] [CrossRef]
  8. Schwarzenbach, H.; Hoon, D.S.B.; Pantel, K. Cell-Free Nucleic Acids as Biomarkers in Cancer Patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef]
  9. Wu, J.; Yang, J.; Cho, W.C.; Zheng, Y. Argonaute Proteins: Structural Features, Functions and Emerging Roles. J. Adv. Res. 2020, 24, 317–324. [Google Scholar] [CrossRef]
  10. Harding, C.V.; Heuser, J.E.; Stahl, P.D. Exosomes: Looking Back Three Decades and into the Future. J. Cell Biol. 2013, 200, 367–371. [Google Scholar] [CrossRef]
  11. Hu, G.; Drescher, K.M.; Chen, X.-M. Exosomal MiRNAs: Biological Properties and Therapeutic Potential. Front. Genet. 2012, 3, 56. [Google Scholar] [CrossRef]
  12. Schwarzenbach, H.; Gahan, P. MicroRNA Shuttle from Cell-To-Cell by Exosomes and Its Impact in Cancer. Noncoding RNA 2019, 5, 28. [Google Scholar] [CrossRef]
  13. Chen, L.; Shan, G. CircRNA in Cancer: Fundamental Mechanism and Clinical Potential. Cancer Lett. 2021, 505, 49–57. [Google Scholar] [CrossRef] [PubMed]
  14. Khan, S.; Jha, A.; Panda, A.C.; Dixit, A. Cancer-Associated CircRNA–MiRNA–MRNA Regulatory Networks: A Meta-Analysis. Front. Mol. Biosci. 2021, 8, 671309. [Google Scholar] [CrossRef] [PubMed]
  15. Park, E.G.; Ha, H.; Lee, D.H.; Kim, W.R.; Lee, Y.J.; Bae, W.H.; Kim, H.S. Genomic Analyses of Non-Coding RNAs Overlapping Transposable Elements and Its Implication to Human Diseases. Int. J. Mol. Sci. 2022, 23, 8950. [Google Scholar] [CrossRef]
  16. Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-Coding RNA Networks in Cancer. Nat. Rev. Cancer 2017, 18, 5–18. [Google Scholar] [CrossRef]
  17. Liu, Y.; Wang, J. Therapeutic Potentials of Noncoding RNAs: Targeted Delivery of NcRNAs in Cancer Cells. In Advances in Experimental Medicine and Biology; Springer: Berlin/Heidelberg, Germany, 2016; Volume 927, pp. 429–458. [Google Scholar]
  18. Chan, J.; Tay, Y. Noncoding RNA:RNA Regulatory Networks in Cancer. Int. J. Mol. Sci. 2018, 19, 1310. [Google Scholar] [CrossRef] [PubMed]
  19. Ho, P.T.B.; Clark, I.M.; Le, L.T.T. MicroRNA-Based Diagnosis and Therapy. Int. J. Mol. Sci. 2022, 23, 7167. [Google Scholar] [CrossRef]
  20. Bortolin-Cavaille, M.-L.; Dance, M.; Weber, M.; Cavaille, J. C19MC MicroRNAs Are Processed from Introns of Large Pol-II, Non-Protein-Coding Transcripts. Nucleic Acids Res. 2009, 37, 3464–3473. [Google Scholar] [CrossRef]
  21. Lee, Y.; Kim, M.; Han, J.; Yeom, K.-H.; Lee, S.; Baek, S.H.; Kim, V.N. MicroRNA Genes Are Transcribed by RNA Polymerase II. EMBO J. 2004, 23, 4051–4060. [Google Scholar] [CrossRef]
  22. Chendrimada, T.P.; Gregory, R.I.; Kumaraswamy, E.; Norman, J.; Cooch, N.; Nishikura, K.; Shiekhattar, R. TRBP Recruits the Dicer Complex to Ago2 for MicroRNA Processing and Gene Silencing. Nature 2005, 436, 740–744. [Google Scholar] [CrossRef] [PubMed]
  23. Vishnoi, A.; Rani, S. MiRNA Biogenesis and Regulation of Diseases: An Updated Overview. In Methods in Molecular Biology; Springer: Berlin/Heidelberg, Germany, 2023; Volume 2595, pp. 1–12. [Google Scholar] [CrossRef]
  24. Schwarzenbach, H.; Nishida, N.; Calin, G.A.; Pantel, K. Clinical Relevance of Circulating Cell-Free MicroRNAs in Cancer. Nat. Rev. Clin. Oncol. 2014, 11, 145–156. [Google Scholar] [CrossRef] [PubMed]
  25. Staicu, C.E.; Predescu, D.V.; Rusu, C.M.; Radu, B.M.; Cretoiu, D.; Suciu, N.; Crețoiu, S.M.; Voinea, S.C. Role of MicroRNAs as Clinical Cancer Biomarkers for Ovarian Cancer: A Short Overview. Cells 2020, 9, 169. [Google Scholar] [CrossRef]
  26. Schwarzenbach, H.; Gahan, P.B. Circulating Non-Coding RNAs in Recurrent and Metastatic Ovarian Cancer. Cancer Drug Resist. 2019, 2, 399. [Google Scholar] [CrossRef]
  27. Liu, H.Y.; Zhang, Y.Y.; Zhu, B.L.; Feng, F.Z.; Yan, H.; Zhang, H.Y.; Zhou, B. MiR-21 Regulates the Proliferation and Apoptosis of Ovarian Cancer Cells through PTEN/PI3K/AKT. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4149–4155. [Google Scholar] [CrossRef] [PubMed]
  28. Yang, H.; Kong, W.; He, L.; Zhao, J.J.; O’Donnell, J.D.; Wang, J.; Wenham, R.M.; Coppola, D.; Kruk, P.A.; Nicosia, S.V.; et al. MicroRNA Expression Profiling in Human Ovarian Cancer: MiR-214 Induces Cell Survival and Cisplatin Resistance by Targeting PTEN. Cancer Res. 2008, 68, 425–433. [Google Scholar] [CrossRef]
  29. Ghafouri-Fard, S.; Shoorei, H.; Taheri, M. MiRNA Profile in Ovarian Cancer. Exp. Mol. Pathol. 2020, 113, 104381. [Google Scholar] [CrossRef]
  30. Li, N.; Yang, L.; Wang, H.; Yi, T.; Jia, X.; Chen, C.; Xu, P. MiR-130a and MiR-374a Function as Novel Regulators of Cisplatin Resistance in Human Ovarian Cancer A2780 Cells. PLoS ONE 2015, 10, e0128886. [Google Scholar] [CrossRef]
  31. Meng, X.; Müller, V.; Milde-Langosch, K.; Trillsch, F.; Pantel, K.; Schwarzenbach, H. Diagnostic and Prognostic Relevance of Circulating Exosomal MiR-373, MiR-200a, MiR-200b and MiR-200c in Patients with Epithelial Ovarian Cancer. Oncotarget 2016, 7, 16923–16935. [Google Scholar] [CrossRef]
  32. Pan, C.; Stevic, I.; Müller, V.; Ni, Q.; Oliveira-Ferrer, L.; Pantel, K.; Schwarzenbach, H. Exosomal MicroRNAs as Tumor Markers in Epithelial Ovarian Cancer. Mol. Oncol. 2018, 12, 1935–1948. [Google Scholar] [CrossRef]
  33. Yu, R.; Cai, L.; Chi, Y.; Ding, X.; Wu, X. MiR-377 Targets CUL4A and Regulates Metastatic Capability in Ovarian Cancer. Int. J. Mol. Med. 2018, 41, 3147–3156. [Google Scholar] [CrossRef] [PubMed]
  34. Huang, Z.; Li, Q.; Luo, K.; Zhang, Q.; Geng, J.; Zhou, X.; Xu, Y.; Qian, M.; Zhang, J.-a.; Ji, L.; et al. MiR-340-FHL2 Axis Inhibits Cell Growth and Metastasis in Ovarian Cancer. Cell Death Dis. 2019, 10, 372. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, W.; He, Y.; Zhang, Y. CircRNA in Ocular Neovascular Diseases: Fundamental Mechanism and Clinical Potential. Pharmacol. Res. 2023, 197, 106946. [Google Scholar] [CrossRef]
  36. Li, X.; Yang, L.; Chen, L.-L. The Biogenesis, Functions, and Challenges of Circular RNAs. Mol. Cell 2018, 71, 428–442. [Google Scholar] [CrossRef]
  37. Hu, Q.; Zhou, T. EIciRNA-Mediated Gene Expression: Tunability and Bimodality. FEBS Lett. 2018, 592, 3460–3471. [Google Scholar] [CrossRef]
  38. Li, J.; Zhang, G.; Liu, C.-G.; Xiang, X.; Le, M.T.N.; Sethi, G.; Wang, L.; Goh, B.-C.; Ma, Z. The Potential Role of Exosomal CircRNAs in the Tumor Microenvironment: Insights into Cancer Diagnosis and Therapy. Theranostics 2022, 12, 87–104. [Google Scholar] [CrossRef]
