Open Access

miR‑539 suppresses proliferation and induces apoptosis in renal cell carcinoma by targeting high mobility group A2

  • Authors:
    • Zhi‑Hua Ye
    • Ding‑Wen Gui
  • View Affiliations

  • Published online on: February 8, 2018     https://doi.org/10.3892/mmr.2018.8578
  • Pages: 5611-5618
  • Copyright: © Ye 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

Renal cell carcinoma (RCC) is one of the most common urinary malignancies with a high rate of morbidity. MicroRNAs (miRNAs) have been shown to be critical post‑transcriptional regulators in tumorigenesis. The present study aimed to investigate the effect of miRNA (miR)‑539 on the proliferation and apoptosis of RCC. The expression of miR‑539 and high mobility group AT‑hook 2(HMGA2) were examined in clinical RCC specimens. The 786‑O RCC cell line was also used and was transfected with miR‑539 mimics or inhibitors. The correlation between miR‑539 and HMGA2 was confirmed using a luciferase reporter assay. Cell viability and apoptosis were detected using MTT and flow cytometry assays. The protein levels of HMGA2, AKT, phosphorylated (p)‑AKT, mammalian target of rapamycin (mTOR) and p‑mTOR were analyzed using western blot analysis. The results revealed that miR‑539 was negatively correlated with the expression of HMGA2 in clinical RCC specimens. Further experiments identified HMGA2 as a direct target of miR‑539. The overexpression of miR‑539 downregulated the expression of HMGA2, reduced cell proliferation and promoted cell apoptosis, whereas the knockdown of miR‑539 led to the opposite results. miR‑539 also suppressed the phosphorylation of AKT and mTOR, without altering the levels of total AKT and mTOR. Taken together, the results of the present study indicated that miR‑539 negatively regulated the expression of HMGA2 in clinical specimens and in vitro. miR539 inhibited cell proliferation and induced apoptosis in RCC cells. This regulatory effect of miR‑539 may be associated with the AKT signaling pathway. Therefore, miR‑539 may be used as a biomarker for predicting the progression of RCC.

Introduction

Renal cell carcinoma (RCC) is a common malignancy and is the third leading cause of cancer-associated mortality in cancer of the urinary system (1). The morbidity rates of RCC continue to increase rapidly, whereas the overall five-year survival rate has not improved significantly, resulting in unsatisfactory prognosis (2). Therefore, examining the molecular interactions occurring in the initiation and progression of RCC may assist in identifying more therapeutic targets and investigating novel approaches in prognosis.

High mobility group AT-hook 2 (HMGA2) is a member of the HMGA group, which functions as a transcriptional enhancer in the alteration of chromatin structure via binding to AT-rich regions in DNA sequences (3). HMGA2 is an oncofetal protein, the expression of which is at low levels or absent with the differentiation of tissues, which is in contrast to its high level of expression with the dedifferentiation of several types of malignant tumor (4,5). Notably, it has been observed that the abnormal expression of HMGA2 shows high correlation with various malignancies, including cancer of the colon, breast, kidney, pancreas, liver and lung (3,68). Previous studies have demonstrated that the level of HMGA2 is positively correlated with tumor size and Fuhrman grade in RCC, and the high expression of HMGA2 leads to poor prognosis of patients, which indicates that HMGA2 is important in the progression of RCC (7).

MicroRNAs (miRNAs) are a series of non-coding, single-stranded RNA molecules, consisting of 20–24 nucleotides (9). They are endogenously expressed and negatively correlated with the transcription of genes through binding to their 3′-untranslated-region (3′UTR) (10). Previous studies have demonstrated that the aberrant expression of miRNAs contributes to tumor growth and is involved in regulating the physiological behavior of tumor cells through the modulation of target genes at mRNA or protein levels. For example, miRNA-129 (miR-129) inhibits the metastasis of prostate cancer by binding to centriolar coiled-coil protein 110 (11), miR-200a regulates the proliferation and metastasis of pancreatic cancer through modulating the DEK gene (12), and miR-543 is downregulated in colorectal cancer samples, acting as tumor suppressor by targeting KRAS, MTA1 and HMGA2 (13).

