MicroRNA‑320 targets mitogen‑activated protein kinase 1 to inhibit cell proliferation and invasion in epithelial ovarian cancer

Retraction in: /10.3892/mmr.2024.13169

  • Authors:
    • Yongqian Xu
    • Jian Hu
    • Chunxia Zhang
    • Yuanyuan Liu
  • View Affiliations

  • Published online on: September 29, 2017     https://doi.org/10.3892/mmr.2017.7664
  • Pages: 8530-8536
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer is the second most frequently occurring cancer and the most fatal gynecological malignancy of all gynecological cancers worldwide. MicroRNAs (miR) have been reported to be downregulated or upregulated in a variety of human malignancies, and involved in the formation and progression of the majority of human cancers, including epithelial ovarian cancer (EOC). miR‑320 has been identified as a tumor suppressor in multiple human cancers. However, the expression levels, biological role and underlying mechanisms of miR‑320 in EOC remain to be elucidated. In the present study, reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was performed to detect miR‑320 expression in EOC tissues and cell lines. Following transfection with miR‑320 mimics, Cell Counting Kit 8 and cell invasion assays were utilized to investigate the effects of miR‑320 on EOC cell proliferation and invasion. Bioinformatic analysis, luciferase reporter assay, RT‑qPCR and western blotting were used to explore the underlying mechanism of how miR‑320 affects cell proliferation and invasion in EOC. Mitogen‑activated protein kinase (MAPK) 1 expression and its association with the miR‑320 expression level was examined in EOC tissues. The role of MAPK1 in EOC cells was additionally evaluated by using a loss‑of‑function assay. The results demonstrated that miR‑320 was markedly downregulated in EOC tissues and cell lines. A decreased miR‑320 expression was significantly correlated with the Federation of Gynecology and Obstetrics stage and lymph node metastasis of EOC patients. Additionally, reintroduction of miR‑320 expression suppressed cell proliferation and invasion in EOC. Furthermore, it was verified that MAPK1 is a direct target gene of miR‑320 in EOC. MAPK1 expression was markedly upregulated in EOC tissues and inversely correlated with miR‑320 expression. Furthermore, silencing of MAPK1 by RNA interference inhibited cell proliferation and invasion of EOC cells. Overall, the present study demonstrated that miR‑320 may act as a useful diagnostic and therapeutic target in the treatment of EOC.

Introduction

Ovarian cancer is the second most common cancer worldwide, and the most fatal gynaecological malignancy of all gynecological cancers, with over 238,700 newly diagnosed cases and 151,900 fatalities per year (1,2). Epithelial ovarian cancer (EOC) accounts for ~90% of all ovarian cancer cases, and consists of five subtypes, including high-grade serous carcinoma (70%), low-grade serous carcinoma (<5%), mucinous carcinoma (3%), endometrioid carcinoma (10%) and clear-cell carcinoma (10%) (3). Despite progress in the traditional treatments for EOC, the overall survival rate for patients with this malignancy remains dissatisfactory over the past 50 years (4). Furthermore, an increased number of patients are presenting with EOC with local or distant metastasis at the time of diagnosis, due to an absence of early diagnostic biomarkers, and this results in poor prognosis and short survival time (5). Therefore, further investigations are necessary in order to elucidate the underlying molecular mechanisms of EOC occurrence and progression, and identify novel efficient targets for diagnosis, therapy and prognosis of this disease.

MicroRNAs (miRNAs) represent a large family of non-coding, single stranded, endogenous and short RNA molecules with 18–25 nucleotides (6). miRNAs regulate gene expression by base-pairing with the 3′ untranslated regions (3′UTRs) of their target genes, resulting in translational suppression or mRNA degradation, and ultimately controlling the protein expression of target genes (7). It has previously been demonstrated that miRNAs are important in various biological processes, including cell proliferation, cell cycle, differentiation and metastasis (810). miRNAs have been reported to be downregulated or upregulated in a variety of human malignancies (1113). Furthermore, previous studies demonstrated that deregulated miRNAs are involved in the formation and progression of the majority of human cancers, including EOC (14), bladder (15), gastric (16), glioma (17) and breast cancers (18). These abnormally expressed miRNAs may function as oncogenes or tumor suppressor genes depending on the roles of their target genes and tumor types (19). These findings suggest that miRNAs may be useful in the diagnosis and prognosis of human cancers, and may additionally act as therapeutic targets for their treatment.

