Mechanisms and functions of long non‑coding RNAs in glioma (Review)

  • Authors:
    • Xingjie Chen
    • Gaochao Guo
    • Yalin Lu
    • Shuaishuai Wang
    • Yu Zhang
    • Qiang Huang
  • View Affiliations

  • Published online on: February 1, 2021     https://doi.org/10.3892/or.2021.7960
  • Article Number: 9
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioma is one of the most common primary malignancies of the adult central nervous system with malignancy grades between I‑IV. Among these four grades, glioblastoma is the most malignant and aggressive type of tumor and is characterized by a poor prognosis, high recurrence rate and short median survival time after initial diagnosis. Existing treatments, such as radiotherapy, chemotherapy and surgical resection, have poor therapeutic effects; therefore, it is necessary to discover novel targeted therapies to enhance the curative effect and improve prognosis. Recently, increasing evidence has shown that long non‑coding RNAs (lncRNAs) participate in the vast majority of key physiological and pathological processes. Moreover, aberrant expression levels of lncRNAs are closely associated with the occurrence and development of glioma and other malignant phenotypes. The present review summarizes new insights into the functions and mechanisms of lncRNAs at the epigenetic, transcriptional and post‑transcriptional levels, describes their ability to encode functional peptides in glioma and discusses their clinical potential as new biomarkers and prospective therapeutic targets.

Introduction

Glioma, the most prevalent primary malignant cancer of the central nervous system of adults (1), is characterized by difficulty of early diagnosis, a high recurrence rate and a poor prognosis, especially for advanced and high-grade types (24). The World Health Organization has classified gliomas into four grades based on their histopathology and clinical prognosis: Grades I and II are routinely viewed as low-grade gliomas, while grades III and IV are deemed as high-grade gliomas (5). Glioblastoma multiforme (GBM), the most malignant and aggressive type, has a median survival time of 12–14 months after initial diagnosis, longer than that of only pancreatic and lung cancer (6). As research has progressed in recent years, medical technology has constantly improved. However, due to the rapid proliferation, high invasive potential and radio/chemotherapeutic resistance of GBM, current treatments, including surgical resection, radiotherapy and chemotherapy, do not have optimal effects, and patients with GBM still have a poor prognosis (7). Therefore, it is necessary to clarify in detail the pathogenetic mechanisms of glioma to achieve improved therapeutic effects and longer survival times in patients after initial diagnosis.

Long non-coding RNAs (lncRNAs) are transcripts that contain >200 nucleotides (nt), but lack protein-coding capacity (8). The structure of lncRNAs is typically similar to that of mRNAs, which have 5′-m7G caps and 3′-poly(A) tails (9), but they are more tissue-specific than mRNAs, indicating that lncRNAs may have specific biological roles and functional mechanisms (10). Recently, an increasing number of studies has reported that lncRNAs participate in a variety of cellular physiological processes, including stemness, tumorigenesis, proliferation, invasion, angiogenesis and drug resistance, by regulating gene expression at the epigenetic, transcriptional and post-transcriptional levels (11,12). It has been demonstrated that most lncRNAs can recruit regulatory complexes through RNA-protein interactions to affect the expression levels of nearby genes, while some lncRNAs can also function as local regulators (13). Overexpression, deficiency or mutation of lncRNA genes has been reported to be associated with numerous human diseases, such as cancer, cardiovascular diseases, metabolic diseases and inflammation (1417). Similarly, in glioma, progressive evidence has illustrated that abnormal expression levels of lncRNAs are closely associated with the occurrence and development of glioma and other malignant phenotypes (12).

Technological advancements, especially the completion of the human genome sequencing, have allowed the discovery of an increasing number of lncRNAs with different targets and functions (18); however, the specific mechanisms and functions of lncRNAs remain unclear. The present review summarizes the functions and mechanisms of lncRNAs at the molecular level in glioma and provides some prospects for their use in the therapy and diagnosis of glioma.

Classification of lncRNAs

Previous human genome studies have reported that lncRNAs are transcripts produced by RNA polymerase II (RNAPol II) that contain >200 nt, but lack an open reading frame (ORF) for translation into proteins (19,20). Accumulating studies have found that lncRNAs are important players at almost every level of gene function and regulation (11,14,21). Based on their genomic location relative to neighboring protein-coding genes and their molecular characteristics, lncRNAs can be classified into five categories: Sense, antisense, bidirectional (22), intronic (23) and intergenic [long intergenic ncRNAs (lincRNAs)] (8) (Fig. 1). Through the classification of unspliced and spliced lncRNAs from mouse and human embryonic stem cells (24), most lncRNAs are either localized to enhancer regions (~20%), called enhancer RNAs (eRNAs) (25,26), or associated with upstream antisense RNAs, which are derived from loci near transcription start sites (TSSs) of coding RNAs (60–70%) (24,27). The remaining lncRNAs are derived from transcripts that overlap with coding sequences (~5%) or from more distal, unannotated regions (~5%) (27). The latter lncRNAs are usually called lincRNAs (27,28). Moreover, eRNAs and promoter upstream transcripts, which are transcribed from enhancers and promoters, respectively, are functionally similar to regulatory DNA molecules (23). For example, eRNAs can promote the interactions of enhancers and promoters to activate target genes (29). However, although a number of lncRNAs are associated with annotated genomic regions, some intervening lncRNAs come from separate transcriptional elements that do not overlap with coding sequences or enhancers; these loci have their own promoters and can function through chromatin modifications as protein-coding genes (30). The lncRNAs metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and nuclear enriched abundant transcript 1 (NEAT1), which are well-known structural intervening lncRNAs, belong to this category (14). Considering the close association between the structures and locations of lncRNAs and their stability and functional mechanisms (14), the identification of lncRNA secondary structures and classification is anticipated to serve a key role in the research and clinical application of lncRNAs (19).

Differential expression of lncRNAs in glioma

Aberrant expression levels of lncRNAs can mediate cell biological processes, such as proliferation, stemness, drug resistance and angiogenesis, and accelerate the progression of glioma malignancy (31). Therefore, an increasing number of studies have focused on the analysis of lncRNA gene expression profiles in GBM to identify the detailed mechanisms. By comparing the expression levels of mRNAs and lncRNAs between GBM and normal brain tissues, Han et al (32) found that 654 lncRNAs were upregulated and 654 lncRNAs were downregulated in GBM. Moreover, 104 matched lncRNA-mRNA pairs were identified, and 90 lncRNAs and 81 lncRNA-mRNA pairs were found to be differentially expressed (32). Chen et al (33) used the significant analysis of microarray (SAM) method in a training dataset to analyze the differential expression of lncRNAs between GBM and normal brain tissues, identifying 299 lncRNAs with differential expression, of which 133 were upregulated and 166 were downregulated in GBM compared with in normal brain tissues (33). The SAM method was then used to analyze the differential expression of lncRNAs between low-grade and high-grade gliomas in the training dataset, and 47 lncRNAs were found to be differentially expressed between low-grade and high-grade gliomas (33). By comparing the expression levels of mRNAs and lncRNAs between normal brain tissues and GBM, Li et al (34) found that 398 lncRNAs were differentially expressed and 1,995 mRNAs were dysregulated in GBM. Among these differentially expressed lncRNAs, 98 participated in 32 gene functions and 30 molecular pathways associated with tumorigenesis, development and metastasis of glioma (34,35).

