Abstract Endometrial cancer (EC) is a prevalent gynecological malignancy worldwide, and 5-methylcytosine (m^5C) modification of mRNA is a crucial epigenetic modification associated with the development and occurrence of several cancers. However, the precise function of m^5C modification in EC remains elusive. This study aimed to investigate the expression and clinical significance of the primary m^5C modification writer, NSUN2, in EC. Our findings indicated that NSUN2 exhibited a substantial up-regulation in EC as a result of an epigenetic augmentation in H3K4me3 levels within the promoter region, which was triggered by the down-regulation of KDM5A. Moreover, gain- and loss-of-function experiments revealed the role of NSUN2 in enhancing m^5C modification of mRNA, thereby promoting EC cell proliferation. RNA bisulfite sequencing and transcriptomic sequencing were employed to elucidate the involvement of NSUN2 in the regulation of ferroptosis. Subsequent in vitro experiments confirmed that the knockdown of NSUN2 significantly up-regulated the levels of lipid peroxides and lipid ROS in EC cells, thereby augmenting the susceptibility of EC to ferroptosis. Mechanistically, NSUN2 stimulated the m^5C modification of SLC7A11 mRNA, and the m^5C reader YBX1 exhibited direct recognition and binding to the m^5C sites on SLC7A11 mRNA via its internal cold shock domain (CSD), leading to an increase in SLC7A11 mRNA stability and elevated levels of SLC7A11. Additionally, rescue experiments showed that NSUN2 functioned as a suppressor of ferroptosis, which was dependent on SLC7A11. Overall, targeting the NSUN2/SLC7A11 axis inhibited tumor growth by increasing lipid peroxidation and ferroptosis of EC cells both in vitro and in vivo. Therefore, our study provides new insight into the role of NSUN2, suggesting that NSUN2 may serve as a prognostic biomarker and therapeutic target in patients with EC. Keywords: NSUN2, SLC7A11, m^5C, Endometrial cancer, Ferroptosis 1. Introduction Endometrial Cancer (EC) is a gynecological tumor originating from the endometrium, and its incidence rate worldwide is steadily increasing [[37]1]. Approximately 90 % of ECs are detected early and have an excellent 5-year survival rate because surgery alone or in combination with local therapy is generally curative [[38]2]. However, the prognosis remains poor for patients with recurrent EC, as the optimal adjuvant therapy is yet to be established [[39]3]. Considering the unsatisfactory treatment results in recurrent EC patients, there is a great need to further explore the mechanisms underlying EC initiation and progression and to develop innovative treatment modalities. More than 100 forms of RNA modifications have been identified, including N6-methyladenosine (m^6A), the most abundant internal mRNA modification that has been shown to play an essential role in cell activities and functions [[40]4]. There is also evidence that 5-methylcytosine (m^5C) regulates RNA metabolism. m^5C is primarily found in tRNAs and rRNAs but has also been identified in mRNAs and other noncoding RNAs [[41]5,[42]6]. Similar to m^6A methylation, m^5C levels of RNAs are also regulated by a series of “writer”, “eraser”, and “reader” proteins. The m^5C methyltransferases consist mainly of NOL1/NOP2/sun (NSUN) family (NSUN1-7) and DNA methyltransferase 2 (DNMT2), which act as “writers” to catalyze the methylation process [[43]7]. In comparison, alpha-ketoglutarate-dependent dioxygenase ABH1 (ALKBH1) and ten-eleven translation family proteins (TET) act as demethylase (“Eraser”), which remove m^5C modifications from RNAs [[44]8,[45]9]. The ultimate fate of m^5C methylated RNAs depends on the “readers”, such as the Aly/REF export factor (ALYREF) and Y-box-binding protein 1 (YBX1). m^5C readers can recognize m^5C-modified sites, which may affect mRNA stability, translation, transcription, nuclear export, and cleavage of RNAs [[46]10,[47]11]. Recently, m^5C modification of RNA was found to play a significant role in tumourigenesis and cancer progression [[48][11], [49][12], [50][13]]. However, the biological significance of m^5C and its underlying regulatory mechanisms in EC remain elusive. Among all the m^5C regulatory proteins, the m^5C writer NSUN2 has been thoroughly investigated. As a major mRNA methyltransferase, NSUN2 is mainly located in the nucleus and catalyzes