Abnormal expression of miR-320 has been reported in multiple types of cancer, including breast (20,21), gastric (22), colorectal (23), glioma (24,25) and bladder cancers (26). However, the role of miR-320 in EOC remains to be elucidated. The present study aimed to investigate the expression pattern and regulatory role of miR-320 in EOC, and its associated underlying mechanism.

Materials and methods

Ethical approval and human tissue

The present study was approved by the Ethics Committee of Shengli Oilfield Central Hospital (Dongying, China). In addition, written informed consent was obtained from all patients. A total of 56 EOC tumor tissues and their paired adjacent normal ovarian epithelium tissues were obtained from the Department of Gynaecology and Obstetrics, Shengli Oilfield Central Hospital, between 2012 and 2015. None of these EOC patients (n=56; female; age, 39–72 years) were treated with other treatments prior to surgery. Tissues specimens were immediately snap-frozen in liquid nitrogen and stored at −70°C in a freezer.

Cell lines

A total of 4 EOC cell lines (CAOV3, OVCAR3, SKOV3, ES-2) and the human normal ovarian epithelial cell line NOEC, were purchased from American Type Culture Collection (Manassas, VA, USA). EOC cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin, all obtained from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). NOEC cells were grown in Ham's F-12 medium (Gibco; Thermo Fisher Scientific Inc.) with 10% FBS and 1% penicillin/streptomycin. All cells were maintained in a humidified environment at 37°C with 5% CO2.

Cell transfection

miR-320 mimics, the corresponding negative miRNA mimics controls (miR-NC), small interfering (si)RNA targeting mitogen activated protein kinase (MAPK; si-MAPK1) and its negative control scrambled siRNA (si-NC) were synthesized by Shanghai GenePharma Co., Ltd (Shanghai, China). The miR-320 mimics sequence was 5′-AAAAGCUGGGUUGAGAGGGGCG-3′ and the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. The si-MAPK1 sequence was 5′-AGUUCGAGUAUACUUCAAGUU-3′ and the si-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. Cells were seeded into 6-well plates at a density of 50–60% confluence in FBS-free RPMI-1640 medium for 1 day prior to transfection. Cells were transfected with miR-320 mimics (100 pmol), miR-NC (100 pmol), si-MAPK1 (100 pmol) or si-NC (100 pmol) by using Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Cell culture medium was replaced with RPMI-1640 medium containing 10% FBS at 8 h post-transfection. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to detect miR-320 and MAPK1 mRNA expression at 48 h post-transfection. Western blotting analysis was applied to detect MAPK1 protein expression at 72 h post-transfection. The Cell Counting kit 8 (CCK8) and cell invasion assays were performed at 24 h and 48 h following transfection, respectively.

RNA extraction and RT-qPCR

Total RNA was isolated from tissues and cells using a TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. For quantification of miR-320, reverse transcription was conducted with TaqMan MicroRNA Reverse Transcription Kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) and followed by qPCR with TaqMan Human MicroRNA assay kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). The cycling conditions were as follows: 50°C for 2 min, 95°C for 10 min, followed by 40 cycles of denaturation at 95°C for 15 sec and annealing/extension at 60°C for 1 min. For MAPK1 mRNA expression, M-MLV Reverse Transcription system (Promega Corporation, Madison, WI, USA) was used to synthesize cDNA. The detection of MAPK1 mRNA expression was conducted using SYBR Premix Ex Taq (Takara Biotechnology Co., Ltd, Dalian, China). The thermocycling conditions were as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. U6 small nuclear RNA and β-actin were used as internal standard references for miR-320 and MAPK1, respectively. The primers sequences were designed as follows: miR-320, 5′-ACACTCCAGCTGGGAAAAGCTGGGTTGAGA-3′ (forward) and 5′-TGGTGTCGTGGAGTCG-3′ (reverse); U6, 5′-CTCGCTTCGGCAGCACA-3′ (forward) and 5′-AACGCTTCACGAATTTGCGT-3′ (reverse); MAPK1, 5′-TGGATTCCCTGGTTCTCTCTAAAG-3′ (forward) and 5′-GGGTCTGTTTTCCGAGGATGA-3′ (reverse); and β-actin, 5′-CCTGGCACCCAGCACAAT-3′ (forward) and 5′-GCTGATCCACATCTGCTGGAA-3′ (reverse). Relative expression was quantified by the 2−ΔΔCq method (27).

