Skip to main content
Erschienen in: Arthritis Research & Therapy 5/2007

Open Access 01.10.2007 | Research article

Microarray gene expression profiling of osteoarthritic bone suggests altered bone remodelling, WNT and transforming growth factor-β/bone morphogenic protein signalling

verfasst von: Blair Hopwood, Anna Tsykin, David M Findlay, Nicola L Fazzalari

Erschienen in: Arthritis Research & Therapy | Ausgabe 5/2007

Abstract

Osteoarthritis (OA) is characterized by alterations to subchondral bone as well as articular cartilage. Changes to bone in OA have also been identified at sites distal to the affected joint, which include increased bone volume fraction and reduced bone mineralization. Altered bone remodelling has been proposed to underlie these bone changes in OA. To investigate the molecular basis for these changes, we performed microarray gene expression profiling of bone obtained at autopsy from individuals with no evidence of joint disease (control) and from individuals undergoing joint replacement surgery for either degenerative hip OA, or fractured neck of femur (osteoporosis [OP]). The OP sample set was included because an inverse association, with respect to bone density, has been observed between OA and the low bone density disease OP. Compugen human 19K-oligo microarray slides were used to compare the gene expression profiles of OA, control and OP bone samples. Four sets of samples were analyzed, comprising 10 OA-control female, 10 OA-control male, 10 OA-OP female and 9 OP-control female sample pairs. Print tip Lowess normalization and Bayesian statistical analyses were carried out using linear models for microarray analysis, which identified 150 differentially expressed genes in OA bone with t scores above 4. Twenty-five of these genes were then confirmed to be differentially expressed (P < 0.01) by real-time PCR analysis. A substantial number of the top-ranking differentially expressed genes identified in OA bone are known to play roles in osteoblasts, osteocytes and osteoclasts. Many of these genes are targets of either the WNT (wingless MMTV integration) signalling pathway (TWIST1, IBSP, S100A4, MMP25, RUNX2 and CD14) or the transforming growth factor (TGF)-β/bone morphogenic protein (BMP) signalling pathway (ADAMTS4, ADM, MEPE, GADD45B, COL4A1 and FST). Other differentially expressed genes included WNT (WNT5B, NHERF1, CTNNB1 and PTEN) and TGF-β/BMP (TGFB1, SMAD3, BMP5 and INHBA) signalling pathway component or modulating genes. In addition a subset of genes involved in osteoclast function (GSN, PTK9, VCAM1, ITGB2, ANXA2, GRN, PDE4A and FOXP1) was identified as being differentially expressed in OA bone between females and males. Altered expression of these sets of genes suggests altered bone remodelling and may in part explain the sex disparity observed in OA.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​ar2301) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

NLF and DMF conceived the study. BH contributed to study design and performed the acquisition of the microarray and real-time PCR data. AT advised on the microarray experiment design and performed the statistical analyses of the microarray data. BH and AT analyzed and interpreted the data. BH, DMF and NLF prepared the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AMF
= Adelaide Microarray facility
BMP
= bone morphogenic protein
CT
= cycle threshold
IL
= interleukin
IT
= intertrochanteric
LEF
= lymphoid enhancer factor
LIMMA
= linear models for microarray analysis
MMP
= matrix metalloproteinase
OA
= osteoarthritis
OP
= osteoporosis
PCR
= polymerase chain reaction
RUNX
= runt-related transcription factor
SD
= standard deviation
TCF
= T-cell factor
TGF
= transforming growth factor
WNT
= wingless MMTV integration.

Introduction

Osteoarthritis (OA) is a complex, multifactorial, age-dependent degenerative disease of the synovial joints. It affects the knee and the hip most commonly, and females at a higher rate than males, particularly after the menopause [1]. OA is characterized by changes to all components of the joint, with degeneration and loss of articular cartilage and changes to the subchondral bone being constant factors in disease progression [2]. Along with the breakdown of the cartilage and joint space narrowing, there is thickening and sclerosis of subchondral bone, development of cysts and bony overgrowth at the margins of the joint. Despite an increase in bone volume fraction, the subchondral bone is mechanically weaker in OA because of hypomineralization, increased collagen metabolism and altered bone remodelling [3, 4]. Evidence from animal models of OA suggests that the changes in the density and metabolism of subchondral bone develop concomitantly with the signs of cartilage damage [57]. In addition, there is now evidence in animal OA models that antiresorptive agents, which inhibit subchondral bone remodelling, also prevent the bone changes and loss of cartilage seen in OA, thus reducing joint damage [8, 9]. A human trial of an antiresorptive agent also showed clear trends toward improvement in both joint structure and symptoms in patients with primary knee OA [10]. These findings are consistent with the hypothesis that OA is a bone disease, rather than – or in addition to – a cartilage disease, and that the structural and compositional changes seen in OA subchondral bone, brought about by altered bone remodelling, contribute to the breakdown of the articular cartilage at the joint [1114].
There is also evidence that the osteoblasts in subchondral bone can influence chondrocyte and cartilage metabolism more directly, leading to abnormal remodelling of OA cartilage [15, 16]. In articular joints there is a complex juxtaposition of vascular elements, subchondral bone and the different cartilage layers, with important communication between these tissues [17]. These observations point to a clear interplay between bone and cartilage at articular joints and show that these tissues represent a functional cellular and molecular unit [18]. Altered angiogenesis could also be contributing to the changes seen in OA bone and cartilage, because important inter-relationships between bone remodelling, chondrogenic and angiogenic processes are now emerging [1921].
In addition to the changes observed in subchondral bone, there is growing evidence for generalized involvement of bone in the pathogenesis of OA. Studies investigating bone at sites distal to the joint cartilage degeneration, such as the intertrochanteric (IT) and medial principal compressive regions of the proximal femur, and the iliac crest, have yielded evidence of altered bone composition and increased bone volume in OA compared with control individuals [2225]. It has been proposed that these structural and compositional changes reflect systemic differences in OA bone remodelling compared with control bone, and when these changes operate in subchondral bone they can contribute to the breakdown of the articular cartilage and eventual failure of the joint [1114]. Furthermore, an inverse association between OA and the low bone density disease osteoporosis (OP) has been observed. OA patients rarely proceed to osteoporotic fracture, suggesting that OA has a protective effect on progression of OP. Conversely, OA is reported to be rare in OP individuals [26].
The structural and compositional changes seen in OA bone are likely to have considerable genetic input because there is a significant heritable component to OA, as judged by genetic studies [27]. Interestingly, many of the candidate susceptibility genes for OA identified by genetic screening approaches have bone-related functions, further suggesting the involvement of bone in OA. Primary OA candidate genes identified, with bone-related functions, include COL1A1, VDR, ESR1, IGF1, SFRP3, BMP5 and TGFB1 [2730]. SFRP3 encodes a decoy receptor for WNT (wingless MMTV integration) ligands and plays a role in osteoblast differentiation [31]. The WNT signalling pathway is a major developmental pathway that is involved in cell fate, differentiation and proliferation. This signalling pathway has also been linked to skeletal development and bone pathologies such as OP [32]. The identification of TGFB1 and BMP5, a member of the transforming growth factor (TGF)-β superfamily, as OA susceptibility loci has implicated the TGF-β/BMP signalling pathway in OA pathogenesis. The TGF-β/BMP signalling pathway plays important roles in development, cell proliferation and differentiation, and it has also been shown to influence bone mass and bone remodelling [33, 34].
Complementing the human genetic studies described above, and in support of altered bone remodelling at sites distal to the active subchondral disease site, we previously identified differences in the expression of known skeletally active genes in human trabecular bone obtained from the IT region from individuals with hip OA, as compared with bone from the same site in control individuals. Genes identified as differentially expressed include downregulated osteoclastogenic factor genes (RANKL, RANK, IL6 and IL11) and upregulated bone formation marker genes (ALPL, BGLAP, SPP1 and COL1A2) [3537]. Others have identified in OA individuals altered levels of insulin-like growth factor-1, insulin-like growth factor-2 and TGF-β1 in cortical bone from the iliac crest [38]; matrix metalloproteinase (MMP)2 and liver alkaline phosphatase in subchondral bone [4]; and IL-1β, IL-6 and TGF-β1 in human primary subchondral osteoblasts [39].
In the present study, we used microarray analysis to survey comprehensively the expression levels of many thousands of genes simultaneously in trabecular bone from the IT region of the proximal femur and to compare gene expression in bone from OA, control and OP individuals. We identified altered expression of WNT and TGF-β/BMP signalling pathway and target genes in OA bone. The genes include those with known or suspected roles in osteoblast, osteocyte and osteoclast differentiation and function, supporting a role for altered bone remodelling in OA pathogenesis.

Materials and methods

Human bone samples

For the OA and OP groups, tube saw bone biopsies (10 mm diameter and 20 to 40 mm long) were obtained from the IT region of the proximal femur. These were obtained from 24 patients (14 females [age range 49 to 83 years] and 10 males [50 to 85 years]) undergoing hip arthroplasty for primary OA and from 10 patients (10 females [74 to 87 years]) undergoing hip arthroplasty for a fractured neck of femur (designated OP). For the control group, trabecular bone from the IT region was obtained during 21 autopsies (11 females [43 to 85 years] and 10 males [50 to 85 years]) of individuals who were known not to have suffered from any chronic condition or disease that may have affected the skeleton. In selecting the OA, OP and control individuals, those with a known history of medication that might have affected bone metabolism were excluded. Informed consent was obtained for the collection of these specimens, with approval from the Royal Adelaide Hospital Research Ethics Committee (protocol number 030309).
The surgical and autopsy femoral heads were graded for OA according to the criteria of Collins [40]. Primary OA femoral heads were either grade III or IV, and the graded autopsy femoral heads were not worse than grade II and predominantly were grade I. Surgical IT trabecular bone specimens from OA and OP individuals were collected within 12 to 24 hours (stored at 4°C in sterile RNase-free phosphate-buffered saline). Control bone was collected within 24 to 72 hours after death.
Trabecular bone in the IT region of the proximal femur, including the marrow, was sampled, permitting analysis of the total contribution of the bone microenvironment. The IT region was also chosen because the trabecular structure in this region depends on stresses in the proximal femoral shaft, while being unaffected by the secondary sclerotic and cystic changes that are often seen in the OA femoral head as the destruction of the cartilage proceeds. By comparing the OA and OP samples with control samples, the contribution to changes in gene expression associated with surgery as opposed to autopsy could be assessed.

RNA extraction

For total RNA extraction, the trabecular bone samples were rinsed briefly in diethylpyrocarbonate-treated water and then separated into small fragments, containing bone and bone marrow, using bone cutters. Total RNA was extracted as described previously [35, 41]. Briefly, bone fragments were placed in 4 mol/l guanidinium thiocyanate solution and homogenized using an Ultra-Turrax (TP 18–10; Janke & Kunkel, IKA-WERK, Staufen, Germany), and the mixture was clarified by centrifugation (1,000 × g for 5 min). After addition of 0.1 vol of 2 mol/l sodium acetate (pH 4.0), the mixture was vortexed and the RNA extracted with 1 vol of phenol and 0.2 vol of chloroform/isoamylalcohol (49:1). Total RNA was precipitated with isopropanol, resuspended in 1 × 10 mmol/l Tris-HCl/1 mmol/l EDTA containing 0.1 vol of 3 mol/l sodium acetate (pH 5.2) and then re-extracted with 0.5 vol phenol, followed by 0.5 vol chloroform/isoamylalcohol. The RNA was then precipitated with 3 vol of 4 mol/l sodium acetate (pH 7.0), to remove contaminating proteoglycans, at -20°C overnight. Total RNA was recovered by centrifugation, washed with 75% ethanol, air dried, dissolved in diethylpyrocarbonate-treated water, and stored at -80°C until further use. RNA concentration and purity (260/280 absorbance ratio) were determined by spectrophotometry. RNA integrity was confirmed by visualization on ethidium bromide stained 1% weight/vol agarose-formaldehyde gels.

Microarray

RNA was further purified using RNeasy columns (Qiagen, Hilden, Germany), in accordance with the manufacturer's instructions. RNA (5 μg) was amplified using a Message Amp II kit (Ambion, Austin, TX, USA) with indirect, amino allyl mediated incorporation of either Cy3 or Cy5 dyes (Amersham Biosciences, Piscataway, NJ, USA), in accordance with the manufacturer's instructions. A Compugen Human 19K-oligo library (Jamesburg, NJ, USA) spotted onto Corning glass slides (Lowell, MA, USA) by the Adelaide Microarray facility (AMF) was used in this study. The Compugen human oligo library consisted of 17,260 oligonucleotide 65-mers each representing a single human gene. The slides were interrogated by competitive hybridization with 5 μg each of Cy3 and Cy5 labelled pairs of OA-control, OA-OP, or OP-control amplified RNA samples. The sample pairs used in the microarray analysis are listed in Table 1. Sample pairs were age-matched as closely as possible.
Table 1
Control versus OA versus OP sample microarray comparisons
  
Sample pair
Slide
GEO accession number
ID
Status
Age (years)
ID
Status
Age (years)
1
GSM207548
1
CTL
85
12
OA
83
2
GSM207549
2
CTL
83
13
OA
82
3
GSM207810
2
CTL
83
14
OA
82
4
GSM207811
3
CTL
72
15
OA
78
5
GSM207550
4
CTL
72
16
OA
77
6
GSM207812
5
CTL
68
21
OA
68
7
GSM207552
6
CTL
68
17
OA
66
8
GSM207553
7
CTL
60
18
OA
60
9
GSM207554
8
CTL
56
19
OA
56
10
GSM207555
9
CTL
43
20
OA
49
11
GSM208577
37
CTL
85
47
OA
85
12
GSM208575
38
CTL
73
48
OA
77
13
GSM208578
39
CTL
71
49
OA
73
14
GSM208576
40
CTL
71
50
OA
70
15
GSM208579
41
CTL
70
51
OA
69
16
GSM208583
42
CTL
69
52
OA
63
17
GSM208580
43
CTL
64
53
OA
63
18
GSM208582
44
CTL
60
54
OA
62
19
GSM208581
45
CTL
57
55
OA
57
20
GSM208584
46
CTL
50
56
OA
50
21
GSM207805
26
OP
91
22
OA
87
22
GSM207813
27
OP
87
12
OA
83
23
GSM207803
34
OP
87
13
OA
82
24
GSM207804
28
OP
84
23
OA
79
25
GSM207808
29
OP
81
24
OA
78
26
GSM207806
35
OP
78
15
OA
78
27
GSM207807
36
OP
78
16
OA
77
28
GSM207556
35
OP
78
25
OA
73
29
GSM208574
32
OP
74
21
OA
68
30
GSM207809
33
OP
74
17
OA
66
32
GSM207798
2
CTL
83
27
OP
87
33
GSM207557
2
CTL
83
34
OP
87
34
GSM207796
10
CTL
83
28
OP
84
35
GSM207797
11
CTL
74
29
OP
81
36
GSM207799
4
CTL
72
36
OP
78
37
GSM207800
3
CTL
72
30
OP
77
38
GSM207551
3
CTL
72
31
OP
75
39
GSM207801
5
CTL
68
32
OP
74
40
GSM207802
6
CTL
68
33
OP
74
'Slide' indicates the microarray slide comparison. Slides 1 to 10 are control (CTL)-osteoarthritis (OA) female sample pairs, slides 11 to 20 are CTL-OA male sample pairs, slides 21 to 30 are OA-osteoporosis (OP) female sample pairs and slides 32 to 40 are CTL-OP female sample pairs. GEO, Gene Expression Omnibus; ID, individual/sample.
A biological dye-swap strategy was employed rather than a replicate dye swap strategy. This involved swapping of Cy3 and Cy5 labelling of the samples in each pair for each group of paired samples to balance for potential dye incorporation and signal intensity bias. It also reduced the number of slides required for the experiment and maximized the statistical power of the experiment with regard to analyzing the biological differences between samples.
Hybridization and washing of slides was carried out according to methods described on the AMF website [42]. The microarray slides were scanned twice at slightly different PMT voltage using a GenePix 4000B Scanner driven by GenePix Pro 4.0 (Axon Instruments, Foster City, CA, USA). All analyses were performed using the statistical programming and graphics environment R [43]. The 'SPOT' software package [44] was used to identify spots using the adaptive segmentation method and subtract backgrounds utilizing the morphological opening approach [45, 46]. Data analysis was performed in R using Bioconductor [47]. The Loess print tip method was used to correct for dye bias and intensity within each group of adjacent spots printed by one pin [48]. Linear modelling was performed using the linear models for microarray analysis (LIMMA) package of Bioconductor [49]. Differentially expressed genes were ranked on moderated t statistics, and those with t scores above 3 were followed up further. The moderated t-statistic score is based on the ratio of the log2 fold change to its standard error. Because there is no consensus on appropriate adjustment of P values in the context of microarrays, genes of interest were chosen based on a combination of statistical and biological indicators. Microarray data have been deposited in the Gene Expression Omnibus [50] and are accessible through Gene Expression Omnibus series number GSE8406.

