MicroRNA‑675 inhibits cell proliferation and invasion in melanoma by directly targeting metadherin

Retraction in: /10.3892/mmr.2023.12986

  • Authors:
    • Ke Liu
    • Junjun Jin
    • Kunjie Rong
    • Lukai Zhuo
    • Pingsong Li
  • View Affiliations

  • Published online on: December 12, 2017     https://doi.org/10.3892/mmr.2017.8264
  • Pages: 3372-3379
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Melanoma is derived from melanocytes and accounts for ~80% of skin cancer-associated fatalities worldwide. The dysregulation of microRNAs (miRNAs/miRs) is involved in the development and progression of melanoma. Therefore, miRNAs may be novel diagnostic or prognostic biomarkers and promising therapeutic targets in the treatment of patients with melanoma. miR‑675 is differentially expressed in several types of human cancer and has important roles in the pathogenesis of several diseases. However, the expression levels and the biological roles of miR‑675 in melanoma remain unclear. Therefore, the present study aimed to assess the expression of miR‑675 in melanoma, explore the effects of miR‑675 on melanoma cells and investigate the underlying molecular mechanisms that may be involved in the actions of miR‑675. The present study indicated that miR‑675 expression was downregulated in melanoma tissues and cell lines. Functional assays demonstrated that the upregulation of miR‑675 impaired cell proliferation and invasion in melanoma. Bioinformatics analysis, luciferase reporter assay, reverse transcription‑quantitative polymerase chain reaction and western blot analysis demonstrated that metadherin (MTDH) was a direct target of miR‑675 in melanoma. The MTDH levels were upregulated in melanoma tissues and inversely correlated with the miR‑675 expression. Furthermore, restored MTDH expression rescued the inhibition effects in melanoma cells caused by miR‑675 overexpression. Thus, miR‑675 may be a potential therapeutic target for melanoma.

Introduction

Melanoma, one of the most aggressive forms of skin cancer, is derived from melanocytes and accounts for approximately 80% of skin cancer-related deaths worldwide (1). The incidence of melanoma has increased considerably compared with that of other malignancies around the world (2). Approximately 200,000 new cases and 46,000 deaths due to melanoma are reported worldwide (3). Currently, the major therapies for patients with melanoma include surgical resection, biotherapy, radiotherapy and chemotherapy (4). Despite the advancements in cancer therapy, melanoma is still considered to be therapy resistant with a 5-year survival rate of less than 15% (5,6). The poor prognosis of melanoma is mainly caused by the melanocytes showing aberrant proliferation, resistance to apoptosis and highly invasive potential and motility capacity, which are important biological characteristics in the aggressive clinical course of the metastatic disease (6,7). Therefore, further investigation on the molecular mechanism underlying the formation and progression of melanoma is needed to develop novel therapeutic targets for melanoma treatment.

MicroRNA (miRNA/miR) is a type of non-coding, single-strand, endogenous and short RNA molecules consisting of 19–22 nucleotides (8). miRNAs play crucial roles in gene regulation through interaction with the 3′-untranslational regions (3′-UTRs) of their target genes in a base-pairing manner, resulting in cleaving or repressing the translation of their target mRNAs at the posttranscriptional level (9). At present, over one thousand miRNAs have been identified in the human genome, which may regulate the expression of approximately 30% of all protein-coding genes (10). Thus, miRNAs are found to be involved in the regulation of diverse physiological and pathological processes, such as cell proliferation, cycle, apoptosis, differentiation, migration, invasion, metastasis, motility and angiogenesis (1113). Many studies have reported that the aberrant expression of miRNAs is associated with many disorders, particularly cancers such as melanoma (1416). A large number of miRNAs have been identified as tumour suppressors or oncogenes in different types of human malignancy, as a result of a change in the expression level of their target genes (1719). These findings provide a strong basis for the importance of miRNAs in the tumorigenesis and tumour development and emphasise the implications of miRNAs as potential therapeutic targets for cancer diagnosis, treatment and prognosis.

miR-675 is differentially expressed in several types of human cancer and plays important roles in the pathogenesis of several diseases (2022), However, the expression levels and the biological roles of miR-675 in melanoma remain unclear. Therefore, the present study aimed to assess the expression of miR-675 in melanoma, explore the effects of miR-675 on melanoma cells and investigate the underlying molecular mechanisms that may be involved in the actions of miR-675.

