Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

LINC00922 promotes deterioration of gastric cancer

  • Hua Ge ,

    Roles Conceptualization, Data curation, Funding acquisition, Methodology, Writing – original draft

    zyyyhuage@126.com

    Affiliation Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

  • Yan Yan,

    Roles Conceptualization, Formal analysis, Software, Supervision, Writing – original draft, Writing – review & editing

    Affiliation Quality Control Department, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

  • Fei Tian,

    Roles Data curation, Software

    Affiliation Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

  • Lingfei Guo,

    Roles Data curation

    Affiliation Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

  • Xueyan He,

    Roles Formal analysis, Validation

    Affiliation Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

  • Can Wang,

    Roles Formal analysis

    Affiliation Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

  • Jiacheng Song,

    Roles Investigation

    Affiliation Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

  • Zhilong Deng

    Roles Investigation

    Affiliation Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, People’s Republic of China

Abstract

Several studies have demonstrated the association of lncRNAs with a variety of cancers. Here, we explored the role of LINC00922 in gastric cancer (GC) using bioinformatics approaches and in vitro experiments. We examined the expression of LINC00922 and the prognosis of GC patients based on data from The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA). LINC00922-related genes were identified by the Multi Experiment Matrix (MEM) database and The Atlas of Noncoding RNAs in Cancer (TANRIC), followed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction analysis. The significance of LINC00922 in cell proliferation, apoptosis, invasion and migration was assessed by MTT assay, flow cytometry, Transwell assay and wound-healing assay. The expression of LINC00922 was increased in GC tissues compared with adjacent non-tumor tissues, and increased LINC00922 expression was correlated with poor overall survival and disease-free survival. In addition, 336 overlapping genes were identified by the MEM database and TANRIC and found to be involved in GC-related biological processes, such as cell adhesion and migration, as well as TGF-β signaling. In the protein-protein interaction network, hub genes, such as FSTL3 and LAMC1, were identified. LINC00922 overexpression significantly promoted cell proliferation and invasion in vitro, whereas LINC00922 knockdown exerted opposite effects. In summary, our findings indicate that LINC00922 is overexpressed in GC tissues, suggesting that it might play a role in the development and progression of GC, and thus, it might serve as a prognostic indicator of GC.

Introduction

Gastric cancer is the fifth most common cancer and the third most fatal cancer worldwide. In 2018, there were more than one million cases of GC, which caused an estimated 783,000 deaths. As such, GC is a public health-related burden for developed and developing countries [1]. Almost half of the world’s GC cases and deaths occur in China, with an estimated 500,000 deaths in 2015 [2]. GC is a multi-factorial disease, and both environmental (68%) and genetic (22%) factors have been reported to associate with its etiology [3]. Personal lifestyle choices, such as insufficient fruits and vegetables in the diet, excessive alcohol consumption and high intake of salt, have been demonstrated to associate with GC [46]. In addition, a family history of GC and Helicobacter pylori infection both increase the risk of developing GC [7,8]. As most patients have non-specific symptoms in the early stages of the disease, GC can progress to advanced stages, which is when most patients are diagnosed. Therefore, it is important to identify reliable diagnostic and prognostic markers of GC, with the hopes of detecting GC in the early stages of the disease.

Long non-coding RNAs (lncRNAs) are a group of non-coding RNAs of more than 200 nucleotides in length without open reading frames [9]. LncRNAs have a variety of functions, including post-transcriptional regulation, gene regulation and intercellular signaling [10,11], and dysregulation of lncRNAs is involved in a variety of diseases such as cancer [12]. Accumulating evidence has revealed that lncRNAs are essential for cancer progression and metastasis [13,14]. Recently, new lncRNAs have been identified, and bioinformatics approaches have been used to understand the roles of lncRNAs [15,16]. However, the function of LINC00922 in the development of GC is unclear. Thus, we investigated the role of LINC00922 in GC using data mining and in vitro experiments.

