Elsevier

Neurochemistry International

Volume 77, November 2014, Pages 78-85
Neurochemistry International

Review
Long non-coding RNAs in glioma: Functional roles and clinical perspectives

https://doi.org/10.1016/j.neuint.2014.05.008Get rights and content

Highlights

  • We discuss the aberrant lncRNA expression in glioma initiation and progression.

  • We discuss the potential mechanisms underlying lncRNA dysregulation in glioma.

  • We discuss the functional roles of lncRNAs in regulating glioma biological behavior.

  • We discuss the potentials of lncRNAs as biomarkers and therapy targets in glioma.

Abstract

Long non-coding RNAs (lncRNAs) are a new class of non-coding gene regulators. But unlike their smaller counterparts, microRNAs, relatively less is known about the roles and functions of lncRNAs. Current evidence suggests that lncRNAs may play important roles in a wide range of biological processes in human cancers, including glioma. By acting as oncogenes or tumor suppressors, lncRNAs may contribute to glioma initiation, progression and other malignant phenotypes. Their expression profiles may also have important clinical implications in glioma subclassification and patients’ prognostication. Here, we review current evidence related to the functional roles of lncRNAs in glioma. We will discuss the aberrant lncRNA expression signatures associated with glioma initiation and progression, as well as the potential mechanisms underlying lncRNA dysregulation. We also discuss the functional roles of lncRNAs in glioma biological behavior. Finally, the potentials and prospects of employing lncRNAs as novel biomarkers and therapeutic targets for glioma clinical practice will also be addressed.

Introduction

The emerging significance of long non-coding RNAs (lncRNAs) in cancer has attracted considerable interests in recent years (Niland et al., 2012, Prensner and Chinnaiyan, 2011). While initially thought to be spurious transcriptional noise, increasing evidence began to suggest that lncRNAs indeed play critical regulatory roles in the development of many human diseases, including cancer (Gibb et al., 2011, Wapinski and Chang, 2011). Functional studies for a handful of lncRNAs revealed that the latter could regulate gene expressions at transcriptional, post-transcriptional and epigenetic levels (Mercer et al., 2009). Aberrant expressions of lncRNAs may potentially alter basic cellular biological processes and contribute to tumorigenesis (Gibb et al., 2011, Gutschner and Diederichs, 2012). Moreover, differential expressions of specific lncRNAs may correlate with disease processes, staging or other malignant phenotypes, and thus could potentially act as therapeutic targets, as well as biomarkers for diagnosis and prognosis (Qi and Du, 2013, Spizzo et al., 2012, Wahlestedt, 2013). LncRNAs are emerging as new players in cancer biology paradigm.

Gliomas account for the great majority of primary tumors in the brain (Wen and Kesari, 2008). Based on their likely cellular origins, gliomas may broadly be subclassified into astrocytomas, oligodendrogliomas, ependymomas and mixed tumors (e.g. oligoastrocytomas) (Louis et al., 2007). Within each histological subtype, they could be further categorized into grades I–IV lesions based on the degree of malignancy (Louis et al., 2007). The clinical outcomes of glioma patients depend heavily on histopathological features (Wen and Kesari, 2008). The most malignant tumor type, glioblastoma multiform (GBM, grade IV astrocytoma), is almost invariably fatal with an overall survival of just over one year only (Wen and Kesari, 2008). Despite the development of multimodal and aggressive treatments that include surgical resection, local radiotherapy and systemic chemotherapy in the past decades, patient outcomes remain unsatisfactory (Omuro and DeAngelis, 2013, Taylor, 2010). To improve treatment efficacy, a better understanding of glioma pathogenesis at the genetic and molecular levels is urgently needed (Chen et al., 2012, Jansen et al., 2010).

Recent evidence indicates that lncRNAs play important roles in glioma pathogenesis (Bian et al., 2014, Sun et al., 2013). It has been reported that lncRNAs may regulate certain tumorigenic processes in glioma such as cellular proliferation and apoptosis (Wang et al., 2012). Aberrant expressions of lncRNAs may not only reflect clinical phenotypes (Zhang et al., 2012) and patient prognosis (Zhang et al., 2013), but also can be exploited as potential therapeutic targets (Amit et al., 2012). In this review, we summarize the recent progress of lncRNA studies in glioma. We will first provide an overview on the dysregulated expressions of lncRNAs associated with glioma initiation and progression, and the potential underlying mechanisms. We will then discuss the functional roles of lncRNAs in regulating glioma biological behavior. Finally, we will discuss the potentials of lncRNAs as biomarkers for glioma diagnosis, prognostication and targeted therapy.

