Open Access

MicroRNA‑365b‑3p represses the proliferation and promotes the apoptosis of non‑small cell lung cancer cells by targeting PPP5C

Retraction in: /10.3892/ol.2023.14010

  • Authors:
    • Xiaomiao Zhang
    • Jin Wang
    • Yuqin Pan
    • Jun Zhao
    • Yingge Pan
    • Yunqi Yan
    • Zhenya Shen
  • View Affiliations

  • Published online on: March 17, 2021     https://doi.org/10.3892/ol.2021.12650
  • Article Number: 389
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA (miR)‑365b‑3p has been recently reported to induce cell cycle arrest and apoptosis in retinoblastoma; however, its expression pattern and biological function in non‑small cell lung cancer (NSCLC) remain unknown. The present study aimed to investigate the functional role of miR‑365b‑3p in NSCLC. The results demonstrated that miR‑365b‑3p expression level was significantly decreased in NSCLC tissues and cell lines compared with controls using reverse transcriptase quantitative PCR. Furthermore, miR‑365b‑3p expression level was overexpressed by miR‑365b‑3p mimics transfection in A549 cells, whereas it was downregulated following H1299 cell transfection with miR‑365b‑3p inhibitor. Restoration of miR‑365b‑3p inhibited cell proliferation, induced cell cycle G0/G1 arrest and stimulated apoptosis in A549 cells using CCK‑8 assay, colony formation and flow cytometry assay. However, miR‑365b‑3p inhibitor had the opposite effects in H1299 cells. Furthermore, results from bioinformatics analysis and luciferase reporter assay confirmed that serine/threonine protein phosphatase 5 (PPP5C) was a direct target of miR‑365b‑3p. In addition, online Kaplan‑Meier plotter software demonstrated that high PPP5C expression level was associated with lower overall survival and disease‑free survival in patients with NSCLC. Furthermore, PPP5C knockdown imitated the effects of miR‑365b‑3p mimics on A549 cell proliferation, cell cycle distribution and apoptosis, whereas its overexpression rescued the effects of miR‑365b‑3p mimics on A549 cell proliferation, cell cycle distribution and apoptosis. In conclusion, the findings from the present study suggested that miR‑365b‑3p may partly suppress NSCLC cell behaviors by targeting PPP5C, which may represent a promising therapeutic target for patients with NSCLC.

Introduction

Lung cancer is an aggressive disease that is considered as the leading cause of cancer-associated mortality worldwide (1), with ~228,150 new cases and 142,670 mortality cases in the United States in 2019 (2). Non-small cell lung cancer (NSCLC) accounts for 85% of lung cancer cases (3). Most patients with NSCLC are diagnosed at an advanced stage, or relapse after curative-intent surgery, chemotherapy, immunotherapy or the use of small molecule tyrosine kinase inhibitors in developed and developing countries (4), causing poor overall survival rates (5). It has been established that numerous aberrantly expressed genes and signaling pathways are involved in the pathogenesis of NSCLC (6). It is therefore crucial to explore the underlying mechanisms of NSCLC development.

MicroRNAs (miRNAs) are endogenous small non-coding RNAs of 19 to 25 nucleotides in length that lack protein-coding ability (7). In the last two decades, miRNAs have received increasing attention due to their critical role in diverse developmental and physiological procedures (8). miRNAs can regulate gene expression by binding to target mRNAs in a sequence specific manner (9). In particular, numerous studies have revealed that abnormal expression of miRNAs function as tumor suppressors or oncogenes in many types of cancer (10), including NSCLC (11). Previous studies reported aberrant expression of certain miRNAs in NSCLC that could regulate proliferation, migration, migration or apoptosis, including miR-361-3p (12), miR-10 (13), miR-126 (14) and miR-187-3p (15). miR-365b-3p has been demonstrated to be downregulated in retinoblastoma (RB) tissues and reported as a tumor suppressor in RB (16). In addition, Tian et al (17) reported that miR-365-3p promotes cell migration and invasion in hepatocellular carcinoma. However, the role of miR-365b-3p in NSCLC remains unknown.

