Comprehensive analysis of the expression and prognostic value for SNRP members in hepatocellular carcinoma

DOI: https://doi.org/10.21203/rs.3.rs-633291/v2

Abstract

Background

Heterogeneity and epigenetic modifications lead to differences among treatment strategies, management and prognosis in hepatocellular carcinomas. The family of small nuclear ribonucleoprotein polypeptides (SNRPs) plays a crucial role in tumorigenesis and progression. However, the expression profile and prognostic impact of these family members are not clear. Here, we discuss the expression levels and prognosis of SNRPs. family members

Methods

We compared the transcript levels of each SNRPs member in pan-cancerous tissues by ONCOMINE and further analyzed the expression levels and tumor staging of these markers in hepatocellular carcinoma using UALCAN and GEPIA online databases, while assessing the prognostic value of their mRNA expression and performing functional enrichment analysis by Metascape software using Kaplan–Meier plotter database.

Results

These results showed that mRNA levels of each member of SNRP (B, D1, D2, D3, E, F, G) were significantly upregulated in hepatocellular carcinoma compared to normal tissue and were more highly expressed in patients with advanced hepatocellular carcinoma. mRNA expression of SNRPB, SNRPD1 and SNRPG was associated with poorer overall survival (OS), recurrence-free survival (RFS) and progression-free survival (PFS), which was considered to be statistically significant.

Conclusion

We systematically analyzed the mRNA expression and prognostic significance of each member of SNRPs in HCC and demonstrated the correlation, interaction network, gene alteration and functional enrichment among SNRPs members. Our data suggest that SNRPs members as oncogenes may be a potential indicator of HCC.

Background

Hepatocellular carcinoma (HCC) is the most common of primary liver cancer, remains a high mortality cancer globally [1]. HCC, which has the highest incidence in China compared to other countries, is now the third leading cause of cancer-related deaths and the fourth most common cancer in China in 2018 [2, 3]. Unlike other malignancies, obvious heterogeneity greatly affects the treatment and prognosis of HCC. More potential indicators need to be identified in the overall management of tumors in clinical practice.

The splicing process is accurately ensured by spliceosomes for stability and normalization [4]. Smith (Sm) proteins play a decisive role in maintaining the integrity of small nuclear ribonucleic acid (snRNA) to avoid nucleases and the downstream RNA processing steps [5]. The formation of heterodimeric (SmD1-SmD2 and SmB-SmD3) or heterotrimeric (SmE-SmF-SmG) subcomplexes is one of the important mechanisms of Sm proteins [6]. The small nuclear ribonucleoprotein polypeptide (SNRP) B, D1, D2, D3, E, F, G genes are core components of the spliceosomal small nuclear ribonucleoproteins (snRNPs), forming a 7-membered ring/Sm-core-complex that is precursors to major and minor spliceosome [6] to ensure RNA stability [5]. These complementary roles of SNRP members tumorigenesis and metastasis roles [7] have attracted value attention.

The increasing evidence showed the indispensable role of the splicing components in the initiation, angiogenesis, apoptosis, and invasion in cancers [710]. Relevant researches [1115] have reported the differences and prognostic value of single SNRP members in different cancer types. The function of SNRPB as an oncogene served as a potential prognostic factor for HCC [11]. Another study [12] displayed the mRNA expression of SNRPB may be an effective therapeutic target for cervical cancer by interfering with alterations in the p53 pathway. In addition, high levels of SNRPD1 are considered as predictive biomarkers of tumorigenesis and poor prognosis in lung adenocarcinoma and ovarian cancers [13, 14]. Even, siRNA deprivation of SNRPE or SNRPD1 drives cell death through autophagy, resulting in a marked reduction in cell viability in breast, lung, and melanoma cancer cell lines [15]. In summary, current studies [1115] described the expression of individual members of the SNRPs family in a variety of tumors; however, few studies have focused on the expression and prognostic value of the Sm core complex family (B, D1, D2, D3, E, F and G) in HCC patients.

In our study, we performed a comprehensive analysis of the expression and prognosis of core family members of seven SNRPs in HCC patients. In addition, we analyzed the interaction network, genetic alterations, and functional enrichment based on multiple datasets.

Methods

The study has been permitted by the Institutional Review Board of Peking University International Hospital. All methods were carried out in accordance with relevant guidelines and regulations. And there is not direct human participation in these databases.

ONCOMINE Database

ONCOMINE (http://oncomine.org.cutestat.com/) is a publicly accessible online cancer database to compute gene expression signatures, clusters and gene-set modules, automatically extracting biological information from datasets [16]. We analyzed the transcriptional levels of each SNRP member and compared them among these members for patients with HCC. Then we used Duncan methods by one-way analysis of variance (ANOVA) (IBM SPSS Statistics v. 21) based on the best gene rank percentile to identify significant differences.

