Skip to main content
Non-coding RNA Research logoLink to Non-coding RNA Research
. 2017 Dec 5;2(3-4):129–136. doi: 10.1016/j.ncrna.2017.11.001

Long noncoding RNAs in the initiation, progression, and metastasis of hepatocellular carcinoma

Johanna K DiStefano 1
PMCID: PMC6084840  PMID: 30159431

Abstract

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Despite awareness of risk factors for the development of HCC and advances in the diagnosis and clinical management of the disease, the molecular mechanisms underlying hepatocarcinogenesis remain poorly understood. Recent experimental studies provide strong evidence that long noncoding RNAs (lncRNAs), non-protein-coding transcripts with lengths >200 basepairs, contribute to the pathogenesis of numerous human diseases. Over the past decade, a role for lncRNAs in the initiation, progression, and metastasis of HCC has likewise emerged and developed into a highly active area of research. Although many lncRNAs appear to be dysregulated in HCC, extensive functional characterization has been performed on only a small proportion of these candidates to date. This review summarizes select lncRNAs that have been shown to wield functional relevance in the initiation, progression, or metastasis of HCC, focusing on the specific mechanisms by which lncRNA effects might be linked to clinical manifestations of the disease. In addition, an overview of circulating lncRNAs that have been identified as potential biomarkers for the diagnosis and prognosis of HCC is provided.

Keywords: Long noncoding RNAs, lncRNAs, Noncoding RNA, Hepatocellular carcinoma, HULC, HOTAIR

1. Introduction

Hepatocellular carcinoma is the most common form of liver cancer, and in the United States, the disease has an annual incidence of at least 6 per 100,000 [1] and represents the fastest rising cause of cancer-related death [2]. Despite recent improvements in diagnostic methods and surgical techniques, the five-year survival rate for advanced HCC remains dismal [3]. Risk factors for the initiation of HCC include viral infection, nonalcoholic fatty liver disease, alcohol overconsumption, aflatoxin, and genetic factors; the majority of these conditions also contribute to the development of hepatic cirrhosis, which promotes HCC formation [2]. In fact, HCC is the most common cause of mortality in cirrhotic patients [4]. Although the risk factors for HCC are well characterized, the molecular mechanisms underlying the malignant transformation of hepatocytes are still incompletely understood [5].

There are approximately 60,000 noncoding RNAs in the human genome, 68% of which are long noncoding RNAs or lncRNAs [6]. Excellent reviews of the origin, biogenesis, and characteristics of lncRNAs, including the manner in which they are distinguished from other ncRNAs, can be found elsewhere [7], [8], [9], [10], and will therefore not be reiterated here. Because lncRNAs are typically expressed at much lower levels than protein-coding transcripts (i.e., messenger RNAs), these transcripts were considered to be transcriptional noise when first discovered in the early 1990s [11], [12], [13]. However, unprecedented advances in both high-throughput sequencing technologies and computational methods have fostered the identification and annotation of a number of biologically relevant lncRNAs. At the same time, a substantial amount of experimental evidence supporting a role for lncRNAs in biological processes underlying disease pathophysiology is accumulating at a rapid pace in the scientific literature.

Over the past decade, a role for lncRNAs in the initiation, progression, and metastasis of HCC has emerged and developed into a highly active area of research. Although many lncRNAs appear to be dysregulated in HCC [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], only a small proportion of these have so far been extensively characterized. This review summarizes select lncRNAs that have been shown to wield functional relevance in the initiation, progression, or metastasis of HCC, focusing on the specific mechanisms by which lncRNA effects might be linked to clinical manifestations of the disease. In addition, an overview of circulating lncRNAs that have been identified as potential biomarkers for the diagnosis and prognosis of HCC is provided.

2. lncRNAs associated with initiation, progression, and metastasis of HCC

A number of studies have investigated lncRNA signatures associated with HCC. Microarray profiling analysis of resected tumor tissues and matched adjacent non-tumor tissues from 29 HCC patients (22 HBV-positive and 3 HCV-positive) revealed 659 differentially expressed lncRNAs [24]. Of these, five lncRNAs were randomly selected for validation using quantitative reverse-transcription PCR (qRT-PCR), four of which were found to be significantly downregulated in tumor tissue: TCONS_00018278, AK093543, D16366, and ENST00000501583. An independent microarray analysis performed using three pairs (two HBV-positive and one HCV-positive) of HCC and adjacent non-tumor tissues reported differential expression of 214 lncRNAs [25]. Fifty of these lncRNAs were subsequently selected for validation with qRT-PCR in three sets of HCC tissues, of which expression patterns of 40 lncRNAs matched the microarray results. Dysregulated expression of eight lncRNAs (BC017743, ENST00000395084, NR_026591, NR_015378, NR_024284, NR_027151, AK056988, and uc003yqb.1) was confirmed in an expanded sample of nineteen matched HCC-non-tumor pairs. Both studies were limited by small sample sizes and differential distribution of patients infected with HBV or HCV, which may explain, in part, the lack of overlap between findings. A third study analyzed expression data from 372 HCC patients from The Cancer Genome Atlas (TCGA) and the NCBI GEO omnibus, and identified 177 cancer-specific lncRNAs, 41 of which were associated with gender, race, tumor grade, tumor stage, and TNM staging [26]. Using the univariate Cox proportional hazards regression model, CECR7, LINC00346, MAPKAPK5-AS1, LOC3338651, FLJ90757, and LOC283663 were found to be associated with overall survival. In addition to the identification of novel lncRNA candidates using a well-powered sample, this study also provided the first evidence of crosstalk among lncRNAs, miRNAs, and mRNAs in the progression of HCC, suggesting that further investigation of relationships between different coding and noncoding transcripts is warranted.

In addition to global lncRNA profiling, a number of studies have examined expression levels of individual lncRNAs. Among these findings, dysregulated expression of ANRIL [27], GAS5 [28] and PCAT-1 [29] has been associated with poor prognosis of HCC. An overview of lncRNAs dysregulated in HCC tumor tissue can be found elsewhere [30], and a comprehensive list of lncRNAs associated with HCC initiation, progression, and metastasis is shown in Table 1. Despite the accumulating evidence supporting dysregulated lncRNA expression in HCC, very few lncRNAs have thus far been functionally characterized with respect to the molecular underpinnings of disease pathogenesis. However, two lncRNAs, HULC and HOTAIR, have been relatively well studied and an overview of the roles played by these candidates in biological processes relevant to HCC is presented below.

Table 1.

lncRNAs associated with HCC initiation, progression, and metastasis in humans.

lncRNA Biological association Ref
AFAP-AS1 Promotes HCC cell proliferation and metastasis [73], [74]
AOC4P Modulates cell proliferation, migration and invasion via EMT inhibition [75]
ANRIL Regulates HCC cell proliferation and apoptosis [76]
CARLo-5 Promotes HCC cell proliferation, invasion, and metastasis [77]
CCAT1 Promotes HCC cell proliferation and invasion [78], [79], [80]
CCHE1 Promotes HCC progression [15]
DANCR Promotes HCC cell proliferation and metastasis; increases stemness features of HCC cells [81], [82]
DBH-AS1 Promotes HCC cell proliferation [83]
FTX Inhibits HCC cell growth and metastasis [84]
GAS5 Promotes HCC cell proliferation and invasion through regulation of vimentin [85], [86]
GPC3-AS1 Promotes HCC cell proliferation and migration. Activates GPC3 to promote HCC progression [87]
H19 Enhances tumorigenicity [88]
HEIH Facilitates tumor growth through enhancer of zeste homolog 2 [89]
HNF1A-AS1 Promotes autophagy and oncogenesis in HCC [90]
HOTAIR Promotes HCC cell migration and invasion and activates autophagy [19], [44], [45], [46], [47], [49]
HOTTIP Promotes HCC cell proliferation and tumorigenicity [91], [92]
HOXA-AS2 Promotes HCC cell proliferation and invasion, regulates apoptosis, and causes tumorigenicity in nude mice [93]
HULC Promotes HCC cell proliferation, triggers autophagy, and enhances tumor angiogenesis [37], [39], [94], [95]
Linc00152 Promotes HCC cell proliferation through activation of mTOR signaling pathway [96]
Linc01225 Promotes HCC cell proliferation and invasion [97]
Linc00974 Promotes HCC cell proliferation and invasion [98]
Linc-cdh4-2 Inhibits migration and invasion of HCC cells via R-cadherin pathway [99]
lincRNA-p21 Inhibits HCC invasion and metastasis [14], [100]
LOC90784 Promotes HCC cell proliferation and invasion [101]
lncRNA-ATB Promotes invasion-metastasis cascade in HCC [102]
lncCAMTA1 Promotes HCC cell proliferation, cancer stem cell-like properties, and tumorigenesis. [103]
lncTCF7 Promotes HCC aggressiveness through EMT activation, promotes cancer stem cell self-renewal and tumor propagation [104], [105]
MALAT1 Promotes tumor progression [106]
NEAT1 Promotes HCC cell proliferation [107]
PANDAR Promotes tumorigenesis in HCC [108]
PCAT1 Promotes HCC cell proliferation and migration; inhibits apoptosis [109]
PlncRNA-1 Promotes HCC ell proliferation, migration, and invasion via EMT signaling [110]
PVT1 Promotes HCC cell proliferation and cancer stem cell-like properties [111]
SNHG1 Promotes tumorigenesis in HCC [112], [113]
SNHG-003 Promotes HCC cell proliferation [114]
Sox2ot Promotes HCC metastasis [115]
SPRY4-IT1 Promotes tumor cell proliferation and invasion [116]
TP73-AS1 Modulates HCC cell proliferation [117]
TUG1 Promotes HCC cell growth and apoptosis through KLF2 sillencing [118]
TUSC7 Suppressed EMT in HCC [119]
Uc001kfo Promotes proliferation, metastasis and EMT in HCC cells by targeting alpha-SMA [120]
Uc-134 Promotes HCC cell proliferation, invasion, and metastasis [121]
Uc-338 Promotes HCC cell proliferation and induces cell cycle progression [122]
UCA1 Promotes HCC cell growth, tumorigenesis, and HCC progression [123], [124]
UFC1 Promotes HCC cell proliferation and cell-cycle progression and inhibits apoptosis [125]
Unigene56159 Promotes cell migration/invasion and EMT in HCC [126]
URHC Regulates HCC cell proliferation and apoptosis via ERK/MAPK signaling pathway [127]
XIST Inhibits HCC cell proliferation and metastasis by targeting miR-92b [128]
ZEB-AS1 Promotes tumor growth and metastasis [129], [130]
ZFAS1 Promotes metastasis in HCC [131]
ZNFX1-AS1 Modulates HCC cell proliferation [17]