In the present study, the discrepant expression between miR-539 and HMGA2 was analyzed in RCC tumor tissues, and it was predicted and confirmed that miR-539 binds to HMGA2. Subsequently, the regulatory effect of miR-539 on the proliferation and apoptosis of RCC cells was confirmed. These findings suggest that miR-539 may be important in the progression of RCC and may be used as a biomarker for predicting the growth of RCC.

Materials and methods

Clinical specimens

The present study was approved by the Institutional Ethics Committee of Ren'min Hospital Affiliated to Wuhan University (Wuhan, China) and performed according to the guidelines on ethical management. Tissue specimens from a 23 cases of RCC and 19 paired normal tissues were collected from the Department of Urology, Ren'min Hospital of Wuhan University between 2015 and 2017. Written informed consent was signed by all participants prior to the study. The tumor stage and grade were classified according to the tumor-node-metastasis staging system of the American Joint Committee on Cancer (14). All tissues were divided into two sections, one of which was fixed in 4% paraformaldehyde, and the other was immediately frozen and stored at −80°C for subsequent analysis. The clinical data of the participants are shown in Table I.

Table I.

Clinicopathological features of participants.

Table I.

Clinicopathological features of participants.

HMGA2 expression (n)

VariablesGroupHighLowTotalP-value
GenderMale7  6130.509
Female4  610
Age (years)<608  9170.901
≥603  3  6
Clinical stageStage I–II510150.056
Stage III–IV6  2  8
T classificationT1-2410140.021
T3-47  2  9
Lymph node metastasisN0711180.103
N1-24  1  5
M classificationM0911200.483
M1-22  1  3
Differentiation degreeWell-moderate412160.007
Low6  1  7

[i] T, tumor; M, metastasis.

Cell lines and cell culture

The 786-O human kidney cancer cell line was purchased from the American Type Culture Collection (Manassas, VA, USA) and cultured in minimal Roswell Parker Memorial Institute-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% FBS (Wuhan Biofavor Biotech Services Co., Ltd., Wuhan, China) at 37°C under normoxic conditions (5% CO2, 95% O2).

Transfection and plasmid construction

The 786-O cells were transfected with miR-539 mimics and miR-539 inhibitors (Biossci Biotechnology Co., Ltd., Wuhan, China) using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. A total of 1.0×106 cells/ml cells were transferred to 24-well plates for group comparison experiments. The cells were serum-starved for 24 h for further analyses prior to reaching a confluence of 90%.

The wild-type sequence of the HMGA2 3′UTR containing predicted miR-539 binding site was amplified from the 786-O cells using the polymerase chain reaction (PCR) method. The mutant 3′UTR sequence of HMGA2 was produced using an overlap-extension PCR method. Subsequently, the wild-type and mutant sequences were subcloned into a psiCHECK-2 vector (Promega Corporation, Madison, WI, USA).

In silico prediction

The binding of miR-539 to HMGA2 was predicted using open access databases TargetScan (www.targetscan.org), miRanda (www.microrna.org). and miRwalk2.0 (http://zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk2/), and a putative binding site in the 3′UTR of HMGA2 for miR-539 was identified (Fig. 2C).

Luciferase reporter assays

For the luciferase reporter assay, the 786-O cells were seeded into a 24-well plate (2×103 cells per well), and then co-transfected with the miR-539 mimics and HMGA2-3′UTR-luciferase plasmids. At 48 h post-transfection, the cells were collected and lysed. The luciferase activities were analyzed using a Dual-Luciferase Reporter Assay system (Promega Corporation).

Immunohistochemistry

The expression of HMGA2 was examined using immunohistochemical staining. The tissues were sliced into 3-µm sections and deparaffinized in xylene, followed by dehydration in gradient ethanol and blocking with 3% hydrogen peroxide for 15 min at room temperature. The sections were then incubated with rabbit polyclonal anti-HMGA2 (cat. no. ab97276; 1:500; Abcam, Cambridge, UK) antibodies at 4°C overnight. Following washing three times with PBS, the sections were stained using diaminobenzidine reagents and counterstained with hematoxylin and the results were observed with an Olympus BX50 light microscope (Olympus Corporation, Tokyo, Japan).

