Open Access

Inhibition of osteoclastogenesis for periprosthetic osteolysis therapy through the suppression of p38 signaling by fraxetin

  • Authors:
    • Jia‑Cheng Liao
    • Zhao‑Xia Wei
    • Chang Zhao
    • Zhong‑Ping Ma
    • Dao‑Zhang Cai
  • View Affiliations

  • Published online on: May 21, 2018     https://doi.org/10.3892/ijmm.2018.3698
  • Pages: 1257-1264
  • Copyright: © Liao 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

Periprosthetic osteolysis belongs to osteolytic diseases, which often occur due to an imbalance between osteoclast and osteoblast number or activity. Fraxetin, a natural plant extract, inhibits osteoblast apoptosis and has therapeutic potential for treating osteolytic diseases. However, data pertaining to the effects of fraxetin on osteoclasts are limited. In the present study, it was demonstrated that the inhibition of osteoclastogenesis by fraxetin had an important role on the therapy of titanium particle‑induced osteolysis in vivo. In addition, fraxetin was demonstrated to suppress receptor activator of nuclear factor‑κB ligand (RANKL)‑mediated osteoclast differentiation and bone resorption in vitro in a dose‑dependent manner. Fraxetin inhibited osteoclast differentiation and function through the suppression of p38 signaling and subsequently, the suppression of osteoclast‑specific gene expression, including tartrate‑resistant acid phosphatase, nuclear factor of activated T‑cells, cytoplasmic 1, and cathepsin K. In conclusion, fraxetin administration may have potential as a treatment method for periprosthetic osteolysis and other osteolytic diseases.

Introduction

Bone tissue is constantly breaking-down and building-up to maintain skeleton balance. The bone remodeling process involves bone formation (osteoblast activities) and bone resorption (osteoclast activities) (1). Excess osteoclast activity may lead to adult skeletal osteolytic diseases, including osteoporosis, rheumatoid arthritis, and periodontal disease (24). Periprosthetic osteolysis (PO) following total hip arthroplasty (THA) is also a typical osteolytic disease. As a highly successful procedure, THA has become the standard method to address serious joint disease. But aseptic loosening, caused by PO, leads frequently to prosthetic failure. Previous studies have reported that PO is initiated by inflammatory response to wear debris (5,6), then excessive osteoclast formation and bone resorption are stimulated, resulting in osteopenia (4,7).

Osteoclasts are large multinucleated cells, which arise from the hematopoietic monocyte/macrophage lineage (8). Macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) are required for osteoclast differentiation and formation (9). RANKL induces osteoclast differentiation by binding to RANK, its cognate receptor (10). Then, downstream signaling pathways are activated, including nuclear factor-κB (NF-κB), c-Jun N-terminal kinase (JNK) 1/2, p38, and extracellular signal-regulated kinase (ERK) 1/2 pathways (10,11). Suppression of these pathways might provide a potential for treating PO through inhibiting osteoclast formation or function.

Fraxetin (7, 8-dihydroxy-6-methoxy coumarin; Fig. 1A), extracted from the Chinese herb Cortex Fraxini, has anti-inflammatory, antitumor, antioxidative, antiviral, antihyperglycemic and neuroprotective effects (12,13). In addition, Kuo et al (14) reported that fraxetin can inhibit interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and anti-first apoptosis signal (Fas) IgM-mediated apoptosis by suppressing the Fas signal pathway in osteoblastic MG-63 cells. These findings indicate that fraxetin might be a potential therapeutic option to prevent osteolytic diseases. However, the role of fraxetin in osteoclasts has not been reported.

The present study aimed to investigate the therapeutic benefits and mechanism of fraxetin on PO in vivo, to assess the effect of fraxetin on osteoclastogenesis and function in vitro, and to determine the mechanisms that mediate the effects of fraxetin on osteoclast development and function.

