Open Access

Silencing of KCNK15‑AS1 inhibits lung cancer cell proliferation via upregulation of miR‑202 and miR‑370

  • Authors:
    • Jun Peng
    • Xin‑Long Chen
    • Hong‑Zhong Cheng
    • Zhe‑Yuan Xu
    • Han Wang
    • Zhi‑Zhou Shi
    • Jun Liu
    • Xian‑Gu Ning
    • Hao Peng
  • View Affiliations

  • Published online on: October 2, 2019     https://doi.org/10.3892/ol.2019.10944
  • Pages: 5968-5976
  • Copyright: © Peng 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

Lung cancer is the most common cause of cancer‑associated mortality globally. Long non‑coding RNAs (lncRNAs) are transcripts with a length of >200 nucleotides, which are not translated into proteins. Growing evidence has indicated that certain lncRNAs are associated with various biological processes in cancer. However, the functions of KCNK15 and WISP2 antisense RNA 1 (KCNK15‑AS1) in lung cancer carcinogenesis and progression have remained elusive. The present study indicated that KCNK15‑AS1 was overexpressed in lung adenocarcinoma tissues compared with paracancerous normal tissues, and the high expression of KCNK15‑AS1 was significantly associated with poor prognosis compared with the patients with low expression (P<0.001). Furthermore, the knockdown of KCNK15‑AS1 was performed in A549 and H460 lung cancer cells with small interfering RNA, resulting in a significant inhibition of the proliferation, a decrease in the mRNA and protein expression of cyclin D1 (CCND1) and epidermal growth factor receptor (EGFR), in addition to the phosphorylation of protein kinase B, with a concomitant increase in the expression of microRNA (miR)‑202 and miR‑370 compared with negative control group. Rescue experiments demonstrated that the inhibition of miR‑202 or miR‑370 partially recovered the EGFR and CCND1 expression and the proliferation rates, which were reduced by KCNK15‑AS1 silencing. In conclusion, these results suggested that KCNK15‑AS1 functions as an oncogene via regulating the miR‑202/miR‑370/EGFR axis in lung cancer and may provide a potential target for lung cancer treatment.

Introduction

Lung cancer is the leading cause of cancer-associated mortality globally. Although progress has been made in the treatment of lung cancer, the survival of patients with lung cancer remains poor with a 5-year survival rate of only 17% (1,2). The characteristics of lung cancer are uncontrolled proliferation and metastasis of tumor cells. Therefore, understanding the regulatory mechanisms underlying lung cancer carcinogenesis and progression is necessary for tumor therapy.

Non-coding RNAs, including microRNAs (miRNAs/miRs) and long non-coding RNAs (lncRNAs) are considered to be potential biomarkers and candidate targets for the treatment of numerous cancer types (3). Certain lncRNAs have key functions in a variety of biological processes, including proliferation, apoptosis, stem cell properties, differentiation and metastasis (4,5). To date, numerous lncRNAs have been reported to be involved in the genesis of lung cancer. The lncRNA activated by transforming growth factor-β was identified to be overexpressed in lung cancer tissues and to promote the proliferation and metastasis of tumor cells by activating the p38 signaling pathway (6). Salt-inducible kinase (SIK)1-LNC, a type of lncRNA adjacent to SIK, was reported to be downregulated in lung cancer tissues and to repress the proliferation, migration and invasion of lung cancer cells (7). LncRNA KCNK15 and WISP2 antisense RNA 1 (KCNK15-AS1) was determined to be overexpressed in lung cancer tissues, and the higher expression of KCNK15-AS1 was associated with a shorter survival (8). However, the functional roles and underlying mechanisms of KCNK15-AS1 in the genesis of lung cancer remain largely elusive.

miR-202 and miR-370 have been previously reported to be decreased in lung cancer (911). miR-202 induces cell cycle arrest and apoptosis by targeting cyclin D1 (CCND1) and inhibits cell proliferation, migration and invasion via targeting signal transducer and activator of transcription (STAT3) in lung cancer (12,13). miR-370 has a tumor suppressive function in lung cancer by targeting tumor necrosis factor receptor-associated factor (TRAF4) and epidermal growth factor receptor (EGFR) (11,14).

In the present study, the regulatory functions of KCNK15-AS1 in lung cancer development, in addition to the associated molecular mechanisms, were investigated.

