Open Access

Detecting the long non‑coding RNA signature related to spinal cord ependymal tumor subtype using a genome‑wide methylome analysis approach

  • Authors:
    • Li Wang
    • Chi Zhang
    • Yujie Xie
    • Wei Jiang
    • Juan Huang
    • Shengmin Guo
    • Fangyuan Xu
    • Jianxiong Wang
  • View Affiliations

  • Published online on: June 18, 2019     https://doi.org/10.3892/mmr.2019.10388
  • Pages: 1531-1540
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Ependymoma is a type of intramedullary tumor that tends to occur in the adult spinal cord. Ependymoma affects the nervous system and has significant impacts on the quality of life, and it may lead to mortality. Previous studies have performed molecular classification of spinal cord ependymal tumors at the DNA methylation level. However, the DNA methylation status of non‑coding regions in spinal cord ependymal tumors remains unclear. In the present study, a genome‑wide methylome method was used to characterize the DNA methylation landscape of long non‑coding RNAs (lncRNAs) in spinal cord ependymal tumor samples. The present study identified lncRNA signatures associated with tumor subtypes based on the methylation status of lncRNA promoters. The present results suggested that the identified lncRNA signatures were associated with cancer‑ or nervous system‑related protein‑coding genes. The majority of the identified lncRNAs was hypomethylated, and may have a role in spinal cord development. The present findings suggested that detection of tumor subtype‑specific lncRNAs may facilitate the identification of novel diagnostic and therapeutic strategies to treat patients with spinal cord ependymal tumor.

Introduction

Spinal tumors are rare and, based on their location, can be classified into various subtypes, such as intramedullary and extramedullary tumors (1). Ependymoma is a type of intramedullary tumor that tends to occur in the spinal cord in adults (2). Ependymoma is the most common type of spinal cord tumor and has significant impacts on the quality of life of patients, and may cause mortality (3). Classifying ependymal tumors has represented a challenge, as the current grading systems do not accurately describe their clinical characteristics (4,5). However, previous studies have found that DNA methylation patterns are reliable biomarkers for the classification of different molecular subtypes of spinal ependymal tumors (6).

Improved detection methods and the identification of disease-associated biomarkers have been shown to facilitate the diagnosis and treatment of patients (711). DNA methylation is one of the most studied epigenetic modifications in mammals (12,13). Notably, aberrant methylation is associated with cancer and aging (14,15). Some DNA methylation markers have been used in commercially available clinical tests, and most of the sites of methylation are located in the promoters of genes (16). Despite several previous studies suggesting that DNA methylation is a reliable molecular biomarker in ependymal tumors, to the best of our knowledge, the role of methylation in long non-coding RNA (lncRNA) genes has not been investigated (4,6). lncRNAs are >200 nucleotides in length and do not encode proteins. Similarly to mRNAs, lncRNAs have their own promoters (17), are transcribed by RNA polymerase II and have a polyadenylated tail (18). An increasing number of studies have identified various roles of lncRNAs in multiple biological processes and diseases through various mechanisms (19,20). Previous studies have demonstrated that lncRNAs can serve as biomarkers for various cancer features (21). Recent studies have provided insight on the mechanism underlying DNA methylation of lncRNA genes in carcinogenesis (22,23). However, to the best of our knowledge, a systematic investigation of the DNA methylation status and the function of lncRNA genes in spinal cord ependymal tumor has yet to be reported.

In the present study, the DNA methylation landscape of lncRNAs in spinal cord ependymal tumors was investigated and certain lncRNAs were identified to have distinct DNA methylation states among various tumor subtypes. Additionally, various tumor subtype-specific lncRNAs were identified to be involved in spinal cord development. In the present study, a functional characterization of lncRNAs was performed using lncRNA-protein interaction data, and a random forest algorithm was used to identify 30 lncRNAs with high classification efficiency. Notably, the majority of subtype-specific lncRNAs was identified to be hypomethylated. A subset of the identified lncRNAs was found to have potential roles in cancer and nervous system development. Furthermore, a functional analysis identified the role of subtype-specific lncRNAs in spinal cord ependymal tumors. To the best of our knowledge, the present study is the first to investigate the DNA methylation status of lncRNA genes in order to identify their clinical significance in various molecular subtypes of spinal cord ependymal tumors.

