Skip to main content
Erschienen in: Arthritis Research & Therapy 2/2007

Open Access 01.04.2007 | Research article

Peroxisome proliferator-activated receptor γ1 expression is diminished in human osteoarthritic cartilage and is downregulated by interleukin-1β in articular chondrocytes

verfasst von: Hassan Afif, Mohamed Benderdour, Leandra Mfuna-Endam, Johanne Martel-Pelletier, Jean-Pierre Pelletier, Nicholas Duval, Hassan Fahmi

Erschienen in: Arthritis Research & Therapy | Ausgabe 2/2007

Abstract

Peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear receptor involved in the regulation of many cellular processes. We and others have previously shown that PPARγ activators display anti-inflammatory and chondroprotective properties in vitro and improve the clinical course and histopathological features in an experimental animal model of osteoarthritis (OA). However, the expression and regulation of PPARγ expression in cartilage are poorly defined. This study was undertaken to investigate the quantitative expression and distribution of PPARγ in normal and OA cartilage and to evaluate the effect of IL-1β, a prominent cytokine in OA, on PPARγ expression in cultured chondrocytes. Immunohistochemical analysis revealed that the levels of PPARγ protein expression were significantly lower in OA cartilage than in normal cartilage. Using real-time RT-PCR, we demonstrated that PPARγ1 mRNA levels were about 10-fold higher than PPARγ2 mRNA levels, and that only PPARγ1 was differentially expressed: its levels in OA cartilage was 2.4-fold lower than in normal cartilage (p < 0.001). IL-1 treatment of OA chondrocytes downregulated PPARγ1 expression in a dose- and time-dependent manner. This effect probably occurred at the transcriptional level, because IL-1 decreases both PPARγ1 mRNA expression and PPARγ1 promoter activity. TNF-α, IL-17, and prostaglandin E2 (PGE2), which are involved in the pathogenesis of OA, also downregulated PPARγ1 expression. Specific inhibitors of the mitogen-activated protein kinases (MAPKs) p38 (SB203580) and c-Jun N-terminal kinase (SP600125), but not of extracellular signal-regulated kinase (PD98059), prevented IL-1-induced downregulation of PPARγ1 expression. Similarly, inhibitors of NF-κB signaling (pyrrolidine dithiocarbamate, MG-132, and SN-50) abolished the suppressive effect of IL-1. Thus, our study demonstrated that PPARγ1 is downregulated in OA cartilage. The pro-inflammatory cytokine IL-1 may be responsible for this downregulation via a mechanism involving activation of the MAPKs (p38 and JNK) and NF-κB signaling pathways. The IL-1-induced downregulation of PPARγ expression might be a new and additional important process by which IL-1 promotes articular inflammation and cartilage degradation.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​ar2151) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

HA conceived the study, designed and performed cell and real-time RT-PCR experiments and some immunohistochemistry experiments. MB participated in the study design and data analysis. LM-E performed some immunohistochemistry experiments. JM-P, J-PP, and ND helped to obtain tissues, participated in some immunohistochemistry studies and gave critical comments on the manuscripts. HF conceived, designed, and coordinated the study, performed some cell experiments, and drafted the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AP-1
= activator protein 1
COX
= cyclooxygenase
DMEM
= Dulbecco's modified Eagle's medium
ERK
– extracellular signal-regulated kinase
FCS
= fetal calf serum
GAPDH
= glyceraldehyde-3-phosphate dehydrogenase
IL
= interleukin
JNK
= c-Jun N-terminal kinase
MAPK
= mitogen-activated protein kinase
MMP
= metalloproteinase
mPGES
= membrane-associated prostaglandin E synthase
NF-κB
= nuclear factor-κB
OA
= osteoarthritis
PDTC
= pyrrolidine dithiocarbamate
PG
= prostaglandin
PGE2
= prostaglandin E2
PPAR
= peroxisome proliferator-activated receptor
RT-PCR
= reverse-transcriptase-mediated polymerase chain reaction
TNF
= tumor necrosis factor.

Introduction

Osteoarthritis (OA) is the most common joint disorder, accounting for a large proportion of disability in adults. It is characterized by the progressive destruction of articular cartilage, and excessive production of several pro-inflammatory mediators [13]. Among these mediators, IL-1β has been shown to be predominantly involved in the initiation and progression of the disease [13]. Exposure of chondrocytes to IL-1 induces a cascade of inflammatory and catabolic events including the upregulation of genes encoding matrix metalloproteinases (MMPs), aggrecanases, inducible nitric oxide synthase, cyclooxygenase-2 (COX-2), and microsomal prostaglandin E synthase-1 (mPGES-1) [14], leading to articular inflammation and destruction. Although the role of increased inflammatory and catabolic responses in OA is well documented, little is known about the endogenous signals and pathways that negatively regulate these events. Thus, identification and characterization of these pathways is of major importance in improving our understanding of the pathogenesis of OA and may be helpful in the development of new efficacious therapeutic strategies.
Peroxisome proliferator-activated receptors (PPARs) are a family of ligand-activated transcription factors belonging to the nuclear receptor superfamily [5]. So far, three PPAR subtypes have been identified: PPARα, PPARβ/δ, and PPARγ. PPARα is present mostly in the liver, heart, and muscle, where it is the target of the fibrate class of drugs and is believed to function in the catabolism of fatty acid [6]. PPARβ/δ is fairly ubiquitous and seems to be important in lipid and energy homeostasis [7]. PPARγ is the most studied form of PPAR. At least two PPARγ isoforms have been identified that are derived from the same gene by the use of alternative promoters and differential mRNA splicing [8, 9]. PPARγ1 is found in a broad range of tissues, whereas PPARγ2 is expressed mainly in adipose tissue [10].
Several lines of evidence suggest that PPARγ activation may have therapeutic benefits in OA and possibly other chronic articular diseases. We and others have shown that PPARγ is expressed and functionally active in chondrocytes and that PPARγ activators modulate the expression of several genes considered essential in the pathogenesis of OA. PPARγ activation inhibits the IL-1-induced expression of inducible nitric oxide synthase, MMP-13, COX-2, and mPGES-1 in chondrocytes [4, 11, 12]. Moreover, pretreatment with PPARγ activators prevents IL-1-induced proteoglycan degradation [13]. Additionally, PPARγ activation in synovial fibroblasts prevents the expression of IL-1, TNF-α, MMP-1, COX-2, and mPGES-1 [1416]. The inhibitory effect of PPARγ is partly due to antagonizing the transcriptional activity of the transcription factors NF-κB, activator protein 1 (AP-1), signal transducers and activators of transcription (STATs), and Egr-1 [16, 17]. The protective effect of PPARγ activators has also been demonstrated in several animal models of arthritis, including a guinea-pig model of OA [18]. In that study, pioglitazone, a PPARγ activator, reduced cartilage degradation as well as IL-1 and MMP-13 expression [18]. Together, these data indicate that PPARγ may constitute a new therapeutic target in treating OA.
Although a considerable amount is known on the effects of PPARγ activation on inflammatory and catabolic responses in articular tissues, little is known about PPARγ expression and regulation in these tissues. To improve our understanding of the biology of PPARγ in OA, we compared the expression of PPARγ in normal and OA cartilage. In addition, we investigated the effect of IL-1 on PPARγ expression in human OA chondrocytes.

Materials and methods

Reagents

Recombinant human IL-1β was obtained from Genzyme (Cambridge, MA, USA), and recombinant human TNF-α and recombinant human IL-17 were from R&D Systems (Minneapolis, MN, USA). Prostaglandin E2 (PGE2) was from Cayman Chemical Co. (Ann Arbor, MI, USA). SB203580, SP600125, PD98059, pyrrolidine dithiocarbamate (PDTC), MG-132 and SN-50 were from Calbiochem (La Jolla, CA, USA). DMEM, penicillin and streptomycin, FCS, and TRIzol® reagent were from Invitrogen (Burlington, ON, Canada). All other chemicals were purchased from either Bio-Rad (Mississauga, ON, Canada) or Sigma-Aldrich Canada (Oakville, ON, Canada).

