Skip to main content

ORIGINAL RESEARCH article

Front. Oncol., 26 January 2021
Sec. Cancer Genetics
This article is part of the Research Topic Interplay between RNA-binding Proteins and non-coding RNAs in Tumor Therapeutic Resistance View all 8 articles

Systematic Construction and Validation of an RNA-Binding Protein-Associated Model for Prognosis Prediction in Hepatocellular Carcinoma

Siyuan Tian&#x;Siyuan Tian1†Jingyi Liu&#x;Jingyi Liu2†Keshuai Sun&#x;Keshuai Sun1†Yansheng Liu&#x;Yansheng Liu1†Jiahao YuJiahao Yu1Shuoyi MaShuoyi Ma1Miao ZhangMiao Zhang1Gui JiaGui Jia1Xia Zhou*Xia Zhou1*Yulong Shang*Yulong Shang1*Ying Han*Ying Han1*
  • 1State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi’an, China
  • 2Department of Radiation Oncology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China

Background: Evidence from prevailing studies show that hepatocellular carcinoma (HCC) is among the top cancers with high mortality globally. Gene regulation at post-transcriptional level orchestrated by RNA-binding proteins (RBPs) is an important mechanism that modifies various biological behaviors of HCC. Currently, it is not fully understood how RBPs affects the prognosis of HCC. In this study, we aimed to construct and validate an RBP-related model to predict the prognosis of HCC patients.

Methods: Differently expressed RBPs were identified in HCC patients based on the GSE54236 dataset from the Gene Expression Omnibus (GEO) database. Integrative bioinformatics analyses were performed to select hub genes. Gene expression patterns were validated in The Cancer Genome Atlas (TCGA) database, after which univariate and multivariate Cox regression analyses, as well as Kaplan-Meier analysis were performed to develop a prognostic model. Then, the performance of the prognostic model was assessed using receiver operating characteristic (ROC) curves and clinicopathological correlation analysis. Moreover, data from the International Cancer Genome Consortium (ICGC) database were used for external validation. Finally, a nomogram combining clinicopathological parameters and prognostic model was established for the individual prediction of survival probability.

Results: The prognostic risk model was finally constructed based on two RBPs (BOP1 and EZH2), facilitating risk-stratification of HCC patients. Survival was markedly higher in the low-risk group relative to the high-risk group. Moreover, higher risk score was associated with advanced pathological grade and late clinical stage. Besides, the risk score was found to be an independent prognosis factor based on multivariate analysis. Nomogram including the risk score and clinical stage proved to perform better in predicting patient prognosis.

Conclusions: The RBP-related prognostic model established in this study may function as a prognostic indicator for HCC, which could provide evidence for clinical decision making.

Introduction

Hepatocellular carcinoma (HCC) is classified as one of the most prevalent cancers globally, posing a serious threat to human health. According to the global cancer statistics, there were nearly 782,000 cancer-associated deaths and 841,000 new cases of HCC, ranking fourth and sixth in all tumors, respectively (1). Although much progress have been made in the diagnosis and treatment of HCC, 5-year survival rate has not been improved largely due to high rates of recurrence and metastasis (2, 3). The occurrence and progression of HCC are driven by multiple processes affected by both genetic and environmental factors (4). In recent years, accumulating evidences have suggested that post-transcriptional modifications play an important role in the malignant phenotype of tumors, and these changes are strongly linked to the clinical course of patients (57). Thus, it’s of great significance to systematically explore relevant biological biomarkers for HCC management.

RNA-binding protein (RBP) is a general term for the group of proteins that exert its function by binding to RNA specifically, such as RNA maturation, transport, localization and translation. To date, human genome-wide screening has identified 1,542 RBP genes, accounting for 7.5% of all protein-coding genes (8). With few exceptions, most of RNAs are required to form RNA-protein complex to perform specific biological functions (9). In this process, RBPs exert essential role, especially in gene expression and the maintenance of genome integrity (10, 11). While the significance of post-transcriptional regulation to tumor initiation and progression has been recognized, the role of RBPs in HCC is still rudimentary. Several studies have demonstrated the aberrant expression of RBPs in HCC (6, 1214). For instance, RBP RPS3 was markedly up regulated in HCC tumor tissues and served as a critical tumor-promoting factor via up-regulating SIRT1. Targeting against the RPS3/SIRT1 pathway holds much promise as a target for therapeutic exploitation for the treatment of HCC (12). RBP eIF3C has been reported to promote cell growth in vitro and tumorigenicity in vivo (14). Furthermore, Yang et al. reported that RBP MEX3A could independently reflect the clinical course of HCC, and overexpression of MEX3A predicted poor prognosis (13). However, large-scale prognostic signature based on RBP genes in HCC has rarely been investigated. Therefore, a systematic study to explore prognosis-related RBP genes will be helpful to understand their roles in the initiation and progression of HCC, and will also have great value in guiding decision-making.

In this study, RBP genes expression data was derived from The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO) databases to develop and validate a prognostic model for long-term prognostic prediction of HCC patients. To make full use of the complementary significance of molecular expression and clinical features, we combined the prognostic model with clinical parameters to construct a comprehensive prognostic nomogram, further improving the predictive power.