2.1. HULC (highly upregulated in liver cancer)

HULC was first identified as a result of a genome-wide search for novel transcripts associated with the molecular pathogenesis of HCC [31]. HCC-specific cDNA libraries and tissue samples from 46 HCCs were analyzed, and one transcript was found to be significantly upregulated (33-fold) in 76% of tumors compared to a non-neoplastic pool of liver samples. Expression levels of this transcript were low in normal tissue and not significantly increased in other neoplastic tissues, suggesting that upregulation of this transcript was specific to HCC; thus this previously unannotated transcript was given the name “highly upregulated in liver cancer”. In a study of 38 patients with HCC, HULC levels were also associated with clinical stage, intrahepatic metastases, HCC recurrence, and postoperative survival [32]. HULC knockdown in hepatoma cell lines resulted in dysregulated expression of many genes, including several with an established role in liver cancer; however, no significant sequence homology was observed between HULC and these potential target genes, indicating that HULC-mediated effects are probably exerted via direct RNA-RNA interactions.

The HULC transcript is a 482 bp, spliced, polyadenylated ncRNA that localizes to the cytoplasm and co-purifies with ribosomes of carcinoma cells [31]. HULC shows evolutionary conservation in primates, although neither the mouse nor rat genome appears to have a HULC homologue [31]. A cAMP response element-binding (CREB) site in the HULC proximal promoter region was shown to be important for transcriptional activity in liver cancer [33]. In addition, HULC RNA can sequester miRNAs to regulate gene expression. For example, HULC was found to bind miR-372, leading to reduced expression of its target gene, protein kinase cAMP-activated catalytic subunit beta (PRKACB) [33].

Chronic infection with the hepatitis B virus (HBV) is associated with HCC development [34]. Levels of HBV X protein (HBx), a protein produced by HBV, are elevated in liver cells from HCC patients [35] and the HBx protein activates genes associated with cellular growth [36]. In an analysis of 33 HCC specimens, expression levels of HBx and HULC were positively correlated [37]. Knockdown of HBx expression corresponded with decreased HULC levels in hepatoma cells, while HBx overexpression resulted in a dose-dependent increase in HULC. Together, these results suggest that HBx regulates HULC expression in liver cells. Further, HULC downregulated expression of p18, a tumor suppressor gene located in close proximity to HULC, leading to proliferation of hepatoma cells. In hepatoma cells, HBx was found to downregulate p18, which was reversed with HULC knockdown, implicating a mechanism in which HBx-mediated upregulation of HULC promotes the proliferation of hepatoma cells through downregulation of p18 [37].

Depletion of insulin growth factor 2 mRNA-binding protein 1 (IGF2BP1) corresponded with increased half-life and steady state transcript levels of HULC [38]. CNOT1 (C-C motif chemokine receptor 4–NOT transcription complex subunit 1), a major component of the cytoplasmic RNA decay machinery, was identified as a novel interaction partner for IGF2BP1, and depletion of CNOT1 also resulted in increased half-life and expression of HULC. Thus, the main finding of this study was the characterization of IGF2BP1 as an adaptor protein capable of recruiting the CNOT1 complex and initiating the degradation of HULC [38].

Additional studies have served to shed light on the role of HULC in HCC development. For example, HULC was found to promote tumor angiogenesis by upregulating sphingosine kinase 1 (SPHK1) [39], an enzyme that generates sphingosine phosphate, which is known to contribute to cell survival, proliferation, differentiation, and angiogenesis [40], [41], [42]. Through a series of experiments, HULC was shown to increase expression of the E2F1 transcription factor, which binds to the SPHK1 promoter. HULC was further found to sequester miR-107, a known regulator of E2F1, resulting in a cascade of upregulated E2F1, increased SPHK1 activation, and eventually, tumor angiogenesis. These findings provided novel insight into molecular mechanisms underlying tumor angiogenesis in HCC.

HULC was also recently shown to enhance epithelial-mesenchymal transition (EMT), which contributes to tumor metastasis and recurrence in HCC via a signaling pathway involving zinc finger E-box binding homeobox 1 (ZEB1) and miR-200a-3p [32]. HULC was found to sequester miR-200a-3p, leading to increased levels of ZEB1, which corresponded to stabilized EMT. Together, these results reveal a potentially novel mechanism by which HULC mediates pathophysiology in HCC.

2.2. HOTAIR (Homeobox [HOX] transcript antisense RNA)

Although HOTAIR was initially identified under the auspices of an ultra-high resolution tiling microarray designed to interrogate the transcriptional and epigenetic landscape of HOX loci in primary human fibroblasts [43], its association with HCC was first reported in a study of 110 HCC samples using qRT-PCR [44]. In that study, HOTAIR was expressed at significantly higher levels in HCC tumor compared to adjacent non-cancerous tissue and was an independent prognostic factor for predicting HCC recurrence in liver transplantation patients. In patients who exceeded the criteria for liver transplantation (single HCC ≤ 5 cm or up to 3 HCCs ≤ 3 cm), high HOTAIR levels were associated with a shorter recurrence-free survival. Functional knockdown of HOTAIR expression in liver cancer cells corresponded with decreased cell viability and invasion, increased TNF-α induced apoptosis, and heightened sensitivity to chemotherapeutic agents [44]. A second study published in the same year reported upregulation of HOTAIR in surgically resected tumor tissue from 63 HCC patients compared with adjacent non-tumor tissues [45]. Patients with high levels of HOTAIR expression showed an elevated risk of post-hepatectomy recurrence and lymph node metastasis. Similar to the previous study [44], HOTAIR knockdown corresponded with decreased HCC cell proliferation, and was also found to be associated with reduced levels of matrix metalloproteinase-9 (MMP9) and vascular endothelial growth factor protein (VEGF), both of which contribute to cell motility and metastasis. These results were replicated in additional studies, providing strong support for HOTAIR in biological mechanisms underlying HCC progression [45], [46], [47].

Expression profiling of coding transcripts following HOTAIR knockdown in HCC cells resulted in the dysregulation of 296 genes, including upregulation of QKI, KH domain containing RNA binding (QKI), CD82, and RNA binding motif protein 38 (RBM38); increased transcript and protein levels of these three genes were further validated by qRT-PCR analysis and western blotting, respectively [46]. However, only levels of RBM38, but not QKI and CD82, were dysregulated in HCC samples compared to adjacent non-tumor paired samples, leading the research team to focus on this gene as being functionally relevant to HOTAIR. Concordant with this finding, knockdown of HOTAIR expression in HepG2 and Bel-7402 cells resulted in increased transcript and protein levels of RBM38, and corresponded with reduced HCC cell migration and invasion, which was specifically rescued by RBM38 downregulation. These findings showed that HOTAIR promotes HCC cell migration and invasion through inhibition of RBM38. A similar investigation of HOTAIR depletion showed downregulated expression of Wnt and β-catenin [48], providing evidence that HOTAIR may affect HCC progression through multiple signaling pathways.

