Differential long non‑coding RNA and mRNA expression in differentiated human glioblastoma stem cells

  • Authors:
    • Hao Li
    • Haowen Li
    • Yajing Hao
    • Yuming Jiao
    • Zhicen Li
    • Haiyan Yue
    • Zhe Xu
    • Shuo Wang
    • Yong Cao
    • Jizong Zhao
  • View Affiliations

  • Published online on: July 12, 2016     https://doi.org/10.3892/mmr.2016.5505
  • Pages: 2067-2076
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Differentiation of glioblastoma stem cells (GSCs) may lead to inhibition of their self‑renewing ability and tumorigenic potential, as well as increasing their sensitivity to treatment. The critical role of long non‑coding RNAs (lncRNAs) in numerous biological processes has been revealed. However, the involvement of lncRNAs in GSC differentiation remains to be elucidated. In the present study, GSCs were isolated from patient samples and differentiation was induced. Using a high‑throughput microarray, the present study identified a profile of 1,545 lncRNAs and 2,729 mRNAs that differed between GSCs and their non‑differentiated counterparts. To ascertain the association between the altered lncRNAs and mRNAs, a co‑expression network was constructed in which 1,087 lncRNAs and 1,928 mRNAs altered upon GSC differentiation formed a total of 19,642 lncRNA‑mRNA pairs. Based on the co‑expression network, the lncRNA functions were additionally predicted by a cis‑ or trans‑ targeting program. Furthermore, three pairs of lncRNAs and their nearby target mRNAs were selected [ENSG00000261924.1‑regulatory associated protein of MTOR complex 1, ENSG00000235427.1‑caveolin 1 and Tax1 binding protein 3 (TAX1BP3)‑purinergic receptor P2X 5 (P2RX5)‑TAX1BP3] and their expression levels were validated by reverse transcription‑quantitative polymerase chain reaction. The altered lncRNAs were also regulated by various pluripotency transcription factors (POU domain, class 3, transcription factor, sex determining region Y‑box 2, spalt‑like transcription factor 2 and oligodendrocyte lineage transcription factor 2). In conclusion, the results of the present study revealed that lncRNAs may function in GSC differentiation by regulating their target mRNAs, and a set of lncRNAs were identified as candidates for further study concerning the future treatment of GSCs.

Introduction

Glioblastoma multiforme (GBM), which accounts for ~40% of all primary brain tumors, is one of the most malignant tumors in humans, with patients having a median overall survival time of 12–15 months (1). Previous studies have demonstrated that tumor cell populations in GBM are heterogeneous in terms of morphology and differentiation status (2). A highly tumorigenic and self-renewing subpopulation of cells, which display stem-like behavior, is the glioblastoma stem cells (GSCs) (2,3). Notably, the property of 'stemness' in GSCs is believed to be key for tumor formation, differentiation, proliferation and resistance to chemo- and radiotherapy, possibly explaining the high frequency of treatment failure and tumor relapse observed in glioblastoma (4,5). The differentiation of GSCs may lead to the inhibition of their self-renewing ability and tumorigenic potential, as well as increasing their sensitivity to treatment. The promotion of differentiation is considered to be a potential strategy to eradicate GSCs, and therefore it is important to understand the detailed molecular mechanisms involved in GSC differentiation (6,7).

Advances in next-generation deep sequencing technologies have identified a large number of non-coding RNAs (ncRNAs) termed long non-coding RNAs (lncRNAs). These molecules are >200 nucleotides in length and are a class of single-stranded RNAs, which lack protein-coding ability (8,9). Rather than being irrelevant transcriptional noise, studies have revealed that lncRNAs possess critical regulatory roles in numerous biological processes, including immune responses, cellular metabolism, stem cell differentiation and tumorigenesis (10,11). Furthermore, certain studies have demonstrated that lncRNA signatures correlate with glioma malignancy grade, histological differentiation and prognosis (12,13). The diverse mechanisms underlying the regulatory roles of lncRNAs include interactions with mRNAs, functioning as miRNA sponges and acting as tethers, guides, decoys and scaffolds for proteins (14,15).

Numerous studies of GSC differentiation have revealed certain underlying molecular mechanisms involved in this process. Certain transcription factors (TFs), protein-coding genes and ncRNAs have been proposed to be involved in the maintenance of GSC stemness, and the regulation of these factors may facilitate GSC differentiation (1618). However, the role of lncRNAs in the GSC differentiation process remains to be elucidated.

In the present study, using patient-derived GSC lines and their differentiation-induced GSC (DGSC) counterparts, a high-throughput microarray analysis was performed to identify the profile changes of lncRNAs and mRNAs. Using bioinformatics analysis, aberrant lncRNA and mRNA expression was integrated and potential regulatory roles of lncRNAs in GSC differentiation were proposed.

