Interaction of long-chain non-coding RNAs and important signaling pathways on human cancers (Review)

  • Authors:
    • Wei Sun
    • Ying Shi
    • Zhifei Wang
    • Jiye Zhang
    • Hanhui Cai
    • Jungang Zhang
    • Dongsheng Huang
  • View Affiliations

  • Published online on: September 27, 2018     https://doi.org/10.3892/ijo.2018.4575
  • Pages: 2343-2355
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Long non-coding RNAs (lncRNAs) usually refer to non-coding RNA transcripts >200 nucleotides in length. In terms of the full genomic transcript, the proportion of lncRNAs far exceeds that of coding RNA. Initially, lncRNAs were considered to be the transcriptional noise of genes, but it has since been demonstrated that lncRNAs serve an important role in the regulation of cellular activities through interaction with DNA, RNA and protein. Numerous studies have demonstrated that various intricate signaling pathways are closely related to lncRNAs. Here, we focus on a large number of studies regarding the interaction of lncRNAs with important signaling pathways. It is comprehensively illustrated that lncRNAs regulate key metabolic components and regulatory factors of signaling pathways to affect the biological activities of tumor cells. Evidence suggests that the abnormal expression or mutation of lncRNAs in human tumor cells, and their interaction with signaling pathways, may provide a basis and potential target for the diagnosis and treatment of human cancers.

1. Introduction

The human genome project has demonstrated that <2% nucleic acid sequences are protein-coding. In 2002, Okazaki et al (1) first identified a class of longer transcripts by sequencing a mouse cDNA library, and termed them long non-coding RNAs (lncRNAs). Due to limitations in our understanding of gene regulation and expression, these non-coding RNAs were considered to be transcriptional noise until HOX transcript antisense RNA (HOTAIR) was identified. HOTAIR is a lncRNA located on the antisense strand of the HOXC gene cluster, which regulates the expression of the HOXD gene and the occurrence of H3K4me2 methylation modifications in certain gene regions, and silences expression at this site (2,3). Although lncRNAs do not encode proteins, they participate in important cellular physiological processes. Numerous studies have reported that lncRNAs are abnormally expressed in various types of tumor, and affect the occurrence and development of tumors through different signaling pathways. lncRNAs are generally defined as long-stranded RNAs with >200 nucleotides, but this is not absolute. For example, BC1 and snaR are <200 nucleotides in length (4). lncRNAs may also be classified according to location, object of action and functional mechanism (5). The progress in lncRNA research is associated with the progress in research technology. It is worth mentioning that RNA-sequencing technology, proposed by the Snyder team at Yale University, has low background noise and is able to detect single-base differences between similar genes or different transcripts caused by variable cleavage in the gene family, as well as low abundance transcripts (6). At present, next-generation sequencing technologies have provided the largest experimental evidence on lncRNA, with the advantages of high flux, short duration, high accuracy and abundant information.

lncRNAs participate in the regulation of various biological activities, including genomic imprinting, X-chromosome inactivation, chromosome modification and telomere biology. They are also associated with the occurrence and development of various human diseases, particularly with the occurrence and development of malignant tumors. Their long ribbon-like molecular structure interacts with multiple types of molecules, thus affecting cell biological activities. Signaling pathways participate in the transduction of cell signals through a series of enzymatic reactions, which are regulated by various types of molecules. lncRNA is an important regulator of signaling pathways, and regulates gene expression at a transcriptional, post-transcriptional and epigenetic level. This review focused on the function of lncRNA in several classical signaling pathways.

2. lncRNA and the p53 signaling pathway

The p53 gene was first reported in 1979 by Lane and Crawford (7), and >50% cancer types have been found to be related to mutations of the p53 gene. A tight and precise regulatory system regulates the activities of p53-associated signaling pathways to prevent abnormal changes in p53 levels from harming cells. lncRNA has gained increasing attention as an important regulator of p53 signaling.

The damage induced noncoding (lncRNA-DINO) is a DNA damage-activated transcriptional lncRNA involved in the lncRNA-directed biological DNA damage response. Numerous abnormal changes in the p53 pathway may increase the probability of tumorigenesis. DINO interacts with the p53 protein to promote its stability, which causes protein accumulation, activation of p53 target proteins and regulates a p53 auto-enhancement loop (Fig. 1). DINO is involved in p53-dependent gene expression, cell cycle arrest and apoptosis. In the absence of DNA damage, artificially upregulating DINO expression can effectively activate the DNA damage response pathway and cell cycle arrest. However, when artificially inhibiting the expression of DINO, the response of cells to p53 signaling is weakened. In mice, inactivation of the DINO gene or the promoter blocks the p53 pathway and improves acute radiation syndrome in vivo (8). Thus, DNA damage-induced lncRNAs form a feedback loop with homologous transcription factors to enhance cell signaling networks. A genome-wide transcriptome analysis of human diploid fibroblasts revealed that lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT1) is involved in regulating the expression of cell cycle-associated genes, and is required for G1/S and mitotic progression. Low expression of MALAT1 leads to the activation of p53 and its target genes, and cell cycle defects observed in MALAT1-depleted cells are sensitive to p53 expression levels, indicating that p53 is a key downstream mediator of MALAT1. Furthermore, reduced expression of an oncogenic transcription factor, B-MYB, which is involved in G2/M progression, is expressed in MALAT1-depleted cells due to altered binding of splicing factors on B-MYB pre-mRNA and aberrant alternative splicing (9). These findings reveal the mechanism by which lncRNA regulates cell proliferation.

lncRNAs also regulate p53, subsequently affecting cell cycle progression, for example via lncRNA regulator of reprogramming (lncRNA-ROR). Unlike MDM2, which leads to p53 degradation through the ubiquitin-proteasome pathway, it directly inhibits p53 through interactions with heterogeneous nuclear ribonucleoprotein I (hnRNP I) at the translational level. ROR carries an hnRNP I binding domain that acts on the 5'-untranslated region of p53 mRNA to suppress the translation of p53, thereby inhibiting p53-mediated cell cycle arrest and apoptosis. The induction of p53 following treatment with doxorubicin increases ROR expression levels, and the ectopic expression of p53 induces ROR production, resulting in a ROR-p53 auto-regulatory feedback loop (10). Similarly to mRNA, the major transcriptional processes of lncRNA are regulated by various factors, including p53, NF-κB, Sox2, Oct4 and Nanog (11). For example, the transcriptional promoter region of ROR is directly affected by pluripotency factors, OCT4, SOX2 and KLF4 (12). lncRNA-p21 binding to hnRNP-K assists hnRNP-K to localize and inhibit p53-regulated genes, and is a p53-dependent transcriptional response inhibitor (13). Competing endogenous RNAs (ceRNAs) regulate gene expression through competitive binding of microRNA. By providing ceRNA or natural microRNA sponge-like functions, lncRNAs are important post-transcriptional regulators of gene expression, regulating microRNAs and competitively inhibiting important receptors involved in various cellular biological activities (14). For example, lncRNA-loc285194, growth-arrest specific 5 (GAS5) and HOTAIR appear to function as competitive endogenous RNAs participating in the inhibition of tumor development (15-17). GAS5 also has the ability to simulate DNA and compete with steroid receptors in cells, which regulates steroid-mediated transcriptional regulation, growth arrest and apoptosis (18). In human meningioma cells, lncRNA maternally expressed gene 3 (MEG3) stimulates p53-mediated transactivation and inhibits tumor proliferation by inducing apoptosis (19), which may be associated with its ability to downregulate MDM2 expression as demonstrated by Zhou et al (20). Certain natural antisense transcripts are important lncRNAs that may be paired with complementary RNAs to form sense-antisense double-stranded RNAs. This causes the degradation or translational inhibition of target mRNA. For example, lncRNA-Wrap53 is a natural antisense transcript of p53. The highly conserved Wrap53 has been demonstrated to regulate endogenous p53 mRNA levels and further induce p53 protein expression by targeting the 5'-untranslated region of p53 mRNA (21). p21-associated lncRNA DNA-damage activated (PANDA) is an lncRNA located ~5 kB upstream of the CDKN1A TSS gene, and is induced by specific DNA damage. PANDA is also a p53-associated regulatory lncRNA involved in cell cycle progression and apoptosis (22). The ectopic expression of lncRNA is involved in numerous disease processes, but the underlying molecular mechanism is not yet fully understood. Table I lists the lncRNAs that interact with the p53 pathway in various types of human cancer.

Table I

lncRNAs interact with p53 signaling pathway in cancers.

Table I

lncRNAs interact with p53 signaling pathway in cancers.