Materials and methods

Methylation and lncRNA data collection

Whole-genome DNA methylation data from spinal cord ependymal tumors were downloaded from Gene Expression Omnibus version 1 (24) [accession no. GSE65362 (4)]. Specifically, these samples were classified into three classes: i) Spinal ependymoma (SP-EPN); ii) spinal myxopapillary EPN (SP-MPE); and iii) spinal subependymoma (SP-SE). lncRNA annotation data were downloaded from GENCODE (version 28) (25). These lncRNAs were classified into nine groups based on their location and features (http://vega.archive.ensembl.org/info/about/gene_and_transcript_types.html) (Fig. 1A). Notably, the To be Experimentally Confirmed (TEC) category has been specifically created for the ENCODE project to indicate regions that could indicate the presence of protein coding genes (PCGs) that require experimental validation (25). Importantly, Ensembl (https://www.ensembl.org/info/genome/genebuild/biotypes.html) and Vega (http://vega.archive.ensembl.org/info/about/gene_and_transcript_types.html) databases classify TEC genes as lncRNAs.

Mapping methylation probes to lncRNA promoters

The genomic locations of the Infinium Human Methylation 450 (HM450) BeadChip (Illumina, Inc.) probes based on GRCh37 were converted to the genomic locations in GRCh38 (The Genome Reference Consortium; http://www.ncbi.nlm.nih.gov/grc) using the University of California, Santa Cruz (UCSC) Batch Coordinate Conversion liftOver tool version 2 (26). Probes exhibiting single nucleotide polymorphisms (SNPs) >5 bp from their 3′-end and probes with non-unique 3′-subsequences of 30 bases were excluded, as previously described (27). lncRNA promoter regions were defined as 3-kb windows from either side of the transcription start site (TSS), as previously described (22). Subsequently, the methylation probes were mapped to the lncRNA promoter regions using BEDtools version 2.24.0 (28). The same approach was used to map the PCG probes. In addition, methylation probes that simultaneously mapped to lncRNAs and PCGs were excluded.

lncRNA DNA methylation in spinal cord ependymal tumors

lncRNA promoter methylation values were calculated as the mean values of all probes located in the corresponding promoter. DNA methylation patterns around TSSs were calculated in 100 bp windows based on the median methylation value across samples. The lncRNA methylation value was used to cluster samples using the pheatmap package (version 1.0.12; http://cran.r-project.org/web/packages/pheatmap/index.html), using the default parameters, on R (version 3.3.0; http://cran.r-project.org/). The similarity between tumor samples was measured by Pearson correlation coefficients using R to determine if tumors within a subtype were more similar to each other than those from other subtypes. In addition, principal component analysis was used to investigate the methylation patterns of different tumor subgroups. Tumor subtype-specific lncRNAs were identified by ANOVA using R, as previously described (29). The RCircos package (version 1.2.1; http://cran.r-project.org/web/packages/RCircos/index.html) was used with R to display the distribution of methylation levels and the locations of tumor subtype-specific lncRNAs in the genome.

Tumor subtype-specific lncRNA-protein interaction network construction

RNA-protein interaction data were downloaded from the RAID database (version 2.0) (30), which integrates experimental and computational prediction interactions from the literature and other database resources under one common framework. lncRNA-protein interaction associations were calculated by mapping subtype-specific lncRNAs to mRNAs.

Tumor subtype-specific lncRNA signature identification

A random forest algorithm (31) was performed using the DNA methylation value of the lncRNAs identified in the constructed network. The lncRNAs with a feature importance value >0, calculated using the random forest algorithm, were considered potential tumor subtype-specific signatures (32). All statistical analyses were performed using R (version 3.3.0; R project).

Functional enrichment analysis of lncRNAs and PCGs

Tumor subtype-specific lncRNAs were annotated using their promoter regions by the Genomic Regions Enrichment of Annotations Tool (GREAT; version 3.3.0) using the default parameters (33); in addition, the lncRNA promoter regions were used as ‘background regions’. Specifically, the genomic coordinates of the lncRNAs used in GREAT were first converted into GRCh37 coordinates using the UCSC liftover tool, as aforementioned. Kyoto Encyclopedia of Genes and Genomes (KEGG; version 73.0) (34) enrichment analysis was performed on the PCGs using the Enrichr online tool (version 2.0), using the default parameters (35), and the significantly enriched KEGG pathways were identified (P<0.01).

