MicroRNA‑433 reduces cell proliferation and invasion in non‑small cell lung cancer via directly targeting E2F transcription factor 3

Retraction in: /10.3892/mmr.2022.12857

  • Authors:
    • Nian Liu
    • Zhiguang Liu
    • Weidong Zhang
    • Yang Li
    • Jun Cao
    • Huan Yang
    • Xiuying Li
  • View Affiliations

  • Published online on: May 14, 2018     https://doi.org/10.3892/mmr.2018.9020
  • Pages: 1155-1164
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNA/miRs) have been associated with the initiation and progression of non‑small‑cell lung cancer (NSCLC). Hence, a comprehensive understanding of the association between dysregulated miRNAs and NSCLC may contribute to the identification of novel therapeutic methods for patients with NSCLC. MiRNA‑433 (miR‑433) has been reported to be dysregulated in numerous types of human cancers; however, its expression pattern, biological roles and associated mechanisms in NSCLC require further investigation. The present study aimed to detect miR‑433 expression and determine its roles and underlying molecular mechanisms in NSCLC. In the present study, reverse transcription‑quantitative polymerase chain reaction revealed that miR‑433 was significantly downregulated in NSCLC tissues and cell lines. This decreased miR‑433 expression was strongly associated with the tumor node metastasis stage and lymph node metastasis of patients with NSCLC. Cell Counting kit‑8 and cell invasion assays revealed that the resumption of miR‑433 expression decreased the proliferation and invasion of NSCLC cells. Bioinformatics analysis predicted E2F transcription factor 3 (E2F3) as a potential target of miR‑433. Luciferase reporter assay, RT‑qPCR and western blot analysis further demonstrated that E2F3 was a direct target of miR‑433 in NSCLC. E2F3 downregulation induced by small interfering RNA exhibited inhibitory effects similar to those of miR‑433 overexpression in NSCLC cells, and the restored E2F3 expression counteracted the suppressive effects on NSCLC cells induced by miR‑433 overexpression. Therefore, miR‑433 may inhibit the progression of NSCLC, at least in part, by targeting E2F3. The present study indicated that miR‑433 may be investigated as an innovative candidate target for the therapy of patients with this fatal disease.

Introduction

Lung cancer is the most common type and leading cause of cancer-associated mortality in men and women worldwide (1,2). It can be divided into small- and non-small-cell lung cancer (NSCLC) (3). NSCLC, the most prevalent lung cancer subtype, constitutes 80–85% of the total number of lung cancer cases (4). Surgical management followed by adjuvant chemotherapy is the major treatment for patients at early disease stages (5); however, over half of NSCLC cases manifest into advanced disease stages and become unfit for surgical resection (6). Improvements in NSCLC treatment have been made; however, the clinical outcomes of these patients are unsatisfactory, with an overall 5-year survival rate of <15% (7). Such outcomes are mainly due to late disease presentation, tumor heterogeneities within histological subtypes and a relatively poor understanding of NSCLC pathogenesis (8,9). Thus, molecular mechanisms associated with the occurrence and development of NSCLC should be fully understood in order to identify novel therapeutic treatments for patients with NSCLC.

MicroRNAs (miRNAs) are a series of endogenous, noncoding and highly conserved short RNAs with a length of 19–25 nucleotides (10). MiRNAs participate in gene regulation by directly interacting with the 3′-untranslated regions (3′-UTRs) of their target genes in a base pairing manner; consequently, mRNA is degraded or translation is suppressed (11). More than a thousand miRNAs are encoded by the mammalian genome and these miRNAs likely modulate over one-third of all human protein-coding genes (12,13). Aberrantly expressed miRNAs are often associated with a variety of disorders, such as NSCLC (14), breast cancer (15) and glioblastoma (16). Providing their regulatory function in gene expression, miRNAs have been reported to be associated with tumorigenesis and tumor development via the regulation of cell proliferation, cell cycle, apoptosis, angiogenesis, migration, invasion and metastasis (1719). Dysregulated miRNAs in human malignancy can serve as tumor suppressors or oncogenes, depending on the biological behaviours of their target genes (20). Hence, miRNAs may be regarded as novel targets for the identification of effective therapeutic methods for patients with cancer.

MiR-433 is aberrantly expressed in numerous human cancers (2124); however, its expression pattern, biological functions and associated mechanisms in NSCLC require further investigation. The present study aimed to investigate the expression of miR-433 and determine its roles and underlying mechanisms in NSCLC.