Serine/threonine protein phosphatase 5 (PPP5C) is a member of the PPP gene family of serine/threonine protein phosphatases, which is broadly expressed in normal tissues (18). High expression level of PPP5C has been reported to be associated with hepatocellular carcinoma (19), ovarian cancer (20), pancreatic cancer (21) and prostate cancer (22). At the molecular level, PPP5C could interact with AMP-activated protein kinase, heat shock protein-90 and extracellular signal-regulated kinases (ERKs), and functions as a multifunctional regulator participating in various cellular processes, including cell proliferation, apoptosis and migration, and glucose homeostasis (23,24). However, the role of PPP5C and its association with miR-365b-3p in NSCLC have not been reported.

The present study explored the biological functions of miR-365b-3p in NSCLC cells by using miR-365b-3p mimics or inhibitor in vitro. Furthermore, luciferase reporter assay was performed to determine the unique regulatory mechanisms between miR-365b-3p and its potential target gene PPP5C in NSCLC cells. The present study further investigated whether miR-365b-3p regulates NSCLC cell proliferation, cell cycle progression and apoptosis by targeting PPP5C.

Materials and methods

Tissue sample collection

A total of 15 paired tumor tissues and adjacent tissues from patients with NSCLC were collected during surgical resection at the First Affiliated Hospital of Soochow University (Jiangsu, China) between September 2015 and December 2017. These tissues were immediately placed in liquid nitrogen and stored at −80°C until subsequent experiments. The clinicopathological characteristics of patients, including sex, age and Tumor-Node-Metastasis stage are summarized in Table I. Patients with one of the following conditions were excluded: i) Patients with recurrent lung cancer; ii) patients who use immune suppressive agents; iii) patients with severe organ diseases; iv) patients suffering from autoimmune diseases; and v) patients who received preoperative radiotherapy or chemotherapy. Each participant provided written informed consent. The present study was approved by the Ethics Committee of the First Affiliated Hospital of Soochow University (approval no. AHS152406).

Table I.

Clinicopathological characteristics of patients with non-small cell lung cancer.

Table I.

Clinicopathological characteristics of patients with non-small cell lung cancer.

CharacteristicCases (n=15)
Sex
  Male10
  Female5
Age, years
  <607
  ≥608
Smoking status
  Never smoked2
  Previously smoked4
  Currently smoke9
Tumor size, cm
  <49
  ≥46
TNM stage
  I+II13
  III2
Lymph node metastasis
  Negative8
  Positive7

[i] TNM, tumor-node-metastasis.

Cell culture

The three human NSCLC cell lines 95D, A549 and H1299 and the normal human bronchial epithelial cell line BEAS-2B were purchased from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences All cells were cultured in RPMI-1640 medium (HyClone) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and placed at 37°C in a humidified atmosphere containing 5% CO2.

Cell transfection

miR-365b-3p mimics, miR-365b-3p inhibitor and a scrambled negative control (miR-NC) were provided by Guangzhou RiboBio Co., Ltd. Plasmid-mediated pcDNA3.1-PPP5C (PPP5C) and the control vector, as well as small interfering (si)RNA targeting PPP5C (si-PPP5C) and si-NC were purchased from Shanghai GenePharma Co., Ltd. A549 or H1299 cells were seeded at 70–80% confluence in six-well plates and cultured overnight. A549 cells were transfected with 50 nM miR-365b-3p mimics or miR-NC, while H1299 cells were transfected with 50 nM miR-365b-3p inhibitor or miR-NC. For PPP5C knockdown, A549 cells were transfected with 30 nM si-PPP5C or si-NC. For the rescue experiments, A549 cells were co-transfected with 50 nM miR-365b-3p mimics and 10 µg empty vector or PPP5C at the same time. All transfections were performed for 48 h using reagent Lipofectamine 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols.