Results

Expression and prognostic value of LINC00922 in GC based on TCGA data

A flow chart of the study design is shown in Fig 1A. In total, we extracted 407 samples from TCGA datasets, including 375 tumor and 32 non-tumor samples. As shown in Table 1, LINC00922 expression was increased in GC tissues (2.908 ± 0.077) and decreased in adjacent non-tumor tissues (0.790 ± 0.189) (P < 0.001). However, LINC00922 expression was not correlated with age, gender, race, tumor depth, lymphnode invasion, remote metastasis, and pathological stage. Next, we used GEPIA to determine the prognostic value of LINC00922 in GC. As shown in Fig 1B and 1C, increased LINC00922 expression was significantly correlated with poor overall survival (P = 0.0056) and disease-free survival (P = 0.0091), indicating that LINC00922 functions as an oncogene in GC.

thumbnail
Fig 1. Flow chart and Kaplan-Meier curves for survival of LINC00922 from GEPIA.

(A) Flow chart. (B) Overall survival. (C) Disease-free survival.

https://doi.org/10.1371/journal.pone.0267798.g001

thumbnail
Table 1. Expression of LINC00922 and clinicopathological features in GC patients in TCGA.

https://doi.org/10.1371/journal.pone.0267798.t001

Prediction of genes related to LINC00922

To identify the genes related to LINC00922, the MEM database and TANRIC were used. For the MEM database, 4,360 related genes were identified when the threshold was set to 10−6. For TANRIC, 1,025 related genes were identified. Among these, 336 genes were overlapping (Fig 2A).

thumbnail
Fig 2. Enrichment analysis of genes related to LINC00922 in gastric cancer.

(A) Venn diagram of the overlapping genes predicted by the MEM database and TANRIC. (B) KEGG pathway enrichment analysis by DAVID. (C) Protein-protein interaction network of genes related to LINC00922. (D) Top ten hub genes of LINC00922.

https://doi.org/10.1371/journal.pone.0267798.g002

GO and KEGG analyses

We used Database for Annotation, Visualization and Integrated Discovery (DAVID) to conduct GO and KEGG analyses. For the GO database, there are three categories of functional annotations, namely, molecular function (MF), cellular component (CC) and biological process (BP). As shown in Table 2, there were 10, 10 and 3 significant terms in BP, CC and MF, respectively (false discovery rate (FDR) < 0.05). For MF, cell adhesion, biological adhesion and cell motion were mostly enriched. For CC, plasma membrane, actin cytoskeleton and contractile fiber part were mostly enriched. For BP, actin binding, cytoskeletal protein binding and growth factor binding were mostly enriched. In the KEGG pathways, there were 11 significant terms (P < 0.05) (Table 3 and Fig 2B). Among these, the TGF-β signaling pathway, dilated cardiomyopathy and focal adhesion were the top three terms.

thumbnail
Table 2. The GO analysis of predicted target genes of LINC00922.

https://doi.org/10.1371/journal.pone.0267798.t002

thumbnail
Table 3. Pathway analysis of the predicted target genes of LINC00922.

https://doi.org/10.1371/journal.pone.0267798.t003

Construction and analysis of protein-protein interaction network

As shown in Fig 2C, the protein-protein interaction network contained 335 nodes and 167 edges, and the degree of connectivity was analyzed by Cytoscape using these features. The genes with more than three degrees were defined as hub genes. As shown in Fig 2D, LAMC1, LTBP1, IGFBP4, IGF1, PENK, SPARCL1, PRSS23, FSTL3, PAK3 and ADRB2 were the top ten hub genes. Next, we examined the expression levels and prognostic values of the most significant hub genes in GEPIA. The results indicated FSTL3 was highly expressed in GC tissues, and correlated with overall survival and disease-free survival (Fig 3A–3C), while LAMC1 expression was increased in GC tissues (Fig 3D), but not correlated with overall survival and disease-free survival of GC patients (Fig 3E and 3F). Thus, FSTL3 and LAMC1 are the likely targets of LINC00922.

thumbnail
Fig 3. Expression level, overall survival and disease-free survival of FSTL3 and LAMC1 in GC patients from GEPIA.