In an investigation of HOTAIR in HCC cell models and a xenograft mouse model, HOTAIR was shown to negatively regulate miR-218 expression in HCC through a promoter regulatory axis involving EZH2-targeting-miR-218-2 [49]. HOTAIR knockdown in vitro was found to inhibit HCC cell viability and induce G1-phase arrest, while in vivo depletion of HOTAIR was shown to suppress tumorigenicity by disinhibiting miR-218 expression. The Bmi-1 oncogene was identified as a functional target of miR-218, and the main downstream targets signaling, P16 (Ink4a) and P14 (ARF), were activated in HOTAIR-suppressed tumorigenesis. In primary human HCC specimens, HOTAIR and Bmi-1 were concordantly upregulated whereas miR-218 was downregulated in these tissues. Furthermore, HOTAIR was inversely associated with miR-218 expression and positively correlated with Bmi-1 expression in these clinical tissues.

Additional studies aimed at understanding the molecular mechanisms by which HOTAIR impacts tumor cell development have been recently undertaken. For example, in one study, overexpression of HOTAIR was found to promote activation of autophagy in HCC cell lines, while depletion of the lncRNA suppressed this pathway [19]. In this model, upregulated HOTAIR corresponded with increased expression autophagy-related 3 (ATG3) and ATG7 expression in HCC cells, suggesting a novel pathway promoting HCC cell proliferation. In another study, RNA-protein complexes formed with HOTAIR were regulated by the RNA helicase DEAD box protein 5 (DDX5), and associated with HBV biosynthesis and poor prognosis in HBV-mediated liver cancer [50]. Specifically, DDX5 was shown to interact with suppressor of zeste 12 homolog (SUZ12), the core subunit of the chromatin-modifying polycomb repressive complex 2 (PRC2), and in association with HOTAIR, repressed the transcription of epithelial cell adhesion molecule (EpCAM) and three pluripotency genes.

In an investigation of HOTAIR, forkhead box C1 (FOXC1), and miR-1, levels of HOTAIR and FOXC1 were increased, while levels of miR-1 were decreased in HCC tissues and HepG2 cells compared to normal liver cells and adjacent non-tumor tissues [51]. Overexpression of HOTAIR in the immunodeficient nude mouse model (nu/nu) resulted in enhanced HCC cell proliferation and progression of tumor xenografts. Functional characterization studies showed that FOXC1 binds to an upstream region of HOTAIR and activates its expression in HCC cells, while HOTAIR negatively regulated miR-1 expression. Results from this work suggested that HOTAIR is a FOXC1-activated driver of malignancy, which acts in part through the repression of miR-1.

Most recently, Zhou et al. [52] demonstrated that HOTAIR knockdown corresponded with reduced cell proliferation and increased G0/G1 cell cycle arrest in Huh7 cells. HOTAIR knockdown was also associated with reduced levels of cyclin D1 (CCND1) transcript and protein, as well as phosphorylated signal transducer and activator of transcription 3 (STAT3), which led to an even greater decrease in CCND1 expression. These results provided evidence that HOTAIR may impact HCC cell proliferation through regulation of cell cycle, STAT3 activity, and CCND1 expression.

Since its annotation in 2007, HOTAIR has emerged as a novel prognostic marker for HCC. While a number of studies have indicated multiple pathways by which HOTAIR may affect HCC cell proliferation and invasion, further investigation of the molecular mechanisms underlying dysregulated HOTAIR expression and the manner in which the lncRNA promotes HCC progression are necessary to nominate its use as a potential therapeutic target in the treatment of HCC.

3. lncRNAs as biomarkers for HCC diagnosis and prognosis

A number of different treatment options are available for HCC patients, including surgical resection, liver transplantation, ablation (radiofrequency, cryoablation, and percutaneous ethanol injection), transcatheter arterial chemoembolization, radioembolization, and chemotherapy with the multi-kinase inhibitor sorafenib [53]. Selection of therapeutic intervention depends on many factors such as tumor stage, the ability of the patient to tolerate surgery, hepatic function, and availability of organs, in the case of liver transplantation; however, in all cases, early diagnosis is key for optimal survival rates. Due to an absence of early-stage symptoms, HCC often remains undiagnosed until advanced stages of disease, which limits opportunities for successful intervention, leading to poor prognosis and reduced five-year survival rate [54]. It is widely recognized that improved screening methods are critical for early diagnosis of HCC and enhanced survival rates for HCC patients.

A variety of imaging techniques are available for HCC screening, each presenting with unique advantages and disadvantages [55]. For example, computed tomography (CT) is an easily accessible imaging modality useful for clinical routine work-up of HCC [55]. However, while CT and magnetic resonance imaging (MRI) both yield acceptable ranges of sensitivity (55%–91%) and specificity (77%–96%), the relatively high cost and the potential harm arising from radiation exposure limit their use for large-scale screening and routine surveillance [56]. On the other hand, liver ultrasound, which represents the primary surveillance modality for HCC, has a sensitivity of ∼60% and a specificity of 85–90% [57], but is less accurate in obese patients or in the presence of a nodular liver [58]. In addition, ultrasound is associated with decreased detection compared to CT and MRI, albeit with a lower false positive rate [59].

In addition to imaging screening modalities, a number of serum biomarkers are available for the detection of HCC [60]. These include alpha-fetoprotein (AFP), gamma-glutamyl transferase (GGT), des-γ-carboxyprothrombin (DCP), glypican-3 (GPC3), osteopontin, and others, all of which having varying levels of sensitivity and specificity [54], [61]. Of these, AFP is the most widely used serum biomarker for the diagnosis and monitoring of HCC; however, the specificity of this glycoprotein is limited. For example, normal levels of AFP are present in ∼30% of HCC patients at the time of diagnosis and remain low even with advanced stages of disease [62]. Likewise, the range of AFP values in HCC is quite wide, from normal concentrations to >100,000 ng/mL [62], and non-cancer liver diseases such as chronic hepatitis or cirrhosis are associated with increased AFP levels [63]. AFP >400–500 ng/ml is considered diagnostic for HCC, although fewer than half of patients present with values in that range [62].

To be clinically useful, biomarkers should demonstrate high sensitivity and specificity for HCC, have concentrations that reflect changes in tumor progression and prognosis, and be easily and non-invasively detectable in body fluids. LncRNAs have emerged as diagnostic biomarkers for human diseases, particularly cancer [64], [65] and cardiovascular disease [66], due to their stability in biofluids. In HCC, hepatic expression levels of many lncRNAs have been found to be dysregulated in tumor samples relative to normal tissue; however, the number of lncRNAs showing differences in circulating concentrations is relatively small [30]. A summary of differentially expressed, circulating lncRNAs is shown in Table 2, and a detailed discussion of several potential biomarkers, including HULC (highly upregulated in liver cancer), MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), and UCA1 (urothelial-associated carcinoma 1), is presented below.

Table 2.

lncRNAs implicated as circulating biomarkers for the diagnosis and prognosis of HCC.

lncRNA lncRNA name Major findings Ref
DANCR Differentiation
antagonizing non-protein coding RNA
High plasma levels differentiated HCC patients from healthy volunteers and individuals with non-cancerous liver disease; high plasma levels associated with microvascular invasion and liver capsule invasion in HCC patients [81]
DGCR5 DiGeorge syndrome critical region gene 5 Low serum levels associated with poor cancer-specific survival and were independent negative prognostic factor for HCC [132]
HEIH High Expression in HCC Plasma levels increased in HCC patients compared to healthy individuals [30]
HULC Highly upregulated in liver cancer Plasma levels increased in HCC patients compared to healthy individuals [67], [68]
LINC00152 Plasma levels increased in HCC patients compared to healthy individuals [68]
LINC01225 Plasma levels increased in HCC patients compared to healthy individuals [97]
MALAT1 Metastasis-associated lung adenocarcinoma transcript 1 Plasma levels increased in HCC patients compared to healthy individuals [70]
PVT1 & uc022mbe.2 Plasmacytoma variant translocation 1 Combined lncRNA signature could distinguish HCC patients from healthy individuals and was associated with associated with tumor size, Barcelona Clinic Liver Cancer stage, and serum bilirubin [20]
RP11-160H22 &XLOC_014172 & LOC149086 Plasma levels of three-lncRNA signatures discriminated HCC patients from healthy individuals; XLOC_014172, LOC149086, showed a higher expression in HCC patients with metastasis [133]
SPRY4-IT1 SPRY4 intronic transcript 1 Plasma levels could distinguish HCC patients from healthy controls [134]
uc001ncr & AX800134 Able to accurately diagnose HBV-positive HCC and early HCC [135]
uc003wbd & AF085935 Serum levels higher in HCC and HBV patients compared with normal controls [136]
UCA1 Urothelial carcinoma-associated 1 Higher expression in HCC, associated with higher tumor grade, large tumor size, positive vascular invasion, and advanced TNM stage [71], [72]
WRAP53 WD repeat containing, antisense to TP5 Higher serum levels in HCC and significant independent prognostic marker in relapse-free survival [71]

