Identification of differentially expressed genes and their subpathways in recurrent versus primary bone giant cell tumors

  • Authors:
    • Shuxin Chen
    • Chunquan Li
    • Bingli Wu
    • Chunlong Zhang
    • Cheng Liu
    • Xiaoxu Lin
    • Xiangqiao Wu
    • Lingling Sun
    • Chunpeng Liu
    • Bo Chen
    • Zhigang Zhong
    • Liyan Xu
    • Enmin Li
  • View Affiliations

  • Published online on: June 18, 2014     https://doi.org/10.3892/ijo.2014.2501
  • Pages: 1133-1142
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Giant cell tumor (GCT) of the bone is a benign but locally aggressive bone neoplasm with a strong tendency to develop local recurrent and metastatic disease. Thus, it provides a useful model system for the identification of biological mechanisms involved in bone tumor progression and metastasis. This study profiled 24 cases of recurrent versus primary bone GCT tissues using QuantiGene 2.0 Multiplex Arrays that included Human p53 80-Plex Panels and Human Stem Cell 80-Plex Panels. A total of 32 differentially expressed genes were identified, including the 20 most upregulated genes and the 12 most downregulated genes in recurrent GCT. The genes identified are related to cell growth, adhesion, apoptosis, signal transduction and bone formation. Furthermore, iSubpathwayMiner analyses were performed to identify significant biological pathway regions (subpathway) associated with this disease. The pathway analysis identified 11 statistically significant enriched subpathways, including pathways in cancer, p53 signaling pathway, osteoclast differentiation pathway and Wnt signaling pathway. Among these subpathways, four genes (IGF1, MDM2, STAT1 and RAC1) were presumed to play an important role in bone GCT recurrence. The differentially expressed MDM2 protein was immunohistochemically confirmed in the recurrent versus primary bone GCT tissues. This study identified differentially expressed genes and their subpathways in recurrent GCT, which may serve as potential biomarkers for the prediction of GCT recurrence.

Introduction

Giant cell tumor of the bone is a relatively uncommon neoplasm, which is a benign but locally aggressive bone neoplasm characterized by massive bone destruction at the epiphysis of the long bone and has a strong tendency to develop local recurrence and metastasis (1). GCT accounts for 4–5% of primary bone tumors and up to 20% of benign bone tumors (2). Statistically, 80% of GCTs have a benign clinical course with a local recurrent rate of 20–50%. Approximately 10% will undergo malignant transformation and 1–4% will have pulmonary metastases even in cases with a benign histology (3). In China, GCT incidence is significantly higher and observed in roughly 20% of all primary bone tumors (4). To date, surgery is the primary treatment for GCT with unresectable tumors being treated with radiotherapy (5), and these treatment regimens have remained unchanged for much of the past three decades, which is partially due to the lack of randomized clinical trials (4) and lack of chemotherapy options. Since the tissue origin of GCT remains to be determined, and its clinical behavior is unpredictable, the accurate prediction of its recurrence and metastasis is still not available using clinical diagnosis, radiology and histology (6). Thus, novel approaches are urgently required to better understand the molecular mechanisms of GCT carcinogenesis and to therefore provide meaningful strategies for the effective control of GCT in the clinic.

Currently, profiling of altered genes and pathways using gene chips is a useful method and an efficient alternative strategy to establish disease-pathway relationships (7,8). Information on disease-related genes, such as from the Genetic Association Database (GAD) (9), is increasingly available for constructing high quality disease-metabolic pathway relationships. Different expression profiles of the p53 pathway and stem cell pathway genes was considered to be a major cause of the occurrence of GCT and promoters of malignant transformation and metastasis (10). However, little is known about the role that the p53 pathway plays underlying tumorigenesis and development of recurrent GCT. Moreover, there is strong evidence showing that the neoplastic cells of GCT are developed from mesenchymal stem cells (11). In recent years, more attention has been paid to subpathway (local area of the entire biological pathway), which can provide more detailed information of complex diseases in high-throughput data analysis, because critical genes may not be significantly enriched in the whole pathway, but nevertheless play key roles (12,13). Therefore, in this study, we profiled differentially expressed genes in recurrent versus primary GCT tissues and identified significant subpathways to further explore the biological mechanisms involved in the recurrence of GCT. Thus, the aim of this study was to improve the understanding of these genes and pathways in the regulation of GCT invasion, recurrence and metastasis, and therefore to evaluate them as potential biomarkers for the early detection and prediction of tumor recurrence.

