Open Access

MicroRNA-9 inhibits the proliferation and migration of malignant melanoma cells via targeting sirituin 1

Retraction in: /10.3892/etm.2021.10569

  • Authors:
    • Pingyuan Bu
    • Chengqun Luo
    • Quanyong He
    • Ping Yang
    • Xi Li
    • Dan Xu
  • View Affiliations

  • Published online on: June 13, 2017     https://doi.org/10.3892/etm.2017.4595
  • Pages: 931-938
  • Copyright: © Bu 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

MicroRNA (miR) are a class of small non‑coding RNA that are able to inhibit gene expression by directly binding to the 3' untranslated region (UTR) of their target mRNA and thus promote translational repression or mRNA degradation. Recently, miR‑9 was reported to have a suppressive role in malignant melanoma; however, the underlying mechanism remains largely unclear. In the present study, reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blotting were used to examine the mRNA and protein expression levels in malignant melanoma tissues and cell lines. The MTT assay and wound healing assay were used to examine the cell viability, proliferation and migratory capacities. Bioinformatics prediction and luciferase reporter assay were performed to investigate the relationship between miR‑9 and its potential target gene. The present data revealed that miR‑9 expression was significantly downregulated in malignant melanoma tissues when compared with their matched adjacent non‑tumor tissues. Furthermore, the expression levels of miR‑9 were reduced in malignant melanoma cell lines when compared with human normal skin HACAT cells. Moreover, the ectopic expression of miR‑9 significantly suppressed the proliferation and migration of malignant melanoma cells, accompanied with a remarkable decrease in the protein expression levels of sirtuin 1 (SIRT1), which were markedly upregulated in malignant melanoma tissues and cell lines. Additionally, restoration of SIRT1 reversed the suppressive effects of miR‑9 on the proliferation and migration of malignant melanoma cells. Luciferase reporter assay data further identified SIRT1 as a direct target gene of miR‑9. To conclude, the present findings indicate that miR‑9 has a suppressive role in malignant melanoma cell viability and migration, at least in part, via directly inhibiting the protein expression of its target gene, SIRT1. Therefore, miR‑9 may serve as a potential candidate for the treatment of malignant melanoma.

Introduction

Malignant melanoma is a malignant tumor of melanocytes, characterized by rapid progression and distant metastasis (1). It ranks as the seventh most common malignancy for women and the fifth most common cancer for men (2,3), and its incidence has been increasing annually by 3.1% over the past decade (3). Therefore, investigations into the molecular mechanism of malignant melanoma are urgently required for the development of effective therapeutic strategies for malignant melanoma (4).

MicroRNA (miR), which are a class of small non-coding RNA, are able to negatively regulate gene expression via directly binding to the 3′ untranslated region (UTR) of their target mRNA, thus resulting in mRNA degradation or translational repression (5). They participate in various cellular biological processes, such as cell survival, proliferation, apoptosis, differentiation, cell cycle progression and migration, predominantly by negatively regulating the protein expression of their target genes (6,7). Recently, miR-9 was reported to have a suppressive role in malignant melanoma (8). For instance, Liu et al (9) identified that miR-9 suppressed the migration and invasion of uveal malignant melanoma cells through the NF-κB1 pathway. Zhao et al (10) reported that Yin Yang 1 promoted the proliferation, cell cycle progression, migration and invasion of malignant melanoma cells, likely via negatively regulating miR-9 expression. However, the molecular mechanism of miR-9 in regulating the proliferation and migration of malignant melanoma cells still remains to be fully elucidated.

Sirtuins (SIRTs) are nicotinamide adenine dinucleotide (NAD+)-dependent class III histone deacetylases (11). SIRT1, a member of the SIRT family, is characterized by a 275-aminoacid catalytic core and the distinctive additional N-terminal and/or C-terminal sequences of variable length (12). Via interactions with acetylating histones and multiple transcription factors, SIRT1 participates in various physiological processes, such as metabolism, senescence, neuroprotection, inflammation and tumorigenesis (1315). Recently, the oncogenic role of SIRT1 in malignant melanoma has gradually been revealed (16). For instance, Wilking et al (17) reported that SIRT1 was upregulated in malignant melanoma and inhibition of SIRT1 by small molecules exhibited an anti-proliferative response via the activation of the tumor suppressor, p53. Ohanna et al (18) indicated that SIRT1 promoted proliferation and inhibited the senescence-like phenotype in malignant melanoma cells. However, the detailed regulatory mechanism of SIRT1 expression in malignant melanoma remains largely unclear.

