MicroRNA‑107 may regulate lung cancer cell proliferation and apoptosis by targeting TP53 regulated inhibitor of apoptosis 1

  • Authors:
    • Peng Cai
    • Jingjing Li
    • Guiming Chen
    • Bing Peng
    • Liuyang Yu
    • Bolin Zhao
    • Yi Yu
  • View Affiliations

  • Published online on: January 7, 2020     https://doi.org/10.3892/ol.2020.11248
  • Pages: 1958-1966
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer causes over 1.6 million mortalities worldwide annually. MicroRNAs (miRs) are involved in various types of cancer‑associated processes. The present study investigated the possible mechanism of miR‑107 in the development of lung cancer in order to identify novel targets for clinical treatment. The expression levels of miR‑107 and its putative target gene TP53 regulated inhibitor of apoptosis 1 (TRIAP1) were measured in lung cancer tumor tissues and non‑tumor adjacent tissues. Subsequently, the association between TRIAP1 and miR‑107 was investigated using a dual‑luciferase reporter assay. Following transfection, the effects of miR‑107 and TRIAP1 on the proliferation and apoptosis of lung cancer cell lines in vitro were investigated using Cell Counting Kit‑8 and flow cytometry assays, respectively. Furthermore, the regulatory effect of miR‑107 on the expression levels of TRIAP1 and associated proteins was analyzed using a western blot assay. The results revealed lower expression levels of miR‑107 and higher expression levels of TRIAP1 in lung cancer tumor tissues compared with non‑tumor adjacent tissues. The dual‑luciferase reporter assay demonstrated that TRIAP1 is a target gene of miR‑107. Additionally, the results revealed that overexpression of miR‑107 resulted in a lower proliferation rate and higher apoptosis rate of A549 cells, compared with the negative control (NC) and control groups (P<0.01). The variation of cell proliferation and apoptosis induced by miR‑107 mimics was reversed by co‑transfection with pcDNA3.1‑TRIAP1. Furthermore, the expression levels of cyclin D1 and proliferating cell nuclear antigen were markedly decreased in the miR‑107 mimics group compared with the NC group (P<0.01). The expression levels of BCL2 associated X apoptosis regulator, tumor protein p53 and caspase 3 were upregulated and the expression levels of TRIAP1 and BCL2 apoptosis regulator were significantly reduced in the miR‑107 mimics group compared with the NC group (P<0.01). The results of the present study suggested that miR‑107 regulates lung cancer cell proliferation and apoptosis by targeting TRIAP1.

Introduction

Lung cancer is a type of malignant tumor with unregulated and rapid proliferation that resulted in >1.6 million deaths worldwide in 2015 (1). Despite advances in clinical therapeutic options, the 5-year survival rate of patients with lung cancer remains <15% (2), which is markedly lower than that of patients with breast, colon or prostate cancer (3). Furthermore, advances in treatments for lung cancer, including surgical excision, medical treatment or radiotherapeutic intervention, have little effect on the long-term survival rate (4). Therefore, the identification of the underlying mechanisms of lung cancer tumorigenesis and progression may aid clinical diagnosis and treatment. Lung cancer tumorigenesis and development are closely associated with dysregulation of microRNAs (miRNAs/miRs) (5,6).

miRNAs are small non-coding RNAs consisting of 19–25 nucleotides (7), which modulate gene expression during cellular processes (8). An increasing number of studies suggest that miRNAs act as either tumor suppressors (9) or oncogenes (10) in the progression of various types of cancer, including lung cancer (11,12). A previous study revealed that c-Myc-activated long non-coding RNA H19 downregulated miR-107 and promoted the cell cycle progression of non-small-cell lung carcinoma (NSCLC) cell lines (13). Another study revealed that the expression of miR-107 was markedly reduced in pathological tissues obtained from patients with lung cancer (14). Furthermore, Takahashi et al (15) reported that the expression levels of miR-107 were decreased in lung tumor tissues compared with healthy tissues and that miR-107 induced cell cycle arrest in NSCLC cell lines in vitro. However, the underlying mechanism by which miR-107 functions in lung cancer progression and development remains largely unknown.

In the current study, the mechanism of miR-107 and its target gene TP53 regulated inhibitor of apoptosis 1 (TRIAP1) in lung cancer was investigated. The results obtained revealed that miR-107 decreased cell proliferation and induced cell apoptosis of lung cancer cell lines in vitro, providing a novel theoretical basis for the treatment of lung cancer.