Specimen selection and chondrocyte culture

Human normal cartilage (from femoral chondyles) was obtained at necropsy, within 12 hours of death, from donors with no history of arthritic diseases (n = 18, age 61 ± 15 years (mean ± SD)). To ensure that only normal tissue was used, cartilage specimens were thoroughly examined both macroscopically and microscopically. Only those with neither alteration were processed further. Human OA cartilage was obtained from patients undergoing total knee replacement (n = 41, age 64 ± 14 years (mean ± SD)). All patients with OA were diagnosed on criteria developed by the American College of Rheumatology Diagnostic Subcommittee for OA [19]. At the time of surgery, the patients had symptomatic disease requiring medical treatment in the form of non-steroidal anti-inflammatory drugs or selective COX-2 inhibitors. Patients who had received intra-articular injections of steroids were excluded. The Clinical Research Ethics Committee of Notre-Dame Hospital approved the study protocol and the use of human tissues.
Chondrocytes were released from cartilage by sequential enzymatic digestion as described previously [11]. In brief, this consisted of 2 mg/ml pronase for 1 hour followed by 1 mg/ml collagenase for 6 hours (type IV; Sigma-Aldrich) at 37°C in DMEM and antibiotics (100 U/ml penicillin, 100 μg/ml streptomycin). The digested tissue was briefly centrifuged and the pellet was washed. The isolated chondrocytes were seeded at high density in tissue culture flasks and cultured in DMEM supplemented with 10% heat-inactivated FCS. At confluence, the chondrocytes were detached, seeded at high density, and allowed to grow in DMEM supplemented as above. The culture medium was changed every second day, and 24 hours before the experiment the cells were incubated in fresh medium containing 0.5% FCS. Only first-passaged chondrocytes were used.

Immunohistochemistry

Cartilage specimens were processed for immunohistochemistry as described previously [4]. The specimens were fixed in 4% paraformaldehyde and embedded in paraffin. Sections (5 μm thick) of paraffin-embedded specimens were deparaffinized in toluene, then dehydrated in a graded ethanol series. The specimens were then preincubated with chondroitinase ABC (0.25 U/ml in PBS, pH 8.0) for 60 minutes at 37°C, followed by incubation with Triton X-100 (0.3%) for 30 minutes at 25°C. Slides were then washed in PBS followed by 2% hydrogen peroxide/methanol for 15 minutes. They were further incubated for 60 minutes with 2% normal serum (Vector Laboratories, Burlingame, CA, USA) and overlaid with primary antibody for 18 hours at 4°C in a humidified chamber. The antibody was a rabbit polyclonal anti-human PPARγ (Santa Cruz Biotechnology, Santa Cruz, CA, USA), used at 10 μg/ml. This antibody recognizes the epitope of the sequence mapping of amino acids 8 to 106 at the N terminus of PPARγ. Each slide was washed three times in PBS, pH 7.4, and stained with the use of the avidin-biotin complex method (Vectastain ABC kit; Vector Laboratories). The color was developed with 3,3'-diaminobenzidine (DAB) (Vector Laboratories) containing hydrogen peroxide. The slides were counterstained with eosin. The specificity of staining was evaluated by using antibody that had been preadsorbed (1 hour at 37°C) with a 20-fold molar excess of the protein fragment corresponding to amino acids 6 to 105 of human PPARγ (Santa Cruz), and by replacing the primary antibody with non-immune rabbit IgG (Chemicon, Temecula, CA, USA; used at the same concentration as the primary antibody). The evaluation of positive-staining chondrocytes was performed with our previously published method [4]. For each specimen, six microscopic fields were examined under ×40 magnification. The total number of chondrocytes and the number of positive-staining chondrocytes were evaluated and results were expressed as the percentage of chondrocytes that stained positive (cell score).

RNA extraction and reverse transcriptase-polymerase chain reaction

Total RNA from homogenized cartilage or stimulated chondrocytes was isolated by using TRIzol® reagent (Invitrogen) in accordance with the manufacturer's instructions. To remove contaminating DNA, isolated RNA was treated with RNase-free DNase I (Ambion, Austin, TX, USA). The RNA was quantified with the RiboGreen RNA quantitation kit (Molecular Probes, Eugene, OR, USA), dissolved in diethylpyrocarbonate-treated water and stored at -80°C until use. Total RNA (1 μg) was reverse-transcribed with Moloney murine leukemia virus reverse transcriptase (Fermentas, Burlington, ON, Canada) as detailed in the manufacturer's guidelines. One-fiftieth of the reverse transcriptase reaction was analyzed by real-time quantitative PCR as described below. The following primers were used: PPARγ1 sense, 5'-AAAGAAGCCAACACTAAACC-3'; PPARγ2 sense, 5'-GCGATTCCTTCACTGATAC-3'; common PPARγ1 and PPARγ2 antisense, 5'-CTTCCATTACGGAGAGATCC-3'; glyceraldehyde-3-phosphate dehydrogenase (GAPDH) sense, 5'-CAGAACATCATCCCTGCCTCT-3'; and GAPDH antisense, 5'-GCTTGACAAAGTGGTCGTTGAG-3'.

Real-time quantitative PCR

Quantitative PCR analysis was performed in a total volume of 50 μl containing template DNA, 200 nM sense and antisense primers, 25 μl of SYBR® Green master mix (Qiagen, Mississauga, ON, Canada) and uracil-N-glycosylase (UNG, 0.5 U; Epicentre Technologies, Madison, WI, USA). After incubation at 50°C for 2 minutes (UNG reaction), and at 95°C for 10 minutes (UNG inactivation and activation of the AmpliTaq Gold enzyme), the mixtures were subjected to 40 amplification cycles (15 s at 95°C for denaturation, and 1 minute for annealing and extension at 60°C). Incorporation of SYBR Green dye into PCR products was monitored in real time with a GeneAmp 5700 Sequence detection system (Applied Biosystems, Foster City, CA, USA) allowing determination of the threshold cycle (Ct) at which exponential amplification of PCR products begins. After PCR, dissociation curves were generated with one peak, indicating the specificity of the amplification. A threshold cycle (Ct value) was obtained from each amplification curve with the software provided by the manufacturer (Applied Biosystems).
Relative amounts of mRNA in normal and OA cartilage were determined with the use of the standard curve method. Serial dilutions of internal standards (plasmids containing cDNA of target genes) were included in each PCR run, and standard curves for the target gene and for GAPDH were generated by linear regression with a plot of log(Ct) against log(cDNA relative dilution). Ct values were then converted to the number of molecules. Relative mRNA expression in cultured chondrocytes was determined with the ΔΔCt method, as detailed in the manufacturer's guidelines (Applied Biosystems). A ΔCt value was first calculated by subtracting the Ct value for the housekeeping gene GAPDH from the Ct value for each sample. A ΔΔCt value was then calculated by subtracting the ΔCt value of the control (unstimulated cells) from the ΔCt value of each treatment. Fold changes compared with the control were then determined by raising 2 to the -ΔΔCt power. Each PCR reaction generated only the expected specific amplicon as shown by the melting-temperature profiles of the final product and by gel electrophoresis of test PCR reactions. Each PCR was performed in triplicate on two separate occasions for each independent experiment.

Plasmids and transient transfection

The luciferase reporter construct pGL3-PPARγ1p3000, containing a 3,000-base-pair fragment of the human PPARγ1 gene promoter, was kindly provided by Dr Johan Auwerx (Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France) [9]. β-Galactosidase reporter vector under the control of SV40 promoter (pSV40-β-galactosidase) was from Promega (Madison, WI, USA). Transient transfection experiments were performed with FuGene-6 (1 μg of DNA to 4 μl of FuGene 6; Roche Applied Science, Laval, QC, Canada) in accordance with the manufacturer's recommended protocol. In brief, chondrocytes were seeded and grown to 50 to 60% confluence. The cells were transfected with 1 μg of the reporter construct and 0.5 μg of the internal control pSV40-β-galactosidase. Six hours later, the medium was replaced with DMEM containing 1% FCS. The next day, the cells were treated for 18 hours with or without IL-1. After harvesting, luciferase activity was determined and normalized to β-galactosidase activity [16].

Western blot analysis

Chondrocytes were lysed in ice-cold lysis buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 2 mM EDTA, 1 mM PMSF, 10 μg/ml each of aprotinin, leupeptin, and pepstatin, 1% Nonidet P40, 1 mM Na3VO4, 1 mM NaF). Lysates were sonicated on ice and centrifuged at 12,000 r.p.m. for 15 minutes. The protein concentration of the supernatant was determined with the bicinchoninic acid method (Pierce, Rockford, IL, USA). Total cell lysate (20 μg) was subjected to SDS-PAGE and electrotransferred to a nitrocellulose membrane (Bio-Rad). After blocking in 20 mM Tris-HCl, pH 7.5, containing 150 mM NaCl, 0.1% Tween 20, and 5% (w/v) non-fat dry milk, blots were incubated overnight at 4°C with the primary antibody and washed with a Tris buffer (Tris-buffered saline, pH 7.5, containing 0.1% Tween 20). The blots were then incubated with horseradish peroxidase-conjugated secondary antibody (Pierce), washed again, incubated with SuperSignal Ultra Chemiluminescent reagent (Pierce), and, finally, exposed to Kodak X-Omat film (Eastman Kodak Ltd, Rochester, NY, USA).

Statistical analysis

Data are expressed as means ± SEM unless stated otherwise. Statistical significance was assessed by the two-tailed Student's t test; p < 0.05 was considered significant.

