Skip to main content
Erschienen in: Arthritis Research & Therapy 2/2005

Open Access 01.12.2004 | Research article

Gene expression profiling in murine autoimmune arthritis during the initiation and progression of joint inflammation

verfasst von: Vyacheslav A Adarichev, Csaba Vermes, Anita Hanyecz, Katalin Mikecz, Eric G Bremer, Tibor T Glant

Erschienen in: Arthritis Research & Therapy | Ausgabe 2/2005

Abstract

We present here an extensive study of differential gene expression in the initiation, acute and chronic phases of murine autoimmune arthritis with the use of high-density oligonucleotide arrays interrogating the entire mouse genome. Arthritis was induced in severe combined immunodeficient mice by using adoptive transfer of lymphocytes from proteoglycan-immunized arthritic BALB/c mice. In this unique system only proteoglycan-specific lymphocytes are transferred from arthritic mice into syngeneic immunodeficient recipients that lack adaptive immunity but have intact innate immunity on an identical (BALB/c) genetic background.
Differential gene expression in response to donor lymphocytes that migrated into the joint can therefore be monitored in a precisely timed manner, even before the onset of inflammation. The initiation phase of adoptively transferred disease (several days before the onset of joint swelling) was characterized by differential expression of 37 genes, mostly related to chemokines, interferon-γ and tumor necrosis factor-α signaling, and T cell functions. These were designated early arthritis 'signature' genes because they could distinguish between the naive and the pre-arthritic state. Acute joint inflammation was characterized by at least twofold overexpression of 256 genes and the downregulation of 21 genes, whereas in chronic arthritis a total of 418 genes with an equal proportion of upregulated and downregulated transcripts were expressed differentially.
Hierarchical clustering and functional classification of inflammation-related and arthritis-related genes indicated that the most common biological activities were represented by genes encoding interleukins, chemokine receptors and ligands, and by those involved in antigen recognition and processing.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​ar1472) contains supplementary material, which is available to authorized users.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

VAA performed essentially all statistical analyses and put together the draft version of the results and figures. CV isolated all RNA samples, prepared biotinylated samples and was involved in Affymetrix hybridization experiments; he also performed preliminary clustering experiments with GeneSpring version 6.2 (not included in this paper). AH performed all in vitro stimulation and adoptive transfer experiments, and assessed arthritis three or four times a day together with KM, who was also involved in all phases of the experimental processes and in the finalization of the manuscript. EGB controlled Affymetrix hybridization and scanning experiments, managed preliminary data analysis and finalized the manuscript. TTG designed experiments, controlled all experimental steps, data analysis, and finalized the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AA
acutely arthritic
AN
absolutely negative (control naive)
CA
chronically arthritic
CV
coefficient of variation
DDA
dimethyldioctadecylammonium bromide
PA
pre-arthritic
PG
cartilage proteoglycan aggrecan
PGIA
PG-induced arthritis
RA
rheumatoid arthritis
SCID
severe combined immunodeficient.

Introduction

The completion of the human and mouse genome sequencing programs and the subsequent annotation of previously unidentified genes have opened a new epoch in biology and biomedical sciences. The genetic information greatly facilitated the discovery of novel disease-related genes and the mapping of signature genes for early diagnosis. More specifically, polynucleotide or oligonucleotide arrays have been applied in both human and experimentally induced disease conditions to determine characteristic expression patterns of signature genes.
In an inflammatory disease such as rheumatoid arthritis (RA), the gene expression profile is extremely complex owing to the diversity of cell types involved in the pathology and the polygenic character of the autoimmune disease [15]. The overall picture of molecular interactions in an inflamed joint, deduced from gene expression studies in both RA and its corresponding animal models, involves proteins participating in immunity, inflammation, apoptosis, proliferation, cellular transformation and cell differentiation, and other processes [38]. Several studies analyzed the patterns of gene expression in peripheral blood or synovial fluid mononuclear cells, and in the inflamed synovium of human patients [1, 35, 7, 911]. However, the genetic heterogeneity of the human population is a serious obstacle to the correct interpretation of data in gene expression studies. Animal models of RA can facilitate the interpretation of genome-wide gene expression by providing genetic and clinical homogeneity, and an opportunity to monitor the onset and progression of the disease [1220]. DNA microarray technology was successfully applied to inflamed paws of mice or rats systemically immunized with arthritogenic compounds to induce arthritis [6, 2123]. Despite the usefulness of the information provided by these studies, the early gene expression events at the site of inflammation (joint and synovium) and the mechanisms of disease initiation remain unknown.
Systemic immunization of genetically susceptible BALB/c mice with human cartilage proteoglycan aggrecan (PG) induces PG-specific immune responses that then trigger inflammation in peripheral joints [13, 19]. PG-induced arthritis (PGIA) is a murine model which bears many similarities to RA as indicated by clinical assessments, radiographic analyses, various laboratory and functional tests, and by histopathologic studies of diarthrodial joints [13, 19, 24, 25]. Moreover, genome-wide screening studies identified multiple genomic loci in PGIA [20, 2629] that are syntenic with those described in RA [25]. Both RA and PGIA are polygenic autoimmune diseases with a major permissive role of the MHC, although non-MHC genes account for a significant portion of the genetic susceptibility. PGIA can be successfully transferred into naive BALB/c or syngeneic severe combined immunodeficient (SCID) mice either with unseparated spleen cells or with antigen (PG)-stimulated T lymphocytes from arthritic donor BALB/c mice [3032].
In the present study, we adoptively transferred the disease (PGIA) into syngeneic BALB/cSCID mice lacking functional T and B cells. SCID mice carry a natural mutation that prevents the V(D)J recombination in B and T lymphocytes, resulting in a failure to generate functional immunoglobulins and T cell receptors [33, 34]. Consequently, adoptively transferred arthritis in BALB/cSCID mice is an ideal model in which activated lymphocytes of arthritic donor BALB/c mice migrate and interact with the intact innate immunity environment in the joints of BALB/cSCID mice. The gene expression profiles in normal, pre-arthritic and arthritic joints of the recipient BALB/cSCID mice were determined by using DNA microarray technology (Affymetrix). Although a significant number of genes were differentially expressed in joints with acute and chronic arthritis, in this study we focused on early genes whose expression occurred before the onset of clinical symptoms.

Methods

Animals, antigen and immunization

The use of human cartilage from joint replacement surgeries for antigen isolation was approved by the Institutional Review Board, and all animal experiments were approved by the Institutional Animal Care and Use Committee. Female BALB/c mice at the age of 24–26 weeks (National Cancer Institute, Kingston Colony, New York, USA) were injected intraperitoneally with 100 μg of cartilage PG (measured as protein) emulsified in dimethyldioctadecylammonium bromide (DDA) adjuvant (Sigma-Aldrich, St Louis, Missouri, USA). The use of adjuvant DDA allowed us to avoid the harmful effects of oil and bacterial proteins present in Freund's adjuvants [35, 36]. Booster injections of the same doses of PG with DDA were given on days 21 and 42. BALB/c mice develop swelling and redness of one or more limbs 7–10 days after the second or third injection with PG in adjuvant [25]. Arthritis was assessed daily, and inflammation was scored from grade 0 to grade 4 for each paw [13, 36, 37]. Female SCID mice of the BALB/c background (NCI/NCrC.B-17-scid/scid; henceforth BALB/cSCID) were used for adoptive cell transfer. BALB/cSCID mice were purchased from the National Cancer Institute and maintained under germ-free conditions.

