Lipopolysaccharide (LPS) promotes osteoclast differentiation and activation by enhancing the MAPK pathway and COX-2 expression in RAW264.7 cells

  • Authors:
    • Guo-Qing Hou
    • Chun Guo
    • Guo-Hua Song
    • Na Fang
    • Wen-Juan Fan
    • Xu-Dong Chen
    • Lei Yuan
    • Zhen-Quan Wang
  • View Affiliations

  • Published online on: June 5, 2013     https://doi.org/10.3892/ijmm.2013.1406
  • Pages: 503-510
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bone degradation is a serious complication of chronic inflammatory diseases such as septic arthritis, osteomyelitis and infected orthopedic implant failure. At present, effective therapeutic treatments for lipopolysaccharide (LPS)‑induced bone destruction are limited to antibiotics and surgical repair in chronic inflammatory diseases. The present study aimed to evaluate the mechanism of LPS on osteoclast differentiation and activation. RAW264.7 cells were non-induced, or induced by the receptor activator of nuclear factor-κB (RANK) ligand (RANKL) and macrophage-colony stimulating factor (M-CSF), and then treated with LPS. Following treatment, the number of osteoclasts and cell viability were measured. The expression of osteoclast-related genes including tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinase-9 (MMP-9), cathepsin K (CK), carbonic anhydrase II (CAII) and cyclooxygenase-2 (COX-2) was determined by RT-PCR. Protein levels of RANK, tumor necrosis factor receptor‑associated factor 6 (TRAF6), COX-2 and mitogen-activated protein kinases (MAPK) were measured using western blotting assays. LPS promoted osteoclast differentiation of RAW264.7 cells and differentiated osteoclasts. LPS significantly increased mRNA expression of osteoclast-related genes in RAW264.7 cells. Differentiated osteoclasts were treated with LPS (100 ng/ml) and the results showed a significantly increased mRNA expression of osteoclast-related genes and protein levels of RANK, TRAF6 and COX-2. Furthermore, LPS at 100 ng/ml significantly promoted the MAPK pathway including increasing the phosphorylation of c‑Jun N-terminal kinases (JNK) and the phosphorylation of the extracellular signal‑regulated kinase (ERK1/2). In conclusion, LPS promoted osteoclast differentiation and activation by enhancing RANK signaling and COX-2 expression. LPS also promoted osteoclast differentiation via activation of the JNK and ERK1/2 cell proliferation pathways.

Introduction

Bone is a dynamic tissue that constantly undergoes remodeling and involves a continuous process of bone formation and resorption (1). This process allows physiological bone growth and repair of damaged bones. Chronic osteomyelitis, an infectious bone disease, is a dysregulation of this process that results in excessive bone resorption (2). The receptor activator of nuclear factor-κB ligand (RANKL) is a key factor in regulating the process of osteoclast differentiation and maintaining the survival of mature osteoclasts (3). RANK signaling plays a critical role in regulating osteoclast differentiation and bone resorptive activity. RANK is activated by RANKL via the tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), which in turn induces the activation of mitogen-activated protein (MAP) kinases (MAPK) (4).

Lipopolysaccharide (LPS), a component of the outer membranes of all Gram-negative bacteria, is the first bacterial component shown to be capable of inducing bone resorption in vitro (5). LPS induces the production of cytokines including TNF-α by fibroblasts, macrophages, and other cells, which are able to induce osteoclast formation and activation (6). Moreover, LPS can promote osteoclast differentiation, fusion, survival and activation independent of IL-1, TNF-α, and RANKL (710). However, at present, effective therapeutic treatments for LPS-induced bone destruction are limited to antibiotics and surgical repair in chronic inflammatory diseases. Moreover, the role of LPS on osteoclast differentiation and activation through RANK signaling and COX-2 expression remains to be determined. Therefore, the research and development of LPS induced osteolysis remains a key objective for evaluating potential drugs in the prevention of bone destruction in infectious bone diseases.