In the present study, miR-9 expression levels in malignant melanoma and the molecular mechanism of miR-9 in regulating the proliferation and migration of malignant melanoma cells involving SIRT1 was investigated.

Materials and methods

Tissue samples

The present study was approved by the Ethic Committee of Third Xiangya Hospital of Central South University (Changsha, China). Primary malignant melanoma (n=24) and matched adjacent non-tumor tissues were collected from malignant melanoma patients who underwent surgical resections between May 2010 and April 2014 at Third Xiangya Hospital (Changsha, China). The patients included 11 males and 13 females, aged 47–77 years (mean, 63.5±11.3 years), who were diagnosed using histopathological analysis. All malignant melanoma patients had received no radiation therapy or chemotherapy prior to the surgery. Tissues were snap-frozen in liquid nitrogen and stored at −70°C prior to use.

Cell culture

Human malignant melanoma cell lines (B16, A375, G361, and HME1), human normal skin HACAT cell line and HEK293 cell line were obtained from the Cell Bank of Central South University (Changsha, China). Cells were cultured in RPMI-1640 medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Inc.) maintained at 37°C in a humidified atmosphere containing 5% CO2.

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

Total RNA was extracted from tissues and cells using TRIzol Reagent (Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. DNase (Takara Biotechnology Co., Ltd., Dalian, China) treatment was used to remove genomic DNA, according to the manufacturer's instructions. For reverse transcription, 5 µl total RNA was mixed with 0.15 µl of 100 mm dNTPs (with dTTP), 1 µl (50 U) MultiScribe reverse transcriptase, 1.5 µl of 10X reverse transcription buffer, 0.19 µl RNase inhibitor (20 U/µl), and 3 µl 1X gene specific primers. Nuclease-free H2O was added to obtain a final volume of 15 µl. Reverse transcription was performed at 16°C for 30 min, followed by an incubation step at 42°C for 30 min and enzyme inactivation at 85°C for 5 min. The resulting cDNA was stored at −20°C until use. For mRNA expression detection, a SYBR-Green RT-PCR kit (Takara Biotechnology Co., Ltd.) was used to perform the RT-qPCR on 7300 Plus thermal cycler (Thermo Fisher Scientific, Inc.), in accordance with the manufacturer's instructions. GAPDH was used as an internal reference. For miRNA analysis, a PrimeScript miRNA RT-PCR kit (Takara Biotechnology Co., Ltd., Dalian, China) was used according to the manufacturer's instructions. U6 was used as an internal reference. For the PCR assay, 10 µl of 1X PCR master mix, 0.33 µl cDNA solution, 2 µl of 1X gene specific primer, and 7.67 µl H2O were mixed to obtain a final reaction volume of 20 µl. For both mRNA and miRNA detection, the reaction conditions were 95°C for 10 min, and 45 cycles of denaturation at 95°C for 15 sec and annealing/elongation at 60°C for 15 sec. The relative expression was analyzed by the 2−ΔΔCq method (19). The specific primers were designed as follows: SIRT1, forward 5′-TGTGTCATAGGTTAGGTGGTGA-3′ and reverse 5′-AGCCAATTCTTTTTGTGTTCGTG-3′; and GAPDH, forward 5′-ACAACTTTGGTATCGTGGAAGG-3′ and reverse 5′-GCCATCACGCCACAGTTTC-3′.

Cell transfection

For ectopic expression of miR-9, a miR-9 mimic (Genepharma, Shanghai, China) was used to transfect malignant melanoma cell lines via Lipofectamine 2000 (Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Scramble miR mimic (Genepharm, Inc., Sunnyvale, CA, USA) was used as a negative control (NC). For SIRT1 reversal experiments, miR-9 mimic and pcDNA3.1-SIRT1 [outside reading frame (ORF)] plasmid (Amspring, Changsha, China) were used to co-transfect B16 cells using Lipofectamine 2000, according to the manufacturer's instructions. Following transfection for 48 h, the expression levels of miR-9 or SIRT1 were evaluated.