Materials and methods

Reagents

Mouse macrophage RAW264.7 cells were obtained from the American Type Culture Collection (Manassas, VA, USA). Fetal bovine serum (FBS) was obtained from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The Cell Counting Kit (CCK)-8 was purchased from Dojindo Molecular Technologies, Inc., (Kumamoto, Japan). Alpha-modified Eagle’s medium (α-MEM) was obtained from Gibco; Thermo Fisher Scientific, Inc. Fraxetin, glycine, Tris, sodium dodecyl sulfate (SDS), NaCl, Acid Phosphatase kit and other reagents were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). M-CSF and RANKL were purchased from R&D Systems, Inc. (Minneapolis, MN, USA). The SYBR Premix Ex Taq II and Prime Script RT reagent kits were purchased from Takara Bio, Inc. (Otsu, Japan). Antibodies against phosphorylated (p-) JNK1/2 (cat. no. 9251), total JNK1/2 (cat. no. 9252), p-p38 (cat. no. 9211), total p38 (cat. no. 8690), p-ERK1/2 (cat. no. 9101), total ERK1/2 (cat. no. 9102), MMK6 (cat. no. 9264) and β-actin (cat. no. 4967) were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA) and the working dilution was 1:1,000 for all.

Animals and experimental design

This study obtained ethics approval by the Institutional Animal Care And Use Committee (IACUC) of Southern Medical University (SMU; Guangzhou, China), and all experiments involving animals were performed according to the guidelines of SMU IACUC. A mouse calvarial osteolysis model was established to investigate the effects of fraxetin in vivo (15). Commercial pure Titanium (Ti) particles were obtained from Johnson Matthey (Royston, UK) (16). The particles were exposed to 180°C for 6 h, and sterilized with 70% ethanol for 48 h to remove adherent endotoxins. The particles with an average diameter of 4.50 mm are similar to wear particles retrieved from periprosthetic tissues (17,18). The concentration of particles in subsequent studies was 0.1 mg/ml. Healthy 8 week-old male C57BL/J6 mice, weighing 23±1 g, were purchased from Southern Medical University Laboratory Animal Center and assigned randomly to three experimental groups (8 mice per group). The sham group was used as a negative control. The vehicle group had calvarial osteolysis mediated by Ti particles. The fraxetin group had calvarial osteolysis and was treated with fraxetin (5 mg/kg/day). For the generation of the calvarial osteolysis model, the Ti particles (30 mg) were embedded on the middle suture of the calvaria (19). Two days later, fraxetin or PBS were locally injected every other day. All animals were housed under conditions of constant temperature and humidity on a 12-h light/dark cycle. Food and water were available ad libitum. After two weeks, the mice were sacrificed, and the calvarias were excised, fixed in 4% paraformaldehyde for 15 h and stored at 70% ethanol for micro-computed tomography (µ-CT) analysis.

µ-CT scanning and histological analysis

The prepared calvarias were analyzed by high resolution µ-CT. Bones were scanned at an isometric voxel resolution of 8 µm. The X-ray energy parameters were 80 kV and 80 µA. A square region of interest (ROI) was selected to perform the subsequent analyses. Porosity, number of pores, and bone volume/tissue volume (BV/TV) were studied for the ROI (20).

After µ-CT scanning, bones were decalcified in 10% ethylenediaminetetraacetic acid (EDTA; pH 7.4) for 2–3 weeks prior to embedding in paraffin. Sections with thickness of 4-µm were prepared and stained with tartrate-resistant acid phosphatase (TRAP) and hematoxylin and eosin (H&E). Stained sections were observed and photographed on a Leica DM1000 light microscope. At least 3 fields per slide per group were analyzed. For calvarias, the osteolysis area was analyzed using Image J (National Institutes of Health, Bethesda, MD, USA). TRAP-positive osteoclast numbers were determined.

Osteoclast culture, TRAP staining assay, resorption pit assay and osteoclastogenesis rescue assay

Primary mouse bone marrow-derived macrophages (BMMs) were obtained from the bone marrow of long bones. BMMs were cultured in α-MEM containing 10% FBS, 100 U/ml penicillin/streptomycin, 2 mM L-glutamine, and 30 ng/ml M-CSF (complete α-MEM). α-MEM containing 10% FBS, 100 U/ml penicillin/streptomycin and 2 mM L-glutamine was used for RAW264.7 cell culture.

BMMs were seeded at a density of 8×103 cells/well into a 96-well plate with complete α-MEM, RANKL (50 ng/ml), and fraxetin (0, 3, 6, or 12 µM). For the resorption pit assay, bovine bone slices were added into the 96-well plate previously. For the osteoclastogenesis rescue assay, anisomycin (2.5 ng/ml), a potent activator of p38, was added following the fraxetin treatment (21,22). After seven days, the mature osteoclasts were fixed with 4% paraformaldehyde for 30 min and stained using the Acid Phosphatase kit. Osteoclast-like (OCL) cells were characterized as TRAP-positive osteoclasts with >3 nuclei. For the resorption pit assay, mechanical agitation and sonication were used to remove OCL cells from the bone slices. Resorption pits were observed using a scanning electron microscope. For each group, three wells were selected randomly for analysis. Image J software was used to analyze the numbers and the area of OCL cells and the % of resorbed bone surface area.