Materials and Methods

Data Source

HCC GeneChip data (GSE 54236) was derived from the National Center for Biorechnology Information (NCBI) GEO DataSets. GSE 54236 was prepared from the Agilent GPL 6480 platform (Agilent-014850 Whole Human Genome Microarray 4×44K G4112F) including 81 human HCC tissue and 80 non-tumor tissue samples. Besides, we downloaded the level-three transcriptome RNA-sequencing data of LIHC from 344 samples with corresponding clinical data in the TCGA database for establishing a prognostic model. Meanwhile, to test the accuracy of the model, a validation set including clinical information and RNA-seq data was also downloaded from the ICGC database (LIRI-JP cohort). The detailed clinical information of TCGA dataset and ICGC dataset was shown in Supplementary Table 1. All data used in this study were freely available online.

Identification of Differently Expressed RNA-Binding Proteins

The GEO2R, an online analysis tool in GEO database, was used to identify differently expressed genes (DEGs) in HCC tissue samples and adjacent normal tissue samples (15). The log2|FC|≥0.5 and adjusted P-value ≤0.05 were set as the cut-off criterion. DEGs were then processed to generate a volcano plot with GraphPad Prism 7 Software. Next, we used the Venn diagram webtool to determine the overlapping between the DEGs and 1542 Human RNA-binding proteins for further analysis. Heat map of the differently expressed RBPs was plotted by the online software NetworkAnalyst (16).

Gene Ontology and Kyoto Encyclopedia of Genes and Genomes Enrichment Analyses

To further investigate the potential function and molecular mechanisms involved by these differently expressed RBPs, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted using the clusterProfiler package in R (version. 4.0). The GO terms covered three biological aspects including molecular function (MF), cellular component (CC) and biological process (BP). P values of <0.05 were regarded as statistically significant.

Protein–Protein Interaction Network Construction and Hub Genes Analysis

Protein-Protein interactions (PPI) were assessed using the online STRING database (https://string-db.org/). The protein-protein pairs of differently expressed RBPs meeting the criteria of combined score > 0.4 were selected and displayed by the Cytoscape software (version 3.7.2). Then, cytoHubba, a plug-in of Cytoscape, was used to screen out the gene with the highest degrees of connectivity. The top ten genes were considered the hub genes and hence used in subsequently analyses.

Screening and Analysis of Prognosis-Related RBPs

The UALCAN database (http://ualcan.path.uab.edu/) is an open network resource for data-mining, primarily based on the respective cancer data in TCGA database. Herein, mRNA levels of hub genes in normal and HCC tissues were compared in the UALCAN database. Besides, the correlation between gene-expression levels and tumor grades were further analyzed. The protein levels of the hub genes were also compared between normal and HCC tissues by immunohistochemical image in Human Protein Atlas (HPA) database (http://www.proteinatlas.org). To further explore the prognostic value of hub RBPs, we conducted a survival analysis using Kaplan-Meier plotter (http://www.kmplot.com). Survival curves and corresponding parameters could be obtained at the website. Finally, genetic alteration data of all prognosis-related hub genes were downloaded and visualized in the form of mutation profiles from the cBioPortal (http://www.cbioportal.org).

Construction and Validation of an RNA-Binding Protein-Associated Prognostic Model

Univariate Cox regression analysis was performed using the “survival” package in R software to identify prognostic factors from above prognosis-related RBPs. Variables with P < 0.05 in univariate analysis were further included in the stepwise multivariate analysis. A predictive model was then designed based on the independent prognostic factors. The following formula was used to determine the risk score: Risk score = β1*Exp1+β2*Exp2+βi*Expi (β, regression coefficient; Exp, prognostic factors). Afterwards, we sub-classified patients into low and high risk groups according to the median risk score. The difference of survival status between the two groups was assessed by Kaplan-Meier curves. Meanwhile, the “time ROC” package was used to establish a ROC curve to assess the prognostic value of the model by calculating the area under the curve (AUC). Finally, the prognostic model was externally validated using the ICGC dataset to test its stability.

Establishing a Nomogram for Prognostic Risk Assessment

To ascertain if the constructed model performed was independently of clinical features, multivariate analysis was conducted using the Cox regression model. Then, we established a nomogram integrating the factors with independent prognostic value, which could provide quantitative methods to predict survival probabilities of HCC patients. The prognostic accuracy of the nomogram was further evaluated using the ROC curve analysis and calibration plots.

Results

Differently Expressed RNA-Binding Proteins in Hepatocellular Carcinoma

A flowchart of the study is provided as Supplementary Figure 1. After removing duplicate samples, 78 HCC tissues and 77 adjacent non-tumor tissues were processed for data-mining by GEO2R, and 2,936 differently expressed genes (DEGs) meeting the screening criteria were identified (Figure 1A). We then used Venn diagram to show the intersection between these DEGs and candidate RBP genes catalog. Finally, a total of 92 RBPs were found, among which 27 were down-regulated and 65 were up-regulated (Figure 1B). Heatmap of the top 50 differently expressed RBPs were plotted in Figure 1C.

