The role of non‑coding RNAs in the regulation, diagnosis, prognosis and treatment of osteosarcoma (Review)

  • Authors:
    • Gu Yang
    • Yanjiao Wu
    • Rongxue Wan
    • Hongxun Sang
    • Huan Liu
    • Wenhua Huang
  • View Affiliations

  • Published online on: July 22, 2021     https://doi.org/10.3892/ijo.2021.5249
  • Article Number: 69
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteosarcoma (OS) is the most common primary bone tumor worldwide. OS exhibits a range of aggressive behaviors, including early metastasis potential, rapid progression, poor clinical prognosis and insensitivity to chemoradiotherapy. Non‑coding RNAs are transcripts that do not encode proteins. A significant number of studies published on OS have been focused on the aberrant expression of non‑coding RNAs and their involvement in tumor initiation and progression. It has been confirmed that non‑coding RNAs exert their regulatory functions at both the transcriptional and post‑transcriptional level, which leads to tumor initiation or progression in OS. According to present knowledge, this review provides a state‑of‑the‑art overview of the functions and mechanisms of microRNAs, long non‑coding RNAs and circular RNAs in terms of their involvement with OS. The review also covers their potential clinical application in the diagnosis, prognosis and treatment of OS. It is hoped that the information presented in this review on the involvement of non‑coding RNAs in OS will lead to a more comprehensive understanding of OS and provide a useful perspective on the potential diagnostic and therapeutic applications of non‑coding RNAs for patients with OS.

1. Introduction

Osteosarcoma (OS) is considered to be the most commonly occurring type of primary bone tumor, with an estimated worldwide incidence of 3-4 new cases per million, which accounts for approximately 60% of the total number of cases of bone malignancy (1,2). OS is derived from the transformation of primitive mesenchymal cells, and typically occurs in the metaphyseal region of long bones, with a peak incidence among young individuals (3,4). OS exhibits a range of aggressive behaviors, including early metastasis potential, rapid progression, poor clinical prognosis and insensitivity to chemoradiotherapy, which collectively lead to a poor overall survival rate (5,6). Prior to the 1970s, surgical resection was the preferred treatment for OS, although, as the 5-year survival rate was <20%, it was insufficient as a means of therapy for numerous patients (7,8).

The current therapeutic strategy for OS includes neoadjuvant chemotherapy after surgical removal of the tumor and adjuvant chemotherapy with or without lesion metastasis, which has led to a marked increase in the survival rate to approximately 65% over the course of the last 30 years (9). However, despite notable improvements achieved in terms of surgical techniques and neoadjuvant chemotherapy, the overall survival time of patients with distant metastasis or multi-drug resistance cannot be effectively prolonged (10-12). Therefore, it is crucial to elucidate the underlying molecular mechanisms that are involved in the tumorigenesis and progression of OS, and to identify novel biomarkers for developing alternative therapies or improving the efficiency of existing treatments.

After having mapped out the transcriptional 'landscape' of the mammalian genome, findings showed that protein-coding mRNAs only account for 1.4% of the genome (13,14). Furthermore, by comparing the number of protein-coding genes with the genome size among different species, it was shown that the more complex eukaryotes carry a larger proportion of non-coding RNA (ncRNA) (15). These phenomena led to the suggestion that the ncRNAs in the human genome may contribute towards complex physiological and pathological processes. Based on a cutoff at 200 bases of length, ncRNAs are standardly categorized as short ncRNAs (sncRNAs) and long ncRNAs (lncRNAs) (16). MicroRNAs (miRNAs/miRs), as a typical class of sncRNAs, have been extensively studied. It has been confirmed that miRNAs exercise a regulatory role on the expression of protein-coding mRNAs, which leads to the initiation or progression of numerous diseases, including cancer (17,18). In addition, there are other types of ncRNAs, including small interfering RNAs (siRNAs), small nuclear RNAs (snRNAs) and small nucleolar RNAs (snoRNAs). The lncRNAs, as observed from a wide spectrum of profiling in mammals, are a class of transcripts >200 nucleotides (nt) in length (13,19,20). Increasing evidence has demonstrated that lncRNAs fulfill functional roles in the occurrence and development of tumors (21,22). Along with the development of high-throughput sequencing technology and novel computational approaches, a set of circular RNAs (circRNAs) have recently been identified. It was revealed that circRNAs are involved in diverse pathological processes, including oncogenesis and tumor progression (23).

With the aim of encouraging further studies on these RNA species, the present review article provides a concise summary of ncRNAs and their biogenesis, their underlying molecular mechanisms and their potential clinical applications in OS. It is the authors' hope that associated research in the future may lead to a more comprehensive understanding of OS and present a reasonable perspective on the potential diagnostic and therapeutic application of ncRNAs in patients with OS.

2. miRNAs

Biogenesis and features of miRNAs

The first miRNA, lin-4, was discovered in Caenorhabditis elegans in 1993 and its involvement in various biological processes has been observed (24-26). Since then, thousands of miRNAs have been identified and studied over the course of the past 20 years. Further research has revealed that genes influenced by miRNAs exist in all metazoans and plants, suggesting that miRNA-associated regulation operates according to a highly conserved mechanism (27).

The biogenesis of miRNAs is a complex process, including nuclear synthesis and cytoplasmic synthesis with the involvement of a specific set of enzymes. First, in the nucleus, a primary miRNA (pri-miRNA) with special hairpin structures (AAAAA and 7MGpppG) is synthesized according to the transcriptional gene that encodes the miRNA by RNA polymerase II. Subsequently, the hairpin domain of the pri-miRNA is cleaved by an RNA-specific nuclease termed Drosha (ribonuclease III) to produce precursor miRNAs (pre-miRNAs) that possess a stem-ring structure and are 70-80 nt in length (28). Then, with the help of cytoplasmic transporter exportin-5, the pre-miRNAs are translocated from the nucleus to the cytoplasm. Within the cytoplasm, these pre-miRNAs are further cleaved into a miRNA duplex of ~19-23 nt by ribonuclease III (Dicerase) (29). The miRNA duplex consists of two strands: A mature miRNA strand and a passenger miRNA strand. After strand unwinding, the mature miRNA strand is transformed into a mature miRNA via its interaction with Argonaute protein, whereas the passenger miRNA is usually degraded (30).

Regulatory functions of miRNAs in OS

Aberrant expression of miRNAs has been reported as a common phenomenon occurring in a diversity of cancer types, including breast, lung, hepatocellular, colon and cervical cancer (31,32). miRNAs exert their regulatory role through interacting with their mRNA target genes. Normally, there are two mechanisms by which mature miRNAs are able to form RNA-induced silencing complex (RISC). In the first scenario, in cases where the miRNA is fully complementary to the target gene, the miRNA degrades the target gene. In the second scenario, where the miRNA is not fully complementary to target gene, miRNA combines with the 3′-untranslated region to inhibit translation of the target gene (33). In addition, it has been confirmed that one single miRNA may affect multiple mRNAs, or conversely, multiple miRNAs may affect one single mRNA (27). Through the mechanism described above, miRNAs are heavily involved in multiple instances of cancer occurrence and development, including proliferation, apoptosis and metastasis (34,35).

