Open Access

Effects of miR‑138‑5p and miR‑204‑5p on the migration and proliferation of gastric cancer cells by targeting EGFR

Retraction in: /10.3892/or.2023.8677

  • Authors:
    • Yi Wang
    • Haiyang Zhang
    • Shaohua Ge
    • Qian Fan
    • Likun Zhou
    • Hongli Li
    • Ming Bai
    • Tao Ning
    • Rui Liu
    • Xia Wang
    • Ting Deng
    • Le Zhang
    • Guoguang Ying
    • Yi Ba
  • View Affiliations

  • Published online on: April 23, 2018     https://doi.org/10.3892/or.2018.6389
  • Pages: 2624-2634
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

GC (gastric cancer) remains one of the most lethal malignancies worldwide. EGFR (epidermal growth factor receptor) plays an important role in the malignant process of GC, therefore, the present study addressed the relationship between EGFR and its potential regulators and examined their regulatory mechanisms in GC. We examined differences in the expression levels of EGFR in GC and adjacent non‑cancerous tissues. Bioinformatics analyses and dual luciferase reporter assays were used to confirm the putative relationship between miR‑138 or miR‑204 and EGFR, and their relationship was further detected using western blotting, RT‑PCR, and a series of cell studies. EGFR proteins were abundantly expressed in GC tissues, however EGFR mRNA levels remained indistinctive. Consequently, EGFR was revealed as a putative target of miR‑138 and miR‑204 which bound to the 3'UTR of EGFR mRNA. Further analysis revealed that miR‑138 and miR‑204 were significantly downregulated in GC tissues and the overexpression of miR‑138 and miR‑204 in GC cell lines resulted in the significant inhibition of EGFR protein levels and GC cell proliferation and metastasis. Rescue experiments confirmed that the roles of the two microRNAs were specific to EGFR. EGFR is a pivotal oncogene in GC progression that may be regulated by miR‑138 and miR‑204.

Introduction

GC is one of the most frequently diagnosed and fatal cancers, remaining the fourth most common type of cancer and the second leading cause of cancer-related deaths worldwide (1). The highest incidence rates for GC have been reported in Eastern Asia, Eastern Europe, and South America, and the lowest incidence rates have been detected in North America and most regions of South Africa (2,3). Many patients succumb to GC following disease progression despite progress in surgical management (staging laparoscopy, nodal dissection and laparoscopic surgery) and adjuvant medical treatments (perioperative chemotherapy, radiation therapy, adjuvant chemo-radiation therapy or adjuvant chemotherapy alone) (4). Therefore, understanding the molecular mechanisms underlying GC development and progression would improve early diagnosis and therapy, leading to improved long-term survival for GC patients.

The epidermal growth factor receptor (EGFR) gene, also called ErbB1, encodes a transmembrane tyrosine kinase receptor, which is a member of the HER family. Other members of this family include ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4 (5). EGFR and its relatives are oncogenic drivers and are observed to be aberrant in many types of tumors (69). According to the updated review, EGFR is generally overexpressed in GC patients and high EGFR expression was significantly correlated with poor clinical outcomes (10). Previous studies have indicated that EGFR is correlated with aggressive tumor growth through the regulation of the cell cycle and angiogenesis (11), and notably, miRNAs affect the growth of GC by targeting EGFR (12). Furthermore, EGFR is considered to be an effective target in the treatment of cancers, such as GC, and targeted therapy, considering EGFR as a target, has become a new hot topic in GC research (13,14).

MicroRNAs (miRNAs) are small non-coding RNAs (usually ~22 nucleotides in length) targeting most protein-coding transcripts (15). In mammals, a majority of miRNAs guide the RNA-induced silencing complex (RISC) to the 3′ untranslated regions (UTRs) of mRNA targets, resulting in the inhibition of target mRNA translation (16). Currently almost 2,000 human miRNAs are listed in the miRBase, and these molecules have been predicted to control more than 30% of all genes (17). Various types of cancers exhibit the aberrant expression of miRNAs that may function as either oncogenes or tumor suppressors under certain conditions (18,19). Many miRNA deficiencies or excesses are correlated with GC progression, including metastasis and proliferation (20,21). As aforementioned, validation of the molecular mechanisms regulated by tumor-suppressive miRNAs can provide new insights into GC oncogenesis and may facilitate the development of novel therapeutic strategies for GC.

