Detection of significant pathways in osteoporosis based on graph clustering

  • Authors:
    • Haijun Xiao
    • Liancheng Shan
    • Haiming Zhu
    • Feng Xue
  • View Affiliations

  • Published online on: September 13, 2012     https://doi.org/10.3892/mmr.2012.1082
  • Pages: 1325-1332
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteoporosis is the most common and serious skeletal disorder among the elderly, characterized by a low bone mineral density (BMD). Low bone mass in the elderly is highly dependent on their peak bone mass (PBM) as young adults. Circulating monocytes serve as early progenitors of osteoclasts and produce significant molecules for bone metabolism. An improved understanding of the biology and genetics of osteoclast differentiation at the pathway level is likely to be beneficial for the development of novel targeted approaches for osteoporosis. The objective of this study was to explore gene expression profiles comprehensively by grouping individual differentially expressed genes (DEGs) into gene sets and pathways using the graph clustering approach and Gene Ontology (GO) term enrichment analysis. The results indicated that the DEGs between high and low PBM samples were grouped into nine gene sets. The genes in clusters 1 and 8 (including GBP1, STAT1, CXCL10 and EIF2AK2) may be associated with osteoclast differentiation by the immune system response. The genes in clusters 2, 7 and 9 (including SOCS3, SOD2, ATF3, ADM EGR2 and BCL2A1) may be associated with osteoclast differentiation by responses to various stimuli. This study provides a number of candidate genes that warrant further investigation, including DDX60, HERC5, RSAD2, SIGLEC1, CMPK2, MX1, SEPING1, EPSTI1, C9orf72, PHLDA2, PFKFB3, PLEKHG2, ANKRD28, IL1RN and RNF19B.

Introduction

Osteoporosis is the most common and serious skeletal disorder among the elderly. Symptomatic osteoporosis occurs due to a decreased bone mineral density (BMD) leading to reduced bone strength and an increased risk of fractures (1). Low bone mass in the elderly is highly dependent on their peak bone mass (PBM) as young adults (2). Therefore, it is necessary to understand and identify the risk factors for impaired PBM in young and middle-aged adults.

Osteopenia may result from an imbalance between increased bone resorption and decreased bone formation (3,4). Bone resorption involves the dissolution of bone mineral and degradation of the organic bone matrix. These two functions are performed by osteoclasts. Osteoclasts are members of the monocyte/macrophage lineage and are formed by multiple instances of cellular fusion of their mononuclear precursors (5). Monocytes differentiate into osteoclasts in the presence of various molecular signals (6). RANKL, one of the most frequently studied, is a ligand for the receptor activator of nuclear factor-κB (NF-κB; RANK) on osteoclast precursor cells (7). RANKL/RANK signaling activates four pathways that mediate osteoclast formation; NF-κB, c-fos and calcineurin/NFATc1 and three pathways that mediate osteoclast activation; Src and MKK6/p38/MITF and survival; Src and extracellular signal-regulated kinase (8). Osteoblasts produce and secrete osteoprotegerin, a decoy receptor that binds to RANKL and blocks RANKL/RANK interactions and hence suppresses the ability of RANK to increase bone resorption (9). Previous studies have shown that blood monocytes also produce a wide variety of inflammatory factors and transcription factors involved in bone metabolism, including interleukin-1 (10), tumor necrosis factor-α (TNF-α) (11), interleukin-6 (12), platelet-derived growth factor (13), transforming growth factor-β (14), resolvinE1 (15), runt-related transcription factor 2 (Runx2; 16), guanylate binding protein 1 (GBP1), signal transducer and activator of transcription 1 (STAT1), CXC chemokine ligand 10 (CXCL10) (17), chemokine receptor 3, histidine decarboxylase and glucocorticoid receptor genes (18).

However, it is unknown whether other mechanisms regulating these factors are significant in the ability of monocytes to affect bone metabolism. Since biological processes are mediated by multiple, co-regulated genes working in synchrony, certain unknown genes may be assigned potential biological functions when studied in gene sets with known genes and ontology groups (19). Thus, the objective of this study was to screen the differential gene expression in monocytes using a high-throughput microarray platform and to explore gene expression profiles comprehensively by grouping individual differentially expressed genes (DEGs) into gene sets and Gene Ontology (GO) terms. The DEGs between high and low PBM samples were grouped into nine gene sets using the graph-clustering approach. GO term enrichment analysis was applied to identify the relevant molecular functions in response to an impaired PBM. The current study revealed that the DEGs, as precursors of osteoclasts, are functionally involved in the immune response. The stimulus response may contribute to differential osteoclastogenesis, leading to differential PBM levels.

