- Research
- Open access
- Published:
Distinct 5-methylcytosine profiles of LncRNA in breast cancer brain metastasis
BMC Cancer volume 25, Article number: 557 (2025)
Abstract
Background
Recent studies have identified a complex relationship between methylation patterns and the development of various cancers. Breast cancer (BC) is the second leading cause of cancer mortality among women. Approximately 5–20% of BC patients are at risk of BC brain metastases (BCBM). Although 5-methylcytosine (m5C) has been identified as an important regulatory modifier, its distribution in BCBM is not well understood. This study aimed to investigate the distribution of m5C in BCBM.
Materials and methods
Samples from BCBM (231-BR cells) and BC (MDA-MB-231 cells) groups were subjected to a comprehensive analysis of the m5C methylation in long non-coding RNA (lncRNA) using methylated RNA immunoprecipitation next-generation sequencing (MeRIP-seq). The expression levels of methylated genes in BC and adjacent tissues were verified through quantitative real-time polymerase chain reaction (RT-qPCR). Enrichment pathway analyses were through Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to predict the potential functions of m5C in BCBM.
Results
The MeRIP-seq analysis identified 23,934 m5C peaks in BCBM and 21,236 m5C in BC. A total of 9,480 annotated genes (BCBM) and 8,481 annotated genes (BC) were mapped. Notably, 1,819 methylation sites in lncRNA were upregulated in BCBM, whereas 2,415 methylation sites were upregulated in BC. Significant m5C hypermethylated lncRNAs included ENST00000477316, ENST00000478098 and uc002gtt.1, whereas hypomethylated lncRNAs included ENST00000600912, ENST00000493668, ENST00000544651 and ENST00000464989. These results were verified by qPCR and MeRIP-qPCR in BC and BCBM. Considering the strong association between m5C RNA methylation regulators and lncRNA, we examined the expression levels of 13 m5C RNA methylation regulators and observed significant differences between BC tissues and adjacent normal tissues. In addition, the interaction between regulators of altered expression and the differentially expressed genes in vitro was analyzed. The GO and KEGG pathways analyses revealed that genes significantly associated with m5C sites in lncRNA were linked to the BCBM signaling pathways.
Conclusion
This uncovered significant variations in the levels and distribution of m5C in BCBM compared to BC. The findings provide a new theoretical understanding of the mechanisms of BCBM.
Introduction
RNA post-transcriptional modifications are increasingly being studied in recent years [1, 2]. These modifications, which exceed 100 in number, include pseudouridine, N6-methyladenosine (m6A) [3], N1-methyladenosine (m1A) [4], and 5-methylcytosine (m5C) [5]. The important role of m6A modification for RNA has been documented by several studies, including its involvement in the development of various cancers, such as colon cancer [3], hepatocellular carcinoma [6], and glioblastoma [7, 8]. Another noteworthy post-transcriptional modification of RNA, m5C, has been identified in stable and abundant tRNAs, as well as in various RNAs and mRNAs [9].
The widespread m5C modification in cells underscores its crucial role in the regulation of gene expression and RNA stability [5]. Catalyzed by RNA methyltransferase or DNA methyltransferase 2 (DNMT2) [10], m5C can be demethylated by tet methylcytosine dioxygenase [11]. m5C-modified mRNA is recognized by ALYREF, which facilitates its nuclear export. In contrast, Y-box binding protein 1 (YBX1) directly interacts with m5C-methylated mRNA, contributing to its stabilization [12]. Recent studies have highlighted the importance of m5C methylation and its related modifying enzymes in tumor initiation and progression. For instance, NSUN2-mediated m5C methylation could promote tumorigenesis and progression of bladder cancer [5], and the m5C methyltransferase NSUN1 alters chromatin structure by interacting with BRD4, influencing the responsiveness of leukemia tumor cells to the chemotherapeutic agent 5-azacytidine [13].
Studies show that m5C modification is essential for the stability and efficiency of tRNA translation, which is critical in the processing, construction, and translation of rRNA [14]. Moreover, m5C modification also occurs in noncoding RNAs (ncRNAs) such as long non-coding RNAs (lncRNAs) and mRNA [15]. Comprehensive analysis of the m5C profiles of mammalian transcriptomes have revealed that the m5C modification contributes to the export of mRNA and post-transcription regulation [16,17,18]. Studies show that the m5C-modified H19 lncRNA can specifically interact with the G3BP1 protein, causing accumulation of the oncoprotein Myc and promoting the occurrence and development of liver cancer [19].
LncRNAs, with lengths exceeding 200 nucleotides and lacking protein-coding functions [20,21,22], have been associated with several epigenetic mechanisms, such as gene silencing, histone modification, regulation of transcription, interference in transcriptional processes, and nuclear transport; which are associated with the progression of various human diseases [23]. Several lncRNAs have been linked to the formation and progression of tumors [23]. For instance, the specific sequence of LINC00942 recruits the methyltransferase METTL14, stabilizing the downstream targets of LNC942 and enhancing the initiation and progression of breast cancer (BC) [24]. Currently, the distribution and functional significance of m5C modification of lncRNA in breast cancer brain metastases (BCBM) is not fully understood.
BC is the second most common cause of cancer-related deaths among women, with the BCBM incidence ranging from 10–20% [25, 26]. BCBM was found to be correlated with unfavorable prognosis, negatively affecting cognitive and sensory functions and severely limiting the patient’s quality of life [25, 27]. Treatment modalities for BCBM comprise surgical intervention, whole-brain radiotherapy (WBRT), stereotactic radiation therapy (SRS), and chemotherapy. These face challenges due to the blood-brain barrier (BBB), leading to unsatisfactory longevity expectations for BCBM patients and limiting penetration and chemoresistance [28]. Recent studies indicates that methylation could be a crucial therapeutic target for BCBM, given its role in the development and progression of the condition [29].
This study adds new insights into the effect of m5C methylation of lncRNAs on BCBM and BC. By using m5C-specific in-depth bioinformatics analyses, we demonstrated that m5C modification was higher in BCBM than in BC, and this phenomenon varied across all chromosomes.
Materials and methods
Preparation of RNA sequencing samples and cell culture
Patricia S Steeg gifted BCBM (231-BR) and BC (MDA-MB-231) cells [30, 31] (National Cancer Institute, NIH, Bethesda, MD, USA), and all subsequent experiments were performed with a comparison between the two groups. Cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM; Sigma Corporation, MO, USA) enriched with 10% (v/v) Fetal Bovine Serum (FBS, Sangon Biotech, Shanghai, China) at 37℃ in a 5% CO2 incubator. Total RNA was extracted from cultured cells utilizing TRIzol reagent (Thermo Fisher, MA, USA) by the manufacturer’s guidelines. The concentration and purity of the RNA were evaluated using a NanoDrop 2000 ultraviolet-visible spectrophotometer (Thermo Fisher, MA, USA), where OD260/OD280 ratios ranging from 1.8 to 2.1 were deemed acceptable.
