Open Access

Transcriptome changes induced by RUNX3 in cervical cancer cells in vitro

  • Authors:
    • Qian‑Qian Gao
    • Bin Zhou
    • Xiu‑Zhang Yu
    • Zhu Zhang
    • Yan‑Yun Wang
    • Ya‑Ping Song
    • Lin Zhang
    • Hong Luo
    • Ming‑Rong Xi
  • View Affiliations

  • Published online on: November 21, 2019     https://doi.org/10.3892/ol.2019.11128
  • Pages: 651-662
  • Copyright: © Gao 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

Runt‑related transcription factor 3 (RUNX3) is a member of Runt domain family that is known to play key roles in various different types of tumor. It was recently demonstrated that RUNX3 may also be associated with cervical cancer. The aim of the present study was to investigate the potential association between transcriptome changes and RUNX3 expression in cervical cancer. A RUNX3 overexpression model was constructed using cervical cancer cell lines by RUNX3 plasmid transfection. It was demonstrated that the upregulated expression of RUNX3 inhibited proliferation of cervical cancer cell lines, particularly SiHa cells, and was associated with the expression of the IL‑6, PTGS2, FOSL1 and TNF genes. In addition, it was revealed that the TNF and FoxO pathways may also be affected by RUNX3. Therefore, the expression of the RUNX3 gene may be involved in the occurrence and progression of cervical cancer.

Introduction

Cervical cancer was reported as the most common gynecological tumor in developing countries in 2012 (1). It was also estimated that 527,600 women were first diagnosed with cervical cancer and 265,700 women succumbed to this disease worldwide in the same year (1). In recent years, cervical cytology screening and vaccination have been used to prevent cervical cancer development due to human papillomavirus (HPV) infection (2). However, thousands of women are still affected by cervical cancer, with detrimental effects on their quality of life (3). Alongside the increased amount of research that makes the molecular mechanism underlying cervical cancer more clear, particularly with the knowledge that HPV and E6 oncoproteins potentially affect the progress of cervical cancer, an increasing number of genes have been demonstrated to be implicated in cervical cancer (4,5). Identifying new molecular targets for the prevention and treatment of cervical cancer would further improve the quality of life of the patients.

Runt-related transcription factor 3 (RUNX3), along with RUNX1 and RUNX2, are members of the Runt domain family (6). RUNX3 is involved in the transforming growth factor β (TGF-β) signaling pathway, which is a key downstream effector affecting the progression of tumors (7). In a number of previous studies on gastric, hepatocellular and breast cancer, RUNX3 has been revealed to play a significant role in tumor suppression (811). Recent research has demonstrated that RUNX3 may also act as a tumor suppressor gene in cervical cancer (12). RUNX3 expression may be suppressed by promoter hypermethylation, gene deletions, inactivating mutations and protein mislocalization (8,11,1316). By contrast, other researchers concluded that RUNX3 acts as an oncogene in skin, ovarian and head and neck cancer, where its expression level was observed as significantly increased (1720). In addition, Lotem et al (21) hypothesized that this gene plays important roles in immunity and inflammation, and may affect the development of epithelial tumors.

In a previous study, it was observed that the polymorphisms of RUNX3 may be associated with cervical cancer, and the mRNA expression of RUNX3 was significantly different between the cervical cancer group and the healthy female subjects (22). However, the specific molecular mechanism through which RUNX3 regulates cancer-associated signaling pathways and affects tumorigenesis remains elusive. The aim of the present study was to investigate the effect of RUNX3 on cervical cancer cell lines, and to identify the transcriptome changes in cervical cancer.

Materials and methods

Cell culture

SiHa, HeLa and C33A cells were obtained from the Laboratory of Molecular Translational Medicine, West China Institute of Women and Children's Health, Key Laboratory of Obstetric & Gynecological and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University (Sichuan, China). SiHa is an HPV-16 infected cervical cancer cell line, HeLa is an HPV-18 infected cervical cell line, and C33A is an HPV-negative cervical cancer cell line. The cells were cultured in DMEM high-glucose medium at 37°C in 5% CO2, supplemented with 10% fetal bovine serum and penicillin/streptomycin (50 U/ml) (both Thermo Fisher Scientific, Inc.). There was no contamination of SiHa and C33A cells by HeLa cells.

