Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-05-31T00:16:21.050Z Has data issue: false hasContentIssue false

Polyphenolic extract from extra virgin olive oil inhibits the inflammatory response in IL-1β-activated synovial fibroblasts

Published online by Cambridge University Press:  26 October 2018

María Ángeles Rosillo
Affiliation:
Department of Pharmacology, Faculty of Pharmacy, University of Seville, Profesor García González, 2, 41012 Seville, Spain
Catalina Alarcón-de-la-Lastra
Affiliation:
Department of Pharmacology, Faculty of Pharmacy, University of Seville, Profesor García González, 2, 41012 Seville, Spain
María Luisa Castejón
Affiliation:
Department of Pharmacology, Faculty of Pharmacy, University of Seville, Profesor García González, 2, 41012 Seville, Spain
Tatiana Montoya
Affiliation:
Department of Pharmacology, Faculty of Pharmacy, University of Seville, Profesor García González, 2, 41012 Seville, Spain
Marta Cejudo-Guillén
Affiliation:
Department of Pharmacology, Faculty of Pharmacy, University of Seville, Profesor García González, 2, 41012 Seville, Spain
Marina Sánchez-Hidalgo*
Affiliation:
Department of Pharmacology, Faculty of Pharmacy, University of Seville, Profesor García González, 2, 41012 Seville, Spain
*
*Corresponding author: Dr M. Sánchez-Hidalgo, fax +34 954 55 6074, email hidalgosanz@us.es
Rights & Permissions [Opens in a new window]

Abstract

The polyphenolic extract (PE) from extra virgin olive oil (EVOO) has been shown to possess important anti-inflammatory and joint protective properties in murine models of rheumatoid arthritis (RA). This study was designed to evaluate the effects of PE on IL-1β-activated human synovial fibroblasts SW982 cell line. PE from EVOO treatment inhibited IL-1β-induced matrix metalloproteases (P<0·001), TNF-α and IL-6 production (P<0·001). Similarly, IL-1β-induced cyclo-oxygenase-2 and microsomal PGE synthase-1 up-regulations were down-regulated by PE (P<0·001). Moreover, IL-1β-induced mitogen-activated protein kinase (MAPK) phosphorylation and NF-κB activation were ameliorated by PE (P<0·001). These results suggest that PE from EVOO reduces the production of proinflammatory mediators in human synovial fibroblasts; particularly, these protective effects could be related to the inhibition of MAPK and NF-κB signalling pathways. Taken together, PE from EVOO probably could provide an attractive complement in management of diseases associated with over-activation of synovial fibroblasts, such as RA.

Type
Full Papers
Copyright
© The Authors 2018 

Rheumatoid arthritis (RA) is a chronic autoimmune disease of unknown origin that primarily affects the joints and ultimately leads to their destruction( Reference Huber, Distler and Tarner 1 ). Nowadays, although the aetiopathogenesis of RA is only partially understood, it is known that the involvement of immune cells and their respective proinflammatory mediators is a common hallmark of RA as of all systemic autoimmune disorders( Reference Smith and Haynes 2 ).

Synovial fibroblasts (SF) are unique cells that populate the intimal lining of the synovium( Reference Bottini and Firestein 3 ). In non-affected healthy joints, resident SF are responsible for continuous physiologic remodelling of the matrix, whereas in RA patients SF present an altered phenotype and play a pivotal role in the pathogenesis of RA( Reference Neumann, Lefevre and Zimmermann 4 ). RA-SF attach to the articular cartilage, produce cartilage matrix-degrading enzymes and once cartilage is eroded they invade the underlying bone and activate osteoclast-mediated bone resorption. Various proinflammatory mediators, including IL-6, IL-1β, TNF-α and matrix metalloproteinases (MMP), released from RA-SF are involved in the destruction of both articular bone and cartilage( Reference Feldmann and Maini 5 ). The regulation of these pro-inflammatory mediator gene expressions involves multiple signal transduction pathways including mitogen-activated protein kinases (MAPK), which comprise extracellular signal-regulated kinases (ERK1/2 or p42/p44), c-Jun N-terminal kinases (JNK)1/2/3 and p38 and the NF-κB, which is also activated in RA-SF and appears to be important in perpetuating disease, as well as in mediating synovial inflammation( Reference Pap, Muller-Ladner and Gay 6 ). Particularly, NF-κB plays an important role in MMP induction and also regulates a wide range of genes that contribute to inflammation, such as IL-1β, TNF-α, IL-6, chemokines and microsomal PG E synthase-1 (mPGES-1), an efficient downstream enzyme co-localised and functionally coupled with the inducible enzyme cyclo-oxygenase (COX)-2( Reference McCoy, Wicks and Audoly 7 ). An excessive production of cytokines by RA-SF has been shown to induce proliferation of these cells and facilitate invasion into the adjacent tissues( Reference Garcia-Vicuna, Gomez-Gaviro and Dominguez-Luis 8 ). These lead to the destruction of articular cartilage and subchondral bone, resulting in joint deformity and a great deal of pain in RA patients.

Although new biological therapies have improved the treatment of chronic inflammatory diseases, including RA, these drugs are effective only in a fraction of patients and have other limitations including adverse effects and a high cost. Given the link between diet and RA, there are a number of studies focusing on certain foods that can help control the inflammation that characterises this autoimmune condition, particularly with respect to cytokine suppression( Reference Rosillo, Alarcón-de-la-Lastra and Sanchez-Hidalgo 9 ). Many of them are found in the so-called Mediterranean diet, which emphasises fish, vegetables and olive oil, among other staples. In fact, the Mediterranean diet has been associated with a reduced incidence of certain pathologies related to chronic inflammation and the immune system( Reference Aparicio-Soto, Sanchez-Hidalgo and Rosillo 10 ). Particularly, extra virgin olive oil (EVOO), the main source of fat in Mediterranean diet, may also help reduce inflammation( Reference Alarcón-de-la-Lastra, Barranco and Motilva 11 ). The health benefits promoted by EVOO cannot only be attributed to its high content in MUFA, but a wide range of evidence indicates that many of the beneficial effects of EVOO intake are owing to its minor highly bioactive components. Among them, phenolic compounds such as hydroxytyrosol (HTy), tyrosol and oleuropein have shown anti-inflammatory and antioxidant effects( Reference Aparicio-Soto, Sanchez-Hidalgo and Rosillo 10 , Reference Cardeno, Sanchez-Hidalgo and Alarcón-de-la-Lastra 12 ).