In the present study, miR-138-5p and miR-204-5p were downregulated, however EGFR was overexpressed in GC tissues. Thus, the aim of the present study was to address the relationship between EGFR and miR-138 or miR-204 and examine their regulatory mechanisms in depth as well as to reveal their contribution in GC. Notably, bioinformatics prediction provided primary evidence for specific binding between miR-138 or miR-204 and EGFR. Moreover, we determined the molecular association between miRs and EGFR in the migration and proliferation of GC cells. The results indicated that EGFR overexpression can accelerate GC progression which may be regulated by miR-138 and miR-204. These findings may help to further elucidate the current understanding of the molecular mechanisms of miRNAs and EGFR in GC.

Materials and methods

Human tissues

Fifteen human GC tissues and corresponding non-cancerous tissues were obtained from surgical patients at Tianjin Medical University Cancer Institute and Hospital (Tianjin, China). These patients, included 9 men and 6 women. The average age was 58 years (range, 43–76), and all patients received radical gastrectomy without any complications. All experimental GC tissues were notarized as adenocarcinoma according to pathological patterns, and non-cancerous tissues were confirmed as negative. Tissues were immediately frozen in liquid nitrogen at the time of surgery and subsequently were stored at −80°C.

Patients and ethics statements

This protocol was approved by the Ethics Committee of Tianjin Medical University Cancer Institute and Hospital and conformed to the standards set by the Declaration of Helsinki. Every patient provided written informed consent.

Cell lines and culture

SGC7901 and MGC803 GC cell lines which were purchased from the Shanghai Institute of Cell Biology of the Chinese Academy of Sciences (Shanghai, China) were grown in DMEM medium supplemented with 10% fetal bovine serum (FBS) (both from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and 1% penicillin/streptomycin (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China). Cells were cultured in a humidified incubator at 37°C with 5% CO2.

High-throughput sequencing

In total, the serum samples of 150 patients with primary GCs and 150 control subjects were subjected to high-throughput sequencing to identify miR-138 and miR-204 that were differentially expressed. Briefly, the serum samples from GC patients and healthy donors were pooled, and the total RNA was extracted. Finally, the reads were processed for in silico analysis (22).

Isolation of total RNA and quantitative RT-PCR

Total RNA was isolated from the cultured cells and tissues using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol. RNA concentrations and quality were confirmed using a Nanodrop 1000 spectrophotometer (Thermo Fisher Scientific, Inc.). Subsequently, both miRNA and mRNA were reverse-transcribed to cDNA. The reverse transcription was performed using AMV reverse transcriptase (Takara Biotechnology, Co., Ltd., Dalian, China) under certain conditions (16°C for 15 min, 42°C for 60 min and 85°C for 5 min). The expression of miR-138-5p and miR-204-5p was calculated by high-throughput sequencing (n=150). The gene-specific PCR products were assessed using qRT-PCR with the SYBR-Green PCR Kit (Takara) on the CFX96 Real-Time RT-PCR System. The PCR products were incubated in a 96-well optical plate, and the reactions were performed in triplicate. The PCR was initiated by a 5-min hold at 95°C, followed by 40 cycles of denaturation at 95°C for 15 sec and annealing/extension at 60°C for 1 min. The EGFR mRNA levels were normalized to GAPDH. The relative expression levels of the target genes were normalized to the control using the equation 2−ΔCt, in which ΔCt = Ct gene-Ct control. The following primers were used: GAPDH forward, 5′-TGGAAGGACTCATGACCACA-3′ and reverse, 5′-TTCAGCTCAGGGATGACCTT-3′; EGFR forward, 5′-TTGCCGCAAAGTGTGTAACG-3′ and reverse, 5′-GTCACCCCTAAATGCCACCG-3′.

Cell transfection

The cells were cultured in 6-well plates transfected with miR-138 or miR-204 mimics and inhibitors using Lipofectamine 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and Opti-MEM (Gibco; Thermo Fisher Scientific, Inc.) for 24 h according to the manufacturer's instructions. We used NC mimics and inhibitors as negative controls. The miR mimics promoted the expression of miRs, and in contrast, miR inhibitors displayed anti-miR effects. siRNA was used to suppress the expression of EGFR (cat. no. sc-29301; Santa Cruz Biotechnology, Inc., Dallas, TX, USA). Scrambled siRNA was used as a negative control. For each well, equal doses (100 pmol) of miRNA mimics, inhibitors, siRNAs, or scrambled negative control RNA molecules were used. The cells were harvested at 24 h after transfection for real-time PCR analysis and western blotting.

