- Research
- Open access
- Published:
The impact of an RNA-binding protein group on regulating the RSPO-LGR4/5-ZNRF3/RNF43 module and the immune microenvironment in hepatocellular carcinoma
BMC Cancer volume 25, Article number: 751 (2025)
Abstract
Background
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality. RNA-binding proteins (RBPs) are potential therapeutic targets because of their role in tumor progression. This study investigated the interactions between specific HCC progression-associated RBPs (HPARBPs), namely, ILF3, PTBP1, U2AF2, NCBP2, RPS3, and SSB, in HCC and their downstream targets, as well as their impact on the immune microenvironment and their clinical value.
Methods
Tissue samples from human HCC, collected from 28 patients who experienced recurrence following postoperative adjuvant therapy were examined. The mRNA levels of RBPs and their prospective targets were quantified through RNA isolation and quantitative real-time PCR. Data from two public datasets were scrutinized for both expression and clinical relevance. Through Student’s t test and logistic regression, HPARBPs were identified. Enhanced cross-linking immunoprecipitation (eCLIP) experiments revealed RBP-RNA interactions in HepG2 cells. For functional enrichment, Metascape was used, whereas CIBERSORT was used to characterize the immune microenvironment.
Results
Public database analysis confirmed widespread RBP expression abnormalities in HCC (false discovery rate < 0.00001 and fold change ≥ 1.15 or ≤ 0.85), leading to the identification of 42 HPARBPs and core modules. eCLIP data analysis revealed the specificity of downstream target genes and binding site features for core HPARBPs (signal value > 3, P value < 0.01). Four core HPARBPs may bind to RNAs of genes in the RSPO-LGR4/5-ZNRF3/RNF43 module, affecting the Wnt pathway and HCC progression. Immunoinfiltration analysis revealed changes in the HCC immune microenvironment due to altered expression of relevant genes.
Conclusion
In our study, we identified core HPARBPs that might contribute to HCC progression by binding to RNAs in the RSPO-LGR4/5-ZNRF3/RNF43 module. Changes in the expression of HPARBPs affect the HCC immune microenvironment. Our findings offer novel insights into the regulatory network of Wnt pathway-related RBPs and their potential clinical value in HCC.
Introduction
Liver cancer, notably hepatocellular carcinoma (HCC), ranks eighth in prevalence and third in tumor-related mortality worldwide [1]. HCC accounts for 80% of liver cancer cases [2]. The landscape of liver diseases research is in continuous evolution, as researchers worldwide are delving deeper into the intricate genetic underpinnings of liver diseases development [3,4,5,6]. Large-scale HCC genome sequencing analyses have identified core driver genes, such as TERT and TP53, as initial molecular events, as well as other low-frequency driver genes [7]. In the field of tumor research, the abnormal changes in genes encoding RNA-binding proteins (RBPs) in tumors and the potential effects of downstream regulatory groups of proteins on tumor progression have gradually attracted the attention of researchers, who have focused on precision targeted therapy [8, 9].
RBPs are highly conserved and diverse proteins that participate in multiple aspects of RNA regulation, including transcription [10], translation [11], splicing [12, 13], polyadenylation [14], stability [15], and localization [16]. RBPs can form dynamic functional complexes by directly interacting with other proteins and RNAs, such as ribonucleoproteins, ultimately forming functional granules or bodies through phase separation [17]. RBPs serve as crucial regulators of cellular homeostasis, and their dysregulation is associated with various human diseases, including cancer [18].
In the study of HCC, researchers have investigated the impact and role of certain RBPs in the regulation of target RNAs. For instance, with respect to transcriptional regulation, the oncogenic factor NELFE directly binds to MYC in the nucleus, regulating its interaction with chromatin and influencing MYC-mediated transcription. NELFE also regulates MYC downstream targets in the cytoplasm, affecting their expression in posttranscriptional processes and ultimately contributing to tumorigenesis in HCC [19, 20]. In terms of alternative splicing, SRSF2 mediates alternative splicing events in HCC, promoting the generation of the GCH1-L isoform of GCH1, thus promoting HCC development [21]. In terms of stability, highly expressed HuR promotes the stability of MAT2A by binding to its 3’ UTR in HCC. It can also bind to the mRNA of β-catenin to inhibit its degradation. Moreover, an imbalance in the expression of HuR and certain other RBPs plays a crucial role in HCC cell proliferation [22, 23].
The aberrant overexpression of RBPs in tumors may lead to abnormal regulation of downstream target RNAs, resulting in tumorigenesis and tumor progression. Consequently, research on targeted RNA-binding protein therapy for tumors is actively underway [24]. For example, studies have shown that the small molecule inhibitor MS-444, which targets HuR, can effectively inhibit the growth of intestinal tumors [25]. Researchers are currently developing small-molecule drugs to disrupt the interaction between the key oncogenic factor HuR and its target mRNAs, offering a novel therapeutic approach for tumors with upregulated HuR expression [26].
Overall, RBPs play a significant role and have a profound impact on HCC, with studies on their function and targets having crucial biological and clinical importance. To explore all the target RNAs bound by RBPs in the physiological or pathological states of living cells or tissues, researchers have developed high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (HITS-CLIP) over the past two decades. This technique integrates the advantages of immunoprecipitation and high-throughput sequencing to elucidate all target RNAs via an antibody against a selected RBP, and its methodology is constantly improving [27].
In this study, we focused primarily on upregulated RBPs such as ILF3 (also known as NF90), PTBP1, U2AF2, NCBP2, RPS3, and SSB because of their differential expression and potential clinical relevance in HCC. Notably, some of these RBPs may share common downstream RNA targets, such as RNF43, ZNRF3, and LGR4. These target genes were identified through the analysis of publicly available high-throughput sequencing data specific to RBP-RNA interactions derived from enhanced cross-linking immunoprecipitation (eCLIP) experiments [28], which can be considered an advanced iteration of HITS-CLIP. Compared to HITS-CLIP, eCLIP demonstrates superior performance by minimizing RNA loss, reducing PCR duplicates, eliminating background noise with higher efficiency, substantially increasing library construction success rates, and enhancing target recognition accuracy. In addition to investigating the downstream regulatory networks of these RBPs, we utilized public database data and our own research cohort to explore the impact of their expression levels on the immune microenvironment and their clinical value in HCC.
Materials and methods
Human HCC samples
A total of 28 paraffin-embedded tissue samples were collected from patients who were diagnosed with HCC by the Pathology Department of Nanjing Drum Tower Hospital and who experienced recurrence following postoperative adjuvant therapy between January 2020 and January 2023 (Table 1). These patients who experienced recurrence were subsequently treated with a combination of anti-angiogenic targeted therapies and immunotherapy. Efficacy was evaluated by the Response Evaluation Criteria in Solid Tumors (RECIST). This study was approved by the Ethics Committee of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, with ethical approval number 2023-646-02. All patients provided written informed consent to participate in this study.
RNA isolation and quantitative real-time PCR (qRT‒PCR) analysis
The FFPE DNA/RNA extraction kit from Aperbio Technologies, Suzhou, China, was used to extract nucleic acids from formalin-fixed, paraffin-embedded tissue samples following the manufacturer’s instructions. Nucleic acid quantification was performed via a Colibri microvolume spectrophotometer (Titertek-Berthold, Pforzheim, Germany) and a Qubit Fluorometer 2.0 (Invitrogen, California, USA). cDNA synthesis was carried out according to the instructions of HiScript® III RT SuperMix for qPCR (+ gDNA wiper) (Vazyme, Nanjing, China). QRT‒PCR was performed via ChamQ Universal SYBR qPCR Master Mix (Vazyme, Nanjing, China) and was repeated three times on an ABI7500 system. Changes in the expression of genes (ILF3, PTBP1, U2AF2, NCBP2, RNF43, ZNRF3, and LGR4) at the mRNA level were detected. The primers used are listed in Table S1. The differences in expression measured by qRT‒PCR were compared via the Wilcoxon method for statistical analysis.