  39. Chen, J.; Gu, J.; Tang, M.; Liao, Z.; Tang, R.; Zhou, L.; Su, M.; Jiang, J.; Hu, Y.; Chen, Y.; et al. Regulation of Cancer Progression by CircRNA and Functional Proteins. J. Cell Physiol. 2022, 237, 373–388. [Google Scholar] [CrossRef]
  40. Donati, B.; Lorenzini, E.; Ciarrocchi, A. BRD4 and Cancer: Going beyond Transcriptional Regulation. Mol. Cancer 2018, 17, 164. [Google Scholar] [CrossRef]
  41. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT Signaling Pathway: From Bench to Clinic. Signal Transduct. Target Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
  42. MacDonald, B.T.; Tamai, K.; He, X. Wnt/Beta-Catenin Signaling: ComponentMacDonald BT; Tamai, K.; He, X. Wnt/Beta-Catenin Signaling: Components, Mechanisms, and Diseases. Dev. Cell 2009, 17, 9–26. [Google Scholar] [CrossRef]
  43. Cargnello, M.; Roux, P.P. Activation and Function of the MAPKs and Their Substrates, the MAPK-Activated Protein Kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef] [PubMed]
  44. Fruman, D.A.; Chiu, H.; Hopkins, B.D.; Bagrodia, S.; Cantley, L.C.; Abraham, R.T. The PI3K Pathway in Human Disease. Cell 2017, 170, 605–635. [Google Scholar] [CrossRef] [PubMed]
  45. Wu, D.; Liu, J.; Yu, L.; Wu, S.; Qiu, X. Circular RNA Hsa_circ_0000144 Aggravates Ovarian Cancer Progression by Regulating ELK3 via Sponging MiR-610. J. Ovarian Res. 2022, 15, 113. [Google Scholar] [CrossRef]
  46. Yu, S.; Yu, M.; Chen, J.; Tang, H.; Gong, W.; Tan, H. Circ_0000471 Suppresses the Progression of Ovarian Cancer through Mediating Mir-135b-5p/Dusp5 Axis. Am. J. Reprod. Immunol. 2023, 89, e13651. [Google Scholar] [CrossRef] [PubMed]
  47. Wu, Y.; Zhou, J.; Li, Y.; Shi, X.; Shen, F.; Chen, M.; Chen, Y.; Wang, J. Hsa_circ_0001445 Works as a Cancer Suppressor via MiR-576-5p/SFRP1 Axis Regulation in Ovarian Cancer. Cancer Med. 2023, 12, 5736–5750. [Google Scholar] [CrossRef] [PubMed]
  48. Zhou, X.; Jiang, J.; Guo, S. Hsa_circ_0004712 Downregulation Attenuates Ovarian Cancer Malignant Development by Targeting the MiR-331-3p/FZD4 Pathway. J. Ovarian Res. 2021, 14, 118. [Google Scholar] [CrossRef]
  49. Zhang, M.; Sun, Y.; Xu, H.; Shi, Y.; Shen, R.; Teng, F.; Xu, J.; Jia, X. Circular Rna Hsa_circ_0007444 Inhibits Ovarian Cancer Progression through Mir-23a-3p/Dicer1 Axis. Acta Biochim. Biophys. Sin. 2023, 55, 574–586. [Google Scholar] [CrossRef]
  50. Wei, W.; Wang, N.; Lin, L. Prognostic Value of Hsa_circ_0007615 in Epithelial Ovarian Cancer and Its Regulatory Effect on Tumor Progression. Horm. Metab. Res. 2023, 55, 801–808. [Google Scholar] [CrossRef]
  51. Li, L.; Yu, P.; Zhang, P.; Wu, H.; Chen, Q.; Li, S.; Wang, Y. Upregulation of Hsa_circ_0007874 Suppresses the Progression of Ovarian Cancer by Regulating the MiR-760/SOCS3 Pathway. Cancer Med. 2020, 9, 2491–2499. [Google Scholar] [CrossRef]
  52. Li, Y.; Lin, S.; An, N. Hsa_circ_0009910: Oncogenic Circular RNA Targets MicroRNA-145 in Ovarian Cancer Cells. Cell Cycle 2020, 19, 1857–1868. [Google Scholar] [CrossRef]
  53. Pan, Y.; Huang, Q.; Peng, X.; Yu, S.; Liu, N. Circ_0015756 Promotes Ovarian Cancer Progression via the MiR-145–5p/PSAT1 Axis. Reprod. Biol. 2022, 22, 100702. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, L.; Han, B.; Liu, L.; Cui, H.; Liu, H.; Jia, R.; Zhang, X.; Lu, X. Circ_0021573 Acts as a Competing Endogenous RNA to Promote the Malignant Phenotypes of Human Ovarian Cancer Cells. Reprod. Biol. 2023, 23, 100704. [Google Scholar] [CrossRef] [PubMed]
  55. Huang, X.; Luo, Y.; Li, X. Circ_0072995 Promotes Ovarian Cancer Progression Through Regulating MiR-122-5p/SLC1A5 Axis. Biochem. Genet. 2022, 60, 153–172. [Google Scholar] [CrossRef]
  56. Jin, Y.; Wang, H. Circ_0078607 Inhibits the Progression of Ovarian Cancer via Regulating the MiR-32-5p/SIK1 Network. J. Ovarian Res. 2022, 15, 3. [Google Scholar] [CrossRef]
  57. Hou, W.; Zhang, Y. Circ_0025033 Promotes the Progression of Ovarian Cancer by Activating the Expression of LSM4 via Targeting MiR-184. Pathol. Res. Pract. 2021, 217, 153275. [Google Scholar] [CrossRef]
  58. Zhang, Y.; Di, Q.; Chen, J.; Chang, M.; Ma, Y.; Yu, J. Circ_0061140 Contributes to the Malignant Progression in Ovarian Cancer Cells by Mediating the RAB1A Level Through Sponging MiR-361-5p. Biochem. Genet. 2022, 60, 1946–1962. [Google Scholar] [CrossRef]
  59. Ma, L.; Liu, W.; Li, M. Circ_0061140 Contributes to Ovarian Cancer Progression by Targeting MiR-761/LETM1 Signaling. Biochem. Genet. 2023, 61, 628–650. [Google Scholar] [CrossRef] [PubMed]
  60. Zhang, N.; Jin, Y.; Hu, Q.; Cheng, S.; Wang, C.; Yang, Z.; Wang, Y. Circular RNA Hsa_circ_0078607 Suppresses Ovarian Cancer Progression by Regulating MiR-518a-5p/Fas Signaling Pathway. J. Ovarian Res. 2020, 13, 64. [Google Scholar] [CrossRef]
  61. Lyu, M.; Li, X.; Shen, Y.; Lu, J.; Zhang, L.; Zhong, S.; Wang, J. CircATRNL1 and CircZNF608 Inhibit Ovarian Cancer by Sequestering MiR-152-5p and Encoding Protein. Front. Genet. 2022, 13, 784089. [Google Scholar] [CrossRef]
  62. Lu, M.; Gong, B.; Wang, Y.; Li, J. CircBNC2 Affects Epithelial Ovarian Cancer Progression through the MiR-223-3p/LARP4 Axis. Anticancer Drugs 2023, 34, 384–394. [Google Scholar] [CrossRef]
  63. Liu, T.; Yuan, L.; Zou, X. Circular RNA Circ-BNC2 (Hsa_circ_0008732) Inhibits the Progression of Ovarian Cancer through MicroRNA-223-3p/FBXW7 Axis. J. Ovarian Res. 2022, 15, 95. [Google Scholar] [CrossRef] [PubMed]
  64. He, S.L.; Zhao, X.; Yi, S.J. CircAHNAK Upregulates EIF2B5 Expression to Inhibit the Progression of Ovarian Cancer by Modulating the JAK2/STAT3 Signaling Pathway. Carcinogenesis 2022, 43, 941–955. [Google Scholar] [CrossRef]
  65. Chen, H.; Mao, M.; Jiang, J.; Zhu, D.; Li, P. Circular RNA CDR1as Acts as a Sponge of MiR-135b-5p to Suppress Ovarian Cancer Progression. Onco. Targets Ther. 2019, 12, 3869–3879. [Google Scholar] [CrossRef] [PubMed]
  66. Du, Y.; Liu, X.; Zhang, S.; Chen, S.; Guan, X.; Li, Q.; Chen, X.; Zhao, Y. CircCRIM1 Promotes Ovarian Cancer Progression by Working as CeRNAs of CRIM1 and Targeting MiR-383-5p/ZEB2 Axis. Reprod. Biol. Endocrinol. 2021, 19, 176. [Google Scholar] [CrossRef]
  67. Xie, J.; Wang, S.; Li, G.; Zhao, X.; Jiang, F.; Liu, J.; Tan, W. CircEPSTI1 Regulates Ovarian Cancer Progression via Decoying MiR-942. J. Cell Mol. Med. 2019, 23, 3597–3602. [Google Scholar] [CrossRef]
  68. Wang, Z.; Zhang, W.; Fang, J.; Xie, P.; Miao, M.; Yang, H. Circular Rna Circexoc6b Inhibits the Progression of Ovarian Cancer by Sponging MiR-421 and Regulating Rus1 Expression. Onco. Targets Ther. 2020, 13, 8233–8243. [Google Scholar] [CrossRef]
