SNRPG is required for the proliferation, migration, and EMT of HCC cells, which interacted with SNRPB
Recently, it has been reported that SNRPB could serve as a prognostic predictor for patients with HCC, and promote tumor progression [11]. To explore how SNRPB functions the tumorigenic potential in HCC, we selected several significantly dysregulated genes after SNRPB knockdown from RNA-sequencing data. Meanwhile, we also selected the top co-expressed genes with SNRPB ranked by the sum of their edges’LLS score through the online tool Coexpedia (https://www.coexpedia.org/hs_single.php?gene=SNRPB). As shown in Fig. 1A, the Venn diagram demonstrates the overlaps of the DEGs from RNA-sequence and the top 10 of co-expressed genes, and then six genes (PCNA, UBE2S, SNRPG, MCM2, PRMT1, and RANBP1) were presented in overlaps part. The table in Fig. 1B showed the expression value of these six genes through RNA sequencing, and a PPI network presented the interaction between SNRPB and six selected genes through the online STRING tool (Fig. 1C). Through RT-qPCR analysis, we observed that a most significant decrease in SNRPG expression after SNRPB expression was inhibited in SK-HEP-1 cells (Fig. 1D). Endogenous SNRPG was immunoprecipitated from 293T cell lysates by SNRPB antibody. The Co-IP assay demonstrated that SNRPB could interact with SNRPG (Fig. 1E). Thus, SNRPG was selected for further analysis.
To investigate whether SNRPG plays a key role in the malignant progression of HCC in vitro, we first evaluated SNRPG expression in two HCC cell lines after being transfected with shSNRPG or shCtrl vectors. The protein expression of SNRPG in Hep3B and SK-HEP-1 cells were significantly downregulated after shSNRPG transfection by western blot (Fig. 1F). The epithelial-mesenchymal transition (EMT) was associated with tumor initiation, invasion, metastasis, and resistance to therapy in several human cancers [12]. To explore whether SNRPG participated in HCC metastasis, we next examined the role of SNPRG in the expression of EMT markers in HCC cells. Western blotting indicated that protein expression levels of EMT markers, including epithelial marker (E-cadherin), were upregulated after knockdown of SNRPG, while mesenchymal markers (N-cadherin and Vimentin) were downregulated in two HCC cells (Fig. 1F). As shown in Fig. 1G, SNRPG silence significantly decreased cell growth of Hep3B and SK-HEP-1 cells. Additionally, the flow cytometer assay showed that the cell cycle was markedly interrupted in the G2 phase in the SNRPG knockdown cells (Fig. 1H), which may be the key event in which SNRPG affects the viability of tumor cells. These findings indicated that SNRPG was a tumor promoter, contributing to the malignant phenotypes of HCC cells.
Pathway’s enrichment analysis
Differential pathways between high- and low SNRPB or SNRPG expressions were enriched through GSVA and the enrichment heat map was drawn according to the enrichment fraction (Fig. 2A). GSVA analysis showed differences in KEGG functional enrichment between high- and low- SNRPG expression groups. The same results were found in the high and low SNRPB groups. Furthermore, a total of nine pathways regulated by SNRPG or SNRPB were significantly correlated with each other (Fig. 2B), including the spliceosome, RNA degradation, cell cycle, DNA replication, glycolysis gluconeogenesis, apoptosis, focal adhesion, the MAPK signaling pathway, and mTOR signaling pathway, which were closely related to Wnt signaling pathways in the development and/or progression of HCC [13–15]. Several Wnt/β-catenin targets, including Cyclin D1 and c-Myc, are vital for the growth and metastasis of HCC [16]. Therefore, we then examined the effect of SNRPG knockdown on related proteins of the Wnt/β-catenin signaling pathway and found that the expressions of β-catenin, Cyclin D1 and c-Myc were significantly reduced in Hep3B and SK-HEP-1 cells after the elimination of SNRPG (Fig. 2C). These results suggest that the roles of SNRPG in HCC proliferation and metastasis may be partly mediated by the activation of the Wnt/β-catenin signaling pathway.