Materials and methods

Acquisition of tissue samples

The present study was approved by the Ethics Committee of Hunan Provincial People's Hospital (Changsha, China). The use of these tissue samples was approved by all of the patients prior to participation in the present study, and written informed consent was obtained from all patients with NSCLC. Paired NSCLC tissues and adjacent non-tumor lung tissues were collected from 47 patients (26 males and 21 females; age range, 39–72 years old) with NSCLC who underwent surgical resection at Hunan Provincial People's Hospital between June 2014 and October 2016. None of the patients underwent chemotherapy or radiotherapy prior to surgery. On the basis of the miR-433 median level, all patients with NSCLC were assigned into either miR-433 low-expression group (n=24) and miR-433 high-expression group (n=23). All tissues were quickly snap-frozen in liquid nitrogen following excision and then stored at −80°C until further experimentation.

Cell culture and transfection

A non-tumorigenic bronchial epithelium BEAS-2B cell line and four NSCLC cell lines (SK-MES-1, A549, H522 and H460) were obtained from the Shanghai Institute of Biochemistry and Cell Biology (Shanghai, China). BEAS-2B cells were cultured in LHC-9 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.). All NSCLC cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM) containing 10% FBS, 100 U/ml penicillin G and 100 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.). All cells were cultured at 37°C in a humidified atmosphere with 5% CO2. A549 and H460 cells were revealed to express relatively low levels of miR-432 compared with SK-MES-1 and H522 cells; therefore, A549 and H460 cell lines were chosen for subsequent functional experiments.

MiR-433 mimics and miRNA mimics negative control (miR-NC) were purchased from Guangzhou RiboBio Co., Ltd. (Guangzhou, China). The miR-433 mimic sequence was 5′-AUCAUGAUGGGCUCCUCGGUGU-3′ and the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. Small interfering RNA (siRNA) against the expression of E2F transcription factor 3 (E2F3; E2F3 siRNA), negative control siRNA (NC siRNA), E2F3 overexpression plasmid pcDNA3.1-E2F3 and blank pcDNA3.1 plasmid were chemically synthesized by GeneCopoeia, Inc. (Rockville, MD, USA). The E2F3 siRNA sequence was 5′-GCACTACGAAGTCCAGATA-3′ and the NC siRNA sequence was 5′-UUUTGATCAUTGATGAAA-3′. Cells were plated into 6-well cell culture plates and cultured to 60–70% confluence. miRNA mimics (100 pmol), siRNAs (100 pmol) or blank plasmids (4 µg) were transfected into A549 and H460 cells using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. A total of 8 h post-transfection, the culture medium in each well was replaced with fresh DMEM medium with 10% FBS.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was prepared from tissues and all five cell lines using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocols. To analyze miR-433 expression, total RNA was reversed transcribed to complementary DNA (cDNA) using a TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. qPCR was performed with a TaqMan MicroRNA PCR kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) on a Bio-Rad CFX96 Real-Time PCR machine (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The thermocycling conditions used for qPCR 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 60 sec. For the detection of E2F3 mRNA expression levels, cDNA was synthesized from total RNA using a M-MLV cDNA Reverse Transcription kit (Invitrogen; Thermo Fisher Scientific, Inc.). Subsequently, qPCR was performed using a SYBR® Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd., Dalian, China). The thermocycling conditions used for qPCR were as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. Relative miR-433 and E2F3 mRNA expression was normalized to U6 small nuclear RNA (U6) and GAPDH, respectively. Each sample was performed in triplicate and analyzed with the 2−ΔΔCq method (25). The primers were designed as follows: miR-433 forward, 5′-GGATCATGATGGGCTCCT-3′ and reverse, 5′-CAGTGCGTGTCGTGGAGT-3′; U6 forward, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ and reverse, 5′-CGCTTCACGAATTTGCGTGTCAT-3′; E2F3 forward, 5′-GATGGGGTCAGATGGAGAGA-3′ and reverse, 5′-GAGACACCCTGGCATTGTTT-3′; and GAPDH forward, 5′-CAAGGTCATCCATGACAACTTTG-3′ and reverse, 5′-GTCCACCACCCTGTTGCTGTAG-3′.

Cell counting kit-8 (CCK-8) assay

The effect of miR-433 on the proliferative ability of A549 and H460 cells was determined using a CCK-8 assay. After 24 h post-transfection, 3,000 transfected cells were plated onto 96-well cell culture plates and cultured for 0, 24, 48 or 72 h. At each time point, a total of 10 µl CCK-8 solution (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was added into each well, and then the cells were incubated at 37°C with 5% CO2 for another 2 h. Finally, the optical density value was detected at a wavelength of 450 nm using a microplate reader (Bio-Rad, Laboratories, Inc.).