FIGURE 1
www.frontiersin.org

Figure 1 Differently expressed RBPs in hepatocellular carcinoma. (A) Volcano plots of DEGs from GSE 54236. Red plots represent genes with log|FC| ≥ 0.5 and adjusted P-value ≤ 0.05. (B) Venn diagram of the overlap between the DEGs from GSE 54236 and candidate RBP genes catalog. (C) Heatmap of top 50 differently expressed RBP genes.

Functional Enrichment of Differently Expressed RNA-Binding Proteins

GO enrichment and KEGG pathway analyses were applied to clarify the gene function of differently expressed RBPs and signaling pathways involved. The GO terms were clustered into 3 major GO ontologies: BP, CC and MF. As shown in Figure 2A, differently expressed RBPs were mostly enriched in BP: including ncRNA metabolic process, regulation of mRNA metabolic process and RNA splicing. For CC, DEGs were mostly enriched in cytoplasmic ribonucleoprotein granule and ribonucleoprotein granule. Molecule Function analysis indicated that the DEGs significantly participated in double-stranded RNA binding, mRNA 3’-UTR binding and translation repressor activity. Moreover, for the KEGG pathway analysis, our results revealed that DEGs were mostly enriched in Spliceosome, Ribosome and mRNA surveillance pathway (Figure 2B).

FIGURE 2
www.frontiersin.org

Figure 2 GO, KEGG enrichment analysis and PPI network. (A) GO analysis of differently expressed RBP genes. MF: molecular function; CC: cellular component, BP: biological process; (B) KEGG analysis of differently expressed RBP genes. (C) PPI network of differently expressed RBP genes.

Protein–Protein Interaction Construction and Hub Genes Identification

In vivo, protein–protein interaction is an important way to exert their biological function. A PPI network of differently expressed RBPs was established based on the STRING database and visualized using Cytoscape Software. The network composed of 74 nodes and 214 links, as shown in Figure 2C. Next, applying the cytoHubba plug-in, top ten genes were selected from the PPI network according to their connectivity degrees. These hub genes including POLR2K, HNRNPM, SNRPE, DDX39A, RRS, HNRNPU, BOP1, SNRPB, EZH2, and ILF3 were all up-regulated. The gene description and connectivity degrees of these hub genes were summarized in Table 1.

TABLE 1
www.frontiersin.org

Table 1 The top 10 hub genes with the highest degree of connectivity in the PPI network.

Verification of the Hub Genes

To confirm the results, above hub genes were verified in the UALCAN database including gene expression patterns and correlation with clinicopathological parameters. The results showed that a total of nine hub genes were matched in the UALCAN database and their expression were significant elevated in tumor tissues (Figure 3). Besides, hub gene expression was positively correlated with tumor pathological grade (Figure 4).

FIGURE 3
www.frontiersin.org

Figure 3 mRNA expression of hub genes in HCC tissues and adjacent normal tissues. All these hub genes were highly expressed in tumor tissues (A–I). *** P < 0.001.

FIGURE 4
www.frontiersin.org

Figure 4 The relationship between hub genes expression and tumor grades in HCC. *P < 0.05, **P < 0.01, ***P < 0.001.

After detecting the hub genes expression at mRNA level, we also compared their protein expression in normal and liver cancer tissues using immunohistochemical results obtained from HPA database (Supplementary Figure 2). We found that RRS1, HNRNPU, SNRPB, EZH2, and ILF3 protein expression levels were increased in HCC tissues than normal tissues, whereas changes for HNRNPM and BOP1 were not significant. Additionally, POLR2K and SNRPE were not detected in either tumor or non-tumor tissues.

Prognosis-Related RNA-Binding Protein Selection and Genetic Alteration Analysis

Considering above results, expression of hub RBPs were strongly correlated with the degree of tumor malignancy. Thus, the association of the hub RBPs expression with the prognosis of HCC patients was explored in the Kaplan-Meier Plotter database. Survival data from 364 HCC patients were available for analysis. As shown in Figure 5, higher expression of HNRNPM (HR = 1.64, 95% CI: 1.16–2.32, and P = 0.0047), SNRPE (HR = 1.65, 95% CI: 1.16–2.35, and P = 0.005), RRS1 (HR = 1.57, 95% CI: 1.11–2.22, and P = 0.011), BOP1 (HR = 1.85, 95% CI: 1.31–2.62, and P = 0.00039), SNRPB (HR = 1.61, 95% CI: 1.13–2.28, and P = 0.0076), EZH2 (HR = 2.23, 95% CI: 1.56–3.19, and P = 6.8×10−6), and ILF3 (HR = 1.55, 95% CI: 1.07–2.24, and P = 0.02) were significantly associated with reduced survival period of patients with liver cancer. However, high expression of POLR2K or HNRNPU didn’t show significant prognostic value.

FIGURE 5
www.frontiersin.org

Figure 5 Survival analyses of the hub genes in patients with HCC (Kaplan–Meier Plotter). P < 0.05 was considered statistically significant.

