Topic Highlight Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Dec 8, 2015; 7(28): 2781-2791
Published online Dec 8, 2015. doi: 10.4254/wjh.v7.i28.2781
Long noncoding RNAs in hepatocellular carcinoma: Novel insights into their mechanism
Yong-Ru Liu, Rui-Xue Tang, Wen-Ting Huang, Fang-Hui Ren, Dian-Zhong Luo, Yi-Wu Dang, Gang Chen, Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
Rong-Quan He, Li-Hua Yang, Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
Author contributions: Liu YR, Tang RX, Huang WT, Ren FH, He RQ and Yang LH performed the literature search, wrote the first draft of the manuscript and approved the final version; Luo DZ, Dang YW and Chen G edited the final draft of the manuscript and approved the final version.
Supported by Partly Fund of Guangxi Provincial Health Bureau Scientific Research Project, No. Z2014054; Youth Science Foundation of Guangxi Medical University, No. GXMUYSF201311; Guangxi University Science and Technology Research Projects, No. LX2014075; Natural Science Foundation of Guangxi, Nos. 2015GXNSFBA139157, 2015GXNSFCA139009; and National Natural Science Foundation of China, Nos. NSFC81360327, NSFC81560489, NSFC81560469.
Conflict-of-interest statement: The authors have not declared any conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Gang Chen, MD, PhD, Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China. chen_gang_triones@163.com
Telephone: +86-771-5356534 Fax: +86-771-5358943
Received: April 30, 2015
Peer-review started: May 8, 2015
First decision: September 8, 2015
Revised: November 3, 2015
Accepted: November 24, 2015
Article in press: November 25, 2015
Published online: December 8, 2015

Abstract

Hepatocellular carcinoma (HCC) is the predominant subject of liver malignancies which arouse global concern. Advanced studies have found that long noncoding RNAs (lncRNAs) are differentially expressed in HCC and implicate they may play distinct roles in the pathogenesis and metastasis of HCC. However, the underlying mechanisms remain largely unclear. In this review, we summarized the functions and mechanisms of those known aberrantly expressed lncRNAs identified in human HCC tissues. We hope to enlighten more comprehensive researches on the detailed mechanisms of lncRNAs and their application in clinic, such as being used as diagnostic and prognostic biomarkers and the targets for potential therapy. Although studies on lncRNAs in HCC are still deficient, an improved understanding of the roles played by lncRNAs in HCC will lead to a much more effective utilization of those lncRNAs as novel candidates in early detection, diagnosis, prevention and treatment of HCC.

Key Words: Hepatocellular carcinoma, Long noncoding RNA, Dysregulation, Mechanism, Pathway

Core tip: Hepatocellular carcinoma (HCC) is a global concern. Long noncoding RNAs (lncRNAs) are likely to play crucial roles in various pathogenesis of HCC, including tumor growth, proliferation, invasion, metastasis and recurrence. Here, we focus on recent studies of human HCC associated lncRNAs and highlight their functions, mechanisms, as well as their potential to act as novel candidates for early detection, diagnosis, prevention and treatment of HCC.



INTRODUCTION

Hepatocellular carcinoma (HCC), one of the most common histologic subtype of primary liver cancers, accounting for 70%-85% of liver cancer cases in most countries, is the third-leading cause of worldwide mortality for various cancers[1-3]. It causes nearly 695900 deaths per year, half of which occur in China, as a result of the high chronic hepatitis B virus (HBV) infection incidence[4]. Besides, infection with hepatitis C virus, hepatosteatosis and chronic exposure to toxic chemical substances are the major HCC risk factors[5,6]. Especially, exposure to aflatoxin plays a key role in inducing HCC in our Guangxi Zhuang Autonomous Region[7]. In addition, many key signal transduction pathways have been verified to be involved in the pathogenesis of HCC, including PI3K/Akt/mTOR pathway, Raf/MAPK/ERK pathway, Jak/Stat pathway, WNT-b-catenin pathway, so on and so forth[8-12]. Usually, the treatments for HCC are finite and are only available in the early stage, while most HCC are only detected in their advanced stage when traditional chemotherapy has marginal effects and leads to poor prognosis. Because of its dismal outcome, etiology and carcinogenesis investigation are in urgent need. Recently, human genome analysis in non-protein coding has made new progress. It discovers massive transcription of large RNA transcripts which lack coding protein function, termed as long noncoding RNAs (lncRNAs).

With recent application of next generation sequencing techniques, significant numbers of non-coding RNAs (ncRNAs) and lncRNAs have been discovered to be associated with HCC. And HCC is characterized by dysregulation of numerous gene networks while both protein-coding genes and ncRNA genes are involved, just like many other cancers. It is estimated that protein-coding genes only account for less than 2% of the human genome while nearly 70% of the human genome is transcribed pervasively[13]. Accordingly, ample ncRNAs are transcribed from human genome, for instance lncRNAs, microRNAs (miRNAs), small interfering RNAs and PIWI-interacting RNAs[14-16].

NcRNAs were once thought to be body “garbage” or transcriptional “noise”. However, accumulating reports have demonstrated that miRNAs and lncRNAs play valid regulatory roles in cancer[17-22]. Recent studies[23] have also elucidated that lncRNAs possess a significant role in epigenetic regulation. Via regulating gene expression by miscellaneous mechanisms, including genomic imprinting, chromatin modification, regulation of protein function, transcription and post-transcriptional processing[24-26], lncRNAs are involved in multitudinous physiological functions and pathological processes.

LncRNAs, larger than 200 nucleotides (nt) in length, are commonly defined as endogenous cellular RNA molecules, which are poorly conserved and not capable of being translated into proteins[21,23,27]. They can be monitored by a high-throughput analysis such as transcriptome analysis and microarrays, or through bioinformatics prediction[28]. LncRNAs can be transcribed by RNA polymerase II, and then undergo cotranscriptional modifications including polyadenylation and pre-RNA splicing[29]. Many studies have pointed out that lncRNA transcripts play vital roles in various biological processes as they function in gene imprinting and splicing, chromatin modification, immunesurveillance, cell fate specification, cell cycle control and cell apoptosis, or act as nuclear architecture, subnuclear compartments, RNA processing enhancer and promoter[30,31].

LncRNAs have assorted mechanisms in biological processes. Generally speaking, the role of lncRNA as a gene expressing regulator could be found in transcriptional level and posttranscriptional level. Cis-regulation and trans-regulation are two main transcriptional regulation means, by which lncRNAs can target local and distant genes, respectively. The posttranscriptional regulating mechanism is involved in posttranscriptional process of mRNAs which includes splicing, editing, trafficking, translation and degradation. LncRNAs can also function as competing endogenous RNAs for shared miRNA[32,33]. In brief, there are four known molecular functions of lncRNAs: Signal, decoy, guide, and scaffold[21,34].

There are five species of lncRNAs, listed as follow[31,35,36]: Sense or antisense (when overlapping at least one exon of another transcript on the opposite or same strand), bidirectional (when a neighboring coding transcript or its expression on the opposite strand is initiated in close genomic proximity), intronic (when derived from an intron of a second transcript), and intergenic (when it lies as an independent unit within the genomic interval between two genes). An updated definition was given to lncRNAs by other researchers regardless of their length and non-protein coding capability[37]. It described lncRNAs as RNA molecules who may have the function as primary or spliced transcripts which do not confirm to the known varieties of small RNAs or structural RNAs[38]. In recent years, the number of articles focused on lncRNAs has increased greatly. Recent studies have demonstrated that certain lncRNAs are specifically correlated with certain classes of cancer and the different expression level of lncRNAs may function as an indicator for metastasis and prognosis[39-41]. Genome-wide transcriptomic analyses have found that a number of lncRNAs are dysregulated in HCC cell lines or cancer tissues[21,28,29,42,43]. Given the fact that such large scales of lncRNAs are aberrantly regulated in HCC, it is of highly possibility that lncRNAs are directly associated with carcinogenesis of HCC. In this review, we concentrated on cancer-related lncRNAs which have been validated in human HCC. Furthermore, we summarized their mechanism and signaling pathways in HCC.