the formation of 5-methylcytosine in RNA [[51]10]. Growing evidence has shown that NSUN2 is highly expressed in multiple cancer types and is involved in various biological functions such as tumourigenesis [[52]14], cell proliferation [[53]12,[54]15], tumor invasion [[55]11,[56]16], and cellular differentiation [[57]13]. Sun et al. revealed through multi-omics analysis that NSUN2 is significantly upregulated in prostate cancer, indicating poor prognosis. Moreover, pathway enrichment suggests a relationship between NSUN2 and signaling pathways associated with endometrial cancer [[58]17]. Wang et al. discovered a significant upregulation of NSUN2 in cervical cancer, noting that silencing NSUN2 markedly inhibited the migration and invasion of cervical cancer cells. Additionally, they analyzed the expression levels of NSUN2 in early-stage endometrial cancer using expression profiling by array ([59]GSE17025) but did not observe any significant differences. However, this observation has not been validated in tissue samples [[60]18]. Furthermore, Yang et al. analyzed expression changes of RNA 5-Methylcytosine regulators in the TCGA database and identified NSUN2 as being significantly overexpressed in endometrial cancer (EC), suggesting its potential as a prognostic factor in EC [[61]19]. However, a complete understanding of the involvement of NSUN2-mediated m^5C modification of mRNA in the pathogenesis of EC remains insufficient. This study aimed to investigate the mechanisms underlying the role of abnormal NSUN2-mediated mRNA m^5C modifications in promoting EC progression. We demonstrated for the first time that NSUN2 is overexpressed in EC and facilitates the malignant biological behavior of EC cells. Using mRNA bisulfite sequencing paired with transcriptome sequencing, we determined that NSUN2 is involved in the regulation of ferroptosis in EC. Further molecular biology experiments revealed a novel mechanism by which NSUN2 renders EC cells resistant to ferroptosis by regulating m^5C modification of SLC7A11 and enhancing its YBX1-dependent mRNA stability. 2. Materials and methods 2.1. Patients and sample collection All surgical specimens used in this study were obtained from the Department of Obstetrics and Gynecology at the Union Hospital of Tong Medical College (Wuhan, China) and were either immediately frozen in liquid nitrogen or fixed in poly-formalin after surgical excision. At least two experienced pathologists histopathologically confirmed the diagnosis of endometrial cancer. The institutional review board of Tongji Medical College at Huazhong University of Science and Technology granted ethical approval prior to the study implementation. All patients were informed, and written informed consent was obtained from the patients. 2.2. Cell culture and treatment The endometrial cancer cell lines used in this study, including HEC-1A, HEC-1B, KLE, Ishikawa, and RL95-2, were purchased from the American Type Culture Collection (ATCC) and cultured in media containing 10 % FBS according to the instructions of the manufacturer. The cells were cultured in a constant temperature incubator at 37 °C with 5 % CO2. Erastin (#S7242), Z-VAD-FMK (#S7023), 3-MA (#S2767), and Ferrostatin-1 (Fer-1) (#S7243), were obtained from Selleck (Shanghai, China). 2.3. Vector construction and cell transfection To construct NSUN2, YBX1, SLC7A11, and KDM5A over-expression plasmids, human NSUN2 ([62]NM_017755.6), YBX1 ([63]NM_004559.5), SLC7A11 ([64]NM_014331.4) and KDM5A ([65]NM_001042603.3) cDNAs were amplified by PCR and cloned into pcDNA 3.1 expression vectors (Shanghai, China). The small hairpin RNAs (shRNAs) of NSUN2 were synthesized and cloned into the PGMLVHU6-MCS-CMV-PGK-Puro vector by Genomeditech (Shanghai, China). Small interfering RNA (siRNA) targeting YBX1 and KDM5A genes were synthesized by Sangon Biotech (Shanghai, China). Transfections with siRNA (20 nM) were performed with RNATransMate (Sangon, #E607402) based on the manufacturer's instructions. Targeted sequences of shRNAs or siRNAs used in this study were listed in [66]Supplementary Table 1. 