MicroRNA‑23a promotes neuroblastoma cell metastasis by targeting CDH1

  • Authors:
    • Lin Cheng
    • Tao Yang
    • Yongqin Kuang
    • Bin Kong
    • Sixun Yu
    • Haifeng Shu
    • Hutian Zhou
    • Jianwen Gu
  • View Affiliations

  • Published online on: January 14, 2014     https://doi.org/10.3892/ol.2014.1794
  • Pages: 839-845
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

CDH1 inactivation is important in tumor metastasis. In the present study, it was suggested that the mRNA and protein levels of CDH1 decreased in metastatic neuroblastoma (NB) tissues compared with those in primary NB tissues. The aim of the study was to explore the regulatory mechanisms of CDH1 downregulation in metastatic NB. MicroRNAs are small non‑coding RNAs (~22 nt in length) that negatively regulate target mRNAs and are involved in various cancer‑related processes, including metastasis. In the current study, miR‑23a was shown to be upregulated in human metastatic NB tissues compared with primary NB tissues. Inhibition of miR‑23a may significantly suppress NB cell migration and invasion. In vitro reporter assay suggested that CDH1 is a direct target gene of miR‑23a. Furthermore, blocking the expression of miR‑23a partly restored the expression of CDH1 in NB cells. These findings provide evidence that miR‑23a is key in promoting NB cell migration and invasion through targeting CDH1, and suggest that exogenous miR‑23a may have therapeutic value in treating NB metastasis.

Introduction

Characterized by spontaneous regression, maturation or aggressive progression, neuroblastoma (NB) is one of the most common forms of cancer in young children. NB typically presents with metastatic disease at diagnosis with a poor outcome (1,2). The most critical step in NB metastasis is that the cells from the primary tumor, which have enhanced capabilities of migration and invasion, invade into the surrounding normal tissues (3). To date, a set of genes associated with tumor progression in NB have been revealed (4). However, the precise mechanisms, particularly the significant molecular pathways in the transition from primary to metastatic NB, remain to be completely understood.

The CDH1 gene encodes the epithelial cell adhesion molecule, E-cadherin, which forms the core of the adherens junctions between adjacent epithelial cells (5). The significance of CDH1 inactivation for metastasis has been demonstrated in a variety of studies (611). However, few studies have analyzed the potential important roles of CDH1 in metastatic NB tissues.

Previously, a group of non-coding single-stranded RNAs, ~22 nt in length, which modulate gene expression post-transcriptionally by interacting with complementary sites within the 3′ untranslated region (UTR) of target mRNA, have emerged as important regulators in cancer-related processes, including metastasis (1215). In a previous study, Guo et al observed differential expression of microRNAs (miRNAs) in metastatic NB compared with primary NB using microarray analysis. In addition, the potential roles of these miRNAs in the NB metastatic process were investigated (16). However, the role of miR-23a in metastatic NB tissues compared with primary NB tissues remains unclear. Hence, we proposed that the upregulated miR 23a enhances the metastatic capability of NB cells. Exploring the miR-23a-mediated NB metastasis molecular mechanism may be useful to characterize the progression from primary to metastatic NB.

Materials and methods

Tissue samples and cell lines

In total, nine pairs of human primary and metastatic NB tissues were acquired from the Chengdu Military General Hospital (Chengdu, China). The study was approved by the ethics committee of Chengdu Military General Hospital and informed written consent was obtained from the patients. All the tissues were confirmed by pathology and immunohistochemistry, frozen in liquid nitrogen and stored at −80°C. Human NB cell lines, SK-N-SH and GI-LA-N, were cultured in Eagle’s minimum essential medium (Gibco-BRL, Carlsbad, CA, USA) supplemented with 10% FBS and 100 U/ml penicillin/streptomycin. All the cells were placed in a humidified incubator with 5% CO2 at 37°C.

RNA isolation and qPCR

Total RNA was isolated using TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA) according to the manufacturer’s instructions. The RNA concentration and quality was measured by a NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA). Next, 1 μg RNA was used for reverse transcription (RT). Specific miR-23a reverse transcription (RT) primers were used for the cDNA from the miRNA RT reaction, while the common primer Oligo (dT) was used for the cDNA from total RNA, for detection of CDH1. qPCR was performed using the SYBR Green PCR master mix (Applied Biosystems, Inc., Foster City, CA, USA) according to the following conditions: 95°C for 5 min followed by 40 cycles of amplification at 95°C for 30 sec, 57°C for 30 sec and 72°C for 30 sec. U6 small nuclear B non-coding RNA was used as the internal control to normalize miR-23a expression and GAPDH was used as the internal control to normalize CDH1 expression.

