Open Access

Silencing of Wnt5a prevents interleukin‑1β‑induced collagen type II degradation in rat chondrocytes

  • Authors:
    • Shiping Shi
    • Zhentao Man
    • Wei Li
    • Shui Sun
    • Wei Zhang
  • View Affiliations

  • Published online on: October 11, 2016     https://doi.org/10.3892/etm.2016.3788
  • Pages: 3161-3166
  • Copyright: © Shi 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

Osteoarthritis (OA) is a joint disease, and few treatments to date have been able to delay OA progression. The degradation of collagen type II (COL2) in the cartilage matrix is an important initiating factor for OA progression; the upregulation of Wnt5a protein activates COL2 degradation. In the present study, small interfering RNA of Wnt‑5a was delivered by a lentiviral vector (LV‑Wnt5a‑RNAi) to silence Wnt‑5a mRNA and prevent COL2 degradation. To determine the function of LV‑Wnt5a‑RNAi, the OA chondrocyte model (OA‑like chondrocytes) were constructed using interleukin (IL)‑1β. Detected using reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR), Wnt‑5a mRNA in the OA‑like chondrocytes were upregulated in a time‑dependent manner, indicating that OA‑like chondrocytes were successfully constructed. The bioactivity of OA‑like chondrocytes was determined using Live‑Dead staining, and the result illustrated that the OA‑like chondrocytes stimulated with IL‑1β for 6 h remained viable, and these were used in Wnt5a silencing. The OA‑like chondrocytes were divided into three groups: Group I, cultivated with common medium; group II, cultivated with common medium supplemented with empty lentiviral vector; group III, cultivated with common medium supplemented with LV‑Wnt5a‑RNAi. The efficiency of LV‑Wnt5a‑RNAi transfection was determined using fluorescence microscopy, the result of which indicated that LV‑Wnt5a‑RNAi could efficiently be transfected into the OA‑like chondrocytes. The LV‑Wnt5a‑RNAi efficiency for the Wnt5a mRNA silencing was determined using RT‑qPCR. The result illustrated that the mRNA of Wnt5a in group III was significantly lower in group I compared with that in group II (P<0.05), indicating that the LV‑Wnt5a‑RNAi could successfully silence Wnt5a mRNA. To further verify whether the silencing of Wnt5a mRNA could prevent COL2 degradation, western blotting and immunohistochemical analyses were performed. The results demonstrated that COL2 in group III was significantly higher compared with that in groups I and II (P<0.05), which illustrated that the silencing of Wnt5a mRNA could prevent COL2 degradation. In conclusion, LV‑Wnt5a‑RNAi was formed successfully and could efficiently silence Wnt5a mRNA expressed by OA‑like chondrocytes. In addition, the silencing of Wnt5a mRNA could prevent the degradation of COL2 in OA‑like chondrocytes, confirming that LV‑Wnt5a‑RNAi may be used as a novel tool for OA treatment.

Introduction

Osteoarthritis (OA) affects ~10% of the world population >60 years old; it is difficult to treat joint disease and it is associated with a heavy financial burden on families and society (1). In developed countries, the cost of OA treatment represents 1.0–2.5% of the gross domestic product per year (2). To date, the treatment of OA is primarily based on symptom management, such as the use of non-steroidal anti-inflammatory drugs (NSAIDs) to relieve pain (3). However, few treatments have the proven ability to delay OA progression (4).

Gene therapy may be a useful method to delay OA progression (5), and it has been studied for nearly 20 years (6). The transferred gene could deliver gene products to the local area of the joint in a sustained manner, which has fewer extra-articular adverse effects (7). Currently, the most prominent transgenes used in OA gene therapy are transforming growth factor β1 (TGF-β1) (8) and insulin-like growth factor-1 (IGF-1) (9), which primarily promote the regeneration of cartilage. However, small interfering RNA (siRNA), which could silence the effect of specific mRNA (10), may be another important tool in OA gene therapy. The present study aimed to explore the biological effect of Wnt5a-specific siRNA in OA.