Next, gene mutations of the seven prognosis-related RBPs were analyzed in cBioPortal database. The results showed that, out of the 345 HCC patients, the seven RBP genes showed altered expression patterns in 214 (62%) samples. Moreover, we found that the mutation rate of BOP1 was 38%, ranking first in all prognosis-related RBPs (Supplementary Figure 3).

Construction and Validation of the Prognostic Model

After identifying the seven prognosis-related RBPs, we then tried to evaluate their independent prognostic value as biomarkers for overall survival (OS) of patients with HCC. Cox regression analysis was performed based on expression data of prognosis-related RBP genes. Univariate analysis showed that all seven RBPs were the influencing factors for unfavorable survival of liver cancer patients. In the multivariate analysis, we found that high expression of BOP1 (HR = 1.259, 95%CI: 1.064–1.489, and p = 0.007) and high expression of EZH2 (HR = 1.801, 95%CI: 1.409–2.303, and p < 0.001) were related to poor prognosis (Table 2). Then, the risk score formula based on regression coefficients of the above two genes can be obtained as follow:

TABLE 2
www.frontiersin.org

Table 2 Association between genes expression and overall survival in HCC patients.

Risk score = 0.2304 * BOP1 + 0.5885 * EZH2.

To assess the ability of the model to predict prognosis, survival analysis was conducted. A total of 344 HCC patients were sub-classified into low and high groups based on the medium risk score value. In the TCGA dataset, the OS rate of patients in the high-risk group was markedly lower compared to that of patients in the low-risk group (Figure 6A). Time-dependent ROC curves at 1, 3 and 5 years were plotted, and the AUC at 1, 3 and 5 years survival was 0.744, 0.688 and 0.713, respectively (Figure 6B). Besides, the distribution of patients’ risk score, survival time and survival status were shown in Figures 6C, D. Then, the stability of this model was examined in an ICGC-JP dataset. Consistent with the TCGA dataset, patients in the high-risk group exhibited a substantially worse outcome (Figure 7A). We also applied ROC curve analysis to evaluate the performance of the prognostic model, and the AUC value at 1 and 3 years was 0.751 and 0.738, respectively (Figure 7B). Risk score and survival information were presented in Figures 7C, D. All these results suggested good accuracy and stability of this model. Compared with OS, disease-free survival (DFS) and disease-specific survival (DSS) can reflect clinical benefits more specifically. To examine whether the prognostic model is also related to DFS and DSS, these two prognostic indicators were used as clinical endpoints for survival analysis. Compared with the low-risk group, the DFS was significantly reduced in high-risk patients (Supplementary Figures 4A, B). The area under the curve (AUC) of time-dependent ROC curves at 1, 3, and 5 years was 0.710, 0.582 and 0.747, respectively (Supplementary Figure 4C). Similar conclusions could also be reached for DSS (Supplementary Figures 4D–F).

FIGURE 6
www.frontiersin.org

Figure 6 Determination of risk scores by the prognostic model in the TCGA dataset. (A) Survival analysis of high- and low-risk groups. (B) ROC curves analysis of the prognostic model. (C) The risk score distribution of HCC patients. (D) The survival status of patients in the TCGA dataset.

FIGURE 7
www.frontiersin.org

Figure 7 Determination of risk scores by the prognostic model in the ICGC dataset. (A) Survival analysis of high- and low-risk groups. (B) ROC curves analysis of the prognostic model. (C) The risk score distribution of HCC patients. (D) The survival status of patients in the ICGC dataset.

Clinical Utility of the Established Prognostic Model

Considering the risk score were closely related to the survival of patients, we subsequently analyzed the relationships between the prognostic model and clinical characteristics. As shown in Figure 8, the risk score was remarkably higher in advanced pathological grade (P < 0.001) and late clinical stage (P < 0.001). No significant difference was found in age > 55 versus age ≤ 55 (P = 0.19), or female versus male (P = 0.91).

FIGURE 8
www.frontiersin.org

Figure 8 The clinicopathological significance of the prognostic model. Age (A) and gender (B) were not associated with the risk score. Advanced pathological grade (C) and late clinical stage (D) were found to be associated with a high risk score.

Construction of a Nomogram for Clinical Practice

Further analysis revealed that risk score and pathological stage were independent prognostic factors for OS (Figure 9A). Then, the R software was used to integrate these two variables to construct a nomogram-plot (Figure 9B). By drawing vertical line to the points axis, we could calculate the sum of the scores of all variables, and the corresponding probability is the survival probability of patients. Calibration curves in TCGA dataset suggested that the predicted values of the nomogram matched well with the actual survival probability of the patients (Figure 9C). We also compared the discrimination performance among the clinical stage, risk score and the nomogram. As shown in Figure 9D, the nomogram exhibited the largest AUC value, indicating good clinical application value. Moreover, we also constructed a nomogram for DFS prediction, and confirmed the model with good predictive power (Supplementary Figure 5).

