Open Access

METTL3‑mediated m6A modification of Bcl‑2 mRNA promotes non‑small cell lung cancer progression

Corrigendum in: /10.3892/or.2023.8502

  • Authors:
    • Yongxi Zhang
    • Shuyuan Liu
    • Tiesuo Zhao
    • Chengxue Dang
  • View Affiliations

  • Published online on: June 15, 2021     https://doi.org/10.3892/or.2021.8114
  • Article Number: 163
  • 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

Methyltransferase‑like 3 (METTL3) is an RNA methyltransferase that mediates modification of N6‑methyladenosine (m6A), which serves as an oncogene in various types of cancer. The role of m6A modification in the onset and progression of cancer has attracted growing attention. However, the functional and regulatory mechanisms of METTL3 in non‑small cell lung cancer (NSCLC) progression are still poorly understood. In the present study, METTL3 expression in NSCLC tissue was analyzed using the Gene Expression Profiling Interactive Analysis database. Western blotting and reverse transcription‑quantitative PCR were performed to evaluate the expression of METTL3 in NSCLC tissue and cell lines. Here, knockdown and overexpression of METTL3 notably decreased NSCLC cell viability, apoptosis and migration in vitro and, as well as tumorigenicity in vivo. Expression of METTL3 was upregulated in NSCLC tissue. METTL3 overexpression promoted cell viability and migration in NSCLC, while knockdown of METTL3 yielded the opposite result in vivo and in vitro. METTL3 increased Bcl‑2 translation via m6A modification, which increased viability and enhanced migration of NSCLC cells. METTL3 served as an oncogene in NSCLC via METTL3‑mediated Bcl‑2 mRNA m6A modification, which indicated that targeting METTL3 may be an effective therapeutic strategy for clinical management of NSCLC.

Introduction

Lung cancer is the most common malignancy worldwide and poses a serious threat to human life and health (1). In addition, non-small-cell lung cancer (NSCLC) accounts for ~85% of total lung cancer diagnoses (2). NSCLC is often asymptomatic at the early stage of the disease and most patients are diagnosed when the disease has progressed to the intermediate or late stage (2,3). Although an increasing number of diagnostic and therapeutic strategies have been developed, the overall survival rate remains unsatisfactory (4). Some patients still die due to distant metastasis and drug resistance (5,6). Thus, it is key to investigate the potential molecular mechanisms of NSCLC and to identify novel treatment approaches to improve outcomes in patients with NSCLC.

RNA N6-methyladenosine (m6A) modification is the most abundant conserved post-translational modification in eukaryotic organisms (7). It primarily regulates gene expression, mRNA stability, alternative splicing, translation efficiency and primary microRNA (miRNA) processing (8,9). The dynamic modification of m6A is regulated by two types of catalytic proteins (m6A ‘writers’ and ‘erasers’) (10). The ‘writers’ primarily include methyltransferase-like (METTL)3, METTL14 and Wilms tumor 1 associated protein (11,12). ‘Erasers’, such as alkylation repair homolog protein 5 and fat mass and obesity-associated protein, serve as demethylases to reverse methylation (13). A previous study demonstrated that m6A modulators exhibit various biological functions in different processes, such as embryonic growth, stem cell differentiation, circadian rhythm and tumor progression (14).

METTL3 is a core methyltransferase for m6A modification (15). Certain studies have reported that METTL3 serves an oncogenic role in gastric, colorectal and bladder cancer, as well as hepatocellular carcinoma (1518). By contrast, a recent study showed that METTL3 is a tumor suppressor in kidney cancer (19). Nevertheless, the biological function of METTL3 and its underlying regulatory mechanisms in NSCLC have not been sufficiently described.

The aim of the present study was to investigate METTL3 expression in NSCLC cells and human NSCLC tissue and to determine the role of METTL3 in NSCLC progression both in vitro and in vivo. The results of the present study may provide a novel therapeutic target for treatment of NSCLC.

Materials and methods

Patients and specimen collection

A total of 77 NSCLC tissue samples and paired adjacent normal tissue samples (distance, ≥3 cm) were obtained from patients undergoing surgery from May 2018 to January 2020 at the First Affiliated Hospital of Xinxiang Medical University (Xinxiang, China). Of these patients, 44 were male (mean age, 60.22 years) and 33 were female (mean age, 58.6 years). The histological type of the tumors was assessed using the World Health Organization Classification (20) by two independent pathologists. Ethics approval was granted by the Ethics Committee of the First Affiliated Hospital of Xinxiang Medical University (approval no. KN201808002). Oral consent was obtained from patients. All experiments were performed in accordance with approved guidelines and regulations (21).

