Open Access

Identification of potential core genes and miRNAs in testicular seminoma via bioinformatics analysis

  • Authors:
    • Kai Wang
    • Yun Chen
    • Zhihong Zhao
    • Meiying Feng
    • Shouquan Zhang
  • View Affiliations

  • Published online on: September 16, 2019     https://doi.org/10.3892/mmr.2019.10684
  • Pages: 4013-4022
  • Copyright: © Wang 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

Testicular seminoma is one of the most common tumours in the field of urology, and its aetiology is still unclear. The aim of the present study was to identify the factors responsible for the development of testicular cancer and to investigate whether mutations in these genes were primarily congenital or acquired. To identify the key genes and miRNAs linked to testicular seminoma, as well as their potential molecular mechanisms, the GSE15220, GSE1818 and GSE59520 microarray datasets were analysed. A total of 5,195 and 1,163 differentially expressed genes (DEGs) were identified after analysing the GSE15220 and GSE1818 datasets, respectively. Among them, 287 genes were common between the two datasets. Of these, 110 were upregulated and 177 were downregulated. Five differentially expressed microRNAs (miRs; DEMs) that were downregulated in seminoma were identified after analysing the GSE59520 dataset. Following protein‑protein interaction network and Gene Ontology analysis, the five nodes with the highest degrees were screened as hub genes. Among them, the high expression of hub genes, such as protein tyrosine phosphatase receptor type C (PTPRC), was associated with worse overall survival. We also predicted the potential target genes of the DEMs. DNA topoisomerase II α (TOP2A), marker of proliferation Ki‑67 (MKI67), PTPRC and ubiquitin conjugating enzyme E2 C were associated with the PI3K/AKT and Wnt/β‑catenin signalling pathways. In addition, hsa‑miR‑650 and hsa‑miR‑665 were associated with the PI3K/AKT and Wnt/β‑catenin signalling pathways. Additionally, TOP2A and MKI67 were strongly associated with the target genes hsa‑miR‑650 and hsa‑miR‑665, respectively. We proposed that the hub genes reported in the present study may have a certain impact on cellular proliferation and migration in testicular seminoma. The roles of these hub genes in seminoma may provide novel insight to improve the diagnosis and treatment of patients with seminoma.

Introduction

The tumour of the testis is one of the most common type of tumours in the field of urology (1). Testicular tumours are divided into seminoma and non-seminoma types, the vast majority of which are seminoma; non-seminoma types are extremely rare. Testicular tumours are almost all malignant, with germ cell tumours accounting for 90–95%, and non-germ cell tumours accounting for 5–10% (2). In germ cell tumours, seminoma is the most common, accounting for 40% to 50% of primary testicular tumours, followed by embryonic cancer (20–30%) and teratoma (~10%) (35). Left and right testicular tumours of other cell types are rare. The treatment of testicular tumours is divided into single treatment, and the comprehensive treatment of surgical treatment radiation therapy and chemotherapy (6). Unfortunately, once a testicular tumour is identified, radical orchiectomy should be performed first, and then a further treatment plan should be implemented based on the pathological findings (710). Hence, it is urgent and necessary to explore novel therapeutic targets for the treatment of seminoma.

In the present study, we selected three gene expression datasets (GSE15220, GSE1818 and GSE59520), which were downloaded from the Gene Expression Omnibus (GEO) database, to obtain differentially expressed genes (DEGs) and differentially expressed microRNAs (DEMs) between testicular seminoma tissues and normal tissue samples. Then, functional enrichment and network analyses were applied to identify the DEGs. Subsequently, we established a protein-protein interaction (PPI) network to identify hub genes related to seminoma. The expression values of these hub genes were determined using the online database UALCAN. Survival analysis of these hub genes was performed using the online database Gene Expression Profiling Interactive Analysis (GEPIA). The potential target genes of the miRNAs were predicted by miRwalk 3.0 and screened by The Cancer Genome Atlas (TCGA) dataset.

Materials and methods

Identification of DEGs and DEMs

Three gene expression profiles, GSE15220, GSE1818 and GSE59520, were acquired from the GEO database (https://www.ncbi.nlm.nih.gov/geo/). The array data of GSE15220 comprising 6 paired seminoma tissues, and adjacent tissues were submitted by Cheung et al (11); GSE1818 consisted of 6 paired seminoma tissues and adjacent tissues. GSE59520 consisted of 14 seminoma tissues and three normal tissues (12). DEGs were obtained from the GEO database by GEO2R analysis (http://www.ncbi.nlm.nih.gov/geo/geo2r/). Adjusted P<0.05 and log fold-change (|logFC|) >2.0 were set as the DEG cutoff criterion. Adjusted P<0.05 and |logFC| >1.0 were set as the DEM cutoff criterion. The common dysregulated genes between GSE1818 and GSE59520 are presented as a Venn diagram and identified using R (version 3.6.1; http://www.r-project.org/). The Search Tool for the Retrieval of Interacting Genes (STRING) database (https://string-db.org/) for annotation, visualization and integrated discovery was employed to facilitate the transition from data collection to biological analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the Enrichr online tool (http://amp.pharm.mssm.edu/Enrichr/). P<0.05 was set as the cut off criterion. Potential target genes of the miRNAs were predicted by miRwalk 3.0 (http://mirwalk.umm.uni-heidelberg.de/) and screened using the TCGA (https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga) database.

GO and KEGG pathway analyses of DEGs

GO analysis is a commonly used method for large-scale functional enrichment research; gene functions can be classified into biological process (BP), molecular function (MF) and cellular component (CC). KEGG is a widely used database that stores data on genomes, biological pathways, diseases, chemical substances and drugs. GO annotation and KEGG pathway enrichment analyses of the DEGs identified in this study were performed using Enrichr tools. P<0.05 was considered to indicate a statistically significant difference.

