Inhibition of RANKL-induced osteoclastogenesis through the suppression of the ERK signaling pathway by astragaloside IV and attenuation of titanium-particle-induced osteolysis

Corrigendum in: /10.3892/ijmm.2018.3617

  • Authors:
    • Mingjun Li
    • Wengang Wang
    • Li Geng
    • Yanru Qin
    • Wenjie Dong
    • Xudong Zhang
    • An Qin
    • Mingzhi Zhang
  • View Affiliations

  • Published online on: August 31, 2015     https://doi.org/10.3892/ijmm.2015.2330
  • Pages: 1335-1344
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Astragaloside IV (AS-IV) is a natural plant extract that enhances osteoblast activity, and therefore, has the potential to treat osteoclast‑related diseases. Such diseases include osteoporosis, periodontal disease, rheumatoid arthritis and aseptic prosthesis loosening. However, data associating the effects of AS‑IV on osteoclasts are limited. The aim of the present study was to assess the effect of AS‑IV on osteoclasts in vitro and in vivo. The in vitro studies demonstrated that AS‑IV exerts potent inhibitory effects on the ligand of the receptor activator of nuclear factor‑κB‑induced osteoclastogenesis and revealed the mechanism of action of AS‑IV, which inhibited osteoclastogenesis by suppression of the extracellular signal‑regulated kinase signaling pathway. The in vivo studies proved that AS‑IV attenuated titanium particle‑induced osteolysis in a mouse calvarial model. Collectively, the findings of the study suggest that AS‑IV is a potential natural agent for the treatment of osteoclast-related diseases.

Introduction

In vivo, the skeleton is constantly being remodeled via a process involving the breakdown (resorption) and build-up (synthesis) of bone, determined by a delicate balance between osteoblast and osteoclast activities (1). As osteoclasts have key roles in the regulation of bone mass and quality, the majority of adult skeletal diseases are due to excess osteoclast activity, resulting in osteopenia (2,3). Such diseases include osteoporosis, periodontal disease, rheumatoid arthritis and aseptic prosthesis loosening (4,5). For individuals with osteoporosis, a condition characterized by low bone mass and skeletal fragility, low trauma bone fractures represent life-threatening events, particularly when they affect the vertebrae, proximal femur (hip), distal forearm or proximal humerus (6).

Osteoclasts are large, multinucleated cells that arise from the hematopoietic stem cell monocyte/macrophage lineage (7). Osteoclast activation was initiated by activation of the receptor activator of the nuclear factor-κB (RANK) and RANK ligand (RANKL) signaling pathways (8,9). RANK and RANKL belong to the tumor necrosis factor (TNF) receptor superfamily (10,11). The binding of RANKL to RANK recruits the adapter protein, TNF receptor-associated factor 6 (TRAF6), to the plasma membrane. RANK, RANKL and TRAF6 are essential for osteoclastogenesis, as mice lacking these molecules show profound bone resorption defects (12). The RANK/TRAF6 complex activates several pathways, including NF-κB signaling and the mitogen-activated protein kinase (MAPK) pathways involving extracellular signal-related kinase (ERK), which induce expression of nuclear factor of activated T cells c1 (NFATc1), considered one of the master transcription factors controlling osteoclastogenesis (1315). Therefore, ERK and NFATc1, which are closely regulated by MAPK activity, are essential for the differentiation, survival and activation of osteoclasts (1618).

Astragaloside IV (AS-IV) (Fig. 1) is a saponin purified from Astragalus membranaceus Bge., one of the most widely used Chinese herbs (19). AS-IV has been reported to have a wide range of treatment effects, with no toxicity (20). Pharmacological activities attributed to AS-IV include cardioprotective (21), anti-inflammatory (22), antioxidant (23), anti-asthmatic (24) and anticancer (25) effects. Some of these pharmacological activities resulted from AS-IV-mediated inhibition of ERK (2628) and NF-κB (26,2931) signaling pathways. Furthermore, AS-IV has been reported to affect osteogenesis (32), and have anti-arthritis activity (33). All these findings indicated that AS-IV may have a negative effect on osteoclastogenesis and may therefore have a significant potential for the treatment of osteoclast-related diseases, including osteoporosis. However, to the best of our knowledge, there is little published information regarding this issue.

Therefore, the present study aimed to i) investigate the potential therapeutic benefits of AS-IV on osteoclast-related osteolytic bone diseases, ii) understand the underlying mechanisms mediating the effects of AS-IV on osteoclast formation and function, and iii) further elucidate the potential molecular mechanisms of AS-IV in osteoclasts.

