MicroRNA-338-3p suppresses tumor growth of esophageal squamous cell carcinoma in vitro and in vivo

Retraction in: /10.3892/mmr.2021.12450

  • Authors:
    • Xinyu Li
    • Zhihong Li
    • Guiyun Yang
    • Zhenxiang Pan
  • View Affiliations

  • Published online on: May 22, 2015     https://doi.org/10.3892/mmr.2015.3820
  • Pages: 3951-3957
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Accumulating evidence has shown that microRNAs (miRNAs) are aberrantly expressed in human esophageal squamous cell carcinoma (ESCC) and are crucial in tumorigenesis, among which miR‑338‑3p has been examined to be downregulated in patients with ESCC. However, the role of miR‑338‑3p in ESCC remains to be elucidated. In the present study, the role of miR‑338‑3p on the growth and survival of an ESCC cell line was determined with several in vitro approaches and in nude mouse models. It was determined that miR‑338‑3p expression was frequently downregulated in ESCC tissue compared with corresponding adjacent non‑tumor tissue, and that its expression was significantly correlated with tumor stage and metastasis. Overexpression of miR‑338‑3p in ESCC cells suppressed cell proliferation, colony formation, migration and invasion, and induced cell arrest at the G0/G1 stage and cell apoptosis in vitro. In addition, it was demonstrated that overexpression of miR‑338‑3p significantly suppresses tumor growth of xenograft tumors in mice (P<0.05). These findings revealed that miR‑338‑3p may act as a tumor suppressor in ESCC, and its dysregulation may be involved in the initiation and development of human ESCC. In addition, it was suggested that miR‑338‑3p may be a potential therapeutic agent for treatment of ESCC.

Introduction

Esophageal cancer (EC) is one of the most common types of cancer worldwide with a variable geographic distribution (1). It ranks eighth in order of occurrence and is the sixth leading cause of cancer-related mortality worldwide, with a higher incidence in males (1). It consists of two histological types, esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC), each with distinct etiologic and pathologic characteristics (2). ESCC is the major histological type of esophageal cancer in developing countries (3). Although advances have been made in the treatment of ESCC, including surgery, chemotherapy, radiation or a combination of these options, the prognosis of patients with ESCC remains poor, with the overall 5-year survival rate of patient after surgery is only 14–22% (3,4). To improve the overall outcome for patients with ESCC, it is important to understand the molecular mechanisms underlying these processes, which may contribute to the indentification of useful biomarkers and novel therapeutic agents.

MicroRNAs (miRNAs), a class of small non-coding RNAs 19–25 nucleotides in size, are involved in multiple biological processes, such as cell cycle, metabolism, cell differentiation, proliferation, oncogenesis, angiogenesis and cell invasion (58). miRNAs are novel posttranscriptional regulators of gene expression that target the 3′ untranslated region (3′-UTR) of mRNAs in a sequence-specific manner for translational repression or degradation (9,10). MicroRNAs (miRNAs) have been recognized as critical regulators of cancer invasion and metastasis, either as promoters or as suppressors in recent years (1113). In view of the close correlation between miRNAs and the biological progression of multiple cancers, miRNAs are presently considered to be potential novel targets for the treatment of various types of cancer (5,6,14,15).

miR-338-3p is a recently identified miRNA and is involved in cell proliferation, differentiation and invasion (1618). Previous investigation using miRNA microarrays has revealed that miR-338-3p is consistently downregulated in ESCC (19). However, knowledge regarding the exact roles of miR-338-3p in ESCC and the underlying molecular mechanisms remain relatively unclear. Therefore, the aim of this study was to identify its role in ESCC cells in vitro and in vivo to determine its utility in ESCC diagnosis and therapy.

Materials and methods

Clinical samples

A total of 48 patients with ESCC were enrolled in the present study and had undergone routine surgery at the Second Hospital of Jilin University (Changchun, China) between July 2011 and August 2013. ESCC samples and the corresponding adjacent esophageal tissues taken from the 48 patients were collected, immediately snap frozen in liquid nitrogen, and stored at −80°C until RNA extraction. This study was approved by the ethical committee of the Affiliated Hospital of Jilin University, and all patients provided written informed consent.