CCK8 assay

Cell proliferation was evaluated by using a CCK8 assay (Dojindo Molecular Technologies, Inc., Kumamoto, Japan). A total of 24 h following transfection, transfected cells were seeded in 96-well plates (3,000 cells/well). Cells were then incubated in a humidified environment at 37°C with 5% CO2 for 4 consecutive days. At every 24 h, a CCK8 assay was performed according to the manufacturer's protocol. Briefly, 10 µl CCK8 reagent was added into each well. Following incubation at 37°C for an additional 4 h, absorbance at a wavelength of 450 nm was measured using a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Each assay was performed in triplicate.

Cell invasion assay

Transwell chambers (8-mm pore size; Costar; Corning Incorporated, Corning, NY, USA) precoated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA) were used to conduct the cell invasion assay. A total of 500 µl RPMI-1640 medium containing 20% FBS was added into the lower chamber, and 5×104 transfected cells in 200 µl FBS-free culture medium were plated in the upper chamber. Following incubation for 48 h at 37°C, cells that remained in the upper surface of the membrane were removed by cotton swabs. The invaded cells were fixed with 4% paraformaldehyde at room temperature for 10 min and stained with 0.1% crystal violet at room temperature for 10 min. Following washing, five randomly selected visual fields per membrane were photographed and counted under an inverted fluorescence microscope (magnification, ×200; CKX41; Olympus Corporation, Tokyo, Japan).

miR-320 target prediction

The computational methods TargetScan (www.targetscan.org) and PicTar (www.pictar.mdc-berlin.de/) were used to predict the potential targets of miR-320.

Luciferase assay

Cells were seeded in 24-well plates at a density of 40–50% confluence. Following incubation overnight, Lipofectamine® 2000 was employed to co-transfect cells with miR-320 mimics or miR-NC, and psiCHECK wild-type MAPK1 3′UTR luciferase plasmid (psiCHECK-Wt-MAPK1-3′UTR; Shanghai GenePharma Co., Ltd) or psiCHECK mutant MAPK1 3′UTR luciferase plasmid (psiCHECK-Mut-MAPK1-3′UTR; Shanghai GenePharma Co., Ltd). At 48 h following transfection, the cells were harvested and subjected to luciferase assay by using the Dual-Luciferase® Reporter Assay system (Promega Corporation). Firefly luciferase activity was normalized to Renilla luciferase activity.

Western blotting

Total protein was extracted from transfected cells at 72 h post-transfection with ice-cold radioimmunoprecipitation assay lysis buffer containing proteinase inhibitor (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). Concentrations of total protein were detected using a bicinchoninic assay kit (Pierce; Thermo Fisher Scientific, Inc.). Equal amounts protein (20 µg) were resolved using SDS-PAGE on a 10% gel. Subsequently, proteins were transferred to polyvinylidene difluoride membranes and then blocked with 5% non-fat milk in Tris-buffered saline containing 0.1% Tween-20 (TBST) at room temperature for 1 h. The membranes were incubated with primary antibodies at 4°C overnight, followed by washing with TBST three times and incubated with a goat anti-mouse horseradish peroxidase-conjugated secondary antibody (1:5,000 dilution; catalog no. sc-2005; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at room temperature for 2 h. Finally, protein bands were visualized using an enhanced chemiluminescence solution (Bio-Rad Laboratories, Inc.) and analyzed using Quantity One software, version 4.62 (Bio-Rad Laboratories, Inc.). Primary antibodies used in the present study included mouse anti-human MAPK1 monoclonal antibody (1:1,000 dilution; catalog no. sc-81459; Santa Cruz Biotechnology, Inc.) and mouse anti-human β-actin monoclonal antibody (1:1,000 dilution; catalog no. sc-47778; Santa Cruz Biotechnology, Inc.). β-actin was used as a loading control.

Statistical analysis

Data are expressed as the mean ± standard deviation. All statistical analyses were performed with Student's t-tests or one-way analysis of variance using SPSS software, version 18.0 (SPSS, Inc., Chicago, IL, USA). The correlation between miR-320 and MAPK1 mRNA expression was analyzed with Spearman's correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-320 expression is downregulated in EOC tissues and cell lines

To assess miR-320 expression levels, RT-qPCR was performed in 56 EOC tumor tissues and matched adjacent normal ovarian epithelium tissues. The data indicated that expression levels of miR-320 were decreased in EOC tissues compared with in matched adjacent normal ovarian epithelium tissues (Fig. 1A; P<0.05). Following this, miR-320 expression was quantified in a panel of EOC cell lines in addition to the human normal ovarian epithelial cell line NOEC. Compared with NOEC, miR-320 expression was significantly downregulated in all four tested EOC cell lines (Fig. 1B).