Aberrant expression levels of lncRNAs in glioma have been identified to serve a crucial role in the tumorigenesis, proliferation and invasion of glioma cells (31). Moreover, abnormal lncRNA expression profiles in clinical glioma specimens are closely associated with histological differentiation and malignancy grade, which have crucial clinical significance in early glioma diagnosis of subclassifications and in patient prognosis (36).

lncRNAs regulate the expression of glioma genes at the epigenetic level

In glioma, lncRNAs can regulate gene expression at the epigenetic level before transcription by recruiting chromatin modifiers such as enhancer of zeste homolog 2 (EZH2)/polycomb repressive complex 2 (PRC2) (37) and WD repeat domain 5/trithorax group proteins (38) to a specific genomic location as scaffolds to regulate the trimethylation or acetylation of histone H3 (31). Therefore, lncRNAs can participate in the regulation of glioma phenotypes, such as tumorigenic behaviors, proliferation, invasion and drug resistance (37,39).

The lncRNA NEAT1 is distributed mainly in the cell nucleus and has two transcripts, NEAT1_1 (3.7 kb) and NEAT1_2 (23 kb) (40). The lncRNA NEAT1 has been demonstrated to promote the occurrence of numerous types of cancer, such as colorectal cancer, breast cancer, liver cancer and glioma (4144). A study on the detailed mechanism of lncRNAs have suggested that the lncRNA NEAT1 can be activated by the upstream EGFR signaling pathway; in addition, it can act as a scaffold to recruit and interact with the chromosome modification enzyme EZH2 (37). Moreover, the interaction between the lncRNA NEAT1 and EZH2 can promote histone H3 trimethylation in the promoter regions of Axin2, inhibitor of β-catenin and T-cell factor and glycogen synthase kinase 3β, which are negative regulatory factors of the WNT/β-catenin signaling pathway (45,46), to silence these downstream target genes, thereby activating the WNT/β-catenin signaling pathway to promote glioma tumorigenesis and proliferation (37) (Fig. 2A). Similarly, in neuroblastoma, the lncRNA neuroblastoma-associated transcript-1 (NBAT-1) functions as a scaffold to recruit and interact with EZH2 to downregulate the expression levels of NBAT-1/EZH2 target genes, such as SRY-box transcription factor 9 (SOX9), oncostatin M receptor and versican, to decrease the risk of neuroblastoma (47). The lncRNA temozolomide-associated lncRNA in GBM recurrence (TALC), with a total length of 418 nt and containing two exons, is highly expressed in temozolomide (TMZ)-resistant glioma cells (39). The lncRNA TALC, induced by AKT-mediated TMZ resistance in GBM, can control the acetylation of histone H3 on lysine 27 (H3K27) in the promoter regions of O6 methylguanine-DNA methyltransferase (MGMT) to trap microRNA (miRNA/miR)-20b-3p, activate c-MET and increase MGMT expression (39). The lncRNA ZFAT antisense RNA 1 (ZFAT-AS1), derived from an imprinted gene located on the long arm of the human genome, can bind to the relevant EZH2 subunit of the PRC2 complex to catalyze histone H3K27 methylation to inhibit transcription of the downstream gene caudal type homeobox 2 (CDX2), in turn promoting glioma cell proliferation, migration and invasion (48).

Transcriptional regulation by lncRNAs

Transcription is an important cellular physiological process that transfers DNA genetic material to the cytoplasm as RNA (49). Based on their patterns of interaction with proteins, lncRNAs regulate transcriptional processes via three mechanisms.

Enhancer-like activity

The transcription of most genes involves the interaction of a proximal promoter with more distant enhancer elements (50). Enhancers are usually located far from the transcriptional initiation site and interact with tissue-specific transcription factors that perform their function to modulate the differential expression of genes (51). Kim et al (52) found that some ncRNAs can be bidirectionally transcribed from activated enhancers, and the expression levels of these eRNAs are associated with the activity of the enhancer. Follow-up studies have shown that eRNAs may exert enhancer-like effects, such as remodeling chromatin, promoting chromatin accessibility (53) and bridging a distal enhancer with a proximal promoter (54). A class of lncRNAs similar to eRNAs is composed of activating ncRNAs (ncRNA-as), which have a transcriptional activation function; these lncRNAs are transcribed from independent loci, not from enhancers, and compose a class of functional molecules that can regulate the enhancing effect (25,55). Depletion of lncRNAs may lead to elevated expression levels of adjacent protein-coding genes at numerous loci in the human genome (25,55). This promotion of gene expression is mediated by RNA, and studies have shown that ncRNA-as can moderate this RNA-dependent transcriptional responsiveness in cis (25,55). These enhancer-like effects of lncRNAs may be functional mechanisms broadly used to modulate gene expression. Moreover, both eRNA and ncRNA-as can link the enhancer and promoter element of the coding gene as a scaffold for a protein complex, thereby regulating the transcription process (56) (Fig. 2B).

Binding to RNAPol II to interfere with transcription

lncRNAs can interfere with transcription by binding to RNAPol II (57). For example, the mRNA of the master regulator heat shock B2, which is transcribed from retrotransposons distributed broadly in the mouse genome (58) and is derived from the short interspersed nuclear element family (59), can induce transcriptional inhibition by interacting directly with RNAPol II and decreasing the production of a functional closed preinitiation complex (PIC) (60) (Fig. 2C).

Acting as a ‘decoy’ for transcription factors

Transcription factors, which are a class of DNA binding proteins, can specifically bind to specific sequences in the TSS of protein-coding genes to modulate the transcription process (61). Accumulating evidence has demonstrated that lncRNAs can interact with transcription factors at the promoter regions of coding genes to regulate transcription (62). For instance, the lncRNA PANDA, transcribed from the CDKN1A promoter, interacts with the nuclear transcription factor Y subunit α or PRCs (PRC1 and PRC2) to either accelerate or suppress senescence (63) (Fig. 2D).