Real-time PCR

First-strand reverse transcription cDNA synthesis was performed on 1 μg amplified RNA from each sample using a first-strand cDNA synthesis kit with Superscript II (Invitrogen, Carlsbad, CA, USA) and 250 ng random hexamer primer (Geneworks, Adelaide, SA, Australia), in accordance with the manufacturer's instructions. Template cDNA (1 μl of 1/100 dilution of cDNA) was amplified using iQ SYBR Green Supermix (BioRad, Hercules, CA, USA) on a Rotor-Gene thermocycler (Corbett Research, Mortlake, NSW, Australia). The reactions were incubated at 94°C for 10 min for 1 cycle, and then 94°C (20 seconds), 60°C, or 65°C (ADAMTS4 and MMP25 only; 20 seconds) and 72°C (30 seconds) for 40 cycles. This set of cycles was followed by an additional extension step at 72°C for 5 minutes. All PCR reactions were validated by the presence of a single peak in the melt curve analysis, and amplification of a single specific product was further confirmed by electrophoresis on a 2.5% weight/vol agarose gel. Primers were designed for each gene that primed in separate exons and spanned at least one intron to avoid contaminating amplification from genomic DNA. Primers were obtained from Geneworks. Amplicons were designed to be in the 100 to 200 base pairs size range. GenBank accession numbers for gene sequences and primer sequences are provided in Table 2. Real-time PCR validation was carried out using the 2-ΔΔCT method [51]. Reactions were performed in duplicate. Normalized gene expression values for each gene based on cycle threshold (CT) values for each of the genes and the housekeeping gene GAPDH were generated. Mean ± standard deviation (SD) values were generated from eight samples from each group of either OA or control samples tested.
Table 2
GenBank accession numbers and primer sequences
Gene/primer (GenBank accession number)
Forward
Reverse
GAPDH (NM_002046)
ACCCAGAAGACTGTGGATGG
CAGTGAGCTTCCCGTTCAG
ADAMTS4 (NM_005099)
GGCTACTACTATGTGCTGGAGC
TCCGCACACCATGCACTTGTCA
ADM (NM_001124)
GGATGAAGCTGGTTTCCGTC
GACTCAGAGCCCACTTATTC
ADFP (NM_001122)
GTTGCCAATACCTATGCCTG
CAGTAGTCGTCACAGCATCT
CD14 (NM_000591)
GAGGTTCGGAAGACTTATCG
ATCTTCATCGTCCAGCTCAC
COL4A1 (NM_001845)
TAGAGAGGAGCGAGATGTTC
GTGACATTAGCTGAGTCAGG
CTNNB1 (NM_001904)
GGTGCTATCTGTCTGCTCTAGT
GACGTTGACTTGGATCTGTCAGG
FST (NM_006350)
GGCAAGATGTAAAGAGCAGC
CATTATTGGTCTGGTCCACC
GADD45B (NM_015675)
TTGCAACATGACGCTGGAAG
CATTCATCAACTTGGCCGAC
IBSP (NM_004967)
CAATCCAGCTTCCCAAGAAG
CTTCTGCTTCGCTTTCTTCG
INHBA (NM_002192)
GAACTTATGGAGCAGACCTC
TGCCTTCCTTGGAAATCTCG
INSIG1 (NM_005542)
TGTATCGACAGTCACCTCGGA
GGACAGCTGGACATTATTGGC
ITGB2 (NM_000211))
AAGTGACGCTTTACCTGCGA
CCTGAGGTCATCAAGCATGG
KLF6 (NM_001300)
TGTGCAGCATCTTCCAGGAG
AACGTTCCAGCTCTAGGCAG
MEPE (NM_020203)
GCAAAGCTGTGTGGAAGAGCAGA
CCCTTATTCTCACTGGCTTCAG
MMP25 (NM_004142)
ATGTCACCGTCAGCAACGCA
CGGTCTTGATGCTGTTCTTG
MT2A (NM_005953)
GCAAATGCACCTCCTGCAAG
GTGGAAGTCGCGTTCTTTAC
NHERF1 (NM_004252)
TCACCAATGGGGAGATACAG
GTCTTGGGAATTCAGCTCCT
PTEN (NM_000314)
AAGACAAAGCCAACCGATAC
GAAGTTGAACTGCTAGCCTC
RUNX2 (NM_004348)
TGATGACACTGCCACCTCTG
GGGATGAAATGCTTGGGAAC
S100A4 (NM_002961)
GTCAGAACTAAAGGAGCTGC
TGTTGCTGTCCAAGTTGCTC
SMAD3 (NM_005902)
TTCAACAACCAGGAGTTCGC
TACTGGTCACAGTA
STC1 (NM_003155)
CCTGTGACACAGATGGGATG
GAATGGCGAGGAAGACCTTG
TIMP4 (NM_003256)
TTGACTGGTCAGGTCCTCAGT
GGTACTGTGTAGCAGGTGGT
TWIST1 (NM_000474)
TCAGCAGGGCCGGAGACCTAGAT
GTCTGGGAATCACTGTCCAC
WNT5B (AY009399)
ACCCTACTCTGGAAACTGTC
TAAACATCTCGGGTCTCTGC
'Slide' indicates the microarray slide comparison. Slides 1 to 10 are control (CTL)-osteoarthritis (OA) female sample pairs, slides 11 to 20 are CTL-OA male sample pairs, slides 21 to 30 are OA-osteoporosis (OP) female sample pairs and slides 32 to 40 are CTL-OP female sample pairs. GEO, Gene Expression Omnibus; ID, individual/sample.

Statistical analysis

The statistical significance of the differences between the means of the OA and control or OP gene expression values was determined using Student's t-test. The critical value for significance was chosen as P < 0.05.

Results

Microarray analysis of OA, control and OP bone samples

This study used Compugen human 19K-oligo human microarray slides to compare the gene expression profiles of OA, control and OP bone samples, with the aim being to identify altered gene expression in OA bone. Microarray analysis was conducted in four sets of samples (39 comparisons in total), comprising 10 OA-control female sample pairs, 10 OA-control male sample pairs, 10 OA-OP female sample pairs and 9 OP-control female sample pairs. Samples from individuals with a range of ages were analyzed in each group, but with sample pairs age-matched as closely as possible (Table 1). Bayesian statistical analysis was carried out using LIMMA to identify statistically significant differentially expressed genes between OA, control and OP bone. Log odds score versus log2 fold change volcano plots of differentially expressed genes from each of the four groups of sample pair comparisons are shown in Figure 1. The log odds (or B statistic) score is the log odds that that gene is differentially expressed. The log2 fold change represents the fold change in expression of the gene. Small levels of differential expression (ranging from 0.38-fold to 2.83-fold change in expression) were detected, with several hundred differentially expressed genes present in each grouping, with t scores above 6. The moderated t-statistic score is based on the ratio of the log2 fold change to its standard error.

Identification and functional classification of top-ranking differentially expressed genes in OA bone

By comparing the lists of ranked differentially expressed genes from each of the four initial groupings, we were able to identify a group of differentially expressed genes that was more likely to be associated with the OA disease process. This group of genes was assembled by filtering out genes that were similarly regulated between OA-control and OP-control samples in order to remove genes that were more likely to be differentially expressed because of potential differences caused by sourcing bone at surgery versus autopsy. Because there were also very few significant differences in gene expression between the male and female OA-control groups, these data were combined because it strengthened the statistical significance of the genes identified as differentially expressed. Using these selection processes, several hundred genes from each initial grouping was reduced to a list of 150 differentially expressed genes in OA bone with t scores above 4.
Gene function and pathway analyses were carried out by searching the National Centre for Biotechnology Information database [52] and by using various analysis programs including OntoExpress [53] and Gostat [54]. We were able to identify a group of 62 top-ranking OA differentially expressed genes from within the initial list of 150 genes, which have known or suspected roles (direct or indirect via angiogenesis) in influencing bone development or bone remodelling (Table 3). For many of the genes both osteogenic and angiogenic roles have been described. In addition, a subset of these genes, particularly those that encode secreted, cell surface and extracellular matrix molecules, also have potential chondrogenic functions, consistent with the proposal that an altered OA subchondral bone microenvironment could interfere with cartilage metabolism.
Table 3
Differentially expressed genes in OA bone with roles in osteogenesis, angiogenesis and chondrogenesis
         
Real-time PCR
Rank
GenBank
Role
Cell type
Pathway
Symbol
Name
t OA/CTL
t OA/OP
Symbol
OA/CTL
         
ADAMTS4
0.11
2
NM_001124
A, B, C
OB, OC, OS, CB
TGF-β/BMP
ADM
Adrenomedullin
-12.620
-7.904
ADM
0.09
3
NM_002450
B
OB
 
MT1L
Metallothionein 1L
-8.342
-6.843
  
4
NM_016109
A
 
WNT
ANGPTL4
Angiopoietin-like 4
-6.165
-8.052
  
5
NM_015675
B
OB, OC, CB
TGF-β/BMP
GADD45B
Growth arrest and DNA-damage-inducible, beta
-8.102
-5.985
GADD45B
0.15
7
NM_000358
A, B
 
TGF-β/BMP
TGF-BI
Transforming growth factor, beta-induced
-6.915
-5.741
  
9
NM_017817
   
RAB20
RAB20, member RAS oncogene family
-5.147
-6.982
  
10
NM_004967
B, C
OB, CB
WNT & TGF-β/BMP
IBSP
Integrin-binding sialoprotein
-5.088
-5.865
IBSP
0.25
12
AB014526
B
OB
 
MFAP3L
Microfibrillar-associated protein 3-like
5.284
4.983
  
13
NM_004142
A, B, C
OB, OC, CB
WNT
MMP25
Matrix metalloproteinase 25
4.942
7.183
MMP25
2.60
15
NM_002025
B
 
TGF-β/BMP
FMR2
Fragile × mental retardation 2
4.915
4.851
  
18
NM_005953
B
OB
 
MT2A
Metallothionein 2A
-4.638
-7.071
MT2A
0.18
20
NM_013258
A, B
M OC
 
PYCARD
PYD and CARD domain containing
6.567
4.355
  
21
NM_014467
   
SRPX2
Sushi-repeat-containing protein, X-linked 2
-4.333
-4.918
  
22
NM_003155
A, B, C
OB, OC, CB
 
STC1
Stanniocalcin 1
-6.656
-4.274
STC1
0.16
23
NM_001109
B
OC
 
ADAM8
A disintegrin and metalloproteinase domain 8
5.426
4.269
  
24
NM_020203
B
OB, OS
TGF-β/BMP
MEPE
Matrix, extracellular phosphoglycoprotein with ASARM motif
-5.036
-4.263
MEPE
0.11
28
S68954
B
OB
 
MT1G
Metallothionein 1G
-4.202
-7.630
  
29
AK026438
B
OB
 
GALNT4
Polypeptide N-acetylgalactosaminyltransferase 4
5.259
4.195
  
30
NM_000474
B
OB
WNT
TWIST1
Twist homolog 1
-4.194
-4.910
TWIST1
0.31
33
NM_000655
B
M
 
SELL
Selectin L
4.852
4.179
  
36
NM_002341
A, B, C
OB, CB
 
LTB
Lymphotoxin beta
4.465
4.086
  
37
NM_003226
A
  
TFF3
Trefoil factor 3
4.080
4.074
  
42
NM_000607
B
M
 
ORM1
Orosomucoid 1
4.284
3.982
  
46
NM_005542
   
INSIG1
Insulin induced gene 1
-7.756
-3.878
INSIG1
0.55
49
NM_001302
B
M
 
CORT
Cortistatin
-6.713
-3.843
  
52
NM_000598
A, B, C
OB, OC, CB
WNT & TGF-β/BMP
IGFBP3
Insulin-like growth factor binding protein 3
-3.772
-3.893
  
54
NM_012479
A, B
OB
 
YWHAG
Tyrosine 3-/tryptophan 5-monooxygenase activation protein, gamma
-3.759
-5.355
  
59
NM_014624
B
OB
WNT
S100A6
S100 calcium binding protein A6
8.214
3.710
  
61
NM_006732
A, B
OB
 
FOSB
FBJ murine osteosarcoma viral oncogene homolog B
3.682
3.926
  
64
NM_002961
A, B
OB
WNT
S100A4
S100 calcium binding protein A4
4.407
3.596
S100A4
2.43
65
NM_016184
B
M
 
CLECSF6
C-type lectin domain family 4, member A
4.575
3.594
  
68
NM_006184
B
  
NUCB1
Nucleobindin 1
-3.551
-5.513
  
69
NM_001300
A
 
WNT & TGF-β/BMP
KLF6
Kruppel-like factor 6
-4.047
-3.544
KLF6
0.41
70
NM_004864
A, B
OB, CB
TGF-β/BMP
GDF15
Growth differentiation factor 15
-3.541
-4.269
  
73
U79271
B
OB, OC
WNT
AKT3
V-akt murine thymoma viral oncogene homolog 3
3.507
4.876
  
76
S83282
A, B
OB, OC
 
MIF
Macrophage migration inhibitory factor
-3.654
-3.496
  
81
NM_005213
B
OC
 
CSTA
Cystatin A
4.450
3.447
  
82
NM_000647
A, B
M OC
 
CCR2
Chemokine (C-C motif) receptor 2
7.277
3.446
  
85
NM_001911
A
  
CTSG
Cathepsin G
8.474
3.411
  
87
NM_001122
  
WNT
ADFP
Adipose differentiation-related protein
-3.368
-10.219
ADFP
0.21
92
NM_006254
A, B
OB
 
PRKCD
Protein kinase C, delta
5.549
3.319
  
94
NM_002162
A
  
ICAM3
Intercellular adhesion molecule 3
3.304
5.798
  
95
NM_001885
B
OB, OC
 
CRYAB
Crystallin, alpha B
-3.296
-7.616
  
96
D17152
A
 
WNT
SOD2
superoxide dismutase 2
-6.347
-3.278
  
102
NM_003256
A
 
WNT
TIMP4
Tissue inhibitor of metalloproteinase 4
-6.319
-3.250
TIMP4
0.08
105
NM_000698
 
M
 
ALOX5
Arachidonate 5-lipoxygenase
3.229
4.736
  
106
NM_014029
B
OC
 
RAC2
Rho family, small GTP binding protein Rac2
3.227
5.325
  
108
NM_003670
B, C
OB, CB
TGF-β/BMP
DEC1
Differentially expressed in chondrocytes 1
-3.203
-3.783
  
109
NM_002067
B
OB
 
GNA11
Guanine nucleotide binding protein, alpha 11
-4.353
-3.202
  
115
AF263545
B
OB
 
SLC14A1
Solute carrier family 14, member 1
3.163
5.759
  
120
NM_005127
B
M
 
CLECSF2
C-type lectin domain family 2, member B
-7.330
-3.141
  
123
NM_004252
B
 
WNT
NHERF1
Sodium/Hydrogen exchanger regulatory factor 1
5.280
3.138
NHERF1
1.71
124
NM_006186
B
OB
 
NR4A2
Nuclear receptor subfamily 4, group A, member 2
-3.128
-3.354
  
133
NM_000104
B
OC
 
CYP1B1
Cytochrome P450, family 1, subfamily B, 1
-3.075
-5.148
  
136
NM_003005
B
  
SELP
Selectin P
8.740
3.066
  
139
NM_004334
B
OC
 
BST1
Bone marrow stromal cell antigen 1 (CD157)
3.037
3.601
  
141
AK023619
A
  
CRIM1
Cysteine-rich motor neuron 1
-3.043
-3.271
  
143
NM_013332
A
 
WNT
HIG2
Hypoxia-inducible protein 2
-6.019
-3.035
  
144
NM_001845
A, B
 
TGF-β/BMP
COL4A1
Collagen, type IV, alpha 1
-3.028
-8.960
COL4A1
0.57
146
NM_016224
A, B
OB
WNT
SNX9
Sorting nexin 9
-3.025
-6.086
  
Rank' indicates the ranking within the top 150 differentially expressed genes in osteoarthritis (OA) bone compared with control (CTL) and osteoporosis (OP) bone. 'Role' indicates the known or suspected role of the gene: A, angiogenic; B, osteogenic; and C, chondrogenic. 'Cell type' indicates the cell type that the gene is expressed in or affects: OB, osteoblast, OC, osteoclast, OS, osteocyte, CB, chondroblast, or M, monocyte. 't OA/CTL' is the t score of OA compared with CTL differential expression of gene: a positive value indicates upregulation in OA and a negative one indicates downregulation in OA. 't OA/OP' is the t score of OA compared with OP differential expression of gene: a positive value indicates upregulation in OA and a negative one indicates downregulation in OA. The moderated t-statistic score is based on the ratio of the log2 fold change to its standard error. 'OA/CTL' under 'Real-time PCR' indicates the fold change in gene expression expressed as ratio of OA to CTL.
Although many of the genes identified in this analysis have pleiotropic effects in bone and other tissues, it was of interest that many of the top-ranking differentially expressed genes in OA bone have known or suspected roles in osteoblast and osteocyte differentiation and function. These genes included ADAMTS4, ADM, GADD45B, IBSP, MMP25, MT2A, STC1, MEPE, TWIST1, IGFBP3, S100A4, AKT3 and COL4A1. There was also a group of differentially expressed genes in OA bone that have known or potential roles in osteoclast function, such as the previously mentioned osteoblast-related genes ADAMTS4, GADD45B, STC1 and IGFB3, as well as ADAM8, CCR2, CSTA, RAC2, CRYAB and CYP1B. Functionally, within the list of genes given in Table 3, there are genes encoding secreted molecules (ADM, ANGPTL4, STC1, CORT, IGFBP3 and MIF), cell surface molecules (SELL, ICAM3, SELP, CRIM1, CLECSF6, CLECSF2, CCR2 and SLC14A1), intracellular signalling molecules (RAB20, YWHAG, RAC2, NHERF1, GNA11 and SNX9), protein kinases (AKT3 and PRKCD), calcium and metal ion binding proteins (S100A4, S100A6, MT1L, MT2A and MT1G), transcription factors (TWIST1, FMR2, KLF6, NR4A2 and DEC1), and both enzymatic (ADAMTS4, MMP25, ADAM8, TIMP4, GALNT4 and CTSG) and structural (TGFBI, IBSP, MEPE, MFAP3L and COL4A1) extracellular matrix molecules.
Because of the small absolute differences in gene expression between the bone tissue samples, real-time PCR was used to confirm a selection of the differentially expressed genes identified by the microarray analysis of OA, control and OP bone. The real-time PCR results (depicted as fold differential expression) are shown alongside the microarray results in Table 3. In total, the differential expression levels of 20 genes were examined using real-time PCR. Results for 16 genes reached statistical significance (P < 0.01) for differential expression between OA and control bone. The differential expression of four genes (TGFBI, S100A6, SLC14A1 and SNX9) could not be confirmed. The female control samples 1–8 (age range 56 to 85 years, mean [ ± SD] age 70.5 ± 10 years) and female OA samples 12–19 (age range 56 to 83 years; mean age 73 ± 10.8 years) were used to confirm the microarray data by real-time PCR (Table 1). The mean age of the OA group did not differ significantly from that in the control group. Interestingly, although the microarray expression ratios were quite small (ranging from 0.62-fold change to 1.47-fold change in expression), the fold difference in expression identified using the real-time PCR reactions was significantly greater in most cases (ranging from 0.08-fold change to 2.6-fold change in expression). This probably reflects differences in sensitivity between the two techniques [55, 56]. The difference is probably also accentuated by the competitive pair-wise comparison of samples used by the microarray platform in this study compared with the individual gene/GAPDH CT expression ratio values generated using real-time PCR. Encouragingly, there was a high confirmation rate with the real-time PCR and consistency between the microarray and PCR detection of expression ratio differences for each of the genes analyzed, suggesting that the majority of the genes identified by the microarray are bona fide differentially expressed genes in OA bone.

