Home Deciphering the role of ELAVL1: Insights from pan-cancer multiomics analyses with emphasis on nasopharyngeal carcinoma
Article Open Access

Deciphering the role of ELAVL1: Insights from pan-cancer multiomics analyses with emphasis on nasopharyngeal carcinoma

  • Jindong Xie , Yi Xie , Wencheng Tan , Yimeng Ye , Xueqi Ou , Xiong Zou , Zhiqing He , Jiarong Wu , Xinpei Deng , Hailin Tang , Longjun He , Kailai Li , Peng Luo , Kunhao Bai , Guoxian Huang ORCID logo EMAIL logo and Jianjun Li ORCID logo EMAIL logo
Published/Copyright: May 8, 2025

Abstract

Background and Objectives

Cancer continues to be a predominant cause of mortality worldwide, underscoring the critical need to identify and develop novel biomarkers to improve prognostic accuracy and therapeutic approaches. The dysregulation of ELAVL1 is linked to various diseases, including cancer. Nevertheless, its role across different cancer types remains insufficiently investigated.

Methods

We conducted a systematic investigation into the expression patterns, prognostic significance, genomic alterations, modifications, and functional implications of ELAVL1 in pan-cancer types. Besides, we performed in vitro and in vivo experiments to confirm the role of ELAVL1 in nasopharyngeal carcinoma (NPC).

Results

By utilizing multi-omics datasets, we found obvious overexpression of ELAVL1 in various cancer types at both the mRNA and protein levels, with predominant expression in malignant cells. Survival analysis revealed that increased ELAVL1 expression was linked to unfavorable outcomes in certain cancers; however, its effect difers among various cancer types. Additionally, we found that the genomic alterations and modifications of ELAVL1 were related to tumor progression. We discovered that ELAVL1 was elevated in NPC tissues. In addition, survival analysis indicated that NPC patients with higher ELAVL1 expression had worse prognoses. Functional assays demonstrated that ELAVL1 suppression led to decreased proliferation and migration in NPC cell lines. Moreover, ELAVL1 knockdown effectively inhibited NPC progression in the lymph node and lung metastasis models.

Conclusions

In summary, ELAVL1 exhibits diverse and complex involvement in tumor progression. Targeting it might inhibit tumor progression, making it a promising biomarker and therapeutic target for enhancing cancer treatment outcomes.

Introduction

Cancer is the primary cause of premature mortality in most countries and is the principal challenge to be addressed in efforts to enhance human life expectancy.[1] According to the Global Cancer Statistics 2022, it accounts for 9.7 million deaths and 20 million newly diagnosed cases worldwide.[2] Its global burden is projected to increase. Cancer incidence and mortality rates are rising because of growing and aging populations, changes in the prevalence and distribution of risk factors associated with the disease, and socioeconomic development.[3] Thus, it is essential to identify novel biomarkers and molecular targets to predict patient prognosis and customize treatment strategies.

Originating from the nasopharyngeal epithelium, nasopharyngeal carcinoma (NPC) is marked by a strong propensity for metastasis and a distinct geographic distribution. Over 75% of the new cases in 2020 were reported in Eastern and Southeastern Asia, with a particularly high incidence in China.[4, 5, 6] The development of NPC has been strongly associated with Epstein–Barr virus infection, environmental influences, and genetic predispositions.[7] Radiotherapy is the primary modality of treatment for it.[8] However, owing to its highly invasive characteristics, approximately 10% to 20% of patients with NPC are likely to face local recurrence, and 7% to 20% might develop distant metastases within two years.[9, 10] Thus, understanding the molecular mechanisms driving NPC progression and finding accurate therapeutic targets for an intervention for it are essential to reduce recurrence and metastatic spread.

ELAVL1 encodes human antigen R (HuR), a well-characterized RNA-binding protein that plays a pivotal role in post-transcriptional regulation.[11] It is a member of the mammalian embryonic lethal abnormal vision-like (ELAVL) protein family, which has three other members: HuB, HuC, and HuD.[12] The potential role of ELAVL1 in carcinogenesis was initially elucidated through a murine study that demonstrated that the abnormal expression levels of ELAVL1 affected the tumorigenic capacity of a colon cancer cell line.[13] Further studies found that ELAVL1 was predominantly localized within the nucleus but could be shuttled to the cytoplasmic compartment in response to various stimuli (e.g., stress signals, ischemia, hypoxia, ultraviolet irradiation). [14, 15, 16] This translocation is a critical component of ELAVL1 function in stabilizing and enhancing the translation of mature target mRNAs. There is growing evidence that ELAVL1 is involved in the progression of multiple cancer types.[17, 18, 19, 20, 21, 22] In addition, recent findings indicate that ELAVL1 participates in metabolic processes and may be associated with immune-related disorders. [23, 24, 25, 26] These observations indicate that ELAVL1 may act in a complex manner to influence both tumor dynamics and immune system interactions.

Nevertheless, there is still a notable deficiency in the comprehensive investigation of ELAVL1’s function across various forms of cancer, and a significant question that emerges is whether ELAVL1 expression can accurately characterize tumor heterogeneity within a specific cancer type and function as a therapeutic target. In addition, no studies have been conducted to investigate ELAVL1 expression or to assess its impact on NPC prognosis. To bridge these knowledge gaps, it is essential to undertake a comprehensive analysis involving large patient cohorts.

An overview of the present study is provided in Figure 1. We conducted a pan-cancer systematic investigation into the expression patterns, prognostic significance, genomic alterations, modifications, and functional implications of ELAVL1 at the multi-omics level. Through in vitro and in vivo validations, we found that ELAVL1 was elevated in NPC tissues and that ELAVL1 knockdown effectively inhibited NPC progression. These findings are expected to elucidate the role of ELAVL1 as a promising biomarker and therapeutic target for enhancing treatment outcomes.

Figure 1 Study flowchart (Created with BioRender.com).
Figure 1

Study flowchart (Created with BioRender.com).

Materials and methods

Pan-cancer data collection and processing

The UCSC Xena platform was utilized to access the TCGA and GTEx databases, focusing on pan-cancer ELAVL1 expression levels and associated clinical characteristics. Abbreviations of the 33 cancer types encompassed in this study were referred to Supplementary TableS1. We obtained the pan-cancer scRNA-seq datasets from the TISCH database.[27] Pan-cancer proteomics and ST datasets were summarized and obtained from Sparkle database (https://grswsci.top/) and the corresponding ID for each ST sample were summarized in Supplementary Table S2. NPC datasets were downloaded from the GEO database. [28, 29, 30, 31, 32] The probes were mapped utilizing the “AnnoProbe” R package, and averaging multiple probes were calculated using the “limma” R package when necessary.[33]

Differential expression and localization analyses

The HPA database was utilized to assess the ELAVL1 expression level in cell lines and tissues. Data from the TCGA and GTEx databases were obtained to compare differences between tumors and normal tissues at mRNA levels, and pan-cancer proteomics datasets were used to compare differences between tumors and normal tissues at protein levels. The 3D configuration of ELAVL1 was obtained from the PDB database (https://www.rcsb.org/) and the location of ELAVL1 was acquired from the UniProt database (https://www.uniprot.org/).

Diagnostic and prognostic analyses

The ROC curves for the cancers of interest were created using the “pROC” R package. Survival curves were conducted using “survival” and “survminer” R packages. Univariate Cox regression analyses were executed using the “survival” and “forestplot” R packages to assess the prognostic significance of ELAVL1 expression as a predictor of OS, DSS, PFI, and DFI.

Genomic alteration analyses

Pan-cancer analyses of genomic mutation frequencies, amplifications, and deep deletions were performed utilizing the Cancer Type Summary module available in the cBioPortal database.[34] The processed SNV data were analyzed using the “maftools” R package, in order to elucidate the mutational landscape of ELAVL1 across pan-cancer datasets.[35] The processed CNV data were obtained from GSCA database and visualized using “ggplot2” R package.[36]

Modification analyses

PTMs was obtained from the PhosphoSite database.[37] Differential expression analyses of phosphorylation and methylation modifications were downloaded from the UALCAN database.[38]

Alternative splicing analyses

The investigation of clinically significant AS of ELAVL1 was conducted utilizing the OncoSplicing server and its ClinicalAS tool.[39] We identify pertinent ELAVL1 AS events within the SplAdder function.

