Open Access

Underlying metastasis mechanism and clinical application of exosomal circular RNA in tumors (Review)

  • Authors:
    • Xuezhi Wei
    • Yaxing Shi
    • Zhijun Dai
    • Pei Wang
    • Xin Meng
    • Bo Yin
  • View Affiliations

  • Published online on: January 28, 2021     https://doi.org/10.3892/ijo.2021.5179
  • Pages: 289-297
  • Copyright: © Wei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Circular RNA (circRNA) is a long non‑coding RNA molecule with a closed loop structure lacking a 5'cap and 3'tail. circRNA is stable, difficult to cleave and resistant to RNA exonuclease or RNase R degradation. circRNA molecules have several clinical applications, especially in tumors. For instance, circRNA may be used for non‑invasive diagnosis, therapy and prognosis. Exosomes play a crucial role in the development of tumors. Exosomal circRNA in particular has led to increased research interest into tumorigenesis and tumor progression. Additionally, exosomal circRNA plays a role in cell‑cell communication. Exosomal circRNA facilitates tumor metastasis by altering the tumor microenvironment and the pre‑metastatic niche. Additionally, studies have revealed the mechanism by which exosomal circRNA affects malignant progression through signal transduction. Moreover, exosomal circRNA promotes tumor metastasis by regulating gene expression, RNA transcription and protein translation. In this review, the biological features and clinical application of exosomal circRNA are described, highlighting the underlying mechanisms through which they regulate tumor metastasis. The application of circRNA as clinical diagnostic biomarkers and in the development of novel therapeutic strategies is also discussed.

1. Introduction

Circular RNA (circRNA) is a type of endogenous non-coding RNA with diverse biological functions in tumors (1). circRNA molecules are involved in the initiation of tumorigenesis and tumor metastasis. Differential expression of circRNA occurs in diverse stages of tumor progression (2). circRNA can sponge microRNA (miRNA/miR) molecules and affect mRNA translation into proteins (3).

Exosomes contain various cargos, including proteins, lipids, DNA and RNA. Exosomes mediate intercellular communication by transferring cargo between cells, including cancer cells. The exosomal contents are transferred to the recipient cell, inducing changes in gene expression and alterations to the tumor microenvironment (TME) (4,5). This process is associated with tumor metastasis. Moreover, circRNA molecules are abundant and stable in exosomes (6). Exosomal circRNA has several extracellular functions and plays critical roles following exosome uptake by recipient cells (7). Exosomal circRNA may induce trans-differentiation processes, such as epithelial-to-mesenchymal transition (EMT), macrophage polarization (8,9) and changes to the TME (10,11). These processes often impact tumor metastasis, as remodeling the extracellular microenvironment is a crucial condition for metastasis. The distant movement of malignant cells requires changes to the pre-metastatic niche (12). However, the detailed mechanism underlying the functions of exosomal circRNA in tumor metastasis are unclear. Thus, deciphering the roles of exosomal circRNA may provide insight into the mechanism of tumor progression.

In this review, research advances into the underlying mechanism and clinical application of exosomal circRNA in tumorigenesis and metastasis are discussed. In particular, the present review focuses on the relationship between circRNA, exosomes and the TME. Furthermore, we propose that exosomal circRNA might indirectly regulate the TME, EMT and macrophage polarization, which in turn affects tumor progression. In addition, exosomal circRNA may represent a diagnostic molecular biomarker and clinical target for cancer treatment.

2. Biogenesis and function of circRNA

Sanger et al (13) first identified the structural characteristics of circRNA, which comprise a single-stranded, highly stable and closed loop structure lacking a 5′cap or 3′poly-A tail. Numerous circRNA molecules have been identified in eukaryotic cells and tissues and were found to be constructed from selective exonic and intronic repeats that base-pair to one another (14). The involvement of back-splicing and exon-skip-ping in circRNA formation have been confirmed in vitro and in vivo (15). circRNA back-splicing involves joining a splice donor to an upstream splice acceptor site. Theoretically, exons in the genome can be circularized. There are four types of circRNA molecules, which are categorized according to their method of formation: i) Exon-intron circRNA (Fig. 1A) (16); ii) exonic circRNA (Fig. 1B) (17); iii) circular intronic RNA (Fig. 1C) (18); and iv) transfer RNA (tRNA) intronic circRNA, which is formed by splicing pre-tRNA introns (Fig. 1D) (16,19). However, the most common type of circRNA is exonic circRNA. Recently, with the development of RNA sequencing (RNA-seq) technologies and novel bioinformatics approaches, numerous circRNA molecules have been detected and identified. circRNA is stable, abundant, prevalent and conserved (20). The biological functions of circRNA include acting as miRNA sponges that regulate gene expression, acting as miRNA 'reservoirs' that stabilize miRNA function, modulating gene expression via ribosome-binding proteins and even encoding proteins (21). Some protein-coding genes may actually produce both non-coding circRNA and protein (22). Das et al (23) have observed that circRNA generated from multiple exons can contain different combinations of these exons, due to alternative splicing, although the back-splicing junction sequence is the same. It was predicted that splice variants would be associated with different subsets of target RNA-binding proteins and miRNA and the results of this previous study showed that different circRNA splice variants can have different biological effects (23).

circRNA has several biological characteristics. It is predominantly expressed in the cytoplasm and regulates the expression of target genes by mediating miRNA activity (24). Most circRNA molecules are found at high levels in extracellular body fluids (liquid biopsy), including serum, urine, saliva and cerebrospinal fluid (25). They are highly resistant to RNA exonuclease or RNase R (26). circRNA may encode a protein under a certain conditions; for example, in vitro and in vivo evidence suggests that hsa_circ_0000423 carries an open reading frame that encodes a functional protein in colon cancer (CC) cells (27).

miRNA are small, non-coding RNA molecules that play pivotal roles in the regulation of target gene expression by regulating mRNA degradation or inhibiting translation (28). Previous studies have demonstrated that miRNA molecules are significantly associated with tumor growth, metastasis and progression (29,30). circRNA may also regulate cell signal transduction and RNA transcription via circRNA-miRNA regulatory networks that control target genes (31). Thus, circRNA can represent a switch that controls miRNA. Currently, circRNA is considered an important factor in the regulation of tumorigenesis and metastasis. For instance, hsa_circRNA_102958 expression is significantly increased in colorectal cancer (CRC) tissue and silencing the expression of this circRNA markedly suppresses CRC growth, migration and invasion (32). Moreover, hsa_circRNA_102958 acts as a sponge for miR-585, whereby hsa_circRNA_102958 promotes cell division cycle 25B expression by inhibiting miR-585 activity in CRC cells (32). The circRNA cerebellar degeneration-related protein 1-antisense (CDR1-AS) binds to miR-7 and acts as its molecular sponge (33). miR-7-5p is the mature form of miR-7 and is closely related to the occurrence and development of tumors (34). For example, miR-7 inhibits the ability of gastric cancer cells to divide, migrate and invade, while promoting their apoptosis (35). circRNA circR-7 acts as a miR-7 sponge that inhibits its biological function (36). Thus, circRNA binding to miRNA inhibits its functions and since these functions can be associated with oncogenesis, circRNA could regulate tumor cell differentiation, growth and metastasis indirectly.