SNRPG is regulated by SNRPB through the Wnt/β-catenin signaling pathway in HCC
To validate the involvement of SNRPB in SNRPG-mediated HCC cell proliferation and metastasis, we stably introduced SNRPG or empty vector controls into SNRPB knockout HCC cells. As shown in Fig. 3A, SNRPG overexpression promoted cell proliferation of Hep3B and SK-HEP-1 cells, and forced expression of SNRPG in SNRPB knockdown cells remarkably reversed the proliferation inhibition. These data suggested that SNRPG could partly rescue the biological effects of SNRPB-knockdown on HCC cells.
Given that chemotherapy is currently commonly used to treat HCC, our study attempted to evaluate the response of SNRPB and SNRPG expressions to the chemo drug, XAV939, which was an inhibitor of the Wnt/β-catenin signaling pathway. Wnt signaling is well known to control many aspects of cell behavior in cancer [17]. The IC50 value for each sample was estimated in the TCGA-LIHC dataset based on the predictive model of XAV939. Notably, it was observed that HCC samples with low-SNRPB and SNRPG were more sensitive to XAV939 chemotherapy as shown in Fig. 3B (p < 0.001). Furthermore, SK-HEP-1 cells were treated with XAV939 (50 µmol/L) after different transfections. As shown in Fig. 3C, overexpression of SNRPG upregulated β-catenin, Cyclin D1, and c-Myc, while shSNRPB slightly inhibited the up-regulation of these proteins. Importantly, XAV939, similar to shSNRPB, could also significantly inhibit the Wnt/β-catenin signaling pathway in SK-HEP-1 cells (Fig. 3C). These data indicated that SNRPG might be regulated by SNRPB through the Wnt/β-catenin signaling pathway in HCC.
An aberrantly high expression of SNRPB and SNRPG correlates with worse survival in patients with HCC
Through the detection of SNRPB in HCC tumor tissues and adjacent non-tumor tissues, it could be observed that proportions of the high expression of SNRPB in HCC tumor tissues were higher than that in adjacent non-tumor tissues (Fig. 4A). The SNRPB protein was predominantly nuclear and significantly elevated in HCC tumor tissues compared to non-tumor tissues through IHC staining (Fig. 4B). Furthermore, we confirmed the higher expression of SNRPB in HCC primary tumor compared to para-cancer from the GSE45114 cohort, as well as SNRPB expression in the metastasis tumor was significantly higher than that in the primary tumor (Fig. 4C). These results confirmed that SNRPB participated in the process of HCC metastasis. Baseline clinical data were combined for 70 cases of patients with HCC, the univariate regression analysis showed that SNRPB and several other factors (tumor size, AJCC, and T stage) were independent risk prognostic factors for HCC with HR ≥ 1 (p < 0.05) (Fig. 4D). Kaplan-Meier analysis showed that higher expression of SNRPB indicated the poor OS of HCC patients (p = 0.00094) (Fig. 4E). Moreover, the results in Fig. 4F showed that HCC patients with higher levels of tumor size and AJCC were related to a lower probability of overall survival. The increased levels of the SNRPB protein in four pairs of HCC tumor and non-tumor tissues were also confirmed by western blot analysis (Fig. 4G). Therefore, SNRPB plays a pro-aggressive role during HCC malignant progression.
Similar to the SNRPB expression in HCC tissues and adjacent non-tumor tissues, we found that proportions of the high expression level of SNRPG in HCC tumor tissues were also higher than in adjacent non-tumor tissues (Fig. 5A). Through IHC staining, the SNRPG protein was also predominantly nuclear in HCC tumor tissues (Fig. 5B). Univariate regression analysis showed that SNRPG and several other factors (tumor size, AJCC, and T stage) also independent risk prognostic factors for HCC with HR ≥ 1 (p < 0.05) (Fig. 5C). The Kaplan-Meier analysis showed that higher SNRPG expression and higher tumor size indicated the poor OS for HCC patients (p = 0.029 and p = 0.02) (Fig. 5D). The increased levels of the SNRPG protein in four pairs of tumor and non-tumor tissues were also confirmed by Western blotting (Fig. 5E). Further Pearson correlation coefficient corroborated a positive correlation between SNRPB and SNRPG in HCC tissues (Fig. 5F). These data indicate that SNRPG, similar to SNRPB, was a potential prognostic marker in HCC.
