Downregulation of long non‑coding RNA GAS5 promotes cell proliferation, migration and invasion in esophageal squamous cell carcinoma

Retraction in: /10.3892/ol.2024.14372

  • Authors:
    • Ke Ke
    • Zhanwen Sun
    • Zhengjun Wang
  • View Affiliations

  • Published online on: May 24, 2018     https://doi.org/10.3892/ol.2018.8797
  • Pages: 1801-1808
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to investigate the potential role of long non‑coding RNA growth arrest‑specific transcript 5 (lncRNA GAS5) in the progression of esophageal squamous cell carcinoma (ESCC) and to reveal its possible regulatory mechanism. The expression of lncRNA GAS5 in ESCC tissues and cell lines was analyzed using reverse transcription‑quantitative polymerase chain reaction and western blot analysis. The overexpression vector pc‑GAS5 and control vector pc‑negative control (NC), containing no GAS5 sequence, were transfected into ESCC cells. The effects of lncRNA GAS5 overexpression on cell proliferation, cell cycle distribution, cell migration and invasion were then analyzed. Besides, the expression levels of ATM‑CHK2 pathway‑associated proteins and epithelial‑mesenchymal transition (EMT)‑associated proteins were measured. Expression of lncRNA GAS5 was downregulated in the ESCC tissues compared with adjacent normal tissues, and was also downregulated in ESCC Kyse450 cells compared with the human esophageal epithelial HET‑1A cell line. Additionally, lncRNA GAS5 was successfully overexpressed in ESCC cells following transfection with pc‑GAS5. Overexpression of lncRNA GAS5 significantly inhibited cell proliferation, induced cell cycle arrest at G2/M phase and suppressed cellular migration and invasion. When cells were transfected with pc‑GAS5, the levels of phosphorylated (p)‑ATM serine/threonine protein kinase, p‑checkpoint kinase 2 (CHK2), p‑cell division cycle 25C, p‑cyclin‑dependent kinase 1, N‑cadherin, vimentin and Snail were significantly increased, whereas that of E‑cadherin were markedly decreased. The results of the present study indicate that overexpression of lncRNA GAS5 may inhibit cell proliferation, migration and invasion in ESCC. lncRNA GAS5 overexpression may induce cell cycle arrest at G2/M stage by activating the ATM‑CHK2 pathway. The results of the current study further indicate that lncRNA GAS5 overexpression may suppress cell migration and invasion via EMT‑associated proteins. lncRNA GAS5 could therefore serve as a potential target for ESCC therapy.

Introduction

Human esophageal cancer is a common malignant tumor with a high mortality rate (>300,000 deaths every year) globally (1,2), with >90% of cases made up of esophageal squamous cell carcinoma (ESCC) (3). The high recurrence rate following surgery is one of the main reasons for the poor prognosis of patients with ESCC (4). However, to the best of our knowledge, the genetic mechanisms underlying the tumorigenesis and poor prognosis of ESCC are largely unknown (5). Therefore, elucidation of the key molecular mechanisms underlying ESCC will aid the development of novel therapeutic approaches.

Long non-coding RNAs (lncRNAs) are mRNA-like transcripts ranging in length from 200 to 100,000 nt, but they do not function as templates for protein synthesis owing to a lack of open-reading frames (6,7). Several lncRNAs have been identified as key players to serve oncogenic or tumor suppressive roles in a variety of cancer types (6,7), including ESCC (8). For instance, upregulation of lncRNA nuclear enriched abundant transcript 1 is associated with the progression and poor prognosis of ESCC (9). Upregulation of lncRNA H19 is also able to promote cellular proliferation and metastasis in ESCC (10). Knockdown of lncRNA P73 antisense RNA 1T has also been shown to inhibit cell proliferation and induce apoptosis in ESCC (11).