Materials and methods

Specimens

A total of 45 pairs of lung cancer tumor tissues and non-tumor adjacent lung tissues were obtained from Jingmen No. 2 People's Hospital (Jingmen, China) between July 2014 and April 2016. Among the patients that the tissues were obtained from, there were 31 males and 14 females, and the average age was 62.3±6.8 years. All patients had been diagnosed with lung cancer and had not undergone any other therapy. The present study was approved by the Ethics Committee of Jingmen No. 2 People's Hospital and written consent was acquired from each patient.

Cell culture

The A549 human NSCLC cell line (cat no. SCSP-503; Type Culture Collection of the Chinese Academy of Sciences), BESA-2B cell line (cat no. CL-0496; Procell Life Science & Technology Co., Ltd.) and 293 cell line (cat no. GNHu43; Type Culture Collection of the Chinese Academy of Sciences) were cultured in RPMI-1640 (cat no. 11875093; Gibco; Thermo Fisher Scientific, Inc.) with 10% fetal bovine serum (cat no. 10099-141; Invitrogen; Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin. The cells were maintained in a humid atmosphere with 5% CO2 at 37°C.

Transfection efficiency

In order to determine the transfection efficiency, cells were divided into three groups as follows: i) Control group (untreated group); ii) microRNA-negative control (NC) mimics group (to eliminate non-sequence-specific effects); and iii) miR-107 mimics group (transfected with miR-107 mimics). The miR-107 mimics and NC oligonucleotides were purchased from Shanghai GenePharma Co., Ltd. The cells were seeded into 6-well plates at a density of 5×105 cells/well, and 50 nM miR-107 mimics or NC mimics were transfected into the cells using Lipofectamine® 2000 (cat no. 11668019; Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Following transfection, cells were cultured for another 24 h before RT-qPCR assays were conducted. The primer sequences were as follows: miR-107 mimics forward, 5′-ATACCGCTCGAGTGCCATGTGTCCACTGAAT-3′; miR-107 mimics reverse, 5′-ATACCGCTCGAGTTCCATGCCTCAACTCCT-3′; miR-NC forward, 5′-ATAGCACAGCCTGGATAGCAACGTAC-3′; miR-NC reverse, 5′-CACCTTCTACAATGAGCTGCGTGTG-3′.

Dual-luciferase reporter assay

TRIAP1 was considered to be a predictive target gene of miR-107 by TargetScan online tool (www.targetscan.org/vert_72). Subsequently, the 293 cell line was transfected with the wild-type TRIAP1 3′untranslated region [UTR; (TRIAP1-3′UTR)] or mutant TRIAP1-3′UTR with either NC mimics or miR-107 mimics using Lipofectamine® RNAiMax Transfection reagent (cat no. 13778075; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Following transfection, the cells were incubated for 4 h in 5% CO2 at 37°C. Subsequently, the firefly and Renilla luciferase activities were detected using a dual-luciferase reporter kit (Beyotime Institute of Biotechnology). Firefly luciferase activity was normalized using Renilla luciferase activity.

Vector construction

In order to obtain pcDNA3.1-TRIAP1, a TRIAP1-expression vector was constructed. In brief, TRIAP1 cDNA was amplified by PCR (as will be described) from the cDNA of BEAS-2B cells (cat no. CL-0496; Procell Life Science & Technology Co., Ltd.). The TRIAP1 cDNA (50 nM) was subsequently inserted into pcDNA3.1 (cat no. VT8071; Yobio) to construct the pcDNA3.1-TRIAP1 expression vector. Subsequently, 50 nM pcDNA3.1 and pcDNA3.1-TRIAP1 plasmids were transfected into A549 cells using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.), following the manufacturer's protocol, and incubated at 37°C for 24 h at 5% CO2 for 2 h. The transfected cells were subsequently incubated at room temperature in 5% CO2 at 37°C for another 48 h, in order to determine cell proliferation and apoptosis. The sequences were described as follows: TRIAP1 forward, 5′-TATCTTGCAGGAACTGTGTGCTA-3′, and TRIAP1 reverse 5′-AATTTAGGTTCTTCCTCCACAGC-3′.

Analysis of cell proliferation

In order to investigate the effect of miR-107 and TRIAP1 on the proliferation of A549 cells, cells were divided into four different groups as follows: i) control group; ii) microRNA-NC mimics group; iii) miR-107 mimics group; iv) miR-107 mimics + pcDNA3.1-TRIAP1 group. Briefly, the transfected cells were seeded onto a 96-well plate at a density of 1×104 cells with 100 µl/well of fresh RPMI-1640 medium. The cells were incubated in a humid atmosphere with 5% CO2 at 37°C for 48 h. Following 12, 24 or 48 h, 10 µl Cell Counting Kit-8 (CCK-8) solution (cat no. HY-P0093; MedChemExpress) was added to each well and the cells were incubated for a further hour at room temperature. The absorbance in each well was measured at a wavelength of 490 nm in order to quantify the proliferation.