Construction and evaluation of double-gene prognostic signature model
In light of the abnormal expression patterns of SNRPB and SNRPG in HCC, their expression levels (FPKM) were used to build the double gene prognostic signature model (Model). Based on the data of patients with HCC from the TCGA data set and 65 HCC patients collected at our hospital, we applied Kaplan-Meier survival and ROC curves to identify the prognostic functions of this model for HCC. As shown in Fig. 6A, the K-M survival curves confirmed that lower levels of the model were associated with better OS (p = 0.022), and the ROC curves showed that this model harbored a promising ability to predict OS for HCC patients in the TCGA data set (training set) (1-year AUC = 0.655, 2-year AUC = 0.637, 3-year AUC = 0.620, 5-year OS = 0.669). Similarly, in the validated set containing 65 HCC patients, a higher level of the model was associated with worse OS (p = 0.0049) and this model also had a promising ability to predict OS for HCC (20 months AUC = 0.662, 30 months AUC = 0.669, and 50 months AUC = 0.673; Fig. 6B). To create a clinically applicable quantitative tool to predict OS for HCC patients, we established a nomogram based on the model, age, tumor stage and gender (Fig. 6C). The OS of patients with HCC was calculated, and then the 1-year, 2-year, 3-year, and 5-year OS were predicted. Calibration plots of 1-, 2-, 3- and 5-year OS indicated that the nomogram was of excellent predictive effect (Figs. 6D), implying that it was incredibly valuable to integrate the double-gene prognostic signature model into the clinical prognostic factors for HCC.
Pan-cancer analysis of SNRPB and SNRPG and the sensitivity of their expression to immunotherapies in HCC
To further explore the possible roles of SNRPB and SNRPG in carcinogenesis, we first analyze the level of their double-gene prognostic signature model in 24 types of human cancer in the TCGA database. As shown in Fig. 7A, compared to normal samples, the model levels were significantly increased in most of the 24 cancer types, including UCEC, THCA, STAD, READ, PRAD, LUSC, LUAD, LIHC, KIRP, KIRC, HNSC, GBM, ESCA, COAD, CHOL, CESC, BRCA, and BLCA. These data indicated that SNRPB and SNRPG could function as crucial regulators in carcinogenesis, as candidate genes for developing independent prognostic factors for these cancer-type tumors.
In addition, tissue-/organ-specific SNRPB and SNRPG were identified by BioGPS. As shown in Fig. 7B, it was observed that SNRPB and SNRPG were both specifically enriched in the immune system, especially in the CD34 + cells, CD105 + Endothelial, and 721_B_lymphoblasts. Meanwhile, SNRPB was specifically enriched in leukemia lymphoblastic (MOLT-4), lymphoma_burkitts (Daudi) and lymphoma_burkitts (Raji), while SNRPG was specifically enriched in bronchial epithelial cells and chronic myelogenous. These data suggested that SNRPB and SNRPG could be involved in the progression of tumors or immune-related diseases by regulating the immune response. Subsequent correlation analysis also confirmed that their double-gene signature model was marked positively correlated with regulatory T cells (Tregs), follicular helper T cells, and memory B cells in the immune system (Fig. 7C), indicating that SNRPB and SNRPG were inextricably linked to the immune process. Additionally, the double-gene prognostic signature model was found to be associated with spliceosome, RNA degradation, cell cycle, DNA replication, glycolysis gluconeogenesis, apoptosis, focal adhesion, MAPK signaling pathway and mTOR signaling pathway in most cancers, including LIHC (Fig. 7D). These enriched signaling pathways are indeed closely related to SNRPB and SNRPG (Fig. 2B), as well as the Wnt signaling pathway, echoing the previously confirmed Wnt/β-Catenin signaling pathway downstream-regulated by the SNRPG in HCC.
To predict the response of SNRPB and SNRPG to ICB immune checkpoint blockade (ICB), we developed TIDE, a computational method to model two primary mechanisms of tumor immune evasion: the induction of T cell dysfunction in tumors with high cytotoxic T lymphocytes (CTL) and the prevention of T cell infiltration in tumors with low CTL level. The findings showed that the TIDE scores were higher in the high SNRPB and high-SNRPG groups than that in the low SNRPB and low-SNRPG groups (P < 0.001) (Fig. 7E), indicating that HCC patients with high-SNRPB and SNRPG were more likely to respond worse to immunotherapy. Therefore, patients with low-SNRPB and SNRPG may benefit more from immunotherapy.