Altered expression of WNT and TGF-β/BMP signalling pathway component and target genes in OA bone

A significant number of the top-ranking differentially expressed genes in OA bone were identified as WNT signalling pathway targets (Table 3). WNT targets included upregulated genes such as MMP25 and S100A4, and downregulated genes such as IBSP, TWIST1 and TIMP4. The altered expression of these genes suggests that WNT signalling may be perturbed in the OA bone microenvironment. This was apparently borne out by closer examination of the extended list of differentially expressed genes in OA bone, which revealed further WNT signalling pathway components and modulators such as WNT5B, FZD3, SFRP5, APC, AXIN2, PTEN and NHERF1. These genes, and additional WNT target genes such as CD14, APOE, ID1, IL6, FST and RUNX2, are listed in Table 4. The differences in expression of this group of genes (t scores above 3) in general were not as pronounced as that seen for the target genes identified from within the top-ranking 150 genes.
Table 4
WNT and TGF-β/BMP signalling pathway components and target genes differentially expressed in OA bone
       
Real-time PCR
GenBank
Role
Cell type
Symbol
Name
t OA/CTL
t OA/OP
Symbol
OA/CTL
WNT pathway components and modulators
AY009399
B, C
OB, OC, CB
WNT5B
Wingless-type MMTV integration site family, member 5B
3.719
0.717
WNT5B
2.52
NM_002332
A, B, C
OB, OC, CB
LRP1
Low density lipoprotein-related protein 1
3.062
-7.071
  
NM_002333
  
LRP3
Low density lipoprotein-related protein 3
5.950
1.146
  
NM_017412
  
FZD3
Frizzled homolog 3
-3.677
0.835
  
AF086500
  
FZD8
Frizzled homolog 8
3.024
-1.557
  
NM_003015
  
SFRP5
Secreted frizzled-related protein 5
-4.338
1.154
  
NM_004655
  
AXIN2
Axin 2
-3.123
2.490
  
NM_000038
  
APC
Adenomatosis polyposis coli
-5.171
1.155
  
AK023892
  
DAAM1
Dishevelled associated activator of morphogenesis 1
-6.704
2.850
  
NM_000314
A, B
OB, OC
PTEN
Phosphatase and tensin homolog
-5.288
-0.216
PTEN
0.30
AF028823
  
TIP1
Tax interaction protein 1
5.057
-5.674
  
AB006630
  
TCF20
Transcription factor 20
8.559
2.719
  
AK026898
B
M
FOXP1
Forkhead box P1
-5.122
1.761
  
WNT inducible/target genes
NM_000591
B
OB, OC, M
CD14
CD14 antigen
5.965
-7.036
CD14
3.47
NM_004039
A, B
OB, OC, M
ANXA2
Annexin A2
5.716
-7.601
  
NM_000211
B
OC
ITGB2
Integrin, beta 2 (CD18)
6.104
-1.243
ITGB2
2.95
NM_018456
  
EAF2
ELL associated factor 2
6.450
0.884
  
NM_000041
B
OB, M
APOE
Apolipoprotein E
6.896
-2.391
  
AB012643
B
OB
ALPL
Alkaline phosphatase, liver/bone/kidney
-5.942
-1.213
  
NM_002229
B
OB
JUNB
Jun B proto-oncogene
5.053
-3.804
  
NM_003377
A, B
 
VEGFB
Vascular endothelial growth factor B
3.238
-3.772
  
NM_005429
A, B
 
VEGFC
Vascular endothelial growth factor C
5.304
0.473
  
NM_000963
B
OB
PTGS2
Prostaglandin-endoperoxide synthase 2
4.346
-0.842
  
NM_005194
B
OB
CEBPB
CCAAT/enhancer binding protein, beta
4.273
-4.092
  
AL353944
A, B
OB
ID1
Inhibitor of DNA binding 1
3.923
-1.996
  
NM_000900
B
OB
MGP
Matrix Gla protein
2.791
-3.073
  
M14584
A, B, C
OB, OC, CB
IL6
Interleukin 6
2.589
-3.612
  
NM_006350
B, C
OB, CB
FST
Follistatin
-2.137
-7.912
FST
0.38
AL353944
B
OB
RUNX2
Runt-related transcription factor 2
1.611
1.050
RUNX2
2.07
TGF-β/BMP pathway components and modulators
M38449
A, B, C
OB, OC, CB
TGFB1
Transforming growth factor, beta 1
2.739
3.054
  
AK021486
B, C
OB, CB
BMP5
Bone morphogenetic protein 5
-10.314
-2.263
  
NM_002192
A, B, C
OB, OC, CB
INHBA
Inhibin, beta A
-7.699
-0.389
INHBA
0.31
NM_006350
B, C
OB, CB
FST
Follistatin
-2.137
-7.912
FST
0.38
NM_004612
B
 
TGFBR1
Transforming growth factor, beta receptor I
-4.915
1.049
  
NM_000118
A
 
ENG
Endoglin
4.131
-2.461
  
NM_001105
B
OB
ACVR1
Activin A receptor, type I
3.791
-0.941
  
NM_003573
  
LTBP4
Latent transforming growth factor beta binding protein 4
-4.060
2.623
  
NM_005902
A, B
OB
SMAD3
SMAD, mothers against DPP homolog 3
4.119
-3.582
SMAD3
2.64
NM_005359
A, B
OB
SMAD4
SMAD, mothers against DPP homolog 4
3.957
0.156
  
NM_002165
A, B
OB
ID1
Inhibitor of DNA binding 1
3.923
-1.996
  
NM_002229
B
OB
JUNB
Jun B proto-oncogene
5.053
-3.804
  
NM_005655
B
OB
KLF10
Kruppel-like factor 10
4.443
-4.449
  
NM_006037
B
OB
HDAC4
Histone deacetylase 4
8.475
2.468
  
NM_000168
B
OB
GLI3
GLI-Kruppel family member GLI3
-4.675
-0.298
  
AL353944
B
OB
RUNX2
Runt-related transcription factor 2
1.611
1.050
RUNX2
2.07
TGF-β/BMP inducible/target genes
AK001052
B
OB
FGFR1
Fibroblast growth factor receptor 1
4.234
1.993
  
NM_001553
A
 
IGFBP7
Insulin-like growth factor binding protein 7
5.137
-6.302
  
NM_000211
B
OC
ITGB2
Integrin, beta 2 (CD18)
6.104
-1.243
ITGB2
2.95
AK001060
B, C
OB, CB
DCN
Decorin
-6.325
2.628
  
NM_000177
B
OC
GSN
Gelsolin
4.725
-2.387
  
X55525
B
OB
COL1A2
Collagen, type I, alpha 2
5.538
-3.633
  
NM_003118
A, B
OB
SPARC
Secreted protein, acidic, cysteine-rich
3.188
-1.844
  
NM_004407
B
OB, OS
DMP1
Dentin matrix acidic phosphoprotein
-3.523
-1.397
  
NM_001831
A, B
 
CLU
Clusterin
4.803
-2.030
  
Not represented on Compugen human 19K microarray
NM_001904
A, B
OB, OC
CTNNB1
Catenin beta 1
  
CTNNB1
2.38
'Role' indicates the known or suspected role of the gene: A, angiogenic; B, osteogenic; and C, chondrogenic. 'Cell type' indicates the cell type that the gene is expressed in or affects: OB, osteoblast, OC, osteoclast, OS, osteocyte, CB, chondroblast, or M, monocyte. 't OA/CTL' is the t score of osteoarthritis (OA) compared with control (CTL) differential expression of gene: a positive value indicates upregulation in OA and a negative one indicates downregulation in OA. 't OA/OP' is the t score of OA compared with osteoporosis (OP) differential expression of gene: a positive value indicates upregulation in OA and a negative one indicates downregulation in OA. The moderated t-statistic score is based on the ratio of the log2 fold change to its standard error. 'OA/CTL' under 'Real-time PCR' indicates the fold change in gene expression expressed as ratio of OA to CTL. BMP, bone morphogenic protein; TGF, transforming growth factor.
In addition, a significant number of TGF-β/BMP signalling pathway target genes were identified as being differentially expressed in OA bone (Table 3). TGF-β/BMP signalling pathway targets included downregulated genes such as ADAMTS4, ADM, GADD45B, MEPE and COL4A1. The altered expression of these genes also suggests that TGF-β/BMP signalling may be perturbed in the OA bone microenvironment. Additional evidence for this was that genes for TGF-β/BMP signalling pathway components and modulators, such as TGFB1, BMP5, INHBA, SMAD3 and FST, were also identified in the extended list of differentially expressed genes in OA bone. These genes, and additional TGF-β/BMP target genes identified, such as COL1A2, GSN, DMP1 and ITGB2, are listed in Table 4. The differences in expression of this group of genes (t scores above 3) was not as pronounced as the target genes identified from within the top-ranking 150 genes.
Like the top-ranking list of 150 differentially expressed genes in OA bone, many of the WNT and TGF-β/BMP signalling pathway related genes identified in Table 4 also have known or suspected roles in either osteoblast (WNT5B, PTEN, CD14, SMAD3, RUNX2, ID1, HDAC4, TGFB1, BMP5, INHBA, DMP1 and FST) or osteoclast (CD14, PTEN, FOXP1, ANXA2, ITGB2, IL6 and GSN) differentiation and function.
The differential expression of a selection of these WNT and TGF-β/BMP signalling pathway component and target genes was confirmed by real-time PCR. In total, the differential expression of 11 genes was examined by real-time PCR (Table 4). The differential expression of two genes (LRP1 and IGFBP7) could not be confirmed. However, results for the other nine genes reached statistical significance (P < 0.01) for differential expression between OA and control bone. CTNNB1 was assayed directly by real-time PCR because it was not represented on the Compugen H19K library. Seven of the remaining nine genes (with FST and RUNX2 being the exceptions) were represented in the top-ranking 300 genes differentially regulated in OA bone. These genes were tested, like those listed in Table 3, using female control samples 1 to 8 and female OA samples 12 to 19 (Table 1). The range of fold difference in expression identified by the real-time PCR reactions was slightly smaller (ranging from 0.3-fold change to 3.47-fold change in expression) than for the group of genes tested from the top ranking 150 genes in Table 3.

Identification of differentially expressed OA genes between females and males

There is a higher incidence of primary hip OA in females than in males [1], and we were interested in identifying differences in gene expression between females and males that may contribute to this disparity. Therefore, we tested for differences between the OA-control female and male microarray datasets. Genes with the greatest difference in expression between females and males in the OA-control microarray comparisons are listed in Table 5. There were very few significant differences in gene expression between females and males. However, approximately 50 genes with t scores above 3, which included the top-ranking 20 genes with t scores above 4, were identified as being differentially expressed between females and males. Interestingly, a significant proportion of these genes have known or suspected roles in osteoclast-lineage cells and osteoclasts (GSN, PTK9, VCAM1, ITGB2, GRN, ANXA2, PDE4A and FOXP1). There are also genes with known roles in osteoblasts (LTF, DF, PRKCG and TGFB1). A number of the highest ranking differentially expressed genes between females and males in OA bone also involve WNT signalling pathway components, including WNT5B, along with the EAF2 and CTBP2 genes, which encode transcription factors that are involved in mediating WNT signalling.
Table 5
Genes differentially expressed between female and male OA and control bone samples
GenBank
Role
Cell type
Symbol
Name
t OAF/OAM
t OA/CTL
t OA/OP
Differentially expressed genes in top 20 regulated genes
NM_000177
B
OC
GSN
Gelsolin
5.983
4.725
-2.387
NM_001078
B
OB, OC
VCAM1
Vascular cell adhesion molecule 1
5.158
-1.358
-2.377
NM_006764
  
IFRD2
Interferon-related developmental regulator 2
-5.081
4.274
2.162
NM_018456
  
EAF2
ELL associated factor 2
4.574
6.450
0.884
NM_002087
B
OC
GRN
Granulin
4.562
11.161
-2.008
NM_000211
B
OC
ITGB2
Integrin, beta 2 (CD18)
4.378
6.104
-1.243
NM_004203
  
PKMYT1
Protein kinase, membrane associated tyrosine/threonine 1
-4.373
-3.375
1.741
NM_001329
  
CTBP2
C-terminal binding protein 2
-4.339
1.548
4.057
NM_006202
B
OB, OC
PDE4A
Phosphodiesterase 4A
-4.131
-1.565
3.337
NM_017845
  
COMMD8
COMM domain containing 8
4.118
3.264
-0.398
AY009399
B, C
OB, OC, CB
WNT5B
Wingless-type MMTV integration site family, member 5B
4.107
3.719
0.717
NM_002822
B
OC
PTK9
PTK9 protein tyrosine kinase 9
4.099
-0.597
-5.894
NM_002343
B
OB
LTF
Lactotransferrin
4.096
4.918
1.882
Additional differentially expressed genes in top 50 regulated genes
NM_005606
A, B
OB, OC
LGMN
Legumain
3.853
9.631
-4.470
NM_015946
A, B, C
OB, CB
ITGA1
Integrin, alpha 1
3.813
3.904
0.131
NM_004039
B
OB, OC
ANXA2
Annexin A2
3.754
5.716
-7.601
M14087
A, B
OB
LGALS1
Lectin, galactoside-binding, soluble,
3.705
4.583
-0.045
NM_006079
A, B
OB
CITED2
Cbp/p300-interacting transactivator with Glu/Asp-rich domain 2
3.591
8.580
-1.403
M38449
A, B, C
OB, OC, CB
TGFB1
Transforming growth factor, beta 1
-3.575
2.739
3.054
Z15114
A, B
OB
PRKCG
Protein kinase C, gamma
-3.570
0.018
3.741
NM_003639
B
OC
IKBKG
I kappa B kinase gamma
3.376
6.775
-1.704
AK026898
B
OC
FOXP1
Forkhead box P1
-3.348
-5.122
1.761
NM_001928
B
OB
DF
D component of complement (adipsin)
3.306
6.851
-0.991
NM_006037
B
OB
HDAC4
Histone deacetylase 4
3.272
8.475
2.468
NM_001742
B
OC
CALCR
Calcitonin receptor
-3.036
0.663
1.800
Not detected by microarray to be differentially regulated between females and males
NM_004142
A, B, C
OB, OC, CB
MMP25
Matrix metalloproteinase 25
0.237
4.942
7.183
'Role' indicates the known or suspected role of the gene: A, angiogenic; B, osteogenic; and C, chondrogenic. 'Cell type' indicates the cell type that the gene is expressed in or affects: OB, osteoblast, OC, osteoclast, OS, osteocyte, or CB, chondroblast. 't OAF/OAM' is the t score of osteoarthritis (OA) female compared with OA male differential expression of gene: a postive value indicates upregulation in OA female and a negative one indicates downregulation in OA female. 't OA/CTL' is the t score of OA compared with CTL differential expression of gene: a positive value indicates upregulation in OA and a negative one indicates downregulation in OA. 't OA/OP' is the t score of OA compared with osteoporosis (OP) differential expression of gene: a positive value indicates upregulation in OA and a negative one indicates downregulation in OA. The moderated t-statistic score is based on the ratio of the log2 fold change to its standard error.
The differential expressions of WNT5B and ITGB2 (along with MMP25) between females and males in OA bone were confirmed by real-time PCR (Figure 2). MMP25 was not originally identified as being differentially expressed between females and males in OA bone by microarray analysis. WNT5B, ITGB2 and MMP25 were all found to be differentially expressed only in females, and not in males, between OA and control bone. The OA/control ratios of expression for the WNT5B gene were 2.52 in female samples (P < 0.01) and 0.92 in male samples (P = 0.7486); those for the ITGB2 gene were 2.95 (P < 0.01) and 1.35 (P = 0.1173), respectively; and those for the MMP25 gene were 2.60 (P < 0.01) and 1.01 (P = 0.4748), respectively. There was also a significant difference in the total expression levels of these three genes between females and males, being approximately 6-fold, 14-fold and 23-fold higher for WNT5B, MMP25 and ITGB2, respectively, in females than in males in OA bone. Thirteen other genes (ADAM8, ADM, ADAMTS4, ADFP, CD14, COL14A1, GADD45B, LRP1, S100A4, SMAD3, TGFBI, TIMP4 and TWIST1) were also tested for differential expression between females and males in OA bone, but none of these was found to be differently expressed (data not shown). Genes were tested using female control samples 1 to 8 and OA samples 12 to 19 as compared with male control samples 37 to 44 (age range 60 to 85 years; mean [ ± SD] age 70.4 ± 7.3 years) and OA samples 47 to 54 (62 to 85 years; 70.3 ± 8 years; Table 1). The mean ages of the female and male OA groups did not significantly differ from those of the control groups. Of the other 13 genes examined by PCR, only SMAD3 had significant differences in total expression levels between males and females, being approximately 2-fold higher in males than in females (Figure 2). However, SMAD3 was similarly differentially regulated between OA and control bone in both females and males (ratio of OA/control: 2.64 in females [P < 0.01] and 2.08 in males [P < 0.01]).

