Open Access

Exploring the molecular mechanisms of osteosarcoma by the integrated analysis of mRNAs and miRNA microarrays

  • Authors:
    • Hao Shen
    • Wei Wang
    • Bingbing Ni
    • Qiang Zou
    • Hua Lu
    • Zhanchao Wang
  • View Affiliations

  • Published online on: March 27, 2018     https://doi.org/10.3892/ijmm.2018.3594
  • Pages: 21-30
  • Copyright: © Shen 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

Osteosarcoma (OS) is the most frequently occurring primary bone malignancy with a rapid progression and poor survival. In the present study, in order to examine the molecular mechanisms of OS, we analyzed the microarray of GSE28425. GSE28425 was downloaded from Gene Expression Omnibus, which also included the miRNA expression profile, GSE28423, and the mRNA expression profile, GSE28424. Each of the expression profiles included 19 OS cell lines and 4 normal bones. The differentially expressed genes (DEGs) and differentially expressed miRNAs (DE-miRNAs) were screened using the limma package in Bioconductor. The DEGs associated with tumors were screened and annotated. Subsequently, the potential functions of the DEGs were analyzed by Gene Ontology (GO) and pathway enrichment analyses. Furthermore, the protein-protein interaction (PPI) network was constructed using the STRING database and Cytoscape software. Furthermore, modules of the PPI network were screened using the ClusterOne plugin in Cytoscape. Additionally, the transcription factor (TF)-DEG regulatory network, DE-miRNA-DEG regulatory network and miRNA-function collaborative network were separately constructed to obtain key DEGs and DE-miRNAs. In total, 1,609 DEGs and 149 DE-miRNAs were screened. Upregulated FOS-like antigen 1 (FOSL1) also had the function of an oncogene. MAD2 mitotic arrest deficient-like 1 (MAD2L1; degree, 65) and aurora kinase A (AURKA; degree, 64) had higher degrees in the PPI network of the DEGs. In the TF-DEG regulatory network, the TF, signal transducer and activator of transcription 3 (STAT3) targeted the most DEGs. Moreover, in the DE-miRNA-DEG regulatory network, downregulated miR‑1 targeted many DEGs and estrogen receptor 1 (ESR1) was targeted by several highly expressed miRNAs. Moreover, in the miRNA-function collaborative networks of upregulated miRNAs, miR‑128 targeted myeloid dendritic associated functions. On the whole, our data indicate that MAD2L1, AURKA, STAT3, ESR1, FOSL1, miR‑1 and miR‑128 may play a role in the development and/or progressio of OS.

Introduction

As a high-grade type and mesenchymally-derived bone sarcoma (1), osteosarcoma (OS) is the most prevalent primary bone cancer and the 8th most frequent type of cancer affecting young patients (2). Being characterized by a high malignant degree, rapid progression and a poor survival, OS consists up to 15% of all solid extracranial cancers in patients aged 15–19 years (3,4). Thus, it is necessary to identify biomarkers involved in OS.

DNA repair gene RecQ protein-like 4 (RECQL4) is overexpressed in OS, and its overexpression is related to overall genomic instability (5). Human epidermal growth factor receptor 2 (Her-2/neu) expression can induce lung metastasis in OS and may be related to gene amplification (6). Overexpressed c-fos (FOS) and runt-related transcription factor 2 (RUNX2) may play a role in OS; in particular, RUNX2 expression may serve as a marker of chemotherapy failure in patients with OS (7,8). The cell cycle regulator, CDC5 cell division cycle 5-like (CDC5L), is essential for the G2-M transition and may be potential oncogene for the 6p12-p21 amplicon in OS (9). It has been reported that genes with the function of transcription factors (TFs) can also play a role in OS, such as Yin Yang 1 (YY1), which is expressed in the early process of osteoblastic transformation and its detection may be used as a promising diagnostic method in human OS (10). In addition, the TF osterix (Osx) can suppress the lung migration of OS tumor cells; thus, the expression of Osx may be implicated in the growth and metastasis of OS (11).

There are also many studies which have investigated the direct or indirect effect of microRNAs (miRNAs or miRs) on OS. For example, by targeting matrix metalloprotease 13 (MMP13) and B-cell CLL/lymphoma 2 (Bcl-2), miR-143 may be involved in the lung metastasis of human OS cells and may thus be used as a target in cancer therapy (12,13). In addition, downregulated miR-199a-3p may function in the growth and proliferation of OS cells; hence, restoring the function of miR-199a-3p may contribute to the treatment of OS (14). By mediating reversion-inducing-cysteine-rich protein with kazal motifs (RECK), miR-21 plays an important role in regulating cell invasion and migration in OS and may be a potential therapeutic target (15). By regulating c-Met and other genes, miR-34a can function as a tumor suppressor gene and suppresses the pulmonary metastasis of OS; thus, it may be a useful gene therapeutic agent (16).

