Long noncoding RNAs (lncRNA) are increasingly implicated in oncogenesis. Here, it is determined that LINC00152/CYTOR is upregulated in glioblastoma multiforme (GBM) and aggressive wild-type IDH1/2 grade 2/3 gliomas and upregulation associates with poor patient outcomes. LINC00152 is similarly upregulated in over 10 other cancer types and associates with a poor prognosis in 7 other cancer types. Inhibition of the mostly cytoplasmic LINC00152 decreases, and overexpression increases cellular invasion. LINC00152 knockdown alters the transcription of genes important to epithelial-to-mesenchymal transition (EMT). PARIS and Ribo-seq data, together with secondary structure prediction, identified a protein-bound 121-bp stem-loop structure at the 3′ end of LINC00152 whose overexpression is sufficient to increase invasion of GBM cells. Point mutations in the stem-loop suggest that stem formation in the hairpin is essential for LINC00152 function. LINC00152 has a nearly identical homolog, MIR4435-2HG, which encodes a near identical hairpin, is equally expressed in low-grade glioma (LGG) and GBM, predicts poor patient survival in these tumors, and is also reduced by LINC00152 knockdown. Together, these data reveal that LINC00152 and its homolog MIR4435-2HG associate with aggressive tumors and promote cellular invasion through a mechanism that requires the structural integrity of a hairpin structure.

Implications: Frequent upregulation of the lncRNA, LINC00152, in glioblastoma and other tumor types combined with its prognostic potential and ability to promote invasion suggests LINC00152 as a potential biomarker and therapeutic target. Mol Cancer Res; 16(10); 1470–82. ©2018 AACR.

Glioblastomas (GBM) are highly aggressive grade 4 gliomas and are the most common type of malignant glioma, with 10,000 new diagnoses each year (1). GBMs are a heterogeneous group of tumors that can be separated into four different subtypes, mesenchymal, classical, proneural, and neural, based on their transcriptional profile. Most of the focus on understanding glioma tumor biology has been on studying protein-coding genes and miRNAs (2). These efforts have identified commonly altered signaling pathways in GBMs, including mutations in EGFR, p53, and mTOR signaling (3, 4). Furthermore, miRNAs have been shown to play a role in many of the oncogenic phenotypes of GBMs, such as invasiveness and stemness of GBM stem cells (5, 6). Although there has been much effort on creating new targeted therapies for GBMs focusing on some of the aforementioned pathways, most have not been effective and the standard-of-care therapy, a combination of surgical resection, radiotherapy, and temozolomide, still leaves patients with a 5-year survival rate of roughly 10% (7).

High-throughput sequencing revealed that a majority of the human genome, long thought to be transcriptionally silent, is actually expressed. Indeed, when surveyed across many different cell types, it was found that nearly 80% of the human genome is actually transcribed (8). Many of these newly discovered transcripts are long noncoding RNAs (lncRNA). lncRNAs are a class of ncRNAs that are longer than 200 bases in length and can be further subdivided into subclasses based on chromosomal position relative to other genes, enhancers, or other genomic regulatory elements. lncRNAs have been shown to play many different functional roles in the cell, in part through regulation of transcription, mRNA stability, and mRNA translational efficiency (9, 10). Most of the research into the role of ncRNA in GBMs has been on miRNAs, with relatively few studies on lncRNAs. This leaves a crucial gap in our understanding of glioma pathogenesis. Indeed, lncRNAs have been shown to function in critical roles in a variety of tumor types, for example, HOTAIR in breast cancer, SChLAP1 in aggressive prostate cancer, MALAT1 in lung cancer, and DRAIC in prostate cancer (11–13).

LINC00152 is an lncRNA that was first identified as being hypomethylated during hepatocellular carcinoma tumorigenesis (14). It is also dysregulated in gastric cancer and esophageal squamous cell carcinoma (15, 16). However, there are conflicting reports on exactly how LINC00152 functions to promote the invasive phenotype. One study has argued that LINC00152 directly interacts with EGFR and affects AKT signaling, whereas others have suggested that LINC00152 acts as a competing endogenous RNA (ceRNA) through titrating miRNAs (5, 6, 17–20). Recently, we identified LINC00152 through an in-depth genomic analysis of gliomas as being highly expressed in GBMs (21). In this study, we characterize LINC00152's association with GBM clinical features and with tumor cell invasion and begin to functionally characterize LINC00152 structurally. Furthermore, we find that LINC00152 is overexpressed in 10 other tumor types compared with matched normal tissue and high LINC00152 expression is associated with a poor prognosis in 7 of these tumors.

Cell culture, knockdown, and overexpression of LINC00152

U87 cells were maintained in MEM supplemented with 1% nonessential amino acids solution (catalog # 11140-050, Gibco), 1 mmol/L sodium pyruvate (catalog # 11360070, Gibco), 0.15% sodium bicarbonate (catalog # 25080094, Gibco), 10% FBS, and 1% penicillin/streptomycin.

For knockdown, U87 cells were transfected during two rounds of transfection. First, cells were reverse transfected with 40 nmol/L of siLINC00152_II (5′-UGACACACUUGAUCGAAUA-3′), siLINC00152_III (5′-CCGGAAUGCAGCUGAAAGA-3′), or a nonspecific siGL2 control siRNA (5′-CGUACGCGGAAUACUUCGA-3′) and 9 μL of Lipofectamine RNAiMAX transfection reagent (Thermo Fisher Scientific). Twenty-four hours later, a second round of transfection was performed using the same quantities of reagents. Twenty-four hours after the final transfection, cells were harvested and used for subsequent analysis.

A total of 500 ηg of LINC00152 or LINC00152-mutant pCDNA3-flag vectors was transfected into U87 cells using 2 μL of Lipofectamine 2000 (Thermo Fisher Scientific). Cells were harvested after 48 hours for downstream analysis.

RNA isolation, cDNA synthesis, qPCR, and Western blotting

Total RNA and nuclear/cytoplasmic RNAs were extracted using TRIzol total RNA isolation reagent (Thermo Fisher Scientific), Protein and RNA Isolation System (Thermo Fisher Scientific), respectively. RNA samples were treated with RQ1 RNase-Free DNase (Promega) according to the manufacturer's instructions. cDNA was produced from 1 μg RNA using Superscript III Kit (Thermo Fisher Scientific) according to the manufacturer's instructions. qPCR and Western blotting were performed according to standard protocols.

LINC00152 subcellular fractionation and in situ hybridization

LINC00152 subcellular fractionation was performed using Protein and RNA Isolation System (Thermo Fisher Scientific) according to the manufacturer's instructions.

For in situ hybridization, 3 × 105 U87 and U251 cells were plated on the top of a cover glass in a 6-well plate. On the next day, cells were washed once with PBS and fixed for 10 minutes with 2% paraformaldehyde. Then, cells were washed 3 times with PBS and incubated with 1 mL of permeabilization buffer (1× PBS/0.5% Triton X-100) for 10 minutes at 4°C. Cells were again washed 3 times with PBS. Next, cells were blocked with 1 mL of prehybridization buffer (3% BSA in 4× SSC) for 20 minutes at 55°C. Ten nanograms of LINC00152 or negative control probe was added to 2 mL of hybridization buffer (10% dextran sulfate in 4× SSC), and cells were incubated overnight at 55°C. On the next day, cells were washed 3 times for 5 minutes using washing buffer I (4× SSC, 0.1% Tween-20), 3 times for 5 minutes using washing buffer II (2× SSC), and 3 times for 5 minutes using washing buffer III (1× SSC). Subsequently, cells were blocked for 15 minutes at room temperature using 2 mL of blocking solution (4% BSA/1× PBS). Next, cells were incubated with 300 μL of antibody solution [2% of BSA/1 × PBS and digoxigenin (1:250)] for 1 hour. Cells were washed with 0.1% Tween-20/PBS 3 times and with alkaline Tris buffer for 5 minutes at room temperature. Finally, the signal was developed by adding 400 μL of BCP/NBT solution until the signal was visible.