Integration of the PPI network and hub gene selection

The STRING database is designed to analyse PPI information. To evaluate the potential PPI relationships, the DEGs we identified were mapped to the STRING database. The PPI pairs with a combined score of 0.4 were extracted. Subsequently, the PPI network was visualized by Cytoscape software (version 3.7.1; www.cytoscape.org/). Nodes with a higher degree of connectivity tend to be more essential in maintaining the stability of the entire network. CytoHubba (version 0.1) (13), a plugin in Cytoscape, was used to calculate the degree of each protein node. In our study, the top five genes were identified as hub genes. miRwalk (version 3.0; http://mirwalk.umm.uni-heidelberg.de/) was used to predict the potential target genes of the miRNAs identified.

Expression profiles of hub genes based on tumour histology and survival analysis

UALCAN (http://UALCAN.path.uab.edu) is a user-friendly, interactive web resource for analysing cancer transcriptome data. According to the median expression of a particular gene, the patients with testicular germ cell tumors (TGCT) were split into high and low expression groups. The overall survival (OS) of TGCT patients was evaluated using GEPIA (14). P<0.05 was considered to indicate a statistically significant result.

Results

Identification of DEGs and DEMs

The gene expression profiles GSE15220, GSE1818 and GSE59520 were selected in this study. Based on the criteria of P<0.05 and |logFC|>2.0, a total of 5,195 DEGs were identified from GSE15220, and 1,163 DEGS were identified from GSE1818. Among them, 287 genes were common to the two datasets (Fig. 1). Of these, 110 were upregulated, and 176 were downregulated (Fig. 2). A total of 8 DEMs that are downregulated in seminoma were identified from GSE59520. All DEGs and DEMs were identified by comparing seminoma samples with normal samples.

Functional and pathway enrichment analyses

GO function and KEGG pathway enrichment analyses for the DEGs were performed using Enrichr (Table I). The enriched GO terms were divided into CC, BP, and MF ontology terms. The results of GO analysis indicated that upregulated genes were mainly enriched in BPs, including ‘regulation of the cellular macromolecule biosynthetic process’, ‘B cell activation’, ‘regulation of nucleic acid-templated transcription’, and ‘regulation of gene expression’, while the downregulated genes were mainly involved in ‘calcium-dependent cell-cell adhesion via plasma membrane cell adhesion molecules (CAMs)’, ‘spermatid development’, ‘spermatogenesis’, and ‘cell-cell adhesion via plasma membrane adhesion molecules’. MF analysis showed that the upregulated genes were significantly enriched in ‘RNA binding’, ‘protein homodimerization activity’, ‘protein heterodimerization activity’, and ‘transcription regulatory region DNA binding; downregulated genes were mainly enriched in ‘motor activity, microtubule motor activity’, ‘actin binding’, and ‘ATPase activity’. For CC ontology, the upregulated genes were mainly enriched in ‘focal adhesion’, ‘phagocytic vesicle membrane’, ‘integral component of the luminal side of the endoplasmic reticulum membrane’, and ‘early endosome membrane’, and the downregulated genes were mainly enriched in ‘condensed nuclear chromosome’, ‘centromeric region’, ‘cytoplasmic dynein complex’, ‘spindle midzone’, and ‘focal adhesion’.

Table I.

Significantly enriched Go terms and KEGG pathways of differentially expressed genes.

Table I.

Significantly enriched Go terms and KEGG pathways of differentially expressed genes.

A, Upregulated

CategoryTermDescriptionCountP-value
BP termGO:2000112Regulation of cellular macromolecule biosynthetic process130.000044
BP termGO: 0042113B cell activation (GO:0042113)40.000741
BP termGO:1903506Regulation of nucleic acid-templated transcription120.000130
BP termGO:0010468Regulation of gene expression150.000557
CC termGO:0005925Focal adhesion80.000804
CC termGO:0030670Phagocytic vesicle membrane40.000061
CC termGO:0071556Integral component of luminal side of endoplasmic reticulum membrane30.000590
CC termGO:0031901Early endosome membrane40.000741
MF termGO:0042803Protein homodimerization activity16 7.14×107
MF termGO:0003723RNA binding180.000550
MF termGO:0046982Protein heterodimerization activity70.000675
MF termGO:0044212Transcription regulatory region DNA binding100.003244
KEGG pathwayhsa05230Viral carcinogenesis40.026682
KEGG pathwayhsa04670Leukocyte transendothelial migration30.027385
KEGG pathwayhsa04514Cell adhesion molecules60.000135

B, Downregulated

CategoryTerm DescriptionCountP-value

BP termGO:0016339Calcium-dependent cell-cell adhesion via plasma membrane cell adhesion molecules40.000500
BP termGO:0007286Spermatid development30.004178
BP termGO:0007283 Spermatogenesis40.047347
BP termGO:0098742Cell-cell adhesion via plasma-membrane adhesion molecules50.008733
CC termGO:0000780Condensed nuclear chromosome, centromeric region20.005635
CC termGO:0005868Cytoplasmic dynein complex20.015846
CC termGO:0051233Spindle midzone20.030298
CC termGO:0005925Focal adhesion70.038965
MF termGO:0003774Motor activity60.000112
MF termGO:0003777Microtubule motor activity40.002046
MF termGO:0003779Actin binding60.025579
MF termGO:0016887ATPase activity50.034558
KEGG pathwayhsa00531Amphetamine addiction40.002886
KEGG pathwayhsa04720Long-term potentiation30.020452
KEGG pathwayhsa04114Oocyte meiosis in diabetic complication40.023439

[i] BP, biological process; CC, cellular component; DEG, differentially expressed gene; ECM, extracellular matrix; GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; MF, molecular function.