Materials and methods

GSC culture and differentiation

GSCs (G0, G1, G2, G3, G4 and G5) were isolated from neurosurgical samples from six patients with GBM who were hospitalized in the Department of Neurosurgery of Beijing Tiantan Hospital (Beijing, China). Informed consent was obtained from the patients and approval was given from the Ethics Committee of Beijing Tiantan Hospital (KY2014-021-02; Beijing, China). Tumor tissue was sectioned into 1-mm3 fragments using scissors and washed with Dulbecco's modified Eagle's medium/nutrient mixture F12 (DMEM/F12; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) three times. The fragments were then digested with 0.02% trypsin for 20 min and dissociated into single cells. The cells were resuspended and maintained in GSC-propagating medium composed of 2 mM GlutaMAX (Invitrogen; Thermo Fisher Scientific, Inc.), 20 ng/ml recombinant human epidermal growth factor (R&D Systems, Inc., Minneapolis, MN, USA), 20 ng/ml basic fibroblast growth factor (Invitrogen; Thermo Fisher Scientific, Inc.), N2 supplement (Invitrogen; Thermo Fisher Scientific, Inc.) and B27 supplement (Invitrogen; Thermo Fisher Scientific, Inc.) in DMEM/F12. The above procedure was completed within 1 h of the surgical removal of tissue. GSCs (2×104/mouse; 6 mice) were subcutaneously injected into the left hind flank of 6-week-old female non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice (VitalStar Biotechnology Co., Ltd., Beijing, China). The mice were sacrificed by decapitation 40 days later, and tumors were harvested and stained with hematoxylin and eosin. The animal use was approved by the Institutional Animal Care and Use Committee of Capital Medical University. All animal procedures were performed in accordance with the 1996 Guide for the Care and Use of Laboratory Animals (19). The rats were housed in an air-conditioned room with a constant temperature of 22°C and a 12 h light/12 h dark cycle. The humidity in the housing room was 50–60%. The rats had free access to food and water. GSCs were sustained in GSC-propagating medium and it took ~18 h for them to undergo symmetric divisions. To induce GSC differentiation, following 5 passages which took about half a month in total following the resuscitation in GSC-propagating medium, GSCs were resuspended and cultured in DMEM/F12 medium containing 10% fetal bovine serum (FBS) (Gibco; Thermo Fisher Scientific, Inc.) without N2 and B27 supplements for 4 days, as previously described (16,20).

Immunofluorescence

GSCs grown as neurospheres were collected by centrifugation at 150 × g for three min at room temperature, then the cells were transferred to microscope slides, while the DGSCs were grown and stained in 24-well plates. The cells were fixed with 4% formaldehyde, permeabilized with 0.1% Triton X-100 for 10 min and then blocked with 10% normal goat serum (Jackson ImmunoResearch Laboratories, Inc., West Grove, PA, USA) for 5 min. The cells were then incubated at 4°C overnight with the following primary antibodies: Mouse anti-cluster of differentiation (CD) 133 (dilution 1:500; catalog no., MAB4399; EMD Millipore, Billerica, MA, USA); rabbit anti-nestin (dilution 1:250; catalog no., ab82375; Abcam, Cambridge, UK); rabbit anti-glial fibrillary acidic protein (GFAP; dilution 1:500; catalog no., ab33922; Abcam); rabbit anti-βIII tubulin (Tuj1; dilution 1:500; catalog no., ab18207; Abcam); mouse anti-O4 (dilution 1:200; catalog no., MAB345; EMD Millipore). Following washing with phosphate-buffered saline 3 times, the cells were incubated with fluorescein isothiocyanate (FITC)-conjugated anti-mouse (dilution 1:1,000, catalog no., F-11,021; Thermo Fisher Scientific, Inc.) and anti-rabbit (dilution 1:2,000; catalog no., 65-6111; Thermo Fisher Scientific, Inc.) secondary antibodies and Texas Red (TR) -conjugated anti-mouse (dilution 1:2,000; catalog no., T-6390; Thermo Fisher Scientific, Inc.) and anti-rabbit (dilution 1:1,000 catalog no., T-2767; Thermo Fisher Scientific, Inc.) secondary antibodies. The cell nuclei were counterstained with DAPI (Sigma-Aldrich, St. Louis, MO, USA). Images were captured using a DMI 4,000 Leica fluorescent microscope and processed using the Leica Application Suite version 4.2 Imaging System software (Leica Microsystems GmbH, Wetzlar, Germany).

RNA extraction and microarray analysis

Total RNA was extracted from GSCs and DGSCs using TRIzol® reagent (Sigma-Aldrich) according to the manufacturer's instructions. RNA purity and concentration was assessed using a NanoDrop ND-1,000 spectrophotometer (Thermo Fisher Scientific, Inc.). RNA integrity was determined by formaldehyde denaturing gel electrophoresis. Microarray hybridization was performed by CapitalBio Corporation (Beijing, China). Briefly, each RNA sample was amplified and transcribed into double-stranded complementary DNA (cDNA). The labeled cDNA was then hybridized to the lncRNA+mRNA Human Gene Expression Microarray version 4.0, 4×180K chip. Data normalization, quality control and the calculation of differences in gene expression were performed with the GeneSpring GX version 11.5.1 software package (Agilent Technologies, Inc., Santa Clara, CA, USA). lncRNAs and mRNAs were defined as differentially expressed (DE) if the fold-change values were >2.0, or if P<0.05. The microarray data was uploaded to the Gene Expression Omnibus (GEO) database (GSE68343; www.ncbi.nlm.nih.gov/geo/). In addition, hierarchical clustering with average linkage was used for calculating the distinguishable lncRNA and mRNA expression patterns with Cluster version 3.0 software (bonsai.hgc.jp/~mdehoon/software/cluster/software.htm#ctv).