Our research group has previously demonstrated that treatment with polyphenolic extract (PE) from EVOO or dietary treatment with PE-enriched EVOO diet improved the progression of damage in a collagen-induced arthritis (CIA) model in mice( Reference Rosillo, Alcaraz and Sanchez-Hidalgo 13 , Reference Rosillo, Sanchez-Hidalgo and Sanchez-Fidalgo 14 ). In addition, Impellizzeri et al. ( Reference Impellizzeri, Esposito and Mazzon 15 ) have shown the anti-arthritic effect of oleuropein aglycone in CIA model.

Taking this background into account, the aim of this study was to explore the potential effects of PE from EVOO treatment, in IL-1β-stimulated SW982 cells (human RA-SF cell line), and provide in-depth insight into the signalling pathways involved in its anti-inflammatory effect. Furthermore, the production of proinflammatory mediators, the protein expression of proinflammatory enzymes and the role of MAPK and NF-κB pathways were also determined.

Methods

Reagents

PE from EVOO was obtained by the method described by Vázquez-Roncero et al. ( Reference Vázquez-Roncero, Janer del Valle and Janer del Valle 16 ), with some modifications( Reference Allouche, Jimenez and Gaforio 17 ). For the characterisation, PE (70–75 mg) extracted from EVOO (75 g) was dissolved in CDCl3 (750 µl) or in dimethylsulfoxide (DMSO)-d6 (750 µl) and a precisely measured volume of the solution (550 µl) was placed in a 5-mm NMR tube for the detection and quantification of phenolic compounds. The phenolic mixture to be analysed in DMSO-d6 was previously dissolved in deuterated methanol (MeOD)–D2O 1:1 (1 ml) and concentrated to dryness at reduced pressure in order to exchange hydroxyl protons with 2H nuclei. The NMR solvents were purchased from Sigma-Aldrich. NMR experiments were conducted on a Bruker Avance III 700 spectrometer, operating at 700·25 MHz. The probe temperature was 24·8±1°C. All chemical shifts were given in parts per million (ppm), and the J couplings in Hz. The solvent itself was used as a chemical shift reference (7·26 ppm for CHCl3, and 2·50 for DMSO-d6). 1H NMR spectra were acquired with the following acquisition parameters: acquisition time 2·3 s, relaxation delay 5 s, spectral width of 0–20 ppm, data points 32k, number of scans 32 and line broadening of 0·3 Hz. Post-acquisition processing included zero-filling to 64k. The spectra were phase-corrected automatically using TOPSPIN, and integration was performed manually. We have determined the levels of oleocanthal and oleacein following a modification of the method developed by Karkoula et al. ( Reference Karkoula, Skantzari and Melliou 18 ) for direct measurement of both dialdehydes in olive oil by quantitative high-resolution 1H NMR in CDCl3. The levels of other phenolic compounds have been determined by the procedure described by Christophoridou & Dais( Reference Christophoridou and Dais 19 ). For the experiments, PE from EVOO was dissolved in DMSO and then diluted with the medium (final DMSO concentration ≤0·05 % (v/v)).

Cell culture

The human synovial cell line SW982 was obtained from American Tissue Culture Collection (ATCC®, Number HTB-93). SW982 cells were routinely cultured in T-150 flasks (NuncTM®) and grown in Dulbecco’s modified Eagle’s medium (DMEM) with 2 mm l-glutamine, 10 % fetal bovine serum (FBS) and 1 % penicillin–streptomycin at 37°C, 5 % CO2.

Cell viability assay

Cells seeded in ninety-six-well plates (1×105 cells/well) were incubated in the presence or absence of PE (1·6–200 µg/ml) for 24 h. At the end of the exposure time, the effect of these compounds on cell growth/viability was analysed by sulforhodamine B (SRB) assay( Reference Skehan, Storeng and Scudiero 20 ). After incubation time, adherent cell cultures were fixed in situ by adding 50 µl of 50 % (w/v) cold TCA (Sigma-Aldrich®) and incubated for 60 min at 4°C. The supernatant was discarded, and plates were washed five times with deionised water and dried. In all, 50 µl of SRB (Sigma-Aldrich®) solution (0·4 % w/v) in 1 % acetic acid (Panreac®) was added to each well and incubated for 30 min at room temperature. Plates containing SRB solution were washed five times with 1 % acetic acid. Then, plates were air-dried, and 100 µl/well of 10 mmol/l TRIS base, pH 10·5 (Sigma-Aldrich®), was added, and the absorbance of each well was read on an ELISA reader at 510 nm (BioRad®). Finally, cell survival was measured as the percentage of absorbance compared with that obtained in control cells (non-treated cells).

Determination of matrix metalloproteinase and proinflammatory cytokines by ELISA

SW982 were plated in twenty-four-well plates (2·5×105 cells/well) for 48 h before treatment. Cells were incubated at 37°C in the presence or absence of PE (50, 25 and 12·5 µg/ml) for 24 h in DMEM containing 10 % (v/v) FBS in a 5 % CO2 atmosphere; then, IL-1β (5 ng/ml) was added and incubated for 24 h. Culture supernatants were collected and stored at –80°C. The levels of MMP-1, MMP-3, IL-6 and TNF-α were determined in culture supernatants from the above experiments using commercially available ELISA kits essentially according to the instructions of the manufacturers: MMP-1, RayBiotech®; MMP-3, R&D System®; and IL-6 and TNF-α, eBioscience®.

