A disintegrin and metallproteinase 15 knockout decreases migration of fibroblast-like synoviocytes and inflammation in rheumatoid arthritis

Retraction in: /10.3892/mmr.2024.13165

  • Authors:
    • Jinliang Gao
    • Wei Zheng
    • Liming Wang
    • Bailin Song
  • View Affiliations

  • Published online on: February 4, 2015     https://doi.org/10.3892/mmr.2015.3302
  • Pages: 4389-4396
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to determine whether the expression of A disintegrin and metallproteinase 15 (ADAM15) affected the inflammatory conditions and cell migration in human fibroblast‑like synoviocytes (FLSs) in a rat model of rheumatoid arthritis (RA). The expression of ADAM15 in FLSs stimulated with lipopolysaccharide (LPS) was confirmed by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. The effects of small interfering RNA targeting ADAM15 (siADAM5) on pro‑inflammatory cytokines and chemokines were assessed using an enzyme‑linked immunosorbent assay. The effects of siADAM15 on cell invasion and migration in FLS were also assessed in vitro. The therapeutic effects and side effects of ADAM15 in a rat model of collagen‑induced arthritis (CIA) were examined in vivo. The present results revealed that ADAM15 expression was significantly elevated at the mRNA and protein level in FLSs stimulated with LPS and that silencing ADAM15 suppressed the expression of pro‑inflammatory cytokines and chemokines, preventing FLS cell migration and invasion via inhibiting vascular endothelial growth factor‑A, matrix metalloproteinase (MMP)1 and MMP‑3 expression. In addition, treatment of CIA rats using siADAM15 significantly reduced the arthritis score and extent of joint damage in the rats. These findings indicated that silencing ADAM15 had anti‑inflammatory effects in FLSs and efficiently inhibited the development of CIA. Therefore, ADAM15 may be a potential target molecule for RA therapies.

Introduction

Rheumatoid arthritis (RA) is an autoimmune disorder characterized by the accumulation of inflammatory cells in the joints, leading to hyperproliferation of synovial cells and tissue destruction (1,2). RA synovium contains high levels of inflammatory cytokines and abundant inflammatory cells, including infiltrating lymphocytes and monocytes (3). Synovial tissue (ST) macrophages produce large quantities of proinflammatory cytokines and proteases, including tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-15 and stromal cell-derived factor 1 (CXCL12) (47). These cytokines and chemokines have been associated with the progression of RA and may have pathogenic roles in the establishment of rheumatoid synovitis (47). Although biological preparations targeting these pro-inflammatory cytokines are widely used clinically in the treatment of RA, there are caveats in using the biological preparations, including risk of infections, such as tuberculosis, high cost and individual variations in efficacy (8). However, the beneficial effects of cytokines are limited to a number of patients (8). Thus, further investigation is required to develop therapies that achieve remission of RA for all patients.

A class of disintegrins and metallproteinases, termed ADAMs, are responsible for the liberation of a variety of cell surface-expressed proteins, and have been implicated in several inflammatory and degenerative pathological conditions (9). ADAM15, a member of the ADAM family, has been observed to be upregulated in a variety of types of cancer and to contribute to cancer progression and metastasis (10-12). In addition to its role in tumorigenesis, ADAM15 has important roles in degenerative joint disease (1315), as well as in inflammatory diseases, such as RA (16). In particular, ADAM15 has been found to be markedly upregulated in the synovial membranes of patients with RA, with a marked level of expression in the hyperplastic synovial lining layer (17,18). ADAM15 has been observed to cleave various inflammatory and angiogenic mediators from the cell surface (16). Previously, Bohm et al (19) revealed that ADAM15 contributes to apoptotic resistance in FLSs by activating the Src/FAK pathway upon Fas ligand exposure (17). These studies imply that ADAM15 may be involved in RA pathophysiology, and that inhibition of ADAM15 may decrease synovitis in RA. Therefore, in the present study, the association between ADAM15 expression and the expression of pro-inflammatory cytokines and chemokines in rat fibroblast-like synoviocytes (FLSs) was examined. The effects of small interfering RNA (siRNA) targeting ADAM15 in a rat model of collagen-induced arthritis (CIA) were also examined, as well as cell migration and invasion of FLSs.

