Investigation of crucial genes and microRNAs in conventional osteosarcoma using gene expression profiling analysis

  • Authors:
    • Chuangang Peng
    • Qi Yang
    • Bo Wei
    • Baoming Yuan
    • Yong Liu
    • Yuxiang Li
    • Dawer Gu
    • Guochao Yin
    • Bo Wang
    • Dehui Xu
    • Xuebing Zhang
    • Daliang Kong
  • View Affiliations

  • Published online on: September 18, 2017     https://doi.org/10.3892/mmr.2017.7506
  • Pages: 7617-7624
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to screen potential genes associated with conventional osteosarcoma (OS) and obtain further information on the pathogenesis of this disease. The microarray dataset GSE14359 was downloaded from the Gene Expression Omnibus. A total of 10 conventional OS samples and two non‑neoplastic primary osteoblast samples in the dataset were selected to identify the differentially expressed genes (DEGs) using the Linear Models for Microarray Data package. The potential functions of the DEGs were predicted using Gene Ontology (GO) and pathway enrichment analyses. Protein‑protein interaction (PPI) data were also obtained using the Search Tool for the Retrieval of Interacting Genes database, and the PPI network was visualized using Cytoscape. Module analysis was then performed using the Molecular Complex Detection module. Additionally, the potential microRNAs (miRNAs) for the upregulated DEGs in the most significant pathway were predicted using the miRDB database, and the regulatory network for the miRNAs‑DEGs was visualized in Cytoscape. In total, 317 upregulated and 670 downregulated DEGs were screened. Certain DEGs, including cyclin‑dependent kinase 1 (CDK1), mitotic arrest deficient 2 like 1 (MAD2L1) and BUB1 mitotic checkpoint serine/threonine‑protein kinase (BUB1), were significantly enriched in the cell cycle phase and oocyte meiosis pathway. DEGs, including replication factor C subunit 2 (RFC2), RFC3, RFC4 and RFC5, were significantly enriched in DNA replication and interacted with each other. RFC4 also interacted with other DEGs, including CDK1, MAD2L1, NDC80 kinetochore complex and BUB1. In addition, RFC4, RFC3 and RFC5 were targeted by miRNA (miR)‑802, miR‑224‑3p and miR‑522‑3p. The DEGs encoding RFC may be important for the development of conventional OS, and their expression may be regulated by a number of miRNAs, including miR‑802, miR‑224‑3p and miR‑522‑3p.

Introduction

Osteosarcoma (OS) is the most common malignancy of bone in early adolescence (1). Conventional OS, also termed classical OS, is a common type of OS and is universally life-threatening due to its rapid growth, high local aggression and metastatic potential (2). During previous years, considerable progress has been made in identifying the key components in conventional OS, including genes, pathways and microRNAs (miRNAs). For example, during osteoblast differentiation, miRNA (miR)-34 is significantly induced by bone morphogenetic protein 2, and regulates multiple components of the Notch signalling pathway, including Notch1, Notch2 and jagged 1, which affects osteoclast differentiation. This regulatory association may be closely associated with the pathogenesis of OS (3). In addition, phosphatase and tensin homolog (PTEN) has been found to be a potent regulator of the phosphatidylinositol 3-kinase (PI3K) /serine-threonine kinase (Akt) pathway (4), and the loss of PTEN is a common occurrence in conventional OS (5). A previous study has showed that the expression of PTEN can be inhibited by miR-221, which potentiates the PI3K/Akt pathway in the conventional pathogenesis of OS (6). PTEN is also a target of miR-92a, and of members of the miR-17 and miR-130/301 families in OS (7).

In 2010, using genome-wide microarrays, Fritsche-Guenther et al (8) found that the aberrant expression of ephrin receptor A2 (EphA2) and its ligand, EFNA1 in OS can modulate the activation of the mitogen-activated protein kinase (MAPK) pathway. In addition, it was found that the expression of CD52 was higher in OS metastases compared with conventional OS metastases, and CAMPATH-1H, an antibody directed against CD52, reduced the number of viable OS cells (9). In 2013, Luo et al (10) found numerous differentially expressed genes (DEGs) and regulatory associations between transcription factors and DEGs in OS using the microarray data deposited by Fritsche-Guenther et al For example, interleukin 6 can be regulated by tumour protein p53 (TP53), nuclear factor I/C (CCAAT-binding transcription factor), retinoic acid receptor α, and CCAAT/enhancer binding protein β. In 2014, Yang et al (11) also identified a number of DEGs, Gene Ontology (GO) terms, including protein binding, and significant pathways, including focal adhesion, in OS based on a meta-analysis of eight expression profiles, including the one deposited by Fritsche-Guenther (8). However, in these previous studies, the potential miRNAs and regulatory associations between miRNAs and DEGs in OS were not examined.