Materials and methods

Tissue samples and cell lines

This study was approved by the Ethics Committee of Subei People's Hospital of Jiangsu province. Written informed consent was provided by all of these patients participated in this research. From May 2014 to February 2016, a total of 21 pairs of melanoma tissues and corresponding adjacent normal tissues were gathered from melanoma patients who suffered a surgical resection at Subei People's Hospital of Jiangsu province. None patients had been treated with chemotherapy, radiotherapy or other treatment prior to surgery. All tissue samples were immediately frozen in liquid nitrogen and stored in −80°C until further use.

Melanoma cell lines, including A375, A2058, HT144 and SK-MEL-28, were obtained from the American Type Culture Collection (Manassas, VA, USA), and cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% foetal bovine serum (FBS), 100 mg/ml penicillin and 100 mg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Human epidermal melanocytes (HEM) were acquired from ScienCell Research Laboratories, Inc. (San Diego, CA, USA) and maintained in melanocyte medium (ScienCell Research Laboratories, Inc.) according to the manufacturer's protocol. All the cell lines were grown in a humid incubator at 37°C with 5% CO2.

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

Total RNA was isolated from tissue samples or cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. To detect the levels of miR-675, complementary DNA (cDNA) was synthesized from total RNA using TaqMan MicroRNA Reverse Transcription Kit (Applied Biosystems; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Quantitative PCR was performed using TaqMan MicroRNA PCR Kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) on an ABI Prism 7500 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). To quantify metadherin (MTDH) mRNA expression level, reverse transcription was carried out with PrimeScript RT Reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). SYBR Premix Ex Taq™ (Takara Biotechnology Co., Ltd.) was used to detect MTDH mRNA expression. U6 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were used as normalization for miR-675 and MTDH mRNA expression, respectively. Each experiment was performed in triplicate and repeated three times. The relative levels of miRNA and mRNA were analysed via the 2−ΔΔCq method (23).

Oligonucleotides, plasmids and cell transfection

The miR-675 mimics and miRNA negative control mimics (miR-NC) were chemically synthesized by GenePharma (Shanghai, China). MTDH overexpression plasmid (pcDNA3.1-MTDH) and empty pcDNA3.1 plasmid were acquired from GeneCopoeia (Guangzhou, China). Cells were seeded into 6-well plates at a density of 5×105 cells per well. Cell transfection was performed using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) as recommended by the manufacturer's instructions. The medium was replaced with fresh DMEM medium containing 10% FBS at 8 h following transfection.

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

MTT assays were employed to evaluate the proliferative ability of the melanoma cells. Transfected cells were collected at 24 h following transfection, and plated into 96-well plates at a density of 3,000 cells/well. At the indicated time (0, 24, 48, or 72 h incubation), MTT assay was performed according to the manufacturer's instructions. Briefly, 20 µl of MTT (5 mg/ml; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) reagent was added into each well. After incubation at 37°C with 5% CO2 for 4 h, the medium was removed followed by addition of 150 µl of DMSO (Sigma-Aldrich; Merck KGaA). Finally, the absorbance was measured using a microplate reader (Omega Bio-Tek, Inc., Norcross, GA, USA) at a wavelength of 490 nm. Experiments were independently repeated in triplicate.

Transwell invasion assay

The invasiveness of melanoma cells was determined using Transwell chambers (24-well plate, 8 µm pore; Corning Incorporated, Corning, NY, USA) coated with Matrigel (BD Bioscience, San Jose, CA, USA). Transfected cells were collected after 48 h of incubation. 5×104 transfected cells in 200 µl FBS-free DMEM medium were seeded in the upper chambers. The lower chambers were filled with DMEM with 20% FBS as a chemoattractant. Following incubation at 37°C for 24 h, the cells remaining on the upper chambers were removed using cotton swabs. The invasive cells were fixed with 4% paraformaldehyde, stained with 0.5% crystal violet and dried in air. A total of five randomly selected fields were then examined under an inverted microscope (IX73; Olympus Corporation, Tokyo, Japan) at a magnification of 200x. Each assay was performed in triplicate and repeated at least three times.

