Open Access

Construction of a model to predict the prognosis of patients with cholangiocarcinoma using alternative splicing events

  • Authors:
    • Hua‑Yu Wu
    • Yi Wei
    • Li‑Min Liu
    • Zhong‑Biao Chen
    • Qi‑Ping Hu
    • Shang‑Ling Pan
  • View Affiliations

  • Published online on: September 10, 2019     https://doi.org/10.3892/ol.2019.10838
  • Pages: 4677-4690
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cholangiocarcinoma (CCA) is a type of malignant tumor that originates in the mucosal epithelial cells of the biliary system. It is a highly aggressive cancer that progresses rapidly, has low surgical resection rates and a high recurrence. At present, no prognostic molecular biomarker for CCA has been identified. However, CCA progression is affected by mRNA precursors that modify gene expression levels and protein structures through alternative splicing (AS) events, which create molecular indicators that may potentially be used to predict CCA outcomes. The present study aimed to construct a model to predict CCA prognosis based on AS events. Using prognostic data available from The Cancer Genome Atlas, including the percent spliced index of AS events obtained from TCGASpliceSeq in 32 CCA cases, univariate and multivariate Cox regression analyses were performed to assess the associations between AS events and the overall survival (OS) rates of patients with CCA. Additional multivariate Cox regression analyses were used to identify AS events that were significantly associated with prognosis, which were used to construct a prediction model with a prognostic index (PI). A receiver operating characteristic (ROC) curve was used to determine the predictive value of the PI, and Pearson's correlation analysis was used to determine the association between OS‑related AS events and splicing factors. A total of 38,804 AS events were identified in 9,673 CCA genes, among which univariate Cox regression analysis identified 1,639 AS events associated with OS (P<0.05); multivariate Cox regression analysis narrowed this list to 23 CCA AS events (P<0.001). The final PI model was constructed to predict the survival of patients with CCA; the ROC curve demonstrated that it had a high predictive power for CCA prognosis, with a highest area under the curve of 0.986. Correlations between 23 OS‑related AS events and splicing factors were also noted, and may thus, these AS events may be used to improve predictions of OS. In conclusion, AS events exhibited potential for predicting the prognosis of patients with CCA, and thus, the effects of AS events in CCA required further examination.

Introduction

Cholangiocarcinoma (CCA) is a malignant tumor that originates in the mucosal epithelial cells of the biliary system. With a high degree of malignancy, rapid progression, low surgical resection rates and high recurrence rates, CCA prognoses are unsatisfactory (14). Recent studies of the molecular pathological mechanisms of CCA have demonstrated that tumor formation, growth, invasion, metastasis and other processes in CCA are regulated by a variety of molecules and signal transduction pathways, such as the aPKC-ι/P-Sp1/Snail signaling pathway and the Merlin/YAP/c-Myc/mTOR signaling pathway (5,6). This information has assisted early diagnosis, prognosis predictions and drug development. However, due to the complexity of CCA development, there are currently no specific biomarkers that meet clinical standards (710). Therefore, there is an urgent need to identify suitable biomolecular markers that can screen for CCA and assist in prognostic evaluations.

Eukaryotic mRNA precursors are known to remove certain unused fragments during the maturation process of mRNAs, allowing the remaining fragments to be rejoined. The fragments that are removed or left in place vary in their effects, depending on the cell type and status. Thus, a gene can encode multiple proteins in a process called alternative splicing (AS) (11,12). Previous studies have demonstrated that AS events occur ubiquitously in eukaryotes, with >95% of genes undergoing AS events (1315). However, AS events are more common in cancer cells and affect cancer development and treatment resistance (1624). In addition, AS events in tumor cells may be used as molecular markers that can differentially diagnose tumor types and predict prognosis (2528), as well as serve as potential targets for cancer treatment (2933). Application of AS events as a means diagnosing and predicting prognosis has been demonstrated in in prostate adenocarcinoma, uteri corpus endometrial carcinoma and colorectal cancer (3436).

CCA involves a high frequency of gene mutations and abnormal epigenetic changes, including DNA methylation, and histone and RNA modifications (37), all of which may lead to the occurrence of AS events (19,3847). AS events modify gene expression and influence protein structure by changing coding regions, thus regulating certain biological processes. For example, the widely reported tumor suppressor genes tumor protein p53, AT-rich interaction domain 1A, PTEN and PI3K, as well as the proto-oncogenes NOTCH1 and MET proto-oncogene, receptor tyrosine kinase, cause variations in gene function through AS events, thus affecting cancer development (30). Similarly, the apoptosis-related genes BCL-X and modulator of VRAC current 1 have been demonstrated to serve opposite functions in promoting and resisting apoptosis due to the occurrence of AS events (4851). AS can also alter the amino acid sequence of a protein, which may destroy the target for certain antitumor drugs and result in drug resistance (20,30,52). AS events therefore serve a unique role in the diagnosis and treatment of tumors.

Previous studies on the association between AS events and tumors have focused on the single-gene level, and few large-scale data mining studies based on high-throughput sequencing exist (5355). The Cancer Genome Atlas (TCGA) database (portal.gdc.cancer.gov/) contains high-throughput sequencing data and comprehensive clinical information from a large number of cancer samples. Previous studies using TCGA data have reported that AS events may be used as prognostic indicators for lung (56), ovarian (57), bladder (58) cancer and gastrointestinal pan-adenocarcinomas (59,60). However, the prognostic value of AS events has not yet been reported for CCA. In the present study, the overall survival (OS)-related AS events in CCA were systemically evaluated. These findings may facilitate development of novel genomic models for clinical cancer management, and construction of novel models based on the prognostic index (PI) to predict CCA survival.

Materials and methods

Data collection

The percent spliced index (PSI) values for CCA AS events were downloaded from the TCGASpliceSeq database (http://bioinformatics.mdanderson.org/TCGASpliceSeq/) (61). PSI is the ratio of normalized read counts indicating inclusion of a transcript element to the total normalized read counts for that event (i.e., both inclusion and exclusion read counts). PSI values range from 0 to 1, indicating the likelihood of the existence of an exon. Information related to seven types of AS events: Exon skip (ES), mutually exclusive exons (ME), retained intron (RI), alternate promoter (AP), alternate terminator (AT), alternate donor (AD) site and alternate acceptor (AA) site events, was included in the analysis. Corresponding clinical information and gene expression levels of the samples were obtained from TCGA database. The database included data from 32 patients with CCA with complete clinical information and total survival time >90 days. Of the 32 patients with CCA, 13 were male and 19 were female. The age of the patients was 29–82, and the median age was 66.5, with 27 patients older than 50 and 5 patients younger than 50.

Association between AS events and survival

R software (R version 3.4.2) (62) was used to perform univariate Cox regression analysis to investigate the PSI prognostic values from the seven types of AS events and the differentially expressed genes in the 32 CCA cases. AS events with P<0.05 were selected for further analysis. The event-dependent survival curve for the top three AS events was plotted (collating P-values from low to high).

Systematic review

To confirm that the identified AS events were CCA-specific, all prognostic AS events from different types of cancer were extracted from published articles on PubMed (https://www.ncbi.nlm.nih.gov/), Wiley Online Library (https://onlinelibrary.wiley.com/), EBSCO (https://www.ebsco.com/), Web of Science (https://www.webofknowledge.com/), and Google Scholar (https://scholar.google.com/) on or prior to 1st May 2019 for comparison. The key words were as follows: ‘cancer OR carcinoma OR adenocarcinoma OR tumour OR tumor OR malignanc* OR neoplas*’ AND ‘prognostic OR prognosis OR predict*’ AND ‘ ‘alternative splicing’ OR AS’. The studies which were included needed to meet the following criteria: i) Studies describing cancer prognosis with a predicted model constructed using prognostic AS events; and ii) the prognostic AS events could be extracted from the studies. All the included studies were assessed using the bias assessment for studies of diagnostic accuracy (QUADAS) guidelines (63) and had a QUADAS score of ≤7. The following data were extracted from the included studies: First author's name, year of publication and prognostic AS events.