Cell lines and cell culture

EC9706 human ESCC cell lines were purchased from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China), were maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS; Gibco BRL, Gaithersburg, MD, USA) and incubated at 37°C and 5% CO2.

miRNA transfection

Cells were transfected with miR-338-3p mimics or corresponding negative control (GenePharma Co. Ltd., Shanghai, China) using Lipofectamine 2000 (Invitrogen Life Technologies, Carlbad, CA, USA) at a final concentration of 50 nM, according to the manufacturer's instructions. Transfection efficiencies were evaluated in every experiment by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) 48 h posttransfection. Cells were subdivided into the following three groups: Untransfected blank group (Blank), transfected negative control group (NC) and miR-338-3p mimic transfection group (miR-338-3p).

RNA preparation and RT-qPCR

Isolation of total RNA from cells was performed using Qiazol reagent and the miRNeasy mini kit (Qiagen, Valencia, CA, USA) at 48 h posttransfection according to the manufacturer's instructions. MicroRNA was reverse transcribed using the One Step Primescript miRNA cDNA Synthesis kit (Qiagen). Then miR-338-3p was quantified as described previously (20), using an Applied Biosystems 7900 Real-Time PCR system (Applied Biosystems Life Technologies, Foster City, CA, USA) and SYBR® Premix Ex Taq™ kits (Takara, Otsu, Japan) according to the manufacturer's instructions. The cycling conditions were as follows: Denaturation at 94°C for 5 min, followed by 40 cycles of the following for amplification, 94°C for 30 sec, 58°C for 60 sec and 72°C for 60 sec. U6 snRNA was used as an endogenous control. The comparative 2−ΔΔCt method was used for relative quantification and statistical analysis. The above experiment was repeated at least three times.

Cell proliferation and colony formation assays

The status of cell proliferation was determined by a 3-(4,5-dimethylthia-zole-2-yl)-2,5-biphenyl tetrazolium bromide (MTT; Amresco, Solon, OH, USA) assay. Exponentially growing ESCC cells were adjusted to 2.5×104 cells/ml with RPMI-1640, and then plated in 96-well plates (Corning, Corning, NY, USA) at 200 µl/well and then incubated for 12 h according to routine procedure. After transfection with miR-338-3p mimics or negative control for 48 h, 20 µl MTT (5 g/l) was added to each well. The medium was then removed after 4 h incubation and 100 µl/well dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO, USA) was added to dissolve the reduced formazan product. Finally, the plate was read in an enzyme-linked immunosorbent microplate reader (Bio-Rad 2550, Bio-Rad, Hercules, CA, USA) at 490 nm. The cellular proliferation inhibition rate was calculated as described previously (21).

For the colony formation assay, after cells were transfected with miR-338-3p mimics or negative control for 48 h, they were seeded in a 6-well plate at a low density (1,000 cells/per well), and cultured for 7 days. Then cells were fixed with 4% paraformaldehyde for 20 min and counted after staining with 1% crystal violet. The experiments were conducted in triplicate wells at least three times.

Cell cycle and cell apoptosis assay

The effects of miR-338-3p on ESCC cell cycle and apoptosis were examined by flow cytometry. In brief, cells were transfected with either miR-338-3p mimics or negative control miRNA for 48 h, then cells were harvested and washed twice with phosphate-buffered saline (PBS), fixed with 70% ethanol at −20°C for 30 min, and stored at 4°C overnight, then washed with PBS again, treated with 100 ml of 100 mg/l RNase at 37°C for 30 min, and stained with 100 ml of 50 mg/l propidium iodide at 4°C for 30 min in the dark. The multiplication cycle and apoptotic rate were assayed using flow cytometry (FACSCalibur; BD Biosciences, Mansfield, MA, USA), and the data were analyzed using CellQuest 2.0 software (BD Biosciences San Jose, CA, USA). The percentages of cells in the G0/G1 phase and S phase, and the apoptotic rate were measured by calculating the ratio of the number of corresponding cells to the number of total cells. In addition, Bcl-2 and survivin protein expression were determined by western blot analysis using specific antibodies as an additional indicator of apoptosis.