Bioinformatics analysis

To predict the potential targets of miR-675, bioinformatics analysis was performed using TargetScan (www.targetscan.org) and miRBase (http://www.mirbase.org/).

Luciferase reporter assay

MTDH was identified as a candidate target of miR-675 using bioinformatics analysis. A wild-type (Wt) MTDH 3′-UTR containing the binding sequences of miR-675 (pGL3-MTDH-3′-UTR Wt) and a mutant type (Mut) MTDH 3′-UTR lacking the binding sequences of miR-675 (pGL3-MTDH-3′-UTR Mut) were chemically synthesized by GenePharma. Cells were plated into 24-well plates at a density of 1.5×105 cells/well. After incubation overnight, cells were cotransfected with miR-675 mimics or miR-NC, and pGL3-MTDH-3′-UTR Wt or pGL3-MTDH-3′-UTR Mut, using Lipofectamine 2000 in accordance with the manufacturer's instructions. Luciferase activities were measured 48 h after transfection using a Dual-Luciferase® Reporter Assay System (Promega Corporation, Madison, WI, USA), according to the manufacturer's instructions. Renilla luciferase activity served as an internal reference.

Western blotting analysis

Total protein was extracted from tissues or cells using cold radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China). The concentration of total protein was examined using a bicinchoninic acid (BCA) Protein Assay kit (Beyotime Institute of Biotechnology), according to the manufacturer's instructions. Equal quantity of total protein was separated by 10% sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) and transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). Subsequently, the membranes were blocked with 5% skimmed milk in TBS/0.1% Tween (TBST) at room temperature for 1 h and incubated overnight at 4°C with primary antibodies: mouse anti-human monoclonal MTDH (sc-517220; 1:1,000 dilution; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and mouse anti-human monoclonal GAPDH (sc-66163; 1:1,000 dilution; Santa Cruz Biotechnology, Inc.). Following three washes with TBST, the membranes were incubated with corresponding horseradish peroxidase-conjugated secondary antibodies (sc-2005; 1:1,000 dilution; Santa Cruz Biotechnology, Inc.) at room temperature for 1 h. Finally, the protein bands were visualized with enhanced chemiluminescence (ECL) reagents (Bio-Rad Laboratories, Hercules, CA, USA), and analyzed with ImageJ 1.49 (National Institutes of Health, Bethesda, MD). GAPDH served as a loading control.

Statistical analysis

Data were expressed as mean ± standard deviation. All statistical analyses were performed via Student's t-test or one-way ANOVA with SPSS 18.0 software (SPSS, Inc., Chicago, IL, USA). Student-Newman-Keuls test was used as a post hoc test following ANOVA. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-675 expression is downregulated in melanoma tissues and cell lines

To investigate the roles of miR-675 in the progression of melanoma, RT-qPCR was performed to measure miR-675 expression in 21 paired melanoma tissues and corresponding adjacent normal tissues. As shown in Fig. 1A, miR-675 was significantly downregulated in melanoma tissues compared with that in corresponding adjacent normal tissues (P<0.05). In addition, the analysis of miR-675 expression in four human melanoma cell lines (A375, A2058, HT144 and SK-MEL-28) and the human epidermal melanocytes (HEMs) indicated that expression levels of miR-675 were lower in melanoma cell lines (Fig. 1B; P<0.05). These results suggested that miR-675 may serve important roles in melanoma formation and progression.

miR-675 inhibits melanoma cell proliferation and invasion in vitro

miR-675 is lowly expressed in melanoma and thus might serve as a tumour suppressor. Therefore, we determined whether the deregulation of miR-675 in malignant melanoma cells affects biological behaviours of melanoma. A375 and HT144 cells, which exhibited relatively low miR-675 expression, were chosen for subsequent functional assays and transfected with miR-675 mimics or miR-NC. The data from RT-qPCR analysis confirmed that miR-675 was markedly upregulated in A375 and HT144 cells after transfection with miR-675 mimics (Fig. 2A; P<0.05).