Analysis of cell apoptosis

In order to investigate the effect of miR-107 and TRIAP1 on the apoptosis of A549 cells, cells were divided into four different groups: i) Control group; ii) microRNA-NC mimics group; iii) miR-107 mimics group; iv) miR-107 mimics + pcDNA3.1-TRIAP1 group. Briefly, the transfected cells in suspension were collected at a density of 1×106 cells/ml. The cells were washed with HEPES buffer solution (cat no. ACC0013A; Seebio Biotech, Inc.) for 5 min at room temperature and centrifuged for 5 min at 5,000 × g on ice. Subsequently, 5 µl Annexin V-fluorescein isothiocyanate (FITC) and 10 µl propidium iodide (BD Biosciences) were added to the cells of the four different groups and the cells were incubated for 10 min in the dark at room temperature. The apoptosis rate was then measured using a flow cytometer (BD Biosciences) and analyzed using FlowJo software (version 10; BD Biosciences).

RNA extraction and reverse transcription-quantitative (RT-q) PCR analysis

miRNA was isolated from the lung tumor and adjacent non-tumor tissues and A549 cells using the miRNeasy Mini kit (Qiagen, Inc.) according to the manufacturer's protocol. Following isolation, the One-Step PrimeScript miRNA cDNA Synthesis kit (Takara Biotechnology Co., Ltd.) was used to synthesize cDNA according to the manufacturer's instructions. The total RNA was extracted using TRIzol reagent (cat. no. 15596018; Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Subsequently, 2 µl RNA was reverse transcribed into cDNA. qPCR analysis was performed using the TaqMan MicroRNA RT kit (cat no. 4366596; Applied Biosystems; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Briefly, 2 µl cDNA, 10 µl SYBRGreen RT-qPCR Master mix (cat. no. AB4106A; Applied Biosystems; Thermo Fisher Scientific, Inc.), 1 µl primers and nuclease-free water were combined during the PCR reaction. The primer sequences were as follows: miR-107 forward, 5′-AGCAGCATTGTACAGGGCTATCA-3′; and reverse, 5′-GCGAGCACAGAATTAATACGAC-3′; U6 forward, 5′-AGAGCCTGTGGTGTCCG-3′; and reverse, 5′-CATCTTCAAAGCACTTCCCT-3′; TRIAP1 forward, 5′-AGGATTTCGCAAGTCCAGAA-3′; and reverse, 5′-GCTGATTCCACCCAAGTAT-3′; and GAPDH forward, 5′-AACGGATTTGGTCGTATTG-3′; and reverse, 5′-GGAAGATGGTGATGGGATT-3′. The PCR reaction conditions were as follows: i) Denaturing for 3 min at 95°C; ii) denaturing for 30 sec at 94°C, annealing for 30 sec at 56°C and extension for 30 sec at 72°C (35 cycles); and iii) extension for 10 min at 72°C. The relative expression of miR-107 was normalized to that of U6 small nuclear RNA (Thermo Fisher Scientific, Inc.) and the expression levels of TRIAP1 mRNA were normalized to those of GAPDH according to the 2−ΔΔCq method (16). U6 or GAPDH was used as the endogenous control.

