International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Skin cutaneous melanoma (SKCM) is a highly aggressive malignancy with strong invasive potential and a markedly high mortality rate (0.6% of all patients with stage-4 melanoma) (1). It tends to progress rapidly, frequently metastasizing to regional lymph nodes and distant organs. The risk of recurrence is positively associated with the clinical stage at diagnosis (1,2). Research indicates that early-stage melanoma is often treatable with radical surgical excision alone, without the need for adjuvant therapy (3). However, in patients with lymph node involvement, the recurrence rate remains high even after complete resection. Notably, the 5-year survival rate for patients with metastatic SKCM is only ~27% (2). Therefore, elucidating the mechanisms underlying melanoma metastasis is essential for improving early diagnosis, prognostic evaluation and the development of novel targeted therapies. A recent comprehensive review highlighted the evolving molecular subclasses of melanoma and emerging roles of vitamin D signaling in tumor-immune crosstalk (4).
The skin functions as an integrated neuro-immuno-endocrine organ that senses environmental cues and coordinates systemic homeostasis (5). Ultraviolet radiation, beyond its carcinogenic potential, modulates cutaneous vitamin D synthesis, neuropeptide release and local immunoregulation, a concept termed ‘photo-neuro-immuno-endocrinology’ (6). These layers of regulation provide additional context for understanding melanoma biology. N6-methyladenosine (m6A), the most abundant internal modification of eukaryotic mRNA, serves a pivotal role in regulating mRNA stability, protein translation, viral infection and embryonic development (7). This modification is dynamically reversible and is orchestrated by three main classes of regulators: ‘Writers’ [methyltransferases such as Vir like m6A methyltransferase associated (VIRMA), methyltransferase m6A complex catalytic subunit (METTL)3/14, RNA binding motif protein (RBM)15/15B and Wilms tumor 1 associated protein (WTAP)], ‘erasers’ [demethylases including fat mass and obesity-associated protein (FTO) and AlkB homolog 5, RNA demethylase (ALKBH5)] and ‘readers’ [m6A-binding proteins such as heterogeneous nuclear ribonucleoprotein (HNRNP)C, HNRNPA2B1, YTH m6A RNA binding protein (YTHD)F1/2/3 and YTHDC1/2] (8). Extensive research has reported that m6A modifications are involved in a wide range of biological processes, including the heat shock response, tissue development, DNA damage repair, and stem cell self-renewal and differentiation (9–12). Moreover, dysregulation of m6A regulators has been increasingly implicated in tumor initiation, progression, metastasis and resistance to therapy (13).
Aberrant m6A modification has been increasingly recognized as a critical contributor to cancer initiation and progression. In acute myeloid leukemia (AML), for example, the m6A methyltransferases METTL14 and METTL3 are markedly upregulated and notably influence patient prognosis (14,15). Similarly, the m6A demethylases FTO and ALKBH5 are overexpressed in several tumor types, where they enhance cancer stemness and promote oncogenic processes (16–18). In uveal melanoma and conjunctival melanoma, m6A-mediated methylation of β-secretase 2 has been reported to trigger intracellular calcium release, thereby accelerating tumor progression (19). Moreover, Dahal et al (20) reported that METTL3 is markedly upregulated in melanoma and promotes cellular invasiveness by upregulating MMP2 and N-cadherin expression. Despite growing evidence supporting the role of m6A modifications in tumor development and prognosis, the specific regulatory mechanisms by which m6A influences metastasis and invasion in SKCM remain poorly understood.
Therefore, the aim of the present study was to uncover the molecular mechanisms by which m6A modification contributes to SKCM metastasis and to provide a theoretical foundation for the development of novel therapeutic targets and prognostic biomarkers. To elucidate the potential role of m6A modification in SKCM metastasis, the present study integrated expression profiles from primary and metastatic SKCM samples in the Gene Expression Omnibus (GEO) database to systematically identify metastasis-related differentially expressed genes (DEGs) and associated signaling pathways. A prognostic prediction model was subsequently constructed using survival data from The Cancer Genome Atlas (TCGA)-SKCM cohort. Key mRNAs strongly associated with both m6A regulation and patient prognosis were further screened. In addition, the present study explored their interactions within a long noncoding (lnc)RNA-micro (mi)RNA-mRNA competing endogenous (ce)RNA network and analyzed their correlation with immune cell infiltration.
Gene expression data for 46 metastatic SKCM samples, 77 primary SKCM samples and 21 normal skin samples were obtained from the GEO database (https://www.ncbi.nlm.nih.gov/geo/), specifically from the GSE8401 (21), GSE15605 (22), GSE46517 (23) and GSE65904 (24) datasets. Raw data were log-transformed and normalized prior to downstream analyses (Fig. S1). A summary of patient characteristics is presented in Tables I and SI. Moreover, RNA sequencing (RNA-seq) data from the TCGA-SKCM dataset were retrieved from the University of California, Santa Cruz Xena platform (https://xena.ucsc.edu/) and converted to transcripts per million (TPM) using the ‘count2tpm’ R package (version 4.3.0; http://www.r-project.org/). Following quality control and filtering for complete survival data, 461/472 patients in the TCGA-SKCM dataset with matched RNA-seq and survival information were included in the subsequent analyses.
In addition, seven pairs of SKCM and adjacent normal tissue samples were obtained from patients who underwent surgical treatment at the Institute of Dermatology, Peking Union Medical College and Chinese Academy of Medical Sciences (Nanjing, China) between 2018 and 2021. A total of 7 patients, aged 26–92 years (4 males and 3 females) were included. All paraffin-embedded tissue sections were confirmed as cutaneous melanoma by two or more dermatopathologists, with cases declining to participate excluded. The fresh tumor and adjacent tissues were promptly frozen and stored at −80°C for subsequent analyses. The present study was approved by the Ethics Committee of the Institute of Dermatology, Peking Union Medical College and Chinese Academy of Medical Sciences (approval no. 2017-KY-044), and all procedures were performed in accordance with approved institutional guidelines. Written informed consent was obtained from all participants.
TCGA-SKCM cases were classified into primary and metastatic groups based on patient clinical information. Least absolute shrinkage and selection operator (LASSO) Cox regression analysis was performed using the ‘glmnet’ package in R (version 4.3.0; http://www.r-project.org/), with the optimal λ value determined using 10-fold cross-validation (25). This approach identified the most predictive gene set, which, combined with primary-metastatic status, was used to construct a risk score model. Individual risk scores were calculated for each patient, who were then stratified into high- and low-risk groups using the median risk score as the cutoff. Survival differences between the two groups were assessed using Kaplan-Meier analysis and compared using the log-rank test, with significance defined as P<0.05. The predictive accuracy of the risk model was evaluated by receiver operating characteristic (ROC) curve analysis, and model performance was quantified using the area under the curve. Internal robustness was assessed using 10-fold cross-validation of the LASSO-Cox model. The resulting risk scores for each fold and the coefficients selected at the λ_min and λ_1se penalty parameters are summarized in Table SII. To address potential violations of the proportional hazard rate assumption due to survival curve crossover for certain genes, the analyzed time range was restricted to periods preceding the crossover points. Survival analyses were re-performed within these truncated time intervals to ensure robustness of the log-rank test results (26).
