Open Access

MicroRNA‑30a suppresses the proliferation, migration and invasion of human renal cell carcinoma cells by directly targeting ADAM9

Retraction in: /10.3892/ol.2024.14232

  • Authors:
    • Lining Jiang
    • Yabin Liu
    • Can Ma
    • Binghui Li
  • View Affiliations

  • Published online on: June 20, 2018     https://doi.org/10.3892/ol.2018.8999
  • Pages: 3038-3044
  • Copyright: © Jiang 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

An increasing number of studies reported that microRNA (miR)‑30a was dysregulated in several types of human cancer and may contribute to cancer carcinogenesis and progression. However, its expression and roles in renal cell carcinoma (RCC) remain unknown. In the present study, reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was performed to quantify miR‑30a expression in RCC tissues and cell lines. The cell counting kit‑8 assay, migration and invasion assays were used to evaluate the roles of miR‑30a on the proliferation, migration and invasion of RCC cells. The target gene of miR‑30a was identified by luciferase reporter assays, RT‑qPCR and western blotting. The results indicated that miR‑30a was downregulated in RCC tissues and cell lines compared with corresponding noncancerous tissues and normal renal cell line, respectively. Re‑expression of miR‑30a inhibited the proliferation, migration and invasion of RCC cells. Additionally, ADAM metallopeptidase domain 9 (ADAM9) was validated as a direct target of miR‑30a. Furthermore, the knockdown of ADAM9 by small interfering RNAs was able to mimic the effects of miR‑30a overexpression in RCC cells. These results highlight the important role for miR‑30a in the occurrence and development of RCC, and the restoration of miR‑30a might be investigated as a potential strategy for treating RCC.

Introduction

Renal cell carcinoma (RCC), the most common neoplasm of the adult kidney, accounts for ~3% of all human malignancies (1). In the USA, the morbidity and mortality of RCC has increased with ~63,920 new cases and 13,860 mortalities due to RCC in 2014 (2,3). Certain environmental and genetic factors have been demonstrated to be associated with RCC; however, the molecular mechanisms underlying RCC carcinogenesis and progression remains poorly understood (4).

Clear cell RCC is the most common renal parenchymal carcinoma, which represents ~70–80% of RCC cases. Papillary (~15%), chromophobe (5%) and other more rare subtypes, including collecting duct carcinoma (<1%) comprises the remaining cases (5). Although significant improvement has been developed in the treatments of RCC, the clinical behaviors and progression are highly variable (6,7). Patients with early-stage RCC have a 90% 5-year overall survival rate; however, the prognosis for metastatic RCC and advanced stage RCC remains poor with a 5-year survival rate <10% (8,9). Therefore, investigating the molecular mechanism of RCC is crucial for investigating novel therapeutic targets that will improve the survival rate.

MicroRNAs (miRNAs) are a large family of single-stranded, non-coding small RNAs with a length of 19–22 nucleotides. miRNAs have important functions in the regulation of gene expression via binding to the 3′ untranslated regions (3′-UTRs) of their target mRNA to mediate translational inhibition or degradation of RNA transcripts in a sequence-specific manner (10,11). In humans, ~1,000 miRNAs identified modulate the expression levels of one third of the total protein-coding transcriptome at the post-transcriptional and translational levels, and therefore have vital roles in a wide range of biological processes, including cell proliferation, apoptosis, cycle, angiogenesis, migration, metastasis and differentiation (12,13). A great deal of studies reported that miRNAs were differentially expressed in human cancers, including RCC. For example, miR-451 (14) and miR-877 (15) expression levels were reduced in RCC, whereas miR-155 (16) and miR-142-3p (17) were upregulated in RCC compared with normal tissues. miRNAs, that are upregulated in cancer tissues, may function as oncogenes by targeting tumor suppressor genes, whereas lowly expressed miRNAs may act as tumor suppressors by negatively regulating the expression of oncogenes (18).

The present study focused on the expression and functional roles of miR-30a in RCC. Firstly, the expression levels of miR-30a in RCC tissues and cell lines were analyzed. Furthermore, the roles of miR-30a in the progression of RCC, including cell proliferation, migration and invasion, were also investigated. ADAM metallopeptidase domain 9 (ADAM9) was validated as a direct target of miR-30a in RCC. The present study may provide a theoretical basis for further study on the pathogenesis and novel treatments for RCC.