Growth arrest-specific transcript 5 (GAS5) was originally identified as a potential tumor suppressor genes involved in arresting cellular growth (12). However, the putative open-reading frame GAS5 is small and poorly conserved, indicating that the biological activity of GAS5 is mediated through the introns (13). Evidence indicates that GAS5 is a tumor-suppressor lncRNA, regulating cellular apoptosis in prostate cancer (14). Downregulation of lncRNA GAS5 can promote cellular proliferation and invasion in hepatocellular carcinoma (15). GAS5 overexpression inhibits cell growth and induces apoptosis in non-small-cell lung cancer cells, indicating that GAS5 has the potential to serve as a diagnostic marker for non-small cell lung cancer (16). Despite these findings, the roles and regulatory mechanism of lncRNA GAS5 in ESCC remain uncertain.

The present study investigated whether lncRNA GAS5 was dysregulated in ESCC tissues and cells. Next, the effects of lncRNA GAS5 overexpression on ESCC cellular proliferation, cell cycle arrest and cell migration and invasion were measured. In addition, the expression levels of ataxia telangiectasia-mutated (ATM)-checkpoint kinase 2 (CHK2) pathway-associated proteins and epithelial-mesenchymal transition (EMT)-associated proteins were assessed. The present study aimed to investigate the potential roles of lncRNA GAS5 in the ESCC development and to elucidate its possible regulatory mechanisms.

Materials and methods

Patient collections

A total of 36 ESCC patients admitted to the Department of Cardiothoracic Surgery of the Affiliated Hospital of Hubei Polytechnic University (Huangshi, China) between March 2013 and January 2016 were included in the present study. The diagnosis of ESCC was pathologically confirmed. Cancer tissues and their adjacent normal tissues were obtained from clinically resected surgical specimens. All the specimens were snap-frozen in liquid nitrogen and stored at −80°C until use. This study was approved by the Affiliated Hospital of Hubei Polytechnic University Protection of Human Ethics Committee, and all patients were informed with consent prior to inclusion in the current study.

Cell culture

The esophageal carcinoma Kyse450 cell line and human esophageal epithelial cell line HET-1A were purchased from the American Type Culture Collection (Manassas, VA, USA). All these cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and maintained at 37°C in a humidified atmosphere with 5% CO2. After 48 h cultivation, cells were collected for the consequent transfection experiment.

Cell transfection

Oligonucleotide primers containing BamHI or HindIII site were synthesized for amplification of coding sequence of GAS5 (Accession No. AF_314752). The two primers were: Forward, 5′-CGCGGATCCGTGCTGGGTGCAGATGCAGTGTGgc-3′ and reverse, 5′-CCGCTCGAGTTTTTTTTTTTTTTTTTTTTTTT-3′. The amplified GAS5 sequence was then cloned into pcDNA3.0 (Sangon Biotech Co., Ltd., Shanghai, China) via BamHI and HindIII sites to construct the overexpression vector pcDNA3.0-GAS5 (pc-GAS5), which was confirmed by sequencing analysis by Sangong Biotech Co., Ltd. The empty vector pc-negative control (NC) with no GAS5 sequence was used as a negative control. Next, pc-GAS5 (50 nM) or pc-NC (50 nM) were transfected into esophageal carcinoma Kyse450 cells using Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in accordance with the instructions of manufacturer.

MTT assay

Cell proliferation was measured using an MTT assay. Briefly, cells (5×103 cells per well) in the logarithmic growth phase were seeded in triplicate in 96-well culture plate. Following incubation for 24 h, the supernatant was removed by centrifugation at 6,000 × g for 5 min at 4°C. Next, 20 µl 10 mg/ml MTT was added to each well and incubated at 37°C for 4 h at different time points (24, 36, 48, 72 and 96 h) following transfection. Following this, 150 µl dimethyl sulfoxide was added into each well for 10 min to sufficiently solubilize the formazan crystals. The optical density at 570 nm was measured using a microplate reader (Synergy4; BioTek Instruments, Inc., Winooski, VT, USA). The cell viability of each well=[optical density (OD) value of test group-OD value of blank group]/(OD value of control group-OD value of blank group).