Reverse transcription quantitative (RT-q) PCR

Total RNA for miRNAs was extracted from tissues or cells using a High Pure miRNA isolation kit (Roche Diagnostics) and reversed transcription was performed using One Step Prime Script miRNA cDNA Synthesis kit (Takara Biotechnology Co., Ltd.) according to the manufacturer's instructions. Quantitative real time PCR was conducted on a ABI 7500 Fast Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) with TaqMan™ Multiplex Master Mix (Invitrogen; Thermo Fisher Scientific, Inc.) according to the following thermocycling conditions: Pre-degeneration at 95°C for 3 min and 40 cycles of 95°C for 30 sec, annealing and elongation at 60°C for 1 min. The sequences of the primers used were as follows: miR-365b-3p, forward, 5′-TAATGCCCCTAAAAAT-3′ and reverse, 5′-CCAGTGCAGGGTCCGAGGT-3′; and U6, forward, 5′-TGCGGGTGCTCGCTTCGGCAGC-3′ and reverse, 5′-CCAGTGCAGGGTCCGAGGT-3′. The expression level of miR-365b-3p was normalized to internal control U6 and was expressed as the 2−ΔΔCq (25).

Cell proliferation assay

The CCK-8 assay was performed to assess proliferation of A549 and H1299 cells in different time points with a Cell Counting Kit-8 (CCK-8; Wuhan Boster Biological Technology, Ltd.) according to the manufacturer's protocol. Briefly, ~3,500 transfected cells were seeded in each well of a 96-well plate and cultured for 0, 24, 48 and 72 h. Subsequently, cells were incubated with 10 µl CCK-8 reagent for 2 h. The optical density was read at 450 nm on a microplate reader (Agilent). To better reflect the proliferative ability of cells in the different groups, proliferation curves were constructed according the OD values at different time points. Each sample was analyzed in triplicate and the experiment was repeated three times.

Colony formation assay

Transfected A549 or H1299 cells were seeded into six-well plates at a density of 500 cells/well. After 2 weeks of cell culture in RPMI-1640 medium at 37°C, the naturally formed colonies were fixed with 4% paraformaldehyde for 30 min at 37°C, stained with 1% crystal violet at room temperature for 15 min and observed under a light microscope (magnification, ×200).

Cell cycle analysis

The cell cycle distribution of A549 and H1299 cells was examined using Cell Cycle Detection Kit (BD Biosciences) according to the manufacturer's instructions. After different cell transfections, cells were harvested, washed twice with PBS and fixed with 70% ethanol at 4°C overnight. Cells were washed again with PBS and stained with propidium iodide (PI; Nanjing KeyGen Biotech Co., Ltd.) staining solution (50 µg/ml; containing 1 mg/ml RNase A and 0.1% Triton X-100 in PBS) for 30 min in the dark. Finally, the percentage of cells at G0/G1, S and G2/M phases was quantified using a flow cytometer (BD Biosciences) equipped with Modfit LT software (version 5.1; BD Biosciences). Each sample was analyzed in triplicate and the experiment was repeated three times.

Cell apoptosis analysis

A549 and H1299 cell apoptosis was examined using an Annexin V-Fluorescein Isothiocyanate (FITC) Apoptosis Detection kit (BD Biosciences) according to the manufacturer's instructions. Briefly, transfected cells were harvested, washed twice with PBS and stained with 5 µl Annexin V-FITC and 5 µl PI for 20 min at room temperature. Subsequently, cell apoptotic rate was detected using a FACS Calibur flow cytometer (BD Biosciences). Each sample was analyzed in triplicate and the experiment was repeated three times.

Bioinformatics analysis and luciferase reporter assay

The target genes of miR-365b-3p were identified using the prediction tool TargetScanHuman7.1 (http://www.targetscan.org/vert_71). The mutant (MUT) type of PPP5C 3′-UTR was constructed using a Fast Mutagenesis System kit (TransGen Biotech Co., Ltd.). Luciferase complexes were constructed by ligating the wild-type (WT) or mutant (MUT) PPP5C in the putative binding sites for miR-365b-3p into the pGL3 luciferase reporter vector (Promega Corporation). For luciferase reporter assay, co-transfection with 50 ng of 3′-UTR-WT or 3′-UTR-MUT PPP5C vector and 20 µM miR-365b-3p mimics or miR-NC were performed in A549 cells. Meanwhile, co-transfection with 50 ng of 3′-UTR-WT or 3′-UTR-MUT PPP5C vector and 20 µM miR-365b-3p inhibitor or miR-NC were carried out in H1299 cells. All transfections were performed for 48 h using Lipofectamine 2000 and the Firefly/Renilla luciferase activity was determined on the Dual-Luciferase Reporter Assay System (Promega Corporation). Firefly luciferase activity was normalized to Renilla luciferase activity.