The identification of circRNA functions provided a novel molecular mechanism for cancer. circRNA exerts several verified functions. For example, a previous study has demonstrated that circzinc finger protein 609 (circ-ZNF609), an oncogenic circRNA, is significantly upregulated in the human glioma cell lines U87 and U251 (37). In addition, circ-ZNF292 silencing suppresses tube formation by inhibiting glioma cell proliferation and cell cycle progression (38). Based on these findings, it has been proposed that abnormal miRNA or circRNA expression may alter cancer growth. Similarly, recent studies have suggested that circRNA can display tissue-dependent expression in neurodegenerative diseases (39). circRNA is differentially expressed in various tumor tissue types, including breast cancer (40), hepatocellular carcinoma (HCC) (41,42) and CRC (32).

3. Biogenesis and functions of exosomal circRNA

Exosomes are nanoscale (30-150 nm) extracellular vesicles (EVs) of endocytic origin that are shed by most types of cells and circulate in bodily fluids, such as blood, urine, saliva and breast milk (43) They contain a specific cargo of diverse growth factors, proteins, lipids and nucleic acids, including long non-coding RNA and circRNA, which can modulate recipient cell behavior and might be used as biomarkers for diagnosis, such as bladder and breast cancer (44,45). Different of exosomes can carry these cargos from cell to cell and mediate intercellular communication. In cancer progression, exosomal cargos are the link between the disruption of normal tissues and oncogenesis (46). Exosomes can facilitate tumor progression by altering their contents and remodeling the microenvironment, thus accelerating the progression of oncogenic processes (44).

RNA-seq analysis has revealed that circRNA are present in exosomes, representing a novel class of RNA species in exosomes, which might be regulated by the levels of associated miRNA in cells and result in the transfer of biological cargos into recipient cells (47). In addition, a previous study has suggested that the concentration of circRNA in exosomes is greater in cholangiocarcinoma cells than in normal cells (48). To date, >1,000 exosomal circRNA molecules have been identified in human serum. For example, circkelch domain containing 10 was identified in a patient with CRC, and it has been demonstrated that this circRNA could be measured by RNA-seq and further validated individually with quantitative reverse transcription PCR analysis in serum from cancer patients (7). Therefore, exosomal circRNA could be used to screen for tumors using patient serum, representing a new type of diagnostic biomarker for cancer (7). Another study has demonstrated that adipose-derived exosomes mediate the delivery of circRNA and promote the tumorigenesis of HCC (42).

Exosomal circRNA not only induces tumorigenesis of HCC by affecting cell signaling, but also facilitates HCC metastasis. Su et al (49) reported that circRNA CDR1-AS was directly transferred from HCC cells into the surrounding normal cells via exosomes, thereby mediating the proliferative and migratory abilities of surrounding cells. Moreover, it was observed that CDR1-AS could sponge miR-1270 and facilitate the expression of α-fetoprotein, a specific biomarker of HCC (49). Another study by Shen et al (36) has also provided insight into the association between exosomal circRNA and tumor metastasis. Therefore, exosomal circRNA may represent a future therapeutic target for tumor metastasis in clinical applications (36).

Exosomes alter the TME and facilitate tumor metastasis

Tumor metastasis is a complex process that involves genetic mutations and TME changes. Evidence shows that the TME facilitates tumor initiation and metastasis. Indeed, the TME comprises multiple categories of substances, such as T cells, exosomes, tumor-associated macrophages (TAMs), cytokines, fibroblasts, endothelial cells, mesenchymal stem cells (MSCs) and neutrophils, which exert a great influence on cancer growth and generate a pre-metastatic niche favorable to tumor progression (50-52). The pre-metastatic niche is a pre-formed microenvironment made possible by exosomes secreted by the primary tumor site before widespread metastasis (53).

Exosomes are released into the TME and bloodstream, subsequently acting as messengers that impact distant tissues (54). Exosomes release various substances into the extracellular milieu to accelerate tumor cell migration. For example, cancer stem cell exosomes might target cancer cells and organs, leading to higher capacity of clear cell renal cell carcinoma to the metastasize to the lungs (55). Matrix metalloproteinases (MMPs) can be transferred by exosomes to facilitate cancer cell invasion by degrading the extracellular stroma (56). Stromal infiltration alters the TME and facilitates tumor metastasis (57). As a constituent of exosomes, exosomal circRNA molecules are directly and indirectly involved in the crosstalk between the TME and tumor cells. The diverse intercellular exchange of exosomal circRNA might drive tumor metastasis.

EVs are circular membrane fragments released from the endosomal compartment as exosomes or shed from the surfaces of the cell membranes. They comprise a lipid bilayer membrane, forming a vesicle containing several types of biomolecules, including proteins, lipids, polysaccharides and nucleic acids. These membrane fragments can be transferred to a recipient cell. When the recipient cell absorbs the biomolecules, changes to the function and gene expression of the recipient cells are elicited (58,59).

Exosomal circRNA regulates TAM differentiation to accelerate tumor metastasis

Non-coding RNA derived from exosomes might stimulate the clearance function of macrophages. TAMs are key cells that create an immunosuppressive TME by inhibiting immune checkpoint proteins to release T cells (60). Macrophages can display very different functions depending on the nature of the microenvironment, such as cancer and inflammatory environments. Activated macrophages are classified into M1 (classically-activated macrophages) and M2 (alternatively-activated macrophages) phenotypes (61). Activated M1 macrophages exhibit anti-tumor activity and elicit anti-tumor adaptive immunity in the early stages of carcinogenesis. Activated M2 macrophages tend to display an immunosuppressive phenotype, suppress tissue repair and promote tumor progression (62). However, M2 macrophage differentiation facilitates tumor cell immune escape; however, the exact ratio of M2/M1 macrophages in response to tumors is unclear (63).

TAMs play multi-functional roles in tumor progression, including cancer initiation and promotion, immune regulation, metastasis and angiogenesis (60). Activated M2 macrophages accumulate in the TME and facilitate tumor cell metastasis. Increased levels of exosomes may modulate the expression of proteins and stimulates M1 macrophages, which can result in the removal of cancer cells. However, further research is needed to test this hypothesis.

Exosomal circRNA molecules affect target genes in order to facilitate biological functions through communication between different cell types. This characteristic distinguishes exosomal circRNA from classical endocrine circulating RNA. In a recent study, exosomal circRNA could be transported throughout the whole body, thus acting as critical mediators of intercellular communication and inducing neoplasm metastasis (64). A previous study has suggested that the level of exosomal circRNA might reflect the expression levels of these circRNA in cells and the physiological activity of the cells to a certain degree (65). CDR1-AS, also known as CiRS-7, a circRNA sponge for miR-7, was confirmed to have ~70 conserved binding-sites for miR-7 (66). CDR1-AS expression levels are increased at metastasis sites compared with primary sites (65). Zou et al (67) have suggested that CDR1-AS plays a specific role in immune and stromal cell infiltration in tumor tissue. They observed that high CDR1-AS expression was associated with a higher proportion of M2 macrophages. Moreover, CDR1-AS expression correlated negatively with the number of CD8+ T cells, activated natural killer cells, monocytes and neutrophils and correlated positively with the M2/M1 macrophage ratio.