Western blot analysis

Radioimmunoprecipitation assay buffer (cat. no. R0010; Beijing Solarbio Science & Technology Co., Ltd.) was used to extract the proteins from tissues and cells. Following centrifugation at 6,000 × g for 5 min at 4°C, a protease inhibitor cocktail (cat. no. 78425; Thermo Fisher Scientific, Inc.) was added to the protein lysates. The Bradford method was used to quantify the concentration of proteins in the supernatant of the lysates. The whole protein lysates (20 µg) were separated by SDS-PAGE (8% gel; cat. no. LC26755; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Subsequently, 10 µg separated protein was transferred to a polyvinylidene fluoride membrane (Shanghai Ofluorine Chemical Technology Co., Ltd.) and the membrane was blocked with 50 ml 5% nonfat milk for 50 min at 37°C. The membrane was then incubated with the following primary antibodies at 37°C for 50 min: Rabbit anti-proliferating cell nuclear antigen (PCNA; 1:1,000; cat. no. ab18197; Abcam); rabbit anti-cyclin D1 (1:100; cat. no. ab16663; Abcam); rabbit anti-TRIAP1 (1:1,000; cat. no. ABIN2970840; 4A Biotechnology Co., Ltd.), rabbit anti-BCL2 apoptosis regulator (BCL2; 1:1,000; cat. no. ab32124; Abcam), rabbit anti-BCL2 associated X apoptosis regulator (BAX; 1:1,000; cat. no. ab32503; Abcam), rabbit anti-tumor protein p53 (TP53; 1:1,000; cat. no. ab131442; Abcam), rabbit anti-caspase 3 (1:500; cat. no. ab13847; Abcam) and rabbit anti-GAPDH (1:2,500; cat. no. ab9485; Abcam). All antibodies were diluted in blocking buffer (concentration, 10×; cat. no. ab126587; Abcam). After washing, the membrane was then incubated for 1 h at 37°C with goat anti-rabbit horseradish peroxidase (HRP) IgG H&L secondary antibody (1:1,000; cat. no. ab7090; Abcam). Subsequently, 200 µl chemiluminescent HRP substrate (cat. no. ab5801; Abcam) was added to the surface of the membranes. The signals were captured and exposed onto X-ray films. ImageJ software (version 1.49; National Institutes of Health) was used to quantify the relative expression levels of the proteins. GAPDH was used as the reference protein. The procedures above were conducted in triplicate.

Statistical analysis

SPSS (version 22.0; IBM SPSS) was used to analyze the results and the data are presented as the mean ± standard deviation. The Student's t-test was used to compare two groups. One-way analysis of variance followed by Newman-Keuls post hoc test was used to distinguish differences among three and more groups. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-107 is downregulated in lung cancer tissues

In order to investigate the putative effects of miR-107 in lung cancer, the expression levels of miR-107 in lung cancer tumor tissues and adjacent non-tumor tissues from 45 patients were compared. As shown in Fig. 1, the expression levels of miR-107 were significantly reduced in the lung cancer tumor tissues compared with the adjacent non-tumor tissues (P<0.01).

TRIAP1 is a direct target of miR-107 in lung cancer

A dual-luciferase reporter assay was conducted in order to determine the association between miR-107 and TRIAP1. The 3′-UTR of the TRIAP1 gene was confirmed to contain binding sequences for miR-107 (Fig. 2A), suggesting that TRIAP1 may be a downstream target of miR-107. Furthermore, the results demonstrated that transfection of miR-107 mimics markedly reduced the luciferase activity in the wild-type TRIAP1-3′UTR plasmid-transfected cells; however, miR-107 had no significant influence on the mutant TRIAP1-3′UTR plasmid-transfected cells (P<0.01; Fig. 2B).

Expression levels of TRIAP1 in lung cancer tumor tissues and adjacent non-tumor tissues

In order to further investigate the potential function of TRIAP1 in lung cancer, the mRNA and protein expression levels of TRIAP1 in lung cancer tumor tissues and adjacent non-tumor tissues were quantified. As shown in Fig. 3, the mRNA and protein expression levels of TRIAP1 were highly increased in the lung cancer tumor tissues compared with the adjacent non-tumor tissues (P<0.01).

miR-107 inhibits lung cancer cell proliferation

A549 cells were transfected with miR-107 mimics or NCs and the transfection efficiency was demonstrated using RT-qPCR analysis. As shown in Fig. 4A, there was no significant difference between the control group and NC group; however, compared with the NC group, the expression levels of miR-107 in the miR-107 mimics group were significantly increased (P<0.01). As demonstrated in Fig. 4B there was no significant difference in the proliferation of cells between the control group and the NC group throughout the whole experiment. As time increased, there was a significantly reduced proportion of proliferative cells in the miR-107 mimics group, in comparison with the control group (P<0.01). However, following transfection with pcDNA3.1 or pcDNA3.1-TRIAP1 (P<0.01; Fig. 5A), the miR-107 mimics-reduced A549 cell proliferation rate was reversed by co-transfection with pcDNA3.1-TRIAP1 (P<0.05; Fig. 5B).

miR-107 promotes lung cancer cell apoptosis

An Annexin V-FITC apoptosis kit was used to identify the apoptotic rate of A549 cells in order to investigate the effect of miR-107 on lung cancer cell apoptosis. The apoptotic rate of the cells in the miR-107 mimics group was almost four times higher than that of the cells in the control group or the NC group (P<0.01; Fig. 6A-D). There was no distinct difference in the apoptotic rate of the cells in the control group and the NC group. However, miR-107 mimics-induced A549 cell apoptosis was reduced by co-transfection with pcDNA3.1-TRIAP1 (P<0.01; Fig. 7A-C).