The lentivirus overexpressing EGFR and the control lentivirus were obtained from GenePharma (Shanghai, China), and an aliquot of 106 lentivirus was added into every single well with DMEM medium and Polybrene at an MOI of 10 according to the manufacturer's instructions.

Luciferase reporter assay

The 3′UTR of wild-type and mutant human EGFR, containing the predicted miR-138 and miR-204 targeting regions, was inserted into the pMIR-REPORT plasmid (Ambion; Thermo Fisher Scientific, Inc.). The following mutant sequences were constructed: miR-138-5p 5′-CGUUCAUAAGUUCCUGUGGUCGA-3′, and miR-204 5′-UUCCGUAUCAAGAAUGUUUCCCUAU-3′. For the luciferase reporter assays, 2 mg of firefly luciferase reporter plasmid, 2 mg of β-galactosidase expression vector (Ambion; Thermo Fisher Scientific, Inc.), and equal amounts (200 pmol) of mimics, inhibitors, or scrambled negative control RNA were transfected into 293T cells. A β-galactosidase expression vector acted as a transfection control. At 24 h after transfection, the cells were assayed using a luciferase assay kit (Promega Corp., Madison, WI, USA).

Protein extraction and western blotting

Protein was extracted from cells and tissues using RIPA buffer containing a freshly added protease inhibitor cocktail. The lysates were separated on 8% SDS-PAGE gels and subsequently transferred onto Immobilon PVDF membranes (EMD Millipore, Billerica, MA, USA). For immunodetection, the membranes were incubated with monoclonal anti-EGFR antibodies (1:2,500; cat. no. sc-31156; Santa Cruz Biotechnology, Inc.) overnight after blocking with 2% BSA. The signals from membranes after incubation with secondary antibodies (1:2,000; cat. no. sc-2768; Santa Cruz Biotechnology, Inc.) were generated using an enhanced chemiluminescence system kit (EMD Millipore) according to the manufacturer's instructions. The resulting values of the proteins of interest were normalized to GAPDH.

Cell proliferation assay

Cells seeded onto 24-well plates were first transfected with miR-138-5p or miR-204-5p mimics, inhibitors, the EGFR-overexpressing lentivirus, EGFR siRNA and the relevant negative control. At 24 h after transfection, EdU was added to the culture medium at a concentration of 50 µM/ml for 5 h to chase the DNA template. Briefly, after fixation in 4% paraformaldehyde and treatment with 0.5% Triton X-100 for 15 min, the cells were incubated in darkness with Apollo®, and the nuclei were stained with DAPI using the Cell-Light EdU DNA cell kit (Apollo® 567/488; Guangzhou RiboBio Co., Ltd., Guangzhou, China) according to the manufacturer's instructions. EdU-labeled and DAPI-labeled cells were manually counted in five fields randomly selected from each well, and the percentages were calculated. All experiments were performed in triplicate to do a statistical analysis.

Transwell migration assay

The cells were first transfected with miR-138-5p/miR-204-5p mimics, inhibitors, the EGFR overexpression lentivirus, EGFR siRNA and the relevant negative control. Twenty-four-well Boyden chambers with 8-µm pore size polycarbonate membranes (Corning Inc., Corning, NY, USA) were used and ~105 cells were seeded onto the upper chamber with 200 µl of serum-free medium at 24 h after transfection. Approximately 600 µl of medium supplemented with 10% serum was added to the lower chamber as a chemoattractant. Twenty-four hours after incubation, the non-migrating cells on the upper surface of the membrane were gently scraped off with cotton swabs. Subsequently, the membranes were fixed using methanol and stained with a three-step staining set (Thermo Fisher Scientific, Inc., Paisley, UK). The migrating cells were calculated in five visual fields randomly selected from each membrane. All experiments were performed in triplicate. The data of experimental group and control group were input to statistical analysis.

Wound-healing assay

The migration ability of SGC7901 and MGC803 cells was assessed using a wound-healing assay. Briefly, the cells were seeded onto six-well plates, and transfected with miR-138-5p or miR-204-5p mimics, inhibitors, the EGFR overexpression lentivirus, EGFR siRNA and the relevant negative control after 24 h. At 90% confluency, a plastic 20-µl pipette tip was used to draw across the centerline of the cultured cells to generate two linear 1-mm wound areas. After incubation for 0, 6, 12 and 24 h with DMEM medium containing 2% FBS (both from Gibco; Thermo Fisher Scientific, Inc.) in a humidified incubator, the migration of the cells into the wound area was examined under the EVOS® FL Cell Imaging System (Thermo Fisher Scientific, Inc.), and five random fields were selected for each well.