Public data sources
The gene expression and clinical data of HCC patients were obtained from the Cancer Genome Atlas (TCGA) liver hepatocellular carcinoma (LIHC) dataset via the Genomic Data Commons Data Portal (https://portal.gdc.cancer.gov/). Furthermore, the gene expression and clinical data of HCC patients from the Liver Cancer Institute (LCI) cohort were obtained from the Gene Expression Omnibus (GEO) GSE14520 dataset (http://www.ncbi.nlm.nih.gov/geo).
Processed data of the eCLIP experiment performed on the HepG2 cell line against six core HPARBP genes (ILF3, PTBP1, NCBP2, U2AF2, SSB, and RPS3) were downloaded from the Encyclopedia of DNA Elements (ENCODE) portal (https://www.encodeproject.org/) with the following identifiers: ENCFF071QDP, ENCFF726SQU, ENCFF692RZM, ENCFF721PWF, ENCFF848JWA and ENCFF301IJW.
Data processing
Differentially expressed RNA-binding proteins (DERBPs) were examined in both the TCGA and LCI cohorts via Student’s t test, with a false discovery rate (FDR) < 0.00001 and a fold change ≥ 1.15 or ≤ 0.85, while low-abundance genes were excluded. Subsequently, hepatocellular carcinoma progression-associated RNA-binding proteins (HPARBPs) were identified by logistic regression analysis with a significant P-value < 0.05 for both TNM stage and alpha-fetoprotein (AFP) level. Following the screening of HPARBPs, only the TCGA-LIHC cohort was utilized for further analysis on the basis of its HCC expression data. Differential analysis of only the TCGA-LIHC cohort was performed via DESeq2 (R package), with an FDR < 0.00001 and a fold change ≥ 1.15 or ≤ 0.85, while low-abundance genes were excluded.
The upstream analysis of eCLIP sequencing data was performed in accordance with the eCLIP-seq Processing Pipeline v2.2 20200409, which was released to the public on the ENCODE website. The bed format files were obtained for further mining. These bed format files, which contain information about specific binding sites (signal value > 3, P-value < 0.01), were sorted and then annotated by HOMER software on the basis of the hg38 reference genome.
Functional and pathway enrichment analysis
The web tool Metascape was used for functional and pathway enrichment analysis of specific gene lists on the basis of Gene Ontology (GO) Biological Processes, GO Cellular Components, GO Molecular Functions and the Kyoto Encyclopedia of Genes and Genomes (KEGG) [29]. All genes in the genome were used as the enrichment background. Terms with a P-value < 0.01, a minimum count of 3, and an enrichment factor > 1.5 (the enrichment factor is the ratio between the observed counts and the counts expected by chance) were collected and grouped into clusters on the basis of their membership similarities.
Immunoinfiltration analysis
CIBERSORT (R package), which uses the principle of linear support vector regression to deconvolute the expression matrix of 22 human immune cell subtypes and the TCGA-LIHC expression matrix normalized by transcripts per million (TPM), was used to explore the estimated proportion of immune cells in HCC patients. We subsequently screened out the immune cells that showed significant differences in infiltration between patients with low and high expression levels of specific genes via the Wilcoxon method and analyzed the correlations with those genes via the Spearman method.
Statistical analysis and visualization
All the statistical analyses were implemented via R software (version 4.2.1) and Microsoft Excel. For comparisons, Student’s t test (two-sided), the Wilcoxon rank-sum test and Spearman’s correlation analysis were performed as indicated. Visualization of the analysis results was accomplished via R packages, TBTools [30], SRplot [31] and Integrative Genomics Viewer (IGV) [32].
Results
Abnormal expression of RBPs appears to be prevalent in HCC
On the basis of public databases and preceding reports, we successfully retrieved expression profiles and clinical data from 887 clinical samples from the TCGA-LIHC and LCI cohorts. We subsequently performed differential analysis between HCC and normal tissues to obtain a list of DERBPs (FDR < 0.00001, fold change ≥ 1.15 or ≤ 0.85). Univariate logistic regression analysis of the aforementioned DERBPs was then performed to screen out those that exhibited significant correlations with TNM stage and the AFP level (P value < 0.05). This analysis yielded the identification of 248 (245 upregulated) and 126 (118 upregulated) HPARBPs in the TCGA and LCI cohorts, respectively (Fig. 1A and Table S2). Notably, almost all 42 overlapping HPARBPs in both cohorts exhibited interactions via protein‒protein interaction (PPI) analysis using STRING (Fig. 1B). The core module consisting of 13 core HPARBPs was discovered within these complex interactions of HPARBPs via the cytoHubba plugin of Cytoscape (Fig. 1C). Functional enrichment analysis revealed that these core HPARBPs are primarily implicated in mRNA metabolism, mRNA process regulation, RNA nuclear output, translation regulation, splicing, and are likely involved in the formation of cytoplasmic ribonucleoprotein particles (Fig. 1D). Additionally, a subset (6 out of 13 core HPARBPs) of the core module was found to have matched high-throughput sequencing data of its binding target RNAs in the liver cancer HepG2 cell line, namely, eCLIP data, available in the ENCODE database for further analysis. In conclusion, our findings indicate that RBPs are commonly upregulated in HCC, and we identified a set of interacting core HPARBPs associated with the progression of HCC that may participate in various RNA processes.
Identification and characterization of the core HPARBP network. (A) Schematic representation of the study design, outlining the approaches employed to achieve the core HPARBP network. (B) PPI network comprising 42 HPARBPs, illustrating the interaction relationships among these RNA-binding proteins. (C) Topological analysis of the HPARBP PPI network via the cytoHubba MNC algorithm, revealing a core module of HPARBPs with key members highlighted in a greater degree of red, indicating their relative importance within the module. The oval shape denotes open-access eCLIP data availability from ENCODE for these members. (D) GO and KEGG functional enrichment analysis results of the core module. The size of the circle represents the number of genes involved, and the abscissa represents the significance of GO terms or KEGG pathways
The core module of HPARBPs may be extensively involved in the binding and regulation of genes related to the Wnt pathway
Through integrative analysis of eCLIP data and genome annotation of binding sites, we observed that the significant binding sites (signal value > 3, P-value < 0.01) of the six core HPARBPs (ILF3, PTBP1, NCBP2, U2AF2, SSB, and RPS3) presented distinct proportions of genomic regions (Fig. 2A) and limited crossover between the downstream target regulatory groups of the core HPARBPs (Fig. 2B and Table S3). A functional enrichment analysis of the shared downstream targets of the core HPARBPs referenced above, excluding SSB due to its low overlap with the others, revealed that the core HPARBPs play a significant role in the regulation of genes associated with the negative control of epithelial cell proliferation and the Wnt signaling pathway (Fig. 2C). By integrating multiple functional annotated gene sets, we identified 485 genes related to the Wnt pathway, nearly half of which are potentially regulated by one or more core HPARBPs (Fig. 2D). Differential gene expression analysis of the TCGA-LIHC cohort revealed that the majority of Wnt pathway-related target genes were not significantly differentially expressed in HCC (adjusted P-value < 0.05 was considered significant) (Fig. 2E). These findings indicate that in HCC, core HPARBPs may engage with the Wnt signaling pathway by modulating downstream RNA target genes, albeit predominantly through mechanisms that do not significantly alter the expression levels of those target genes.