In 2012, Namløs et al (17) used global microarray analyses to identify the differentially expressed miRNAs (DE-miRNAs) between OS cell lines and normal bones, and obtained 177 DE-miRNAs. In this study, using the same data by Namløs et al (17), we aimed to further screen the differentially expressed genes (DEGs) and DE-miRNAs. The potential functions of the DEGs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Subsequently, the interaction associations of the proteins encoded by the DEGs were investigated by protein-protein interaction network (PPI) network and modules of PPI network. In addition, the TF-DEG regulatory network, DE-miRNA-DEG regulatory network and miRNA-function collaborative network were separately constructed to obtain key DEGs and DE-miRNAs.

Data collection methods and analysis

Microarray data

The microarray of GSE28425 deposited by Namløs et al (17) was downloaded from Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo/), which included the miRNA expression profile, GSE28423, and the mRNA expression profile, GSE28424. Each of the expression profiles included a collection of 19 OS cell lines and 4 normal bones. GSE28423 was based on the platform of GPL8227 Agilent-019118 Human miRNA Microarray 2.0 G4470B (Agilent Technologies Inc., Santa Clara, CA, USA). GSE28424 was based on the platform of GPL13376 Illumina HumanWG-6 v2.0 expression beadchip (Illumina, San Diego, CA, USA). The OS samples were from a panel collected within EuroBoNeT and from the Norwegian Radium Hospital. Meanwhile, normal bones were from Capital Biosciences or from amputations of cancer patients at the University College London and Norwegian Radium Hospital.

Screening of DEGs and DE-miRNAs

After GSE28425 was downloaded, the microarray data was pre-processed using the limma package (18) in Bioconductor (http://www.bioconductor.org/packages/release/bioc/html/limma.html). In brief, the pre-processing process included Background Correction, Quantile Normalization and Probe Summarization. The limma (linear models for microarray data) package (18) was used to analyze the DEGs and DE-miRNAs between the OS cell lines and normal bones. The FDR (that is, adjusted p-value) <0.05 and |log2fold-change (FC)| >1 were used as the cut-off criteria. Screening the tumor suppressor (TS) gene (http://bioinfo.mc.vanderbilt.edu/TSGene/download.cgi) (19) and tumor-associated gene (TAG) (http://www.binfo.ncku.edu.tw/TAG/GeneDoc.php) (20) databases, the DEGs associated with tumors were screened and annotated.

Functional and pathway enrichment analysis

GO provides controlled and structured vocabularies which model biological process (BP), cell components (CC) and molecular function (MF) (21). KEGG is a database containing 16 main databases, roughly divided into systems information, chemical information and genomic information (22). GO functional enrichment analyses, which involved the BP, MF and CC categories, as well as KEGG pathway enrichment analyses were performed for the DEGs and the DE-miRNAs. A p-value <0.05 was used as the cut-off criterion.

PPI network and module construction

The interaction associations of the proteins encoded by the DEGs were searched using STRING online software (http://string-db.org; v9.05) (23), and the combined score of >0.7 was used as the cut-off criterion. The PPI network was visualized using Cytoscape software (http://www.cytoscape.org) (24). Modules of the PPI network were screened using the ClusterOne plugin (25) in Cytoscape, and the significant p-value of the modules were set to 1.1E-6.

TF-DEG regulatory network construction

Human TF-gene regulatory pairs were downloaded from the UCSC database (http://genome.ucsc.edu/) (26). The DEGs which can also function as TFs and their target genes were then identified. Moreover, Cytoscape software (24) was used to visualize the the TF-DEG regulatory network.

DE-miRNA-DEG regulatory network construction

By comparing the experimental validated miRNA-mRNA pairs in the miRecords (http://www.mirecords.umn.edu) (27) and mirWalk (http://mirwalk.uni-hd.de/) (28) databases, pairs of DE-miRNAs from the miRNA expression profile, GSE28423, and DEGs from the mRNA expression profile, GSE28424, were obtained. The DE-miRNA-DEG pairs should appear in either miRecords database or mirWalk database.

miRNA-function collaborative network construction

According to the functional enrichment results of the DE-miRNAs, the DE-miRNAs which targeted the genes involved in one BP term were identified. Subsequently, miRNA-function collaborative network was constructed. A p-value <0.01 was used as the cut-off criterion.

Results

DEGs analysis

Compared with normal bones, there were 1,609 DEGs (including 774 upregulated and 835 downregulated mRNAs) and 149 DE-miRNAs (including 76 upregulated and 73 downregulated miRNAs) screened in the OS cell lines. The DEGs associated with tumors were annotated and are listed in Table I. Importantly, upregulated FOS-like antigen 1 (FOSL1) also had the function of an oncogene.