Cell culture

Human NSCLC cell lines (A549, PC9, H1299, H1975 and HCC827) and normal pulmonary epithelial cells (BEAS-2B) were obtained from the American Type Culture Collection. A549, PC9, H1299, H1975 and HCC827 cells were cultured in RPMI-1640 medium (Invitrogen; Thermo Fisher Scientific, Inc.). BEAS-2B cells were cultured in DMEM (Invitrogen; Thermo Fisher Scientific, Inc.). All media were supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) and all cells were incubated at 37°C with 5% CO2.

Establishment of stable cell lines

Stable cell lines were constructed using lentivirus system. The plasmids used to make stably overexpressed/knockdown cell lines were based on the pcDNA3/Flag-METTL3 plasmid (Addgene; cat. no. 53739) or short hairpin (sh)RNA-METTL3 (GenePharma Co., Ltd.). To generate lentivirus particles, H1299 or H1975 cells were co-transfected with plasmids together with Trans-lentiviral packaging plasmids using Trans-lentiviral Packaging kit (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Lentivirus particles were harvested at 48 h post transfection and concentrated using Lenti-X concentrator according to manufactory instruction (Clontech Laboratories, Inc.). Stable cell lines was transduced with lentivirus particles in serum-free medium (Invitrogen; Thermo Fisher Scientific, Inc.) containing 8 µg/ml polybrene (Sigma-Aldrich; Merck KGaA) followed by selection with 2 µg/ml puromycin for ≥10 days. The cells with stable overexpression or knockdown of METTL3 were s identified by RT-qPCR and immunoblot analysis. The target sequences of shRNAs were as follows: shMETTL3#1, 5′-GCCAAGGAACAATCCATTGTT-3′; shMETTL3#2, 5′-GCTGCACTTCAGACGAATTAT-3′; and shMETTL3#, 5′GCTTACTATCTAGCATCACAT-3′.

RNA isolation and reverse transcription-quantitative (RT-q)PCR

Total RNA was isolated from cells using TRIzol® Reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Then, 1 µg total RNA was reverse-transcribed to complementary DNA using PrimeScript™ RT Master Mix (Takara Bio, Inc.). The reaction conditions were 37°C for 15 min and 85°C for 15 sec. mRNA expression was measured using a TB Green™ kit (Takara Bio, Inc.), according to the manufacturer's instructions. Quantification of the target and reference (GAPDH) genes was performed in triplicate using a LightCycler® 480 II (Roche Diagnostics GmbH) with an initial denaturing step at 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec and 60°C for 30 sec. The primers used were as follows: GAPDH forward, 5′-CTGGGCTACACTGAGCACC-3′ and reverse, 5′-GGAACGCTTCACGAATTTG-3′; and METTL3 forward, 5′-AAGTGGTCGTTGAGGGCAATG-3′ and reverse, 5′-GCTTGGCGTGTGGTCTTT-3′. All primers were synthesized by Shanghai Shenggong Biology Engineering Technology Service, Ltd.. The analysis was conducted with the 2−ΔΔCq quantification method (22).

Cell viability assay

Cells were seeded in 96-well plates at a density of 2,000 cells/well. Cells were incubated at 37°C in an incubator with 5% CO2 for 0, 24, 48 and 72 h. Cell Counting Kit-8 (CCK-8; Sigma-Aldrich; Merck KGaA) was used to measure cell viability according to the manufacturer's instructions. The absorbance was measured at a wavelength of 450 nm using a plate reader (Bio-Rad Laboratories, Inc.).

Apoptosis

Apoptosis was measured using flow cytometry with a FITC-Annexin V/PI detection kit (Wanleibio Co., Ltd.). A total of 1×106 cells per well was seeded in 6-well plates and cells were then harvested after culturing for 48 h at 37°C. Afterwards, the cells were stained in the dark with FITC-Annexin V and PI at room temperature for 15 min. Subsequently, the percentages of apoptotic cells were measured using FACS flow cytometry (BD Biosciences) and analyzed using FlowJo 8.6.3 (Tree Star, Inc.).

