Open Access

Receptor tyrosine kinase ligands and inflammatory cytokines cooperatively suppress the fibrogenic activity in temporomandibular‑joint‑derived fibroblast‑like synoviocytes via mitogen‑activated protein kinase kinase/extracellular signal‑regulated kinase

  • Authors:
    • Shikino Matsumoto
    • Seiji Yokota
    • Naoyuki Chosa
    • Seiko Kyakumoto
    • Hitomichi Kimura
    • Masaharu Kamo
    • Kazuro Satoh
    • Akira Ishisaki
  • View Affiliations

  • Published online on: June 25, 2020     https://doi.org/10.3892/etm.2020.8944
  • Pages: 1967-1974
  • Copyright: © Matsumoto 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)-related fibrosis is a possible cause of temporomandibular joint (TMJ) stiffness. However, the molecular mechanisms underlying the fibrogenic activity in fibroblast‑like synoviocytes (FLSs) remain to be clarified. The present study examined the effects of receptor tyrosine kinase (RTK) ligands, such as fibroblast growth factor (FGF)‑1 and epidermal growth factor (EGF), on myofibroblastic differentiation of the FLS cell line FLS1, which is derived from the mouse TMJ. The present study revealed that both FGF‑1 and EGF dose‑dependently suppressed the expression of the myofibroblast (MF) markers, including α‑smooth muscle actin (α‑SMA) and type I collagen, in FLS1 cells. Additionally, both FGF‑1 and EGF activated extracellular signal‑regulated kinase (ERK) in FLS1 cells. In addition, the mitogen‑activated protein kinase (MAPK)/ERK kinase (MEK) inhibitor U0126 abrogated the FGF‑1‑ and EGF‑mediated suppression of MF marker expression. On the other hand, inflammatory cytokines, such as interleukin‑1β and tumor necrosis factor‑α, also suppressed the expression of MF markers in FLS1 cells. Importantly, U0126 abrogated the inflammatory cytokine‑mediated suppression of MF marker expression. Interestingly, RTK ligands and inflammatory cytokines additively suppressed the expression of type I collagen. These results suggested that RTK ligands and inflammatory cytokines cooperatively inhibited the fibrogenic activity in FLSs derived from the TMJ in a MEK/ERK‑dependent manner. The present findings partially clarify the molecular mechanisms underlying the development of OA‑related fibrosis in the TMJ and may aid in identifying therapeutic targets for this condition. Additionally, FGF‑1 and EGF could be therapeutically utilized to prevent OA‑related fibrosis around the inflammatory TMJ.

Introduction

The temporomandibular joint (TMJ) is a synovial joint that is composed of the mandibular fossa of the temporal bone and the mandibular condyle (1). TMJ-osteoarthritis (OA) symptoms include cartilage degeneration, subchondral bone remodeling, and synovitis, which result in TMJ dysfunction (2). Intriguingly, histological studies have shown the presence of extensive fibrosis in the TMJ-OA synovial tissue (3,4), suggesting that fibrotic tissue formation may be responsible for restricted joint movements (5).

We have previously established a fibroblast-like synoviocyte (FLS) cell line, FLS1, from fibroblastic cells derived from a mouse TMJ and found that these cells exhibit myofibroblast (MF)-like fibrogenic characteristics (6). We have also demonstrated that fibroblast growth factor (FGF)-1 alone significantly suppresses the MF differentiation markers α-smooth muscle actin (α-SMA) and type I collagen in FLS1 cells (6). The FGF family consists of 24 members that share 13-71% amino acid identity (7). Although FGF-11-15 are generally considered to belong to the FGF family, they do not activate any FGF receptor (FGFR) (8). However, four FGFRs belong to the receptor tyrosine kinase (RTK) family (9). In general, FGF-1 binds to FGFR1-4(10) to activate various intracellular signaling factors, including phosphoinositide 3-kinase (PI3K)/Akt and mitogen-activated protein kinases (MAPKs), such as extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and p38 MAPK (11). Intriguingly, human synovial fibroblasts derived from the knee synovial tissues express FGF-1 and FGF-R1 proteins (12). However, it remains to be determined which FGF-1-induced intracellular signaling pathway negatively controls the fibrogenic activity in FLSs.

Epidermal growth factor (EGF) was first purified from the mouse salivary gland as a soluble factor that accelerated corneal wound healing (13); however, EGF was soon after found to be a general growth factor that affected various cellular functions involving cell proliferation and differentiation (14). The EGF receptor (EGFR) family consists of four members, namely EGFR/ErbB1/HER1, ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4, all of which belong to the RTK family (15). EGFR dimerizes upon its association with EGF. The cytoplasmic tyrosine kinase domains of EGFR can be autophosphorylated to relay extracellular signals to various intracellular signaling proteins. The carboxy terminal tyrosine residues on EGFR, Tyr1068 and Tyr1173, are the major sites of the autophosphorylation, which occurs as a result of EGF-binding and converts the extracellular EGF signal to intracellular signals (16,17). Autophosphorylated EGFR activates various types of intracellular signaling molecules, including MAPKs and PI3K/Akt (18). Intriguingly, EGF has been detected in the human knee synovial fluid (19). In addition, the EGFR signaling is critical for maintaining the superficial layer of the articular cartilage and preventing OA initiation (20). However, whether EGF-induced intracellular signaling affects the fibrogenic activity in FLSs remains elusive. Furthermore, the mechanism whereby EGF affects the FGF-1-mediated suppression of the fibrogenic activity in FLSs derived from the TMJ synovial tissues warrants investigation.