Immunohistochemistry assay

GC and the paired adjacent non-cancerous tissues were cut from paraffin block, and then incubated with the anti-EGFR monoclonal antibody (cat. no. sc-31156; Santa Cruz Biotechnology, Inc.) at a 1:50 dilution at 4°C overnight. The DAB system (Zhongshanjinqiao, Beijing, China) was used to identify the positive staining. All the samples were identified as positive or negative by two pathologists.

Biomaterial analysis of target predictions

miRNA target prediction and analysis were performed with the publically available algorithms from TargetScan (http://www.targetscan.org/), PicTar (http://pictar.mdc-berlin.de/) and miRanda (http://www.microrna.org/). RNAhybrid (http://bibiserv.cebitec.uni-bielefeld.de/rnahybrid/) was used to describe the probability of interaction by target accessibility. ΔG (minimum free energy) scores are computed as the free energy gained by microRNA to EGFR, and only ΔG ≤20 kcal/mol was considered to be the better match.

Statistical analysis

The results are presented as the average of at least three experiments, each performed in triplicate, with standard errors. Statistical analyses were performed using analysis of variance followed by Student's t-test using SPSS 20.0 (IBM Corp., Armonk, NY, USA). P-values of 0.05 were considered significant and are indicated with asterisks. In this study, ‘*’ indicates ‘P<0.05’, ‘**’ indicates ‘P<0.01’, and ‘***’ indicates ‘P<0.001’.

Results

EGFR is upregulated in GC

To evaluate the expression of EGFR at the protein level, we detected 15 pairs of GC tissues and corresponding non-cancerous tissues using western blotting. As is shown in Fig. 1A, the EGFR protein levels were significantly upregulated in GC tissues compared with normal adjacent tissues. The differences between the GC and NC groups were statistically significant (P=0.014). However, the expression of EGFR mRNA levels revealed little difference between cancer tissues and adjacent non-cancerous tissues (Fig. 1C). The disparity between the protein and mRNA levels suggested that the expression of EGFR was regulated at post-transcriptional levels in GC. Immunohistochemical analysis (IHC) revealed that EGFR was overexpressed in GC tissues but not adjacent non-cancerous tissues (Fig. 1D).

EGFR-related miR-138 and miR-204 are downregulated in GC

miRNAs function in the regulation of gene expression (23), and microRNAs in cancer have been underlined for patient prognosis and clinical responses for which many clinical trials are now underway (24). Using bioinformatics tools (TargetScan, miRanda, PicTar), we deduced that miR-138 and miR-204 potentially target EGFR, which bind to the 3′UTR position of EGFR mRNA as predicted (Figs. 1E and 2A). As shown in Fig. 2A, ΔG (minimum free energy) that was less than −20 kcal/mol was regarded as a perfect match. The binding site is highly conserved among many species, and studies have reported that miR-204 was greatly downregulated in GC tissues (25). Therefore, we detected the expression of miR-138 and miR-204 in the serums of 150 pairs of GC and NC patients through high-throughput sequencing, and observed decreased expression in GC patients (Fig. 1F).

miR-138 and miR-204 were selected to clarify potential associations with EGFR and the biological effects in GC.

Conformation of miR-138 or miR-204 direct targeting to EGFR

The results aforementioned revealed that the levels of miR-138 or miR-204 and EGFR have opposite correlations in GC tissues. Moreover, miR-138 and miR-204 may have a suppressive influence on EGFR according to the results of the bioinformatics analysis. Thus, the luciferase assay was performed to confirm the correlation between miR-138 or miR-204 and EGFR. The relative luciferase activity was definitely inhibited by the co-transfection of miR-138 or miR-204 mimics and the luciferase reporters containing the predicted target regions of EGFR mRNA (Fig. 2B and C), and naturally, when the binding sites in the 3′UTR were mutated, the inhibition was absent. Moreover, the results were antipodal when miR-138 or miR-204 inhibitors were transfected (Fig. 2B and C).

Expectedly, in SGC7901 and MGC803 cell lines, the overexpression of miR-138 or miR-204 through transfection with mimics resulted in the inhibition of EGFR proteins, whereas transfection with inhibitors could enhance EGFR protein levels (Fig. 3A and C). However, the EGFR mRNA levels exhibited no differences when treated with miR-138 or miR-204 (Fig. 3B and D), which also indicating that EGFR was regulated by miR-138 and miR-204 through a post-transcriptional pathway.