2.4. RNA extraction and RT-qPCR Total RNA from tissues and cell lines was extracted using TRIeasy Total RNA Extraction Reagent (Yeasen, #10606ES60) according to the manufacturer's protocol. Total RNA was reverse-transcribed into cDNA using ABScript III RT Master Mix for qPCR (Abclonal, #RK20428). RT-qPCR analysis was conducted on a CFX Connect Real-Time PCR Detection System (Bio-Rad, Hercules, CA, USA) using Universal SYBR Green Fast qPCR Mix (Abclonal, #RK21203). The housekeeping gene GAPDH was used as the internal reference gene for normalization. The primers used are listed in [67]Supplementary Table 2. 2.5. Western blot Protein extracts containing protease inhibitors (Bimake, #[68]B14001) were prepared using RIPA buffer (Servicebio, #G2002). Protein concentrations were measured using the BCA kit (Biosharp, #BL521A). Equal amounts of total protein were separated by SDS-polyacrylamide gel electrophoresis and transferred onto a PVDF membrane (Merck Millipore, ISEQ00010). The membrane was then blocked for 1 h with 5 % non-fat dry milk in 0.1 % TBST. The blots were incubated with primary antibodies on a shaker at 4 °C overnight and then incubated with secondary antibodies for 1 h at room temperature. After 3 × 5 min washes in TBST, chemiluminescent signals were detected using ECL detection reagent (Biosharp, #BL520A). Antibodies against the interest proteins are shown in [69]Supplementary Table 3. 2.6. Chromatin immunoprecipitation assays (ChIP) The cells were subjected to appropriate disposal procedures prior to conducting the ChIP assay, which was performed using the ChIP Assay Kit (Beyotime, #P2078) in accordance with the manufacturer's instructions. Immunoprecipitation was carried out using anti-H3K4me3 and anti-KDM5A antibodies, while normal rabbit IgG was utilized as a negative control. The primer for ChIP-qPCR targeting NSUN2 promoter is provided in [70]Supplementary Table 2. 2.7. CUT&Tag CUT&Tag was performed using a commercial kit (Vazyme, China, TD903) according to the manufacturer's protocol. In brief, a total of 50,000 cells were collected and subjected to a washing procedure, after which they were incubated with magnetic beads coated with Concanavalin A. Subsequently, the cells were exposed to an anti-H3K4me3 primary antibody (Abcam, #ab12209) for a duration of 2 h, followed by an additional hour of incubation with a secondary antibody at room temperature. To facilitate tagmentation, the hyperactive pG-Tn5 transposonase was introduced. The reaction was then halted, and the resulting DNA fragments were extracted using PCI. Finally, PCR amplification was performed utilizing indexed P5 and P7 primers. The library products underwent enrichment, quantification, and subsequent sequencing using a Novaseq 6000 sequencer (Illumina) with the PE150 model. The CUT&Tag experiment, as well as high-throughput sequencing and data analysis, were performed by Seqhealth Technology Co., LTD (Wuhan, China). 2.8. RNA immunoprecipitation (RIP) and m^5C-mRNA immunoprecipitation The RIP experiment was carried out with the Magna RIP RNA-Binding Protein Immunoprecipitation Kit (Millipore, Bedford, MA, USA) according to the manufacturer's instructions. Cell lysates were incubated with 5 μg of primary antibodies against NSUN2, YBX1, and FLAG. The controls used were mouse or rabbit IgG. m^5C-mRNA immunoprecipitation was performed according to a previous description [[71]14]. Briefly, mRNA was purified from total RNA using a Hieff NGS mRNA Isolation Master Kit (Yeasen, #12603ES24). Anti-m^5C antibody was mixed with 50 μL protein G magnetic beads (Thermo Fisher, #10007D) and incubated for 30 min at room temperature with rotation. Subsequently, the purified mRNA was mixed with the antibody-bead mixture and incubated on a rotator overnight at 4 °C. The mRNA bound to the antibody was isolated using proteinase K (Beyotime, # ST532), followed by an additional round of purification with RNA Clean Magnetic Beads (Beyotime, #R0081), and the purified mRNA was then subjected to reverse transcription and quantitative PCR analysis. 2.9. Dot blot assays for m^5C The total RNA of EC cell lines was subjected to denaturation at 95 °C for 3 min, followed by cooling on ice. Equal amounts of RNA were spotted on a positively charged nylon membrane (Biosharp, #BS-NY-45), crosslinked with UV for 3 min at 120 mJ/cm2, and then washed with buffer to remove unbound RNA. After blocking with 5 % non-fat milk, the membrane was incubated with m^5C antibody overnight at 4 °C. The membrane was washed three times with buffer and incubated with a secondary antibody. Finally, chemiluminescence was used for detection, and methylene blue staining was used as a loading control. 