miR-138 inhibits gastric cancer growth by suppressing SOX4

Retraction in: /10.3892/or.2022.8258

  • Authors:
    • Lei Pang
    • Bai Li
    • Baisong Zheng
    • Liang Niu
    • Liang Ge
  • View Affiliations

  • Published online on: June 22, 2017     https://doi.org/10.3892/or.2017.5745
  • Pages: 1295-1302
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA-138 (miR-138) has been reported to be downregulated and function as a tumor suppressor in several cancers. However, the role and molecular mechanisms of miR-138 in the progression of gastric cancer (GC) remain to be clarified. The aim of the present study was to determine the role of miR-138 in GC progression. In the present study we found that miR-138 expression was downregulated in GC tissues and cell lines. Statistical analysis demonstrated that low expression levels of miR-138 were associated with advanced tumor-node-metastasis (TNM) stage, and lymph node metastasis. Function assays demonstrated that overexpression of miR-138 impaired GC cell proliferation, colony formation, migration and invasion in vitro, as well as suppressed tumor growth in vivo. Through reporter gene, qRT-PCR and western blot assays, SRY-related high mobility group box 4 (SOX4), a master mediator in epithelial-mesenchymal transition (EMT), was confirmed to be a direct target of miR-138 in GC cells. Western blot assay revealed that miR-138 overexpression inhibited EMT procession in GC cells by upregulation of epithelial marker E-cadherin and downregulation of mesenchymal markers, N-cadherin and vimentin. Furthermore, the levels of miR-138 were inversely correlated with those of SOX4 expression in GC tissues. Overexpression of SOX4 rescued the inhibition effect in GC cells caused by miR-138. Collectively, these findings indicate that miR-138 may be a potential therapeutic target for GC.

Introduction

Gastric cancer (GC) is the fourth most common malignancy worldwide, and the second leading cause of cancer-related deaths worldwide (1). Despite recent advances in the diagnosis and management of GC, patients with advanced GC have a poor prognosis and the 5-year overall survival rate remains low at ~25% due to tumor recurrence, metastasis and poor response to chemotherapy and radiotherapy (2,3). In addition, the unclear pathophysiological mechanisms of GC have limited its clinical treatment options (4). Thus, it is essential to identify the molecular characterizations of GC in order to find new diagnostic strategies and potential drugs targets.

MicroRNAs (miRNAs), a class of small non-coding RNAs that regulate the gene expression by binding to the 3′-untranslated region (3-UTR) of target mRNAs, have been reported to be involved in various biological processes, such as cell proliferation, the cell cycle, migration, invasion and apoptosis (5,6). Emerging evidence has indicated that miRNAs function as oncogenes or tumor-suppressor genes by regulating cancer cell proliferation, invasion, migration, differentiation and apoptosis (79). Recently, the aberrant expression of miRNAs has been revealed to play essential roles in tumorigenesis and progression of GC (10,11).

Growing evidence has revealed that microRNA-138 (miR-138) is frequently downregulated in hepatocellular carcinoma (12), colorectal cancer (13), osteosarcoma (14), glioma (15), non-small-cell lung (16) and oral squamous cell carcinoma (17), cervical (18) and breast cancer (19), suggesting a tumor-suppressive role of miR-138 in human tumorigenesis. However, the roles of miR-138 as well as its downstream targets in the regulation of GC progression remain unclear. Therefore, in the present study we aimed to investigate the expression levels of miR-138 and its clinical significance in GC. We also studied the molecular mechanism by which miR-138 mediated the growth and metastasis of GC.

Materials and methods

Tumor tissue sample

GC tissues and their matched adjacent normal tissues were collected from 48 patients during surgical resection at The First Hospital of Jilin University (Changchun, China) from July 2014 to July 2015. All samples were immediately snap-frozen in liquid nitrogen after surgical removal and stored at −80°C until use. The pathological diagnosis was made at the Department of Pathology of The First Hospital of Jilin University. Written informed consents were obtained from all patients. The present study was approved by the Ethics Committee of Jilin University and all aspects of the study complied with the Declaration of Helsinki.

Cell culture and transfection

Human GC HGC-27, MGC-803, SGC-7901 and BGC-823 cell lines and immortalized gastric epithelial cells GES-1 were purchased from the Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (Shanghai, China). All cells were cultured in Roswell Park Memorial Institute-1640 (RPMI-1640) medium supplemented with 10% fetal bovine serum (FBS) (both from Life Technologies, Carlsbad, CA, USA) at 37°C with 5% CO2.

