Open Access

Vicenin 3 ameliorates ECM degradation by regulating the MAPK pathway in SW1353 chondrocytes

  • Authors:
    • Yue-Yuan Chen
    • Xiao-Jie Yan
    • Xiao-Hua Jiang
    • Feng-Lai Lu
    • Xue-Rong Yang
    • Dian-Peng Li
  • View Affiliations

  • Published online on: October 20, 2021     https://doi.org/10.3892/etm.2021.10896
  • Article Number: 1461
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Aberrant destruction of the articular extracellular matrix (ECM) has been considered to be one of the pathological features of osteoarthritis (OA) which results in chondrocyte changes and articular cartilage degeneration. The MAPK signaling pathway serves a key role by releasing cartilage‑degrading enzymes from OA chondrocytes. However, the use of MAPK inhibitors for OA is hindered by their potential long‑term toxicity. Vicenin 3 is one of the major components of the Jian‑Gu injection which is effective in the clinical treatment of OA. However, its potential impact on OA remain poorly understood. Therefore, the present study aimed to assess the effects of vicenin 3 on interleukin (IL)‑1β‑treated SW1353 chondrocytes, which mimic the microenvironment of OA. These chondrocytes were pretreated with vicenin 3 (0, 5 and 20 µM) for 1 h and subsequently stimulated with IL‑1β (10 ng/ml) for 24 h. Nitric oxide (NO) production was measured using the Griess reaction, whereas the production of prostaglandin E2 (PGE2), matrix metalloproteinases (MMPs), A disintegrin‑like and metalloproteinase with thrombospondin motifs (ADAMTSs), collagen type II and aggrecan were measured using ELISA. The mRNA expression of MMPs and ADAMTSs were measured using reverse transcription‑quantitative PCR. The protein expression levels of MAPK were measured using western blotting. Vicenin 3 was found to significantly inhibit IL‑1β‑induced production of NO and PGE. Increments in the expression levels of MMP‑1, MMP‑3, MMP‑13, ADAMTS‑4 and ADAMTS‑5 induced by IL‑1β, in addition to the IL‑1β‑induced degradation of collagen type II and aggrecan, were all reversed by vicenin 3 treatment. Furthermore, vicenin 3 suppressed IL‑1β‑stimulated MAPK activation, an effect that was similar to that exerted by SB203580, a well‑known p38 MAPK inhibitor. In conclusion, vicenin 3 may confer therapeutic potential similar to that of the p38 MAPK inhibitor for the treatment of OA.

Introduction

Osteoarthritis (OA) is characterized by the slow, progressive but irreversible erosion of the articular cartilage (1,2). It is also one of the most common musculoskeletal disorders and is a leading cause of disability, which results in the reduction in the quality of life in older adults. Among the various causes of OA, cartilage matrix degradation caused by inflammation is considered to be one of the main causes, which finally leads to the degradation of the extracellular matrix (ECM) (3,4). In particular, the proinflammatory cytokine interleukin (IL)-1β is considered to be a key inducer of OA acceleration by stimulating the release of cartilage degrading enzymes, including matrix metalloproteinases (MMPs), A disintegrin-like and metalloproteinase with thrombospondin motifs (ADAMTSs) and other inflammatory mediators, such as nitric oxide (NO) and prostaglandin E2 (PGE2) (5,6). In addition, elevated levels of IL-1β have been previously detected in the synovial fluid and cartilage tissue of patients with OA (7). The MMP family of matrix-degrading enzymes serves an important role in OA cartilage degradation, of which MMP-13 is a key component in the catabolic processes of OA (8). It hydrolyzes protein structures in the ECM in the articular cartilage, including collagen type II and aggrecan (9). In addition, aggrecanases such as ADAMT-4 and -5 have been shown to induce degradation of the ECM by causing cleavage of proteoglycans and aggrecans within the matrix (10). These repeated cycles of inflammation and catabolism impair chondrocyte homeostasis and promote the irreversible degradation of the ECM in the cartilage, which occurs during OA (11). At present, conservative treatment options for OA mainly involve controlling pain and inflammation using anti-inflammatory agents, including analgesics and nonsteroidal anti-inflammatory drugs, to attenuate articular cartilage injury at the early stages of OA (12,13). However, the majority of these treatment options are only short term that do not alleviate or prevent the progression of OA. In addition, they have been previously reported to cause adverse side effects, including the gastrointestinal hemorrhage (14) and renal impairment (15). By contrast, the other therapeutic strategy available, surgery, can increase risk (such as postoperative infection) and financial burden of the patient and family. Since there is currently no effective treatment strategies available to reverse OA progression, there is a demand to develop an alternative and efficient strategy to alleviate, delay or even reverse the process of OA.