In addition, KEGG pathway analysis showed that the DEGs were mainly enriched in ‘viral carcinogenesis’, ‘leukocyte transendothelial migration’ and ‘CAMs’, while the downregulated genes were mainly enriched in ‘amphetamine addiction’, ‘long-term potentiation’, and ‘oocyte meiosis in diabetic complications’.

PPI network construction and the analysis of hub genes

A total of 123 nodes and 269 edges were mapped in the PPI network of the identified DEGs (Fig. 3). The 5 nodes with the highest degrees, including DNA topoisomerase II α (TOP2A), ubiquitin conjugating enzyme E2 C (UBE2C), protein tyrosine phosphatase receptor type C (PTPRC), marker of proliferation Ki-67 (MKI67), and centromere protein A (CENPA), were screened as hub genes (Fig. 4, Table II). GO term enrichment analysis showed that in BPs, the genes in this module were mainly associated with ‘negative regulation of chromosome organization’, ‘regulation of metaphase/anaphase transition of cell cycle’, ‘regulation of cell cycle process’, and ‘condensed nuclear chromosome kinetochore’ (Table III). The genes were significantly enriched in the ‘condensed nuclear chromosome kinetochore’, ‘nuclear ubiquitin ligase complex’ ‘chromosome’, and ‘nucleolus’ (Table III). MF analysis showed that the genes were mainly enriched in ‘ubiquitin-like protein conjugating enzyme activity’, ‘protein kinase binding’, and ‘ubiquitin binding’ (Table III). KEGG analysis revealed that the genes were mainly enriched in ‘cell adhesion molecules’, ‘primary immunodeficiency’, and ‘Fc gamma R-mediated phagocytosis’ (Table III).

Table II.

Top five hub genes with the highest degrees of connectivity.

Table II.

Top five hub genes with the highest degrees of connectivity.

GeneGene descriptionDegree
TOP2ATopoisomerase (DNA) II α 170 kDa33
UBE2C Ubiquitin-conjugating enzyme E2C16
PTPRCProtein tyrosine phosphatase, receptor type, C15
MKI67Antigen identified by monoclonal antibody Ki-6713
CENPACentromere protein A13

Table III.

Significantly enriched GO terms and KEGG pathways of the top five hub genes.

Table III.

Significantly enriched GO terms and KEGG pathways of the top five hub genes.

CategoryTermDescriptionCountP-value
BP termGO:2001251Negative regulation of chromosome organization10.001749
BP termGO:1902099Regulation of metaphase/anaphase transition of cell cycle10.002248
BP termGO:0010564Regulation of cell cycle process10.022500
CC termGO:0000778Condensed nuclear chromosome kinetochore20.002997
CC termGO:0005730Nucleolus20.010690
CC termGO:0005694Chromosome20.000235
CC termGO:0000152Nuclear ubiquitin ligase complex10.010460
MF termGO:0061650Ubiquitin-like protein conjugating enzyme activity10.005488
MF termGO:0019198Transmembrane receptor protein phosphatase activity10.003994
MF termGO:0043130Ubiquitin binding10.015650
MF termGO:0019901Protein kinase binding20.005841
KEGG pathwayhsa04514Cell adhesion molecules10.035000
KEGG pathwayhsa05340Primary immunodeficiency10.009217
KEGG pathwayhsa04666Fc gamma R-mediated phagocytosis10.023040

[i] BP, biological process; CC, cellular component; DEG, differentially expressed gene; ECM, extracellular matrix; GO, Gene Ontology; hsa, homo sapiens; KEGG, Kyoto Encyclopedia of Genes and Genomes; MF, molecular function.

Expression profiles of the hub genes and survival analysis

To investigate the expression and prognostic values of the five potential hub genes, the UALCAN bioinformatics analysis platform was used. All of the hub genes were significant (Fig. 5). We found that the high expression of these hub genes was associated with an unfavourable OS of patients with testicular seminoma by GEPIA (Fig. 6). However, because of the better prognosis of testicular cancer and few mortalities, the overexpression of only PTPRC was identified as an unfavourable prognostic overall survival in patients with seminoma.

Potential target genes of the miRNAs

To predict the potential target genes of the miRNAs, miRwalk and Cytoscape were used. miR-661 was found to be associated with PTPRC, miR-640 and miR-665 to MKI67, miR-1204 with CENPA, miR-1203 with UBE2C, miR-650 and 934 with TOP2A, and miR-1182 with TOP2A, UBE2C and MKI67 (Fig. 7).

Discussion

Although the cure and survival rates of testicular cancer are high, the main treatment methods are chemotherapy and resection (15), which presents a great burden on the patient quality of life. In addition, the incidence of seminoma is rising. The lifestyles of males in different regions vary, while the incidence rate also differs (16). It is estimated that by 2030, there will be 65,827 new cases worldwide, an increase of 10,561 cases from 2012 (17). Although multiple approaches have reduced mortality, treatment resistance, disease relapse and treatment-derived side effects in particular, are important issues at present (18). Therefore, it is important to develop novel specific targeted therapies for the treatment of seminoma.