Effect of miR-107 on the expression levels of cyclin D1 and PCNA

In order to further demonstrate the effect of miR-107 on regulating A549 cell proliferation, the expression levels of cyclin D1 and PCNA were measured. As demonstrated in Fig. 8, the protein expression levels of cyclin D1 and PCNA were significantly decreased in the miR-107 mimics group, as compared with the NC group (P<0.01). However, there were no significant differences between the NC and control groups.

Effect of miR-107 on the protein expression of BCL2, BAX, TP53 and caspase 3

In order to further investigate the association between miR-107 and its target gene TRIAP1, a western blot assay was performed to examine the protein expression levels of TRIAP1 and related apoptotic proteins among the different groups. As shown in Fig. 9, the protein expression levels of TRIAP1 and BCL2 were significantly reduced and the expression levels of BAX, TP53 and caspase 3 were significantly increased in the miR-107 mimics group, compared with those in the NC group (P<0.01). The protein expression levels of BCL2, BAX, TP53 and caspase 3 in the control group and the NC group were similar.

Discussion

A previous study revealed that miR-107 promoted the proliferation of hepatocellular carcinoma cells by targeting axin 2 (17). Furthermore, miR-107 promoted the proliferation and invasion of gastric adenocarcinoma cells through large tumor suppressor kinase 2 (18). However, miR-107 inhibited cell proliferation and metastasis in gastric cancer (19) and osteosarcoma (20). Therefore, miR-107 acts as a tumor suppressor or oncogene in different types of cancer. The present study demonstrated that miR-107 may function as a tumor suppressor in lung cancer, which corresponds with previous studies (1720).

In the present study, the results of the CCK-8 and flow cytometry assays demonstrated that the overexpression of miR-107 reduced proliferation and promoted apoptosis in lung cancer cells in vitro. A previous study demonstrated that miR-107-5p suppressed NSCLC by directly targeting the oncogene epidermal growth factor receptor (21). Another study indicated that miR-107 inhibited tumor growth by targeting the brain derived neurotrophic factor-mediated PI3K/AKT signaling pathway in human NSCLC (14). The results of the present study were consistent with those of previous studies; however, a target gene of miR-107, which may provide novel approaches for clinical treatment, was also identified.

miRNAs regulate cell proliferation and apoptosis by targeting specific genes during cellular processes (22). According to previous studies, TRIAP1 was predicted to be a candidate oncogene in various types of cancer, including ovarian cancer (23), nasopharyngeal carcinoma (24) and lung cancer (25). In the present study, TRIAP1 was revealed to be upregulated in lung cancer tumor tissues compared with adjacent non-tumor tissue. TRIAP1 was subsequently predicted to be a target gene of miR-107 using TargetScan and a dual-luciferase reporter assay. The CCK-8 and flow cytometry assays further demonstrated that the effects on A549 cell proliferation and apoptosis induced by miR-107 mimics were reversed by co-transfection with pcDNA3.1-TRIAP1. Collectively, miR-107 may inhibit cell proliferation and promote cell apoptosis in lung cancer by targeting TRIAP1. However, the association between miR-107 and TRIAP1 requires further investigation.

In order to demonstrate the function of miR-107 on cell proliferation of lung cancer, the expression levels of the proliferation-associated factors, cyclin D1 and PCNA were measured via a western blot assay. Previous studies reported that inhibitory expression of cyclin D1 decreased lung cancer cell proliferation (26,27). Furthermore, the decreased expression of PCNA may inhibit lung cancer cell proliferation (28,29). In the present study, the expression levels of cyclin D1 and PCNA were significantly decreased in the miR-107 mimics group compared with the miR-NC mimics group. Thus, it was hypothesized that the overexpression of miR-107 may inhibit lung cancer cell proliferation by reducing the expression levels of cyclin D1 and PCNA.

