Mori folium inhibits interleukin-1β-induced expression of matrix metalloproteinases and inflammatory mediators by suppressing the activation of NF-κB and p38 MAPK in SW1353 human chondrocytes

  • Authors:
    • Jin-Woo Jeong
    • Hye Hyeon Lee
    • Ki Won Lee
    • Ki Young Kim
    • Sung Goo Kim
    • Su Hyun Hong
    • Gi-Young Kim
    • Cheol Park
    • Ho Kyoung Kim
    • Young Whan Choi
    • Yung Hyun Choi
  • View Affiliations

  • Published online on: December 23, 2015     https://doi.org/10.3892/ijmm.2015.2443
  • Pages: 452-460
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The pro-inflammatory cytokine interleukin-1β (IL-1β) is known to play a crucial role in the pathogenesis of osteoarthritis (OA) by stimulating several mediators that contribute to cartilage degradation. Mori folium, the leaves of Morus alba L., has long been used in traditional medicine to treat diabetes, protect the liver, and lower blood pressure; however, the role of Mori folium in OA is not yet fully understood. Therefore, in the present study, we investigated whether Mori folium water extract (MF) inhibited the catabolic effects of IL-1β in vitro, and also whether it inhibited the matrix metalloproteinases (MMPs), inducible nitric oxide (NO) synthase (iNOS) and cyclooxygenase-2 (COX-2) through the attenuation of nuclear factor-κB (NF-κB) and mitogen activated protein kinase (MAPK) pathways in SW1353 human chondrocytes. MMP proteins in culture medium were determined using a cytokine‑specific enzyme-linked immunosorbent assay (ELISA). The production of NO and prostaglandin E2 (PGE2) were evaluated using Griess reagent and ELISA. Subsequently, the mRNA and protein levels of MMPs, iNOS, COX-2, NF-κB and MAPKs were examined by RT-qPCR and/or western blot analysis. The results indicate that MF significantly reduced the IL-1β‑induced release of MMP-1 and -13 in SW1353 cells, which was associated with the inhibition of MMP-1 and -13 mRNA and protein expression in a concentration‑dependent manner at concentrations with no cytotoxicity. MF also attenuated the IL-1β-induced production of NO and PGE2, and reduced iNOS and COX-2 expression. Furthermore, we noted that MF markedly suppressed the IL-1β‑induced nuclear translocation of NF-κB, which correlated with the inhibitory effects of MF on inhibitor-κB (IκB) degradation, and the phosphorylation of p38 MAPK was selectively restored by MF upon IL-1β stimulation. These results indicate that MF inhibited the production and expression of MMP-1 and -13 and inflammatory mediators, at least in part, through suppressing the activation of either NF-κB or p38 MAPK in IL-1β-treated SW1353 chondrocytes. Therefore, the novel findings of the present study suggest that MF is a potential therapeutic choice for chondroprotection against the collagen matrix breakdown in the cartilage of diseased tissues, such as those found in patients with arthritic disorders.

Introduction

Currently, osteoarthritis (OA) is a significant clinical issue worldwide, and this situation is expected to worsen with the growth of aging populations (1,2). OA, also known as degenerative arthritis, degenerative joint disease, or osteoarthrosis, is a type of chronic and degenerative joint disease characterized by an increase in protease activity, which results in the degradation of critical extracellular matrix (ECM) proteins as well as pain which results in disabilities (1,2).

In addition to mechanical and genetic factors, chronic and excessive production of inflammatory cytokines and mediators is an important characteristic of OA (2,3). Of the inflammatory cytokines, interleukin-1β (IL-1β) is considered to be one of the most potent catabolic factors in arthritis (35). IL-1β plays a pivotal role in the destruction of the cartilage matrix by upregulating the production of proteases, such as matrix metal-loproteinases (MMPs) and zinc-dependent endopeptidases, which are specifically controlled by the tissue inhibitors of matrix metalloproteinases (TIMPs) (3,6). In healthy cartilage, there is homeostasis between MMPs and TIMPs, which is disturbed in OA (2,6). In the joint cartilage, MMPs are synthesized and secreted by the residing chondrocytes, particularly under arthritic conditions (6,7). Of the various MMPs, collagenases such as MMP-1, -8 and -13, which are also known as collagenase-1, -2 and -3, are of particular importance, as they are elevated in joint disorders and closely related to pathological conditions, such as joint inflammation and joint degenerative diseases (6). Thus, reducing the activity of collagenases may have beneficial results, in the form of chondroprotection against pathological conditions such as OA.

It is also well documented that the expression of collagenases in chondrocytes is regulated by the activation of mitogen-activated protein kinase (MAPK) family members, including c-Jun N-terminal kinase (JNK), p38 MAPK, and extracellular signal-regulated kinase (ERK), and several transcription factors, including nuclear factor-κB (NF-κB), which is known to be activated in human chondrocytes (810). In addition, IL-1β induces excessive inducible nitric oxide (NO) synthase (iNOS) and cyclooxygenase-2 (COX-2) expression in chondrocytes, leading to the elevated production of NO and prostaglandin E2 (PGE2), which accelerate cartilage degradation by inhibiting proteoglycan biosynthesis (11,12). Accordingly, NO and PGE2, which are found in high levels in the synovial fluid of OA patients, are also regarded as beneficial therapeutic targets in the treatment of OA; thus, non-steroidal anti-inflammatory drugs (NSAIDs) have been used for treatment for several years. However, adverse effects, particularly gastrointestinal diseases and cardiovascular risks, are commonly associated with these agents (13,14). Therefore, there is currently considerable interest in the development of new drugs from natural sources, which can be safely used for the prolonged treatment of OA.