Materials and methods

Study population

A total of 24 cases of bone GCT, including 12 primary and 12 recurrent tumors, were obtained from 17 GCT patients who were surgically treated in the Department of Orthopedics Surgery, Shantou Hospital of Zhongshan University, between March 2001 and April 2010. All cases were diagnosed by the experienced subspecialty bone and soft-tissue pathologists, and confirmed by another study pathologist. The clinicopathological data of each patient were retrieved from their medical records and are summarized in Table I. The Institutional Review Board of Shantou Hospital of Zhongshan University approved the study protocol and each patient signed an informed consent form before recruitment into this study.

Table I

Clinical characteristics of recurrent and primary bone GCT patients.

Table I

Clinical characteristics of recurrent and primary bone GCT patients.

Case Primary/recurrentSexAge (years)SiteCampanacci’s gradingSurgical treatment
1PrimaryFemale25FemurICurettage
RecurrencesFemale27FemurIIIWide resection
2PrimaryMale44RadiusICurettage
RecurrencesMale46RadiusIICurettage
RecurrencesMale48RadiusIIIWide resection
3PrimaryMale23TibiaICurettage
RecurrencesMale24TibiaIICurettage
4PrimaryFemale20VertebraICurettage
RecurrencesFemale21VertebraIICurettage
5PrimaryFemale26TibiaICurettage
RecurrencesFemale26TibiaICurettage
RecurrencesFemale27TibiaIICurettage
6PrimaryMale18FibulaIWide resection
7RecurrencesMale40HumerusIIIWide resection
8RecurrencesMale49FemurIIIWide resection
9RecurrencesMale28TibiaIIICurettage
10RecurrencesMale48FemurIIIWide resection
11PrimaryFemale45RadiusIICurettage
12RecurrencesMale28FemurIIIWide resection
13PrimaryFemale19FemurIICurettage
14PrimaryMale50HumerusIICurettage
15PrimaryFemale31FemurIICurettage
16PrimaryFemale20MetacarpusICurettage
17PrimaryMale23TibiaICurettage
Whole genome cDNA QuantiGene 2.0 microarray analysis

Formalin-fixed and paraffin-embedded (FFPE) non-cancer and cancer tissues were isolated, separately by scraping, and placed into 1.5-ml microcentrifuge tubes for processing of tissue homogenates according to the procedure as described in the QuantiGene sample processing kit for FFPE tissues (Affymetrix, Inc., Santa Clara, CA, USA). Briefly, 300 μl of homogenizing tissue mixture, containing 10 deparaffinized 10-μm sections, were supplemented with 3 μl of proteinase K (50 μg/μl) and incubated overnight at 65°C. The following day, the tissue homogenates were separated from debris by brief centrifugation and transferred to a new tube. The resulting tissue homogenates were frozen at −80°C and stored until further use.

A QuantiGene 2.0 Multiplex assay system, containing Human p53 80-Plex Panels and Human Stem Cell 80-Plex Panels, was purchased from Affymetrix, Inc. The QuantiGene 80-Plex assay was performed according to the recommended protocol of QuantiGene 2.0 reagent systems (Affymetrix, Inc.). Briefly, 40 μl of tissue homogenate was mixed with 33.3 μl of lysis mixture, l μl of blocking reagent, 0.3 μl of 2.0 probe set, and 25.4 μl of nuclease-free water. The reactions were placed in a 96-well capture plate covalently coated with capture probes and incubated for 16 h at 54°C. Wells were washed three times with wash buffer to remove unbound material. For signal amplification, with 100 μl of 2.0 Pre-Amplifier working reagent, 100 μl of Amplifier working reagent was added to each sample and incubated for 1 h each at 50°C, respectively. To detect the signal, to each sample was added 100 μl of 2.0 substrate, the samples were sealed and incubated for 5 min. Luminescence levels were then measured using a luminometer (Victor Light; Perkin-Elmer, Waltham, MA, USA). Duplicate assays were performed for all samples, and homogenizing buffer was used as background control. To verify that the resulting assay signals were linearly proportional to the sample input, a 2-fold dilution series of each sample was performed. The RNA level of PGK1, TBP, HPRT1, GUSB and TFRC (reference genes) were measured to normalize the data.

Function enrichment analysis

We used the iSubpathwayMiner package that was developed by our laboratory (12) to identify the pathways of the differentially expressed genes in recurrent vs. primary bone GCT tissues. The tool was an R package for flexible biological pathway identification from the KEGG database (14), which covered not only the entire pathway level but also the subpathway level. During enrichment analyses, we performed entire pathway and subpathway identification for the differentially expressed genes based on the hypergeometric test. The corresponding GCT data were integrated with p53 gene and stem cell data and then into the corresponding gene product nodes (referred to as signature nodes) within the pathway. The lenient distance similar to the signature nodes within the pathway structure were analyzed to locate key cascade subpathway regions. Finally, a hypergeometric test was used to evaluate the enrichment significance of these subpathway regions.

