microRNA-363-3p inhibits cell growth and invasion of non‑small cell lung cancer by targeting HMGA2

Retraction in: /10.3892/mmr.2023.13034

  • Authors:
    • Chuanfu Jiang
    • Yang Cao
    • Ting Lei
    • Yu Wang
    • Junfeng Fu
    • Ze Wang
    • Zhenyang Lv
  • View Affiliations

  • Published online on: November 22, 2017     https://doi.org/10.3892/mmr.2017.8131
  • Pages: 2712-2718
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer is the second most common cancer and is the leading cause of cancer-related death worldwide. For decades, increasing evidence revealed that microRNAs may contribute to non‑small cell lung cancer (NSCLC) carcinogenesis and progression and could provide novel therapeutic targets for treatments of patients with NSCLC. Accumulated studies indicate that microRNA (miR)‑363‑3p serves important roles in tumorigenesis and tumor development; however, the role of miR‑363‑3p in NSCLC is still unclear. The current study reported that miR‑363‑3p exhibited reduced expression in NSCLC tissues and cell lines. Reduced miR‑363‑3p expression was correlated with tumor node metastasis classification and distant metastasis of NSCLC patients. Notably, miR‑363‑3p re‑expression significantly suppressed cell proliferation and invasion of NSCLC. Furthermore, bioinformatics analysis, luciferase reporter assay, reverse transcription‑quantitative polymerase chain reaction and western blotting indicated that (high mobility group AT-hook 2) HMGA2 was a direct target gene of miR‑363‑3p. HMGA2 was increased in NSCLC tissues and inversely associated with HMGA2 expression. Moreover, HMGA2 underexpression had similar effects to miR‑363‑3p overexpression in NSCLC cells. Thus, the current study suggested that miR‑363‑3p may act as a tumor suppressor in NSCLC and that the miR‑363‑3p could be investigated as a therapeutic target for the patients with this disease.

Introduction

Lung cancer is the second most common cancer and the leading cause of cancer-related death worldwide (1). Non-small cell lung cancer (NSCLC), the most common type, accounts for ~85% of total cases of lung cancer (2). NSCLC can be subdivided into four histological types: Squamous cell carcinoma, adenocarcinoma, adenosquamous cell carcinoma and large cell carcinoma (3). Thus far, studies have demonstrated that many risk factors are involved in NSCLC, including environmental pollution, smoking and occupational carcinogens (47). Currently, considerable advances have been developed in the early detection and therapeutic strategies for patients with NSCLC; however, NSCLC is still refractory to treatment (8). In all, ~70% of patients with this disease are at locally advanced stages or have distant metastasis at the time of diagnosis (9). The overall 5-year overall survival rate for NSCLC is only 17.1% (10). Therefore, it is important to elucidate the mechanisms underlying tumorigenesis and tumor development in NSCLC, and to investigate novel therapeutic methods for NSCLC cases.

For decades, an increasing number of studies revealed that microRNAs (miRs) may contribute to NSCLC carcinogenesis and progression and could provide novel therapeutic targets for treatments of patients with NSCLC. miRs are a family of endogenous, non-protein-coding and short RNAs that range in size from 17 to 23 nucleotides (11). miRs act as post-transcriptional regulators of gene expression through imperfect binding to the 3′untranslated regions (3′UTRs) of their target genes, and leading to translational inhibition or mRNA degradation (12,13). miRs serve roles in a great deal of important physiological processes, such as cell growth, cell cycle, apoptosis, differentiation, migration and metastasis (14). miRs have stable molecular structure and therefore can be detected in body fluid and tissues (15). Previous studies indicated that expression levels of miRs may be significantly correlated with diagnosis, treatment and prognosis in human cancer (1618). Accumulated evidence has demonstrated that miRs are abnormally expressed in various kinds of human cancer, such as bladder cancer (19), gastric cancer (20), colorectal cancer (21), glioma (22) and NSCLC (23). The aberrantly overexpressed miRs can act as oncogenes through downregulation of tumor suppressor genes, whereas lowly expressed miRs can function as tumor suppressors by negatively regulation of oncogenes (24).

miR-363-3p has been investigated in many cancers (2527). However, the role of miR-363-3p in NSCLC is still unclear. The objective of the present study was to elucidate the expression and biological roles of miR-363-3p in NSCLC, and to investigate its underlying molecular mechanisms.

