Identification of genes associated with osteoarthritis by microarray analysis

  • Authors:
    • Jianwei Sun
    • Bingshan Yan
    • Wangping Yin
    • Xinchao Zhang
  • View Affiliations

  • Published online on: July 7, 2015     https://doi.org/10.3892/mmr.2015.4048
  • Pages: 5211-5216
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 investigate the mechanisms of osteoarthritis (OA). Raw microarray data (GSE51588) were downloaded from Gene Expression Omnibus, including samples from OA (n=20) and non‑OA (n=5) knee lateral and medial tibial plateaus. Differentially expressed genes (DEGs) were identified using Student's t‑test. Functional and pathway enrichment analyses were performed for the upregulated and downregulated DEGs. A protein‑protein interaction network (PPI) was constructed according to the Search Tool for the Retrieval of Interacting Genes/Proteins database, and module analysis of the PPI network was performed using CFinder. The protein domain enrichment analysis for genes in modules was performed using the INTERPRO database. A total of 869 upregulated and 508 downregulated DEGs were identified. The enriched pathways of downregulated and upregulated DEGs were predominantly associated with the cell cycle (BUB1, BUB1B, CCNA2, CCNB1 and CCNE1), and extracellular matrix (ECM)‑receptor interaction (CD36, COL11A2, COL1A1, COL2A1 and COL3A1). Functional enrichment analysis of the DEGs demonstrated that FGF19, KIF11 and KIF2C were involved in the response to stress and that ACAN, ADAMTS10 and BGN were associated with proteinaceous ECM. The top protein domain was IPR001752: Kinesin motor region involving three genes (KIF2C, KIF11 and KIF20A). The identified DEGs, including KIF2C, KIF11 and KIF20A, may be significant in the pathogenesis of OA.

Introduction

Osteoarthritis (OA) is the most prevalent chronic joint disease and results in a large economic burden due to the associated costs of medical care and lost earnings (1). OA is characterized by the disappearance of the cartilage, combined with sub-chondral bone sclerosis, formation of osteophytes and a mild inflammation of the synovial membrane (2).

The pathology of OA is complex and a large number of studies have focused on the pathogenesis of OA to identify a therapeutic method for patients with OA. The sub-chondral bone is involved in the pathophysiology of OA through biochemical and mechanical pathways (3). OA microarray analysis has predominantly focused on the articular cartilage, meniscus or synovium; however, only few studies on sub-chondral bone are available (4,5). Therefore, the present study assessed the sub-chondral bone in order to enhance the understanding of the pathology of OA. Zhen et al (6) reported that the pathological changes of OA may be caused by high concentrations of active transforming growth factor-β1 in the subchondral bone, and that inhibition of this process may represent a potential therapeutic method. According to a study by Valverde-Franco et al (7), bone-specific overexpression of EphB4 on subchondral bone and cartilage has a protective effect on OA. With the number of studies increasing, further genes associated with OA are being identified.

Microarray analysis has been used to analyze the gene expression of thousands of transcripts in OA samples. Several novel candidate genes, including bone formation-associated genes (CLEC3B, CDH11, GPNMB, CLEC3A, CHST11, MSX1, MSX2) and genes encoding collagens (COL13A1, COL14A1, COL15A1, COL8A2), were identified by microarray analysis (8). Chou et al (4) only performed functional and pathway enrichment analyses of the identified differentially expressed genes (DEGs). In the present study, the microarray data (GSE51588) (4) were downloaded from Gene Expression Omnibus using additional bioinformatics analysis to gain further insight into the molecular mechanisms of OA. Gene Ontology (GO) functional terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed for the DEGs. In addition, a protein-protein interaction (PPI) network was constructed and protein domain enrichment of the genes in the modules of the PPI network were performed to screen the significant genes, which were involved in the pathogenesis of OA.

Materials and methods

Microarray data and data pre-processing

The microarray data (GSE51588) deposited by Chou et al (4) were downloaded from the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/). The platform used was the GPL13497 Agilent-026652 Whole Human Genome Microarray 4×44 K v2 (Agilent Technologies, Palo Alto, CA, USA). A total of 50 samples were available, including 20 OA knee lateral tibial plateau samples, 20 OA knee medial plateau samples, 5 non-OA knee lateral tibial plateau samples and 5 non-OA knee medial plateau samples.