For thousands of years, herbal medicines have been used safely and effectively for alleviating and treating various diseases in many countries; there is increasing interest in the pharmacological activity of traditional herbs that are widely used in traditional medicine, and numerous studies support their potential clinical benefits for diseases that are difficult to treat (15,16). Among them, Morus alba L., or white mulberry, a deciduous tree that belongs to the Moraceae family, is widely distributed in Asia (16). In particular, Mori folium, the leaves of Morus alba L., is one of the most widely consumed medicinal herbs in Asia, including Korea, and medications made from Mori folium have been found to be useful in the treatment of metabolic disorders, such as diabetes, hyperlipidemia, and high blood pressure (1719). Previous research has shown that Mori folium exerts diverse pharmacological effects, including anti-microbial (20), anti-inflammatory (21), antioxidant (22), antitumor (23,24), anti-atherosclerotic (25), anti-obesity (17,26), anti-hypotensive (25), and anti-hypoglycemic effects (21), and has the potential to protect the liver (25,27). To the best of our knowledge, no study has been conducted to date on the potential chondroprotective effects and cellular action mechanisms of Mori folium. In this study, as a part of our research on novel biologically active substances for the prevention and treatment of OA from traditional medicinal resources, we investigated the potential of Mori folium water extract (MF) on the production of MMPs and inflammatory mediators in IL-1β-stimulated SW1353 human chondrocyte cells.

Materials and methods

Preparation of MF

The leaves of Morus alba L., Mori folium, were obtained from Bio-Port Korea, Inc. (Busan, Korea) and authenticated by Professor Su Hyun Hong, Department of Biochemistry, Dongeui University College of Korean Medicine (Busan, Korea). The dried leaves were subsequently cut into small pieces, ground into a fine powder, and then boiled with distilled water (50 g/500 ml) for 3 h. The extract was filtered through Whatman no. 3 filter paper (Sigma-Aldrich Chemical Co., St. Louis, MO, USA) twice in order to remove any insoluble materials, and the filtrate was lyophilized and then crushed into a thin powder. The extracts (MF) were dissolved in a 100 mg/ml concentration with distilled water, and the stock solution was then diluted with Dulbecco's modified Eagle's medium (Gibco-BRL, Grand Island, NY, USA) to the desired concentration prior to use.

Cell culture

SW1353 chondrocytes were purchased from the American Type Culture Collection (Manassas, VA, USA). The cells were cultured in Dulbecco's modified Eagle's medium (Gibco-BRL) which contained 10% v/v fetal bovine serum, 100 U/ml penicillin, and 100 mg/ml streptomycin in the presence or absence of MF at 37°C in humidified air with 5% CO2.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

Cell viability was measured using an MTT assay. Briefly, SW1353 cells were seeded in 6-well plates at a density of 2×105 cells/well. After incubation for 24 h, the cells were treated with various concentrations of the MF or 40 ng/ml IL-1β (R&D Systems, Inc., Minneapolis, MN, USA) individually or were pretreated with different concentrations of MF for 1 h before the IL-1β treatment. After 24 h, the medium was removed, and the MTT working solution (0.5 mg/ml; Sigma-Aldrich Chemical Co.) was then added to the culture plates and incubated continuously at 37°C for 2 h. Subsequently, the culture supernatant was removed from the wells, and dimethyl sulfoxide (DMSO; Sigma-Aldrich Chemical Co.) was added to dissolve the formazan crystals. After shaking, the absorbance of each well was measured at 540 nm with a microplate reader (Dynatech Laboratories, Chantilly VA, USA), and the results were expressed as cell viability relative to the untreated control, which was considered 100% viable.

Enzyme-linked immunosorbent assay (ELISA)

The inhibitory effects of MF on the production of MMPs (MMP-1, -2, -3 and -13) and PGE2 were examined using commercial ELISA kits (purchased from R&D Systems, Inc.). Briefly, cells were treated with IL-1β (40 ng/ml) only or alternatively were pretreated with various concentrations of MF for 1 h before IL-1β treatment. After 24 h, the concentrations of the MMPs and PGE2 in the culture medium were determined using selective ELISA kits, according to the manufacturer's instructions.

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Total RNA from the cultured cells was isolated using TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA), according to the manufacturer's instructions. cDNA was synthesized from 1 µg total RNA using AccuPower® RT PreMix (Bioneer, Daejeon, Korea) containing moloney murine leukemia virus reverse transcriptase. RT-generated cDNA was amplified by PCR using selected primers (Table I), which were purchased from Bioneer. After amplification, the PCR reactions were electrophoresed in 1% agarose gels and visualized using ethidium bromide (EtBr; Sigma-Aldrich Chemical Co.) staining. In a parallel experiment, glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as an internal control.

Table I

Sequences of the primer pairs employed in the RT-qPCR reactions.

Table I

Sequences of the primer pairs employed in the RT-qPCR reactions.