In the present study, to screen and identify additional DEGs and miRNAs in conventional OS, the microarray data deposited by Fritsche-Guenther (8) were downloaded. Following GO and pathway enrichment analyses, and construction of a protein-protein interaction (PPI) network for the DEGs, the potential miRNAs in the most significant pathway for the upregulated DEGs were identified, and a regulatory network for the miRNAs-DEGs was constructed. The results were expected to assist in elucidating the aetiology of conventional OS, and provide more information to assist in the clinical diagnosis and treatment of this disease.

Materials and methods

Affymetrix microarray data

The GSE14359 (8) gene expression profile data were acquired from the Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/), which was based on the platform of the GPL96 [HG-U133A] Affymetrix Human Genome U133A Array. This dataset contains 10 conventional OS samples from the femur or tibia (two replicates each) from five consenting patients with grade 2–3 conventional OS between 7 and 74 years of age; eight OS lung metastasis tumour samples (two replicates each) from four consenting patients with a grade 1–3 OS lung metastatic tumour; and two non-neoplastic primary osteoblast cell samples with limited life span in vitro from one patient (two replicates). These 10 conventional OS samples and two non-neoplastic primary osteoblast samples were selected for further analysis.

The CEL files and probe annotation files were downloaded, and the gene expression data of all samples were preprocessed via background correction, quantile normalization and probe summarization using the Gene Chip Robust Multi Array algorithm (12) in the Affy software package (version 1.32.0; http://www.bioconductor.org/packages/release/bioc/html/affy.html) (13).

DEG screening

The Linear Models for Microarray Data package (version 3.10.3; http://www.bioconductor.org/packages/2.9/bioc/html/limma.html) (14) of R was used to identify genes, which were significantly differentially expressed in the conventional OS samples. The raw P-value was adjusted by the Benjamin and Hochberg method (15), and only genes meeting the cut-off criteria of |log2 fold-change|>1 and adjusted P<0.01 were selected as DEGs.

Hierarchical clustering analysis of the DEGs

Hierarchical clustering is a common method used to determine clusters of similar data points in multidimensional spaces (16). The pheatmap package (version 1.08; https://cran.r-project.org/web/packages/pheatmap/) (17) was used to perform hierarchical clustering via joint between-within distances for the DEGs in the conventional OS and non-neoplastic primary osteoblasts samples.

GO and pathway enrichment analyses

The Database for Annotation, Visualization and Integrated Discovery (DAVID) provides a set of comprehensive functional annotation tools, which can be used to identify the biological meanings of abundant genes (18). P<0.01 was used as the cut-off criterion for GO and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis using DAVID (version 6.7; https://david-d.ncifcrf.gov/), based on the hypergeometric distribution algorithm.

PPI network construction

The Search Tool for the Retrieval of Interacting Genes database (version 10.0; http://string-db.org/), which provides experimental and predicted interaction information (19), was used to analyse the PPIs for DEGs by calculating the combined score, and a score >0.4 was selected as the cut-off criterion. Subsequently, the PPI network of the upregulated and downregulated DEGs was visualized using Cytoscape (version 3.2.0; http://cytoscape.org/) (20).

Screening and analysis of network modules

The network modules were obtained based on Molecular Complex Detection (MCODE) analysis (21) of the original PPI networks. The default parameters (degree cut-off, 2; node score cut-off, 0.2; K-core, 2) were used as the cut-off criteria for the network module screening.

In order to obtain further information on the gene functions and identify pathways closely associated with the DEGs, functional annotation analysis and subsequent pathway enrichment analysis of the network module with the highest MCODE score were performed using DAVID, with a P<0.01 cut-off.

Integrated miRNA-DEG regulatory network construction

The potential miRNAs for upregulated DEGs in the most significant pathway were predicted using the miRDB database (version 1.24.0; http://www.bioconductor.org/packages/2.8/bioc/html/maDB.html) (22), with a cut-off for the target score of ≥60. The binding sites of miRNAs in the human mRNAs > 800 were abandoned. The integrated miRNA-DEG regulatory network was then visualised with Cytoscape.