For data pre-processing, the expression profile chip was pre-processed using the Affy package in Bioconductor (http://www.bioconductor.org/packages/release/bioc/html/affy.html) (9) and Affymetrix annotation files from Brain Array Lab (Affymetrix, Santa Clara, CA, USA; http://www.affymetrix.com/analysis/). The background correction, quartile data normalization and probe summarization were performed using the robust multiarray average algorithm (http://www.bioconductor.org) (10) to obtain a gene expression matrix.

Identification of DEGs

The expression values for the normalized data were calculated using the Limma package in R (Affymetrix; http://www.affymetrix.com/analysis/) (11). DEGs were identified by Student's t-test. The raw P-value was adjusted into false discovery rate (FDR) using the Benjamini & Hochberg method (12). An FDR <0.01 and |log2fold change|>1 were selected as cut-off criteria.

Enrichment analysis for DEGs

Functional enrichment of the DEGs in the biological process, molecular function and cellular component categories was performed using the GO database (http://geneontology.org/) (13). Based on the KEGG database (http://www.genome.jp/kegg/pathway.html) (14), pathway enrichment of the DEGs was performed. P<0.01 was selected as a cut-off criterion.

Construction of the PPI network and module analysis

The identified DEGs were mapped to the Search Tool for the Retrieval of Interacting Genes version 9.1 database (http://www.string-db.org/) (15) to search for the interaction associations between the proteins, and a confidence score >0.4 was selected as cut-off criterion. Cytoscape software (http://www.cytoscape.org/) (16) was subsequently used to visualize the PPI network.

Module analysis of the PPI network was performed by CFinder (http://www.cfnder.org/) (17). The search algorithm using the Clique Percolation Method was used to identify the k-clique percolation clusters of the network (18). Larger k-clique values represented higher stringency during the identification of dense groups and provided smaller groups with a higher density of links inside them. A k-cliques value of 5 was selected as the cut-off criterion. The protein domain enrichment analysis of genes in the module was analyzed using the INTERPRO database (http://www.ebi.ac.uk/interpro/) (17,19) and P<0.05 was selected as the cut-off criterion.

Results

DEG analysis

For microarray analysis, 1,377 DEGs were identified between the OA group and the non-OA group. Of these genes, 869 DEGs were upregulated and 508 DEGs were downregulated.

Functional and pathway enrichment analysis of the DEGs

The functional enrichment analysis of the downregulated DEGs revealed that the top five enriched pathways were associated with systemic lupus erythematosus (P=1.17×10−13), the cell cycle (P=1.47×10−6), complement and coagulation cascades (P=0.000685), nitrogen metabolism (P=0.001086) and amoebiasis (P=0.001278) (Table I). BUB1, BUB1B, CCNA2, CCNB1 and CCNE1 were the genes involved in the cell cycle. The top five enriched GO terms included response to stress (P = 4.91×10−14), involving CCNA2, CCNB1, FGF19, KIF11 and KIF2C, immune system-associated processes (P=1.87×10−12) involving FGF19, KIF11 and KIF2C, response to wounding (P=2.04×10−12), including CCNB1, KIF11 and KIF2C, defense response (P=2.37×10−12), involving FGF19, and mitotic cell cycle (P=2.60×10−12) associated with BUB1, BUB1B, CCNA2, CCNB1, CCNE1, KIF20, KIF11 and KIF2C (Table II).

Table I

Top five enriched KEGG pathways for downregulated differentially expressed genes.

Table I

Top five enriched KEGG pathways for downregulated differentially expressed genes.

KEGG pathwayGene countP-valueGenes
Systemic lupus erythematosus27 1.17×10−13C9, CTSG, ELANE, H2AFX, HIST1H2AC
Cell cycle17 1.47×10−6BUB1, BUB1B, CCNA2, CCNB1, CCNE1
Complement and coagulation cascades90.000685C5AR1, C9, CR1, F12, F3
Nitrogen metabolism50.001086CA1, CA2, CA8, GLUL, HAL
Amoebiasis110.001278ARG1, ARG2, C9, COL4A6, CTSG

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes.

