IMR Press / FBL / Volume 28 / Issue 9 / DOI: 10.31083/j.fbl2809199
Open Access Original Research
N6-methyladenosine Methyltransferase METTL3 Enhances PTGER2 Expression to Increase Ovarian Cancer Stemness and Chemoresistance
Show Less
1 Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, 350014 Fuzhou, Fujian, China
2 Department of Radiation Oncology, Fujian Medical University Union Hospital, 350001 Fuzhou, Fujian, China
3 Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), 350001 Fuzhou, Fujian, China
4 Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), 350001 Fuzhou, Fujian, China
*Correspondence: linyibin@fjmu.edu.cn (Yi-Bin Lin); benhuaxu@163.com (Ben-Hua Xu)
Front. Biosci. (Landmark Ed) 2023, 28(9), 199; https://doi.org/10.31083/j.fbl2809199
Revised: 7 April 2023 | Accepted: 9 May 2023 | Published: 14 September 2023
(This article belongs to the Special Issue Diagnostic, Prognostic, and Therapeutic Biomarkers for Ovarian Cancer)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Ovarian cancer is the second leading cause of gynecologic cancer-associated deaths. Cancer stemness and chemoresistance are responsible for ovarian cancer metastasis and the poor prognosis of patients. In this study, we determined the function of N6-methyladenine (m6A) RNA methylation and prostaglandin E receptor 2 (PTGER2) in ovarian cancer progression. Methods: The m6A RNA methylation-associated PTGER2 in ovarian cancer was identified using bioinformatics analysis. The role of PTGER2 in ovarian cancer was elucidated in cell lines and clinical samples with cellular and molecular experiments. Results: In this investigation, bioinformatics analysis based on a public cancer database was used to elucidate the impact of m6A modification on the prognosis of patients with ovarian cancer. Moreover, PTGER2 was identified as a potential oncogene associated with the distant metastasis of ovarian cancer and poor patient prognosis. Interestingly, PTGER2 expression was experimentally shown to be enhanced by N6-adenosine-methyltransferase 70 kDa subunit (METTL3)-mediated m6A modification. In addition, PTGER2 enhanced cancer stem cell self-renewal properties, the epithelial-mesenchymal transition, and DNA damage repair, thus potentiating cell stemness, therapy resistance to carboplatin, proliferation, and metastasis of ovarian cancer. Importantly, PTGER2 expression in clinical samples was associated with distant metastasis, predicted poor patient prognosis, and independently served as a prognostic predictor in ovarian cancer. Conclusions: Our work defines PTGER2 as an oncogene and reveals that PTGER2 is a prognostic predictor and novel therapeutic target for the management of ovarian cancer.

Keywords
N6-methyladenosine
PTGER2
ovarian cancer
stemness
chemoresistance
1. Introduction

Ovarian cancer is characterized by a high prevalence and high mortality rate and ranks as the second leading factor causing gynecologic cancer-related deaths in women worldwide [1]. Platinum-based chemotherapy is still the main treatment for advanced ovarian cancer [2]. Although ovarian cancer mortality has declined in recent years [3], ovarian cancer stemness, chemoresistance, and metastasis are common reasons for treatment failure and poor patient prognosis [4]. Therefore, identifying novel targets to overcome these tumorigenic properties is now an urgent priority for this disease.

N6-methyladenine (m6A) RNA methylation is the most abundant RNA modification type in non-coding RNAs and mRNAs and can regulate downstream effectors to affect ovarian cancer progression [5]. The m6A RNA methylation process is mediated by many proteins including methyltransferases, demethylases, and m6A readers [6]. Previous studies have suggested that m6A regulator-mediated methylation modification patterns can affect cancer progression [7, 8]. Copy number variation (CNV) gains are associated with an increase in gene expression, whereas CNV deletions are correlated with a decrease in gene expression. Moreover, advanced-stage cancers harbor more CNV events of m6A regulators [9], and patients with CNV gain of N6-adenosine-methyltransferase 70 kDa subunit (METTL3) have a poor prognosis [10]. METTL3 functions as a methyltransferase and plays an oncogenic role in ovarian cancer [11, 12]. However, the dysregulation of downstream effectors of these m6A regulators remains largely undetermined.

Prostaglandin E2 receptor (subtype EP2), namely PTGER2, belongs to the G protein-coupled receptor superfamily. The activation of PTGER2 results in the increased activity of cAMP-dependent signaling [13]. Previous studies have revealed that PTGER2 acts as an oncogene in colorectal cancer [14] and prostate cancer [15] and serves as a tumor suppressor in neuroblastoma [16]. However, the biological role and clinical significance of PTGER2 and the mechanism of PTGER2 dysregulation in ovarian cancer remain unclear.