In glioma, lncRNAs generally bind to transcription factors at the promoter region of target genes to regulate transcription. For instance, the lncRNA paxillin interacting protein 1-antisense RNA 1 (PAXIP1-AS1), a critical mediator of cell death, has been found to recruit the transcription factor ETS proto-oncogene 1 (ETS1) to the promoter region of kinesin family member 14 (KIF14) to upregulate its expression (64). Thus, the lncRNA PAXIP1-AS1 promotes glioma cell migration, invasion and angiogenesis via the PAXIP1-AS1/ETS1/KIF14 axis (64). Furthermore, the lncRNA growth arrest-specific transcription 5 (GAS5), a member of the 5′ terminal oligopyrimidine class of genes (65), can inhibit tumorigenesis by recruiting the transcription factor TFAP2A to its promoter region under physiological conditions (66). However, an indel genetic polymorphism of the lncRNA GAS5 increases glioma susceptibility by blocking the binding of the transcription factor TFAP2A (66).

Post-transcriptional regulation by lncRNAs

In addition to participating in transcriptional and epigenetic regulation, lncRNAs often modulate gene expression by post-transcriptional regulation (11). A number of published results have shown that lncRNAs can act as competing endogenous RNAs (ceRNAs) to decrease the expression levels of specific mRNAs (67), mediate alternative splicing (68) and affect mRNA stability (69) and intercellular communication (70). This section focusses on these functions to summarize the post-transcriptional mechanisms of lncRNAs in glioma.

ceRNAs

ceRNAs are a class of transcripts that can affect the expression levels of mRNAs at the post-transcriptional level by competing for binding to miRNAs (67). In 2011, a review proposed the ceRNA hypothesis, which states that mRNAs contain miRNA response elements (MREs) to which miRNAs can specifically bind, and that ncRNA species, including lncRNAs, circular RNAs (circRNAs), pseudogene RNAs and mRNA-mRNA pairs, also contain MREs and can potentially compete for a limited pool of miRNAs to regulate gene expression (71) (Fig. 3A). Subsequently, accumulating evidence has shown that lncRNAs may function as ceRNAs to sponge miRNAs and prevent them from interacting with their downstream target genes, thereby silencing these genes to affect the expression levels of the corresponding proteins (18,7275).

The lncRNA CRNDE, encoded by the colorectal neoplasia differentially expressed gene, has been shown to be the most highly upregulated lncRNA among 129 differentially expressed lncRNAs in glioma (76). Li et al (75) demonstrated that the lncRNA CRNDE can act as a ceRNA to interact with miR-136-5p, thus competitively inhibiting miR-136-5p-mediated inhibition of Wnt-2 and Bcl-2. This event leads to an increase in the post-transcriptional expression levels of Wnt-2 and Bcl-2, and in the activation of the PI3K/AKT/mTOR signaling pathway (75). Moreover, Zheng et al (77,78) found that the lncRNA CRNDR can facilitate the proliferation, invasion and migration of glioma cells, and decrease their apoptosis through competitive inhibition of miR-384 and miR-186. Similarly, the lncRNA X-inactive specific transcript (XIST), transcribed from the X inactivation centre (79), can also act as a ceRNA of miR-126 to regulate the IRS1/PI3K/Akt signaling pathway in order to promote the viability, migration, invasion, apoptosis resistance and glucose metabolism of GBM cells (80). The lncRNAs HOTAIR and MEG3 can promote or suppress, respectively, the proliferation, invasion and migration of glioma cells by acting as ceRNAs of miR-141 and miR-19a (74).

In summary, lncRNAs act as ceRNAs through a ceRNA/miRNA/mRNA regulatory axis at the post-trans-criptional level. The concept of a ceRNA/miRNA/mRNA regulatory pathway offers a novel concept for studying the underlying molecular mechanisms of glioma, as well as improves the understanding of lncRNAs and identifies a specific and sensitive profile of interactions between lncRNAs and mRNAs, which may contribute to the development of methods for both the earlier diagnosis and targeted therapy of glioma.

Alternative splicing

Alternative splicing of pre-mRNAs serves a crucial role in the regulation and diversity of gene functions, and is used by higher eukaryotes to increase the complexity of the transcriptome and proteome (81). Alternative splicing is mediated mainly by trans-acting protein factors, including serine/arginine-rich (SR)-associated proteins, heterogeneous nuclear ribonucleoproteins, the SR family of nuclear phosphoproteins (SR proteins) and small nuclear ribonucleoproteins (82). Among these factors, SR proteins, a class of proteins that can specifically bind to RNA, typically serve a key role in alternative splicing (83). Some results have shown that lncRNAs in glioma can modulate alternative splicing by controlling these trans-acting protein factors (68).

The lncRNA MALAT1 is one of the most abundant lncRNAs in normal human physiological tissues (84). Initially, lncRNA MALAT1 was known as a prognostic molecular marker of advanced lung cancer (85); however, previous studies have found an association between MALAT1 and other types of cancer, such as pancreatic cancer, prostate cancer, breast cancer, glioma and leukemia (8690). Tripathi et al (91) reported that the lncRNA MALAT1 can bind to SR proteins and act as a molecular sponge to modulate the phosphorylation levels of SR proteins. By regulating the phosphorylation status of SR proteins, the lncRNA MALAT1 can indirectly mediate the intranuclear transfer of SR proteins between nuclear speckles and transcription sites to control their distribution to nuclear speckles, thereby regulating alternative splicing (91) (Fig. 3C). Furthermore, by modulating the activation levels of SR proteins, MALAT1 regulates alternative splicing, as well as controlling other post-transcriptional gene regulatory mechanisms associated with SR proteins, such as translation, nonsense-mediated decay and RNA export (92,93).

mRNA stability and protein modification

Regulation of mRNA stability and protein modification are important processes in post-transcriptional regulation. From the perspective of modulating mRNA stability, lncRNAs can either enhance mRNA stability by forming protective lncRNA-mRNA duplexes (94) or accelerate mRNA degradation by recruiting RNA-binding proteins, such as polypyrimidine tract binding protein 1 (PTBP1), to target pre-mRNAs in order to promote mRNA degradation (95). For instance, the lncRNA PTB-AS can directly bind to the PTBP1 3′-untranslated region via staphylococcal nuclease domain-containing 1 to stabilize PTBP1 mRNA, which significantly promotes the proliferation and migration of glioma cells (96). The lncRNA FMR1 autosomal homolog 1 can maintain the stability of miR-17-92a-1 cluster host gene mRNA to upregulate the downstream protein TAL bHLH transcription factor 1 in order to regulate the biological behavior of glioma cells (97).

Moreover, lncRNAs can directly interact with key proteins of signaling pathways, thus influencing their expression levels and regulating their functions (31). For example, in addition to acting as a ceRNA, the lncRNA CRNDE can also bind to the P70S6K protein, a direct downstream effector of the mTOR signaling pathway, and enhance its phosphorylation level, suggesting that CRNDE may modulate the mTOR signaling pathway by modifying this downstream protein (98).