CDR1-AS regulates the TGF-β signaling pathway and alters the TME (67). Stimulating TGF-β signaling can exert a great influence on the TME by remodeling the extracellular matrix (67,68). Moreover, exosomal circRNA likely accelerates M2 macrophage differentiation by altering the TME through the TGF-β signaling pathway (Fig. 2A).

Exosomal circRNA triggers EMT to promote tumor metastasis

EMT is essential in tumor metastasis. During EMT, epithelial cells lose their polarity and obtain invasive properties to become MSCs (69). Exosomal circRNA is closely associated with EMT in tumors (70). Chen et al (71) have demonstrated that exosomal circ-protein arginine methyltransferase 5 (circ-PRMT5) was upregulated in patients with urinary carcinoma of the bladder (UCB) and might predict metastatic progress. Indeed, upregulated exosomal circ-PRMT5 was positively associated with advanced clinical stage and poor survival. In addition, circ-PRMT5 also promoted EMT in UCB.

Other studies have also indicated that exosomal circRNA might induce EMT. In a mouse model, Zhang et al (72) investigated the role of exosomal circ-nuclear receptor interacting protein 1 (NRIP1) in distant metastasis via tail vein injection of gastric cancer cells co-cultured with exosomes. This study confirmed that exosomal circ-NRIP1 could promote EMT and metastasis in vivo (72). However, in another study, the opposite observation was made (73).

CDR1-AS1 binds to miR-7, which inhibits insulin like growth factor 1 receptor expression, thereby increasing the expression of E-cadherin to partially reverse EMT, subsequently inhibiting tumor cell metastasis (Fig. 2B) (74).

4. Bidirectional biological function of exosomal circRNA secreted by mesenchymal stem cells

Human MSCs restore damaged tissues in the body

MSCs may migrate to tumor sites and secrete a variety of factors, such as VEGF and platelet-derived growth factor (75). Exosomes exert number of functions that induce and support regenerative processes in necrotic tissue (76). MSC-derived exosomes also have antitumor effects. Exosomes derived from placental MSCs selectively inhibit the growth of prostate cancer cells through numerous molecular species within their cargo, such as mRNA, miRNA, lipids and proteins (77). MSC-derived exosomes transport mRNA and non-coding RNA to target cells and induce endothelial cell proliferation (78). For example, the transfer of miR-143 from MSC-derived exosomes to recipient cells can decrease the migration of osteosarcoma cells in vitro (79). Moreover, tumor cells can also reprogram surrounding MSCs to support the growth of tumors via inter-cellular communication, especially by releasing EVs. This long-term 'education' by tumor cells favors tumorigenesis (80). MSCs may secrete exosomes to promote the migration and invasion of breast cancer cells (81).

circRNA controls MSC identity and differentiation by sponging miRNA. For example, circforkhead box P1 (circ-FOXP1) is also enriched in MSCs. Silencing circ-FOXP1 expression markedly impairs MSC differentiation, Indeed, a previous study demonstrated that circ-FOXP1 regulates MSC molecular networks through the Wnt pathway (82). Tumor-derived exosomes also regulate MSCs in the distant metastatic microenvironment to promote metastasis (83). However, further experiments are required to verify the regulatory relationship between MSCs and exosomal circRNA.

Exosomal circRNA sponges miRNA and accelerates tumor metastasis

Tumor cells affect surrounding cells through direct contact, paracrine secretion and autocrine secretion (84). Intercellular communication can occur via exosomes, which has an important role in tumor metastasis and invasion (85). Exosomes containing non-coding RNA molecules are detectable in body fluids, particularly the blood, and are frequently released by tumor cells, implying that exosomes can act as messengers between cancer cells and immune cells to promote cancer cell escape from immune surveillance, thus contributing to tumor formation (86). As aforementioned, a previous study has indicated that circRNA is abundant and stable in exosomes and can continue to exert its role following uptake by recipient cells (7).

Metastasis is a phenomenon that can occur in the majority of cancer types. Tumor cells and normal cells can communicate with each other through exosomes, which secrete substances that affect tumor metastasis (87). The first metastasis mechanism occurs when exosomal circRNA induce the progression of a tumor by regulating cell signal transduction pathways (88). In ovarian cancer cells, circ-Wolf-Hirschhorn syndrome candidate 1 binds to miR-145 and miR-1182, which upregulates the expression of downstream targets mucin 1 and telomerase reverse transcriptase, thus promoting proliferation and invasion (89). It can also facilitate peritoneal dissemination via exosomes (89). Li et al (90) also found that high expression levels of exosomal circ-phosphodiesterase 8A (circ-PDE8A) in pancreatic ductal adenocarcinoma tissue was positively associated with invasion. Further research revealed that circ-PED8A promoted tumor cell growth by upregulating the MET proto-oncogene (91). Exosomal circRNA also regulates the progress of tumor metastasis via cell signal transduction. Moreover, circ-protein phosphatase 1 regulatory subunit 12A (circ-PPP1R12A) encodes a conserved 73-amino-acid peptide, PPP1R12A-C, which promotes the proliferation, migration and invasion of CC cells by activating the Hippo/YES-activating protein signaling pathway (27). Another study suggested that exosomal circ-NRIP1 promoted energy production by activating the protein kinase B/AKT/mTOR signaling pathway to facilitate gastric cancer tumor growth (72). Moreover, exosomal circ-NRIP1 promoted gastric cancer metastasis via EMT (72).

The second mechanism of tumor metastasis occurs through endothelial monolayer permeability and tumor cell invasion. In pancreatic cancer cells, exosomal circisoleucyl-tRNA synthetase 1 (circ-IARS) increases the expression levels of RhoA and F-actin and downregulates zona occludens-1, which increases the permeability of endothelial monolayer cells (92). Animal experiments have confirmed that circ-IARS can increase the number of tumor cells that can pass through the endothelial monolayer, which promotes metastasis (92).

Other evidence has suggested that exosomal circRNA modulates tumor cell migration by sponging miRNA, thus affecting metastasis. Exosomal circRNA may shorten the duration of tumor metastasis by sponging miRNA and regulating mRNA expression. For example, circ-PTGR1 is detectable in three HCC cell lines, namely the non-metastatic HepG2, the low-metastatic 97L and the highly metastatic LM3 cell lines (91). Exosomal circ-prostaglandin reductase 1 (circ-PTGR1) derived from highly metastatic HCC cells can promote metastasis in cells that normally have low metastatic potential (93). In particular, LM3 exosome-derived circ-PTGR1 promotes the progression of HepG2 and 97L cells in vitro and in vivo, which results from circ-PTGR1 competing with MET to target miR-449a. Thus, circ-PTGR1 can promote migration and metastasis in HCC through the circ-PTGR1/miR-449a/MET pathway (93).