Materials and methods

Cell culture

FLSs were obtained from the synovium of active anti-citrullinated protein antibody-positive RA patients during knee joint arthroscopy following ethical approval from the Ethics Committee of Changchun University of Chinese Medicine (Changchun, China; no. CZY2013-976). Informed consent was obtained from all patients. FLSs were isolated from synovial tissues via enzymatic digestion as previously described by Yoshioka et al (20). FLSs were grown in Dulbecco’s modified Eagle’s medium (DMEM; Gibco-BRL, Gaithersburg, MD, USA) containing 10% heat-inactivated fetal calf serum (FCS; Invitrogen Life Technologies, Carlsbad, CA, USA), supplemented with antibiotics (100 mg/ml streptomycin and 100 U/ml penicillin) in a humidified incubator at 37°C in 5% CO2. Cells used for experiments were from the third to sixth passages.

Silencing of ADAM15 in synovial fibroblasts by RNA interference

Silencing of ADAM15 was performed using small interfering RNAs (siRNAs; Ambion, Austin, TX, USA) targeting ADAM15 (siADAM5) as described previously (21). The nonsilencing siRNA control #1 (Ambion) was used as the negative control (siNC). FLSs (1×104) were seeded in 96-well culture plates and grown for 24 h. A total of 100 pmol siRNAs were mixed with 7.5 ml RNAiFect transfection reagent (Qiagen, Hilden, Germany) followed by incubation for 15 min. After 12 h, the cells were transfected with 20 μM siRNA using RNAiFect according to the manufacturer’s instructions.

The expression of ADAM15 was examined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting with an antibody against ADAM15 to validate the silencing efficiency of the target gene following RNAi.

RT-qPCR

Total RNA was extracted from cultured FLSs using TRIzol reagent (Invitrogen Life Technologies) according to the manufacturer’s instructions. Subsequently, RNA was reverse-transcribed into cDNA using a Primescript™ RT reagent kit according to the manufacturer’s instructions (Takara Bio Inc., Dalian, China). RT-qPCR was conducted using the SYBR green fluorescent dye method and a Rotor-Gene 3000 real-time PCR apparatus (Corbett Life Science, Sydney, Australia). ADAM15 gene-specific amplification was confirmed using PCR with specific primers sequences as follows: Sense: 5′-GGCAATCGAGGCAGCAAAT-3′ and antisense: 5′-TGGTGGAGATCAGCCCAAAC-3′ and then subjected to melting curve analysis. β-actin was used as an internal control for standardization. The primer sequences for β-actin were as follows: Sense: 5′-GATCATTGCTCCTCCTGAGC-3′ and antisense: 5′-ACTCCTGCTTGCTGATCCAC-3′. The amplification was performed with an initial denaturation at 95°C for 5 min followed by 40 cycles of 95°C for 10 sec and 60°C for 30 sec. All RT-qPCR assessments were performed in triplicate and were performed after the third day of siRNA transfection. The data were analyzed using the comparative Ct method.

Western blot analysis

The cells were collected and then homogenized in radioimmunoprecipitation assay lysis buffer (Sigma-Aldrich Chemie GmbH, Steinheim, Germany) on ice for 30 min. Cell lysates were clarified by centrifugation at 10,000 × g for 15 min, and protein concentrations were determined using the Bradford reagent (Sigma-Aldrich Chemie GmbH). Equal quantities of protein (15 μg/lane) from the cell lysates were separated on an 8–15% SDS-polyacrylamide gel (SDS-PAGE) and transferred onto nitrocellulose membranes (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). The membrane was incubated for 2 h in phosphate-buffered saline (PBS) plus 0.1% Tween-20 (PBST) and 5% non-fat skimmed milk to block nonspecific binding. Subsequently, the membranes were incubated overnight at 4°C with primary antibodies. Following washing, proteins were visualized using an electrochemiluminescence detection kit (PerkinElmer, Inc., Waltham, MA, USA) with the rabbit anti-mouse horseradish peroxidase-conjugated IgG (1:12,000 dilution; cat. no. L100903) secondary antibody (Amersham Pharmacia Biotech, Piscataway, NJ, USA) for 2 h. All assays were performed after the third day of siRNA transfection. The primary antibodies used in the western blot analysis were as follows: Mouse monoclonal anti-human VEGF-A (1:1,000 dilution; cat. no. CB11008356; Santa Cruz Biotechnology, Inc.), mouse monoclonal anti-human MMP-3 (1:3,000 dilution; cat. no. CB0364331; Santa Cruz Biotechnology, Inc.), mouse monoclonal anti-human ADAM15 (1:1,000 dilution; cat. no. CB2556697; Santa Cruz Biotechnology, Inc.) and mouse monoclonal anti-human β-actin (1:5,000 dilution; cat. no. CB7664544; Santa Cruz Biotechnology, Inc.).