MTT and Matrigel invasion assays

For measuring cell growth, 1,000 cells were plated in quadruplets in 96-well plates, and cell growth was measured using standard MTT reagent (Promega). To measure invasion, 2 × 105 U87 cells in serum-free media were seeded into 24-well Matrigel Invasion Chambers (BD Biosciences), and the bottom was filled with media and 10% FBS as the chemoattractant. Cells were allowed to invade for 8 hours and then fixed and stained with crystal violet/methanol and invaded cells were counted.

Expression of LINC00152 in TCGA datasets and survival analysis

The expression of LINC00152 in GBMs and low-grade glioma (LGG) compared with normal brain and tumor subtypes was performed as described previously (21). Expression of LINC00152 in all other The Cancer Genome Atlas (TCGA) tumors was determined by comparing expression data of only those tumors that had a matched normal tissue sample. Statistical significance was determined using a paired t test. TCGA patient survival data for GBMs and LGGs were retrieved from cBioPortal (www.cbioportal.org), and survival data for the remaining tumor types were retrieved from OncoLnc (www.oncolnc.org) in December 2016 (22–24). The expression threshold used to separate patients is outlined in the main text. Kaplan–Meier plots, HRs, and P values based on these separations were generated using the “survminer” package for R.

RNA-seq analysis

U87 cells were treated with a combination of the two siRNAs as mentioned earlier and total cell RNA was isolated using TRIzol and subsequently purified using RNeasy Isolation Kit (Qiagen). Sequencing libraries were generated using NEB NEXT Ultra Directional RNA Library Prep Kit and samplers were barcoded with NEBNext Multiplexing oligos per standard manufacturer protocols. Libraries were sequenced with 75-bp paired-end reads NextSeq500 instrument, in the Biomolecular Analysis Facility, University of Virginia School of Medicine. Sequencing reads were aligned to the hg38 reference genome using HISAT (25). Gene abundances and identification of differentially expressed genes were performed using HTSeq and DESeq2 (26, 27). An adjusted P value (obtained by DESeq2) cutoff of 0.05 and log2 fold change of 2 was used to define differentially expressed genes. GSEA analysis was performed on preranked gene list based on fold change (siLINC00152/siGL2) against 50 hallmark gene sets (28). For plotting enrichment score obtained by GSEA analysis (as shown in Fig. 4B), we have included only the genes that are either induced or repressed 1.5-folds upon siLINC00152. The raw and processed data were deposited in Gene Expression Omnibus (GEO) under accession number GSE111652.

LINC00152 structure predictions

Secondary structure predictions of LINC00152 were determined using mfold (29). The two structures with the lowest predicted free energies were selected for comparisons with psoralen analysis of RNA interactions and structures (PARIS) and Ribo-seq. For PARIS data analysis of LINC00152, raw sequencing data from Lu and colleagues were aligned to the hg19 genome using STAR (spliced transcripts alignment to a reference) with the alignment parameters outlines in Lu and colleagues' work (30, 31). Aligned reads were then processed to identify gapped mapping to LINC00152 and visualized with IGV (32). We used ribosome profiling data from Gonzalez and colleagues and aligned reads to the hg19 genome using HISAT2 (33). We then examined reads that mapped to LINC00152 for their distribution along the message to ensure that they were not legitimate ribosome footprints using IGV (32). The predicted secondary structure elements and protein-bound region were then compared with the in silico secondary structure predictions.

LINC00152 is an lncRNA overexpressed in aggressive gliomas

We first identified LINC00152 from a comprehensive analysis of lncRNAs in gliomas (21). LINC00152 was one of the most differentially expressed lncRNAs in GBMs compared with normal brain tissue; however, it is not upregulated in grade 2 and 3 gliomas (Fig. 1A; Supplementary Fig. S1A). We have validated the upregulation of LINC00152 in an independent set of patients with GBM compared with normal FFPE brain tissue (21).

Figure 1.

LINC00152 is upregulated in aggressive gliomas and in many cancer types. A, Boxplot of LINC00152 expression in normal brain tissue, G2 (grade 2 glioma), G3 (grade 3 glioma), and GBM. B, Boxplot of LINC00152 expression in LGG subtypes and normal brain tissue. C–L, Expression (RSEM) of LINC00152 in tumors and matched normal tissue from the TCGA in head and neck squamous carcinoma (HNSCC), renal papillary tumor (KIRP), hepatocellular carcinoma (LIHC), colorectal carcinoma (COADREAD), renal clear cell carcinoma (KIRC), breast-invasive carcinoma (BRCA), stomach adenocarcinoma (STAD), uterine carcinoma (UCEC), thyroid carcinoma (THCA), and lung adenocarcinoma (LUAD), respectively.

Figure 1.

LINC00152 is upregulated in aggressive gliomas and in many cancer types. A, Boxplot of LINC00152 expression in normal brain tissue, G2 (grade 2 glioma), G3 (grade 3 glioma), and GBM. B, Boxplot of LINC00152 expression in LGG subtypes and normal brain tissue. C–L, Expression (RSEM) of LINC00152 in tumors and matched normal tissue from the TCGA in head and neck squamous carcinoma (HNSCC), renal papillary tumor (KIRP), hepatocellular carcinoma (LIHC), colorectal carcinoma (COADREAD), renal clear cell carcinoma (KIRC), breast-invasive carcinoma (BRCA), stomach adenocarcinoma (STAD), uterine carcinoma (UCEC), thyroid carcinoma (THCA), and lung adenocarcinoma (LUAD), respectively.

Close modal

We tested whether LINC00152 is preferentially expressed in a particular GBM subtype, but that did not appear to be the case. The differences in LINC00152 expression between the subtypes were not statistically significant, although the median expression of LINC00152 is lowest in the proneural GBM subtype (P < 0.1; Supplementary Fig. S1B). Even though LINC00152 is not upregulated in LGGs as a whole, the IDHwt LGG subtype expresses 4 times as much LINC00152 as normal brains (P < 0.00001; Fig. 1B). This is interesting, because IDHwt LGGs are far more aggressive than the other LGG subtypes and display clinical properties similar to GBMs (34).

LINC00152 expression predicts survival in GBMs and LGGs

Because LINC00152 is upregulated in brain tumors compared with normal brain tissue, we next elucidated the association of LINC00152 association with survival of patients with GBM and LGG. To do this, we assessed the survival difference of patients expressing high (top 33% highest expressing LINC00152 cohort) and low level of LINC00152 expression from the TCGA for both GBM and LGG. In GBMs, patients who had high expression of LINC00152 had a poor prognosis (P = 0.02) compared with the patients expressing low level of LINC00152, with a median survival of 11.9 and 15.4 months, respectively (Fig. 2A). Furthermore, LINC00152 expression was also able to separate patients into two distinct prognostic groups in LGGs. LGG patients with high expression of LINC00152 had a median survival of 62.1 months, whereas the low expressing group had a median survival of 98.2 months (P < 0.0001; Fig. 2B). These results demonstrate that not only is LINC00152 overexpressed in gliomas, but that this overexpression is associated with poor patient outcome.

