Calcitonin gene‑related peptide reduces Porphyromonas gingivalis LPS‑induced TNF‑α release and apoptosis in osteoblasts

  • Authors:
    • Yan Zhou
    • Huiyu Zhang
    • Gang Zhang
    • Yong He
    • Ping Zhang
    • Ziqi Sun
    • Yuxia Gao
    • Yinghui Tan
  • View Affiliations

  • Published online on: December 6, 2017     https://doi.org/10.3892/mmr.2017.8205
  • Pages: 3246-3254
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Periodontal diseases comprise mixed bacterial infections mainly caused by Gram‑negative anaerobic bacteria. Lipopolysaccharides (LPS) are important virulence factors and periodontal pathogens, which change local cytokine levels and promote osteoblast apoptosis, thereby leading to an imbalance in bone remodeling mechanisms and accelerating bone loss. Calcitonin gene‑related peptide (CGRP) is a vasoactive neuropeptide that is released from sensory nerves and has a positive effect on osteoblast proliferation and differentiation. In addition, this small molecule peptide is an important immune regulator in the inflammatory response. The aim of the present study was to assess the in vitro effects of CGRP on Porphyromonas gingivalis (Pg)LPS‑induced osteoblast apoptosis. Osteoblast cultures were stimulated either with various concentrations of PgLPS (0, 25, 50, 100, 500 and 1,000 ng/ml) for 48 h or with 500 ng/ml PgLPS for various lengths of time (0, 6, 12, 24, 48 and 72 h). The PgLPS‑stimulated cells were pretreated with different concentrations of CGRP (0, 1, 10, 100 and 1,000 nM) and cell viability and apoptotic rates were measured by Cell Counting kit‑8 assays and flow cytometry, respectively. CGRP, cleaved (c)‑Caspase‑8 and c‑Caspase‑3 protein expression levels were analyzed by western blotting. Changes in cytokine expression levels, which included tumor necrosis factor (TNF)‑α, interleukin (IL)‑1β, IL‑6, monocyte chemotactic protein (MCP)‑1 and MCP‑2, were measured by ELISA. PgLPS was demonstrated to inhibit osteoblast viability and promote apoptosis in a time‑ and concentration‑dependent manner. CGRP expression was revealed to reduce PgLPS‑induced cytostatic activity and apoptosis in osteoblasts. CGRP also suppressed the PgLPS‑induced release of TNF‑α and inhibited the activation of c‑Caspase‑3 and c‑Caspase‑8, thus preventing apoptosis in osteoblasts. CGRP may be an important neuropeptide in bone remodeling and may reduce osteoblast apoptosis in inflammatory conditions. These results may provide a solid foundation for CGRP to serve as a new target for the treatment of periodontitis.

Introduction

Bone loss may be caused by the imbalance of bone remodeling. Pathological changes in bone tissue not only depend on the number of osteoclasts and osteoblasts but also are closely associated with the apoptosis-mediated lifecycle of these cells (1). A number of studies have reported that apoptosis participates in the development and progression of periodontitis (2,3). Periodontal pathogens mainly include Gram-negative anaerobic bacteria. Lipopolysaccharides (LPSs) are a major component of the cell wall of Gram-negative bacteria and are considered to be a pivotal factor for inducing the destruction of alveolar bone tissue and for the development and progression of periodontitis (4,5). LPS has previously been demonstrated to directly inhibit the differentiation of periodontal target cells and to increase the absorption of periodontal tissues (68). Therefore, LPS is of growing concern for its role in alveolar bone resorption and periodontitis incidence. Previous studies have revealed that LPS and the metabolites of periodontal pathogens may affect matrix formation and cellular apoptosis. (9,10). Regarding this imbalanced process, the biological behavior of osteoblasts is particularly important. LPS may induce inflammatory responses in osteoblasts and abnormal osteoblast apoptosis, which may lead to a disorder in the number of cells coupled between bone resorption and formation, thereby causing bone loss (10). Currently, there are few effective treatments for bone destruction caused by bacteria, and a major objective of bone-loss prevention is to investigate the protection of osteoblast function and activity in the imbalanced state and to search for potential drug targets.