Cell proliferation detection

B16 cells (10,000 cells/well) were plated into a 96-well plate, and cultured at 37°C in an atmosphere containing 5% CO2 for 0, 12, 24, 48, and 72 h. Subsequently, 10 µl of MTT (5 mg/ml; Thermo Fisher Scientific, Inc.) was added. Following incubation at 37°C for 4 h, 100 µl of dimethyl sulfoxide was added and samples were incubated at room temperature for 20 min. Subsequently, formazan production was detected by determining the optical density at 570 nm using an enzyme immunoassay analyzer.

Wound healing assay

A wound healing assay was performed to evaluate the cell migratory capacity and proliferation of B16 malignant melanoma cells. B16 cells were cultured to full confluence and a wound of ~1 mm width was created with a plastic scriber. Subsequently, cells were washed in Dulbecco's phosphate-buffered saline (Thermo Fisher Scientific, Inc.) and incubated in serum-free RPMI-1640 at 37°C for 24 h. Subsequently, cells were incubated in RPMI-1640 supplemented with 10% FBS. Once cultured for 48 h, cells were fixed by 90% ethanol for 20 min at room temperature and observed under a light microscope (CX22; Olympus Corporation, Tokyo, Japan).

Western blotting

Tissues and cells were lysed in cold radioimmunoprecipitation assay buffer (Thermo Fisher Scientific, Inc.). The concentration of protein was determined using a bicinchoninic acid (BCA) Protein Assay kit (Pierce; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Protein was separated using 12% SDS-PAGE, transferred to a polyvinylidene fluoride membrane (PVDF; Thermo Fisher Scientific, Inc.), and blocked in 5% non-fat dried milk in phosphate-buffered saline (Thermo Fisher Scientific, Inc.) at 4°C overnight. The PVDF membrane was subsequently incubated with rabbit anti-human SIRT1 monoclonal antibody (1:200; ab32441; Abcam, Cambridge, MA, USA), or rabbit anti-human GAPDH monoclonal antibody (1:100; ab9485, Abcam) for 3 h at room temperature. The membrane was washed three times using Dulbecco's phosphate-buffered saline, incubated with goat anti-rabbit IgG (1:20,000; ab6721; Abcam) for 40 min at room temperature and washed three times with Dulbecco's phosphate-buffered saline. An enhanced chemiluminescence (ECL) Western Blotting kit (Pierce; Thermo Fisher Scientific, Inc.) was used to detect the immune complex on the PVDF membrane. Protein expression was analyzed using Image-Pro Plus software 6.0 (Media Cybernetics, Inc., Rockville, MD, USA). GAPDH was used as an internal control.

Bioinformatics analysis and luciferase reporter assay

TargetScan software (targetsan.org) was used to analyze whether SIRT1 was a potential target of miR-9. The wild type (WT) of SIRT1 3′UTR containing the putative binding sites of miR-9 was amplified and subcloned into the psiCHECK-2 vector (Promega Corp., Madison, WI, USA), downstream to the luciferase gene sequence. The mutant type (MT) of SIRT1 3′UTR was generated using the Quick-Change Site-Directed Mutagenesis kit (Agilent Technologies, Inc., Santa Clara, CA, USA), in accordance with the manufacturer's protocol and was subcloned into the psiCHECK-2 vector. In the control group, B16 cells were transfected with 100 ng of WT-SIRT1-3′UTR vector or MT-SIRT1-3′UTR vector using Lipofectamine 2000, according to the manufacturer's instructions. In the NC group, cells were co-transfected with 50 nM of scramble miR mimic and 100 ng of WT-SIRT1-3′UTR vector or MT-SIRT1-3′UTR vector, respectively. In the experimental group, cells were co-transfected with 50 nM of miR-9 mimic and 100 ng of SIRT1 3′UTR WT vector or SIRT1 3′UTR MT vector, respectively. Following transfection for 48 h, the activities of Renilla luciferase and Firefly luciferase were examined using the luciferase reporter assay system (Promega Corp.), according to the manufacturer's instructions. The Renilla luciferase activity was normalized to Firefly luciferase activity.