Materials and methods

Clinical specimens and cell culture

The protocol of the present study was approved by the Ethical Committee of The Second Affiliated Hospital of Dalian Medical University (Dalian, China). All patients provided written informed consent. Primary NSCLC tissues and paired adjacent normal lung tissues were obtained from 57 patients who had been treated with surgery resection at Department of Thoracic Surgery, The Second Affiliated Hospital of Dalian Medical University (Dalian, China) between January 2012 and July 2014. None of these patients received radiotherapy or chemotherapy before surgery. All tissues were immediately snap-frozen in liquid nitrogen and stored at −80°C until use.

Human NSCLC cell lines (SK-MES-1, H23, H522, SPC-A1, A549) and a normal human bronchial epithelial cell line (16HBE) were purchased from American Type Culture Collection (Manassas, VA, USA). Cells were maintained in Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin G (Gibco; Thermo Fisher Scientific, Inc.) and 100 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a humidified atmosphere containing 5% CO2.

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

Total RNA was extracted form tissues (1 g) or cells (1.5×106) with TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) following to the manufacturer's protocols. The concentration of total RNA was measured using an ND-2000 spectrophotometer (NanoDrop; Thermo Fisher Scientific, Inc., Wilmington, DE, USA). Total RNA was reverse transcribed into cDNA using M-MLV reverse transcriptase (Promega Corporation, Madison, WI, USA), according to the manufacturer's protocols.

The expression of miR-363-3p and (high mobility group AT-hook 2) HMGA2 mRNA was quantified by using SYBR Premix Ex Taq™ kits (Takara Bio, Inc., Otsu, Japan) on an Applied Biosystems® 7900HT Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocols. The cycling conditions for qPCR were as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. U6 and GADPH were used as reference for miR-363-3p and HMGA2 mRNA, respectively. Each sample was analyzed in triplicate. The data were calculated using the relative quantification method (2−ΔΔCq) (28).

Cell transfection

Mature miR-363-3p mimic, miR mimic negative control (miR-NC), small interfering RNA targeting HMGA2 (si-HMGA2) and its negative control (si-NC) were synthesized by Shanghai GenePharma Co., Ltd. (Shanghai, China). For functional analysis, cells were transfected with miR-363-3p mimics, miR-NC, si-HMGA2 or si-NC using Lipofectamine 2,000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

Cell counting kit (CCK)-8 assay

Cell proliferation was determined using the CCK8 assay (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) according to the manufacturer's protocol. At 24 h following transfection, cells were harvested and re-suspended in culture medium. A total of 3×103 cells were seeded in the 96-well plates and incubated at 37°C in a humidified air atmosphere of 5% CO2. Cell proliferation was detected following 24, 48, 72 and 96 h of incubation. Briefly, 10 µl CCK8 solution was added into each well and then incubated at 37°C for another 4 h. The absorbance at 450 nm was measured with an ELISA reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Each assay was performed in triplicate and repeated at least three times.

Cell invasion assay

For cell invasion assay, transwell chambers (BD Biosciences, Franklin Lakes, NJ, USA) were coated with Matrigel (BD Biosciences). Following 72 h of transfection, cells were harvested and re-suspended in FBS-free culture medium. A total of 1×105 cells in 100 µl FBS-free medium were added into the upper chamber. As a chemoattractant, 500 µl culture medium containing 20% FBS was added to the lower chamber. Chambers were then incubated for 48 h at 37°C in a humidified air atmosphere of 5% CO2. Cells that did not invade through the pores of the membranes were carefully wiped away with cotton wool. The invaded cells were fixed with 95% methanol, stained with 0.5% crystal violet (Beyotime Institute of Biotechnology, Haimen, China), and washed with PBS. Finally, the invaded cells were photographed and counted with an inverted microscope (CKX41; Olympus Corporation, Tokyo, Japan).