Cytotoxicity assay

The effects of fraxetin on the viability of BMMs were determined by CCK-8 assay. BMMs were plated at a density of 8,000 cells/well in 96-well plates, and cultured in complete α-MEM containing fraxetin (0, 6.25, 12.5, 25, 50, 100, 200, 400 and 800 µM) for 48 h. GraphPad Prism (GraphPad Software, Inc., La Jolla, CA, USA) was used to calculate the half-maximal inhibitory concentration (IC50).

Western blot analysis

RAW264.7 cells were seeded in 6-well plates at a density of 5×105 cells/well. First, RAW264.7 cells were treated with placebo or fraxetin for 2 h. Then, the RAW264.7 cells were stimulated with RANKL for 0, 5, 10, 20, 30, or 60 min. Subsequently, cells lysates were collected and western blotting was performed (23). The Odyssey infrared imaging system (LI-COR Biosciences, Lincoln, NE, USA) was used to visualize the antibody reactivity.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay

BMMs were seeded at a density of 2×105 cells/well in 6-well plates and cultured in complete α-MEM supplemented with 50 ng/ml RANKL. Then, the BMMs were treated with either 0, 3, or 12 µM fraxetin for 5 days, or 12 µM fraxetin for 0–5 days. A Qiagen RNeasy Mini kit (Qiagen GmbH, Hilden, Germany) was used to extract total RNA. Reverse transcription was performed with the Prime Script RT kit. qPCR was performed using SYBR Premix Ex Taq II with the following specific primers: TRAP, forward 5′-CTG GAG TGC ACG ATG CCA GCG ACA-3′ and reverse 5′-TCC GTG CTC GGC GAT GGA CCA GA-3′; nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1), forward 5′-CCG TTG CTT CCA GAA AAT AAC A-3′ and reverse 5′-TGT GGG ATG TGA ACT CGG AA-3′; cathepsin K (CTSK), forward 5′-CTT CCA ATA CGT GCA GCA GA-3′ and reverse 5′-TCT TCA GGG CTT TCT CGT TC-3′; GAPDH, forward 5′-ACC CAG AAG ACT GTG GAT GG-3′ and reverse 5′-CAC ATT GGG GGT AGG AAC AC-3′. The following cycling conditions were used: 40 cycles of denaturation at 95°C for 5 sec and amplification at 60°C for 24 sec. GAPDH expression was used to normalize transcript levels for the genes under investigation, and all reactions were run in triplicate. The 2−ΔΔCq method was used for quantification (24).

Statistical analysis

Results are expressed as the mean ± standard deviation. The significance of differences between groups was analyzed with one-way analysis of variance with a post hoc Bonferroni test, using SPSS 18.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Fraxetin protects against bone loss induced by wear particles

A Ti particle-induced calvarial osteolysis model was used to simulate PO. Analysis of parietal bones by µ-CT revealed extensive bone resorption in mice exposed to Ti particles (vehicle group; received Ti injection) compared with the negative controls (sham group; received PBS only injection; Fig. 1B). Bone resorption was lower in mice administered a 5 mg/kg/day concentration of fraxetin compared with the vehicle group (Fig. 1B), demonstrating that fraxetin suppressed osteolysis induced by Ti-particles. Quantification of bone parameters revealed that exposure to fraxetin increased bone volume (Fig. 1C) and decreased porosity (Fig. 1D and E) compared with the vehicle group.

Histological assessment results demonstrated that fraxetin administration prevented parietal bones from Ti particle-induced osteolysis. H&E staining revealed few osteolytic changes in the sham group mice. Multiple osteolytic changes occurred in the vehicle group, whereas fraxetin administration reversed opposed this effect (Fig. 1B). Accordingly, TRAP staining revealed that the number of OCLs was increased following Ti particle stimulation, while fraxetin administration reversed this effect (Fig. 1B and F). In summary, fraxetin could prevent wear particle-induced bone loss by suppressing osteoclastogenesis.