Western blot analysis

The NB cells were seeded into six-well plates at a density of 3×104 cells/well and cells were transfected once cell density reached ~80% confluence on the second day. At 48 h following transfection, the cells were lysed using RIPA buffer for 30 min at 4°C. The protein concentration was measured by the bicinchoninic acid method using a Pierce® BCA Protein Assay kit (Thermo Fisher Scientific) and then 20 μg protein was loaded on SDS-PAGE for analysis. The primary antibodies used were rabbit polyclonal anti-human CDH1 (1:500; Abcam, Cambridge, MA, USA) and rabbit monoclonal anti-human GAPDH (1:1,000; Abcam). The secondary antibody used was a goat anti-rabbit IgG conjugated with horseradish peroxidase (1:1,000; Abcam). The bound antibodies were detected by ECL Plus Western blotting detection system (GE Healthcare, Amersham, UK) and the chemiluminescent signals were detected by high-performance chemiluminescence film (GE Healthcare).

EGFP reporter assay

The 3′UTR of CDH1 was amplified and cloned downstream of the pcDNA3/EGFP vector. Next, the mutant 3′UTR of CDH1 (in which several nucleotides within the binding sites were deleted) was amplified using pcDNA3/EGFP-CDH1 3′UTR as the template and cloned downstream of the pcDNA3/EGFP vector. For the EGFP reporter assay, the cells were co-transfected with miR-23a mimics, ASO-miR-23a and pcDNA3/EGFP-CDH1 3′UTR or the mutant 3′UTR, together with the controls. The plasmid expressing RFP was transfected as the spike-in control. At 48 h following transfection, the cells were lysed using RIPA buffer, and EGFP and RFP intensity was measured by an F-4500 fluorescence spectrophotometer (Hitachi, Ltd., Tokyo, Japan).

Transwell migration and invasion assays

The migration and invasion assays were performed using the Transwell chamber (Millipore, Billerica, MA, USA). For the migration assay, following transfection, the cells were seeded into the upper chamber (2.5×104 cells/well) in 250 μl serum-free medium, but the bottom chamber was incubated with 750 μl medium containing 10 or 20% serum. For the invasion assay, the chamber was coated with Matrigel and the cells were seeded into the upper chamber (2.5×104 cells/well) in 250 μl serum-free medium, while the bottom chamber was incubated with 750 μl medium containing 10 or 20% serum. Following cell migration or invasion for 20 h, the cells that had not migrated or invaded were scraped off with a cotton swab. The cells which had migrated or invaded into the chamber membrane were fixed and then stained with crystal violet for ~15 min. Finally, images of the cells were captured under a microscope (ME21 digital microscope, Olympus, Guangzhou, China) and cells were counted.

Statistics

Data are presented as the mean ± standard deviation and the difference between groups was determined by a two-tailed Student’s t-test. P<0.01 or P<0.05 were considered to indicate a statistically significant difference.

Results

Quantitative analysis of CDH1 expression in metastatic NB tissues

CDH1 is an important cancer metastasis-related molecule. To test the expression of CDH1 in metastatic NB tissues and matched primary NB tissues, qPCR assay was performed in nine pairs of NB tissues. It was shown that CDH1 mRNA expression levels were generally and significantly (P<0.01) lower in metastatic NB tissues than in the matched primary NB tissues (Fig. 1A). In addition, western blot analysis confirmed that the CDH1 protein expression was lower in metastatic NB tissues compared with that in primary NB tissues within the representative patients 3, 6 and 7.