Materials and methods

Affymetrix microarray data

Circulating monocyte affymetrix microarray datasets were accessible from the National Center for Biotechnology Information Gene Expression Omnibus (GEO) data repository (http://www.ncbi.nlm.nih.gov/geo/) using the series accession number GSE7158. Fourteen subjects with extremely high PBM levels and 12 subjects with extremely low PBM levels were selected for DNA microarray experiments. All the recruited volunteers signed an informed consent form prior to entering this project.

Statistical analysis

The limma method (20) was used to identify DEGs. The raw expression datasets from all conditions were normalized using the Robust Multiarray Average (RMA) method with the default settings implemented in Bioconductor and then the linear model was constructed. DEGs with a fold change >1.5 and P<0.05 were selected.

The Pearson correlation coefficient (r) was used to compare the potential correlations between DEGs. Statistical significance was set at r>0.95 and P<0.05. All statistical tests were performed using R language (21).

Network analyses and graph clustering

To identify co-expressed groups, DPClus, a graph clustering algorithm that extracts densely connected nodes as a cluster, was used (22). DPClus is based on the density and periphery tracking of clusters and is freely available from http://kanaya.naist.jp/DPClus/. In the current study, the overlapping mode with the DPClus settings were used. The parameter settings of cluster properties were set; density values were set to 0.5 (23) and minimum cluster size was set to 2.

GO term enrichment analysis

The GO (24) project is a major bioinformatics initiative with the aim of standardizing the representation of genes and gene product attributes across species and databases. The project provides a controlled vocabulary of terms for describing gene product characteristics and gene product annotation data from GO Consortium members, as well as tools to access and process this data.

The DAVID tool (25) was used to identify overrepresented GO terms in biological process. P<0.05 and counts of >2 were set as the threshold for the analysis using the hypergeometric distribution.

Results

Differential gene expression profiling and co-expression network construction

GSE7158 microarray datasets were publicly available from the GEO database. Following microarray analysis, a total of 49 genes were selected as DEGs with a fold change >1.5 and P<0.05. The expression profiling of these 49 DEGs is presented in Fig. 1.

To form the correlations between DEGs, r>0.7 and P<0.05 were selected as the cut-off points. A correlation network was constructed with a total of 159 correlations among 49 DEGs (Fig. 2).

Graph clustering identifies modules significantly enriched for DEGs contained in GO term pathways

At r>0.7, DPClus (22) identified 9 clusters in the correlation network for osteoporosis, ranging in size from 3–14 genes. Clusters 1, 2, 7, 8 and 9 were connected as they shared the same genes. For example, one gene (epithelial stromal interaction 1, EPSTI1) was shared between clusters 1 and 8; three genes (suppressor of cytokine signaling, SOCS3; superoxide dismutase, SOD2 and activating transcription factor 3, ATF3) were shared between cluster 2 and 7 and one gene (adrenomedullin, ADM) was shared between clusters 2 and 9. The higher the number of genes shared, the more connectivity among them (corresponding to the thicker lines; Fig. 3).

To assess the significance of the obtained clusters, the overrepresented GO terms were used. Enrichment analysis was performed using the hypergeometrical distribution to find the significant GO term enrichment pathways. In accordance with the graph clustering results, the genes in clusters 1 and 8 were enriched in similar pathways, including immune responses and circulatory system processes. The genes in clusters 2, 7 and 9 were enriched in similar pathways regulating apoptosis and responding to various stimuli, including insulin, hypoxia, nutrients, drugs, radiation and hormones (Table I). Clusters 2 and 7 had the most similar GO term enrichment pathways. These GO biological processes may be relevant to the differentiation of monocytes into osteoclasts.

Table I

List of enriched GO terms in clusters 1, 2, 7, 8 and 9 detected by DPClus.

Table I

List of enriched GO terms in clusters 1, 2, 7, 8 and 9 detected by DPClus.