In our study, a total of 287 DEGs were screened, including 110 upregulated genes and 177 downregulated genes. The upregulated genes were enriched in ‘viral carcinogenesis’, ‘leukocyte transendothelial migration’ and ‘CAMs’, while the downregulated genes were mainly enriched in ‘amphetamine addiction’, ‘long-term potentiation’ and ‘oocyte meiosis in diabetic complications’. Moreover, by constructing the PPI network, we identified five high-degree hub genes, including TOP2A, UBE2C, PTPRC, MKI67 and CENPA. Among them, all hub genes were upregulated in seminoma. Finally, we used UALCAN to analyse the expression of these hub genes; all the hub genes were reported to be significant. Then, we predicted the association between the expression of the hub genes and the prognosis of TGCT patients. Based on GEPIA, the overexpression of all hub genes was related to an unfavourable prognosis in patients with testicular cancer. Among them, we found that the overexpression of PTPRC was an unfavourable prognostic factor for patients with seminoma.

TOP2A is the molecular target of several clinically useful chemotherapeutic drugs and has been used to treat a variety of tumours, including breast cancer, prostate cancer and endometrial cancer (19). Certain studies have suggested that the overexpression of TOP2A may be associated with the poor prognosis of these malignant diseases. For seminoma, TOP2A overexpression was associated with aggressive clinical behaviours (20). Coleman et al (21) and Sano and Shuhin (22) suggested that TOP2A may be a marker of seminoma cell proliferation, and TOP2A was easily detected in seminoma. Additionally, studies have also revealed that TOP2A is related to primary tumours (2325). In prostate cancer, Labbé et al (26) found that TOP2A and EZH2 mRNA and protein upregulation was linked to a subgroup of primary and metastatic patients with more aggressive disease, and exhibited a notable overlap of genes involved in mitotic regulation, these results further support the hypothesis of TOP2A as a biomarker for the early identification of patients with increased metastatic potential that may benefit from adjuvant or neoadjuvant targeted therapy approaches (26). UBE2C serves as the key component in the ubiquitin proteasome system by partnering with the anaphase-promoting complex (APC/C). Upregulated UBE2C protein expression has been reported in various types of human tumours (27). Mo et al (28) conducted an immunoassay to examine 209 breast cancer (BRCA) tissue samples and 53 normal tissue samples, and found that UBE2C is highly expressed in BRCA. Furthermore, the expression of UBE2C was positively correlated with tumour size (24). Wang et al (29) revealed that the level of phosphorylated aurora kinase A (p-AURKA) decreased markedly via the Wnt/β-catenin and PI3K/AKT signalling pathways following the knockdown of UBE2C with a small interfering RNA. The signalling pathway suppressed the occurrence and development of gastric cancer, and their data suggested that the activity of AURKA may be regulated by UBE2C via modulation of the activity of APC/C (29), UBE2C may be a novel marker in the diagnosis of gastric cancer (25); however, little is known about UBE2C in seminoma. PTPRC is a member of the protein tyrosine phosphatase (PTP) family. PTPs are signalling molecules that regulate a variety of cellular processes, including cell growth, differentiation, mitosis, and oncogenic transformation (30). Porcu et al (31) demonstrated that the downregulation of CD45 (encoded by PTPRC) expression sensitizes T cells to cytokine stimulation, as observed by increased JAK/STAT signalling, whereas the overexpression of CD45 decreases cytokine-induced signalling. In our study, PTPRC, which may serve an important role in cytokine induction, was upregulated in seminoma. The expression of MKI67 is strongly associated with tumour cell proliferation and growth, and is widely employed in routine pathological investigations as a proliferation marker (32). It was revealed that the expression of P53 and Wnt signalling correlated with that the expression of MKI-67 in several types of cancer (3336). Downregulated MKI67 may be involved in cancer; in our study, it was downregulated in seminoma. Five pivotal genes detected in this study have been reported to be overexpressed in various human cancers, are associated with their prognosis and are significantly expressed in testicular seminoma tissues; but no significant difference in prognosis was reported. The role of these genes in TGCT is unclear; thus, further investigation is required.