SGC-7901 cells were plated into 6-well plates at density of 5×105 cells/well. Twenty-four hours after plating, 100 nM of miR-138 mimic or corresponding negative control (miR-NC) (Ambion, Carlsbad, CA, USA) or SRY-related high mobility group box 4 (SOX4) overexpression plasmid (pCDNA3.1-SOX4 from Dr Gou Wang, Jilin University) were transfected into the cells using Oligofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's protocol.

RNA extraction and quantitative RT-PCR

Total RNA including miRNAs were extracted from culture cells or tissues using the TRIzol reagent (Invitrogen) following the manufacturer's instructions. The quality and concentration of RNA were identified by the A260/A280 ratio and 1% agarose gel electrophoresis. For the detection of the expression level of miR-138, miRNAs were reversely transcribed into cDNA by the High Capacity cDNA Synthesis kit (Applied Biosystems, Foster City, CA, USA). Quantitative PCR (qPCR) was performed on an Applied Biosystems 7900 Real-Time PCR machine with miRNA-specific primers using TaqMan Gene Expression kit (Applied Biosystems). The miR-138-specific primer and the internal control U6 primer were purchased from Ambion. For the detection of SOX4 mRNA level, cDNAs were synthesized using a PrimeScript® RT reagent kit (Takara Biotechnology Co., Ltd., Dalin, China). qPCR was performed on ABI7900 sequence detector (Applied Biosystems) using the SYBR Premix Ex Taq kit (Takara Biotechnology Co., Ltd.). The SOX4 and the internal control GAPDH primer were used in the present study as previously described (20). Their relative expression levels were calculated using the 2−ΔΔCt method (95% confidence interval) with calibration to the corresponding endogenous control.

Cell proliferation, colony formation, migration and invasion assays

Cell Counting Kit-8 (CCK-8) assay was used to assess the ability of cell proliferation. Briefly, transfected cells were plated in 96-well plates at a density of 5×104 cells/well. At 24, 48 and 72 h post-plating, 10 µl of CCK-8 solution (Sigma-Aldrich, St. Louis, MO, USA) was added to each well and incubated for 4 h at room temperature. The absorbance at 450 nm was assessed on a microplate reader (Molecular Devices, Menlo Park, CA, USA).

For colony formation assay, transfected cells were trypsinized, counted and seeded into 6-well plates at 300 cells/well at 37°C and 5% CO2 in a humidified incubator. During colony growth, the culture medium was replaced every 2 days. At 10 days after seeding, the colonies were fixed with 10% formaldehyde for 5 min and stained with 1.0% crystal violet for 1 min. The colony number in each well was counted under an inverted phase-contrast microscope (Olympus IX71; Olympus, Tokyo, Japan).

Wound healing was performed to examine the capacity of cell migration. Briefly, transfected cells were grown to confluence in a 24-well plate, and a wound was created into the cell monolayer with a sterile pipette tip. Cells were then washed using phosphate-buffered saline (PBS), and cultured in serum-free RPMI-1640 medium for 24 h. The width of the wound was measured at 0 and 24 h under an inverted phase-contrast microscope (Olympus IX71).

Cell invasion was determined using a 24-well plate Transwell® system with a polycarbonate filter membrane of 8-µm pore size (Corning, Cambridge, UK) assay kit. In brief, 5×104 transfected cells in serum-free medium were plated in the top chamber precoated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA). Medium supplemented with 10% FBS serum was used as a chemoattractant in the lower chamber. After incubation at 37°C for 48 h, the top chamber was wiped using a cotton swab to remove the non-invasive cells. The invading cells on the underside of the membrane were fixed in 100% methanol for 10 min, air-dried, stained in 0.1% crystal violet for 5 min. Five randomly selected fields of the fixed cells were imaged and counted under an inverted phase-contrast microscope (Olympus IX71).