Functional enrichment and immunological interaction analyses

Based on ELAVL1 mRNA expression levels, the top quarter of samples were defined as the high expression group, and the bottom quarter were considered as the low expression group in each tumor type. We utilized the “limma” R package to calculate the log2 fold change (log2FC) for each gene, and subsequently, all genes were ranked based on their log2FC values.[33] GSEA was executed using the “clusterProfiler” R package and the hallmark gene set.[40] Genome-wide screening CRISPR were downloaded from DepMap database,[41] and candidate genes were calculated using CERES algorithm. A negative score means cell growth inhibition and/or death after gene knockout.

For immunological interaction analyses, we applied “immunedeconv” R package to calculate the infiltration levels from the TCGA pan-cancer datasets,[42] and immune-related genes were collected from a previous study.[43] The verification of genome status and immune subtypes were originated from another previous study.[44] Pan-cancer immunotherapy cohorts were obtained from the BEST database.[45]

Cell lines and cultures

Human NPC cell lines (CNE1, CNE2, HNE1, C666-1, SUNE-1, and HONE-1) were maintained in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS). N2-Tert was cultured in keratinocyte serum-free medium (Invitrogen), with supplementation of bovine pituitary extract. The cell lines mentioned above were authenticated and kindly provided by Prof. Wei Xiong (Central South University, China). Radiotherapy-sensitive and resistant HK1 cell lines were kindly provided by Prof. Jian Zhang (Southern Medical University, China).[46, 47]

Western blotting

Cell lysates were prepared using RIPA lysis buffer to extract proteins, which were then separated by SDS-PAGE (Beyotime) and transferred onto PVDF membranes (Millipore). Following 1 hour of blocking in nonfat milk, membranes were incubated with a primary antibody overnight at 4°C, followed by incubation with secondary antibodies at room temperature for 1 hour. The following primary antibodies were used: anit-ELAVL1 (Rabbit, 11910–1-AP, Proteintech), and anti-α-Tubulin (Rabbit, 66031-1-Ig, Proteintech).

Transient transfection and stable cell line construction

For siRNA-mediated RNA interference, siRNAs specifically targeting ELAVL1, as well as non-targeting control, were synthesized and procured from GenePharma (Suzhou, China). The siRNAs were transfected into the cells using Lipofectamine 3000 (Invitrogen), following the protocol provided by the manufacturer. For stable cell line construction, NPC cells were transfected with the shRNA‐ELAVL1 or control viruses. Targeted sequences for siRNA and shRNA are provided in Supplementary TableS3 and Supplementary TableS4.

CCK-8 assays

A total of 2, 000 cells were incubated in 96‐well plates. Subsequently, the CCK-8 solution was introduced into each well and incubated for a duration of 2 h for evaluating cell viability. OD450 values were measured over 5 days.

Transwell assays

A total of 4 × 104 HONE-1 cells and 6 × 104 SUNE-1 cells underwent enzymatic digestion and were subsequently resuspended. Cells were introduced into the upper chambers devoid of FBS, whereas the lower cross-pore compartment contained a medium supplemented with 20% FBS. After a 20-hour incubation, the migrated NPC cells were fixed with methanol and stained with 0.1% crystal violet.

Wound healing assays

The transfected NPC cells were cultured in 6-well plates, and wounds were created using a 100 μL pipette tip. Microscopic images were taken at 0 and 24 hours. ImageJ software was utilized to quantify the scratch area.

Colony formation assays

A total of 1, 000 NPC cells were seeded into each well of 6-well plates using a medium supplemented with 10% FBS. After 2 weeks, the colonies were fixed using methanol for 20 minutes and stained with 0.1% crystal violet for 20 minutes.

Animal experiments

Animal experiments were performed in accordance with the guidelines of the Institutional Animal Care and Use Ethics Committee of SYSUCC (L102012024120B). Female BALB/c nude mice aged 4 weeks were purchased from Zhuhai BesTest Bio‐Tech Co. Ltd. and housed at the Animal Facility of SYSUCC under controlled conditions.

For the popliteal lymph node metastatic model, ELAVL1-KD or control HONE-1 cells (1 × 106 cells) in PBS were subcutaneously injected into the footpads of mice. After one month, the mice were humanely euthanized. The primary footpad along with lymph node metastatic tumors were surgically excised.

For the lung metastasis model, ELAVL1-KD or control HONE-1 cells (1 × 106 cells) were administered via tail vein injection. After one month, the mice were humanely euthanized, and their lungs were surgically excised.

For bioluminescence imaging, 100 μL of D-luciferin (Yeason, Shanghai, China) was injected into each mouse and images were subsequently captured. The results were then analyzed using the Living Image software (Xenogen Corp.).

IHC staining

Three NPC and three adjacent normal paraffin-embedded tissues were collected from Guangdong Provincial People’s Hospital. All patients were pathologically confirmed. This study was approved by the Ethics Committee of Guangdong Provincial People’s Hospital. Tissue sections underwent deparaffinization using xylene followed by rehydration with a series of ethanol concentrations (100%, 95%, 85%, and 75%). Antigen retrieval was conducted before overnight incubation with the primary antibody at 4°C to inhibit endogenous peroxidase activity. Subsequently, the sections were incubated for 20 minutes with an HRP-conjugated secondary antibody at room temperature, and staining was carried out using diaminobenzidine (DAB) substrate (Dako). Following DAB treatment, the sections were counterstained with hematoxylin.

Statistical analysis

Statistical analyses within this research were executed using R and GraphPad Prism software. For comparisons between two groups, Student’s t-test and Wilcoxon test was used. When comparing more than two groups, one-way ANOVA and Kruskal-Wallis test was applied. Cox regression, log-rank tests were used to assess the prognostic significance of ELAVL1. Correlations between variables were assessed using Spearman’s correlation for non-parametric data and Pearson’s correlation for parametric data. The Benjamini–Hochberg method was applied to adjust the false discovery rate (FDR), with an adjusted P-value < 0.05 considered significant.

Results

Expression patterns of ELAVL1 in pan-cancer and its localization

The human protein atlas (HPA) and FANTOM5 datasets revealed that ELAVL1 expression was most prominent within the thymus, testis, and lymph node tissues (Figure 2A and Supplementary Figure S1A). We also gathered data regarding ELAVL1 expression across various pan-cancer cell lines and found that it is highly expressed in adrenocortical cancer, leukemia, and lymphoma (Supplementary Figure S1B). Subsequently, we conducted a comparative analysis of ELAVL1 mRNA expression levels between tumor and normal tissues using data compiled from the TCGA and GTEx datasets, and we observed significantly increased expression of ELAVL1 in most cancer types, except for LAML, PCPG, and TGCT (Figure 2B and Supplementary Figure S1C). Similarly, ELAVL1 exhibited elevated mRNA expression levels in tumors compared to paired normal tissues, based on the TCGA pan-cancer dataset (Figure 2C and Supplementary Figure S1D). To verify its abnormal expression at the protein level, we collected pan-cancer proteomics datasets, and we noted that in most tumor types, there was a general upregulation of ELAVL1 protein levels (Figure 2D).

Figure 2 Expression patterns of ELAVL1 in pan-cancer and its localization. (A) The expression levels of ELAVL1 based on HPA database. (B) Violin plots showing the different mRNA expression levels of ELAVL1 in TCGA-GTEx pan-cancer datasets. (C) Paired differential analysis showing the mRNA expression levels between tumor and normal tissues in TCGA pan-cancer dataset. (D) Boxplots showing the different protein expression levels of ELAVL1 in proteomics pan-cancer datasets. (E) 3D structure of ELAVL1 obtained from PDB database. (F) Subcellular locations of ELAVL1 from the UniProt database. (G) Bar plots showing the AUC values evaluating the diagnostic efficacy of ELAVL1 expression in tumor and normal tissues. (H) Heatmap showing the mRNA expression level of ELAVL1 in pan-cancer datasets at the spatial transcriptome level. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. HPA, human protein atlas; 3D, three-dimensional; PDB, Protein Data Bank.
Figure 2

Expression patterns of ELAVL1 in pan-cancer and its localization. (A) The expression levels of ELAVL1 based on HPA database. (B) Violin plots showing the different mRNA expression levels of ELAVL1 in TCGA-GTEx pan-cancer datasets. (C) Paired differential analysis showing the mRNA expression levels between tumor and normal tissues in TCGA pan-cancer dataset. (D) Boxplots showing the different protein expression levels of ELAVL1 in proteomics pan-cancer datasets. (E) 3D structure of ELAVL1 obtained from PDB database. (F) Subcellular locations of ELAVL1 from the UniProt database. (G) Bar plots showing the AUC values evaluating the diagnostic efficacy of ELAVL1 expression in tumor and normal tissues. (H) Heatmap showing the mRNA expression level of ELAVL1 in pan-cancer datasets at the spatial transcriptome level. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. HPA, human protein atlas; 3D, three-dimensional; PDB, Protein Data Bank.