In tumor tissues, exosomal circRNA mediates the progression of cancer by modulating surrounding normal cells to accelerate invasion and metastasis. Su et al (49) found that exosomal circRNA CDR1-AS from HCC cells accelerated the proliferative and migratory abilities of surrounding normal cells. Exosomal circRNA CDR1-AS serves as a competing endogenous RNA (ceRNA) to promote the progression of HCC (94). It is directly transferred from HCC cells to surrounding normal cells via exosomes to further mediate its biological functions (49). In nasopharyngeal carcinoma (NPC) tissues, CDR1-AS upregulates E2F transcription factor 3 expression by binding to miR-7-5p, which promotes the growth of NPC cells. Moreover, CDR1-AS could promote NPC progression by negatively regulating miR-7-5p in a nasopharyngeal carcinoma xenograft tumor model (95).

In prostate cancer, exosomal circRNA acts as miRNA sponge to induce EMT and facilitate tumor cell migration and invasion. Li et al (96) found that exosome circ-0044516 was significantly upregulated in patients with prostate cancer and in cell lines. Exosomal circ-0044516 downregulates miR-29a-3p expression, which inhibits the progression of prostate cancer cells (96). High expression levels of exosomal circ-0000284 enhance the migration, invasion and proliferation abilities of cholangiocarcinoma cells by acting as a ceRNA that binds to miR-637, further regulating lymphocyte antigen 6 family member E expression in vivo and in vitro (97). In bladder cancer, circhomeodomain interacting protein kinase 33 suppresses bladder cell migration, invasion and angiogenesis by sponging miR-558, which subsequently inhibits heparanase, MMP-9 and VEGF expression in vitro (98) (Fig. 2C).

5. Clinical applications of exosomal circRNA in cancer

Currently, clinical therapeutic progress includes improvements in early diagnosis, surgery, radiotherapy, chemotherapy and other types of therapeutic methods. Early effective diagnosis biomarkers are insufficient. Final diagnosis is often established by performing a biopsy, which may delay early diagnosis of the majority cancer patients. Biopsies can be painful, invasive for the patients and can lead to associated complications. Nevertheless, non-invasive early diagnostic biomarkers are lacking for clinical use and many types of carcinoma lack an authoritative indicator. Thus, exosomal circRNA could be developed as authoritative indicators. Interestingly, a previous study has shown circRNA are abundant and stable in exosomes, which can be detected in the circulation and urine (99). The most effective approach is RNA-seq analyses detection of circRNA molecules in exosomes (100,101).

Exosomal circRNA may represent diagnostic biomarkers in cancer

There is an urgent need to identify novel diagnostic biomarkers and develop more efficient therapeutic molecular targets in cancer. A recent study has revealed that the exosomes secreted by tumor cells are more abundant compared with those from normal cells (102). In addition, the contents of exosomes are different under normal physiological conditions compared with pathological conditions, even for the same tissue or body fluid (103). Exosomal circRNA is resistant to degradation and its secretion into the extracellular environment could be exploited in many biological applications; for example, as novel diagnostic biomarkers. For example, exosomal circRNA has been proposed as a potential diagnostic biomarker of idiopathic membranous nephropathy (104). A previous study has shown that both esophageal epithelial cells and esopha-geal squamous-cell carcinoma (ESCC) cells secrete exosomal circRNA and suggested that circRNA from patient plasma could be used for early diagnosis (105). hsa-circ-0001946 and hsa-circ-0043603 are found in exosomes of cell-conditioned culture conditioned media and may be potential diagnostic biomarkers for ESCC (106).

High-throughput sequencing analysis has demonstrated that the expression levels of three types of circRNA (hsa-circ-007293, hsa-circ-031752 and hsa-circ-020135) are upregulated in the serum of patients with papillary thyroid carcinoma. Thus, these exosome circRNA might represent potential diagnostic molecular biomarkers (107).

In cholangiocarcinoma, exosome-transmitted circ-0000284 stimulates the migration and proliferation of surrounding normal cells and promotes the progression of the tumor; thus, exosomal circ-0000284 may serve as a potential metastatic diagnostic biomarker (97). Exosomal circ-PTGR1 is abundant and aberrantly expressed in malignant cells of patients with metastatic HCC. Thus, circ-PTGR1 could act as a prognostic biomarker and therapeutic target (93). Circulating exosomal hsa-circ-0004771 is significantly upregulated in patients with CRC and may serve as a novel potential diagnostic biomarker of CRC (108). A previous study also suggested that hsa_circ_0001492 and hsa_circ_0001346 might be novel potential early diagnosis candidate markers in lung adenocarcinoma (109). Altogether, these studies indicated the possibility of using circRNA as tumor diagnostic markers. However, further research remains to be done to in different diseases and tumor subtypes.

Exosomal circRNA is associated with drug resistance

Drug resistance (chemotherapy insensitivity) is common during cancer therapy. Exosomal miRNA associated with drug resistance have been revealed in certain types of cancer, such as ovarian cancer (110). However, the detailed molecular mechanism underlying the relationship between exosomal circRNA and drug resistance is unclear and should be investigated to develop new target drugs. A study has highlighted epigenetic changes, such as resistance to cytotoxic drugs and angiogenic properties conferred by exosomes in the recipient cells through this 'cargo delivery' process (111). Zhao et al (112) have proposed that exosomal circRNA affects drug sensitivity. CDR1-AS is 1,500 nucleotides in length and is transcribed in the antisense orientation with respect to the CDR1 gene. Upregulation of exosomal CDR1-AS could increase the cisplatin sensitivity of ovarian cancer cells (112). CDR1-AS functions as a molecular sponge for miR-1270, which weakens the inhibitory effect of the miRNA on the downstream target gene suppressor of cancer cell invasion (113). CDR1-AS might regulate the sensitivity of ovarian cancer cells to cisplatin and the progression of ovarian cancer (112,114). Modulating the release of exosomal circRNA might be a potential therapeutic strategy to improve drug sensitivity in tumors. However, further trials are required to test this hypothesis.

6. Conclusions

Exosomal circRNA has stable biological function and structure. Importantly, circRNA is resistant to degradation by RNA exonuclease or RNase R and easily obtained from body fluid or serum (115). Recently, exosomal circRNA molecules have been proposed as novel cancer diagnostic biomarkers that can be extracted non-invasively. They have been used to study cancer progression and to evaluate prognosis. Several studies have demonstrated that exosomes are closely related to tumor development. The function of exosomes as biological delivery vehicles is of considerable interest in cancer research because modulating the TME using variety of RNA species may help increase the sensitivity of chemotherapeutics. Exosomal circRNA might promote the generation of the TME. Upregulation of exosomal circRNA expression can aggravate tumor progression. By contrast, downregulation of exosomal circRNA expression might inhibit tumor metastasis. The exosomal circRNA functions that impact on tumor progression can be divided into three pathways: i) sponging miRNA and regulating its function; ii) regulating transcription and translation; and iii) interacting with proteins to modulate gene expression. Thus, an improved understanding of exosome circRNA characteristics and functional roles in tumor progression is required. Although exosomal circRNA are known to display various functions, controlled studies in the majority of disease are lacking. In conclusion, it is essential to understand the detailed molecular mechanisms underlying the functions of exosomal circRNA, as this may lead to the development of novel diagnostic tools and treatment targets for tumors. Thus, further investigation of exosomal circRNA candidates and tumor metastasis may contribute to the control of cancer.