Results

Global DNA methylation patterns in lncRNA promoters in spinal cord ependymal tumors

To characterize lncRNA methylation patterns, a computational pipeline was used to annotate HM450 probes to lncRNA promoters. In total, 485,506 HM450 probes were successfully converted into GRCh38 coordinates, and 433,532 probes were obtained after filtering for SNPs and copy number-associated probes. The present analysis resulted in a set of 29,402 probes annotated in 6,967 lncRNA promoter regions (Fig. 1A). In total, 12,668 and 11,711 methylation probes were found in the promoters of long intergenic noncoding RNAs (lincRNAs) and antisense lncRNAs, respectively. Additionally, lncRNAs exhibited methylation states similar to those of PCGs around TSSs; both had low methylation values in proximity to the TSS and had higher methylation values as the distance from the TSS increased (Fig. 1B). However, the overall methylation level in lncRNAs was higher compared with PCGs (Fig. 1B). Cluster analysis suggested that tumor samples from patients with the same molecular subtype had similar methylation patterns in lncRNAs and that the methylation levels of various lncRNAs in the same class were not similar (Fig. 1C). Additionally, the majority of lncRNAs identified in the present study had a methylation value >0.4 (Fig. 1C). Certain lncRNAs, including those classified as ‘processed_transcript’ and ‘bidirectional_promoter_lncRNA’ lncRNAs, exhibited bimodally distributed methylation levels in tumor samples, whereas other lncRNAs, including ‘antisense’ and ‘sense_intronic’ lncRNAs, were hypermethylated in the majority of the patients (Fig. 1D).

DNA methylation in lncRNAs is correlated with histological characteristics of spinal cord ependymal tumors

The patterns of DNA methylation in the lncRNA genes were investigated in different tumor histopathological subtypes. In total, there were 57 patients with spinal cord tumors, 29 (50.9%) patients were in the SP-MPE group, whereas 21 (36.8%) and 7 (12.3%) were in the SP-EPN and SP-SE group, respectively. DNA methylation correlation values were calculated between each pair of samples, and tumor samples were clustered using their methylation correlation values. lncRNA methylation levels were found to be more similar within groups than between groups (Fig. 2A). In particular, patients in the SP-SE group exhibited the most consistent methylation status, with a mean methylation correlation value of 0.96. The mean methylation correlation values in the SP-EPN and SP-MPE were 0.93 and 0.92, respectively. This effect may be due to the little tumor heterogeneity among SE samples. According to the World Health Organization classification (36,37), SP-SE is considered a grade I tumor and its prognosis is more favorable compared with the majority of ependymal tumors (36). In addition, principal component analysis was performed to analyze the tumor samples based on the DNA methylation of the lncRNAs. The first three principal components had the highest proportion of variance values (Fig. 2B). These three components were used to characterize the DNA methylation features in these tumor subtypes, and the results suggested that patients with the same tumor subtype were likely to have similar lncRNA methylation levels (Fig. 2C). The present results suggested that the DNA methylation in lncRNA genes was correlated with histological characteristics of spinal cord ependymal tumors.

DNA methylation-based detection of tumor subtypes-specific lncRNAs

Tumor subtype-specific lncRNAs were identified based on their DNA methylation values using ANOVA. In total, 1,046 subtype-specific lncRNAs were identified [false discovery rate (FDR) <0.05; Figs. 3 and S1]. These lncRNAs tended to have higher methylation values (Fig. 3A), and the majority had methylation values ~0.9. Furthermore, the patients in the SP-EPN group had the highest methylation levels, whereas the patients in the SP-SE had the lowest methylation levels (Fig. 3B and C). GREAT was used to investigate the potential mechanisms of lncRNAs in spinal cord ependymal tumors. Among the significant Gene Ontology (38) terms (FDR q-value <0.01), three of them were associated with spinal cord development (Fig. 3D). ‘Ventral spinal cord development’ and ‘spinal cord development’ processes can affect cell differentiation of spinal cord cells, and a previous study demonstrated that cell differentiation is associated with tumorigenesis (39). Therefore, the distinct DNA methylation levels of various lncRNAs may affect the expression levels of lncRNAs in each tumor subtype, thus affecting genes associated with the nervous system and spinal cord development. The present results indicated the role of lncRNAs in human spinal cord development and suggested that they could be lncRNA signatures in spinal cord ependymal tumors.