The three-dimensional (3D) configuration of ELAVL1 was retrieved from the Protein Data Bank (PDB) database (Figure 2E). We then explored the cellular location of ELAVL1. Using the UniProt database, we found that ELAVL1 could be detected in the nucleoplasm, nucleoli, and cytosol (Figure 2F). The emergence of multi-omics technologies enables a more accurate exploration of the expression patterns of genes.[48] To identify the particular cell types expressing ELAVL1, we examined its expression levels using the TISCH database-which contains pan-caner single-cell RNA sequencing (scRNA-seq) datasets-and the results showed that ELAVL1 was mainly expressed in malignant cells, proliferating T cells, and fibroblasts (Supplementary Figure S1E). To verify our finding, we collected pan-cancer spatial transcriptomics (ST) datasets. As expected, ELAVL1 was predominantly localized within the regions occupied by tumor cells (Figure 2H). In summary, our findings indicate that ELAVL1 is overexpressed in various cancers, which may underscore its potential as a clinical diagnostic biomarker.

ELAVL1’s provision of both diagnostic and prognostic value in different cancers

Receiver operating characteristic (ROC) curve analyses confirmed that ELAVL1 demonstrated high accuracy in distinguishing tumor and normal tissues for most cancer types such as BLCA, BRCA, and CESC (Figure 2G). Moreover, we performed univariate logistic regression, and the results showed that the odds ratio (OR) values were over 1 in most cancer types, suggesting a positive correlation between elevated ELAVL1 expression and tumor status and a possible association between increased ELAVL1 expression and an elevated risk of tumor development (Figure 3A).

To evaluate the prognostic utility of ELAVL1 across pan-cancer, we conducted Cox regression and log-rank tests for survival analyses encompassing overall survival (OS), disease-specific survival (DSS), progression-free interval (PFI), and disease-free interval (DFI) (Figure 3B). OS analysis showed that high ELAVL1 expression was associated with shorter OS in KIRC, KIRP, LGG, MESO, and SKCM, whereas it was a protective factor in CESC, KICH, LAML, OV, THCA, and THYM (Figure 3C). ELAVL1 was also a risk factor significantly associated with worse DFS in KIRC, KIRP, LGG, LUAD, MESO, and SKCM, while it was a protective factor in HNSC, KICH, and OV (Figure 3D). For PFI, ELAVL1 was a risk factor for KIRC, KIRP, and LGG, while it was protective in GBM and KICH (Figure 3E). With respect to DFI, ELAVL1 was a risk factor in BRCA and PRAD (Figure 3F). Overall, these findings indicate that ELAVL1 provides both diagnostic and prognostic value in different cancer types.

Analyses of ELAVL1 genomic alterations

Genomic approaches are a powerful means of cancer analysis.[49, 50] Single nucleotide variation (SNV) mainly refers to the change in a DNA sequence caused by the alteration of a single nucleotide at the genomic level, which plays a significant role in tumor progression.[51,52] Abnormal copy number variation (CNV) is also widely recognized as a fundamental molecular mechanism in tumor progression.[53, 54] ELAVL1 amplification was chiefly detected in SARC, whereas deep deletions were frequently identified in UCEC, DLBC, SKCM, and SNVs were notably frequent in UCEC, DLBC, and STAD (Figure 4A–4D). We then explored CNV patterns in pan-cancer (including homozygous amplification, heterozygous amplification, homozygous deletion, and heterozygous deletion) (Figure 4D). Our analysis revealed that the CNV profiles of ELAVL1 predominantly exhibited heterozygosity (Figure 4E). Moreover, we analyzed the correlation of CNV with mRNA expression and found that most cancer types showed a positive correlation (Figure 4F).

Figure 3 ELAVL1 offers diagnostic and prognostic utility in different cancers. (A) Univariate logistic regression analysis of ELAVL1 mRNA expression and tumor status. (B) Heatmap showing relationship between ELAVL1 mRNA expression and different survival outcomes in pan-cancer. (C–F) Forest plots were used for pan-cancer analyses of ELAVL1 and OS (C), DFS (D), PFI (E), and DFI (F). OS, overall survival; DSS, disease-specific survival; PFI, progression-free interval; DFI, disease-free interval.
Figure 3

ELAVL1 offers diagnostic and prognostic utility in different cancers. (A) Univariate logistic regression analysis of ELAVL1 mRNA expression and tumor status. (B) Heatmap showing relationship between ELAVL1 mRNA expression and different survival outcomes in pan-cancer. (C–F) Forest plots were used for pan-cancer analyses of ELAVL1 and OS (C), DFS (D), PFI (E), and DFI (F). OS, overall survival; DSS, disease-specific survival; PFI, progression-free interval; DFI, disease-free interval.

Figure 4 Analyses of ELAVL1 genomic alterations. (A) Stacked bar plots showing mutation frequencies of ELAVL1 in pan-cancer. (B) Heatmap showing mutation of ELAVL1 and several classical carcinogenic signaling pathways in pan-cancer. (C) Oncoplot of the mutation distribution of ELAVL1 in pan-cancer. (D) Percentage pie charts showing CNV profile of ELAVL1 in each tumor type. (E) The heterozygous and homozygous CNV profile of ELAVL1 in each tumor type, including the percentage of amplification and deletion. (F) Bubble plots showing the correlation of CNV with ELAVL1 mRNA expression. FDR, false discovery rate. CNV, copy number variation.
Figure 4

Analyses of ELAVL1 genomic alterations. (A) Stacked bar plots showing mutation frequencies of ELAVL1 in pan-cancer. (B) Heatmap showing mutation of ELAVL1 and several classical carcinogenic signaling pathways in pan-cancer. (C) Oncoplot of the mutation distribution of ELAVL1 in pan-cancer. (D) Percentage pie charts showing CNV profile of ELAVL1 in each tumor type. (E) The heterozygous and homozygous CNV profile of ELAVL1 in each tumor type, including the percentage of amplification and deletion. (F) Bubble plots showing the correlation of CNV with ELAVL1 mRNA expression. FDR, false discovery rate. CNV, copy number variation.

Modification patterns of ELAVL1 in pan-cancer

Epigenetic modifications and post-transcriptional modifications (PTMs) are crucial in influencing the onset and progression of cancers. [55, 56, 57, 58] By utilizing the UALCAN database, we first examined the promoter methylation levels of tumor and normal tissues, and the results showed that the ELAVL1 promoter methylation levels were upregulated in COAD, HNSC, KIRC, LUSC, and PAAD, whereas they were downregulated in PCPG (Supplementary Figure S2A). In addition, we observed a negative correlation between ELAVL1 mRNA expression levels and methylation levels (Supplementary Figure S2B).

We then explored the PTMs of ELAVL1. Figure 5A shows the common PTM sites of ELAVL1 obtained from the PhosphoSite database, indicating that S197, S200, and S202 were the most frequent phosphorylation sites. These findings were subsequently verified using the UACLAN database. We found that the ELAVL1 phosphorylation levels at these specific sites were elevated in tumor tissues (Figure 5B). Collectively, these results indicate the potential involvement of ELAVL1 in epigenetic modifications and PTMs.

Figure 5 Modification and alternative splicing patterns of ELAVL1 in pan-cancer. (A) An overview of Modification sites of ELAVL1 obtained from PhosphoSite database. (B) Box plots showing the different phosphorylated ELAVL1 levels between tumor and normal tissues using UALCAN database. (C) Reads-in, reads-out, and PSI values of ELAVL1_alt_3prime_134894 among pan-cancer, adjacent samples, and healthy tissues using OncoSplicing database. (D) PSI differences when comparing tumors and corresponding healthy or adjacent tissues and the association between ELAVL1_alt_3prime_134894 events and prognosis using OncoSplicing database. *P < 0.05, **P < 0.01, and ****P < 0.0001.
Figure 5

Modification and alternative splicing patterns of ELAVL1 in pan-cancer. (A) An overview of Modification sites of ELAVL1 obtained from PhosphoSite database. (B) Box plots showing the different phosphorylated ELAVL1 levels between tumor and normal tissues using UALCAN database. (C) Reads-in, reads-out, and PSI values of ELAVL1_alt_3prime_134894 among pan-cancer, adjacent samples, and healthy tissues using OncoSplicing database. (D) PSI differences when comparing tumors and corresponding healthy or adjacent tissues and the association between ELAVL1_alt_3prime_134894 events and prognosis using OncoSplicing database. *P < 0.05, **P < 0.01, and ****P < 0.0001.

