Skip to main content
Erschienen in: BMC Musculoskeletal Disorders 1/2010

Open Access 01.12.2010 | Research article

Analysis of meniscal degeneration and meniscal gene expression

verfasst von: Yubo Sun, David R Mauerhan, Patrick R Honeycutt, Jeffrey S Kneisl, James H Norton, Edward N Hanley Jr, Helen E Gruber

Erschienen in: BMC Musculoskeletal Disorders | Ausgabe 1/2010

Abstract

Background

Menisci play a vital role in load transmission, shock absorption and joint stability. There is increasing evidence suggesting that OA menisci may not merely be bystanders in the disease process of OA. This study sought: 1) to determine the prevalence of meniscal degeneration in OA patients, and 2) to examine gene expression in OA meniscal cells compared to normal meniscal cells.

Methods

Studies were approved by our human subjects Institutional Review Board. Menisci and articular cartilage were collected during joint replacement surgery for OA patients and lower limb amputation surgery for osteosarcoma patients (normal control specimens), and graded. Meniscal cells were prepared from these meniscal tissues and expanded in monolayer culture. Differential gene expression in OA meniscal cells and normal meniscal cells was examined using Affymetrix microarray and real time RT-PCR.

Results

The grades of meniscal degeneration correlated with the grades of articular cartilage degeneration (r = 0.672; P < 0.0001). Many of the genes classified in the biological processes of immune response, inflammatory response, biomineral formation and cell proliferation, including major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), integrin, beta 2 (ITGB2), ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), ankylosis, progressive homolog (ANKH) and fibroblast growth factor 7 (FGF7), were expressed at significantly higher levels in OA meniscal cells compared to normal meniscal cells. Importantly, many of the genes that have been shown to be differentially expressed in other OA cell types/tissues, including ADAM metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS5) and prostaglandin E synthase (PTGES), were found to be expressed at significantly higher levels in OA meniscal cells. This consistency suggests that many of the genes detected in our study are disease-specific.

Conclusion

Our findings suggest that OA is a whole joint disease. Meniscal cells may play an active role in the development of OA. Investigation of the gene expression profiles of OA meniscal cells may reveal new therapeutic targets for OA therapy and also may uncover novel disease markers for early diagnosis of OA.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2474-11-19) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

YS, HEG and ENH conceived the study and participated in design and coordination. YS wrote the manuscript. DRM and JSK provided surgical tissues and participated in the discussion of experimental results. HJN assisted with statistical analysis. PRH, DRM and YS graded menisci and articular cartilage. PRH prepared cell culture and extracted RNA. HEG assisted with manuscript preparation. All authors read and approved the final manuscript.
Abkürzungen
OA
osteoarthritis
FLS
fibroblast-like synoviocytes
MMP
matrix metalloproteinase
ITGB
integrin: beta
ADAMTS5
metallopeptidase with thrombospondin type 1 motif 5
ANKH
ankylosis: progressive homolog
BST1
bone marrow stromal cell antigen 1
CACNA1C
calcium channel: voltage-dependent: L type: alpha 1C subunit
COL5A1
collagen, type V, alpha 1
COL11A1
collagen, type XI, alpha 1
CTSS
cathepsin S
ENPP1
ectonucleotide pyrophosphatase/phosphodiesterase 1
FGF7
fibroblast growth factor 7
FMOD
fibromodul
FUS
fusion
GAPDH
glyceraldehyde-3-phosphate dehydrogenase
HLA-DPA1
major histocompatibility complex, class II, DP alpha 1
IGFBP7
insulin-like growth factor binding protein 7
MCAM
melanoma cell adhesion molecule
MGP
matrix Gla protein
PTGES
prostaglandin E synthase
SFRP4
secreted frizzled-related protein 4
TUFT1
tuftelin
TBX5
T-box 5
TNXB
tenascin XB.

Background

Osteoarthritis (OA) is a disease characterized by the breakdown of articular cartilage and the formation of osteophytes. However, it has been gradually realized that OA is not merely an articular cartilage disease, but a disease of the whole joint [1, 2]. OA synovial membrane and subchondral bone have drawn considerably attention recently. Aberrant gene expression in OA synovium, OA fibroblast-like synoviocytes (FLS) and OA subchondral bone has been detected [36]. These findings suggest that OA synoviocytes and subchondral bone cells may be involved in the disease process of OA.
Few studies have investigated the potential role of OA meniscal cells in the disease process of OA. The knee menisci are specialized tissues that play a vital role in load transmission, shock absorption and joint stability. Knee menisci may absorb most of the shock generated at the joint because their combined mass is greater than that of the articular cartilage. The current dogma is that menisci protect the articular cartilage, but play a minimal role in the disease process of OA unless they are injured. However, increasing evidence suggests that knee menisci may not be passive bystanders in the disease process of OA. It has been reported that meniscal degeneration is a feature of OA knee joints as revealed by magnetic resonance imaging [7, 8]. There is a strong association between meniscal damage/degenerative tears and cartilage loss [9]. In addition, it has been found that meniscal degeneration contributes to joint space narrowing [10]. Taken together, these findings and observations suggest that pathological changes and extracellular matrix degeneration also occur in OA menisci. OA menisci, similar to OA synovial membrane and OA subchondral bone, may play an active role in the disease process of OA.
In the present study, we examined the prevalence of meniscal degeneration in OA patients who underwent joint replacement surgery, and analyzed the correlation between the degeneration of menisci and the degeneration of articular cartilage. We also examined differential gene expression between OA meniscal cells and normal meniscal cells to test the hypothesis that OA meniscal cells are different from normal meniscal cells and may display a disease-specific gene expression profile. The determination of the differential gene expression between OA and normal meniscal cells may not only provide experimental evident to support our hypothesis, but also may reveal new therapeutic targets for OA therapy and uncover novel disease markers for early diagnosis of OA.

Methods

Menisci and articular cartilage specimens

Menisci and articular cartilage specimens were collected from OA patients who underwent joint replacement surgery and from osteosarcoma patients who underwent lower limb amputation surgery with the approval of the authors' Institutional Review Board. The need for informed consent was waived since the menisci and articular cartilage were surgical waste of routine joint replacement surgery and lower limb amputation surgery, and there was no patient private information being collected. Specimens were transported to the laboratory from the operation room at our hospital in sterile tissue culture medium. The medial menisci and the medial articular cartilage of the tibia were graded (by DRM, PRH and YS) according to following scales. For the medial meniscus: 0 = normal appearing surface; 1 = minimal fibrillation and degeneration; 2 = moderate fibrillation and degeneration; 3 = severe fibrillation and degeneration, no tears; 4 = severe fibrillation and degeneration, multiple incomplete tears, or complete tears. For medial articular cartilage of the tibia: 0 = normal-appearing surface; 1 = minimal fibrillation and degeneration; 2 = erosion extending to middle layers; 3 = erosion extending into the deep layers; 4 = erosion extending to the subchondral bone, and 5 = the majority of articular cartilage completely absent.

Preparation and culture of meniscal cells

Dulbecco's modified eagle medium, fetal bovine serum, stock antibiotic and antimycotic mixture were products of Invitrogen (Carlsbad, CA). Meniscal cells were isolated from meniscal specimens as previously described [11]. Briefly, when meniscal specimens were arrived in sterile tissue culture medium, the central region of the medial meniscus was incised, and processed to remove fatty and synovial tissues. These meniscal specimens were then minced into small pieces (3 mm × 3 mm), and cultured in 100 mm plates at 37°C in medium containing 0.5% antibiotic/antimycotic solution and 10% serum. Every three days, medium was changed. Meniscal cells proliferated, migrated out the tissues, attached to the plate and expanded. When meniscal cells reached 80% confluence, they were replated and expanded in monolayer culture. Passage two cells were used in all studies. Meniscal cells prepared from meniscal specimens derived from osteosarcoma patients were used as normal control meniscal cells.