Table I

The DEGs associated with tumors.

Table I

The DEGs associated with tumors.

CategoryOncogeneTSGTAG
UPCDC5L, FOSL1, HMMR, AURKA, MLF1, CDK4, MET, TRIO, NRAS, HOXA10, WHSC1, PIK3CAS100A2, TUSC3, PAWR, LZTS1, YAP1, GADD45GIP1, PTPRG, RND3, DFNA5, HOXB13, BAI2, ZDHHC2, NF2, BCL10, FANCG, AMH, RCN2, HLTF, NME1, REV3L, DAPK3, FH, MEN1, HECA, TRIM3, SCRIB, BRMS1, EXTL3, SMARCB1, PCGF2TFAP2A, BUB1, NKX3-1, DNMT3B, PMS1, SHC1, YEATS4, FADD, C1QBP
DOWNFGF20, LYN, BCL6, TAL1, ESR1, WISP2, LMO2, LCN2, LYL1HSD17B7, PRODH, MAL, DUSP22, TSC22D1, COL4A3, BAI3, BNIP3L, PER1, PAEP, RASSF4, FOXC1, EXTL1, ARHGAP20, CMTM5, NGFR, TXNIP, NOTCH1, MRVI1, MTSS1, MTUS1, PPAP2A, TCF4, ST5, PYHIN1, PRKCD, TGFBR3, CBFA2T3, MT1G, TSPAN32, RASSF2, CEBPA, LTF, RARRES1, MAP4K1, BTG2, PLA2G2A, ZBTB16, SYK, GPX3, PYCARD, H19, PTPN6, C2orf40TAL2, WISP3, STAT3, CBLB, NR4A2, LYST, RGS2, FES, MGP

[i] TSG, tumor suppressor gene; TAG, tumor-associated gene.

Functional and pathway enrichment analysis

The top 5 enriched GO functions in the BP, CC and MF categories separately for the upregulated and downregulated genes are listed in Table II. For the upregulated genes, the enriched functions included cell cycle (p=0), intracellular membrane-bounded organelle (p=0) and catalytic activity (p=3.05E-10). For the downregulated genes, the enriched functions included cell activation (p=0), extracellular region (p=0) and carbohydrate derivative binding (p=1.55E-08).

Table II

The top 5 enriched GO functions in BP, CC and MF categories, as well as the top 10 enriched KEGG pathways separately for the upregulated and downregulated genes.

Table II

The top 5 enriched GO functions in BP, CC and MF categories, as well as the top 10 enriched KEGG pathways separately for the upregulated and downregulated genes.

CategoryTermDescriptionGene no.Gene symbolp-value
UP_BPGO:0007049Cell cycle133KPNA2, UBE2C0
GO:0000278Mitotic cell cycle90CDCA3, E2F72.22E-16
GO:0022402Cell cycle process108FAM83D, SPC251.22E-15
GO:0051301Cell division62UBE2C, CDCA33.86E-14
GO:0048285Organelle fission52FAM83D, SPC257.72E-14
UP_CCGO:0005622Intracellular611TFAP2A, CBS0
GO:0031981Nuclear lumen161CBS, KPNA20
GO:0043231Intracellular membrane-bounded organelle509KPNA2, JPH30
GO:0044422Organelle part384SHROOM3, UBE2C0
GO:0044424Intracellular part607FOXD1, UBE2C0
UP_MFGO:0003824Catalytic activity300PSAT1, UBE2C3.05E-10
GO:0016740Transferase activity123PSAT1, CCNB13.95E-09
GO:0005515Protein binding382TFAP2A, CBS1.13E-07
GO:0032549Ribonucleoside binding115UBE2C, KIF2C3.57E-06
GO:0035639Purine ribonucleoside triphosphate binding114SEPT3, PTK74.27E-06
DOWN_BPGO:0001775Cell activation104GRAP2, IL12RB10
GO:0001816Cytokine production73STAT5B, LIPA0
GO:0002376Immune system process252FGF20, FCGR3A0
GO:0002682Regulation of immune system process153BLK, CD200R10
GO:0002684Positive regulation of immune system process99FCGR3A, GRAP20
DOWN_CCGO:0005576Extracellular region191FGF20, FCGR3A0
GO:0005615Extracellular space98CCL25, APOC20
GO:0005886Plasma membrane316IL12RB1, BLK0
GO:0044421Extracellular region part120IL12RB1, BLK0
GO:0044459Plasma membrane part174OPRD1, MAL0
DOWN_MFGO:0097367Carbohydrate derivative binding29FGF7, TLR21.55E-08
GO:0005515Protein binding417FGF20, HMGN33.54E-08
GO:0046983Protein dimerization activity80ADD2, APOC25.30E-08
GO:0008307Structural constituent of muscle13DMD, MYL49.25E-08
GO:0042803Protein homodimerization activity55MZF1, ADD11.31E-07
UP_KEGG01100Metabolic pathways89CBS, PSAT11.14E-06
00100Steroid biosynthesis7DHCR24, SQLE2.02E-05
03040Spliceosome14CDC5L, SMNDC10.003765459
03008Ribosome biogenesis in eukaryotes10NXT2, NMD30.005664746
00270Cysteine and methionine metabolism6CBS, DNMT3B0.007673661
00510N-Glycan biosynthesis7TUSC3, ALG10B0.009627025
00970Aminoacyl-tRNA biosynthesis8MARS, YARS0.011699718
00620Pyruvate metabolism6ME1, ACAT20.012824724
00290Valine, leucine and isoleucine biosynthesis3BCAT1, VARS, LARS0.014647016
01040Biosynthesis of unsaturated fatty acids4PTPLA, ELOVL5, PTPLB, SCD0.017934809
DOWN_KEGG05150Staphylococcus aureus infection21FCAR, C3AR12.29E-12
04640Hematopoietic cell lineage21IL4R, CR14.15E-08
04145Phagosome27TLR2, NOX14.14E-07
05140Leishmaniasis17CR1, IFNGR19.79E-07
04060Cytokine-cytokine receptor interaction36CCL25, TNFSF83.76E-06
04380Osteoclast differentiation22LILRA6, NOX17.87E-06
04650Natural killer cell mediated cytotoxicity22IFNGR1, NFATC32.14E-05
04514Cell adhesion molecules (CAMs)21MAG, F11R4.77E-05
05310Asthma9MS4A2, EPX4.91E-05
05416Viral myocarditis14DMD, SGCA6.63E-05