Furthermore, correlation between miR-320 and the clinicopathological variables of patients with EOC was investigated. As presented in Table I, miR-320 expression was strongly correlated with FIGO stage (P=0.013) and lymph node metastasis (P=0.001). However, no correlation was observed with other clinicopathological characteristics, including age, differentiation and tumor size (all P>0.05). These results suggested that miR-320 may be important in EOC formation and progression.

Table I.

Correlation of miRNA-320 with clinical characteristics in patients with epithelial ovarian cancer.

Table I.

Correlation of miRNA-320 with clinical characteristics in patients with epithelial ovarian cancer.

miR-320 expression

FeaturesNo. of patientsLowHighP-value
Age (years) 0.269
  <60291415
  ≥60271710
FIGO stage 0.013
  I–II301218
  III–IV26197
Differentiation 0.420
  1/2281711
  3281414
Tumor size (cm) 0.243
  <525169
  ≥5311516
Lymph node metastasis 0.001
  Negative27918
  Positive29227

[i] FIGO, Federation of Gynecology and Obstetrics; miR-320, micro RNA-320.

Overexpression of miR-320 suppresses cell proliferation and invasion in EOC

To explore the biological role of miR-320 in EOC, a gain-of-function analysis was conducted. CAOV3 and OVCAR3 cells were transfected with miR-320 mimics or miR-NC. A total of 48 h following transfection, RT-qPCR was performed to detect miR-320 expression and it was observed that miR-320 expression levels in CAOV3 and OVCAR3 cells were significantly increased following transfection with miR-320 mimics (Fig. 2A; P<0.05). To determine if miR-320 contributes to EOC progression, CCK8 and cell invasion assays were performed in CAOV3 and OVCAR3 cells following transfection with miR-320 mimics or miR-NC. CCK8 assay demonstrated that upregulation of miR-320 decreased CAOV3 and OVCAR3 cell proliferation (Fig. 2B; P<0.05). Similarly, overexpression of miR-320 resulted in a significant reduction of cell invasion capacity of the CAOV3 and OVCAR3 cells (Fig. 2C; P<0.05). These findings suggested that miR-320 may suppress EOC cell growth and metastasis.

MAPK1 is a direct target of miR-320 in EOC

To explore the mechanisms underlying the tumor suppressive role of miR-320 in EOC, Targetscan and PicTar were used to predict the potential targets of miR-320. As presented in Fig. 3A, the seed sequence of miR-320 was complementary to the 3′UTR of MAPK1. MAPK1 is overexpressed in EOC tissues and cell lines (28,29), and contributes to the tumorigenesis and tumor development in EOC (28), which led to the hypothesis that MAPK1 may be a direct target of miR-320 in EOC. To determine whether MAPK1 is a direct target gene of miR-320, a luciferase reporter assay was performed in CAOV3 and OVCAR3 cells co-transfected with miR-320 mimics or miR-NC, and luciferase reporter vector containing the wild type or mutant 3′UTR of MAPK1. The results demonstrated that transfection of miR-320 resulted in a significant inhibition of luciferase activities by psiCHECK-Wt-MAPK1-3′UTR (Fig. 3B; P<0.05). However, these repressive effects of miR-320 on luciferase activities were reversed following transfection with psiCHECK-Mut-MAPK1-3′UTR. The present study then sought to investigate whether ectopic expression of miR-320 regulated endogenous MAPK1 expression. RT-qPCR and western blotting verified that upregulation of miR-320 suppressed MAPK1 expression in CAOV3 and OVCAR3 cells at the mRNA (Fig. 3C; P<0.05) and protein (Fig. 3D; P<0.05) levels. Collectively, these results suggested that miR-320 decreased MAPK1 expression by targeting specific sites within the 3′UTR of MAPK1.

MAPK1 is upregulated in EOC tissues and negatively correlated with miR-320 expression

MAPK1 was identified as a direct target gene of miR-320 in EOC; therefore, MAPK1 expression in EOC tissues and matched adjacent normal ovarian epithelium tissues was measured. As expected, MAPK1 mRNA was significantly increased in EOC tissues, compared with normal ovarian epithelium tissues (Fig. 4A; P<0.05). Furthermore, Spearman's correlation analysis indicated an inverse correlation between MAPK1 mRNA and miR-320 expression (Fig. 4B; r=-0.4078; P=0.0018).

miR-320 inhibits cell proliferation and invasion of EOC by downregulation of MAPK1