It is worth noting that inflammatory cytokines, such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) are important factors participating in the pathogenesis of OA. However, the roles of IL-1β and TNF-α in the onset of OA have not been comprehensively studied yet (21). IL-1β binds to the cell membrane receptors IL-1R1 (IL-1RI and CD121a) and IL-1R2 (IL-1RII and CD121b) (22), whereas TNF-α binds to the cell membrane receptors TNF-R1 (p55, CD120a, and TNFRSF1a) and TNFR-2 (p75, CD120b, and TNFRSF1b) (23). In general, IL-1β and TNF-α use similar signal transduction mechanisms to activate nuclear factor-kappa B (NF-κB) and MAPKs, including ERK1/2, JNK, and p38 MAPK. Interestingly, IL-1β and TNF-α both induce MF differentiation in mesenchymal cells in an NF-κB-dependent manner (24,25). In addition, transforming growth factor-β1 (TGF-β1) promotes MF differentiation in an NF-κB-dependent manner as well (26), suggesting that NF-κB-mediated signals positively regulate MF differentiation in mesenchymal cells. Interestingly, IL-1β promotes TGF-β1-induced MF differentiation in nasal fibroblasts in MAPK/ERK kinase (MEK)/ERK-, JNK-, and p38 MAPK-dependent manners (24), whereas TNF-α attenuates TGF-β1-induced MF differentiation in pulmonary fibroblasts in a MEK/ERK-dependent manner (27). These results suggest that MAPK-mediated signals positively or negatively regulate the MF differentiation of mesenchymal cells in a cell-type-specific manner.

Here, we examined the mechanisms whereby the RTK ligands FGF-1 and EGF affect the fibrogenic activity in the myofibroblastic FLS cell line FLS1. We also investigated the effects of FGF-1 and EGF on the activity of PI3K/Akt and MAPKs, such as ERK, JNK, and p38 MAPK, in FLS1 cells and examined whether FGF-1-, or EGF-activated PI3K/Akt or MAPKs affected the status of myofibroblastic differentiation in FLS1 cells. In addition, we examined the cooperative and non-cooperative effects of RTK ligands and inflammatory cytokines, such as IL-1β and TNF-α, on the myofibroblastic differentiation in FLS1 cells. Our study clarified the molecular mechanisms underlying the development of OA-related fibrosis in TMJ and may aid in identifying new therapeutic targets for this condition.

Materials and methods

Reagents

Recombinant mouse EGF was purchased from PeproTech, Inc. Recombinant human IL-1β and TNF-α were obtained from Miltenyi Biotec, GmbH (Bergisch Gladbach). The MEK inhibitors U0126 and PD98059, and the EGFR inhibitor PD153035 were purchased from Calbiochem (Merck KGaA). The NF-κB inhibitor BAY 11-7085 was obtained from Cayman Chemical. Recombinant human FGF-1 and the NF-κB kinase-2 (IKK-2) inhibitor TPCA-1 were purchased from R&D Systems, Inc. The FGFR1 inhibitor SU-5402 was obtained from Wako Pure Chemical Industries, Ltd. We confirmed that dimethyl sulfoxide (DMSO), the vehicle used for the U0126, PD98059, PD153035, BAY 11-7085, TPCA-1, and SU-5402 treatments, did not affect the expression of the MF markers α-SMA and type I collagen (data not shown). Heparin sodium salt was obtained from Merck KGaA. Heparin was included to achieve the optimal FGF-1 activity (28).

Cell culture

The FLS cell line FLS1 was previously established and reported (6): Briefly, to prepare FLSs derived from the mouse TMJ, TMJ synovial tissue was obtained from eight-week-old female mice (C57BL/6J). The tissue was then immersed in digestion solution composed of 20 ml of Ham's F-12 containing 2 mg/ml collagenases consisting of class I and class II collagenases (Collagenase NB4; Wako), at 37˚C for 30 min with continuous vigorous rocking. The cells released from the tissue were transfected with pBABE-puro-simian virus 40 large T antigen (SV40LT) expression plasmid (cat. no. 13970) obtained from Addgene, Inc., with Lipofectamine LTX Reagent (ThermoFisher Scientific, Inc.) according to the manufacturer's protocol. The immortalized FLSs, FLS1 cells were maintained in culture with Ham's F-12 supplemented with 2 mM glutamine, 10% FBS, and penicillin-streptomycin (Invitrogen). These cells were then sub-cultured at a ratio of 1:4 when they reached sub-confluency.

