Open Access

MicroRNA‑206 regulates the epithelial‑mesenchymal transition and inhibits the invasion and metastasis of prostate cancer cells by targeting Annexin A2

  • Authors:
    • Ning Yang
    • Ling Wang
    • Jun Liu
    • Li Liu
    • Jiangbo Huang
    • Xian Chen
    • Zhigang Luo
  • View Affiliations

  • Published online on: March 30, 2018     https://doi.org/10.3892/ol.2018.8395
  • Pages: 8295-8302
  • Copyright: © Yang 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

The present study investigated the molecular mechanism by which microRNA‑206 (miR‑206) targets Annexin A2 (ANXA2) expression and inhibits the invasion and metastasis of prostatic cancer cells through regulation of the epithelial‑mesenchymal transition (EMT). Using bioinformatics analysis, miR‑206 was identified as the most promising candidate miRNA that targeted ANXA2. Prostate tissue specimens from 60 patients with prostate cancer, 30 patients with metastatic prostate cancer and 20 patients with benign prostatic hyperplasia (BPH) were examined for ANXA2 protein expression by immunohistochemistry and western blotting and for miR‑206 expression by reverse transcription‑quantitative polymerase chain reaction. Additionally, human prostate cancer PC‑3 cells were transfected with miR‑206 mimics, miR‑206 inhibitors or a negative control sequence, and expression of ANXA2, E‑cadherin and N‑cadherin was detected by western blotting. Transwell assays were performed to determine the effect of altered miR‑206 expression on the invasive behavior of PC‑3 cells. Bioinformatics analysis predicted complementary binding between miR‑206 and ANXA2 mRNA. ANXA2 protein expression was detected in a significantly higher proportion of BPH tissues (95%, 19/20) when compared with prostate cancer tissues (51.7%, 31/60; P<0.05). Similarly, ANXA2 was expressed in a significantly higher proportion of metastatic prostate cancer samples than that of prostate cancer samples (P<0.05). Expression of miR‑206 was higher than that of ANXA2 in prostate cancer samples, but lower in BPH samples. Inhibition of miR‑206 expression in PC‑3 cells upregulated ANXA2 and E‑cadherin protein expression levels, downregulated N‑cadherin and vimentin, and promoted cell invasion in vitro. These data suggested that binding between miRNA‑206 and ANXA2 mRNA may regulate EMT signaling, thereby suppressing the invasion and metastasis of prostatic cancer cells.

Introduction

The growth rate of prostate cancer is slow and early symptoms are often not obvious and are easily overlooked, which may delay diagnosis and initiation of treatment (1,2). The molecular mechanisms underlying prostate cancer metastasis remain unclear, and there is a requirement for the identification of molecular markers for metastasis to enable the treatment and to improve the prognosis of patients with prostate cancer.

The membrane-associated Annexin family is a group of highly evolutionarily conserved calcium-dependent phospholipid-binding proteins (3). The Annexin family has numerous critical functions in cell signaling, inflammation, cell proliferation and differentiation, and maintenance of the extracellular matrix integrity (4,5). Downregulation or dysfunction of Annexins may serve important roles in the development and progression of malignant tumors. Annexin A2 (ANXA2) is one of nearly 20 members of the Annexin superfamily and contains a ~70 amino acid conserved repeat region (6). ANXA2 is involved in diverse cellular functions, including cell apoptosis, membrane transport, signal transduction and cell-cell interaction (7). ANXA2 is a multifunctional protein that serves an important regulatory role in biofilm formation, endocytosis and exocytosis, osteoblast formation, bone resorption, DNA synthesis, and cell proliferation and differentiation (8). ANXA2 is also associated with a number of cellular activities by connecting membrane protein complexes with the actin cytoskeleton, and is involved in ion channel formation, plasminogen activation and cellular matrix interactions (9,10).

Epithelial-mesenchymal transition (EMT) is a highly regulated process by which epithelial cells transform into a mesenchymal cell phenotype (11). EMT is associated with primary tumor invasion, cell migration and secondary metastasis formation in a variety of tumor types, particularly in those of epithelial origin (12,13).

Bioinformatics analysis is a major tool for the identification of putative microRNA (miRNA/miR)-targeted genes. The present study used bioinformatics software analysis to identify ANXA2 mRNA as a target of miR-206 and an aim of the present study was to examine the molecular mechanism by which miR-206 targeting of ANXA2 regulates the EMT, and the invasive and metastatic activity of prostate cancer cells.