Construction of plasmids, RNA interference and transfection

Homo sapiens RUNX3 mRNA sequence (23) was used to design and identify the RUNX3 overexpression model and empty plasmid (plasmid EX-NEG-M98; Guangzhou FulenGen Co., Ltd.), was used as the control; in addition, RUNX3 short-hairpin RNA (shRNA) was designed and identified (Guangzhou FulenGen Co., Ltd.). The following base pairs of shRNA (shRNA31, shRNA32, shRNA33 and shRNA34) and the non-targeting sequence control shRNA (shRNA001) were used for RUNX3 gene interference (Table I). After the single clones were obtained, the extracted plasmids were identified and sequenced via digestion, following the instructions of endotoxin-free plasmid extraction kit (Tiangen Biotech Co., Ltd.). Briefly, 8 ml buffer P1 with RNase A was added to the collected bacteria and vortexed. Subsequently, 8 ml buffer P2 was added, and the samples were shaken gently and left to stand at 25°C for 5 min. This was repeated with buffer P4, after which the samples were centrifuged at 8,228 × g for 10 min at 25°C. Following which, 0.3 of the volume of isopropanol and 2.5 ml buffer BL was added, and the samples were transferred to CP6 columns and centrifuged at 8,228 × g for 2 min at 25°C, twice. A total of 10 ml buffer PW with absolute ethanol was added and the samples were centrifuged at 8,228 g for 2 min at 25°C, twice. Subsequently, cervical cancer cells were transfected with RUNX3 overexpression plasmid or shRNA (0.5 µg plasmid) by X-tremeGene HP DNA Transfection Reagent (Roche Diagnostics). In the present study, cells with RUNX3 overexpression were referred to as ‘ORF’ RUNX3 cells, the empty plasmid was the control of ORF RUNX3 cells as ‘NEG’ control cells, and those with RUNX3 shRNA as ‘shRNA’ RUNX3 cells. In addition, the control groups were treated only with transfection reagent in SiHa, HeLa and C33A cells, respectively which were referred to as ‘Control’, and without any intervention as ‘cell line name’ (baseline controls). At 24, 48 and 72 h after transfection, the cells were harvested and prepared for subsequent experiments.

Table I.

shRNAs used in the present study.

Table I.

shRNAs used in the present study.

Clone nameSymbolChromosome locationLength5′-3′
HSH021393-31-LVRH1GP(OS503663)RUNX381521 GGCAATGACGAGAACTACTCC
HSH021393-32-LVRH1GP(OS545091)RUNX315921 GGAATCCAAATTCTTGGGTAC
HSH021393-33-LVRH1GP(OS503664)RUNX3289521 GGTCTCTTACAGGTATAGTTC
HSH021393-34-LVRH1GP(OS545092)RUNX3385821 GGGATAGTAAATAAATTGCTC
CSHCTR001-1-LVRH1GP(OSNEG20) 19 GCTTCGCGCCGTAGTCTTA

[i] shRNA, short hairpin RNA; RUNX3, runt-related transcription factor 3.

RNA isolation and reverse transcription-quantitative PCR (RT-qPCR) analysis

Total RNA was extracted from cervical cancer cells and purified using TRIzol® reagent according to the manufacturer's protocol (Thermo Fisher Scientific, Inc.). RT-PCR was performed using a One-Step RT-PCR kit (Bioneer), according to the manufacturer's protocol. RT-qPCR was performed using the SYBR Green PCR Master Mix (Roche Diagnostics). The samples of all genes were amplified in a thermocycler as follows: 95°C for 10 min (1 cycle), 95°C for 15 sec and 60°C for 1 min (48 cycles). The primer sequences are presented in Table II. Data were normalized against β-actin expression with the comparative quantification cycle method. Triplicate Cq values were averaged and the relative expression levels were determined as 2−ΔΔCq (24).

Table II.

Primer sequences used in the present study.

Table II.

Primer sequences used in the present study.

GeneGene IDForward (5′-3′)Reverse (5′-3′)Product size, bp
TNCNM_002160.3 TCGCTACAAGCTGAAGGTGG GTTAACGCCCTGACTGTGGT214
PTGS2NM_000963.3 CAAATTGCTGGCAGGGTTGC AGGGCTTCAGCATAAAGCGT139
ICAM1NM_000201.2 ATGGCAACGACTCCTTCTCG GCCGGAAAGCTGTAGATGGT142
TNF SF10NM_001190942.1 TGCGTGCTGATCGTGATCTT TCTTGGAGTCTTTCTAACGAGC234
IL6NM_000600.3 TTCAATGAGGAGACTTGCCTGG CTGGCATTTGTGGTTGGGTC206
IL7RNM_002185.3 TGAAATATGTGGGGCCCTCG GTCATTGGCTCCTTCCCGAT223
FOSL1NM_005438.4 AGCCCAGCAGAAGTTCCAC CCTCTTCCTCCGGGCTGAT227
IL32NM_001308078 AGACAGTGGCGGCTTATTATGAGG GCCTCGGCACCGTAATCC  86
TGF-βNM_000660.4 TATCGACATGGAGCTGGTGAAG CAGCTTGGACAGGATCTGGC  67
β-actinNM_001101.3 TGACGTGGACATCCGCAAAG CTGGAAGGTGGACAGCGAGG205
WST-1 measurement and flow cytometry

WST-1 measurement was used to detect cell viability using a WST-1 cell proliferation and cytotoxicity assay kit (Boster Biological Technology Co., Ltd.) according to the manufacturer's protocol. Briefly, the cervical cancer cells were seeded at a density of 104 cells/ml into 96-well plates and incubated overnight at 37°C. Subsequently, the plasmids were transfected into cancer cells for 24/48/72 h 3 times, as aforementioned. WST-1 (10 µl) was added, followed by incubation for a further 2 h at 37°C. In order to exclude the effect of the WST-1 reagent, the same concentrations of transfection reagent were added to the cells directly when the WST-1 measurement was performed. The absorbance of cells was monitored at 450 nm.

Apoptosis was also analyzed via flow cytometry. First, the transfected tumor cells in each group were lysed with trypsin without EDTA (HyClone; GE Healthcare Life Sciences), and the cells were stained using the FITC Annexin V Apoptosis Detection kit with propidium iodide (PI; both BestBio, http://bestbio.bioon.com.cn/). Finally, for the cell cycle analysis, cells in each group were stained with PI and then analyzed by flow cytometry (Guava® easyCyte™), using nCyte v2.7 software (both EMD Millipore; Merck KGaA).

