Genome‑wide analysis reveals the emerging roles of long non‑coding RNAs in cancer (Review)

  • Authors:
    • Xiaoxia Ren
  • View Affiliations

  • Published online on: November 22, 2019     https://doi.org/10.3892/ol.2019.11141
  • Pages: 588-594
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer is the most intractable human disease that is primarily caused by genetic alterations. Recently, the general application of microarrays and high‑throughput sequencing technology has revealed various important roles of long noncoding RNAs (lncRNAs) in cancer. This review summarizes the function, mechanism, diagnostic and treatment potential of lncRNAs identified through genome‑wide analysis in cancer. Cell‑, tissue‑ and development stage‑specific expression patterns are major characteristics of cancer‑associated lncRNAs, and various genetic alterations are also implicated. Microarray and sequencing analyses serve important roles in mechanistic studies of either nuclear or cytoplasmic lncRNAs. Collectively, genome‑wide analysis is the inexorable trend of future studies or clinical applications of lncRNAs and offers a novel perspective regarding the prognosis and treatment of cancer.

Introduction

Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nucleotides that lack protein-coding potential. Due to the simultaneous and synergistic availability of genome and transcriptome sequences, lncRNAs were discovered by analyzing the mouse transcriptome based on the functional annotation of 60,770 full-length cDNAs in 2002 (1). Djebali et al (2) have also used sequencing technology to reveal that <2% of the human genome encodes proteins, and three-quarters of the human genome is actively transcribed into non-coding RNAs. LncRNAs are highly conserved (>95%) in mammals (3), indicating their biological functionality. However, for a long time, a large number of ncRNAs have been regard as dark matter or transcriptional noise.

In recent years, the general application of genome-wide analysis, which includes microarrays and high-throughput sequencing technologies, have revealed various important biological functions of lncRNAs in cell differentiation, development and numerous diseases, especially cancer (4). In cancer, lncRNAs exhibit tissue-specific expression and are transcriptionally regulated by key tumor suppressors or oncogenes, which can influence cell cycle regulation, survival, immune response and pluripotency (5). For example, lncRNA taurine upregulated gene 1 (TUG1) is a direct transcriptional target of p53 and affects cell proliferation in human non-small cell lung and laryngeal cancer (6). Likewise, lncRNA plasmacytoma variant translocation 1 (PVT1) is adjacent to proto-oncogene Myc in the same genomic region of 8q24, and its amplification is associated with the Myc gene copy number gain, thus increasing Myc protein levels in cancer (7).

Although the above examples represent only a small proportion of lncRNA effects, they indicate the large diversity in the functions of lncRNAs in cancer. Based on these mechanisms, lncRNAs are attractive potential therapeutic targets and biomarkers of cancer, and genome-wide analysis in combination with novel computational strategies may further advance the clinical application of lncRNAs. In this review, the emerging roles of lncRNAs in cancer based on genome-wide analysis and the potential clinical applications of lncRNAs were summarized.

Identification of lncRNAs in cancer using genome analysis

Aberrant expression of lncRNAs in cancer

LncRNAs exhibit tissue-specific expression, and their expression level consistently changes in cancer (8). Considering their limitations in large-sample tests and tedious operation protocols, traditional molecular methods such as reverse transcription-PCR and fluorescence in situ hybridization do not suffice for the study of lncRNAs (9). The general implementation of microarrays (Table I) and high-throughput sequencing (Table II) technology overcomes these difficulties and makes great progress in cancer. To delineate genome-wide lncRNA expression patterns, polyA+ RNA sequencing (RNA-seq) data from 6,503 samples, including tumors, normal samples and cancer cell lines, were applied (10). Based on these data, a non-parametric method termed Sample Set Enrichment Analysis was developed to assess differential expression. Using this approach, known lncRNAs in breast and prostate cancer, including upregulated oncogenic lncRNA hox transcript antisense intergenic RNA (HOTAIR) and prostate cancer antigen-3 (PCA3), downregulated tumor suppressor lncRNA maternally expressed gene 3 (MEG3) were identified, and a myriad of unannotated lncRNAs exhibiting potential tissue- and cancer-specific gene signatures were also identified (Fig. 1A) (10).

Table I.

Typical cancer-associated lncRNAs identified by microarray.

Table I.

Typical cancer-associated lncRNAs identified by microarray.