Colony formation assay

Cell proliferation was also assessed using a clongenic assay with a modification of previously described method. Briefly, at 96 h after transfection, cells (100 cells per dish) in the logarithmic growth phase were plated into 60-mm culture dishes containing RPMI-1640 medium supplemented with 10% FBS. After 14 days, colonies were fixed with ethanol at 4°C for 10 min and stained with 0.1% crystal violet (Sigma-Aldrich, Merck KGaA, Darmstadt, Germany) for 30 min at room temperature. Finally, colonies were counted under a light microscope (magnification, ×400). The cell number each colony was at least 30 cells. Each experiment was performed in triplicate.

Cell-cycle assay

The cell cycle distribution following transfection was detected by flow cytometry. Briefly, at 96 h after transfection, the transfected cells were cultured in RPMI-1640 with 10% FBS. Cells were then suspended with cold PBS buffer at a volume ratio of 2.5:1 and fixed with methanol at 4°C for 30 min. Next, Cells were stained at room temperature with propidium iodide (PI; Sangong Biotech, Co., Ltd.) solution for 30 min. Finally, the cell cycle distribution and DNA content were analyzed using flow cytometry (FACSDiva software, version 6.0; BD Biosciences, San Jose, CA, USA).

Transwell assay

Cell invasion and migration were measured using Transwell assay as previously described. The Transwell chamber (8-µm pore size; Corning Incorporated, Corning, NY, USA) were uncoated for the migration assays or coated with Matrigel (BD Biosciences) for the invasion assays. Briefly, 96 h after transfection with pc-GAS5 and pc-NC, the transfected cells (1×105) cultured in serum-free RPMI 1640 medium were added into the upper layer of Transwell chamber for 30 min at 37°C. RPMI 1640 medium mixed with 10% FBS (as a chemoattractant) was added into the lower layer of the transwell chamber. After 48 h of incubation, the lower transwell chamber in each group was washed with PBS buffer 3 times, fixed with methanol at 4°C for 10 min, and stained with 0.1% Giemsa for 30 min at room temperature. Finally, the migrated and invaded cells were counted from 10 random fields under a light microscope (magnification, ×400). Each experiment was performed in triplicate.

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Total RNA was isolated from tissues and the cells at 96 h after transfection using TRIzol Reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) following the manufacturer's instructions. Following treatment with RNse-free Dnase I (Promega Corporation, Madison, WI, USA) and quantitation with SMA 400 UV-VIS (Merinton Instrument, Ltd., Shanghai, China), 0.5 µg/µl purified RNA was used for cDNA synthesis using the PrimerScript 1st Strand cDNA Synthesis kit (Invitrogen; Thermo Fisher Scientific, Inc.). qPCR was then performed using the SYBR ExScript RT-qPCR kit (Takara Biotechnology Co., Ltd., Dailan, China) to detect the expression of targets. The PCR reaction was pre-incubated at 95°C for 10 min, incubated at 95°C for 30 sec followed by 40 cycles of 95°C for 15 sec, 60°C for 1 min and then extension at 72°C for 5 min. Each sample was analyzed in triplicate. Phosphoglyceraldehyde dehydrogenase (GAPDH) was used as an endogenous control to normalize the data and the 2−ΔΔCq method (17) was used to calculate the relative expressions of targets. Primers used for the amplification of targets are shown in Table I.

Table I.

Primers used for targets amplification.

Table I.

Primers used for targets amplification.

Accession no.NameForward, 5′-3′Reverse, 5′-3′Length, bp
NR_002578.2GAS5 GAGAGTGGTGTGGGGAACTG CAGAGGTCCCACTGCATGTT651
Pr032301916E-cadherin AACGCATTGCCACATACAC AACGCATTGCCACATACAC144
Pr032246378N-cadherin CATCCCTCCAATCAACTTGCATGTGC CCTCAAATGAAACC  71
Pr032475850Vimentin TCCAAGTTGCTGACCTCTC TCAACGGCAAAGTTCTCTTC206
Pr032301930Snail TTCAACTGCAAATACTGCAACAAG CGTGTGGCTTCGGATGTG131
Pr032754117GAPDH TTGTCAAGCTCGTTTCTTGGT CCTAGTCTCCATGGTCTCACT202

[i] GAS5, growth arrest-specific transcript 5.