Immunoblotting detection

Cells (2·5×105 cells/ml) were left untreated or treated with PE (50, 25 and 12·5 µg/ml) for 24 h and stimulated with IL-1β at different time points depending on the protein assayed. After incubation, cells were rinsed, scraped off, collected in ice-cold PBS containing a cocktail of protease and phosphatase inhibitors and processed as described by Sanchez-Hidalgo et al. ( Reference Sanchez-Hidalgo, Martin and Villegas 21 ) to isolate proteins. Protein concentration was measured for each sample using a protein assay reagent (Bio-Rad®) according to Bradford’s method and using γ-globulin as a standard( Reference Bradford 22 ). Aliquots of supernatant containing equal amounts of protein (20 µg) were separated on 10 % acrylamide gel by SDS–PAGE. In the next step, the proteins were electrophoretically transferred into a nitrocellulose membrane and incubated with specific primary antibodies overnight at 4°C: rabbit anti-COX-2 (1:1000) and rabbit polyclonal anti-mPGES-1 (1:200) (Cayman Chemical®); rabbit anti-IκBα (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, α) (1:1000), rabbit anti-phosphorylated ERK 1/2 (anti-ppERK1/2) (1:1000) and mouse anti-pERK(1/2) (1:1000) (Cell Signaling®); and mouse anti-pJNK (1:200), rabbit anti-JNK (1:200), mouse anti-p38 mitogen-activated protein kinase (anti-pp38) (1:200) and rabbit anti-p38 (1:200) (Santa Cruz Biotechnology®). After rinsing, the membranes were incubated with a horseradish peroxidase-labelled secondary antibody anti-rabbit (Cayman Chemical®) (1:50 000) or anti-mouse (Dako®) (1:2000) containing blocking solution for 1–2 h at room temperature. To prove equal loading, the blots were analysed for β-actin expression using an anti-β-actin antibody (Sigma-Aldrich®). Immunodetection was performed using enhanced chemiluminescence light-detecting kit (Pierce®). The immunosignals were captured using an LAS-3000 Imaging System from Fujifilm Image Reader (Stamford®), and densitometry data were studied following normalisation to the housekeeping loading control. The signals were analysed and quantified by ImageJ software.

Statistical evaluation

All values in the figures and text are expressed as arithmetic means with their standard errors. Experiments were carried out in triplicate. Data were evaluated with Graph Pad Prism® version 5.01 software. The statistical significance of any difference in each parameter among the groups was evaluated by one-way ANOVA, using Tukey’s multiple-comparisons test as post hoc test. P values <0·05 were considered statistically significant. In the experiments involving densitometry, the figures shown are representative of at least three different experiments performed on different days.

Results

Chemical composition of polyphenolic extract from extra virgin olive oil

Table 1 shows the result of qualitative and quantitative analyses of PE from EVOO. A total of nine different phenolic compounds have been identified, with oleocanthal and oleacein being the main components present in PE.

Table 1 Phenolic content of polyphenolic extract determined by 1H NMR spectroscopy

ppm, Parts per million.

Effects of polyphenolic extract from extra virgin olive oil on cell viability

To evaluate the effects of PE from EVOO on the viability of SW982 cells, the SRB assay was performed. The SRB assay is an efficient method for the toxicity screening of compounds to adherent cells, based on the measurement of cellular protein content. After 24 h, our data demonstrated that the viability of SW982 cells treated with PE was not reduced at concentrations of 1·6–50 µg/ml, showing a cell viability >90 % (Fig. 1).

Fig. 1 Effect of polyphenolic extract (PE) from extra virgin olive oil (EVOO) on cell viability. The concentrations used in this study (50, 25 and 12·5 µg/ml) did not affect viability of SW982 cells. Cells were treated with PE from EVOO (1·6–200 µg/ml) for 24 h. Cell survival was measured as the percentage of absorbance compared with that obtained in control cells (non-treated cells). SRB, sulforhodamine B. , PE; , dimethylsulfoxide.

Effects of polyphenolic extract from extra virgin olive oil on matrix metalloproteinase production

As shown in Fig. 2, IL-1β at 5 ng/ml significantly increased the MMP-1 and MMP-3 production in SW982 cells. In contrast, pre-treatment with PE for 24 h blocked the IL-1β-induced MMP-1 and MMP-3 up-regulation (Fig. 2).

Fig. 2 Inhibitory effects of polyphenolic extract (PE) from extra virgin olive oil on the production of matrix metalloproteinase (MMP) and proinflammatory cytokines by SW982 cells. SW982 cells were cultured for 24 h in the presence or absence of PE and stimulated with IL-1β (5 ng/ml). As controls (C), cells were untreated in the absence of IL-1β. Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (not stimulated) (P<0·001). Mean values were significantly different from IL-1β-stimulated control cells: †† P<0·01, ††† P<0·001.

Effects of polyphenolic extract from extra virgin olive oil on proinflammatory cytokine production

Treatment of SW982 cells with IL-1β increased the levels of IL-6 and TNF-α when compared with cells in the absence of stimulus (Fig. 2). However, pre-treatment with PE resulted in a significant inhibition of the production of both proinflammatory cytokines induced by IL-1β (Fig. 2).

Polyphenolic extract from extra virgin olive oil down-regulated the cyclo-oxygenase-2 and microsomal PGE synthase-1 over-expression induced by IL-1β

We investigated the possible effects of PE on COX-2 and mPGES-1 inflammation-related enzymes as PG play a crucial role in the pathogenesis of RA. COX-2 and mPGES-1 protein expressions were remarkably increased after 24-h IL-1β stimulation, whereas a significant down-regulation in both pro-inflammatory protein expression was observed in SW982 treated with PE (Fig. 3).