(A) Expression level of FSTL3. (B) Overall survival of FSTL3. (C) Disease-free survival of FSTL3. (D) Expression level of LAMC1. (E) Overall survival of LAMC1. (F) Disease-free survival of LAMC1.

https://doi.org/10.1371/journal.pone.0267798.g003

LINC00922 is essential for GC cell proliferation

To examine the function of LINC00922, we assessed the expression of LINC00922 in GC cell lines HGC27, SGC7901 and AGS as well as the normal gastric cell line GES-1. As shown in Fig 4A, all three GC cell lines expressed a high level of LINC00922, and the HGC27 cell line expressed the highest level of expression. Next, we silenced LINC00922 in HGC27 cells (Fig 4B). To assess cell proliferation, the MTT assay was used, and the results showed that LINC00922 knockdown could significantly affect cell viability in the ShRNA group (P < 0.05) (Fig 4C). Furthermore, decreased LINC00922 expression could increase HCG27 cell apoptosis (P < 0.05) (Fig 4D). These findings indicate that LINC00922 is important in cell proliferation.

thumbnail
Fig 4. Expression of LINC00922 and GC cell proliferation and apoptosis.

(A) Expression of LINC00922 in different GC cell lines, as assessed by RT-PCR. (B) LINC00922 expression in HGC27 cells was examined by RT-PCR. LINC00922 expression was decreased in the ShRNA group compared with the control group (P < 0.05). (C) Cell growth curve of control cells and those transfected with shRNAs and NC. (D) Cell apoptosis was determined by flow cytometry. The apoptosis rate was increased in the ShRNA group compared with the control group (P < 0.05). **P < 0.01, and ***P < 0.001.

https://doi.org/10.1371/journal.pone.0267798.g004

LINC00922 associates with gastric cancer cell invasion and migration

The Transwell assay was performed to assess cell invasion. As shown in Fig 5A and 5C, decreased LINC00922 expression could inhibit cell invasion. Next, the wound-healing assay was used to evaluate the effects of LINC00922 on cell migration. As shown in Fig 5B and 5D, LINC00922 knockdown could significantly decrease cell migration. These results reveal that LINC00922 is an important regulator of cell invasion and migration.

thumbnail
Fig 5. Effects of LINC00922 knockdown on HCG27 cell invasion.

(A) Representative images of the Transwell assay of control cells and those transfected with shRNAs and NC. (B) Representative images of the wound-healing assay showing that cell migration was inhibited in cells transfected with shRNAs. (C) Quantification of Transwell assays in control cells and those transfected with shRNAs and NC. (D) Quantification of wound-healing assays in control cells and those transfected with shRNAs and NC. ***P < 0.001.

https://doi.org/10.1371/journal.pone.0267798.g005

Discussion

According to The National Central Cancer Registry of China, the mortality rate of patients with GC is the second highest among all cancers in China [17]. Although advances have been made in the treatment of GC, such as surgery, chemotherapy and radiotherapy, the prognosis of GC patients still remains poor. Among the prognostic and therapeutic biomarkers, HER2 is the only biomarker screened in clinical settings. However, HER2 is overexpressed in only 9%–38% of GC patients [18]. Thus, a better understanding of GC will lead to the identification of new biomarkers.

Our results indicated that LINC00922 was overexpressed in GC tissues. Recently, several studies have demonstrated the association of lncRNAs with a variety of cancers. For example, the increased expression of lncRNA HOXA11-AS has been reported to associate with cell cycle progression, invasion and migration in GC [19]. Another lncRNA MALAT1, regulates VE-cadherin/β-catenin, ERK/MMP and FAK/paxillin pathways to facilitate angiogenesis and promote metastasis [20]. Here, we demonstrated that LINC00922 was overexpressed in GC tissues and correlated with poor prognosis in GC patients. These findings are consistent with those of other studies, which reported several lncRNAs, such as PEG10, MT1JP and BLACAT1, to associate with cancer patient survival [2123].

We identified 366 genes related to LINC00922 by the MEM database and TANRIC. By GO and KEGG analyses, adhesion-related processes, such as cell and biological adhesion, were significantly enriched. Cell migration was also related to LINC00922, which was confirmed by in vitro experiments that showed LINC00922 to be required for cell invasion and migration. To date, several studies have revealed that lncRNAs play critical roles in metastasis. For example, Sun et al. demonstrated that YAP1 is highly up-regulated in GC, accelerates tumor growth and metastasis through ERK1/2 phosphorylation and regulates lncRNAs, including HOTAIR, MALAT1, LATS2 and LATS2-AS1-001 [24]. HOTAIR and LINC00261 can regulate the epithelial-mesenchymal transition by modulating the expression of E-cadherin, N-cadherin and vimentin [25,26].