Materials and methods

Patients and samples

Fresh lung adenocarcinoma (LAD) and adjacent normal tissue samples from 40 patients were collected at the Department of Thoracic Surgery of the First People's Hospital of Yunnan (Kunming, China) between June 2014 and September 2015 and immediately stored at −70°C. All patients with LAD were treated using radical surgery and no patients received any pre-operative treatment. All samples were residual specimens following diagnostic sampling, and all patients provided written informed consent for sampling and molecular analysis separately. The present study was ethically approved by the Institutional Ethics Committee of the First People's Hospital of Yunnan Province (Kunming, China). Paracancerous tissue samples were collected at a 2-cm distance from the tumor edge as previously described (15), and the normal tissues were pathologically confirmed. The samples were graded by the AJCC staging classification system (8th edition) (16). The mean age of the patients was 62 years old (range, 38–79), and 65% of the patients (n=40) were male. Overall survival (OS) was defined as the time from surgery treatment to mortality or to the last follow-up. The clinical-pathological characteristics of the samples are presented in Table I.

Table I.

Association between KCNK15-AS1 expression and clinicopathological characteristics.

Table I.

Association between KCNK15-AS1 expression and clinicopathological characteristics.

KCNK15-AS1 expression

CharacteristicnHigh (n=19)Low (n=21)P-value
Sex 0.7475
  Female1468
  Male261313
Age, years 0.5266
  <501569
  ≥50251312
Degree of differentiation 0.0309
  Well or moderate18513
  Poor or undifferentiated22148
Clinical stage 0.0270
  I–II16412
  III–IV24159
T-stage 0.7518
  T1-T221912
  T3-T419109
N classification 0.0309
  N0-N118513
  N2-N322148
M classification 0.5962
  M0371720
  M1321

[i] KCNK15-AS1, KCNK15 and WISP2 antisense RNA 1; T, tumor; N, nodes; M, metastasis.

Cell culture

A549 (cat. no. CCL-185) and H460 cells (cat. no. HTB-177) were purchased from the American Type Culture Collection (Manassas, VA, USA). All cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 mg/ml streptomycin in an incubator at 37°C with 5% CO2.

Small interfering (si)RNA transfection

Cancer cells (2×105) were seeded in six-well plates and cultured with DMEM. KCNK15-AS1 siRNAs, miR-202 inhibitor, miR-370 inhibitor and a negative control were purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China). siRNAs or inhibitors (50 nM) were transfected into cells using Lipofectamine® RNAiMAX Transfection reagent for 48 h according to the manufacturer's protocol (Thermo Fisher Scientific, Inc.). The sequences of the KCNK15-AS1 siRNAs and negative control were as follows: siRNA-1 sense, 5′-GUCAUCACUACCAUCGGUGATT-3′ and antisense, 5′-UCACCGAUGGUAGUGAUGACTT-3′; siRNA-2 sense, 5′-GUCCGAGGCGGAAAGCGGTT-3′ and antisense, 5′-CCGCUUUCCGCCUCGGACTT-3′; negative control sense, 5′-UUCUCCGAACGUGUCACGUTT-3′ and antisense, 5′-ACGUGACACGUUCGGAGAATT-3′; miR-202 inhibitor, 5′-CAAAGAAGUAUAUGCAUAGGAA-3′; miR-370 inhibitor, 5′-GUAACUGCAGAGACGUGACCUG-3′; inhibitor negative control, 5′-CAGUACUUUUGUGUAGUACAA-3′. The miR-202 inhibitor and miR-370 inhibitor were used at a final concentration of 20 nM.

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

Total RNA was isolated from cells using RNAiso Plus (Takara Bio, Inc., Otsu, Japan). RT-qPCR was used to determine the expression levels of KCNK15-AS1, CCND1, EGFR and GAPDH. PCR was performed in a total volume of 20 µl, including 10 µl PowerUp™ SYBR™ Green Mix (Thermo Fisher Scientific, Inc.), 2 µl complementary DNA and 1 µl primer mix (10 µM each). PCR was performed in an ABI 7300 real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) as follows: Initial denaturation at 95°C for 10 min, followed by 40 cycles of 95°C for 10 sec and 60°C for 1 min. The PCR data were normalized to GAPDH and the relative expression of each gene was calculated using the 2−∆∆Cq method (17). The following primers were used: KCNK15-AS1 forward, 5′-AGCAGATGCAGAGAACCCAAA-3′ and reverse, 5′-TTGCAAGGCAGGTGTTTGTTC-3′; CCND1 forward, 5′-GCTGCGAAGTGGAAACCATC-3′ and reverse, 5′-CCTCCTTCTGCACACATTTGAA-3′; EGFR forward, 5′-AGGCACGAGTAACAAGCTCAC-3′ and reverse, 5′-ATGAGGACATAACCAGCCACC-3′; GAPDH forward, 5′-AAATCCCATCACCATCTTCCAG-3′ and reverse, 5′-GAGTCCTTCCACGATACCAAAGTTG-3′.