Statistical analysis

Data were expressed as mean ± standard deviation. SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA) was used for statistical analysis. Data were analyzed by a Student's t-test for two-group comparison and one-way analysis of variance for multiple-group comparison. Tukey's post hoc test was also used. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-9 is downregulated in malignant melanoma

To reveal the role of miR-9 in malignant melanoma, RT-qPCR was used to detect miR-9 expression levels in malignant melanoma tissues and their matched adjacent non-tumor tissues. As shown in Fig. 1A, the miR-9 expression level was lower in 83.3% (20/24) of malignant melanoma tissues when compared with their matched adjacent non-tumor tissues. miR-9 expression levels were significantly downregulated in malignant melanoma tissues when compared with that of adjacent non-tumor tissues (P=0.0019; Fig. 1B). In addition, the expression levels of miR-9 in human malignant melanoma cell lines, including B16, A375, G361, and HME1 and human normal skin HACAT cells were determined. The data suggested that miR-9 expression levels were significantly decreased in malignant melanoma cell lines when compared with HACAT cells (P<0.01; Fig. 1C). Accordingly, miR-9 was indicated to be downregulated in malignant melanoma.

Ectopic expression of miR-9 suppressed the proliferation and migration of malignant melanoma cells

As miR-9 was downregulated in malignant melanoma cell lines, B16 cells were transfected with miR-9 mimic to restore its expression. Scramble miR mimic was used as the NC. RT-qPCR data indicated that the miR-9 expression level was significantly increased in B16 cells transfected with miR-9 mimic, when compared with that in the control group (P<0.01; Fig. 2A). However, no significant difference was observed in the miR-9 expression levels in the NC group when compared with that in the control group (Fig. 2A). MTT and wound healing assays were used to examine cell viability, proliferation and migratory capacities in each group. Ectopic expression of miR-9 resulted in a significant decrease in viability, proliferation and migration of B16 cells when compared with the control group (P<0.01; Fig. 2B and C, respectively). However, no significant difference in the viability and migration of B16 cells in the NC group were observed when compared with those in the control group (Fig. 2B and C, respectively). These results suggest that miR-9 may have a suppressive role in regulating the viability, proliferation and migration of malignant melanoma B16 cells.

Overexpression of miR-9 decreased the expression of SIRT1 in malignant melanoma cells

Recently, SIRT1 has been demonstrated to promote the proliferation and migration of malignant melanoma cells (17,18). Therefore, the expression of SIRT1 in B16 cells in each group was examined. As indicated in Fig. 3A, RT-qPCR data indicated no significant difference in SIRT1 mRNA expression levels between the miR-9 overexpression group and the control group. However, western blot data indicated that overexpression of miR-9 resulted in a significant decrease in the protein expression levels of SIRT1 in B16 cells when compared with the control group (P<0.01; Fig. 3B). Therefore, the present findings suggest that miR-9 may negatively regulate SIRT1 expression at post-transcriptional level.

Furthermore, the present study indicated that the mRNA expression levels of SIRT1 were significantly increased in malignant melanoma tissues when compared with their matched adjacent non-tumor tissues (P<0.0001; Fig. 3C). In addition, SIRT1 protein expression levels were significantly upregulated in malignant melanoma cell lines when compared with the normal skin HACAT cells (P<0.01; Fig. 3D). Moreover, a reversed correlation between the miR-9 and SIRT1 expression levels in malignant melanoma tissues was observed (Fig. 3E). The present findings suggest that the increased mRNA and protein expression levels of SIRT1 may be due to the downregulation of miR-9 in malignant melanoma tissues and cell lines.

Overexpression of SIRT1 reversed the miR-9-mediated proliferation and migration of malignant melanoma cells

As overexpression of miR-9 was able to decrease the protein expression levels of SIRT1, which has an oncogenic role in malignant melanoma, subsequently, whether SIRT1 was involved in the miR-9-mediated viability, proliferation and migration of malignant melanoma cells was investigated. B16 cells were transfected with miR-9 mimic, or co-transfected with miR-9 mimic and SIRT1 ORF plasmid, respectively. Following transfection, western blotting was performed to examine the protein expression levels of SIRT1 in each group. The data revealed that SIRT1 protein expression levels were significantly increased in B16 cells co-transfected with miR-9 mimic and SIRT1 ORF plasmid when compared with B16 cells transfected with miR-9 group (P<0.01; Fig. 4A). This suggested that transfection with SIRT1 ORF plasmid reversed the suppressive effect of miR-9 on the protein expression levels of SIRT1 in B16 cells.