Gene nameSequence
MMP-1
 Sense 5′-CTG-TTC-AGG-GAC-AGA-ATG-TGC-3′
 Antisense 5′-TTG-GAC-TCA-CAC-CAT-GTG-TT-3′
MMP-2
 Sense 5′-GTC-AGT-GAG-AAG-GAA-GTG-GAC-TCT-3′
 Antisense 5′-ATG-TTC-TTC-TCT-GTG-ACC-CAG-TC-3′
MMP-3
 Sense 5′-TGC-GTG-GCA-GTT-TGC-TCA-GCC-3′
 Antisense 5′-GAA-TGT-GAG-TGG-AGT-CAC-CTC-3′
MMP-13
 Sense 5′-GGC-TCC-GAG-AAA-TGC-AGT-CTT-TCT-T-3′
 Antisense 5′-ATC-AAA-TGG-GTA-GAA-GTC-GCC-ATG-C-3′
iNOS
 Sense 5′-GTG-AGG-ATC-AAA-AAC-TGG-GG-3′
 Antisense 5′-ACC-TGC-AGG-TTG-GAC-CAC-3′
COX-2
 Sense 5′-TCA-GCC-ACG-CAG-CAA-ATC-CT-3′
 Antisense 5′-GTG-ATC-TGG-ATG-TCA-CG-3′
GAPDH
 Sense5′-CGG AGT CAA CGG ATT TGG TCG TAT-3′
 Antisense5′-AGC CTT CTC CAT GGT GGT GAA GAC-3′

[i] MMP, matrix metalloproteinase; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2.

Protein extraction and western blot analysis

For total protein extraction, the cells were harvested and lysed in lysis buffer [25 mM Tris-Cl (pH 7.5), 250 mM NaCl, 5 mM ethylene-diaminetetraacetic acid (EDTA), 1% Nonidet-P40 (NP-40), 1 mM phenylmethylsulfonyl fluoride (PMSF), and 5 mM dithiothreitol (DTT)] for 1 h. In a parallel experiment, nuclear and cytosolic proteins were prepared using nuclear extraction reagents (Pierce, Rockford, IL, USA), according to the manufacturer's instructions. The insoluble materials were discarded by centrifugation at 13,000 x g for 20 min at 4°C. Protein concentration was measured using a Bio-Rad protein assay kit (Bio-Rad Laboratories, Hercules, CA, USA), according to the manufacturer's instructions. For the western blot analysis, equivalent amounts of proteins were separated by electrophoresis on sodium dodecyl sulfate-polyacrylamide gels (SDS-PAGE) and transferred to nitrocellulose membranes (Schleicher and Schuell, Keene, NH, USA). After blocking with 5% skimmed milk, the membranes were incubated with protein specific antibodies [MMP-1 (1:1,000; ab52631, rabbit monoclonal; Abcam, Cambridge, UK), MMP-2 (1:1,000; SC-10736, rabbit polyclonal; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), MMP-3 (1:1,000; ab52915, rabbit monoclonal; Abcam), MMP-13 (1:1,000; ab51072, rabbit monoclonal; Abcam), iNOS (1:500; SC-7271, mouse monoclonal, Santa Cruz Biotechnology, Inc.), COX-2 (1:500; SC-19999, mouse monoclonal; Santa Cruz Biotechnology, Inc.), NF-κB p65 (1:500; SC-109, rabbit polyclonal; Santa Cruz Biotechnology, Inc.), IκB-α (1:500; SC-371, rabbit polyclonal; Santa Cruz Biotechnology, Inc.), ERK (1:1,000; SC-154, rabbit polyclonal; Santa Cruz Biotechnology, Inc.), p-ERK (1:500; #9106S, mouse monoclonal; Cell Signaling Technology, Inc., Danvers, MA, USA), p38 (1:1,000; SC-535, rabbit polyclonal; Santa Cruz Biotechnology, Inc.), p-p38 (1:500; #9211S, rabbit polyclonal; Cell Signaling Technology, Inc.), JNK (1:1,000; #9252S, rabbit polyclonal; Cell Signaling Technology, Inc.), p-JNK (1:500; #9255S, mouse monoclonal; Cell Signaling Technology, Inc.), Lamin B (1:500; SC-6216, goat polyclonal; Santa Cruz Biotechnology, Inc.) and β-actin (1:1,000; sc-1616, goat polyclonal; Santa Cruz Biotechnology, Inc.)] for 1 h, subsequently incubated with appropriate enzyme-linked secondary antibodies [mouse IgG, HRP-linked whole antibody (NA931) and rabbit IgG, HRP-linked whole antibody (NA934); Amersham Corp., Arlington Heights, IL, USA], and visualized using an enhanced chemiluminescence (ECL) solution (Amersham Corp.), according to the manufacturer's instructions. The primary antibodies were purchased from Santa Cruz Biotechnology, Inc. and Abcam.

Measurement of NO production

The concentration of NO in the culture supernatants was determined by measuring nitrite, a stable oxidation product of nitric oxide, using Griess reagent (Sigma-Aldrich Chemical Co.). The cell culture conditions were the same as those used for the ELISA. Following incubation with IL-1β and/or MF for 24 h, 100 µl of each culture supernatant was mixed with an equal volume of Griess reagent for 10 min at room temperature. Subsequently, absorbance was measured at 540 nm with a microplate reader, and nitrite production was determined by NaNO2 standard curve, as previously described (28).

Immunofluorescence staining

For the detection of the translocation of NF-κB p65, the SW1353 cells were grown on glass coverslips in 6-well plates for 24 h. Cells were pretreated with MF (800 µg/ml) for 1 h prior to stimulation with IL-1β (40 ng/ml). Following incubation for 30 min, the cells were washed twice with phosphate-buffered saline (PBS), fixed with 3.7% paraformaldehyde, treated with 0.2% Triton X-100, and subsequently blocked with 2% bovine serum albumin. The cells were sequentially incubated with an anti-NF-κB p65 antibody (SC-109; Santa Cruz Biotechnology, Inc.) and fluorescein isothiocyanate-conjugated donkey anti-rabbit immunoglobulin G (IgG; Cat. no. 711-001-003; Jackson ImmunoResearch Laboratories, Inc., West Grove, PA, USA). After washing with PBS, the nuclei were stained with 4′6-diamidino-2-phenylindole (DAPI; Sigma-Aldrich Chemical Co.), and fluorescence was visualized using a fluorescence microscope (Carl Zeiss, Jena, Germany), as previously described (29).