Tumor subtype-specific lncRNA signature discovery

Biological networks represent the interactions between molecules in vivo, and can be used to identify regulatory pathways and processes (40,41). lncRNAs may contribute to cancer by interacting with proteins (42), and an increasing number of studies have investigated lncRNA-protein interactions (43). lncRNA-protein interaction data in humans have been downloaded from the RAID database (30). In total, 32 subtype-specific lncRNAs exhibited associations with proteins (Fig. 4A). The network degrees of lncRNAs and proteins were separately analyzed (Fig. 4B and C). lncRNAs exhibited more associations compared with proteins. The present results indicated the important role of lncRNAs in interacting with proteins and suggested that these 32 lncRNAs may affect the disease status by affecting protein function in spinal cord ependymal tumors. Furthermore, in order to identify the lncRNA signatures in ependymal tumors, a random forest classification method was used. In total, 30 lncRNAs with importance values >0, ranging between 0.48 and 2.98, were identified (Fig. 4D). Using the methylation values of these 30 lncRNAs, the three tumor subtypes could be reliably distinguished with sensitivity and specificity values of 1 (Fig. 4E). These lncRNAs may be potential biomarkers associated with tumor subtypes, and may facilitate the diagnosis and treatment of patients with spinal cord ependymal tumors.

Long intergenic non-coding RNA 52 (LINC00052) is a lincRNA, and exhibited the highest importance value in the classification of the three tumor subtypes (Fig. 4D and F). LINC00052 has been previously shown to promote breast cancer and hepatocarcinoma development (44,45). HOX transcript antisense RNA (HOTAIR) is an oncogenic lncRNA in multiple types of cancer, including breast, gastric, colorectal and cervical cancer (46). Additionally, its DNA methylation status can serve as a biomarker in primary ovarian cancer (47). HOTAIR has been investigated as a prognostic factor in mesenchymal glioma, another type of tumor affecting the nervous system (48). In the network constructed in the present study, HOTAIR was identified to exhibit the features of a hub. HOTAIR interacted with 51 proteins and displayed tumor subtype-specific DNA methylation characteristics (Fig. 4A and G). The lncRNAs listed in Fig. 4D may be potential novel biomarkers for treating and diagnosing spinal cord ependymal tumors. Analyzing the DNA methylation values of these lncRNAs in patients with ependymal tumors may facilitate the classification of the tumors and the development of personalized therapies.

Functional and epigenomic characteristics suggest the roles of lncRNAs in spinal cord ependymal tumors

KEGG pathway enrichment analysis was performed in order to analyze the PCGs interacting with the identified lncRNA signatures. In total, 128 PCGs were analyzed. A total of 39 significantly enriched pathways (adjusted P<0.01; Fig. 5A) were identified. Out of 39 pathways, 17 were associated with cancer, including ‘transcriptional misregulation in cancer’, ‘p53 signalling pathway’, ‘colorectal cancer’ and ‘bladder cancer’. Furthermore, the pathway ‘proteoglycans in cancer’ was identified, which is involved in central nervous system-associated functions and diseases (49). Chondroitin sulphate proteoglycans (CSPGs) are enriched in the nervous system and contribute to neural cell migration and axon extension (50). A previous study demonstrated that CSPGs are upregulated after spinal cord injury (51). Additionally, the tumor necrosis factor (TNF) signaling pathway was identified, which is involved in various diseases (Fig. 5C). Interestingly, most of the lncRNAs identified to exhibit subtype-specific expression were hypomethylated compared with the total amount of lncRNAs identified (Figs. 1C and 5B).

Discussion

Ependymal tumor is a rare type of malignant tumor (52). Ependymal tumors can arise from both the brain and spinal cord (53). Notably, depending on its origin, there is a large genetic difference between these two types of ependymal tumor (54). Previous studies have investigated ependymal tumor arising from the brain (6,55), and the current knowledge of the molecular characteristics of spinal cord-derived ependymal tumor remains limited.