Materials and methods

Media, reagents and cells

Fetal bovine serum and α-modification of Eagle's medium (α-MEM) were obtained from Gibco-BRL (Sydney, Australia). The cell counting kit-8 (CCK-8) was purchased from Dojindo (Kumamoto, Japan). Soluble human recombinant macrophage-colony stimulating factor (M-CSF) and bacteria-derived recombinant mouse RANKL were supplied by R&D Systems (Minneapolis, MN, USA). AS-IV (purity >98%) was purchased from Sigma-Aldrich (St. Louis, MO, USA) and dissolved in dimethyl sulfoxide prior to dilution to the appropriate concentrations in culture medium (34). Western blot-specific antibodies were obtained from Cell Signaling Technology (Cambridge, MA, USA). RAW264.7 cells were purchased from the American Type Culture Collection (Rockville, MD, USA).

Cytotoxicity assay

The cytotoxic effects of AS-IV were determined using a CCK-8 assay. Bone marrow macrophages (BMMs) of C57/BL6 mice were seeded in 96-well plates at a density of 8×103 cells/well, and cultured for 24 h. Cells were subsequently treated with different concentrations of AS-IV for 24, 48, 72 and 96 h. CCK-8 buffer (10 µl) was added to each well, and the cells were incubated at 37°C for an additional 2 h. The absorbance was subsequently measured at a wavelength of 450 nm (650 nm reference). The half-maximal inhibitory concentration (IC50) value was calculated.

BMM isolation and osteoclast culture

For primary cell culture, BMMs were obtained from the femurs and tibias of 6-week-old C57/BL6 mice. Cells were cultured in a T75 flask for 24 h. Floating cells were removed, and the adherent cells were cultured for another 3 days. The BMMs were subsequently seeded in a 96-well plate at a density of 8×103 cells/well in complete α-MEM supplemented with 30 ng/ml M-CSF, 50 ng/ml RANKL and different concentrations of AS-IV (0, 25, 50 or 100 µM). Culture media were replaced every 2 days until mature osteoclasts had formed. Osteoclasts were identified by positive staining for tartrate-resistant acid phosphatase (TRAP). TRAP-positive cells with >3 nuclei were counted under a microscope.

F-actin ring immunofluorescence

For F-actin ring immunofluorescent staining, osteoclasts were fixed with 4% paraformaldehyde for 15 min at room temperature and permeabilized for 5 min with 0.1% v/v Triton X-100. Cells were incubated with rhodamine-conjugated phalloidin (1:100; Invitrogen, Carlsbad, CA, USA) for 1 h at room temperature. Cells were subsequently incubated with Hoechst 3342 dye (1:5,000) for visualization of the nuclei, washed with phosphate-buffered saline (PBS), and mounted with ProLong Gold anti-fade mounting medium (both from Invitrogen). Fluorescence detection was performed on Nikon A1Si spectral detector confocal system equipped with 20X (dry) lenses (Nikon, Tokyo, Japan). Images were collected using NIS-C elements software and analyzed using ImageJ Software (National Institutes of Health, Bethesda, MD, USA).

Bone resorption pit assay

BMMs were seeded on bone slices in 96-well plates at a density of 8×103 cells/well and stimulated (in triplicate) with M-CSF (30 ng/ml), RANKL (50 ng/ml) and AS-IV (0, 25, 50 or 100 µM). Bone slices were imaged using field-emission scanning electron microscopy (FESEM, Hitachi S-4800, CamScan; Hitachi, Tokyo, Japan) with a magnification of 10 kV.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay

RT-qPCR was used to measure specific gene expression. BMMs were plated in 6-well plates at a density of 1×105 cells/well and cultured in complete α-MEM supplemented with 30 ng/ml M-CSF and 50 ng/ml RANKL. For the concentration gradient assay, cells were incubated with AS-IV (0, 25 or 50 µM) for 5–7 days until mature osteoclasts formed. For the time gradient assay, cells were incubated with 50 µM AS-IV for 0, 1, 3 or 5 days. Total RNA was extracted using the Qiagen RNeasy Mini kit (Qiagen, Valencia, CA, USA) and cDNA was synthesized from 1 µg of total RNA using reverse transcriptase (Takara Biotechnology, Otsu, Japan). qPCR was performed using the SYBR Premix Ex Tag kit (Takara Biotechnology) and an ABI 7500 Sequencing Detection system (Applied Biosystems, Foster City, CA, USA) with 40 cycles of denaturation at 95°C for 5 sec and amplification at 60°C for 24 sec. GAPDH was used as the reference gene, and all the reactions were run in triplicate. The mouse primer sequences for cathepsin K (CtsK), TRAP, dendritic cell-specific trans membrane protein (DC-STAMP), V-ATPase d2, c-fos, NFATc1 and GAPDH were as follows: CtsK forward, 5′-CTTCCAATACGTGCA GCAGA-3′ and reverse, 5′-TCTTCAGGGCTTTCTCGTTC-3′; TRAP forward, 5′-CTGGAGTGCACGATGCCAGCGACA-3′ and reverse, 5′-TCCGTGCTCGGCGATGGACCAGA-3′; DC-STAMP forward, 5′-AAAACCCTTGGGCTGTTCTT-3′ and reverse, 5′-AATCATGGACGACTCCTTGG-3′; V-ATPase d2 forward, 5′-AAGCCTTTGTTTGACGCTGT-3′ and reverse, 5′-TTCGATGCCTCTGTGAGATG-3′; c-fos forward, 5′-CCA GTCAAGAGCATCAGCAA-3′ and reverse, 5′-AAGTAG TGCAGCCCGGAGTA-3′; NFATc1 forward, 5′-CCGTTG CTTCCAGAAAATAACA-3′ and reverse, 5′-TGTGGGATG TGAACTCGGAA-3′; and GAPDH forward, 5′-ACCCAG AAGACTGTGGATGG-3′ and reverse, 5′-CACATTGGG GGTAGGAACAC-3′.