  69. Sun, D.; Liu, J.; Zhou, L. Upregulation of Circular RNA Circ-FAM53B Predicts Adverse Prognosis and Accelerates the Progression of Ovarian Cancer via the MiR-646/VAMP2 and MiR-647/MDM2 Signaling Pathways. Oncol. Rep. 2019, 42, 2728–2737. [Google Scholar] [CrossRef]
  70. Wu, M.; Qiu, Q.; Zhou, Q.; Li, J.; Yang, J.; Zheng, C.; Luo, A.; Li, X.; Zhang, H.; Cheng, X.; et al. CircFBXO7/MiR-96-5p/MTSS1 Axis Is an Important Regulator in the Wnt Signaling Pathway in Ovarian Cancer. Mol. Cancer 2022, 21, 137. [Google Scholar] [CrossRef]
  71. Hu, J.; Wang, L.; Chen, J.; Gao, H.; Zhao, W.; Huang, Y.; Jiang, T.; Zhou, J.; Chen, Y. The Circular RNA Circ-ITCH Suppresses Ovarian Carcinoma Progression through Targeting MiR-145/RASA1 Signaling. Biochem. Biophys. Res. Commun. 2018, 505, 222–228. [Google Scholar] [CrossRef]
  72. Song, R.; Chai, T.; Liu, J.; Chu, A.; Sun, C.; Liu, Z. Knockdown of CircMFN2 Inhibits Cell Progression and Glycolysis by MiR-198/CUL4B Pathway in Ovarian Cancer. J. Biochem. Mol. Toxicol. 2023, 37, e23383. [Google Scholar] [CrossRef]
  73. Yang, H.; Guo, Y.; Zhang, Y.; Wang, D.; Zhang, G.; Hou, J.; Yang, J. Circ_MUC16 Attenuates the Effects of Propofol to Promote the Aggressive Behaviors of Ovarian Cancer by Mediating the MiR-1182/S100B Signaling Pathway. BMC Anesthesiol. 2021, 21, 297. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, R.; Ye, H.; Yang, B.; Ao, M.; Yu, X.; Wu, Y.; Xi, M.; Hou, M. M6A-Modified CircNFIX Promotes Ovarian Cancer Progression and Immune Escape via Activating IL-6R/JAK1/STAT3 Signaling by Sponging MiR-647. Int. Immunopharmacol. 2023, 124, 110879. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, C.; Li, Y.; Zhao, W.; Liu, G.; Yang, Q. Circ-PGAM1 Promotes Malignant Progression of Epithelial Ovarian Cancer through Regulation of the MiR-542-3p/CDC5L/PEAK1 Pathway. Cancer Med. 2020, 9, 3500–3521. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, H.; Lan, S.; Wang, L.; Zhao, J.; Jia, X.; Xu, J.; Sun, G.; Liu, L.; Gong, S.; Wang, N.; et al. Expression of Circ-PHC3 Enhances Ovarian Cancer Progression via Regulation of the MiR-497-5p/SOX9 Pathway. J. Ovarian Res. 2023, 16, 142. [Google Scholar] [CrossRef] [PubMed]
  77. Sun, Y.; Li, X.; Chen, A.; Shi, W.; Wang, L.; Yi, R.; Qiu, J. CircPIP5K1A Serves as a Competitive Endogenous RNA Contributing to Ovarian Cancer Progression via Regulation of MiR-661/IGFBP5 Signaling. J. Cell. Biochem. 2019, 120, 19406–19414. [Google Scholar] [CrossRef]
  78. Wu, S.G.; Zhou, P.; Chen, J.X.; Lei, J.; Hua, L.; Dong, Y.; Hu, M.; Lian, C.L.; Yang, L.C.; Zhou, J. Circ-PTK2 (Hsa_circ_0008305) Regulates the Pathogenic Processes of Ovarian Cancer via MiR-639 and FOXC1 Regulatory Cascade. Cancer Cell Int. 2021, 21, 277. [Google Scholar] [CrossRef]
  79. Song, W.; Zeng, Z.; Zhang, Y.; Li, H.; Cheng, H.; Wang, J.; Wu, F. CircRNF144B/MiR-342-3p/FBXL11 Axis Reduced Autophagy and Promoted the Progression of Ovarian Cancer by Increasing the Ubiquitination of Beclin-1. Cell Death Dis. 2022, 13, 857. [Google Scholar] [CrossRef]
  80. Wang, L.L.; Zong, Z.H.; Liu, Y.; Guan, X.; Chen, S.; Zhao, Y. CircRhoC Promotes Tumorigenicity and Progression in Ovarian Cancer by Functioning as a MiR-302e Sponge to Positively Regulate VEGFA. J. Cell Mol. Med. 2019, 23, 8472–8481. [Google Scholar] [CrossRef]
  81. Li, B.; Zhang, L. CircSETDB1 Knockdown Inhibits the Malignant Progression of Serous Ovarian Cancer through MiR-129-3p-Dependent Regulation of MAP3K3. J. Ovarian Res. 2021, 14, 160. [Google Scholar] [CrossRef]
  82. Zong, Z.H.; Du, Y.P.; Guan, X.; Chen, S.; Zhao, Y. CircWHSC1 Promotes Ovarian Cancer Progression by Regulating MUC1 and HTERT through Sponging MiR-145 and MiR-1182. J. Exp. Clin. Cancer Res. 2019, 38, 437. [Google Scholar] [CrossRef]
  83. Sheng, M.; Wei, N.; Yang, H.Y.; Yan, M.; Zhao, Q.X.; Jing, L.J. CircRNA UBAP2 Promotes the Progression of Ovarian Cancer by Sponging MicroRNA-144. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7283–7294. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, L.; Zhou, Q.; Qiu, Q.; Hou, L.; Wu, M.; Li, J.; Li, X.; Lu, B.; Cheng, X.; Liu, P.; et al. CircPLEKHM3 Acts as a Tumor Suppressor through Regulation of the MiR-9/BRCA1/DNAJB6/KLF4/AKT1 Axis in Ovarian Cancer. Mol. Cancer 2019, 18, 144. [Google Scholar] [CrossRef] [PubMed]
  85. Sun, S.; Fang, H. Curcumin Inhibits Ovarian Cancer Progression by Regulating Circ-PLEKHM3/MiR-320a/SMG1 Axis. J. Ovarian Res. 2021, 14, 158. [Google Scholar] [CrossRef] [PubMed]
  86. Zhao, Y.; Hu, Y.; Shen, Q.; Chen, Q.; Zhu, X.J.; Jiang, S.S.; Zhang, Q. CircRNA-MYLK Promotes Malignant Progression of Ovarian Cancer through Regulating MicroRNA-652. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 5281–5291. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, H.; Liang, C.; Lin, J.; Dong, Y.; Wang, Y.; Xia, L. Hsa_circ_0001741 Suppresses Ovarian Cancer Cell Proliferations Through Adsorption of MiR-188-5p and Promotion of FOXN2 Expression. Mol. Biotechnol. 2024, 66, 1477–1483. [Google Scholar] [CrossRef]
  88. Xu, F.; Ni, M.; Li, J.; Cheng, J.; Zhao, H.; Zhao, J.; Huang, S.; Wu, X. Circ0004390 Promotes Cell Proliferation through Sponging MiR-198 in Ovarian Cancer. Biochem. Biophys. Res. Commun. 2020, 526, 14–20. [Google Scholar] [CrossRef]
  89. Luo, L.; Gao, Y.Q.; Sun, X.F. Circular RNA ITCH Suppresses Proliferation and Promotes Apoptosis in Human Epithelial Ovarian Cancer Cells by Sponging MiR-10a-α. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8119–8126. [Google Scholar]
  90. Sun, X.; Luo, L.; Gao, Y. Circular RNA PVT1 Enhances Cell Proliferation but Inhibits Apoptosis through Sponging MicroRNA-149 in Epithelial Ovarian Cancer. J. Obstet. Gynaecol. Res. 2020, 46, 625–635. [Google Scholar] [CrossRef]
  91. Xu, Q.; Deng, B.; Li, M.; Chen, Y.; Zhuan, L. CircRNA-UBAP2 Promotes the Proliferation and Inhibits Apoptosis of Ovarian Cancer Though MiR-382-5p/PRPF8 Axis. J. Ovarian Res. 2020, 13, 81. [Google Scholar] [CrossRef]
  92. Tian, Q.; Mu, Q.; Liu, S.; Huang, K.; Tang, Y.; Zhang, P.; Zhao, J.; Shu, C. M6A-Modified CircASXL1 Promotes Proliferation and Migration of Ovarian Cancer through the MiR-320d/RACGAP1 Axis. Carcinogenesis 2023, 44, 859–870. [Google Scholar] [CrossRef]