CategoryTermDescriptionCountP-valueFDR
Cluster 1GO:0009615Response to virus4 5.86e−50.006833
GO:0006955Immune response50.001100570.062388
GO:0006952Defense response40.008827950.292359
Cluster 2GO:0032868Response to insulin stimulus30.001471490.378515
GO:0007568Aging30.001776870.249653
GO:0001666Response to hypoxia30.002622430.246268
GO:0070482Response to oxygen levels30.002898630.208958
GO:0043434Response to peptide hormone stimulus30.003446540.19991
GO:0006915Apoptosis40.004151340.200641
GO:0012501Programmed cell death40.004329560.181443
GO:0031667Response to nutrient levels30.005576390.202104
GO:0006916Anti-apoptosis30.006082640.196651
GO:0008219Cell death40.006846520.199007
GO:0009991Response to extracellular stimulus30.006910870.184238
GO:0016265Death40.006980580.171843
GO:0042981Regulation of apoptosis40.009347380.208115
GO:0043067Regulation of programmed cell death40.009607510.19967
GO:0010941Regulation of cell death40.009706180.189438
GO:0010332Response to gamma radiation20.013524240.240339
GO:0031100Organ regeneration20.015276410.253599
GO:0048666Neuron development30.015864320.249457
GO:0043066Negative regulation of apoptosis30.017224420.255741
GO:0043069Negative regulation of programmed cell death30.017688780.250411
GO:0060548Negative regulation of cell death30.017782310.241164
GO:0009725Response to hormone stimulus30.018443080.23914
GO:0009628Response to abiotic stimulus30.018538350.231094
GO:0006873Cellular ion homeostasis30.019114450.228747
GO:0055082Cellular chemical homeostasis30.019698290.226664
GO:0009719Response to endogenous stimulus30.02221310.24351
GO:0050801Ion homeostasis30.022627620.239518
GO:0030182Neuron differentiation30.025731120.259709
GO:0019725Cellular homeostasis30.028888010.27855
GO:0048878Chemical homeostasis30.034404230.314043
GO:0010212Response to ionizing radiation20.034944920.309692
GO:0031099Regeneration20.04009340.338357
GO:0032496Response to lipopolysaccharide20.044649630.360509
GO:0009266Response to temperature stimulus20.048054370.373652
GO:0002237Response to molecule of bacterial origin20.049752760.375599
Cluster 7GO:0070482Response to oxygen levels3 3.21e−40.079885
GO:0009314Response to radiation3 6.46e−40.080292
GO:0042493Response to drug3 7.53e−40.062989
GO:0009991Response to extracellular stimulus3 7.81e−40.049354
GO:0055093Response to hyperoxia20.001773180.087833
GO:0043066Negative regulation of apoptosis30.002013090.08331
GO:0043069Negative regulation of programmed cell death30.002069920.073801
GO:0060548Negative regulation of cell death30.002081380.06523
GO:0009628Response to abiotic stimulus30.002174170.060715
GO:0010332Response to gamma radiation20.005092240.123857
GO:0031100Organ regeneration20.005755170.12707
GO:0048145Regulation of fibroblast proliferation20.007742190.154437
GO:0042127Regulation of cell proliferation30.00974870.177311
GO:0042981Regulation of apoptosis30.010165810.172238
GO:0043067Regulation of programmed cell death30.010364990.164649
GO:0010941Regulation of cell death30.010440160.156241
GO:0010212Response to ionizing radiation20.013247780.183871
GO:0031099Regeneration20.01522480.198085
GO:0007568Aging20.024197810.283882
GO:0014070Response to organic cyclic substance20.026595890.294663
GO:0001666Response to hypoxia20.029424920.308128
GO:0048545Response to steroid hormone stimulus20.041979970.396429
GO:0031667Response to nutrient levels20.04305720.390801
GO:0010035Response to inorganic substance20.04477910.39006
GO:0006916Anti-apoptosis20.044994190.379328
Cluster 8GO:0008015Blood circulation20.013749260.839184
GO:0003013Circulatory system process20.013749260.839184
Cluster 9GO:0051384Response to glucocorticoid stimulus20.011498820.709892
GO:0031960Response to corticosteroid stimulus20.012527510.490566
GO:0048545Response to steroid hormone stimulus20.028185170.6393

[i] GO, gene ontology; FDR, false discovery rate.

Discussion

In the current study, differential expression profiling was systematically investigated and its possible role in the differentiation of osteoclasts was explored. A total of 49 DEGs were identified and correlated to produce 159 network connections. These DEGs were assigned into nine clusters using the graph clustering method in response to different PBM levels. A total of 14 genes were included in cluster 1 [GBP1; interferon-induced protein with tetratricopeptide repeats 2, IFIT2; eukaryotic translation initiation factor 2-α kinase 2, EIF2AK2; interferon-induced protein 44, IFI44; IFI44L; DEAD (Asp-Glu-Ala-Asp) box polypeptide 60, DDX60; HECT and RLD domain containing E3 ubiquitin protein ligase 5, HERC5; radical S-adenosyl methionine domain containing 2, RSAD2; sialic acid binding Ig-like lectin 1, sialoadhesin, SIGLEC1; cytidine monophosphate kinase 2, CMPK2; EPSTI1; interferon, α-inducible protein 6, IFI6; CXCL10; and myxovirus resistance 1, interferon-inducible protein p78, MX1] and 3 genes were involved in cluster 8 (STAT1; EPSTI1; and serpin peptidase inhibitor, clade G, SERPING1). Notably, cluster 8 was connected with all the genes of cluster 1 by STAT1 and EPSTI1 in order to be involved in immune responses and circulatory system processes, as demonstrated in previous studies.