Wound-healing assays

Cells were treated with miR-338-3p mimics or corresponding negative controls when cells were grown to 80–90% confluence in 24-well plates. After 24 h of transfection, linear scratch wounds were created on the confluent cell monolayers with a pipette tip. To stop cells from entering the cell cycle prior to wounding, cells were maintained in serum-free medium. To visualize migrating cells and wound healing, images were captured at 0 and 24 h under an inverted microscope (IX51; Olympus Corporation, Tokyo, Japan). More than five field areas were selected randomly from each well and the cells in three wells of each group were determined.

Transwell assay

Cell invasion was measured using an 8-µm-pore polycarbonate membrane Boyden chamber insert in a Transwell apparatus (Millipore, MA, USA). In brief, the concentration of cells in each group was adjusted to 2.5×105 cells/ml at 48 h post-transfection. The upper chamber of a 24-well transwell permeable support with an 8-µm pore size was loaded with 200 µl cell suspension, and the lower chamber was filled with 500 µl RPMI-1640 medium containing 10% FBS, and then cultured for 48 h. Five wells were used for each group. After incubation, the media was removed from the upper chamber, and cells were scraped out of the upper chamber with a cotton swab. Cells that had migrated to the other side of the membrane were fixed with methanol, stained with hematoxylin, mounted and dried at 80°C for 30 min. The number of cells invading the Matrigel was counted in three randomly selected fields using an inverted microscope (IX51; Olympus Corporation). Experiments were performed in triplicate.

Western blot analysis

After 48 h of transfection, total proteins were prepared from the cells, quantities using a bicinchoninic acid protein assay (Beyotime Institute of Biotechnology, Haimen, China). Proteins were fractionated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis transferred to a polyvinylidene fluoride membrane (Invitrogen Life Technologies), blocked in 5% dry milk at room temperature for 1 h and immunostained with antibodies at 4°C overnight using anti-MMP-2 (1:1,000; cat. no. 13132; Cell Signaling Technology, Inc., Danvers, MA, USA); and anti-MMP-9 (1:2,000; cat. no. sc-12759; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), anti-survivin (1:5,000; cat. no. 2802s; Cell Signaling Technology, Inc.); and anti-Bcl-2 (1:3,000; cat. no. sc-7382; Santa Cruz Biotechnology, Inc). Anti-GAPDH (1:5,000; cat. no. sc-47724; Santa Cruz Biotechnology, Inc.) was used as a loading control. The membranes were incubated with the goat anti-mouse horseradish peroxidase (HRP)-conjugated IgG (1:10,000; cat. no. sc-2005; Santa Cruz Biotechnology, Inc.) or goat anti-rabbit HRP-conjugated IgG (1:10,000; cat. no. sc-2004; Santa Cruz Biotechnology, Inc.) for 2 h at room temperature. All results were visualized through a chemiluminescent detection system (Pierce, Pittsburgh, PA, USA) and then exposed in Molecular Imager ChemiDoc XRS system (Bio-Rad). The integrated density of the band was quantified by Quantity One v4.62 software (Bio-Rad).

Tumor growth in vivo

Thirty male BALB mice 5–6 weeks old, were purchased from Jilin Institute of Experimental Animals (Changchun, China). The research protocol was approved and mice were maintained in accordance with the Institutional Guidelines of the Experimental Animals of Jilin University.

Then, 2×106 untreated EC9706 cells, stably expressing miR-338-3p or corresponding negative control suspended in 50 µl PBS were injected into the flanks of mice (n=10), respectively. Mice were monitored weekly for tumor growth. Tumor volume was measured every week and calculated as 0.5236 × width2 × length. Three weeks after inoculation, mice were sacrificed by cervical dislocation, and tumors were resected and weighed, and part of tumors was used to measure the miR-338-3p level.

Statistical analysis

All data are expressed as the mean ± standard deviation. Comparisons between the groups were analyzed with one-way analysis of variance or two-tailed Student's t-test from SPSS version 19.0 software (SPSS Inc., Chicago, IL, USA) and GraphPad Prism version 5.01 (GraphPad Software, San Diego, CA, USA) for Windows®. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-338-3p was decreased in human ESCC tissues

The expression of miR-338-3p was analyzed in 48 EC samples and corresponding adjacent tissues by RT-qPCR. Significantly, miR-338-3p expression was lower in EC tissues than corresponding adjacent non-tumor tissues (P<0.01, Fig. 1). The correlation between miR-338-3p expression and clinicopathological characteristics is shown Table I. No positive correlation with gender, age or tumor location was observed; however, there was a significant correlation with tumor stage (P<0.01) and metastasis (P<0.01). The aberrant expression level of miR-338-3p implied that miR-338-3p may be key in ESCC development.