Premna fulva Craib (Verbenaceae), also called ‘Zhangu’, is an ethnomedicine of the Zhuang people (a Chinese minority) that is used for the preparation of Jian-Gu injection (16). Clinically, Jian-Gu injection has been widely applied for the treatment of cervical spondylosis, lumbar sprain and OA (17-19). However, despite its use in the hospital, the mechanism of action has not been characterized in detail. Vicenin 3, which is also called (apigenin 6-C-β-glucopyranosyl-8-C-β-xylopyranoside (Fig. 1A), is a flavone di-C-glycoside purified from Premna fulva Craib that has been reported to be one of the major components in the Jian-Gu injection (20). Recently, flavonoid C-glycosides were reported to exert significant antioxidant, anti-tumor, anti-inflammatory and anti-diabetic activities (21,22). In general, C-glycosyl flavonoids exhibit more potent activity compared with that mediated by their corresponding O-glycosyl flavonoid and aglycone counterparts (23). Previous studies have shown that treatment with flavonoid C-glycosides are associated with a number of health benefits in the prevention and management of diseases such as cancer, diabetes, and cardiovascular disease (23), especially when used as a treatment for antigen-induced arthritis (24,25). However, the underlying mechanism of the effects of vicenin 3 on articular cartilage degeneration during OA remain poorly understood.

MAPK is a super family of intracellular serine-threonine protein kinases that serves as a major node of numerous signal transduction pathways (5). The MAPK pathway mainly includes three signal cascades: JNK, p38 MAPK and ERK (26,27). Previous studies have shown that the p38 signaling pathway serves a key role in the progression of several human diseases including cardiovascular disease, diabetes and cancer, especially in the development of OA (28,29). In addition, activation of the p38 MAPK signaling pathway may increase the expression of proinflammatory cytokines (IL-6, TNF-α), chemokines (CCL3, CCL5), MMPs (MMP-3, MMP-13) and signaling enzymes (iNOS, COX-2) in human OA chondrocytes (30). Blocking the p38 MAPK pathway with a p38 inhibitor has been revealed to inhibit chondrocyte apoptosis and reduce the production of inflammatory cytokines to prevent the recruitment of inflammatory cells, which may alleviate bone and cartilage degradation (29). Therefore, in the present study, the effects of vicenin 3 on chondrocytes in an in vitro model of OA was investigated. In addition, the underlying mechanism of these effects, which particular focus on the MAPK signaling cascade, was also assessed.

Materials and methods

Reagents

Vicenin 3 (purity ≥98%) was isolated from Premna fulva Craib by recycling counter-current chromatography in Guangxi Key Laboratory of Functional Phytochemicals Research and Utilization as previously described (20). FBS and DMEM were purchased from Gibco (Thermo Fisher Scientific, Inc.). Recombinant human IL-1β was purchased from R&D Systems, Inc. The Cell Counting Kit-8 (CCK-8) was purchased from Beyotime Institute of Biotechnology. The Griess reagent for NO estimation was obtained from Nanjing Jiancheng Bioengineering Institute. The ELISA kits for aggrecan (cat. no. E-EL-H0294c), collagen type II (cat. no. E-EL-H0777c), PGE2 (cat. no. E-EL-0034c), MMP-1 (cat. no. E-EL-H6073c) and MMP-13 (cat. no. E-EL-H0134c) were obtained from Elabscience Biotechnology Co., Ltd. whereas the MMP-3 (cat. no. CSB-E04677h) ELISA kit was purchased from Cusabio Biotech Co., Ltd. TRIzol reagent was purchased from Ambion; Thermo Fisher Scientific, Inc. HiScript QuantiTect Reverse Transcription kit was obtained from Vazyme Biotech Co., Ltd. Antibodies against JNK (#9252), phosphorylated (p-)-JNK (#9251), ERK (#9102), p-ERK (#9101), p38 (#8690), p-p38 (#9211) and GAPDH (#5174) were purchased from Cell Signaling Technology Inc. The p38 inhibitor SB203580 was obtained from MedChemExpress to block the activation of p38. Cultured chondrocytes were pretreated with vicenin 3 (20 µM) or SB203580 (10 µM) at 37˚C for 1 h and then treated with 10 ng/ml IL-1β at 37˚C for 24 h, western blot was used to detect the expression of p38, p-p38; PGE2, MMP-1, MMP-3, MMP-13, aggrecan and collagen type II were measured in the culture medium using an ELISA kit.

Cell culture

The human chondrocyte cell line SW1353 was acquired from the Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences. They were cultured in DMEM containing 10% FBS and 2 mM glutamine, 100 U/ml penicillin and 100 µg/ml streptomycin at 37˚C in humidified atmosphere with 5% CO2.

Cell viability assay

The effects of vicenin 3 (6.25-100 µM) on chondrocytes were determined using a CCK-8 kit according to the manufacturer's protocols. The SW1353 cells were seeded into 96-well plates (5,000 cells/well) at 37˚C for 12 h and then treated with various concentrations of either vicenin 3 or IL-1β (10 ng/ml) alone at 37˚C for 24 h. In an additional protocol, the cells were also pretreated with vicenin 3 (5 and 20 µM) at 37˚C for 1 h before IL-1β treatment (at 37˚C for 24 h). Subsequently, 10 µl CCK-8 was added to each well and incubated at 37˚C for 4 h. Absorbance in each well was then measured at 450 nm using a microplate reader (Leica Microsystems GmbH). All experiments were performed in triplicate.

NO measurement and ELISA

The SW1353 cells (5,000 cells/well) were pretreated with different concentrations (5 and 20 M) of vicenin 3 at 37˚C for 1 h, followed by treatment with IL-1β (10 ng/ml) at 37 ˚C for 24 h, before the levels of NO accumulation in the culture medium were determined using the Griess reaction according to the manufacturer's protocols.