Here, we elucidated the role of m6A modification in ovarian cancer and identified specific genes modulated by m6A regulators using a public cancer database. In particular, we focused on the mechanism by which METTL3-mediated m6A modification of PTGER2 mRNA results in its upregulation, and explored the impact of these aberrant interactions on ovarian cancer cell stemness, chemoresistance, proliferation, and metastasis. Our work further suggests the potential application of PTGER2 as a reliable prognostic predictor and a novel therapeutic target for ovarian cancer treatment by using ovarian cancer cell lines and clinical samples.

2. Materials and Methods
2.1 Bioinformatics Analysis

Bioinformatics analysis was performed according to our previous study [17]. The genomic data, mRNA expression profiles, and clinical data of The Cancer Genome Atlas (TCGA) ovarian serous cystadenocarcinoma (OV) database were downloaded for conducting differential analyses, survival analyses, correlation analyses, unsupervised clustering analyses, gene set variation analysis (GSVA), and gene set enrichment analysis (GSEA). The gene set representing platinum drug resistance was constructed using data obtained from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database as previously reported [18]. Based on the best cut-off value obtained from survival analysis, samples were assigned as low and high expression groups.

2.2 Cell Cultures

Ovarian cancer cell lines OV90 and SKOV3 with short tandem repeat verification were purchased from American Type Culture Collection (Manassas, VA, USA) and cultured in Dulbecco’s Modified Eagle Medium (DMEM) harboring 10% fetal bovine serum (FBS) at 37 °C with 5% CO2. It was confirmed that the cell lines did not have mycoplasma contamination.

2.3 Cell Transfections

Single hairpin RNA (shRNA) negative control and shRNA targeting PTGER2 (sh-PTGER2) were constructed and inserted into lentiviral particles by GeneChem Corporation (Shanghai, China). Small interfering RNA (siRNA) against METTL3 (si-METTL3) was synthesized by RiboBio Corporation (Guangzhou, China). Polybrene and Lipofectamine 3000 were used for lentivirus particle transfection and siRNA transduction, respectively. After transfection or transduction for 48–72 h, ovarian cancer cells were subjected to further investigation.

2.4 RT-PCR and Quantitative PCR

Total RNAs of ovarian cancer cells were used for reverse transcription with a kit purchased from TaKaRa Corporation (Dalian, China). RT-PCR and quantitative PCR (qPCR) were conducted with the generated cDNAs and specific primers. Bio-Rad GelDoc XR+ (Hercules, CA, USA) was applied to capture images for RT-PCR, and the 2-ΔΔCt method was used to determine the relative expression of mRNA for qPCR.

2.5 Western Blot Analysis

Total proteins of ovarian cancer cells in lysis buffer were quantified by the BCA method as previously described in our investigation [19]. Then the separated and transferred proteins were incubated with antibodies against the following proteins: METTL3, PTGER2, Myc, cyclin D1 (CCND1), vimentin, and β-actin. The chemiluminescence method was adopted for protein detection, and the Bio-Rad GelDoc XR+ was applied to record images.

2.6 Methylated RNA Immunoprecipitation

The Methylated RNA Immunoprecipitation (MeRIP) m6A Kit purchased from RiboBio Corporation (Guangzhou, China) was used to conduct MeRIP as previously described in our investigation [8]. Briefly, total RNAs of ovarian cancer cells were used to generate fragments that were pulled down with magnetic beads harboring the m6A antibody. Enriched RNA was purified and used for RT-PCR and qPCR.

2.7 Luciferase Reporter Assay

PmirGLO luciferase reporter vectors (Promega Co., Madison, WI, USA) containing wild-type or mutant 3’-untranslated region (UTR) of PTGER2 were transfected into ovarian cancer cells. After transfection for 48 h, the luciferase activity in cells was measured to evaluate the effects of METTL3 on PTGER2 transcriptional levels on the BioTek Luminometer Synergy H1 (Winooski, VT, USA) using the Dual-Luciferase Reporter Assay System (Promega, Madison, WI, USA).

2.8 Tumorsphere Formation Assay

Ovarian cancer cells seeded on 6-well ultra-low-attachment plates were incubated in serum-free DMEM/F12 (20 ng/mL epidermal growth factor, 20 ng/mL fibroblast growth factor, and 2% B27). The images of tumorspheres were captured, and the number of tumorspheres was counted after a 14-day culture. Three continuous generations of tumorspheres number were calculated for analysis.

2.9 Immunofluorescence

Ovarian cancer cells seeded on coverslips were treated with paraformaldehyde (4%) for fixation, incubated with Triton X-100 (0.2%) for permeabilization, and subjected to incubation with antibodies against the following proteins: cluster of differentiation 44 (CD44), CD133, and gamma-H2A histone family member X (γH2AX). DAPI was used to co-stain the cells, and the images of stained cells were captured with a fluorescence confocal microscope.

