Long non‑coding RNAs in small cell lung cancer: A potential opening to combat the disease (Review)

  • Authors:
    • Tian‑Tian Li
    • Rong‑Quan He
    • Jie Ma
    • Zu‑Yun Li
    • Xiao‑Hua Hu
    • Gang Chen
  • View Affiliations

  • Published online on: August 7, 2018     https://doi.org/10.3892/or.2018.6635
  • Pages: 1831-1842
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer is the top cause of cancer‑associated mortality in men and women worldwide. Small cell lung cancer (SCLC) is a subtype that constitutes ~15% of all lung cancer cases. Long non‑coding RNAs (lncRNAs), possessing no or limited protein‑coding ability, have gained extensive attention as a potentially promising avenue by which to investigate the biological regulation of human cancer. lncRNAs can modulate gene expression at the transcriptional, post‑transcriptional and epigenetic levels. The current review highlights the developing clinical implications and functional roles of lncRNAs in SCLC, and provides directions for their future utilization in the diagnosis and treatment of SCLC.

Introduction

Lung cancer, recognized as the leading cause of cancer-associated mortality worldwide, is classified into small cell lung cancer (SCLC) and non-SCLC (NSCLC). SCLC constitutes ~15% of all confirmed cases of lung cancer worldwide (13). Distinct from NSCLC, SCLC is unique in its inclination for quick metastasis and sensitivity to initial systemic cytotoxic chemotherapy. Systemic chemotherapy is the solid foundation of treatment for the limited and extensive stages of this disease. Nevertheless, the commonly adopted management standard of platinum-oriented chemotherapy has reached an efficacy bottleneck, mainly due to chemoresistance and relapse in SCLC patients (4). Despite plentiful clinical trials in the past four decades, systematic treatment for SCLC patients has not changed markedly. As a result, the majority of patients live for only 1 year or less following diagnosis, with the overall 5-year survival rate staying low at <7% (5). The widely employed technique to diagnose SCLC from a tiny amount of malignant cells, in combination with a lack of proved predictive biomarkers that would require tissue biopsies and relatively rare surgical resection, has cut down the source of SCLC tissue for more profound studies (5). In NSCLC, a growing number of gene fusions or mutations instruct treatment selections for specific patient subgroups, particularly those with anaplastic lymphoma kinase or epidermal growth factor receptor (EGFR) (6) mutations. In marked contrast, numerous experimental and targeted agents regarding SCLC have failed to yield convincing clinical benefits (7). Novel and effective therapies for SCLC patients are urgently required. Regarding the molecular mechanisms of carcinogenesis, medical communities have mostly concentrated on genes with protein-coding capacity. Unexpectedly, the ENCODE project identified that up to three-quarters of the human genome could be transcribed, though <3% of it encodes protein. This unexpected fact indicated that non-coding RNAs (ncRNAs) make up the vast majority of the human transcriptome (8). Long ncRNAs (lncRNAs) are >200 nucleotides in size and possess no or a very low protein-coding ability. Bioinformatics platforms and high-throughput sequencing emerging in recent years have facilitated uncovering the mystique of lncRNAs, which function as key molecules in wide-ranging cellular processes, including cell growth, adhesion, proliferation and apoptosis (9,10). lncRNA deregulation is involved in numerous human diseases, and there is also increasing evidence suggesting that lncRNAs are involved in SCLC pathogenesis and clinical outcomes (1115). Digging deeper into the biological functions and molecular mechanisms of lncRNAs will enable researchers to further understand the biology of SCLC and develop lncRNA-oriented therapeutics.

Molecular mechanisms of lncRNAs

What is lncRNA? The manifestation of lncRNAs owes much to the studies on the size, function and evolution of the human genome. Along with the development of DNA-RNA hybridization techniques, scientists have gradually become aware that the majority of the genome, which was initially labeled as ‘junk DNA’, does not encode proteins (16). However, subsequent studies demonstrating that small nucleolar RNAs (snoRNAs) and small nuclear RNAs (snRNAs) have a certain impact on post-transcriptional RNA processing propelled further investigation into non-coding sequences (17,18). In the early 2000s, whole-transcriptome sequencing arose and carried forward the identification and annotation of numerous lncRNAs (1921). ncRNAs can be large or small in size. Linearized ncRNAs with a length of >200 bp and with no or low protein-coding ability are known as lncRNAs. Small ncRNAs (<200 bp) are categorized into PIWI-interacting RNAs, small interfering RNAs, microRNAs (miRNAs) and classical housekeeping ncRNAs, including transfer RNAs, ribosomal RNAs, snRNAs and snoRNAs. The FANTOM5 project has identified 19,175 potential functional lncRNAs in the human genome (22), yet few of them have been thoroughly investigated (23). Accumulating studies have supported the theory that at different levels, aberrant expression of lncRNAs serves crucial roles in cancer development, affecting cell growth, proliferation, apoptosis and metastasis via diverse mechanisms (12,13,15,24).

Four archetypes of lncRNAs

lncRNAs are a set of ubiquitous genes participating in various biological mechanisms. There are four archetypes in which lncRNAs execute their molecular functions, namely as signals, decoys, guides and scaffolds (25). The signal archetype of lncRNAs may serve as markers of functionally significant biological events, as their expression exhibits cell type, time and space specificity. For example, lncRNA homeobox (HOX) transcript antisense intergenic RNA (HOTAIR) located in the HOXC locus exists in posterior and distal cells, whereas another HOXC lncRNA, Frigidair, is expressed in an anterior pattern. Conversely, lncRNA HOXA transcript at the distal tip (HOTTIP), located in the far end of the human HOXA cluster, is expressed in distal cells (26,27). The decoys archetype is a type of lncRNA that regulates transcription through binding to and then carrying away protein targets, yet it does not exert extra functions. Decoys display as ‘molecular sinks’ for chromatin modifiers, transcription factors or other regulatory factors, all of which are RNA-binding proteins (25). For instance, by directly binding to and sequestering nuclear transcription factor Y subunit that drives a DNA damage-induced apoptotic program, lncRNA p21-associated ncRNA DNA damage-activated suppresses apoptotic gene expression to facilitate cell cycle arrest, leading to the promotion of cell survival (28). Knockdown of lncRNAs of this archetype may imitate the gain-of-function of the target proteins, while a rescue phenotype could be induced by loss-of-function of the lncRNA and its effector (25). The guides archetype of lncRNA can bind chromatin modifying proteins and direct the localization of ribonucleoprotein complexes to specific targets in a cis or trans manner. The well-known cis mechanism, mammalian X inactivation center, specifies a set of ncRNAs, X-inactive specific transcript (Xist) included (29,30). A 1.6-kb lncRNA, RepA RNA, stemming from the 5′ end of Xist, produces polycomb repressive complex 2 (PRC2) in cis. PRC2 is involved in extra X-chromosome inactivation (31). In contrast to cis-regulatory lncRNAs, certain lncRNAs serve their chromosome-wide transcriptional roles in trans, such as lncRNA HOTAIR, which is capable of directing PRC2 to target genes in trans (3234). The scaffolds archetype of lncRNA can act a platform where components are assembled, precisely regulating the sophisticated molecular interactions and signaling transductions involved in diverse biological signaling processes (35). For example, telomerase catalytic activity necessitates the combination of two common telomerase units, the telomerase RNA (TERC) and the telomerase reverse transcriptase (TERT). TERC is an essential lncRNA unit that offers the template for repeat synthesis, and it also possesses domains that promote TERT binding, catalytic activity and stability of the complex (36). Certain morbid states, including dyskeratosis congenital, presumably result from mutations altering the equilibrium between different conformations of TERC, more specifically, through destruction of the RNA scaffold structure where modular biding sites for telomeric regulatory proteins are located (37).

lncRNAs modulate gene expression at distinct levels

lncRNAs exert functions in an enormous range of biological processes by promoting or inhibiting the transcription and translation of protein-coding genes. Unlike highly conserved small ncRNAs that participate in gene silencing transcriptionally and post-transcriptionally (3840), lncRNAs are poorly conserved and can modulate target gene expression via various mechanisms at different levels.

Transcriptional level

At the transcriptional level, lncRNAs have the following roles: i) Functioning as decoys for RNA polymerase II or transcription factors (TFs) to inhibit their binding to enhancers or promoters of target genes, therefore specifically promoting or repressing target gene expression (26); ii) alteration of TF localization or modification to promote or inhibit gene transcription (40); iii) interaction with DNA to form a triple helix structure, thereby affecting target gene transcription (41); and iv) presenting as competitive endogenous RNAs (ceRNAs) to inhibit the transcription of target genes (42).

Post-transcriptional level

At the post-transcriptional level, lncRNAs have the following roles: i) Providing different transcripts by regulating pre-mRNA alternative splicing (43); ii) combining with mRNAs to synthesize double-stranded RNA complexes, thereby effectively enhancing the stability of mRNAs (44); and iii) interaction with miRNAs to regulate signaling events (45).