Co-expression network construction and lncRNA target prediction

The lncRNA-mRNA co-expression network was constructed based on associations between DE lncRNAs and DE mRNAs during GSC differentiation. Using the open source bioinformatics software Cytoscape (Institute of Systems Biology, Seattle, WA, USA), a network of the selected lncRNA-mRNA pairs was drawn (Pearson correlation coefficient >0.99 or <−0.99; and P<0.05). Subsequently, the lncRNA-mRNA co-expression network was analyzed using a cis- or trans-regulatory prediction program. A target gene was defined as cis if the mRNA transcribed was within a 10 kilo-base (kb) window upstream or downstream of the lncRNA genomic location, using gene annotations from the University of California, Santa Cruz (Santa Cruz, CA, USA; genome.ucsc.edu/). As lncRNAs may act as competing endogenous RNAs, which regulate the mRNA transcripts by competing for shared microRNAs, the trans- predictions were primarily executed by searching the pairs of lncRNAs and mRNAs that had similar sequences in the 3′ untranslated region using the BLAST-Like Alignment Tool (default parameter settings) software (genome.ucsc.edu/cgi-bin/hgBlat).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) validation

Total RNA was reverse transcribed to cDNA using the PrimeScript RT Reagent kit (Takara Bio, Inc., Otsu, Japan). Using a 7500 Real Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) with SYBR® Premix Ex Taq™ II (Tli RNaseH Plus; Takara Bio, Inc.), RT-qPCR was performed according to the manufacturer's instructions. The specific primers for each gene are presented in Table I. All experiments were performed in triplicate. Gene expression was normalized to GAPDH in each sample.

Table I

Reverse transcription-quantitative polymerase chain reaction primers.

Table I

Reverse transcription-quantitative polymerase chain reaction primers.

NamePrimer sequence
ForwardReverse
ENSG00000235427.1 5′-AAAACCACTGAGACACGGAGGC-3′ 5′-CCAGGGACAGGCAGACATCA-3′
TAX1BP3 5′-CCTACATCCCGGGCCAGC-3′5′- CACCTCCAATGCTGAAACCCA-3′
CAV1 5′-CGACCCTAAACACCTCAACGA-3′5′- GGCAGACAGCAAGCGGTAAAA-3′
RPTOR 5′-GTGGTGGACTGGGAGCAGGAGA-3′ 5′-TGAGCGGTGGGAATCACAGGA-3′
ENSG00000261924.1 5′-AGGAATGACATGAACACGAGGGAA-3′ 5′-CCAGGGCGATATGTGGAGCAA-3′
P2RX5-TAX1BP3 5′-CGAGGCGAAGCGTGGAA-3′ 5′-TGGTGTAAGGGAGAAGCAGAGG-3′
CD133 5′-TACCAAGGACAAGGCGTTCACAGA-3′ 5′-GTGCAAGCTCTTCAAGGTGCTG-3′
Nestin 5′-CGTTGGAACAGAGGTTGGAG-3′ 5′-TAAGAAAGGCTGGCACAGGT-3′
Tuj1 5′-GTACGAAGACGACGAGGAGG-3′ 5′-GCCTGGAGCTGCAATAAGAC-3′
GFAP 5′-GTCCATGTGGAGCTTGACG-3′ 5′-GCAGGTCAAGGACTGCAACT-3′
SOX2 5′-ATGCACAACTCGGAGATCAG-3′ 5′-TATAATCCGGGTGCTCCTTC-3′
GAPDH 5′-CTATAAATTGAGCCCGCAGCC-3′ 5′-GCGCCCAATACGACCAAATC-3′

[i] TAX1BP3, Tax1 binding protein 3; CAV1, caveolin 1; RPTOR, regulatory associated protein of MTOR complex 1; P2RX5, purinergic receptor P2X 5; CD133, cluster of differentiation 133; Tuj1, βIII tubulin; GFAP, glial fibrillary acidic protein; SOX2, sex determining region Y-box 2.

Analysis of TF binding sites and lncRNA conservation

Genomic annotations of human lncRNA genes and transcripts were downloaded from the GENCODE website (www.gencodegenes.org/). Peak lists of chromatin immunoprecipitation sequencing (ChIP-Seq) datasets performed on 4 TFs [POU domain, class 3, transcription factor (POU3F), sex determining region Y-box 2 (SOX2), spalt-like transcription factor 2 (SALL2) and oligodendrocyte lineage transcription factor (OLIG) 2] were available from the deep sequencing of ChIP-Seq libraries in GSCs (21). Subsequently, the peaks of the TFs were overlapped with the regulatory regions of the lncRNA genes (5 kb upstream from the transcription start site) and the gene body of the lncRNA using the InterSectBed tool from Bedtools (bedtools.readthedocs.io/en/latest/). If the regulatory regions or the gene body of an lncRNA gene had at least one TF peak, the lncRNA gene was determined as having the potential to be regulated by the TF. Conservation analysis was conducted using phastCons data (compgen.cshl.edu/phast/) as described previously (22). If the average phastCons score was >0.1, the lncRNA was defined as conserved.