2.10. RNA-bisulfite sequencing The total RNA of EC cell lines was extracted using Trizol, and the RNA samples were fragmented to lengths suitable for sequencing. Subsequently, EZ RNA Methylation Kit (Zymo Research, CA, USA) was used for Bisulfite treatment of RNA, followed by RNA library construction using KC UMI RNA Library Kit (Seqhealth, Wuhan, China), which was subjected to high-throughput sequencing. The raw FASTQ format sequencing data was quality controlled, the Clean Reads were aligned to the reference genome using Bismark (v0.22.3) software, and duplicate reads were removed. Bismark software was used to identify the methylation sites of cytosine (C). The detected methylation sites' corresponding methylation levels were calculated by mC/(mC + umC). Differentially methylated regions (DMRs) are regions where the methylation level between samples exhibits significant differences. The detection of DMRs between the two groups of samples was conducted using metilene software (v0.2-8) [[72]20], with filtering parameters of methylation difference greater than 0.1 and P-value less than 0.05. A complete list of differentially methylated genes was provided in [73]Supplementary Table 4. 2.11. RNA sequencing Total RNAs were extracted from EC cell lines using TRIzol Reagent (Invitrogen, c#15596026). RNA quality was evaluated by examining A260/A280 with Nanodrop OneCspectrophotometer. A total of 2 μg RNAs were used for stranded RNA sequencing library preparation using KCTM Stranded mRNA Library Prep Kit following the manufacturer's instructions. On the DNBSEQ-T7 sequencer (MGI Tech Co., Ltd. China), 200–500 bps PCR products were enriched, quantified, and sequenced. Clean data were mapped to the hg38 human reference genome using STRA software (version 2.5.3a) with default parameters. Reads mapped to the exon regions of each gene were counted by featureCounts (Subread-1.5.1; Bioconductor), and then RPKMs were calculated. Differentially expressed genes between groups were identified using the edgeR package (version 3.12.1). A p-value cutoff of 0.05 and a fold-change cutoff of 2 were used to judge the statistical significance of gene expression differences. A complete list of differentially expressed genes was provided in [74]Supplementary Table 5. RNA-Bisulfite sequencing and RNA sequencing experiments were conducted by Seqhealth Technology Co. (Wuhan, China). 2.12. RNA stability assay The EC cell lines were evenly seeded in a 6-well plate and treated with 5 μg/mL actinomycin D (ActD) (Selleck, #S8964) for 0, 2, 4, 6, and 8 h, respectively. The collected cells were used for total RNA extraction and quantitative PCR detection of mRNA levels of target genes. The mRNA half-life was calculated based on the previously described method [[75]21]. 2.13. RNA pull-down Biotin-labeled RNA fragments (50 pmol) containing SLC7A11 RNA sequences with (oligo-m^5C) or without m^5C modification at the m^5C site (chr4: 138,242,036) were synthesized from Sangon (Shanghai, China; [76]Supplementary Table 2). RNA pull-down assays were performed using the Pierce™ Magnetic RNA-Protein Pull-Down Kit (20,164, Thermo Scientific), according to the manufacturer's instructions. The resulting RNA-protein complexes were further analyzed by immunoblotting or mass spectrometry. 2.14. Silver staining and mass spectrometry (MS) analysis After a duration of 75 min of electrophoresis at a voltage of 120 V, the gel underwent silver staining using the Fast Silver Stain Kit (P0017S, Beyotime) in accordance with the instructions provided by the manufacturer. The process of mass spectrometry (MS) was carried out by SpecAlly Life Technology Co., Ltd. (Wuhan, China). To summarize, the complexes bound to immobilized magnetic beads were subjected to cleaning and digestion using sequencing-grade modified trypsin. Following extraction and purification, the peptide samples were identified through mass spectrometry using the Q Exactive instrument manufactured by Thermo Finnigan in the United States. The acquired raw mass spectrometric data were analyzed using MaxQuant software (V1.6.2.10). 