Calcitonin gene-related peptide (CGRP) was first extracted from medullary thyroid carcinoma tissue in 1971 (11). CGRP comprises 37 amino acids and includes two isomers, α-CGRP and β-CGRP. CGRP was revealed to be stored as secretory granules in sensory nerve endings (12), and recent studies have demonstrated that CGRP is widely distributed in the nervous, cardiovascular, respiratory and digestive systems (1316). CGRP exhibits diverse physiological effects in various tissues and in the immune response; exogenous CGRP may increase the number and size of osteoblast colonies (17). In bone tissues, CGRP is not only produced in sensory nerve fibers and endings, but also generated by osteoblasts, and it functions in both autocrine and paracrine signaling (18). It has been recently reported that CGRP may have a potential role in regulating inflammation, as it lies at the intersection of the nervous and immune systems (19,20). CGRP was reported to directly affect CD4+ T helper cells and influence the function of antigen-presenting cells to regulate adaptive immune responses (21). Additionally, CGRP was revealed to suppress the release of inflammatory cytokines, such as interleukin (IL)-1β, tumor necrosis factor (TNF)-α and carbon tetrachloride, from monocyte-macrophage cells and dendritic cells upon stimulation by inactivated bacteria or Toll-like receptor agonists (22,23). Numerous studies have provided an understanding that CGRP may be a potential regulator of inflammation that might inhibit the production of pro-inflammatory cytokines, as CGRP serves a major regulatory role in the inflammatory process (24,25). However, the effects of CGRP on Porphyromonas gingivalis (Pg)LPS-induced osteoblast apoptosis remained unclear. The present study demonstrated that CGRP blocks PgLPS-induced cytostatic activity and apoptosis, whereas TNF-α serves an important opposing role in this process.

Materials and methods

Cell culture and reagents

Primary osteoblasts were obtained from the calvaria of BALB/C mice according to the method described previously (26) and cultured in Dulbecco's modified Eagle's medium (DMEM; cat. no. 11965; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (cat. no. 10100; Gibco; Thermo Fisher Scientific, Inc.), 1.5 g/l sodium bicarbonate (cat. no. 25080; Gibco; Thermo Fisher Scientific, Inc.), 0.11 g/l sodium pyruvate (cat. no. 11360; Gibco; Thermo Fisher Scientific, Inc.) and 100 µg/ml penicillin/streptomycin (cat. no. 15140; Gibco; Thermo Fisher Scientific, Inc.). Cells were maintained in a humidified atmosphere containing 5% CO2 at 37°C. Peptide α-CGRP (cat. no. C0167) and TNF-α (cat. no. T6674) were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). PgLPS (Invitrogen; Thermo Fisher Scientific, Inc.) was reconstituted in distilled and deionized water, according to the manufacturer's protocol.

Cell viability assay

Cell viability was evaluated using a Cell Counting kit-8 (CCK-8; cat. no. C0038; Beyotime Institute of Biotechnology, Haimen, China), according to the manufacturer's protocol. Osteoblasts (1.0×104 cells/well) were seeded in 96-well plates and cultured at 4°C overnight. Cells were treated with PgLPS at different concentrations (0, 25, 50, 100, 500 and 1,000 ng/ml) in the aforementioned culture medium for different lengths of time (0, 6, 12, 24, 48 or 72 h). Cells cultured in medium with 0 ng/ml PgLPS or for 0 h incubation were used as the controls. Following treatment, 10 µl of CCK-8 was added to each well and incubated for 2 h, and the absorbance of each well was measured with an iMark 680 Microplate Absorbance Reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA) at 450 nm. All experiments were performed independently and repeated three times. The cell viabilities were normalized to the control group using SPSS software for Windows (version 18.0; SPSS, Inc., Chicago, IL, USA).

ELISA

Osteoblasts (1×104 cells/well) were pretreated with 100 nM CGRP at 37°C for 30 min, followed by treatment with 500 ng/ml PgLPS at 37°C for 48 h. Cells cultured in medium with 0 ng/ml PgLPS or for 0 h incubation were used as the controls ELISA kits for TNF-α (cat. no. MTA00B), IL-1β (cat. no. MLB00C), IL-6 (cat. no. M6000B), monocyte chemotactic protein 1 (MCP-1; cat. no. MJE00) and MCP-2 (cat. no. DY790) were all purchased from R&D Systems China Co., Ltd. (Shanghai, China) and used to determine the concentrations in cell culture supernatants according to the manufacturer's protocols.

The cells were centrifuged at 71.04 × g for 5 min at 25°C, and then 100 µl supernatant was collected and assessed for concentration of the above proteins. The optical density of each well was measured with a microplate reader at 450 nm and normalized to the control group. The level of absorbance for each tested sample was measured using the Microplate Reader 550 (Bio-Rad Laboratories, Inc.). The data were analyzed by using SPSS software for Windows (version 18.0; SPSS, Inc., Chicago, IL, USA).