Stimulation of lymphocytes in vitro, and adoptive transfer of arthritis

To ensure uniformity and reproducibility of disease transfer, donor spleen cells were isolated from arthritic BALB/c mice within 1–2 weeks after the onset of inflammation. At least two paws of donor BALB/c mice were arthritic, and the cumulative inflammation score (for four paws) was in the range 5–8. Spleen cells of arthritic BALB/c mice were collected and cultured in six-well plates (2.5 × 106 cells/ml) with cartilage PG (50 μg/ml) for 4 days in Dulbecco's modified Eagle's medium supplemented with 5% fetal bovine serum (HyClone Laboratories, Logan, Utah, USA). After stimulation in vitro for 4 days with cartilage PG, non-adherent cells were collected, and live cells (lymphocytes) were separated on Lympholyte-M (Cedarlane, Ontario, Canada). Finally, 2 × 107 lymphocytes were injected intraperitoneally on days 0 and 7 into recipient BALB/cSCID mice as described [32].
A standard scoring system used for primary arthritis was applied to the assessment of disease severity in BALB/cSCID mice [24, 37]. Typically, one to four paws became inflamed simultaneously 3–5 days after the second cell transfer, and the rest of the peripheral joints became inflamed within 2–4 days after the onset of the first symptoms. BALB/cSCID mice were scored twice daily, and were killed as soon as the inflamed paw reached an individual arthritis score of 2, but not later than 24 hours after the onset of arthritis. This paw was designated as acute arthritic (AA), and contralateral or ipsilateral paws that were not inflamed at that time were used as pre-arthritic (PA) samples. The PA joints did not show evidence of inflammation on histopathological examination, although thickening of the synovial lining in small joints was observed occasionally (data not shown). Several arthritic BALB/cSCID mice were scored daily and were killed 8–10 days after disease onset. These joint samples represented subacute-chronically arthritic (CA) samples. In addition to PA, AA and CA experimental conditions, paws of naive non-immunized BALB/cSCID mice were used as 'absolutely negative' (control naive; AN) samples for RNA isolation and subsequent hybridization. Each sample represented RNA pooled from four paws of two mice.

Probe preparation

Synthesis and biotinylation of cRNA and hybridization were performed in accordance with the manufacturer's instructions (Affymetrix, Santa Clara, California, USA). In brief, total RNA was isolated from normal or inflamed paws of mice by using TRIzol reagent (Invitrogen, Gaithersburg, Maryland, USA) with additional purification on RNeasy columns (Qiagen, Valencia, California, USA). RNA quality was confirmed by spectrophotometry and electrophoresis on formaldehyde gels [38]. Double-stranded complementary DNA was synthesized with the T7-dT24 primer incorporating a T7 RNA polymerase promoter. Biotinylated cRNA was prepared with the Enzo BioArray High Yield RNA Transcript Labeling Kit (Enzo Diagnostics, Inc., Farmingdale, New York, USA) and hybridized to the murine genome Affymetrix U74v2 chip set, which included three DNA chips, MG_U74Av2, MG_U74Bv2 and MG_U74Cv2, interrogating more than 36,000 genes that represented essentially the entire mouse genome [3942]. Fluorescent hybridization signals were developed with phycoerythrin-conjugated streptavidin and were further enhanced with fluorescently labeled anti-streptavidin antibodies. DNA chips were scanned to obtain quantitative gene expression levels. DNA chip hybridization, Fluidics Station operations, scanning, and preliminary data management were performed in accordance with Affymetrix protocols as described previously [43, 44].

Microarray analysis

Fluorescent intensity data from Affymetrix Microarray Suite version 5 were exported as CEL files and imported into DNA-Chip Analyzer version 1.3 [45]. Data were normalized, and expression values, based on the perfect match/mismatch (PM/MM) model, were calculated for each DNA chip. All chips were examined for the image spikes, chip and gene outliers. Exported expression values for each DNA chip were combined into a single file (three chips × four experimental conditions × three to five replicates), and imported back to DNA-Chip Analyzer; the resulting data were normalized by using an array with median probe intensity.
For the pairwise comparison of experimental conditions, signals were filtered by using several criteria. Gene expression was considered above the background if it showed the signal on most chips (more than 50%; that is, for three replicates, the gene should be detectable on at least two chips; for five replicates, the gene should be present on at least three DNA chips). Fold changes for gene expression were calculated when any of three following criteria were met: (1) the gene was present in the experimental condition but absent in the basal condition; (2) the gene was present in the basal condition but absent in the experimental condition; (3) the gene was present in both basal condition and experimental conditions. Student's t-test was used to determine the statistical significance of the difference in gene expression between basal and experimental conditions (P < 0.05 was taken as significant). An additional cut-off threshold of twofold change in gene expression (either upregulation or downregulation) was used to characterize a gene as being differentially regulated (for example, a negative twofold value corresponded to a twofold downregulation). The Fisher exact test (implemented by us in Visual Basic code for MS Excel 2000) and the Mann–Whitney U-test (SPSS, Chicago, Illinois, USA) were used to verify non-paired Student's t-test calculations of the probability of gene expression differences in pairwise comparisons. Finally, the false discovery rate was established with 500 permutations for each pairwise comparison to estimate the proportion of false-positive genes.
To characterize gene expression patterns, hierarchical gene clustering was performed with a DNA-Chip Analyzer program [45, 46]. The algorithm was based on the distance between two genes defined as 1 - r, where r is the Pearson correlation coefficient between the standardized expression values of the two genes across the samples used. To characterize functional relationships between differentially expressed genes, Gene Ontology terms classification [47], incorporated in DNA-Chip Analyzer, was performed [48]. The significance level for a functional cluster was set at P < 0.05, and the minimum size of a cluster was three genes. Venn diagram calculations were performed in Visual Basic code for MS Excel 2000 to analyze overlapping of sets of genes differentially expressed in the samples at different phases of arthritis.

Results

The major goal of the present study was to find and characterize early signature genes whose expressions were different (at least twofold change in the threshold level) and statistically significant (P < 0.05) between experimental groups at different phases of joint inflammation. The induction of arthritis in BALB/cSCID mice was a multi-step process. First, donor BALB/c mice were immunized with cartilage PG to induce arthritis. Second, spleen cells from acutely arthritic (AA) donor mice were stimulated in vitro with cartilage PG, and live lymphocytes were isolated on a Lympholyte-M density gradient. Third, these antigen-stimulated donor lymphocytes were injected into BALB/cSCID mice. For gene expression profiling during the time course of the adoptively transferred arthritis, RNA was isolated from pre-arthritic paws (PA) and diseased paws (AA and CA) (Table 1). In addition, RNA was isolated from normal paws of naive BALB/cSCID mice and served as a baseline non-arthritic control condition (AN). Three pairwise comparisons were performed: PA versus AN, AA versus AN, and CA versus AN (hereafter denoted as PA/AN, AA/AN and CA/AN).
Table 1
Experimental groups used for adoptive disease transfer and differential expression analysis
Group
RNA source
Treatment
Days after injection
No. of animals
AN
Naive control (absolute negative) BALB/cSCID paw
None
N/A
3
PA
Normal paw from arthritic BALB/cSCID mouse
Cell transfer
6
3
AA
Acute arthritic paw of BALB/cSCID mouse
Cell transfer
6
5
CA
Chronically arthritic paw of BALB/cSCID mouse
Cell transfer
12–14
3
Group AN represents naive BALB/cSCID mice that received no cells. Experimental groups PA, AA, and CA received antigen-stimulated lymphocytes from arthritic BALB/c donor mice. RNA was isolated from four paws of two mice at the indicated number of days after injection, and pooled.
Each experimental condition was reproduced three to five times (RNA isolation, probe preparation, and independent hybridizations), and each replicate contained RNA samples pooled from a total of four paws of two arthritic animals. When the number of replicates is low and the distribution of data in the general population is basically unknown, the applicability of Student's t-test is questionable. We therefore analyzed data by using both Student's t-test and the Fisher exact test, in which the first approach requires normal data distribution, whereas the second test does not have this requirement [45, 49, 50]. Setting the significance level for the difference between groups at P < 0.05 and no threshold for the fold change in expression, 1805 genes passed the Fisher exact test and 1752 genes passed the DNA-Chip Analyzer Student's t-test [45] for the PA/AN comparison. In AA/AN pairwise comparisons, 3676 genes passed the Fisher exact test and 3305 genes passed Student's t-test. Concluding that Student's t-test provided similar results and was even more conservative than the Fisher exact test, we employed the former for all further analyses.

Effect of the numbers of replicates on data variability

Being aware of the importance of data reproducibility, we determined the optimal number of arrays to be included in experimental design by monitoring the convergence of variance for gene expression signals in five replicates representing the condition AA. For each replicate, we pooled equal amounts of quality-controlled RNA samples, isolated from two inflamed paws of two BALB/cSCID mice that had been identically treated (in terms of the number of donor cells and antigen stimulation) and had similar disease onset and severity. A total of five replicates represented 20 paws of 10 arthritic mice. We used the coefficient of variation (CV) to measure data variability. The CV for each gene on the chip and the mean CV for the entire probe set were calculated. Mean CV reached a plateau when the number of replicates increased beyond three (Fig. 1, experimental condition AA) and there was no significant change afterwards. Therefore, for all other experimental conditions, we used three replicates representing three independent hybridization experiments of three RNA samples isolated from six paws. Mean CV after sampling of the three repeats ranged between 0.21 and 0.25 for all experimental conditions.

