Open Access

miR‑613 suppresses migration and invasion in esophageal squamous cell carcinoma via the targeting of G6PD

Retraction in: /10.3892/etm.2023.12246

  • Authors:
    • Xiangyu Su
    • Chanchan Gao
    • Xiaoyao Feng
    • Ming Jiang
  • View Affiliations

  • Published online on: February 21, 2020     https://doi.org/10.3892/etm.2020.8540
  • Pages: 3081-3089
  • Copyright: © Su 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

Esophageal squamous cell carcinoma (ESCC) is a common cancer in China and has a high mortality rate. MicroRNAs (miRs) are a family of post‑transcriptional regulators, which negatively regulate target gene expression. miR‑613 has been revealed to be a diagnostic and prognostic biomarker in ESCC. However, the role of miR‑613 in ESCC remains unclear. In the present study, miR‑613 expression was identified to be reduced in tumor tissues in comparison with corresponding adjacent normal tissues. TargetScan and a dual‑luciferase reporter assay verified glucose‑6‑phosphate dehydrogenase (G6PD) as a direct target of miR‑613. In contrast with miR‑613, G6PD expression was increased in tumor tissues compared with matched healthy tissues. Furthermore, overexpression of miR‑613 inhibited cell migration and invasion of Eca109 cells compared with controls, while G6PD overexpression reversed the inhibition induced by miR‑613, as determined by wound healing and Transwell assays. In addition, miR‑613 overexpression decreased the mRNA and protein expression of G6PD, matrix metalloproteinase (MMP)2 and MMP9, and reduced the phosphorylation of signal transducer and activator of transcription 3 (STAT3) compared with controls, while G6PD reversed the effects of miR‑613. However, miR‑613 and G6PD did not affect the expression of STAT3. In conclusion, the aforementioned results suggest that miR‑613 targets G6PD to suppress ESCC cell migration and invasion through reduced MMP2 and MMP9 expression and inactivation of the STAT3 signaling pathway. Thus, the present study may provide a new molecular foundation for treatment of ESCC.

Introduction

Esophageal cancer (EC) is the sixth most common cause of cancer-associated mortality worldwide (1). Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent ECs in China, particularly in North-Central China, and accounts for 90% of all cases of EC (2). Surgery alone remains the standard treatment for patients at the early stage (3). However, ESCC is typically diagnosed at an advanced stage, due to a lack of clinical diagnostic techniques (4). Lymph node metastasis occurs in ~40% of patients with ESCC, which results in poor prognosis (5). The overall 5-year survival rate of ESCC patients is only 15-25% (6). The development of targeted therapy and improved understanding of the pathogenesis and molecular mechanisms underlying ESCC should facilitate the early diagnosis and treatment of ESCC.

MicroRNAs (miRNAs/miRs) are a family of short non-coding RNAs, which have been identified as post-transcriptional regulators (7). miRNAs can negatively regulate translation processes through binding to complementary sequences in the 3'-untranslated region (3'UTR) of target mRNAs (7,8). Alterations to the expression of miRNAs are the cause of numerous human malignancies (9). Furthermore, miRNA expression profiling is associated with the diagnosis, staging, progression and treatment of human cancers (10). A number of miRNAs are involved in biological and pathological processes in ESCC (11). Previous studies have reported that miR-613 functions as a tumor suppressor in human cancers, including glioma (12), hepatocellular carcinoma (13), non-small cell lung cancer (14) and laryngeal squamous cell carcinoma (15). In ESCC, miR-613 has been identified as a diagnostic and prognostic biomarker for patients (16). However, the molecular mechanism underlying the action of miR-613 in ESCC remains largely unknown.

The present study confirmed the expression of miR-613 in ESCC tissues. Overexpression of miR-613 was then revealed to suppress cell migration and invasion in vitro in comparison with controls. Notably, glucose-6-phosphate dehydrogenase (G6PD) was identified as a direct target of miR-613, and the overexpression of G6PD reversed the effects of miR-613. The present study may improve understanding of ESCC and assist with the development of future therapeutic targets.