Western blotting

Total protein was extracted from clinical tissues or NSCLC cells using RIPA lysis buffer (Beyotime Institute of Biotechnology) and protein concentration was measured using a BCA assay kit (Beyotime Institute of Biotechnology). Proteins (30 µg) were separated by 10% SDS-PAGE and transferred onto polyvinylidene difluoride membranes (EMD Millipore). Membranes were blocked with 5% non-fat milk at room temperature for 1 h and incubated with the primary antibodies (1:1,000) against PPP5C (cat. no. 11715-1-AP; ProteinTech Group, Inc.), CDK4 (cat. no. ab226474; Abcam), Cyclin D1 (cat. no. ab226977; Abcam), Bax (cat. no. ab270742; Abcam), Bcl-2 (cat. no. ab196495; Abcam) and GAPDH (cat. no. 10494-1-AP; ProteinTech Group, Inc.) overnight at 4°C, followed by incubation with horseradish peroxidase-conjugated secondary antibody (1:5,000; cat. no. SC-2054; Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. GAPDH was served as a loading control. Protein bands were visualized with an Enhanced Chemiluminescence kit (Pierce; Thermo Fisher Scientific, Inc.).

Kaplan-Meier plotter analysis

Considering the relatively small sample size (n=15) in the clinical collection, the Kaplan-Meier Plotter database (http://kmplot.com/analysis) was used to assess the prognostic value of PPP5C in lung cancer. The patient samples were split into two groups, a high and a low expression group. The two groups were compared using a Kaplan-Meier survival plot. The hazard ratio (HR) with 95% confidence intervals and log rank P-value was calculated.

Statistical analysis

Statistical analysis was conducted using the SPSS 19.0 software version (SPSS, Inc.). Data were expressed as means ± standard deviation from at least three repeated experiments. Student's t-test and ANOVA followed by Tukey post hoc test were performed to evaluate differences between two groups and multiple groups, respectively. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-365b-3p expression is downregulated in NSCLC tissues and cell lines

Using RT-qPCR, the expression level of miR-365b-3p in paired tumor tissues and adjacent normal tissues derived from 15 patients with NSCLC was evaluated. As presented in Fig. 1A and B, miR-365b-3p expression level was significantly decreased in NSCLC tissues compared with adjacent normal tissues. In addition, miR-365b-3p expression level was significantly decreased in NSCLC cell lines compared with the normal lung epithelial cell line BEAS-2B (Fig. 1C).

miR-365b-3p regulates proliferation, cell cycle distribution and apoptosis in NSCLC cells

To explore the biological function of miR-356b-3p in NSCLC cells, A549 and H1299 cells, which had a relative lower and higher miR-356b-3p expression level, respectively, were selected for gain-of-function and loss-of-function assays, respectively. At first, miR-356b-3p expression was significantly up- and down regulated in transfected A549 and H1299 cells, respectively (Fig. 2A). Subsequently, the results from CCK-8 (Fig. 2B) and colony formation (Fig. 2C) assays demonstrated that cell proliferation was significantly decreased in A549 cells transfected with miR-356b-3p mimics compared with miR-NC. Conversely, H1299 cells transfected with miR-356b-3p inhibitor exhibited a significantly increased cell proliferation compared with miR-NC. Furthermore, flow cytometry was used to analyze the cell cycle distribution. As presented in Fig. 2D and E, miR-356b-3p overexpression induced A549 cell cycle G0/G1 phase arrest, as reflected by significantly increased cell population in the G0/G1 phase and decreased cell populations in S and G2/M phase; however, miR-356b-3p knockdown reversed G0/G1 phase arrest in H1299 cells. Subsequently, cell apoptosis was assessed. The results demonstrated that apoptotic rate was significantly increased in the miR-356b-3p mimics group in A549 cells but declined when H1299 cells were transfected with miR-356b-3p inhibitor (Fig. 2F and G). The results from western blotting further demonstrated that miR-365b-3p mimics downregulated and miR-365b-3p inhibitor upregulated the expression of CDK4, cyclin D1 and Bcl-2 compared with miR-NC. In addition, the pro-apoptotic Bax expression was increased in A549 cells following miR-365b-3p overexpression, whereas it was decreased in H1299 cells following miR-365b-3p knockdown (Fig. 2H). Taken together, these findings suggested that miR-356b-3p may inhibit the proliferation, induce cell cycle arrest and stimulate apoptosis in NSCLC cells.