Fraxetin inhibits osteoclast formation and function in vitro

Based on the aforementioned in vivo results, it was hypothesized that fraxetin can inhibit osteoclastogenesis in vitro. The role and mechanism of fraxetin in normal osteoclast formation and bone resorption function were therefore explored further in vitro. First, CCK-8 assays were performed in order to exclude the possibility that a potential cytotoxic effect of fraxetin may be responsible for the decrease in OCL cell numbers. The IC50 value of fraxetin was 217.9 µM at 48 h, suggesting that fraxetin might suppress the proliferation of BMM cells at concentrations >217.9 µM (Fig. 2A). Then, differentiated BMMs were induced into TRAP-positive OCL cells (Fig. 2B). Exposure to fraxetin decreased osteoclast formation in a dose dependent manner. The numbers of TRAP-positive OCL cells was suppressed by ~30% in the presence of 3 µM fraxetin (Fig. 2B–D), and was almost completely suppressed by exposure to fraxetin at concentrations >12 µM (Fig. 2 B–D). Since the IC50 value of fraxetin was demonstrated to be 217.9 µM, fraxetin could not have affected the proliferation of BMMs at doses 6–12 µM, suggesting that at these latter concentrations of fraxetin impaired osteoclast formation without cell cytotoxicity.

Bone resorption is the most important function of osteoclasts. Therefore, bone resorption assays were conducted in vitro in order to evaluate the effect of fraxetin on bone resorption. Scanning electron microscopy analysis revealed osteoclastic bone resorption pits present in BMM cultures in the presence of RANKL. Exposure of BMMs to 3 µM fraxetin resulted in a reduction in resorptive activity >50%, and at fraxetin concentrations >12 µM, resorption was completely suppressed (Fig. 2B and E). These results demonstrate that fraxetin administration reduced Ti particle-induced bone resorption in the presence of RANKL in vitro.

Fraxetin suppresses osteoclast activity by downregulating osteoclast-specific gene expression

RT-qPCR results demonstrated that the mRNA expression levels of TRAP, CTSK and NFATc1 were significantly decreased following fraxetin treatment, compared with untreated control cells, in a time- and dose-dependent manner (Fig. 3). These findings suggest that fraxetin inhibited osteoclast differentiation by suppressing osteoclast-specific gene expression.

Fraxetin suppresses osteoclast activity by specifically inhibiting the p38 signaling pathway

Osteoclasts were differentiated from monocytes and macrophages in response to stimulation with M-CSF and RANKL. The RAW264.7 cell line has good stability and can substitute for preosteoclasts in vitro. Western blot analysis was conducted in RAW264.7 cells in order to investigate the activity of signaling pathways that determine osteoclast differentiation. Well known, mitogen-activated protein kinase (MAPK) pathways (namely ERK1/2, p38 and JNK1/2) have an important role in osteoclast differentiation downstream of RANK signaling (25). The present western blot results revealed that stimulation of RAW264.7 cells with RANKL induced peak p38 phosphorylation within 10 min, and this effect was suppressed following treatment with 12 µM fraxetin (Fig. 4A). Densitometry analysis of western blot images confirmed the significant inhibition of p38 phosphorylation by fraxetin (Fig. 4B). To confirm these findings, BMMs exposed to fraxetin were treated with anisomycin, a p38 agonist (21,22). In accordance with previous observations, fraxetin inhibited osteoclast formation, whereas anisomycin precluded this effect (Fig. 4C–E). Conversely, in both the control and fraxetin-treated groups, ERK and JNK phosphorylation were induced, and fraxetin treatment did not oppose this effect (Fig. 4A). Furthermore, MKK6, an upstream regulator of p38, was not affected by RANKL stimulation or fraxetin treatment (Fig. 4A). Taken together, these results demonstrated that fraxetin suppressed osteoclast differentiation by inhibiting p38 signaling, whereas JNK and ERK activity was not affected.

Discussion

Previous studies have reported that fraxetin is a potential anti-osteolytic agent, based on its properties to inhibit osteoblast apoptosis induced by inflammatory cytokines (14). However, osteoclasts serve an important role in osteolytic diseases, and the effects of fraxetin on these cells have not been characterized. In the present study, fraxetin was demonstrated to suppress PO via inhibition of the stimulatory effects of RANKL-induced p38 signaling on osteoclast formation and bone resorption in vitro and vivo. Thus, the present findings provide a mechanistic justification for the application of fraxetin to the treatment of PO.