Materials and methods

Patients and tissue specimens

A total of 110 male patients were enrolled in the present study; 60 with prostate cancer (median age, 72.8 years; age range, 56–85 years), 30 with metastatic prostate cancer (median age, 73.5 years; age range, 57–85 years), and 20 with benign prostatic hyperplasia (BPH; median age, 68.6 years; age range, 52–83 years) as control. Patients with prostate cancer included in the present study received no preoperative medication and had no history of surgical castration or radiotherapy. Patients with BPH had no long-term medication history prior to surgery. Tissue samples were obtained by surgical resection at the Department of Urology at the Second Affiliated Hospital, University of South China (Hengyang, China) and were stored at −80°C prior to use. All specimens were reviewed independently by two senior pathologists and the diagnoses were confirmed by histopathological examination. The present study was certified by the Ethics Committee of the Second Affiliated Hospital of University of South China, and all participants provided written informed consent.

Cell lines

The prostate cancer PC-3 cell line was purchased from The Cell Centre of Central South University (Changsha, China). Cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), supplemented with 10% bovine serum albumin, and were incubated at 37°C in a 5% CO2 atmosphere.

Reagents

The immunohistochemical streptavidin peroxidase (S-P) kit and 3,3′-diaminobenzidine developer were obtained from Fuzhou Maixin Biotech Co., Ltd. (Fuzhou, Fujian province, China). Mouse anti-human monoclonal antibodies against ANXA2, GAPDH, E-cadherin, N-cadherin and β-actin were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Lipofectamine 2000 was purchased from Invitrogen; Thermo Fisher Scientific, Inc., the Transwell assay kit was purchased from Corning Incorporated (Corning, NY, USA), and Matrigel was obtained from BD Biosciences (Franklin Lakes, NJ, USA).

Bioinformatics analysis

miRNAs predicted to bind to ANXA2 mRNA were identified using the miRWalk online program, which contains 10 software programs (http://zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk/predictedmirnagene.html). The miRNAs with the highest predicted binding scores were identified using miRanda software (version: August 2010 release; http://www.microrna.org/microrna/home.do), which computes thermodynamic stability scores and sequence conservation scores.

Immunohistochemistry (IHC)

The prostate tissue specimens were fixed using by 10% formalin for 24–48 h at room temperature, and then embedded in paraffin. The sample was sliced into sections 4 µm thick. Immunohistochemical staining of prostate tissue specimens was performed using the S-P immunohistochemical method (14). The cytoplasmic staining intensity was scored by two pathologists as follows: No color, negative (−); pale yellow, weakly positive (+); brown, positive (++); and tan, strongly positive (+++). The percentage of tissue samples with positive expression was calculated as [(total number of samples with weakly positive + positive + strongly positive staining)/total number of samples evaluated] ×100.

RNA extraction

Total RNA was extracted from fresh prostate cancer and BPH tissues by homogenization using TRIzol reagent (Thermo Fisher Scientific, Inc. Waltham, MA, USA). Following incubation for 5 min at room temperature, the samples were mixed with 200 ml of chloroform, incubated for 5 min at room temperature, and then centrifuged at 12,000 × g for 15 min at 4°C. The supernatant was removed, combined with 200 ml isopropanol, mixed by inversion, incubated for 10 min at room temperature, and centrifuged at 12,000 × g for 15 min at 4°C. The supernatant was removed and the pellet was washed by addition of 1 ml ethanol followed by centrifugation at 12,000 × g for 15 min at 4°C. The supernatant was removed and the pellet was vacuum dried. Finally, the RNA was resuspended in deionized water and subsequently used for reverse transcription-quantitative polymerase chain reactions (RT-qPCR).

RT-qPCR

PCR amplification of miR-206 was performed using a two-step method using an RT-qPCR kit (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). RT were performed in a water bath at 50°C for 60 min and 15 min with inactivated reverse transcriptase (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) at 70°C in a water bath. Primers were designed based on the precursor sequence of miR-206 by Primer Premier 5.0 (Primer Biosoft, Paolo Alto, CA, USA). Primer sequences were as follows: miR-206 forward, 5′-TGCTTCCCGAGGCCACATGC-3′ and reverse, 5′-GTGTGTGGTTTCGGCAAGTG-3′; and U6 forward, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ and reverse, 5′-CGCTTCACGAATTTGCGTGTCAT-3′. All primers were synthesized by Guangzhou RiboBio Co., Ltd. (Guangzhou, China). The reactions were performed on a fluorescence RT-qPCR instrument, according to the manufacturer's protocol. The PCR reaction system is as follows: 2.5 µl dNTP (2.5 mM each); 2.5 µl 10× PCR buffer; 1.5 µl MgCl2 solution; 1 U Taq polymerase; 0.25× SYBRGreen I (Sigma-Aldrich; Merck KGaA) (final concentration); 1 µl 10 µM PCR forward and reverse primer; 1 µl cDNA; water (to a total volume of 25 µl). The U6 reaction was as follows: 95°C for 5 min and 35 cycles of 95°C for 10 sec, 59°C for 15 sec, 72°C for 20 sec and 82°C for 5 sec. The miRNA reaction was as follows: 95°C for 15 min and 40 cycles of 94°C for 15 sec, 55°C for 30 sec and 70°C for 30 sec). The 2−ΔΔCq was used to quantify the results (15).