REPRESENTATIVE LNCRNAS DYSREGULATED IN HCC

Abnormal expressed lncRNAs have been found to be associated with hepatocarcinogenesis and play a key role in metastasis and prognosis[29,34,42]. Accumulating studies recently have focused on the contributions of lncRNAs in HCC development. Here, we summarized differential expressions of lncRNAs and their potential roles in HCC (Tables 1 and 2).

Table 1 Upregulated long noncoding RNAs in hepatocellular carcinoma.
NameGene locusSize (bp)DysregulationPotential role in HCCRef.
HULC6p24.31638UpregulatedAssociate with HBV infection or histological grade. Associate with tumor growth[22,29,44-50]
H1911p15.52660UpregulatedSuppress progression, metastasis. Promote cell proliferation[22,29,50-57]
TUC33812q13.13590UpregulatedIncreased in liver cirrhosis. Modulate cell growth[58]
MALAT111q13.18708UpregulatedAssociate with tumor metastasis, recurrence[21,22,29,59,60]
HOTAIR12q13.1312649UpregulatedAssociate with invasion and metastasis. Increases chemosensitivity[21,22,29,61-65]
HOTTIP7p15.26839UpregulatedAssociate with tumor progression and disease outcome[28,66-68]
HEIH5q35.31665UpregulatedAssociated with HBV-HCC. Associate with prognosis[21,29,69]
MDIG3q11.230635UpregulatedAssociate with DNA repair and prognosis[70]
PVT18q24.21210626UpregulatedAssociate with HCC progression and predict recurrence[71,72]
Linc0097417q21.314890UpregulatedPredict tumor growth and metastasis[73]
UFC11q23.35113UpregulatedPromote HCC cell proliferation, inhibit cell apoptosis and induce cell cycle progression[10,12,74]
PCNA-AS120p12.3384UpregulatedPromote tumor growth[75]
UCA119p13.127375UpregulatedInvolved in chemotherapeutic resistance. Associatewith TNM stage, metastasis and postoperative survival[50,76]
CCAT18q24.2111887UpregulatedPromotes HCC progression[77-79]
ATB19q13.32895UpregulatedAssociat with poor prognosis[80,81]
URHC2q24.2192173UpregulatedPromote cell proliferation and inhibit apoptosis[82]
Table 2 Downregulated long noncoding RNAs in hepatocellular carcinoma.
NameGene locusSize (bp)DysregulationPotential role in HCCRef.
MEG3 (GTL2)14q32.334919DownregulatedAssociate with methylation. Predictive biomarker for monitoring epigenetic therapy[21,83-87]
LncRNA-LET15q24.12606DownregulatedReduces hepatic invasion and abdominal metastases[88]
PTENP19p13.33917DownregulatedRepress the tumorigenic properties of HCC cells[22,89]
SRHC5p15.316365DownregulatedInhibit cancer proliferation[90]
MT1DP16q131255DownregulatedAct as a tumor suppressor. Overexpression of MT1DP decreases cell proliferation but increases apoptosis[91]
MECHANISMS AND SIGNALING PATHWAY IN HCC

After attentive study, we divided the researched lncRNAs into 3 groups and made the above conclusive table. And with the purpose of identifying the roles of different lncRNAs who might play in the early detection and diagnosis or prevention and treatment of HCC, we further summarized their function and mechanism as follow.

LNCRNAS ASSOCIATED WITH TUMOR GROWTH AND PROLIFERATION IN HCC
Highly upregulated in liver cancer

Highly upregulated in liver cancer (HULC), a well-researched lncRNA who associates with HBV infection and HCC tumor growth is upregulated in HCC and associated with its grades[45]. By viewing dozens of research articles, we found that HULC can accelerate the growth of HCC via downregulating its neighbor gene p18 (known as eukaryotic translation elongation factor 1, EEF1E1 or AIMP3) and can disturb the circadian rhythm of HCC via upregulating oscillator CLOCK[47]. The complementary base which pairs between 5’UTR of CLOCK mRNA and HULC takes responsibility for the modulation of CLOCK mediated by HULC[91]. With regard to the mechanism of HULC upregulation, HBx regulates the transcription of CREB-dependent promoters by interacting with CREB, and then, the transcription factor CREB contributes to the activation of HULC promoter[47,48]. The variant genotypes of rs7763881 in HULC contribute to decreasing HCC susceptibility in persistent HBV carriers. And single nucleotide polymorphisms (SNPs) in HULC contribute to the risk of HBV chronic infection and HCC[92]. Besides, HULC acts as an endogenous “sponge”, who downregulates a series of miRNAs activities, including miR-372. Studies indicate that inhibition of miR-372 results in decreased translational repression of its target gene, PRKACB, and inducing phosphorylation of CREB in turn[48].

Based on the above mechanisms, HULC may have the potential of predicting prognosis in clinical practice. However, only cell lines research were done by researchers, other confirmatory experiments are requisite.

LncRNA-hPVT1

Two studies investigated closely into lncRNA-hPVT1 and concluded that it has a function of promoting cell proliferation, cell cycling and it also functions as an acquisition of stem-cell like contents in HCC cells. LncRNA-hPVT1 upregulates nucleolar protein p120 (NOP2) via enhancing the stability of NOP2 proteins and its above functions depend on the presence of NOP2. Studies show that the transforming growth factor (TGF)-β1/lncRNA-hPVT1/NOP2 pathway is compromised in the progression of HCC. Hence, lncRNA-hPVT1 influences the stem-cell like potential of HCC cells and promotes the growth of HCC. Regulation of the lncRNA-hPVT1/NOP2 pathway has a beneficial effect in the treatment of HCC[71]. More researches are needed to be done in order to find effective therapy targeted at lncRNA-hPVT1.

UFC1

Other than the above three lncRNAs, another well-studied lncRNA associated with HCC proliferation is lncRNA-UFC1 (GenBank Accession No. KJ809564), who promotes HCC cell proliferation, induces cell cycle progression and inhibits cell apoptosis[73]. It induces HuR translocation and by silencing HuR expression can abrogate the function of lncRNA-UFC1 function in HCC. Moreover, lncRNA-UFC1 is targeted by miR-34a and the overexpression of miR-34a significantly suppresses the expression levels of cell cycle related proteins, cellular proliferation and HuR expression in lncRNA-UFC1-overexpressing cells[10]. As molecularly targeted therapies are heated studied, UFC1 offers us a new aspect, clinical research are in the urgent need.

ZNRD1 antisense RNA 1

A large case-control study including 1344 HBV natural-clearance subjects, 1344 HBV persistent carriers and 1300 HBV-positive HCC patients was done[93]. The study found out that ZNRD1 antisense RNA 1 (ZNRD1-AS1) is a crucial regulator of ZNRD1 (human zinc ribbon domain containing 1). In ZNRD1-AS1, several SNPs (nucleotide polymorphisms) is identified as expression quantitative trait loci (eQTLs) SNPs, which are connected with the expression of ZNRD1[94,95]. ZNRD1 is involved in DNA damage and repair via regulating the expression of excision repair cross-complementing 1 (ERCC1)[96], to restrain cell proliferation and to regulate the expression of miRNAs in cancers[97,98]. Furthermore, ZNRD1 eQTLs SNPs in lncRNA ZNRD1-AS1 have an increased risk for persistent HBV-carriers HCC but a protective influence against chronic HBV infection[93]. As a result, the different roles which ZNRD1-AS1 play make a difference in the treatment of HBV-positive HCC patients and HBV-negative HCC patients.