Western blot assay

At 96 h after transfection with pc-GAS5 and pc-NC, cells were incubated in radioimmunoprecipitation assay buffer (Sangon Biotech Co., Ltd., Shanghai, China) containing phenylmethanesufonyl fluoride on ice for 20 min and mixed 2-3 times. The supernatant was collected and then quantitated using a bicinchoninic acid (BCA) assay. Next, a total of 20 µg protein per lane was subjected to 10% SDS-PAGE and then transferred onto a polyvinylidene fluoride membrane. Following blocking in PBST (0.1% Triton X-100 in PBS), the membranes were incubated with primary antibodies against ATM serine/threonine protein kinase (hereafter ATM; cat. no. ab95037), phosphorylated (p)-ATM (cat. no. ab95037), checkpoint kinase 2 (CHK2; cat. no. ab26338), p-CHK2 (cat. no. ab26338), cell division cycle 25C (CDC25C; cat. no. ab32444), p-CDC25C (cat. no. ab32444), cyclin-dependent kinase 1 (CDK1; cat. no. ab133327), p-CDK1 (cat. no. ab133327), E-cadherin (cat. no. ab197751), N-cadherin (cat. no. ab76057), vimentin (cat. no. ab45939) and Snail (cat. no. ab53519; 1:1,000 dilution for all primary antibodies; Abcam, Cambridge, UK) overnight at 4°C, and subsequently probed with horseradish peroxidase-labeled secondary antibody (1:1,000 dilution; cat. no. ab6721; Abcam) at room temperature for 1 h. Finally, the bands were visualized using an enhanced chemiluminescence (CEL; Thermo Fisher Scientific, Inc.). GAPDH (1:5,000 dilution; cat. no. ab8245) was considered as the internal control. All primary antibodies were purchased from Abcam. To further quantify the results, the protein blots were scanned and analyzed using ImageQuant software (version TL 7.0; Molecular Dynamics, LLC, Sunnyvale, CA, USA).

Statistical analysis

All statistical analyses in this study were performed using SPSS 19.0 (IBM Corp, Armonk, NY, USA). All experiments in the present study were repeated at least 3 times, and data collected from 3 independent experiments were presented as the mean ± standard deviation. Two-tailed Student's t-test was used to analyze the difference in GAS5 expression between cancer tissues and their adjacent normal tissues. Pearson's correlation coefficient analysis was used to calculate the correlation between the expression of GAS5 and p-ATM or p-CHK2 in cancer tissues. One-way analysis of variance was used to detect the significant differences in the effects of GAS5 on cell proliferation, migration and invasion in differently transfected groups. A threshold of P<0.05 was considered to indicate a statistically significant difference.

Results

LncRNA GAS5 is downregulated in ESCC tissues and cells

The present study investigated the expression of lncRNA GAS5 in ESCC tissues and cells by RT-qPCR analysis. As shown in Fig. 1A, GAS5 expression was significantly downregulated in ESCC tissues compared with the adjacent normal tissues (P<0.01). In addition, the expression of GAS5 in esophageal carcinoma cell line Kyse450 was significantly lower than that in the normal esophageal epithelial cell line HET-1A (P<0.01; Fig. 1B).

Overexpression of lncRNA GAS5 inhibits cell proliferation

To investigate the role of GAS5 in ESCC, GAS5 was successfully overexpressed in ESCC cells following transfection with pc-GAS5 (P<0.01; Fig. 2A). Subsequently, the effects of GAS5 on cell proliferation were assessed using MTT and colony-formation assays. Similar results were obtained using the two assays, which found that, in comparison with the control group, cell viability and the number of colony of pc-GAS5 group were significantly decreased (P<0.05; Fig. 2B-D), indicating that the overexpression of lncRNA GAS5 significantly inhibited cell proliferation.