Cell migration assay

Cell migration assays were performed using a cell migration assay kit (Corning, Inc.) and Transwell inserts. Briefly, 2×104 cells were seeded in the upper chamber of a Boyden chamber (without serum in the RPMI-1640 medium, Invitrogen; Thermo Fisher Scientific, Inc.). The lower portion of the chamber was filled with 600 µl RPMI-1640 medium (Invitrogen; Thermo Fisher Scientific, Inc.) supplemented with 20% FBS (Gibco; Thermo Fisher Scientific, Inc.). Following incubation for 24 h at 37°C, migrated cells in the lower chambers were fixed with 4% paraformaldehyde for 15 min at room temperature and stained with 0.05% crystal violet (Sigma-Aldrich; Merck KGaA) for 10 min at room temperature. Cells were counted in five randomly selected fields of view under a light microscope (Leica GmbH DMI4000B) at 200× magnification.

m6A qPCR

m6A qPCR was performed as reported previously (23) to assess the relative abundance of the selected mRNA in m6A antibody immunoprecipitation (IP) and input samples. Isolated m6A-RIP RNA was quantified by qPCR. The primers used were as follows: Bcl-2 forward, 5′-ATCGCCCTGTGGATGACTGAGT-3′ and reverse: 5′-GCCAGGAGAAATCAAACAGAGGC-3′.

m6A sequencing (m6A-seq)

Total RNA was isolated and purified using PolyATtract mRNA Isolation Systems kit (cat. no. Z5310; Promega Corporation). m6A-seq was performed as previously described (24). The NEBNext Ultra Directional RNA Library Prep kit (cat. no. E7420L; Illumina, Inc.) was used for library construction. Clustered libraries were sequenced with an Illumina HiSeq 4000 (Illumina, Inc.) by Novogene Co., Ltd.

Western blotting

Total protein was extracted from cells using RIPA buffer containing protease and phosphatase inhibitor cocktail (both from Sigma-Aldrich; Merck KGaA). The protein concentration was determined with a BCA Protein Assay kit (Beyotime Institute of Biotechnology). Cellular proteins were separated by 10% SDS-PAGE and then transferred to PVDF membranes. The membranes were blocked with 5% non-fat milk and 0.1% Tween-20 in Tris-buffered saline at room temperature for 2 h. Membranes were probed at 4°C overnight with primary antibodies against METTL3 (1:2,000; cat. no. 86132S; Cell Signaling Technology, Inc.), Bcl-2 (1:1,000; cat. no. ab32124; Abcam) and GAPDH (1:5,000; cat. no. 5174S; Cell Signaling Technology, Inc.). After washing in PBST (0.5% Tween-20). Three times, the blots were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (1:1,000; cat. no. BA1070; Wuhan Boster Biological Technology, Ltd.) at room temperature for 1 h. The signal was visualized using ECL western blotting detection reagents (Tanon Science and Technology Co., Ltd). Protein expression levels were quantified using ImageJ software (version 1.50; National Institutes of Health).

Experimental mouse techniques

A total of 15 female BALB/c nude mice (age, 6 weeks; weight, 18–20 g) were purchased from the Model Animal Center at Nanjing University (Nanjing, China) and raised under pathogen-free conditions in the Xinxiang Medical University Animal Center (Xinxiang, China). The mice were housed with filtered air, 12 h light/dark cycle, constant temperature (25°C), relative humidity (50±5%) and free access to food and water. In order to establish a human NSCLC xenograft model, 5×106 H1299, sh-METTL3-H1299 and METTL3 stably overexpressed H1299 cells (2×106 per mouse) were subcutaneously injected into mice. Tumor growth was observed daily. Tumor volume was calculated as follows: 0.5× (length × width2). At 24 days post-inoculation, the maximum diameter exhibited by a single subcutaneous tumor was 15 mm and mice were anesthetized by intraperitoneal administration of sodium pentobarbital (50 mg/kg), then sacrificed by cervical dislocation. Tumors were harvested and weighed for histological analysis. Lungs were extracted and fixed for 24 h at room temperature using 10% (v/v) neutral-buffered formalin and transferred to 70% ethanol until embedding in paraffin then sectioned at 3 µm. For hematoxylin-eosin staining, sections were incubated with hematoxylin for 2 min and eosin for 1 min. The experimental procedures were approved by the Laboratory Animal Care Committee at Xinxiang Medical College (approval no. XXMU-2020-146367).

