Integrated analysis of gene expression and copy number variations in MET proto‑oncogene‑transformed human primary osteoblasts

  • Authors:
    • Ru‑Jiang Jia
    • Chun‑Gen Lan
    • Xiu‑Chao Wang
    • Chun‑Tao Gao
  • View Affiliations

  • Published online on: November 22, 2017     https://doi.org/10.3892/mmr.2017.8135
  • Pages: 2543-2548
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to screen the potential osteosarcoma (OS)‑associated genes and to obtain additional insight into the pathogenesis of OS. Transcriptional profile (ID: GSE28256) and copy number variations (CNV) profile were downloaded from Gene Expression Omnibus database. Differentially expressed genes (DEGs) between MET proto‑oncogene‑transformed human primary osteoblast (MET‑HOB) samples and the control samples were identified using the Linear Models for Microarray Data package. Subsequently, CNV areas and CNVs were identified using cut‑off criterion of >30%‑overlap within the cases using detect_cnv.pl in PennCNV. Genes shared in DEGs and CNVs were obtained and discussed. Additionally, the Database for Annotation, Visualization and Integrated Discovery was used to identify significant Gene Ontology (GO) functions and pathways in DEGs with P<0.05. A total of 1,601 DEGs were screened out in MET‑HOBs and compared with control samples, including 784 upregulated genes, such as E2F transcription factor 1 (E2F1) and 2 (E2F2) and 817 downregulated genes, such as retinoblastoma 1 (RB1) and cyclin D1 (CCND1). DEGs were enriched in 344 GO terms, such as extracellular region part and extracellular matrix and 14 pathways, including pathways in cancer and extracellular matrix‑receptor interaction. Additionally, 239 duplications and 439 deletions in 678 genes from 1,313 chromosome regions were detected. A total of 12 genes were identified to be CNV‑driven genes, including cadherin 18, laminin subunit α 1, spectrin β, erythrocytic, ciliary rootlet coiled‑coil, rootletin pseudogene 2, β‑1,4-N-acetyl-galactosaminyltransferase 1, G protein regulated inducer of neurite outgrowth 1, EH domain binding protein 1‑like 1, growth factor independent 1, cathepsin Z, WNK lysine deficient protein kinase 1, glutathione S‑transferase mu 2 and microsomal glutathione S‑transferase 1. Therefore, cell cycle‑associated genes including E2F1, E2F2, RB1 and CCND1, and cell adhesion‑associated genes, such as CDH18 and LAMA1 may be used as diagnosis and/or therapeutic markers for patients with OS.

Introduction

Osteosarcoma (OS), also termed bone sarcoma, originates from bone and particularly from the mesenchymal stem cell lineage (1). OS, the most common bone tumor, is highly aggressive and usually has poor prognosis (2). Additionally, OS primarily affects adolescents and children and ~60% of neoplasms occur in patients under the age of 20 (3,4). Current treatment frequently involves a combination of surgery and chemotherapy; however, OS still leads to a high mortality and morbidity, particularly in children and adolescents (1).

Currently, considerable progress has been made in identifying the critical factors in the development and progression of OS, including genes, pathways and copy number variants (CNVs) (5). Alterations of tumor suppressor gene expression including protein kinase, cAMP-dependent, regulatory, and type I a and deregulation of major signaling pathways such as the wingless-type MMTV integration site family, transforming growth factor-β, Notch and sonic hedgehog have been previously associated with OS (6,7). It has also been previously demonstrated that OS development is dependent on loss of P53 and enhanced by loss of retinoblastoma 1 (RB1) (8). CNVs are DNA segments 1 kb in length which are present in a variable population frequency in the genome (9). During the 1990s, CNVs with duplications and deletions were expressed as an inducement of a quantity of single gene disorders (10). Various differentially expressed genes (DEGs) and several candidate CNVs in OS have been identified to be involved in the development of OS by analyzing the microarray data and high-resolution single nucleotide polymorphism (SNP)/CNV arrays (11,12). However, frequently only one of these approaches has been used in previous studies to identify the candidate molecule, and the molecular mechanism of OS remains to be elucidated (911).