Based on the risk scores generated by the machine learning model, the TCGA-SKCM cohort was stratified into high- and low-risk groups. Differential expression analyses were performed to compare gene expression profiles among primary tumors, metastatic tumors and normal tissues from the GEO datasets, as well as between the high- and low-risk groups in the TCGA cohort. For the TCGA-SKCM RNA-seq data, raw read-count matrices were analyzed using the ‘DESeq2’ package (R version 4.3.0; http://www.r-project.org/) and the median-of-ratios procedure was used to normalize library size and estimate dispersion, followed by Wald tests to identify DEGs. Genes with adjusted P<0.05 and |log2FoldChange (FC)|>1 were deemed significant. TPM values, calculated using the count2tpm tool (R version 4.3.0; http://www.r-project.org/), were used only for downstream visualization (27). For the GEO datasets, raw or processed expression matrices were analyzed using the ‘limma’ package (R version 4.3.0; http://www.r-project.org/) with empirical Bayes moderation to identify DEGs, applying the same adjusted P-value and fold-change thresholds. DEGs identified from multiple GEO datasets were then integrated using the ‘RobustRankAggreg’ (RRA) package (R version 4.3.0; http://www.r–project.org/) (28) to obtain a robust DEG list.
For single-gene differential expression analyses, thresholds were set at |log2FC|>1 and adjusted P<0.05 to identify differentially expressed lncRNAs and mRNAs associated with each of the four candidate genes. Overlapping lncRNAs and mRNAs were determined using the ‘VennDiagram’ package in R (version 4.3.0; http://www.r-project.org/). Visualization of DEGs was achieved using volcano plots and heatmaps generated using the ‘ggplot2’ package (R version 4.3.0; http://www.r-project.org/).
To assess the biological processes and signaling pathways underlying SKCM metastasis, enrichment analyses were performed on DEGs identified from GEO datasets comparing primary and metastatic melanoma, and co-expressed DEGs associated with the risk-related genes [keratin (KRT)17, plakophilin 1 (PKP1), cadherin 3 (CDH3) and cellular retinoic acid binding protein 2 (CRABP2)]. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the Metascape database (http://metascape.org/) and the ‘clusterProfiler’ R package (version 4.3.0; http://www.r–project.org/) (29). The top 20 enriched GO and KEGG terms were visualized using network analysis in Cytoscape 3.6.1 (30). Additionally, gene set enrichment analysis (GSEA) was performed using clusterProfiler, with significance thresholds set at false discovery rate (FDR) <0.25 and adjusted P<0.05 (31).
Based on previous studies (32), 21 key m6A regulatory genes [FTO, ALKBH5, RBM15, RBM15B, METTL3, METTL14, METTL16, WTAP, VIRMA, zinc finger CCCH-type containing 13 (ZC3H13), HNRNPC, HNRNPA2B1, insulin like growth factor 2 mRNA binding protein (IGF2BP)1, IGF2BP2, IGF2BP3, RBMX, YTHDC1, YTHDC2, YTHDF1, YTHDF2 and YTHDF3] were selected to assess the potential association between m6A methylation regulation and metastatic SKCM. Pearson correlation analysis was performed to assess the relationships between the expression levels of these m6A regulators and key genes, with |R|>0.2 and P<0.05 considered statistically significant.
Potential miRNAs targeting key mRNAs were predicted using the miRWalk database (http://mirwalk.umm.uni-heidelberg.de/). Concurrently, lncRNA-miRNA interactions were identified using the TarBase v8 database (http://carolina.imis.athena-innovation.gr/diana_tools/web/index.php?r=lncbasev2%2Findex). Common miRNAs predicted by both approaches were determined using Venn diagram analysis using the VennDiagram R package. A lncRNA-miRNA-mRNA ceRNA network was subsequently constructed and visualized using Cytoscape (v3.6.1; http://cytoscape.org/release_notes_3_6_1.html). Key regulatory nodes within the network were identified using the ‘cytoHubba’ plugin. To further characterize the lncRNAs, sequences were retrieved from LNCipedia (https://lncipedia.org/), and their subcellular localizations were predicted using lncLocator (http://www.csbio.sjtu.edu.cn/bioinf/lncLocator/). The subcellular localization of mRNAs was determined using the DM3Loc web tool (http://dm3loc.lin-group.cn/). Potential miRNA binding sites within genomic sequences were identified using miRanda (https://www.bioinformatics.com.cn/local_miranda_miRNA_target_prediction_120) (33). Only lncRNAs and mRNAs predicted to localize mainly in the cytoplasm (lncLocator or DM3Loc probability ≥0.6) were retained, ensuring structural plausibility for ceRNA crosstalk.
Relative enrichment scores for 24 immune subsets were calculated from log2(TPM+1) matrices with the GSVA package (v1.34.0; method=‘ssgsea’, kcdf=‘Gaussian’) (34,35). Absolute cell fractions were inferred using CIBERSORTx (https://cibersortx.stanford.edu/index.php) in ‘absolute mode’ with the LM22 signature and 1,000 permutations; samples with P≥0.05 were discarded. Deconvolution was repeated using the EPIC algorithm implemented in TIMER2.0 (http://timer.cistrome.org/TIMER2.0) to provide an additional independent estimate. To evaluate the immune and stromal components of the tumor microenvironment, the Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithm was applied using the ‘estimate’ R package (version 4.3.0; http://www.r-project.org/). Immune scores, stromal scores and tumor purity were calculated for each sample according to the original algorithm. These packages were used to assess the correlation between the expression of m6A-associated genes (KRT17, PKP1, CDH3 and CRABP2) and the infiltration levels of 24 immune cell types. Pearson correlation analysis was applied, with |R|>0.2 and P<0.05 considered statistically significant.