Immunohistochemistry

We also performed immunohistochemistry using the PV-9000 2-step plus Poly-HRP anti-mouse/rabbit IgG detection system (ZSGB-BIO) and the liquid DAB substrate kit (ZSGB-BIO) to assess expression of MDM2 in GCT tissues using an MDM2 antibody (cat no. ZA-0519; ZSGB-BIO, Beijing, China). Briefly, 24 cases of GCT tissues were built to form a tissue microarray (TMA) and prepared for 4 μm sections. For immunohistochemistry, the TMA sections were subjected to dewaxing in xylene and rehydration in a series of graded alcohols, and then subjected to antigen retrieval with a pressure cooker for 10 min in 0.01 M sodium citrate buffer (pH 6.0). After that, the sections were submerged in a peroxidase quenching solution, containing one part of 30% hydrogen peroxide to nine parts of distilled water, for 10 min and then washed with phosphate-buffered saline (PBS) three times for 2 min each. The sections were incubated in a moist chamber with 0.1 ml of blocking serum solution for 10 min and then further incubated with 0.1 ml primary antibody for 30 min. After rinsing with PBS three times for 2 min each, 0.1 ml of HRP polymer conjugate was added to each section and incubated for 10 min, followed by a rinse with PBS. Next, the sections were incubated with DAB chromogen solution for 3–10 min and subsequently counterstained with Mayer’s hematoxylin, dehydrated and mounted. The negative controls were incubated with 10% normal goat serum to substitute the primary antibody. Immunostained TMA sections were then reviewed and scored in a blinded manner by at least two independent investigators. The positive signal was observed in tumor cell cytoplasm, and scored as the estimated percentage of staining. MDM2 immunoreactivity was classified into three categories as negative (<20% tumor cells displaying cytoplasmic staining); heterogeneous (20–79% tumor cells with cytoplasmic reactivity); and homogeneous (>80% tumor cells with intense cytoplasmic staining).

Statistical analysis

All statistical analyses were performed by using SPSS 11.0 software (SPSS, Chicago, IL, USA). Statistical analyses between primary and recurrent groups were determined by using the Kruskal Wallis test. A P-value <0.05 was considered statistically significant.

Results

Identification of differentially expressed genes in recurrent vs. primary bone GCT

In this study, we analyzed differentially expressed genes in recurrent vs. primary bone GCT tissues using QuantiGene 2.0 Multiplex assay. We identified a total of 32 differentially expressed genes using fold-change (FD >2 or FD <0.5), including 20 most upregulated genes and 12 most downregulated genes in recurrent bone giant cell tumor tissues versus the primary tumors (Table II). These genes are related to cell growth, adhesion, apoptosis, signal transduction, and bone formation, indicating that they may play roles in bone GCT progression, such as recurrence or metastasis.

Table II

Differentially expressed genes between the primary and recurrent bone giant cell tumor tissues.

Table II

Differentially expressed genes between the primary and recurrent bone giant cell tumor tissues.

NameFull nameFold change
NANOGNanog homeobox62.01
CD4CD4 molecule23.97
TIMP3Tissue inhibitor of metalloproteinases 313.31
ADARAdenosine deaminase, RNA-specific10.63
MDM2Murine double minute 26.17
NUMBNumb homolog (Drosophila)4.98
STAT1Signal transducer and activator of transcription 14.89
BAXBCL2-associated X protein4.16
PAFAH1B1Platelet-activating factor acetylhydrolase 1b, regulatory subunit 1 (45 kDa)3.96
RAC1Ras-related C3 botulinum toxin substrate 13.62
CDH2Cadherin 2, type 1, N-cadherin (neuronal)3.47
NFKB1Nuclear factor of κ light polypeptide gene enhancer in B-cells 13.32
NCSTNNicastrin3.00
CD8ACD8a molecule2.27
PGK1Phosphoglycerate kinase 11.71
IGF1Insulin-like growth factor 1 (somatomedin C)1.47
BTG2BTG family, member 21.34
MMEMembrane metallo-endopeptidase1.17
CXCL12Chemokine (C-X-C motif) ligand 121.10
JUNJun proto-oncogene0.21
FZD1Frizzled family receptor 1−1.81
HK2Hexokinase 2−1.86
TFRCTransferrin receptor (p90, CD71)−2.22
E2F1E2F transcription factor 1−3.38
MYCv-myc myelocytomatosis viral oncogene homolog (avian)−7.67
DVL3Dishevelled, dsh homolog 3 (Drosophila)−8.62
BMP1Bone morphogenetic protein 1−10.28
EGR1Early growth response 1−14.41
FGFR1Fibroblast growth factor receptor 1−20.25
BMP2Bone morphogenetic protein 2−36.26
COL1A1Collagen, type I, α 1−469.8