2.10 Colony Formation Assay

To detect cell proliferation and chemoresistance, ovarian cancer cells were subjected to treatment with carboplatin for 6 h at the indicated concentrations and then seeded in wells for 10 days. The generated colonies were subjected to fixation, staining, and recording for analysis.

2.11 Transwell Assays

The migration and invasion ability of ovarian cancer cells were detected by transwell assays. For the migration assay, serum-free cells were seeded in the upper chamber of a transwell without Matrigel coating. For the invasion assay, serum-free cells were plated in the upper chamber of a transwell with Matrigel coating. DMEM containing 10% FBS was added to the lower chambers of a transwell were for both the migration and invasion assays. The migrated and invaded cells were subjected to fixation, staining, and recording for analysis.

2.12 Patient Tissues

One hundred and fifty-eight ovarian cancer specimens described in our previous study [19] were collected for analysis. All specimens had a pathological diagnosis. Patient written consent and ethics approval from the Ethics Committee of the hospital were obtained (K2020-036-01). All procedures strictly adhered to the policies approved by the Institutional Ethics Committee and Declaration of Helsinki.

2.13 Immunohistochemistry

Immunohistochemistry (IHC) and staining evaluation were carried out as previously described [19]. Paraffin-fixed sections from ovarian cancer specimens were deparaffinized and rehydrated before antigen retrieval in citrate buffer. Then the sections were subjected to the eradication of endogenous peroxidase activity and blocking of non-specific antigens. After incubation with anti-PTGER2 antibody, the sections were treated with Diaminobenzidine substrate and subjected to the evaluation of staining intensities. For statistical analysis, a score of 7–12 was assigned as high expression, and a score of 0–6 was considered low expression.

2.14 Statistical Analyses

SPSS 22.0 (IBM SPSS Inc., Chicago, IL, USA) or RStudio 9.0 was applied for analysis of the data expressed as the mean ± standard deviation from three independent experiments. The Wilcoxon rank-sum test and Student’s two-tailed t-test were used to test the statistical significance between two groups. One-way analysis of variance was applied to reveal the statistical significance among multiple groups. Spearman’s rank correlation test and Chi-square test were applied for the correlation analyses. Survival analysis was performed by plotting Kaplan-Meier survival curves with log-rank tests. Univariate and multivariate Cox regression models were applied to identify the correlation between PTGER2 expression and the overall survival of patients with ovarian cancer. Statistical significance was achieved with p < 0.05.

3. Results
3.1 Bioinformatics Analyses Indicate the Important Role of m6A RNA Methylation in Ovarian Cancer Progression

To explore the role of m6A regulators in ovarian cancer progression, we adopted 9 m6A writers (including Cbl Proto-Oncogene Like 1 [CBLL1], METTL3/14/16, RNA-binding motif protein 15/15B [RBM15/15B], Vir-like m6A methyltransferase associated, WT1-associated protein [WTAP], and zinc finger CCCH-type containing 13 [ZC3H13]), 2 m6A erasers (AlkB homolog 5, RNA demethylase [ALKBH5] and fat mass and obesity-associated protein), and 15 m6A readers (including eukaryotic translation initiation factor 3 subunit A, ELAV like RNA-binding protein 1 [ELAVL1], fragile X messenger ribonucleoprotein 1 [FMR1], heterogeneous nuclear ribonucleoprotein A2/B1 [hnRNPA2B1], hnRNPC, insulin-like growth factor 2 mRNA-binding protein 1/2/3 [IGF2BP1/2/3], leucine rich pentatricopeptide repeat containing, YTH domain-containing protein 1/2 [YTHDC1/], YTHDF1/2/3 and RNA-binding motif protein X-linked), which are dysregulated in human cancers. Since the deletion or amplification of gene copy number affects the corresponding gene expression and is regarded as the trigger of cancer development [8], we applied bioinformatics analyses based on TCGA OV database to identify CNVs in these 26 m6A regulators and found that 23 m6A regulators contained CNVs in ovarian cancer (Supplementary Fig. 1). Then the 23 CNV-carrying genes in TCGA database were used to continue our analysis of the clinical significance of these m6A regulators in ovarian cancer. Gene interaction network analysis suggested that the expression of ALKBH5, CBLL1, ELAVL1, FMR1, HNRNPA2B1, HNRNPC, IGF2BP2, METTL14, METTL16, METTL3, YTHDC2, YTHDF2, and WTAP was correlated with the overall survival of patients with ovarian cancer and indicated a strong correlation among the expression of 23 m6A regulators (Supplementary Fig. 2A), suggesting these dysregulated m6A regulators as contributors to ovarian cancer development and progression.