PPP5C is a direct gene of miR-365b-3p

To explore the molecular mechanisms underlying miR-365b-3p regulation of NSCLC cell proliferation, the prediction tool TargetScanHuman7.1 was used to predict the potential target genes of miR-365b-3p. Among these predicted target genes, PPP5C was selected for as a putative target gene of miR-365b-3p for further analysis. The predicted possible binding sites between PPP5C and miR-365b-3p were presented in Fig. 3A. Subsequently, luciferase reporter assay was performed to confirm this prediction. The results demonstrated that miR-365b-3p overexpression in A549 cells significantly decreased the relative luciferase activity of WT PPP5C (Fig. 3B), whereas miR-365b-3p knockdown in H1299 cells significantly increased the relative luciferase activity of WT PPP5C (Fig. 3C). However, modulating miR-365b-3p level did not affect the relative luciferase activity of MUT PPP5C in both A549 and H1299 cells. Furthermore, the protein expression of PPP5C was decreased following miR-365b-3p overexpression in A549 cells, but was elevated in H1299 cells after miR-365b-3p knockdown (Fig. 3D). In addition, PPP5C protein expression was upregulated in the three NSCLC cell lines 95D, A549 and H1299 compared with the normal lung epithelial cell line BEAS-2B (Fig. 3E). These findings demonstrated that miR-365b-3p could negatively regulate PPP5C expression by directly binding to its 3′UTR in NSCLC cells.

PPP5C knockdown inhibits proliferation, induces cell cycle arrest and stimulates apoptosis in NSCLC cells

PPP5C protein expression was analyzed in NSCLC tissues using western blotting. As presented in Fig. 4A, PPP5C expression was increased in tumor tissues compared with adjacent normal tissues from six representative cases of NSCLC. Through online Kaplan-Meier plotter software, the association between PPP5C expression and overall survival in patients with lung cancer was further evaluated. The results revealed that high PPP5C expression was associated with lower overall survival (Fig. 4B; HR=1.25; 95% CI=1.10–1.42; P<0.001) and disease-free survival (Fig. 4C; HR=1.79; 95% CI=1.47–2.18; P<0.001) in patients with lung cancer. To confirm the role of PPP5C in NSCLC, loss-of-function assays were performed in A549 cells by transfection with si-PPP5C or si-NC. As presented in Fig. 5A, si-PPP5C efficiently downregulated PPP5C protein expression in A549 cells. Consistent with miR-365b-3p overexpression, the results demonstrated that PPP5C significantly inhibited cell proliferation (Fig. 5B), induced cell cycle G0/G1 phase arrest (Fig. 5C) and stimulated cell apoptosis (Fig. 5D) in A549 cells. These findings demonstrated that PPP5C may be considered as an oncogene in NSCLC.

PPP5C overexpression partially rescues the effects of miR-365b-3p mimics in NSCLC cells

Rescue experiments were performed to investigate whether PPP5C could be a downstream function regulator involved in miR-365b-3p regulation of cell proliferation, cell cycle and cell apoptosis. The interaction between miR-365b-3p and PPP5C was first confirmed through co-transfecting the miR-365b-3p mimics and the PPP5C vector into A549 cells. As presented in Fig. 6A, a decrease in PPP5C expression induced by the miR-365b-3p mimics was restored following transfection with PPP5C vector in A549 cells. Furthermore, the results form CCK-8 assay demonstrated that PPP5C upregulation impaired the inhibitory effect of miR-365b-3p mimics on A549 cell proliferation (Fig. 6B). In addition, the inhibitory effects of miR-365b-3p mimics on cell cycle G0/G1 phase arrest (Fig. 6C) and apoptosis (Fig. 6D) were also reversed following PPP5C overexpression in A549 cells. Taken together, these results suggested that PPP5C overexpression impaired the suppressive effect of miR-365b-3p in NSCLC cells.