[i] SERPINE1 Serpin peptidase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1 −37.99

Functional analysis of the differentially expressed genes

We performed pathway enrichment analysis using the differentially expressed genes in recurrent GCT tissues and identified six gene pathways (Table III). We then used the gene pathway data to locate the important pathway regions, and tested the regions by entering the differentially expressed genes into the p53 and stem data set of 150 genes for the pathway enrichment. Thus, we found a total of 11 subpathways (Table IV). It needs to be pointed out that 6 of these subpathways were not identifiable by the entire pathway identification method. Only focal adhesion pathway is significant in both entire and subpathway identification methods. If we only adopted the entire pathway identification method, these pathways could be ignored due to their high P-values threshold. However, these pathways were found statistically significant using the subpathway identification method. The result indicated that these subpathways may be associated with GCT recurrence.

Table III

The newly identified gene pathways after the pathway enrichment analysis of the differentially expressed genes.

Table III

The newly identified gene pathways after the pathway enrichment analysis of the differentially expressed genes.

Pathway IDPathway nameP-valueAnn molecule listAnnBg molecule list
path:04640Hematopoietic cell lineage0.007708CD8A; TFRC; MME; CD4CD8A; TFRC; MME; CD44; IL6; TNF; CD4
path:04145Phagosome0.018792TFRC; RAC1TFRC; RAC1
path:04974Protein digestion and absorption0.018792MME; COL1A1MME; COL1A1
path:05340Primary immunodeficiency0.018792CD8A; CD4CD8A; CD4
path:04514Cell adhesion molecules (CAMs)0.036157CD8A; CDH2; CD4CD8A; VCAN; NCAM1; CDH2; CDH1; CD4
path:04510Focal adhesion0.049058IGF1; COL1A1; JUN; RAC1MAPK8; RHOA; IGF1; CCND1; COL1A1; PTEN; JUN; BCL2; PRKCA; IGF1R; RAC1

Table IV

The statistically significant subpathways identified by iSubpathwayMiner.

Table IV

The statistically significant subpathways identified by iSubpathwayMiner.

Subpathway IDPathway nameP-valueDifferential genes within the subpathwayGenes within the subpathway
path:05200_1Pathways in cancer0.002258DVL3; FZD1; IGF1; STAT1; FGFR1; NFKB1; MDM2; MYC; RAC1DVL3; DVL1; FZD1; RHOA; FZD8; FZD3; IGF1; STAT1; TCF7; FGFR1; NFKB1; MDM2; MYC; KRAS; RELA; IGF1R; RAC1
path:04062_1Chemokine signaling pathway0.007513STAT1; NFKB1; CXCL12; RAC1STAT1; NFKB1; CXCL12; RELA; RAC1
path:04115_1p53 signaling pathway0.007513IGF1; SERPINE1; MDM2; BAXIGF1; TP53; SERPINE1; MDM2; BAX
path:04380_1Osteoclast differentiation0.007513STAT1; NFKB1; JUN; RAC1STAT1; NFKB1; JUN; RELA; RAC1
path:04010_1MAPK signaling pathway0.019088NFKB1; MYC; JUN; RAC1MAPK8; NFKB1; MYC; JUN; RELA; RAC1
path:04510_1Focal adhesion0.019088IGF1; COL1A1; JUN; RAC1MAPK8; IGF1; COL1A1; JUN; IGF1R; RAC1
path:05215_1Prostate cancer0.019088IGF1; FGFR1; NFKB1; MDM2IGF1; FGFR1; NFKB1; MDM2; RELA; IGF1R
path:05220_1Chronic myeloid leukemia0.030663NFKB1; MDM2; MYCNFKB1; MDM2; MYC; RELA
path:04310_2Wnt signaling pathway0.036904DVL3; FZD1; MYC; JUN; RAC1DVL3; DVL1; FZD1; FZD8; FZD3; TCF7; AXIN1; MYC; JUN; RAC1
path:04722_1Neurotrophin signaling pathway0.037737NFKB1; JUN; BAX; RAC1MAPK8; TP53; NFKB1; JUN; BAX; RELA; RAC1
path:00010_1 Glycolysis/gluconeogenesis0.044385HK2; PGK1HK2; PGK1

The most significant subpathway was the ‘pathways in cancer’. GCT, as tumor disease was obviously associated with the dysregulation of the pathway. We found that the differential gene IGF1, a growth factor, was located in the starting regions in the pathway (Fig. 1).