2.15. Transmission electron microscope (TEM) For the observation of the ultrastructure of cells, a TEM was used. The EC cells were collected and resuspended in TEM fixative solution at 4 °C for 2–4 h. Then, the cells were pre-embedded in agarose, and 2 h of fixation was performed at room temperature using 1 % glutaraldehyde. After gradient alcohol dehydration, the samples were infiltrated and polymerized using a solution of propylene oxide and epoxy resin. Ultrathin sections were prepared and stained before observation using an HT7800 TEM (HITACHI). Images were collected for analysis. 2.16. Tumor xenograft model Four-week-old female nude mice were purchased from Vital River (Beijing, China) and raised in a separate ventilated SPF-level animal facility. Tumor cell suspension (1 × 10^6/point) was subcutaneously injected into the backs of the nude mice. Tumor size was measured every four days using a caliper. After four weeks, the mice were euthanized, and the xenograft tumors were harvested, weighed, and measured for volume. The xenograft tumors were fixed with formalin and embedded in paraffin for subsequent IHC staining. 2.17. Malondialdehyde (MDA) measurement The relative MDA content in the cell lysate was measured using a Lipid Peroxidation Assay Kit (Beyotime, #S0131S) according to the manufacturer's instructions. Briefly, EC cells were lysed using lysis buffer (Beyotime, #P0013) and centrifuged at 12,000 g for 10 min. A total of 100 μL supernatant was mixed with 200 μL MDA working solution and incubated at 100 °C for 15 min, followed by cooling to room temperature. The absorbance value at 532 nm was then measured. 2.18. Lipid reactive oxygen species (ROS) measurement EC cells were seeded in a six-well plate before treatment. After removing the culture medium, the cells were incubated with 5 μM C11-BODIPY (Thermo Fisher Scientific, #D3861) in HBSS at 37 °C for 30 min. After washing the cells twice with PBS, trypsin was used to digest the cells, followed by centrifugation. The cells were resuspended in a serum-free medium for flow cytometric analysis (DxFLEX, Beckman Coulter, USA), and data were analyzed using FlowJo software (TreeStar). 2.19. Immunohistochemistry (IHC) The surgically resected tissues were immediately fixed with 4 % paraformaldehyde and embedded in paraffin. For immunohistochemical analysis, the tissues were dewaxed, dehydrated, and rehydrated, followed by overnight incubation with primary antibodies at 4 °C. The tissues were then incubated with secondary antibodies the next day, and DAB was used for chromogenic detection. Images were captured using a microscope equipped with a digital camera. The immunohistochemical score (IHC score) was defined as the product of staining intensity and staining area, where the staining intensity was scored as negative (0), weak (1), moderate (2), or strong (3), and staining area was scored as follows: 0 = no positive cells; 1 = <10 % of positive cells; 2 = 10–50 % positive cells; 3 = 51–80 % positive cells; 4 = >80 % positive cells. 2.20. Cell proliferation and viability assays Cell proliferation assays were performed as previously described [[77]22]. Cell viability was evaluated using a CCK8 Cell Counting Kit (Topscience, Shanghai, China). In brief, 5 × 10^3 EC cells were seeded in a 96-well plate before treatment. After removing the culture medium, 100 μL complete medium containing 10 μL of CCK8 reagent was added and incubated at 37 °C for 2 h in a CO2 incubator. The absorbance at 450 nm was measured, and the cell viability was calculated. 2.21. Statistical analysis Data are expressed as mean ± SD. The experiments were repeated at least three times with a minimum of three technical repeats. Statistical significance was determined with students' t-test. The Kaplan-Meier method was used to calculate OS. P-values less than 0.05 were considered statistically significant. Statistical analyses were performed using Prism 8.1.2 (GraphPad Software Inc.). 