RNA isolation and RT-qPCR

FLS1 cells were seeded into 12-well tissue culture plates at a density of 1x105 cells/well in FLS1 growth medium and maintained for 24 h. The growth medium was replaced with Ham's F-12 containing 0.5% FBS for 24 h for cell starvation. Subsequently, the cells were cultured with or without FGF-1, heparin, EGF, IL-1β, or TNF-α for the indicated periods. Total RNA was isolated from FLS-1 cells using ISOGEN reagent (Nippon Gene) according to the manufacturer's protocol. First-strand cDNA was synthesized from total RNA using the PrimeScript RT reagent Kit (Takara-Bio). PCR was subsequently performed on a Thermal Cycler Dice Real Time System (Takara-Bio) using SYBR Premix Ex Taq II (Takara-Bio), with the following specific oligonucleotide primers: Mouse α-SMA, 5'-CAGATGTGGATACAGCAAACAGGA-3' (forward) and 5'-GACTTAGAAGCATTTGCGGTGGA-3' (reverse); mouse α1 chain of collagen type I (colIα1), 5'-GACATGTTCAGCTTTGTGGACCTC-3' (forward) and 5'-GGGACCCTTAGGCCATTGTGTA-3' (reverse); and mouse GAPDH, 5'-TGTGTCCGTCGTGGATCTG-3' (forward) and 5'-TTGCTGTTGAAGTCGCAGGAG-3' (reverse). The mRNA levels of α-SMA and colIα1 were normalized to GAPDH mRNA levels, and the relative expression levels were calculated as the fold increase or decrease relative to the control.

Western blot analysis

Cells were seeded into 6-well tissue culture plates at a density of 2x105 cells/well in FLS1 growth medium and maintained for 24 h. Afterward, the cells were starved for 24 h as indicated above and cultured with or without FGF-1 plus heparin, EGF, IL-1β, or TNF-α for the indicated periods. Eventualy, the cells were lysed in RIPA buffer [Sigma; 50 mM Tris-HCl (pH 7.2), 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, and 0.1% SDS] or lysis buffer [20 mM HEPES (pH 7.5), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100] containing protease and phosphatase inhibitor cocktails (Sigma). The protein contents of the cell extracts were measured using BCA reagent (Pierce). Extracts containing equal amounts of protein were separated on 10% SDS-polyacrylamide gels and transferred onto polyvinylidenedifluoride membranes (Millipore). After blocking the membranes with 1% BSA or 1% skim milk in T-TBS (50 mM Tris-HCl, pH 7.2, 150 mM NaCl, and 0.05% Tween 20), they were incubated with the appropriate primary antibody. The primary antibodies used included rabbit anti-p44/42 (ERK1/2; cat. no. 9102), rabbit anti-p38 MAPK (cat. no. 9212), rabbit anti-SAPK/JNK (cat. no. 9252), rabbit anti-Akt (cat. no. 9272), rabbit anti-phospho-p44/42 (ERK1/2, Thr202/Tyl204; cat. no. 9101), rabbit anti-phospho-p38 MAPK (Thr180/Tyr182; cat. no. 9211), rabbit anti-phospho-SAPK/JNK (Ther183/185; cat. no. 9251), rabbit anti-phospho-Akt (Ser473; cat. no. 9271) polyclonal antibodies (1:1,000; Cell Signaling Technology), and anti-β-actin antibody (cat. no. sc-47778, 1:1,000; Santa Cruz Biotechnology). The blots were then incubated with the appropriate alkaline phosphatase-conjugated secondary antibody, and signals were detected using an alkaline phosphatase substrate kit (BCIP/NBT Substrate Kit; Vector Laboratories Inc.). Especially, β-actin blots ware obtained from the same membrane as the total ERK1/2 blots after stripping anti-total ERK1/2 antibody from the membranes according to the manufacturer's protocol.

Statistical analysis

Data were presented as mean ± standard deviation (SD; n=4) and statistically analyzed by Tukey's multiple comparison test except for the data analysis in Fig. S1. In Fig. S1, the data were statistically analyzed by Student's t-test. Values of *P<0.01 and **P<0.05 were considered to indicate a statistically significant difference. The results shown in all the experiments are representatives of at least two separate experiments.

Results

FGF-1 suppressed the expression of myofibroblast markers in FLSs

As shown in Fig. 1A, FGF-1 (0.1-1 ng/ml) with heparin (15 µg/ml) significantly downregulated the α-SMA mRNA level in FLS1 cells in a dose-dependent manner. In addition, FGF-1 (0.01-1 ng/ml) with heparin (15 µg/ml) significantly downregulated the colIa1 mRNA level in FLS1 cells in a dose-dependent manner (Fig. 1B). Importantly, we confirmed that the FGFR1 inhibitor SU-5402 (5 µM) significantly abrogated this FGF-1-mediated suppression of α-SMA and colIa1 expression (Fig. 1A and B, respectively). We also confirmed that 15 µM of heparin alone did not significantly affect the mRNA levels of the MF markers α-SMA and type I collagen (Fig. S1) relative to the control cells.