Funding

This work was supported by The Department of Science and Technology of Liaoning province (grant no. 2017225038) and 345 Talent Project funding from Sheng Jing Hospital of China Medical University (grant no. 2019345).

Availability and of data and materials

Not applicable.

Authors' contributions

XW wrote and edited the manuscript. BY was involved in acquisition of the data and provided direction and guidance throughout the preparation of this manuscript. XM generated the figures and revised the manuscript. YS, ZD and PW conceived the review and revised the manuscript. All authors read and approved the final manuscript. YS and ZD confirm the authenticity of the data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests

Acknowledgments

Not applicable.

Abbreviations:

CC

colon cancer

circRNA

circular RNA

CRC

colorectal cancer

EMT

epithelial-to-mesenchymal transition

ESCC

esophageal squamous-cell carcinoma

EV

extracellular vesicle

HCC

hepatocellular carcinoma

miRNA

microRNA

MMP

matrix metalloproteinase

MSC

mesenchymal stem cell

NPC

nasopharyngeal carcinoma

RNA-seq

RNA sequencing

TME

tumor microenvironment

UCB

urothelial carcinoma of the bladder

References

1 

Kristensen LS, Hansen TB, Venø MT and Kjems J: Circular RNAs in cancer: Opportunities and challenges in the field. Oncogene. 37:555–565. 2018. View Article : Google Scholar :

2 

Lin J, Zhang Y, Zeng X, Xue C and Lin X: CircRNA CircRIMS Acts as a MicroRNA sponge to promote gastric cancer metastasis. ACS Omega. 5:23237–23246. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Wang Y, Mo Y, Gong Z, Yang X, Yang M, Zhang S, Xiong F, Xiang B, Zhou M, Liao Q, et al: Circular RNAs in human cancer. Mol Cancer. 16:252017. View Article : Google Scholar : PubMed/NCBI

4 

Martins VR, Dias MS and Hainaut P: Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr Opin Oncol. 25:66–75. 2013. View Article : Google Scholar

5 

Whiteside TL: Tumor-derived exosomes and their role in cancer progression. Adv Clin Chem. 74:103–141. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF and Sharpless NE: Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA. 19:141–157. 2013. View Article : Google Scholar :

7 

Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, Chen D, Gu J, He X and Huang S: Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 25:981–984. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Funes SC, Rios M, Escobar-Vera J and Kalergis AM: Implications of macrophage polarization in autoimmunity. Immunology. 154:186–195. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Mantovani A, Sozzani S, Locati M, Allavena P and Sica A: Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 23:549–555. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Wang JJ, Lei KF and Han F: Tumor microenvironment: Recent advances in various cancer treatments. Eur Rev Med Pharmacol Sci. 22:3855–3864. 2018.PubMed/NCBI

11 

Sun Y: Tumor microenvironment and cancer therapy resistance. Cancer Lett. 380:205–215. 2016. View Article : Google Scholar

12 

Xie F, Zhou X, Fang M, Li H, Su P, Tu Y, Zhang L and Zhou F: Extracellular vesicles in cancer immune microenvironment and cancer immunotherapy. Adv Sci (Weinh). 6:19017792019. View Article : Google Scholar

13 

Sanger HL, Klotz G, Riesner D, Gross HJ and Kleinschmidt AK: Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci USA. 73:3852–3856. 1976. View Article : Google Scholar : PubMed/NCBI

14 

Li J, Mohammed-Elsabagh M, Paczkowski F and Li Y: Circular nucleic acids: Discovery, functions and applications. Chembiochem. 21:1547–1566. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Jeck WR and Sharpless NE: Detecting and characterizing circular RNAs. Nat Biotechnol. 32:453–461. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, et al: Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 22:256–264. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Patop IL, Wüst S and Kadener S: Past, present, and future of circRNAs. EMBO J. 38:e1008362019. View Article : Google Scholar : PubMed/NCBI

18 

Meng X, Chen Q, Zhang P and Chen M: CircPro: An integrated tool for the identification of circRNAs with protein-coding potential. Bioinformatics. 33:3314–3316. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Zhao X, Cai Y and Xu J: Circular RNAs: Biogenesis, mechanism, and function in human cancers. Int J Mol Sci. 20:39262019. View Article : Google Scholar :

20 

Wilusz JE: A 360° view of circular RNAs: From biogenesis to functions. Wiley interdisciplinary reviews. RNA. 9:e14782018.

21 

Chen X, Yang T, Wang W, Xi W, Zhang T, Li Q, Yang A and Wang T: Circular RNAs in immune responses and immune diseases. Theranostics. 9:588–607. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Liang D and Wilusz JE: Short intronic repeat sequences facilitate circular RNA production. Genes Dev. 28:2233–2247. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Das A, Rout PK, Gorospe M and Panda AC: Rolling Circle cDNA synthesis uncovers circular RNA splice variants. Int J Mol Sci. 20:39882019. View Article : Google Scholar :

24 

Chan JJ and Tay Y: Noncoding RNA:RNA Regulatory networks in cancer. Int J Mol Sci. 19:13102018. View Article : Google Scholar :

25 

Pardini B, Sabo AA, Birolo G and Calin GA: Noncoding RNAs in extracellular fluids as cancer biomarkers: The New Frontier of Liquid Biopsies. Cancers. 11:11702019. View Article : Google Scholar :

26 

Liu K, Zhang Q, Pan F, Wang XD, Wenjing H and Tong H: Expression of circular RNAs in gynecological tumors: A systematic review. Medicine. 98:e157362019. View Article : Google Scholar : PubMed/NCBI

27 

Zheng X, Chen L, Zhou Y, Wang Q, Zheng Z, Xu B, Wu C, Zhou Q, Hu W, Wu C and Jiang J: A novel protein encoded by a circular RNA circPPP1R12A promotes tumor pathogenesis and metastasis of colon cancer via Hippo-YAP signaling. Mol Cancer. 18:472019. View Article : Google Scholar : PubMed/NCBI

28 

Correia de Sousa M, Gjorgjieva M, Dolicka D, Sobolewski C and Foti M: Deciphering miRNAs' Action through miRNA Editing. Int J Mol Sci. 20:62492019. View Article : Google Scholar

29 

O'Brien J, Hayder H, Zayed Y and Peng C: Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol. 9:4022018. View Article : Google Scholar

30 

Tan W, Liu B, Qu S, Liang G, Luo W and Gong C: MicroRNAs and cancer: Key paradigms in molecular therapy. Oncol Lett. 15:2735–2742. 2018.PubMed/NCBI

31 

Xue D, Wang H, Chen Y, Shen D, Lu J, Wang M, Zebibula A, Xu L, Wu H, Li G and Xia L: Circ-AKT3 inhibits clear cell renal cell carcinoma metastasis via altering miR-296-3p/E-cadherin signals. Mol Cancer. 18:1512019. View Article : Google Scholar : PubMed/NCBI

32 

Li R, Wu B, Xia J, Ye L and Yang X: Circular RNA hsa_ circRNA_102958 promotes tumorigenesis of colorectal cancer via miR-585/CDC25B axis. Cancer Manag Res. 11:6887–6893. 2019. View Article : Google Scholar :