Results

Identification of DEGs

Following the data preprocessing, 11,107 probes were obtained. Based on the cut-off criteria, a total of 987 DEGs were screened from the conventional OS samples, including 317 upregulated genes and 670 downregulated genes. The hierarchical cluster analysis of the data revealed that it was possible to use the DEGs to accurately distinguish the conventional OS samples from the non-neoplastic primary osteoblast cell samples (Fig. 1).

Enrichment analysis of upregulated and downregulated DEGs

According to the GO functional annotation, the upregulated DEGs were predominantly enriched in GO terms associated with DNA replication, including MCM3, replication factor C (RFC)5, replication protein A3 (RPA3) and flap endonuclease 1 (FEN1), and cell cycle, including cyclin-dependent kinase 1 (CDK1), NDC80 kinetochore complex (NDC80), BUB1 mitotic checkpoint serine/threonine-protein kinase (BUB1) and mitotic arrest deficient 2 like 1 (MAD2L1). A number of downregulated DEGs, including caveolin 1 (CAV1), cadherin 13 (CDH13), vascular endothelial growth factor C (VEGFC) and transforming growth factor β receptor 3 (TGFBR3), were relevant to blood vessel development, whereas epidermal growth factor receptor (EGFR), TP53, VEGFB and MAPK1 were associated with the regulation of cell proliferation (Table IA).

Table I.

GO terms and pathways enriched for the upregulated and downregulated DEGs.

Table I.

GO terms and pathways enriched for the upregulated and downregulated DEGs.

A, Top 10 GO terms for the upregulated and downregulated DEGs

CategoryTermDescriptionP-valuenGenes
UpGO:0006259DNA metabolic process8.09E-1338RPA3, FANCL, FEN1, DTL, CENPF, MCM3, RFC5, RFC3, RFC4, RFC2
GO:0006260DNA replication3.90E-1021DTL, CENPF, MCM3, RPA3, RFC5, RFC3, RFC4, RFC2, RRM2, FEN1
GO:0022403Cell cycle phase8.15E-1030AURKA, NCAPG, BUB1, CDK1, KIF11, DLGAP5, CENPF, BIRC5, NDC80, MAD2L1
GO:0000278Mitotic cell cycle1.34E-0928NCAPG, BUB1, CDK1, KIF11, CENPF, BIRC5, NDC80, CDKN3, MAD2L1, ZWINT
GO:0051301Cell division1.36E-0925NCAPG, BUB1, ASPM, CDK1, KIF11, CENPF, BIRC5, NDC80, MCM5, MAD2L1
GO:0000280Nuclear division5.16E-0921CDK1, KIF11, CENPF, CDC23, NDC80, BIRC5, SMC4, MAD2L1, NCAPG, BUB1
GO:0007067Mitosis5.16E-0921CDK1, KIF11, CENPF, CDC23, NDC80, BIRC5, SMC4, MAD2L1, NCAPG, BUB1
GO:0000087M phase of mitotic cell cycle7.03E-0921CDK1, KIF11, CENPF, CDC23, NDC80, BIRC5, SMC4, MAD2L1, NCAPG, BUB1
GO:0048285Organelle fission1.03E-0821CDK1, KIF11, CENPF, CDC23, NDC80, BIRC5, SMC4, MAD2L1, NCAPG, BUB1
GO:0000279M phase1.18E-0825NCAPG, BUB1, CDK1, KIF11, DLGAP5, CDC23, CENPF, BIRC5, NDC80, MAD2L1
DownGO:0001568Blood vessel development1.57E-0628CAV1, THBS1, MMP14, PNPLA6, CDH13, VEGFC, NTRK2, TGFBR3, ENG, TNFAIP2
GO:0042127Regulation of cell proliferation1.63E-0660EGFR, CTBP1, TP53, MFGE8, HOXC10, VEGFB, MAPK1, VEGFC, SMAD3, SMAD2
GO:0007242Intracellular signaling cascade2.08E-0684RRAS, TP53, CAV1, MAPKAPK3, FHL2, TGFBR3, GRK5, ABL1, CRK, IGFBP5
GO:0010033Response to organic substance2.28E-0656TIMP3, STAT6, SRR, PPP3CB, COL6A2, SMAD2, CDH13, ADCY9, SMPD1, TGFBR3
GO:0051270Regulation of cell motion2.47E-0624SMAD3, ACTN1, MAPK1, VEGFC, SEMA3F, TGFBR3, RRAS, THBS1, IGFBP3, IGFBP5
GO:0001944Vasculature development2.52E-0628CAV1, MYH9, MMP14, PNPLA6, CDH13, VEGFC, NTRK2, TGFBR3, ENG, TNFAIP2
GO:0008285Negative regulation of cell proliferation7.25E-0634CAV1, TP53, SMAD3, SMAD2, TGFBR3, ADAMTS1, IGFBP3, ENG, IGFBP5, TOB1
GO:0040007Growth1.22E-0522SMAD2, LAMB2, NUPR1, DHCR7, SERPINE1, TGFBR3, BIN3, ADD1, IGFBP5, ERCC2
GO:0030334Regulation of cell migration1.24E-0521EGFR, SMAD3, MAPK1, VEGFC, PTP4A1, RRAS, TGFBR3, THBS1, IGFBP3, IGFBP5
GO:0048514Blood vessel morphogenesis3.44E-0523CAV1, CDH13, VEGFC, SEMA3C, PLCD1, NR2F2, THBS1, TNFAIP2, ENG, CYR61