[i] GO, Gene Ontology; BP, biological process; CC, cell components; MF, molecular function; KEGG, Kyoto Encyclopedia of Genes and Genomes.

The top 10 enriched KEGG pathways separately for the upregulated and downregulated genes are also listed in Table II. For the upregulated genes, the enriched pathways included metabolic pathways (p=1.14E-06), steroid biosyn-thesis (p=2.02E-05) and spliceosome (p=0.003765459). For the downregulated genes, the enriched pathways included cytokine-cytokine receptor interaction (p=3.76E-06) and osteoclast differentiation (p=7.87E-06).

PPI network and module analysis

The PPI network of the DEGs had 844 nodes and 3,400 interactions. In particular, MAD2 mitotic arrest deficient-like 1 (MAD2L1, degree, 65), cyclin B1 (CCNB1, degree, 65) and aurora kinase A (AURKA, degree, 64) had high degrees in the PPI network. In addition, 3 modules (module 1, module 2 and module 3) of the PPI network were screened (Fig. 1). In module 1, TAO kinase 1 (TAOK1) was the only downregulated gene. The enriched KEGG pathways for the DEGs in module 1 included oocyte meiosis (p=2.04E-08), cell cycle (p=4.16E-08) and progesterone-mediated oocyte maturation (p=0.000112373) (Table III). In module 2, guanine nucleotide binding protein, α inhibiting 1 (GNAI1) and regulator of G-protein signaling 20 (RGS20) were downregulated. The enriched KEGG pathways for the DEGs in module 2 included chemokine signaling pathway (p=0) and cytokine-cytokine receptor interaction (p=9.77E-15) (Table III). Furthermore, the DEGs involved in module 3 were all upregulated genes. The enriched KEGG pathways for the DEGs in module 3 included ribosome (p=1.26E-12) and protein processing in endoplasmic reticulum (p=0.043084724) (Table III).

Table III

The enriched KEGG pathways for the DEGs in module 1, module 2 and module 3 of the PPI network.

Table III

The enriched KEGG pathways for the DEGs in module 1, module 2 and module 3 of the PPI network.

