IMR Press / FBL / Volume 28 / Issue 4 / DOI: 10.31083/j.fbl2804075
Open Access Original Research
Prognostic Significance of DNA Topoisomerase II Alpha (TOP2A) in Cholangiocarcinoma
Show Less
1 Division of Gastroenterology & General Surgery, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
2 Department of Medical Technology, Chung Hwa University of Medical Technology, 717 Tainan, Taiwan
3 Institute of Biomedical Sciences, National Sun Yat-sen University, 804 Kaohsiung, Taiwan
4 Department of Pharmacology, School of Medicine, China Medical University, 404333 Taichung, Taiwan
5 Department of Clinical Pathology, Chi Mei Medical Center, 710 Tainan, Taiwan
6 Division of Urology, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
7 Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, 711 Tainan, Taiwan
8 Division of Colon and Rectal Surgery, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
9 Institute of Precision Medicine, National Sun Yat-sen University, 804 Kaohsiung, Taiwan
10 Department of Medical Research, Chi Mei Medical Center, 710 Tainan, Taiwan
11 National Institute of Cancer Research, National Health Research Institutes, 704 Tainan, Taiwan
12 Trans-Omic Laboratory for Precision Medicine, Chi Mei Medical Center, 710 Tainan, Taiwan
13 Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, 71004 Tainan, Taiwan
14 College of Pharmacy and Science, Chia Nan University, 71710 Tainan, Taiwan
*Correspondence: beethovan@gmail.com (Yu-Hsuan Kuo)
Front. Biosci. (Landmark Ed) 2023, 28(4), 75; https://doi.org/10.31083/j.fbl2804075
Submitted: 24 December 2022 | Revised: 3 March 2023 | Accepted: 27 March 2023 | Published: 19 April 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Cholangiocarcinoma (CCA) is a malignant tumor with an increasing incidence worldwide. Although radiation therapy has improved the therapeutic efficiency of CCA treatment, differential expression of genes among cholangiocarcinoma subtypes has been revealed through precise sequencing. However, no specific molecular therapeutic targets or biomarkers have been figured out for use in precision medicine, and the exact mechanism by which antitumorigenic effects occur is still unclear. Therefore, it is necessary to conduct further studies on the development and mechanisms associated with CCA. Methods: We examined the clinical data and pathological features of patients with cholangiocarcinomas. We investigated the associations between DNA Topoisomerase II Alpha (TOP2A) expression and patient outcomes, such as metastasis-free survival (MFS) and disease-specific survival (DSS), as well as clinical characteristics and pathological results. Results: TOP2A expression was shown to be upregulated in CCA tissue sections by immunohistochemistry staining and data mining. Moreover, we observed that the TOP2A expression correlated with clinical features, such as the primary tumor stage, histological variants, and patients with hepatitis. Furthermore, high expression of TOP2A was associated with worse survival outcomes in terms of the overall survival (p < 0.0001), disease-specific survival (p < 0.0001), and metastasis-free survival (p < 0.0001) compared with patients in the low TOP2A expression group. This indicates that a high level of TOP2A expression is related to an unfavorable prognosis. Conclusions: Our results show that TOP2A is highly expressed in CCA tissues, and its upregulation is correlated with the primary disease stage and poor prognosis significantly. Consequently, TOP2A is a prognostic biomarker and a novel therapeutic target for the treatment of CCA.

Keywords
cholangiocarcinoma
DNA topoisomerase (ATP-hydrolyzing activity) activity
topoisomerase II α (TOP2A)
prognostic biomarker
1. Introduction

Cholangiocarcinoma, which is a type of cancer that affects the epithelial cells, can arise from various locations of the bile duct [1]. Depending on its location, cholangiocarcinoma can be classified as intrahepatic, perihilar, or distal. Intrahepatic cholangiocarcinoma starts from the secondary bile ducts located close to the liver, while perihilar cholangiocarcinoma occurs between the cystic duct and the secondary bile ducts. Distal cholangiocarcinoma is found between the ampulla and the origin of the cystic duct [2, 3]. According to previous studies, the risk factors for cholangiocarcinoma (CCA) are sporadic and are correlated with geographic variations [4, 5]. For example, infection with hepatobiliary flukes is associated with chronic inflammation and is also correlated with the development of CCA in individuals from Southeast Asia [6]; Hepatolithiasis is also a risk factor of developing CCA (mainly intrahepatic cholangiocarcioma (iCCA)) in Asians [5]. In the West, primary sclerosing cholangitis (PSC) carries the greatest risk for patients with CCA [7, 8]. Accordingly, CCA is diagnosed within two years of developing PSC. Although risk factors for CCA development, such as smoking and alcohol, have been identified in PSC patients, direct research data are lacking [9].

The TOP2A gene is responsible for regulating the chromosome bond pathway by encoding DNA topoisomerase, which in turn controls the topological state of DNA transcription and replication [10]. Studies have linked the expression of TOP2A with various levels of cancer progression and the development of nasopharyngeal carcinoma [11], breast cancer [12], adrenal cancer [13], and endometrial cancer [14]. Abnormal expression of TOP2A is often linked to irregular cell proliferation, while decreased expression of the gene can lead to changes in several molecular signaling pathways, such as the β-catenin pathway in pancreatic cancer and EPK/AKT in colon cancer [15, 16, 17]. Furthermore, TOP2A is considered a prognostic factor or a driver gene that affects the survival of patients with different types of cancer [16].

In recent years, few studies involving genetic factors (tumor suppressors and oncogenes) and epigenetic alterations associated with CCA progression have been con-ducted [18]. However, there is no definitive result to identify the major oncogenes or suppressor genes associated with CCA development. In this study, we explore the molecular role of TOP2A in CCA and try to identify the biological functions of TOP2A in CCA progression.

2. Material and Methods
2.1 Analysis of Published Transcriptomic Datasets

The gene expression profile of cholangiocarcinoma (GSE26566) was investigated through the Gene Expression Omnibus (GEO) database. Significant gene expression changes were identified, particularly those in molecular pathways involving DNA topoisomerase (ATP-hydrolyzing activity) activity in Gene Ontology (GO:2000371), through comparative and functional analyses. The databases were analyzed, and focus was placed on p-values of <0.01 and log2-transformed expression fold changes of >0.1.