Western blot analysis

To extract total cellular protein, tissues or cells were mixed with precooled lysis buffer (Sigma-Aldrich; Merck KGaA), centrifuged for 10 min at 4°C at 10,000 × g, and placed on ice for 30 min. Following centrifugation, the supernatants were removed and protein concentrations were determined using a Bradford assay. A total of 40 µg protein was separated using SDS-PAGE (10% gel). The proteins were denatured by incubation for 5 min at 100°C, separated by gel electrophoresis and transferred onto polyvinylidene difluoride membranes. The membranes were blocked at room temperature with in 5% skimmed milk powder for 1–2 h and were then washed and incubated with the following primary antibodies: Anti-ANXA2 (1:1,000; cat. no. 28385), anti-β-actin (1:10,000; cat. no. 58673), anti-vimentin (1:1,500; cat. no. 6260), anti N-cadherin (1:1,500; cat. no. 8424) and anti-E-cadherin (1:1,500; cat. no. 8426), overnight at 4°C, and added the secondary antibody (1:1,000; HRP-conjugated mouse anti-rabbit IgG; cat. no. 2357) for 2 h at temperature. All the primary antibodies and the secondary antibody were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). The proteins were treated using enhanced chemiluminescence chromogenic solution (Boster Biological Engineering Co., Ltd. Wuhan, China). Visualization of the protein on the membrane occurred upon exposure to X-ray film. The Gray value was detected by Image J software (version K1.45; National Institutes of Health, Bethesda, MD, USA).

In vitro cell invasion assay

All the cell lines were cultured in RPMI-1640 and 10% fetal bovine serum (Hangzhou Sijiqing Biological Engineering Co., Ltd., Hangzhou, China). PC-3 cells were seeded onto 6-well plates (~2×105 cells/plate) and cultured to 80% confluence prior to transfection at 37°C. Cells were transfected with miR-206 mimic or inhibitor sequences or a negative control (NC) sequence using a Lipofectamine 2000 transfection kit (Santa Cruz Biotechnology, Inc.). After 48 h of transfection, the cell extractive was analyzed to detect the activity of the reported gene. For the Transwell invasion assay, Matrigel was diluted at 1:100 in serum-free medium and 100 µl was added to coat the upper chamber at room temperature for 24 h. To detect invasion, the upper chamber was removed, and the cells in the membrane were fixed in ethanol at room temperature for 30 min and stained with 0.5% crystal violet at room temperature for 20 min. The number of invaded tumor cells in the membrane was counted under an optical microscope (Nikon Corporation, Tokyo, Japan) at ×400 magnification.

Statistical analysis

Data were analyzed using SPSS version 17 software (SPSS, Inc., Chicago, IL, USA). Data are expressed as the mean ± standard deviation. Comparisons were performed using an unpaired Student's t-test or Mann-Whitney U test for two groups or with a one-way analysis of variance, followed by the least significant difference post hoc test, for three groups. P<0.05 was considered to indicate a statistically significant difference.

Results

miRNA-ANXA2 interactions predicted by miRWalk

Using miRWalk, which combines the power of 10 programs, the following 12 miRNAs predicted to interact with ANXA2 mRNA were identified: hsa-miR-206, hsa-miR-1, hsa-miR-9, hsa-miR-613, hsa-miR-185, hsa-miR-425, hsa-miR-890, hsa-miR-155, hsa-miR-20b, hsa-miR-520h, hsa-miR-579 and hsa-miR-767-5p (Table I).

Table I.

Results of intersection analysis by miRWalk.

Table I.

Results of intersection analysis by miRWalk.