Overexpression of lncRNA GAS5 induces cell cycle arrest at G2/M stage by activating the ATM-CHK2 pathway

The effect of lncRNA GAS5 was assessed on the cell cycle using flow cytometry. As presented in Fig. 3A, the percentage of cells in the G0/G1 stage in the pc-GAS5 group was significantly decreased compared with that in the pc-NC group (P<0.05), whereas the percentage of cells in the G2/M stage in the pc-GAS5 group was markedly increased, indicating that overexpression of lncRNA GAS5 may induce cell cycle arrest at G2/M stage.

To investigate the possible mechanism of lncRNA GAS5 on cell-cycle arrest at G2/M stage, the expression levels of ATM-CHK2 pathway-associated proteins were analyzed. As presented in Fig. 3B and C, there were no significant differences in the protein expression levels of ATM, CHK2, CDC25C and CDK1, whereas the levels of p-ATM, p-CHK2, p-CDC25C and p-CDK1 proteins were significantly increased when cells were transfected with pc-GAS5 compared with that in the pc-NC group (P<0.05). In addition, the levels of p-ATM and p-CHK2 in ESCC tissues were found to be significantly lower than that in the adjacent normal tissues (P<0.05; Fig. 3D and E). Pearson's correlation analysis revealed that the levels of p-ATM and p-CHK2 were positive correlated with GAS5 expression in cancer tissues (P<0.001; Fig. 3F and G).

Overexpression of lncRNA GAS5 suppresses cell migration and invasion via inhibiting expression of EMT-associated proteins

The roles of lncRNA GAS5 in regulating cell migration and invasion were further evaluated using transwell migration and invasion assays. As shown in Fig. 4, the number of migrated cells and invaded cells in the pc-GAS5 group were all significantly than those in the pc-NC group (P<0.05).

The expression levels of EMT-associated proteins, including E-cadherin, N-cadherin, vimentin and Snail, were investigated. The results of this analysis revealed that once GAS5 was overexpressed, the expressions of E-cadherin were significantly decreased, whereas the expression levels of N-cadherin, vimentin and Snail were markedly increased (P<0.05; Fig. 5), indicating that overexpression of lncRNA GAS5 may suppress cell migration and invasion by inhibiting EMT-associated proteins.

Discussion

Prior studies identified a large number of lncRNAs that are involved in the cancer progression (68); however, to the best of our knowledge, their function remains largely unknown. The present study focused on the roles of lncRNA GAS5 in the development of ESCC. GAS5 was downregulated in ESCC tissues compared with adjacent normal tissues. Consistently, previous findings have demonstrated that GAS5 and/or its small nucleolar RNAs are downregulated in multiple cancer types, including breast cancer (18), renal cell carcinoma (19) and glioma (20). Additionally, the results of the present study revealed that overexpression of GAS5 significantly inhibited cell proliferation, arrested cell cycle and suppressed cell migration and invasion. Therefore, the observations from the current study indicate that deregulation of GAS5 may serve roles in the progression of ESCC.

It has reported that activating checkpoints, particularly S and G2 phase, can effectively arrest cell proliferation in various types of cancer (21). Loss of ATM-CHK2-tumor protein p53 pathway components has been shown to accelerate tumor development in gliomas (22). Additionally, hepatitis B virus X protein can delay the cell cycle progression in liver cancer by activating the ATM-Chk2 pathway (23). Naphthalimides have been confirmed to induce G2 arrest in HCT116 cells via activating the ATM-CHK2 pathway (24). In the present study, the levels of key proteins involved in ATM-CHK2 pathway, including p-ATM and p-CHK2, were significantly increased in pc-GAS5 transfected cells and ESCC tissues. Pearson's correlation analysis demonstrated that the expression levels of p-ATM and p-CHK2 were positively correlated with GAS5 expression. On the basis of the experimental results of the current study, we hypothesize that GAS5 may induce cell cycle arrest at the G2/M stage in ESCC via activation of the ATM-CHK2 pathway. Furthermore, bendamustine also induces G2 phase cell cycle arrest in myeloma cells via regulating ATM-CHK2-CDC25A pathway (25). Phosphatase and tensin homolog (PTEN) contributes to the DNA damage response and induces G2/M phase arrest in etoposide-treated MCF-7 cells through activation of the ATM-CHK2 pathway (26). It can therefore be speculated that GAS5 could be involved in drug therapy for ESCC via activation of the ATM-CHK2 pathway.