Colon cancer associated transcript-1

Dysregulation of colon cancer associated transcript-1 (CCAT1) is in association with tumor size, microvascular invasion, AFP and prognosis in patients with HCC. Besides, it is demonstrated that in vitro CCAT1 could promote proliferation and migration in HCC by binding to let-7, which contributes to the up-regulation of HMGA2 and c-Myc[76]. Herein, the complex of CCAT1 and let-7 may have the diagnostic function in early detection of HCC and its migration.

Maternally expressed gene 3

Maternally expressed gene 3 (MEG3) regulates tumor cell proliferation and apoptosis in HCC partially through the accumulation of p53[83]. UHRF1, as a new identified oncogene, contributes to the upregulation of MEG3 in HCC by regulating DNMT1, while upregulation of MEG3 in HCC cells can partially diminish the promotion of proliferation produced by UHRF1. In addition, UHRF1/DNMT1/MEG3/p53 axis signaling pathway is involved in HCC progression[99]. Furthermore, loss of MEG3 gene expression is related to hypermethylation of the promoter region in HCC[82]. Impressively, enforced expression of MEG3 in HCC remarkably decreases both anchorage-independent and anchorage-dependent cell growth, and induces cell apoptosis[83]. Associated with anti-oncogene p53, MEG3 shows a promising future in being one of the therapeutic targets for HCC treatment.

PTENP1

The over-expression of PTENP1 (a pseudogene of PTEN) represses the oncogenic PI3K/AKT pathway and elicits pro-death autophagy by sequestering miR-20a, miR-19b and miR-17 in vitro. It also inhibits tumor growth in vivo. These are accompanied by dampened angiogenesis or neovasculature maturation, enhanced apoptosis and autophagy[88]. It’s necessary to do further in vivo experiments to confirm its detailed mechanism.

The above 7 lncRNAs have been widely studied, and they are considered to be associated with tumor growth and proliferation in HCC. With further clinical trials, their application in the prediction and diagnosis of HCC would be possible.

LNCRNAS ASSOCIATED WITH METASTASIS AND PROGNOSIS IN HCC

Invasion and metastasis often adumbrate an advanced stage while recurrence often indicates a poor prognosis. It’s the same in HCC development. The following lncRNAs were found associated with metastases and recurrence which may predict a dismal outcome.

H19

A dozen of studies have elucidated that H19 serves as a potential prognostic marker as well as potential target for HCC therapy. By decreasing the expression of markers for epithelial-to-mesenchymal transition, such as claudin 1, cytokeratin-8 (KRT-8), KRT-19 and CDH1 (E-cadherin), H19 suppresses the progression of HCC. By mediating hnRNPU/PCAF/RNAPol II, H19 suppresses the migration of HCC. By increasing histone acetylation, H19 can epigenetically activates miR-200 family, and thus, it suppresses HCC metastasis[50]. Moreover, the identification of AKT/GSK-3β/Cdc25A signaling pathway as the downstream signaling pathway of H19 explains the molecular mechanism of metastasis and invasion in HCC[100]. And it has also been demonstrated that the deletion of H19 endodermal enhancer can regulate expression of insulin-like growth factor 2 (IGF2) and H19 in the early stage of liver carcinogenesis as well as that paternal inheritance of the deletion of H19 endodermal enhancer can delay tumor formation by increasing apoptosis of the hepatocytes and reducing IGF2 expression[101]. In the main, by various signaling pathway, H19 can be a promising indicator for prognosis and can be targeted in the treatment of HCC.

Metastasis-associated lung adenocarcinoma transcript 1

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a critical regulator of maintaining the transformative phenotype in HCC, it associates with tumor metastasis and recurrence[102]. MALAT1 is a genuine target gene of the Wnt/TCF/β-catenin and Hippo/yes associated protein (YAP) signaling pathway. It is negatively regulated by SRSF1 via the following two pathways. The first pathway is by accelerating the degradation of MALAT1 which is blocked by YAP at the stage of post-transcriptional. When overexpressed, YAP stimulates the translocation of SRSF1 from the nucleus to the cytoplasm, thus the nuclear-retained MALAT1 avoids degradation[103]. The second pathway is by binding to YAP. SRSF1 inhibits the transcriptional activity of YAP and prevents the recruitment of YAP on the MALAT1 promoter at a transcriptional stage[104].

MALAT1 can interact with the arginine/serine SR proteins and modulate their distribution to the nuclear speckles. Furthermore, MALAT1 can regulate alternative splicing of pre-mRNAs via controlling active SR proteins’ levels[103].

To sum up, by participating in different pathway, MALAT1 shows a crucial role in the carcinogenesis of HCC which lays the foundation of early detection of metastases and assessment of recurrence.

HOX transcript antisense RNA

High expression of HOX transcript antisense RNA (HOTAIR) indicates a notably poorer prognosis with respect to overall survival (OS) and a remarkably larger tumor size in HCC patients[62]. HOTAIR is selectively required to target polycomb repressive complex 2 (PRC2) occupancy, thus, induces histone H3 tri-methylated at lysine 27 (H3K27) trimethylation and silenced transcription of the HOXD locus[105]. The 5’ domain of HOTAIR can bind to PRC2, while the 3’ domain of HOTAIR can bind to the LSD1 (lysine specific demethylase 1)/CoREST (Co-repressor of RE1-silencing transcription factor)/REST complex; after that, the complexes are targeted and assembled to the HOXD locus; and coordinately regulate histone H3K27 methylation and histone histone 3 methylated at lysine 4 demethylation; consequently, the transcription across 40 kb of the HOXD locus is silenced in trans by DNA methylation[106]. These indicate that the HOTAIR-induced assembly and targeting of LSD1 and PRC2 complexes is a general mechanism for gene silencing across the genome, which plays a vital role in the association with invasion and metastasis in HCC[64]. Expression of HOTAIR is also associated with tumor size and lymph node metastasis in HCC patients[62]. Knockdown of HOTAIR reduces the levels of matrix metallopeptidase 9 and vascular endothelial growth factor, which play an important role in metastasis and cell motility[60].

In addition, HOTAIR is found to promote invasion and migration in HCC by inhibiting RBM38. Silencing of RBM38 restores cell motility, while knockdown of HOTAIR conspicuously reduces cell motility as the downregulation of HOTAIR increases the expression of RBM38 both on mRNA levels and protein levels[63].

With its association with tumor size, tumor metastasis and OS, HOTAIR is regarded as one of the prognosis indicator. It’s of great potential value to use it in the clinic in the future.

HOTTIP

HOTTIP (the transcript of HOXA at the distal tip with 3.8 kb), transcribed from the 5’ tip of the HOXA locus, coordinates the activation of some 5’HOXA genes and is negatively regulated by miR-125b. High HOTTIP expression indicates an increased metastasis formation and a decreased overall survival[65]. HOTTIP binds the adaptor protein WDR5 directly and targets WDR5/MLL complexes across HOXA, thereby driving histone H3 lysine 4 trimethylation and the gene transcription and influences HCC cell proliferation rates as a result[27]. Its overexpression contributes to hepatocarcinogenesis through regulating the expression of its neighboring protein-coding genes[67]. With these function, HOTTIP can be potentially used in clinic as a prognosis predictor.