RNA MeRIP-seq library construction & sequencing
MeRIP-Seq was conducted using Cloudseq Biotech Inc (Shanghai, China) following a slightly modified version of an established protocol [32]. Fragmented RNA was incubated with an anti-m5C antibody for 2 h at 4 °C in IPP buffer. The mixture was then subjected to immunoprecipitation using protein-A beads (Thermo Fisher, MA, USA) at 4 °C for an additional 2 h. The RNA was subsequently eluted using a free m5C adenosine analog. RNA MeRIP-seq was performed with three biological replicates per group. The eluted RNA was extracted using Trizol reagent (Thermo Fisher, MA, USA). Purified RNA was used for library construction with RNA Library Kit (New England Biolabs, Inc, USA), and paired-end 150 bp sequencing was performed on an Illumina Hiseq sequencer (Fig. 1).
Analysis of sequencing data
Quality control was conducted on paired-end reads (Q30 > 80%, Supplementary Table S1), followed by the trimming of 3’ adapters, and low-quality reads were filtered out using Cutadapt software (v1.9.3). The clean reads derived from the input library were accurately mapped to the reference genome using the STAR software [33], subsequently leveraging the DCC software to identify circRNAs [34]. The clean reads were mapped to the reference genome using Hisat2 software (v2.0.4) [35]. Methylated genes in each sample were identified using MACS software [36]. The portions that overlap with the exons of known lncRNAs were selected using the proprietary program of Shanghai CloudSeq Biotechnology Company. The threshold for MACS identification was set as P < = 0.00001 and Fold enrichment > = 2. Subsequently, the diffReps software [37] was utilized to identify differentially methylated sites and conduct corresponding annotations. The dataset can be accessed at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE246721.
Analysis of pathway enrichment
Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses used DAVID to interpret the functional enrichment and annotation of differentially methylated genes. P-values < 0.05 were regarded as significant for both analyses.
Quantitative real-time PCR (RT-qPCR) analysis
Total RNA from all samples was isolated using Trizol reagent (Thermo Fisher, MA, USA). The first complementary DNA (cDNA) strand was synthesized using reverse transcription kits (Beyotime, Beijing, China). cDNA underwent RT-qPCR utilizing a SYBR Green qPCR Master Kit (Toroivd, Shanghai, China) along with specific primers (sequences provided in Supplementary Table S2) under the reaction condition of 95℃ for 5s and 60 ℃ for 30s for 35 cycles.
Western blotting
Protein was extracted using RIPA buffer (Applygen, Beijing, China). The BCA protein quantification kit was then used to assess the protein concentration in the sample (Beyotime, Beijing, China). The protein samples were separated through SDS-PAGE gel electrophoresis and transferred to a polyvinylidene fluoride (PVDF) membrane. The membrane was blocked with 5% non-fat milk at room temperature for 1 h to prevent nonspecific binding and then incubated overnight at 4 °C with primary antibodies targeting NSUN2 (1:1000, #52901S, Cell Signaling Technology, USA) or β-actin (1:1000, #sc-47778, Santa, USA). Once the primary antibody incubation was finished, the PVDF membrane was rinsed three times using TBST buffer. An HRP-conjugated secondary antibody was then applied at room temperature. Finally, the PVDF membrane was treated with an enhanced chemiluminescence reagent (Beyotime, Beijing, China). The protein signal was captured and analyzed using a chemiluminescent imaging system (Tanon, Shanghai, China).
Knockdown of NSUN2 in BCBM cells via lentiviral infection
For NSUN2 gene knockdown, the BCBM cells were transfected with Lv-shNSUN2 (Genechem, Shanghai, China) or the negative control Lv-NC at a multiplicity of infection (MOI) of 10 for 12 h, following the manufacturer’s protocol. The successfully infected cells were then selected by treating them with 2 µg/mL puromycin (Beyotime, Beijing, China) for 48 h.
Statistical analysis
Data analysis was conducted using GraphPad Prism v5.0 software. Student’s t-test was utilized to compare statistical significance, and P < 0.05 was considered statistically significant. All experiments were conducted in triplicate.
Results
General features of m5C methylation in BC and BCBM
Transcriptome-wide m5C methylation was assessed in BC and BCBM cells through RNA MeRIP-seq of LncRNA. BCBM cells exhibited 23,934 m5C peaks compared to 21,236 in BC cells. The BCBM and BC cells shared 7030 methylation sites, 16,904 were exclusive to BCBM, while 14,206 were exclusive to BC (Fig. 2A). A total of 9,480 annotated genes were mapped in BCBM, and 8,481 in BC, with 6,246 genes shared between the two (Fig. 2B). Moreover, the results showed that the number of up-methylated transcript sites per gene exclusive to BCBM (5.23 sites/gene) or BC (6.36 sites/gene) was markedly higher than the methylated transcript sites shared between BCBM and BC cells (1.13 sites/gene). This observation highlights the intricate nature of epigenetic regulation in BCBM. The distinct patterns of m5C methylation suggest potential mechanisms by which these cells adapt to their microenvironment and acquire unique functional properties.
RNA MeRIP-seq was performed to determine the transcriptome-wide m5C methylation and the overall characteristics of lncRNA in BCBM. (A) A Venn diagram illustrating the m5C methylation sites in lncRNA from BCBM and BC. (B) A Venn diagram displaying the m5C genes in both BCBM and BC. (C) The proportion of lncRNAs with varying m5C methylation peaks between BCBM and BC cells, with the majority having only a single m5C peak. (D) The histogram showing the distribution of m5C methylation sites in various chromosomes
Statistical evaluation of peak counts for each LncRNA
Analysis of m5C peaks on lncRNAs showed that the majority had only one methylation peak, with a higher proportion observed in BCBM (93.24%) compared to BC (92.44%). Furthermore, the percentages for two or more methylation peaks on lncRNAs were comparable between BCBM (5.82%) and BC (6.19%). Three methylation peaks (0.71% BCBM vs. 0.99% BC) and more than three methylation peaks (0.235% BCBM vs. 0.39% BC) were detected (Fig. 2C).
Chromosome visualization of m5C in LncRNAs
The distribution analysis of m5C methylation sites across chromosomes in BCBM and BC cells revealed a broad and widespread pattern in all chromosomes. Notably, there were distinct differences in both methylation levels and distribution between these two cell types. Specifically, chromosome 21 exhibited significantly lower methylation levels compared to the other chromosomes in both BCBM and BC cells, as illustrated in Fig. 2D.
Motif analysis of methylation sites
The sequence of the m5C methylation peak was analyzed to clarify the presence of the m5C motif, which revealed CCAGSCUG (S = C/G) as the most common and reliable motif in BCBM cells (E-value = 2.1e-088) (Fig. 3A). The most consistent motif for m5C peaks in BC cells was identified as GRAGRA (R = G/A) (E-value = 1.2e-119) (Fig. 3B), highlighting distinct motif preferences for m5C peaks in each cell type.
Motif analysis of methylation sites. (A-B) The motif of m5C in BCBM and BC. (C) Heat map displaying the methylation patterns between BCBM and BC cells. (D) Differential expression of lncRNAs between BCBM and BC cells. Up-regulated methylated genes are shown in red, while down-regulated methylated genes are represented by purple
Cluster analysis of differential methylation peaks
Unsupervised hierarchical cluster analysis revealed varying methylation patterns between BCBM and BC cells (Fig. 3C), highlighting both consistency and inter-cellular variability between the two cell types. Differential methylation sites in lncRNAs were identified, with 1819 and 2415 methylation sites in lncRNA upregulated in BCBM and BC cells, respectively. The top ten up and hypomethylated sites in BCBM cells compared to BC cells are shown in Tables 1 and 2.