EGF suppressed the expression of myofibroblast markers in FLSs

As shown in Fig. 2A, EGF (0.01-0.1 ng/ml) significantly downregulated the α-SMA mRNA level in FLS1 cells in a dose-dependent manner. In addition, EGF (0.1-1 ng/ml) significantly downregulated the colIa1 mRNA level in FLS1 cells (Fig. 2B). Importantly, we confirmed that the EGFR inhibitor PD153035 (0.5 µM) significantly abrogated this EGF-mediated suppression of α-SMA and colIa1 expression (Fig. 2A and B, respectively).

FGF-1 and EGF promoted phosphorylation of ERK1/ERK2 in FLSs

We used western blotting to evaluate the phosphorylation statuses of ERK1/2, p38 MAPK, JNK, and AKT after the stimulation of FLS1 cells with EGF or FGF-1. As shown in Fig. 3A, strong phosphorylation of ERK1/2 was observed between 5 and 60 min after stimulation with FGF-1 (10 ng/ml) with heparin (15 µg/ml). On the other hand, strong phosphorylation of ERK1/2 was observed at 5-15 min after EGF (10 ng/ml) treatment (Fig. 3B). However, phosphorylated p38 MAPK, JNK, or AKT were not at detectable levels even after treatment with FGF-1 (10 ng/ml) and heparin (15 µg/ml) or EGF (10 ng/ml) alone (data not shown). We also confirmed that β-actin expression was unaffected by the administrations of FGF-1 (10 ng/ml) with heparin (15 µg/ml) or EGF (10 ng/ml) alone at any time points of the treatments (Fig. 3A and B).

FGF-1 and EGF downregulated the mRNA levels of the myofibroblast markers α-SMA and colIa1 in FLSs in a MEK-dependent manner

As shown in Fig. 4A, the MEK inhibitor U0126 (1 µM) partially and significantly reversed the FGF-1 (1 ng/ml) plus heparin (15 µg/ml)-mediated suppression of α-SMA (left graph) and colIa1 (right graph) expression in FLS1 cells, respectively. In addition, U0126 (1 µM) partially and significantly reversed the EGF (5 ng/ml)-mediated suppression of α-SMA (left graph) and colIa1 (right graph) expression, respectively, in FLS1 cells (Fig. 4B). We also found that the MEK inhibitor PD98059 similarly abrogated the FGF-1 (0.25 ng/ml) and heparin (15 µg/ml)-, or EGF (5 ng/ml)-mediated suppression of MF marker expression at concentrations of 5 and 1 µM, respectively (data not shown). Interestingly, FGF-1 (0.1 ng/ml) plus heparin (15 µg/ml) and EGF (0.1 ng/ml) additively downregulated the mRNA levels of α-SMA (left graph) and colIa1 (right graph) in FLS1 cells (Fig. 4C).

RTK ligands and inflammatory cytokines cooperatively inhibited the fibrogenic activity in FLSs in a MEK/ERK-dependent manner

As shown in Fig. 5A, the inflammatory cytokines IL-1β (10 ng/ml) and TNF-α (10 ng/ml) significantly suppressed α-SMA (left graph) and colIa1 (right graph) expression in FLS1 cells. Interestingly, the suppression of α-SMA expression by the combination of the inflammatory cytokines IL-1β (10 ng/ml) and TNF-α (10 ng/ml) was evidently abrogated by U0126 (1 µM) or PD98059 (5 µM; Fig. 5B, left graph). In addition, the suppression of colIa1 expression by the combinatorial stimulation of the inflammatory cytokines was partially but significantly abrogated by U0126 (1 µM), but not by PD98059 (5 µM; Fig. 5B, right graph). However, the NF-κB inhibitor BAY 11-7085 (5 µM) or the IKK-2 inhibitor TPCA-1 (5 µM) did not abrogate the suppression of MF marker expression caused by the combination of the inflammatory cytokines and instead, further downregulated the levels of these MF markers (Fig. S2). As shown in Fig. 5C (left panels), strong phosphorylation of ERK1/2 was observed at 5-15 min after IL-1β (10 ng/ml) stimulation. In addition, strong phosphorylation of ERK1/2 was observed at 30 min after TNF-α (10 ng/ml) stimulation (Fig. 5C, right panels). We also confirmed that β-actin expression was unaffected by the administration of IL-1β (10 ng/ml) or TNF-α (10 ng/ml) at any time point after the stimulation (Fig. 5C). Interestingly, FGF-1 (10 ng/ml) plus heparin (15 µg/ml)- or EGF (10 ng/ml)-mediated suppression of colIa1 expression was further enhanced by the administration of IL-1β (10 ng/ml) or TNF-α (10 ng/ml) (Fig. 5D; left and right graphs, respectively).