AS event interaction analysis and gene network construction

The interactions of the seven types of AS events were analyzed using the UpSetR package for R (64), and an UpSet plot was drawn. The network was constructed using the Reactome FI plug-in (65) for Cytoscape 3.6.0 (66), with AS events identified using univariate Cox regression analysis (P<0.01). Genes with AS events were processed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) gene enrichment analyses. Due to the occurrence of a large number of significant GO pathways, only the top five pathways per group were presented using the ggplot2 package for R (67).

PI construction and prognostic value assessment

To assess the association between AS events and OS, univariate Cox regression was carried out with a significance threshold of P<0.05. The top ten most promising AS events, based on their prognostic value, AS events selected for each splicing type. Multivariate Cox regression analysis was performed, splicing events with P<0.05 were selected as PIs. The model was established by grouping together all splicing events of the seven splicing event types. The samples were divided into two groups according to the median PSI value of the splicing events. The formula used to calculate the PI was as follows:

Risk score =

∑inPSIi*βi

The clinical prognostic value of the PI was assessed using Kaplan-Meier (K-M) curve analysis and time-dependent receiver operator characteristic (ROC) curve analysis. The time-dependent ROC curve was drawn using the survival ROC package for R (68).

Experimental validation of AS events in clinical CCA tissue samples

In addition to the aforementioned analyses, the existence of the AS events predicted by the TCGASpliceSeq database was verified using PCR analyses with six samples of CCA tumor tissues separately collected from a 37-year-old male patient and a 38-year-old female patient during surgery at the Pathology Department of the First Affiliated Hospital of Guangxi Medical University (Nanning, China) between February 2017 and October 2017. The study was approved by the Ethics Committee of the First Affiliated Hospital of Guangxi Medical University, and written informed consent was obtained from all patients. Total RNA was extracted using the AxyPrep Multisource Total RNA Miniprep kit (Axygen; Corning, Inc.), quantified using a NanoDrop 2000 (Thermo Fisher Scientific, Inc.) and 500 ng total RNA was reverse-transcribed into cDNA using MiScript® II RT SuperMix kit (Vazyme). AS events in phytanoyl-CoA 2-hydroxylase (PHYH)_100582_ES and transferrin receptor 2 (TFR2)_80979_ES were selected for verification. The mean PSI of PHYH_100582_ES in TCGASpliceSeq database was 0.78, and ES was present in exons 7 and 8. The forward primer (primer 1) was designed based on exon 6, and the sequence was 5′-GATACTGCACTCTCCCCGAG-3′. The reverse primer (primer 2) was designed based on exon 9, and the primer sequence was 5′-GACCAGATCCGTGGATGAGC-3′. The PCR system contained 10 µl 2X PCR Master mix (Thermo Fisher Scientific, Inc), 1 µl each of the forward and reverse primers (10 µM), 1 µl cDNA and 7 µl nuclease-free water (total volume, 20 µl). The thermocycling conditions were as follows: 95°C for 3 min, followed by 35 cycles of 95°C for 30 sec, 60°C for 30 sec and 72°C for 1 min. PCR products were subjected to electrophoresis on 2% agarose gels. The mean PSI of TFR2_80979_ES in the TCGASpliceSeq database was 1, and ES was present in exon 10. The forward primer (primer 3) was designed based on exon 9, and the primer sequence was 5′-CAGCCCATCAGTGCAGACAT-3′. The reverse primer (primer 4) was designed based on exon 11, and the primer sequence was 5′-TTGTTGACCACTAGCCGCAG-3′. The PCR system and thermocycling conditions were as mentioned above.

Correlation analysis between splicing factors and prognosis-related AS events

A splicing factor that serves as a splicing activator when bound to an intronic enhancer element may function as a repressor when bound to its splicing element in the context of an exon (69). Therefore, the correlations between splicing factors and the prognosis-related AS events were investigated. Splicing factor information was obtained from the SpliceAid 2 database (http://193.206.120.249/splicing_tissue.html) (70). Univariate Cox regression analysis was performed on the splicing events and splicing factors, followed by Pearson's correlation analysis between the significant splicing events, splicing factors and the OS-related AS events identified by multivariate Cox analysis. The results were visualized using Cytoscape version 3.4.0 (66).

Results

CCA AS events

A total of 38,804 AS events were obtained from 9,673 genes in 32 CCA cases from TCGA SpliceSeq dataset. The numbers of each type of AS event and the corresponding genes are presented in Fig. 1. The results demonstrated that a single gene may undergo multiple AS events simultaneously. ES was the most frequent AS event in this dataset.

AS events associated with CCA OS rates

Univariate Cox analysis revealed that 1,639 AS events were associated with CCA OS rates (P<0.05) and that certain genes had undergone multiple OS-related AS events. Visualization of the intersecting sets was performed using UpSet plotting software (Fig. 2), and ≤3 AS events were identified to be associated with OS rates within the same gene. KEGG pathway enrichment analysis revealed that the genes corresponding to these AS events were primarily enriched in ‘biosynthesis of antibiotics’, ‘axon guidance’, ‘pancreatic cancer’, ‘RAP1 signaling pathway’ and ‘SNARE interactions in vesicular transport’ (Fig. 3A). GO gene enrichment analysis demonstrated that the genes with OS-related AS events corresponded to ‘protein transport’, ‘cell-cell adhesion’, ‘apoptotic process’, ‘protein localization to the nucleus’ and ‘positive regulation of transcription from RNA polymerase II promoters’ (Fig. 3B). Using a network map of these genes, MYC associated factor X (MAX), mitogen-activated protein kinase 11 (MAPK11), γ-aminobutyric acid type A receptor-associated protein-like 1 (GABARAPL1), checkpoint kinase 1 (CHEK1), X-linked inhibitor of apoptosis (XIAP) and forkhead box A1 (FOXA1) exhibited the highest degrees of connectivity and were thus considered critical positions of the network (Fig. 4).

OS-related AS events are CCA-specific

Comparisons with other types of cancer demonstrated that ME-GTF2H3-306194 occurred in papillary thyroid cancer and CCA, whereas the other identified AS events were CCA-specific (Table I).

Table I.

Prognostic alternative splicing events in different types of cancer.

Table I.

Prognostic alternative splicing events in different types of cancer.