Materials and methods

Clinical tissue samples

A total of 35 pairs of tumor tissues and matched adjacent healthy tissues were obtained from patients with ESCC who underwent surgery at Jiangsu Cancer Hospital (Nanjing, China) from July 2017 to July 2018. The tissue samples were frozen in liquid nitrogen and stored at -80˚C for further experiments. The average age of the 20 male and 15 female patients was 57.54 years (age range from 47-69 years). All patients had underwent surgery with no treatment after completion of pathological diagnosis and provided their written informed consent, prior to the study. The present study was approved by the Ethics Committee of Jiangsu Cancer Hospital.

Cell culture

The human ESCC cell line Eca109 was obtained from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. Cells were maintained in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin (all Gibco; Thermo Fisher Scientific, Inc.) in a humidified incubator at 37˚C with 5% CO2.

Cell transfection

The miR-613 mimic (cat. no. miR10003281-1-5; sequence: 5'-AGGAAUGUUCCUUCUUUGCC-3') and negative control (miR-NC; cat. no. miR01201-1-5; sequence: 5'-UUCUCCGAACGUGUCACGUTT-3') were purchased from Guangzhou RiboBio Co., Ltd. pcDNA3.1 vector was purchased from Shaanxi YouBio Technology Co., Ltd. The coding sequence of G6PD (NCBI accession no. NM_000402.4; forward, 5'-CCCAAGCTTATGGGCCGGCGGGGCTCAGC-3'; reverse, 5'-CGGGAATTCTCAGAGCTTGTGGGGGTTCA-3') was inserted into an empty pcDNA3.1 plasmid to prepare pcDNA3.1-G6PD. Eca109 cells were seeded into 6-well plates at a density of 2x105 cells/well. When confluence reached 50-70%, the cells were placed in serum-free medium and then transfected with 30 nM miR-613 mimic, miR-NC, empty vector pcDNA3.1 or pcDNA3.1-G6PD using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. After 6 h of transfection, the medium was replaced with complete medium and the cells were cultured for a further 48 h. Transfection efficiency was assessed by reverse transcription-quantitative PCR (RT-qPCR) as described below.

miR-613 target prediction and dual-luciferase reporter assay

Bioinformatics prediction tools [MicroRNA Target Prediction Database (miRDB): http://www.mirdb.org/ and TargetScan Human version 7.2: http://www.targetscan.org/vert_72/] were used to predict the potential targets of miR-613, and the data suggested that G6PD was a target of miR-613. To confirm this prediction, a dual-luciferase reporter assay was performed. G6PD 3'UTR wild type (WT; 5'-CCGAGCCCAGCUACAUUCCU-3') and mutant (5'-CCGAGCCCAGCUCACGCAAU-3') fragments, containing the predicted binding sites of miR-613, were separately inserted in the pmirGLO vector (Promega Corporation). Eca109 cells at a density of 2x105 were seeded into 24-well plates and co-transfected with 200 ng WT plasmid or mutant plasmid together with 50 nM miR-613 mimic or miR-NC mimic using Lipofectamine® 2000, according to the manufacturer's protocol. At 48 h post-transfection, luciferase activity was measured using a Dual-Luciferase Reporter assay system (Promega Corporation). Luciferase activity of Renilla was used for normalization.