Statistical analysis

The values are expressed as the means ± SD. A Student's t-test was used to evaluate the differences between the samples treated with IL-1β only and the samples treated with IL-1β and MF. A p-value <0.05 was considered to indicate a statistically significant difference.

Results

Effects of MF on the viability of SW1353 chondrocytes

To evaluate whether or not MF exerted cytotoxic effects on SW1353 cells, the cells were treated with various concentrations of MF for 24 h. In the MTT assays, we noted that MF concentrations up to 800 µg/ml did not induce cytotoxicity in the SW1353 cells, whereas 1,000 µg/ml MF significantly reduced viability (approximately 70%; Fig. 1A). In a subsequent experiment, when we administered MF to IL-1β-treated SW1353 cells at concentrations below 800 µg/ml, no adverse effects on cell viability were noted (Fig. 1B). Therefore, concentrations of MF up to 800 µg/ml were used in the remaining experiments.

Suppressive effects of MF on IL-1β-induced production of MMP-1 and -13 in SW1353 chondrocytes

Previous studies have suggested that various MMPs are involved in both physiological collagen turnover in articular cartilage and matrix degradation in OA cartilage (2,6). Therefore, to examine the inhibitory effect of MF on IL-1β-induced MMP production, SW1353 cells were stimulated with IL-1β (40 ng/ml) for 24 h in the presence and absence of MF, and subsequently the release of MMPs from SW1353 cells was detected in the culture supernatants using ELISA. As also reported previously (30,31), increased release of MMPs (MMP-1, -2, -3 and -13) was detected in the culture supernatants after stimulation with IL-1β (Fig. 2); however, pretreatment of the SW1353 cells with MF dose-dependently reduced the production of IL-1β-stimulated collagenases MMP-1 and -13, but not MMP-2 (gelatinase A) or -3 (a stromelysin), and levels were comparable to those found in cells not treated with MF.

Inhibition of IL-1β-induced expression of MMP-1 and -13 by MF in SW1353 chondrocytes

To evaluate the effects of MF on IL-1β-induced expression of MMPs in SW1353 cells, cells were treated with IL-1β alone or with different concentrations of MF for 24 h. RT-qPCR and western blot analysis revealed that the levels of all MMPs in the SW1353 cells treated with IL-1β alone markedly increased compared to the levels detected in the untreated cells (Fig. 3); however, pretreatment with MF was found to inhibit the increase in MMP-1 and -13 but not MMP-2 and -3 caused by treatment with IL-1β in a concentration-dependent manner. These results suggest that MF is a potent inhibitor of IL-1β-mediated MMP-1 and -13 transcription in SW1353 cells, whereas it appears not to interfere with MMP-2 and -3 gene expression.

Attenuation of IL-1β-induced NO and PGE2 production by MF in SW1353 chondrocytes

It is well-known that inflammatory mediators, such as NO and PGE2, play key roles in the progression of cartilage destruction in OA (32,33), and thus the effects of MF on the level of NO and PGE2 in IL-1β-stimulated SW1353 cells were examined in this study. To verify the effects of MF on the levels of NO and PGE2 produced by SW1353 cells, supernatants of treated cell cultures were collected and subjected to Griess reagent and ELISA, respectively. Consistent with previous reports (34,35), treating SW1353 cells with IL-1β alone markedly elevated the levels of NO and PGE2 compared to the control group (Fig. 4); however, MF considerably abrogated the release of NO and PGE2 in IL-1β-treated SW1353 cells in a concentration-dependent manner at a range of 200–800 µg/ml.

Reduction of IL-1β-induced iNOS and COX-2 expression by MF in SW1353 chondrocytes

Subsequently, RT-qPCR and western blot analysis were performed to determine whether or not the inhibition of NO and PGE2 production by MF in the IL-1β-stimulated SW1353 cells was associated with decreased levels of iNOS and COX-2 expression. The results indicate that the IL-1β-induced increase in the iNOS and COX-2 mRNA levels was reversed by MF in a concentration-dependent manner (Fig. 5A). In a parallel experiment, the elevated protein levels of iNOS and COX-2 resulting from stimulation with IL-1β were also decreased following pretreatment with MF (Fig. 5B). These results indicate that the reduced expression of iNOS and COX-2 at the transcriptional levels contributed to the inhibitory effect of MF on IL-1β-induced NO and PGE2 production.

Effect of MF on IL-1β-induced nuclear translocation of NF-κB in SW1353 chondrocytes

Previous studies have reported that IL-1β-induced NF-κB activation is involved in upregulating MMPs, iNOS and COX-2 transcriptional activities (36,37). Therefore, we determined whether or not the inhibitory effect of MF on the IL-1β-induced activation of MMPs, iNOS and COX-2 was mediated by the suppression of NF-κB signaling by measuring the nuclear translocation of NF-κB. Western blot analyses revealed that treatment with IL-1β enhanced the nuclear accumulation of NF-κB proteins within 30 min, concomitantly with the degradation of IκB-α in the cytosol (Fig. 6A); however, pretreatment with MF reduced the IL-1β-induced nuclear accumulation of NF-κB and the degradation of IκB-α compared to those in cells treated with IL-1β alone. The immunofluorescence images also revealed that the nuclear translocation of NF-κB p65 was strongly induced after the stimulation of cells with IL-1β, and the shift in NF-κB p65 to the nucleus was completely abolished after pretreating the cells with MF (Fig. 6B). These findings indicate that MF inhibits IL-1β-induced NF-κB activation in SW1353 cells through the suppression of IκB degradation and the nuclear translocation of NF-κB.