An MTT assay was conducted to explore effect of miR-675 overexpression on melanoma cell proliferation. Restoration expression of miR-675 suppressed the proliferation of A375 and HT144 compared with the miR-NC group (Fig. 2B; P<0.05), indicating that miR-675 may serve a suppressive role in the proliferation of melanoma cells. To further characterise the effect of miR-675 on cell invasion ability, Transwell invasion assays were performed. As depicted in Fig. 2C, the upregulation of miR-675 decreased the invasion capacities of A375 and HT144 cells, relative to the miR-NC group (P<0.05). Collectively, these data suggested that miR-675 may play tumour-suppressing roles in melanoma.

MTDH is a direct target of miR-675 in melanoma

To investigate the underlying mechanism responsible for miR-675-mediated tumour-suppressing roles in melanoma, bioinformatics analysis was performed to predict the potential targets of miR-675. Among these potential target genes, MTDH (Fig. 3A) caught our attention because it is upregulated in melanoma and participated in the regulation of biological behaviours of melanoma (2426). Luciferase reporter assays were used to verify a direct interaction between miR-675 and the 3′-UTR of MTDH. miR-675 mimics or miR-NC was transfected into A375 and HT144 cells, together with pGL3-MTDH-3′-UTR Wt or pGL3-MTDH-3′-UTR Mut. As shown in Fig. 3B, miR-675 overexpression decreased the luciferase activities of the MTDH with a wild-type 3′-UTR vector (P<0.05) but not the mutant vector, implying that miR-675 directly binds the 3′-UTR of MTDH. To further confirm whether miR-675 has regulation effect on MTDH endogenous in melanoma, we detected MTDH mRNA and protein expression in A375 and HT144 cells after transfection with miR-675 mimics or miR-NC. The results revealed that MTDH mRNA (Fig. 3C; P<0.05) and protein (Fig. 3D; P<0.05) expressions were significantly suppressed by miR-675 overexpression in A375 and HT144 cells. Overall, these results suggested that MTDH is a direct target gene of miR-675 in melanoma.

MTDH is upregulated in melanoma tissues and negatively correlated with miR-675 expression

To further explore the relationship between miR-675 and MTDH, we measured MTDH mRNA expression in melanoma tissues and corresponding adjacent normal tissues. The data from RT-qPCR analysis revealed that that levels of MTDH mRNA were significantly upregulated in melanoma tissues compared with those in adjacent normal tissues (Fig. 4A; P<0.05). In addition, Western blotting analysis indicated that MTDH protein was increased in melanoma tissues than adjacent normal tissues (Fig. 4B). Furthermore, Spearman's correlation analysis demonstrated an inverse association between MTDH mRNA and miR-675 expression levels in melanoma tissues (Fig. 4C; r=−0.7251; P=0.0002). These results suggested that increased levels of MTDH in melanoma, at least in part, may be attributed to a downregulation in miR-675.

Restoration of MTDH expression reverses the antitumor effects of miR-675 in melanoma

To further determine whether MTDH is a functional target of miR-675 in melanoma, we performed a rescue experiment involving the transfection of MTDH overexpression plasmids into miR-675-expressing A375 and HT144 cells. As shown in Fig. 5A, the decreased expression of MTDH protein was markedly restored by the transfection of pcDNA3.1-MTDH in A375 and HT144 cells (P<0.05). Functional experiments demonstrated that the upregulation of MTDH could effectively reverse the tumour-suppressing effects on cell proliferation (Fig. 5B; P<0.05) and invasion (Fig. 5C; P<0.05) induced by miR-675 overexpression in A375 and HT144 cells. These results suggested that miR-675 exerted its suppressive effects in melanoma, at least partially, by repressing MTDH.