The protein levels of PGE2, MMP-1, MMP-3, MMP-13, aggrecan and collagen type II released from the chondrocyte cultured medium under the same conditions were evaluated using the ELISA kits according to the manufacturer's protocols. All assays were performed in triplicate.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was isolated from chondrocytes using TRIzol reagent (Ambion; Thermo Fisher Scientific, Inc., 15596-026) according to the manufacturer's protocol. The concentration was spectrophotometrically measured at 260 nm using NanoDrop 2000 (Thermo Fisher Scientific, Inc.). The A260/A280 ratio was calculated to test the quality and purity of the RNA samples. First-strand cDNA was synthesized from 3 µg total RNA with the HiScript QuantiTect RT kit (Vazyme Biotech Co., Ltd, R101-01/02) at 25˚C for 5 min, 50˚C for 15 min, 85˚C for 5 min and at 4˚C for 10 min. qPCR was performed in the Bio-Rad CFX96 Touch Real-Time PCR Detection System (Bio Rad Laboratories, Inc., 1855196) under the following thermocycling conditions: 10 min at 95˚C, followed by 40 cycles of 15 sec at 95˚C and 1 min at 60˚C. The level of target mRNA was normalized to the level of GAPDH and then compared with the control. mRNA expression was quantified using the 2-∆∆Cq method (31). This assay was performed in triplicate. The primer sequences of the targeted genes are listed in Table I.

Table I

Primer sequences used for reverse transcription-quantitative PCR in the present study.

Table I

Primer sequences used for reverse transcription-quantitative PCR in the present study.

GenePrimerSequence (5'-3')PCR products
GAPDHForward CAGCCTCAAGATCATCAGCA106 bp
 Reverse TGTGGTCATGAGTCCTTCCA 
MMP-1Forward CCAGGTATTGGAGGGGATG273 bp
 Reverse GTCACACGCTTTTGGGGTT 
MMP-13Forward CCCAACCCTAAACATCCAA147 bp
 Reverse AAACAGCTCCGCATCAACC 
MMP-3Forward TTCCTTGGATTGGAGGTGAC248 bp
 Reverse AGCCTGGAGAATGTGAGTGG 
ADAMTS-4Forward CAATCCTGTCAGCTTGGTGG162 bp
 Reverse GCTGTGTCAAAGTGGTCAGG 
ADAMTS-5Forward CTGCCACCACACTCAAGAAC208 bp
 Reverse TGGAGGCCATCGTCTTCAAT 

[i] MMP, matrix metalloproteinase; ADAMTS, A disintegrin-like and metalloproteinase with thrombospondin motifs.

Western blot analysis

Western blotting was used to detect the protein expression and phosphorylation of JNK, ERK and p38. Total proteins were extracted from chondrocytes using RIPA (Beyotime Institute of Biotechnology) and PMSF (Shanghai Aladdin Biochemical Technology Co., Ltd) buffers. Lysates were sonicated (sonicated time: 40 sec, interval time 60 sec, power/frequency: 100 W/16 KHz) three times on finely-crushed dry ice for 5 min and centrifuged at 119 (x100 g) for 30 min at 4˚C. Protein concentration was determined using the BCA protein assay kit. Protein samples (40 µg/lane) was then separated by SDS-PAGE (10%) and transferred onto PVDF membranes. The membranes were blocked with 5% non-fat dry milk for 2 h at room temperature and subsequently washed three times for 5 min in TBS with 0.1% Tween-20 (TBST). The membranes were incubated sequentially with primary antibodies (1:1,000) against JNK, p-JNK, ERK, p-ERK, p38, p-p38 and GAPDH overnight at 4˚C. After washing three times with TBST for 5 min, the membranes were incubated with HRP-conjugated secondary antibodies (BA1054, Wuhan Boster Biological Technology Co., Ltd.; 1:50,000) at room temperature for 2 h. Finally, proteins were visualized using the Immobilon Western Chemiluminescent HRP substrate (cat. no. WBKLS0500; EMD Millipore), and then the blots were analyzed using the ChemiDoc XRS system (Bio Rad Laboratories, Inc.). The density of the protein bands was quantified using Image Lab 3.0 software (Bio-Rad Laboratories, Inc.). The relative protein levels of the target protein in each sample were determined before the levels of phosphorylated protein were normalized to those of their corresponding total protein.

Statistical analysis

The results are expressed as the mean ± standard deviation. All assays were performed in triplicate. One-way analysis of variance followed by the Dunnett's test was used for multiple comparisons using the GraphPad Prism 8.0 software (Graphpad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of vicenin 3 on SW1353 chondrocyte cytotoxicity

The potential cytotoxicity of vicenin 3 on SW1353 chondrocytes was tested using CCK-8 assay. Vicenin 3 at the concentration range of 6.25-25 µM did not exert cytotoxic effects on the cells. However, significant reductions in cell viability was observed at concentrations >50 µM (Fig. 1B). In addition, 5 and 20 µM vicenin 3 did not alter chondrocyte viability in combination with inflammatory stimuli (10 ng/ml IL-1β; Fig. 1C). Therefore, 5 and 20 µM vicenin 3 were chosen for subsequent experiments.