3.1. HULC

In the initial work identifying HULC as a ncRNA upregulated in HCC, a pilot study comprised of four HCC patients, ten patients with hepatic cirrhosis, and nine healthy individuals found that peripheral blood levels of HULC RNA were substantially higher in patients with HCC compared to individuals with no evidence of liver disease, and that for two of the HCC patients, blood levels of HULC mirrored those in surgically resected tumors [31]. These findings were subsequently corroborated in a larger study of 30 HCC patients and 20 healthy individuals, in which HULC was detected in plasma samples of 63% of the HCC patients [67]. In these patients, HULC detection frequencies increased in accord with Edmondson and Steiner grade, an established method of histological classification. HULC was also detected more frequently in HCC patients with HBV versus those without HBV (90% vs. 25%), although the implications of this finding are not yet clear. In an investigation of eight lncRNA candidates in 24 HCC patients and an equal number of healthy individuals, HULC levels were significantly higher in HCC patients and were correlated with tumor size and tumor capsular [68]. Receiver operating characteristic (ROC) curve analysis for HULC was 0.78, which increased when combined with AFP concentration [68]. Together, these findings demonstrated that circulating HULC levels are elevated in HCC patients, are reflective of neoplasm expression levels, and are associated with tumor aggressiveness and progression. While the prognostic power of HULC requires further substantiation by longitudinal analysis in prospective studies, these reports provide a significant step toward establishing the utility of HULC expression as a prognostic indicator for HCC.

3.2. MALAT1

In an analysis of gene expression from HCC, hepatoblastomas, tissue adjacent to HCC tumors, and normal tissue, MALAT1 showed significant upregulation in neoplastic samples [69]. This finding laid the groundwork for a subsequent study in which plasma levels of MALAT1 were assessed in 88 HCC patients, 51 healthy controls, and 28 individuals with non-cancerous liver disease [70]. Circulating levels of MALAT1 were elevated in HCC samples (9.17 ± 43.62) compared to individuals with hepatic disease (1.10 ± 0.82) and healthy controls (0.85 ± 1.10). Interestingly, plasma MALAT1 levels in HCC patients with HBV infection were lower than those with HCV, and in those HCC patients with liver damage or cirrhosis, MALAT1 levels were substantially higher. ROC analysis between HCC patients and individuals with hepatic disease revealed a cut-off value of 1.60 and an AUC of 0.66. Sensitivity and specificity for the detection of HCC were highest (88.6% and 75%, respectively) using a combination of MALAT1, AFP, and DCP levels, compared to results using individual measures. Although these findings require additional validation, they provide preliminary evidence supporting clinical utility of plasma MALAT1 levels for predicting development of HCC and as a marker of liver damage. However, given the association of this lncRNA in many different kinds of cancer, the specificity of MALAT1 as a biomarker for HCC will need to be addressed.

3.3. UCA1

Kamel et al. [71] used in silico pathway enrichment analysis to identify urothelial carcinoma associated 1 (UCA1) as a potential candidate for carcinogenesis. Serum levels of UCA1 were significantly higher in HCC patients (N = 82) compared to chronic HCV patients (N = 34) and healthy individuals (N = 44) and were associated with age, smoking, HCV antibodies-positive patients, and Child-Pugh score. Serum UCA1 levels were also strongly correlated with expression levels in paired HCC tissue (r = 0.823; P < 0.01), suggesting that UCA1 may not only represent a biomarker for HCC, but may also play a role in carcinogenesis. In comparisons of HCC patients and healthy controls, the threshold of UCA1 was 1.04, with a sensitivity of 92.7%, indicating that the lncRNA could accurately distinguish HCC patients from individuals without cancer. In comparisons of HCC patients with chronic HCV patients, a value of 1.5 could discriminate the two groups, but with a reduced sensitivity and specificity. The area under the ROC was 0.861 for discriminating HCC patients from healthy controls, and 0.728 for discriminating HCC patients from chronic HCV patients. In an independent investigation comprised of 105 HCC patients, 105 individuals with benign liver disease (BLD), and 105 healthy volunteers, serum UCA1 levels were significantly higher in HCC patients compared with BLD patients or healthy controls [72]. ROC curve analysis demonstrated that serum UCA1 levels could distinguish HCC patients from healthy controls (AUC = 0.902) and at a cut-off threshold of 1.85; the sensitivity and specificity were 73.3% and 99.0%, respectively. Similar findings were obtained for comparisons of HCC and BLD patients. In HCC patients, elevated serum UCA1 levels were associated with high tumor grade, large tumor size, positive vascular invasion, and advanced TNM stage. In addition, the 5-year overall survival in HCC patients with high serum UCA1 levels was significantly worse than those with low serum levels. While these findings suggest that UCA1 may be a useful prognostic marker for HCC, other studies have found elevated plasma levels of this lncRNA in other kinds of cancer, suggesting that it may not be specific for HCC. However, given the evidence supporting association between serum UCA1 levels and clinicopathological features of HCC, investigation of this lncRNA in prospective studies with large sample sizes appears to be warranted.

4. Conclusions

The discovery of dysregulated lncRNAs has added a new layer of complexity to the molecular architecture of human disease. However, there are still many gaps in our current understanding of lncRNA function, and further study of these molecules is expected to yield deeper insights into mechanisms underlying the pathogenesis of many human diseases, development of new RNA-based targets for the prevention and treatment of disease, and improved methods for early detection of pathology.

Numerous studies have demonstrated a role for lncRNAs in multiple biological processes relevant to HCC, including the initiation, progression, metastasis, recurrence, treatment, and prognosis of the disease. Additional work has shown dysregulation of lncRNAs in tumor tissues is associated with these biological processes. Finally, a rapidly growing set of circulating lncRNAs is showing diagnostic and prognostic promise in HCC research. While the role of lncRNAs in HCC development and progression is still not completely understood, elucidation of the molecular mechanisms by which these noncoding transcripts affect disease pathogenesis will be critical for identifying potential targets for therapeutic development. Further investigation is expected to unravel the regulatory networks and molecular characteristics of lncRNAs, leading to improvements in the diagnosis, treatment, and possible prevention of HCC.