Effect of methylation on transcriptional expression
To investigate the impact of m5C methylation on lncRNA expression, RNA sequencing data (GSE246721) was used to analyze the expression patterns of lncRNA in BCBM cells and their parental BC cells. The results showed that methylation increased the expression level of numerous lncRNAs in BCBM cells (Fig. 3D). In contrast, methylation decreased the expression levels of lncRNAs in BC cells.
GO and KEGG enrichment analysis
The function of differentially methylated lncRNA-associated genes in BCBM and BC cells was determined using GO functional analysis. The main enrichment in biological processes (BP), cell components (CC), and molecular functions (MF) are shown in Fig. 4 (A-F). The analysis of biological processes (BP) revealed that genes with up-methylated m5C in BCBM were mainly involved in RNA splicing through transesterification reactions with bulged adenosine as a nucleophile, along with RNA splicing and regulation of chromosome organization. In contrast, genes with down-methylated m5C in BCBM were primarily associated with tube morphogenesis, protein localization, organelle organization, and macromolecule localization. Regarding cellular components (CC), genes exhibiting up-methylated m5C in BCBM were associated with organelles, nucleoplasm, nuclear lumen, and bounded organelles. In contrast, genes that were down-methylated m5C in BCBM were associated with organelle membranes, membrane-bounded organelles, and intracellular vesicles. For the MF category, the up-regulated methylation sites in BCBM were enriched in transcription coregulator activity, anion antiporter activity, protein binding and nucleoside-triphosphatase regulator activity and small GTPase binding, protein serine/threonine kinase activity, protein binding and phosphotransferase activity alcohol group as acceptor were down-regulated methylated.
Gene Ontology (GO) enrichment analysis of the differentially methylated lncRNA-associated genes in BCBM cells. The top 10 GO terms in the (A) biological processes (BP), (B) cellular components (CC), and (C) molecular functions (MF), illustrating the enrichment of the up-methylated m5C genes in BCBM. The top 10 GO terms in the (D) BP, (E) CC, and (F) MF, showing enrichment of the down-methylated m5C genes in BCBM
The KEGG pathway analysis highlighted pathways enriched in hyper- and hypo-methylated lncRNA (Fig. 5A-B), providing insights into the molecular mechanisms these methylation changes might affect. The KEGG pathway analysis showed that genes associated with methylated lncRNAs were significantly involved in choline metabolism in cancer, oocyte meiosis, and the P53 and MAPK signaling pathways. Conversely, down-methylated genes were linked to regulating the actin cytoskeleton, pathways governing stem cell pluripotency, VEGF, and the Hippo signaling pathway. The dot plot (GeneRatio) shows the ratio of the number of differentially methylated The top 10 pathway items with significant enrichment, based on high fold enrichment, were associated with lncRNA-related genes among the total number of differentially methylated LncRNA-associated genes in BCBM and BC (Fig. 5C-D).
KEGG pathway analysis for the m5C genes among BCBM lncRNAs. (A) The bar chart illustrating the top ten enrichment scores for significant pathways associated with up-methylated m5C genes in BCBM. (B) The bar chart showing the top ten enrichment scores of the significant pathways associated with down-methylated m5C genes in BCBM. (C) The dot plot indicating the gene ratio values for the ten most significantly enriched pathways associated with the up-methylated m5C genes in BCBM. (D) The dot plot showing the gene ratio values of the ten most significantly enriched pathways associated with the down-methylated m5C genes in BCBM
Differential methylation LncRNAs expression in BCBM and BC cells
The top ten up and hypomethylated sites in BCBM cells in comparison with BC cells were validated using qPCR. The results showed that BCBM and BC shared three upregulated lncRNAs (P < 0.05), ENST00000477316, ENST00000478098 and uc002gtt.1. Furthermore, there are four downregulated lncRNAs which covered ENST00000493668, ENST00000544651, ENST00000600912 and ENST00000464989 (P < 0.05) (Fig. 6A-G). The findings from MeRIP-qPCR aligned with the results above (Fig. 7A-G). The log2 fold changes of seven methylation genes on lncRNAs were calculated (Fig. 7H). The qPCR results were consistent with RNA MeRIP-seq sequencing data on the relative expression level of these seven methylated genes (P = 0.0105), indicating that our RNA MeRIP-seq of lncRNA results are reliable (Fig. 7I).
(A-G) MeRIP-qPCR analysis of the three up-regulated genes and four down-regulated genes following methylation in the BCBM and BC. (n = 3; data are presented as the mean ± SD; P < 0.05). (H) Comparison of mean fold change (log2 conversion) between MeRIP-qPCR and RNA MeRIP-seq. (I) The correlation of the mean fold changes (log2 transformed) between MeRIP-qPCR and the RNA MeRIP-seq data
Analytical integration of m5C LncRNA methylation and the expression levels of LncRNA transcripts
Significant differences in global expression patterns of lncRNAs were detected between BCBM and BC cells, with 199 upregulated and 175 downregulated lncRNAs identified in BCBM (Fig. 8A, B). The differences in the expression patterns of lncRNAs in BCBM were displayed in Volcano plots (Fig. 8C; Fold Change > 2, P < 0.05). The nine-quadrant diagram showed a strong positive correlation between the lncRNA transcriptome and m5C methylation, along with significant variations in this relationship (Fig. 8D; R = 0.991, P < 0.01). The Upset diagram showed that two genes were identified in the up-regulated transcriptional group of lncRNAs and the up-regulated methylation group of m5C-modified lncRNAs. In contrast, two genes were identified in both the down-regulated transcriptional group of lncRNAs and the down-regulated methylation group of m5C-modified lncRNAs. Furthermore, three genes were found in the up-regulated transcriptional group of lncRNAs and the down-regulated methylation group of m5C-modified lncRNAs (Fig. 8E).
Integration analysis of m5C lncRNA methylation and lncRNA transcript expression. (A) Cluster analysis of the lncRNA levels of BCBM and BC. (B) Scatter Plot analysis of differential lncRNA expression in BC and BCBM. Up-regulated lncRNA are shown in red, while down-regulated lncRNA are indicated in green. (C) A volcano plot showing significant differences in lncRNA expression between the BCBM and BC (Fold change > 2.0 and P < 0.05). (D) Nine-quadrant diagram for m5C methylation and lncRNA expression. (E) Up-set graph for different m5C methylation and lncRNA expression profiles
Validation of differentially methylated LncRNA in BC tissues
Validation of methylation sites in BC and adjacent tissues confirmed differential expression patterns observed in cell experiments. Increased expression of ENST00000477316, ENST00000478098 and uc002gtt.1, and decreased expression of ENST00000493668, ENST00000544651, ENST00000600912 and ENST00000464989 was observed in BC compared to adjacent tissues (P < 0.05) (Fig. 9A-G).