Western blot analysis

Total cellular proteins were extracted with radioimmunoprecipitation buffer (Beyotime Institute of Biotechnology, Jiangsu, China). The protein concentration was measured using a bicinchoninic acid assay (Beyotime Institute of Biotechnology). Briefly, equivalent quantities of protein sample (40 µg/lane) were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and subsequently electrotransferred onto polyvinylidene fluoride membranes (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Subsequently, the membrane was blocked at 4°C for 1 h with TBS containing 0.05% Tween-20 (TBST) buffer with 5% non-fat milk and then incubated with the following primary antibodies against HMGA2 (1:1,000; cat. no. ab97276; Abcam), AKT (1:1,000; cat. no. sc-8312; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), phosphorylated (p)-AKT (1:1,000; cat. no. sc-33437; Santa Cruz Biotechnology, Inc.), mammalian target of rapamycin (1:500, mTOR; cat. no. sc-8319; Santa Cruz Biotechnology, Inc.) and p-mTOR (1:500, cat. no. sc-101738; Santa Cruz Biotechnology, Inc.) at 4°C overnight. Following incubation with secondary antibodies conjugated with horseradish peroxidase (LK2001 and LK2003; 1:100; Sungene Biotech, Co., Ltd., Tianjin, China) for 1 h at room temperature, the bands were examined using an enhanced chemiluminescence system (MultiSciences Biotech Co., Ltd., Hangzhou, China).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the clinical specimens using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Subsequently, the RNAs were reversed transcribed into cDNA using a reverse transcription reagent kit (Takara Biotechnology Co., Ltd., Dalian China). Subsequently, cDNA was amplified using an SYBR Green mix kit and the ABI 7900 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. PCR was performed with the following thermocycling conditions: Initial denaturation at 94°C for 4 min, 40 cycles of 94°C for 30 sec, 56°C for 30 sec and 72°C for 25 sec, using the ABI 7900 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific Inc.). The primer sequences are shown in Table II. Relative levels of miR-539 and mRNA expression levels of HMGA2 were normalized to that of small nuclear RNA U6 (for miRNAs) or GAPDH (for mRNAs) respectively. The relative expression of miRNA or mRNA was calculated using the 2−ΔΔCt method (15).

Table II.

Primers for reverse transcription-quantitative polymerase chain reaction analysis.

Table II.

Primers for reverse transcription-quantitative polymerase chain reaction analysis.

GenePrimer sequence (5′-3′)
HMGA2F: CGAAAGGTGCTGGGCAGCTCCGG
R: CCATTTCCTAGGTCTGCCTCTTG
miR-539F: GGAGAAAUUUCCUUGUGUGU
R: UUUCUUUAAAGGAACAUACA
U6 snRNAF: CTCGCTTCGGCAGCACATATACT
R: ACGCTTCACGAATTTGCGTGTC
GAPDHF: TGAAGGTCGGTGTGAACGGATTTGGTC
R: CATGTAGGCCATGAGGTCCACCAC

[i] HMGA2, HMGA2, high mobility group AT-hook 2; miR, microRNA; snRNA, small nuclear RNA; F, forward; R, reverse.

Cell proliferation assay

The 786-O cells were cultured in 96-well plates (2×103 cells per well) for 24 h. The cells were then stained with 10 µl of 5 mg/ml MTT per well (Sigma-Aldrich; Merck Millipore; Darmstadt, Germany) for 4 h at 37°C. The culture medium was then discarded and 150 µl of dimethyl sulfoxide was added. The absorbance was detected at 490 nm with an ELX-800 spectrometer reader (Bio-Tek Instruments, Inc., Winooski, VT, USA).

Cell apoptosis assay

Cell apoptosis was measured using Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining (BD Pharmingen, San Diego, CA, USA) according to the manufacturer's protocol. In brief, the 786-O cells were collected in 6-well plates at a concentration of 105 cells/ml. Annexin V-FITC (5 µl) and PI (5 µl) were then distributed into each well, and the cells were incubated in the dark for 15 min to undergo flow cytometry (BD LSRII; BD Pharmingen).

Statistical analysis

All data are presented as the mean ± standard deviation. Differences were assessed by two-tailed Student's t-test and χ2 test, as appropriate. P≤0.05 was considered to indicate a statistically significant difference. All experiments were performed at least three times. Statistical analyses were performed using SPSS 19.0 (IBM SPSS, Armonk, NY, USA).