Author, yearlncRNAAssociated diseaseFunctionEffect on pathway(Refs.)
Liu et al, 2013LOC285194OsteosarcomaInhibit tumor cell growthPositive(15)
Tano et al, 2017MALAT1Lung adenocarcinomaInhibit cell cycle arrestNegative(90)
Yang et al, 2016RORColorectal cancerDecrease sensitivity to radiotherapyNegative(91)
Chen et al, 2016
Zhu et al, 2015
MEG3HepatomaInhibit proliferation and apoptosisPositive(92,95)
Li et al, 2016MEG3GliomaInhibit tumorigenesisPositive(93)
Lu et al, 2013MEG3Non-small cell lung cancerInhibit proliferation and apoptosisPositive(94)
Sun et al, 2016MEG3Breast cancerInhibit proliferation, metastasis and invasionPositive(96)
Zhang et al, 2014TUG1Non-small cell lung cancerInhibit tumorigenesisPositive(97)
Huang et al, 2015UCA1Breast cancerPromote tumorigenesisNegative(98)
Zhai et al, 2016HOTAIRNon-small cell lung cancerPromote proliferation and invasionNegative(99)
Su et al, 2017PRALLung cancerInhibit proliferationPositive(100)
Gong et al, 2014LOC401317Nasopharyngeal carcinomaInhibit proliferationPositive(101)
Zhai et al, 2015ASLNC04080Endometrial carcinomaPromote proliferation and invasionNegative(102)
Thorenoor et al, 2016ZFAS1Colorectal cancerPromote proliferationNegative(103)

3. lncRNA and the NF-κB signaling pathway

NF-κB is a widely expressed pleiotropic transcription factor. It is a heterotrimer composed of p50, p65 and IκB. The NF-κB signaling pathway is activated by a variety of extracellular stimuli, serving a central role in transcriptional regulation, and in the expression and regulation of various genes. It is a marker of cell activation, and mediates a variety of biological processes, including inflammation, cell proliferation and tumor metastasis. The NF-κB pathway is regulated by various lncRNAs.

lncRNA-Lethe was the first lncRNA identified to be involved in regulation of the NF-κB signaling pathway. In human 293 cells, Lethe is selectively induced by pro-inflammatory cytokines through the NF-κB pathway, and acts on RelA (the p65 subunit of NF-κB) to inhibit RelA-DNA binding and subsequently target gene activation (23). Lethe has an active role in regulating the NF-κB pathway and is involved in inflammation through a typical negative feedback mechanism. Activated NF-κB promotes the expression of Lethe, and Lethe directly inhibits this pathway by interacting with RelA, thereby reducing the production of various inflammatory proteins (23). A novel lncRNA, namely nuclear transcription factor NF-κB interacting lncRNA (NKILA), which participates in the inhibition of breast cancer metastasis through the NF-κB signaling pathway, was discovered by analyzing a large number of lncRNAs in human breast cancer (24). NF-κB upregulates the expression of NKILA by binding to the promoter of NKILA, and a large quantity of NKILA is able to bind to NF-κB/IκB to form a stable complex, thus directly masking the phosphorylation site of IκB to prevent IKK-mediated phosphorylation of IκB and activation of NF-κB. The negative feedback of NKILA and NF-κB serves a key role in preventing overactivation of the NF-κB pathway in inflammation-stimulated mammary epithelial cells. However, highly expressed microRNA-103/107 mediates the degradation of NKILA, which results in overactivation of the NF-κB pathway, thus contributing to cancer metastasis and poor patient outcome (24). As a key enzyme in prostaglandin synthesis, COX-2 serves an important role in the prognosis of tumors, particularly neoangiogenesis and tumor progression (25,26). lncRNA p50-associated COX-2 extragenic RNA (PACER), localized in the nucleus, binds directly to free p50, inhibits the formation of homodimers, promotes the formation of heterodimers, increases the transcription enhancement effect and increases the expression of COX-2 (27). Pearson et al (28) achieved results, which support these observations, whereby primary human osteoarthritic chondrocytes treated with IL-1β rapidly induced the formation of PACER, indicating that PACER expression is associated with COX2. In addition to PACER, the lncRNA cyclooxygenase 2 (COX2) locus near the COX2 gene is essential for the transcription of NF-κB-associated late inflammatory genes in endotoxin-treated mouse macrophages (29). In the nucleus, MALAT1 interacts with the P65-p50 heterodimer to inhibit downstream TNF/ IL-6 expression. Bioinformatics analysis has revealed that MALAT1 affects NF-κB/RelA activity in the epithelial-mesenchymal transition process (30). The abnormal expression of HOTAIR in ovarian cancer induces NF-κB activation by inhibiting Iκ-Bα in the DNA damage response. It also increases the expression of MMP-9 and IL-6, key target genes of NF-κB, and serves an important role in DNA damage response, cell senescence and chemotherapy resistance (31). In addition, certain lncRNAs regulate signaling pathways indirectly through signaling elements or associated upstream molecules in the NF-κB pathway. For example, MIR31HG is frequently deleted in glioblastoma, and significantly reduces the levels of micro (mi)RNA-31, while loss of miRNA-31 expression enhances NF-κB signaling by targeting TRADD, its upstream activator, and promotes tumor growth (32). C2dat1 promotes neural cell survival by enhancing CAMK2D expression and activating the NF-κB pathway (33). Expression of IL1β-eRNA, IL1β-RBT46, AS-IL1α, ANRIL, THRIL and IL7R is induced by the NF-κB signaling pathway, and regulates the expression of NF-κB-associated target genes (34-38). Table II lists the lncRNAs that interact with the NF-κB pathway in cancer.

Table II

lncRNAs interact with NF-κB signaling pathway in cancers.

Table II

lncRNAs interact with NF-κB signaling pathway in cancers.

Author, yearlncRNAAssociated diseaseFunctionEffect on pathway(Refs.)
Rajbhandari et al, 2015MIR31HGGlioblastomaInhibit tumor growthNegative(32)
Chen et al, 2016MEG3HepatomaInhibit cell apoptosisPositive(92)
Özeş et al, 2016HOTAIROvarian cancerPromote cellular senescence and chemotherapy resistancePositive(31)
Li et al, 2017HOTAIRColorectal cancerPromote 5FU resistancePositive(104)
Liao et al, 2018H19Malignant melanomaPromote the metastasis and invasionPositive(105)
Liu et al, 2015NKILABreast cancerInhibit metastasisNegative(24)
Yang et al, 2018NKILALaryngeal cancerInhibit proliferation and metastasis Enhance radiosensitivityNegative(106)
Bian et al, 2017NKILAMalignant melanomaInhibit invasion and metastasisNegative(107)
Huang et al, 2016NKILATongue squamouscell carcinomaInhibit migration and invasionNegative(108)
Ma et al, 2018DANCRGliomaPromote cisplatin resistancePositive(109)

4. lncRNA and the Wnt signaling pathway

The Wnt gene was first named Int1, because its activation is dependent on the insertion of the mouse breast cancer-associated virus gene (39). Since then, a large number of studies have reported that Int1 serves an important role in the normal embryonic development of mice and is equivalent to the Wingless gene of drosophila. Therefore, combining Wingless with Int1 it was named Wnt. The Wnt signaling pathway is an evolutionarily highly conserved signaling system involved in important physiological processes, including embryonic development, tissue differentiation and cell homeostasis (40,41). Specific lncRNAs associated with the Wnt signaling pathway are discussed below.

Numerous lncRNAs target and affect the accumulation of β-catenin, a key molecule that acts on the Wnt pathway, thereby regulating the expression of Wnt target genes and the function of cancer cells (42,43). Numerous lncRNAs regulate target genes to promote tumorigenesis and development. In breast cancer, patients exhibiting high CCAT2 expression have a significantly poorer prognosis and lower overall survival rate compared with patients with low CCAT2 expression (44). A previous study on MCF-7 and MDA-MB-231 breast cancer cells demonstrated that the suppression of CCAT2 expression decreases the levels of β-catenin in the cytoplasm and nucleus, and reduces the expression of CCND1 and c-myc, which are classic downstream genes of the Wnt/β-catenin signaling pathway. These results indicate the possible role of CCAT2 in breast cancer (44). The abnormal expression of HOTAIR has been demonstrated in certain types of human cancer. In colorectal cancer, Wnt/0205-catenin signaling is inhibited by HOTAIR-knockdown and miR-203a-3p overexpression (45). The downregulation of HOTAIR expression is essential for reducing cell proliferation and chemoresistance via modulation of miR-203a-3p expression levels and the activity of the Wnt/0205-catenin signaling pathway (45). lncRNA-CRNDE is significantly overexpressed in renal cell carcinoma. Elevated CRNDE expression may regulate the PI3K/Akt/GSK3b signaling pathway, increase the level of β-catenin in the nucleus, and ultimately regulate the expression of the Wnt target gene to regulate the proliferation of renal cancer cells (46). lncRNA-TCF7 promotes the self-renewal and proliferation of hepatocellular carcinoma cells by activating the Wnt signaling pathway. It may be that TCF7 recruits SWI/SNF complexes to act on its own promoter to regulate expression and activate the Wnt pathway (47). Similarly, numerous studies have reported that lncRNAs inhibit tumorigenesis. In a large number of non-small cell lung cancer tissue samples, low expression of AK126698 often predicts larger tumor diameter and advanced tumor stage. Conversely, an increase in AK126698 expression targets Frizzled-8, one of the receptors acting of the Wnt/β-catenin signaling pathway, inhibiting the proliferation and metastasis of cancer cells and inducing apoptosis (48). The expression of lncRNA-CTD903 is significantly upregulated in human colorectal cancer and may be used as an independent prognostic factor for colorectal cancer. When CTD903 was knocked down in the RKO and SW480 cell lines, the invasion and metastasis of the tumor cells is increased, and epithelial-mesenchymal transition-like characteristics are exhibited (49). Further results demonstrated that downregulated CTD903 expression allows the Wnt/β-catenin signaling pathway to be more easily activated, increasing the expression of transcription factors, Twist and Snail, as well as the protein expression of the mesenchymal marker, Vimentin, and reducing that of the epithelial marker, ZO-1 (49). Chemotherapy resistance can result in poor prognosis. Furthermore, lncRNAs serve a role in cell resistance. The level of lncRNA-Meg3 is significantly lower in the cisplatin-resistant lung cancer cell line, A549/DDP, compared with that in the A549 cell line (50). Further results demonstrated that the upregulation of Meg3 expression in vitro increases the sensitivity of the A549/DDP lung cancer cell line to cisplatin. However, the sensitivity of A549 cells to cisplatin decreased following RNA interference downregulation of Meg3 expression. Researchers hypothesized that Meg3 mediates increased cell cycle arrest and apoptosis by regulating p53, β-catenin and survivin, resulting in increased sensitivity to chemotherapy (50). In osteosarcoma, HOTTIP induces tumor occurrence and resistance by activating the Wnt signaling pathway, which may be a potential target for the treatment of osteosarcoma (51). Table III lists the lncRNAs that interact with the Wnt signaling pathway and their general roles in cancer.

Table III

lncRNAs that interact with the Wnt signaling pathway in cancer.

Table III

lncRNAs that interact with the Wnt signaling pathway in cancer.