The proto-oncogene MET protein, a hepatocyte growth factor receptor, encodes tyrosine-kinase activity (13), which has been revealed to be aberrantly expressed in OS and closely associated with cancer (1416). Therefore, overexpression of the MET oncogene may convert human primary osteoblasts (HOB) into OS cells.

The present study extracted the transcriptional and CNV profiles from Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) and CNVs were screened in the transcriptional profile of MET-HOB cells, which were previously turned into OS cells by lentiviral vector (LV)-driven overexpression of the MET oncogene. Subsequently, the shared genes in the expression and CNV profiles were analyzed. The present study obtained a series of candidate markers in OS and may provide the foundation for treatment of OS.

Materials and methods

Microarray and CNV data

Transcriptional profile (ID: GSE28256) was downloaded from the GEO database (www.ncbi.nlm.nih.gov/geo/) which was based on the platform of GPL6098 (Illumina humanRef-8 v1.0 expression beadchip) (17). The dataset contained 15 samples, including 6 HOB cell lines and 9 MET-HOBs clones, which were previously turned into osteosarcoma cells by over-expression of MET oncogene driven by a LV. The CNV data were extracted from the GSE32964 dataset in the GEO database, which included 36 samples for detecting SNP and 32 samples for CNV. A total of 32 CNV samples of OS tumor tissues based on the platform of GPL6985 (Illumina HumanCNV370-QuadV3 DNA Analysis BeadChip) were analyzed in the present study.

Data preprocessing

The probe-level data of the transcriptional profile were initially converted into expression values. Probes that mapped with the gene names labeled in the annotation platform were transformed using log2 and normalized using preprocess-Core package in R version 2.9.0. According to the annotation platform, the values of probes corresponding to the same transcript were averaged and then defined as the final expression value of a transcript. The PennCNV tool (version 2014 May 07; http://penncnv.openbioinformatics.org) was used in the subsequent processing of data, the profile of CNV samples was converted into specific format for PennCNV, which contained log R Ratio: LRR and B Allele frequency: BAF. In addition, to investigate the differences among samples, a heatmap was generated to compare their expression values using the Gplots package in R.

Identification of DEGs and CNV

A Student's t-test was conducted on the gene expression values between testing and control samples. The Linear Models for Microarray Data (LIMMA) package was used to normalize the data and identify the DEGs in MET-HOB samples compared with control samples using cut-offs of P<0.05 and |log2 fold-change (FC)|>2. Additionally, detect_cnv.pl in PennCNV was applied to select CNV areas and CNVs were identified with the cut-off criteria of >30% overlap within the cases. Genes shared in CNVs and DEGs were identified as critical genes associated with the development of OS.

Functional enrichment of DEGs

The Database for Annotation, Visualization and Integrated Discovery (DAVID) provides numerous comprehensive functional annotation which contributes to the understanding of the biological meanings behind abundant genes (18). Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses have become commonly used approaches for functional and pathway studies of large-scale genomic or transcription data, respectively (19). Therefore, they were used in the present study. Next, DAVID was used to screen the enriched GO terms and KEGG pathways in the DEGs. P<0.05 was used as a cut-off criterion.

Results

Data preprocessing and DEGs screening

In the present study, 24,350 probes were detected in the original data and 21,454 non-redundant genes were obtained following data preprocessing. The raw data in all samples have been normalized (Fig. 1). A total of 1601 DEGs were screened out in MET-HOBs compared with controls. Among these genes, 784 were upregulated in MET-HOBs and 817 were downregulated. The hierarchical clustering analysis revealed a clearly distinct expression of all DEGs between MET-HOBs and HOBs (Fig. 2).

Function enrichment of DEGs

In order to identify the functions of the DEGs, they were performed GO (P<0.01) and KEGG (P<0.01) enrichment analyses. The results indicated that 344 GO terms were obtained and the top 10% terms were listed in Table I, such as extracellular region (P=2.68E-24), extracellular matrix (ECM; P=4.08E-24) and proteinaceous extracellular matrix (P=4.79E-24). Besides, 14 KEGG pathways were obtained and most of them were related to cancers, such as hsa05200: pathways in cancer (P=4.12E-09), hsa04512: ECM-receptor interaction (P=2.41E-08) and hsa05222: small cell lung cancer (P=1.06E-05; Table II).