Analysis of m5C RNA methylation regulators
RNA methylation regulators for m5C consist of “writers” (methyltransferases), “erasers” (demethylases), and “readers” (binding proteins)[11]. Considering the dynamic regulatory role of m5C in LncRNA methylation, we began by performing a thorough comparison of the expression levels of 13 regulators associated with m5C RNA methylation (Supplementary Table S2) between BC and adjacent tissues (due to the challenge in acquiring specimens of BCBM). The analysis compared the alterations in the expression of m5C RNA methylation regulators between BC and adjacent tissues). Given their elevated expression in BC tissues, NSUN2, NSUN5, YBX1, and ALYREF may play important roles in cancer progression (P < 0.05). Methyltransferase DNMT3B, DNMT3A, DNMT1, and TET3 were downregulated (P < 0.05), and no difference was observed between BC and adjacent tissues for NSUN3, NSUN4, NSUN5, NSUN6, and TET2 (Fig. 10A).
Analysis of m5C RNA methylation regulators. (A) Analysis of the relative expression of eight RNA methylation regulators in BC and adjacent tissues by qPCR. The expression of NSUN2 in BC and BCBM was quantified by qPCR (B) and western-blot (C-D, Supplementary Original western blots). NSUN2 knockdown efficiency in BCBM cells measured by immunofluorescent (E, scale bars: 100 μm), qPCR (F), and Western-blot (G-H, Original blots/gels are shown in Supplementary Original western blots). (I) The relative expression levels of 7 different methylation genes in BCBM following NSUN2 knock-down. (n = 3; Data are presented as the mean ± SD; P < 0.05)
Further comprehensive analysis of the interplay between regulators of altered expression and differentially expressed genes revealed significant upregulation of NSUN2 expression in BC tissues. Similarly, qPCR and western-blot showed that the expression of NSUN2 was increased in BCBM cells (Fig. 10B-D) implying that NSUN2 is involved in the modulation of BC proliferation and metastatic capabilities. The NSUN2 in BCBM cells were knocked down in vitro to investigate the relationship between NSUN2 and differentially expressed genes. First, the expression of NSUN2 in BCBM cells was knocked out, and then the knockout efficiency of NSUN2 was verified through GFP-positive images (Fig. 10E). It was confirmed that NSUN2 was inhibited in BCBM using western-blot and qPCR (Fig. 10F-H). After knocking down NSUN2 in BCBM, three genes with upregulated methylation lncRNAs expression ENST00000477316, ENST00000478098 and uc002gtt.1, were positively correlated with NSUN2. In contrast, ENST00000493668, ENST00000544651, ENST00000600912 and ENST00000464989, which were associated with downregulated methylation expression, were negatively correlated (P < 0.05), suggesting NSUN2 plays an essential role in the process of lncRNA methylation (Fig. 10E).
Discussion
BC is one of the most common cancers among women worldwide, with a mortality rate ranking second among female cancers [26], and cancer metastasis is the main cause of death [38, 39]. In the exploration of new treatment approaches for BC, tumor mutational burden (TMB) has attracted much attention as a biomarker for immunotherapy, but a high TMB score is associated with a reduced survival rate in BC [39]. Immune-related genes such as CCL18 are markers of poor prognosis, while B-cell infiltration is a marker of good prognosis [39]. Another study has shown that bone metastases, compared to primary breast tumors, exhibit a large number of stromal cells and an inactive immune microenvironment. This suggests that the application of combined immunotherapy in specific bone metastasis patients warrants further exploration [40]. In addition, a good systemic immune activity and the effect of neoadjuvant chemotherapy may be related to lipid metabolism in triple-negative breast cancer (TNBC) [41]. Research has demonstrated that overexpression or abnormal synthesis of proteins in breast tissue influence the occurrence and development of BC [42]. Therefore, identifying BC-related proteins may provide important insights for improving the diagnosis and treatment of BC patients.
BC is a significant health challenge due to its propensity for brain metastasis, which drastically reduces patient prognosis [43, 44]. Thus, it is imperative to study the mechanism driving brain metastasis in BC to generate ideas for developing effective treatments [45]. Epigenetic modifications, including RNA modifications, have emerged as key players in cancer metastasis [46, 47]. To date, over 250 RNA modifications have been identified, with m6A being the most prevalent internal modification of mRNA. Disruption in its regulation has been closely linked to carcinogenesis [48, 49]. For instance, m6A reader YTHDF3 was found to enhance the translation of m6A-enriched transcripts of brain metastasis-related genes [46].
The m5C is another common RNA modification in human [9, 50]. While m6A modification has received considerable attention, the role of m5C modification in cancer, particularly in BCBM, is relatively understudied. This study aimed to address this gap by comprehensively analyzing m5C methylation profiles in both BCBM and BC cells by MeRIP-seq.
Distinct patterns of m5C methylation were observed between BCBM and BC cells, indicating a potential role for these m5C modifications in driving metastatic progression. We hypothesize that m5C modifications in BCBM lncRNAs contribute to enhanced RNA stability under conditions of oxidative stress and nutrient deprivation in the brain microenvironment. For example, m5C peaks in NEAT1, a lncRNA essential for nuclear paraspeckle formation, may prevent RNA degradation, thereby facilitating the continuation of pro-survival signaling [51]. This hypothesis aligns with previous studies that have associated m5C modifications with cellular adaptation to stress. The higher number of m5C sites in BCBM cells, particularly within lncRNAs and genes associated with m5C sites, suggests a relationship between m5C methylation and BCBM. Our sequencing analysis revealed that TGFβ1 is among the genes associated with highly methylated LncRNA. Given its role in tumor biology, we found that m5C modification could influence the cytokine profile secreted by tumor cells. This, in turn, appears to regulate the migration and invasion of BC cells, potentially promoting the development of BCBM. Analysis of m5C distribution across chromosomes revealed significant differences between BCBM and BC cells, showing wide-ranging effects of m5C in BCBM and the potential importance of spatial regulation in cancer epigenetics. The differential distribution of m5C methylation sites on chromosomes may alter the features of BCBM cells by regulating gene expression and cellular functions. m5C methylation may not only regulate gene expression by altering the spatial structure of chromosomes but also influence gene expression by modulating the interaction between RNA and chromatin. For instance, m5C reader proteins (such as YBX1) can recognize m5C-modified RNA, thereby orchestrating its binding to chromatin to regulate gene expression and affect tumor progression [52]. The differences in motif preferences for m5C peaks between BCBM and BC cells may be attributed to the concerted action of multiple molecular mechanisms and regulatory factors. The observed motif preference for “CCAGSCUG” in BCBM may be linked to the upregulation of methyltransferases, such as NSUN2. Our validation confirmed elevated NSUN2 expression in BCBM, which preferentially targets cytosine residues within specific sequence contexts. Cluster analysis uncovered consistent methylation patterns within groups but significant differences between BCBM and BC cells. Verification of methylated lncRNAs in BCBM corroborated sequencing results.
LncRNAs are implicated in diverse biological processes and pathological processes [53]. A previous study found that lncRNAs also regulate cancer occurrence, development, and metastasis [54]. In BC mouse models, inactivating the purported metastasis-promoting lncRNA MALAT1 enhances lung metastasis. Conversely, knocking out MALAT1 in BC cells increases their metastatic potential [55]. The exosomes of BCBM cells breach the blood-brain barrier by transferring lncRNA GS1-600g8.5 [56]. The role of lncRNAs in cancer metastasis remains unclear despite their effect in m5C-mediated BCBM. Our findings revealed dysregulated expression of m5C-methylated lncRNA genes in BCBM compared to BC cells. We hypothesize that m5C modification may influence the expression of lncRNAs in BCBM by modulating their stability or function, thereby facilitating BCBM. Previous studies have demonstrated that NSUN2 promotes the upregulation of lncRNA NR_033928 in gastric cancer (GC) through its stabilization via m5C modification. NR_033928 promotes GC proliferation and inhibits apoptosis by enhancing glutaminase (GLS) expression [57]. The precise mechanisms underlying the differential expression of lncRNAs in BCBM need to be further investigated.