Compared with normal testis samples, 11 DEMs were acquired in GSE59520 in seminoma samples. In the present study, hsa-miR-1182 was downregulated in seminoma samples. Zhou et al (37) reported that hsa-miR-1182 is dysregulated in bladder cancer tissues and cell lines, in which functional assays were then performed, the overexpression of miR-1182 significantly inhibits bladder cancer cell proliferation, colony formation and invasion (37,38); however, the effects of hsa-miR-1182 in seminoma are yet to be determined. Hsa-miR-650 has been reported in many cancers. For example, Zhou et al (39) observed that the expression of miR-650 in tumour tissues had a positive association with OS. Hsa-miR-650 inhibited cell growth and invasion in vitro and in vivo (40,41). Furthermore, miR-650 targeted AKT2 and suppressed the activation of the AKT2/glycogen synthase kinase-3β/E-cadherin (39). Interestingly, Yang et al (42) suggested that phosphatase and tensin homolog/AKT signalling affects the expression of TOP2A, reducing cell growth and inducing the apoptosis of human breast cancer MCF-7 cells through ATP and caspase-3 signalling pathways (42). In our study, TOP2A was identified as the target gene of hsa-miR-650, and hsa-miR-650 was downregulated in seminoma, while TOP2A was upregulated. Thus, we proposed that in seminoma, the downregulation of hsa-miR-650 could lead to the activation of the AKT pathway, upregulating TOP2A to affect seminoma cell migration and proliferation. At present, few studies have been conducted in the investigation of hsa-miR-934. For hsa-miR-1204, Xu et al (43) revealed that hsa-miR-1204 expression was significantly correlated with tumour size. The expression levels of hsa-miR-1204 and glucose transporter-1 (GLUT-1) were significantly high in ovarian cancer (OC) patients (43). The expression levels of hsa-miR-1204 were positively correlated with the expression levels of GLUT-1 in OC patients. Hsa-miR-1204 overexpression significantly promoted GLUT-1 expression, glucose uptake and cell proliferation. In our study, the target gene of hsa-miR-1204, CENPA, was revealed to mostly function in kinetochores and regulate cell division. The overexpression of CENPA is significantly related to colon cancer and neoplastic germ cells (44,45). Hsa-miR-1204 and its target gene CENPA were both upregulated in seminoma; kinetochores are unique centromere macromolecular protein structures that attach chromosomes to the spindle for proper movement and segregation, during this process, an increasing amount of ATP is required (46), and the overexpression of hsa-miR-1204 can significantly promote GLUT-1 expression, glucose uptake and ATP production (43). Thus, this pathway may affect the expression of CENPA; however, the association between CENPA and hsa-miR-1204 requires further investigation. In addition, hsa-miR-661 has been reported in non-small-cell lung cancer and OC (47,48). Hoffman et al (49) found that low miR-661 expression correlates with poor outcomes in BRCA that typically express wild-type p53. In the present study, miR-661 was downregulated in seminoma. Of note, hsa-miR-1203 has been reported to be dysregulated in prostate cancer, small cell carcinoma of the oesophagus and oesophageal squamous cell carcinoma (5052). In addition, Prashad et al (53) reported that miR-665 suppresses neuroblastoma tumorigenesis by inhibiting c-MYC and suggested the potential of hsa-miR-665 as an antineuroblastoma therapeutic factor. Dong et al (54) found that miR-665 was downregulated in osteosarcoma tissues compared with nontumourous tissues, and the expression of miR-665 was inversely associated with the expression of Rab23 in the osteosarcoma tissues. These results suggest that miR-665 could act as a tumour suppressor gene in the development of osteosarcoma (54); it has also been reported to be related to the Wnt/β-catenin signalling pathway (55). In our study, miR-665 was downregulated in seminoma, and the target gene MKI67 was also associated with the Wnt/β-catenin signalling pathway (34). For hsa-miR-640, Li et al (56) demonstrated that miR-640 was downregulated in paclitaxel-resistant formalin-fixed paraffin-embedded tumour samples; however, Zhou et al (57) found that miR-640 is related to the vascular endothelial growth factor receptor 2-mTOR pathway. In the present study, hsa-miR-640 was downregulated in seminoma, and its target gene, MKI67, was also associated with mTOR (58).

In summary, TOP2A, MKI67, PTPRC and UBE2C were revealed to be potentially associated with the PI3K/AKT and Wnt/β-catenin signalling pathways, while hsa-miR-650 and hsa-miR-665 were proposed to be linked the PI3K/AKT and Wnt/β-catenin signalling pathways. Moreover, TOP2A and MKI67 were strongly associated with the target genes of hsa-miR-650 and hsa-miR-665, respectively.

At present, few bioinformatics analyses have been conducted to investigate DEGs and DEMs in seminoma. Of the DEGs screened in this study, only TOP2A and PTPRC have been reported in seminoma (59,60), to the best of our knowledge. Additionally, we screened the differential expression of genes from array data and predicted target genes of DEMs which were proposed to act together on the PI3K/AKT and Wnt/β-catenin signalling pathways with a strong correlation (29,39). The PI3K/AKT and Wnt/β-catenin signalling pathways participate in the growth, invasion, and migration of cancer cells in a variety of ways (6163). TOP2A, MKI67, PTPRC and UBE2C could be used as potential diagnostic biomarkers and therapeutic molecular targets for seminoma. In addition, the main treatment methods for seminoma in clinical practice are chemotherapy and testicular resection, which undoubtedly cause great physical burden to patients, particularly in individuals aged 15–24 years (7,64). Therefore, our findings of the present study, including the genes identified, may serve as a basis for exploring gene therapy for seminoma in the future.

In this study, 287 differential genes for testicular seminoma were detected from the GEO database, and 5 pivotal genes and 8 miRNAs were screened, all of which were significantly differentially expressed in testicular cancer tissues. Among them, we predicted the target genes of the miRNAs. TOP2A, MKI67, PTPRC and UBE2C were determined to be associated with the PI3K/AKT and Wnt/β-catenin signalling pathways, and hsa-miR-650 and hsa-miR-665 were associated with the PI3K/AKT and Wnt/β-catenin signalling pathways. Furthermore, TOP2A and MKI67 were strongly associated with the target genes of hsa-miR-650 and hsa-miR-665, respectively. To the best of our knowledge, hsa-miR-934 has not been investigated. There may be certain associations that are yet to be identified; the hub genes we reported could have notable impact on cell proliferation and migration in testicular seminoma. In addition, in patients with testicular seminoma, PTPRC overexpression is an unfavourable prognostic factor, and further studies are needed to verify our findings. The results of the present study suggested that TOP2A, MKI67, CENPA, PTPRC, UBE2C, hsa-miR-650, hsa-miR-665, hsa-miR-640, hsa-miR-1182, hsa-miR-1203, hsa-miR-661 and hsa-miR-1204 may be potential targets for seminoma therapy.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasests generated and/or analyzed during the current study are available from the Gene Expression Omnibus repository, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE15220, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE1818 and https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE59520.

