Review Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Sep 15, 2018; 10(9): 260-270
Published online Sep 15, 2018. doi: 10.4251/wjgo.v10.i9.260
Emerging role of long non-coding RNA in the development of gastric cancer
Hang Yu, Long Rong, Department of Endoscopic Center, Peking University First Hospital, Beijing 100034, China
ORCID number: Hang Yu (0000-0001-7419-3579); Long Rong (0000-0002-3635-4682).
Author contributions: Yu H designed the study, searched the scientific literature for the latest developments in the field, wrote and edited the manuscript; Rong L revised the manuscript.
Conflict-of-interest statement: No potential conflicts of interest. No financial support.
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: Long Rong, MD, Chief Doctor, Professor, Department of Endoscopic Center, Peking University First Hospital, No.8 Xishiku Street, Beijing 100034, China. ronglong8@sina.com
Telephone: +86-10-83572437 Fax: +86-10-83572437
Received: March 28, 2018
Peer-review started: March 29, 2018
First decision: April 26, 2018
Revised: June 14, 2018
Accepted: June 26, 2018
Article in press: June 27, 2018
Published online: September 15, 2018

Abstract

Gastric cancer is a common, worldwide malignancy and has a poor prognosis due to late diagnosis. Long non-coding RNAs (lncRNAs) are a significant subtype of RNA molecules with a length longer than 200 nucleotides (nt) that rarely encode proteins. In recent decades, deregulation of lncRNAs has been shown to be involved in tumorigenesis and tumor progression in various human carcinomas, including gastric cancer. Accumulating evidence has shown that some lncRNAs may function as diagnostic biomarkers or therapeutic targets for gastric cancer. Thus, exploring the specific functions of lncRNAs will help both gain a better understanding of the pathogenesis and develop novel treatments for gastric cancer. In this review, we highlight the expression and functional roles of lncRNAs in gastric cancer, and analyze the potential applications of lncRNAs as diagnostic markers and therapeutic targets.

Key Words: Function, Tumorigenesis, Gastric cancer, Therapeutic target, Long non-coding RNAs, Diagnostic marker

Core tip: Gastric cancer is a common, worldwide malignancy that has a poor prognosis. The promising regulatory potential of long non-coding RNAs (lncRNAs) in tumorigenesis and cancer development, including gastric cancer, has been widely demonstrated. Thus, exploring the function of lncRNAs can help to both gain a better understanding of the pathogenesis and develop novel treatments for gastric cancer. In this review, we aim to elucidate the expression patterns and functional roles of lncRNAs in gastric cancer, and analyze the latent applications of lncRNAs as diagnostic markers and therapeutic targets.



INTRODUCTION

Gastric cancer is one of the most common malignancies of the digestive tract[1]. Furthermore, there has been consistent growth in the incidence and mortality rates of gastric cancer due to late diagnosis. The 5-year survival rate could reach 90%-97% if patients are diagnosed early and get prompt treatment, either by endoscopy or surgery. Nevertheless, the 5-year survival rate is currently under 20% for terminal cancer patients[2-8]. As a result, prompt diagnosis of gastric cancer would significantly improve prognosis. An exploration of the molecular mechanisms involved in the initiation and development of gastric cancer is needed to help discover credible markers, and further reduce mortality rate, decrease disability and improve prognosis.

In the past, most non-coding RNAs were considered “junk RNAs” of the transcriptome. Nevertheless, with the rapid evolution of whole-genome analysis of gene expression, it has been demonstrated that most of the genome is transcribed into RNAs that have no protein-coding functions[9,10]. Although non-coding RNAs do not encode proteins, they regulate gene expression through various mechanisms. Non-coding RNA-mediated gene silencing is an important part of epigenetic changes, which have been demonstrated to be involved in human carcinogenesis[11]. During the last decade, more attention has been paid to the functional significance of non-coding RNAs in oncogenesis and tumor progression[12]. Long non-coding RNAs (LncRNAs), defined as transcripts > 200 nt in length, are an important group of non-coding RNAs[13]. It has been revealed that in various carcinomas, lncRNAs are frequently deregulated, which may indicate their potential role in the initiation of cancers[14-16]. Thus, understanding the roles of lncRNAs will help elucidate the underlying biological events in different cancers, including gastric cancer, and ultimately lead to the development of novel diagnostic tools and targeted therapies. Furthermore, multiple lncRNAs have been shown to be related to diverse biological processes associated with gastric cancer, which enable lncRNAs to serve as diagnostic biomarkers and therapeutic targets. Here, we aim to review the recent progress made in elucidating the function of gastric cancer-related lncRNAs, and to also explore their potential capacity to serve as diagnostic or prognostic biomarkers.

Structure of lncRNAs

The length of transcript > 200 nts and the limited protein-coding potential are two of the main characteristics that distinguish lncRNAs from others[17]. Researchers first identified lncRNAs when trying to sequence full-length cDNAs in mice[18]. Thereafter, a 2.2 kilobase functional lncRNA termed “HOTAIR” was shown to be involved in multiple processes of epigenetic regulation[19]. In the past decade, with the development of transcriptomics, more lncRNAs have been recognized as important functional products of the genome[20]. The polyadenylation and transcription of lncRNAs are commonly performed by RNA polymerase (RNAP) II[21-23]. The length of lncRNAs typically varies from 1000 to 10000 nts, and some lncRNAs can reach 100000 nts[20]. To date, the sequence and molecular structure of many lncRNAs still need to be elucidated. For sequence elements, some lncRNAs may perfectly match Watson-Crick base-pairing in order to function properly, while others would utilize imperfect pairing, which is when Watson-Crick base pairs are interspersed with non-Watson-Crick pairs[17]. In a previous study, an analysis of 204 lncRNAs and their comparison to protein-coding transcripts showed that a paucity of introns, low GC content, and lack of start codons were some of the sequence traits of lncRNAs. Some of the biological features of lncRNAs, including positioning within the nucleus and low transcription levels, are generated from the sequence traits formerly mentioned[24]. The secondary elements and three-dimensional structures of RNA also play a vital role in their action mode, but structural studies of lncRNAs have not been performed.

Category of lncRNAs

Thus far, there has been no systematic classification of lncRNAs. In fact, lncRNA entries are a mixture of multiple functions and mechanisms, only a small proportion of which has been functionally annotated. Many lncRNAs cannot be classified into any particular category. As a result, it is difficult to classify lncRNAs based on only one principal.

Different classification methods are used according to different features of lncRNAs. For example, based on genomic location and context relative to protein-coding genes, lncRNAs can be divided into five broad categories: (1) Sense lncRNA is transcribed from the sense strand and contains several overlapping exons; (2) Antisense lncRNA, on the contrary, is transcribed from the antisense strand; (3) Bidirectional lncRNA is transcribed on one strand, while an adjacent protein-coding gene initiates expression on the same strand, simultaneously; (4) Intronic lncRNA is transcribed entirely from within the introns of protein-coding genes; and (5) Intergenic lncRNA is transcribed within genomic intervals of neighboring protein-coding genes[21]. According to their effects exerted on DNA sequences, lncRNAs can be classified into cis-lncRNAs (cis-acting lncRNAs) and trans-lncRNAs (trans-acting lncRNAs). The expression levels of adjacent genes can be regulated by cis-lncRNAs, while those of remote genes by trans-lncRNAs[25].