The differentially expressed lncRNAs may influence tumor metastasis and progression by regulating specific oncogenic pathways, such as the MAPK signaling pathway and epithelial-mesenchymal transition (EMT). For example, the lncRNA SNHG25, which is linked to the MAPK signaling pathway, promotes this pathway by activating MAP2K2 upon overexpression. This activation enhances the invasive capacity of glioma cells, thereby facilitating tumor progression [58]. LncRNA PVT1 promote tumor cell invasion and metastasis by regulating the expression of EMT-related transcription factors, such as TGF-β1 [59]. In this study, we found two significantly upregulated lncRNAs (ENST00000483869 and ENST00000564176), which were associated with MAPK3 and TGFβ1 genes, and involved in the MAPK signaling pathway and EMT process, respectively. In BCBM, the interaction between lncRNAs and m5C methylation significantly affects tumor metastasis and progression by regulating these oncogenic pathways. Through the integration of the lncRNA transcriptome profile with m5C methylation sequencing data, we identified a notable correlation between the two. Specifically, m5C methylation appears to influence the interaction between lncRNA and RNA-binding proteins, such as YBX1, by modifying the secondary structure of lncRNA. This alteration may, in turn, regulate its functional activity [60]. The interaction between lncRNA and m5C methylation may carry some therapeutic benefits in controlling BCBM. Specifically, inhibition of metastasis-promoting lncRNAs (such as DIAPH2-AS1) and simultaneously targeting m5C methyltransferases (such as NSUN2) may more effectively suppress tumor metastasis [61].
The m5C is also regulated by corresponding writers, readers, and erasers [13, 62, 63]. These regulatory factors are involved in the pathological process of BC by dynamically modulating the m5C modification level, affecting the activity, stability and function of lncRNAs [60].The analysis revealed differential expression patterns of certain m5C RNA methylation regulators in BC and adjunct tissues, supporting the relevance of m5C methylation in cancer progression. The elevated expression of NSUN2, NSUN5, YBX1, and ALYREF in BC tissues suggests their potential to regulate metastatic processes. The NSUN2 methyltransferase is a typical nucleolar protein for tRNA, mRNA, and lncRNA [64]. NSUN2 was implicated in various cellular processes such as proliferation [65], ageing, the cell cycle, differentiation of epidermal stem cells, differentiation of neural stem cells, and testicular differentiation [66, 67]. Furthermore, NSUN2, a major m5C-modifying methyltransferase (writer), plays a key role in the methylation of mRNA and lncRNA. Its upregulation has been linked to cancer development. In BC, NSUN2 enhances the stability of lncRNA through m5C modification, facilitating its binding to target genes and thereby promoting the invasion and metastasis of tumor cells [68]. NSUN2 forms functional complexes with m5C-recognizing proteins, such as YBX1, to regulate the expression of target genes. This collaboration further enhances the proliferation and metastatic ability of BC cells [69]. The up- or down-regulation of the different types of regulators and their corresponding changes in NSUN2-knockdown BCBM cells implicates their potential role in driving metastatic processes. Previous studies have shown that differentially expressed genes (DEGs) are significantly enriched in extracellular matrix (ECM)-receptor interaction, TGF-β, and PI3K-Akt signaling pathways after NSUN2 knockout. For instance, the down-regulation of genes such as TGFB1, THBS1 and LOXL2 results in the suppression of the ECM remodeling capacity, thereby inhibiting tumor invasion [70]. Combined with our research results, we infer that NSUN2 deficiency induces significant dysregulation of multiple metastasis-related genes to suppress tumor cell growth. Using m5C sequencing analysis, we identified two oncogenic lncRNAs in BC, ENST00000461842 and ENST00000564176, which are highly expressed in tumor tissues. Their expression levels are significantly negatively correlated with patient prognosis, suggesting their potential markers for BC metastasis.
The m5C modification contributes to the RNA nucleation, cell differentiation processes [71], regulating stem cell function and stress [72]. Functional enrichment analysis identified the diverse cellular functions and pathways modulating m5C methylation. The enrichment of hyper-methylated genes in cancer-related pathways, such as the MAPK and EMT signaling pathway, underscores the potential oncogenic role of m5C methylation in BCBM. Conversely, the association of hypo-methylated genes with pathways such as the Hippo signaling pathway suggests a regulatory role in tumor suppressor pathways. This indicates that the coordinated regulation of key pathways by methylation balance plays a significant role in tumor metastasis. However, the underlying mechanism requires further study.
As an important mechanism of the epitranscriptome, the m5C methylation modification of RNA plays a key role in the progression of breast cancer, particularly during the metastatic stage, offering a new direction for targeted therapy. Studies have shown that NSUN2, the main m5C methyltransferase, is abnormally overexpressed in BC. It enhances the mRNA stability of the target gene HGH1 (FAM203) through m5C modification, thereby promoting tumor cell proliferation, migration, and invasion [58]. Targeting the NSUN2/YBX1 axis, developing m5C-related biomarkers, and integrating stratified treatment strategies offer new perspectives for epigenetic therapy. Small molecule inhibitors that disrupt this complex may inhibit BC metastasis. Furthermore, repurposing drugs and administering various vitamins, such as vitamin D, as prophylactic agents with modulatory effects could have a positive impact on BC [73]. The dysregulation of lncRNA expression observed in BCBM cells underscores the importance of lncRNAs in cancer progression. The dysregulated lncRNAs are potential biomarkers for monitoring metastatic progression and developing drug targets for BCBM.
Conclusion
This study highlights the significance of m5C methylation in lncRNAs within the context of BCBM. It emphasizes the necessity for further mechanistic investigations to elucidate the functional implications of its dysregulation in cancer metastasis. These findings lay the foundation for developing targeted therapies for preventing BC metastasis and progression.
Data availability
Sequence data that support the findings of this study have been deposited in the NCBI GEO Archive with the submission ID GSE246721.