Authors' contributions

KW and SZ made substantial contributions to the conception of the present study. KW performed the primary bioinformatics analysis and was a major contributor in writing the manuscript; YC made substantial contributions to data analysis, including the biological significance of hub genes and figure editing. ZZ and MF were involved in the interpretation of the hub genes data and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Pedersen MR, Rafaelsen SR, Møller H, Vedsted P and Osther PJ: Testicular microlithiasis and testicular cancer: Review of the literature. Int Urol Nephrol. 48:1079–1086. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Smith ZL, Werntz RP and Eggener SE: Testicular cancer: Epidemiology, diagnosis, and management. Med Clin North Am. 102:251–264. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Ostrowski KA and Walsh TJ: Infertility with testicular cancer. Urol Clin North Am. 42:409–420. 2015. View Article : Google Scholar : PubMed/NCBI

4 

McNally RJ, Basta NO, Errington S, James PW, Norman PD, Hale JP and Pearce MS: Socioeconomic patterning in the incidence and survival of teenage and young adult men aged between 15 and 24 years diagnosed with non-seminoma testicular cancer in northern england. Urol Oncol. 33:506.e9–e14. 2015. View Article : Google Scholar

5 

Sharma P, Dhillon J and Sexton WJ: Intratubular germ cell neoplasia of the testis, bilateral testicular cancer, and aberrant histologies. Urol Clin North Am. 42:277–285. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Baird DC, Meyers GJ and Hu JS: Testicular cancer: Diagnosis and treatment. Am Fam Physician. 97:261–268. 2018.PubMed/NCBI

7 

Kamel MH, Elfaramawi M, Jadhav S, Saafan A, Raheem OA and Davis R: Insurance status and differences in treatment and survival of testicular cancer patients. Urology. 87:140–145. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Shen AH, Howell D, Edwards E, Warde P, Matthew A and Jones JM: The experience of patients with early-stage testicular cancer during the transition from active treatment to follow-up surveillance. Urol Oncol. 34:168.e11–e20. 2016. View Article : Google Scholar

9 

Brand S, Williams H and Braybrooke J: How has early testicular cancer affected your life? A study of sexual function in men attending active surveillance for stage one testicular cancer. Eur J Oncol Nurs. 19:278–281. 2015. View Article : Google Scholar : PubMed/NCBI

10 

McBride D: Surveillance is as effective as chemotherapy in stage 1 testicular cancer. ONS Connect. 29:102014.

11 

Cheung HH, Lee TL, Davis AJ, Taft DH, Rennert OM and Chan WY: Genome-wide DNA methylation profiling reveals novel epigenetically regulated genes and non-coding RNAs in human testicular cancer. Br J Cancer. 102:419–427. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Skotheim RI, Lind GE, Monni O, Nesland JM, Abeler VM, Fosså SD, Duale N, Brunborg G, Kallioniemi O, Andrews PW and Lothe RA: Differentiation of human embryonal carcinomas in vitro and in vivo reveals expression profiles relevant to normal development. Cancer Res. 65:5588–5598. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Chin CH, Chen SH, Wu HH, Ho CW, Ko MT and Lin CY: CytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 8 (Suppl 4):S112014. View Article : Google Scholar : PubMed/NCBI

14 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Bender JL, Wiljer D, To MJ, Bedard PL, Chung P, Jewett MA, Matthew A, Moore M, Warde P and Gospodarowicz M: Testicular cancer survivors' supportive care needs and use of online support: A cross-sectional survey. Support Care Cancer. 20:2737–2746. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Trabert B, Chen J, Devesa SS, Bray F and McGlynn KA: International patterns and trends in testicular cancer incidence, overall and by histologic subtype, 1973–2007. Andrology. 3:4–12. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Lobo J, Costa AL, Vilela-Salgueiro B, Rodrigues Â, Guimarães R, Cantante M, Lopes P, Antunes L, Jerónimo C and Henrique R: Testicular germ cell tumors: Revisiting a series in light of the new WHO classification and AJCC staging systems, focusing on challenges for pathologists. Hum Pathol. 82:113–124. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Russell SS: Testicular cancer: Overview and implications for health care providers. Urol Nurs. 34:172–176, 192. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Delgado JL, Hsieh CM, Chan NL and Hiasa H: Topoisomerases as anticancer targets. Biochem J. 475:373–398. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Panvichian R, Tantiwetrueangdet A, Angkathunyakul N and Leelaudomlipi S: TOP2A amplification and overexpression in hepatocellular carcinoma tissues. Biomed Res Int. 2015:3816022015. View Article : Google Scholar : PubMed/NCBI

21 

Coleman LW, Perkins SL, Bronstein IB and Holden JA: Expression of DNA toposiomerase I and DNA topoisomerase II-alpha in testicular seminomas. Hum Pathol. 31:728–733. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Sano K and Shuhin T: A study of topoisomerase activity in human testicular cancers. Anticancer Res. 15:2117–2120. 1995.PubMed/NCBI

23 

Meng H, Chen R, Li W and Xu L and Xu L: Correlations of TOP2A gene aberrations and expression of topoisomerase IIα protein and TOP2A mRNA expression in primary breast cancer: A retrospective study of 86 cases using fluorescence in situ hybridization and immunohistochemistry. Pathol Int. 62:391–399. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Jensen JD, Knoop A, Ewertz M and Laenkholm AV: ER, HER2, and TOP2A expression in primary tumor, synchronous axillary nodes, and asynchronous metastases in breast cancer. Breast Cancer Res Treat. 132:511–521. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Konecny GE, Pauletti G, Untch M, Wang HJ, Möbus V, Kuhn W, Thomssen C, Harbeck N, Wang L, Apple S, et al: Association between HER2, TOP2A, and response to anthracycline-based preoperative chemotherapy in high-risk primary breast cancer. Breast Cancer Res Treat. 120:481–489. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Labbé DP, Sweeney CJ, Brown M, Galbo P, Rosario S, Wadosky KM, Ku SY, Sjöström M, Alshalalfa M, Erho N, et al: TOP2A and EZH2 provide early detection of an aggressive prostate cancer subgroup. Clin Cancer Res. 23:7072–7083. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Xie C, Powell C, Yao M, Wu J and Dong Q: Ubiquitin-conjugating enzyme E2C: A potential cancer biomarker. Int J Biochem Cell Biol. 47:113–117. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Mo CH, Gao L, Zhu XF, Wei KL, Zeng JJ, Chen G and Feng ZB: The clinicopathological significance of UBE2C in breast cancer: A study based on immunohistochemistry, microarray and RNA-sequencing data. Cancer Cell Int. 17:832017. View Article : Google Scholar : PubMed/NCBI