Results

Decreased expression of PPARγ1 in OA cartilage

To examine the expression and localization of PPAR-γ protein in cartilage, we performed an immunohistochemical analysis. We found that chondrocytes in both normal and OA cartilage express PPARγ protein. The immunostaining for PPARγ was essentially located in the superficial zones, and was lower in OA cartilage than in normal cartilage. Statistical evaluation of the cell score for PPARγ indicated significant differences between normal cartilage (22 ± 2.5% (mean ± SEM)) and cartilage from mild to moderate OA (11 ± 3%; Figure 1a,b). Similarly, PPARγ expression was significantly reduced in severe OA cartilage (10 ± 2%, data not shown). By contrast, in intact OA cartilage, the positive staining seemed lower, but the differences were not significant (data not shown). The specificity of the staining was confirmed by using antibodies that had been preadsorbed (1 hour, 37°C) with a 20-fold molar excess of the protein fragment corresponding to amino acids 6 to 105 of human PPARγ (Figure 1c) or non-immune serum (Figure 1c). PPARα and PPARβ were also expressed in normal, mild to moderate, and severe OA cartilage, but no significant differences were observed between the cartilage groups (Additional file 1).
PPARγ has two isoforms, PPARγ1 and PPARγ2, which are generated by alternative promoters and differential splicing [9]. To examine which PPARγ transcripts were expressed in cartilage, we determined absolute mRNA concentrations of PPARγ1 and PPARγ2 by quantitative real-time PCR. As shown in Figure 2, PPARγ1 abundance represents about 90% of the total PPARγ mRNA. Thus, human cartilage expresses high levels of γ1 mRNA, the isoform that is generally expressed in various tissues, and low levels of the γ2 isoform, which is more selectively expressed in adipose tissue [10]. The level of PPARγ1 expression in OA cartilage was 2.4-fold lower than in normal cartilage (p < 0.005). However, no significant differences in mRNA levels of PPARγ2 were seen between normal and OA cartilage (Figure 2). These observations demonstrate a selective downregulation of PPARγ1 in OA cartilage. In preliminary experiments we showed that the amplification efficiency of PPARγ1, PPARγ2, and GAPDH were approximately equal, ranging between 1.95 and 2.

Time-course and dose-dependent effect of IL-1 on PPARγ1 expression in chondrocytes

The reduced expression of PPARγ1 in OA cartilage suggests that humoral factors produced in the OA joint downregulate PPARγ1 expression. We therefore evaluated the effect of IL-1, one of the most prominent mediators in OA, on PPARγ1 expression in cultured chondrocytes. OA chondrocytes were treated with 100 pg/ml IL-1 for 0, 3, 6, 12, and 24 hours; the levels of PPARγ1 protein were then analyzed by Western blotting. In preliminary experiments we found that, as in cartilage, cultured chondrocytes express predominantly the PPARγ1 isoform but not the adipocyte-specific PPARγ2 isoform. As shown in Figure 3a, PPARγ1 protein expression was not significantly affected after 3 hours of stimulation with IL-1. The level of PPARγ1 protein then started to decline gradually at 6 hours and remained low until at least 24 hours. Subsequently, we examined the effect of various concentrations of IL-1 on PPARγ1 protein expression. As shown in Figure 3b, the expression of PPARγ1 was downregulated by IL-1 in a concentration-dependent manner; significant decreases were observed at a concentration as low as 10 pg/ml. Maximal decreases were obtained at an IL-1 concentration of 100 pg/ml (Figure 3b). No modulation of PPARα and PPARβ expression was seen (Additional file 2).
In addition to IL-1, the pro-inflammatory mediators TNF-α, IL-17, and PGE2 also contribute to the pathogenesis of OA [13]. We therefore examined their effects on PPARγ1 protein expression. Cultured OA chondrocytes were incubated for 24 hours with IL-1 (100 pg/ml), TNF-α (1 and 10 ng/ml), IL-17 (10 and 100 ng/ml), and PGE2 (0.1 and 1 μM), and the expression levels of PPARγ1 were determined by Western blotting. As shown in Figure 4, and like IL-1, TNF-α, IL-17, and PGE2 also downregulated PPARγ1 protein expression. Similar results were obtained with normal chondrocytes (n = 3; data not shown).

Downregulation by IL-1 of PPARγ1 expression at the transcriptional level

To elucidate the mechanism responsible for the changes in amounts of PPARγ1 protein, we measured the steady-state level of PPARγ1 mRNA by quantitative real-time PCR. Expression of the gene encoding GAPDH was used for normalization. The relative expression level of PPARγ1 mRNA was plotted as a percentage decrease compared with untreated control cells (Figure 5a). Consistent with its effects on protein expression (Figure 3b), IL-1 downregulates PPARγ1 mRNA expression in a dose-dependent manner in OA chondrocytes. The effect of IL-1 on PPARγ1 mRNA expression was maximal (about 85% decrease) at 100 pg/ml. A dose-dependent effect of IL-1 on PPARγ1 mRNA expression was also observed in normal chondrocytes (n = 3; data not shown).
To characterize the effect of IL-1 on PPARγ1 expression further, we performed transient transfection experiments with the reporter construct pGL3-PPARγ1p3000, containing about 3,000 base pairs of regulatory sequence of the gene encoding human PPARγ1 [9]. As shown in Figure 5b, IL-1 suppressed PPARγ1 promoter activity in a dose-dependent manner. The effect of IL-1 on PPARγ1 promoter activity was optimal at 100 pg/ml (about 65% decrease). Taken together, these data strongly suggest that IL-1 suppressed PPARγ1 expression at the transcriptional level.

The MAPKs JNK and p38, but not ERK, are involved in IL-1-induced downregulation of PPARγ1

IL-1 is known to induce its effects in chondrocytes through activation of a plethora of signaling pathways, including the mitogen-activated protein kinases (MAPKs) c-Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK) [20]. To assess the contribution of these pathways in the IL-1-mediated downregulation of PPARγ1, OA chondrocytes were pretreated for 30 minutes with selective inhibitors for the above pathways, and then stimulated or not with IL-1 for 18 hours. Total cell lysates were analyzed for PPARγ1 protein expression by Western blotting. As shown in Figure 6a, IL-1 reduced PPARγ1 expression remarkably, confirming the results seen previously (Figure 3). Pretreatment with SB203580, a specific p38 MAPK inhibitor, as well as pretreatment with SP600125, a selective inhibitor of JNK, dose-dependently abolished IL-1-induced downregulation of PPARγ1 expression. Conversely, PD98059, a selective inhibitor of ERK, had no effect on IL-1-induced downregulation of PPARγ expression, even at a high concentration (20 μM). None of the MAPK inhibitors had an effect on PPARγ expression in the absence of IL-1. These results suggest that the MAPKs JNK and p38, but not ERK, are involved in the suppression of PPARγ1 expression by IL-1.

Mediation of IL-1-induced downregulation of PPARγ1 by NF-κB

Because NF-κB mediates many of the effects of IL-1 in a variety of cell types including chondrocytes, we examined the role of this transcription factor in the repression of PPARγ1. We used three different pharmacological inhibitors of the NF-κB pathway: the antioxidant PDTC, a proteasome inhibitor MG-132, and an inhibitor of NF-κB translocation (SN-50). Cells were pretreated with increasing concentrations of each inhibitor for 30 minutes and then subsequently treated with 100 pg of IL-1 for 18 hours.
As shown in Figure 6b, treatment with IL-1 decreased PPARγ1 expression, but this IL-1 effect was dose-dependently abolished in the presence of each of the three NF-κB inhibitors (PDTC, MG-132, and SN-50). None of the NF-κB inhibitors had an effect on basal PPARγ1 expression. These results imply that NF-κB activation participates in the IL-1-mediated downregulation of PPARγ1 expression.