33 

Xu H, Guo S, Li W and Yu P: The circular RNA Cdr1as, via miR-7 and its targets, regulates insulin transcription and secretion in islet cells. Sci Rep. 5:124532015. View Article : Google Scholar : PubMed/NCBI

34 

Zhu W, Wang Y, Zhang D, Yu X and Leng X: MiR-75p functions as a tumor suppressor by targeting SOX18 in pancreatic ductal adenocarcinoma. Biochem Biophys Res Commun. 497:963–970. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Lin J, Liu Z, Liao S, Li E, Wu X and Zeng W: Elevated microRNA-7 inhibits proliferation and tumor angiogenesis and promotes apoptosis of gastric cancer cells via repression of Raf-1. Cell Cycle. 19:2496–2508. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Shen B, Wang Z, Li Z, Song H and Ding X: Circular RNAs: An emerging landscape in tumor metastasis. Am J Cancer Res. 9:630–643. 2019.PubMed/NCBI

37 

Tong H, Zhao K, Wang J, Xu H and Xiao J: CircZNF609/miR-134-5p/BTG-2 axis regulates proliferation and migration of glioma cell. J Pharm Pharmacol. 72:68–75. 2020. View Article : Google Scholar

38 

Yang P, Qiu Z, Jiang Y, Dong L, Yang W, Gu C, Li G and Zhu Y: Silencing of cZNF292 circular RNA suppresses human glioma tube formation via the Wnt/β-catenin signaling pathway. Oncotarget. 7:63449–63455. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Rybak-Wolf A, Stottmeister C, Glažar P, Jens M, Pino N, Giusti S, Hanan M, Behm M, Bartok O, Ashwal-Fluss R, et al: Circular RNAs in the mammalian brain are highly abundant, conserved, and dynamically expressed. Mol Cell. 58:870–885. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Li Z, Chen Z, Hu G and Jiang Y: Roles of circular RNA in breast cancer: Present and future. Am J Transl Res. 11:3945–3954. 2019.PubMed/NCBI

41 

Sun S, Wang W, Luo X, Li Y, Liu B and Li X, Zhang B, Han S and Li X: Circular RNA circ-ADD3 inhibits hepatocellular carcinoma metastasis through facilitating EZH2 degradation via CDK1-mediated ubiquitination. Am J Cancer Res. 9:1695–1707. 2019.PubMed/NCBI

42 

Liu Z, Yu Y, Huang Z, Kong Y, Hu X, Xiao W, Quan J and Fan X: CircRNA-5692 inhibits the progression of hepatocellular carcinoma by sponging miR-328-5p to enhance DAB2IP expression. Cell Death Dis. 10:9002019. View Article : Google Scholar : PubMed/NCBI

43 

Boriachek K, Islam MN, Möller A, Salomon C, Nguyen NT, Hossain MSA, Yamauchi Y and Shiddiky MJA: Biological functions and current advances in isolation and detection strategies for exosome nanovesicles. Small. 14:17021532018. View Article : Google Scholar

44 

Braicu C, Tomuleasa C, Monroig P, Cucuianu A, Berindan-Neagoe I and Calin GA: Exosomes as divine messengers: Are they the Hermes of modern molecular oncology? Cell Death Differ. 22:34–45. 2015. View Article : Google Scholar

45 

Pant S, Hilton H and Burczynski ME: The multifaceted exosome: Biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem Pharmacol. 83:1484–1494. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Ngalame NNO, Luz AL, Makia N and Tokar EJ: Arsenic alters exosome quantity and cargo to mediate stem cell recruitment into a cancer stem cell-like phenotype. Toxicol Sci. 165:40–49. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Shi X, Wang B, Feng X, Xu Y, Lu K and Sun M: circRNAs and exosomes: A mysterious frontier for human cancer. Mol Ther Nucleic Acids. 19:384–392. 2020. View Article : Google Scholar :

48 

Li S, Li Y, Chen B, Zhao J, Yu S, Tang Y, Zheng Q, Li Y, Wang P, He X and Huang S: exoRBase: A database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 46:D106–D112. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Su Y, Lv X, Yin W, Zhou L, Hu Y, Zhou A and Qi F: CircRNA Cdr1as functions as a competitive endogenous RNA to promote hepatocellular carcinoma progression. Aging (Albany NY). 11:8182–8203. 2019.

50 

Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, et al: Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 24:541–550. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Ridge SM, Sullivan FJ and Glynn SA: Mesenchymal stem cells: Key players in cancer progression. Mol Cancer. 16:312017. View Article : Google Scholar : PubMed/NCBI

52 

Kalluri R: The biology and function of fibroblasts in cancer. Nat Rev Cancer. 16:582–598. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Li K, Chen Y, Li A, Tan C and Liu X: Exosomes play roles in sequential processes of tumor metastasis. Int J Cancer. 144:1486–1495. 2019. View Article : Google Scholar

54 

Matei I, Kim HS and Lyden D: Unshielding exosomal RNA unleashes tumor growth and metastasis. Cell. 170:223–225. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Wang L, Yang G, Zhao D, Wang J, Bai Y, Peng Q, Wang H, Fang R, Chen G, Wang Z, et al: CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote MiR-19b-3p. Mol Cancer. 18:862019. View Article : Google Scholar : PubMed/NCBI

56 

Hakulinen J, Sankkila L, Sugiyama N, Lehti K and Keski-Oja J: Secretion of active membrane type 1 matrix metalloproteinase (MMP-14) into extracellular space in microvesicular exosomes. J Cell Biochem. 105:1211–1218. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Turley SJ, Cremasco V and Astarita JL: Immunological hall-marks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 15:669–682. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Lai CP, Kim EY, Badr CE, Weissleder R, Mempel TR, Tannous BA and Breakefield XO: Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters. Nat Commun. 6:70292015. View Article : Google Scholar : PubMed/NCBI

59 

Yoon YJ, Kim OY and Gho YS: Extracellular vesicles as emerging intercellular communicasomes. BMB Rep. 47:531–539. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Lin Y, Xu J and Lan H: Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J Hematol Oncol. 12:762019. View Article : Google Scholar : PubMed/NCBI

61 

Biswas SK and Mantovani A: Macrophage plasticity and inter-action with lymphocyte subsets: Cancer as a paradigm. Nat Immunol. 11:889–896. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Najafi M, Hashemi Goradel N, Farhood B, Salehi E, Nashtaei MS, Khanlarkhani N, Khezri Z, Majidpoor J, Abouzaripour M, Habibi M, et al: Macrophage polarity in cancer: A review. J Cell Biochem. 120:2756–2765. 2019. View Article : Google Scholar

63 

Allavena P, Sica A, Garlanda C and Mantovani A: The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol Rev. 222:155–161. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Preußer C, Hung LH, Schneider T, Schreiner S, Hardt M, Moebus A, Santoso S and Bindereif A: Selective release of circRNAs in platelet-derived extracellular vesicles. J Extracell Vesicles. 7:14244732018. View Article : Google Scholar