UALCAN

UALCAN (http://ualcan.path.uab.edu/) is a user-friendly, and interactive web resource for providing genes or miRNA analysis based on TCGA from 31 cancer types [17]. In our study, it was utilized to analyze the expression levels of tumor and normal tissues. Wilcoxon rank sum test was used to determine whether a significant p value or not. The multiple comparisons by Duncan according to the high expression of SNRP members in primary tissues were described. The cross-correlation coefficients among different genes were calculated by “ggstatsplot” in R version 3.6.1 (http://www.r-pro gect.org/) using spearman correlation analysis.

GEPIA

Gene Expression Profiling Interactive Analysis (GEPIA) (http://gepia.cancer-pku.cn/), is a newly web server to offer customizable functions such as tumor/normal differential expression, profiling according to cancer types or pathological stages, survival analysis, similar gene detection, correlation analysis, and dimensionality reduction analysis based on TCGA and Genotype-tissue Expression (GTE) data [18]. The expression of SNRP members and the tumor stages of HCC were analyzed in this study. The method using pathological stage as variable to calculate differential expression analysis is one-way ANOVA [18].

Kaplan–Meier (KM) Plotter

The Kaplan-Meier Plotter (http://kmplot.com/analysis/) includes the data of patients on survival of 21 cancer types [19]. The prognostic value of mRNA expression was evaluated using the KM plotter with the hazard ratio (HR) with 95% confidence intervals (CI), log rank p-value and median overall survival (OS), relapse-free survival (RFS), progress-free survival (PFS) in lower and upper groups. We split patients by auto select best cutoff [20]. The use of false discovery rate (FDR) control of multiple testing in each member survival analysis can provide a adjust p value for drawing conclusions about statistical significance. It was considered to be significant when a adjust p value was less than 0.05. Multivariable Cox regression analysis was conducted to determine the independent predictors of OS, PFS or RFS by using R version 3.6.1.

cBioPortal

cBioPortal (http://www.cbioportal.org/) is an open web tool for interactive exploration of multiple cancer genomic datasets [21]. It was assessed in terms of genetic alteration frequency, the association between alterations, and survival outcome. The survival data were also recorded with respect of OS, PFS, and RFS (numbers of total and events, median survival, and p value) of each member.

Gene MANIA and STRING

Gene MANIA (http://genemania.org) is an online tool for predicting genes and gene sets [22]. It covered 2277 association networks containing 597 million interactions mapped to 163000 genes from 9 organisms [22]. In this study, Gene MANIA was used to describe the genes network of SNRP members and neighbouring genes. STRING (https://cn.string-db.org/) is a database that analyzed protein -protein interactions (PPI) networks [23]. It was applied to perform reciprocities among the PPI networks of co-expressed genes, and the species were set to Homo sapiens. The relations of expression level about the gene and protein by Cytoscape were identified. Herein, we showed 26 related genes, 47 related nodes, and 927 edges by STRING tool.

Metascape

Metascape (http://metascape.org), is an online website focusing on enrichments pathway analysis [24]. In this study, the pathways and enrichments were analyzed by Metascape.

Results

Differential expression of SNRP members in patients with HCC

Firstly, it was determined that genes for SNRP members are located on definite genomic sites [25,26] (Table 1). We analyzed the transcript levels of the entire cohort of 1858 assays in various cancer types using the ONCOMINE database [16] (Fig. 1). The results showed that significant unique analyses were found in the SNRPB (26 tests), SNRPD1 (32 tests), SNRPD2 (22 tests), SNRPD3 (12 tests), SNRPE (39 tests), SNRPF (15 tests) and SNRPG (23 tests) groups, respectively. SNRPB was significantly elevated in tumor tissues, especially in bladder, cervical, colorectal, gastric, head and neck, kidney, liver and breast cancers, with 2,136 samples. In addition, the levels of each SNRP member were high in patients with gastric and colorectal cancers. Compared with other cancers, liver cancer had the large sample size and strong mRNA expression of SNRPB, SNRPD1, SNRPD2, and SNRPE. In contrast, high levels of SNRPD3 are rarely found in tumor tissue. 