Author, yearlncRNAAssociated diseaseFunctionEffect on pathway(Refs.)
Cai et al, 2015CCAT2Breast cancerPromote cell growth and tumor formationPositive(44)
Shao et al, 2016CRNDERenal cell carcinomaPromote cell proliferationPositive(46)
Wang et al, 2015TCF7Hepatocellular carcinomaPromote liver cancer stem cells self-renewal and tumor propagationPositive(47)
Fu et al, 2016AK126698Non-small cell lung cancerInhibit proliferation and migrationNegative(48)
Yuan et al, 2016CTD903Colorectal cancerInhibit cancer invasion and migrationNegative(49)
Xia et al, 2015MEG3Lung cancerEnhance chemosensitivityNegative(50)
Rajbhandari et al, 2015TUG1Oral squamous cell carcinomaPromote proliferation and metastasisPositive(110)
Fan et al, 2014UCA1Bladder cancerPromote chemoresistancePositive(43)
Yang et al, 2016UCA1Oral squamous cell carcinomaPromote proliferation and metastasisPositive(111)
Xiao et al, 2018HOTAIRColorectal cancerPromote cell proliferation and chemoresistancePositive(45)
Ge et al, 2013HOTAIREsophageal squamous cell cancerPromote metastasis and invasionPositive(112)
Wu et al, 2017H19Colorectal cancerPromote methotrexate resistancePositive(113)
Cao et al, 2017CCAT1Epithelial ovarian cancerPromote metastasis and poor prognosisPositive(114)
Li et al, 2015HOTTIPOsteosarcomaPromote tumor occurrencePositive(51)
Fu et al, 2017HOTTIPPancreatic cancerPromote tumorigenesisPositive(115)
Yue et al, 2018CYTORColon cancerPromote epithelial mesenchymal transformation and metastasisPositive(116)
Liu et al, 2016LETLung adenocarcinomaInhibit tumor growth and epithelial mesenchymal transformationNegative(117)
Wang et al, 2017PTCSC3Papillary thyroid carcinomaInhibit proliferation and metastasisNegative(118)
Zhang et al, 2016CASC11Colorectal cancerPromote proliferation and metastasisPositive(119)
Zhang et al, 2017PCAT1Extrahepatic cholangiocarcinomaPromote tumorigenesisPositive(120)
Wang et al, 2018LINC00968Non-small cell lung cancerPromote tumorigenesisPositive(121)

5. lncRNA and the Notch signaling pathway

The notch signaling pathway is a highly evolutionarily conserved cell signaling pathway. It mediates direct cell-cell contact and affects the normal morphogenesis of cells. It regulates differentiation of multipotent progenitor cells, apoptosis, cell proliferation and the formation of cell borders (52). lncRNA also interacts with the Notch signaling pathway and affects tumor progression.

It has been reported that the non-coding sequences that accompany the genes are often involved in the regulation of peripheral genes. In the human genome, lncRNA-NALT was identified at a distance of ~100 bases from the NOTCH1 gene. In T-cell acute lymphoblastic leukemia cells, increased expression of NALT significantly promotes cell proliferation. It may be that NALT activates transcription in the Notch pathway and regulatory elements in the nucleus (53). lncRNA-SNHG12 expression is significantly elevated in osteosarcoma tissues and cell lines, and predicts poor prognosis. A study of the biological function of SNHG12 in the 143B and U2OS cell lines revealed that the downregulation of SNHG12 inhibits cell proliferation by blocking the G0/G1 phase of the cell cycle, and reduces the potential for cell invasion and metastasis. SNHG12 sponges miR-195-5p in osteosarcoma cells, similarly to endogenously-competent RNA, to regulate Notch2 gene expression, thereby affecting signal transduction in the Notch pathway (54). In a study of CRC specimens, and matched adjacent normal tissues and CRC cell lines, the expression of FOXD2-AS1 was significantly increased in CRC tissues as well as in CRC cell lines, and downregulation of FOXD2-AS1 expression suppressed proliferation, invasion and migration in vitro. Further study examined associated markers of the epithelial-mesenchymal transition and Notch signaling pathways, and confirmed that the two pathways are inactivated in CRC cells following FOXD2-AS1-knockdown (55). When studying the expression profile of lncRNA-MEG3 and the two Notch pathway-associated molecules, Notch1 and Hes1, in human endometrial tissues and cell lines, Guo et al (56) reported that, compared with the normal control group, MEG3 expression is significantly downregulated in cancer tissue. However, the expression levels of Notch1 and Hes1 are significantly increased. Upregulation of MEG3 in tumor cells in vivo inhibits the growth of tumors by inhibiting the Notch pathway (56). The Notch pathway serves an important role in maintaining the pluripotency of glioma stem cells. The activation of Notch1 induces lncRNA-TUG1 expression in specific glioma cell lines. In the cytoplasm, TUG1 promotes cell self-renewal by decoying miR-145. In the nucleus, TUG1 binds YY1 by recruiting multi-comb complexes to allow site-specific H3K27 histones to methylate, thereby inhibiting gene mutation. In addition, intravenous administration of TSG1 with antisense oligonucleotides induces the differentiation of glioma stem cell lines in vivo, and effectively inhibits their proliferation (57). Table IV lists the lncRNAs that interact with the Notch pathway in cancer.

Table IV

lncRNAs that interact with the Notch signaling pathway in cancer.

Table IV

lncRNAs that interact with the Notch signaling pathway in cancer.

Author, yearlncRNAAssociated diseaseFunctionEffect on pathway(Refs.)
Wang et al, 2015NALTPediatric T cell acute lymphoblastic leukemiaPromote proliferationPositive(53)
Zhou et al, 2018SNHG12OsteosarcomaPromote cell invasion and migrationPositive(54)
Yang et al, 2018FOXD2-AS1Colorectal cancerPromote cell proliferation and migrationPositive(55)
Guo et al, 2016MEG3Endometrial carcinomaInhibit proliferationNegative(56)
Cai et al, 2017HOTAIRPancreatic cancerPromote tumor growthPositive(17)
Hang et al, 2015AK022798Gastric cancerPromotes cisplatin-resistantPositive(122)
Lu et al, 2018FAM83H-AS1Colorectal carcinomaPromote proliferationPositive(123)
Chen et al, 201700673Hepatocellular carcinomaPromote proliferation and metastasisPositive(124)

6. lncRNA and the PI3K/AKT signaling pathway

AKT, also known as protein kinase B, is an important crossover molecule in multiple signaling pathways and is activated by various substances, including hormones, growth factors, cytokines and intercellular matrix. The PI3K/AKT signaling pathway is involved in important physiological activities, including cell proliferation and survival, and is associated with cancer, diabetes, rheumatoid arthritis and other diseases (58,59). In recent years, the regulatory mechanism of lncRNAs in the PI3K/AKT pathway and the impact on diseases, have gradually been revealed.

Using a CRISPR/Cas9-based synergistic activation mediator system, Koirala et al (60) studied numerous lncRNAs that may affect AKT activity. They reported that lncRNA-AK023948 is a positive regulator of the AKT pathway. In malignant cells, upregulation of AK023948 and DHX9 expression promotes the activation of the AKT pathway. Furthermore, they may interact with ATP-dependent RNA helicase A (RHA/DHX9) and P58 to participate in the AKT pathway, and respond to growth factors or internal environmental stimuli, such as acid-alkali poisoning (60). It has been reported that the upregulation of lncRNA-AB073614 expression in ovarian cancer (61) and glioma (62,63) is associated with the occurrence and development of tumors. In vitro, the knockdown of AB073614 expression significantly inhibits the proliferation, differentiation, apoptosis, metastasis and invasion of colorectal cancer cells, and results in increased rates of apoptosis and G1 phase cell cycle arrest, while overexpression of AB073614 results in the opposite effects. The agonist (740Y-P) and an inhibitor (LY294002) of the PI3K/AKT signaling pathway were used to demonstrate the role of AB073614 in colorectal tumor cells (64). Using similar research methods, a team reported that PlncRNA-1 (65) and NEAT1 (66) act on the PI3K/AKT pathway, and participate in the proliferation and apoptosis of colorectal cancer cells. A study on 102 gastric cancer tissues and adjacent normal tissues indicated that the abnormal expression of lncRNA-UCA1 serves an important role in the development, metastasis and invasion of gastric cancer. In vitro and in vivo experiments demonstrated that the molecular mechanism of UCA1 regulation on the PI3K/AKT pathway occurs by regulating PI3K-Akt-mTOR signaling proteins and downstream molecules to influence the growth process of gastric cancer cells (67). MALAT1 regulates multiple signaling pathways. In breast and ovarian cancer, it was recently discovered that MALAT1 influences the epithelial-mesenchymal transition and regulates the tumorigenicity of cells by targeting the PI3K/AKT pathway (68,69). In bovine epithelial cells, lncRNA-H19 also affects the epithelial-mesenchymal transition via PI3K/AKT regulation (70). Table V lists the lncRNAs associated with the PI3K/AKT signaling pathway in cancer.

Table V

lncRNAs that interact with the PI3K/AKT signaling pathway in cancer.

Table V

lncRNAs that interact with the PI3K/AKT signaling pathway in cancer.