In summary, miR-138 and miR-204 directly target EGFR by binding to the 3′UTR of EGFR mRNA.

Overexpression of miR-138 and miR-204 suppresses migration and proliferation in SGC7901/MGC803 cells

Proliferation and metastasis, two hallmarks of malignancy, are the leading causes of cancer-related deaths (26). miR-138 and miR-204 targeting EGFR may influence many biological activities of cancer cells including migration and proliferation. Therefore, we transfected SGC7901 and MGC803 cells with miR-138 or miR-204 mimics/inhibitors and examined the effects on cellular proliferation and migration.

We conducted a Transwell assay to characterize the migration capacity of the transfected cells. After treating GC cells with miR-138 mimics, the migration capacity was markedly suppressed, while the transfection of miR-138 inhibitors enhanced migration, and the same properties were observed for miR-204 respectively (Figs. 4C and D, and 5C and D).

The wound healing assay was also used to verify the migration ability of SGC7901 and MGC803 cells transfected with the selected miRNAs. A single scratch was established in each well at 24 h after transfection and wound closure was subsequently monitored. As shown in Figs. 4E and 5E, we concluded that the upregulation of miR-138 and miR-204 prevented cell migration (Figs. 4E and 5E).

The proliferation of SGC7901 and MGC803 cells was confirmed using the Cell-Light EdU DNA Cell Kit. Undoubtedly, the upregulation of miR-138 and miR-204 suppressed cell proliferation. However, inhibiting the expression of miR-138 and miR-204 markedly promoted cell proliferation capacity, respectively (Figs. 4A and B, and 5A and B).

These results demonstrated that miR-138 and miR-204 act as suppressors in the migration and proliferation of SGC7901 and MGC803 cells.

Overexpression and silencing of EGFR regulates the biological effects of SGC7901 cells. To evaluate the biological functions of the impact of EGFR on SGC7901 cells, siRNA sequences and lentivirus particles were used to overexpress or silence EGFR respectively. RT-PCR for RNA and western blotting for protein were used to determine the silencing and overexpression efficiencies. Silencing of EGFR exhibited low-level protein and mRNA expression, while the protein and mRNA levels of EGFR lentivirus overexpression were markedly improved (Fig. 6A-D).

To understand the EGFR-related biological effects of SGC7901 cells, we also performed Transwell and wound healing assays, and used the Cell-Light EdU DNA Cell Kit. Expectedly, the transfection of EGFR siRNA resulted in significantly decreased migration and proliferation compared with the transfection of EGFR lentivirus particles (Fig. 7A-D).

Therefore, EGFR acts as a cancer promoter, and EGFR overexpression markedly accelerated the migration and proliferation of GC cells.

Co-transfection of Si.EGFR plus miR-138 or miR-204 inhibitors further confirms the specificity of miRs for EGFR

Given that EGFR is a potent cancer promoter in GC and miR-138 and miR-204 consistently suppressed the biological effects of GC cells, we focused on the cooperation between Si.EGFR and miR-138 or miR-204 inhibitors. We next correlated Si.EGFR and miR-138 or miR-204 inhibitors via co-transfection in the MGC803 cell line, further underscoring the biological relevance of these molecules. We assessed the transfection efficacy through western blotting, and as predicted, the co-transfection of Si.EGFR plus miR-138 or miR-204 inhibitors significantly relieved the inhibitory effect of Si.EGFR (Fig. 8A and B). Moreover, the Cell-Light EdU DNA Cell Kit and wound healing assay were further used to evaluate the coordinated regulation of these two molecules. As shown in Fig. 8C-E, the silencing of EGFR inhibited the migration and proliferation of GC cells, but the co-transfection of Si.EGFR plus miR-138 or miR-204 inhibitors significantly relieved this inhibitory effect, consistent with the results of western blotting.

In summary, this rescue experiment further confirmed the specificity of miR-138 and miR-204 for EGFR.

Discussion

Despite improvements in diagnosis and treatment, the outcomes of patients with GC remain poor (27). Thus, a better understanding of gastric carcinogenesis and the identification of novel molecular targets to improve the diagnosis and therapy of GC are warranted.