Figure 2.

High level of LINC00152 expression is associated with poor patient prognosis in GBMs, LGGs, and many other tumors. A, Kaplan–Meier plot of patients with GBM separated into the top 33% highest expressing LINC00152 cohort and lower expressing cohort. B, Kaplan–Meier plot of patients with LGG separated into the 33% highest expressing LINC00152 cohort and lower expressing cohort. C–F, Kaplan–Meier plots of the highest LINC00152-expressing quartile and lowest LINC00152-expressing quartiles for head and neck squamous carcinoma (HNSCC), lung adenocarcinoma (LUAD), renal clear cell carcinoma (KIRC), and hepatocellular carcinoma (LIHC), respectively. 95% CI, 95% confidence interval.

Figure 2.

High level of LINC00152 expression is associated with poor patient prognosis in GBMs, LGGs, and many other tumors. A, Kaplan–Meier plot of patients with GBM separated into the top 33% highest expressing LINC00152 cohort and lower expressing cohort. B, Kaplan–Meier plot of patients with LGG separated into the 33% highest expressing LINC00152 cohort and lower expressing cohort. C–F, Kaplan–Meier plots of the highest LINC00152-expressing quartile and lowest LINC00152-expressing quartiles for head and neck squamous carcinoma (HNSCC), lung adenocarcinoma (LUAD), renal clear cell carcinoma (KIRC), and hepatocellular carcinoma (LIHC), respectively. 95% CI, 95% confidence interval.

Close modal

LINC00152 in other cancers

It was intriguing to examine LINC00152 expression in other cancers compared with their respective normal tissues. We compared the expression of LINC00152 in all TCGA tumor samples with paired normal and tumor RNA sequencing (RNA-seq) data. Surprisingly, LINC00152 is upregulated in nearly every tumor type we analyzed, including head and neck squamous carcinoma, renal papillary tumor, hepatocellular carcinoma, colorectal carcinoma, renal clear cell carcinoma, breast-invasive carcinoma, stomach adenocarcinoma, uterine carcinoma, thyroid carcinoma, and lung adenocarcinoma (Fig. 1C–L).

Because LINC00152 is overexpressed in the majority of tumors that we have analyzed, we next wanted to determine whether LINC00152 expression is associated with patient survival in the TCGA tumors that had higher levels of LINC00152 compared with the paired normal samples. To do this, we performed Kaplan–Meier analysis for each tumor type by separating patients into two groups, the top quartile LINC00152-expressing tumors and the lowest quartile LINC00152-expressing tumors. From the original list of tumors, LINC00152 expression was associated with poor patient outcome in head and neck squamous cell carcinoma, lung adenocarcinoma, renal clear cell carcinoma, and hepatocellular carcinoma (Fig. 2C–F). The poor outcome of patients with renal papillary carcinoma was not statistically significant comparing the top and bottom quartiles of LINC00152 expression (P = 0.1), but the poor outcome was statistically significant (P = 0.015) when we compared patients in the top third and bottom third based on LINC00152 expression (Supplementary Fig. S1C).

Although LINC00152 was not overexpressed in LGGs relative to normal brain, it was upregulated in an aggressive subpopulation of LGGs (those with IDH wild type) and was associated with poor patient outcome. This made us realize that even if a tumor type does not overexpress LINC00152 globally relative to normal tissue, overexpression of the lncRNA in specific tumors may still be associated with poor outcome. We therefore examined other TCGA tumors that did not show a global increase of LINC00152 expression in the cancers relative to normal tissue for the predictive value of the expression of this lncRNA. Interestingly, even among these tumors, LINC00152 expression was associated with poor patient outcome in pancreatic adenocarcinoma when we compare the tumors in the top third and bottom third (Supplementary Fig. S1D), and acute myeloid leukemia, with the top quartile and bottom quartile for LINC00152 expression (Supplementary Fig. S1E). These results highlight the fact that in nine tumor types (GBMs, LGGs, head and neck squamous cell carcinoma, renal clear cell carcinoma, hepatocellular carcinoma, lung adenocarcinoma, renal papillary carcinoma, pancreatic adenocarcinoma, and acute myeloid leukemia), LINC00152 appears to function as unfavorable gene whose expression is associated with a poor patient outcome.

LINC00152 expression controls GBM cell invasion

Subcellular fractionation (Fig. 3A and B) and in situ hybridization (Fig. 3C) revealed that LINC00152 is primarily localized in the cytoplasm of U87 cells. We next sought to determine whether the upregulation of LINC00152 seen in GBMs is associated with any cancer phenotypes in GBM cell lines. LINC00152 has previously been shown to affect multiple cellular phenotypes, including cell growth, migration, invasion, and epithelial-to-mesenchymal transition (EMT; refs. 35, 36). We knocked down LINC00152 expression using two separate siRNAs or overexpressed the lncRNA and found that LINC00152 knockdown or overexpression did not affect cell proliferation for a period of 10 days (Supplementary Fig. S2B and S2D). We next assayed whether LINC00152 expression was associated with tumor cell invasion using a transwell migration assay. Knockdown of LINC00152 in U87 cell lines led to a statistically significant reduction in cell invasion with both siRNAs targeting LINC00152 (Fig. 3D and E). Conversely, overexpression of LINC00152 led to an increase of over 2-fold in the number of invaded cells (Fig. 3F and G). These findings suggest that LINC00152 knockdown decreases invasion of GBM cells, whereas upregulation in GBMs promotes the invasive phenotype that is commonly seen in patient tumors.

Figure 3.

LINC00152 is a cytoplasmic lncRNA that promotes cell invasion in U87 cells. A, Western blot analysis of Lamin A/C and actin, markers of the nucleus and cytoplasm, respectively. B, qRT-PCR of LINC00152 and a cytoplasmic RNA marker, GAPDH, and a nuclear RNA marker, MALAT1. C,In situ hybridization of LINC00152 in U87 and U251 cell lines; DRAIC lncRNA probes were used as negative control (“− control”). Purple color, positive signal. D, qRT-PCR showing knockdown of LINC00152 after treatment with two different siRNAs. E, Invasion assay with U87 cells after treatment with two different siRNAs against LINC00152; *, P < 0.05. Pictures were adjusted by −20% in brightness and +40% in contrast. F, qRT-PCR showing overexpression of LINC00152 after transient overexpression. G, Invasion assay with U87 cells overexpressing LINC00152; *, P < 0.05. Pictures were adjusted by +40% in contrast.

Figure 3.

LINC00152 is a cytoplasmic lncRNA that promotes cell invasion in U87 cells. A, Western blot analysis of Lamin A/C and actin, markers of the nucleus and cytoplasm, respectively. B, qRT-PCR of LINC00152 and a cytoplasmic RNA marker, GAPDH, and a nuclear RNA marker, MALAT1. C,In situ hybridization of LINC00152 in U87 and U251 cell lines; DRAIC lncRNA probes were used as negative control (“− control”). Purple color, positive signal. D, qRT-PCR showing knockdown of LINC00152 after treatment with two different siRNAs. E, Invasion assay with U87 cells after treatment with two different siRNAs against LINC00152; *, P < 0.05. Pictures were adjusted by −20% in brightness and +40% in contrast. F, qRT-PCR showing overexpression of LINC00152 after transient overexpression. G, Invasion assay with U87 cells overexpressing LINC00152; *, P < 0.05. Pictures were adjusted by +40% in contrast.