The immune system has been correlated with bone resorption through a complex interaction involving T and B lymphocytes, dendritic cells (DCs), cytokines and cell-cell interactions (26). There is strong evidence that STAT1 is significant in bone metabolism as STAT1 has been reported to be upregulated in the femur tissue of osteoporotic mice (27) and humans (18). STAT1 may serve as a primary mediator of interferon (IFN) signaling pathways involving osteoclast differentiation. Through the p38 MAPK pathway, RANKL stimulates the serine phosphorylation of STAT1, resulting in the migration and adhesion of osteoclast precursors (28). STAT1 interacts with Runx2, an essential transcription factor for osteoblast differentiation, in its latent form in the cytoplasm, thereby inhibiting the nuclear localization of Runx2. This function of STAT1 does not require the Tyr 701 that is phosphorylated when STAT1 becomes a transcriptional activator (29).

The GBP1 gene is also predicted to be involved in bone metabolism or osteoclast differentiation (30) in a STAT1-dependent manner (31). The sumoylation-defective STAT1 mutant exhibits increased induction of GBP1 and transporters associated with antigen presentation 1 (TAP1) transcription (32). The mutation in the STAT1 gene dramatically reduces the inducibility of the GBP1 and TAP1 genes by IFN (33). In this study, STAT1 and GBP1 directly interacted with each other (Figs. 2 and 3).

Chemokines have a potential role in the regulation of osteoclast functions. For example, IFN-γ-inducible protein-10 (CXCL10) is expressed in human osteoclasts with changing expression levels during osteoclast differentiation (34). CXCL10 has been suggested to contribute to osteoclastogenesis by increasing RANKL expression in CD4+ T cells in an animal model of rheumatoid arthritis (35). Notably, previous studies have shown that osteoblasts secrete IFN-β in response to viral infections and that endogenous IFN-β induces CXCL10 and IFI44L production via an IFN-α/β receptor-STAT1 pathway (36,37).

EIF2AK2 is also reported to interact with STAT1 and increase its degradation. Reduction of EIF2AK2 activity also reduces RUNX2 activity and murine osteoblast differentiation (38,39). Therefore, it appears illogical that EIF2AK2 is upregulated in human osteoblasts following IFN-β treatment which results in an inhibition of mineralization (40).

Ten genes were included in cluster 2 [ADM; early growth response 2, EGR2; BCL2-related protein A1, BCL2A1; chromosome 9 open reading frame 72, C9orf72; pleckstrin homology-like domain, family A, member 2, PHLDA2; ATF3; SOCS3; SOD2; 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, PFKFB3; and pleckstrin homology domain containing, family G (with RhoGef domain) member 2, PLEKHG2], five genes were included in cluster 7 (SOCS3; SOD2; ATF3; cyclin-dependent kinase inhibitor 1A, CDKN1A; and ankyrin repeat domain 28, ANKRD28) and three genes were included in cluster 9 (ADM; interleukin 1 receptor antagonist, IL1RN; and ring finger protein 19B, RNF19B). Cluster 7 was connected with all the genes of cluster 2 and 9 by SOCS3, SOD2 and ATF3. Cluster 9 was connected with all the genes of clusters 2 and 7 by ADM and IL1RN. These findings indicate that SOCS3, SOD2, ATF3 and ADM are significant genes for responding to various stimuli, including insulin, hypoxia, nutrients, drugs, radiation and hormones regulating apoptosis.

The SOCS3 family are cytoplasmic adaptor proteins that negatively regulate various cytokine responses in leukocytes. SOCS3 overexpression augments TGF-β, TNF-α and RANKL-induced osteoclast formation, priming precursors to the osteoclast lineage by suppressing specific anti-osteoclastic JAK/STAT signals (41). Zhang et al demonstrated that a higher SOCS3 expression level is associated with RANKL-mediated alveolar bone loss and enhances CD11c+ DC-derived osteoclastogenesis in vivo and in vitro. The reduced expression of functional SOCS3 in CD11c+ DCs results in significantly lower osteoclastogenesis and dendritic cell-derived osteoclasts development during immune interactions with T cells, based on TRAP expression and bone resorptive activity (42). In SOCS3-deficient bone marrow-derived monocytes, the expression levels of TNF-receptor-associated factor-6 and IκB are drastically reduced. The receptor activation of NF-κB ligand-induced IκB phosphorylation is severely impaired, indicating that SOCS3 regulates osteoclastogenesis by blocking the inhibitory effect of inflammatory cytokines on receptor activation of the NF-κB ligand-mediated osteoclast differentiation signals (43).