EGFR is a member of the receptor tyrosine kinase family and overexpressed in GC (28). EGFR may contribute to malignant progression through intracellular kinase domain activation immediately following the formation of ligand-induced dimerization (29). To date, a number of miRNAs have been associated with tumor type or stage, and may be developed as diagnostic markers or a promising strategy for cancer therapy (3032). Previously, microRNA-138 was identified as a critical tumor suppressor in different human cancers such as malignant melanoma and osteosarcoma (3335). Moreover, miRNA-204 was downregulated in GC tissues and serum samples (25,36), and reduced miR-204 levels may be employed as a novel biomarker for monitoring the treatment response and predicting the prognosis of GC (37). Hence, we validated the biological relevance of these two miRs in GC tissues and GC cell lines.

However, in the present study, we initially observed that miR-138 and miR-204 were decreased in GC tissues through high-throughput sequencing (n=150), indicating that the dysregulation of miR-138 or miR-204 is a crucial event of GC tumorigenesis. Consequently, an increased understanding of the functional roles of miR-138 and miR-204 in gastric carcinogenesis could provide insights into the mechanisms of tumor development and identify therapeutic targets.

In the present study, EGFR and miR-138 or miR-204 exhibited opposing trends in GC tissues and normal adjacent tissues. Consistent with this conclusion, EGFR is overexpressed in GC tissues and may play a common oncogenic role in GC progression, however the EGFR mRNA levels remained indistinctive, indicating a potential post-transcriptional pathway in the regulation of EGFR. Moreover, we further identified the potential targeting relationship between miR-138 or miR-204 and EGFR using luciferase reporter assays. These data suggested that miR-138 and miR-204 may bind to the 3′UTR of EGFR mRNA, and influence the biological activity of EGFR in GC cells. Subsequently, miR-138 and miR-204 upregulation played an important role in inhibiting the proliferation and migration of GC cells. Similarly, we also detected an inverse correlation between EGFR protein and mRNA levels in SGC7901 and MGC803 cells, demonstrating that miR-138 or miR-204 regulated EGFR expression through a post-transcriptional pathway. Furthermore, we performed a rescue experiment to specify the targeting regulation of miR-138 or miR-204 to EGFR in GC cells.

Collectively, these results demonstrated that miR-138 and miR-204 may potentially target EGFR and negatively regulate EGFR expression in GC.

To the best of our knowledge, this study is the first to examine the relationship between miR-138 or miR-204 and EGFR in GC. The results of the present study revealed that these two molecules play an important role in GC tumorigenesis by regulating EGFR. Admittedly, there are several potential limitations of the present study. A correction for multiple testing was performed, and some of the experimental sample sizes were small. Whether the two miRNAs could act as diagnosis markers and useful therapeutic targets still warrant long-term follow-ups and multicenter clinical trials.

In conclusion, miR-138 and miR-204 acted as novel players with tumor suppressor functions that targeted EGFR in the tumorigenesis of GC. These findings may contribute to the development of more effective therapeutic strategies for GC patients in the future.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (nos. 81772629, 81602158, 81602156, 81702437, 81702431, 81702275 and 81772843) and the Tianjin Health and Family Planning Commission Foundation of Science and Technology (15KG142). This study was also supported by the Tianjin Science Foundation (no. 16PTSYJC00170). The funders had no role in the study design, collection, analysis, and interpretation of data, in the writing of the report, and in the decision to submit this article for publication.

Availability of data and materials

The datasets used during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YW, HZ, SG and QF performed most of the experiments. YW and HZ analyzed the data, and wrote the manuscript. LZ, HL, TN and LZ performed some experiments. MB, RL, XW and TD reviewed and edited the manuscript. YB and GY designed the experiments and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of Tianjin Medical University Cancer Institute and Hospital.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferro A, Peleteiro B, Malvezzi M, Bosetti C, Bertuccio P, Levi F, Negri E, La Vecchia C and Lunet N: Worldwide trends in gastric cancer mortality (1980–2011), with predictions to 2015, and incidence by subtype. Eur J Cancer. 50:1330–1344. 2014. 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 

Carcas LP: Gastric cancer review. J Carcinog. 13:142014. View Article : Google Scholar : PubMed/NCBI