Statistical analysis

Statistical analyses were performed using SPSS software, version 13.0 (SPSS, Inc., Chicago, IL, USA). Values are presented as the mean ± standard deviation, and Student's t-test was used for comparing sample sets. P<0.05 was considered to indicate a statistically significant difference.

Results

Characterization of GSCs isolated from surgical GBM samples and determination of GSC differentiation

GSCs were cultured in GSC-propagating media, where they formed characteristic renewable neurospheres and were able to proliferate indefinitely (Fig. 1Aa). Immunofluorescence revealed that GSCs exhibited an increased proportion of cells positive for CD133 (Fig. 1Ab), and increased expression of nestin, a neural progenitor cell marker (Fig. 1Ac), compared with DGSCs (Fig. 1Ad and e). The tumorigenesis of GSCs was confirmed by tumor formation in NOD/SCID mice following inoculation with 2×104 GSCs (data not shown).

The differentiation of GSCs was induced in DMEM/F12 medium containing 10% FBS for 4 days. Following serum exposure, the GSCs acquired glial- and neurite-like cell features, with protrusions and adherence to the flask, observed using an optical microscope (Fig. 1Af). In addition, immunofluorescent staining analysis confirmed astrocytic- and neural-cell differentiation, indicated by the positive staining with anti-GFAP and anti-Tuj1 in the majority of DGSCs (Fig. 1Ag and h), but not in GSCs (Fig. 1i and j). Furthermore, it was observed that a small proportion of the cells differentiated into the oligodendrocytic lineage, as demonstrated by positive staining for O4 sulfatides in DGSCs (Fig. 1Ak), but not in GSCs (Fig. 1Al). The results of the present study are consistent with a previous study (16), and indicate that GSCs may be efficiently induced into astrocytic and neural lineages, and partly induced into oligodendrocytic lineages. RT-qPCR detected similar trends in CD133 (P=0.02), nestin (P=0.03), GFAP (P<0.001) and Tuj1 (P<0.001) expression in the GSC differentiation process. In addition, SOX2 (P<0.001), which functions in the maintenance of GSC stemness, was markedly decreased in DGSCs compared with the expression observed in GSCs (Fig. 1B).

lncRNAs and mRNAs are differentially expressed during GSC differentiation

Microarray analysis was performed to investigate the expression of lncRNAs and mRNAs during GSC differentiation. A total of 1,545 lncRNAs were differentially expressed greater than 2-fold (P<0.05) during the GSC differentiation process, of which 650 lncRNAs were upregulated and 895 were downregulated. In addition, 2,729 mRNAs were differentially expressed greater than 2-fold (P<0.05) between GSCs and DGSCs, of which 2,179 were upregulated and 550 were downregulated. Subsequently, Cluster 3.0 software was used for the clustering of the lncRNA and mRNA expression data. As presented in Fig. 2A, the GSCs exhibited similar expression patterns of the lncRNAs and mRNAs, but were distinct from the DGSCs. The results of the present study indicated that the expression patterns of lncRNAs and mRNAs may be associated with stemness, and that lncRNAs and mRNAs may have potential roles in regulating GSC differentiation.

To aid the interpretation of the functionality of the lncRNAs, the DE lncRNAs were classified into five categories: Antisense, intergenic, intronic, divergent and others, based on their locations relative to nearby protein-coding genes (23). The anatomy of the lncRNA loci implied that the lncRNAs have potential functions in regulating their neighboring protein-coding genes. According to this categorization method, nearly half the lncRNAs (593/1,545) (Fig. 2B) belonged to the intergenic lncRNA category, of which a large proportion may have important functions due to their clear conservation across mammalian species and their association with chromatin-modifying complexes and gene expression (24).