In the current study, the expression levels of the apoptosis-associated factors BAX, TP53, caspase 3 and BCL2 were measured using a western blot assay. A previous study demonstrated that overexpression of caspase 3 may inhibit the apoptosis of lung cancer cells (30). Another study reported that activation of BAX contributed to the apoptosis of lung cancer cells (31). TP53 functions as a tumor suppressor and blocks cancer progression (32). Overexpression of miR-107 may increase the expression levels of caspase 3 (33), BAX (34) and TP53 (35); while the inhibition of TRIAP1 may increase the expression levels of TP53 and caspase 3 (36,37). In the present study, the protein expression levels of BAX, TP53 and caspase 3 were significantly increased, and those of TRIAP1 were decreased, in the miR-107 mimics group compared with the NC group. Therefore, it was hypothesized that miR-107 may induce the apoptosis of lung cancer cells by targeting TRIAP1 and increasing the expression levels of BAX, TP53 and caspase 3, which are three known tumor suppressors in lung cancer (32,33). Furthermore, previous studies have demonstrated that BCL2 knockdown decreased the viability and increased apoptosis of lung cancer cells (38), while overexpression of BCL2 decreased apoptosis of human lung cancer cells (39). Therefore, BCL2 may act as a tumor promoter in lung cancer. In the present study, the expression levels of TRIAP1 and BCL2 were decreased in the miR-107 mimics group compared with the NC group. Therefore, the inhibition of TRIAP1 induced by the overexpression of miR-107 may reduce the expression of BCL2, a known tumor promoter in lung cancer (38,39), which contributes to apoptosis in the process of lung cancer. However, the underlying mechanisms involved in TRIAP1 and apoptosis-associated proteins require further investigation.

In conclusion, miR-107 may inhibit A549 lung cancer cell proliferation and promote apoptosis by targeting TRIAP1, indicating that miR-107 may be a novel target in lung cancer treatment. However, the current study had the following limitations: i) The association and interaction of miR-107 and its target TRIAP1 require further investigation; ii) the effects of miR-107 and other targets on regulating lung cancer cell proliferation and apoptosis requires further study; iii) the histopathological patterns of lung cancer tissues were not distinguished; and iv) further experiments on different lung cancer cell lines and in in vivo models are required.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

PC collected lung cancer tumor and non-tumor adjacent tissues, analyzed and interpreted the patient data. JL, GC, BP, LY and BZ analyzed all the experimental data. YY performed all the cell experiments and was a major contributor in writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Jingmen No. 2 People's Hospital (Jingmen, China) and written consent was acquired from each patient.

Patients consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Didkowska J, Wojciechowska U, Mańczuk M and Łobaszewski J: Lung cancer epidemiology: Contemporary and future challenges worldwide. Ann Transl Med. 4:1502016. View Article : Google Scholar : PubMed/NCBI

2 

Nadel E, Truini A, Nakata A, Lin J, Reddy RM, Change AC, Ramnath N, Gotoh N, Beer DG and Chen G: A novel serum-4-microRNA signature for lung cancer detection. Sci Rep. 23:124642015. View Article : Google Scholar

3 

Tarver T: American cancer society cancer facts & figures 2014. Jour Consum Heal Intern. 16:366–367. 2012. View Article : Google Scholar

4 

Nanavaty P, Alvarez MS and Alberts WM: Lung cancer screening: Advantages, controversies, and applications. Cancer Control. 21:9–14. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Fortunato O, Boeri M, Moro M, Verri C, Mensah M, Conte D, Calecal L, Roz L, Pastorino U and Sozzi G: Mir-660 is downregulated in lung cancer patients and its replacement inhibits lung tumorigenesis by targeting MDM2-p53 interaction. Cell Death Dis. 11:e15642014. View Article : Google Scholar

6 

Huang J, Sun C, Wang S, He Q and Li D: MicroRNA miR-10b inhibition reduces cell proliferation and promotes apoptosis in non-small cell lung cancer (NSCLC) cells. Mol Biosyst. 11:2051–2059. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Kent OA and Mendell JT: A small piece in the cancer puzzle: microRNAs as a tumor suppressors and oncogenes. Oncogene. 25:6199–6196. 2006. View Article : Google Scholar

8 

Farazi TA, Hoell JI, Morozov P and Tuschl T: MicroRNAs in human cancer. Adv Exp Med Biol. 774:1–20. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Imamura T, Komatsu S, Ichikawa D, Miyamae M, Okajima W, Ohashi T, Kiuchi J, Nishibeppu K, Konishi H, Shiozaki A, et al: Depleted tumor suppressor miR-107 in plasma relates to tumor progression and is a novel therapeutic target in pancreatic cancer. Sci Rep. 7:57082017. View Article : Google Scholar : PubMed/NCBI

10 

Liu P, Qi X, Bian C, Yang F, Lin X, Zhou S, Xie C, Zhao X and Yi T: MicroRNA-18a inhibits ovarian cancer growth via directly targeting TRIAP1 and IPMK. Oncol Lett. 13:4039–4046. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Yang JZ, Bian L, Hou JG and Wang HY: MiR-550a-3p promotes non-small cell lung cancer cell proliferation and metastasis through down-regulating TIMP2. Eur Rev Med Pharmacol Sci. 22:4156–4165. 2018.PubMed/NCBI