Author, yearLncRNACancer typeBiological functionMechanism(Refs.)
Rinn et al, 2007; Botti et al, 2017HOTAIRBreast, gastric, colorectal and cervicalPromotes metastasisActs as scaffold for the chromatin repressors PRC2 and LSD1. Silences HoxD and other gene loci(11,12)
He et al, 2017MEG3HepatocellularTumor suppressor; inhibits cell growth and induces apoptosismiR-29 regulates expression of MEG3 in methylation-dependent pattern(13)
Tee et al, 2016MALAT1Neuroblastoma, bladder, lung and gastricPromotes cell proliferation and metastasisPromotes tumor-driven angiogenesis by upregulating pro-angiogenic gene expression(31)
Ji et al, 2015; Zhang et al, 2017LINC00152Hepatocellular and lungPromotes proliferation, invasion, metastasis and apoptosisAffects mechanistic targets of mTOR, Akt and EGFR pathways(33,35)
Zhu et al, 2016LOC572558BladderTumor suppressor; inhibits proliferation and induces apoptosisRegulates the p53 signaling pathway(37)
Prensner et al, 2014; Smolle et al, 2017SCHLAP1ProstateBiomarker for metastaticUnknown progression(54,55)
Li et al, 2016GAS5BreastPrognostic marker; suppresses cancer proliferationActs as a molecular sponge for miR-21, leading to the de-repression of phosphatase and tensin homologs(57)

[i] LncRNAs, long non-coding RNAs; HOTAIR, hox transcript antisense intergenic RNA; PRC2, polycomb repressive complex 2; LSD1, histone demethylase lysine-specific demethylase; MEG3, maternally expressed gene 3; miR, microRNA; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; mTOR, mechanistic target of rapamycin; EGFR, epidermal growth factor receptor; SChLAP1, second chromosome locus associated with prostate-1; GAS5, growth arrest-specific transcript 5.

Table II.

Typical cancer-associated lncRNAs identified by high-throughput sequencing.

Table II.

Typical cancer-associated lncRNAs identified by high-throughput sequencing.

Author, yearLncRNACancer typeBiological functionMechanism(Refs.)
Tseng et al, 2014PVT1Breast, ovarian and hepatocellularPromotes proliferationControls Myc levels by regulating protein stability(7)
Russell et al, 2015CASC15Neuroblastoma and metastatic melanomaTumor suppressor; inhibits proliferation and migratoryAs a mediator of neural growth and differentiation to impact initiation and progression(17)
Pan et al, 2016GAS8-AS1Papillary thyroidTumor suppressor; inhibits cancer growthUnknown(22)
Liu et al, 2015SIRT1-ASHepatocellularPromotes proliferationBinds to SIRT1 mRNA at 3′UTR, masks the microRNA-29c binding site and stabilizes SIRT1 mRNA(23)
Sánchez et al, 2014PR-lncRNA-1ColorectalTumor suppressor; inhibits cell survival and proliferationModulates gene expression response to DNA damage downstream of p53(41)
Hünten et al, 2015; Kaller et al, 2017LINC01021ColorectalInhibits proliferationA novel direct p53 target gene(42,43)
Merry et al, 2015; Somasundaram et al, 2018DACOR1ColonGrowth suppressorChanges DNA methylation patterns without affecting DNMT1 protein levels(47,48)
Kondo et al, 2017JHDM1D-AS1PancreaticPromotes tumorigenesisRegulates angiogenesis in response to nutrient starvation(49)
Hu et al, 2019LINK-ABreastDownregulates cancer antigenicityInactivation of protein kinase A pathways(50)

[i] lncRNAs, long non-coding RNAs; PVT1, plasmacytoma variant translocation 1; CASC15, cancer-associated susceptibility candidate 15; GAS8-AS1, GAS8 antisense RNA 1; SIRT1-AS, sirtuin 1-antisense; SIRT1, sirtuin 1; 3′UTR, 3′-untranslated region; PR-lncRNA-1, p53-regulated lncRNA-1; DACOR1, DNMT1-associated colon cancer repressed lncRNA 1; JHDM1D-AS1, JHDM1D antisense 1; LINK-A, lncRNA long intergenic non-coding RNA for kinase activation.