Close modal

LINC00152 knockdown decreases expression of proinvasive genes

To better understand how LINC00152 affects cellular invasion, we performed RNA-seq on U87 following knockdown of LINC00152 using a combination of two different siRNAs. Knockdown of LINC00152 leads to large changes in gene expression, with 259 genes significantly upregulated and 295 downregulated at least 2-fold (Fig. 4A). Thus, to determine the most significant molecular pathways regulated by LINC00152, we performed gene set enrichment analysis (GSEA), a method that can identify pathway enrichment from fold change–based preranked gene list from RNA-seq (28). This analysis showed a significant enrichment of upregulated genes upon siLINC00152 involved in EMT (Fig. 4B). Among the differentially expressed genes involved in EMT, the changes were validated by qPCR on 12 of 13 genes after siLIN00152 treatment (Supplementary Table S1). More interestingly, six of the genes that were downregulated by LINC00152 knockdown were conversely upregulated by overexpression of the lncRNA: TPM2 (Tropomyosin 2), PTX3 (Pentraxin 3), IGFBP4 (Insulin growth factor binding protein 4), TGM2 (Transglutaminase 2), SPP1 (Secreted phosphoprotein 1), and LUM (Lumican; Supplementary Table S1). Moreover, overexpression of the siRNA-resistant M8 was sufficient to upregulate these genes even after knockdown of endogenous LINC00152 (Supplementary Fig. S3). These results indicate that LINC00152 may induce U87 cell invasion by regulating the expression of at least these six genes.

Figure 4.

LINC00152 regulates genes involved in invasion in U87 cells. A, Volcano plot of statistical significance against fold change highlighting differentially regulated genes in black color upon siLINC00152 in U87 cells. B, Plot from GSEA showing the gene set involved in EMT enriched among upregulated genes (left black end of spectrum) after LINC00152 knockdown in U87 cells. C, Cumulative distribution frequency (CDF) plots of miRNA target mRNAs (as predicted by TargetScan: black line) or nontargets (gray line) showing fraction of genes with fold change less than that indicated on the x-axis after LINC00152 knockdown. None of the miRNAs previously proposed to be sponged by LINC00152 are released as evident from the fact that their targets are not repressed upon LINC00152 knockdown.

Figure 4.

LINC00152 regulates genes involved in invasion in U87 cells. A, Volcano plot of statistical significance against fold change highlighting differentially regulated genes in black color upon siLINC00152 in U87 cells. B, Plot from GSEA showing the gene set involved in EMT enriched among upregulated genes (left black end of spectrum) after LINC00152 knockdown in U87 cells. C, Cumulative distribution frequency (CDF) plots of miRNA target mRNAs (as predicted by TargetScan: black line) or nontargets (gray line) showing fraction of genes with fold change less than that indicated on the x-axis after LINC00152 knockdown. None of the miRNAs previously proposed to be sponged by LINC00152 are released as evident from the fact that their targets are not repressed upon LINC00152 knockdown.

Close modal

LINC00152 is not involved in sponging of miRNAs

Several previous studies have suggested that LINC00152 acts as an miRNA sponge by titrating different miRNAs (miR-376c-3p, miR-4775, miR-4767, miR-138-5p, miR-103, and miR-205) in different types of tumors, including GBMs (5, 6, 17–20). However, suggestions that an lncRNA acts as an miRNA sponge are sometimes questioned because the abundance of the lncRNA is often far less than that of the targets of the miRNAs and of the miRNAs themselves. If LINC00152 acts as an miRNA sponge in U87 cells, we would expect that the targets of these miRNAs would be repressed upon knockdown of the lncRNA and the subsequent release of the miRNAs from interaction with the lncRNA. However, we find that there is a statistically significant upregulation of the targets of these six miRNAs compared with nontargets when LINC00152 is knocked down, ruling out the possibility of LINC00152 acting as a ceRNA for these miRNAs (Fig. 4C).

Secondary structure components of LINC00152

Over the past decade, several new technologies have been developed to examine the secondary structures of lncRNAs on a global basis, one such technique is PARIS (30). PARIS is based on reversibly crosslinking RNA duplexes (stems of stem-loops) and gentle digestion with a single-strand RNase, S1 nuclease, to cut looped single-stranded portions of an RNA's secondary structure. The surviving RNA duplexes from the stems are then ligated to each other and subjected to high-throughput sequencing. RNAs containing stem-loops will have sequencing reads corresponding to the stems with gaps (corresponding to the loops) that do not overlap with a splice site. We analyzed publicly available PARIS data from HeLa cells to determine whether LINC00152 contains any secondary structure elements that could be detected by PARIS. Following alignment, we identified reads with a 2-nt gap that were present in the PARIS libraries (Supplementary Fig. S4C). These reads are positioned from position 285 to 373 of the 496 nt long LINC00152, with a small 2 base gap starting at position 342 (Fig. 5A). Sequence analysis of this region revealed some complementarity, suggesting that this region might in fact form a stem-loop structure (Fig. 5A).

Figure 5.

A 120-nucleotide hairpin at the 3′ end of LINC00152 (M8) is sufficient for promoting cell invasion in U87 cells. A, Predicted secondary structure of LINC00152 and the stem loop and protein-bound regions identified by PARIS (RNA Duplex) and Ribo-seq (Supplementary Fig. S4). B, Schematics of LINC00152 deletion mutants. C,LINC00152 qRT-PCR confirming overexpression levels of the different constructs. D, Invasion of U87 cells after overexpressing the different LINC00152 deletion mutants; *, P < 0.05. Pictures were adjusted by −10% in brightness and +40% in contrast. E, Invasion of U87 cells decreases after treatment with si00152_II but is rescued by LINC00152 m8 overexpression; *, P < 0.05. Pictures were adjusted by +20% in brightness and +40% in contrast.

Figure 5.

A 120-nucleotide hairpin at the 3′ end of LINC00152 (M8) is sufficient for promoting cell invasion in U87 cells. A, Predicted secondary structure of LINC00152 and the stem loop and protein-bound regions identified by PARIS (RNA Duplex) and Ribo-seq (Supplementary Fig. S4). B, Schematics of LINC00152 deletion mutants. C,LINC00152 qRT-PCR confirming overexpression levels of the different constructs. D, Invasion of U87 cells after overexpressing the different LINC00152 deletion mutants; *, P < 0.05. Pictures were adjusted by −10% in brightness and +40% in contrast. E, Invasion of U87 cells decreases after treatment with si00152_II but is rescued by LINC00152 m8 overexpression; *, P < 0.05. Pictures were adjusted by +20% in brightness and +40% in contrast.

Close modal

To get a better understanding of overall LINC00152 secondary structure, we used publicly available RNA secondary structure prediction tool, mfold, to identify secondary structure predictions for LINC00152 that are consistent with a stem-loop being present from 285 to 373 (29). The top 2 secondary structures with the lowest free energy differed in their exact base-pairing, but the overall stem-loop structure was largely the same. Importantly, both structures were consistent with a stem-loop being present from position 285 to 373 (Fig. 5A; Supplementary Fig. S4A and S4B). Furthermore, the resulting loop from the stem formation is rather small, 4 nt, which is consistent with the small 2-nt gap seen by PARIS.