Fig. 3 Polyphenolic extract (PE) from extra virgin olive oil inhibits cyclo-oxygenase-2 (COX-2, ) and microsomal PGE synthase-1 (m-PGES-1, ) protein expressions in IL-1β-stimulated SW982 cells. Cells were untreated or treated with PE (50, 25 or 12·5 µg/ml) for 24 h in the presence of IL-1β (5 ng/ml). Control cells (C) were incubated in the absence of IL-1β. The plots represent band intensity. β-Actin served as an equal loading control for normalisation. Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (not stimulated) (P<0·001). ††† Mean values were significantly different from IL-1β-stimulated control cells (P<0·001).

Effects of polyphenolic extract from extra virgin olive oil on IL-1β-induced mitogen-activated protein kinase activation in SW982 cells

To explore possible signalling pathways underlying the anti-inflammatory effects, we evaluated whether PE was able to modulate JNK, p38 and ERK MAPK activation after 30 min of IL-1β stimulation by Western blotting. Our findings demonstrated that SW982 cells stimulated with IL-1β induced remarkably higher levels of phosphorylation of JNK, p38 and ERK MAPK in comparison with unstimulated cells. In contrast, PE treatment was able to reduce the IL-1β-induced phosphorylation of JNK, p38 and ERK MAPK to basal levels similar to those detected in unstimulated cells (Fig. 4).

Fig. 4 Effects of polyphenolic extract (PE) from extra virgin olive oil on the mitogen-activated protein kinase signalling pathway in IL-1β-stimulated SW982 cells. Cells were untreated or treated with PE (50, 25 or 12·5 µg/ml) for 24 h and stimulated with IL-1β (5 ng/ml) for 30 min. Control cells (C) were incubated in the absence of IL-1β. Densitometry was performed following normalisation to the control housekeeping genes (JNK, p38 and ERK1/2). Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (not stimulated) (P<0·001). Mean values were significantly different from IL-1β-stimulated control cells: † P<0·05, †† P<0·01, ††† P<0·001. , Phospho-c-Jun N-terminal kinase; , pp38; , phospho-extracellular signal-regulated kinase 1/2.

Polyphenolic extract from extra virgin olive oil prevented IL-1β-induced IκBα degradation on SW982 cells

NF-κB is a pleiotropic mediator in the control of several inducible and tissue-specific genes and is one of the key regulators of the cellular responses to oxidative stress in mammalian cells. NF-κB activation is initiated by the degradation of IκBα. After IκBα is degraded, NF-κB, in the NF-κB–IκBα complex, is free to be translocated into the nucleus, where it can induce the expression of pro-inflammatory genes contributing to the damage. To determine whether proinflammatory protein down-regulation is regulated by the IκBα pathway, we measured the expression levels of IκBα in SW982 cells pre-treated with PE (24 h) in the presence or absence of stimulus (5 ng/ml IL-1β) for 6 h. As shown in Fig. 5, IκB-α was reduced in SW982 cells stimulated with IL-1β, whereas pre-treatment with PE from EVOO was able to prevent IκB-α degradation in IL-1β-stimulated SW082 cells (Fig. 5).

Fig. 5 Polyphenolic extract (PE) from extra virgin olive oil pre-treatment prevented IκB-α degradation in IL-1β-stimulated SW982 cells. Cells were untreated or treated with PE (50, 25 or 12·5 µg/ml) for 24 h and stimulated with IL-1β (5 ng/ml) for 6 h. Control cells were incubated in the absence of IL-1β. Densitometry was performed following normalisation to the control (β-actin housekeeping gene). Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (no stimulated) (P<0·001). Mean values were significantly different from IL-1β-stimulated control cells: † P<0·05 and †† P<0·01.

Discussion

The development of effective strategies for the prevention and therapy of RA is highly desired. On the basis of our assessment in cultured SW082 SF, PE from EVOO may have promising potential for the development of a novel and effective nutritional supplement for RA suppressing key proinflammatory mediators and cytokines involved in the pathogenesis of RA through blockage of MAPK and NF-κB signalling pathways.

In RA, SF act as a major cell population in the invasive pannus to participate in the chronic inflammatory responses( Reference Firestein 23 ). In the current study, we have shown, for the first time, which PE from EVOO was able to inhibit the activation of SW982 human synovial cells induced by IL-1β, preventing the inflammatory response.

Inflammatory changes of SF play a vital role in the progression of RA. In this sense, it has been reported that synovial reaction in RA patients is characterised by an over-production of pro-inflammatory cytokines, such as TNF-α, IL-1β and IL-6, which are known to play key roles in the pathogenesis of RA( Reference Tanaka 24 ). In addition, it has been demonstrated that an increase of serum c-globulin and the emergence of rheumatoid factors are related to IL-6 levels. In fact, high levels of IL-6 have been detected in both sera and synovial fluids from the affected joints of RA patients( Reference Baillet, Gossec and Paternotte 25 ). On the other hand, TNF-α, a pro-inflammatory agent that is formed in the macrophages and T cells, is reportedly involved in early joint swelling, chronic joint inflammation and the concomitant erosive changes in cartilage and bone( Reference Sommerfelt, Feuerherm and Jones 26 ). In fact, development of TNF-α inhibitors such as infliximab and etanercept has been shown to induce the clinical remission of RA and delay or halt the clinical and radiological progression of the disease, thus improving the quality of life of many patients( Reference Smolen, Landewe and Breedveld 27 ). However, the high cost and the adverse effect profile associated with these drugs limit their wide use as first-line medications. In this context, novel treatment possibilities are necessary for the treatment and/or control of chronic inflammatory state in RA. At present, many dietary phenolic compounds such as curcumin, resveratrol, HTy, oleuropein and epigallocatechin, among others, have reported a beneficial role in the prevention and management of RA, by suppressing the expression of proinflammatory cytokines or mediators, adhesion molecules and MMP in SF( Reference Rosillo, Alarcón-de-la-Lastra and Sanchez-Hidalgo 9 ). In the search of new strategies in RA, the present study confirmed, for the first time, that PE from EVOO treatment caused a significant inhibition of IL-1β-induced TNF-α and IL-6 release in human synovial SW982 cells. These results are in agreement with our previous observations in CIA mouse model, the most commonly studied autoimmune model of RA where PE decreased joint oedema, cell migration, cartilage degradation and bone erosion( Reference Rosillo, Alcaraz and Sanchez-Hidalgo 13 ).