Inhibition of IL-1β-induced activation of p38 MAPK by MF in SW1353 chondrocytes

As it is well-known that the MAPK pathway is involved in IL-1β-induced MMP and inflammatory mediator expression (10,38), we subsequently investigated whether or not the inhibitory effect of MF on their expressions was mediated by MAPK signaling cascades. Western blot analysis confirmed that IL-1β treatment enhanced the phosphorylation of JNK, p38 MAPK and ERK without markedly affecting their total protein levels (Fig. 7A). Therefore, the effect of MF on IL-1β-induced phosphorylation of MAPKs in SW1353 cells was evaluated. The results reveal that pretreatment with MF significantly reduced the IL-1β- stimulated activation of p38 MAPK to near the control levels in a concentration-dependent manner, but it had no such effect on the activity of JNK and ERK (Fig. 7B). The results suggest that the suppression of IL-1β-induced p38 MAPK activation by MF is responsible for the observed condroprotective effect of MF on IL-1β-stimulated SW1353 cells.

Discussion

In this study, we investigated whether or not MF inhibited the release and expression of MMPs and inflammatory mediators induced by a representative pro-inflammatory cytokine, IL-1β, in SW1353 chondrocytes. As the data demonstrated, pretreating SW1353 cells with MF effectively suppressed the IL-1β-mediated release of MMP-1, -3, NO and PGE2, as indicated by the attenuation of their corresponding gene expressions, at least in part via blocking NF-κB and p38 MAPK activation.

It is well-known that OA, a heterogeneous and complicated joint disorder, is characterized by the destruction of articular cartilage due to an imbalance between the biosynthesis and degradation of the ECM (1,8). Since MMPs are usually abundantly expressed in joint disorders, and the effect of these proteolytic enzymes is largely due to their ability to degrade the ECM, it is widely accepted that MMPs represent promising pharmacological targets for the treatment of OA (2,6). In particular, collagenases including MMP-1, -8 and -13 provide a suitable microenvironment for the development and progression of OA; moreover, they specifically degrade type II collagen and proteoglycans through other MMPs in the ECM of cartilage (6,9). Of the MMPs, MMP-1 is expressed ubiquitously and is found in a broad range of normal tissue cells, including chondrocytes, whereas MMP-13 is more closely linked to the degradation of type II collagen than MMP-1 or -8 and has long been regarded as a key mediator of cartilage degradation in joint disorders (6,9). In the present study, the OA microenvironment was mimicked by cultured SW1353 cells stimulated with IL-1β. As indicated in the Results, the production of MMP-1 and -13 were significantly promoted in SW1353 cells after stimulation with IL-1β, and MF inhibited MMP-13 release more than MMP-1. Conversely, MF pretreatment effectively suppressed IL-1β-stimulated MMP-1 and MMP-13 release in SW1353 cells by downregulating the overexpression of MMP-1 and -13 but not MMP-2 and -3. Thus, it is likely that treatment with MF has a significant impact on cartilage homeostasis, as it consistently inhibited the IL-1β-induced upregulation of MMP-1 and -13 at the transcriptional level in SW1353 chondrocytes.

In addition to examining the roles of MMPs in OA, elevated levels of inflammatory mediators, such as NO and PGE2, have been previously observed in the cartilage and serum of OA patients (39,40). IL-1β-mediated overproduction of NO has also been reported to act as an important inflammatory mediator that plays a critical role in the pathogenesis of OA through inducing chondrocyte and synoviocyte deaths (41). IL-1β also stimulates the expression of COX-2 to increase the synthesis of PGE2, which is responsible for PGE2 and is implicated in bone resorption and joint pain in OA (42). Moreover, both NO and PGE2 are capable of upregulating the production of MMPs and other inflammatory cytokines (43,44). Therefore, the inhibition of the production of IL-1β-induced inflammatory mediators has been shown to be useful in the treatment of OA. In the present study, we demonstrated that MMP-1 and -13 expression was suppressed by MF, and pretreatment with MF significantly decreased IL-1β-induced NO and PGE2 production by attenuating the expression of their upstream molecules, iNOS and COX-2, on both mRNA and protein levels. These observations indicate that the expression of iNOS and COX-2 was regulated by MF at the transcriptional level and also that MF exerts anti-inflammatory effects on IL-1β-induced inflammation in chondrocytes.

It has previously been noted that NF-κB is the most important transcription factor that regulates the expression of MMPs, COX-2, and iNOS in OA (9,38). Despite the previously demonstrated function of the activator protein-1 in regulating MMP expression (45,46), data demonstrating the NF-κB-mediated induction of MMPs, in particular MMP-1 and -13, indicate that NF-κB is a potential therapeutic target in OA (37,47). Normally, NF-κB is located in the cytoplasm in an inactive form in which the heterodimer is bound to IκB inhibitor proteins. Stimulation with inflammatory molecules, such as IL-1β, leads to phosphorylation and the subsequent degradation of IκB-α, which leads to the nuclear translocation of NF-κB and the promotion of the regulation of transcription of response genes (48,49). Therefore, previous research has focused on the inhibition of MMP and inflammatory mediator expression by blocking the mobilization of NF-κB into the nucleus in chondrocytes. In addition, the high-level expression of MMPs, iNOS, and COX-2 in arthritic joints results from the activation of a tightly regulated and synchronized signaling cascade activated by IL-1β and involving the MAPKs signaling pathway (10,38). MAPKs are activated through the phosphorylation of specific tyrosine and threonine residues by the upstream kinases in response to inflammatory signals (10,11,38). In the present study, it was demonstrated that IL-1β enhances the degradation of IκB-α and enhances the translocation of NF-κB p65 from the cytoplasm to the nucleus; however, pretreatment with MF effectively attenuated the IL-1β-induced degradation of IκB-α and the nuclear translocation of NF-κB p65 in SW1353 cells. It was also observed that following stimulation of SW1353 cells with IL-1β, the activation of the JNK, p39 MAPK and ERK signaling pathways was evident. Interestingly, it was found that MF selectively inhibited IL-1β-induced p38 MAPK activation without markedly affecting ERK and JNK activation. Although other signaling pathways also play an important role in the regulation of OA-related gene expression, the results of our study suggest that the attenuation of NF-κB and p38 activity partly accounts for the inhibitory effects of MF on the gene expression of MMP-1, MMP-13, iNOS and COX-2. As previously mentioned, traditional herbal medicines have become one of the most important alternative treatments for OA. At present, the wider public is also more aware of the significance of prophylactic medications which complement traditional treatments or prevent OA. Therefore, we consider our water extract of Mori folium to be a useful nutritional supplement for OA therapy. In the future, we will further characterize the active compounds from our extract and examined its chondroprotective effect against OA and investigate the molecular mechanisms responsible in detail.