Bioinformatics analysis

TargetScan (http://www.targetscan.org/) and PicTar (http://pictar.mdcberlin.de/) were used to explore the potential target genes of miR-363-3p.

Luciferase reporter assay

To determine whether HMGA2 was a direct target of miR-363-3p, a luciferase reporter assay was performed. Luciferase reporter plasmids (pmirGLO-HMGA2-3′UTR WT and pmirGLO-HMGA2-3′UTR MUT) were synthesized and purified by Shanghai GenePharma Co., Ltd. HEK293T cells were seeded in 12-well plates at a density of 60–70% confluence and transfected with pmirGLO-HMGA2-3′UTR WT or pmirGLO-HMGA2-3′UTR MUT along with miR-363-3p mimics or miR-NC using Lipofectamine 2000. Following 48 h of transfection, the firefly and renilla luciferase activities were detected using a Dual-Luciferase® Reporter Assay system (Promega Corporation). Firefly luciferase activity served as an internal control. Each sample was analyzed in triplicate and the assay was repeated three times.

Western blotting

Following 72 h of transfection, cellular protein was extracted using radioimmunoprecipitation assay lysis buffer (50 mM Tris-HCl, pH 7.4; 1% NP-40; 0.25% Na-deoxycholate; 150 mM NaCl; 1 mM EDTA; 1 mM PMSF; aprotinin, leupeptin, pepstatin: 1 µg/ml each; 1 mM Na3VO4; 1 mM NaF) containing protease and phosphatase inhibitors (Roche Diagnostics GmbH, Mannheim, Germany). The concentration of total protein was measured by using a bicinchoninic acid assay kit (Nanjing KeyGen Biotech. Co., Ltd., Nanjing, China). Equal amounts of protein (30 µg) were separated by 10% SDS-PAGE electrophoresis and electrotransferred to polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). Membranes were blocked in TBS containing 0.05% Tween-20 (TBST; Beyotime Institute of Biotechnology, Haimen, China) containing 5% non-fat dry milk for 1 h at room temperature and incubated with mouse anti-human HMGA2 monoclonal primary antibody (ab184616; 1:1,000; Abcam, Cambridge, UK) and mouse anti-human monoclonal GADPH antibody (ab184616; 1:1,000; Abcam), at 4°C overnight. The membranes were then probed with goat anti-mouse horseradish peroxidase conjugated secondary antibody (ab6785; 1:5,000; Abcam) at room temperature for 1 h. Finally, the protein bands were developed with enhanced chemiluminescence reagents (Pierce; Thermo Fisher Scientific, Inc.). GADPH was use as a loading control. ImageJ version 1.49 (National Institutes of Health, Bethesda, MD) was used to quantify protein expression.

Statistical analysis

All data were presented as the mean ± standard deviation. SPSS software version, 15.0 (SPSS Inc., Chicago, IL, USA) was used for statistical analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-363-3p is downregulated in NSCLC and its association with clinicopathological factors

The present study measured miR-363-3p expression in NSCLC tissues and corresponding adjacent normal lung tissues using RT-qPCR. As presented in Fig. 1A, miR-363-3p was lower in NSCLC tissues than corresponding adjacent normal tissues (P<0.05). The expression levels of miR-363-3p in NSCLC cell lines were also determined. The results demonstrated that miR-363-3p expression levels were reduced in NSCLC cell lines (SK-MES-1, H23, H522, SPC-A1, A549) compared with 16HBE cell line (P<0.05; Fig. 1B).

The study then examined whether miR-363-3p expression level is associated with clinicopathological factors of NSCLC patients. The statistical analysis demonstrated that miR-363-3p expression was significantly associated with the tumor node metastasis classification and distant metastasis (presented in Table I). These results suggested that miR-363-3p may serve important functions in NSCLC.

Table I.