Discussion

There is considerable evidence for the importance of PPARγ in OA because of its potential beneficial effects. It is expressed by all major cells in joints, including chondrocytes [11, 13]. Natural and synthetic ligands of PPARγ were shown to inhibit the expression of several inflammatory and catabolic genes in cultured chondrocytes [4, 11, 12] and to exhibit anti-inflammatory and chondroprotective effects in an experimental animal model of OA [18]. However, little is known about the expression and regulation of PPARγ expression in cartilage. Here, we analyzed the expression of PPARγ in OA and normal cartilage, and studied the effect of IL-1, a prominent cytokine in OA, on PPARγ expression in cultured chondrocytes.
This is the first study to demonstrate that human cartilage expresses predominantly PPARγ1 mRNA and that the levels of PPARγ1 are decreased in OA in comparison with normal cartilage. Our immunohistochemistry analysis showed that PPARγ was located essentially in the superficial zone of cartilage and that the levels of PPARγ expression in OA cartilage were lower than in normal cartilage.
Altered expression of PPARγ was observed in several other inflammatory disorders. For instance, PPARγ expression was shown to be reduced in atherosclerotic tissues [21], in epithelial cells from patients with ulcerative colitis [22], in peripheral blood mononuclear cells from patients with multiple sclerosis [23], in alveolar macrophages from patients with allergic asthma [24], and in nasal polyposis from patients with allergic rhinitis [25]. In contrast, PPARγ expression was shown to be elevated in brains of patients with Alzheimer's disease [26], in bronchial epithelium and airway smooth muscle cells of asthmatic patients [27], and in T cells isolated from patients with sepsis [28]. Taken together, these results suggest that tissue-specific regulation of PPARγ expression is extremely complex.
To determine which factors might downregulate PPARγ expression in cartilage, we tested the impact of IL-1, which accumulates in chondrocytes in the superficial zone of OA cartilage [29, 30] and has a pivotal role in the initiation and progression of OA [13]. Our results revealed that exposure to IL-1 downregulates PPARγ protein expression in chondrocytes in a time- and dose-dependent manner. It should be noted that TNF-α, IL-17, and PGE2, which are known to contribute to the pathogenesis of OA, also downregulate PPARγ gene expression. We therefore cannot exclude the possibility of a role for these inflammatory mediators in PPARγ downregulation in cartilage in vivo. Given the anti-inflammatory and anti-catabolic functions of PPARγ, it is reasonable to speculate that the suppression of PPARγ expression by inflammatory mediators in chondrocytes presents a new and additional mechanism by which these mediators contribute to the pathogenesis of OA. Our findings are consistent with other studies showing that pro-inflammatory stimuli downregulate PPARγ expression in chondrocytes [3133] and synovial fibroblasts [34, 35]. In contrast, Shan and colleagues [36] found that IL-1 upregulates PPARγ expression in chondrocytes. The reasons for these discrepancies are not clear and could be due to small differences in chondrocyte preparation, culture conditions, and/or detection methods.
Suppression of PPARγ1 expression by IL-1 in chondrocytes probably occurs at the transcriptional level, because reporter gene assays revealed a decrease in PPARγ1 promoter activity by IL-1. As an alternative to an effect on PPARγ1 promoter, we could not exclude a specific effect of IL-1 on the stability of PPARγ1 mRNA.
The MAPKs JNK, p38, and ERK are activated by IL-1 and mediate many of the effects of IL-1 in chondrocytes [20]. To determine whether these MAPKs are involved in the IL-1-mediated downregulation of PPARγ1 expression, we employed specific inhibitors of the three MAPKs. We found that SB203580 and SP600125 – specific inhibitors of the MAPKs p38 and JNK, respectively – almost completely abolished the IL-1-mediated downregulation of PPARγ1 expression, whereas PD98059 – an inhibitor of the MAPK ERK- was without effect. These data suggest that the MAPKs JNK and p38, but not ERK, mediate IL-1-induced downregulation of PPARγ1 expression in chondrocytes. The NF-κB pathway also mediates many effects of IL-1 in chondrocytes [3741]. We demonstrate here that three compounds that interfere with NF-κB activation, the anti-oxidant PDTC, the proteasome inhibitor MG-132, and an inhibitor of NF-κB translocation SN-50, blocked the suppressive effect of IL-1, suggesting the involvement of NF-κB in the IL-1-mediated downregulation of PPARγ1 in chondrocytes. Thus, IL-1 engages both the MAPK (JNK and p38) and the NF-κB pathways to suppress PPARγ1 expression, although it is not clear whether these pathways act on the same axis or in parallel. Downstream nuclear events in JNK, p38, and NF-κB signaling pathways leading to the regulation of gene expression in chondrocytes include the activation of the transcription factors AP-1 and NF-κB [20, 37, 38, 4043]. The human PPARγ1 promoter contains binding sites for both AP-1 and NF-κB [9]. It is therefore possible that AP-1 and NF-κB mediate IL-1-induced downregulation of PPARγ1 expression. Although they are historically characterized as transcriptional activators, several reports have recently defined AP-1 and NF-κB as transcriptional repressors [4450]. Analysis of PPARγ1 promoter in a promoter reporter construct, with mutation of the AP-1 and NF-κB response elements and the use of small interfering RNA technology, will contribute to our understanding of the importance of AP-1 and NF-κB in the IL-1-induced downregulation of PPARγ1 expression.
The physiological significance of reduced expression of PPARγ in OA cartilage is of considerable interest, given the protective functions of PPARγ in cartilage. Indeed, we and others have previously reported that PPARγ activators inhibit several inflammatory and catabolic events involved in the pathogenesis of OA [4, 11, 12, 3234]. PPARγ activation was also shown to prevent the proteoglycan degradation induced by pro-inflammatory cytokines [13]. Furthermore, PPARγ ligands were shown to reduce the incidence and severity of OA in an experimental model, preventing inflammatory and catabolic responses as well as cartilage degradation [18]. All these data suggest that PPARγ has a protective role in OA. This is strengthened by the observation that PPARγ haploinsufficiency exacerbates experimentally induced arthritis [51]. It is therefore tempting to speculate that diminished expression of PPARγ in OA cartilage may, at least in part, be involved in increased expression of inflammatory and catabolic genes, promoting articular inflammation and cartilage degradation. In addition, the observation that IL-1 and other pro-inflammatory mediators downregulate PPARγ1 expression in chondrocytes has important implications for our understanding of the pathophysiology of OA.

Conclusion

The decreased expression of PPARγ in OA cartilage and the literature supporting a protective role for PPARγ in OA raise the possibility that upregulation of PPARγ may be beneficial in the context of preventing and treating OA. Additional studies to define the molecular mechanisms controlling the expression of PPARγ are therefore urgently needed. Such research will no doubt add to our understanding of the pathogenesis of OA, and could lead to the development of new therapeutic strategies in the prevention and treatment of OA and possibly other arthritic diseases.

Acknowledgements

The authors thank J Auwerx for the PPARg1 promoter, and M Boily for help and critical comments. This work was supported by the Canadian Institutes of Health Research (CIHR) Grant IMH-63168, and the Fonds de la Recherche du Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CHUM). HF is a Research Scholar of the Fonds de Recherche en Santé du Québec (FRSQ).