The results of ONCOMINE [16] described that the transcript levels of SNRPB, SNRPD1, and SNRPD2 were significantly elevated in HCC tissues [25,27]. But SNRPD3, SNRPF, and SNRPG had no data set to study. For SNRPE mRNA levels, it was upregulated in all three datasets of the TCGA database [24-27]. It was summarized in Figure 1 and Table 2. We also compared the transcript levels of SNRPs between HCC and normal tissues by using UALCAN [17] (Fig. 2a-2g). We found that SNRPB, D1, D2, D3, E, F, and G were all upregulated in tumor tissues. Besides, we analyzed the correlation among different genes in HCC tissues and determined that SNRPB and SNRPD1 had the highest correlation (Fig. 2h). In conclusion, our results showed that transcriptional expression of SNRPB, SNRPD1, SNRPD2, SNRPD3, SNRPE, SNRPF, and SNRPG was overexpressed in HCC patients. Table 3 shows multiple comparisons of significant unique analyses or median expression in HCC patients using ONCOMINE and UALCAN servers. SNRPB, SNRPD2, SNRPD3 and SNRPF; SNRPD1 and SNRPD2; SNRPD1, SNRPE and SNRPG were analyzed by best gene ranking percentile, with no differences. In addition, there was no significance between SNRPB and SNRPE; SNRPD1, SNRPD2, SNRPD3, SNRPF, and SNRPG by median expression.

Correlation between mRNA expression and tumor stages of SNRP members

We used GEPIA [18] to analyze the correlation between mRNA expression and cancer stage in patients treated with HCC. Although there were significant differences between SNRPB, D1, D2, D3, F and G groups at stages I, II, III and IV, there were no differences between SNRPE groups and tumor stage (Fig. 3). That is, the mRNA expression of SNRPB, D1, D2, D3, F and G had a significant relationship with the cancer stage of the patients and appeared higher in advanced stage cancers.

Prognostic value of SNRP members in patients undergoing HCC

Prognostic significance of mRNA expression, including OS, PFS, and RFS was under observation. It could be found that patients were classified as low (black) and high (red) risk based on their respective OS thresholds (Fig. 4). High levels of SNRPB, SNRPD1, SNRPE and SNRPG mRNA levels suggest a trend towards worse OS, but without significant differences. The cutoff values distinguishing between the high and low groups based on automatic selection of the best cutoff [20], can be seen in Fig. S1.

Increases in SNRPB, SNRPD1, SNRPD2, and SNRPG were associated with poor PFS (Fig. S2). The relevant cutoff values can be seen in Fig. S4. Furthermore, high mRNA levels of SNRPB SNRPD1, SNRPD2, SNRPE, and SNRPG led to shorter RFS (Fig. S5), while no similar findings were found for SNRPD3, and SNRPDF.  The cutoff value for each member can be showed in Fig. S7. 

There are several known prognostic factors for HCC, such as age, gender, WHO grade, p_T stage, and p_TNM stage. It was necessary to examine whether each member could independently predict prognosis. Univariate Cox analysis presented seven genes were positively associated with survival prognosis (Fig. 5, S3, S6). Moreover, p_T and p_TNM stages were also significantly related to OS (Fig. 5a), PFS (Fig. S3a), and RFS (Fig. S6a). Subsequent multivariate Cox regression analysis indicated that SNRPD1 was significantly correlated with OS (Fig. 5b), but failed to be an independent prognostic factor for PFS (Fig. S3b) or RFS (Fig. S6b). In conclusion, high levels of SNRPB, SNRPD1 and SNRPG were associated with prognosis of OS, PFS and RFS. Details are shown in Table 4.

Genetic alterations and correlations of SNRP members

 We analyzed genatic mutations and interactions in HCC patients by cBioPortal [21] (INSERM Cancer Cell 2014 dataset, MSK Clin Cancer Res 2018 dataset, INSERM Nat Genet 2015 dataset, MSK PLOS One 2018, AMC Hepatology 2014 dataset, RIKEN Nat Genet 2012 dataset, TCGA Firehose Legacy dataset, TCGA PanCancer Atlas dataset). The results (TCGA PanCancer Atlas dataset) illustrated that the percentages of gene alterations were 0.27% mutations (1/372), 0.53% deep deletions (2/372), 7.53% amplifications (28/372) in all SNRP members, respectively (Fig. 6a). The frequency of alterations in SNRPs b was analyzed by using the AMC Hepatology 2014 dataset, TCGA Firehose Legacy dataset, and TCGA PanCancer Atlas dataset (SNRPB, 0.5%; SNRPD1, 0.3%; SNRPD2, 0.8%; SNRPD3, 0.3%; SNRPE, 5%; SNRPF, 0.2%; SNRPG, 0.2%) (Fig. 6b). Then, we further presented the survival results based on genetic alterations. Unfortunately, we did not find significance among genetic alterations, OS or PFS, respectively (p = 0.792, 0.662, Fig. 6c, 6e). However, disease-free survival (DFS) was significant in the genetically altered and unaltered groups (Fig. 6d). The reasons why genetic alterations in SNRPs and prognosis did not seem to be associated with prognosis might be related to the context of the study in the database, the material methodology and the small sample size. Table 5 summarizes the OS or DFS data for the genetically altered and unaltered groups, including the total number and number of events. Only the DFS data for SNRPE had a P value <0.05.