Section snippets

Dysregulation of lncRNA expression in glioma

Genome-wide profiling studies have demonstrated that lncRNAs are aberrantly expressed in gliomas as compared with non-tumoral brain tissues. Differential expressions also occur across gliomas with different malignancy grades, suggesting the potential role of lncRNAs in glioma initiation as well as progression. The lncRNA profiling studies in glioma reported so far are summarized in Table 1.

For each study reviewed here, and also in the following sections of this manuscript, the significantly

Potential mechanisms underlying lncRNA dysregulation in glioma

The mechanisms underlying the dysregulation of lncRNAs remain incompletely understood. Generally, there are three mechanisms commonly reported to underlie the regulation of gene expressions in human cancer cells: transcriptional regulation, epigenetic regulation and genetic regulation. These mechanisms can function independently or in concert. Abundant evidence is now available to demonstrate the regulatory effects of these mechanisms in mRNA and miRNA expressions, and it is likely that they

Functional roles of lncRNAs in glioma

LncRNAs have been implicated in the biological behavior of glioma cells. However, as a newly emerging gene class, there is currently a little direct evidence on the functional roles of lncRNA in glioma. As mentioned above and in our previous study (Zhang et al., 2012), the tentative link between lncRNA and the specific biological behavior of glioma indeed may be inferred indirectly from results obtained by mining previously published data in public databases. Although the underlying molecular

LncRNAs in relation to glioma treatment response

Aberrant lncRNAs expression has also been linked with differential treatment responses in glioma patients. Garcia-Claver et al. (2013) analyzed the gene expression profiles of a panel of human glioma cell lines before and after erlotinib (ERL) treatment, a tyrosine kinase inhibitor that acts on the epidermal growth factor receptor (EGFR). They found that lncRNA GAS5 was significantly increased after ERL treatment in both ERL-resistant and ERL-sensitive glioma cell lines. Moreover, knock-down of

LncRNAs as diagnostic and prognostic biomarkers in glioma

The differential expressions of lncRNAs between tumors and normal tissues, or in tumors with different clinical features provide a rationale for their use in diagnostics and prognostication.

By comparative analysis of the lncRNA expression signatures between the gliomas with different histological subtypes (astrocytic and oligodendroglial), we have reported differential expressions of 23 lncRNAs (Zhang et al., 2012). By using this set of lncRNAs, astrocytic tumors could be distinguished from

LncRNAs as therapeutic targets in glioma

The fact that lncRNA dysregulation in cancer has a pathogenic effect provides the theoretical rationale for using lncRNAs as potential therapeutic targets. At present, the therapeutic potential of lncRNAs has been reported in several cancer types, including glioma. Amit et al. (2012) tested the therapeutic effect of H19 on GBM in vitro and in vivo. They first constructed a DNA plasmid that expressed diphtheria toxin A-fragment (DTA, an intracellular toxin) under the control of both H19 and

Conclusion

The discovery of lncRNAs has generated a lot of excitement in the field of cancer research in the past several years. Reported studies have demonstrated the associations between lncRNA expressions and glioma malignant phenotypes, and this may potentially lead to the development of novel diagnostic and prognostic biomarkers. However, more studies in large cohorts at independent laboratories are needed before the utilization of lncRNA-based biomarkers can be further exploited. In addition, given

Acknowledgement

We thank Miss. Karrie Kiang and Mr. Derek Lee for their critical reading of, and valuable input into, the manuscript.

Reference (76)

  • Y.Z. Sun et al.

    Long non-coding RNAs as potential biomarkers and therapeutic targets for gliomas

    Med. Hypotheses

    (2013)
  • H. Suzuki et al.

    DNA methylation and microRNA dysregulation in cancer

    Mol. Oncol.

    (2012)
  • K. Tano et al.

    MALAT-1 enhances cell motility of lung adenocarcinoma cells by influencing the expression of motility-related genes

    FEBS Lett.

    (2010)
  • O. Wapinski et al.

    Long noncoding RNAs and human disease

    Trends Cell Biol.

    (2011)
  • X. Zhang et al.

    Long non-coding RNA expression profiles predict clinical phenotypes in glioma

    Neurobiol. Dis.