We next asked whether we could use a separate method to independently validate the hairpin formation in LINC00152. Ribo-seq (ribosome profiling) is a technique that has been used to identify RNAs that interact with the ribosome and how the ribosome is distributed across those RNAs (37). This information has also been used to ascertain that some lncRNAs are associated with ribosomes, but not translating ribosomes (38). Recently, it was determined that the polysomes isolated for Ribo-seq are contaminated with other ribonucleoprotein (RBP) complexes. As a result, RNA footprints from RBPs that are not ribosome proteins can be detected in Ribo-seq data (33). To determine whether we could identify RBP–RNA footprints from LINC00152, we analyzed publicly available Ribo-seq data from normal brain samples (39). In two of the three normal brain Ribo-seq samples, we detected an RBP footprint at positions 303–330 of LINC00152. In addition, in one of the samples, there was an RBP footprint from 354 to 382 (Fig. 5A; Supplementary Fig. S4D). These two footprinted areas are located on opposite strands of the same stem-loop that was detected by PARIS, providing additional evidence of the existence of this stem-loop and suggesting that this stem is bound by a protein in an RBP (Fig. 5A).

LINC00152 stem-loop, M8, is sufficient to promote cell invasion

To determine whether this newly identified, potentially protein-bound, stem-loop plays a role in LINC00152 function, we created a series of LINC00152 deletion mutants (Fig. 5B; Supplementary Fig. S5A). The sites of the deletions were chosen based on PARIS and Ribo-seq analysis as well as two in silico predicted structures of LINC00152 (Fig. 5A; Supplementary Fig. S4C and S4D). We assessed whether independent overexpression of the mutants was able to stimulate U87 cell invasion. Overexpression of M2 (which removed the minimal amount of the protein bound stem-loop, nucleotides 280–401) or M3 (which removed the stem-loop and the remaining 3′ end) led to a decreased cell invasion significantly compared with full-length LINC00152 (P < 0.05; Fig. 5D). On the other hand, the mutant M4 (which removed the 3′ end but preserved the stem-loop) or M7 (which removed the extreme 3′ end, and also preserved the stem-loop) increased U87 cell invasion. Other deletion mutants that removed regions of LINC00152 5′ to the stem loop (M5 or M6) stimulated cellular invasion to a similar extent as full-length LINC00152. Finally, overexpression of M8, containing only the protein-bound stem-loop (nucleotides 280–401) was sufficient to stimulate invasion of U87 cells (Fig. 5D). These results suggest that M8 stem-loop is necessary and sufficient for stimulation of cell invasion.

Consistent with this conclusion, overexpression of the stem-loop also induced the six genes involved in EMT to the same extent as the full-length LINC00152 (Supplementary Table S1). In addition, knockdown of LINC00152 by siLINC00152_II (a siRNA that targets a region on LINC00152 outside of M8) decreased cell invasion, while the siRNA-resistant M8 was sufficient to rescue cell invasion (Fig. 5E).

The M8 stem-loop structure is important for stimulating invasion

We next tested with point mutations whether the ability to stimulate invasion of U87 cells depends on the LINC00152 stem-loop structure. Two mutants on opposite side of the stem disrupt the stem-loop (mutA: changes bases 333–336 and mutB: changes bases 349–352; Fig. 6D). Neither mutA nor mutB stimulated the invasion of U87 cells as well as full-length LINC00152 or M8 (Fig. 6A). In contrast, when the two mutations were combined in mutAB, the stem-loop structure was reconstructed and this promoted invasion to the same extent as full-length LINC00152 or M8 (Fig. 6A). Therefore, we can conclude that the stem-loop structure itself is essential for LINC00152 to stimulate cellular invasion.

Figure 6.

Point mutation of nucleotides 333-336 or 349-352 of LINC00152 shows the importance of M8 hairpin for stimulating invasion. A, Invasion of U87 cells after the different LINC00152 deletion mutants are overexpressed. Mut A or mut B are incapable of inducing invasion in U87 cells. Combining the two mutants (mut AB) restores the hairpin and induces invasion to the same level as full length LINC00152; *, P < 0.05. Pictures were adjusted by +20% in brightness and +40% in contrast. B, qRT-PCR confirming overexpression of the different LINC00152 constructs. C, Predicted secondary structure of full-length LINC00152 with the black line marking the sequence at the tip of the hairpin and the light gray and dark gray lines marking the residues that are mutated in Mut A or B, respectively. D, Predicted secondary structures of LINC00152 mutants A, B, and AB. The black, light gray, and dark gray lines mark the corresponding residues as in C.

Figure 6.

Point mutation of nucleotides 333-336 or 349-352 of LINC00152 shows the importance of M8 hairpin for stimulating invasion. A, Invasion of U87 cells after the different LINC00152 deletion mutants are overexpressed. Mut A or mut B are incapable of inducing invasion in U87 cells. Combining the two mutants (mut AB) restores the hairpin and induces invasion to the same level as full length LINC00152; *, P < 0.05. Pictures were adjusted by +20% in brightness and +40% in contrast. B, qRT-PCR confirming overexpression of the different LINC00152 constructs. C, Predicted secondary structure of full-length LINC00152 with the black line marking the sequence at the tip of the hairpin and the light gray and dark gray lines marking the residues that are mutated in Mut A or B, respectively. D, Predicted secondary structures of LINC00152 mutants A, B, and AB. The black, light gray, and dark gray lines mark the corresponding residues as in C.

Close modal

MIR4435-2HG, a homolog of LINC00152

As reported previously (40), LINC00152 is a close homolog of another lncRNA on chromosome 2, MIR4435-2HG; both have nearly identical sequences (with only 6 base mismatches) and both contain M8 sequence (Fig. 7A). LINC00152 and MIR4435-2HG are both transcribed from chromosome 2, LINC00152 is located at chr2: 87455476-87606739, and MIR4435-2HG is transcribed from chr2:111196350-111495115. In order to estimate the expression level of these two RNAs, we considered RNA-seq reads that uniquely mapped without any mismatch to either LINC00152 or MIR4435-2HG. This analysis showed that LINC00152 and MIR4435-2HG are expressed at the same level in U87 cells, and both of the RNAs are knocked down upon treatment of siLINC00152 to a similar extent (Fig. 7B and C). Thus, the phenotype that we observe with siRNA directed toward LINC00152 is also likely through knocking down the highly similar MIR4435-2HG. Moreover, given the high similarity between the two transcripts, and the fact that, from nucleotides 382 to 478, MIR4435-2HG forms a 97 nucleotides long stem-loop in the same position as LINC00152, it is likely that MIR4435-2HG overexpression phenocopies the effects of LINC00152 on cell invasion. However, we see an increase in cell invasion when we exogenously express LINC00152, saying that LINC00152 by itself can promote cell invasion. Again, upon considering uniquely mapped reads in TCGA RNA-seq data, we found that both LINC00152 and MIR4435-2HG are equally expressed in LGG and GBM (Fig. 7D). Analysis of TCGA RNA-seq data also revealed a positive correlation between the expression of the two RNAs in GBM and LGG (Fig. 7E and G), suggesting that these two RNAs may be coregulated. Moreover, the Kaplan–Meier plot to estimate survival showed that the expression of either RNA is associated with poor patient survival (Fig. 7G and H).