Discussion

The dysregulation of miRNAs is involved in the development and progression of melanoma (2729). Therefore, miRNAs may be investigated as novel diagnostic or prognostic biomarkers and promising therapeutic targets in the treatment of patients with melanoma. In the present study, the expression of miR-675 was significantly downregulated in melanoma tissues and cell lines. In addition, we illustrated that miR-675 overexpression inhibited cell proliferation and invasion of melanoma in vitro. Importantly, this is the first study to identify MTDH as a direct target gene of miR-675 in melanoma. MTDH is highly expressed in melanoma tissues and negatively correlated with miR-675 expression level. The upregulation of MTDH rescued the antitumor effects of miR-675 in melanoma. Thus, miR-675 may play tumour-suppressing roles in melanoma by targeting MTDH in pathophysiologic process of melanoma.

An increasing number of studies reported that miR-675 is upregulated and plays an oncogenic role in the development and progression of multiple kinds of human cancer. For example, miR-675 was upregulated in breast cancer tissues and correlated with tumour grade (20). miR-675 overexpression increased the aggressive phenotype of breast cancer cells including promoted cell proliferation and migration in vitro and increased tumour growth and metastasis in vivo (21). miR-675 was also identified as an oncogene in bladder cancer (22), head and neck squamous cell carcinoma (30) and hepatocellular carcinoma (31,32). However, miR-675 was found to be lowly expressed in non-small cell lung cancer. Low miR-675 expression was correlated with lymph node metastasis and TNM stage (33). Resumption expression of miR-675 suppressed non-small cell lung cancer growth, colony formation and metastasis in vitro and decreased the tumorigenicity graft growth of nude mice in vivo (33). The downregulation of miR-675 was also observed in pancreatic cancer (34), adrenocortical adenomas (35), prostate cancer (36) and glioma (37). These findings suggested that miR-675 expression and roles exhibits tissue specificity and may be a prognostic marker and promising molecular therapeutic target in these specific types of cancer.

The identification of the miRNA target genes is important for understanding its roles in carcinogenesis and progression (38). Multiple targets of miR-675 have been identified, including c-Cbl(21), Cbl-b(21) in breast cancer, ZEB1(34) in pancreatic cancer, GPR55(33) in non-small cell lung cancer, TGFBI(36) in prostate cancer, CDK6(37) in glioma, p53(22) in bladder cancer, AKT(32) and Cdc25A(31) in hepatocellular carcinoma. In the current study, MTDH, also known as astrocyte elevated gene-1, was identified as a direct target of miR-675 in melanoma. It was first discovered in human foetal astrocytes in 2002 (39) and was found to be overexpressed in several types of human cancer, such as colorectal cancer (40), breast cancer (41), cervical cancer (42), gastric cancer (43) and bladder cancer (44). Increasing evidence indicated that MTDH is participated in the regulation of tumorigenesis and tumour development by regulating cell proliferation, cell cycle, apoptosis, migration, invasion, metastasis, epithelial-to-mesenchymal transition and angiogenesis (4547). In melanoma, MTDH expression level increased in tumour tissues and is significantly associated with metastatic rate (24,48). Functional study indicated that MTDH underexpression inhibited the proliferation, migration and invasion and induced cell cycle arrest and apoptosis in melanoma. Additionally, in vivo experiments revealed that the downregulation of MTDH reduced the growth of melanoma xenografts in nude mice (26). Combined with the present findings, miR-675/MTDH axis may present a potentially effective therapeutic strategy for the treatments of patients with melanoma.

In conclusion, the results of the present study provide evidence suggesting that miR-675 may play a tumour-suppressing role in melanoma, partly by targeting MTDH. The miR-675/MTDH pathway may provide novel insights into the pathogenesis of melanoma and could be investigated as a novel potential therapeutic target for the treatment of this disease. Future work is needed to explore whether the potential of miR-675 may be fully realised in melanoma.