2.2 Cholangiocarcinoma Patient Tissues

The Chi Mei Medical Center enrolled 182 iCCA patients who underwent curative surgery between 1990 and 2010. Patients with metastatic disease or nodal metastases were excluded. This study was authorized by the Institutional Review Board (IRB) of The Chi Mei Medical Center with approval number 09912003. All participants provided their informed consent. Patients’ demographics and clinical details was collected retrospectively, including pathological characteristics, oncological survival follow-up, and cause of death. The Tumor, Node, Metastasis (TNM) system created by the eighth edition American Joint Committee on Cancer (AJCC) in 2017 was used to measure the tumor stage. Two pathologists examined the tumor samples.

2.3 Immunohistochemistry (IHC) Staining

The formalin-fixed tissues were embedded in paraffin and sectioned into 10 μm sections. Subsequently, immunohistochemical (IHC) staining of TOP2A was conducted using a primary monoclonal antibody directed towards TOP2A (1:200, EP1102Y, Epitomics, Cambridge, UK) for a duration of 1 hour. To detect the primary antibodies, the ChemMate EnVision kit (DAKO, K5001, Carpinteria, CA, USA) was utilized. The secondary antibody was incubated with the slides for 30 minutes and then developed with 3,3-diaminobenzidine for 5 minutes. Subsequently, the slides were counterstained with hematoxylin. Cholangiocarcinoma expressing TOP2A was utilized as a positive control, while rabbit serum IgG replaced the primary antibody as a negative control. Two pathologists used the formula H-score = SPi (i + 1) to compute the H-score, where Pi stands for the percentage of stained tumor cells in different intensities ranging from 0% to 100% and i for the degree of staining (0 to 3+). The two pathologists evaluated the slides concurrently and decided on an H-score in case there were any scoring disagreements. Based on the median H-score, the immunostaining was categorized as having low or high expression levels.

2.4 Real-Time Reverse-Transcription Polymerase Chain Reaction (RT-PCR)

The relative expression and knockdown efficacy of the TOP2A gene in iCCA cell lines were determined using RT-PCR, as previously published [19]. Total RNA was collected from iCCA cell lines, and real-time RT-PCR was employed to determine the transcription level of TOP2A. The mRNA abundance of TOP2A (Hs01032137_mL) was assessed using predesigned TaqMan assay reagents (Cat. No. Hs01032137_mL, Applied Biosystems, Waltham, MA, USA) and the ABI StepOnePlusTM system (Applied Biosystems, Waltham, MA, USA), with POLR2A (Hs01108291_mL) used as the internal control for normalization.

2.5 Cell Culture

The SNU1079 and SNU1196 cell lines were procured from a cell bank based in Seoul, South Korea. Initially, these cell lines were cultured in ACL-4 medium supplemented with 5% heat-inactivated fetal bovine serum. After establishment, the cell cultures were maintained in RPMI 1640 medium and supplemented with 10% heat-inactivated fetal bovine serum. These cells were cultured under controlled conditions in a humidified incubator at 37 °C in the presence of 5% CO2 and 95% air as previously described [20]. On the other hand, the Huh28 and HuCCT1 cell lines were obtained from a cell bank in Osaka, Japan. These cell lines were cultured in either Roswell Park Memorial Institute 1640 medium (HuCCT1) or minimal essential medium (Huh28), which were both supplemented with 100 U/mL penicillin, 100 μg/mL streptomycin, and 10% fetal bovine serum as previously reported [21]. All the cell lines were authenticated by short tandem repeat genotyping (ThermoFisher, Waltham, MA, USA), periodically confirmed to be mycoplasma free using PlasmoTest (Invivogen, San Diego, CA, USA).

2.6 RNA Interference

The SNU1196 and Huh28 cell lines were modified to create stable TOP2A-silenced clones using lentiviral vectors pLKO.1-shLacZ (TRCN0000072223: 5′-TGTTCGCATTATCCGAACCAT-3′) and pLKO.1-shTOP2A (#1: TRCN0000049280: 5′-GCTCCAAATCAA TATGTGATT-3′; #2: TRCN0000049278: 5′-GCCCAA GTGTTCTTTAGCTTT-3′), which were obtained from the Taiwan National RNAi Core Facility in Taipei, Taiwan. These cell lines initially had high TOP2A expression that was reduced by shRNAs against TOP2A (shTOP2A). To generate viruses for the modification, HEK293 cells were transfected with the three vectors mentioned above, using Lipofectamine 2000 from Thermo Fisher Scientific in Waltham, MA, USA, as previously described [19].

2.7 Western Blotting

Primary antibodies against TOP2A (clone AA6, 1:500; Millipore, Beverly, MA, USA) were utilized, and a previously reported western blotting technique was utilized to assess TOP2A expression and the effect of TOP2Aknockdown in SNU1196 and Huh28 cell lines. Glyceraldehyde 3-phosphate dehydrogenase (GADPH) was used as a control for protein loading (6C5, 1:10,000; Millipore, Beverly, MA, USA) [22]. To immobilize the protein, cell lysates containing 25 μg of protein were separated on a 4% to 12% gradient NuPAGE gel (Invitrogen, Carlsbad, CA, USA) and transferred to polyvinylidene difluoride membranes (Amersham Biosciences, Buckinghamshire, UK). To prevent non-specific binding, membranes were blocked with TBST buffer and 5% skimmed milk at room temperature for one hour. Primary antibodies were then exposed to the membranes at 4 °C overnight. After that, the membranes were incubated with the secondary antibody for 1.5 hours at room temperature, and proteins were identified using a chemiluminescence system (Amersham Biosciences, Buckinghamshire, UK).