Discussion

Bernasconi et al (29), have investigated the morphological modifications occurring in synovial tissue after severe derangement of the articular structure, with dislocation or perforation of the TMJ disks. After histological examination, they have reported remarkable hyperplasia of the synovial tissue, with an increase in the number of myofibroblastic fibroblast-like cells (29). Interestingly, post-traumatic joint stiffness is characterized by an increase in the number of MFs in the joint capsules (30,31), suggesting that MFs retain crucial roles in the pathogenesis of joint stiffness.

MFs are the cells primarily responsible for inducing fibrosis in scleroderma, renal fibrosis, pulmonary fibrosis, and liver fibrosis (32). MFs retain contractile properties and produce a large quantity of extracellular molecules, such as type I collagen (33). The most widely recognized molecular marker of differentiated and activated MFs is the de novo expression of α-SMA (34). We have previously established an FLS cell line, FLS1, which is derived from a mouse TMJ. We have reported that FLS1 cells express higher levels of MF marker molecules, such as α-SMA and type I collagen than mouse NIH3T3 embryonic fibroblasts, which are frequently used as a standard fibroblast control (6). Thus, the FLS1 cell line is a suitable experimental model for the investigation of the molecular mechanisms underlying the extensive fibrosis observed in the TMJ-OA synovial tissue. We have previously demonstrated that Rho-associated coiled-coil-forming kinase (ROCK)-mediated actin-polymerization, which induces the translocation of myocardin-related transcription factor (MRTF) from the cytoplasm to the nucleus, promotes MF differentiation in FLS1 cells (6). However, it remains to be clarified what intra-cellular signals other than the ROCK-mediated signal affect the fibrogenic activity in FLS1 cells.

Here, we demonstrated that FGF-1 and EGF dose-dependently downregulated the mRNA levels of the MF differentiation markers α-SMA and type I collagen in FLS1 cells (Figs. 1 and 2). In addition, we found that ERK1/2-mediated signaling played an important role in these anti-fibrogenic effects of FGF-1 and EGF (Figs. 3 and 4). These results strongly suggested that FGF-1 and EGF suppressed the fibrogenic activity in FLSs in a MEK/ERK-dependent manner. However, U0126 only partially abrogated the FGF-1- or EGF-mediated downregulation of MF differentiation markers in FLS1 cells, suggesting that signaling pathways other than MEK/ERK play important roles (Fig. 4). Interestingly, we have previously demonstrated that EGF suppresses the expression of MF differentiation markers in periodontal ligament-derived endothelial progenitor cells (EPCs, SCDC2) through MEK- and JNK-dependent signaling pathways (35), whereas the activation of JNK was not detected after the EGF stimulation of FLS1 cells (data not shown). These results suggested that EGF differentially induced intracellular signals in EPCs and FLSs and had a negative effect on myofibroblastic differentiation. On the other hand, the combination of the inflammatory cytokines IL-1β and TNF-α suppressed the expression of the MF markers α-SMA (Fig. 5B, left graph) and type I collagen (Fig. 5B, right graph), and this effect was significantly abrogated by the MEK1/2 inhibitor U0126. In contrast, PD98059, which is known as a MEK1 inhibitor, but not a MEK1/2 inhibitor, in a cell-type specific manner (36), significantly abrogated the suppression of α-SMA expression by the combinatorial treatment with the inflammatory cytokines (Fig. 5B, left graph), but did not abrogate the suppression of type I collagen expression (Fig. 5B, right graph), suggesting that MEK2-mediated intracellular signaling played an important role in the suppression of type I collagen expression in FLSs after stimulation with inflammatory cytokines. Importantly, neither an NF-κB inhibitor nor an IKK-2 inhibitor abrogated the IL-1β- and TNF-α-mediated suppression of MF marker expression (Fig. S2), suggesting that these effects of the inflammatory cytokines in FLSs were not mediated by NF-κB. Interestingly, the NF-κB and IKK-2 inhibitors further decreased the expression levels of the MF markers in FLS1 cells treated with the inflammatory cytokines (Fig. S2), suggesting that the NF-κB-mediated signaling possibly positively regulated the MF marker expression in FLSs. In addition, we confirmed that IL-1β and TNF-α induced ERK1/2 phosphorylation in FLS1 cells (Fig. 5C). Moreover, these inflammatory cytokines further enhanced the FGF-1- or EGF-mediated suppression of type I collagen expression (Fig. 5D). These results strongly suggested that RTK ligands and inflammatory cytokines cooperatively inhibited the fibrogenic activity in FLSs in a MEK/ERK-dependent manner. Interestingly, Ma et al (37) have previously reported that EGF and IL-1β synergistically promote ERK1/2-mediated invasive breast ductal cancer cell migration and invasion. In addition, Ziv et al (38) and Kakiashvili et al (39) have reported that TNF-α activates ERK1/2 through EGFR activation in epithelial cells. However, it remains to be determined whether IL-1β and TNF-α activate ERK1/2 through EGFR activation in FLSs.