To evaluate the role of MAPK1 in EOC, a loss-of-function assay was performed. CAOV3 and OVCAR3 cells were injected with si-MAPK1 or si-NC. Western blotting verified that MAPK1 protein was downregulated in CAOV3 and OVCAR3 cells following transfection with si-MAPK1 (Fig. 5A; P<0.05). CCK8 assay demonstrated that downregulation of MAPK1 suppressed proliferation in CAOV3 and OVCAR3 cells, which was consistent with miR-320 overexpression (Fig. 5B; P<0.05). Furthermore, consistent with miR-320 mimics, cell invasive abilities were decreased in si-MAPK1-transfected CAOV3 and OVCAR3 cell lines (Fig. 5C; P<0.05). These results demonstrated that miR-320 inhibits cell proliferation and invasion of EOC by negative regulation of MAPK1.

Discussion

Previous studies have suggested that miRNAs are important in tumorigenesis and tumor development, and thus may prove as novel targets for the treatment and prognosis of various cancers (30,31). Abnormal expression of miR-320 has been reported in various types of cancers, including breast (20,21), gastric (22), colorectal (23), glioma (24,25), bladder (26) and cervical cancers (32). In addition, expression levels of miR-320 have been demonstrated to be correlated with clinicopathological variables of multiple human cancers. For example, in non-small cell lung cancer, miR-320 is correlated with TNM classification and metastasis (33). In breast cancer, a significant correlation has been observed between low miR-320 expression level and tumor size, clinical stage, lymph node metastasis and distant metastasis (34). The present study measured miR-320 expression in EOC tissues and cell lines. Data from RT-qPCR demonstrated that miR-320 was significantly downregulated in EOC tissues and cell lines. Low miR-320 expression was significantly correlated with FIGO stage and lymph node metastasis of EOC patients. These findings suggested that miR-320 deregulation is a common event in human cancer, and may be important in tumorigenesis and tumor development.

Previous studies have demonstrated that miR-320 regulates the formation and progression of human cancer. Introduction of miR-320 inhibits cell proliferation in osteosarcoma (35), colorectal adenoma (36), non-small cell lung cancer (33), cervical cancer (32), glioma (25), multiple myeloma (37) and breast cancer (21). Additionally, upregulation of miR-320 results in a significant decrease in the motility of breast cancer (20,21), salivary adenoid cystic carcinoma (38), nasopharyngeal carcinoma (39), glioma (24) and non-small cell lung cancer (33). It has previously been demonstrated that miR-320 promotes Fluorouracil resistance in pancreatic cancer (40), enhances the chemosensitivity of tamoxifen-resistant breast cancer cells to tamoxifen (41), improves the chemosensitivity and radiosensitivity of colon cancer (42), and represses tube formation of vascular endothelial cells in oral cancer (43). In the present study, the CCK8 assay revealed that miR-320 inhibited cell growth in EOC cells. The cell invasion assay indicated that restoration of expression of miR-320 decreased invasion activity in EOC cells. Collectively, these experiments indicated that miR-320 may act as a tumor suppressor in human cancers, and may be used as a novel molecular therapeutic target for anti-tumor treatments.

The present study then aimed to investigate the molecular mechanism by which miR-320 acts as a tumor suppressor in EOC. Previous studies identified numerous targets of miR-320 including E2F transcription factor 1 (35), cyclin dependent kinase 6 (36), MCI1 (32), PBX Homeobox 3 (37), RAB11A (21) and metadherin (20). To explore the targets of miR-320, Targetscan and PicTar were used. MAPK1 was predicated to be a potential target of miR-320. A luciferase reporter assay was then performed to verify that miR-320 directly targeted the 3′UTR of MAPK1. Subsequently, it was demonstrated that miR-320 negatively regulated MAPK1 expression at the mRNA and protein level in EOC cells. Furthermore, MAPK1 expression was upregulated in EOC tissues and negatively correlated with miR-320 expression. Additionally, consistent with miR-320 overexpression, cell proliferation and invasion were decreased in si-MAPK1-transfected EOC cells. These results verified MAPK1 as a novel direct target of miR-320 in EOC.

The MAPK signaling cascades are composed of membrane-to-nucleus signaling modules which are important in multiple physiological processes (44). MAPK1, additionally termed, extracellular regulated kinase 2, has been reported to be abnormally expressed in various human cancers, including cervical (45), myeloma (46), sacral chordoma (47), non-small cell lung cancer (48) and gastric cancer (49). Rahman et al (29) reported that MAPK1 is highly expressed in ovarian cancer tissues and cell lines (28,29). Functional assays have demonstrated that MAPK1 underexpression suppresses growth and metastasis in ovarian cancer SKOV3 cells (28). In accordance with previous studies, the results of the present study demonstrated that MAPK1 was significantly upregulated in EOC tissues, and the downregulation of MAPK1 repressed the proliferation and invasive ability of EOC cells. MAPK1 may be investigated as a useful therapeutic target for the treatment of patients with EOC.