Dual-luciferase reporter assays

Potential miR-138 targets were predicted and analyzed using 3 publicly available algorithms: PicTar, TargetScan and miRanda. SOX4 was predicted as a target of miR-138. According to the prediction results the wild-type SOX4 3′-UTR was amplified by PCR using human gastric cDNA and inserted into the psiCHECKTM-2 vector (Promega, Madison, WI, USA). The mutant-type SOX4 3′-UTR was generated using QuikChange Site-Directed Mutagenesis kit (Stratagene, La Jolla, CA, USA) as per the manufacturer's protocol, and then inserted into the psiCHECKTM-2 vector. For the dual-luciferase reporter assay, SGC-7901 cells were grown to ~80% confluence, and co-transfected with wild- or mutant-type SOX4 3′UTR reporter plasmid (50 ng), and miR-138 mimic or miR-NC (100 nM), using Lipofectamine 2000 as per the manufacturer's protocol. After 48 h, the activities of Renilla luciferase and firefly luciferase were determined using dual-luciferase reporter assay system (Promega). Renilla-luciferase was used for normalization.

Western blot analysis

Cells or tissues were harvested and lysed with RIPA buffer (Sigma-Aldrich), and western blotting was performed as previously described. The primary antibodies used for the analysis were mouse anti-human SOX4 (dilution, 1:1,000), mouse anti-human vimentin (dilution, 1:500), mouse anti-human N-cadherin (dilution, 1:500), mouse anti-human E-Cadherin (dilution, 1:500), and mouse anti-human GAPDH (dilution, 1:2,000), and horseradish peroxidase-conjugated (HRP) antibodies against mouse (dilution, 1:5,000) (all from Santa Cruz Biotechnology, Santa Cruz, CA, USA) were used as the secondary antibodies. The protein bands were detected with SuperSignal West Pico Chemiluminescent Substrate (Thermo Fisher Scientific, Inc., Waltham, MA, USA) and visualized on an UVP BioImaging System (Upland, CA, USA).

Xenograft model experiment

Twenty male BALB/c nude mice aged 6 weeks were purchased from the Laboratory Animal Center of Jilin University, and maintained under specific pathogen-free (SPF) conditions in the animal care facility at Jilin University. All animal experiments in the present study were approved by the Committee on Animal Welfare of Jilin University. A total of 2×106 SGC-7901 cells transfected with miR-138 mimic or miR-NC were suspended in 0.2 ml of Matrigel matrix (BD Biosciences), and then subcutaneously injected into the right side of the posterior flank of each mouse (n=10 each group), respectively. The length (L, cm) and width (W, cm) of the tumors were assessed every 5 days starting on the 10th day after inoculation using vernier callipers. The tumor volume was calculated using the formula: V = 0.5 × W2 × L. The mice were sacrificed on the 35th day after injection and the tumor tissues were stripped, weighed and stored for further assays.

Statistical analysis

Data were analyzed using SPSS version 19.0 software (SPSS, Inc., Chicago, IL, USA) and GraphPad Prism 5 (GraphPad Software, Inc., San Diego, CA, USA). The difference was determined using two-tailed Students t-test or one-way ANOVA. The relationship between miR-138 levels and SOX4 mRNA expression was tested with Pearson's correlation. Statistical significance was established at P<0.05.

Results

miR-138 is downregulated in GC tissues and cell lines

To explore the role of miR-138 in GC, we performed real-time quantitative RT-PCR in 48 pairs of GC and adjacent normal tissues. Compared with the adjacent normal tissues, miR-138 expression was significantly decreased in GC tissues (Fig. 1A). In clinical stages, miR-138 expression in cancer tissues of stages III and IV was significantly lower than in cancer tissues of stages I and II (Fig. 1B). Concomitantly, we compared the expression of miR-138 GC tissues with lymph node metastasis and GC tissues without lymph node metastasis. As the data revealed in Fig. 1C, the level of miR-138 in GC with lymph node metastasis was significantly lower than GC without lymph node metastasis. Moreover, we also investigated the expression of miR-138 in 4 human GC cell lines (HGC-27, MGC-803, SGC-7901 and BGC-823) and immortalized gastric epithelial cells GES-1 as control. The results demonstrated that miR-138 was significantly downregulated in GC cell lines compared with the human normal gastric cell line (Fig. 1D). These results revealed that miR-138 may be involved in gastric cancer tumorigenesis.