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

HA conceived the study, designed and performed cell and real-time RT-PCR experiments and some immunohistochemistry experiments. MB participated in the study design and data analysis. LM-E performed some immunohistochemistry experiments. JM-P, J-PP, and ND helped to obtain tissues, participated in some immunohistochemistry studies and gave critical comments on the manuscripts. HF conceived, designed, and coordinated the study, performed some cell experiments, and drafted the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Goldring MB: The role of cytokines as inflammatory mediators in osteoarthritis: lessons from animal models. Connect Tissue Res. 1999, 40: 1-11.CrossRefPubMed Goldring MB: The role of cytokines as inflammatory mediators in osteoarthritis: lessons from animal models. Connect Tissue Res. 1999, 40: 1-11.CrossRefPubMed
2.
Zurück zum Zitat Pelletier JP, Martel-Pelletier J, Abramson SB: Osteoarthritis, an inflammatory disease: potential implication for the selection of new therapeutic targets. Arthritis Rheum. 2001, 44: 1237-1247. 10.1002/1529-0131(200106)44:6<1237::AID-ART214>3.0.CO;2-F.CrossRefPubMed Pelletier JP, Martel-Pelletier J, Abramson SB: Osteoarthritis, an inflammatory disease: potential implication for the selection of new therapeutic targets. Arthritis Rheum. 2001, 44: 1237-1247. 10.1002/1529-0131(200106)44:6<1237::AID-ART214>3.0.CO;2-F.CrossRefPubMed
3.
Zurück zum Zitat Goldring MB, Berenbaum F: The regulation of chondrocyte function by proinflammatory mediators: prostaglandins and nitric oxide. Clin Orthop Relat Res. 2004, S37-S46. 10.1097/01.blo.0000144484.69656.e4. 427 Suppl Goldring MB, Berenbaum F: The regulation of chondrocyte function by proinflammatory mediators: prostaglandins and nitric oxide. Clin Orthop Relat Res. 2004, S37-S46. 10.1097/01.blo.0000144484.69656.e4. 427 Suppl
4.
Zurück zum Zitat Li X, Afif H, Cheng S, Martel-Pelletier J, Pelletier JP, Ranger P, Fahmi H: Expression and regulation of microsomal prostaglandin E synthase-1 in human osteoarthritic cartilage and chondrocytes. J Rheumatol. 2005, 32: 887-895.PubMed Li X, Afif H, Cheng S, Martel-Pelletier J, Pelletier JP, Ranger P, Fahmi H: Expression and regulation of microsomal prostaglandin E synthase-1 in human osteoarthritic cartilage and chondrocytes. J Rheumatol. 2005, 32: 887-895.PubMed
5.
Zurück zum Zitat Fahmi H, Pelletier JP, Martel-Pelletier J: PPARγ ligands as modulators of inflammatory and catabolic responses on arthritis. An overview. J Rheumatol. 2002, 29: 3-14.PubMed Fahmi H, Pelletier JP, Martel-Pelletier J: PPARγ ligands as modulators of inflammatory and catabolic responses on arthritis. An overview. J Rheumatol. 2002, 29: 3-14.PubMed
6.
Zurück zum Zitat Braissant O, Foufelle F, Scotto C, Dauca M, Wahli W: Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-α, -β, and -γ in the adult rat. Endocrinology. 1996, 137: 354-366. 10.1210/en.137.1.354.PubMed Braissant O, Foufelle F, Scotto C, Dauca M, Wahli W: Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-α, -β, and -γ in the adult rat. Endocrinology. 1996, 137: 354-366. 10.1210/en.137.1.354.PubMed
7.
8.
Zurück zum Zitat Zhu Y, Qi C, Korenberg JR, Chen XN, Noya D, Rao MS, Reddy JK: Structural organization of mouse peroxisome proliferator-activated receptor γ (mPPAR γ) gene: alternative promoter use and different splicing yield two mPPAR γ isoforms. Proc Natl Acad Sci USA. 1995, 92: 7921-7925. 10.1073/pnas.92.17.7921.PubMedCentralCrossRefPubMed Zhu Y, Qi C, Korenberg JR, Chen XN, Noya D, Rao MS, Reddy JK: Structural organization of mouse peroxisome proliferator-activated receptor γ (mPPAR γ) gene: alternative promoter use and different splicing yield two mPPAR γ isoforms. Proc Natl Acad Sci USA. 1995, 92: 7921-7925. 10.1073/pnas.92.17.7921.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, Saladin R, Najib J, Laville M, Fruchart JC, Deeb S, et al: The organization, promoter analysis, and expression of the human PPARγ gene. J Biol Chem. 1997, 272: 18779-18789. 10.1074/jbc.272.30.18779.CrossRefPubMed Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, Saladin R, Najib J, Laville M, Fruchart JC, Deeb S, et al: The organization, promoter analysis, and expression of the human PPARγ gene. J Biol Chem. 1997, 272: 18779-18789. 10.1074/jbc.272.30.18779.CrossRefPubMed
10.
Zurück zum Zitat Vidal-Puig A, Jimenez-Linan M, Lowell BB, Hamann A, Hu E, Spiegelman B, Flier JS, Moller DE: Regulation of PPAR γ gene expression by nutrition and obesity in rodents. J Clin Invest. 1996, 97: 2553-2561.PubMedCentralCrossRefPubMed Vidal-Puig A, Jimenez-Linan M, Lowell BB, Hamann A, Hu E, Spiegelman B, Flier JS, Moller DE: Regulation of PPAR γ gene expression by nutrition and obesity in rodents. J Clin Invest. 1996, 97: 2553-2561.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Fahmi H, Di Battista JA, Pelletier JP, Mineau F, Ranger P, Martel-Pelletier J: Peroxisome proliferator-activated receptor γ activators inhibit interleukin-1β-induced nitric oxide and matrix metalloproteinase 13 production in human chondrocytes. Arthritis Rheum. 2001, 44: 595-607. 10.1002/1529-0131(200103)44:3<595::AID-ANR108>3.0.CO;2-8.CrossRefPubMed Fahmi H, Di Battista JA, Pelletier JP, Mineau F, Ranger P, Martel-Pelletier J: Peroxisome proliferator-activated receptor γ activators inhibit interleukin-1β-induced nitric oxide and matrix metalloproteinase 13 production in human chondrocytes. Arthritis Rheum. 2001, 44: 595-607. 10.1002/1529-0131(200103)44:3<595::AID-ANR108>3.0.CO;2-8.CrossRefPubMed
12.
Zurück zum Zitat Fahmi H, Pelletier JP, Mineau F, Martel-Pelletier J: 15d-PGJ2 is acting as a 'dual agent' on the regulation of COX-2 expression in human osteoarthritic chondrocytes. Osteoarthritis Cartilage. 2002, 10: 845-848. 10.1053/joca.2002.0835.CrossRefPubMed Fahmi H, Pelletier JP, Mineau F, Martel-Pelletier J: 15d-PGJ2 is acting as a 'dual agent' on the regulation of COX-2 expression in human osteoarthritic chondrocytes. Osteoarthritis Cartilage. 2002, 10: 845-848. 10.1053/joca.2002.0835.CrossRefPubMed
13.
Zurück zum Zitat Bordji K, Grillasca JP, Gouze JN, Magdalou J, Schohn H, Keller JM, Bianchi A, Dauca M, Netter P, Terlain B: Evidence for the presence of peroxisome proliferator-activated receptor (PPAR) α and γ and retinoid Z receptor in cartilage. PPARγ activation modulates the effects of interleukin-1β on rat chondrocytes. J Biol Chem. 2000, 275: 12243-12250. 10.1074/jbc.275.16.12243.CrossRefPubMed Bordji K, Grillasca JP, Gouze JN, Magdalou J, Schohn H, Keller JM, Bianchi A, Dauca M, Netter P, Terlain B: Evidence for the presence of peroxisome proliferator-activated receptor (PPAR) α and γ and retinoid Z receptor in cartilage. PPARγ activation modulates the effects of interleukin-1β on rat chondrocytes. J Biol Chem. 2000, 275: 12243-12250. 10.1074/jbc.275.16.12243.CrossRefPubMed
14.
Zurück zum Zitat Ji JD, Cheon H, Jun JB, Choi SJ, Kim YR, Lee YH, Kim TH, Chae IJ, Song GG, Yoo DH, et al: Effects of peroxisome proliferator-activated receptor-γ (PPAR-γ) on the expression of inflammatory cytokines and apoptosis induction in rheumatoid synovial fibroblasts and monocytes. J Autoimmun. 2001, 17: 215-221. 10.1006/jaut.2001.0542.CrossRefPubMed Ji JD, Cheon H, Jun JB, Choi SJ, Kim YR, Lee YH, Kim TH, Chae IJ, Song GG, Yoo DH, et al: Effects of peroxisome proliferator-activated receptor-γ (PPAR-γ) on the expression of inflammatory cytokines and apoptosis induction in rheumatoid synovial fibroblasts and monocytes. J Autoimmun. 2001, 17: 215-221. 10.1006/jaut.2001.0542.CrossRefPubMed
15.