RNA extraction and microarrays

OA meniscal cells and normal meniscal cells (passage 2) were plated in 100 mm plates at 75% confluence. On the second day, medium containing 1% serum was added and cells were cultured for twenty-four hours. Medium (1% serum) was changed again and cells cultured for another twenty-four hours. Total RNA was extracted using Trizol reagent (Invitrogen, Carlsbad, CA), and purified using Oligotex kit (Qiagen, Valencia, CA). Microarray analyses were performed using these RNA samples. For microarray analysis, double stranded DNA was synthesized from RNA samples using SuperScript double-stranded cDNA synthesis kit (Invitrogen, Carlsbad, CA). The DNA product was purified using GeneChip sample cleanup module (Affymetrix, Santa Clara, CA). cRNA was synthesized and biotin labeled using BioArray high yield RNA transcript labeling kit (Enzo Life Sciences, Farmingdale, NY). The product was purified using GeneChip sample cleanup module and subsequently chemically fragmented. The fragmented, biotinylated cRNA was hybridized to HG-U133_Plus_2 gene chip (Affymetrix, Santa Clara, CA) using Affymetrix Fluidics Station 400. The fluorescent signal was quantified during two scans by Agilent Gene Array Scanner G2500A (Agilent Technologies, Palo Alto, CA), and GeneChip operating Software (Affymetrix, Santa Clara, CA).

Real-time RT-PCR

cDNA was synthesized using TaqMan® Reverse Transcription Reagents (Applied Biosystems, Inc., University Park, IL). Quantification of relative transcript levels for selected genes and the housekeeping gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was performed using the ABI7000 Real Time PCR system (Applied Biosystems, Inc., University Park, IL). TaqMan® Gene Expression assays were used, which contains a FAM-MGB probe for fluorescent detection. cDNA samples were amplified with an initial Taq DNA polymerase activation step at 95°C for 10 minutes, followed by 40 cycles of denaturation at 95°C for 15 seconds and annealing at 60°C for one minute. For each gene, Ct values were obtained in triplicates. Fold change was calculated [12] and the expression level of the genes of interest was normalized to GAPDH. Each real time RT-PCR experiment was repeated twice in triplicate.

Statistical Analysis

The Spearman's correlations between the grades of meniscal degeneration and the grades of articular cartilage degeneration were calculated with SAS® software (version 9.1). A two-tailed p-value of < 0.05 was considered statistically significant. For microarray analysis, Genesifter software (VizX Labs, Seattle, WA) was used to determine fold changes in gene expression, and gene ontologies. Statistical significance was determined using the Student t-test (p < 0.05). A correction factor for false discovery rate was applied using the Benjamini and Hochberg method [13].

Results

Degeneration of menisci in OA patients

Demographic patient features and assigned grades are presented in Table 1. All menisci had signs of degeneration. More than 80% menisci showed signs of severe degeneration (grade 4 or grade 3). Representative images of grade 4 OA menisci and grade 0 normal control menisci are shown in Figure 1. Grade 4 OA menisci displayed discolorations and an extremely rough surface. Severe degeneration and fissures/tears were apparent. In contrast, grade 0 control menisci displayed a smooth, white and glistening surface, with no any signs of degeneration.
Table 1
Demographic data and grades of articular cartilage and menisci*
I
Cartilage Grade
# Subjects
Mean Age ± SD
 
0
1 F
39
 
0
1 F
12
 
0
1 M
43
 
2
3 F, 1 M
62 ± 13
 
3
6 F, 5 M
64 ± 9
 
4
15 F, 4 M
62 ± 11
 
5
12 F, 5 M
67 ± 9
II
Meniscus Grade
# Subjects
Mean Age ± SD
 
0
1 F
39
 
0
1 F
12
 
0
1 M
43
 
2
4 F, 3 M
57 ± 12
 
3
12 F, 6 M
64 ± 9
 
4
20 F, 6 M
66 ± 9
*The first three patients are osteosarcoma patients. The rest fifty-one patients are OA patients. F, female; M, male
The relationship between the grade of menisci and the grade of articular cartilage was examined. A correlation between the grade of menisci and the grade of articular cartilage was found. The Spearman's correlation coefficient (r) was 0.672 (p < 0.0001). Of the seventeen OA patients who had grade 5 (the highest grade of articular cartilage degeneration) articular cartilage, fourteen patients (82%) had grade 4 medial meniscus (the highest grade of meniscal degeneration) and three patients had grade 3 medial meniscus. Of the nineteen OA patients who had grade 4 articular cartilages, ten patients had grade 4 (58.82%) and seven had grade 3 (41.17%) medial meniscus. Only two patients had grade 2 (0.11%) medial meniscus.

Differential gene expression

Microarray analyses of the differential gene expression between OA meniscal cells prepared from menisci (grade 4) derived from five OA patients and normal meniscal cells prepared from menisci (grade 0) derived from three osteosarcoma patients were carried out as described in Methods. The row microarray data can be found in Gene Expression Omnibus (GEO; http://​www.​ncbi.​nlm.​nih.​gov/​projects/​geo/​) with the accession number #GSE19060. Table 2 presents the demographic data of the meniscal specimens which were the basis of this analysis. Of more than 50,000 transcripts examined, 505 transcripts that displayed differential gene expression (more than 1.2 fold) were identified; 149 transcripts were expressed at significantly higher levels and 356 transcripts at significantly lower levels in OA meniscal cells compared to normal meniscal cells. The differentially expressed genes were classified according to gene ontology category biological process using GeneSifter software (VizX Labs, Seattle, WA). Selected genes, which fell into specific biological processes previously implicated in OA or suspected to play a role in OA, or displayed a distinct expression pattern, are listed in Table 3.
Table 2
Demographic data and grades of menisci that were used to prepare meniscal cells*
Patients
Age/Gender/Diagnosis
Grade Meniscus
A
39/F (osteosarcoma)
0
B
43/M (osteosarcoma)
0
C
12/F (osteosarcoma)
0
1
65/F (OA patient)
4
2
56/F (OA patient)
4
3
50/F (OA patient)
4
4
52/F (OA patient)
4
5
61/M (OA patient)
4
* F, female; M, male; OA, osteoarthritis
Table 3
Genes differentially expressed in OA meniscal cells compared to normal meniscal cells.
Biological process
Gene Name
Gene ID
Differ
Exp
(fold)*
Description
Immune response
    
 
HLA-DPA1
M27487
5.3
Major histocompatibility complex, class II, DP alpha 1
 
IFI6
NM_022873
2.0
Interferon, alpha-inducible protein 6
 
CFH
X04697
1.7
Complement factor H
 
CTSS
NM_004079
1.6
Cathepsin S
 
FYN
S74774
1.5
FYN oncogene related to SRC, FGR, YES
 
ILF2
NM_004515
-1.3
Interleukin enhancer binding factor 2, 45 kDa
 
PSMB9
AI375915
-1.2
Proteasome (prosome, macropain) subunit, beta type, 9
Inflammatory
Response
    
 
ITGB2
NM_000211
6.6
Integrin, beta 2 (complement component 3 receptor 3 and 4 subunit)
 
CFH
X04697
1.7
Complement factor H
 
BDKRB2
NM_000623
1.3
Bradykinin receptor B2
Cytokine production
    
 
SRGN
NM_002727
2.8
Serglycin
 
ARNT
AI768497
2.3
Aryl hydrocarbon receptor nuclear translocator
 
INHBA
M13436
1.9
Inhibin, beta A
Calcium ion transport
    
 
CACNA1C
F11066
1.5
Calcium channel, voltage-dependent, L type, alpha 1C subunit
 
FYN
S74774
1.5
FYN oncogene related to SRC, FGR, YES
 
CHRNB2
NM_000748
1.4
Cholinergic receptor, nicotinic, beta 2 (neuronal)
Biomineral formation
    
 
ENPP1
BF057080
1.7
Ectonucleotide pyrophosphatase/phosphodiesterase 1
 
SRGN
NM_002727
2.8
Serglycin
 
ACVR2A
AI149508
1.5
Activin A receptor, type IIA
Cell proliferation
    
 
ITGB2
NM_000211
6.6
Integrin, beta 2 (complement component 3 receptor 3 and 4 subunit)
 