It is understood that OA results from cartilage degeneration. Specifically, the destruction of collagen type II (COL2) in the cartilage matrix is an important initiating factor for cartilage degeneration and OA progression. COL2 forms the skeletal structure of cartilage, which provides structural and biochemical support (11). As a result of the poor self-repair ability of cartilage, preventing COL2 degradation in cartilage injury is the key factor in inhibiting further cartilage degeneration and OA progression (12). Interleukin (IL)-1β is the most important proinflammatory cytokine in the pathophysiology of OA, which serves a key role in COL2 degradation (13,14). It has been reported that IL-1β could upregulate the Wnt5a protein, and the Wnt5a protein could activate the Jun amino-terminal kinase (JNK) signaling pathway, inducing the upregulation of matrix metalloproteinases (MMPs) (15,16). Ultimately, the MMPs cause COL2 degradation and destruction, thus inducing OA. That is to say, the Wnt5a protein is the core site of IL-1β-induced COL2 degradation in chondrocytes (Fig. 1). Consequently, Wnt5a mRNA can be chosen as a therapeutic target of siRNA.

The present study aimed to silence Wnt5a mRNA with lentiviral vector-mediated Wnt5a-specific siRNA (LV-Wnt5a-RNAi) to prevent COL2 degradation. OA-like chondrocyte injury was mimicked using IL-1β in vitro, and LV-Wnt5a-RNAi was used to transfect the OA-like chondrocytes. Following this, the efficiency of Wnt5a mRNA silencing was determined. Finally, the expression of COL2 proteins were determined to assess whether the silencing of Wnt5a mRNA can prevent COL2 degradation in vitro.

Materials and methods

Articular chondrocyte culture and identification

Chondrocytes from Sprague-Dawley rats (n=24 rats; age, 9 weeks; weight, 180±12 g) were harvested according to a previous study (17). These rats were fed with standard laboratory food (containing 1.56% calcium, 0.8% phosphorus and 800 IU/kg vitamin D) and kept under a 12 h light/dark cycle at 24°C and 50% humidity. After sacrifice with an overdose of anesthetic (12 ml/kg 20% urethane; Hengyuan Biological Technology Co., Ltd., Shanghai, China), the cartilage was sliced into small pieces from the femoral trochlea and digested with 2.0 mg/ml type II collagenase (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C for 4 h. Then, the digest solution was filtered with 200 mesh-filtrating screen to remove large fragments. The collected chondrocytes were seeded into culture dishes and cultured with Dulbecco's modified Eagle's medium (DMEM; Hyclone, GE Healthcare Life Sciences, Logan, UT, USA) supplemented with 15% fetal bovine serum (Hyclone), and 50 U/ml penicillin and streptomycin. At 90% confluence, the chondrocytes were passaged; chondrocytes of passage 2 were used, as in a previous study (18). All animal experiments were approved by the ethics committee of Shandong Provincial Hospital Affiliated to Shandong University, China, and complying with the ‘Guide for the Care and Use of Laboratory Animals’ published by the National Academy Press (NIH Publication no. 85–23, revised 1996).

To verify that the cultured cells were chondrocytes, the Toluidine Blue staining for sulfated glycosaminoglycans (GAGs) and immunohistochemical staining for COL2 were performed according to previous studies, respectively (19,20). The GAGs and COL2 were specific markers of hyaline cartilage.

To mimic the OA-like chondrocyte injury in vitro, IL-1β was used, as previously described (21). The chondrocytes of passage 2 were cultivated with serum-free DMEM for 24 h, and stimulated with medium containing 10 ng/ml IL-1β for 0, 1, 6, 12 or 24 h, respectively, as previously described (22).