Link of alternative ELAVL1 splicing to survival outcomes

Specific alterations in gene splicing within tumor cells might facilitate tumor progression, and discovering these alternative splicing (AS) events is important for enhancing survival outcomes. [59, 60, 61] The detection of clinically relevant ELAVL1 AS events was achieved using the OncoSplicing server. Figure 5C illustrates the percent spliced in (PSI) of the ELAVL1_alt_3prime event, designated 134894, and Figure 5D presents the PSI differences between tumor tissues and normal or adjacent tissues, as well as their relationships with prognosis. Although minimal differences in expression levels were observed, we found that ELAVL1 AS events might be correlated with the clinical outcomes of patients with ACC, KICH, LGG, UCEC, etc.

Involvement of ELAVL1 in multiple oncogenic pathways and its correlation with immune activity

A functional enrichment analysis was performed to clarify the role of ELAVL1 in tumor cells. We collected hallmark gene sets and performed GSEA. The result indicated that ELAVL1 was positively associated with several proliferation biological processes (e.g., G2M checkpoint, mitotic spindle, and DNA repair), and negatively related to classic inhibitory oncogenic pathways (e.g., p53 pathways and TNF-α pathways) and immune activity (e.g., interferon responses and complement) (Figure 6A). We then collected CRISPR screening data for ELAVL1 among pan-cancer cell lines and found a negative score for ELAVL1 across a substantial number of cell lines, indicating inhibition of cell proliferation and/or cell death following ELAVL1 knockout (Figure 6B).

Subsequently, we assessed the correlation between ELAVL1 expression levels and both genomic status and immune cell infiltration based on the immunogenicity and DNA damage scores. The results were mainly consistent with our previous findings, according to the quartile values of ELAVL1 expression levels (Figure 6C). In addition, we investigated the potential distinction among immune subtypes based on ELAVL1 expression levels and found that a high ELAVL1 expression level had a greater proportion of C1 subtype (increasing angiogenic gene expression and a higher proliferation percentage) but a lower proportion of C3 subtype (fewer aneuploidies and fewer CNV changes) (Figure 6D). The tumor microenvironment (TME) is vital in regulating tumor progression and modulating the response to standard-of-care therapies. [62, 63, 64, 65] Therefore, we additionally assessed the correlation between ELAVL expression levels and TME components and observed an immunosuppressive tendency (Supplementary Figure S3). Moreover, inverse negative correlations between ELAVL and immune-related genes existed in most cancer types, except THCA (Supplementary Figure S4). Pan-cancer immunotherapy cohorts were also summarized and the results indicated that ELAVL might felicitously predict immunotherapy response (Figure 6E).

ELAVL1 as a promising biomarker of NPC progression

Since no studies have examined ELAVL1 expression or its effect on NPC prognosis, we then focused on whether ELAVL1 could be a promising biomarker of NPC progression. We first collected four GEO datasets (GSE12452, GSE53819, GSE61218, and GSE103611) containing NPC and normal tissues. Compared to normal tissues, NPC tissues exhibited significantly increased ELAVL1 mRNA expression, and ELAVL1 was elevated in metastatic NPC tissues (Figure 7A). The AUC values for these datasets were 0.884, 0.778, 0.950, and 0.726, respectively, demonstrating the diagnostic efficacies of elevated ELAVL1 expression in NPC (Figure S6A). Besides, we found that elevated ELAVL1 mRNA expression levels were correlated with higher pathological N, stage, grade, and more specific mutations in the TCGAHNSC dataset (Figure S6B and S6C). We also observed upregulation of ELAVL1 mRNA expression levels in the NPC cell line with radiotherapy resistance (Figure 7B).

We then stratified the patients derived from the GSE102349 dataset into two groups characterized by the optimal cutoff value. We found that higher ELAVL1 expression levels were linked to unfavorable progression-free survival in NPC patients (Figure 7C). Taken together, our results suggest that ELAVL1 might be an effective biomarker of NPC progression.

ELAVL1 knockdown inhibited NPC progression in vitro

Subsequently, we performed western blotting to verify the upregulation of ELAVL1 protein levels in NPC cell lines compared to the normal nasopharyngeal cell line N2-Tert. We then selected SUNE-1 and HONE-1 cell lines to determine whether ELAVL1 knockdown could inhibit NPC progression (Figure 7D). Small interfering RNAs (siRNAs) targeting ELAVL1 mRNA were designed and validated in NPC cell lines (Figure 7E). CCK-8 assays confirmed that ELAVL1 suppression led to a significantly decreased viability of NPC cell lines (Figure 7F). Additionally, the results of the Transwell and wound healing assays confirmed that ELAVL1 suppression markedly hindered NPC cell line migration (Figure 7G, 7H, and 7J). We subsequently performed colony formation assays and found that ELAVL1 knockdown significantly led to a reduction in NPC cell proliferation and stemness (Figure 7I and 7J). Moreover, we performed immunohistochemical (IHC) staining, and the results demonstrated that ELAVL1 was highly expressed in NPC tissues compared to adjacent normal tissues (Figure 8A). These in vitro findings underscore that ELAVL1 knockdown could impede the proliferation and migration of NPC cell lines.

ELAVL1 knockdown suppressed NPC progression in vivo

We established popliteal lymph node metastatic and lung metastatic models using the HONE-1 NPC cell line to further substantiate the role of ELAVL1 in vivo. Short hairpin RNA (shRNA) targeting ELAVL1 was applied and verified through western blotting (Figure 8B). ELAVL1-KD and control NPC cells were administered via injection into the footpads or tail veins of BALB/C nude mice (Figure 8C). For the popliteal lymph node metastatic model, we performed bioluminescence imaging and HE staining and observed that ELAVL1 knockdown inhibited the metastatic spread of NPC cells from the primary tumor located in the footpad to the popliteal space, resulting in tumor-draining lymph nodes with reduced sizes (Figure 8D, 8E, 8G, and 8H). Meanwhile, the tumor volumes at the primary site exhibited a significant reduction in the ELAVL1-KD group (Figure 8F–8H). For the lung metastatic model, the intravenous injection of ELAVL1-KD NPC cells exhibited a reduced lung metastatic burden compared to the control group, as evidenced by a decrease in overt metastatic lesions (Figure 8I–8L). Moreover, in these metastatic models, ELAVL1 knockdown inhibited the infiltration of NPC cells into normal tissues. Collectively, our data suggest that suppressing ELAVL1 expression might limit the migratory behavior of NPC cells and hinder metastasis in vivo.

Discussion

Cancer is a global public health challenge and a major threat to human life, and the appearance of bulk RNA sequencing enables the proper exploration of tumor progression. scRNA-seq is a precise methodology for identifying both intrinsic and extrinsic tumor characteristics.[66, 67] It is capable of distinguishing distinct cellular subsets, elucidating clonal diversity, and critically identifying the key factors that influence tumor heterogeneity. [68, 69, 70] Moreover, recent advancements in various omics methodologies, such as ST, proteomics, and metabolomics, have offered new insights into tumor heterogeneity. [71, 72, 73, 74, 75] Consequently, it is imperative to employ multi-omics approaches to elucidate the molecular characteristics associated with tumor progression.

The ELAVL1 gene encodes HuR, a protein located on human chromosome 19p13.2.[76] Numerous previous studies have characterized ELAVL1, demonstrating its significant role in post-transcriptional regulation. Under typical physiological conditions, ELAVL1 is primarily localized within the nucleus. However, in response to cellular stress, such as hypoxic conditions or the presence of proliferative signals, it interacts with AU-rich elements (AREs) commonly situated in the 3′ UTRs of mRNAs, including the encoding cytokines, growth factors, oncogenes, and inflammatory molecules, and translocates to the cytoplasm, where it modulates the stability and processing of these mRNAs.[77] Given its vital role in the regulation of numerous genes, ELAVL1 is considered a therapeutic target for cancer treatment. In recent decades, inhibitors specifically targeting it (e.g., MS-444, CMLD-2, and KH-3) have been developed and have exhibited effective outcomes. [78, 79, 80] However, no ELAVL1 inhibitors have progressed to clinical trials. Consequently, the safety and efficacy of ELAVL1 inhibitors in clinical applications require further investigation and validation.