Table II

Top five enriched GO terms for the downregulated differentially expressed genes.

Table II

Top five enriched GO terms for the downregulated differentially expressed genes.

GO IDTermGene countP-valueGenes
0006950Response to stress155 4.91×10−14KIF11, KIF2C, FGF19, ADORA3, AEN
0002376Immune system process111 1.87×10−12KIF11, KIF2C, FGF19, AHCY, APOBEC3B
0009611Response to wounding75 2.04×10−12KIF11, KIF2C, ADM, ADORA3, AHCY
0006952Defense response85 2.37×10−12FGF19, AHCY, ALOX5, ALOX5AP, APOBEC3B
0000278Mitotic cell cycle61 2.60×10−12KIF11, KIF2C, KIF20A, AURKA, AURKB

[i] GO, gene ontology.

The functional enrichment analysis of the upregulated DEGs demonstrated that the top five enriched pathways were associated with extracellular matrix (ECM)-receptor interaction (P=3.01×10−5), axon guidance (P=0.000472), amoebiasis (P=0.003458), focal adhesion (P=0.003709) and cancer-associated pathways (P=0.00407) (Table III). Certain genes, including CD36, COL11A2, COL1A1, COL2A1 and COL3A1, were involved in the ECM-receptor interaction pathway. In addition, the GO functional enrichment analysis demonstrated that ACAN, ADAMTS10 and BGN were involved in proteinaceous ECM (P=1.11×10−15) and ECM (P=2.22×10−15) terms, A2M, ABCC6 and ACAN were involved in single-multicellular organism process (P=4.66×10−15), and multicellular organism process (P=8.27×10−14) terms, while ADAMTS10, COL10A1 and COL11A2 were involved in the ECM part (P=5.17×10−14) (Table IV).

Table III

Top five enriched KEGG pathways for the upregulated differentially expressed genes.

Table III

Top five enriched KEGG pathways for the upregulated differentially expressed genes.

KEGG pathwayGene countP-valueGenes
ECM-receptor interaction12 3.01×10−5CD36, COL11A2, COL1A1, COL2A1, COL3A1
Axon guidance130.000472EPHB3, LRRC4C, NTN1, NTN4, PLXNA3
Amoebiasis100.003458COL11A2, COL1A1, COL2A1, COL3A1, COL4A5
Focal adhesion150.003709AKT2, COL11A2, COL1A1, COL2A1, COL3A1
Pathways in cancer210.00407AKT2, ARNT2, COL4A5, CSF1R, DAPK2

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; ECM, extracellular matrix.

Table IV

Top five enriched GO terms for the upregulated differentially expressed genes.

Table IV

Top five enriched GO terms for the upregulated differentially expressed genes.

GO IDTermGene countP-valueGenes
0005578Proteinaceous extracellular matrix51 1.11×10−15ACAN, ADAMTS10, BGN, C1QTNF8, C1QT-
NF9
0031012Extracellular matrix55 2.22×10−15ACAN, ADAMTS10, BGN, C1QTNF8, C1QTNF9
0044707 Single-multicellular organism process305 4.66×10−15A2M, ABCC6, ACAN, ADAM20, ADAP2
0044420Extracellular matrix part33 5.17×10−14ADAMTS10, C1QTNF8, C1QTNF9, COL10A1/2
0032501Multicellular organismal process309 8.27×10−14A2M, ABCC6, ACAN, ADAM20, ADAP2

[i] GO, gene ontology.

Analysis of PPI network and modules

Two modules, including module 2 (Fig. 1) and module 5 (Fig. 2) were extracted from the constructed PPI network. Protein domain enrichment analysis revealed that the genes in module 5 were not significantly enriched in any protein domain. A total of 10 protein domains were enriched for the genes in module 2 (Table V), including IPR001752: Kinesin, motor region (P=0.001518) involving three genes (KIF2C, KIF11 and KIF20A), IPR013212: Mad3/BUB1 homology region 1 (P=0.002759) associated with two genes (BUB1 and BUB1B) and IPR014400: Cyclin A/B/D/E (P=0.016448) involving two genes (CCNB1 and CCNA2).