miR-138 suppresses the proliferation and colony formation of GC cells

To investigate the potential functions of miR-138 in GC cells, SGC-7901 cells, with the lowest expression of miR-138 of the 4 GC cell lines (Fig. 1D), were transfected with the miR-138 mimic or miR-NC to enhance miR-138 expression, and then cell proliferation and colony formation were determined. Firstly, we performed qRT-PCR to verify the efficacy of transfection. As indicated in Fig. 2A, transfection of cells with miR-138 mimic revealed a significant increase of miR-138 expression compared to cell transfection with miR-NC. The results from the CCK-8 assay revealed that overexpression of miR-138 significantly inhibited cell proliferation in SGC7901 cells (Fig. 2B). Consistent with this result, overexpression of miR-138 significantly inhibited cell colony formation in SGC7901 cells (Fig. 2C). These results revealed that miR-138 inhibited GC growth in vitro.

miR-138 inhibits cell invasion and migration in GC cells

Since the expression of miR-138 was correlated with lymph node metastasis, we explored the effect of miR-138 on cell migration and invasion in GC cells. The results from the wound-healing assay revealed that overexpression of miR-138 markedly decreased the migration of SGC-7901 cells (Fig. 3A). Matrigel Transwell assays revealed that miR-138 overexpression significantly inhibited the invasion of SGC-7901 cells (Fig. 3B). These results implied that miR-138 is involved in metastasis.

SOX4 is a direct target of miR-138 in GC cells

To identify the potential target mRNA of miRNA-138, the bioinformatics algorithms PicTar, TargetScan and miRanda were used. All of these databases predicted SOX4 as a target of miR-138, and the 3′-UTR of SOX4 mRNA contained a highly conserved binding site from positions 1,233 to 1,239 with a miR-138 seed sequence (Fig. 4A). Luciferase activity analysis further revealed that miR-138 significantly inhibited the luciferase activity of the wild-type SOX4-3-UTR reporter, but did not affect the mutant-type SOX4-3′-UTR reporter (Fig. 4B). Furthermore, qRT-PCR and western blot analysis revealed that the SOX4 expression at the mRNA and protein levels was downregulated in SGC7901 cells transfected with miR-138 compared to cells transfected with miR-NC (Fig. 4C and D). It has been demonstrated that SOX4 could act as a downstream regulator of epithelial-mesenchymal transition (EMT). Therefore, we wondered whether miR-138 regulates EMT. Thus, we also detected the expression of EMT epithelial marker E-cadherin and mesenchymal markers, N-cadherin and vimentin expression in SGC7901 cells after transfection with miR-138 mimic or miR-NC. As shown in Fig. 4D, E-cadherin was upregulated, while N-cadherin and vimentin were downregulated in SGC7901 cells after transfection with miR-138, suggesting that miR-138 regulates EMT progression. These data revealed that the SOX4 gene is one of the direct targets of miR-138 in GC cells.

SOX4 is upregulated in GC tissues, and inversely correlated with miR-138 expression

As the aforementioned results revealed, SOX4 is a direct target of miR-138 in GC cells. Therefore we next detected the expression of SOX4 in GC tissues and the corresponding adjacent normal tissues by qRT-PCR. We found that SOX4 mRNA expression was upregulated in GC tissues compared to adjacent normal tissues (Fig. 5A). Through Pearson's correlation analysis, we found that SOX4 mRNA expression was inversely correlated with miR-138 in GC tissues (Fig. 5B; r=−0.455; P=0.002).

miR-138 modulates cell growth, migration and invasion by targeting SOX4

To verify whether the inhibitory effects of miR-138 are mediated through SOX4, we rescued the expression of SOX4 in miR-138 mimic-transfected cells. qRT-PCR and western blot assays revealed that transfection of SOX4 overexpression plasmid (pCDNA3.1-SOX4) in miR-138 mimic-transfected cells could restore SOX4 expression (Fig. 6A and B). In addition, we also found that restoration of SOX4 expression partially reversed the inhibitory effect on cell proliferation, colony formation, migration and invasion in SGC7901 cells induced by miR-138 overexpression (Fig. 6C-F). These results revealed that miR-138 modulates cell growth, migration and invasion by targeting SOX4.

miR-138 suppresses tumor growth in vivo

To determine the effects of miR-138 on tumorigenicity in vivo, transfected cells were injected into the flanks of nude mice to form ectopic tumors. We found that tumor growth was slower in the miR-138 transfected cell group than that of the miR-NC transfected cell group (Fig. 7A). In addition, the size and weight of the tumors from the miR-138 transfected cell group were significantly decreased compared to the miR-NC transfected cell group (Fig. 7B and C). We also analyzed the expression of miR-138 and SOX4 in xenograft tumors. In the miR-138 transfected cell group, miR-138 expression was upregulated (Fig. 7D), whereas SOX4 expression was downregulated at the mRNA (Fig. 7E) and protein levels (Fig. 7F). These data indicated that miR-138 suppresses tumor growth in vivo by targeting SOX4.