Arthritis 'signature' genes in pre-inflamed joints

Paws of naive BALB/cSCID mice and still non-inflamed (PA) paws were clinically normal with no sign of inflammation, and comparison of these two experimental conditions (PA/AN) identified a relatively small number of differentially expressed genes. Only 37 of the 36,000 screened genes were differentially expressed (that is, showed greater than a ± twofold change relative to threshold level), of which 11 genes were over the ± threefold threshold, and seven genes changed beyond ± fivefold (Fig. 2). The seven genes with the most significant change in expression levels encoded chemokine CC motif receptor 5 (Ccr5), chemokine CXC motif ligand 1 (Cxcl1), interferon-γ-inducible protein (Ifi47), membrane-spanning 4-domains subfamily A member 6C (Ms4a6c), tumor necrosis factor-α-induced protein 6 (Tnfip6), T cell receptor β variable 13 (Tcrbv13), and Terf1-interacting nuclear factor2 (Tinf2) (Table 2). Although the upregulation of Tcrbv13, Tgtp and interferon-induced genes might indicate the appearance of antigen-specific T cells in the synovium (Table 2), the significant upregulation of Tnfip6 suggests the activation of an anti-inflammatory cascade [51]. Thus, gene expression related to pro-inflammatory and anti-inflammatory events can be detected even before the migration of inflammatory leukocytes into the joints.
Table 2
Array-based expression values of upregulated or downregulated genes in pre-inflamed joint
Affy ID
Description
Gene
Mean AN expression
AN presence call
Mean PA expression
PA presence call
Fold
Cluster
161968_f_at
Chemokine (CC motif) receptor 5
Ccr5
1
A
57.1
P
57.1
D
95349_g_at
Chemokine (CXC motif) ligand 1
Cxcl1
1
A
55.9
P
55.9
D
104750_at
Interferon-γ inducible protein
Ifi47
0.69
A
15.69
P
22.7
D
130509_at
Membrane-spanning 4-domains member A6C
Ms4a6c
1.17
A
10.27
P
8.78
D
93106_i_at
T-cell receptor beta, variable 13
Tcrbv13
1.86
A
10.34
P
5.55
A
98474_r_at
Tumor necrosis factor-α induced protein 6
Tnfaip6
3.61
A
19.97
P
5.54
D
94761_at
Chemokine (CC motif) ligand 7
Ccl7
24.47
A
115.33
P
4.71
D
101578_f_at
Actin, β, cytoplasmic
Actg
188.33
P
859.16
P
4.56
A
102736_at
Chemokine (CC motif) ligand 2
Ccl2
29.89
P
125.48
P
4.20
D
95121_at
Polymerase (DNA-directed) ε 4 p12
Pole4
8.95
P
26.13
P
2.92
C
93397_at
Chemokine (CC) receptor 2
Ccr2
93.67
P
259.21
P
2.77
D
102906_at
T cell-specific GTPase
Tgtp
24.27
A
66.03
P
2.72
D
97322_at
Membrane-spanning 4-domains member A6B
Ms4a6b
9.43
A
25.16
P
2.67
D
93514_at
Myosin, light polypeptide 3
Myl3
29.28
A
77.42
P
2.64
A
103089_at
CD48 antigen
Cd48
34.17
A
85.82
P
2.51
C
103507_at
EGF-like hormone receptor-like sequence 1
Emr1
59.04
A
145.95
P
2.47
D
96764_at
Interferon-inducible GTPase
Ifigtp
66.38
A
157.88
P
2.38
D
102326_at
Neutrophil cytosolic factor 2
Ncf2
23.67
P
55.88
P
2.36
D
104388_at
Chemokine (CC motif) ligand 9
Ccl9
246.56
P
568.47
P
2.31
D
93321_at
Interferon-activated gene 203
Ifi203
43.58
P
99.27
P
2.28
C
94085_at
Proteoglycan, secretory granule
Prg
329.68
P
747.2
P
2.27
D
92762_at
C-type lectin, superfamily member 6
Clecsf6
35.73
A
80.64
P
2.26
D
93136_at
Dermatan sulphate proteoglycan 3
Dspg3
30.23
A
67.77
P
2.24
B
101753_s_at
P lysozyme structural
Lzps
635.78
P
1398.72
P
2.20
D
94939_at
CD53 antigen
Cd53
112.16
P
243.17
P
2.17
D
94958_at
RIKEN cDNA 1110013L07 gene
1110013L07Rik
24.91
A
53.7
P
2.16
A
162066_f_at
Rho interacting protein 3
Rip3
19.15
A
40.54
P
2.12
A
93039_at
RIKEN cDNA 1190003P12 gene
1190003P12Rik
36.44
P
77.03
P
2.11
A
101048_at
Protein tyrosine phosphatase, receptor type, C
Ptprc
138.7
A
289.19
P
2.09
D
92217_s_at
Glycoprotein 49 B
Gp49b
72.11
P
148.01
P
2.05
D
93869_s_at
Hematopoietic-specific A1-d protein
Bcl2a1a
49.43
A
100.11
P
2.03
D
103989_at
RIKEN cDNA 4432417F03 gene
4432417F03Rik
35.66
A
72.18
P
2.02
B
160611_at
Cytochrome P450 polypeptide 4v3
Cyp4v3
82
P
164.59
P
2.01
B
162107_r_at
Tissue inhibitor of metalloproteinase 1
Timp1
9.64
P
4.39
A
-2.20
E
164493_i_at
Makorin, ring finger protein, 1
Mkrn1
8.23
P
3.21
A
-2.56
F
167637_i_at
RIKEN cDNA 4833424O15 gene
4833424O15Rik
8.19
P
2.64
A
-3.11
F
167950_r_at
Terf1 (TRF1)-interacting nuclear factor 2
Tinf2
8.82
P
1.65
P
-5.35
E
Affy ID, unique Affymetrix probe set identifier. Description, gene description. Gene, gene abbreviation. Mean AN expression, average expression value in basal experimental condition of clinically normal paws of naive severe combined immunodeficient mice without cell transfer. Mean PA expression, average expression value in pre-arthritic joints. Presence call, average presence call for gene in AN or PA experimental condition: P, transcript was actually present in the majority of samples; A, transcript was actually absent in the majority of samples. Fold, fold change in gene expression in PA joint compared with AN basal expression. Cluster, cluster designation from Fig. 5. Difference in expression was significant by Mann–Whitney U-test, P < 0.05. Differential expression for listed genes was either greater than twofold overexpression or less than twofold downregulation (negative values). Genes that were differentially expressed in both pre-inflamed paws and in vitro-stimulated lymphocytes used for cell transfer are shown in bold type.
To characterize major biological functions in context with the initiation phase of the disease, we assigned the 37 early genes (Table 2, Additional file 1) to separate groups according to the corresponding protein functions and Gene Ontology classification [47, 48]. We found that differentially expressed genes in PA joints were related to immune responses, chemokine activity (including chemotaxis), cell adhesion, proteolysis regulation, inflammation and wounding, cytokines, and cytoskeletal activity (Fig. 3, yellow circles). All clustered genes were upregulated at the pre-inflamed phase of arthritis.

Gene expression profile in acute and chronic arthritis

To monitor the progression of disease, we analyzed genes that were differentially expressed in paws with acute and chronic joint inflammation. Both AA and CA experimental conditions were associated with the activity of a large number of genes: 256 genes were upregulated and 21 were downregulated in acute arthritis (AA/AN comparison), and 201 genes were upregulated and 217 were downregulated in chronic inflammation (CA/AN) (Fig. 2, Additional files 2 and 3). A Venn diagram summarizes the relationships between gene sets that were differentially expressed at different phases of the disease. Only 15 genes were differentially expressed in all three phases of the disease (PA, AA, and CA), 25 genes were differentially expressed both at the PA phase and during acute inflammation, 127 genes were active both in acute and chronic phases, and 17 transcripts shared a common expression pattern in pre-inflamed and chronically inflamed joints (Fig. 2).
Using Gene Ontology terms for the functional classification of genes differentially expressed in acute and chronic arthritis [47], dozens of cell signaling pathways and gene clusters were identified. By further filtering of functional clusters, and by combining clusters encoding proteins with similar functions, we found that the acute and chronic phases of the disease can be comprehensively described by the differential expression of 15 macro-clusters (Fig. 3). Six clusters were found in all three phases of inflammation; they were related to immune response, chemokine activity, cytokines, inflammation and wounding, cell adhesion, and proteolysis regulation. The most abundantly represented genes in inflamed joints were those involved in immune responses: 51 genes in AA and 25 genes in CA. These genes were upregulated as much as 31-fold (group average) in acute arthritis and 15-fold in chronic arthritis (Fig. 3). Cytokine and chemokine genes demonstrated the highest overexpression levels: about 64-fold in acute and 28-fold in chronic arthritis, where both groups included more than a dozen genes. Proteolysis-regulating genes (proteases and their inhibitors) were highly represented at the acute phase (45 genes), but were less abundant in chronic arthritis (19 genes). Extracellular matrix-related genes, mostly relevant to tissue repair and healing, were more abundant in chronic than acute disease. Some functional clusters were phase-specific, such as lysosome, antigen presentation, scavenger receptors, immunoglobulin binding, and complement cascade; these genes were preferentially expressed in acute joint inflammation. Suppression of genes related to the respiratory chain complex was specific to chronic inflammation (Fig. 3).