Discussion

miR-365b-3p has been identified as a novel miRNA described in various types of tumor, including retinoblastoma (16) and hepatocellular carcinoma (17); however, it has not been reported in NSCLC. Although PPP5C has been widely demonstrated to be an oncogene, its role remains unclear in NSCLC. Based on the prediction analysis between miR-365b-3p and PPP5C, the present study aimed to explore the function of miR-365b-3p and validate whether PPP5C could be a downstream functional regulator involved in miR-365b-3p participating in NSCLC cell functions. The results demonstrated that miR-365b-3p expression was significantly decreased in NSCLC tissue samples compared with adjacent normal tissues. Furthermore, miR-365b-3p expression was significantly downregulated in the three NSCLC cell lines H1299, A549 and H1975 compared with the human lung cell line BEAS-2B. These results were consistent with those from Wang et al (16) demonstrating that miR-365b-3p expression level is significantly decreased in RB tissues. miR-365b-3p may therefore be a tumor suppressor in NSCLC.

Abnormal abundance of miRNAs has been shown to influence the biological behavior, including sustaining cell proliferation, stimulating survival signaling and angiogenesis in cancer (26). In order to determine the role of miR-365b-3p in NSCLC, A549 and H1299 cells were selected for gain-of-function and loss-of-function assays, respectively. Despite major difference between these two cell lines lies in terms of p53 oncogene activity, this did not affect the role of miR-365b-3p in NSCLC cell proliferation, cell cycle progression and apoptosis. As expected, the results from the present study demonstrated that restoration of miR-365-3p inhibited cell proliferation and induced G0/G1 cell-cycle arrest and apoptosis; however, miR-365b-3p knockdown in H1299 cells showed the opposite results. These results indicated that miR-365b-3p may function as a tumor suppressor in NSCLC cells.

To explore the molecular mechanisms of miR-365b-3p in NSCLC, the target genes of miR-365b-3p were predicted, PPP5C was selected as a potential target of miR-365b-3p. The results from luciferase reporter gene assay and western blotting demonstrated that miR-365b-3p could directly target the 3′UTR of PPP5C and decrease the PPP5C protein expression in NSCLC cells. In addition, PPP5C knockdown could imitate the effects of miR-365b-3p on the biological behaviors of NSCLC cells, whereas PPP5C overexpression reversed the effects of miR-365b-3p on the biological behaviors of NSCLC cells. PPP5C belongs to the PPP family of serine/threonine protein phosphatases and is actively involved in the progression of numerous types of cancer (1921). PPP5C has been demonstrated to be highly expressed in prostate cancer tissues and induce cancer cell proliferation and survival by decreasing the phosphorylation of JNK and ERK1/2 (27). Inversely, PPP5C exerts a tumor suppressor in hepatocellular carcinoma via interaction with AMP-activated protein kinase (23). A previous study reported that PPP5C serves a crucial role in the canonical and non-canonical WNT signaling pathways (28). The findings from the present study indicated that miR-365b-3p may inhibit NSCLC cell proliferation by targeting PPP5C.

In conclusion, the results of the present study demonstrated that upregulated miR-365b-3p expression was associated with decreased cell proliferation, increased G0/G1 cell cycle arrest, and stimulated apoptosis in NSCLC cells. However, the present study is not without limitations, including the lack of migration and invasion assays, relatively small sample sizes were used, the lack of cell cycle arrest analysis in clinical samples or a mouse model, and the lack of deeper investigation on the underlying molecular mechanism.

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 upon reasonable request.