To further examine how the 23 m6A regulators exert their clinical significance in ovarian cancer, we performed unsupervised clustering and identified two distinct patterns of m6A modification in TCGA OV patients according to the expression of 23 m6A regulators (Supplementary Fig. 2B). Survival analyses showed the superior prognosis of patients with ovarian cancer in cluster A patterns than in cluster B patterns (Supplementary Fig. 2C). Differently enriched Gene Ontology (GO) and KEGG terms between these two clusters were elucidated by conducting GSVA. The analysis verified that cluster B was involved in RNA processing (Supplementary Fig. 2D) and cancer-associated signaling pathways (Supplementary Fig. 2E), such as the Notch and Wnt pathways, as well as DNA replication, cell cycle, base excision repair, nucleotide excision repair, homologous recombination, non-homologous end joining, and mismatch repair compared to cluster A patterns, further indicating the critical role of m6A modification in ovarian cancer progression.

3.2 PTGER2 Expression is Associated with Aberrant m6A RNA Methylation and Functions as a Tumorigenic Gene in Ovarian Cancer

To identify specific genes that could be affected by m6A RNA methylation in ovarian cancer, we carried out differential expression analyses to compare the identified cluster A with cluster B and elucidated 492 differentially expressed genes (fold change 2.0, p < 0.05). Then we integrated and analyzed the differential expression data with another database to identify specific genes participating in ovarian cancer progression. First, we calculated the index of platinum drug resistance by GSVA in TCGA OV patients and performed correlation analyses to elucidate genes associated with chemoresistance (Geneset A). Furthermore, we conducted survival analyses to reveal genes correlated with the prognosis of TCGA OV patients (|r| 2.0, p < 0.05) (Geneset B). In addition, we extracted genes potentially modified by m6A RNA methylation as suggested by the m6A-Atlas database with experimental evidence (Geneset C). Interestingly, the intersection of these three genesets only identified PTGER2 as an overlapping gene (Fig. 1A).

Fig. 1.

PTGER2 expression serves as an m6A RNA methylation-mediated oncogene in ovarian cancer as per The Cancer Genome Atlas (TCGA) database. (A) Venn diagram showing genes correlated with chemoresistance, m6A modification, and patient prognosis in ovarian cancer. (B–H) The associations among Myc, CD44, CD133, L1CAM, p53, CCND1, vimentin, and PTGER2 expression. (I,J) Gene set enrichment analysis (GSEA) of PTGER2 showing enrichment of dysregulated pathways and processes in Hallmark (Top 30) and Kyoto Encyclopedia of Genes and Genomes (KEGG) (Top 30). (K) Comparison of PTGER2 expression in ovarian cancer patients with or without distant metastasis. (L) The Kaplan-Meier curve delineating the overall survival of patients with ovarian cancer based on PTGER2 expression.

Subsequent correlation analyses revealed a positive correlation between PTGER2 expression and the expression of Myc, CD44, CD133, L1 cell adhesion molecule (CAM), CCND1, and vimentin, which serve as stemness, proliferation, and epithelial-mesenchymal transition (EMT)-associated markers, and showed a negative association between PTGER2 expression and expression of the DNA damage repair-associated marker p53 (Fig. 1B–H). Moreover, GSEA confirmed that PTGER2 positively participated in oncogenic processes and pathways such as DNA repair, E2F targets, EMT, Myc targets, Wnt/β-catenin signaling, CAMs, focal adhesion, and the extracellular matrix receptor interaction (Fig. 1I,J). Importantly, PTGER2 expression was upregulated in ovarian cancer patients with distant metastasis compared to those without distant metastasis (Fig. 1K), and high PTGER2 expression predicted poor patient prognosis in TCGA (Fig. 1L). These findings collectively suggest PTGER2 as a potential oncogene in ovarian cancer.

3.3 METTL3-Mediated m6A Modification Elevates PTGER2 Expression

Based on the findings of the above-mentioned bioinformatics analyses, we further investigated whether PTGER2 expression is modulated by m6A modification. PTGER2 was potentially modified by METTL3, METTL14, and IGF2BP1 in the m6A2Target database (http://m6a2target.canceromics.org/). Since our above survival analysis suggests METTL3 as a risk factor for the prognosis of ovarian cancer patients and PTGER2 serves as a potential oncogene, we explored the impact of METTL3 on PTGER2 and found that silencing METTL3 downregulated the mRNA and protein levels of PTGER (Fig. 2A–C). Moreover, m6A individual-nucleotide-resolution cross-linking and immunoprecipitation and m6A MeRIP-sequencing data collected in the RMVar database (http://rmvar.renlab.org) revealed three potential m6A modification sites in the coding sequence (CDS) and 3 UTR of PTGER mRNA (Fig. 2D). Considering the location relationship of these three sites, we synthesized two pairs of primers covering site 1, and sites 2 and 3, respectively. Furthermore, m6A MeRIP analyses validated that PTGER mRNA was immunoprecipitated by anti-m6A antibody and could be detected by qPCR with the primers covering sites 2 and 3, but not site 1 (Supplementary Fig. 3). Moreover, the decrease in m6A modification of PTGER mRNA was detected in METTL3-silenced ovarian cancer cells (Fig. 2E,F). To further elucidate the site responsible for m6A modification of PTGER mRNA, we constructed plasmids harboring mutant sites 2 or site 3 (Fig. 2G). Luciferase reporter assays further indicated that METTL3 knockdown can inhibit the transcription of wild-type PTGER, whereas mutation at site 2 but not at site 3 abolished this suppression (Fig. 2H). These findings demonstrate that METTL3-mediated m6A RNA methylation contributes to controlling PTGER2 expression in ovarian cancer.