Table I

Correlation between relative level of miR-338-3p in ESCC tissue, and clinicopathological features in patients with ESCC.

Table I

Correlation between relative level of miR-338-3p in ESCC tissue, and clinicopathological features in patients with ESCC.

FeaturenmiR-338-3p expression levelP-value
Age (years)>0.05
 <60220.3186±0.023
 ≥60260.3212±0.024
Gender>0.05
 Male320.3210±0.022
 Female160.3142±0.019
Tumor location>0.05
 Middle300.3174±0.019
 Lower180.3323±0.023
Metastasis<0.05
 Yes240.2623±0.015
 No240.3613±0.027
TNM stage<0.05
 0–I310.3831±0.026
 II–IV170.2234±0.011

[i] TNM, tumor-node-metastasis.

miR-338-3p inhibits cell proliferation and colony formation in EC9706 cells

As it was shown that miR-338-3p is significantly downregulated in ESCC tissue, the present study aimed to determine how miR-338-3p affects ESCC cell behavior. To this end, miR-338-3p mimics and corresponding negative controls were transfected into EC9706 cells, respectively. The results of RT-qPCR demonstrated that expression of miR-338-3p in was increased in the miR-338-3p group compared with the NC and blank groups (P<0.05, Fig. 2A). Then cell proliferation was determined by an MTT assay. As shown in Fig. 2B, the viability of EC9706 cells was markedly decreased in the miR-338-3p group compared with the NC and blank groups (P<0.01).

The effect of miR-338-3p on cell colony formation in EC9706 cells was also determined and it was revealed that transfection with miR-338-3p mimics significantly inhibited cell colony formation compared with cells transfected with negative control and untransfected cells (P<0.05, Fig. 2C). These findings suggested that the miR-338-3p expression greatly inhibited cell proliferation and colony formation in EC9706 cells.

miR-338-3p induced cell cycle arrest and apoptosis in EC9706 cells. To determine the effects on miR-338-3p cell cycle, FACScan flow cytometry assays were performed. Flow cytometric analysis revealed that the G1-phase cell population was increased in the miR-338-3p group compared to blank group and NC group (P<0.05, Fig. 3A and B). In addition, miR-338-3p overexpression resulted in a lower percentage of cells in S phase compared with those of blank group and NC group (P<0.05, Fig. 3A and B).

In addition, flow cytometric analysis also showed that cells transfected with miR-338-3p could significantly induce cell apoptosis compared with untreated cells and cells transfected with negative control (P<0.05, Fig. 3C).

To determine the potential mechanism underlying cell apoptosis in vitro, survivin and Bcl-2 expression was detected using western blot analysis. It was found that survivin and Bcl-2 protein expression was significantly decreased in miR-338-3p treatment groups compared with the blank and NC groups (Fig. 3D).

miR-338-3p inhibits cell migration and invasion in EC9706 cells

To test whether miR-338-3p overexpressing cells possessed a reduced propensity for migration and invasion, wound-healing and Transwell assays were performed. For the wound-healing assay, microscopic observations of the three groups were recorded 24 h after scratching the cell surface. The capacity for wound healing was lower for the miR-338-3p group than for the blank and NC groups (P<0.05; Fig. 4A).

Using a Transwell assay, it was demonstrated that the mean number of cells penetrating the membrane was not identified to be significantly different for the blank and NC groups (P>0.05; Fig. 4B). However, the mean number of cells penetrating the Transwell membrane was significantly lower in the miR-338-3p group compared with the blank and NC groups (P<0.05; Fig. 4B). Based on these results, it was concluded that exogenous overexpression of miR-338-3p decreases the invasive ability of EC9706 cells.