miR-23a is a candidate regulator of CDH1 in NB

miRNAs function as tumor suppressors or oncogenes, through the direct regulation of associated oncogenes or tumor suppressor genes (14,17,18). The oncogene miRNAs (onco-miRs), such as miR-23a, are usually upregulated in tumors and may cause the loss of expression of tumor suppressors and contribute to the tumorigenesis of cancer, including metastasis. The loss of CDH1 expression in metastatic NB tissues implies that there may be important miRNAs involved in the regulation of CDH1 in NB metastasis. Greater specificity in miRNA predictions may be attained by the consensus of multiple algorithms. Therefore, three programs (TargetScan, microRNA.org and miRDB) were used for the prediction of which miRNAs target the 3′UTR of CDH1 and regulate its expression. Table I shows the 20 miRNAs that were identified to target the 3′UTR of CDH1. In addition, Table II shows the 34 downregulated miRNAs collected from metastatic NB tissues to compare with primary NB tissues, according to the previous study by Guo et al using the microarray analysis (16). Based on these two tables, miR-23a (Fig. 2) was identified as a candidate for directly targeting CDH1 whose mRNA 3′UTR contains a putative binding site. This analysis is consistent with the model in which tumor onco-miRs were found to promote tumor development by targeting and negatively regulating tumor suppressors (13).

Table I

Predicted miRNAs targeting CDH1 by three algorithms.

Table I

Predicted miRNAs targeting CDH1 by three algorithms.

Gene namemiRNAmicroRNA.orgmiRDBTargetScan
CDH1hsa-miR-1248+++
CDH1hsa-miR-219-2-3p+++
CDH1hsa-miR-504+++
CDH1hsa-miR-340+++
CDH1hsa-miR-580+++
CDH1hsa-miR-296-3p+++
CDH1hsa-miR-1299+++
CDH1hsa-miR-1296+++
CDH1hsa-miR-515-5p+++
CDH1hsa-miR-1323+++
CDH1hsa-miR-1264+++
CDH1hsa-miR-548o+++
CDH1hsa-miR-619+++
CDH1hsa-miR-544+++
CDH1hsa-miR-339-5p+++
CDH1hsa-miR-23b+++
CDH1hsa-miR-23a+++
CDH1 hsa-miR-1207-5p+++
CDH1hsa-miR-219-5p+++
CDH1hsa-miR-338-3p+++

[i] miRNA, microRNA.

Table II

Differential expression of miRNAs in NB between metastatic and primary tumors (increased expression, >2-fold).

Table II

Differential expression of miRNAs in NB between metastatic and primary tumors (increased expression, >2-fold).

miRNAFold changes (metastatic/primary tumors)
hsa-miR-342-3p6.2879
hsa-miR-92b11.8088
hsa-miR-130b2.5771
hsa-miR-129-3p3.0205
hsa-miR-483-3p9.9583
hsa-miR-3453.1005
hsa-miR-245.0395
hsa-miR-486-5p2.4739
hsa-miR-99b3.2086
hsa-miR-1454.9442
hsa-miR-3753.5245
hsa-miR-92a5.3560
hsa-miR-361-5p4.0653
hsa-miR-4842.9674
hsa-miR-339-5p3.8349
hsa-miR-148b4.3914
hsa-miR-140-3p3.7209
hsa-miR-3352.5920
hsa-miR-483-5p3.2567
hsa-miR-23a2.5497
hsa-miR-29a2.4728
hsa-miR-423-3p2.0051
hsa-miR-128a2.3610
hsa-miR-130a3.1655
hsa-miR-2142.8887
hsa-miR-15a2.5875
hsa-miR-1912.3789
hsa-miR-1002.2647
hsa-miR-10b3.1618
hsa-miR-29b2.4477
hsa-miR-30c2.5702
hsa-miR-324-5p2.2561
hsa-miR-6024.4085
hsa-miR-1072.1387

[i] Taken from (16). miRNA, microRNA; NB, neuroblastoma.

miRNA-23a is upregulated in metastatic NB tissues and regulates NB cell migration and invasion

miR-23a has been previously reported to play a role as an onco-miR in several types of cancer, and the microarray analysis identified that miR-23a is upregulated in metastatic NB tissues compared with primary NB tissues. Next, qPCR assay was used to detect the expression of miR-23a in nine pairs of NB samples (metastatic and matched primary NB tissues). It was shown that miR-23a expression levels were generally higher in metastatic NB tissues compared with primary NB tissues (Fig. 3A). Transwell migration (without Matrigel) and invasion (with Matrigel) assays were used to evaluate the effect of miR-23a on NB cell migration and invasion. As Fig. 3C shows, blocking the expression of miR-23a with miR-23a ASO significantly decreased the number of invaded SK-N-SH and GI-LA-N cells compared with the ASO control group. This implies that the upregulated miR-23a promotes the invasion of NB cells. Consistent with the invasion assay results, blockage of miR-23a significantly inhibited the migration of SK-N-SH and GI-LA-N cells compared with the control group (Fig. 3D). Thus, it was concluded that miR-23a is overexpressed in metastatic NB tissues and inhibition of miR-23a suppresses the migration and invasion of NB cells.