Author, yearlncRNAAssociated diseaseFunctionEffect on pathway(Refs.)
Cheng et al, 2015AB073614Ovarian cancerPromote tumorigenesisPositive(61)
Wang et al, 2017AB073614Colorectal cancerPromote tumor growthPositive(64)
Song et al, 2017PlncRNA-1Colorectal carcinomaPromote tumor growthPositive(65)
Peng et al, 2017NEAT1Colorectal cancerPromote tumor growthPositive(66)
Zhang et al, 2017MEG3GliomaInhibit tumorigenesisNegative(125)
Li et al, 2018UCA1OsteosarcomaPromote proliferationPositive(126)
Yun et al, 2016TUG1OsteosarcomaPromote proliferationPositive(127)
Yan et al, 2016HOTAIRGastric cancerPromotes cisplatin resistancePositive(128)
Wang et al, 2017HOTAIRChronic myeloid leukemiaPromote imatinib resistancePositive(129)
Jiang et al, 2017DANCROsteosarcomaPromote tumor progressionPositive(130)
Yuan et al, 2018HOTTIPPapillary thyroid carcinomaPromote proliferationPositive(131)
Xue et al, 2016GAS5Prostate cancerInhibit tumor growthNegative(132)
Huang et al, 2016XistBreast cancerInhibit tumor growthNegative(133)
Lu et al, 2017HULCChronic myeloid leukemiaPromote proliferationPositive(134)
Wang et al, 2017HULCBladder cancerPromote proliferationPositive(135)
He et al, 2017NONHSAT062994Colorectal cancerInhibit tumor growthNegative(136)
Han et al, 2018p21OsteosarcomaInhibit proliferationNegative(137)
Wang et al, 2018AB209630Pancreatic ductal adenocarcinomaInhibit gemcitabine resistanceNegative(138)
Wang et al, 2017MIR31HGNon-small cell lung cancerPromote gefitinib resistancePositive(139)
Chen et al, 2017PTENP1Breast cancerInhibit proliferation and metastasisNegative(140)

7. lncRNA and the MAPK signaling pathway

The MAPK pathway is a classical signaling pathway, and is involved in the regulation of cell proliferation, differentiation, transformation and apoptosis. The MAPK pathway is associated with the occurrence of various diseases, including inflammation and tumors, by phosphorylating nuclear transcription factors, cytoskeletal proteins, and enzymes. MAPK signaling primarily includes four pathways: ERK, JNK/SAPK, P38 and ERK5/BMK1. The ERK, JNK, P38 and ERK5/BMK1 pathways can be activated by different stimuli, and there is wide crosstalk between these pathways, resulting in mutual synergy or inhibition between them. As important regulatory genes, numerous lncRNAs have been identified to regulate the MAPK pathway.

An important role of the SNHG12-miR-181a-MAPK/Slug axis was described in multidrug resistance in non-small cell lung cancer cells. In non-small cell lung cancer cells, silencing of lncRNA-SNHG12 upregulates miR-181a expression, and inhibits MAPK1 and MAP2K1 expression. This is accompanied by decreased phosphorylation of MAPK1 and MAP2K1, and decreased Slug expression levels. Ultimately, lncRNA-SNHG12 inhibits the activation of the MAPK/Slug pathway and increases the drug sensitivity of tumor cells (71). Similarly, lncRNA-XIST acts via microRNA to influence the activation of pathways. XIST regulates MAPK1 by targeting miR-194-5p-like ceRNA and reducing its expression. Silenced XIST inhibits the proliferation of hepatoma cells and reduces their invasiveness (72). In previous in vitro and in vivo studies of gallbladder carcinoma, the knockdown of MALAT1 was demonstrated to reduce the phosphorylation of MEK1/2, ERK1/2, MAPK and JNK 1/2/3, resulting in significant inhibition of the ERK/MAPK pathway. The metastasis and invasiveness of cancer cells is also weakened simultaneously (73). In a cecal ligation and puncture (CLP) rat model, the expression levels of p38, MAPK and NF-κB protein were significantly higher in the CLP group compared with that in the sham group (74). Overexpression of MALAT1 alone significantly increases the levels of p38/MAPK and NF-κB. An elaborative study demonstrated that MALAT1 mediates the development of heart failure and the inflammatory response through the activation of p38/MAPK/NF-κB. Furthermore, in the inflammatory response to sepsis, p38/MAPK/NF-κB may be downstream of MALAT1 signaling (74). In neuro-genic hyperplasia of neuroblastoma-derived Neuro-2a cells, silencing of MALAT1 results in significant activation of the MAPK pathway, and abnormal activation of the peroxisome proliferator-activated receptor and the p53 pathway (75). Thus, the same lncRNA appears to exhibit different effects in different types of tumors. When lncRNA-BANCR is deleted in lung cancer cells, the proliferation and metastasis of cancer cells is increased. In addition, when the expression of BANCR is increased, the proliferation of lung cancer cells is inhibited, acting as a tumor suppressor gene (76). In in vivo and in vitro experiments on malignant melanoma, tumor growth is inhibited following BANCR-knockout. The underlying mechanism may involve the silencing of BANCR making it difficult to activate the MAPK pathway, and to inhibit important components of the pathway, ERK1/2 and JNK (77). Table VI lists the lncRNAs that interact with the MAPK pathway in cancer.

Table VI

lncRNAs that interact with the MAPK signaling pathway in cancer.

Table VI

lncRNAs that interact with the MAPK signaling pathway in cancer.

Author, yearlncRNAAssociated diseaseFunctionEffect on pathway(Refs.)
Wang et al, 2017SNHG12Non-small cell lung cancerPromote multidrug resistancePositive(71)
Kong et al, 2017XistHepatocellular carcinomaPromote proliferation, migration and invasionPositive(72)
Wu et al, 2014MALAT1Gallbladder cancerPromote proliferation and metastasisPositive(73)
Liu et al, 2017MALAT1RetinoblastomaPromote proliferation and metastasisPositive(143)
Jiang et al, 2015BANCRLung carcinomaPromotes proliferation and migrationPositive(76)
Li et al, 2014BANCRMalignant melanomaPromote proliferationPositive(77)
Huang et al, 2015TUG1Non-small cell lung cancerInhibit tumorigenesisNegative(98)
Chen et al, 2017PTENP1Breast cancerInhibit proliferation and metastasisNegative(140)
Li et al, 2018HOTAIRCervical cancerPromote metastasis and invasionPositive(141)
Gao et al, 2017CCAT1 MedulloblastomaPromote proliferation and metastasisPositive(142)
Huang et al, 2015DBH-AS1Hepatocellular carcinomaPromote proliferationPositive(144)
Peng et al, 2017RoRBreast cancerPromote estrogen-independent tumor growthPositive(145)
Wu et al, 2017NEAT1Non-small cell lung cancerPromote tumorigenesisPositive(146)
Peng et al, 2016CCHE1Hepatocellular carcinomaPromote carcinogenesisPositive(147)

8. Discussion

The regulatory roles of lncRNAs are diverse and complex, and the current understanding of them is limited. The ENCODE research project identified <10,000 lncRNAs in the human genome (78), but only ~100 lncRNAs are known to be biologically functional. This is not only due to limited research techniques, but also the nature of lncRNAs. The intracellular abundance of lncRNA is typically low, and the function between different species is not conserved. For example, the human HOTAIR gene has a transregulatory effect on the HOXD gene, but this function is not observed in mice (79). Previous studies have demonstrated that lncRNAs primarily act as signal transduction molecules, decoys, transcription factors and scaffolds, interacting with proteins, DNA, RNA or other molecules to participate in transcriptional regulation, post-transcriptional regulation and epigenetic regulation of genes. The transcriptional regulation and posttranscriptional regulation of lncRNAs have been detailed, but their regulatory role in epigenetics is also an important function, and lncRNA-Xist is an interesting example (80,81). lncRNA-Xist is able to extensively bind to the X-chromosome through an interaction with polycomb repressive complex 2, leading to the inactivation of the X-chromosome (82,83). Recently, the novel silencing effect of Xist on the X-chromosome identified by Chen et al (84) has been debated. In the experiments by Chen et al (84), it was demonstrated that Xist interacts with Lamin B Receptor (LBR), a nuclear inner membrane protein involved in the establishment of the three-dimensional structures of chromosomes by regulating the anchoring of chromatin in the nucleus. It was hypothesized that the combination of Xist and LBR triggers alterations to the three-dimensional structure of the X-chromosome and thus participates in the silencing of the X-chromosome. This hypothesis was validated by constructing stable cell lines, LBS-Xist and LBS-Xist-BoxB, associated with Xist and LBR (84). However, a study by Wang et al questioned these findings. According to the analysis of the original sequencing data, it was considered that the Xist-associated stable cell lines, LBS-Xist and LBS-Xist-BoxB, were unsuccessfully constructed, and the sequencing depth and data normalization were also problematic (85). In response, the original research team provided more detailed evidence to confirm the results of their own research (86). Similar disputes reflect that, although lncRNAs have been recognized as having important biological functions, our understanding remains far from thorough. However, rational academic controversy makes lncRNA-associated research more rigorous.

The lncRNA research surge began with HOTAIR, first reported in cell by Rinn et al, in 2007 (2). As the first lncRNA reported to have a transregulatory effect, it became a popular topic of research. However, researchers still question the role of HOTAIR in mouse development, and it is considered that it may not be as important as previous studies have described (87,88). Furthermore, enrichment of the H3K27me3 modification of HOTAIR-regulated HOXD sites is not as pronounced as originally thought (79), and reports directly indicate that HOTAIR-knockout mice exhibit no apparent phenotypic changes (89). With some controversy, the differential expression of lncRNAs in a variety of types of cancer and chronic diseases have been confirmed. Several of the complex molecular mechanisms of lncRNA interaction have also been revealed. lncRNA research has improved our understanding of cell biological regulatory network, which is a step towards understanding the molecular mechanisms involved in disease. Although lncRNAs are currently at an early stage of research as potential targets for the treatment of cancer, certain small molecule lncRNAs have entered clinical trials as targeted drugs. It is expected that the targeted treatment of lncRNAs will become clinically available, improve the quality of life and prolong the survival of patients.

Funding

This grant was supported by National Science Foundation Committee of China (grant no. 81502482 and 81672474), Zhejiang Provincial National Science Foundation Committee of China (grant no. LQ14H160017) and Zhejiang Provincial research projects of medical and healthy industries (grant nos. 2015KYB026, 2017KY193 and 2017KY210).

Availability of data and materials

Not applicable.