Target binding profile of core HPARBPs. (A) Pie chart of target binding genomic regions of the six core HPARBPs. (B) UpSet plot depicting the target genes of the six core HPARBPs. (C) Results of the GO and KEGG functional enrichment analyses of the common targets of the core HPARBPs. Wnt-related terms and pathways are highlighted with red characters. (D) Pie chart displaying the characteristics of Wnt pathway-related genes targeted by the core HPARBPs, providing a detailed view of the regulatory landscape of these genes. (E) Volcano map of Wnt pathway-related genes identified via TCGA-LIHC differential analysis, together with labels of target characteristics
Specific core HPARBPs may coregulate the RSPO-LGR4/5-ZNRF3/RNF43 module upstream of the Wnt pathway in HCC
Among the potential Wnt pathway-related targets of the six core HPARBPs, three members of the RSPO-LGR4/5-ZNRF3/RNF43 module upstream of the Wnt pathway, specifically RNF43, ZNRF3, and LGR4, can be commonly bound by specific core HPARBPs, including ILF3, NCBP2, PTBP1, and U2AF2, thereby exerting regulatory functions (Fig. 3). The IGV browser revealed that the binding regions of these specific HPARBPs in RNF43, ZNRF3, and LGR4 are consistent with the proportion characteristics observed in previous results (Figs. 2A and 3). ILF3, PTBP1, and U2AF2 were found to bind to the intron regions of the three targets, whereas NCBP2 bound to the promoter-TSS regions. Given their reported expression in the nucleus, we speculated that these four specific core HPARBPs (ILF3, NCBP2, PTBP1, and U2AF2) may interact with three target pre-mRNAs (RNF43, ZNRF3, and LGR4) in the nucleus, potentially influencing posttranscriptional processes.
The expression levels of four specific core HPARBPs significantly correlate with immune cell infiltration patterns in HCC, demonstrating high similarity
On the basis of the analysis of gene expression data from the TCGA-LIHC cohort and the leukocyte signature matrix (LM22), we identified the predominant immune cell types infiltrating HCC, which included M0 macrophages, CD8+ T cells, M2 macrophages, and resting memory CD4+ T cells (Fig. 4A). LIHC patients were then divided into a high expression group and a low expression group using the median expression level of a certain gene as a cutoff. The classification of expression levels for four specific core HPARBPs revealed common patterns: γδ T cells were significantly increased in the low-expression group for all four specific core HPARBPs (P-value < 0.05), follicular helper T cells were significantly elevated in the high-expression groups for three core HPARBPs (ILF3, PTBP1, and U2AF2) (P-value < 0.05), and activated natural killer (NK) cells were significantly more abundant in the low-expression groups for two specific core HPARBPs (ILF3 and U2AF2) (P-value < 0.05) (Fig. 4B-E). Spearman correlation analysis revealed a significant negative correlation between the expression levels of these four core HPARBPs and γδ T cells (P-value < 0.05), which aligns with previous findings (Fig. S4A-D). These comprehensive analyses shed light on the intricate interplay between the expression of specific core HPARBPs and the immune cell composition within the tumor microenvironment. We offer valuable insights into the potential regulatory functions of HPARBPs in modulating immune responses in HCC, particularly concerning γδ T cells.
Immunoinfiltration analysis in HCC patients on the basis of the expression of the four specific core HPARBPs. (A) Box plot illustrating the estimated proportion of immune cells in HCC patients. (B-E) Comparison of immune cell infiltration between the high- and low-expression groups of the core HPARBPs via the Wilcoxon test, with the median used as the group cutoff. Immune cell subsets with P-values < 0.05 are denoted in red. *P-value < 0.05; **P-value < 0.01; ***P-value < 0.001
The potential clinical guiding significance of core HPARBPs and their targets in the RSPO-LGR4/5-ZNRF3/RNF43 module awaits further validation
After analyzing tumor tissue samples from 28 patients who experienced HCC recurrence following post-adjuvant therapy at our institution (as detailed in Table 1) and comparing them with two normal control tissues via qRT‒PCR and differential analysis, we detected no significant differences in HPARBP expression levels (P-value >0.05) (Fig. S5A-D and Table 2). Additionally, the expression levels of LGR4 and RNF43, which are considered as downstream targets of the specific core HPARBPs in the RSPO-LGR4/5-ZNRF3/RNF43 module, did not significantly change (P-value >0.05), whereas the expression level of ZNRF3 was significantly downregulated (P-value <0.05) (Fig. 5A-C and Table 2). Furthermore, we performed an immunoinfiltration analysis focusing on these three target genes. By analyzing the differences between the high- and low-expression groups via correlation analysis, we discovered that the expression levels of these genes were significantly positively correlated with those of naive B cells and negatively correlated with those of activated NK cells (P-value <0.05) (Fig. 5D-F and S4E-G). Notably, the immune microenvironment attributes of ZNRF3 and RNF43 display substantial similarity. To delve deeper into the correlation between the expression levels of these genes and the efficacy of combining anti-angiogenesis targeted drugs with immunotherapy, we reclassified the locally sourced HCC cohort using the RECIST guidelines and performed differential analysis of qRT‒PCR data from various HCC immunotherapy response groups. The results revealed that the expression levels of these genes did not significantly differ among the various immunotherapy response groups and normal tissues (P-value > 0.05), except for ZNRF3, which was significantly downregulated in the stable disease (SD) group compared with the normal group (P-value < 0.05) (Fig. 6A-G and Table 2). Furthermore, we employed ordinal logistic regression analysis to explore the relationships between the expression levels of seven genes and treatment response outcomes. However, the model lacked statistical significance and did not demonstrate any significant correlations (P-value > 0.05) (data not shown). The weighted average expression levels of all seven genes also showed no significant differences (P-value > 0.05) (Fig. 6H). Taken together, our analysis of HCC tumor tissues subsequent to adjuvant therapy revealed no significant change in the expression levels of HPARBPs together with downstream LGR4 and RNF43. However, ZNRF3 was significantly downregulated both in patients who experienced recurrence after adjuvant therapy for HCC and in those in the SD group after immunotherapy. The expression of these three downstream targets was correlated with the infiltration levels of naive B cells and activated NK cells.
Comparison of the expression levels and immune microenvironments of common target genes. (A-C) Box plot of RT‒qPCR expression levels of target genes between posttreatment HCC tumor and normal samples via the Wilcoxon rank-sum test for comparison. (D-F) Comparison of immune cell infiltration between the high- and low-expression groups of target genes, using the median as the group cutoff. Immune cell subsets with P-values < 0.05 are marked in red. *P-value < 0.05; **P-value < 0.01
Comparative analysis of gene expression levels across HCC immunotherapy response groups and normal tissues. (A-G) Box plot of the expression levels of relevant genes across the normal group and various HCC immunotherapy response groups generated via the Wilcoxon rank-sum test for comparison. (H) Box plot of the total expression levels of all relevant genes across the normal group and various HCC immunotherapy response groups generated via the Wilcoxon rank-sum test for comparison. AverageExpr calculates the weighted average expression of seven relevant genes for each sample within different groups. ns, not significant; *P-value < 0.05; PD, progressive disease; PR, partial response; SD, stable disease
Discussion
Historically, studies of RBPs have focused primarily on one-on-one interactions with target RNAs. However, in the 1990s, Gao et al. introduced a groundbreaking method using human brain RNAs that revealed that HuB could bind multiple mRNAs in vitro [33]. This pivotal work paved the way for understanding global mRNA targeting and established the foundation for the posttranscriptional RNA regulon theory [34, 35]. RBPs play crucial roles in cancer by controlling numerous mRNAs encoding proto-oncogenes, growth factors, and cell cycle regulators. Changes in RBP expression or localization can profoundly affect gene expression patterns, as reflected by the stark differences in global RNA expression levels between cancerous and normal tissues [36,37,38].