Furthermore, the effects of miR-338-3p on the expression of cell invasion relevant proteins, MMP-2 and MMP-9 were analyzed by western blot analysis. As shown in Fig. 4C and D, MMP-2 and MMP-9 protein expression significantly decreased in the miR-338-3p treatment group compared with the blank and NC groups (P<0.05).

miR-338-3p suppresses tumor growth in a mouse xenograft model

It was investigated whether miR-338-3p could function as a tumor suppressor (as it does in other malignancies) using a mouse xenograft model. miR-338-3p mimic transfected EC9706 cells produced tumors with significantly reduced weight and volume 21 days after injection compared with tumors initiated by cells treated with negative control miRNA or untreated cells (P<0.05, Fig. 5A–C). To clarify miR-338-3p transfection activity, miR-338-3p expression was examined in xenograft tumors 21 days after injection. The results showed that miR-338-3p expression was upregulated in the xenograft tumors of miR-338-3p group compared with the xenograft tumors of the NC or control group (untreated group). These results may suggest that overexpression of miR-338-3p could inhibit tumor growth of ESCC in vivo.

Discussion

Increasing evidence showed that miRNAs have critical regulatory roles in cancer biology (22,23). Specifically for ESCC, it has been reported that miRNA contributes to proliferation, apoptosis, migration and invasion (24,25). For example, Ma et al (26) found that miR-21 was overexpressed in ESCC cell lines and ESCC tissue, and overexpression of miR-21 promoted cell proliferation and invasion of ESCC cells by targeting phosphatase and tensin homolog. Zhang et al (27) suggest that miR-518b may function as a tumor suppressor by targeting Rap1b in the development of ESCC, and that overexpression miR-518b inhibited ESCC cell proliferation and invasion. Wang et al (28) demonstrated that high expression of miR-625 inhibited proliferation and invasion in ESCC cells via controlling Sox2 expression. Wang et al (29) found that miR-655 is expressed at low levels in primary ESCC tissues, and upregulation of miR-655 inhibits ESCC cell invasiveness by targeting tumor-transforming gene-1 (PTTG1). The present aimed to provide evidence that the overexpression of miR-338-3p inhibited proliferation, colony formation, migration and invasion in ESCC.

The miR-338 gene is located on chromosome 17q25 within the eighth intron of the apoptosis-associated tyrosine kinase (AATK) gene. There are two mature forms of miR-338: miR-338-3p and miR-338-5p (30). It has been shown that miR-338-3p is involved in a variety of physiological and pathological processes, and was downregulated in several malignancies. For instance, Huang et al (31) showed that miR-338-3p expression is downregulated in hepatocellular carcinoma, in which miR-338-3p downregulation was significantly associated with TNM stage, vascular invasion, intrahepatic metastasis, tumor size and tumor grade. Xue et al (16) found that miR-338-3p downregulation in certain selective CRC samples, and that the miR-338-3p expression was not only associated with TNM stage but also with tumor metastasis. Peng et al (32) found that the level of miR-338 expression was significantly reduced in the tumor tissues compared with the adjacent normal mucosa tissues. In line with these results, our studies showed that miR-338-3p was frequently downregulated in ESCC tissues, and that miR-338-3p expression was significantly correlated with tumor stage and metastasis. These findings indicate that miR-338-3p may be a novel tumor suppressor miRNA for treatment of ESCC.

miR-338-3p has been shown to be involved in the development of several types of cancer (1618,3032); however, the underlying mechanism and its involvement in the development in ESCC remains to be elucidated. To reveal the exact role of miR-338-3p in ESCC, the effect of miR-338-3p on proliferation, colony formation, cell cycle, apoptosis, migration and invasion was analyzed by upregulating the expression level of miR-338-3p. The results showed that upregulation of miR-338-3p inhibited cell proliferation colony formation, migration and invasion, and induced cell apoptosis and cell arrest G0/G1 stage. In addition, it was demonstrated that overexpression of miR-338-3p could inhibit tumor growth of ESCC in vivo. These results suggest that miR-338-3p may be involved in the development of ESCC.

The degradation of basement membrane (BM) and extracellular matrix (ECM) is a critical event in tumour invasion and metastasis (33). Matrix metalloproteinases (MMPs), particularly MMP2 and MMP9, are a major group of enzymes that regulate ECM and BM composition during normal development and pathological responses (34,35). The results showed that enforced expression of miR-338-3p in EC9706 cells inhibited migration and invasion, and decreased MMP-2 and MMP-9 expression. These findings suggest that upregulation of miR-338-3p suppresses ESCC cell migration and invasion probably through inhibition of MMP-9 and MMP-2 expression.