Zurück zum Zitat Fahmi H, Pelletier JP, Di Battista JA, Cheung HS, Fernandes J, Martel-Pelletier J: Peroxisome proliferator-activated receptor γ activators inhibit MMP-1 production in human synovial fibroblasts by reducing the activity of the activator protein 1. Osteoarthritis Cartilage. 2002, 10: 100-108. 10.1053/joca.2001.0485.CrossRefPubMed Fahmi H, Pelletier JP, Di Battista JA, Cheung HS, Fernandes J, Martel-Pelletier J: Peroxisome proliferator-activated receptor γ activators inhibit MMP-1 production in human synovial fibroblasts by reducing the activity of the activator protein 1. Osteoarthritis Cartilage. 2002, 10: 100-108. 10.1053/joca.2001.0485.CrossRefPubMed
16.
Zurück zum Zitat Cheng S, Afif H, Martel-Pelletier J, Pelletier JP, Li X, Farrajota K, Lavigne M, Fahmi H: Activation of peroxisome proliferator-activated receptor γ inhibits interleukin-1β-induced membrane-associated prostaglandin E2 synthase-1 expression in human synovial fibroblasts by interfering with Egr-1. J Biol Chem. 2004, 279: 22057-22065. 10.1074/jbc.M402828200.CrossRefPubMed Cheng S, Afif H, Martel-Pelletier J, Pelletier JP, Li X, Farrajota K, Lavigne M, Fahmi H: Activation of peroxisome proliferator-activated receptor γ inhibits interleukin-1β-induced membrane-associated prostaglandin E2 synthase-1 expression in human synovial fibroblasts by interfering with Egr-1. J Biol Chem. 2004, 279: 22057-22065. 10.1074/jbc.M402828200.CrossRefPubMed
17.
Zurück zum Zitat Jiang C, Ting AT, Seed B: PPAR-γ agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998, 391: 82-86. 10.1038/35154.CrossRefPubMed Jiang C, Ting AT, Seed B: PPAR-γ agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998, 391: 82-86. 10.1038/35154.CrossRefPubMed
18.
Zurück zum Zitat Kobayashi T, Notoya K, Naito T, Unno S, Nakamura A, Martel-Pelletier J, Pelletier JP: Pioglitazone, a peroxisome proliferator-activated receptor γ agonist, reduces the progression of experimental osteoarthritis in guinea pigs. Arthritis Rheum. 2005, 52: 479-487. 10.1002/art.20792.CrossRefPubMed Kobayashi T, Notoya K, Naito T, Unno S, Nakamura A, Martel-Pelletier J, Pelletier JP: Pioglitazone, a peroxisome proliferator-activated receptor γ agonist, reduces the progression of experimental osteoarthritis in guinea pigs. Arthritis Rheum. 2005, 52: 479-487. 10.1002/art.20792.CrossRefPubMed
19.
Zurück zum Zitat Altman R, Asch E, Bloch D, Bole G, Borenstein D, Brandt K, Christy W, Cooke TD, Greenwald R, Hochberg M, et al: Development of criteria for the classification and reporting of osteoarthritis. Classification of osteoarthritis of the knee. Diagnostic and Therapeutic Criteria Committee of the American Rheumatism Association. Arthritis Rheum. 1986, 29: 1039-1049. 10.1002/art.1780290816.CrossRefPubMed Altman R, Asch E, Bloch D, Bole G, Borenstein D, Brandt K, Christy W, Cooke TD, Greenwald R, Hochberg M, et al: Development of criteria for the classification and reporting of osteoarthritis. Classification of osteoarthritis of the knee. Diagnostic and Therapeutic Criteria Committee of the American Rheumatism Association. Arthritis Rheum. 1986, 29: 1039-1049. 10.1002/art.1780290816.CrossRefPubMed
20.
Zurück zum Zitat Geng Y, Valbracht J, Lotz M: Selective activation of the mitogen-activated protein kinase subgroups c-Jun NH2 terminal kinase and p38 by IL-1 and TNF in human articular chondrocytes. J Clin Invest. 1996, 98: 2425-2430.PubMedCentralCrossRefPubMed Geng Y, Valbracht J, Lotz M: Selective activation of the mitogen-activated protein kinase subgroups c-Jun NH2 terminal kinase and p38 by IL-1 and TNF in human articular chondrocytes. J Clin Invest. 1996, 98: 2425-2430.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Soumian S, Gibbs R, Davies A, Albrecht C: mRNA expression of genes involved in lipid efflux and matrix degradation in occlusive and ectatic atherosclerotic disease. J Clin Pathol. 2005, 58: 1255-1260. 10.1136/jcp.2005.026161.PubMedCentralCrossRefPubMed Soumian S, Gibbs R, Davies A, Albrecht C: mRNA expression of genes involved in lipid efflux and matrix degradation in occlusive and ectatic atherosclerotic disease. J Clin Pathol. 2005, 58: 1255-1260. 10.1136/jcp.2005.026161.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Dubuquoy L, Jansson EA, Deeb S, Rakotobe S, Karoui M, Colombel JF, Auwerx J, Pettersson S, Desreumaux P: Impaired expression of peroxisome proliferator-activated receptor gamma in ulcerative colitis. Gastroenterology. 2003, 124: 1265-1276. 10.1016/S0016-5085(03)00271-3.CrossRefPubMed Dubuquoy L, Jansson EA, Deeb S, Rakotobe S, Karoui M, Colombel JF, Auwerx J, Pettersson S, Desreumaux P: Impaired expression of peroxisome proliferator-activated receptor gamma in ulcerative colitis. Gastroenterology. 2003, 124: 1265-1276. 10.1016/S0016-5085(03)00271-3.CrossRefPubMed
23.
Zurück zum Zitat Klotz L, Schmidt M, Giese T, Sastre M, Knolle P, Klockgether T, Heneka MT: Proinflammatory stimulation and pioglitazone treatment regulate peroxisome proliferator-activated receptor γ levels in peripheral blood mononuclear cells from healthy controls and multiple sclerosis patients. J Immunol. 2005, 175: 4948-4955.CrossRefPubMed Klotz L, Schmidt M, Giese T, Sastre M, Knolle P, Klockgether T, Heneka MT: Proinflammatory stimulation and pioglitazone treatment regulate peroxisome proliferator-activated receptor γ levels in peripheral blood mononuclear cells from healthy controls and multiple sclerosis patients. J Immunol. 2005, 175: 4948-4955.CrossRefPubMed
24.
Zurück zum Zitat Kobayashi M, Thomassen MJ, Rambasek T, Bonfield TL, Raychaudhuri B, Malur A, Winkler AR, Barna BP, Goldman SJ, Kavuru MS: An inverse relationship between peroxisome proliferator-activated receptor γ and allergic airway inflammation in an allergen challenge model. Ann Allergy Asthma Immunol. 2005, 95: 468-473.CrossRefPubMed Kobayashi M, Thomassen MJ, Rambasek T, Bonfield TL, Raychaudhuri B, Malur A, Winkler AR, Barna BP, Goldman SJ, Kavuru MS: An inverse relationship between peroxisome proliferator-activated receptor γ and allergic airway inflammation in an allergen challenge model. Ann Allergy Asthma Immunol. 2005, 95: 468-473.CrossRefPubMed
25.
Zurück zum Zitat Cardell LO, Hagge M, Uddman R, Adner M: Downregulation of peroxisome proliferator-activated receptors (PPARs) in nasal polyposis. Respir Res. 2005, 6: 132-10.1186/1465-9921-6-132.PubMedCentralCrossRefPubMed Cardell LO, Hagge M, Uddman R, Adner M: Downregulation of peroxisome proliferator-activated receptors (PPARs) in nasal polyposis. Respir Res. 2005, 6: 132-10.1186/1465-9921-6-132.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Kitamura Y, Shimohama S, Koike H, Kakimura J, Matsuoka Y, Nomura Y, Gebicke-Haerter PJ, Taniguchi T: Increased expression of cyclooxygenases and peroxisome proliferator-activated receptor-γ in Alzheimer's disease brains. Biochem Biophys Res Commun. 1999, 254: 582-586. 10.1006/bbrc.1998.9981.CrossRefPubMed Kitamura Y, Shimohama S, Koike H, Kakimura J, Matsuoka Y, Nomura Y, Gebicke-Haerter PJ, Taniguchi T: Increased expression of cyclooxygenases and peroxisome proliferator-activated receptor-γ in Alzheimer's disease brains. Biochem Biophys Res Commun. 1999, 254: 582-586. 10.1006/bbrc.1998.9981.CrossRefPubMed
27.
Zurück zum Zitat Benayoun L, Letuve S, Druilhe A, Boczkowski J, Dombret MC, Mechighel P, Megret J, Leseche G, Aubier M, Pretolani M: Regulation of peroxisome proliferator-activated receptor γ expression in human asthmatic airways: relationship with proliferation, apoptosis, and airway remodeling. Am J Respir Crit Care Med. 2001, 164: 1487-1494.CrossRefPubMed Benayoun L, Letuve S, Druilhe A, Boczkowski J, Dombret MC, Mechighel P, Megret J, Leseche G, Aubier M, Pretolani M: Regulation of peroxisome proliferator-activated receptor γ expression in human asthmatic airways: relationship with proliferation, apoptosis, and airway remodeling. Am J Respir Crit Care Med. 2001, 164: 1487-1494.CrossRefPubMed
28.