Fig. 2.

METTL3 is responsible for m6A modification of PTGER2 mRNA. (A) quantitative PCR (qPCR) analysis determining the efficiency of METTL3 knockdown in OV90 and SKOV3 cells. (B) qPCR analysis showing the effect of METTL3 knockdown on PTGER2 mRNA expression in OV90 and SKOV3 cells. (C) Western blot analysis suggesting the impact of METTL3 knockdown on PTGER2 protein expression in OV90 and SKOV3 cells. (D) Bioinformatics analysis identifying m6A modification sites within the CDS and 3’UTR regions of PTGER2 mRNA. (E,F) m6A MeRIP assays measuring the influence of METTL3 knockdown on m6A modification of PTGER2 mRNA in OV90 and SKOV3 cells. The enriched RNAs were further subjected to qPCR (E) and RT-PCR (F). (G) The sequences of wild-type and mutant m6A modification sites. (H) Luciferase reporter assays revealing the impact of METTL3 depletion on the post-transcriptional inhibition of PTGER2 in OV90 and SKOV3 cells. ***p < 0.001. NC, negative control.

3.4 PTGER2 Knockdown Inhibits Ovarian Cancer Stemness, Chemoresistance, Proliferation, and Metastasis

According to the results obtained from the above bioinformatics analyses, we determined the impact of PTGER2 on ovarian cancer cell stemness, chemoresistance, proliferation, and metastasis. First, shRNA targeting PTGER2 (sh-PTGER2) was used for PTGER2 silencing in ovarian cells. Then tumorsphere formation and immunofluorescence assays showed that PTGER2 knockdown suppressed the stemness of ovarian cancer cells and impaired the expression of stem cell markers CD44 and CD133 in ovarian cancer cells (Fig. 3A–C). Colony formation assays indicated that PTGER2 depletion inhibited the carboplatin resistance and proliferation of ovarian cancer cells (Fig. 3D,E), and transwell assays measured the inhibition of migration and invasion by PTGER2 knockdown in ovarian cancer cells (Fig. 3F,G). Furthermore, immunofluorescence assays detected the upregulation of γH2AX by PTGER2 depletion in ovarian cancer cells treated with carboplatin, indicating enhanced DNA damage (Fig. 3H). Moreover, the expression of cell stemness, proliferation, and EMT-associated proteins including Myc, CCND1, and vimentin were impaired in PTGER2-silenced ovarian cancer cells (Fig. 3I). These findings demonstrate that PTGER2 functions as an oncogene by stimulating cell stemness, chemoresistance, proliferation, and metastasis.

Fig. 3.

PTGER2 increases ovarian cancer stemness, chemoresistance, proliferation, and metastasis. (A–G) Tumorsphere formation (A,B), immunofluorescence (scale bar: 12.5 µm) (C), Clonogenic (D,E), transwell (Scale bar: 50 µm) (F,G) assays were adopted for detecting the stemness, chemoresistance, proliferation, and metastasis of PTGER2-depleted OV90 and SKOV3 cells and the controls. (H) Immunofluorescence analysis (scale bar: 8 µm) was applied to measure γH2AX expression in PTGER2-depleted OV90 cultured with carboplatin, PTGER2-depleted SKOV3 cells cultured with carboplatin, and the controls. (I) Western blot analysis showing the expression of proteins correlated with stemness and chemoresistance (Myc), proliferation (CCND1), and metastasis (vimentin) in PTGER2-silenced OV90 and SKOV3 cells and the controls. **p < 0.01, ***p < 0.001 (compared with the control group). ##p < 0.01, ###p < 0.001 (compared with the carboplatin group). NC, negative control.