Figure 7.

LINC00152 is highly similar to the lncRNA MIR4435-2HG. A, Sequence alignment of LINC00152 and MIR4435-2HG. M8 is highlighted in the boxed area. B,LINC00152 specific RNA-seq reads in cells treated with siGL2 or siLINC00152. C, MIR4435-2HG–specific RNA-seq reads in cells treated with siGL2 or siLINC00152. D,LINC00152 or MIR4435-2HG–specific reads in TCGA RNA-seq data for LGG and GBM. E, Correlation of expression of LINC00152 and MIR4435-2HG in LGGs (spearman correlation 0.68 P < 2.2 e−16). F, Correlation of expression of LINC00152 and MIR4435-2HG in GBMs (Spearman correlation: 0.86; P < 2.2e−16). G, Kaplan–Meier plot of patients with LGG separated into the 50% highest expressing LINC00152 cohort and the lowest 50% expressing cohort. H, Kaplan–Meier plot of patients with LGG separated into the 50% highest expressing MIR4435-2HG cohort and the lowest 50% expressing cohort. 95% CI, 95% confidence interval.

Figure 7.

LINC00152 is highly similar to the lncRNA MIR4435-2HG. A, Sequence alignment of LINC00152 and MIR4435-2HG. M8 is highlighted in the boxed area. B,LINC00152 specific RNA-seq reads in cells treated with siGL2 or siLINC00152. C, MIR4435-2HG–specific RNA-seq reads in cells treated with siGL2 or siLINC00152. D,LINC00152 or MIR4435-2HG–specific reads in TCGA RNA-seq data for LGG and GBM. E, Correlation of expression of LINC00152 and MIR4435-2HG in LGGs (spearman correlation 0.68 P < 2.2 e−16). F, Correlation of expression of LINC00152 and MIR4435-2HG in GBMs (Spearman correlation: 0.86; P < 2.2e−16). G, Kaplan–Meier plot of patients with LGG separated into the 50% highest expressing LINC00152 cohort and the lowest 50% expressing cohort. H, Kaplan–Meier plot of patients with LGG separated into the 50% highest expressing MIR4435-2HG cohort and the lowest 50% expressing cohort. 95% CI, 95% confidence interval.

Close modal

The human genome was once thought to be mainly dormant and most of the transcription was devoted in producing protein-coding genes. We now know that the genome is transcriptionally vibrant and only a small fraction of the expressed genome, roughly 2%, encodes for protein-coding genes. GWAS and high-throughput sequencing studies have found that many of the genomic lesions and expression alterations seen in cancer and other pathologies fall within nonprotein-coding regions of the genome and may lead to dysregulation of ncRNAs (41–43). Furthermore, there is a growing body of evidence implicating lncRNAs in playing a direct role in normal cellular physiology, as well as driving pathogenesis in a variety of disorders, including cancer (43–46). Indeed, recent work has illustrated the critical role that lncRNAs play in cancer, including iconic examples such as HOTAIR in breast cancer and HULC in hepatocellular carcinoma and DRAIC in prostate cancer (11, 14, 47).

In this study, we have shown that LINC00152 is an lncRNA that is upregulated in many different cancer types and is highly upregulated in GBMs. Although LINC00152 is not upregulated in all LGGs relative to normal brain tissue, it is upregulated in the highly malignant IDHwt LGG subtype, further supporting LINC00152's association with aggressive tumors. This raised the interesting possibility that in tumors where LINC00152 is not differentially overexpressed or is moderately upregulated in the tumor population relative to normal tissue, LINC00152 could still be highly upregulated in a more aggressive subgroup of the tumors. This was indeed found to be true in pancreatic adenocarcinomas and acute myeloid leukemias. LINC00152 expression is associated with patient survival in nine different cancer types, including GBMs and LGGs. LINC00152 expression promotes cell invasion, which is consistent with its association with poor patient outcomes.

To assess the coding probability of LINC00152, we used the Coding Potential Assessment Tool (CPAT) and found a score of 0.0289, suggesting that this lncRNA has no coding potential, because a CPAT score of <0.5242 is considered noncoding (48). In addition, ExPasy (49) predicts the first LINC00152 ORF of 42 amino acids (126 nucleotides) from nucleotides 64 to 189. This ORF is not similar to any ORF predicted for the mouse transcript Gm14005. Moreover, blastx of the translated 126 nucleotide ORF did not show any hits in mouse protein database. In a different frame, there is a longer ORF of 92 amino acids (which also does not have a homolog in the mouse transcript) that is preceded by a short ORF that has three stop codons. So, the longer ORF is also unlikely to be translated.

Previous studies have shown that LINC00152 is an oncogenic lncRNA involved in regulating invasion in different types of tumors (5, 6, 18–20), including gliomas (5, 6). Yu and colleagues (5) have reported an in vivo tumor xenograft study; downregulation of LINC00152 produced smaller tumors and increased survival rates when compared with control. Thus, our findings reinforce the idea that LINC00152 is an oncogenic lncRNA that is associated with aggressive tumors by promoting cell invasion. Moreover, through analysis of global RNA structure mapping and RNA–protein interaction data, we identified a protein-bound stem-loop in the 3′ region of LINC00152. The structure–function analysis demonstrated that this stem-loop is necessary and sufficient for stimulating invasion of U87 cells, that it can rescue the loss of invasion seen after knockdown of LINC00152, and that the base-pairing of the opposite strands of the stem-loop, rather than the sequence at the mutated sites, is more important for stimulating invasion of U87 cells. However, it is likely that there are specific sequences along the M8 stem-loop that are important for LINC00152 function that will be examined in a future study.

GSEA of RNA-seq from LINC00152 knocked down cells also supports the idea that LINC00152 is involved in promoting invasion. More specifically, TPM2, PTX3, IGFBP4, TGM2, SPP1, and LUM were downregulated by siLINC00152 and upregulated after LINC00152 overexpression. Because we did not compare the global gene expression changes with LINC00152 overexpression and knockdown by RNA-seq, there are likely to be many other genes that will be regulated similarly to the six genes we tested in this study. Because siLINC00152 decreased invasion, we focused on genes in the RNA-seq data whose change (up or down) will decrease invasion. Of these, 13 transcripts qRT-PCR after siLINC00152 validated the changes in 12. Out of these 12, 6 genes were changed in the opposite direction when LINC00152 full length or M8 was overexpressed (Supplementary Table S1).

In addition, despite previous suggestions, analysis of RNA-seq showed us that LINC00152 is not acting as a ceRNA that sponges miRNAs.

LINC00152 is expressed from a syntenic location from mouse transcript. Mouse has a single gene, MIR4435-2HG (Gm14005 or MORRBID), but humans have two closely related genes, LINC00152 and MIR4435-2HG.

MIR4435-2HG is a host gene for an miRNA, as miR-4435 is transcribed from an intron of MIR4435-2HG. However, none of the effects observed by overexpression of LINC00152 are due to miR-4435, because the functional assays were done using the cDNA of LINC00152. Furthermore, miR-4435 is not detected in any of the short RNA-seq libraries (such as miRGator v3.0 and miRmine) from brain, glial cell lines, and gliomas.