Epigenetic level

At the epigenetic level, lncRNAs have the following roles: i) The regulation of histone modifications, including acetylation, methylation and ubiquitination, among others (46); ii) participating in chromatin remodeling and conformational alterations by combining with chromatin modification complexes that are crucial for gene transcription (47); and iii) participating in gene silencing via modulating DNA methylation in the promoter region of target genes (48).

To summarize, lncRNAs participate in diverse transcriptional, post-transcriptional and epigenetic molecular mechanisms, covering regulation of chromatin structure or modification, transcription, splicing and translation, therefore regulating a multitude of physiological and pathological courses, including cell proliferation, differentiation, apoptosis, the heat shock response, cancer development and chemoresistance (4951). Amongst these functions, the regulation of gene expression is of paramount significance in elucidating how lncRNAs promote or suppress tumorigenesis. Genome-wide studies of tumor samples have verified plentiful lncRNAs that are linked to distinct types of cancer. Dysregulated expression of lncRNAs can stimulate carcinogenesis and metastasis. However, from an overall perspective, the function of lncRNAs may not be one-sided, and could be tumor-promoting or tumor-suppressing.

Expression of lncRNAs in SCLC

Representing one of the largest classes of transcripts, lncRNAs possess highly diverse characteristics and functions. Progress in high-throughput sequencing technology has accelerated the identification of lncRNAs as key regulatory molecules participating in various cellular processes and their dysregulation in human diseases. Although only a few lncRNAs have been well described thus far, accumulating evidence suggests that lncRNAs contribute to tumor biology. Given the aforementioned difficulties, breakthroughs in SCLC research remain stagnant compared with those in other types of cancer. However, it remains worthwhile to investigate the research status of SCLC from an lncRNA point of view, as this field may open novel and optimistic windows to elucidate SCLC molecular mechanisms. In the following text, previous findings in the expression of lncRNAs in SCLC are reviewed. The roles that HOTTIP, HOTAIR, taurine upregulated gene 1 (TUG1), colon cancer-associated transcript 2 (CCAT2) and plasmacytoma variant translocation 1 (PVT1) serve in SCLC are discussed and also briefly presented in Table I. A schematic diagram of these genes in SCLC and their mechanisms is shown in Fig. 1. The dysregulations and functions of these five lncRNAs in other malignancies are also summarized, as a contrast and enlightenment to their roles in SCLC (Table II).

Table I.

Information of five lncRNAs involved In SCLC.

Table I.

Information of five lncRNAs involved In SCLC.

lncRNAGenomic locationDysregulationFunctionsMechanismPublication year(Refs.)
HOTTIP7p15.2UpregulationChemoresistance, shorter survival, clinical stage HOTTIP/miR-574-5p/EZH1 axis, HOTTIP/miR-216a/BCL-2 axis2017, 2018(57,58)
HOTAIR12q13.13UpregulationLymphatic invasion, chemoresistanceRegulation of HOXA1 methylation and target genes2013, 2016(66,67)
TUG122q12.2UpregulationChemoresistance, clinical stage and shorter survivalRegulation of LIMK2b via binding with EZH22017(76)
CCAT28q24.21UpregulationMalignant status, poor prognosisUnknown2016(85)
PVT18q24.21UpregulationLymph node metastasis, distal metastasis, and clinical stage lncRNAUnknown2016(95)

[i] SCLC, small cell lung cancer; HOTTIP, HOXA transcript at the distal tip; EZH1, enhancer of zeste homolog 1; BCL-2, B-cell leukemia/lymphoma-2; HOTAIR, HOX transcript antisense intergenic RNA; TUG1, taurine upregulated gene 1; CCAT2, colon cancer-associated transcript 2; PVT1, plasmacytoma variant translocation 1; HOXA1, homeobox A1; LIMK2b, LIM domain kinase 2; EZH2, enhancer of zeste homolog 2; miR, microRNA; lncRNA, long non-coding RNA.

Table II.

Differential expression and mechanisms of five lncRNAs in SCLC and other malignancies.

Table II.

Differential expression and mechanisms of five lncRNAs in SCLC and other malignancies.

lncRNAMalignancyDysregulation Functions/mechanisms(Refs.)
HOTTIPSCLCUpregulationChemoresistance, tumor progression. HOTTIP/miR-574-5p/EZH1 network, HOTTIP/miR-216a/BCL-2 network(57,58)
Liver cancerUpregulationMetastasis and tumor progression. Regulation of HOXA13(55)
Gastric cancerUpregulationCell proliferation, migration and invasion. Regulation of HOXA13 and HOTTIP/miR-331-3p/HER2 network(54,117)
Pancreatic cancerUpregulationCell cycle, proliferation and invasion. Regulation of HOXA13(56)
Prostate cancerUpregulationCell proliferation and apoptosis. Regulation of HOXA13, BAX and BCL-2(118)
HOTAIRSCLCUpregulationCell proliferation, lymphatic invasion, and chemoresistance. Regulation of HOXA1 methylation and target genes(66,67)
Breast cancerUpregulationMetastasis, invasion, poor prognosis, and shorter survival. Regulation of Wnt signaling pathway(80,119)
Colorectal cancerUpregulationMetastasis, poor prognosis, and low survival. Chromatin modification and EMT(120122)
Cervical cancerUpregulationFIGO stage, aggression and lymph node metastasis. Regulation of microRNA(123125)
Gastric cancerUpregulationVenous infiltration, lymph node metastasis, chemoresistance and tumor staging. Regulation of E-cadherin, regulation of PI3K/AKT/MRP1 genes(126129)
TUG1SCLCUpregulationClinical stage, chemoresistance, and shorter survival. Regulation of LIMK2b(76)
Liver cancerUpregulationCell proliferation, apoptosis, metastasis and glycolysis. Regulation of KLF2 transcription. Regulation of AMPKβ2 and HK2(69,130)
Gastric cancerUpregulationCell proliferation and cell cycle arrest. Regulation of cyclin-dependent protein kinase inhibitors(74)
Pancreatic cancerUpregulationCell proliferation and cell migration. Regulation of EZH2 and EMT(131)
Bladder cancerUpregulationCell proliferation and apoptosis. Regulation of Wnt/β-catenin pathway(72,132)
CCAT2SCLCUpregulationMalignant status and poor prognosis(85)
Colorectal cancerUpregulationLymph node metastasis, cell proliferation and differentiation. Regulation of microRNA(77,133)
Ovarian cancerUpregulationFIGO stage, cell proliferation, migration, and invasion. Regulation of microRNA(82,134)
Prostate cancerUpregulationEMT, cell proliferation, invasion, and migration. Regulation of EMT(83)
Breast cancerUpregulationCell proliferation, invasion, chemoresistance, and poor prognosis. Regulation of Wnt signaling pathway(80,81)
PVT1SCLCUpregulationLymph node metastasis, distal metastasis, and clinical stage(95)
Gastric cancerUpregulationLymph node invasion, cell proliferation and invasion. Regulation of microRNA(89,135,136)
LivercancerUpregulationCell proliferation and cell cycling. Regulation of NOP2(90)
Pancreatic cancerUpregulationCell proliferation, migration, shorter survival and poor prognosis. Regulation of microRNA(92,137)
Thyroid cancerUpregulationCell proliferation and cell cycle arrest. Regulation of EZH2 and TSHR(91)

[i] SCLC, small cell lung cancer; HOTTIP, HOXA transcript at the distal tip; EZH1, enhancer of zeste homolog 1, BCL-2, B-cell leukemia/lymphoma-2; HOXA13, homeobox A13; HER2, erb-b2 receptor tyrosine kinase 2; BAX, BCL-2 associated X; HOTAIR, HOX transcript antisense intergenic RNA; TUG1, taurine upregulated gene 1; CCAT2, colon cancer-associated transcript 2; PVT1, plasmacytoma variant translocation 1; HOXA1, homeobox A1; EMT, epithelial-to-mesenchymal transition; FIGO, International Federation of Gynecology and Obstetrics; PI3K, phosphatidylinositol 3-kinase; AKT, protein kinase B; MRP1, multidrug resistance-associated protein 1; LIMK2b, LIM domain kinase 2; KLF2, Kruppel like factor 2; MPKβ2, AMP-activated protein kinase subunit β2; HK2, hexokinase 2; NOP2, NOP2 nucleolar protein; EZH2, enhancer of zeste homolog 2; TSHR, thyroid stimulating hormone receptor; miR, microRNA; lncRNA, long non-coding RNA.