The third subpathway (path:04115_1) in Table I belonged to the p53 signaling pathway. The pathway was reported to be highly associated with giant cell tumor of bone (GCTB)(1517). Gene p53 was located in the center of the pathway and identified within the subpathway path:04115_1 (Fig. 2). Moreover, MDM2 was localized in the central region of this subpathway.

The fourth subpathway path:04380_1, which belonged to the osteoclast differentiation pathway, was identified as significant in iSubpathwayMiner, which yielded a P-value of 0.0075. This pathway was associated with the localized bone destruction of GCTB (18). Mononuclear stromal cells in GCTs of bone have shown characteristics of the osteoblast lineage by expressing many osteoblast-associated differentiation markers (19). The bone resorption activity of osteoclasts can cause destructive osteolysis and consequent morbidity in GCT (18). The subpathway path:04380_1 contained four differential genes: JUN, NFKB1, STAT1 and RAC1 (Fig. 3).

The ninth subpathway (path:04310_2) belonged to the Wnt signaling pathway. The subpathway path:04310_2 contained MYC, JUN, and RAC1, most of which were localized in the endpoint of this pathway (Fig. 4).

Immunohistochemical validation of MDM2 expression in bone GCT tissues

After the above pathway analysis, we identified many risk genes in recurrent GCT. Among these risk genes, we chose MDM2 for immunohistochemical analysis in bone GCT tissues. MDM2 protein was observed in both multinucleated giant cells and mononuclear stromal cells in GCT tissues (Fig. 5). Comparing immunohistochemical results between the primary and recurrent bone GCT tissues, MDM2 was statistically significantly higher in recurrent tumors than primary tumors (P=0.015, χ2=5.86). MDM2 is involved in the occurrence and development of GCT.

Discussion

Bone GCT is a benign but locally aggressive bone neoplasm with a strong tendency to develop a local recurrent and metastatic disease. Recently, two studies attempted to identify differentially expressed genes associated with GCT development and progression (20,21). Altered expression of Ephrin A receptor, Claudin 7, CD52, FGFR3, and AMFR was found by Guenther et al (20), whereas Skubitz et al (21) reported that genes found to be overexpressed in GCTs included tartrate-resistant acid phosphatase, the lysosomal H+-transporting ATPase, and osteoprotegrin ligand (OPGL). In our present study, we identified 32 differentially expressed genes in the primary vs. recurrent bone GCT tissues and we then used iSubpathwayMiner to annotate them into gene subpathways resulting 11 statistically significantly enriched subpathways. This study is just the first step to identify genes that are associated with bone GCT recurrence and further study is warranted to investigate them mechanistically in bone GCT to provide biomarkers or therapeutic targets.

The present study identified several critical genes and gene pathways that are associated with bone GCT recurrence. The first gene was IGF1, which regulates cell proliferation, differentiation and survival (22). IGFI plays an important role in normal bone growth, bone cell turnover and metabolism, and is a key factor in osteoblast proliferation and bone formation (23). IGF1 may be a possible prognostic marker useful in the identification of GCT patients at a higher risk of relapse with potential for development into a therapeutic agent against GCT. Moreover, the third subpathway (path:04115_1) belonged to the p53 signaling pathway and p53 is localized in the center of this pathway and identified within the subpathway path:04115_1. MDM2 was localized in the central region of this subpathway. Indeed, p53 is frequently mutated in GCT and could be useful in predicting tumor progression and local recurrence (15). p53 mutations were detected in the cases of secondary malignant giant-cell tumor without irradiation therapy (10). Masui et al showed that p53 expression levels correlated with the rates of lung metastasis and recurrence of GCT (16). These results suggest that p53 mutations may play an important role in malignant transformation of conventional GCT (17). MDM2 is a negative regulator of p53 and plays an important role in the p53-signaling pathway, suggesting a potentially high association with bone GCT development. Indeed, MDM2 has been found widely expressed in GCT (24). Thus, we further evaluated MDM2 expression in bone GCT tissues and the immune-reactivity of anti-MDM2 antibody was observed in osteoclast-like giant cells and mononuclear stromal cells. Statistical analyses showed that MDM2 expression was significantly higher in recurrent tumors than in primary tumors, suggesting that MDM2 might be associated with bone GCT recurrence.