Author, yearTypePMIDAlternative splicing events(Refs.)
Lin et al, 2019Papillary thyroid cancer30986203AA-SHPRH-78032, AA-CASK-88861, AD-FBXL19-36205, AD-SAT2-39030, AD-TRO-89255, AD-CSTF2-89611, AP-ZC3H11A-9456, AP-STK32C-13483, AP-GRB2-43439, AP-CRTC1-48500, AP-ERCC1-50440, AP-ESR1-78161, AT-MAGI3-4271, AT-TPM1-30982, AT-ATP8B3-46544, AT-MAST1-47878, AT-SPAG16-57327, AT-CBWD5-86498, AT-OLFM1-88103, ME-NSMF-193275, ME-GTF2H3-306194, RI-C11orf49-15609, RI-ZNF276-38138, RI-USP36-43917, RI-NUDT18-82937, RI-NAPRT1-85430(95)
Gao et al, 2019Uteri corpus endometrial carcinoma30640723AP-BDNF-14763, AP-DDX58-86057, AP-FYTTD1-68310, AP-GNAL-44643, AP-GPATCH2L-28538, AP-HUS1-79610, AP-MAP4-64545, AT-IPO11-72190, AT-ZFAND4-11368, ES-CKMT2-72660, ES-CMC2-37735, ES-FBXL12-47421, ES-NDUFB1-28987, ES-PSMD12-43112, ES-ZNF528-51457, RI-AP3M2-83565, RI-DNASE1L3-65424, RI-GABARAP-38871(34)
Huang et al, 2018Prostate adenocarcinoma30221674ES-TCEB2-33303, AD-ABHD17A-46558, AP-FKBP2-16603, ES-TXN-87183, AP-FKBP2-16602, AD-YPEL3-36074, ES-STXBP2-47124, AT-PTGDS-88235, AT-HMGA2-22879, ES-NHLRC3-25701(36)
He et al, 2018Bladder urothelial carcinoma30048970AA-B4GALT2-1228, ES-TMTC2-9217, ES-TIMM9-11224, ES-APOBEC3D-26508, AP-TPD52-35921, ES-MICU1-4164, ES-DDX11-8115, ES-SMC6-22132(94)
Zhang et al, 2019Breast carcinoma30984247AA-CARM1-47598, AA-ZBTB25-27884, AA-GPBP1-72126, AA-ZNRF1-37578, AA-DDX41-74796, AA-CTDSP1-57478, AD-OS9-22701, AD-HN1-43371, AD-THTPA-26757, AD-NTMT1-87866, AD-MGME1-58753, AD-SEC31A-69735, AP-SEC22A-66462, AP-ALG3-67851, AP-PACS2-29630, AP-ECE2-67857, AP-HSP90AB1-76378, AT-MAGT1-89535, AT-RCBTB1-25898, AT-SIN3B-48214, AT-SARNP-22252, AT-ZNF675-48822, AT-STOX2-71289, AT-NIPAL3-1110, ES-NDUFA12-23737, ES-UBR4-880, ES-COPS3-39468, ES-ABCE1-70753, ES-CCNI-69628, ES-RPAP1-30096, ME-HLCS-96019, RI-RBM48-80441, RI-RBM6-64936, RI-RPAP1-30095, RI-METTL17-26476, RI-POMGNT1-2787, RI-TRABD-62792, RI-WDR6-64794, RI-FASTK-82335, RI-NAA38-81579(96)
Lin et al, 2018Esophageal adenocarcinoma30131306AA-U2AF1L4-49280, AA-TICRR-32428, AA-RSRC2-24968, AA-PREPL-53439, AA-PPIL2-61247, AA-FAM135A-76637, AA-CDV3-66839, AA-ABCB7-89517, AD-ZNF384-19927, AD-RPP14-65434, AD-PQBP1-89028, AD-MFSD11-43690, AD-COX6C-84682, AP-ZNF623-85469, AP-KIAA0513-37876, AP-FAM19A5-62732, AP-ALDH6A1-28367, AT-TRIM4-80864, AT-RNASEH2B-25927, AT-RNASEH2B-25926, AT-MCPH1-82574, AT-ARL6-65732, AT-AHI1-77886, ES-TNC-87345, ES-PML-31651, ES-NBPF15-91080, ES-MYL6-22384, ES-MRPL43-12857, ES-IRF9-117161, ME-SDR39U1-27012, ME-KLHL2-71038, ME-CMC2-37707, RI-ZNF131-71926, RI-SLC52A3-58464, RI-PPARGC1B-74051, RI-PCGF3-68404, RI-MDK-15570, RI-MAF-37687, RI-FAM9C-88504(60)
Lin et al, 2018Stomach adenocarcinoma30131306AA-RPLP0-24727, AA-NAT6-64990, AA-MRVI1-14373, AA-LMO7-26065, AA-BDKRB2-29192, AD-YIPF2-47605, AD-SPHK2-50793, AD-SENP1-21411, AD-PGAP2-14004, AD-NFATC1-46241, AD-CCDC51-64653, AP-RCAN1-60494, AP-PLCD1-64009, AP-LTBP1-53179, AP-FAM65B-75537, AP-ABL2-9101, AT-ZNF846-47399, AT-ZFYVE28-68559, AT-STEAP4-80362, AT-STEAP4-80361, AT-KIF1B-602, AT-KIF1B-601, AT-CXCL12-11344, AT-CLDN11-67617, AT-ABCB5-78909, ES-UBXN11-1263, ES-TMEM230-58637, ES-SRSF3-75985, ES-SORBS1-12641, ES-P4HA2-73263, ES-CREM-11245, ME-N4BP2L1-25590, ME-KDM6A-98323, ME-FYN-77273, ME-CCDC53-106010, RI-TREX1-64682, RI-SRSF7-53276, RI-RPS15-46490, RI-LDHA-14642, RI-BICD2-86883, RI-ALS2CL-64462(60)
Lin et al, 2018Colon adenocarcinoma30131306AA-RASSF7-13691, AA-PTGR1-87219, AA-FAM173A-32964, AA-DPP3-17040, AA-CDV3-66842, AD-RNF14-73855, AD-IP6K2-64759, AD-HPS4-61506, AD-HDGF-8323, AD-ANKRD46-84712, AD-ADPGK-31594, AP-TUBB3-38167, AP-RAB3IP-23345, AP-MAZ-35938, AP-FADS2-16289, AP-ENO2-20011, AT-ZNF765-51718, AT-UPK3B-80182, AT-RASEF-86677, AT-RASEF-86676, AT-NRG4-31911, AT-AIG1-77972, ES-VTI1B-28083, ES-STRN3-27098, ES-RHOC-4236, ES-PRMT1-51042, ES-PLEKHM2-767, ES-DMWD-50528, ES-D2HGDH-58423, ME-CNOT10-63822, RI-ZNF226-50290, RI-NPIPA5-34148, RI-ELP5-38889, RI-ALS2CL-64463(60)
Lin et al, 2018Rectal adenocarcinoma30131306AA-ZNF467-82205, AA-RNPC3-3907, AA-GGT1-61440, AA-BTN3A1-75660, AD-OSBPL9-2975, AD-METTL23-43637, AD-BCS1L-57522, AP-TADA2B-68732, AP-PTCH1-86955, AP-DAB2IP-87442, AT-PUS10-53676, AT-NOTCH2NL-4437, ES-SPAG9-42496, ES-SERPINA1-29134, ES-PHB2-20048, ES-FGFR1OP2-20856, ME-RBMS2-22465, RI-ZNF692-10557, RI-WDR33-55246, RI-TMEM91-50046, RI-SIDT2-18886, RI-EXOSC9-70501, RI-ADARB1-60863(60)

[i] PMID, PubMed manuscript ID.

AS events that may be used as CCA prognostic indicators

The three most statistically significant AS events were selected, and their gene expression levels were used to calculate their prognostic values in CCA (Fig. 5). K-M analysis revealed that these prognostic signatures significantly separated patients with distinct prognoses. To assess the ability of these splicing events to predict CCA prognosis, a time-dependent ROC curve was established. The results of the ROC curve suggested that a single indicator was not enough to predict patient prognosis, as the area under the curve (AUC) values were low for the AS events and gene expression values (Fig. 5). Therefore, ten AS events with the most significant prognostic values in each splicing type were selected to construct the PIs (Table II). The results suggested that these PIs had higher predictive values compared with single indicators (Fig. 6). The AUCs for each of the seven subtypes of AS were as follows: AA, 0.901; AD, 0.858; AP, 0.986; AT, 0.839; ES, 0.939; ME, 0.779; and RI, 0.859. In addition, the ten most significant AS events from all types combined were selected to construct a final prognostic model; the AUC was 0.984 (Fig. 6). It is worth noting that the predictive efficacy of the prognostic model with seven combined splicing events (0.984) was slightly lower than that obtained with AP alone (0.986).

Table II.

Prediction models for cholangiocarcinoma based on each type of splicing event.

Table II.

Prediction models for cholangiocarcinoma based on each type of splicing event.