Intercellular communication

Previous studies have shown that lncRNAs can serve important roles in intercellular communication. Barile and Vassalli (99) found that lncRNAs can be incorporated into exosomes and secreted into recipient cells passing through blood vessels, thereby controlling target signaling pathways and regulating cell phenotypes. Exosomes are the most clearly defined vesicles known to date; these vesicles have diameters ranging between 40 and 150 nm, and can be secreted by numerous different types of cells (100). Their promising diagnostic and therapeutic potential and value have received increasing attention, particularly in cancer, such as glioma, breast cancer, prostate cancer and pancreatic cancer (101106). Accumulated evidence has demonstrated that exosomes can decelerate lncRNA degradation in the circulation and that exosomal lncRNAs can be utilized for early diagnosis of cancer (107). Moreover, a number of studies have confirmed that exosomal lncRNAs can function as intercellular carriers to transmit cellular messenger molecules, including lncRNAs (108110).

For instance, a recent study identified a lncRNA, lncSBF2-AS1 (ENSG00000246273), that can be activated by the transcription factor ZEB1 and promote the TMZ resistance of glioma cells (110). Zhang et al (110) revealed that lncSBF2-AS1 released from TMZ-resistant glioma cells via exosomes can endow neighboring TMZ-sensitive cells with a TMZ-resistant phenotype; by contrast, deficiency of exosomal lncSBF2-AS1 can partially reverse the drug resistance phenotype of the parental cells, suggesting that exosomal lncSBF2-AS1 can induce TMZ resistance via intercellular communication (Fig. 3B). Similarly, the lncRNA HOTAIR can be secreted into adjacent cells via serum exosomes and modulate TMZ resistance through the miR-519a-3p/ribonucleotide reductase catalytic subunit M1 axis (111).

lncRNAs encode tumor-associated functional polypeptides

In addition to the aforementioned functional mechanisms, lncRNAs have been proven to have other functions in normal and cancer tissues, such as encoding functional peptides. As mass spectrometry, deep RNA sequencing and bioinformatics techniques have improved, accumulating evidence suggests that lncRNAs that were previously considered non-coding may have the ability to encode small biologically active peptides (112). lncRNAs may have small ORFs (sORFs) that can be translated into small peptides containing <100 amino acids (aa) (113). Some studies have identified that these functional peptides encoded by lncRNA sORFs can regulate biological processes and influence tumorigenesis, proliferation, invasion and metastasis.

Wang et al (114) identified that the lncRNA LINC00908 can encode a 60-aa functional peptide known as ASPRS and is differentially expressed in normal and triple-negative breast cancer tissues. ASPRS is a small regulatory peptide of STAT3 and can directly bind to STAT3 and downregulate its phosphorylation to decrease VEGF expression (114). Through regulation of VEGF, the ASPRS protein can decrease angiogenesis and suppress tumorigenesis in breast cancer (114). Moreover, in glioma, the circRNA SNF2 histone linker PHD RING helicase (SHPRH) can be translated into SHPRH-146-aa; this protein protects the full-length SHPRH from degradation by ubiquitin proteases to inhibit cell proliferation and tumorigenicity (115,116). The lncRNA LINC-PINT can form the circular molecule circPINT by self-cyclization, which contains an sORF that encodes the functional peptide PINT-78-aa; this peptide can directly interact with polymerase-associated factor 1 (PAF1) to regulate the PAF1/POLII complex, inhibit transcriptional elongation of downstream oncogenes, including c-Myc, sox-2, cyclin D1 and cpeb1, and suppress the proliferation and tumorigenesis of GBM cells (117).

Clinical potential of lncRNAs

Based on these functional mechanisms of lncRNAs, we speculate that lncRNAs may function as novel biomarkers and therapeutic targets in glioma to improve patient prognosis. For example, lncRNAs have highly specific expression patterns in different cells and tissues; thus, they may be used to distinguish different subtypes of glioma and evaluate patient prognosis (10). A previous study revealed that using small interfering RNAs to target tumor-associated lncRNAs achieves therapeutic effects (118). Moreover, given that lncRNAs always interact with other molecules to regulate gene expression, the binding sites of these interactions may become new therapeutic targets using methods such as a peptide nuclein acid-based strategy, which can block the interaction between lncRNA HOTAIR and EZH2, subsequently decreasing HOTAIR-EZH2 complex activity (119). Based on the differential expression of lncRNAs between normal and glioma tissues, we speculate that lncRNAs may act as potential biomarkers to diagnose glioma in early stages. Considering that exosomes are extremely stable and are readily accessible in nearly all types of human biofluids, and that lncRNAs can be secreted into the circulation through packaging into exosomes (120), exosomal lncRNAs may be one of the most promising biomarkers. Since exosomes can cross the blood-brain barrier, the strategy of exploiting exosomes to deliver glioma-suppressive lncRNAs to target sites may be a promising therapeutic option for glioma (121).

Conclusions

Currently, accumulating evidence has proven that lncRNAs are closely associated with the malignant progression of cancer and serve important roles in the onset and progression of glioma. Therefore, the present review described the classification of lncRNAs, the functional mechanisms of lncRNAs in glioma at the epigenetic, transcriptional and post-transcriptional levels, and the ability of lncRNAs to encode functional peptides. However, numerous questions about lncRNAs remain unanswered. Considering the significance of lncRNAs in numerous physiological processes and their close association with the occurrence of diseases, research on lncRNAs is expected to grow exponentially in the future. We predict that further exploration will focus on the following aspects: Detecting the secondary structures of lncRNA interaction sites; investigating lncRNA binding patterns to seek new RNA-based targets; establishing complete ceRNA/miRNA/mRNA regulatory networks and translating these findings from theories to clinical applications; improving the identification and isolation of tumor-specific exosomal lncRNAs, further revealing the detailed mechanism underlying the intercellular transfer of exosomal lncRNAs; increasing the intracellular uptake efficiency and the relative stability of lncRNA-based drugs; accurately delivering lncRNAs to target sites to enhance the therapeutic effects of lncRNA-associated drugs; further studying the encoding function of lncRNAs; and evaluating the clinical potential of functional polypeptides. The understanding of lncRNAs remains incomplete, but the clinical potential of lncRNAs is worth exploring. The present review summarized new insights into the functional mechanisms of lncRNAs from different aspects and may be useful for future research in similar areas.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 81572490 and 81172405), the Tianjin Science and Technology Committee (grant no. 18JCZDJC98600) and the Science and Technology fund of Tianjin Binhai New Area Health and Family Planning Commission (grant no. 2018BWKZ003).

Availability of data and materials

Not applicable.

Authors' contributions

XC was a major contributor in writing the manuscript. GG provided the major ideas and outlines, and gave the final approval of the version to be published. YL contributed to conception and design, and acquisition of data. SW contributed to acquisition of data and revision of the text. YZ contributed to acquisition of data and revision of the figures. YL and QH confirmed the authenticity of the data. QH was the corresponding author and primarily responsible for revision of the manuscript. All authors read and approved the final 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.