By investigating the DNA methylation status of lncRNAs, the present study provided novel insight into the understanding of ependymal tumors and the possible treatments of patients with this disease. The present study investigated the DNA methylation profile of multiple lncRNA promoters and identified that the majority of lncRNAs in ependymal tumors presented high methylation levels. The present study suggested that the DNA methylation level in lncRNA promoters are consistent among samples belonging to the same tumor subtype. The regions with differential methylation levels corresponded to lncRNA signatures that may be involved in spinal cord development.

The present study suggested that tumor subtype-specific lncRNAs may be involved in spinal cord development. By integrating lncRNA-PCG interaction data, a total of 30 lncRNAs were identified, and their DNA methylation levels may be a signature in spinal cord ependymal tumors. Some of these lncRNAs were identified to be associated with cancer and nervous system diseases. The PCGs regulated by the lncRNA signatures were identified to be enriched in many cancer- and nervous system-associated pathways. Interestingly, most of the lncRNAs interacting with cancer- and nervous system-associated proteins were found to be hypomethylated. In addition, the TNF signaling pathway was identified as being involved in spinal cord ependymal tumorigenesis. In previous studies, TNF-α was shown to be involved in cell survival, apoptosis, inflammation and immunity (5658), and its role has been investigated in various diseases, particularly in cancer (59). A previous study investigating the function of TNF in the nervous system demonstrated that TNF affects the nervous system, and it is involved in neurodegenerative diseases (60). Although previous studies have examined the function of the TNF signalling pathway in the nervous system, the present study may provide insight for future studies aimed to investigate its function in spinal cord tumors. In the present study, lncRNAs with target genes associated with cancer and the nervous system were identified, and their methylation patterns in tumor samples were investigated. Therefore, DNA methylation may be a principal factor in regulating the biological features of lncRNAs, thus influencing the function of proteins downstream of these lncRNAs.

Collectively, spinal cord ependymal tumors were investigated using a novel approach, and the methylation status of lncRNAs in ependymal tumors was characterized. The present study may lay the foundations for future studies aimed to investigate spinal cord tumors. However, the present results require validation using a high number of tumor samples. The present findings may contribute to the development of novel strategies for diagnosing and treating spinal cord ependymal tumors.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

JW conceived and designed the experiments. LW performed most of the experiments. CZ and YX performed certain experiments. JH and SG identified tumor-subtype specific lncRNA signatures. FX and WJ performed functional enrichment analysis. JH, SG, FX and WJ aided in the interpretation of the results and wrote 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.

Glossary

Abbreviations

Abbreviations:

lncRNA

long non-coding RNA

PCGs

protein coding genes

SP-EPN

spinal ependymoma

SP-SE

spinal subependymoma

SP-MPE

spinal myxopapillary

TSS

transcript start site

GREAT

Genomic Regions Enrichment of Annotations Tool

HM450

Human Methylation 450

CSPGs

chondroitin sulfate proteoglycans

SNP

single nucleotide polymorphism

TEC

To be Experimentally Confirmed

References

1 

Arnautovic K and Arnautovic A: Extramedullary intradural spinal tumors: A review of modern diagnostic and treatment options and a report of a series. Bosn J Basic Med Sci. 9 (Suppl 1):S40–S45. 2009. View Article : Google Scholar :

2 

Allen JC, Siffert J and Hukin J: Clinical manifestations of childhood ependymoma: A multitude of syndromes. Pediatr Neurosurg. 28:49–55. 1998. View Article : Google Scholar : PubMed/NCBI

3 

Thuppal S, Propp JM and McCarthy BJ: Average years of potential life lost in those who have died from brain and CNS tumors in the USA. Neuroepidemiology. 27:22–27. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Pajtler KW, Witt H, Sill M, Jones DT, Hovestadt V, Kratochwil F, Wani K, Tatevossian R, Punchihewa C, Johann P, et al: Molecular classification of ependymal tumors across all CNS compartments, histopathological grades, and age groups. Cancer Cell. 27:728–743. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Pfeffer C and Olsen BR: Editorial: Journal of negative results in biomedicine. J Negative Results Biomed. 1:22002. View Article : Google Scholar

6 

Witt H, Gramatzki D, Hentschel B, Pajtler KW, Felsberg J, Schackert G, Löffler M, Capper D, Sahm F, Sill M, et al: DNA methylation-based classification of ependymomas in adulthood: Implications for diagnosis and treatment. Neuro Oncology. 20:1616–1624. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Ma DL, Lin S, Leung KH, Zhong HJ, Liu LJ, Chan DS, Bourdoncle A, Mergny JL, Wang HM and Leung CH: An oligonucleotide-based label-free luminescent switch-on probe for RNA detection utilizing a G-quadruplex-selective iridium(III) complex. Nanoscale. 6:8489–8494. 2014. View Article : Google Scholar : PubMed/NCBI