Results

Expression of miR-539 negatively correlates with the expression of HMGA2 in RCC samples

To examine the expression of HMGA2 in RCC samples and adjacent normal control samples, immunohistochemistry was performed to stain tissues from 23 cases of RCC tissues and 19 paired normal tissues. As shown in Fig. 1A, HMGA2 was predominantly expressed in the nucleus of cells and the expression of HMGA2 was significantly increased in the RCC tissues, compared with that in the adjacent normal tissues. In addition to the progression of RCC, HMGA2 was expressed at a higher level in advanced samples. By contrast, the RT-qPCR results revealed a lower expression of miR-539 in RCC samples, compared with that in paired normal samples (Fig. 1B and C). Pearson's correlation analysis was performed, and the result showed that the expression of miR-539 was negatively correlated with the expression of HMGA2 (Fig. 1D).

miR-539 directly regulates the expression of HMGA2

To further confirm the hypothesis that miR-539 downregulates the expression of HMGA2, the 786-O RCC cell line was used. The 786-O cells were transfected with miR-539 mimics or inhibitors to obtain cells with miR-539 overexpression or knockdown. Using western blot analysis, the expression of HMGA2 was measured. As the data revealed, the expression of HMGA2 was significantly downregulated in the cells overexpressing miR-539, but was moderately increased in the miR-539-knockdown cells (Fig. 2A and B). Subsequently, it was predicted that miR-539 was an upstream regulator of HMGA2 using open access databases and a putative binding site in the 3′UTR of HMGA2 for miR-539 was identified (Fig. 2C). To confirm this prediction, a luciferase reporter assay was performed. The result revealed that the reporter activity of the HMGA2 3′UTR was significantly suppressed in the cells overexpressing miR-539. However, this effect was abrogated when the putative binding site in the 3′UTR of HMGA2 was mutated (Fig. 2D). Taken together, these results suggested that miR-539 negatively regulated the expression of HMGA2 by directly targeting it.

miR-539 suppresses proliferation and promotes apoptosis of RCC cells

To investigate the effect of miR-539 on the proliferation of RCC cells, an MTT assay was performed and the data showed that the viability of 786-O cells transfected with miR-539 mimics was markedly suppressed, compared with that in the control group, whereas transfection with miR-539 inhibitors increased cell viability (Fig. 3A). Subsequently, the effect of miR-539 on the apoptosis of RCC cells was observed. The results of the flow cytometry revealed that the overexpression of miR-539 led to a significant increase in apoptotic rate, compared with that in the control group, and this promotion of cell apoptosis was reversed following the use of miR-539 inhibitor (Fig. 3B and C). Collectively, these results indicated that miR-539 regulated the proliferation and apoptosis of RCC cells.

Effects of miR-539 on the AKT signaling pathway

To investigate whether the AKT signaling pathway was involved in the potential mechanism of miR-539, the present study detected the phosphorylation levels of AKT and mTOR. The results of the western blot analysis showed that the overexpression of miR-539 suppressed the expression of p-AKT and p-mTOR, compared with the expression in the control group, whereas the knockdown of miR-539 led to the opposite results. The total levels of AKT and mTOR were not affected by miR-539 (Fig. 4A and B). These data indicated that miR-539 is involved in regulation of the AKT signaling pathway, which may be an important factor to the growth of 786-O cells.

Discussion

RCC is not sensitive to radiotherapy or chemotherapy; therefore, partial or radical nephrectomy is the preferred treatment for localized RCC in patients (16,17). Unfortunately, RCC usually occurs without typical symptoms, therefore, it is difficult to diagnose prior to its development into advanced stages or distant metastasis (18). According to previous studies, up to one-third of patients with RCC exhibit metastatsis at the time of diagnosis, whereas almost 40% of patients with a localized lesion have been shown to experience recurrence following surgical therapies (19,20). Therefore, it is important to elucidate the mechanism underlying the progression of RCC and examine novel molecular interactive targets to develop more effective therapeutic approaches. Previous studies have indicated that miRNA can regulate tumor proliferation and apoptosis in cancer cells by targeting specific genetic markers (21). Based on these results, the present study aimed to identify miRNAs involved in regulating the progression of RCC.