ELISA

To quantify TNF-α, IL-6, IL-15 and CXCL12 production, cells transfected with siRNA were incubated for 8 h, followed by stimulation with 10 μg/ml lipopolysaccaride (LPS; Sigma-Aldrich, St. Louis, MO, USA), and then incubated for 24 h. At 24 h after LPS stimulation, the culture supernatant was harvested and the concentrations of TNF-α, IL-6, IL-15 and CXCL12 were measured using an ELISA kit for human TNF-α, IL-6, IL-15 and CXCL12 (Genzyme Techne, Minneapolis, MN, USA) according to the manufacturer’s instructions. The concentrations of each were normalized relative to the total number of cells. The determinations were performed in duplicate for each cell culture preparation.

Detection of cell apoptosis

In order to measure the effect of siADAM15 on cell apoptosis, a terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL) assay was performed. Briefly, FLSs were transfected with siADAM15, siNC for 24 h, followed by stimulation with 10 μg/ml LPS (Sigma-Aldrich) and then incubated for 48 h. Cellular DNA fragmentation was measured with the ApoTag Red in situ Apoptosis detection kit (Chemicon International, Temecula, CA, USA) according to the manufacturer’s instructions. To quantify the apoptotic cells, the TUNEL-positive cells were counted using a confocal microscope (LEXT-OLS3100; Olympus, Tokyo, Japan). In addition, at the molecular level, caspase 3/7 activity was also detected as an additional indicator of apoptosis.

Determination of caspase 3/7 activity

A total of 1×104 synovial fibroblasts were seeded in 96-well culture plates and grown for 24 h in DMEM containing 10% FCS. Following silencing of ADAM15 for 24 h, cells were treated with 10 μg/ml LPS (Sigma-Aldrich) for 48 h, and then caspase 3/7 activity was measured using the Caspase-Glo 3/7 assay (Promega Corporation, Madison, WI, USA) on a Mithras LB 940 luminometer plate reader (Berthold Technologies, Bad Wildbad, Germany) as described previously (21).

Cell migration and invasion assay

To assess the effect of siADAM15 on cell migration, a wound-healing assay was performed. Briefly, FLS were seeded at a density of 4×103 cells/well in a 96-well plate. After 48 h of siADAM15 transfection, the cells formed a fluent monolayer and were observed under a fluorescent microscope (CKX31; Olympus). A linear scratch was formed using a 10 μl pipette tip 120 h after infection. Wounded monolayers were washed with PBS to remove detached cells and debris. Transwell migration assays were performed using a 24-well Boyden chamber (6.5 mm diameter, 8.0 μm; BD Biosciences, Mountain View, CA, USA) according to the manufacturer’s instructions. Photomicrographs of ten random fields were obtained (original magnification, ×100), and cells were counted to calculate the average number of cells that had migrated.

For the in vitro invasion assay, similar experiments were performed using inserts coated using a Matrigel basement membrane matrix (BD Biosciences). Briefly, the Matrigel was diluted in serum-free cold media, placed into the upper chambers of a 24-well Transwell and incubated at 37°C for 1 h. Cells were resuspended with serum-free DMEM media at a density of 5×104 cells/well and incubated for 48 h to evaluate cell migration. All experiments were performed in duplicate.

Induction of CIA and treatment CIA with siADAM15 in vivo

A total of 30 6–8 week-old male DBA/1 rats (200–250 g) were purchased from the Institute of Laboratory Animal Science, Jilin University (Changchun, China), were maintained under specific pathogen-free conditions and provided with food and water ad libitum. All animal experiments were conducted according to the standards of animal care as outlined in the Guide for the Care and Use of Experimental Animals of Jilin University. To induce CIA, collagen type II (Collagen Research Center, Tokyo, Japan) was dissolved in 0.01 M acetic acid (2 mg/ml) and emulsified at 1:1 in Freund’s incomplete adjuvant (Sigma-Aldrich; CII/FIA) on ice. DBA/1 rats were injected intradermally with 200 ml CII/FIA solution at the base of the tail.