65 

Hou J, Jiang W, Zhu L, Zhong S, Zhang H, Li J, Zhou S, Yang S, He Y, Wang D, et al: Circular RNAs and exosomes in cancer: A mysterious connection. Clin Transl Oncol. 20:1109–1116. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Sekar S, Cuyugan L, Adkins J, Geiger P and Liang WS: Circular RNA expression and regulatory network prediction in posterior cingulate astrocytes in elderly subjects. BMC Genomics. 19:3402018. View Article : Google Scholar : PubMed/NCBI

67 

Zou Y, Zheng S, Deng X, Yang A and Xie X, Tang H and Xie X: The role of circular RNA CDR1as/ciRS-7 in regulating tumor micro-environment: A pan-cancer analysis. Biomolecules. 9:4292019. View Article : Google Scholar

68 

Pickup M, Novitskiy S and Moses HL: The roles of TGFβ in the tumour microenvironment. Nature Revi Cancer. 13:788–799. 2013. View Article : Google Scholar

69 

Yang J and Weinberg RA: Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell. 14:818–829. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Shang BQ, Li ML, Quan HY, Hou PF, Li ZW, Chu SF, Zheng JN and Bai J: Functional roles of circular RNAs during epithelial-to-mesenchymal transition. Mol Cancer. 18:1382019. View Article : Google Scholar : PubMed/NCBI

71 

Chen X, Chen RX, Wei WS, Li YH, Feng ZH, Tan L, Chen JW, Yuan GJ, Chen SL, Guo SJ, et al: PRMT5 circular RNA promotes metastasis of urothelial carcinoma of the bladder through sponging miR-30c to induce epithelial-mesenchymal transition. Clin Cancer Res. 24:6319–6330. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Zhang X, Wang S, Wang H, Cao J, Huang X, Chen Z, Xu P, Sun G, Xu J, Lv J and Xu Z: Circular RNA circNRIP1 acts as a microRNA-149-5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol Cancer. 18:202019. View Article : Google Scholar : PubMed/NCBI

73 

Ma C, Shi T, Qu Z, Zhang A, Wu Z, Zhao H, Zhao H and Chen H: CircRNA_ACAP2 suppresses EMT in head and neck squamous cell carcinoma by targeting the miR-21-5p/STAT3 signaling axis. Front Oncol. 10:5836822020. View Article : Google Scholar : PubMed/NCBI

74 

Zhao X, Dou W, He L, Liang S, Tie J, Liu C, Li T, Lu Y, Mo P, Shi Y, et al: MicroRNA-7 functions as an anti-metastatic microRNA in gastric cancer by targeting insulin-like growth factor-1 receptor. Oncogene. 32:1363–1372. 2013. View Article : Google Scholar

75 

Ball SG, Shuttleworth CA and Kielty CM: Mesenchymal stem cells and neovascularization: Role of platelet-derived growth factor receptors. J Cell Mol Med. 11:1012–1030. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Altaner C, Altanerova U and Jakubechova J: Intracellular acting tumor cell-targeted chemotherapy by MSC-suicide gene exosomes. Oncotarget. 10:5573–5575. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Brossa A, Fonsato V and Bussolati B: Anti-tumor activity of stem cell-derived extracellular vesicles. Oncotarget. 10:1872–1873. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Ferguson SW, Wang J, Lee CJ, Liu M, Neelamegham S, Canty JM and Nguyen J: The microRNA regulatory landscape of MSC-derived exosomes: A systems view. Sci Rep. 8:14192018. View Article : Google Scholar : PubMed/NCBI

79 

Shimbo K, Miyaki S, Ishitobi H, Kato Y, Kubo T, Shimose S and Ochi M: Exosome-formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem Biophys Res Commun. 445:381–387. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Li X, Wang S, Zhu R, Li H, Han Q and Zhao RC: Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J Hematol Oncol. 9:422016. View Article : Google Scholar

81 

Menck K, Klemm F, Gross JC, Pukrop T, Wenzel D and Binder C: Induction and transport of Wnt 5a during macrophage-induced malignant invasion is mediated by two types of extracellular vesicles. Oncotarget. 4:2057–2066. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Cherubini A, Barilani M, Rossi RL, Jalal MMK, Rusconi F, Buono G, Ragni E, Cantarella G, Simpson HARW, Péault B and Lazzari L: FOXP1 circular RNA sustains mesenchymal stem cell identity via microRNA inhibition. Nucleic Acids Res. 47:5325–5340. 2019. View Article : Google Scholar : PubMed/NCBI

83 

Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, et al: Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biol. 17:816–826. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Ahmed N, Escalona R, Leung D, Chan E and Kannourakis G: Tumour microenvironment and metabolic plasticity in cancer and cancer stem cells: Perspectives on metabolic and immune regulatory signatures in chemoresistant ovarian cancer stem cells. Semin Cancer Biol. 53:265–281. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Steinbichler TB, Dudás J, Riechelmann H and Skvortsova II: The role of exosomes in cancer metastasis. Semin Cancer Biol. 44:170–181. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Raposo G and Stoorvogel W: Extracellular vesicles: Exosomes, microvesicles, and friends. J Cell Biol. 200:373–383. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Wortzel I, Dror S, Kenific CM and Lyden D: Exosome-mediated metastasis: Communication from a distance. Dev Cell. 49:347–360. 2019. View Article : Google Scholar : PubMed/NCBI

88 

Zhang H, Zhu L, Bai M, Liu Y, Zhan Y, Deng T, Yang H, Sun W, Wang X, Zhu K, et al: Exosomal circRNA derived from gastric tumor promotes white adipose browning by targeting the miR-133/PRDM16 pathway. Int J Cancer. 144:2501–2515. 2019. View Article : Google Scholar

89 

Zong ZH, Du YP, Guan X, Chen S and Zhao Y: CircWHSC1 promotes ovarian cancer progression by regulating MUC1 and hTERT through sponging miR-145 and miR-1182. J Exp Clin Cancer Res. 38:4372019. View Article : Google Scholar : PubMed/NCBI

90 

Li Z, Yanfang W, Li J, Jiang P, Peng T, Chen K, Zhao X, Zhang Y, Zhen P, Zhu J and Li X: Tumor-released exosomal circular RNA PDE8A promotes invasive growth via the miR-338/MACC1/MET pathway in pancreatic cancer. Cancer Lett. 432:237–250. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Luna J, Boni J, Cuatrecasas M, Bofill-De Ros X, Núñez-Manchón E, Gironella M, Vaquero EC, Arbones ML, de la Luna S and Fillat C: DYRK1A modulates c-MET in pancreatic ductal adenocarcinoma to drive tumour growth. Gut. 68:1465–1476. 2019. View Article : Google Scholar

92 

Li J, Li Z, Jiang P, Peng M, Zhang X, Chen K, Liu H, Bi H, Liu X and Li X: Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res. 37:1772018. View Article : Google Scholar : PubMed/NCBI