Several studies have confirmed that miRNA is important in tumorigenesis and metastasis. It has been elucidated that miRNA regulates gene expression post-transcriptionally and acts as an oncogene or tumor suppressor in different types of cancer (22,23). miR-539 has been reported to be downregulated in osteosarcoma and suppresses tumor metastasis by targeting MMP-8 (24). It has also been shown that miR-539 functions as a tumor suppressor in prostate cancer through binding to sperm-associated antigen 5 (25). A previous study revealed that miR-539 can be used as a prognostic biomarker for colon cancer (26). However, the effect of miR-539 on RCC and its underlying mechanisms had not been elucidated. In the present study, a negative correlation was found between miR-539 and HMGA2 in RCC specimens. Compared with adjacent normal tissues, miR-539 was significantly downregulated, whereas HMGA2 was upregulated in tumors. Subsequently, the binding correlation between miR-539 and HMGA2 was predicted using open access databases, and HMGA2 was confirmed as a direct downstream target of miR-539 using a luciferase reporter assay.

HMGA2 has been reported to be abnormally expressed in various types of cancer, and to regulate the proliferation and apoptosis of tumor cells through multiple pathways (27,28). For example, HMGA2 promotes the proliferation and metastasis of nasopharyngeal cancer cells through activation by the transforming growth factor-β/small mothers against decapentaplegic 3 signaling pathway (29). In addition, the downregulation of HMGA2 effectively inhibits the proliferation process and promotes apoptosis in prostate cancer (30). In the present study, in vitro experiments were performed using the 786-O renal carcinoma cell line to simulate the progression of RCC. As expected, the overexpression of miR-539 suppressed the expression of HMGA2, whereas the inhibition of miR-539 caused a significant upregulation in the expression of HMGA2. These data demonstrated that miR-539 negatively regulated the expression of HMGA2 in RCC cells, which was in agreement with the observations from clinical samples.

A previous study showed that the phosphoinositide-3-kinase/-Akt signaling pathway is involved in the regulation of cell proliferation and apoptosis (31,32). It has also been reported that HMGA2 promotes cell proliferation by activating the AKT pathway (33). Accordingly, the present study aimed to investigate whether the AKT signaling pathway mediated the tumor suppression induced by miR-539. The data revealed that the overexpression of miR-539 inhibited the phosphorylation of AKT and mTOR, rather than altering the expression of total AKT and mTOR. Taken together, the above results suggested that miR-539 may be important in regulating the AKT pathway. This regulatory effect may be initiated via the modulation of the expression of HMGA2 by miR-539.

In conclusion, the present study indicated that miR-539 acted as a tumor suppressor in RCC cells by suppressing cell proliferation and inducing cell apoptosis, and this suppression may be due, at least in part, to the modulation of HMGA2 through the AKT signaling pathway. These results suggested that miR-539 may be used as a diagnostic biomarker for RCC treatment.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

ZY was responsible for conception and design of the study, data collection and analysis, and manuscript writing. DG, designed the study, performed critical revision and supervised all phases of the study.

Ethics approval and consent to participate

The present study was approved by the Institutional Ethics Committee of Ren'min Hospital Affiliated to Wuhan University (Wuhan, China) and performed according to the guidelines on ethical management. Written informed consent was signed by all participants prior to the study.

Consent for publication

Written informed consent was signed by all participants prior to the study.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel R, Ward E, Brawley O and Jemal A: Cancer statistics, 2011: The impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 61:212–236. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Yang CM, Ji S, Li Y, Fu LY, Jiang T and Meng FD: β-Catenin promotes cell proliferation, migration, and invasion but induces apoptosis in renal cell carcinoma. Onco Targets Ther. 10:711–724. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Wu J, Zhang S, Shan J, Hu Z, Liu X, Chen L, Ren X, Yao L, Sheng H, Li L, et al: Elevated HMGA2 expression is associated with cancer aggressiveness and predicts poor outcome in breast cancer. Cancer Lett. 376:284–292. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Piscuoglio S, Zlobec I, Pallante P, Sepe R, Esposito F, Zimmermann A, Diamantis I, Terracciano L, Fusco A and Karamitopoulou E: HMGA1 and HMGA2 protein expression correlates with advanced tumour grade and lymph node metastasis in pancreatic adenocarcinoma. Histopathology. 60:397–404. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Parameswaran S, Xia X, Hegde G and Ahmad I: Hmga2 regulates self-renewal of retinal progenitors. Development. 141:4087–4097. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Pallante P, Sepe R, Puca F and Fusco A: High mobility group a proteins as tumor markers. Front Med (Lausanne). 2:152015.PubMed/NCBI