Tissues were fixed in 10% neutral buffered formalin at 4°C overnight, followed by paraffin embedding and sectioning at a 4–5 µm thickness. Sections were sequentially deparaffinized in xylene, dehydrated and rehydrated through a graded ethanol series, and finally immersed in distilled water. Subsequently, 10% goat serum (Gibco; Thermo Fisher Scientific, Inc.) was applied to the sections and incubated at room temperature for 1 h to complete blocking. Sections were then incubated overnight at 4°C with primary antibodies against PKP1 (1:500; cat. no. ab154622; Abcam), KRT17 (1:200; cat. no. ab109725; Abcam), CRABP2 (1:1,000; cat. no. ab211927; Abcam) and CDH3 (1:250; cat. no. ab242060; Abcam). After washing, the slides were incubated with HRP-conjugated secondary antibody (1:2,000; cat. no. ab6759; Abcam) in a humidified chamber for 30 min, followed by color development using a DAB detection kit (Beyotime Institute of Biotechnology). A total of two independent pathologists evaluated the stained sections under an optical microscope (Olympus Corporation). The proportion of positively stained cells was scored as follows: 0, 0–5%; 1, 6–25%; 2, 26–50%; 3, 51–75%; and 4, >75%. Staining intensity was graded as follows: 0, negative; 1, light yellow; 2, yellow-to-brown; and 3, brown. The final immunohistochemical score was calculated as the product of the proportion and intensity scores, and classified into four categories: Negative (−; 0), weakly positive (+; 1–4), moderately positive (++; 5–8) and strongly positive (+++; 9–12).
Total RNA was extracted from SKCM and matched adjacent normal tissues using TRIzol™ reagent (Invitrogen™; Thermo Fisher Scientific, Inc.). cDNA synthesis was performed using the EVO M-MLV RT Mix Kit [cat. no. AG11728; Accurate Biotechnology (Hunan) Co., Ltd.] according to the manufacturer's instructions. qPCR was then performed using the SYBR Green Premix Pro Taq HS qPCR Kit [cat. no. AG11701; Accurate Biotechnology (Hunan) Co., Ltd.] on a LightCycler® 480 Real-Time PCR System (Roche Diagnostics). Each 10 µl reaction mixture contained 1 µl cDNA, 5 µl 2X SYBR Green Pro Taq HS Premix, 0.5 µM of each primer and nuclease-free water. The qPCR cycling conditions were as follows: Initial denaturation at 94°C for 30 sec, followed by 40 cycles at 94°C for 5 sec, 60°C for 10 sec and 50°C for 30 sec. Relative gene expression levels were calculated using the 2−ΔΔCq method (36). The primer sequences used in the present study are listed in Table II.
All statistical analyses, except those involving transcriptomic sequencing data, were performed using GraphPad Prism (version 8.0; Dotmatics) and SPSS software (version 23.0; IBM Corp.). For comparisons of immunohistochemistry scores between tumor and adjacent normal tissues, non-parametric tests were applied: The Mann-Whitney U test for two-group comparisons or the Kruskal-Wallis test for multiple groups, followed by Dunn's post hoc test with Benjamini-Hochberg correction for multiple comparisons. For comparisons among more than two groups with normally distributed data, one-way ANOVA was used, followed by Tukey's post hoc test for multiple pairwise comparisons. RT-qPCR data were analyzed using the Wilcoxon signed-rank test to compare expression levels between tumor and matched normal tissues. Unless otherwise specified, P-values were adjusted for multiple comparisons using the Benjamini-Hochberg false-discovery-rate (FDR) procedure. P<0.05 was considered to indicate a statistically significant difference.
To identify key genes associated with SKCM metastasis, a comprehensive analysis of multiple GEO datasets was performed. Prior to analysis, gene expression data were normalized (Fig. S1), and the ‘limma’ package in R was used to identify DEGs within each microarray dataset, using adjusted P<0.05 and |log2FC|>1 as cutoff criteria (Fig. S2). Subsequently, DEGs from all datasets were integrated using the RRA package, resulting in the identification of 94 consistently differentially expressed mRNAs, 84 upregulated and 10 downregulated, in metastatic SKCM samples compared with in primary SKCM samples across the four datasets (Fig. 1A).
Subsequently, GO biological process enrichment analysis was performed using the ‘clusterProfiler’ package in R. The results indicated that pathways related to cornification, epidermis development, and keratinization were significantly enriched, suggesting their critical involvement in SKCM metastasis. Notably, genes such as cathepsin G (CTSG), serine peptidase inhibitor kazal type 1 and S100 calcium binding protein A8 (S100A8) were identified as key contributors within these pathways (Fig. 1B and C).
In addition, the correlations between the 94 metastasis-related DEGs and 21 m6A regulatory genes were evaluated using Pearson correlation analysis, with thresholds set at |R|>0.5 and P<0.05. DEGs whose expression levels were significantly correlated with ≥1 m6A regulator were defined as m6A-related mRNAs. Based on this criterion, a total of 45 mRNAs were identified that showed significant correlations with m6A (Fig. 1D).
A LASSO-Cox analysis of the upregulated pathways and related genes involved in SKCM metastasis was subsequently performed. The partial likelihood deviance in relation to log(λ) was derived from the LASSO-Cox regression model (Fig. 2A and B). Using the TCGA transcriptomic data and survival information, 12 SKCM metastasis regulators associated with overall survival (OS) were identified: KRT5, KRT17, PKP1, KRT15, filaggrin (FLG), cystatin A (CSTA), CRABP2, cystatin E/M (CST6), calmodulin like (CALML)5, CDH3, S100A8 and CTSG. The risk score and survival status distribution indicated that patient survival time decreased with an elevated risk score. Moreover, combined with the gene expression distribution, higher expression of these genes correlated with a higher risk score (Fig. 2C). Furthermore, survival analysis revealed that patients in the high-risk group exhibited significantly worse long-term survival than those in the low-risk group [hazard ratio (HR), 2.62; P<0.001; Fig. 2D]. Ultimately, the present study focused on four mRNAs strongly related to m6A regulation (CDH3, KRT17, PKP1 and CRABP2). These genes appear to serve crucial roles in SKCM metastasis.
According to previous studies, mRNA expression is modulated by miRNAs, whilst lncRNAs can competitively bind miRNAs and thus indirectly regulate mRNA expression. This lncRNA-miRNA-mRNA triple regulatory network is known as a ceRNA network (37). To clarify the regulation of metastasis-related mRNAs in SKCM by lncRNAs via miRNAs, the present study constructed a ceRNA network centered on CDH3, KRT17, PKP1 and CRABP2. First, single-gene differential expression analysis was performed for each of the four genes in the TCGA-SKCM cohort, identifying significantly co-expressed or differentially up or downregulated transcripts, and for common differentially expressed mRNAs and lncRNAs (Fig. S3). Ultimately, 494 differentially expressed mRNAs and 91 differentially expressed lncRNAs were identified (Fig. 3A).