References

1 

Miller AJ and Mihm MC Jr: Melanoma. N Engl J Med. 355:51–65. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Eggermont AM, Suciu S, Rutkowski P, Kruit WH, Punt CJ, Dummer R, Salès F, Keilholz U, de Schaetzen G and Testori A; EORTC Melanoma Group, : Long term follow up of the EORTC 18952 trial of adjuvant therapy in resected stage IIB-III cutaneous melanoma patients comparing intermediate doses of interferon-alpha-2b (IFN) with observation: Ulceration of primary is key determinant for IFN-sensitivity. Eur J Cancer. 55:111–121. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Pacheco I, Buzea C and Tron V: Towards new therapeutic approaches for malignant melanoma. Expert Rev Mol Med. 13:e332011. View Article : Google Scholar : PubMed/NCBI

7 

Fang W, Fan Y, Fa Z, Xu J, Yu H, Li P and Gu J: microRNA-625 inhibits tumorigenicity by suppressing proliferation, migration and invasion in malignant melanoma. Oncotarget. 8:13253–13263. 2017.PubMed/NCBI

8 

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

9 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Chen K and Rajewsky N: The evolution of gene regulation by transcription factors and microRNAs. Nat Rev Genet. 8:93–103. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Adams BD, Parsons C, Walker L, Zhang WC and Slack FJ: Targeting noncoding RNAs in disease. J Clin Invest. 127:761–771. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Li S, Xu X, Xu X, Hu Z, Wu J, Zhu Y, Chen H, Mao Y, Lin Y, Luo J, et al: MicroRNA-490-5p inhibits proliferation of bladder cancer by targeting c-Fos. Biochem Biophys Res Commun. 441:976–981. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Zhang J, Wu W, Xu S, Zhang J, Zhang J, Yu Q, Jiao Y, Wang Y, Lu A, You Y, et al: MicroRNA-105 inhibits human glioma cell malignancy by directly targeting SUZ12. Tumour Biol. 39:10104283177057662017.PubMed/NCBI

16 

Asahchop EL, Akinwumi SM, Branton WG, Fujiwara E, Gill MJ and Power C: Plasma microRNA profiling predicts HIV-associated neurocognitive disorder. AIDS. 30:2021–2031. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Wang W, Zhang H, Tang M, Liu L, Zhou Z, Zhang S and Wang L: MicroRNA-592 targets IGF-1R to suppress cellular proliferation, migration and invasion in hepatocellular carcinoma. Oncol Lett. 13:3522–3528. 2017.PubMed/NCBI

18 

Zhang P, Kong F, Deng X, Yu Y, Hou C, Liang T and Zhu L: MicroRNA-326 suppresses the proliferation, migration and invasion of cervical cancer cells by targeting ELK1. Oncol Lett. 13:2949–2956. 2017.PubMed/NCBI

19 

Liang Z, Wang X, Xu X, Xie B, Ji A, Meng S, Li S, Zhu Y, Wu J, Hu Z, et al: MicroRNA-608 inhibits proliferation of bladder cancer via AKT/FOXO3a signaling pathway. Mol Cancer. 16:962017. View Article : Google Scholar : PubMed/NCBI

20 

Zhai LL, Wang P, Zhou LY, Yin JY, Tang Q, Zhang TJ, Wang YX, Yang DQ, Lin J and Deng ZQ: Over-expression of miR-675 in formalin-fixed paraffin-embedded (FFPE) tissues of breast cancer patients. Int J Clin Exp Med. 8:11195–11201. 2015.PubMed/NCBI

21 

Vennin C, Spruyt N, Dahmani F, Julien S, Bertucci F, Finetti P, Chassat T, Bourette RP, Le Bourhis X and Adriaenssens E: H19 non coding RNA-derived miR-675 enhances tumorigenesis and metastasis of breast cancer cells by downregulating c-Cbl and Cbl-b. Oncotarget. 6:29209–29223. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Liu C, Chen Z, Fang J, Xu A, Zhang W and Wang Z: H19-derived miR-675contributes to bladder cancer cell proliferation by regulating p53 activation. Tumour Biol. 37:263–270. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Kang DC, Su ZZ, Sarkar D, Emdad L, Volsky DJ and Fisher PB: Cloning and characterization of HIV-1-inducible astrocyte elevated gene-1, AEG-1. Gene. 353:8–15. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Huang Y and Li LP: Progress of cancer research on astrocyte elevated gene-1/Metadherin (Review). Oncol Lett. 8:493–501. 2014.PubMed/NCBI