    (2012)
  • X.Q. Zhang et al.

    A long non-coding RNA signature in glioblastoma multiforme predicts survival

    Neurobiol. Dis.

    (2013)
  • Aldaz, B., Malumbres, R., Luna, J., Martinez-Climent, J., Guruceaga, E., Segura, V., 2013. Microarray analysis of...
  • V. Amberger-Murphy

    Hypoxia helps glioma to fight therapy

    Curr. Cancer Drug Targets

    (2009)
  • D. Amit et al.

    Transcriptional targeting of glioblastoma by diphtheria toxin-A driven by both H19 and IGF2-P4 promoters

    Int. J. Clin. Exp. Med.

    (2012)
  • G. Anders et al.

    DoRiNA: a database of RNA interactions in post-transcriptional regulation

    Nucleic Acids Res.

    (2012)
  • Araki, N., Niibori, A., Midorikawa, U., 2013. Expression profiling of glioma initiating cells (GICs) in the sphere and...
  • T. Arita et al.

    Circulating long non-coding RNAs in plasma of patients with gastric cancer

    Anticancer Res.

    (2013)
  • G.P. Atkinson et al.

    NF-kappaB and STAT3 signaling in glioma: targets for future therapies

    Expert Rev. Neurother.

    (2010)
  • D. Barsyte-Lovejoy et al.

    The c-Myc oncogene directly induces the H19 noncoding RNA by allele-specific binding to potentiate tumorigenesis

    Cancer Res.

    (2006)
  • M. Bellucci et al.

    Predicting protein associations with long noncoding RNAs

    Nat. Methods

    (2011)
  • E.B. Bian et al.

    LncRNAs: new players in gliomas, with special emphasis on the interaction of lncRNAs With EZH2

    J. Cell. Physiol.

    (2014)
  • G.A. Calin et al.

    Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers

    Proc. Natl. Acad. Sci. USA

    (2004)
  • M.E. Davis et al.

    Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles

    Nature

    (2010)
  • B.C. Ellis et al.

    CRNDE: a long non-coding RNA involved in CanceR, Neurobiology, and DEvelopment

    Front. Genet.

    (2012)
  • H.E. Gee et al.

    Hsa-mir-210 is a marker of tumor hypoxia and a prognostic factor in head and neck cancer

    Cancer

    (2010)
  • A. Giannakakis et al.

    MiR-210 links hypoxia with cell cycle regulation and is deleted in human epithelial ovarian cancer

    Cancer Biol. Ther.

    (2008)
  • E.A. Gibb et al.

    The functional role of long non-coding RNA in human carcinomas

    Mol. Cancer

    (2011)
  • L.D. Graham et al.

    Colorectal neoplasia differentially expressed (CRNDE), a novel gene with elevated expression in colorectal adenomas and adenocarcinomas

    Genes Cancer

    (2011)
  • L.A.M. Gravendeel et al.

    Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology

    Cancer Res.

    (2009)
  • M. Grzmil et al.

    MAP kinase-interacting kinase 1 regulates SMAD2-dependent TGF-beta signaling pathway in human glioblastoma

    Cancer Res.

    (2011)
  • T. Gutschner et al.

    The hallmarks of cancer: a long non-coding RNA point of view

    RNA Biol.

    (2012)
  • T. Gutschner et al.

    The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells

    Cancer Res.

    (2013)
  • L. Han et al.

    LncRNA profile of glioblastoma reveals the potential role of lncRNAs in contributing to glioblastoma pathogenesis

    Int. J. Oncol.

    (2012)
  • Cited by (72)

    • Long Non-coding RNA EPIC1 Promotes Cell Proliferation and Motility and Drug Resistance in Glioma

      2020, Molecular Therapy Oncolytics
      Citation Excerpt :

      lncRNAs play an oncogenic or tumor-suppressive role in glioma initiation and progression.13 Aberrant expression signatures of lncRNAs have been revealed to be correlated with glioma development and malignant progression.13 For example, linc00645 enhanced transforming growth factor beta (TGF-β)-triggered epithelial mesenchymal transition (EMT) through regulation of microRNA-205-3p (miR-205-3p) and zinc finger E-box binding homeobox 1 (ZEB1) in glioma.14

    • Long non-coding SNHG1 in cancer

      2019, Clinica Chimica Acta
    View all citing articles on Scopus
    View full text