2.8 Bromodeoxyuridine (BrdU) Assay to Assess DNA Synthesis

The enzyme-linked immunosorbent assay (ELISA)-based and colorimetric bromodeoxyuridine (BrdU) assay (Roche Holding AG, Basel, Switzerland) was utilized to quantify DNA synthesis. At 24, 48, and 72 hours, the amount of DNA synthesis was measured in the TOP2A-knockdown or shLacA control SNU1196 and Huh28 cell lines. After three hours of BrdU incubation at 37 °C and 5% CO2, the labeling medium was removed and the cells were fixed before being incubated with an anti-BrdU-POD solution. Using an ELISA reader (Promega Corp., Madison, WI, USA), the absorbance at 450 nm was measured, with the reference absorbance set to 690 nm.

2.9 Migration and Invasion Assays

The experimental protocol for cell migration and invasion was carried out in accordance with a previously documented procedure [22]. For the cell invasion experiment, the 24-well Collagen-based Cell Invasion Assay from Millipore, Beverly, MA, USA, and Falcon HTS FluoroBlok 24-well inserts from BD Biosciences, Franklin Lakes, NJ, USA, were used. The inserts were rehydrated using serum-free medium and then placed in the upper chamber, which contained a serum-free suspension with an equal number of cells. Over a 12- to 24-hour incubation period, the cells were allowed to migrate towards the lower chamber, which contained a medium with 10% fetal bovine serum. Following removal of the non-invading cells from the upper chamber, the invading cells were stained, lysed in extraction buffer, and then transferred to 96-well plates for 560 nm colorimetric readings.

2.10 Functional Annotation

In order to determine the unknown functions of TOP2A in iCCA, the transcription level of TOP2A and its coexpressed genes contained in the cholangiocarcinoma dataset (n = 51, Firehose Legacy, TCGA) were analyzed to establish correlations. Subsequently, the top 200 differentially expressed genes with positive or negative correlations with TOP2A were selected for functional annotation using the Gene Ontology (GO) classification system and rated by fold enrichment.

2.11 Statistical Analysis

The statistical analyses were conducted using SPSS software (version 28, IBM Corp., Chicago, IL, USA). The Chi-square test was utilized to evaluate the association between TOP2A expression stage and clinicopathologic characteristics. Local recurrence-free survival (LRFS), metastasis-free survival (MFS), and disease-specific survival (DSS) were calculated from the beginning of therapy to the date of the event, and the latest follow-up date was noted for patients who were lost to follow-up during the study period. Kaplan-Meier analysis was employed to generate survival curves, and log-rank tests were utilized to identify prognostic differences between groups. All analyses were performed using two-sided significance tests, and a p-value less than 0.05 was considered significant.

3. Results
3.1 TOP2A Expression was Associated with DNA Topoisomerase Activity in CCA

We mined the public CCA transcriptome dataset (GSE26566) and compared the results with DNA topoisomerase (ATP-hydrolyzing activity) activity (GO:2000371). We discovered that five genes were significantly associated with DNA topoisomerase (ATP-hydrolyzing activity) activity (Table 1), and TOP2Awas found to be a high-ranking differently expressed candidate gene that showed a significant difference (p < 0.0001) and a log-ratio of >0.1 (Table 1). Moreover, the heat map data mining results also showed that expression of DNA topoisomerase-related genes differed between CCA tissue and the surrounding liver and non-tumor biliary epithelium tissues, and TOP2A was shown to be highly expressed in CCA patients (Fig. 1). The above data demonstrate that TOP2A might play a critical role in cancer progression in CCA patients.

Table 1.Summary of the alterations of gene associated with DNA topoisomerase (ATP-hydrolyzing activity) activity (GO:2000371) in cholangiocarcinoma (GSE26566).
Probe CCA vs Non-tumor# CCA vs Normal intrahepatic bile duct& Gene Symbol Molecular function Biological process
log ratio p-value log ratio p-value
ILMN_1686097 1.7717 <0.0001* 1.7928 <0.0001* TOP2A DNA topoisomerase (ATP-hydrolyzing) activity, protein homodimerization activity, histone deacetylase binding, protein heterodimerization activity, ATP binding, chromatin binding, DNA-dependent ATPase activity, nucleotide binding, protein C-terminus binding, ubiquitin binding, protein kinase C binding, drug binding DNA ligation, DNA topological change, positive regulation of apoptosis, phosphoinositide-mediated signaling, DNA repair, chromosome segregation, DNA replication, apoptotic chromosome condensation
ILMN_1777663 0.3019 0.0018* 0.339 0.1852 TOP2B DNA topoisomerase (ATP-hydrolyzing) activity, histone deacetylase binding, protein heterodimerization activity, ATP binding, chromatin binding, nucleotide binding, protein C-terminus binding, protein kinase C binding DNA topological change
ILMN_1659651 0.2556 <0.0001* 0.6474 <0.0001* TOP1MT DNA topoisomerase (ATP-hydrolyzing) activity, DNA topoisomerase type I activity DNA topological change
ILMN_1796508 0.185 0.0010* 0.4342 0.0122*
ILMN_1735572 0.0875 0.1265 0.1408 0.3608 TOP1 DNA topoisomerase (ATP-hydrolyzing) activity, chromatin binding, DNA topoisomerase type I activity, protein binding DNA topological change
ILMN_1687970 –0.1256 0.0006* –0.1417 0.1639 SPO11 DNA topoisomerase (ATP-hydrolyzing) activity, hydrolase activity, DNA binding, ATP binding DNA topological change, female gamete generation, spermatogenesis, meiotic recombination, meiosis
ILMN_1796655 –0.1309 0.0009* –0.0693 0.5052 DNA topological change, female gamete generation, spermatogenesis, meiotic recombination, meiosis
#, Comparing cholangiocarcinoma (CCA, n = 104) to surrounding liver (n = 59) and normal intrahepatic bile duct (n = 6); &, Comparing cholangiocarcinoma (CCA, n = 104) to normal intrahepatic bile duct (n = 6); * statistically significant.
Fig. 1.

Data mining of genes expression compared between cholangiocarcinoma, surrounding liver and non-tumor biliary epithelium from the public domain (GSE26566).