Risk scoreAlgorithmHR (95% CI)P-valueAUC
Risk score (AA)PSIST3GAL4_19399_AA × 0.849 +10.990 (3.818–31.61)<0.0010.901
PSITECR_47998_AA × (−6.242) +
PSITGIF1_44506_AA × (−0.713)
Risk score (AD)PSISYNGR1_62301_AD × (−0.645) +3.375 (1.227–9.283)0.0180.858
PSIZHX3_59398_AD × (−0.277)
Risk score (AP) PSIC12orf65_25058_AP × (−0.147) +17.910 (5.658–56.67)<0.0010.986
PSICHMP3_54439_AP × 1.274 +
PSISH3KBP1_88640_AP × 0.432
Risk score (AT)PSIMBLAC2_72765_AT × 0.166 +7.622 (2.576–22.55)<0.0010.839
PSITGFB3_28531_AT × (−1.244)
Risk score (ES)PSIACAD9_66674_ES × (−1.952) +19.090 (5.705–63.86)<0.0010.939
PSIFRMD8_16850_ES × (−0.701) +
PSIPLEKHG2_49826_ES × (−1.49) +
PSITP53I11_15489_ES × 2.014 +
PSIUBE2F_58170_ES × 8.384
Risk score (ME)PSIFGFR3_68513_ME × (−0.15) +4.977 (1.790–13.84)0.0020.779
PSIGRB10_79717_ME × (−0.068) +
PSIGTF2H3_306194_ME × 1.421 +
PSIRNF146_114496_ME × (−0.085) +
PSISORBS2_71377_ME × (−0.282)
Risk score (RI) PSIC11orf88_18667_RI × (−0.073) +6.358 (2.295–17.61)<0.0010.859
PSIDET1_32385_RI × (−0.214) +
PSIGAREML_52884_RI × (−0.332)
Risk score (merged)PSITFDP1_26387_AD × (−0.57) +19.670 (5.842–66.22)<0.0010.984
PSIMBLAC2_72766_AT × (−0.838) +
PSIHACL1_63592_ES × (−0.62) +
PSIPLEKHG2_49826_ES × (−4.314) +
PSIUBE2F_58170_ES × 7.06

[i] PSI, percent spliced index; HR, Hazard ratio; CI, confidence interval; AUC, area under the curve; AA, alternate acceptor; AD, alternate donor; AP, alternate promoter; AT, alternate terminator; ES, exon skip; ME, mutually exclusive exon; RI, retained intron; ‘ST3GAL4_19399_AA’ represents ‘gene symbol_alternative splicing event ID_splice type’.

Confirmation of PHYH_100582_ES and TFR2_80979_ES by clinical samples

Towards PHYH_100582_ES, the PCR products were predicted to comprise two bands; the band without ES was predicted to be 414 bp in length, whereas the band size following ES was predicted to be 144 bp in length (Fig. 7A). If the TFR2_80979_ES event did not occur, the PCR product was predicted to be 238 bp; following the ES event, the PCR product size was predicted to be 140 bp (Fig. 7B). Since the PSI value of this ES event was 1, the ES event occurred in all TFR2 mRNAs; if the TCGASpliceSeq database was accurate, the PCR product was predicted to be a single band. The results of PHYH_100582_ES and TFR2_80979_ES were consistent with those predicted by the TCGASpliceSeq database. The PCR products of PHYH_100582_ES were 414 and 144 bp (Fig. 7C), suggesting that, in CCA tissues, a proportion of PHYH mRNA had undergone an ES event. The PSI of TFR2_80979_ES in the TCGASpliceSeq database was 1, indicating that ES events occurred in all TFR2 mRNAs. The electrophoresis result demonstrated that the product of TFR2_80979_ES was a single band of 140 bp; no band of 238 bp was observed (Fig. 7D), suggesting that ES events occurred in all TFR2 mRNA. These results were consistent with those suggested by the RNA sequencing data from TCGASpliceSeq.

Correlation between splicing factors and prognosis-related AS events

A total of 11 AS events and corresponding splicing factors significantly related to CCA OS rates (P<0.05) were identified. Multivariate Cox analysis identified 23 prognosis-related AS events (Table III; P<0.001); 17 of these exhibited positive correlations with OS, whereas 6 exhibited negative correlations with OS. Of these 23 AS events, 15 were correlated with a group of splicing factors, the expression of which was directly associated with AS events (|r|>0.3; P<0.05). AS events are primarily regulated by splicing factors, which often bind to pre-mRNAs and regulate RNA splicing by influencing exon selection and splicing site. Therefore, the associations between survival-related AS events and splicing factors were determined (Fig. 8).

Table III.

Predictive factors identified for cholangiocarcinoma using multivariate logistic regression.

Table III.

Predictive factors identified for cholangiocarcinoma using multivariate logistic regression.

Splicing typeGene symbolAS ID
AAST3GAL419399
AATECR47998
AATGIF144506
ADSYNGR162301
ADZHX359398
APC12orf6525058
APCHMP354439
APSH3KBP188640
ATMBLAC272765
ATTGFB328531
ESPLEKHG249826
ESACAD966674
ESTP53I1115489
ESFRMD816850
ESUBE2F58170
MEFGFR368513
MERNF146114496
MEGRB1079717
MESORBS271377
MEGTF2H3306194
RIC11orf8818667
RIDET132385
RIGAREML52884

[i] AA, alternate acceptor; AD, alternate donor; AP, alternate promoter; AT, alternate terminator; ES, exon skip; ME, mutually exclusive exon; RI, retained intron; AS ID, alternative splicing event ID.

Discussion

Using data mined from TCGASpliceSeq and in silico approaches, the present study identified that a number of AS events are closely associated with survival in CCA. The present study is the first to report this type of result. The results of the present study also demonstrated that AS events may be used to construct a PI model that effectively determines CCA prognosis.

The incidence of CCA has increased in recent years. The prognosis of advanced CCA is poor, with an extremely low 5-year survival rate (7). The prognosis depends on the synergistic effects of various factors; however, no clear and effective molecular markers have been identified for CCA diagnosis and treatment. Carcinoembryonic antigens (CEAs) and carbohydrate antigen (CA) 199, CA 125, CA 50 and CA 242 are currently used as CCA tumor markers, but these biomarkers have disadvantages in clinical application. For example, the majority of studies that examined CA 199 as a biomarker for the detection of CCA have reported its suboptimal accuracy, with wide variation in reported sensitivity (38–93%) and specificity (67–98%) (71). CA 125 is upregulated in 65% of patients with CCA and is of value in predicting survival (72); however, there are no further studies on CA 125 in CCA. CA 50 exhibits cross-antigenicity with CA 199, and elevated serum CA 50 levels are commonly used to diagnose or prognose pancreatic and colorectal cancers (73). However, Shan et al (74) demonstrated that elevated serum CA 50 levels are not always associated with the expression of CA 50 in cancer tissues. Despite the clinical utility of CA 242, it is not sufficiently effective in the early detection of cancer, since elevated classical tumor biomarker levels indicate the presence of a significant number of cancer cells (75).

At the genetic level, a number of genes are abnormally expressed in CCA. For example, the abnormal expression of genes such as transforming growth factor β1, SMAD4, c-MET, matrix metallopeptidase 7, vascular endothelial growth factor (VEGF)-A, VEGF-B, VEGF-C and VEGF-D are associated with poor prognosis (7679). Mutations in genes such as human epidermal growth factor receptor 2, TP53, KRAS, cytosolic NADP-dependent isocitrate dehydrogenase and mitochondrial NADP-dependent isocitrate dehydrogenase can also affect the prognosis of patients with CCA, but the prognostic value of these genes remains controversial and has not been applied clinically (72). Due to the complexity of cancer, diagnosis or prognosis based on a single molecule is limited. The advent of high-throughput sequencing technologies allows us to address this issue by identifying more genomic abnormalities related to CCA.