To assess whether the OA-like chondrocytes were successfully generated, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to determine the gene expression of Wnt5a at different time points. Briefly, total RNA was harvested from chondrocytes of different groups using 1 ml TRIzol reagent (Takara Bio, Inc., Otsu, Japan). Subsequently, 1 µg total RNA was reverse transcribed in each group using PrimeScript RT Reagent Kit with gDNA Eraser (Takara Bio, Inc., Otsu, Japan), under incubation conditions of 37°C for 15 min, followed by 85°C for 5 sec. The primer sequences of Wnt5a and β-actin are reported in Table I. RT-qPCR was performed on an Applied Biosystems 7300 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) with the amplification program as follows: Initial denaturation at 95°C for 10 min; followed by denaturation at 95°C for 5 sec, annealing at 55°C for 30 sec and extension at 72°C for 30 sec for a total of 40 cycles; and a final extension at 60°C for 1 min. The total PCR reaction volume was 15 µl, including 1 µl cDNA, 7.5 µl SYBR® Green PCR Master Mix (Toyobo Co., Ltd., Osaka, Japan), 1 µl of each primer and 5.5 µl sterile water. The expression of Wnt5a relative to β-actin was calculated with the 2−∆∆Cq method (23) using SPSS v. 19.0 software (IBM SPSS, Armonk, NY, USA). This procedure was performed in triplicate for each gene, with each reaction also performed without reverse transcriptase as a negative control.

Table I.

Primer sequence of reverse transcription-quantitative polymerase chain reaction.

Table I.

Primer sequence of reverse transcription-quantitative polymerase chain reaction.

GenePrimer sequence
Wnt5a
  Forward 5′-TGTGGTTTAATGGTGCCTGA-3′
  Reverse 5′-TTCGTCGTGCTCAAGGTATG-3′
β-actin
  Forward 5′-CTAAGGCCAACCGTGAAAAG-3′
  Reverse 5′-AACACAGCCTGGATGGCTAC-3′

To verify that the OA-like chondrocytes were viable and could be used in the present study, Live-Dead staining was performed according to the manufacturer's instructions (Invitrogen; Thermo Fisher Scientific, Inc.) and as previously described (24). Briefly, the OA-like chondrocytes were cultivated with reagent containing 2 mM calcein AM and 4 mM ethidium homodimer-1 for 30 min at 37°C. After washing with phosphate-buffed saline (PBS), the specimens were assessed using a fluorescence microscope with 488 or 568 nm excitation. Live cells were colored green by calcein AM, while dead cells were colored red by ethidium homodimer-1.

Lentiviral vector packaging and transfection

The siRNA of Wnt-5a was designed and packaged into the LV-Wnt5a-RNAi by Genechem Co., Ltd. (Shanghai, China). The operation process of lentiviral vector formation followed the recommendations of the manufacturer.

In Wnt-5a mRNA silencing, the passage 2 chondrocytes were stimulated for 6 h with IL-1β as previously described (25). The OA-like chondrocytes were divided into three groups: Group I, incubated with complete DMEM for 7 days; group II, incubated with complete DMEM supplemented with empty lentiviral vector for 7 days; group III, incubated with complete DMEM supplemented with LV-Wnt5a-RNAi for 7 days. The concentration of empty lentiviral vector in group II was the same with the concentration of LV-Wnt5a-RNAi in group III, which ensured that the multiplicity-of-infection (MOI) was 50.

Western blotting analysis of COL2

Western blotting analysis was performed to determine whether the silencing of Wnt5a mRNA could prevent COL2 degradation in OA-like chondrocytes. Protein was harvested from the cells using lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) and the protein concentration was determined using a BCA protein assay kit (Pierce Biotechnology, Inc., Rockford, IL, USA). Protein (30 µg from each sample) was run on 10% sodium dodceyl sulfate-polyacrylamide gels and electrotransferred onto nitrocellulose membranes. Subsequently, the membranes were blocked with 5% bovine serum albumin (BSA; Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) for 1 h at room temperature. The blots were probed with monoclonal mouse anti-collagen II (cat. no. CP18; Calbiochem; Merck Millipore; 1:300 dilution) and monoclonal mouse anti-GAPDH (cat. no. AG019; Beyotime Institute of Biotechnology, Haimen, China, 1:500 dilution) antibodies overnight at 4°C, then incubated with a 1:4,000 dilution of goat anti-mouse, conjugated to horseradish peroxidase (HRP) (cat. no. sc-2005; Santa Cruz Biotechnology, Inc., Dallas, TX, USA; 1:1,000 dilution) for 1 h at room temperature. The blots were visualized using the enhanced chemiluminescence method according to the manufacturer's recommendations (EMD Millipore, Billerica, MA, USA). GAPDH was used as an internal control. Finally, the band density values were quantified with ImageJ v. 1.48 software (www.imagej.nih.gov/ij/).