3. Results 3.1. Elevated NSUN2 expression correlates with poor prognosis in patients with EC To investigate the potential role of m^5C modification in EC progression, we preliminarily analyzed the expression of critical m^5C regulatory genes in TCGA UCEC dataset. We found that NSUN2, which catalyzes mRNA m^5C, was significantly upregulated ([78]Fig. 1A, [79]Supplementary Figs. S1A and B). We also compared the mRNA levels of NSUN2 in paired EC tissues and adjacent normal tissues in the TCGA database, which suggested high NSUN2 expression in EC ([80]Fig. 1B). Moreover, based on public data from the CPTAC, we found higher protein expression of NSUN2 in EC than in normal endometrial tissues ([81]Fig. 1C). RT-qPCR and western blotting were employed to assess the mRNA and protein levels of NSUN2 in surgically resected specimens, which confirmed the overexpression of NSUN2 in EC tissues compared to normal tissues ([82]Fig. 1D and E). Immunohistochemistry revealed that NSUN2 was highly expressed in EC tissues and predominantly located in the nucleus ([83]Fig. 1F). We evaluated the prognostic significance of NSUN2 by analyzing clinicopathological factors and found that the expression levels of NSUN2 in EC were closely related to FIGO stage, histological grade, and pathological type ([84]Fig. 1G–I). Moreover, patients with high expression of NSUN2 showed a much worse prognosis ([85]Fig. 1J). Collectively, these findings demonstrated that NSUN2 is often upregulated in EC, making it a potential prognostic indicator. Fig. 1. [86]Fig. 1 [87]Open in a new tab NSUN2 overexpression predicts poor prognosis in endometrial cancer. (A) The mRNA levels of NSUN2 were compared between EC and normal tissues in TCGA UCEC dataset. (B) Different mRNA expression of NSUN2 between EC and the paired normal tissues. (C) The protein expression levels of NSUN2 based on the CPTAC database. (D) RT-qPCR analysis displaying the mRNA levels of NSUN2 in surgical EC and normal samples. (E) Western blot was used to detect the protein levels of NSUN2 in EC and paired normal tissues. (F) Representative images of immunohistochemical staining for NSUN2 and the comparison of the ISH scores in EC and normal tissues. The expression levels of NSUN2 were presented in different FIGO stages (G), histological grade (H), and pathological type (I). (J) Kaplan-Meier (KM) curve for OS of patients with UCEC in TCGA and patients were stratified into low and high expression based on the median NSUN2 expression. *P < 0.05; **P < 0.01; ***P < 0.001. 3.2. NSUN2 upregulation is induced by the epigenetic alteration of H3K4me3 Post-translational modifications (PTMs) of histones play essential roles in the regulation of gene expression in cancer cells [[88]23]. Consequently, we used the WashU Epigenome Browser and the Cistrome Data Browser to explore whether NSUN2 is regulated by epigenetic mechanisms [[89]24,[90]25], which discovered significant enrichment of H3K4me3 in the promoter region of NSUN2 ([91]Fig. 2A). Gene promoters enriched for H3K4me3 are generally associated with transcriptional activation [[92]26], suggesting that elevated NSUN2 might be regulated by histone methylation. Moreover, chromatin immunoprecipitation (ChIP) assays were performed to further determine histone methylation levels in EC cells, and the ChIP-PCR results indicated that histone H3 lysine 4 (H3K4) trimethylation (H3K4me3) was significantly enriched in the promoter region of NSUN2 ([93]Fig. 2B). To yield more insight into the underlying mechanisms, we analyzed the expression of lysine methylases and demethylases of H3K4me3 in TCGA UCEC dataset. The findings indicate that the decreased expression of KDM5A may be the primary factor contributing to the elevated levels of H3K4me3 at the NSUN2 promoter region ([94]Supplementary Fig. S2A). We then used siRNA technology to knockdown endogenous KDM5A expression in EC cells and found that the mRNA level of NSUN2 increased significantly ([95]Fig. 2C). Furthermore, Western blot results also indicated that KDM5A knockdown led to an increase in NSUN2 protein levels and H3K4me3 levels, whereas over-expression of KDM5A produced the opposite result ([96]Fig. 2D, [97]Supplementary Figs. S2B and C). Consistently, ChIP-PCR was performed in KDM5A-overexpressed EC cells, and the results suggested that the overexpression of KDM5A resulted in a significant decrease of H3K4me3 enrichment in the NSUN2 promoter region ([98]Fig. 2E). Similarly, we also found the enrichment of KDM5A in the NSUN2 promoter region in the Cistrome Data Browser, and ChIP-PCR assay using KDM5A antibody also confirmed that KDM5A bound the NSUN2 promoter in EC ([99]Fig. 2F and G). Moreover, we proceeded to suppress KDM5A in EC cell lines and conducted CUT & Tag analysis. The findings indicated that diminishing KDM5A levels can augment the accumulation of H3K4me3 in the promoter region ([100]Fig. 2H). Additionally, the IGV map reveals an enhanced signal of H3K4me3 