Authors' contributions

YS, JYZ and HHC consulted and preliminarily summarized the literature required for the manuscript. WS was the major contributor in writing the manuscript. ZFW contributed to the writing of the manuscript. DSH and JGZ approved the final version of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

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 

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

3 

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

4 

Amaral PP, Clark MB, Gascoigne DK, Dinger ME and Mattick JS: lncRNAdb: A reference database for long noncoding RNAs. Nucleic Acids Res. 39(Suppl 1): D146–D151. 2011. View Article : Google Scholar :

5 

St Laurent G, Wahlestedt C and Kapranov P: The Landscape of long noncoding RNA classification. Trends Genet. 31:239–251. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Wang Z, Gerstein M and Snyder M: RNA-Seq: A revolutionary tool for transcriptomics. Nat Rev Genet. 10:57–63. 2009. View Article : Google Scholar

7 

Lane DP and Crawford LV: T antigen is bound to a host protein in SV40-transformed cells. Nature. 278:261–263. 1979. View Article : Google Scholar : PubMed/NCBI

8 

Schmitt AM, Garcia JT, Hung T, Flynn RA, Shen Y, Qu K, Payumo AY, Peres-da-Silva A, Broz DK, Baum R, et al: An inducible long noncoding RNA amplifies DNA damage signaling. Nat Genet. 48:1370–1376. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Tripathi V, Shen Z, Chakraborty A, Giri S, Freier SM, Wu X, Zhang Y, Gorospe M, Prasanth SG, Lal A, et al: Long noncoding RNA MALAT1 controls cell cycle progression by regulating the expression of oncogenic transcription factor B-MYB. PLoS Genet. 9:e10033682013. View Article : Google Scholar : PubMed/NCBI

10 

Zhang A, Zhou N, Huang J, Liu Q, Fukuda K, Ma D, Lu Z, Bai C, Watabe K and Mo YY: The human long non-coding RNA-RoR is a p53 repressor in response to DNA damage. Cell Res. 23:340–350. 2013. View Article : Google Scholar :

11 

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

12 

Loewer S, Cabili MN, Guttman M, Loh YH, Thomas K, Park IH, Garber M, Curran M, Onder T, Agarwal S, et al: Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat Genet. 42:1113–1117. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D, Khalil AM, Zuk O, Amit I, Rabani M, et al: A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 142:409–419. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Tay Y, Rinn J and Pandolfi PP: The multilayered complexity of ceRNA crosstalk and competition. Nature. 505:344–352. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Liu Q, Huang J, Zhou N, Zhang Z, Zhang A, Lu Z, Wu F and Mo YY: LncRNA loc285194 is a p53-regulated tumor suppressor. Nucleic Acids Res. 41:4976–4987. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Zhang XF, Ye Y and Zhao SJ: LncRNA Gas5 acts as a ceRNA to regulate PTEN expression by sponging miR-222-3p in papillary thyroid carcinoma. Oncotarget. 9:3519–3530. 2017.

17 

Cai H, Yao J, An Y, Chen X, Chen W, Wu D, Luo B, Yang Y, Jiang Y, Sun D, et al: LncRNA HOTAIR acts a competing endogenous RNA to control the expression of notch3 via sponging miR-613 in pancreatic cancer. Oncotarget. 8:32905–32917. 2017.PubMed/NCBI

18 

Hudson WH, Pickard MR, de Vera IM, Kuiper EG, Mourtada-Maarabouni M, Conn GL, Kojetin DJ, Williams GT and Ortlund EA: Conserved sequence-specific lincRNA-steroid receptor interactions drive transcriptional repression and direct cell fate. Nat Commun. 5:53952014. View Article : Google Scholar : PubMed/NCBI

19 

Zhang X, Gejman R, Mahta A, Zhong Y, Rice KA, Zhou Y, Cheunsuchon P, Louis DN and Klibanski A: Maternally expressed gene 3, an imprinted noncoding RNA gene, is associated with meningioma pathogenesis and progression. Cancer Res. 70:2350–2358. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Zhou Y, Zhong Y, Wang Y, Zhang X, Batista DL, Gejman R, Ansell PJ, Zhao J, Weng C and Klibanski A: Activation of p53 by MEG3 non-coding RNA. J Biol Chem. 282:24731–24742. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Mahmoudi S, Henriksson S, Corcoran M, Méndez-Vidal C, Wiman KG and Farnebo M: Wrap53, a natural p53 antisense transcript required for p53 induction upon DNA damage. Mol Cell. 33:462–471. 2009. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Rapicavoli NA, Qu K, Zhang J, Mikhail M, Laberge RM and Chang HY: A mammalian pseudogene lncRNA at the interface of inflammation and anti-inflammatory therapeutics. eLife. 2:e007622013. View Article : Google Scholar : PubMed/NCBI

24 

Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, Lin L, Yao H, Su F, Li D, et al: A cytoplasmic NF-κB interacting long noncoding RNA blocks IκB phosphorylation and suppresses breast cancer metastasis. Cancer Cell. 27:370–381. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Rozic JG, Chakraborty C and Lala PK: Cyclooxygenase inhibitors retard murine mammary tumor progression by reducing tumor cell migration, invasiveness and angiogenesis. Int J Cancer. 93:497–506. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Subbaramaiah K and Dannenberg AJ: Cyclooxygenase 2: A molecular target for cancer prevention and treatment. Trends Pharmacol Sci. 24:96–102. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Krawczyk M and Emerson BM: p50-associated COX-2 extragenic RNA (PACER) activates COX-2 gene expression by occluding repressive NF-κB complexes. eLife. 3:e017762014. View Article : Google Scholar

28 

Pearson MJ, Philp AM, Heward JA, Roux BT, Walsh DA, Davis ET, Lindsay MA and Jones SW: Long intergenic noncoding RNAs mediate the human chondrocyte inflammatory response and are differentially expressed in osteoarthritis cartilage. Arthritis Rheumatol. 68:845–856. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Hu G, Gong A-Y, Wang Y, Ma S, Chen X, Chen J, Su CJ, Shibata A, Strauss-Soukup JK, Drescher KM, et al: LincRNA-Cox2 promotes late inflammatory gene transcription in macrophages through modulating SWI/SNF-mediated chromatin remodeling. J Immunol. 196:2799–2808. 2016. View Article : Google Scholar : PubMed/NCBI

30 

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

31 

Özeş AR, Miller DF, Özeş ON, Fang F, Liu Y, Matei D, Huang T and Nephew KP: NF-κB-HOTAIR axis links DNA damage response, chemoresistance and cellular senescence in ovarian cancer. Oncogene. 35:5350–5361. 2016. View Article : Google Scholar

32 

Rajbhandari R, McFarland BC, Patel A, Gerigk M, Gray GK, Fehling SC, Bredel M, Berbari NF, Kim H, Marks MP, et al: Loss of tumor suppressive microRNA-31 enhances TRADD/NF-κB signaling in glioblastoma. Oncotarget. 6:17805–17816. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Xu Q, Deng F, Xing Z, Wu Z, Cen B, Xu S, Zhao Z, Nepomuceno R, Bhuiyan MI, Sun D, et al: Long non-coding RNA C2dat1 regulates CaMKIIδ expression to promote neuronal survival through the NF-κB signaling pathway following cerebral ischemia. Cell Death Dis. 7:e21732016. View Article : Google Scholar

34 

IIott NE, Heward JA, Roux B, Tsitsiou E, Fenwick PS, Lenzi L, Goodhead I, Hertz-Fowler C, Heger A, Hall N, et al: Long non-coding RNAs and enhancer RNAs regulate the lipopolysac-charide-induced inflammatory response in human monocytes. Nat Commun. 5:39792014. View Article : Google Scholar

35 

Chan J, Atianand M, Jiang Z, Carpenter S, Aiello D, Elling R, Fitzgerald KA and Caffrey DR: Cutting edge: A natural antisense transcript, AS-IL1α, controls inducible transcription of the proin-flammatory cytokine IL-1α. J Immunol. 195:1359–1363. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Zhou X, Han X, Wittfeldt A, Sun J, Liu C, Wang X, Gan LM, Cao H and Liang Z: Long non-coding RNA ANRIL regulates inflammatory responses as a novel component of NF-κB pathway. RNA Biol. 13:98–108. 2016. View Article : Google Scholar

37 

Li Z, Chao TC, Chang KY, Lin N, Patil VS, Shimizu C, Head SR, Burns JC and Rana TM: The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL. Proc Natl Acad Sci USA. 111:1002–1007. 2014. View Article : Google Scholar

38 

Cui H, Xie N, Tan Z, Banerjee S, Thannickal VJ, Abraham E and Liu G: The human long noncoding RNA lnc-IL7R regulates the inflammatory response. Eur J Immunol. 44:2085–2095. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Nusse R and Varmus HE: Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell. 31:99–109. 1982. View Article : Google Scholar : PubMed/NCBI

40 

Miki T, Yasuda SY and Kahn M: Wnt/β-catenin signaling in embryonic stem cell self-renewal and somatic cell reprogramming. Stem Cell Rev. 7:836–846. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Serio RN: Wnt of the two horizons: Putting stem cell self-renewal and cell fate determination into context. Stem Cells Dev. 23:1975–1990. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Ma Y, Yang Y, Wang F, Moyer MP, Wei Q, Zhang P, Yang Z, Liu W, Zhang H, Chen N, et al: Long non-coding RNA CCAL regulates colorectal cancer progression by activating Wnt/β-catenin signalling pathway via suppression of activator protein 2α. Gut. 65:1494–1504. 2015. View Article : Google Scholar

43 

Fan Y, Shen B, Tan M, Mu X, Qin Y, Zhang F and Liu Y: Long non-coding RNA UCA1 increases chemoresistance of bladder cancer cells by regulating Wnt signaling. FEBS J. 281:1750–1758. 2014. View Article : Google Scholar : PubMed/NCBI

44 

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

45 

Xiao Z, Qu Z, Chen Z, Fang Z, Zhou K, Huang Z, Guo X and Zhang Y: LncRNA HOTAIR is a prognostic biomarker for the proliferation and chemoresistance of colorectal cancer via MiR-203a-3p-mediated Wnt/β-catenin signaling pathway. Cell Physiol Biochem. 46:1275–1285. 2018. View Article : Google Scholar

46 

Shao K, Shi T, Yang Y, Wang X, Xu D and Zhou P: Highly expressed lncRNA CRNDE promotes cell proliferation through Wnt/β-catenin signaling in renal cell carcinoma. Tumour Biol. 37:15997–16004. 2016. View Article : Google Scholar

47 

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

48 

Fu X, Li H, Liu C, Hu B, Li T and Wang Y: Long noncoding RNA AK126698 inhibits proliferation and migration of non-small cell lung cancer cells by targeting Frizzled-8 and suppressing Wnt/β-catenin signaling pathway. OncoTargets Ther. 9:3815–3827. 2016. View Article : Google Scholar

49 

Yuan Z, Yu X, Ni B, Chen D, Yang Z, Huang J, Wang J, Chen D and Wang L: Overexpression of long non-coding RNA-CTD903 inhibits colorectal cancer invasion and migration by repressing Wnt/β-catenin signaling and predicts favorable prognosis. Int J Oncol. 48:2675–2685. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Xia Y, He Z, Liu B, Wang P and Chen Y: Downregulation of Meg3 enhances cisplatin resistance of lung cancer cells through activation of the WNT/β-catenin signaling pathway. Mol Med Rep. 12:4530–4537. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Li Z, Zhao L and Wang Q: Overexpression of long non-coding RNA HOTTIP increases chemoresistance of osteosarcoma cell by activating the Wnt/β-catenin pathway. Am J Transl Res. 8:2385–2393. 2016.