B, Pathways enriched for the upregulated and downregulated DEGs

CategoryTermDescriptionP-valuenGenes

Uphsa03030DNA replication1.50E-06  9RFC5, RFC3, RFC4, POLE3, RFC2, MCM3, FEN1, MCM5, RPA3
hsa04110Cell cycle1.77E-0412CDC7, CDK1, CDKN1B, MAD2L1, CREBBP, BUB1, PRKDC, CDC23, MCM3, SMC1A
hsa03040Spliceosome1.90E-0412RBM22, HNRNPA3, SF3B1, SNRPA1, MAGOH, TRA2B, SNRNP200, LSM5, LSM3, SNRPE
hsa03430Mismatch repair1.96E-03  5RFC5, RFC3, RFC4, RFC2, RPA3
hsa03420Nucleotide excision repair3.76E-03  6RFC5, RFC3, RFC4, POLE3, RFC2, RPA3
hsa04114Oocyte meiosis4.63E-03  9CDK1, MAD2L1, SLK, BUB1, CDC23, AURKA, PPP1CC, SMC1A, PPP1CB
hsa03410Base excision repair9.30E-03  5HMGB1, POLE3, TDG, POLB, FEN1
Downhsa04510Focal adhesion4.08E-0729CAV1, COL6A1, THBS1, FN1, EGFR, FLNC, VEGFB, LAMA2, MAPK1, VEGFC
hsa04142Lysosome1.74E-0417SGSH, CLN3, NAGLU, PLA2G15, CLTB, PSAP, CTSA, GLB1, DNASE2, LAMP1
hsa04520Adherens junction3.35E-0413EGFR, WASF3, SMAD3, SMAD2, CTNNA1, TCF7L1, CSNK2A2, MAPK1, FYN, MAPK3
hsa04115p53 signaling pathway4.26E-0412CCND1, TNFRSF10B, ZMAT3, SERPINE1, DDB2, TP53, FAS, PERP, THBS1, IGFBP3
hsa05219Bladder cancer8.61E-04  9EGFR, VEGFB, MAPK1, VEGFC, CCND1, MAP2K2, MAPK3, TP53, THBS1
hsa04540Gap junction1.28E-0313ADCY3, EGFR, GNAI2, MAP2K2, GNA11, LPAR1, ITPR3, MAPK1, ADCY9, CSNK1D
hsa00980Metabolism of xenobiotics by cytochrome P4502.44E-0310GSTM1, AKR1C3, GSTM2, AKR1C2, CYP1B1, ADH5, GSTT1, EPHX1, AKR1C1, ALDH3B1
hsa05216Thyroid cancer2.48E-03  7MAPK1, CCND1, MAP2K2, RXRA, MAPK3, TP53, TCF7L1
hsa05212Pancreatic cancer2.57E-0311EGFR, VEGFB, MAPK1, VEGFC, CCND1, RELA, MAPK3, TP53, SMAD3, SMAD2
hsa05220Chronic myeloid leukemia3.49E-0311MAPK1, CTBP1, CCND1, MAP2K2, RELA, MAPK3, TP53, SMAD3, BCL2L1, ABL1

[i] DEGs, differentially expressed genes; GO, Gene Ontology; Up, upregulated; Down, downregulated.