Correlation between miR-363-3p expression and clinicopathological factors of patients with non-small cell lung cancer.

Table I.

Correlation between miR-363-3p expression and clinicopathological factors of patients with non-small cell lung cancer.

miR-363-3p expression

Clinical factorsCases, nLowHighP-value
Gender 0.930
  Male2415  9
  Female332112
Age, years 0.503
  <60 years321913
  ≥60 years2517  8
Smoking history, years 0.341
  <10 years1511  4
  ≥10 years422517
Tumor differentiation, grade 0.816
  I–II312011
  III–IV261610
TNM classification, stage 0.005
  I–II271215
  III–IV3024  6
Distant metastasis 0.001
  Negative321418
  Positive2522  3

[i] miR, microRNA; TNM classification, tumor-node-metastasis classification.

miR-363-3p inhibits cell proliferation and invasion of NSCLC

To investigate the biological roles of miR-363-3p in NSCLC, miR-363-3p mimics was introduced into H522 and A549 cells to increase its expression (Fig. 2A; P<0.05). Following transfection, CCK8 and cell invasion assay were performed to examine the effects of miR-363-3p overexpression on cell proliferation and invasion, respectively. As reported in Fig. 2B, upregulation of miR-363-3p inhibited H522 and A549 cell proliferation (P<0.05). The results of cell invasion assay indicated that restoration expression of miR-363-3p evidently decreased the invasion ability of H522 and A549 cells (P<0.05; Fig. 2C). These results suggested that miR-363-3p acts as a tumor suppressor in NSCLC.

HMGA2 is a direct target of miR-363-3p

The present study further explored the mechanisms underlying the tumor suppressive roles of miR-363-3p in NSCLC. Bioinformatics analysis was performed to identify the potential targets of miR-363-3p. As indicated in Fig. 3A, HMGA2 was predicted to be a candidate target gene of miR-363-3p. Luciferase reporter assay was then used to determine whether the 3′UTR of HMGA2 could be directly targeted by miR-363-3p. The results identified that ectopic expression of miR-363-3p suppressed the luciferase activities of pmirGLO-HMGA2-3′UTR WT (P<0.05), but not pmirGLO-HMGA2-3′UTR MUT, in HEK293T cells (Fig. 3B).

A further experiment was performed to measure HMGA2 expression in NSCLC tissues and corresponding adjacent normal lung tissues using RT-qPCR. As reported in Fig. 3C, HMGA2 mRNA was significantly upregulated in NSCLC tissues (P<0.05). Spearman's correlation analysis indicated that miR-363-3p expression was negative correlated with HMGA2 mRNA level in NSCLC tissues (Fig. 3D; r=−0.5640; P<0.01).

RT-qPCR and western blotting were performed to investigate whether HMGA2 was negatively modulated by miR-363-3p in H522 and A549. As demonstrated in Fig. 3E, miR-363-3p re-expression reduced HMGA2 mRNA expression in both H522 and A549 cells (P<0.05). The data of western blotting showed that miR-363-3p overexpression significantly decreased HMGA2 protein expression in H522 and A549 cells (Fig. 3F, P<0.05). These findings suggested that HMGA2 is a direct target gene of miR-363-3p.

HMGA2 knockdown has similar effects to miR-363-3p overexpression in NSCLC cells

To investigate whether the tumor suppressive roles of miR-363-3p in NSCLC cells were mediated by HMGA2, RNA interference experiments were performed. H522 and A549 cells were transfected with si-HMGA2 or si-NC. Following transfection, HMGA2 protein was downregulated in H522 and A549 cells transfected with si-HMGA2 (Fig. 4A, P<0.05). Following, CCK8 assay and cell invasion assay were conducted in H522 and A549 cells to evaluate its effects on cell proliferation and invasion. As shown in Fig. 4B and C, HMGA2 knockdown suppressed the proliferation (P<0.05) and invasion (P<0.05) of H522 and A549 cells. These results suggested the biological roles of HMGA2 underexpression were similar to the effects exerted by miR-363-3p in NSCLC cells, indicating that HMGA2 is a functional target of miR-363-3p in NSCLC.