TermDescriptionGene no.Gene symbolp-value
Module 104114Oocyte meiosis7AURKA, SGOL12.04E-08
04110Cell cycle7PCNA, MCM24.16E-08
04914 Progesterone-mediated oocyte maturation4CCNB2, BUB1, MAD2L1, CCNB10.000112373
04115p53 signaling pathway3CCNB2, CCNB1,GTSE10.001071355
03430Mismatch repair2PCNA, EXO10.002163033
03030DNA replication2PCNA, MCM20.00526065
Module 204062Chemokine signaling pathway17ADCY2, CX3CR10
04060Cytokine-cytokine receptor interaction15CX3CR1, CXCR69.77E-15
04080Neuroactive ligand-receptor interaction7OPRD1, P2RY139.93E-05
05150Staphylococcus aureus infection3C3AR1, FPR1,C5AR10.001547235
04916Melanogenesis3ADCY2, POMC, GNAI10.008639105
04620Toll-like receptor signaling pathway3CCL3, CXCL9, CXCL100.008876016
04672Intestinal immune network for IgA production2CCL25, CCR90.017427247
04610Complement and coagulation cascades2C3AR1, C5AR10.034329309
04971Gastric acid secretion2ADCY2, GNAI10.039017822
Module 303010Ribosome8RPL27A, RPL37A1.26E-12
04141Protein processing in endoplasmic reticulum2DDOST, SSR30.043084724

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; DEGs, differentially expressed genes; PPI, protein-protein interaction.

TF-DEG regulatory network analysis

The TF-DEG regulatory network had 311 interactions (involving 10 transcription factors and 285 DEGs) (Fig. 2). Importantly, the TFs, signal transducer and activator of transcription 3 (STAT3, degree, 158) and forkhead box A1 (FOXA1, degree, 106) targeted the most DEGs.

DE-miRNA-DEG regulatory network analysis

The DE-miRNA-DEG regulatory network involved 23 upregulated miRNAs and 64 downregulated miRNAs (Fig. 3). In the DE-miRNA-DEG regulatory network, downregulated miR-1 targeted and activated many DEGs. Moreover, downregulated estrogen receptor 1 (ESR1) was targeted by several high-expressed miRNAs, including miR-221, miR-20b and miR-18a. The enriched GO functions for the upregulated and downregulated miRNAs are listed in Table IV. For the upregulated miRNAs, the enriched functions included positive regulation of retinoic acid receptor signaling pathway (p=0.000211583) and type 1 metabotropic glutamate receptor binding (p=0.000150457). For the downregulated miRNAs, the enriched functions included response to inactivity (p=0.001989302) and potassium ion binding (p=0.006643278).

Table IV

The enriched GO functions for the upregulated and downregulated miRNAs involved in the DE-miRNA-DEG regulatory network.

Table IV

The enriched GO functions for the upregulated and downregulated miRNAs involved in the DE-miRNA-DEG regulatory network.

CategoryTermDescriptionmiRNA no.miRNA symbolp-value
UP_BP0048386Positive regulation of retinoic acid Receptor signaling pathway6miR-221, miR-18a0.000211583
0060523Prostate epithelial cord elongation6miR-20b, miR-18a0.001058491
0060745Mammary gland branching involved in pregnancy6miR-221, miR-20b0.001196154
0001766Membrane raft polarization2miR-125a-5p, miR-1280.002429205
0030885Regulation of myeloid dendritic cell activation2miR-125a-5p, miR-1280.002429205
0030887Positive regulation of myeloid dendritic cell activation2miR-125a-5p, miR-1280.002429205
UP_MF0031798Type 1 metabotropic glutamate receptor binding6miR-221, miR-20b0.000150457
0030235Nitric-oxide synthase regulator activity6miR-19b, miR-20b0.000413057
0035256G-protein coupled glutamate receptor binding6miR-19b, miR-18a0.000413057
0030284Estrogen receptor activity6miR-19a, miR-18a0.002145221
0034056Estrogen response element binding6miR-19b, miR-19a0.003216968
0031779Melanocortin receptor binding3miR-455-5p, miR-125a-5p, miR-4840.009585627
0031781Type 3 melanocortin receptor binding3miR-455-5p, miR-484, miR-125a-5p0.009585627
DOWN_BP0014854Response to inactivity4miR-133b, miR-2060.001989302
0014870Response to muscle inactivity4miR-1, miR-133b0.001989302
0014877Response to muscle inactivity involved in regulation of muscle adaptation4miR-206, miR-10.001989302
0014894Response to denervation involved in regulation of muscle adaptation4miR-1, miR-133b0.001989302
0002368B cell cytokine production2miR-206, miR-10.002474699
0002424T cell mediated immune response to tumor cell2miR-1, miR-2060.002474699
DOWN_ MF0005008Hepatocyte growth factor-activated receptor activity4miR-133b, miR-2060.001415624
0030955Potassium ion binding4miR-206, miR-140-3p0.006643278
0031420Alkali metal ion binding4miR-133b, miR-10.007459611
0003688DNA replication origin binding2miR-206, miR-10.014287542
0031078Histone deacetylase activity (H3-K14 specific)4miR-206, miR-140-3p0.016613831
0032041NAD-dependent histone deacetylase activity (H3-K14 specific)4miR-1, miR-2060.016613831

[i] DEG, differentially expressed gene; BP, biological process; MF, molecular function.

miRNA-function collaborative network analysis

The miRNA-function collaborative networks of upregulated (Fig. 4) and downregulated (Fig. 5) miRNAs were constructed, respectively. In the miRNA-function collaborative networks of upregulated miRNAs, myeloid dendritic associated functions were targeted by miR-128 and miR-125a-5p.