Results from the Ti particle-induced bone loss assay indicated that the inhibition of osteoclastogenesis by fraxetin in vivo may have an important role on the therapy of PO. Additionally, the inhibitory effect of fraxetin on osteoclastogenesis and bone resorption were further investigated in vitro. The results demonstrated that fraxetin prevented the stimulatory effects of RANKL through the p38 cascade. Activated p38 signaling is a key procedure in bone destruction, while suppression of p38 activity reduces osteoclast formation and bone resorption (26,27). p38 is involved in the activation of activator protein (AP)-1 components (28), but the molecular mechanisms of their functions are not well understood. A major component of the transcription factor AP-1 is Fos proto-oncogene (also known as c-fos), which also induces NFATc1, a master regulator of osteoclastogenesis, by binding to its promoter region (29,30). The activity of c-fos and NFATc1 has been reported to be regulated by p38 (26,2932). In addition, the present study demonstrated that MKK6, the kinase upstream of p38 (33), was unaffected by fraxetin administration. It was therefore speculated that fraxetin may prevent the stimulatory effects of RANKL by p38/c-fos/NFATc1 signaling, and subsequently inhibit osteoclastic-specific gene expression (10).

Ultra-high molecular weight polyethylene, Ti, and poly-methyl methacrylate (PMMA) are common particles present in tissues around joint prostheses, which are important for today’s joint replacement surgeons (34,35). Though metal particles are relatively less frequently used, osteolysis induced by Ti particles is typical (36,37). Thus, Ti particle-induced osteolysis could be representative and suitable for studying the effect of fraxetin on osteolysis caused by wear debris (16,17). The current results demonstrated that fraxetin administration prevented Ti particle-induced osteolysis in a mouse osteolysis model and did not result in any obvious side effects, confirming that fraxetin inhibits osteoclast differentiation and function in vivo and suggesting that it may hold potential therapeutic benefits on PO.

The present study has several strengths. It is the first that characterized the suppression effects of fraxetin on osteo-clastogenesis. Chinese herbs are less prone to lead to drug resistance and have fewer side effects compared with synthetic drugs. Therefore, investigating the effects of fraxetin further may be a promising approach to develop treatments for PO. The present study also presents a limitation. PO is a complex process that consists of multiple interactions between bone cells and inflammatory reactions in vivo. However, the present study only explored the mechanism for osteolytic diseases therapy through the inhibition of osteoclast activities.

Collectively, the present findings demonstrated that fraxetin administration improved PO in vitro and in vivo, through the suppression of osteoclast development and function via inhibition of the p38 signaling pathway. Therefore, fraxetin may be a potential agent for the treatment of PO and other osteolytic diseases.

Acknowledgments

Not applicable.

Funding

No funding was received.

Availability of data and materials

The analyzed datasets generated during the study are available from the corresponding author on reasonable request.

Authors’ contributions

DZC designed the study; JCL, ZXW, and ZPM performed the research; JCL and CZ contributed new reagents or analytical tools; JCL, CZ and ZXW analyzed data; JCL wrote the study.

Ethics approval and consent to participate

This study obtained ethics approval by the Institutional Animal Care and Use Committee (IACUC) of Southern Medical University (SMU; Guangzhou, China), and all experiments involving animals were performed according to the guidelines of SMU IACUC.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Teitelbaum SL: Bone resorption by osteoclasts. Science. 289:1504–1508. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Rodan GA and Martin TJ: Therapeutic approaches to bone diseases. Science. 289:1508–1514. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Helfrich MH: Osteoclast diseases and dental abnormalities. Arch Oral Biol. 50:115–122. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Abu-Amer Y, Darwech I and Clohisy JC: Aseptic loosening of total joint replacements: Mechanisms underlying osteolysis and potential therapies. Arthritis Res Ther. 9(Suppl 1): S62007. View Article : Google Scholar : PubMed/NCBI

5 

Anderson JM, Rodriguez A and Chang DT: Foreign body reaction to biomaterials. Semin Immunol. 20:86–100. 2008. View Article : Google Scholar

6 

Long M and Rack HJ: Titanium alloys in total joint replacement-a materials science perspective. Biomaterials. 19:1621–1639. 1998. View Article : Google Scholar : PubMed/NCBI

7 

Holt G, Murnaghan C, Reilly J and Meek RM: The biology of aseptic osteolysis. Clin Orthop Relat Res. 460:240–252. 2007.PubMed/NCBI