Western blot analysis

Cells were lysed using radioimmunoprecipitation assay buffer [50 mM Tris-HCl (pH 7.4), 150 mM NaCl and 1% Nonidet P-40]. Following incubation on ice for 45 min, the homogenates were centrifuged at 13,000 × g for 15 min at 4°C. The concentrations of samples were detected using the Bradford method (Beyotime Institute of Biotechnology, Haimen, China). Proteins (10–20 µg/lane) were separated by 12% SDS-PAGE and then transferred onto a 0.45-µm polyvinylidene difluoride membrane (EMD Millipore, Billerica, MA, USA). Subsequently, the membranes were blocked with 5% skimmed milk for 30 min at room temperature. The membranes were incubated with primary antibody at 4°C overnight. Prior to incubation with secondary antibodies [anti-mouse immunoglobulin G (IgG), horseradish peroxidase (HRP)-linked antibody, cat no. 7075, 1:10,000 dilution, Cell Signaling Technology, Inc., Danvers, MA, USA; anti-rabbit IgG, HRP-linked antibody, cat no. 7074, 1:10,000 dilution, Cell Signaling Technology, Inc.] at room temperature for 1 h, the membranes were washed with Tris-buffered saline containing 0.3% Tween-20. The protein signals were visualized using the Enhanced Chemiluminescence Detection reagent (cat no. PE0020; Beijing Solarbio Bioscience & Technology Co., Ltd., Beijing, China). The primary antibodies and dilution ratios were as follows: CCND1 (cat no. sc-450; 1:1,000 dilution; Santa Cruz Biotechnology, Inc.); EGFR (cat no. 4267, 1:1,000 dilution; Cell Signaling Technology, Inc., Dallas, TX, USA); phosphorylated protein kinase B (AKT; cat no. 4060, 1:1,000 dilution; Cell Signaling Technology, Inc.); AKT (cat no. 9272, 1:1,000 dilution; Cell Signaling Technology, Inc.) and β-actin (cat no. 4970, 1:5,000 dilution; Cell Signaling Technology, Inc.).

Cell proliferation assay

A Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was used to quantify the proliferation of A549 and H460 cells according to the manufacturer's protocol. Cells were cultured at 1,000 cells/well in 96-well plates and transfected for 24 h. Following incubation for 24, 48, 72, 96 or 120 h at 37°C, 10 µl CCK-8 reagent was added to each well, followed by incubation at 37°C for 1 h. The absorbance of each well was read at 450 nm on a microplate spectrophotometer (SPECTRAMax 190; Molecular Devices, LLC, Sunnyvale, CA, USA). Three independent experiments were performed.

Statistical analysis

Data were presented as the mean ± standard deviation, and statistical analysis was performed using GraphPad Prism 6.0 (GraphPad Software, Inc., La Jolla, CA, USA). The expression levels of KCNK15-AS1 between LAD tissues and paracancerous normal tissues were analyzed using a Student's t-test (two-tailed, paired). Other data were analyzed using a Student's t-test (two-tailed, unpaired) between two groups and one-way analysis of variance followed by a Tukey's post-hoc test for multiple comparisons. The association between KCNK15-AS1 expression and clinicopathological characteristics were analyzed using a χ2 test. Estimation of survival time distribution was performed using the Kaplan-Meier method. P<0.05 was considered to indicate a statistically significant difference. All experiments were independently performed at least three times.