Mouse MIR4435-2HG has been proposed to be a prosurvival lncRNA that represses a gene in cis, the proapoptic gene BCL2L11 (BIM) by recruiting the polycomb repressive complex, PRC2, to the BCL2L11 promoter (50). We considered the possibility that LINC00152, although cytoplasmic, is acting as a prooncogenic RNA by similarly suppressing BCL2L11. siLINC00152 increases BCL2L11 RNA (Supplementary Fig. S6B–S6D), but this is not expected to decrease cell invasion. Second, overexpression of LINC00152 from a heterologous site or the M8 hairpin of the lncRNA did not decrease BCL2L11 (Supplementary Fig. S6E–S6G) and yet increased cell invasion. Third, analyzing previously published mouse PAR-CLIP data, we determined that EZH2 from the polycomb complex associates with an intronic region of MORRBID (mouse MIR4435-2HG/MORRBID), which is not near the M8 region. Fourth mouse MIR4435-2HG encodes only the first third of the M8 hairpin that we have found to be functions in human LINC00152 or human MIR4435-2HG. Finally, LINC00152 is predominantly cytoplasmic, arguing against any role in recruiting any factors to the genome. Collectively, these results suggest that interaction with PRC2 is not necessary for the stimulation of invasion seen upon overexpression of the LINC00152 or the M8 hairpin RNA.

In conclusion, LINC00152/CYTOR and its homolog MIR4435-2HG functions as an oncogenic lncRNA in GBMs through the action of a protein-bound stem-loop and potentially plays a critical oncogenic role in a wide variety of cancer types. The results rule out a mechanism of action involving the sponging of miRNAs as proposed in the literature, or interaction with the Polycomb complex proposed for the mouse MIR4435-2HG/MORRBID RNA. LINC00152 could also serve as a tumor biomarker or a target for future cancer therapeutics.

No potential conflicts of interest were disclosed.

Conception and design: B.J. Reon, A. Dutta

Development of methodology: B.J. Reon, A. Dutta

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B.J. Reon, B. Takao Real Karia, A. Dutta

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B.J. Reon, B. Takao Real Karia, M. Kiran, A. Dutta

Writing, review, and/or revision of the manuscript: B.J. Reon, B. Takao Real Karia, M. Kiran, A. Dutta