Materials and methods

Surgical specimens

The present study was approved by Hebei Medical University Fourth Hospital (Shijiazhuang, China). Written informed consent was obtained form all patients. A total of twenty-three paired RCC tissues and corresponding noncancerous tissues (NCTs) were collected from patients with RCC undergoing surgery between July 2012 and April 2014 at Hebei Medical University Fourth Hospital. These patients did not receive adjuvant treatment including radiotherapy or chemotherapy prior to surgery. RCC tissues and corresponding NCTs were immediately snap-frozen in liquid nitrogen until use.

Cell culture

The human RCC cell lines, A498, 786-O, 769-P and one normal renal cell line (HK-2) were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). RCC cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), while HK-2 was maintained in keratinocyte-SFM (Invitrogen; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 mg/ml streptomycin (both Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a humidified atmosphere containing 5% CO2.

Transient transfection

All small RNA molecules were obtained from Shanghai GenePharma Co., Ltd. (Shanghai, China), including miR-30a mimics, mimics negative controls (NC), ADAM9 siRNA and NC siRNA. The cells (1×106) were seeded in 6-well plates and transfected with these small RNA molecules (50 nM) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's procedure. The time interval between transfection and subsequent experimentation was 24 h. Untransfected cells were not used as controls.

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

TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used to isolate total RNA from the surgical specimens and cell lines. The RNA concentration and purity of total RNA was determined using a NanoDrop® ND-1000 (NanoDrop Technologies; Thermo Fisher Scientific, Inc.). For miR-30a expression, cDNA was synthesized using the miScript II RT kit (Qiagen GmbH, Hilden, Germany) followed by PCR analysis with QuantiFast SYBR-Green PCR kit (Qiagen GmbH). ADAM9 mRNA expression was detected using the cDNA reverse transcription kit (Thermo Fisher Scientific, Inc.) followed by PCR analysis using the QuantiFast SYBR Green PCR kit according to the manufacturer's protocol. The relative expression of miR-30a and ADAM9 mRNA was calculated using the 2−∆∆Cq method, and normalized to RNU6 and β-actin expression, respectively.

Cell counting kit (CCK)-8

Cell proliferation was determined using the CCK-8 assay (Dojindo Molecular Technologies, Inc., Rockville, MD, USA). In brief, transfected cells were harvested at 24 h post-transfection, and re-seeded into 96-well plates at a density of 3,000 cells per well. Following incubation for 24, 48, 72 and 96 h at 37°C, CCK-8 assay was performed according the manufacturer's instructions. 10 µl CCK-8 solution was plated into each well and incubated for additional 2 h. The absorbance at 450 nm was detected using an immunoassay analyzer (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Cell migration and invasion assay

Cell migratory and invasive abilities were evaluated using Transwell chambers (Corning Incorporated, Corning, NY, USA) with a polycarbonate membrane (pore size, 8 µm). Transfected cells were harvested at 48 h post-transfection and resuspended in FBS-free culture medium. A total of 5×104 cells in 200 µl FBS-free DMEM were added into the upper chamber, and the lower chamber was filled with 500 µl culture medium containing 20% FBS. Following incubation for 48 h at 37°C in a humidified atmosphere containing 5% CO2, the chambers were fixed with 95% ethanol, stained with 0.5% crystal violet at 37°C for 30 min, washed with PBS (Gibco; Thermo Fisher Scientific, Inc.) and counted with a light microscope (Olympus IX53; Olympus Corporation, Tokyo, Japan). For cell invasion assay, the membranes of the Transwell chambers were pre-coated with Matrigel (BD Biosciences, San Jose, CA, USA). Except for the use of Matrigel pre-coated chambers, cell invasion assay were performed with the same procedure of the cell migration assay.

Prediction of miR-30a targets

Two independent online databases, miRanda (http://www.microrna.org) and TargetScan 7.0 (http://www.targetscan.org/), were used to predict miR-30a target genes.

Luciferase reporter assay

Luciferase reporter vectors, pMIR-ADAM9-3′UTR wild-type (Wt) and pMIR- ADAM9-3′UTR mutant-type (Mut) were synthesized and purified by Shanghai GenePharma Co., Ltd. For the luciferase reporter assay, 293T cells were seeded in 24-well plates at a density of 60–70% confluence. Following incubation overnight, 293T cells were co-transfected with luciferase reporter vectors and miR-30a mimics or NC using Lipofectamine® 2000. After incubation 48 h at 37°C in a humidified atmosphere containing 5% CO2, the activity of firefly and Renilla luciferase were measured using the Dual-Luciferase Reporter Assay system (Promega Corporation, Madison, WI, USA) according to the manufacturer's instructions.