Immunohistochemistry (IHC) assay

Tissue sections were incubated with primary antibodies against METTL3 (1:50; cat. no. 86132S; Cell Signaling Technology, Inc.), Bcl-2 (1:50; cat. no. ab32124; Abcam) and Ki67 (1:100; cat. no. ab16667; Abcam) at 4°C overnight. Following washing with PBST containing 0.05% Tween-20, the sections were incubated with HRP-conjugated anti-rabbit secondary antibody (1:5,000; cat. no. BM3894; Wuhan Boster Biological Technology, Ltd.) at room temperature for 1 h. The sections were developed with 0.05% 3-diaminobenzidine tetrahydrochloride at room temperature for 10 sec, which was followed by counterstaining with 10% Mayer's hematoxylin for 4 min at room temperature. IHC results were analyzed by two experienced pathologists. A total of five random fields under a light microscope (Leica GmbH DMI4000B) at 200× magnification were chosen to calculate the percentage of positively stained cells vs. the total number of tumor cells. The staining proportion of the positive cells was divided into four groups as follows: -, 0 positive cells observed; +, <30% vs. positive tumor cells observed; ++, 30–60% vs. positive tumor cells observed; and +++: >60% vs. positive tumor cells observed. The - and + groups were classed as METTL3-low expression group, while the ++ and +++ groups were classed as METTL3-high expression group for subsequent analysis. Low- and high-expression groups were combined following scoring and analyzed by Fisher's exact t-test.

Bioinformatics analysis

The Gene Expression Profiling Interactive Analysis (GEPIA) database (gepia.cancer-pku.cn/) was used to evaluate the mRNA expression levels of METTL3 in NSCLC and normal tissue. Log rank test was used for Kaplan-Meier analysis. The Kaplan-Meier plotting tool (kmplot.com/analysis/) was used to evaluate the relapse-free survival of patients with NSCLC expressing low or high levels of METTL3. The association between METTL3 and Bcl-2 mRNA expression levels in NSCLC was analyzed using GEPIA database (gepia.cancer-pku.cn/) and Pearson's correlation coefficient.

Statistical analysis

A total of three independent repeats of experiments were performed. All data are shown as the mean ± SD. Data were analyzed using SPSS software 22.0 (IBM Corp.). Associations between two groups were evaluated using χ2 test and Pearson's correlation analysis. Comparisons between two groups were performed using Student's unpaired two-tailed or paired t-test. Multiple groups were analyzed using one-way ANOVA followed by Bonferroni's post hoc test for pairwise comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

METTL3 expression is elevated in NSCLC tissue and cells

First, it was determined that the expression of METTL3 was upregulated in the NSCLC cohort using bioinformatics tools (gepia.cancer-pku.cn/; Fig. 1A). In addition, RT-qPCR analysis of METTL3 mRNA expression in samples from patients with NSCLC showed that the expression of METTL3 was significantly higher in NSCLC tumor samples than in normal paracancerous tissue (Fig. 1B). m6A quantitative analysis showed that the m6A content level was significantly higher in tumor than in normal paracancerous tissue (Fig. 1C). Western blot and IHC analysis showed that the protein expression of METTL3 was increased in NSCLC compared with normal paracancerous tissue (Fig. 1D and E). Clinical and pathological data are shown in Fig. 1F. A significant positive association between high METTL3 expression and tumor differentiation, tumor stage and metastasis was observed. These results indicated that METTL3 served a significant role in NSCLC development and progression. Consistently, METTL3 expression was increased in the NSCLC cell lines compared with the normal human lung epithelial cell line BEAS-2B (Fig. 1G). Kaplan-Meier survival analysis indicated that patients with tumors with high METTL3 expression had a poor survival rate and outcome (Fig. 1H).

METTL3 knockdown inhibits NSCLC cell viability and migration in vitro

In order to investigate the functional role of METTL3 in NSCLC, METTL3 was knocked down using three independent shRNAs (shRNA-1, shRNA-2 and shRNA-3) in an NSCLC cell line (H1299). Efficient knockdown of METTL3 was confirmed by RT-qPCR (Fig. 2A) and western blotting (Fig. 2B). The CCK-8 assay results showed significantly decreased viability of NSCLC cell lines (H1299, H1975) after silencing METTL3 (Fig. 2C). Moreover, flow cytometry assay revealed that METTL3 silencing accelerated apoptosis in NSCLC cell lines (H1299, H1975; Fig. 2D). Migration assay revealed that METTL3 silencing repressed migration (Fig. 2E). m6A-qPCR validation showed that the percentage of m6A content was significantly decreased when METTL3 was knocked down in H1299 and H1975 cell lines (Fig. 2F). Collectively, these results indicated that silencing METTL3 suppressed viability and migration and promoted apoptosis in NSCLC cell lines.