Similarly, certain miRNAs have been demonstrated to regulate malignancy by serving either as an oncogene or as an onco-suppressor in OS. miR-210-5p was recently shown to be upregulated in human OS tissues and cell lines, closely correlating with the advanced tumor-node-metastasis (TNM) stage, tumor size and pulmonary metastasis. Overexpressed miR-210-5p led to an increase in the rates of tumor invasion, migration and autophagy by suppressing the downstream target, phosphoinositide-3-kinase regulatory subunit 5 (PIK3R5). Of note, miR-210-5p-mediated autophagy facilitates miR-210-5p-induced tumor invasion and migration promotion via inhibiting the AKT/mTOR pathway (36). Another upregulated miRNA, miR-624-5p, has been identified in clinical OS specimens and cell lines. Further functional analysis suggested that miR-624-5p may promote cell proliferation, migration and invasion both in vitro and in vivo. Identified as the target gene of miR-624-5p, protein tyrosine phosphatase receptor type B (PTPRB) was found to be negatively correlated with miR-624-5p. Furthermore, PTPRB restored the effects of miR-624-5p on OS migration and invasion (37). miR-627-3p was shown to be downregulated in OS tissues using biochip analysis, and was also shown to be expressed at a lower level in OS cell lines compared with human osteoblastic cells. Moreover, miR-627-3p significantly suppressed the expression and activity of pleiotrophin (PTN), and PTN affected the proliferation and migration of OS cells via regulation of a range of different proteins, including cyclin D1 and matrix metalloproteinase-2 (38). Also through microarray analysis, miR-15b was found to be markedly downregulated in the doxorubicin-resistance cell lines, KHOS and U-2OS (39). Furthermore, patients with OS who had high expression levels of miR-15b received a significantly improved clinical prognosis compared with those with low expression levels. Wee1, the direct target of miR-15b, was shown to mediate both the cytotoxic effect of doxorubicin and the multidrug-resistance capabilities in OS (39).

Although the participation of miRNAs in OS is a complex and multifactorial process, emerging evidence has strongly supported the involvement of numerous miRNAs in the processes of OS initiation and progression, including cell proliferation, apoptosis, immigration, invasion and drug resistance. These miRNAs and their roles are shown in Table I (36-51).

Table I

Examples of the involvement of miRNAs in OS.

Table I

Examples of the involvement of miRNAs in OS.

NameExpressionRoleRelated moleculesPhenotypes affectedExperimental models(Refs.)
miR-627-3pTumor suppressorPTNProliferation (−), migration (−),invasion (−)HOS, U2OS, MG63 and MG63/dox cells(38)
miR-15bTumor suppressorWee1Drug resistance (+)KHOS, U-2OS and corresponding drug-resistant cells(39)
miR-382Tumor suppressorKLF12 and HIPK3Cell growth (−),chemosensitivity (+)MNNG/HOS, U2OS and MG63 cells(40)
miR-145Tumor suppressorFLI-1Cell growth (−)HOS, Saos-2, U2OS and MG-63(41)
miR-143Tumor suppressorMMP-13Metastasis (−)143B cells(42)
miR-26aTumor suppressorJagged1Cell growth (−)U2OS, MG63, Saos-2 and 143B cells(43)
miR-29bTumor suppressorCDK6Proliferation (−), migration (−)MG-63 cells(44)
miR-539Tumor suppressorMMP-8Migration (−), invasion (−)MG-63, U2OS and Saos2 cells(45)
miR-210-5pOncogenePIK3R5Migration (+), invasion (+),autophagy (+)HOS, Saos-2, SW1353, U2OS, and MG63 cells(36)
miR-624-5pOncogenePTPRBMigration (+), invasion (+)U2OS, MG63, SW1353, HOS, and Saos-2 cells(37)
miR-21OncogeneCaspase-8Apoptosis (−)Saos-2 cells(46)
miR-543OncogenePRMT9Proliferation (+)Saos2, MNNG/HOS, U2OS and MG63 cells(47)
miR-27aOncogeneCBFA2T3Metastasis (+)HOS, KHOS, SAOS2, 143B and U2OS cells(48)
miR-191OncogeneCHEK2Proliferation (+)Saos2 and MG-63 cells(49)
miR-199a-5pOncogenePIAS3 and p27Cell growth (+)MG63, Saos-2 and 143B cells(50)
miR-9OncogeneGCIPCell growth (+)U2OS and HOS cells(51)

[i] ↓, downregulated; ↑, upregulated; PTN, pleiotrophin; Wee1, WEE1 G2 checkpoint kinase; KLF12, Kruppel-like factor 12; HIPK3, homeodomain interacting protein kinase 3; FLI-1, friend leukemia virus integration 1; MMP-13, matrix metallopeptidase 13; CDK6, cyclin-dependent kinase 6; MMP-8, matrix metallopeptidase 8; PIK3R5, phosphoinositide-3-kinase regulatory subunit 5; PRMT9, protein arginine methyltransferase 9; CBFA2T3, CBFA2/RUNX1 partner transcriptional co-repressor 3; CHEK2, checkpoint kinase 2; PIAS3, protein inhibitor of activated STAT 3; GCIP, Grap2 and cyclin D interacting protein.

Role of miRNAs in the diagnosis, prognosis and treatment of OS

MiRNAs have consistently attracted a great deal of attention from researchers as putative diagnostic or prognostic biomarkers for OS due to their stability in the plasma and serum (52). miR-21 overexpression has been shown to be strongly associated with advanced Enneking stage, chemotherapeutic resistance and an unfavorable prognosis, and therefore miR-21 may be used as an individual marker for OS staging and prognosis (53). Low levels of miR-101 have been observed in serum samples from patients with OS; however, the miR-101 expression levels reverted to significantly higher levels following treatment (54). miR-195-5p and miR-199a-3p have been shown to have remarkable potential in terms of distinguishing between metastatic and non-metastatic statuses in patients with OS, whereas miR-320a and miR-199a-3p were associated with the histological subtype (55).

At present, the standard clinical treatments mainly comprise surgical resection and neoadjuvant therapy. The involvement of miRNAs in the initiation and progression of OS renders miRNAs suitable as possible therapeutic targets. The corresponding approach would involve the use of miRNA mimics to substitute for the loss of expression of a tumor-suppressor miRNA or to block the expression of an oncomiR using oligonucleotides or anti-viral constructs (56). A mimic of miR-34 as a tumor suppressor for cancer treatment was entered into Phase I clinical trials (57). Furthermore, it was identified that a miR-34 mimic significantly suppressed lung metastasis in OS mouse models, strongly suggesting that miR-34 may serve as a potential therapeutic target (58).

3. lncRNAs

Biogenesis and features of lncRNA

lncRNAs form a large subgroup of ncRNAs with transcripts >200 bases in length that lack the ability to encode proteins. For the most part, they are transcribed by RNA polymerase II, the same as mRNAs, and they are often 5′-capped, polyadenylated and spliced without a translated open reading frame (59,60). Based on their location with the neighboring protein-coding genes, they can be divided into the following five classes: Sense, antisense, bidirectional, intronic and intergenic lncRNAs (61-63). lncRNAs are also characterized by their low abundance and through the tissue- and developmental stage-specific manner of their expression (64,65).

Regulatory functions of lncRNAs in OS

Similar to miRNAs, numerous lncRNAs have been demonstrated to have key roles as contributors to tumor initiation or progression. The manner in which a given lncRNA exerts its regulatory effect depends on its subcellular localization. lncRNAs in the cytoplasm that share miRNA response elements with mRNAs contain similar sequences to these target-coding RNAs and inhibit the interactions between miRNAs and mRNAs. These lncRNAs, which are termed competing endogenous RNAs (ceRNAs), act as 'sponges' for miRNAs and regulate the process of translation mediated by miRNAs on their target mRNAs (66). lncRNAs in the nucleus mainly act at the epigenetic and genetic levels by binding to the transcription preinitiation complex at the promoter (67).

The lncRNA DANCR has been shown to be elevated in OS tissue specimens and cell lines and is closely correlated with poor prognosis among clinical patients. DANCR serves as an oncogene, regulating ROCK1-mediated proliferation and metastasis through sequestering both miR-335-5p and miR-1972 as a ceRNA (68). The lncRNA HIF1A-AS2 has also been shown to be upregulated in OS, and is associated with poor survival. HIF1A-AS2 regulates the tumorigenesis of OS, as demonstrated by its effects on cell proliferation, cell cycle progression and invasion, through 'sponging' miR-129-5p (69). In OS, the lncRNA TTN-AS1 has been shown to facilitate cell growth, apoptosis and drug resistance via the miR-134-5p/MBTD1 axis (70). By contrast, the lncRNA TTN-AS1 also acts as a ceRNA on miRNA-376a, enhancing the malignancy of OS via upregulating dickkopf-1 (71). Other confirmed lncRNAs are presented in Table II (68-79).