FIGURE 9
www.frontiersin.org

Figure 9 A nomogram for overall survival prediction for HCC patients. (A) Univariate and multivariate Cox analyses for the prognostic model and clinical parameters. (B). A nomogram to predict OS at 1 year, 3 years, and 5 years of patients with HCC. (C). Calibration plots showing the prediction of OS at 1 year, 3 years, and 5 years by the nomogram (D). Comparison of time-dependent ROC curves among nomogram, clinical stage and the prognostic model.

Discussion

HCC is a highly heterogeneous cancer with high mortality and different clinical outcomes. So far, there have been no solidly established prognostic biomarkers for risk prediction in clinical practice. Hence, the search for accurate, reliable and simple prognostic biomarkers is especially significant for HCC patients (17). Being important epigenetic factors involved in post-transcriptional regulation, RBPs participate in the initiation and development of various tumors including HCC, revealing their potential to serve as prognostic biomarkers (1821). In studies by Feng et al. (22), RBM43 was identified as a tumor suppressor by inhibiting carcinogenesis and tumor growth, and its low expression was negatively correlated with HCC prognosis. Similarly, low expression of SORBS2 was regarded as an adverse prognostic marker (23, 24). Moreover, several other studies revealed that overexpression of certain RBPs exhibited positive correlations with the recurrence and poor prognosis of HCC patients (13, 25, 26). Although these studies have explored the possible functions of certain RBPs and confirmed their prognostic value, further study is required to investigate the clinical value of RBPs in HCC, comprehensively and systematically. Herein, we mined expression profiles of RBP from GEO, TCGA and ICGC datasets and aimed to explore the impact of RBP gene expression level on the prognosis of HCC patients.

A total of 92 differently expressed RBP genes between HCC and non-tumor tissues were selected from the GEO dataset, which were mainly enriched in the processes involved in the regulation of RNA. Through the PPI network construction, ten hub genes exhibiting the highest degrees were identified in the network. After validation in the TCGA database, survival analyses, univariate, and multivariate Cox regression analyses were performed to screen out survival-related RBP genes. Based on two survival-related RBP genes, a prognostic model was finally constructed for prediction of clinical outcomes. Patients with higher risk scores showed worse overall survival, which was confirmed in both TCGA and ICGC databases. To specifically reflect clinical benefits, DFS and DSS were additionally used as the clinical endpoints, and this model still exhibited good predictive capability. Furthermore, multivariate analysis revealed that the prognostic model could be used as an independent prognostic biomarker for HCC patients. A nomogram combined pathological stage with prognostic model will provide a more precise individualized prediction.

Two RBP genes identified in this study were previously reported to function as important regulators driving the initiation and progression of diverse tumors. The block of proliferation (BOP1) gene, located on chromosome 8q24.3, belongs to the WD40 protein family (27). As part of the PES1-BOP1-WDR12 (PeBow) complex, the BOP1 gene is mainly involved in 60S ribosome maturation process (28). Abnormal expression of BOP1 modifies various aspects of colorectal cancer and melanoma (29, 30). In colorectal cancer cells, BOP1 was identified as a target gene of the Wnt/β-catenin pathway. Overexpression of BOP1 promoted cell migration, epithelial-mesenchymal-transition and experimental metastasis of CRC cells as a function of its crosstalk with the JNK signaling pathway and downstream genes (29). In melanoma, Gupta et al. reported that BOP1 knockdown played a crucial role in the acquisition of BRAF kinase inhibitors resistance by targeting MAPK phosphatases DUSP4 and DUSP6 (30). Recently, it has been demonstrated that the BOP1 gene was overexpressed in HCC tissues, and showed strong association with microvascular invasion and advanced disease stage. Moreover, evidence from in vitro studies suggested that BOP1 gene exerted oncogenic effects by inducing EMT and actin cytoskeleton remodeling (31). Similar roles of BOP1 gene in HCC progression were also found in the present study. Our results showed that the expression level of BOP1 was significantly higher in HCC tissues compared with paracancerous tissues. Besides, high expression of BOP1 was associated with tumor grade and poor survival, acting as an independent prognostic factor for HCC patients.

The enhancer of zeste homolog2 (EZH2) gene encodes the histone-lysine N-methyltransferase enzyme, participating in the silencing of many tumor suppressor genes (32, 33). As expected, over-expression of EZH2 was found in a variety of malignancies, such as prostate cancers (34), breast cancers (35) and gastric cancers (36). Studies from Xu et al. showed that EZH2 was over-expressed in HCC tissues and cell lines, and miR-101 could target EZH2 to inhibit HCC progression and increase chemotherapeutic treatment sensitivity (37). Interestingly, Hibino et al. found that the inhibition of EZH2 by small molecule agents could also activate tumor-suppressor miRNAs to exert antitumor effects in HepG2 cells (38). In recent years, immune checkpoint inhibitors appear to be a promising approach for HCC treatment. Xiao et al. found epigenetic modifications mediated by EZH2 could also affect immunotherapy effect. Mechanistically, EZH2 could negatively regulate the expression of PD-L1 by regulating the promoter H3K27me3 levels of CD274 (39). Herein, the protein and mRNA expression of EZH2 were found significantly higher in HCC tissues, and mRNA expression of EZH2 was highly correlated with tumor grades and prognosis of HCC patients.