Hierarchical clustering of arthritis phase-specific genes

To identify genes whose expression might be specific for the actual phase of arthritis, and to combine transcripts by the pattern of their expression through all disease phases, we applied a hierarchical clustering technique [46]. Genes that were specific for pairwise comparisons (PA/AN, AA/AN, and CA/AN) were combined into one single file (excluding redundant genes); the merged set included 507 genes. Hierarchical clustering was performed for all experimental conditions studied (AN, PA, AA, and CA), and four major gene clusters were identified, each with a distinct expression pattern (Fig. 4, clusters I–IV). Using further classification analysis with Gene Ontology terms, to examine the functions of genes inside each cluster, we identified genes encoding proteins whose biological functions were the most relevant to arthritis development and progression.
Cluster I contained genes with major functions in collagen turnover and tissue repair; the expression of these genes reached a peak in chronically inflamed joints.
Cluster II was the largest cluster including about half of all phase-specific genes (Fig. 4). The cluster included genes with roles in immune, inflammatory and stress responses, extracellular matrix formation, cell growth, and receptor activity. The expression of cluster II genes reached a peak at the acute phase of joint inflammation.
Transcription of genes in clusters III and IV gradually decreased during disease progression (Fig. 4). These genes were mostly related to cytoskeleton remodeling, the formation of cell junctions, and the production of structural molecules such as desmin, β-3 laminin, envoplakin, and dystonin (for a detailed gene list see Additional files 1, 2, 3). Genes associated with early arthritis (Table 2) were found in clusters III and IV, further underlining the importance of cell adhesion and cytoskeleton remodeling during the initiation phase of arthritis.

Expression patterns of early arthritis genes

Hierarchical clustering of a large number of phase-specific genes (n = 507) (Fig. 4) obscured the expression pattern of a relatively small number (n = 37) of early arthritis genes (Table 2). A separate hierarchical clustering was therefore performed for these 37 early genes, and the levels of expression were monitored at later phases of the disease. Six distinct expression patterns were identified (Fig. 5, clusters A–F) using this approach. Clusters A–D contained early arthritis genes whose transcription increased as the disease progressed, reaching a peak in the pre-inflamed joint or during inflammation. Cluster A included genes that coded for variable parts of the T cell receptor, together with genes related to cytoskeleton reorganization such as Rho interacting protein 3, myosin, and β-actin (reviewed in [52, 53]). Cluster A genes were at the peak of their expression in the PA joint. However, most early arthritis genes in clusters C and D showed an expression peak later, at the acute phase of inflammation (Fig. 5), and encoded chemokine receptors (Ccr2 and Ccr5) and chemokine ligands (Cxcl1, Ccl2, Ccl7, and Ccl9). Clusters C and D also included interferon-activating genes Ifi203, Ifi47, and Ifigtp, and cell differentiation antigens such as CD48 and CD53.
Hierarchical clusters E and F contained four genes whose expression was downregulated in the pre-inflamed joint but returned to a 'normal' level (as expressed in naive paws) during arthritis progression. Clusters E and F included genes encoding Terf1-interacting nuclear factor 2, tissue inhibitor of metalloproteinase 1, makorin, and DNA clone 4833424O15 with unknown function (Table 2 and Fig. 5).

Discussion

This study describes genome-wide gene activity taking place in mouse joints during three major phases of autoimmune arthritis: initiation, acute inflammation, and chronic inflammation. Spleen cells from PG-immunized arthritic BALB/c mice were used to transfer the disease into non-immunized syngeneic SCID mice [30, 32]. This adoptive transfer system minimized the individual differences that are typical in primary arthritis (induced by systemic immunization), and also excluded antigen-independent stimulation of the immune system by the adjuvant. Additional benefits of the cell transfer included a decrease in the time needed for arthritis development, and uniformity and synchronization of joint inflammation in recipient mice [32].
Two major criteria were used to select genes that might be important for arthritis development: (1) significant differences in expression levels between experimental groups and (2) the fold change in expression levels. When only the first criterion was applied, genome-wide analysis identified a large number of genes whose expression was significantly (P < 0.05) different between any pair of the experimental conditions compared. Irrespective of the statistics used (either unpaired Student's t-test, the Fisher exact test or the Mann–Whitney U-test), the number of differentially expressed genes was found to represent about 5–10% of the entire mouse genome. We further 'filtered' these genes by using a cut-off threshold set at twofold change of expression, because this threshold could reflect a physiologically important change in gene activity, and a twofold change exceeded the average CV for all pairwise comparisons. Decreasing the number of 'false positive' genes by application of these two filtering procedures proved to be an effective technique for the identification of genes that are likely to be involved in arthritis development.
The present study indicates that the number of differentially expressed genes increases with the progression of the disease. At the initiation phase, when no clinical symptoms of inflammation were yet detected, only 37 genes were upregulated or downregulated. However, a differential expression of 277 genes was observed at the acute phase, and chronic inflammation was characterized by the differential activity of 418 genes. Interestingly, most early arthritis signature genes (27 of 37) remained upregulated or downregulated in inflamed joints (Fig. 2). A different set of genes was also involved in acute inflammation. At the chronic phase, less than half of AA-specific genes (127 of 277) were differentially expressed, and another half was CA-specific. A very limited number of transcripts (n = 15) remained upregulated or downregulated in all three phases of arthritis.
Activated T cells must be present in the peripheral blood of recipient BALB/cSCID mice after the transfer, but donor lymphocytes can be detected in joints as early as 3–5 days after the second transfer [32]. In earlier studies [31], and in control experiments (data not shown), using fluorescein-labeled or isotope-labeled donor lymphocytes, only very few cells were found in joints during the first week of transfer, and a second cell transfer was needed to induce a significant influx of lymphocytes into the joints and cause subsequent inflammation. In this study, we detected overexpression of a T cell-specific GTPase (Tgtp) and T cell receptor β (Tcrbv13) in still non-inflamed (pre-arthritic) paws of recipient BALB/cSCID mice as early as 3–5 days after the second injection, indicating the presence of donor BALB/c lymphocytes. Thus, the initiation and development of arthritis in adoptively transferred PGIA must depend on cooperation between adaptive immunity cells (represented by donor BALB/c lymphocytes) and cells of innate immunity (represented by non-lymphoid cells in the recipient BALB/cSCID mice). Analysis of the cellular and tissue specificity of gene expression, using public gene expression databases [5456], indicated that genes encoding CD48 (Cd48), membrane-spanning 4A6B and 4A6C (Ms4a6b and Ms4a6c), epidermal growth factor-like receptor-like protein 1 (Emr1), and interferon-induced 47 kDa protein (Ifi47) were most probably originating from donor lymphoid cells, whereas other early arthritis genes (Table 2) were related to the activation of the innate immune system (represented by macrophages, dendritic cells, and cells of myeloid lineage) of recipient BALB/cSCID mice.
Transcriptional control of gene activity is only one component of the complex cellular regulatory pathways. In other words, the functional activity of a protein depends on several factors such as interaction with other proteins, phosphorylation/dephosphorylation, subcellular compartmentalization, and other post-translational modifications. All of these factors might be involved in the regulation of interactions between the donor lymphocytes and the synovial/joint cells of recipient mice that lack an adaptive immune system. The list of genes we present in this study is rather short; that is, it includes only genes profoundly affected during arthritis initiation and progression at the level of transcription. Genes and proteins that are under subtle regulatory pressure, or are controlled by non-genetic mechanisms such as protein phosphorylation and other post-translational events, could not be detected and analyzed in this study. The development of new proteomics assays, and the synthesis of existing knowledge in cellular signaling pathways with information provided by gene expression studies, will be necessary to build up a complete arthritis-related regulatory network and to unravel the mechanisms involved in the development and progression of autoimmune arthritis.