Discussion

In this study we identified altered expression of WNT and TGF-β/BMP signalling pathway component and target genes in OA bone distal to the disease site (from the IT region of the proximal femur). This was accomplished by using microarray analysis to compare gene expression in bone from individuals with end-stage OA disease and individuals without obvious OA (control or OP). The genes identified include those with known or suspected roles in osteoblast, osteocyte and osteoclast differentiation and function, as well as angiogenesis, suggesting perturbation of these processes and a role for altered bone remodelling in OA pathogenesis.
The trabecular bone sampled in this study included all of the cellular elements of bone, including the bone marrow. This has provided a 'snapshot' of the gene expression in OA bone, with contributions from all of the different cells in the bone microenvironment. Although the specific cell type(s) contributing to the altered gene expression cannot readily be identified, osteoblasts and osteocytes, which represent the most abundant cells in the trabecular bone [57], would be expected to contribute significantly to the altered gene expression measured. In addition, because we have analyzed bone from end-stage OA, it is difficult to determine unequivocally that the altered gene expression detected in the OA bone is causal or secondary to the disease. However, by sampling the IT region we have avoided secondary pathogenic changes that the subchondral bone undergoes at the joint as the disease progresses (such as sclerosis, osteophytes and cysts), which could confound identification of altered gene expression potentially responsible for the underlying subchondral bone remodelling. Therefore, we suggest that the altered gene expression identified in the IT region from OA bone may be informative about underlying systemic OA disease mechanisms that also operate at the joint in the subchondral bone.
The first main finding of this study is that many of the top-ranking differentially expressed genes in OA bone (Table 3) have known or suspected roles in osteoblast and osteocyte differentiation and function. (Also see Table 6 for descriptions of the functions of selected genes: MEPE, IBSP, MT2A, ADM, STC1, IGFBP3, GADD45B, ADAMTS4, S100A4 and MMP25.) Significantly, the changes in expression of these genes as a group suggest altered osteoblast and osteocyte activity in OA bone, which is consistent with increased bone volume fraction and under-mineralization previously reported in OA bone [3, 4, 11, 2225]. For instance, MEPE (matrix extracellular phosphoglycoprotein), which is highly expressed in osteoblasts and osteocytes, appears to be an important regulator of bone formation and mineralization. Targeted disruption of MEPE has been found to result in increased bone formation and bone mass [58]. MEPE was found to be downregulated in OA bone, which is consistent with the increased trabecular bone volume in OA. MMP25 (upregulated in OA bone) encodes a metalloproteinase that plays a role in MMP2 activation [59], and MMP2 is a major effector in osteocytes, with MMP2-/- mice exhibiting disrupted osteocytic networks and altered bone remodelling and mineralization [60]. The MMP2-/- mice have a complex bone phenotype that includes reduced bone volume in the long bones. Upregulation of MMP25 in OA bone is consistent with increased activity of MMP2 and increased bone volume.
Table 6
Bone related-functions of a selection of differentially expressed genes in osteoarthritis bone
Gene
Description/function
References
Bone remodeling, osteoblast: upregulated in osteoarthritis (OA) bone
    S100A4
Negative regulator of matrix mineralization in osteoblasts
[88,89]
    MMP25
Metalloproteinase with role in matrix metalloproteinase (MMP)2 activation. Mice lacking MMP2 have disrupted osteocytes and altered bone mineralization
[59,60]
Bone remodeling, osteoblast: downregulated in OA bone
    MEPE
Extracellular matrix protein, highly expressed in osteocytes
[58]
    IBSP
Major constituent of the bone matrix, thought to initiate and regulate mineralization
[90]
    MT2A
Metallothionein proteins (also MT1L and MT1G) have roles in regulating osteoblast differentiation and mineralization
[91,92]
    ADM
Adrenomedullin stimulates osteoblast activity, but also interacts with and influences the effects of key bone regulators insulin-like growth factor (IGF)1 and transforming growth factor (TGF)-β
[93,94]
    STC1
Stanniocalcin inhibits calcium uptake and has inhibitory effect on bone growth during development
[95,96]
    IGFBP3
Constitutive over-expression of IGF1-binding protein (IGFBP)3 impairs osteoblast proliferation and bone formation
[97]
    GADD45B
Mice deficient in GADD45B protein have defective bone mineralization
[98]
    ADAMTS4
Metalloproteinase involved in remodelling extracellular matrix. Upregulated in fracture healing. Expressed in osteocytes and osteoblasts
[99,100]
    LTF
Lactotransferrin is an anabolic bone factor
[101]
    DF
Adipsin inhibits osteoblastogenesis
[102]
Bone remodeling, osteoclast: upregulated in OA bone
    ADAM8
Stimulatory role in osteoclast formation and differentiation
[103]
    CCR2
Receptor for CC chemokine ligand (CCL)2, which promotes recruitment and fusion of monocytes/osteoclast precursors
[104]
    RAC2
Member of the Rho-GTPase subfamily. Involved in organisation of cytoskeleton and adhesion of osteoclasts to bone
[105]
    CD14
Monocyte/osteoclast precursor marker. CD14-deficient mice have increased bone mass
[106,107]
    ANXA2
Stimulates osteoclast precursor proliferation and differentiation through production of granulocyte-macrophage colony-stimulating factor (GMCSF) and receptor activator of nuclear factor-kB ligand (RANKL)
[108]
    GSN
Gelsolin deficiency in mice blocks podosome assembly in osteoclasts and produces increased bone mass
[109]
    ITGB2
Adhesion molecule important in cell-to-cell contacts during the early stage of osteoclast development
[110,111]
Bone remodeling, osteoclast: downregulated in OA bone
    IGFBP3
Over-expression of IGFBP3 in mice increases osteoclast number and bone resorption
[97]
    STC1
Stanniocalcin suppresses osteoclast activity
[96]
    PTEN
Over-expression of PTEN suppresses RANKL-stimulated signal transduction during osteoclast differentiation
[112]
    FOXP1
Transcriptional repressor that has role in modulating monocyte differentiation
[113]
    PDE4A
Down-regulated during monocyte to macrophage/osteoclast differentiation
[114]
WNT pathway components and modulators: upregulated in OA bone
    WNT5B
WNT ligand with roles in osteoblastogenic and chondrogenic differentiation
[68,69,70]
    CTNNB1
Mice lacking b-catenin in osteoblasts develop severe osteopenia with increased osteoclastogenesis and impaired osteoblastogenesis
[115]
    AKT3
Member of the AKT kinase family. Role in regulating osteoblast lifespan
[116,117]
    NHERF1
Mediates parathyroid hormone receptor signalling. Interacts with b-catenin, potentiating the effects of parathyroid hormone (PTH) on WNT signalling in bone.
[118,119]
WNT pathway components and modulators: downregulated in OA bone
    FZD3
WNT5B co-receptor
[120]
    PTEN
Modulates/antagonises WNT signalling. Roles in osteoclast and osteoblast differentiation
[112,117]
    APC
Loss of APC in mice leads to increased bone mass
[115]
    AXIN2
Negatively regulates both expansion of osteoprogenitors and maturation of osteoblasts through its modulation of WNT signalling
[121]
TGF-β/bone morphogenic protein (BMP) pathway components and modulators: upregulated in OA bone
    ACVR1
BMP and activin A receptor. Mutation in receptor causes ectopic osteogenesis
[122]
    SMAD3
Important mediator of TGF-β signalling and regulator of osteoblastogenesis and bone formation
[80,82]
    ID1
Transcription factor with roles in bone formation and osteoblast proliferation and differentiation
[123]
    RUNX2
Key transcription factor involved in promoting osteoblast differentiation
[76]
TGF-β/BMP pathway components and modulators: downregulated in OA bone
    TGFB1
Growth factor with key role in regulating bone development and metabolism
[34,80]
    INHBA
TGF-β family member. Can act as either inhibitor or activator of bone formation and osteoblast differentiation
[124,125]
    BMP5
Secreted signalling molecule involved in skeletal development and genetically implicated in OA
[29]
    FST
TGF-β/BMP antagonist that inhibits osteoblast differentiation
[126]
Interestingly, and in contrast to the large number of differentially expressed genes identified in OA bone with osteoblast-related and osteocyte-related roles, a substantial group of top-ranking differentially expressed genes identified in OP bone (data not shown) have known or suspected roles in osteoclast-lineage cells. The change in expression of these genes as a group is consistent with increased osteoclast numbers, activity and bone resorption, leading to the reduction of bone volume seen in OP. There was also a group of differentially expressed genes identified in OA bone that have known or potential roles in osteoclast function (Tables 3 to 5). However, the overall change in expression of these genes, as a group, was not as consistent as for those genes identified with osteoblast-related functions. For instance, the change in expression of a subset of these genes (ADAMTS4, GADD45B, IGFBP3 and CSTA) is consistent with decreased osteoclast activity and increased bone volume in OA, whereas the change in expression of another subset of these osteoclast-related genes (ADAM8, STC1, CCR2, RAC2, CRYAB, CYP1B, CD14, PTEN, ANXA2 and GSN) suggests upregulated osteoclast activity (Table 6). It is now becoming clear that, in addition to osteoblasts being intimately involved in influencing osteoclast-lineage cell differentiation and function [61], the converse may also be true [62, 63]. As a result, perturbed osteoblast or osteoclast gene expression could lead to complex changes in communication between these bone cell types and their remodelling behaviour in OA bone. Therefore, differential expression of a subset of the osteoclast-related genes, suggesting upregulated osteoclast activity, is consistent with the increased levels of bone remodelling seen in OA bone and perhaps a net gain of under-mineralized bone rather than the net loss of bone volume seen in OP.
Important inter-relationships between bone remodelling and angiogenesis are also now emerging, and so perturbations to angiogenic molecular pathways could contribute to the changes seen in OA bone. Consistent with a role for increased angiogenesis in OA bone, leading to increased bone volume and potentially turnover, is the upregulation of a group of top-ranking differentially expressed genes with recognized pro-angiogenic functions (MMP25, S100A4, FOSB, TFF3, CTSG and LTB) and downregulation of a group of genes that negatively influence angiogenesis (HIG2, ADAMTS4, ANGPTL4, STC1, KLF6, IGFBP3, TIMP4 and GDF15).
In addition, a subset of the genes with roles in osteoblasts, particularly those that encode secreted, cell surface and extracellular matrix molecules, including ADM, IBSP, MMP25, STC1, IGFBP3, WNT5B, FST, TGFB1, BMP5 and INHBA, also have potential chondrogenic functions. Altered expression of many of these genes is consistent with the proposal that similar altered expression of these genes in osteoblasts in the subchondral bone microenvironment could interfere with chondrocyte or cartilage metabolism. For instance, ADM, which is downregulated in OA bone has a pro-chondrogenic role [64]. Reduced levels of ADM could negatively affect chondrocyte/cartilage metabolism.
The second significant and connected observation from this study was that many of the top-ranking differentially expressed genes identified in OA bone (with osteoblast, osteocyte and osteoclast related roles) were WNT or TGF-β/BMP signalling pathway target genes. This observation, on closer examination of the ranked list of differentially expressed in OA bone, led to the identification of additional sets of differentially expressed genes that were WNT or TGF-β/BMP signalling pathway component or modulator genes. These data together suggest that the WNT and TGF-β/BMP signalling pathways are altered in OA bone and may play a role(s) in OA pathogenesis. Both the WNT and TGF-β/BMP signalling pathways have been implicated in influencing bone mass and bone remodelling [3234] and have been demonstrated to do this by controlling both osteoblast and osteoclast differentiation and function [6567].
WNT signalling, in terms of bone mass and bone remodelling, is a very complex process that depends on the interplay of a large number of WNT ligands, the receptors they complex with, prevailing antagonists and particular combinations of β-catenin/transcription factor complexes that ultimately control the expression of the target genes. Interestingly, the only gene encoding a WNT ligand, WNT5B, that was identified as being differentially expressed (upregulated in OA bone) in the present study was recently demonstrated to increase in expression during in vitro osteoblast differentiation [68]. The protein encoded by WNT5B is known to have both stimulatory and inhibitory effects on bone and cartilage cells, and signals through both the canonical and noncanonical WNT signalling pathways, depending on the receptor it complexes with at the cell surface [6870]. Along with WNT5B, there were also several other important WNT pathway related genes that were altered in their expression in OA bone. Genes for the WNT5B co-receptor FZD3 and extracellular WNT antagonist SFRP5 were under-expressed, relative to controls, suggesting increased WNT signalling. Downregulation of the intracellular signalling cascade genes PTEN, APC and AXIN2, and upregulation of CTNNB1, AKT3 and NHERF1 are also consistent with increased WNT signalling. CTNNB1 encodes β-catenin, which is the central downstream mediator of canonical WNT signalling, which forms a complex with lymphoid enhancer factor/T cell factor (LEF/TCF) transcription factors to modulate target gene expression [71], whereas PTEN, APC, AXIN2, AKT3 and NHERF1 gene products modulate β-catenin activity (Table 6).
Like WNT signalling, TGF-β/BMP signalling is similarly complex, with a large family of ligands, cognate receptors and intracellular signalling molecules involved in the pathway, exerting both stimulatory and inhibitory effects on bone remodelling. Several TGF-β/BMP signalling pathway component and modulator genes that influence osteoblast function, bone remodelling and bone mineralization were identified as altered in OA bone. These included TGFB1, INHBA, ACVR1, BMP5, FST and SMAD3 (Table 6).
There is significant crosstalk between the WNT and TGF-β/BMP signalling pathways. β-Catenin, SMAD3 and runt-related transcription factor (RUNX)2 potentially play important roles in mediating the crosstalk between the WNT and TGF-β/BMP signalling pathways via direct interactions and in complexes with the TCF/LEF transcription factor family members in the nucleus [7275]. We observed increased RUNX2 expression in OA bone in this study, which is consistent with increased osteoblast differentiation and activity in OA bone. RUNX2, which is a WNT inducible gene, encodes a transcription factor that plays roles in mediating both WNT and TGF-β/BMP signalling, and is essential for osteoblast differentiation and skeletal development [66, 7577]. Intriguingly, decreased expression of RUNX2 has been shown to reduce cartilage destruction and subchondral bone changes in a mouse joint instability OA model [78], suggesting a role for increased RUNX2 expression in OA pathogenesis. An important role for RUNX2 in OA pathogenesis is supported by our microarray data. The products of several of the top-ranking differentially expressed genes identified, such as TWIST1 (twist homologue 1) [79], FOXF1 (forkhead box F1), ID1 (inhibitor of DNA binding 1), HDAC4 (histone deacetylase 4) and SMAD3, modify RUNX2 expression or interact with and modify RUNX2 function. SMAD3 is an important mediator of TGF-β regulation of bone mechanical properties and composition [80]. TGF-β represses RUNX2, and one of the ways it does this is through recruitment of the histone deacetylase HDAC4 by SMAD3 [81]. However, SMAD3 over-expression (SMAD3 was upregulated in OA bone in the present study) has also been reported to induce RUNX2 expression and osteoblast differentiation [82]. Significantly, several of the highest ranking differentially expressed genes identified in this study in OA bone are targets of RUNX2 and SMAD3, such as GADD45B [83], ADAMTS4 [84] and MEPE [82].
Finally, the molecular mechanisms that are responsible for the greater incidence of OA in females are not known. Genes may operate differently in the two sexes, at different body sites and on different disease features within body sites [27]. Interestingly, of the relatively small number of differences between females and males in OA bone identified in this study, there were significant numbers of genes that were involved in both osteoclast (for example, ANXA2, GSN, ITGB2, FOXP1 and PDE4A) and osteoblast (LTF, DF and TGFB1) function and hence bone remodelling (Tables 5 and 6). Collectively, the differential expression of these genes is consistent with increased bone turnover in OA females compared with males, suggesting an OA disease mechanism and perhaps partly accounting for a greater incidence of OA in females than in males.
A number of the highest ranking differentially expressed genes between OA females and males include WNT signalling pathway components such as WNT5B and the transcription factor genes EAF2 and CTBP2. In addition MMP25 and ITGB2 are WNT target genes, suggesting a difference between females and males in WNT signalling that may have an impact on the OA bone microenvironment. There is also evidence of crosstalk between WNT and oestrogen signalling pathways via functional interaction between β-catenin and oestrogen receptor-α [85]. A number of the genes identified in our study, including WNT5B, ITGB2, GSN, VCAM1, LTF and DF, are affected by oestrogen, potentially providing a mechanism by which they are differentially expressed in females compared with males. Examples of sexual dimorphism in mammalian gene expression related to different responses to disease by females and males are beginning to be identified [86]. The differences in expression levels of WNT5B, ITGB2 and MMP25 detected between females and males in OA bone is of interest and marks these genes as good candidates for further investigation into the sex disparity in OA.
In this study we observed small gene expression ratios in both microarray and real-time PCR analyses. These are probably contributed by the complex mix of cells being assayed, along with the subtle changes to bone that are observed in OA distal to the affected joint, and the often slow, age-dependent onset of the disease. Furthermore, OA is a multifactorial, multigene disorder (and perhaps even a heterogenous group of disorders that lead to similar bone changes, cartilage degeneration and ultimately loss of joint function). Therefore, it is to be expected that many genes and small changes in the expression of these genes would be involved in OA pathogenesis. Microarray analysis is able to reliably detect small (<2-fold) changes that prove to be biologically relevant [87], and in our study we were able to confirm the large majority of the differentially expressed genes by real-time PCR analysis. Furthermore, the power of the microarray analysis approach lies in its ability to detect genome-wide, coordinated, or similarly regulated differential gene expression, pointing to perturbed signalling pathways and importantly downstream molecular processes. Our study has identified such relationships between commonly regulated target genes (via WNT and TGF-β/BMP signalling pathways) that play roles, in particular, in osteoblasts, osteocytes and osteoclasts, potentially influencing bone formation, mineralization and remodelling.

Conclusion

In conclusion, we identified altered gene expression in bone from individuals with primary hip OA at a site distal to the diseased joint. This information is of interest because it identifies genes that potentially play roles in systemic physiological bone turnover or in skeletal disease processes, and implicate altered WNT and TGF-β/BMP signalling in OA pathogenesis. Further work sampling from individuals with early OA will be required to determine whether the genes identified as differentially expressed in OA bone are causal or secondary to the altered bone seen in OA.