7 

Na N, Si T, Huang Z, Miao B, Hong L, Li H and Qiu J and Qiu J: High expression of HMGA2 predicts poor survival in patients with clear cell renal cell carcinoma. Onco Targets Ther. 9:7199–7205. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Huang W, Li J, Guo X, Zhao Y and Yuan X: miR-663a inhibits hepatocellular carcinoma cell proliferation and invasion by targeting HMGA2. Biomed Pharmacother. 81:431–438. 2016. View Article : Google Scholar : PubMed/NCBI

9 

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

10 

Kala R, Peek GW, Hardy TM and Tollefsbol TO: MicroRNAs: An emerging science in cancer epigenetics. J Clin Bioinforma. 3:62013. View Article : Google Scholar : PubMed/NCBI

11 

Bijnsdorp IV, Hodzic J, Lagerweij T, Westerman B, Krijgsman O, Broeke J, Verweij F, Nilsson RJ, Rozendaal L, van Beusechem VW, et al: miR-129-3p controls centrosome number in metastatic prostate cancer cells by repressing CP110. Oncotarget. 7:16676–16687. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Wu X, Wu G, Wu Z, Yao X and Li G: MiR-200a suppresses the proliferation and metastasis in pancreatic ductal adenocarcinoma through downregulation of DEK gene. Transl Oncol. 9:25–31. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Fan C, Lin Y, Mao Y, Huang Z, Liu AY, Ma H, Yu D, Maitikabili A, Xiao H, Zhang C, et al: MicroRNA-543 suppresses colorectal cancer growth and metastasis by targeting KRAS, MTA1 and HMGA2. Oncotarget. 7:21825–21839. 2016.PubMed/NCBI

14 

Martínez-Salamanca JI, Huang WC, Millán I, Bertini R, Bianco FJ, Carballido JA, Ciancio G, Hernández C, Herranz F, Haferkamp A, et al: Prognostic impact of the 2009 UICC/AJCC TNM staging system for renal cell carcinoma with venous extension. Eur Urol. 59:120–127. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Rao X, Huang X, Zhou Z and Lin X: An improvement of the 2^(-delta delta CT) method for quantitative real-time polymerase chain reaction data analysis. Biostat Bioinforma Biomath. 3:71–85. 2013.PubMed/NCBI

16 

Chow TF, Youssef YM, Lianidou E, Romaschin AD, Honey RJ, Stewart R, Pace KT and Yousef GM: Differential expression profiling of microRNAs and their potential involvement in renal cell carcinoma pathogenesis. Clin Biochem. 43:150–158. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Pan XD, Gu DH, Mao JH, Zhu H, Chen X, Zheng B and Shan Y: Concurrent inhibition of mTORC1 and mTORC2 by WYE-687 inhibits renal cell carcinoma cell growth in vitro and in vivo. PLoS One. 12:e1725552017.

18 

Wu SW, Chen PN, Lin CY, Hsieh YS and Chang HR: Everolimus suppresses invasion and migration of renal cell carcinoma by inhibiting FAK activity and reversing epithelial to mesenchymal transition in vitro and in vivo. Environ Toxicol. 32:1888–1898. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Ko JJ, Xie W, Kroeger N, Lee JL, Rini BI, Knox JJ, Bjarnason GA, Srinivas S, Pal SK, Yuasa T, et al: The international metastatic renal cell carcinoma database consortium model as a prognostic tool in patients with metastatic renal cell carcinoma previously treated with first-line targeted therapy: A population-based study. Lancet Oncol. 16:293–300. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Chatzizacharias NA, Rosich-Medina A, Dajani K, Harper S, Huguet E, Liau SS, Praseedom RK and Jah A: Surgical management of hepato-pancreatic metastasis from renal cell carcinoma. World J Gastrointest Oncol. 9:70–77. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Jiang T, Li M, Li Q, Guo Z, Sun X, Zhang X, Liu Y, Yao W and Xiao P: MicroRNA-98-5p inhibits cell proliferation and induces cell apoptosis in hepatocellular carcinoma via targeting IGF2BP1. Oncol Res. 25:1117–1127. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Okato A, Goto Y, Kurozumi A, Kato M, Kojima S, Matsushita R, Yonemori M, Miyamoto K, Ichikawa T and Seki N: Direct regulation of LAMP1 by tumor-suppressive microRNA-320a in prostate cancer. Int J Oncol. 49:111–122. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Lin Y, Liu AY, Fan C, Zheng H, Li Y, Zhang C, Wu S, Yu D, Huang Z, Liu F, et al: MicroRNA-33b Inhibits Breast Cancer Metastasis by Targeting HMGA2, SALL4 and Twist1. Sci Rep. 5:99952015. View Article : Google Scholar : PubMed/NCBI