Table I.

The top 10% enriched GO terms for DEGs.

Table I.

The top 10% enriched GO terms for DEGs.

CategoryGO IDGO nameGene numberP-value
CCGO:0044421Extracellular region part134 2.68×10−24
CCGO:0031012Extracellular matrix  74 4.08×10−24
CCGO:0005578Proteinaceous extracellular matrix  71 4.79×10−24
BPGO:0001501Skeletal system development  60 4.57×10−15
MFGO:0019838Growth factor binding  32 2.32×10−14
BPGO:0001944Vasculature development  50 1.22×10−13
BPGO:0001568Blood vessel development  49 1.88×10−13
MFGO:0005201Extracellular matrix structural constituent  28 2.11×10−13
BPGO:0042127Regulation of cell proliferation  98 4.21×10−12
BPGO:0006260DNA replication  40 8.03×10−12
BPGO:0007049Cell cycle  96 1.09×10−11
BPGO:0051726Regulation of cell cycle  55 1.23×10−11
BPGO:0022403Cell cycle phase  63 1.57×10−11
CCGO:0005576Extracellular region179 1.87×10−11
CCGO:0044427Chromosomal part  57 2.60×10−11
BPGO:0051270Regulation of cell motion  39 5.52×10−11
BPGO:0006259DNA metabolic process  70 8.61×10−11
CCGO:0044420Extracellular matrix part  28 1.55×10−10
BPGO:0040012Regulation of locomotion  38 1.89×10−10
BPGO:0051301Cell division  49 1.93×10−10
BPGO:0030334Regulation of cell migration  35 3.13×10−10
BPGO:0007155Rell adhesion  85 5.11×10−10
BPGO:0022610Biological adhesion  85 5.66×10−10
CCGO:0005615Extracellular space  79 6.44×10−10
BPGO:0006928Cell motion  65 6.97×10−10
BPGO:0022402Cell cycle process  73 7.17×10−10
CCGO:0005694Chromosome  60 1.09×10−9
BPGO:0000278Mitotic cell cycle  54 2.53×10−9
BPGO:0000279M phase  49 7.98×10−9
BPGO:0048514Blood vessel morphogenesis  37 1.03×10−8
BPGO:0065004Protein-DNA complex assembly  23 1.29×10−8
CCGO:0005581Collagen  14 1.97×10−8
BPGO:0030198Extracellular matrix organization  24 3.69×10−8
BPGO:0008283Cell proliferation  57 4.64×10−8

[i] GO, Gene Ontology; DEGs, differentially expressed genes; CC, cellular component; MF, molecular function; BP, biological process.

Table II.

Enriched Kyoto Encyclopedia of Genes and Genomes pathways for DEGs.

Table II.

Enriched Kyoto Encyclopedia of Genes and Genomes pathways for DEGs.

KEGG IDPathway nameGene numberP-value
hsa05200Pathways in cancer53 4.12×10−9
hsa04512ECM-receptor interaction23 2.41×10−8
hsa05222Small cell lung cancer19 1.06×10−5
hsa03030DNA replication12 1.72×10−5
hsa04510Focal adhesion30 8.78×10−5
hsa04115p53 signaling pathway15 1.62×10−4
hsa05210Colorectal cancer16 4.93×10−4
hsa05218Melanoma14 9.09×10−4
hsa05219Bladder cancer10 1.79×10−3
hsa00980Metabolism of xenobiotics by cytochrome P45012 2.20×10−3
hsa05215Prostate cancer15 2.68×10−3
hsa05217Basal cell carcinoma11 3.69×10−3
hsa05214Glioma11 9.89×10−3

[i] DEGs, differentially expressed genes.