With the development of microarray or RNA sequence techniques, several prediction models have been proposed to facilitate the prediction of long-term outcomes of HCC patients. However, some limitations should be considered and addressed in these studies. For instance, Sun et al. constructed a prognostic signature containing 33 immune gene pairs (40). The model incorporating too many variables was not easy to use, which limited its clinical application. Besides, the robustness of predictive models was not given enough attention in the research of Zheng et al. (41). An external validation in different population was needed to test the reliability of predictive models. Moreover, some predictive models did not include relevant clinical parameters, which to some extent weakened the diagnostic effectiveness of the models (42, 43).

Recently, a similar study has been published. In that study, the investigators constructed a gene signature containing 37 gene pairs to predict the overall survival of HCC patients (44). The gene signature in our study contained only two genes, which seemed to be less comprehensive than the former. However, the gene signature in our study still showed good predictive ability and high stability. In contrast, it could be more easy-to-use and cost-effective in clinical practice. The robustness of this prognostic signature was also validated in the ICGC-JP series from the high incidence area of HCC, further extending its applicability across different race groups. Besides, the prognostic signature was confirmed using different study endpoints including OS, DFS and DSS, making the application more convenient and accurate. On the basis of the model, pathological stage was added to establish a nomogram, further enhancing the performance for the prediction of survival. All these findings may provide a better guidance for clinical decision-making of individual therapy.

Meanwhile, some limitations of this study should also be noted. Although we attempted to perform a comprehensive analysis to conduct the retrospective study, further prospective studies are needed to verify the conclusion. In addition, considering the heterogeneity between study populations, it will be necessary to validate the effectiveness and reliability of the model with more patients’ samples from different centers.

Data Availability Statement

The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found below: https://www.ncbi.nlm.nih.gov/, GSE54236 https://cancergenome.nih.gov/, TCGA-LIHC https://dcc.icgc.org/, ICGC-JP.

Author Contributions

YH, YS, and XZ conceived and designed the experiments. JY, MZ, and GJ collected the data. ST, JL, KS, YL, JY, and SM analyzed the data. ST, JL, KS, and YL drafted the paper. All authors critically reviewed the manuscript. All authors contributed to the article and approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The study was funded by the National Natural Science Foundation of China (nos. 81770569, 81870421, and 81773072), International Cooperation and Exchange of the National Natural Science Foundation of China (no. 81820108005).

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2020.597996/full#supplementary-material

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin (2018) 68:394–424. doi: 10.3322/caac.21492

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Singal AG, Marrero JA. Recent advances in the treatment of hepatocellular carcinoma. Curr Opin gastroenterol (2010) 26:189–95. doi: 10.1097/MOG.0b013e3283383ca5

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Bruix J, Gores GJ, Mazzaferro V. Hepatocellular carcinoma: clinical frontiers and perspectives. Gut (2014) 63:844–55. doi: 10.1136/gutjnl-2013-306627

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Chuang SC, La Vecchia C, Boffetta P. Liver cancer: descriptive epidemiology and risk factors other than HBV and HCV infection. Cancer Lett (2009) 286:9–14. doi: 10.1016/j.canlet.2008.10.040

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Wong CM, Tsang FH, Ng IO. Non-coding RNAs in hepatocellular carcinoma: molecular functions and pathological implications. Nat Rev Gastroenterol hepatol (2018) 15:137–51. doi: 10.1038/nrgastro.2017.169

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Han L, Huang C, Zhang S. The RNA-binding protein SORBS2 suppresses hepatocellular carcinoma tumourigenesis and metastasis by stabilizing RORA mRNA. Liver Int Off J Int Assoc Study Liver (2019) 39:2190–203. doi: 10.1111/liv.14202

CrossRef Full Text | Google Scholar

7. Lan T, Li H, Zhang D, Xu L, Liu H, Hao X, et al. KIAA1429 contributes to liver cancer progression through N6-methyladenosine-dependent post-transcriptional modification of GATA3. Mol Cancer (2019) 18:186. doi: 10.1186/s12943-019-1106-z

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Gerstberger S, Hafner M, Tuschl T. A census of human RNA-binding proteins. Nat Rev Genet (2014) 15:829–45. doi: 10.1038/nrg3813

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Marchese D, de Groot NS, Lorenzo Gotor N, Livi CM, Tartaglia GG. Advances in the characterization of RNA-binding proteins. Wiley Interdiscip Rev RNA (2016) 7:793–810. doi: 10.1002/wrna.1378

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Masuda K, Kuwano Y. Diverse roles of RNA-binding proteins in cancer traits and their implications in gastrointestinal cancers. Wiley Interdiscip Rev RNA (2019) 10:e1520. doi: 10.1002/wrna.1520

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Nishida K, Kuwano Y, Nishikawa T, Masuda K, Rokutan K. RNA Binding Proteins and Genome Integrity. Int J Mol Sci (2017) 18:1341. doi: 10.3390/ijms18071341