RT-qPCR

To detect the expression of miR-613, total RNA was extracted from tissue samples or Eca109 cells using a miRNeasy mini kit (Qiagen GmbH). RT was performed using a One Step Primer Script miRNA cDNA Synthesis kit (Takara Biotechnology, Co., Ltd.). The RT conditions were 37˚C for 60 min and 85˚C for 5 sec. To detect the expression of G6PD, matrix metalloproteinase (MMP) 2 and MMP9, total RNA was isolated from tissues or Eca109 cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). First strand cDNA was synthesized from total RNA using a PrimeScript 1st strand cDNA Synthesis kit (Takara Biotechnology, Co., Ltd.). The RT reaction conditions were 30˚C for 10 min, 42˚C for 60 min and 95˚C for 5 min. qPCR was used to determine the expression of miRNA and mRNA using a Realtime PCR Master mix (SYBR Green; Toyobo Inc.) on an ABI PRISM 7700 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The reaction conditions were as follows: 95˚C for 60 sec, followed by 40 cycles of 95˚C for 15 sec and 60˚C for 60 sec. The specific primers were as follows: miR-613 forward: 5'-GTGAGTGCGTTTCCAAGTGT-3', reverse: 5'-TGAGTGGCAAAGAAGGAACATT-3'; U6 forward: 5'-GCACCTTAGGCTGAACA-3', reverse: 5'-AGCTTATGCCGAGCTCTTGT-3'; G6PD forward: 5'-AGCTGGAGGACTTCTTTGCC-3', reverse: 5'-TGATGCGGTTCCAGCCTATC-3'; MMP2 forward: 5'-GGGGCCTCTCCTGACATT-3', reverse: 5'-TCACAGTCCGCCAAATGAA-3'; MMP9 forward: 5'-TCCAACCACCACCACACCGC-3', reverse: 5'-CAGAGAATCGCCAGTACTT-3'; GAPDH forward: 5'-CTGGGCTACACTGAGCACC-3', reverse: 5'-AAGTGGTCGTTGAGGGCAATG-3'. miR-613 quantification was normalized to U6 and GAPDH was used for the normalization of G6PD, MMP2 and MMP9. RT-qPCR data were quantified according to the 2-ΔΔCq method (17).

Western blot analysis

Proteins from tissues or cellular lysates were obtained using RIPA lysis buffer (Beyotime Institute of Biotechnology) on ice. Protein concentrations were determined using a Pierce BCA protein assay kit (Thermo Fisher Scientific, Inc.). Total protein (30 µg) was separated by 10% SDS-PAGE and then transferred to a PVDF membrane (EMD Millipore). The membrane was blocked with 5% non-fat milk for 1 h at room temperature. Subsequently, the membrane was incubated with primary antibodies (all supplied by Abcam) against G6PD (cat. no. ab993; 1:1,000), MMP2 (cat. no. ab92536; 1:1,000), MMP9 (cat. no. ab76003; 1:1,000), signal transducer and activator of transcription 3 (STAT3; cat. no. ab68153; 1:1,000), phosphorylated (p)-STAT3 (cat. no. ab30647; 1:1,000) or GAPDH (cat. no. ab181602; 1:1,0000) at 4˚C overnight. The membrane was then incubated with goat anti-rabbit IgG (cat. no. ab205718; 1:2,000; Abcam) at room temperature for 1 h. The protein bands were visualized using an ECL Substrate kit (Abcam), and the density of each band was quantified with Image-Pro Plus software (version 6.0; Media Cybernetics, Inc.). GAPDH was used as an internal control.

Cell migration assay

Cell migratory ability was assessed by wound healing assay. Briefly, 5x105 transfected cells were seeded into six-well plates and incubated at 37˚C with 5% CO2 until the cell confluence reached 90-100%. A wound was then generated using a 10-µl sterile pipette tip and the resulting cell debris removed by washing twice with PBS. Immediately and again 24 h after making the wound, the width of the wound in each well was photographed under an inverted microscope (magnification x200; Olympus Corporation). The percentage of the wound area at 24 h compared with 0 h was quantified using a caliper.