Discussion

In this study, we screened 1,609 DEGs (including 774 upregulated and 835 downregulated mRNAs) and 149 DE-miRNAs (including 76 upregulated and 73 downregulated miRNAs) in the OS cell lines compared with normal bones. Importantly, upregulated FOSL1 also had the function of an oncogene. MAD2L1 (degree, 65) and AURKA (degree, 64) had higher degrees in the PPI network of the DEGs. In the DE-miRNA-DEG regulatory network, downregulated miR-1 targeted many DEGs and ESR1 were targeted by several highly expressed miRNAs. Moreover, in the miRNA-function collaborative networks of upregulated miRNAs, miR-128 targeted myeloid dendritic associated functions.

In the PPI network of the DEGs, MAD2L1 and AURKA were with high degrees. The overexpression of Mad2 can induce early dyscrasia, lung metastasis and poor survival in OS (29). The knockdown of Mad2 leads to OS cell death through apoptosis associated with Rad21 cleavage; thus, Mad2 may serve as a target for cancer therapy (30). AURKA can promote cell cycle and suppress cell apoptosis, and the inhibition of AURKA by specific short hairpin RNA (shRNA) may be a promising therapeutic strategy of OS (31). Furthermore, in the TF-DEG regulatory network, the TF, STAT3, targeted the most DEGs. By binding to the promoter region of miR-125b and acting as a transactivator, STAT3 regulates miR-125b which serves as a potential target in the therapy of OS (32). The overexpression of phosphorylated-STAT3 in OS cells is implicated in poor prognosis and may function as a prognostic indicator and therapeutic target for OS (33,34). These data suggest that MAD2L1, AURKA and STAT3 may be closely associated with OS.

Some other molecules have also been involved in OS. The deregulation of miR-1 and miR-133b may correlate with cell cycle and cell proliferation of OS by mediating c-met (MET) protein expression (35). Through directly regulating PTEN/AKT signaling, miR-128 functions in the proliferation of human OS cells (36). The hypermethylation of p14ARF and ESR1 separately correlates with the absence of metastases at diagnoses and poor survival, therefore, p14ARF and ESR1 hypermethylation may be used as prognostic indicators for in OS (37). In 143B OS cells, phosphorylated and activated c-Jun and Fra-1 (also known as FOSL1) can induce MMP1 gene expression which may be a target for invasive and pulmonary metastases of OS, therefore, phosphorylated c-Jun and Fra-1 may affect invasion of OS through mediating MMP1 (38).

In conclusion, this study identified key genes or miRNAs involved in OS. We screened 1,609 DEGs and 149 DE-miRNAs in the OS cell lines compared with normal bones. Besides, some molecules may correlate with OS, such as MAD2L1, AURKA, STAT3, ESR1, FOSL1, miR-1 and miR-128. However, experimental researches are still necessary to validate the functions of these molecules in OS.

References

1 

Wittig JC, Bickels J, Priebat D, Jelinek J, Kellar-Graney K, Shmookler B and Malawer MM: Osteosarcoma: A multidisciplinary approach to diagnosis and treatment. Am Fam Physician. 65:1123–1132. 2002.PubMed/NCBI

2 

Ottaviani G and Jaffe N: The epidemiology of osteosarcoma. Pediatric and Adolescent Osteosarcoma. Springer; pp. 3–13. 2010

3 

Stiller CA, Bielack SS, Jundt G and Steliarova-Foucher E: Bone tumours in European children and adolescents, 1978–1997. Report from the Automated Childhood Cancer Information System project. Eur J Cancer. 42:2124–2135. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Stiller CA, Craft AW and Corazziari I; EUROCARE Working Group: Survival of children with bone sarcoma in Europe since 1978: Results from the EUROCARE study. Eur J Cancer. 37:760–766. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Maire G, Yoshimoto M, Chilton-MacNeill S, Thorner PS, Zielenska M and Squire JA: Recurrent RECQL4 imbalance and increased gene expression levels are associated with structural chromosomal instability in sporadic osteosarcoma. Neoplasia. 11:260–268. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Zhou H, Randall RL, Brothman AR, Maxwell T, Coffin CM and Goldsby RE: Her-2/neu expression in osteosarcoma increases risk of lung metastasis and can be associated with gene amplification. J Pediatr Hematol Oncol. 25:27–32. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Sadikovic B, Thorner P, Chilton-Macneill S, Martin JW, Cervigne NK, Squire J and Zielenska M: Expression analysis of genes associated with human osteosarcoma tumors shows correlation of RUNX2 overexpression with poor response to chemotherapy. BMC Cancer. 10:2022010. View Article : Google Scholar : PubMed/NCBI