Gene namemiRNADIANAmTmiRandamiRDBmiRWalkRNAhybridPICTAR4PICTAR5PITARNA22Target scanSUM
ANXA2hsa-miR-20610111010016
ANXA2hsa-miR-110111010016
ANXA2hsa-miR-89010110010015
ANXA2hsa-miR-911011000015
ANXA2hsa-miR-61310110010015
ANXA2hsa-miR-18511011000015
ANXA2hsa-miR-42510110010015
ANXA2hsa-miR-15511010000014
ANXA2hsa-miR-20b11010000014
ANXA2hsa-miR-520h11010000014
ANXA2hsa-miR-57911010000014
ANXA2hsa-miR-767-5b11010000014

[i] 0, not predicted; 1, predicted; ANXA2, Annexin A2; miR/miRNA, microRNA.

Optimal miRNA-ANXA2 interactions predicted by miRanda

miRNAs with the highest binding scores for ANXA2 mRNA were identified using miRanda software, which incorporates thermodynamic and sequence conservation scores. This analysis identified the following 4 miRNAs that met the imposed criteria (mirSVR, ≤-0.1; phastCons, 0.5–0.7): miR-206, miR-1, miR-613 and miR-425. miR-206 was predicted to bind to ANXA2 mRNA with the highest stability and specificity, suggesting that it may exhibit preferential binding to ANXA2 mRNA (Table II).

Table II.

Scores of mirSVR and PhastCons among 12 predicted miRNAs.

Table II.

Scores of mirSVR and PhastCons among 12 predicted miRNAs.

mirSVR scoreRankingPhastCons score
hsa-miR-206−1.196510.6944
hsa-miR-1−1.196420.6944
hsa-miR-613−1.186530.6944
hsa-miR-425−1.178740.6451
hsa-miR-520h−0.867850.6697
hsa-miR-185−0.735760.6459
hsa-miR-579−0.451170.5177
hsa-miR-9−0.225280.6459
hsa-miR-155−0.209890.5084
hsa-miR-767−0.1488100.5022
hsa-miR-890−0.1193110.6944
hsa-miR-20b−0.0235120.6697

[i] miR/miRNA, microRNA.

Expression of ANXA2 protein in prostate cancer and BPH tissues

Expression of ANXA2 protein in tissue specimens was assessed by IHC. The proportion of samples positively stained for ANXA2 was significantly higher in the BPH samples (95%, 19/20) compared with the prostate cancer samples (51.7%, 31/60; P<0.001). Additionally, 76.7% (23/30) of the metastatic prostate cancer tissues exhibited positive ANXA2 staining, which was a significantly higher proportion than that of the prostate cancer samples (P<0.001 Fig. 1; Table III).

Table III.

Expression of ANXA2 in prostate cancer and BPH tissues.

Table III.

Expression of ANXA2 in prostate cancer and BPH tissues.

Immunohistochemical staining scores

nWeak positive or negative (0–2)Positive (3–4)Strong positive (5–6)P-value
BPH201316 <0.001a
Prostate cancer <0.001b
  Highly-differentiated206122
  Moderately-differentiated3418160
  Poorly-differentiated6510
Metastatic prostate cancer307815 <0.001c

{ label (or @symbol) needed for fn[@id='tfn3-ol-0-0-8395'] } ANXA2, Annexin A2; BPH, benign prostatic hyperplasia.

a P<0.001, BPH vs. prostate cancer

b P<0.001, among the three groups

c P<0.05 Metastatic prostate cancer vs. prostate cancer. Data were analyzed using Mann-Whitney U test or one-way analysis of variance followed by the least significant difference post hoc test.

Association between the expression of miR-206 and ANXA2 in prostate cancer and BPH tissues

The expression of miR-206 was significantly higher in prostate cancer tissues than in BPH tissues, while an inverse expression pattern was observed for ANXA2 (P<0.001; Fig. 2). miR-206 expression was significantly increased in the prostate cancer samples compared with the BPH samples (P<0.001). ANXA2 expression levels were significantly decreased in the prostate cancer samples, but increased in the BPH samples (P<0.001). By contrast, miR-206 was lowly expressed and ANXA2 was highly expressed in the BPH samples. Therefore, there was a negative association between the expression of miR-206 and ANXA2.

In vitro invasion assay of a prostate cancer cell line

The invasion assay demonstrated that a significantly lower number of PC-3-miR-206-inhibitor cells exhibited invasive behavior than untransfected PC-3 or PC-3-miR-206-NC cells (Fig. 3; P<0.001, data not shown). By contrast, the number of invasive PC-3-miR-206-mimic cells was significantly higher than the number of invasive PC-3 or PC-3-miR-206-NC cells. These results indicated that downregulation of miR-206 may promote the invasive capacity of PC-3 prostate cancer cells in vitro (Table IV; Fig. 3; P<0.001).

