Abstract
Background and purpose:
A main challenge in the therapy of osteoarthritis (OA) is the development of drugs that will modify the disease. Reliable test systems are necessary to enable an efficient screening of therapeutic substances. We therefore established a chondrocyte-based in vitro cell culture model in order to characterize different p38MAPK inhibitors.
Experimental approach:
Interleukin-1β (IL-1β)-stimulated human OA chondrocytes were treated with the p38MAPK inhibitors Birb 796, pamapimod, SB203580 and the new substance CBS-3868. Birb 796- and SB203580-treated cells were analysed in a genome-wide microarray analysis. The efficacy of all inhibitors was characterized by quantitative gene expression analysis and the quantification of PGE2 and NO release.
Key results:
Microarray analysis revealed inhibitor-specific differences in gene expression. Whereas SB203580 had a broad effect on chondrocytes, Birb 796 counteracted the IL-1β effect more specifically. All p38MAPK inhibitors significantly inhibited the IL-1β-induced gene expression of COX-2, mPGES1, iNOS, matrix metalloproteinase 13 (MMP13) and TNFRSF11B, as well as PGE2 release. Birb 796 and CBS-3868 showed a higher efficacy than SB203580 and pamapimod at inhibiting the expression of COX-2 and MMP13 genes, as well as PGE2 release. In the case of mPGES1 and TNFRSF11B gene expression, CBS-3868 exceeded the efficacy of Birb 796.
Conclusions and implications:
Our test system could differentially characterize inhibitors of the same primary pharmaceutical target. It reflects processes relevant in OA and is based on chondrocytes that are mainly responsible for cartilage degradation. It therefore represents a valuable tool for drug screening in between functional in vitro testing and in vivo models.
Keywords: chondrocytes, interleukin 1β, in vitro model, osteoarthritis, p38MAPK inhibition, whole-genome array, Birb 796, pamapimod, SB203580
Introduction
The central role of p38MAP kinases (p38MAPK), foremost the α-isoform, in the production of inflammatory response proteins such as TNF-α, interleukin-1β (IL-1β), COX-2 and microsomal prostaglandin E synthase (mPGES1) is well documented (Masuko-Hongo et al., 2004; Schieven, 2005). Activated p38α MAPK up-regulates cytokine production by several independent mechanisms, including direct phosphorylation of transcription factors, and direct or indirect stabilization and increased translation of mRNAs by phosphorylation of adenylate/uridylate-rich element-binding proteins (Ashwell, 2006). Since its identification as a protein that binds cytokine-suppressing anti-inflammatory molecules (Lee et al., 1994), p38α MAPK has been considered to be an attractive target for drug-mediated modulation of inflammatory processes. Many small molecules have been described in the scientific literature and in patent application, and a few have been clinically developed as a treatment for conditions such as rheumatoid arthritis (RA), Crohn's disease or psoriasis (Dominguez et al., 2005; Genovese, 2009). Most drug discovery programmes have focused on the inhibition of the α-form, but essentially all p38α MAPK inhibitors also interact with the β-isoform. However, recently published results of clinical studies, which investigated the efficacy of pamapimod (Cohen et al., 2009; Alten et al., 2010) and VX-702 (Damjanov et al., 2009) for treatment of RA, were disappointing. During a 12 week treatment of patients with p38α/β MAPK inhibitor either alone or in combination with methotrexate, a significant benefit was not observed. The reasons for this failure of p38α/β MAPK inhibitors in clinical studies are unknown and somehow surprising as they generally show good efficacy in experimental models of arthritis and in clinical pharmacodynamic studies (Sweeney, 2009). Systemically, after intravenous LPS stimulation in healthy subjects, a dose-dependent inhibition of TNF-α release following a single administration of the earlier clinical candidates doramapimod (Birb 796) and RWJ-67657 was observed (Fijen et al., 2001; Branger et al., 2002).
Based on the outcome of the RA proof-of-concept studies, it was hypothesized that biological adaptations allow the re-constitution of the inflammatory process by bypassing the p38α-signalling pathway (Genovese, 2009). Another not-yet-explored explanation relates to different cell- and tissue-specific potencies of drugs. For example, the p38α/β MAPK inhibitors SB239063 and SD-282 (Smith et al., 2006), as well as RWJ-67657 (Westra et al., 2004), exhibited different potencies regarding the inhibition of LPS-induced cytokine release in monocytes and macrophages (Smith et al., 2006). Similar results were obtained when the efficacy of p38α/β MAPK inhibitors was investigated by high-content analysis in SW1353 chondrocytes and baby hamster kidney cells (Ross et al., 2006).
Tissue-specific differences may play an important role in diseases such as RA and osteoarthritis (OA), where articular chondrocytes significantly contribute to the overall pathophysiology. A potent and sustained inhibition of inflammatory processes in this compartment might be pivotal for the efficacy of p38α/β MAPK inhibitors, and therefore, a suitable and reliable in vitro chondrocyte model may deliver important information for defining the molecular properties required of clinical candidates.
The relevance of p38α MAPK signalling in chondrocytes is well documented. Experimental data on the effect of extracellular stimuli such as IL-1β or TNF-α, however, indicate that the other members of the MAP kinase family, the extracellular regulated kinases ERK1/2 and the c-Jun terminal kinases JNK1/2, become activated and contribute to the release of pro-inflammatory mediators (Nieminen et al., 2005). To address the complex interactions in chondrocyte signalling and its assumed relevance for the anti-arthritic efficacy of p38α/β MAPK inhibitors, a global gene expression analysis in primary human chondrocytes after stimulation with IL-1β, in the absence and presence of SB203580 (Joos et al., 2009) or Birb 796, was performed. Many genes that were up-regulated by IL-1β and counter-regulated by the inhibitors were identified (Joos et al., 2009). To characterize the pharmacological profile of different p38α/β inhibitors in IL-1β-stimulated chondrocytes, based on the microarray analysis, a panel of genes was selected and quantitative real-time PCR assays were developed. In the present paper, the effects of different p38α/β inhibitors on the expression of selected genes are presented, and the potential relevance of this model as a screening tool that specifically addresses OA-relevant processes is discussed.
Methods
Cartilage samples
Human osteoarthritic cartilage was obtained from donors undergoing total knee joint replacement due to OA; informed consent was obtained from the patients according to the terms of the Ethics Committee of the University of Ulm. Overall, tissue samples from 30 patients were included in the study; the mean age of the donors was 66 ± 8 years.