FGF7
NM_002009
4.1
Fibroblast growth factor 7 (keratinocyte growth factor)
 
ELN
AA479278
2.5
Elastin (supravalvular aortic stenosis, Williams-Beuren syndrome)
 
IGFBP7
NM_001553
2.4
Insulin-like growth factor binding protein 7
 
SKAP2
AB014486
2.2
Src kinase associated phosphoprotein 2
 
ARNT
AI768497
2.3
Aryl hydrocarbon receptor nuclear translocator
 
FOXO1
NM_002015
1.5
Forkhead box O1
 
BCAT1
AL390172
1.5
Branched chain aminotransferase 1, cytosolic
 
ACVR2A
AI149508
1.5
Activin A receptor, type IIA
 
TCF7L2
AA664011
1.4
Transcription factor 7-like 2 (T-cell specific, HMG-box)
 
CHRNB2
NM_000748
1.4
Cholinergic receptor, nicotinic, beta 2 (neuronal)
 
TCFL5
NM_006602
1.2
Transcription factor-like 5 (basic helix-loop-helix)
 
NRP1
AA609131
-1.3
Neuropilin 1
 
PHB
AL560017
-1.3
Prohibitin
 
TBX5
NM_000192
-1.3
T-box 5
 
NCK2
BC000103
-1.2
NCK adaptor protein 2
 
PRPF19
NM_014502
-1.2
PRP19/PSO4 pre-mRNA processing factor 19 homolog (S. cerevisiae)
Integrin-mediated signaling pathway
    
 
ITGB2
NM_000211
6.6
Integrin, beta 2
 
ITGB8
BF513121
3.6
Integrin, beta 8
 
ITGA11
AF109681
1.5
integrin, alpha 11
 
GAB2
NM_012296
1.5
GRB2-associated binding protein 2
Skeletal system/Tissue development
    
 
DSP
NM_004415
5.2
Desmoplakin
 
FGF7
NM_002009
4.2
Fibroblast growth factor 7 (keratinocyte growth factor)
 
SRGN
NM_002727
2.8
Serglycin
 
INHBA
M13436
1.9
Inhibin, beta A
 
TIMP3
AW338933
1.7
TIMP metallopeptidase inhibitor 3
 
ENPP1
BF057080
1.7
Ectonucleotide pyrophosphatase/phosphodiesterase 1
 
COL5A1
N30339
1.7
Collagen, type V, alpha 1
 
ACVR2A
AI149508
1.5
Activin A receptor, type IIA
 
FBN1
AW955612
1.4
Fibrillin 1
 
BARX1
NM_021570N
-1.9
BARX homeobox 1
 
GABBR1
NM_001470
-1.9
Gamma-aminobutyric acid (GABA) B receptor, 1
 
HOXA11
H94842
-1.5
Homeobox A11
 
CHST11
NM_018413
-1.5
Carbohydrate (chondroitin 4) sulfotransferase 11
 
NSDHL
BC000245
-1.4
NAD(P) dependent steroid dehydrogenase-like
 
ZNRD1
AF230337
-1.3
Zinc ribbon domain containing 1
 
SMURF1
AF199364
-1.3
SMAD specific E3 ubiquitin protein ligase 1
DNA repair
    
 
ZSWIM7
BE645222
-1.6
Zinc finger, SWIM-type containing 7
 
WDR33
AB044749
-1.5
WD repeat domain 33
 
ASF1A
NM_014034
-1.5
ASF1 anti-silencing function 1 homolog A (S. cerevisiae)
 
ATM
U82828
-1.3
ataxia telangiectasia mutated
 
PMS2L1
AI375694
-1.3
Postmeiotic segregation increased 2-like 1
 
ALKBH2
AI865555
-1.2
AlkB, alkylation repair homolog 2 (E. coli)
 
XRCC6
NM_001469
-1.2
X-ray repair complementing defective repair in Chinese hamster cells 6
 
DCLRE1A
D42045
-1.2
DNA cross-link repair 1A (PSO2 homolog, S. cerevisiae)
 
PRPF19
NM_014502
-1.2
PRP19/PSO4 pre-mRNA processing factor 19 homolog (S. cerevisiae)
 
ELN
AA479278
2.5
Elastin (supravalvular aortic stenosis, Williams-Beuren syndrome)
Cellular biosynthetic process
    
 
PITX2
NM_000325
-6.4
Paired-like homeodomain 2
 
NR3C2
NM_000901
-5.8
Nuclear receptor subfamily 3, group C, member 2
 
GALNT6
AW014155
-4.5
Polypeptide N-acetylgalactosaminyltransferase 6
 
RGMB
BE855765
-2.7
RGM domain family, member B
 
FOXF2
NM_001452
-2.2
Forkhead box F2
 
ZNF529
AL109722
-2.1
Zinc finger protein 529
 
BARX1
NM_021570
-1.9
BARX homeobox 1
 
DHODH
M94065
-1.9
Dihydroorotate dehydrogenase
 
LMO4
BC003600
-1.8
LIM domain only 4
 
BRWD1
NM_018963
-1.7
Bromodomain and WD repeat domain containing 1
 
TARS2
NM_025150
-1.7
Threonyl-tRNA synthetase 2, mitochondrial (putative)
 
HLX
M60721
-1.7
H2.0-like homeobox
 
BNC2
AI767962
-1.7
Basonuclin 2
 
PGD
NM_002631
-1.6
Phosphogluconate dehydrogenase
 
MEIS1
AL832770
-1.6
Meis homeobox 1
 
ZNF420
AI339586
-1.6
Zinc finger protein 420
 
ZFP28
AA831323
-1.5
Zinc finger protein 28 homolog (mouse)
 
CARS2
NM_024537
-1.5
Cysteinyl-tRNA synthetase 2, mitochondrial (putative)
 
HOXA11
H94842
-1.5
Homeobox A11
 
PDLIM1
BC000915
13.2
PDZ and LIM domain 1 (elfin)
 
ITGB2
NM_000211
6.6
Integrin, beta 2 (complement component 3 receptor 3 and 4 subunit)
 
LMCD1
NM_014583
3.5
LIM and cysteine-rich domains 1
 
KRT7
BC002700
3.1
Keratin 7
 
ARNT
AI768497
2.3
Aryl hydrocarbon receptor nuclear translocator
 
ZNF415
NM_018355
2.2
Zinc finger protein 415
 
ZNF630
AK000580
2.0
Zinc finger protein 630
 
INHBA
M13436
1.9
Inhibin, beta A
 
GALNT1
U41514
1.7
Polypeptide N-acetylgalactosaminyltransferase 1
 
ENPP1
BF057080
1.7
Ectonucleotide pyrophosphatase/phosphodiesterase 1
Miscellaneous
    
 
PTGES
AF010316
3.2
Prostaglandin E synthase
 
ADAMTS5
NM_007038
1.7
ADAM metallopeptidase with thrombospondin type 1 motif, 5 (aggrecanase-2)
 
DSP
NM_004415
5.2
Desmoplakin
 
FMOD
NM_002023
2.7
Fibromodulin
 
sFRP4
AW089415
3.4
Secreted frizzled-related protein 4
 
MCAM
M28882
2.3
Melanoma cell adhesion molecule
 
TNXB
NM_004381
-1.3
Tenascin XB
 
FUS
NM_004960
-1.5
Fusion (involved in t(12;16) in malignant liposarcoma)
 