References

1 

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P and Ellison DW: The 2016 world health organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 131:803–820. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Reni M, Mazza E, Zanon S, Gatta G and Vecht CJ: Central nervous system gliomas. Crit Rev Oncol Hematol. 113:213–234. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Chen R, Smith-Cohn M, Cohen AL and Colman H: Glioma subclassifications and their clinical significance. Neurotherapeutics. 14:284–297. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Alexander BM and Cloughesy TF: Adult glioblastoma. J Clin Oncol. 35:2402–2409. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Taylor OG, Brzozowski JS and Skelding KA: Glioblastoma multiforme: An overview of emerging therapeutic targets. Front Oncol. 9:9632019. View Article : Google Scholar : PubMed/NCBI

7 

Cheng J, Meng J, Zhu L and Peng Y: Exosomal noncoding RNAs in Glioma: Biological functions and potential clinical applications. Mol Cancer. 19:662020. View Article : Google Scholar : PubMed/NCBI

8 

Ulitsky I and Bartel DP: lincRNAs: Genomics, evolution, and mechanisms. Cell. 154:26–46. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Lagarde J, Uszczynska-Ratajczak B, Santoyo-Lopez J, Gonzalez JM, Tapanari E, Mudge JM, Steward CA, Wilming L, Tanzer A, Howald C, et al: Extension of human lncRNA transcripts by RACE coupled with long-read high-throughput sequencing (RACE-Seq). Nat Commun. 7:123392016. View Article : Google Scholar : PubMed/NCBI

10 

Deveson IW, Hardwick SA, Mercer TR and Mattick JS: The dimensions, dynamics, and relevance of the mammalian noncoding transcriptome. Trends Genet. 33:464–478. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Bonasio R and Shiekhattar R: Regulation of transcription by long noncoding RNAs. Annu Rev Genet. 48:433–455. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Yan Y, Xu Z, Li Z, Sun L and Gong Z: An insight into the increasing role of lncRNAs in the pathogenesis of gliomas. Front Mol Neurosci. 10:532017. View Article : Google Scholar : PubMed/NCBI

13 

Engreitz JM, Haines JE, Perez EM, Munson G, Chen J, Kane M, McDonel PE, Guttman M and Lander ES: Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature. 539:452–455. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Quinn JJ and Chang HY: Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 17:47–62. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Huang CK, Kafert-Kasting S and Thum T: Preclinical and clinical development of noncoding RNA therapeutics for cardiovascular disease. Circ Res. 126:663–678. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Kato M and Natarajan R: Epigenetics and epigenomics in diabetic kidney disease and metabolic memory. Nat Rev Nephrol. 15:327–345. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Atianand MK, Caffrey DR and Fitzgerald KA: Immunobiology of long noncoding RNAs. Annu Rev Immunol. 35:177–198. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Cheng Z, Li Z, Ma K, Li X, Tian N, Duan J, Xiao X and Wang Y: Long non-coding RNA XIST promotes glioma tumorigenicity and angiogenesis by acting as a molecular sponge of miR-429. J Cancer. 8:4106–4116. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Qian X, Zhao J, Yeung PY, Zhang QC and Kwok CK: Revealing lncRNA structures and interactions by sequencing-based approaches. Trends Biochem Sci. 44:33–52. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Mercer TR and Mattick JS: Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol. 20:300–307. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Rinn JL and Chang HY: Genome regulation by long noncoding RNAs. Annu Rev Biochem. 81:145–166. 2012. View Article : Google Scholar : PubMed/NCBI

23 

St Laurent G, Wahlestedt C and Kapranov P: The Landscape of long noncoding RNA classification. Trends Genet. 31:239–251. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Sigova AA, Mullen AC, Molinie B, Gupta S, Orlando DA, Guenther MG, Almada AE, Lin C, Sharp PA, Giallourakis CC and Young RA: Divergent transcription of long noncoding RNA/mRNA gene pairs in embryonic stem cells. Proc Natl Acad Sci USA. 110:2876–2881. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Ørom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, et al: Long noncoding RNAs with enhancer-like function in human cells. Cell. 143:46–58. 2010. View Article : Google Scholar : PubMed/NCBI

26 

de Santa F, Barozzi I, Mietton F, Ghisletti S, Polletti S, Tusi BK, Muller H, Ragoussis J, Wei CL and Natoli G: A large fraction of extragenic RNA pol II transcription sites overlap enhancers. PLoS Biol. 8:e10003842010. View Article : Google Scholar : PubMed/NCBI

27 

Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, et al: Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 458:223–227. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Ulitsky I, Shkumatava A, Jan CH, Sive H and Bartel DP: Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell. 147:1537–1550. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Fitz J, Neumann T, Steininger M, Wiedemann EM, Garcia AC, Athanasiadis A, Schoeberl UE and Pavri R: Spt5-mediated enhancer transcription directly couples enhancer activation with physical promoter interaction. Nat Genet. 52:505–515. 2020. View Article : Google Scholar : PubMed/NCBI

30 

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 : PubMed/NCBI

31 

Peng Z, Liu C and Wu M: New insights into long noncoding RNAs and their roles in glioma. Mol Cancer. 17:612018. View Article : Google Scholar : PubMed/NCBI

32 

Han L, Zhang K, Shi Z, Zhang J, Zhu J, Zhu S, Zhang A, Jia Z, Wang G, Yu S, et al: lncRNA profile of glioblastoma reveals the potential role of lncRNAs in contributing to glioblastoma pathogenesis. Int J Oncol. 40:2004–2012. 2012.PubMed/NCBI

33 

Chen G, Cao Y, Zhang L, Ma H, Shen C and Zhao J: Analysis of long non-coding RNA expression profiles identifies novel lncRNA biomarkers in the tumorigenesis and malignant progression of gliomas. Oncotarget. 8:67744–67753. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Li Q, Jia H, Li H, Dong C, Wang Y and Zou Z: lncRNA and mRNA expression profiles of glioblastoma multiforme (GBM) reveal the potential roles of lncRNAs in GBM pathogenesis. Tumour Biol. 37:14537–14552. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Xi J, Sun Q, Ma L and Kang J: Long non-coding RNAs in glioma progression. Cancer Lett. 419:203–209. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Malissovas N, Ninou E, Michail A and Politis PK: Targeting long non-coding RNAs in nervous system cancers: New insights in prognosis, diagnosis and therapy. Curr Med Chem. 26:5649–5663. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Chen Q, Cai J, Wang Q, Wang Y, Liu M, Yang J, Zhou J, Kang C, Li M and Jiang C: Long noncoding RNA NEAT1, regulated by the EGFR pathway, contributes to glioblastoma progression through the WNT/β-catenin pathway by scaffolding EZH2. Clin Cancer Res. 24:684–695. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Quagliata L, Matter MS, Piscuoglio S, Arabi L, Ruiz C, Procino A, Kovac M, Moretti F, Makowska Z, Boldanova T, et al: Long noncoding RNA HOTTIP/HOXA13 expression is associated with disease progression and predicts outcome in hepatocellular carcinoma patients. Hepatology. 59:911–923. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Wu P, Cai J, Chen Q, Han B, Meng X, Li Y, Li Z, Wang R, Lin L, Duan C, et al: lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p. Nat Commun. 10:20452019. View Article : Google Scholar : PubMed/NCBI