The aim of the present study was to evaluate the effect of LPS on osteoclast differentiation and activation. We also aimed to clarify the mechanism of LPS on MAPK phosphorylation, as well as on the expression of RANK/TRAF6 and COX-2 in preosteoclasts.

Materials and methods

Reagents

Escherichia coli LPS (serotype 055:B5), Leukocyte Acid Phosphatase kit 387-A, and 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) were purchased from Sigma Co. (St. Louis, MO, USA). α modified-minimum essential medium (α-MEM), 10% fetal bovine serum (FBS), penicillin/streptomycin and TRIzol® reagent were purchased from Gibco (Rockville, MD, USA). RANKL and macrophage-colony stimulating factor (M-CSF) were purchased from Peprothec (Rocky Hill, NJ, USA). Primary antibodies against total c-Jun N-terminal kinase (JNK), phosphorylated JNK, extracellular signal-regulated kinase (ERK1/2), phosphorylated ERK1/2 and COX-2 were purchased from Cell Signaling Technology (Beverly, MA, USA). Primary antibodies against RANK, TRAF6 and β-actin were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). The mouse macrophage RAW264.7 cell line that can be induced to differentiate into osteoclasts was obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). Other chemicals and reagents used in this study were of analytical grade.

Cell culture

RAW264.7 cells were grown in α-MEM supplemented with 10% (v/v) FBS, 1% (v/v) penicillin-streptomycin solution, 10 mM HEPES solution and incubated at 37°C in 5% CO2 humidified air. The medium was changed every 3 days. For the cell viability assay, cells were seeded at 1×104 cells/cm2; for other experiments, cells were seeded at 4×105 cells/cm2. RAW264.7 were seeded into culture dishes and incubated overnight prior to treatment.

To evaluate the effect of LPS-induced osteoclastogenesis, the cells were treated with 10, 50, 100, 200 or 1000 ng/ml LPS or without LPS for 4 days until differentiation. To evaluate the effect of LPS on differentiated osteoclasts, RAW264.7 cells were treated with 50 ng/ml RANKL and 50 ng/ml M-CSF for 4 days. Then, 10, 50, 100, 200 or 1000 ng/ml LPS or without LPS were administered into cells in the presence of RANKL (50 ng/ml) and M-CSF (50 ng/ml) for 24 h.

Tartrate-resistant acid phosphatase staining (TRAP)

Cells were washed with phosphate-buffered saline (PBS) and fixed in 4% (v/v) paraformaldehyde at room temperature for 15 min. The cells were stained with TRAP using the Leukocyte Acid Phosphatase kit 387-A at 37°C for 60 min. Stained cells were then applied for microscopy (Olympus IX71, Olympus Optical, Tokyo, Japan). Osteoclasts were identified as TRAP-positive cells with ≥3 nuclei in each cell.

Cell viability assay

Cell viability was measured using MTT assay. RAW264.7 cells (1×103 cells/cm2) were seeded into 96-well flat bottom plates and incubated overnight prior to the experiment. To evaluate the effect of LPS on cell viability, RAW264.7 cells were treated with 10, 50, 100, 200 or 1000 ng/ml LPS or without LPS for 1 and 4 days. At the end of treatment, 20 μl MTT (5 mg/ml) was added to each well. Cells were cultured for an additional 4 h, the supernatant was removed, and 100 μl dimethyl sulfoxide was added to each well. Following agitation for 30 sec, absorbance was measured at 570 nm using a microplate reader (TECAN, Salzburg, Austria). The experiment was performed in triplicate.