Acknowledgements

The authors thank the donors and donors' families for their kind donation of bone tissue used for this study. The authors thank the orthopaedic surgeons and nursing staff of The Department of Orthopaedics and Trauma in the Royal Adelaide Hospital for support and cooperation in the collection of femoral specimens and the mortuary staff of the Institute of Medical and Veterinary Science for the collection of autopsy specimens. Thanks to Julia Kuliwaba and Helen Tsangari for processing and preparation of RNA from a subset of the samples used in this work. This work was supported by the National Health and Medical Research Council (NHMRC). The Compugen human oligonucleotide library for the microarray was purchased with Australian Cancer Research Foundation (ACRF) funds through the AMF. We thank Ashley Connolly and Mark Van der Hoek from the AMF for their guidance with the microarray experiments. We thank Tina Vincent from Discipline of Orthopaedics and Trauma, University of Adelaide, for advice with real-time PCR experiments.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

NLF and DMF conceived the study. BH contributed to study design and performed the acquisition of the microarray and real-time PCR data. AT advised on the microarray experiment design and performed the statistical analyses of the microarray data. BH and AT analyzed and interpreted the data. BH, DMF and NLF prepared the manuscript. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Felson DT, Lawrence RC, Dieppe PA, Hirsch R, Helmick CG, Jordan JM, Kington RS, Lane NE, Nevitt MC, Zhang Y, et al: Osteoarthritis: new insights. Part 1: the disease and its risk factors. Ann Intern Med. 2000, 133: 635-646.PubMed Felson DT, Lawrence RC, Dieppe PA, Hirsch R, Helmick CG, Jordan JM, Kington RS, Lane NE, Nevitt MC, Zhang Y, et al: Osteoarthritis: new insights. Part 1: the disease and its risk factors. Ann Intern Med. 2000, 133: 635-646.PubMed
2.
Zurück zum Zitat Hough AJ: Pathology of osteoarthritis. Arthritis and Allied Conditions: a Text Book of Rheumatology. Edited by: Koopman WJ. 2001, Philadelphia, PA: Lippincott Williams and Wilkins, 2: 2167-2194. 14 Hough AJ: Pathology of osteoarthritis. Arthritis and Allied Conditions: a Text Book of Rheumatology. Edited by: Koopman WJ. 2001, Philadelphia, PA: Lippincott Williams and Wilkins, 2: 2167-2194. 14
3.
Zurück zum Zitat Li B, Aspden RM: Composition and mechanical properties of cancellous bone from the femoral head of patients with osteoporosis or osteoarthritis. J Bone Miner Res. 1997, 12: 641-651. 10.1359/jbmr.1997.12.4.641.PubMed Li B, Aspden RM: Composition and mechanical properties of cancellous bone from the femoral head of patients with osteoporosis or osteoarthritis. J Bone Miner Res. 1997, 12: 641-651. 10.1359/jbmr.1997.12.4.641.PubMed
4.
Zurück zum Zitat Mansell JP, Tarlton JF, Bailey AF: Biochemical evidence for altered subchondral bone collagen metabolism in osteoarthritis of the hip. Br J Rheumatol. 1997, 36: 16-19. 10.1093/rheumatology/36.1.16.PubMed Mansell JP, Tarlton JF, Bailey AF: Biochemical evidence for altered subchondral bone collagen metabolism in osteoarthritis of the hip. Br J Rheumatol. 1997, 36: 16-19. 10.1093/rheumatology/36.1.16.PubMed
5.
Zurück zum Zitat Carlson CS, Loeser RF, Jayo MJ, Weaver DS, Adams MR, Jerome CP: Osteoarthritis in cynomolgus macaques: a primate model of naturally occurring disease. J Orthop Res. 1994, 12: 331-339. 10.1002/jor.1100120305.PubMed Carlson CS, Loeser RF, Jayo MJ, Weaver DS, Adams MR, Jerome CP: Osteoarthritis in cynomolgus macaques: a primate model of naturally occurring disease. J Orthop Res. 1994, 12: 331-339. 10.1002/jor.1100120305.PubMed
6.
Zurück zum Zitat Watson PJ, Hall LD, Malcolm A, Tyler JA: Degenerative joint disease in the guinea pig. Use of magnetic resonance imaging to monitor progression of bone pathology. Arthritis Rheum. 1996, 39: 1327-1337. 10.1002/art.1780390810.PubMed Watson PJ, Hall LD, Malcolm A, Tyler JA: Degenerative joint disease in the guinea pig. Use of magnetic resonance imaging to monitor progression of bone pathology. Arthritis Rheum. 1996, 39: 1327-1337. 10.1002/art.1780390810.PubMed
7.
Zurück zum Zitat Hayami T, Pickarski M, Zhuo Y, Wesolowski GA, Rodan GA, Duong le T: Characterization of articular cartilage and subchondral bone changes in the rat anterior cruciate ligament transection and meniscectomized models of osteoarthritis. Bone. 2006, 38: 234-243. 10.1016/j.bone.2005.08.007.PubMed Hayami T, Pickarski M, Zhuo Y, Wesolowski GA, Rodan GA, Duong le T: Characterization of articular cartilage and subchondral bone changes in the rat anterior cruciate ligament transection and meniscectomized models of osteoarthritis. Bone. 2006, 38: 234-243. 10.1016/j.bone.2005.08.007.PubMed
8.
Zurück zum Zitat Hayami T, Pickarski M, Wesolowski GA, McLane J, Bone A, Destefano J, Rodan GA, Duong le T: The role of subchondral bone remodeling in osteoarthritis: reduction of cartilage degeneration and prevention of osteophyte formation by alendronate in the rat anterior cruciate ligament transection model. Arthritis Rheum. 2004, 50: 1193-1206. 10.1002/art.20124.PubMed Hayami T, Pickarski M, Wesolowski GA, McLane J, Bone A, Destefano J, Rodan GA, Duong le T: The role of subchondral bone remodeling in osteoarthritis: reduction of cartilage degeneration and prevention of osteophyte formation by alendronate in the rat anterior cruciate ligament transection model. Arthritis Rheum. 2004, 50: 1193-1206. 10.1002/art.20124.PubMed
9.
Zurück zum Zitat Behets C, Williams JM, Chappard D, Devogelaer JP, Manicourt DH: Effects of calcitonin on subchondral trabecular bone changes and on osteoarthritic cartilage lesions after acute anterior cruciate ligament deficiency. J Bone Miner Res. 2004, 19: 1821-1826. 10.1359/JBMR.040609.PubMed Behets C, Williams JM, Chappard D, Devogelaer JP, Manicourt DH: Effects of calcitonin on subchondral trabecular bone changes and on osteoarthritic cartilage lesions after acute anterior cruciate ligament deficiency. J Bone Miner Res. 2004, 19: 1821-1826. 10.1359/JBMR.040609.PubMed
10.
Zurück zum Zitat Spector TD, Conaghan PG, Buckland-Wright JC, Garnero P, Cline GA, Beary JF, Valent DJ, Meyer JM: Effect of risedronate on joint structure and symptoms of knee osteoarthritis: results of the BRISK randomized, controlled trial [ISRCTN01928173]. Arthritis Res Ther. 2005, 7: R625-R633. 10.1186/ar1716.PubMedCentralPubMed Spector TD, Conaghan PG, Buckland-Wright JC, Garnero P, Cline GA, Beary JF, Valent DJ, Meyer JM: Effect of risedronate on joint structure and symptoms of knee osteoarthritis: results of the BRISK randomized, controlled trial [ISRCTN01928173]. Arthritis Res Ther. 2005, 7: R625-R633. 10.1186/ar1716.PubMedCentralPubMed
11.
Zurück zum Zitat Dequeker J, Luyten FP: Bone mass and osteoarthritis. Clin Exp Rheumatol. 2000, 18: S21-S26. Dequeker J, Luyten FP: Bone mass and osteoarthritis. Clin Exp Rheumatol. 2000, 18: S21-S26.
12.
Zurück zum Zitat Radin EL, Rose RM: Role of subchondral bone in the initiation and progression of cartilage damage. Clin Orthop Relat Res. 1986, 213: 34-40.PubMed Radin EL, Rose RM: Role of subchondral bone in the initiation and progression of cartilage damage. Clin Orthop Relat Res. 1986, 213: 34-40.PubMed
13.
Zurück zum Zitat Bailey AJ, Buckland-Wright C, Metz D: The role of bone in osteoarthritis. Age Ageing. 2001, 30: 374-378. 10.1093/ageing/30.5.374.PubMed Bailey AJ, Buckland-Wright C, Metz D: The role of bone in osteoarthritis. Age Ageing. 2001, 30: 374-378. 10.1093/ageing/30.5.374.PubMed
14.
Zurück zum Zitat Lajeunesse D: The role of bone in the treatment of osteoarthritis. Osteoarthritis Cartilage. 2004, 12: S34-S38. 10.1016/j.joca.2003.09.013.PubMed Lajeunesse D: The role of bone in the treatment of osteoarthritis. Osteoarthritis Cartilage. 2004, 12: S34-S38. 10.1016/j.joca.2003.09.013.PubMed
15.
Zurück zum Zitat Westacott CI, Webb GR, Warnock MG, Sims JV, Elson CJ: Alteration of cartilage metabolism by cells from osteoarthritic bone. Arthritis Rheum. 1997, 40: 1282-1291.PubMed Westacott CI, Webb GR, Warnock MG, Sims JV, Elson CJ: Alteration of cartilage metabolism by cells from osteoarthritic bone. Arthritis Rheum. 1997, 40: 1282-1291.PubMed
16.
Zurück zum Zitat Sanchez C, Deberg MA, Piccardi N, Msika P, Reginster JY, Henrotin YE: Osteoblasts from the sclerotic subchondral bone downregulate aggrecan but upregulate metalloproteinases expression by chondrocytes. This effect is mimicked by interleukin-6, -1beta and oncostatin M pre-treated non-sclerotic osteoblasts. Osteoarthritis Cartilage. 2005, 13: 979-987. 10.1016/j.joca.2005.03.008.PubMed Sanchez C, Deberg MA, Piccardi N, Msika P, Reginster JY, Henrotin YE: Osteoblasts from the sclerotic subchondral bone downregulate aggrecan but upregulate metalloproteinases expression by chondrocytes. This effect is mimicked by interleukin-6, -1beta and oncostatin M pre-treated non-sclerotic osteoblasts. Osteoarthritis Cartilage. 2005, 13: 979-987. 10.1016/j.joca.2005.03.008.PubMed
17.
Zurück zum Zitat Lyons TJ, McClure SF, Stoddart RW, McClure J: The normal human chondro-osseous junctional region: evidence for contact of uncalcified cartilage with subchondral bone and marrow spaces. BMC Musculoskelet Disord. 2006, 7: 52-10.1186/1471-2474-7-52.PubMedCentralPubMed Lyons TJ, McClure SF, Stoddart RW, McClure J: The normal human chondro-osseous junctional region: evidence for contact of uncalcified cartilage with subchondral bone and marrow spaces. BMC Musculoskelet Disord. 2006, 7: 52-10.1186/1471-2474-7-52.PubMedCentralPubMed
18.
Zurück zum Zitat Imhof H, Sulzbacher I, Grampp S, Czerny C, Youssefzadeh S, Kainberger F: Subchondral bone and cartilage disease: a rediscovered functional unit. Invest Radiol. 2000, 35: 581-588. 10.1097/00004424-200010000-00004.PubMed Imhof H, Sulzbacher I, Grampp S, Czerny C, Youssefzadeh S, Kainberger F: Subchondral bone and cartilage disease: a rediscovered functional unit. Invest Radiol. 2000, 35: 581-588. 10.1097/00004424-200010000-00004.PubMed
19.
Zurück zum Zitat Gerber HP, Vu TH, Ryan AM, Kowalski J, Werb Z, Ferrara N: VEGF couples hypertrophic cartilage remodeling, ossification and angiogenesis during endochondral bone formation. Nat Med. 1999, 5: 623-628. 10.1038/9467.PubMed Gerber HP, Vu TH, Ryan AM, Kowalski J, Werb Z, Ferrara N: VEGF couples hypertrophic cartilage remodeling, ossification and angiogenesis during endochondral bone formation. Nat Med. 1999, 5: 623-628. 10.1038/9467.PubMed
20.
Zurück zum Zitat Fang TD, Salim A, Xia W, Nacamuli RP, Guccione S, Song HM, Carano RA, Filvaroff EH, Bednarski MD, Giaccia AJ, et al: Angiogenesis is required for successful bone induction during distraction osteogenesis. J Bone Miner Res. 2005, 20: 1114-1124. 10.1359/JBMR.050301.PubMed Fang TD, Salim A, Xia W, Nacamuli RP, Guccione S, Song HM, Carano RA, Filvaroff EH, Bednarski MD, Giaccia AJ, et al: Angiogenesis is required for successful bone induction during distraction osteogenesis. J Bone Miner Res. 2005, 20: 1114-1124. 10.1359/JBMR.050301.PubMed
21.
Zurück zum Zitat Meury T, Verrier S, Alini M: Human endothelial cells inhibit BMSC differentiation into mature osteoblasts in vitro by interfering with osterix expression. J Cell Biochem. 2006, 98: 992-1006. 10.1002/jcb.20818.PubMed Meury T, Verrier S, Alini M: Human endothelial cells inhibit BMSC differentiation into mature osteoblasts in vitro by interfering with osterix expression. J Cell Biochem. 2006, 98: 992-1006. 10.1002/jcb.20818.PubMed
22.
Zurück zum Zitat Gevers G, Dequeker J, Martens M, Van Audekercke R, Nyssen-Behets C, Dhem A: Biomechanical characteristics of iliac crest bone in elderly women according to osteoarthritis grade at the hand joints. J Rheumatol. 1989, 16: 660-663.PubMed Gevers G, Dequeker J, Martens M, Van Audekercke R, Nyssen-Behets C, Dhem A: Biomechanical characteristics of iliac crest bone in elderly women according to osteoarthritis grade at the hand joints. J Rheumatol. 1989, 16: 660-663.PubMed
23.
Zurück zum Zitat Fazzalari NL, Vernon-Roberts B, Manthey BA, Parkinson IH: Relationship between changes in articular cartilage and bone in the femoral head in osteoarthritis of the hip. J Orthopaed Rheumatol. 1990, 3: 155-169. Fazzalari NL, Vernon-Roberts B, Manthey BA, Parkinson IH: Relationship between changes in articular cartilage and bone in the femoral head in osteoarthritis of the hip. J Orthopaed Rheumatol. 1990, 3: 155-169.
24.
Zurück zum Zitat Fazzalari NL, Parkinson IH: Femoral trabecular bone of osteoarthritic and normal subjects in an age and sex matched group. Osteoarthritis Cartilage. 1998, 6: 377-382. 10.1053/joca.1998.0141.PubMed Fazzalari NL, Parkinson IH: Femoral trabecular bone of osteoarthritic and normal subjects in an age and sex matched group. Osteoarthritis Cartilage. 1998, 6: 377-382. 10.1053/joca.1998.0141.PubMed
25.
Zurück zum Zitat Nevitt MC, Lane NE, Scott JC, Hochberg MC, Pressman AR, Genant HK, Cummings SR: Radiographic osteoarthritis of the hip and bone mineral density. The Study of Osteoporotic Fractures Research Group. Arthritis Rheum. 1995, 38: 907-916. 10.1002/art.1780380706.PubMed Nevitt MC, Lane NE, Scott JC, Hochberg MC, Pressman AR, Genant HK, Cummings SR: Radiographic osteoarthritis of the hip and bone mineral density. The Study of Osteoporotic Fractures Research Group. Arthritis Rheum. 1995, 38: 907-916. 10.1002/art.1780380706.PubMed
26.
Zurück zum Zitat Dequeker J, Boonen S, Aerssens J, Westhovens R: Inverse relationship osteoarthritis-osteoporosis: what is the evidence? What are the consequences?. Br J Rheumatol. 1996, 35: 813-818. 10.1093/rheumatology/35.9.813.PubMed Dequeker J, Boonen S, Aerssens J, Westhovens R: Inverse relationship osteoarthritis-osteoporosis: what is the evidence? What are the consequences?. Br J Rheumatol. 1996, 35: 813-818. 10.1093/rheumatology/35.9.813.PubMed
27.
Zurück zum Zitat Spector TD, MacGregor AJ: Risk factors for osteoarthritis: genetics. Osteoarthritis Cartilage. 2004, 12: S39-S44. 10.1016/j.joca.2003.09.005.PubMed Spector TD, MacGregor AJ: Risk factors for osteoarthritis: genetics. Osteoarthritis Cartilage. 2004, 12: S39-S44. 10.1016/j.joca.2003.09.005.PubMed
28.
Zurück zum Zitat Brandi ML, Gennari L, Cerinic MM, Becherini L, Falchetti A, Masi L, Gennari C, Reginster JY: Genetic markers of osteoarticular disorders: facts and hopes. Arthritis Res. 2001, 3: 270-280. 10.1186/ar316.PubMedCentralPubMed Brandi ML, Gennari L, Cerinic MM, Becherini L, Falchetti A, Masi L, Gennari C, Reginster JY: Genetic markers of osteoarticular disorders: facts and hopes. Arthritis Res. 2001, 3: 270-280. 10.1186/ar316.PubMedCentralPubMed
29.
Zurück zum Zitat Peach CA, Carr AJ, Loughlin J: Recent advances in the genetic investigation of osteoarthritis. Trends Mol Med. 2005, 11: 186-191. 10.1016/j.molmed.2005.02.005.PubMed Peach CA, Carr AJ, Loughlin J: Recent advances in the genetic investigation of osteoarthritis. Trends Mol Med. 2005, 11: 186-191. 10.1016/j.molmed.2005.02.005.PubMed