Recent advances in high-throughput transcriptome sequencing technologies have made it feasible to conduct deep mining of both the function and mechanism of more lncRNAs, which will eventually enable us to optimize the arbitrary classifications of lncRNAs.

MECHANISMS AND FUNCTION OF LNCRNAS

With the rapid development of experimental and computational technologies, more and more lncRNAs have been identified, among which only a small proportion has been functionally annotated. However, researchers have shown that the process of chromatin remodeling, transcription and post-transcriptional modification could be regulated by lncRNAs[20,26-28].

Chromatin remodeling

Chromatin remodeling was one of the first identified functions of lncRNAs. It has been elucidated that lncRNAs could alter the structure of chromatin and modulate the expression level of genes[29]. LncRNAs can confine chromatin remodeling complexes to particular regions in the genome, which is frequently achieved by interaction with polycomb repressive complex 2 (PRC2), so as to epigenetically regulate gene expression[20,26]. In association with PRC2, small interfering RNAs (siRNAs) have been shown to mediate deletion of specific lncRNAs and thus further alter their expression levels[30]. In addition to acting through PRC2, some lncRNAs directly recruit DNA methyltransferases or other complexes to modify chromatin conformation[31-33].

Transcriptional regulation

LncRNAs regulate transcription by interfering with the transcription of enhancers and promoters[34,35]. Some lncRNAs are transcribed within adjacent gene promoters. These lncRNAs can modulate the function of specific genes by interfering with the binding of protein factors. For example, the non-coding RNA SRG1 is transcribed across the promoter of the SER3 gene, and the expression of SRG1 can remarkably repress SER3. Mechanistically, the transcription of SRG1 in the promoter area disturbs the binding of activators[36]. LncRNAs can also be transcribed within distal enhancers and recruit transcription factors to these loci to regulate the expression level of neighboring genes[37]. Furthermore, lncRNAs can act by regulating RNAP II activity[38]. Some lncRNAs could regulate the transcription of key apoptotic genes, which is one of the vital pathways for carcinogenesis control[39]. For instance, the lncRNA INXS is transcribed from the intron of the BCL-X gene. Under the regulation of INXS, BCL-X can splice into BCL-XS, which is a pro-apoptosis isomer of BCL-X[40].

Post-transcriptional regulation

LncRNAs can recognize complementary sequences, and thus can regulate multiple procedures in the post-transcriptional modification of messenger RNAs (mRNAs). For instance, the complementarity of lncRNA Xist and Tsix can form complex dimers in vivo. These dimers are then spliced into small RNAs, which can balance the effect of X-chromosome inactivation through RNAi-mediated silencing[41]. Some lncRNAs could also act as competing endogenous RNAs (ceRNAs). Studies showed that these lncRNAs were able to bind miRNAs (sponging) and diminish the inhibitory effect on their natural targets[42]. LncRNA sponges are widely involved in cancer tumorigenesis. For example, in hepatocellular carcinoma, the lncRNA CCAT1 could act as a molecular sponge for let-7 and de-repress the function of its endogenous targets HMGA2 and c-Myc[43].

By exploring the function of lncRNAs in various aspects of cell transformation and metastasis, we will finally gain a better understanding of cancer biology. Nevertheless, many other functions of lncRNAs remain to be discovered.

ROLES OF LNCRNAS IN CANCER

Aberrant gene expression is the foundation of cancer pathogenesis. Intensive study of the genetic causes of cancer has found that variation in non-coding sequences is responsible for a large proportion of cancer susceptibility[44]. In fact, most single nucleotide polymorphisms (SNPs) associated with malignant tumors are found to be located in non-protein-coding loci. Recent studies have shown that many cancer risk loci are transcribed into non-coding RNAs, particularly lncRNAs, which play vital roles in the process of tumorigenesis and progression.

The underlying mechanisms of the regulatory function of lncRNAs in the progression of cancer remain largely unknown. Evidence to date shows that some lncRNAs can recruit protein factors to particular regions of the genome in order to epigenetically modify chromatin, while others can regulate the protein signaling pathways underlying carcinogenesis. LncRNAs can functionally control cellular growth, division and differentiation, thus making them the focus of current cancer research.

As mentioned above, lncRNAs are key regulators of cancer initiation and progression, suggesting they may have applications in diagnosis and therapeutics. Many lncRNAs are highly correlated with particular cancer states and are useful as diagnostic and prognostic markers. For instance, the lncRNA prostate cancer non-coding RNA 1 (PRNCR1) is upregulated in both prostate cancer and precursor lesion prostatic intraepithelial neoplasia. In addition, PRNCR1 expression levels are elevated in patient urine samples, thus making it a fine noninvasive indicator of prostate cancer[45].

Deregulations of lncRNAs in gastric caner

The above data showed that lncRNAs have strong correlations with cancer state, and their deregulation can lead to cancer initiation and progression. Many lncRNAs have also been shown to be involved in gastric cancer. Among lncRNAs associated with gastric cancer, some of them function as oncogenes and are upregulated during tumorigenesis, while others are downregulated and serve as tumor suppressors. In this section, we briefly review some of the well-studied lncRNAs involved in gastric cancer.

HOX transcript antisense RNA

Located on chromosome 12, the HOX transcript antisense RNA (HOTAIR) contains 6232 nts and encodes 2.2 kbs of long non-coding RNA. HOTAIR is a non-protein-coding RNA with significant regulatory potential via gene remodeling[46]. High levels of HOTAIR expression are associated with cancer cell proliferation, apoptosis, invasion, and progression in a variety of malignancies, making it a significant predictor of subsequent metastasis and death[47-50].

In gastric cancer tissues, HOTAIR expression levels are remarkably elevated, which suggests that HOTAIR functions as an oncogene in gastric cancer. Song et al[51] observed that HOTAIR was overexpressed in gastric cancer, and that by inhibiting miR-152, HOTAIR was responsible for the elevation of human leukocyte antigen G. Furthermore, Endo et al[52] elucidated that upregulation of HOTAIR was correlated with lymph node metastasis, invasion into vessels, and reduction of survival time in gastric cancer. Chen et al[53] also found that HOTAIR was significantly upregulated in gastric cancer tissues, and that its overexpression was associated with migration and invasion.

The mechanism of HOTAIR overexpression in gastric cancer is currently unknown. Previous studies have proposed several potential mechanisms of how deregulated HOTAIR functions in tumorigenesis. Epithelial-to-mesenchymal transition (EMT) is generally considered to be the foundation of metastasis. Liu et al[54] found that by suppressing HOTAIR, the EMT process could be reversed in gastric cancer cells. Other research showed that HOTAIR promoted gastric cell EMT and metastasis by inhibiting E-cadherin expression through its interaction with EZH2[53]. The functional SNP rs4759314 of HOTAIR had strong associations with gastric cancer susceptibility. SNP rs4759314, which resides in the promoter region of an intron, has been demonstrated to influence the expression of HOTAIR by interfering with this promoter[55].