Using the TCGASpliceSeq CCA data, 38,804 AS events were identified in 9,673 CCA genes, some of which exhibited multiple AS events occurring simultaneously. AS events are widespread in CCA and may be related to its occurrence and development. For instance, the first AS event reported to be associated with CCA prognosis was exon 2 skipping of the trefoil factor 2 (TFF2) gene (80). TFF2 is highly expressed in a variety of tumors, including CCA. Exon 2 skipping AS events lead to the loss of exon 2, resulting in a decrease in wild-type TFF2 proteins. A high proportion of this AS event in TFF2 is associated with improved prognosis. Another example is P53, as alterations in the N-terminus of this protein by AS has been demonstrated to worsen the prognosis in patients with CCA (81).

The present study identified a large number of CCA AS events, 1,639 of which were correlated with patient prognosis (P<0.05). Bioinformatics analyses of all genes with significant prognostic values identified the following genes to be located at the center of the gene network: MAX, MAPK11, GABARAPL1, CHEK1, XIAP and FOXA1. These genes have been broadly reported to be involved in cell cycle regulation, autophagy, proliferation, apoptosis and have been previously demonstrated to be associated with cancer (8287). Tumor-specific AS events have been intensively studied by investigating the differentially alternatively spliced genes between tumor and adjacent normal tissues (88) in certain cancer types, such as breast (89), lung (56) and ovarian (57) cancer. Yosudjai et al (90) also demonstrated that aberrant AS of anterior gradient protein 2 homolog promoted cell proliferation, migration, invasion and adhesive potential in CCA; however, there has been a lack of research on its association with patient prognoses. Among the genes identified above, MAX forms a heterodimer with the proto-oncogene MYC, which binds to DNA to regulate the transcription of multiple genes, thus regulating cell proliferation, differentiation and apoptosis (82). The MAPK11 gene encodes a serine/threonine kinase that is widely involved in various cellular signal transduction pathways, such as p38 the MAPK signaling pathway by phosphorylating multiple target proteins (87). CHEK1 is a cell cycle monitoring-related protein, abnormalities of which result in DNA damage and the bypassing of the cell cycle checkpoints (91). FOXA1 is a transcription factor, the aberrant expression of which is closely associated with hepatocellular carcinoma (84); however, its association with CCA has not been previously reported.

The current study is the first to identify the characteristics of alternative CCA splicing events and establish a model that can predict CCA prognosis using PIs from AS events. Li et al (56), who studied non-small cell lung cancer, were the first to demonstrate a combined survival and correlation network between the expression of splicing factors and AS events; the present study provides a more accurate potential network by screening the significant prognostic AS events identified by univariate Cox regression.

The present study attempted to predict the prognosis of patients with CCA using the three most significant AS events and genes. The results were not satisfactory (the maximum AUC was 0.747; data not shown), likely due to individual differences between patients. Similarly, no single factor was able to accurately predict patient prognosis. Therefore, ten AS events with the most significant prognostic values, as calculated by multivariate Cox regression analysis for each type of AS event, were selected to construct a PI. The prognostic models based on the combined seven types of splicing events demonstrated comparable predictive efficacy to the model that used only the AP splicing events. However, the combined model was more successful at predicting patient prognosis compared with models based on any other single type of splicing event.

The results of the present study were compared with those reported in previous studies. All AS events identified in the present study were uniquely associated with CCA, with the exception of ME-GTF2H3-306194, which has been previously identified to have prognostic value in papillary thyroid cancer and CCA, indicating that ME-GTF2H3-306194 may serve similar functions in CCA and papillary thyroid cancer and suggesting a new treatment possibility for CCA and papillary thyroid cancer by targeting ME-GTF2H3-306194.

In addition, the existence of two AS events predicted by the TCGASpliceSeq database was verified using PCR analysis of six CCA tumor tissue samples. Metastasis was common in cholangiocarcinoma (7,92). The direct infiltration of cholangiocarcinoma cells along the bile duct wall is one of the main features of cholangiocarcinoma metastasis (7). Cancer cells diffuse invasively in the bile duct wall and coexist with the bile duct and surrounding connective tissue, making the infiltration range of cholangiocarcinoma difficult to identify, and thus non-cancerous bile duct tissues cannon be easily obtained during surgery (93). Therefore, only two AS events were verified, and the difference of the splicing events between tumor and non-tumor tissues was not determined.

To investigate the causes of AS events in CCA, the relationship between AS events and splicing factors was analyzed. Splicing factors are a class of RNA-binding proteins that affect the selection of cleavage sites by recognizing the cis-acting elements of mRNA precursors (27). Previous studies on lung (56), ovarian (57) and bladder (94) cancer have demonstrated that the expression of splicing factors was correlated with prognosis-related AS events; however, this phenomenon was not observed in the current study. Pearson's correlation analysis between a group of splicing factors, the expression of which was directly related to AS events, and general OS-related splicing events identified 15 AS events that were mutually associated. Therefore, in CCA, splicing factors may affect the structure of their own proteins by creating their own AS event and then subsequently affecting the AS events of other genes.

In summary, TCGASpliceSeq data were mined to identify the characteristics of AS events associated with CCA to establish a model that can predict CCA prognosis using PIs from AS events. The prognostic effect was estimated using ROC curves and was favorable, with AUCs >0.9. However, due to the small sample size, the validity of this model should be verified with additional clinical samples and in different populations. Several additional limitations of the present study also need to be addressed. The data in the current study were obtained from online databases and thus remain at the bioinformatics level. Additionally, among the AS events selected to construct the PI model, one event was not CCA-specific, which reduced the specificity of the PI model. In addition, although splicing factors have been demonstrated to contribute to changes in the splicing patterns of target genes and tumorigenesis, and certain cancer-specific splicing factors are associated with OS rates, there has been a lack of research on CCA-related splicing factors, and whether the splicing factors identified in the present study are truly CCA-specific requires additional research. Finally, although an accurate PI model was constructed by screening the significant prognostic AS events identified by univariate and multivariate Cox regression, the results are based on a single cohort and should be validated in additional cohorts; this is intended to be performed in our subsequent study.

Acknowledgements

Not applicable.

Funding

The study was supported by the Promoting Project of Basic Capacity for Young and Middle-aged University Teachers in Guangxi (grant no. 2017KY0111), the Innovation Project of Guangxi Graduate Education (grant no. YCBZ2017045), the National Natural Science Foundation of China (grant nos. 31760319, 81360066 and 81660241) and the Guangxi First-class Discipline Project for Pharmaceutical Sciences (grant no. GXFCDP-PS-2018).

Availability of data and materials

The datasets generated and analyzed during the current study are available from the TCGA data portal (portal.gdc.cancer.gov/).