Rinn et al (11) have reported that lncRNA HOTAIR is a spliced and polyadenylated RNA with 2,158 nucleotides and 6 exons, according to a DNA microarray. Patients with high HOTAIR expression levels exhibit an increased incidence of malignancy and lymph node metastasis (12). Another lncRNA, MEG3, which is expressed in normal tissues, has been demonstrated to be either lost or decreased in a number of human tumors and tumor-derived cell lines; MEG3 is a tumor suppressor involved in the etiology, progression and chemosensitivity of cancers (13). In addition to well-known lncRNAs, a growing number of novel lncRNAs have been identified, such as adenocarcinoma- and immune-associated lncRNAs. A lncRNA expression profile in lung adenocarcinoma and matched adjacent normal lung tissues measured by microarray demonstrated that 1,048 lncRNAs were upregulated and 1,997 lncRNAs were downregulated in lung adenocarcinoma (14). Among these lncRNAs, nine lncRNAs were validated; the expression of NONHSAT077036 was associated with N classification and clinical stage (14). Wang et al (15) also used the Chinese Glioma Genome Atlas microarray to identify 344 immune-associated lncRNAs. Further validation indicated that the nine immune-associated lncRNA signature, including SNHG8, ST20-AS1, PGM5-AS1, LINC00937, MIR155HG, AGAP2-AS1, MAPKAPK5-AS1, HCG18 and TUG1, exhibited prognostic value for anaplastic gliomas.

Genetic alteration of lncRNAs in cancer

Microarrays and sequencing technologies, especially whole genome sequencing, have been prominently applied to identify the genetic alterations of lncRNAs in cancer, including single nucleotide polymorphisms (SNPs), copy number alterations (CNAs) and point mutations (16). For example, a common disease genome-wide association study identified a SNP within a tumor suppressor lncRNA cancer-associated susceptibility candidate 15 (CASC15) at 6p22, which was associated with CASC15-S differential expression (17).

CBioPortal (http://www.cbioportal.org/) is an open-access resource for the interactive examination of multidimensional cancer genomics data sets that include information regarding somatic mutations, CNAs, RNA expression, DNA methylation and protein and phosphoprotein abundance (18,19). CBioPortal provides access to data from >5,000 tumor samples from 105 cancer studies in The Cancer Genome Atlas (TCGA) (20). A previous study assessed 1,000 cases of breast invasive carcinoma in this database; the results demonstrated that 577 lncRNAs exhibited alterations among the 2,730 analyzed lncRNAs (21). The deregulation of 11 lncRNAs, primarily due to CNA, was associated with poor overall survival rate; the results also suggested that greater distance from the oncogene Myc was associated with smaller alterations of CNA, which indicated that 8q24.21 was the center of CAN (21). Another team also supports this conclusion; Tseng et al (7) analyzed genome-wide data from two large cancer databases, including Progenetix and TCGA, and consistently observed in 98% of cases a co-gain of Myc and PVT1 across a wide variety of cancer types harboring the amplified 8q24 region, such as ovarian, oesophageal and breast cancer. According to their study, PVT1 RNA and Myc protein expression were associated in primary human tumors, and the copy number of PVT1 was co-increased in >98% of cancer cases with the increase of Myc copy numbers.

Pan et al (22) identified somatic mutations for papillary thyroid carcinoma (PTC) in the Chinese population using 402 pairs of tumor and normal tissues, of which 91 were characterized by exome sequencing and 311 by Sanger sequencing; the results demonstrated that the lncRNA GAS8-AS1 was the secondary most frequently altered gene and acted as a novel tumor suppressor in PTC. Liu et al (23) identified a single-nucleotide mutation (622U>C) in the SIRT1-AS sequence using gene-sequencing technology; bioinformatics analysis revealed that this mutation led to an alteration in the secondary structure of lncRNA SIRT1-AS and prevented it from binding to SIRT1 mRNA.

Mechanisms of lncRNAs identified by genome-wide analyses in cancer

The mechanisms of lncRNAs likely depend on their secondary or tertiary structures (24). Nuclear lncRNAs may act as molecular scaffolds and aid in alternative splicing or chromatin structure modification (25). Cytoplasmic lncRNAs may modulate translation and promote or inhibit mRNA degradation, acting as miRNA sponges (25,26). In addition to identifying novel lncRNAs, genome-wide analysis also serves important roles in lncRNA mechanistic studies, such as to determine its functional importance or to probe its mechanistic properties (27). Recently, these techniques were combined with approaches used to study gene-gene, gene-protein and protein-protein interactions, including chromatin immunoprecipitation (ChIP), RNA binding protein immunoprecipitation (RIP) and co-immunoprecipitation (Co-IP), to further assess the mechanistic role of lncRNAs in cancer (Fig. 1B) (4).