Reverse transcription-PCR

Total RNA was isolated using TRIzol reagent (Invitrogen) and quantified by spectrophotometry. After isolation, 2 μg total RNA from each sample was reverse transcribed (RT) utilizing the M-MLV First Strand kit (Invitrogen) according to the manufacturer’s instructions. The primer sequences of TRAP, matrix metalloproteinase-9 (MMP-9), cathepsin K (CK) and carbonic anhydrase II (CAII) and β-actin (Generay Biotech Co., Ltd., Shanghai, China) and annealing temperatures used in this study are listed Table I. In each reaction, 1 μl cDNA, 12.5 μl 2X Taq Master Mix (Beijing CoWin Biotech Co., Ltd., Beijing, China) and 0.4 μM forward and reverse primers in a total volume of 25 μl were used. The initial denaturation was performed at 94°C for 3 min. The products were then subjected to denaturation at 94°C for 30 sec, annealing temperature for 30 sec, extension at 72°C for 30 sec for 32 cycles, and a final elongation at 72°C for 5 min. The PCR products were separated by 1.5% agarose gel electrophoresis, photographed using Gel-Doc (Bio-Rad, Hercules, CA, USA), and quantified by density determination using Quantity One image analysis software (Bio-Rad). β-actin was used as the internal control. Results were normalized to β-actin, and data were expressed as a ratio of gene to β-actin expression.

Table I

Primer sequences used in the RT-PCR analysis.

Table I

Primer sequences used in the RT-PCR analysis.

GeneAccession no.Sequences (5′→3′)PCR product size (bp)
TRAPNM_007388F: CACTCCCACCCTGAGATTTGT
R: CATCGTCTGCACGGTTCTG
118
MMP-9NM_013599F: CTGGACAGCCAGACACTAAAG
R: CTCGCGGCAAGTCTTCAGAG
145
CKNM_007802F: GAAGAAGACTCACCAGAAGCAG
R: TCCAGGTTATGGGCAGAGATT
102
CAIINM_009801F: TCCCACCACTGGGGATACAG
R: CTCTTGGACGCAGCTTTATCATA
170
COX-2NM_022415F: GGATGCGCTGAAACGTGGA
R: CAGGAATGAGTACACGAAGCC
133
β-actinNM_007393F: GGCTGTATTCCCCTCCATCG
R: CCAGTTGGTAACAATGCCATGT
154

[i] TRAP, tartrate-resistant acid phosphatase; MMP-9, matrix metalloproteinase-9; CK, cathepsin K; CAII, carbonic anhydrase II; COX-2, cyclooxygenase-2; F, forward; R, reverse.

Western blot analysis

At the end of treatment, the cell culture medium was aspirated and cells were detached in PBS by scrapping. Detached cells were transferred to fresh microcentrifuge tubes and centrifuged at 12,500 × g at 4°C for 10 min. Cell pellets were then lysed in 300 μl lysis buffer (Cytobuster protein extraction reagent, Novagen, Darmstadt, Germany) with 25 mM NaF, 1 mM Na3VO4, 1X protease inhibitor cocktail. Protein concentrations were determined using a standard Bradford assay. For western blotting, equal amounts of protein from each sample were loaded on SDS-PAGE and electrotransferred onto PVDF membranes (Millipore, Bedford, MA, USA). These membranes were then blocked with 5% (w/v) bovine serum albumin in TBST [10 mM Tris, 150 mM NaCl, and 0.1% (v/v) Tween-20, pH 7.5] for 1 h at room temperature, and incubated with primary antibodies overnight at 4°C. Secondary antibody incubation was at room temperature for 2 h (Santa Cruz Biotechnology, Inc.). Chemiluminescence ECL (Amersham, Arlington Heights, IL, USA) was used to detect immunoreactive protein signals. Protein signals were then visualized on films and scanned and quantified using the ImageJ software (National Institutes of Health Image, USA). For re-probing, the PVDF membranes were stripped with 0.2 M NaOH for 10 min before blocking with another primary antibody. The expression of molecules of interest was determined relative to β-actin.

Statistical analysis

Each experiment was repeated at least three times. Quantified results were presented as the mean ± SD. Significant differences were determined using factorial analysis of variance (ANOVA). P<0.05 was considered to indicate a statistically significant difference.

Results

LPS promoted osteoclast differentiation

The population of osteoclasts in LPS-treated cultures was higher than that in non-treated cultures (Fig. 1). LPS at 100 ng/ml induced the most osteoclast formation compared with other concentrations of LPS-treated cultures in RAW264.7 cells (P<0.01) and differentiated osteoclasts (P<0.01).