Discussion

MicroRNAs (miRNAs) are important epigenetic regulators in the development and progression of human gastric cancer, and function as oncogenes or tumor suppressors in gastric cancer (GC) progression (10,11). Thus, identification of the exact biological role of miRNAs in GC progression and development may contribute to finding novel diagnosis markers and therapy targets for GC. In the present study we first confirmed that miR-138 was downregulated in GC tissues and cell lines, and its expression was closely associated with advanced tumor-node-metastasis (TNM) stage and lymph node metastasis. The in vitro and in vivo experiments revealed that restoration of miR-138 suppressed the cell proliferation, colony formation, migration and invasion of GC cells in vitro, as well as suppressed tumor growth by directly targeting SOX4. These data implied that miR-138 may be a promising therapeutic target of GC.

MicroRNA-138 (miR-138), a family of microRNA precursors, has been reported to play crucial roles in various biological processes, such as embryological morphogenesis, cell proliferation, cell invasion and developmental events tied to stem cell differentiation (21). Several studies have demonstrated that miR-138 played a suppressive role in multiple human cancers (1219). For instance, Liu et al revealed that miR-138 inhibited cell proliferation, colony formation, migration and invasion in hepatocellular carcinoma cells partially suppressing SOX9 expression (12). Xiao et al reported that miR-138 suppressed the growth and metastasis of non-small cell lung cancer cells partly at least via targeting YAP1 (22). Zhang et al demonstrated that miR-138 suppressed metastasis and EMT in breast cancer cells by targeting vimentin (19). In the present study, we found that the miR-138 expression level was significantly decreased in GC tissues compared to their matched adjacent normal tissues, and that decreased miR-138 was associated with advanced TNM stage and lymph node metastasis. However, a previous study reported that miR-138 was among 24 miRNAs that were significantly upregulated in GC tissues compared to normal gastric mucosae (23), yet, this previous study did not specify how many cases of GC tissues were used in the miRNA array analysis and did not verify their results by qRT-PCR (23). In addition, our results also revealed that miR-138 was downregualted in 4 GC cell lines compared to normal gastric cell lines, which further suggested that miR-138 may be downregulated in GC tissues and cell lines. We also found that enforced expression of miR-138 inhibited proliferation, colony formation, migration and invasion in GC cells, as well as suppressed tumor growth in nude mice. These results revealed that miR-138 functioned as a tumor suppressor by inhibiting GC progression.

It is well known that miRNAs exert their biological function primarily depending on the target gene (5,6). To explore the mechanisms underlying how miR-138 inhibited GC progression, we searched for potential targets of miR-138 in GC cells using several computational algorithms. Among the candidate target genes, we focused on SOX4 since it has been demonstrated to serve as an oncogene in various human cancers (2426). SOX4, a member of the Sox (Sry-related high-mobility group box) family of transcription factors, has been reported to promote tumor cell growth and metastasis, through regulation of multiple signaling pathways, such as the Wnt, Notch1 and p53 pathway (27,28). In addition, SOX4 could serve as a master mediator in epithelial-mesenchymal transition (EMT) (29,30), which is a critical step in tumor progression and metastasis (31). For GC, a previous study revealed that SOX4 expression was upregulated in GC tissues, and that overexpression of SOX4 was significantly correlated with depth of invasion, nodal status, distant metastasis, stage and vascular invasion, suggesting that SOX4 could serve as an oncogene in GC. Furthermore, SOX4 has been reported to be a target of miR-138 in ovarian cancer (32), however, the interaction between miR-138 and SOX4 has not been experimentally validated in GC. In the present study, luciferase reporter assay, qRT-PCR and western blot assays further confirmed their targeting relationship, and found that the expression of SOX4 was negatively regulated by miR-138 in GC cells. We also found that miR-138 downregulated vimentin expression and upregulated E-cadherin expression, which contributed to EMT inhibition. In addition, an inverse correlation between miR-138 and SOX4 mRNA expression was observed in GC tissues. Notably, overexpression of SOX4 markedly reversed the inhibitory effect of miR-138 on GC cell proliferation, migration and invasion. Our in vivo experiments also revealed that miR-138 inhibited tumor growth in nude mice by regulating SOX4. These results demonstrated that miR-138 played a tumor-suppressive role in GC, at least in part, by suppressing SOX4.