Functional enrichment analysis of SNRP members 

To understand the function of SNRP members and their neighboring proteins, we used GO and KEGG pathways by Metascape [24]. The result indicated two major GO-biological processes (Fig. 7a), spliceosomal snRNP assembly (GO:0000387) and spliceosomal complex assembly (GO:0000245). U1 snRNP binding (GO:1990446) was only associated with the molecular function (Fig. 7b). The top 4 GO enrichments (Fig. 7c) were cellular components: methylosome, plCIn-Sm protein complex, U5 snRNP, and U7 snRNP. The top 2 KEGG enrichments (Fig. 7d) were structural complexes: Sm core complex; pathway: systemic lupus erythematosus. 

Interaction of correlated genes and proteins of SNRP members 

We briefly illustrated the correlation of SNRP members at the genatic level by Gene MANIA online tool [22] (Fig. S8a). The results demonstrated SNRP members share genetic thresholds very closely. Markedly, relationships were found among SNRP members regarding the PPI network. We used STRING [23] to determine the correlation of SNRP members at the protein expression level (Figure S8b).

Discussion

A growing number of studies have illustrated that SNRP members are upregulated in various types of cancer and play a vital role in cancer initiation and progression [7]. Recent studies [13, 14] have shown that SNRPD1 is a predictive biomarker for tumorigenesis and poor prognosis in lung and ovarian cancers. In addition, siRNA depletion of SNRPE, D1 led to a reduction of cell viability in breast, lung, and melanoma cancer cell lines [15]. However, since SNRPs are the major spliceosomal precursors of the Sm core complex [6], we need to study the complex as an integrator to explore the different roles in HCC tissues. We hypothesized that SNRP members might act as onco-promoters to affect the prognosis for HCC patients. Therefore, we performed a systematic analysis of the transcript levels and prognostic value of SNRP members in HCC.

We discussed that the mRNA levels of SNRPB, SNRPD1, SNRPD2, and SNRPE were upregulated in HCC tissues compared to normal tissues and illustrated high levels of SNRPD3, F and G did not present a significant disadvantage by ONCOMINE [16]. However, the high levels of expression of SNRPB, D1, D2, D3, E, F, G were found to be present in malignant tumors compared to normal tissues using UALCAN [17]. These two inconsistent results may be due to the diversity of the background and materials of such abundant researches.

The protein encoded by SNRPB is one of nuclear proteins found in U1, U2, U4, U6, and U5 snRNPs which affected pre-mRNA splicing and it may play an important role in snRNP combination [25]. Peng NF and his colleagues [11] showed that SNRPB expression was increased in HCC tissues. In our study, we found that SNRPB expression was significantly elevated in patients with advanced cancer, which is similar to the findings of Peng et al. The gene of SNRPD1 encodes for snRNP [25]. Studies [13, 14] reported the use of free-scale gene co-expression networks to assess the relationship between multiple gene datasets and clinical characteristics of patients, followed by confirmation of predictors by weighted gene co-expression network analysis (WGCNA). The mentioned studies indicated that the mRNA expression of SNRPD1 and its encoded protein were highly specific and sensitive for identifying tumor lesions as one of the predictive biomarkers of tumorigenesis and poor prognosis. In our report, we found mRNA expression of SNRPD1 was upregulated in HCC tissues and led to shorter OS, RFS and PFS, and these results were similar to previous studies. Furthermore, SNRPD1 was reported systematically for the first time in HCC patients. The protein encoded by SNRPD2 and SNRPD3 also belonged to the snRNP core protein [27]. It was shown to be involved in pre-mRNA splicing and snRNP biogenesis. There were few studies on SNRPD2 and SNRPD3 because SNRPD1-SNRPD2 or SNRPB-SNRPD3 preferentially form heterodimeric subcomplexes before forming Smcomplex [6]. We revealed that mRNA expression of SNRPD2 and D3 was up-regulated in HCC and correlated with cancer staging. However, there was no correlation between abnormal levels of SNRPD2 and SNRPD3. This suggested that SNRPD2 and D3 were at the high levels in tumor tissues, but may not be suitable as potential prognostic indicators. As with the heterodimeric subcomplexes of SNRPD2 and SNRPD3, SNRPE, SNRPF and SNRPG could form heterotrimeric subcomplexes that cooperate with other SNRP members to form 7- member ring structure/complex and participate in the splicing process [6]. The current study [15] assumed that SNRPE knockdown obviously led to reduced expression in mTOR pathway and protein levels, which partly explained the SNRPE-based autophagy phenomenon. According to Blijlevens and co-workers [28], high levels of SNRPG protein in v various types of cancer interact positively with cancer initiation, progression and metastasis. The expression of SNRPG in different cancers can be explained by high levels of protein, the mis-localization of unassembled or misassembled proteins [29]. Thus, SNRPG might contribute to the initiation and progression of different cancers [3034]. We found that SNRPE was highly expressed in tumor tissues by using ONCOMINE [16], but the results of SNRPF and SNRPG groups were not similar in this study. In addition, the mRNA of SNRPE did not correlate with cancer stage and PFS. The different results compared to previous studies might be the small sample sizes or different cancer types.