According to the results of the pathway enrichment analysis, the upregulated DEGs were predominantly enriched in seven pathways. In accordance with the GO term analysis, the DNA replication pathway, including RFC2, RFC3, RFC4 and RFC5, and cell cycle pathway, including CDK1, minichromosome maintenance complex component 3 (MCM3) and BUB1, were also enriched in the upregulated genes. The downregulated DEGs were predominantly enriched in the focal adhesion, including CAV1, collagen type VI α1 (COL6A1), thrombospondin 1 (THBS1) and EGFR, and p53 signalling pathways, including TP53, Fas cell surface death receptor (FAS) and TP53 apoptosis effector (PERP), as shown in Table IB.

Construction and analysis of the PPI network

The PPI network for the upregulated and downregulated DEGs consisted of 442 pairs of PPIs. The degrees of DEGs, including CDK1, MAD2L1, NDC80, non-SMC condensin I complex subunit G (NCAPG), BUB1, centromere protein F (CENPF) and kinesin family member 11 (KIF11), were >17 (Table II), indicating that they were important genes in OS.

Table II.

Differentially expressed genes with a connectivity degree of ≥10 in the protein-protein interaction network.

Table II.

Differentially expressed genes with a connectivity degree of ≥10 in the protein-protein interaction network.

IDDegree
CDK129
MAD2L123
NDC8020
NCAPG20
BUB119
CENPF19
KIF1118
DLGAP517
CREBBP17
BIRC517
RFC416
RRM216
TP5316
AURKA16
SF3A214
ASPM14
SNRPG13
MAPK112
HMMR11
NUP10711
CDKN311
PPP1CC11
SRSF111
RACGAP110
NUP16010
ZWINT10
SRSF310
Analysis of network modules

A total of 10 network modules were obtained from the PPI network using the default criteria, and the module with the highest score contained 16 nodes and 102 edges. In this module, CDK1 interacted with other DEGs, including MAD2L1, BUB1, NCAPG, NDC80 and CENPF (Fig. 2).

The functional enrichment analysis of the module with the highest score showed that the majority of the DEGs in this module were predominantly associated with the cell cycle. Certain DEGs, including CDK1, MAD2L1, BUB1 and NDC80, were implicated in mitosis and the M phase of mitotic cell cycle; other DEGs, including Rac GTPase-activating protein 1 (RACGAP1) and MAD2L1, were correlated with cell cycle process (Table IIIA). CDK1, MAD2L1, BUB1 and aurora kinase A (AURKA) were significantly enriched in the oocyte meiosis pathway (Table IIIB).

Table III.

GO terms and pathways enriched for the DEGs in the most significant module.

Table III.

GO terms and pathways enriched for the DEGs in the most significant module.

A, Top 10 GO terms enriched for the DEGs in the most significant module

TermDescriptionP-valuenGenes
GO:0000280Nuclear division9.92E-1611CDK1, MAD2L1, KIF11, NCAPG, DLGAP5, BUB1, CENPF, BIRC5, NDC80, AURKA
GO:0007067Mitosis9.92E-1611CDK1, MAD2L1, KIF11, NCAPG, DLGAP5, BUB1, CENPF, BIRC5, NDC80, AURKA
GO:0000087M phase of mitotic cell cycle1.19E-1511CDK1, MAD2L1, KIF11, NCAPG, DLGAP5, BUB1, CENPF, BIRC5, NDC80, AURKA
GO:0048285Organelle fission1.49E-1511CDK1, MAD2L1, KIF11, NCAPG, DLGAP5, BUB1, CENPF, BIRC5, NDC80, AURKA
GO:0000278Mitotic cell cycle1.86E-1512CDK1, MAD2L1, KIF11, NCAPG, DLGAP5, BUB1, CENPF, BIRC5, NDC80, AURKA
GO:0022402Cell cycle process2.11E-1513CDK1, KIF11, DLGAP5, CENPF, AURKA, NDC80, BIRC5, MAD2L1, NCAPG, BUB1
GO:0022403Cell cycle phase6.44E-1512CDK1, MAD2L1, KIF11, NCAPG, DLGAP5, BUB1, BIRC5, NDC80, AURKA, ASPM
GO:0000279M phase5.81E-1411CDK1, MAD2L1, KIF11, NCAPG, DLGAP5, BUB1, CENPF, BIRC5, NDC80, AURKA
GO:0007049Cell cycle9.57E-1413CDK1, KIF11, DLGAP5, CENPF, AURKA, NDC80, BIRC5, MAD2L1, NCAPG, BUB1
GO:0051301Cell division1.80E-1210CDK1, MAD2L1, KIF11, NCAPG, BUB1, CENPF, BIRC5, NDC80, RACGAP1, ASPM

B, Pathways enriched for the DEGs in the most significant module

TermDescriptionP-valuenGenes

hsa04114Oocyte meiosis1.88E-04  4CDK1, MAD2L1, BUB1, AURKA
hsa04914 Progesterone-mediated oocyte maturation4.06E-03  3CDK1, MAD2L1, BUB1
hsa04110Cell cycle8.43E-03  3CDK1, MAD2L1, BUB1

[i] DEGs, differentially expressed genes; GO, Gene Ontology.