In conclusion, we have demonstrated in the present study that MF effectively suppresses the IL-1β-induced expression of MMP-1, -13, iNOS and COX-2 in SW1353 cells, all of which play a pivotal role in the progression of OA. Furthermore, we have also shown that this effect is mediated, at least in part, through the regulation of NF-κB and p38 signaling pathways. Our findings suggest that MF merits consideration as a therapeutic agent in the treatment and prevention of OA. Therefore, an in-depth study to define the bioavailability of active agent(s) in MF which are capable of exerting cartilage and chondroprotective effects in vivo is warranted.

Acknowledgments

This study was supported by the High Value-added Food Technology Development Program (314043-3), Ministry of Agriculture, Food and Rural Affairs, Republic of Korea.

References

1 

Hardingham T: Extracellular matrix and pathogenic mechanisms in osteoarthritis. Curr Rheumatol Rep. 10:30–36. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Wilusz RE, Sanchez-Adams J and Guilak F: The structure and function of the pericellular matrix of articular cartilage. Matrix Biol. 39:25–32. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Bian Q, Wang YJ, Liu SF and Li YP: Osteoarthritis: genetic factors, animal models, mechanisms, and therapies. Front Biosci (Elite Ed). 4:74–100. 2012. View Article : Google Scholar

4 

Daheshia M and Yao JQ: The interleukin 1beta pathway in the pathogenesis of osteoarthritis. J Rheumatol. 35:2306–2312. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Hwang HS, Park IY, Kim DW, Choi SY, Jung YO and Kim HA: PEP-1-FK506BP12 inhibits matrix metalloproteinase expression in human articular chondrocytes and in a mouse carrageenan-induced arthritis model. BMB Rep. 48:407–412. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Burrage PS, Mix KS and Brinckerhoff CE: Matrix metalloproteinases: role in arthritis. Front Biosci. 11:529–543. 2006. View Article : Google Scholar

7 

Hadler-Olsen E, Fadnes B, Sylte I, Uhlin-Hansen L and Winberg JO: Regulation of matrix metalloproteinase activity in health and disease. FEBS J. 278:28–45. 2011. View Article : Google Scholar

8 

Berenbaum F: Signaling transduction: target in osteoarthritis. Curr Opin Rheumatol. 16:616–622. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Vincenti MP and Brinckerhoff CE: Transcriptional regulation of collagenase (MMP-1, MMP-13) genes in arthritis: integration of complex signaling pathways for the recruitment of gene-specific transcription factors. Arthritis Res. 4:157–164. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Sondergaard BC, Schultz N, Madsen SH, Bay-Jensen AC, Kassem M and Karsdal MA: MAPKs are essential upstream signaling pathways in proteolytic cartilage degradation - divergence in pathways leading to aggrecanase and MMP-mediated articular cartilage degradation. Osteoarthritis Cartilage. 18:279–288. 2010. View Article : Google Scholar

11 

Mengshol JA, Vincenti MP, Coon CI, Barchowsky A and 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 kappaB: differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum. 43:801–811. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Studer R, Jaffurs D, Stefanovic-Racic M, Robbins PD and Evans CH: Nitric oxide in osteoarthritis. Osteoarthritis Cartilage. 7:377–379. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Farkouh ME, Greenberg JD, Jeger RV, Ramanathan K, Verheugt FW, Chesebro JH, Kirshner H, Hochman JS, Lay CL, Ruland S, et al: Cardiovascular outcomes in high risk patients with osteoarthritis treated with ibuprofen, naproxen or lumiracoxib. Ann Rheum Dis. 66:764–770. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Herndon CM: Topical delivery of nonsteroidal anti-inflammatory drugs for osteoarthritis. J Pain Palliat Care Pharmacother. 26:18–23. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Mobasheri A: Intersection of inflammation and herbal medicine in the treatment of osteoarthritis. Curr Rheumatol Rep. 14:604–616. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Wang X, Wei S, Liu T, Pang J, Gao N, Ding D, Duan T, Cao Y, Zheng Y and Zhan H: Effectiveness, medication patterns, and adverse events of traditional chinese herbal patches for osteoarthritis: a systematic review. Evid Based Complement Alternat Med. 2014:3431762014. View Article : Google Scholar : PubMed/NCBI

17 

Ann JY, Eo H and Lim Y: Mulberry leaves (Morus alba L.) ameliorate obesity-induced hepatic lipogenesis, fibrosis, and oxidative stress in high-fat diet-fed mice. Genes Nutr. 10:462015. View Article : Google Scholar : PubMed/NCBI

18 

Jo SP, Kim JK and Lim YH: Antihyperlipidemic effects of stilbenoids isolated from Morus alba in rats fed a high-cholesterol diet. Food Chem Toxicol. 65:213–218. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Kim HG, Jeong HU, Park G, Kim H, Lim Y and Oh MS: Mori folium and mori fructus mixture attenuates high-fat diet-induced cognitive deficits in mice. Based Complement Alternat Med. 2015:3794182015.