In addition, the cartilage destruction of RA is mostly caused by the activity of MMP, and MMP are pivotal in the recruitment of leucocytes and macrophages into joints. Particularly, MMP-1 (collagenase 1) is one of the principal neutral proteinases that degrade native fibrillar collagens in the extracellular matrix( Reference Yamanishi and Firestein 28 ). Alternatively, MMP-3 (stromelysin) can degrade proteoglycan, type IV and type IX collagens, denatured type I and type II collagens, fibronectin, gelatin and laminin and is considered to be especially important because, in addition to its direct enzyme activity, its activation is necessary for full activation of collagenases( Reference Chen and Mattey 29 ). Our data showed that the production of both MMP-1 and MMP-3 was induced after IL-1β stimulation in human SW982 cells, whereas PE pre-treatment induced a significant down-regulation of both MMP levels in IL-1β-stimulated SW982 cells.

At the same time, cytokine and MMP production directly correlate with increased levels of PG in response to IL-1β in RA-SF( Reference Egg 30 ). Besides, COX-2 and mPGES-1, enzymes responsible for the over-production of PGE2 in inflammation, are up-regulated in RA patients( Reference Lazarus, Kubata and Eguchi 31 ), contributing to the progression of RA through EP4 receptor activation( Reference McCoy, Wicks and Audoly 7 ). In this study, we have shown that PE from EVOO decreased the expression of both COX-2 and mPGES-1 in IL-1β-stimulated SW982 cells. Our data are in agreement with our previous study where we confirmed that PE decreased mPEG-1 and COX-2 protein expression by Western blotting and immunohistochemical staining, respectively, in mice subjected to CIA( Reference Rosillo, Alcaraz and Sanchez-Hidalgo 13 ).

There is evidence that multiple stress signalling pathways, such as MAPK and NF-κB, on activation, act as pivotal regulators of proliferation, differentiation and cellular survival in RA, contributing to the pathogenic mechanisms of joint destruction and inflammation in RA( Reference Liu, Sun and Tao 32 ). It is well known that MAPK and NF-κB pathways are involved in regulating the expression of IL-6, IL-8, MMP-1 and MMP-3 in RA-SF( Reference Han, Boyle and Chang 33 , Reference Tak, Gerlag and Aupperle 34 ) and their signal transduction pathways have been found to be involved in the pathogenesis of RA( Reference Morel and Berenbaum 35 ). Particularly, the three families of MAPK, ERK, JNK and p38 are activated by various mitogens, growth factors and pro-inflammatory cytokines, such as IL-1β and TNF-α. MAPK play a critical role in the regulation of the synthesis of chemokines, cytokines, adhesion molecules and PG involved in RA and are considered as the major tyrosine phosphorylation proteins in human synovial cells stimulated with IL-1β ( Reference Barchowsky, Frleta and Vincenti 36 ). In addition, MAPK modulate MMP production by SF and drive osteoclast differentiation in RA( Reference Han, Boyle and Chang 33 ).

The transcription factor NF-kB has been well recognised as a pivotal regulator of inflammation in RA. It has been evidenced that NF-kB is involved in abnormal apoptosis and proliferation of RA fibroblast-like synovial cells, as well as differentiation and activation of bone resorbing activity of osteoclasts( Reference Makarov 37 ). In RA, NF-κB is over-expressed in the inflamed synovium( Reference Han, Boyle and Manning 38 ), where its activity may enhance recruitment of inflammatory cells and production of proinflammatory mediators such as IL-1β, IL-6, IL-8 and TNF-α ( Reference Tak and Firestein 39 ).

Consistent with other observations, we described that IL-1β enhanced the activation of MAPK; increased the phosphorylation of ERK1/2, JNK and p38; and endorsed IκBα degradation( Reference Castejon, Rosillo and Montoya 40 ). Nevertheless, PE from EVOO pre-treatment prevented both MAPK and NF-κB signalling pathway activation. These results are in concordance with our previous studies in which oral administration of PE from EVOO was able to down-regulate the arthritic process in a CIA murine model blocking these signalling pathways( Reference Rosillo, Alcaraz and Sanchez-Hidalgo 13 ). Similar results were also described by Cardeno et al. ( Reference Cardeno, Sanchez-Hidalgo and Aparicio-Soto 41 ), where PE from EVOO down-regulated efficiently the inflammatory response in lipopolysaccharide-activated murine peritoneal macrophages suppressing NF-κB and MAPK signalling pathways.

Among the components present in the PE from EVOO, oleocanthal was the main component detected in our extract. Nevertheless, although oleocanthal has been shown to exert a key role in the anti-inflammatory and anti-arthritic effects in an in vitro model of degenerative joint disease( Reference Iacono, Gomez and Sperry 42 , Reference Scotece, Gomez and Conde 43 ), other minor bioactive compounds present in the PE, such as HTy and hydroxytyrosol acetate (HTy-Ac), among others, might contribute synergically to these beneficial effects. In fact, previous studies carried out by our research group have demonstrated the anti-inflammatory and joint protective effects of both HTy and HTy-Ac in in vitro ( Reference Rosillo, Sánchez-Hidalgo and Castejón 44 ) and in vivo ( Reference Rosillo, Sanchez-Hidalgo and Gonzalez-Benjumea 45 ) models of RA.