4 

De Mestier L, Lardière-Deguelte S, Volet J, Kianmanesh R and Bouché O: Recent insights in the therapeutic management of patients with gastric cancer. Dig Liver Dis. 48:984–994. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Park JS, Kim HS, Bae YS, Cheong JH, Rha SY, Noh SH and Kim H: Prognostic significance and frequency of EGFR expression and amplification in surgically resected advanced gastric cancer. Jpn J Clin Oncol. 46:507–516. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Feng X, Qin JJ, Zheng BS, Huang LL, Xie XY and Zhou HF: Association of epidermal growth factor receptor (EGFR) gene polymorphism with lung cancer risk: A systematic review. J Recept Signal Transduct Res. 34:333–334. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Lee HJ, Seo AN, Kim EJ, Jang MH, Kim YJ, Kim JH, Kim SW, Ryu HS, Park IA, Im SA, et al: Prognostic and predictive values of EGFR overexpression and EGFR copy number alteration in HER2-positive breast cancer. Br J Cancer. 112:103–111. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Berger MS, Greenfield C, Gullick WJ, Haley J, Downward J, Neal DE, Harris AL and Waterfield MD: Evaluation of epidermal growth factor receptors in bladder tumours. Br J Cancer. 56:533–537. 1987. View Article : Google Scholar : PubMed/NCBI

9 

Onguru O, Scheithauer BW, Kovacs K, Vidal S, Jin L, Zhang S, Ruebel KH and Lloyd RV: Analysis of epidermal growth factor receptor and activated epidermal growth factor receptor expression in pituitary adenomas and carcinomas. Mod Pathol. 17:772–780. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Zhang Z, Tang H, Lin J, Hu Y, Luo G, Luo Z, Cheng C and Wang P: Clinicopathologic and prognostic significance of human epidermal growth factor receptor in patients with gastric cancer: An updated meta-analysis. Oncotarget. 8:17202–17215. 2017.PubMed/NCBI

11 

Hong L, Han Y and Brain L: The role of epidermal growth factor receptor in prognosis and treatment of gastric cancer. Expert Rev Gastroenterol Hepatol. 8:111–117. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Li S, Zhang H, Ning T, Wang X, Liu R, Yang H, Han Y, Deng T, Zhou L, Zhang L, et al: MiR-520b/e regulates proliferation and migration by simultaneously targeting EGFR in gastric cancer. Cell Physiol Biochem. 40:1303–1315. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Thiel A and Ristimäki A: Targeted therapy in gastric cancer. APMIS. 123:365–372. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Zhang L, Yang J, Cai J, Song X, Deng J, Huang X, Chen D, Yang M, Wery JP, Li S, et al: A subset of gastric cancers with EGFR amplification and overexpression respond to cetuximab therapy. Sci Rep. 3:29922013. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Soifer HS, Rossi JJ and Saetrom P: MicroRNAs in disease and potential therapeutic applications. Mol Ther. 15:2070–2079. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Kozomara A and Griffiths-Jones S: MiRBase: Integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 39:D152–D157. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Shenouda SK and Alahari SK: MicroRNA function in cancer: Oncogene or a tumor suppressor? Cancer Metastasis Rev. 28:369–378. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Garzon R, Calin GA and Croce CM: MicroRNAs in cancer. Annu Rev Med. 60:167–179. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Qu Y, Zhang H, Duan J, Liu R, Deng T, Bai M, Huang D, Li H, Ning T, Zhang L, et al: MiR-17-5p regulates cell proliferation and migration by targeting transforming growth factor-β receptor 2 in gastric cancer. Oncotarget. 7:33286–33296. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Zhang H, Duan J, Qu Y, Deng T, Liu R, Zhang L, Bai M, Li J, Ning T, Ge S, et al: Onco-miR-24 regulates cell growth and apoptosis by targeting BCL2L11 in gastric cancer. Protein Cell. 7:141–151. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Chen X, Li Q, Wang J, Guo X, Jiang X, Ren Z, Weng C, Sun G, Wang X, Liu Y, et al: Identification and characterization of novel amphioxus microRNAs by Solexa sequencing. Genome Biol. 10:R782009. View Article : Google Scholar : PubMed/NCBI

23 

Song JH and Meltzer SJ: MicroRNAs in pathogenesis, diagnosis, and treatment of gastroesophageal cancers. Gastroenterology. 143:35–47. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Hayes J, Peruzzi PP and Lawler S: MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol Med. 20:460–469. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Canu V, Sacconi A, Lorenzon L, Biagioni F, Lo Sardo F, Diodoro MG, Muti P, Garofalo A, Strano S, D'Errico A, et al: MiR-204 down-regulation elicited perturbation of a gene target signature common to human cholangiocarcinoma and gastric cancer. Oncotarget. 8:29540–29557. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Greenlee RT, Murray T, Bolden S and Wingo PA: Cancer statistics, 2000. CA Cancer J Clin. 50:7–33. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Bennett C, Paterson IM, Corbishley CM and Luqmani YA: Expression of growth factor and epidermal growth factor receptor encoded transcripts in human gastric tissues. Cancer Res. 49:2104–2111. 1989.PubMed/NCBI