LncRNA high expression in HCC

LncRNA high expression in HCC (HEIH), associated with recurrence and overall survival in HBV-related HCC, is overexpressed in HCC tissues[68]. HEIH influences the expression of enhancer of zeste homolog 2 (EZH2) target genes by increasing the binding of EZH2 levels. Downregulation of HEIH induces G(0)/G(1) arrest which is caused by the interaction of EZH2 with HEIH. Thus, level of HEIH is significantly associated with recurrence and is an independent prognostic factor for survival in HCC[68]. In summary, HEIH connects with HBV-related HCC and plays a role as the risk factor for HCC recurrence. Therefore, predicting the possibility of HCC recurrence by detecting the relative change of the HBV virus and HEIH level in serum is worth further studying.

ATB

Overexpression of lncRNA-ATB (lncRNA-activated by TGF-β) significantly correlates with EMT gene signature expression, macrovascular invasion, microvascular invasion, portal vein tumor thrombus and encapsulation. Moreover, higher expression of lncRNA-ATB is significantly correlated with shorter recurrence-free survival and overall survival, suggesting that lncRNA-ATB contributes to HCC progression[27].

LncRNA-ATB upregulates ZEB1 and ZEB2 via competitively binding with the miR-200 family, then it induces EMT and invasion. Besides, lncRNA-ATB promotes organ colonization of disseminated HCC tumor cells through binding with autocrine induction of interleukin 11 (IL-11), IL-11 mRNA and triggering signal transducer and activator of transcription 3 signaling[79,80]. On the whole, lncRNA-ATB facilitates the invasion-metastasis cascade, which makes it a predictor for HCC prognosis.

LncRNA-p21

LncRNA-p21, located upstream of CDKN1A gene, who triggers apoptosis in HCC, is a transcriptional target of p53 and p53 gene is an important tumor suppressor gene[107,108]. Importantly, lncRNA-p21 can bind to heterogeneous nuclear ribonucleoprotein K (hnRNP-K), therefore it contributes to the localization of hnRNP-K and transcriptional repression of p53-regulated genes[109] and as a result, triggers apoptosis. Theoretically, lncRNA-p21 should be down-regulated in HCC, however, corroborating experiments are needed to delineate the exact underlying mechanism. As the transcriptional target of anti-oncogene p53, p21 shows a promising future in being one of the therapeutic targets for inducing cellular apoptosis.

The above 7 lncRNAs are considered to be associated with metastasis and prognosis in HCC. Although many researches have been complicated, studies with more cases and further ward clinical trials would be needed in order to develop novel therapeutics and treatment for HCC patients.

LATEST FIND OF LNCRNAS RELATED TO HCC
Linc00974

Dysregulation of Linc00974 increases KRT19 levels, which results in the activation of both TGF-β and Notch signaling pathways, which causes the invasion and proliferation of HCC both in vivo and in vitro. Linc00974 influenced KRT19 expression by interacting with miR-642[72]. Being significantly correlated with tumor differentiation grade, size and metastasis makes Linc00974 a feasible predictor in the carcinogenesis of HCC.

Up-regulated in hepatocellular carcinoma

High level of up-regulated in hepatocellular carcinoma (URHC) expression is significantly associated with tumor size, tumor number and shorter overall survival after surgery[81]. URHC can regulate cell proliferation and apoptosis by repressing ZAK expression. ZAK, also known as MLK-like MAP triple kinase-α or ZAK-α, belongs to the mixed lineage kinase family and functions as a tumor-suppressor gene in HCC[110]. Inactivation of the ERK/MAPK pathway is required for the increase in HCC growth, which is induced by URHC-ZAK regulation[81]. However, only microarray analysis was done, which limited its conviction. Experiments in vivo and in vitro are desperately needed.

SRHC

SRHC (NCBI No: uc003jdr) is an important downstream target gene of HNF-4A and it is correlated with α-fetoprotein (AFP) levels and the degree of differentiated tumors[89]. SRHC is combined with HNF-4A to promote its transcription, thus inhibiting the proliferation of tumor cells and promoting cell differentiation in HCC[89]. Serum AFP level monitoring is well developed and have been used as a standardized index of HCC diagnosis, therefore, detecting SRHC in predicting HCC may have great value in clinical practice as well.

Metallothionein 1D, pseudogene

Metallothionein 1D, pseudogene (MT1DP) inhibits tumor cell growth could be rescued by a combination of overexpression of Runt related transcription factor 2, FoxA1 and YAP. In addition, MT1DP inhibited transformative phenotype of liver cancer cells and cell proliferation by reducing protein synthesis of FoxA1[90]. With this inhibiting function, it’s of great research value whether MT1DP can be a potential therapeutic target in the treatment of HCC or not.

LncRNA low expression in tumor

LncRNA low expression in tumor (LncRNA-LET) plays a decisive role in hypoxia-induced metastasis in HCC through a hypoxia-inducible factor 1, alpha subunit (HIF-1a)/histone deacetylase 3 (HDAC3)/LET/NF90 pathway[110]. Precisely, HDAC3 represses LET by decreasing the LET promoter region’s histone acetylation-mediated modulation under hypoxic conditions. And then, downregulation of LET recedes the direct interactions between LET and NF90, then enhances the stabilization of NF90 and increases the expression of HIF-1a (a target mRNA of NF90 involved in hypoxia-induced metastasis). As a result, LET inhibits the metastasis of HCC via this positive feedback loop[87]. Therefore, when LET is downregulated, patients usually face a poor prognosis.

For these 5 newly found lncRNAs, investigations with more preclinical models of HCC would be desired in order to further strengthen the conclusions and provide a more rational support for ward clinical application.

CONCLUSION

In conclusion, lncRNAs play a crucial role in various biological processes in HCC, such as initiation, progression, metastasis, treatment and prognosis.

Two latest published articles also reached the same conclusion. On the basis of dozens of studies, Yang et al[111] discussed the probable molecular mechanisms depended on lncRNA level change, and drew the conclusion that lncRNAs can be applied in HCC diagnosis and treatment. However, their summary was omissive as they only analyzed the upregulated lncRNAs, and even some upregulated lncRNAs were left out, such as HOTTIP, MVIH, UFC1, UCA1, CCAT1, etc. In our article, we not only analyzed the differential expression of lncRNAs in human HCC (upregulated as well as downregulated), but also summarized their specific mechanisms and pathways and gave an outlook in their potential as candidates in diagnosis and treatment of HCC. Another review[111] provided different lncRNAs compared to us, such as RP11-160H22.5, XLOC_014172 and LOC149086. However, these lncRNAs were not embodied in NCBI database, and their mechanisms were unavailable, which brought us new research direction.

Although plenty of studies[28,112-115] cast light on the characteristics and mechanisms of different lncRNAs involved in HCC as we summarized above, till now, research on lncRNA still remains in its infancy and a large portion of lncRNAs surely remains to be further discovered. As systematic identification of lncRNAs and their well-understanding of mechanisms can pave the way for early diagnosis and therapeutics designing for HCC, there is still a long way to go in the research field of HCC-related lncRNAs.

Continuous researches are needed to verify the detailed function and mechanisms of revealed lncRNAs and to find innovative lncRNAs and sequentially, the predictive and diagnostic roles of lncRNAs in HCC can be validated. Thereby, diagnosis of HCC in an early stage and controlling its development and progression will become possible. Further clinical and ward trials are also in the urge, so that we can develop therapeutic roles of lncRNAs in HCC. In a word, future studies should aim at investigating how can a discovered lncRNA be used to identify HCC patients and used as a guidance being applied in treatment to have optimal responses and to reduce the likelihood of relapse.