Table II

Examples of the involvement of lncRNAs in OS.

Table II

Examples of the involvement of lncRNAs in OS.

NameExpressionRoleRelated moleculesPhenotypes affectedExperimental models(Refs.)
CASC2Tumor suppressormiR-181a and RASSF6Proliferation (−), invasion (−)MG-63, SW1353, Saos-2, SOSP-9607 and U2OS cells(72)
CTATumor suppressormiR-210Drug resistance (+)Saos-2, U-2OS and MG-63 cells(73)
GAS5Tumor suppressorDownregulate ARHI via miR-221Cell growth (−), migration (−),EMT (−)KHOS, 143B, SAOS-2, U2OS, and MG-63 cells(74)
FENDRRTumor suppressorABCB1 and ABCC1Apoptosis (+), drug resistance (−)MG63, SaoS2 and HOS cells(75)
DANCROncogeneUpregulate ROCK1 via miR-335-5p and miR-1972Proliferation (+), invasion (+),migration (+)MG-63, U2OS, MNNG/HOS and 143B cells(68)
HIF1A-AS2OncogenemiR-129-5pProliferation (+), invasion (+)U2OS, SoSP-M, SaOS2, MG-63 cells(69)
TTN-AS1OncogenemiR-134-5p/MBTD1 axis and miR-376a/DKK1 axisProliferation (+), apoptosis (+), invasion (+), migration (+) drug resistance (−)Saos-2, HOS, MG-63 and U2OS cells(70,71)
H19OncogeneInactivate NF-κB pathway via PI3K/AKT signaling pathwayMigration (+), invasion (+)MG-63, U2OS and SAOS-2 cells(76)
SNHG12OncogeneUpregulate Notch2 via miR-195-5pProliferation (+), invasion (+), migration (+)143B, U2OS, MG63 and HOS cells(77)
NEAT1OncogeneUpregulate HOXA13 via miR-34a-5pProliferation (+), cells apoptosis (−)Saos2, MG63, U2OS, SJSA1, and HOS cells(78)
APTROncogeneUpregulate YAP1 via miR-132-3pProliferation (+), invasion (+), migration (+), apoptosis (−)MG63 and 143B(79)

[i] ↓, downregulated; ↑, upregulated; CASC2, cancer susceptibility 2; RASSF6, Ras association domain family member 6; GAS5, growth arrest specific 5; ARHI, aplasia Ras homolog member 1; FENDRR, FOXF1 adjacent non-coding developmental regulatory RNA; ABCB1, ATP binding cassette subfamily B member 1; ABCC1, ATP binding cassette subfamily C member 1; DANCR, differentiation antagonizing non-protein coding RNA; ROCK1, Rho associated coiled-coil containing protein kinase 1; HIF1A-AS2, hypoxia inducible factor 1 subunit α antisense RNA 2; TTN-AS1, titin antisense RNA 1; MBTD1, mbt domain containing 1; DKK1, dickkopf WNT signaling pathway inhibitor 1; PI3K, plasmodium gallinaceum phosphatidylinositol 3-kinase; SNHG12, small nucleolar RNA host gene 12; NOTCH2, notch receptor 2; NEAT1, nuclear paraspeckle assembly transcript 1; HOXA13, homeobox A13; APTR, Alu-mediated CDKN1A/p21 transcriptional regulator; YAP1, Yes1-associated transcriptional regulator.

Role of lncRNAs in the diagnosis, prognosis and treatment of OS

At present, the main surveillance methods of OS are limited to physical examination, blood biochemistry and radiographic examination (80). Due to the lack of an effective and noninvasive measure to monitor patient status or predict overall survival, lncRNAs are considered as potential candidates for prognosis prediction and treatment guidance. The level of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been shown to be upregulated in 162 OS tissues, closely correlating with advanced clinical stage, distant metastasis and shorter survival times. Therefore, MALAT1 is able to serve as an independent prognostic factor for OS (81). The clinical potential of taurine-upregulated gene 1 (TUG1) has also been demonstrated through its marked elevation in patients with OS progression and relapse. Furthermore, the serum levels of TUG1 were shown to be decreased after surgical resection of OS tissues in postoperative patients (82). The most common obstacles in the treatment of OS are the poor therapeutic response to traditional chemo- and radio-therapies and the emergence of resistance during treatment (83). lncRNAs that mediate acquired resistance are potential candidates as targets for OS therapies.

4. circRNAs

Biogenesis and features of circRNAs

CircRNAs, a novel class of non-protein-coding RNAs, were first discovered in the 1970s, and were then considered as 'junk' molecules with little functional potential (84). They are generated from pre-mRNAs through back-splicing, and are expressed in a tissue- and developmental stage-specific manner (85,86). In addition, they are evolutionarily conserved and highly abundant in the brain. Unlike traditional linear RNAs, circRNAs are characterized by a continuous covalently closed loop structure lacking either a 5′-cap or a 3′-polyadenylated tail, which gives them stronger resistance to ribonucleases compared with their corresponding linear counterparts (87,88).

Regulatory functions of circRNAs in OS

Emerging evidence has shown that circRNAs fulfill essential roles in both physiological and pathological processes, including oncogenesis and tumor progression (23). circRNAs have multiple functions, such as regulating gene expression at the transcriptional or post-transcriptional level by interacting with miRNAs as 'sponges', binding to RNA-binding protein and initiating protein translation in a splicing-dependent, cap-independent manner (89,90). The circRNA circTADA2A has been reported to be highly upregulated in OS, and acts as a sponge for the miRNA miR-203a-3p, which regulates CREB3 expression to promote the proliferation, migration and invasion of OS cells in vitro (91). The circRNA hsa_circ_0001564, detected through circRNA microarray analysis, has been shown to be upregulated in OS tissues. A further study revealed that hsa_circ_001564 aggravates OS proliferation and apoptosis via sponging miR-29c-3p (92). circPVT1 facilitates the doxorubicin and cisplatin resistance of OS via increasing the expression of the classical drug resistance-associated gene, ABCB1 (93). In patients with OS, circNASP expression was found to be positively correlated with tumor size and lung metastasis. Upregulated circNASP acts as a sponge of miR-1253, targeting the transcription factor FOXF1 to markedly promote the proliferation, cell cycle progression and invasion of OS cells (94). Other novel circRNAs are listed in Table III (91-103).

Table III

The expression and function of circRNAs in OS.

Table III

The expression and function of circRNAs in OS.