40 

Ghafouri-Fard S and Taheri M: Nuclear enriched abundant transcript 1 (NEAT1): A long non-coding RNA with diverse functions in tumorigenesis. Biomed Pharmacother. 111:51–59. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Zhao W, Li W, Jin X, Niu T, Cao Y, Zhou P and Zheng M: Silencing long non-coding RNA NEAT1 enhances the suppression of cell growth, invasion, and apoptosis of bladder cancer cells under cisplatin chemotherapy. Int J Clin Exp Pathol. 12:549–558. 2019.PubMed/NCBI

42 

Wu Y, Yang L, Zhao J, Li C, Nie J, Liu F, Zhuo C, Zheng Y, Li B, Wang Z and Xu Y: Nuclear-enriched abundant transcript 1 as a diagnostic and prognostic biomarker in colorectal cancer. Mol Cancer. 14:1912015. View Article : Google Scholar : PubMed/NCBI

43 

Fujimoto A, Furuta M, Totoki Y, Tsunoda T, Kato M, Shiraishi Y, Tanaka H, Taniguchi H, Kawakami Y, Ueno M, et al: Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat Genet. 48:500–509. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Jiang X, Zhou Y, Sun AJ and Xue JL: NEAT1 contributes to breast cancer progression through modulating miR-448 and ZEB1. J Cell Physiol. 233:8558–8566. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Jho EH, Zhang T, Domon C, Joo CK, Freund JN and Costantini F: Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol. 22:1172–1183. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Tago K, Nakamura T, Nishita M, Hyodo J, Nagai S, Murata Y, Adachi S, Ohwada S, Morishita Y, Shibuya H and Akiyama T: Inhibition of Wnt signaling by ICAT, a novel beta-catenin-interacting protein. Genes Dev. 14:1741–1749. 2000.PubMed/NCBI

47 

Pandey GK, Mitra S, Subhash S, Hertwig F, Kanduri M, Mishra K, Fransson S, Ganeshram A, Mondal T, Bandaru S, et al: The risk-associated long noncoding RNA NBAT-1 controls neuroblastoma progression by regulating cell proliferation and neuronal differentiation. Cancer Cell. 26:722–737. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Zhang F, Ruan X, Ma J, Liu X, Zheng J, Liu Y, Liu L, Shen S, Shao L, Wang D, et al: DGCR8/ZFAT-AS1 promotes CDX2 transcription in a PRC2 complex-dependent manner to facilitate the malignant biological behavior of glioma cells. Mol Ther. 28:613–630. 2020. View Article : Google Scholar : PubMed/NCBI

49 

de Klerk E and t Hoen PA: Alternative mRNA transcription, processing, and translation: Insights from RNA sequencing. Trends Genet. 31:128–139. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Ong CT and Corces VG: Enhancer function: New insights into the regulation of tissue-specific gene expression. Nat Rev Genet. 12:283–293. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Field A and Adelman K: Evaluating enhancer function and transcription. Annu Rev Biochem. 89:213–234. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Kim TK, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, Harmin DA, Laptewicz M, Barbara-Haley K, Kuersten S, et al: Widespread transcription at neuronal activity-regulated enhancers. Nature. 465:182–187. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Mousavi K, Zare H, Dell'orso S, Grontved L, Gutierrez-Cruz G, Derfoul A, Hager GL and Sartorelli V: eRNAs promote transcription by establishing chromatin accessibility at defined genomic loci. Mol Cell. 51:606–617. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Li W, Notani D, Ma Q, Tanasa B, Nunez E, Chen AY, Merkurjev D, Zhang J, Ohgi K, Song X, et al: Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature. 498:516–520. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Ørom UA, Derrien T, Guigo R and Shiekhattar R: Long noncoding RNAs as enhancers of gene expression. Cold Spring Harb Symp Quant Biol. 75:325–331. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Dykes IM and Emanueli C: Transcriptional and post-transcriptional gene regulation by long non-coding RNA. Genomics Proteomics Bioinformatics. 15:177–186. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Long Y, Wang X, Youmans DT and Cech TR: How do lncRNAs regulate transcription? Sci Adv. 3:eaao21102017. View Article : Google Scholar : PubMed/NCBI

58 

Zovoilis A, Cifuentes-Rojas C, Chu HP, Hernandez AJ and Lee JT: Destabilization of B2 RNA by EZH2 activates the stress response. Cell. 167:1788–1802.e13. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Hernandez AJ, Zovoilis A, Cifuentes-Rojas C, Han L, Bujisic B and Lee JT: B2 and ALU retrotransposons are self-cleaving ribozymes whose activity is enhanced by EZH2. Proc Natl Acad Sci USA. 117:415–425. 2020. View Article : Google Scholar : PubMed/NCBI

60 

Espinoza CA, Allen TA, Hieb AR, Kugel JF and Goodrich JA: B2 RNA binds directly to RNA polymerase II to repress transcript synthesis. Nat Struct Mol Biol. 11:822–829. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Lambert SA, Jolma A, Campitelli LF, Das PK, Yin Y, Albu M, Chen X, Taipale J, Hughes TR and Weirauch MT: The human transcription factors. Cell. 172:650–665. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Hudson WH and Ortlund EA: The structure, function and evolution of proteins that bind DNA and RNA. Nat Rev Mol Cell Biol. 15:749–760. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD, Horlings HM, Shah N, Umbricht C, Wang P, et al: Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet. 43:621–629. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Xu H, Zhao G, Zhang Y, Jiang H, Wang W, Zhao D, Yu H and Qi L: Long non-coding RNA PAXIP1-AS1 facilitates cell invasion and angiogenesis of glioma by recruiting transcription factor ETS1 to upregulate KIF14 expression. J Exp Clin Cancer Res. 38:4862019. View Article : Google Scholar : PubMed/NCBI

65 

Schneider C, King RM and Philipson L: Genes specifically expressed at growth arrest of mammalian cells. Cell. 54:787–793. 1988. View Article : Google Scholar : PubMed/NCBI

66 

Yuan J, Zhang N, Zheng Y, Chen YD, Liu J and Yang M: lncRNA GAS5 indel genetic polymorphism contributes to glioma risk through interfering binding of transcriptional factor TFAP2A. DNA Cell Biol. 37:750–757. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Qi X, Zhang DH, Wu N, Xiao JH, Wang X and Ma W: ceRNA in cancer: Possible functions and clinical implications. J Med Genet. 52:710–718. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Amirkhah R, Naderi-Meshkin H, Shah JS, Dunne PD and Schmitz U: The intricate interplay between epigenetic events, alternative splicing and noncoding RNA deregulation in colorectal cancer. Cells. 8:9292019. View Article : Google Scholar