Western blot

At 72 h post-transfection, total protein was isolated from transfected cells using RIPA lysis buffer (Beyotime Institute of Biotechnology, Haimen, China). Briefly, equal quantities of protein (20 µg) were separated by 10% SDS-PAGE and then transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). After blocking with 5% non-fat dried milk in Tris-buffered saline containing 0.1% Tween-20 (TBST) for 1 h at room temperature, the membranes were probed with primary antibodies at 4°C overnight. The primary antibodies used in the present study included mouse anti-human monoclonal ADAM9 antibody (cat. no. sc-377233; dilution, 1:1,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and mouse anti-human monoclonal GAPDH antibody (cat. sc-166574; dilution, 1:1,000; Santa Cruz Biotechnology, Inc.). Thereafter, the membranes were washed with TBST for three times, incubated with the corresponding horseradish peroxidase-conjugated goat anti-mouse secondary ImmunoglobulinG (cat. ab150113; dilution, 1:1,000; Abcam, Cambridge, UK) at room temperature for 1 h, and visualized by chemiluminescence using the ECL detection system (Pierce; Thermo Fisher Scientific, Inc.). The protein expression levels were normalized to GAPDH. Protein expression was analyzed using BandScan 5.0 software (Glyko, Inc., Novato, CA, USA). All experiments were repeated ≥3 times.

Statistical analysis

The data are expressed as the mean ± standard deviation and analyzed using SPSS software (version 18.0; Chicago, IL, USA). Statistical analysis involved the use of Student's t-test for the comparison of two groups or one-way analysis of variance for multiple comparisons. All analyses were performed using SPSS version 18.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-30a is downregulated in RCC

Firstly, the levels of miR-30a expression in RCC tissues and corresponding NCTs were detected using RT-qPCR. As shown in Fig. 1A, miR-30a expression was significantly reduced in RCC tissues in comparison with the corresponding NCTs (P<0.05). miR-30a expression in RCC cell lines was also analyzed. As indicated in Fig. 1B, miR-30a was generally downregulated in RCC cell lines compared with HK-2 (P<0.05). These results suggested that miR-30a might act as a tumor suppressor in RCC carcinogenesis and progression.

miR-30a inhibits the proliferation, migration and invasion of RCC cells

The CCK-8 assay was used to investigate whether miR-30a overexpression had any effect on the proliferation of RCC cells. Firstly, miR-30a mimics were introduced into 786-O and A498 cells, which exhibited higher miR-30a expression levels, compared with the NC group. The transfection efficiency of miR-30a was determined using RT-qPCR. As indicated in Fig. 2A, miR-30a was markedly increased in 786-O and A498 cells that were transfected with miR-30a mimics compared with the NC (P<0.05). The results of the CCK-8 assay showed that the overexpression of miR-30a decreased the proliferation of 786-O and A498 cells (Fig. 2B).

Cell migration and invasion assays were performed to investigate the role of miR-30a in the migration and invasion of RCC cells. The results revealed that the migratory and invasive abilities of the miR-30a mimics group were significantly decreased in 786-O and A498 cells in comparison with the NC groups (Fig. 2C, P<0.05). These results indicated that the upregulation of miR-30a inhibited the growth and metastasis of RCC cells.

miR-30a directly targets ADAM9 in RCC cells

miRNAs usually performs its functions via the negatively regulation of their target genes. Therefore, the next aim of the present study was to investigate the direct target genes of miR-30a that contribute to the suppressive effects in cell proliferation, migration and invasion.

Bioinformatics analysis with miRanda and TargetScan indicated that the 3′UTR of ADAM9 contains complementary binding sites for miR-30a (Fig. 3A). To investigate whether ADAM9 was a direct target gene of miR-30a, luciferase reporter assays were performed. Compared with the NC, the miR-30a mimic markedly repressed the luciferase activity of pMIR-ADAM9-3′UTR Wt (P<0.05) but did not alter the luciferase activity of the pMIR-ADAM9-3′UTR Mut (Fig. 3B, P>0.05). To investigate whether miR-30a was able to regulate ADAM9 expression, RT-qPCR and western blotting were performed in 786-O and A498 cells that were transfected with miR-30a mimics or NC. The mRNA (Fig. 3C, P<0.05) and protein (Fig. 3D, P<0.05) levels of ADAM9 were significantly reduced in miR-30a mimics-transfected-786-O and A498 cells. These results indicated that miR-30a directly targeted ADAM9 in RCC.