Discussion

Abnormally expressed miRs have been verified to be involved in initiation and progression of various kinds of human cancer; however, their biological roles and molecular mechanism remains to be elucidated (29). The current study reported that expression levels of miR-363-3p were reduced in NSCLC tumor tissues and cell lines. Decreased miR-363-3p expression was obviously associated with aggressive clinicopathological features. In addition, resumption expression of miR-363-3p significantly suppressed cell proliferation and invasion of NSCLC. Moreover, HMGA2 was identified as a direct target gene of miR-363-3p. To the best of the authors' knowledge, the present study is the first to investigate the expression pattern, clinical significance, biological roles and molecular mechanism of miR-363-3p in NSCLC.

miR-363-3p, derived from the miR-106a-363 cluster on chromosome X, has been identified as a tumor suppressor in several kinds of human cancer. For instance, in colorectal cancer, miR-363-3p exhibited a reduced expression in tumor tissues. In addition, upregulation of miR-363-3p inhibited colorectal carcinogenesis through directly targeting the GATA6/Lgr5 pathway (25). Moreover, in vitro and in vivo experiments revealed that miR-363-3p underexpression enhanced cell migration, invasion and EMT of colorectal cancer via blockade of SOX4 (26). Zhou et al (27) reported that miR-363-3p targeted S1PR1 to decrease hepatocellular carcinoma cells proliferation by directly targeting. Ou et al (30) reported that miR-363-3p was lower in HCC tissues treated with cisplatin-based chemotherapy. miR-363-3p overexpression overcame cisplatin resistance in cisplatin-resistant HepG2 cells through downregulation of Mcl-1. In osteosarcoma, miR-363-3p expression level was decreased in tumor tissues and cell lines. Reduced miR-363-3p expression was correlated with tumor size, clinical stage and distant metastasis. miR-363-3p re-expression suppressed cell growth and metastasis through downregulation of MAP2K4 (31).

In contrast to the aforementioned antitumor properties, miR-363-3p also functions as an oncogene in glioma, prostate cancer and gastric cancer. In glioma, miR-363-3p was significantly increased in both tumor samples and cell lines, and significantly associated with tumor grading. High expression of miR-363-3p enhanced cell survival and proliferation of glioma (32). Chen et al (33) suggested that miR-363-3p was higher in prostate cancer cells. Ectopic expression of miR-363-3p promoted cell proliferation and transformation properties in addition to promoting EMT by negative regulation of c-myc. In gastric cancer, exogenous miR-363-3p enhanced cell growth, viability, progression, EMT and tumorsphere formation of gastric cancer via directly targeting MBP-1 (34). Zhang et al (35) also demonstrated that miR-363-3p was upregulated in gastric cancer tissues. Restoration expression of miR-363-3p promotes cell proliferation and chemo-resistance in gastric cancer via negatively regulation of FBW7. Taken together, these findings suggested that the expression pattern, functional roles of miR-363-3p in human malignancies may be multifaceted which mainly depending on the involved tissue and their target genes.

Currently, it is well established that miRs exert their oncogenic or tumor suppressor functions through negatively regulation of their target genes (29). In the present study, HMGA2 was subsequently demonstrated to be a novel direct target of miR-363-3p. Firstly, bioinformatic analysis indicated that HMGA2 was a candidate target of miR-363-3p. Secondly, luciferase reporter assays revealed that the 3′UTR of HMGA2 could be directly targeted by miR-363-3p. Thirdly, HMGA2 mRNA was significantly upregulated in NSCLC tissues and inverse correlated with miR-363-3p expression. Upregulation of miR-363-3p decreased HMGA2 expression in NSCLC cells at both mRNA and protein level. Finally, the biological roles of HMGA2 underexpression were similar to the effects exerted by miR-363-3p in NSCLC cells, also indicating that HMGA2 is a functional target of miR-363-3p in NSCLC.