TBX5
NM_000192
-1.3
T-box 5
*Positive number indicates elevated expression (fold) in OA meniscal cells compared to normal meniscal cells. Negative number indicates decreased expression (fold) in OA meniscal cells compared to normal meniscal cells.
We were particularly interested in identifying distinct differential gene expression patterns. As shown in Table 3, distinct differential gene expression patterns were apparent. For example, of the seven differentially-expressed genes classified in the immune response biological process, five genes had elevated expressions but only two genes had decreased expression in OA meniscal cells compared to the control meniscal cells. Of the three differentially-expressed genes classified in the inflammation response biological process, cytokine production biological process, calcium ion transport biological process or the biomineral formation biological process, all these genes had elevated expression in OA meniscal cells compared to the control meniscal cells. Of the four differentially-expressed genes classified in the regulation of phosphate metabolic process, three genes had elevated expression but only one gene had decreased expression in OA meniscal cells compared to the control meniscal cells. Of the eighteen genes classified in the cell proliferation biological process, thirteen genes had elevated expression but only five genes had decreased expression in OA meniscal cells compared to the control meniscal cells. Similar distinct expression patterns were also found in other categories of biological processes, including integrin-mediated signaling pathway and skeletal system/tissue development biological processes (Table 3).
In contrast, most of the genes with decreased expressions in OA meniscal cells compared to the control meniscal cells fell into a different set of biological processes. For example, of the ten differentially-expressed genes classified in the DNA repair biological process, nine genes had decreased expression but only one gene had elevated expression in OA meniscal cells compare to the control meniscal cells. Of the ninety-one differentially expressed genes classified in the cellular biosynthetic process biological process, seventy-four genes had decreased expression in OA meniscal cells and only seventeen genes had elevated expression (Partial results are listed in Table 3). These distinct distributions of the differentially-expressed genes in different sets of biological processes suggest that OA meniscal cells are different from normal meniscal cells. We also analyzed the differential gene expression between individual OA meniscal cells (OA1, OA2, OA3, OA4, and OA5) and the normal control meniscal cells together as a control group. Selected results are listed in Table 4.
Table 4
Genes differentially expressed in individual OA meniscal cells compared to normal meniscal cells.
Gene
Name
Gene ID
OA1
OA1
OA3
OA4
OA5
Description
BST1
NM_004334
3.9
3.4
3.3
1.6
2.2
bone marrow stromal cell antigen 1
FGF9
NM_002010
3.7
2.4
3.7
2.5
2.9
Fibroblast growth factor 9
ACAN
NM_013227
2.4
1.6
1.6
0.0
1.6
Aggrecan
COL11A1
J04177
0.0
11.5
5.8
2.2
3.6
Collagen, type XI, alpha 1
ANKH
AL833238
0.0
1.8
1.7
1.5
1.8
Ankylosis, progressive homolog (mouse)
MGP
NM_000900
11.8
4.7
21.6
0.0
17.6
Matrix Gla protein
TUFT1
NM_020127
2.8
2.7
1.9
0.0
0.0
Tuftelin 1
TFIP11
NM_012143
2.2
1.6
0.0
0.0
0.0
Tuftelin interacting protein 11
IL20RB
AL578102
2.3
2.7
0.0
2.6
7.8
Interleukin 20 receptor beta
IL26
NM_018402
1.7
1.6
1.6
0.0
3.0
Interleukin 26
COL4A5
AW052179
-3.1
-3.0
-3.1
-3.0
-3.1
Collagen, type IV, alpha 5 (Alport syndrome)
MMP9
NM_004994
-2.2
-2.1
-2.1
0.0
-2.2
Matrix metallopeptidase 9
MMP10
NM_002425
-2.7
-2.6
-2.7
-1.5
-2.6
Matrix metallopeptidase 10
MMP12
NM_002426
-2.1
-2.0
-2.1
0.0
-2.1
Matrix metallopeptidase 12
FZD10
NM_007197
-1.6
-1.5
-1.6
-1.5
-1.5
Frizzled homolog 10 (Drosophila)
CYTL1
NM_018659
-2.6
-2.6
-2.6
-2.6
-2.5
Cytokine-like 1
CHODL
NM_024944
-2.6
-2.6
-2.6
 
-2.6
Chondrolectin
*Positive number indicates elevated expression (fold) in OA meniscal cells compared to normal meniscal cells. Negative number indicates decreased expression (fold) in OA meniscal cells compared to normal meniscal cells. 0.0 indicates there was no differential gene expression being detected.

Validation of differential expression of selected genes

The genes selected for validation by quantitative real-time PCR included seven genes that displayed elevated expression in OA meniscal cells compared to normal meniscal cells (Table 5). They were major histocompatibility complex, class II, DP alpha (HLA-DPA1), integrin, beta 2 (ITGB2), ectonucleotide phosphodiesterase 1 (ENPP1), ADAM metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS5), prostaglandin E synthase (PTGES), etc. Matrix metalloproteinase 10 (MMP10), which displayed decreased expression in OA meniscal cells compared to normal meniscal cells, was also selected for validation. RNA sample extracted from OA1 meniscal cells (Table 4) and RNA sample (mixture) extracted from the three normal control meniscal cells were used in the quantitative real time RT-PCR experiments. As shown in Table 5, the differential expressions of these genes were confirmed.
Table 5
Differential gene expression confirmed by real time RT-PCR*
Gene name
Gene ID
Differential Expression* Microarray
Differential Expression* RT-PCR
Description
HLA-DPA1
M27487
5.3
10.5
Major histocompatibility complex, class II, DP alpha
ITGB2
NM_000211
6.6
5.2
Integrin, beta 2
PTGES
AF010316
3.2
3.4
Prostaglandin E synthase
ENPP1
BF057080
1.7
2.1
Ectonucleotide pyrophosphatase/phosphodiesterase 1
ADAMTS5
BI254089
1.7
1.9
ADAM metallopeptidase with thrombospondin type 1 motif, 5
IL26
NM_018402
1.7
4.3
Interleukin 26
TUFT1
NM_020127
2.8
3.9
Matrix metalloproteinase 1
MMP10
NM_002425
-2.7
-3.1
Matrix metalloproteinase 10
*Differential Expression -- the numbers are the ratio of the relative expression level of a specific gene in OA1 meniscal cells to the relative expression level of the specific gene in the control meniscal cells (mixture of RNA prepared from the three normal control meniscal cells).