NF-κB luciferase reporter assay

RAW264.7 cells were stably transfected with a p-NF-κB-TA-Luc luciferase reporter construct. Briefly, cells were plated in a 24-well plate at a density of 1×105 cells/well. The cells were treated 24 h later with different AS-IV concentrations (0, 12.5, 25, 50, 100 or 200 µM) for 1 h, prior to incubation with 50 ng/ml RANKL for a further 8 h. Cells were subsequently lysed and luciferase activity was measured using the Promega Luciferase Assay system (Promega, Madison, WI, USA).

Western blotting

RAW264.7 cells were seeded in 6-well plates at a density of 5×105 cells/well. After pretreatment with or without AS-IV (200 µM) for 4 h, the cells were stimulated with RANKL for 0, 5, 10, 20, 30 or 60 min. The cells were subsequently washed twice in PBS and lysed in radioimmuno-precipitation assay lysis buffer. The lysates were centrifuged at 12,000 x g for 15 min and supernatants were collected.

Protein concentrations were determined using the bicinchoninic acid assay. Protein from each lysate (20 µg) was resolved using 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis gels, and transferred to polyvinylidene difluoride membranes (Millipore, Bedford, MA, USA). Membranes were subsequently blocked with 5% skimmed milk in TBS-Tween-20 for 1 h, and incubated with primary antibodies overnight at 4°C. The membranes were incubated with horseradish peroxidase-conjugated secondary antibodies (1:5,000) for 1 h. Antibody reactivity was detected using an Odyssey infrared imaging system (LI-COR Biosciences, Lincoln, NE, USA).

Titanium particle-induced calvarial osteolysis model

An in vivo wear particle-induced osteolysis model was generated as previously reported (35). All the animal care and experimental procedures complied with Directive 2010/63/EU revising Directive 86/609/EEC approved by Animal Care and Use Committee of Zhengzhou University (approved on Feb 19th 2014, the approval number is 001381). Twenty healthy 8-week-old C57BL/J6 mice were randomly assigned to four groups: Sham PBS control (sham), Ti particles with PBS (vehicle), and Ti particles with low (10 mg/kg/day) and high (25 mg/kg/day) concentrations of AS-IV (low and high, respectively). AS-IV was used by intraperitoneal injection every other day for 14 days. At the end of the experiment, the mice were sacrificed, and the calvaria were excised and fixed in 4% paraformaldehyde for micro-computed tomography (micro-CT) analysis.

Micro-CT scanning

A high-resolution micro-CT scanner (Skyscan 1176; Skyscan; Aartselaar, Belgium) was used with the following settings: X-ray voltage, 50 kV; electric current, 500 mA; and rotation step, 0.7°. Following reconstruction, a square region of interest (ROI) around the midline suture was chosen for further qualitative and quantitative analyses. The bone volume against tissue volume (BV/TV), number of porosities and percentage of total porosity were determined for each sample as described previously (36).

Statistical analysis

The data Are expressed as mean ± standard deviation. The results were analyzed using the analysis of variance and post hoc tests with the SPSS 13.0 software (SPSS Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference between groups.

Results

AS-IV cytotoxicity

CCK-8 cell viability assays were performed to examine the potential AS-IV cytotoxicity. The results showed that AS-IV cytotoxicity was concentration- and time-dependent (Fig. 2). These assays indicated that the maximum concentration used in the subsequent studies (200 µM) showed no cytotoxic effects in BMMs, even after 96-h exposure. Based on these data, the AS-IV concentrations used in subsequent experiments were considered non-cytotoxic.

Effect of AS-IV on osteoclastogenesis

As osteoclasts have key roles in osteoclast-related diseases (2), the effect of AS-IV on osteoclastogenesis was investigated. BMMs were exposed to AS-IV (0, 25, 50 and 100 µM) during osteoclast formation. The control group formed numerous TRAP-positive multinucleated osteoclasts (Fig. 3A). Osteoclast formation was inhibited by AS-IV (Fig. 3A–C). Furthermore, comparison of the control and 100 µM AS-IV cells after 3, 5 and 7 days produced similar results (Fig. 3D–F).