Cell culture
Well-preserved cartilage from femoral condyles was used for chondrocyte isolation as described previously (Joos et al., 2008). The cartilage was minced and digested for 45 min with 9 U·mL−1 Pronase (Sigma-Aldrich, Munich, Germany), and for 14 h with 80 U·mL−1 Collagenase type IV (Sigma-Aldrich). After being washed and ltered, the isolated cells were cultivated in complete medium consisting of 1:1 DMEM/Hams F12 supplemented with 10% fetal bovine serum, 0.5% penicillin/streptomycin, 0.5% l-glutamine and 10 µg·mL−1 2-phospho-l-ascorbic acid trisodium salt (Sigma-Aldrich, Fluka, Seelze, Germany). After 24 h incubation, adhered chondrocytes were treated with trypsin (trypsin/EDTA 0.05%/0.02% in PBS) and frozen in complete medium containing 5% dimethyl sulphoxide (DMSO, Roth, Karlsruhe, Germany). All chemicals were obtained from Biochrom, Berlin, Germany, unless indicated otherwise.
Cell stimulation and treatment with inhibitor
For all experiments, thawed cells were cultivated for 1–3 days, treated with trypsin and pooled as indicated below and seeded at a density of 5 × 104 cells cm−2 in complete medium. For the microarray analysis, 3.8 × 106 cells were used per experiment, for the other experiments, 1.8 × 105 cells per batch were applied. After 24 h of adherence, they were silenced for 24 h in serum-free medium [DMEM containing 0.5% penicillin/streptomycin, 0.5% l-glutamine, 1% non-essential amino acids 100-fold concentrate, 1 mM pyruvate, 0.1% SES1 solution A (supplement for serum-free media) and 0.1% freshly added SES1 solution B (supplement containing insulin for serum-free media); all from Biochrom]. Cells were stimulated for the indicated time with 10 ng·mL−1 rhIL-1β (tebu-bio, Offenbach, Germany) in serum-free medium. Inhibitor-treated cells were incubated for 15 min prior to stimulation and subsequently co-incubated with 10 ng·mL−1 rhIL-1β and inhibitor, with a final DMSO concentration of 0.1% in the cultivation medium, for the indicated time span. For comparability, the same amount of DMSO was added to control cells. The inhibitors used were CBS-3868 ([4-[6-(4-fluoro-phenyl)-1-oxo-2,3-dihydro-1H-1lambda*4*-imidazo[2,1-b]thiazol-5-yl]-pyridin-2-yl]-((S)-1-phenyl-ethyl)-amine), Birb 796 (1-(5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl)-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]urea), pamapimod (pyrido[2,3-d]pyrimidin-7(8H)-one, 6-(2,4-difluorophenoxy)-2-[[3-hydroxy-1-(2-hydroxyethyl)propyl] amino]-8-methyl-) and SB203580 (4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole) (all provided by c-a-i-r biosciences GmbH, Tübingen, Germany). The kinase interactions of these p38 inhibitors are given in Table 1. At the end of the stimulation period, cells were washed twice in sterile PBS (PAA Laboratories, Egelsbach, Germany) and lysed in 600 µL lysis buffer RLT (Qiagen, Hilden, Germany) per 106 cells.
Table 1.
Specificities of the inhibitors for p38α MAPK and other kinases
| Gene symbol | Kinase target | Kd (nM) |
CBS-3868* | ||
|---|---|---|---|---|---|
| Birb 796 | SB203580 | Pamapimod | |||
| ABL1 | ABL1(T315I) | 42 | |||
| CSNK1D | CSNK1D | 37 | |||
| CSNK1E | CSNK1E | 100 | 260 | ||
| DDR1 | DDR1 | 1.9 | 1000 | ||
| DDR2 | DDR2 | 33 | 5000 | ||
| GAK | GAK | 19 | |||
| MAPK8 | JNK1 | 1100 | 190 | ||
| MAPK9 | JNK2 | 7.3 | 130 | 16 | × |
| MAPK10 | JNK3 | 110 | 35 | 19 | × |
| MAP4K4 | MAP4K4 | 90 | 3700 | ||
| MAPK11 | p38 Beta | 1500 | 70 | 120 | × |
| MAPK12 | p38 Gamma | 19 | 1500 | ||
| MAPK14 | p38 alpha | 0.37 | 12 | 1.3 | × |
| NLK | NLK | 1000 | 25 | 170 | |
| RIPK2 | RIPK2 | 24 | |||
| STK10 | LOK | 12 | |||
| TEK | TIE2 | 20 | |||
| TIE1 | TIE1 | 8.3 | |||
Kd values are according to Karaman et al. (2008; Birb 796 and SB203580) and Hill et al. (2008; pamapimod). Only those kinases at which at least one inhibitor shows a Kd < 100 nM are listed.
For CBS-3868, no Kd values were determined, but the inhibitor shows a relevant affinity for the indicated kinases (personal communications).
Microarray experiment
To obtain enough RNA for the microarray experiment, cells of six different donors were pooled after they had been thawed and treated as described above. After cell lysis, a whole human genome oligo-microarray (Human Genome Oligo-Set-Version 2.0, Operon, Ebersberg, Germany), representing 21 329 genes, was conducted at the Chip Facility of Ulm according to Buchholz et al. (2005). The experiment was performed in triplicate with six different donors each. By the use of this experimental design with biological replication, we could assess biological variation in spite of the need for pooling different donors (Kendziorski et al., 2005).
GoMiner analysis
Genes that showed at least a twofold regulation and a signicance level of P < 0.05 in the microarray analysis were assigned to Gene Ontologies by an analysing tool called GoMiner (http://discover.nci.nih.gov/gominer/) (Zeeberg et al., 2003). The Gene Ontology (GO) consortium offers three main ontologies, namely ‘biological process’, ‘cellular component’ and ‘molecular function’, subsuming subsequent terms that are organized in a tree-like structure. We used the ontology ‘biological process’. In brief, given a set of regulated genes, the set of all unique GO terms within the ontology was first identified that was associated with one or more of these genes. Next, the number of the regulated genes and the number of the genes that were assayed (all the genes represented on the microarray) were annotated at each term (Zeeberg et al., 2003).
Isolation of mRNA and cDNA synthesis
For isolating RNA from cultured cells, the RNeasy mini kit (Qiagen) was used according to the manufacturer's instructions. Briefly, the cell lysate was mixed 1:1 with 70% ethanol, loaded on a mini column and, after several washing steps and DNase digestion, the RNA was eluted in 30 µL of RNase free water. cDNA was synthesized with Omniscript RT (Qiagen) in accordance with the manufacturer's instructions using 12 µL of RNA solution.