Immunohistochemical analysis of COL2

To determine the COL2 protein expression in OA-like chondrocytes following the silencing of Wnt5a mRNA, immunohistochemical staining was performed, according to the previous report (26). Briefly, the chondrocytes of the three groups were fixed with 4% paraformaldehyde and blocked with 5% BSA in PBS. Then, the samples were incubated overnight at 4°C with anti-collagen II antibody (as with the Western blot). After three PBS washes, the cells were immersed in polyclonal goat anti-mouse secondary antibody (cat. no. PV-9002, Polink-2 plus Polymer HRP Detection System; Zhongshan Goldenbridge Co. Ltd., Beijing, China) at 37°C for 1 h. Following another round of washing, 3,3′-diaminobenzidine reagent (Zhongshan Golden Bridge Biotechnology Co., Ltd., Beijing, China) was used to determine the immunoactivity, and then the cell nuclei were counterstained with hematoxylin. The samples were observed under a light microscope, and the integrated optical density of different images were analyzed with Image-Pro Plus version 6.0 software (Media Cybernetics, Inc., Rockville, MD, USA).

Statistical analysis

All experiments were performed in triplicate, and the results were analyzed using one-way analysis of variance, followed by Tukey's test. SPSS software version 19.0 (IBM SPSS, Armonk, NY, USA) was used to perform the statistical analysis. The data were presented as mean ± standard deviation. P<0.05 were considered to indicate a statistically significant difference.

Results

Identification of chondrocytes

As shown in Fig. 2A, the cultured cells of passage 2 were not elongated, which indicated that the cultured cells were chondrocytes. Furthermore, the Toluidine Blue staining confirmed that the passage 2 cells were chondrocytes (Fig. 2B). Consequently, the chondrocytes were successfully cultured, which resulted in approval for the follow-up study.

Properties and bioactivity of OA-like chondrocytes

RT-qPCR was performed to determine whether the OA-like chondrocytes were successfully induced with IL-1β. The data in Fig. 3A illustrates that the mRNA of Wnt5a was upregulated in a time-dependent manner. The results indicate that the OA-like chondrocytes were successfully induced with IL-1β.

The bioactivity of OA-like chondrocytes were assessed using Live-Dead staining. As shown in Fig. 3B, although the inflammatory reaction was activated in the OA-like chondrocytes due to the stimulation of IL-1β, the cells remained viable. From these data, it can be concluded that the OA-like chondrocytes stimulated with IL-1β for 6 h could be used in subsequent studies of Wnt5a silencing.

LV-Wnt5a-RNAi transfected chondrocytes and Wnt5a mRNA silencing

After 7 days of incubation, the LV-Wnt5a-RNAi could be transfected into the OA-like chondrocytes with MOI = 50 (Fig. 4A), as determined by fluorescence microscope (Fig. 4B).

RT-qPCR demonstrated that Wnt5a mRNA in group III was significantly lower compared with that in groups I and II (P<0.05), while there was no significant difference between groups I and II (Fig. 4C). In conclusion, the LV-Wnt5a-RNAi used in group III could silence the Wnt5a mRNA expressed by the OA-like chondrocytes.