Furthermore, the fourth subpathway path:04380_1 was the osteoclast differentiation pathway. This subpathway path:04380_1 contained four differentially expressed genes, including JUN, NFKB1, STAT1 and RAC1. c-Jun is a component of the heterodimeric AP-1 transcription factor and was highly expressed in GCT stromal cells (25). JUN may also be involved in upregulation of matrix-metalloproteinases in GCT. MMP-2, MMP-13, and MMP-9 have been shown to be highly expressed in GCT tissues (2628). Both MMP-2 and MMP-9 display several AP-1 consensus sequences within their promoter regions and may be directly upregulated by JUN (29). MMP-13 is responsible for optimizing the bone resorption capability of the giant cells, which is likely to be due to recruiting them to the bone surface (27,30). JUN could influence normal ECM physiology, thereby promoting growth and destructiveness of GCT (27). NF-κB has been shown to play an important role in many types of cancer and may also regulate tumor angiogenesis and invasiveness. NF-κB provides a mechanistic link between inflammation and cancer and is a major transcriptional factor controlling the ability of both pre-neoplastic and malignant cells to resist apoptosis-based tumor-surveillance mechanisms (30). RANKL, as a negative regulator of NF-κB, was identified as essential for osteoclast physiology (31,32). RANKL is highly expressed in stromal cells of GCT (33,34). As a potential therapeutic target of bone disease, Amgen developed a monoclonal antibody to RANKL (denosumab). Denosumab was studied in a recent proof-of-principle phase II study of 35 patients with recurrent or unresectable GCT (35). Twenty-six of the 31 patients with data reported reduced pain or improvement in the functional status. Radiologic evidence of bone repair was reported in nine patients. The treatment was generally well tolerated without treatment-related serious adverse events. Thus, blockade of RANKL signaling in patients with advanced or unresectable GCT could provide objective changes in tumor composition, reduced bony destruction, and clinical benefit. Signal transducer and activator of transcription 1 (STAT1), localized in the starting region of the subpathway path:04380_1 (Fig. 3), was reported to be associated with human breast cancer, melanoma, leukemia, lymphoma, and other cancers (36,37). STAT1 has a critical role in regulation of bone growth and bone formation (38).

In addition, the ninth subpathway (path:04310_2) belongs to the Wnt signaling pathway. Several studies have shown that this pathway plays an important role in regulation of skeletal function, and the activation of Wnt signaling may induce osteoblast differentiation and osteoclastogenesis during the bone resorption process (3941). Bone GCT was found associated with activation of the Wnt signaling pathway (42). This subpathway contained MYC, JUN, and RAC1, most of which were localized at the endpoint of this pathway. Gamberi et al (43) showed a strong correlation between c-Myc overexpression and GCT occurrence and its metastases. c-Myc protein was overexpressed in both giant cells and mononuclear cells, suggesting that both cell types are involved in progression of this tumor type (43). Previous studies found that Rac1 regulated a diverse array of cellular events, including formation of lamellipodia and membrane ruffles, cell cycle, cell adhesion and mobility (4446). Rac1 is thought to play a significant role in the development of various cancers, including melanoma (45) and non-small cell lung cancer (46). As a result, it is now considered as a therapeutic target for these diseases (30). Rac1 can regulate survival signaling of osteoclasts and their bone resorption activity (47). A transgenic mouse model was used to confirm that Rac1 was the primary Rac isoform in regulating ROS production and the cytoskeleton organization during the multiple stages of osteoclast differentiation (48).

In conclusion, in the present study, we identified 32 genes that were differentially expressed in recurrent vs. primary bone GCT tissues and found them in multiple subpathways. Among them, four genes (IGF1, MDM2, STAT1 and RAC1) were located in key positions in these pathways. Further studies will confirm our current data and investigate their roles and functions in bone GCT progression.

References

1 

Gupta R, Seethalakshmi V, Jambhekar NA, et al: Clinico-pathologic profile of 470 giant cell tumors of bone from a cancer hospital in western India. Ann Diagn Pathol. 12:239–248. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Gamberi G, Serra M, Ragazzini P, et al: Identification of markers of possible prognostic value in 57 giant cell tumors of bone. Oncol Rep. 10:351–356. 2003.PubMed/NCBI

3 

Szendröi M: Giant cell tumor of bone. J Bone Joint Surg Br. 86:5–12. 2004.PubMed/NCBI

4 

Thomas DM and Skubitz KM: Giant cell tumour of bone. Curr Opin Oncol. 21:338–344. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Mendenhall WM, Zlotecki RA, Scarborough MT, Gibbs CP and Mendenhall NP: Giant cell tumour of bone. Am J Clin Oncol. 29:96–99. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Eckardt JJ and Grogan TJ: Giant cell tumor of bone. Clin Orthop Relat Res. 204:45–58. 1986.PubMed/NCBI