Real-time PCR
Quantitative real-time PCR was used to detect human COX-2, mPGES1, iNOS, matrix metalloproteinase 13 (MMP13) and TNFRSF11B mRNA in human articular chondrocytes. The primers (Table 2) were designed using Primer 3 software (http://www.ncbi.nlm.nih.gov/tools/primer-blast/) except for the MMP13 primer which was assumed from literature (Bau et al., 2002). A gene-specific cDNA fragment was amplified for each gene, using the specified primers (Table 2), and sequenced with the Thermo Sequenase Primer Cycle Sequencing Kit (Amersham Biosciences, Freiburg, Germany), according to the manufacturer's instructions, on a fluorescent automated DNA sequencer (ALFexpress, Amersham Biosciences) to confirm the correct amplification products of the specific primers. Amplifications were done with an AbiPrism 7000 system (Applera, Darmstadt, Germany). For all the genes analysed, the Power SYBR Green PCR Master Mix (Applied Biosystems, Darmstadt, Germany) was used according to the manufacturer's instructions, except for MMP13, for which Invitrogen Platinum SYBR qPCR SuperMix UDG (Invitrogen, Karlsruhe, Germany) was utilized. 18S-rRNA was used as endogenous control; the concentration of primers applied was 1 µM (18S-rRNA, COX-2, MMP13 and TNFRSF11B) or 0.3 µM (mPGES1 and iNOS).
Table 2.
Sequences of primers used for quantitative real-time PCR
|
Gene |
GenBank |
Amplification |
Forward |
Reverse |
|---|---|---|---|---|
| symbol | accession no | length (bases) | 5′ 3′ | 5′ 3′ |
| 18SrRNA | X03205 | 65 | cgcagctaggaataatggaatagg | catggcctcagttccgaaa |
| COX-2 | NM_000963 | 123 | cccttgggtgtcaaaggtaa | ggcaaagaatgcaaacatca |
| MMP13 | NM_002427 | 97 | tcctcttcttgagctggactcatt | cgctctgcaaactggaggtc |
| iNOS | NM_000625 | 117 | attcactcagctgtgcatcg | tcaggtgggatttcgaagag |
| TNFRSF11B | NM_002546 | 117 | ggcaacacagctcacaagaa | cgctgttttcacagaggtca |
| mPGES1 | NM_004878 | 155 | cccccagtattgcaggag | ggaagaccaggaagtgcatc |
PGE2 and NO assays
Absolute concentrations of nitrite, a stable end product of NO metabolism, were determined in the media of the cell culture using a spectrophotometric method based on the Griess assay (Griess Reagent System, Promega, Mannheim, Deutschland) according to the manufacturer's instructions. Absorbance was measured at 550 nm, and nitrite concentration was determined by comparison with standard solutions of sodium nitrite.
PGE2 production was measured in the media by a high sensitivity, commercially available enzyme immunoassay kit (PGE2 ELISA Correlate EIA, Biotrend, Assay Designs, Köln, Germany) according to the manufacturer's instructions. The sensitivity was 13 pg·mL−1. PGE2 concentration was determined in duplicate and was read against a standard curve.
Determination of % inhibition and IC50 values
The results of quantitative real-time PCR and metabolite determination were used to calculate % inhibition normalized to stimulatability of the cells according to the following formula:
with Hpn = normalized percentage inhibition, vC = value of control, vS = value of stimulation and vI = value of inhibition probe.
The IC50 values were determined by the two-point form of a linear equation. Given the lowest inhibitor concentration (x1) with which inhibition of more than 50% was achieved, and the highest inhibitor concentration (x2) with which an inhibition of less than 50% was achieved, the IC50 value was calculated accordingly as follows:
with y1 = Hpn in x1 and y2 = Hpn in x2.
Nomenclature
The nomenclature of genes and molecular targets conforms to BJP's Guide to Receptors and Channels (Alexander et al., 2008).
Statistical analysis
For the microarray results, a print-tip loess normalization according to Buchholz et al. (2005) and a moderated t-test (Smyth, 2004) were performed at the Chip Facility of Ulm. In the GoMiner analysis, the two-sided Fisher's exact test evaluated whether there were more genes of interest at the term than one might expect by chance (Zeeberg et al., 2003). For standardization of the gene expression levels determined by TaqMan analysis, mRNA expression was normalized to 18SrRNA expression. Differential regulation was determined by calculating the ratios of gene expression in different treatments. A two-tailed paired t-test of the calculated ratios was performed to evaluate significant differences from the relative control treatment; as always, identical donors were compared.
Results
Control of cell phenotype and viability
The OA cartilage used for the experiments, macroscopically, had a smooth surface and no severe osteoarthritic changes. As described elsewhere (Joos et al., 2008), analysis of collagen type II, aggrecan and cartilage oligomeric matrix protein expression confirmed a stable differentiation stage of the chondrocytes during the experimental period. Trypan blue staining showed comparable cell viability with and without IL-1β stimulation.
Microarray experiment
The first objective was to characterize the effects of p38MAPK inhibition on global gene expression in IL-1β-stimulated human chondrocytes. To this end, three chondrocyte cultures, each composed of cells from six different donors, were stimulated with 10 ng·mL−1 IL-1β in the presence or absence of 10 µM SB203580 and 10 µM Birb 796 (Doramapimod) respectively. After 24 h of stimulation, RNA was isolated and subjected to a whole human genome oligo-microarray analysis (Human Genome Oligo-Set-Version 2.0, Operon) as described previously (Joos et al., 2008). All microarray data have been deposited on ArrayExpress (http://www.ebi.ac.uk/microarray-as/aer; Accession number: E-MEXP-1434).
Effects of SB203580 and Birb 796 on whole-genome gene expression
Stimulation of chondrocytes with IL-1β resulted in an up- or down-regulation of the expression of 1141 genes when compared to controls. This IL-1β-induced gene expression profile was used as a reference to analyse the effects of the p38α/β MAPK inhibitors on gene expression. In the presence of SB203580, 646 genes were modulated and 116 thereof (=18%) were co-regulated by IL-1β and SB203580. Most of the co-modulated genes (87%) were regulated in opposite directions, 13% moved unidirectional, whereas most genes were up-regulated. Co-incubation with Birb 796 affected 503 genes with 208 genes (=41%) co-regulated by Birb 796 and IL-1β; 98% of these co-modulated genes were regulated in opposite directions. Among the genes analysed on the microarray, some are hypothetical or unknown. The list of known co-regulated genes with their accession numbers, fold change and P values for IL-1β and Birb 796 regulation is shown in Supporting Information Table S1. The genes that were co-regulated by IL-1β and SB203580 have been presented in a previous study (Joos et al., 2009).