To ensure the effect of LPS on osteoclast differentiation, we evaluated cell viability in RAW264.7 cells. LPS at 100 ng/ml significantly increased cell viability at 24 h, while 1000 ng/ml LPS decreased cell viability at 24 h and 4 days (Fig. 2).

LPS promoted the mRNA expression of osteoclast-specific genes in RAW264.7 cells and differentiated osteoclasts

On differentiation day 4, LPS at 100 ng/ml significantly increased mRNA expression of TRAP, MMP-9, CK, CAII and COX-2 compared to other concentrations of LPS-treated groups as well as the non-treated group in RAW264.7 cells and differentiated osteoclasts (Fig. 3).

LPS promoted the expression of RANK, TRAF6 and COX-2 protein in differentiated osteoclasts

On differentiation day 4, LPS at 100 ng/ml significantly increased the protein expression of RANK, TRAF6 and COX-2 in differentiated osteoclasts. However, higher concentrations of LPS inhibited the protein expression of these molecules (Fig. 4).

LPS selectively regulated the activation of MAPK in RAW264.7 cells

Since MAP kinases are important regulators of inflammatory mediators and osteoclast differentiation, the effect of LPS on the activation of MAPKs in RAW264.7 cells were examined by western blotting. LPS treatment at 50, 100, 200 or 1000 ng/ml enhanced the protein levels of phosphorylated forms of ERK1/2 and JNK in RAW264.7 cells at 2 h (Fig. 5). When MAPK inhibitors SP600125 and PD98059 were added 2 h prior to LPS treatment, the enhanced phosphorylation of ERK1/2 and JNK in RAW264.7 cells was selectively attenuated (Fig. 6). This suggests LPS-induced osteoclast differentiation through selective activations of ERK1/2 and JNK, which were involved in the cell proliferation pathway.

Discussion

Bacteria-induced inflammatory response is an important contributor to excessive bone resorption in chronic inflammatory diseases such as septic arthritis, osteomyelitis, and infected orthopedic implant failure (2). LPS, a pro-inflammatory glycolipid and main component of the cell wall of Gram-negative bacteria, is well documented in Gram-negative bacteria-induced bone destruction. LPS stimulates osteoclastic bone resorption in vivo (11,12) and promotes osteoclast differentiation in whole bone marrow cell culture (13) and in preosteoclasts (7,8). However, LPS on osteoclast differentiation and activation through RANK signaling and COX-2 expression remains to be determined. In the present study, LPS increased the protein expression of RANK, TRAF6 and COX-2 and mRNA expression of osteoclast-related genes in differentiated osteoclasts (100 ng/ml LPS). In our previous study, LPS increased the protein expression of RANK, TRAF6 and COX-2 in RAW264.7 cells (14). Those results confirmed that LPS was able to stimulate osteoclast differentiation directly.

The effect of LPS on osteoclast resorptive activity is confirmed by evaluating mRNA expression levels of osteoclast-related genes. TRAP has been suggested to be involved in bone resorptive activity in osteoclasts (15,16). The proteolytic enzymes such as MMP-9, CAII and CK are highly expressed in osteoclasts and are involved in degradation of the bone matrix during bone resorption (1722). In the present study, the mRNA expression levels of TRAP, MMP-9, CAII and CK in RAW264.7 cells and mature osteoclasts were significantly enhanced by LPS treatment. Taken together, our data indicate that LPS promoted osteoclast differentiation and activity due to the upregulated osteoclast-related genes.

COX-2 plays a critical role in osteoclastogenic signaling pathways (23,24). Inhibition of COX-2 blocks osteoclast formation in vitro (25,26). In a previous study, we found that LPS upregulated the protein expression of COX-2 of RAW264.7 cells (14). In the current study, LPS upregulated the protein expression of COX-2 of differentiated osteoclasts. Taken together, these findings indicate that LPS promoted osteoclast differentiation by enhancing COX-2 expression.