ADM is a 52-amino acid peptide first described in a human phaeochromocytoma but has since been identified in numerous tissues, including the bone (44). Systemic administration of ADM stimulates the proliferation of osteoblasts and promotes bone growth (45). Treatment with ADM significantly blunts the apoptosis of serum-deprived osteoblastic cells, evaluated by caspase-3 activity, DNA fragmentation quantification and Annexin V-FITC labeling. This effect is eliminated by calcitonin-related polypeptide α (CGRP1) and insulin-like growth factor-I (46). The selective inhibitor of MAPK kinase (MEK), PD98059, also eliminates the protective effect of ADM on apoptosis and prevents ADM activation of ERK1/2. These data show that ADM acts as a survival factor in osteoblastic cells via a CGRP1 receptor-MEK-ERK pathway, which provides further understanding on the physiological function of ADM in osteoblasts (47).

The SOD2 gene encodes a free radical-scavenging enzyme that removes superoxidate and catalyzes the production of hydrogen peroxide. Oxidative stress is significant in the pathogenesis of osteoporosis (48). Previous studies have revealed that SOD2 is significantly upregulated in circulating monocytes at the mRNA and protein level in vivo in Chinese patients with low versus high hip BMD levels (49). Women with postmenopausal osteoporosis have significantly higher plasma SOD enzyme activity levels than those in controls (50). This indicates that SOD2 is significant in the pathogenesis of osteoporosis, promoting osteoclast differentiation, formation and activity (51).

EGR2 is a highly conserved transcription factor involved in bone remodeling. The upregulation of EGR2 is involved in the biological affinity of titanium for osteogenic cells and in the promotion of osteoblast differentiation (52). Macrophage colony-stimulating factor activates MEK/ERK and induces the MEK-dependent expression of the immediate early gene EGR2. Inhibition of either MEK1/2 or EGR2 increases osteoclast apoptosis (53). Previous studies have revealed a novel role for EGR2 in postnatal skeletal metabolism. EGR2+/− mice reveal a low bone mass phenotype. EGR2 silencing in pre-osteoclasts increases the expression of cFms and the response to macrophage colony-stimulating factor, leading to a cell-autonomous stimulation of cell-cycle progression. Thus, the anti-mitogenic role of EGR2 in pre-osteoclasts is the predominant mechanism underlying the low bone mass phenotype of EGR2-deficient mice (54).

The osteoporotic state increases ATF3 expression in dorsal root ganglia neurons innervating L3 vertebrae (55). BCL2A1, an anti-apoptotic activated macrophage protein, is also heavily overexpressed in osteolysis patients, providing a possible mechanism for the persistence of the particle-laden cells expressing macrophage phenotype activation markers (56).

In conclusion, the present findings shed new light on the biology of osteoporosis and have implications for future research. The changes in the immune system (GBP1, STAT1, CXCL10 and EIF2AK2) and stimulus response (SOCS3, SOD2, ATF3, ADM EGR2 and BCL2A1) may be associated with osteoclast differentiation. This study provides a number of candidate genes that warrant further investigation, including DDX60, HERC5, RSAD2, SIGLEC1, CMPK2, MX1, SERPING1, EPSTI1, C9orf72, PHLDA2, PFKFB3, PLEKHG2, ANKRD28, IL1RN and RNF19B.

References

1 

Chao TH, Yu HN, Huang CC, Liu WS, Tsai YW and Wu WT: Association of interleukin-1β (-511C/T) polymorphisms with osteoporosis in postmenopausal women. Ann Saudi Med. 30:437–441. 2010.

2 

Lapauw BM, Taes Y, Bogaert V, Vanbillemont G, Goemaere S, Zmierczak HG, De Bacquer D and Kaufman JM: Serum estradiol is associated with volumetric BMD and modulates the impact of physical activity on bone size at the age of peak bone mass: a study in healthy male siblings. J Bone Miner Res. 24:1075–1085. 2009.PubMed/NCBI

3 

Andersen TL, Sondergaard TE, Skorzynska KE, Dagnaes-Hansen F, Plesner TL, Hauge EM, Plesner T and Delaisse JM: A physical mechanism for coupling bone resorption and formation in adult human bone. Am J Pathol. 174:239–247. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Marie PJ: The calcium-sensing receptor in bone cells: a potential therapeutic target in osteoporosis. Bone. 46:571–576. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Quinn JM, Neale S, Fujikawa Y, McGee JO and Athanasou NA: Human osteoclast formation from blood monocytes, peritoneal macrophages and bone marrow cells. Calcif Tissue Int. 62:527–531. 1998. View Article : Google Scholar : PubMed/NCBI