29 

Lemmon MA, Schlessinger J and Ferguson KM: The EGFR family: Not so prototypical receptor tyrosine kinases. Cold Spring Harb Perspect Biol. 6:a0207682014. View Article : Google Scholar : PubMed/NCBI

30 

Song S and Ajani JA: The role of microRNAs in cancers of the upper gastrointestinal tract. Nat Rev Gastroenterol Hepatol. 10:109–118. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Nassar FJ, Nasr R and Talhouk R: MicroRNAs as biomarkers for early breast cancer diagnosis, prognosis and therapy prediction. Pharmacol Therap. 172:34–49. 2016. View Article : Google Scholar

32 

Naidu S, Magee P and Garofalo M: MiRNA-based therapeutic intervention of cancer. J Hematol Oncol. 8:682015. View Article : Google Scholar : PubMed/NCBI

33 

Chen Y, Cao KE, Wang S, Chen J, He B, He G, Chen Y, Peng B and Zhou J: MicroRNA-138 suppresses proliferation, invasion and glycolysis in malignant melanoma cells by targeting HIF-1α. Exp Ther Med. 11:2513–2518. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Jiang B, Mu W, Wang J, Lu J, Jiang S, Li L, Xu H and Tian H: MicroRNA-138 functions as a tumor suppressor in osteosarcoma by targeting differentiated embryonic chondrocyte gene 2. J Exp Clin Cancer Res. 35:692016. View Article : Google Scholar : PubMed/NCBI

35 

Sun DK, Wang JM, Zhang P and Wang YQ: MicroRNA-138 regulates metastatic potential of bladder cancer through ZEB2. Cell Physiol Biochem. 37:2366–2374. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Yuan X, Wang S, Liu M, Lu Z, Zhan Y, Wang W and Xu AM: Histological and pathological assessment of miR-204 and SOX4 levels in gastric cancer patients. Biomed Res Int. 2017:68946752017. View Article : Google Scholar : PubMed/NCBI

37 

Chen X, Liu XS, Liu HY, Lu YY and Li Y: Reduced expression of serum miR-204 predicts poor prognosis of gastric cancer. Genet Mol Res. 15:doi: 10.4238/gmr.15027702. 2016.

Related Articles

Journal Cover

June-2018
Volume 39 Issue 6

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
Wang Y, Zhang H, Ge S, Fan Q, Zhou L, Li H, Bai M, Ning T, Liu R, Wang X, Wang X, et al: Effects of miR‑138‑5p and miR‑204‑5p on the migration and proliferation of gastric cancer cells by targeting EGFR Retraction in /10.3892/or.2023.8677. Oncol Rep 39: 2624-2634, 2018
APA
Wang, Y., Zhang, H., Ge, S., Fan, Q., Zhou, L., Li, H. ... Ba, Y. (2018). Effects of miR‑138‑5p and miR‑204‑5p on the migration and proliferation of gastric cancer cells by targeting EGFR Retraction in /10.3892/or.2023.8677. Oncology Reports, 39, 2624-2634. https://doi.org/10.3892/or.2018.6389
MLA
Wang, Y., Zhang, H., Ge, S., Fan, Q., Zhou, L., Li, H., Bai, M., Ning, T., Liu, R., Wang, X., Deng, T., Zhang, L., Ying, G., Ba, Y."Effects of miR‑138‑5p and miR‑204‑5p on the migration and proliferation of gastric cancer cells by targeting EGFR Retraction in /10.3892/or.2023.8677". Oncology Reports 39.6 (2018): 2624-2634.
Chicago
Wang, Y., Zhang, H., Ge, S., Fan, Q., Zhou, L., Li, H., Bai, M., Ning, T., Liu, R., Wang, X., Deng, T., Zhang, L., Ying, G., Ba, Y."Effects of miR‑138‑5p and miR‑204‑5p on the migration and proliferation of gastric cancer cells by targeting EGFR Retraction in /10.3892/or.2023.8677". Oncology Reports 39, no. 6 (2018): 2624-2634. https://doi.org/10.3892/or.2018.6389