3.2 The Association between TOP2A Expression and Important Clinic Pathological Parameters in CCA

Our previous results indicate that high expression of TOP2A might be correlated with CCA progression. We further analyzed associations between TOP2A expression and the clinicopathologic characteristics of CCA patients. We included data from 182 iCCA patients, 108 males and 74 females, where 107 patients were older than 65 and 75 patients were younger than 65. These cases were classified as 105 of the large duct type and 77 of the small duct type based on histological variants; 61 cases were classified as well-differentiated, 66 as moderately differentiated, and 55 as poorly differentiated according to histological grades. A total of 163 cases were classified as R0 and 19 cases were classified as R1 based on the surgical margin, and in terms of the primary tumor size, 87 cases were classified as T1, 61 as T2, and 34 as T3 (Table 2). Taken together, our results indicate that TOP2A overexpression is highly correlated with the co-occurrence of hepatitis (p-value = 0.002), the primary tumor size (p-value = 0.001), and the histological variants (p-value = 0.002) of clinicopathological parameters in cholangiocarcinoma patients with iCCA (Table 2). Moreover, we determined that TOP2A is overexpressed in cholangiocarcinoma patients (Fig. 1). We confirmed this by IHC staining of cholangiocarcinoma patient tissue sections (Fig. 2). Moreover, we calculated the immunohistochemical staining H-score which showed significantly higher in iCCA cells than hepatocytes and cholangiocytes (Fig. 3). These data suggest that TOP2A expression is significantly associated with clinicopathological variables in iCCA patients.

Table 2.Correlations between TOP2A expression and other important clinicopathological parameters in primary localized IHCC.
Parameter Category Case No. TOP2A expression p-value
Low High
Gender Male 108 51 57 0.365
Female 74 40 34
Age (years) <65 107 49 58 0.175
65 75 42 33
Hepatitis Hepatitis B 72 38 34 0.002*
Hepatitis C 29 22 7
Non-B, non-C 81 31 50
Intrahepatic lithiasis Not identified 102 56 46 0.135
Present 80 35 45
Surgical margin R0 163 84 79 0.225
R1 19 7 12
Primary tumor (T) T1 87 56 21 0.001*
T2 61 24 37
T3 34 11 23
Histological variants Large duct type 105 42 63 0.002*
Small duct type 77 49 28
Histological grade Well differentiated 61 31 30 0.887
Moderately differentiated 66 34 32
Poorly differentiated 55 26 29
* Statistically significant.
Fig. 2.

Identify the TOP2A expression in iCCA tissues. (A) In low-stage tumor tissues, TOP2A positive cells are weak detected (a–d). (B) In high-stage tumor tissues, tumor cells showed positive TOP2A staining (e–h). (C) A comparison of H-score showed significantly higher TOP2A expression in iCCA cells than hepatocytes and cholangiocytes (N = 182 in each group).

Fig. 3.

Kaplan–Meier survival curves in the CCA patients according to surgical margin, primary tumor staging and TOP2A expression. R0 Resection, pT1 stage and low TOP2A expression correlated with better overall survival (A,B,C), better disease-specific survival (D,E,F), better local recurrence-free survival (G,H,I) and better metastasis-free survival (J,K,L).

3.3 Identified TOP2A in Comparison with the Patient Survival Analysis

To determine whether TOP2A expression is correlated with CCA patients’ survival outcomes, we analyzed the association between TOP2A expression and the overall survival rate, disease-specific survival rate, local recurrence-free survival rate, and metastasis-free survival rate in CCA patients. For overall survival, an R0 stage surgical margin, T1 stage primary tumor, and low TOP2A expression (Fig. 3A–C) were associated with better outcomes in both the univariate and multivariate analyses (Table 3). The dis-ease-specific survival endpoints analysis showed the same results whereby R0 stage, T1 stage, and low TOP2A expression patients had better outcomes (Table 3) (Fig. 3D–F) than other patients. Moreover, patients with a T1 stage primary tumor, large duct type histological variant, and histological low grade were shown to have better outcomes in the univariate analysis of local recurrence-free survival (Table 4). Importantly, an R0 stage surgical margin, T1 stage primary tumor, and low TOP2A expression were all significantly associated with better outcomes (Fig. 3G–I) in both the univariate analysis and the multivariate analysis of local recurrence-free survival and metastasis-free survival (Table 4) (Fig. 3J–L).