It is well established that an imbalance between cell proliferation and survival is critical for cancer progression. For example, H19 is up-regulated in GC and suppresses the expression of p53 and p53 target B-cell lymphoma-associated X protein, which induce GC cell proliferation [27]. H19 also regulates GC cell proliferation by acting as a competing endogenous RNA of miR-141 and competing for binding to its target genes, including insulin-like growth factor (IGF) 1, IGF receptor 2 and zinc finger E-box-binding homeobox 1 [28]. Gastric carcinoma high expressed transcript (GHET1) interacts with IGF2 mRNA binding protein 1 (IGF2BP1), which enhances the interaction between IGF2BP1 and c-Myc and increases the expression of c-Myc, thereby promoting cell proliferation [29]. Our findings demonstrate that LINC00922 inhibits cell apoptosis and promotes cell proliferation, indicating that the expression of LINC00922 might disrupt several cancer-related processes and signaling pathways, and contribute to GC.

We investigated the prognostic significance of LINC00922 in GC using GEPIA, and the results revealed that increased LINC00922 expression correlated with shorter overall survival and disease-free survival. In addition, we obtained the predicted hub genes, FSTL3 and LAMC1, of LINC00922 using STRING and Cytoscape. FSTL3 is a member of the follistatin family, which binds to and inactivates activin to regulate cell growth and differentiation [30]. FSTL3 has been reported to participate in tumorigenesis and associate with nuclear grade and tumor size in breast cancer. LAMC1 encodes laminin γ1, which is involved in a variety of processes such as normal tissue development, cancer cell invasion and metastasis [31,32]. In hepatocellular carcinoma (HCC), LAMC1 competes for miR-124 binding and acts as a trans-regulator to induce the expression of CD151 [33]. Zhang et al. reported that overexpression of LAMC1 in HCC enhances invasion and migration and predicts poor prognosis [34]. Taken collectively, these findings suggest that FSTL3 and LAMC1 are involved in tumor biology and targeting LINC00922 may be an effective therapy in GC patients.

There were several limitations in this study. First, lncRNAs play a variety of roles in GC, many of which were not examined here. As such, the functions of LINC00922, such as the ceRNA mechanism, should be further explored and combined with bioinformatics approaches and in vitro and in vivo experiments [3539]. Second, a larger cohort and a computational model are needed to assess the potential of LINC00922 as a biomarker for GC [4042]. Despite these limitations, our results indicate that LINC00922 is critical in the progression, invasion and migration of GC cells and can be used as a potential target for the treatment of GC.

Materials and methods

Data collection

GC tissue and non-tumor tissue gene expression profiles were obtained from TCGA dataset (https://portal.gdc.cancer.gov/). To normalize the expression levels of lncRNAs in different samples, the data were log2-scaled. Differential expression analysis of lncRNAs was performed with the Limma package (http://www.bioconductor.org/). P-value < 0.05 and fold change (FC) > 1 were set as the threshold. Among the differentially expressed lncRNAs, LINC00922 was selected for further study.

GEPIA dataset analysis

GEPIA is an online web server that includes 9,736 tumor tissue and 8,587 normal samples tissue specimens in TCGA and genotype-tissue expression datasets. Differential gene expression analysis and Kaplan-Meier analysis of overall survival and disease free survival were conducted by GEPIA.

Genes potentially related to LINC00922 in gastric cancer

Online bioinformatics databases MEM and TANRIC were used to predict the genes potentially related to LINC00922 [43,44]. In this study, overlapping genes from the aforementioned programs were recorded as target genes of LINC00922 in GC.

Analysis of associated gene-enriched pathways and their functions

DAVID (https://david.ncifcrf.gov/) was used to conduct GO and KEGG analyses [45]. The results were considered to be significant when the FDR was < 0.05 in GO analysis and the P-value was < 0.05 in KEGG analysis.