H19

The LncRNA H19, discovered in 1991 by Bartolomei[56], was the first imprinted lncRNA gene identified. H19, residing on chromosome 11p15.5, is transcribed from the H19/IGF2 gene[57,58]. Similar to mRNA, the H19 gene contains five exons and is transcribed by RNA polymerase II. However, it does not contain a common open reading frame. In general, the high conservation in H19 structure is considered to be responsible for the universality of its functions[59]. Deregulation of H19 has been reported in various malignancies, such as breast cancer, bladder cancer and cervical carcinomas, which indicates its oncogenic role[60-64]. H19 has also been reported to function as an oncogene in gastric cancer, and its overexpression may contribute to gastric carcinogenesis. Li et al[58] demonstrated the upregulation of lncRNA H19 in gastric cancer tissues compared with paired normal tissues, and its positive correlation with lymph node metastasis and clinical stage. In vitro, upregulation of H19 could accelerate the proliferation, migration and invasion of gastric cancer cells, while knockdown of H19 caused apoptosis[61,65-67]. Moreover, Hashad et al[68] demonstrated that H19 was upregulated in the plasma of gastric cancer patients, making it a potential non-invasive diagnostic biomarker for gastric cancer.

Multiple previous researchers have presented the potential functional mechanisms of H19 as an oncogene in gastric cancer. Studies have shown that H19 and miR-675, the primary precursor of H19, act together as oncogenes by promoting cell growth and malignant transformation in gastric cancer[58]. H19 expression was negatively related to the expression of miR-141 in gastric cancer. The proliferation and invasion of gastric cancer could be accelerated by H19, but suppressed by miR-141. The competitive inhibitory relationship between H19 and miR-141 plays significant roles in the development of gastric cancer[69]. Other research demonstrated that the H19-PEG10 axis is involved in EMT, and that the knockdown of this axis could induce tremendous changes in the expression of EMT-associated proteins, making it a potential therapeutic target in gastric cancer[70].

Growth arrest-specific transcript 5

Growth arrest-specific transcript 5 (GAS5), a long non-coding RNA of approximately 650 nts, was originally isolated when screening for potential tumor suppressor genes during growth arrest[71]. The aberrant expression of GAS5 has been found in a variety of human malignancies, including prostate cancer, renal cell carcinoma, and breast cancer. Furthermore, by regulating apoptosis and the cell cycle, GAS5 managed to arrest the growth of many cancer cell lines[72-74]. Given the statistics above, the potential tumor suppressor role of GAS5 is clear. In a study that retrospectively analyzed the expression of GAS5 in 89 patients with gastric carcinoma, Sun et al[75] found that the decreased GAS5 expression was a common event, and that downregulation of GAS5 was positively correlated with tumor size, tumor stage, invasion depth and regional lymph nodes. Another study also demonstrated lower expression levels of GAS5 in gastric cancer tissues vs non-cancerous tissues, and its positive relation to tumor size and clinical stage[76].

The downregulation of GAS5 in gastric cancer has been generally proven, however the functional mechanisms of it remain to be elucidated. Accumulating evidence shows that GAS5 could function by binding with miRNA during the process of tumorigenesis. Li et al[77] found that overexpression of GAS5 could suppress cell proliferation in gastric cancer cells by negatively regulating miR-222, which was proven to be an oncogenic miRNA. Liu et al[78] showed that GAS5 expression in gastric cancer cells was inversely correlated with upregulated expression of miR-23a, indicating that GAS5 affected the biological behavior of gastric cancer by negatively regulating miR-23a expression. GAS5 has also been reported to be further downregulated in Adriamycin (ADM)-resistant gastric cancer cells. Nevertheless, when ADM-resistant gastric cell lines were transfected to promote GAS5 overexpression, they were more sensitive to ADM treatment, suggesting that GAS5 may act as a potential therapeutic target in gastric cancer treatment[79].

Maternally expressed gene 3

Located on chromosome 14q32.3, maternally expressed gene 3 (MEG3) is downregulated in multiple cancer tissues and cells[80,81]. It has been proven that MEG3 is a tumor suppressor gene involved in various types of cancers, including gastric cancer. A previous study that detected MEG3 expression in 31 patients with gastric cancer showed that MEG3 was significantly downregulated in gastric cancer tissues vs adjacent non-cancerous tissues. Furthermore, it demonstrated that MEG3 expression was negatively-related to tumor size and positively-related to overall survival rates of gastric cancer patients[82]. Accumulating studies demonstrated that overexpression of MEG3 could inhibit proliferation and metastasis, and that MEG3 was strongly correlated with deep tumor invasion, advanced metastasis and poor gastric cancer prognosis [82-84].

Increasing evidence reveals that lncRNA might play a crucial role in the occurrence and development of gastric cancer by interacting with miRNAs and promoting signaling pathways[85,86]. Studies showed that MEG3 could act as a competing endogenous RNA that sponges different miRNAs, such as miR-148a, miR-770, miR-181 and miR-141, to regulate the malignant activity of gastric cancer[83,87-89]. Other studies showed that overexpression of MEG3 promoted the expression of p53 in gastric cancer cell lines, indicating that MEG3 may suppress the proliferation and metastasis of gastric cancer via p53-dependent transcription pathways[82].

Long intergenic non-coding RNA 00152

Located on chromosome 2p11.2, long intergenic non-coding RNA 00152 (LINC00152) has an 828 nt-long transcript[90]. In a study in which the expression level of LINC00152 was detected in 71 gastric cancer tissues and their paired non-cancerous tissues, Pang et al[91] found remarkable overexpression of LINC00152 in gastric carcinoma, making it a potential novel biomarker for predicting gastric cancer. Moreover, high expression of LINC00152 was positively-correlated with tumor size, invasion depth and prognosis[92].

Functional analysis showed that silencing LINC00152 could inhibit cell proliferation, arrest cell cycle at the G1 phase, induce late apoptosis, suppress EMT and inhibit cell migration and invasion[93]. Another study demonstrated that gastric cancer cell proliferation could be remarkably inhibited by knocking down LINC00152. Moreover, LINC00152 could exert its function by binding to the oncogenic driver epidermal growth factor receptor (EGFR), leading to subsequent EGFR activation, which is a significant step in the tumorigenesis of gastric cancer[94]. Huang et al[92] discovered that LINC00152 was inversely-related to miR-193a-3p, which could significantly reduce gastric cancer cell proliferation and inhibit tumor growth by targeting MCL1. Thus, LINC00152 exerts its biological effects in gastric cancer development through the LINC00152/miR-193a-3p/MCL1 pathway[92]. LINC00152 could also bind to the enhancer of zeste homolog 2 (EZH2), which might lead to the repression of p15 and p21, thereby inducing gastric cancer cell cycle progression[95].

Urothelial carcinoma-associated 1

Researchers first discovered urothelial carcinoma-associated 1 (UCA1) in urinary bladder carcinoma. UCA1 was then shown to be an oncogenic long non-coding RNA[96,97]. Deregulation of UCA1 has been reported in a variety of additional human malignancies as well, such as melanoma, breast cancer, colorectal cancer, and tongue squamous cell carcinoma[98-100]. Recently, UCA1 has consistently been proven to play significant roles in the pathogenesis of gastric cancer. In a previous study, which analyzed UCA1 expression in 112 tumor and adjacent normal tissue samples from gastric cancer patients, researchers found that UCA1 was dramatically overexpressed in gastric cancer tissues and cell lines. Further clinicopathological analysis showed that the expression level of UCA1 was positively related to tumor size, invasion depth, TNM stage and poor overall survival[101].