3.5 PTGER2 Expression is Associated with the CLinical and Pathological Characteristics of Patients with Ovarian Cancer

To gain further knowledge of the vital role of PTGER2 in the progression of ovarian cancer, we performed IHC in 158 ovarian cancer tissues. IHC showed the different levels of PTGER2 protein expression in the detected ovarian cancer samples (Fig. 4A), and PTGER2 protein expression was upregulated in ovarian cancer samples with metastasis compared to those without metastasis (p = 0.021) (Fig. 4B, Supplementary Table 1). However, PTGER2 protein expression had no significant correlation with other clinical and pathological characteristics. Survival analysis was performed and revealed that high PTGER2 protein expression conferred poor overall survival for patients with ovarian cancer (p = 0.009, log-rank test) (Fig. 4C). Univariate analyses were subsequently conducted and elucidated the positive associations between high PTGER2 protein expression, old age, advanced T classification, advanced N classification, distant metastasis, and poor overall survival of patients with ovarian cancer (Fig. 4D). Multivariate analyses were performed and revealed that PTGER2 protein expression (HR = 1.095, 95% confidence interval [CI] 1.061–1.180, p = 0.027), T classification, and lymph node metastasis served as independent and unfavorable prognostic factors for patients with ovarian cancer (Fig. 4E).

Fig. 4.

PTGER2 expression predicts the prognosis of ovarian cancer patients. (A) Images representing the differential PTGER2 expression in ovarian cancer tissues (scale bar: 125 µm). (B) Images representing the differential PTGER2 expression between ovarian cancer patients with or without distant metastasis (scale bar: 125 µm). (C) Kaplan-Meier curve displaying the overall survival of patients with ovarian cancer based on PTGER2 protein levels. (D) Univariate analysis was used to correlate PTGER2 protein levels, clinicopathological parameters, and overall survival of patients with ovarian cancer. (E) Multivariate analysis was conducted to correlate PTGER2 protein levels, clinicopathological parameters, and overall survival of patients with ovarian cancer.

4. Discussion

Developing new biomarkers for cancer diagnosis, treatment, and prognosis evaluation are the key issues in cancer research [20, 21]. Stemness and chemoresistance are responsible for proliferation and metastasis and are regarded as contributors to poor prognosis of patients with ovarian cancer [19]. m6A modification plays a key role in cancer development and progression by regulating many biological and pathological processes including cell stemness, drug resistance, proliferation, and metastasis [22]. In this investigation, notable links between m6A modification and ovarian cancer progression were identified using bioinformatics analyses. In addition, our study suggested that METTL3-mediated m6A RNA methylation modulated the expression of PTGER2, which subsequently promoted ovarian cancer stemness, chemoresistance, proliferation, and metastasis by affecting the expression of associated key regulators. Importantly, high PTGER2 expression predicted the poor overall survival of patients with ovarian cancer.

Distinct m6A modification patterns can affect the prognosis of cancer patients [7]. In this work, we conducted bioinformatics analyses and established two m6A modification patterns according to 23 CNV-harboring m6A regulators. The expression of 23 m6A regulators was affected by DNA CNV, suggesting the significance of their gene expression in cancer progression [23]. The aberrant expression of these m6A regulators together with their prognostic role in ovarian cancer further revealed the importance of m6A modification in cancer progression. Based on the vital value of m6A modification in ovarian cancer, we further identified PTGER2 as an oncogene potentially modified by m6A modification in a subsequent investigation.

Previous studies have shown that METTL3 promotes ovarian cancer proliferation, migration, and invasion [24, 25]. In addition, METTL3 is an inducer of cancer stem cell self-renewal and chemoresistance in human cancers [26, 27]. In this work, the regulatory impact of m6A RNA methylation on the expression of PTGER2, which served as a promoter of ovarian cancer stemness, chemoresistance, proliferation, and metastasis, was experimentally verified by the observation that METTL3 enhanced PTGER2 expression through m6A modification, further suggesting the oncogenic role of METTL3 in ovarian cancer. Comprehensive analysis has suggested that METTL3 mainly regulates m6A RNA methylation in the CDS region, stop codon, and 3’ UTR of its targets [28]. In this investigation, we also found that METTL3 modulated m6A RNA methylation of PTGER2 in the 3’ UTR region, thus enhancing PTGER2 expression. However, some experts and scholars have proposed that m6A-sensitive PCR and MeRIP mapping of specific mRNA do not provide definitive evidence of m6A. Therefore, additional studies are needed to confirm the m6A RNA methylation of PTGER2 mRNA.