Table IV.

Changes in the effect of miR-206 expression on the invasion of prostate cancer cells in vitro.

Table IV.

Changes in the effect of miR-206 expression on the invasion of prostate cancer cells in vitro.

Groupno. invaded cells (/HPF)P-value
PC-3-miR-206-inhibitors107±16a<0.001
PC-352±9
PC-3-miR-206-NC48±8
PC-3-miR-206-mimics21±4b<0.001

a P<0.001 PC-3-miR-206-inhibitors vs. PC-3/NC

b P<0.05 PC-3-miR-206-mimics vs. PC-3/NC. miR, microRNA; NC, negative control; HPF, high power field. One-way analysis of variance followed by the least significant difference post hoc test.

Association between the expression of miR-206 and that of ANXA2 and WNT pathway proteins

Downregulation of miR-206 in PC-3-miR-206-inhibitor cells was associated with increased expression of ANXA2 and E-cadherin and decreased expression of N-cadherin and vimentin (Fig. 4). By contrast, upregulation of miR-206 in PC-3-miR-206-mimic cells resulted in downregulation of ANXA2 and E-cadherin and upregulation of N-cadherin and vimentin (P<0.05; Fig. 4). PC-3 and PC-3-miR-206-NC cells exhibited no significant differences in the expression of ANXA2, E-cadherin, N-cadherin or vimentin (Fig. 4).

The results indicated that transfection of miR-206 mimics into PC-3 cells inhibited the expression of ANXA2, resulting in the downregulation of E-cadherin and vimentin and the upregulation of N-cadherin. However, transfection of miR-206 inhibitors into PC-3 cells upregulated ANXA2 expression and concomitantly downregulated E-cadherin. Therefore, the miR-206 target ANXA2 regulates the expression of E-cadherin, N-cadherin and vimentin and may be involved in the EMT.

Discussion

Changes in the expression and distribution of ANXA2 have been detected in many types of cancer, such as laryngeal cancer, pancreatic cancer, breast cancer, and so on (1517). ANXA2 expression has also been associated with the differentiation of squamous cell lung cancer (18). ANXA2 content was demonstrated to be high in human hepatocellular cancer cells but not in embryonic tissues, normal liver cells or injured liver cells, suggesting that ANXA2 may serve a key role in hepatocellular carcinoma (19). A study undertaken by Kirshner et al (20) revealed that respiratory cilia, pleural mesothelial and alveolar epithelial cells exhibit a high expression of ANXA2. Expression of ANXA2 in prostate cancer differs from that in other tumor types. Immunohistochemical studies have demonstrated that ANXA2 highly expressed in >50% of glands in the majority (>85%) of patients with BPH (21). A number of studies have demonstrated that changes in the expression and distribution of ANXA2 are associated with the invasion and metastasis of various tumor types (22,23). However, the exact molecular mechanism by which this occurs remains unclear.

In the present study, a significantly higher proportion of BPH samples than prostate cancer samples were positively stained for ANXA2 protein. Furthermore, a significantly higher proportion of metastatic cancer samples than prostate cancer samples expressed ANXA2. Therefore, the expression of ANXA2 may be a potential marker for the progression and development of prostate cancer.

The interaction between miRNAs and mRNAs occurs through complementary binding of base pairs. However, the interaction is not usually fully complementary, and several programs have been developed to predict miRNA-mRNA binding and to identify the specific target genes of miRNAs (24). Due to the fact that the bioinformatics programs use algorithms that focus on different aspects of the interactions between miRNAs and mRNAs, the prediction results are not entirely consistent between programs. Therefore, a previous study employed a combination of predictive programs for analyses (25). In the present study, ANXA2 mRNA was predicted by miRWalk online program, which contains 10 software programs), to be a target for 12 miRNAs, of which the interaction with miR-206 was predicted to have the highest stability and specificity. This suggested that miR-206 may preferentially target ANXA2 mRNA.

The present study also revealed that the expression of miR-206 was higher than that of ANXA2 in prostate cancer samples, while an inverse association was observed in BPH tissues, suggesting a negative association between the expression of miR-206 and ANXA2. These findings further supported the possibility that ANXA2 may be a target of miR-206.