Discussion

In the present study, we found that more than eighty five percent (85.7%) of the OA patients who underwent joint replacement surgery in our Medical Center had severe degenerative menisci (grades 4 or 3), indicating that meniscal degeneration is common in OA patients. We also found that meniscal degeneration correlated positively with articular cartilage degeneration in OA patients. The Spearman's correlation coefficient (r) was 0.672 (p < 0.0001). Our finding is consistent with previous reports that meniscal degeneration/tears is a feature of OA knee joints [7], that meniscal degeneration contributes to joint space narrowing [10], that there is a strong association between meniscal damage and cartilage loss [9], and that degenerative meniscal tears are positively associated with the severity of articular degeneration compared with other types of meniscal tears [14]. Taken together, our findings suggest that meniscal degeneration in OA, similar to cartilage degeneration, is a major degenerative process regardless of whether it is primary or secondary. Further studies such as examination of the complete patient histories may provide information regarding the cause-result relationship between meniscal degeneration and cartilage degeneration.
If meniscal degeneration is a general feature of OA, one would like to assess whether OA meniscal cells are different from normal meniscal cells and may play a role in the development of OA. To this end, we examined the differential gene expression between OA meniscal cells and normal control meniscal cells. We have recently reported that numerous genes that were classified in the biological process of immune response displayed elevated expression in OA FLS compared to rheumatoid arthritis (RA) FLS. HLA-DPA1 was expressed in OA FLS (hTERT-OA 13A FLS) 16 fold higher than that in RA FLS (hTERT-RA 516 FLS) [4]. Consistently, many genes that were classified in the biological process of immune response including HLA-DPA1 also displayed elevated expression in OA meniscal cells (Table 3). The findings indicate that OA meniscal cells, similar to OA FLS, may be involved in the inflammatory process observed in OA.
Calcium-containing crystals are found in the joint fluid of up to 65% of OA patients, and the presence of these crystals correlates with the radiographic evidence of cartilaginous degeneration [1518]. There is evidence indicating that crystals may promote joint degeneration [1921]. Most recently, it was demonstrated that the inhibition of meniscal calcification by calcium phosphocitrate, a potent anti-calcification agent, was accompanied by a significant reduction in the degeneration of articular cartilage in Hartley guinea pigs [22]. These data suggest that excessive meniscal calcification may play a role in OA. However, there have been no studies to investigate the alterations in OA meniscal cells or to identify the candidate disease genes that are potentially responsible for the excessive meniscal calcification in OA. In the present study, we found that many genes which are involved in biomineral formation such as ENPP1 [23], in phosphate metabolic process such as ITGB2 and in calcium ion transport such as calcium channel, voltage-dependent, L type, alpha 1C subunit (CACNA1C) are expressed at an elevated levels in OA meniscal cells compared to normal meniscal cells. These findings are consistent with clinical observations that meniscal calcification is more severe in OA menisci [24, 25] and that calcium content in OA menisci is positively correlated with the stage of meniscal degeneration [26]. In addition to the genes listed in Table 3, several other genes that have been previously implicated in pathological calcification were also detected (Table 4). They were ankylosis progressive homolog (ANKH) [27, 28], matrix Gla protein (MGP) [29, 30] and tuftelin (TUFT1) [3133]. Taken together, our findings suggest that OA meniscal cells may be actively involved in the meniscal calcification process in OA.
Many genes that have been previously found to be expressed at elevated levels in other types of OA cells or tissues such as in OA articular cartilage and OA bone were also detected in this study. They were integrin, beta 8 (ITGB8) [34], insulin-like growth factor binding protein 7 (IGFBP7) [34], fibromodul (FMOD) [28], cathepsin S (CTSS) [35], secreted frizzled-related protein 4 (SFRP4) [34], bone marrow stromal cell antigen 1 (BST1) [35], collagen, type XI, alpha 1 (COL11A1) [34], collagen, type V, alpha 1 (COL5A1) [34], PTGES [36] and ADAMTS5 [37]. These genes were expressed at significantly elevated levels in OA meniscal cells compared to normal meniscal cells (Tables 3 and 4). ADAMTS5 is a major cartilage matrix degrading enzyme and has been implicated in articular cartilage degeneration previously [38, 39]. The elevated expression of ADAMTS5 in OA meniscal cells suggests that ADAMTS5 may play a role in meniscal degeneration.
Furthermore, many genes that have been previously found to be expressed at decreased levels in other types of OA cells or tissues were also detected in this study. They are MMP10 [34], MMP12 [40], tenascin XB (TNXB) [34], fusion (FUS) [28], melanoma cell adhesion molecule (MCAM) [35], and T-box 5 (TBX5) [5]. These genes were expressed at decreased levels in OA meniscal cells compared to normal meniscal cells (Tables 3 and 4). The consistency between our findings and the previous findings provides strong support for our hypothesis that OA meniscal cells are different from normal meniscal cells and may play an active role in the development of OA.
Our study has some limitations which should be considered. The control meniscal cells we used were derived from the menisci of osteosarcoma patients and were not optimal normal control meniscal cells. To minimize this limitation, we only collected overtly normal-appearing meniscal and cartilage specimens (grade 0) from osteosarcoma patients whose tumors were located far away from the knee joints. In addition, we analyzed the differential gene expression using meniscal cells rather than using meniscal tissue specimens directly to eliminate the effect of different drugs that might be taken by OA and osteosarcoma patients at the time of surgery.
Most meniscal cells synthesize type I collagen as their major collagen product. The meniscal cells in the inner, central nonvascularized region of meniscus synthesize type II collagen. It is an open question at this time as to whether a single cell type exists in the meniscus that displays a fibroblast-like phenotype or a chondrocyte-like phenotype depending on its environment, or whether two or more distinct cell types exist. Therefore, it is not absolutely certain at this time whether the differences in the gene expressions detected in this study reflect the overall differences between OA meniscal cells and the control meniscal cells or only reflect the differences between a subpopulation of OA meniscal cells and a subpopulation of the control meniscal cells. Another limitation is that the age of osteosarcoma patients was younger than the age of OA patients. Therefore, certain genes we detected may be age-related rather disease-specific. It is difficult to obtain age-matched control meniscal specimens because osteosarcoma occurs often in younger patients while OA occurs mostly in older patients. We will continue this line of study when more age matched normal control meniscal specimens become available in the future. In spite of these limitations, the consistency between our findings and the previous findings that many of the genes we detected are also abnormally expressed in other OA cell types/tissues [4, 5, 23, 28, 3436, 40] suggests that many of the differential gene expressions detected in this study are disease-specific.

Conclusions

Our findings suggest that OA is not merely a cartilage disease, but also a disease of the menisci. OA meniscal cells may play an active role in the disease process of OA. Investigation of the gene expression profile of OA meniscal cells compared to normal meniscal cells may reveal new therapeutic targets for OA therapy and uncover novel disease markers for early diagnosis of OA.

Acknowledgements

This study is supported in part by Charlotte-Mecklenburg Education and Research Foundation Grant (to YS) and Mecklenburg County Medical Society Smith Arthritis Fund Grant (to YS). This study was performed at Carolinas Medical Center, Charlotte, NC.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