8 

Udagawa N, Takahashi N, Akatsu T, Tanaka H, Sasaki T, Nishihara T, Koga T, Martin TJ and Suda T: Origin of osteo-clasts: Mature monocytes and macrophages are capable of differentiating into osteoclasts under a suitable microenvironment prepared by bone marrow-derived stromal cells. Proc Natl Acad Sci USA. 87:7260–7264. 1990. View Article : Google Scholar

9 

Novack DV: Role of NF-kappaB in the skeleton. Cell Res. 21:169–182. 2011. View Article : Google Scholar

10 

Boyle WJ, Simonet WS and Lacey DL: Osteoclast differentiation and activation. Nature. 423:337–342. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Feng X: RANKing intracellular signaling in osteoclasts. IUBMB Life. 57:389–395. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Witaicenis A, Seito LN, da Silveira Chagas A, de Almeida LD Jr, Luchini AC, Rodrigues-Orsi P, Cestari SH and Di Stasi LC: Antioxidant and intestinal anti-inflammatory effects of plant-derived coumarin derivatives. Phytomedicine. 21:240–246. 2014. View Article : Google Scholar

13 

Murali R, Srinivasan S and Ashokkumar N: Antihyperglycemic effect of fraxetin on hepatic key enzymes of carbohydrate metabolism in streptozotocin-induced diabetic rats. Biochimie. 95:1848–1854. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Kuo PL, Huang YT, Chang CH and Chang JK: Fraxetin inhibits the induction of anti-Fas IgM, tumor necrosis factor-alpha and interleukin-1beta-mediated apoptosis by Fas pathway inhibition in human osteoblastic cell line MG-63. Int Immunopharmacol. 6:1167–1175. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Jin S, Park JY, Hong JM, Kim TH, Shin HI, Park EK and Kim SY: Inhibitory effect of (-)-epigallocatechin gallate on titanium particle-induced TNF-alpha release and in vivo osteolysis. Exp Mol Med. 43:411–418. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Liu F, Zhu Z, Mao Y, Liu M, Tang T and Qiu S: Inhibition of titanium particle-induced osteoclastogenesis through inactivation of NFATc1 by VIVIT peptide. Biomaterials. 30:1756–1762. 2009. View Article : Google Scholar : PubMed/NCBI

17 

von Knoch M, Jewison DE, Sibonga JD, Sprecher C, Morrey BF, Loer F, Berry DJ and Scully SP: The effectiveness of polyethylene versus titanium particles in inducing osteolysis in vivo. J Orthop Res. 22:237–243. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Lee SS, Woo CH, Chang JD and Kim JH: Roles of Rac and cytosolic phospholipase A2 in the intracellular signalling in response to titanium particles. Cell Signal. 15:339–345. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Qin A, Cheng TS, Lin Z, Cao L, Chim SM, Pavlos NJ, Xu J, Zheng MH and Dai KR: Prevention of wear particle-induced osteolysis by a novel V-ATPase inhibitor saliphenylhalamide through inhibition of osteoclast bone resorption. PloS One. 7:e341322012. View Article : Google Scholar : PubMed/NCBI

20 

Wedemeyer C, Xu J, Neuerburg C, Landgraeber S, Malyar NM, von Knoch F, Gosheger G, von Knoch M, Löer F and Saxler G: Particle-induced osteolysis in three-dimensional micro-computed tomography. Calcif Tissue Int. 81:394–402. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Cano E, Hazzalin CA and Mahadevan LC: Anisomycin-activated protein kinases p45 and p55 but not mitogen-activated protein kinases ERK-1 and -2 are implicated in the induction of c-fos and c-jun. Mol Cell Biol. 14:7352–7362. 1994. View Article : Google Scholar : PubMed/NCBI