Effect of vicenin 3 on NO and PGE2 production in chondrocytes

The possible effects of vicenin 3 on IL-1β-induced NO and PGE2 production in SW1353 cells were next investigated. SW1353 cells were pretreated with vicenin 3 (5 and 20 µM) for 1 h before subsequent IL-1β (10 ng/ml) stimulation for 24 h. NO concentration in the cell suspension was then determined using the Griess reaction whereas PGE2 levels were measured using ELISA. As shown in Fig. 2, the levels of NO and PGE2 in the supernatant were significantly increased after IL-1β treatment, which were significantly reversed by vicenin 3 in a dose-dependent manner.

Effect of vicenin 3 on expression and secretion of MMP-1, MMP-3, MMP-13, ADAMTS-4 and ADAMTS-5 in IL-1β-induced SW1353 chondrocytes

ECM degradation by matrix degrading enzymes, such as MMPs is a characteristic feature of OA (32). Therefore, the effect of vicenin 3 on the secretion levels of MMP-1, MMP-3 and MMP-13 was evaluated in IL-1β-induced SW1353 cells. As shown in Fig. 3A and B, IL-1β significantly upregulated the secretion and mRNA expression levels of MMP-1, MMP-3 and MMP-13, whilst treatment with the higher dose of vicenin 3 (20 µM) resulted in the significant reversal of this IL-1β-induced increase in MMP-1, MMP-3 and MMP-13 mRNA expression and protein secretion. Although 5 µM vicenin 3 did not significantly affect the secretion of MMP-3, it did significantly reverse the IL-1β-induced increase in MMP-1, MMP-3 and MMP-13 mRNA expression and MMP-1 and MMP-13 secretion. In addition, treatment with vicenin 3 significantly reversed the IL-1β-induced increase in ADAMTS-4 and ADAMTS-5 mRNA expression (Fig. 3C and D). These results suggest that vicenin 3 reduced the IL-1β-induced expression of matrix degrading enzymes in SW1353 chondrocytes.

Effect of vicenin 3 on collagen type II and aggrecan secretion by IL-1β-induced SW1353 chondrocytes

The effects of vicenin 3 on the secretion of collagen type II and aggrecan, major components of ECM in the articular cartilage (33), by IL-1β-stimulated SW1353 chondrocytes was next assessed using ELISA. As shown in Fig. 4, collagen type II and aggrecan levels were significantly decreased by IL-1β, whereas pre-treatment with vicenin 3 (5 and 20 µM) significantly prevented this IL-1β-induced decrease in the secretion of collagen type II. In addition, vicenin 3 (20 µM) significantly prevented the IL-1β-induced decrease in aggrecan expression. These results suggest that vicenin 3 has potent cartilage matrix-protective effects by suppressing IL-1β-induced degradation of collagen type II and aggrecan in SW1353 chondrocytes.

Effect of vicenin 3 on MAPK activation in IL-1β-induced SW1353 chondrocytes

Since IL-1β-induced MMP expression was previously found to be mediated by members of the MAPK kinase family, such as ERK, JNK, and p38 MAPK (34), the potential effects of vicenin 3 on the activity of these three MAPK kinases were next examined. In IL-1β-stimulated chondrocytes, the phosphorylation of ERK, JNK and p38 MAPK was found to be activated compared with that in the control group but their corresponding total protein expression levels were not significantly affected (Fig. 5). However, pretreatment with vicenin 3 significantly reversed the IL-1β-induced phosphorylation of ERK, JNK and p38 MAPK (Fig. 5). These results suggest that the suppressive effects of vicenin 3 on the IL-1β-induced expression of cartilage degrading enzymes was mediated through MAPK signaling in SW1353 chondrocytes.

Vicenin 3 ameliorates the degradation of ECM in IL-1β-treated SW1353 chondrocytes by blocking the p38 signaling pathway

Western blot analysis demonstrated that vicenin 3 exerted a significant inhibitory influence on IL-1β-induced activation of ERK, JNK and p38 MAPK (Fig. 5). p38 MAPK has been reported to be associated with OA and has been considered to be a target for drug-mediated modulation in OA (35,36). Therefore, it was investigated whether vicenin 3 and the p38MAPK inhibitor (SB203580) had similar efficacy in IL-1β-stimulated SW1353 chondrocytes. As shown in Fig. 6, vicenin 3 was also found to exert similar effects compared with those mediated by SB203580, a p38 inhibitor, on blocking p38 activation in IL-1β-stimulated SW1353 chondrocytes. Subsequently, in IL-1β-incubated SW1353 chondrocytes, which exhibited significantly higher protein secretion levels of PGE2, MMP-1, MMP-3 and MMP-13 compared with those in the control group, treatment with vicenin 3 or SB203580 significantly reduced the secretion of these enzymes (Fig. 6). By contrast, analysis of ELISA results revealed that the secretion levels of collagen type II by SW1353 chondrocytes was significantly decreased by IL-1β, which was significantly reversed by vicenin 3 or SB203580 treatment (Fig. 6). However, vicenin 3 exhibited a more potent inhibitory effect on the secretion of aggrecan compared with that by SB203580 (Fig. 6). Therefore, these results demonstrated that vicenin 3-related amelioration of ECM degradation by SW1353 chondrocytes was associated at least in part with the blocking of p38 MAPK signaling.