Results

Silencing of KCNK15-AS1 inhibits the proliferation and decreases the expression of CCND1 in lung cancer cells

A previous study indicated that KCNK15-AS1 was overexpressed in lung cancer and that the high expression of KCNK15-AS1 was associated with poor prognosis (8). In the present study, 40 patients with lung cancer were assessed to confirm these results, and the analysis suggested that KCNK15-AS1 was significantly overexpressed in lung cancer compared with paracancerous tissues (P<0.001) and that its high expression was associated with poor overall survival (Fig. 1A and B). The high expression of KCNK15-AS1 was significantly associated with a poor degree of differentiation, advanced clinical stage and lymph node metastasis (P<0.05; Table I). In order to investigate the function of KCNK15-AS1, it was knocked down using siRNAs (Fig. 2A). The results indicated that the silencing of KCNK15-AS1 significantly inhibited the proliferation of A549 and H460 lung cancer cells (P<0.05; Fig. 2B). CCND1 is involved in the regulation of the cell cycle (18). Notably, the knockdown of KCNK15-AS1 caused a significant downregulation of the expression of CCND1 at the mRNA levels (P<0.001; Fig. 2C) and downregulation at the protein level (Fig. 2D). Furthermore, the expression of CCND1 in lung cancer tissues was significantly higher compared with that in paracancerous normal tissues (P<0.001; Fig. 2E).

Silencing of KCNK15-AS1 decreases the expression of EGFR and inhibits the phosphorylation of AKT

The EGFR/AKT axis has important functions in cancer cell proliferation (19,20); therefore, the present study assessed whether the silencing of KCNK15-AS1 affects EGFR/AKT signaling. RT-qPCR and western blot analyses indicated that the silencing of KCNK15-AS1 reduced the protein expression and significantly reduced the mRNA levels of EGFR (P<0.001) and inhibited the phosphorylation of AKT (Fig. 3A and B).

In tumors, EGFR has been reported to be targeted and regulated by miR-202 and miR-370 (14,21,22). The results of the present study suggested that the silencing of KCNK15-AS1 caused a significant upregulation of the expression of miR-202 and miR-370 in A549 and H460 lung cancer cells (P<0.001; Fig. 3C and D).

Inhibition of miR-202 or miR-370 partially recovers the inhibitory effect of KCNK15-AS1 knockdown on the proliferation of lung cancer cells

To confirm the involvement of miR-202 or miR-370 in the regulatory effect of KCNK15-AS1 on lung cancer cell proliferation, initially, miR-202 and miR-370 were knocked-down using inhibitors in A549 and H460 cells. A RT-qPCR assay confirmed the efficiency of miR-202 and miR-370 silencing (Fig. 4A and B). miR-202 and miR-370 inhibitors also significantly decreased the expression levels of miR-202 and miR-370 in the KCNK15-AS1-silenced lung cancer cells compared with the negative control (P<0.001; Fig. 4C and D). Inhibition of miR-202 or miR-370 partially recovered the EGFR expression levels in the KCNK15-AS1 silenced lung cancer cells (Fig. 4E-H). Furthermore, the knockdown of miR-202 or miR-370 partially recovered the cell proliferation ability and CCND1 expression in the KCNK15-AS1-silenced lung cancer cells (Fig. 5A-C).

Discussion

LncRNAs, a class of RNAs with a length of >200 nucleotides, have important functions in numerous biological processes, including tumor formation and development (23). A previous study indicated that KCNK15-AS1 was overexpressed in lung cancer tissues, and the higher expression of KCNK15-AS1 was associated with poor survival (8). On the other hand, KCNK15-AS1 was reported to be decreased in pancreatic cancer tissues, and to inhibit the migration and invasion of pancreatic cancer cells (24). Furthermore, the expression of KCNK15-AS1 was regulated by the m6A eraser AlkB homolog 5, RNA demethylase (24). Therefore, KCNK15-AS1 may have different functions in different types of cancer, and elucidation of the mechanisms associated with KCNK15-AS1 overexpression is urgently required.

The results of the present study initially confirmed the overexpression and association with the poor prognosis of KCNK15-AS1 in lung cancer. Furthermore, it was determined that the knockdown of KCNK15-AS1 significantly inhibited the proliferation of lung cancer cells and caused the downregulation of the cell cycle-associated gene CCND1 at the mRNA and protein levels.