NameExpressionRoleRelated moleculesPhenotypes affectedExperimental models(Refs.)
circ_0002052Tumor suppressorInactivate Wnt/β-catenin pathway via miR-1205/APC2 axisProliferation (−), apoptosis (+), migration (−), invasion (−)HOS, U2OS, MG63 and 143B cells(95)
circITCHTumor suppressormiR-22 and miR-7Proliferation (−), apoptosis (+), migration (−), invasion (−)MG63, U2OS, Saos-2 and SJSA-1 cells(96,97)
circ_HIPK3Tumor suppressor-Proliferation (−), migration (−), invasion (−)SaoS2, HOS, KH-OS, MG63, 143B and U2OS cells(98)
circTADA2AOncogenemiR-203a-3p/CREB3 axisProliferation (+), migration (+), invasion (+)HOS, 143B, MG-63, U2OS and SJSA-1 cells(91)
circ_0001564OncogenemiR-29c-3pProliferation (+), apoptosis (−)U2OS, Saos-2, HOS and MG-63 cells(92)
circPVT1OncogeneABCB1Drug resistance (+)SaoS2, KHOS, U2OS, MG63 cells(93)
circNASPOncogenemiR-1253/FOXF1 axisProliferation (+), invasion (+)143B and MG63 cells(94)
circ_0102049Oncogeneupregulate MDM2 via miR-1304-5pCell growth (+), migration (−), invasion (+), apoptosis (−)MG63, HOS, Saos2, and U2OS cells(99)
circ_0001721OncogenemiR-569 and miR-599Proliferation (+), apoptosis (−), migration (+), invasion (+)HOS, Saos2, MG63 and U2OS cells(100)
circGLI2OncogenemiR125b-5pProliferation (+), migration (+),invasion (+)MG63, Saos-2, HOS and U2OS cells(101)
circ_0001785Oncogeneregulate HOXB2 via miR-1200Proliferation (+), apoptosis (−)Saos2, U2OS, SJSA1, HOS and MG63 cells(102)
circ_0060428Oncogeneregulate RBPJ via miR-375Proliferation (+), apoptosis (−)U2OS, 143B, SAOS-2 and HOS cells(103)

[i] ↓, downregulated; ↑, upregulated; ITCH, itchy E3 ubiquitin protein ligase; HIPK3, homeodomain interacting protein kinase 3; TADA2A, transcriptional adaptor 2A; CREB3, cAMP responsive element binding protein 3; NASP, nuclear autoantigenic sperm protein; FOXF1, forkhead box F1; GLI2, GLI family zinc finger 2; HOXB2, homeobox B2; RBPJ, recombination signal binding protein for immunoglobulin κ J region.

Role of circRNAs in the diagnosis, prognosis and treatment of OS

CircRNAs can be secreted in body fluids where they are circulated, and the structures of circRNAs are geared towards a high level of resistance to cleavage by RNA exonucleases or ribonuclease R. These features, along with high specificity and sensitivity, demonstrate that circRNAs may serve as good candidates for OS (104,105). For example, the high expression level of serum circPVT1 enabled patients with OS to be distinguished from healthy individuals, suggesting that circPVT1 may be more reliable as a diagnostic biomarker compared to the traditional biomarker alkaline phosphatase (93). In another study, evaluated expression levels of circUBAP2 were found via Kaplan-Meier survival analysis to be correlated with reduced survival and poor prognosis, and they were also significantly correlated with the tumor stages (106).

CircRNAs, as ceRNAs, are natural miRNA inhibitors that bind to their corresponding miRNA to regulate the malignant behavior of OS. This property ensures that circRNAs have great potential in terms of therapeutic strategies. Recently, a newly designed artificial miRNA sponge has been developed. This artificial circRNA can sponge multiple miR-21 molecules, and has been reported to upregulate the expression of the tumor suppressor gene DAXX to inhibit the proliferation of gastric cancer cells (107).

5. Conclusions and perspectives

The identification of ncRNAs and their role in cancer initiation and progression has provided revolutionary insights into how the research efforts for OS may be directed. MiRNAs exert their regulatory functions via RISC, whereas lncRNAs and circRNAs function according to mechanisms involving ceRNAs (Fig. 1). However, since the majority of these studies have focused on miRNAs, lncRNAs and circRNAs, rather than other types of ncRNA (such as snoRNAs), the data remain incomplete. Of the numerous human ncRNAs, only a few have been thoroughly studied, and only a limited number of these have an important biological impact. The RNA world is much more complicated than was once considered to be the case, and the clinical environment for OS progression should be greatly improved if more research is devoted to the study of ncRNAs and their involvement in this type of cancer.

Although the significant clinical potential of ncRNAs as biomarkers in OS has been acknowledged, there exist several limitations. In most of the studies that have been performed to date, the cohort of patients with OS was relatively small; thus, long-term, controlled and large-sample experiments are required. In addition, the detected ncRNA biomarkers do not perform entirely consistently even for a particular cancer type, which is an important obstacle in terms of their usefulness as biomarkers. Therefore, it is necessary to identify combinations of several ncRNAs with a high degree of specificity and sensitivity, and to develop a standardized approach in methodology to normalize the expression of ncRNAs.

NcRNAs possess a unique advantage in terms of their clinical application in OS treatment. A single ncRNA simultaneously targets multiple downstream factors and is involved in multiple signaling pathways, which brings important benefits for refractory cancers with genomic heterogeneity. However, there are certain disadvantages or challenges associated with ncRNA-targeted treatment strategy. First, they may break the balance of gene expression profiles in cells due to unrelated genes being targeted by the same ncRNAs. It has been observed that miRNAs may exert different effects, which means that miRNA antagonists or mimics must be carefully selected according to different conditions. Furthermore, the existence of off-target effects for ncRNA antagonists cannot be ignored. Therefore, extensive toxicity studies and preclinical safety requirements should be assessed before an ncRNA-based therapeutic approach may be considered as being appropriate for patients with OS (108). At present, researchers have also made efforts to develop effective drug delivery systems for chemotherapy. The reduction-responsive polypeptide micelles were developed as multifunctional nanoparticle-based drug delivery systems based on methoxy poly-block-poly copolymers. These micelles can selectively accumulate in OS tumors, which induces antitumor effects with less systematic toxicity (109). As for nucleic acid therapeutics, approaches of delivery systems for targeting ncRNAs have been developing at a rapid pace. There are some general problems of nucleic acid delivery strategies including short half-life, off-target effects and low transfection efficiency in RNA delivery, which makes nucleic acid drugs remain at a low bioavailability in vivo (110,111). To overcome the aforementioned obstacles, a variety of ncRNA carriers or systems have been investigated, including nanoparticles, ncRNA modification, and oncolytic adenovirus strategy (112). Several of the delivery strategies have been applied in the research of hepatic carcinoma (113,114). Despite the lack of nucleic acid drug delivery strategies and associated research in OS, nucleic acid therapy may prove beneficial for OS treatment through further study.

In the present review, we have made detailed predictions on the future research directions:

One single ncRNA is able to affect multiple downstream target molecules associated with cancer development, and one single downstream target can be regulated by multiple upstream molecules. For instance, the lncRNA DANCR promotes cell proliferation and metastasis via sponging miR-335-5p and miR-1972 in OS (68). Therefore, understanding the complicated connections between the ncRNA regulatory networks, as well as determining some important core ncRNAs in OS, requires further investigation.

Typically, researchers obtain tissue samples for early diagnosis and prognosis via surgery resections, which is a difficult and inconvenient procedure. Findings have shown that the serum expression of the lncRNA UCA1 was significantly higher in patients with OS compared with healthy controls. In addition, the upregulation of UCA1 was correlated with clinical stage and metastasis (115). Although the data reported are only preliminary, it is possible to predict that liquid biopsy, such as human peripheral blood, is a promising non-invasive technique for OS diagnosis and prognosis in clinical practice. Furthermore, clinical specimens can vary from puncture fluid to sputum if patients have lung metastasis.

At present, a limited number of proteins or peptides encoded by ncRNAs have been verified, and these have important biological and/or pathological functions in the occurrence and development of different tumors. A conserved 53-amino-acid peptide encoded by the lncRNA HOXB-AS3 was shown to suppress the proliferation, migration, invasion of colon cancer cells and tumor growth both in vitro and in vivo (116). FBXW7-185aa is encoded by the circRNA FBXW7, and inhibits glioma growth (117). Therefore, it is important to distinguish whether ncRNAs exert functions by acting directly as RNA molecules, or through encoding peptides or proteins.

Avoiding immune surveillance is an important hallmark of tumor initiation and progression (118). Recent findings have shown that ncRNAs are able to facilitate tumor immune escape to enhance malignant behaviors. The PD-1/PD-L1 pathway provides a key immunosuppressive mechanism for cancer cells, and miR-140 is associated with anti-tumor immunity via its effects on the PD-L1/PD-1 immune checkpoint signaling pathway in OS (119). At present, however, there are only limited numbers of studies on ncRNAs and their association with the tumor immune response in OS. Thus, further investigation to develop promising immunotherapies for patients with OS is necessary.