Complexes of siRNA and atelocollagen (Boppard Co., Ltd., Beijing, China) were prepared as previously described (22). The siRNA/atelocollagen complex was formed by mixing siRNAs with atelocollagen and these complexes were prepared in an injectable form. Subsequently, the siRNA/atelocollagen complexes (0.5 mg/kg body weight) were administered to rats with CIA twice weekly for three weeks. This protocol has been well established by Li et al (23).

Arthritic score and histological analysis

Clinical arthritic assessment was performed every three days according to a previously described scoring system (23,24). The maximum score per paw was 3 with a total score of 12 per mouse. At the termination of the experiment the mice were sacrificed by CO2 asphyxiation, and the hind limbs were fixed with 4% paraformaldehyde, decalcified and embedded in paraffin. Serial 4-μm sections were cut and stained with hematoxylin and eosin according to standard protocols for morphological analysis. The sections were analyzed microscopically (CX41; Olympus) for the degree of inflammation and arthritic changes, including infiltration of inflammatory cells, synovial proliferation, destruction of articular cartilage and bone destruction following a previously described method (23,25).

Statistical analysis

Statistical analysis of data was performed using SPSS 19.0 (IBM, Armonk, NY, USA) and GraphPad Prism 5.01 software (GraphPad Software, Inc., La Jolla, CA, USA). Data points and the bars indicate the mean ± standard deviation of three independent determinations. Data were analyzed using a one-way analysis of variance followed by Tukey’s multiple comparison tests. P<0.01 was considered to indicate a statistically significant difference.

Results

siADAM15 suppresses the over-expression of ADAM15 induced by LPS in FLSs

ADAM15 mRNA levels and protein levels in FLSs were measured using RT-qPCR and western blotting, respectively. The present result revealed that the ADAM15 mRNA level and protein level in the FLSs were significantly upregulated by LPS-stimulation compared with normal FLSs (without stimulation; P<0.01, Fig. 1A and B). In addition, it was also identified that ADAM15 expression in siADAM15 treated FLSs was significantly decreased compared with untreated FLSs and FLSs treated with siNC (P<0.01; Fig. 1A and B). These findings suggest that ADAM15 expression, which was upregulated by LPS-stimulation, was significantly inhibited by siADAM15 (Fig. 1).

siADAM15 inhibits LPS-induced pro-inflammatory cytokines and CXCL16 expression in FLS

To quantify TNF-α, IL-6, IL-15 and CXCL12 production, an ELISA was performed. It was found that transfection of si-ADAM15 significantly inhibited the expression of TNF-α, IL-6 and IL-15 upregulated by LPS (P<0.01; Fig. 2A–C). The levels of CXCL-12 were also significantly reduced by transfection with siADAM15 (P<0.01; Fig. 2D).

siADAM15 induces cell apoptosis of FLSs stimulated with LPS

It was also examined whether silencing the ADAM15 gene had any effect on cell apoptosis using a TUNEL assay. FLSs were silenced for 24 h with the specific siRNAs or treated with a negative siRNA control and then subsequently exposed to LPS for 48 h, finally a TUNEL assay was performed. The present results demonstrated that silencing ADAM15 led to a marked increase in the number of apoptotic cells compared with the untreated control group and the si-NC treatment group (P<0.01; Fig. 3A). In addition, no significant difference was identified between the control group and the si-NC group in the induction of FLS apoptosis.

In order to examine the possible mechanism of the pro-apoptotic effect of silencing ADAM15, caspase3/7 activity was determined using an ELISA. The results are shown in Fig. 3B. The results demonstrated that silencing ADAM15 significantly increased caspase 3/7 activity compared with the control group and the siNC group.

siADAM15 inhibits FLS migration and invasion

To demonstrate the effect of ADAM15 on FLS migration and invasion, cell migration and invasion were analyzed. RA-FLS cells were treated with siADAM15 or siNC. As a result, ADAM15 silencing significantly attenuated the migration and invasion of RA-FLS (P<0.01; Fig. 4). These results imply that the presence of siADAM15 attenuates chemokine-induced FLS migratory behavior.