Repression of ADAM9 contributes to the inhibition of the malignant phenotypes of RCC

To investigate the roles of ADAM9 in the initiation and progression of RCC, ADAM9 siRNA was used to knockdown ADAM9 expression in 786-O and A498 cells. The transfection efficiency of ADAM9 siRNA was analyzed using RT-qPCR and western blotting. The results indicated that ADAM9 was downregulated at mRNA (Fig. 4A, P<0.05) and protein (Fig. 4B, P<0.05) levels in 786-O and A498 cells that were transfected with ADAM9 siRNA compared with the NC siRNA groups.

Subsequently, CCK-8 assay, cell migration and invasion assays were performed in 786-O and A498 cells that were transfected with ADAM9 siRNA. CCK-8 assay indicated that the downregulation of ADAM9 inhibited the proliferation in 786-O and A498 cells (Fig. 4C; P<0.05). Additionally, cell migration and invasion assays revealed that ADAM9 underexpression were able to decrease the migratory and invasive abilities in 786-O and A498 cells (Fig. 4D; P<0.05). These findings suggested that ADAM9 might be a functional target of miR-30a in RCC.

Discussion

miR-30a is one of the most prominent miRNAs, and it has been demonstrated to be dysregulated in multiple types of human cancer. For example, it was downregulated in breast cancer and negatively associated with the extent of lymph node and lung metastasis in patients with breast cancer (19). miR-30a expression levels were also reduced in lung cancer, compared with a healthy individual, and significantly associated with tumor size, lymphatic metastasis, clinical tumor-node metastasis stage, pathological grading and histological classification. Survival time in patients with lung cancer and a low miR-30a expression was remarkably shorter compared with the high expression group (20). miR-30a was downregulated in bladder cancer tissues, and the expression of miR-30a was negatively associated with shorter overall survival and disease-free survival (21). The downregulation of miR-30a was also verified in hepatocellular carcinoma (22), gastric cancer (23) and colorectal cancer (24). However, glioma (25) tissues and cells expressed high expression levels of miR-30a. These findings suggested that the expression of miR-30a was tissue specific, and it might become a prognostic marker for these types of human cancer.

miR-30a has been reported to be involved in diverse biological functions in many types of human cancer. In breast cancer, miR-30a inhibited cell proliferation, migration and invasion (26). In a xenograft mouse model, the suppressive effects of miR-30a on tumor growth and distal pulmonary metastasis of breast cancer cells were demonstrated (19). It was indicated that the ectopic expression of miR-30a in non-small cell lung cancer suppressed cell growth and metastasis (27,28). Li et al (22) demonstrated that the restoration of miR-30a expression decreased cell proliferation and induced apoptosis in hepatocellular carcinoma. In colorectal cancer, the overexpression of miR-30a significantly decreased the proliferation, growth and motility of tumor cells (24,29). These findings indicated that miR-30a acted as a tumor suppressor in human cancer. However, in glioma, miR-30a promoted the proliferation and invasion of glioma cells (30,31). Furthermore, the exogenous expression of miR-30a improved the ability of nasopharyngeal carcinoma cells to metastasize and invade in vivo and in vitro (32). These conflicting results demonstrated that the roles of miR-30a are tissue specific, and this might be explained by the ‘imperfect complementarity’ of the interactions between miRNAs and their target genes.

Previous studies validated that miR-30a was able to directly target multiple genes, including ubiquitin protein ligase E3C (33), Slug (34), eyes absent homolog 2 (28), Notch1 (21), endothelin receptor type A (35), runt related transcription factor 2 (36) and metadherin (22). In the present study, the molecular mechanism by which miR-30a inhibits the proliferation, migration and invasion of RCC cells was revealed to be at least in part by negatively regulating ADAM9. The potential targets of ADAM9 for miR-30a was predicted by miRanda and TargetScan. In addition, the results of the luciferase reporter assay suggested that miR-30a was able to directly target the 3′UTR of ADAM9. Furthermore, the mRNA and protein expression levels of ADAM9 in RCC cells that were transfected with miR-30a mimics were analyzed. The restoration of miR-30a expression decreased ADAM9 expression at the level of mRNA and protein in RCC cells. Furthermore, the knockdown of ADAM9 was able to mimic the suppressive effects of miR-30a overexpression on the proliferation, migration and invasion of RCC cells. Taken together, ADAM9 might be a direct and functional downstream target gene of miR-30a in RCC.