Subsequently, miRNAs predicted to target the 91 lncRNAs (TarBase) and the 4 mRNAs (miRWalk) were screened, identifying the shared miRNA interactions between lncRNAs and mRNAs. Consequently, 13 lncRNAs, 20 miRNAs and 4 mRNAs were integrated into the ceRNA network (Fig. 3B). Kaplan-Meier survival analysis revealed that KRT17, PKP1, CDH3, CRABP2, SOX21-AS1, hsa-miR-3200-3p, hsa-miR-1976, hsa-miR-1276 and hsa-miR-151a-3p were all significantly associated with OS in SKCM. HR analysis also demonstrated that KRT17, PKP1, CDH3, CRABP2, SOX21-AS1, hsa-miR-1276 and hsa-miR-151a-3p are risk factors for SKCM, whereas hsa-miR-3200-3p and hsa-miR-1976 serve as protective factors (Fig. 3C).
Additionally, co-expressed differentially expressed mRNAs associated with KRT17, PKP1, CDH3, and CRABP2 were subjected to functional enrichment analysis using the Metascape online tool. The results revealed that these genes are closely associated with the estrogen signaling pathway, Ras signaling pathway, cytokine-cytokine receptor interaction, melanogenesis and chemical carcinogenesis, with the CALML3/5 and phospholipase A2 group IVA families serving important roles in these pathways (Fig. 3D).
To further elucidate the abnormal m6A regulatory mechanisms in SKCM, the expression of 21 m6A regulators in SKCM and normal tissues from TCGA were analyzed. A total of four downregulated regulators (METTL3, WTAP, YTHDC1 and YTHDC2) and 13 upregulated regulators (ALKBH5, VIRMA, RBM15, ZC3H13, IGF2BP1, IGF2BP2, IGF2BP3, YTHDF1, YTHDF2, YTHDF3, HNRNPC, HNRNPA2B1 and RBMX) were identified in SKCM (Fig. 4A). The present study also evaluated how these 21 m6A regulators vary across different clinical stages (I–IV), and the ANOVA results revealed that ALKBH5, METTL3, WTAP, RBM15, YTHDF2, HNRNPC, HNRNPA2B1, RBMX, YTHDC1 and IGF2BP3 displayed continuous patterns of increased or decreased mRNA expression with advancing SKCM stage (Fig. 4B). This suggests that certain m6A regulators may be closely associated with SKCM tumorigenesis and progression.
The associations between the four key metastatic prognostic mRNAs (KRT17, PKP1, CDH3 and CRABP2) and the aforementioned 21 m6A regulators were subsequently assessed. The results showed that the expression level of KRT17 was significantly associated with METTL3, RBM15, ALKBH5, IGF2BP3 and HNRNPC. The expression level of PKP1 was significantly associated with METTL16, ALKBH5, IGF2BP3, YTHDF1 and YTHDF2. The expression level of CDH3 was significantly associated with VIRMA, RBM15B, YTHDC2, IGF2BP1 and RBMX, whereas the expression level of CRABP2 was significantly associated with METTL3, RBM15B, FTO, ALKBH5, IGF2BP1 and YTHDF1 (Fig. 5).
To thoroughly assess the influence of KRT17, PKP1, CDH3 and CRABP2 expression on immune cell infiltration, we used single-sample (ss)GSEA to assess correlations between these genes and 24 tumor-infiltrating immune cell types in 472 SKCM samples. The results indicated that PKP1 is positively correlated with mast cells and Th17 cells (Pearson-value >0.2; P<0.05; (Fig. 6A and B); KRT17 is negatively correlated with Tγδ (Tgd) cells, T helper cells (Th cells), and Th1 cells (Pearson-value <-0.2; P<0.05); CDH3 is positively correlated with mast cells and Th17 cells but negatively correlated with B cells and T helper cells; and CRABP2 is positively correlated with mast cells, natural killer (NK) cells, immature dendritic cells and neutrophils, and negatively correlated with B cells (Fig. 6A and B). To corroborate the ssGSEA-derived landscape, the immune infiltration data was re-evaluated using ESTIMATE and CIBERSORTx. The results revealed that tumors with a high expression of PKP1, KRT17, CRABP2 or CDH3 displayed significantly lower StromalScore, ImmuneScore and composite ESTIMATEScore than their low-expression counterparts (all FDR <0.01; Fig. S4), mirroring the global immune-suppressed phenotype observed with ssGSEA. Consistent with the ssGSEA results, CIBERSORTx deconvolution analysis further characterized the tumor immune microenvironment. Specifically, high CRABP2 expression was associated with significantly reduced levels of CD8+ T cells (Figs. 6B and S4). By contrast, both high-PKP1 and high-CRABP2 tumors showed increased levels of NK cells (Fig. 6B). Furthermore, both tumor subtypes shared a common feature of an increased M0-to-M2 macrophage fraction (all FDR <0.05; Fig. S4). The concordant results from the three independent algorithms strengthen the conclusion that elevated expression of these keratin-related genes is associated with an immunologically ‘cold’ microenvironment in melanoma.
ROC curves revealed that KRT17, PKP1, CDH3 and CRABP2 demonstrated predictive power for OS in the TCGA cohort of patients with SKCM (Fig. 7A). The associations of these genes with clinical factors was then further assessed. KRT17, PKP1, CDH3, and CRABP2 demonstrated a positive association with pathological stage (Fig. 7B). For tumor (T)-node (N)-metastasis (M) staging, their expression levels were significantly associated with the T stage, but had no discernable association with N or M stages (Fig. 7C). Similarly, no significant association was revealed between their expression and patient age (Fig. 7D).
Representative immunohistochemistry images demonstrated markedly higher cytoplasmic staining for all four genes in tumor tissues compared with in adjacent skin (Fig. 8). Consistently, RT-qPCR analysis on the same sample set showed 2.3- to 4.5-fold upregulation at the mRNA level (Fig. S5). These concordant protein and transcript results corroborate the bioinformatic prediction of the present study that the four hub genes are overexpressed in metastatic SKCM.
SKCM is a highly malignant tumor derived from melanocytes, known for its poor prognosis and strong invasiveness. Risk factors such as ultraviolet exposure, excessive pigmented nevi, a family history of melanoma and a higher number of nevi lead to DNA damage that can initiate carcinogenesis (38,39). Despite emerging diagnostic and therapeutic approaches, clinical options remain limited as SKCM often progresses rapidly and is frequently asymptomatic in the early stages (1). Hence, elucidating the molecular mechanisms underlying SKCM is paramount for improving treatment outcomes and patient prognosis.
Research has reported that abnormal m6A modification is closely associated with tumorigenesis and progression, as m6A modulates numerous processes (such as mRNA splicing, 3′-end processing, nuclear export, translational regulation, mRNA decay and miRNA biogenesis) and that its dynamic reversibility influences tumor cell growth and differentiation (40). This role has been reported in several cancers, including cervical cancer (41), acute myeloid leukemia (AML) (42), pancreatic cancer (43) and hepatocellular carcinoma (44). However, the specific molecular mechanisms mediated by m6A in SKCM are not yet fully understood.