HOTTIP

HOTTIP, as the lncRNA encoded by the HOTTIP gene that is located at the HOXA locus, was initially identified in human fibroblasts distributed in anatomically distal regions of the body (52). Wang et al (53) verified the direct coupling of HOTTIP and the adaptor protein, WD repeat domain 5 (WDR5) to target WDR5/lysine methyltransferase 2A complexes across HOXA, thereby impelling histone lysine 4 trimethylation and the transcription of various 5′ HOXA genes. Multiple studies confirmed the positive correlation between the expression level of HOTTIP and HOXA genes in a variety of malignancies (52,5456). In brief, HOTTIP could activate HOX genes by recruiting histone-modifying enzymes to suppress tumor-suppressor genes. Sun et al (57,58) completed pioneering studies unveiling the underlying molecular mechanism of HOTTIP in SCLC utilizing a series of experiments conducted in vitro and in vivo. At first, gene expression array analysis revealed the overexpression of HOTTIP in H69 and H69R cell lines, and the result was further supported by the significant overexpression of HOTTIP, as detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in 50 clinical SCLC tissues prior to chemotherapy, compared with their non-cancerous counterparts. In addition, higher HOTTIP expression was significantly associated with a poorer prognosis. Manipulation of HOTTIP loss- and gain- of function experiments in SCLC cell lines also demonstrated that HOTTIP overexpression contributed to cell proliferation, as it led to a decreased number of G2-phase cells and an increased number of S-phase cells. In vivo, HOTTIP loss and gain of function experiments conducted in xenograft nude mice showed that mice with knockdown of HOTTIP had a smaller mean tumor volume in comparison to those in the negative control group. Afterwards, by employing web-based bioinformatics platform RNA22-seq (https://cm.jefferson.edu/), miR-574-5p and enhancer of zeste homolog 1 (EZH1) were predicted to possess targeted binding sites for HOTTIP, and this association was later verified by RT-qPCR. Therefore, it was assumed that HOTTIP may exert its effect on SCLC through a regulatory network of miRNA-574-5p-HOTTIP-EZH1 (57). Notably, the hypothesis was verified by a subsequent co-transfection dual luciferase reporter assay, indicating that HOTTIP acts as an oncogene by sponging miR-574-5p to abrogate the expression of polycomb group protein EZH1 induced by miR-574-5p, thereby promoting the progression of SCLC (57). In another study by Sun et al (58), a similar experimental design was applied to investigate the role of HOTTIP in SCLC, and the association of HOTTIP with SCLC chemoresistance was also investigated, which enriched the clinical value of the study. The expression of HOTTIP and HOXA13 was markedly upregulated in SCLC cell lines and biopsy samples. Overexpression of HOTTIP impaired the anti-chemoresistance effects of etoposide, irinotecan and cisplatin toward SCLC cells in vitro and in vivo, whereas knockdown of HOTTIP exhibited a reversed effect. In addition, the finding that knockdown of HOTTIP suppressed HOXA13 expression, combined with the result of a rescue experiment by HOXA13 overexpression implied that HOTTIP exerts its function in SCLC chemoresistance and progression partly via manipulating HOXA13. Likewise, the online bioinformatics tool RNA22-seq excavated miR-216a as possessing targeted binding sites with HOTTIP, and unexpectedly, an atoptosis-related gene, B-cell leukemia/lymphoma-2 (BCL-2). Subsequent experiments confirmed that HOTTIP could function as a competing ‘sponge’ through binding miR-216a, thereby diminishing its silencing effect toward BCL-2, contributing to the chemoresistance and progression of SCLC (58). Although the aforementioned findings may only be the tip of the iceberg, they widen the landscape of research into the molecular mechanism of SCLC, as a novel network composed of lncRNA, miRNA and specific cancer-related genes is put forward, providing inspiration for developing prognostic and therapeutic agents.

HOTAIR

HOTAIR is one of the most well-characterized lncRNAs and is overexpressed in certain malignancies, including breast, colorectal, hepatocellular, gastrointestinal and non-small cell lung cancer (59). First identified in 2007, HOTAIR resides in the HOXC locus. Previous reports revealed that the molecular mechanism of HOTAIR is its transcription from the HOXC gene as an antisense transcript and then binding to PRC2 (composed of EZH2, polycomb protein SUZ12 and polycomb protein EED) and lysine-specific demethylase/CoREST/REST complex as a scaffold, leading to catalyzation of trimethylation of histone H3 on lysine 27 (H3K27) and spontaneous demethylation of H3K4, and repression of the transcription of HOXD genes (27,60). With regard to DNA methylation, EZH2, a compartment of PRC2, directly interacts with DNA methyltransferases. This interaction assists in maintaining DNA methylation and stabilizing the repression of certain genes, including various tumor suppressors (61). As targets of HOTAIR, the homeobox-containing genes are a set of regulators that transcriptionally encode DNA-binding homeodomains that participate in controlling normal development (51,62). In addition, abnormal expression of HOX genes is associated with oncogenesis and paramorphia (61,63). Additionally, by inducing epithelial-to-mesenchymal transition (EMT), HOTAIR associates with tumorigenesis (64). HOTAIR also triggers ubiquitin-mediated proteolysis via interaction with RNA-binding protein MEX3B and E3 ubiquitin-protein ligase DZIP3 (65). Ono et al (66) studied the association of HOTAIR with SCLC cellular processes and clinical characteristics. The study assessed HOTAIR expression in 35 surgically resected SCLC tissues and 10 SCLC cell lines, and observed that expression of HOTAIR in pure SCLC was markedly overexpressed compared with that in those combined with lung adenocarcinoma (LUAD), large cell carcinoma or large cell neuroendocrine carcinoma, and that HOTAIR overexpression was clearly associated with relapse and lymphatic invasion. In vitro experiments indicated that the expression of HOTAIR in half of the SCLC cell lines was elevated compared with that in normal cells. Knockdown of HOTAIR reduced cellular invasiveness and proliferative activity of SBC-3 cells. Gene expression analysis revealed that a reduction in HOTAIR led to upregulated expression of mucin production-related genes, including mucin 5AC, and cell adhesion-related genes, including astrotactin 1 and protocadherin α1, and downregulated expression of genes such as neurotrimin and protein tyrosine kinase 2β, participating in neuronal growth and signal transduction, respectively (66). Recently, Fang et al (67) investigated the role of HOTAIR expression in the chemoresistance of SCLC and its underlying mechanism. The study assessed the impact of HOTAIR on SCLC chemoresistance in vitro and observed that HOTAIR expression was also markedly upregulated in drug-resistant cell lines compared with that in the parental cell lines. The study showed that downregulated expression of HOTAIR promoted cell cycle arrest and apoptosis to increase sensitivity to antitumor drugs, while repressing tumor growth in vivo. In addition, increased HOXA1 methylation was observed in the drug-resistant cells. An enzyme-linked immunosorbent assay revealed that a reduction of HOTAIR lessened HOXA1 methylation via reducing the expression of DNA (cytosine-5)-methyltransferase (DNMT)3b and DNMT1. RNA immunoprecipitation validated the interaction between HOXA1 and HOTAIR. Together, these findings indicated that HOTAIR mediates chemoresistance by increasing HOXA1 methylation. Hence, HOTAIR could serve as a possible target for novel therapeutics to combat chemoresistance. Based on these previous findings, lncRNA HOTAIR is involved in the relapse and lymphatic invasion in SCLC patients, and it could also act as a biomarker for prognosis and chemotherapy response, and as a therapeutic target to overcome the chemoresistance of SCLC.

TUG1

TUG1 was initially described as a spliced and polyadenylated lncRNA necessary for the development of photoreceptors in mice retina (68). Increasing evidence has demonstrated that TUG1 serves a crucial role in a number of human tumors, including hepatocellular carcinoma, osteosarcoma, glioma, esophageal, gastric and bladder cancer (6974). Aberrant expression of PRC2-related lncRNAs is involved in tumorigenesis and progression. In a previous study, TUG1 was found to be induced by p53, prior to binding to PCR2 and influencing certain genes involved in the modulation of mitosis, spindle construction and cell-cycle phasing (34). Yang et al (75) revealed that a combination of methylated PRC2 and TUG1 manipulates the relocation of growth-control genes between interchromatin granules and polycomb bodies in response to growth signals, therefore portraying a role that TUG1 serves in the relocation of transcription units to coordinate gene expression (75). Niu et al (76) investigated the functions of TUG1 in the cell proliferation and chemoresistance of SCLC, and its underlying molecular mechanism (76). The study analyzed TUG1 expression in tissue samples from SCLC patients (n=33) who had undergone biopsy or bronchofiberscopy, and elevated TUG1 expression was found in cancerous tissues compared with that in adjacent non-cancerous tissues. Statistical analysis showed that higher expression of TUG1 was associated with shorter survival time, advanced clinical stage and cigarette smoking. In vitro Cell Counting Kit-8 and colony formation assays indicated that silencing TUG1 markedly reduced cell growth. The results from flow cytometry analysis conducted to assess the effect of TUG1 on cell apoptosis suggested that knockdown of TUG1 promoted apoptosis and led to a significant accumulation of G1-phase cells, and that downregulated TUG1 expression increased apoptosis in H44DDP and H69AR cell lines exposed to anticancer drugs. The chemoresistance-inducing ability of TUG1 in vivo was further investigated using a mouse xenograft model, and the result was consistent with that of the in vitro experiment. Moreover, TUG1 could modulate LIM domain kinase 2 expression through binding with EZH2, and subsequently led to increased cell growth and chemoresistance in SCLC. Outcomes of this study could be guidance to the development of innovative TUG1-directed prognostic and therapeutic strategies.