miR-23a directly targets CDH1 expression in NB cells

To verify that the regulation of CDH1 expression by miR-23a is direct, an EGFP reporter system was used in SK-N-SH cells. The alignment of miR-23a with the CDH1 3′UTR insert is illustrated in Fig. 4A. In addition, a mutated 3′UTR was constructed by deleting 4 nt in the seed sequence of miR-23a (asterisk marked in Fig. 4A). The EGFP reporter analysis showed that with the CDH1-3′UTR insert, the EGFP intensity was significantly lower than that in the CDH1-3′UTR-mutant group when treated with control oligo. When miR-23a was blocked by miR-23a ASO, the EGFP intensity levels were significantly higher than those in the control group. However, the EGFP intensity with the mutated 3′UTR was not affected by miR-23a (Fig. 4B). Next, it was investigated whether the endogenous CDH1 is regulated by miR-23a in NB cells. qPCR assay revealed that the CDH1 mRNA levels were inversely correlated with the miR-23a levels (Fig. 4C). The trend of CDH1 protein levels was similar to that of the mRNA levels, according to the western blot analysis (Fig. 4D). Therefore, it was concluded that CDH1 is a direct target of miR-23a in SK-N-SH cells, and that miR-23a regulates the expression of CDH1 at the mRNA and protein levels.

CDH1 suppresses NB cell migration and invasion in vitro

Previous studies have shown that the downregulation of CDH1 is an important feature of a number of transformed cells (19), and that CDH1 functions as a tumor suppressor. Accordingly, the present study investigated whether CDH1 affects NB cell metastasis. Overexpression plasmids (pCMV6/CDH1) were constructed that ectopically overexpress the CDH1 protein in NB cells. Subsequently, the effect of CDH1 on cell metastasis was evaluated by Transwell assay. The results showed that the invasion rate was reduced by ~50 and ~60% with the overexpression of CDH1 in SK-N-SH and GI-LA-N cells, respectively, compared with that of the control group (Fig. 5A). In addition, the migration rate of the SK-N-SH and GI-LA-N cells transfected with pCMV6/CDH1 was markedly lower than that of the control group (Fig. 5B). These results provided further evidence that CDH1 is a tumor suppressor in NB metastasis. The tumor suppressor role of CDH1 in NB metastasis may explain the manner in which the upregulation of miR-23a promotes NB cell migration and invasion and contribute to the transformation of primary to metastatic NB (Fig. 5C).

Discussion

As in the case of various types of cancer metastasis, the transformation of primary to metastatic NB is a complex multistep process including cell adhesion, migration, angiogenesis, immune escape and homing to target organs. The increased NB cell migration and invasion capabilities are essential features of the metastatic transformation process. Identifying the molecules and pathways that control NB cell migration and invasion is critical to understanding NB metastasis.

Accumulating previous evidence has suggested that genetic or epigenetic alterations in CDH1 or alterations in their protein expression, often result in tissue disorder, cellular dedifferentiation, increased invasiveness of tumor cells and ultimately metastasis (1921). It has been previously demonstrated that the downregulation of CDH1 leads to loss of cell polarity and derangement of normal tissue architecture (22,23). In various types of cancer, CDH1-mediated cell-cell adhesion is lost concomitantly with acquisition of an invasive phenotype, high tumor grade and low patient survival rates (2429). Based on the important role of CDH1 in cancer tumorigenesis, the investigation of the regulatory mechanisms of CDH1 in the process of primary to metastatic NB transition may highlight new prognostic markers and therapy targets for the metastasis of NB in the future. To date, somatic mutations, chromosomal deletions, proteolytic cleavage and silencing of the CDH1 promoter have been reported to cause the loss or reduction of CDH1 expression (3032). In a previous study by Hoebeeck et al, the authors tested 42 primary NB tumors and the frequencies of methylation were 8% with CDH1 (19). This implies that there may be potential other mechanisms that are involved in the downregulation of CDH1 in the NB metastatic process. In the present study, nine pairs of metastatic NB tissues and matched primary NB tissues were tested, and the mRNA and protein levels of CDH1 were found to be significantly decreased in the NB metastatic transformation process (Fig. 1).