30.
Zurück zum Zitat Loughlin J, Dowling B, Chapman K, Marcelline L, Mustafa Z, Southam L, Ferreira A, Ciesielski C, Carson DA, Corr M: Functional variants within the secreted frizzled-related protein 3 gene are associated with hip osteoarthritis in females. Proc Natl Acad Sci USA. 2004, 101: 9757-9762. 10.1073/pnas.0403456101.PubMedCentralPubMed Loughlin J, Dowling B, Chapman K, Marcelline L, Mustafa Z, Southam L, Ferreira A, Ciesielski C, Carson DA, Corr M: Functional variants within the secreted frizzled-related protein 3 gene are associated with hip osteoarthritis in females. Proc Natl Acad Sci USA. 2004, 101: 9757-9762. 10.1073/pnas.0403456101.PubMedCentralPubMed
31.
Zurück zum Zitat Chung YS, Baylink DJ, Srivastava AK, Amaar Y, Tapia B, Kasukawa Y, Mohan S: Effects of secreted frizzled-related protein 3 on osteoblasts in vitro. J Bone Miner Res. 2004, 19: 1395-1402. 10.1359/JBMR.040412.PubMed Chung YS, Baylink DJ, Srivastava AK, Amaar Y, Tapia B, Kasukawa Y, Mohan S: Effects of secreted frizzled-related protein 3 on osteoblasts in vitro. J Bone Miner Res. 2004, 19: 1395-1402. 10.1359/JBMR.040412.PubMed
32.
Zurück zum Zitat Krishnan V, Bryant HU, Macdougald OA: Regulation of bone mass by Wnt signaling. J Clin Invest. 2006, 116: 1202-1209. 10.1172/JCI28551.PubMedCentralPubMed Krishnan V, Bryant HU, Macdougald OA: Regulation of bone mass by Wnt signaling. J Clin Invest. 2006, 116: 1202-1209. 10.1172/JCI28551.PubMedCentralPubMed
33.
Zurück zum Zitat Canalis E, Economides AN, Gazzerro E: Bone morphogenetic proteins, their antagonists, and the skeleton. Endocr Rev. 2003, 24: 218-235. 10.1210/er.2002-0023.PubMed Canalis E, Economides AN, Gazzerro E: Bone morphogenetic proteins, their antagonists, and the skeleton. Endocr Rev. 2003, 24: 218-235. 10.1210/er.2002-0023.PubMed
34.
Zurück zum Zitat Janssens K, ten Dijke P, Janssens S, Van Hul W: Transforming growth factor-beta1 to the bone. Endocr Rev. 2005, 26: 743-774. 10.1210/er.2004-0001.PubMed Janssens K, ten Dijke P, Janssens S, Van Hul W: Transforming growth factor-beta1 to the bone. Endocr Rev. 2005, 26: 743-774. 10.1210/er.2004-0001.PubMed
35.
Zurück zum Zitat Kuliwaba JS, Findlay DM, Atkins GJ, Forwood MR, Fazzalari NL: Enhanced expression of osteocalcin mRNA in human osteoarthritic trabecular bone of the proximal femur is associated with decreased expression of interleukin-6 and interleukin-11 mRNA. J Bone Miner Res. 2000, 15: 332-341. 10.1359/jbmr.2000.15.2.332.PubMed Kuliwaba JS, Findlay DM, Atkins GJ, Forwood MR, Fazzalari NL: Enhanced expression of osteocalcin mRNA in human osteoarthritic trabecular bone of the proximal femur is associated with decreased expression of interleukin-6 and interleukin-11 mRNA. J Bone Miner Res. 2000, 15: 332-341. 10.1359/jbmr.2000.15.2.332.PubMed
36.
Zurück zum Zitat Fazzalari NL, Kuliwaba JS, Atkins GJ, Forwood MR, Findlay DM: The ratio of messenger RNA levels of receptor activator of nuclear factor kappaB ligand to osteoprotegerin correlates with bone remodeling indices in normal human cancellous bone but not in osteoarthritis. J Bone Miner Res. 2001, 16: 1015-1027. 10.1359/jbmr.2001.16.6.1015.PubMed Fazzalari NL, Kuliwaba JS, Atkins GJ, Forwood MR, Findlay DM: The ratio of messenger RNA levels of receptor activator of nuclear factor kappaB ligand to osteoprotegerin correlates with bone remodeling indices in normal human cancellous bone but not in osteoarthritis. J Bone Miner Res. 2001, 16: 1015-1027. 10.1359/jbmr.2001.16.6.1015.PubMed
37.
Zurück zum Zitat Truong LH, Kuliwaba JS, Tsangari H, Fazzalari NL: Differential gene expression of bone anabolic factors and trabecular bone architectural changes in the proximal femoral shaft of primary hip osteoarthritis patients. Arthritis Res Ther. 2006, 8: R188-10.1186/ar2101.PubMedCentralPubMed Truong LH, Kuliwaba JS, Tsangari H, Fazzalari NL: Differential gene expression of bone anabolic factors and trabecular bone architectural changes in the proximal femoral shaft of primary hip osteoarthritis patients. Arthritis Res Ther. 2006, 8: R188-10.1186/ar2101.PubMedCentralPubMed
38.
Zurück zum Zitat Dequeker J, Mohan S, Finkelman RD, Aerssens J, Baylink DJ: Generalized osteoarthritis associated with increased insulin-like growth factor types I and II and transforming growth factor beta in cortical bone from the iliac crest. Possible mechanism of increased bone density and protection against osteoporosis. Arthritis Rheum. 1993, 36: 1702-1708. 10.1002/art.1780361209.PubMed Dequeker J, Mohan S, Finkelman RD, Aerssens J, Baylink DJ: Generalized osteoarthritis associated with increased insulin-like growth factor types I and II and transforming growth factor beta in cortical bone from the iliac crest. Possible mechanism of increased bone density and protection against osteoporosis. Arthritis Rheum. 1993, 36: 1702-1708. 10.1002/art.1780361209.PubMed
39.
Zurück zum Zitat Massicotte F, Lajeunesse D, Benderdour M, Pelletier JP, Hilal G, Duval N, Martel-Pelletier J: Can altered production of interleukin-1beta, interleukin-6, transforming growth factor-beta and prostaglandin E2 by isolated human subchondral osteoblasts identify two subgroups of osteoarthritic patients. Osteoarthritis Cartilage. 2002, 10: 491-500. 10.1053/joca.2002.0528.PubMed Massicotte F, Lajeunesse D, Benderdour M, Pelletier JP, Hilal G, Duval N, Martel-Pelletier J: Can altered production of interleukin-1beta, interleukin-6, transforming growth factor-beta and prostaglandin E2 by isolated human subchondral osteoblasts identify two subgroups of osteoarthritic patients. Osteoarthritis Cartilage. 2002, 10: 491-500. 10.1053/joca.2002.0528.PubMed
40.
Zurück zum Zitat Collins D: The Pathology of Articular and Spinal Diseases. 1949, London, UK: Edward Arnold and Co Collins D: The Pathology of Articular and Spinal Diseases. 1949, London, UK: Edward Arnold and Co
41.
Zurück zum Zitat Kuliwaba JS, Fazzalari NL, Findlay DM: Stability of RNA isolated from human trabecular bone at post-mortem and surgery. Biochim Biophys Acta. 2005, 1740: 1-11.PubMed Kuliwaba JS, Fazzalari NL, Findlay DM: Stability of RNA isolated from human trabecular bone at post-mortem and surgery. Biochim Biophys Acta. 2005, 1740: 1-11.PubMed
45.
Zurück zum Zitat Yang YH, Buckley MJ, Speed TP: Analysis of cDNA microarray images. Brief Bioinform. 2001, 2: 341-349. 10.1093/bib/2.4.341.PubMed Yang YH, Buckley MJ, Speed TP: Analysis of cDNA microarray images. Brief Bioinform. 2001, 2: 341-349. 10.1093/bib/2.4.341.PubMed
46.
Zurück zum Zitat Ritchie ME: Quantitative quality control and background correction for two-colour microarray data. PhD thesis. 2004, University of Melbourne, Department of Medical Biology Ritchie ME: Quantitative quality control and background correction for two-colour microarray data. PhD thesis. 2004, University of Melbourne, Department of Medical Biology
47.
Zurück zum Zitat 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. 2004, 5: R80-10.1186/gb-2004-5-10-r80.PubMedCentralPubMed 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. 2004, 5: R80-10.1186/gb-2004-5-10-r80.PubMedCentralPubMed
48.
Zurück zum Zitat Yang YH, Dudoit S, Luu P, Lin DM, Peng V, Ngai J, Speed TP: Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res. 2002, 30: e15-10.1093/nar/30.4.e15.PubMedCentralPubMed Yang YH, Dudoit S, Luu P, Lin DM, Peng V, Ngai J, Speed TP: Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res. 2002, 30: e15-10.1093/nar/30.4.e15.PubMedCentralPubMed
49.
Zurück zum Zitat Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004, 3: Article 3 Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004, 3: Article 3
51.
Zurück zum Zitat KJ Livak, TD Schmittgen: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta CT) Method. Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262. KJ Livak, TD Schmittgen: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta CT) Method. Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262.
53.
Zurück zum Zitat Draghici S, Khatri P, Bhavsar B, Shah A, Krawetz SA, Tainsky MA: Onto-Tools, the toolkit of the modern biologist: Onto-Express, Onto-Compare, Onto-Design and Onto-Translate. Nucleic Acids Res. 2003, 31: 3775-3781. 10.1093/nar/gkg624.PubMedCentralPubMed Draghici S, Khatri P, Bhavsar B, Shah A, Krawetz SA, Tainsky MA: Onto-Tools, the toolkit of the modern biologist: Onto-Express, Onto-Compare, Onto-Design and Onto-Translate. Nucleic Acids Res. 2003, 31: 3775-3781. 10.1093/nar/gkg624.PubMedCentralPubMed
54.
Zurück zum Zitat Beissbarth T, Speed TP: GOstat: find statistically overrepresented Gene Ontologies within a group of genes. Bioinformatics. 2004, 20: 1464-1465. 10.1093/bioinformatics/bth088.PubMed Beissbarth T, Speed TP: GOstat: find statistically overrepresented Gene Ontologies within a group of genes. Bioinformatics. 2004, 20: 1464-1465. 10.1093/bioinformatics/bth088.PubMed
55.
Zurück zum Zitat Dallas PB, Gottardo NG, Firth MJ, Beesley AH, Hoffmann K, Terry PA, Freitas JR, Boag JM, Cummings AJ, Kees UR: Gene expression levels assessed by oligonucleotide microarray analysis and quantitative real-time RT-PCR: how well do they correlate?. BMC Genomics. 2005, 6: 59-10.1186/1471-2164-6-59.PubMedCentralPubMed Dallas PB, Gottardo NG, Firth MJ, Beesley AH, Hoffmann K, Terry PA, Freitas JR, Boag JM, Cummings AJ, Kees UR: Gene expression levels assessed by oligonucleotide microarray analysis and quantitative real-time RT-PCR: how well do they correlate?. BMC Genomics. 2005, 6: 59-10.1186/1471-2164-6-59.PubMedCentralPubMed
56.
Zurück zum Zitat Wang Y, Barbacioru C, Hyland F, Xiao W, Hunkapiller KL, Blake J, Chan F, Gonzalez C, Zhang L, Samaha RR: Large scale real-time PCR validation on gene expression measurements from two commercial long-oligonucleotide microarrays. BMC Genomics. 2006, 7: 59-10.1186/1471-2164-7-59.PubMedCentralPubMed Wang Y, Barbacioru C, Hyland F, Xiao W, Hunkapiller KL, Blake J, Chan F, Gonzalez C, Zhang L, Samaha RR: Large scale real-time PCR validation on gene expression measurements from two commercial long-oligonucleotide microarrays. BMC Genomics. 2006, 7: 59-10.1186/1471-2164-7-59.PubMedCentralPubMed
57.
Zurück zum Zitat Franz-Odendaal TA, Hall BK, Witten PE: Buried alive: how osteoblasts become osteocytes. Dev Dyn. 2006, 235: 176-190. 10.1002/dvdy.20603.PubMed Franz-Odendaal TA, Hall BK, Witten PE: Buried alive: how osteoblasts become osteocytes. Dev Dyn. 2006, 235: 176-190. 10.1002/dvdy.20603.PubMed
58.
Zurück zum Zitat Gowen LC, Petersen DN, Mansolf AL, Qi H, Stock JL, Tkalcevic GT, Simmons HA, Crawford DT, Chidsey-Frink KL, Ke HZ, et al: Targeted disruption of the osteoblast/osteocyte factor 45 gene (OF45) results in increased bone formation and bone mass. J Biol Chem. 2003, 278: 1998-2007. 10.1074/jbc.M203250200.PubMed Gowen LC, Petersen DN, Mansolf AL, Qi H, Stock JL, Tkalcevic GT, Simmons HA, Crawford DT, Chidsey-Frink KL, Ke HZ, et al: Targeted disruption of the osteoblast/osteocyte factor 45 gene (OF45) results in increased bone formation and bone mass. J Biol Chem. 2003, 278: 1998-2007. 10.1074/jbc.M203250200.PubMed
59.
Zurück zum Zitat Nie J, Pei D: Direct activation of pro-matrix metalloproteinase-2 by leukolysin/membrane-type 6 matrix metalloproteinase/matrix metalloproteinase 25 at the asn(109)-Tyr bond. Cancer Res. 2003, 63: 6758-6762.PubMed Nie J, Pei D: Direct activation of pro-matrix metalloproteinase-2 by leukolysin/membrane-type 6 matrix metalloproteinase/matrix metalloproteinase 25 at the asn(109)-Tyr bond. Cancer Res. 2003, 63: 6758-6762.PubMed
60.
Zurück zum Zitat Inoue K, Mikuni-Takagaki Y, Oikawa K, Itoh T, Inada M, Noguchi T, Park JS, Onodera T, Krane SM, Noda M, et al: A crucial role for matrix metalloproteinase 2 in osteocytic canalicular formation and bone metabolism. J Biol Chem. 2006, 281: 33814-33824. 10.1074/jbc.M607290200.PubMed Inoue K, Mikuni-Takagaki Y, Oikawa K, Itoh T, Inada M, Noguchi T, Park JS, Onodera T, Krane SM, Noda M, et al: A crucial role for matrix metalloproteinase 2 in osteocytic canalicular formation and bone metabolism. J Biol Chem. 2006, 281: 33814-33824. 10.1074/jbc.M607290200.PubMed
61.
Zurück zum Zitat Atkins GJ, Kostakis P, Welldon KJ, Vincent C, Findlay DM, Zannettino AC: Human trabecular bone-derived osteoblasts support human osteoclast formation in vitro in a defined, serum-free medium. J Cell Physiol. 2005, 203: 573-582. 10.1002/jcp.20255.PubMed Atkins GJ, Kostakis P, Welldon KJ, Vincent C, Findlay DM, Zannettino AC: Human trabecular bone-derived osteoblasts support human osteoclast formation in vitro in a defined, serum-free medium. J Cell Physiol. 2005, 203: 573-582. 10.1002/jcp.20255.PubMed
62.
Zurück zum Zitat Kim HJ, Zhao H, Kitaura H, Bhattacharyya S, Brewer JA, Muglia LJ, Ross FP, Teitelbaum SL: Glucocorticoids suppress bone formation via the osteoclast. J Clin Invest. 2006, 116: 2152-2160. 10.1172/JCI28084.PubMedCentralPubMed Kim HJ, Zhao H, Kitaura H, Bhattacharyya S, Brewer JA, Muglia LJ, Ross FP, Teitelbaum SL: Glucocorticoids suppress bone formation via the osteoclast. J Clin Invest. 2006, 116: 2152-2160. 10.1172/JCI28084.PubMedCentralPubMed
63.
Zurück zum Zitat Karsdal MA, Martin TJ, Bollerslev J, Christiansen C, Henriksen K: Are nonresorbing osteoclasts sources of bone anabolic activity?. J Bone Miner Res. 2007, 22: 487-494. 10.1359/jbmr.070109.PubMed Karsdal MA, Martin TJ, Bollerslev J, Christiansen C, Henriksen K: Are nonresorbing osteoclasts sources of bone anabolic activity?. J Bone Miner Res. 2007, 22: 487-494. 10.1359/jbmr.070109.PubMed
64.
Zurück zum Zitat Cornish J, Naot D: Amylin and adrenomedullin: novel regulators of bone growth. Curr Pharm Des. 2002, 8: 2009-2021. 10.2174/1381612023393341.PubMed Cornish J, Naot D: Amylin and adrenomedullin: novel regulators of bone growth. Curr Pharm Des. 2002, 8: 2009-2021. 10.2174/1381612023393341.PubMed
65.
Zurück zum Zitat Glass DA, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, Taketo MM, Long F, McMahon AP, Lang RA, et al: Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005, 8: 751-764. 10.1016/j.devcel.2005.02.017.PubMed Glass DA, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, Taketo MM, Long F, McMahon AP, Lang RA, et al: Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005, 8: 751-764. 10.1016/j.devcel.2005.02.017.PubMed
66.
Zurück zum Zitat Alliston T, Choy L, Ducy P, Karsenty G, Derynck R: TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. Embo J. 2001, 20: 2254-2272. 10.1093/emboj/20.9.2254.PubMedCentralPubMed Alliston T, Choy L, Ducy P, Karsenty G, Derynck R: TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. Embo J. 2001, 20: 2254-2272. 10.1093/emboj/20.9.2254.PubMedCentralPubMed
67.