Effects of SB203580 and Birb 796 on biological processes
For the identification of the biological processes regulated, the genes regulated by IL-1β, SB203580 or Birb 796 were analysed with the GoMiner software tool and classified into biological coherent categories. A Fisher's exact test evaluated GO terms with a significant accumulation of changed genes on all levels of the hierarchical GO tree structure. In the GO ‘biological processes’, 215, 145 and 58 processes were found to be regulated by IL-1β, Birb 796 and SB203580, respectively (see Supporting Information Tables S2–S4). A comparison of the regulated processes revealed 27 terms that were co-regulated by IL-1β and Birb 796 (including 19% of all Birb 796-regulated genes), and five terms that were co-regulated by IL-1β and SB203580 (including 9% of all SB203580-regulated genes). The terms of the subcategories were topically allocated to main fields. IL-1β mainly affected genes in the field ‘response to stimulus’ that involved 43 subterms. The main topics in this field were ‘immune response’, ‘response to oxidative stress’ and ‘cytokine production’. In addition, eight cytoskeleton-associated terms were affected. In contrast, only a few of the SB203580-affected processes could be allocated to main fields. Thirteen terms could be assigned to the field ‘response to stimulus’, involving three oxidative stress processes. Five terms were associated with the cytoskeleton. Birb 796 influenced the main term ‘response to stimulus’ with 26 subterms like ‘immune response’ and ‘response to oxidative stress’, but revealed no impact on the cytoskeleton. Furthermore, Birb 796 affected 12 processes involved in apoptosis and cell death. A closer look at the genes involved revealed that, for example, death-associated protein kinase 3 and programmed cell death 2 were up-regulated by Birb 796, whereas the anti-apoptotic gene baculoviral IAP repeat-containing 3 was down-regulated.
Cluster analysis of regulated genes
Cluster analysis of the microarray data was used to study the gene expression patterns of IL-1β- and p38MAPK inhibitor-treated chondrocytes. Microarray data were assigned to the software tool ‘Genesis’ in order to perform a hierarchical clustering. Tools for visualization of the gene expression data allowed us to identify 334 genes that were up-regulated by IL-1β and differentially regulated by SB203580 and/or Birb 796. A possible role in RA and OA was ascribed to 35 of those genes, which are listed in Supporting Information Table S5. In order to investigate pathophysiological parameters of OA with widely accepted relevance for in vivo models, COX-2, MMP13, inducible NOS (iNOS) and TNFRSF11B (osteoprotegerin) were chosen as panel of genes for further quantitative analyses. They are all actively involved in the pathogenesis of OA and RA, and are expected to correlate with the course of the disease. COX-2 and iNOS are involved in the synthesis of inflammatory mediators, MMP13 is a major catabolic protease in OA and osteoprotegerin has been shown to play a role in the progression of OA (Schieven, 2005; Goldring and Goldring, 2007; Schett et al., 2008; Kwan Tat et al., 2009). The expression of these genes may be used to distinguish different p38α MAPK inhibitors and may form a suitable test system for inhibitor characterization.
Quantitative characterization of p38α/β MAPK inhibitors
For inhibitor characterization, the gene expression of COX-2, MMP13, iNOS and TNFRSF11B was quantitatively analysed. In addition to the gene expression of mPGES1, the release of prostaglandin E2 (PGE2) was measured as an indicator of the activity of COX-2 and mPGES1 on protein levels. The inhibition of the NO synthesis pathway was further analysed by determination of NO release. To evaluate this test system, several inhibitors were administered to IL-1β-stimulated chondrocytes, and the specified outcome parameters were determined. The tested substances included the three established p38MAPK inhibitors Birb 796, SB203580 and pamapimod, as well as a new p38α/β selective agent under development (CBS-3868).
Effects of p38MAPK inhibitors on PGE2 synthesis pathway
The effects of the inhibitors on the PGE2 synthesis pathway are shown in Figure 1. The stimulation of OA chondrocytes increased gene expression of COX-2 after 4 and 24 h by a factor of 30 and 150 respectively. The p38 inhibitors repressed this stimulation, in a concentration-dependent manner, up to 90%. The IC50 values were all below 0.1 µM, except for that of SB203580 (IC50 = 0.9 µM). The IC50 values for all measurements are given in Table 3.
Figure 1.

Inhibition of IL-1β-induced PGE2 synthesis, and gene expression of the enzymes involved, by anti-inflammatory substances. Chondrocytes from two donors were pooled and cultivated with inhibitor and 10 ng·mL−1 IL-1β. mRNA was isolated for quantitative gene expression analysis, and PGE2 levels in the supernatant were determined by elisa. Data were obtained from at least six independent experiments with 12 different donors, except for 10 µM Birb 796 and 10 µM CBS-3868 where three independent experiments with six different donors were used. (A) % Inhibition of IL-1β-induced COX-2 gene expression after 4 and 24 h. (B) % Inhibition of IL-1β-induced mPGES1 gene expression after 24 h. (C) % Inhibition of IL-1β-induced PGE2 release after 4 and 24 h. Significant inhibition and concentration-related dependence is marked (*P < 0.05; **P < 0.01).
Table 3.
IC50 values of the inhibitors tested for inhibition of gene expression and metabolite release in µM
| Time (h) | Birb 796 | CBS-3868 | Pamapimod | SB203580 | ||
|---|---|---|---|---|---|---|
| Gene expression | COX-2 | 4 | <0.1 | <0.1 | 0.1 | 0.9 |
| COX-2 | 24 | <0.1 | <0.1 | 0.2 | 0.6 | |
| mPGES1 | 24 | 3 | 0.6 | 1 | 0.7 | |
| iNOS | 24 | 7 | 2 | >1 | 10 | |
| MMP13 | 24 | <0.1 | <0.1 | 0.7 | 0.6 | |
| TNFRSF11B | 24 | >10 | <0.1 | >1 | 0.9 | |
| Release | PGE2 | 4 | <0.1 | <0.1 | 0.9 | 0.1 |
| PGE2 | 24 | <0.1 | <0.1 | 0.6 | 0.9 | |
| NO | 24 | 6 | 6 | >1 | >10 |
The gene expression of mPGES1 was augmented threefold after 4 h (P = 0.001) and 11-fold after 24 h (P < 0.001) by IL-1β, respectively. As seen in Figure 1B, co-incubation with p38α/β MAPK inhibitors resulted in an approx. 50% inhibition of the IL-1β-induced expression with IC50 values between 0.6 and 3 µM. The inhibitory effect on mPGES1 gene expression, determined 4 h after chondrocyte stimulation was statistically not significant.
To estimate the activity of the enzymes COX-2 and mPGES1 in IL-1β-treated chondrocytes, the release of their product PGE2 was measured in the presence and absence of p38 α/β inhibitors. IL-1β stimulation augmented the PGE2 concentration in the supernatant from 0.9 to 6.0 ng·mL−1 after 4 h, and from 1.3 to 11.6 ng·mL−1 after 24 h. All tested substances acted as strong inhibitors (Figure 1C) with IC50 values below or around 0.1 µM; only pamapimod and SB203580 showed IC50 values up to 0.9 µM (Table 3). The effects of all the inhibitors, except for Birb 796, were concentration dependent.