Previously, miRNAs have been identified as important regulators of gene expression. Several miRNAs have been found to exhibit prometastatic (miR-10b, -21 and -373/520c) or antimetastatic (miR-34b/c, -126, -148a, -206 and -335) activity (3335). In a previous study, miRNA microarray assay was performed to screen out the miRNAs that were differentially expressed between metastatic and primary NB tissues (16). In addition, a previous study has shown that miR-9, which is directly bound and upregulated by MYC and MYCN in breast cancer cells, directly targets CDH1, leading to increased cell motility and invasiveness (26). miR-10b has been found to modulate breast cancer metastasis by targeting CDH1 and may be a useful biomarker of advanced progression and metastasis of breast cancer (36). miR-25 has also been demonstrated to promote esophageal squamous cell carcinoma cell migration and invasion by directly targeting CDH1 (37). Recently, Cao et al (38) found that miR-23a regulates TGF-β-induced epithelial-mesenchymal transition by targeting E-cadherin (CDH1) in lung cancer cells. However, few studies have considered the manner in which the miRNAs contribute to the transformation of primary to metastatic NB through the CDH1-induced pathway. Therefore, the current study analyzed the microarray results (Table II) and miRNA target prediction algorithms (Table I). miR-23a was identified as a candidate molecule in the regulation of NB metastasis by the CDH1 pathway (Fig. 2).

miR-23a belongs to the miR-23a/24/27a cluster, which is located in chromosome 19p13.12 and may be induced by TGF-β. In several types of human cancer, this gene cluster functions as an oncogenic miRNA and has been previously reported to be upregulated in types of human cancer. As one of the most famous members of the miRNA-cluster, miR-23a has been shown to promote the growth of gastric adenocarcinoma cells by targeting the interleukin-6 receptor (39,40). miR-23a has also been demonstrated to promote colon carcinoma cell growth, invasion and metastasis through inhibition of the MTSS gene (41,42). The current study revealed that miR-23a is upregulated in metastatic NB tissues when compared with primary NB tissues. In addition, blockage of miR-23a may reduce the capability of NB cell migration and invasion using Transwell assays (Fig. 3). To identify the direct regulation of CDH1 by miR-23a in NB cells, qPCR, western blot analysis and EGFP report system were used to demonstrate that CDH1 is a direct target gene of miR-23a (Fig. 4). The Transwell assay results by overexpression of CDH1 in SK-N-SH and GI-LA-N cells showed that the CDH1 may inhibit the capability of NB cell migration and invasion (Fig. 5). This was found to correlate with the blocking of the expression of miR-23a. These results suggest that there exists a miR-23a/CDH1 pathway, which is important in the regulation of NB, particularly in the process of primary to metastatic NB transition (Fig. 5C).

In conclusion, the results of the present study may be summarized by the following six major observations: i) CDH1 was inactive in the metastatic NB tissues compared with the primary NB tissues at the mRNA and protein levels; ii) bioinformatic software, including TargetScan, microRNA.org and miRDB algorithms, were used and miR-23a was identified as a candidate regulator of CDH1 in the transformation process of primary to metastatic NB; iii) miR-23a was found to be upregulated in metastatic NB tissues compared with primary NB tissues; iv) CDH1 is negatively regulated by miR-23a at the mRNA and protein levels and is a direct target in NB; v) inhibition of miR-23a suppresses the migration and invasiveness of SK-N-SH and GI-LA-N cells; and vi) there exists a miR-23a/CDH1 pathway in the transition of primary to metastatic NB. However, whether miR-23a may become a new prognostic marker and therapy target for the metastasis of NB must be explored in future studies.

Acknowledgements

The current study was supported by the National Natural Science Foundation of China (grant nos. 81071037 and 81271395).