Identification of CNVs

A total of 1,313 chromosome regions were identified and 678 genes (239 duplications and 439 deletions) were obtained, which were spread among 22 pairs of autosomes (Fig. 3). Then these CNVs were checked for overlap with the DEGs. Finally, 12 genes were identified in both in CNVs and DEGs, including the six upregulated genes cadherin 18 (CDH18), spectrin β, erythrocytic (SPTB), ciliary rootlet coiled-coil, rootletin pseudogene 2 (CROCCP2), β-1,4-N-acetyl-galactosaminyltransferase 1 (B4GALNT), G protein regulated inducer of neurite outgrowth 1 (GPRIN1) and growth factor independent 1 (GFI1). A total of six downregulated genes were identified, including laminin subunit α 1 (LAMA1), EH domain binding protein 1-like 1 (EHBP1L1), cathepsin Z (CTSZ), WNK lysine deficient protein kinase 1 (PRKWNK1), glutathione S-transferase µ 2 (GSTM2) and microsomal glutathione S-transferase 1 (MGST1) (Table III).

Table III.

CNV-driven genes.

Table III.

CNV-driven genes.

GenelogFC
LAMA1−2.583
EHBP1L1−5.75702
SPTB5.224308
CTSZ−7.39063
CROCCP22.491733
B4GALNT3.6299
CDH182.227242
GPRIN12.361558
PRKWNK1−2.75581
GFI13.577933
GSTM2−2.62289
MGST1−2.30508

[i] CNV, copy number variations; FC, fold-change.

Discussion

OS is a universally fatal disease, due to the rapid growth, high local aggressiveness, and metastasizing potential (20). Numerous DEGs and regulatory relationships between transcription factors and DEGs in OS have been identified using microarray data (12). Additionally, susceptibility genes associated with OS were also reported by analyzing SNP/CNV arrays (11). However, the underlying molecular mechanism of OS remains to be elucidated. In the present study, 1,601 DEGs were identified, including 784 upregulated and 817 downregulated DEGs and CNVs in 678 genes (239 duplications and 439 deletions) were observed in MET-HOBs samples when compared with controls. By analyzing the transcriptional profile and SNP/CNV arrays, CDH18, LAMA1, SPTB, CROCCP2, B4GALNT, GPRIN1, GFI1, EHBP1L1, CTSZ, PRKWNK1, GSTM2 and MGST1 were identified as CNV-driven DEGs.

The DEGs obtained in the current study suggested that several genes such as E2F transcription factor 1 (E2F1) and 2 (E2F2), retinoblastoma 1 (RB1) and cyclin D1 (CCND1) were involved in various pathways. E2F1 and E2F2, members of the E2F family of transcription factors, were upregulated in the MET-HOBs samples. E2F proteins regulate the transcription of genes required for DNA synthesis (21). The E2F family has an important role in cell cycle regulation and action of tumor suppressor proteins, and is also a target of the transforming proteins of small DNA tumor viruses (22,23). Additionally, the RB protein has been previously identified to bind to E2F transcription factors (24). It is evident that the RB/E2F pathway is very important in regulating the initiation of DNA replication and the pathway is disrupted in the majority of human cancers (25). CCND1, is part of the highly conserved cyclin family, is a nuclear protein required for cell cycle progression in G1 phase (26). It has been previously reported that CCND1 has an important role in the regulation of OS cell proliferation (26). Consistently, the findings of the present study revealed that those genes were involved in several cell cycle-associated GO terms, such as cell cycle, cell cycle phase, regulation of cell cycle and cell division, and cancer-associated KEGG pathways, including pathways in cancer, small cell lung cancer and melanoma. Therefore, the present study is reliable and may suggest that the screened DEGs such as E2F1, E2F2, RB1 and CCND1 are closely associated with the cell cycle and cell division of OS.