Cell invasion assay

The invasive ability of Eca109 cells was measured using Transwell chambers pre-coated with Matrigel (24-well Transwell; 8-µm pore size filter; BD Biosciences). Transfected cells (200 µl) suspended in RPMI-1640 medium without serum were plated in the upper chambers at a density of 3x105 cells/ml. RPMI-1640 medium (600 µl) containing 10% FBS was added to the lower chamber. After incubation at 37˚C with 5% CO2 for 24 h, cells on the upper surface of the filter were removed. Cells on the under surface of the filter were fixed in 4% paraformaldehyde for 10 min and then stained with 0.1% crystal violet for 15 min at room temperature. The stained cells were imaged and counted using a light microscope (magnification x200; Olympus Corporation) in five random fields.

Statistical analysis

All data analyses were performed using GraphPad Prism version 7 (GraphPad Software, Inc.). All data are presented as the mean ± standard error of the mean. Statistical analyses were performed by a paired Student's t-test between tissue samples, and one-way analysis of variance followed by Tukey's post hoc test for multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-613 levels are reduced in ESCC tissues compared with matched controls

To investigate the expression of miR-613, 35 pairs of tumor tissues and matched adjacent healthy tissues were obtained. RT-qPCR was used to determine the miR-613 expression level. As presented in Fig. 1, the expression of miR-613 was significantly reduced in tumor tissue samples compared with matched healthy tissues (P<0.01). These results indicated that miR-613 expression is lower in ESCC compared with healthy controls.

G6PD is a potential target of miR-613

The potential targets of miR-613 were predicted using TargetScan and miRDB. G6PD was identified as a potential direct target of miR-613. It was indicated that miR-613 could bind to the 165-171 nucleotide position of the G6PD 3'UTR but not the corresponding mutated position of the G6PD 3'UTR (Fig. 2A). To confirm the binding of miR-613 and G6PD, a dual-luciferase reporter assay was performed. A G6PD 3'UTR WT or mutant-containing plasmid was co-transfected with miR-613 mimic or miR-NC into Eca109 cells, and the luciferase activity was measured. The results demonstrated that miR-613 significantly reduced luciferase activity when combined with the WT G6PD 3'UTR (P<0.01 vs. miR-NC). However, there was no significant difference when the mutant G6PD 3'UTR was co-transfected with miR-613 mimic or miR-NC (Fig. 2B).

G6PD expression is increased in ESCC tissues compared with control tissues

To further verify that G6PD is a target of miR-613, the mRNA and protein expression levels of G6PD in ESCC tissues and matched non-tumor tissues were measured by RT-qPCR and western blotting, respectively. As presented in Fig. 3A, the mRNA expression of G6PD was significantly increased in tumor tissues compared with matched non-tumor tissues (P<0.01). Similarly, the protein level of G6PD was significantly increased in ESCC tissues compared with matched non-tumor tissues (P<0.01; Fig. 3B).

Overexpression of miR-613 suppresses cell migration and invasion, while G6PD rescues the suppression induced by miR-613 in vitro

The present study evaluated the role of miR-613 and G6PD in ESCC cells. miR-613 mimic, miR-NC, pcDNA3.1 and pcDNA3.1-G6PD were transfected into Eca109 cells, and untransfected cells served as the control group. The transfection efficiency was measured by RT-qPCR. The expression of miR-613 was significantly upregulated in the miR-613 mimic group compared with the miR-NC and control groups (P<0.01; Fig. 4A). The expression of G6PD was significantly increased in the pcDNA3.1-G6PD group compared with the pcDNA3.1 and control groups (P<0.01; Fig. 4B).

Following transfection with miR-613 mimic and miR-NC, G6PD expression was determined at the mRNA and protein levels. The expression of G6PD was significantly reduced when cells were transfected with miR-613 mimic compared with miR-NC (P<0.01; Fig. 5). These results suggested that G6PD was negatively regulated by miR-613.