lncRNAs are significantly correlated with mRNAs

While the differential expression of lncRNAs and protein-coding mRNAs indicated that this subset of lncRNAs may be associated with the local transcriptional activity of protein-coding mRNAs, a lncRNA-mRNA co-expression network was constructed to ascertain the correlation between DE lncRNAs and DE mRNAs in GSC differentiation. A total of 19,642 lncRNA-mRNA pairs, composed of 1,087 DE lncRNAs and 1,928 DE mRNAs, were identified. In the network, single lncRNAs were associated with multiple (between 1 and 10) mRNAs and vice versa (data not shown). The lncRNA-mRNA network was large and complex; therefore, to present the association between lncRNAs and mRNAs more clearly, DE lncRNAs and DE mRNAs with fold-change values >3.0 and P<0.05 were selected to generate a 'core' sub-network map (data not shown). From the core sub-network, a schematic of eight RNA hub nodes (RNA nodes were designated as hub nodes if they were associated with multiple RNAs, implying their important functional roles) and their associated RNAs was drawn (Fig. 3A). Hub nodes were selected based on the degree distribution graph (Fig. 3B; degree was used to depict the number of RNAs associated with one RNA node, and one hub node was defined when its degree was ≥10). Among them, C20orf27 was a protein-coding mRNA, and the remaining 7 hub nodes (uc.359-, XLOC_006350, ENSG00000232956.3, ENSG00000172965.7, ENSG00000242808.2, ENSG00000172965.7 and XLOC_003079) were lncRNAs, suggesting that lncRNAs were more likely to be executing functions via the regulation of other RNAs due to their multiple connections with other RNAs. These lncRNA hub nodes were associated with protein-coding mRNAs, including leukemia inhibitory factor, OLIG1 and fibroblast growth factor 2, which have key roles in the regulation of biological processes associated with stem cells. Together, these results suggest that DE lncRNAs may potentially be involved in GSC differentiation via associations with protein-coding mRNAs.

lncRNA functional prediction via cis- and trans- target prediction

To improve the prediction accuracy of the functions of the lncRNAs, cis- and trans- target prediction programs were utilized based on the lncRNA-mRNA co-expression network (containing 19,642 lncRNA-mRNA pairs, as described previously) obtained. A total of 30 lncRNA-mRNA matched pairs were fitted with cis- regulatory effects and 13 lncRNA-mRNA matched pairs were fitted with trans- regulatory effects (Fig. 4). Among the matched pairs, lncRNA purinergic receptor P2X 5 (P2RX5)-Tax1 binding protein 3 (TAX1BP3) had 3 cis- or trans-genes (TAX1BP3, derlin 2 and proteasome 26S subunit, non-ATPase 12), lncRNA ENSG00000261924.1 had 2 cis- or trans-genes [kinesin family member 1C and regulatory associated protein of MTOR complex 1 (RPTOR)], and the remaining lncRNAs had 1 cis- or trans-gene each. The functional roles of the target protein-coding mRNAs, including RPTOR, in stem cell biology or glioma malignancy (25,26) raised the possibility that lncRNAs regulate GSC differentiation through regulation of their target mRNAs. Thus, the results identified the most plausible functional lncRNAs and their target mRNAs through a series of bioinformatics filter strategies.

Validation of the lncRNAs and their predicted protein-coding mRNA genes

To evaluate the consistency of the microarray and confirm expression of lncRNAs and their target mRNAs, RT-qPCR was performed in samples G0-G5 and their differentiated counterparts. A total of three pairs [RPTOR-ENSG00000261924.1, cave ol i n 1 (CAV1) -ENSG00000235427.1 a nd TAX1BP3-P2RX5-TAX1BP3] were selected for investigation, according to fold-change values and reported gene functions. Whilst the fold-change values of the lncRNAs and mRNAs varied between samples, the expression trends were similar to the microarray results. These data, presented in Fig. 5, verify the consistency of the microarray and confirm the expression of lncRNAs and their target mRNAs despite the tumor-specific genetic background. The P-values for the expression level of DGSCs compared to that of GSCs for each transcript in each sample were as follows: RPTOR G0, P=0.002; G1, P=0.04; G2, P<0.001; G3, P<0.001; G4, P=0.004; G5, P=0.01; ENSG00000261924.1 G0, P=0.008; G1, P=0.012; G2, P=0.04; G3, P=0.04; G4, P=0.02; G5, P<0.001; CAV1 G0, P<0.001; G1, P<0.001; G2, P<0.001; G3, P<0.001; G4, P<0.001; G5, P<0.001; ENSG00000235427.1 G0, P<0.001; G1, P<0.001; G2, P<0.001; G3, P=0.003; G4, P<0.001; G5, P=0.08; TAX1BP3 G0, P<0.001; G1, P=0.008; G2, P=0.01; G3, P<0.001; G4, P<0.001; G5, P<0.001; P2RX5-TAX1BP3 G0, P=0.001; G1, P<0.001; G2, P=0.004; G3, P=0.02; G4, P=0.01; G5, P<0.001.

Identification of DE lncRNAs regulated by a set of core TFs

A set of core TFs (POU3F, SOX2, SALL2 and OLIG2) have previously been demonstrated to bind to, and activate, GSC-specific regulatory elements, and thus determine and reprogram the differentiation status of the GSCs (21). In the present study, microarray and RT-qPCR revealed that the expression of all four TFs decreased upon differentiation of GSCs to DGSCs, which was consistent with a previous study (21). A total of 262 lncRNAs were revealed to contain at least one TF binding site in their regulatory regions or gene body, and 21 lncRNAs contained binding sites for all four TFs. The results are summarized as a Venn diagram in Fig. 6A. In addition, a representative schematic of the binding of the four TFs in the ENSG00000242808 regions is presented in Fig. 6B. Furthermore, as evidence has suggested that conservation often indicates functionality (27), the sequence conservation of the 21 lncRNAs was then evaluated with the phastCons score, with 10 lncRNAs confirmed to be conserved among vertebrates (data not shown). Taken together, the present results suggest that the DE lncRNAs are regulated by a set of core TFs, and that they may be integrated into known pluripotency TF networks regulating GSC differentiation.