Change history
07 April 2025
A Correction to this paper has been published: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-025-14042-x
Abbreviations
- BC:
-
Breast Cancer
- BCBM:
-
Breast Cancer Brain Metastases
- m5C:
-
5-Methylcytosine
- LncRNA:
-
Long Non-coding RNA
- MeRIP-seq:
-
Methylated RNA Immunoprecipitation Next-generation Sequencing
- RT-PCR:
-
Quantitative Real-time PCR
- GO:
-
Gene Ontology
- KEGG:
-
Kyoto Encyclopedia of Genes and Genomes
- BP:
-
Biological Processes
- CC:
-
Cell Components
- MF:
-
Molecular Functions
- ANKMY1:
-
Ankyrin Repeat and MYND Domain Containing 1
- KCNAB2:
-
Potassium Voltage-gated Channel, Shaker-related Subfamily, Beta Member 2
- HECW1:
-
HECT, C2 and WW Domain Containing E3 Ubiquitin Protein Ligase 1
- SMARCD3:
-
SWI/SNF Related, Matrix Associated, Actin Dependent Regulator of Chromatin, Subfamilyd, Member 3
- CERS4:
-
Ceramide Synthase 4
- LLGL2:
-
LLGL Scribble Cell Polarity Complex Component 2
- SHROOM2:
-
Shroom Family Member 2
- ACACB:
-
Acetyl-CoA Carboxylase Beta
- SPEG:
-
Striated Muscle Enriched Protein Kinase
- NSUN2:
-
NOP2/Sun RNA methyltransferase 2
- NSUN5:
-
NOP2/Sun RNA methyltransferase 5
- YBX1:
-
Y-Box Binding Protein 1
- ALYREF:
-
Aly/REF Export Factor
- DNMT3B:
-
DNA Methyltransferase 3 Aeta
- DNMT3A:
-
DNA Methyltransferase 3 Alpha
- DNMT1:
-
DNA Methyltransferase 1
- TET3:
-
Tet Methylcytosine Dioxygenase 3
- NSUN3:
-
NOP2/Sun RNA methyltransferase 3
- NSUN4:
-
NOP2/Sun RNA methyltransferase 4
- NSUN5:
-
NOP2/Sun RNA methyltransferase 5
- NSUN6:
-
NOP2/Sun RNA methyltransferase 6
- TET2:
-
Tet Methylcytosine Dioxygenase 2
References
Yuan H, Liu J, Zhao L, Wu P, Chen G, Chen Q, Shen P, Yang T, Fan S, Xiao B, et al. Prognostic risk model and tumor immune environment modulation of m5C-Related LncRNAs in pancreatic ductal adenocarcinoma. Front Immunol. 2021;12:800268.
Delaunay S, Frye M. RNA modifications regulating cell fate in cancer. Nat Cell Biol. 2019;21(5):552–9.
Yang Z, Wang T, Wu D, Min Z, Tan J, Yu B. RNA N6-methyladenosine reader IGF2BP3 regulates cell cycle and angiogenesis in colon cancer. J Exp Clin Cancer Res. 2020;39(1):203.
Liu J, Chen C, Wang Y, Qian C, Wei J, Xing Y, Bai J. Comprehensive of N1-Methyladenosine modifications patterns and immunological characteristics in ovarian cancer. Front Immunol. 2021;12:746647.
Chen X, Li A, Sun BF, Yang Y, Han YN, Yuan X, Chen RX, Wei WS, Liu Y, Gao CC, et al. 5-methylcytosine promotes pathogenesis of bladder cancer through stabilizing mRNAs. Nat Cell Biol. 2019;21(8):978–90.
Li Q, Ni Y, Zhang L, Jiang R, Xu J, Yang H, Hu Y, Qiu J, Pu L, Tang J, et al. HIF-1alpha-induced expression of m6A reader YTHDF1 drives hypoxia-induced autophagy and malignancy of hepatocellular carcinoma by promoting ATG2A and ATG14 translation. Signal Transduct Target Ther. 2021;6(1):76.
Dixit D, Prager BC, Gimple RC, Poh HX, Wang Y, Wu Q, Qiu Z, Kidwell RL, Kim LJY, Xie Q, et al. The RNA m6A reader YTHDF2 maintains oncogene expression and is a targetable dependency in glioblastoma stem cells. Cancer Discov. 2021;11(2):480–99.
Hamdy NM, Gabr MM, AbdelHamid SG, Swellam MM. Scrutinizing the landscape of DNA methylation epigenetic face(s) in glioblastomas. Nutraceutical Fruits and Foods for Neurodegenerative Disorders, Academic Press. 2024:467–504.
Liu J, Xiao S, Chen J, Lou W, Chen X. A comprehensive analysis for expression, diagnosis, and prognosis of m(5)C regulator in breast cancer and its ncRNA-mRNA regulatory mechanism. Front Genet. 2022;13:822721.
Li M, Tao Z, Zhao Y, Li L, Zheng J, Li Z, Chen X. 5-methylcytosine RNA methyltransferases and their potential roles in cancer. J Translational Med. 2022;20(1):214.
Shen Q, Zhang Q, Shi Y, Shi Q, Jiang Y, Gu Y, Li Z, Li X, Zhao K, Wang C, et al. Tet2 promotes pathogen infection-induced myelopoiesis through mRNA oxidation. Nature. 2018;554(7690):123–7.
Wang Y, Wei J, Feng L, Li O, Huang L, Zhou S, Xu Y, An K, Zhang Y, Chen R, et al. Aberrant m5C hypermethylation mediates intrinsic resistance to gefitinib through NSUN2/YBX1/QSOX1 axis in EGFR-mutant non-small-cell lung cancer. Mol Cancer. 2023;22(1):81.
Cheng JX, Chen L, Li Y, Cloe A, Yue M, Wei J, Watanabe KA, Shammo JM, Anastasi J, Shen QJ, et al. RNA cytosine methylation and methyltransferases mediate chromatin organization and 5-azacytidine response and resistance in leukaemia. Nat Commun. 2018;9(1):1163.
Zhang Q, Zheng Q, Yu X, He Y, Guo W. Overview of distinct 5-methylcytosine profiles of messenger RNA in human hepatocellular carcinoma and paired adjacent non-tumor tissues. J Translational Med. 2020;18(1):245.
David R, Burgess A, Parker B, Li J, Pulsford K, Sibbritt T, Preiss T, Searle IR. Transcriptome-Wide mapping of RNA 5-Methylcytosine in Arabidopsis mRNAs and noncoding RNAs. Plant Cell. 2017;29(3):445–60.
Yang X, Yang Y, Sun BF, Chen YS, Xu JW, Lai WY, Li A, Wang X, Bhattarai DP, Xiao W, et al. 5-methylcytosine promotes mRNA export - NSUN2 as the methyltransferase and ALYREF as an m(5)C reader. Cell Res. 2017;27(5):606–25.
Huang T, Chen W, Liu J, Gu N, Zhang R. Genome-wide identification of mRNA 5-methylcytosine in mammals. Nat Struct Mol Biol. 2019;26(5):380–8.
Abaza T, El-Aziz MKA, Daniel KA, Karousi P, Papatsirou M, Fahmy SA, Hamdy NM, Kontos CK, Youness RA. Emerging role of circular RNAs in hepatocellular carcinoma immunotherapy. Int J Mol Sci 2023, 24(22).
Sun Z, Xue S, Zhang M, Xu H, Hu X, Chen S, Liu Y, Guo M, Cui H. Aberrant NSUN2-mediated m(5)C modification of H19 LncRNA is associated with poor differentiation of hepatocellular carcinoma. Oncogene. 2020;39(45):6906–19.
Zhou C, Yi C, Yi Y, Qin W, Yan Y, Dong X, Zhang X, Huang Y, Zhang R, Wei J, et al. LncRNA PVT1 promotes gemcitabine resistance of pancreatic cancer via activating Wnt/beta-catenin and autophagy pathway through modulating the miR-619-5p/Pygo2 and miR-619-5p/ATG14 axes. Mol Cancer. 2020;19(1):118.
Qian X, Zhao J, Yeung PY, Zhang QC, Kwok CK. Revealing LncRNA structures and interactions by Sequencing-Based approaches. Trends Biochem Sci. 2019;44(1):33–52.