Analysis of the miRNA-DEG regulatory network

The miRNA-DEG regulatory network contained 63 miRNAs, nine of their corresponding DEGs and 16 DEGs, which interacted with these nine DEGs. DNA polymerase ζ subunit 3 (POLE3) was regulated by 18 miRNAs, including miR-4310, miR-4680-3p, miR-583 and miR-4269; RFC3 was regulated by 16 miRNAs, including miR-802 and miR-649; RFC3 and RFC5 were modulated by miR-522-3p and miR-224-3p. In addition, RFC2, RFC3, RFC4 and RFC5 interacted with each other (Fig. 3).

Discussion

In the present study, 317 DEGs were found to be significantly upregulated and 670 were significantly downregulated in the conventional OS samples. A majority of the DEGs were associated with cell cycle. According to the miRNA-DEG regulatory network for the DEGs enriched in DNA replication, RFC2, RFC3, RFC4 and RFC5 were found to interact with each other.

RFC2, RFC3, RFC4 and RFC5 encode members of RFC family, also termed activator 1. These DEGs were enriched in DNA replication, which agreed with the results of previous studies (23,24). DNA replication is an essential event in tumour growth (25). The deregulation of protein complexes involved in the initiation of DNA replication can lead to cancer (26). Several DEGs in the network module, including CDK1, MAD2L1, NDC80 and BUB1, which had higher degrees in the PPI network, were found to interact with RFC4. These four DEGs were predominantly enriched in cell mitosis and cell cycle. Alterations in cell cycle regulation occur in several types of cancer, including OS (27). Cyclin-dependent kinase 1 (CDK1) is an important G2/M checkpoint protein (28), and its inhibitor, SCH 727965 (dinacliclib) can trigger the apoptosis of U-2 OS cells (29). MAD2L1, BUB1 and NDC80 are involved in the spindle checkpoint pathway (30,31). MAD2 has been reported to be commonly overexpressed in human conventional OS (32), and BUB1 has been found to be ectopically expressed in SAOS and U-2 OS cell lines (33). In addition, CDK1, MAD2L1 and BUB1 have been found to be significantly enriched in the pathway of oocyte meiosis, which was found to be markedly altered in high-grade OS cell lines when compared with osteoblasts (34). RFC3 was also modulated by a cluster of miRNAs, including miR-802. The expression of miR-802 has been reported to be upregulated in OS tissues, and to promote cell proliferation by targeting p27 in U-2 OS and MG-63 cells (35). RFC3 and RFC5 are also modulated by miR-224-3p and miR-522-3p. There is no previous evidence indicating that miR-224-3p and miR-522-3p are associated with conventional OS. Therefore, miR-224-3p and miR-522-3p are predicted to be novel biomarkers in conventional OS. Therefore, RFC2-5, together with certain DEGs, including CDK1, MAD2L1, NDC80 and BUB1, and a series of miRNAs, including miR-802, miR-224-3p and miR-522-3p, may be responsible for the initiation and development of conventional OS.

In conclusion, the present study found that the majority of DEGs, including CDK1, MAD2L1, NDC80 and BUB1, were associated with the cell cycle. Other DEGs, including RFC2, RFC3, RFC4 and RFC5, were associated with DNA replication. These, in addition to a number of miRNAs, including miR-802, miR-224-3p and miR-522-3p, may be essential in the pathogenesis of conventional OS, providing novel information to assist in the clinical diagnosis of this disease. However due to limitations in the present study, additional experiments are required to shed light on the molecular mechanisms involved in this life-threatening disease.