Table V

Enriched protein domains for genes in module 2 of the protein-protein interaction network (category, Interpro).

Table V

Enriched protein domains for genes in module 2 of the protein-protein interaction network (category, Interpro).

TermName/functionCountP-valueGenes
IPR001752Kinesin, motor region30.001518KIF2C, KIF11, KIF20A
IPR019821Kinesin, motor region, conserved site30.001518KIF2C, KIF11, KIF20A
IPR013212Mad3/BUB1 homology region 120.002759BUB1, BUB1B
IPR015661Mitotic checkpoint, serine/threonine protein kinase, BUB120.002759BUB1, BUB1B
IPR008271Serine/threonine protein kinase, active site40.012294BUB1, BUB1B, AURKA, AURKB
IPR014400Cyclin A/B/D/E20.016448CCNB1, CCNA2
IPR004367Cyclin, C-terminus20.019164CCNB1, CCNA2
IPR017441Protein kinase, ATP binding site40.023893BUB1, BUB1B, AURKA, AURKB
IPR006671Cyclin, N-terminus20.044611CCNB1, CCNA2
IPR013763 Cyclin-associated20.049887CCNB1, CCNA2

[i] IPR, interpro; ATP, adenosine triphosphate.

Discussion

In the present study, the gene expression profiles of the OA group and the non-OA group were analyzed and 1,377 DEGs were identified, including 869 upregulated DEGs and 508 downregulated DEGs. The functional enrichment for the downregulated DEGs revealed that the FGF19 gene was involved in the response to stress as well as the defense response. According to Tew et al (20), apoptosis and proliferation are involved in the progression of cartilage wounding during the development of OA. Costouros and Kim (21) reported that the use of programmed cell death inhibitors can improve the results of cartilage repair surgeries. fibroblast growth factor (FGF)2 is a mitogen that induces chondrocyte proliferation in wounded cartilage (22). FGF18 induces the increase of cartilage thickness of the tibial plateau and also increases remodeling of the subchondral bone by repairing damaged cartilage in OA rats (23). FGF2 and FGF18 are involved in cartilage wounding, which may be considered as one of the responses to stress. Therefore, the present study hypothesized that FGF19 may be involved in the repair of damaged cartilage in OA.

Functional enrichment analysis of the downregulated DEGs further revealed that KIF11 and KIF2C were involved in the response to stress, immune system process, response to wounding and mitotic cell cycle. In addition, the protein domain enrichment analysis of the genes in module 2 showed that KIF2C, KIF11 and KIF20A, were significantly enriched in the protein domain of IPR001752: Kinesin, motor region. KIF2C, KIF11 and KIF20A belong to the Kinesin family of proteins. Kinesins are important in cell division and at least 12 kinesins are involved in mitosis and cytokinesis (24). KIF22 is a member of the kinesin-like protein family and KIF22 mRNA was detected in human bone, cartilage, joint capsules, ligaments, skin and primary cultured chondrocytes (25). Mutations in KIF22 may be the cause of spondyloepimetaphyseal dysplasia with joint laxity, which is an autosomal dominant skeletal disorder (26). Phosphocitrate is known to downregulate multiple genes responsible for cell proliferation. KIF23 and KIFC1 were shown to be differentially expressed between phosphocitrate-treated and -untreated telomerase-transduced OA 13A fibroblast-like synoviocytes (27). KIF2C was also identified as a DEG in phosphocitrate-treated vs. untreated OA meniscal cells (28). These results suggested that KIF2C, KIF11 and KIF20A may be involved in the mitotic cell cycle and contribute to the development of OA.

In addition, functional enrichment analysis of the down-regulated DEGs revealed that BUB1, BUB1B, CCNB1 and CCNA2 were involved in the mitotic cell cycle process. Furthermore, the protein domain enrichment analysis of the genes in module 2, including BUB1 and BUB1B, revealed that these genes were significantly enriched in the protein domain of IPR013212: Mad3/BUB1 homology region (1), and the genes in module 2, including CCNB1 and CCNA2, were significantly enriched in the protein domain of IPR014400: Cyclin A/B/D/E. Several previous studies have reported that BUB1, BUB1B, CCNB1 and CCNA2 had roles in the regulation of the cell cycle (2932). According to Tew et al (20), apoptosis and proliferation were involved in processes of cartilage wounding. Based on these results, the present study hypothesized that BUB1, BUB1B, CCNB1 and CCNA2 affected the development of OA through the cell cycle.