We initially used data from two HCC cohorts to identify HPARBPs and core modules, followed by enrichment analysis. These findings suggest that HPARBPs likely interact with specific target groups as functional complexes, regulating various posttranscriptional processes of target RNAs. Previous studies have reported that functional complexes are formed by several RBPs and other proteins/RNAs, such as spliceosomes [39], stress granules [40], and processing bodies [41]. Our study identified potential molecular targets for future targeted therapies. However, further experiments are necessary to ascertain whether specific core HPARBPs form protein functional complexes that bind and regulate targets within the same spatial and temporal context.
The eCLIP experiment uses immunoprecipitation to isolate complexes of specific RBPs and their bound RNAs [28]. All target RNAs can be separated and sequenced after processing. The sequencing reads, followed by steps such as mapping and peak calling, provide the genomic coordinates of significant binding sites. Genome annotation on the basis of these coordinates allows us to uncover more information about binding sites. RBPs may perform diverse posttranscriptional regulatory functions by binding to different regions of their target RNAs [42]. We found that ILF3, PTBP1, and U2AF2 tend to bind to the intron regions of target RNAs, suggesting that they may play crucial roles in pre-mRNA splicing regulation [43]. NCBP2 and SSB, which preferentially bind to promoter-TSS regions, likely play significant roles in translational control and mRNA stability [44, 45].
Although our initial screening of core HPARBPs was based on data from public databases via bioinformatics, statistics, and topology methods, the four specific core HPARBPs we ultimately focused on have been reported to have important clinical significance in HCC. ILF3, PTBP1, U2AF2, and NCBP2 are significantly upregulated in HCC and contribute to tumor progression through distinct molecular mechanisms involving nuclear RNA processing and gene posttranscriptional regulation, which affect the proliferation, migration, and invasion of tumor cells, with the expression levels of some of these genes closely related to patient prognosis [46,47,48,49,50,51,52,53]. These proteins reportedly interact with downstream target RNAs and play a role in tumorigenesis and tumor development. Specifically, ILF3 primarily acts in the nucleus by modulating the mRNAs of specific genes, such as CCNE1, IRF3, and IRF9, influencing the cell cycle and immune response [54]. PTBP1 binds to the precursor mRNAs of AXL and NUMB, leading to splicing variations that produce aberrant transcripts, thereby promoting tumor migration and invasion [50, 55, 56]. Concurrently, PTBP1 and U2AF2 competitively interact to regulate AXL splicing, further impacting tumor development [55]. As a splicing factor, U2AF2 is overexpressed in HCC and enhances the stability of EXO1 mRNA through interaction with the lncRNA CECR7, facilitating tumor progression [57]. These findings not only reveal the complexity of multiple crucial gene expression regulatory networks within HCC but also validate the rationality of our screening methodology and highlight the significant role of these four specific core HPARBPs. However, our findings indicate that the majority of these aberrant alterations in target RNAs are not directly manifested at the transcriptional level.
According to our results, the four specific core HPARBPs mentioned above may collectively bind and regulate the RNAs of LGR4, ZNRF3, and RNF43 within the RSPO-LGR4/5-ZNRF3/RNF43 module, which in turn exerts profound effects on downstream Wnt signaling pathways. In the mature healthy liver, the Wnt pathway is predominantly inactive. However, during processes of cell renewal and regeneration, as well as in certain pathological conditions, diseases, precancerous states, and cancers, the Wnt pathway can be reactivated [58]. The Wnt/β-catenin pathway governs numerous cellular processes involved in the initiation, proliferation, survival, migration, differentiation, and apoptosis of HCC [59]. The Wnt/β-catenin pathway is activated in up to 50% of HCC cases in the liver [60, 61]. The activated Wnt pathway collaborates with multiple signaling cascades to drive the formation and development of HCC and exerts its effects through downstream effector molecules [62].
RNF43 and ZNRF3, two negative feedback regulators of the Wnt pathway, function as E3 ubiquitin ligases that specifically target Frizzled (FZD) receptors for rapid endolysosomal degradation. Conversely, R-spondins (RSPO), ligands of LGR4-5-6 receptors, interact with ZNRF3/RNF43 to counteract FZD membrane clearance mediated by these proteins, thereby increasing both the strength and duration of Wnt signaling [63,64,65,66,67]. LGR4 and RNF43 have been reported to be overexpressed in HCC tissues [68, 69], but the changes in ZNRF3 expression remain unclear. ZNRF3 expression was found to be decreased in liver cancer tissues and HepG2 cells, which contradicts the results reported in the TCGA database [70]. In vitro experiments revealed that LGR4 can act as an oncogene in HCC and that its expression level is positively correlated with the tumor size, microvascular invasion, TNM stage and pathological differentiation grade of HCC patients [69, 71]. As Wnt pathway-related genes, ZNRF3 and RNF43 have been reported to be mutated in HCC, and patients with ZNRF3 mutations have been reported to have a poorer prognosis [63, 72,73,74]. ZNRF3 and RNF43 predispose people to liver cancer by controlling the proliferative, differentiation and lipid metabolic states of hepatocytes [63].
The upregulation of RNF43 expression appears to contradict the activation of the Wnt pathway in HCC, which may be the result of the combined activation of other bypass pathways. The influence of aberrant HPARBP expression on downstream targets in HCC remains largely unexplored. Given that the regulatory impact of HPARBPs is mostly absent at the transcriptional level of their targets, we propose that HPARBP abnormalities could lead to inactivated, unstable, or oncogenic products in the RSPO-LGR4/5-ZNRF3/RNF43 module and other carcinogenic pathways, potentially hindering their inherent suppression of the Wnt pathway. Perturbations within the complex Wnt upstream network could trigger compensatory mechanisms, resulting in abnormal activation of the Wnt pathway [75]. Furthermore, epigenetic alterations and crosstalk with other signaling pathways are also conjectured to substantially contribute to this activation process [76, 77]. Our hypothesis provides new insights into the tumor progression and regulatory mechanisms of HCC and offers a potential research direction for those who are interested in the interaction between RBP groups and downstream target groups in the tumor environment. However, the mechanism by which RBPs control the RSPO-LGR4/5-ZNRF3/RNF43 module to affect the Wnt pathway still requires further validation in in vivo and in vitro models.
Additionally, some RBPs have been reported to significantly influence antitumor immunity by modulating immune cell activity or infiltration within the tumor microenvironment, ultimately affecting tumor progression [78,79,80,81,82,83]. Therefore, we explored the impact of specific core HPARBPs on the immune microenvironment. The altered expression of the four HPARBPs has a similar effect on specific immune cell infiltration patterns, suggesting that they may synergistically regulate downstream oncogenes or tumor suppressor genes through common pathways or mechanisms, ultimately affecting the tumor microenvironment. Notably, their elevated expression in HCC could impair γδ T-cell functionality or hinder γδ T-cell production pathways. γδ T cells, which exhibit a distinctive capacity to recognize stress-associated antigens on tumor cells without major histocompatibility complex restriction, present a promising avenue for hematological and solid tumor immunotherapy. These cells circumvent immune evasion, mitigate rejection, and function as potent antigen-presenting cells (APCs), thereby increasing the prospects of cancer therapy [84]. In the context of HCC, γδ T cells contribute to the antitumor response and are linked to a favorable prognosis. However, within the tumor microenvironment, these cells may acquire protumorigenic attributes [85]. Our research sheds new light on the potential RBP regulators of γδ T cells in HCC and related immunotherapeutic strategies. Nonetheless, whether the observed alterations in γδ T cells correlate with the upstream modulation of the Wnt pathway by HPARBPs, specifically through the RSPO-LGR4/5-ZNRF3/RNF43 module, warrants additional investigation.