Transcriptome sequencing

A total of 3 µg RNA from each sample was used, which was prepared for input material of the RNA samples. Sequencing libraries were generated, using the NEBNext® Ultra™ RNA Library Prep kit for Illumina® (New England Biolabs) according to the manufacturer's protocol. The index codes were added to assign sequences to each sample. Briefly, mRNA was purified from total RNA using poly-T oligo-attached magnetic beads NEBNext® Ultra™ Directional RNA Library Prep kit for Illumina® (New England Biolabs, Inc.) according to the manufacturer's protocol. Fragmentation was then performed using divalent cations at increasing temperatures of 25°C for 10 min, 42°C for 15 min, and 70°C for 15 min after which the samples were held at 4°C, in NEBNext First-Strand Synthesis Reaction Buffer (5X) (New England Biolabs, Inc.). Finally, the PCR products were purified (AMPure XP system), and the library quality was assessed on the Agilent Bioanalyzer 2100 system. According to the manufacturer's instructions, clustering of the index-coded samples was performed on a cBot Cluster Generation System, which used a TruSeq PE Cluster kit v3-cBot-HS (Illumina, Inc.). The library preparations were then sequenced on an Illumina Hiseq 2000/2500 platform, and 100/50 bp single-end reads were generated. Gene Ontology (GO; http://geneontology.org) enrichment analysis of differentially expressed genes was implemented by the GOseq R package (clusterProfiler v3.4.4; http://bioconductor.org/), in which gene length bias was corrected. GO terms with corrected P<0.05 were considered significantly enriched by differentially expressed genes. The Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.jp/kegg) is a database resource for understanding high-level functions and utilities of the biological system, such as the cell, the organism and the ecosystem, from molecular-level information, particularly from large-scale molecular datasets generated by genome sequencing and other high-throughput experimental technologies. The present study used clusterProfiler v3.4.4 software (http://bioconductor.org/) in order to test the statistical enrichment of differentially expressed genes in the KEGG pathways. Through this analysis, different genes were identified as potential regulators of cervical cancer, which may be downstream or upstream of RUNX3.

Statistical analysis

GraphPad Prism software (version 5.01; GraphPad Software, Inc.) was used for the data analysis and to assess the normal distribution and equal variance of all data. The baseline characteristics of the participants were assessed by Student's t-test and single-factor Pearson's χ2 test. Only the difference between two groups were evaluated by Student's t test. Differences among multiple groups were evaluated by the one-way ANOVA, followed by Bonferroni's multiple comparisons test. The RUNX3 mRNA expression levels were compared between transfected cells, or between groups of cervical cancer cells and controls using Bonferroni's multiple comparisons test. P<0.05 was considered to indicate a statistically significant difference. According to transcriptome sequencing, differential expression analysis of two groups was performed using the DESequencing (DESeq) R package (v1.10.1; http://www.bioconductor.org/). DESeq provides statistical analysis for determining differential expression in digital gene expression data using a model based on the negative binomial distribution. The resulting P-values were adjusted using the Benjamini and Hochberg's approach for controlling the false discovery rate. Genes with an adjusted P-value (q value) found by DESeq were classified as differentially expressed. q<0.05 was considered to indicate a statistically significant difference.

Results

Expression of RUNX3 in cervical cancer lines

The efficiency of exogenous RUNX3 expression and RUNX3 shRNA in cervical cancer cells was verified via RT-qPCR analysis. The mRNA levels of RUNX3 were markedly higher in ORF RUNX3 cells compared with the NEG control group of the three cervical cancer cell lines (P<0.001; Fig. 1), which were also extremely significantly higher than the ‘Control’ and ‘cell name’ groups (P<0.001; Fig. 1). It also demonstrated the low expression levels of RUNX3 in all the control groups, with or without intervention. In addition, the shRNA RUNX3 and control groups (shRNA001) were significantly different in the three cell lines, particularly in SiHa cells (P<0.001 and P<0.05, respectively; Fig. 2). As the RUNX3 gene was successfully suppressed by shRNA34 in SiHa, HeLa and C33A cells, particularly the SiHa cell line at 24 (P<0.001) and 48 h (P<0.05), shRNA34 was selected as the interference plasmid for subsequent experiments (Fig. 2).

Role of RUNX3 in proliferation and apoptosis of cervical cancer cells

The WST-1 results suggested that there were significant differences among ORF and NEG plasmid transfection groups and shRNA34 and shRNA001 groups in the SiHa cell line (P<0.05; Fig. 3A), However, the shRNA34 and shRNA001, and ORF and NEG groups exhibited less prominent differences in HeLa and C33A cell lines, respectively, although the results were still significant; (Fig. 3B and C). There was no significant difference between the groups of cervical cancer cells with WST-1 or without WST-1 (Fig. 3). In addition, the results revealed that there were significant differences at different transfection times; the ORF and NEG groups were significantly different at 24, 48, 72 and 96 h in the SiHa cell line, the shRNA34 and shRNA001 groups were significantly different at 72 and 96 h in the HeLa cell line and the ORF and NEG groups were significantly different at 24, 48 and 96 h in the C33A cell line (P<0.05). The results also demonstrated that the effects on different cervical cancer cells varied and indicated that RUNX3 may inhibit the proliferation of cervical cancer cells.