52 

High FA and Epstein JA: The multifaceted role of Notch in cardiac development and disease. Nat Rev Genet. 9:49–61. 2008. View Article : Google Scholar

53 

Wang Y, Wu P, Lin R, Rong L, Xue Y and Fang Y: LncRNA NALT interaction with NOTCH1 promoted cell proliferation in pediatric T cell acute lymphoblastic leukemia. Sci Rep. 5:137492015. View Article : Google Scholar : PubMed/NCBI

54 

Zhou S, Yu L, Xiong M and Dai G: LncRNA SNHG12 promotes tumorigenesis and metastasis in osteosarcoma by upregu-lating Notch2 by sponging miR-195-5p. Biochem Biophys Res Commun. 495:1822–1832. 2018. View Article : Google Scholar

55 

Yang X, Duan B and Zhou X: Long non-coding RNA FOXD2-AS1 functions as a tumor promoter in colorectal cancer by regulating EMT and Notch signaling pathway. Eur Rev Med Pharmacol Sci. 21:3586–3591. 2017.PubMed/NCBI

56 

Guo Q, Qian Z, Yan D, Li L and Huang L: LncRNA-MEG3 inhibits cell proliferation of endometrial carcinoma by repressing Notch signaling. Biomed Pharmacother. 82:589–594. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Katsushima K, Natsume A, Ohka F, Shinjo K, Hatanaka A, Ichimura N, Sato S, Takahashi S, Kimura H, Totoki Y, et al: Targeting the Notch-regulated non-coding RNA TUG1 for glioma treatment. Nat Commun. 7:136162016. View Article : Google Scholar : PubMed/NCBI

58 

Osaki M, Oshimura M and Ito H: PI3K-Akt pathway: Its functions and alterations in human cancer. Apoptosis. 9:667–676. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Bellacosa A, Kumar CC, Di Cristofano A and Testa JR: Activation of AKT kinases in cancer: Implications for therapeutic targeting. Adv Cancer Res. 94:29–86. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Koirala P, Huang J, Ho TT, Wu F, Ding X and Mo YY: LncRNA AK023948 is a positive regulator of AKT. Nat Commun. 8:144222017. View Article : Google Scholar : PubMed/NCBI

61 

Cheng Z, Guo J, Chen L, Luo N, Yang W and Qu X: A long noncoding RNA AB073614 promotes tumorigenesis and predicts poor prognosis in ovarian cancer. Oncotarget. 6:25381–25389. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Hu L, Lv QL, Chen SH, Sun B, Qu Q, Cheng L, Guo Y, Zhou HH and Fan L: Up-regulation of long non-coding RNA AB073614 predicts a poor prognosis in patients with glioma. Int J Environ Res Public Health. 13:4332016. View Article : Google Scholar : PubMed/NCBI

63 

Li J, Wang YM and Song YL: Knockdown of long noncoding RNA AB073614 inhibits glioma cell proliferation and migration via affecting epithelial-mesenchymal transition. Eur Rev Med Pharmacol Sci. 20:3997–4002. 2016.PubMed/NCBI

64 

Wang Y, Kuang H, Xue J, Liao L, Yin F and Zhou X: LncRNA AB073614 regulates proliferation and metastasis of colorectal cancer cells via the PI3K/AKT signaling pathway. Biomed Pharmacother. 93:1230–1237. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Song W, Mei JZ and Zhang M: lncRNA PlncRNA-1 promotes colorectal cancer cell progression by regulating PI3K/Akt signaling pathway. Oncol Res. 26:261–268. 2018. View Article : Google Scholar

66 

Peng W, Wang Z and Fan H: lncRNA NEAT1 impacts cell proliferation and apoptosis of colorectal cancer via regulation of Akt signaling. Pathol Oncol Res. 23:651–656. 2017. View Article : Google Scholar

67 

Li C, Liang G, Yang S, Sui J, Yao W, Shen X, Zhang Y, Peng H, Hong W, Xu S, et al: Dysregulated lncRNA-UCA1 contributes to the progression of gastric cancer through regulation of the PI3K-Akt-mTOR signaling pathway. Oncotarget. 8:93476–93491. 2017.PubMed/NCBI

68 

Xu S, Sui S, Zhang J, Bai N, Shi Q, Zhang G, Gao S, You Z, Zhan C, Liu F, et al: Downregulation of long noncoding RNA MALAT1 induces epithelial-to-mesenchymal transition via the PI3K-AKT pathway in breast cancer. Int J Clin Exp Pathol. 8:4881–4891. 2015.PubMed/NCBI

69 

Jin Y, Feng SJ, Qiu S, Shao N and Zheng JH: LncRNA MALAT1 promotes proliferation and metastasis in epithelial ovarian cancer via the PI3K-AKT pathway. Eur Rev Med Pharmacol Sci. 21:3176–3184. 2017.PubMed/NCBI

70 

Yang W, Li X, Qi S, Li X, Zhou K, Qing S, Zhang Y and Gao MQ: lncRNA H19 is involved in TGF-β1-induced epithelial to mesenchymal transition in bovine epithelial cells through PI3K/AKT Signaling Pathway. PeerJ. 5:e39502017. View Article : Google Scholar

71 

Wang P, Chen D, Ma H and Li Y: LncRNA SNHG12 contributes to multidrug resistance through activating the MAPK/Slug pathway by sponging miR-181a in non-small cell lung cancer. Oncotarget. 8:84086–84101. 2017.PubMed/NCBI

72 

Kong Q, Zhang S, Liang C, Zhang Y, Kong Q, Chen S, Qin J and Jin Y: lncRNA XIST functions as a molecular sponge of miR-194-5p to regulate MAPK1 expression in hepatocellular carcinoma cell. J Cell Biochem. 119:4458–4468. 2018. View Article : Google Scholar

73 

Wu XS, Wang XA, Wu WG, Hu YP, Li ML, Ding Q, Weng H, Shu YJ, Liu TY, Jiang L, et al: MALAT1 promotes the proliferation and metastasis of gallbladder cancer cells by activating the ERK/MAPK pathway. Cancer Biol Ther. 15:806–814. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Chen H, Wang X, Yan X, Cheng X, He X and Zheng W: LncRNA MALAT1 regulates sepsis-induced cardiac inflammation and dysfunction via interaction with miR-125b and p38 MAPK/NFκB. Int Immunopharmacol. 55:69–76. 2018. View Article : Google Scholar

75 

Chen L, Feng P, Zhu X, He S, Duan J and Zhou D: Long non-coding RNA Malat1 promotes neurite outgrowth through activation of ERK/MAPK signalling pathway in N2a cells. J Cell Mol Med. 20:2102–2110. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Jiang W, Zhang D, Xu B, Wu Z, Liu S, Zhang L, Tian Y, Han X and Tian D: Long non-coding RNA BANCR promotes proliferation and migration of lung carcinoma via MAPK pathways. Biomed Pharmacother. 69:90–95. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Li R, Zhang L, Jia L, Duan Y, Li Y, Bao L and Sha N: Long non-coding RNA BANCR promotes proliferation in malignant melanoma by regulating MAPK pathway activation. PLoS One. 9:e1008932014. View Article : Google Scholar : PubMed/NCBI

78 

Rosenbloom KR, Dreszer TR, Long JC, Malladi VS, Sloan CA, Raney BJ, Cline MS, Karolchik D, Barber GP, Clawson H, et al: ENCODE whole-genome data in the UCSC Genome Browser: Update 2012. Nucleic Acids Res. 40:D912–D917. 2012. View Article : Google Scholar :

79 

Schorderet P and Duboule D: Structural and functional differences in the long non-coding RNA hotair in mouse and human. PLoS Genet. 7:e10020712011. View Article : Google Scholar : PubMed/NCBI

80 

Brown CJ, Ballabio A, Rupert JL, Lafreniere RG, Grompe M, Tonlorenzi R and Willard HF: A gene from the region of the human X inactivation centre is expressed exclusively from the inactive X chromosome. Nature. 349:38–44. 1991. View Article : Google Scholar : PubMed/NCBI

81 

Brown CJ, Hendrich BD, Rupert JL, Lafrenière RG, Xing Y, Lawrence J and Willard HF: The human XIST gene: Analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell. 71:527–542. 1992. View Article : Google Scholar : PubMed/NCBI

82 

Engreitz JM, Pandya-Jones A, McDonel P, Shishkin A, Sirokman K, Surka C, Kadri S, Xing J, Goren A, Lander ES, et al: The Xist lncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science. 341:12379732013. View Article : Google Scholar : PubMed/NCBI