ADAM15 silencing inhibits expression of VEGF-A, MMP-1, and MMP-3 in human RA-FLS

To illustrate the possible mechanism of effect on the migration of silencing ADAM15, VEGF-A, MMP-1 and MMP-3 were further investigated in the human RA-FLSs. The cells were pretreated with siADAM15 or siNC for 24 h followed by stimulation with human LPS for 48 h. The supernatants were then assayed for VEGF-A, MMP-3 and MMP-9 expression using western blot analysis. The present results demonstrated that silencing ADAM15 resulted in a marked decrease in the levels of VEGF-A, MMP-1 and MMP-3 in the supernatants compared with the controls and the si-NC group (Fig. 5).

ADAM15 silencing lowers arthritis score and histological damage in rats with CIA

The effect of silencing ADAM15 in a CIA rat model was also evaluated. The current results demonstrated that silencing ADAM15 reduced the arthritis score of CIA in rats during the treatment (P<0.01; Fig. 6A). Furthermore, the histological examination at the end of the experiment revealed that the articular histological damage was markedly reduced by siADAM15 as compared with siNC treated rats (P<0.01; Fig. 6B and C).

Discussion

RA is an inflammatory degenerative joint disease involving tissue remodeling, in which the affected joints develop chronic synovitis, which is characterized by abundant neovascularization and infiltration of inflammatory cells. It has been observed that activated FLSs migrate throughout the joint and even invade other joints, leading to extensive cartilage and bone deterioration, which contributes to the development of RA (26). FLSs have also been found to secrete multiple cytokines, such as TNF-α, IL-6, IL-1β and growth factors, including VEGF that in part activate an autocrine loop, resulting in further FLS hyperplasia (27). A growing quantity of evidence indicates that the ADAM family is involved in the regulation of inflammatory responses, in which ADAM family members may be novel therapeutic targets for the treatment of inflammatory disorders (16). ADAM15, a member of the ADAM family, has been observed to be highly expressed in the RA synovial membrane as well as in osteoarthritic chondrocytes, whereas its expression levels are markedly low in normal, nondiseased cartilage and synovial tissue (17,18). Thus, ADAM15 was examined as a viable therapeutic target for RA treatment. In the present study, the results demonstrated that LPS-stimulation of human FLSs increased the expression of ADAM15, which were consistent with those of previous studies (13,17,18). It was also identified that silencing ADAM15 suppressed the expression of pro-inflammatory cytokines and chemokines. In addition, the present results clearly demonstrated that treatment with siRNA against ADAM15 for three weeks reduced the arthritis score and extent of joint damage in the rats. These findings indicate that silencing ADAM15 may be a viable therapeutic target in the amelioration of disease progression in RA.

TNF-α and IL-6 are cytokines, which have major roles in the etiology of experimental arthritis in rats with CIA, as well as in human RA (28). It has been demonstrated that TNF-α is a key inflammatory cytokine involved in the pathogenesis of rheumatoid arthritis and inhibiting TNF-α expression via antagonism or alternate drug treatments is an effective treatment for RA (29,30). In the present study, it was determined whether the expression of ADAM15 effected the inflammatory conditions in human FLS. The present results demonstrated that transfection of siADAM15 in vitro inhibited LPS-induced TNF-α and IL-6 expression, suggesting that siADAM15 may have great potential for use as a therapeutic tool in the treatment of RA patients resistant to anti-cytokine therapies.

Matrix metalloproteinases (MMPs) are an important proteolytic enzyme family with a zinc-activated region that degrades numerous components of the extracellular matrix. They have an important role in tissue repair, cell invasion and metastasis (31). It was found that MMP-1 and MMP-3 are produced by synovial lining cells in RA and exhibit a major role in the process of cartilage destruction in RA joints (32,33). In the present study, the results demonstrated that the expression of VEGF-A, MMP1 and MMP-3 was decreased by ADAM15 silencing and that RA-FLS migration and invasion were significantly attenuated by ADAM15 knockdown. These findings indicated that silencing ADAM15 attenuated FLS migration and invasion via inhibiting VEGF-A, MMP1 and MMP-3 protein expression.

In conclusion, the present study demonstrated that knockdown of ADAM15 by siRNA provided protection against the development of inflammation and joint destruction in a rat model of RA, and that ADAM15 silencing blocked FLS migration and invasion via inhibiting VEGF-A, MMP1 and MMP-3 expression. These findings indicated that ADAM15 may be a target molecule in therapies for RA.

Acknowledgments

The present study was supported by the Science and Technology Research and Innovation Team funded by Jilin Province (grant no. JL2012058).