PI/Annexin V flow cytometry analysis was performed in the present study in order to detect the apoptosis rates in RUNX3-transfected cell lines. The apoptosis rates of different cervical cancer cell groups were significantly different, especially in SiHa cells (Fig. 4). The apoptosis rate of the ORF group was significantly higher compared with that in the control group in SiHa cell at 24, 48 and 72 h (P<0.001), indicating an apoptosis-promoting role of RUNX3 in cervical cancer. Furthermore the apoptosis rate of the shRNA34 group was significantly higher compared with that in the control group at 24, 48 and 72 h, which were inconsistent as expected (P<0.001, Fig. 4). In addition, the apoptosis ratios of HeLa and C33A cells were not significantly higher in the ORF group compared with that in the control group at 24 and 48 h, which were not consistent with those of the SiHa cells in ORF and control group, but in the 48 and 72 h time periods, shRNA34 group was significantly higher compared with that in the control group, in both the HeLa and C33A cells, which is consistent with the SiHa cells (P<0.05; Fig. 4). Finally the apoptosis ratios of ORF group were significantly lower compared with that in the shRNA34 group at 48 h in SiHa, HeLa and C33A cell lines (P<0.05; Fig. 4), and at 72 h in SiHa and HeLa cell lines (P<0.05; Fig. 4). The results may suggest the low expression of RUNX3 in cervical cancer is complexity to interfere and further research should be preceded.

Effects of RUNX3 on cervical cancer cells by transcriptome sequencing

The molecular mechanisms through which RUNX3 may affect cervical cancer were investigated in SiHa cells in vitro. Relative transcript levels were tested by transcriptome sequencing in order to determine the difference in transcripts mediated by RUNX3 in SiHa cells. The present study identified 31 genes that were differentially expressed in RUNX3-overexpressing SiHa cells. A total of 9 genes showed differences in the three groups simultaneously, including IL-6, PTGS2, FOSL1, TNC, ICAM1, IL-7R, IL-32, TGF-β and TNFSF10 (Figs. 5 and 6), which may be potential regulators of cervical cancer combined with RUNX3. The mRNA expressions of the 9 genes confirmed the results of the transcriptome sequencing (Fig. 7), although further research is required to clarify the connection between RUNX3 and these genes. However, in the shRNA34 groups, except with the TNC and IL-6 gene, the expression levels of these genes were not changed, which may be due to the low expression of RUNX3 mRNA in cervical cancer and limited interference of shRNA34. Additional research is required in order to develop an improved understanding. Furthermore, following verification of the mRNA expression levels, there was no significant difference between the control groups. In addition, the GO enrichment analysis of biological processes revealed that certain genes were enriched, such as those involved in the regulation of carboxypeptidase, exopeptidase and hydrolase activity. Furthermore, the GO enrichment analyses of the KEGG pathways concluded that these genes were enriched in signaling pathways such as the tumor necrosis factor (TNF) pathway, Forkhead box O (FoxO) pathway, African trypanosomiasis and malaria (Fig. 8). Transcription factors and proto-oncogenes, such as HMGA1 and FOSL1, IL7R and MALAT1, were demonstrated as likely to be associated with RUNX3 in cervical cancer (Fig. 6) and may be associated with microfollicular thyroid adenoma, various benign mesenchymal tumors and renal cell carcinoma (25,26). Finally, the transcriptome sequencing analysis results indicated that two long non-coding RNAs, RP11-54O7.3 and MALAT1, were significantly different between groups, suggesting that RUNX3 may affect their expression level (Fig. 6). These findings indicate that the expression of RUNX3 may affect the expression of other genes, which may also be associated with cervical cancer in vitro.

Discussion

Cervical cancer has been confirmed to be associated with HPV infection; furthermore 0.7% of women initially found to be infected with high-risk HPV will develop invasive carcinomas within 3 years (2,3). In recent years, immunological mechanisms and genetic factors have been demonstrated to play critical roles in cervical cancer (4,2730). A previous study demonstrated that RUNX3 is likely a potential gene involved in cervical cancer susceptibility, and was associated with the type of HPV infection and cervical intraepithelial neoplasia progression (22). In the present study, three types of cervical cancer cell lines were selected: SiHa, HeLa and C33A, infected by HPV-16, HPV-18 or not infected, respectively. It was recently reported that RUNX3 may play a key role in the development and progression of cervical cancer (12). It was also observed in the present study that RUNX3 may inhibit cervical cancer cell proliferation, particularly in the SiHa cell lines, indicating its potential function as a tumor suppressor; inconsistencies in these results may be due to the different cell lines. In addition, the expression levels of RUNX3 were lower in the three cell lines, which were almost the same level in the control groups with or without any intervention. The molecular mechanisms and signaling pathways underlying the role of RUNX3 in cervical cancer remain elusive. The aim of the present study was to elucidate the potential effects of RUNX3 on cervical cancer by transcriptome sequencing, which may help improve the current therapy options and prognosis of patients with cervical cancer.