Conclusions

The development and progression of a complex polygenic autoimmune disease such as RA are controlled by hundreds or thousands of genes, in addition to the MHC. Despite the relatively high incidence of RA in the human population, only a few studies have applied gene array methods to the monitoring of disease progression and efficacy of treatment, or to predicting the prognosis of the disease. The major obstacles in the human studies are the relatively late diagnosis of RA, the large variety of cell types (cells of the immune system and of synovial joints) involved in autoimmune arthritic processes, and the extreme genetic heterogeneity of the human population. The present study applied an adoptively transferred murine model of RA and a microarray approach to detect differentially expressed, disease-related signature genes in PA (still non-inflamed) joints, days before the clinical symptoms or histopathological abnormalities of joint inflammation could be observed.
However, the detection of early arthritis signature genes in joints can be done only in an experimental system in which particular joints have already been affected before the inflammatory symptoms can be identified. To make this experimental system uniform, that is, to exclude individual variations, we adoptively transferred antigen (PG)-specific lymphocytes (representing cells of adaptive immunity) from primarily arthritic mice into syngeneic SCID mice, which lack an adaptive immune system. In this highly synchronized and uniform system we were able to detect differentially expressed genes in still non-inflamed paws of arthritis-'prone' animals. We identified a relatively small number of mostly upregulated early arthritis signature genes (known to be involved in arthritic processes and/or autoimmunity), some of which were expressed at even higher levels in the acute phase of arthritis. These early arthritis signature genes, originating from donor cells, indicated the involvement of adaptive immunity, whereas the innate immunity genes were differentially expressed by cells of the recipients.
The early signature genes, together with those that were differentially expressed in the acute (277 genes) and chronic (418 genes) phase of arthritis, are listed in the Additional files. Although many of these differentially expressed genes, detected either in the acute phase or during the progression of the disease, have been implicated in inflammation or autoimmunity, the list contains a significant number of differentially expressed genes whose function, or association with arthritis, is unknown at present.

Acknowledgements

We thank Dr Kira Adaricheva for algorithmic and mathematical support during data analysis. We are indebted to David George and Yonghong Zhang for Affymetrix fluidic station operation and data management. This research was supported in part by grants AR40310, AR45652, and AR51163 from the National Institutes of Health, and the JO Galante MD DSc endowment fund.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