Discussion

There is a growing understanding of the importance of lncRNAs in the regulation of pluripotency and differentiation in stem cells, including cancer stem cells (14,28). In addition, certain classical lncRNAs, including HOX transcript antisense RNA (HOTAIR), metastasis associated lung adenocarcinoma transcript 1 (MALAT-1) and long intergenic non-protein coding RNA, regulator of reprogramming (Linc-RoR), whose roles in numerous biological processes have been widely investigated, have been reported to possess regulatory roles in stemness maintenance and differentiation in various cancer stem cells (2931). However, no significant alterations in HOTAIR, MALAT-1 and Linc-RoR expression were observed in the present study, which may be partly explained by the differences between cell lines of different types of cancer. Based on transcriptome microarray analysis and deep sequencing technologies, a number of novel lncRNAs were observed to regulate stem cell stemness and differentiation in the present study, notably LncTCF7 and pnky. Previously, LncTCF7 was reported to maintain the stemness of human liver cancer stem cells via the activation of Wnt signaling (32). Pnky, a conserved lncRNA, has been demonstrated to interact with and regulate the neuronal differentiation of embryonic and postnatal neural stem cells (33).

Despite a number of lncRNAs being examined in a variety of cancer stem cells, few studies have reported lncRNA profile and their roles in GSC differentiation. In the current study, the use of an lncRNA-mRNA human gene expression microarray platform identified for the first time a total of 1,545 lncRNA profile changes during the differentiation of patient-derived GSCs. Using a series of bioinformatics strategies for lncRNA prediction, it was observed that DE lncRNAs may interact with protein-coding mRNAs by associating with them and in addition through cis- or trans-targeting. Certain protein-coding mRNAs that are regulated by lncRNAs possess significant biological functions in cancer or stem cells. For example, RPTOR, a regulatory protein that forms the stoichiometric mammalian target of rapamycin complex 1 (mTORC1), is indispensable for the kinase activity of mTORC1, through which it may exert its functions (25). A previous study demonstrated that RPTOR-deficient mesenchymal stem cells have impaired mTORC1 signaling and a reduced capacity to form lipid-laden adipocytes (34). Furthermore, a differentiation regulation capacity for mTORC1 in GSCs has been suggested (26). In addition to RPTOR, certain mRNAs, including CAV1, tropomyosin 1, TAX1BP3 and NADPH quinone dehydrogenase 1, have previously been reported to be involved in stem cell differentiation or glioma malignancy (3539). These results highlight the roles of lncRNAs in regulating GSC differentiation through associating with, and targeting, a series of functional protein-coding mRNAs.

A previous study has suggested that pluripotent lncRNAs may be regulated by TFs known to regulate pluripotency (40). It is well-established that the GSC differentiation process may be artificially manipulated via the induction of combinations of a set of core pluripotent TFs including POU3F2, SOX2, SALL2 and OLIG2 (21). The partially identified functional targets to which the four core TFs bind are responsible for reprograming the differentiation process in GSCs (21). In the present study, DE lncRNAs were integrated into the core pluripotency TF network to provide a more comprehensive network involved in the regulation of GSC differentiation. The results of the present study not only reveal that a large proportion of DE lncRNAs are regulated by TFs, but in addition provide an alternative method to interpret the functions of the core TFs that bind to and activate DE lncRNAs and thus direct the GSC differentiation process.

In conclusion, the present study identified a lncRNA and mRNA expression profile that clearly distinguished GSCs from DGSCs. Using a series of bioinformatics strategies for lncRNA prediction, it was observed that DE lncRNAs were able to regulate the GSC differentiation process through an association with protein-coding mRNAs and additionally through cis- or trans-targeting. Furthermore, the DE lncRNAs were regulated by a set of core pluripotency TFs. The results of the present study provide a potential direction for future research via the comprehensive integration of lncRNAs and mRNAs, and the identification of a set of lncRNAs as candidates for further study.

Acknowledgments

The authors would like to thank Dr Anchen Guo at the Laboratory of Clinical Medicine Research, Beijing Tiantan Hospital for providing the GSC-propagating medium. The present study was supported by a grant from the China National Clinical Research Center for Neurological Diseases, the Training Plan for Beijing High-Level Healthcare Personnel (grant no. 2011-3-28).