References

1 

Messerschmitt PJ, Garcia RM, Abdul-Karim FW, Greenfield EM and Getty PJ: Osteosarcoma. J Am Acad Orthop Surg. 17:515–527. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Mohseny AB, Tieken C, Van Der Velden PA, Szuhai K, de Andrea C, Hogendoorn PC and Cleton-Jansen AM: Small deletions but not methylation underlie CDKN2A/p16 loss of expression in conventional osteosarcoma. Genes Chromosomes Cancer. 49:1095–1103. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Bae Y, Yang T, Zeng HC, Campeau PM, Chen Y, Bertin T, Dawson BC, Munivez E, Tao J and Lee BH: miRNA-34c regulates Notch signaling during bone development. Hum Mol Genet. 21:2991–3000. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Silva A, Yunes JA, Cardoso BA, Martins LR, Jotta PY, Abecasis M, Nowill AE, Leslie NR, Cardoso AA and Barata JT: PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability. J Clin Invest. 118:3762–3774. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Freeman SS, Allen SW, Ganti R, Wu J, Ma J, Su X, Neale G, Dome JS, Daw NC and Khoury JD: Copy number gains in EGFR and copy number losses in PTEN are common events in osteosarcoma tumors. Cancer. 113:1453–1461. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Zhao G, Cai C, Yang T, Qiu X, Liao B, Li W, Ji Z, Zhao J, Zhao H, Guo M, et al: MicroRNA-221 induces cell survival and cisplatin resistance through PI3K/Akt pathway in human osteosarcoma. PLoS One. 8:e539062013. View Article : Google Scholar : PubMed/NCBI

7 

Namløs HM, Meza-Zepeda LA, Barøy T, Østensen IH, Kresse SH, Kuijjer ML, Serra M, Bürger H, Cleton-Jansen AM and Myklebost O: Modulation of the osteosarcoma expression phenotype by microRNAs. PLoS One. 7:e480862012. View Article : Google Scholar : PubMed/NCBI

8 

Fritsche-Guenther R, Noske A, Ungethüm U, Kuban RJ, Schlag PM, Tunn PU, Karle J, Krenn V, Dietel M and Sers C: De novo expression of EphA2 in osteosarcoma modulates activation of the mitogenic signalling pathway. Histopathology. 57:836–850. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Fritsche-Guenther R, Gruetzkau A, Noske A, Melcher I, Schaser KD, Schlag PM, Kasper HU, Krenn V and Sers C: Therapeutic potential of CAMPATH-1H in skeletal tumours. Histopathology. 57:851–861. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Luo Y, Deng Z and Chen J: Pivotal regulatory network and genes in osteosarcoma. Arch Med Sci. 9:569–575. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Yang Z, Chen Y, Fu Y, Yang Y, Zhang Y, Chen Y and Li D: Meta-analysis of differentially expressed genes in osteosarcoma based on gene expression data. BMC Med Genet. 15:802014. View Article : Google Scholar : PubMed/NCBI

12 

Wu Z, Irizarry RA, Gentleman R, Martinez-Murillo F and Spencer F: A model-based background adjustment for oligonucleotide expression arrays. Journal of the American Statistical Association. 99:909–917. 2004. View Article : Google Scholar

13 

Seo J and Hoffman EP: Probe set algorithms: Is there a rational best bet? BMC bioinformatics. 7:3952006. View Article : Google Scholar : PubMed/NCBI

14 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:Article32004. View Article : Google Scholar : PubMed/NCBI

15 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: A practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B (Methodological). 57:289–300. 1995.

16 

Olson CF: Parallel algorithms for hierarchical clustering. Parallel Computing. 21:1313–1325. 1995. View Article : Google Scholar

17 

Kolde R: pheatmap: Pretty Heatmaps. R package. version 0.7. 7. 2012.

18 

Huang DW, Sherman BT, Tan Q, Collins JR, Alvord WG, Roayaei J, Stephens R, Baseler MW, Lane HC and Lempicki RA: The DAVID gene functional classification tool: A novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 8:R1832007. View Article : Google Scholar : PubMed/NCBI

19 

Von Mering C, Huynen M, Jaeggi D, Schmidt S, Bork P and Snel B: STRING: A database of predicted functional associations between proteins. Nucleic Acids Res. 31:258–261. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

22 

Wang X: miRDB: A microRNA target prediction and functional annotation database with a wiki interface. RNA. 14:1012–1017. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Reynolds N, Fantes PA and MacNeill SA: A key role for replication factor C in DNA replication checkpoint function in fission yeast. Nucleic Acids Res. 27:462–469. 1999. View Article : Google Scholar : PubMed/NCBI

24 

Redondo-Muñoz J, Rodríguez MJ, Silió V, Pérez-García V, Valpuesta JM and Carrera AC: Phosphoinositide 3-kinase beta controls replication factor C assembly and function. Nucleic Acids Res. 41:855–868. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Loeb LA, Springgate CF and Battula N: Errors in DNA replication as a basis of malignant changes. Cancer Res. 34:2311–2321. 1974.PubMed/NCBI