The expression of the HPARBP targets, ZNRF3 and RNF43 in HCC is inversely related to the infiltration of activated NK cells. Acting as E3-ubiquitin ligases, these proteins target the cytoplasmic loops of FZD receptors for ubiquitination, leading to their accelerated endolysosomal degradation [63]. This synergistic role aligns with their consistent immune microenvironment attributes in our study. Furthermore, LGR4, ZNRF3, and RNF43 each exhibited positive correlations with naive B cells, indicating a potential regulatory relationship between the RSPO-LGR4/5-ZNRF3/RNF43 axis and naive B cells. However, the immune microenvironment properties of these four HPARBPs and their downstream targets are not uniformly matched, supporting our prior findings that HPARBP-mediated modulation of target RNAs may not directly manifest at the transcriptional levels of those targets. Notably, the results of the immune infiltration analysis need to be validated in HCC tissue samples using experiments such as tissue staining to verify the key genes and associated immune markers, which is what we will be working on in the future.
As mentioned earlier, the specific core HPARBPs and their downstream target genes, with the exception of ZNRF3, exhibit widespread upregulation in HCC. However, in recurrent HCC tissues following postoperative adjuvant therapy, their expression levels were not significantly different from those in normal tissues. This finding suggests that the current clinical approach may intervene in the expression of these genes from overexpression to normal levels and highlights the importance of our research subjects for further investigation into the clinical therapy of HCC. The expression of ZNRF3 remained lower than that in normal tissues both before and after recurrence, and it continued to be downregulated in the SD group following immunotherapy for recurrent patients. These findings suggest that the expression level of ZNRF3 may serve as a potential biomarker to predict the efficacy of adjuvant therapy after hepatectomy, recurrence, and the response to immunotherapy postrecurrence in HCC patients. In addition to ZNRF3, the expression levels of other related genes were not significantly different among the various immune response groups or between the treatment groups and the control group. This finding indicates that the predictive value of these related genes is not as anticipated and that they may serve only as prognostic indicators. In our exploration, it was not possible to entirely exclude the influence of factors such as limited sample size, individual variability in response to immunotherapy, and tumor genetic expression heterogeneity on determining whether these genes can serve as predictive markers for the efficacy of immunotherapy [86, 87]. Nonetheless, clues about the close relationships between these genes and tumor immunotherapy have been discovered, particularly regarding therapeutic outcomes [88,89,90,91]. Therefore, additional studies employing larger sample sizes, particularly when combined with HCC tumor samples prior to any treatment, are necessary to validate and update these preliminary findings. In addition, we will focus more on protein expression levels rather than transcriptional expression levels when we study the relationship between the expression levels of these key genes and clinical outcomes in the future.
Conclusions
In summary, we identified core HPARBPs that might contribute to HCC progression by binding to RNAs in the RSPO-LGR4/5-ZNRF3/RNF43 module (Fig. 7). Changes in the expression of HPARBPs affect the HCC immune microenvironment. Our findings offer novel insights into the regulatory network of Wnt pathway-related RBPs and their potential clinical value in HCC.
A schematic representation of the underlying molecular mechanisms of the specific HPARBPs and their common binding targets regulating the Wnt signaling pathway. Under normal physiological conditions, Wnt proteins interact with FZD and LRP5/6 to initiate Wnt/β-catenin signaling. Acting as negative regulators, ZNRF3 and RNF43 impede Wnt/β-catenin signaling by promoting the ubiquitination and subsequent degradation of the Wnt receptors FZD and LRP6, thereby deactivating Wnt/β-catenin signaling. RSPO interacts with LGR4/5 and ZNRF3/RNF43, triggering their ubiquitination and degradation. In the HCC state, four highly expressed specific core HPARBPs bind to downstream target RNAs, leading to the generation of abnormal mRNA and protein products. This aberrant process results in the abnormal degradation of FZD and ZNRF3/RNF43, ultimately causing the failure of Wnt signaling shutdown. Sustained Wnt signaling furthers tumor progression and, when combined with other unidentified factors, ultimately influences the tumor immune microenvironment
Data availability
In this study, publicly available data sets and online tools were utilized as described in the main text. The specific details are as follows: The Genomic Data Commons Data Portal (https://portal.gdc.cancer.gov/), Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo), the Encyclopedia of DNA Elements (ENCODE) portal (https://www.encodeproject.org/), STRING website (https://string-db.org/), Metascape website (https://metascape.org) and SRplot (https://www.bioinformatics.com.cn/en). The analyzed result data are provided in the SUPPLEMENTARY INFORMATION section.
Abbreviations
- AFP:
-
Alpha fetoprotein
- APCs:
-
Antigen presenting cells
- AXL:
-
AXL Receptor tyrosine kinase
- BMI:
-
Body mass index
- CECR7:
-
Cat eye syndrome chromosome region candidate 7
- CCNE1:
-
Cyclin E1
- DERBPs:
-
Differentially expressed RNA-binding proteins
- ENCODE:
-
Encyclopedia of DNA Elements
- eCLIP:
-
Enhanced cross-linking immunoprecipitation
- EXO1:
-
Exonuclease 1
- FDR:
-
False discovery rate
- FZD:
-
Frizzled
- GEO:
-
Gene Expression Omnibus
- GO:
-
Gene Ontology
- GCH1:
-
GTP cyclohydrolase 1
- HCC:
-
Hepatocellular carcinoma
- HPARBPs:
-
Hepatocellular carcinoma progression-associated RNA-binding proteins
- HITS-CLIP:
-
High-throughput sequencing of RNA isolated by cross-linking immunoprecipitation
- HuR:
-
Human antigen R
- IGV:
-
Integrative Genomics Viewer
- IRF3:
-
Interferon regulatory factor 3
- IRF9:
-
Interferon regulatory factor 9
- ILF3:
-
Interleukin enhancer-binding factor 3
- KEGG:
-
Kyoto Encyclopedia of Genes and Genomes
- LGR4:
-
Leucine-rich repeat-containing G protein–coupled receptor
- LCI:
-
Liver Cancer Institute
- LIHC:
-
Liver hepatocellular carcinoma
- MAT2A:
-
Methionine adenosyltransferase 2 A
- NK:
-
Natural killer
- NELFE:
-
Negative elongation factor complex member E
- NCBP2:
-
Nuclear cap binding protein subunit 2
- NF90:
-
Nuclear factor 90
- NUMB:
-
NUMB endocytic adaptor protein
- PR:
-
Partial response
- PTBP1:
-
Polypyrimidine tract binding protein 1
- PD:
-
Progressive disease
- PPI:
-
Protein‒protein interaction
- qRT‒PCR:
-
Quantitative real-time PCR
- RECIST:
-
Response evaluation criteria in solid tumors
- RPS3:
-
Ribosomal protein S3
- RNF43:
-
Ring finger protein 43
- RBPs:
-
RNA-binding proteins
- RSPO:
-
R-spondins
- SRSF2:
-
Serine- and arginine-rich splicing factor 2
- SSB:
-
Single-stranded DNA binding protein
- SD:
-
Stable disease
- TERT:
-
Telomerase reverse transcriptase
- TCGA:
-
The Cancer Genome Atlas
- TPM:
-
Transcript per million
- TP53:
-
Tumor protein P53
- U2AF2:
-
U2 small nuclear RNA auxiliary factor 2
- ZNRF3:
-
Zinc and ring finger protein 3
- CTNNB1:
-
β-catenin
References
Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clinicians. 2021;71(3):209–249. https://doiorg.publicaciones.saludcastillayleon.es/10.3322/caac.21660
Chrysavgis L, Giannakodimos I, Diamantopoulou P, Cholongitas E. Non-alcoholic fatty liver disease and hepatocellular carcinoma: Clinical challenges of an intriguing link. World Journal of Gastroenterology. 2022;28(3):310–331. https://doiorg.publicaciones.saludcastillayleon.es/10.3748/wjg.v28.i3.310
Toh MR, Wong EYT, Wong SH, et al. Global Epidemiology and Genetics of Hepatocellular Carcinoma. Gastroenterology. 2023;164(5):766–782. https://doiorg.publicaciones.saludcastillayleon.es/10.1053/j.gastro.2023.01.033
Zhai X, Zhang H, Xia Z, et al. Oxytocin alleviates liver fibrosis via hepatic macrophages. JHEP Rep. 2024;6(6):101032. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jhepr.2024.101032
Zhang H, Xia T, Xia Z, et al. KIF18A inactivates hepatic stellate cells and alleviates liver fibrosis through the TTC3/Akt/mTOR pathway. Cell Mol Life Sci. 2024;81(1):96. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-024-05114-5
Zhang X, Zhang P, Cong A, et al. Unraveling molecular networks in thymic epithelial tumors: deciphering the unique signatures. Front Immunol. 2023;14:1264325. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1264325
Shibata T. Genomic landscape of hepatocarcinogenesis. J Hum Genet. 2021;66(9):845–851. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s10038-021-00928-8
Zhang K, Barry AE, Lamm R, Patel K, Schafer M, Dang H. The role of RNA binding proteins in hepatocellular carcinoma. Advanced Drug Delivery Reviews. 2022;182. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2022.114114
Wang S, Sun Z, Lei Z, Zhang HT. RNA-binding proteins and cancer metastasis. Semin Cancer Biol. 2022;86(Pt 2):748–768. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcancer.2022.03.018
Kechavarzi B, Janga S. Dissecting the expression landscape of RNA-binding proteins in human cancers. Genome Biol. 2014;15(1):R14. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/gb-2014-15-1-r14
Xiao R, Chen JY, Liang Z, et al. Pervasive Chromatin-RNA Binding Protein Interactions Enable RNA-Based Regulation of Transcription. Cell. 2019;178(1):107–121.e18. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2019.06.001
David CJ, Manley JL. Alternative pre-mRNA splicing regulation in cancer: pathways and programs unhinged. Genes Dev. 2010;24(21):2343–2364. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/gad.1973010
Gautrey H, Jackson C, Dittrich AL, Browell D, Lennard T, Tyson-Capper A. SRSF3 and hnRNP H1 regulate a splicing hotspot of HER2 in breast cancer cells. RNA Biol. 2015;12(10):1139–1151. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/15476286.2015.1076610
Masamha CP, Xia Z, Yang J, et al. CFIm25 links alternative polyadenylation to glioblastoma tumour suppression. Nature. 2014;510(7505):412–416. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature13261
Guo X, Hartley RS. HuR contributes to cyclin E1 deregulation in MCF-7 breast cancer cells. Cancer Res. 2006;66(16):7948–7956. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-05-4362
Hüttelmaier S, Zenklusen D, Lederer M, et al. Spatial regulation of beta-actin translation by Src-dependent phosphorylation of ZBP1. Nature. 2005;438(7067):512–515. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature04115
Ripin N, Parker R. Formation, function, and pathology of RNP granules. Cell. 2023;186(22):4737–4756. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2023.09.006
Qin H, Ni H, Liu Y, et al. RNA-binding proteins in tumor progression. J Hematol Oncol. 2020;13(1):90. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-020-00927-w
Dang H, Takai A, Forgues M, et al. Oncogenic Activation of the RNA Binding Protein NELFE and MYC Signaling in Hepatocellular Carcinoma. Cancer Cell. 2017;32(1):101–114.e8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccell.2017.06.002
Dang H, Pomyen Y, Martin SP, et al. NELFE-Dependent MYC Signature Identifies a Unique Cancer Subtype in Hepatocellular Carcinoma. Sci Rep. 2019;9(1):3369. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-019-39727-9
Luo C, Cheng Y, Liu Y, et al. SRSF2 Regulates Alternative Splicing to Drive Hepatocellular Carcinoma Development. Cancer Res. 2017;77(5):1168–1178. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-16-1919
Ale-Agha N, Galban S, Sobieroy C, et al. HuR regulates gap junctional intercellular communication by controlling beta-catenin levels and adherens junction integrity. Hepatology. 2009;50(5):1567–1576. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hep.23146
Vázquez-Chantada M, Fernández-Ramos D, Embade N, et al. HuR/methyl-HuR and AUF1 regulate the MAT expressed during liver proliferation, differentiation, and carcinogenesis. Gastroenterology. 2010;138(5):1943–1953. https://doiorg.publicaciones.saludcastillayleon.es/10.1053/j.gastro.2010.01.032
Mohibi S, Chen X, Zhang J. Cancer the‘RBP’eutics–RNA-binding proteins as therapeutic targets for cancer. Pharmacology & Therapeutics. 2019;203:107390. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pharmthera.2019.07.001
Lang M, Berry D, Passecker K, et al. HuR Small-Molecule Inhibitor Elicits Differential Effects in Adenomatosis Polyposis and Colorectal Carcinogenesis. Cancer Res. 2017;77(9):2424–2438. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-15-1726
Wu X, Lan L, Wilson DM, et al. Identification and validation of novel small molecule disruptors of HuR-mRNA interaction. ACS Chem Biol. 2015;10(6):1476–1484. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/cb500851u
Licatalosi DD, Mele A, Fak JJ, et al. HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature. 2008;456(7221):464–469. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature07488
Van Nostrand EL, Pratt GA, Shishkin AA, et al. Robust transcriptome-wide discovery of RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nat Methods. 2016;13(6):508–514. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nmeth.3810
Zhou Y, Zhou B, Pache L, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10(1):1523. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-019-09234-6
Chen C, Wu Y, Li J, et al. TBtools-II: A “one for all, all for one” bioinformatics platform for biological big-data mining. Mol Plant. 2023;16(11):1733–1742. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molp.2023.09.010
Tang D, Chen M, Huang X, et al. SRplot: A free online platform for data visualization and graphing. PLoS One. 2023;18(11):e0294236. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0294236
Robinson JT, Thorvaldsdóttir H, Winckler W, et al. Integrative Genomics Viewer. Nat Biotechnol. 2011;29(1):24–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nbt.1754
Gao FB, Carson CC, Levine T, Keene JD. Selection of a subset of mRNAs from combinatorial 3’ untranslated region libraries using neuronal RNA-binding protein Hel-N1. Proc Natl Acad Sci U S A. 1994;91(23):11207–11211. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.91.23.11207
Keene JD. The globalization of messenger RNA regulation. Natl Sci Rev. 2014;1(2):184–186. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nsr/nwu004
Blackinton JG, Keene JD. Post-transcriptional RNA regulons affecting cell cycle and proliferation. Seminars in Cell & Developmental Biology. 2014;34:44–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2014.05.014