The present study comprehensively analyzed four metastatic SKCM cohorts from the GEO database (GSE8401, GSE15605, GSE46517 and GSE65904), identifying a total of 823 DEGs, comprising 283 upregulated and 540 downregulated genes. GO enrichment analysis indicated that the DEGs are predominantly involved in biological pathways such as cornification, epidermis development and keratinization, with genes including CTSG, sphingosine-1-phosphate lyase 1 and S100A8 serving pivotal roles. These findings underscore that SKCM metastasis is accompanied by proliferation and keratinization in epidermal/epithelial cells. Similarly, keratinization has been associated with a poor prognosis in human papillomavirus-negative oropharyngeal carcinoma (45).
Subsequently, the present study constructed a survival prediction model in the TCGA database using LASSO-Cox analysis. The results revealed that KRT5, KRT17, PKP1, KRT15, FLG, CSTA, CRABP2, CST6, CALML5, CDH3, S100A8 and CTSG were significantly associated with OS. Elevated expression of these genes was closely associated with poor outcomes. Multiple studies have reported that m6A RNA modification, such as DNA methylation and histone modifications, exerts regulatory control over tumorigenesis via methyltransferases and demethylases (46). However, the precise mechanism by which m6A may affect melanoma progression in an mRNA-dependent manner remains elusive. Therefore, the present study employed Pearson correlation analysis to identify m6A-related metastasis genes in SKCM, and the findings revealed that KRT17, PKP1, CDH3 and CRABP2 exhibit strong associations with m6A regulation and OS in patients with melanoma.
Further single-gene differential expression analysis of KRT17, PKP1, CDH3 and CRABP2 revealed 91 commonly differentially expressed lncRNAs. A lncRNA-miRNA-mRNA triple regulatory network comprising 20 miRNAs and 13 lncRNAs was then constructed. In addition, enrichment analysis demonstrated that 494 co-differentially expressed mRNAs associated with KRT17, PKP1, CDH3 and CRABP2 are primarily enriched in the estrogen signaling pathway, Ras signaling pathway, cytokine-cytokine receptor interaction, melanogenesis and chemical carcinogenesis. Notably, melanin and its synthesis process itself can affect melanoma behavior by regulating oxidative stress, drug sensitivity and immune escape, thus having a profound impact on prognosis (47).
Previous studies indicate that KRTs are crucial in epidermal development, intermediate filament organization, cytoskeletal reorganization, keratinization and keratinocyte migration in patients with melanoma (48–50). In particular, KRT17 has been identified as a diagnostic and prognostic marker in breast cancer (51), epithelial ovarian cancer (52), cervical squamous cell carcinoma (53) and gastric cancer (54). Moreover, PKP1, a key component of desmosomes, has been reported to be abnormally expressed in multiple cancers, including prostate cancer (54), oral squamous cell carcinoma (55) and esophageal adenocarcinoma (56), regulating tumor cell proliferation, colony formation, migration/invasion and apoptosis. In lung cancer, high PKP1 expression has been markedly associated with favorable clinical prognosis, whereas its downregulation is associated with hypermethylation of its promoter region (57). CDH3, which encodes P-cadherin, is a core component of adherens junctions and contributes to the development and progression of several malignancies (58). In patients with adenomyosis, aberrant expression of m6A regulators has been associated with dysregulated CDH3, sodium voltage-gated channel β subunit 4 and placenta associated 8 (59). CRABP2 also serves important roles in multiple tumors. In hepatocellular carcinoma, downregulation of CRABP2 has been reported to inhibit tumor formation in vivo (59). Additionally, Yan et al (60) reported that mutations in ciliary neurotrophic factor receptor, CRABP2, galanin And GMAP prepropeptide, and progestagen associated endometrial protein markedly affected immune infiltration in melanoma.
Furthermore, the investigation of 21 m6A regulators revealed significant differences in the expression of several factors (METTL3, WTAP, YTHDC1, YTHDF2, ALKBH5, VIRMA, RBM15, ZC3H13, IGF2BP1, IGF2BP2, IGF2BP3, YTHDF1, YTHDF2, YTHDF3, HNRNPC, HNRNPA2B1 and RBMX) between normal and tumor samples. Among these, ALKBH5, an important ‘eraser’ in the m6A regulatory system, influences tumor growth and metastasis by demethylating specific transcripts. In breast cancer, ALKBH5-mediated demethylation upregulates NANOG expression, promoting cancer stem cell specification and metastatic capacity (61). ALKBH5 is also highly expressed in lung adenocarcinoma cells, and its knockout can suppress cancer cell proliferation and invasion by increasing m6A modification of forkhead box M1 (62).
Additionally, the m6A methyltransferase WTAP forms a complex with METTL3 and METTL14 and co-localizes in the nucleus to participate in RNA methylation. Research has reported that, under the influence of the hepatocellular carcinoma suppressor ETS1, WTAP regulates the cell cycle in liver cancer via a p21/p27-dependent mechanism (63,64). METTL3 and METTL14 are likewise implicated in oncogenic proliferation and metastasis (65,66). In the cohort in the present study, all three genes were significantly upregulated in metastatic SKCM relative to normal skin, suggesting a potential but as-yet unproven role in melanoma progression. To move beyond bioinformatic association, follow-up experiments, such as chromatin immunoprecipitation-qPCR to confirm direct binding of the complex to target promoters and RNA immunoprecipitation-qPCR to assess m6A modification on specific transcripts, will be necessary to clarify causality in future studies.
However, the ceRNA network presented in the present study is exploratory. Although most miRNA-RNA edges were retrieved from the TarBase and miRWalk records supported by cross-linking immunoprecipitation (CLIP)-sequencing, immunoprecipitation or reporter assays, in-situ validation (such as argonaute-CLIP, biotinylated RNA pull-down or dual-luciferase assays) was not performed in the present study. Furthermore, the localization filter relied on computational predictors rather than fractionation experiments. Future work will need to verify key triplets, particularly the SOX21-AS1/hsa-miR-1276/KRT17 and hsa-miR-151a-3p/PKP1 axes, in melanoma cell lines and patient-derived samples.