The EGFR/AKT signaling pathway has important functions in lung carcinogenesis and certain miRNAs are known to regulate this signaling pathway. miR-133a was reported to decrease the expression of EGFR and inhibit the phosphorylation of AKT in human non-small cell lung cancer cells (25). miR-145 was reported to induce apoptosis and inhibit the migratory ability of non-small cell lung cancer cells by inhibiting the EGFR/phosphoinositide-3-kinase/AKT signaling pathway (26). The present study indicated that KCNK15-AS1 activated the EGFR/AKT signaling pathway via reducing miR-202 and miR-370.

Previous studies have indicated that miR-202 exhibits tumor suppressive functions and is downregulated in numerous types of cancer, including prostate cancer, breast cancer, osteosarcoma and lung cancer (12,2729). miR-202 has been reported to be significantly downregulated in bladder cancer tissues and cell lines, and the overexpression of miR-202 inhibited cell proliferation, colony formation, invasion and migration in vitro, as well as suppressed tumor growth in vivo; of note, miR-202 exerted its tumor suppressive functions via targeting EGFR (21). In lung cancer, miR-202 was decreased, and the overexpression of miR-202 enhanced the sensitivity of lung cancer cells to cisplatin by inactivating the Ras/mitogen-activated protein kinase signaling pathway (30). miR-202 was also demonstrated to have a tumor suppressive function in lung cancer by targeting STAT3 and CCND1 (12,13). miR-370 was downregulated in lung cancer tissues and the overexpression of miR-370 inhibited the proliferation, colony formation, migration and invasion of lung cancer cells. Furthermore, miR-370 was indicated to target EGFR and regulate its expression, and also to inhibit AKT phosphorylation (14). miR-370 also inhibited the CCND1/CCND kinase (CDK)4/CDK6 pathway by increasing p21 expression in lung cancer cells, and suppressing the progression of non-small cell lung cancer via targeting and reducing the expression of TRAF4 (11,31). In gastric cancer, the expression of miR-370 was negatively associated with EGFR, and the overexpression of miR-370 suppressed the proliferation and migration of gastric cancer cells by targeting EGFR (22). The present study indicated that KCNK15-AS1 knockdown inhibited lung cancer cell proliferation, reduced CCND1 expression and inactivated the EGFR/AKT axis via upregulating miR-202 and miR-370 by a rescue experiment.

In conclusion, the results of the present study suggested that KCNK15-AS1 promoted lung cancer cell proliferation and inactivated the EGFR/AKT signaling pathway via downregulating miR-202 and miR-370. Future studies should focus on the mechanisms of how KCNK15-AS1 regulates miR-202 and miR-370, in addition to the diagnostic and therapeutic potential of KCNK15-AS1 in lung cancer.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

JP and HP designed the study. JP, XC, HC, ZX, HW, ZS, JL and XN performed the experiments. JP, XC and HP analyzed the data. JP and HP wrote the paper.

Ethics approval and consent to participate

The present study was ethically approved by the Institutional Ethics Committee of the First People's Hospital of Yunnan Province (Kunming, China). All samples were residual specimens following diagnostic sampling, and all patients provided written informed consent for sampling and molecular analysis separately.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

lncRNA

long non-coding RNA

miRNA

microRNA

KCNK15-AS1

KCNK15 and WISP2 antisense RNA 1

EGFR

epidermal growth factor receptor

References

1 

Hao Y, Yang X, Zhang D, Luo J and Chen R: Long noncoding RNA LINC01186, regulated by TGF-β/SMAD3, inhibits migration and invasion through Epithelial-Mesenchymal-Transition in lung cancer. Gene. 608:1–12. 2017. View Article : Google Scholar : PubMed/NCBI

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. View Article : Google Scholar : PubMed/NCBI

3 

Wang Y, Liu Z, Yao B, Li Q, Wang L, Wang C, Dou C, Xu M, Liu Q and Tu K: Long non-coding RNA CASC2 suppresses epithelial-mesenchymal transition of hepatocellular carcinoma cells through CASC2/miR-367/FBXW7 axis. Mol Cancer. 16:1232017. View Article : Google Scholar : PubMed/NCBI

4 

Jiang R, Tang J, Chen Y, Deng L, Ji J, Xie Y, Wang K, Jia W, Chu WM and Sun B: The long noncoding RNA lnc-EGFR stimulates T-regulatory cells differentiation thus promoting hepatocellular carcinoma immune evasion. Nat Commun. 8:151292017. View Article : Google Scholar : PubMed/NCBI