References

1 

Quick A, Patel D, Hadziahmetovic M, Chakravarti A and Mehta M: Current therapeutic paradigms in glioblastoma. Rev Recent Clin Trials. 5:14–27. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Orza A, Soriţău O, Tomuleasa C, Olenic L, Florea A, Pana O, Bratu I, Pall E, Florian S, Casciano D and Biris AS: Reversing chemoresistance of malignant glioma stem cells using gold nanoparticles. Int J Nanomedicine. 8:689–702. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Friedmann-Morvinski D: Glioblastoma heterogeneity and cancer cell plasticity. Crit Rev Oncog. 19:327–336. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Jackson M, Hassiotou F and Nowak A: Glioblastoma stem-like cells: At the root of tumor recurrence and a therapeutic target. Carcinogenesis. 36:177–185. 2015. View Article : Google Scholar

5 

Nakano I: Stem cell signature in glioblastoma: Therapeutic development for a moving target. J Neurosurg. 122:324–330. 2015. View Article : Google Scholar

6 

Binello E and Germano IM: Targeting glioma stem cells: A novel framework for brain tumors. Cancer Sci. 102:1958–1966. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Stockhausen MT, Kristoffersen K, Stobbe L and Poulsen HS: Differentiation of glioblastoma multiforme stem-like cells leads to downregulation of EGFR and EGFRvIII and decreased tumorigenic and stem-like cell potential. Cancer Biol Ther. 15:216–224. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Jiang YJ and Bikle DD: LncRNA profiling reveals new mechanism for VDR protection against skin cancer formation. J Steroid Biochem Mol Biol. 144:87–90. 2014. View Article : Google Scholar

9 

Thum T and Condorelli G: Long noncoding RNAs and microRNAs in cardiovascular pathophysiology. Circ Res. 116:751–762. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Curtale G and Citarella F: Dynamic nature of noncoding RNA regulation of adaptive immune response. Int J Mol Sci. 14:17347–17377. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Lin N, Chang KY, Li Z, Gates K, Rana ZA, Dang J, Zhang D, Han T, Yang CS, Cunningham TJ, et al: An evolutionarily conserved long noncoding RNA TUNA controls pluripotency and neural lineage commitment. Mol Cell. 53:1005–1019. 2014. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Zhang XQ, Sun S, Lam KF, Kiang KM, Pu JK, Ho AS, Lui WM, Fung CF, Wong TS and Leung GK: A long non-coding RNA signature in glioblastoma multiforme predicts survival. Neurobiol Dis. 58:123–131. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Eades G, Zhang YS, Li QL, Xia JX, Yao Y and Zhou Q: Long non-coding RNAs in stem cells and cancer. World J Clin Oncol. 5:134–141. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Zhu J, Liu S, Ye F, Shen Y, Tie Y, Zhu J, Jin Y, Zheng X, Wu Y and Fu H: The long noncoding RNA expression profile of hepatocellular carcinoma identified by microarray analysis. PLoS One. 9:e1017072014. View Article : Google Scholar : PubMed/NCBI

16 

Aldaz B, Sagardoy A, Nogueira L, Guruceaga E, Grande L, Huse JT, Aznar MA, Díez-Valle R, Tejada-Solís S, Alonso MM, et al: Involvement of miRNAs in the differentiation of human glioblastoma multiforme stem-like cells. PLoS One. 8:e770982013. View Article : Google Scholar : PubMed/NCBI

17 

Rheinbay E, Suvà ML, Gillespie SM, Wakimoto H, Patel AP, Shahid M, Oksuz O, Rabkin SD, Martuza RL, Rivera MN, et al: An aberrant transcription factor network essential for Wnt signaling and stem cell maintenance in glioblastoma. Cell Rep. 3:1567–1579. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Katsushima K and Kondo Y: Non-coding RNAs as epigenetic regulator of glioma stem-like cell differentiation. Front Genet. 5:142014. View Article : Google Scholar : PubMed/NCBI

19 

Clark JD, Gebhart GF, Gonder JC, Keeling ME and Kohn DF: Special Report: The 1996 guide for the care and use of laboratory animals. ILAR J. 38:41–48. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Nogueira L, Ruiz-Ontañon P, Vazquez-Barquero A, Lafarga M, Berciano MT, Aldaz B, Grande L, Casafont I, Segura V, Robles EF, et al: Blockade of the NFκB pathway drives differentiating glioblastoma-initiating cells into senescence both in vitro and in vivo. Oncogene. 30:3537–3548. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Suvà ML, Rheinbay E, Gillespie SM, Patel AP, Wakimoto H, Rabkin SD, Riggi N, Chi AS, Cahill DP, Nahed BV, et al: Reconstructing and reprogramming the tumor-propagating tumor-propagating potential of glioblastoma stem-like cells. Cell. 157:580–594. 2014. View Article : Google Scholar

22 

Siepel A, Bejerano G, Pedersen JS, Hinrichs AS, Hou M, Rosenbloom K, Clawson H, Spieth J, Hillier LW, Richards S, et al: Evolutionarily conserved elements in vertebrate, insect, worm, and yeast genomes. Genome Res. 15:1034–1050. 2005. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

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

25 

Kim DH and Sabatini DM: Raptor and mTOR: Subunits of a nutrient-sensitive complex. Curr Top Microbiol Immunol. 279:259–270. 2004.