The mechanisms of lncRNAs in cancer identified by microarrays

Microarrays involve hybridizing a modified complementary DNA (cDNA) or a complementary RNA template of target transcripts to a set of short oligonucleotide probes immobilized on a solid surface (28). When bound to the probes, these modifications enable either direct or indirect florescence detection, and a digital signal is used to quantify the level of gene expression (28). Therefore, microarray offers a highly sensitive, highly selective and high-throughput method for the analysis of mRNAs and lncRNAs in biology and disease (29).

The intergenic lncRNA MALAT1, was originally identified as a prognostic marker of lung cancer metastasis, and was associated with metastasis in other cancer types, such as breast, prostate, and pancreatic cancer (30). Microarray-based differential gene expression analysis has revealed that FGF2 is significantly downregulated by MALAT1, and that MALAT1-mediated FGF2 protein secretion from neuroblastoma cells induces vasculature formation to promote tumor angiogenesis (31). In addition, LINC00152 is involved in the pathogenesis of cancer; the detailed mechanism was unknown until microarray-based analysis was performed (32). LINC00152 can affect the mammalian target of rapamycin, phosphatidylinositol 3-kinase/AKT and the epidermal growth factor receptor signaling pathways as a molecular indicator that assists in the regulation of the associated target gene expression (3336). Furthermore, using a high-throughput phospho-proteome microarray, Zhu et al (37) have demonstrated that Akt, mouse double minute 2 homolog and p53 phosphorylation are reduced when lncRNA LOC572558 is stably upregulated, suggesting that LOC572558 may be a tumor suppressor and regulate the p53 signaling pathway in bladder cancer.

The mechanisms of lncRNAs in cancer identified by sequencing

Sequencing involves the generation of cDNA-containing primers by PCR for library construction and analysis (38). The library is subsequently sequenced and quantified for each oligonucleotide base (38). The advantages of sequencing include a direct measure of the genome and its ability to discover novel transcripts, alternate transcript starts and stops, splicing variants and single nucleotide polymorphisms (39). Compared with microarray, sequencing offers more information when combined with functional experiments in mechanistic studies of lncRNAs (9).

A number of studies have demonstrated the association between endogenous p53 activity and the expression of lncRNAs in cancer progression (4043). By integrating RNA-seq with p53 ChIP-seq analyses of HCT116 cells under DNA damage, two lncRNAs, PR-lncRNA-1 and PR-lncRNA-10, were identified (41). These lncRNAs are required for the efficient binding of p53 to its target genes, modulating p53 activity at the transcriptional, but not the post-transcriptional level, and contributing to apoptotic induction in response to DNA damage in cancer (41). The effect of p53 activation and genome-wide DNA binding of p53 was also determined by RNA-seq in combination with ChIP-seq analysis in SW480 cells (42). A total of 393 lncRNAs, including 270 upregulated and 123 downregulated lncRNAs, were identified as differentially regulated by p53. In addition, 18 lncRNAs demonstrating p53 binding near the corresponding gene TSS and 17 lncRNAs displaying p53 chromatin occupancy in a 20-kbp region surrounding the corresponding gene TSS were identified (42). LINC01021 has been identified as a novel direct p53 target gene, the ectopic expression of which inhibits colorectal cancer cell proliferation and chemosensitivity (42,43).

A number of lncRNAs affect gene expression though interacting with epigenetic processes (25). Enhancer of zeste homolog 2 (EZH2), the catalytic component of polycomb repressive complex 2 (PRC2), possesses histone-lysine N-methyltransferase activity and plays an essential role in cancer (44). RIP-seq has demonstrated that the lncRNA MALAT1 binds with EZH2 to suppresses the tumor suppressor PCDH10 and promote gastric cellular migration and invasion (45). In addition to histone methylation, DNA methylation is another crucial epigenetic marker typically associated with repressed genes in human cells (46). Merry et al (47) identified a subset of lncRNAs that interacted with DNMT1 in HCT116 cells though RIP-seq. DNMT1-associated colon cancer-repressed lncRNA 1 (DACOR1) exhibited high tissue-specific expression in the normal colon tissue, but was repressed in a panel of colon tumors and patient-derived colon cancer cell lines (47). The genomic occupancy sites of DACOR1 significantly overlap with known differentially methylated regions, and dysregulation of DACOR1 contributes to aberrant DNA methylation patterns in colon tumors (47,48).