Footnotes

P- Reviewer: Dalay N, Slomiany BL, Tomizawa M S- Editor: Ji FF L- Editor: A E- Editor: Liu SQ

References
1.  Schmieder R, Puehler F, Neuhaus R, Kissel M, Adjei AA, Miner JN, Mumberg D, Ziegelbauer K, Scholz A. Allosteric MEK1/2 inhibitor refametinib (BAY 86-9766) in combination with sorafenib exhibits antitumor activity in preclinical murine and rat models of hepatocellular carcinoma. Neoplasia. 2013;15:1161-1171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (1)]
2.  Su JC, Tseng PH, Wu SH, Hsu CY, Tai WT, Li YS, Chen IT, Liu CY, Chen KF, Shiau CW. SC-2001 overcomes STAT3-mediated sorafenib resistance through RFX-1/SHP-1 activation in hepatocellular carcinoma. Neoplasia. 2014;16:595-605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
3.  Xia H, Ooi LL, Hui KM. MicroRNA-216a/217-induced epithelial-mesenchymal transition targets PTEN and SMAD7 to promote drug resistance and recurrence of liver cancer. Hepatology. 2013;58:629-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 315]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
4.  El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology. 2007;132:2557-2576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3846]  [Cited by in F6Publishing: 4102]  [Article Influence: 241.3]  [Reference Citation Analysis (2)]
5.  Sakurai T, Kudo M, Umemura A, He G, Elsharkawy AM, Seki E, Karin M. p38α inhibits liver fibrogenesis and consequent hepatocarcinogenesis by curtailing accumulation of reactive oxygen species. Cancer Res. 2013;73:215-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
6.  Zhang T, Zhang J, Cui M, Liu F, You X, Du Y, Gao Y, Zhang S, Lu Z, Ye L. Hepatitis B virus X protein inhibits tumor suppressor miR-205 through inducing hypermethylation of miR-205 promoter to enhance carcinogenesis. Neoplasia. 2013;15:1282-1291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
7.  Qi LN, Bai T, Chen ZS, Wu FX, Chen YY, De Xiang B, Peng T, Han ZG, Li LQ. The p53 mutation spectrum in hepatocellular carcinoma from Guangxi, China : role of chronic hepatitis B virus infection and aflatoxin B1 exposure. Liver Int. 2015;35:999-1009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 60]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
8.  Aravalli RN, Cressman EN, Steer CJ. Cellular and molecular mechanisms of hepatocellular carcinoma: an update. Arch Toxicol. 2013;87:227-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 166]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
9.  Shin JW, Chung YH. Molecular targeted therapy for hepatocellular carcinoma: current and future. World J Gastroenterol. 2013;19:6144-6155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 5.8]  [Reference Citation Analysis (2)]
10.  Cao C, Sun J, Zhang D, Guo X, Xie L, Li X, Wu D, Liu L. The long intergenic noncoding RNA UFC1, a target of MicroRNA 34a, interacts with the mRNA stabilizing protein HuR to increase levels of β-catenin in HCC cells. Gastroenterology. 2015;148:415-426.e18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 199]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
11.  Sherwood V. WNT signaling: an emerging mediator of cancer cell metabolism? Mol Cell Biol. 2015;35:2-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 113]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
12.  Collins JF. Long noncoding RNAs and hepatocellular carcinoma. Gastroenterology. 2015;148:291-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
13.  Huang B, Zhang R. Regulatory non-coding RNAs: revolutionizing the RNA world. Mol Biol Rep. 2014;41:3915-3923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
14.  Gehrau RC, Mas VR, Villamil FG, Dumur CI, Mehta NK, Suh JL, Maluf DG. MicroRNA signature at the time of clinical HCV recurrence associates with aggressive fibrosis progression post-liver transplantation. Am J Transplant. 2013;13:729-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
15.  Dong P, Yu F, Fan X, Lin Z, Chen Y, Li J. Inhibition of ATIR by shRNA prevents collagen synthesis in hepatic stellate cells. Mol Cell Biochem. 2010;344:195-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
16.  Gomes AQ, Nolasco S, Soares H. Non-coding RNAs: multi-tasking molecules in the cell. Int J Mol Sci. 2013;14:16010-16039.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 179]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
17.  Li X, Wu J, Zheng J, Li Y, Yang T, Hu G, Dai J, Yang Q, Dai L, Jiang Y. Altered miRNA expression profiles and miR-1a associated with urethane-induced pulmonary carcinogenesis. Toxicol Sci. 2013;135:63-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
18.  Zheng L, Pu J, Qi T, Qi M, Li D, Xiang X, Huang K, Tong Q. miRNA-145 targets v-ets erythroblastosis virus E26 oncogene homolog 1 to suppress the invasion, metastasis, and angiogenesis of gastric cancer cells. Mol Cancer Res. 2013;11:182-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 71]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
19.  Wang Y, Zhang X, Li H, Yu J, Ren X. The role of miRNA-29 family in cancer. Eur J Cell Biol. 2013;92:123-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 151]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
20.  Li J, Kong X, Zhang J, Luo Q, Li X, Fang L. MiRNA-26b inhibits proliferation by targeting PTGS2 in breast cancer. Cancer Cell Int. 2013;13:7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 73]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
21.  He Y, Meng XM, Huang C, Wu BM, Zhang L, Lv XW, Li J. Long noncoding RNAs: Novel insights into hepatocelluar carcinoma. Cancer Lett. 2014;344:20-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 283]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
22.  Qiu MT, Hu JW, Yin R, Xu L. Long noncoding RNA: an emerging paradigm of cancer research. Tumour Biol. 2013;34:613-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 308]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
23.  Cheetham SW, Gruhl F, Mattick JS, Dinger ME. Long noncoding RNAs and the genetics of cancer. Br J Cancer. 2013;108:2419-2425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 582]  [Cited by in F6Publishing: 596]  [Article Influence: 54.2]  [Reference Citation Analysis (0)]
24.  Huang JF, Guo YJ, Zhao CX, Yuan SX, Wang Y, Tang GN, Zhou WP, Sun SH. Hepatitis B virus X protein (HBx)-related long noncoding RNA (lncRNA) down-regulated expression by HBx (Dreh) inhibits hepatocellular carcinoma metastasis by targeting the intermediate filament protein vimentin. Hepatology. 2013;57:1882-1892.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 268]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
25.  Pandey RR, Mondal T, Mohammad F, Enroth S, Redrup L, Komorowski J, Nagano T, Mancini-Dinardo D, Kanduri C. Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol Cell. 2008;32:232-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 900]  [Cited by in F6Publishing: 890]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
26.  Wang X, Arai S, Song X, Reichart D, Du K, Pascual G, Tempst P, Rosenfeld MG, Glass CK, Kurokawa R. Induced ncRNAs allosterically modify RNA-binding proteins in cis to inhibit transcription. Nature. 2008;454:126-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 739]  [Cited by in F6Publishing: 757]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
27.  Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, Lajoie BR, Protacio A, Flynn RA, Gupta RA. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature. 2011;472:120-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1461]  [Cited by in F6Publishing: 1505]  [Article Influence: 115.8]  [Reference Citation Analysis (0)]
28.  Wang Z, Li X. The role of noncoding RNA in hepatocellular carcinoma. Gland Surg. 2013;2:25-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 19]  [Reference Citation Analysis (0)]
29.  Shibata C, Otsuka M, Kishikawa T, Ohno M, Yoshikawa T, Takata A, Koike K. Diagnostic and therapeutic application of noncoding RNAs for hepatocellular carcinoma. World J Hepatol. 2015;7:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
30.  Yan B, Wang Z. Long noncoding RNA: its physiological and pathological roles. DNA Cell Biol. 2012;31 Suppl 1:S34-S41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