Protein-protein interaction network construction and confirmation of hub genes

To identify the interactions between overlapping genes, STRING 11.0 (http://www.string-db.org) was used to construct the protein-protein interaction network [46]. Genes with combined scores > 0.9 were obtained. The degree of connectivity in the protein-protein interaction network was analyzed with Cytoscape software 3.0, and the hub gene network of LINC00922 was constructed. In the protein-protein interaction network, each node indicated a protein, and each color corresponded to a cluster. The edges represented the functional associations of prediction, and the thickness of lines represented the strength of evidence.

Cell culture and transfection

HGC27, SGC7901 and AGC cell lines were obtained from the Cancer Hospital of the Chinese Academy of Medical Sciences (China). The GES-1 cell line was obtained from the American Type Culture Collection (USA). The cells were cultured in Roswell Park Memorial Institute (RPMI) medium supplemented with 10% fetal bovine serum (FBS) (Gibco, USA) and 100 μg/ml streptomycin (Invitrogen, USA), and the cells were cultured in conditions of 5% CO2 at 37˚C.

The LINC00922 sequence was obtained from GenBank, and shRNAs against LINC00922 were synthesized with the following sequences: shRNA-1 (5’-CCTGCACCTACAGATCTACACCTCGAGGTGTAGATCTGTAGGTGCAGG-3’), shRNA-2 (5’-TGCAGGAAGTGTTCATCTAAGCTCGAGCTTAGATGAACACTTCCTGCA-3’). ShRNAs against LINC00922 or the negative control were transfected into the pSICOR-GFP plasmid (Shanghai Geneline Bioscience, China). Flow cytometry was used to confirm knockdown. The cells with the highest knockdown efficiency were used in subsequent experiments.

Real-time polymerase chain reaction (RT-PCR)

Total RNA was isolated from cells using TRIzol reagent (Invitrogen, USA) according to the manufacturer’s instructions, and cDNA was synthesized. Next, the mRNA transcripts were amplified in the Eppendorf PCR system (China). The relative expressions of each target gene was normalized to that of GADPH. The primer sequences were as follows: LINC00922 (5’-CAAGACCGCCAAGCATTAAGAT-3’ [forward] and 5’-AGTGGCTCCTCCACCACCTCCT-3’ [reverse]) and GADPH (5’-GTCAACGGATTTGGTCTGTATT-3’ [forward] and, 5’-AGTCTTCTGGGTGGCAGTGAT-3’ [reverse]). The 2-ΔΔcq method was used to calculate the relative expression level of LINC00922.

MTT assay and apoptosis analysis

For the MTT assay, cells (1 × 103 cells/well) were seeded in 96-well plates and transfected with shRNAs or controls, and cell proliferation was measured every 1 to 4 days. MTT (20 μl; stock concentration, 5 mg/ml) (Solarbio, China) was added to each well, and the plate was incubated at 37˚C for 4 h. The absorbance at 540nm was measured. For the apoptosis assay, cells were harvested and stained with Annexin V-FITC/PI and analyzed with an Accuri C6 flow cytometer (Becton-Dickinson, USA) and Flow Jo 7.6.1 software (Tree Star Inc., USA).

Transwell assay and wound-healing assay

The invasion assay was performed using Transwell chambers (Corning, USA). In brief, 1 × 105 cells were added into the upper chamber, and the lower chamber was filled with medium supplemented with FBS (10%, 600 μl), which served as the chemo-attractant, and the plates were incubated for 24h at 37˚C. Next, the cells on the lower membrane were fixed with 100% methanol and stained with 0.1% crystal violet. Five fields were randomly selected and viewed under 200× magnification. The number of cells was counted. For the wound-healing assay, cells (1 × 103) were added into 6-well plates and cultured until confluence. Next, a 200-μL pipette was used to create a wound. The cells were imaged after 24 hours.

Statistical analysis

GraphPad Prism 6 (USA) was used for statistical analysis. The data were presented as mean ± standard deviation (SD). Student’s t-test was used for comparisons between two groups, and one-way ANOVA was used for comparisons among three or more groups. All experiments were repeated three independent times. P-vales < 0.05 were statistically significant.