8 

Gamberi G, Benassi MS, Bohling T, Ragazzini P, Molendini L, Sollazzo MR, Pompetti F, Merli M, Magagnoli G, Balladelli A, et al: C-myc and c-fos in human osteosarcoma: Prognostic value of mRNA and protein expression. Oncology. 55:556–563. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Lu XY, Lu Y, Zhao YJ, Jaeweon K, Kang J, Xiao-Nan L, Ge G, Meyer R, Perlaky L, Hicks J, et al: Cell cycle regulator gene CDC5L, a potential target for 6p12-21 amplicon in osteosarcoma. Mol Cancer Res. 6:937–946. 2008. View Article : Google Scholar : PubMed/NCBI

10 

de Nigris F, Botti C, de Chiara A, Rossiello R, Apice G, Fazioli F, Fiorito C, Sica V and Napoli C: Expression of transcription factor Yin Yang 1 in human osteosarcomas. Eur J Cancer. 42:2420–2424. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Cao Y, Zhou Z, de Crombrugghe B, Nakashima K, Guan H, Duan X, Jia SF and Kleinerman ES: Osterix, a transcription factor for osteoblast differentiation, mediates antitumor activity in murine osteosarcoma. Cancer Res. 65:1124–1128. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Osaki M, Takeshita F, Sugimoto Y, Kosaka N, Yamamoto Y, Yoshioka Y, Kobayashi E, Yamada T, Kawai A, Inoue T, et al: MicroRNA-143 regulates human osteosarcoma metastasis by regulating matrix metalloprotease-13 expression. Mol Ther. 19:1123–1130. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Zhang H, Cai X, Wang Y, Tang H, Tong D and Ji F: microRNA-143, downregulated in osteosarcoma, promotes apoptosis and suppresses tumorigenicity by targeting Bcl-2. Oncol Rep. 24:1363–1369. 2010.PubMed/NCBI

14 

Duan Z, Choy E, Harmon D, Liu X, Susa M, Mankin H and Hornicek F: MicroRNA-199a-3p is downregulated in human osteosarcoma and regulates cell proliferation and migration. Mol Cancer Ther. 10:1337–1345. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Ziyan W, Shuhua Y, Xiufang W and Xiaoyun L: MicroRNA-21 is involved in osteosarcoma cell invasion and migration. Med Oncol. 28:1469–1474. 2011. View Article : Google Scholar

16 

Yan K, Gao J, Yang T, Ma Q, Qiu X, Fan Q and Ma B: MicroRNA-34a inhibits the proliferation and metastasis of osteosarcoma cells both in vitro and in vivo. PLoS One. 7:e337782012. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Smyth GK: Limma: linear models for microarray data. Bioinformatics and computational biology solutions using R and Bioconductor. Springer; pp. 397–420. 2005, View Article : Google Scholar

19 

Zhao M, Sun J and Zhao Z: TSGene: A web resource for tumor suppressor genes. Nucleic Acids Res. 41:D970–D976. 2013. View Article : Google Scholar :

20 

Chen JS, Hung WS, Chan HH, Tsai SJ and Sun HS: In silico identification of oncogenic potential of fyn-related kinase in hepatocellular carcinoma. Bioinformatics. 29:420–427. 2013. View Article : Google Scholar

21 

Boyle EI, Weng S, Gollub J, Jin H, Botstein D, Cherry JM and Sherlock G: GO:TermFinder - open source software for accessing Gene Ontology information and finding significantly enriched Gene Ontology terms associated with a list of genes. Bioinformatics. 20:3710–3715. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Kanehisa M, Goto S, Furumichi M, Tanabe M and Hirakawa M: KEGG for representation and analysis of molecular networks involving diseases and drugs. Nucleic Acids Res. 38:D355–D360. 2010. View Article : Google Scholar :

23 

Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, Minguez P, Bork P, von Mering C, et al: STRING v9.1: Protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41:D808–D815. 2013. View Article : Google Scholar :

24 

Kohl M, Wiese S and Warscheid B: Cytoscape: software for visualization and analysis of biological networks. Data Mining in Proteomics. Springer; pp. 291–303. 2011

25 

Nepusz T, Yu H and Paccanaro A: Detecting overlapping protein complexes in protein-protein interaction networks. Nat Methods. 9:471–472. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Fujita PA, Rhead B, Zweig AS, Hinrichs AS, Karolchik D, Cline MS, Goldman M, Barber GP, Clawson H, Coelho A, et al: The UCSC genome browser database: Update 2011. Nucleic Acids Res. 39:D876–D882. 2011. View Article : Google Scholar :