8 

He HZ, Chan DS, Leung CH and Ma DL: A highly selective G-quadruplex-based luminescent switch-on probe for the detection of gene deletion. Chem Commun (Camb). 48:9462–9464. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Leung KH, He HZ, Chan DS, Fu WC, Leungb CH and Maa DL: An oligonucleotide-based switch-on luminescent probe for the detection of kanamycin in aqueous solution. Sensors Actuators B: Chemical. 177:487–492. 2013. View Article : Google Scholar

10 

Wu C, Wu KJ, Kang TS, Wang HD, Leung CH, Liu JB and Ma DL: Iridium-based probe for luminescent nitric oxide monitoring in live cells. Sci Rep. 8:124672018. View Article : Google Scholar : PubMed/NCBI

11 

Kiltschewskij D and Cairns MJ: Temporospatial guidance of activity-dependent gene expression by microRNA: Mechanisms and functional implications for neural plasticity. Nucleic Acids Res. 47:533–545. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Kulis M and Esteller M: DNA methylation and cancer. Adv Genet. 70:27–56. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Xiao Y, Yu F, Pang L, Zhao H, Liu L, Zhang G, Liu T, Zhang H, Fan H, Zhang Y, et al: MeSiC: A model-based method for estimating 5 mC levels at Single-CpG resolution from MeDIP-seq. Sci Rep. 5:146992015. View Article : Google Scholar : PubMed/NCBI

14 

Klutstein M, Nejman D, Greenfield R and Cedar H: DNA methylation in cancer and aging. Cancer Res. 76:3446–3450. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Yu F, Quan F, Xu J, Zhang Y, Xie Y, Zhang J, Lan Y, Yuan H, Zhang H, Cheng S, et al: Breast cancer prognosis signature: Linking risk stratification to disease subtypes. Brief Bioinform. Sep 3–2018.doi: 10.1093/bib/bby073 (Epub ahead of print). View Article : Google Scholar

16 

Koch A, Joosten SC, Feng Z, de Ruijter TC, Draht MX, Melotte V, Smits KM, Veeck J, Herman JG, Van Neste L, et al: Analysis of DNA methylation in cancer: Location revisited. Nat Rev Clin Oncol. 15:459–466. 2018. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Wu Q, Kim YC, Lu J, Xuan Z, Chen J, Zheng Y, Zhou T, Zhang MQ, Wu CI and Wang SM: Poly A-transcripts expressed in HeLa cells. PLoS One. 3:e28032008. View Article : Google Scholar : PubMed/NCBI

19 

Yu F, Zhang G, Shi A, Hu J, Li F, Zhang X, Zhang Y, Huang J, Xiao Y, Li X and Cheng S: LnChrom: A resource of experimentally validated lncRNA-chromatin interactions in human and mouse. Database (Oxford). 20182018.doi: 10.1093/database/bay039.

20 

Zhang Y, Li X, Zhou D, Zhi H, Wang P, Gao Y, Guo M, Yue M, Wang Y, Shen W, et al: Inferences of individual drug responses across diverse cancer types using a novel competing endogenous RNA network. Mol Oncol. 12:1429–1446. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Bolha L, Ravnik-Glavac M and Glavac D: Long noncoding RNAs as biomarkers in cancer. Dis Markers. 2017:72439682017. View Article : Google Scholar : PubMed/NCBI