In addition, upregulated DEGs, including CD36, COL11A 2, COL1A1, COL2A1 and COL3A1, were revealed to be involved in the ECM and ECM-receptor interaction pathway. The structural matrix macromolecules in the ECM and growth factors regulate the chondrocyte function via specific membrane receptors (33). TSP-1 is involved in the cell-matrix interactions of various tissues in cartilage. The number of CD36-positive chondrocytes, which is considered to be the receptor of TSP-1, is significantly increased in severely osteoarthritic cartilage (34). The expression of CD36 patterning receptor has attenuating effects on the inflammatory, pro-catabolic responses to S100A11 and tumor necrosis factor α in chondrocytes (35). The α1(X) collagen gene COL10A1 is a hypertrophic chondrocytes-specific molecular marker and a transcriptional target of RUNX2 during chondrogenesis (36,37). RUNX2 also regulates the expression of COL10A1 in hypertrophic chondrocytes and matrix metallopeptidase 13 in terminal hypertrophic chondrocytes (38). The collagen genes (COL11A 2, COL1A1 and COL2A1) are involved in the progression of OA through the mediation of sensitivity to OA (39). The upregulated expression of COL1A1 and COL2A1 reflects the metabolic activation of OA chondrocytes, and the expression of COL11A2 is also upregulated with the progression of OA (40). The results suggested that the DEGs associated with the ECM are important in the regulation of chondrocyte function.

In conclusion, in the present study, gene expression analysis was performed and 1,377 DEGs were identified, among which 869 DEGs were upregulated and 508 DEGs were downregulated. The identified DEGs may be involved in the pathogenesis of OA, particularly FGF19, BUB1, KIF2C, CD36 and COL11A2, which are involved in the cellular stress response, cell cycle and the ECM. However, the results of the present study require confirmation by further studies.

References

1 

Fransen M, Bridgett L, March L, Hoy D, Penserga E and Brooks P: The epidemiology of osteoarthritis in Asia. Int J Rheum Dis. 14:113–121. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Mahjoub M, Berenbaum F and Houard X: Why subchondral bone in osteoarthritis? The importance of the cartilage bone interface in osteoarthritis. Osteoporos Int. 23(Suppl 8): S841–S846. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Henrotin Y, Pesesse L and Sanchez C: Subchondral bone and osteoarthritis: Biological and cellular aspects. Osteoporos Int. 23:S847–S851. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Chou CH, Wu CC, Song IW, Chuang HP, Lu LS, Chang JH, Kuo SY, Lee CH, Wu JY, Chen YT, et al: Genome-wide expression profiles of subchondral bone in osteoarthritis. Arthritis Res Ther. 15:R1902013. View Article : Google Scholar : PubMed/NCBI

5 

Chou CH, Wu CC, Song IW, Chuang HP, Lu LS, Chang JH, Kuo SY, Lee CH, Wu JY and Chen YT: Genome-wide expression profiles of subchondral bone in osteoarthritis. Arthritis Res Ther. 15:R1902013. View Article : Google Scholar : PubMed/NCBI

6 

Zhen G, Wen C, Jia X, Li Y, Crane JL, Mears SC, Askin FB, Frassica FJ, Chang W, Yao J, et al: Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nat Med. 19:704–712. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Valverde-Franco G, Pelletier JP, Fahmi H, Hum D, Matsuo K, Lussier B, Kapoor M and Martel-Pelletier J: In vivo bone-specific EphB4 overexpression in mice protects both subchondral bone and cartilage during osteoarthritis. Arthritis Rheum. 64:3614–3625. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Karlsson C, Dehne T, Lindahl A, Brittberg M, Pruss A, Sittinger M and Ringe J: Genome-wide expression profiling reveals new candidate genes associated with osteoarthritis. Osteoarthritis Cartilage. 18:581–592. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Gautier L, Cope L, Bolstad BM and Irizarry RA: Affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Smyth G and Limma: Linear models for microarray data Bioinformatics and Computational Biology Solutions using R and Bioconductor 2005. Springer; New York: pp. 397–420. 2005

12 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: A practical and powerful approach to multiple testing. JR Stat Soc. 57:289–300. 1995.