References

1 

Feldmann M, Brennan FM and Maini RN: Rheumatoid arthritis. Cell. 85:307–310. 1996. View Article : Google Scholar : PubMed/NCBI

2 

Brennan FM and McInnes IB: Evidence that cytokines play a role in rheumatoid arthritis. J Clin Invest. 118:3537–3545. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Ritchlin C: Fibroblast biology. Effector signals released by the synovial fibroblast in arthritis. Arthritis Res. 2:356–360. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Buchan G, Barrett K, Turner M, Chantry D, Maini RN and Feldmann M: Interleukin-1 and tumour necrosis factor mRNA expression in rheumatoid arthritis: prolonged production of IL-1 alpha. Clin Exp Immunol. 73:449–455. 1988.PubMed/NCBI

5 

Gabay C, Lamacchia C and Palmer G: IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol. 6:232–241. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Choy EH and Panayi GS: Cytokine pathways and joint inflammation in rheumatoid arthritis. N Engl J Med. 344:907–916. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Nanki T, Hayashida K, El-Gabalawy HS, et al: Stromal cell-derived factor-1-CXC chemokine receptor 4 interactions play a central role in CD4+ T cell accumulation in rheumatoid arthritis synovium. J Immunol. 165:6590–6598. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Scott DL: Biologics-based therapy for the treatment of rheumatoid arthritis. Clin Pharmacol Ther. 91:30–43. 2012. View Article : Google Scholar

9 

Pruessmeyer J and Ludwig A: The good, the bad and the ugly substrates for ADAM10 and ADAM17 in brain pathology, inflammation and cancer. Semin Cell Dev Biol. 20:164–174. 2009. View Article : Google Scholar

10 

Baren JP, Stewart GD, Stokes A, et al: mRNA profiling of the cancer degradome in oesophago-gastric adenocarcinoma. Br J Cancer. 107:143–149. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Kuefer R, Day KC, Kleer CG, et al: ADAM15 disintegrin is associated with aggressive prostate and breast cancer disease. Neoplasia. 8:319–329. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Lucas N and Day ML: The role of the disintegrin metalloproteinase ADAM15 in prostate cancer progression. J Cell Biochem. 106:967–974. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Böhm BB, Aigner T, Gehrsitz A, Blobel CP, Kalden JR and Burkhardt H: Up-regulation of MDC15 (metargidin) messenger RNA in human osteoarthritic cartilage. Arthritis Rheum. 42:1946–1950. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Böhm BB, Schirner A and Burkhardt H: ADAM15 modulates outside-in signalling in chondrocyte-matrix interactions. J Cell Mol Med. 13:2634–2644. 2009. View Article : Google Scholar

15 

Böhm BB, Aigner T, Roy B, Brodie TA, Blobel CP and Burkhardt H: Homeostatic effects of the metalloproteinase disintegrin ADAM15 in degenerative cartilage remodeling. Arthritis Rheum. 52:1100–1109. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Charrier-Hisamuddin L, Laboisse CL and Merlin D: ADAM-15: a metalloprotease that mediates inflammation. FASEB J. 22:641–653. 2008. View Article : Google Scholar

17 

Komiya K, Enomoto H, Inoki I, et al: Expression of ADAM15 in rheumatoid synovium: up-regulation by vascular endothelial growth factor and possible implications for angiogenesis. Arthritis Res Ther. 7:R1158–R1173. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Böhm BB, Aigner T, Blobel CP, Kalden JR and Burkhardt H: Highly enhanced expression of the disintegrin metalloproteinase MDC15 (metargidin) in rheumatoid synovial tissue. Arthritis Rheum. 44:2046–2054. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Böhm BB, Freund I, Krause K, Kinne RW and Burkhardt H: ADAM15 adds to apoptosis resistance of synovial fibroblasts by modulating focal adhesion kinase signaling. Arthritis Rheum. 65:2826–2834. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Yoshioka Y, Kozawa E, Urakawa H, et al: Suppression of hyaluronan synthesis alleviates inflammatory responses in murine arthritis and in human rheumatoid synovial fibroblasts. Arthritis Rheum. 65:1160–1170. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Fried D, Böhm BB, Krause K and Burkhardt H: ADAM15 protein amplifies focal adhesion kinase phosphorylation under genotoxic stress conditions. J Biol Chem. 287:21214–21223. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Minakuchi Y, Takeshita F, Kosaka N, et al: Atelocollagen-mediated synthetic small interfering RNA delivery for effective gene silencing in vitro and in vivo. Nucleic Acids Res. 32:e1092004. View Article : Google Scholar : PubMed/NCBI