References

  • 1.El-Serag H.B., Kanwal F. Epidemiology of hepatocellular carcinoma in the United States: where are we? Where do we go? Hepatology. 2014;60:1767–1775. doi: 10.1002/hep.27222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Mittal S., El-Serag H.B. Epidemiology of hepatocellular carcinoma consider the population. J Clin Gastroenterol. 2013;47(Suppl):S2–S6. doi: 10.1097/MCG.0b013e3182872f29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ilikhan S.U., Bilici M., Sahin H. Assessment of the correlation between serum prolidase and alpha-fetoprotein levels in patients with hepatocellular carcinoma. World J. Gastroenterol. 2015;21:6999–7007. doi: 10.3748/wjg.v21.i22.6999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Forner A., Llovet J.M., Bruix J. Hepatocellular carcinoma. Lancet. 2012;379:1245–1255. doi: 10.1016/S0140-6736(11)61347-0. [DOI] [PubMed] [Google Scholar]
  • 5.Cicinnati V.R., Sotiropoulos G.C., Beckebaum S. Established and emerging therapies for hepatocellular carcinoma. Minerva Med. 2010;101:405–418. [PubMed] [Google Scholar]
  • 6.Iyer M.K., Niknafs Y.S., Malik R. The landscape of long noncoding RNAs in the human transcriptome. Nat. Genet. 2015;47:199–208. doi: 10.1038/ng.3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Leti F., DiStefano J.K. Long noncoding RNAs as diagnostic and therapeutic targets in type 2 diabetes and related complications. Genes (Basel) 2017:8. doi: 10.3390/genes8080207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Li L.J., Leng R.X., Fan Y.G., Pan H.F., Ye D.Q. Translation of noncoding RNAs: focus on lncRNAs, pri-miRNAs, and circRNAs. Exp. Cell Res. 2017;361:1–8. doi: 10.1016/j.yexcr.2017.10.010. [DOI] [PubMed] [Google Scholar]
  • 9.Marchese F.P., Raimondi I., Huarte M. The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 2017;18:206. doi: 10.1186/s13059-017-1348-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sun W., Yang Y., Xu C., Guo J. Regulatory mechanisms of long noncoding RNAs on gene expression in cancers. Cancer Genet. 2017;216-217:105–110. doi: 10.1016/j.cancergen.2017.06.003. [DOI] [PubMed] [Google Scholar]
  • 11.Borsani G., Tonlorenzi R., Simmler M.C. Characterization of a murine gene expressed from the inactive X chromosome. Nature. 1991;351:325–329. doi: 10.1038/351325a0. [DOI] [PubMed] [Google Scholar]
  • 12.Brannan C.I., Dees E.C., Ingram R.S., Tilghman S.M. The product of the H19 gene may function as an RNA. Mol. Cell Biol. 1990;10:28–36. doi: 10.1128/mcb.10.1.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brockdorff N., Ashworth A., Kay G.F. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell. 1992;71:515–526. doi: 10.1016/0092-8674(92)90519-i. [DOI] [PubMed] [Google Scholar]
  • 14.Jia M., Jiang L., Wang Y.D., Huang J.Z., Yu M., Xue H.Z. lincRNA-p21 inhibits invasion and metastasis of hepatocellular carcinoma through Notch signaling-induced epithelial-mesenchymal transition. Hepatol. Res. 2016 Oct;46(11):1137–1144. doi: 10.1111/hepr.12659. [DOI] [PubMed] [Google Scholar]
  • 15.Peng W., 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. 2016;83:450–455. doi: 10.1016/j.biopha.2016.06.056. [DOI] [PubMed] [Google Scholar]
  • 16.Sui C.J., Zhou Y.M., Shen W.F. Long noncoding RNA GIHCG promotes hepatocellular carcinoma progression through epigenetically regulating miR-200b/a/429. J. Mol. Med. Berl. 2016 Nov;94(11):1281–1296. doi: 10.1007/s00109-016-1442-z. [DOI] [PubMed] [Google Scholar]
  • 17.Wang T., Ma S., Qi X. Long noncoding RNA ZNFX1-AS1 suppresses growth of hepatocellular carcinoma cells by regulating the methylation of miR-9. Onco Targets Ther. 2016;9:5005–5014. doi: 10.2147/OTT.S103329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Xiong D., Sheng Y., Ding S. LINC00052 regulates the expression of NTRK3 by miR-128 and miR-485-3p to strengthen HCC cells invasion and migration. Oncotarget. 2016 Jul 26;7(30):47593–47608. doi: 10.18632/oncotarget.10250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yang L., Zhang X., Li H., Liu J. The long noncoding RNA HOTAIR activates autophagy by upregulating ATG3 and ATG7 in hepatocellular carcinoma. Mol. Biosyst. 2016;12:2605–2612. doi: 10.1039/c6mb00114a. [DOI] [PubMed] [Google Scholar]
  • 20.Yu J., Han J., Zhang J. The long noncoding RNAs PVT1 and uc002mbe.2 in sera provide a new supplementary method for hepatocellular carcinoma diagnosis. Med. Baltim. 2016;95 doi: 10.1097/MD.0000000000004436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yuan P., Cao W., Zang Q., Li G., Guo X., Fan J. The HIF-2alpha-MALAT1-miR-216b axis regulates multi-drug resistance of hepatocellular carcinoma cells via modulating autophagy. Biochem. Biophys. Res. Commun. 2016 Sep 23;478(3):1067–1073. doi: 10.1016/j.bbrc.2016.08.065. [DOI] [PubMed] [Google Scholar]
  • 22.Zhou N., Si Z., Li T., Chen G., Zhang Z., Qi H. Long non-coding RNA CCAT2 functions as an oncogene in hepatocellular carcinoma, regulating cellular proliferation, migration and apoptosis. Oncol. Lett. 2016;12:132–138. doi: 10.3892/ol.2016.4580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zhu X.T., Yuan J.H., Zhu T.T., Li Y.Y., Cheng X.Y. Long noncoding RNA GPC3-AS1 promotes hepatocellular carcinoma progression via epigenetically activating GPC3. FEBS J. 2016 Jul;12(1):132–138. doi: 10.1111/febs.13839. [DOI] [PubMed] [Google Scholar]
  • 24.Liu W.T., Lu X., Tang G.H. LncRNAs expression signatures of hepatocellular carcinoma revealed by microarray. World J. Gastroenterol. 2014;20:6314–6321. doi: 10.3748/wjg.v20.i20.6314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhu J., Liu S., Ye F. The long noncoding RNA expression profile of hepatocellular carcinoma identified by microarray analysis. PLoS One. 2014;9 doi: 10.1371/journal.pone.0101707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zhang J., Fan D., Jian Z., Chen G.G., Lai P.B. Cancer specific long noncoding RNAs show differential expression patterns and competing endogenous RNA potential in hepatocellular carcinoma. PLoS One. 2015;10 doi: 10.1371/journal.pone.0141042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hua L., Wang C.Y., Yao K.H., Chen J.T., Zhang J.J., Ma W.L. High expression of long non-coding RNA ANRIL is associated with poor prognosis in hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2015;8:3076–3082. [PMC free article] [PubMed] [Google Scholar]
  • 28.Tu Z.Q., Li R.J., Mei J.Z., Li X.H. Down-regulation of long non-coding RNA GAS5 is associated with the prognosis of hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2014;7:4303–4309. [PMC free article] [PubMed] [Google Scholar]
  • 29.Yan T.H., Yang H., Jiang J.H. Prognostic significance of long non-coding RNA PCAT-1 expression in human hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2015;8:4126–4131. [PMC free article] [PubMed] [Google Scholar]
  • 30.Bao H., Su H. Long noncoding RNAs act as novel biomarkers for hepatocellular carcinoma: progress and prospects. Biomed. Res. Int. 2017;2017:6049480. doi: 10.1155/2017/6049480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Panzitt K., Tschernatsch M.M., Guelly C. Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology. 2007;132:330–342. doi: 10.1053/j.gastro.2006.08.026. [DOI] [PubMed] [Google Scholar]
  • 32.Li S.P., Xu H.X., Yu Y. LncRNA HULC enhances epithelial-mesenchymal transition to promote tumorigenesis and metastasis of hepatocellular carcinoma via the miR-200a-3p/ZEB1 signaling pathway. Oncotarget. 2016 Jul 5;7(27):42431–42446. doi: 10.18632/oncotarget.9883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wang J., Liu X., Wu H. CREB up-regulates long non-coding RNA, HULC expression through interaction with microRNA-372 in liver cancer. Nucleic Acids Res. 2010;38:5366–5383. doi: 10.1093/nar/gkq285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Di Bisceglie A.M. Hepatitis B and hepatocellular carcinoma. Hepatology. 2009;49:S56–S60. doi: 10.1002/hep.22962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kim C.M., Koike K., Saito I., Miyamura T., Jay G. HBx gene of hepatitis B virus induces liver cancer in transgenic mice. Nature. 1991;351:317–320. doi: 10.1038/351317a0. [DOI] [PubMed] [Google Scholar]