It was previously demonstrated that the tumorigenicity of human gastric cancer cell lines was significantly associated with the level of RUNX3 in nude mice, which suggested that suppression of RUNX3 function was directly associated with the occurrence and progression of human gastric cancer (9). In a previous study on ovarian cancer, the researchers observed that the overexpression of RUNX3 in A2780s cells rendered them more resistant to carboplatin, whereas the sensitivity of A2780cp cells to carboplatin increased significantly following inhibition of RUNX3 (31). In the present study, the upregulated expression of RUNX3 consistently inhibited the proliferation and promoted the apoptosis of cervical cancer cells, particularly in the SiHa cell line. The results were similar to those of a recent study on cervical cancer, which reported that upregulated expression of RUNX3 inhibited the proliferation, migration and invasion of cervical cancer cells (12). According to these results, it may be concluded that RUNX3 acts as a tumor suppressor gene in cervical cancer (12).

As the mechanisms of action of RUNX3 have not yet been fully elucidated, the molecular mechanisms and signaling pathways of RUNX3 have become a focus in cancer research. It is generally recognized that the transcription factor RUNX3, which is a key effector of the TGF-β signaling pathway, acts on the TGF-β receptor, thereby promoting cell proliferation and apoptosis through the TGF-β signal transduction pathway (7,10,3234). This may explain its wide involvement in tumorigenesis (35). However, several other signaling pathways are affected by RUNX3. The Wnt signaling pathway was confirmed to be associated with RUNX3 in intestinal tumorigenesis (36) through the formation of a ternary complex with β-catenin/TCF4 and attenuation of Wnt signaling activity, whereas the inactivation of RUNX3 may promote intestinal adenoma formation (36). In addition, the mitochondria-mediated pathway has been demonstrated to be associated with RUNX3, inducing apoptosis in gastric cancer cells (37). In a study on hepatocellular carcinoma (HCC), the researchers reported that RUNX3 suppressed Notch signaling in HCC SMMC7721 cells (38). In the present study, the TNF and FoxO signaling pathways were demonstrated to be associated with RUNX3.

TNF is a member of the TNF superfamily of cytokines, which mediates cell processes such as differentiation, inflammation, proliferation and apoptosis (39), and has double the effect in cancer cells. More specifically, it has been reported that TNF is associated with cervical cancer (40). In addition, the TNF-α/TNFR1/NF-κB pathway is potentially implicated in tongue cancer and lung metastasis (41). The NF-κB signaling axis (defined by the interactions of NF-κB dimers, IκB regulators and IKK complexes) is responsive to external stimuli and signal received (42). FoxO is a subfamily member of the forkhead transcription factor family. FoxO has been revealed as a key determinant of cell fate, and to play an important functional role as a tumor suppressor in different types of cancer (43,44). During apoptosis, FoxO is involved in mitochondria-dependent and -independent processes that trigger the expression of death receptor ligands, such as Fas ligand, TNF apoptosis ligand, Bcl XL, bNIP3 and Bim (43,44). The most important pathway associated with FoxO is the PI3K/AKT pathway. The PI3K/AKT pathway is also dysregulated and activated in a wide variety of cancers, such as breast, thyroid and cervical cancers (43). In addition, the Ras/MEK/ERK, IKK and AMPK pathways have also been demonstrated to be associated with FoxO, and they may play a role in tumorigenesis (4345). However, the role of FoxO in cervical cancer has not been extensively investigated. An association between FoxO and TGF-β in tumors has also been reported (46,47). In an HCC study, the Thr32 residue of FoxO3 was proven to be a critical factor for TGF-β-induced apoptosis, which was mediated via Bim (47). Due to the complexity and uncertainty of the associations between RUNX3 and signaling pathways, further research is required in order to confirm the association between RUNX3 and the TNF/FoxO pathway in cervical cancer.

Recently, the associations of lncRNAs with RUNX3 in different cancers were investigated. A previous study identified a potential competing endogenous RNA regulatory network involving MT1JP and the regulation of RUNX3 expression and progression of gastric cancer (48). In a study of human colorectal cancer, miR-532-5p mimic was revealed to markedly downregulate the mRNA and protein levels of RUNX3, potentially acting as an oncogenic miRNA (49). Finally, the present study demonstrated that RUNX3 may affect the expression levels of RP11-54O7.3 and MALAT1. According to previous reports, MALAT1 may participate in tumor formation, such as lung, prostate and ovarian cancer (5053). It was recently reported that overexpression of MALAT1 could sponge miR-429 and regulate cervical cancer pathogenesis in vivo and in vitro (54), while the associations between MALAT1 and RUNX3 remain unclear. However, the role of RP11-54O7.3 has not yet been fully elucidated, and the association between RP11-54O7.3 and RUNX3 remains unknown (55,56). These results indicate that RUNX3 may affect the expression of lncRNAs, which may be associated with cervical cancer in vitro. The specific mechanisms of action and role of RUNX3 in cervical cancer will be further investigated in future studies.

In conclusion, the present study demonstrated that RUNX3 inhibited proliferation and promoted the apoptosis of cervical cancer cells. In addition, the TNF and FoxO pathways were demonstrated to be affected by RUNX3, and the effects of MALAT1 and RP11-54O7.3 are likely mediated by RUNX3 in cervical cancer. However, further research is required in order to achieve an improved understanding of the molecular complexities and functions of RUNX3 in cervical cancer.

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant nos. 81572573 and 81172440), and the New Bud Research Foundation of West China Second University Hospital (grant no. kx091), Sichuan University.

Availability of data and materials

All data generated or analyzed during the present study are included in this published article.