CNVs such as deletions, duplications and amplifications across the whole genome may contribute to OS tumorigenesis (27). It is of note that CNVs in cyclin-dependent kinase inhibitor 2A (CDKN2A), sex determining region Y-box 6 (SOX6) and phosphatase and tensin homolog (PTEN) were associated with Ewing sarcoma (28). The trail of CDKN2A/B locus was detected in OS cell lines (29), whereas two SNPs in the SOX6 gene were identified to be associated with both hip bone mineral density and body mass index in Caucasians (30). In addition, copy number losses in PTEN were common events in OS (31). In the present study, further analysis identified 12 CNV-driven genes in MET-HOBs samples, such as CDH18 (upregulated) and LAMA1 (downregulated), which were associated with cell adhesion. Since the 1990s, many cadherins and cadherin-associated proteins had been identified and implicated in cancers as candidate tumor suppressors or proto-oncogenes (32). Deregulation of cadherin-catenin complexes may contribute to tumor development by influencing the adhesion of epithelial cells (33). CDH18 is a member of the cadherin superfamily that mediates calcium-dependent cell-cell adhesion (34). Although, no previous studies have not identified a direct association between CDH18 and OS, it has been revealed that an Exon 2 deletion of CDH18 may be associated with human colorectal cancer and CDH18 may act as novel candidate gene involved in colorectal cancer predisposition (35). In the current study, CDH18 was upregulated and CNVs were detected in CDH18 of MET-HOBs samples; therefore, CDH18 may have a role in cell-cell adhesion of OS. Additionally, LAMA1, also termed EHS laminin, was downregulated in MET-HOBs samples when compared with controls. Laminin is a complex glycoprotein and is considered to control the attachment, migration and organization of cells during embryonic development by interacting with other ECM components (36,37). Additionally, it has been previously reported that metadherin, as a laminin receptor, has an important role in controlling tumorigenesis and metastasis in many human cancers (3840). Metadherin, a type II membrane protein in OS cells, may enhance cell invasion by regulating cell adhesion to the ECM through interaction with laminin (41). Therefore, LAMA1 may be involved in cell adhesion and cell-cell interactions in OS.

It is of note that there were some limitations in the present study. Only the OS-associated CNVs and DEGs were identified, whereas the transcription factors and protein-protein interaction network remain to be determined. The current findings were obtained by bioinformatics analysis and the corresponding validations were not performed. Therefore, future studies should involve in performing experiments such as reverse transcription-quantitative polymerase reaction and western blotting to validate the CNVs and DEGs identified.

In conclusion, a series of DEGs were identified to be associated with cell cycle and cell division of human OS, specifically E2F1, E2F2, RB1 and CCND1. Additionally, 12 CNV-driven DEGs were obtained and the cell adhesion-associated genes such as CDH18 and LAMA1 may contribute to OS cell-cell adhesion. These genes may act as alternative diagnosis and/or therapeutic markers for patients with OS. The present study developed the current understanding about the etiology of OS and provided the foundation for the development of novel treatment strategies for OS. However, further experiments are required to confirm these findings.

References

1 

Heymann D and Rédini F: Targeted therapies for bone sarcomas. Bonekey Rep. 2:3782013. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Li C, Cheng Q, Liu J, Wang B, Chen D and Liu Y: Potent growth-inhibitory effect of TRAIL therapy mediated by double-regulated oncolytic adenovirus on osteosarcoma. Mol Cell Biochem. 364:337–344. 2012. View Article : Google Scholar : PubMed/NCBI

4 

He JP, Hao Y, Wang XL, Yang XJ, Shao JF, Guo FJ and Feng JX: Review of the molecular pathogenesis of osteosarcoma. Asian Pac J Cancer Prev. 15:5967–5976. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Xiong Y, Wu S, Du Q, Wang A and Wang Z: Integrated analysis of gene expression and genomic aberration data in osteosarcoma (OS). Cancer Gene Ther. 22:524–529. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Molyneux SD, Di Grappa MA, Beristain AG, McKee TD, Wai DH, Paderova J, Kashyap M, Hu P, Maiuri T, Narala SR, et al: Prkar1a is an osteosarcoma tumor suppressor that defines a molecular subclass in mice. J Clin Invest. 120:3310–3325. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Tang N, Song WX, Luo J, Haydon RC and He TC: Osteosarcoma development and stem cell differentiation. Clin Orthop Relat Res. 466:2114–2130. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Walkley CR, Qudsi R, Sankaran VG, Perry JA, Gostissa M, Roth SI, Rodda SJ, Snay E, Dunning P, Fahey FH, et al: Conditional mouse osteosarcoma, dependent on p53 loss and potentiated by loss of Rb, mimics the human disease. Genes Dev. 22:1662–1676. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Kirov G, Rees E, Walters JT, Escott-Price V, Georgieva L, Richards AL, Chambert KD, Davies G, Legge SE, Moran JL, et al: The penetrance of copy number variations for schizophrenia and developmental delay. Biol Psychiatry. 75:378–385. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Riccardi VM and Lupski JR: Duplications, deletions, and single-nucleotide variations: The complexity of genetic arithmetic. Genet Med. 15:172–173. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Porat RM, Pasic I, Shlien A, Golgoz N, Andrulis I, Wunder JS and Malkin D: Genome-wide copy number analysis reveals two novel loci for susceptibility to sporadic osteosarcoma. Cancer Res. 71 8 Suppl:S53342011. View Article : Google Scholar