Zurück zum Zitat Karst M, Gorny G, Galvin RJ, Oursler MJ: Roles of stromal cell RANKL, OPG, and M-CSF expression in biphasic TGF-beta regulation of osteoclast differentiation. J Cell Physiol. 2004, 200: 99-106. 10.1002/jcp.20036.PubMedCentralPubMed Karst M, Gorny G, Galvin RJ, Oursler MJ: Roles of stromal cell RANKL, OPG, and M-CSF expression in biphasic TGF-beta regulation of osteoclast differentiation. J Cell Physiol. 2004, 200: 99-106. 10.1002/jcp.20036.PubMedCentralPubMed
68.
Zurück zum Zitat Hurson CJ, Butler JS, Keating DT, Murray DW, Sadlier DM, O'Byrne JM, Doran PP: Gene expression analysis in human osteoblasts exposed to dexamethasone identifies altered developmental pathways as putative drivers of osteoporosis. BMC Musculoskelet Disord. 2007, 8: 12-10.1186/1471-2474-8-12.PubMedCentralPubMed Hurson CJ, Butler JS, Keating DT, Murray DW, Sadlier DM, O'Byrne JM, Doran PP: Gene expression analysis in human osteoblasts exposed to dexamethasone identifies altered developmental pathways as putative drivers of osteoporosis. BMC Musculoskelet Disord. 2007, 8: 12-10.1186/1471-2474-8-12.PubMedCentralPubMed
69.
Zurück zum Zitat Church V, Nohno T, Linker C, Marcelle C, Francis-West P: Wnt regulation of chondrocyte differentiation. J Cell Sci. 2002, 115: 4809-4818. 10.1242/jcs.00152.PubMed Church V, Nohno T, Linker C, Marcelle C, Francis-West P: Wnt regulation of chondrocyte differentiation. J Cell Sci. 2002, 115: 4809-4818. 10.1242/jcs.00152.PubMed
70.
Zurück zum Zitat Shea CM, Edgar CM, Einhorn TA, Gerstenfeld LC: BMP treatment of C3H10T1/2 mesenchymal stem cells induces both chondrogenesis and osteogenesis. J Cell Biochem. 2003, 90: 1112-1127. 10.1002/jcb.10734.PubMed Shea CM, Edgar CM, Einhorn TA, Gerstenfeld LC: BMP treatment of C3H10T1/2 mesenchymal stem cells induces both chondrogenesis and osteogenesis. J Cell Biochem. 2003, 90: 1112-1127. 10.1002/jcb.10734.PubMed
71.
Zurück zum Zitat Logan CY, Nusse R: The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004, 20: 781-810. 10.1146/annurev.cellbio.20.010403.113126.PubMed Logan CY, Nusse R: The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004, 20: 781-810. 10.1146/annurev.cellbio.20.010403.113126.PubMed
72.
Zurück zum Zitat Jian H, Shen X, Liu I, Semenov M, He X, Wang XF: Smad3-dependent nuclear translocation of beta-catenin is required for TGF-beta1-induced proliferation of bone marrow-derived adult human mesenchymal stem cells. Genes Dev. 2006, 20: 666-674. 10.1101/gad.1388806.PubMedCentralPubMed Jian H, Shen X, Liu I, Semenov M, He X, Wang XF: Smad3-dependent nuclear translocation of beta-catenin is required for TGF-beta1-induced proliferation of bone marrow-derived adult human mesenchymal stem cells. Genes Dev. 2006, 20: 666-674. 10.1101/gad.1388806.PubMedCentralPubMed
73.
Zurück zum Zitat Reinhold MI, Naski MC: Direct interactions of Runx2 and canonical Wnt signaling induce FGF18. J Biol Chem. 2007, 282: 3653-3663. 10.1074/jbc.M608995200.PubMed Reinhold MI, Naski MC: Direct interactions of Runx2 and canonical Wnt signaling induce FGF18. J Biol Chem. 2007, 282: 3653-3663. 10.1074/jbc.M608995200.PubMed
74.
Zurück zum Zitat Labbe E, Letamendia A, Attisano L: Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathways. Proc Natl Acad Sci USA. 2000, 97: 8358-8363. 10.1073/pnas.150152697.PubMedCentralPubMed Labbe E, Letamendia A, Attisano L: Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathways. Proc Natl Acad Sci USA. 2000, 97: 8358-8363. 10.1073/pnas.150152697.PubMedCentralPubMed
75.
Zurück zum Zitat Kahler RA, Westendorf JJ: Lymphoid enhancer factor-1 and beta-catenin inhibit Runx2-dependent transcriptional activation of the osteocalcin promoter. J Biol Chem. 2003, 278: 11937-11944. 10.1074/jbc.M211443200.PubMed Kahler RA, Westendorf JJ: Lymphoid enhancer factor-1 and beta-catenin inhibit Runx2-dependent transcriptional activation of the osteocalcin promoter. J Biol Chem. 2003, 278: 11937-11944. 10.1074/jbc.M211443200.PubMed
76.
Zurück zum Zitat Komori T: Regulation of osteoblast differentiation by transcription factors. J Cell Biochem. 2006, 99: 1233-1239. 10.1002/jcb.20958.PubMed Komori T: Regulation of osteoblast differentiation by transcription factors. J Cell Biochem. 2006, 99: 1233-1239. 10.1002/jcb.20958.PubMed
77.
Zurück zum Zitat Gaur T, Lengner CJ, Hovhannisyan H, Bhat RA, Bodine PV, Komm BS, Javed A, van Wijnen AJ, Stein JL, Stein GS, et al: Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem. 2005, 280: 33132-33140. 10.1074/jbc.M500608200.PubMed Gaur T, Lengner CJ, Hovhannisyan H, Bhat RA, Bodine PV, Komm BS, Javed A, van Wijnen AJ, Stein JL, Stein GS, et al: Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem. 2005, 280: 33132-33140. 10.1074/jbc.M500608200.PubMed
78.
Zurück zum Zitat Kamekura S, Kawasaki Y, Hoshi K, Shimoaka T, Chikuda H, Maruyama Z, Komori T, Sato S, Takeda S, Karsenty G, et al: Contribution of runt-related transcription factor 2 to the pathogenesis of osteoarthritis in mice after induction of knee joint instability. Arthritis Rheum. 2006, 54: 2462-2470. 10.1002/art.22041.PubMed Kamekura S, Kawasaki Y, Hoshi K, Shimoaka T, Chikuda H, Maruyama Z, Komori T, Sato S, Takeda S, Karsenty G, et al: Contribution of runt-related transcription factor 2 to the pathogenesis of osteoarthritis in mice after induction of knee joint instability. Arthritis Rheum. 2006, 54: 2462-2470. 10.1002/art.22041.PubMed
79.
Zurück zum Zitat Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, Wu H, Yu K, Ornitz DM, Olson EN, et al: A twist code determines the onset of osteoblast differentiation. Dev Cell. 2004, 6: 423-435. 10.1016/S1534-5807(04)00058-9.PubMed Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, Wu H, Yu K, Ornitz DM, Olson EN, et al: A twist code determines the onset of osteoblast differentiation. Dev Cell. 2004, 6: 423-435. 10.1016/S1534-5807(04)00058-9.PubMed
80.
Zurück zum Zitat Balooch G, Balooch M, Nalla RK, Schilling S, Filvaroff EH, Marshall GW, Marshall SJ, Ritchie RO, Derynck R, Alliston T: TGF-beta regulates the mechanical properties and composition of bone matrix. Proc Natl Acad Sci USA. 2005, 102: 18813-18818. 10.1073/pnas.0507417102.PubMedCentralPubMed Balooch G, Balooch M, Nalla RK, Schilling S, Filvaroff EH, Marshall GW, Marshall SJ, Ritchie RO, Derynck R, Alliston T: TGF-beta regulates the mechanical properties and composition of bone matrix. Proc Natl Acad Sci USA. 2005, 102: 18813-18818. 10.1073/pnas.0507417102.PubMedCentralPubMed
81.
Zurück zum Zitat Kang JS, Alliston T, Delston R, Derynck R: Repression of Runx2 function by TGF-beta through recruitment of class II histone deacetylases by Smad3. Embo J. 2005, 24: 2543-2555. 10.1038/sj.emboj.7600729.PubMedCentralPubMed Kang JS, Alliston T, Delston R, Derynck R: Repression of Runx2 function by TGF-beta through recruitment of class II histone deacetylases by Smad3. Embo J. 2005, 24: 2543-2555. 10.1038/sj.emboj.7600729.PubMedCentralPubMed
82.
Zurück zum Zitat Kaji H, Naito J, Sowa H, Sugimoto T, Chihara K: Smad3 differently affects osteoblast differentiation depending upon its differentiation stage. Horm Metab Res. 2006, 38: 740-745. 10.1055/s-2006-955085.PubMed Kaji H, Naito J, Sowa H, Sugimoto T, Chihara K: Smad3 differently affects osteoblast differentiation depending upon its differentiation stage. Horm Metab Res. 2006, 38: 740-745. 10.1055/s-2006-955085.PubMed
83.
Zurück zum Zitat Major MB, Jones DA: Identification of a gadd45beta 3' enhancer that mediates SMAD3- and SMAD4-dependent transcriptional induction by transforming growth factor beta. J Biol Chem. 2004, 279: 5278-5287. 10.1074/jbc.M311517200.PubMed Major MB, Jones DA: Identification of a gadd45beta 3' enhancer that mediates SMAD3- and SMAD4-dependent transcriptional induction by transforming growth factor beta. J Biol Chem. 2004, 279: 5278-5287. 10.1074/jbc.M311517200.PubMed
84.
Zurück zum Zitat Thirunavukkarasu K, Pei Y, Moore TL, Wang H, Yu XP, Geiser AG, Chandrasekhar S: Regulation of the human ADAMTS-4 promoter by transcription factors and cytokines. Biochem Biophys Res Commun. 2006, 345: 197-204. 10.1016/j.bbrc.2006.04.023.PubMed Thirunavukkarasu K, Pei Y, Moore TL, Wang H, Yu XP, Geiser AG, Chandrasekhar S: Regulation of the human ADAMTS-4 promoter by transcription factors and cytokines. Biochem Biophys Res Commun. 2006, 345: 197-204. 10.1016/j.bbrc.2006.04.023.PubMed
85.
Zurück zum Zitat Kouzmenko AP, Takeyama K, Ito S, Furutani T, Sawatsubashi S, Maki A, Suzuki E, Kawasaki Y, Akiyama T, Tabata T, et al: Wnt/beta-catenin and estrogen signaling converge in vivo. J Biol Chem. 2004, 279: 40255-40258. 10.1074/jbc.C400331200.PubMed Kouzmenko AP, Takeyama K, Ito S, Furutani T, Sawatsubashi S, Maki A, Suzuki E, Kawasaki Y, Akiyama T, Tabata T, et al: Wnt/beta-catenin and estrogen signaling converge in vivo. J Biol Chem. 2004, 279: 40255-40258. 10.1074/jbc.C400331200.PubMed
86.
Zurück zum Zitat Rinn JL, Snyder M: Sexual dimorphism in mammalian gene expression. Trends Genet. 2005, 21: 298-305. 10.1016/j.tig.2005.03.005.PubMed Rinn JL, Snyder M: Sexual dimorphism in mammalian gene expression. Trends Genet. 2005, 21: 298-305. 10.1016/j.tig.2005.03.005.PubMed
87.
Zurück zum Zitat Yao B, Rakhade RN, Li Q, Ahmed S, Krauss R, Draghici S, Loeb JA: Accuracy of cDNA microarray methods to detect small gene expression changes induced by neuregulin on breast epithelial cells. BMC Bioinformatics. 2004, 5: 99-10.1186/1471-2105-5-99.PubMedCentralPubMed Yao B, Rakhade RN, Li Q, Ahmed S, Krauss R, Draghici S, Loeb JA: Accuracy of cDNA microarray methods to detect small gene expression changes induced by neuregulin on breast epithelial cells. BMC Bioinformatics. 2004, 5: 99-10.1186/1471-2105-5-99.PubMedCentralPubMed
88.
Zurück zum Zitat Duarte WR, Shibata T, Takenaga K, Takahashi E, Kubota K, Ohya K, Ishikawa I, Yamauchi M, Kasugai S: S100A4: a novel negative regulator of mineralization and osteoblast differentiation. J Bone Miner Res. 2003, 18: 493-501. 10.1359/jbmr.2003.18.3.493.PubMed Duarte WR, Shibata T, Takenaga K, Takahashi E, Kubota K, Ohya K, Ishikawa I, Yamauchi M, Kasugai S: S100A4: a novel negative regulator of mineralization and osteoblast differentiation. J Bone Miner Res. 2003, 18: 493-501. 10.1359/jbmr.2003.18.3.493.PubMed
89.
Zurück zum Zitat Mathisen B, Lindstad RI, Hansen J, El-Gewely SA, Maelandsmo GM, Hovig E, Fodstad O, Loennechen T, Winberg JO: S100A4 regulates membrane induced activation of matrix metalloproteinase-2 in osteosarcoma cells. Clin Exp Metastasis. 2003, 20: 701-711. 10.1023/B:CLIN.0000006819.21361.03.PubMed Mathisen B, Lindstad RI, Hansen J, El-Gewely SA, Maelandsmo GM, Hovig E, Fodstad O, Loennechen T, Winberg JO: S100A4 regulates membrane induced activation of matrix metalloproteinase-2 in osteosarcoma cells. Clin Exp Metastasis. 2003, 20: 701-711. 10.1023/B:CLIN.0000006819.21361.03.PubMed
90.
Zurück zum Zitat Ogata Y, Niisato N, Furuyama S, Cheifetz S, Kim RH, Sugiya H, Sodek J: Transforming growth factor-beta 1 regulation of bone sialoprotein gene transcription: identification of a TGF-beta activation element in the rat BSP gene promoter. J Cell Biochem. 1997, 65: 501-512. 10.1002/(SICI)1097-4644(19970615)65:4<501::AID-JCB6>3.0.CO;2-S.PubMed Ogata Y, Niisato N, Furuyama S, Cheifetz S, Kim RH, Sugiya H, Sodek J: Transforming growth factor-beta 1 regulation of bone sialoprotein gene transcription: identification of a TGF-beta activation element in the rat BSP gene promoter. J Cell Biochem. 1997, 65: 501-512. 10.1002/(SICI)1097-4644(19970615)65:4<501::AID-JCB6>3.0.CO;2-S.PubMed
91.
Zurück zum Zitat Dohi Y, Shimaoka H, Ikeuchi M, Ohgushi H, Yonemasu K, Minami T: Role of metallothionein isoforms in bone formation processes in rat marrow mesenchymal stem cells in culture. Biol Trace Elem Res. 2005, 104: 57-70. 10.1385/BTER:104:1:057.PubMed Dohi Y, Shimaoka H, Ikeuchi M, Ohgushi H, Yonemasu K, Minami T: Role of metallothionein isoforms in bone formation processes in rat marrow mesenchymal stem cells in culture. Biol Trace Elem Res. 2005, 104: 57-70. 10.1385/BTER:104:1:057.PubMed
92.
Zurück zum Zitat Liu AL, Zhang ZM, Zhu BF, Liao ZH, Liu Z: Metallothionein protects bone marrow stromal cells against hydrogen peroxide-induced inhibition of osteoblastic differentiation. Cell Biol Int. 2004, 28: 905-911. 10.1016/j.cellbi.2004.09.004.PubMed Liu AL, Zhang ZM, Zhu BF, Liao ZH, Liu Z: Metallothionein protects bone marrow stromal cells against hydrogen peroxide-induced inhibition of osteoblastic differentiation. Cell Biol Int. 2004, 28: 905-911. 10.1016/j.cellbi.2004.09.004.PubMed
93.
Zurück zum Zitat Cornish J, Grey A, Callon KE, Naot D, Hill BL, Lin CQ, Balchin LM, Reid IR: Shared pathways of osteoblast mitogenesis induced by amylin, adrenomedullin, and IGF-1. Biochem Biophys Res Commun. 2004, 318: 240-246. 10.1016/j.bbrc.2004.04.020.PubMed Cornish J, Grey A, Callon KE, Naot D, Hill BL, Lin CQ, Balchin LM, Reid IR: Shared pathways of osteoblast mitogenesis induced by amylin, adrenomedullin, and IGF-1. Biochem Biophys Res Commun. 2004, 318: 240-246. 10.1016/j.bbrc.2004.04.020.PubMed
94.
Zurück zum Zitat Bodegas E, Martinez A, Ozbun LL, Garayoa M, Letterio JJ, Montuenga LM, Jakowlew SB: Depressed adrenomedullin in the embryonic transforming growth factor-beta1 null mouse becomes elevated postnatally. Int J Dev Biol. 2004, 48: 67-70. 10.1387/ijdb.15005577.PubMed Bodegas E, Martinez A, Ozbun LL, Garayoa M, Letterio JJ, Montuenga LM, Jakowlew SB: Depressed adrenomedullin in the embryonic transforming growth factor-beta1 null mouse becomes elevated postnatally. Int J Dev Biol. 2004, 48: 67-70. 10.1387/ijdb.15005577.PubMed
95.
Zurück zum Zitat Wu S, Yoshiko Y, De Luca F: Stanniocalcin 1 acts as a paracrine regulator of growth plate chondrogenesis. J Biol Chem. 2006, 281: 5120-5127. 10.1074/jbc.M506667200.PubMed Wu S, Yoshiko Y, De Luca F: Stanniocalcin 1 acts as a paracrine regulator of growth plate chondrogenesis. J Biol Chem. 2006, 281: 5120-5127. 10.1074/jbc.M506667200.PubMed
96.
Zurück zum Zitat Filvaroff EH, Guillet S, Zlot C, Bao M, Ingle G, Steinmetz H, Hoeffel J, Bunting S, Ross J, Carano RA, et al: Stanniocalcin 1 alters muscle and bone structure and function in transgenic mice. Endocrinology. 2002, 143: 3681-3690. 10.1210/en.2001-211424.PubMed Filvaroff EH, Guillet S, Zlot C, Bao M, Ingle G, Steinmetz H, Hoeffel J, Bunting S, Ross J, Carano RA, et al: Stanniocalcin 1 alters muscle and bone structure and function in transgenic mice. Endocrinology. 2002, 143: 3681-3690. 10.1210/en.2001-211424.PubMed
97.
Zurück zum Zitat Silha JV, Mishra S, Rosen CJ, Beamer WG, Turner RT, Powell DR, Murphy LJ: Perturbations in bone formation and resorption in insulin-like growth factor binding protein-3 transgenic mice. J Bone Miner Res. 2003, 18: 1834-1841. 10.1359/jbmr.2003.18.10.1834.PubMed Silha JV, Mishra S, Rosen CJ, Beamer WG, Turner RT, Powell DR, Murphy LJ: Perturbations in bone formation and resorption in insulin-like growth factor binding protein-3 transgenic mice. J Bone Miner Res. 2003, 18: 1834-1841. 10.1359/jbmr.2003.18.10.1834.PubMed