In conclusion, the results of the present study demonstrated that miR-320 was decreased in EOC tissues and cell lines. Low miR-320 expression was significantly correlated with FIGO stage and lymph node metastasis of EOC patients. Furthermore, ectopic expression of miR-320 inhibited EOC cell proliferation and invasion through directly targeting MAPK1. These findings may provide a novel insight into the potential carcinogenic and progressive mechanisms in EOC, and may be used in the development of novel treatment strategies for patients with this malignancy. Further investigations are required to explore whether the potential of miR-320 may be fully realised in EOC.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Prat J: Ovarian carcinomas: Five distinct diseases with different origins, genetic alterations, and clinicopathological features. Virchows Arch. 460:237–249. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Wang Y, Kim S and Kim IM: Regulation of metastasis by microRNAs in ovarian cancer. Front Oncol. 4:1432014. View Article : Google Scholar : PubMed/NCBI

5 

Tung CS, Wong KK and Mok SC: Biomarker discovery in ovarian cancer. Womens Health (Lond). 4:27–40. 2008. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Vasudevan S: Posttranscriptional upregulation by microRNAs. Wiley Interdiscip Rev RNA. 3:311–330. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Esteller M: Non-coding RNAs in human disease. Nat Rev Genet. 12:861–874. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Seashols-Williams SJ, Budd W, Clark GC, Wu Q, Daniel R, Dragoescu E and Zehner ZE: miR-9 acts as an oncomiR in prostate cancer through multiple pathways that drive tumour progression and metastasis. PLoS One. 11:e01596012016. View Article : Google Scholar : PubMed/NCBI

10 

Sulaiman SA, Ab Mutalib NS and Jamal R: miR-200c regulation of metastases in ovarian cancer: Potential role in epithelial and mesenchymal transition. Front Pharmacol. 7:2712016. View Article : Google Scholar : PubMed/NCBI

11 

Xiao L, Zhou H, Li XP, Chen J, Fang C, Mao CX, Cui JJ, Zhang W, Zhou HH, Yin JY and Liu ZQ: MicroRNA-138 acts as a tumor suppressor in non small cell lung cancer via targeting YAP1. Oncotarget. 7:40038–40046. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Das DK, Naidoo M, Ilboudo A, Park JY, Ali T, Krampis K, Robinson BD, Osborne JR and Ogunwobi OO: miR-1207-3p regulates the androgen receptor in prostate cancer via FNDC1/fibronectin. Exp Cell Res. 348:190–200. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Gopalan V, Islam F, Pillai S, Tang JC, Tong DK, Law S, Chan KW and Lam AK: Overexpression of microRNA-1288 in oesophageal squamous cell carcinoma. Exp Cell Res. 348:146–154. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Liu J, Dou Y and Sheng M: Inhibition of microRNA-383 has tumor suppressive effect in human epithelial ovarian cancer through the action on caspase-2 gene. Biomed Pharmacother. 83:1286–1294. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Xiao J, Lin HY, Zhu YY, Zhu YP and Chen LW: MiR-126 regulates proliferation and invasion in the bladder cancer BLS cell line by targeting the PIK3R2-mediated PI3K/Akt signaling pathway. Onco Targets Ther. 9:5181–5193. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wu D, Niu X, Pan H, Zhou Y, Zhang Z, Qu P and Zhou J: Tumor-suppressing effects of microRNA-429 in human renal cell carcinoma via the downregulation of Sp1. Oncol Lett. 12:2906–2911. 2016.PubMed/NCBI