RANK signal transduction has been reported to be essential for osteoclast differentiation, bone resorptive function, and survival (27,28). Mice deficient in RANKL or RANK genes exhibit severe osteopetrosis as they do not form osteoclasts (2932). The selective inhibition of RANK with RANK:Fc or RANK receptor inhibitor blocks osteoclast maturation and function in vivo or in vitro (3335). The selective inhibition of TRAF6 also blocks osteoclast maturation and function (36,27). Taken together with our previous study (14), results of the present study have shown that LPS enhanced the protein expression of RANK and TRAF6 in RAW264.7 cells and differentiated osteoclasts. These results suggest that RANK/TRAF6 signaling plays a prominent role in osteoclastogenesis downstream of LPS.

MAPKs are activated by a variety of stresses including LPS and then affect apoptosis either in a positive or negative manner (37). In many cell types, ERK1/2 is involved in cell proliferation and cell cycle progression, while the JNK pathway has been involved in both apoptosis and survival signaling (38). A novel ERK to JNK cross-activation has been reported, with JNK being the final mediator for the stimulation of cell proliferation by ERK (39). In the current study, LPS promoted osteoclast differentiation through the enhanced phosphorylation of ERK1/2 and JNK which were involved in the cell proliferation pathway.

In conclusion, our data suggest that LPS promoted osteoclast formation and activation by enhancing RANK signaling and COX-2 expression. LPS also promoted osteoclast differentiation through an enhanced cell proliferation pathway of ERK1/2 and JNK. Our findings indicate that blocking RANK signaling and COX-2 expression may be used as an evaluation agent to treat bacteria-induced bone resorption.

Acknowledgements

This study was supported by School Fund of Luohe Medical College (no. 2012-DF001).

References

1 

Roodman D: Advances in bone biology: the osteoclast. Endocr Rev. 17:308–332. 1996.PubMed/NCBI

2 

Nair SP, Meghji S, Wilson M, Reddi K, White P and Henderson B: Bacterially induced bone destruction: mechanisms and misconceptions. Infect Immun. 64:2371–2380. 1996.PubMed/NCBI

3 

Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT and Martin TJ: Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev. 20:345–357. 1999. View Article : Google Scholar : PubMed/NCBI

4 

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

5 

Shuto T, Jimi E, Kukita T, Hirata M and Koga T: Granulocytemacrophage colony stimulating factor suppresses lipopolysaccharide-induced osteoclast-like cell formation in mouse bone marrow cultures. Endocrinology. 134:831–837. 1994.

6 

Abu-Amer Y, Ross FP, Edwards J and Teitelbaum SL: Lipopolysaccharide-stimulated osteoclastogenesis is mediated by tumor necrosis factor via its P55 receptor. J Clin Invest. 100:1557–1565. 1997. View Article : Google Scholar : PubMed/NCBI

7 

Islam S, Hassan F, Tumurkhuu G, et al: Bacterial lipopolysaccharide induces osteoclast formation in RAW 264.7 macrophage cells. Biochem Biophys Res Commun. 360:346–351. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Mörmann M, Thederan M, Nackchbandi I, Giese T, Wagner C and Hänsch GM: Lipopolysaccharides (LPS) induce the differentiation of human monocytes to osteoclasts in a tumour necrosis factor (TNF) alpha-dependent manner: a link between infection and pathological bone resorption. Mol Immunol. 45:3330–3337. 2008.PubMed/NCBI

9 

Itoh K, Udagawa N, Kobayashi K, et al: Lipopolysaccharide promotes the survival of osteoclasts via toll-like receptor 4, but cytokine production of osteoclasts in response to lipopolysaccharide is different from that of macrophages. J Immunol. 170:3688–3695. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Takami M, Kim N, Rho J and Choi Y: Stimulation by toll-like receptors inhibits osteoclast differentiation. J Immunol. 169:1516–1523. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Orcel P, Feuga M, Bielakoff J and De Vernejoul MC: Local bone injections of LPS and M-CSF increase bone resorption by different pathways in vivo in rats. Am J Physiol. 264:E391–E397. 1993.PubMed/NCBI

12 

Chiang CY, Kyritsis G, Graves DT and Amar S: Interleukin-1 and tumor necrosis factor activities partially account for calvarial bone resorption induced by local injection of lipopolysaccharide. Infect Immun. 67:4231–4236. 1999.PubMed/NCBI

13 

Ito HO, Shuto T, Takada H and Koga T, Aida Y, Hirata M and Koga T: Lipopolysaccharides from Porphyromonas gingivalis, Prevotella intermedia and Actinobacillus actinomycetemcomitans promote osteoclastic differentiation in vitro. Arch Oral Biol. 41:439–444. 1996.