Authors' contributions

ZS contributed to the conception and design of the present study. XZ and JW performed the experiments. YGP and YY helped perform the literature review and collected the data. JZ analyzed and interpretated the data. YQP participated in data acquisition and data analysis. All authors confirmed the agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of First Affiliated Hospital of Soochow University (Jiangsu, China; approval no. AHS152406) and performed in accordance with the Declaration of Helsinki. All participants provided written informed consent prior to the study start.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

NSCLC

non-small cell lung cancer

miR-365b-3p

microRNA-365b-3p

PPP5C

serine/threonine protein phosphatase 5

References

1 

Muller M, Schouten RD, De Gooijer CJ and Baas P: Pembrolizumab for the treatment of non-small cell lung cancer. Expert Rev Anticancer Ther. 17:399–409. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Bremnes RM, Donnem T and Busund LT: Importance of tumor infiltrating lymphocytes in non-small cell lung cancer? Ann Transl Med. 4:1422016. View Article : Google Scholar : PubMed/NCBI

4 

Herbst RS, Morgensztern D and Boshoff C: The biology and management of non-small cell lung cancer. Nature. 553:446–454. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, Mok TS, Reck M, Van Schil PE, Hellmann MD, et al: Metastatic non-small cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 29 (Suppl 4):iv192–iv237. 2018. View Article : Google Scholar

6 

Hong L, Sun H, Lv X, Yang D, Zhang J and Shi Y: Expression of periostin in the serum of NSCLC and its function on proliferation and migration of human lung adenocarcinoma cell line (A549) in vitro. Mol Biol Rep. 37:2285–2293. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Favara MT, Aghai Z, Gayen nee' Betal S, Fong G and Addya S: Effects of chorioamnionitis on microRNA profile in cord blood mononuclear leukocytes. Pediatrics. 144:6312019.

8 

Lin S and Gregory RI: MicroRNA biogenesis pathways in cancer. Nat Rev Cancer. 15:321–333. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Catalanotto C, Cogoni C and Zardo G: MicroRNA in control of gene expression: An overview of nuclear functions. Int J Mol Sci. 17:17122016. View Article : Google Scholar

10 

Di Leva G, Garofalo M and Croce CM: microRNAs in cancer. Annu Rev Pathol. 9:287–314. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Leonetti A, Assaraf YG, Veltsista PD, El Hassouni B, Tiseo M and Giovannetti E: MicroRNAs as a drug resistance mechanism to targeted therapies in EGFR-mutated NSCLC: Current implications and future directions. Drug Resist Updat. 42:1–11. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Chen W, Wang J, Liu S, Wang S, Cheng Y, Zhou W, Duan C and Zhang C: MicroRNA-361-3p suppresses tumor cell proliferation and metastasis by directly targeting SH2B1 in NSCLC. J Exp Clin Cancer Res. 35:762016. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Song L, Li D, Gu Y, Wen ZM, Jie J, Zhao D and Peng LP: MicroRNA-126 targeting PIK3R2 inhibits NSCLC A549 cell proliferation, migration, and invasion by regulation of PTEN/PI3K/AKT pathway. Clin Lung Cancer. 17:e65–e75. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Sun C, Li S, Yang C, Xi Y, Wang L, Zhang F and Li D: MicroRNA-187-3p mitigates non-small cell lung cancer (NSCLC) development through down-regulation of BCL6. Biochem Biophys Res Commun. 471:82–88. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wang J, Wang X, Wu G, Hou D and Hu Q: MiR-365b-3p, down-regulated in retinoblastoma, regulates cell cycle progression and apoptosis of human retinoblastoma cells by targeting PAX6. FEBS Lett. 587:1779–1786. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Tian Q, Sun HF, Wang WJ, Li Q, Ding J and Di W: miRNA-365b promotes hepatocellular carcinoma cell migration and invasion by downregulating SGTB. Future Oncol. 15:2019–2028. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Amable L, Grankvist N, Largen JW, Ortsäter H, Sjöholm Å and Honkanen RE: Disruption of serine/threonine protein phosphatase 5 (PP5:PPP5c) in mice reveals a novel role for PP5 in the regulation of ultraviolet light-induced phosphorylation of serine/threonine protein kinase Chk1 (CHEK1). J Biol Chem. 286:40413–40422. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Feng L, Sun P, Li Z, Liu M and Sun S: Knockdown of PPP5C inhibits growth of hepatocellular carcinoma cells in vitro. Appl Biochem Biotechnol. 175:526–534. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Zheng X, Zhang L, Jin B, Zhang F, Zhang D and Cui L: Knockdown of protein phosphatase 5 inhibits ovarian cancer growth in vitro. Oncol Lett. 11:168–172. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Zhu J, Ji Y, Yu Y, Jin Y, Zhang X, Zhou J and Chen Y: Knockdown of serine/threonine protein phosphatase 5 enhances gemcitabine sensitivity by promoting apoptosis in pancreatic cancer cells in vitro. Oncol Lett. 15:8761–8769. 2018.PubMed/NCBI