31.  Isin M, Dalay N. LncRNAs and neoplasia. Clin Chim Acta. 2015;444:280-288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 77]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
32.  Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell. 2011;146:353-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4127]  [Cited by in F6Publishing: 5080]  [Article Influence: 390.8]  [Reference Citation Analysis (0)]
33.  Li X, Wang Z. The role of noncoding RNA in thyroid cancer. Gland Surg. 2012;1:146-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
34.  Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell. 2011;43:904-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2963]  [Cited by in F6Publishing: 3284]  [Article Influence: 252.6]  [Reference Citation Analysis (0)]
35.  Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136:629-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3469]  [Cited by in F6Publishing: 3785]  [Article Influence: 252.3]  [Reference Citation Analysis (0)]
36.  Ma L, Bajic VB, Zhang Z. On the classification of long non-coding RNAs. RNA Biol. 2013;10:925-933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 755]  [Cited by in F6Publishing: 866]  [Article Influence: 78.7]  [Reference Citation Analysis (0)]
37.  Spizzo R, Almeida MI, Colombatti A, Calin GA. Long non-coding RNAs and cancer: a new frontier of translational research? Oncogene. 2012;31:4577-4587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 717]  [Cited by in F6Publishing: 816]  [Article Influence: 68.0]  [Reference Citation Analysis (0)]
38.  Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10:155-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3924]  [Cited by in F6Publishing: 4236]  [Article Influence: 282.4]  [Reference Citation Analysis (0)]
39.  Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464:1071-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3723]  [Cited by in F6Publishing: 4051]  [Article Influence: 289.4]  [Reference Citation Analysis (0)]
40.  Matouk IJ, Abbasi I, Hochberg A, Galun E, Dweik H, Akkawi M. Highly upregulated in liver cancer noncoding RNA is overexpressed in hepatic colorectal metastasis. Eur J Gastroenterol Hepatol. 2009;21:688-692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 158]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
41.  Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003;22:8031-8041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1584]  [Cited by in F6Publishing: 1716]  [Article Influence: 81.7]  [Reference Citation Analysis (0)]
42.  Huang JL, Zheng L, Hu YW, Wang Q. Characteristics of long non-coding RNA and its relation to hepatocellular carcinoma. Carcinogenesis. 2014;35:507-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 155]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
43.  Ma L, Chua MS, Andrisani O, So S. Epigenetics in hepatocellular carcinoma: an update and future therapy perspectives. World J Gastroenterol. 2014;20:333-345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 75]  [Cited by in F6Publishing: 73]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
44.  Zhao Y, Guo Q, Chen J, Hu J, Wang S, Sun Y. Role of long non-coding RNA HULC in cell proliferation, apoptosis and tumor metastasis of gastric cancer: a clinical and in vitro investigation. Oncol Rep. 2014;31:358-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 218]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
45.  Xie H, Ma H, Zhou D. Plasma HULC as a promising novel biomarker for the detection of hepatocellular carcinoma. Biomed Res Int. 2013;2013:136106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 218]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
46.  Hämmerle M, Gutschner T, Uckelmann H, Ozgur S, Fiskin E, Gross M, Skawran B, Geffers R, Longerich T, Breuhahn K. Posttranscriptional destabilization of the liver-specific long noncoding RNA HULC by the IGF2 mRNA-binding protein 1 (IGF2BP1). Hepatology. 2013;58:1703-1712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 193]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
47.  Du Y, Kong G, You X, Zhang S, Zhang T, Gao Y, Ye L, Zhang X. Elevation of highly up-regulated in liver cancer (HULC) by hepatitis B virus X protein promotes hepatoma cell proliferation via down-regulating p18. J Biol Chem. 2012;287:26302-26311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 274]  [Cited by in F6Publishing: 306]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
48.  Wang J, Liu X, Wu H, Ni P, Gu Z, Qiao Y, Chen N, Sun F, Fan Q. CREB up-regulates long non-coding RNA, HULC expression through interaction with microRNA-372 in liver cancer. Nucleic Acids Res. 2010;38:5366-5383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 744]  [Cited by in F6Publishing: 812]  [Article Influence: 58.0]  [Reference Citation Analysis (0)]
49.  Yang Z, Lu Y, Xu Q, Tang B, Park CK, Chen X. HULC and H19 Played Different Roles in Overall and Disease-Free Survival from Hepatocellular Carcinoma after Curative Hepatectomy: A Preliminary Analysis from Gene Expression Omnibus. Dis Markers. 2015;2015:191029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
50.  Zhang L, Yang F, Yuan JH, Yuan SX, Zhou WP, Huo XS, Xu D, Bi HS, Wang F, Sun SH. Epigenetic activation of the MiR-200 family contributes to H19-mediated metastasis suppression in hepatocellular carcinoma. Carcinogenesis. 2013;34:577-586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 269]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
51.  Shi X, Sun M, Liu H, Yao Y, Song Y. Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett. 2013;339:159-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 823]  [Cited by in F6Publishing: 912]  [Article Influence: 82.9]  [Reference Citation Analysis (0)]
52.  Lv J, Yu YQ, Li SQ, Luo L, Wang Q. Aflatoxin B1 promotes cell growth and invasion in hepatocellular carcinoma HepG2 cells through H19 and E2F1. Asian Pac J Cancer Prev. 2014;15:2565-2570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
53.  Tsang WP, Kwok TT. Riboregulator H19 induction of MDR1-associated drug resistance in human hepatocellular carcinoma cells. Oncogene. 2007;26:4877-4881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 158]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
54.  Iizuka N, Oka M, Yamada-Okabe H, Mori N, Tamesa T, Okada T, Takemoto N, Tangoku A, Hamada K, Nakayama H. Comparison of gene expression profiles between hepatitis B virus- and hepatitis C virus-infected hepatocellular carcinoma by oligonucleotide microarray data on the basis of a supervised learning method. Cancer Res. 2002;62:3939-3944.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Ariel I, Miao HQ, Ji XR, Schneider T, Roll D, de Groot N, Hochberg A, Ayesh S. Imprinted H19 oncofetal RNA is a candidate tumour marker for hepatocellular carcinoma. Mol Pathol. 1998;51:21-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 106]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
56.  Gabory A, Jammes H, Dandolo L. The H19 locus: role of an imprinted non-coding RNA in growth and development. Bioessays. 2010;32:473-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 461]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
57.  Braconi C, Valeri N, Kogure T, Gasparini P, Huang N, Nuovo GJ, Terracciano L, Croce CM, Patel T. Expression and functional role of a transcribed noncoding RNA with an ultraconserved element in hepatocellular carcinoma. Proc Natl Acad Sci USA. 2011;108:786-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 178]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
58.  Lai MC, Yang Z, Zhou L, Zhu QQ, Xie HY, Zhang F, Wu LM, Chen LM, Zheng SS. Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol. 2012;29:1810-1816.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 464]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
59.  Lin R, Maeda S, Liu C, Karin M, Edgington TS. A large noncoding RNA is a marker for murine hepatocellular carcinomas and a spectrum of human carcinomas. Oncogene. 2007;26:851-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 432]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