METTL3 overexpression promotes NSCLC cell viability and migration in vitro

Next, stable METTL3-overexpressing NSCLC cell lines (H1299 and H1975) were constructed by lentiviral infection. Upregulation of METTL3 expression was confirmed by RT-qPCR (Fig. 3A) and western blotting (Fig. 3B). METTL3 overexpression significantly promoted NSCLC cell line (H1299 and H1975) viability, as determined by CCK-8 assay (Fig. 3C). Moreover, flow cytometry revealed that overexpression of METTL3 inhibited the apoptosis of NSCLC cell lines (H1299 and H1975; Fig. 3D). Furthermore, METTL3 overexpression significantly increased migration of NSCLC cell lines in the Transwell assay (Fig. 3E). Quantitative analysis of m6A showed that METTL3 overexpression increased m6A content (Fig. 3F). Based on these results, it was concluded that METTL3 overexpression promoted NSCLC cell viability and migration.

Bcl-2 is a downstream target of METTL3

A previous study reported that Bcl-2 may be mediated by METTL3 in breast cancer (21). In order to identify the underlying molecular mechanisms by which METTL3 promotes NSCLC progression, m6A-seq was performed. The results showed that the GGAC motif was highly enriched within m6A sites in the immunopurified RNA (Fig. 4A). In order to verify that METTL3 targets Bcl-2 mRNA for m6A modification, ethylated RNA immunoprecipitation qPCR was performed to validate the m6A-seq data. The results showed that METTL3 knockdown significantly decreased m6A levels of Bcl-2 mRNA (Fig. 4B). RT-qPCR analysis showed that Bcl-2 expression was significantly decreased at the mRNA level following METTL3 knockdown in H1299 and H1975 cells (Fig. 4C). Bcl-2 protein expression was significantly downregulated in METTL3-knockdown H1299 and H1975 cells (Fig. 4D). Then, the online bioinformatics tool GEPIA database (gepia.cancer-pku.cn/) was used for correlation analysis and our results showed that the METTL3 expression level also showed a positive correlation with Bcl-2 in tumors from individuals with NSCLC (Fig. 4E). Thus, it was concluded that Bcl-2 was a target of METTL3.

METTL3 facilitates NSCLC tumorigenesis by enhancing expression of Bcl-2 in vivo

In order to confirm the potential contribution of METTL3 to tumor progression in vivo, BALB/c nude mice were subcutaneously injected with H1299 cells with METTL3 overexpression or knockdown to develop an NSCLC xenograft model. After 24 days, the tumors were removed, photographed and weighed (Fig. 5A and B). Tumors arising from cells with METTL3 overexpression were significantly larger and heavier than those arising from control cells (Fig. 5C). By contrast, knockdown of METTL3 significantly repressed tumor growth (volume) compared with that of tumors arising from control cells (Fig. 5C). Hematoxylin-eosin staining indicated that knockdown of METTL3 decreased the number of metastatic nodules, while overexpression of METTL3 increased the number of lung metastatic nodules compared with control (Fig. 5D). In order to confirm whether METTL3 promoted tumor growth in vivo by regulating the expression of Bcl-2, IHC staining and western blot analysis were performed. The results indicated that METTL3 overexpression increased Bcl-2 expression and METTL3 knockdown inhibited Bcl-2 expression (Fig. 5E-F). Collectively, these results suggested that METTL3-mediated m6A modification promoted NSCLC progression by enhancing expression of Bcl-2 in vivo.

Discussion

Recently, the role of m6A modification in mRNA translation has emerged as a hot spot in the field of epigenetics (25). Emerging evidence indicates that m6A modification serves important and diverse biological functions in multiple cellular processes, such as RNA synthesis, processing, translation and metabolism (26,27). m6A modification regulates the tumorigenesis of cancers (28,29). For example, in breast cancer tumorigenesis, m6A demethylase ALKBH5 mediates m6A modification in the 3′-UTR of NANOG mRNA (30). Therefore, more detailed work is needed to define the role and underlying mechanisms of m6a modification in certain types of cancer.