enrichment in the NSUN2 promoter region subsequent to KDM5A knockdown([101]Fig. 2I). Taken together, the above results further revealed that NSUN2 upregulation occurs because of an increase in H3K4me3 mediated by a decrease in KDM5A. Fig. 2. [102]Fig. 2 [103]Open in a new tab NSUN2 upregulation is regulated by the epigenetic alteration of H3K4me3. (A) ChIP-seq data for mapping H3K4me3 modifications near the NSUN2 promoter in the different cell lines were obtained from the Cistrome Data in the WashU Epigenome Browser. (B) ChIP-PCR analysis of H3K4me3 modification in the promoter of NSUN2. (C) The mRNA levels of NSUN2 were detected by qPCR after the knockdown of KDM5A. (D) Protein expression in HEC-1B or Ishikawa cells with overexpression or silencing of KDM5A was assessed using Western blot; GAPDH and H3 were used as the loading control. (E) ChIP-PCR analysis of H3K4me3 modification of the NSUN2 promoter in KDM5A overexpression EC cells. (F) KDM5A ChIP-seq of different cell lines at NSUN2 promoter is displayed using the WashU Epigenome Browser. (G) ChIP-PCR analysis of KDM5A in the promoter of NSUN2. (H) Heatmaps illustrating H3K4me3 levels around gene body regions. (I) IGV tracks presenting the enrichments of H3K4me3 by CUT&Tag. ***P < 0.001. 3.3. NSUN2 deposits m^5C on mRNA and promotes EC proliferation in vitro To further demonstrate the regulatory effects of NSUN2 on tumor progression in vivo, we evaluated the mRNA and protein expression of NSUN2 in different EC cell lines and in normal endometrium (NE). Increased NSUN2 levels were observed in cancer cell lines in contrast to primary normal cells ([104]Fig. 3A and B), and HEC-1B, KLE, and Ishikawa cell lines were selected for subsequent studies. We silenced NSUN2 in HEC-1B and KLE cells using shRNA knockdown and overexpressed NSUN2 in Ishikawa cells using NSUN2 expression vectors ([105]Supplementary Figs. S3A–C). We purified mRNA from all three EC cell lines transfected with the indicated vectors and performed dot blot analysis with an antibody recognizing m^5C, which demonstrated that NSUN2 knockdown resulted in reduced m^5C levels, whereas NSUN2 overexpression resulted in an increased level of m^5C mRNA ([106]Fig. 3C). Subsequently, a series of experiments were conducted to assess the effect of NSUN2 on EC cell proliferation, including CCK-8, clone formation, and EDU assays. As shown in [107]Fig. 3D–F and [108]Supplementary Figs. S3D and E, NSUN2-knockdown robustly inhibited the proliferation of HEC-1B and KLE cells, whereas overexpression of NSUN2 promoted the proliferation of Ishikawa cells. Fig. 3. [109]Fig. 3 [110]Open in a new tab NSUN2 is involved in the regulation of m^5C levels and the proliferation ability of EC cells. (A, B) The mRNA and protein levels of NSUN2 in normal endometrium and five different EC cell lines. (C) The m^5C levels of total RNA in NSUN2 knockdown HEC-1B and KLE cells or NSUN2 overexpression Ishikawa cells were indicated by m^5C dot blot. (D–F) CCK-8, colony formation, and EdU assays were carried out to detect the proliferation ability of EC cells with NSUN2 knockdown or overexpression. *P < 0.05; **P < 0.01; ***P < 0.001. As the primary mRNA m^5C methyltransferase, it has been confirmed that mutations in the releasing (cysteine 271) and catalytic (cysteine 321) sites of NSUN2 completely abrogated its regulatory role on mRNA m^5C levels [[111]10]. Thus, we constructed wild-type and double-mutant NSUN2 overexpression plasmids and transfected them into EC cells with stable knockdown of NSUN2. In comparison with wild-type NSUN2, the mutant NSUN2 failed to restore the proliferation ability of EC cells ([112]Supplementary Figs. S4A–C). Collectively, these results indicate that NSUN2 facilitates EC proliferation in vitro via m^5C modification. 