7 

Khatri P, Sellamuthu S, Malhotra P, et al: Recent additions and improvements to the Onto-Tools. Nucleic Acids Res. 33:W762–W765. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Wang K, Li M and Hakonarson H: Analysing biological pathways in genomewide association studies. Nat Rev Genet. 11:843–854. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Becker KG, Barnes KC, Bright TJ and Wang SA: The genetic association database. Nat Genet. 36:431–432. 2004. View Article : Google Scholar

10 

Papanastassiou I, Ioannou M, Papagelopoulos PJ, et al: P53 expression as a prognostic marker in giant cell tumor of bone: a pilot study. Orthopedics. 33:3072010.PubMed/NCBI

11 

Wülling M, Delling G and Kaiser E: The origin of the neoplastic stromal cell in giant cell tumor of bone. Hum Pathol. 34:983–993. 2003.PubMed/NCBI

12 

Li C, Li X, Miao Y, et al: SubpathwayMiner: a software package for flexible identification of pathways. Nucleic Acids Res. 37:e1312009. View Article : Google Scholar : PubMed/NCBI

13 

Li X, Li C, Shang D, et al: The implications of relationships between human diseases and metabolic subpathways. PLoS One. 6:e211312011. View Article : Google Scholar : PubMed/NCBI

14 

Kanehisa M, Goto S, Sato Y, Furumichi M and Tanabe M: KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40(Database issue): D109–D114. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Oda Y, Sakamoto A, Saito T, et al: Secondary malignant giant- cell tumor of bone: molecular abnormalities of p53 and H-ras gene correlated with malignant transformation. Histopathology. 39:6292001. View Article : Google Scholar : PubMed/NCBI

16 

Masui F, Ushigome S and Fujii K: Giant cell tumor of bone: a clinicopathologic study of prognostic factors. Pathol Int. 48:7231998. View Article : Google Scholar : PubMed/NCBI

17 

Gong L, Liu W, Sun X, et al: Histological and clinical characteristics of malignant giant cell tumor of bone. Virchows Arch. 460:327–334. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Boyle WJ, Simonet SW and Lacey DL: Osteoclast differentiation and activation. Nature. 423:337–342. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Murata A, Fujita T, Kawahara N, Tsuchiya H and Tomita K: Osteoblast lineage properties in giant cell tumors of bone. J Orthop Sci. 10:581–588. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Guenther R, Krenn V, Morawietz L, et al: Giant cell tumors of bone: molecular profiling and expression analysis of Ephrin A receptor, Claudin 7, CD52, FGFR3 and AMFR. Pathol Res Pract. 201:649–663. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Skubitz KM, Cheng EY, Clohisy DR, Thompson RC and Skubitz AP: Gene expression in giant cell tumors. J Lab Clin Med. 144:193–200. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Kappel CC, Velez-Yanguas MC, Hirschfeld S and Helman LJ: Human osteosarcoma cell lines are dependent on insulin-like growth factor I for in vitro growth. Cancer Res. 54:2803–2807. 1994.PubMed/NCBI

23 

Bonjour JP, Schurch MA, Chevalley T, Ammann P and Rizolli R: Protein intake. IGF-1 and osteoporosis. Osteoporosis Int. 7:36–42. 1997. View Article : Google Scholar

24 

de Souza PE, Paim JF, Carvalhais JN and Gomez RS: Immunohistochemical expression of p53, MDM2, Ki-67 and PCNA in central giant cell granuloma and giant cell tumor. J Oral Pathol Med. 28:54–58. 1999.PubMed/NCBI

25 

Mak IW, Turcotte RE, Popovic S, Singh G and Ghert M: AP-1 as a regulator of MMP-13 in the stromal cell of giant cell tumor of bone. Biochem Res Int. 2011:1641972011.PubMed/NCBI

26 

Rao VH, Singh RK, Delimont DC, et al: Interleukin-1beta upregulates MMP-9 expression in stromal cells of human giant cell tumor of bone. J Interferon Cytokine Res. 19:1207–1217. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Teti A, Farina AR, Villanova I, et al: Activation of MMP-2 by human GCT23 giant cell tumor cells induced by osteopontin, bone sialoprotein and GRGDSP peptides is RGD and cell shape change dependent. Int J Cancer. 77:82–93. 1998. View Article : Google Scholar