Figure 6 ELAVL1 is involved in multiple oncogenic pathways and is correlated with immune activity. (A) Bubble plots showing the correlation between ELAVL1 mRNA expression levels and each hallmark gene set activity in TCGA pan-cancer dataset. NES, normalized enrichment score. (B) Violin plots showing the CRISPR scores of ELAVL1 in cell lines originated from different organs. (C) Heatmap showing mean values of immune responses and genomic status at different expression levels of ELAVL1 mRNA expression levels in pan-cancer. Q1, Q2, Q3, and Q4 means top 25%, 25%-50%, 50%-75%, and bottom 25% of ELAVL1 mRNA expression levels, respectively. (D) The differences in immune subtypes across pan-cancer samples based on high and low ELAVL1 mRNA expression levels. (E) Bar plots showing the AUC values evaluating the predictive efficacy of ELAVL1 expression in pan-cancer immunotherapy cohorts.
Figure 6

ELAVL1 is involved in multiple oncogenic pathways and is correlated with immune activity. (A) Bubble plots showing the correlation between ELAVL1 mRNA expression levels and each hallmark gene set activity in TCGA pan-cancer dataset. NES, normalized enrichment score. (B) Violin plots showing the CRISPR scores of ELAVL1 in cell lines originated from different organs. (C) Heatmap showing mean values of immune responses and genomic status at different expression levels of ELAVL1 mRNA expression levels in pan-cancer. Q1, Q2, Q3, and Q4 means top 25%, 25%-50%, 50%-75%, and bottom 25% of ELAVL1 mRNA expression levels, respectively. (D) The differences in immune subtypes across pan-cancer samples based on high and low ELAVL1 mRNA expression levels. (E) Bar plots showing the AUC values evaluating the predictive efficacy of ELAVL1 expression in pan-cancer immunotherapy cohorts.

Figure 7 ELAVL1 is a promising biomarker of NPC progression and ELAVL1 knockdown inhibited NPC progression in vitro. (A) Histogram analysis of ELAVL1 mRNA expression levels between normal and NPC tissues. (B) Histogram analysis of ELAVL1 mRNA expression levels between radiotherapy-sensitive and radiotherapy-resistant NPC cell lines. (C) Kaplan-Meier survival analysis of ELAVL1 in GSE102349 dataset. (D) Western blot of ELAVL1 protein expression level in N2-Tert and NPC cell lines. (E) Western blot analysis showing the verification of the silent efficiency of ELAVL1 in NPC cell lines. (F) CCK-8 assays evaluated cellular growth curves across groups. (G) Transwell assays evaluated the efficacy of migration across groups. (H) Wound healing assays evaluated the efficacy of migration across groups. (I) Colony formation assays evaluated the efficacy of proliferation and stemness across groups. (J) Histogram analysis of the results of Transwell assays, wound healing assays, and colony formation assays across groups. **P < 0.01, and ***P < 0.001.
Figure 7

ELAVL1 is a promising biomarker of NPC progression and ELAVL1 knockdown inhibited NPC progression in vitro. (A) Histogram analysis of ELAVL1 mRNA expression levels between normal and NPC tissues. (B) Histogram analysis of ELAVL1 mRNA expression levels between radiotherapy-sensitive and radiotherapy-resistant NPC cell lines. (C) Kaplan-Meier survival analysis of ELAVL1 in GSE102349 dataset. (D) Western blot of ELAVL1 protein expression level in N2-Tert and NPC cell lines. (E) Western blot analysis showing the verification of the silent efficiency of ELAVL1 in NPC cell lines. (F) CCK-8 assays evaluated cellular growth curves across groups. (G) Transwell assays evaluated the efficacy of migration across groups. (H) Wound healing assays evaluated the efficacy of migration across groups. (I) Colony formation assays evaluated the efficacy of proliferation and stemness across groups. (J) Histogram analysis of the results of Transwell assays, wound healing assays, and colony formation assays across groups. **P < 0.01, and ***P < 0.001.

Figure 8 ELAVL1 knockdown inhibited NPC progression in vivo. (A) IHC staining of the NPC and adjacent normal tissues. (B) Western blot analysis showing the verification of the efficiency of ELAVL1-KD in HONE-1 NPC cell line. (C) Anatomic representation for stablishing the nude mice model of popliteal lymph node metastasis. (D) Bioluminescence imaging showing the different metastatic efficacy between ELAVL1-KD and control groups in popliteal lymph node metastatic models. (E) Representative images of resected popliteal lymph nodes. (F) Representative images of resected primary footpad tumors. (G) HE staining images of popliteal lymph nodes and primary footpad tumors. (H) Histogram analysis of lymph nodes and primary footpad tumor volumes between ELAVL1-KD and control groups. (I) Bioluminescence imaging showing the different metastatic efficacy between ELAVL1-KD and control groups in lung metastatic models. (J) Representative resected and bioluminescence images of metastatic lung tissues. (K) HE staining images of lung metastatic models. (L) Histogram analysis of metastatic nodes per lung between ELAVL1-KD and control groups. **P < 0.01, and ***P < 0.001. IHC, immunohistochemical; NPC, nasopharyngeal carcinoma.
Figure 8

ELAVL1 knockdown inhibited NPC progression in vivo. (A) IHC staining of the NPC and adjacent normal tissues. (B) Western blot analysis showing the verification of the efficiency of ELAVL1-KD in HONE-1 NPC cell line. (C) Anatomic representation for stablishing the nude mice model of popliteal lymph node metastasis. (D) Bioluminescence imaging showing the different metastatic efficacy between ELAVL1-KD and control groups in popliteal lymph node metastatic models. (E) Representative images of resected popliteal lymph nodes. (F) Representative images of resected primary footpad tumors. (G) HE staining images of popliteal lymph nodes and primary footpad tumors. (H) Histogram analysis of lymph nodes and primary footpad tumor volumes between ELAVL1-KD and control groups. (I) Bioluminescence imaging showing the different metastatic efficacy between ELAVL1-KD and control groups in lung metastatic models. (J) Representative resected and bioluminescence images of metastatic lung tissues. (K) HE staining images of lung metastatic models. (L) Histogram analysis of metastatic nodes per lung between ELAVL1-KD and control groups. **P < 0.01, and ***P < 0.001. IHC, immunohistochemical; NPC, nasopharyngeal carcinoma.

We conducted a broad pan-cancer analysis to explore the role of ELAVL1. Our analyses demonstrated that it is overexpressed across various cancer types at both the mRNA and protein levels. The scRNA-seq and ST datasets revealed that it was predominantly localized in tumor cells within the TME. Besides, its high expression is significantly correlated with unfavorable outcomes in various cancer types, although its effects differ among cancer types. Genomic alterations and modifications of ELAVL1 were also found to be associated with tumor progression. NPC is regarded as having a strong propensity for metastasis and a distinct geographic distribution, and understanding the molecular mechanisms driving its progression and finding accurate therapeutic targets for NPC intervention are essential to reduce recurrence and metastatic spread. ELAVL1 was previously confirmed upregulated in NPC cell lines at mRNA levels,[81] and a recent study found that its ubiquitin degradation could affect NPC metastasis, indicating its essential role in NPC progression.[82] We found that ELAVL1 was elevated in NPC tissues and high ELAVL1 expression was correlated with an unfavorable prognosis in NPC patients. Besides, functional experiments revealed that ELAVL1 suppression hindered NPC cell proliferation and migration both in vitro and in vivo, further underscoring its potential as a therapeutic target in NPC.

Notably, the present study had certain limitations. First, our pan-cancer results were mainly obtained by combining data from various databases. Despite providing broad insights, this comprehensive approach might be prone to systematic errors typical of such analytical methods. To address these biases, normalization techniques were employed to decrease technical variations, and multiple datasets were used to verify the findings. Second, although our findings corroborate the involvement of ELAVL1 in NPC progression, additional explorations concentrating on specific molecular mechanisms are needed, such as how ELAVL1 interacts with other components in the TME and whether its dysregulation could impact immune escape.[83] Third, it is essential to explore the potential integration of ELAVL1 inhibitors with conventional therapeutic regimens in the treatment of NPC patients.

Conclusion

In summary, our study conducted the first comprehensive investigation into the significance of ELAVL1 across a pan-cancer scale and corroborated its functional role in NPC progression. ELAVL1 might play a diverse role during cancer progression, and targeting it could effectively inhibit NPC progression. These findings highlight ELAVL1 as a promising target for cancer treatment. Integrating its expression levels into clinical decisions has the potential to optimize treatment strategies and enhance patient outcomes.

Supplementary Information

Supplementary materials are only available at the official site of the journal (www.intern-med.com).


Address for Correspondence: Jianjun Li and Guoxian Huang, Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
#

These authors contributed equally to this work.


Funding statement: This work was supported by the National Natural Science Foundation of China (82373016, Xiong Zou), Guangzhou Clinical High-Tech Project (2024P-GX15, Jianjun Li), and the Youth Program of Esophageal Cancer Institute (Q202421, Wencheng Tan).