Preventing COL2 degradation by silencing Wnt5a mRNA

To determine whether the silencing of Wnt5a by LV-Wnt5a-RNAi could prevent COL2 degradation, COL2 proteins expressed in the three groups were assessed using western blotting and immunohistochemical analysis. The results of western blotting indicated that the COL2 content in group III was significantly higher compared with that in groups I and II (P<0.05; Fig. 5A and B), while no significant difference were observed between groups I and II. In the immunohistochemical analysis, the immunoactivity of group III was the highest among the three groups (Fig. 5C-F); this is consistent with the results of western blotting analysis. From these data, it can be concluded that the silencing of Wnt5a mRNA may prevent COL2 protein degradation.

Discussion

OA is a degenerative joint disease which affects a large number of individuals worldwide (1). In developed countries, the cost of OA treatment is about 1.0–2.5% of gross domestic product per year (2). To date, no highly effective drug can delay OA progression, because the existing treatment of OA is primarily based on symptom management, such as the use of NSAIDs to relieve pain (7). Gene therapy may be a useful method to delay OA progression, which has been studied for >20 years (6). The gene therapy of OA could be more effective and less expensive than the existing method, and be associated with fewer extra-articular adverse effects (10). Currently, the most prominent gene therapy of OA is the transgene of TGF-β1 (8) and IGF-1 (9), which primarily promote the regeneration of cartilage. However, siRNA may be another useful tool of gene therapy in OA and may silence the biological effects of specific mRNA (10). For example, Chen et al (27) used the adenoviral vector-mediated nuclear factor-κB p65-specific siRNA to alleviate inflammation of the synovium in OA.

It is understood that IL-1β is the most important proinflammatory cytokine in the pathophysiology of OA. IL-1β may upregulate the Wnt5a protein, and therefore activate the JNK signaling pathway to increase the expression of MMPs. MMPs result in the degradation and destruction of COL2, thus inducing OA (15,16). That is to say, the Wnt5a protein is the core site for IL-1β-induced COL2 degradation in OA. Consequently, the silencing of Wnt5a mRNA was chosen as the therapeutic target of Wnt5a-specific siRNA to prevent COL2 degradation in the present study.

The Wnt5a-specific siRNA was packaged in a lentiviral vector to improve the transfection efficiency. Previous studies have reported that the lentiviral vector is an effective siRNA delivery system, which can protect the enclosed siRNA and transport the siRNA to targeted cells (10). In the current study, green fluorescence could be observed in the majority of the chondrocytes, as shown in Fig. 4B, which indicated that the transfection efficiency of LV-Wnt5a-RNAi was excellent and the MOI used was appropriate.

The Wnt5a mRNA was silenced at least in part by LV-Wnt5a-RNAi, since the expression of Wnt5a mRNA in group III was significantly lower compared with that in groups I and II (Fig. 4C). With the action of LV-Wnt5a-RNAi, the Wnt5a mRNA becomes the component of RNA-induced silencing complexes (28). As a result, the Wnt5a mRNA is silenced and loses its biological activity.

To further explore whether silencing Wnt5a mRNA with LV-Wnt5a-RNAi can prevent COL2 degradation, the synthesis of COL2 was determined in the three groups. As shown in Fig. 5, the content of COL2 in group III was significantly higher compared with that in groups I and II. These results illustrate that the silencing of Wnt5a may prevent the degradation of COL2, the underlying mechanism being the silencing of Wnt5a reducing the synthesis of Wnt5a protein. The decrease of Wnt5a protein may reduce the activation of the JNK sigaling pathway, further inducing the downregulation of MMPs (15,16). Consequently, the silencing of Wnt5a may protect COL2 from degradation in vitro, which may be a useful method of treating OA. Further animal experiments should be performed in future studies to fully assess the protection of COL2 by the silencing of Wnt5a mRNA.

In conclusion, the present constructed LV-Wnt5a-RNAi, which is siRNA of Wnt-5a packaged into a lentiviral vector. The LV-Wnt5a-RNAi could successfully silence the mRNA of Wnt5a. This silencing of Wnt5a mRNA may prevent the degradation of COL2, which is the key component in cartilage matrix. Therefore, LV-Wnt5a-RNAi may be a useful tool to prevent the progression of OA.