References

1 

Maris JM, Hogarty MD, Bagatell R and Cohn SL: Neuroblastoma. Lancet. 369:2106–2120. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Maris JM: Recent advances in neuroblastoma. N Engl J Med. 362:2202–2211. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Steeg PS: Tumor metastasis: mechanistic insights and clinical challenges. Nat Med. 12:895–904. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Nevo I, Oberthuer A, Botzer E, et al: Gene-expression-based analysis of local and metastatic neuroblastoma variants reveals a set of genes associated with tumor progression in neuroblastoma patients. Int J Cancer. 126:1570–1581. 2010.

5 

Gumbiner BM: Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol. 6:622–634. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Ceteci F, Ceteci S, Karreman C, et al: Disruption of tumor cell adhesion promotes angiogenic switch and progression to micrometastasis in RAF-driven murine lung cancer. Cancer Cell. 12:145–159. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Derksen PW, Liu X, Saridin F, et al: Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell. 10:437–449. 2006. View Article : Google Scholar

8 

Onder TT, Gupta PB, Mani SA, Yang J, Lander ES and Weinberg RA: Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 68:3645–3654. 2008. View Article : Google Scholar

9 

Vleminckx K, Vakaet L Jr, Mareel M, Fiers W and van Roy F: Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 66:107–119. 1991. View Article : Google Scholar : PubMed/NCBI

10 

Frixen UH, Behrens J, Sachs M, et al: E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. J Cell Biol. 113:173–185. 1991. View Article : Google Scholar : PubMed/NCBI

11 

Perl AK, Wilgenbus P, Dahl U, Semb H and Christofori G: A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 392:190–193. 1998. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Gabriely G, Wurdinger T, Kesari S, et al: MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol Cell Biol. 28:5369–5380. 2008. View Article : Google Scholar : PubMed/NCBI

14 

O’Donnell KA, Wentzel EA, Zeller KI, Dang CV and Mendell JT: c-Myc-regulated microRNAs modulate E2F1 expression. Nature. 435:839–843. 2005.PubMed/NCBI

15 

Pillai RS, Bhattacharyya SN and Filipowicz W: Repression of protein synthesis by miRNAs: how many mechanisms? Trends Cell Biol. 17:118–126. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Guo J, Dong Q, Fang Z, et al: Identification of miRNAs that are associated with tumor metastasis in neuroblastoma. Cancer Biol Ther. 9:446–452. 2010. View Article : Google Scholar

17 

Johnson SM, Grosshans H, Shingara J, et al: RAS is regulated by the let-7 microRNA family. Cell. 120:635–647. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Cimmino A, Calin GA, Fabbri M, et al: miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA. 102:13944–13949. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Hoebeeck J, Michels E, Pattyn F, et al: Aberrant methylation of candidate tumor suppressor genes in neuroblastoma. Cancer Lett. 273:336–346. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Paredes J, Figueiredo J, Albergaria A, et al: Epithelial E- and P-cadherins: role and clinical significance in cancer. Biochim Biophys Acta. 1826:297–311. 2012.PubMed/NCBI

21 

Jiang T, Jiang H, Su XM, et al: Expressions of E-cadherin and alpha-catenin in benign, malignant and metastatic prostate tumors. Zhonghua Nan Ke Xue. 18:499–503. 2012.(In Chinese).

22 

Larue L, Ohsugi M, Hirchenhain J and Kemler R: E-cadherin null mutant embryos fail to form a trophectoderm epithelium. Proc Natl Acad Sci USA. 91:8263–8267. 1994. View Article : Google Scholar : PubMed/NCBI

23 

Karayiannakis AJ, Syrigos KN, Savva A, Polychronidis A, Karatzas G and Simopoulos C: Serum E-cadherin concentrations and their response during laparoscopic and open cholecystectomy. Surg Endosc. 16:1551–1554. 2002. View Article : Google Scholar

24 

Alexiou D, Karayiannakis AJ, Syrigos KN, et al: Serum levels of E-selectin, ICAM-1 and VCAM-1 in colorectal cancer patients: correlations with clinicopathological features, patient survival and tumour surgery. Eur J Cancer. 37:2392–2397. 2001. View Article : Google Scholar

25 

Cavallaro U and Christofori G: Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 4:118–132. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Karatzas G, Karayiannakis AJ, Syrigos KN, et al: E-cadherin expression correlates with tumor differentiation in colorectal cancer. Hepatogastroenterology. 46:232–235. 1999.PubMed/NCBI