28 

Mak IW, Seidlitz EP, Cowan RW, et al: Evidence for the role of matrix metalloproteinase-13 in bone resorption by giant cell tumor of bone. Hum Pathol. 41:1320–1329. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Yoon SO, Park SJ, Yun CH and Chung AS: Roles of matrix metalloproteinases in tumor metastasis and angiogenesis. J Biochem Mol Biol. 36:128–137. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Karin M: Nuclear factor-kappaB in cancer development and progression. Nature. 441:431–436. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Dougall WC, Glaccum M, Charrier K, et al: RANK is essential for osteoclast and lymph node development. Genes Dev. 13:2412–2424. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Simonet WS, Lacey DL, Dunstan CR, et al: Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 89:309–319. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Morgan T, Atkins GJ, Trivett MK, et al: Molecular profiling of giant cell tumor of bone and the osteoclastic localization of ligand for receptor activator of nuclear factor kappaB. Am J Pathol. 167:117–128. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Geldyyev A, Koleganova N, Piecha G, et al: High expression level of bone degrading proteins as a possible inducer of osteolytic features in pigmented villonodular synovitis. Cancer Lett. 255:275–283. 2007. View Article : Google Scholar

35 

Thomas D, Henshaw R, Skubitz K, et al: Denosumab in patients with giant-cell tumor of bone: an open-label, phase 2 study. Lancet Oncol. 11:275–280. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Bowman T, Garcia R, Turkson J and Jove R: STATs in oncogenesis. Oncogene. 19:2474–2488. 2000. View Article : Google Scholar

37 

Boudný V, Kocák I, Lauerová L and Kovarík J: Interferon inducibility of STAT 1 activation and its prognostic significance in melanoma patients. Folia Biol. 49:142–146. 2003.PubMed/NCBI

38 

Sims NA, Jenkins BJ, Quinn JM, et al: Glycoprotein 130 regulates bone turnover and bone size by distinct downstream signaling pathways. J Clin Invest. 113:379–389. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Holmen SL, Zylstra CR, Mukherjee A, et al: Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem. 280:21162–21168. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Gaur T, Lengner CJ, Hovhannisyan H, et al: Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem. 280:33132–33140. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Goldring SR and Goldring MB: Eating bone or adding it: the Wnt pathway decides. Nat Med. 13:133–134. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Matsubayashi S, Nakashima M, Kumagai K, et al: Immunohistochemical analyses of beta-catenin and cyclin D1 expression in giant cell tumor of bone (GCTB): a possible role of Wnt pathway in GCTB tumorigenesis. Pathol Res Pract. 205:626–633. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Gamberi G, Benassi MS, Böhling T, et al: Prognostic relevance of C-myc gene expression in giant-ceIl tmnor of bone. J Orthop Res. 16:1–7. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Ridley AJ: Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol. 16:522–529. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Stallings-Mann ML, Waldmann J, Zhang Y, Miller E and Gauthier ML: Matrix metalloproteinase induction of Rac1b, a key effector of lung cancer progression. Sci Transl Med. 4:142ra952012.PubMed/NCBI

46 

McAllister SS: Got a light? Illuminating lung cancer. Sci Transl Med. 4:142fs222012.PubMed/NCBI

47 

Wang L, Kuang L, Pan X, et al: Isoalvaxant hone inhibits colon cancer cell proliferation, migration and invasion through inactivating Racl and AP-1. Int J Cancer. 127:1220–1229. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Fukuda A, Hikita A, Wakeyama H, et al: Regulation of osteoclast apoptosis and motility by small GTPase binding protein Rac1. J Bone Miner Res. 20:2245–2253. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2014
Volume 45 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen S, Li C, Wu B, Zhang C, Liu C, Lin X, Wu X, Sun L, Liu C, Chen B, Chen B, et al: Identification of differentially expressed genes and their subpathways in recurrent versus primary bone giant cell tumors. Int J Oncol 45: 1133-1142, 2014
APA
Chen, S., Li, C., Wu, B., Zhang, C., Liu, C., Lin, X. ... Li, E. (2014). Identification of differentially expressed genes and their subpathways in recurrent versus primary bone giant cell tumors. International Journal of Oncology, 45, 1133-1142. https://doi.org/10.3892/ijo.2014.2501
MLA
Chen, S., Li, C., Wu, B., Zhang, C., Liu, C., Lin, X., Wu, X., Sun, L., Liu, C., Chen, B., Zhong, Z., Xu, L., Li, E."Identification of differentially expressed genes and their subpathways in recurrent versus primary bone giant cell tumors". International Journal of Oncology 45.3 (2014): 1133-1142.
Chicago
Chen, S., Li, C., Wu, B., Zhang, C., Liu, C., Lin, X., Wu, X., Sun, L., Liu, C., Chen, B., Zhong, Z., Xu, L., Li, E."Identification of differentially expressed genes and their subpathways in recurrent versus primary bone giant cell tumors". International Journal of Oncology 45, no. 3 (2014): 1133-1142. https://doi.org/10.3892/ijo.2014.2501