Acknowledgements

We gratefully acknowledge contributions from the public databases for free use. We thank Sparkle database (https://grswsci.top/) for providing the platform for multi-omics analyses.

  1. Author Contributions

    Research design: Jianjun Li and Guoxian Huang; data collection: Jindong Xie, Yi Xie, and Wencheng Tan; data analysis: Jindong Xie, Yi Xie, Wencheng Tan,and Yimeng Ye; manuscript preparation: Jindong Xie, Yi Xie, Wencheng Tan, Yimeng Ye, Xueqi Ou, Xiong Zou, ZhiqingHe, Jiarong Wu, Xinpei Deng, Hailin Tang, Longjun He, Kailai Li, Peng Luo,and Kunhao Bai. Manuscript editing: Jianjun Li and Guoxian Huang. All authors contributed to the article and approved the submitted version.

  2. Ethical Approval

    Animal experiments were performed in accordance with the guidelines of the Institutional Animal Care and Use Ethics Committee of SYSUCC (L102012024120B). This study was approved by the Ethics Committee of Guangdong Provincial People’s Hospital.

  3. Informed Consent

    The collection of samples was executed with informed consent.

  4. Conflict of Interest

    Not applicable.

  5. Use of Large Language Models, AI and Machine Learning Tools

    None declared.

  6. Data Availability Statement

    All data generated or analyzed during this study are included in this published article. Processed data support the findings of this study are available from the corresponding author upon reasonable request.

References

1 Soerjomataram I, Bray F. Planning for tomorrow: global cancer incidence and the role of prevention 2020-2070. Nat Rev Clin Oncol 2021;18:663–672.10.1038/s41571-021-00514-zSearch in Google Scholar PubMed

2 Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229–263.10.3322/caac.21834Search in Google Scholar PubMed

3 Cao W, Chen HD, Yu YW, Li N, Chen WQ. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020. Chin Med J (Engl) 2021;134:783–791.10.1097/CM9.0000000000001474Search in Google Scholar PubMed PubMed Central

4 Chen YP, Chan ATC, Le QT, Blanchard P, Sun Y, Ma J. Nasopharyngeal carcinoma. Lancet 2019;394:64-80.10.1016/S0140-6736(19)30956-0Search in Google Scholar PubMed

5 Toumi N, Ennouri S, Charfeddine I, Daoud J, Khanfir A. Prognostic factors in metastatic nasopharyngeal carcinoma. Braz J Otorhinolaryngol 2022;88:212–219.10.1016/j.bjorl.2020.05.022Search in Google Scholar PubMed PubMed Central

6 Yang Z, Peng Y, Wang Y, Yang P, Huang Z, Quan T, et al. KLF5 regulates actin remodeling to enhance the metastasis of nasopharyngeal carcinoma. Oncogene 2024;43:1779–1795.10.1038/s41388-024-03033-0Search in Google Scholar PubMed

7 Su ZY, Siak PY, Lwin YY, Cheah SC. Epidemiology of nasopharyngeal carcinoma: current insights and future outlook. Cancer Metastasis Rev 2024;43:919–939.10.1007/s10555-024-10176-9Search in Google Scholar PubMed

8 Wang Z, Fang M, Zhang J, Tang L, Zhong L, Li H, et al. Radiomics and Deep Learning in Nasopharyngeal Carcinoma: A Review. IEEE Rev Biomed Eng 2024;17:118–135.10.1109/RBME.2023.3269776Search in Google Scholar PubMed

9 Pua LJW, Mai CW, Chung FF, Khoo AS, Leong CO, Lim WM, et al. Functional Roles of JNK and p38 MAPK Signaling in Nasopharyngeal Carcinoma. Int J Mol Sci 2022;23:1108.10.3390/ijms23031108Search in Google Scholar PubMed PubMed Central

10 Siak PY, Heng WS, Teoh SSH, Lwin YY, Cheah SC. Precision medicine in nasopharyngeal carcinoma: comprehensive review of past, present, and future prospect. J Transl Med 2023;21:786.10.1186/s12967-023-04673-8Search in Google Scholar PubMed PubMed Central

11 Srikantan S, Gorospe M. HuR function in disease. Front Biosci (Landmark Ed) 2012;17:189–205.10.2741/3921Search in Google Scholar PubMed PubMed Central

12 Ma WJ, Cheng S, Campbell C, Wright A, Furneaux H. Cloning and characterization of HuR, a ubiquitously expressed Elav-like protein. J Biol Chem 1996;271:8144–8151.10.1074/jbc.271.14.8144Search in Google Scholar PubMed

13 López de Silanes I, Fan J, Yang X, Zonderman AB, Potapova O, Pizer ES, et al. Role of the RNA-binding protein HuR in colon carcinogenesis. Oncogene 2003;22:7146–7154.10.1038/sj.onc.1206862Search in Google Scholar PubMed

14 Ding F, Lu L, Wu C, Pan X, Liu B, Zhang Y, et al. circHIPK3 prevents cardiac senescence by acting as a scaffold to recruit ubiquitin ligase to degrade HuR. Theranostics 2022;12:7550–7566.10.7150/thno.77630Search in Google Scholar PubMed PubMed Central

15 Wu X, Xu L. The RNA-binding protein HuR in human cancer: A friend or foe?Adv Drug Deliv Rev 2022;184:114179.10.1016/j.addr.2022.114179Search in Google Scholar PubMed PubMed Central

16 Seufert L, Benzing T, Ignarski M, Müller RU. RNA-binding proteins and their role in kidney disease. Nat Rev Nephrol 2022;18:153–170.10.1038/s41581-021-00497-1Search in Google Scholar PubMed

17 Schultz CW, Preet R, Dhir T, Dixon DA, Brody JR. Understanding and targeting the disease-related RNA binding protein human antigen R (HuR). Wiley Interdiscip Rev RNA 2020;11:e1581.10.1002/wrna.1581Search in Google Scholar PubMed PubMed Central

18 Heinonen M, Bono P, Narko K, Chang SH, Lundin J, Joensuu H, et al. Cytoplasmic HuR expression is a prognostic factor in invasive ductal breast carcinoma. Cancer Res 2005;65:2157–2161.10.1158/0008-5472.CAN-04-3765Search in Google Scholar PubMed

19 Mitsunari K, Miyata Y, Asai A, Matsuo T, Shida Y, Hakariya T, et al. Human antigen R is positively associated with malignant aggressiveness via upregulation of cell proliferation, migration, and vascular endothelial growth factors and cyclooxygenase-2 in prostate cancer. Transl Res 2016;175:116–128.10.1016/j.trsl.2016.04.002Search in Google Scholar PubMed

20 Costantino CL, Witkiewicz AK, Kuwano Y, Cozzitorto JA, Kennedy EP, Dasgupta A, et al. The role of HuR in gemcitabine efficacy in pancreatic cancer: HuR Up-regulates the expression of the gemcitabine metabolizing enzyme deoxycytidine kinase. Cancer Res 2009;69:4567–4572.10.1158/0008-5472.CAN-09-0371Search in Google Scholar PubMed PubMed Central

21 Lim SJ, Lee SH, Joo SH, Song JY, Choi SI. Cytoplasmic expression of HuR is related to cyclooxygenase-2 expression in colon cancer. Cancer Res Treat 2009;41:87–92.10.4143/crt.2009.41.2.87Search in Google Scholar PubMed PubMed Central

22 Palomo-Irigoyen M, Pérez-Andrés E, Iruarrizaga-Lejarreta M, Barreira-Manrique A, Tamayo-Caro M, Vila-Vecilla L, et al. HuR/ELAVL1 drives malignant peripheral nerve sheath tumor growth and metastasis. J Clin Invest 2020;130:3848–3864.10.1172/JCI130379Search in Google Scholar PubMed PubMed Central

23 Majumder M, Chakraborty P, Mohan S, Mehrotra S, Palanisamy V. HuR as a molecular target for cancer therapeutics and immune-related disorders. Adv Drug Deliv Rev 2022;188:114442.10.1016/j.addr.2022.114442Search in Google Scholar PubMed PubMed Central

24 Cai Z, Zhai X, Xu J, Hong T, Yang K, Min S, et al. ELAVL1 regulates PD-L1 mRNA stability to disrupt the infiltration of CD4-positive T cells in prostate cancer. Neoplasia 2024;57:101049.10.1016/j.neo.2024.101049Search in Google Scholar PubMed PubMed Central

25 Adamoski D, M Dos Reis L, Mafra ACP, Corrêa-da-Silva F, Moraes-Vieira PMM, Berindan-Neagoe I, et al. HuR controls glutaminase RNA metabolism. Nat Commun 2024;15:5620.10.1038/s41467-024-49874-xSearch in Google Scholar PubMed PubMed Central