LncRNAs also serve pivotal roles in tumor progression and malignant transformation by regulating the nutrient-starved tumor microenvironment or immune checkpoints. Kondo et al (49) identified the nutrient starvation-responsive lncRNA JHDM1D antisense 1 using ChIP-seq and formaldehyde-assisted isolation of regulatory element sequencing, which was demonstrated to be upregulated and to promote tumorigenesis by regulating angiogenesis in response to nutrient starvation. In addition, Hu et al (50) reported that the oncogenic lncRNA long intergenic non-coding RNA for kinase activation (LINK-A) inactivated tumor suppressor pathways and downregulated antigen presentation through inactivation of protein kinase A pathways, thus contributing to decreased immunosurveillance and escaping from immune checkpoints by regulating immune cell gene expression programs. These results provided the basis for developing combination immunotherapy treatment regimens.

Cancer diagnostics and therapies based on genome-wide analysis of lncRNAs

The emerging role of lncRNAs, especially their highly tissue- and developmental stage-specific expression, provides a new range of possibilities for the diagnosis and treatment of cancer. The lncRNAs may serve as diagnostic biomarkers detected in the plasma, urine or tissue or contained in exosomes, microvesicles or apoptotic bodies and as treatment targets for drugs or radiotherapy (Fig. 1C) (51). Based on genome-wide analysis, the prognostic values of various lncRNAs for patients with cancer were evaluated, and the reported number of lncRNAs with clinical implications has increased (52).

The first lncRNA to be approved by the Food and Drug Administration was PCA3, which was used as a urine biomarker in patients with prostate cancer (53). A large-scale high-throughput study was performed using a microarray of prostate tumors from patients who experienced metastasis (n=212) and those who did not (n=333), and the lncRNA SCHLAP1 was discovered as one of the optimal predictive genes for metastasis (54). This was further validated using a clinical-grade urine test in a Clinical Laboratory Improvement Amendments-certified laboratory; SCHLAP1 has been suggested to be a promising biomarker of the aggressive clinical course of prostate cancer (55). Therapeutic resistance to trastuzumab caused by dysregulation of lncRNAs often poses a major obstacle in the clinical management of HER2-positive breast cancer (56). In addition, microarray results indicated that the expression of the lncRNA GAS5 was decreased in SKBR-3/Tr cells and breast cancer tissues from trastuzumab-treated patients, suggesting that GAS5 may serve as a novel prognostic marker and candidate drug target for HER2-positive breast cancer (57). Collectively, the prognostic values of these lncRNAs for patients with cancer were assessed by meta-analysis, which may facilitate improvements in the diagnosis and prognosis of cancer treatment (5860). Although the majority of pioneering studies remain in the early stage, they still encourage further examination of the diagnostics and therapeutic opportunities of lncRNAs in cancer.

Conclusion and future perspective

The development of genome-wide analysis has resulted in a tremendous advance in the understanding of the underlying mechanisms of lncRNAs. The extensive functions and mechanisms of lncRNAs in cancers are a hot area of research. This review provides an overview of lncRNAs that were identified by microarray and sequencing in cancer, including their functions and mechanisms, as well as their potential use in cancer diagnosis and therapies. However, this review also provides information that remains far from the realistic clinical application of lncRNAs in the treatment of cancer. The techniques that detect lncRNA interactions with key genes and proteins, the use of dependable animal models for cancer-associated lncRNAs and the implementation of reliable computational calculation methods of genome-wide analysis should be developed for future functional and mechanistic studies of lncRNAs. In addition, large clinical trials should be performed to confirm these potential lncRNA biomarkers. Furthermore, genome-wide analyses, which reduce analytic time and cost, will be indispensable in future studies and in clinical applications of lncRNAs in cancer.

Acknowledgments

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

XXR conceived the review, researched the literature and wrote the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The author declares that they have no competing interests.