F-actin ring formation

To further examine the effects of AS-IV on osteoclastogenesis, RANKL-induced osteoclast F-actin ring formation was studied, which is the most well-known characteristic of mature osteoclasts and a prerequisite for osteoclast bone resorption during osteoclastogenesis (37). As expected, confocal microscopy revealed F-actin ring formation and characteristic podosomal condensation in control osteoclasts. However, the size and number of F-actin rings significantly decreased in cells incubated with AS-IV (Fig. 4A), suggesting that AS-IV suppressed F-actin ring formation.

Osteoclast bone resorption

As the formation of a well-polarized F-actin ring is an essential prerequisite for efficient bone resorption by osteoclasts, we inferred that osteoclast bone resorption would also be inhibited by AS-IV. Numerous bone resorption pits were observed on the surface of control bone slices (Fig. 5). In bone slices exposed to AS-IV, the resorption area was decreased. These findings demonstrated that AS-IV impaired osteoclast bone resorption in vitro.

RANKL-induced gene expression

The expression levels of several specific genes are upregulated during osteoclast differentiation. Thus, RT-qPCR was used to examine and quantify the RANKL-induced mRNA expression of osteoclast-related genes (including NFATc1, TRAP, V-ATPase d2, CtsK, DC-STAMP and c-fos). The results showed that the expression of these genes was inhibited by AS-IV in a dose- and time-dependent manner (Fig. 6).

RANKL-induced ERK and NFATc1 expression signaling

RANKL-induced activation of ERK is essential for osteoclast differentiation and function (38,39). The effects of AS-IV on RANKL-induced signaling pathways were therefore investigated. ERK phosphorylation increased within 5–30 min of stimulation with RANKL in the control group. However, ERK phosphorylation was significantly reduced by exposure to AS-IV (Fig. 7A). Quantitative analysis confirmed these results (Fig. 7C). These results suggested that AS-IV inhibited phosphorylation of ERK during osteoclast differentiation.

NFATc1 is an important master regulator of osteoclastogenesis and osteoclast function (40). Therefore, the effects of AS-IV on RANKL-induced NFATc1 expression were investigated. The data presented in Fig. 6A and B indicated that NFATc1 transcriptional activity increased when the cells were stimulated by RANKL. AS-IV inhibited this activity in a dose- and time-dependent manner. To confirm this effect of AS-IV on NFATc1 expression, the NFATc1 protein level was examined using western blot analysis. NFATc1 protein levels increased when cells were exposed to RANKL, and AS-IV attenuated this increase (Fig. 7D and E), suggesting that AS-IV suppressed RANKL-induced NFATc1 expression.

RANKL-induced NF-κB signaling

RANKL-induced NF-κB activation is also a dominant mediator of osteoclast differentiation, resorption function and survival (4143). Western blot analysis and luciferase assays were used to investigate the NF-κB signaling pathway. Similar levels of IκBα phosphorylation and degradation were observed in control and AS-IV groups (Fig. 7A). This observation was supported by luciferase reporter gene assays (Fig. 7B). These data indicated that AS-IV inhibited osteoclastogenesis without affecting the NF-κB signaling pathway.

Titanium particle-induced osteolysis

To explore the effects of AS-IV on pathological osteolysis, a Ti particle-induced mouse calvarial osteolysis model was used. The degree of particle-induced osteolysis was assessed using high-resolution micro-CT. Compared with the sham group (no Ti particles), the vehicle group (administration of Ti particles in PBS) showed significant calvarial osteolysis. When AS-IV (10 mg/kg, low; 25 mg/kg, high) was administered with the Ti particles, this osteolytic bone loss reduced (Fig. 8A). Quantitative analysis of bone parameters further demonstrated that the high or low concentrations of AS-IV significantly increased the BV/TV (Fig. 8B), and decreased the number of porosities and the percentage of total porosity in the ROI in the calvaria (Fig. 8C and D).

Discussion

Diseases associated with osteoclasts include osteoporosis, rheumatoid arthritis, multiple myeloma, periprosthetic osteolysis and metastatic cancers (2,4446). During the last two decades, numerous advances have been made in the treatment of osteoclast-related diseases; however, treatment options are not optimal thus far.