  • 36.Muroyama R., Kato N., Yoshida H. Nucleotide change of codon 38 in the X gene of hepatitis B virus genotype C is associated with an increased risk of hepatocellular carcinoma. J. Hepatol. 2006;45:805–812. doi: 10.1016/j.jhep.2006.07.025. [DOI] [PubMed] [Google Scholar]
  • 37.Du Y., Kong G., You 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. doi: 10.1074/jbc.M112.342113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hammerle M., Gutschner T., Uckelmann H. Posttranscriptional destabilization of the liver-specific long noncoding RNA HULC by the IGF2 mRNA-binding protein 1 (IGF2BP1) Hepatology. 2013;58:1703–1712. doi: 10.1002/hep.26537. [DOI] [PubMed] [Google Scholar]
  • 39.Lu Z., Xiao Z., Liu F. Long non-coding RNA HULC promotes tumor angiogenesis in liver cancer by up-regulating sphingosine kinase 1 (SPHK1) Oncotarget. 2016;7:241–254. doi: 10.18632/oncotarget.6280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Alvarez S.E., Harikumar K.B., Hait N.C. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature. 2010;465:1084–1088. doi: 10.1038/nature09128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Liu Y., Deng J., Wang L. S1PR1 is an effective target to block STAT3 signaling in activated B cell-like diffuse large B-cell lymphoma. Blood. 2012;120:1458–1465. doi: 10.1182/blood-2011-12-399030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nagahashi M., Ramachandran S., Kim E.Y. Sphingosine-1-phosphate produced by sphingosine kinase 1 promotes breast cancer progression by stimulating angiogenesis and lymphangiogenesis. Cancer Res. 2012;72:726–735. doi: 10.1158/0008-5472.CAN-11-2167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rinn J.L., Kertesz M., Wang J.K. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129:1311–1323. doi: 10.1016/j.cell.2007.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Yang Z., Zhou L., Wu L.M. Overexpression of long non-coding RNA HOTAIR predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation. Ann. Surg. Oncol. 2011;18:1243–1250. doi: 10.1245/s10434-011-1581-y. [DOI] [PubMed] [Google Scholar]
  • 45.Geng Y.J., Xie S.L., Li Q., Ma J., Wang G.Y. Large intervening non-coding RNA HOTAIR is associated with hepatocellular carcinoma progression. J. Int. Med. Res. 2011;39:2119–2128. doi: 10.1177/147323001103900608. [DOI] [PubMed] [Google Scholar]
  • 46.Ding C., Cheng S., Yang Z. 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. doi: 10.3390/ijms15034060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ishibashi M., Kogo R., Shibata K. Clinical significance of the expression of long non-coding RNA HOTAIR in primary hepatocellular carcinoma. Oncol. Rep. 2013;29:946–950. doi: 10.3892/or.2012.2219. [DOI] [PubMed] [Google Scholar]
  • 48.Gao J.Z., Li J., Du J.L., Li X.L. Long non-coding RNA HOTAIR is a marker for hepatocellular carcinoma progression and tumor recurrence. Oncol. Lett. 2016;11:1791–1798. doi: 10.3892/ol.2016.4130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Fu W.M., Zhu X., Wang W.M. Hotair mediates hepatocarcinogenesis through suppressing miRNA-218 expression and activating P14 and P16 signaling. J. Hepatol. 2015;63:886–895. doi: 10.1016/j.jhep.2015.05.016. [DOI] [PubMed] [Google Scholar]
  • 50.Zhang H., Xing Z., Mani S.K. RNA helicase DEAD box protein 5 regulates Polycomb repressive complex 2/Hox transcript antisense intergenic RNA function in hepatitis B virus infection and hepatocarcinogenesis. Hepatology. 2016;64:1033–1048. doi: 10.1002/hep.28698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Su D.N., Wu S.P., Chen H.T., He J.H. HOTAIR, a long non-coding RNA driver of malignancy whose expression is activated by FOXC1, negatively regulates miRNA-1 in hepatocellular carcinoma. Oncol. Lett. 2016;12:4061–4067. doi: 10.3892/ol.2016.5127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhou J.J., Cheng D., He X.Y., Meng Z., Li W.Z., Chen R.F. Knockdown of Hotair suppresses proliferation and cell cycle progression in hepatocellular carcinoma cell by downregulating CCND1 expression. Mol. Med. Rep. 2017;16:4980–4986. doi: 10.3892/mmr.2017.7162. [DOI] [PubMed] [Google Scholar]
  • 53.Raza A., Sood G.K. Hepatocellular carcinoma review: current treatment, and evidence-based medicine. World J. Gastroenterol. 2014;20:4115–4127. doi: 10.3748/wjg.v20.i15.4115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tsuchiya N., Sawada Y., Endo I., Saito K., Uemura Y., Nakatsura T. Biomarkers for the early diagnosis of hepatocellular carcinoma. World J. Gastroenterol. 2015;21:10573–10583. doi: 10.3748/wjg.v21.i37.10573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Schraml C., Kaufmann S., Rempp H. Imaging of HCC-current state of the art. Diagn. (Basel) 2015;5:513–545. doi: 10.3390/diagnostics5040513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hao Q.Q., Chen G.Y., Zhang J.H., Sheng J.H., Gao Y. Diagnostic value of long noncoding RNAs for hepatocellular carcinoma: a PRISMA-compliant meta-analysis. Med. Baltim. 2017;96 doi: 10.1097/MD.0000000000007496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Singal A., Volk M.L., Waljee A. Meta-analysis: surveillance with ultrasound for early-stage hepatocellular carcinoma in patients with cirrhosis. Aliment. Pharmacol. Ther. 2009;30:37–47. doi: 10.1111/j.1365-2036.2009.04014.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Tong M.J., Blatt L.M., Kao V.W. Surveillance for hepatocellular carcinoma in patients with chronic viral hepatitis in the United States of America. J. Gastroenterol. Hepatol. 2001;16:553–559. doi: 10.1046/j.1440-1746.2001.02470.x. [DOI] [PubMed] [Google Scholar]
  • 59.Kobayashi K., Sugimoto T., Makino H. Screening methods for early detection of hepatocellular carcinoma. Hepatology. 1985;5:1100–1105. doi: 10.1002/hep.1840050607. [DOI] [PubMed] [Google Scholar]
  • 60.El-Serag H.B., Davila J.A. Surveillance for hepatocellular carcinoma: in whom and how? Ther. Adv. Gastroenterol. 2011;4:5–10. doi: 10.1177/1756283X10385964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zheng C., Liu X., Chen L., Xu Z., Shao J. lncRNAs as prognostic molecular biomarkers in hepatocellular carcinoma: a systematic review and meta-analysis. Oncotarget. 2017;8:59638–59647. doi: 10.18632/oncotarget.19559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bialecki E.S., Di Bisceglie A.M. Diagnosis of hepatocellular carcinoma. HPB Oxf. 2005;7:26–34. doi: 10.1080/13651820410024049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Johnson P.J. The role of serum alpha-fetoprotein estimation in the diagnosis and management of hepatocellular carcinoma. Clin. Liver Dis. 2001;5:145–159. doi: 10.1016/s1089-3261(05)70158-6. [DOI] [PubMed] [Google Scholar]
  • 64.Huang Y.K., Yu J.C. Circulating microRNAs and long non-coding RNAs in gastric cancer diagnosis: an update and review. World J. Gastroenterol. 2015;21:9863–9886. doi: 10.3748/wjg.v21.i34.9863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kishikawa T., Otsuka M., Ohno M., Yoshikawa T., Takata A., Koike K. Circulating RNAs as new biomarkers for detecting pancreatic cancer. World J. Gastroenterol. 2015;21:8527–8540. doi: 10.3748/wjg.v21.i28.8527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Shi Q., Yang X. Circulating MicroRNA and long noncoding RNA as biomarkers of cardiovascular diseases. J. Cell Physiol. 2016;231:751–755. doi: 10.1002/jcp.25174. [DOI] [PubMed] [Google Scholar]
  • 67.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. doi: 10.1155/2013/136106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Li J., Wang X., Tang J. HULC and Linc00152 act as novel biomarkers in predicting diagnosis of hepatocellular carcinoma. Cell Physiol. Biochem. 2015;37:687–696. doi: 10.1159/000430387. [DOI] [PubMed] [Google Scholar]
  • 69.Luo J.H., Ren B., Keryanov S. Transcriptomic and genomic analysis of human hepatocellular carcinomas and hepatoblastomas. Hepatology. 2006;44:1012–1024. doi: 10.1002/hep.21328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Konishi H., Ichikawa D., Yamamoto Y. Plasma level of metastasis-associated lung adenocarcinoma transcript 1 is associated with liver damage and predicts development of hepatocellular carcinoma. Cancer Sci. 2016;107:149–154. doi: 10.1111/cas.12854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kamel M.M., Matboli M., Sallam M., Montasser I.F., Saad A.S., El-Tawdi A.H.F. Investigation of long noncoding RNAs expression profile as potential serum biomarkers in patients with hepatocellular carcinoma. Transl. Res. 2016;168:134–145. doi: 10.1016/j.trsl.2015.10.002. [DOI] [PubMed] [Google Scholar]