In conclusion, the findings of the present study demonstrate that miR-338-3p was downregulated in ESCC tissues compared with corresponding adjacent tissues, which were associated with tumor depth, stage and metastasis, and that enforced expression of miR-338-3p significantly inhibited cell proliferation, clonogenicity, migration and invasion, and induced G1 arrest and cell apoptosis in vitro, as well as suppressed tumor growth in a nude mouse model. These findings suggest that miR-338-3p may be a potential therapeutic target in ESCC treatment.

References

1 

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

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

McCann J: Esophageal cancers: changing character, increasing incidence. J Natl Cancer Inst. 91:497–498. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Koshy M, Esiashvilli N, Landry JC, Thomas CR Jr and Matthews RH: Multiple management modalities in esophageal cancer: combined modality management approaches. Oncologist. 9:147–159. 2004. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Lin J, Huang S, Wu S, et al: MicroRNA-423 promotes cell growth and regulates G(1)/S transition by targeting p21Cip1/Waf1 in hepatocellular carcinoma. Carcinogenesis. 32:1641–1647. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Jovanovic M and Hengartner MO: miRNAs and apoptosis: RNAs to die for. Oncogene. 25:6176–6187. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Stevanato L and Sinden JD: The effects of microRNAs on human neural stem cell differentiation in two- and three-dimensional cultures. Stem Cell Res Ther. 5:492009. View Article : Google Scholar

9 

Djuranovic S, Nahvi A and Green R: A parsimonious model for gene regulation by miRNAs. Science. 331:550–553. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Kasinski AL and Slack FJ: Epigenetics and genetics. MicroRNAs en route to the clinic: progress in validating and targeting microRNAs for cancer therapy. Nat Rev Cancer. 11:849–864. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Valastyan S, Reinhardt F, Benaich N, et al: A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell. 137:1032–1046. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Iorio MV, Visone R, Di Leva G, et al: MicroRNA signatures in human ovarian cancer. Cancer Res. 67:8699–8707. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Iorio MV and Croce CM: MicroRNAs in cancer: small molecules with a huge impact. J Clin Oncol. 27:5848–5856. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Cho WC: OncomiRs: the discovery and progress of microRNAs in cancers. Mol Cancer. 6:602007. View Article : Google Scholar : PubMed/NCBI

16 

Xue Q, Sun K, Deng HJ, Lei ST, Dong JQ and Li GX: MicroRNA-338-3p inhibits colorectal carcinoma cell invasion and migration by targeting smoothened. Jpn J Clin Oncol. 44:13–21. 2014. View Article : Google Scholar

17 

Sun K, Guo C, Deng HJ, Dong JQ, Lei ST and Li GX: Construction of lentivirus-based inhibitor of hsamicroRNA-338-3p with specific secondary structure. Acta Pharmacol Sin. 34:167–175. 2013. View Article : Google Scholar

18 

Tsuchiya S, Oku M, Imanaka Y, et al: MicroRNA-338-3p and microRNA-451 contribute to the formation of basolateral polarity in epithelial cells. Nucleic Acids Res. 37:3821–3827. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Yang M, Liu R, Sheng J, et al: Differential expression profiles of microRNAs as potential biomarkers for the early diagnosis of esophageal squamous cell carcinoma. Oncol Rep. 29:169–176. 2013.