12 

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

13 

MacEwen EG, Kutzke J, Carew J, Pastor J, Schmidt JA, Tsan R, Thamm DH and Radinsky R: c-Met tyrosine kinase receptor expression and function in human and canine osteosarcoma cells. Clin Exp Metastasis. 20:421–430. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Ferracini R, Angelini P, Cagliero E, Linari A, Martano M, Wunder J and Buracco P: MET oncogene aberrant expression in canine osteosarcoma. J Orthop Res. 18:253–256. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Coltella N, Manara MC, Cerisano V, Trusolino L, Di Renzo MF, Scotlandi K and Ferracini R: Role of the MET/HGF receptor in proliferation and invasive behavior of osteosarcoma. FASEB J. 17:1162–1164. 2003.PubMed/NCBI

16 

Naka T, Iwamoto Y, Shinohara N, Ushijima M, Chuman H and Tsuneyoshi M: Expression of c-met proto-oncogene product (c-MET) in benign and malignant bone tumors. Mod Pathol. 10:832–838. 1997.PubMed/NCBI

17 

Dani N, Olivero M, Mareschi K, van Duist MM, Miretti S, Cuvertino S, Patanè S, Calogero R, Ferracini R, Scotlandi K, et al: The MET oncogene transforms human primary bone-derived cells into osteosarcomas by targeting committed osteo-progenitors. J Bone Miner Res. 27:1322–1334. 2012. View Article : Google Scholar : PubMed/NCBI

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 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Lim JH, Chang YC, Park YB, Park JW and Kwon TK: Transcriptional repression of E2F gene by proteasome inhibitors in human osteosarcoma cells. Biochem Biophys Res Commun. 318:868–872. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Tsantoulis PK and Gorgoulis VG: Involvement of E2F transcription factor family in cancer. Eur J Cancer. 41:2403–2414. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Helt AM and Galloway DA: Mechanisms by which DNA tumor virus oncoproteins target the Rb family of pocket proteins. Carcinogenesis. 24:159–169. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Lees JA, Saito M, Vidal M, Valentine M, Look T, Harlow E, Dyson N and Helin K: The retinoblastoma protein binds to a family of E2F transcription factors. Mol Cell Biol. 13:7813–7825. 1993. View Article : Google Scholar : PubMed/NCBI

25 

Nevins JR: The Rb/E2F pathway and cancer. Hum Mol Genet. 10:699–703. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Baldin V, Lukas J, Marcote MJ, Pagano M and Draetta G: Cyclin D1 is a nuclear protein required for cell cycle progression in G1. Genes Dev. 7:812–821. 1993. View Article : Google Scholar : PubMed/NCBI

27 

Gokgoz N, Wunder JS and Andrulis IL: Genome-wide analysis of DNA copy number variations in osteosarcoma. Cancer Res. 72 8 Suppl:S50752012. View Article : Google Scholar