98.
Zurück zum Zitat Ijiri K, Zerbini LF, Peng H, Correa RG, Lu B, Walsh N, Zhao Y, Taniguchi N, Huang XL, Out H, et al: A novel role for GADD45beta as a mediator of MMP-13 gene expression during chondrocyte terminal differentiation. J Biol Chem. 2005, 280: 38544-38555. 10.1074/jbc.M504202200.PubMedCentralPubMed Ijiri K, Zerbini LF, Peng H, Correa RG, Lu B, Walsh N, Zhao Y, Taniguchi N, Huang XL, Out H, et al: A novel role for GADD45beta as a mediator of MMP-13 gene expression during chondrocyte terminal differentiation. J Biol Chem. 2005, 280: 38544-38555. 10.1074/jbc.M504202200.PubMedCentralPubMed
99.
Zurück zum Zitat Wang K, Vishwanath P, Eichler GS, Al-Sebaei MO, Edgar CM, Einhorn TA, Smith TF, Gerstenfeld LC: Analysis of fracture healing by large-scale transcriptional profile identified temporal relationships between metalloproteinase and ADAMTS mRNA expression. Matrix Biol. 2006, 25: 271-281. 10.1016/j.matbio.2006.02.001.PubMed Wang K, Vishwanath P, Eichler GS, Al-Sebaei MO, Edgar CM, Einhorn TA, Smith TF, Gerstenfeld LC: Analysis of fracture healing by large-scale transcriptional profile identified temporal relationships between metalloproteinase and ADAMTS mRNA expression. Matrix Biol. 2006, 25: 271-281. 10.1016/j.matbio.2006.02.001.PubMed
100.
Zurück zum Zitat Sone S, Nakamura M, Maruya Y, Takahashi I, Mizoguchi I, Mayanagi H, Sasano Y: Expression of versican and ADAMTS during rat tooth eruption. J Mol Histol. 2005, 36: 281-288. 10.1007/s10735-005-5534-2.PubMed Sone S, Nakamura M, Maruya Y, Takahashi I, Mizoguchi I, Mayanagi H, Sasano Y: Expression of versican and ADAMTS during rat tooth eruption. J Mol Histol. 2005, 36: 281-288. 10.1007/s10735-005-5534-2.PubMed
101.
Zurück zum Zitat Cornish J, Callon KE, Naot D, Palmano KP, Banovic T, Bava U, Watson M, Lin JM, Tong PC, Chen Q, et al: Lactoferrin is a potent regulator of bone cell activity and increases bone formation in vivo. Endocrinology. 2004, 145: 4366-4374. 10.1210/en.2003-1307.PubMed Cornish J, Callon KE, Naot D, Palmano KP, Banovic T, Bava U, Watson M, Lin JM, Tong PC, Chen Q, et al: Lactoferrin is a potent regulator of bone cell activity and increases bone formation in vivo. Endocrinology. 2004, 145: 4366-4374. 10.1210/en.2003-1307.PubMed
102.
Zurück zum Zitat Zayzafoon M, Gathings WE, McDonald JM: Modeled microgravity inhibits osteogenic differentiation of human mesenchymal stem cells and increases adipogenesis. Endocrinology. 2004, 145: 2421-2432. 10.1210/en.2003-1156.PubMed Zayzafoon M, Gathings WE, McDonald JM: Modeled microgravity inhibits osteogenic differentiation of human mesenchymal stem cells and increases adipogenesis. Endocrinology. 2004, 145: 2421-2432. 10.1210/en.2003-1156.PubMed
103.
Zurück zum Zitat Choi SJ, Han JH, Roodman GD: ADAM8: a novel osteoclast stimulating factor. J Bone Miner Res. 2001, 16: 814-822. 10.1359/jbmr.2001.16.5.814.PubMed Choi SJ, Han JH, Roodman GD: ADAM8: a novel osteoclast stimulating factor. J Bone Miner Res. 2001, 16: 814-822. 10.1359/jbmr.2001.16.5.814.PubMed
104.
Zurück zum Zitat Kim MS, Magno CL, Day CJ, Morrison NA: Induction of chemokines and chemokine receptors CCR2b and CCR4 in authentic human osteoclasts differentiated with RANKL and osteoclast like cells differentiated by MCP-1 and RANTES. J Cell Biochem. 2006, 97: 512-518. 10.1002/jcb.20649.PubMed Kim MS, Magno CL, Day CJ, Morrison NA: Induction of chemokines and chemokine receptors CCR2b and CCR4 in authentic human osteoclasts differentiated with RANKL and osteoclast like cells differentiated by MCP-1 and RANTES. J Cell Biochem. 2006, 97: 512-518. 10.1002/jcb.20649.PubMed
105.
Zurück zum Zitat Razzouk S, Lieberherr M, Cournot G: Rac-GTPase, osteoclast cytoskeleton and bone resorption. Eur J Cell Biol. 1999, 78: 249-255.PubMed Razzouk S, Lieberherr M, Cournot G: Rac-GTPase, osteoclast cytoskeleton and bone resorption. Eur J Cell Biol. 1999, 78: 249-255.PubMed
106.
Zurück zum Zitat Sorensen MG, Henriksen K, Schaller S, Henriksen DB, Nielsen FC, Dziegiel MH, Karsdal MA: Characterization of osteoclasts derived from CD14+ monocytes isolated from peripheral blood. J Bone Miner Metab. 2007, 25: 36-45. 10.1007/s00774-006-0725-9.PubMed Sorensen MG, Henriksen K, Schaller S, Henriksen DB, Nielsen FC, Dziegiel MH, Karsdal MA: Characterization of osteoclasts derived from CD14+ monocytes isolated from peripheral blood. J Bone Miner Metab. 2007, 25: 36-45. 10.1007/s00774-006-0725-9.PubMed
107.
Zurück zum Zitat Johnson GB, Riggs BL, Platt JL: A genetic basis for the 'Adonis' phenotype of low adiposity and strong bones. Faseb J. 2004, 18: 1282-1284.PubMed Johnson GB, Riggs BL, Platt JL: A genetic basis for the 'Adonis' phenotype of low adiposity and strong bones. Faseb J. 2004, 18: 1282-1284.PubMed
108.
Zurück zum Zitat Li F, Chung H, Reddy SV, Lu G, Kurihara N, Zhao AZ, Roodman GD: Annexin II stimulates RANKL expression through MAPK. J Bone Miner Res. 2005, 20: 1161-1167. 10.1359/JBMR.050207.PubMed Li F, Chung H, Reddy SV, Lu G, Kurihara N, Zhao AZ, Roodman GD: Annexin II stimulates RANKL expression through MAPK. J Bone Miner Res. 2005, 20: 1161-1167. 10.1359/JBMR.050207.PubMed
109.
Zurück zum Zitat Chellaiah M, Kizer N, Silva M, Alvarez U, Kwiatkowski D, Hruska KA: Gelsolin deficiency blocks podosome assembly and produces increased bone mass and strength. J Cell Biol. 2000, 148: 665-678. 10.1083/jcb.148.4.665.PubMedCentralPubMed Chellaiah M, Kizer N, Silva M, Alvarez U, Kwiatkowski D, Hruska KA: Gelsolin deficiency blocks podosome assembly and produces increased bone mass and strength. J Cell Biol. 2000, 148: 665-678. 10.1083/jcb.148.4.665.PubMedCentralPubMed
110.
Zurück zum Zitat Tani-Ishii N, Penninger JM, Matsumoto G, Teranaka T, Umemoto T: The role of LFA-1 in osteoclast development induced by co-cultures of mouse bone marrow cells and MC3T3-G2/PA6 cells. J Periodontal Res. 2002, 37: 184-191. 10.1034/j.1600-0765.2002.00610.x.PubMed Tani-Ishii N, Penninger JM, Matsumoto G, Teranaka T, Umemoto T: The role of LFA-1 in osteoclast development induced by co-cultures of mouse bone marrow cells and MC3T3-G2/PA6 cells. J Periodontal Res. 2002, 37: 184-191. 10.1034/j.1600-0765.2002.00610.x.PubMed
111.
Zurück zum Zitat Miura Y, Miura M, Gronthos S, Allen MR, Cao C, Uveges TE, Bi Y, Ehirchiou D, Kortesidis A, Shi S, et al: Defective osteogenesis of the stromal stem cells predisposes CD18-null mice to osteoporosis. Proc Natl Acad Sci USA. 2005, 102: 14022-14027. 10.1073/pnas.0409397102.PubMedCentralPubMed Miura Y, Miura M, Gronthos S, Allen MR, Cao C, Uveges TE, Bi Y, Ehirchiou D, Kortesidis A, Shi S, et al: Defective osteogenesis of the stromal stem cells predisposes CD18-null mice to osteoporosis. Proc Natl Acad Sci USA. 2005, 102: 14022-14027. 10.1073/pnas.0409397102.PubMedCentralPubMed
112.
Zurück zum Zitat Sugatani T, Alvarez U, Hruska KA: PTEN regulates RANKL- and osteopontin-stimulated signal transduction during osteoclast differentiation and cell motility. J Biol Chem. 2003, 278: 5001-5008. 10.1074/jbc.M209299200.PubMed Sugatani T, Alvarez U, Hruska KA: PTEN regulates RANKL- and osteopontin-stimulated signal transduction during osteoclast differentiation and cell motility. J Biol Chem. 2003, 278: 5001-5008. 10.1074/jbc.M209299200.PubMed
113.
Zurück zum Zitat Shi C, Zhang X, Chen Z, Sulaiman K, Feinberg MW, Ballantyne CM, Jain MK, Simon DI: Integrin engagement regulates monocyte differentiation through the forkhead transcription factor Foxp1. J Clin Invest. 2004, 114: 408-418. 10.1172/JCI200421100.PubMedCentralPubMed Shi C, Zhang X, Chen Z, Sulaiman K, Feinberg MW, Ballantyne CM, Jain MK, Simon DI: Integrin engagement regulates monocyte differentiation through the forkhead transcription factor Foxp1. J Clin Invest. 2004, 114: 408-418. 10.1172/JCI200421100.PubMedCentralPubMed
114.
Zurück zum Zitat Gantner F, Kupferschmidt R, Schudt C, Wendel A, Hatzelmann A: In vitro differentiation of human monocytes to macrophages: change of PDE profile and its relationship to suppression of tumour necrosis factor-alpha release by PDE inhibitors. Br J Pharmacol. 1997, 121: 221-231. 10.1038/sj.bjp.0701124.PubMedCentralPubMed Gantner F, Kupferschmidt R, Schudt C, Wendel A, Hatzelmann A: In vitro differentiation of human monocytes to macrophages: change of PDE profile and its relationship to suppression of tumour necrosis factor-alpha release by PDE inhibitors. Br J Pharmacol. 1997, 121: 221-231. 10.1038/sj.bjp.0701124.PubMedCentralPubMed
115.
Zurück zum Zitat Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, Deng L, Clemens TL, Williams BO: Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem. 2005, 280: 21162-21168. 10.1074/jbc.M501900200.PubMed Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, Deng L, Clemens TL, Williams BO: Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem. 2005, 280: 21162-21168. 10.1074/jbc.M501900200.PubMed
116.
Zurück zum Zitat Almeida M, Han L, Bellido T, Manolagas SC, Kousteni S: Wnt proteins prevent apoptosis of both uncommitted osteoblast progenitors and differentiated osteoblasts by beta-catenin-dependent and -independent signaling cascades involving Src/ERK and phosphatidylinositol 3-kinase/AKT. J Biol Chem. 2005, 280: 41342-41351. 10.1074/jbc.M502168200.PubMed Almeida M, Han L, Bellido T, Manolagas SC, Kousteni S: Wnt proteins prevent apoptosis of both uncommitted osteoblast progenitors and differentiated osteoblasts by beta-catenin-dependent and -independent signaling cascades involving Src/ERK and phosphatidylinositol 3-kinase/AKT. J Biol Chem. 2005, 280: 41342-41351. 10.1074/jbc.M502168200.PubMed
117.
Zurück zum Zitat Liu X, Bruxvoort KJ, Zylstra CR, Liu J, Cichowski R, Faugere MC, Bouxsein ML, Wan C, Williams BO, Clemens TL: Lifelong accumulation of bone in mice lacking Pten in osteoblasts. Proc Natl Acad Sci USA. 2007, 104: 2259-2264. 10.1073/pnas.0604153104.PubMedCentralPubMed Liu X, Bruxvoort KJ, Zylstra CR, Liu J, Cichowski R, Faugere MC, Bouxsein ML, Wan C, Williams BO, Clemens TL: Lifelong accumulation of bone in mice lacking Pten in osteoblasts. Proc Natl Acad Sci USA. 2007, 104: 2259-2264. 10.1073/pnas.0604153104.PubMedCentralPubMed
118.
Zurück zum Zitat Sneddon WB, Syme CA, Bisello A, Magyar CE, Rochdi MD, Parent JL, Weinman EJ, Abou-Samra AB, Friedman PA: Activation-independent parathyroid hormone receptor internalization is regulated by NHERF1 (EBP50). J Biol Chem. 2003, 278: 43787-43796. 10.1074/jbc.M306019200.PubMed Sneddon WB, Syme CA, Bisello A, Magyar CE, Rochdi MD, Parent JL, Weinman EJ, Abou-Samra AB, Friedman PA: Activation-independent parathyroid hormone receptor internalization is regulated by NHERF1 (EBP50). J Biol Chem. 2003, 278: 43787-43796. 10.1074/jbc.M306019200.PubMed
119.
Zurück zum Zitat Kulkarni NH, Halladay DL, Miles RR, Gilbert LM, Frolik CA, Galvin RJ, Martin TJ, Gillespie MT, Onyia JE: Effects of parathyroid hormone on Wnt signaling pathway in bone. J Cell Biochem. 2005, 95: 1178-1190. 10.1002/jcb.20506.PubMed Kulkarni NH, Halladay DL, Miles RR, Gilbert LM, Frolik CA, Galvin RJ, Martin TJ, Gillespie MT, Onyia JE: Effects of parathyroid hormone on Wnt signaling pathway in bone. J Cell Biochem. 2005, 95: 1178-1190. 10.1002/jcb.20506.PubMed
120.
Zurück zum Zitat Katoh M: WNT/PCP signaling pathway and human cancer [review]. Oncol Rep. 2005, 14: 1583-1588.PubMed Katoh M: WNT/PCP signaling pathway and human cancer [review]. Oncol Rep. 2005, 14: 1583-1588.PubMed
121.
Zurück zum Zitat Liu B, Yu HM, Hsu W: Craniosynostosis caused by Axin2 deficiency is mediated through distinct functions of beta-catenin in proliferation and differentiation. Dev Biol. 2007, 301: 298-308. 10.1016/j.ydbio.2006.10.018.PubMedCentralPubMed Liu B, Yu HM, Hsu W: Craniosynostosis caused by Axin2 deficiency is mediated through distinct functions of beta-catenin in proliferation and differentiation. Dev Biol. 2007, 301: 298-308. 10.1016/j.ydbio.2006.10.018.PubMedCentralPubMed
122.
Zurück zum Zitat Shore EM, Xu M, Feldman GJ, Fenstermacher DA, Cho TJ, Choi IH, Connor JM, Delai P, Glaser DL, LeMerrer M, et al: A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva. Nat Genet. 2006, 38: 525-527. 10.1038/ng1783.PubMed Shore EM, Xu M, Feldman GJ, Fenstermacher DA, Cho TJ, Choi IH, Connor JM, Delai P, Glaser DL, LeMerrer M, et al: A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva. Nat Genet. 2006, 38: 525-527. 10.1038/ng1783.PubMed
123.
Zurück zum Zitat Maeda Y, Tsuji K, Nifuji A, Noda M: Inhibitory helix-loop-helix transcription factors Id1/Id3 promote bone formation in vivo. J Cell Biochem. 2004, 93: 337-344. 10.1002/jcb.20154.PubMed Maeda Y, Tsuji K, Nifuji A, Noda M: Inhibitory helix-loop-helix transcription factors Id1/Id3 promote bone formation in vivo. J Cell Biochem. 2004, 93: 337-344. 10.1002/jcb.20154.PubMed
124.
Zurück zum Zitat de Jong DS, van Zoelen EJ, Bauerschmidt S, Olijve W, Steegenga WT: Microarray analysis of bone morphogenetic protein, transforming growth factor beta, and activin early response genes during osteoblastic cell differentiation. J Bone Miner Res. 2002, 17: 2119-2129. 10.1359/jbmr.2002.17.12.2119.PubMed de Jong DS, van Zoelen EJ, Bauerschmidt S, Olijve W, Steegenga WT: Microarray analysis of bone morphogenetic protein, transforming growth factor beta, and activin early response genes during osteoblastic cell differentiation. J Bone Miner Res. 2002, 17: 2119-2129. 10.1359/jbmr.2002.17.12.2119.PubMed
125.
Zurück zum Zitat Wiater E, Vale W: Inhibin is an antagonist of bone morphogenetic protein signaling. J Biol Chem. 2003, 278: 7934-7941. 10.1074/jbc.M209710200.PubMed Wiater E, Vale W: Inhibin is an antagonist of bone morphogenetic protein signaling. J Biol Chem. 2003, 278: 7934-7941. 10.1074/jbc.M209710200.PubMed
126.
Zurück zum Zitat Abe Y, Abe T, Aida Y, Hara Y, Maeda K: Follistatin restricts bone morphogenetic protein (BMP)-2 action on the differentiation of osteoblasts in fetal rat mandibular cells. J Bone Miner Res. 2004, 19: 1302-1307. 10.1359/JBMR.040408.PubMed Abe Y, Abe T, Aida Y, Hara Y, Maeda K: Follistatin restricts bone morphogenetic protein (BMP)-2 action on the differentiation of osteoblasts in fetal rat mandibular cells. J Bone Miner Res. 2004, 19: 1302-1307. 10.1359/JBMR.040408.PubMed
Metadaten
Titel
Microarray gene expression profiling of osteoarthritic bone suggests altered bone remodelling, WNT and transforming growth factor-β/bone morphogenic protein signalling
verfasst von
Blair Hopwood
Anna Tsykin
David M Findlay
Nicola L Fazzalari
Publikationsdatum
01.10.2007
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 5/2007
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/ar2301

Weitere Artikel der Ausgabe 5/2007

Arthritis Research & Therapy 5/2007 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.