Apoptosis assay

Osteoblasts (1×104 cells/well) were pretreated with 100 nM CGRP at 37°C for 30 min, followed by treatment with 500 ng/ml PgLPS at 37°C for 48 h. Cells (1×104 cells/well) were treated with PgLPS at different concentrations (0, 25, 50, 100, 500 and 1,000 ng/ml) in the aforementioned culture medium for different lengths of time (0, 6, 12, 24, 48 or 72 h). Cells cultured in medium with 0 ng/ml PgLPS or for 0 h incubation were used as the controls.

Apoptosis was assessed by flow cytometry using an Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining kit (cat. no. 556547; BD Biosciences, Franklin Lakes, NJ, USA), according to the manufacturer's protocol. Briefly, cells were collected and washed twice with cold PBS, resuspended in staining buffer containing Annexin V-FITC (0.025 µg/ml) and PI (1 µg/ml) and incubated for 15 min at 25°C in the dark. Cells were washed twice with PBS and apoptotic cells were analyzed by FACScan flow cytometer and CELLQuest software (version 4.0.2; BD Biosciences, Franklin Lakes, NJ, USA).

Western blot analysis

Osteoblast cultures (1×104 cells/well) were stimulated with 500 ng/ml PgLPS at 37°C for various lengths of time (0, 6, 12, 24, and 48 h). CGRP protein expression was assessed. Osteoblasts (1×104 cells/well) were pretreated with 100 nM CGRP at 37°C for 30 min, followed by treating with 500 ng/ml PgLPS at 37°C for 48 h. Cleaved (c)-Caspase-8, (c)-Caspase-3 and TNF-α protein expression was assessed.

Cells were treated and collected by centrifugation at 71.04 × g for 1 min at 4°C. Following washing with PBS, cells were lysed in radioimmunoprecipitation assay buffer [50 mM Tris, (pH 7.4) 150 mM NaCl; 1% sodium deoxycholate, 1 mM sodium orthovanadate; 1% Triton X-100; 0.1% SDS; 10 µg/ml aprotinin; 10 µg/ml leupeptin and 1 mM phenylmethylsulfonyl fluoride] on ice for 30 min. Supernatants were collected by centrifugation at 18,759 × g for 5 min at 4°C. Protein concentrations were measured using an Enhanced BCA Protein Assay Reagent (cat. no. P0010; Beyotime Institute of Biotechnology). A total of 30 µg cellular protein was boiled for 10 min and separated by 12% SDS-PAGE. Proteins were transferred to a polyvinylidene difluoride membrane (cat. no. 162-0177; Bio-Rad Laboratories, Inc.) at 50 V for 3 h at 4°C. Following blocking with 5% non-fat dried milk for 2 h at room temperature, membranes were incubated with primary antibodies as follows: CRGP (1:1,000; cat. no. 14959; CST Biological Reagents Company Ltd., Shanghai, China), cleaved (c)-Caspase-8 (1:500; cat. no. 9505; CST Biological Reagents Company Ltd.), c-Caspase-3 (1:500; cat. no. 9664; CST Biological Reagents Company Ltd.), TNF-α (1:1,000; cat. no. ab6671; Abcam, Shanghai, China) and actin (1:1,000; cat. no. sc-8432; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) overnight at 4°C. Membranes were washed three times in 1X TBS +0.1% Tween-20 (TBST), followed by incubation with horseradish peroxidase (HRP)-conjugated goat anti-rabbit (1:2,000; cat. no. 474-1506; Kirkegaard & Perry Laboratories, Inc.; SeraCare Life Sciences, Milford, MA, USA) or HRP-conjugated goat anti-mouse (1:2,000; cat. no. 474-1806; Kirkegaard & Perry Laboratories, Inc.; SeraCare Life Sciences) secondary antibodies for 2 h at room temperature. Following washing by 1X TBST, protein bands were visualized using the Enhanced Chemiluminescence kit (cat. no. 170-5061; Bio-Rad Laboratories, Inc.), according to the manufacturer's protocol. Densitometric analysis of the blots was performed using Quantity One Software (version 4-2; Bio-Rad Laboratories, Inc.) and normalized to actin expression levels.