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Kiran, A. Dutta

Study supervision: A. Dutta

The authors thank Dutta laboratory members for advice and helpful discussions. This work was supported by grants from the NIH R01 CA166054, AR067712, and a V foundation award D2018-002 to A. Dutta. B. Takao Real Karia was partly supported by CAPES BEX 0320-13-7. M. Kiran was partly supported by a DOD award PC151085.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ostrom
QT
,
Gittleman
H
,
Fulop
J
,
Liu
M
,
Blanda
R
,
Kromer
C
, et al
CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2008–2012
.
Neuro Oncol
2015
;
17
(
suppl 4
):
iv1
iv62
.
2.
Huse
JT
,
Holland
EC
. 
Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma
.
Nat Rev Cancer
2010
;
10
:
319
31
.
3.
Cancer Genome Atlas Research Network
. 
Comprehensive genomic characterization defines human glioblastoma genes and core pathways
.
Nature
2008
;
455
:
1061
8
.
4.
Brennan
CW
,
Verhaak
RGW
,
McKenna
A
,
Campos
B
,
Noushmehr
H
,
Salama
SR
, et al
The somatic genomic landscape of glioblastoma
.
Cell
2013
;
155
:
462
77
.
5.
Yu
M
,
Xue
Y
,
Zheng
J
,
Liu
X
,
Yu
H
,
Liu
L
, et al
Linc00152 promotes malignant progression of glioma stem cells by regulating miR-103a-3p/FEZF1/CDC25A pathway
.
Mol Cancer
2017
;
16
:
110
.
6.
Zhu
Z
,
Dai
J
,
Liao
Y
,
Ma
J
,
Zhou
W
. 
Knockdown of long noncoding RNA LINC0000125 suppresses cellular proliferation and invasion in glioma cells by regulating MiR-4775
.
Oncol Res
2017 Aug 11
[Epub ahead of print]
.
7.
Stupp
R
,
Hegi
ME
,
Mason
WP
,
van den Bent
MJ
,
Taphoorn
MJ
,
Janzer
RC
, et al
Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial
.
Lancet Oncol
2009
;
10
:
459
66
.
8.
ENCODE Project Consortium
. 
An integrated encyclopedia of DNA elements in the human genome
.
Nature
2012
;
489
:
57
74
.
9.
Kretz
M
,
Siprashvili
Z
,
Chu
C
,
Webster
DE
,
Zehnder
A
,
Qu
K
, et al
Control of somatic tissue differentiation by the long non-coding RNA TINCR
.
Nature
2013
;
493
:
231
5
.
10.
Gupta
RA
,
Shah
N
,
Wang
KC
,
Kim
J
,
Horlings
HM
,
Wong
DJ
, et al
Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis
.
Nature
2010
;
464
:
1071
6
.
11.
Prensner
JR
,
Iyer
MK
,
Sahu
A
,
Asangani
IA
,
Cao
Q
,
Patel
L
, et al
The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex
.
Nat Genet
2013
;
45
:
1392
8
.
12.
Gutschner
T
,
Hämmerle
M
,
Eissman
M
,
Hsu
J
,
Kim
Y
,
Hung
G
, et al
The non-coding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells
.
Cancer Res
2013
;
73
:
1180
9
.
13.
Sakurai
K
,
Reon
BJ
,
Anaya
J
,
Dutta
A
. 
The lncRNA DRAIC/PCAT29 locus constitutes a tumor-suppressive nexus
.
Mol Cancer Res
2015
;
13
:
828
38
.
14.
Neumann
O
,
Kesselmeier
M
,
Geffers
R
,
Pellegrino
R
,
Radlwimmer
B
,
Hoffmann
K
, et al
Methylome analysis and integrative profiling of human HCCs identify novel protumorigenic factors
.
Hepatology
2012
;
56
:
1817
27
.
15.
Cao
WJ
,
Wu
HL
,
He
BS
,
Zhang
YS
,
Zhang
ZY
. 
Analysis of long non-coding RNA expression profiles in gastric cancer
.
World J Gastroenterol
2013
;
19
:
3658
64
.
16.
Yang
S
,
Ning
Q
,
Zhang
G
,
Sun
H
,
Wang
Z
,
Li
Y
. 
Construction of differential mRNA-lncRNA crosstalk networks based on ceRNA hypothesis uncover key roles of lncRNAs implicated in esophageal squamous cell carcinoma
.
Oncotarget
2016
;
7
:
85728
40
.
17.
Zhang
YH
,
Fu
J
,
Zhang
ZJ
,
Ge
CC
,
Yi
Y
. 
LncRNA-LINC00152 down-regulated by miR-376c-3p restricts viability and promotes apoptosis of colorectal cancer cells
.
Am J Transl Res
2016
;
8
:
5286
97
.
18.
Teng
W
,
Qiu
C
,
He
Z
,
Wang
G
,
Xue
Y
,
Hui
X
. 
Linc00152 suppresses apoptosis and promotes migration by sponging miR-4767 in vascular endothelial cells
.
Oncotarget
2017
;
8
:
85014
23
.
19.
Cai
Q
,
Wang
Z
,
Wang
S
,
Weng
M
,
Zhou
D
,
Li
C
, et al
Long non-coding RNA LINC00152 promotes gallbladder cancer metastasis and epithelial-mesenchymal transition by regulating HIF-1α via miR-138
.
Open Biol
2017
;
7
:
pii:160247
.
20.
Wang
Y
,
Liu
J
,
Bai
H
,
Dang
Y
,
Lv
P
,
Wu
S
. 
Long intergenic non-coding RNA 00152 promotes renal cell carcinoma progression by epigenetically suppressing P16 and negatively regulates miR-205
.
Am J Cancer Res
2017
;
7
:
312
22
.
21.
Reon
BJ
,
Anaya
J
,
Zhang
Y
,
Mandell
J
,
Purow
B
,
Abounader
R
, et al
Expression of lncRNAs in low-grade gliomas and glioblastoma multiforme: an in silico analysis
.
PLoS Med
2016
;
13
:
e1002192
.
22.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
, et al
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.
23.
Gao
J
,
Aksoy
BA
,
Dogrusoz
U
,
Dresdner
G
,
Gross
B
,
Sumer
SO
, et al
Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal
.
Sci Signal
2013
;
6
:
pl1
.
24.
Anaya
J
. 
OncoLnc: linking TCGA survival data to mRNAs, miRNAs, and lncRNAs
.
PeerJ Comput Sci
2016
;
2
:
e67
.
25.
Kim
D
,
Langmead
B
,
Salzberg
SL
. 
HISAT: a fast spliced aligner with low memory requirements
.
Nat Meth
2015
;
12
:
357
60
26.
Anders
S
,
Pyl
PT
,
Huber
W
. 
HTSeq—a Python framework to work with high-throughput sequencing data
.
Bioinformatics
2014
;
31
:
166
9
.
27.
Love
MI
,
Huber
W
,
Anders
S
. 
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
.
28.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
29.
Zuker
M
. 
Mfold web server for nucleic acid folding and hybridization prediction
.
Nucleic Acids Res
2003
;
31
:
3406
15
.
30.
Lu
Z
,
Zhang
QC
,
Lee
B
,
Flynn
RA
,
Smith
MA
,
Robinson
JT
, et al
RNA duplex map in living cells reveals higher-order transcriptome structure
.
Cell
2016
;
165
:
1267
79
.
31.
Dobin
A
,
Davis
CA
,
Schlesinger
F
,
Drenkow
J
,
Zaleski
C
,
Jha
S
, et al
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
2013
;
29
:
15
21
.
32.
Robinson
JT
,
Thorvaldsdottir
H
,
Winckler
W
,
Guttman
M
,
Lander
ES
,
Getz
G
, et al
Integrative genomics viewer
.
Nat Biotech
2011
;
29
:
24
6
.
33.
Ji
Z
,
Song
R
,
Huang
H
,
Regev
A
,
Struhl
K
. 
Transcriptome-scale RNase-footprinting of RNA-protein complexes
.
Nat Biotech
2016
;
34
:
410
3
.
34.
Cancer Genome Atlas Research Network. 
Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas
.
N Engl J Med
2015
;
372
:
2481
98
.
35.
Ji
J
,
Tang
J
,
Deng
L
,
Xie
Y
,
Jiang
R
,
Li
G
, et al
LINC00152 promotes proliferation in hepatocellular carcinoma by targeting EpCAM via the mTOR signaling pathway
.
Oncotarget
2015
;
6
:
42813
24
.
36.
Zhao
J
,
Liu
Y
,
Zhang
W
,
Zhou
Z
,
Wu
J
,
Cui
P
, et al
Long non-coding RNA Linc00152 is involved in cell cycle arrest, apoptosis, epithelial to mesenchymal transition, cell migration and invasion in gastric cancer
.
Cell Cycle
2015
;
14
:
3112
23
.
37.
Ingolia
NT
,
Ghaemmaghami
S
,
Newman
JR
,
Weissman
JS
. 
Genome-wide analysis in vivo of translation with nucleotide resolution using ribosome profiling
.
Science
2009
;
324
:
218
23
.
38.
Guttman
M
,
Russell
P
,
Ingolia
NT
,
Weissman
JS
,
Lander
ES
. 
Ribosome profiling provides evidence that large noncoding RNAs do not encode proteins
.
Cell
2013
;
154
:
240
51
.
39.
Gonzalez
C
,
Sims
JS
,
Hornstein
N
,
Mela
A
,
Garcia
F
,
Lei
L
, et al
Ribosome profiling reveals a cell-type-specific translational landscape in brain tumors
.
J Neurosci
2014
;
34
:
10924
36
.
40.
Nötzold
L
,
Frank
L
,
Gandhi
M
,
Polycarpou-Schwarz
M
,
Groß
M
,
Gunkel
M
, et al
The long non-coding RNA LINC00152 is essential for cell cycle progression through mitosis in HeLa cells
.
Sci Rep
2017
;
7
:
2265
.
41.
Mirza
AH
,
Kaur
S
,
Brorsson
CA
,
Pociot
F
. 
Effects of GWAS-associated genetic variants on lncRNAs within IBD and T1D candidate loci
.
PLoS One
2014
;
9
:
e105723
.
42.
Huarte
M
. 
The emerging role of lncRNAs in cancer
.
Nat Med
2015
;
21
:
1253
61
.
43.
Iyer
MK
,
Niknafs
YS
,
Malik
R
,
Singhal
U
,
Sahu
A
,
Hosono
Y
, et al
The landscape of long noncoding RNAs in the human transcriptome
.
Nat Genet
2015
;
47
:
199
208
.
44.
Grote
P
,
Wittler
L
,
Währisch
S
,
Hendrix
D
,
Beisaw
A
,
Macura
K
, et al
The tissue-specific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse
.
Dev Cell
2013
;
24
:
206
14
.
45.
Flockhart
RJ
,
Webster
DE
,
Qu
K
,
Mascarenhas
N
,
Kovalski
J
,
Kretz
M
, et al
BRAFV600E remodels the melanocyte transcriptome and induces BANCR to regulate melanoma cell migration
.
Genome Res
2012
;
22
:
1006
14
.
46.
Van Grembergen
O
,
Bizet
M
,
de Bony
EJ
,
Calonne
E
,
Putmans
P
,
Brohée
S
, et al
Portraying breast cancers with long noncoding RNAs
.
Sci Adv
2016
;
2
:
e1600220
.
47.
Panzitt
K
,
Tschernatsch
MM
,
Guelly
C
,
Moustafa
T
,
Stradner
M
,
Strohmaier
HM
, et al
Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA
.
Gastroenterology
2007
;
132
:
330
42
.
48.
Wang
L
,
Park
HJ
,
Dasari
S
,
Wang
S
,
Kocher
JP
,
Li
W
. 
CPAT: Coding-Potential Assessment Tool using an alignment-free logistic regression model
.
Nucleic Acids Res
2013
;
41
:
e74
.
49.
Gasteiger
E
,
Gattiker
A
,
Hoogland
C
,
Ivanyi
I
,
Appel
RD
,
Bairoch
A
. 
ExPASy: The proteomics server for in-depth protein knowledge and analysis
.
Nucleic Acids Res
2003
;
31
:
3784
8
.
50.
Kotzin
JJ
,
Spencer
SP
,
McCright
SJ
,
Kumar
DBU
,
Collet
MA
,
Mowel
WK
, et al
The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan
.
Nature
2016
;
537
:
239
243
.

Supplementary data