MTT assay and wound healing assay were subsequently conducted to examine the cell viability, proliferation and migration of B16 cells. The viability, proliferation and migration of B16 cells in the miR-9 + SIRT1 group were significantly increased when compared with the miR-9 group (P<0.01; Fig. 4B and C). The present findings suggested that overexpression of SIRT1 reversed the suppressive effects of miR-9 on B16 cell viability, proliferation and migration.

SIRT1 is a direct target of miR-9 in B16 cells

TargetScan was used to analyze whether SIRT1 was a potential target of miR-9. As indicated in Fig. 5A and B, SIRT1 was predicted to be an evolutionarily conserved direct target of miR-9. To further confirm the targeting relationship between miR-9 and SIRT1, the WT-SIRT1-3′UTR luciferase reporter vector and the MT-SIRT1-3′UTR luciferase reporter vector were generated (Fig. 5C and D). HEK293 cells were further co-transfected with WT-SIRT1-3′UTR vector or MT-SIRT1-3′UTR vector, with miR-9 mimic or miR-NC, respectively. Subsequently, the luciferase reporter assay was performed. As indicated in Fig. 5E, the luciferase activity was significantly decreased in HEK293 cells co-transfected with miR-9 mimic and WT-SIRT1-3′UTR vector when compared to the control cells that were transfected with WT-SIRT1-3′UTR vector alone (P<0.01). However, the downregulation of luciferase activity was abolished when co-transfection with miR-9 mimic and MT-SIRT1-3′UTR vector (Fig. 5E), indicating that SIRT1 is a direct target gene of miR-9. To conclude, the present findings demonstrate that miR-9 may have a suppressive role in malignant melanoma cell viability, proliferation and migration, at least in part, via directly inhibiting the protein expression levels of its target gene, SIRT1.

Discussion

miR-9 has recently been demonstrated to have a suppressive role in malignant melanoma (9); however, the underlying mechanism remains to be fully elucidated. In the present study, miR-9 was indicated to be significantly downregulated in malignant melanoma tissues and cell lines when compared with matched adjacent non-tumor tissues or normal skin HACAT cells, respectively. Ectopic expression of miR-9 suppressed malignant melanoma cell viability, proliferation and migration, accompanied with the decreased protein expression levels of SIRT1, which was upregulated in malignant melanoma tissues and cell lines. Moreover, overexpression of SIRT1 reversed the suppressive effects of miR-9 on the viability, proliferation and migration of malignant melanoma cells. Furthermore, luciferase reporter assay data identified SIRT1 as a direct target gene of miR-9.

Deregulations of various miR have been implicated in malignant melanoma (2022). For instance, miR-365 was significantly downregulated in malignant melanoma tissues and cell lines and its expression levels were associated with lymph node metastasis, clinical stage, and survival of this disease (23). Moreover, miR-365 has been identified to inhibit the growth, invasion and metastasis of malignant melanoma by targeting NRP1 expression (23). Additionally, miR-193b is significantly downregulated in the melanoma tissues and was able inhibit melanoma cell proliferation via targeting Cyclin D1 (24). Recently, the miR-9 expression was reported to be significantly reduced in metastatic melanoma when compared with primary melanoma (8). In the present study, miR-9 was revealed to be downregulated in melanoma tissues when compared to their matched adjacent non-tumor tissues. Furthermore, miR-9 levels were decreased in melanoma cell lines when compared with normal skin HACAT cells. These findings suggest that miR-9 may have a role in melanoma. To further study the exact role of miR-9 in malignant melanoma, B16 cells were transfected with miR-9 mimic to upregulate its expression levels. The present data indicated that overexpression of miR-9 significantly decreased the viability and migration of B16 cells, suggesting that miR-9 has suppressive effects on melanoma growth and metastasis. These findings were consistent with several alternative studies (9,10). For instance, Zhao et al (10) reported that overexpression of miR-9 reduced the proliferation, cell cycle progression, migration and invasion of melanoma cells. Liu et al (9) showed that miR-9 was able to suppress the migration and invasion of highly invasive melanoma cells. These findings and the present study indicate that miR-9 acts a tumor suppressor in malignant melanoma.