Authors' contributions

HYW analyzed and interpreted data, and drafted the manuscript. YW, LML, ZBC, QPH and SLP obtained information from the database, supervised the data mining, and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of the First Affiliated Hospital of Guangxi Medical University (ethics approval no. 20170303-2). All the patients provided written informed consent to participate in this study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CCA

cholangiocarcinoma

TCGA

The Cancer Genome Atlas

K-M

Kaplan-Meier

AUC

area under the curve

ROC

receiver operator characteristic

PSI

percent spliced index

ES

exon skip

ME

mutually exclusive exons

RI

retained intron

AP

alternate promoter

AT

alternate terminator

AD

alternate donor site

AA

alternate acceptor site

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

References

1 

Popat K, McQueen K and Feeley TW: The global burden of cancer. Best Pract Res Clin Anaesthesiol. 27:399–408. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Verathamjamras C, Weeraphan C, Chokchaichamnankit D, Watcharatanyatip K, Subhasitanont P, Diskul-Na-Ayudthaya P, Mingkwan K, Luevisadpaibul V, Chutipongtanate S, Champattanachai V, et al: Secretomic profiling of cells from hollow fiber bioreactor reveals PSMA3 as a potential cholangiocarcinoma biomarker. Int J Oncol. 51:269–280. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Lu X, Zhou C, Li R, Deng Y, Zhao L and Zhai W: Long noncoding RNA AFAP1-AS1 promoted tumor growth and invasion in cholangiocarcinoma. Cell Physiol Biochem. 42:222–230. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Cai X, Li J, Yuan X, Xiao J, Dooley S, Wan X, Weng H and Lu L: CD133 expression in cancer cells predicts poor prognosis of non-mucin producing intrahepatic cholangiocarcinoma. J Transl Med. 16:502018. View Article : Google Scholar : PubMed/NCBI

5 

Luo G, Li B, Duan C, Cheng Y, Xiao B, Yao F, Wei M, Tao Q, Feng C, Xia X, et al: cMyc promotes cholangiocarcinoma cells to overcome contact inhibition via the mTOR pathway. Oncol Rep. 38:2498–2506. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Qian Y, Yao W, Yang T, Yang Y, Liu Y, Shen Q, Zhang J, Qi W and Wang J: aPKC-iota/P-Sp1/Snail signaling induces epithelial-mesenchymal transition and immunosuppression in cholangiocarcinoma. Hepatology. 66:1165–1182. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Razumilava N and Gores GJ: Cholangiocarcinoma. Lancet. 383:2168–2179. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Zhou G, Yang Z, Wang X, Tao R and Zhou Y: TRAIL enhances shikonin induced apoptosis through ROS/JNK signaling in cholangiocarcinoma cells. Cell Physiol Biochem. 42:1073–1086. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Tang J, Liao Y, He S, Shi J, Peng L, Xu X, Xie F, Diao N, Huang J, Xie Q, et al: Autocrine parathyroid hormone-like hormone promotes intrahepatic cholangiocarcinoma cell proliferation via increased ERK/JNK-ATF2-cyclin D1 signaling. J Transl Med. 15:2382017. View Article : Google Scholar : PubMed/NCBI

10 

Chng KR, Chan SH, Ng AHQ, Li C, Jusakul A, Bertrand D, Wilm A, Choo SP, Tan DMY, Lim KH, et al: Tissue microbiome profiling identifies an enrichment of specific enteric bacteria in opisthorchis viverrini associated cholangiocarcinoma. Ebiomedicine. 8:195–202. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Breitbart RE, Andreadis A and Nadalginard B: Alternative splicing: A ubiquitous mechanism for the generation of multiple protein isoforms from single genes. Annu Rev Biochem. 56:467–495. 1987. View Article : Google Scholar : PubMed/NCBI

12 

Blencowe BJ: Alternative splicing: New insights from global analyses. Cell. 126:37–47. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Oltean S and Bates DO: Hallmarks of alternative splicing in cancer. Oncogene. 33:5311–5318. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Wang ET, Rickard S, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP and Burge CB: Alternative isoform regulation in human tissue transcriptomes. Nature. 456:470–476. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Pan Q, Shai O, Lee LJ, Frey BJ and Blencowe BJ: Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet. 40:1413–1415. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Dvinge H and Bradley RK: Widespread intron retention diversifies most cancer transcriptomes. Genome Medi. 7:452015. View Article : Google Scholar

17 

Bechara EG, Sebestyén E, Bernardis I, Eyras E and Valcárcel J: RBM5, 6 and 10 differentially regulate NUMB alternative splicing to control cancer cell proliferation. Mol Cell. 52:720–733. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Ghigna C, Giordano S, Shen H, Benvenuto F, Castiglioni F, Comoglio PM, Green MR, Riva S and Biamonti G: Cell motility is controlled by SF2/ASF through alternative splicing of the Ron Protooncogene. Mol Cell. 20:881–890. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Jung H, Lee D, Lee J, Park D, Kim YJ, Park WY, Hong D, Park PJ and Lee E: Intron retention is a widespread mechanism of tumor-suppressor inactivation. Nat Genet. 47:1242–1248. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Goldstein L, Lee J, Gnad F, Klijn C, Schaub A, Reeder J, Daemen A, Bakalarski CE, Holcomb T, Shames DS, et al: Recurrent loss of NFE2L2 exon 2 is a mechanism for Nrf2 pathway activation in human cancers. Cell Rep. 16:2605–2617. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Kornblihtt AR: Epigenetics at the base of alternative splicing changes that promote colorectal cancer. J Clin Invest. 127:3281–3283. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Poulikakos PI, Persaud Y, Janakiraman M, Kong X, Ng C, Moriceau G, Shi H, Atefi M, Titz B, Gabay MT, et al: RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF (V600E). Nature. 480:387–390. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, Sussman R, Lanauze C, Ruella M, Gazzara MR, et al: Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 5:1282–1295. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Wang BD, Ceniccola K, Hwang S, Andrawis R, Horvath A, Freedman JA, Olender J, Knapp S, Ching T, Garmire L, et al: Alternative splicing promotes tumour aggressiveness and drug resistance in African American prostate cancer. Nat Commun. 8:159212017. View Article : Google Scholar : PubMed/NCBI

25 

Sebestyén E, Zawisza M and Eyras E: Detection of recurrent alternative splicing switches in tumor samples reveals novel signatures of cancer. Nucleic Acids Res. 43:1345–1356. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Trincado JL, Sebestyén E, Pagés A and Eyras E: The prognostic potential of alternative transcript isoforms across human tumors. Genome Med. 8:852016. View Article : Google Scholar : PubMed/NCBI

27 

Shen S, Wang Y, Wang C, Ying NW and Yi X: SURVIV for survival analysis of mRNA isoform variation. Nat Commun. 7:115482016. View Article : Google Scholar : PubMed/NCBI

28 

Parsons DW, Li M, Zhang X, Jones S, Leary RJ, Lin JC, Boca SM, Carter H, Samayoa J, Bettegowda C, et al: The genetic landscape of the childhood cancer medulloblastoma. Science. 331:435–439. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Ghigna C, De Toledo M, Bonomi S, Valacca C, Gallo S, Apicella M, Eperon I, Tazi J and Biamonti G: Pro-metastatic splicing of Ron proto-oncogene mRNA can be reversed: Therapeutic potential of bifunctional oligonucleotides and indole derivatives. RNA Biol. 7:495–503. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Lee CW and Abdelwahab O: Therapeutic targeting of splicing in cancer. Nat Med. 22:976–986. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Koh CM, Bezzi M, Low DH, Ang WX, Teo SX, Gay FP, Al-Haddawi M, Tan SY, Osato M, Sabò A, et al: MYC regulates the core pre-mRNA splicing machinery as an essential step in lymphomagenesis. Nature. 523:96–100. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Havens MA and Hastings ML: Splice-switching antisense oligonucleotides as therapeutic drugs. Nucleic Acids Res. 44:6549–6563. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Salton M and Misteli T: Small molecule modulators of Pre-mRNA splicing in cancer therapy. Trends Mol Med. 22:28–37. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Gao L, Xie ZC, Pang JS, Li TT and Chen G: A novel alternative splicing-based prediction model for uteri corpus endometrial carcinoma. Aging (Albany NY). 11:263–283. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Xiong Y, Deng Y, Wang K, Zhou H, Zheng X, Si L and Fu Z: Profiles of alternative splicing in colorectal cancer and their clinical significance: A study based on large-scale sequencing data. EBioMedicine. 36:183–195. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Huang ZG, He RQ and Mo ZN: Prognostic value and potential function of splicing events in prostate adenocarcinoma. Int J Oncol. 53:2473–2487. 2018.PubMed/NCBI