Hamdy NM, Zaki MB, Abdelmaksoud NM, Ismail RA, Abd-Elmawla MA, Rizk NI, Fathi D, Abulsoud AI. Insights into the genetic and epigenetic mechanisms governing X-chromosome-linked-miRNAs expression in cancer; a step-toward NcRNA precision. Int J Biol Macromol. 2025;289:138773.
Bridges MC, Daulagala AC, Kourtidis A. LNCcation: LncRNA localization and function. J Cell Biol 2021, 220(2).
Sun T, Wu Z, Wang X, Wang Y, Hu X, Qin W, Lu S, Xu D, Wu Y, Chen Q, et al. LNC942 promoting METTL14-mediated m(6)A methylation in breast cancer cell proliferation and progression. Oncogene. 2020;39(31):5358–72.
Achrol AS, Rennert RC, Anders C, Soffietti R, Ahluwalia MS, Nayak L, Peters S, Arvold ND, Harsh GR, Steeg PS, et al. Brain metastases. Nat Rev Dis Primers. 2019;5(1):5.
Hamdy NM, El-Sisi MG, Ibrahim SM, ElNokoudy H, Hady AA, Abd-Ellatef GEF, Sallam AM, Barakat BM. In Silico analysis and comprehensive review of circular-RNA regulatory roles in breast diseases; a step-toward non-coding RNA precision. Pathol Res Pract. 2024;263:155651.
Wang Y, Ye F, Liang Y, Yang Q. Breast cancer brain metastasis: insight into molecular mechanisms and therapeutic strategies. Br J Cancer. 2021;125(8):1056–67.
Wang S, Liang K, Hu Q, Li P, Song J, Yang Y, Yao J, Mangala LS, Li C, Yang W, et al. JAK2-binding long noncoding RNA promotes breast cancer brain metastasis. J Clin Investig. 2017;127(12):4498–515.
Huang Z, Pan J, Wang H, Du X, Xu Y, Wang Z, Chen D. Prognostic significance and tumor immune microenvironment heterogenicity of m5C RNA methylation regulators in Triple-Negative breast cancer. Front Cell Dev Biol. 2021;9:657547.
Fu B, Zhang A, Li M, Pan L, Tang W, An M, Liu W, Zhang J. Circular RNA profile of breast cancer brain metastasis: identification of potential biomarkers and therapeutic targets. Epigenomics. 2018;10(12):1619–30.
Dun MD, Chalkley RJ, Faulkner S, Keene S, Avery-Kiejda KA, Scott RJ, Falkenby LG, Cairns MJ, Larsen MR, Bradshaw RA, et al. Proteotranscriptomic profiling of 231-BR breast cancer cells: identification of potential biomarkers and therapeutic targets for brain metastasis. Mol Cell Proteom. 2015;14(9):2316–30.
Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3’ UTRs and near stop codons. Cell. 2012;149(7):1635–46.
Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.
Cheng J, Metge F, Dieterich C. Specific identification and quantification of circular RNAs from sequencing data. Bioinformatics. 2016;32(7):1094–6.
Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12(4):357–60.
Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers RM, Brown M, Li W, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 2008;9(9):R137.
Shen L, Shao NY, Liu X, Maze I, Feng J, Nestler EJ. DiffReps: detecting differential chromatin modification sites from ChIP-seq data with biological replicates. PLoS ONE. 2013;8(6):e65598.
DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Goding Sauer A, Jemal A, Siegel RL. Breast cancer statistics, 2019. Cancer J Clin. 2019;69(6):438–51.
Cui S, Feng J, Tang X, Lou S, Guo W, Xiao X, Li S, Chen X, Huan Y, Zhou Y, et al. The prognostic value of tumor mutation burden (TMB) and its relationship with immune infiltration in breast cancer patients. Eur J Med Res. 2023;28(1):90.
Chao X, Zhang Y, Zheng C, Huang Q, Lu J, Pulver EM, Houthuijzen J, Hutten S, Luo R, He J, et al. Metastasis of breast cancer to bones alters the tumor immune microenvironment. Eur J Med Res. 2023;28(1):119.
Goto W, Kashiwagi S, Takada K, Asano Y, Ogisawa K, Morisaki T, Shibutani M, Tanaka H, Maeda K. Clinical verification of the relationship between serum lipid metabolism and immune activity in breast cancer patients treated with neoadjuvant chemotherapy. Eur J Med Res. 2023;28(1):2.
Xu J, Yu C, Zeng X, Tang W, Xu S, Tang L, Huang Y, Sun Z, Yu T. Visualization of breast cancer-related protein synthesis from the perspective of bibliometric analysis. Eur J Med Res. 2023;28(1):461.
Morgan AJ, Giannoudis A, Palmieri C. The genomic landscape of breast cancer brain metastases: a systematic review. Lancet Oncol. 2021;22(1):e7–17.
Mills MN, Figura NB, Arrington JA, Yu HM, Etame AB, Vogelbaum MA, Soliman H, Czerniecki BJ, Forsyth PA, Han HS, et al. Management of brain metastases in breast cancer: a review of current practices and emerging treatments. Breast Cancer Res Treat. 2020;180(2):279–300.
Chiang YF, Huang KC, Chen HY, Hamdy NM, Huang TC, Chang HY, Shieh TM, Huang YJ, Hsia SM. Hinokitiol Inhibits Breast Cancer Cells In Vitro Stemness-Progression and Self-Renewal with Apoptosis and Autophagy Modulation via the CD44/Nanog/SOX2/Oct4 Pathway. International journal of molecular sciences 2024, 25(7).
Chang G, Shi L, Ye Y, Shi H, Zeng L, Tiwary S, Huse JT, Huo L, Ma L, Ma Y, et al. YTHDF3 induces the translation of m(6)A-Enriched gene transcripts to promote breast cancer brain metastasis. Cancer Cell. 2020;38(6):857–e871857.
Wang P, Wu M, Tu Z, Tao C, Hu Q, Li K, Zhu X, Huang K. Identification of RNA: 5-Methylcytosine Methyltransferases-Related signature for predicting prognosis in glioma. Front Oncol. 2020;10:1119.
Li Y, Xiao J, Bai J, Tian Y, Qu Y, Chen X, Wang Q, Li X, Zhang Y, Xu J. Molecular characterization and clinical relevance of m(6)A regulators across 33 cancer types. Mol Cancer. 2019;18(1):137.
Deng X, Su R, Weng H, Huang H, Li Z, Chen J. RNA N(6)-methyladenosine modification in cancers: current status and perspectives. Cell Res. 2018;28(5):507–17.
Han X, Wang M, Zhao YL, Yang Y, Yang YG. RNA methylations in human cancers. Sem Cancer Biol. 2021;75:97–115.
Lin G, Cai H, Hong Y, Yao M, Ye W, Li W, Liang W, Feng S, Lv Y, Ye H, et al. Implications of m(5)C modifications in ribosomal proteins on oxidative stress, metabolic reprogramming, and immune responses in patients with mid-to-late-stage head and neck squamous cell carcinoma: insights from nanopore sequencing. Heliyon. 2024;10(14):e34529.
Chen B, Deng Y, Hong Y, Fan L, Zhai X, Hu H, Yin S, Chen Q, Xie X, Ren X, et al. Metabolic recoding of NSUN2-Mediated m(5)C modification promotes the progression of colorectal cancer via the NSUN2/YBX1/m(5)C-ENO1 positive feedback loop. Adv Sci. 2024;11(28):e2309840.