Cell invasion assay

Matrigel (BD Biosciences, San Jose, CA, USA) coated Transwell plates with 8 µm pore polycarbonate membranes (BD Biosciences) were employed to assess the invasive capacity of A549 and H460 cells. A total of 1×105 transfected cells in FBS-free DMEM were seeded into the upper chambers, and 600 µl DMEM supplemented with 20% FBS was added to the lower chambers as a chemoattractant. Following incubation at 37°C with 5% CO2 for 24 h, the cells remaining on the upper side of the polycarbonate membranes were wiped with cotton swabs. The invasive cells were fixed with 100% methanol at room temperature for 20 min and stained with 0.5% crystal violet solution (Beyotime Institute of Biotechnology, Shanghai, China). Finally, the stained cells were photographed and counted under a light microscope (magnification, ×200) using five randomly selected fields per membrane.

Bioinformatics analysis

TargetScan 7.1 (http://www.targetscan.org/) and miRanda (http://www.microrna.org/) were applied to predict the potential targets of miR-433.

Luciferase reporter assay

The wild type and mutant sequences containing the predicted target sites of miR-433 in the 3′-UTR of E2F3 mRNA were synthesised by Shanghai GenePharma Co., Ltd., (Shanghai, China), cloned into the pMIR-REPORT luciferase reporter plasmids (Promega Corporation, Madison, WI, USA) and named as pMIR-Wt-E2F3-3′-UTR and pMIR-Mut-E2F3-3′-UTR, respectively. Cells were plated into 24-well cell culture plates at a density of 60–70% confluence. MiR-433 mimics or miR-NC with or without pMIR-Wt-E2F3-3′-UTR or pMIR-Mut-E2F3-3′-UTR, were transfected into A549 and H460 cells using Lipofectamine® 2000 according to the manufacturer's protocols. A pRL-TK plasmid with a constitutive expression of Renilla luciferase (Promega Corporation) was also transfected into A549 and H460 cells to serve as a negative control. Following transfection for 48 h, the transfected cells were collected and the relative luciferase activity was measured using a Dual-Luciferase® Reporter Assay System (Promega Corporation) according to the manufacturer's protocols. The activity of firefly luciferase was normalised to that of Renilla luciferase.

Western blot analysis

The primary antibodies used in the present study were acquired from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA) and included mouse anti-human monoclonal E2F3 (1:1,000; cat. no. sc-28308) and mouse anti-human monoclonal GAPDH (1:1,000; cat. no. sc-365062) antibodies. A total of 72 h post-transfection, A549 and H460 cells were harvested. Total protein isolated from tissues and A549 and H460 cells following transfection was isolated using a radioimmunoprecipitation assay lysis buffer (Nanjing KeyGen Biotech Co., Ltd. Nanjing, China). The concentration of total protein was detected using a Bicinchoninic Acid Protein Assay kit (Nanjing KeyGen Biotech Co., Ltd.). Equal amounts of protein (30 µg) was separated via 10% SDS-PAGE and transferred onto polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were then blocked with 5% non-fat milk in Tris-buffered saline containing 0.1% Tween-20 (TBST) at room temperature for 1 h, washed with TBST three times and incubated with primary antibodies overnight at 4°C. Subsequent to washing with TBST three times, the membranes were further probed with goat anti-mouse IgG horseradish peroxidase-conjugated secondary antibodies (1:5,000; cat. no. sc-2005; Santa Cruz Biotechnology, Inc.) at room temperature for 1 h. Finally, the protein signals were visualized using an ECL™ Western Blotting Detection Reagents kit (GE Healthcare, Chicago, IL, USA), and analyzed with Quantity One software (version 4.62; Bio-Rad Laboratories, Inc.). GAPDH served as a loading control.

Statistical analysis

Data were expressed as the mean ± standard deviation of at least 3 independent experiments and analysed using a statistical software package (SPSS 19.0, IBM Corp., Armonk, NY, USA). Differences between groups were compared with Student's t-test or one-way analysis of variance for multiple comparisons, combined with post hoc analysis (Student-Newman-Keuls test). The association between miR-433 and clinicopathological features in NSCLC was evaluated by chi-square test. P<0.05 was considered to indicate a statistically significant difference.