37 

Jusakul A, Cutcutache I, Yong CH, Lim JQ, Huang MN, Padmanabhan N, Nellore V, Kongpetch S, Ng AWT, Ng LM, et al: Whole-genome and epigenomic landscapes of etiologically distinct subtypes of cholangiocarcinoma. Cancer Discov. 7:1116–1135. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Supek F, Miñana B, Valcárcel J, Gabaldón T and Lehner B: Synonymous mutations frequently act as driver mutations in human cancers. Cell. 156:1324–1335. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Sterneweiler T and Sanford JR: Exon identity crisis: Disease-causing mutations that disrupt the splicing code. Genome Biol. 15:2012014. View Article : Google Scholar : PubMed/NCBI

40 

Diederichs S, Bartsch L, Berkmann JC, Fröse K, Heitmann J, Hoppe C, Iggena D, Jazmati D, Karschnia P, Linsenmeier M, et al: The dark matter of the cancer genome: Aberrations in regulatory elements, untranslated regions, splice sites, non-coding RNA and synonymous mutations. EMBO Mol Med. 8:442–457. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Singh S, Narayanan SP, Biswas K, Gupta A, Ahuja N, Yadav S, Panday RK, Samaiya A, Sharan SK and Shukla S: Intragenic DNA methylation and BORIS-mediated cancer-specific splicing contribute to the Warburg effect. Proc Natl Acad Sci USA. 114:11440–11445. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Gelfman S, Cohen N, Yearim A and Ast G: DNA-methylation effect on cotranscriptional splicing is dependent on GC architecture of the exon-intron structure. Genome Res. 23:789–799. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Shukla S, Kavak E, Gregory M, Imashimizu M, Shutinoski B, Kashlev M, Oberdoerffer P, Sandberg R and Oberdoerffer S: CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing. Nature. 479:74–79. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Yuan H, Li N, Fu D, Ren J, Hui J, Peng J, Liu Y, Qiu T, Jiang M, Pan Q, et al: Histone methyltransferase SETD2 modulates alternative splicing to inhibit intestinal tumorigenesis. J Clin Invest. 127:3375–3391. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Ding X, Liu S, Tian M, Zhang W, Zhu T, Li D, Wu J, Deng H, Jia Y, Xie W, et al: Activity-induced histone modifications govern Neurexin-1 mRNA splicing and memory preservation. Nat Neurosci. 20:690–699. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Sharma A, Nguyen H, Geng C, Hinman MN, Luo G and Lou H: Calcium-mediated histone modifications regulate alternative splicing in cardiomyocytes. Proc Natl Acad Sci USA. 111:E4920–E4928. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Kim S, Kim H, Fong N, Erickson B and Bentley DL: Pre-mRNA splicing is a determinant of histone H3K36 methylation. Proc Natl Acad Sci USA. 108:13564–13569. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Takehara T, Liu X, Fujimoto J, Friedman SL and Takahashi H: Expression and role of Bcl-xL in human hepatocellular carcinomas. Hepatology. 34:55–61. 2001. View Article : Google Scholar : PubMed/NCBI

49 

Boise LH, Gonzálezgarcía M, Postema CE, Ding L, Lindsten T, Turka LA, Mao X, Nuñez G and Thompson CB: bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death. Cell. 74:597–608. 1993. View Article : Google Scholar : PubMed/NCBI

50 

Bingle CD, Craig RW, Swales BM, Singleton V, Zhou P and Whyte MK: Exon skipping in Mcl-1 results in a Bcl-2 homology domain 3 only gene product that promotes cell death. J Biol Chem. 275:22136–22146. 2000. View Article : Google Scholar : PubMed/NCBI

51 

Bae J, Leo CP, Hsu SY and Hsueh AJ: MCL-1S, a splicing variant of the antiapoptotic BCL-2 family member MCL-1, encodes a proapoptotic protein possessing only the BH3 domain. J Biol Chem. 275:25255–25261. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Salton M, Kasprzak WK, Voss T, Shapiro BA, Poulikakos PI and Misteli T: Inhibition of vemurafenib-resistant melanoma by interference with pre-mRNA splicing. Nat Commun. 6:71032015. View Article : Google Scholar : PubMed/NCBI

53 

Chen K, Xiao H, Zeng J, Yu G, Zhou H, Huang C, Yao W, Xiao W, Hu J, Guan W, et al: Alternative splicing of EZH2 pre-mRNA by SF3B3 contributes to the tumorigenic potential of renal cancer. Clin Cancer Res. 23:3428–3441. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Luo C, Cheng Y, Liu Y, Chen L, Liu L, Wei N, Xie Z, Wu W and Feng Y: SRSF2 regulates alternative splicing to drive hepatocellular carcinoma development. Cancer Res. 77:1168–1178. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Calabretta S, Bielli P, Passacantilli I, Pilozzi E, Fendrich V, Capurso G, Fave GD and Sette C: Modulation of PKM alternative splicing by PTBP1 promotes gemcitabine resistance in pancreatic cancer cells. Oncogene. 35:2031–2039. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Li Y, Sun N, Lu Z, Sun S, Huang J, Chen Z and He J: Prognostic alternative mRNA splicing signature in non-small cell lung cancer. Cancer Lett. 393:40–51. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Zhu J, Chen Z and Yong L: Systematic profiling of alternative splicing signature reveals prognostic predictor for ovarian cancer. Gynecol Oncol. 148:368–374. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Miles S, Swift L and Leinster SJ: The dundee ready education environment measure (DREEM): A review of its adoption and use. Med Teach. 34:e620–e634. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Shi Y, Chen Z, Gao J, Wu S, Gao H and Feng G: Transcriptome-wide analysis of alternative mRNA splicing signature in the diagnosis and prognosis of stomach adenocarcinoma. Oncol Rep. 40:2014–2022. 2018.PubMed/NCBI

60 

Lin P, He RQ, Ma FC, Liang L, He Y, Yang H, Dang YW and Chen G: Systematic analysis of survival-associated alternative splicing signatures in gastrointestinal pan-adenocarcinomas. EBioMedicine. 34:46–60. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Ryan M, Wong WC, Brown R, Akbani R, Su X, Broom B, Melott J and Weinstein J: TCGASpliceSeq a compendium of alternative mRNA splicing in cancer. Nucleic Acids Res. 44:D1018–D1022. 2016. View Article : Google Scholar : PubMed/NCBI

62 

R Core Team. R, . A language and environment for statistical computingR Foundation for Statistical Computing; Vienna, Austria: 2012, ISBN 3-900051-07-0. http://www.R-project.org/

63 

Smidt N DJaMT: Guide to the contents of a Cochrane review and protocol. Cochrane handbook for systematic reviews of diagnostic test accuracy. 2011.