Discussion

OA is a degenerative joint disease that is frequently observed among the elderly and exerts great burden on the society and economy due to the lack of therapeutic methods (37). The underlying pathological mechanism of OA remains to be fully elucidated. However, it is generally accepted that OA is caused at least in part by degradation of the cartilaginous matrix because of the inflammation-induced upregulation of catabolism in chondrocytes (4). A number of studies have highlighted the importance of chondrocyte function in the development of OA, since they are the only type of cells in the articular cartilage and are responsible for maintaining the anabolic and catabolic balance in the cartilage ECM (38,39). In the present study, a protective effect of vicenin 3 on chondrocyte was found, which was mediated through the inhibition of various pathological factors affecting OA, including NO-induce stress, degradation of articular ECM and the expression of proinflammatory cytokines and mediators. This effect was found to be due to the inhibition of MAPK signaling. Application of various p38 inhibitors, including synthetic pyridine imidazole small molecular compounds, for OA has conferred disappointing results owing to their long-term toxicities, such as gastrointestinal disorders, dizziness and rashes (40). Vicenin 3 is a natural flavone di-C-glycoside that can be purified from Premna fulva Craib (20). In the present study, it was found to suppress IL-1β-stimulated MAPK activation in a similar manner to that exerted by the p38 MAPK inhibitor SB203580. Taken together, these findings suggest that vicenin 3 may have therapeutic potential as ap38 MAPK inhibitor for the treatment of OA.

Inflammation is a potent catabolic inducer of the cartilage during OA pathogenesis, which apparently disrupts tissue maintenance and functionality as a result of chondrocyte apoptosis and hinders the regenerative ability of the joint (41). In particular, IL-1β has been found to trigger the initiation and acceleration of ECM degradation by chondrocytes, by stimulating the release of cartilage-degrading enzymes in the MMP and ADAMTS families and other catabolic factors, including NO and PGE2(6). This consequently contributes to chondrocyte dysfunction (6). As a mediator of the inflammatory response, NO and PGE2 is involved in a number of inflammatory degenerative diseases, including OA and chronic neuronal diseases (42,43). In particular, the increase of inflammatory mediators such as NO and PGE2 by IL-1β alleviate OA pathogenesis (44). Therefore, inhibition of these inflammatory mediators would be of therapeutic potential by assessing the effect of potential anti-inflammatory drugs on the joint degenerative process induced by inflammation. In the present study, the production of NO and PGE2 were increased by IL-1β treatment, but pre-treatment with vicenin 3 reverse this effect.

MMPs belong to a family of proteolytic enzymes and can facilitate OA development by mediating the irreversible degradation of collagen type II and proteoglycan in the cartilage (4). In particular, MMP-13 is considered to be a key player in the catabolic processes during OA because of its ability to degrade collagen type II and proteoglycan (9). In addition, ADAMTS-4 and -5 are considered to be the main enzymes responsible for the degradation of aggrecan and associated with OA (10). Therefore, the level of cartilage matrix components (collagen type II and proteoglycan) can be used as indicators for assessing the progression of cartilage destruction. In the present study, vicenin 3 inhibited the IL-1β-induced secretion and mRNA expression of MMP-1, MMP-3, MMP-13, ADAMTS-4 and ADAMTS-5 in SW1353 chondrocytes. In addition, vicenin 3 reversed the IL-1β-induced reduction of collagen type II and aggrecan secretion. These results suggest that vicenin 3 has a protective effect on chondrocytes to inhibit the development of OA following IL-1β-related induction.

Numerous intracellular signaling systems have been reported to participate in regulating OA (45,46). Among these, MAPKs have been extensively studied, where they were reported to be involved in the development and progression of OA (47). In addition, activation of p38MAPK has been revealed to be associated with cartilage collagen degradation, chondrocyte apoptosis and the inflammation process in OA (35,48). Therefore, p38 MAPK was proposed to be a therapeutic target for OA (36). In the present study, vicenin 3 was able to reduce the phosphorylation of MAPKs (ERK1/2, JNK and p38) in vitro induced by IL-1β stimulation. These results suggest that vicenin 3 may have the therapeutic potential for the treatment of OA by blocking MAPK signaling pathways.

In conclusion, IL-1β-induced production of inflammatory factors NO and PGE2 and cartilage-degrading enzymes MMP-1, MMP-3, MMP-13, ADAMTS-4 and ADAMTS-5 by SW1353 chondrocytes were found to be significantly reversed by vicenin 3 (especially at a concentration of 20 µM) in the present study. Furthermore, vicenin 3 reversed the reduction in the production of ECM components collagen type II and aggrecan by SW1353 chondrocytes following IL-1β treatment. These findings suggest that vicenin 3, which is a main component of Jian-Gu injection, exerts protective effects against IL-1β-treated OA development.

Despite determining the discovery of the protective effects of vicenin 3 and the associated underlying mechanism of action using an in vitro OA model, the present study also has limitations. Although comparison to a previous study showed that SW1353 human chondrosarcoma is commensurate with the in vitro primary experimental human chondrocyte system (49), it would be more valuable to use chondrocytes isolated from patients with OA. In addition, further validation studies, such as the recommended optimal dose for use in vivo, are also required. Vicenin 3 is one of the five structurally related flavone C-glycoside components reported to be present in the Jian-Gu injection (20). Therefore, further studies are required to clarify the protective effects of other flavone C-glycoside compounds on chondrocytes.