From a technical perspective, the research technology of circular RNA (circRNA) is similar to the classic RNA research technology, such as RT-PCR, qPCR, FISH (RNA positioning) and NB (northern blot) (RNA expression). However, with the emergence of new technologies, the research methods of circular RNA (circRNA) have become more abundant, such as using CRISPR/Cas13 system to efficiently knock down the expression of circular RNA without affecting the expression of its parental linear mRNA (120).

In conclusion, the investigation of emerging functional ncRNAs has led to a deeper understanding of the pathologies that control initiation and progression in OS. Moreover, the potential applications of ncRNAs in the diagnosis, prognosis and therapy of OS have been revealed in recent years. Further efforts, however, are required to elucidate the ncRNA-associated regulatory mechanisms and to establish ncRNA-targeted therapeutic options.

Availability of data and materials

Not applicable.

Authors' contributions

GY, YW, HS and HL made substantial contributions to the concept and design of the review, collected information and wrote the manuscript. RW and WH collected references, and reviewed and edited the manuscript. HS was the major contributor in drafting and revising the manuscript. The authenticity of all the raw data have been assessed by GY and YW to ensure its legitimacy. All authors read and approved the final version of this manuscript.

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.

References

1 

Mirabello L, Troisi RJ and Savage SA: Osteosarcoma incidence and survival rates from 1973 to 2004: Data from the surveillance, epidemiology, and end results program. Cancer. 115:1531–1543. 2009. View Article : Google Scholar

2 

Ma O, Cai WW, Zender L, Dayaram T, Shen J, Herron AJ, Lowe SW, Man TK, Lau CC and Donehower LA: MMP13, Birc2 (cIAP1), and Birc3 (cIAP2), amplified on chromosome 9, collaborate with p53 deficiency in mouse osteosarcoma progression. Cancer Res. 69:2559–2567. 2009. View Article : Google Scholar

3 

Sampson VB, Kamara DF and Kolb EA: Xenograft and genetically engineered mouse model systems of osteosarcoma and Ewing's sarcoma: Tumor models for cancer drug discovery. Expert Opin Drug Discov. 8:1181–1189. 2013. View Article : Google Scholar

4 

Moore DD and Luu HH: Osteosarcoma. Cancer Treat Res. 162:65–92. 2014. View Article : Google Scholar

5 

Shi ZW, Wang JL, Zhao N, Guan Y and He W: Single nucleotide polymorphism of hsa-miR-124a affects risk and prognosis of osteosarcoma. Cancer Biomark. 17:249–257. 2016. View Article : Google Scholar

6 

He F, Zhang W, Shen Y, Yu P, Bao Q, Wen J, Hu C and Qiu S: Effects of resection margins on local recurrence of osteosarcoma in extremity and pelvis: Systematic review and meta-analysis. Int J Surg. 36:283–292. 2016. View Article : Google Scholar

7 

Marcove RC, Miké V, Hajek JV, Levin AG and Hutter RV: Osteogenic sarcoma under the age of twenty-one. A review of one hundred and forty-five operative cases. J Bone Joint Surg Am. 52:411–423. 1970. View Article : Google Scholar

8 

Dahlin DC and Coventry MB: Osteogenic sarcoma. A study of six hundred cases. J Bone Joint Surg Am. 49:101–110. 1967. View Article : Google Scholar

9 

Isakoff MS, Bielack SS, Meltzer P and Gorlick R: Osteosarcoma: Current treatment and a collaborative pathway to success. J Clin Oncol. 33:3029–3035. 2015. View Article : Google Scholar

10 

Wang B, Xu M, Zheng K and Yu X: Effect of unplanned therapy on the prognosis of patients with extremity osteosarcoma. Sci Rep. 6:387832016. View Article : Google Scholar

11 

Liu K, Huang J, Ni J, Song D, Ding M, Wang J, Huang X and Li W: MALAT1 promotes osteosarcoma development by regulation of HMGB1 via miR-142-3p and miR-129-5p. Cell Cycle. 16:578–587. 2017. View Article : Google Scholar

12 

Chen L, Wang Q, Wang GD, Wang HS, Huang Y, Liu XM and Cai XH: miR-16 inhibits cell proliferation by targeting IGF1R and the Raf1-MEK1/2-ERK1/2 pathway in osteosarcoma. FEBS Lett. 587:1366–1372. 2013. View Article : Google Scholar

13 

Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, Oyama R, Ravasi T, Lenhard B, Wells C, et al: The transcriptional landscape of the mammalian genome. Science. 309:1559–1563. 2005. View Article : Google Scholar

14 

Baltimore D: Our genome unveiled. Nature. 409:814–816. 2001. View Article : Google Scholar

15 

Mattick JS: The genetic signatures of noncoding RNAs. PLoS Genet. 5:e10004592009. View Article : Google Scholar

16 

Kour S and Rath PC: Long noncoding RNAs in aging and age-related diseases. Ageing Res Rev. 26:1–21. 2016. View Article : Google Scholar

17 

Ji Q, Xu X, Song Q, Xu Y, Tai Y, Goodman SB, Bi W, Xu M, Jiao S, Maloney WJ and Wang Y: miR-223-3p inhibits human osteosarcoma metastasis and progression by directly targeting CDH6. Mol Ther. 26:1299–1312. 2018. View Article : Google Scholar

18 

Andersen GB, Knudsen A, Hager H, Hansen LL and Tost J: miRNA profiling identifies deregulated miRNAs associated with osteosarcoma development and time to metastasis in two large cohorts. Mol Oncol. 12:114–131. 2018. View Article : Google Scholar

19 

ENCODE Project Consortium; Birney E, Stamatoyannopoulos JA, Dutta A, Guigó R, Gingeras TR, Margulies EH, Weng Z, Snyder M, Dermitzakis ET, et al: Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 447:799–816. 2007. View Article : Google Scholar

20 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012. View Article : Google Scholar

21 

Evans JR, Feng FY and Chinnaiyan AM: The bright side of dark matter: lncRNAs in cancer. J Clin Invest. 126:2775–2782. 2016. View Article : Google Scholar

22 

Lin C and Yang L: Long noncoding RNA in cancer: Wiring signaling circuitry. Trends Cell Biol. 28:287–301. 2018. View Article : Google Scholar

23 

Barrett SP and Salzman J: Circular RNAs: Analysis, expression and potential functions. Development. 143:1838–1847. 2016. View Article : Google Scholar

24 

Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE and Mello CC: Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 391:806–811. 1998. View Article : Google Scholar

25 

Ebert MS and Sharp PA: Roles for microRNAs in conferring robustness to biological processes. Cell. 149:515–524. 2012. View Article : Google Scholar

26 

Hwang HW and Mendell JT: MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer. 94:776–780. 2006. View Article : Google Scholar

27 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar

28 

Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S and Kim VN: The nuclear RNase III Drosha initiates microRNA processing. Nature. 425:415–419. 2003. View Article : Google Scholar

29 

Foulkes WD, Priest JR and Duchaine TF: DICER1: Mutations, microRNAs and mechanisms. Nat Rev Cancer. 14:662–672. 2014. View Article : Google Scholar

30 

Stappert L, Roese-Koerner B and Brüstle O: The role of microRNAs in human neural stem cells, neuronal differentiation and subtype specification. Cell Tissue Res. 359:47–64. 2015. View Article : Google Scholar

31 

Bottai G, Pasculli B, Calin GA and Santarpia L: Targeting the microRNA-regulating DNA damage/repair pathways in cancer. Expert Opin Biol Ther. 14:1667–1683. 2014. View Article : Google Scholar

32 

Adams BD, Kasinski AL and Slack FJ: Aberrant regulation and function of microRNAs in cancer. Curr Biol. 24:R762–R776. 2014. View Article : Google Scholar