23 

Li F, Li X, Kou L, Li Y, Meng F and Ma F: SUMO-conjugating enzyme UBC9 promotes proliferation and migration of fibroblast-like synoviocytes in rheumatoid arthritis. Inflammation. 37:1134–1141. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Tarrant TK, Liu P, Rampersad RR, et al: Decreased Th17 and antigen-specific humoral responses in CX3 CR1-deficient mice in the collagen-induced arthritis model. Arthritis Rheum. 64:1379–1387. 2012. View Article : Google Scholar

25 

Nishikawa M, Myoui A, Tomita T, Takahi K, Nampei A and Yoshikawa H: Prevention of the onset and progression of collagen-induced arthritis in rats by the potent p38 mitogen-activated protein kinase inhibitor FR167653. Arthritis Rheum. 48:2670–2681. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Chang SK, Gu Z and Brenner MB: Fibroblast-like synoviocytes in inflammatory arthritis pathology: the emerging role of cadherin-11. Immunol Rev. 233:256–266. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Afuwape AO, Kiriakidis S and Paleolog EM: The role of the angiogenic molecule VEGF in the pathogenesis of rheumatoid arthritis. Histol Histopathol. 17:961–972. 2002.PubMed/NCBI

28 

Mori T, Miyamoto T, Yoshida H, et al: IL-1beta and TNFalpha-initiated IL-6-STAT3 pathway is critical in mediating inflammatory cytokines and RANKL expression in inflammatory arthritis. Int Immunol. 23:701–712. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Feldmann M: Development of anti-TNF therapy for rheumatoid arthritis. Nat Rev Immunol. 2:364–371. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Ehrenstein MR, Evans JG, Singh A, et al: Compromised function of regulatory T cells in rheumatoid arthritis and reversal by anti-TNFalpha therapy. J Exp Med. 200:277–285. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Folgueras AR, Pendas AM, Sanchez LM and Lopez-Otin C: Matrix metalloproteinases in cancer: from new functions to improved inhibition strategies. Int J Dev Biol. 48:411–424. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Okada Y, Gonoji Y, Nakanishi I, Nagase H and Hayakawa T: Immunohistochemical demonstration of collagenase and tissue inhibitor of metalloproteinases (TIMP) in synovial lining cells of rheumatoid synovium. Virchows Arch B Cell Pathol Incl Mol Pathol. 59:305–312. 1990. View Article : Google Scholar : PubMed/NCBI

33 

Ding L, Guo D, Homandberg GA, Buckwalter JA and Martin JA: A single blunt impact on cartilage promotes fibronectin fragmentation and upregulates cartilage degrading stromelysin-1/matrix metalloproteinase-3 in a bovine ex vivo model. J Orthop Res. 32:811–818. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2015
Volume 11 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao J, Zheng W, Wang L and Song B: A disintegrin and metallproteinase 15 knockout decreases migration of fibroblast-like synoviocytes and inflammation in rheumatoid arthritis Retraction in /10.3892/mmr.2024.13165. Mol Med Rep 11: 4389-4396, 2015
APA
Gao, J., Zheng, W., Wang, L., & Song, B. (2015). A disintegrin and metallproteinase 15 knockout decreases migration of fibroblast-like synoviocytes and inflammation in rheumatoid arthritis Retraction in /10.3892/mmr.2024.13165. Molecular Medicine Reports, 11, 4389-4396. https://doi.org/10.3892/mmr.2015.3302
MLA
Gao, J., Zheng, W., Wang, L., Song, B."A disintegrin and metallproteinase 15 knockout decreases migration of fibroblast-like synoviocytes and inflammation in rheumatoid arthritis Retraction in /10.3892/mmr.2024.13165". Molecular Medicine Reports 11.6 (2015): 4389-4396.
Chicago
Gao, J., Zheng, W., Wang, L., Song, B."A disintegrin and metallproteinase 15 knockout decreases migration of fibroblast-like synoviocytes and inflammation in rheumatoid arthritis Retraction in /10.3892/mmr.2024.13165". Molecular Medicine Reports 11, no. 6 (2015): 4389-4396. https://doi.org/10.3892/mmr.2015.3302