ADAM9, a membrane-anchored metalloproteinase, is one of the first ADAM proteins to be identified and characterized. ADAM9 comprises an N-terminal prodomain followed by a metalloprotease domain, a disintegrin domain and cysteine-rich region, an epidermal growth factor repeat, a transmembrane domain and a cytoplasmic tail with potential SH3 ligand domains (37,38). In RCC, ADAM9 was significantly upregulated in tumor tissues in comparison with adjacent normal tissues in the present study. ADAM9 was reported to be significantly associated with higher tumor grade, positive nodal status and distant metastasis (39). In addition, the levels of ADAM9 expression were associated with shorter patient survival in univariate analysis (39). In the present study, it was demonstrated that the knockdown of ADAM9 inhibited the proliferation, migration and invasion of RCC cells. The present study suggested that ADAM9 may act as an oncogene in RCC, and the inhibition of ADAM9 might serve as a novel therapeutic method for RCC.

Previous studies also reported that miRNAs might target ADAM9 and contribute to diverse biological functions. For example, in osteosarcoma, miR-126 targeted ADAM9 to inhibit cell proliferation and invasion (40). In hepatocellular carcinoma, ADAM9 might be targeted by miR-1274a (41). Together with the results of the present study, the miR-30a/ADAM9 axis might be investigated as a potential target for the treatment of RCC.

The present study suggested that miR-30a was a potential tumor suppressor in RCC. miR-30a, by targeting ADAM9, might inhibit the proliferation, migration and invasion of RCC cells. The restoration of miR-30a might be a potential strategy for treating RCC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

LJ and YL designed the study. CM and BL performed the experiments. BL analyzed the data.

Ethics approval and consent to participate

All patients were required to provide written informed consent prior to their inclusion. The study was approved by the Ethics Committee of Hebei Medical University Fourth Hospital.

Patient consent for publication

All patients provided written informed consent for the publication of their data.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Siegel R, Desantis C and Jemal A: Colorectal cancer statistics, 2014. CA Cancer J Clin. 64:104–117. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Cairns P: Renal cell carcinoma. Cancer Biomark. 9:461–473. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Janzen NK, Kim HL, Figlin RA and Belldegrun AS: Surveillance after radical or partial nephrectomy for localized renal cell carcinoma and management of recurrent disease. Urol Clin North Am. 30:843–852. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Decastro GJ and McKiernan JM: Epidemiology, clinical staging, and presentation of renal cell carcinoma. Urol Clin North Am. 35(581–592): vi2008.

6 

Ljungberg B, Cowan NC, Hanbury DC, Hora M, Kuczyk MA, Merseburger AS, Patard JJ, Mulders PF and Sinescu IC: European Association of Urology Guideline Group: EAU guidelines on renal cell carcinoma: The 2010 update. Eur Urol. 58:398–406. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Park I, Lee JL, Ahn JH, Lee DH, Lee KH, Jeong IG, Song C, Hong B, Hong JH and Ahn H: Active surveillance for metastatic or recurrent renal cell carcinoma. J Cancer Res Clin Oncol. 140:1421–1428. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Chaffer CL and Weinberg RA: A perspective on cancer cell metastasis. Science. 331:1559–1564. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Hirata H, Hinoda Y, Ueno K, Nakajima K, Ishii N and Dahiya R: MicroRNA-1826 directly targets beta-catenin (CTNNB1) and MEK1 (MAP2K1) in VHL-inactivated renal cancer. Carcinogenesis. 33:501–508. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Bushati N and Cohen SM: microRNA functions. Annu Rev Cell Dev Biol. 23:175–205. 2007. View Article : Google Scholar : PubMed/NCBI

11 

de Planell-Saguer M and Rodicio MC: Analytical aspects of microRNA in diagnostics: A review. Anal Chim Acta. 699:134–152. 2011. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Ha TY: MicroRNAs in human diseases: From cancer to cardiovascular disease. Immune Netw. 11:135–154. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Tang Y, Wan W, Wang L, Ji S and Zhang J: microRNA-451 inhibited cell proliferation, migration and invasion through regulation of MIF in renal cell carcinoma. Int J Clin Exp Pathol. 8:15611–15621. 2015.PubMed/NCBI