The bisphosphonates were the first drugs approved specifically for the treatment and prevention of osteoclast-related diseases. However, they often cause gastrointestinal toxicity, including dyspepsia, abdominal pain, gastritis and esophagitis. Other serious adverse effects include osteonecrosis of the jaw, femur fractures and atrial fibrillation (47). Estrogens have also been used for the treatment of osteoporosis, but can have serious adverse effects, such as breast cancer, endometrial cancer and thromboembolism (48). The first effective bone anabolic agent teriparatide [parathyroid hormone (PTH) 1–34] has also been used clinically. However, high cost and the requirement for daily subcutaneous injections are major limitations to its use (49). Denosumab is a human monoclonal antibody that inhibits RANKL and, consequently, osteoclastogenesis. However, cellulitis was more frequent in patients taking denosumab compared with the placebo (50) and patients also had a high risk of fractures (51). Strontium ranelate is also used clinically; however, it has the common side effects of nausea, diarrhea, and mild and transient elevation in creatine kinase, and is contraindicated in patients with a high risk of thromboembolic events. In addition, a few cases of hypersensitivity have been reported (52). Due to the limitations of present therapies, attempts to develop improved treatment options are being pursued.

Previous studies indicated that AS-IV had positive effects on osteogenesis (32) and arthritis (33), and to the best of our knowledge, the present study demonstrated for the first time that it significantly inhibited osteoclast differentiation and formation, impaired F-actin ring formation, and significantly decreased the number and area of bone resorption pits in vitro. In the in vivo studies, three-dimensional reconstruction of micro-CT images from mouse calvarias showed that AS-IV treatment markedly suppressed Ti particle-induced osteolysis in a dose-dependent manner.

Activation of RANK by its ligand leads to the expression of osteoclast-specific genes during differentiation, and the activation of resorption by mature osteoclasts (11). RANK signaling is mediated by cytoplasmic factors that activate downstream pathways controlling these various functions. At least five distinct protein kinase-mediated signaling cascades are induced during osteoclastogenesis and activation; inhibitor of NF-κB kinase (IKK), c-Jun N-terminal kinase, p38, ERK and Src pathways (11). ERK is essential for osteoclast differentiation, survival and activation (16,17,53,54). The inhibition of ERK has been proven to possess a therapeutic potential for osteoclast-related diseases (39). Activated ERK stimulates transcription factors, such as NFATc1 (15), which is a master regulator of osteoclast differentiation (40,55,56). Overexpression of NFATc1 accelerates RANKL-induced osteoclast differentiation and can also increase osteoclast formation independently of RANKL. Additionally, NFATc1-deficient embryonic stem cells failed to differentiate into osteoclasts, even in the presence of RANKL (40). Using western blot analysis, AS-IV was revealed to inhibit RANKL-induced ERK signaling. NF-κB was unaffected and this result was confirmed using an NF-κB luciferase reporter assay. AS-IV also inhibited NFATc1 mRNA and protein expression. NFATc1 regulates the expression of a range of genes associated with osteoclast differentiation and function. In the present study, the expression of NFATc1-regulated genes (TRAP, CtsK and V-ATPase d2) was downregulated by AS-IV, suggesting that AS-IV not only affected the expression of NFATc1, but also affected the expression of its downstream genes.

In conclusion, the present study demonstrated that AS-IV had inhibitory effects on osteoclastogenesis and osteoclast function in vitro and in vivo. Additionally, these inhibitory effects appeared to operate via blockade of the ERK and NFATc1 pathways. Taken together, the data strongly suggested that AS-IV may be developed as a potential agent for the treatment of osteoclast-related diseases, including osteoporosis.

Acknowledgments

The present study was supported by the Department of Oncology, The First Affiliated Hospital of Zhengzhou University.

Abbreviations:

AS-IV

astragaloside IV

BMMs

bone marrow macrophages

ERK

extracellular signal-regulated kinase

GAPDH

glyceraldehyde-3-phosphate dehydrogenase

MAPK

mitogen-activated protein kinase

M-CSF

macrophage colony-stimulating factor

NFATc1

nuclear factor of activated T cells c1

RANK

receptor activator of the nuclear factor-κB

RANKL

receptor activator of the nuclear factor-κB ligand

TNF

tumor necrosis factor

TRAF6

tumor necrosis factor receptor-associated factor 6

References

1 

Kim HS, Suh KS, Sul D, Kim BJ, Lee SK and Jung WW: The inhibitory effect and the molecular mechanism of glabridin on RANKL-induced osteoclastogenesis in RAW264.7 cells. Int J Mol Med. 29:169–177. 2012.