  93. Qu, B.; Sun, L.; Xiao, P.; Shen, H.; Ren, Y.; Zhang, J. CircCDK17 Promotes the Proliferation and Metastasis of Ovarian Cancer Cells by Sponging MiR-22-3p to Regulate CD147 Expression. Carcinogenesis 2024, 45, 83–94. [Google Scholar] [CrossRef] [PubMed]
  94. Han, Y.; Zheng, Y.; You, J.; Han, Y.; Lu, X.; Wang, X.; Shi, C.; Zhu, W. Hsa_circ_0001535 Inhibits the Proliferation and Migration of Ovarian Cancer by Sponging MiR-593-3p, Upregulating PTEN Expression. Clin. Transl. Oncol. 2023, 25, 2901–2910. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, H.; Zhang, X.; Qiao, L.; Wang, H. CircRNA Circ_0000554 Promotes Ovarian Cancer Invasion and Proliferation by Regulating MiR-567. Environ. Sci. Pollut. Res. 2022, 29, 19072–19080. [Google Scholar] [CrossRef] [PubMed]
  96. Liang, Y.; Meng, K.; Qiu, R. Circular RNA Circ_0013958 Functions as a Tumor Promoter in Ovarian Cancer by Regulating MiR-637/PLXNB2 Axis. Front. Genet. 2021, 12, 644451. [Google Scholar] [CrossRef]
  97. Lin, X.; Chen, Y.; Ye, X.; Xia, X. Circular RNA ABCB10 Promotes Cell Proliferation and Invasion, but Inhibits Apoptosis via Regulating the MicroRNA-1271-Mediated Capn4/Wnt/β-Catenin Signaling Pathway in Epithelial Ovarian Cancer. Mol. Med. Rep. 2021, 23, 387. [Google Scholar] [CrossRef]
  98. Wang, N.; Cao, Q.X.; Tian, J.; Ren, L.; Cheng, H.L.; Yang, S.Q. Circular RNA MTO1 Inhibits the Proliferation and Invasion of Ovarian Cancer Cells Through the MiR-182-5p/KLF15 Axis. Cell Transplant. 2020, 29, 963689720943613. [Google Scholar] [CrossRef] [PubMed]
  99. Fu, Y.; Sun, H. The Molecular Mechanism of CircRHOBTB3 Inhibits the Proliferation and Invasion of Epithelial Ovarian Cancer by Serving as the CeRNA of MiR-23a-3p. J. Ovarian Res. 2022, 15, 66. [Google Scholar] [CrossRef]
  100. Gong, J.; Xu, X.; Zhang, X.; Zhou, Y. Circular RNA-9119 Suppresses in Ovarian Cancer Cell Viability via Targeting the MicroRNA-21-5p-PTEN-Akt Pathway. Aging 2020, 12, 14314–14328. [Google Scholar] [CrossRef]
  101. Lu, H.; Zheng, G.; Gao, X.; Chen, C.; Zhou, M.; Zhang, L. Propofol Suppresses Cell Viability, Cell Cycle Progression and Motility and Induces Cell Apoptosis of Ovarian Cancer Cells through Suppressing MEK/ERK Signaling via Targeting CircVPS13C/MiR-145 Axis. J. Ovarian Res. 2021, 14, 30. [Google Scholar] [CrossRef]
  102. Wei, X.; Lv, H.; Yang, S.; Yang, X. CircRNA PLOD2 Enhances Ovarian Cancer Propagation by Controlling MiR-378. Saudi J. Biol. Sci. 2021, 28, 6260–6265. [Google Scholar] [CrossRef]
  103. Zhang, M.; Xia, B.; Xu, Y.; Zhang, Y.; Xu, J.; Lou, G. Circular RNA (Hsa:Circ_0051240) Promotes Cell Proliferation, Migration and Invasion in Ovarian Cancer through MiR-637/KLK4 Axis. Artif. Cells Nanomed. Biotechnol. 2019, 47, 224–1233. [Google Scholar] [CrossRef]
  104. Li, Q.-h.; Liu, Y.; Chen, S.; Zong, Z.-h.; Du, Y.-p; Sheng, X.-j.; Zhao, Y. Circ-CSPP1 Promotes Proliferation, Invasion and Migration of Ovarian Cancer Cells by Acting as a MiR-1236-3p Sponge. Biomed. Pharmacother. 2019, 114, 108832. [Google Scholar] [CrossRef] [PubMed]
  105. Ye, H.; Wang, R.Y.; Yu, X.Z.; Wu, Y.K.; Yang, B.W.; Ao, M.Y.; Xi, M.R.; Hou, M.M. Exosomal CircNFIX Promotes Angiogenesis in Ovarian Cancer via MiR-518a-3p/TRIM44 Axis. Kaohsiung J. Med. Sci. 2023, 39, 26–39. [Google Scholar] [CrossRef]
  106. Yang, X.; Wang, J.; Li, H.; Sun, Y.; Tong, X. Downregulation of Hsa_circ_0026123 Suppresses Ovarian Cancer Cell Metastasis and Proliferation through the MiR-124-3p/EZH2 Signaling Pathway. Int. J. Mol. Med. 2021, 47, 668–676. [Google Scholar] [CrossRef]
  107. Zeng, X.Y.; Yuan, J.; Wang, C.; Zeng, D.; Yong, J.H.; Jiang, X.Y.; Lan, H.; Xiao, S.S. CircCELSR1 Facilitates Ovarian Cancer Proliferation and Metastasis by Sponging MiR-598 to Activate BRD4 Signals. Mol. Med. 2020, 26, 70. [Google Scholar] [CrossRef]
  108. Wang, J.; Li, Y.; Zhou, J.H.; Shen, F.R.; Shi, X.; Chen, Y.G. CircATRNL1 Activates Smad4 Signaling to Inhibit Angiogenesis and Ovarian Cancer Metastasis via MiR-378. Mol. Oncol. 2021, 15, 1217–1233. [Google Scholar] [CrossRef]
  109. Wang, F.; Niu, Y.; Chen, K.; Yuan, X.; Qin, Y.; Zheng, F.; Cui, Z.; Lu, W.; Wu, Y.; Xia, D. Extracellular Vesicle-Packaged CircATP2B4 Mediates M2 Macrophage Polarization via MiR-532-3p/SREBF1 Axis to Promote Epithelial Ovarian Cancer Metastasis. Cancer Immunol. Res. 2023, 11, 199–216. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, X.; Ren, X.; Jiaoting, E.; Zhou, Y.; Bian, R. Exosome-Transmitted CircIFNGR2 Modulates Ovarian Cancer Metastasis via MiR-378/ST5 Axis. Mol. Cell Biol. 2023, 43, 22–42. [Google Scholar] [CrossRef]
  111. Xie, W.; Liu, L.; He, C.; Zhao, M.; Ni, R.; Zhang, Z.; Shui, C. Circ_0002711 Knockdown Suppresses Cell Growth and Aerobic Glycolysis by Modulating MiR-1244/ROCK1 Axis in Ovarian Cancer. J. Biosci. 2021, 46, 21. [Google Scholar] [CrossRef]
  112. Deng, G.; Zhou, X.; Chen, L.; Yao, Y.; Li, J.; Zhang, Y.; Luo, C.; Sun, L.; Tang, J. High Expression of ESRP1 Regulated by Circ-0005585 Promotes Cell Colonization in Ovarian Cancer. Cancer Cell Int. 2020, 20, 174. [Google Scholar] [CrossRef]
  113. Tang, Q.; Wen, H.; Hu, H.; Chen, X.; Xu, S.; Fan, L.; Liu, L.; Li, J. Circ_0070203 Promotes Epithelial-Mesenchymal Transition in Ovarian Serous Cystadenocarcinoma through MiR-370-3p/TGFβR2 Axis. Recent Pat. Anticancer Drug Discov. 2023, 19, 233–246. [Google Scholar] [CrossRef]
  114. Lv, J.; Zhang, Y.; Yang, M.; Qiao, L.; Wang, H.; Jiang, H.; Fu, M.; Qin, J.; Xu, S. Hsa_circ_0013561 Promotes Epithelial-Mesenchymal Transition and Tumor Progression by Regulating ANXA2 via MiR-23b-3p in Ovarian Cancer. Cancer Gene Ther. 2024, 31, 108–118. [Google Scholar] [CrossRef] [PubMed]
  115. Zhou, J.; Dong, Z.N.; Qiu, B.Q.; Hu, M.; Liang, X.Q.; Dai, X.; Hong, D.; Sun, Y.F. CircRNA FGFR3 Induces Epithelial-Mesenchymal Transition of Ovarian Cancer by Regulating MiR-29a-3p/E2F1 Axis. Aging 2020, 12, 14080–14091. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, F.; Xu, Y.; Ye, W.; Jiang, J.; Wu, C. Circular RNA S-7 Promotes Ovarian Cancer EMT via Sponging MiR-641 to up-Regulate ZEB1 and MDM2. Biosci. Rep. 2020, 40, BSR20200825. [Google Scholar] [CrossRef]
  117. Wang, S.; Li, Z.; Zhu, G.; Hong, L.; Hu, C.; Wang, K.; Cui, K.; Hao, C. RNA-Binding Protein IGF2BP2 Enhances Circ_0000745 Abundancy and Promotes Aggressiveness and Stemness of Ovarian Cancer Cells via the MicroRNA-3187-3p/ERBB4/PI3K/AKT Axis. J. Ovarian Res. 2021, 14, 154. [Google Scholar] [CrossRef]