Certain studies have demonstrated that METTL3 serves a regulatory role in a variety of cancer types, such as breast, bladder and lung cancer, as well as hepatocellular carcinoma (3135). For example, METTL3 can act as an oncogene in bladder tumorigenesis (36); however, METTL3 and METTL14 also serve as tumor suppressors in hepatocellular carcinoma (37,38). Certain studies have reported that METTL3 contributes to transforming growth factor-β-induced epithelial-mesenchymal transition of lung cancer cells (39). An additional study revealed that there is a significant correlation between METTL3 expression and the occurrence of chronic obstructive pulmonary disease (40). However, the role of METTL3-mediated m6A modification in NSCLC initiation and progression is not fully known. The present study demonstrated that METTL3 expression was significantly upregulated in NSCLC. The role of METTL3 in promoting NSCLC progression was demonstrated both in vitro and in vivo. Bcl-2 was shown to be a direct target of METTL3-mediated m6A modification in NSCLC. Finally, the present findings shed new light on m6A methylation-mediated Bcl-2 overexpression during progression of NSCLC and identified that METTL3 serves an oncogenic role as an m6A ‘writer’ in NSCLC. Thus, METTL3 is a promising therapeutic target for NSCLC treatment.

A recent study indicated that METTL3 regulates expression of various mRNAs in bladder cancer, including MYC, NF-κB and AF4/FMR2 family member 4 (41). Hence, it was hypothesized that METTL3 may promote the translational efficiency of Bcl-2 mRNA to regulate NSCLC progression. Similarly, in breast cancer, METTL3-mediated m6A modification of Bcl-2 promotes its expression (27). Furthermore, METTL3 and Bcl-2 expression levels were positively correlated. Certain studies have revealed that METTL3 overexpression directly mediates m6A methylation and inhibits apoptosis in various types of cancer (42,43). For example, overexpression of Bcl-2 blocks adriamycin-induced apoptosis in bladder cancer cells (44). Another report revealed that high Bcl-2 expression is associated with recurrence and poor survival in gastric cancer (4546).

Taken together, the results of the present study highlight the critical role of METTL3 in NSCLC progression. METTL3 regulated cellular growth, survival and migration in NSCLC. METTL3 promoted NSCLC progression by modulating the level of Bcl-2. This finding may contribute to understanding of the oncogenic role of METTL3, which may be an effective therapeutic target for NSCLC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Henan Provincial Medical Science and Technology Research Joint Co-Construction Project (grant no. LHGJ20190485).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in the Sequence Read Archive repository, accession no. PRJNA721237.

Authors' contributions

The project was designed and conceived by YZ and CD. SL, YZ and TZ analyzed the data. YZ wrote the manuscript. YZ and CD guided the study and revised the manuscript. YZ, SL, TZ and CD confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Ethics approval was granted by the Ethics Committee of the First Affiliated Hospital of Xinxiang Medical University (approval no. KN201808002). Oral consent was obtained from patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Neal RD, Sun F, Emery JD and Callister ME: Lung cancer. BMJ. 365:l17252019. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Reck M and Rabe KF: Precision diagnosis and treatment for advanced non-small-cell lung cancer. N Engl J Med. 377:849–861. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Tandberg DJ, Tong BC, Ackerson BG and Kelsey CR: Surgery versus stereotactic body radiation therapy for stage I non-small cell lung cancer: A comprehensive review. Cancer. 124:667–678. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Wu SG and Shih JY: Management of acquired resistance to EGFR TKI-targeted therapy in advanced non-small cell lung cancer. Mol Cancer. 17:382018. View Article : Google Scholar : PubMed/NCBI

6 

Liu WJ, Du Y, Wen R, Yang M and Xu J: Drug resistance to targeted therapeutic strategies in non-small cell lung cancer. Pharmacol Ther. 206:1074382020. View Article : Google Scholar : PubMed/NCBI

7 

Dai D, Wang H, Zhu L, Jin H and Wang X: N6-methyladenosine links RNA metabolism to cancer progression. Cell Death Dis. 9:1242018. View Article : Google Scholar : PubMed/NCBI

8 

Maity A and Das B: N6-methyladenosine modification in mRNA: Machinery, function and implications for health and diseases. FEBS J. 283:1607–1630. 2016. View Article : Google Scholar : PubMed/NCBI

9 

He L, Li H, Wu A, Peng Y, Shu G and Yin G: Functions of N6-methyladenosine and its role in cancer. Mol Cancer. 18:1762019. View Article : Google Scholar : PubMed/NCBI