6 

Kim HJ, Minashima T, McCarthy EF, Winkles JA and Kirsch T: Progressive ankylosis protein (ANK) in osteoblasts and osteoclasts controls bone formation and bone remodeling. J Bone Miner Res. 25:1771–1783. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Wada T, Nakashima T, Hiroshi N and Penninger JM: RANKL-RANK signaling in osteoclastogenesis and bone disease. Trends Mol Med. 12:17–25. 2006. View Article : Google Scholar

8 

Kobayashi Y, Udagawa N and Takahashi N: Action of RANKL and OPG for osteoclastogenesis. Crit Rev Eukaryot Gene Expr. 19:61–72. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Boyce BF and Xing L: Functions of RANKL/RANK/OPG in bone modeling and remodeling. Arch Biochem Biophys. 473:139–146. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Yao Z, Xing L, Qin C, Schwarz EM and Boyce BF: Osteoclast precursor interaction with bone matrix induces osteoclast formation directly by an interleukin-1-mediated autocrine mechanism. J Biol Chem. 283:9917–9924. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Goto H, Hozumi A, Osaki M, Fukushima T, Sakamoto K, Yonekura A, Tomita M, Furukawa K, Shindo H and Baba H: Primary human bone marrow adipocytes support TNFα-induced osteoclast differentiation and function through RANKL expression. Cytokine. 56:662–668. 2011.

12 

Axmann R, Böhm C, Krönke G, Zwerina J, Smolen J and Schett G: Inhibition of interleukin-6 receptor directly blocks osteoclast formation in vitro and in vivo. Arthritis Rheum. 60:2747–2756. 2009. View Article : Google Scholar : PubMed/NCBI

13 

McCarthy HS, Williams JH, Davie MW and Marshall MJ: Platelet-derived growth factor stimulates osteoprotegerin production in osteoblastic cells. J Cell Physiol. 218:350–354. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Futakuchi M, Nannuru KC, Varney ML, Sadanandam A, Nakao K, Asai K, Shirai T, Sato S and Singh RK: Transforming growth factor-β signaling at the tumor-bone interface promotes mammary tumor growth and osteoclast activation. Cancer Sci. 100:71–81. 2009.

15 

Herrera BS, Ohira T, Gao L, Omori K, Yang R, Zhu M, Muscara MN, Serhan CN, Van Dyke TE and Gyurko R: An endogenous regulator of inflammation, resolvin E1, modulates osteoclast differentiation and bone resorption. Br J Pharmacol. 155:1214–1223. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR and de Crombrugghe B: The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 108:17–29. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Lei SF, Wu S, Li LM, Deng FY, Xiao SM, Jiang C, Chen Y, Jiang H, Yang F, Tan LJ, et al: An in vivo genome wide gene expression study of circulating monocytes suggested GBP1, STAT1 and CXCL10 as novel risk genes for the differentiation of peak bone mass. Bone. 44:1010–1014. 2009.

18 

Chen XD, Xiao P, Lei SF, Liu YZ, Guo YF, Deng FY, Tan LJ, Zhu XZ, Chen FR, Recker RR and Deng HW: Gene expression profiling in monocytes and SNP association suggest the importance of the STAT1 gene for osteoporosis in both Chinese and Caucasians. J Bone Miner Res. 25:339–355. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Horan K, Jang C, Bailey-Serres J, Mittler R, Shelton C, Harper JF, Zhu JK, Cushman JC, Gollery M and Girke T: Annotating genes of known and unknown function by large-scale coexpression analysis. Plant Physiol. 147:41–57. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:Article 3. 2004.PubMed/NCBI

21 

Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, et al: Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5:R802004. View Article : Google Scholar : PubMed/NCBI

22 

Altaf-Ul-Amin M, Shinbo Y, Mihara K, Kurokawa K and Kanaya S: Development and implementation of an algorithm for detection of protein complexes in large interaction networks. BMC Bioinformatics. 7:2072006. View Article : Google Scholar : PubMed/NCBI

23 

Fukushima A, Kusano M, Redestig H, Arita M and Saito K: Metabolomic correlation-network modules in Arabidopsis based on a graph-clustering approach. BMC Syst Biol. 5:12011. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009.PubMed/NCBI

26 

Clowes JA, Riggs BL and Khosla S: The role of the immune system in the pathophysiology of osteoporosis. Immunol Rev. 208:207–227. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Orlić I, Borovecki F, Simić P and Vukicević S: Gene expression profiling in bone tissue of osteoporotic mice. J Ind Hyg Toxicol. 58:3–11. 2007.PubMed/NCBI

28 

Kwak HB, Lee SW, Jin HM, Ha H, Lee SH, Takeshita S, Tanaka S, Kim HM, Kim HH and Lee ZH: Monokine induced by interferon-γ is induced by receptor activator of nuclear factor κB ligand and is involved in osteoclast adhesion and migration. Blood. 105:2963–2969. 2005.