15 

Shi Q, Xu X, Liu Q, Luo F, Shi J and He X: MicroRNA-877 acts as a tumor suppressor by directly targeting eEF2K in renal cell carcinoma. Oncol Lett. 11:1474–1480. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Gao Y, Ma X, Yao Y, Li H, Fan Y, Zhang Y, Zhao C, Wang L, Ma M, Lei Z and Zhang X: miR-155 regulates the proliferation and invasion of clear cell renal cell carcinoma cells by targeting E2F2. Oncotarget. 7:20324–20337. 2016.PubMed/NCBI

17 

Li Y, Chen D, Jin LU, Liu J, Li Y, Su Z, Qi Z, Shi M, Jiang Z, Yang S, et al: Oncogenic microRNA-142-3p is associated with cellular migration, proliferation and apoptosis in renal cell carcinoma. Oncol Lett. 11:1235–1241. 2016. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Zhang N, Wang X, Huo Q, Sun M, Cai C, Liu Z, Hu G and Yang Q: MicroRNA-30a suppresses breast tumor growth and metastasis by targeting metadherin. Oncogene. 33:3119–3128. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Tang R, Liang L, Luo D, Feng Z, Huang Q, He R, Gan T, Yang L and Chen G: Downregulation of MiR-30a is associated with poor prognosis in lung cancer. Med Sci Monit. 21:2514–2520. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Zhang C, Ma X, Du J, Yao Z, Shi T, Ai Q, Chen X, Zhang Z, Zhang X and Yao X: MicroRNA-30a as a prognostic factor in urothelial carcinoma of bladder inhibits cellular malignancy by antagonising Notch1. BJU Int. 118:578–589. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Li WF, Dai H, Ou Q, Zuo GQ and Liu CA: Overexpression of microRNA-30a-5p inhibits liver cancer cell proliferation and induces apoptosis by targeting MTDH/PTEN/AKT pathway. Tumour Biol. 37:5885–5895. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Liu Z, Chen L, Zhang X, Xu X, Xing H, Zhang Y, Li W, Yu H, Zeng J and Jia J: RUNX3 regulates vimentin expression via miR-30a during epithelial-mesenchymal transition in gastric cancer cells. J Cell Mol Med. 18:610–623. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Zhong M, Bian Z and Wu Z: miR-30a suppresses cell migration and invasion through downregulation of PIK3CD in colorectal carcinoma. Cell Physiol Biochem. 31:209–218. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Wang K, Jia Z, Zou J, Zhang A, Wang G, Hao J, Wang Y, Yang S and Pu P: Analysis of hsa-miR-30a-5p expression in human gliomas. Pathol Oncol Res. 19:405–411. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Cheng CW, Wang HW, Chang CW, Chu HW, Chen CY, Yu JC, Chao JI, Liu HF, Ding SL and Shen CY: MicroRNA-30a inhibits cell migration and invasion by downregulating vimentin expression and is a potential prognostic marker in breast cancer. Breast Cancer Res Treat. 134:1081–1093. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Wen XP, Ma HL, Zhao LY, Zhang W and Dang CX: MiR-30a suppresses non-small cell lung cancer progression through AKT signaling pathway by targeting IGF1R. Cell Mol Biol (Noisy-le-grand). 61:78–85. 2015.PubMed/NCBI

28 

Yuan Y, Zheng S, Li Q, Xiang X, Gao T, Ran P, Sun L, Huang Q, Xie F, Du J and Xiao C: Overexpression of miR-30a in lung adenocarcinoma A549 cell line inhibits migration and invasion via targeting EYA2. Acta Biochim Biophys Sin (Shanghai). 48:220–228. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Baraniskin A, Birkenkamp-Demtroder K, Maghnouj A, Zöllner H, Munding J, Klein-Scory S, Reinacher-Schick A, Schwarte-Waldhoff I, Schmiegel W and Hahn SA: MiR-30a-5p suppresses tumor growth in colon carcinoma by targeting DTL. Carcinogenesis. 33:732–739. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Wang Z, Dai X, Chen Y, Sun C, Zhu Q, Zhao H, Liu G, Huang Q and Lan Q: MiR-30a-5p is induced by Wnt/β-catenin pathway and promotes glioma cell invasion by repressing NCAM. Biochem Biophys Res Commun. 465:374–380. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Jia Z, Wang K, Wang G, Zhang A and Pu P: MiR-30a-5p antisense oligonucleotide suppresses glioma cell growth by targeting SEPT7. PLoS One. 8:e550082013. View Article : Google Scholar : PubMed/NCBI