VAA performed essentially all statistical analyses and put together the draft version of the results and figures. CV isolated all RNA samples, prepared biotinylated samples and was involved in Affymetrix hybridization experiments; he also performed preliminary clustering experiments with GeneSpring version 6.2 (not included in this paper). AH performed all in vitro stimulation and adoptive transfer experiments, and assessed arthritis three or four times a day together with KM, who was also involved in all phases of the experimental processes and in the finalization of the manuscript. EGB controlled Affymetrix hybridization and scanning experiments, managed preliminary data analysis and finalized the manuscript. TTG designed experiments, controlled all experimental steps, data analysis, and finalized the manuscript. All authors read and approved the final manuscript.
Anhänge
Literatur
1.
Zurück zum Zitat Barnes MG, Aronow BJ, Luyrink LK, Moroldo MB, Pavlidis P, Passo MH, Grom AA, Hirsch R, Giannini EH, Colbert RA, et al: Gene expression in juvenile arthritis and spondyloarthropathy: pro-angiogenic ELR+ chemokine genes relate to course of arthritis. Rheumatology (Oxford). 2004, 43: 973-979.CrossRef Barnes MG, Aronow BJ, Luyrink LK, Moroldo MB, Pavlidis P, Passo MH, Grom AA, Hirsch R, Giannini EH, Colbert RA, et al: Gene expression in juvenile arthritis and spondyloarthropathy: pro-angiogenic ELR+ chemokine genes relate to course of arthritis. Rheumatology (Oxford). 2004, 43: 973-979.CrossRef
2.
Zurück zum Zitat Heller RA, Schena M, Chai A, Shalon D, Bedilion T, Gilmore J, Woolley DE, Davis RW: Discovery and analysis of inflammatory disease-related genes using cDNA microarrays. Proc Natl Acad Sci USA. 1997, 94: 2150-2155. 10.1073/pnas.94.6.2150.PubMedCentralCrossRefPubMed Heller RA, Schena M, Chai A, Shalon D, Bedilion T, Gilmore J, Woolley DE, Davis RW: Discovery and analysis of inflammatory disease-related genes using cDNA microarrays. Proc Natl Acad Sci USA. 1997, 94: 2150-2155. 10.1073/pnas.94.6.2150.PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Zanders ED, Goulden MG, Kennedy TC, Kempsell KE: Analysis of immune system gene expression in small rheumatoid arthritis biopsies using a combination of subtractive hybridization and high-density cDNA arrays. J Immunol Methods. 2000, 233: 131-140. 10.1016/S0022-1759(99)00126-X.CrossRefPubMed Zanders ED, Goulden MG, Kennedy TC, Kempsell KE: Analysis of immune system gene expression in small rheumatoid arthritis biopsies using a combination of subtractive hybridization and high-density cDNA arrays. J Immunol Methods. 2000, 233: 131-140. 10.1016/S0022-1759(99)00126-X.CrossRefPubMed
4.
Zurück zum Zitat Watanabe N, Ando K, Yoshida S, Inuzuka S, Kobayashi M, Matsui N, Okamoto T: Gene expression profile analysis of rheumatoid synovial fibroblast cultures revealing the overexpression of genes responsible for tumor- like growth of rheumatoid synovium. Biochem Biophys Res Commun. 2002, 294: 1121-1129. 10.1016/S0006-291X(02)00608-3.CrossRefPubMed Watanabe N, Ando K, Yoshida S, Inuzuka S, Kobayashi M, Matsui N, Okamoto T: Gene expression profile analysis of rheumatoid synovial fibroblast cultures revealing the overexpression of genes responsible for tumor- like growth of rheumatoid synovium. Biochem Biophys Res Commun. 2002, 294: 1121-1129. 10.1016/S0006-291X(02)00608-3.CrossRefPubMed
5.
Zurück zum Zitat Grant EP, Pickard MD, Briskin MJ, Gutierrez-Ramos JC: Gene expression profiles: creating new perspectives in arthritis research. Arthritis Rheum. 2002, 46: 874-884. 10.1002/art.10014.CrossRefPubMed Grant EP, Pickard MD, Briskin MJ, Gutierrez-Ramos JC: Gene expression profiles: creating new perspectives in arthritis research. Arthritis Rheum. 2002, 46: 874-884. 10.1002/art.10014.CrossRefPubMed
6.
Zurück zum Zitat Thornton S, Sowders D, Aronow B, Witte DP, Brunner HI, Giannini EH, Hirsch R: DNA microarray analysis reveals novel gene expression profiles in collagen-induced arthritis. Clin Immunol. 2002, 105: 155-168. 10.1006/clim.2002.5227.CrossRefPubMed Thornton S, Sowders D, Aronow B, Witte DP, Brunner HI, Giannini EH, Hirsch R: DNA microarray analysis reveals novel gene expression profiles in collagen-induced arthritis. Clin Immunol. 2002, 105: 155-168. 10.1006/clim.2002.5227.CrossRefPubMed
7.
Zurück zum Zitat Maas K, Chan S, Parker J, Slater A, Moore J, Olsen N, Aune TM: Cutting edge: molecular portrait of human autoimmune disease. J Immunol. 2002, 169: 5-9.CrossRefPubMed Maas K, Chan S, Parker J, Slater A, Moore J, Olsen N, Aune TM: Cutting edge: molecular portrait of human autoimmune disease. J Immunol. 2002, 169: 5-9.CrossRefPubMed
8.
Zurück zum Zitat Sweeney SE, Firestein GS: Signal transduction in rheumatoid arthritis. Curr Opin Rheumatol. 2004, 16: 231-237. 10.1097/00002281-200405000-00011.CrossRefPubMed Sweeney SE, Firestein GS: Signal transduction in rheumatoid arthritis. Curr Opin Rheumatol. 2004, 16: 231-237. 10.1097/00002281-200405000-00011.CrossRefPubMed
9.
Zurück zum Zitat Justen HP, Grunewald E, Totzke G, Gouni-Berthold I, Sachinidis A, Wessinghage D, Vetter H, Schulze-Osthoff K, Ko Y: Differential gene expression in synovium of rheumatoid arthritis and osteoarthritis. Mol Cell Biol Res Commun. 2000, 3: 165-172. 10.1006/mcbr.2000.0211.CrossRefPubMed Justen HP, Grunewald E, Totzke G, Gouni-Berthold I, Sachinidis A, Wessinghage D, Vetter H, Schulze-Osthoff K, Ko Y: Differential gene expression in synovium of rheumatoid arthritis and osteoarthritis. Mol Cell Biol Res Commun. 2000, 3: 165-172. 10.1006/mcbr.2000.0211.CrossRefPubMed
10.
Zurück zum Zitat van der Pouw Kraan TC, van Gaalen FA, Huizinga TW, Pieterman E, Breedveld FC, Verweij CL: Discovery of distinctive gene expression profiles in rheumatoid synovium using cDNA microarray technology: evidence for the existence of multiple pathways of tissue destruction and repair. Genes Immun. 2003, 4: 187-196. 10.1038/sj.gene.6363975.CrossRefPubMed van der Pouw Kraan TC, van Gaalen FA, Huizinga TW, Pieterman E, Breedveld FC, Verweij CL: Discovery of distinctive gene expression profiles in rheumatoid synovium using cDNA microarray technology: evidence for the existence of multiple pathways of tissue destruction and repair. Genes Immun. 2003, 4: 187-196. 10.1038/sj.gene.6363975.CrossRefPubMed
11.
Zurück zum Zitat Jarvis JN, Dozmorov I, Jiang K, Frank MB, Szodoray P, Alex P, Centola M: Novel approaches to gene expression analysis of active polyarticular juvenile rheumatoid arthritis. Arthritis Res Ther. 2004, 6: R15-R32. 10.1186/ar1018.PubMedCentralCrossRefPubMed Jarvis JN, Dozmorov I, Jiang K, Frank MB, Szodoray P, Alex P, Centola M: Novel approaches to gene expression analysis of active polyarticular juvenile rheumatoid arthritis. Arthritis Res Ther. 2004, 6: R15-R32. 10.1186/ar1018.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Trentham DE, Townes AS, Kang AH: Autoimmunity to type II collagen: an experimental model of arthritis. J Exp Med. 1977, 146: 857-868. 10.1084/jem.146.3.857.CrossRefPubMed Trentham DE, Townes AS, Kang AH: Autoimmunity to type II collagen: an experimental model of arthritis. J Exp Med. 1977, 146: 857-868. 10.1084/jem.146.3.857.CrossRefPubMed
13.
Zurück zum Zitat Glant TT, Mikecz K, Arzoumanian A, Poole AR: Proteoglycan-induced arthritis in BALB/c mice. Clinical features and histopathology. Arthritis Rheum. 1987, 30: 201-212.CrossRefPubMed Glant TT, Mikecz K, Arzoumanian A, Poole AR: Proteoglycan-induced arthritis in BALB/c mice. Clinical features and histopathology. Arthritis Rheum. 1987, 30: 201-212.CrossRefPubMed
14.
Zurück zum Zitat Zhang Y, Guerassimov A, Leroux J-Y, Cartman A, Webber C, Lalic R, de Miguel E, Rosenberg LC, Poole AR: Induction of arthritis in BALB/c mice by cartilage link protein. Involvement of distinct regions recognized by T- and B lymphocytes. Am J Pathol. 1998, 153: 1283-1291.PubMedCentralCrossRefPubMed Zhang Y, Guerassimov A, Leroux J-Y, Cartman A, Webber C, Lalic R, de Miguel E, Rosenberg LC, Poole AR: Induction of arthritis in BALB/c mice by cartilage link protein. Involvement of distinct regions recognized by T- and B lymphocytes. Am J Pathol. 1998, 153: 1283-1291.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Verheijden GFM, Rijnders AWM, Bos E, De Roo CJJC, van Staveren CJ, Miltenburg AMM, Meijerink JH, Elewaut D, de Keyser F, Veys E, et al: Human cartilage glycoprotein-39 as a candidate autoantigen in rheumatoid arthritis. Arthritis Rheum. 1997, 40: 1115-1125.CrossRefPubMed Verheijden GFM, Rijnders AWM, Bos E, De Roo CJJC, van Staveren CJ, Miltenburg AMM, Meijerink JH, Elewaut D, de Keyser F, Veys E, et al: Human cartilage glycoprotein-39 as a candidate autoantigen in rheumatoid arthritis. Arthritis Rheum. 1997, 40: 1115-1125.CrossRefPubMed