  • 72.Zheng Z.K., Pang C., Yang Y., Duan Q., Zhang J., Liu W.C. Serum long noncoding RNA urothelial carcinoma-associated 1: a novel biomarker for diagnosis and prognosis of hepatocellular carcinoma. J. Int. Med. Res. 2017 doi: 10.1177/0300060517726441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lu X., Zhou C., Li R. Critical role for the long non-coding RNA AFAP1-AS1 in the proliferation and metastasis of hepatocellular carcinoma. Tumour Biol. 2016;37:9699–9707. doi: 10.1007/s13277-016-4858-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zhang J.Y., Weng M.Z., Song F.B. Long noncoding RNA AFAP1-AS1 indicates a poor prognosis of hepatocellular carcinoma and promotes cell proliferation and invasion via upregulation of the RhoA/Rac2 signaling. Int. J. Oncol. 2016;48:1590–1598. doi: 10.3892/ijo.2016.3385. [DOI] [PubMed] [Google Scholar]
  • 75.Wang T.H., Lin Y.S., Chen Y. Long non-coding RNA AOC4P suppresses hepatocellular carcinoma metastasis by enhancing vimentin degradation and inhibiting epithelial-mesenchymal transition. Oncotarget. 2015;6:23342–23357. doi: 10.18632/oncotarget.4344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Huang M.D., Chen W.M., Qi F.Z. Long non-coding RNA ANRIL is upregulated in hepatocellular carcinoma and regulates cell apoptosis by epigenetic silencing of KLF2. J. Hematol. Oncol. 2015;8:50. doi: 10.1186/s13045-015-0146-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wang F., Xie C., Zhao W., Deng Z., Yang H., Fang Q. Long non-coding RNA CARLo-5 expression is associated with disease progression and predicts outcome in hepatocellular carcinoma patients. Clin. Exp. Med. 2017;17:33–43. doi: 10.1007/s10238-015-0395-9. [DOI] [PubMed] [Google Scholar]
  • 78.Deng L., Yang S.B., Xu F.F., Zhang J.H. Long noncoding RNA CCAT1 promotes hepatocellular carcinoma progression by functioning as let-7 sponge. J. Exp. Clin. Cancer Res. 2015;34:18. doi: 10.1186/s13046-015-0136-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhu H., Zhou X., Chang H. CCAT1 promotes hepatocellular carcinoma cell proliferation and invasion. Int. J. Clin. Exp. Pathol. 2015;8:5427–5434. [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhu H.Q., Zhou X., Chang H. Aberrant expression of CCAT1 regulated by c-Myc predicts the prognosis of hepatocellular carcinoma. Asian Pac J. Cancer Prev. 2015;16:5181–5185. doi: 10.7314/apjcp.2015.16.13.5181. [DOI] [PubMed] [Google Scholar]
  • 81.Ma X., Wang X., Yang C. DANCR acts as a diagnostic biomarker and promotes tumor growth and metastasis in hepatocellular carcinoma. Anticancer Res. 2016;36:6389–6398. doi: 10.21873/anticanres.11236. [DOI] [PubMed] [Google Scholar]
  • 82.Yuan S.X., Wang J., Yang F. Long noncoding RNA DANCR increases stemness features of hepatocellular carcinoma by derepression of CTNNB1. Hepatology. 2016;63:499–511. doi: 10.1002/hep.27893. [DOI] [PubMed] [Google Scholar]
  • 83.Huang J.L., Ren T.Y., Cao S.W. HBx-related long non-coding RNA DBH-AS1 promotes cell proliferation and survival by activating MAPK signaling in hepatocellular carcinoma. Oncotarget. 2015;6:33791–33804. doi: 10.18632/oncotarget.5667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Liu F., Yuan J.H., Huang J.F. Long noncoding RNA FTX inhibits hepatocellular carcinoma proliferation and metastasis by binding MCM2 and miR-374a. Oncogene. 2016;35:5422–5434. doi: 10.1038/onc.2016.80. [DOI] [PubMed] [Google Scholar]
  • 85.Chang L., Li C., Lan T. Decreased expression of long non-coding RNA GAS5 indicates a poor prognosis and promotes cell proliferation and invasion in hepatocellular carcinoma by regulating vimentin. Mol. Med. Rep. 2016;13:1541–1550. doi: 10.3892/mmr.2015.4716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hu L., Ye H., Huang G. Long noncoding RNA GAS5 suppresses the migration and invasion of hepatocellular carcinoma cells via miR-21. Tumour Biol. 2016;37:2691–2702. doi: 10.1007/s13277-015-4111-x. [DOI] [PubMed] [Google Scholar]
  • 87.Zhu X.T., Yuan J.H., Zhu T.T., Li Y.Y., Cheng X.Y. Long noncoding RNA glypican 3 (GPC3) antisense transcript 1 promotes hepatocellular carcinoma progression via epigenetically activating GPC3. FEBS J. 2016;283:3739–3754. doi: 10.1111/febs.13839. [DOI] [PubMed] [Google Scholar]
  • 88.Matouk I.J., DeGroot N., Mezan S. The H19 non-coding RNA is essential for human tumor growth. PLoS One. 2007;2 doi: 10.1371/journal.pone.0000845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Yang F., Zhang L., Huo X.S. Long noncoding RNA high expression in hepatocellular carcinoma facilitates tumor growth through enhancer of zeste homolog 2 in humans. Hepatology. 2011;54:1679–1689. doi: 10.1002/hep.24563. [DOI] [PubMed] [Google Scholar]
  • 90.Liu Z., Wei X., Zhang A., Li C., Bai J., Dong J. Long non-coding RNA HNF1A-AS1 functioned as an oncogene and autophagy promoter in hepatocellular carcinoma through sponging hsa-miR-30b-5p. Biochem. Biophys. Res. Commun. 2016;473:1268–1275. doi: 10.1016/j.bbrc.2016.04.054. [DOI] [PubMed] [Google Scholar]
  • 91.Ge Y., Yan X., Jin Y. MiRNA-192 [corrected] and miRNA-204 directly suppress lncRNA HOTTIP and interrupt GLS1-mediated glutaminolysis in hepatocellular carcinoma. PLoS Genet. 2015;11 doi: 10.1371/journal.pgen.1005726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Tsang F.H., Au S.L., Wei L. 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. doi: 10.1111/liv.12746. [DOI] [PubMed] [Google Scholar]
  • 93.Wang F., Yang H., Deng Z., Su Y., Fang Q., Yin Z. HOX antisense lincRNA HOXA-AS2 promotes tumorigenesis of hepatocellular carcinoma. Cell Physiol. Biochem. 2016;40:287–296. doi: 10.1159/000452545. [DOI] [PubMed] [Google Scholar]
  • 94.Xiong H., Li B., He J., Zeng Y., Zhang Y., He F. lncRNA HULC promotes the growth of hepatocellular carcinoma cells via stabilizing COX-2 protein. Biochem. Biophys. Res. Commun. 2017;490:693–699. doi: 10.1016/j.bbrc.2017.06.103. [DOI] [PubMed] [Google Scholar]
  • 95.Xiong H., Ni Z., He J. LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. Oncogene. 2017;36:3528–3540. doi: 10.1038/onc.2016.521. [DOI] [PubMed] [Google Scholar]
  • 96.Ji J., Tang J., Deng L. LINC00152 promotes proliferation in hepatocellular carcinoma by targeting EpCAM via the mTOR signaling pathway. Oncotarget. 2015;6:42813–42824. doi: 10.18632/oncotarget.5970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wang X., Zhang W., Tang J. LINC01225 promotes occurrence and metastasis of hepatocellular carcinoma in an epidermal growth factor receptor-dependent pathway. Cell Death Dis. 2016;7 doi: 10.1038/cddis.2016.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Tang J., Zhuo H., Zhang X. A novel biomarker Linc00974 interacting with KRT19 promotes proliferation and metastasis in hepatocellular carcinoma. Cell Death Dis. 2014;5 doi: 10.1038/cddis.2014.518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gao Y., Wang G., Zhang C. Long non-coding RNA linc-cdh4-2 inhibits the migration and invasion of HCC cells by targeting R-cadherin pathway. Biochem. Biophys. Res. Commun. 2016;480:348–354. doi: 10.1016/j.bbrc.2016.10.048. [DOI] [PubMed] [Google Scholar]
  • 100.Ding G., Peng Z., Shang J., Kang Y., Ning H., Mao C. LincRNA-p21 inhibits invasion and metastasis of hepatocellular carcinoma through miR-9/E-cadherin cascade signaling pathway molecular mechanism. Onco Targets Ther. 2017;10:3241–3247. doi: 10.2147/OTT.S134910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Xu J.H., Chang W.H., Fu H.W., Shu W.Q., Yuan T., Chen P. Upregulated long non-coding RNA LOC90784 promotes cell proliferation and invasion and is associated with poor clinical features in HCC. Biochem. Biophys. Res. Commun. 2017;490:920–926. doi: 10.1016/j.bbrc.2017.06.141. [DOI] [PubMed] [Google Scholar]
  • 102.Yuan J.H., Yang F., Wang F. A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 2014;25:666–681. doi: 10.1016/j.ccr.2014.03.010. [DOI] [PubMed] [Google Scholar]