Functional studies revealed that UCA1 expression could enhance cell proliferation, colony formation, and invasion of gastric cancer cells, and that silencing of UCA1 inhibits tumor growth. Gu et al[102] found that UCA1 might function by both negatively regulating miR-590-3p expression and activating the expression of its downstream target CREB1. UCA1/miR590-3p/CREB1 may be a potential target for the treatment of gastric cancer[102]. Another study indicated that knockdown of UCA1 reduced EMT-related protein levels, and that this effect could be partially rescued by treatment with transforming growth factor β1 (TGFβ1). Hypothetically, UCA1 might promote the proliferation, invasion and metastasis of gastric cancer upon TGFβ1 induction[103]. Moreover, Shang et al[104] demonstrated that chemotherapeutic resistance to ADM in gastric cancer cells was depressed, and that the half maximal inhibitory concentration (IC50) of ADM was also strongly decreased by silencing UCA1, thus making it a potential chemotherapeutic target for gastric cancer.

Metastasis-associated lung adenocarcinoma transcript 1

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), encoded on chromosome 11q13.1, is a long non-coding RNA with a length of more than 8000 nts. In response to growth signals, MALAT1 could bind to unmethylated PRC2 proteins and thus activate the growth control program[105]. At first, researchers found that MALAT1 could function as a metastatic biomarker for early-stage lung carcinoma[106]. Recently, MALAT1 overexpression has been observed in a variety of solid carcinomas, including gastric cancer, indicating that MALAT1 plays an important role in cancer development and metastasis[107-112]. By analyzing expression levels of MALAT1 in gastric cancer tissues and paired non-cancerous tissues, researchers revealed the upregulation of MALAT1 and the positive correlation between expression level and local invasion, lymph node invasion, peritoneal metastasis and short overall survival time[113,114]{Feng, 2017 #301;Okugawa, 2014 #329;Li, 2017 #333}. Another study showed that MALAT1 was aberrantly highly expressed in gastric cancer patients with distant metastasis compared to those without metastasis. Furthermore, functional studies demonstrated that EMT could be prevented by epigenetically silencing MALAT1, thus inhibiting cancer cell migration and invasion[115,116]. According to this evidence, the diagnostic potential of MALAT1 for gastric cancer is unequivocal.

An in vitro study confirmed that MALAT1 was negatively-correlated with miR-1297 expression, which promotes cell proliferation and invasion by targeting HMGA2. Moreover, silencing MALAT1 could reduce HMGA2 protein levels by eliminating miR-1297 inhibition, thus indicating that MALAT1 functions as an oncogenic lncRNA in part by modulating HMGA2 expression[113]. Another report illustrated that MALAT1 inhibited the expression of tumor suppressor PCDH10 by binding to EZH2, leading to gastric cancer cell migration and invasion[117]. MALAT1 could also serve as a competing endogenous RNA for miR-23b-3p and diminish its inhibitory effect on ATG12, which is a significant regulator of autophagy. This would thus promote chemo-induced autophagy and chemoresistance in gastric cancer cells. These findings revealed that MALAT1 could function as a therapeutic target for gastric cancer[118].

In addition to the well-documented lncRNAs discussed above, many other lncRNAs play important pathological roles in gastric cancer (Table 1). ANRIL is an antisense lncRNA located in the INK4 gene area. ANRIL has been shown to be overexpressed in gastric cancer and positively-related to tumor size, TNM stage and decreased survival. ANRIL regulates the development of gastric cancer by modulating miR-99a/miR-449a through the mTOR and CDK6/E2F1 pathways[119-121]. FENDRR is one of the lncRNAs that plays significant roles in tumorigenesis. Researchers have demonstrated the downregulation of FENDRR and its correlation with invasion depth, metastatic lymph nodes and poor patient prognosis. FENDRR exerts its function by targeting FN1 and MMP2/MMP9[122]. Other lncRNAs found to be overexpressed in gastric cancer include AFAP1-AS1, Sox2ot and CCAT2, while Linc00261, SNHG5 and LincRNA717 were downregulated in gastric cancer[123-129].

Table 1 Deregulations of long non-coding RNA associated with gastric cancer in this review.
LncRNADeregulationBiological rolesRef.
HOTAIRUpregulatedInduces EMT and promotes metastasis[46-55]
H19UpregulatedPromotes cell growth, proliferation, invasion Promotes EMT[56-70]
GAS5DownregulatedSuppresses cell proliferation Sensitizes cells to ADM treatment[71-79]
MEG3DownregulatedSuppresses cell proliferation and metastasis[80-89]
LINC00152UpregulatedPromotes cell proliferation and tumor growth[90-95]
UCA1UpregulatedPromotes cell proliferation, invasion, metastasis Depresses resistance to ADM treatment[96-104]
MALAT1UpregulatedPromotes cell proliferation and invasion Promotes chemo-induced autophagy and chemoresistance[105-118]
ANRILUpregulatedPromotes tumor growth and metastasis[119-121]
FENDRRDownregulatedInhibits migration and invasion[122]
AFAP1-AS1UpregulatedPromotes cell proliferation and cell cycle progression[123, 124]
Sox2otUpregulatedPromotes cell growth and motility[125]
CCAT2UpregulatedPromotes EMT[126]
Linc00261DownregulatedRepresses metastasis Inhibits EMT[127]
SNHG5DownregulatedSuppresses cell proliferation and metastasis[128]
LincRNA717DownregulatedInhibits tumor growth and invasion[129]
CONCLUSION

In summary, with the rapid development of various bioinformatic techniques, thousands of lncRNAs have been discovered. Thus far, various studies have proven the significant functions of lncRNAs in tumorigenesis of gastric cancer. Aberrantly-expressed lncRNAs might be used as diagnostic biomarkers, prognostic markers, and therapeutic targets for gastric cancer. However, our current understanding of lncRNAs in relation to gastric cancer remains limited. As a result, more investigations are necessary to gain a better understanding of lncRNAs and their mechanisms in gastric cancer.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Oncology

Country of origin: China

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Chivu-Economescu M, Gazouli M, Sugimura H, Yoshiyama H S- Editor: Ji FF L- Editor: Filipodia E- Editor: Tan WW