  118. Huang, K.; Liu, D.; Su, C. Circ_0007841 Accelerates Ovarian Cancer Development through Facilitating MEX3C Expression by Restraining MiR-151-3p Activity. Aging 2021, 13, 12058–12066. [Google Scholar] [CrossRef] [PubMed]
  119. Ma, H.; Qu, S.; Zhai, Y.; Yang, X. Circ_0025033 Promotes Ovarian Cancer Development via Regulating the Hsa_miR-370-3p/SLC1A5 Axis. Cell. Mol. Biol. Lett. 2022, 27, 94. [Google Scholar] [CrossRef]
  120. Li, X.; Jiang, X.; Lu, J.; Lin, Y.; Jiang, L.; Li, Y.; Wan, F.; Wang, C. CircCERS6 Suppresses the Development of Epithelial Ovarian Cancer Through Mediating MiR-630/RASSF8. Biochem. Genet. 2022, 60, 2611–2629. [Google Scholar] [CrossRef]
  121. Wu, Y.; Xu, M.; Feng, Z.; Wu, H.; Wu, J.; Ha, X.; Wu, Y.; Chen, S.; Xu, F.; Wen, H.; et al. AUF1-Induced Circular RNA Hsa_circ_0010467 Promotes Platinum Resistance of Ovarian Cancer through MiR-637/LIF/STAT3 Axis. Cell. Mol. Life Sci. 2023, 80, 256. [Google Scholar] [CrossRef]
  122. Dai, C.; Dai, S.Y.; Gao, Y.; Yan, T.; Zhou, Q.Y.; Liu, S.-j.; Liu, X.; Deng, D.N.; Wang, D.H.; Qin, Q.F.; et al. Circ_0078607 Increases Platinum Drug Sensitivity via MiR-196b-5p/GAS7 Axis in Ovarian Cancer. Epigenetics 2023, 18, 2175565. [Google Scholar] [CrossRef]
  123. Gao, Y.; Huang, Y. Circ_0007841 Knockdown Confers Cisplatin Sensitivity to Ovarian Cancer Cells by Down-Regulation of NFIB Expression in a MiR-532-5p-Dependent Manner. J. Chemother. 2023, 35, 117–130. [Google Scholar] [CrossRef] [PubMed]
  124. Wei, L.; He, W.; Zhao, H.; Zhao, P. Circ_0026123 Promotes Cisplatin Resistance and Progression of Ovarian Cancer by Upregulating RAB1A through Sequestering MiR-543. Anticancer Drugs 2022, 33, 1069–1080. [Google Scholar] [CrossRef] [PubMed]
  125. You, J.; Han, Y.; Qiao, H.; Han, Y.; Lu, X.; Lu, Y.; Wang, X.; Kai, H.; Zheng, Y. Hsa_circ_0063804 Enhances Ovarian Cancer Cells Proliferation and Resistance to Cisplatin by Targeting MiR-1276/CLU Axis. Aging 2022, 14, 4699–4713. [Google Scholar] [CrossRef]
  126. Yin, Y.; Li, J.; Rong, J.; Zhang, B.; Wang, X.; Han, H. Circ_0067934 Reduces JNK Phosphorylation through a MicroRNA-545-3p/PPA1 Axis to Enhance Tumorigenesis and Cisplatin Resistance in Ovarian Cancer. Immunopharmacol. Immunotoxicol. 2022, 44, 261–274. [Google Scholar] [CrossRef]
  127. Luo, Y.; Gui, R. Circulating Exosomal Circfoxp1 Confers Cisplatin Resistance in Epithelial Ovarian Cancer Cells. J. Gynecol. Oncol. 2020, 31, e75. [Google Scholar] [CrossRef]
  128. Sheng, H.; Wang, X. Knockdown of Circ-PIP5K1A Overcomes Resistance to Cisplatin in Ovarian Cancer by MiR-942-5p/NFIB Axis. Anticancer Drugs 2023, 34, 214–226. [Google Scholar] [CrossRef]
  129. Li, H.; Lin, R.; Zhang, Y.; Zhu, Y.; Huang, S.; Lan, J.; Lu, N.; Xie, C.; He, S.; Zhang, W. N6-Methyladenosine-Modified CircPLPP4 Sustains Cisplatin Resistance in Ovarian Cancer Cells via PIK3R1 Upregulation. Mol. Cancer 2024, 23, 5. [Google Scholar] [CrossRef]
  130. 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. Onco. Targets Ther. 2020, 13, 13211–13224. [Google Scholar] [CrossRef]
  131. Huang, H.; Yan, L.; Zhong, J.; Hong, L.; Zhang, N.; Luo, X. Circ_0025033 Deficiency Suppresses Paclitaxel Resistance and Malignant Development of Paclitaxel-Resistant Ovarian Cancer Cells by Modulating the MiR-532-3p/FOXM1 Network. Immunopharmacol. Immunotoxicol. 2022, 44, 275–286. [Google Scholar] [CrossRef]
  132. Zhu, J.; Luo, J.-e.; Chen, Y.; Wu, Q. Circ_0061140 Knockdown Inhibits Tumorigenesis and Improves PTX Sensitivity by Regulating MiR-136/CBX2 Axis in Ovarian Cancer. J. Ovarian Res. 2021, 14, 136. [Google Scholar] [CrossRef]
  133. Ying, H.; Zhao, R.; Yu, Q.; Zhang, K.; Deng, Q. CircATL2 Enhances Paclitaxel Resistance of Ovarian Cancer via Impacting MiR-506-3p/NFIB Axis. Drug Dev. Res. 2022, 83, 512–524. [Google Scholar] [CrossRef]
  134. Wei, S.; Qi, L.; Wang, L. Overexpression of Circ-CELSR1 Facilitates Paclitaxel Resistance of Ovarian Cancer by Regulating MiR-149-5p/SIK2 Axis. Anticancer Drugs 2021, 32, 496–507. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, S.; Cheng, J.; Quan, C.; Wen, H.; Feng, Z.; Hu, Q.; Zhu, J.; Huang, Y.; Wu, X. CircCELSR1 (Hsa_circ_0063809) Contributes to Paclitaxel Resistance of Ovarian Cancer Cells by Regulating FOXR2 Expression via MiR-1252. Mol. Ther. Nucleic Acids 2020, 19, 718–730. [Google Scholar] [CrossRef] [PubMed]
  136. Zheng, Y.; Li, Z.; Yang, S.; Wang, Y.; Luan, Z. CircEXOC6B Suppresses the Proliferation and Motility and Sensitizes Ovarian Cancer Cells to Paclitaxel Through MiR-376c-3p/FOXO3 Axis. Cancer Biother. Radiopharm. 2022, 37, 802–814. [Google Scholar] [CrossRef]
  137. Huang, C.; Qin, L.; Chen, S.; Huang, Q. CircSETDB1 Contributes to Paclitaxel Resistance of Ovarian Cancer Cells by Sponging MiR-508-3p and Regulating ABCC1 Expression. Anticancer Drugs 2023, 34, 395–404. [Google Scholar] [CrossRef]
  138. Chen, Y.-y.; Tai, Y. chun Hsa_circ_0006404 and Hsa_circ_0000735 Regulated Ovarian Cancer Response to Docetaxel Treatment via Regulating P-GP Expression. Biochem. Genet. 2022, 60, 395–414. [Google Scholar] [CrossRef] [PubMed]
  139. Xu, X.; Guan, R.; Gong, K.; Xie, H.; Shi, L. Circ_FURIN Knockdown Assuages Testosterone-Induced Human Ovarian Granulosa-like Tumor Cell Disorders by Sponging MiR-423-5p to Reduce MTM1 Expression in Polycystic Ovary Syndrome. Reprod. Biol. Endocrinol. 2022, 20, 32. [Google Scholar] [CrossRef] [PubMed]
  140. Gan, X.; Zhu, H.; Jiang, X.; Obiegbusi, S.C.; Yong, M.; Long, X.; Hu, J. CircMUC16 Promotes Autophagy of Epithelial Ovarian Cancer via Interaction with ATG13 and MiR-199a. Mol. Cancer 2020, 19, 45. [Google Scholar] [CrossRef]
  141. Zhang, Z.; Zhu, H.; Hu, J. CircRAB11FIP1 Promoted Autophagy Flux of Ovarian Cancer through DSC1 and MiR-129. Cell Death Dis. 2021, 12, 219. [Google Scholar] [CrossRef]
  142. Qu, D.D.; Zou, X.; Liu, Z.L. Propofol Modulates Glycolysis Reprogramming of Ovarian Tumor via Restraining Circular RNA-Zinc Finger RNA-Binding Protein/MicroRNA-212-5p/Superoxide Dismutase 2 Axis. Bioengineered 2022, 13, 11881–11892. [Google Scholar] [CrossRef]
  143. Zhang, W.; Wang, Y.; Liu, Y.; Liu, C.; Wang, Y.; He, L.; Cheng, X.; Peng, Y.; Xia, L.; Wu, X.; et al. NFIB Facilitates Replication Licensing by Acting as a Genome Organizer. Nat. Commun. 2023, 14, 5076. [Google Scholar] [CrossRef] [PubMed]
  144. Yoo, H.C.; Park, S.J.; Nam, M.; Kang, J.; Kim, K.; Yeo, J.H.; Kim, J.K.; Heo, Y.; Lee, H.S.; Lee, M.Y.; et al. A Variant of SLC1A5 Is a Mitochondrial Glutamine Transporter for Metabolic Reprogramming in Cancer Cells. Cell Metab. 2020, 31, 267–283. [Google Scholar] [CrossRef] [PubMed]