References

1 

Okazaki Y, Furuno M, Kasukawa T, Adachi J, Bono H, Kondo S, Nikaido I, Osato N, Saito R, Suzuki H, et al: Analysis of the mouse transcriptome based on functional annotation of 60,770 full-length cDNAs. Nature. 420:563–573. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Jathar S, Kumar V, Srivastava J and Tripathi V: Technological developments in lncRNA biology. Adv Exp Med Biol. 1008:283–323. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Zheng GX, Do BT, Webster DE, Khavari PA and Chang HY: Dicer-microRNA-Myc circuit promotes transcription of hundreds of long noncoding RNAs. Nat Struct Mol Biol. 21:585–590. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Zhang Z, Wang X, Cao S, Han X, Wang Z, Zhao X, Liu X, Li G, Pan X and Lei D: The long noncoding RNA TUG1 promotes laryngeal cancer proliferation and migration. Cell Physiol Biochem. 49:2511–2520. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Tseng YY, Moriarity BS, Gong W, Akiyama R, Tiwari A, Kawakami H, Ronning P, Reuland B, Guenther K, Beadnell TC, et al: PVT1 dependence in cancer with MYC copy-number increase. Nature. 512:82–86. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Sánchez Y and Huarte M: Long non-coding RNAs: Challenges for diagnosis and therapies. Nucleic Acid Ther. 23:15–20. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Veneziano D, Di Bella S, Nigita G, Laganà A, Ferro A and Croce CM: Noncoding RNA: Current deep sequencing data analysis approaches and challenges. Hum Mutat. 37:1283–1298. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al: The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 47:199–208. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Botti G, Marra L, Malzone MG, Anniciello A, Botti C, Franco R and Cantile M: LncRNA HOTAIR as prognostic circulating marker and potential therapeutic target in patients with tumor diseases. Curr Drug Targets. 18:27–34. 2017. View Article : Google Scholar : PubMed/NCBI

13 

He Y, Luo Y, Liang B, Ye L, Lu G and He W: Potential applications of MEG3 in cancer diagnosis and prognosis. Oncotarget. 8:73282–73295. 2017.PubMed/NCBI

14 

Peng Z, Wang J, Shan B, Yuan F, Li B, Dong Y, Peng W, Shi W, Cheng Y, Gao Y, et al: Genome-wide analyses of long noncoding RNA expression profiles in lung adenocarcinoma. Sci Rep. 7:153312017. View Article : Google Scholar : PubMed/NCBI

15 

Wang W, Zhao Z, Yang F, Wang H, Wu F, Liang T, Yan X, Li J, Lan Q, Wang J and Zhao J: An immune-related lncRNA signature for patients with anaplastic gliomas. J Neurooncol. 136:263–271. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Evans JR, Feng FY and Chinnaiyan AM: The bright side of dark matter: lncRNAs in cancer. J Clin Invest. 126:2775–2782. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Russell MR, Penikis A, Oldridge DA, Alvarez-Dominguez JR, McDaniel L, Diamond M, Padovan O, Raman P, Li Y, Wei JS, et al: CASC15-S is a tumor suppressor lncRNA at the 6p22 neuroblastoma susceptibility locus. Cancer Res. 75:3155–3166. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

19 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Lin C, Yuan G, Hu Z, Zeng Y, Qiu X, Yu H and He S: Bioinformatics analysis of the interactions among lncRNA, miRNA and mRNA expression, genetic mutations and epigenetic modifications in hepatocellular carcinoma. Mol Med Rep. 19:1356–1364. 2019.PubMed/NCBI

21 

Liu H, Li J, Koirala P, Ding X, Chen B, Wang Y, Wang Z, Wang C, Zhang X and Mo YY: Long non-coding RNAs as prognostic markers in human breast cancer. Oncotarget. 7:20584–20596. 2016.PubMed/NCBI

22 

Pan W, Zhou L, Ge M, Zhang B, Yang X, Xiong X, Fu G, Zhang J, Nie X, Li H, et al: Whole exome sequencing identifies lncRNA GAS8-AS1 and LPAR4 as novel papillary thyroid carcinoma driver alternations. Hum Mol Genet. 25:1875–1884. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Liu J, Wu W and Jin J: A novel mutation in SIRT1-AS leading to a decreased risk of HCC. Oncol Rep. 34:2343–2350. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Zampetaki A, Albrecht A and Steinhofel K: Long non-coding RNA structure and function: Is there a link? Front Physiol. 9:12012018. View Article : Google Scholar : PubMed/NCBI

25 

Yao RW, Wang Y and Chen LL: Cellular functions of long noncoding RNAs. Nat Cell Biol. 21:542–551. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Wu XS, Wang F, Li HF, Hu YP, Jiang L, Zhang F, Li ML, Wang XA, Jin YP, Zhang YJ, et al: LncRNA-PAGBC acts as a microRNA sponge and promotes gallbladder tumorigenesis. EMBO Rep. 18:1837–1853. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Signal B, Gloss BS and Dinger ME: Computational approaches for functional prediction and characterisation of long noncoding RNAs. Trends Genet. 32:620–637. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Jaksik R, Iwanaszko M, Rzeszowska-Wolny J and Kimmel M: Microarray experiments and factors which affect their reliability. Biol Direct. 10:462015. View Article : Google Scholar : PubMed/NCBI