References
1.  Li PF, Chen SC, Xia T, Jiang XM, Shao YF, Xiao BX, Guo JM. Non-coding RNAs and gastric cancer. World J Gastroenterol. 2014;20:5411-5419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 98]  [Cited by in F6Publishing: 112]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
2.  Suzuki H, Oda I, Abe S, Sekiguchi M, Mori G, Nonaka S, Yoshinaga S, Saito Y. High rate of 5-year survival among patients with early gastric cancer undergoing curative endoscopic submucosal dissection. Gastric Cancer. 2016;19:198-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 162]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
3.  Kim YI, Kim YW, Choi IJ, Kim CG, Lee JY, Cho SJ, Eom BW, Yoon HM, Ryu KW, Kook MC. Long-term survival after endoscopic resection versus surgery in early gastric cancers. Endoscopy. 2015;47:293-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
4.  Tanabe S, Ishido K, Higuchi K, Sasaki T, Katada C, Azuma M, Naruke A, Kim M, Koizumi W. Long-term outcomes of endoscopic submucosal dissection for early gastric cancer: a retrospective comparison with conventional endoscopic resection in a single center. Gastric Cancer. 2014;17:130-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 94]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
5.  Oda I, Oyama T, Abe S, Ohnita K, Kosaka T, Hirasawa K, Ishido K, Nakagawa M, Takahashi S. Preliminary results of multicenter questionnaire study on long-term outcomes of curative endoscopic submucosal dissection for early gastric cancer. Dig Endosc. 2014;26:214-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 66]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
6.  Choi IJ. Endoscopic gastric cancer screening and surveillance in high-risk groups. Clin Endosc. 2014;47:497-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 54]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
7.  Lazăr D, Tăban S, Dema A, Cornianu M, Goldiş A, Raţiu I, Sporea I. Gastric cancer: the correlation between the clinicopathological factors and patients’ survival (I). Rom J Morphol Embryol. 2009;50:41-50.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Yu H, Yang AM, Lu XH, Zhou WX, Yao F, Fei GJ, Guo T, Yao LQ, He LP, Wang BM. Magnifying narrow-band imaging endoscopy is superior in diagnosis of early gastric cancer. World J Gastroenterol. 2015;21:9156-9162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 3.8]  [Reference Citation Analysis (1)]
9.  Kapranov P, Cawley SE, Drenkow J, Bekiranov S, Strausberg RL, Fodor SP, Gingeras TR. Large-scale transcriptional activity in chromosomes 21 and 22. Science. 2002;296:916-919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 635]  [Cited by in F6Publishing: 686]  [Article Influence: 31.2]  [Reference Citation Analysis (0)]
10.  Kapranov P, Willingham AT, Gingeras TR. Genome-wide transcription and the implications for genomic organization. Nat Rev Genet. 2007;8:413-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 528]  [Cited by in F6Publishing: 529]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
11.  Taby R, Issa JP. Cancer epigenetics. CA Cancer J Clin. 2010;60:376-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 314]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
12.  Majem B, Rigau M, Reventós J, Wong DT. Non-coding RNAs in saliva: emerging biomarkers for molecular diagnostics. Int J Mol Sci. 2015;16:8676-8698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
13.  Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, Stadler PF, Hertel J, Hackermüller J, Hofacker IL. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 2007;316:1484-1488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1800]  [Cited by in F6Publishing: 1823]  [Article Influence: 107.2]  [Reference Citation Analysis (0)]
14.  Takenaka K, Chen BJ, Modesitt SC, Byrne FL, Hoehn KL, Janitz M. The emerging role of long non-coding RNAs in endometrial cancer. Cancer Genet. 2016;209:445-455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
15.  Kobayashi R, Miyagawa R, Yamashita H, Morikawa T, Okuma K, Fukayama M, Ohtomo K, Nakagawa K. Increased expression of long non-coding RNA XIST predicts favorable prognosis of cervical squamous cell carcinoma subsequent to definitive chemoradiation therapy. Oncol Lett. 2016;12:3066-3074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
16.  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)]
17.  Novikova IV, Hennelly SP, Sanbonmatsu KY. Sizing up long non-coding RNAs: do lncRNAs have secondary and tertiary structure? Bioarchitecture. 2012;2:189-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 103]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
18.  Okazaki Y, Furuno M, Kasukawa T, Adachi J, Bono H, Kondo S, Nikaido I, Osato N, Saito R, Suzuki H. Analysis of the mouse transcriptome based on functional annotation of 60,770 full-length cDNAs. Nature. 2002;420:563-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1203]  [Cited by in F6Publishing: 1225]  [Article Influence: 55.7]  [Reference Citation Analysis (0)]
19.  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)]
20.  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)]
21.  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)]
22.  Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3103]  [Cited by in F6Publishing: 3195]  [Article Influence: 213.0]  [Reference Citation Analysis (0)]
23.  Gong C, Maquat LE. lncRNAs transactivate STAU1-mediated mRNA decay by duplexing with 3’ UTRs via Alu elements. Nature. 2011;470:284-288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 915]  [Cited by in F6Publishing: 954]  [Article Influence: 73.4]  [Reference Citation Analysis (0)]
24.  Niazi F, Valadkhan S. Computational analysis of functional long noncoding RNAs reveals lack of peptide-coding capacity and parallels with 3’ UTRs. RNA. 2012;18:825-843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 115]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
25.  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)]
26.  Bartonicek N, Maag JL, Dinger ME. Long noncoding RNAs in cancer: mechanisms of action and technological advancements. Mol Cancer. 2016;15:43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 329]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
27.  Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S. The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 2015;47:199-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1788]  [Cited by in F6Publishing: 1975]  [Article Influence: 219.4]  [Reference Citation Analysis (0)]
28.  Cao J. The functional role of long non-coding RNAs and epigenetics. Biol Proced Online. 2014;16:11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 242]  [Article Influence: 24.2]  [Reference Citation Analysis (0)]
29.  Zhang R, Xia LQ, Lu WW, Zhang J, Zhu JS. LncRNAs and cancer. Oncol Lett. 2016;12:1233-1239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 77]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
30.  Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci USA. 2009;106:11667-11672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2182]  [Cited by in F6Publishing: 2276]  [Article Influence: 151.7]  [Reference Citation Analysis (0)]
31.  Mohammad F, Mondal T, Guseva N, Pandey GK, Kanduri C. Kcnq1ot1 noncoding RNA mediates transcriptional gene silencing by interacting with Dnmt1. Development. 2010;137:2493-2499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 207]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
32.  Merry CR, Forrest ME, Sabers JN, Beard L, Gao XH, Hatzoglou M, Jackson MW, Wang Z, Markowitz SD, Khalil AM. DNMT1-associated long non-coding RNAs regulate global gene expression and DNA methylation in colon cancer. Hum Mol Genet. 2015;24:6240-6253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 120]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
33.  Arab K, Park YJ, Lindroth AM, Schäfer A, Oakes C, Weichenhan D, Lukanova A, Lundin E, Risch A, Meister M. Long noncoding RNA TARID directs demethylation and activation of the tumor suppressor TCF21 via GADD45A. Mol Cell. 2014;55:604-614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 200]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
34.  Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA. A chromatin landmark and transcription initiation at most promoters in human cells. Cell. 2007;130:77-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1484]  [Cited by in F6Publishing: 1495]  [Article Influence: 87.9]  [Reference Citation Analysis (0)]