Subsequently, the association between KRT17, PKP1, CDH3 and CRABP2 expression and immune infiltration in SKCM was assessed, and the results indicated that these four genes are positively correlated with mast cells and Th17 cells and negatively correlated with B cells and Th1 cells. Immunomodulatory shifts in the tumor microenvironment serve vital roles in tumor metastasis. In a murine melanoma endothelial model, upregulated indoleamine 2,3-dioxygenase 1 was reported to respond to CD40 agonist immunotherapy-induced IFN-γ production, suggesting a novel immunosuppressive feedback mechanism (67). Similarly, Singh et al (23) reported that focal ultrasound heating combined with anti-CD40 agonist antibodies markedly improved T-cell and macrophage function in melanoma. These findings initially suggest that m6A regulators may alter SKCM progression and prognosis by influencing KRT17, PKP1, CDH3 and CRABP2 expression and remodeling the tumor immune microenvironment. Notably, in the present study, KRT17, PKP1, CDH3 and CRABP2 demonstrated significant differential expression across T stages but not in N and M stages, implying that these genes may have a greater role in local tumor growth and tissue infiltration rather than lymph node or distal metastasis. However, further large-scale multicenter studies are needed to validate these findings and explore the underlying molecular mechanisms. Furthermore, there is growing evidence that cancer cells can ‘rewire’ the neuro-endocrine network of the host by secreting hormone-like factors and neurotransmitters to shape a supportive microenvironment (68). This autonomous regulation capability suggests that when targeting m6A-related pathways, its potential interaction with the whole-body steady-state axis should also be considered. If future experiments confirm that genes such as KRT17 or PKP1 critically regulate m6A modifications and the immune microenvironment, they may hold promise for early diagnosis, risk stratification and novel immunotherapeutic approaches in SKCM.
Lastly, RT-qPCR experiments demonstrated higher expression levels of KRT17, PKP1, CDH3 and CRABP2 in SKCM tumor tissues compared with in adjacent normal tissues. Thus, we hypothesize that m6A modification may influence SKCM metastasis by modulating the expression of these four genes. Nevertheless, certain limitations should be noted. First, although the in silico analyses in the present study are generally in-line with previous findings, larger, multicenter clinical cohorts are required to enhance reliability. Moreover, further in vitro and in vivo functional experiments are needed to clarify the mechanisms by which m6A modification and KRT17, PKP1, CDH3 and CRABP2 operate in SKCM. Furthermore, although analyzing the correlation between these four genes and clinicopathological variables has important research value for the prognosis of SKCM patients, disease-free survival (DFS) and tumor-stage-stratified correlations were not incorporated as in the TCGA-SKCM cohort. This is since, in the dataset used for this study, DFS data are missing for 65% of patients and the recorded follow-up periods are highly heterogeneous (median, 6.2 months; interquartile range, 1.8–11.5 months). Preliminary modelling under these conditions yielded unstable HR estimates that risk over-interpretation. Consequently, survival analyses were restricted to OS, for which follow-up is more complete, and immune-infiltration profiling plus experimental validation was focused on. Future studies in prospective cohorts with comprehensive DFS information will be required to extend and confirm the prognostic relevance of the selected markers across multiple clinical endpoints.
In summary, the results of the present study demonstrate that KRT17, PKP1, CDH3 and CRABP2 expression are regulated by m6A modification and may influence SKCM metastasis, ultimately altering patient survival. Moreover, immune microenvironment analyses suggested that mast cells, Th17 cells, B cells and Th1 cells serve critical roles in this process. These findings provide new insights into the mechanisms of SKCM metastasis and potential therapeutic targets. Nonetheless, further research and larger clinical samples are required for more in-depth validation and exploration.
Not applicable.
The present study was supported by the National Natural Science Foundation of China (grant nos. 81772916 and 82103470) and the Natural Science Foundation of Jiangsu Province (grant no. BK20171132).
The data generated in the present study may be requested from the corresponding author.
YG was responsible for study concept design, data organization, formal analysis and drafting the initial manuscript. SZ contributed to data visualization and analysis, manuscript drafting, revision, and final approval. TG was responsible for data validation, study concept design, data review and editing. XLX contributed to study concept design, provided and obtained research resources, and participated in manuscript review and revision. XZ contributed to study concept design, data organization, manuscript writing and final manuscript approval. YG and XZ confirm the authenticity of all the raw data. All authors read and approved the final manuscript.
The present study was approved by the Ethics Committee of the Institute of Dermatology, Peking Union Medical College and Chinese Academy of Medical Sciences (approval no. 2017-KY-044). Written informed consent was obtained from all participants.
Not applicable.
The authors declare that they have no competing interests.
|
Schadendorf D, van Akkooi ACJ, Berking C, Griewank KG, Gutzmer R, Hauschild A, Stang A, Roesch A and Ugurel S: Melanoma. Lancet. 392:971–984. 2018. View Article : Google Scholar | |
|
Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Gershenwald JE, Scolyer RA, Hess KR, Sondak VK, Long GV, Ross MI, Lazar AJ, Faries MB, Kirkwood JM, McArthur GA, et al: Melanoma staging: Evidence-Based changes in the American joint committee on cancer eighth edition cancer staging manual. CA Cancer J Clin. 67:472–492. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Slominski RM, Kim TK, Janjetovic Z, Brożyna AA, Podgorska E, Dixon KM, Mason RS, Tuckey RC, Sharma R, Crossman DK, et al: Malignant melanoma: An overview, new perspectives, and vitamin D signaling. Cancers (Basel). 16:22622024. View Article : Google Scholar : PubMed/NCBI | |
|
Slominski RM, Raman C, Jetten AM and Slominski AT: Neuro-immuno-endocrinology of the skin: How environment regulates body homeostasis. Nat Rev Endocrinol. 21:495–509. 2025. View Article : Google Scholar | |
|
Slominski RM, Chen JY, Raman C and Slominski AT: Photo-neuro-immuno-endocrinology: How the ultraviolet radiation regulates the body, brain, and immune system. Proc Natl Acad Sci USA. 121:e23083741212024. View Article : Google Scholar : PubMed/NCBI | |