Effects of p38MAPK inhibitors on NO synthesis pathway
To examine the effect of the pharmaceutical agents on the NO synthesis pathway, modulation of iNOS gene expression and NO release was analysed. The results are shown in Figure 2. As NO is rapidly oxidized, nitrite concentration was determined in the supernatant of treated chondrocytes as an indicator for NO production. IL-1β stimulation caused a 250- and 370-fold increase in iNOS gene expression after 4 and 24 h respectively. No significant down-regulation could be detected after 4 h incubation with inhibitor. After 24 h, Birb 796, CBS-3868 and SB203580 caused a significant repression of iNOS gene expression of 50–70% with IC50 values between 2 and 10 µM. Nitrite release was increased by IL-1β after 24 h, but not after 4 h from 1.2 to 6.2 µM (P < 0.01). This IL-1β-induced increase in NO was inhibited by high concentrations of the inhibitors, but the effects were not statistically significant. The IC50 values were 6 µM except for SB203580 where the IC50 > 10 µM (Table 3).
Figure 2.

Inhibition of IL-1β-induced iNOS gene expression and NO synthesis by anti-inflammatory substances. Chondrocytes from two donors were pooled and cultivated with inhibitor and 10 ng·mL−1 IL-1β. mRNA was isolated for quantitative gene expression analysis, and NO levels in the supernatant were obtained by determination of nitrite concentration with the Griess assay. Data were obtained from at least six independent experiments with 12 different donors, except for 10 µM Birb 796 and 10 µM CBS-3868 where three independent experiments with six different donors were used. (A) % Inhibition of IL-1β-induced iNOS gene expression after 24 h. (B) % Inhibition of IL-1β-induced nitrite release after 24 h. Significant inhibition and concentration-related dependence is marked (+P = 0.054; *P < 0.05; **P < 0.01).
Effects of p38MAPK inhibitors on MMP13 and TNFRSF11B gene expression
In addition, the impact of the anti-inflammatory substances on MMP13 and TNFRSF11B gene expression was examined. MMP13 was threefold (P = 0.08) and 43-fold (P = 0.014) up-regulated by IL-1β after 4 and 24 h respectively. In samples analysed 4 h after IL-1β stimulation, in the presence of CBS-3868, the up-regulation of MMP13 gene expression was significantly inhibited, whereas the other inhibitors had no significant effect. The drug-mediated effects, determined after 24 h, are shown in Figure 3A. At 10 µM, the test compounds inhibited MMP13 expression by 80% to almost 100%. The IC50 values of Birb 796 and CBS-3868 were below 0.1 µM, and the IC50 values of SB203580 and pamapimod were 0.6 and 0.7 µM, respectively (Table 3). TNFRSF11B was increased three- and fivefold by IL-1β after 4 and 24 h respectively. CBS-3868 (1 µM, P = 0.0103) and pamapimod (1 µM, P = 0.037) inhibited the increase in TNFRSF11B after 4 h significantly. CBS-3868 and SB203580 significantly down-regulated TNFRSF11B gene expression after 24 h (Figure 3B). Birb 796 did not show a significant inhibitory effect at either time-point. IC50 values are given in Table 3. In addition, IC75 values were determined from all the concentration–effect curves and included in Supporting Information Table S6).
Figure 3.

Inhibition of IL-1β-induced MMP13 and TNFRSF11B gene expression by anti-inflammatory substances. Chondrocytes from two donors were pooled and cultivated with inhibitor and 10 ng·mL−1 IL-1β, and then mRNA was isolated for quantitative gene expression analysis. Data were obtained from at least six independent experiments with 12 different donors, except for 10 µM Birb 796 and 10 µM CBS-3868 where three independent experiments with six different donors were used. (A) % Inhibition of IL-1β-induced MMP13 gene expression after 24 h. (B) % Inhibition of IL-1β-induced TNFRSF11B gene expression after 24 h. Significant inhibition and concentration-related dependence is marked (*P < 0.05; **P < 0.01).
Discussion and conclusions
In the present study, the effect of p38α/β MAPK inhibitors on the expression of inflammation-related genes in IL-1β-stimulated, primary human chondrocytes was investigated. Initially, a genome-wide microarray analysis was performed with stimulated chondrocytes in the presence or absence of the test article Birb 796, and the results were compared with those obtained with SB203580. The objective of this analysis was to identify genes, which are related to the IL-1β-induced inflammatory processes in chondrocytes and, which are suitable to qualitatively and quantitatively determine the effects of anti-inflammatory drugs on the expression of these genes. A rather high inhibitor concentration of 10 µM was chosen to detect the effects of all relevant inhibitors, as chondrocytes are less sensitive than blood cells (unpublished observation). As published, the results of IL-1β-mediated induction of gene expression and the effect of SB203580 demonstrated that the model produces reliable and reproducible results (Joos et al., 2008; 2009;). Many of the IL-1β-regulated genes (e.g. MMP13, SOD2 or GADD45 β) have also been shown to be differentially expressed in OA tissue (Aigner et al., 2006; Sato et al., 2006; Ijiri et al., 2008). These findings support the relevance of the in vitro microarray results to in vivo processes. The lists of genes regulated that were obtained also represent a valuable database for the selection of further OA-related genes that could extend the set of parameters used for the testing of anti-inflammatory substances.
The comparison of effects of Birb 796 and SB203580 (10 µM each) demonstrated that the number of genes modulated by SB203580 was higher than that of Birb 796. Quantitative evaluation of global expression analysis also demonstrated that SB203580 and Birb 796 counter-regulated 87 and 98%, respectively, of those genes that were regulated by IL-1β and the test articles. This analysis indicates that both compounds may modulate additional targets at the concentration used, 10 µM, in line with the knowledge that both compounds are not absolutely selective for p38α/β MAPK (Table 1). Therefore, it is possible that off-target effects contribute to the results of this study. In addition, the analysis of regulated biological processes would allow for the screening for undesired side effects of the test articles. Apoptosis of chondrocytes for example plays an important role in OA (Kuhn et al., 2004). While the application of SB203580 did not reveal alarming gene regulation with respect to pro-apoptotic genes (Joos et al., 2009), Birb 796 influenced apoptotic processes in the GoMiner analysis. The direction of regulation indicated a pro-apoptotic effect, which might represent a possible negative side effect and warrants further investigation.
Based on the results of global gene expression analysis, a set of genes involved in inflammatory processes (PGE2 and NO pathway), matrix degradation (MMP13) and bone metabolism (TNFRSF11B) were selected to investigate the effect of different p38α/β MAPK inhibitors on gene expression. Analysis of the PGE2 synthesis pathway included the specification of COX-2 and mPGES1 gene expression, and release of PGE2 into the supernatant of chondrocyte cultures. All inhibitors significantly abolished IL-1β-induced up-regulation of COX-2 after 4 and 24 h incubation. This effect is in accordance with another work and indicates that p38α MAPK inhibition directly blocks COX-2 transcription (Cieslik et al., 2002). When compared to the potency of Birb 796, CBS-3868 and pamapimod, SB203580 was less efficient at inhibiting COX-2 expression. This outcome is in line with the relative potencies of the compounds regarding p38α MAPK inhibition. In an enzyme assay, the IC50 of SB203580 was 19.5-, 14.1- and 2.2-fold higher than that of Birb 796, CBS-3868 and pamapimod, respectively (unpublished results, c-a-i-r biosciences GmbH).