In the study, we found that Gene MANIA [22] and STRING [23] analysis revealed close co-expression between SNRP members at the gene level, while co-expression at the protein level was compactly correlated.

To investigate the correlation of gene alterations, we revealed the frequency of gene alterations in SNRPs by using cBioPortal [21]. We studied the functional enrichment of the SNRPs by Metascape [24]. Our results indicated that SNRPs members are involved in functions that may include methylosome, U1, U5, U7 snRNP binding, Sm core complex, etc., which have been studied as involved in cell cycle, signal transduction, angiogenesis, apoptosis and invasion [8–10,35−40]. The spliceosome complex is formed by snRNP [41, 42]. Each snRNP (U1, U2, U4, U6, and U5) includes an snRNA integrated with a set of Sm core complex. The Sm core complexes (B, D1, D2, D3, E, F and G) form a 7-ring core structure/complex to encapsulate RNA. All SNRP proteins have conserved Sm structural domains to help form the Sm core of the snRNPs [5, 43], thereby determining pre-mRNA processing [44]. However, further work is needed to understand the role and function of SNRP members.

There were some limitations in our research. First, all data were analyzed in our study from various online database tools, which c may be derived from various research contexts, bases and samples, and therefore further studies in larger samples are needed to demonstrate these. Then, no biological experiments, clinical specimens and cases were performed to validate the results. Next, in vitro and in vivo studies will be performed and may provide some further conclusions.

Conclusion

In this study, we systematically analyzed the mRNA expression and prognostic significance of SNRP members in HCC. In addition, we presented the correlations of co-expression and interaction networks, genetic alterations and function enrichment of SNRP members. Expression of SNRPB, SNRPD1, SNRPD2, SNRPD3, SNRPE, SNRPF, and SNRPG was upregulated in tumor tissues compared to normal tissues, and high levels of SNRPB, SNRPD1 and SNRPG resulted in poorer OS, RFS and PFS. In conclusion, SNRPB, SNRPD1, and SNRPG could act as the gene promoters and novel prognostic biomarkers for HCC.

Declarations

Ethics approval and consent to participate: All ethical approval, guidelines, and informed consent are available in each article, which is published and searchable in a public database. All methods were carried out in accordance with relevant guidelines and regulations. And there is not direct human participation in these databases.

Consent for publication: Not Applicable.

Availability of data and materials: Oncomine (http://oncomine.org.cutestat.com/); UALCAN (http://ualcan.path.uab.edu/); Gene Expression Profiling Interactive Analysis (GEPIA) (http://gepia.cancer-pku.cn/); The Kaplan-Meier (KM) Plotter (http://kmplot.com/analysis/); cBioPortal for Cancer Genomics (http://www.cbioportal.org/); GeneMANIA (http://genemania.org); STRING (https://cn.string-db.org/); Metascape (http://metascape.org);

Competing interests: All authors have completed the ICMJE uniform disclosure form. The authors have no conflicts of interest to declare.

Funding: Not applicable.

Authors' contribution:

(I) Conception and design: Ziwei Guo
(II) Administrative support: Jun Liang
(III) Provision of study materials or patients: Ziwei Guo, Chuanhao Tang
(IV) Collection and assembly of data: Ziwei Guo, Chuanhao Tang
(V) Data analysis and interpretation: Ziwei Guo, Chuanhao Tang
(VI) Manuscript writing: All authors
(VII) Final approval of manuscript: All authors

Acknowledgements: A preprint has previously been published [45].

Supplementary description: In supplementary materials, we split patients by auto select best cutoff for OS, PFS, and RFS in Fig. S1, S4, S7, and the prognostic value of PFS and RFS were showed in Fig S2, S5, the subgroup analyses shown in Fig S3, S6. Interaction network of SNRPs at the gene and protein levels in HCC patients in Fig. S8.