5 

Liu YY, Chen ZH, Peng JJ, Wu JL, Yuan YJ, Zhai ET, Cai SR, He YL and Song W: Up-regulation of long non-coding RNA XLOC_010235 regulates epithelial-to-mesenchymal transition to promote metastasis by associating with Snail1 in gastric cancer. Sci Rep. 7:24612017. View Article : Google Scholar : PubMed/NCBI

6 

Wei L, Wu T, He P, Zhang JL and Wu W: LncRNA ATB promotes the proliferation and metastasis of lung cancer via activation of the p38 signaling pathway. Oncol Lett. 16:3907–3912. 2018.PubMed/NCBI

7 

Yang L, Xie N, Huang J, Huang H, Xu S, Wang Z and Cai J: SIK1-LNC represses the proliferative, migrative, and invasive abilities of lung cancer cells. Onco Targets Ther. 11:4197–4206. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Zhang X, Chi Q and Zhao Z: Up-regulation of long non-coding RNA SPRY4-IT1 promotes tumor cell migration and invasion in lung adenocarcinoma. Oncotarget. 8:51058–51065. 2017.PubMed/NCBI

9 

Wang R, Chen XF and Shu YQ: Prediction of non-small cell lung cancer metastasis-associated microRNAs using bioinformatics. Am J Cancer Res. 5:32–51. 2014.PubMed/NCBI

10 

Nymark P, Guled M, Borze I, Faisal A, Lahti L, Salmenkivi K, Kettunen E, Anttila S and Knuutila S: Integrative analysis of microRNA, mRNA and aCGH data reveals asbestos- and histology-related changes in lung cancer. Genes Chromosomes Cancer. 50:585–597. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Chen T, Gao F, Feng S, Yang T and Chen M: MicroRNA-370 inhibits the progression of non-small cell lung cancer by downregulating oncogene TRAF4. Oncol Rep. 34:461–468. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Zhao Z, Lv B, Zhang L, Zhao N and Lv Y: miR-202 functions as a tumor suppressor in non-small cell lung cancer by targeting STAT3. Mol Med Rep. 16:2281–2289. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Jiang J, Huang J, Wang XR and Quan YH: MicroRNA-202 induces cell cycle arrest and apoptosis in lung cancer cells through targeting cyclin D1. Eur Rev Med Pharmacol Sci. 20:2278–2284. 2016.PubMed/NCBI

14 

Liu X, Huang YG, Jin CG, Zhou YC, Chen XQ, Li J, Chen Y, Li M, Yao Q, Li K, et al: MicroRNA-370 inhibits the growth and metastasis of lung cancer by down-regulating epidermal growth factor receptor expression. Oncotarget. 8:88139–88151. 2017.PubMed/NCBI

15 

Yong-Hao Y, Xian-Guo W, Ming X and Jin-Ping Z: Expression and clinical significance of miR-139-5p in non-small cell lung cancer. J Int Med Res. 47:867–874. 2019. View Article : Google Scholar : PubMed/NCBI

16 

In H, Solsky I, Palis B, Langdon-Embry M, Ajani J and Sano T: Validation of the 8th edition of the AJCC TNM staging system for gastric cancer using the National Cancer Database. Ann Surg Oncol. 24:3683–3691. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Varnholt H, Drebber U, Schulze F, Wedemeyer I, Schirmacher P, Dienes HP and Odenthal M: MicroRNA gene expression profile of hepatitis C virus-associated hepatocellular carcinoma. Hepatology. 47:1223–1232. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Chen C, Zhang Z, Li J and Sun Y: SNHG8 is identified as a key regulator in non-small-cell lung cancer progression sponging to miR-542-3p by targeting CCND1/CDK6. Onco Targets Ther. 11:6081–6090. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Liu F, Shangli Z and Hu Z: CAV2 promotes the growth of renal cell carcinoma through the EGFR/PI3K/Akt pathway. Onco Targets Ther. 11:6209–6216. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Gao J, Qiu X, Xi G, Liu H, Zhang F, Lv T and Song Y: Downregulation of GSDMD attenuates tumor proliferation via the intrinsic mitochondrial apoptotic pathway and inhibition of EGFR/Akt signaling and predicts a good prognosis in non-small cell lung cancer. Oncol Rep. 40:1971–1984. 2018.PubMed/NCBI