29 

Kim S, Koga T, Isobe M, Kern BE, Yokochi T, Chin YE, Karsenty G, Taniguchi T and Takayanagi H: Stat1 functions as a cytoplasmic attenuator of Runx2 in the transcriptional program of osteoblast differentiation. Genes Dev. 17:1979–1991. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Lei SF, Wu S, Li LM, Deng FY, Xiao SM, Jiang C, Chen Y, Jiang H, Yang F and Tan LJ: An in vivo genome wide gene expression study of circulating monocytes suggested GBP1, STAT1 and CXCL10 as novel risk genes for the differentiation of peak bone mass. Bone. 44:1010–1014. 2009.

31 

Cheng YS, Patterson CE and Staeheli P: Interferon-induced guanylate-binding proteins lack an N (T) KXD consensus motif and bind GMP in addition to GDP and GTP. Mol Cell Biol. 11:4717–4725. 1991.PubMed/NCBI

32 

Ungureanu D, Vanhatupa S, Grönholm J, Palvimo JJ and Silvennoinen O: SUMO-1 conjugation selectively modulates STAT1-mediated gene responses. Blood. 106:224–226. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Kovarik P, Mangold M, Ramsauer K, Heidari H, Steinborn R, Zotter A, Levy DE, Müller M and Decker T: Specificity of signaling by STAT1 depends on SH2 and C-terminal domains that regulate Ser727 phosphorylation, differentially affecting specific target gene expression. EMBO J. 20:91–100. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Grassi F, Piacentini A, Cristino S, Toneguzzi S, Cavallo C, Facchini A and Lisignoli G: Human osteoclasts express different CXC chemokines depending on cell culture substrate: molecular and immunocytochemical evidence of high levels of CXCL10 and CXCL12. Histochem Cell Biol. 120:391–400. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Lee EY, Seo M, Juhnn YS, Kim JY, Hong YJ, Lee YJ, Lee EB and Song YW: Potential role and mechanism of IFN-γ-inducible protein-10 on receptor activator of nuclear factor kappa-B ligand (RANKL) expression in rheumatoid arthritis. Arthritis Res Ther. 13:R1042011.

36 

Nakamura K, Deyama Y, Yoshimura Y, Suzuki K and Morita M: Toll-like receptor 3 ligand-induced antiviral response in mouse osteoblastic cells. Int J Mol Med. 19:771–775. 2007.PubMed/NCBI

37 

Woeckel V, Eijken M, van de Peppel J, Chiba H, van der Eerden B and van Leeuwen J: IFNβ impairs extracellular matrix formation leading to inhibition of mineralization by effects in the early stage of human osteoblast differentiation. J Cell Physiol. 227:2668–2676. 2011.

38 

Yoshida K, Okamura H, Amorim BR, Hinode D, Yoshida H and Haneji T: PKR-mediated degradation of STAT1 regulates osteoblast differentiation. Exp Cell Res. 315:2105–2114. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Yoshida K, Okamura H, Amorim BR, Ozaki A, Tanaka H, Morimoto H and Haneji T: Double-stranded RNA-dependent protein kinase is required for bone calcification in MC3T3-E1 cells in vitro. Exp Cell Res. 311:117–125. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Woeckel VS, Koedam M, van de Peppel J, Chiba H, van der Eerden BC and van Leeuwen JP: Evidence of Vitamin D and interferon-β cross-talk in human osteoblasts with 1α, 25-dihydroxyvitamin D3 being dominant over interferon-β in stimulating mineralization. J Cell Physiol. 227:3258–3266. 2011.

41 

Lovibond AC, Haque SJ, Chambers TJ and Fox SW: TGFβ-induced SOCS3 expression augments TNFα-induced osteoclast formation. Biochem Biophys Res Commun. 309:762–767. 2003.