10 

Zhou J, Wan J, Gao X, Zhang X, Jaffrey SR and Qian SB: Dynamic m(6)A mRNA methylation directs translational control of heat shock response. Nature. 526:591–594. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, et al: A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 10:93–95. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, Adhikari S, Shi Y, Lv Y, Chen YS, et al: Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 24:177–189. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Roundtree IA, Evans ME, Pan T and He C: Dynamic RNA modifications in gene expression regulation. Cell. 169:1187–1200. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Sun T, Wu R and Ming L: The role of m6A RNA methylation in cancer. Biomed Pharmacother. 112:1086132019. View Article : Google Scholar : PubMed/NCBI

15 

Lin S, Choe J, Du P, Triboulet R and Gregory RI: The m(6)A Methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 62:335–345. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Peng W, Li J, Chen R, Gu Q, Yang P, Qian W, Ji D, Wang Q, Zhang Z, Tang J and Sun Y: Upregulated METTL3 promotes metastasis of colorectal Cancer via miR-1246/SPRED2/MAPK signaling pathway. J Exp Clin Cancer Res. 38:3932019. View Article : Google Scholar : PubMed/NCBI

17 

Wang Q, Chen C, Ding Q, Zhao Y, Wang Z, Chen J, Jiang Z, Zhang Y, Xu G, Zhang J, et al: METTL3-mediated m6A modification of HDGF mRNA promotes gastric cancer progression and has prognostic significance. Gut. 69:1193–1205. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, Tsang LH, Ho DW, Chiu DK, Lee JM, et al: RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 67:2254–2270. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Tao Z, Zhao Y and Chen X: Role of methyltransferase-like enzyme 3 and methyltransferase-like enzyme 14 in urological cancers. PeerJ. 8:e95892020. View Article : Google Scholar : PubMed/NCBI

20 

Travis WD, Brambilla E, Burke AP, Marx A and Nicholson AG: Introduction to the 2015 world health organization classification of tumors of the lung, pleura, thymus and heart. J Thorac Oncol. 10:1240–1242. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Ni WJ and Leng XM: Down-regulated miR-495 can target programmed cell death 10 in ankylosing spondylitis. Mol Med. 26:502020. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Zhang S, Zhao BS, Zhou A, Lin K, Zheng S, Lu Z, Chen Y, Sulman EP, Xie K, Bögler O, et al: m6A Demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell. 31:591–606.e6. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Dominissini D, Moshitch-Moshkovitz S, Salmon-Divon M, Amariglio N and Rechavi G: Transcriptome-wide mapping of N(6)-methyladenosine by m(6)A-seq based on immunocapturing and massively parallel sequencing. Nat Protoc. 8:176–189. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Zaccara S, Ries RJ and Jaffrey SR: Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 20:608–624. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, Wang Y, Li X, Xiong XF, Wei B, et al: RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Mol Cancer. 18:462019. View Article : Google Scholar : PubMed/NCBI

27 

Wang H, Xu B and Shi J: N6-methyladenosine METTL3 promotes the breast cancer progression via targeting Bcl-2. Gene. 22:1440762020. View Article : Google Scholar

28 

Zhang H, Shi X, Huang T, Zhao X, Chen W, Gu N and Zhang R: Dynamic landscape and evolution of m6A methylation in human. Nucleic Acids Res. 48:6251–6264. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Liang Z, Kidwell RL, Deng H and Xie Q: Epigenetic N6-methyladenosine modification of RNA and DNA regulates cancer. Cancer Biol Med. 17:9–19. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Zhang C, Samanta D, Lu H, Bullen JW, Zhang H, Chen I, He X and Semenza GL: Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc Natl Acad Sci USA. 113:E2047–E2056. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Liu ZX, Li LM, Sun HL and Liu SM: Link Between m6A modification and cancers. Front Bioeng Biotechnol. 6:892018. View Article : Google Scholar : PubMed/NCBI

32 

Zeng C, Huang W, Li Y and Weng H: Roles of METTL3 in cancer: Mechanisms and therapeutic targeting. J Hematol Oncol. 13:1172020. View Article : Google Scholar : PubMed/NCBI

33 

Cai X, Wang X, Cao C, Gao Y, Zhang S, Yang Z, Liu Y, Zhang X, Zhang W and Ye L: HBXIP-elevated methyltransferase METTL3 promotes the progression of breast cancer via inhibiting tumor suppressor let-7g. Cancer Lett. 415:11–19. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Lin Y, Wei X, Jian Z and Zhang X: METTL3 expression is associated with glycolysis metabolism and sensitivity to glycolytic stress in hepatocellular carcinoma. Cancer Med. 9:2859–2867. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Wang H, Deng Q, Lv Z, Ling Y, Hou X, Chen Z, Dinglin X, Ma S, Li D, Wu Y, et al: N6-methyladenosine induced miR-143-3p promotes the brain metastasis of lung cancer via regulation of VASH1. Mol Cancer. 18:1812019. View Article : Google Scholar : PubMed/NCBI