35.  Ashe HL, Monks J, Wijgerde M, Fraser P, Proudfoot NJ. Intergenic transcription and transinduction of the human beta-globin locus. Genes Dev. 1997;11:2494-2509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 292]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
36.  Martens JA, Laprade L, Winston F. Intergenic transcription is required to repress the Saccharomyces cerevisiae SER3 gene. Nature. 2004;429:571-574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 459]  [Cited by in F6Publishing: 450]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
37.  Feng J, Bi C, Clark BS, Mady R, Shah P, Kohtz JD. The Evf-2 noncoding RNA is transcribed from the Dlx-5/6 ultraconserved region and functions as a Dlx-2 transcriptional coactivator. Genes Dev. 2006;20:1470-1484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 548]  [Cited by in F6Publishing: 545]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
38.  Martianov I, Ramadass A, Serra Barros A, Chow N, Akoulitchev A. Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript. Nature. 2007;445:666-670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 552]  [Cited by in F6Publishing: 579]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
39.  Rossi MN, Antonangeli F. LncRNAs: New Players in Apoptosis Control. Int J Cell Biol. 2014;2014:473857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 98]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
40.  DeOcesano-Pereira C, Amaral MS, Parreira KS, Ayupe AC, Jacysyn JF, Amarante-Mendes GP, Reis EM, Verjovski-Almeida S. Long non-coding RNA INXS is a critical mediator of BCL-XS induced apoptosis. Nucleic Acids Res. 2014;42:8343-8355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
41.  Ogawa Y, Sun BK, Lee JT. Intersection of the RNA interference and X-inactivation pathways. Science. 2008;320:1336-1341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 224]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
42.  Tay Y, Rinn J, Pandolfi PP. The multilayered complexity of ceRNA crosstalk and competition. Nature. 2014;505:344-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2305]  [Cited by in F6Publishing: 2785]  [Article Influence: 278.5]  [Reference Citation Analysis (0)]
43.  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)]
44.  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)]
45.  Chung S, Nakagawa H, Uemura M, Piao L, Ashikawa K, Hosono N, Takata R, Akamatsu S, Kawaguchi T, Morizono T. Association of a novel long non-coding RNA in 8q24 with prostate cancer susceptibility. Cancer Sci. 2011;102:245-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 226]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
46.  Feng X, Huang S. Effect and mechanism of lncRNA HOTAIR on occurrence and development of gastric cancer. J Cell Biochem. 2017; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
47.  Fagoonee S, Durazzo M. HOTAIR and gastric cancer: a lesson from two meta-analyses. Panminerva Med. 2017;59:201-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
48.  Zhou X, Chen J, Tang W. The molecular mechanism of HOTAIR in tumorigenesis, metastasis, and drug resistance. Acta Biochim Biophys Sin (Shanghai). 2014;46:1011-1015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
49.  Wu Y, Zhang L, Wang Y, Li H, Ren X, Wei F, Yu W, Wang X, Zhang L, Yu J. Long noncoding RNA HOTAIR involvement in cancer. Tumour Biol. 2014;35:9531-9538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 105]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
50.  Hajjari M, Salavaty A. HOTAIR: an oncogenic long non-coding RNA in different cancers. Cancer Biol Med. 2015;12:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 215]  [Reference Citation Analysis (0)]
51.  Song B, Guan Z, Liu F, Sun D, Wang K, Qu H. Long non-coding RNA HOTAIR promotes HLA-G expression via inhibiting miR-152 in gastric cancer cells. Biochem Biophys Res Commun. 2015;464:807-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 62]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
52.  Endo H, Shiroki T, Nakagawa T, Yokoyama M, Tamai K, Yamanami H, Fujiya T, Sato I, Yamaguchi K, Tanaka N. Enhanced expression of long non-coding RNA HOTAIR is associated with the development of gastric cancer. PLoS One. 2013;8:e77070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 202]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
53.  Chen WM, Chen WD, Jiang XM, Jia XF, Wang HM, Zhang QJ, Shu YQ, Zhao HB. HOX transcript antisense intergenic RNA represses E-cadherin expression by binding to EZH2 in gastric cancer. World J Gastroenterol. 2017;23:6100-6110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
54.  Liu YW, Sun M, Xia R, Zhang EB, Liu XH, Zhang ZH, Xu TP, De W, Liu BR, Wang ZX. LincHOTAIR epigenetically silences miR34a by binding to PRC2 to promote the epithelial-to-mesenchymal transition in human gastric cancer. Cell Death Dis. 2015;6:e1802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 151]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
55.  Du M, Wang W, Jin H, Wang Q, Ge Y, Lu J, Ma G, Chu H, Tong N, Zhu H. The association analysis of lncRNA HOTAIR genetic variants and gastric cancer risk in a Chinese population. Oncotarget. 2015;6:31255-31262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 88]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
56.  Bartolomei MS, Zemel S, Tilghman SM. Parental imprinting of the mouse H19 gene. Nature. 1991;351:153-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 889]  [Cited by in F6Publishing: 881]  [Article Influence: 26.7]  [Reference Citation Analysis (0)]
57.  Zhang L, Zhou Y, Huang T, Cheng AS, Yu J, Kang W, To KF. The Interplay of LncRNA-H19 and Its Binding Partners in Physiological Process and Gastric Carcinogenesis. Int J Mol Sci. 2017;18:pii: E450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
58.  Li H, Yu B, Li J, Su L, Yan M, Zhu Z, Liu B. Overexpression of lncRNA H19 enhances carcinogenesis and metastasis of gastric cancer. Oncotarget. 2014;5:2318-2329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 345]  [Cited by in F6Publishing: 416]  [Article Influence: 46.2]  [Reference Citation Analysis (0)]
59.  Smits G, Mungall AJ, Griffiths-Jones S, Smith P, Beury D, Matthews L, Rogers J, Pask AJ, Shaw G, VandeBerg JL. Conservation of the H19 noncoding RNA and H19-IGF2 imprinting mechanism in therians. Nat Genet. 2008;40:971-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 142]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
60.  Luo M, Li Z, Wang W, Zeng Y, Liu Z, Qiu J. Upregulated H19 contributes to bladder cancer cell proliferation by regulating ID2 expression. FEBS J. 2013;280:1709-1716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 118]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
61.  Berteaux N, Aptel N, Cathala G, Genton C, Coll J, Daccache A, Spruyt N, Hondermarck H, Dugimont T, Curgy JJ. A novel H19 antisense RNA overexpressed in breast cancer contributes to paternal IGF2 expression. Mol Cell Biol. 2008;28:6731-6745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 121]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
62.  Byun HM, Wong HL, Birnstein EA, Wolff EM, Liang G, Yang AS. Examination of IGF2 and H19 loss of imprinting in bladder cancer. Cancer Res. 2007;67:10753-10758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 99]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
63.  Berteaux N, Lottin S, Monté D, Pinte S, Quatannens B, Coll J, Hondermarck H, Curgy JJ, Dugimont T, Adriaenssens E. H19 mRNA-like noncoding RNA promotes breast cancer cell proliferation through positive control by E2F1. J Biol Chem. 2005;280:29625-29636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 283]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
64.  Kim SJ, Park SE, Lee C, Lee SY, Jo JH, Kim JM, Oh YK. Alterations in promoter usage and expression levels of insulin-like growth factor-II and H19 genes in cervical carcinoma exhibiting biallelic expression of IGF-II. Biochim Biophys Acta. 2002;1586:307-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
65.  Gao T, He B, Pan Y, Xu Y, Li R, Deng Q, Sun H, Wang S. Long non-coding RNA 91H contributes to the occurrence and progression of esophageal squamous cell carcinoma by inhibiting IGF2 expression. Mol Carcinog. 2015;54:359-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
66.  Livingstone C. IGF2 and cancer. Endocr Relat Cancer. 2013;20:R321-R339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 207]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