CCAT2

CCAT2 was first introduced in 2013 as an lncRNA located in the 8q24 gene desert region, and it possesses a tumor-related single nucleotide polymorphism rs6983276. Additionally, overexpression of CCAT2 in colon cancer was observed, and it was considered to serve an oncogenic role, promoting colorectal cancer cell proliferation and motility, metastasis and chromosomal instability by regulating myc and Wnt pathways (77). A CCAT2 genetic polymorphism, rs6983267, is associated with platinum-based chemotherapy sensitivity in lung cancer patients (78). Since its discovery, the oncogenic role of CCAT 2 has been increasingly demonstrated in different tumors, including gastric, breast, lung, liver, colon, cervical, ovarian, bladder, prostate and esophageal cancer (7984). The stimulatory effects on the Wnt/β-catenin signaling pathway, cancer metabolism and EMT may underlie its oncogenic action (79). CCAT2 is upregulated in an estimated two-thirds of breast cancer patients (80). High CCAT2 expression was associated with a poor curative effect from cyclophosphamide/methotrexate/fluorouracil-containing adjuvant chemotherapy in breast cancer patients with lymph node metastasis (80). Chen et al (85) detected CCAT2 expression in 102 human SCLC tissues, 15 paired non-tumor tissues, SCLC cell lines (DMS-53 and H446) and a normal bronchial epithelial cell line (16HBE). The association between clinicopathological factors and CCAT2 expression was subsequently analyzed. The study reported that CCAT2 level was significantly overexpressed in SCLC tissue and cell lines compared with that in normal lung tissues. Subgroup analyses also indicated that higher expression of CCAT2 was correlated with malignant status and poor prognosis in SCLC patients. Moreover, knockdown of CCAT2 to inhibit SCLC cell growth and metastasis in vitro was observed. To conclude, CCAT2 may serve as an oncogene and a negative prognostic indicator in SCLC.

PVT1

PVT1 is an lncRNA homologous to the mouse plasmacytoma variant translocation gene (Pvt1), which was first identified as being frequently involved in a variant translocation in plasmacytoma in the mid-80s in mice (86,87). Soon after, the PVT1 locus emerged as a site of variant translocations in Burkitt lymphoma. Subsequent studies support the role of PVT1 as a cancer risk locus in relation to the well-known myc oncogene (88). PVT1 presents the capacity to facilitate cell growth and suppress cell apoptosis in the tumorigenesis of various types of cancer, including gastric (89), liver (90), thyroid (91) and pancreatic (92) cancer, and non-small cell lung cancer (93). The expression of PVT1 in tumor samples of these types of cancer is elevated. In vitro and in vivo experiments conducted by Wang et al (90) demonstrated that PVT1 promotes cell proliferation, cell cycling and the acquisition of stem cell-like properties in hepatocellular carcinoma cell by stabilizing NOP2 nucleolar protein (90). However, the underlying mechanisms of the functional exertion of PVT1 and its interaction with downstream targets remain largely unknown. Partially known molecular functions of PVT1 can be categorized into three key pathways: Partaking in DNA rearrangement, encoding microRNAs and intercommunicating with myc (94). Recently, Huang et al (95) first identified the role of PVT1 in SCLC. In the study, PVT1 expression was detected in SCLC tissues, paired normal gastric tissues and two SCLC cell lines. Meanwhile, the association of PVT1 expression levels with clinical features of 120 enrolled SCLC patients was analyzed. RT-PCR analysis showed that PVT1 expression was significantly higher in SCLC tissues and cell lines than in their normal counterparts, and positive correlations between PVT1 overexpression and the status of clinical stage, lymph node metastasis and distal metastasis in SCLC were noted. Furthermore, multivariate analysis revealed that PVT overexpression could be an independent prognostic biomarker for the survival of SCLC patients. Cell migration and invasion were significantly suppressed in vitro by silencing of PVT1 in SCLC. To conclude, PVT1 possesses the potency to be a novel marker and a prospect to develop targeted therapy for SCLC. However, further investigations are required for thorough elucidation of the molecular mechanism of PVT1 in SCLC.

Thus far, the present review has summarized and discussed five lncRNAs (HOTTIP, HOTAIR, TUG1, CCAT2 and PVT1) involved in SCLC. However, these lncRNAs are also involved in NSCLC, which accounts for ~85% of all lung cancer cases. It is of merit to include information on the roles that these lncRNAs serve in NSCLC, as a contrast and possibly, enlightenment. HOTTIP was reported to be significantly upregulated in NSCLC and to function as an oncogene by regulating HOXA13, which coincides with the findings observed in other malignancies, implying that HOXA13 is a key element through which HOTTIP promotes carcinogenesis (96). Moreover, overexpression of HOTTIP was found to motivate LUAD cell proliferation and chemoresistance via regulating the protein kinase B (AKT) signaling pathway (97). As for HOTAIR, multiple studies also demonstrated its overexpression in NSCLC, and it is involved in the initiation and development of NSCLC through interacting with unc-51-like autophagy-activating kinase 1 to suppress autophagy (98), targeting caveolin 1 (99) and miR-613 (100). Based on the available literature, there is controversy with regard to the expression of TUG1 in NSCLC. Studies by Lin et al (101) and Zhang et al (102) revealed that TUG1 was downregulated in NSCLC, while a study by Liu et al (103) showed that it was upregulated. According to the study by Liu et al, TUG1 could inhibit apoptosis by silencing BCL-2 associated X via interacting with EZH2 (103). TUG1 RNA could target PRC2 in the promotor region of CUGBP Elav-like family member 1 (CELF1) and CELF1 expression was therefore negatively regulated (101). Zhang et al (102) suggested that TUG1 acted as a growth regulator in NSCLC partly through controlling HOXB7. CCAT2 exerted overexpression in NSCLC (104,105), and could promote oncogenesis via overexpression of zinc finger and BTB domain containing 7A (104). With regard to PVT1, it is of note that accumulating recent studies (106109) investigated the roles of upregulated PVT1 in NSCLC via the ceRNA-regulated network, which is a trending hotspot in the research field. PVT1 was demonstrated to exert its oncogenic functions by sponging miR199a5p (106), miR-126 (107), miR-497 (108) and miR-195 (109).

Conclusions and future directions

SCLC is a fatal disease with an aggressive and brutal nature; it comprises ~15% of all lung cancer cases. The management of SCLC remains challenging, while disease outcome has remained poor, mainly due to limited options for effective treatment. The majority of the cases are at an irreversible advanced stage when diagnosed and rapidly develop treatment resistance despite a high success rate of initial chemotherapy and radiation. The pathogenesis of SCLC has been investigated by researchers across the world; nevertheless, the implicit molecular mechanism remains mostly unidentified. In light of next-generation sequencing techniques and bioinformatics tools, lncRNAs have been shown to exert distinguishable functions in a broad range of human diseases, including the most concerning types of cancer. Although great discoveries and advances in cancer pathogenesis and therapeutics have been made over the decade, the elucidation of the SCLC molecular mechanism and its frontline treatment have developed slowly due to obstacles from various aspects, including difficulty in sample collection, research funding, late diagnosis, rapid progression and chemoresistance. Investigation into the role of lncRNAs in SCLC is underway, yet no lncRNAs have been extensively investigated, let alone clinically utilized for prognosis, diagnosis or therapeutic design. According to the available published literature, the current research state of SCLC is relatively superficial compared with that of NSCLC. Thus, more research is urgently required.