2 

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

3 

Kim TH, Kim HJ, Lee SH and Kim SY: Potent inhibitory effect of Foeniculum vulgare Miller extract on osteoclast differentiation and ovariectomy-induced bone loss. Int J Mol Med. 29:1053–1059. 2012.PubMed/NCBI

4 

Guo H, Zhang J, Hao S and Jin Q: Adenovirus-mediated small interfering RNA targeting tumor necrosis factor-α inhibits titanium particle-induced osteoclastogenesis and bone resorption. Int J Mol Med. 32:296–306. 2013.PubMed/NCBI

5 

Hou GQ, Guo C, Song GH, Fang N, Fan WJ, Chen XD, Yuan L and Wang ZQ: Lipopolysaccharide (LPS) promotes osteoclast differentiation and activation by enhancing the MAPK pathway and COX-2 expression in RAW264.7 cells. Int J Mol Med. 32:503–510. 2013.PubMed/NCBI

6 

Kanis JA: Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: Synopsis of a WHO report. Osteoporos Int. 4:368–381. 1994. View Article : Google Scholar : PubMed/NCBI

7 

Udagawa N, Takahashi N, Akatsu T, Tanaka H, Sasaki T, Nishihara T, Koga T, Martin TJ and Suda T: Origin of osteoclasts: 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 : PubMed/NCBI

8 

Purdue PE, Koulouvaris P, Potter HG, Nestor BJ and Sculco TP: The cellular and molecular biology of periprosthetic osteolysis. Clin Orthop Relat Res. 454:251–261. 2007. View Article : Google Scholar

9 

Ren W, Wu B, Peng X, Hua J, Hao HN and Wooley PH: Implant wear induces inflammation, but not osteoclastic bone resorption, in RANK(−/−) mice. J Orthop Res. 24:1575–1586. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Raggatt LJ and Partridge NC: Cellular and molecular mechanisms of bone remodeling. J Biol Chem. 285:25103–25108. 2010. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Asagiri M and Takayanagi H: The molecular understanding of osteoclast differentiation. Bone. 40:251–264. 2007. View Article : Google Scholar

13 

Strait K, Li Y, Dillehay DL and Weitzmann MN: Suppression of NF-kappaB activation blocks osteoclastic bone resorption during estrogen deficiency. Int J Mol Med. 21:521–525. 2008.PubMed/NCBI

14 

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

15 

Meissner JD, Freund R, Krone D, Umeda PK, Chang KC, Gros G and Scheibe RJ: Extracellular signal-regulated kinase 1/2-mediated phosphorylation of p300 enhances myosin heavy chain I/beta gene expression via acetylation of nuclear factor of activated T cells c1. Nucleic Acids Res. 39:5907–5925. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Grigoriadis AE, Wang ZQ, Cecchini MG, Hofstetter W, Felix R, Fleisch HA and Wagner EF: c-Fos: A key regulator of osteoclast-macrophage lineage determination and bone remodeling. Science. 266:443–448. 1994. View Article : Google Scholar : PubMed/NCBI

17 

Mansky KC, Sankar U, Han J and Ostrowski MC: Microphthalmia transcription factor is a target of the p38 MAPK pathway in response to receptor activator of NF-kappa B ligand signaling. J Biol Chem. 277:11077–11083. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Kim YW, Baek SH, Lee SH, Kim TH and Kim SY: Fucoidan, a sulfated polysaccharide, inhibits osteoclast differentiation and function by modulating RANKL signaling. Int J Mol Sci. 15:18840–18855. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Xuying W, Jiangbo Z, Yuping Z, Xili M, Yiwen Z, Tianbao Z and Weidong Z: Effect of astragaloside IV on the general and peripartum reproductive toxicity in Sprague-Dawley rats. Int J Toxicol. 29:505–516. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Ren S, Zhang H, Mu Y, Sun M and Liu P: Pharmacological effects of astragaloside IV: A literature review. J Tradit Chin Med. 33:413–416. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Jia Y, Zuo D, Li Z, Liu H, Dai Z, Cai J, Pang L and Wu Y: Astragaloside IV inhibits doxorubicin-induced cardiomyocyte apoptosis mediated by mitochondrial apoptotic pathway via activating the PI3K/Akt pathway. Chem Pharm Bull (Tokyo). 62:45–53. 2014. View Article : Google Scholar

22 

Tan S, Wang G, Guo Y, Gui D and Wang N: Preventive effects of a natural anti-inflammatory agent, astragaloside IV, on ischemic acute kidney injury in rats. Evid Based Complement Alternat Med. 2013:2840252013.PubMed/NCBI

23 

Hu JY, Han J, Chu ZG, Song HP, Zhang DX, Zhang Q and Huang YS: Astragaloside IV attenuates hypoxia-induced cardiomyocyte damage in rats by upregulating superoxide dismutase-1 levels. Clin Exp Pharmacol Physiol. 36:351–357. 2009. View Article : Google Scholar

24 

Huang X, Tang L, Wang F and Song G: Astragaloside IV attenuates allergic inflammation by regulation Th1/Th2 cytokine and enhancement CD4(+)CD25(+)Foxp3 T cells in ovalbumin-induced asthma. Immunobiology. 219:565–571. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Qi H, Wei L, Han Y, Zhang Q, Lau AS and Rong J: Proteomic characterization of the cellular response to chemopreventive triterpenoid astragaloside IV in human hepatocellular carcinoma cell line HepG2. Int J Oncol. 36:725–735. 2010.PubMed/NCBI