IL-1β-induced increase in mPGES1 gene expression was less pronounced than that of COX-2, and a significant inhibition of gene expression was observed only after 24 h. With an IC50 value of 3 µM, Birb 796 was the weakest inhibitor of mPGES1 expression, while CBS-3868 and SB203580 showed a somewhat higher potency with IC50 values of 0.7 and 0.6 µM. Pamapimod inhibited the IL-1β-induced mPGES1 expression with an IC50 value of 1 µM. Masuko-Hongo et al. investigated the regulation of mPGES1 expression based on the effects of the p38α/β MAPK inhibitor SB203580 and a known p38α-selective inhibitor SC-906. The authors concluded that the expression of mPGES1 is regulated by p38β rather than p38α (Masuko-Hongo et al., 2004). The results of the present study support the concept of an involvement of p38β MAPK, as the published Kd values of the ligand/p38β MAPK affinity determined for SB203580 (70 nM), pamapimod (120 nM) and Birb 796 (1500 nM) (Hill et al., 2008; Karaman et al., 2008) correlate with the inhibition of mPGES1 expression. Yet, other mechanisms like ERK1/2 signalling may contribute to our findings (Masuko-Hongo et al., 2004).
Birb 796 and CBS-3868, according to their effect on COX-2 gene expression, exerted the strongest effect on PGE2 synthesis with an IC50 of <0.1 µM. The efficacy of pamapimod and SB203580 was weaker by a factor of 10, and correlates with their IC50 values of COX-2 gene expression. Consequently, the drug-mediated effect on COX-2 expression was thought to be more relevant for the inhibition of PGE2 synthesis than their effect on mPGES1.
The effects of the inhibitors on the NO synthesis pathway were examined by the analysis of iNOS gene expression and nitrite release as an indicator of NO formation. Although it has been suggested that a p38-dependent mechanism is involved in the regulation of iNOS expression and NO synthesis (Mendes et al., 2002), the p38α/β MAPK inhibitors tested did not seem to directly prevent the induction of iNOS. A significant inhibition of iNOS expression was achieved with SB203580 (50% at 10 µM) and CBS-3868 (70% at 10 µM) only after 24 h. Both the modest-to-moderate extent of inhibition and the time-course are in agreement with the multiple mechanisms of regulation for iNOS gene expression described previously, and confirm that p38α MAPK is neither an immediate nor the only regulator of iNOS expression and NO synthesis (Otero et al., 2005; Chockalingam et al., 2007; Chowdhury et al., 2008). The IL-1β-mediated induction of MMP13 gene expression was efficiently and concentration dependently inhibited by all test compounds. At concentrations of 1 and 10 µM, the extent of inhibition was similar. At 0.1 µM, the high degree of inhibition, which was achieved with Birb 796 and CBS-3868 compared to pamapimod and SB203580, correlated with the inhibitory potencies of the test compounds on p38α MAPK activity. A promising new approach for inhibition of cartilage degradation was recently introduced by Kimura et al. (2009) who presented a new inhibitor that, in contrast to SB203580, inhibited MMP13 expression, but not the expression of other MMPs.
The effects of the p38MAPK inhibitors on TNFRSF11B gene expression were divergent. In vivo, the so-called decoy receptor TNFRSF11B interferes with RANK/RANKL signalling, thereby preventing the RANKL-mediated osteoclastogenesis (Schett et al., 2003). An up-regulation of TNFRSF11B protein has not only been described in IL-1β-treated chondrocytes (Kwan Tat et al., 2009), but also in OA cartilage and in the synovium of RA patients (Komuro et al., 2001). The low extent of induction, three- and fivefold up-regulation after 4 and 24 h, made it difficult to detect the drug-mediated effects unequivocally. Birb 796 was the weakest inhibitor of TNFRSF11B gene expression, indicating the contribution of another mechanism rather than p38α-mediated signalling. However, in osteosarcoma cells, p38α/β, but not JNK, ERK or NFκB, inhibition was shown to influence IL-1β-induced TNFRSF11B gene expression (Lambert et al., 2007). It is possible that a comparable mechanism exists in chondrocytes, and therefore an effect mediated by p38β could play a role in the regulation of TNFRSF11B expression.
In summary, in the present study, a reliable in vitro model using IL-1β-stimulated human primary chondrocytes was established with the objective to investigate and compare the effects of different p38α/β MAPK inhibitors on gene expression. The role of p38MAPK and JNK isoforms in the regulation of the analysed biomarkers is illustrated in Figure 4. It was demonstrated that the effects of the test compounds on COX-2 and MMP13 expression, as well as on PGE2 release, correlated well with their potency at inhibiting p38α MAPK. In contrast, their effect on mPGES1 and TNFRSF11B expression appeared to be associated with the affinity of the test compounds for p38β rather than the α-form of MAPK. These observations shed new light on the role of p38β MAPK in chondrocytes and on the required α/β-specificity of p38MAPK inhibitors. Although in the case of mPGES1, they confirm those obtained in a previous study (Masuko-Hongo et al., 2004). Undoubtedly, further studies are required to unequivocally verify these findings. iNOS expression and NO release appear to be useful, as biomarkers of inflammation, for differentiating the efficacy of p38α/β MAPK inhibitors. Marked differences were observed with the inhibitors tested, especially at low concentrations, which may be more relevant in vivo because of their limited bioavailability within cartilage tissue. Despite the selection of candidate genes for differential analysis of test substances with respect to well-known relevance to the in vivo situation, the correlation of our results with in vivo models remains to be determined. Overall, our tissue-specific test system could be successfully applied for differential characterization of inhibitors with the same primary pharmaceutical target. It therefore represents a valuable tool for drug screening between functional in vitro testing and in vivo models in the field of OA.
Figure 4.

Schematic presentation of inhibitor effects on the expression of the gene analysed and on metabolite release. The size of the blocking bar indicates the affinity of the inhibitor for the respective target according to their Kd values (Hill et al., 2008; Karaman et al., 2008). The estimation of CBS-3868 affinities is based on IC50 determinations (personal communication, c-a-i-r biosciences GmbH, Tübingen, Germany). Dashed arrows indicate signalling pathways with intermediate steps. AA, arachidonic acid; IL-1β, interleukin 1β; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; TNFRSF11B, osteoprotegerin. The scheme gives an overview of regulatory mechanisms discussed in this paper. The different target affinities and the different off-target specificities of the inhibitors are a possible explanation for their different impacts described in the presented study.
Acknowledgments
The authors would like to thank Dr Holzmann from the Microarray-Facility Ulm for his support in the chip analysis. This work was in part supported by Merckle GmbH, Ulm and the German Research Council (DFG, Grant KFO 200).