Subsequently, cell migratory ability was measured using a wound healing assay. The results suggested that the overexpression of miR-613 significantly inhibited the migration capabilities of Eca109 cells compared with the miR-NC group (P<0.01) while G6PD promoted cell migration and reversed the inhibition induced by the miR-613 mimic (all P<0.01; Figs. 6A and 7A). Similarly, Transwell assay results demonstrated that miR-613 overexpression significantly reduced cell invasion in vitro compared with that of miR-NC-transfected cells (P<0.01). Restoration of G6PD reversed the suppression induced by miR-613 (P<0.01), and there was no significant difference in the number of invaded cells between the miR-NC group and the miR-613 mimic + pcDNA3.1-G6PD group (Figs. 6B and 7B).

miR-613 inhibits MMP2 and MMP9 expression and the STAT3 signaling pathway, while G6PD attenuates the inhibitory effects caused by miR-613

The mRNA expression levels of MMP2 and MMP9 were determined by RT-qPCR. The results indicated that miR-613 markedly suppressed the expression of MMP2 and MMP9 when compared with miR-NC, while G6PD abolished the suppression induced by miR-613 (all P<0.01; Figs. 8A and B, 9A and B). In addition, western blotting was performed to examine the protein levels of STAT3, p-STAT3, MMP2 and MMP9. The results demonstrated that overexpression of miR-613 significantly inhibited MMP2, MMP9 and p-STAT3 levels, while G6PD reversed the effect caused by miR-613 (P<0.01). However, there was no significant difference in the total protein level of STAT3 among the different groups (Figs. 8C and 9C). These findings suggest miR-613 targets G6PD to inhibit MMP2 and MMP9 expression and the STAT3 pathway.

Discussion

In the present study, the role of miR-613 and its molecular mechanism in ESCC were revealed. miR-613 levels were identified to be reduced in ESCC tissues compared with matched healthy control tissues, and miR-613 overexpression inhibited cell migration and invasion in vitro when compared with miR-NC. The target gene of miR-613, G6PD, was highly expressed in tumor tissues and reversed the inhibition of migration and invasion induced by miR-613.

The role of miR-613 in cancer is unclear, as it has been reported to serve both promoter and inhibitor functions and it is involved in tumor cell migration and invasion. miR-613 acts as an oncogene in cervical cancer, in which it suppresses tumor cell proliferation, migration and invasion (18). In addition, miR-613 promotes the proliferation, migration and invasion of colon cancer via the targeting of protein atonal homolog 1(19). By contrast, miR-613 functions as a tumor suppressor in glioma, and has been demonstrated to inhibit cell proliferation, colony formation, migration and invasion in vitro and tumor growth in vivo (12). Additionally, miR-613 suppresses cell migration and invasion of triple-negative breast cancer (20). In the present study, the expression of miR-613 was decreased in ESCC tissues compared with corresponding non-tumor tissues. Furthermore, miR-613 inhibited the migration and invasion of ESCC cells in vitro. These findings suggest that miR-613 functions as a tumor suppressor in ESCC, which is similar to other cancer types (12-15,20).

The underlying molecular mechanism of miR-613 was investigated in the present study. G6PD was identified as a potential target of miR-613 using bioinformatics analysis. A dual-luciferase reporter assay was then performed to confirm this finding. It is understood that G6PD is a housekeeping gene in all cells, and it is a rate-limiting enzyme of the pentose phosphate pathway (PPP) (21). The PPP serves a critical role in cancer cell metabolism and survival, as it provides NADPH to synthesize fatty acids and generates pentose phosphates to promote nucleic acid synthesis (22,23). It has been revealed that the expression of G6PD is elevated in several cancer types, including renal cell carcinoma (24), hepatocellular carcinoma (25) and cervical cancer (26). Furthermore, G6PD is associated with tumor cellular processes, including proliferation, apoptosis, migration and invasion (24-27). A previous study of ESCC revealed that G6PD inhibits cell growth and apoptosis (28). However, to the best of our knowledge, its effects on migration and invasion are unknown. In the present study, G6PD was upregulated in ESCC tissues, which is consistent with its expression in other cancer types. In addition, G6PD expression was downregulated when miR-613 was overexpressed in Eca109 cells. Furthermore, G6PD reversed the inhibition of cell migration and invasion induced by miR-613. This indicates that G6PD acts as an oncogene and is negatively regulated by miR-613. Furthermore, it appears that miR-613 suppressed the migration and invasion by targeting G6PD.