Identification of miR-363-3p target gene is critical for understanding its biological roles in NSCLC occurrence and tumor development.

HMGA2, a member of the high mobility group A proteins, is a non-histone chromatin-binding protein (36). Previous studies reported that HMGA2 was highly expressed in many types of human cancer, such as bladder cancer (37), breast cancer (38), colorectal cancer (39) and prostate cancer (40). In NSCLC, HMGA2 was significantly increased in NSCLC tissues. High expression level of HMGA2 was associated with lymph node metastasis of NSCLC patients. Besides, HMGA2 was identified as an independent prognostic factor for patients with NSCLC (41). Moreover, HMGA2 was demonstrated to be involved in NSCLC progression and metastasis (42). The current study revealed that HMGA2 expression was significantly upregulated in NSCLC tissues. HMGA2 knockdown suppressed cell proliferation and invasion of NSCLC. Taken together, these findings suggested that miR-363-3p/HMGA2 based targeted therapy could be a novel therapeutic strategy for NSCLC patients.

In conclusion, the present study provided novel evidences that miR-363-3p expression level was significantly reduced in NSCLC and was associated with tumor development. The tumor suppressive roles of miR-363-3p were also identified in the functional analysis, and HMGA2 was confirmed as a direct target of miR-363-3p. These findings suggested that miR-363-3p could be investigated as a potential therapeutic target for NSCLC cases.

References

1 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Debevec L and Debeljak A: Multidisciplinary management of lung cancer. J Thorac Oncol. 2:5772007. View Article : Google Scholar : PubMed/NCBI

3 

Spira A and Ettinger DS: Multidisciplinary management of lung cancer. N Engl J Med. 350:379–392. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Boffetta P and Nyberg F: Contribution of environmental factors to cancer risk. Br Med Bull. 68:71–94. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Didkowska J, Manczuk M, McNeill A, Powles J and Zatonski W: Lung cancer mortality at ages 35–54 in the European Union: Ecological study of evolving tobacco epidemics. BMJ. 331:189–191. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Ridge CA, McErlean AM and Ginsberg MS: Epidemiology of lung cancer. Semin Intervent Radiol. 30:93–98. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Paliogiannis P, Attene F, Cossu A, Budroni M, Cesaraccio R, Tanda F, Trignano M and Palmieri G: Lung cancer epidemiology in North Sardinia, Italy. Multidiscip Respir Med. 8:452013. View Article : Google Scholar : PubMed/NCBI

8 

Søes S, Daugaard IL, Sørensen BS, Carus A, Mattheisen M, Alsner J, Overgaard J, Hager H, Hansen LL and Kristensen LS: Hypomethylation and increased expression of the putative oncogene ELMO3 are associated with lung cancer development and metastases formation. Oncoscience. 1:367–374. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Reungwetwattana T, Weroha SJ and Molina JR: Oncogenic pathways, molecularly targeted therapies, and highlighted clinical trials in non-small-cell lung cancer (NSCLC). Clin Lung Cancer. 13:252–266. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Sánchez de Cos J, Sojo González MA, Montero MV, Pérez Calvo MC, Vicente MJ and Valle MH: Non-small cell lung cancer and silent brain metastasis. Survival and prognostic factors. Lung Cancer. 63:140–145. 2009. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Lai EC: Micro RNAs are complementary to 3′UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet. 30:363–364. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Iorio MV and Croce CM: MicroRNA dysregulation in cancer: Diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med. 4:143–159. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, et al: Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 105:pp. 10513–10518. 2008; View Article : Google Scholar : PubMed/NCBI

16 

Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, et al: Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 18:997–1006. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Vychytilova-Faltejskova P, Radova L, Sachlova M, Kosarova Z, Slaba K, Fabian P, Grolich T, Prochazka V, Kala Z, Svoboda M, et al: Serum-based microRNA signatures in early diagnosis and prognosis prediction of colon cancer. Carcinogenesis. 37:941–950. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Li P, Liu H, Wang Z, He F, Wang H, Shi Z, Yang A and Ye J: MicroRNAs in laryngeal cancer: Implications for diagnosis, prognosis and therapy. Am J Transl Res. 8:1935–1944. 2016.PubMed/NCBI