33 

Huntzinger E and Izaurralde E: Gene silencing by microRNAs: Contributions of translational repression and mRNA decay. Nat Rev Genet. 12:99–110. 2011. View Article : Google Scholar

34 

Ell B and Kang Y: MicroRNAs as regulators of bone homeostasis and bone metastasis. Bonekey Rep. 3:5492014. View Article : Google Scholar

35 

Nugent M: MicroRNA function and dysregulation in bone tumors: The evidence to date. Cancer Manag Res. 6:15–25. 2014. View Article : Google Scholar

36 

Liu W, Jiang D, Gong F, Huang Y, Luo Y, Rong Y, Wang J, Ge X, Ji C, Fan J and Cai W: miR-210-5p promotes epithelial-mesenchymal transition by inhibiting PIK3R5 thereby activating oncogenic autophagy in osteosarcoma cells. Cell Death Dis. 11:932020. View Article : Google Scholar

37 

Luo Y, Liu W, Tang P, Jiang D, Gu C, Huang Y, Gong F, Rong Y, Qian D, Chen J, et al: miR-624-5p promoted tumorigenesis and metastasis by suppressing hippo signaling through targeting PTPRB in osteosarcoma cells. J Exp Clin Cancer Res. 38:4882019. View Article : Google Scholar

38 

He M, Shen P, Qiu C and Wang J: miR-627-3p inhibits osteosarcoma cell proliferation and metastasis by targeting PTN. Aging (Albany NY). 11:5744–5756. 2019. View Article : Google Scholar

39 

Duan Z, Gao Y, Shen J, Choy E, Cote G, Harmon D, Bernstein K, Lozano-Calderon S, Mankin H and Hornicek FJ: miR-15b modulates multidrug resistance in human osteosarcoma in vitro and in vivo. Mol Oncol. 11:151–166. 2017. View Article : Google Scholar

40 

Xu M, Jin H, Xu CX, Sun B, Mao Z, Bi WZ and Wang Y: miR-382 inhibits tumor growth and enhance chemosensitivity in osteosarcoma. Oncotarget. 5:9472–9483. 2014. View Article : Google Scholar

41 

Wu P, Liang J, Yu F, Zhou Z, Tang J and Li K: miR-145 promotes osteosarcoma growth by reducing expression of the transcription factor friend leukemia virus integration 1. Oncotarget. 7:42241–42251. 2016. View Article : Google Scholar

42 

Hirahata M, Osaki M, Kanda Y, Sugimoto Y, Yoshioka Y, Kosaka N, Takeshita F, Fujiwara T, Kawai A, Ito H, et al: PAI-1, a target gene of miR-143, regulates invasion and metastasis by upregulating MMP-13 expression of human osteosarcoma. Cancer Med. 5:892–902. 2016. View Article : Google Scholar

43 

Lu J, Song G, Tang Q, Yin J, Zou C, Zhao Z, Xie X, Xu H, Huang G, Wang J, et al: MiR-26a inhibits stem cell-like phenotype and tumor growth of osteosarcoma by targeting Jagged1. Oncogene. 36:231–241. 2017. View Article : Google Scholar

44 

Zhu K, Liu L, Zhang J, Wang Y, Liang H, Fan G, Jiang Z, Zhang CY, Chen X and Zhou G: MiR-29b suppresses the proliferation and migration of osteosarcoma cells by targeting CDK6. Protein Cell. 7:434–444. 2016. View Article : Google Scholar

45 

Jin H and Wang W: MicroRNA-539 suppresses osteosarcoma cell invasion and migration in vitro and targeting matrix metallopeptidase-8. Int J Clin Exp Pathol. 8:8075–8082. 2015.

46 

Xu B, Xia H, Cao J, Wang Z, Yang Y and Lin Y: MicroRNA-21 inhibits the apoptosis of osteosarcoma cell line SAOS-2 via targeting caspase 8. Oncol Res. 25:1161–1168. 2017. View Article : Google Scholar

47 

Zhang H, Guo X, Feng X, Wang T, Hu Z, Que X, Tian Q, Zhu T, Guo G, Huang W and Li X: MiRNA-543 promotes osteosarcoma cell proliferation and glycolysis by partially suppressing PRMT9 and stabilizing HIF-1α protein. Oncotarget. 8:2342–2355. 2017. View Article : Google Scholar

48 

Salah Z, Arafeh R, Maximov V, Galasso M, Khawaled S, Abou-Sharieha S, Volinia S, Jones KB, Croce CM and Aqeilan RI: miR-27a and miR-27a* contribute to metastatic properties of osteosarcoma cells. Oncotarget. 6:4920–4935. 2015. View Article : Google Scholar

49 

Huang YZ, Zhang J, Shao HY, Chen JP and Zhao HY: MicroRNA-191 promotes osteosarcoma cells proliferation by targeting checkpoint kinase 2. Tumour Biol. 36:6095–6101. 2015. View Article : Google Scholar

50 

Wang C, Ba X, Guo Y, Sun D, Jiang H, Li W, Huang Z, Zhou G, Wu S, Zhang J and Chen J: MicroRNA-199a-5p promotes tumour growth by dual-targeting PIAS3 and p27 in human osteosarcoma. Sci Rep. 7:414562017. View Article : Google Scholar

51 

Zhu SW, Li JP, Ma XL, Ma JX, Yang Y, Chen Y and Liu W: miR-9 modulates osteosarcoma cell growth by targeting the GCIP tumor suppressor. Asian Pac J Cancer Prev. 16:4509–4513. 2015. View Article : Google Scholar

52 

Zhou S, Wang B, Hu J, Zhou Y, Jiang M, Wu M, Qin L and Yang X: miR-421 is a diagnostic and prognostic marker in patients with osteosarcoma. Tumour Biol. 37:9001–9007. 2016. View Article : Google Scholar

53 

Yuan J, Chen L, Chen X, Sun W and Zhou X: Identification of serum microRNA-21 as a biomarker for chemosensitivity and prognosis in human osteosarcoma. J Int Med Res. 40:2090–2097. 2012. View Article : Google Scholar

54 

Yao ZS, Li C, Liang D, Jiang XB, Tang JJ, Ye LQ, Yuan K, Ren H, Yang ZD, Jin DX, et al: Diagnostic and prognostic implications of serum miR-101 in osteosarcoma. Cancer Biomark. 22:127–133. 2018. View Article : Google Scholar

55 

Lian F, Cui Y, Zhou C, Gao K and Wu L: Identification of a plasma four-microRNA panel as potential noninvasive biomarker for osteosarcoma. PLoS One. 10:e01214992015. View Article : Google Scholar

56 

Garzon R, Marcucci G and Croce CM: Targeting microRNAs in cancer: Rationale, strategies and challenges. Nat Rev Drug Discov. 9:775–789. 2010. View Article : Google Scholar

57 

Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017. View Article : Google Scholar

58 

Jian C, Tu MJ, Ho PY, Duan Z, Zhang Q, Qiu JX, DeVere White RW, Wun T, Lara PN, Lam KS, et al: Co-targeting of DNA, RNA, and protein molecules provides optimal outcomes for treating osteosarcoma and pulmonary metastasis in spontaneous and experimental metastasis mouse models. Oncotarget. 8:30742–30755. 2017. View Article : Google Scholar

59 

Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012. View Article : Google Scholar

60 

Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A and Rinn JL: Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25:1915–1927. 2011. View Article : Google Scholar

61 

Zhang K, Shi ZM, Chang YN, Hu ZM, Qi HX and Hong W: The ways of action of long non-coding RNAs in cytoplasm and nucleus. Gene. 547:1–9. 2014. View Article : Google Scholar

62 

Chen LL: Linking long noncoding RNA localization and function. Trends Biochem Sci. 41:761–772. 2016. View Article : Google Scholar

63 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009. View Article : Google Scholar

64 

Sun J, Lin Y and Wu J: Long non-coding RNA expression profiling of mouse testis during postnatal development. PLoS One. 8:e757502013. View Article : Google Scholar

65 

Ríos-Barrera LD, Gutiérrez-Pérez I, Domínguez M and Riesgo-Escovar JR: acal is a long non-coding RNA in JNK signaling in epithelial shape changes during drosophila dorsal closure. PLoS Genet. 11:e10049272015. View Article : Google Scholar

66 

Tao F, Tian X, Ruan S, Shen M and Zhang Z: miR-211 sponges lncRNA MALAT1 to suppress tumor growth and progression through inhibiting PHF19 in ovarian carcinoma. FASEB J. fj201800495RR2018.Online ahead of print.