Statistical analysis

All data are expressed as the mean ± standard deviation from at least three independent experiments. Statistical analysis was performed using one-way analysis of variance, followed by a Newman-Keuls Student's t-test for multiple comparisons. The analysis was conducted using SPSS software for Windows (version 18.0; SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

PgLPS induces the cell viability inhibition and apoptosis

To assess the effects of PgLPS on osteoblast viability, osteoblasts were first cultured with different concentrations of PgLPS for 48 h and Cell viability was assessed by CCK-8 and apoptosis was assessed by flow cytometry assay as stated above.

Osteoblast viability was significantly reduced by PgLPS treatment at concentrations of 50, 100, 500 and 1,000 ng/ml (P<0.05; Fig. 1A); no significant difference was identified between cells treated with 500 or 1,000 ng/ml PgLPS. Osteoblasts were subsequently stimulated with 500 ng/ml PgLPS for 0-72 h, followed by measurement of viability. The results indicated that as the stimulation time increased, the viability of the osteoblasts significantly decreased within 48 h (P<0.01; Fig. 1B), no significant difference was identified between 48 and 72 h treatment. Analysis of apoptotic rates revealed that osteoblast apoptosis increased with increasing concentrations of PgLPS and with increasing treatment time (Fig. 1C and D, respectively).

CGRP attenuates cell viability inhibition and apoptosis induced by PgLPS

Osteoblast cultures (1×104 cells/well) were stimulated with 500 ng/ml PgLPS at 37°C for various lengths of time (0, 6, 12, 24, and 48 h). Western blot analysis demonstrated that a transient increase occurred in CGRP protein expression at 6 h following PgLPS stimulation (Fig. 2A); thereafter, CGRP protein levels gradually and significantly decreased over time, between 12 and 48 h.

Figure 2.

Effects of CGRP pretreatment on PgLPS-induced cell viability inhibition and apoptosis. (A) Osteoblasts were treated with 500 ng/ml PgLPS for 0, 6, 12, 24 and 48 h, and the expression of CGRP protein levels were detected by western blot analysis. Actin was used as a loading control; band intensities normalized to actin are represented as the mean ± standard deviation from three separate experiments; **P<0.01 vs. untreated control. (B) Osteoblasts were pretreated with 0, 1, 10, 100 and 1,000 nM CGRP for 1 h, followed by treatment with or without 500 ng/ml PgLPS for 48 h, and cell viability was measured by Cell Counting kit-8 assay. Data are presented as the mean ± standard deviation (n=3) and normalized to the untreated control group; **P<0.01. (C and D) Osteoblasts were pretreated with 100 nM CGRP for 30 min, followed by treatment with 500 ng/ml PgLPS for 48 h. Apoptotic rates were measured by flow cytometry and data are present the mean ± standard deviation from three separate experiments; **P<0.01 vs. 0 h control. (E and F) Cells were treated as indicated in C; whole cell lysates were prepared and subjected to immunoblotting using antibodies against c-Caspase-8, c-Caspase3 and Actin. Band intensities were quantified by densitometric analysis and normalized to Actin. Data are presented as the mean ± standard deviation; **P<0.01. C, cleaved; CGRP, calcitonin gene-related peptide; FITC, fluorescein isothiocyanate; PgLPS, Porphyromonas gingivalis lipopolysaccharide; PI, propidium iodide.

Osteoblasts (1×104 cells/well) were pretreated with 100 nM CGRP at 37°C for 30 min, followed by treating with 500 ng/ml PgLPS at 37°C for 48 h. Results from the CCK-8 assay indicated that PgLPS-stimulated (500 ng/ml) osteoblasts pretreated with CGRP (100 nM) significantly reduced the cytostatic activity of PgLPS on osteoblasts (Fig. 2B). Apoptotic rates were measured by flow cytometry and, consistent with the above results, 100 nM CGRP pretreatment markedly suppressed the 500 ng/ml PgLPS-induced apoptosis in osteoblasts (Fig. 2C and D). In addition, CGRP (100 nM) pretreatment was demonstrated to suppress the PgLPS-induced upregulation of c-Caspase-3 and c-Caspase 8 protein expression levels (Fig. 2E and F).