Acknowledgements

Not applicable.

Funding

The present study was financially supported by the Natural Science Foundation of Guangxi Zhuang Autonomous Region (grant no. 2018GXNSFAA294031), Key-Area Research and Development Program of Guangdong Province (grant no. 2020B1111110003)and Guangxi Innovation-Driven Development Special Project (grant no. GuikeAA18118015).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YYC and DPL designed this study. XJY and XHJ performed mechanistic and phenotypic experiments, respectively. XJY, XRY, and YYC analyzed the data and interpreted the results of the experiments. FLL analyzed the data and contributed to manuscript revision. DPL and FLL confirmed the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Di Nicola V: Degenerative osteoarthritis a reversible chronic disease. Regen Ther. 15:149–160. 2020.PubMed/NCBI View Article : Google Scholar

2 

Hunter DJ and Bierma-Zeinstra S: Osteoarthritis. Lancet. 393:1745–1759. 2019.PubMed/NCBI View Article : Google Scholar

3 

Blasioli DJ and Kaplan DL: The roles of catabolic factors in the development of osteoarthritis. Tissue Eng Part B Rev. 20:355–363. 2014.PubMed/NCBI View Article : Google Scholar

4 

Guilak F, Nims RJ, Dicks A, Wu CL and Meulenbelt I: Osteoarthritis as a disease of the cartilage pericellular matrix. Matrix Biol. 71-72:40–50. 2018.PubMed/NCBI View Article : Google Scholar

5 

Zeng L, Rong XF, Li RH and Wu XY: Icariin inhibits MMP-1, MMP-3 and MMP-13 expression through MAPK pathways in IL-1β-stimulated SW1353 chondrosarcoma cells. Mol Med Rep. 15:2853–2858. 2017.PubMed/NCBI View Article : Google Scholar

6 

Liu-Bryan R and Terkeltaub R: Emerging regulators of the inflammatory process in osteoarthritis. Nat Rev Rheumatol. 11:35–44. 2015.PubMed/NCBI View Article : Google Scholar

7 

Matejova JP, Spakova T, Harvanova D, Lacko M, Filip V, Sepitka R, Mitro I and Rosocha J: A Preliminary Study of Combined Detection of COMP, TIMP-1, and MMP-3 in Synovial Fluid: Potential Indicators of Osteoarthritis Progression. Cartilage: Aug 4, 2020 (Epub ahead of print).

8 

Murphy G, Knäuper V, Atkinson S, Butler G, English W, Hutton M, Stracke J and Clark I: Matrix metalloproteinases in arthritic disease. Arthritis Res. 4 (Suppl 3):S39–S49. 2002.PubMed/NCBI View Article : Google Scholar

9 

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

10 

Majumdar MK, Askew R, Schelling S, Stedman N, Blanchet T, Hopkins B, Morris EA and Glasson SS: Double-knockout of ADAMTS-4 and ADAMTS-5 in mice results in physiologically normal animals and prevents the progression of osteoarthritis. Arthritis Rheum. 56:3670–3674. 2007.PubMed/NCBI View Article : Google Scholar

11 

Dancevic CM and McCulloch DR: Current and emerging therapeutic strategies for preventing inflammation and aggrecanase-mediated cartilage destruction in arthritis. Arthritis Res Ther. 16:429–440. 2014.PubMed/NCBI View Article : Google Scholar

12 

Zhang Z, Huang C, Jiang Q, Zheng Y, Liu Y, Liu S, Chen Y, Mei Y, Ding C, Chen M, et al: Guidelines for the diagnosis and treatment of osteoarthritis in China (2019 edition). Ann Transl Med. 8:1213. 2020.PubMed/NCBI View Article : Google Scholar

13 

Latourte A, Kloppenburg M and Richette P: Emerging pharmaceutical therapies for osteoarthritis. Nat Rev Rheumatol. 16:673–688. 2020.PubMed/NCBI View Article : Google Scholar

14 

Li Z, Meng D, Li G, Xu J, Tian K and Li Y: Celecoxib Combined with Diacerein Effectively Alleviates Osteoarthritis in Rats via Regulating JNK and p38MAPK Signaling Pathways. Inflammation. 38:1563–1572. 2015.PubMed/NCBI View Article : Google Scholar

15 

Konijnenbelt-Peters J, van der Heijden C, Ekhart C, Bos J, Bruhn J and Kramers C: Metamizole (Dipyrone) as an Alternative Agent in Postoperative Analgesia in Patients with Contraindications for Nonsteroidal Anti-Inflammatory Drugs. Pain Pract. 17:402–408. 2017.PubMed/NCBI View Article : Google Scholar

16 

Pharmacopoeia Committee of the Ministry of Health of the People's Republic of China: Jian gu zhu she ye (Jian-gu injection). In: Prescription preparation of Chinese Medicine. Vol 14. Pharmacopoeia Committee of the Ministry of Health of the People's Republic of China, pp59-60, 1997 (In Chinese).