In summary, we investigated the biological functions of miR-138 in GC for the first time, and found that miR-138 was downregulated in GC cell lines and tissues, and was closely associated with advanced TNM stage and lymph node metastasis, and that miR-138 could inhibit malignant progression of GC by targeting SOX4. These findings revealed that miR-138 may function as a diagnostic marker and a therapeutic target for GC.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A, et al: Treatment of gastric cancer. World J Gastroenterol. 20:1635–1649. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Kim C, Mulder K and Spratlin J: How prognostic and predictive biomarkers are transforming our understanding and management of advanced gastric cancer. Oncologist. 19:1046–1055. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Guo H, Ingolia NT, Weissman JS and Bartel DP: Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 466:835–840. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

8 

McManus MT: MicroRNAs and cancer. Semin Cancer Biol. 13:253–258. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Farazi TA, Spitzer JI, Morozov P and Tuschl T: miRNAs in human cancer. J Pathol. 223:102–115. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Shrestha S, Hsu SD, Huang WY, Huang HY, Chen W, Weng SL and Huang HD: A systematic review of microRNA expression profiling studies in human gastric cancer. Cancer Med. 3:878–888. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Zhu X, Lv M, Wang H and Guan W: Identification of circulating microRNAs as novel potential biomarkers for gastric cancer detection: A systematic review and meta-analysis. Dig Dis Sci. 59:911–919. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Liu Y, Zhang W, Liu K, Liu S, Ji B and Wang Y: miR-138 suppresses cell proliferation and invasion by inhibiting SOX9 in hepatocellular carcinoma. Am J Transl Res. 8:2159–2168. 2016.PubMed/NCBI

13 

Zhao L, Yu H, Yi S, Peng X, Su P, Xiao Z, Liu R, Tang A, Li X, Liu F, et al: The tumor suppressor miR-138-5p targets PD-L1 in colorectal cancer. Oncotarget. 7:45370–45384. 2016.PubMed/NCBI

14 

Zhu Z, Tang J, Wang J, Duan G, Zhou L and Zhou X: MiR-138 acts as a tumor suppressor by targeting EZH2 and enhances cisplatin-induced apoptosis in osteosarcoma cells. PLoS One. 11:e01500262016. View Article : Google Scholar : PubMed/NCBI

15 

Wei J, Nduom EK, Kong LY, Hashimoto Y, Xu S, Gabrusiewicz K, Ling X, Huang N, Qiao W, Zhou S, et al: MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol. 18:639–648. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Li J, Wang Q, Wen R, Liang J, Zhong X, Yang W, Su D and Tang J: MiR-138 inhibits cell proliferation and reverses epithelial-mesenchymal transition in non-small cell lung cancer cells by targeting GIT1 and SEMA4C. J Cell Mol Med. 19:2793–2805. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Xu R, Zeng G, Gao J, Ren Y, Zhang Z, Zhang Q, Zhao J, Tao H and Li D: miR-138 suppresses the proliferation of oral squamous cell carcinoma cells by targeting Yes-associated protein 1. Oncol Rep. 34:2171–2178. 2015.PubMed/NCBI

18 

Li B, Yang XX, Wang D and Ji HK: MicroRNA-138 inhibits proliferation of cervical cancer cells by targeting c-Met. Eur Rev Med Pharmacol Sci. 20:1109–1114. 2016.PubMed/NCBI

19 

Zhang J, Liu D, Feng Z, Mao J, Zhang C, Lu Y, Li J, Zhang Q, Li Q and Li L: MicroRNA-138 modulates metastasis and EMT in breast cancer cells by targeting vimentin. Biomed Pharmacother. 77:135–141. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Jin Y, Zhao M, Xie Q, Zhang H, Wang Q and Ma Q: MicroRNA-338-3p functions as tumor suppressor in breast cancer by targeting SOX4. Int J Oncol. 47:1594–1602. 2015.PubMed/NCBI