CGRP blocks PgLPS-induced TNF-α expression in osteoblasts

PgLPS is a classic endotoxin and has long been considered a trigger of periodontal diseases (27). PgLPS can also induce cells to release large amounts of inflammatory cytokines, such as TNF-α, IL-1β and IL-6, and cause a series of inflammatory reactions (28). Cells (1×104 cells/well) were treated with PgLPS (500 ng/ml) in the aforementioned culture medium for different lengths of time (0, 6, 12, 24 or 48 h). Cells cultured in medium with 0 ng/ml PgLPS or for 0 h incubation were used as the controls. Results from the ELISA assays demonstrated that PgLPS (500 ng/ml) treatment promoted the expression of TNF-α, IL-1β, IL-6 and MCP-1 production in osteoblasts in a time-dependent manner (Fig. 3A). Although no significant changes were identified for MCP-2 expression between 0 and 6 h, expression significantly increased at 12, 24 and 48 h following PgLPS stimulation. In addition, pretreatment with CGRP did not effectively reduce IL-1β, IL-6, MCP-1 and MCP-2 production in osteoblasts treated with PgLPS, whereas TNF-α production was significantly inhibited (Fig. 3B). Osteoblasts (1×104 cells/well) were pretreated with 100 nM CGRP at 37°C for 30 min, followed by treating with 500 ng/ml PgLPS at 37°C for 48 h. Western blot analysis also demonstrated that the PgLPS-induced increase in TNF-α protein expression was suppressed by CGRP pretreatment (Fig. 3C and D).

TNF-α is a key molecule in osteoblasts viability inhibition and apoptosis induced by PgLPS and reversed by CGRP

CCK-8 cell viability assay and flow cytometric results revealed that CGRP pretreatment reversed the PgLPS-induced inhibition of osteoblast cell viability and increase in apoptosis; however, these effects were not observed in the additional presence of exogenous TNF-α (Fig. 4A and B). Furthermore, western blot protein expression analysis confirmed that CGRP only inhibited the PgLPS-induced upregulation of c-Caspase-8 and c-Caspase-3 levels; whereas, CGRP treatment was not able to inhibit the upregulation of c-Caspase-8 and c-Caspase-3 expression in the presence of both PgLPS and TNF-α. TNF-α is an important pro-inflammatory cytokine and a major bone resorption factor. TNF-α mainly acts on osteoclasts and osteoblasts, and it may cause osteoblast apoptosis (29). The current results demonstrate that CGRP inhibited PgLPS-induced apoptosis; however, this phenomenon was reversed by TNF-α expression. The present study hypothesized that TNF-α was the key factor serving an opposing role in the CGRP-induced inhibition of PgLPS-stimulated osteoblast apoptosis.

Discussion

The present study assessed the effects of CGRP on PgLPS-induced osteoblast apoptosis in vitro. The results revealed that PgLPS may inhibit osteoblast viability and promote apoptosis in a time- and concentration-dependent manner. CGRP expression was demonstrated to reduce PgLPS-induced cytostatic activity and apoptosis in osteoblasts, suggesting that CGRP may be a potential agent for the prevention and treatment of periodontitis.

A number of previous reports have focused on the effects of CGRP on cultured osteoblasts and have revealed that CGRP can regulate bone metabolism and stimulate osteoblasts differentiation (17,30,31). Nevertheless, only a few reports have reported on LPS-induced osteoblast apoptosis. A study published in 1997 (32) demonstrated that CGRP can inhibit LPS-induced TNF-α production in osteoblasts, which is in line with the results of the present study. However, the present study also further assessed the expression of several cytokines induced by LPS and evaluated the effect of CGRP on them. The present study successfully established a LPS-treated osteoblast cell model and elucidated more mechanisms associated with apoptosis.