  145. Liao, G.-B.; Li, X.Z.; Zeng, S.; Liu, C.; Yang, S.M.; Yang, L.; Hu, C.J.; Bai, J.Y. Regulation of the Master Regulator FOXM1 in Cancer. Cell Commun. Signal. 2018, 16, 57. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, J.; Liao, J.; Liu, J.; Meng, C.; Liu, B.; Shao, C. Analysis of Correlation between Rab1A Expression and Its Prognosis in Cancers: A Meta-Analysis. Clin. Lab. 2024, 70, 1. [Google Scholar] [CrossRef]
  147. Bertorello, A.M.; Zhu, J.K. SIK1/SOS2 Networks: Decoding Sodium Signals via Calcium-Responsive Protein Kinase Pathways. Pflug. Arch. 2009, 458, 613–619. [Google Scholar] [CrossRef]
  148. Green, D.R.; Llambi, F. Cell Death Signaling. Cold Spring Harb. Perspect. Biol. 2015, 7, a006080. [Google Scholar] [CrossRef] [PubMed]
  149. Hu, D.; Du, J.H.; Xing, Y.J.; Cheng, Y.M.; He, R.F.; Liang, X.L.; Li, H.L.; Yang, Y.X. SIK2: A Critical Glucolipid Metabolic Reprogramming Regulator and Potential Target in Ovarian Cancer. J. Obstet. Gynaecol. Res. 2023, 49, 2000–2009. [Google Scholar] [CrossRef]
  150. Karin, M.; Baud, V. Is NF-KappaB a Good Target for Cancer Therapy? Hopes and Pitfalls. Nat. Rev. Drug Discov. 2009, 8, 33–40. [Google Scholar]
  151. Brzozowa-Zasada, M.; Piecuch, A.; Michalski, M.; Segiet, O.; Kurek, J.; Harabin-Słowińska, M.; Wojnicz, R. Notch and Its Oncogenic Activity in Human Malignancies. Eur. Surg. 2017, 49, 199–209. [Google Scholar] [CrossRef]
  152. Zhang, Y.; Li, Y.; Wang, Q.; Su, B.; Xu, H.; Sun, Y.; Sun, P.; Li, R.; Peng, X.; Cai, J. Role of RASA1 in Cancer: A Review and Update (Review). Oncol. Rep. 2020, 44, 2386–2396. [Google Scholar] [CrossRef]
  153. Damia, G.; Broggini, M. Platinum Resistance in Ovarian Cancer: Role of DNA Repair. Cancers 2019, 11, 119. [Google Scholar] [CrossRef] [PubMed]
  154. Zhu, L.; Chen, L. Progress in Research on Paclitaxel and Tumor Immunotherapy. Cell. Mol. Biol. Lett. 2019, 24, 40. [Google Scholar] [CrossRef] [PubMed]
  155. Yélamos, J.; Moreno-Lama, L.; Jimeno, J.; Ali, S.O. Immunomodulatory Roles of PARP-1 and PARP-2: Impact on PARP-Centered Cancer Therapies. Cancers 2020, 12, 392. [Google Scholar] [CrossRef]
  156. Irusta, G. Roads to the Strategic Targeting of Ovarian Cancer Treatment. Reproduction 2021, 161, R1–R11. [Google Scholar] [CrossRef]
  157. Nusse, R.; Clevers, H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985–999. [Google Scholar] [CrossRef] [PubMed]
  158. Beilerli, A.; Begliarzade, S.; Sufianov, A.; Ilyasova, T.; Liang, Y.; Beylerli, O. Circulating CiRS-7 as a Potential Non-Invasive Biomarker for Epithelial Ovarian Cancer: An Investigative Study. Noncoding RNA Res. 2022, 7, 197–204. [Google Scholar] [CrossRef]
  159. Liu, N.; Zhang, J.; Zhang, L.Y.; Wang, L. CircHIPK3 Is Upregulated and Predicts a Poor Prognosis in Epithelial Ovarian Cancer. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3713–3718. [Google Scholar] [CrossRef]
  160. Ning, L.; Long, B.; Zhang, W.; Yu, M.; Wang, S.; Cao, D.; Yang, J.; Shen, K.; Huang, Y.; Lang, J. Circular RNA Profiling Reveals CircEXOC6B and CircN4BP2L2 as Novel Prognostic Biomarkers in Epithelial Ovarian Cancer. Int. J. Oncol. 2018, 53, 2637–2646. [Google Scholar] [CrossRef] [PubMed]
  161. Ning, L.; Lang, J.; Long, B.; Wu, L. Diagnostic Value of CircN4BP2L2 in Type I and Type II Epithelial Ovarian Cancer. BMC Cancer 2022, 22, 1210. [Google Scholar] [CrossRef]
  162. Gao, Y.; Zhang, C.; Liu, Y.; Wang, M. Circular RNA Profiling Reveals CircRNA1656 as a Novel Biomarker in High Grade Serous Ovarian Cancer. Biosci. Trends 2019, 13, 204–211. [Google Scholar] [CrossRef]
  163. Ge, L.; Sun, Y.; Shi, Y.; Liu, G.; Teng, F.; Geng, Z.; Chen, X.; Xu, H.; Xu, J.; Jia, X. Plasma CircRNA Microarray Profiling Identifies Novel CircRNA Biomarkers for the Diagnosis of Ovarian Cancer. J. Ovarian Res. 2022, 15, 58. [Google Scholar] [CrossRef] [PubMed]
  164. Zhang, N.; Yang, Z.; Jin, Y.; Cheng, S.; Yang, J.; Wang, Y. Low Expression of Circular RNA Hsa_Circ_0078607 Predicts Poor Prognosis in High-Grade Serous Ovarian Cancer. Cancer Manag. Res. 2021, 13, 2877–2883. [Google Scholar] [CrossRef] [PubMed]
  165. Zou, T.; Wang, P.L.; Gao, Y.; Liang, W.T. Circular RNA-LARP4 Is Lower Expressed and Serves as a Potential Biomarker of Ovarian Cancer Prognosis. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7178–7182. [Google Scholar]
  166. Bao, L.; Zhong, J.; Pang, L. Upregulation of Circular RNA VPS13C-Has-Circ-001567 Promotes Ovarian Cancer Cell Proliferation and Invasion. Cancer Biother. Radiopharm. 2019, 34, 110–118. [Google Scholar] [CrossRef] [PubMed]
  167. Chen, S.; Wu, W.; Li, Q.H.; Xie, B.M.; Shen, F.; Du, Y.P.; Zong, Z.H.; Wang, L.L.; Wei, X.Q.; Zhao, Y. Circ-NOLC1 Promotes Epithelial Ovarian Cancer Tumorigenesis and Progression by Binding ESRP1 and Modulating CDK1 and RhoA Expression. Cell Death Discov. 2021, 7, 22. [Google Scholar] [CrossRef]
  168. Pei, C.; Wang, H.; Shi, C.; Zhang, C.; Wang, M. CircRNA Hsa_circ_0013958 May Contribute to the Development of Ovarian Cancer by Affecting Epithelial-Mesenchymal Transition and Apoptotic Signaling Pathways. J. Clin. Lab. Anal. 2020, 34, e23292. [Google Scholar] [CrossRef]
  169. Hu, Y.; Zhu, Y.; Zhang, W.; Lang, J.; Ning, L. Utility of Plasma CircBNC2 as a Diagnostic Biomarker in Epithelial Ovarian Cancer. Onco. Targets Ther. 2019, 12, 9715–9723. [Google Scholar] [CrossRef]
  170. Wang, W.; Wang, J.; Zhang, X.; Liu, G. Serum CircSETDB1 Is a Promising Biomarker for Predicting Response to Platinum-Taxane-Combined Chemotherapy and Relapse in High-Grade Serous Ovarian Cancer. Onco Targets Ther. 2019, 12, 7451–7457. [Google Scholar] [CrossRef]
  171. Chen, Y.; Ye, X.; Xia, X.; Lin, X. Circular RNA ABCB10 Correlates with Advanced Clinicopathological Features and Unfavorable Survival, and Promotes Cell Proliferation While Reduces Cell Apoptosis in Epithelial Ovarian Cancer. Cancer Biomark. 2019, 26, 151–161. [Google Scholar] [CrossRef]
  172. Esquela-Kerscher, A.; Slack, F.J. Oncomirs—MicroRNAs with a Role in Cancer. Nat. Rev. Cancer 2006, 6, 259–269. [Google Scholar] [CrossRef]
  173. Xing, Y.; Wang, Z.; Lu, Z.; Xia, J.; Xie, Z.; Jiao, M.; Liu, R.; Chu, Y. MicroRNAs: Immune Modulators in Cancer Immunotherapy. Immunother. Adv. 2021, 1, ltab006. [Google Scholar] [CrossRef] [PubMed]