42 

Zhang X, Alnaeeli M, Singh B and Teng YT: Involvement of SOCS3 in regulation of CD11c+ dendritic cell-derived osteoclastogenesis and severe alveolar bone loss. Infect Immun. 77:2000–2009. 2009.PubMed/NCBI

43 

Yoshimura A, Sanada T, Takaki H and Ohishi M: Suppressor of cytokine signalling-1 (SOCS1) and SOCS3 regulate osteoclastogenesis by blocking inflammatory cytokine signals. Nihon Riumachi Gakkai, Gakujutsu Shukai, Kokusai Riumachi Simpojiumu Puroguramu, Shorokushu. 50:152006.PubMed/NCBI

44 

Cornish J, Callon KE, Bava U, Coy DH, Mulvey TB, Murray MA, Cooper GJ and Reid IR: Systemic administration of adrenomedullin (27–52) increases bone volume and strength in male mice. J Endocrinol. 170:251–257. 2001.

45 

Cornish J, Callon KE, Coy DH, Jiang NY, Xiao L, Cooper GJ and Reid IR: Adrenomedullin is a potent stimulator of osteoblastic activity in vitro and in vivo. Am J Physiol. 273:E1113–1120. 1997.PubMed/NCBI

46 

Cornish J, Grey A, Callon KE, Naot D, Hill BL, Lin CQ, Balchin LM and Reid IR: Shared pathways of osteoblast mitogenesis induced by amylin, adrenomedullin and IGF-1. Biochem Biophys Res Commun. 318:240–246. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Uzan B, Villemin A, Garel JM and Cressent M: Adrenomedullin is anti-apoptotic in osteoblasts through CGRP1 receptors and MEK-ERK pathway. J Cell Physiol. 215:122–128. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Abdollahi M, Larijani B, Rahimi R and Salari P: Role of oxidative stress in osteoporosis. Therapy. 2:787–796. 2005. View Article : Google Scholar

49 

Deng FY, Lei SF, Chen XD, Tan LJ, Zhu XZ and Deng HW: An integrative study ascertained SOD2 as a susceptibility gene for osteoporosis in Chinese. J Bone Miner Res. 26:2695–2701. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Ozgocmen S, Kaya H, Fadillioglu E, Aydogan R and Yilmaz Z: Role of antioxidant systems, lipid peroxidation and nitric oxide in postmenopausal osteoporosis. Mol Cell Biochem. 295:45–52. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Deng FY, Liu YZ, Li LM, Jiang C, Wu S, Chen Y, Jiang H, Yang F, Xiong JX and Xiao P: Proteomic analysis of circulating monocytes in Chinese premenopausal females with extremely discordant bone mineral density. Proteomics. 8:4259–4272. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Levi G, Topilko P, Schneider-Maunoury S, Lasagna M, Mantero S, Pesce B, Ghersi G, Cancedda R and Charnay P: Role of Krox-20 in endochondral bone formationa. Ann NY Acad Sci. 785:288–291. 1996. View Article : Google Scholar : PubMed/NCBI

53 

Bradley EW, Ruan MM and Oursler MJ: Novel pro-survival functions of the Kruppel-like transcription factor EGR2 in promotion of M-CSF-mediated osteoclast survival downstream of the MEK/ERK pathway. J Biol Chem. 283:8055–8064. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Gabet Y, Baniwal SK, Leclerc N, Shi Y, Kohn-Gabet AE, Cogan J, Dixon A, Bachar M, Guo L, Turman JE Jr and Frenkel B: Krox20/EGR2 deficiency accelerates cell growth and differentiation in the monocytic lineage and decreases bone mass. Blood. 116:3964–3971. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Suzuki M, Ohtori S, Inoue G, Orita S, Ishikawa T, Miyagi M, Kamoda H, Eguchi Y, Arai G, et al: ATF3 and GAP43 immunoreactive DRG neurons innervate the vertebral body in a rat model of osteoporosis: GP18. Spine. 2011.

56 

Purdue PE: Alternative macrophage activation in periprosthetic osteolysis. Autoimmunity. 41:212–217. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December 2012
Volume 6 Issue 6

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
Xiao H, Shan L, Zhu H and Xue F: Detection of significant pathways in osteoporosis based on graph clustering. Mol Med Rep 6: 1325-1332, 2012
APA
Xiao, H., Shan, L., Zhu, H., & Xue, F. (2012). Detection of significant pathways in osteoporosis based on graph clustering. Molecular Medicine Reports, 6, 1325-1332. https://doi.org/10.3892/mmr.2012.1082
MLA
Xiao, H., Shan, L., Zhu, H., Xue, F."Detection of significant pathways in osteoporosis based on graph clustering". Molecular Medicine Reports 6.6 (2012): 1325-1332.
Chicago
Xiao, H., Shan, L., Zhu, H., Xue, F."Detection of significant pathways in osteoporosis based on graph clustering". Molecular Medicine Reports 6, no. 6 (2012): 1325-1332. https://doi.org/10.3892/mmr.2012.1082