CrossRef Full Text | Google Scholar

12. Zhao L, Cao J, Hu K, Wang P, Li G, He X, et al. RNA-binding protein RPS3 contributes to hepatocarcinogenesis by post-transcriptionally up-regulating SIRT1. Nucleic Acids Res (2019) 47:2011–28. doi: 10.1093/nar/gky1209

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Yang D, Jiao Y, Li Y, Fang X. Clinical characteristics and prognostic value of MEX3A mRNA in liver cancer. PeerJ (2020) 8:e8252. doi: 10.7717/peerj.8252

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Li T, Li S, Chen D, Chen B, Yu T, Zhao F, et al. Transcriptomic analyses of RNA-binding proteins reveal eIF3c promotes cell proliferation in hepatocellular carcinoma. Cancer Sci (2017) 108:877–85. doi: 10.1111/cas.13209

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res (2013) 41:D991–5. doi: 10.1093/nar/gks1193

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Xia J, Gill EE, Hancock RE. NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nat Protoc (2015) 10:823–44. doi: 10.1038/nprot.2015.052

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet (London England) (2018) 391:1301–14. doi: 10.1016/s0140-6736(18)30010-2

CrossRef Full Text | Google Scholar

18. Zhao L, Wang W, Huang S, Yang Z, Xu L, Yang Q, et al. The RNA binding protein SORBS2 suppresses metastatic colonization of ovarian cancer by stabilizing tumor-suppressive immunomodulatory transcripts. Genome Biol (2018) 19:35. doi: 10.1186/s13059-018-1412-6

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Hu Z, Dong L, Li S, Li Z, Qiao Y, Li Y, et al. Splicing Regulator p54 /Non-POU Domain-Containing Octamer-Binding Protein Enhances Carcinogenesis Through Oncogenic Isoform Switch of MYC Box-Dependent Interacting Protein 1 in Hepatocellular Carcinoma. Hepatol (Baltimore Md) (2019) 72:548–68. doi: 10.1002/hep.31062

CrossRef Full Text | Google Scholar

20. 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 tumour growth and metastasis. J Clin Invest (2020) 130:3848–64. doi: 10.1172/jci130379

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Correa BR, de Araujo PR, Qiao M, Burns SC, Chen C, Schlegel R, et al. Functional genomics analyses of RNA-binding proteins reveal the splicing regulator SNRPB as an oncogenic candidate in glioblastoma. Genome Biol (2016) 17:125. doi: 10.1186/s13059-016-0990-4

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Feng H, Liu J, Qiu Y, Liu Y, Saiyin H, Liang X, et al. RNA-binding motif protein 43 (RBM43) suppresses hepatocellular carcinoma progression through modulation of cyclin B1 expression. Oncogene (2020) 39:5495–506. doi: 10.1038/s41388-020-1380-7

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Yan B, Peng Z, Xing C. SORBS2, mediated by MEF2D, suppresses the metastasis of human hepatocellular carcinoma by inhibitiing the c-Abl-ERK signaling pathway. Am J Cancer Res (2019) 9:2706–18.

PubMed Abstract | Google Scholar

24. Han L, Huang C, Zhang S. The RNA-binding protein SORBS2 suppresses hepatocellular carcinoma tumourigenesis and metastasis by stabilizing RORA mRNA. Liver Int Off J Int Assoc Study Liver (2019) 39:2190–203. doi: 10.1111/liv.14202

CrossRef Full Text | Google Scholar

25. Jeng Y, Chang C, Hu F, Chou H, Kao H, Wang T, et al. RNA-binding protein insulin-like growth factor II mRNA-binding protein 3 expression promotes tumor invasion and predicts early recurrence and poor prognosis in hepatocellular carcinoma. Hepatol (Baltimore Md) (2008) 48:1118–27. doi: 10.1002/hep.22459

CrossRef Full Text | Google Scholar

26. Zhang K, Pomyen Y, Barry A, Martin S, Khatib S, Knight L, et al. MYCAGO2 Mediates mRNA Stability in Hepatocellular Carcinoma. Mol Cancer Res : MCR (2020) 18:612–22. doi: 10.1158/1541-7786.mcr-19-0805

CrossRef Full Text | Google Scholar

27. Pestov DG, Grzeszkiewicz TM, Lau LF. Isolation of growth suppressors from a cDNA expression library. Oncogene (1998) 17:3187–97. doi: 10.1038/sj.onc.1202260

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Strezoska Z, Pestov DG, Lau LF. Bop1 is a mouse WD40 repeat nucleolar protein involved in 28S and 5. 8S RRNA processing and 60S ribosome biogenesis. Mol Cell Biol (2000) 20:5516–28. doi: 10.1128/mcb.20.15.5516-5528.2000

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Qi J, Yu Y, Akilli Öztürk Ö, Holland JD, Besser D, Fritzmann J, et al. New Wnt/β-catenin target genes promote experimental metastasis and migration of colorectal cancer cells through different signals. Gut (2016) 65:1690–701. doi: 10.1136/gutjnl-2014-307900