Glossary
Abbreviations
- IL-1β
interleukin-1β
- iNOS
inducible NOS
- MAPK
mitogen-activated protein kinase
- MMP13
matrix metalloproteinase 13
- mPGES1
prostaglandin E synthase
- PGE2
prostaglandin E2
- TNFRSF11B
osteoprotegerin
Conflicts of interest
The authors declare that they have no competing interests.
Supporting information
Additional Supporting Information may be found in the online version of this article.
Table S1 Co-regulated genes by IL-1beta and Birb 796.
Table S2 Regulated biological processes by IL-1beta.
Table S3 Regulated biological processes by Birb 796.
Table S4 Regulated biological processes by SB203580.
Table S5 Microarray data of differentially regulated genes selected by hierarchical clustering and associated with OA.
Table S6 IC75 values of the tested inhibitors for inhibition of gene expression and metabolite release.
Please note: Wiley-Blackwell are not responsible for the content or functionality of any supporting materials supplied by the authors. Any queries (other than missing material) should be directed to the corresponding author for the article.
References
- Aigner T, Soeder S, Haag J. IL-1beta and BMPs – interactive players of cartilage matrix degradation and regeneration. Eur Cell Mater. 2006;12:49–56. doi: 10.22203/ecm.v012a06. discussion 56. [DOI] [PubMed] [Google Scholar]
- Alexander SP, Mathie A, Peters JA. Guide to Receptors and Channels (GRAC), 3rd edition. Br J Pharmacol. 2008;153:S1–209. doi: 10.1038/sj.bjp.0707746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alten RE, Zerbini C, Jeka S, Irazoque F, Khatib F, Emery P, et al. Efficacy and safety of pamapimod in patients with active rheumatoid arthritis receiving stable methotrexate therapy. Ann Rheum Dis. 2010;69:364–367. doi: 10.1136/ard.2008.104802. [DOI] [PubMed] [Google Scholar]
- Ashwell JD. The many paths to p38 mitogen-activated protein kinase activation in the immune system. Nat Rev Immunol. 2006;6:532–540. doi: 10.1038/nri1865. [DOI] [PubMed] [Google Scholar]
- Bau B, Gebhard PM, Haag J, Knorr T, Eckart B, Aigner T. Relative messenger RNA expression profiling of collagenases and aggrecanases in human articular chondrocytes in vivo and in vitro. Arthritis Rheum. 2002;46:2648–2657. doi: 10.1002/art.10531. [DOI] [PubMed] [Google Scholar]
- Branger J, Blink B, Weijer S, Madwed J, Bos CL, Gupta A, et al. Anti-inflammatory effects of a p38 mitogen-activated protein kinase inhibitor during human endotoxemia. J Immunol. 2002;168:4070–4077. doi: 10.4049/jimmunol.168.8.4070. [DOI] [PubMed] [Google Scholar]
- Buchholz M, Braun M, Heidenblut A, Kestler HA, Kloeppel G, Schmiegel W, et al. Transcriptome analysis of microdissected pancreatic intraepithelial neoplastic lesions. Oncogene. 2005;24:6626–6636. doi: 10.1038/sj.onc.1208804. [DOI] [PubMed] [Google Scholar]
- Chockalingam PS, Varadarajan U, Sheldon R, Fortier E, LaVallie ER, Morris EA, et al. Involvement of protein kinase Czeta in interleukin-1beta induction of ADAMTS-4 and type 2 nitric oxide synthase via NF-kappaB signaling in primary human osteoarthritic chondrocytes. Arthritis Rheum. 2007;56:4074–4083. doi: 10.1002/art.23043. [DOI] [PubMed] [Google Scholar]
- Chowdhury TT, Salter DM, Bader DL, Lee DA. Signal transduction pathways involving p38 MAPK, JNK, NFkappaB and AP-1 influences the response of chondrocytes cultured in agarose constructs to IL-1beta and dynamic compression. Inflamm Res. 2008;57:306–313. doi: 10.1007/s00011-007-7126-y. [DOI] [PubMed] [Google Scholar]
- Cieslik K, Zhu Y, Wu KK. Salicylate suppresses macrophage nitric-oxide synthase-2 and cyclo-oxygenase-2 expression by inhibiting CCAAT/enhancer-binding protein-beta binding via a common signaling pathway. J Biol Chem. 2002;277:49304–49310. doi: 10.1074/jbc.M205030200. [DOI] [PubMed] [Google Scholar]
- Cohen SB, Cheng T-T, Chindalore V, Damjanov N, Burgos-Vargas R, Delora P, et al. Evaluation of the efficacy and safety of pamapimod, a p38 MAP kinase inhibitor, in a double-blind, methotrexate-controlled study of patients with active rheumatoid arthritis. Arthritis Rheum. 2009;60:335–344. doi: 10.1002/art.24266. [DOI] [PubMed] [Google Scholar]
- Damjanov N, Kauffman RS, Spencer-Green GT. Efficacy, pharmacodynamics, and safety of VX-702, a novel p38 MAPK inhibitor, in rheumatoid arthritis: results of two randomized, double-blind, placebo-controlled clinical studies. Arthritis Rheum. 2009;60:1232–1241. doi: 10.1002/art.24485. [DOI] [PubMed] [Google Scholar]
- Dominguez C, Powers DA, Tamayo N. p38 MAP kinase inhibitors: many are made, but few are chosen. Curr Opin Drug Discov Devel. 2005;8:421–430. [PubMed] [Google Scholar]
- Fijen JW, Zijlstra JG, De Boer P, Spanjersberg R, Tervaert JW, Van Der Werf TS, et al. Suppression of the clinical and cytokine response to endotoxin by RWJ-67657, a p38 mitogen-activated protein-kinase inhibitor, in healthy human volunteers. Clin Exp Immunol. 2001;124:16–20. doi: 10.1046/j.1365-2249.2001.01485.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Genovese MC. Inhibition of p38: has the fat lady sung? Arthritis Rheum. 2009;60:317–320. doi: 10.1002/art.24264. [DOI] [PubMed] [Google Scholar]
- Goldring MB, Goldring SR. Osteoarthritis. J Cell Physiol. 2007;213:626–634. doi: 10.1002/jcp.21258. [DOI] [PubMed] [Google Scholar]
- Hill RJ, Dabbagh K, Phippard D, Li C, Suttmann RT, Welch M, et al. Pamapimod, a novel p38 mitogen-activated protein kinase inhibitor: preclinical analysis of efficacy and selectivity. J Pharmacol Exp Ther. 2008;327:610–619. doi: 10.1124/jpet.108.139006. [DOI] [PubMed] [Google Scholar]