In addition, EMT is shown to be implicated in the migration and invasion in a variety of cancer types (27,28), including ESCC (29). Aberrant expression of E-cadherin is widely involved in the invasion and metastasis in ESCC (30). Certain tumor-associated molecules, such as a disintegrin and metalloprotease 10 (ADAM10), have been shown to mediate cell invasion and metastasis in ESCC by regulating E-cadherin (31). Additionally, Snail has been reported to serve a notable role in E-cadherin-preserved ESCC (32). In addition, the knockdown of N-cadherin in vitro can decrease the invasiveness of ESCC (33). Wang et al (34) demonstrated that N-cadherin contributed to the invasion and metastasis in ESCC by regulating the formation of vasculogenic mimicry. Additionally, vimentin has been reported to drive lymph node metastasis in ESCC in an aggressive manner (35). The results of the current study also verified that the number of migrated cells and invaded cells in pc-GAS5 group were significantly lower that of the pc-NC group. Following overexpression of GAS5, the expression of E-cadherin was significantly decreased, whereas those of N-cadherin, vimentin and Snail were markedly increased. Although the regulatory mechanism of GAS5 in cellular migration and invasion has not been fully investigated, based on the experimental results of the present study we hypothesize that lncRNA GAS5 may regulate cell migration and invasion in ESCC by altering the expression of EMT-associated proteins.

The present study did not consider whether lncRNA GAS5 dysregulation in ESCC was dependent on the stage, invasion, lymph node/distant metastasis, differentiation or any other clinicopathological characteristic of patients, which was a limitation of the present study. If the associations between the expression of lncRNA GAS5 and clinicopathological characteristics of patients are to be analyzed, the findings could provide a broader perspective on the potential applications of lncRNA GAS5 in cancer therapy.

In conclusion, the findings of the present study indicate that the overexpression of lncRNA GAS5 may inhibit cell proliferation, migration and invasion in ESCC. GAS5 may induce cell cycle arrest at G2/M phase by activating the ATM-CHK2 pathway. GAS5 may suppress cellular migration and invasion via EMT-associated proteins. The lncRNA GAS5 could therefore serve as a potential target for the therapy of ESCC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contribution

KK prepared the manuscript and conducted the experiments. ZS designed the present study and prepared the patients samples and cell lines. ZW analyzed the data and the performed statistical analysis.

Ethics approval and consent to participate

This study was approved by the Affiliated Hospital of Hubei Polytechnic University Protection of Human Ethics Committee, and all patients provided informed consent prior to their inclusion in the current study.

Consent for publication

The authors declare that all patients enrolled in this study provided informed consent for this publication.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Holmes RS and Vaughan TL: Epidemiology and pathogenesis of esophageal cancer. Semin Radiat Oncol. 17:2–9. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Enzinger PC and Mayer RJ: Esophageal cancer. N Engl J Med. 349:2241–2252. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Hu D, Zhang M, Wang S and Wang Z: High expression of cyclooxygenase 2 is an indicator of prognosis for patients with esophageal squamous cell carcinoma after Ivor Lewis esophagectomy. Thorac Cancer. 7:310–315. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Qin HD, Liao XY, Chen YB, Huang SY, Xue WQ, Li FF, Ge XS, Liu DQ, Cai Q, Long J, et al: Genomic characterization of esophageal squamous cell carcinoma reveals critical genes underlying tumorigenesis and poor prognosis. Am J Hum Genet. 98:709–727. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Gibb EA, Brown CJ and Wan LL: The functional role of long non-coding RNA in human carcinomas. Mol Cancer. 10:382011. View Article : Google Scholar : PubMed/NCBI