28 

Lynn M, Wang Y, Slater J, Shah N, Conroy J, Ennis S, Morris T, Betts DR, Fletcher JA and O'Sullivan MJ: High-resolution genome-wide copy-number analyses identify localized copy-number alterations in Ewing sarcoma. Diagn Mol Pathol. 22:76–84. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Ottaviano L, Schaefer KL, Gajewski M, Huckenbeck W, Baldus S, Rogel U, Mackintosh C, de Alava E, Myklebost O, Kresse SH, et al: Molecular characterization of commonly used cell lines for bone tumor research: A trans-European EuroBoNet effort. Genes Chromosomes Cancer. 49:40–51. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Liu YZ, Pei YF, Liu JF, Yang F, Guo Y, Zhang L, Liu XG, Yan H, Wang L, Zhang YP, et al: Powerful bivariate genome-wide association analyses suggest the SOX6 gene influencing both obesity and osteoporosis phenotypes in males. PLoS One. 4:e68272009. View Article : Google Scholar : PubMed/NCBI

31 

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

32 

Berx G and Van Roy F: Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol. 1:a0031292009. View Article : Google Scholar : PubMed/NCBI

33 

Goss KH and Groden J: Biology of the adenomatous polyposis coli tumor suppressor. J Clin Oncol. 18:1967–1979. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Yagi T and Takeichi M: Cadherin superfamily genes: Functions, genomic organization, and neurologic diversity. Genes Dev. 14:1169–1180. 2000.PubMed/NCBI

35 

Venkatachalam R, Verwiel ET, Kamping EJ, Hoenselaar E, Görgens H, Schackert HK, Van Krieken JH, Ligtenberg MJ, Hoogerbrugge N, van Kessel AG and Kuiper RP: Identification of candidate predisposing copy number variants in familial and early-onset colorectal cancer patients. Int J Cancer. 129:1635–1642. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Dziadek M: Role of laminin-nidogen complexes in basement membrane formation during embryonic development. Experientia. 51:901–913. 1995. View Article : Google Scholar : PubMed/NCBI

37 

Kleinman HK, Cannon FB, Laurie GW, Hassell JR, Aumailley M, Terranova VP, Martin GR and DuBois-Dalcq M: Biological activities of laminin. J Cell Biochem. 27:317–325. 1985. View Article : Google Scholar : PubMed/NCBI

38 

Li X, Kong X, Huo Q, Guo H, Yan S, Yuan C, Moran MS, Shao C and Yang Q: Metadherin enhances the invasiveness of breast cancer cells by inducing epithelial to mesenchymal transition. Cancer Sci. 102:1151–1157. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Zhu K, Dai Z, Pan Q, Wang Z, Yang GH, Yu L, Ding ZB, Shi GM, Ke AW, Yang XR, et al: Metadherin promotes hepatocellular carcinoma metastasis through induction of epithelial-mesenchymal transition. Clin Cancer Res. 17:7294–7302. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Wei Y, Hu G and Kang Y: Metadherin as a link between metastasis and chemoresistance. Cell Cycle. 8:2132–2137. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Zhu L, Zhang P, Yang Y, Buford AS, Wang WL, Thomas DG and Hughes DP: Abstract A53: Metadherin functions as a laminin receptor that is essential for metastasis and is associated with poor survival in osteosarcoma. Cancer Res. 74 20 Suppl:A532014. View Article : Google Scholar

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

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
Jia RJ, Lan CG, Wang XC and Gao CT: Integrated analysis of gene expression and copy number variations in MET proto‑oncogene‑transformed human primary osteoblasts. Mol Med Rep 17: 2543-2548, 2018
APA
Jia, R., Lan, C., Wang, X., & Gao, C. (2018). Integrated analysis of gene expression and copy number variations in MET proto‑oncogene‑transformed human primary osteoblasts. Molecular Medicine Reports, 17, 2543-2548. https://doi.org/10.3892/mmr.2017.8135
MLA
Jia, R., Lan, C., Wang, X., Gao, C."Integrated analysis of gene expression and copy number variations in MET proto‑oncogene‑transformed human primary osteoblasts". Molecular Medicine Reports 17.2 (2018): 2543-2548.
Chicago
Jia, R., Lan, C., Wang, X., Gao, C."Integrated analysis of gene expression and copy number variations in MET proto‑oncogene‑transformed human primary osteoblasts". Molecular Medicine Reports 17, no. 2 (2018): 2543-2548. https://doi.org/10.3892/mmr.2017.8135