Authors' contributions

XY, ZZ and YS contributed to analysis and manuscript preparation. QG and MX performed the data analyses and wrote the manuscript. BZ, YW, LZ and HL helped perform the analysis with constructive discussions.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Berkhof J, de Bruijne MC, Zielinski GD and Meijer CJ: Natural history and screening model for high-risk human papillomavirus infection, neoplasia and cervical cancer in the Netherlands. Int J Cancer. 115:268–275. 2005. View Article : Google Scholar : PubMed/NCBI

3 

McCredie MR, Sharples KJ, Paul C, Baranyai J, Medley G, Jones RW and Skegg DC: Natural history of cervical neoplasia and risk of invasive cancer in women with cervical intraepithelial neoplasia 3: A retrospective cohort study. Lancet Oncol. 9:425–434. 2008. View Article : Google Scholar : PubMed/NCBI

4 

González-Herrera L, Rodríguez-Morales P, Gonza Lez-Losa Mdel R, Pérez-Mendoza G, Canul-Canché J, Rosado-López I and Cetina TC: MTHFR/p53 polymorphisms as genetic factors for cervical intraepithelial neoplasia and cervical cancer in HPV-infected Mexican women. Int J Biol Markers. 29:e142–e149. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Kuglik P, Kasikova K, Smetana J, Vallova V, Lastuvkova A, Moukova L, Cvanova M and Brozova L: Molecular cytogenetic analyses of hTERC (3q26) and MYC (8q24) genes amplifications in correlation with oncogenic human papillomavirus infection in Czech patients with cervical intraepithelial neoplasia and cervical carcinomas. Neoplasma. 62:130–139. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Lund AH and van Lohuizen M: RUNX: A trilogy of cancer genes. Cancer Cell. 1:213–215. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Ito Y and Miyazono K: RUNX transcription factors as key targets of TGF-beta superfamily signaling. Curr Opin Genet Dev. 13:43–47. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Hwang KT, Han W, Bae JY, Hwang SE, Shin HJ, Lee JE, Kim SW, Min HJ and Noh DY: Downregulation of the RUNX3 gene by promoter hypermethylation and hemizygous deletion in breast cancer. J Korean Med Sci. 22 (Suppl):S24–S31. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Li QL, Ito K, Sakakura C, Fukamachi H, Inoue K, Chi XZ, Lee KY, Nomura S, Lee CW, Han SB, et al: Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell. 109:113–124. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Subramaniam MM, Chan JY, Yeoh KG, Quek T, Ito K and Salto-Tellez M: Molecular pathology of RUNX3 in human carcinogenesis. Biochim Biophys Acta. 1796:315–331. 2009.PubMed/NCBI

11 

Xiao WH and Liu WW: Hemizygous deletion and hypermethylation of RUNX3 gene in hepatocellular carcinoma. World J Gastroenterol. 10:376–380. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Li Z, Fan P, Deng M and Zeng C: The roles of RUNX3 in cervical cancer cells in vitro. Oncol Lett. 15:8729–8734. 2018.PubMed/NCBI

13 

Gao F, Huang C, Lin M, Wang Z, Shen J, Zhang H, Jiang L and Chen Q: Frequent inactivation of RUNX3 by promoter hypermethylation and protein mislocalization in oral squamous cell carcinomas. J Cancer Res Clin Oncol. 135:739–747. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Ito K, Liu Q, Salto-Tellez M, Yano T, Tada K, Ida H, Huang C, Shah N, Inoue M, Rajnakova A, et al: RUNX3, a novel tumor suppressor, is frequently inactivated in gastric cancer by protein mislocalization. Cancer Res. 65:7743–7750. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Kim WJ, Kim EJ, Jeong P, Quan C, Kim J, Li QL, Yang JO, Ito Y and Bae SC: RUNX3 inactivation by point mutations and aberrant DNA methylation in bladder tumors. Cancer Res. 65:9347–9354. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Lau QC, Raja E, Salto-Tellez M, Liu Q, Ito K, Inoue M, Putti TC, Loh M, Ko TK, Huang C, et al: RUNX3 is frequently inactivated by dual mechanisms of protein mislocalization and promoter hypermethylation in breast cancer. Cancer Res. 66:6512–6520. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Kudo Y, Tsunematsu T and Takata T: Oncogenic role of RUNX3 in head and neck cancer. J Cell Biochem. 112:387–393. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Tsunematsu T, Kudo Y, Iizuka S, Ogawa I, Fujita T, Kurihara H, Abiko Y and Takata T: RUNX3 has an oncogenic role in head and neck cancer. PLoS One. 4:e58922009. View Article : Google Scholar : PubMed/NCBI

19 

Lee JH, Pyon JK, Kim DW, Lee SH, Nam HS, Kang SG, Kim CH, Lee YJ, Chun JS and Cho MK: Expression of RUNX3 in skin cancers. Clin Exp Dermatol. 36:769–774. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Häfner N, Steinbach D, Jansen L, Diebolder H, Dürst M and Runnebaum IB: RUNX3 and CAMK2N1 hypermethylation as prognostic marker for epithelial ovarian cancer. INT J Cancer. 138:217–228. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Lotem J, Levanon D, Negreanu V, Bauer O, Hantisteanu S, Dicken J and Groner Y: Runx3 at the interface of immunity, inflammation and cancer. Biochim Biophys Acta. 1855:131–143. 2015.PubMed/NCBI

22 

Gao QQ, Zhou B, Yu XZ, Zeng X, Zhang Z, Quan Y, Wang YY, Pu Y, Cheng P, Song YP, et al: RUNX3 polymorphisms and the susceptibility to cervical cancer and cervical intraepithelial neoplasia in Western China. Int J Clin Exp Pathol. 9:10617–10626. 2016.