Zurück zum Zitat Soller M, Tautenhahn A, Brune B, Zacharowski K, John S, Link H, von Knethen A: Peroxisome proliferator-activated receptor γ contributes to T lymphocyte apoptosis during sepsis. J Leukoc Biol. 2006, 79: 235-243. 10.1189/jlb.0205058.CrossRefPubMed Soller M, Tautenhahn A, Brune B, Zacharowski K, John S, Link H, von Knethen A: Peroxisome proliferator-activated receptor γ contributes to T lymphocyte apoptosis during sepsis. J Leukoc Biol. 2006, 79: 235-243. 10.1189/jlb.0205058.CrossRefPubMed
29.
Zurück zum Zitat Tetlow LC, Adlam DJ, Woolley DE: Matrix metalloproteinase and proinflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: associations with degenerative changes. Arthritis Rheum. 2001, 44: 585-594. 10.1002/1529-0131(200103)44:3<585::AID-ANR107>3.0.CO;2-C.CrossRefPubMed Tetlow LC, Adlam DJ, Woolley DE: Matrix metalloproteinase and proinflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: associations with degenerative changes. Arthritis Rheum. 2001, 44: 585-594. 10.1002/1529-0131(200103)44:3<585::AID-ANR107>3.0.CO;2-C.CrossRefPubMed
30.
Zurück zum Zitat Towle CA, Hung HH, Bonassar LJ, Treadwell BV, Mangham DC: Detection of interleukin-1 in the cartilage of patients with osteoarthritis: a possible autocrine/paracrine role in pathogenesis. Osteoarthritis Cartilage. 1997, 5: 293-300. 10.1016/S1063-4584(97)80008-8.CrossRefPubMed Towle CA, Hung HH, Bonassar LJ, Treadwell BV, Mangham DC: Detection of interleukin-1 in the cartilage of patients with osteoarthritis: a possible autocrine/paracrine role in pathogenesis. Osteoarthritis Cartilage. 1997, 5: 293-300. 10.1016/S1063-4584(97)80008-8.CrossRefPubMed
31.
Zurück zum Zitat Boyault S, Simonin MA, Bianchi A, Compe E, Liagre B, Mainard D, Becuwe P, Dauca M, Netter P, Terlain B, Bordji K: 15-Deoxy-δ 12,14-PGJ2, but not troglitazone, modulates IL-1β effects in human chondrocytes by inhibiting NF-κB and AP-1 activation pathways. FEBS Lett. 2001, 501: 24-30. 10.1016/S0014-5793(01)02614-X.CrossRefPubMed Boyault S, Simonin MA, Bianchi A, Compe E, Liagre B, Mainard D, Becuwe P, Dauca M, Netter P, Terlain B, Bordji K: 15-Deoxy-δ 12,14-PGJ2, but not troglitazone, modulates IL-1β effects in human chondrocytes by inhibiting NF-κB and AP-1 activation pathways. FEBS Lett. 2001, 501: 24-30. 10.1016/S0014-5793(01)02614-X.CrossRefPubMed
32.
Zurück zum Zitat Francois M, Richette P, Tsagris L, Raymondjean M, Fulchignoni-Lataud MC, Forest C, Savouret JF, Corvol MT: Peroxisome proliferator-activated receptor-γ down-regulates chondrocyte matrix metalloproteinase-1 via a novel composite element. J Biol Chem. 2004, 279: 28411-28418. 10.1074/jbc.M312708200.CrossRefPubMed Francois M, Richette P, Tsagris L, Raymondjean M, Fulchignoni-Lataud MC, Forest C, Savouret JF, Corvol MT: Peroxisome proliferator-activated receptor-γ down-regulates chondrocyte matrix metalloproteinase-1 via a novel composite element. J Biol Chem. 2004, 279: 28411-28418. 10.1074/jbc.M312708200.CrossRefPubMed
33.
Zurück zum Zitat Poleni PE, Bianchi A, Etienne S, Koufany M, Sebillaud S, Netter P, Terlain B, Jouzeau JY: Agonists of peroxisome proliferators-activated receptors (PPAR) α, β/δ or γ reduce transforming growth factor (TGF)-β-induced proteoglycans' production in chondrocytes. Osteoarthritis Cartilage. 2006 Poleni PE, Bianchi A, Etienne S, Koufany M, Sebillaud S, Netter P, Terlain B, Jouzeau JY: Agonists of peroxisome proliferators-activated receptors (PPAR) α, β/δ or γ reduce transforming growth factor (TGF)-β-induced proteoglycans' production in chondrocytes. Osteoarthritis Cartilage. 2006
34.
Zurück zum Zitat Simonin MA, Bordji K, Boyault S, Bianchi A, Gouze E, Becuwe P, Dauca M, Netter P, Terlain B: PPAR-γ ligands modulate effects of LPS in stimulated rat synovial fibroblasts. Am J Physiol Cell Physiol. 2002, 282: C125-C133.PubMed Simonin MA, Bordji K, Boyault S, Bianchi A, Gouze E, Becuwe P, Dauca M, Netter P, Terlain B: PPAR-γ ligands modulate effects of LPS in stimulated rat synovial fibroblasts. Am J Physiol Cell Physiol. 2002, 282: C125-C133.PubMed
35.
Zurück zum Zitat Moulin D, Bianchi A, Boyault S, Sebillaud S, Koufany M, Francois M, Netter P, Jouzeau JY, Terlain B: Rosiglitazone induces interleukin-1 receptor antagonist in interleukin-1β-stimulated rat synovial fibroblasts via a peroxisome proliferator-activated receptor β/δ-dependent mechanism. Arthritis Rheum. 2005, 52: 759-769. 10.1002/art.20868.CrossRefPubMed Moulin D, Bianchi A, Boyault S, Sebillaud S, Koufany M, Francois M, Netter P, Jouzeau JY, Terlain B: Rosiglitazone induces interleukin-1 receptor antagonist in interleukin-1β-stimulated rat synovial fibroblasts via a peroxisome proliferator-activated receptor β/δ-dependent mechanism. Arthritis Rheum. 2005, 52: 759-769. 10.1002/art.20868.CrossRefPubMed
36.
Zurück zum Zitat Shan ZZ, Masuko-Hongo K, Dai SM, Nakamura H, Kato T, Nishioka K: A potential role of 15-deoxy-δ (12,14)-prostaglandin J2 for induction of human articular chondrocyte apoptosis in arthritis. J Biol Chem. 2004, 279: 37939-37950. 10.1074/jbc.M402424200.CrossRefPubMed Shan ZZ, Masuko-Hongo K, Dai SM, Nakamura H, Kato T, Nishioka K: A potential role of 15-deoxy-δ (12,14)-prostaglandin J2 for induction of human articular chondrocyte apoptosis in arthritis. J Biol Chem. 2004, 279: 37939-37950. 10.1074/jbc.M402424200.CrossRefPubMed
37.
Zurück zum Zitat Ding GJ, Fischer PA, Boltz RC, Schmidt JA, Colaianne JJ, Gough A, Rubin RA, Miller DK: Characterization and quantitation of NF-κB nuclear translocation induced by interleukin-1 and tumor necrosis factor-α. Development and use of a high capacity fluorescence cytometric system. J Biol Chem. 1998, 273: 28897-28905. 10.1074/jbc.273.44.28897.CrossRefPubMed Ding GJ, Fischer PA, Boltz RC, Schmidt JA, Colaianne JJ, Gough A, Rubin RA, Miller DK: Characterization and quantitation of NF-κB nuclear translocation induced by interleukin-1 and tumor necrosis factor-α. Development and use of a high capacity fluorescence cytometric system. J Biol Chem. 1998, 273: 28897-28905. 10.1074/jbc.273.44.28897.CrossRefPubMed
38.
Zurück zum Zitat Mengshol JA, Vincenti MP, Coon CI, Barchowsky A, Brinckerhoff CE: Interleukin-1 induction of collagenase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requires p38, c-Jun N-terminal kinase, and nuclear factor κB: differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum. 2000, 43: 801-811. 10.1002/1529-0131(200004)43:4<801::AID-ANR10>3.0.CO;2-4.CrossRefPubMed Mengshol JA, Vincenti MP, Coon CI, Barchowsky A, Brinckerhoff CE: Interleukin-1 induction of collagenase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requires p38, c-Jun N-terminal kinase, and nuclear factor κB: differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum. 2000, 43: 801-811. 10.1002/1529-0131(200004)43:4<801::AID-ANR10>3.0.CO;2-4.CrossRefPubMed
39.
Zurück zum Zitat Mendes AF, Caramona MM, Carvalho AP, Lopes MC: Role of mitogen-activated protein kinases and tyrosine kinases on IL-1-induced NF-κB activation and iNOS expression in bovine articular chondrocytes. Nitric Oxide. 2002, 6: 35-44. 10.1006/niox.2001.0378.CrossRefPubMed Mendes AF, Caramona MM, Carvalho AP, Lopes MC: Role of mitogen-activated protein kinases and tyrosine kinases on IL-1-induced NF-κB activation and iNOS expression in bovine articular chondrocytes. Nitric Oxide. 2002, 6: 35-44. 10.1006/niox.2001.0378.CrossRefPubMed
40.
Zurück zum Zitat Liacini A, Sylvester J, Li WQ, Huang W, Dehnade F, Ahmad M, Zafarullah M: Induction of matrix metalloproteinase-13 gene expression by TNF-α is mediated by MAP kinases, AP-1, and NF-κB transcription factors in articular chondrocytes. Exp Cell Res. 2003, 288: 208-217. 10.1016/S0014-4827(03)00180-0.CrossRefPubMed Liacini A, Sylvester J, Li WQ, Huang W, Dehnade F, Ahmad M, Zafarullah M: Induction of matrix metalloproteinase-13 gene expression by TNF-α is mediated by MAP kinases, AP-1, and NF-κB transcription factors in articular chondrocytes. Exp Cell Res. 2003, 288: 208-217. 10.1016/S0014-4827(03)00180-0.CrossRefPubMed