Table 3.Univariate log-rank and multivariate analyses for overall and disease-specific survivals in primary localized IHCC.
Parameter Category Case No. Overall survival Disease-specific survival
Univariate analysis Multivariate analysis Univariate analysis Multivariate analysis
No. of event p-value R.R. 95% CI p-value No. of event p-value R.R. 95% CI p-value
Gender Male 108 50 0.0254* 1 - 0.087 9 0.0072* 1 - 0.044*
Female 74 21 1.569 0.937–2.629 - 32 2.158 1.021–4.558 -
Age (years) <65 107 37 0.2626 - - - 28 0.2125 - - -
65 75 34 - - - 13 - - -
Hepatitis Hepatitis B 72 32 0.2379 - - - 16 0.4561 - - -
Hepatitis C 29 8 - - - 19 - - -
Non-B, non-C 81 31 - - - 6 - - -
Intrahepatic lithiasis Not identified 102 36 0.2831 - - - 19 0.1613 - - -
Present 80 35 - - - 22 - - -
Surgical margin R0 163 59 <0.0001* 1 - 0.002* 31 <0.0001* 1 - <0.001*
R1 19 12 2.913 1.466–5.789 10 4.962 2.196–11.213
Primary tumor (T) T1 87 25 0.0001* 1 - 0.037* 9 <0.0001* 1 - 0.020*
T2 61 27 1.608 0.920–2.812 - 19 2.769 1.234–6.211 -
T3 34 19 2.317 1.205–4.455 - 13 3.281 1.321–8.153 -
Histological variants Large duct type 105 43 0.4281 - - - 27 0.1984 - - -
Small duct type 77 28 - - - 14 - - -
Histological grade (Differentiation) Well 61 20 0.1663 - - - 12 0.3881 - - -
Moderately 66 28 - - - 16 - - -
Poorly 55 23 - - - 13 - - -
TOP2A Exp. Low expression 91 28 0.0011 1 - 0.036*- 10 <0.0001* 1 - 0.002*-
High expression 91 43 1.714 1.035–2.838 - 31 1.929 1.198–3.886 -
* Statistically significant.
Table 4.Univariate log-rank and multivariate analyses for local recurrence-free and metastasis-free survivals in primary localized IHCC.
Parameter Category Case No. Local recurrence-free survival Metastasis-free survival
Univariate analysis Multivariate analysis Univariate analysis Multivariate analysis
No. of event p-value R.R. 95% CI p-value No. of event p-value R.R. 95% CI p-value
Gender Male 108 54 0.2170 - - - 21 0.1008 - - -
Female 74 31 - - - 44 - - -
Age (years) <65 107 55 0.2993 - - - 42 0.2936 - - -
65 75 30 - - - 23 - - -
Hepatitis Hepatitis B 72 33 0.7333 - - - 26 0.8762 - - -
Hepatitis C 29 13 - - - 11 - - -
Non-B, non-C 81 39 - - - 28 - - -
Intrahepatic lithiasis Not identified 102 41 0.0551 - - - 31 0.1000 - - -
Present 80 44 - - - 34 - - -
Surgical margin R0 163 71 <0.0001* 1 - 0.010* 54 <0.0001* 1 0.009*
R1 19 14 2.702 1/294–5.640 11 2.674 1.306–5.474
Primary tumor (T) T1 87 28 <0.0001* 1 - 0.051 21 <0.0001* 1 - 0.046*
T2 61 32 1.741 0.961–3.154 26 1.759 0.978–3.164
T3 34 25 1.899 0.958–3.764 18 1.912 0.965–3.788
Histological variants Large duct type 105 58 0.0085* 1 - 0.988 43 0.0759 - - -
Small duct type 77 27 1.108 0.592–1.751 22 - - -
Histological grade (Differentiation) Well 61 28 0.0299* 1 - 0.829 22 0.1794 - - -
Moderately 66 27 1.154 0.630–2.114 22 - - -
Poorly 55 30 1.208 0.638–2.283 21 - - -
TOP2A Exp. Low expression 91 15 <0.0001* 1 - <0.001 14 <0.0001* 1 - <0.001
High expression 91 70 4.658 2.481–8.745 51 4.695 2.514–8.768
* Statistically significant.

Taken together, our results indicate that TOP2A is highly expressed in CCA patients, and high TOP2A expression is correlated with poor prognosis and notably worse overall survival (p = 0.0011, Fig. 3C), disease-specific survival (p < 0.0001, Fig. 3F), local recurrence-free survival (p < 0.0001, Fig. 3I), and metastasis-free survival (p < 0.0001, Fig. 3L) rates.

3.4 Bioinformatic Analysis and Functional Prediction of TOP2A

To determine the unknown functions of TOP2A in IHCC, a set of the top 200 differentially expressed genes with positive (Supplementary Table 1) or negative correlations (Supplementary Table 2) with TOP2A were downloaded from the cholangiocarcinoma dataset (n = 51, Firehose Legacy, TCGA). These genes were then utilized for a functional annotation analysis using the Gene Ontology (GO) classification system. The biological processes most positively correlated with TOP2A were positive regulation of DNA endoreduplication (GO: 0032877, fold enrichment: >100), cell cycle DNA replication maintenance of fidelity (GO: 1902298, fold enrichment: >100), and mitotic DNA replication maintenance of fidelity (GO: 1990505, fold enrichment: >100) (Fig. 4A). DNA repair protein RAD51 homolog 1 (RAD51) and breast cancer type 2 susceptibility protein (BRCA2) genes, involved in most of the biological processes mentioned above, were also identified. As for molecular functions, the most positively correlated term with TOP2A was D-loop DNA binding (GO: 0062037, fold enrichment: 60.85) (Fig. 4B), and the RAD51-associated protein 1 (RAD51AP1) gene was identified.

Fig. 4.

The characteristic GO terms enriched in TOP2A upregulation. The top 200 differentially expressed genes with positive relationship to TOP2A were annotated utilizing the GO classification system depending on (A) biological processes and (B) molecular functions and were rated by fold enrichment.

3.5 TOP2A Promotes the Cell Proliferation, Migration, and Invasion of CCA Cell Lines

In order to assess the impact of TOP2A, we initially measured the levels of endogenous TOP2A expression in four different cholangiocarcinoma cell lines. We found that SNU1196 and Huh28 cells had the highest amounts of TOP2A transcripts and protein expression (Fig. 5A). We then used short hairpin RNA (shRNA) to successfully knock down TOP2A in both SNU1196 (Fig. 5B, left) and Huh28 (Fig. 5B, right) cell lines. TOP2A-silenced SNU1196 (Fig. 5E, left) and Huh28 (Fig. 5E, right) cells had considerably reduced proliferation (viability). We also looked at TOP2A’s role in cholangiocarcinoma cell migration and invasion. TOP2A knockdown significantly reduced SNU1196 (Fig. 5C, left and Fig. 5D, left) and Huh28 (Fig. 5C, right and Fig. 5D, right) cell migratory and invasive abilities.

Fig. 5.

TOP2A expression promotes the growth of CCA cells in vitro. (A) SNU1196 and Huh28 cells had the highest amounts of TOP2A mRNA and protein expression among four CCA cell lines. (B) The two cell lines with high endogenous TOP2A expression were stably silenced by a lentiviral vector bearing one of the two clones of TOP2A shRNA with different sequences for both SNU1196 (left panel) and Huh28 (right panel) cells. (C) Cell migration ability was significantly reduced in TOP2A- knockdown SNU1196 and Huh28 cell lines. (D) Cell invasion ability was significantly reduced in TOP2A- knockdown SNU1196 and Huh28 cell lines. (E) Using an ELISA-based colorimetric assay to assess the rate of BrdU uptake, cell proliferation was significantly reduced in TOP2A-knockdown SNU1196 and Huh28 cell lines (* p < 0.05, ** p < 0.01).