MMPs are proteolytic enzymes that participate in the degradation of extracellular matrix, which can promote cancer progression by increasing its growth, migration, invasion, metastasis and angiogenesis (29,30). Downregulation of MMP2 and MMP9 inhibits tumor cell migration and invasion (31,32). STAT proteins, particularly STAT3, are activated in a large number of human cancer types (33). Aberrant expression of STAT3 induces tumorigenesis and promotes cell proliferation, angiogenesis, invasion and migration (34,35). Previous studies have revealed that knockdown of G6PD reduces the expression of MMP2, MMP9 and STAT3 (25,28). In the present study, overexpression of miR-613 suppressed the expression of MMP2 and MMP9, and decreased the phosphorylation of STAT3, while G6PD rescued the suppressive role of miR-613. Additionally, neither miR-613 nor G6PD affected the total protein level of STAT3. These findings suggest that miR-613 suppresses MMP2 and MMP9, and inactivates the STAT3 signaling pathway by targeting G6PD. They also indicate that the phosphorylation of STAT3, not the total expression of STAT3, served a role in the underlying mechanism.

In conclusion, miR-613 was demonstrated to function as a tumor suppressor in ESCC by suppressing the expression of MMP2 and MMP9, and inactivating the STAT3 signaling pathway via G6PD, which inhibited cell migration and invasion in vitro. These findings indicate that miR-613 and G6PD may be therapeutic targets for ESCC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

XS and MJ designed the study. Patient information was provided by MJ. XS, CG and XF performed the experiments and analyzed the data. XS was a major contributor in writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Jiangsu Cancer Hospital. All patients provided written informed consent prior to the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–386. 2015.PubMed/NCBI View Article : Google Scholar

2 

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.PubMed/NCBI View Article : Google Scholar

3 

Kelsen DP, Ginsberg R, Pajak TF, Sheahan DG, Gunderson L, Mortimer J, Estes N, Haller DG, Ajani J, Kocha W, et al: Chemotherapy followed by surgery compared with surgery alone for localized esophageal cancer. N Engl J Med. 339:1979–1984. 1998.PubMed/NCBI View Article : Google Scholar

4 

Liu YT, Zong D, Jiang XS, Yin L, Wang LJ, Wang TT, Zhu J and He X: miR-32 promotes esophageal squamous cell carcinoma metastasis by targeting CXXC5. J Cell Biochem. 120:6250–6263. 2019.PubMed/NCBI View Article : Google Scholar

5 

Wang H, Deng F, Liu Q and Ma Y: Prognostic significance of lymph node metastasis in esophageal squamous cell carcinoma. Pathol Res Pract. 213:842–847. 2017.PubMed/NCBI View Article : Google Scholar

6 

Pennathur A, Gibson MK, Jobe BA and Luketich JD: Oesophageal carcinoma. Lancet. 381:400–412. 2013.PubMed/NCBI View Article : Google Scholar

7 

Krol J, Loedige I and Filipowicz W: The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet. 11:597–610. 2010.PubMed/NCBI View Article : Google Scholar

8 

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

9 

Croce CM: Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 10:704–714. 2009.PubMed/NCBI View Article : Google Scholar

10 

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

11 

Mei LL, Qiu YT, Zhang B and Shi ZZ: MicroRNAs in esophageal squamous cell carcinoma: Potential biomarkers and therapeutic targets. Cancer Biomark. 19:1–9. 2017.PubMed/NCBI View Article : Google Scholar