Results

MiR-433 is underexpressed in NSCLC tissues and cell lines

To evaluate the expression pattern of miR-433 in NSCLC, total RNA from 47 paired NSCLC tissues and adjacent non-tumor lung tissues was obtained. The data of RT-qPCR revealed that miR-433 was significantly underexpressed in the NSCLC tissues relative to that in the adjacent non-tumor lung tissues (Fig. 1A; P<0.05). After confirming the downregulation of miR-433 in NSCLC, the association between the miR-433 expression levels and clinicopathological data in the patients with NSCLC was investigated. On the basis of the miR-433 median level, patients with NSCLC were separated into two groups as follows: MiR-433 low-expression group (n=24) and miR-433 high-expression group (n=23). Decreased miR-433 expression levels were significantly associated with the tumor-node-metastasis (TNM) stage (P=0.006) and lymph node metastasis (P=0.028) but not associated with the other clinicopathological factors in NSCLC (Table I). Lastly, the miR-433 expression levels in four NSCLC cell lines and a non-tumorigenic bronchial epithelium BEAS-2B cell line were analysed. Compared with in BEAS-2B cells, the expression levels of miR-433 were lower in all the examined NSCLC cell lines (Fig. 1B; P<0.05). These results suggested that the downregulation of miR-433 may be associated with the progression of NSCLC.

Table I.

Association between miR-433 expression and clinicopathological factors of patients with non-small-cell lung cancer.

Table I.

Association between miR-433 expression and clinicopathological factors of patients with non-small-cell lung cancer.

miR-433 expression

Clinicopathological factorsCasesLowHighP-value
Sex 0.181
  Male261115
  Female2113  8
Age 0.676
  <55 years19  910
  ≥55 years281513
Tumor size 0.376
  <5 cm19  811
  ≥5 cm281612
Smoking history 0.423
  <10 years1710  7
  ≥10 years301416
Tumor differentiation 0.917
  I–II16  8  8
  III–IV311615
Tumor node metastasis stage 0.006a
  I–II21  615
  III–IV2618  8
Lymph node metastasis 0.028a
  Negative25  916
  Positive2215  7

a P<0.05.

MiR-433 overexpression attenuates cell proliferation and invasion of NSCLC

To determine the effects of miR-433 on the progression of NSCLC, miR-433 mimics were transfected into A549 and H460 cells, which were revealed to express relatively low miR-433 levels among the four NSCLC cell lines. RT-qPCR analysis revealed that miR-433 was significantly overexpressed in A549 and H460 cells following transfection with miR-433 mimics compared with corresponding miR-NC-transfected cells (Fig. 2A; P<0.05). Subsequently, CCK-8 and cell invasion assays were employed to examine the effects of miR-433 overexpression on NSCLC cell proliferation and invasion, respectively. CCK-8 assay indicated that ectopic miR-433 expression significantly decreased A549 and H460 cell proliferation at both 48 and 72 h (Fig. 2B; P<0.05). As presented in Fig. 2C, the invasive capacities of A549 and H460 cells transfected with miR-433 mimics were significantly lower than cells transfected with miR-NC (P<0.05). These results suggested that miR-433 may exhibit a tumor suppressive role in NSCLC progression.

E2F3 is a direct target of miR-433 in NSCLC

To elucidate the mechanisms underlying the inhibitory effects of miR-433 in NSCLC cells, bioinformatics analysis was performed to predict the potential targets of miR-433. E2F3 (Fig. 3A) was predicted as a primary target of miR-433 and selected for investigation, to further verify its previously reported contribution to NSCLC formation and progression (2630). To confirm this hypothesis, luciferase reporter assays were performed using A549 and H460 cells cotransfected with miR-433 mimics or miR-NC and pMIR-Wt-E2F3-3′-UTR or pMIR-Mut-E2F3-3′-UTR. The results indicated that the ectopic expression of miR-433 significantly decreased the luciferase activities of the wild-type 3′-UTR of E2F3 compared with cells transfected with miR-NC (P<0.05), but did not affect the luciferase activities of the mutant 3′-UTR of E2F3 in the A549 and H460 cells (Fig. 3B). RT-qPCR and western blot analyses were performed to investigate whether miR-433 may exert regulatory effects on E2F3 expression in NSCLC cells. MiR-433 upregulation significantly suppressed E2F3 mRNA and protein expression levels in the A549 and H460 cells compared with miR-NC groups (Fig. 3C and D; P<0.05). The results of the present study indicated that E2F3 may be a direct target of miR-433 in NSCLC.