67.  Schmidt JV, Levorse JM, Tilghman SM. Enhancer competition between H19 and Igf2 does not mediate their imprinting. Proc Natl Acad Sci USA. 1999;96:9733-9738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 79]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
68.  Hashad D, Elbanna A, Ibrahim A, Khedr G. Evaluation of the Role of Circulating Long Non-Coding RNA H19 as a Promising Novel Biomarker in Plasma of Patients with Gastric Cancer. J Clin Lab Anal. 2016;30:1100-1105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 74]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
69.  Zhou X, Ye F, Yin C, Zhuang Y, Yue G, Zhang G. The Interaction Between MiR-141 and lncRNA-H19 in Regulating Cell Proliferation and Migration in Gastric Cancer. Cell Physiol Biochem. 2015;36:1440-1452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 154]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
70.  Ishii S, Yamashita K, Harada H, Ushiku H, Tanaka T, Nishizawa N, Yokoi K, Washio M, Ema A, Mieno H. The H19-PEG10/IGF2BP3 axis promotes gastric cancer progression in patients with high lymph node ratios. Oncotarget. 2017;8:74567-74581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
71.  Schneider C, King RM, Philipson L. Genes specifically expressed at growth arrest of mammalian cells. Cell. 1988;54:787-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 724]  [Cited by in F6Publishing: 773]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
72.  Qiao HP, Gao WS, Huo JX, Yang ZS. Long non-coding RNA GAS5 functions as a tumor suppressor in renal cell carcinoma. Asian Pac J Cancer Prev. 2013;14:1077-1082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 194]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
73.  Pickard MR, Mourtada-Maarabouni M, Williams GT. Long non-coding RNA GAS5 regulates apoptosis in prostate cancer cell lines. Biochim Biophys Acta. 2013;1832:1613-1623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 268]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
74.  Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT. GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 2009;28:195-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 596]  [Cited by in F6Publishing: 615]  [Article Influence: 38.4]  [Reference Citation Analysis (0)]
75.  Sun M, Jin FY, Xia R, Kong R, Li JH, Xu TP, Liu YW, Zhang EB, Liu XH, De W. Decreased expression of long noncoding RNA GAS5 indicates a poor prognosis and promotes cell proliferation in gastric cancer. BMC Cancer. 2014;14:319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 256]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
76.  Guo X, Deng K, Wang H, Xia J, Shan T, Liang Z, Yao L, Jin S. GAS5 Inhibits Gastric Cancer Cell Proliferation Partly by Modulating CDK6. Oncol Res Treat. 2015;38:362-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
77.  Li Y, Gu J, Lu H. The GAS5/miR-222 Axis Regulates Proliferation of Gastric Cancer Cells Through the PTEN/Akt/mTOR Pathway. Dig Dis Sci. 2017;62:3426-3437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
78.  Liu X, Jiao T, Wang Y, Su W, Tang Z, Han C. Long non-coding RNA GAS5 acts as a molecular sponge to regulate miR-23a in gastric cancer. Minerva Med. 2016;.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Zhang N, Wang AY, Wang XK, Sun XM, Xue HZ. GAS5 is downregulated in gastric cancer cells by promoter hypermethylation and regulates adriamycin sensitivity. Eur Rev Med Pharmacol Sci. 2016;20:3199-3205.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Benetatos L, Vartholomatos G, Hatzimichael E. MEG3 imprinted gene contribution in tumorigenesis. Int J Cancer. 2011;129:773-779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 210]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
81.  Lin SP, Youngson N, Takada S, Seitz H, Reik W, Paulsen M, Cavaille J, Ferguson-Smith AC. Asymmetric regulation of imprinting on the maternal and paternal chromosomes at the Dlk1-Gtl2 imprinted cluster on mouse chromosome 12. Nat Genet. 2003;35:97-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 377]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
82.  Wei GH, Wang X. lncRNA MEG3 inhibit proliferation and metastasis of gastric cancer via p53 signaling pathway. Eur Rev Med Pharmacol Sci. 2017;21:3850-3856.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Peng W, Si S, Zhang Q, Li C, Zhao F, Wang F, Yu J, Ma R. Long non-coding RNA MEG3 functions as a competing endogenous RNA to regulate gastric cancer progression. J Exp Clin Cancer Res. 2015;34:79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 213]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
84.  Sun M, Xia R, Jin F, Xu T, Liu Z, De W, Liu X. Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumour Biol. 2014;35:1065-1073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 200]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
85.  Yoon JH, Abdelmohsen K, Gorospe M. Functional interactions among microRNAs and long noncoding RNAs. Semin Cell Dev Biol. 2014;34:9-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 435]  [Cited by in F6Publishing: 498]  [Article Influence: 49.8]  [Reference Citation Analysis (0)]
86.  Sen R, Ghosal S, Das S, Balti S, Chakrabarti J. Competing endogenous RNA: the key to posttranscriptional regulation. ScientificWorldJournal. 2014;2014:896206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 227]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
87.  Guo W, Dong Z, Liu S, Qiao Y, Kuang G, Guo Y, Shen S, Liang J. Promoter hypermethylation-mediated downregulation of miR-770 and its host gene MEG3, a long non-coding RNA, in the development of gastric cardia adenocarcinoma. Mol Carcinog. 2017;56:1924-1934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 40]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
88.  Zhou X, Ji G, Ke X, Gu H, Jin W, Zhang G. MiR-141 Inhibits Gastric Cancer Proliferation by Interacting with Long Noncoding RNA MEG3 and Down-Regulating E2F3 Expression. Dig Dis Sci. 2015;60:3271-3282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
89.  Yan J, Guo X, Xia J, Shan T, Gu C, Liang Z, Zhao W, Jin S. MiR-148a regulates MEG3 in gastric cancer by targeting DNA methyltransferase 1. Med Oncol. 2014;31:879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 102]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
90.  Yu J, Liu Y, Guo C, Zhang S, Gong Z, Tang Y, Yang L, He Y, Lian Y, Li X. Upregulated long non-coding RNA LINC00152 expression is associated with progression and poor prognosis of tongue squamous cell carcinoma. J Cancer. 2017;8:523-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 89]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
91.  Pang Q, Ge J, Shao Y, Sun W, Song H, Xia T, Xiao B, Guo J. Increased expression of long intergenic non-coding RNA LINC00152 in gastric cancer and its clinical significance. Tumour Biol. 2014;35:5441-5447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 138]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
92.  Huang Y, Luo H, Li F, Yang Y, Ou G, Ye X, Li N. LINC00152 down-regulated miR-193a-3p to enhance MCL1 expression and promote gastric cancer cells proliferation. Biosci Rep. 2018;38:pii: BSR20171607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
93.  Zhao J, Liu Y, Zhang W, Zhou Z, Wu J, Cui P, Zhang Y, Huang G. Long non-coding RNA Linc00152 is involved in cell cycle arrest, apoptosis, epithelial to mesenchymal transition, cell migration and invasion in gastric cancer. Cell Cycle. 2015;14:3112-3123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 188]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
94.  Zhou J, Zhi X, Wang L, Wang W, Li Z, Tang J, Wang J, Zhang Q, Xu Z. Linc00152 promotes proliferation in gastric cancer through the EGFR-dependent pathway. J Exp Clin Cancer Res. 2015;34:135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 98]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
95.  Chen WM, Huang MD, Sun DP, Kong R, Xu TP, Xia R, Zhang EB, Shu YQ. Long intergenic non-coding RNA 00152 promotes tumor cell cycle progression by binding to EZH2 and repressing p15 and p21 in gastric cancer. Oncotarget. 2016;7:9773-9787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 117]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
96.  Wang Y, Chen W, Yang C, Wu W, Wu S, Qin X, Li X. Long non-coding RNA UCA1a(CUDR) promotes proliferation and tumorigenesis of bladder cancer. Int J Oncol. 2012;41:276-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
97.  Wang F, Li X, Xie X, Zhao L, Chen W. UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion. FEBS Lett. 2008;582:1919-1927.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 241]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