29 

Wang R, Song Y, Liu X, Wang Q, Wang Y, Li L, Kang C and Zhang Q: UBE2C induces EMT through Wnt/β-catenin and PI3K/Akt signaling pathways by regulating phosphorylation levels of Aurora-A. Int J Oncol. 50:1116–1126. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Vang T, Miletic AV, Arimura Y, Tautz L, Rickert RC and Mustelin T: Protein tyrosine phosphatases in autoimmunity. Annu Rev Immunol. 26:29–55. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Porcu M, Kleppe M, Gianfelici V, Geerdens E, De Keersmaecker K, Tartaglia M, Foà R, Soulier J, Cauwelier B, Uyttebroeck A, et al: Mutation of the receptor tyrosine phosphatase PTPRC (CD45) in T-cell acute lymphoblastic leukemia. Blood. 119:4476–4479. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Menon SS, Guruvayoorappan C, Sakthivel KM and Rasmi RR: Ki-67 protein as a tumour proliferation marker. Clin Chim Acta. 491:39–45. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Yang CK, Yu TD, Han CY, Qin W, Liao XW, Yu L, Liu XG, Zhu GZ, Su H, Lu SC, et al: Genome-wide association study of MKI67 expression and its clinical implications in HBV-related hepatocellular carcinoma in Southern China. Cell Physiol Biochem. 42:1342–1357. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Bleckmann A, Conradi LC, Menck K, Schmick NA, Schubert A, Rietkötter E, Arackal J, Middel P, Schambony A, Liersch T, et al: β-catenin-independent WNT signaling and Ki67 in contrast to the estrogen receptor status are prognostic and associated with poor prognosis in breast cancer liver metastases. Clin Exp Metastasis. 33:309–323. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Li LT, Jiang G, Chen Q and Zheng JN: Ki67 is a promising molecular target in the diagnosis of cancer (review). Mol Med Rep. 11:1566–1572. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Gallegos I, Valdevenito JP, Miranda R and Fernandez C: Immunohistochemistry expression of P53, Ki67, CD30, and CD117 and presence of clinical metastasis at diagnosis of testicular seminoma. Appl Immunohistochem Mol Morphol. 19:147–152. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Zhou J, Dai W and Song J: miR-1182 inhibits growth and mediates the chemosensitivity of bladder cancer by targeting hTERT. Biochem Biophys Res Commun. 470:445–452. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Zhang T, Zhao D, Wang Q, Yu X, Cui Y, Guo L and Lu SH: MicroRNA-1322 regulates ECRG2 allele specifically and acts as a potential biomarker in patients with esophageal squamous cell carcinoma. Mol Carcinog. 52:581–590. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Zhou C, Cui F, Li J, Wang D, Wei Y, Wu Y, Wang J, Zhu H and Wang S: MiR-650 represses high-risk non-metastatic colorectal cancer progression via inhibition of AKT2/GSK3β/E-cadherin pathway. Oncotarget. 8:49534–49547. 2017.PubMed/NCBI

40 

Ningning S, Libo S, Chuanbin W, Haijiang S and Qing Z: MiR-650 regulates the proliferation, migration and invasion of human oral cancer by targeting growth factor independent 1 (Gfi1). Biochimie. 156:69–78. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Xu X, Chen H, Zhang Q, Xu J, Shi Q and Wang M: MiR-650 inhibits proliferation, migration and invasion of rheumatoid arthritis synovial fibroblasts by targeting AKT2. Biomed Pharmacother. 88:535–541. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Yang Z, Liu Y, Shi C, Zhang Y, Lv R, Zhang R, Wang Q and Wang Y: Suppression of PTEN/AKT signaling decreases the expression of TUBB3 and TOP2A with subsequent inhibition of cell growth and induction of apoptosis in human breast cancer MCF-7 cells via ATP and caspase-3 signaling pathways. Oncol Rep. 37:1011–1019. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Xu J, Gu X, Yang X and Meng Y: MiR-1204 promotes ovarian squamous cell carcinoma growth by increasing glucose uptake. Biosci Biotechnol Biochem. 1–6. 2018.(Epub ahead of print).