4. Discussion

Cholangiocarcinoma is a heterogeneous group of tumors that initiate from a number of cells from the biliary tree [23]. According to previous research, the risk and the molecular mechanisms associated with CCA pathogenesis involve inflammation and cholestasis or CCA development [24]. Accordingly, it has been observed that CCA patients have increased total serum bilirubin, alkaline phosphatase, and gamma-glutamyl transpeptidase levels or obstructive jaundice [25, 26]. Although no specific biomarkers for CCA have been identified, some references indicate that Carbohydrate antigen 19-9 (CA 19-9) could be used as be a marker for the detection of CCA, since CA 19-9 levels >100 U/mL have been reported to be a sensitive method for diagnosing CCA patients [27]. However, markers such as CA 19-9 can also be detected in gastric, pancreatic, colorectal, and gynecologic cancer patients or in individuals with other diseases, such as cholestasis, liver injury, and benign biliary obstruction [27, 28, 29]. Although researchers have explored the risks [30, 31, 32] and therapeutic methods [33, 34, 35] associated with CCA and have explored imaging diagnosis [36], there is no direct biomarker or powerful therapeutic target available to improve the treatment efficiency.

TOP2A is a member of the TOP2 family that plays a vital role in promoting transcriptional initiation in DNA replication, chromosome condensation, and mitosis [37]. Furthermore, TOP2A has been reported to play a crucial role in cancer progression. For example, TOP2A expression is usually increased or down-regulated depending on the presence of Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2). Based on this characteristic, these two genes are often co-tested before treating patients with Herceptin as an anti-cancer drug [38, 39]. Other studies have demonstrated that TOP2A is upregulated in lung cancer patients by IHC staining, and it has also been discovered that TOP2A expression is correlated with poor overall survival in lung cancer patients [40]. Moreover, high expression of TOP2A and EZH2 is positively correlated with aggressive disease progression in prostate cancer patients [41]. Recently, TOP2A was considered as a therapeutic target of anti-cancer drugs. For example, TOP2A may serve as a predictor of responses to anthracycline therapy in breast cancer patients, and etoposide is used for the treatment of different cancers (lung cancer, ovarian, lymphoma, and acute myeloid leukemia) [42, 43, 44]. Furthermore, several researchers have clarified that topoisomerase II inhibitors affect cancer cell proliferation by inducing apoptosis, altering metabolism, and regulating the JAK2-STAT1-CXCL1 molecular pathway [45, 46]. In addition, topoisomerase inhibitors directly affect nucleic acid metabolism, which hints at their potential lethality [47]. Nevertheless, the functions and regulation of TOP2A in CCA patients were still unknown prior to this study.

In order to gain insight into the impact of TOP2A on CCA progression and prognosis, we conducted an analysis of TOP2A expression in patient tissues using an online database. The results revealed that TOP2A expression was markedly elevated in CCA patients, particularly in those with advanced primary T stage and those with poor overall survival rates and R1 stage surgical margins.

DNA double-strand breaks (DSBs) can be repaired by homologous recombination (HR) or non-homologous end joining (NHEJ) in response to DNA damage [48]. Homologous recombination is a DNA repair process that involves RAD51 recombinase and its regulator BRCA2 [49]. Since topoisomerase II may also generate transient DSBs in DNA [50], it is not surprisingly that the BRCA2, RAD51, and RAD51AP1 genes were found to be significantly positively correlated with TOP2A (Supplementary Table 1 and Fig. 4). BRCA2 is generally regarded as a tumor suppressor, and its mutation may contribute to an increased risk for the development of various cancers, especially breast and ovarian cancers [51]. However, the recovery of BRCA2 function owing to secondary BRCA2 mutation has been considered a mechanism associated with acquired resistance to cisplatin, suggesting that this genetic reversion is beneficial for cell survival [52]. In addition, high RAD51 expression has also been indicated to increase drug resistance and genome instability in tumor cells [53]. Moreover, as an RAD51 activator, RAD51AP1 upregulation has been correlated with inferior survival in hepatocellular carcinoma patients [54]. Collectively, the involvement of TOP2A, BRCA2, RAD51, and RAD51AP1 in IHCC development deserves further investigation.

5. Conclusions

In summary, the expression of TOP2A has been recognized to be upregulated in CCA patients, and this was confirmed by immunohistochemistry staining of CCA tissue sections. The expression of TOP2A was significantly correlated with the primary tumor stage (according to AJCC stages) and histological variant (large duct type). Moreover, high expression of TOP2A is predictive of worse overall survival, disease-specific survival, and metastasis-free survival rates. In addition, increased expression of TOP2A may contribute to tumor progression in CCA patients. This information indicates that TOP2A can be considered for use in future prospective prognostic analyses. Moreover, we need more evidence to clarify the molecular mechanisms and explore the biological functions of TOP2A to determine its potential as a therapeutic target for CCA in clinical trials.

Availability of Data and Materials

The transcriptome dataset (GSE26566) used in this study is publicly available in the Gene Expression Omnibus (GEO) database, which is maintained by the National Center for Biotechnology Information in Bethesda, MD, USA.

Author Contributions

Conceptualization—KHO and YHK; methodology—KHO, HYL, DPS, TJC, SKHH, YFT, CLC, YLS, TCC, and CFL; investigation—KHO, HYL, DPS, TJC, SKHH, YFT, CLC, YLS, TCC, and CFL; formal analysis—KHO, HYL, DPS, TJC, SKHH, YFT, CLC, YLS, TCC, and CFL; resources—CLC, YLS, TCC, and CFL; validation—KHO, HYL, DPS, TJC, SKHH, and YFT; visualization—KHO, HYL, DPS, TJC, SKHH, and YFT; writing - original draft—KHO and YHK; writing - review & editing—KHO and YHK; funding acquisition—YHK; supervision—KHO and YHK. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