21 

Zhang L, Xu J, Yang G, Li H and Guo X: miR-202 inhibits cell proliferation, migration, and invasion by targeting epidermal growth factor receptor in human bladder cancer. Oncol Res. 26:949–957. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Ning T, Zhang H, Wang X, Li S, Zhang L, Deng T, Zhou L, Liu R, Wang X, Bai M, et al: miR-370 regulates cell proliferation and migration by targeting EGFR in gastric cancer. Oncol Rep. 38:384–392. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Zou Y, Zhang B, Mao Y, Zhang H and Hong W: Long non-coding RNA OECC promotes cell proliferation and metastasis through the PI3K/Akt/mTOR signaling pathway in human lung cancer. Oncol Lett. 18:3017–3024. 2019.PubMed/NCBI

24 

He Y, Hu H, Wang Y, Yuan H, Lu Z, Wu P, Liu D, Tian L, Yin J, Jiang K and Miao Y: ALKBH5 inhibits pancreatic cancer motility by decreasing long non-coding RNA KCNK15-AS1 methylation. Cell Physiol Biochem. 48:838–846. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Guo N, Zhao Y, Zhang W, Li S, Li S and Yu J: MicroRNA-133a downregulated EGFR expression in human non-small cell lung cancer cells via AKT/ERK signaling. Oncol Lett. 16:6045–6050. 2018.PubMed/NCBI

26 

Li B, Ding CM, Li YX, Peng JC, Geng N and Qin WW: MicroRNA145 inhibits migration and induces apoptosis in human non-small cell lung cancer cells through regulation of the EGFR/PI3K/AKT signaling pathway. Oncol Rep. 40:2944–2954. 2018.PubMed/NCBI

27 

Zhang S, Cai J, Xie W, Luo H and Yang F: miR-202 suppresses prostate cancer growth and metastasis by targeting PIK3CA. Exp Ther Med. 16:1499–1504. 2018.PubMed/NCBI

28 

Gao S, Cao C, Dai Q, Chen J and Tu J: miR-202 acts as a potential tumor suppressor in breast cancer. Oncol Lett. 16:1155–1162. 2018.PubMed/NCBI

29 

Li C, Ma D, Yang J, Lin X and Chen B: miR-202-5p inhibits the migration and invasion of osteosarcoma cells by targeting ROCK1. Oncol Lett. 16:829–834. 2018.PubMed/NCBI

30 

Sun W, Ping W, Tian Y, Zou W, Liu J and Zu Y: miR-202 enhances the anti-tumor effect of cisplatin on non-small cell lung cancer by targeting the Ras/MAPK pathway. Cell Physiol Biochem. 51:2160–2171. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Li C, Ge Q, Liu J, Zhang Q, Wang C, Cui K and Chen Z: Effects of miR-1236-3p and miR-370-5p on activation of p21 in various tumors and its inhibition on the growth of lung cancer cells. Tumour Biol. 39:10104283177108242017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Peng J, Chen XL, Cheng HZ, Xu ZY, Wang H, Shi ZZ, Liu J, Ning XG and Peng H: Silencing of KCNK15‑AS1 inhibits lung cancer cell proliferation via upregulation of miR‑202 and miR‑370. Oncol Lett 18: 5968-5976, 2019
APA
Peng, J., Chen, X., Cheng, H., Xu, Z., Wang, H., Shi, Z. ... Peng, H. (2019). Silencing of KCNK15‑AS1 inhibits lung cancer cell proliferation via upregulation of miR‑202 and miR‑370. Oncology Letters, 18, 5968-5976. https://doi.org/10.3892/ol.2019.10944
MLA
Peng, J., Chen, X., Cheng, H., Xu, Z., Wang, H., Shi, Z., Liu, J., Ning, X., Peng, H."Silencing of KCNK15‑AS1 inhibits lung cancer cell proliferation via upregulation of miR‑202 and miR‑370". Oncology Letters 18.6 (2019): 5968-5976.
Chicago
Peng, J., Chen, X., Cheng, H., Xu, Z., Wang, H., Shi, Z., Liu, J., Ning, X., Peng, H."Silencing of KCNK15‑AS1 inhibits lung cancer cell proliferation via upregulation of miR‑202 and miR‑370". Oncology Letters 18, no. 6 (2019): 5968-5976. https://doi.org/10.3892/ol.2019.10944