Our findings partially clarify the molecular mechanisms underlying the development of OA-related fibrosis in the TMJ and may aid in identifying novel therapeutic targets for this condition. In addition, FGF-1 and EGF may be used to prevent OA-related fibrosis around inflammatory TMJs.

Supplementary Material

Heparin itself does not affect the expression of the myofibroblast markers α-SMA and colIα1 in fibroblast-like synoviocytes. Cells were starved and then cultured with heparin (15 μg/ml) alone for 24 h. The relative expression levels of the myofibroblast markers α-SMA and colIα1 were then evaluated using reverse transcription-quantitative PCR. Data represent the mean ± SD (n=4). α-SMA, α-smooth muscle actin; colIα1, α1 chain of collagen type I.
Neither the NF‑κB inhibitor BAY 11-7085 nor the IKK-2 inhibitor TPCA-1 abrogated the IL-1β- and TNF-α-mediated suppression of the myofibroblast marker expression in fibroblast-like synoviocytes. Cells were starved and then cultured with IL-1β (10 ng/ml) and TNF-α (10 ng/ml), with or without the NF-κB inhibitor BAY 11-7085 or the IKK-2 inhibitor TPCA-1 for 24 h. The relative expression levels of the myofibroblast markers α-SMA and colIα1 were then evaluated using reverse transcription-quantitative PCR. Data are presented as the mean ± SD (n=4). *P<0.01. α-smooth muscle actin; colIα1, α1 chain of collagen type I; IL, interleukin; TNF-α, tumor necrosis factor α.

Acknowledgements

Not applicable.

Funding

The present study was supported by JSPS KAKENHI (grant nos. JP16H05534 to AI, JP16K11654 to NC, JP17K11851 to MK and JP19K19277 to SY).

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

SM, SY, NC, SK, HK, MK and KS performed RT-qPCR and western blotting. SM and AI designed the present study. AI was a major contributor in writing the manuscript. 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 

Ibi M: Inflammation and temporomandibular joint derangement. Biol Pharm Bull. 42:538–542. 2019.PubMed/NCBI View Article : Google Scholar

2 

Wang XD, Zhang JN, Gan YH and Zhou YH: Current understanding of pathogenesis and treatment of TMJ osteoarthritis. J Dent Res. 94:666–673. 2015.PubMed/NCBI View Article : Google Scholar

3 

Dijkgraaf LC, Liem RS and de Bont LG: Synovial membrane involvement in osteoarthritic temporomandibular joints: A light microscopic study. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 83:373–386. 1997.PubMed/NCBI View Article : Google Scholar

4 

Dijkgraaf LC, Liem RS and de Bont LG: Ultrastructural characteristics of the synovial membrane in osteoarthritic temporomandibular joints. J Oral Maxillifac Surg. 55:1269–1279. 1997.PubMed/NCBI View Article : Google Scholar

5 

Dijkgraaf LC, Zardeneta G, Cordewener FW, Liem RS, Schmitz JP, de Bont LG and Milam SB: Crosslinking of fibrinogen and fibronectin by free radicals: A possible initial step in adhesion formation in osteoarthritis of the temporomandibular joint. J Oral Maxillofac Surg. 61:101–111. 2003.PubMed/NCBI View Article : Google Scholar

6 

Yokota S, Chosa N, Kyakumoto S, Kimura H, Ibi M, Kamo M, Satoh K and Ishisaki A: ROCK/actin/MRTF signaling promotes the fibrogenic phenotype of fibroblast-like synoviocytes derived from the temporomandibular joint. Int J Mol Med. 39:799–808. 2017.PubMed/NCBI View Article : Google Scholar

7 

Chen TM, Chen YH, Sun HS and Tsai SJ: Fibroblast growth factors: Potential targets for regenerative therapy of osteoarthritis. Chin J Physiol. 62:2–10. 2019.PubMed/NCBI View Article : Google Scholar

8 

Beenken A and Mohammadi M: The fgf family: Biology, pathophysiology and therapy. Nat Rev Drug Discov. 8:235–253. 2009.PubMed/NCBI View Article : Google Scholar

9 

Liu F and Zhuang S: Role of receptor tyrosine kinase signaling in renal fibrosis. Int J Mol Sci. 17(E972)2016.PubMed/NCBI View Article : Google Scholar

10 

Omitz DM, Xu J, Colvin JS, McEwen DG, MacArthur CA, Couller F, Gao G and Goldfarb M: Receptor specificity of the fibroblast growth factor family. J Biol Chem. 271:15292–15297. 1996.PubMed/NCBI View Article : Google Scholar

11 

Raju R, Palapetta SM, Sandhya VK, Sahu A, Alipoor A, Balakrishnan L, Advani J, George B, Kini KR, Geetha NP, et al: A network map of FGF-1/FGFR signaling system. J Signal Transduct. 2014(962962)2014.PubMed/NCBI View Article : Google Scholar