19 

Shin SS, Park SS, Hwang B, Moon B, Kim WT, Kim WJ and Moon SK: MicroRNA-892b influences proliferation, migration and invasion of bladder cancer cells by mediating the p19ARF/cyclin D1/CDK6 and Sp-1/MMP-9 pathways. Oncol Rep. 36:2313–2320. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Qiu X, Zhu H, Liu S, Tao G, Jin J, Chu H, Wang M, Tong N, Gong W, Zhao Q, et al: Expression and prognostic value of microRNA-26a and microRNA-148a in gastric cancer. J Gastroenterol Hepatol. 32:819–827. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Chandrasekaran KS, Sathyanarayanan A and Karunagaran D: MicroRNA-214 suppresses growth, migration and invasion through a novel target, high mobility group AT-hook 1, in human cervical and colorectal cancer cells. Br J Cancer. 115:741–751. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Feng F, Kuai D, Wang H, Li T, Miao W, Liu Y and Fan Y: Reduced expression of microRNA-497 is associated with greater angiogenesis and poor prognosis in human gliomas. Hum Pathol. 58:47–53. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Huang T, She K, Peng G, Wang W, Huang J, Li J, Wang Z and He J: MicroRNA-186 suppresses cell proliferation and metastasis through targeting MAP3K2 in non-small cell lung cancer. Int J Oncol. 49:1437–1444. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Esquela-Kerscher A and Slack FJ: Oncomirs-microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Tsuji S, Kawasaki Y, Furukawa S, Taniue K, Hayashi T, Okuno M, Hiyoshi M, Kitayama J and Akiyama T: The miR-363-GATA6-Lgr5 pathway is critical for colorectal tumourigenesis. Nat Commun. 5:31502014. View Article : Google Scholar : PubMed/NCBI

26 

Hu F, Min J, Cao X, Liu L, Ge Z, Hu J and Li X: MiR-363-3p inhibits the epithelial-to-mesenchymal transition and suppresses metastasis in colorectal cancer by targeting Sox4. Biochem Biophys Res Commun. 474:35–42. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Zhou P, Huang G, Zhao Y, Zhong D, Xu Z, Zeng Y, Zhang Y, Li S and He F: MicroRNA-363-mediated downregulation of S1PR1 suppresses the proliferation of hepatocellular carcinoma cells. Cell Signal. 26:1347–1354. 2014. View Article : Google Scholar : PubMed/NCBI

28 

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. View Article : Google Scholar : PubMed/NCBI

29 

Hou Y, Zhen J, Xu X, Zhen K, Zhu B, Pan R and Zhao C: miR-215 functions as a tumor suppressor and directly targets ZEB2 in human non-small cell lung cancer. Oncol Lett. 10:1985–1992. 2015.PubMed/NCBI

30 

Ou Y, Zhai D, Wu N and Li X: Downregulation of miR-363 increases drug resistance in cisplatin-treated HepG2 by dysregulating Mcl-1. Gene. 572:116–122. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Li X, Liu X, Fang J, Li H and Chen J: microRNA-363 plays a tumor suppressive role in osteosarcoma by directly targeting MAP2K4. Int J Clin Exp Med. 8:20157–20167. 2015.PubMed/NCBI