64 

Conway JR, Lex A and Gehlenborg N: UpSetR: An R package for the visualization of intersecting sets and their properties. Bioinformatics. 33:2938–2940. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Wu G, Dawson E, Duong A, Haw R and Stein L: ReactomeFIViz: A Cytoscape app for pathway and network-based data analysis. F1000Res. 3:1462014. View Article : Google Scholar : PubMed/NCBI

66 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

67 

Wickham H: ggplot2: Elegant Graphics for Data AnalysisSpringer-Verlag; New York, NY: pp. 77–186. 2016

68 

Heagerty PJ and Zheng Y: Survival model predictive accuracy and ROC curves. Biometrics. 61:92–105. 2005. View Article : Google Scholar : PubMed/NCBI

69 

Lee Y and Rio DC: Mechanisms and regulation of alternative Pre-mRNA splicing. Annu Rev Biochem. 84:291–323. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Piva F, Giulietti M, Burini AB and Principato G: SpliceAid 2: A database of human splicing factors expression data and RNA target motifs. Hum Mutat. 33:81–85. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Rahnemai-Azar AA, Weisbrod A, Dillhoff M, Schmidt C and Pawlik TM: Intrahepatic cholangiocarcinoma: Molecular markers for diagnosis and prognosis. Surg Oncol. 26:125–137. 2017. View Article : Google Scholar : PubMed/NCBI

72 

Khan SA, Davidson BR, Goldin RD, Heaton N, Karani J, Pereira SP, Rosenberg WM, Tait P, Taylor-Robinson SD, Thillainayagam AV, et al: Guidelines for the diagnosis and treatment of cholangiocarcinoma: An update. Gut. 61:1657–1669. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Pasquali C, Sperti C, D'Andrea AA, Costantino V, Filipponi C and Pedrazzoli S: CA50 as a serum marker for pancreatic carcinoma: Comparison with CA19-9. Eur J Cancer 30A. 1042–1043. 1994. View Article : Google Scholar

74 

Shan M, Tian Q and Zhang L: Serum CA50 levels in patients with cancers and other diseases. Prog Mol Biol Transl Sci. 162:187–198. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Dou H, Sun G and Zhang L: CA242 as a biomarker for pancreatic cancer and other diseases. Prog Mol Biol Transl Sci. 162:229–239. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Huang CK, Aihara A, Iwagami Y, Yu T, Carlson R, Koga H, Kim M, Zou J, Casulli S and Wands JR: Expression of transforming growth factor β1 promotes cholangiocarcinoma development and progression. Cancer Lett. 380:153–162. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, Zuo M, Zinner R, Hong D, Meric-Bernstam F, et al: Mutation profiling in cholangiocarcinoma: Prognostic and therapeutic implications. PLoS One. 9:e1153832014. View Article : Google Scholar : PubMed/NCBI

78 

Dana P, Kariya R, Vaeteewoottacharn K, Sawanyawisuth K, Seubwai W, Matsuda K, Okada S and Wongkham S: Upregulation of CD147 promotes metastasis of cholangiocarcinoma by modulating the Epithelial-to-Mesenchymal transitional process. Oncol Res. 25:1047–1059. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Schweitzer N and Vogel A: Systemic therapy of cholangiocarcinoma: From chemotherapy to targeted therapies. Best Pract Res Clin Gastroenterol. 29:345–353. 2015. View Article : Google Scholar : PubMed/NCBI

80 

Kamlua S, Patrakitkomjorn S, Jearanaikoon P, Menheniott TR, Giraud AS and Limpaiboon T: A novel TFF2 splice variant (ΔEX2TFF2) correlates with longer overall survival time in cholangiocarcinoma. Oncol Rep. 27:1207–1212. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Nutthasirikul N, Limpaiboon T, Leelayuwat C, Patrakitkomjorn S and Jearanaikoon P: Ratio disruption of the ∆133p53 and TAp53 isoform equilibrium correlates with poor clinical outcome in intrahepatic cholangiocarcinoma. Int J Oncol. 42:1181–1188. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Comino-Mendez I, Leandro-Garcia LJ, Montoya G, Inglada-Pérez L, de Cubas AA, Currás-Freixes M, Tysoe C, Izatt L, Letón R, Gómez-Graña Á, et al: Functional and in silico assessment of MAX variants of unknown significance. J Mol Med (Berl). 93:1247–1255. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Huang X, Wu Z, Mei Y and Wu M: XIAP inhibits autophagy via XIAP-Mdm2-p53 signalling. EMBO J. 32:2204–2216. 2013. View Article : Google Scholar : PubMed/NCBI

84 

Li Z, Tuteja G, Schug J and Kaestner KH: Foxa1 and Foxa2 are essential for sexual dimorphism in liver cancer. Cell. 148:72–83. 2012. View Article : Google Scholar : PubMed/NCBI

85 

McNeely S, Beckmann R and Bence Lin AK: CHEK again: Revisiting the development of CHK1 inhibitors for cancer therapy. Pharmacol Ther. 142:1–10. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Mo J, Zhang D and Yang R: MicroRNA-195 regulates proliferation, migration, angiogenesis and autophagy of endothelial progenitor cells by targeting GABARAPL1. Biosci Rep. 36(pii): e003962016. View Article : Google Scholar : PubMed/NCBI

87 

Wagner EF and Nebreda AR: Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 9:537–549. 2009. View Article : Google Scholar : PubMed/NCBI

88 

Eswaran J, Horvath A, Godbole S, Reddy SD, Mudvari P, Ohshiro K, Cyanam D, Nair S, Fuqua SA, Polyak K, et al: RNA sequencing of cancer reveals novel splicing alterations. Sci Rep. 3:16892013. View Article : Google Scholar : PubMed/NCBI

89 

Suo C, Hrydziuszko O, Lee D, Pramana S, Saputra D, Joshi H, Calza S and Pawitan Y: Integration of somatic mutation, expression and functional data reveals potential driver genes predictive of breast cancer survival. Bioinformatics. 31:2607–2613. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Yosudjai J, Inpad C, Chomwong S, Dana P, Sawanyawisuth K, Phimsen S, Wongkham S, Jirawatnotai S and Kaewkong W: An aberrantly spliced isoform of anterior gradient-2, AGR2vH promotes migration and invasion of cholangiocarcinoma cell. Biomed Pharmacother. 107:109–116. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Mcneely S, Beckmann R and Bence Lin AK: CHEK again: Revisiting the development of CHK1 inhibitors for cancer therapy. Pharmacol Ther. 142:1–10. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Zhang H, Yang T, Wu M and Shen F: Intrahepatic cholangiocarcinoma: Epidemiology, risk factors, diagnosis and surgical management. Cancer Lett. 379:198–205. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Goere D, Wagholikar GD, Pessaux P, Carrère N, Sibert A, Vilgrain V, Sauvanet A and Belghiti J: Utility of staging laparoscopy in subsets of biliary cancers: Laparoscopy is a powerful diagnostic tool in patients with intrahepatic and gallbladder carcinoma. Surg Endosc. 20:721–725. 2006. View Article : Google Scholar : PubMed/NCBI

94 

He RQ, Zhou XG, Yi QY, Deng CW, Gao JM, Chen G and Wang QY: Prognostic signature of alternative splicing events in bladder urothelial carcinoma based on spliceseq data from 317 cases. Cell Physiol Biochem. 48:1355–1368. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Lin P, He RQ, Huang ZG, Zhang R, Wu HY, Shi L, Li XJ, Li Q, Chen G, Yang H and He Y: Role of global aberrant alternative splicing events in papillary thyroid cancer prognosis. Aging (Albany NY). 11:2082–2097. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Zhang D, Duan Y, Cun J and Yang Q: Identification of prognostic alternative splicing signature in breast carcinoma. Front Genet. 10:2782019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 18 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu HY, Wei Y, Liu LM, Chen ZB, Hu QP and Pan SL: Construction of a model to predict the prognosis of patients with cholangiocarcinoma using alternative splicing events. Oncol Lett 18: 4677-4690, 2019
APA
Wu, H., Wei, Y., Liu, L., Chen, Z., Hu, Q., & Pan, S. (2019). Construction of a model to predict the prognosis of patients with cholangiocarcinoma using alternative splicing events. Oncology Letters, 18, 4677-4690. https://doi.org/10.3892/ol.2019.10838
MLA
Wu, H., Wei, Y., Liu, L., Chen, Z., Hu, Q., Pan, S."Construction of a model to predict the prognosis of patients with cholangiocarcinoma using alternative splicing events". Oncology Letters 18.5 (2019): 4677-4690.
Chicago
Wu, H., Wei, Y., Liu, L., Chen, Z., Hu, Q., Pan, S."Construction of a model to predict the prognosis of patients with cholangiocarcinoma using alternative splicing events". Oncology Letters 18, no. 5 (2019): 4677-4690. https://doi.org/10.3892/ol.2019.10838