12 

Sang Q, Liu X and Sun D: Role of miR-613 as a tumor suppressor in glioma cells by targeting SOX9. Onco Targets Ther. 11:2429–2438. 2018.PubMed/NCBI View Article : Google Scholar

13 

Jiang X, Wu J, Zhang Y, Wang S, Yu X, Li R and Huang X: MiR-613 functions as tumor suppressor in hepatocellular carcinoma by targeting YWHAZ. Gene. 659:168–174. 2018.PubMed/NCBI View Article : Google Scholar

14 

Li D, Li DQ, Liu D and Tang XJ: MiR-613 induces cell cycle arrest by targeting CDK4 in non-small cell lung cancer. Cell Oncol (Dordr). 39:139–147. 2016.PubMed/NCBI View Article : Google Scholar

15 

Wang J, Yang S, Ge W, Wang Y, Han C and Li M: MiR-613 suppressed the laryngeal squamous cell carcinoma progression through regulating PDK1. J Cell Biochem. 119:5118–5125. 2018.PubMed/NCBI View Article : Google Scholar

16 

Guan S, Wang C, Chen X, Liu B, Tan B, Liu F, Wang D, Han L, Wang L, Huang X, et al: MiR-613: A novel diagnostic and prognostic biomarker for patients with esophageal squamous cell carcinoma. Tumour Biol. 37:4383–4391. 2016.PubMed/NCBI View Article : Google Scholar

17 

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

18 

Li WT, Wang BL, Yang CS, Lang BC and Lin YZ: MiR-613 promotes cell proliferation and invasion in cervical cancer via targeting PTPN9. Eur Rev Med Pharmacol Sci. 22:4107–4114. 2018.PubMed/NCBI View Article : Google Scholar

19 

Yang X, Zhang L, Song X, He W, Zhang D, Lu Q, Wu J, Wu C and Jiang J: MicroRNA-613 promotes colon cancer cell proliferation, invasion and migration by targeting ATOH1. Biochem Biophys Res Commun. 504:827–833. 2018.PubMed/NCBI View Article : Google Scholar

20 

Xiong H, Yan T, Zhang W, Shi F, Jiang X, Wang X, Li S, Chen Y, Chen C and Zhu Y: miR-613 inhibits cell migration and invasion by downregulating Daam1 in triple-negative breast cancer. Cell Signal. 44:33–42. 2018.PubMed/NCBI View Article : Google Scholar

21 

Luzzatto L, Nannelli C and Notaro R: Glucose-6-phosphate dehydrogenase deficiency. Hematol Oncol Clin North Am. 30:373–393. 2016.PubMed/NCBI View Article : Google Scholar

22 

Patra KC and Hay N: The pentose phosphate pathway and cancer. Trends Biochem Sci. 39:347–354. 2014.PubMed/NCBI View Article : Google Scholar

23 

Jiang P, Du W and Wu M: Regulation of the pentose phosphate pathway in cancer. Protein Cell. 5:592–602. 2014.PubMed/NCBI View Article : Google Scholar

24 

Zhang Q, Yang Z, Han Q, Bai H, Wang Y, Yi X, Yi Z, Yang L, Jiang L, Song X, et al: G6PD promotes renal cell carcinoma proliferation through positive feedback regulation of p-STAT3. Oncotarget. 8:109043–109060. 2017.PubMed/NCBI View Article : Google Scholar

25 

Lu M, Lu L, Dong Q, Yu G, Chen J, Qin L, Wang L, Zhu W and Jia H: Elevated G6PD expression contributes to migration and invasion of hepatocellular carcinoma cells by inducing epithelial-mesenchymal transition. Acta Biochim Biophys Sin (Shanghai). 50:370–380. 2018.PubMed/NCBI View Article : Google Scholar

26 

Cui J, Pan Y, Wang J, Liu Y, Wang H and Li H: MicroRNA-206 suppresses proliferation and predicts poor prognosis of HR-HPV-positive cervical cancer cells by targeting G6PD. Oncol Lett. 16:5946–5952. 2018.PubMed/NCBI View Article : Google Scholar