As miR function predominantly through the inhibition of their target genes (25), we further focused on the potential targets of miR-9 in melanoma. SIRT1, an NAD+-dependent class III histone deacetylase, exhibits an oncogenic role in human cancers (14,15). For instance, Wu et al (26) demonstrated that SIRT1 participated in the tumorigenesis, metastasis, and chemoresistance of hepatocellular carcinoma. Qu et al (27) reported that SIRT1 promoted proliferation and inhibited apoptosis of glioma cells. Recently, SIRT1 was indicated to be involved in malignant melanoma. Knockdown of SIRT1 resulted in cell cycle arrest and a senescence-like phenotype of melanoma cells as well as inhibition of tumor growth, while overexpression of SIRT1 relieved the senescence-like phenotype and the proliferation arrest (18). However, the regulatory mechanism of SIRT1 expression in melanoma has not yet been fully studied. The present study identified that overexpression of miR-9 led to decreased protein expression levels of SIRT1 in melanoma cells; however, this did not affect the mRNA expression level of SIRT1. Further investigation showed that SIRT1 was upregulated in melanoma tissues and cell lines, which was reversely correlated with the miR-9 expression levels in melanoma tissues. These findings suggest that downregulation of miR-9 may contribute to the upregulation of SIRT1 in melanoma. As bioinformatics analysis predicted that SIRT1 was a direct target gene of miR-9, we used a luciferase reporter assay to confirm their relationship. Luciferase activity was indicated to be significantly decreased in HEK293 cells co-transfected with miR-9 mimic and WT-SIRT1-3′UTR vector; however, this downregulation was markedly abolished in cells co-transfected with miR-9 mimic and WT-SIRT1-3′UTR vector, suggesting that miR-9 can directly bind to the seed sequences in the SIRT1 3′UTR. Therefore, SIRT1 is indeed a target gene of miR-9. Accordingly, the present study reveals that the miR-9/SIRT1 axis is involved in malignant melanoma.

In conclusion, to the best of our knowledge, our study is the first to demonstrate a suppressive role of miR-9 in regulating malignant melanoma cell proliferation and migration via inhibition of SIRT1. Therefore, we suggest that miR-9 may serve as a potential candidate for the treatment of malignant melanoma.

References

1 

Rastrelli M, Tropea S, Rossi CR and Alaibac M: Melanoma: Epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo. 28:1005–1011. 2014.PubMed/NCBI

2 

Trotter SC, Sroa N, Winkelmann RR, Olencki T and Bechtel M: A global review of melanoma follow-up guidelines. J Clin Aesthet Dermatol. 6:18–26. 2013.PubMed/NCBI

3 

Linos E, Swetter SM, Cockburn MG, Colditz GA and Clarke CA: Increasing burden of melanoma in the United States. J Invest Dermatol. 129:1666–1674. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Populo H, Soares P and Lopes JM: Insights into melanoma: targeting the mTOR pathway for therapeutics. Expert Opin Ther Targets. 16:689–705. 2012. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

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

7 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2:e3632004. View Article : Google Scholar : PubMed/NCBI

8 

Liu S, Kumar SM, Lu H, Liu A, Yang R, Pushparajan A, Guo W and Xu X: MicroRNA-9 up-regulates E-cadherin through inhibition of NF-κB1-Snail1 pathway in melanoma. J Pathol. 226:61–72. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Liu N, Sun Q, Chen J, Li J, Zeng Y, Zhai S, Li P, Wang B and Wang X: MicroRNA-9 suppresses uveal melanoma cell migration and invasion through the NF-κB1 pathway. Oncol Rep. 28:961–968. 2012.PubMed/NCBI

10 

Zhao G, Li Q, Wang A and Jiao J: YY1 regulates melanoma tumorigenesis through a miR-9 ~ RYBP axis. J Exp Clin Cancer Res. 34:662015. View Article : Google Scholar : PubMed/NCBI

11 

Opitz CA and Heiland I: Dynamics of NAD-metabolism: Everything but constant. Biochem Soc Trans. 43:1127–1132. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Qiu G, Li X, Che X, Wei C, He S, Lu J, Jia Z, Pang K and Fan L: SIRT1 is a regulator of autophagy: Implications in gastric cancer progression and treatment. FEBS Lett. 589:2034–2042. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Zhang X, Chen S, Cheng M, Cao F and Cheng Y: The expression and correlation of SIRT1 and Phospho-SIRT1 in colorectal cancer. Int J Clin Exp Med. 8:809–817. 2015.PubMed/NCBI

14 

Lin L, Zheng X, Qiu C, Dongol S, Lv Q, Jiang J, Kong B and Wang C: SIRT1 promotes endometrial tumor growth by targeting SREBP1 and lipogenesis. Oncol Rep. 32:2831–2835. 2014.PubMed/NCBI