Previous studies have demonstrated that miR-206 acts as a tumor suppressor, and that its expression is downregulated in many tumor types compared with normal cells or tissues (26). The expression of miR-206 is downregulated in renal cell carcinoma, and overexpression of miR-206 targets cell division kinase CDK9 and CDK4, inducing cell cycle arrest and inhibiting cell proliferation (26). miR-206 expression is also downregulated in colon cancer cells, and overexpression of miR-206 targets d-MET and inhibits cell proliferation and metastasis (27). miR-206 directly targets the oncogenes Kirsten rat sarcoma proto-oncogene and ANXA2, thereby acting as tumor suppressor in human pancreatic ductal adenocarcinoma cells by blocking cell cycle progression, and cell proliferation, migration and invasion (28). The miRNA functional analysis and a luciferase reporter assay demonstrated that miR-206 effectively downregulated the expression of ANXA2 by binding to the 3′-untranslated region of the ANXA2 mRNA in pulmonary artery smooth muscle cells (29). However, there have been no previous studies on miR-206 expression in prostate cancer.

EMT is a complex and dynamic process. One of the early events is remodeling of the cytoskeleton, which is accompanied by increased expression of mesenchymal markers, including N-cadherin and vimentin, and decreased expression of epithelial markers, such as E-cadherin (30). A previous study demonstrated that low expression of E-cadherin may induce EMT (31). N-cadherin is a cell surface adhesion molecule and a major structural element in intercellular adhesion. Its principal function is to mediate cell adhesion and migration (32). The expression of N-cadherin is increased in epithelial malignant tumors and is associated with the EMT (33). Recent studies have demonstrated that E-cadherin and N-cadherin are aberrantly expressed in numerous tumor types and serve an important role in the occurrence, development, invasion and metastasis of tumors (34,35).

In the present study, miR-206 was demonstrated to promote the invasive ability of prostate cancer cells in vitro. Increased expression of miR-206 inhibited the expression of ANXA2, resulting in downregulation of E-cadherin, and upregulation of N-cadherin and vimentin. By contrast, decreased expression of miR-206 increased the expression of ANXA2, resulting in upregulation of E-cadherin and downregulation of N-cadherin and vimentin. These results indicated that miR-206 may bind to ANXA2 and inhibit its expression, which, in turn, regulates the EMT and inhibits the invasion and metastasis of prostate cancer cells.

Acknowledgements

Not applicable.

Funding

The Hunan Province Science Foundation (grant no. 2015JC3088).

Availability of data and materials

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

Author's contributions

NY contributed to the conception and design of the work. LW, JL, LL, JH and XC acquired and analyzed the data for the work. ZL designed the work and corrected the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was certified by the Ethics Committee of the Second Affiliated Hospital of University of South China, and all participants provided written informed consent.

Consent for publication

All participants provided written consent agreeing for publication of the present study.

Competing interests

The authors declare that they have no competing interests.

References

1 

Nandana S and Chung LW: Prostate cancer progression and metastasis: Potential regulatory pathways for therapeutic targeting. Am J Clin Exp Urol. 2:92–101. 2014.PubMed/NCBI

2 

Simons JW: Prostate cancer immunotherapy: Beyond immunity to curability. Cancer Immunol Res. 2:1034–1043. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Monastyrskaya K: Functional association between regulatory RNAs and the annexins. Int J Mol Sci. 19:E5912018. View Article : Google Scholar : PubMed/NCBI