41.
Zurück zum Zitat Fan Z, Bau B, Yang H, Aigner T: IL-1β induction of IL-6 and LIF in normal articular human chondrocytes involves the ERK, p38 and NFκB signaling pathways. Cytokine. 2004, 28: 17-24. 10.1016/j.cyto.2004.06.003.CrossRefPubMed Fan Z, Bau B, Yang H, Aigner T: IL-1β induction of IL-6 and LIF in normal articular human chondrocytes involves the ERK, p38 and NFκB signaling pathways. Cytokine. 2004, 28: 17-24. 10.1016/j.cyto.2004.06.003.CrossRefPubMed
42.
Zurück zum Zitat Liacini A, Sylvester J, Li WQ, Zafarullah M: Inhibition of interleukin-1-stimulated MAP kinases, activating protein-1 (AP-1) and nuclear factor κB (NF-κB) transcription factors down-regulates matrix metalloproteinase gene expression in articular chondrocytes. Matrix Biol. 2002, 21: 251-262. 10.1016/S0945-053X(02)00007-0.CrossRefPubMed Liacini A, Sylvester J, Li WQ, Zafarullah M: Inhibition of interleukin-1-stimulated MAP kinases, activating protein-1 (AP-1) and nuclear factor κB (NF-κB) transcription factors down-regulates matrix metalloproteinase gene expression in articular chondrocytes. Matrix Biol. 2002, 21: 251-262. 10.1016/S0945-053X(02)00007-0.CrossRefPubMed
43.
Zurück zum Zitat Miyazaki Y, Tsukazaki T, Hirota Y, Yonekura A, Osaki M, Shindo H, Yamashita S: Dexamethasone inhibition of TGF β-induced cell growth and type II collagen mRNA expression through ERK-integrated AP-1 activity in cultured rat articular chondrocytes. Osteoarthritis Cartilage. 2000, 8: 378-385. 10.1053/joca.1999.0313.CrossRefPubMed Miyazaki Y, Tsukazaki T, Hirota Y, Yonekura A, Osaki M, Shindo H, Yamashita S: Dexamethasone inhibition of TGF β-induced cell growth and type II collagen mRNA expression through ERK-integrated AP-1 activity in cultured rat articular chondrocytes. Osteoarthritis Cartilage. 2000, 8: 378-385. 10.1053/joca.1999.0313.CrossRefPubMed
44.
Zurück zum Zitat Bushel P, Kim JH, Chang W, Catino JJ, Ruley HE, Kumar CC: Two serum response elements mediate transcriptional repression of human smooth muscle α-actin promoter in ras-transformed cells. Oncogene. 1995, 10: 1361-1370.PubMed Bushel P, Kim JH, Chang W, Catino JJ, Ruley HE, Kumar CC: Two serum response elements mediate transcriptional repression of human smooth muscle α-actin promoter in ras-transformed cells. Oncogene. 1995, 10: 1361-1370.PubMed
45.
Zurück zum Zitat Schreiber M, Kolbus A, Piu F, Szabowski A, Mohle-Steinlein U, Tian J, Karin M, Angel P, Wagner EF: Control of cell cycle progression by c-Jun is p53 dependent. Genes Dev. 1999, 13: 607-619.PubMedCentralCrossRefPubMed Schreiber M, Kolbus A, Piu F, Szabowski A, Mohle-Steinlein U, Tian J, Karin M, Angel P, Wagner EF: Control of cell cycle progression by c-Jun is p53 dependent. Genes Dev. 1999, 13: 607-619.PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Takakura M, Kyo S, Inoue M, Wright WE, Shay JW: Function of AP-1 in transcription of the telomerase reverse transcriptase gene (TERT) in human and mouse cells. Mol Cell Biol. 2005, 25: 8037-8043. 10.1128/MCB.25.18.8037-8043.2005.PubMedCentralCrossRefPubMed Takakura M, Kyo S, Inoue M, Wright WE, Shay JW: Function of AP-1 in transcription of the telomerase reverse transcriptase gene (TERT) in human and mouse cells. Mol Cell Biol. 2005, 25: 8037-8043. 10.1128/MCB.25.18.8037-8043.2005.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Toliver-Kinsky T, Wood T, Perez-Polo JR: Nuclear factor κB/p49 is a negative regulatory factor in nerve growth factor-induced choline acetyltransferase promoter activity in PC12 cells. J Neurochem. 2000, 75: 2241-2251. 10.1046/j.1471-4159.2000.0752241.x.CrossRefPubMed Toliver-Kinsky T, Wood T, Perez-Polo JR: Nuclear factor κB/p49 is a negative regulatory factor in nerve growth factor-induced choline acetyltransferase promoter activity in PC12 cells. J Neurochem. 2000, 75: 2241-2251. 10.1046/j.1471-4159.2000.0752241.x.CrossRefPubMed
48.
Zurück zum Zitat Wachtel M, Bolliger MF, Ishihara H, Frei K, Bluethmann H, Gloor SM: Down-regulation of occludin expression in astrocytes by tumour necrosis factor (TNF) is mediated via TNF type-1 receptor and nuclear factor-κB activation. J Neurochem. 2001, 78: 155-162. 10.1046/j.1471-4159.2001.00399.x.CrossRefPubMed Wachtel M, Bolliger MF, Ishihara H, Frei K, Bluethmann H, Gloor SM: Down-regulation of occludin expression in astrocytes by tumour necrosis factor (TNF) is mediated via TNF type-1 receptor and nuclear factor-κB activation. J Neurochem. 2001, 78: 155-162. 10.1046/j.1471-4159.2001.00399.x.CrossRefPubMed
49.
Zurück zum Zitat Sohur US, Dixit MN, Chen CL, Byrom MW, Kerr LA: Rel/NF-κB represses bcl-2 transcription in pro-B lymphocytes. Gene Expr. 1999, 8: 219-229.PubMed Sohur US, Dixit MN, Chen CL, Byrom MW, Kerr LA: Rel/NF-κB represses bcl-2 transcription in pro-B lymphocytes. Gene Expr. 1999, 8: 219-229.PubMed
50.
Zurück zum Zitat Todorov VT, Volkl S, Muller M, Bohla A, Klar J, Kunz-Schughart LA, Hehlgans T, Kurtz A: Tumor necrosis factor-α activates NFκB to inhibit renin transcription by targeting cAMP-responsive element. J Biol Chem. 2004, 279: 1458-1467. 10.1074/jbc.M308697200.CrossRefPubMed Todorov VT, Volkl S, Muller M, Bohla A, Klar J, Kunz-Schughart LA, Hehlgans T, Kurtz A: Tumor necrosis factor-α activates NFκB to inhibit renin transcription by targeting cAMP-responsive element. J Biol Chem. 2004, 279: 1458-1467. 10.1074/jbc.M308697200.CrossRefPubMed
51.
Zurück zum Zitat Setoguchi K, Misaki Y, Terauchi Y, Yamauchi T, Kawahata K, Kadowaki T, Yamamoto K: Peroxisome proliferator-activated receptor-γ haploinsufficiency enhances B cell proliferative responses and exacerbates experimentally induced arthritis. J Clin Invest. 2001, 108: 1667-1675. 10.1172/JCI200113202.PubMedCentralCrossRefPubMed Setoguchi K, Misaki Y, Terauchi Y, Yamauchi T, Kawahata K, Kadowaki T, Yamamoto K: Peroxisome proliferator-activated receptor-γ haploinsufficiency enhances B cell proliferative responses and exacerbates experimentally induced arthritis. J Clin Invest. 2001, 108: 1667-1675. 10.1172/JCI200113202.PubMedCentralCrossRefPubMed
Metadaten
Titel
Peroxisome proliferator-activated receptor γ1 expression is diminished in human osteoarthritic cartilage and is downregulated by interleukin-1β in articular chondrocytes
verfasst von
Hassan Afif
Mohamed Benderdour
Leandra Mfuna-Endam
Johanne Martel-Pelletier
Jean-Pierre Pelletier
Nicholas Duval
Hassan Fahmi
Publikationsdatum
01.04.2007
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 2/2007
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/ar2151

Weitere Artikel der Ausgabe 2/2007

Arthritis Research & Therapy 2/2007 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neue S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Intervallfasten zur Regeneration des Herzmuskels?

14.05.2024 Herzinfarkt Nachrichten

Die Nahrungsaufnahme auf wenige Stunden am Tag zu beschränken, hat möglicherweise einen günstigen Einfluss auf die Prognose nach akutem ST-Hebungsinfarkt. Darauf deutet eine Studie an der Uniklinik in Halle an der Saale hin.

Klimaschutz beginnt bei der Wahl des Inhalators

14.05.2024 Klimawandel Podcast

Auch kleine Entscheidungen im Alltag einer Praxis können einen großen Beitrag zum Klimaschutz leisten. Die neue Leitlinie zur "klimabewussten Verordnung von Inhalativa" geht mit gutem Beispiel voran, denn der Wechsel vom klimaschädlichen Dosieraerosol zum Pulverinhalator spart viele Tonnen CO2. Leitlinienautor PD Dr. Guido Schmiemann erklärt, warum nicht nur die Umwelt, sondern auch Patientinnen und Patienten davon profitieren.

Zeitschrift für Allgemeinmedizin, DEGAM

Update Innere Medizin

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