13 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene Ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

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

16 

Saito R, Smoot ME, Ono K, Ruscheinski J, Wang PL, Lotia S, Pico AR, Bader GD and Ideker T: A travel guide to Cytoscape plugins. Nat Methods. 9:1069–1076. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Adamcsek B, Palla G, Farkas IJ, Derényi I and Vicsek T: CFinder: Locating cliques and overlapping modules in biological networks. Bioinformatics. 22:1021–1023. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Derényi I, Palla G and Vicsek T: Clique percolation in random networks. Phys Rev Lett. 94:1602022005. View Article : Google Scholar : PubMed/NCBI

19 

Apweiler R, Attwood TK, Bairoch A, Bateman A, Birney E, Biswas M, Bucher P, Cerutti L, Corpet F, Croning MD, et al: The InterPro database, an integrated documentation resource for protein families, domains and functional sites. Nucleic Acids Res. 29:37–40. 2001. View Article : Google Scholar :

20 

Tew SR, Kwan AP, Hann A, Thomson BM and Archer CW: The reactions of articular cartilage to experimental wounding: Role of apoptosis. Arthritis Rheum. 43:215–225. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Costouros JG and Kim HT: Preventing chondrocyte programmed cell death caused by iatrogenic injury. Knee. 14:107–111. 2007. View Article : Google Scholar

22 

Khan IM, Palmer EA and Archer CW: Fibroblast growth factor-2 induced chondrocyte cluster formation in experimentally wounded articular cartilage is blocked by soluble Jagged-1. Osteoarthritis Cartilage. 18:208–219. 2010. View Article : Google Scholar

23 

Moore EE, Bendele AM, Thompson DL, Littau A, Waggie KS, Reardon B and Ellsworth JL: Fibroblast growth factor-18 stimulates chondrogenesis and cartilage repair in a rat model of injury-induced osteoarthritis. Osteoarthritis Cartilage. 13:623–631. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Zhu C, Zhao J, Bibikova M, Leverson JD, Bossy-Wetzel E, Fan JB, Abraham RT and Jiang W: Functional analysis of human microtubule-based motor proteins, the kinesins and dyneins, in mitosis/cytokinesis using RNA interference. Mol Biol Cell. 16:3187–3199. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Min BJ, Kim N, Chung T, Kim OH, Nishimura G, Chung CY, Song HR, Kim HW, Lee HR, Kim J, et al: Whole-exome sequencing identifies mutations of KIF22 in spondyloepimetaphyseal dysplasia with joint laxity, leptodactylic type. Am J Hum Genet. 89:760–766. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Boyden ED, Campos-Xavier AB, Kalamajski S, Cameron TL, Suarez P, Tanackovic G, Andria G, Ballhausen D, Briggs MD, Hartley C, et al: Recurrent dominant mutations affecting two adjacent residues in the motor domain of the monomeric kinesin KIF22 result in skeletal dysplasia and joint laxity. Am J Hum Genet. 89:767–772. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Sun Y, Mauerhan DR, Franklin AM, Norton J, Hanley EN Jr and Gruber HE: Phosphocitrate is potentially a disease-modifying drug for noncrystal-associated osteoarthritis. Biomed Res Int. 2013:3262672013. View Article : Google Scholar : PubMed/NCBI

28 

Sun Y, Roberts A, Mauerhan DR, Sun AR, Norton HJ and Hanley EN Jr: Biological activities of phosphocitrate: A potential meniscal protective agent. Biomed Res Int. 2013:7265812013. View Article : Google Scholar : PubMed/NCBI

29 

Taylor SS and Mckeon F: Kinetochore localization of murine Bub1 is required for normal mitotic timing and checkpoint response to spindle damage. Cell. 89:727–735. 1997. View Article : Google Scholar : PubMed/NCBI