60.  Geng YJ, Xie SL, Li Q, Ma J, Wang GY. Large intervening non-coding RNA HOTAIR is associated with hepatocellular carcinoma progression. J Int Med Res. 2011;39:2119-2128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 316]  [Article Influence: 26.3]  [Reference Citation Analysis (0)]
61.  Yang Z, Zhou L, Wu LM, Lai MC, Xie HY, Zhang F, Zheng SS. Overexpression of long non-coding RNA HOTAIR predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation. Ann Surg Oncol. 2011;18:1243-1250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 530]  [Cited by in F6Publishing: 595]  [Article Influence: 45.8]  [Reference Citation Analysis (0)]
62.  Ishibashi M, Kogo R, Shibata K, Sawada G, Takahashi Y, Kurashige J, Akiyoshi S, Sasaki S, Iwaya T, Sudo T. Clinical significance of the expression of long non-coding RNA HOTAIR in primary hepatocellular carcinoma. Oncol Rep. 2013;29:946-950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 193]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
63.  Ding C, Cheng S, Yang Z, Lv Z, Xiao H, Du C, Peng C, Xie H, Zhou L, Wu J. Long non-coding RNA HOTAIR promotes cell migration and invasion via down-regulation of RNA binding motif protein 38 in hepatocellular carcinoma cells. Int J Mol Sci. 2014;15:4060-4076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 132]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
64.  Cai B, Song XQ, Cai JP, Zhang S. HOTAIR: a cancer-related long non-coding RNA. Neoplasma. 2014;61:379-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 130]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
65.  Quagliata L, Matter MS, Piscuoglio S, Arabi L, Ruiz C, Procino A, Kovac M, Moretti F, Makowska Z, Boldanova T. Long noncoding RNA HOTTIP/HOXA13 expression is associated with disease progression and predicts outcome in hepatocellular carcinoma patients. Hepatology. 2014;59:911-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 341]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
66.  Cillo C, Schiavo G, Cantile M, Bihl MP, Sorrentino P, Carafa V, D’ Armiento M, Roncalli M, Sansano S, Vecchione R. The HOX gene network in hepatocellular carcinoma. Int J Cancer. 2011;129:2577-2587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
67.  Tsang FH, Au SL, Wei L, Fan DN, Lee JM, Wong CC, Ng IO, Wong CM. Long non-coding RNA HOTTIP is frequently up-regulated in hepatocellular carcinoma and is targeted by tumour suppressive miR-125b. Liver Int. 2015;35:1597-1606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 117]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
68.  Yang F, Zhang L, Huo XS, Yuan JH, Xu D, Yuan SX, Zhu N, Zhou WP, Yang GS, Wang YZ. Long noncoding RNA high expression in hepatocellular carcinoma facilitates tumor growth through enhancer of zeste homolog 2 in humans. Hepatology. 2011;54:1679-1689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 535]  [Article Influence: 41.2]  [Reference Citation Analysis (0)]
69.  Chen B, Yu M, Chang Q, Lu Y, Thakur C, Ma D, Yi Z, Chen F. Mdig de-represses H19 large intergenic non-coding RNA (lincRNA) by down-regulating H3K9me3 and heterochromatin. Oncotarget. 2013;4:1427-1437.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Ding C, Yang Z, Lv Z, DU C, Xiao H, Peng C, Cheng S, Xie H, Zhou L, Wu J. Long non-coding RNA PVT1 is associated with tumor progression and predicts recurrence in hepatocellular carcinoma patients. Oncol Lett. 2015;9:955-963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 106]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
71.  Wang F, Yuan JH, Wang SB, Yang F, Yuan SX, Ye C, Yang N, Zhou WP, Li WL, Li W. Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology. 2014;60:1278-1290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 348]  [Article Influence: 34.8]  [Reference Citation Analysis (0)]
72.  Tang J, Zhuo H, Zhang X, Jiang R, Ji J, Deng L, Qian X, Zhang F, Sun B. A novel biomarker Linc00974 interacting with KRT19 promotes proliferation and metastasis in hepatocellular carcinoma. Cell Death Dis. 2014;5:e1549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 142]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
73.  Liu H, Gong M, French BA, Li J, Tillman B, French SW. Mallory-Denk Body (MDB) formation modulates Ufmylation expression epigenetically in alcoholic hepatitis (AH) and non-alcoholic steatohepatitis (NASH). Exp Mol Pathol. 2014;97:477-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
74.  Yuan SX, Tao QF, Wang J, Yang F, Liu L, Wang LL, Zhang J, Yang Y, Liu H, Wang F. Antisense long non-coding RNA PCNA-AS1 promotes tumor growth by regulating proliferating cell nuclear antigen in hepatocellular carcinoma. Cancer Lett. 2014;349:87-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 83]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
75.  Wang F, Ying HQ, He BS, Pan YQ, Deng QW, Sun HL, Chen J, Liu X, Wang SK. Upregulated lncRNA-UCA1 contributes to progression of hepatocellular carcinoma through inhibition of miR-216b and activation of FGFR1/ERK signaling pathway. Oncotarget. 2015;6:7899-7917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 305]  [Article Influence: 38.1]  [Reference Citation Analysis (0)]
76.  Deng L, Yang SB, Xu FF, Zhang JH. Long noncoding RNA CCAT1 promotes hepatocellular carcinoma progression by functioning as let-7 sponge. J Exp Clin Cancer Res. 2015;34:18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 233]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
77.  Zhu H, Zhou X, Chang H, Li H, Liu F, Ma C, Lu J. CCAT1 promotes hepatocellular carcinoma cell proliferation and invasion. Int J Clin Exp Pathol. 2015;8:5427-5434.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Zhu HQ, Zhou X, Chang H, Li HG, Liu FF, Ma CQ, Lu J. Aberrant Expression of CCAT1 Regulated by c-Myc Predicts the Prognosis of Hepatocellular Carcinoma. Asian Pac J Cancer Prev. 2015;16:5181-5185.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Sun T, Wong N. Transforming growth factor-β-induced long noncoding RNA promotes liver cancer metastasis via RNA-RNA crosstalk. Hepatology. 2015;61:722-724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
80.  Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC. A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 2014;25:666-681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1118]  [Cited by in F6Publishing: 1223]  [Article Influence: 122.3]  [Reference Citation Analysis (0)]
81.  Xu WH, Zhang JB, Dang Z, Li X, Zhou T, Liu J, Wang DS, Song WJ, Dou KF. Long non-coding RNA URHC regulates cell proliferation and apoptosis via ZAK through the ERK/MAPK signaling pathway in hepatocellular carcinoma. Int J Biol Sci. 2014;10:664-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 83]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
82.  Anwar SL, Krech T, Hasemeier B, Schipper E, Schweitzer N, Vogel A, Kreipe H, Lehmann U. Loss of imprinting and allelic switching at the DLK1-MEG3 locus in human hepatocellular carcinoma. PLoS One. 2012;7:e49462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 107]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
83.  Braconi C, Kogure T, Valeri N, Huang N, Nuovo G, Costinean S, Negrini M, Miotto E, Croce CM, Patel T. microRNA-29 can regulate expression of the long non-coding RNA gene MEG3 in hepatocellular cancer. Oncogene. 2011;30:4750-4756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 487]  [Cited by in F6Publishing: 529]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
84.  Huang J, Zhang X, Zhang M, Zhu JD, Zhang YL, Lin Y, Wang KS, Qi XF, Zhang Q, Liu GZ. Up-regulation of DLK1 as an imprinted gene could contribute to human hepatocellular carcinoma. Carcinogenesis. 2007;28:1094-1103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 82]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
85.  Zhang X, Zhou Y, Mehta KR, Danila DC, Scolavino S, Johnson SR, Klibanski A. A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J Clin Endocrinol Metab. 2003;88:5119-5126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 318]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
86.  Zhou Y, Zhong Y, Wang Y, Zhang X, Batista DL, Gejman R, Ansell PJ, Zhao J, Weng C, Klibanski A. Activation of p53 by MEG3 non-coding RNA. J Biol Chem. 2007;282:24731-24742.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 454]  [Cited by in F6Publishing: 483]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