All participants in this study provided informed consent before their samples were deposited in the biobank. The Ethics Committee and Institutional Review Board of Chi Mei Medical Center (IRB09912003) approved the study’s use of deidentified tumor samples from the biobank, indicating that the research was conducted in compliance with ethical guidelines outlined in the Declaration of Helsinki and regulations set forth by the government.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Razumilava N, Gores GJ. Cholangiocarcinoma. Lancet. 2014; 383: 2168–2179.
[2]
Battal M, Gürbulak B, Bostanci O, Yılmaz MB, Ozdenkaya Y, Karatepe O. Cholangiocarcinoma presenting with hypercalcemia and thrombocytopenia. Case Reports in Medicine. 2014; 2014: 246817.
[3]
Razumilava N, Gores GJ. Combination of gemcitabine and cisplatin for biliary tract cancer: a platform to build on. Journal of Hepatology. 2011; 54: 577–578.
[4]
Khan SA, Davidson BR, Goldin RD, Heaton N, Karani J, Pereira SP, et al. Guidelines for the diagnosis and treatment of cholangiocarcinoma: an update. Gut. 2012; 61: 1657–1669.
[5]
Tyson GL, El-Serag HB. Risk factors for cholangiocarcinoma. Hepatology. 2011; 54: 173–184.
[6]
Shin HR, Oh JK, Lim MK, Shin A, Kong HJ, Jung KW, et al. Descriptive epidemiology of cholangiocarcinoma and clonorchiasis in Korea. Journal of Korean Medical Science. 2010; 25: 1011–1016.
[7]
Matchar DB, Samsa GP, Matthews JR, Ancukiewicz M, Parmigiani G, Hasselblad V, et al. The Stroke Prevention Policy Model: linking evidence and clinical decisions. Annals of Internal Medicine. 1997; 127: 704–711.
[8]
Razumilava N, Gores GJ. Classification, diagnosis, and management of cholangiocarcinoma. Clinical Gastroenterology and Hepatology: the Official Clinical Practice Journal of the American Gastroenterological Association. 2013; 11: 13–21.e1.
[9]
Wadsworth CA, Lim A, Taylor-Robinson SD, Khan SA. The risk factors and diagnosis of cholangiocarcinoma. Hepatology International. 2013; 7: 377–393.
[10]
Lee SK, Wang W. Roles of Topoisomerases in Heterochromatin, Aging, and Diseases. Genes. 2019; 10: 884.
[11]
Lan J, Huang HY, Lee SW, Chen TJ, Tai HC, Hsu HP, et al. TOP2A overexpression as a poor prognostic factor in patients with nasopharyngeal carcinoma. Tumour Biology. 2014; 35: 179–187.
[12]
Zhu L, Ma N, Wang B, Zhou C, Yan Y, Wang K, et al. Clinical analysis of 21-gene recurrence score test in hormone receptor-positive early-stage breast cancer. Oncology Letters. 2019; 17: 5469–5480.
[13]
Xiao H, Xu D, Chen P, Zeng G, Wang X, Zhang X. Identification of Five Genes as a Potential Biomarker for Predicting Progress and Prognosis in Adrenocortical Carcinoma. Journal of Cancer. 2018; 9: 4484–4495.
[14]
Liu L, Lin J, He H. Identification of Potential Crucial Genes Associated With the Pathogenesis and Prognosis of Endometrial Cancer. Frontiers in Genetics. 2019; 10: 373.
[15]
Chen T, Sun Y, Ji P, Kopetz S, Zhang W. Topoisomerase IIα in chromosome instability and personalized cancer therapy. Oncogene. 2015; 34: 4019–4031.
[16]
Pei YF, Yin XM, Liu XQ. TOP2A induces malignant character of pancreatic cancer through activating β-catenin signaling pathway. Biochimica et Biophysica Acta. Molecular Basis of Disease. 2018; 1864: 197–207.
[17]
Zhang R, Xu J, Zhao J, Bai JH. Proliferation and invasion of colon cancer cells are suppressed by knockdown of TOP2A. Journal of Cellular Biochemistry. 2018; 119: 7256–7263.
[18]
Andersen JB, Thorgeirsson SS. Genetic profiling of intrahepatic cholangiocarcinoma. Current Opinion in Gastroenterology. 2012; 28: 266–272.
[19]
Chan TC, Hsing CH, Shiue YL, Huang SK, Hsieh KL, Kuo YH, et al. Angiogenesis Driven by the CEBPD-hsa-miR-429-VEGFA Signaling Axis Promotes Urothelial Carcinoma Progression. Cells. 2022; 11: 638.
[20]
Ku JL, Yoon KA, Kim IJ, Kim WH, Jang JY, Suh KS, et al. Establishment and characterisation of six human biliary tract cancer cell lines. British Journal of Cancer. 2002; 87: 187–193.
[21]
Merdrignac A, Angenard G, Allain C, Petitjean K, Bergeat D, Bellaud P, et al. A novel transforming growth factor beta-induced long noncoding RNA promotes an inflammatory microenvironment in human intrahepatic cholangiocarcinoma. Hepatology Communications. 2018; 2: 254–269.
[22]
Chan TC, Chen YT, Tan KT, Wu CL, Wu WJ, Li WM, et al. Biological significance of MYC and CEBPD coamplification in urothelial carcinoma: Multilayered genomic, transcriptional and posttranscriptional positive feedback loops enhance oncogenic glycolysis. Clinical and Translational Medicine. 2021; 11: e674.
[23]
Rimassa L, Personeni N, Aghemo A, Lleo A. The immune milieu of cholangiocarcinoma: From molecular pathogenesis to precision medicine. Journal of Autoimmunity. 2019; 100: 17–26.
[24]
Labib PL, Goodchild G, Pereira SP. Molecular Pathogenesis of Cholangiocarcinoma. BMC Cancer. 2019; 19: 185.
[25]
Nakeeb A, Lipsett PA, Lillemoe KD, Fox-Talbot MK, Coleman J, Cameron JL, et al. Biliary carcinoembryonic antigen levels are a marker for cholangiocarcinoma. American Journal of Surgery. 1996; 171: 147–153.