20 

Chen X, Pan M, Han L, Lu H, Hao X and Dong Q: miR-338-3p suppresses neuroblastoma proliferation, invasion and migration through targeting PREX2a. FEBS Lett. 587:3729–3737. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Sun K, Deng HJ, Lei ST, Dong JQ and Li GX: miRNA-338-3p suppresses cell growth of human colorectal carcinoma by targeting smoothened. World J Gastroenterol. 19:2197–2207. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Bartel DP: MicroRNAs: target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Kloosterman WP and Plasterk RH: The diverse functions of microRNAs in animal development and disease. Dev Cell. 11:441–450. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Akanuma N, Hoshino I, Akutsu Y, et al: MicroRNA-133a regulates the mRNAs of two invadopodia-related proteins, FSCN1 and MMP14, in esophageal cancer. Br J Cancer. 110:189–198. 2014. View Article : Google Scholar :

25 

Lee KH, Goan YG, Hsiao M, et al: MicroRNA-373 (miR-373) post-transcriptionally regulates large tumor suppressor, homolog 2 (LATS2) and stimulates proliferation in human esophageal cancer. Exp Cell Res. 315:2529–2538. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Ma WJ, Lv GD, Tuersun A, et al: Role of microRNA-21 and effect on PTEN in Kazakh's esophageal squamous cell carcinoma. Mol Biol Rep. 38:3253–3260. 2011. View Article : Google Scholar

27 

Zhang M, Zhou S, Zhang L, et al: miR-518b is downregulated, and involved in cell proliferation and invasion by targeting Rap1b in esophageal squamous cell carcinoma. FEBS Lett. 586:3508–3521. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Wang Z, Qiao Q, Chen M, et al: miR-625 downregulation promotes proliferation and invasion in esophageal cancer by targeting Sox2. FEBS Lett. 588:915–921. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Wang Y, Zang W, Du Y, et al: Mir-655 upregulation suppresses cell invasion by targeting pituitary tumor-transforming gene-1 in esophageal squamous cell carcinoma. J Transl Med. 11:3012013. View Article : Google Scholar

30 

Kos A, Olde Loohuis NF, Wieczorek ML, et al: A potential regulatory role for intronic microRNA-338-3p for its host gene encoding apoptosis-associated tyrosine kinase. PLoS One. 7:e310222012. View Article : Google Scholar : PubMed/NCBI

31 

Huang XH, Wang Q, Chen JS, et al: Bead-based microarray analysis of microRNA expression in hepatocellular carcinoma: miR-338 is downregulated. Hepatol Res. 39:786–794. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Peng Y, Liu YM, Li LC, Wang LL and Wu XL: MicroRNA-338 inhibits growth, invasion and metastasis of gastric cancer by targeting NRP1 expression. PLoS One. 9:e944222014. View Article : Google Scholar : PubMed/NCBI

33 

Rowe RG and Weiss SJ: Navigating ECM barriers at the invasive front: the cancer cell-stroma interface. Annu Rev Cell Dev Biol. 25:567–595. 2009. View Article : Google Scholar

34 

Buchheit CL, Weigel KJ and Schafer ZT: Cancer cell survival during detachment from the ECM: multiple barriers to tumour progression. Nat Rev Cancer. 14:632–641. 2014. View Article : Google Scholar : PubMed/NCBI

35 

MacDougall JR and Matrisian LM: Contributions of tumor and stromal matrix metalloproteinases to tumor progression, invasion and metastasis. Cancer Metastasis Rev. 14:351–362. 1995. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2015
Volume 12 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li X, Li Z, Yang G and Pan Z: MicroRNA-338-3p suppresses tumor growth of esophageal squamous cell carcinoma in vitro and in vivo Retraction in /10.3892/mmr.2021.12450. Mol Med Rep 12: 3951-3957, 2015
APA
Li, X., Li, Z., Yang, G., & Pan, Z. (2015). MicroRNA-338-3p suppresses tumor growth of esophageal squamous cell carcinoma in vitro and in vivo Retraction in /10.3892/mmr.2021.12450. Molecular Medicine Reports, 12, 3951-3957. https://doi.org/10.3892/mmr.2015.3820
MLA
Li, X., Li, Z., Yang, G., Pan, Z."MicroRNA-338-3p suppresses tumor growth of esophageal squamous cell carcinoma in vitro and in vivo Retraction in /10.3892/mmr.2021.12450". Molecular Medicine Reports 12.3 (2015): 3951-3957.
Chicago
Li, X., Li, Z., Yang, G., Pan, Z."MicroRNA-338-3p suppresses tumor growth of esophageal squamous cell carcinoma in vitro and in vivo Retraction in /10.3892/mmr.2021.12450". Molecular Medicine Reports 12, no. 3 (2015): 3951-3957. https://doi.org/10.3892/mmr.2015.3820