17 

Ma YH, Sun Q, Yang L, Zhang J and Zhai WT: Effect of Jiangu injection by shoulder injection combined with arthroscopic subacromial space decompression on shoulder function recovery in patients with subacromial impingement syndrome. Mod J Integr Tradi Chin West Med. 27:2415–2418. 2018.(In Chinese).

18 

Chen Z, Hao LD and Zhong W: Clinical efficacy of pain spot injection with Jiangu injection in acute lumbar sprain. J Guandong Med Coll. 34:295–297. 2016.(In Chinese).

19 

Wang HQ, Huang YR and Qin XL: Treatment of 40 cases of knee osteoarthritis with acupoint injection of Jiangu injection. J Guangxi Trad Chin Med Univ. 14:23–24. 2011.(In Chinese).

20 

Chen Y, Yan X, Lu F, Jiang X, Friesen JB, Pauli GF, Chen SN and Li DP: Preparation of flavone di-C-glycoside isomers from Jian-Gu injection (Premna fulva Craib.) using recycling counter-current chromatography. J Chromatogr A. 1599:180–186. 2019.PubMed/NCBI View Article : Google Scholar

21 

Xue Y, Liu Y, Xie Y, Cong C, Wang G, An L, Teng Y, Chen M and Zhang L: Antioxidant activity and mechanism of dihydrochalcone C-glycosides: Effects of C-glycosylation and hydroxyl groups. Phytochemistry. 179(112393)2020.PubMed/NCBI View Article : Google Scholar

22 

Chen GL, Fan MX, Wu JL, Li N and Guo MQ: Antioxidant and anti-inflammatory properties of flavonoids from lotus plumule. Food Chem. 277:706–712. 2019.PubMed/NCBI View Article : Google Scholar

23 

Xiao J, Capanoglu E, Jassbi AR and Miron A: Advance on the Flavonoid C-glycosides and Health Benefits. Crit Rev Food Sci Nutr. 56 (Suppl 1):S29–S45. 2016.PubMed/NCBI View Article : Google Scholar

24 

Khan MA, Khurana N, Ahmed RS, Umar S, Md G, Sarwar AH, Alam Q, Kamal MA and Ashraf GM: Chemokines: A Potential Therapeutic Target to Suppress Autoimmune Arthritis. Curr Pharm Des. 25:2937–2946. 2019.PubMed/NCBI View Article : Google Scholar

25 

Garcia EF, de Oliveira MA, Candido LCM, Coelho FM, Costa VV, Queiroz-Junior CM, Boff D, Amaral FA, de Souza DG, Teixeira MM, et al: Effect of the Hydroethanolic Extract from Echinodorus grandiflorus Leaves and a Fraction Enriched in Flavone-C-Glycosides on Antigen-Induced Arthritis in Mice. Planta Med. 82:407–413. 2016.PubMed/NCBI View Article : Google Scholar

26 

Keshet Y and Seger R: The MAP kinase signaling cascades: a system of hundreds of components regulates a diverse array of physiological functions. Methods Mol Biol. 661:3–38. 2010.PubMed/NCBI View Article : Google Scholar

27 

Thalhamer T, McGrath MA and Harnett MM: MAPKs and their relevance to arthritis and inflammation. Rheumatology (Oxford). 47:409–414. 2008.PubMed/NCBI View Article : Google Scholar

28 

Wang H, Shan XB and Qiao YJ, Wang H, Shan XB and Qiao YJ: PDK2 promotes chondrogenic differentiation of mesenchymal stem cells by upregulation of Sox6 and activation of JNK/MAPK/ERK pathway. Braz J Med Biol Res. 50(e5988)2017.PubMed/NCBI View Article : Google Scholar

29 

Sun HY, Hu KZ and Yin ZS: Inhibition of the p38-MAPK signaling pathway suppresses the apoptosis and expression of proinflammatory cytokines in human osteoarthritis chondrocytes. Cytokine. 90:135–143. 2017.PubMed/NCBI View Article : Google Scholar

30 

Feng Z, Zheng W, Li X, Lin J, Xie C, Li H, Cheng L, Wu A and Ni W: Cryptotanshinone protects against IL-1β-induced inflammation in human osteoarthritis chondrocytes and ameliorates the progression of osteoarthritis in mice. Int Immunopharmacol. 50:161–167. 2017.PubMed/NCBI View Article : Google Scholar

31 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

32 

Visse R and Nagase H: Matrix metalloproteinases and tissue inhibitors of metalloproteinases: Structure, function, and biochemistry. Circ Res. 92:827–839. 2003.PubMed/NCBI View Article : Google Scholar

33 

Sharma S, Vazquez-Portalatin N, Calve S and Panitch A: Biomimetic molecules lower catabolic expression and prevent chondroitin sulfate degradation in an osteoarthritic ex vivo model. ACS Biomater Sci Eng. 2:241–250. 2016.PubMed/NCBI View Article : Google Scholar

34 

Yao M, Wang X, Zhao Y, Wang X and Gao F: Expression of MMPs is dependent on the activity of mitogen-activated protein kinase in chondrosarcoma. Mol Med Rep. 15:915–921. 2017.PubMed/NCBI View Article : Google Scholar