Despite the aforementioned challenges, there have been certain notable novel findings that have the potential to advance the field. Given the scarcity of SCLC tissues, a multidisciplinary, interoperable, cross-institutional approach is required to collect adequate SCLC tissues for more translational research projects. Emerging techniques, including next-generation sequencing and bioinformatics, have created opportunities to conduct larger scale and deeper studies on SCLC. The past decade has witnessed the emergence of lncRNAs involved in various types of cancer. For example, lncRNA prostate cancer antigen 3 and lncRNA highly upregulated in liver cancer can be detected in prostate and liver cancer, respectively, and serve as sensitive diagnostic markers (110,111). As an intensively studied lncRNA, lncRNA metastasis-associated lung adenocarcinoma transcript 1 is found to be involved in multiple malignancies, including lung, colon, breast and liver cancer, indicating its general participation in cancer cell proliferation (112). Overexpression of lncRNA antisense noncoding RNA in the INK4 locus is observed in a number of types of cancer and is associated with a poor prognosis in gastric and prostate cancer (113). lncRNA CCAT1 could be used as a clinically detectable marker to predict the therapeutic responsiveness of bromodomain and extraterminal inhibitors in patients with colorectal cancer (114). lncRNA maternally expressed gene 3, which acts as a tumor-suppressor via promoting p53 accumulation and recruiting PRC2, is downregulated in multiple primary human tumors (115). As these lncRNAs have been verified as promising predictive markers for diagnosis, prognosis and chemotherapy sensitivity in cancer patients, they also have the potential to lead to a greater understanding of SCLC tumorigenesis and chemoresistance, and could serve as efficient therapeutic targets. The diagnostic sensitivity and specificity may be enhanced by joint detection of disparate lncRNAs, and this may become particularly useful in non-invasive screening for early-stage SCLC patients. The functional roles of lncRNAs involve diverse signaling pathways and investigation into these pathways may yield crucial signaling targets that could be blocked to impede tumor progression. Signaling pathways frequently altered in cancer include phosphoinositide 3-kinase/AKT, Kirsten rat sarcoma viral oncogene homolog/V-raf murine sarcoma b-viral oncogene homolog B1, retrovirus-associated DNA sequences/mitogen-activated protein kinase, EGFR, fibroblast growth factor receptor, Wnt and myc, among others (116). Currently, among nearly 20,000 identified lncRNAs, only five have been investigated in SCLC. Therefore, more efforts should be put into this field of great potential. As a large genetic information treasury and a potential opening to combat diseases, lncRNAs will play no small part in identifying SCLC mechanisms.

Acknowledgements

Not applicable.

Funding

The present study was supported by funds from the National Natural Science Foundation of China (grant no. NSFC81560469 and NSFC81360327), the Natural Science Foundation of Guangxi, China (grant no. 2015GXNSFCA139009, 2016GXNSFAA380255 and 2017GXNSFAA198016) and the Guangxi Medical University Training Program for Distinguished Young Scholars (grant no. 2017).

Availability of data and materials

Not applicable.

Authors' contributions

TTL contributed to the literature retrieval and manuscript preparation. RQH and JM contributed to the manuscript modification and commented on multiple aspects of the manuscript. ZYL, XHH and GC jointly supervised the construction of the study, and contributed to the design and approval of the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

SCLC

small cell lung cancer

NSCLC

non-SCLC

EGFR

epidermal growth factor receptor

lncRNAs

long non-coding RNAs

snoRNAs

small nucleolar RNAs

snRNAs

small nuclear RNAs

ncRNAs

non-coding RNAs

miRNAs

microRNAs

HOTAIR

HOX transcript antisense intergenic RNA

HOX

homeobox

PRC2

polycomb repressive complex 2

TERC

telomerase RNA

TERT

telomerase reverse transcriptase

TFs

transcription factors

ceRNAs

competitive endogenous RNAs

HOTTIP

HOXA transcript at the distal tip

WDR5

WD repeat domain 5

EZH1

enhancer of zeste homolog 1

BCL-2

B-cell leukemia/lymphoma-2

CELF1

CUGBP Elav-like family member 1

EZH2

enhancer of zeste homolog 2

EMT

epithelial-to-mesenchymal transition

HOXA1

homeobox A1

TUG1

taurine upregulated gene 1

CCAT2

colon cancer-associated transcript 2

PVT1

plasmacytoma variant translocation 1

CCAT1

colon cancer associated transcript 1

References

1 

van Meerbeeck JP, Fennell DA and De Ruysscher DK: Small-cell lung cancer. Lancet. 378:1741–1755. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Barnes H, See K, Barnett S and Manser R: Surgery for limited-stage small-cell lung cancer. Cochrane Database Syst Rev. 4:CD0119172017.PubMed/NCBI

3 

Sabari JK, Lok BH, Laird JH, Poirier JT and Rudin CM: Unravelling the biology of SCLC: Implications for therapy. Nat Rev Clin Oncol. 14:549–561. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Pillai RN and Owonikoko TK: Small cell lung cancer: Therapies and targets. Semin Oncol. 41:133–142. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Byers LA and Rudin CM: Small cell lung cancer: Where do we go from here? Cancer. 121:664–672. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Dolly SO, Collins DC, Sundar R, Popat S and Yap TA: Advances in the development of molecularly targeted agents in non-small-cell lung cancer. Drugs. 77:813–827. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Seeber A, Leitner C, Philipp-Abbrederis K, Spizzo G and Kocher F: What's new in small cell lung cancer-extensive disease? An overview on advances of systemic treatment in 2016. Future Oncol. 13:1427–1435. 2017. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

George J, Lim JS, Jang SJ, Cun Y, Ozretić L, Kong G, Leenders F, Lu X, Fernández-Cuesta L, Bosco G, et al: Comprehensive genomic profiles of small cell lung cancer. Nature. 524:47–53. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Pleasance ED, Stephens PJ, O'Meara S, McBride DJ, Meynert A, Jones D, Lin ML, Beare D, Lau KW, Greenman C, et al: A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature. 463:184–190. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Kung JT, Colognori D and Lee JT: Long noncoding RNAs: Past, present, and future. Genetics. 193:651–669. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Ulitsky I and Bartel DP: LincRNAs: Genomics, evolution, and mechanisms. Cell. 154:26–46. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Gibb EA, Brown CJ and Lam WL: The functional role of long non-coding RNA in human carcinomas. Mol Cancer. 10:382011. View Article : Google Scholar : PubMed/NCBI

14 

Huang J, Peng J and Guo L: Non-coding RNA: A new tool for the diagnosis, prognosis, and therapy of small cell lung cancer. J Thorac Oncol. 10:28–37. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Kwok ZH and Tay Y: Long noncoding RNAs: Lincs between human health and disease. Biochem Soc Trans. 45:805–812. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Ohno S: So much ‘junk’ DNA in our genome. Brookhaven Symp Biol. 23:366–370. 1972.PubMed/NCBI

17 

Spornraft M, Kirchner B, Pfaffl MW and Riedmaier I: Comparison of the miRNome and piRNome of bovine blood and plasma by small RNA sequencing. Biotechnol Lett. 37:1165–1176. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Busch H, Reddy R, Rothblum L and Choi YC: SnRNAs, SnRNPs, and RNA processing. Annu Rev Biochem. 51:617–654. 1982. View Article : Google Scholar : PubMed/NCBI

19 

Ota T, Suzuki Y, Nishikawa T, Otsuki T, Sugiyama T, Irie R, Wakamatsu A, Hayashi K, Sato H, Nagai K, et al: Complete sequencing and characterization of 21,243 full-length human cDNAs. Nat Genet. 36:40–45. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Bertone P, Stolc V, Royce TE, Rozowsky JS, Urban AE, Zhu X, Rinn JL, Tongprasit W, Samanta M, Weissman S, et al: Global identification of human transcribed sequences with genome tiling arrays. Science. 306:2242–2246. 2004. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Hon CC, Ramilowski JA, Harshbarger J, Bertin N, Rackham OJ, Gough J, Denisenko E, Schmeier S, Poulsen TM, Severin J, et al: An atlas of human long non-coding RNAs with accurate 5′ ends. Nature. 543:199–204. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Hombach S and Kretz M: Non-coding RNAs: Classification, biology and functioning. Adv Exp Med Biol. 937:3–17. 2016. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Wang KC and Chang HY: Molecular mechanisms of long noncoding RNAs. Mol Cell. 43:904–914. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Wang KC, Helms JA and Chang HY: Regeneration, repair and remembering identity: The three Rs of Hox gene expression. Trends Cell Biol. 19:268–275. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E, et al: 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

28 

Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD, Horlings HM, Shah N, Umbricht C, Wang P, et al: Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet. 43:621–629. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Plath K, Mlynarczyk-Evans S, Nusinow DA and Panning B: Xist RNA and the mechanism of X chromosome inactivation. Annu Rev Genet. 36:233–278. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Lee JT: The X as model for RNA's niche in epigenomic regulation. Cold Spring Harb Perspect Biol. 2:a0037492010. View Article : Google Scholar : PubMed/NCBI