26 

He CL, Yi PF, Fan QJ, Shen HQ, Jiang XL, Qin QQ, Song Z, Zhang C, Wu SC, Wei XB, et al: Xiang-Qi-Tang and its active components exhibit anti-inflammatory and anticoagulant properties by inhibiting MAPK and NF-κB signaling pathways in LPS-treated rat cardiac microvascular endothelial cells. Immunopharmacol Immunotoxicol. 35:215–224. 2013. View Article : Google Scholar

27 

Wang Q, Shao X, Xu W, Qi C, Gu L, Ni Z and Mou S: Astragalosides IV inhibits high glucose-induced cell apoptosis through HGF activation in cultured human tubular epithelial cells. Ren Fail. 36:400–406. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Zheng R, Deng Y, Chen Y, Fan J, Zhang M, Zhong Y, Zhu R and Wang L: Astragaloside IV attenuates complement membranous attack complex induced podocyte injury through the MAPK pathway. Phytother Res. 26:892–898. 2012. View Article : Google Scholar

29 

Gui D, Huang J, Guo Y, Chen J, Chen Y, Xiao W, Liu X and Wang N: Astragaloside IV ameliorates renal injury in streptozotocin-induced diabetic rats through inhibiting NF-κB-mediated inflammatory genes expression. Cytokine. 61:970–977. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Li M, Yu L, She T, Gan Y, Liu F, Hu Z, Chen Y, Li S and Xia H: Astragaloside IV attenuates Toll-like receptor 4 expression via NF-κB pathway under high glucose condition in mesenchymal stem cells. Eur J Pharmacol. 696:203–209. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Sun L, Li W, Li W, Xiong L, Li G and Ma R: Astragaloside IV prevents damage to human mesangial cells through the inhibition of the NADPH oxidase/ROS/Akt/NF-κB pathway under high glucose conditions. Int J Mol Med. 34:167–176. 2014.PubMed/NCBI

32 

Bian Q, Huang JH, Liang QQ, Shu B, Hou W, Xu H, Zhao YJ, Lu S, Shi Q and Wang YJ: The osteogenetic effect of astragaloside IV with centrifugating pressure on the OCT-1 cells. Pharmazie. 66:63–68. 2011.PubMed/NCBI

33 

Wang B and Chen MZ: Astragaloside IV possesses antiarthritic effect by preventing interleukin 1β-induced joint inflammation and cartilage damage. Arch Pharm Res. 37:793–802. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Xie J, Wang H, Song T, Wang Z, Li F, Ma J, Chen J, Nan Y, Yi H and Wang W: Tanshinone IIA and astragaloside IV promote the migration of mesenchymal stem cells by up-regulation of CXCR4. Protoplasma. 250:521–530. 2013. View Article : Google Scholar

35 

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

36 

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

37 

Qin A, Cheng TS, Pavlos NJ, Lin Z, Dai KR and Zheng MH: V-ATPases in osteoclasts: Structure, function and potential inhibitors of bone resorption. Int J Biochem Cell Biol. 44:1422–1435. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Monje P, Hernández-Losa J, Lyons RJ, Castellone MD and Gutkind JS: Regulation of the transcriptional activity of c-Fos by ERK. A novel role for the prolyl isomerase PIN1. J Biol Chem. 280:35081–35084. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Seo SW, Lee D, Minematsu H, Kim AD, Shin M, Cho SK, Kim DW, Yang J and Lee FY: Targeting extracellular signal-regulated kinase (ERK) signaling has therapeutic implications for inflammatory osteolysis. Bone. 46:695–702. 2010. View Article : Google Scholar :

40 

Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, Saiura A, Isobe M, Yokochi T and Inoue J: 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

41 

Franzoso G, Carlson L, Xing L, Poljak L, Shores EW, Brown KD, Leonardi A, Tran T, Boyce BF and Siebenlist U: Requirement for NF-kappaB in osteoclast and B-cell development. Genes Dev. 11:3482–3496. 1997. View Article : Google Scholar

42 

Iotsova V, Caamaño J, Loy J, Yang Y, Lewin A and Bravo R: Osteopetrosis in mice lacking NF-kappaB1 and NF-kappaB2. Nat Med. 3:1285–1289. 1997. View Article : Google Scholar : PubMed/NCBI

43 

Lee SH, Kim JK and Jang HD: Genistein inhibits osteoclastic differentiation of RAW 264.7 cells via regulation of ROS production and scavenging. Int J Mol Sci. 15:10605–10621. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Inacio MC, Ake CF, Paxton EW, Khatod M, Wang C, Gross TP, Kaczmarek RG, Marinac-Dabic D and Sedrakyan A: Sex and risk of hip implant failure: Assessing total hip arthroplasty outcomes in the United States. JAMA Intern Med. 173:435–441. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Rizzoli R: A new treatment for post-menopausal osteoporosis: Strontium ranelate. J Endocrinol Invest. 28(Suppl): 50–57. 2005.PubMed/NCBI