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Gupta R, Bugide S, Wang B, Green MR, Johnson DB, Wajapeyee N. Loss of BOP1 confers resistance to BRAF kinase inhibitors in melanoma by activating MAP kinase pathway. Proc Natl Acad Sci USA (2019) 116:4583–91. doi: 10.1073/pnas.1821889116

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Chung K, Cheng I, Ching A, Chu J, Lai P, Wong N. Block of proliferation 1 (BOP1) plays an oncogenic role in hepatocellular carcinoma by promoting epithelial-to-mesenchymal transition. Hepatol (Baltimore Md) (2011) 54:307–18. doi: 10.1002/hep.24372

CrossRef Full Text | Google Scholar

32. Zhao JC, Yu J, Runkle C, Wu L, Hu M, Wu D, et al. Cooperation between Polycomb and androgen receptor during oncogenic transformation. Genome Res (2012) 22:322–31. doi: 10.1101/gr.131508.111

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Yu J, Yu J, Mani RS, Cao Q, Brenner CJ, Cao X, et al. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell (2010) 17:443–54. doi: 10.1016/j.ccr.2010.03.018

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature (2002) 419:624–9. doi: 10.1038/nature01075

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A (2003) 100:11606–11. doi: 10.1073/pnas.1933744100

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Pan YM, Wang CG, Zhu M, Xing R, Cui JT, Li WM, et al. STAT3 signaling drives EZH2 transcriptional activation and mediates poor prognosis in gastric cancer. Mol Cancer (2016) 15:79. doi: 10.1186/s12943-016-0561-z

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Xu L, Beckebaum S, Iacob S, Wu G, Kaiser GM, Radtke A, et al. MicroRNA-101 inhibits human hepatocellular carcinoma progression through EZH2 downregulation and increased cytostatic drug sensitivity. J Hepatol (2014) 60:590–8. doi: 10.1016/j.jhep.2013.10.028

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Hibino S, Saito Y, Muramatsu T, Otani A, Kasai Y, Kimura M, et al. Inhibitors of enhancer of zeste homolog 2 (EZH2) activate tumor-suppressor microRNAs in human cancer cells. Oncogenesis (2014) 3:e104. doi: 10.1038/oncsis.2014.17

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Xiao G, Jin LL, Liu CQ, Wang YC, Meng YM, Zhou ZG, et al. EZH2 negatively regulates PD-L1 expression in hepatocellular carcinoma. J immunother Cancer (2019) 7:300. doi: 10.1186/s40425-019-0784-9

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Sun X, Yu S, Zhang H, Li J, Guo W, Zhang S. A signature of 33 immune-related gene pairs predicts clinical outcome in hepatocellular carcinoma. Cancer Med (2020) 9:2868–78. doi: 10.1002/cam4.2921

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Zheng Z, Wen Y, Nie K, Tang S, Chen X, Lan S, et al. Construction of a 13-microRNA-based signature and prognostic nomogram for predicting overall survival in patients with hepatocellular carcinoma. Hepatol Res Off J Japan Soc Hepatol (2020) 50:1151–63. doi: 10.1111/hepr.13538

CrossRef Full Text | Google Scholar

42. Jiang H, Zhao L, Chen Y, Sun L. A four-long noncoding RNA signature predicts survival of hepatocellular carcinoma patients. J Clin Lab Anal (2020) 34:e23377. doi: 10.1002/jcla.23377

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Du B, Wang F, Jarad B, Wang Z, Zhang Y. A novel signature based on microvascular invasion predicts the recurrence of HCC. J Transl Med (2020) 18:272. doi: 10.1186/s12967-020-02432-7

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Kang C, Jia X, Liu H. Development and validation of a RNA binding protein gene pair-associated prognostic signature for prediction of overall survival in hepatocellular carcinoma. Biomed Eng Online (2020) 19:68. doi: 10.1186/s12938-020-00812-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: prognostic model, survival analysis, hepatocellular carcinoma, RNA-binding proteins, The Cancer Genome Atlas (TCGA)

Citation: Tian S, Liu J, Sun K, Liu Y, Yu J, Ma S, Zhang M, Jia G, Zhou X, Shang Y and Han Y (2021) Systematic Construction and Validation of an RNA-Binding Protein-Associated Model for Prognosis Prediction in Hepatocellular Carcinoma. Front. Oncol. 10:597996. doi: 10.3389/fonc.2020.597996

Received: 23 August 2020; Accepted: 07 December 2020;
Published: 26 January 2021.

Edited by:

Wentao Wang, Sichuan University, China

Reviewed by:

Jianjun Lu, First Affiliated Hospital of Sun Yat-Sen University, China
Dongjun Lee, Pusan National University, South Korea

Copyright © 2021 Tian, Liu, Sun, Liu, Yu, Ma, Zhang, Jia, Zhou, Shang and Han. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xia Zhou, zhouxialyg@163.com; Yulong Shang, shangyul870222@163.com; Ying Han, hanying1@fmmu.edu.cn

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.