27 

Hu H, Ding X, Yang Y, Zhang H, Li H, Tong S, An X, Zhong Q, Liu X, Ma L, et al: Changes in glucose-6-phosphate dehydrogenase expression results in altered behavior of HBV-associated liver cancer cells. Am J Physiol Gastrointest Liver Physiol. 307:G611–G622. 2014.PubMed/NCBI View Article : Google Scholar

28 

Wang X, Liu H, Zhang X, Li X, Gu H, Zhang H and Fan R: G6PD downregulation triggered growth inhibition and induced apoptosis by regulating STAT3 signaling pathway in esophageal squamous cell carcinoma. Tumour Biol. 37:781–789. 2016.PubMed/NCBI View Article : Google Scholar

29 

Visse R and Nagase H: Matrix metalloproteinases and tissue inhibitors of metalloproteinases: Structure, function, and biochemistry. Circ Res. 92:827–839. 2003.PubMed/NCBI View Article : Google Scholar

30 

Egeblad M and Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2:161–174. 2002.PubMed/NCBI View Article : Google Scholar

31 

Zhang JF, Wang P, Yan YJ, Li Y, Guan MW, Yu JJ and Wang XD: IL-33 enhances glioma cell migration and invasion by upregulation of MMP2 and MMP9 via the ST2-NF-kB pathway. Oncol Rep. 38:2033–2042. 2017.PubMed/NCBI View Article : Google Scholar

32 

Yang F, Yu N, Wang H, Zhang C, Zhang Z, Li Y, Li D, Yan L, Liu H and Xu Z: Downregulated expression of hepatoma-derived growth factor inhibits migration and invasion of prostate cancer cells by suppressing epithelial-mesenchymal transition and MMP2, MMP9. PLoS One. 13(e0190725)2018.PubMed/NCBI View Article : Google Scholar

33 

Yu H and Jove R: The STATs of cancer-new molecular targets come of age. Nat Rev Cancer. 4:97–105. 2004.PubMed/NCBI View Article : Google Scholar

34 

Siveen KS, Sikka S, Surana R, Dai X, Zhang J, Kumar AP, Tan BK, Sethi G and Bishayee A: Targeting the STAT3 signaling pathway in cancer: Role of synthetic and natural inhibitors. Biochim Biophys Acta. 1845:136–154. 2014.PubMed/NCBI View Article : Google Scholar

35 

Wake MS and Watson CJ: STAT3 the oncogene-still eluding therapy? FEBS J. 282:2600–2611. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2020
Volume 19 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Su X, Gao C, Feng X and Jiang M: miR‑613 suppresses migration and invasion in esophageal squamous cell carcinoma via the targeting of G6PD Retraction in /10.3892/etm.2023.12246. Exp Ther Med 19: 3081-3089, 2020
APA
Su, X., Gao, C., Feng, X., & Jiang, M. (2020). miR‑613 suppresses migration and invasion in esophageal squamous cell carcinoma via the targeting of G6PD Retraction in /10.3892/etm.2023.12246. Experimental and Therapeutic Medicine, 19, 3081-3089. https://doi.org/10.3892/etm.2020.8540
MLA
Su, X., Gao, C., Feng, X., Jiang, M."miR‑613 suppresses migration and invasion in esophageal squamous cell carcinoma via the targeting of G6PD Retraction in /10.3892/etm.2023.12246". Experimental and Therapeutic Medicine 19.4 (2020): 3081-3089.
Chicago
Su, X., Gao, C., Feng, X., Jiang, M."miR‑613 suppresses migration and invasion in esophageal squamous cell carcinoma via the targeting of G6PD Retraction in /10.3892/etm.2023.12246". Experimental and Therapeutic Medicine 19, no. 4 (2020): 3081-3089. https://doi.org/10.3892/etm.2020.8540