12 

Zhu L, Weng Z, Shen P, Zhou J, Zeng J, Weng F, Zhang X and Yang H: S100B regulates inflammatory response during osteoarthritis via fibroblast growth factor receptor 1 signaling. Mol Med Rep. 18:4855–4864. 2018.PubMed/NCBI View Article : Google Scholar

13 

Cohen S and Elliott GA: The stimulation of epidermal keratinization by a protein isolated from the submaxillary gland of the mouse. J Invest Dermatol. 40:1–5. 1963.PubMed/NCBI View Article : Google Scholar

14 

Tsai CJ and Nussinov R: Emerging mechanism of EGFR activation in physiological and pathological contexts. Biophys J. 117:5–13. 2019.PubMed/NCBI View Article : Google Scholar

15 

Lemmmon MA and Schlessinger J: Cell signaling by receptor tyrosine kinase. Cell. 141:1117–1134. 2010.PubMed/NCBI View Article : Google Scholar

16 

Dawnward J, Waterfield MD and Parker PJ: Autophosphorylation and protein kinase C phosphorylation of the epidermal growth factor receptor. Effect on tyrosine kinase activity and ligand binding affinity. J Biol Chem. 260:14538–14546. 1985.PubMed/NCBI

17 

Helin K, Velu T, Martin P, Vass WC, Allevato G, Lowy DR and Beguinot L: The biological activity of the human epidermal growth factor receptor is positively regulated by its C-terminal tyrosines. Oncogene. 6:825–832. 1991.PubMed/NCBI

18 

Wee P and Wang Z: Epidermal growth factor receptor cell proliferation signaling pathways. Cancers (Basel). 9(E52)2017.PubMed/NCBI View Article : Google Scholar

19 

Ren G, Lutz L, Raiton P, Wiley JP, McAllister J, Powell J and Krawetz RJ: Serum and synovial fluid cytokine profiling in hip osteoarthritis: Distinct from knee osteoarthritis and correlated with pain. BMC Musculoskelet Disord. 19(39)2018.PubMed/NCBI View Article : Google Scholar

20 

Jia H, Ma X, Tong W, Doyran B, Sun Z, Wang L, Zhang X, Zhou Y, Badar F, Chandra A, et al: EGFR signaling is critical for maintaining the superficial layer of articular cartilage and preventing osteoarthritis initiation. Proc Natl Acad Sci USA. 113:14360–14365. 2016.PubMed/NCBI View Article : Google Scholar

21 

Wojdasiewicz P, Poniatowski ŁA and Szukiewicz D: The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm. 2014(561459)2014.PubMed/NCBI View Article : Google Scholar

22 

Boraschi D and Tagliabue A: The interleukin-1 receptor family. Semin Immunol. 25:394–407. 2013.PubMed/NCBI View Article : Google Scholar

23 

MacEwan DJ: TNF receptor subtype signaling: Differences and cellular consequences. Cell Signal. 14:477–492. 2002.PubMed/NCBI View Article : Google Scholar

24 

Shin JM, Kang JH, Lee SA, Park IH and Lee HM: Baicalin downregulates IL-1β-stimulated extracellular matrix production in nasal fibroblasts. PLoS One. 11(e0168195)2016.PubMed/NCBI View Article : Google Scholar

25 

Hou J, Ma T, Cao H, Chen Y, Wang C, Chen X, Xiang Z and Han X: TNF-α induced NF-κB activation promotes myofibroblast differentiation of LR-MSCs and exacerbates bleomycin-induced pulmonary fibrosis. J Cell Physiol. 233:2409–2419. 2018.PubMed/NCBI View Article : Google Scholar

26 

Mia MM and Bank RA: The IκB kinase inhibitor strongly attenuates TGFβ1-induced myofibroblast formation and collagen synthesis. J Cell Mol Med. 19:2780–2792. 2015.PubMed/NCBI View Article : Google Scholar

27 

Liu X, Kelm RJ Jr and Strauch AR: Transforming growth factor beta1-mediated activation of the smooth muscle alpha-actin gene in human pulmonary myofibroblasts is inhibited by tumor necrosis factor-alpha via mitogen-activated protein kinase 1-dependent induction of the Egr-1 transcriptional repressor. Mol Biol Cell. 20:2174–2185. 2009.PubMed/NCBI View Article : Google Scholar

28 

Harmer NJ: Insights into the role of heparin sulphate in fibroblast growth factor signaling. Biochem Soc Trans. 34:442–445. 2006.PubMed/NCBI View Article : Google Scholar

29 

Bernasconi G, Marchetti C, Reguzzoni M and Baciliero U: Synovia hyperplasia and calcification in the human TMJ disk: A clinical, surgical, and histologic study. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 84:245–252. 1997.PubMed/NCBI View Article : Google Scholar

30 

Hildebrand KA, Zhang M and Hart DA: Myofibroblast upregulators are elevated in joint capsules in posttraumatic contractures. Clin Orthop Relat Res. 456:85–91. 2007.PubMed/NCBI View Article : Google Scholar