20 

Tirupathi RG, Suresh BK, Ujwal KJ, Sujana P, Raoa AV and Sreedhar AS: Anti-microbial principles of selected remedial plants from Southern India. Asian Pac J Trop Biomed. 1:298–305. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Qin J, Fan M, He J, Wu XD, Peng LY, Su J, Cheng X, Li Y, Kong LM, Li RT and Zhao Q: New cytotoxic and anti-inflammatory compounds isolated from Morus alba L. Nat Prod Res. 29:1711–1718. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Khan MA, Rahman AA, Islam S, Khandokhar P, Parvin S, Islam MB, Hossain M, Rashid M, Sadik G, Nasrin S, et al: A comparative study on the antioxidant activity of methanolic extracts from different parts of Morus alba L. (Moraceae). BMC Res Notes. 6:242013. View Article : Google Scholar : PubMed/NCBI

23 

Deepa M, Sureshkumar T, Satheeshkumar PK and Priya S: Antioxidant rich Morus alba leaf extract induces apoptosis in human colon and breast cancer cells by the downregulation of nitric oxide produced by inducible nitric oxide synthase. Nutr Cancer. 65:305–310. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Deepa M and Priya S: Purification and characterization of a novel anti-proliferative lectin from Morus alba L. leaves. Protein Pept Lett. 19:839–845. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Enkhmaa B, Shiwaku K, Katsube T, Kitajima K, Anuurad E, Yamasaki M and Yamane Y: Mulberry (Morus alba L.) leaves and their major flavonol quercetin 3-(6-malonylglucoside) attenuate atherosclerotic lesion development in LDL receptor-deficient mice. J Nutr. 135:729–734. 2005.PubMed/NCBI

26 

Sugimoto M, Arai H, Tamura Y, Murayama T, Khaengkhan P, Nishio T, Ono K, Ariyasu H, Akamizu T, Ueda Y, et al: Mulberry leaf ameliorates the expression profile of adipocytokines by inhibiting oxidative stress in white adipose tissue in db/db mice. Atherosclerosis. 204:388–394. 2009. View Article : Google Scholar

27 

Lee YJ, Choi DH, Kim EJ, Kim HY, Kwon TO, Kang DG and Lee HS: Hypotensive, hypolipidemic, and vascular protective effects of Morus alba L. in rats fed an atherogenic diet. Am J Chin Med. 39:39–52. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Lee H, Pyo MJ, Bae SK, Heo Y, Kim CG, Kang C and Kim E: Improved Therapeutic Profiles of PLA2-Free Bee Venom Prepared by Ultrafiltration Method. Toxicol Res. 31:33–40. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Kim BH, Oh I, Kim JH, Jeon JE, Jeon B, Shin J and Kim TY: Anti-inflammatory activity of compounds isolated from Astragalus sinicus L. in cytokine-induced keratinocytes and skin. Exp Mol Med. 46:e872014. View Article : Google Scholar : PubMed/NCBI

30 

Gebauer M, Saas J, Sohler F, Haag J, Söder S, Pieper M, Bartnik E, Beninga J, Zimmer R and Aigner T: Comparison of the chondrosarcoma cell line SW1353 with primary human adult articular chondrocytes with regard to their gene expression profile and reactivity to IL-1beta. Osteoarthritis Cartilage. 13:697–708. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Toegel S, Wu SQ, Otero M, Goldring MB, Leelapornpisid P, Chiari C, Kolb A, Unger FM, Windhager R and Viernstein H: Caesalpinia sappan extract inhibits IL1β-mediated overexpression of matrix metalloproteinases in human chondrocytes. Genes Nutr. 7:307–318. 2012. View Article : Google Scholar :

32 

Torzilli PA, Tehrany AM, Grigiene R and Young E: Effects of misoprostol and prostaglandin E2 on proteoglycan biosynthesis and loss in unloaded and loaded articular cartilage explants. Prostaglandins. 52:157–173. 1996. View Article : Google Scholar : PubMed/NCBI

33 

Amin AR, Attur M and Abramson SB: Nitric oxide synthase and cyclooxygenases: distribution, regulation, and intervention in arthritis. Curr Opin Rheumatol. 11:202–209. 1999. View Article : Google Scholar : PubMed/NCBI

34 

Wada Y, Shimada K, Sugimoto K, Kimura T and Ushiyama S: Novel p38 mitogen-activated protein kinase inhibitor R-130823 protects cartilage by downregulating matrix metal-loproteinase-1,-13 and prostaglandin E2 production in human chondrocytes. Int Immunopharmacol. 6:144–155. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Park SJ, Cheon EJ and Kim HA: MicroRNA-558 regulates the expression of cyclooxygenase-2 and IL-1β-induced catabolic effects in human articular chondrocytes. Osteoarthritis Cartilage. 21:981–989. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Lu YC, Jayakumar T, Duann YF, Chou YC, Hsieh CY, Yu SY, Sheu JR and Hsiao G: Chondroprotective role of sesamol by inhibiting MMPs expression via retaining NF-κB signaling in activated SW1353 cells. J Agric Food Chem. 59:4969–4978. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Rigoglou S and Papavassiliou AG: The NF-κB signalling pathway in osteoarthritis. Int J Biochem Cell Biol. 45:2580–2584. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Liacini A, Sylvester J, Li WQ, Huang W, Dehnade F, Ahmad M and Zafarullah M: Induction of matrix metalloproteinase-13 gene expression by TNF-alpha is mediated by MAP kinases, AP-1, and NF-kappaB transcription factors in articular chondrocytes. Exp Cell Res. 288:208–217. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Salvatierra J, Escames G, Hernandez P, Cantero J, Crespo E, Leon J, Salvatierra D, Acuña-Castroviejo D and Vives F: Cartilage and serum levels of nitric oxide in patients with hip osteoarthritis. J Rheumatol. 26:2015–2017. 1999.PubMed/NCBI