27 

Karayiannakis AJ, Syrigos KN, Chatzigianni E, et al: Aberrant E-cadherin expression associated with loss of differentiation and advanced stage in human pancreatic cancer. Anticancer Res. 18:4177–4180. 1998.PubMed/NCBI

28 

Karayiannakis AJ, Syrigos KN, Chatzigianni E, Papanikolaou S and Karatzas G: E-cadherin expression as a differentiation marker in gastric cancer. Hepatogastroenterology. 45:2437–2442. 1998.PubMed/NCBI

29 

Syrigos KN, Krausz T, Waxman J, et al: E-cadherin expression in bladder cancer using formalin-fixed, paraffin-embedded tissues: correlation with histopathological grade, tumour stage and survival. Int J Cancer. 64:367–370. 1995. View Article : Google Scholar

30 

Berx G, Becker KF, Höfler H and van Roy F: Mutations of the human E-cadherin (CDH1) gene. Hum Mutat. 12:226–237. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Maretzky T, Reiss K, Ludwig A, et al: ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and beta-catenin translocation. Proc Natl Acad Sci USA. 102:9182–9187. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Strathdee G: Epigenetic versus genetic alterations in the inactivation of E-cadherin. Semin Cancer Biol. 12:373–379. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Voorhoeve PM, le Sage C, Schrier M, et al: A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell. 124:1169–1181. 2006. View Article : Google Scholar

34 

Zhu S, Si ML, Wu H and Mo YY: MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem. 282:14328–14336. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Ma L, Teruya-Feldstein J and Weinberg RA: Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 449:682–688. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Liu Y, Zhao J, Zhang PY, et al: MicroRNA-10b targets E-cadherin and modulates breast cancer metastasis. Med Sci Monit. 18:BR299–308. 2012.PubMed/NCBI

37 

Xu X, Chen Z, Zhao X, et al: MicroRNA-25 promotes cell migration and invasion in esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 421:640–645. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Cao M, Seike M, Soeno C, et al: MiR-23a regulates TGF-β-induced epithelial-mesenchymal transition by targeting E-cadherin in lung cancer cells. Int J Oncol. 41:869–875. 2012.

39 

Zhu LH, Liu T, Tang H, et al: MicroRNA-23a promotes the growth of gastric adenocarcinoma cell line MGC803 and downregulates interleukin-6 receptor. FEBS J. 277:3726–3734. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Huang S, He X, Ding J, et al: Upregulation of miR-23a approximately 27a approximately 24 decreases transforming growth factor-beta-induced tumor-suppressive activities in human hepatocellular carcinoma cells. Int J Cancer. 123:972–978. 2008. View Article : Google Scholar

41 

Jahid S, Sun J, Edwards RA, et al: miR-23a promotes the transition from indolent to invasive colorectal cancer. Cancer Discov. 2:540–553. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Wang Z, Wei W and Sarkar FH: miR-23a, a critical regulator of ‘migR’ation and metastasis in colorectal cancer. Cancer Discov. 2:489–491. 2012.

Related Articles

Journal Cover

2014-March
Volume 7 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
Cheng L, Yang T, Kuang Y, Kong B, Yu S, Shu H, Zhou H and Gu J: MicroRNA‑23a promotes neuroblastoma cell metastasis by targeting CDH1. Oncol Lett 7: 839-845, 2014
APA
Cheng, L., Yang, T., Kuang, Y., Kong, B., Yu, S., Shu, H. ... Gu, J. (2014). MicroRNA‑23a promotes neuroblastoma cell metastasis by targeting CDH1. Oncology Letters, 7, 839-845. https://doi.org/10.3892/ol.2014.1794
MLA
Cheng, L., Yang, T., Kuang, Y., Kong, B., Yu, S., Shu, H., Zhou, H., Gu, J."MicroRNA‑23a promotes neuroblastoma cell metastasis by targeting CDH1". Oncology Letters 7.3 (2014): 839-845.
Chicago
Cheng, L., Yang, T., Kuang, Y., Kong, B., Yu, S., Shu, H., Zhou, H., Gu, J."MicroRNA‑23a promotes neuroblastoma cell metastasis by targeting CDH1". Oncology Letters 7, no. 3 (2014): 839-845. https://doi.org/10.3892/ol.2014.1794