29 

Shi Y and Shang J: Long noncoding RNA expression profiling using arraystar LncRNA microarrays. Methods Mol Biol. 1402:43–61. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Sun Y and Ma L: New insights into long non-coding RNA MALAT1 in cancer and metastasis. Cancers (Basel). 11:2019. View Article : Google Scholar

31 

Tee AE, Bing L, Song R, Li J, Pasquier E, Cheung BB, Jiang C, Marshall GM, Haber M, Norris MD, et al: The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression. Oncotarget. 7:8663–8675. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Xia T, Liao Q, Jiang X, Shao Y, Xiao B, Xi Y and Guo J: Long noncoding RNA associated-competing endogenous RNAs in gastric cancer. Sci Rep. 4:60882014. View Article : Google Scholar : PubMed/NCBI

33 

Ji J, Tang J, Deng L, Xie Y, Jiang R, Li G and Sun B: LINC00152 promotes proliferation in hepatocellular carcinoma by targeting EpCAM via the mTOR signaling pathway. Oncotarget. 6:42813–42824. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Wang Y, Li M, Dong C, Ma Y, Xiao L, Zuo S, Gong Y, Ren T and Sun B: Linc00152 knockdown inactivates the Akt/mTOR and Notch1 pathways to exert its anti-hemangioma effect. Life Sci. 223:22–28. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Zhang Y, Xiang C, Wang Y, Duan Y, Liu C, Jin Y and Zhang Y: lncRNA LINC00152 knockdown had effects to suppress biological activity of lung cancer via EGFR/PI3K/AKT pathway. Biomed Pharmacother. 94:644–651. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Xu J, Guo J, Jiang Y, Liu Y, Liao K, Fu Z and Xiong Z: Improved characterization of the relationship between long intergenic non-coding RNA Linc00152 and the occurrence and development of malignancies. Cancer Med. 8:4722–4731. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Zhu Y, Dai B, Zhang H, Shi G, Shen Y and Ye D: Long non-coding RNA LOC572558 inhibits bladder cancer cell proliferation and tumor growth by regulating the AKT-MDM2-p53 signaling axis. Cancer Lett. 380:369–374. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Podnar J, Deiderick H, Huerta G and Hunicke-Smith S: Next-generation sequencing RNA-Seq library construction. Curr Protoc Mol Biol. 106:4.21.1–19. 2014. View Article : Google Scholar

39 

Metzker ML: Sequencing technologies-the next generation. Nat Rev Genet. 11:31–46. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Lin T, Hou PF, Meng S, Chen F, Jiang T, Li ML, Shi ML, Liu JJ, Zheng JN and Bai J: Emerging roles of p53 related lncRNAs in cancer progression: A systematic review. Int J Biol Sci. 15:1287–1298. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Sánchez Y, Segura V, Marín-Béjar O, Athie A, Marchese FP, González J, Bujanda L, Guo S, Matheu A and Huarte M: Genome-wide analysis of the human p53 transcriptional network unveils a lncRNA tumour suppressor signature. Nat Commun. 5:58122014. View Article : Google Scholar : PubMed/NCBI

42 

Hünten S, Kaller M, Drepper F, Oeljeklaus S, Bonfert T, Erhard F, Dueck A, Eichner N, Friedel CC, Meister G, et al: p53-regulated networks of protein, mRNA, miRNA and lncRNA expression revealed by integrated pulsed stable isotope labeling with amino acids in cell culture (pSILAC) and next generation sequencing (NGS) analyses. Mol Cell Proteomics. 14:2609–2629. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Kaller M, Götz U and Hermeking H: Loss of p53-inducible long non-coding RNA LINC01021 increases chemosensitivity. Oncotarget. 8:102783–102800. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Yamaguchi H and Hung MC: Regulation and role of EZH2 in cancer. Cancer Res Treat. 46:209–222. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Qi Y, Ooi HS, Wu J, Chen J, Zhang X, Tan S, Yu Q, Li YY, Kang Y, Li H, et al: MALAT1 long ncRNA promotes gastric cancer metastasis by suppressing PCDH10. Oncotarget. 7:12693–12703. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Michalak EM, Burr ML, Bannister AJ and Dawson MA: The roles of DNA, RNA and histone methylation in ageing and cancer. Nat Rev Mol Cell Biol. 20:573–589. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Merry CR, Forrest ME, Sabers JN, Beard L, Gao XH, Hatzoglou M, Jackson MW, Wang Z, Markowitz SD and Khalil AM: DNMT1-associated long non-coding RNAs regulate global gene expression and DNA methylation in colon cancer. Hum Mol Genet. 24:6240–6253. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Somasundaram S, Forrest ME, Moinova H, Cohen A, Varadan V, LaFramboise T, Markowitz S and Khalil AM: The DNMT1-associated lincRNA DACOR1 reprograms genome-wide DNA methylation in colon cancer. Clin Epigenetics. 10:1272018. View Article : Google Scholar : PubMed/NCBI