References

  1. Justin Hartke, Matthew Johnson, Marwan Ghabril. The diagnosis and treatment of hepatocellular carcinoma. Semin Diagn Pathol. 2017; 34: 153-9.
  2. Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics? Cancer Commun. 2019; 39: 22.
  3. Michael C Wallace, David Preen, Gary P Jeff, Leon Adams. The evolving epidemiology of hepatocellular carcinoma: a global perspective. Expert Rev Gastroenterol Hepatol. 2015; 9: 765-79.
  4. Lerner MR, Boyle JA, Mount SM, Wolin SL, Steitz JA. Are snRNPs involved in splicing? Nature. 1980; 283: 220-4.
  5. Salgado-Garrido J, Bragado-Nilsson E, Kandels-Lewis S, Seraphin B. Sm and Sm-like proteins assemble in two related complexes of deep evolutionary origin. EMBO J. 1999; 18: 3451-62.
  6. A. Gregory Matera, Zefeng Wang. A day in the life of the spliceosome. Nat Rev Mol Cell Biol. 2014; 15: 108-21.
  7. Lloyd Mabonga, Abidemi Paul Kappo. The oncogenic potential of small nuclear ribonucleoprotein polypeptide G: a comprehensive and perspective view. Am J Transl Res. 2019; 11: 6702-16.
  8. Fackenthal JD, Godley LA. Aberrant RNA splicing and its functional consequences in cancer cells. Dis Model Mech. 2008; 1: 37-42.
  9. Pajares MJ, Ezponda T, Catena R, Calvo A, Pio R, Montuenga LM. Alternative splicing: an emerging topic in molecular and clinical oncology. Lancet Oncol. 2007; 8: 349-57.
  10. Skotheim RI, Nees M. Alternative splicing in cancer: noise, functional, or systematic? Int J Biochem Cell Biol. 2007; 39: 1432-49.
  11. Peng NF, Li J, He JR, Shi XM, Huang H, Mo YS, et al. c‐Myc‐mediated SNRPB upregulation functions as an oncogene in hepatocellular carcinoma. Cell Biol. 2020; 44: 1103-11.
  12. Lei Z, Zhang XZ, Sun ZQ. SNRPB promotes cervical cancer progression through repressing p53 expression. Biomedicine & Pharmacotherapy. 2020: 109948.
  13. Bao MY, Zhang LH, Hu YQ. Novel gene signatures for prognosis prediction in ovarian cancer. J Cell Mol Med. 2020;24: 9972-84.
  14. Yi M, Li TY, Qin S, Yu SN, Chu Q, Li A, et al. Identifying Tumorigenesis and Prognosis-Related Genes of Lung Adenocarcinoma: Based on Weighted Gene Coexpression Network Analysis. BioMed Research International. 2020: 4169691.
  15. Virginie Quidville, Samar Alsafadi, Aïcha Goubar, Fred eric Commo, Veronique Scott, Catherine Pioche-Durieu, et al. Targeting the Deregulated Spliceosome Core Machinery in Cancer Cells Triggers mTOR Blockade and Autophagy. Cancer Research. 2013; 73: 2247-58.
  16. Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, et al. ONCOMINE: a cancer microarray database and integrated data-mining platform, Neoplasia. 2004; 6: 1-6.
  17. Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Rodriguez IP, Chakravarthi BVSK, et al. UALCAN: A portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 2017; 19: 649-58.
  18. Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses, Nucleic Acids Res. 2017; 45: 98-102.
  19. Menyhart O, Nagy A, Gyorffy B. Determining consistent prognostic biomarkers of overall survival and vascular invasion in hepatocellular carcinoma. R Soc Open Sci. 2018; 5: 181006.
  20. Lánczky A, Győrffy B. Web-Based Survival Analysis Tool Tailored for Medical Research (KMplot): Development and Implementation. J Med Internet Res. 2021; 23(7): e27633.
  21. G. Jianjiong, A. Bülent Arman, D. Ugur, D. Gideon, G. Benjamin, S.S. Onur, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013; 6: l1.
  22. S. Mostafavi, D. Ray, D. Wardefarley, C. Grouios, Q. Morris. GeneMANIA: a realtime multiple association network integration algorithm for predicting gene function. Genome Biol. 2008; 9: 1-15.