22 

Hazzalin CA, Le Panse R, Cano E and Mahadevan LC: Anisomycin selectively desensitizes signalling components involved in stress kinase activation and fos and jun induction. Mol Cell Biol. 18:1844–1854. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Lin C, Shao Y, Zeng C, Zhao C, Fang H, Wang L, Pan J, Liu L, Qi W, Feng X, et al: Blocking PI3K/AKT signaling inhibits bone sclerosis in subchondral bone and attenuates post-traumatic osteoarthritis. J Cell Physiol. 233:6135–6147. 2018. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Stevenson DA, Schwarz EL, Carey JC, Viskochil DH, Hanson H, Bauer S, Weng HY, Greene T, Reinker K, Swensen J, et al: Bone resorption in syndromes of the Ras/MAPK pathway. Clin Genet. 80:566–573. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Huang H, Chang EJ, Ryu J, Lee ZH, Lee Y and Kim HH: Induction of c-Fos and NFATc1 during RANKL-stimulated osteoclast differentiation is mediated by the p38 signaling pathway. Biochem Biophys Res Commun. 351:99–105. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Zwerina J, Hayer S, Redlich K, Bobacz K, Kollias G, Smolen JS and Schett G: Activation of p38 MAPK is a key step in tumor necrosis factor-mediated inflammatory bone destruction. Arthritis Rheum. 54:463–472. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Chang L and Karin M: Mammalian MAP kinase signalling cascades. Nature. 410:37–40. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Lucas JJ, Yamamoto A, Scearce-Levie K, Saudou F and Hen R: Absence of fenfluramine-induced anorexia and reduced c-Fos induction in the hypothalamus and central amygdaloid complex of serotonin 1B receptor knock-out mice. J Neurosci. 18:5537–5544. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, Saiura A, Isobe M, Yokochi T, Inoue J, et al: Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell. 3:889–901. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Sharma SM, Bronisz A, Hu R, Patel K, Mansky KC, Sif S and Ostrowski MC: MITF and PU.1 recruit p38 MAPK and NFATc1 to target genes during osteoclast differentiation. J Biol Chem. 282:15921–15929. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Matsumoto M, Kogawa M, Wada S, Takayanagi H, Tsujimoto M, Katayama S, Hisatake K and Nogi Y: Essential role of p38 mitogen-activated protein kinase in cathepsin K gene expression during osteoclastogenesis through association of NFATc1 and PU.1. J Biol Chem. 279:45969–45979. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Kyriakis JM and Avruch J: Mammalian MAPK signal transduction pathways activated by stress and inflammation: A 10-year update. Physiol Rev. 92:689–737. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Hirakawa K, Bauer TW, Stulberg BN and Wilde AH: Comparison and quantitation of wear debris of failed total hip and total knee arthroplasty. J Biomed Mater Res. 31:257–263. 1996. View Article : Google Scholar : PubMed/NCBI

35 

Baumann B, Seufert J, Jakob F, Nöth U, Rolf O, Eulert J and Rader CP: Activation of NF-kappaB signalling and TNFalpha-expression in THP-1 macrophages by TiAlV- and polyethylene-wear particles. J Orthop Res. 23:1241–1248. 2005.PubMed/NCBI

36 

Masui T, Sakano S, Hasegawa Y, Warashina H and Ishiguro N: Expression of inflammatory cytokines, RANKL and OPG induced by titanium, cobalt-chromium and polyethylene particles. Biomaterials. 26:1695–1702. 2005. View Article : Google Scholar

37 

Baumann B, Rader CP, Seufert J, Nöth U, Rolf O, Eulert J and Jakob F: Effects of polyethylene and TiAlV wear particles on expression of RANK, RANKL and OPG mRNA. Acta Orthop Scand. 75:295–302. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 42 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liao JC, Wei ZX, Zhao C, Ma ZP and Cai DZ: Inhibition of osteoclastogenesis for periprosthetic osteolysis therapy through the suppression of p38 signaling by fraxetin. Int J Mol Med 42: 1257-1264, 2018
APA
Liao, J., Wei, Z., Zhao, C., Ma, Z., & Cai, D. (2018). Inhibition of osteoclastogenesis for periprosthetic osteolysis therapy through the suppression of p38 signaling by fraxetin. International Journal of Molecular Medicine, 42, 1257-1264. https://doi.org/10.3892/ijmm.2018.3698
MLA
Liao, J., Wei, Z., Zhao, C., Ma, Z., Cai, D."Inhibition of osteoclastogenesis for periprosthetic osteolysis therapy through the suppression of p38 signaling by fraxetin". International Journal of Molecular Medicine 42.3 (2018): 1257-1264.
Chicago
Liao, J., Wei, Z., Zhao, C., Ma, Z., Cai, D."Inhibition of osteoclastogenesis for periprosthetic osteolysis therapy through the suppression of p38 signaling by fraxetin". International Journal of Molecular Medicine 42, no. 3 (2018): 1257-1264. https://doi.org/10.3892/ijmm.2018.3698