67 

Schaukowitch K and Kim TK: Emerging epigenetic mechanisms of long non-coding RNAs. Neuroscience. 264:25–38. 2014. View Article : Google Scholar

68 

Wang Y, Zeng X, Wang N, Zhao W, Zhang X, Teng S, Zhang Y and Lu Z: Long noncoding RNA DANCR, working as a competitive endogenous RNA, promotes ROCK1-mediated proliferation and metastasis via decoying of miR-335-5p and miR-1972 in osteosarcoma. Mol Cancer. 17:892018. View Article : Google Scholar

69 

Wang X, Peng L, Gong X, Zhang X and Sun R: LncRNA HIF1A-AS2 promotes osteosarcoma progression by acting as a sponge of miR-129-5p. Aging (Albany NY). 11:11803–11813. 2019. View Article : Google Scholar

70 

Fu D, Lu C, Qu X, Li P, Chen K, Shan L and Zhu X: LncRNA TTN-AS1 regulates osteosarcoma cell apoptosis and drug resistance via the miR-134-5p/MBTD1 axis. Aging (Albany NY). 11:8374–8385. 2019. View Article : Google Scholar

71 

Li S, Liu F, Pei Y, Wang W, Zheng K and Zhang X: Long noncoding RNA TTN-AS1 enhances the malignant characteristics of osteosarcoma by acting as a competing endogenous RNA on microRNA-376a thereby upregulating dickkopf-1. Aging (Albany NY). 11:7678–7693. 2019. View Article : Google Scholar

72 

Ba Z, Gu L, Hao S, Wang X, Cheng Z and Nie G: Downregulation of lncRNA CASC2 facilitates osteosarcoma growth and invasion through miR-181a. Cell Prolif. 51:e124092018. View Article : Google Scholar

73 

Wang Z, Liu Z and Wu S: Long non-coding RNA CTA sensitizes osteosarcoma cells to doxorubicin through inhibition of autophagy. Oncotarget. 8:31465–31477. 2017. View Article : Google Scholar

74 

Ye K, Wang S, Zhang H, Han H, Ma B and Nan W: Long noncoding RNA GAS5 suppresses cell growth and epithelial-mesenchymal transition in osteosarcoma by regulating the miR-221/ARHI pathway. J Cell Biochem. 118:4772–4781. 2017. View Article : Google Scholar

75 

Kun-Peng Z, Xiao-Long M and Chun-Lin Z: LncRNA FENDRR sensitizes doxorubicin-resistance of osteosarcoma cells through down-regulating ABCB1 and ABCC1. Oncotarget. 8:71881–71893. 2017. View Article : Google Scholar

76 

Zhao J and Ma ST: Downregulation of lncRNA H19 inhibits migration and invasion of human osteosarcoma through the NF-κB pathway. Mol Med Rep. 17:7388–7394. 2018.

77 

Zhou S, Yu L, Xiong M and Dai G: LncRNA SNHG12 promotes tumorigenesis and metastasis in osteosarcoma by upregulating Notch2 by sponging miR-195-5p. Biochem Biophys Res Commun. 495:1822–1832. 2018. View Article : Google Scholar

78 

Ji S, Wang S, Zhao X and Lv L: Long noncoding RNA NEAT1 regulates the development of osteosarcoma through sponging miR-34a-5p to mediate HOXA13 expression as a competitive endogenous RNA. Mol Genet Genomic Med. 7:e6732019. View Article : Google Scholar

79 

Guan H, Shang G, Cui Y, Liu J, Sun X, Cao W, Wang Y and Li Y: Long noncoding RNA APTR contributes to osteosarcoma progression through repression of miR-132-3p and upregulation of yes-associated protein 1. J Cell Physiol. 234:8998–9007. 2019. View Article : Google Scholar

80 

Bielack S, Carrle D and Casali PG; ESMO Guidelines Working Group: Osteosarcoma: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 20(Suppl): S137–S139. 2009. View Article : Google Scholar

81 

Dong Y, Liang G, Yuan B, Yang C, Gao R and Zhou X: MALAT1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/Akt pathway. Tumour Biol. 36:1477–1486. 2015. View Article : Google Scholar

82 

Ma B, Li M, Zhang L, Huang M, Lei JB, Fu GH, Liu CX, Lai QW, Chen QQ and Wang YL: Upregulation of long non-coding RNA TUG1 correlates with poor prognosis and disease status in osteosarcoma. Tumour Biol. 37:4445–4455. 2016. View Article : Google Scholar

83 

Holohan C, Van Schaeybroeck S, Longley DB and Johnston PG: Cancer drug resistance: An evolving paradigm. Nat Rev Cancer. 13:714–726. 2013. View Article : Google Scholar

84 

Nigro JM, Cho KR, Fearon ER, Kern SE, Ruppert JM, Oliner JD, Kinzler KW and Vogelstein B: Scrambled exons. Cell. 64:607–613. 1991. View Article : Google Scholar

85 

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

86 

Lei K, Bai H, Wei Z, Xie C, Wang J, Li J and Chen Q: The mechanism and function of circular RNAs in human diseases. Exp Cell Res. 368:147–158. 2018. View Article : Google Scholar

87 

Chen LL and Yang L: Regulation of circRNA biogenesis. RNA Biol. 12:381–388. 2015. View Article : Google Scholar

88 

Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al: Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 495:333–338. 2013. View Article : Google Scholar

89 

de Almeida RA, Fraczek MG, Parker S, Delneri D and O'Keefe RT: Non-coding RNAs and disease: The classical ncRNAs make a comeback. Biochem Soc Trans. 44:1073–1078. 2016. View Article : Google Scholar

90 

Han B, Chao J and Yao H: Circular RNA and its mechanisms in disease: From the bench to the clinic. Pharmacol Ther. 187:31–44. 2018. View Article : Google Scholar

91 

Wu Y, Xie Z, Chen J, Chen J, Ni W, Ma Y, Huang K, Wang G, Wang J, Ma J, et al: Circular RNA circTADA2A promotes osteosarcoma progression and metastasis by sponging miR-203a-3p and regulating CREB3 expression. Mol Cancer. 18:732019. View Article : Google Scholar

92 

Song YZ and Li JF: Circular RNA hsa_circ_0001564 regulates osteosarcoma proliferation and apoptosis by acting miRNA sponge. Biochem Biophys Res Commun. 495:2369–2375. 2018. View Article : Google Scholar

93 

Kun-Peng Z, Xiao-Long M and Chun-Lin Z: Overexpressed circPVT1, a potential new circular RNA biomarker, contributes to doxorubicin and cisplatin resistance of osteosarcoma cells by regulating ABCB1. Int J Biol Sci. 14:321–330. 2018. View Article : Google Scholar

94 

Huang L, Chen M, Pan J and Yu W: Circular RNA circNASP modulates the malignant behaviors in osteosarcoma via miR-1253/FOXF1 pathway. Biochem Biophys Res Commun. 500:511–517. 2018. View Article : Google Scholar

95 

Wu Z, Shi W and Jiang C: Overexpressing circular RNA hsa_ circ_0002052 impairs osteosarcoma progression via inhibiting Wnt/β-catenin pathway by regulating miR-1205/APC2 axis. Biochem Biophys Res Commun. 502:465–471. 2018. View Article : Google Scholar

96 

Ren C, Liu J, Zheng B, Yan P, Sun Y and Yue B: The circular RNA circ-ITCH acts as a tumour suppressor in osteosarcoma via regulating miR-22. Artif Cells Nanomed Biotechnol. 47:3359–3367. 2019. View Article : Google Scholar

97 

Li H, Lan M, Liao X, Tang Z and Yang C: Circular RNA cir-ITCH promotes osteosarcoma migration and invasion through cir-ITCH/miR-7/EGFR pathway. Technol Cancer Res Treat. 19:15330338198987282020.