22 

Wang Z, Yang B, Zhang M, Guo W, Wu Z, Wang Y, Jia L, Li S; Cancer Genome Atlas Research Network, ; Xie W and Yang D: lncRNA epigenetic landscape analysis identifies EPIC1 as an oncogenic lncRNA that interacts with MYC and promotes cell-cycle progression in cancer. Cancer Cell. 33:706–720.e9. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Heilmann K, Toth R, Bossmann C, Klimo K, Plass C and Gerhauser C: Genome-wide screen for differentially methylated long noncoding RNAs identifies Esrp2 and lncRNA Esrp2-as regulated by enhancer DNA methylation with prognostic relevance for human breast cancer. Oncogene. 36:6446–6461. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Clough E and Barrett T: The gene expression omnibus database. Methods Mol Biol. 1418:93–110. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Harrow J, Frankish A, Gonzalez JM, Tapanari E, Diekhans M, Kokocinski F, Aken BL, Barrell D, Zadissa A, Searle S, et al: GENCODE: The reference human genome annotation for The ENCODE Project. Genome Res. 22:1760–1774. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM and Haussler D: The human genome browser at UCSC. Genome Res. 12:996–1006. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Zhou W, Laird PW and Shen H: Comprehensive characterization, annotation and innovative use of Infinium DNA methylation BeadChip probes. Nucleic Acids Res. 45:e222017.PubMed/NCBI

28 

Quinlan AR and Hall IM: BEDTools: A flexible suite of utilities for comparing genomic features. Bioinformatics. 26:841–842. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Aine M, Sjödahl G, Eriksson P, Veerla S, Lindgren D, Ringnér M and Höglund M: Integrative epigenomic analysis of differential DNA methylation in urothelial carcinoma. Genome Med. 7:232015. View Article : Google Scholar : PubMed/NCBI

30 

Yi Y, Zhao Y, Li C, Zhang L, Huang H, Li Y, Liu L, Hou P, Cui T, Tan P, et al: RAID v2.0: An updated resource of RNA-associated interactions across organisms. Nucleic Acids Res. 45:D115–D118. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Chen X and Ishwaran H: Random forests for genomic data analysis. Genomics. 99:323–329. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Nguyen TT, Huang JZ and Nguyen TT: Unbiased feature selection in learning random forests for high-dimensional data. TheScientificWorldJournal. 2015:4713712015. View Article : Google Scholar : PubMed/NCBI

33 

McLean CY, Bristor D, Hiller M, Clarke SL, Schaar BT, Lowe CB, Wenger AM and Bejerano G: GREAT improves functional interpretation of cis-regulatory regions. Nat Biotechnol. 28:495–501. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, et al: Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44:W90–W97. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Prayson RA and Suh JH: Subependymomas: Clinicopathologic study of 14 tumors, including comparative MIB-1 immunohistochemical analysis with other ependymal neoplasms. Arch Pathol Lab Med. 123:306–309. 1999.PubMed/NCBI

37 

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

38 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Yao J, Zhang L, Hu L, Guo B, Hu X, Borjigin U, Wei Z, Chen Y, Lv M, Lau JT, et al: Tumorigenic potential is restored during differentiation in fusion-reprogrammed cancer cells. Cell Death Dis. 7:e23142016. View Article : Google Scholar : PubMed/NCBI

40 

Zhang Y, Liu D, Wang L, Wang S, Yu X, Dai E, Liu X, Luo S and Jiang W: Integrated systems approach identifies risk regulatory pathways and key regulators in coronary artery disease. J Mol Med (Berl). 93:1381–1390. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Jiang W, Zhang Y, Meng F, Lian B, Chen X, Yu X, Dai E, Wang S, Liu X, Li X, et al: Identification of active transcription factor and miRNA regulatory pathways in Alzheimer's disease. Bioinformatics. 29:2596–2602. 2013. View Article : Google Scholar : PubMed/NCBI

42 

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

43 

Ferre F, Colantoni A and Helmer-Citterich M: Revealing protein-lncRNA interaction. Brief Bioinform. 17:106–116. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Xiong D, Sheng Y, Ding S, Chen J, Tan X, Zeng T, Qin D, Zhu L, Huang A and Tang H: LINC00052 regulates the expression of NTRK3 by miR-128 and miR-485-3p to strengthen HCC cells invasion and migration. Oncotarget. 7:47593–47608. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Salameh A, Fan X, Choi BK, Zhang S, Zhang N and An Z: HER3 and LINC00052 interplay promotes tumor growth in breast cancer. Oncotarget. 8:6526–6539. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Hajjari M and Salavaty A: HOTAIR: An oncogenic long non-coding RNA in different cancers. Cancer Biol Med. 12:1–9. 2015.PubMed/NCBI