15 

Li L and Bhatia R: Role of SIRT1 in the growth and regulation of normal hematopoietic and leukemia stem cells. Curr Opin Hematol. 22:324–329. 2015.PubMed/NCBI

16 

Hu Z, Fan H, Lv G, Zhou Q, Yang B, Zheng J and Cao W: 5-Aminolevulinic acid-mediated sonodynamic therapy induces anti-tumor effects in malignant melanoma via p53-miR-34a-Sirt1 axis. J Dermatol Sci. 79:155–162. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Wilking MJ, Singh C, Nihal M, Zhong W and Ahmad N: SIRT1 deacetylase is overexpressed in human melanoma and its small molecule inhibition imparts anti-proliferative response via p53 activation. Arch Biochem Biophys. 563:94–100. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Ohanna M, Bonet C, Bille K, Allegra M, Davidson I, Bahadoran P, Lacour JP, Ballotti R and Bertolotto C: SIRT1 promotes proliferation and inhibits the senescence-like phenotype in human melanoma cells. Oncotarget. 5:2085–2095. 2014. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Kozubek J, Ma Z, Fleming E, Duggan T, Wu R, Shin DG and Dadras SS: In-depth characterization of microRNA transcriptome in melanoma. PLoS One. 8:e726992013. View Article : Google Scholar : PubMed/NCBI

21 

Xu D, Tan J, Zhou M, Jiang B, Xie H, Nie X, Xia K and Zhou J: Let-7b and microRNA-199a inhibit the proliferation of B16F10 melanoma cells. Oncol Lett. 4:941–946. 2012.PubMed/NCBI

22 

Strong AM Poenitzsch, Setaluri V and Spiegelman VS: microRNA-340 as a modulator of RAS-RAF-MAPK signaling in melanoma. Arch Biochem Biophys. 563:118–124. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Bai J, Zhang Z, Li X and Liu H: MicroRNA-365 inhibits growth, invasion and metastasis of malignant melanoma by targeting NRP1 expression. Int J Clin Exp Pathol. 8:4913–4922. 2015.PubMed/NCBI

24 

Chen J, Feilotter HE, Paré GC, Zhang X, Pemberton JG, Garady C, Lai D, Yang X and Tron VA: MicroRNA-193b represses cell proliferation and regulates cyclin D1 in melanoma. Am J Pathol. 176:2520–2529. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Esquela-Kerscher A and Slack FJ: Oncomirs-microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Wu Y, Meng X, Huang C and Li J: Emerging role of silent information regulator 1 (SIRT1) in hepatocellular carcinoma: A potential therapeutic target. Tumour Biol. 36:4063–4074. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Qu Y, Zhang J, Wu S, Li B, Liu S and Cheng J: SIRT1 promotes proliferation and inhibits apoptosis of human malignant glioma cell lines. Neurosci Lett. 525:168–172. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 14 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bu P, Luo C, He Q, Yang P, Li X and Xu D: MicroRNA-9 inhibits the proliferation and migration of malignant melanoma cells via targeting sirituin 1 Retraction in /10.3892/etm.2021.10569. Exp Ther Med 14: 931-938, 2017
APA
Bu, P., Luo, C., He, Q., Yang, P., Li, X., & Xu, D. (2017). MicroRNA-9 inhibits the proliferation and migration of malignant melanoma cells via targeting sirituin 1 Retraction in /10.3892/etm.2021.10569. Experimental and Therapeutic Medicine, 14, 931-938. https://doi.org/10.3892/etm.2017.4595
MLA
Bu, P., Luo, C., He, Q., Yang, P., Li, X., Xu, D."MicroRNA-9 inhibits the proliferation and migration of malignant melanoma cells via targeting sirituin 1 Retraction in /10.3892/etm.2021.10569". Experimental and Therapeutic Medicine 14.2 (2017): 931-938.
Chicago
Bu, P., Luo, C., He, Q., Yang, P., Li, X., Xu, D."MicroRNA-9 inhibits the proliferation and migration of malignant melanoma cells via targeting sirituin 1 Retraction in /10.3892/etm.2021.10569". Experimental and Therapeutic Medicine 14, no. 2 (2017): 931-938. https://doi.org/10.3892/etm.2017.4595