24 

Jin H and Wang W: MicroRNA-539 suppresses osteosarcoma cell invasion and migration in vitro and targeting Matrix metallopeptidase-8. Int J Clin Exp Pathol. 8:8075–8082. 2015.PubMed/NCBI

25 

Zhang H, Li S, Yang X, Qiao B, Zhang Z and Xu Y: miR-539 inhibits prostate cancer progression by directly targeting SPAG5. J Exp Clin Cancer Res. 35:602016. View Article : Google Scholar : PubMed/NCBI

26 

Bobowicz M, Skrzypski M, Czapiewski P, Marczyk M, Maciejewska A, Jankowski M, Szulgo-Paczkowska A, Zegarski W, Pawłowski R, Polańska J, et al: Prognostic value of 5-microRNA based signature in T2-T3N0 colon cancer. Clin Exp Metastasis. 33:765–773. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Zou Q, Xiong L, Yang Z, Lv F, Yang L and Miao X: Expression levels of HMGA2 and CD9 and its clinicopathological significances in the benign and malignant lesions of the gallbladder. World J Surg Oncol. 10:922012. View Article : Google Scholar : PubMed/NCBI

28 

Li Y, Zhao Z, Xu C, Zhou Z, Zhu Z and You T: HMGA2 induces transcription factor Slug expression to promote epithelial-to-mesenchymal transition and contributes to colon cancer progression. Cancer Lett. 355:130–140. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Xia YY, Yin L, Jiang N, Guo WJ, Tian H, Jiang XS, Wu J, Chen M, Wu JZ and He X: Downregulating HMGA2 attenuates epithelial-mesenchymal transition-induced invasion and migration in nasopharyngeal cancer cells. Biochem Biophys Res Commun. 463:357–363. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Shi Z, Wu D, Tang R, Li X, Chen R, Xue S, Zhang C and Sun X: Silencing of HMGA2 promotes apoptosis and inhibits migration and invasion of prostate cancer cells. J Biosci. 41:229–236. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Xie HX, Xu ZY, Tang JN, DU YA, Huang L, Yu PF and Cheng XD: Effect of Huaier on the proliferation and apoptosis of human gastric cancer cells through modulation of the PI3K/AKT signaling pathway. Exp Ther Med. 10:1212–1218. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Deng W, Zhang Y, Gu L, Cui J, Duan B, Wang Y and Du J: Heat shock protein 27 downstream of P38-PI3K/Akt signaling antagonizes melatonin-induced apoptosis of SGC-7901 gastric cancer cells. Cancer Cell Int. 16:52016. View Article : Google Scholar : PubMed/NCBI

33 

Tan L, Wei X, Zheng L, Zeng J, Liu H, Yang S and Tan H: Amplified HMGA2 promotes cell growth by regulating Akt pathway in AML. J Cancer Res Clin Oncol. 142:389–399. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 17 Issue 4

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
Ye ZH and Ye ZH: miR‑539 suppresses proliferation and induces apoptosis in renal cell carcinoma by targeting high mobility group A2. Mol Med Rep 17: 5611-5618, 2018
APA
Ye, Z., & Ye, Z. (2018). miR‑539 suppresses proliferation and induces apoptosis in renal cell carcinoma by targeting high mobility group A2. Molecular Medicine Reports, 17, 5611-5618. https://doi.org/10.3892/mmr.2018.8578
MLA
Ye, Z., Gui, D."miR‑539 suppresses proliferation and induces apoptosis in renal cell carcinoma by targeting high mobility group A2". Molecular Medicine Reports 17.4 (2018): 5611-5618.
Chicago
Ye, Z., Gui, D."miR‑539 suppresses proliferation and induces apoptosis in renal cell carcinoma by targeting high mobility group A2". Molecular Medicine Reports 17, no. 4 (2018): 5611-5618. https://doi.org/10.3892/mmr.2018.8578