31 

Wutz A, Rasmussen TP and Jaenisch R: Chromosomal silencing and localization are mediated by different domains of Xist RNA. Nat Genet. 30:167–174. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Zhao J, Ohsumi TK, Kung JT, Ogawa Y, Grau DJ, Sarma K, Song JJ, Kingston RE, Borowsky M and Lee JT: Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol Cell. 40:939–953. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Morales Rivea D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, et al: Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci USA. 106:11667–11672. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Spitale RC, Tsai MC and Chang HY: RNA templating the epigenome: Long noncoding RNAs as molecular scaffolds. Epigenetics. 6:539–543. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Collins K: Physiological assembly and activity of human telomerase complexes. Mech Ageing Dev. 129:91–98. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Chen JL and Greider CW: Telomerase RNA structure and function: Implications for dyskeratosis congenita. Trends Biochem Sci. 29:183–192. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Moretti F, Thermann R and Hentze MW: Mechanism of translational regulation by miR-2 from sites in the 5′ untranslated region or the open reading frame. RNA. 16:2493–2502. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Ørom UA, Nielsen FC and Lund AH: MicroRNA-10a binds the 5′UTR of ribosomal protein mRNAs and enhances their translation. Mol Cell. 30:460–471. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Hu X, Feng Y, Zhang D, Zhao SD, Hu Z, Greshock J, Zhang Y, Yang L, Zhong X, Wang LP, et al: A functional genomic approach identifies FAL1 as an oncogenic long noncoding RNA that associates with BMI1 and represses p21 expression in cancer. Cancer Cell. 26:344–357. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Yin Y, Yan P, Lu J, Song G, Zhu Y, Li Z, Zhao Y, Shen B, Huang X, Zhu H, et al: Opposing roles for the lncRNA Haunt and its genomic locus in regulating HOXA gene activation during embryonic stem cell differentiation. Cell Stem Cell. 16:504–516. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A and Bozzoni I: A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 147:358–369. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, et al: The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell. 39:925–938. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, et al: A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 25:666–681. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Steck E, Boeuf S, Gabler J, Werth N, Schnatzer P, Diederichs S and Richter W: Regulation of H19 and its encoded microRNA-675 in osteoarthritis and under anabolic and catabolic in vitro conditions. J Mol Med. 90:1185–1195. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Houseley J, Rubbi L, Grunstein M, Tollervey D and Vogelauer M: A ncRNA modulates histone modification and mRNA induction in the yeast GAL gene cluster. Mol Cell. 32:685–695. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Hainer SJ, Gu W, Carone BR, Landry BD, Rando OJ, Mello CC and Fazzio TG: Suppression of pervasive noncoding transcription in embryonic stem cells by esBAF. Genes Dev. 29:362–378. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Berghoff EG, Clark MF, Chen S, Cajigas I, Leib DE and Kohtz JD: Evf2 (Dlx6as) lncRNA regulates ultraconserved enhancer methylation and the differential transcriptional control of adjacent genes. Development. 140:4407–4416. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Geisler S and Coller J: RNA in unexpected places: Long non-coding RNA functions in diverse cellular contexts. Nat Rev Mol Cell Biol. 14:699–712. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Chen G, Wang Z, Wang D, Qiu C, Liu M, Chen X, Zhang Q, Yan G and Cui Q: LncRNADisease: A database for long-non-coding RNA-associated diseases. Nucleic Acids Res. 41:D983–D986. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Lian Y, Cai Z, Gong H, Xue S, Wu D and Wang K: HOTTIP: A critical oncogenic long non-coding RNA in human cancers. Mol biosyst. 12:3247–3253. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, Lajoie BR, Protacio A, Flynn RA, Gupta RA, et al: A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature. 472:120–124. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Chang S, Liu J, Guo S, He S, Qiu G, Lu J, Wang J, Fan L, Zhao W and Che X: HOTTIP and HOXA13 are oncogenes associated with gastric cancer progression. Oncol Rep. 35:3577–3585. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Quagliata L, Matter MS, Piscuoglio S, Arabi L, Ruiz C, Procino A, Kovac M, Moretti F, Makowska Z, Boldanova T, et al: Long noncoding RNA HOTTIP/HOXA13 expression is associated with disease progression and predicts outcome in hepatocellular carcinoma patients. Hepatology. 59:911–923. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Li Z, Zhao X, Zhou Y, Liu Y, Zhou Q, Ye H, Wang Y, Zeng J, Song Y, Gao W, et al: The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer. J Transl Med. 13:842015. View Article : Google Scholar : PubMed/NCBI

57 

Sun Y, Zhou Y, Bai Y, Wang Q, Bao J, Luo Y, Guo Y and Guo L: A long non-coding RNA HOTTIP expression is associated with disease progression and predicts outcome in small cell lung cancer patients. Mol Cancer. 16:1622017. View Article : Google Scholar : PubMed/NCBI

58 

Sun Y, Hu B, Wang Q, Ye M, Qiu Q, Zhou Y, Zeng F, Zhang X, Guo Y and Guo L: Long non-coding RNA HOTTIP promotes BCL-2 expression and induces chemoresistance in small cell lung cancer by sponging miR-216a. Cell Death Dis. 9:852018. View Article : Google Scholar : PubMed/NCBI

59 

Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E and Chang HY: Long noncoding RNA as modular scaffold of histone modification complexes. Science. 329:689–693. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Sun G, Alzayady K, Stewart R, Ye P, Yang S, Li W and Shi Y: Histone demethylase LSD1 regulates neural stem cell proliferation. Mol Cell Biol. 30:1997–2005. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Mercer TR, Dinger ME and Mattick JS: Long non-coding RNAs: Insights into functions. Nat Rev Genet. 10:155–159. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Tsai MC, Spitale RC and Chang HY: Long intergenic noncoding RNAs: New links in cancer progression. Cancer Res. 71:3–7. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Alves Pádua C, Fonseca AS, Muys BR, de Barros E Lima, Bueno R, Bürger MC, de Souza JE, Valente V, Zago MA and Silva WA Jr: Brief report: The lincRNA Hotair is required for epithelial-to-mesenchymal transition and stemness maintenance of cancer cell lines. Stem Cells. 31:2827–2832. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Yoon JH, Abdelmohsen K, Kim J, Yang X, Martindale JL, Tominaga-Yamanaka K, White EJ, Orjalo AV, Rinn JL, Kreft SG, et al: Scaffold function of long non-coding RNA HOTAIR in protein ubiquitination. Nat Commun. 4:29392013. View Article : Google Scholar : PubMed/NCBI

66 

Ono H, Motoi N, Nagano H, Miyauchi E, Ushijima M, Matsuura M, Okumura S, Nishio M, Hirose T, Inase N, et al: Long noncoding RNA HOTAIR is relevant to cellular proliferation, invasiveness, and clinical relapse in small-cell lung cancer. Cancer Med. 3:632–642. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Fang S, Gao H, Tong Y, Yang J, Tang R, Niu Y, Li M and Guo L: Long noncoding RNA-HOTAIR affects chemoresistance by regulating HOXA1 methylation in small cell lung cancer cells. Lab Invest. 96:60–68. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Young TL, Matsuda T and Cepko CL: The noncoding RNA taurine upregulated gene 1 is required for differentiation of the murine retina. Curr Biol. 15:501–512. 2005. View Article : Google Scholar : PubMed/NCBI

69 

Huang MD, Chen WM, Qi FZ, Sun M, Xu TP, Ma P and Shu YQ: Long non-coding RNA TUG1 is up-regulated in hepatocellular carcinoma and promotes cell growth and apoptosis by epigenetically silencing of KLF2. Mol Cancer. 14:1652015. View Article : Google Scholar : PubMed/NCBI

70 

Ma B, Li M, Zhang L, Huang M, Lei JB, Fu GH, Liu CX, Lai QW, Chen QQ and Wang YL: Upregulation of long non-coding RNA TUG1 correlates with poor prognosis and disease status in osteosarcoma. Tumour Biol. 37:4445–4455. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Liu Q, Sun S, Yu W, Jiang J, Zhuo F, Qiu G, Xu S and Jiang X: Altered expression of long non-coding RNAs during genotoxic stress-induced cell death in human glioma cells. J Neurooncol. 122:283–292. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Han Y, Liu Y, Gui Y and Cai Z: Long intergenic non-coding RNA TUG1 is overexpressed in urothelial carcinoma of the bladder. J Surg Oncol. 107:555–559. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Xu Y, Wang J, Qiu M and Xu L, Li M, Jiang F, Yin R and Xu L: Upregulation of the long noncoding RNA TUG1 promotes proliferation and migration of esophageal squamous cell carcinoma. Tumour Biol. 36:1643–1651. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Zhang E, He X, Yin D, Han L, Qiu M, Xu T, Xia R, Xu L, Yin R and De W: Increased expression of long noncoding RNA TUG1 predicts a poor prognosis of gastric cancer and regulates cell proliferation by epigenetically silencing of p57. Cell Death Dis. 7:e21092016. View Article : Google Scholar : PubMed/NCBI

75 

Yang L, Lin C, Liu W, Zhang J, Ohgi KA, Grinstein JD, Dorrestein PC and Rosenfeld MG: ncRNA- and Pc2 methylation-dependent gene relocation between nuclear structures mediates gene activation programs. Cell. 147:773–788. 2011. View Article : Google Scholar : PubMed/NCBI

76 

Niu Y, Ma F, Huang W, Fang S, Li M, Wei T and Guo L: Long non-coding RNA TUG1 is involved in cell growth and chemoresistance of small cell lung cancer by regulating LIMK2b via EZH2. Mol Cancer. 16:52017. View Article : Google Scholar : PubMed/NCBI