References

  1. 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. http://doi.org/10.3322/caac.21492 pmid:30207593.
  2. 2. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32. http://doi.org/10.3322/caac.21338 pmid:26808342.
  3. 3. Kelley JR, Duggan JM. Gastric cancer epidemiology and risk factors. J CLIN EPIDEMIOL. 2003;56(1):1–9. pmid:12589864.
  4. 4. Fang X, Wei J, He X, An P, Wang H, Jiang L, et al. Landscape of dietary factors associated with risk of gastric cancer: A systematic review and dose-response meta-analysis of prospective cohort studies. EUR J CANCER. 2015;51(18):2820–32. http://doi.org/10.1016/j.ejca.2015.09.010 pmid:26589974.
  5. 5. Rota M, Pelucchi C, Bertuccio P, Matsuo K, Zhang ZF, Ito H, et al. Alcohol consumption and gastric cancer risk-A pooled analysis within the StoP project consortium. INT J CANCER. 2017;141(10):1950–62. http://doi.org/10.1002/ijc.30891 pmid:28718913.
  6. 6. Salt Tsugane S., salted food intake, and risk of gastric cancer: epidemiologic evidence. CANCER SCI. 2005;96(1):1–6. http://doi.org/10.1111/j.1349-7006.2005.00006.x pmid:15649247.
  7. 7. Plummer M, Franceschi S, Vignat J, Forman D, de Martel C. Global burden of gastric cancer attributable to Helicobacter pylori. INT J CANCER. 2015;136(2):487–90. http://doi.org/10.1002/ijc.28999 pmid:24889903.
  8. 8. Fock KM, Talley N, Moayyedi P, Hunt R, Azuma T, Sugano K, et al. Asia-Pacific consensus guidelines on gastric cancer prevention. J Gastroenterol Hepatol. 2008;23(3):351–65. http://doi.org/10.1111/j.1440-1746.2008.05314.x pmid:18318820.
  9. 9. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. CELL. 2009;136(4):629–41. http://doi.org/10.1016/j.cell.2009.02.006 pmid:19239885.
  10. 10. Engreitz JM, Ollikainen N, Guttman M. Long non-coding RNAs: spatial amplifiers that control nuclear structure and gene expression. Nat Rev Mol Cell Biol. 2016;17(12):756–70. http://doi.org/10.1038/nrm.2016.126 pmid:27780979.
  11. 11. Li D, Liu X, Zhou J, Hu J, Zhang D, Liu J, et al. Long noncoding RNA HULC modulates the phosphorylation of YB-1 through serving as a scaffold of extracellular signal-regulated kinase and YB-1 to enhance hepatocarcinogenesis. HEPATOLOGY. 2017;65(5):1612–27. http://doi.org/10.1002/hep.29010 pmid:28027578.
  12. 12. 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(2):159–66. http://doi.org/10.1016/j.canlet.2013.06.013 pmid:23791884.
  13. 13. Gibb EA, Brown CJ, Lam WL. The functional role of long non-coding RNA in human carcinomas. MOL CANCER. 2011;10:38. http://doi.org/10.1186/1476-4598-10-38 pmid:21489289.
  14. 14. Peng WX, Koirala P, Mo YY. LncRNA-mediated regulation of cell signaling in cancer. ONCOGENE. 2017;36(41):5661–7. http://doi.org/10.1038/onc.2017.184 pmid:28604750.
  15. 15. Chen X, Yan CC, Zhang X, You ZH. Long non-coding RNAs and complex diseases: from experimental results to computational models. BRIEF BIOINFORM. 2017;18(4):558–76. http://doi.org/10.1093/bib/bbw060 pmid:27345524.
  16. 16. Chen X, Sun YZ, Guan NN, Qu J, Huang ZA, Zhu ZX, et al. Computational models for lncRNA function prediction and functional similarity calculation. BRIEF FUNCT GENOMICS. 2019;18(1):58–82. http://doi.org/10.1093/bfgp/ely031 pmid:30247501.
  17. 17. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32. http://doi.org/10.3322/caac.21338 pmid:26808342.