46 

Weitzmann MN and Pacifici R: Estrogen regulation of immune cell bone interactions. Ann NY Acad Sci. 1068:256–274. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Marx RE: The deception and fallacies of sponsored randomized prospective double-blinded clinical trials: The bisphosphonate research example. Int J Oral Maxillofac Implants. 29:e37–e44. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Maeda SS and Lazaretti-Castro M: An overview on the treatment of postmenopausal osteoporosis. Arq Bras Endocrinol Metabol. 58:162–171. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Nakamura T, Sugimoto T, Nakano T, Kishimoto H, Ito M, Fukunaga M, Hagino H, Sone T, Yoshikawa H, Nishizawa Y, et al: Randomized Teriparatide [human parathyroid hormone (PTH) 1-34] Once-Weekly Efficacy Research (TOWER) trial for examining the reduction in new vertebral fractures in subjects with primary osteoporosis and high fracture risk. J Clin Endocrinol Metab. 97:3097–3106. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, et al: FREEDOM Trial: Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 361:756–765. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Hiligsmann M, Boonen A, Dirksen CD, Ben Sedrine W and Reginster JY: Cost-effectiveness of denosumab in the treatment of postmenopausal osteoporotic women. Expert Rev Pharmacoecon Outcomes Res. 13:19–28. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Meunier PJ, Roux C, Ortolani S, Diaz-Curiel M, Compston J, Marquis P, Cormier C, Isaia G, Badurski J, Wark JD, et al: Effects of long-term strontium ranelate treatment on vertebral fracture risk in postmenopausal women with osteoporosis. Osteoporos Int. 20:1663–1673. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Gingery A, Bradley E, Shaw A and Oursler MJ: Phosphatidylinositol 3-kinase coordinately activates the MEK/ERK and AKT/NFkappaB pathways to maintain osteoclast survival. J Cell Biochem. 89:165–179. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Liu X, Qu X, Wu C, Zhai Z, Tian B, Li H, Ouyang Z, Xu X, Wang W, Fan Q, et al: The effect of enoxacin on osteoclastogenesis and reduction of titanium particle-induced osteolysis via suppression of JNK signaling pathway. Biomaterials. 35:5721–5730. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Zhai Z, Qu X, Li H, Yang K, Wan P, Tan L, Ouyang Z, Liu X, Tian B, Xiao F, et al: The effect of metallic magnesium degradation products on osteoclast-induced osteolysis and attenuation of NF-κB and NFATc1 signaling. Biomaterials. 35:6299–6310. 2014. View Article : Google Scholar : PubMed/NCBI

56 

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

Related Articles

Journal Cover

November-2015
Volume 36 Issue 5

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
Li M, Wang W, Geng L, Qin Y, Dong W, Zhang X, Qin A and Zhang M: Inhibition of RANKL-induced osteoclastogenesis through the suppression of the ERK signaling pathway by astragaloside IV and attenuation of titanium-particle-induced osteolysis Corrigendum in /10.3892/ijmm.2018.3617. Int J Mol Med 36: 1335-1344, 2015
APA
Li, M., Wang, W., Geng, L., Qin, Y., Dong, W., Zhang, X. ... Zhang, M. (2015). Inhibition of RANKL-induced osteoclastogenesis through the suppression of the ERK signaling pathway by astragaloside IV and attenuation of titanium-particle-induced osteolysis Corrigendum in /10.3892/ijmm.2018.3617. International Journal of Molecular Medicine, 36, 1335-1344. https://doi.org/10.3892/ijmm.2015.2330
MLA
Li, M., Wang, W., Geng, L., Qin, Y., Dong, W., Zhang, X., Qin, A., Zhang, M."Inhibition of RANKL-induced osteoclastogenesis through the suppression of the ERK signaling pathway by astragaloside IV and attenuation of titanium-particle-induced osteolysis Corrigendum in /10.3892/ijmm.2018.3617". International Journal of Molecular Medicine 36.5 (2015): 1335-1344.
Chicago
Li, M., Wang, W., Geng, L., Qin, Y., Dong, W., Zhang, X., Qin, A., Zhang, M."Inhibition of RANKL-induced osteoclastogenesis through the suppression of the ERK signaling pathway by astragaloside IV and attenuation of titanium-particle-induced osteolysis Corrigendum in /10.3892/ijmm.2018.3617". International Journal of Molecular Medicine 36, no. 5 (2015): 1335-1344. https://doi.org/10.3892/ijmm.2015.2330