E2F3 knockdown exhibits similar inhibitory effects to miR-433 overexpression on NSCLC cells

E2F3 was confirmed as a direct target of miR-433 in NSCLC in the present study. Hence, the tumor suppressive role of miR-433 in NSCLC cells may be induced by the downregulation of E2F3. To test this hypothesis, A549 and H460 cells were transfected with E2F3 siRNA to significantly knockdown the endogenous E2F3 expression levels compared with in cells transfected NC siRNA. The results were further confirmed by western blot analysis (Fig. 4A; P<0.05). Functional experiments demonstrated that E2F3 knockdown significantly reduced the proliferation (Fig. 4B; P<0.05) and invasion (Fig. 4C; P<0.05) of the A549 and H460 cells. These effects were similar to those observed with miR-433 overexpression. Hence, miR-433 may have prohibited the proliferation and invasion in NSCLC, at least partly, by E2F3 downregulation.

Restored E2F3 expression counteracts the suppressive effects of miR-433 overexpression on NSCLC cells

Rescue experiments were performed to determine whether the tumor-suppressing roles of miR-433 in NSCLC cells were mediated by E2F3. A549 and H460 cells were cotransfected with miR-433 mimics and E2F3 overexpression plasmid pcDNA3.1-E2F3 or blank pcDNA3.1 plasmid. Western blot analysis indicated that the cotransfection of pcDNA3.1-E2F3 significantly increased the level of E2F3 protein expression in A549 and H460 cells compared with cells transfected with miR-433 mimics and blank pcDNA3.1 plasmids (Fig. 5A; P<0.05). Subsequent functional assays revealed that the cotransfection of pcDNA3.1-E2F3 significantly decreased the proliferation (Fig. 5B; P<0.05) and invasion (Fig. 5C; P<0.05) of A549 and H460 cells compared with cells transfected with miR-433 mimics and blank pcDNA3.1 plasmids. Thus, the results of the present study suggested that the tumor suppressive roles of miR-433 on NSCLC cells may depend, at least in part, on the inhibition of E2F3 expression.

Discussion

MiRNAs have been associated with the initiation and progression of NSCLC (3133). As such, a comprehensive understanding of the association between dysregulated miRNA expression and NSCLC may contribute to the identification of novel therapeutic methods for patients with this disease. In the present study, miR-433 was significantly downregulated in the NSCLC tissues and cell lines; low miR-433 expression levels were significantly associated with TNM stage and lymph node metastasis. In addition, the resumption of miR-433 expression attenuated the proliferation and invasion of NSCLC cells. E2F3 was also identified as a direct target of miR-433 in NSCLC. E2F3 knockdown may mimic the inhibitory roles of miR-433 overexpression in NSCLC cell proliferation and invasion; however, restored E2F3 expression rescued NSCLC cells of the suppressive effects exhibited by miR-433 overexpression. The findings of the present study suggested that miR-433 may be considered as a potential therapeutic target for the treatment of NSCLC.

MiR-433 dysregulation is involved in numerous types of human cancer (2123). For example, miR-433 is downregulated within gastric cancer tissues and cell lines (21). Decreased miR-433 expression levels are correlated with distant metastasis and pathological TNM stage in patients with gastric cancer (21). MiR-433 downregulation has also been reported in colorectal cancer (22), hepatocellular carcinoma (23,24), myeloproliferative neoplasms (34), oral squamous cell carcinoma (35), ovarian cancer (36), retinoblastoma (37) and glioma (38). Conversely, miR-433 is overexpressed in osteosarcoma (39). These conflicting findings suggest that the expression pattern of miR-433 in human malignancies exhibits tissue specificity; miR-433 may be considered as a marker for the diagnosis of certain tumors.

MiR-433 is closely associated with numerous malignant human cancer phenotypes. For instance, ectopic miR-433 expression notably decreases the rate of gastric cancer cell growth, metastasis and cell cycle progression (21). Li et al (22) reported that miR-433 overexpression negatively regulates cell viability and promotes apoptosis in colorectal cancer. Xue et al (23) and Yang et al (24) demonstrated that the upregulation of miR-433 inhibits the proliferation and invasion of hepatocellular carcinoma. Lin et al (34) revealed that miR-433 reduces the hematopoietic cell growth and differentiation in myeloproliferative neoplasms. Wang et al (35) reported that the induction of miR-433 attenuates cell growth, migration and invasion in oral squamous cell carcinoma. Liang et al (36) demonstrated that the enforced expression of miR-433 considerably inhibits ovarian cancer cell migration and invasion. Li et al (37) indicated that miR-433 overexpression notably suppresses cell growth and metastasis and promotes cell cycle arrest and apoptosis in retinoblastoma. In addition, Sun et al (38) reported that restoring miR-433 expression prohibits cell proliferation and motility in vitro, induces apoptosis in vitro, reduces tumor growth in vivo and increases the chemosensitivity of cells to temozolomide in vitro and in vivo. However, miR-433 has been identified as an oncogene in osteosarcoma by regulating cell apoptosis and growth both in vitro and in vivo (39). These conflicting findings indicated the tissue specificity of the biological roles of miR-433 in tumor occurrence and development, suggesting that miR-433 may be investigated as a potential anticancer drug for particular types of cancer.