77 

Ling H, Spizzo R, Atlasi Y, Nicoloso M, Shimizu M, Redis RS, Nishida N, Gafà R, Song J, Guo Z, et al: CCAT2, a novel noncoding RNA mapping to 8q24, underlies metastatic progression and chromosomal instability in colon cancer. Genome Res. 23:1446–1461. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Gong WJ, Yin JY, Li XP, Fang C, Xiao D, Zhang W, Zhou HH, Li X and Liu ZQ: Association of well-characterized lung cancer lncRNA polymorphisms with lung cancer susceptibility and platinum-based chemotherapy response. Tumour Biol. 37:8349–8358. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Xin Y, Li Z, Zheng H, Chan MTV and Ka Kei Wu W: CCAT2: A novel oncogenic long non-coding RNA in human cancers. Cell Prolif. 50:2017. View Article : Google Scholar

80 

Redis RS, Sieuwerts AM, Look MP, Tudoran O, Ivan C, Spizzo R, Zhang X, de Weerd V, Shimizu M, Ling H, et al: CCAT2, a novel long non-coding RNA in breast cancer: Expression study and clinical correlations. Oncotarget. 4:1748–1762. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Cai Y, He J and Zhang D: Long noncoding RNA CCAT2 promotes breast tumor growth by regulating the Wnt signaling pathway. Onco Targets Ther. 8:2657–2664. 2015.PubMed/NCBI

82 

Huang S, Qing C, Huang Z and Zhu Y: The long non-coding RNA CCAT2 is up-regulated in ovarian cancer and associated with poor prognosis. Diagn Pathol. 11:492016. View Article : Google Scholar : PubMed/NCBI

83 

Zheng J, Zhao S, He X, Zheng Z, Bai W, Duan Y, Cheng S, Wang J, Liu X and Zhang G: The up-regulation of long non-coding RNA CCAT2 indicates a poor prognosis for prostate cancer and promotes metastasis by affecting epithelial-mesenchymal transition. Biochem Biophys Res Commun. 480:508–514. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Wang CY, Hua L, Yao KH, Chen JT, Zhang JJ and Hu JH: Long non-coding RNA CCAT2 is up-regulated in gastric cancer and associated with poor prognosis. Int J Clin Exp Pathol. 8:779–785. 2015.PubMed/NCBI

85 

Chen S, Wu H, Lv N, Wang H, Wang Y, Tang Q, Shao H and Sun C: LncRNA CCAT2 predicts poor prognosis and regulates growth and metastasis in small cell lung cancer. Biomed Pharmacother. 82:583–588. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Webb E, Adams JM and Cory S: Variant (6;15) translocation in a murine plasmacytoma occurs near an immunoglobulin kappa gene but far from the myc oncogene. Nature. 312:777–779. 1984. View Article : Google Scholar : PubMed/NCBI

87 

Cory S, Graham M, Webb E, Corcoran L and Adams JM: Variant (6;15) translocations in murine plasmacytomas involve a chromosome 15 locus at least 72 kb from the c-myc oncogene. EMBO J. 4:675–681. 1985.PubMed/NCBI

88 

Colombo T, Farina L, Macino G and Paci P: PVT1: A rising star among oncogenic long noncoding RNAs. Biomed Res Int. 2015:3042082015. View Article : Google Scholar : PubMed/NCBI

89 

Ding J, Li D, Gong M, Wang J, Huang X, Wu T and Wang C: Expression and clinical significance of the long non-coding RNA PVT1 in human gastric cancer. Onco Targets Ther. 7:1625–1630. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Wang F, Yuan JH, Wang SB, Yang F, Yuan SX, Ye C, Yang N, Zhou WP, Li WL, Li W, et al: Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology. 60:1278–1290. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Zhou Q, Chen J, Feng J and Wang J: Long noncoding RNA PVT1 modulates thyroid cancer cell proliferation by recruiting EZH2 and regulating thyroid-stimulating hormone receptor (TSHR). Tumour Biol. 37:3105–3113. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Huang C, Yu W, Wang Q, Cui H, Wang Y, Zhang L, Han F and Huang T: Increased expression of the lncRNA PVT1 is associated with poor prognosis in pancreatic cancer patients. Minerva Med. 106:143–149. 2015.PubMed/NCBI

93 

Cui D, Yu CH, Liu M, Xia QQ, Zhang YF and Jiang WL: Long non-coding RNA PVT1 as a novel biomarker for diagnosis and prognosis of non-small cell lung cancer. Tumour Biol. 37:4127–4134. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Cui M, You L, Ren X, Zhao W, Liao Q and Zhao Y: Long non-coding RNA PVT1 and cancer. Biochem Biophys Res Commun. 471:10–14. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Huang C, Liu S, Wang H, Zhang Z, Yang Q and Gao F: LncRNA PVT1 overexpression is a poor prognostic biomarker and regulates migration and invasion in small cell lung cancer. Am J Transl Res. 8:5025–5034. 2016.PubMed/NCBI

96 

Sang Y, Zhou F, Wang D, Bi X, Liu X, Hao Z, Li Q and Zhang W: Up-regulation of long non-coding HOTTIP functions as an oncogene by regulating HOXA13 in non-small cell lung cancer. Am J Transl Res. 8:2022–2032. 2016.PubMed/NCBI

97 

Zhang GJ, Song W and Song Y: Overexpression of HOTTIP promotes proliferation and drug resistance of lung adenocarcinoma by regulating AKT signaling pathway. Eur Rev Med Pharmacol Sci. 21:5683–5690. 2017.PubMed/NCBI

98 

Yang Y, Jiang C, Yang Y, Guo L, Huang J, Liu X, Wu C and Zou J: Silencing of LncRNA-HOTAIR decreases drug resistance of non-small cell lung cancer cells by inactivating autophagy via suppressing the phosphorylation of ULK1. Biochem Biophys Res Commun. 497:1003–1010. 2018. View Article : Google Scholar : PubMed/NCBI

99 

Liu W, Yin NC, Liu H and Nan KJ: Cav-1 promote lung cancer cell proliferation and invasion through lncRNA HOTAIR. Gene. 641:335–340. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Jiang C, Yang Y, Yang Y, Guo L, Huang J, Liu X, Wu C and Zou J: Long noncoding RNA (lncRNA) HOTAIR affects tumorigenesis and metastasis of non-small cell lung cancer by up-regulating miR-613. Oncol Res. 26:725–734. 2018. View Article : Google Scholar : PubMed/NCBI

101 

Lin PC, Huang HD, Chang CC, Chang YS, Yen JC, Lee CC, Chang WH, Liu TC and Chang JG: Long noncoding RNA TUG1 is downregulated in non-small cell lung cancer and can regulate CELF1 on binding to PRC2. BMC Cancer. 16:5832016. View Article : Google Scholar : PubMed/NCBI

102 

Zhang EB, Yin DD, Sun M, Kong R, Liu XH, You LH, Han L, Xia R, Wang KM, Yang JS, et al: P53-regulated long non-coding RNA TUG1 affects cell proliferation in human non-small cell lung cancer, partly through epigenetically regulating HOXB7 expression. Cell Death Dis. 5:e12432014. View Article : Google Scholar : PubMed/NCBI

103 

Liu H, Zhou G, Fu X, Cui H, Pu G, Xiao Y, Sun W, Dong X, Zhang L, Cao S, et al: Long noncoding RNA TUG1 is a diagnostic factor in lung adenocarcinoma and suppresses apoptosis via epigenetic silencing of BAX. Oncotarget. 8:101899–101910. 2017.PubMed/NCBI

104 

Zhao Z, Wang J, Wang S, Chang H, Zhang T and Qu J: LncRNA CCAT2 promotes tumorigenesis by over-expressed Pokemon in non-small cell lung cancer. Biomed Pharmacother. 87:692–697. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Qiu M, Xu Y, Yang X, Wang J, Hu J, Xu L and Yin R: CCAT2 is a lung adenocarcinoma-specific long non-coding RNA and promotes invasion of non-small cell lung cancer. Tumour Biol. 35:5375–5380. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Wang C, Han C, Zhang Y and Liu F: LncRNA PVT1 regulate expression of HIF1α via functioning as ceRNA for miR199a5p in nonsmall cell lung cancer under hypoxia. Mol Med Rep. 17:1105–1110. 2018.PubMed/NCBI

107 

Li H, Chen S, Liu J, Guo X, Xiang X, Dong T, Ran P, Li Q, Zhu B, Zhang X, et al: Long non-coding RNA PVT1-5 promotes cell proliferation by regulating miR-126/SLC7A5 axis in lung cancer. Biochem Biophys Res Commun. 495:2350–2355. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Guo D, Wang Y, Ren K and Han X: Knockdown of LncRNA PVT1 inhibits tumorigenesis in non-small-cell lung cancer by regulating miR-497 expression. Exp Cell Res. 362:172–179. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Wu D, Li Y, Zhang H and Hu X: Knockdown of lncrna PVT1 enhances radiosensitivity in non-small cell lung cancer by sponging Mir-195. Cell Physiol Biochem. 42:2453–2466. 2017. View Article : Google Scholar : PubMed/NCBI

110 

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:E4732017. View Article : Google Scholar : PubMed/NCBI