Our study reveals interesting insights into the antioxidant, anti-inflammatory and immunomodulatory properties of PE from EVOO in an establishment in vitro model for human RA, IL-1β-stimulated SW982 cells. These beneficial effects were accompanied by a remarkable reduction of proinflammatory mediators including MMP-1, MMP-3, COX-2, mPGES-1 and cytokines IL-6 and TNF-α. The possible signalling pathways involved in this protective effect could be related to the inhibition of MAPK and NF-κB controlling the production of inflammatory mediators. The observations reported at bench are promising and suggest that the dietary PE from EVOO may influence the course of this disease in humans, ameliorating the RA symptoms and down-regulating the inflammation at the molecular level, opening a new interesting field in which is necessary to explore and bringing them to the forefront of the treatment of this chronic human disease. Nevertheless, it is also important to mention that the bioavailability and the mechanism by which PE from EVOO is absorbed remains unknown and is very limited to their individual compounds( Reference Vissers, Zock and Roodenburg 46 Reference Tan, Tuck and Stupans 48 ). Therefore, it is necessary to provide deep insight into the processes of absorption and biotransformation of this fraction in order to establish the role of the flora of the colon in these processes and to determine the types of metabolites formed that can contribute to the biological effects finally observed.

Conclusions

PE from EVOO could play an important role in the anti-inflammatory effect of EVOO and probably provide an attractive complement in management of diseases associated with over-activation of SF, such as RA. Nevertheless, future studies should focus more on understanding the biochemical and biological activities of PE from EVOO underlying their effective doses in humans and the dose dependence of their effects.

Acknowledgements

The authors thank I+D+i de Oleoestepa SAC department who kindly provided the EVOO. The authors gratefully acknowledge the assistance of Centre for Technology and Innovation Research, University of Seville (CITIUS).

This work was supported by Junta de Andalucía (P-10AGR-6609). M. A. R. thanks Junta de Andalucía for the award of a pre-doctoral and a post-doctoral grant associated with P-10AGR-6609.

M. A. R. performed the experiments and data analysis. M. L. C, T. M. and M. C.-G. performed part of the analysis. C. A.-d.-l.-L. and M. S.-H. designed the study and prepared the manuscript. All authors read and approved the final content of the manuscript.

The authors declare that there are no conflicts of interest.