Numerous miR-433 targets, including Kirsten murine sarcoma virus 2 (21), mitogen-activated protein kinase 4 (21) in gastric cancer, metastasis-associated in colon cancer protein 1 (22) in colorectal cancer, p21 protein-activated kinase 4 (23) and cyclic adenosine 5′-phosphate responsive element binding protein (CREB) 1 (24) in hepatocellular carcinoma, guanylate binding protein 2 (34) in myeloproliferative neoplasms, histone deacetylase 6 (35) in oral squamous cell carcinoma, Notch1 (36) in ovarian cancer, Notch1 (37) and paired box 6 (37) in retinoblastoma and CREB (38) in glioma, have been previously identified. In the present study, E2F3 was validated as a direct target of miR-433 in NSCLC. The transcription factor E2F3, a key regulator of the G1/S phase transition, has been reported to be upregulated in numerous types of human cancer, including bladder (40), gastric (41), colorectal (42) and breast cancers (43). E2F3 activation serves important roles in carcinogenesis and progression via the regulation of cell cycle, apoptosis, differentiation, migration and invasion (4446). E2F3 is upregulated in NSCLC tissues, cell lines and serum (26,27) and contributes to the regulation of NSCLC initiation and progression (2830). Hence, targeting E2F3 may provide novel and promising therapies for this aggressive cancer in particular.

In conclusion, miR-433 was downregulated in NSCLC tissues and cell lines, and this dysregulation was associated with the TNM stage and lymph node metastasis. Functional experiments also demonstrated that miR-433 overexpression repressed cell proliferation and invasion in NSCLC; E2F3 was verified to be a direct target of miR-433 in NSCLC. Collectively, the results of the present study may improve the understanding of the mechanisms of miR-433 in regulating the progression of NSCLC. The present study also suggested that miR-433 may potentially serve as a therapeutic target for the treatment of patients with this malignancy.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

NL and ZL designed the present study. NL and WZ performed reverse transcription-quantitative polymerase chain reaction, Cell Counting kit-8 assays and cell invasion assays. YL and JC performed western blot analyses. HY and XL performed luciferase reporter assays and analyzed the data in the present study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Hunan Provincial People's Hospital (Changsha, China), and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of Hunan Provincial People's Hospital (Changsha, China). Written informed consent was obtained from all patients for the use of their clinical tissues.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. 2010. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Zarogoulidis K, Zarogoulidis P, Darwiche K, Boutsikou E, Machairiotis N, Tsakiridis K, Katsikogiannis N, Kougioumtzi I, Karapantzos I, Huang H and Spyratos D: Treatment of non-small cell lung cancer (NSCLC). J Thorac Dis. 5 Suppl 4:S389–S396. 2013.PubMed/NCBI

4 

Li X, Shi Y, Yin Z, Xue X and Zhou B: An eight-miRNA signature as a potential biomarker for predicting survival in lung adenocarcinoma. J Transl Med. 12:1592014. View Article : Google Scholar : PubMed/NCBI

5 

Naito Y, Kubota K, Nihei K, Fujii T, Yoh K, Niho S, Goto K, Ohmatsu H, Saijo N and Nishiwaki Y: Concurrent chemoradiotherapy with cisplatin and vinorelbine for stage III non-small cell lung cancer. J Thorac Oncol. 3:617–622. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Paz-Ares L, Soulières D, Melezínek I, Moecks J, Keil L, Mok T, Rosell R and Klughammer B: Clinical outcomes in non-small-cell lung cancer patients with EGFR mutations: Pooled analysis. J Cell Mol Med. 14:51–69. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Fassina A, Cappellesso R and Fassan M: Classification of non-small cell lung carcinoma in transthoracic needle specimens using microRNA expression profiling. Chest. 140:1305–1311. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Simon J: Technology radiation technology targets tumors. Surgical precision without the incision. S D Med. 67:3622014.PubMed/NCBI