  174. Bertoli, G.; Cava, C.; Castiglioni, I. Micrornas: New Biomarkers for Diagnosis, Prognosis, Therapy Prediction and Therapeutic Tools for Breast Cancer. Theranostics 2015, 5, 1122–1143. [Google Scholar] [CrossRef]
  175. Holdt, L.M.; Kohlmaier, A.; Teupser, D. Circular RNAs as Therapeutic Agents and Targets. Front. Physiol. 2018, 9, 1262. [Google Scholar] [CrossRef]
  176. 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]
  177. Wesselhoeft, R.A.; Kowalski, P.S.; Anderson, D.G. Engineering Circular RNA for Potent and Stable Translation in Eukaryotic Cells. Nat. Commun. 2018, 9, 2629. [Google Scholar] [CrossRef] [PubMed]
  178. Rossbach, O. Artificial Circular RNA Sponges Targeting MicroRNAs as a Novel Tool in Molecular Biology. Mol. Ther. Nucleic Acids 2019, 17, 452–454. [Google Scholar] [CrossRef]
  179. Yang, Q.; Li, F.; He, A.T.; Yang, B.B. Circular RNAs: Expression, Localization, and Therapeutic Potentials. Mol. Ther. 2021, 29, 1683–1702. [Google Scholar] [CrossRef] [PubMed]
  180. Li, S.; Li, X.; Xue, W.; Zhang, L.; Yang, L.Z.; Cao, S.M.; Lei, Y.N.; Liu, C.X.; Guo, S.K.; Shan, L.; et al. Screening for Functional Circular RNAs Using the CRISPR–Cas13 System. Nat. Methods 2021, 18, 51–59. [Google Scholar] [CrossRef]
  181. Meganck, R.M.; Borchardt, E.K.; Castellanos Rivera, R.M.; Scalabrino, M.L.; Wilusz, J.E.; Marzluff, W.F.; Asokan, A. Tissue-Dependent Expression and Translation of Circular RNAs with Recombinant AAV Vectors In Vivo. Mol. Ther. Nucleic Acids 2018, 13, 89–98. [Google Scholar] [CrossRef]
  182. Ma, J.; Du, W.W.; Zeng, K.; Wu, N.; Fang, L.; Lyu, J.; Yee, A.J.; Yang, B.B. An Antisense Circular RNA CircSCRIB Enhances Cancer Progression by Suppressing Parental Gene Splicing and Translation. Mol. Ther. 2021, 29, 2754–2768. [Google Scholar] [CrossRef]
  183. Paunovska, K.; Loughrey, D.; Dahlman, J.E. Drug Delivery Systems for RNA Therapeutics. Nat. Rev. Genet. 2022, 23, 265–280. [Google Scholar] [CrossRef] [PubMed]
  184. Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H.; Sun, D. Engineering Exosomes as Refined Biological Nanoplatforms for Drug Delivery. Acta Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef] [PubMed]
  185. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, L.; Liao, Y.; Tang, L. MicroRNA-34 Family: A Potential Tumor Suppressor and Therapeutic Candidate in Cancer. J. Exp. Clin. Cancer Res. 2019, 38, 53. [Google Scholar] [CrossRef]
  187. Beg, M.S.; Brenner, A.J.; Sachdev, J.; Borad, M.; Kang, Y.K.; Stoudemire, J.; Smith, S.; Bader, A.G.; Kim, S.; Hong, D.S. Phase I Study of MRX34, a Liposomal MiR-34a Mimic, Administered Twice Weekly in Patients with Advanced Solid Tumors. Investig. New Drugs 2017, 35, 180–188. [Google Scholar] [CrossRef]
Figure 1. Biogenesis and function of miRNAs and circRNAs. Important components of the biogenesis of miRNAs and circRNAs, as described in more detail in the text, are depicted. As shown, EIciRNAs and ciRNAs promote the transcription by RNA polymerase II (RNA Pol II). MiRNAs bind to and inhibits the translation of mRNA, while the binding of circRNAs to miRNAs abrogates the inhibition, resulting in the translation of mRNAs by the ribosome.
Figure 1. Biogenesis and function of miRNAs and circRNAs. Important components of the biogenesis of miRNAs and circRNAs, as described in more detail in the text, are depicted. As shown, EIciRNAs and ciRNAs promote the transcription by RNA polymerase II (RNA Pol II). MiRNAs bind to and inhibits the translation of mRNA, while the binding of circRNAs to miRNAs abrogates the inhibition, resulting in the translation of mRNAs by the ribosome.
Ncrna 10 00051 g001
Figure 2. CircRNA/miRNA interactions and their involvement in different signaling pathways.
Figure 2. CircRNA/miRNA interactions and their involvement in different signaling pathways.
Ncrna 10 00051 g002
Table 1. Interactions of circRNAs and miRNAs and their targets in EOC, arranged according to their function.
Table 1. Interactions of circRNAs and miRNAs and their targets in EOC, arranged according to their function.
circRNAsmiRNAsTargetsFunctionRef.
* Increasing** Decreasing
circ_0000144miR-610ELK3progression-[45]
circ_0000471miR-135bdusp5 progression[46]
circ_0001445miR-576SFRP1-progression[47]
circ_0004712miR-331FZD4progression-[48]
circ_0007444miR-23aDicer1-progression[49]
circ_0007615miR-874TUBB3progression-[50]
circ_0007874miR-760SOCS3-progression[51]
circ_0009910miR-145-progression-[52]
circ_0015756miR-145PSAT1progression-[53]
circ_0021573miR-936CUL4Bprogression-[54]
circ_0072995miR-122SLC1A5progression-[55]
circ_0078607miR-32SIK1-progression[56]
circ_0025033miR-184LSM4progression-[57]
circ_0061140miR-361RAB1Aprogression-[58]
miR-761LETM1progression-[59]
circ_0070203miR-518aFas-progression[60]
circ_ATRNL1miR-152-progression-[61]
circ_BNC2miR-223LARP4-progression[62]
miR-223FBXW7-progression[63]
circ_AHNAKmiR-28EIF2B5-progression[64]
circ_CDR1miR-135b--progression[65]
circ_CRIM1miR-383ZEB2progression-[66]
circ_EPSTI1miR-942-progression-[67]
circ_EXOC6BmiR-421RUS1-progression[68]
circ_FAM53BmiR-646VAMP2progression-[69]
miR-647MDM2progression-[69]
circ_FBXO7miR-96-5pMTSS1/Wntprogression-[70]
circ_ITCHmiR-145RASA1-progression[71]
circ_MFN2miR-198CUL4Bprogression, glycolysis-[72]
circ_MUC16miR-1182S100Bprogression-[73]
circ_NFIXmiR-647JAK1/STAT3progression-[74]
circ_PGAM1miR-542CDC5L/PEAK1progression-[75]
circ_PHC3miR-497SOX9progression-[76]
circ_PIP5K1AmiR-661IGFBP5progression-[77]
circ-PTK2miR-639FOXC1progression-[78]
circ_RNF144BmiR-342FBXL11progression-[79]
circ_RhoCmiR-302eVEGFAprogression-[80]
circ_SETDB1miR-129MAP3K3progression-[81]
circ_WHSC1miR-145MUC1progression-[82]
circ_WHSC1miR-1182hTERTprogression-[82]
circ_ZNF608miR-152-progression-[61]
circ_UBAP2miR-144--progression[83]
circ_PLEKHM3miR-9BRCA1/DNAJB6/KLF4/AKT1-progression[84]
miR-320aSMG1-progression[85]
circ_MYLKmiR-652-progression-[86]
circ_0001741miR-188FOXN2-proliferation[87]
circ_0004390miR-198-proliferation-[88]
circ_ITCHmiR-10a--proliferation[89]
circ_PVT1miR-149-proliferation-[90]
circ_UBAP2miR-382PRPF8proliferation-[91]
circ_ASXL1miR-320dRACGAP1proliferation, migration-[92]
circ_CDK17miR-22CD147proliferation, migration-[93]
circ_0001535miR-593PTEN-proliferation, migration [94]
circ_0000554miR-567-proliferation,
invasion
-[95]
circ_0013958miR-637PLXNB2proliferation, invasion-[96]
circ_ABCB10miR-1271Capn4/Wntproliferation, invasion-[97]
circ_MTO1miR-182KLF15-proliferation, invasion[98]