31 

Hildebrand KA, Zhang M, van Snellenberg W, King GJ and Hart DA: Myofibroblast numbers are elevated in human elbow capsules after trauma. Clin Orthop Relat Res. 189–197. 2004.PubMed/NCBI View Article : Google Scholar

32 

Hinz B, Phan SM, Thannickal VJ, Plunotto M, Desmouliére A, Varga J, De Wever O, Mareel M and Gabbiani G: Recent developments in myofibroblast biology: Paradigms for connective tissue remodeling. Am J Pathol. 180:1340–1355. 2012.PubMed/NCBI View Article : Google Scholar

33 

van Caam A, Vonk M, van den Hoogen F, van Lent P and van der Kraan P: Unraveling SSc pathophysiology; The myofibroblast. Front Immunol. 9(2452)2018.PubMed/NCBI View Article : Google Scholar

34 

Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML and Gabbiani G: The myofibroblast: one function, multiple organs. Am J Pathol. 170:1807–1816. 2007.PubMed/NCBI View Article : Google Scholar

35 

Kimura H, Okubo N, Chosa N, Kyakumoto S, Kamo M, Miura H and Ishisaki A: EGF positively regulates the proliferation and migration, and negatively regulates myofibroblast differentiation of periodontal ligament-derived endothelial progenitor cells through MEK/ERK- and JNK-dependent signals. Cell Physiol Biochem. 32:899–914. 2013.PubMed/NCBI View Article : Google Scholar

36 

Lee HR, Lee J and Kim HJ: Differential effects of MEK inhibitor on rat neural stem cell differentiation: Repressive roles of MEK2 in neurogenesis and induction of astrocytogenesis by PD98059. Pharmacol Res. 149(104466)2019.PubMed/NCBI View Article : Google Scholar

37 

Ma L, Lan F, Zheng Z, Xie F, Wang L, Liu W, Han J, Zheng F, Xie Y and Huang Q: Epidermal growth factor (EGF) and interleukin (IL)-1β synergistically promote ERK1/2-mediated invasive breast ductal cancer cell migration and invasion. Mol Cancer. 11(79)2012.PubMed/NCBI View Article : Google Scholar

38 

Ziv E, Rotem C, Miodovnik M, Ravid A and Koren R: Two modes of ERK activation by TNF in keratinocytes: Different cellular outcomes and bi-directional modulation by vitamin D. J Cell Biochem. 104:606–619. 2008.PubMed/NCBI View Article : Google Scholar

39 

Kakiashvili E, Dan Q, Vandermeer M, Zhang Y, Waheed F, Pham M and Száski K: The epidermal growth factor receptor mediates tumor necrosis factor-alpha-induced activation of the ERK/GEF-H1/RhoA pathway in tubular epithelium. J Biol Chem. 286:9268–9279. 2011.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2020
Volume 20 Issue 3

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
Matsumoto S, Yokota S, Chosa N, Kyakumoto S, Kimura H, Kamo M, Satoh K and Ishisaki A: Receptor tyrosine kinase ligands and inflammatory cytokines cooperatively suppress the fibrogenic activity in temporomandibular‑joint‑derived fibroblast‑like synoviocytes via mitogen‑activated protein kinase kinase/extracellular signal‑regulated kinase. Exp Ther Med 20: 1967-1974, 2020
APA
Matsumoto, S., Yokota, S., Chosa, N., Kyakumoto, S., Kimura, H., Kamo, M. ... Ishisaki, A. (2020). Receptor tyrosine kinase ligands and inflammatory cytokines cooperatively suppress the fibrogenic activity in temporomandibular‑joint‑derived fibroblast‑like synoviocytes via mitogen‑activated protein kinase kinase/extracellular signal‑regulated kinase. Experimental and Therapeutic Medicine, 20, 1967-1974. https://doi.org/10.3892/etm.2020.8944
MLA
Matsumoto, S., Yokota, S., Chosa, N., Kyakumoto, S., Kimura, H., Kamo, M., Satoh, K., Ishisaki, A."Receptor tyrosine kinase ligands and inflammatory cytokines cooperatively suppress the fibrogenic activity in temporomandibular‑joint‑derived fibroblast‑like synoviocytes via mitogen‑activated protein kinase kinase/extracellular signal‑regulated kinase". Experimental and Therapeutic Medicine 20.3 (2020): 1967-1974.
Chicago
Matsumoto, S., Yokota, S., Chosa, N., Kyakumoto, S., Kimura, H., Kamo, M., Satoh, K., Ishisaki, A."Receptor tyrosine kinase ligands and inflammatory cytokines cooperatively suppress the fibrogenic activity in temporomandibular‑joint‑derived fibroblast‑like synoviocytes via mitogen‑activated protein kinase kinase/extracellular signal‑regulated kinase". Experimental and Therapeutic Medicine 20, no. 3 (2020): 1967-1974. https://doi.org/10.3892/etm.2020.8944