93 

Wang G, Liu W, Zou Y, Wang G, Deng Y, Luo J, Zhang Y, Li H, Zhang Q, Yang Y and Chen G: Three isoforms of exosomal circPTGR1 promote hepatocellular carcinoma metastasis via the miR449a-MET pathway. EBioMedicine. 40:432–445. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Yang X, Xiong Q, Wu Y, Li S and Ge F: Quantitative proteomics reveals the regulatory networks of circular RNA CDR1as in hepatocellular carcinoma cells. J Proteome Res. 16:3891–3902. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Zhong Q, Huang J, Wei J and Wu R: Circular RNA CDR1as sponges miR-7-5p to enhance E2F3 stability and promote the growth of nasopharyngeal carcinoma. Cancer Cell Int. 19:2522019. View Article : Google Scholar : PubMed/NCBI

96 

Li T, Sun X and Chen L: Exosome circ_0044516 promotes prostate cancer cell proliferation and metastasis as a potential biomarker. J Cell Biochem. 121:2118–2126. 2020. View Article : Google Scholar

97 

Wang S, Hu Y, Lv X, Li B, Gu D, Li Y, Sun Y and Su Y: Circ-0000284 arouses malignant phenotype of cholangiocarcinoma cells and regulates the biological functions of peripheral cells through cellular communication. Clin Sci (Lond). 133:1935–1953. 2019. View Article : Google Scholar

98 

Li Y, Zheng F, Xiao X, Xie F, Tao D, Huang C, Liu D, Wang M, Wang L, Zeng F and Jiang G: CircHIPK3 sponges miR-558 to suppress heparanase expression in bladder cancer cells. EMBO Rep. 18:1646–1659. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Ren GL, Zhu J, Li J and Meng XM: Noncoding RNAs in acute kidney injury. J Cell Physiol. 234:2266–2276. 2019. View Article : Google Scholar

100 

Xu Y, Ku X, Wu C, Cai C, Tang J and Yan W: Exosomal proteome analysis of human plasma to monitor sepsis progression. Biochem Biophys Res Commun. 499:856–861. 2018. View Article : Google Scholar : PubMed/NCBI

101 

Fanale D, Taverna S, Russo A and Bazan V: Circular RNA in Exosomes. Adv Exp Med Biol. 1087:109–117. 2018. View Article : Google Scholar : PubMed/NCBI

102 

Chen R, Xu X, Qian Z, Zhang C, Niu Y, Wang Z, Sun J, Zhang X and Yu Y: The biological functions and clinical applications of exosomes in lung cancer. Cell Mol Life Sci. 76:4613–4633. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Lakhal S and Wood MJ: Exosome nanotechnology: An emerging paradigm shift in drug delivery: Exploitation of exosome nanovesicles for systemic in vivo delivery of RNAi heralds new horizons for drug delivery across biological barriers. Bioessays. 33:737–741. 2011. View Article : Google Scholar : PubMed/NCBI

104 

Ma H, Xu Y, Zhang R, Guo B, Zhang S and Zhang X: Differential expression study of circular RNAs in exosomes from serum and urine in patients with idiopathic membranous nephropathy. Arch Med Sci. 15:738–753. 2019. View Article : Google Scholar : PubMed/NCBI

105 

Liu S, Lin Z, Rao W, Zheng J, Xie Q, Lin Y, Lin X, Chen H, Chen Y and Hu Z: Upregulated expression of serum exosomal hsa_circ_0026611 is associated with lymph node metastasis and poor prognosis of esophageal squamous cell carcinoma. J Cancer. 12:918–926. 2021. View Article : Google Scholar : PubMed/NCBI

106 

Fan L, Cao Q, Liu J, Zhang J and Li B: Circular RNA profiling and its potential for esophageal squamous cell cancer diagnosis and prognosis. Mol Cancer. 18:162019. View Article : Google Scholar : PubMed/NCBI

107 

Yang C, Wei Y, Yu L and Xiao Y: Identification of altered circular RNA expression in serum exosomes from patients with papillary thyroid carcinoma by high-throughput sequencing. Med Sci Monit. 25:2785–2791. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Pan B, Qin J, Liu X, He B, Wang X, Pan Y, Sun H, Xu T, Xu M, Chen X, et al: Identification of serum exosomal hsa-circ-0004771 as a novel diagnostic biomarker of colorectal cancer. Front Genet. 10:10962019. View Article : Google Scholar : PubMed/NCBI

109 

Chen F, Huang C, Wu Q, Jiang L, Chen S and Chen L: Circular RNAs expression profiles in plasma exosomes from early-stage lung adenocarcinoma and the potential biomarkers. J Cell Biochem. 121:2525–2533. 2020. View Article : Google Scholar

110 

Feng Y, Hang W, Sang Z, Li S, Xu W, Miao Y, Xi X and Huang Q: Identification of exosomal and non-exosomal microRNAs associated with the drug resistance of ovarian cancer. Mol Med Rep. 19:3376–3392. 2019.PubMed/NCBI

111 

Sousa D, Lima RT and Vasconcelos MH: Intercellular transfer of cancer drug resistance traits by extracellular vesicles. Trends Mol Med. 21:595–608. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Zhao Z, Ji M, Wang Q, He N and Li Y: Circular RNA Cdr1as upregulates SCAI to suppress cisplatin resistance in ovarian cancer via miR-1270 suppression. Mol Ther Nucleic Acids. 18:24–33. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Tang W, Ji M, He G, Yang L, Niu Z, Jian M, Wei Y, Ren L and Xu J: Silencing CDR1as inhibits colorectal cancer progression through regulating microRNA-7. OncoTargets Ther. 10:2045–2056. 2017. View Article : Google Scholar

114 

Sang M, Meng L, Sang Y, Liu S, Ding P, Ju Y, Liu F, Gu L, Lian Y, Li J, et al: Circular RNA ciRS-7 accelerates ESCC progression through acting as a miR-876-5p sponge to enhance MAGE-A family expression. Cancer Lett. 426:37–46. 2018. View Article : Google Scholar : PubMed/NCBI

115 

Burd CE, Jeck WR, Liu Y, Sanoff HK, Wang Z and Sharpless NE: Expression of linear and novel circular forms of an INK4/ARF-associated non-coding RNA correlates with atherosclerosis risk. PLoS Genet. 6:e10012332010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 58 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei X, Shi Y, Dai Z, Wang P, Meng X and Yin B: Underlying metastasis mechanism and clinical application of exosomal circular RNA in tumors (Review). Int J Oncol 58: 289-297, 2021
APA
Wei, X., Shi, Y., Dai, Z., Wang, P., Meng, X., & Yin, B. (2021). Underlying metastasis mechanism and clinical application of exosomal circular RNA in tumors (Review). International Journal of Oncology, 58, 289-297. https://doi.org/10.3892/ijo.2021.5179
MLA
Wei, X., Shi, Y., Dai, Z., Wang, P., Meng, X., Yin, B."Underlying metastasis mechanism and clinical application of exosomal circular RNA in tumors (Review)". International Journal of Oncology 58.3 (2021): 289-297.
Chicago
Wei, X., Shi, Y., Dai, Z., Wang, P., Meng, X., Yin, B."Underlying metastasis mechanism and clinical application of exosomal circular RNA in tumors (Review)". International Journal of Oncology 58, no. 3 (2021): 289-297. https://doi.org/10.3892/ijo.2021.5179