9 

Johtatsu T, Noguchi S, Yatera K, Shinohara S, Oka S, Yamasaki K, Nishida C, Kawanami T, Kawanami Y, Ishimoto H, et al: A case of lung adenocarcinoma with uncontrollable myocardial metastasis and pericardial effusion. J UOEH. 36:199–203. 2014.(In Japanese). View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Wilson RC and Doudna JA: Molecular mechanisms of RNA interference. Annu Rev Biophys. 42:217–239. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Berezikov E, van Tetering G, Verheul M, van de Belt J, van Laake L, Vos J, Verloop R, van de Wetering M, Guryev V, Takada S, et al: Many novel mammalian microRNA candidates identified by extensive cloning and RAKE analysis. Genome Res. 16:1289–1298. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Fadejeva I, Olschewski H and Hrzenjak A: MicroRNAs as regulators of cisplatin-resistance in non-small cell lung carcinomas. Oncotarget. 8:115754–115773. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Lü L, Mao X, Shi P, He B, Xu K, Zhang S and Wang J: MicroRNAs in the prognosis of triple-negative breast cancer: A systematic review and meta-analysis. Medicine (Baltimore). 96:e70852017. View Article : Google Scholar : PubMed/NCBI

16 

Ahir BK, Ozer H, Engelhard HH and Lakka SS: MicroRNAs in glioblastoma pathogenesis and therapy: A comprehensive review. Crit Rev Oncol Hematol. 120:22–33. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Cho WC: MicroRNAs: Potential biomarkers for cancer diagnosis, prognosis and targets for therapy. Int J Biochem Cell Biol. 42:1273–1281. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Cortés-Sempere M and de Cáceres Ibáñez I: microRNAs as novel epigenetic biomarkers for human cancer. Clin Transl Oncol. 13:357–362. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Calin GA and Croce CM: MicroRNA-cancer connection: The beginning of a new tale. Cancer Res. 66:7390–7394. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:2257–2261. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Guo LH, Li H, Wang F, Yu J and He JS: The tumor suppressor roles of miR-433 and miR-127 in gastric cancer. Int J Mol Sci. 14:14171–14184. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Li J, Mao X, Wang X, Miao G and Li J: miR-433 reduces cell viability and promotes cell apoptosis by regulating MACC1 in colorectal cancer. Oncol Lett. 13:81–88. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Xue J, Chen LZ, Li ZZ, Hu YY, Yan SP and Liu LY: MicroRNA-433 inhibits cell proliferation in hepatocellular carcinoma by targeting p21 activated kinase (PAK4). Mol Cell Biochem. 399:77–86. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Yang Z, Tsuchiya H, Zhang Y, Hartnett ME and Wang L: MicroRNA-433 inhibits liver cancer cell migration by repressing the protein expression and function of cAMP response element-binding protein. J Biol Chem. 288:28893–28899. 2013. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Al Ahmed HA and Nada O: E2F3 transcription factor: A promising biomarker in lung cancer. Cancer Biomark. 19:21–26. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Cooper CS, Nicholson AG, Foster C, Dodson A, Edwards S, Fletcher A, Roe T, Clark J, Joshi A, Norman A, et al: Nuclear overexpression of the E2F3 transcription factor in human lung cancer. Lung Cancer. 54:155–162. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Ren J, Ding L, Xu Q, Shi G, Li X, Li X, Ji J, Zhang D, Wang Y, Wang T and Hou Y: LF-MF inhibits iron metabolism and suppresses lung cancer through activation of P53-miR-34a-E2F1/E2F3 pathway. Sci Rep. 7:7492017. View Article : Google Scholar : PubMed/NCBI

29 

Zhang J, Li Y, Dong M and Wu D: Long non-coding RNA NEAT1 regulates E2F3 expression by competitively binding to miR-377 in non-small cell lung cancer. Oncol Lett. 14:4983–4988. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Trikha P, Sharma N, Pena C, Reyes A, Pécot T, Khurshid S, Rawahneh M, Moffitt J, Stephens JA, Fernandez SA, et al: E2f3 in tumor macrophages promotes lung metastasis. Oncogene. 35:3636–3646. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Zhao D, Han W, Liu X, Cui D and Chen Y: MicroRNA-128 promotes apoptosis in lung cancer by directly targeting NIMA-related kinase 2. Thorac Cancer. 8:304–311. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Zhao X, Lu C, Chu W, Zhang B, Zhen Q, Wang R, Zhang Y, Li Z, Lv B, Li H and Liu J: MicroRNA-124 suppresses proliferation and glycolysis in non-small cell lung cancer cells by targeting AKT-GLUT1/HKII. Tumour Biol. 39:10104283177062152017. View Article : Google Scholar : PubMed/NCBI