27 

Xiao F, Zuo Z, Cai G, Kang S, Gao X and Li T: miRecords: An integrated resource for microRNA-target interactions. Nucleic Acids Res. 37:D105–D110. 2009. View Article : Google Scholar

28 

Dweep H, Sticht C, Pandey P and Gretz N: miRWalk - database: Prediction of possible miRNA binding sites by 'walking' the genes of three genomes. J Biomed Inform. 44:839–847. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Yu L, Liu S, Guo W, Zhang B, Liang Y and Feng Q: Upregulation of Mad2 facilitates in vivo and in vitro osteosarcoma progression. Oncol Rep. 28:2170–2176. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Yu L, Guo W, Zhao S, Tang J and Liu J: Knockdown of Mad2 induces osteosarcoma cell apoptosis-involved Rad21 cleavage. J Orthop Sci. 16:814–820. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Jiang Z, Jiang J, Yang H, Ge Z, Wang Q, Zhang L, Wu C and Wang J: Silencing of Aurora kinase A by RNA interference inhibits tumor growth in human osteosarcoma cells by inducing apoptosis and G2/M cell cycle arrest. Oncol Rep. 31:1249–1254. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Liu LH, Li H, Li JP, Zhong H, Zhang HC, Chen J and Xiao T: miR-125b suppresses the proliferation and migration of osteosarcoma cells through downregulation of STAT3. Biochem Biophys Res Commun. 416:31–38. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Ryu K, Choy E, Yang C, Susa M, Hornicek FJ, Mankin H and Duan Z: Activation of signal transducer and activator of transcription 3 (Stat3) pathway in osteosarcoma cells and overexpression of phosphorylated-Stat3 correlates with poor prognosis. J Orthop Res. 28:971–978. 2010.PubMed/NCBI

34 

Wang YC, Zheng LH, Ma BA, Zhou Y, Zhang MH, Zhang DZ and Fan QY: Clinical value of signal transducers and activators of transcription 3 (STAT3) gene expression in human osteosarcoma. Acta Histochem. 113:402–408. 2011. View Article : Google Scholar

35 

Novello C, Pazzaglia L, Cingolani C, Conti A, Quattrini I, Manara MC, Tognon M, Picci P and Benassi MS: miRNA expression profile in human osteosarcoma: Role of miR-1 and miR-133b in proliferation and cell cycle control. Int J Oncol. 42:667–675. 2013. View Article : Google Scholar

36 

Shen L, Chen XD and Zhang YH: MicroRNA-128 promotes proliferation in osteosarcoma cells by downregulating PTEN. Tumour Biol. 35:2069–2074. 2014. View Article : Google Scholar

37 

Sonaglio V, de Carvalho AC, Toledo SR, Salinas-Souza C, Carvalho AL, Petrilli AS, de Camargo B and Vettore AL: Aberrant DNA methylation of ESR1 and p14ARF genes could be useful as prognostic indicators in osteosarcoma. Onco Targets Ther. 6:713–723. 2013.PubMed/NCBI

38 

Kimura R, Ishikawa C, Rokkaku T, Janknecht R and Mori N: Phosphorylated c-Jun and Fra-1 induce matrix metalloproteinase-1 and thereby regulate invasion activity of 143B osteosarcoma cells. Biochim Biophys Acta. 1813:1543–1553. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 42 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen H, Wang W, Ni B, Zou Q, Lu H and Wang Z: Exploring the molecular mechanisms of osteosarcoma by the integrated analysis of mRNAs and miRNA microarrays. Int J Mol Med 42: 21-30, 2018
APA
Shen, H., Wang, W., Ni, B., Zou, Q., Lu, H., & Wang, Z. (2018). Exploring the molecular mechanisms of osteosarcoma by the integrated analysis of mRNAs and miRNA microarrays. International Journal of Molecular Medicine, 42, 21-30. https://doi.org/10.3892/ijmm.2018.3594
MLA
Shen, H., Wang, W., Ni, B., Zou, Q., Lu, H., Wang, Z."Exploring the molecular mechanisms of osteosarcoma by the integrated analysis of mRNAs and miRNA microarrays". International Journal of Molecular Medicine 42.1 (2018): 21-30.
Chicago
Shen, H., Wang, W., Ni, B., Zou, Q., Lu, H., Wang, Z."Exploring the molecular mechanisms of osteosarcoma by the integrated analysis of mRNAs and miRNA microarrays". International Journal of Molecular Medicine 42, no. 1 (2018): 21-30. https://doi.org/10.3892/ijmm.2018.3594