16.
Zurück zum Zitat Vingsbo-Lundberg C, Nordquist N, Olofsson P, Sundvall M, Saxne T, Pettersson U, Holmdahl R: Genetic control of arthritis onset, severity and chronicity in a model for rheumatoid arthritis in rats. Nat Genet. 1998, 20: 401-404. 10.1038/3887.CrossRefPubMed Vingsbo-Lundberg C, Nordquist N, Olofsson P, Sundvall M, Saxne T, Pettersson U, Holmdahl R: Genetic control of arthritis onset, severity and chronicity in a model for rheumatoid arthritis in rats. Nat Genet. 1998, 20: 401-404. 10.1038/3887.CrossRefPubMed
17.
Zurück zum Zitat Remmers EF, Longman RE, Du Y, O'Hare A, Cannon GW, Griffiths MM, Wilder RL: A genome scan localizes five non-MHC loci controlling collagen-induced arthritis in rats. Nat Genet. 1996, 14: 82-85. 10.1038/ng0996-82.CrossRefPubMed Remmers EF, Longman RE, Du Y, O'Hare A, Cannon GW, Griffiths MM, Wilder RL: A genome scan localizes five non-MHC loci controlling collagen-induced arthritis in rats. Nat Genet. 1996, 14: 82-85. 10.1038/ng0996-82.CrossRefPubMed
18.
Zurück zum Zitat Jirholt J, Cook A, Emahazion T, Sundvall M, Jansson L, Nordquist N, Pettersson U, Holmdahl R: Genetic linkage analysis of collagen-induced arthritis in the mouse. Eur J Immunol. 1998, 28: 3321-3328. 10.1002/(SICI)1521-4141(199810)28:10<3321::AID-IMMU3321>3.0.CO;2-M.CrossRefPubMed Jirholt J, Cook A, Emahazion T, Sundvall M, Jansson L, Nordquist N, Pettersson U, Holmdahl R: Genetic linkage analysis of collagen-induced arthritis in the mouse. Eur J Immunol. 1998, 28: 3321-3328. 10.1002/(SICI)1521-4141(199810)28:10<3321::AID-IMMU3321>3.0.CO;2-M.CrossRefPubMed
19.
Zurück zum Zitat Mikecz K, Glant TT, Poole AR: Immunity to cartilage proteoglycans in BALB/c mice with progressive polyarthritis and ankylosing spondylitis induced by injection of human cartilage proteoglycan. Arthritis Rheum. 1987, 30: 306-318.CrossRefPubMed Mikecz K, Glant TT, Poole AR: Immunity to cartilage proteoglycans in BALB/c mice with progressive polyarthritis and ankylosing spondylitis induced by injection of human cartilage proteoglycan. Arthritis Rheum. 1987, 30: 306-318.CrossRefPubMed
20.
Zurück zum Zitat Adarichev VA, Bárdos T, Christodoulou S, Phillips MT, Mikecz K, Glant TT: Major histocompatibility complex controls susceptibility and dominant inheritance, but not the severity of the disease in mouse models of rheumatoid arthritis. Immunogenetics. 2002, 54: 184-192. 10.1007/s00251-002-0462-8.CrossRefPubMed Adarichev VA, Bárdos T, Christodoulou S, Phillips MT, Mikecz K, Glant TT: Major histocompatibility complex controls susceptibility and dominant inheritance, but not the severity of the disease in mouse models of rheumatoid arthritis. Immunogenetics. 2002, 54: 184-192. 10.1007/s00251-002-0462-8.CrossRefPubMed
21.
Zurück zum Zitat Ibrahim SM, Koczan D, Thiesen HJ: Gene-expression profile of collagen-induced arthritis. J Autoimmun. 2002, 18: 159-167. 10.1006/jaut.2001.0580.CrossRefPubMed Ibrahim SM, Koczan D, Thiesen HJ: Gene-expression profile of collagen-induced arthritis. J Autoimmun. 2002, 18: 159-167. 10.1006/jaut.2001.0580.CrossRefPubMed
22.
Zurück zum Zitat Firneisz G, Zahevi I, Vermes C, Hanyecz A, Frieman JA, Glant TT: Identification and quantification of disease-related gene clusters. Bioinformatics. 2003, 19: 1781-1786. 10.1093/bioinformatics/btg252.CrossRefPubMed Firneisz G, Zahevi I, Vermes C, Hanyecz A, Frieman JA, Glant TT: Identification and quantification of disease-related gene clusters. Bioinformatics. 2003, 19: 1781-1786. 10.1093/bioinformatics/btg252.CrossRefPubMed
23.
Zurück zum Zitat Wester L, Koczan D, Holmberg J, Olofsson P, Thiesen HJ, Holmdahl R, Ibrahim S: Differential gene expression in pristane-induced arthritis susceptible DA versus resistant E3 rats. Arthritis Res Ther. 2003, 5: R361-R372. 10.1186/ar993.PubMedCentralCrossRefPubMed Wester L, Koczan D, Holmberg J, Olofsson P, Thiesen HJ, Holmdahl R, Ibrahim S: Differential gene expression in pristane-induced arthritis susceptible DA versus resistant E3 rats. Arthritis Res Ther. 2003, 5: R361-R372. 10.1186/ar993.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Glant TT, Cs-Szabó G, Nagase H, Jacobs JJ, Mikecz K: Progressive polyarthritis induced in BALB/c mice by aggrecan from human osteoarthritic cartilage. Arthritis Rheum. 1998, 41: 1007-1018. 10.1002/1529-0131(199806)41:6<1007::AID-ART7>3.0.CO;2-6.CrossRefPubMed Glant TT, Cs-Szabó G, Nagase H, Jacobs JJ, Mikecz K: Progressive polyarthritis induced in BALB/c mice by aggrecan from human osteoarthritic cartilage. Arthritis Rheum. 1998, 41: 1007-1018. 10.1002/1529-0131(199806)41:6<1007::AID-ART7>3.0.CO;2-6.CrossRefPubMed
25.
Zurück zum Zitat Glant TT, Finnegan A, Mikecz K: Proteoglycan-induced arthritis: immune regulation, cellular mechanisms and genetics. Crit Rev Immunol. 2003, 23: 199-250. 10.1615/CritRevImmunol.v23.i3.20.CrossRefPubMed Glant TT, Finnegan A, Mikecz K: Proteoglycan-induced arthritis: immune regulation, cellular mechanisms and genetics. Crit Rev Immunol. 2003, 23: 199-250. 10.1615/CritRevImmunol.v23.i3.20.CrossRefPubMed
26.
Zurück zum Zitat Otto JM, Chandrasekaran R, Vermes C, Mikecz K, Finnegan A, Rickert SE, Enders JT, Glant TT: A genome scan using a novel genetic cross identifies new susceptibility loci and traits in a mouse model of rheumatoid arthritis. J Immunol. 2000, 165: 5278-5286.CrossRefPubMed Otto JM, Chandrasekaran R, Vermes C, Mikecz K, Finnegan A, Rickert SE, Enders JT, Glant TT: A genome scan using a novel genetic cross identifies new susceptibility loci and traits in a mouse model of rheumatoid arthritis. J Immunol. 2000, 165: 5278-5286.CrossRefPubMed
27.
Zurück zum Zitat Adarichev VA, Valdez JC, Bárdos T, Finnegan A, Mikecz K, Glant TT: Combined autoimmune models of arthritis reveal shared and independent qualitative (binary) and quantitative trait loci. J Immunol. 2003, 170: 2283-2292.CrossRefPubMed Adarichev VA, Valdez JC, Bárdos T, Finnegan A, Mikecz K, Glant TT: Combined autoimmune models of arthritis reveal shared and independent qualitative (binary) and quantitative trait loci. J Immunol. 2003, 170: 2283-2292.CrossRefPubMed
28.
Zurück zum Zitat Otto JM, Cs-Szabó G, Gallagher J, Velins S, Mikecz K, Buzás EI, Enders JT, Li Y, Olsen BR, Glant TT: Identification of multiple loci linked to inflammation and autoantibody production by a genome scan of a murine model of rheumatoid arthritis. Arthritis Rheum. 1999, 42: 2524-2531. 10.1002/1529-0131(199912)42:12<2524::AID-ANR4>3.0.CO;2-0.CrossRefPubMed Otto JM, Cs-Szabó G, Gallagher J, Velins S, Mikecz K, Buzás EI, Enders JT, Li Y, Olsen BR, Glant TT: Identification of multiple loci linked to inflammation and autoantibody production by a genome scan of a murine model of rheumatoid arthritis. Arthritis Rheum. 1999, 42: 2524-2531. 10.1002/1529-0131(199912)42:12<2524::AID-ANR4>3.0.CO;2-0.CrossRefPubMed
29.
Zurück zum Zitat Adarichev VA, Nesterovitch AB, Bárdos T, Biesczat D, Chandrasekaran R, Vermes C, Mikecz K, Finnegan A, Glant TT: Sex effect on clinical and immunological quantitative trait loci in a murine model of rheumatoid arthritis. Arthritis Rheum. 2003, 48: 1708-1720. 10.1002/art.11016.CrossRefPubMed Adarichev VA, Nesterovitch AB, Bárdos T, Biesczat D, Chandrasekaran R, Vermes C, Mikecz K, Finnegan A, Glant TT: Sex effect on clinical and immunological quantitative trait loci in a murine model of rheumatoid arthritis. Arthritis Rheum. 2003, 48: 1708-1720. 10.1002/art.11016.CrossRefPubMed
30.
Zurück zum Zitat Mikecz K, Glant TT, Buzás E, Poole AR: Proteoglycan-induced polyarthritis and spondylitis adoptively transferred to naive (nonimmunized) BALB/c mice. Arthritis Rheum. 1990, 33: 866-876.CrossRefPubMed Mikecz K, Glant TT, Buzás E, Poole AR: Proteoglycan-induced polyarthritis and spondylitis adoptively transferred to naive (nonimmunized) BALB/c mice. Arthritis Rheum. 1990, 33: 866-876.CrossRefPubMed
31.
Zurück zum Zitat Mikecz K, Glant TT: Migration and homing of lymphocytes to lymphoid and synovial tissues in proteoglycan-induced murine arthritis. Arthritis Rheum. 1994, 37: 1395-1403.CrossRefPubMed Mikecz K, Glant TT: Migration and homing of lymphocytes to lymphoid and synovial tissues in proteoglycan-induced murine arthritis. Arthritis Rheum. 1994, 37: 1395-1403.CrossRefPubMed
32.