17 

Zhou Y, Liu Y, Hu C and Jiang Y: MicroRNA-16 inhibits the proliferation, migration and invasion of glioma cells by targeting Sal-like protein 4. Int J Mol Med. 38:1768–1776. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Pan Y, Jiao G, Wang C, Yang J and Yang W: MicroRNA-421 inhibits breast cancer metastasis by targeting metastasis associated 1. Biomed Pharmacother. 83:1398–1406. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Ventura A and Jacks T: MicroRNAs and cancer: Short RNAs go a long way. Cell. 136:586–591. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Yu J, Wang JG, Zhang L, Yang HP, Wang L, Ding D, Chen Q, Yang WL, Ren KH, Zhou D, et al: MicroRNA-320a inhibits breast cancer metastasis by targeting metadherin. Oncotarget. 7:38612–38625. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Wang B, Yang Z, Wang H, Cao Z, Zhao Y, Gong C, Ma L, Wang X, Hu X and Chen S: MicroRNA-320a inhibits proliferation and invasion of breast cancer cells by targeting RAB11A. Am J Cancer Res. 5:2719–2729. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Wang Y, Zeng J, Pan J, Geng X, Li L, Wu J, Song P, Wang Y, Liu J and Wang L: MiR-320a inhibits gastric carcinoma by targeting activity in the FoxM1-P27KIP1 axis. Oncotarget. 7:29275–29286. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Zhao H, Dong T, Zhou H, Wang L, Huang A, Feng B, Quan Y, Jin R, Zhang W, Sun J, et al: miR-320a suppresses colorectal cancer progression by targeting Rac1. Carcinogenesis. 35:886–895. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Guo T, Feng Y, Liu Q, Yang X, Jiang T, Chen Y and Zhang Q: MicroRNA-320a suppresses in GBM patients and modulates glioma cell functions by targeting IGF-1R. Tumour Biol. 35:11269–11275. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Sun JY, Xiao WZ, Wang F, Wang YQ, Zhu YH, Wu YF, Miao ZL and Lin YC: MicroRNA-320 inhibits cell proliferation in glioma by targeting E2F1. Mol Med Rep. 12:2355–2359. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Shang C, Zhang H, Guo Y, Hong Y, Liu Y and Xue Y: MiR-320a down-regulation mediates bladder carcinoma invasion by targeting ITGB3. Mol Biol Rep. 41:2521–2527. 2014. View Article : Google Scholar : PubMed/NCBI

27 

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

28 

Yiwei T, Hua H, Hui G, Mao M and Xiang L: HOTAIR interacting with MAPK1 regulates ovarian cancer skov3 cell proliferation, migration, and invasion. Med Sci Monit. 21:1856–1863. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Rahman MT, Nakayama K, Rahman M, Katagiri H, Katagiri A, Ishibashi T, Ishikawa M, Sato E, Iida K, Nakayama N, et al: KRAS and MAPK1 gene amplification in type II ovarian carcinomas. Int J Mol Sci. 14:13748–13762. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Wu WK, Lee CW, Cho CH, Fan D, Wu K, Yu J and Sung JJ: MicroRNA dysregulation in gastric cancer: A new player enters the game. Oncogene. 29:5761–5771. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Miao J, Wu S, Peng Z, Tania M and Zhang C: MicroRNAs in osteosarcoma: Diagnostic and therapeutic aspects. Tumour Biol. 34:2093–2098. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Zhang T, Zou P, Wang T, Xiang J, Cheng J, Chen D and Zhou J: Down-regulation of miR-320 associated with cancer progression and cell apoptosis via targeting Mcl-1 in cervical cancer. Tumour Biol. 37:8931–8940. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Lei T, Zhu Y, Jiang C, Wang Y, Fu J, Fan Z and Qin H: MicroRNA-320 was downregulated in non-small cell lung cancer and inhibited cell proliferation, migration and invasion by targeting fatty acid synthase. Mol Med Rep. 14:1255–1262. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Yang H, Yu J, Wang L, Ding D, Zhang L, Chu C, Chen Q, Xu Z, Zou Q and Liu X: miR-320a is an independent prognostic biomarker for invasive breast cancer. Oncol Lett. 8:1043–1050. 2014.PubMed/NCBI

35 

Wu H, Li W, Zhang M, Zhu S, Zhang D and Wang X: Inhibitory roles of miR-320 in osteosarcoma via regulating E2F1. J Cancer Res Ther. 12:68–71. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Tadano T, Kakuta Y, Hamada S, Shimodaira Y, Kuroha M, Kawakami Y, Kimura T, Shiga H, Endo K, Masamune A, et al: MicroRNA-320 family is downregulated in colorectal adenoma and affects tumor proliferation by targeting CDK6. World J Gastrointest Oncol. 8:532–542. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Lu Y, Wu D, Wang J, Li Y, Chai X and Kang Q: miR-320a regulates cell proliferation and apoptosis in multiple myeloma by targeting pre-B-cell leukemia transcription factor 3. Biochem Biophys Res Commun. 473:1315–1320. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Sun L, Liu B, Lin Z, Yao Y, Chen Y, Li Y, Chen J, Yu D, Tang Z, Wang B, et al: MiR-320a acts as a prognostic factor and Inhibits metastasis of salivary adenoid cystic carcinoma by targeting ITGB3. Mol Cancer. 14:962015. View Article : Google Scholar : PubMed/NCBI