44 

Biermann K, Heukamp LC, Steger K, Zhou H, Franke FE, Guetgemann I, Sonnack V, Brehm R, Berg J, Bastian PJ, et al: Gene expression profiling identifies new biological markers of neoplastic germ cells. Anticancer Res. 27:3091–3100. 2007.PubMed/NCBI

45 

Liu R, Zhang W, Liu ZQ and Zhou HH: Associating transcriptional modules with colon cancer survival through weighted gene co-expression network analysis. Bmc Genomics. 18:3612017. View Article : Google Scholar : PubMed/NCBI

46 

Collins CM, Malacrida B, Burke C, Kiely PA and Dunleavy EM: ATP synthase F1 subunits recruited to centromeres by CENP-A are required for male meiosis. Nat Commun. 9:27022018. View Article : Google Scholar : PubMed/NCBI

47 

Liu F, Cai Y, Rong X, Chen J, Zheng D, Chen L, Zhang J, Luo R, Zhao P and Ruan J: MiR-661 promotes tumor invasion and metastasis by directly inhibiting RB1 in non small cell lung cancer. Mol Cancer. 16:1222017. View Article : Google Scholar : PubMed/NCBI

48 

Zhu T, Yuan J, Wang Y, Gong C, Xie Y and Li H: MiR-661 contributed to cell proliferation of human ovarian cancer cells by repressing INPP5J expression. Biomed Pharmacother. 75:123–128. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Hoffman Y, Bublik DR, Pilpel Y and Oren M: miR-661 downregulates both Mdm2 and Mdm4 to activate p53. Cell Death Differ. 21:302–309. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Haldrup C, Kosaka N, Ochiya T, Borre M, Høyer S, Orntoft TF and Sorensen KD: Profiling of circulating microRNAs for prostate cancer biomarker discovery. Drug Deliv Transl Res. 4:19–30. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Okumura T, Shimada Y, Omura T, Hirano K, Nagata T and Tsukada K: MicroRNA profiles to predict postoperative prognosis in patients with small cell carcinoma of the esophagus. Anticancer Res. 35:719–727. 2015.PubMed/NCBI

52 

Okumura T, Kojima H, Miwa T, Sekine S, Hashimoto I, Hojo S, Nagata T and Shimada Y: The expression of microRNA 574-3p as a predictor of postoperative outcome in patients with esophageal squamous cell carcinoma. World J Surg Oncol. 14:2282016. View Article : Google Scholar : PubMed/NCBI

53 

Prashad N: miR-665 targets c-MYC and HDAC8 to inhibit murine neuroblastoma cell growth. Oncotarget. 9:33186–33201. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Dong C, Du Q, Wang Z, Wang Y, Wu S and Wang A: MicroRNA-665 suppressed the invasion and metastasis of osteosarcoma by directly inhibiting RAB23. Am J Transl Res. 8:4975–4981. 2016.PubMed/NCBI

55 

Li K, Pan J, Wang J, Liu F and Wang L: MiR-665 regulates VSMCs proliferation via targeting FGF9 and MEF2D and modulating activities of Wnt/β-catenin signaling. Am J Transl Res. 9:4402–4414. 2017.PubMed/NCBI

56 

Li X, Lu Y, Chen Y, Lu W and Xie X: MicroRNA profile of paclitaxel-resistant serous ovarian carcinoma based on formalin-fixed paraffin-embedded samples. Bmc Cancer. 13:2162013. View Article : Google Scholar : PubMed/NCBI

57 

Zhou Y, Li XH, Zhang CC, Wang MJ, Xue WL, Wu DD, Ma FF, Li WW, Tao BB and Zhu YC: Hydrogen sulfide promotes angiogenesis by downregulating miR-640 via the VEGFR2/mTOR pathway. Am J Physiol Cell Physiol. 310:C305–C317. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Yanai A, Inoue N, Yagi T, Nishimukai A, Miyagawa Y, Murase K, Imamura M, Enomoto Y, Takatsuka Y, Watanabe T, et al: Activation of mTOR/S6K But Not MAPK pathways might be associated with high Ki-67, ER(+), and HER2(−) breast cancer. Clin Breast Cancer. 15:197–203. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Dimov ND, Zynger DL, Luan C, Kozlowski JM and Yang XJ: Topoisomerase II alpha expression in testicular germ cell tumors. Urology. 69:955–961. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Bo H, Cao K, Tang R, Zhang H, Gong Z, Liu Z, Liu J, Li J and Fan L: A network-based approach to identify DNA methylation and its involved molecular pathways in testicular germ cell tumors. J Cancer. 10:893–902. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Yang Q, Jiang W and Hou P: Emerging role of PI3K/AKT in tumor-related epigenetic regulation. Semin Cancer Biol. Apr 2–2019.(Epub ahead of print). View Article : Google Scholar

62 

Nusse R and Clevers H: Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169:985–999. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Fruman DA and Rommel C: PI3K and cancer: Lessons, challenges and opportunities. Nat Rev Drug Discov. 13:140–156. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Rajpert-De Meyts E, McGlynn KA, Okamoto K, Jewett MA and Bokemeyer C: Testicular germ cell tumours. Lancet. 387:1762–1774. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 20 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang K, Chen Y, Zhao Z, Feng M and Zhang S: Identification of potential core genes and miRNAs in testicular seminoma via bioinformatics analysis. Mol Med Rep 20: 4013-4022, 2019
APA
Wang, K., Chen, Y., Zhao, Z., Feng, M., & Zhang, S. (2019). Identification of potential core genes and miRNAs in testicular seminoma via bioinformatics analysis. Molecular Medicine Reports, 20, 4013-4022. https://doi.org/10.3892/mmr.2019.10684
MLA
Wang, K., Chen, Y., Zhao, Z., Feng, M., Zhang, S."Identification of potential core genes and miRNAs in testicular seminoma via bioinformatics analysis". Molecular Medicine Reports 20.5 (2019): 4013-4022.
Chicago
Wang, K., Chen, Y., Zhao, Z., Feng, M., Zhang, S."Identification of potential core genes and miRNAs in testicular seminoma via bioinformatics analysis". Molecular Medicine Reports 20, no. 5 (2019): 4013-4022. https://doi.org/10.3892/mmr.2019.10684