26 

Jhanwar-Uniyal M, Jeevan D, Neil J, Shannon C, Albert L and Murali R: Deconstructing mTOR complexes in regulation of Glioblastoma Multiforme and its stem cells. Adv Biol Regul. 53:202–210. 2013. View Article : Google Scholar

27 

Roberts TC, Morris KV and Wood MJ: The role of long non-coding RNAs in neurodevelopment, brain function and neurological disease. Philos Trans R Soc Lond B Biol Sci. 369:pii: 20130507. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Luo M, Jeong M, Sun D, Park HJ, Rodriguez BA, Xia Z, Yang L, Zhang X, Sheng K, Darlington GJ, et al: Long non-coding RNAs control hematopoietic stem cell function. Cell Stem Cell. 16:426–438. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Jiao F, Hu H, Han T, Yuan C and Wang L, Jin Z, Guo Z and Wang L: Long noncoding RNA MALAT-1 enhances stem cell-like phenotypes in pancreatic cancer cells. Int J Mol Sci. 16:6677–6693. 2015. View Article : Google Scholar :

30 

Pádua Alves C, Fonseca AS and Muys BR: Brief report: The lincRNA Hotair is required for epithelial-to-mesenchymal transition and stemness maintenance of cancer cell lines. Stem Cells. 31:2827–2832. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Zhou X, Gao Q, Wang J, Zhang X, Liu K and Duan Z: Linc-RNA-RoR acts as a 'sponge' against mediation of the differentiation of endometrial cancer stem cells by microRNA-145. Gynecol Oncol. 133:333–339. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, et al: The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell. 16:413–425. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Ramos AD, Andersen RE, Liu SJ, Nowakowski TJ, Hong SJ, Gertz CC, Salinas RD, Zarabi H, Kriegstein AR and Lim DA: The long noncoding RNA Pnky regulates neuronal differentiation of embryonic and postnatal neural stem cells. Cell Stem Cell. 16:439–447. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Martin SK, Fitter S, Dutta AK, Matthews MP, Walkley CR, Hall MN, Ruegg MA, Gronthos S and Zannettino AC: Brief report: The differential roles of mTORC1 and mTORC2 in mesenchymal stem cell differentiation. Stem Cells. 33:1359–1365. 2015. View Article : Google Scholar

35 

Cosset EC, Godet J, Entz-Werlé N, Guérin E, Guenot D, Froelich S, Bonnet D, Pinel S, Plenat F, Chastagner P, et al: Involvement of the TGFβ pathway in the regulation of α5 β1 integrins by caveolin-1 in human glioblastoma. Int J Cancer. 131:601–611. 2012. View Article : Google Scholar

36 

Luanpitpong S, Wang L, Stueckle TA, Tse W, Chen YC and Rojanasakul Y: Caveolin-1 regulates lung cancer stem-like cell induction and p53 inactivation in carbon nanotube-driven tumorigenesis. Oncotarget. 5:3541–3554. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Du HQ, Wang Y, Jiang Y, Wang CH, Zhou T, Liu HY and Xiao H: Silencing of the TPM1 gene induces radioresistance of glioma U251 cells. Oncol Rep. 33:2807–2814. 2015.PubMed/NCBI

38 

Wang H, Han M, Whetsell W Jr, Wang J, Rich J, Hallahan D and Han Z: Tax-interacting protein 1 coordinates the spatiotemporal activation of Rho GTPases and regulates the infiltrative growth of human glioblastoma. Oncogene. 33:1558–1569. 2014. View Article : Google Scholar :

39 

Okamura T, Kurisu K, Yamamoto W, Takano H and Nishiyama M: NADPH/quinone oxidoreductase is a priority target of glioblastoma chemotherapy. Int J Oncol. 16:295–303. 2000.PubMed/NCBI

40 

Ng SY, Johnson R and Stanton LW: Human long non-coding RNAs promote pluripotency and neuronal differentiation by association with chromatin modifiers and transcription factors. EMBO J. 31:522–533. 2012. View Article : Google Scholar :

Related Articles

Journal Cover

September-2016
Volume 14 Issue 3

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
Li H, Li H, Hao Y, Jiao Y, Li Z, Yue H, Xu Z, Wang S, Cao Y, Zhao J, Zhao J, et al: Differential long non‑coding RNA and mRNA expression in differentiated human glioblastoma stem cells. Mol Med Rep 14: 2067-2076, 2016
APA
Li, H., Li, H., Hao, Y., Jiao, Y., Li, Z., Yue, H. ... Zhao, J. (2016). Differential long non‑coding RNA and mRNA expression in differentiated human glioblastoma stem cells. Molecular Medicine Reports, 14, 2067-2076. https://doi.org/10.3892/mmr.2016.5505
MLA
Li, H., Li, H., Hao, Y., Jiao, Y., Li, Z., Yue, H., Xu, Z., Wang, S., Cao, Y., Zhao, J."Differential long non‑coding RNA and mRNA expression in differentiated human glioblastoma stem cells". Molecular Medicine Reports 14.3 (2016): 2067-2076.
Chicago
Li, H., Li, H., Hao, Y., Jiao, Y., Li, Z., Yue, H., Xu, Z., Wang, S., Cao, Y., Zhao, J."Differential long non‑coding RNA and mRNA expression in differentiated human glioblastoma stem cells". Molecular Medicine Reports 14, no. 3 (2016): 2067-2076. https://doi.org/10.3892/mmr.2016.5505