33 

Castro D, Moreira M, Gouveia AM, Pozza DH and De Mello RA: MicroRNAs in lung cancer. Oncotarget. 8:81679–81685. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Lin X, Rice KL, Buzzai M, Hexner E, Costa FF, Kilpivaara O, Mullally A, Soares MB, Ebert BL, Levine R and Licht JD: miR-433 is aberrantly expressed in myeloproliferative neoplasms and suppresses hematopoietic cell growth and differentiation. Leukemia. 27:344–352. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Wang XC, Ma Y, Meng PS, Han JL, Yu HY and Bi LJ: miR-433 inhibits oral squamous cell carcinoma (OSCC) cell growth and metastasis by targeting HDAC6. Oral Oncol. 51:674–682. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Liang T, Guo Q, Li L, Cheng Y, Ren C and Zhang G: MicroRNA-433 inhibits migration and invasion of ovarian cancer cells via targeting Notch1. Neoplasma. 63:696–704. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Li X, Yang L, Shuai T, Piao T and Wang R: MiR-433 inhibits retinoblastoma malignancy by suppressing Notch1 and PAX6 expression. Biomed Pharmacother. 82:247–255. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Sun S, Wang X, Xu X, Di H, Du J, Xu B, Wang Q and Wang J: MiR-433-3p suppresses cell growth and enhances chemosensitivity by targeting CREB in human glioma. Oncotarget. 8:5057–5068. 2017.PubMed/NCBI

39 

Sun Y, Wang F, Wang L, Jiao Z, Fang J and Li J: MicroRNA-433 regulates apoptosis by targeting PDCD4 in human osteosarcoma cells. Oncol Lett. 14:2353–2358. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Olsson AY, Feber A, Edwards S, Te Poele R, Giddings I, Merson S and Cooper CS: Role of E2F3 expression in modulating cellular proliferation rate in human bladder and prostate cancer cells. Oncogene. 26:1028–1037. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Guo Y, Qi Y, Guo A, Du C, Zhang R and Chu X: miR-564 is downregulated in gastric carcinoma and targets E2F3. Oncol Lett. 13:4155–4160. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Chang SW, Yue J, Wang BC and Zhang XL: miR-503 inhibits cell proliferation and induces apoptosis in colorectal cancer cells by targeting E2F3. Int J Clin Exp Pathol. 8:12853–12860. 2015.PubMed/NCBI

43 

Lee M, Oprea-Ilies G and Saavedra HI: Silencing of E2F3 suppresses tumor growth of Her2+ breast cancer cells by restricting mitosis. Oncotarget. 6:37316–37334. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Wang JP, Jiao Y, Wang CY, Xu ZB and Zhang B: Rb knockdown accelerates bladder cancer progression through E2F3 activation. Int J Oncol. 50:149–160. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Oeggerli M, Tomovska S, Schraml P, Calvano-Forte D, Schafroth S, Simon R, Gasser T, Mihatsch MJ and Sauter G: E2F3 amplification and overexpression is associated with invasive tumor growth and rapid tumor cell proliferation in urinary bladder cancer. Oncogene. 23:5616–5623. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Dong D, Gong Y, Zhang D, Bao H and Gu G: miR-874 suppresses the proliferation and metastasis of osteosarcoma by targeting E2F3. Tumour Biol. 37:6447–6455. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 18 Issue 1

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 N, Liu Z, Zhang W, Li Y, Cao J, Yang H and Li X: MicroRNA‑433 reduces cell proliferation and invasion in non‑small cell lung cancer via directly targeting E2F transcription factor 3 Retraction in /10.3892/mmr.2022.12857. Mol Med Rep 18: 1155-1164, 2018
APA
Liu, N., Liu, Z., Zhang, W., Li, Y., Cao, J., Yang, H., & Li, X. (2018). MicroRNA‑433 reduces cell proliferation and invasion in non‑small cell lung cancer via directly targeting E2F transcription factor 3 Retraction in /10.3892/mmr.2022.12857. Molecular Medicine Reports, 18, 1155-1164. https://doi.org/10.3892/mmr.2018.9020
MLA
Liu, N., Liu, Z., Zhang, W., Li, Y., Cao, J., Yang, H., Li, X."MicroRNA‑433 reduces cell proliferation and invasion in non‑small cell lung cancer via directly targeting E2F transcription factor 3 Retraction in /10.3892/mmr.2022.12857". Molecular Medicine Reports 18.1 (2018): 1155-1164.
Chicago
Liu, N., Liu, Z., Zhang, W., Li, Y., Cao, J., Yang, H., Li, X."MicroRNA‑433 reduces cell proliferation and invasion in non‑small cell lung cancer via directly targeting E2F transcription factor 3 Retraction in /10.3892/mmr.2022.12857". Molecular Medicine Reports 18, no. 1 (2018): 1155-1164. https://doi.org/10.3892/mmr.2018.9020