30 

Davenport JW, Fernandes ER, Harris LD, Neale GA and Goorha R: The mouse mitotic checkpoint gene bub1b, a novel bub1 family member, is expressed in a cell cycle-dependent manner. Genomics. 55:113–117. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Sartor H, Ehlert F, Grzeschik KH, Müller R and Adolph S: Assignment of two human cell cycle genes, CDC25C and CCNB1, to 5q31 and 5q12, respectively. Genomics. 13:911–912. 1992. View Article : Google Scholar : PubMed/NCBI

32 

Zhang C, Wang L, Wu D, Chen H, Chen Z, Thomas-Ahner JM, Zynger DL, Eeckhoute J, Yu J, Luo J, et al: Defnition of a FoxA1 Cistrome that is crucial for G1 to S-phase cell-cycle transit in castration-resistant prostate cancer. Cancer Res. 71:6738–6748. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Van Der Kraan PM, Buma P, Van Kuppevelt T and Van Den Berg WB: Interaction of chondrocytes, extracellular matrix and growth factors: Relevance for articular cartilage tissue engineering. Osteoarthritis Cartilage. 10:631–637. 2002. View Article : Google Scholar : PubMed/NCBI

34 

Pfander D, Cramer T, Deuerling D, Weseloh G and Swoboda B: Expression of thrombospondin-1 and its receptor CD36 in human osteoarthritic cartilage. Ann Rheum Dis. 59:448–454. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Cecil DL, Appleton CT, Polewski MD, Mort JS, Schmidt AM, Bendele A, Beier F and Terkeltaub R: The pattern recognition receptor CD36 is a chondrocyte hypertrophy marker associated with suppression of catabolic responses and promotion of repair responses to inflammatory stimuli. J Immunol. 182:5024–5031. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Zheng Q, Zhou G, Morello R, Chen Y, Garcia-Rojas X and Lee B: Type X collagen gene regulation by Runx2 contributes directly to its hypertrophic chondrocyte-specific expression in vivo. J Cell Biol. 162:833–842. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Lin AC, Seeto BL, Bartoszko JM, Khoury MA, Whetstone H, Ho L, Hsu C, Ali SA and Alman BA: Modulating hedgehog signaling can attenuate the severity of osteoarthritis. Nat Med. 15:1421–1425. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Komori T: Regulation of bone development and extracellular matrix protein genes by RUNX2. Cell Tissue Res. 339:189–195. 2010. View Article : Google Scholar

39 

Jin SY, Hong SJ, Yang HI, Park SD, Yoo MC, Lee HJ, Hong MS, Park HJ, Yoon SH, Kim BS, et al: Estrogen receptor-alpha gene haplotype is associated with primary knee osteoarthritis in Korean population. Arthritis Res Ther. 6:R415–R421. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Lamas JR, Rodríguez-Rodríguez L, Vigo AG, Alvarez-Lafuente R, López-Romero P, Marco F, Camafeita E, Dopazo A, Callejas S, Villafuertes E, et al: Large-scale gene expression in bone marrow mesenchymal stem cells: A putative role for COL10A1 in osteoarthritis. Ann Rheum Dis. 69:1880–1885. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 12 Issue 4

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
Sun J, Yan B, Yin W and Zhang X: Identification of genes associated with osteoarthritis by microarray analysis. Mol Med Rep 12: 5211-5216, 2015
APA
Sun, J., Yan, B., Yin, W., & Zhang, X. (2015). Identification of genes associated with osteoarthritis by microarray analysis. Molecular Medicine Reports, 12, 5211-5216. https://doi.org/10.3892/mmr.2015.4048
MLA
Sun, J., Yan, B., Yin, W., Zhang, X."Identification of genes associated with osteoarthritis by microarray analysis". Molecular Medicine Reports 12.4 (2015): 5211-5216.
Chicago
Sun, J., Yan, B., Yin, W., Zhang, X."Identification of genes associated with osteoarthritis by microarray analysis". Molecular Medicine Reports 12, no. 4 (2015): 5211-5216. https://doi.org/10.3892/mmr.2015.4048