3.4. NSUN2 stimulates m^5C hypermethylation in EC As NSUN2 is a major mRNA methyltransferase, we performed bisulfite sequencing (BS-seq) of mRNA derived from HEC-1B cells transfected with control or sh-NSUN2 vectors ([113]Fig. 4A). As shown in [114]Fig. 4B, NSUN2 knockdown resulted in a considerable reduction in m^5C modification sites. A higher frequency of C[A/C]GGGG downstream of the m^5C site was observed in the probability sequence context generated using WebLogo ([115]Fig. 4C). The distribution density map of m^5C sites on the mRNA of HEC-1B was drawn, and it indicated that m^5C sites are located in the coding DNA sequences (CDS): 5′UTR and 3′UTR ([116]Fig. 4D). Differentially Methylated Regions (DMRs) are regions with significant differences in methylation levels between samples that may play an essential role in the regulation of gene expression. Using Metilene software, we identified differentially methylation-modified genes between the control and NSUN2 knockdown groups. The cut-off for significant DMR was |methylation changes|> 0.1 and p < 0.05 ([117]Fig. 4E). Among these differentially methylated genes, there were more genes whose methylation levels were decreased by more than one after NSUN2 knockdown ([118]Fig. 4F). Differentially methylated genes were also subjected to pathway enrichment analysis using KOBAS3.0, and significant enrichment for the oxidative stress response, ferroptosis, and iron uptake was observed ([119]Fig. 4G). Fig. 4. [120]Fig. 4 [121]Open in a new tab Identification of NSUN2-mediated regulation of m^5C-modified mRNAs. (A) Flow chart of the bisulfite sequencing. (B) The number of m^5C modified sites on different chromosomes in control or NSUN2 knockdown HEC-1B cells. (C) The sequence context of 10 bases upstream and downstream of NSUN2-m^5C sites is depicted in the probability pattern. (D) Distribution and methylation level of all m^5C sites in different segments of mRNAs. (E) The heatmap showed differentially methylated genes in HEC-1B cells with NSUN2 knockdown. The screening criteria were as follows: methylation difference greater than 0.1, P-value less than 0.05. (F) The number distribution histogram of differentially methylated mRNAs. (G) Pathway enrichment analysis of differentially methylated genes. 3.5. The downregulation of NSUN2 renders EC susceptible to ferroptosis Considering that differentially methylated genes were enriched in ferroptosis-related pathways, we speculated that NSUN2 may participate in the regulation of ferroptosis in EC. Ferroptosis is characterized by iron-dependent cell death induced by cysteine depletion and massive lipid peroxidation [[122]27]. In previous studies, we found that significant ferroptosis resistance exists in EC cells and that targeting ferroptosis is a potential treatment for EC [[123]28]. Functionally, NSUN2 knockdown significantly promoted erastin-induced cell death in a concentration-dependent manner ([124]Fig. 5A and B). Notably, Fer-1, an inhibitor of ferroptosis, rather than inhibitors of other forms of cell death such as apoptosis (Z-VAD-FMK) and autophagy (3-methylademine, 3-MA), suppressed the erastin-induced increase in cell death in HEC-1B and KLE cells with NSUN2 knockdown ([125]Fig. 5C and D). This indicated that NSUN2 is involved in the regulation of ferroptosis in EC cells. Similarly, EC cells were treated with erastin or DMSO for 12h and subjected to live or dead staining, which suggested that NSUN2 knockdown significantly increased erastin-induced cell death ([126]Fig. 5E). Furthermore, the relative intracellular MDA concentration and lipid ROS levels suggested that NSUN2 knockdown caused a robust increase in the extent of lipid peroxidation and lipid ROS ([127]Fig. 5F and G). Transmission electron microscope (TEM) analysis further revealed that NSUN2 knockdown EC cells exhibited typical morphological features of ferroptosis, including shrunken mitochondria with elevated membrane density ([128]Fig. 5H). Taken together, these observations support the conclusion that the downregulation of NSUN2 renders EC susceptible to ferroptosis. Fig. 5. [129]Fig. 5 [130]Open in a new tab NSUN2 knockdown promotes ferroptosis in EC. (A, B) CCK-8 assay was performed to determine the cell viability after treatment with different concentrations of erastin in HEC-1B or KLE cells. (C, D) Evaluation of cell viability after treatment with DMSO, erastin, erastin and Fer-1, erastin and Z-VAD, erastin and 3-MA in the indicated cells. (E) Calcein-AM (green)/PI (red) staining was performed to visualize living and dead cells, respectively. (F) Intracellular lipid peroxides MDA levels were measured with an MDA assay kit. (G) Lipid peroxidation was measured by flow cytometry after C11-BODIPY staining in the indicated cells. (H) Representative TEM images of mitochondria in control or NSUN2 knockdown EC cells. ***P < 0.001; ns, not statistically significant. (For interpretation of the references to