12 

Han L, Chen W, Xia Y, Song Y, Zhao Z, Cheng H and Jiang T: MiR-101 inhibits the proliferation and metastasis of lung cancer by targeting zinc finger E-box binding homeobox 1. Am J Transl Res. 10:1172–1183. 2018.PubMed/NCBI

13 

Cui J, Mo J, Luo M, Yu Q, Zhou S, Li T, Zhang Y and Luo W: c-Myc-activated long non-coding RNA h19 downregulates miR-107 and promotes cell cycle progression of non-small cell lung cancer. Int J Clin Exp Pathol. 8:12400–12409. 2015.PubMed/NCBI

14 

Xia H, Li Y and Lv X: MicroRNA-107 inhibits tumor growth and metastasis by targeting the BDNF-mediated PI3K/AKT pathway in human non-small lung cancer. Int J Oncol. 49:1325–1333. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Takahashi Y, Forrest AR, Maeno E, Hashimoto T, Daub CO and Yasuda J: MiR-107 and miR-185 can induce cell cycle arrest in human non small cell lung cancer cell lines. PLoS One. 4:e66772009. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Zhang JJ, Wang CY, Hua L, Yao KH, Chen JT and Hu JH: MiR-107 promotes hepatocellular carcinoma cell proliferation by targeting Axin2. Int J Clin Exp Pathol. 8:5168–5174. 2015.PubMed/NCBI

18 

Zhang M, Wang X, Li W and Cui Y: MiR-107 and miR-25 simultaneously target LATS2 and regulate proliferation and invasion of gastric adenocarcinoma (GAC) cells. Biochem Biophys Res Commun. 460:806–812. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Cheng F, Yang Z, Huang F, Yin L, Yan G and Gong G: MicroRNA-107 inhibits gastric cancer cell proliferation and metastasis by targeting PI3K/AKT pathway. Microb Pathog. 121:110–114. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Yu M, Guo D, Cao Z, Xiao L and Wang G: Inhibitory effect of microRNA-107 on osteosarcoma malignancy through regulation of wnt/β catenin signaling in vitro. Cancer Invest. 36:175–184. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Wang P, Liu X, Shao Y, Wang H, Liang C, Han B and Ma Z: MicroRNA-107-5p suppresses non-small cell lung cancer by directly targeting oncogene epidermal growth factor receptor. Oncotarget. 8:57012–57023. 2017.PubMed/NCBI

22 

Chen JF, Mandel EM, Thomson JM, Wu QL, Callis T, Hammond SM, Conlon FL and Wang DZ: The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet. 38:228–233. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Liu P, Qi X, Bian C, Yang F, Lin X, Zhou S, Xie C, Zhao X and Yi T: MicroRNA-18a inhibits ovarian cancer growth via directly targeting TRIAP1 and IPMK. Oncol Lett. 13:4039–4046. 2017. View Article : Google Scholar : PubMed/NCBI

24 

He Q, Yang X, Ren X, Wen X, Zhang J, Wang Y, Liu N and Ma J: Overexpression of mitochondria mediator gene TRIAP1 by miR-320b loss is associated with progression in nasopharyngeal carcinoma. PLoS Genet. 12:e10061832016. View Article : Google Scholar : PubMed/NCBI

25 

Wang B, Zuo ZJ, Lv F, Zhao L, Du MJ and Gao YS: MiR-107 inhibits proliferation of lung cancer cells through regulating TP53 regulated inhibitor of apoptosis 1 (TRIAP1). Open Life Sci. 12:200–205. 2017.

26 

Tian XP, Jin XH, Li M, Huang WJ, Xie D and Zhang JX: The depletion of PinX1 involved in the tumorigenesis of non-small cell lung cancer promotes cell proliferation via p15/cyclin D1 pathway. Mol Cancer. 16:742017. View Article : Google Scholar : PubMed/NCBI

27 

Du B, Wang Z, Zhang X, Feng S, Wang G, He J and Zhang B: MicroRNA-545 suppresses cell proliferation by targeting cyclin D1 and CDK4 in lung cancer cells. PLoS One. 9:e880222014. View Article : Google Scholar : PubMed/NCBI

28 

Wang Y, Chen T, Huang H, Jiang Y, Yang L, Lin Z, He H, Liu T, Wu B, Chen J, et al: miR-363-3p inhibits tumor growth by targeting PCNA in lung adenocarcinoma. Oncotarget. 8:20133–20144. 2017.PubMed/NCBI