22 

Grankvist N, Amable L, Honkanen RE, Sjöholm A and Ortsäter H: Serine/threonine protein phosphatase 5 regulates glucose homeostasis in vivo and apoptosis signalling in mouse pancreatic islets and clonal MIN6 cells. Diabetologia. 55:2005–2015. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Chen YL, Hung MH, Chu PY, Chao TI, Tsai MH, Chen LJ, Hsiao YJ, Shih CT, Hsieh FS and Chen KF: Protein phosphatase 5 promotes hepatocarcinogenesis through interaction with AMP-activated protein kinase. Biochem Pharmacol. 138:49–60. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Mazalouskas MD, Godoy-Ruiz R, Weber DJ, Zimmer DB, Honkanen RE and Wadzinski BE: Small G proteins Rac1 and Ras regulate serine/threonine protein phosphatase 5 (PP5)·extracellular signal-regulated kinase (ERK) complexes involved in the feedback regulation of Raf1. J Biol Chem. 289:4219–4232. 2014. 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 

Peng Y and Croce CM: The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 1:150042016. View Article : Google Scholar : PubMed/NCBI

27 

Lv JM, Chen L, Gao Y, Huang H, Pan XW, Liu X, Chen M, Qu FJ, Li L, Wang JK, et al: PPP5C promotes cell proliferation and survival in human prostate cancer by regulating of the JNK and ERK1/2 phosphorylation. Onco Targets Ther. 11:5797–5809. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Xie J, Han M, Zhang M, Deng H and Wu W: PP5 (PPP5C) is a phosphatase of Dvl2. Sci Rep. 8:27152018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

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
Zhang X, Wang J, Pan Y, Zhao J, Pan Y, Yan Y and Shen Z: MicroRNA‑365b‑3p represses the proliferation and promotes the apoptosis of non‑small cell lung cancer cells by targeting PPP5C Retraction in /10.3892/ol.2023.14010. Oncol Lett 21: 389, 2021
APA
Zhang, X., Wang, J., Pan, Y., Zhao, J., Pan, Y., Yan, Y., & Shen, Z. (2021). MicroRNA‑365b‑3p represses the proliferation and promotes the apoptosis of non‑small cell lung cancer cells by targeting PPP5C Retraction in /10.3892/ol.2023.14010. Oncology Letters, 21, 389. https://doi.org/10.3892/ol.2021.12650
MLA
Zhang, X., Wang, J., Pan, Y., Zhao, J., Pan, Y., Yan, Y., Shen, Z."MicroRNA‑365b‑3p represses the proliferation and promotes the apoptosis of non‑small cell lung cancer cells by targeting PPP5C Retraction in /10.3892/ol.2023.14010". Oncology Letters 21.5 (2021): 389.
Chicago
Zhang, X., Wang, J., Pan, Y., Zhao, J., Pan, Y., Yan, Y., Shen, Z."MicroRNA‑365b‑3p represses the proliferation and promotes the apoptosis of non‑small cell lung cancer cells by targeting PPP5C Retraction in /10.3892/ol.2023.14010". Oncology Letters 21, no. 5 (2021): 389. https://doi.org/10.3892/ol.2021.12650