YS, HEG and ENH conceived the study and participated in design and coordination. YS wrote the manuscript. DRM and JSK provided surgical tissues and participated in the discussion of experimental results. HJN assisted with statistical analysis. PRH, DRM and YS graded menisci and articular cartilage. PRH prepared cell culture and extracted RNA. HEG assisted with manuscript preparation. 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 Goldring MB, Goldring SR: Osteoarthritis. J Cell Physiol. 2007, 213: 626-634. 10.1002/jcp.21258.CrossRefPubMed Goldring MB, Goldring SR: Osteoarthritis. J Cell Physiol. 2007, 213: 626-634. 10.1002/jcp.21258.CrossRefPubMed
2.
Zurück zum Zitat Samuels J, Krasnokutsky S, Abramson SB: Osteoarthritis: a tale of three tissues. Bull NYU Hosp Jt Dis. 2008, 66: 244-250.PubMed Samuels J, Krasnokutsky S, Abramson SB: Osteoarthritis: a tale of three tissues. Bull NYU Hosp Jt Dis. 2008, 66: 244-250.PubMed
3.
Zurück zum Zitat H Kato, Matsumine A, Wakabayashi T, Hasegawa M, Sudo A, Shintani K, Fukuda A, Kato K, Ide N, Orita S, Hasegawa T, Matsumura C, Furukawa M, Tasaki T, Sonoda H, Uchida A: Large-scale gene expression profiles, differentially represented in osteoarthritic synovium of the knee joint using cDNA microarray technology. Biomarkers. 2007, 12: 384-402. 10.1080/13547500601162482.CrossRef H Kato, Matsumine A, Wakabayashi T, Hasegawa M, Sudo A, Shintani K, Fukuda A, Kato K, Ide N, Orita S, Hasegawa T, Matsumura C, Furukawa M, Tasaki T, Sonoda H, Uchida A: Large-scale gene expression profiles, differentially represented in osteoarthritic synovium of the knee joint using cDNA microarray technology. Biomarkers. 2007, 12: 384-402. 10.1080/13547500601162482.CrossRef
4.
Zurück zum Zitat Sun Y, Mauerhan DR, Firestein GS, Loeffler BJ, Hanley EN, Gruber HE: Telomerase transduced osteoarthritis fibroblast-like synoviocytes display a distinct gene expression profile. J Rheumatol. 2009, 36: 141-155.PubMedPubMedCentral Sun Y, Mauerhan DR, Firestein GS, Loeffler BJ, Hanley EN, Gruber HE: Telomerase transduced osteoarthritis fibroblast-like synoviocytes display a distinct gene expression profile. J Rheumatol. 2009, 36: 141-155.PubMedPubMedCentral
5.
Zurück zum Zitat Galligan CL, Baig E, Bykerk V, Keystone EC, Fish EN: Distinctive gene expression signatures in rheumatoid arthritis synovial tissue fibroblast cells: correlates with disease activity. Genes Immun. 2007, 8: 480-491. 10.1038/sj.gene.6364400.CrossRefPubMed Galligan CL, Baig E, Bykerk V, Keystone EC, Fish EN: Distinctive gene expression signatures in rheumatoid arthritis synovial tissue fibroblast cells: correlates with disease activity. Genes Immun. 2007, 8: 480-491. 10.1038/sj.gene.6364400.CrossRefPubMed
6.
Zurück zum Zitat Hopwood B, Tsykin A, Findlay DM, Fazzalari NL: Microarray gene expression profiling of osteoarthritic bone suggests altered bone remodelling, WNT and transforming growth factor-beta/bone morphogenic protein signalling. Arthritis Res Ther. 2007, 9: R100-10.1186/ar2301.CrossRefPubMedPubMedCentral Hopwood B, Tsykin A, Findlay DM, Fazzalari NL: Microarray gene expression profiling of osteoarthritic bone suggests altered bone remodelling, WNT and transforming growth factor-beta/bone morphogenic protein signalling. Arthritis Res Ther. 2007, 9: R100-10.1186/ar2301.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Chan WP, Lang P, Stevens MP, Sack K, Majumdar S, Stoller DW, Basch C, Genant HK: Osteoarthritis of the knee: comparison of radiography, CT, and MR imaging to assess extent and severity. AJR Am J Roentgenol. 1991, 157: 799-806.CrossRefPubMed Chan WP, Lang P, Stevens MP, Sack K, Majumdar S, Stoller DW, Basch C, Genant HK: Osteoarthritis of the knee: comparison of radiography, CT, and MR imaging to assess extent and severity. AJR Am J Roentgenol. 1991, 157: 799-806.CrossRefPubMed
8.
Zurück zum Zitat Bennett LD, Buckland-Wright JC: Meniscal and articular cartilage changes in knee osteoarthritis: a cross-sectional double-contrast macroradiographic study. Rheumatology. 2002, 41: 917-923. 10.1093/rheumatology/41.8.917.CrossRefPubMed Bennett LD, Buckland-Wright JC: Meniscal and articular cartilage changes in knee osteoarthritis: a cross-sectional double-contrast macroradiographic study. Rheumatology. 2002, 41: 917-923. 10.1093/rheumatology/41.8.917.CrossRefPubMed
9.
Zurück zum Zitat Hunter DJ, Zhang YQ, Niu JB, Tu X, Amin S, Clancy M, Guermazi A, Grigorian M, Gale D, Felson DT: The association of meniscal pathologic changes with cartilage loss in symptomatic knee osteoarthritis. Arthritis & Rheumatism. 2006, 54: 795-801.CrossRef Hunter DJ, Zhang YQ, Niu JB, Tu X, Amin S, Clancy M, Guermazi A, Grigorian M, Gale D, Felson DT: The association of meniscal pathologic changes with cartilage loss in symptomatic knee osteoarthritis. Arthritis & Rheumatism. 2006, 54: 795-801.CrossRef
10.
Zurück zum Zitat Hunter DJ, Zhang YQ, Tu X, Lavalley M, Niu JB, Amin S, Guermazi A, Genant H, Gale D, Felson DT: Change in joint space width: hyaline articular cartilage loss or alteration in meniscus?. Arthritis Rheum. 2006, 54: 2488-2495. 10.1002/art.22016.CrossRefPubMed Hunter DJ, Zhang YQ, Tu X, Lavalley M, Niu JB, Amin S, Guermazi A, Genant H, Gale D, Felson DT: Change in joint space width: hyaline articular cartilage loss or alteration in meniscus?. Arthritis Rheum. 2006, 54: 2488-2495. 10.1002/art.22016.CrossRefPubMed
11.
Zurück zum Zitat Gruber HE, Mauerhan D, Chow Y, Ingram JA, Norton HJ, Hanley EN, Sun Y: Three-dimensional culture of human meniscal cells: extracellular matrix and proteoglycan production. BMC Biotechnol. 2008, 8: 54-10.1186/1472-6750-8-54.CrossRefPubMedPubMedCentral Gruber HE, Mauerhan D, Chow Y, Ingram JA, Norton HJ, Hanley EN, Sun Y: Three-dimensional culture of human meniscal cells: extracellular matrix and proteoglycan production. BMC Biotechnol. 2008, 8: 54-10.1186/1472-6750-8-54.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Pfaffl M, Meyer HH, Sauerwein H: Quantification of insulin-like growth factor-1 (IGF-1) mRNA: development and validation of an internally standardised competitive reverse transcription-polymerase chain reaction. Exp Clin Endocrinol Diabetes. 1998, 106: 506-513. 10.1055/s-0029-1212025.CrossRefPubMed Pfaffl M, Meyer HH, Sauerwein H: Quantification of insulin-like growth factor-1 (IGF-1) mRNA: development and validation of an internally standardised competitive reverse transcription-polymerase chain reaction. Exp Clin Endocrinol Diabetes. 1998, 106: 506-513. 10.1055/s-0029-1212025.CrossRefPubMed
13.
Zurück zum Zitat Reiner A, Yekutieli D, Benjamini Y: Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics. 2003, 19: 368-375. 10.1093/bioinformatics/btf877.CrossRefPubMed Reiner A, Yekutieli D, Benjamini Y: Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics. 2003, 19: 368-375. 10.1093/bioinformatics/btf877.CrossRefPubMed
14.
Zurück zum Zitat Christoforakis J, Pradhan R, Sanchez-Ballester J, Hunt N, Strachan RK: Is there an association between articular cartilage changes and degenerative meniscus tears?. Arthroscopy. 2005, 21: 1366-1369. 10.1016/j.arthro.2005.08.031.CrossRefPubMed Christoforakis J, Pradhan R, Sanchez-Ballester J, Hunt N, Strachan RK: Is there an association between articular cartilage changes and degenerative meniscus tears?. Arthroscopy. 2005, 21: 1366-1369. 10.1016/j.arthro.2005.08.031.CrossRefPubMed
15.