Acknowledgements

The present work was supported by grants from the National Natural Science Foundation of China (grant no. 30672115) and the Science and Technology Development Plan of Shandong Province (grant no. 2012GSF21809).

References

1 

Ni GX, Li Z and Zhou YZ: The role of small leucine-rich proteoglycans in osteoarthritis pathogenesis. Osteoarthr Cartil. 22:896–903. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Hiligsmann M, Cooper C, Arden N, Boers M, Branco JC, Brandi M Luisa, Bruyère O, Guillemin F, Hochberg MC, Hunter DJ, et al: Health economics in the field of osteoarthritis: An expert's consensus paper from the European Society for Clinical and Economic Aspects of Osteoporosis and Osteoarthritis (ESCEO). Semin Arthritis Rheum. 43:303–313. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Cutolo M, Berenbaum F, Hochberg M, Punzi L and Reginster JY: Commentary on recent therapeutic guidelines for osteoarthritis. Semin Arthritis Rheum. 44:611–617. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Glyn-Jones S, Palmer AJ, Agricola R, Prince AJ, Vincent TL, Weinans H and Carr AJ: Osteoarthritis. Lancet. 386:376–387. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Evans CH and Huard J: Gene therapy approaches to regenerating the musculoskeletal system. Nat Rev Rheumatol. 11:234–242. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Bandara G, Robbins PD, Georgescu HI, Mueller GM, Glorioso JC and Evans CH: Gene transfer to synoviocytes: Prospects for gene treatment of arthritis. DNA Cell Biol. 11:227–231. 1992. View Article : Google Scholar : PubMed/NCBI

7 

Evans CH, Ghivizzani SC and Robbins PD: Getting arthritis gene therapy into the clinic. Nat Rev Rheumatol. 7:244–249. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Ivkovic A, Pascher A, Hudetz D, Maticic D, Jelic M, Dickinson S, Loparic M, Haspl M, Windhager R and Pecina M: Articular cartilage repair by genetically modified bone marrow aspirate in sheep. Gene Ther. 17:779–789. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Haupt JL, Frisbie DD, McIlwraith CW, Robbins PD, Ghvizzani S, Evans CH and Nixon AJ: Dual transduction of insulin-like growth factor-I and interleukin-1 receptor antagonist protein controls cartilage degradation in an osteoarthritic culture model. J Orth Res. 23:118–126. 2005. View Article : Google Scholar

10 

Shi Q, Zhang XL, Dai KR, Benderdour M and Fernandes JC: siRNA therapy for cancer and non-lethal diseases such as arthritis and osteoporosis. Expert Opin Biol Ther. 11:5–16. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Gao Y, Liu S, Huang J, Guo W, Chen J, Zhang L, Zhao B, Peng J, Wang A, Wang Y, et al: The ECM-cell interaction of cartilage extracellular matrix on chondrocytes. BioMed Res Int. 2014:6484592014. View Article : Google Scholar : PubMed/NCBI

12 

Tchetina EV, Squires G and Poole AR: Increased type II collagen degradation and very early focal cartilage degeneration is associated with upregulation of chondrocyte differentiation related genes in early human articular cartilage lesions. J Rheumatol. 32:876–886. 2005.PubMed/NCBI

13 

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. View Article : Google Scholar : PubMed/NCBI

14 

Mabey T and Honsawek S: Cytokines as biochemical markers for knee osteoarthritis. J Orthop. 6:95–105. 2015.