40 

Miyaura C, Inada M, Suzawa T, Sugimoto Y, Ushikubi F, Ichikawa A, Narumiya S and Suda T: Impaired bone resorption to prostaglandin E2 in prostaglandin E receptor EP4-knockout mice. J Biol Chem. 275:19819–19823. 2000. View Article : Google Scholar : PubMed/NCBI

41 

Chen WP, Wang YL, Tang JL, Hu PF, Bao JP and Wu LD: Morin inhibits interleukin-1β-induced nitric oxide and prostaglandin E2 production in human chondrocytes. Int Immunopharmacol. 12:447–452. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Graham S, Gamie Z, Polyzois I, Narvani AA, Tzafetta K and Tsiridis E, Helioti M, Mantalaris A and Tsiridis E: Prostaglandin EP2 and EP4 receptor agonists in bone formation and bone healing: in vivo and in vitro evidence. Expert Opin Investig Drugs. 18:746–766. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Sasaki K, Hattori T, Fujisawa T, Takahashi K, Inoue H and Takigawa M: Nitric oxide mediates interleukin-1-induced gene expression of matrix metalloproteinases and basic fibroblast growth factor in cultured rabbit articular chondrocytes. J Biochem. 123:431–439. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Tung JT, Arnold CE, Alexander LH, Yuzbasiyan-Gurkan V, Venta PJ, Richardson DW and Caron JP: Evaluation of the influence of prostaglandin E2 on recombinant equine interleukin-1beta-stimulated matrix metalloproteinases 1, 3, and 13 and tissue inhibitor of matrix metalloproteinase 1 expression in equine chondrocyte cultures. Am J Vet Res. 63:987–993. 2002. View Article : Google Scholar : PubMed/NCBI

45 

Barchowsky A, Frleta D and Vincenti MP: 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. 12:1469–1479. 2000. View Article : Google Scholar : PubMed/NCBI

46 

White LA and Brinckerhoff CE: Two activator protein-1 elements in the matrix metalloproteinase-1 promoter have different effects on transcription and bind Jun D, c-Fos, and Fra-2. Matrix Biol. 14:715–725. 1995. View Article : Google Scholar : PubMed/NCBI

47 

Roman-Blas JA and Jimenez SA: NF-kappaB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis. Osteoarthritis Cartilage. 14:839–848. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Itthiarbha A, Phitak T, Sanyacharernkul S, Pothacharoen P, Pompimon W and Kongtawelert P: Polyoxypregnane glycoside from Dregea volubilis extract inhibits IL-1β-induced expression of matrix metalloproteinase via activation of NF-κB in human chondrocytes. In Vitro Cell Dev Biol Anim. 48:43–53. 2012. View Article : Google Scholar

49 

Sunaga T, Oh N, Hosoya K, Takagi S and Okumura M: Inhibitory effects of pentosan polysulfate sodium on MAP-kinase pathway and NF-κB nuclear translocation in canine chondrocytes in vitro. J Vet Med Sci. 74:707–711. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 37 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jeong J, Lee HH, Lee KW, Kim KY, Kim SG, Hong SH, Kim G, Park C, Kim HK, Choi YW, Choi YW, et al: Mori folium inhibits interleukin-1β-induced expression of matrix metalloproteinases and inflammatory mediators by suppressing the activation of NF-κB and p38 MAPK in SW1353 human chondrocytes. Int J Mol Med 37: 452-460, 2016
APA
Jeong, J., Lee, H.H., Lee, K.W., Kim, K.Y., Kim, S.G., Hong, S.H. ... Choi, Y.H. (2016). Mori folium inhibits interleukin-1β-induced expression of matrix metalloproteinases and inflammatory mediators by suppressing the activation of NF-κB and p38 MAPK in SW1353 human chondrocytes. International Journal of Molecular Medicine, 37, 452-460. https://doi.org/10.3892/ijmm.2015.2443
MLA
Jeong, J., Lee, H. H., Lee, K. W., Kim, K. Y., Kim, S. G., Hong, S. H., Kim, G., Park, C., Kim, H. K., Choi, Y. W., Choi, Y. H."Mori folium inhibits interleukin-1β-induced expression of matrix metalloproteinases and inflammatory mediators by suppressing the activation of NF-κB and p38 MAPK in SW1353 human chondrocytes". International Journal of Molecular Medicine 37.2 (2016): 452-460.
Chicago
Jeong, J., Lee, H. H., Lee, K. W., Kim, K. Y., Kim, S. G., Hong, S. H., Kim, G., Park, C., Kim, H. K., Choi, Y. W., Choi, Y. H."Mori folium inhibits interleukin-1β-induced expression of matrix metalloproteinases and inflammatory mediators by suppressing the activation of NF-κB and p38 MAPK in SW1353 human chondrocytes". International Journal of Molecular Medicine 37, no. 2 (2016): 452-460. https://doi.org/10.3892/ijmm.2015.2443