|
Tan B, Liu H, Zhang S, da Silva SR, Zhang L, Meng J, Cui X, Yuan H, Sorel O, Zhang SW, et al: Viral and cellular N6-Methyladenosine and N6,2′-O-Dimethyladenosine epitranscriptomes in the KSHV life cycle. Nat Microbiol. 3:108–120. 2018. View Article : Google Scholar | |
|
Pfeifer GP: Defining driver DNA methylation changes in human cancer. Int J Mol Sci. 19:11662018. View Article : Google Scholar | |
|
Wang Y, Li Y, Toth JI, Petroski MD, Zhang Z and Zhao JC: N6-Methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol. 16:191–198. 2014. View Article : Google Scholar | |
|
Alarcón CR, Lee H, Goodarzi H, Halberg N and Tavazoie SF: N6-Methyladenosine marks primary microRNAs for processing. Nature. 519:482–485. 2015. View Article : Google Scholar | |
|
Huang H, Weng H and Chen J: m6A modification in coding and non-coding RNAs: Roles and therapeutic implications in cancer. Cancer Cell. 37:270–288. 2020. View Article : Google Scholar | |
|
Huang H, Weng H and Chen J: The biogenesis and precise control of RNA m6A methylation. Trends Genet. 36:44–52. 2020. View Article : Google Scholar | |
|
Huang H, Weng H, Deng X and Chen J: RNA modifications in cancer: Functions, mechanisms, and therapeutic implications. Annu Rev Cancer Biol. 4:221–240. 2020. View Article : Google Scholar | |
|
Weng H, Huang H, Wu H, Qin X, Zhao BS, Dong L, Shi H, Skibbe J, Shen C, Hu C, et al: METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m6A modification. Cell Stem Cell. 22:191–205. 2018. View Article : Google Scholar | |
|
Zhang C, Chen Y, Sun B, Wang L, Yang Y, Ma D, Lv J, Heng J, Ding Y, Xue Y, et al: m6A modulates haematopoietic stem and progenitor cell specification. Nature. 549:273–276. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Weng H, Su R, Weng X, Zuo Z, Li C, Huang H, Nachtergaele S, Dong L, Hu C, et al: FTO plays an oncogenic role in acute myeloid leukemia as a N6-methyladenosine RNA demethylase. Cancer Cell. 31:127–141. 2017. View Article : Google Scholar | |
|
Zhang S, Zhao BS, Zhou A, Lin K, Zheng S, Lu Z, Chen Y, Sulman EP, Xie K, Bögler O, et al: m6A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by Sustaining FOXM1 expression and cell proliferation program. Cancer Cell. 31:591–606.e6. 2017. View Article : Google Scholar | |
|
Zhang C, Samanta D, Lu H, Bullen JW, Zhang H, Chen I, He X and Semenza GL: Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc Natl Acad Sci USA. 113:E2047–E2056. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
He F, Yu J, Yang J, Wang S, Zhuang A, Shi H, Gu X, Xu X, Chai P and Jia R: m6A RNA hypermethylation-induced BACE2 boosts intracellular calcium release and accelerates tumorigenesis of ocular melanoma. Mol Ther. 29:2121–2133. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dahal U, Le K and Gupta M: RNA m6A methyltransferase METTL3 regulates invasiveness of melanoma cells by matrix metallopeptidase 2. Melanoma Res. 29:382–389. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xu L, Shen SS, Hoshida Y, Subramanian A, Ross K, Brunet JP, Wagner SN, Ramaswamy S, Mesirov JP and Hynes RO: Gene expression changes in an animal melanoma model correlate with aggressiveness of human melanoma metastases. Mol Cancer Res. 6:760–769. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Cabrita R, Lauss M, Sanna A, Donia M, Larsen MS, Mitra S, Johansson I, Phung B, Harbst K, Vallon-Christersson J, et al: Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 577:561–565. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Singh MP, Sethuraman SN, Ritchey J, Fiering S, Guha C, Malayer J and Ranjan A: In-situ vaccination using focused ultrasound heating and anti-CD-40 agonistic antibody enhances T-cell mediated local and abscopal effects in murine melanoma. Int J Hyperthermia. 36:64–73. 2019. View Article : Google Scholar | |
|
Raskin L, Fullen DR, Giordano TJ, Thomas DG, Frohm ML, Cha KB, Ahn J, Mukherjee B, Johnson TM and Gruber SB: Transcriptome profiling identifies HMGA2 as a biomarker of melanoma progression and prognosis. J Invest Dermatol. 133:2585–2592. 2013. View Article : Google Scholar | |
|
Xu F, Lin H, He P, He L, Chen J, Lin L and Chen Y: A TP53-associated gene signature for prediction of prognosis and therapeutic responses in lung squamous cell carcinoma. Oncoimmunology. 9:17319432020. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Han D, Hou Y, Chen H and Chen Z: Statistical inference methods for two crossing survival curves: A comparison of methods. PLoS One. 10:e01167742015. View Article : Google Scholar : PubMed/NCBI | |
|
Love MI, Huber W and Anders S: Moderated estimation of fold change and dispersion for RNA-Seq data with DESeq2. Genome Biol. 15:5502014. View Article : Google Scholar | |
|
Kolde R, Laur S, Adler P and Vilo J: Robust rank aggregation for gene list integration and meta-analysis. Bioinformatics. 28:573–580. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi A.H, Tanaseichuk O, Benner C and Chanda SK: Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 10:15232019. View Article : Google Scholar : PubMed/NCBI | |
|
Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Zaccara S, Ries RJ and Jaffrey SR: Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 20:608–624. 2019. View Article : Google Scholar | |
|
Enright AJ, John B, Gaul U, Tuschl T, Sander C and Marks DS: MicroRNA targets in drosophila. Genome Biol. 5:R12003. View Article : Google Scholar | |
|
Hänzelmann S, Castelo R and Guinney J.GSVA: Gene set variation analysis for microarray and RNA-Seq data. BMC Bioinformatics. 14:72013. | |
|
Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, Angell H, Fredriksen T, Lafontaine L, Berger A, et al: Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 39:782–795. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The rosetta stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar | |
|
Markovic SN, Erickson LA, Rao RD, Weenig RH, Pockaj BA, Bardia A, Vachon CM, Schild SE, McWilliams RR, Hand JL, et al: Malignant melanoma in the 21st century, part 1: Epidemiology, risk factors, screening, prevention, and diagnosis. Mayo Clin Proc. 82:364–380. 2007. View Article : Google Scholar | |
|
Osipov M and Sokolnikov M: Previous malignancy as a risk factor for the second solid cancer in a cohort of nuclear workers. SciMedicine J. 3:8–15. 2021. View Article : Google Scholar | |
|
Jiang X, Liu B, Nie Z, Duan L, Xiong Q, Jin Z, Yang C and Chen Y: The role of m6A modification in the biological functions and diseases. Signal Transduct Target Ther. 6:742021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Li Z, Kong B, Song C, Cong J, Hou J and Wang S: Reduced m6A mRNA methylation is correlated with the progression of human cervical cancer. Oncotarget. 8:98918–98930. 2017. View Article : Google Scholar | |