49 

Kondo A, Nonaka A, Shimamura T, Yamamoto S, Yoshida T, Kodama T, Aburatani H and Osawa T: Long noncoding RNA JHDM1D-AS1 promotes tumor growth by regulating angiogenesis in response to nutrient starvation. Mol Cell Biol. 37:2017. View Article : Google Scholar

50 

Hu Q, Ye Y, Chan LC, Li Y, Liang K, Lin A, Egranov SD, Zhang Y, Xia W, Gong J, et al: Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression. Nat Immunol. 20:835–851. 2019. View Article : Google Scholar : PubMed/NCBI

51 

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

52 

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

53 

de Kok JB, Verhaegh GW, Roelofs RW, Hessels D, Kiemeney LA, Aalders TW, Swinkels DW and Schalken JA: DD3(PCA3), a very sensitive and specific marker to detect prostate tumors. Cancer Res. 62:2695–2698. 2002.PubMed/NCBI

54 

Prensner JR, Zhao S, Erho N, Schipper M, Iyer MK, Dhanasekaran SM, Magi-Galluzzi C, Mehra R, Sahu A, Siddiqui J, et al: RNA biomarkers associated with metastatic progression in prostate cancer: A multi-institutional high-throughput analysis of SChLAP1. Lancet Oncol. 15:1469–1480. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Smolle MA, Bauernhofer T, Pummer K, Calin GA and Pichler M: Current insights into long non-coding RNAs (LncRNAs) in prostate cancer. Int J Mol Sci. 18:2017. View Article : Google Scholar

56 

Merry CR, Mcmahon S, Thompson CL, Miskimen KL, Harris LN and Khalil AM: Integrative transcriptome-wide analyses reveal critical HER2-regulated mRNAs and lincRNAs in HER2+ breast cancer. Breast Cancer Res Treat. 150:321–334. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Li W, Zhai L, Wang H, Liu C, Zhang J, Chen W and Wei Q: Downregulation of LncRNA GAS5 causes trastuzumab resistance in breast cancer. Oncotarget. 7:27778–27786. 2016.PubMed/NCBI

58 

Fradet V, Toren P, Nguile-Makao M, Lodde M, Lévesque J, Léger C, Caron A, Bergeron A, Ben-Zvi T, Lacombe L, et al: Prognostic value of urinary prostate cancer antigen 3 (PCA3) during active surveillance of patients with low-risk prostate cancer receiving 5α-reductase inhibitors. BJU Int. 121:399–404. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Ma W, Chen X, Ding L, Ma J, Jing W, Lan T, Sattar H, Wei Y, Zhou F and Yuan Y: The prognostic value of long noncoding RNAs in prostate cancer: A systematic review and meta-analysis. Oncotarget. 8:57755–57765. 2017.PubMed/NCBI

60 

Liu L, Meng T, Yang XH, Sayim P, Lei C, Jin B, Ge L and Wang HJ: Prognostic and predictive value of long non-coding RNA GAS5 and mircoRNA-221 in colorectal cancer and their effects on colorectal cancer cell proliferation, migration and invasion. Cancer Biomark. 22:283–299. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 19 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ren X: Genome‑wide analysis reveals the emerging roles of long non‑coding RNAs in cancer (Review). Oncol Lett 19: 588-594, 2020
APA
Ren, X. (2020). Genome‑wide analysis reveals the emerging roles of long non‑coding RNAs in cancer (Review). Oncology Letters, 19, 588-594. https://doi.org/10.3892/ol.2019.11141
MLA
Ren, X."Genome‑wide analysis reveals the emerging roles of long non‑coding RNAs in cancer (Review)". Oncology Letters 19.1 (2020): 588-594.
Chicago
Ren, X."Genome‑wide analysis reveals the emerging roles of long non‑coding RNAs in cancer (Review)". Oncology Letters 19, no. 1 (2020): 588-594. https://doi.org/10.3892/ol.2019.11141