Bisogno LS, Keene JD. Analysis of post-transcriptional regulation during cancer progression using a donor-derived isogenic model of tumorigenesis. Methods. 2017;126:193–200. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ymeth.2017.05.012
Kechavarzi B, Janga SC. Dissecting the expression landscape of RNA-binding proteins in human cancers. Genome Biol. 2014;15(1):R14. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/gb-2014-15-1-r14
Bisogno LS, Keene JD. RNA regulons in cancer and inflammation. Curr Opin Genet Dev. 2018;48:97–103. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.gde.2017.11.004
Yang H, Beutler B, Zhang D. Emerging roles of spliceosome in cancer and immunity. Protein Cell. 2022;13(8):559–579. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13238-021-00856-5
Marcelo A, Koppenol R, de Almeida LP, Matos CA, Nóbrega C. Stress granules, RNA-binding proteins and polyglutamine diseases: too much aggregation? Cell Death Dis. 2021;12(6):592. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-021-03873-8
Ivanov P, Kedersha N, Anderson P. Stress Granules and Processing Bodies in Translational Control. Cold Spring Harb Perspect Biol. 2019;11(5):a032813. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a032813
Van Nostrand EL, Freese P, Pratt GA, et al. A large-scale binding and functional map of human RNA-binding proteins. Nature. 2020;583(7818):711–719. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-020-2077-3
Ule J, Blencowe BJ. Alternative Splicing Regulatory Networks: Functions, Mechanisms, and Evolution. Molecular Cell. 2019;76(2):329–345. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molcel.2019.09.017
Hawkins S, Mondaini A, Namboori SC, et al. ePRINT: exonuclease assisted mapping of protein-RNA interactions. Genome Biology. 2024;25(1):140. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13059-024-03271-1
Policastro RA, Zentner GE. Global approaches for profiling transcription initiation. Cell Reports Methods. 2021;1(5):100081. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.crmeth.2021.100081
Wang H, Wang R, Fang J. A spliceosome-associated gene signature aids in predicting prognosis and tumor microenvironment of hepatocellular carcinoma. Aging (Albany NY). 2023;15(11):4906–4925. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/aging.204765
Qiu J, Wu X, Luo Y, et al. Prognostic and immunotherapeutic predictive value of interleukin enhancer-binding factor 3 in hepatocellular carcinoma: Integrated bioinformatics and experimental analysis. Gene. 2023;856:147132. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.gene.2022.147132
Jiang W, Huang H, Ding L, et al. Regulation of cell cycle of hepatocellular carcinoma by NF90 through modulation of cyclin E1 mRNA stability. Oncogene. 2015;34(34):4460–4470. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2014.373
Kang H, Heo S, Shin JJ, et al. A miR-194/PTBP1/CCND3 axis regulates tumor growth in human hepatocellular carcinoma. The Journal of Pathology. 2019;249(3):395–408. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/path.5325
Cho CY, Chung SY, Lin S, et al. PTBP1-mediated regulation of AXL mRNA stability plays a role in lung tumorigenesis. Sci Rep. 2019;9(1):16922. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-019-53097-2
Mo Z, Qu H, Su R, et al. Effect of U2AF2 expression on proliferation and migration of hepatocellular carcinoma and its relationship with prognosis (in Chinese). Chinese Journal of Hepatic Surgery (Electronic Edition). 2023;12(3):336–341. https://doiorg.publicaciones.saludcastillayleon.es/10.3877/cma.j.issn.2095-3232.2023.03.017
Glasser E, Maji D, Biancon G, et al. Pre-mRNA splicing factor U2AF2 recognizes distinct conformations of nucleotide variants at the center of the pre-mRNA splice site signal. Nucleic Acids Res. 2022;50(9):5299–5312. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkac287
53. Zhou K, Yang J, Li X, Xiong W, Zhang P, Zhang X. N7-Methylguanosine Regulatory Genes Profoundly Affect the Prognosis, Progression, and Antitumor Immune Response of Hepatocellular Carcinoma. Front Surg. 2022;9:893977. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fsurg.2022.893977
Lodde V, Floris M, Munk R, et al. Systematic identification of NF90 target RNAs by iCLIP analysis. Scientific Reports. 2022;12(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-021-04101-1
Shen L, Lei S, Zhang B, et al. Skipping of exon 10 in Axl pre-mRNA regulated by PTBP1 mediates invasion and metastasis process of liver cancer cells. Theranostics. 2020;10(13):5719–5735. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.42010
He Z, Ni Q, Li X, Zhao M, Mo Q, Duo Y. PTBP1 promotes hepatocellular carcinoma progression by regulating the skipping of exon 9 in NUMB pre-mRNA. Heliyon. 2023;9(6):e17387. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.heliyon.2023.e17387
Zhao L, Zang Q, Liang G, Yao X. LncRNA CECR7 boosts hepatocellular carcinoma progression by recruiting RNA binding protein U2AF2 to enhance the stability of EXO1 mRNA. Heliyon. 2023;9(9):e19862. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.heliyon.2023.e19862
Perugorria MJ, Olaizola P, Labiano I, et al. Wnt-β-catenin signalling in liver development, health and disease. Nat Rev Gastroenterol Hepatol. 2019;16(2):121–136. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41575-018-0075-9
Khalaf AM, Fuentes D, Morshid AI, et al. Role of Wnt/β-catenin signaling in hepatocellular carcinoma, pathogenesis, and clinical significance. J Hepatocell Carcinoma. 2018;5:61–73. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/JHC.S156701
Vilchez V, Turcios L, Marti F, Gedaly R. Targeting Wnt/β-catenin pathway in hepatocellular carcinoma treatment. World J Gastroenterol. 2016;22(2):823–832. https://doiorg.publicaciones.saludcastillayleon.es/10.3748/wjg.v22.i2.823
Lee JM, Yang J, Newell P, et al. β-Catenin signaling in hepatocellular cancer: Implications in inflammation, fibrosis, and proliferation. Cancer Lett. 2014;343(1):90–97. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.canlet.2013.09.020
Xu C, Xu Z, Zhang Y, Evert M, Calvisi DF, Chen X. β-Catenin signaling in hepatocellular carcinoma. J Clin Invest. 2022;132(4):e154515. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI154515
Belenguer G, Mastrogiovanni G, Pacini C, et al. RNF43/ZNRF3 loss predisposes to hepatocellular-carcinoma by impairing liver regeneration and altering the liver lipid metabolic ground-state. Nat Commun. 2022;13(1):334. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-021-27923-z
Planas-Paz L, Orsini V, Boulter L, et al. The RSPO-LGR4/5-ZNRF3/RNF43 module controls liver zonation and size. Nat Cell Biol. 2016;18(5):467–479. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncb3337
Sun T, Annunziato S, Bergling S, et al. ZNRF3 and RNF43 cooperate to safeguard metabolic liver zonation and hepatocyte proliferation. Cell Stem Cell. 2021;28(10):1822–1837 e10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.stem.2021.05.013
Hao HX, Jiang X, Cong F. Control of Wnt Receptor Turnover by R-spondin-ZNRF3/RNF43 Signaling Module and Its Dysregulation in Cancer. Cancers. 2016;8(6):54. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers8060054
Annunziato S, Sun T, Tchorz JS. The RSPO-LGR4/5-ZNRF3/RNF43 module in liver homeostasis, regeneration, and disease. Hepatology. 2022;76(3):888–899. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hep.32328
Xie H, Xing C, Cao G, et al. Association of RNF43 with cell cycle proteins involved in p53 pathway. Int J Clin Exp Pathol. 2015;8(11):14995–15000.