98 

Xiao-Long M, Kun-Peng Z and Chun-Lin Z: Circular RNA circ_HIPK3 is down-regulated and suppresses cell proliferation, migration and invasion in osteosarcoma. J Cancer. 9:1856–1862. 2018. View Article : Google Scholar

99 

Jin Y, Li L, Zhu T and Liu G: Circular RNA circ_0102049 promotes cell progression as ceRNA to target MDM2 via sponging miR-1304-5p in osteosarcoma. Pathol Res Pract. 215:1526882019. View Article : Google Scholar

100 

Li L, Guo L, Yin G, Yu G, Zhao Y and Pan Y: Upregulation of circular RNA circ_0001721 predicts unfavorable prognosis in osteosarcoma and facilitates cell progression via sponging miR-569 and miR-599. Biomed Pharmacother. 109:226–232. 2019. View Article : Google Scholar

101 

Li JF and Song YZ: Circular RNA GLI2 promotes osteosarcoma cell proliferation, migration, and invasion by targeting miR-125b-5p. Tumour Biol. 39:10104283177099912017. View Article : Google Scholar

102 

Li S, Pei Y, Wang W, Liu F, Zheng K and Zhang X: Circular RNA 0001785 regulates the pathogenesis of osteosarcoma as a ceRNA by sponging miR-1200 to upregulate HOXB2. Cell Cycle. 18:1281–1291. 2019. View Article : Google Scholar

103 

Cao J and Liu XS: Circular RNA 0060428 sponges miR-375 to promote osteosarcoma cell proliferation by upregulating the expression of RPBJ. Gene. 740:1445202020. View Article : Google Scholar

104 

Li S, Sun X, Miao S, Lu T, Wang Y, Liu J and Jiao W: hsa_circ_0000729, a potential prognostic biomarker in lung adenocarcinoma. Thorac Cancer. 9:924–930. 2018. View Article : Google Scholar

105 

Li XM, Ge HM, Yao J, Zhou YF, Yao MD, Liu C, Hu HT, Zhu YX, Shan K, Yan B and Jiang Q: Genome-wide identification of circular RNAs as a novel class of putative biomarkers for an ocular surface disease. Cell Physiol Biochem. 47:1630–1642. 2018. View Article : Google Scholar

106 

Zhang H, Wang G, Ding C, Liu P, Wang R, Ding W, Tong D, Wu D, Li C, Wei Q, et al: Increased circular RNA UBAP2 acts as a sponge of miR-143 to promote osteosarcoma progression. Oncotarget. 8:61687–61697. 2017. View Article : Google Scholar

107 

Liu X, Abraham JM, Cheng Y, Wang Z, Wang Z, Zhang G, Ashktorab H, Smoot DT, Cole RN, Boronina TN, et al: Synthetic circular RNA functions as a miR-21 sponge to suppress gastric carcinoma cell proliferation. Mol Ther Nucleic Acids. 13:312–321. 2018. View Article : Google Scholar

108 

Xu S, Gong Y, Yin Y, Xing H and Zhang N: The multiple function of long noncoding RNAs in osteosarcoma progression, drug resistance and prognosis. Biomed Pharmacother. 127:1101412020. View Article : Google Scholar

109 

Yin F, Wang Z, Jiang Y, Zhang T, Wang Z, Hua Y, Song Z, Liu J, Xu W, Xu J, et al: Reduction-responsive polypeptide nanomedicines significantly inhibit progression of orthotopic osteosarcoma. Nanomedicine. 23:1020852020. View Article : Google Scholar

110 

Matsui M and Corey DR: Non-coding RNAs as drug targets. Nat Rev Drug Discov. 16:167–179. 2017. View Article : Google Scholar

111 

Li Z and Rana TM: Therapeutic targeting of microRNAs: Current status and future challenges. Nat Rev Drug Discov. 13:622–638. 2014. View Article : Google Scholar

112 

Wang WT, Han C, Sun YM, Chen TQ and Chen YQ: Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol. 12:552019. View Article : Google Scholar

113 

Huang KW, Lai YT, Chern GJ, Huang SF, Tsai CL, Sung YC, Chiang CC, Hwang PB, Ho TL, Huang RL, et al: Galactose derivative-modified nanoparticles for efficient siRNA delivery to hepatocellular carcinoma. Biomacromolecules. 19:2330–2339. 2018. View Article : Google Scholar

114 

Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, Hoekstra M, Kandasamy P, Kel'in AV, Milstein S, et al: Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 136:16958–16961. 2014. View Article : Google Scholar

115 

Wen JJ, Ma YD, Yang GS and Wang GM: Analysis of circulating long non-coding RNA UCA1 as potential biomarkers for diagnosis and prognosis of osteosarcoma. Eur Rev Med Pharmacol Sci. 21:498–503. 2017.

116 

Huang JZ, Chen M, Chen D, Gao XC, Zhu S, Huang H, Hu M, Zhu H and Yan GR: A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cell. 68:171–184.e6. 2017. View Article : Google Scholar

117 

Yang Y, Gao X, Zhang M, Yan S, Sun C, Xiao F, Huang N, Yang X, Zhao K, Zhou H, et al: Novel role of FBXW7 circular RNA in repressing glioma tumorigenesis. J Natl Cancer Inst. 110:3042018. View Article : Google Scholar

118 

Ribatti D: The concept of immune surveillance against tumors. The first theories Oncotarget. 8:7175–7180. 2017. View Article : Google Scholar

119 

Ji X, Wang E and Tian F: MicroRNA-140 suppresses osteosarcoma tumor growth by enhancing anti-tumor immune response and blocking mTOR signaling. Biochem Biophys Res Commun. 495:1342–1348. 2018. View Article : Google Scholar

120 

Li S, Li X, Xue W, Zhang L, Yang LZ, Cao SM, Lei YN, Liu CX, Guo SK, Shan L, et al: Screening for functional circular RNAs using the CRISPR-Cas13 system. Nat Methods. 18:51–59. 2021. View Article : Google Scholar

Related Articles

Journal Cover

September-2021
Volume 59 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
Yang G, Wu Y, Wan R, Sang H, Liu H and Huang W: The role of non‑coding RNAs in the regulation, diagnosis, prognosis and treatment of osteosarcoma (Review). Int J Oncol 59: 69, 2021
APA
Yang, G., Wu, Y., Wan, R., Sang, H., Liu, H., & Huang, W. (2021). The role of non‑coding RNAs in the regulation, diagnosis, prognosis and treatment of osteosarcoma (Review). International Journal of Oncology, 59, 69. https://doi.org/10.3892/ijo.2021.5249
MLA
Yang, G., Wu, Y., Wan, R., Sang, H., Liu, H., Huang, W."The role of non‑coding RNAs in the regulation, diagnosis, prognosis and treatment of osteosarcoma (Review)". International Journal of Oncology 59.3 (2021): 69.
Chicago
Yang, G., Wu, Y., Wan, R., Sang, H., Liu, H., Huang, W."The role of non‑coding RNAs in the regulation, diagnosis, prognosis and treatment of osteosarcoma (Review)". International Journal of Oncology 59, no. 3 (2021): 69. https://doi.org/10.3892/ijo.2021.5249