  18. 18. Baniak N, Senger JL, Ahmed S, Kanthan SC, Kanthan R. Gastric biomarkers: a global review. WORLD J SURG ONCOL. 2016;14(1):212. http://doi.org/10.1186/s12957-016-0969-3 pmid:27514667.
  19. 19. Liu Z, Chen Z, Fan R, Jiang B, Chen X, Chen Q, et al. Over-expressed long noncoding RNA HOXA11-AS promotes cell cycle progression and metastasis in gastric cancer. MOL CANCER. 2017;16(1):82. http://doi.org/10.1186/s12943-017-0651-6 pmid:28441948.
  20. 20. Li Y, Wu Z, Yuan J, Sun L, Lin L, Huang N, et al. Long non-coding RNA MALAT1 promotes gastric cancer tumorigenicity and metastasis by regulating vasculogenic mimicry and angiogenesis. CANCER LETT. 2017;395:31–44. http://doi.org/10.1016/j.canlet.2017.02.035 pmid:28268166.
  21. 21. Ge H, Yan Y, Wu D, Huang Y, Tian F. Prognostic value of PEG10 in Asian solid tumors: A meta-analysis. CLIN CHIM ACTA. 2018;483:197–203. http://doi.org/10.1016/j.cca.2018.04.041 pmid:29727698.
  22. 22. Zhu C, Ma J, Li Y, Zhang Y, Da M. Low expression of long noncoding RNA MT1JP is associated with poor overall survival in gastric cancer patients: Protocol for meta-analysis. Medicine (Baltimore). 2018;97(21):e10394. http://doi.org/10.1097/MD.0000000000010394 pmid:29794726.
  23. 23. Lu H, Liu H, Yang X, Ye T, Lv P, Wu X, et al. LncRNA BLACAT1 May Serve as a Prognostic Predictor in Cancer: Evidence from a Meta-Analysis. BIOMED RES INT. 2019;2019:1275491. http://doi.org/10.1155/2019/1275491 pmid:31061820.
  24. 24. Sun D, Li X, He Y, Li W, Wang Y, Wang H, et al. YAP1 enhances cell proliferation, migration, and invasion of gastric cancer in vitro and in vivo. Oncotarget. 2016;7(49):81062–76. http://doi.org/10.18632/oncotarget.13188 pmid:27835600.
  25. 25. Liu YW, Sun M, Xia R, Zhang EB, Liu XH, Zhang ZH, et al. 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. http://doi.org/10.1038/cddis.2015.150 pmid:26136075.
  26. 26. Fan Y, Wang YF, Su HF, Fang N, Zou C, Li WF, et al. Decreased expression of the long noncoding RNA LINC00261 indicate poor prognosis in gastric cancer and suppress gastric cancer metastasis by affecting the epithelial-mesenchymal transition. J HEMATOL ONCOL. 2016;9(1):57. http://doi.org/10.1186/s13045-016-0288-8 pmid:27439973.
  27. 27. Yang F, Bi J, Xue X, Zheng L, Zhi K, Hua J, et al. Up-regulated long non-coding RNA H19 contributes to proliferation of gastric cancer cells. FEBS J. 2012;279(17):3159–65. http://doi.org/10.1111/j.1742-4658.2012.08694.x pmid:22776265.
  28. 28. 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(4):1440–52. http://doi.org/10.1159/000430309 pmid:26160158.
  29. 29. Yang F, Xue X, Zheng L, Bi J, Zhou Y, Zhi K, et al. Long non-coding RNA GHET1 promotes gastric carcinoma cell proliferation by increasing c-Myc mRNA stability. FEBS J. 2014;281(3):802–13. http://doi.org/10.1111/febs.12625 pmid:24397586.
  30. 30. Wankell M, Kaesler S, Zhang YQ, Florence C, Werner S, Duan R. The activin binding proteins follistatin and follistatin-related protein are differentially regulated in vitro and during cutaneous wound repair. J ENDOCRINOL. 2001;171(3):385–95. pmid:11739004.
  31. 31. Givant-Horwitz V, Davidson B, Reich R. Laminin-induced signaling in tumor cells. CANCER LETT. 2005;223(1):1–10. http://doi.org/10.1016/j.canlet.2004.08.030 pmid:15890231.