26 

Tsaniras S Champeris, Kanellakis N, Symeonidou IE, Nikolopoulou P, Lygerou Z and Taraviras S: Licensing of DNA replication, cancer, pluripotency and differentiation: An interlinked world? Semin Cell Dev Biol. 30:174–180. 2014. View Article : Google Scholar : PubMed/NCBI

27 

PosthumaDeBoer J, van Royen B and Helder M: Mechanisms of therapy resistance in osteosarcoma: A review. Oncol Discov. 1:82013. View Article : Google Scholar

28 

Kim MJ, Lee JY and Lee SJ: Transient suppression of nuclear Cdc2 activity in response to ionizing radiation. Oncol Rep. 19:1323–1329. 2008.PubMed/NCBI

29 

Fu W, Ma L, Chu B, Wang X, Bui MM, Gemmer J, Altiok S and Pledger WJ: The cyclin-dependent kinase inhibitor SCH 727965 (dinacliclib) induces the apoptosis of osteosarcoma cells. Mol Cancer Ther. 10:1018–1027. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Doak SH, Jenkins GJ, Parry EM, Griffiths AP, Baxter JN and Parry JM: Differential expression of the MAD2, BUB1 and HSP27 genes in Barrett's oesophagus-their association with aneuploidy and neoplastic progression. Mutat Res. 547:133–144. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Giantin M, Granato A, Baratto C, Marconato L, Vascellari M, Morello EM, Vercelli A, Mutinelli F and Dacasto M: Global gene expression analysis of canine cutaneous mast cell tumor: Could molecular profiling be useful for subtype classification and prognostication? PLoS One. 9:e954812014. View Article : Google Scholar : PubMed/NCBI

32 

Yu L, Guo WC, Zhao SH, Tang J and Chen JL: Mitotic arrest defective protein 2 expression abnormality and its clinicopathologic significance in human osteosarcoma. Apmis. 118:222–229. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Trougakos IP, Chondrogianni N, Amarantos I, Blake J, Schwager C, Wirkner U, Ansorge W and Gonos ES: Genome-wide transcriptome profile of the human osteosarcoma Sa OS and U-2 OS cell lines. Cancer Genet Cytogenet. 196:109–118. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Kuijjer ML, Peterse EF, van den Akker BE, Briaire-de Bruijn IH, Serra M, Meza-Zepeda LA, Myklebost O, Hassan AB, Hogendoorn PC and Cleton-Jansen AM: IR/IGF1R signaling as potential target for treatment of high-grade osteosarcoma. BMC Cancer. 13:2452013. View Article : Google Scholar : PubMed/NCBI

35 

Cao ZQ, Shen Z and Huang WY: MicroRNA-802 promotes osteosarcoma cell proliferation by targeting p27. Asian Pac J Cancer Prev. 14:7081–7084. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 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
Peng C, Yang Q, Wei B, Yuan B, Liu Y, Li Y, Gu D, Yin G, Wang B, Xu D, Xu D, et al: Investigation of crucial genes and microRNAs in conventional osteosarcoma using gene expression profiling analysis. Mol Med Rep 16: 7617-7624, 2017
APA
Peng, C., Yang, Q., Wei, B., Yuan, B., Liu, Y., Li, Y. ... Kong, D. (2017). Investigation of crucial genes and microRNAs in conventional osteosarcoma using gene expression profiling analysis. Molecular Medicine Reports, 16, 7617-7624. https://doi.org/10.3892/mmr.2017.7506
MLA
Peng, C., Yang, Q., Wei, B., Yuan, B., Liu, Y., Li, Y., Gu, D., Yin, G., Wang, B., Xu, D., Zhang, X., Kong, D."Investigation of crucial genes and microRNAs in conventional osteosarcoma using gene expression profiling analysis". Molecular Medicine Reports 16.5 (2017): 7617-7624.
Chicago
Peng, C., Yang, Q., Wei, B., Yuan, B., Liu, Y., Li, Y., Gu, D., Yin, G., Wang, B., Xu, D., Zhang, X., Kong, D."Investigation of crucial genes and microRNAs in conventional osteosarcoma using gene expression profiling analysis". Molecular Medicine Reports 16, no. 5 (2017): 7617-7624. https://doi.org/10.3892/mmr.2017.7506