26 Liu C, Lin Y, Wang Y, Lin S, Zhou J, Tang H, et al. HuR promotes triglyceride synthesis and intestinal fat absorption. Cell Rep 2024;43:114238.10.1016/j.celrep.2024.114238Search in Google Scholar PubMed

27 Sun D, Wang J, Han Y, Dong X, Ge J, Zheng R, et al. TISCH: a comprehensive web resource enabling interactive single-cell transcriptome visualization of tumor microenvironment. Nucleic Acids Res 2021;49:D1420-D1430.10.1093/nar/gkaa1020Search in Google Scholar PubMed PubMed Central

28 Sengupta S, den Boon JA, Chen IH, Newton MA, Dahl DB, Chen M, et al. Genome-wide expression profiling reveals EBV-associated inhibition of MHC class I expression in nasopharyngeal carcinoma. Cancer Res 2006;66:7999–8006.10.1158/0008-5472.CAN-05-4399Search in Google Scholar PubMed

29 Bao YN, Cao X, Luo DH, Sun R, Peng LX, Wang L, et al. Urokinase-type plasminogen activator receptor signaling is critical in nasopharyngeal carcinoma cell growth and metastasis. Cell Cycle 2014;13:1958–1969.10.4161/cc.28921Search in Google Scholar PubMed PubMed Central

30 Fan C, Wang J, Tang Y, Zhang S, Xiong F, Guo C, et al. Upregulation of long non-coding RNA LOC284454 may serve as a new serum diagnostic biomarker for head and neck cancers. BMC Cancer 2020;20:917.10.1186/s12885-020-07408-wSearch in Google Scholar PubMed PubMed Central

31 Tang XR, Li YQ, Liang SB, Jiang W, Liu F, Ge WX, et al. Development and validation of a gene expression-based signature to predict distant metastasis in locoregionally advanced nasopharyngeal carcinoma: a retrospective, multicentre, cohort study. Lancet Oncol 2018;19:382–393.10.1016/S1470-2045(18)30080-9Search in Google Scholar PubMed

32 Zhang L, MacIsaac KD, Zhou T, Huang PY, Xin C, Dobson JR, et al. Genomic Analysis of Nasopharyngeal Carcinoma Reveals TME-Based Subtypes. Mol Cancer Res 2017;15:1722–1732.10.1158/1541-7786.MCR-17-0134Search in Google Scholar PubMed

33 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015;43:e47.10.1093/nar/gkv007Search in Google Scholar PubMed PubMed Central

34 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6(269):pl1.10.1126/scisignal.2004088Search in Google Scholar PubMed PubMed Central

35 Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res 2018;28:1747–1756.10.1101/gr.239244.118Search in Google Scholar PubMed PubMed Central

36 Liu CJ, Hu FF, Xie GY, Miao YR, Li XW, Zeng Y, et al. GSCA: an integrated platform for gene set cancer analysis at genomic, pharmacogenomic and immunogenomic levels. Brief Bioinform 2023;24:bbac558.10.1093/bib/bbac558Search in Google Scholar PubMed

37 Hornbeck PV, Zhang B, Murray B, Kornhauser JM, Latham V, Skrzypek E. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res 2015;43:D512-D520.10.1093/nar/gku1267Search in Google Scholar PubMed PubMed Central

38 Chandrashekar DS, Karthikeyan SK, Korla PK, Patel H, Shovon AR, Athar M, et al. UALCAN: An update to the integrated cancer data analysis platform. Neoplasia 2022;25:18–27.10.1016/j.neo.2022.01.001Search in Google Scholar PubMed PubMed Central

39 Zhang Y, Yao X, Zhou H, Wu X, Tian J, Zeng J, et al. OncoSplicing: an updated database for clinically relevant alternative splicing in 33 human cancers. Nucleic Acids Res 2022;50:D1340–D1347.10.1093/nar/gkab851Search in Google Scholar PubMed PubMed Central

40 Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Camb) 2021;2:100141.10.1016/j.xinn.2021.100141Search in Google Scholar PubMed PubMed Central

41 Meyers RM, Bryan JG, McFarland JM, Weir BA, Sizemore AE, Xu H, et al. Computational correction of copy number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells. Nat Genet 2017;49:1779–1784.10.1038/ng.3984Search in Google Scholar PubMed PubMed Central

42 Sturm G, Finotello F, Petitprez F, Zhang JD, Baumbach J, Fridman WH, et al. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics 2019;35:i436–i445.10.1093/bioinformatics/btz363Search in Google Scholar PubMed PubMed Central

43 Li R, Yan L, Jiu J, Liu H, Li D, Li X, et al. PSME2 offers value as a biomarker of M1 macrophage infiltration in pan-cancer and inhibits osteosarcoma malignant phenotypes. Int J Biol Sci 2024;20:1452–1470.10.7150/ijbs.90226Search in Google Scholar PubMed PubMed Central

44 Huang TX, Fu L. The immune landscape of esophageal cancer. Cancer Commun (Lond) 2019;39:79.10.1186/s40880-019-0427-zSearch in Google Scholar PubMed PubMed Central

45 Liu Z, Liu L, Weng S, Xu H, Xing Z, Ren Y, et al. BEST: a web application for comprehensive biomarker exploration on large-scale data in solid tumors. J Big Data 2023;10:165.10.1186/s40537-023-00844-ySearch in Google Scholar

46 Zhou X, Ying H, Sun Y, Zhang W, Luo P, Zhu S, et al. Homologous recombination deficiency (HRD) is associated with better prognosis and possibly causes a non-inflamed tumour microenvironment in nasopharyngeal carcinoma. J Pathol Clin Res 2024;10:e12391.10.1002/2056-4538.12391Search in Google Scholar PubMed PubMed Central

47 Yang Q, Zhou X, Fang J, Lin A, Zhang H, Cheng Q, et al. Development and validation of a radiosensitivity model to evaluate radiotherapy benefits in pan-cancer. Cancer Sci 2024;115:1820–1833.10.1111/cas.16168Search in Google Scholar PubMed PubMed Central

48 Xie J, Deng X, Xie Y, Zhu H, Liu P, Deng W, et al. Multi-omics analysis of disulfidptosis regulators and therapeutic potential reveals glycogen synthase 1 as a disulfidptosis triggering target for triple-negative breast cancer. MedComm (2020) 2024;5:e502.10.1002/mco2.502Search in Google Scholar PubMed PubMed Central

49 Birkbak NJ, McGranahan N. Cancer Genome Evolutionary Trajectories in Metastasis. Cancer Cell 2020;37:8–19.10.1016/j.ccell.2019.12.004Search in Google Scholar PubMed

50 Hackshaw A, Clarke CA, Hartman AR. New genomic technologies for multi-cancer early detection: Rethinking the scope of cancer screening. Cancer Cell 2022;40:109–113.10.1016/j.ccell.2022.01.012Search in Google Scholar PubMed

51 Zhang L, Wang Y, Guo Y, Chen H, Yu W, Zhang Z, et al. A comprehensive system for detecting rare single nucleotide variants based on competitive DNA probe and duplex-specific nuclease. Anal Chim Acta 2021;1166:338545.10.1016/j.aca.2021.338545Search in Google Scholar PubMed

52 Hamanaka K, Miyake N, Mizuguchi T, Miyatake S, Uchiyama Y, Tsuchida N, et al. Large-scale discovery of novel neurodevelopmental disorder-related genes through a unified analysis of single-nucleotide and copy number variants. Genome Med 2022;14:40.10.1186/s13073-022-01042-wSearch in Google Scholar PubMed PubMed Central

53 Graf RP, Eskander R, Brueggeman L, Stupack DG. Association of Copy Number Variation Signature and Survival in Patients With Serous Ovarian Cancer. JAMA Netw Open 2021;4:e2114162.10.1001/jamanetworkopen.2021.14162Search in Google Scholar PubMed PubMed Central

54 Tan RSYC, Ong WS, Lee KH, Lim AH, Park S, Park YH, et al. HER2 expression, copy number variation and survival outcomes in HER2-low non-metastatic breast cancer: an international multicentre cohort study and TCGA-METABRIC analysis. BMC Med 2022;20:105.10.1186/s12916-022-02284-6Search in Google Scholar PubMed PubMed Central

55 Deng X, Qing Y, Horne D, Huang H, Chen J. The roles and implications of RNA m6A modification in cancer. Nat Rev Clin Oncol 2023;20:507–526.10.1038/s41571-023-00774-xSearch in Google Scholar PubMed