14 

Guo C, Hou GQ, Li XD, et al: Quercetin triggers apoptosis of lipopolysaccharide (LPS)-induced osteoclasts and inhibits bone resorption in RAW264.7 cells. Cell Physiol Biochem. 30:123–136. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Halleen JM, Raisanen S, Salo JJ, et al: Intracellular fragmentation of bone resorption products by reactive oxygen species generated by osteoclastic tartrate-resistant acid phosphatase. J Biol Chem. 274:22907–22910. 1999. View Article : Google Scholar

16 

Angel NZ, Walsh N, Forwood MR, Ostrowski MC, Cassady AI and Hume DA: Transgenic mice overexpressing tartrate-resistant acid phosphatase exhibit an increased rate of bone turnover. J Bone Miner Res. 15:103–110. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Reponen P, Sahlberg C, Munaut C, Thesleff I and Tryggvason K: High expression of 92-kD type IV collagenase (gelatinase B) in the osteoclast lineage during mouse development. J Cell Biol. 124:1091–1102. 1994. View Article : Google Scholar : PubMed/NCBI

18 

Yu X, Huang Y, Collin-Osdoby P and Osdoby P: Stromal cell-derived factor-1 (SDF-1) recruits osteoclast precursors by inducing chemotaxis, matrix metalloproteinase-9 (MMP-9) activity, and collagen transmigration. J Bone Miner Res. 18:1404–1418. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Wilson SR, Peters C, Saftig P and Brömme D: Cathepsin K activity-dependent regulation of osteoclast actin ring formation and bone resorption. J Biol Chem. 284:2584–2592. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Ishikawa T, Kamiyama M, Tani-Ishii N, et al: Inhibition of osteoclast differentiation and bone resorption by cathepsin K antisense oligonucleotides. Mol Carcinog. 32:84–91. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Kozloff KM, Quinti L, Patntirapong S, Hauschka PV, Tung CH, Weissleder R and Mahmood U: Non-invasive optical detection of cathepsin K-mediated fluorescence reveals osteoclast activity in vitro and in vivo. Bone. 44:190–198. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Lehenkari P, Hentunen TA, Laitala-Leinonen T, Tuukkanen J and Väänänen HK: Carbonic anhydrase II plays a major role in osteoclast differentiation and bone resorption by effecting the steady state intracellular pH and Ca2+. Exp Cell Res. 242:128–137. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Li L, Pettit AR, Gregory LS and Forwood MR: Regulation of bone biology by prostaglandin endoperoxide H synthases (PGHS): a rose by any other name. Cytokine Growth Factor Rev. 17:203–216. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Ono K, Akatsu T, Murakami T, et al: Involvement of cyclooxygenase-2 in osteoclast formation and bone destruction in bone metastasis of mammary carcinoma cell lines. J Bone Miner Res. 17:774–781. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Ono K, Akatsu T, Kugai N, Pilbeam CC and Raisz LG: The effect of deletion of cyclooxygenase-2, prostaglandin receptor EP2, or EP4 in bone marrow cells on osteoclasts induced by mouse mammary cancer cell lines. Bone. 33:798–804. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Kellinsalmi M, Parikka V, Risteli J, et al: Inhibition of cyclooxygenase-2 down-regulates osteoclast and osteoblast differentiation and favours adipocyte formation in vitro. Eur J Pharmacol. 572:102–110. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Armstrong AP, Tometsko ME, Glaccum M, Sutherland CL, Cosman D and Dougall WC: A RANK/TRAF6-dependent signal transduction pathway is essential for osteoclast cytoskeletal organization and resorptive function. J Biol Chem. 277:44347–44356. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Crockett JC, Mellis DJ, Scott DI and Helfrich MH: New knowledge on critical osteoclast formation and activation pathways from study of rare genetic diseases of osteoclasts: focus on the RANK/RANKL axis. Osteoporos Int. 22:1–20. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Kong YY, Yoshida H, Sarosi I, et al: OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 397:315–323. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Kim N, Odgren PR, Kim DK, Marks SC Jr and Choi Y: Diverse roles of the tumor necrosis factor family member TRANCE in skeletal physiology revealed by TRANCE deficiency and partial rescue by a lymphocyte-expressed TRANCE transgene. Proc Natl Acad Sci USA. 97:10905–10910. 2000. View Article : Google Scholar