[26]
Nehls O, Gregor M, Klump B. Serum and bile markers for cholangiocarcinoma. Seminars in Liver Disease. 2004; 24: 139–154.
[27]
Levy C, Lymp J, Angulo P, Gores GJ, Larusso N, Lindor KD. The value of serum CA 19-9 in predicting cholangiocarcinomas in patients with primary sclerosing cholangitis. Digestive Diseases and Sciences. 2005; 50: 1734–1740.
[28]
Koopmann J, Thuluvath PJ, Zahurak ML, Kristiansen TZ, Pandey A, Schulick R, et al. Mac-2-binding protein is a diagnostic marker for biliary tract carcinoma. Cancer. 2004; 101: 1609–1615.
[29]
Patel AH, Harnois DM, Klee GG, LaRusso NF, Gores GJ. The utility of CA 19-9 in the diagnoses of cholangiocarcinoma in patients without primary sclerosing cholangitis. The American Journal of Gastroenterology. 2000; 95: 204–207.
[30]
Shaib Y, El-Serag HB. The epidemiology of cholangiocarcinoma. Seminars in Liver Disease. 2004; 24: 115–125.
[31]
Bergquist A, Glaumann H, Persson B, Broomé U. Risk factors and clinical presentation of hepatobiliary carcinoma in patients with primary sclerosing cholangitis: a case-control study. Hepatology. 1998; 27: 311–316.
[32]
Scott J, Shousha S, Thomas HC, Sherlock S. Bile duct carcinoma: a late complication of congenital hepatic fibrosis. Case report and review of literature. The American Journal of Gastroenterology. 1980; 73: 113–119.
[33]
Khan SA, Davidson BR, Goldin R, Pereira SP, Rosenberg WMC, Taylor-Robinson SD, et al. Guidelines for the diagnosis and treatment of cholangiocarcinoma: consensus document. Gut. 2002; 51: VI1–9.
[34]
Jarnagin WR, Shoup M. Surgical management of cholangiocarcinoma. Seminars in Liver Disease. 2004; 24: 189–199.
[35]
Ortner MA. Photodynamic therapy in cholangiocarcinomas. Best Practice & Research. Clinical Gastroenterology. 2004; 18: 147–154.
[36]
Nieveen van Dijkum EJ, de Wit LT, Gouma DJ. Imaging of the hepatobiliary tract. The New England Journal of Medicine. 1997; 337: 1391–1392.
[37]
Madabhushi R. The Roles of DNA Topoisomerase IIβ in Transcription. International Journal of Molecular Sciences. 2018; 19: 1917.
[38]
Chen JR, Chien HP, Chen KS, Hwang CC, Chen HY, Yeh KY, et al. Amplification of HER2 and TOP2A and deletion of TOP2A genes in a series of Taiwanese breast cancer. Medicine. 2017; 96: e5582.
[39]
Huijsmans CJJ, van den Brule AJC, Rigter H, Poodt J, van der Linden JC, Savelkoul PHM, et al. Allelic imbalance at the HER2/TOP2A locus in breast cancer. Diagnostic Pathology. 2015; 10: 56.
[40]
Ma W, Wang B, Zhang Y, Wang Z, Niu D, Chen S, et al. Prognostic significance of TOP2A in non-small cell lung cancer revealed by bioinformatic analysis. Cancer Cell International. 2019; 19: 239.
[41]
Labbé DP, Sweeney CJ, Brown M, Galbo P, Rosario S, Wadosky KM, et al. TOP2A and EZH2 Provide Early Detection of an Aggressive Prostate Cancer Subgroup. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2017; 23: 7072–7083.
[42]
Wang S, Zimmermann S, Parikh K, Mansfield AS, Adjei AA. Current Diagnosis and Management of Small-Cell Lung Cancer. Mayo Clinic Proceedings. 2019; 94: 1599–1622.
[43]
Wu CC, Li TK, Farh L, Lin LY, Lin TS, Yu YJ, et al. Structural basis of type II topoisomerase inhibition by the anticancer drug etoposide. Science. 2011; 333: 459–462.
[44]
Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Jr, Wu YL, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017; 389: 299–311.
[45]
Wasim L, Chopra M. Synergistic anticancer effect of panobinostat and topoisomerase inhibitors through ROS generation and intrinsic apoptotic pathway induction in cervical cancer cells. Cellular Oncology. 2018; 41: 201–212.
[46]
Du J, Liu S, He J, Liu X, Qu Y, Yan W, et al. MicroRNA-451 regulates stemness of side population cells via PI3K/Akt/mTOR signaling pathway in multiple myeloma. Oncotarget. 2015; 6: 14993–15007.
[47]
Chowdhury SR, Majumder HK. DNA Topoisomerases in Unicellular Pathogens: Structure, Function, and Druggability. Trends in Biochemical Sciences. 2019; 44: 415–432.
[48]
Vítor AC, Huertas P, Legube G, de Almeida SF. Studying DNA Double-Strand Break Repair: An Ever-Growing Toolbox. Frontiers in Molecular Biosciences. 2020; 7: 24.
[49]
West SC. Molecular views of recombination proteins and their control. Nature Reviews. Molecular Cell Biology. 2003; 4: 435–445.
[50]
Deweese JE, Osheroff N. The DNA cleavage reaction of topoisomerase II: wolf in sheep’s clothing. Nucleic Acids Research. 2009; 37: 738–748.
[51]
Moynahan ME. The cancer connection: BRCA1 and BRCA2 tumor suppression in mice and humans. Oncogene. 2002; 21: 8994–9007.
[52]
Dhillon KK, Swisher EM, Taniguchi T. Secondary mutations of BRCA1/2 and drug resistance. Cancer Science. 2011; 102: 663–669.
[53]
Klein HL. The consequences of Rad51 overexpression for normal and tumor cells. DNA Repair. 2008; 7: 686–693.
[54]
Zhuang L, Zhang Y, Meng Z, Yang Z. Oncogenic Roles of RAD51AP1 in Tumor Tissues Related to Overall Survival and Disease-Free Survival in Hepatocellular Carcinoma. Cancer Control. 2020; 27: 1073274820977149.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top