35 

Kühn K, Shikhman AR and Lotz M: Role of nitric oxide, reactive oxygen species, and p38 MAP kinase in the regulation of human chondrocyte apoptosis. J Cell Physiol. 197:379–387. 2003.PubMed/NCBI View Article : Google Scholar

36 

Joos H, Albrecht W, Laufer S and Brenner RE: Influence of p38MAPK inhibition on IL-1beta-stimulated human chondrocytes: A microarray approach. Int J Mol Med. 23:685–693. 2009.PubMed/NCBI View Article : Google Scholar

37 

Rahmati M, Nalesso G, Mobasheri A and Mozafari M: Aging and osteoarthritis: Central role of the extracellular matrix. Ageing Res Rev. 40:20–30. 2017.PubMed/NCBI View Article : Google Scholar

38 

Schroeppel JP, Crist JD, Anderson HC and Wang J: Molecular regulation of articular chondrocyte function and its significance in osteoarthritis. Histol Histopathol. 26:377–394. 2011.PubMed/NCBI View Article : Google Scholar

39 

Alexopoulos LG, Youn I, Bonaldo P and Guilak F: Developmental and osteoarthritic changes in Col6a1-knockout mice: Biomechanics of type VI collagen in the cartilage pericellular matrix. Arthritis Rheum. 60:771–779. 2009.PubMed/NCBI View Article : Google Scholar

40 

Joos H, Albrecht W, Laufer S and Brenner RE: Differential effects of p38MAP kinase inhibitors on the expression of inflammation-associated genes in primary, interleukin-1β-stimulated human chondrocytes. Br J Pharmacol. 160:1252–1262. 2010.PubMed/NCBI View Article : Google Scholar

41 

Robinson WH, Lepus CM, Wang Q, Raghu H, Mao R, Lindstrom TM and Sokolove J: Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 12:580–592. 2016.PubMed/NCBI View Article : Google Scholar

42 

Woo JH, Lee JH, Kim H, Park SJ, Joe EH and Jou I: Control of Inflammatory Responses: A New Paradigm for the Treatment of Chronic Neuronal Diseases. Exp Neurobiol. 24:95–102. 2015.PubMed/NCBI View Article : Google Scholar

43 

Frizziero A, Bonsangue V, Trevisan M, Ames PRJ and Masiero S: Foot tendinopathies in rheumatic diseases: Etiopathogenesis, clinical manifestations and therapeutic options. Clin Rheumatol. 32:547–555. 2013.PubMed/NCBI View Article : Google Scholar

44 

Rai MF and Sandell LJ: Inflammatory mediators: Tracing links between obesity and osteoarthritis. Crit Rev Eukaryot Gene Expr. 21:131–142. 2011.PubMed/NCBI View Article : Google Scholar

45 

Stampella A, Monteagudo S and Lories R: Wnt signaling as target for the treatment of osteoarthritis. Best Pract Res Clin Rheumatol. 31:721–729. 2017.PubMed/NCBI View Article : Google Scholar

46 

Sun K, Luo J, Guo J, Yao X, Jing X and Guo F: The PI3K/AKT/mTOR signaling pathway in osteoarthritis: A narrative review. Osteoarthritis Cartilage. 28:400–409. 2020.PubMed/NCBI View Article : Google Scholar

47 

Chowdhury TT, Salter DM, Bader DL and Lee DA: Signal transduction pathways involving p38 MAPK, JNK, NFkappaB and AP-1 influences the response of chondrocytes cultured in agarose constructs to IL-1β and dynamic compression. Inflamm Res. 57:306–313. 2008.PubMed/NCBI View Article : Google Scholar

48 

Masuko-Hongo K, Berenbaum F, Humbert L, Salvat C, Goldring MB and Thirion S: Up-regulation of microsomal prostaglandin E synthase 1 in osteoarthritic human cartilage: Critical roles of the ERK-1/2 and p38 signaling pathways. Arthritis Rheum. 50:2829–2838. 2004.PubMed/NCBI View Article : Google Scholar

49 

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-1β. Osteoarthritis Cartilage. 13:697–708. 2005.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2021
Volume 22 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Y, Yan X, Jiang X, Lu F, Yang X and Li D: Vicenin 3 ameliorates ECM degradation by regulating the MAPK pathway in SW1353 chondrocytes. Exp Ther Med 22: 1461, 2021
APA
Chen, Y., Yan, X., Jiang, X., Lu, F., Yang, X., & Li, D. (2021). Vicenin 3 ameliorates ECM degradation by regulating the MAPK pathway in SW1353 chondrocytes. Experimental and Therapeutic Medicine, 22, 1461. https://doi.org/10.3892/etm.2021.10896
MLA
Chen, Y., Yan, X., Jiang, X., Lu, F., Yang, X., Li, D."Vicenin 3 ameliorates ECM degradation by regulating the MAPK pathway in SW1353 chondrocytes". Experimental and Therapeutic Medicine 22.6 (2021): 1461.
Chicago
Chen, Y., Yan, X., Jiang, X., Lu, F., Yang, X., Li, D."Vicenin 3 ameliorates ECM degradation by regulating the MAPK pathway in SW1353 chondrocytes". Experimental and Therapeutic Medicine 22, no. 6 (2021): 1461. https://doi.org/10.3892/etm.2021.10896