circ_RHOBTB3miR-23a--proliferation, invasion[99]
circ_9119miR-21PTEN/Akt-cell viability[100]
circ_VPS13CmiR-145MEK/ERKcell cycle, motility-[101]
circ_PLOD2miR-378-propagation-[102]
circ_0051240miR-637KLK4migration, invasion-[103]
circ_CSPP1miR-1236-invasion, migration-[104]
circ_NFIXmiR-518aTRIM44angiogenesis-[105]
circ_0026123miR-124EZH2proliferation, metastasis-[106]
circ_CELSR1miR-598BRD4proliferation, metastasis-[107]
circ_ATRNL1miR-378Smad4-angiogenesis, metastasis[108]
circ_ATP2B4miR-532SREBF1metastasis-[109]
circ_IFNGR2miR-378ST5metastasis-[110]
circ_0002711miR-1244ROCK1cell growth, glycolysis-[111]
circ_0005585 miR-23a/b/ 15a/15b/16ESRP1colonization-[112]
circ_0070203miR-370TGFβR2EMT-[113]
circ_0013561miR-23bANXA2EMT-[114]
circ_FGFR3miR-29aE2F1EMT-[115]
circ_S-7miR-641ZEB1, MDM2EMT-[116]
circ_0000745miR-3187ERBB4/PI3K/AKTcell stemness-[117]
circ_0007841miR-151MEX3Cdevelopment-[118]
circ_002503miR-370SLC1A5development-[119]
circ_CERS6miR-630RASSF8-development[120]
circ_0010467miR-637LIF/STAT3platinum resistance-[121]
circ_0070203miR-196bGAS7platinum sensitivity-[122]
circ_0007841miR-532NFIBcisplatin resistance-[123]
circ_0026123miR-543RAB1Acisplatin resistance-[124]
circ_0063804miR-1276CLUcisplatin resistance-[125]
circ_0067934miR-545PPA1-cisplatin resistance[126]
circ_Foxp1miR-22CEBPGcisplatin resistance-[127]
miR-150FMNL3cisplatin resistance-[127]
circ_NFIXmiR-942NFIBcisplatin resistance-[128]
circ_PLPP4miR-136PIK3R1cisplatin resistance-[129]
circ_0000714miR-370RAB17, CDK6/RBpaclitaxel resistance-[130]
circ_0025033miR-532FOXM1paclitaxel resistance-[131]
circ_0061140miR-136CBX2-paclitaxel sensitivity[132]
circ_ATL2miR-506NFIBpaclitaxel resistance-[133]
circ_CELSR1miR-149SIK2paclitaxel resistance-[134]
miR-1252FOXR2paclitaxel resistance-[135]
circ_EXOC6BmiR-376cFOXO3-paclitaxel sensitivity[136]
circ_SETDB1miR-508ABCC1paclitaxel resistance-[137]
circ_0000735miR-526bp-GPdocetaxel resistance-[138]
circ_0006404miR-346p-GP-docetaxel[138]
circ_FURINmiR-423MTM1testosterone effect-[139]
circ_MUC16miR-199aATG13autophagy-[140]
circ_RAB11FIP1 miR-129DSC1autophagy-[141]
circ_zinc fingermiR-212superoxide dismutase 2glycolysis-[142]
Via sponging of miRNAs by circRNAs, the cancer features continued to be either increased * or decreased **. Abbreviations are explained at the end of the article.
Table 2. Interactions of circRNAs and miRNAs and their targets arranged to miRNAs which are sponged by two or more circRNAs.
Table 2. Interactions of circRNAs and miRNAs and their targets arranged to miRNAs which are sponged by two or more circRNAs.
circRNAsmiRNAsTargetsFunctionRef.
* Increasing** Decreasing
circ_CDK17miR-22CD147proliferation, migration-[93]
circ_Foxp1 CEBPGcisplatin resistance-[127]
circ_0007444miR-23aDicer1-progression[49]
circ_RHOBTB3 --proliferation, invasion[99]
circ_0005585 miR-23a/bESRP1colonization-[112]
circ_0013561miR-23bANXA2EMT-[114]
circ_RAB11FIP1 miR-129DSC1autophagy-[141]
circ_SETDB1 MAP3K3progression-[81]
circ_0000471miR-135bdusp5-progression [46]
circ_CDR1 --progression[65]
circ_0061140miR-136CBX2-paclitaxel sensitivity[132]
circ_PLPP4 PIK3R1cisplatin resistance-[129]
circ_0009910miR-145-progression-[52]
circ_0015756 PSAT1progression-[53]
circ_ITCH RASA1-progression[71]
circ_VPS13C MEK/ERKcell cycle, motility-[101]
circ_WHSC1 MUC1progression-[82]
circ_CELSR1miR-149SIK2paclitaxel resistance-[134]
circ_PVT1 -proliferation-[90]
circ_ATRNL1miR-152-progression-[61]
circ_ZNF608 -progression-[61]
circ_0004390miR-198-proliferation-[88]
circ_MFN2 CUL4Bprogression, glycolysis-[72]
circ_BNC2miR-223LARP4-progression[62]
circ_BNC2 FBXW7-progression[63]
circ_0025033miR-370SLC1A5 development-[119]
circ_0070203 TGFβR2EMT-[113]
circ_0000714 RAB17, CDK6/RBpaclitaxel resistance-[130]
circ_ATRNL1miR-378Smad4-angiogenesis, metastasis[108]
circ_IFNGR2 ST5metastasis-[110]
circ_PLOD2 -propagation-[102]
circ_0078607miR-518aFas-progression[60]
circ_NFIX TRIM44angiogenesis-[105]
circ_0007841miR-532NFIBcisplatin-[123]
circ_0025033 FOXM1paclitaxel resistance-[131]
circ_ATP2B4 SREBF1metastasis-[109]
circ_0010467miR-637LIF/STAT3platinum resistance-[121]
circ_0013958 PLXNB2proliferation, invasion-[96]
circ_0051240 KLK4migration, invasion-[103]
circ_EPSTI1miR-942-progression-[67]
circ_PIP5K1A NFIBcisplatin resistance-[128]
circ_MUC16miR-1182S100Bprogression-[73]
circ_WHSC1 hTERTprogression-[82]
By sponging of miRNAs by circRNAs, the cancer features continued to be either increased * or inhibited **. Abbreviations are explained at the end of the article.
Table 3. CircRNAs as biomarkers and their relevance in prognosis.
Table 3. CircRNAs as biomarkers and their relevance in prognosis.
circRNAsLevelsFunctionAssociationsOCRef.
StimulationInhibition Subtype
circ_RS-7up--FIGO stage EOC[158]
lymph node
distant metastasis
circ_HIPK3upproliferation, migration apoptosis FIGO stageEOC[159]
invasion lymph node
circ_EXOC6Bup--FIGO stageEOC[160]
circ_N4BP2L2up--FIGO stageEOC[161]
circ_RNA1656down--FIGO stageHGSOC [162]
circ_0003972down---EOC[163]
circ_0007288down--lymph nodeEOC [163]
circ_0078607downapoptosisproliferation -HGSOC[164]
circLARP4down--FIGO, lymph node EOC[165]
circ-001567 proliferation apoptosis E-/N-cadherin EOC[166]
circ-NOLC1upproliferation, migration-FIGO stageEOC[167]
invasion differentiation
circ_0013958upproliferation, migration-FIGO stage EOC[168]
invasion lymph node
circ_BNC2down--FIGO stageEOC[169]
lymph node
circ_SETDB1uprelapse-FIGO stage HGSOC[170]
lymph node
circ-ABCB10upproliferationapoptosisFIGO stageEOC[171]
differentiation
tumor size
Up, upregulated; down, downregulated; HGSOC, high-grade ovarian serous carcinoma.
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

Schwarzenbach, H. Interplay of microRNAs and circRNAs in Epithelial Ovarian Cancer. Non-Coding RNA 2024, 10, 51. https://doi.org/10.3390/ncrna10050051

AMA Style

Schwarzenbach H. Interplay of microRNAs and circRNAs in Epithelial Ovarian Cancer. Non-Coding RNA. 2024; 10(5):51. https://doi.org/10.3390/ncrna10050051

Chicago/Turabian Style

Schwarzenbach, Heidi. 2024. "Interplay of microRNAs and circRNAs in Epithelial Ovarian Cancer" Non-Coding RNA 10, no. 5: 51. https://doi.org/10.3390/ncrna10050051

APA Style

Schwarzenbach, H. (2024). Interplay of microRNAs and circRNAs in Epithelial Ovarian Cancer. Non-Coding RNA, 10(5), 51. https://doi.org/10.3390/ncrna10050051

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