39 

Qi X, Li J, Zhou C, Lv C and Tian M: MicroRNA-320a inhibits cell proliferation, migration and invasion by targeting BMI-1 in nasopharyngeal carcinoma. FEBS Lett. 588:3732–3738. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Wang W, Zhao L, Wei X, Wang L, Liu S, Yang Y, Wang F, Sun G, Zhang J, Ma Y, et al: MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Sci Rep. 6:276412016. View Article : Google Scholar : PubMed/NCBI

41 

Lu M, Ding K, Zhang G, Yin M, Yao G, Tian H, Lian J, Liu L, Liang M, Zhu T and Sun F: MicroRNA-320a sensitizes tamoxifen-resistant breast cancer cells to tamoxifen by targeting ARPP-19 and ERRγ. Sci Rep. 5:87352015. View Article : Google Scholar : PubMed/NCBI

42 

Wan LY, Deng J, Xiang XJ, Zhang L, Yu F, Chen J, Sun Z, Feng M and Xiong JP: miR-320 enhances the sensitivity of human colon cancer cells to chemoradiotherapy in vitro by targeting FOXM1. Biochem Biophys Res Commun. 457:125–132. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Wu YY, Chen YL, Jao YC, Hsieh IS, Chang KC and Hong TM: miR-320 regulates tumor angiogenesis driven by vascular endothelial cells in oral cancer by silencing neuropilin 1. Angiogenesis. 17:247–260. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Seger R and Krebs EG: The MAPK signaling cascade. FASEB J. 9:726–735. 1995.PubMed/NCBI

45 

Li XW, Tuergan M and Abulizi G: Expression of MAPK1 in cervical cancer and effect of MAPK1 gene silencing on epithelial-mesenchymal transition, invasion and metastasis. Asian Pac J Trop Med. 8:937–943. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Tsubaki M, Takeda T, Ogawa N, Sakamoto K, Shimaoka H, Fujita A, Itoh T, Imano M, Ishizaka T, Satou T and Nishida S: Overexpression of survivin via activation of ERK1/2, Akt, and NF-κB plays a central role in vincristine resistance in multiple myeloma cells. Leuk Res. 39:445–452. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Zhang K, Chen H, Zhang B, Sun J, Lu J, Chen K and Yang H: Overexpression of Raf-1 and ERK1/2 in sacral chordoma and association with tumor recurrence. Int J Clin Exp Pathol. 8:608–614. 2015.PubMed/NCBI

48 

You B, Yang YL, Xu Z, Dai Y, Liu S, Mao JH, Tetsu O, Li H, Jablons DM and You L: Inhibition of ERK1/2 down-regulates the Hippo/YAP signaling pathway in human NSCLC cells. Oncotarget. 6:4357–4368. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Fei B and Wu H: MiR-378 inhibits progression of human gastric cancer MGC-803 cells by targeting MAPK1 in vitro. Oncol Res. 20:557–564. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 16 Issue 6

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
Xu Y, Hu J, Zhang C and Liu Y: MicroRNA‑320 targets mitogen‑activated protein kinase 1 to inhibit cell proliferation and invasion in epithelial ovarian cancer Retraction in /10.3892/mmr.2024.13169. Mol Med Rep 16: 8530-8536, 2017
APA
Xu, Y., Hu, J., Zhang, C., & Liu, Y. (2017). MicroRNA‑320 targets mitogen‑activated protein kinase 1 to inhibit cell proliferation and invasion in epithelial ovarian cancer Retraction in /10.3892/mmr.2024.13169. Molecular Medicine Reports, 16, 8530-8536. https://doi.org/10.3892/mmr.2017.7664
MLA
Xu, Y., Hu, J., Zhang, C., Liu, Y."MicroRNA‑320 targets mitogen‑activated protein kinase 1 to inhibit cell proliferation and invasion in epithelial ovarian cancer Retraction in /10.3892/mmr.2024.13169". Molecular Medicine Reports 16.6 (2017): 8530-8536.
Chicago
Xu, Y., Hu, J., Zhang, C., Liu, Y."MicroRNA‑320 targets mitogen‑activated protein kinase 1 to inhibit cell proliferation and invasion in epithelial ovarian cancer Retraction in /10.3892/mmr.2024.13169". Molecular Medicine Reports 16, no. 6 (2017): 8530-8536. https://doi.org/10.3892/mmr.2017.7664