98.  Hong HH, Hou LK, Pan X, Wu CY, Huang H, Li B, Nie W. Long non-coding RNA UCA1 is a predictive biomarker of cancer. Oncotarget. 2016;7:44442-44447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
99.  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)]
100.  Gao J, Cao R, Mu H. Long non-coding RNA UCA1 may be a novel diagnostic and predictive biomarker in plasma for early gastric cancer. Int J Clin Exp Pathol. 2015;8:12936-12942.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Zheng Q, Wu F, Dai WY, Zheng DC, Zheng C, Ye H, Zhou B, Chen JJ, Chen P. Aberrant expression of UCA1 in gastric cancer and its clinical significance. Clin Transl Oncol. 2015;17:640-646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 101]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
102.  Gu L, Lu LS, Zhou DL, Liu ZC. UCA1 promotes cell proliferation and invasion of gastric cancer by targeting CREB1 sponging to miR-590-3p. Cancer Med. 2018;7:1253-1263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
103.  Zuo ZK, Gong Y, Chen XH, Ye F, Yin ZM, Gong QN, Huang JS. TGFβ1-Induced LncRNA UCA1 Upregulation Promotes Gastric Cancer Invasion and Migration. DNA Cell Biol. 2017;36:159-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
104.  Shang C, Guo Y, Zhang J, Huang B. Silence of long noncoding RNA UCA1 inhibits malignant proliferation and chemotherapy resistance to adriamycin in gastric cancer. Cancer Chemother Pharmacol. 2016;77:1061-1067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 88]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
105.  Yang L, Lin C, Liu W, Zhang J, Ohgi KA, Grinstein JD, Dorrestein PC, Rosenfeld MG. ncRNA- and Pc2 methylation-dependent gene relocation between nuclear structures mediates gene activation programs. Cell. 2011;147:773-788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 458]  [Cited by in F6Publishing: 493]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
106.  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)]
107.  Xu C, Yang M, Tian J, Wang X, Li Z. MALAT-1: a long non-coding RNA and its important 3’ end functional motif in colorectal cancer metastasis. Int J Oncol. 2011;39:169-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 178]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
108.  Sun Y, Wu J, Wu SH, Thakur A, Bollig A, Huang Y, Liao DJ. Expression profile of microRNAs in c-Myc induced mouse mammary tumors. Breast Cancer Res Treat. 2009;118:185-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 66]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
109.  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)]
110.  Fellenberg J, Bernd L, Delling G, Witte D, Zahlten-Hinguranage A. Prognostic significance of drug-regulated genes in high-grade osteosarcoma. Mod Pathol. 2007;20:1085-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 91]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
111.  Yamada K, Kano J, Tsunoda H, Yoshikawa H, Okubo C, Ishiyama T, Noguchi M. Phenotypic characterization of endometrial stromal sarcoma of the uterus. Cancer Sci. 2006;97:106-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 110]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
112.  Luo JH, Ren B, Keryanov S, Tseng GC, Rao UN, Monga SP, Strom S, Demetris AJ, Nalesnik M, Yu YP. Transcriptomic and genomic analysis of human hepatocellular carcinomas and hepatoblastomas. Hepatology. 2006;44:1012-1024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 389]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
113.  Li J, Gao J, Tian W, Li Y, Zhang J. Long non-coding RNA MALAT1 drives gastric cancer progression by regulating HMGB2 modulating the miR-1297. Cancer Cell Int. 2017;17:44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 57]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
114.  Okugawa Y, Toiyama Y, Hur K, Toden S, Saigusa S, Tanaka K, Inoue Y, Mohri Y, Kusunoki M, Boland CR. Metastasis-associated long non-coding RNA drives gastric cancer development and promotes peritoneal metastasis. Carcinogenesis. 2014;35:2731-2739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 214]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
115.  Lee NK, Lee JH, Ivan C, Ling H, Zhang X, Park CH, Calin GA, Lee SK. MALAT1 promoted invasiveness of gastric adenocarcinoma. BMC Cancer. 2017;17:46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 47]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
116.  Xia H, Chen Q, Chen Y, Ge X, Leng W, Tang Q, Ren M, Chen L, Yuan D, Zhang Y. The lncRNA MALAT1 is a novel biomarker for gastric cancer metastasis. Oncotarget. 2016;7:56209-56218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 88]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
117.  Qi Y, Ooi HS, Wu J, Chen J, Zhang X, Tan S, Yu Q, Li YY, Kang Y, Li H. MALAT1 long ncRNA promotes gastric cancer metastasis by suppressing PCDH10. Oncotarget. 2016;7:12693-12703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 87]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
118.  YiRen H, YingCong Y, Sunwu Y, Keqin L, Xiaochun T, Senrui C, Ende C, XiZhou L, Yanfan C. Long noncoding RNA MALAT1 regulates autophagy associated chemoresistance via miR-23b-3p sequestration in gastric cancer. Mol Cancer. 2017;16:174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 245]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
119.  Lan WG, Xu DH, Xu C, Ding CL, Ning FL, Zhou YL, Ma LB, Liu CM, Han X. Silencing of long non-coding RNA ANRIL inhibits the development of multidrug resistance in gastric cancer cells. Oncol Rep. 2016;36:263-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 74]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
120.  Deng W, Wang J, Zhang J, Cai J, Bai Z, Zhang Z. TET2 regulates LncRNA-ANRIL expression and inhibits the growth of human gastric cancer cells. IUBMB Life. 2016;68:355-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 45]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
121.  Zhang EB, Kong R, Yin DD, You LH, Sun M, Han L, Xu TP, Xia R, Yang JS, De W. Long noncoding RNA ANRIL indicates a poor prognosis of gastric cancer and promotes tumor growth by epigenetically silencing of miR-99a/miR-449a. Oncotarget. 2014;5:2276-2292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 299]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
122.  Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, Chen WM, Han L, Zhang EB, Kong R. Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol. 2014;7:63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 216]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
123.  Feng Y, Zhang Q, Wang J, Liu P. Increased lncRNA AFAP1-AS1 expression predicts poor prognosis and promotes malignant phenotypes in gastric cancer. Eur Rev Med Pharmacol Sci. 2017;21:3842-3849.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Zhang F, Li J, Xiao H, Zou Y, Liu Y, Huang W. AFAP1-AS1: A novel oncogenic long non-coding RNA in human cancers. Cell Prolif. 2018;51:e12397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 46]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
125.  Zhang Y, Yang R, Lian J, Xu H. LncRNA Sox2ot overexpression serves as a poor prognostic biomarker in gastric cancer. Am J Transl Res. 2016;8:5035-5043.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Wang YJ, Liu JZ, Lv P, Dang Y, Gao JY, Wang Y. Long non-coding RNA CCAT2 promotes gastric cancer proliferation and invasion by regulating the E-cadherin and LATS2. Am J Cancer Res. 2016;6:2651-2660.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Yu Y, Li L, Zheng Z, Chen S, Chen E, Hu Y. Long non-coding RNA linc00261 suppresses gastric cancer progression via promoting Slug degradation. J Cell Mol Med. 2017;21:955-967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 69]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
128.  Zhao L, Guo H, Zhou B, Feng J, Li Y, Han T, Liu L, Li L, Zhang S, Liu Y. Long non-coding RNA SNHG5 suppresses gastric cancer progression by trapping MTA2 in the cytosol. Oncogene. 2016;35:5770-5780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 87]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
129.  Shao Y, Chen H, Jiang X, Chen S, Li P, Ye M, Li Q, Sun W, Guo J. Low expression of lncRNA-HMlincRNA717 in human gastric cancer and its clinical significances. Tumour Biol. 2014;35:9591-9595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 52]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]