  23. D. Szklarczyk, J.H. Morris, H. Cook, M. Kuhn, S. Wyder, M. Simonovic, et al. The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible. Nucleic Acids Res. 2017; 45: 362-8.
  24. D. Warde-Farley, S.L. Donaldson, O. Comes, K. Zuberi, R. Badrawi, P. Chao, et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010; 38: 214-20.
  25. Roessler S, Jia HL, Budhu A, Forgues M, Je QH, Lee JS, et al. A Unique Metastasis Gene Signature Enables Prediction of Tumor Relapse in Early-Stage Hepatocellular Carcinoma Patients. Cancer Res. 2010; 70: 10202-12.
  26. Chen X, Cheung ST, So S, Fan ST, Barry C, Higgins J, et al. Gene expression patterns in human liver cancers. Mol Biol Cell. 2002; 13: 1929-39.
  27. Roessler E, Muenke M. The molecular genetics of holoprosencephaly. Am J Med Genet C Semin Med Genet. 2010;154C:52-61.
  28. Blijlevens M, Meulen-Muileman IH, Menezes RX, Smit EF and van Beusechem VW. Highthroughput RNAi screening reveals cancer-selective lethal targets in the RNA spliceosome. Oncogene. 2019; 38: 4142-53.
  29. Prusty AB, Meduri R, Prusty BK, Vanselow J, Schlosser A and Fischer U. Impaired spliceosomal UsnRNP assembly leads to Sm mRNA down-regulation and Sm protein degradation. J Cell Biol. 2017; 216: 2391-407.
  30. Schwer B, Kruchten J and Shuman S. Structure-function analysis and genetic interactions of the SmG, SmE, and SmF subunits of the yeast Sm protein ring. RNA. 2016; 22: 1320-8.
  31. Conte N, Charafe-Jauffret E, Delaval B, Adéla-ïde J, Ginestier C, Geneix J, et al. Carcinogenesis and translational controls: TACC1 is down-regulated in human cancers and associates with mRNA regulators. Oncogene. 2002; 21:5 619-30.
  32. Shi Y, Di Giammartino DC, Taylor D, Sarkeshik A, Rice WJ, Yates JR 3rd, et al. Molecular architecture of the human premRNA 3’-processing complex. Mol Cell. 2009; 33: 365-76.
  33. Yoshitake Y, Nakatsura T, Monji M, Senju S, Matsuyoshi H, Tsukamoto H, et al. Proliferation potential-related protein, an ideal esophageal cancer antigen for immunotherapy, identifed using complementary DNA microarray analysis. Clin Cancer Res. 2004; 10: 6437-48.
  34. Ye F, Song J and Wang Y. Proliferation potentialrelated protein promotes the esophageal cancer cell proliferation, migration and suppresses apoptosis by mediating the expression of p53 and interleukin-17. Pathobiology. 2018; 85: 322-31.
  35. Dutertre M, Vagner S, Auboeuf D. Alternative splicing and breast cancer. RNA Biol. 2010; 7: 403-11.
  36. Pettigrew CA, Brown MA. Pre-mRNA splicing aberrations and cancer. Front Biosci. 2008; 13: 1090-105.
  37. Srebrow A, Kornblihtt AR. The connection between splicing and cancer. J Cell Sci. 2006; 119: 2635-41.
  38. Venables JP. Unbalanced alternative splicing and its significance in cancer. Bioessays. 2006; 28: 378-86.
  39. Kalnina Z, Zayakin P, Silina K, Line A. Alterations of pre-mRNA splicing in cancer. Genes Chromosomes Cancer. 2005; 42: 342-57.
  40. Brinkman BM. Splice variants as cancer biomarkers. Clin Biochem. 2004; 37: 584-94.
  41. Jurica MS, Moore MJ. Pre-mRNA splicing: awash in a sea of proteins. Mol Cell. 2003; 12: 5-14.
  42. Zhou Z, Licklider LJ, Gygi SP, Reed R. Comprehensive proteomic analysis of the human spliceosome. Nature. 2002; 419: 182-5.
  43. Hermann H, Fabrizio P, Raker VA, Foulaki K, Hornig H, Brahms H, et al. snRNP Sm proteins share two evolutionarily conserved sequence motifs which are involved in Sm protein-protein interactions. EMBO J. 1995; 14: 2076-88.
  44. Wahl MC, Will CL, Luhrmann R. The spliceosome: design principles of a dynamic RNP machine. Cell. 2009; 136: 701-18.
  45. Ziwei Guo, Jun Liang. Comprehensive analysis of the expression and prognostic value for SNRP members in hepatocellular carcinoma. DOI: 10.21203/rs.3.rs-633291/v1.