Zhuo H, Miao S, Jin Z, et al. Metformin Suppresses Hepatocellular Carcinoma through Regulating Alternative Splicing of LGR4. J Oncol. 2022;2022:1774095. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2022/1774095
Liu M, Zhao H, Peng S, et al. Comprehensive analysis of zinc and ring finger 3 in prognostic value and pan-cancer immunity. The FASEB Journal. 2024;38(5):e23523. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.202301161R
Bi Y, Zhang L, Song Y, et al. Rspo2-LGR4 exacerbates hepatocellular carcinoma progression via activation of Wnt/β-catenin signaling pathway. Gastroenterol Hepatol. 2024;47(4):352–365. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.gastrohep.2023.05.016
Schulze K, Imbeaud S, Letouzé E, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 2015;47(5):505–511. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.3252
Ong CK, Subimerb C, Pairojkul C, et al. Exome sequencing of liver fluke-associated cholangiocarcinoma. Nat Genet. 2012;44(6):690–693. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.2273
Guichard C, Amaddeo G, Imbeaud S, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012;44(6):694–698. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.2256
Liu LJ, Xie SX, Chen YT, Xue JL, Zhang CJ, Zhu F. Aberrant regulation of Wnt signaling in hepatocellular carcinoma. World J Gastroenterol. 2016;22(33):7486–7499. https://doiorg.publicaciones.saludcastillayleon.es/10.3748/wjg.v22.i33.7486
Collu GM, Hidalgo-Sastre A, Brennan K. Wnt–Notch signalling crosstalk in development and disease. Cell Mol Life Sci. 2014;71(18):3553–3567. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-014-1644-x
Shorning BY, Dass MS, Smalley MJ, Pearson HB. The PI3K-AKT-mTOR Pathway and Prostate Cancer: At the Crossroads of AR, MAPK, and WNT Signaling. Int J Mol Sci. 2020;21(12):4507. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21124507
Zeng Q, Saghafinia S, Chryplewicz A, et al. Aberrant hyperexpression of the RNA binding protein FMRP in tumors mediates immune evasion. Science. 2022;378(6621):eabl7207. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.abl7207
Marasca F, Sinha S, Vadalà R, et al. LINE1 are spliced in non-canonical transcript variants to regulate T cell quiescence and exhaustion. Nat Genet. 2022;54(2):180–193. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41588-021-00989-7
Elcheva IA, Gowda CP, Bogush D, et al. IGF2BP family of RNA-binding proteins regulate innate and adaptive immune responses in cancer cells and tumor microenvironment. Front Immunol. 2023;14:1224516. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1224516
Xie H, Xi X, Lei T, Liu H, Xia Z. CD8 + T cell exhaustion in the tumor microenvironment of breast cancer. Front Immunol. 2024;15:1507283. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2024.1507283
Xia Z, Chen S, He M, et al. Editorial: Targeting metabolism to activate T cells and enhance the efficacy of checkpoint blockade immunotherapy in solid tumors. Front Immunol. 2023;14:1247178. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1247178
Deng Y, Shi M, Yi L, Naveed Khan M, Xia Z, Li X. Eliminating a barrier: Aiming at VISTA, reversing MDSC-mediated T cell suppression in the tumor microenvironment. Heliyon. 2024;10(17):e37060. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.heliyon.2024.e37060
Hu Y, Hu Q, Li Y, et al. γδ T cells: origin and fate, subsets, diseases and immunotherapy. Sig Transduct Target Ther. 2023;8(1):1–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-023-01653-8
He W, Hu Y, Chen D, et al. Hepatocellular carcinoma-infiltrating γδ T cells are functionally defected and allogenic Vδ2 + γδ T cell can be a promising complement. Clin Transl Med. 2022;12(4):e800. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ctm2.800
Craig AJ, von Felden J, Garcia-Lezana T, Sarcognato S, Villanueva A. Tumour evolution in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2020;17(3):139–152. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41575-019-0229-4
Nault JC, Villanueva A. Intratumor molecular and phenotypic diversity in hepatocellular carcinoma. Clin Cancer Res. 2015;21(8):1786–1788. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-14-2602
Zhou G, Sprengers D, Boor PPC, et al. Antibodies Against Immune Checkpoint Molecules Restore Functions of Tumor-Infiltrating T Cells in Hepatocellular Carcinomas. Gastroenterology. 2017;153(4):1107–1119.e10. https://doiorg.publicaciones.saludcastillayleon.es/10.1053/j.gastro.2017.06.017
Chen J, Lin Z, Liu L, et al. GOLM1 exacerbates CD8 + T cell suppression in hepatocellular carcinoma by promoting exosomal PD-L1 transport into tumor-associated macrophages. Signal Transduct Target Ther. 2021;6(1):397. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-021-00784-0
Nazitto R, Amon LM, Mast FD, et al. ILF3 Is a Negative Transcriptional Regulator of Innate Immune Responses and Myeloid Dendritic Cell Maturation. J Immunol. 2021;206(12):2949–2965. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.2001235
Jiang J, Mei J, Yi S, et al. Tumor associated macrophage and microbe: The potential targets of tumor vaccine delivery. Adv Drug Deliv Rev. 2022;180:114046. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2021.114046
Acknowledgements
The authors thank Professor Tanjun Tong’s team at Peking University for providing inspiration and references that were used in the screening of HPARBPs described in this paper. The authors also thank members of the Pathology Department and Oncology Department of Nanjing Drum Tower Hospital for their assistance in the collection of samples and clinical information.
Funding
Supported by fundings for Clinical Trials from the Affiliated Drum Tower Hospital, Medical School of Nanjing University (2024-LCYJ-PY-59); Medical New Technology Development Project of Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School (XJSFZLX202321); Project of The National Health Commission Health Development Research Center (No. WKZX2023CX020001).
Author information
Authors and Affiliations
Contributions
XZY, DZY, LZY, LSY, LJJ and SJ designed the research, interpreted the data, and revised the paper. XZY, DZY and LJJ performed the data analysis. LZY, CYQ, HST and LSY collected the tissue specimen and performed validation experiments. XZY drafted the paper. All of the authors approved the submitted and final versions.
Corresponding authors
Ethics declarations
Ethical approval
This study was approved by the Ethics Committee of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, with ethical approval number (2023-646-02).
Human ethics declaration
The study was conducted according to the guidelines laid down in the Declaration of Helsinki.
Consent for publication
The patient’s written informed consent was obtained for publication.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Additional File 1. Table S1.
Primers used for qRT-PCR experiments in this study
Additional File 2. Table S2.
Relevant results during the screening process of HPARBPs, related to Figure 1. DERBPs and HPARBPs in the TCGA and LCI cohort are listed in this file
Additional File 3. Table S3.
The binding target gene lists for the core HPARBPs, related to Figure 2. The target genes of each core HPARBPs arelisted and gathered in this file
Addtional File 4. Figure S4.
Relationship of core HPARBPs expression and immune cell subtypes in HCC patients. (A-G) The number on the abscissa represents the correlation coefficient based on Spearman’s correlation analysis, and the number on the ordinate represents the P-value, where P-values < 0.05 are marked in red. They all share the legend in graph G
Additional File 5. Figure S5.
Comparison of immunotherapy effectiveness of HPARBPs. (A-D) Box plot of RT-qPCR expression level of HPARBPs between post-treatment HCC tumor and normal samples using Wilcoxon rank-sum test for comparison
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Xie, Z., Dai, Z., Liu, Z. et al. The impact of an RNA-binding protein group on regulating the RSPO-LGR4/5-ZNRF3/RNF43 module and the immune microenvironment in hepatocellular carcinoma. BMC Cancer 25, 751 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-025-13874-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-025-13874-x