32 

Wang HY, Li YY, Fu S, Wang XP, Huang MY, Zhang X, Shao Q, Deng L, Zeng MS, Zeng YX and Shao JY: MicroRNA-30a promotes invasiveness and metastasis in vitro and in vivo through epithelial-mesenchymal transition and results in poor survival of nasopharyngeal carcinoma patients. Exp Biol Med (Maywood). 239:891–898. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Xiong J, Wei B, Ye Q and Liu W: MiR-30a-5p/UBE3C axis regulates breast cancer cell proliferation and migration. Biochem Biophys Res Commun. 18–Mar;2016.(Epub ahead of print). View Article : Google Scholar

34 

Chang CW, Yu JC, Hsieh YH, Yao CC, Chao JI, Chen PM, Hsieh HY, Hsiung CN, Chu HW, Shen CY and Cheng CW: MicroRNA-30a increases tight junction protein expression to suppress the epithelial-mesenchymal transition and metastasis by targeting Slug in breast cancer. Oncotarget. 7:16462–16478. 2016.PubMed/NCBI

35 

Sestito R, Cianfrocca R, Rosanò L, Tocci P, Semprucci E, Di Castro V, Caprara V, Ferrandina G, Sacconi A, Blandino G and Bagnato A: miR-30a inhibits endothelin A receptor and chemoresistance in ovarian carcinoma. Oncotarget. 7:4009–4023. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Jiang D, Zheng X, Shan W and Shan Y: The overexpression of miR-30a affects cell proliferation of chondrosarcoma via targeting Runx2. Tumour Biol. 37:5933–5940. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Guaiquil V, Swendeman S, Yoshida T, Chavala S, Campochiaro PA and Blobel CP: ADAM9 is involved in pathological retinal neovascularization. Mol Cell Biol. 29:2694–2703. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Duffy MJ, McKiernan E, O'Donovan N and McGowan PM: Role of ADAMs in cancer formation and progression. Clin Cancer Res. 15:1140–1144. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Fritzsche FR, Wassermann K, Jung M, Tölle A, Kristiansen I, Lein M, Johannsen M, Dietel M, Jung K and Kristiansen G: ADAM9 is highly expressed in renal cell cancer and is associated with tumour progression. BMC Cancer. 8:1792008. View Article : Google Scholar : PubMed/NCBI

40 

Wang S, Wang X, Guo Q, Wang G, Han X, Li X, Shi ZW and He W: MicroRNA-126 overexpression inhibits proliferation and invasion in osteosarcoma cells. Technol Cancer Res Treat. 15:NP49–NP59. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Zhou C, Liu J, Li Y, Liu L, Zhang X, Ma CY, Hua SC, Yang M and Yuan Q: microRNA-1274a, a modulator of sorafenib induced a disintegrin and metalloproteinase 9 (ADAM9) down-regulation in hepatocellular carcinoma. FEBS Lett. 585:1828–1834. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 16 Issue 3

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
Jiang L, Liu Y, Ma C and Li B: MicroRNA‑30a suppresses the proliferation, migration and invasion of human renal cell carcinoma cells by directly targeting ADAM9 Retraction in /10.3892/ol.2024.14232. Oncol Lett 16: 3038-3044, 2018
APA
Jiang, L., Liu, Y., Ma, C., & Li, B. (2018). MicroRNA‑30a suppresses the proliferation, migration and invasion of human renal cell carcinoma cells by directly targeting ADAM9 Retraction in /10.3892/ol.2024.14232. Oncology Letters, 16, 3038-3044. https://doi.org/10.3892/ol.2018.8999
MLA
Jiang, L., Liu, Y., Ma, C., Li, B."MicroRNA‑30a suppresses the proliferation, migration and invasion of human renal cell carcinoma cells by directly targeting ADAM9 Retraction in /10.3892/ol.2024.14232". Oncology Letters 16.3 (2018): 3038-3044.
Chicago
Jiang, L., Liu, Y., Ma, C., Li, B."MicroRNA‑30a suppresses the proliferation, migration and invasion of human renal cell carcinoma cells by directly targeting ADAM9 Retraction in /10.3892/ol.2024.14232". Oncology Letters 16, no. 3 (2018): 3038-3044. https://doi.org/10.3892/ol.2018.8999