56 Zhuang H, Yu B, Tao D, Xu X, Xu Y, Wang J, et al. The role of m6A methylation in therapy resistance in cancer. Mol Cancer 2023;22:91.10.1186/s12943-023-01782-2Search in Google Scholar PubMed PubMed Central

57 Ou X, Tan Y, Xie J, Yuan J, Deng X, Shao R, et al. Methylation of GPRC5A promotes liver metastasis and docetaxel resistance through activating mTOR signaling pathway in triple negative breast cancer. Drug Resist Updat 2024;73:101063.10.1016/j.drup.2024.101063Search in Google Scholar PubMed

58 Yang L, Shi J, Zhong M, Sun P, Zhang X, Lian Z, et al. NXPH4 mediated by m5C contributes to the malignant characteristics of colorectal cancer via inhibiting HIF1A degradation. Cell Mol Biol Lett 2024;29:111.10.1186/s11658-024-00630-5Search in Google Scholar PubMed PubMed Central

59 Bonnal SC, López-Oreja I, Valcárcel J. Roles and mechanisms of alternative splicing in cancer - implications for care. Nat Rev Clin Oncol 2020;17:457–474.10.1038/s41571-020-0350-xSearch in Google Scholar PubMed

60 Zhang Y, Qian J, Gu C, Yang Y. Alternative splicing and cancer: a systematic review. Signal Transduct Target Ther 2021;6:78.10.1038/s41392-021-00486-7Search in Google Scholar PubMed PubMed Central

61 Bradley RK, Anczuków O. RNA splicing dysregulation and the hallmarks of cancer. Nat Rev Cancer 2023;23:135–155.10.1038/s41568-022-00541-7Search in Google Scholar PubMed PubMed Central

62 Bejarano L, Jordāo MJC, Joyce JA. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov 2021;11:933–959.10.1158/2159-8290.CD-20-1808Search in Google Scholar PubMed

63 Liu L, Xie Y, Yang H, Lin A, Dong M, Wang H, et al. HPVTIMER: A shiny web application for tumor immune estimation in human papillomavirusassociated cancers. Imeta 2023;2:e130.10.1002/imt2.130Search in Google Scholar PubMed PubMed Central

64 Xie J, Lin X, Deng X, Tang H, Zou Y, Chen W, et al. Cancer-associated fibroblast-derived extracellular vesicles: regulators and therapeutic targets in the tumor microenvironment. Cancer Drug Resist 2025;8:2.10.20517/cdr.2024.152Search in Google Scholar PubMed PubMed Central

65 Glaviano A, Lau HS, Carter LM, Lee EHC, Lam HY, Okina E, et al. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025;18:6.10.1186/s13045-024-01634-6Search in Google Scholar PubMed PubMed Central

66 Ding S, Chen X, Shen K. Single-cell RNA sequencing in breast cancer: Understanding tumor heterogeneity and paving roads to individualized therapy. Cancer Commun (Lond) 2020;40:329–344.10.1002/cac2.12078Search in Google Scholar PubMed PubMed Central

67 Olbrecht S, Busschaert P, Qian J, Vanderstichele A, Loverix L, Van Gorp T, et al. High-grade serous tubo-ovarian cancer refined with single-cell RNA sequencing: specific cell subtypes influence survival and determine molecular subtype classification. Genome Med 2021;13:111.10.1186/s13073-021-00922-xSearch in Google Scholar PubMed PubMed Central

68 Biermann J, Melms JC, Amin AD, Wang Y, Caprio LA, Karz A, et al. Dissecting the treatment-naive ecosystem of human melanoma brain metastasis. Cell 2022;185:2591–2608.10.1016/j.cell.2022.06.007Search in Google Scholar PubMed PubMed Central

69 Xie J, Deng W, Deng X, Liang JY, Tang Y, Huang J, et al. Single-cell histone chaperones patterns guide intercellular communication of tumor microenvironment that contribute to breast cancer metastases. Cancer Cell Int 2023;23:311.10.1186/s12935-023-03166-4Search in Google Scholar PubMed PubMed Central

70 Xie J, Yang A, Liu Q, Deng X, Lv G, Ou X, et al. Single-cell RNA sequencing elucidated the landscape of breast cancer brain metastases and identified ILF2 as a potential therapeutic target. Cell Prolif 2024;57:e13697.10.1111/cpr.13697Search in Google Scholar PubMed PubMed Central

71 Mund A, Brunner AD, Mann M. Unbiased spatial proteomics with single-cell resolution in tissues. Mol Cell 2022;82:2335–2349.10.1016/j.molcel.2022.05.022Search in Google Scholar PubMed

72 Rao A, Barkley D, França GS, Yanai I. Exploring tissue architecture using spatial transcriptomics. Nature 2021;596:211–220.10.1038/s41586-021-03634-9Search in Google Scholar PubMed PubMed Central

73 Pan Y, Lei X, Zhang Y. Association predictions of genomics, proteinomics, transcriptomics, microbiome, metabolomics, pathomics, radiomics, drug, symptoms, environment factor, and disease networks: A comprehensive approach. Med Res Rev 2022;42:441–461.10.1002/med.21847Search in Google Scholar PubMed

74 Yang F, Xiao Y, Ding JH, Jin X, Ma D, Li DQ, et al. Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab 2023;35:84–100.10.1016/j.cmet.2022.09.021Search in Google Scholar PubMed

75 Liu C, Xie J, Lin B, Tian W, Wu Y, Xin S, et al. Pan-Cancer Single-Cell and Spatial-Resolved Profiling Reveals the Immunosuppressive Role of APOE+ Macrophages in Immune Checkpoint Inhibitor Therapy. Adv Sci (Weinh) 2024;11:e2401061.10.1002/advs.202401061Search in Google Scholar PubMed PubMed Central

76 Ma WJ, Furneaux H. Localization of the human HuR gene to chromosome 19p13.2. Hum Genet 1997;99:32–33.10.1007/s004390050305Search in Google Scholar PubMed

77 Fan XC, Steitz JA. Overexpression of HuR, a nuclear-cytoplasmic shuttling protein, increases the in vivo stability of ARE-containing mRNAs. EMBO J 1998;17:3448–3460.10.1093/emboj/17.12.3448Search in Google Scholar PubMed PubMed Central

78 Lang M, Berry D, Passecker K, Mesteri I, Bhuju S, Ebner F, et al. HuR Small-Molecule Inhibitor Elicits Differential Effects in Adenomatosis Polyposis and Colorectal Carcinogenesis. Cancer Res 2017;77:2424–2438.10.1158/0008-5472.CAN-15-1726Search in Google Scholar PubMed PubMed Central

79 Wu X, Lan L, Wilson DM, Marquez RT, Tsao WC, Gao P, et al. Identification and validation of novel small molecule disruptors of HuR-mRNA interaction. ACS Chem Biol 2015;10:1476–1484.10.1021/cb500851uSearch in Google Scholar PubMed PubMed Central

80 Wu X, Gardashova G, Lan L, Han S, Zhong C, Marquez RT, et al. Targeting the interaction between RNA-binding protein HuR and FOXQ1 suppresses breast cancer invasion and metastasis. Commun Biol 2020;3:193.10.1038/s42003-020-0933-1Search in Google Scholar PubMed PubMed Central

81 Hu W, Li H, Wang S. LncRNA SNHG7 promotes the proliferation of nasopharyngeal carcinoma by miR-514a-5p/ELAVL1 axis. BMC Cancer 2020;20:376.10.1186/s12885-020-06775-8Search in Google Scholar PubMed PubMed Central

82 Zhang P, Wang T, Chen K, Sun R, Cao X, Du M, et al. CircINADL promotes nasopharyngeal carcinoma metastasis by inhibiting HuR ubiquitin degradation and disrupting the hippo signaling pathway. Cell Signal 2025;126:111526.10.1016/j.cellsig.2024.111526Search in Google Scholar PubMed

83 Peng S, Lin A, Jiang A, Zhang C, Zhang J, Cheng Q, et al. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer 2024;23:58.10.1186/s12943-024-01972-6Search in Google Scholar PubMed PubMed Central

Published Online: 2025-05-08

© 2025 Jindong Xie, Yi Xie, Wencheng Tan, Yimeng Ye, Xueqi Ou, Xiong Zou, Zhiqing He, Jiarong Wu, Xinpei Deng, Hailin Tang, Longjun He, Kailai Li, Peng Luo, Kunhao Bai, Guoxian Huang, Jianjun Li, published by De Gruyter on behalf of the SMP

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Downloaded on 9.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/jtim-2025-0009/html
Scroll to top button