87.  Yang F, Huo XS, Yuan SX, Zhang L, Zhou WP, Wang F, Sun SH. Repression of the long noncoding RNA-LET by histone deacetylase 3 contributes to hypoxia-mediated metastasis. Mol Cell. 2013;49:1083-1096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 402]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
88.  Chen CL, Tseng YW, Wu JC, Chen GY, Lin KC, Hwang SM, Hu YC. Suppression of hepatocellular carcinoma by baculovirus-mediated expression of long non-coding RNA PTENP1 and MicroRNA regulation. Biomaterials. 2015;44:71-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 173]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
89.  Zheng H, Yang S, Yang Y, Yuan SX, Wu FQ, Wang LL, Yan HL, Sun SH, Zhou WP. Epigenetically silenced long noncoding-SRHC promotes proliferation of hepatocellular carcinoma. J Cancer Res Clin Oncol. 2015;141:1195-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
90.  Yu W, Qiao Y, Tang X, Ma L, Wang Y, Zhang X, Weng W, Pan Q, Yu Y, Sun F. Tumor suppressor long non-coding RNA, MT1DP is negatively regulated by YAP and Runx2 to inhibit FoxA1 in liver cancer cells. Cell Signal. 2014;26:2961-2968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 84]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
91.  Cui M, Zheng M, Sun B, Wang Y, Ye L, Zhang X. A long noncoding RNA perturbs the circadian rhythm of hepatoma cells to facilitate hepatocarcinogenesis. Neoplasia. 2015;17:79-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 77]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
92.  Liu Y, Pan S, Liu L, Zhai X, Liu J, Wen J, Zhang Y, Chen J, Shen H, Hu Z. A genetic variant in long non-coding RNA HULC contributes to risk of HBV-related hepatocellular carcinoma in a Chinese population. PLoS One. 2012;7:e35145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 139]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
93.  Wen J, Liu Y, Liu J, Liu L, Song C, Han J, Zhu L, Wang C, Chen J, Zhai X. Expression quantitative trait loci in long non-coding RNA ZNRD1-AS1 influence both HBV infection and hepatocellular carcinoma development. Mol Carcinog. 2015;54:1275-1282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
94.  Stranger BE, Nica AC, Forrest MS, Dimas A, Bird CP, Beazley C, Ingle CE, Dunning M, Flicek P, Koller D. Population genomics of human gene expression. Nat Genet. 2007;39:1217-1224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 894]  [Cited by in F6Publishing: 946]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
95.  Veyrieras JB, Kudaravalli S, Kim SY, Dermitzakis ET, Gilad Y, Stephens M, Pritchard JK. High-resolution mapping of expression-QTLs yields insight into human gene regulation. PLoS Genet. 2008;4:e1000214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 453]  [Cited by in F6Publishing: 471]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
96.  Guo W, Zhao YP, Jiang YG, Wang RW, Hong L, Fan DM. ZNRD1 might mediate UV irradiation related DNA damage and repair in human esophageal cancer cells by regulation of ERCC1. Dis Esophagus. 2008;21:730-736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
97.  Hong L, Han Y, Li S, Yang J, Gong T, Li J, Zheng J, Zhang H, Zhao Q, Wu K. Role of ZNRD1 (zinc ribbon domain-containing 1) in angiogenesis of leukaemia cells. Cell Biol Int. 2011;35:321-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
98.  Hong L, Han Y, Shi R, Shao X, Sun L, Zhang Y, Huang D, Chen Z, Zhang G, Liang J. ZNRD1 gene suppresses cell proliferation through cell cycle arrest in G1 phase. Cancer Biol Ther. 2005;4:60-64.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Zhuo H, Tang J, Lin Z, Jiang R, Zhang X, Ji J, Wang P, Sun B. The aberrant expression of MEG3 regulated by UHRF1 predicts the prognosis of hepatocellular carcinoma. Mol Carcinog. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 106]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
100.  Lv J, Ma L, Chen XL, Huang XH, Wang Q. Downregulation of LncRNAH19 and MiR-675 promotes migration and invasion of human hepatocellular carcinoma cells through AKT/GSK-3β/Cdc25A signaling pathway. J Huazhong Univ Sci Technolog Med Sci. 2014;34:363-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
101.  Vernucci M, Cerrato F, Besnard N, Casola S, Pedone PV, Bruni CB, Riccio A. The H19 endodermal enhancer is required for Igf2 activation and tumor formation in experimental liver carcinogenesis. Oncogene. 2000;19:6376-6385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
102.  Wang J, Wang H, Zhang Y, Zhen N, Zhang L, Qiao Y, Weng W, Liu X, Ma L, Xiao W. Mutual inhibition between YAP and SRSF1 maintains long non-coding RNA, Malat1-induced tumourigenesis in liver cancer. Cell Signal. 2014;26:1048-1059.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell. 2010;39:925-938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1521]  [Cited by in F6Publishing: 1636]  [Article Influence: 116.9]  [Reference Citation Analysis (0)]
104.  Bernard D, Prasanth KV, Tripathi V, Colasse S, Nakamura T, Xuan Z, Zhang MQ, Sedel F, Jourdren L, Coulpier F. A long nuclear-retained non-coding RNA regulates synaptogenesis by modulating gene expression. EMBO J. 2010;29:3082-3093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 523]  [Cited by in F6Publishing: 559]  [Article Influence: 39.9]  [Reference Citation Analysis (0)]
105.  Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129:1311-1323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3230]  [Cited by in F6Publishing: 3249]  [Article Influence: 191.1]  [Reference Citation Analysis (0)]
106.  Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E, Chang HY. Long noncoding RNA as modular scaffold of histone modification complexes. Science. 2010;329:689-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2434]  [Cited by in F6Publishing: 2542]  [Article Influence: 181.6]  [Reference Citation Analysis (0)]
107.  Lujambio A, Akkari L, Simon J, Grace D, Tschaharganeh DF, Bolden JE, Zhao Z, Thapar V, Joyce JA, Krizhanovsky V. Non-cell-autonomous tumor suppression by p53. Cell. 2013;153:449-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 490]  [Cited by in F6Publishing: 539]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
108.  Mellert H, Espinosa JM. Tumor suppression by p53: is apoptosis important or not? Cell Rep. 2013;3:1335-1336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
109.  Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D, Khalil AM, Zuk O, Amit I, Rabani M. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 2010;142:409-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1559]  [Cited by in F6Publishing: 1640]  [Article Influence: 117.1]  [Reference Citation Analysis (0)]
110.  Yang JJ, Lee YJ, Hung HH, Tseng WP, Tu CC, Lee H, Wu WJ. ZAK inhibits human lung cancer cell growth via ERK and JNK activation in an AP-1-dependent manner. Cancer Sci. 2010;101:1374-1381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
111.  Yang X, Xie X, Xiao YF, Xie R, Hu CJ, Tang B, Li BS, Yang SM. The emergence of long non-coding RNAs in the tumorigenesis of hepatocellular carcinoma. Cancer Lett. 2015;360:119-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 127]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
112.  Amicone L, Citarella F, Cicchini C. Epigenetic regulation in hepatocellular carcinoma requires long noncoding RNAs. Biomed Res Int. 2015;2015:473942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
113.  Sun J, Bie B, Zhang S, Yang J, Li Z. Long non-coding RNAs: critical players in hepatocellular carcinoma. Int J Mol Sci. 2014;15:20434-20448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
114.  Tang J, Jiang R, Deng L, Zhang X, Wang K, Sun B. Circulation long non-coding RNAs act as biomarkers for predicting tumorigenesis and metastasis in hepatocellular carcinoma. Oncotarget. 2015;6:4505-4515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 113]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
115.  Li CH, Chen Y. Targeting long non-coding RNAs in cancers: progress and prospects. Int J Biochem Cell Biol. 2013;45:1895-1910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 344]  [Article Influence: 31.3]  [Reference Citation Analysis (0)]