  32. 32. Patarroyo M, Tryggvason K, Virtanen I. Laminin isoforms in tumor invasion, angiogenesis and metastasis. SEMIN CANCER BIOL. 2002;12(3):197–207. http://doi.org/10.1016/S1044-579X(02)00023-8 pmid:12083850.
  33. 33. Yang ZP, Ma HS, Wang SS, Wang L, Liu T. LAMC1 mRNA promotes malignancy of hepatocellular carcinoma cells by competing for MicroRNA-124 binding with CD151. IUBMB LIFE. 2017;69(8):595–605. http://doi.org/10.1002/iub.1642 pmid:28524360.
  34. 34. Zhang Y, Xi S, Chen J, Zhou D, Gao H, Zhou Z, et al. Overexpression of LAMC1 predicts poor prognosis and enhances tumor cell invasion and migration in hepatocellular carcinoma. J CANCER. 2017;8(15):2992–3000. http://doi.org/10.7150/jca.21038 pmid:28928891.
  35. 35. Zhang L, Liu T, Chen H, Zhao Q, Liu H. Predicting lncRNA-miRNA interactions based on interactome network and graphlet interaction. GENOMICS. 2021;113(3):874–80. http://doi.org/10.1016/j.ygeno.2021.02.002 pmid:33588070.
  36. 36. Zhang L, Yang P, Feng H, Zhao Q, Liu H. Using Network Distance Analysis to Predict lncRNA-miRNA Interactions. INTERDISCIP SCI. 2021;13(3):535–45. http://doi.org/10.1007/s12539-021-00458-z pmid:34232474.
  37. 37. Liu W, Jiang Y, Peng L, Sun X, Gan W, Zhao Q, et al. Inferring Gene Regulatory Networks Using the Improved Markov Blanket Discovery Algorithm. INTERDISCIP SCI. 2021. http://doi.org/10.1007/s12539-021-00478-9 pmid:34495484.
  38. 38. Wang CC, Han CD, Zhao Q, Chen X. Circular RNAs and complex diseases: from experimental results to computational models. BRIEF BIOINFORM. 2021;22(6). http://doi.org/10.1093/bib/bbab286 pmid:34329377.
  39. 39. Liu H, Ren G, Chen H, Liu Q, Yang Y, Zhao Q. Predicting lncRNA–miRNA interactions based on logistic matrix factorization with neighborhood regularized. KNOWL-BASED SYST. 2020;191:105261. http://doi.org/https://doi.org/10.1016/j.knosys.2019.105261.
  40. 40. Chen X, Wang L, Qu J, Guan NN, Li JQ. Predicting miRNA-disease association based on inductive matrix completion. BIOINFORMATICS. 2018;34(24):4256–65. http://doi.org/10.1093/bioinformatics/bty503 pmid:29939227.
  41. 41. Chen X, Xie D, Zhao Q, You ZH. MicroRNAs and complex diseases: from experimental results to computational models. BRIEF BIOINFORM. 2019;20(2):515–39. http://doi.org/10.1093/bib/bbx130 pmid:29045685.
  42. 42. Chen X, Yin J, Qu J, Huang L. MDHGI: Matrix Decomposition and Heterogeneous Graph Inference for miRNA-disease association prediction. PLOS COMPUT BIOL. 2018;14(8):e1006418. http://doi.org/10.1371/journal.pcbi.1006418 pmid:30142158.
  43. 43. Adler P, Kolde R, Kull M, Tkachenko A, Peterson H, Reimand J, et al. Mining for coexpression across hundreds of datasets using novel rank aggregation and visualization methods. GENOME BIOL. 2009;10(12):R139. http://doi.org/10.1186/gb-2009-10-12-r139 pmid:19961599.
  44. 44. Li J, Han L, Roebuck P, Diao L, Liu L, Yuan Y, et al. TANRIC: An Interactive Open Platform to Explore the Function of lncRNAs in Cancer. CANCER RES. 2015;75(18):3728–37. http://doi.org/10.1158/0008-5472.CAN-15-0273 pmid:26208906.
  45. 45. Dennis GJ, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery. GENOME BIOL. 2003;4(5):P3.pmid:12734009.
  46. 46. Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. NUCLEIC ACIDS RES. 2015;43(Database issue):D447–52. http://doi.org/10.1093/nar/gku1003 pmid:25352553.