7 

Cheetham SW, Gruhl F, Mattick JS and Dinger ME: Long noncoding RNAs and the genetics of cancer. Br J Cancer. 108:2419–2425. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Yao J, Huang JX, Lin M, Wu ZD, Yu H, Wang PC, Ye J, Chen P, Wu J and Zhao GJ: Microarray expression profile analysis of aberrant long non-coding RNAs in esophageal squamous cell carcinoma. Int J Oncol. 48:2543–2557. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Chen X, Kong J, Ma Z, Gao S and Feng X: Up regulation of the long non-coding RNA NEAT1 promotes esophageal squamous cell carcinoma cell progression and correlates with poor prognosis. Am J Cancer Res. 5:2808–2815. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Tan D, Wu Y, Hu L, He P, Xiong G, Bai Y and Yang K: Long noncoding RNA H19 is up-regulated in esophageal squamous cell carcinoma and promotes cell proliferation and metastasis. Dis Esophagus. 30:1–9. 2017.

11 

Zang W, Wang T, Wang Y, Chen X, Du Y, Sun Q, Li M, Dong Z and Zhao G: Knockdown of long non-coding RNA TP73-AS1 inhibits cell proliferation and induces apoptosis in esophageal squamous cell carcinoma. Oncotarget. 7:19960–19974. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Schneider C, King RM and Philipson L: Genes specifically expressed at growth arrest of mammalian cells. Cell. 54:787–793. 1988. View Article : Google Scholar : PubMed/NCBI

13 

Smith CM and Steitz JA: Classification of gas5 as a multi-small-nucleolar-RNA (snoRNA) host gene and a member of the 5′-terminal oligopyrimidine gene family reveals common features of snoRNA host genes. Mol Cell Biol. 18:6897–6909. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Pickard MR, Mourtada-Maarabouni M and Williams GT: Long non-coding RNA GAS5 regulates apoptosis in prostate cancer cell lines. Biochim Biophys Acta. 1832:1613–1623. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Chang L, Li C, Lan T, Wu L, Yuan Y, Liu Q and Liu Z: Decreased expression of long non-coding RNA GAS5 indicates a poor prognosis and promotes cell proliferation and invasion in hepatocellular carcinoma by regulating vimentin. Mol Med Rep. 13:1541–1550. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Shi X, Sun M, Liu H, Yao Y, Kong R, Chen F and Song Y: A critical role for the long non-coding RNA GAS5 in proliferation and apoptosis in non-small-cell lung cancer. Mol Carcinog. 54 Suppl 1:E1–E12. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F and Williams GT: GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 28:195–208. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Qiao HP, Gao WS, Huo JX and Yang ZS: Long non-coding RNA GAS5 functions as a tumor suppressor in renal cell carcinoma. Asian Pac J Cancer Prev. 14:1077–1082. 2013. View Article : Google Scholar : PubMed/NCBI

20 

García-Claver A, Lorente M, Mur P, Campos-Martín Y, Mollejo M, Velasco G and Meléndez B: Gene expression changes associated with erlotinib response in glioma cell lines. Eur J Cancer. 49:1641–1653. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Chen T, Stephens PA, Middleton FK and Curtin NJ: Targeting the S and G2 checkpoint to treat cancer. Drug Discov Today. 17:194–202. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Squatrito M, Brennan CW, Helmy K, Huse JT, Petrini JH and Holland EC: Loss of ATM/Chk2/p53 pathway components accelerates tumor development and contributes to radiation resistance in gliomas. Cancer Cell. 18:619–629. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Kim S, Lee HS, Ji JH, Cho MY, Yoo YS, Park YY, Cha HJ, Lee Y, Kim Y and Cho H: Hepatitis B virus X protein activates the ATM-Chk2 pathway and delays the cell cycle progression. J Gen Virol. 96:2242–2251. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Zhu H, Miao ZH, Huang M, Feng JM, Zhang ZX, Lu JJ, Cai YJ, Tong LJ, Xu YF, Qian XH and Ding J: Naphthalimides induce G(2) arrest through the ATM-activated Chk2-executed pathway in HCT116 cells. Neoplasia. 11:1226–1234. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Gaul L, Mandl-Weber S, Baumann P, Emmerich B and Schmidmaier R: Bendamustine induces G2 cell cycle arrest and apoptosis in myeloma cells: The role of ATM-Chk2-Cdc25A and ATM-p53-p21-pathways. J Cancer Res Clin Oncol. 134:245–253. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Zhang R, Zhu L, Zhang L, Xu A, Li Z, Xu Y, He P, Wu M, Wei F and Wang C: PTEN enhances G2/M arrest in etoposide-treated MCF-7 cells through activation of the ATM pathway. Oncol Rep. 35:2707–2714. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Yang J and Weinberg RA: Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell. 14:818–829. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Thompson EW, Newgreen DF and Tarin D: Carcinoma invasion and metastasis: A role for epithelial-mesenchymal transition? Cancer Res. 65:5991–5995. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Ju JY, Yu WJ and Zhao CL: PLK1 promotes invasion of esophageal squamous cell carcinoma cells through inducing epithelial-mesenchymal transition. Adv Mater Res. 998-999:279–282. 2014. View Article : Google Scholar