Zurück zum Zitat Bárdos T, Mikecz K, Finnegan A, Zhang J, Glant TT: T and B cell recovery in arthritis adoptively transferred to SCID mice: antigen-specific activation is required for restoration of autopathogenic CD4+ Th1 cells in a syngeneic system. J Immunol. 2002, 168: 6013-6021.CrossRefPubMed Bárdos T, Mikecz K, Finnegan A, Zhang J, Glant TT: T and B cell recovery in arthritis adoptively transferred to SCID mice: antigen-specific activation is required for restoration of autopathogenic CD4+ Th1 cells in a syngeneic system. J Immunol. 2002, 168: 6013-6021.CrossRefPubMed
33.
Zurück zum Zitat Schaible UE, Kramer MD, Museteanu C, Zimmer G, Mossmann H, Simon MM: The severe combined immunodeficiency (scid) mouse. A laboratory model for the analysis of lyme arthritis and carditis. J Exp Med. 1989, 170: 1427-1432. 10.1084/jem.170.4.1427.CrossRefPubMed Schaible UE, Kramer MD, Museteanu C, Zimmer G, Mossmann H, Simon MM: The severe combined immunodeficiency (scid) mouse. A laboratory model for the analysis of lyme arthritis and carditis. J Exp Med. 1989, 170: 1427-1432. 10.1084/jem.170.4.1427.CrossRefPubMed
34.
Zurück zum Zitat Carlow DA, Marth J, Clark-Lewis I, Teh HS: Isolation of a gene encoding a developmentally regulated T cell-specific protein with a guanine nucleotide triphosphate-binding motif. J Immunol. 1995, 154: 1724-1734.PubMed Carlow DA, Marth J, Clark-Lewis I, Teh HS: Isolation of a gene encoding a developmentally regulated T cell-specific protein with a guanine nucleotide triphosphate-binding motif. J Immunol. 1995, 154: 1724-1734.PubMed
35.
Zurück zum Zitat Hanyecz A, Berlo SE, Szanto S, Broeren CPM, Mikecz K, Glant TT: Achievement of a synergistic adjuvant effect on arthritis induction by activation of innate immunity and forcing the immune response toward the Th1 phenotype. Arthritis Rheum. 2004, 50: 1665-1676. 10.1002/art.20180.CrossRefPubMed Hanyecz A, Berlo SE, Szanto S, Broeren CPM, Mikecz K, Glant TT: Achievement of a synergistic adjuvant effect on arthritis induction by activation of innate immunity and forcing the immune response toward the Th1 phenotype. Arthritis Rheum. 2004, 50: 1665-1676. 10.1002/art.20180.CrossRefPubMed
36.
Zurück zum Zitat Glant TT, Mikecz K: Proteoglycan aggrecan-induced arthritis. A murine autoimmune model of rheumatoid arthritis. Autoimmunity Methods and Protocols. Edited by: Perl A. 2004, Totowa, NJ: Humana Press, 313-338. Glant TT, Mikecz K: Proteoglycan aggrecan-induced arthritis. A murine autoimmune model of rheumatoid arthritis. Autoimmunity Methods and Protocols. Edited by: Perl A. 2004, Totowa, NJ: Humana Press, 313-338.
37.
Zurück zum Zitat Mikecz K, Brennan FR, Kim JH, Glant TT: Anti-CD44 treatment abrogates tissue edema and leukocyte infiltration in murine arthritis. Nat Med. 1995, 1: 558-563. 10.1038/nm0695-558.CrossRefPubMed Mikecz K, Brennan FR, Kim JH, Glant TT: Anti-CD44 treatment abrogates tissue edema and leukocyte infiltration in murine arthritis. Nat Med. 1995, 1: 558-563. 10.1038/nm0695-558.CrossRefPubMed
38.
Zurück zum Zitat Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning: a Laboratory Manual. 1989, New York: Cold Spring Harbor Laboratory Press Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning: a Laboratory Manual. 1989, New York: Cold Spring Harbor Laboratory Press
39.
Zurück zum Zitat Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, An P, et al: Initial sequencing and comparative analysis of the mouse genome. Nature. 2002, 420: 520-562. 10.1038/nature01262.CrossRefPubMed Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, An P, et al: Initial sequencing and comparative analysis of the mouse genome. Nature. 2002, 420: 520-562. 10.1038/nature01262.CrossRefPubMed
40.
Zurück zum Zitat Hubbard T, Barker D, Birney E, Cameron G, Chen Y, Clark L, Cox T, Cuff J, Curwen V, Down T, et al: The Ensembl genome database project. Nucleic Acids Res. 2002, 30: 38-41. 10.1093/nar/30.1.38.PubMedCentralCrossRefPubMed Hubbard T, Barker D, Birney E, Cameron G, Chen Y, Clark L, Cox T, Cuff J, Curwen V, Down T, et al: The Ensembl genome database project. Nucleic Acids Res. 2002, 30: 38-41. 10.1093/nar/30.1.38.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Blake JA, Eppig JT, Richardson JE, Davisson MT, Mouse Genome Informatics Group: The mouse genome database (MGD): a community resource. Status and enhancements. Nucleic Acids Res. 1998, 26: 130-137. 10.1093/nar/26.1.130.PubMedCentralCrossRefPubMed Blake JA, Eppig JT, Richardson JE, Davisson MT, Mouse Genome Informatics Group: The mouse genome database (MGD): a community resource. Status and enhancements. Nucleic Acids Res. 1998, 26: 130-137. 10.1093/nar/26.1.130.PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Mayanil CS, George D, Freilich L, Miljan EJ, Mania-Farnell B, McLone DG, Bremer EG: Microarray analysis detects novel Pax3 downstream target genes. J Biol Chem. 2001, 276: 49299-49309. 10.1074/jbc.M107933200.CrossRefPubMed Mayanil CS, George D, Freilich L, Miljan EJ, Mania-Farnell B, McLone DG, Bremer EG: Microarray analysis detects novel Pax3 downstream target genes. J Biol Chem. 2001, 276: 49299-49309. 10.1074/jbc.M107933200.CrossRefPubMed
44.
Zurück zum Zitat Underhill GH, George D, Bremer EG, Kansas GS: Gene expression profiling reveals a highly specialized genetic program of plasma cells. Blood. 2003, 101: 4013-4021. 10.1182/blood-2002-08-2673.CrossRefPubMed Underhill GH, George D, Bremer EG, Kansas GS: Gene expression profiling reveals a highly specialized genetic program of plasma cells. Blood. 2003, 101: 4013-4021. 10.1182/blood-2002-08-2673.CrossRefPubMed
45.
Zurück zum Zitat Li C, Wong WH: Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc Natl Acad Sci USA. 2001, 98: 31-36. 10.1073/pnas.011404098.PubMedCentralCrossRefPubMed Li C, Wong WH: Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc Natl Acad Sci USA. 2001, 98: 31-36. 10.1073/pnas.011404098.PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Eisen MB, Spellman PT, Brown PO, Botstein D: Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA. 1998, 95: 14863-14868. 10.1073/pnas.95.25.14863.PubMedCentralCrossRefPubMed Eisen MB, Spellman PT, Brown PO, Botstein D: Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA. 1998, 95: 14863-14868. 10.1073/pnas.95.25.14863.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000, 25: 25-29. 10.1038/75556.PubMedCentralCrossRefPubMed Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000, 25: 25-29. 10.1038/75556.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Shoukri MM, Edge VL: Statistical Methods for Health Sciences. 1996, Boca Raton: CRC Press Shoukri MM, Edge VL: Statistical Methods for Health Sciences. 1996, Boca Raton: CRC Press
50.
Zurück zum Zitat Feinstein AR: Principles of Medical Statistics. 2002, Boca Raton: Chapman & Hall/CRC Feinstein AR: Principles of Medical Statistics. 2002, Boca Raton: Chapman & Hall/CRC
51.
Zurück zum Zitat Glant TT, Kamath RV, Bárdos T, Gál I, Szanto S, Murad YM, Sandy JD, Mort JS, Roughley PJ, Mikecz K: Cartilage-specific constitutive expression of TSG-6 protein (product of tumor necrosis factor α-stimulated gene 6) provides a chondroprotective, but not anti-inflammatory, effect in antigen-induced arthritis. Arthritis Rheum. 2002, 46: 2207-2218. 10.1002/art.10555.CrossRefPubMed Glant TT, Kamath RV, Bárdos T, Gál I, Szanto S, Murad YM, Sandy JD, Mort JS, Roughley PJ, Mikecz K: Cartilage-specific constitutive expression of TSG-6 protein (product of tumor necrosis factor α-stimulated gene 6) provides a chondroprotective, but not anti-inflammatory, effect in antigen-induced arthritis. Arthritis Rheum. 2002, 46: 2207-2218. 10.1002/art.10555.CrossRefPubMed
52.
Zurück zum Zitat Suetsugu S, Takenawa T: Regulation of cortical actin networks in cell migration. Int Rev Cytol. 2003, 229: 245-286.CrossRefPubMed Suetsugu S, Takenawa T: Regulation of cortical actin networks in cell migration. Int Rev Cytol. 2003, 229: 245-286.CrossRefPubMed
53.
Zurück zum Zitat Arthur WT, Noren NK, Burridge K: Regulation of Rho family GTPases by cell–cell and cell–matrix adhesion. Biol Res. 2002, 35: 239-246.CrossRefPubMed Arthur WT, Noren NK, Burridge K: Regulation of Rho family GTPases by cell–cell and cell–matrix adhesion. Biol Res. 2002, 35: 239-246.CrossRefPubMed
Metadaten
Titel
Gene expression profiling in murine autoimmune arthritis during the initiation and progression of joint inflammation
verfasst von
Vyacheslav A Adarichev
Csaba Vermes
Anita Hanyecz
Katalin Mikecz
Eric G Bremer
Tibor T Glant
Publikationsdatum
01.12.2004
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 2/2005
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/ar1472

Weitere Artikel der Ausgabe 2/2005

Arthritis Research & Therapy 2/2005 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

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