References

1. Huber, LC, Distler, O, Tarner, I, et al. (2006) Synovial fibroblasts: key players in rheumatoid arthritis. Rheumatology 45, 669675.Google Scholar
2. Smith, JB & Haynes, MK (2002) Rheumatoid arthritis–a molecular understanding. Ann Intern Med 136, 908922.Google Scholar
3. Bottini, N & Firestein, GS (2013) Duality of fibroblast-like synoviocytes in RA: passive responders and imprinted aggressors. Nat Rev Rheumatol 9, 2433.Google Scholar
4. Neumann, E, Lefevre, S, Zimmermann, B, et al. (2010) Rheumatoid arthritis progression mediated by activated synovial fibroblasts. Trends Mol Med 16, 458468.Google Scholar
5. Feldmann, M & Maini, SR (2008) Role of cytokines in rheumatoid arthritis: an education in pathophysiology and therapeutics. Immunol Rev 223, 719.Google Scholar
6. Pap, T, Muller-Ladner, U, Gay, RE, et al. (2000) Fibroblast biology. Role of synovial fibroblasts in the pathogenesis of rheumatoid arthritis. Arthritis Res 2, 361367.Google Scholar
7. McCoy, JM, Wicks, JR & Audoly, LP (2002) The role of prostaglandin E2 receptors in the pathogenesis of rheumatoid arthritis. J Clin Invest 110, 651658.Google Scholar
8. Garcia-Vicuna, R, Gomez-Gaviro, MV, Dominguez-Luis, MJ, et al. (2004) CC and CXC chemokine receptors mediate migration, proliferation, and matrix metalloproteinase production by fibroblast-like synoviocytes from rheumatoid arthritis patients. Arthritis Rheum 50, 38663877.Google Scholar
9. Rosillo, MA, Alarcón-de-la-Lastra, C & Sanchez-Hidalgo, M (2016) An update on dietary phenolic compounds in the prevention and management of rheumatoid arthritis. Food Funct 7, 29432969.Google Scholar
10. Aparicio-Soto, M, Sanchez-Hidalgo, M, Rosillo, MA, et al. (2016) Extra virgin olive oil: a key functional food for prevention of immune-inflammatory diseases. Food Funct 7, 44924505.Google Scholar
11. Alarcón-de-la-Lastra, C, Barranco, MD, Motilva, V, et al. (2001) Mediterranean diet and health: biological importance of olive oil. Curr Pharm Design 7, 933950.Google Scholar
12. Cardeno, A, Sanchez-Hidalgo, M & Alarcón-de-la-Lastra, C (2013) An up-date of olive oil phenols in inflammation and cancer: molecular mechanisms and clinical implications. Curr Med Chem 20, 47584776.Google Scholar
13. Rosillo, MA, Alcaraz, MJ, Sanchez-Hidalgo, M, et al. (2014) Anti-inflammatory and joint protective effects of extra-virgin olive-oil polyphenol extract in experimental arthritis. J Nutr Biochem 25, 12751281.Google Scholar
14. Rosillo, MA, Sanchez-Hidalgo, M, Sanchez-Fidalgo, S, et al. (2016) Dietary extra-virgin olive oil prevents inflammatory response and cartilage matrix degradation in murine collagen-induced arthritis. Eur J Nutr 55, 315325.Google Scholar
15. Impellizzeri, D, Esposito, E, Mazzon, E, et al. (2011) Oleuropein aglycone, an olive oil compound, ameliorates development of arthritis caused by injection of collagen type II in mice. J Pharmacol Exp Ther 339, 859869.Google Scholar
16. Vázquez-Roncero, A, Janer del Valle, C & Janer del Valle, ML (1976) Componentes fenolicos de la aceituna. III Polifenoles del aceite (Phenolic components of the olive. III Oil polyphenols). Grasas Aceites 27, 185190.Google Scholar
17. Allouche, Y, Jimenez, A, Gaforio, JJ, et al. (2007) How heating affects extra virgin olive oil quality indexes and chemical composition. J Agric Food Chem 55, 96469654.Google Scholar
18. Karkoula, E, Skantzari, A, Melliou, E, et al. (2012) Direct measurement of oleocanthal and oleacein levels in olive oil by quantitative (1)H NMR. Establishment of a new index for the characterization of extra virgin olive oils. J Agric Food Chem 60, 1169611703.Google Scholar
19. Christophoridou, S & Dais, P (2009) Detection and quantification of phenolic compounds in olive oil by high resolution 1H nuclear magnetic resonance spectroscopy. Anal Chim Acta 633, 283292.Google Scholar
20. Skehan, P, Storeng, R, Scudiero, D, et al. (1990) New colorimetric cytotoxicity assay for anticancer-drug screening. J Natl Cancer Inst 82, 11071112.Google Scholar
21. Sanchez-Hidalgo, M, Martin, AR, Villegas, I, et al. (2005) Rosiglitazone, an agonist of peroxisome proliferator-activated receptor gamma, reduces chronic colonic inflammation in rats. Biochem Pharmacol 69, 17331744.Google Scholar
22. Bradford, MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72, 248254.Google Scholar
23. Firestein, GS (1996) Invasive fibroblast-like synoviocytes in rheumatoid arthritis. Passive responders or transformed aggressors? Arthritis Rheum 39, 17811790.Google Scholar
24. Tanaka, Y (2001) The role of chemokines and adhesion molecules in the pathogenesis of rheumatoid arthritis. Drug Today 37, 477484.Google Scholar
25. Baillet, A, Gossec, L, Paternotte, S, et al. (2015) Evaluation of serum interleukin-6 level as a surrogate marker of synovial inflammation and as a factor of structural progression in early rheumatoid arthritis: results from a French national multicenter cohort. Arthritis Care Res 67, 905912.Google Scholar
26. Sommerfelt, RM, Feuerherm, AJ, Jones, K, et al. (2013) Cytosolic phospholipase A2 regulates TNF-induced production of joint destructive effectors in synoviocytes. PLOS ONE 8, e83555.Google Scholar
27. Smolen, JS, Landewe, R, Breedveld, FC, et al. (2010) EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs. Ann Rheum Dis 69, 964975.Google Scholar
28. Yamanishi, Y & Firestein, GS (2001) Pathogenesis of rheumatoid arthritis: the role of synoviocytes. Rheum Dis Clin North Am 27, 355371.Google Scholar
29. Chen, Y & Mattey, DL (2012) Age at onset of rheumatoid arthritis: association with polymorphisms in the vascular endothelial growth factor A (VEGFA) gene and an intergenic locus between matrix metalloproteinase (MMP) 1 and 3 genes. Clin Exp Rheumatol 30, 894898.Google Scholar
30. Egg, D (1984) Concentrations of prostaglandins D2, E2, F2 alpha, 6-keto-F1 alpha and thromboxane B2 in synovial fluid from patients with inflammatory joint disorders and osteoarthritis. Z Rheumatol 43, 8996.Google Scholar
31. Lazarus, M, Kubata, BK, Eguchi, N, et al. (2002) Biochemical characterization of mouse microsomal prostaglandin E synthase-1 and its colocalization with cyclooxygenase-2 in peritoneal macrophages. Arch Biochem Biophys 397, 336341.Google Scholar
32. Liu, Z, Sun, C, Tao, R, et al. (2016) Pyrroloquinoline quinone decelerates rheumatoid arthritis progression by inhibiting inflammatory responses and joint destruction via modulating NF-kappaB and MAPK pathways. Inflammation 39, 248256.Google Scholar
33. Han, Z, Boyle, DL, Chang, L, et al. (2001) c-Jun N-terminal kinase is required for metalloproteinase expression and joint destruction in inflammatory arthritis. J Clin Invest 108, 7381.Google Scholar
34. Tak, PP, Gerlag, DM, Aupperle, KR, et al. (2001) Inhibitor of nuclear factor kappaB kinase beta is a key regulator of synovial inflammation. Arthritis Rheum 44, 18971907.Google Scholar
35. Morel, J & Berenbaum, F (2004) Signal transduction pathways: new targets for treating rheumatoid arthritis. Joint Bone Spine 71, 503510.Google Scholar
36. Barchowsky, A, Frleta, D & Vincenti, MP (2000) Integration of the NF-kappaB and mitogen-activated protein kinase/AP-1 pathways at the collagenase-1 promoter: divergence of IL-1 and TNF-dependent signal transduction in rabbit primary synovial fibroblasts. Cytokine 12, 14691479.Google Scholar
37. Makarov, SS (2001) NF-kappa B in rheumatoid arthritis: a pivotal regulator of inflammation, hyperplasia, and tissue destruction. Arthritis Res 3, 200206.Google Scholar
38. Han, Z, Boyle, DL, Manning, AM, et al. (1998) AP-1 and NF-kappaB regulation in rheumatoid arthritis and murine collagen-induced arthritis. Autoimmunity 28, 197208.Google Scholar
39. Tak, PP & Firestein, GS (2001) NF-kappaB: a key role in inflammatory diseases. J Clin Invest 107, 711.Google Scholar
40. Castejon, ML, Rosillo, MA, Montoya, T, et al. (2017) Oleuropein down-regulated IL-1beta-induced inflammation and oxidative stress in human synovial fibroblast cell line SW982. Food Funct 8, 18901898.Google Scholar
41. Cardeno, A, Sanchez-Hidalgo, M, Aparicio-Soto, M, et al. (2014) Extra virgin olive oil polyphenolic extracts downregulate inflammatory responses in LPS-activated murine peritoneal macrophages suppressing NFkappaB and MAPK signalling pathways. Food Funct 5, 12701277.Google Scholar
42. Iacono, A, Gomez, R, Sperry, J, et al. (2010) Effect of oleocanthal and its derivatives on inflammatory response induced by lipopolysaccharide in a murine chondrocyte cell line. Arthritis Rheum 62, 16751682.Google Scholar
43. Scotece, M, Gomez, R, Conde, J, et al. (2012) Further evidence for the anti-inflammatory activity of oleocanthal: inhibition of MIP-1alpha and IL-6 in J774 macrophages and in ATDC5 chondrocytes. Life Sci 91, 12291235.Google Scholar
44. Rosillo, MA, Sánchez-Hidalgo, M, Castejón, ML, et al. (2017) Extra-virgin olive oil phenols hydroxytyrosol and hydroxytyrosol acetate, down-regulate the production of mediators involved in joint erosion in human synovial cells. J Funct Foods 36, 2733.Google Scholar
45. Rosillo, MA, Sanchez-Hidalgo, M, Gonzalez-Benjumea, A, et al. (2015) Preventive effects of dietary hydroxytyrosol acetate, an extra virgin olive oil polyphenol in murine collagen-induced arthritis. Mol Nutr Food Res 59, 25372546.Google Scholar
46. Vissers, MN, Zock, PL, Roodenburg, AJ, et al. (2002) Olive oil phenols are absorbed in humans. J Nutr 132, 409417.Google Scholar
47. Katsoulieris, EN (2016) The olive leaf extract oleuropein exerts protective effects against oxidant-induced cell death, concurrently displaying pro-oxidant activity in human hepatocarcinoma cells. Redox Rep 21, 9097.Google Scholar
48. Tan, HW, Tuck, KL, Stupans, I, et al. (2003) Simultaneous determination of oleuropein and hydroxytyrosol in rat plasma using liquid chromatography with fluorescence detection. J Chromatogr B Analyt Technol Biomed Life Sci 785, 187191.Google Scholar
Figure 0