Zurück zum Zitat Carroll GJ, Stuart RA, Armstrong JA, Breidahl PD, Laing BA: Hydroxyapatite crystals are a frequent finding in osteoarthritic synovial fluid, but are not related to increased concentrations of keratan sulfate or interleukin 1 beta. The Journal of Rheumatology. 1991, 18: 861-866.PubMed Carroll GJ, Stuart RA, Armstrong JA, Breidahl PD, Laing BA: Hydroxyapatite crystals are a frequent finding in osteoarthritic synovial fluid, but are not related to increased concentrations of keratan sulfate or interleukin 1 beta. The Journal of Rheumatology. 1991, 18: 861-866.PubMed
16.
Zurück zum Zitat Derfus BA, Kurian JB, Butler JJ, Daft LJ, Carrera GF, Ryan LM, Rosenthal AK: The high prevalence of pathologic calcium crystals in pre-operative knees. J Rheumatol. 2002, 29: 570-574.PubMed Derfus BA, Kurian JB, Butler JJ, Daft LJ, Carrera GF, Ryan LM, Rosenthal AK: The high prevalence of pathologic calcium crystals in pre-operative knees. J Rheumatol. 2002, 29: 570-574.PubMed
17.
Zurück zum Zitat Felson DT, Anderson JJ, Naimark A, Kannel W, Meenan RF: The prevalence of chondrocalcinosis in the elderly and its association with knee osteoarthritis: the Framingham Study. The Journal of Rheumatology. 1989, 16: 1241-1245.PubMed Felson DT, Anderson JJ, Naimark A, Kannel W, Meenan RF: The prevalence of chondrocalcinosis in the elderly and its association with knee osteoarthritis: the Framingham Study. The Journal of Rheumatology. 1989, 16: 1241-1245.PubMed
18.
Zurück zum Zitat Nalbant S, Martinez JA, Kitumnuaypong T, Clayburne G, Sieck M, Schumacher HR: Synovial fluid features and their relations to osteoarthritis severity: new findings from sequential studies. Osteoarthritis and Cartilage. 2003, 11: 50-54. 10.1053/joca.2002.0861.CrossRefPubMed Nalbant S, Martinez JA, Kitumnuaypong T, Clayburne G, Sieck M, Schumacher HR: Synovial fluid features and their relations to osteoarthritis severity: new findings from sequential studies. Osteoarthritis and Cartilage. 2003, 11: 50-54. 10.1053/joca.2002.0861.CrossRefPubMed
19.
Zurück zum Zitat Cheung HS, Mitchell PG, Pledger WJ: Induction of expression of c-fos and c-myc protooncogenes by basic calcium phosphate crystal: effect of beta-interferon. Cancer Res. 1989, 49: 134-138.PubMed Cheung HS, Mitchell PG, Pledger WJ: Induction of expression of c-fos and c-myc protooncogenes by basic calcium phosphate crystal: effect of beta-interferon. Cancer Res. 1989, 49: 134-138.PubMed
20.
Zurück zum Zitat McCarthy GM, Cheung HS, Abel SM, Ryan LM: Basic calcium phosphate crystal-induced collagenase production: role of intracellular crystal dissolution. Osteoarthritis Cartilage. 1998, 6: 205-213. 10.1053/joca.1998.0113.CrossRefPubMed McCarthy GM, Cheung HS, Abel SM, Ryan LM: Basic calcium phosphate crystal-induced collagenase production: role of intracellular crystal dissolution. Osteoarthritis Cartilage. 1998, 6: 205-213. 10.1053/joca.1998.0113.CrossRefPubMed
21.
Zurück zum Zitat Sun Y, Zeng XR, Wenger L, Cheung HS: Basic calcium phosphate crystals stimulate the endocytotic activity of cells--inhibition by anti-calcification agents. Biochem Biophys Res Commun. 2003, 312: 1053-1059. 10.1016/j.bbrc.2003.11.048.CrossRefPubMed Sun Y, Zeng XR, Wenger L, Cheung HS: Basic calcium phosphate crystals stimulate the endocytotic activity of cells--inhibition by anti-calcification agents. Biochem Biophys Res Commun. 2003, 312: 1053-1059. 10.1016/j.bbrc.2003.11.048.CrossRefPubMed
22.
Zurück zum Zitat Cheung HS, Sallis JD, Demadis KD, Wierzbicki A: Phosphocitrate blocks calcification-induced articular joint degeneration in a guinea pig model. Arthritis Rheum. 2006, 54: 2452-2461. 10.1002/art.22017.CrossRefPubMed Cheung HS, Sallis JD, Demadis KD, Wierzbicki A: Phosphocitrate blocks calcification-induced articular joint degeneration in a guinea pig model. Arthritis Rheum. 2006, 54: 2452-2461. 10.1002/art.22017.CrossRefPubMed
23.
Zurück zum Zitat Zhang Y, Brown MA, Peach C, Russell G, Wordsworth BP: Investigation of the role of ENPP1 and TNAP genes in chondrocalcinosis. Rheumatology (Oxford). 2007, 46: 586-589. 10.1093/rheumatology/kel338.CrossRef Zhang Y, Brown MA, Peach C, Russell G, Wordsworth BP: Investigation of the role of ENPP1 and TNAP genes in chondrocalcinosis. Rheumatology (Oxford). 2007, 46: 586-589. 10.1093/rheumatology/kel338.CrossRef
24.
Zurück zum Zitat Canhao H, Fonseca JE, Leandro MJ, Romeu JC, Pimentao JB, Costa JT, Queiroz MV: Cross-sectional study of 50 patients with calcium pyrophosphate dihydrate crystal arthropathy. Clin Rheumatol. 2001, 20: 119-122.CrossRefPubMed Canhao H, Fonseca JE, Leandro MJ, Romeu JC, Pimentao JB, Costa JT, Queiroz MV: Cross-sectional study of 50 patients with calcium pyrophosphate dihydrate crystal arthropathy. Clin Rheumatol. 2001, 20: 119-122.CrossRefPubMed
25.
Zurück zum Zitat Brandes A, Muller KM: [Calcinosis of the meniscus. Morphologic and roentgenographic findings for zonal classification]. Pathologe. 1995, 16: 269-277. 10.1007/s002920050101.CrossRefPubMed Brandes A, Muller KM: [Calcinosis of the meniscus. Morphologic and roentgenographic findings for zonal classification]. Pathologe. 1995, 16: 269-277. 10.1007/s002920050101.CrossRefPubMed
26.
Zurück zum Zitat Habata T, Ohgushi H, Takakura Y, Tohno Y, Moriwake Y, Minami T, Fujisawa Y: Relationship between meniscal degeneration and element contents. Biological Trace Element Research. 2001, 79: 247-256. 10.1385/BTER:79:3:247.CrossRefPubMed Habata T, Ohgushi H, Takakura Y, Tohno Y, Moriwake Y, Minami T, Fujisawa Y: Relationship between meniscal degeneration and element contents. Biological Trace Element Research. 2001, 79: 247-256. 10.1385/BTER:79:3:247.CrossRefPubMed
27.
Zurück zum Zitat Williams CJ, Zhang Y, Timms A, Bonavita G, Caeiro F, Broxholme J, Cuthbertson J, Jones Y, Marchegiani R, Reginato A, Russell RG, Wordsworth BP, Carr AJ, Brown MA: Autosomal dominant familial calcium pyrophosphate dihydrate deposition disease is caused by mutation in the transmembrane protein ANKH. Am J Hum Genet. 2002, 71: 985-991. 10.1086/343053.CrossRefPubMedPubMedCentral Williams CJ, Zhang Y, Timms A, Bonavita G, Caeiro F, Broxholme J, Cuthbertson J, Jones Y, Marchegiani R, Reginato A, Russell RG, Wordsworth BP, Carr AJ, Brown MA: Autosomal dominant familial calcium pyrophosphate dihydrate deposition disease is caused by mutation in the transmembrane protein ANKH. Am J Hum Genet. 2002, 71: 985-991. 10.1086/343053.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Aigner T, Zien A, Gehrsitz A, Gebhard PM, McKenna L: Anabolic and catabolic gene expression pattern analysis in normal versus osteoarthritic cartilage using complementary DNA-array technology. Arthritis Rheum. 2001, 44: 2777-2789. 10.1002/1529-0131(200112)44:12<2777::AID-ART465>3.0.CO;2-H.CrossRefPubMed Aigner T, Zien A, Gehrsitz A, Gebhard PM, McKenna L: Anabolic and catabolic gene expression pattern analysis in normal versus osteoarthritic cartilage using complementary DNA-array technology. Arthritis Rheum. 2001, 44: 2777-2789. 10.1002/1529-0131(200112)44:12<2777::AID-ART465>3.0.CO;2-H.CrossRefPubMed
29.