|
Kwok CT, Marshall AD, Rasko JEJ and Wong JJL: Genetic alterations of m6A regulators predict poorer survival in acute myeloid leukemia. J Hematol Oncol. 10:392017. View Article : Google Scholar | |
|
Cho SH, Ha M, Cho YH, Ryu JH, Yang K, Lee KH, Han ME, Oh SO and Kim YH: ALKBH5 gene is a novel biomarker that predicts the prognosis of pancreatic cancer: A retrospective multicohort study. Ann Hepatobiliary Pancreat Surg. 22:305–309. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao X, Chen Y, Mao Q, Jiang X, Jiang W, Chen J, Xu W, Zhong L and Sun X: Overexpression of YTHDF1 is associated with poor prognosis in patients with hepatocellular carcinoma. Cancer Biomarkers. 21:859–868. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Slebos RJC, Jehmlich N, Brown B, Yin Z, Chung CH, Yarbrough WG and Liebler DC: Proteomic analysis of oropharyngeal carcinomas reveals novel HPV-associated biological pathways. Int J Cancer. 132:568–579. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Deng X, Su R, Feng X, Wei M and Chen J: Role of N6-methyladenosine modification in cancer. Curr Opin Genet Dev. 48:1–7. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Slominski RM, Sarna T, Płonka PM, Raman C, Brożyna AA and Slominski AT: Melanoma, melanin, and melanogenesis: The yin and yang relationship. Front Oncol. 12:8424962022. View Article : Google Scholar | |
|
Han W, Hu C, Fan ZJ and Shen GL: Transcript levels of keratin 1/5/6/14/15/16/17 as potential prognostic indicators in melanoma patients. Sci Rep. 11:10232021. View Article : Google Scholar : PubMed/NCBI | |
|
Han Y, Li X, Yan J, Ma C, Wang X, Pan H, Zheng X, Zhang Z, Gao B and Ji XY: Bioinformatic analysis identifies potential key genes in the pathogenesis of melanoma. Front Oncol. 10:5819852020. View Article : Google Scholar | |
|
Kodet O, Lacina L, Krejčí E, Dvořánková B, Grim M, Štork J, Kodetová D, Vlček Č, Šáchová J, Kolář M, et al: Melanoma cells influence the differentiation pattern of human epidermal keratinocytes. Mol Cancer. 14:12015. View Article : Google Scholar : PubMed/NCBI | |
|
van de Rijn M, Perou CM, Tibshirani R, Haas P, Kallioniemi O, Kononen J, Torhorst J, Sauter G, Zuber M, Köchli OR, et al: Expression of cytokeratins 17 and 5 identifies a group of breast carcinomas with poor clinical outcome. Am J Pathol. 161:1991–1996. 2002. View Article : Google Scholar | |
|
Wang YF, Lang HY, Yuan J, Wang J, Wang R, Zhang XH, Zhang J, Zhao T, Li YR, Liu JY, et al: Overexpression of keratin 17 is associated with poor prognosis in epithelial ovarian cancer. Tumour Biol. 34:1685–1689. 2013. View Article : Google Scholar | |
|
Escobar-Hoyos LF, Yang J, Zhu J, Cavallo JA, Zhai H, Burke S, Koller A, Chen EI and Shroyer KR: Keratin 17 in premalignant and malignant squamous lesions of the cervix: Proteomic discovery and immunohistochemical validation as a diagnostic and prognostic biomarker. Mod Pathol. 27:621–630. 2014. View Article : Google Scholar | |
|
Chivu-Economescu M, Dragu DL, Necula LG, Matei L, Enciu AM, Bleotu C and Diaconu CC: Knockdown of KRT17 by siRNA induces antitumoral effects on gastric cancer cells. Gastric Cancer. 20:948–959. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Harris TM, Du P, Kawachi N, Belbin TJ, Wang Y, Schlecht NF, Ow TJ, Keller CE, Childs GJ, Smith RV, et al: Proteomic analysis of oral cavity squamous cell carcinoma specimens identifies patient outcome-associated proteins. Arch Pathol Lab Med. 139:494–507. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Kaz AM, Luo Y, Dzieciatkowski S, Chak A, Willis JE, Upton MP, Leidner RS and Grady WM: Aberrantly methylated PKP1 in the progression of Barrett's esophagus to esophageal adenocarcinoma. Genes Chromosomes Cancer. 51:384–393. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Haase D, Cui T, Yang L, Ma Y, Liu H, Theis B, Petersen I and Chen Y: Plakophilin 1 is methylated and has a tumor suppressive activity in human lung cancer. Exp Mol Pathol. 108:73–79. 2019. View Article : Google Scholar | |
|
Xu Y, Zhao J, Dai X, Xie Y and Dong M: High expression of CDH3 predicts a good prognosis for colon adenocarcinoma patients. Exp Ther Med. 18:841–847. 2019.PubMed/NCBI | |
|
Zhai J, Li S, Sen S, Opoku-Anane J, Du Y, Chen ZJ and Giudice LC: m6A RNA methylation regulators contribute to eutopic endometrium and myometrium dysfunction in adenomyosis. Front Genet. 11:71620202020. View Article : Google Scholar | |
|
Yan J, Wu X, Yu J, Zhu Y and Cang S: Prognostic role of tumor mutation burden combined with immune infiltrates in skin cutaneous melanoma based on multi-omics analysis. Front Oncol. 10:5706542020. View Article : Google Scholar | |
|
Zhang C, Zhi WI, Lu H, Samanta D, Chen I, Gabrielson E and Semenza GL: Hypoxia-Inducible factors regulate pluripotency factor expression by ZNF217- and ALKBH5-mediated modulation of RNA methylation in breast cancer cells. Oncotarget. 7:64527–64542. 2016. View Article : Google Scholar | |
|
Chao Y, Shang J and Ji W: ALKBH5-m6A-FOXM1 signaling axis promotes proliferation and invasion of lung adenocarcinoma cells under intermittent hypoxia. Biochem Biophys Res Commun. 521:499–506. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Schöller E, Weichmann F, Treiber T, Ringle S, Treiber N, Flatley A, Feederle R, Bruckmann A and Meister G: Interactions, localization, and phosphorylation of the m6A generating METTL3-METTL14-WTAP complex. RNA. 24:499–512. 2018. View Article : Google Scholar | |
|
Chen Y, Peng C, Chen J, Chen D, Yang B, He B, Hu W, Zhang Y, Liu H, Dai L, et al: WTAP facilitates progression of hepatocellular carcinoma via m6A-HuR-dependent epigenetic silencing of ETS1. Mol Cancer. 18:1272019. View Article : Google Scholar : PubMed/NCBI | |
|
Ma JZ, Yang F, Zhou CC, Liu F, Yuan JH, Wang F, Wang TT, Xu QG, Zhou WP and Sun SH: METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6-methyladenosine-dependent primary MicroRNA processing. Hepatology. 65:529–543. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Chen M, Wei L, Law CT, Tsang FHC, Shen J, Cheng CLH, Tsang LH, Ho DWH, Chiu DKC, Lee JMF, et al: RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 67:2254–2270. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Georganaki M, Ramachandran M, Tuit S, Núñez NG, Karampatzakis A, Fotaki G, van Hooren L, Huang H, Lugano R, Ulas T, et al: Tumor endothelial cell up-regulation of IDO1 is an immunosuppressive feed-back mechanism that reduces the response to CD40-stimulating immunotherapy. Oncoimmunology. 9:17305382020. View Article : Google Scholar : PubMed/NCBI | |
|
Slominski RM, Raman C, Chen JY and Slominski AT: How cancer hijacks the body's homeostasis through the neuroendocrine system. Trends Neurosci. 46:263–275. 2023. View Article : Google Scholar |