69 

Zhang XZ, Liu H and Chen SR: Mechanisms of long non-coding RNAs in cancers and their dynamic regulations. Cancers (Basel). 12:12452020. View Article : Google Scholar

70 

Li Y, Yin Z, Fan J, Zhang S and Yang W: The roles of exosomal miRNAs and lncRNAs in lung diseases. Signal Transduct Target Ther. 4:472019. View Article : Google Scholar : PubMed/NCBI

71 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The Rosetta Stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A and Bozzoni I: A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 147:358–369. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Sa L, Li Y, Zhao L, Liu Y, Wang P, Liu L, Li Z, Ma J, Cai H and Xue Y: The Role of HOTAIR/miR-148b-3p/USF1 on regulating the permeability of BTB. Front Mol Neurosci. 10:1942017. View Article : Google Scholar : PubMed/NCBI

74 

Qin N, Tong GF, Sun LW and Xu XL: Long noncoding RNA MEG3 suppresses glioma cell proliferation, migration, and invasion by acting as a competing endogenous RNA of miR-19a. Oncol Res. 25:1471–1478. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Li DX, Fei XR, Dong YF, Cheng CD, Yang Y, Deng XF, Huang HL, Niu WX, Zhou CX, Xia CY and Niu CS: The long non-coding RNA CRNDE acts as a ceRNA and promotes glioma malignancy by preventing miR-136-5p-mediated downregulation of Bcl-2 and Wnt2. Oncotarget. 8:88163–88178. 2017. View Article : Google Scholar : PubMed/NCBI

76 

Zhang X, Sun S, Pu JK, Tsang AC, Lee D, Man VO, Lui WM, Wong ST and Leung GK: Long non-coding RNA expression profiles predict clinical phenotypes in glioma. Neurobiol Dis. 48:1–8. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Zheng J, Li XD, Wang P, Liu XB, Xue YX, Hu Y, Li Z, Li ZQ, Wang ZH and Liu YH: CRNDE affects the malignant biological characteristics of human glioma stem cells by negatively regulating miR-186. Oncotarget. 6:25339–25355. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Zheng J, Liu X, Wang P, Xue Y, Ma J, Qu C and Liu Y: CRNDE promotes malignant progression of glioma by attenuating miR-384/PIWIL4/STAT3 axis. Mol Ther. 24:1199–1215. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Brockdorff N, Bowness JS and Wei G: Progress toward understanding chromosome silencing by Xist RNA. Genes Dev. 34:733–744. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Cheng Z, Luo C and Guo Z: lncRNA-XIST/microRNA-126 sponge mediates cell proliferation and glucose metabolism through the IRS1/PI3K/Akt pathway in glioma. J Cell Biochem. 121:2170–2183. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Ule J and Blencowe BJ: Alternative splicing regulatory networks: Functions, mechanisms, and evolution. Mol Cell. 76:329–345. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Shepard PJ and Hertel KJ: The SR protein family. Genome Biol. 10:2422009. View Article : Google Scholar : PubMed/NCBI

83 

Blencowe BJ: Alternative splicing: New insights from global analyses. Cell. 126:37–47. 2006. View Article : Google Scholar : PubMed/NCBI

84 

Sun Y and Ma L: New insights into long non-coding RNA MALAT1 in cancer and metastasis. Cancers (Basel). 11:2162019. View Article : Google Scholar

85 

Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, et al: MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 22:8031–8041. 2003. View Article : Google Scholar : PubMed/NCBI

86 

Arun G and Spector DL: MALAT1 long non-coding RNA and breast cancer. RNA Biol. 16:860–863. 2019. View Article : Google Scholar : PubMed/NCBI

87 

Chang J, Xu W, Du X and Hou J: MALAT1 silencing suppresses prostate cancer progression by upregulating miR-1 and downregulating KRAS. Onco Targets Ther. 11:3461–3473. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Le L, Chen H, Gao Y, Wang YW, Zhang GQ, Pan SH, Ji L, Kong R, Wang G, Jia YH, et al: Long noncoding RNA MALAT1 promotes aggressive pancreatic cancer proliferation and metastasis via the stimulation of autophagy. Mol Cancer Ther. 15:2232–2243. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Liao K, Lin Y, Gao W, Xiao Z, Medina R, Dmitriev P, Cui J, Zhuang Z, Zhao X, Qiu Y, et al: Blocking lncRNA MALAT1/miR-199a/ZHX1 axis inhibits glioblastoma proliferation and progression. Mol Ther Nucleic Acids. 18:388–399. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Wen F, Cao YX, Luo ZY, Liao P and Lu ZW: lncRNA MALAT1 promotes cell proliferation and imatinib resistance by sponging miR-328 in chronic myelogenous leukemia. Biochem Biophys Res Commun. 507:1–8. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, et al: The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell. 39:925–938. 2010. View Article : Google Scholar : PubMed/NCBI

92 

Long JC and Caceres JF: The SR protein family of splicing factors: Master regulators of gene expression. Biochem J. 417:15–27. 2009. View Article : Google Scholar : PubMed/NCBI

93 

Stamm S: Regulation of alternative splicing by reversible protein phosphorylation. J Biol Chem. 283:1223–1227. 2008. View Article : Google Scholar : PubMed/NCBI

94 

Huang B, Song JH, Cheng Y, Abraham JM, Ibrahim S, Sun Z, Ke X and Meltzer SJ: Long non-coding antisense RNA KRT7-AS is activated in gastric cancers and supports cancer cell progression by increasing KRT7 expression. Oncogene. 35:4927–4936. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Zhang L, Yang Z, Trottier J, Barbier O and Wang L: Long noncoding RNA MEG3 induces cholestatic liver injury by interaction with PTBP1 to facilitate shp mRNA decay. Hepatology. 65:604–615. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Zhu L, Wei Q, Qi Y, Ruan X, Wu F, Li L, Zhou J, Liu W, Jiang T, Zhang J, et al: PTB-AS, a novel natural antisense transcript, promotes glioma progression by improving PTBP1 mRNA stability with SND1. Mol Ther. 27:1621–1637. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Cao S, Zheng J, Liu X, Liu Y, Ruan X, Ma J, Liu L, Wang D, Yang C, Cai H, et al: FXR1 promotes the malignant biological behavior of glioma cells via stabilizing MIR17HG. J Exp Clin Cancer Res. 38:372019. View Article : Google Scholar : PubMed/NCBI

98 

Wang Y, Wang Y, Li J, Zhang Y, Yin H and Han B: CRNDE, a long-noncoding RNA, promotes glioma cell growth and invasion through mTOR signaling. Cancer Lett. 367:122–128. 2015. View Article : Google Scholar : PubMed/NCBI