26 

Zhang Y, Peng G, Wang Y, Cui L, Wu W, Wang L, Liu C and Han X: Silencing of astrocyte elevated gene-1 inhibits proliferation and migration of melanoma cells and induces apoptosis. Clin Exp Pharmacol Physiol. 44:815–826. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Bustos MA, Ono S, Marzese DM, Oyama T, Iida Y, Cheung G, Nelson N, Hsu SC, Yu Q and Hoon DSB: miR-200a regulates CDK4/6 Inhibitor effect by targeting CDK6 in metastatic melanoma. J Invest Dermatol. 137:1955–1964. 2017. View Article : Google Scholar : PubMed/NCBI

28 

He J, Tian N, Yang Y, Jin L, Feng X, Hua J, Lin S, Wang B, Li H and Wang J: miR-185 enhances the inhibition of proliferation and migration induced by ionizing radiation in melanoma. Oncol Lett. 13:2442–2448. 2017.PubMed/NCBI

29 

Zeng HF, Yan S and Wu SF: MicroRNA-153-3p suppress cell proliferation and invasion by targeting SNAI1 in melanoma. Biochem Biophys Res Commun. 487:140–145. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Guan GF, Zhang DJ, Wen LJ, Xin D, Liu Y, Yu DJ, Su K, Zhu L, Guo YY and Wang K: Overexpression of lncRNA H19/miR-675 promotes tumorigenesis in head and neck squamous cell carcinoma. Int J Med Sci. 13:914–922. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Yu YQ, Weng J, Li SQ, Li B and Lv J: miR-675 promotes the growth of hepatocellular carcinoma cells through the Cdc25A pathway. Asian Pac J Cancer Prev. 17:3881–3885. 2016.PubMed/NCBI

32 

Lv J, Ma L, Chen XL, Huang XH and Wang Q: Downregulation of LncRNAH19 and miR-675 promotes migration and invasion of human hepatocellular carcinoma cells through AKT/GSK-3β/Cdc25A signaling pathway. J Huazhong Univ Sci Technolog Med Sci. 34:363–369. 2014. View Article : Google Scholar : PubMed/NCBI

33 

He D, Wang J, Zhang C, Shan B, Deng X, Li B, Zhou Y, Chen W, Hong J, Gao Y, et al: Down-regulation of miR-675-5p contributes to tumor progression and development by targeting pro-tumorigenic GPR55 in non-small cell lung cancer. Mol Cancer. 14:732015. View Article : Google Scholar : PubMed/NCBI

34 

Wang J, Zhang Y, Wei H, Zhang X, Wu Y, Gong A, Xia Y, Wang W and Xu M: The mir-675-5p regulates the progression and development of pancreatic cancer via the UBQLN1-ZEB1-mir200 axis. Oncotarget. 8:24978–24987. 2017.PubMed/NCBI

35 

Schmitz KJ, Helwig J, Bertram S, Sheu SY, Suttorp AC, Seggewiss J, Willscher E, Walz MK, Worm K and Schmid KW: Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in benign and malignant adrenocortical tumours. J Clin Pathol. 64:529–535. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Zhu M, Chen Q, Liu X, Sun Q, Zhao X, Deng R, Wang Y, Huang J, Xu M, Yan J and Yu J: lncRNA H19/miR-675 axis represses prostate cancer metastasis by targeting TGFBI. FEBS J. 281:3766–3775. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Li C, Lei B, Huang S, Zheng M, Liu Z, Li Z and Deng Y: H19 derived microRNA-675 regulates cell proliferation and migration through CDK6 in glioma. Am J Transl Res. 7:1747–1764. 2015.PubMed/NCBI

38 

Yao J, Zhang P, Li J and Xu W: MicroRNA-215 acts as a tumor suppressor in breast cancer by targeting AKT serine/threonine kinase 1. Oncol Lett. 14:1097–1104. 2017.PubMed/NCBI