36 

Jin H, Ying X, Que B, Wang X, Chao Y, Zhang H, Yuan Z, Qi D, Lin S, Min W, et al: N6-methyladenosine modification of ITGA6 mRNA promotes the development and progression of bladder cancer. EBioMedicine. 47:195–207. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Ma JZ, Yang F, Zhou CC, Liu F, Yuan JH, Wang F, Wang TT, Xu QG, Zhou WP and Sun SH: METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6-methyladenosine-dependent primary MicroRNA processing. Hepatology. 65:529–543. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Liu X, Qin J, Gao T, Li C, Chen X, Zeng K, Xu M, He B, Pan B, Xu X, et al: Analysis of METTL3 and METTL14 in hepatocellular carcinoma. Aging (Albany NY). 12:21638–21659. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Wanna-Udom S, Terashima M, Lyu H, Ishimura A, Takino T, Sakari M, Tsukahara T and Suzuki T: The m6A methyltransferase METTL3 contributes to Transforming Growth Factor-beta-induced epithelial-mesenchymal transition of lung cancer cells through the regulation of JUNB. Biochem Biophys Res Commun. 524:150–155. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Huang X, Lv D, Yang X, Li M and Zhang H: m6A RNA methylation regulators could contribute to the occurrence of chronic obstructive pulmonary disease. J Cell Mol Med. 24:12706–12715. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Cheng M, Sheng L, Gao Q, Xiong Q, Zhang H, Wu M, Liang Y, Zhu F, Zhang Y, Zhang X, et al: The m6A methyltransferase METTL3 promotes bladder cancer progression via AFF4/NF-κB/MYC signaling network. Oncogene. 38:3667–3680. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Yang Z and Jiang X, Li D and Jiang X: HBXIP promotes gastric cancer via METTL3-mediated MYC mRNA m6A modification. Aging (Albany NY). 12:24967–24982. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Xiang S, Liang X, Yin S, Liu J and Xiang Z: N6-methyladenosine methyltransferase METTL3 promotes colorectal cancer cell proliferation through enhancing MYC expression. Am J Transl Res. 12:1789–1806. 2020.PubMed/NCBI

44 

Chui K and Zhang Z: Bcl-2 overexpression inhibits generation of intracellular reactive oxygen species and blocks adriamycin-induced apoptosis in bladder cancer cells. Asian Pac J Cancer Prev. 14:895–901. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Xu G and Tang X: Troxerutin (TXN) potentiated 5-Fluorouracil (5-Fu) treatment of human gastric cancer through suppressing STAT3/NF-κB and Bcl-2 signaling pathways. Biomed Pharmacother. 92:95–107. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Kim R, Emi M, Tanabe K and Toge T: Therapeutic potential of antisense Bcl-2 as a chemosensitizer for cancer therapy. Cancer. 101:2491–502. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2021
Volume 46 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Liu S, Zhao T and Dang C: METTL3‑mediated m6A modification of Bcl‑2 mRNA promotes non‑small cell lung cancer progression Corrigendum in /10.3892/or.2023.8502. Oncol Rep 46: 163, 2021
APA
Zhang, Y., Liu, S., Zhao, T., & Dang, C. (2021). METTL3‑mediated m6A modification of Bcl‑2 mRNA promotes non‑small cell lung cancer progression Corrigendum in /10.3892/or.2023.8502. Oncology Reports, 46, 163. https://doi.org/10.3892/or.2021.8114
MLA
Zhang, Y., Liu, S., Zhao, T., Dang, C."METTL3‑mediated m6A modification of Bcl‑2 mRNA promotes non‑small cell lung cancer progression Corrigendum in /10.3892/or.2023.8502". Oncology Reports 46.2 (2021): 163.
Chicago
Zhang, Y., Liu, S., Zhao, T., Dang, C."METTL3‑mediated m6A modification of Bcl‑2 mRNA promotes non‑small cell lung cancer progression Corrigendum in /10.3892/or.2023.8502". Oncology Reports 46, no. 2 (2021): 163. https://doi.org/10.3892/or.2021.8114