23 

Lee JW, Kim DM, Jang JW, Park TG, Song SH, Lee YS, Chi XZ, Park IY, Hyun JW, Ito Y and Bae SC: RUNX3 regulates cell cycle-dependent chromatin dynamics by functioning as a pioneer factor of the restriction-point. Nat Commun. 10:18972019. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Dal Cin P, Fusco A, Belge G, Chiappetta G, Fedele M, Pauwels P, Bullerdiek J and Van den Berghe H: Involvement of the HMGI(Y) gene in a microfollicular adenoma of the thyroid. Genes Chromosomes Cancer. 24:286–289. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Hirata H, Hinoda Y, Shahryari V, Deng G, Nakajima K, Tabatabai ZL, Ishii N and Dahiya R: Long Noncoding RNA MALAT1 promotes aggressive renal cell carcinoma through Ezh2 and interacts with miR-205. Cancer Res. 75:1322–1331. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Quan Y, Zhou B, Wang Y, Duan R, Wang K, Gao Q, Shi S, Song Y, Zhang L and Xi M: Association between IL17 polymorphisms and risk of cervical cancer in Chinese women. Clin Dev Immunol. 2012:258–293. 2012. View Article : Google Scholar

28 

Walch-Rückheim B, Mavrova R, Henning M, Vicinus B, Kim YJ, Bohle RM, Juhasz-Böss I, Solomayer EF and Smola S: Stromal fibroblasts induce CCL20 through IL6/C/EBPβ to support the recruitment of Th17 cells during cervical cancer progression. Cancer Res. 75:5248–5259. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Zidi S, Sghaier I, Gazouani E, Mezlini A and Yacoubi-Loueslati B: Evaluation of Toll-like receptors 2/3/4/9 gene polymorphisms in cervical cancer evolution. Pathol Oncol Res. 22:323–330. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Zidi S, Stayoussef M, Gazouani E, Mezlini A, Yacoubi-Loueslati B and Almawi WY: Relationship of common vascular endothelial growth factor polymorphisms and haplotypes with the risk of cervical cancer in Tunisians. Cytokine. 74:108–112. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Barghout SH, Zepeda N, Vincent K, Azad AK, Xu Z, Yang C, Steed H, Postovit LM and Fu Y: RUNX3 contributes to carboplatin resistance in epithelial ovarian cancer cells. Gynecol Oncol. 138:647–655. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Yano T, Ito K, Fukamachi H, Chi XZ, Wee HJ, Inoue K, Ida H, Bouillet P, Strasser A, Bae SC and Ito Y: The RUNX3 tumor suppressor upregulates Bim in gastric epithelial cells undergoing transforming growth factor beta-induced apoptosis. Mol Cell Biol. 26:4474–4488. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Yamamura Y, Lee WL, Inoue K, Ida H and Ito Y: RUNX3 cooperates with FoxO3a to induce apoptosis in gastric cancer cells. J Biol Chem. 281:5267–5276. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Chi XZ, Yang JO, Lee KY, Ito K, Sakakura C, Li QL, Kim HR, Cha EJ, Lee YH, Kaneda A, et al: RUNX3 suppresses gastric epithelial cell growth by inducing p21(WAF1/Cip1) expression in cooperation with transforming growth factor {beta}-activated SMAD. Mol Cell Biol. 25:8097–8107. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Farooqi AA, Khalid S and Ahmad A: Regulation of cell signaling pathways and miRNAs by resveratrol in different cancers. Int J Mol Sci. 19(pii): E6522018. View Article : Google Scholar : PubMed/NCBI

36 

Ito K, Lim AC, Salto-Tellez M, Motoda L, Osato M, Chuang LS, Lee CW, Voon DC, Koo JK, Wang H, et al: RUNX3 attenuates beta-catenin/T cell factors in intestinal tumorigenesis. Cancer Cell. 14:226–237. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Nagahama Y, Ishimaru M, Osaki M, Inoue T, Maeda A, Nakada C, Moriyama M, Sato K, Oshimura M and Ito H: Apoptotic pathway induced by transduction of RUNX3 in the human gastric carcinoma cell line MKN-1. Cancer Sci. 99:23–30. 2008.PubMed/NCBI