Table 1 Phenolic content of polyphenolic extract determined by 1H NMR spectroscopy

Figure 1

Fig. 1 Effect of polyphenolic extract (PE) from extra virgin olive oil (EVOO) on cell viability. The concentrations used in this study (50, 25 and 12·5 µg/ml) did not affect viability of SW982 cells. Cells were treated with PE from EVOO (1·6–200 µg/ml) for 24 h. Cell survival was measured as the percentage of absorbance compared with that obtained in control cells (non-treated cells). SRB, sulforhodamine B. , PE; , dimethylsulfoxide.

Figure 2

Fig. 2 Inhibitory effects of polyphenolic extract (PE) from extra virgin olive oil on the production of matrix metalloproteinase (MMP) and proinflammatory cytokines by SW982 cells. SW982 cells were cultured for 24 h in the presence or absence of PE and stimulated with IL-1β (5 ng/ml). As controls (C), cells were untreated in the absence of IL-1β. Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (not stimulated) (P<0·001). Mean values were significantly different from IL-1β-stimulated control cells: †† P<0·01, ††† P<0·001.

Figure 3

Fig. 3 Polyphenolic extract (PE) from extra virgin olive oil inhibits cyclo-oxygenase-2 (COX-2, ) and microsomal PGE synthase-1 (m-PGES-1, ) protein expressions in IL-1β-stimulated SW982 cells. Cells were untreated or treated with PE (50, 25 or 12·5 µg/ml) for 24 h in the presence of IL-1β (5 ng/ml). Control cells (C) were incubated in the absence of IL-1β. The plots represent band intensity. β-Actin served as an equal loading control for normalisation. Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (not stimulated) (P<0·001). ††† Mean values were significantly different from IL-1β-stimulated control cells (P<0·001).

Figure 4

Fig. 4 Effects of polyphenolic extract (PE) from extra virgin olive oil on the mitogen-activated protein kinase signalling pathway in IL-1β-stimulated SW982 cells. Cells were untreated or treated with PE (50, 25 or 12·5 µg/ml) for 24 h and stimulated with IL-1β (5 ng/ml) for 30 min. Control cells (C) were incubated in the absence of IL-1β. Densitometry was performed following normalisation to the control housekeeping genes (JNK, p38 and ERK1/2). Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (not stimulated) (P<0·001). Mean values were significantly different from IL-1β-stimulated control cells: † P<0·05, †† P<0·01, ††† P<0·001. , Phospho-c-Jun N-terminal kinase; , pp38; , phospho-extracellular signal-regulated kinase 1/2.

Figure 5

Fig. 5 Polyphenolic extract (PE) from extra virgin olive oil pre-treatment prevented IκB-α degradation in IL-1β-stimulated SW982 cells. Cells were untreated or treated with PE (50, 25 or 12·5 µg/ml) for 24 h and stimulated with IL-1β (5 ng/ml) for 6 h. Control cells were incubated in the absence of IL-1β. Densitometry was performed following normalisation to the control (β-actin housekeeping gene). Values are means (n 5), with their standard errors represented by vertical bars. *** Mean values were significantly different from control cells (no stimulated) (P<0·001). Mean values were significantly different from IL-1β-stimulated control cells: † P<0·05 and †† P<0·01.