83 

Zhao J, Sun BK, Erwin JA, Song J-J and Lee JT: Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science. 322:750–756. 2008. View Article : Google Scholar : PubMed/NCBI

84 

Chen CK, Blanco M, Jackson C, Aznauryan E, Ollikainen N, Surka C, Chow A, Cerase A, McDonel P and Guttman M: Xist recruits the X chromosome to the nuclear lamina to enable chromosome-wide silencing. Science. 354:468–472. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Wang CY, Froberg JE, Blum R, Jeon Y and Lee JT: Comment on ‘Xist recruits the X chromosome to the nuclear lamina to enable chromosome-wide silencing’. Science. 356:3562017. View Article : Google Scholar

86 

Chen CK, Chow A, Lai M and Guttman M: Response to Comment on ‘Xist recruits the X chromosome to the nuclear lamina to enable chromosome-wide silencing’. Science. 356:54392017. View Article : Google Scholar

87 

Suemori H and Noguchi S: Hox C cluster genes are dispensable for overall body plan of mouse embryonic development. Dev Biol. 220:333–342. 2000. View Article : Google Scholar

88 

Amândio AR, Necsulea A, Joye E, Mascrez B and Duboule D: Hotair is dispensible for mouse development. PLoS Genet. 12:e10062322016. View Article : Google Scholar : PubMed/NCBI

89 

Lai KM, Gong G, Atanasio A, Rojas J, Quispe J, Posca J, White D, Huang M, Fedorova D, Grant C, et al: Diverse phenotypes and specific transcription patterns in twenty mouse lines with ablated LincRNAs. PLoS One. 10:e01255222015. View Article : Google Scholar : PubMed/NCBI

90 

Tano K, Onoguchi-Mizutani R, Yeasmin F, Uchiumi F, Suzuki Y, Yada T and Akimitsu N: Identification of Minimal p53 Promoter Region Regulated by MALAT1 in Human Lung Adenocarcinoma Cells. Front Genet. 8:2082018. View Article : Google Scholar :

91 

Yang P, Yang Y, An W, Xu J, Zhang G, Jie J and Zhang Q: The long noncoding RNA-ROR promotes the resistance of radiotherapy for human colorectal cancer cells by targeting the p53/miR-145 pathway. J Gastroenterol Hepatol. 32:837–845. 2017. View Article : Google Scholar

92 

Chen RP, Huang ZL, Liu LX, Xiang MQ, Li GP, Feng JL, Liu B and Wu LF: Involvement of endoplasmic reticulum stress and p53 in lncRNA MEG3-induced human hepatoma HepG2 cell apoptosis. Oncol Rep. 36:1649–1657. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Li J, Bian EB, He XJ, Ma CC, Zong G, Wang HL and Zhao B: Epigenetic repression of long non-coding RNA MEG3 mediated by DNMT1 represses the p53 pathway in gliomas. Int J Oncol. 48:723–733. 2016. View Article : Google Scholar

94 

Lu KH, Li W, Liu XH, Sun M, Zhang ML, Wu WQ, Xie WP and Hou YY: Long non-coding RNA MEG3 inhibits NSCLC cells proliferation and induces apoptosis by affecting p53 expression. BMC Cancer. 13:4612013. View Article : Google Scholar : PubMed/NCBI

95 

Zhu J, Liu S, Ye F, Shen Y, Tie Y, Zhu J, Wei L, Jin Y, Fu H, Wu Y, et al: Long noncoding RNA MEG3 interacts with p53 protein and regulates partial p53 target genes in hepatoma cells. PLoS One. 10:e01397902015. View Article : Google Scholar : PubMed/NCBI

96 

Sun L, Li Y and Yang B: Downregulated long non-coding RNA MEG3 in breast cancer regulates proliferation, migration and invasion by depending on p53's transcriptional activity. Biochem Biophys Res Commun. 478:323–329. 2016. View Article : Google Scholar : PubMed/NCBI

97 

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

98 

Huang J, Zhou N, Watabe K, Lu Z, Wu F, Xu M and Mo YY: Long non-coding RNA UCA1 promotes breast tumor growth by suppression of p27 (Kip1). Cell Death Dis. 5:e10082014. View Article : Google Scholar : PubMed/NCBI

99 

Zhai N, Xia Y, Yin R, Liu J and Gao F: A negative regulation loop of long noncoding RNA HOTAIR and p53 in non-small-cell lung cancer. OncoTargets Ther. 9:5713–5720. 2016. View Article : Google Scholar

100 

Su P, Wang F, Qi B, Wang T and Zhang S: p53 regulation-association long non-coding RNA (LncRNA PRAL) inhibits cell proliferation by regulation of P53 in human lung cancer. Med Sci Monit. 23:1751–1758. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Gong Z, Zhang S, Zeng Z, Wu H, Yang Q, Xiong F, Shi L, Yang J, Zhang W, Zhou Y, et al: LOC401317, a p53-regulated long non-coding RNA, inhibits cell proliferation and induces apoptosis in the nasopharyngeal carcinoma cell line HNE2. PLoS One. 9:e1106742014. View Article : Google Scholar : PubMed/NCBI

102 

Zhai W, Li X, Wu S, Zhang Y, Pang H and Chen W: Microarray expression profile of lncRNAs and the upregulated ASLNC04080 lncRNA in human endometrial carcinoma. Int J Oncol. 46:2125–2137. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Thorenoor N, Faltejskova-Vychytilova P, Hombach S, Mlcochova J, Kretz M, Svoboda M and Slaby O: Long non-coding RNA ZFAS1 interacts with CDK1 and is involved in p53-dependent cell cycle control and apoptosis in colorectal cancer. Oncotarget. 7:622–637. 2016. View Article : Google Scholar :

104 

Li P, Zhang X, Wang L, Du L, Yang Y, Liu T, Li C and Wang C: lncRNA HOTAIR Contributes to 5FU Resistance through Suppressing miR-218 and Activating NF-κB/TS Signaling in Colorectal Cancer. Mol Ther Nucleic Acids. 8:356–369. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Liao Z, Zhao J and Yang Y: Downregulation of lncRNA H19 inhibits the migration and invasion of melanoma cells by inactivating the NF κB and PI3K/Akt signaling pathways. Mol Med Rep. 17:7313–7318. 2018.PubMed/NCBI

106 

Yang T, Li S, Liu J, Yin D, Yang X and Tang Q: lncRNA-NKILA/ NF-κB feedback loop modulates laryngeal cancer cell proliferation, invasion, and radioresistance. Cancer Med. 7:2048–2063. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Bian D, Gao C, Bao K and Song G: The long non-coding RNA NKILA inhibits the invasion-metastasis cascade of malignant melanoma via the regulation of NF-ĸB. Am J Cancer Res. 7:28–40. 2017.

108 

Huang W, Cui X, Chen J, Feng Y, Song E, Li J and Liu Y: Long non-coding RNA NKILA inhibits migration and invasion of tongue squamous cell carcinoma cells via suppressing epithelial-mesenchymal transition. Oncotarget. 7:62520–62532. 2016.PubMed/NCBI

109 

Ma Y, Zhou G, Li M, Hu D, Zhang L, Liu P and Lin K: Long noncoding RNA DANCR mediates cisplatin resistance in glioma cells via activating AXL/PI3K/Akt/NF-κB signaling pathway. Neurochem Int. 118:233–241. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Liang S, Zhang S, Wang P, Yang C, Shang C, Yang J and Wang J: LncRNA, TUG1 regulates the oral squamous cell carcinoma progression possibly via interacting with Wnt/β-catenin signaling. Gene. 608:49–57. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Yang YT, Wang YF, Lai JY, Shen SY, Wang F, Kong J, Zhang W and Yang HY: Long non-coding RNA UCA1 contributes to the progression of oral squamous cell carcinoma by regulating the WNT/β-catenin signaling pathway. Cancer Sci. 107:1581–1589. 2016. View Article : Google Scholar : PubMed/NCBI

112 

Ge XS, Ma HJ, Zheng XH, Ruan HL, Liao XY, Xue WQ, Chen YB, Zhang Y and Jia WH: HOTAIR, a prognostic factor in esophageal squamous cell carcinoma, inhibits WIF-1 expression and activates Wnt pathway. Cancer Sci. 104:1675–1682. 2013. View Article : Google Scholar : PubMed/NCBI

113 

Wu KF, Liang WC, Feng L, Pang JX, Waye MM, Zhang JF and Fu WM: H19 mediates methotrexate resistance in colorectal cancer through activating Wnt/β-catenin pathway. Exp Cell Res. 350:312–317. 2017. View Article : Google Scholar

114 

Cao Y, Shi H, Ren F, Jia Y and Zhang R: Long non-coding RNA CCAT1 promotes metastasis and poor prognosis in epithelial ovarian cancer. Exp Cell Res. 359:185–194. 2017. View Article : Google Scholar : PubMed/NCBI

115 

Fu Z, Chen C, Zhou Q, Wang Y, Zhao Y, Zhao X, Li W, Zheng S, Ye H, Wang L, et al: LncRNA HOTTIP modulates cancer stem cell properties in human pancreatic cancer by regulating HOXA9. Cancer Lett. 410:68–81. 2017. View Article : Google Scholar : PubMed/NCBI

116 

Yue B, Liu C, Sun H, Liu M, Song C, Cui R, Qiu S and Zhong M: A Positive Feed-Forward Loop between LncRNA-CYTOR and Wnt/β-Catenin Signaling Promotes Metastasis of Colon Cancer. Mol Ther. 26:1287–1298. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Liu B, Pan CF, He ZC, Wang J, Wang PL, Ma T, Xia Y and Chen YJ: Long noncoding RNA-LET suppresses tumor growth and EMT in lung adenocarcinoma. Biomed Res Int. 2016:46934712016. View Article : Google Scholar : PubMed/NCBI