  • 103.Ding L.J., Li Y., Wang S.D. Long noncoding RNA lncCAMTA1 promotes proliferation and cancer stem cell-like properties of liver cancer by inhibiting CAMTA1. Int. J. Mol. Sci. 2016:17. doi: 10.3390/ijms17101617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang Y., He L., Du Y. The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell. 2015;16:413–425. doi: 10.1016/j.stem.2015.03.003. [DOI] [PubMed] [Google Scholar]
  • 105.Wu J., Zhang J., Shen B. Long noncoding RNA lncTCF7, induced by IL-6/STAT3 transactivation, promotes hepatocellular carcinoma aggressiveness through epithelial-mesenchymal transition. J. Exp. Clin. Cancer Res. 2015;34:116. doi: 10.1186/s13046-015-0229-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lai M.C., Yang Z., Zhou L. Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med. Oncol. 2012;29:1810–1816. doi: 10.1007/s12032-011-0004-z. [DOI] [PubMed] [Google Scholar]
  • 107.Fang L., Sun J., Pan Z. Long non-coding RNA NEAT1 promotes hepatocellular carcinoma cell proliferation through the regulation of miR-129-5p-VCP-IkappaB. Am. J. Physiol. Gastrointest. Liver Physiol. 2017;313:G150–G156. doi: 10.1152/ajpgi.00426.2016. [DOI] [PubMed] [Google Scholar]
  • 108.Peng W., Fan H. Long non-coding RNA PANDAR correlates with poor prognosis and promotes tumorigenesis in hepatocellular carcinoma. Biomed. Pharmacother. 2015;72:113–118. doi: 10.1016/j.biopha.2015.04.014. [DOI] [PubMed] [Google Scholar]
  • 109.Wen J., Xu J., Sun Q., Xing C., Yin W. Upregulation of long non coding RNA PCAT-1 contributes to cell proliferation, migration and apoptosis in hepatocellular carcinoma. Mol. Med. Rep. 2016;13:4481–4486. doi: 10.3892/mmr.2016.5075. [DOI] [PubMed] [Google Scholar]
  • 110.Dong L., Ni J., Hu W., Yu C., Li H. Upregulation of long non-coding RNA PlncRNA-1 promotes metastasis and induces epithelial-mesenchymal transition in hepatocellular carcinoma. Cell Physiol. Biochem. 2016;38:836–846. doi: 10.1159/000443038. [DOI] [PubMed] [Google Scholar]
  • 111.Wang F., Yuan J.H., Wang S.B. Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology. 2014;60:1278–1290. doi: 10.1002/hep.27239. [DOI] [PubMed] [Google Scholar]
  • 112.Zhang H., Zhou D., Ying M. Expression of long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) exacerbates hepatocellular carcinoma through suppressing miR-195. Med. Sci. Monit. 2016;22:4820–4829. doi: 10.12659/MSM.898574. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 113.Zhang M., Wang W., Li T. Long noncoding RNA SNHG1 predicts a poor prognosis and promotes hepatocellular carcinoma tumorigenesis. Biomed. Pharmacother. 2016;80:73–79. doi: 10.1016/j.biopha.2016.02.036. [DOI] [PubMed] [Google Scholar]
  • 114.Cao C., Zhang T., Zhang D. The long non-coding RNA, SNHG6-003, functions as a competing endogenous RNA to promote the progression of hepatocellular carcinoma. Oncogene. 2017;36:1112–1122. doi: 10.1038/onc.2016.278. [DOI] [PubMed] [Google Scholar]
  • 115.Shi X.M., Teng F. Up-regulation of long non-coding RNA Sox2ot promotes hepatocellular carcinoma cell metastasis and correlates with poor prognosis. Int. J. Clin. Exp. Pathol. 2015;8:4008–4014. [PMC free article] [PubMed] [Google Scholar]
  • 116.Zhou M., Zhang X.Y., Yu X. Overexpression of the long non-coding RNA SPRY4-IT1 promotes tumor cell proliferation and invasion by activating EZH2 in hepatocellular carcinoma. Biomed. Pharmacother. 2017;85:348–354. doi: 10.1016/j.biopha.2016.11.035. [DOI] [PubMed] [Google Scholar]
  • 117.Li S., Huang Y., Huang Y. The long non-coding RNA TP73-AS1 modulates HCC cell proliferation through miR-200a-dependent HMGB1/RAGE regulation. J. Exp. Clin. Cancer Res. 2017;36:51. doi: 10.1186/s13046-017-0519-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Huang M.D., Chen W.M., Qi F.Z. Long non-coding RNA TUG1 is up-regulated in hepatocellular carcinoma and promotes cell growth and apoptosis by epigenetically silencing of KLF2. Mol. Cancer. 2015;14:165. doi: 10.1186/s12943-015-0431-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Wang Y., Liu Z., Yao B. Long non-coding RNA TUSC7 acts a molecular sponge for miR-10a and suppresses EMT in hepatocellular carcinoma. Tumour Biol. 2016;37:11429–11441. doi: 10.1007/s13277-016-4892-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Pan Y., Qin T., Yin S., Zhang X., Gao X., Mu L. Long non-coding RNA UC001kfo promotes hepatocellular carcinoma proliferation and metastasis by targeting alpha-SMA. Biomed. Pharmacother. 2017;87:669–677. doi: 10.1016/j.biopha.2017.01.018. [DOI] [PubMed] [Google Scholar]
  • 121.Ni W., Zhang Y., Zhan Z. A novel lncRNA uc.134 represses hepatocellular carcinoma progression by inhibiting CUL4A-mediated ubiquitination of LATS1. J. Hematol. Oncol. 2017;10:91. doi: 10.1186/s13045-017-0449-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Bo C., Li N., Li X., Liang X., An Y. Long noncoding RNA uc.338 promotes cell proliferation through association with BMI1 in hepatocellular carcinoma. Hum. Cell. 2016;29:141–147. doi: 10.1007/s13577-016-0140-z. [DOI] [PubMed] [Google Scholar]
  • 123.Hu J.J., Song W., Zhang S.D. HBx-upregulated lncRNA UCA1 promotes cell growth and tumorigenesis by recruiting EZH2 and repressing p27Kip1/CDK2 signaling. Sci. Rep. 2016;6:23521. doi: 10.1038/srep23521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Xiao J.N., Yan T.H., Yu R.M. Long non-coding RNA UCA1 regulates the expression of Snail2 by miR-203 to promote hepatocellular carcinoma progression. J. Cancer Res. Clin. Oncol. 2017;143:981–990. doi: 10.1007/s00432-017-2370-1. [DOI] [PubMed] [Google Scholar]
  • 125.Cao C., Sun J., Zhang D. The long intergenic noncoding RNA UFC1, a target of MicroRNA 34a, interacts with the mRNA stabilizing protein HuR to increase levels of beta-catenin in HCC cells. Gastroenterology. 2015;148 doi: 10.1053/j.gastro.2014.10.012. 415–426. [DOI] [PubMed] [Google Scholar]
  • 126.Lv J., Fan H.X., Zhao X.P. Long non-coding RNA Unigene56159 promotes epithelial-mesenchymal transition by acting as a ceRNA of miR-140-5p in hepatocellular carcinoma cells. Cancer Lett. 2016;382:166–175. doi: 10.1016/j.canlet.2016.08.029. [DOI] [PubMed] [Google Scholar]
  • 127.Xu W.H., Zhang J.B., Dang Z. 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. doi: 10.7150/ijbs.8232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Zhuang L.K., Yang Y.T., Ma X. MicroRNA-92b promotes hepatocellular carcinoma progression by targeting Smad7 and is mediated by long non-coding RNA XIST. Cell Death Dis. 2016;7 doi: 10.1038/cddis.2016.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lan T., Chang L., Wu L., Yuan Y. Downregulation of ZEB2-AS1 decreased tumor growth and metastasis in hepatocellular carcinoma. Mol. Med. Rep. 2016;14:4606–4612. doi: 10.3892/mmr.2016.5836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Li T., Xie J., Shen C. Upregulation of long noncoding RNA ZEB1-AS1 promotes tumor metastasis and predicts poor prognosis in hepatocellular carcinoma. Oncogene. 2016;35:1575–1584. doi: 10.1038/onc.2015.223. [DOI] [PubMed] [Google Scholar]
  • 131.Li T., Xie J., Shen C. Amplification of long noncoding RNA ZFAS1 promotes metastasis in hepatocellular carcinoma. Cancer Res. 2015;75:3181–3191. doi: 10.1158/0008-5472.CAN-14-3721. [DOI] [PubMed] [Google Scholar]
  • 132.Huang R., Wang X., Zhang W. Down-regulation of LncRNA DGCR5 correlates with poor prognosis in hepatocellular carcinoma. Cell Physiol. Biochem. 2016;40:707–715. doi: 10.1159/000452582. [DOI] [PubMed] [Google Scholar]
  • 133.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. doi: 10.18632/oncotarget.2934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Jing W., Gao S., Zhu M. Potential diagnostic value of lncRNA SPRY4-IT1 in hepatocellular carcinoma. Oncol. Rep. 2016;36:1085–1092. doi: 10.3892/or.2016.4859. [DOI] [PubMed] [Google Scholar]
  • 135.Wang K., Guo W.X., Li N. Serum LncRNAs profiles serve as novel potential biomarkers for the diagnosis of HBV-positive hepatocellular carcinoma. PLoS One. 2015;10 doi: 10.1371/journal.pone.0144934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lu J., Xie F., Geng L., Shen W., Sui C., Yang J. Investigation of serum lncRNA-uc003wbd and lncRNA-AF085935 expression profile in patients with hepatocellular carcinoma and HBV. Tumour Biol. 2015;36:3231–3236. doi: 10.1007/s13277-014-2951-4. [DOI] [PubMed] [Google Scholar]

Articles from Non-coding RNA Research are provided here courtesy of KeAi Publishing

RESOURCES