32 

Conti A, Romeo SG, Cama A, La Torre D, Barresi V, Pezzino G, Tomasello C, Cardali S, Angileri FF, Polito F, et al: MiRNA expression profiling in human gliomas: Upregulated miR-363 increases cell survival and proliferation. Tumour Biol. 37:14035–14048. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Chen Y, Lu X, Wu B, Su Y, Li J and Wang H: MicroRNA 363 mediated positive regulation of c-myc translation affect prostate cancer development and progress. Neoplasma. 62:191–198. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Hsu KW, Wang AM, Ping YH, Huang KH, Huang TT, Lee HC, Lo SS, Chi CW and Yeh TS: Downregulation of tumor suppressor MBP-1 by microRNA-363 in gastric carcinogenesis. Carcinogenesis. 35:208–217. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Zhang PF, Sheng LL, Wang G, Tian M, Zhu LY, Zhang R, Zhang J and Zhu JS: miR-363 promotes proliferation and chemo-resistance of human gastric cancer via targeting of FBW7 ubiquitin ligase expression. Oncotarget. 7:35284–35292. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Zhuo HC, Song YF, Ye J, Lai GX and Liu DL: MicroRNA-154 functions as a tumor suppressor and directly targets HMGA2 in human non-small cell lung cancer. Genet Mol Res. 15:2016. View Article : Google Scholar

37 

Yang GL, Zhang LH, Bo JJ, Hou KL, Cai X, Chen YY, Li H, Liu DM and Huang YR: Overexpression of HMGA2 in bladder cancer and its association with clinicopathologic features and prognosis HMGA2 as a prognostic marker of bladder cancer. Eur J Surg Oncol. 37:265–271. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Sun M, Song CX, Huang H, Frankenberger CA, Sankarasharma D, Gomes S, Chen P, Chen J, Chada KK, He C and Rosner MR: HMGA2/TET1/HOXA9 signaling pathway regulates breast cancer growth and metastasis. Proc Natl Acad Sci USA. 110:pp. 9920–9925. 2013; View Article : Google Scholar : PubMed/NCBI

39 

Wang X, Liu X, Li AY, Chen L, Lai L, Lin HH, Hu S, Yao L, Peng J, Loera S, et al: Overexpression of HMGA2 promotes metastasis and impacts survival of colorectal cancers. Clin Cancer Res. 17:2570–2580. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Winkler S, Murua Escobar H, Meyer B, Simon D, Eberle N, Baumgartner W, Loeschke S, Nolte I and Bullerdiek J: HMGA2 expression in a canine model of prostate cancer. Cancer Genet Cytogenet. 177:98–102. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Wu Y, Song Y and Liu H: Expression and its clinical significance of HMGA2 in the patients with non-small cell lung cancer. Zhongguo Fei Ai Za Zhi. 11:377–381. 2008.(In Chinese). PubMed/NCBI

42 

Kumar MS, Armenteros-Monterroso E, East P, Chakravorty P, Matthews N, Winslow MM and Downward J: HMGA2 functions as a competing endogenous RNA to promote lung cancer progression. Nature. 505:212–217. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang C, Cao Y, Lei T, Wang Y, Fu J, Wang Z and Lv Z: microRNA-363-3p inhibits cell growth and invasion of non‑small cell lung cancer by targeting HMGA2 Retraction in /10.3892/mmr.2023.13034. Mol Med Rep 17: 2712-2718, 2018
APA
Jiang, C., Cao, Y., Lei, T., Wang, Y., Fu, J., Wang, Z., & Lv, Z. (2018). microRNA-363-3p inhibits cell growth and invasion of non‑small cell lung cancer by targeting HMGA2 Retraction in /10.3892/mmr.2023.13034. Molecular Medicine Reports, 17, 2712-2718. https://doi.org/10.3892/mmr.2017.8131
MLA
Jiang, C., Cao, Y., Lei, T., Wang, Y., Fu, J., Wang, Z., Lv, Z."microRNA-363-3p inhibits cell growth and invasion of non‑small cell lung cancer by targeting HMGA2 Retraction in /10.3892/mmr.2023.13034". Molecular Medicine Reports 17.2 (2018): 2712-2718.
Chicago
Jiang, C., Cao, Y., Lei, T., Wang, Y., Fu, J., Wang, Z., Lv, Z."microRNA-363-3p inhibits cell growth and invasion of non‑small cell lung cancer by targeting HMGA2 Retraction in /10.3892/mmr.2023.13034". Molecular Medicine Reports 17, no. 2 (2018): 2712-2718. https://doi.org/10.3892/mmr.2017.8131