Tables

Table 1 The chromosomal locations of SNRP members

SNRP familys

SNRPB

SNRPD1

SNRPD2

SNRPD3

SNRPE

SNRPF

SNRPG

Chromosomal location

20p13

18q11.2

19q13.2

22q11.23

1q32

12q32.1

2p13.3


Table 2 Differential expression analyses of SNRP family in transcription level in hepatocellular carcinoma (ONCOMINE).

 

Types of cancer vs. normal

Fold change

p-value

t-test

References

SNRPB

Hepatocellular carcinoma vs. normal

2.315

4.23E-75

22.843

Roessler et al., 2010

 

Hepatocellular carcinoma vs. normal

2.338

1.12E-6

5.744

Roessler et al., 2010

SNRPD1

Hepatocellular carcinoma vs. normal

3.270

2.55E-97

27.765

Roessler et al., 2010

 

Hepatocellular carcinoma vs. normal

2.880

7.91E-9

7.287

Roessler et al., 2010

SNRPD2

Hepatocellular carcinoma vs. normal

2.160

4.05E-82

24.017

Roessler et al., 2010

 

Hepatocellular carcinoma vs. normal

2.052

5.00E-9

7.882

Roessler et al., 2010

SNRPE

Hepatocellular carcinoma vs. normal

2.971

1.81E-103

28.959

Roessler et al., 2010

 

Hepatocellular carcinoma vs. normal

2.046

1.10E-25

12.246

Chen et al., 2002

 

Hepatocellular carcinoma vs. normal

2.344

2.12E-7

6.554

Roessler et al., 2010

 

Table 3 The multiple comparisons for SNRP members about the expression levels

SNRP members

Significant unique analyses*

High expression

N

Best gene rank

N

Median expression

SNRPB

22

4.82 ± 3.05a

90

203.73 ± 162.09a’

SNRPD1

32

2.96 ± 2.91bc

89

24.41± 18.35b’

SNRPD2

21

4.19 ± 3.08ab

89

221.16 ± 145.40a’

SNRPD3

12

5.08 ± 1.93a

92

87.90 ± 51.95b’

SNRPE

38

2.00 ± 2.35c

90

44.74 ± 31.68b’

SNRPF

16

5.13 ± 2.33a

88

73.91 ± 50.38b’

SNRPG

23

2.09 ± 2.31c

88

64.82 ± 40.17b’

F


6.208

 

3.799

P


<0.001

 

0.007

*  by ONCOMINE database; a, b, c represent post hoc by one-way ANOVA

♯  by UALCAN online server; a’, b’ represent post hoc by one-way ANOVA

 

Table 4. The prognostic values of SNRP family members in liver cancer patients (Kaplan–Meier plotter).

SNRP family

OS

 

RFS

 

PFS

Cases

HR

95%Cl

p

 

Cases

HR

95%Cl

p

 

Cases

HR

95%Cl

p

SNRPB

364

1.61

1.13−2.28

0.0076

 

316

1.65

1.16-2.36

0.0048

 

370

1.52

1.11-2.08

0.0083

SNRPD1

364

1.94

1.36-2.76

0.00021

 

316

1.68

1.2-2.35

0.0022

 

370

1.57

1.17-2.1

0.0026

SNRPD2

364

1.4

0.98-2

0.062

 

316

1.53

1.1-2.13

0.01

 

370

1.41

1.05-1.89

0.02

SNRPD3

364

0.73

0.5-1.06

0.097

 

316

1.33

0.95-1.86

0.091

 

370

1.28

0.95-1.73

0.11

SNRPE

364

1.65

1.16-2.35

0.005

 

316

1.59

1.05-2.41

0.026

 

370

1.38

0.97-1.97

0.076

SNRPF

364

1.3

0.9-1.88

0.16

 

316

1.34

0.93-1.94

0.12

 

370

1.36

0.98-1.89

0.066

SNRPG

364

1.64

1.16-2.32

0.0048

 

316

1.52

1.09-2.11

0.012

 

370

1.45

1.08-1.94

0.013

Bold values mean p < 0.05

  

Table 5 OS and DFS for the altered and unaltered groups.

 

Variables

 

levels

Altered groups

Unaltered groups

 

p

Total (n)

Events (n)

Months

(95%Cl)

Total (n)

Events (n)

Median months

SNRPB

OS

5

0

NA

973

300

81.73

0.141

 

DFS

5

5

38.00

870

417

29.36

0.192

SNRPD1

OS

4

0

NA

974

300

83.18

0.420

 

DFS

4

0

NA

871

422

29.36

0.236

SNRPD2

OS

10

1

43.10

968

299

83.18

0.398

 

DFS

9

3

NA

866

419

29.66

0.657

SNRPD3

OS

4

2

3.35

974

298

83.18

0.637

 

DFS

NA

SNRPE

OS

60

16

NA

917

284

83.18

0.768

 

DFS

54

32

12.69

820

389

31.90

0.031

SNRPF

OS

3

0

NA

975

300

81.73

0.161

 

DFS

3

1

16.40

872

421

29.66

0.539

SNRPG

OS

NA

 

DFS

NA

OS: Overall survival;

DFS: Disease-free survival