118 

Wang X, Lu X, Geng Z, Yang G and Shi Y: lncRNA PTCSC3/ miR-574-5p governs cell proliferation and migration of papillary thyroid carcinoma via Wnt/β-catenin signaling. J Cell Biochem. 118:4745–4752. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Zhang Z, Zhou C, Chang Y, Zhang Z, Hu Y, Zhang F, Lu Y, Zheng L, Zhang W, Li X, et al: Long non-coding RNA CASC11 interacts with hnRNP-K and activates the WNT/β-catenin pathway to promote growth and metastasis in colorectal cancer. Cancer Lett. 376:62–73. 2016. View Article : Google Scholar : PubMed/NCBI

120 

Zhang F, Wan M, Xu Y, Li Z, Leng K, Kang P, Cui Y and Jiang X: Long noncoding RNA PCAT1 regulates extrahepatic cholangiocarcinoma progression via the Wnt/β-catenin-signaling pathway. Biomed Pharmacother. 94:55–62. 2017. View Article : Google Scholar : PubMed/NCBI

121 

Wang Y, Zhou J, Xu YJ and Hu HB: Long non-coding RNA LINC00968 acts as oncogene in NSCLC by activating the Wnt signaling pathway. J Cell Physiol. 233:3397–3406. 2018. View Article : Google Scholar

122 

Hang Q, Sun R, Jiang C and Li Y: Notch 1 promotes cisplatin-resistant gastric cancer formation by upregulating lncRNA AK022798 expression. Anticancer Drugs. 26:632–640. 2015.PubMed/NCBI

123 

Lu S, Dong W, Zhao P and Liu Z: lncRNA FAM83H-AS1 is associated with the prognosis of colorectal carcinoma and promotes cell proliferation by targeting the Notch signaling pathway. Oncol Lett. 15:1861–1868. 2018.PubMed/NCBI

124 

Chen H, Liu JZ, Hu GJ, Shi LL and Lan T: Promotion of proliferation and metastasis of hepatocellular carcinoma by LncRNA00673 based on the targeted-regulation of notch signaling pathway. Eur Rev Med Pharmacol Sci. 21:3412–3420. 2017.PubMed/NCBI

125 

Zhang L, Liang X and Li Y: Long non-coding RNA MEG3 inhibits cell growth of gliomas by targeting miR-93 and inactivating PI3K/AKT pathway. Oncol Rep. 38:2408–2416. 2017. View Article : Google Scholar : PubMed/NCBI

126 

Li T, Xiao Y and Huang T: HIF-1α-induced upregulation of lncRNA UCA1 promotes cell growth in osteosarcoma by inactivating the PTEN/AKT signaling pathway. Oncol Rep. 39:1072–1080. 2018.PubMed/NCBI

127 

Yun-Bo F, Xiao-Po L, Xiao-Li L, Guo-Long C, Pei Z and Fa-Ming T: LncRNA TUG1 is upregulated and promotes cell proliferation in osteosarcoma. Open Med (Wars). 11:163–167. 2016.

128 

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. 37:16345–16355. 2016. View Article : Google Scholar

129 

Wang H, Li Q, Tang S, Li M, Feng A, Qin L, Liu Z and Wang X: The role of long noncoding RNA HOTAIR in the acquired multidrug resistance to imatinib in chronic myeloid leukemia cells. Hematology. 22:208–216. 2017. View Article : Google Scholar

130 

Jiang N, Wang X, Xie X, Liao Y, Liu N, Liu J, Miao N, Shen J and Peng T: lncRNA DANCR promotes tumor progression and cancer stemness features in osteosarcoma by upregulating AXL via miR-33a5p inhibition. Cancer Lett. 405:46–55. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Yuan Q, Liu Y, Fan Y, Liu Z, Wang X, Jia M, Geng Z, Zhang J and Lu X: LncRNA HOTTIP promotes papillary thyroid carcinoma cell proliferation, invasion and migration by regulating miR-637. Int J Biochem Cell Biol. 98:1–9. 2018. View Article : Google Scholar : PubMed/NCBI

132 

Xue D, Zhou C, Lu H, Xu R, Xu X and He X: LncRNA GAS5 inhibits proliferation and progression of prostate cancer by targeting miR-103 through AKT/mTOR signaling pathway. Tumour Biol. 37:16187–16197. 2016. View Article : Google Scholar

133 

Huang YS, Chang CC, Lee SS, Jou YS and Shih HM: Xist reduction in breast cancer upregulates AKT phosphorylation via HDAC3-mediated repression of PHLPP1 expression. Oncotarget. 7:43256–43266. 2016.PubMed/NCBI

134 

Lu Y, Li Y, Chai X, Kang Q, Zhao P, Xiong J and Wang J: Long noncoding RNA HULC promotes cell proliferation by regulating PI3K/AKT signaling pathway in chronic myeloid leukemia. Gene. 607:41–46. 2017. View Article : Google Scholar : PubMed/NCBI

135 

Wang J, Ma W and Liu Y: Long non-coding RNA HULC promotes bladder cancer cells proliferation but inhibits apoptosis via regulation of ZIC2 and PI3K/AKT signaling pathway. Cancer Biomark. 20:425–434. 2017. View Article : Google Scholar : PubMed/NCBI

136 

He XS, Guo LC, Du MZ, Huang S, Huang RP, Zhan SH, Gu DM, Liu WS, Wang XM, Wu H, et al: The long non-coding RNA NONHSAT062994 inhibits colorectal cancer by inactivating Akt signaling. Oncotarget. 8:68696–68706. 2017.PubMed/NCBI

137 

Han W and Liu J: LncRNA-p21 inhibited the proliferation of osteosarcoma cells via the miR-130b/PTEN/AKT signaling pathway. Biomed Pharmacother. 97:911–918. 2018. View Article : Google Scholar

138 

Wang L, Wang F, Na L, Yu J, Huang L, Meng ZQ, Chen Z, Chen H, Ming LL and Hua YQ: LncRNA AB209630 inhibits gemcitabine resistance cell proliferation by regulating PI3K/ AKT signaling in pancreatic ductal adenocarcinoma. Cancer Biomark. 22:169–174. 2018. View Article : Google Scholar

139 

Wang B, Jiang H, Wang L, Chen X, Wu K, Zhang S, Ma S and Xia B: Increased MIR31HG lncRNA expression increases gefitinib resistance in non-small cell lung cancer cell lines through the EGFR/PI3K/AKT signaling pathway. Oncol Lett. 13:3494–3500. 2017. View Article : Google Scholar : PubMed/NCBI

140 

Chen S, Wang Y, Zhang JH, Xia QJ, Sun Q, Li ZK, Zhang JG, Tang MS and Dong MS: Long non-coding RNA PTENP1 inhibits proliferation and migration of breast cancer cells via AKT and MAPK signaling pathways. Oncol Lett. 14:4659–4662. 2017. View Article : Google Scholar : PubMed/NCBI

141 

Li Q, Feng Y, Chao X, Shi S, Liang M, Qiao Y, Wang B, Wang P and Zhu Z: HOTAIR contributes to cell proliferation and metastasis of cervical cancer via targetting miR-23b/MAPK1 axis. Biosci Rep. 38:BSR201715632018. View Article : Google Scholar :

142 

Gao R, Zhang R, Zhang C, Zhao L and Zhang Y: Long noncoding RNA CCAT1 promotes cell proliferation and metastasis in human medulloblastoma via MAPK pathway. Tumori. 104:43–50. 2017. View Article : Google Scholar : PubMed/NCBI

143 

Liu S, Yan G, Zhang J and Yu L: Knockdown of long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) inhibits proliferation, migration, and invasion and promoted apoptosis by targeting miR-124 in retinoblastoma. Oncol Res. May 21–2017.Epub ahead of print. View Article : Google Scholar

144 

Huang JL, Ren TY, Cao SW, Zheng SH, Hu XM, Hu YW, Lin L, Chen J, Zheng L and Wang Q: HBx-related long non-coding RNA DBH-AS1 promotes cell proliferation and survival by activating MAPK signaling in hepatocellular carcinoma. Oncotarget. 6:33791–33804. 2015. View Article : Google Scholar : PubMed/NCBI

145 

Peng WX, Huang JG, Yang L, Gong AH and Mo YY: Linc-RoR promotes MAPK/ERK signaling and confers estrogen-independent growth of breast cancer. Mol Cancer. 16:1612017. View Article : Google Scholar : PubMed/NCBI

146 

Wu F, Mo Q, Wan X, Dan J and Hu H: NEAT1/has-mir-98 5p/MAPK6 axis is involved in non-small-cell lung cancer (NSCLC) development. J Cell Biochem. Nov 2–2017.Epub ahead of print. View Article : Google Scholar

147 

Peng W and Fan H: Long noncoding RNA CCHE1 indicates a poor prognosis of hepatocellular carcinoma and promotes carcinogenesis via activation of the ERK/MAPK pathway. Biomed Pharmacother. 83:450–455. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 53 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun W, Shi Y, Wang Z, Zhang J, Cai H, Zhang J and Huang D: Interaction of long-chain non-coding RNAs and important signaling pathways on human cancers (Review). Int J Oncol 53: 2343-2355, 2018
APA
Sun, W., Shi, Y., Wang, Z., Zhang, J., Cai, H., Zhang, J., & Huang, D. (2018). Interaction of long-chain non-coding RNAs and important signaling pathways on human cancers (Review). International Journal of Oncology, 53, 2343-2355. https://doi.org/10.3892/ijo.2018.4575
MLA
Sun, W., Shi, Y., Wang, Z., Zhang, J., Cai, H., Zhang, J., Huang, D."Interaction of long-chain non-coding RNAs and important signaling pathways on human cancers (Review)". International Journal of Oncology 53.6 (2018): 2343-2355.
Chicago
Sun, W., Shi, Y., Wang, Z., Zhang, J., Cai, H., Zhang, J., Huang, D."Interaction of long-chain non-coding RNAs and important signaling pathways on human cancers (Review)". International Journal of Oncology 53, no. 6 (2018): 2343-2355. https://doi.org/10.3892/ijo.2018.4575