31 

Li J, Sarosi I, Yan XQ, et al: RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc Natl Acad Sci USA. 97:1566–1571. 2000. View Article : Google Scholar : PubMed/NCBI

32 

Dougall WC, Glaccum M, Charrier K, et al: RANK is essential for osteoclast and lymph node development. Genes Dev. 13:2412–2424. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Feeley BT, Liu NQ, Conduah AH, et al: Mixed metastatic lung cancer lesions in bone are inhibited by noggin overexpression and RANK:Fc administration. J Bone Miner Res. 21:1571–1580. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Childs LM, Paschalis EP, Xing L, et al: In vivo RANK signaling blockade using the receptor activator of NF-kappaB:Fc effectively prevents and ameliorates wear debris-induced osteolysis via osteoclast depletion without inhibiting osteogenesis. J Bone Miner Res. 17:192–199. 2002. View Article : Google Scholar

35 

Kim H, Choi HK, Shin JH, et al: Selective inhibition of RANK blocks osteoclast maturation and function and prevents bone loss in mice. J Clin Invest. 119:813–825. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Poblenz AT, Jacoby JJ, Singh S and Darnay BG: Inhibition of RANKL-mediated osteoclast differentiation by selective TRAF6 decoy peptides. Biochem Biophys Res Commun. 359:510–515. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Pearson G, Robinson F, Beers Gibson T, et al: Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 22:153–183. 2001.PubMed/NCBI

38 

Zhang W and Liu HT: MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 12:9–18. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Pedram A, Razandi M and Levin ER: Extracellular signalregulated protein kinase/Jun kinase cross-talk underlies vascular endothelial cell growth factor-induced endothelial cell proliferation. J Biol Chem. 273:26722–26728. 1998. View Article : Google Scholar

Related Articles

Journal Cover

August 2013
Volume 32 Issue 2

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
Hou G, Guo C, Song G, Fang N, Fan W, Chen X, Yuan L and Wang Z: 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
APA
Hou, G., Guo, C., Song, G., Fang, N., Fan, W., Chen, X. ... Wang, Z. (2013). Lipopolysaccharide (LPS) promotes osteoclast differentiation and activation by enhancing the MAPK pathway and COX-2 expression in RAW264.7 cells. International Journal of Molecular Medicine, 32, 503-510. https://doi.org/10.3892/ijmm.2013.1406
MLA
Hou, G., Guo, C., Song, G., Fang, N., Fan, W., Chen, X., Yuan, L., Wang, Z."Lipopolysaccharide (LPS) promotes osteoclast differentiation and activation by enhancing the MAPK pathway and COX-2 expression in RAW264.7 cells". International Journal of Molecular Medicine 32.2 (2013): 503-510.
Chicago
Hou, G., Guo, C., Song, G., Fang, N., Fan, W., Chen, X., Yuan, L., Wang, Z."Lipopolysaccharide (LPS) promotes osteoclast differentiation and activation by enhancing the MAPK pathway and COX-2 expression in RAW264.7 cells". International Journal of Molecular Medicine 32, no. 2 (2013): 503-510. https://doi.org/10.3892/ijmm.2013.1406