29 

Wang X, Shi W, Shi H, Lu S, Wang K, Sun C, He J, Jin W, Lv X, Zhou H and Shu Y: TRIM11 overexpression promotes proliferation, migration and invasion of lung cancer cells. J Exp Clin Cancer Res. 35:1002016. View Article : Google Scholar : PubMed/NCBI

30 

Xue Y, Wu L, Liu Y, Ma Y, Zhang L, Ma X, Yang Y and Chen J: ENTPD5 induces apoptosis in lung cancer cells via regulating caspase 3 expression. PLoS One. 10:e01200462015. View Article : Google Scholar : PubMed/NCBI

31 

Gu JJ, Qiao KS, Sun P, Chen P and Li Q: Study of EGCG induced apoptosis in lung cancer cells by inhibiting PI3K/Akt signaling pathway. Eur Rev Med Pharmacol Sci. 22:4557–4563. 2018.PubMed/NCBI

32 

Stegh AH: Targeting the p53 signaling pathway in cancer therapy-the promises, challenges and perils. Expert Opin Ther Targets. 16:67–82. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Zhang ZC, Liu JX, Shao ZW, Pu FF, Wang BC, Wu Q, Zhang YK, Zeng XL, Guo XD, Yang SH and He TC: In vitro effect of microRNA-107 targeting Dkk-1 by regulation of Wnt/β-catenin signaling pathway in osteosarcoma. Medicine (Baltimore). 96:e72452017. View Article : Google Scholar : PubMed/NCBI

34 

Sirotkin AV, Laukova M, Ovcharenko D, Brenaut P and Mlyncek M: Identification of microRNAs controlling human ovarian cell proliferation and apoptosis. J Cell Physiol. 223:49–56. 2010.PubMed/NCBI

35 

Yamakuchi M, Lotterman CD, Bao C, Hruban RH, Karim B, Mendell JT, Huso D and Lowenstein CJ: P53-induced microRNA-107 inhibits HIF-1 and tumor angiogenesis. Proc Natl Acad Sci USA. 107:6334–6339. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Andrysil Z, Kim J, Tan AC and Espinosa JM: A genetic screen identifies TCF3/ESA and TRIAP1 as pathway-specific regulators of the cellular response to p53 activation. Cell Rep. 3:1346–1354. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Liu P, Qi X, Bian C, Yang F, Lin XJ, Zhou S, Xie C, Zhao X and Yi T: MicroRNA-18a inhibits ovarian cancer growth via directly targeting TRIAP1 and IPMK. Oncol Lett. 13:4039–4046. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Zhao J, Li X, Zou M, He J, Han Y, Wu D, Yang H and Wu J: MiR-135a inhibition protects A549 cells from LPS-induced apoptosis by targeting Bcl-2. Biochem Biophys Res Commun. 452:951–957. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Sun SY, Yue P, Zhou JY, Wang Y, Choi Kim HR, Lotan R and Wu GS: Overexpression of BCL2 blocks TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in human lung cancer cells. Biochem Biophys Res Commun. 280:788–797. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2020
Volume 19 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cai P, Li J, Chen G, Peng B, Yu L, Zhao B and Yu Y: MicroRNA‑107 may regulate lung cancer cell proliferation and apoptosis by targeting TP53 regulated inhibitor of apoptosis 1. Oncol Lett 19: 1958-1966, 2020
APA
Cai, P., Li, J., Chen, G., Peng, B., Yu, L., Zhao, B., & Yu, Y. (2020). MicroRNA‑107 may regulate lung cancer cell proliferation and apoptosis by targeting TP53 regulated inhibitor of apoptosis 1. Oncology Letters, 19, 1958-1966. https://doi.org/10.3892/ol.2020.11248
MLA
Cai, P., Li, J., Chen, G., Peng, B., Yu, L., Zhao, B., Yu, Y."MicroRNA‑107 may regulate lung cancer cell proliferation and apoptosis by targeting TP53 regulated inhibitor of apoptosis 1". Oncology Letters 19.3 (2020): 1958-1966.
Chicago
Cai, P., Li, J., Chen, G., Peng, B., Yu, L., Zhao, B., Yu, Y."MicroRNA‑107 may regulate lung cancer cell proliferation and apoptosis by targeting TP53 regulated inhibitor of apoptosis 1". Oncology Letters 19, no. 3 (2020): 1958-1966. https://doi.org/10.3892/ol.2020.11248