30 

Ping FM, Liu GJ, Liu ZJ, Li HB, Zhai JW, Li SX, Liu YM, Li BW and Wei H: Expression of RKIP, E-cadherin and NF-kB p65 in esophageal squamous cell carcinoma and their correlations. Int J Clin Exp Pathol. 8:10164–10170. 2014.

31 

Ma B, Zhang HY, Bai X, Wang F, Ren XH, Zhang L and Zhang MZ: ADAM10 mediates the cell invasion and metastasis of human esophageal squamous cell carcinoma via regulation of E-cadherin activity. Oncol Rep. 35:2785–2794. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Natsugoe S, Uchikado Y, Okumura H, Matsumoto M, Setoyama T, Tamotsu K, Kita Y, Sakamoto A, Owaki T, Ishigami S and Aikou T: Snail plays a key role in E-cadherin-preserved esophageal squamous cell carcinoma. Oncol Rep. 17:517–523. 2007.PubMed/NCBI

33 

Li K, He W, Lin N, Wang X and Fan QX: N-cadherin knock-down decreases invasiveness of esophageal squamous cell carcinoma in vitro. World J Gastroenterol. 15:697–704. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Wang F, Li XK, Xu HY, Shan ZZ, Wang T, Yang ZC, He W, Wang LX and Fan QX: N-cadherin participated in invasion and metastasis of human esophageal squamous cell carcinoma via taking part in the formation of vasculogenic mimicry. Med Oncol. 32:4802015. View Article : Google Scholar : PubMed/NCBI

35 

Jin H, Morohashi S, Sato F, Kudo Y, Akasaka H, Tsutsumi S, Ogasawara H, Miyamoto K, Wajima N, Kawasaki H, et al: Vimentin expression of esophageal squamous cell carcinoma and its aggressive potential for lymph node metastasis. Biomed Res. 31:105–112. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 16 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ke K, Sun Z and Wang Z: Downregulation of long non‑coding RNA GAS5 promotes cell proliferation, migration and invasion in esophageal squamous cell carcinoma Retraction in /10.3892/ol.2024.14372. Oncol Lett 16: 1801-1808, 2018
APA
Ke, K., Sun, Z., & Wang, Z. (2018). Downregulation of long non‑coding RNA GAS5 promotes cell proliferation, migration and invasion in esophageal squamous cell carcinoma Retraction in /10.3892/ol.2024.14372. Oncology Letters, 16, 1801-1808. https://doi.org/10.3892/ol.2018.8797
MLA
Ke, K., Sun, Z., Wang, Z."Downregulation of long non‑coding RNA GAS5 promotes cell proliferation, migration and invasion in esophageal squamous cell carcinoma Retraction in /10.3892/ol.2024.14372". Oncology Letters 16.2 (2018): 1801-1808.
Chicago
Ke, K., Sun, Z., Wang, Z."Downregulation of long non‑coding RNA GAS5 promotes cell proliferation, migration and invasion in esophageal squamous cell carcinoma Retraction in /10.3892/ol.2024.14372". Oncology Letters 16, no. 2 (2018): 1801-1808. https://doi.org/10.3892/ol.2018.8797