- Ijiri K, Zerbini LF, Peng H, Otu HH, Tsuchimochi K, Otero M, et al. Differential expression of GADD45beta in normal and osteoarthritic cartilage: potential role in homeostasis of articular chondrocytes. Arthritis Rheum. 2008;58:2075–2087. doi: 10.1002/art.23504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joos H, Albrecht W, Laufer S, Reichel H, Brenner RE. IL-1beta regulates FHL2 and other cytoskeleton-related genes in human chondrocytes. Mol Med. 2008;14:150–159. doi: 10.2119/2007-00118.Joos. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joos H, Albrecht W, Laufer S, Brenner RE. Influence of p38MAPK inhibition on IL-1beta-stimulated human chondrocytes: a microarray approach. Int J Mol Med. 2009;23:685–693. doi: 10.3892/ijmm_00000181. [DOI] [PubMed] [Google Scholar]
- Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–132. doi: 10.1038/nbt1358. [DOI] [PubMed] [Google Scholar]
- Kendziorski C, Irizarry RA, Chen KS, Haag JD, Gould MN. On the utility of pooling biological samples in microarray experiments. Proc Natl Acad Sci USA. 2005;102:4252–4257. doi: 10.1073/pnas.0500607102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura H, Yukitake H, Suzuki H, Tajima Y, Gomaibashi K, Morimoto S, et al. The chondroprotective agent ITZ-1 inhibits interleukin-1beta-induced matrix metalloproteinase-13 production and suppresses nitric oxide-induced chondrocyte death. J Pharmacol Sci. 2009;110:201–211. doi: 10.1254/jphs.09076fp. [DOI] [PubMed] [Google Scholar]
- Komuro H, Olee T, Kühn K, Quach J, Brinson DC, Shikhman A, et al. The osteoprotegerin/receptor activator of nuclear factor kappaB/receptor activator of nuclear factor kappaB ligand system in cartilage. Arthritis Rheum. 2001;44:2768–2776. doi: 10.1002/1529-0131(200112)44:12<2768::aid-art464>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
- Kuhn K, D'Lima DD, Hashimoto S, Lotz M. Cell death in cartilage. Osteoarthritis Cartilage. 2004;12:1–16. doi: 10.1016/j.joca.2003.09.015. [DOI] [PubMed] [Google Scholar]
- Kwan Tat S, Amiable N, Pelletier JP, Boileau C, Lajeunesse D, Duval N, et al. Modulation of OPG, RANK and RANKL by human chondrocytes and their implication during osteoarthritis. Rheumatology (Oxford) 2009;48:1482–1490. doi: 10.1093/rheumatology/kep300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lambert C, Oury C, Dejardin E, Chariot A, Piette J, Malaise M, et al. Further insights in the mechanisms of interleukin-1beta stimulation of osteoprotegerin in osteoblast-like cells. J Bone Miner Res. 2007;22:1350–1361. doi: 10.1359/jbmr.070508. [DOI] [PubMed] [Google Scholar]
- Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, et al. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature. 1994;372:739–746. doi: 10.1038/372739a0. [DOI] [PubMed] [Google Scholar]
- Masuko-Hongo K, Berenbaum F, Humbert L, Salvat C, Goldring MB, Thirion S. Up-regulation of microsomal prostaglandin E synthase 1 in osteoarthritic human cartilage: critical roles of the ERK-1/2 and p38 signaling pathways. Arthritis Rheum. 2004;50:2829–2838. doi: 10.1002/art.20437. [DOI] [PubMed] [Google Scholar]
- Mendes AF, Caramona MM, Carvalho AP, Lopes MC. Role of mitogen-activated protein kinases and tyrosine kinases on IL-1-induced NF-kappaB activation and iNOS expression in bovine articular chondrocytes. Nitric Oxide. 2002;6:35–44. doi: 10.1006/niox.2001.0378. [DOI] [PubMed] [Google Scholar]
- Nieminen R, Leinonen S, Lahti A, Vuolteenaho K, Jalonen U, Kankaanranta H, et al. Inhibitors of mitogen-activated protein kinases downregulate COX-2 expression in human chondrocytes. Mediators Inflamm. 2005;2005:249–255. doi: 10.1155/MI.2005.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otero M, Lago R, Lago F, Reino JJG, Gualillo O. Signalling pathway involved in nitric oxide synthase type II activation in chondrocytes: synergistic effect of leptin with interleukin-1. Arthritis Res Ther. 2005;7:R581–R591. doi: 10.1186/ar1708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ross S, Chen T, Yu V, Tudor Y, Zhang D, Liu L, et al. High-content screening analysis of the p38 pathway: profiling of structurally related p38alpha kinase inhibitors using cell-based assays. Assay Drug Dev Technol. 2006;4:397–409. doi: 10.1089/adt.2006.4.397. [DOI] [PubMed] [Google Scholar]
- Sato T, Konomi K, Yamasaki S, Aratani S, Tsuchimochi K, Yokouchi M, et al. Comparative analysis of gene expression profiles in intact and damaged regions of human osteoarthritic cartilage. Arthritis Rheum. 2006;54:808–817. doi: 10.1002/art.21638. [DOI] [PubMed] [Google Scholar]
- Schett G, Redlich K, Smolen JS. The role of osteoprotegerin in arthritis. Arthritis Res Ther. 2003;5:239–245. doi: 10.1186/ar990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schett G, Zwerina J, Firestein G. The p38 mitogen-activated protein kinase (MAPK) pathway in rheumatoid arthritis. Ann Rheum Dis. 2008;67:909–916. doi: 10.1136/ard.2007.074278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schieven GL. The biology of p38 kinase: a central role in inflammation. Curr Top Med Chem. 2005;5:921–928. doi: 10.2174/1568026054985902. [DOI] [PubMed] [Google Scholar]
- Smith SJ, Fenwick PS, Nicholson AG, Kirschenbaum F, Finney-Hayward TK, Higgins LS, et al. Inhibitory effect of p38 mitogen-activated protein kinase inhibitors on cytokine release from human macrophages. Br J Pharmacol. 2006;149:393–404. doi: 10.1038/sj.bjp.0706885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3 doi: 10.2202/1544-6115.1027. Article3. [DOI] [PubMed] [Google Scholar]
- Sweeney SE. The as-yet unfulfilled promise of p38 MAPK inhibitors. Nat Rev Rheumatol. 2009;5:475–477. doi: 10.1038/nrrheum.2009.171. [DOI] [PubMed] [Google Scholar]
- Westra J, Doornbos-van der Meer B, de Boer P, van Leeuwen MA, van Rijswijk MH, Limburg PC. Strong inhibition of TNF-alpha production and inhibition of IL-8 and COX-2 mRNA expression in monocyte-derived macrophages by RWJ 67657, a p38 mitogen-activated protein kinase (MAPK) inhibitor. Arthritis Res Ther. 2004;6:R384–R392. doi: 10.1186/ar1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeeberg BR, Feng W, Wang G, Wang MD, Fojo AT, Sunshine M, et al. GoMiner: a resource for biological interpretation of genomic and proteomic data. Genome Biol. 2003;4:R28. doi: 10.1186/gb-2003-4-4-r28. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