Zurück zum Zitat Yagami K, Suh JY, Enomoto-Iwamoto M, Koyama E, Abrams WR, Shapiro IM, Pacifici M, Iwamoto M: Matrix GLA protein is a developmental regulator of chondrocyte mineralization and, when constitutively expressed, blocks endochondral and intramembranous ossification in the limb. The Journal of Cell Biology. 1999, 147: 1097-1108. 10.1083/jcb.147.5.1097.CrossRefPubMedPubMedCentral Yagami K, Suh JY, Enomoto-Iwamoto M, Koyama E, Abrams WR, Shapiro IM, Pacifici M, Iwamoto M: Matrix GLA protein is a developmental regulator of chondrocyte mineralization and, when constitutively expressed, blocks endochondral and intramembranous ossification in the limb. The Journal of Cell Biology. 1999, 147: 1097-1108. 10.1083/jcb.147.5.1097.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Taylor BC, Schreiner PJ, Doherty TM, Fornage M, Carr JJ, Sidney S: Matrix Gla protein and osteopontin genetic associations with coronary artery calcification and bone density: the CARDIA study. Hum Genet. 2005, 116: 525-528. 10.1007/s00439-005-1258-3.CrossRefPubMed Taylor BC, Schreiner PJ, Doherty TM, Fornage M, Carr JJ, Sidney S: Matrix Gla protein and osteopontin genetic associations with coronary artery calcification and bone density: the CARDIA study. Hum Genet. 2005, 116: 525-528. 10.1007/s00439-005-1258-3.CrossRefPubMed
31.
Zurück zum Zitat Paine CT, Paine ML, Luo W, Okamoto CT, Lyngstadaas SP, Snead ML: A tuftelin-interacting protein (TIP39) localizes to the apical secretory pole of mouse ameloblasts. J Biol Chem. 2000, 275: 22284-22292. 10.1074/jbc.M000118200.CrossRefPubMed Paine CT, Paine ML, Luo W, Okamoto CT, Lyngstadaas SP, Snead ML: A tuftelin-interacting protein (TIP39) localizes to the apical secretory pole of mouse ameloblasts. J Biol Chem. 2000, 275: 22284-22292. 10.1074/jbc.M000118200.CrossRefPubMed
32.
Zurück zum Zitat Cerny R: [Mineralized dental enamel matrix proteins]. Bratisl Lek Listy. 2000, 101: 288-293.PubMed Cerny R: [Mineralized dental enamel matrix proteins]. Bratisl Lek Listy. 2000, 101: 288-293.PubMed
33.
Zurück zum Zitat Deutsch D, Leiser Y, Shay B, Fermon E, Taylor A, Rosenfeld E, Dafni L, Charuvi K, Cohen Y, Haze A, Fuks A, Mao Z: The human tuftelin gene and the expression of tuftelin in mineralizing and nonmineralizing tissues. Connect Tissue Res. 2002, 43: 425-434.CrossRefPubMed Deutsch D, Leiser Y, Shay B, Fermon E, Taylor A, Rosenfeld E, Dafni L, Charuvi K, Cohen Y, Haze A, Fuks A, Mao Z: The human tuftelin gene and the expression of tuftelin in mineralizing and nonmineralizing tissues. Connect Tissue Res. 2002, 43: 425-434.CrossRefPubMed
34.
Zurück zum Zitat Ijiri K, Zerbini LF, Peng H, Otu HH, Tsuchimochi K, Otero M, Dragomir C, Walsh N, Bierbaum BE, Mattingly D, van FG, Komiya S, Aigner T, Libermann TA, Goldring MB: Differential expression of GADD45beta in normal and osteoarthritic cartilage: potential role in homeostasis of articular chondrocytes. Arthritis Rheum. 2008, 58: 2075-2087. 10.1002/art.23504.CrossRefPubMedPubMedCentral Ijiri K, Zerbini LF, Peng H, Otu HH, Tsuchimochi K, Otero M, Dragomir C, Walsh N, Bierbaum BE, Mattingly D, van FG, Komiya S, Aigner T, Libermann TA, Goldring MB: Differential expression of GADD45beta in normal and osteoarthritic cartilage: potential role in homeostasis of articular chondrocytes. Arthritis Rheum. 2008, 58: 2075-2087. 10.1002/art.23504.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Appleton CT, Pitelka V, Henry J, Beier F: Global analyses of gene expression in early experimental osteoarthritis. Arthritis Rheum. 2007, 56: 1854-1868. 10.1002/art.22711.CrossRefPubMed Appleton CT, Pitelka V, Henry J, Beier F: Global analyses of gene expression in early experimental osteoarthritis. Arthritis Rheum. 2007, 56: 1854-1868. 10.1002/art.22711.CrossRefPubMed
36.
Zurück zum Zitat Sato T, Konomi K, Yamasaki S, Aratani S, Tsuchimochi K, Yokouchi M, Masuko-Hongo K, Yagishita N, Nakamura H, Komiya S, Beppu M, Aoki H, Nishioka K, Nakajima T: Comparative analysis of gene expression profiles in intact and damaged regions of human osteoarthritic cartilage. Arthritis Rheum. 2006, 54: 808-817. 10.1002/art.21638.CrossRefPubMed Sato T, Konomi K, Yamasaki S, Aratani S, Tsuchimochi K, Yokouchi M, Masuko-Hongo K, Yagishita N, Nakamura H, Komiya S, Beppu M, Aoki H, Nishioka K, Nakajima T: Comparative analysis of gene expression profiles in intact and damaged regions of human osteoarthritic cartilage. Arthritis Rheum. 2006, 54: 808-817. 10.1002/art.21638.CrossRefPubMed
37.
Zurück zum Zitat Plaas A, Osborn B, Yoshihara Y, Bai Y, Bloom T, Nelson F, Mikecz K, Sandy JD: Aggrecanolysis in human osteoarthritis: confocal localization and biochemical characterization of ADAMTS5-hyaluronan complexes in articular cartilages. Osteoarthritis Cartilage. 2007, 15: 719-734. 10.1016/j.joca.2006.12.008.CrossRefPubMed Plaas A, Osborn B, Yoshihara Y, Bai Y, Bloom T, Nelson F, Mikecz K, Sandy JD: Aggrecanolysis in human osteoarthritis: confocal localization and biochemical characterization of ADAMTS5-hyaluronan complexes in articular cartilages. Osteoarthritis Cartilage. 2007, 15: 719-734. 10.1016/j.joca.2006.12.008.CrossRefPubMed
38.
Zurück zum Zitat Majumdar MK, Askew R, Schelling S, Stedman N, Blanchet T, Hopkins B, Morris EA, Glasson SS: Double-knockout of ADAMTS-4 and ADAMTS-5 in mice results in physiologically normal animals and prevents the progression of osteoarthritis. Arthritis Rheum. 2007, 56: 3670-3674. 10.1002/art.23027.CrossRefPubMed Majumdar MK, Askew R, Schelling S, Stedman N, Blanchet T, Hopkins B, Morris EA, Glasson SS: Double-knockout of ADAMTS-4 and ADAMTS-5 in mice results in physiologically normal animals and prevents the progression of osteoarthritis. Arthritis Rheum. 2007, 56: 3670-3674. 10.1002/art.23027.CrossRefPubMed
39.
Zurück zum Zitat Chia SL, Sawaji Y, Burleigh A, McLean C, Inglis J, Saklatvala J, Vincent T: Fibroblast growth factor 2 is an intrinsic chondroprotective agent that suppresses ADAMTS-5 and delays cartilage degradation in murine osteoarthritis. Arthritis Rheum. 2009, 60: 2019-2027. 10.1002/art.24654.CrossRefPubMed Chia SL, Sawaji Y, Burleigh A, McLean C, Inglis J, Saklatvala J, Vincent T: Fibroblast growth factor 2 is an intrinsic chondroprotective agent that suppresses ADAMTS-5 and delays cartilage degradation in murine osteoarthritis. Arthritis Rheum. 2009, 60: 2019-2027. 10.1002/art.24654.CrossRefPubMed
40.
Zurück zum Zitat Davidson RK, Waters JG, Kevorkian L, Darrah C, Cooper A, Donell ST, Clark IM: Expression profiling of metalloproteinases and their inhibitors in synovium and cartilage. Arthritis Res Ther. 2006, 8: R124-10.1186/ar2013.CrossRefPubMedPubMedCentral Davidson RK, Waters JG, Kevorkian L, Darrah C, Cooper A, Donell ST, Clark IM: Expression profiling of metalloproteinases and their inhibitors in synovium and cartilage. Arthritis Res Ther. 2006, 8: R124-10.1186/ar2013.CrossRefPubMedPubMedCentral
Metadaten
Titel
Analysis of meniscal degeneration and meniscal gene expression
verfasst von
Yubo Sun
David R Mauerhan
Patrick R Honeycutt
Jeffrey S Kneisl
James H Norton
Edward N Hanley Jr
Helen E Gruber
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
BMC Musculoskeletal Disorders / Ausgabe 1/2010
Elektronische ISSN: 1471-2474
DOI
https://doi.org/10.1186/1471-2474-11-19

Weitere Artikel der Ausgabe 1/2010

BMC Musculoskeletal Disorders 1/2010 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Arthroskopie kann Knieprothese nicht hinauszögern

25.04.2024 Gonarthrose Nachrichten

Ein arthroskopischer Eingriff bei Kniearthrose macht im Hinblick darauf, ob und wann ein Gelenkersatz fällig wird, offenbar keinen Unterschied.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Ärztliche Empathie hilft gegen Rückenschmerzen

23.04.2024 Leitsymptom Rückenschmerzen Nachrichten

Personen mit chronischen Rückenschmerzen, die von einfühlsamen Ärzten und Ärztinnen betreut werden, berichten über weniger Beschwerden und eine bessere Lebensqualität.

Update Orthopädie und Unfallchirurgie

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