PTGER2 plays dual roles as an oncogene or a tumor suppressor in human cancers [13, 15, 16]. PTGER2 facilitates cancer stemness, chemoresistance, proliferation, and metastasis [14, 29]. In the present work, we also revealed the role of PTGER2 in promoting ovarian cancer stemness, chemoresistance, proliferation, and metastasis. Cancer stem cell self-renewal properties contribute to regulating the EMT process and DNA damage repair [30]. The facilitation of cancer chemoresistance can be regulated by several mechanisms, including modulation of cancer stemness, DNA damage repair, and the EMT [31]. In our investigation, we further discovered that PTGER2 increased cancer stem cell self-renewal properties, the EMT, and DNA damage repair to enhance cell stemness, resistance to carboplatin, proliferation, and metastasis, thus potentiating ovarian cancer progression. Moreover, bioinformatics analysis of TCGA ovarian cancer samples and immunohistochemical assays in our collected clinical samples confirmed that PTGER2 functioned as an oncogene and was correlated with ovarian cancer distant metastasis. Importantly, PTGER2 expression was identified as an independent and unfavorable factor in patient tissues, further supporting the conclusions drawn from ovarian cancer cells.

5. Conclusions

Overall, our study provides a new regulatory axis consisting of METTL3 and PTGER2 in the modulation of ovarian cancer progression. METTL3-mediated m6A RNA methylation of PTGER2 is responsible for the oncogenic role of PTGER2 in ovarian cancer. This work delineates the role of m6A RNA methylation in a specific network of signal transduction and highlights the foundation for the clinical translation of PTGER2 both as a prognostic predictor and as a novel target for the management of ovarian cancer.

Availability of Data and Materials

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

Author Contributions

YL and BX conceived and designed the present study. YL wrote the manuscript. BX checked and revised the manuscript. Both authors contributed to the article and approved the submitted version. Both authors read and approved the final manuscript. Both authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Patient written consent and ethics approval from the Ethics Committee of the hospital were obtained (K2020-036-01).

Acknowledgment

Not applicable.