21 

Schröder J, Ansaloni S, Schilling M, Liu T, Radke J, Jaedicke M, Schjeide BM, Mashychev A, Tegeler C, Radbruch H, et al: MicroRNA-138 is a potential regulator of memory performance in humans. Front Hum Neurosci. 8:5012014.PubMed/NCBI

22 

Xiao L, Zhou H, Li XP, Chen J, Fang C, Mao CX, Cui JJ, Zhang W, Zhou HH, Yin JY, et al: MicroRNA-138 acts as a tumor suppressor in non small cell lung cancer via targeting YAP1. Oncotarget. 7:40038–40046. 2016.PubMed/NCBI

23 

Yao Y, Suo AL, Li ZF, Liu LY, Tian T, Ni L, Zhang WG, Nan KJ, Song TS and Huang C: MicroRNA profiling of human gastric cancer. Mol Med Rep. 2:963–970. 2009.PubMed/NCBI

24 

Chen J, Ju HL, Yuan XY, Wang TJ and Lai BQ: SOX4 is a potential prognostic factor in human cancers: a systematic review and meta-analysis. Clin Transl Oncol. 18:65–72. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Han R, Huang S, Bao Y, Liu X, Peng X, Chen Z, Wang Q, Wang J, Zhang Q, Wang T, et al: Upregulation of SOX4 antagonizes cellular senescence in esophageal squamous cell carcinoma. Oncol Lett. 12:1367–1372. 2016.PubMed/NCBI

26 

Liu Y, Cui L, Huang J, Ji EH, Chen W, Messadi D and Hu S: SOX4 promotes progression in OLP-associated squamous cell carcinoma. J Cancer. 7:1534–1540. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Vervoort SJ, van Boxtel R and Coffer PJ: The role of SRY-related HMG box transcription factor 4 (SOX4) in tumorigenesis and metastasis: Friend or foe? Oncogene. 32:3397–3409. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Parvani JG and Schiemann WP: Sox4, EMT programs, and the metastatic progression of breast cancers: Mastering the masters of EMT. Breast Cancer Res. 15:R722013. View Article : Google Scholar : PubMed/NCBI

29 

Vervoort SJ, Lourenço AR, van Boxtel R and Coffer PJ: SOX4 mediates TGF-β-induced expression of mesenchymal markers during mammary cell epithelial to mesenchymal transition. PLoS One. 8:e532382013. View Article : Google Scholar : PubMed/NCBI

30 

Tiwari N, Tiwari VK, Waldmeier L, Balwierz PJ, Arnold P, Pachkov M, Meyer-Schaller N, Schübeler D, van Nimwegen E and Christofori G: Sox4 is a master regulator of epithelial-mesenchymal transition by controlling Ezh2 expression and epigenetic reprogramming. Cancer Cell. 23:768–783. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Ishiwata T: Cancer stem cells and epithelial-mesenchymal transition: Novel therapeutic targets for cancer. Pathol Int. 66:601–608. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Yeh YM, Chuang CM, Chao KC and Wang LH: MicroRNA-138 suppresses ovarian cancer cell invasion and metastasis by targeting SOX4 and HIF-1α. Int J Cancer. 133:867–878. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 38 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pang L, Li B, Zheng B, Niu L and Ge L: miR-138 inhibits gastric cancer growth by suppressing SOX4 Retraction in /10.3892/or.2022.8258. Oncol Rep 38: 1295-1302, 2017
APA
Pang, L., Li, B., Zheng, B., Niu, L., & Ge, L. (2017). miR-138 inhibits gastric cancer growth by suppressing SOX4 Retraction in /10.3892/or.2022.8258. Oncology Reports, 38, 1295-1302. https://doi.org/10.3892/or.2017.5745
MLA
Pang, L., Li, B., Zheng, B., Niu, L., Ge, L."miR-138 inhibits gastric cancer growth by suppressing SOX4 Retraction in /10.3892/or.2022.8258". Oncology Reports 38.2 (2017): 1295-1302.
Chicago
Pang, L., Li, B., Zheng, B., Niu, L., Ge, L."miR-138 inhibits gastric cancer growth by suppressing SOX4 Retraction in /10.3892/or.2022.8258". Oncology Reports 38, no. 2 (2017): 1295-1302. https://doi.org/10.3892/or.2017.5745