111 

Yu X, Zheng H, Chan MT and Wu WK: HULC: An oncogenic long non-coding RNA in human cancer. J Cell Mol Med. 21:410–417. 2017. View Article : Google Scholar : PubMed/NCBI

112 

Gutschner T, Hämmerle M and Diederichs S: MALAT1-a paradigm for long noncoding RNA function in cancer. J Mol Med. 91:791–801. 2013. View Article : Google Scholar : PubMed/NCBI

113 

Huarte M: The emerging role of lncRNAs in cancer. Nat Med. 21:1253–1261. 2015. View Article : Google Scholar : PubMed/NCBI

114 

McCleland ML, Mesh K, Lorenzana E, Chopra VS, Segal E, Watanabe C, Haley B, Mayba O, Yaylaoglu M, Gnad F, et al: CCAT1 is an enhancer-templated RNA that predicts BET sensitivity in colorectal cancer. J Clin Invest. 126:639–652. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Zhou Y, Zhang X and Klibanski A: MEG3 noncoding RNA: A tumor suppressor. J Mol Endocrinol. 48:R45–R53. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Yap TA, Omlin A and de Bono JS: Development of therapeutic combinations targeting major cancer signaling pathways. J Clin Oncol. 31:1592–1605. 2013. View Article : Google Scholar : PubMed/NCBI

117 

Liu XH, Sun M, Nie FQ, Ge YB, Zhang EB, Yin DD, Kong R, Xia R, Lu KH, Li JH, et al: Lnc RNA HOTAIR functions as a competing endogenous RNA to regulate HER2 expression by sponging miR-331-3p in gastric cancer. Mol Cancer. 13:922014. View Article : Google Scholar : PubMed/NCBI

118 

Zhang SR, Yang JK, Xie JK and Zhao LC: Long noncoding RNA HOTTIP contributes to the progression of prostate cancer by regulating HOXA13. Cell Mol Biology. 62:84–88. 2016.

119 

Sørensen KP, Thomassen M, Tan Q, Bak M, Cold S, Burton M, Larsen MJ and Kruse TA: Long non-coding RNA HOTAIR is an independent prognostic marker of metastasis in estrogen receptor-positive primary breast cancer. Breast Cancer Res Treat. 142:529–536. 2013. View Article : Google Scholar : PubMed/NCBI

120 

Wu ZH, Wang XL, Tang HM, Jiang T, Chen J, Lu S, Qiu GQ, Peng ZH and Yan DW: Long non-coding RNA HOTAIR is a powerful predictor of metastasis and poor prognosis and is associated with epithelial-mesenchymal transition Rep. 32:395–402. 2014.

121 

Svoboda M, Slyskova J, Schneiderova M, Makovicky P, Bielik L, Levy M, Lipska L, Hemmelova B, Kala Z, Protivankova M, et al: HOTAIR long non-coding RNA is a negative prognostic factor not only in primary tumors, but also in the blood of colorectal cancer patients. Carcinogenesis. 35:1510–1515. 2014. View Article : Google Scholar : PubMed/NCBI

122 

Kogo R, Shimamura T, Mimori K, Kawahara K, Imoto S, Sudo T, Tanaka F, Shibata K, Suzuki A, Komune S, et al: Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res. 71:6320–6326. 2011. View Article : Google Scholar : PubMed/NCBI

123 

Huang L, Liao LM, Liu AW, Wu JB, Cheng XL, Lin JX and Zheng M: Overexpression of long noncoding RNA HOTAIR predicts a poor prognosis in patients with cervical cancer. Arch Gynecol Obstet. 290:717–723. 2014. View Article : Google Scholar : PubMed/NCBI

124 

Ji F, Wuerkenbieke D, He Y and Ding Y: Long noncoding RNA HOTAIR: An oncogene in human cervical cancer interacting with MicroRNA-17-5p. Oncol Res. 2017.Doi: 10.3727/096504017X15002869385155. View Article : Google Scholar :

125 

Wu X, Cao X and Chen F: LncRNA-HOTAIR activates tumor cell proliferation and migration by suppressing MiR-326 in cervical cancer. Oncol Res. 2017.Doi: 10.3727/096504017X15037515496840. View Article : Google Scholar :

126 

Endo H, Shiroki T, Nakagawa T, Yokoyama M, Tamai K, Yamanami H, Fujiya T, Sato I, Yamaguchi K, Tanaka N, et al: Enhanced expression of long non-coding RNA HOTAIR is associated with the development of gastric cancer. PLoS One. 8:e770702013. View Article : Google Scholar : PubMed/NCBI

127 

Hajjari M, Behmanesh M, Sadeghizadeh M and Zeinoddini M: Up-regulation of HOTAIR long non-coding RNA in human gastric adenocarcinoma tissues. Med Oncol. 30:6702013. View Article : Google Scholar : PubMed/NCBI

128 

Chen WM, Chen WD, Jiang XM, Jia XF, Wang HM, Zhang QJ, Shu YQ and Zhao HB: HOX transcript antisense intergenic RNA represses E-cadherin expression by binding to EZH2 in gastric cancer. World J Gastroenterol. 23:6100–6110. 2017. View Article : Google Scholar : PubMed/NCBI

129 

Yan J, Dang Y, Liu S, Zhang Y and Zhang G: LncRNA HOTAIR promotes cisplatin resistance in gastric cancer by targeting miR-126 to activate the PI3K/AKT/MRP1 genes. Tumour Biol. Nov 30–2016.(Epub ahead of print). View Article : Google Scholar :

130 

Lin YH, Wu MH, Huang YH, Yeh CT, Cheng ML, Chi HC, Tsai CY, Chung IH, Chen CY and Lin KH: Taurine upregulated gene 1 functions as a master regulator to coordinate glycolysis and metastasis in hepatocellular carcinoma. Hepatology. 67:188–203. 2018. View Article : Google Scholar : PubMed/NCBI

131 

Zhao L, Sun H, Kong H, Chen Z, Chen B and Zhou M: The lncrna-TUG1/EZH2 axis promotes pancreatic cancer cell proliferation, migration and EMT phenotype formation through sponging mir-382. Cell Physiol Biochem. 42:2145–2158. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Liu Q, Liu H, Cheng H, Li Y, Li X and Zhu C: Downregulation of long noncoding RNA TUG1 inhibits proliferation and induces apoptosis through the TUG1/miR-142/ZEB2 axis in bladder cancer cells. Onco Targets Ther. 10:2461–2471. 2017. View Article : Google Scholar : PubMed/NCBI

133 

Yu Y, Nangia-Makker P, Farhana L and Majumdar AP: A novel mechanism of lncRNA and miRNA interaction: CCAT2 regulates miR-145 expression by suppressing its maturation process in colon cancer cells. Mol Cancer. 16:1552017. View Article : Google Scholar : PubMed/NCBI

134 

Hua F, Li CH, Chen XG and Liu XP: Long noncoding RNA CCAT2 Knockdown suppresses tumorous progression by sponging miR-424 in epithelial ovarian cancer. Oncol Res. 26:241–247. 2018. View Article : Google Scholar : PubMed/NCBI

135 

Li T, Meng XL and Yang WQ: Long noncoding RNA PVT1 acts as a ‘Sponge’ to inhibit microRNA-152 in gastric cancer cells. Dig Dis Sci. 62:3021–3028. 2017. View Article : Google Scholar : PubMed/NCBI

136 

Huang T, Liu HW, Chen JQ, Wang SH, Hao LQ, Liu M and Wang B: The long noncoding RNA PVT1 functions as a competing endogenous RNA by sponging miR-186 in gastric cancer. Biomed Pharmacother. 88:302–308. 2017. View Article : Google Scholar : PubMed/NCBI

137 

Zhao L, Kong H, Sun H, Chen Z, Chen B and Zhou M: LncRNA-PVT1 promotes pancreatic cancer cells proliferation and migration through acting as a molecular sponge to regulate miR-448. J Cell Physiol. 233:4044–4055. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 40 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li TT, He RQ, Ma J, Li ZY, Hu XH and Chen G: Long non‑coding RNAs in small cell lung cancer: A potential opening to combat the disease (Review). Oncol Rep 40: 1831-1842, 2018
APA
Li, T., He, R., Ma, J., Li, Z., Hu, X., & Chen, G. (2018). Long non‑coding RNAs in small cell lung cancer: A potential opening to combat the disease (Review). Oncology Reports, 40, 1831-1842. https://doi.org/10.3892/or.2018.6635
MLA
Li, T., He, R., Ma, J., Li, Z., Hu, X., Chen, G."Long non‑coding RNAs in small cell lung cancer: A potential opening to combat the disease (Review)". Oncology Reports 40.4 (2018): 1831-1842.
Chicago
Li, T., He, R., Ma, J., Li, Z., Hu, X., Chen, G."Long non‑coding RNAs in small cell lung cancer: A potential opening to combat the disease (Review)". Oncology Reports 40, no. 4 (2018): 1831-1842. https://doi.org/10.3892/or.2018.6635