Funding

This work was founded by Startup Fund for scientific research, Fujian Medical University (Grant number: 2021QH1141).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Lheureux S, Braunstein M, Oza AM. Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA: A Cancer Journal for Clinicians. 2019; 69: 280–304.
[2]
Zhou D, Liu J, Liu R, Li H, Huang Y, Ma D, et al. Effectiveness and Safety of Niraparib as Neoadjuvant Therapy in Advanced Ovarian Cancer With Homologous Recombination Deficiency (NANT): Study Protocol for a Prospective, Multicenter, Exploratory, Phase 2, Single-Arm Study. Frontiers in Oncology. 2022; 12: 852772.
[3]
Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA: A Cancer Journal for Clinicians. 2023; 73: 17–48.
[4]
Roy L, Cowden Dahl KD. Can Stemness and Chemoresistance Be Therapeutically Targeted via Signaling Pathways in Ovarian Cancer? Cancers. 2018; 10: 241.
[5]
Chang LL, Xu XQ, Liu XL, Guo QQ, Fan YN, He BX, et al. Emerging role of m6A methylation modification in ovarian cancer. Cancer Cell International. 2021; 21: 663.
[6]
Zhao Y, Shi Y, Shen H, Xie W. m6A-binding proteins: the emerging crucial performers in epigenetics. Journal of Hematology & Oncology. 2020; 13: 35.
[7]
Zhang B, Wu Q, Li B, Wang D, Wang L, Zhou YL. m6A regulator-mediated methylation modification patterns and tumor microenvironment infiltration characterization in gastric cancer. Molecular Cancer. 2020; 19: 53.
[8]
Lin X, Wang F, Chen J, Liu J, Lin YB, Li L, et al. N6-methyladenosine modification of CENPK mRNA by ZC3H13 promotes cervical cancer stemness and chemoresistance. Military Medical Research. 2022; 9: 19.
[9]
Li W, Gao Y, Jin X, Wang H, Lan T, Wei M, et al. Comprehensive analysis of N6-methylandenosine regulators and m6A-related RNAs as prognosis factors in colorectal cancer. Molecular Therapy. Nucleic Acids. 2021; 27: 598–610.
[10]
Wang X, Yu J, Chen J, Hou Y, Du Z, Huang H, et al. Copy number variation analysis of m6 A regulators identified METTL3 as a prognostic and immune-related biomarker in bladder cancer. Cancer Medicine. 2021; 10: 7804–7815.
[11]
Hua W, Zhao Y, Jin X, Yu D, He J, Xie D, et al. METTL3 promotes ovarian carcinoma growth and invasion through the regulation of AXL translation and epithelial to mesenchymal transition. Gynecologic Oncology. 2018; 151: 356–365.
[12]
Ma Z, Li Q, Liu P, Dong W, Zuo Y. METTL3 regulates m6A in endometrioid epithelial ovarian cancer independently of METTl14 and WTAP. Cell Biology International. 2020; 44: 2524–2531.
[13]
Jiang J, Dingledine R. Prostaglandin receptor EP2 in the crosshairs of anti-inflammation, anti-cancer, and neuroprotection. Trends in Pharmacological Sciences. 2013; 34: 413–423.
[14]
Iwamoto K, Takahashi H, Okuzaki D, Osawa H, Ogino T, Miyoshi N, et al. Syntenin-1 promotes colorectal cancer stem cell expansion and chemoresistance by regulating prostaglandin E2 receptor. British Journal of Cancer. 2020; 123: 955–964.
[15]
Merz C, von Mässenhausen A, Queisser A, Vogel W, Andrén O, Kirfel J, et al. IL-6 Overexpression in ERG-Positive Prostate Cancer Is Mediated by Prostaglandin Receptor EP2. The American Journal of Pathology. 2016; 186: 974–984.
[16]
Sugino Y, Misawa A, Inoue J, Kitagawa M, Hosoi H, Sugimoto T, et al. Epigenetic silencing of prostaglandin E receptor 2 (PTGER2) is associated with progression of neuroblastomas. Oncogene. 2007; 26: 7401–7413.
[17]
Pan S, Sun S, Liu B, Hou Y. Pan-cancer Landscape of the RUNX Protein Family Reveals their Potential as Carcinogenic Biomarkers and the Mechanisms Underlying their Action. Journal of Translational Internal Medicine. 2022; 10: 156–174.
[18]
Lin X, Luo L, Zou Y, Chen J. Cancer stemness-associated LINC02475 serves as a novel biomarker for diagnosis and prognosis prediction of hepatocellular carcinoma. Frontiers in Genetics. 2022; 13: 991936.
[19]
Xu Q, Lin YB, Li L, Liu J. LncRNA TLR8-AS1 promotes metastasis and chemoresistance of ovarian cancer through enhancing TLR8 mRNA stability. Biochemical and Biophysical Research Communications. 2020; 526: 857–864.
[20]
Pilvenyte G, Ratautaite V, Boguzaite R, Ramanavicius A, Viter R, Ramanavicius S. Molecularly Imprinted Polymers for the Determination of Cancer Biomarkers. International Journal of Molecular Sciences. 2023; 24: 4105.
[21]
Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, et al. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Frontiers in Immunology. 2020; 11: 1280.
[22]
Li B, Jiang J, Assaraf YG, Xiao H, Chen ZS, Huang C. Surmounting cancer drug resistance: New insights from the perspective of N6-methyladenosine RNA modification. Drug Resistance Updates: Reviews and Commentaries in Antimicrobial and Anticancer Chemotherapy. 2020; 53: 100720.
[23]
Lin X, Zheng X, Yang B, Chen J, Xu Q, Wang Q. Clinical significance and immune landscapes of stemness-related and immune gene set-based signature in oral cancer. Clinical and Translational Medicine. 2021; 11: e343.
[24]
Bi X, Lv X, Liu D, Guo H, Yao G, Wang L, et al. METTL3 promotes the initiation and metastasis of ovarian cancer by inhibiting CCNG2 expression via promoting the maturation of pri-microRNA-1246. Cell Death Discovery. 2021; 7: 237.
[25]
Yang L, Chen X, Qian X, Zhang J, Wu M, Yu A. Comprehensive Analysis of the Transcriptome-Wide m6A Methylome in Endometrioid Ovarian Cancer. Frontiers in Oncology. 2022; 12: 844613.
[26]
Liu X, Su K, Sun X, Jiang Y, Wang L, Hu C, et al. Sec62 promotes stemness and chemoresistance of human colorectal cancer through activating Wnt/β-catenin pathway. Journal of Experimental & Clinical Cancer Research: CR. 2021; 40: 132.
[27]
Zhang J, Bai R, Li M, Ye H, Wu C, Wang C, et al. Excessive miR-25-3p maturation via N6-methyladenosine stimulated by cigarette smoke promotes pancreatic cancer progression. Nature Communications. 2019; 10: 1858.
[28]
Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012; 485: 201–206.
[29]
Jiang J, Dingledine R. Role of prostaglandin receptor EP2 in the regulations of cancer cell proliferation, invasion, and inflammation. The Journal of Pharmacology and Experimental Therapeutics. 2013; 344: 360–367.
[30]
Lin X, Li AM, Li YH, Luo RC, Zou YJ, Liu YY, et al. Silencing MYH9 blocks HBx-induced GSK3β ubiquitination and degradation to inhibit tumor stemness in hepatocellular carcinoma. Signal Transduction and Targeted Therapy. 2020; 5: 13.
[31]
Zou Y, Lin X, Bu J, Lin Z, Chen Y, Qiu Y, et al. Timeless-Stimulated miR-5188-FOXO1/β-Catenin-c-Jun Feedback Loop Promotes Stemness via Ubiquitination of β-Catenin in Breast Cancer. Molecular Therapy: the Journal of the American Society of Gene Therapy. 2020; 28: 313–327.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top