38 

Gao J, Chen Y, Wu KC, Liu J, Zhao YQ, Pan YL, Du R, Zheng GR, Xiong YM, Xu HL and Fan DM: RUNX3 directly interacts with intracellular domain of Notch1 and suppresses Notch signaling in hepatocellular carcinoma cells. Exp Cell Res. 316:149–157. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Holdbrooks AT, Britain CM and Bellis SL: ST6Gal-I sialyltransferase promotes tumor necrosis factor (TNF)-mediated cancer cell survival via sialylation of the TNF receptor 1 (TNFR1) death receptor. J Biol Chem. 293:1610–1622. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Zidi S, Stayoussef M, Zouidi F, Benali S, Gazouani E, Mezlini A and Yacoubi-Loueslati B: Tumor necrosis factor alpha (−238/-308) and TNFRII–VNTR (−322) polymorphisms as genetic biomarkers of susceptibility to develop cervical cancer among tunisians. Pathol Oncol Res. 21:339–345. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Tanaka T, Imamura T, Yoneda M, Irie A, Ogi H, Nagata M, Yoshida R, Fukuma D, Kawahara K, Shinohara M and Nakayama H: Enhancement of active MMP release and invasive activity of lymph node metastatic tongue cancer cells by elevated signaling via the TNF-α-TNFR1-NF-κB pathway and a possible involvement of angiopoietin-like 4 in lung metastasis. Int J Oncol. 49:1377–1384. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Mitchell S, Vargas J and Hoffmann A: Signaling via the NFκB system. Wiley Interdiscip Rev Syst Biol Med. 8:227–241. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Farhan M, Wang H, Gaur U, Little PJ, Xu J and Zheng W: foxo signaling pathways as therapeutic targets in cancer. Int J Biol Sci. 13:815–827. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Coomans de Brachène A and Demoulin JB: FOXO transcription factors in cancer development and therapy. Cell Mol Life Sci. 73:1159–1172. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Link W and Fernandez-Marcos PJ: FOXO transcription factors at the interface of metabolism and cancer. Int J Cancer. 141:2379–2391. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Yadav H, Devalaraja S, Chung ST and Rane SG: TGF-β1/Smad3 pathway targets PP2A-AMPK-FoxO1 signaling to regulate hepatic gluconeogenesis. J Biol Chem. 292:3420–3432. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Zhao X, Liu Y, Du L, He L, Ni B, Hu J, Zhu D and Chen Q: Threonine 32 (Thr32) of FoxO3 is critical for TGF-β-induced apoptosis via Bim in hepatocarcinoma cells. Protein Cell. 6:127–138. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Xu Y, Zhang G, Zou C, Zhang H, Gong Z, Wang W, Ma G, Jiang P and Zhang W: LncRNA MT1JP suppresses gastric cancer cell proliferation and migration through MT1JP/MiR-214-3p/RUNX3 axis. Cell Physiol Biochem. 46:2445–2459. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Zhang J, Zhou W, Liu Y, Liu T, Li C and Wang L: Oncogenic role of microRNA-532-5p in human colorectal cancer via targeting of the 5′UTR of RUNX3. Oncol Lett. 15:7215–7220. 2018.PubMed/NCBI

50 

Xue D, Lu H, Xu HY, Zhou CX and He XZ: Long noncoding RNA MALAT1 enhances the docetaxel resistance of prostate cancer cells via miR-145-5p-mediated regulation of AKAP12. J Cell Mol Med. 22:3223–3237. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Bai L, Wang A, Zhang Y, Xu X and Zhang X: Knockdown of MALAT1 enhances chemosensitivity of ovarian cancer cells to cisplatin through inhibiting the Notch1 signaling pathway. Exp Cell Res. 366:161–171. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Fang Z, Chen W, Yuan Z, Liu X and Jiang H: LncRNA-MALAT1 contributes to the cisplatin-resistance of lung cancer by upregulating MRP1 and MDR1 via STAT3 activation. Biomed Pharmacother. 101:536–542. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Xiping Z, Bo C, Shifeng Y, Feijiang Y, Hongjian Y, Qihui C and Binbin T: Roles of MALAT1 in development and migration of triple negative and Her-2 positive breast cancer. Oncotarget. 9:2255–2267. 2017.PubMed/NCBI

54 

Shen F, Zheng H, Zhou L, Li W and Xu X: Overexpression of MALAT1 contributes to cervical cancer progression by acting as a sponge of miR-429. J Cell Physiol. 234:11219–11226. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Li J, Wang W, Xia P, Wan L, Zhang L, Yu L, Wang L, Chen X, Xiao Y and Xu C: Identification of a five-lncRNA signature for predicting the risk of tumor recurrence in patients with breast cancer. Int J cancer. 143:2150–2160. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Jiang W, Zhan H, Jiao Y, Li S and Gao W: A novel lncRNA-miRNA-mRNA network analysis identified the hub lncRNA RP11-159F24.1 in the pathogenesis of papillary thyroid cancer. Cancer Med. 7:6290–6298. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 19 Issue 1

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
Gao QQ, Zhou B, Yu XZ, Zhang Z, Wang YY, Song YP, Zhang L, Luo H and Xi MR: Transcriptome changes induced by RUNX3 in cervical cancer cells in vitro. Oncol Lett 19: 651-662, 2020
APA
Gao, Q., Zhou, B., Yu, X., Zhang, Z., Wang, Y., Song, Y. ... Xi, M. (2020). Transcriptome changes induced by RUNX3 in cervical cancer cells in vitro. Oncology Letters, 19, 651-662. https://doi.org/10.3892/ol.2019.11128
MLA
Gao, Q., Zhou, B., Yu, X., Zhang, Z., Wang, Y., Song, Y., Zhang, L., Luo, H., Xi, M."Transcriptome changes induced by RUNX3 in cervical cancer cells in vitro". Oncology Letters 19.1 (2020): 651-662.
Chicago
Gao, Q., Zhou, B., Yu, X., Zhang, Z., Wang, Y., Song, Y., Zhang, L., Luo, H., Xi, M."Transcriptome changes induced by RUNX3 in cervical cancer cells in vitro". Oncology Letters 19, no. 1 (2020): 651-662. https://doi.org/10.3892/ol.2019.11128