99 

Barile L and Vassalli G: Exosomes: Therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 174:63–78. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Jiang L, Gu Y, Du Y and Liu J: Exosomes: Diagnostic biomarkers and therapeutic delivery vehicles for cancer. Mol Pharm. 16:3333–3349. 2019. View Article : Google Scholar : PubMed/NCBI

101 

D'Asti E, Chennakrishnaiah S, Lee TH and Rak J: Extracellular vesicles in brain tumor progression. Cell Mol Neurobiol. 36:383–407. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Katsuda T, Kosaka N and Ochiya T: The roles of extracellular vesicles in cancer biology: Toward the development of novel cancer biomarkers. Proteomics. 14:412–425. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Vader P, Breakefield XO and Wood MJ: Extracellular vesicles: Emerging targets for cancer therapy. Trends Mol Med. 20:385–393. 2014. View Article : Google Scholar : PubMed/NCBI

104 

Jabbari N, Akbariazar E, Feqhhi M, Rahbarghazi R and Rezaie J: Breast cancer-derived exosomes: Tumor progression and therapeutic agents. J Cell Physiol. 235:6345–6356. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Lorenc T, Klimczyk K, Michalczewska I, Słomka M, Kubiak-Tomaszewska G and Olejarz W: Exosomes in prostate cancer diagnosis, prognosis and therapy. Int J Mol Sci. 21:21182020. View Article : Google Scholar

106 

Sun W, Ren Y, Lu Z and Zhao X: The potential roles of exosomes in pancreatic cancer initiation and metastasis. Mol Cancer. 19:1352020. View Article : Google Scholar : PubMed/NCBI

107 

Zhu L, Li J, Gong Y, Wu Q, Tan S, Sun D, Xu X, Zuo Y, Zhao Y, Wei YQ, et al: Exosomal tRNA-derived small RNA as a promising biomarker for cancer diagnosis. Mol Cancer. 18:742019. View Article : Google Scholar : PubMed/NCBI

108 

Shah S, Wittmann S, Kilchert C and Vasiljeva L: lncRNA recruits RNAi and the exosome to dynamically regulate pho1 expression in response to phosphate levels in fission yeast. Genes Dev. 28:231–244. 2014. View Article : Google Scholar : PubMed/NCBI

109 

Han M, Gu Y, Lu P, Li J, Cao H, Li X, Qian X, Yu C, Yang Y, Yang X, et al: Exosome-mediated lncRNA AFAP1-AS1 promotes trastuzumab resistance through binding with AUF1 and activating ERBB2 translation. Mol Cancer. 19:262020. View Article : Google Scholar : PubMed/NCBI

110 

Zhang Z, Yin J, Lu C, Wei Y, Zeng A and You Y: Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J Exp Clin Cancer Res. 38:1662019. View Article : Google Scholar : PubMed/NCBI

111 

Yuan Z, Yang Z, Li W, Wu A, Su Z and Jiang B: Exosome-mediated transfer of long noncoding RNA HOTAIR regulates temozolomide resistance by miR-519a-3p/RRM1 axis in glioblastoma. Cancer Biother Radiopharm. Jul 24–2020.(Epub ahead of print). doi: 10.1089/cbr.2019.3499. View Article : Google Scholar

112 

Li LJ, Leng RX, Fan YG, Pan HF and Ye DQ: Translation of noncoding RNAs: Focus on lncRNAs, pri-miRNAs, and circRNAs. Exp Cell Res. 361:1–8. 2017. View Article : Google Scholar : PubMed/NCBI

113 

Wu P, Mo Y, Peng M, Tang T, Zhong Y, Deng X, Xiong F, Guo C, Wu X, Li Y, et al: Emerging role of tumor-related functional peptides encoded by lncRNA and circRNA. Mol Cancer. 19:222020. View Article : Google Scholar : PubMed/NCBI

114 

Wang Y, Wu S, Zhu X, Zhang L, Deng J, Li F, Guo B, Zhang S, Wu R, Zhang Z, et al: lncRNA-encoded polypeptide ASRPS inhibits triple-negative breast cancer angiogenesis. J Exp Med. 217:jem.20190950. 2020. View Article : Google Scholar

115 

Begum S, Yiu A, Stebbing J and Castellano L: Novel tumour suppressive protein encoded by circular RNA, circ-SHPRH, in glioblastomas. Oncogene. 37:4055–4057. 2018. View Article : Google Scholar : PubMed/NCBI

116 

Zhang M, Huang N, Yang X, Luo J, Yan S, Xiao F, Chen W, Gao X, Zhao K, Zhou H, et al: A novel protein encoded by the circular form of the SHPRH gene suppresses glioma tumorigenesis. Oncogene. 37:1805–1814. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Zhang M, Zhao K, Xu X, Yang Y, Yan S, Wei P, Liu H, Xu J, Xiao F, Zhou H, et al: A peptide encoded by circular form of LINC-PINT suppresses oncogenic transcriptional elongation in glioblastoma. Nat Commun. 9:44752018. View Article : Google Scholar : PubMed/NCBI

118 

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

119 

Özeş AR, Wang Y, Zong X, Fang F, Pilrose J and Nephew KP: Therapeutic targeting using tumor specific peptides inhibits long non-coding RNA HOTAIR activity in ovarian and breast cancer. Sci Rep. 7:8942017. View Article : Google Scholar : PubMed/NCBI

120 

Bullock MD, Silva AM, Kanlikilicer-Unaldi P, Filant J, Rashed MH, Sood AK, Lopez-Berestein G and Calin GA: Exosomal non-coding RNAs: Diagnostic, prognostic and therapeutic applications in cancer. Noncoding RNA. 1:53–68. 2015.PubMed/NCBI

121 

H Rashed M, Bayraktar E, K Helal G, Abd-Ellah MF, Amero P, Chavez-Reyes A and Rodriguez-Aguayo C: Exosomes: From garbage bins to promising therapeutic targets. Int J Mol Sci. 18:5382017. View Article : Google Scholar

Related Articles

Journal Cover

April-2021
Volume 45 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Guo G, Lu Y, Wang S, Zhang Y and Huang Q: Mechanisms and functions of long non‑coding RNAs in glioma (Review). Oncol Rep 45: 9, 2021
APA
Chen, X., Guo, G., Lu, Y., Wang, S., Zhang, Y., & Huang, Q. (2021). Mechanisms and functions of long non‑coding RNAs in glioma (Review). Oncology Reports, 45, 9. https://doi.org/10.3892/or.2021.7960
MLA
Chen, X., Guo, G., Lu, Y., Wang, S., Zhang, Y., Huang, Q."Mechanisms and functions of long non‑coding RNAs in glioma (Review)". Oncology Reports 45.4 (2021): 9.
Chicago
Chen, X., Guo, G., Lu, Y., Wang, S., Zhang, Y., Huang, Q."Mechanisms and functions of long non‑coding RNAs in glioma (Review)". Oncology Reports 45, no. 4 (2021): 9. https://doi.org/10.3892/or.2021.7960