47 

Teschendorff AE, Lee SH, Jones A, Fiegl H, Kalwa M, Wagner W, Chindera K, Evans I, Dubeau L, Orjalo A, et al: HOTAIR and its surrogate DNA methylation signature indicate carboplatin resistance in ovarian cancer. Genome Med. 7:1082015. View Article : Google Scholar : PubMed/NCBI

48 

Zhang JX, Han L, Bao ZS, Wang YY, Chen LY, Yan W, Yu SZ, Pu PY, Liu N, You YP, et al: HOTAIR, a cell cycle-associated long noncoding RNA and a strong predictor of survival, is preferentially expressed in classical and mesenchymal glioma. Neuro Oncol. 15:1595–1603. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Heindryckx F and Li JP: Role of proteoglycans in neuro-inflammation and central nervous system fibrosis. Matrix Biol. 68-69:589–601. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Silver DJ and Silver J: Contributions of chondroitin sulfate proteoglycans to neurodevelopment, injury, and cancer. Curr Opin Neurobiol. 27:171–178. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Rolls A, Shechter R, London A, Segev Y, Jacob-Hirsch J, Amariglio N, Rechavi G and Schwartz M: Two faces of chondroitin sulfate proteoglycan in spinal cord repair: A role in microglia/macrophage activation. PLoS Med. 5:e1712008. View Article : Google Scholar : PubMed/NCBI

52 

Villano JL, Parker CK and Dolecek TA: Descriptive epidemiology of ependymal tumours in the United States. Br J Cancer. 108:2367–2371. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Armstrong TS, Vera-Bolanos E, Bekele BN, Aldape K and Gilbert MR: Adult ependymal tumors: Prognosis and the M. D. Anderson Cancer Center experience. Neuro Oncol. 12:862–870. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Lee CH, Chung CK and Kim CH: Genetic differences on intracranial versus spinal cord ependymal tumors: A meta-analysis of genetic researches. Eur Spine J. 25:3942–3951. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Gittleman H, Boscia A, Ostrom QT, Truitt G, Fritz Y, Kruchko C and Barnholtz-Sloan JS: Survivorship in adults with malignant brain and other central nervous system tumor from 2000–2014. Neuro Oncol. 20 (Suppl 7):vii6–vii16. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Olmos G and Lladó J: Tumor necrosis factor alpha: A link between neuroinflammation and excitotoxicity. Mediators Inflamm. 2014:8612312014. View Article : Google Scholar : PubMed/NCBI

57 

Mak TW and Yeh WC: Signaling for survival and apoptosis in the immune system. Arthritis Res. 4 (Suppl 3):S243–S252. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Parameswaran N and Patial S: Tumor necrosis factor-alpha signaling in macrophages. Crit Rev Eukaryot Gene Expr. 20:87–103. 2010. View Article : Google Scholar : PubMed/NCBI

59 

van Horssen R, Ten Hagen TL and Eggermont AM: TNF-alpha in cancer treatment: Molecular insights, antitumor effects, and clinical utility. Oncologist. 11:397–408. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Probert L: TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience. 302:2–22. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang L, Zhang C, Xie Y, Jiang W, Huang J, Guo S, Xu F and Wang J: Detecting the long non‑coding RNA signature related to spinal cord ependymal tumor subtype using a genome‑wide methylome analysis approach. Mol Med Rep 20: 1531-1540, 2019
APA
Wang, L., Zhang, C., Xie, Y., Jiang, W., Huang, J., Guo, S. ... Wang, J. (2019). Detecting the long non‑coding RNA signature related to spinal cord ependymal tumor subtype using a genome‑wide methylome analysis approach. Molecular Medicine Reports, 20, 1531-1540. https://doi.org/10.3892/mmr.2019.10388
MLA
Wang, L., Zhang, C., Xie, Y., Jiang, W., Huang, J., Guo, S., Xu, F., Wang, J."Detecting the long non‑coding RNA signature related to spinal cord ependymal tumor subtype using a genome‑wide methylome analysis approach". Molecular Medicine Reports 20.2 (2019): 1531-1540.
Chicago
Wang, L., Zhang, C., Xie, Y., Jiang, W., Huang, J., Guo, S., Xu, F., Wang, J."Detecting the long non‑coding RNA signature related to spinal cord ependymal tumor subtype using a genome‑wide methylome analysis approach". Molecular Medicine Reports 20, no. 2 (2019): 1531-1540. https://doi.org/10.3892/mmr.2019.10388