39 

Su ZZ, Kang DC, Chen Y, Pekarskaya O, Chao W, Volsky DJ and Fisher PB: Identification and cloning of human astrocyte genes displaying elevated expression after infection with HIV-1 or exposure to HIV-1 envelope glycoprotein by rapid subtraction hybridization, RaSH. Oncogene. 21:3592–3602. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Gnosa S, Shen YM, Wang CJ, Zhang H, Stratmann J, Arbman G and Sun XF: Expression of AEG-1 mRNA and protein in colorectal cancer patients and colon cancer cell lines. J Transl Med. 10:1092012. View Article : Google Scholar : PubMed/NCBI

41 

Li J, Zhang N, Song LB, Liao WT, Jiang LL, Gong LY, Wu J, Yuan J, Zhang HZ, Zeng MS and Li M: Astrocyte elevated gene-1 is a novel prognostic marker for breast cancer progression and overall patient survival. Clin Cancer Res. 14:3319–3326. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Yu JQ, Zhou Q, Zhu H, Zheng FY and Chen ZW: Overexpression of astrocyte elevated gene-1 (AEG-1) in cervical cancer and its correlation with angiogenesis. Asian Pac J Cancer Prev. 16:2277–2281. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Dong L, Qin S, Li Y, Zhao L, Dong S, Wang Y, Zhang C and Han S: High expression of astrocyte elevated gene-1 is associated with clinical staging, metastasis and unfavorable prognosis in gastric carcinoma. Tumour Biol. 36:2169–2178. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Yang G, Zhang L, Lin S, Li L, Liu M, Chen H, Cao M, Liu D, Huang YR and Bo J: AEG-1 is associated with tumor progression in nonmuscle-invasive bladder cancer. Med Oncol. 31:9862014. View Article : Google Scholar : PubMed/NCBI

45 

Hu G, Wei Y and Kang Y: The multifaceted role of MTDH/AEG-1 in cancer progression. Clin Cancer Res. 15:5615–5620. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Emdad L, Lee SG, Su ZZ, Jeon HY, Boukerche H, Sarkar D and Fisher PB: Astrocyte elevated gene-1 (AEG-1) functions as an oncogene and regulates angiogenesis. Proc Natl Acad Sci USA. 106:pp. 21300–21305. 2009; View Article : Google Scholar : PubMed/NCBI

47 

Du Y, Jiang B, Song S, Pei G, Ni X, Wu J, Wang S, Wang Z and Yu J: Metadherin regulates actin cytoskeletal remodeling and enhances human gastric cancer metastasis via epithelial-mesenchymal transition. Int J Oncol. 51:63–74. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Zou B and Wang S: Expression and prognostic value of MTDH, Bcl-2 and E-cadherin in sinonasal malignant mucosal melanoma. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 50:1009–1014. 2015.(In Chinese). PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu K, Jin J, Rong K, Zhuo L and Li P: MicroRNA‑675 inhibits cell proliferation and invasion in melanoma by directly targeting metadherin Retraction in /10.3892/mmr.2023.12986. Mol Med Rep 17: 3372-3379, 2018
APA
Liu, K., Jin, J., Rong, K., Zhuo, L., & Li, P. (2018). MicroRNA‑675 inhibits cell proliferation and invasion in melanoma by directly targeting metadherin Retraction in /10.3892/mmr.2023.12986. Molecular Medicine Reports, 17, 3372-3379. https://doi.org/10.3892/mmr.2017.8264
MLA
Liu, K., Jin, J., Rong, K., Zhuo, L., Li, P."MicroRNA‑675 inhibits cell proliferation and invasion in melanoma by directly targeting metadherin Retraction in /10.3892/mmr.2023.12986". Molecular Medicine Reports 17.2 (2018): 3372-3379.
Chicago
Liu, K., Jin, J., Rong, K., Zhuo, L., Li, P."MicroRNA‑675 inhibits cell proliferation and invasion in melanoma by directly targeting metadherin Retraction in /10.3892/mmr.2023.12986". Molecular Medicine Reports 17, no. 2 (2018): 3372-3379. https://doi.org/10.3892/mmr.2017.8264