4 

Mussunoor S and Murray GI: The role of annexins in tumour development and progression. J Pathol. 216:131–140. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Mortimer JC, Laohavisit A, Macpherson N, Webb A, Brownlee C, Battey NH and Davies JM: Annexins: Multifunctional components of growth and adaptation. J Exp Bot. 59:533–544. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Zhang M, Chen D, Zhen Z, Ao J, Yuan X and Gao X: Annexin A2 positively regulates milk synthesis and proliferation of bovine mammary epithelial cells through the mTOR signaling pathway. J Cell Physiol. 233:2464–2475. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Sun MY, Xing RH, Gao XJ, Yu X, He HM, Gao N, Shi HY, Hu YY, Wang QX, Xu JH and Hou YC: ANXA2 regulates the behavior of SGC-7901 cells. Asian Pac J Cancer Prev. 14:6007–6012. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Jeon YR, Kim SY, Lee EJ, Kim YN, Noh DY, Park SY and Moon A: Identification of annexin II as a novel secretory biomarker for breast cancer. Proteomics. 13:3145–3156. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Lokman NA, Elder AS, Ween MP, Pyragius CE, Hoffmann P, Oehler MK and Ricciardelli C: Annexin A2 is regulated by ovarian cancer-peritoneal cell interactions and promotes metastasis. Oncotarget. 4:1199–1211. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Zhang HJ, Yao DF, Yao M, Huang H, Wang L, Yan MJ, Yan XD, Gu X, Wu W and Lu SL: Annexin A2 silencing inhibits invasion, migration, and tumorigenic potential of hepatoma cells. World J Gastroenterol. 19:3792–3801. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Zare M, Bastami M, Solali S and Alivand MR: Aberrant miRNA promoter methylation and EMT-involving miRNAs in breast cancer metastasis: Diagnosis and therapeutic implications. J Cell Physiol. 233:3729–3744. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Feng J, Fu Z, Guo J, Lu W, Wen K, Chen W, Wang H, Wei J and Zhang S: Overexpression of peroxiredoxin 2 inhibits TGF-β1-induced epithelial-mesenchymal transition and cell migration in colorectal cancer. Mol Med Rep. 10:867–873. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Bezdenezhnykh N, Semesiuk N, Lykhova O, Zhylchuk V and Kudryavets Y: Impact of stromal cell components of tumor microenvironment on epithelial-mesenchymal transition in breast cancer cells. Exp Oncol. 36:72–78. 2014.PubMed/NCBI

14 

Mou H, Yu L, Zheng X, Liao Q, Hou X and Wu Y: p16 gene expression in pancreatic cancer tissue and its importance in diagnosis. J Biol Regul Homeost Agents. 31:1043–1047. 2017.PubMed/NCBI

15 

Luo S, Xie C, Wu P, He J, Tang Y, Xu J and Zhao S: Annexin A2 is an independent prognostic biomarker for evaluating the malignant progression of laryngeal cancer. Exp Ther Med. 14:6113–6118. 2017.PubMed/NCBI

16 

Murphy AG, Foley K, Rucki AA, Xia T, Jaffee EM, Huang CY and Zheng L: Stromal Annexin A2 expression is predictive of decreased survival in pancreatic cancer. Oncotarget. 8:106405–106414. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Shetty P, Patil VS, Mohan R, D'souza LC, Bargale A, Patil BR, Dinesh US, Haridas V and Kulkarni SP: Annexin A2 and its downstream IL-6 and HB-EGF as secretory biomarkers in the differential diagnosis of Her-2 negative breast cancer. Ann Clin Biochem. 54:463–471. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Cui JW and Wang YL: Expression and function of Annexin II in lung cancer tissue. Asian Pac J Trop Med. 6:150–152. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Dong Z, Yao M, Zhang H, Wang L, Huang H, Yan M, Wu W and Yao D: Inhibition of Annexin A2 gene transcription is a promising molecular target for hepatoma cell proliferation and metastasis. Oncol Lett. 7:28–34. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Kirshner J, Schumann D and Shively JE: CEACAM1, a cell-cell adhesion molecule, directly associates with annexin II in a three-dimensional model of mammary morphogenesis. J Bio Chem. 278:50338–50345. 2003. View Article : Google Scholar

21 

Yee DS, Narula N, Ramzy I, Boker J, Ahlering TE, Skarecky DW and Ornstein DK: Reduced annexin II protein expression in high-grade prostatic intraepithelial neoplasia and prostate cancer. Arch Pathol Lab Med. 131:902–908. 2007.PubMed/NCBI

22 

Ohno Y, Izumi M, Kawamura T, Nishimura T, Mukai K and Tachibana M: Annexin II represents metastatic potential in clear-cell renal cell carcinoma. Br J Cancer. 101:287–294. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Zhang W, Zhao P, Xu XL, Cai L, Song ZS, Cao DY, Tao KS, Zhou WP, Chen ZN and Dou KF: Annexin A2 promotes the migration and invasion of human hepatocellular carcinoma cells in vitro by regulating the shedding of CD147-harboring microvesicles from tumor cells. PLoS One. 8:e672682013. View Article : Google Scholar : PubMed/NCBI

24 

Oh M, Rhee S, Moon JH, Chae H, Lee S, Kang J and Kim S: Literature-based condition-specific miRNA-mRNA target prediction. PLoS One. 12:e01749992017. View Article : Google Scholar : PubMed/NCBI