Beermann J, Piccoli MT, Viereck J, Thum T. Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches. Physiol Rev. 2016;96(4):1297–325.
Schmitt AM, Chang HY. Long noncoding RNAs in cancer pathways. Cancer Cell. 2016;29(4):452–63.
Kim J, Piao HL, Kim BJ, Yao F, Han Z, Wang Y, Xiao Z, Siverly AN, Lawhon SE, Ton BN, et al. Long noncoding RNA MALAT1 suppresses breast cancer metastasis. Nat Genet. 2018;50(12):1705–15.
Lu Y, Chen L, Li L, Cao Y. Exosomes Derived from Brain Metastatic Breast Cancer Cells Destroy the Blood-Brain Barrier by Carrying lncRNA GS1-600G8.5. BioMed research international, 2020:7461727.
Fang L, Huang H, Lv J, Chen Z, Lu C, Jiang T, Xu P, Li Y, Wang S, Li B, et al. m5C-methylated LncRNA NR_033928 promotes gastric cancer proliferation by stabilizing GLS mRNA to promote glutamine metabolism reprogramming. Cell Death Dis. 2023;14(8):520.
Wu Z, Lun P, Ji T, Niu J, Sun X, Liu X, Xu J. LncRNA SNHG25 promotes glioma progression through activating MAPK signaling. Mol Neurobiol. 2022;59(11):6993–7005.
Zhang X, Feng W, Zhang J, Ge L, Zhang Y, Jiang X, Peng W, Wang D, Gong A, Xu M. Long non–coding RNA PVT1 promotes epithelial–mesenchymal transition via the TGF–beta/Smad pathway in pancreatic cancer cells. Oncol Rep. 2018;40(2):1093–102.
Song H, Zhang J, Liu B, Xu J, Cai B, Yang H, Straube J, Yu X, Ma T. Biological roles of RNA m(5)C modification and its implications in cancer immunotherapy. Biomark Res. 2022;10(1):15.
Li Y, Xia Y, Jiang T, Chen Z, Shen Y, Lin J, Xie L, Gu C, Lv J, Lu C, et al. Long noncoding RNA DIAPH2-AS1 promotes neural invasion of gastric cancer via stabilizing NSUN2 to enhance the m5C modification of NTN1. Cell Death Dis. 2023;14(4):260.
Boo SH, Kim YK. The emerging role of RNA modifications in the regulation of mRNA stability. Exp Mol Med. 2020;52(3):400–8.
He Y, Yu X, Li J, Zhang Q, Zheng Q, Guo W. Role of m(5)C-related regulatory genes in the diagnosis and prognosis of hepatocellular carcinoma. Am J Translational Res. 2020;12(3):912–22.
Chellamuthu A, Gray SG. The RNA methyltransferase NSUN2 and its potential roles in cancer. Cells 2020, 9(8).
Xing J, Yi J, Cai X, Tang H, Liu Z, Zhang X, Martindale JL, Yang X, Jiang B, Gorospe M, et al. NSun2 promotes cell growth via elevating Cyclin-Dependent kinase 1 translation. Mol Cell Biol. 2015;35(23):4043–52.
Sajini AA, Choudhury NR, Wagner RE, Bornelov S, Selmi T, Spanos C, Dietmann S, Rappsilber J, Michlewski G, Frye M. Loss of 5-methylcytosine alters the biogenesis of vault-derived small RNAs to coordinate epidermal differentiation. Nat Commun. 2019;10(1):2550.
Hu Y, Chen C, Tong X, Chen S, Hu X, Pan B, Sun X, Chen Z, Shi X, Hu Y, et al. NSUN2 modified by SUMO-2/3 promotes gastric cancer progression and regulates mRNA m5C methylation. Cell Death Dis. 2021;12(9):842.
Wang X, Liu D, Hua K, Fang L. LncRNA HOST2 promotes NSUN2-mediated breast cancer progression via interaction with ELAVL1. Cell Signal. 2024;117:111112.
Zhang X, An K, Ge X, Sun Y, Wei J, Ren W, Wang H, Wang Y, Du Y, He L, et al. NSUN2/YBX1 promotes the progression of breast cancer by enhancing HGH1 mRNA stability through m(5)C methylation. Breast cancer Research: BCR. 2024;26(1):94.
Zou S, Huang Y, Yang Z, Zhang J, Meng M, Zhang Y, Feng J, Sun R, Li W, Wang W, et al. NSUN2 promotes colorectal cancer progression by enhancing SKIL mRNA stabilization. Clin Translational Med. 2024;14(3):e1621.
Flores JV, Cordero-Espinoza L, Oeztuerk-Winder F, Andersson-Rolf A, Selmi T, Blanco S, Tailor J, Dietmann S, Frye M. Cytosine-5 RNA methylation regulates neural stem cell differentiation and motility. Stem Cell Rep. 2017;8(1):112–24.
Blanco S, Bandiera R, Popis M, Hussain S, Lombard P, Aleksic J, Sajini A, Tanna H, Cortes-Garrido R, Gkatza N, et al. Stem cell function and stress response are controlled by protein synthesis. Nature. 2016;534(7607):335–40.
Chen YC, Chiang YF, Lin YJ, Huang KC, Chen HY, Hamdy NM, Huang TC, Chang HY, Shieh TM, Huang YJ et al. Effect of vitamin D supplementation on primary dysmenorrhea: A systematic review and Meta-Analysis of randomized clinical trials. Nutrients 2023, 15(13).
Acknowledgements
The authors would like to thank all the reviewers who participated in the review and MJEditor (www.mjeditor.com) for its linguistic assistance during the preparation of this manuscript.
Funding
This work was supported by the National Natural Science Foundation of China (Grant no. 81702884), Natural Science Foundation of Shandong Province (Grant no. ZR2022MH272 and ZR2023QH115), Medicine and Health Science and Technology Foundation of Shandong Province (Grant no. 202111000399, 202203020733 and 202202080721), Liaocheng Key R&D Project (Grant no. 2022YDSF31 and Grant no. 2022YDSF35) and Science Foundation of Liaocheng People’s Hospital (Grant no. LYQN201901 and LYQN201902).
Author information
Authors and Affiliations
Contributions
S W, B F, J.R.G: Conceptualization, Formal analysis, Writingoriginal draft. D Y, X.M.X, M L, Y.F.Z: Data Collection, Methodology, Software. G.H.G, Y.J.L, L P, S C, X.H.Z: Formal analysis and interpretation. S W, M A, B F: Conceptualization, Funding acquisition, Project administration. S W, A.Q.Z, B F: Writing - review & editing, Resources. All the authors agreed with the final version of the manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
All human samples studies have been approved by the Ethics Committee of Liaocheng People’s Hospital (2022261), and all participants have signed informed consent forms.
Consent to for publication
All authors have read, approved, and agreed to publish this manuscript.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
The original version of this article was revised: Figure 9 was a duplication of figure 10. This has been corrected.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Wang, S., Guo, J., Xian, X. et al. Distinct 5-methylcytosine profiles of LncRNA in breast cancer brain metastasis. BMC Cancer 25, 557 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-025-13948-w
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-025-13948-w