15 

Ge X, Ma X, Meng J, Zhang C, Ma K and Zhou C: Role of Wnt-5A in interleukin-1beta-induced matrix metalloproteinase expression in rabbit temporomandibular joint condylar chondrocytes. Arthritis Rheum. 60:2714–2722. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Ryu JH and Chun JS: Opposing roles of WNT-5A and WNT-11 in interleukin-1beta regulation of type II collagen expression in articular chondrocytes. J Biol Chem. 281:22039–22047. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Yu DG, Ding HF, Mao YQ, Liu M, Yu B, Zhao X, Wang XQ, Li Y, Liu GW, Nie SB, et al: Strontium ranelate reduces cartilage degeneration and subchondral bone remodeling in rat osteoarthritis model. Acta Pharmacol Sin. 34:393–402. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Wang PY and Tsai WB: Modulation of the proliferation and matrix synthesis of chondrocytes by dynamic compression on genipin-crosslinked chitosan/collagen scaffolds. J Biomater Sci Polym Ed. 24:507–519. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Yu L, Weng Y, Shui X, Fang W, Zhang E and Pan J: Multipotent Adult Progenitor Cells from Bone Marrow Differentiate into Chondrocyte-Like Cells. J Arthroplasty. 30:1273–1276. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Tekari A, Luginbuehl R, Hofstetter W and Egli RJ: Chondrocytes expressing intracellular collagen type II enter the cell cycle and co-express collagen type I in monolayer culture. J Orthop Res. 32:1503–1511. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Chen WP and Wu LD: Chlorogenic acid suppresses interleukin-1β-induced inflammatory mediators in human chondrocytes. Int J Clin Exp Pathol. 7:8797–8801. 2014.PubMed/NCBI

22 

Tao R, Wang S, Xia X, Wang Y, Cao Y, Huang Y, Xu X, Liu Z, Liu P, Tang X, et al: Pyrroloquinoline Quinone Slows Down the Progression of Osteoarthritis by Inhibiting Nitric Oxide Production and Metalloproteinase Synthesis. Inflammation. 38:1546–1555. 2015. View Article : Google Scholar : PubMed/NCBI

23 

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. View Article : Google Scholar : PubMed/NCBI

24 

Xu HG, Zhang XH, Wang H, Liu P, Wang LT, Zuo CJ, Tong WX and Zhang XL: Intermittent Cyclic Mechanical Tension-Induced Calcification and downregulation of ankh gene expression of end plate chondrocytes. Spine. 37:1192–1197. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Lu J, Sun Y, Ge Q, Teng H and Jiang Q: Histone deacetylase 4 alters cartilage homeostasis in human osteoarthritis. BMC Musculoskelet Disord. 15:4382014. View Article : Google Scholar : PubMed/NCBI

26 

Wu PC, Tsai CL, Gordon GM, Jeong S, Itakura T, Patel N, Shi S and Fini ME: Chondrogenesis in scleral stem/progenitor cells and its association with form-deprived myopia in mice. Mol Vision. 21:138–147. 2015.

27 

Chen LX, Lin L, Wang HJ, Wei XL, Fu X, Zhang JY and Yu CL: Suppression of early experimental osteoarthritis by in vivo delivery of the adenoviral vector-mediated NF-kappaBp65-specific siRNA. Osteoarthritis Cartilage. 16:174–184. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Hammond SM, Bernstein E, Beach D and Hannon GJ: An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature. 404:293–296. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 12 Issue 5

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
Shi S, Man Z, Li W, Sun S and Zhang W: Silencing of Wnt5a prevents interleukin‑1β‑induced collagen type II degradation in rat chondrocytes. Exp Ther Med 12: 3161-3166, 2016
APA
Shi, S., Man, Z., Li, W., Sun, S., & Zhang, W. (2016). Silencing of Wnt5a prevents interleukin‑1β‑induced collagen type II degradation in rat chondrocytes. Experimental and Therapeutic Medicine, 12, 3161-3166. https://doi.org/10.3892/etm.2016.3788
MLA
Shi, S., Man, Z., Li, W., Sun, S., Zhang, W."Silencing of Wnt5a prevents interleukin‑1β‑induced collagen type II degradation in rat chondrocytes". Experimental and Therapeutic Medicine 12.5 (2016): 3161-3166.
Chicago
Shi, S., Man, Z., Li, W., Sun, S., Zhang, W."Silencing of Wnt5a prevents interleukin‑1β‑induced collagen type II degradation in rat chondrocytes". Experimental and Therapeutic Medicine 12, no. 5 (2016): 3161-3166. https://doi.org/10.3892/etm.2016.3788