25 

Dweep H, Sticht C, Pandey P and Gretz N: miRWalk-database: Prediction of possible miRNA binding sites by ‘walking’ the genes of three genomes. J Biomed Inform. 44:839–847. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Xiao H, Xiao W, Cao J, Li H, Guan W, Guo X, Chen K, Zheng T, Ye Z, Wang J and Xu H: miR-206 functions as a novel cell cycle regulator and tumor suppressor in clear-cell renal cell carcinoma. Cancer Lett. 374:107–116. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Ren XL, He GY, Li XM, Men H, Yi LZ, Lu GF, Xin SN, Wu PX, Li YL, Liao WT, et al: MicroRNA-206 functions as a tumor suppressor in colorectal cancer by targeting FMNL2. J Cancer Res Clin Oncol. 142:581–592. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Keklikoglou I, Hosaka K, Bender C, Bott A, Koerner C, Mitra D, Will R, Woerner A, Muenstermann E, Wilhelm H, et al: MicroRNA-206 functions as a pleiotropic modulator of cell proliferation, invasion and lymphangiogenesis in pancreatic adenocarcinoma by targeting ANXA2 and KRAS genes. Oncogene. 34:4867–4878. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Chen L, Li YS, Cui J, Ning JN, Wang GS, Qian GS, Lu KZ and Yi B: MiR-206 controls the phenotypic modulation of pulmonary arterial smooth muscle cells induced by serum from rats with hepatopulmonary syndrome by regulating the target gene, annexin A2. Cell Physiol Biochem. 34:1768–1779. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Scanlon CS, Van Tubergen EA, Inglehart RC and D'Silva NJ: Biomarkers of epithelial-mesenchymal transition in squamous cell carcinoma. J Dent Res. 92:114–121. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Martínez-Ramírez AS, Garay E, García-Carrancá A and Vázquez-Cuevas FG: The P2RY2 receptor induces carcinoma cell migration and EMT through cross-talk with epidermal growth factor receptor. J Cell Biochem. 117:1016–1026. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Carvalho-Cruz P, Alisson-Silva F, Todeschini AR and Dias WB: Cellular glycosylation senses metabolic changes and modulates cell plasticity during epithelial to mesenchymal transition. Dev Dyn. 247:481–491. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Kaushik NK, Kaushik N, Yoo KC, Uddin N, Kim JS, Lee SJ and Choi EH: Low doses of PEG-coated gold nanoparticles sensitize solid tumors to cold plasma by blocking the PI3K/AKT-driven signaling axis to suppress cellular transformation by inhibiting growth and EMT. Biomaterials. 87:118–130. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Yu AQ, Ding Y, Li CL, Yang Y, Yan SR and Li DS: TALEN-induced disruption of Nanog expression results in reduced proliferation, invasiveness and migration, increased chemosensitivity and reversal of EMT in HepG2 cells. Oncol Rep. 35:1657–1663. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Chen HN, Yuan K, Xie N, Wang K, Huang Z, Chen Y, Dou Q, Wu M, Nice EC, Zhou ZG and Huang C: PDLIM1 Stabilizes the E-Cadherin/β-Catenin complex to prevent epithelial-mesenchymal transition and metastatic potential of colorectal cancer cells. Cancer Res. 76:1122–1134. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 15 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang N, Wang L, Liu J, Liu L, Huang J, Chen X and Luo Z: MicroRNA‑206 regulates the epithelial‑mesenchymal transition and inhibits the invasion and metastasis of prostate cancer cells by targeting Annexin A2. Oncol Lett 15: 8295-8302, 2018
APA
Yang, N., Wang, L., Liu, J., Liu, L., Huang, J., Chen, X., & Luo, Z. (2018). MicroRNA‑206 regulates the epithelial‑mesenchymal transition and inhibits the invasion and metastasis of prostate cancer cells by targeting Annexin A2. Oncology Letters, 15, 8295-8302. https://doi.org/10.3892/ol.2018.8395
MLA
Yang, N., Wang, L., Liu, J., Liu, L., Huang, J., Chen, X., Luo, Z."MicroRNA‑206 regulates the epithelial‑mesenchymal transition and inhibits the invasion and metastasis of prostate cancer cells by targeting Annexin A2". Oncology Letters 15.6 (2018): 8295-8302.
Chicago
Yang, N., Wang, L., Liu, J., Liu, L., Huang, J., Chen, X., Luo, Z."MicroRNA‑206 regulates the epithelial‑mesenchymal transition and inhibits the invasion and metastasis of prostate cancer cells by targeting Annexin A2". Oncology Letters 15, no. 6 (2018): 8295-8302. https://doi.org/10.3892/ol.2018.8395