Inflammation is characterized by an increase in the expression of inflammatory cytokines produced by cells of the activated innate and adaptive immune systems. It has been previously reported that the inflammatory response is closely related to the extent of systemic and local bone loss (33). LPS exposure was reported to stimulate osteoclastic bone resorption in vivo and inhibit osteoblast differentiation (34). An increasing amount of data has also demonstrated that PgLPS may directly induce cell death or apoptosis in osteoblasts (10,34). Additional studies have demonstrated that apoptosis serves an essential role in the development and progression of infectious diseases, autoimmune diseases and tumors (35). Osteoblasts cells are capable of secreting matrix and mineralizing into bone tissue; they are the major cells involved in bone remodeling (36). Osteoblast proliferation and apoptosis are of great importance to maintaining the balance of bone formation (37). In the course of periodontitis, bacterial-induced osteoblast apoptosis may be a major contributor to bone loss. A number of studies have revealed that LPS is present in plaque, saliva, gingival crevicular fluid, inflammatory cavities and diseased cementum, with high toxicity to periodontal tissues (3840). In addition, a previous study suggested that during alveolar bone resorption and periodontitis, bacteria do not directly invade gingival tissue; instead, the destruction of gingival tissue may be mediated by LPS cytotoxicity in the gingival crevice (37). A number of inflammatory cytokines are synthesized and secreted by periodontal tissues and cells in response to LPS exposure, and research has shown that LPS may inhibit osteoblast proliferation and differentiation, and suppress bone formation (34). A previous study reported that PgLPS was able to significantly inhibit alkaline phosphatase (ALP) activity in osteoblasts and decrease the formation of mineralization nodules in a dose- and time-dependent manner (7). Additionally, PgLPS was revealed to increase the protein expression levels of CD14 and LPS receptors, and the mRNA expression level of the bone resorption factor IL-1β. Similarly, LPS exposure was demonstrated to significantly inhibit ALP activity and collagen synthesis in the osteoblast cell line MC3T3-E1 (34). LPS expression has also been reported to facilitate osteoclast differentiation and activation by activating the mitogen-activated protein kinase signaling pathway (41), as well as induce osteoblast apoptosis by activating the c-Jun N-terminal kinase pathway (42). The present study demonstrated that PgLPS stimulation inhibited the viability and induced apoptosis in osteoblasts, which was in agreement with previous reports. In addition, CGRP pretreatment was revealed to reduce apoptotic rates that were induced by PgLPS exposure. Following PgLPS stimulation, osteoblasts released a large amount of inflammatory cytokines, including TNF-α, IL-1β, IL-6, MCP-1 and MCP-2; PgLPS stimulation also increased the expression of apoptotic signals c-Caspase-3 and c-Caspase-8. A number of previous studies reported that the change in cytokine expression levels, including IL-1β, IL-6, TNF-α, interferon-γ and leukemia inhibitory factor, was an important cause of bone loss (43,44). Among these, the increase of TNF-α expression was identified as one of the characteristics of bone loss (45). It is worth noting that, results from the present study demonstrated that TNF-α was markedly suppressed by CGRP pretreatment, whereas CGRP inhibited the PgLPS-activated caspase signal, which ultimately led to apoptosis. These results indicated that CGRP was able to block PgLPS-induced initiation of inflammation and thus may have a protective effect on osteoblasts.

In the serum of patients suffering from postmenopausal osteoporosis, the level of TNF-α expression was reportedly increased (46). Similarly, TNF-α expression was also revealed to be increased in the serum of patients with type 2 diabetes, which indicated that the change in TNF-α level may be an important cause for bone metabolism diseases (47). TNF-α has many physiological functions in the regulation of cell proliferation, differentiation and survival. In models of pathological bone loss, such as senile osteoporosis and bone resorption caused by chronic inflammation, the concentrations of TNF-α are increased, indicating that TNF-α may also be an important factor in regulating bone metabolism (48). Indeed, TNF-α alone was reported to cause osteoblast apoptosis and, in addition, TNF-α was revealed to inhibit collagen synthesis, ALP activity and osteocalcin synthesis (49). TNF-α may directly promote osteoclast maturation and differentiation by inducing the expression of macrophage colony-stimulating factor and receptor activator of nuclear factor κB ligand (RANKL) in osteoblasts (50). Additionally, TNF-α may directly promote RANKL-exposed precursor cells to differentiate into osteoclasts, activate mature osteoclasts and inhibit osteoclast apoptosis (51). According to the present results, CGRP pretreatment inhibited PgLPS-induced osteoblast apoptosis; however, this phenomenon was reversed by the addition of TNF-α. Although an accurate explanation of this phenomenon is difficult, it may be speculated that TNF-α serves a pivotal role in CGRP inhibition of PgLPS-induced apoptosis in osteoblasts; however, the exact mechanism involved requires further investigation.

Current treatments for periodontitis mainly consist of antibiotics; despite the therapeutic effects, long-term use of antibiotics may lead to oral dysbiosis, increased bacterial resistance to these antibiotics and gastrointestinal irritation, among other side effects. These factors have affected the clinical application of antibiotics for periodontitis (52). The present in vitro study demonstrated that the vasoactive peptide CGRP reduced the expression of inflammatory cytokines and osteoblast apoptosis. In particular, its significant inhibition of TNF-α suggested a new potential target of action for this peptide in the treatment of periodontitis. In addition, it may be also interesting to integrate the present results with the effects of the neuropeptidergic innervation in dental pulp as, in pulpitis, CGRP-positive fibers in the pulp increased, which also suggested that CGRP may serve a potential role in the regulation of this process. The present study on CGRP may also lead to a new direction for the treatment of pulpitis, which will be the focus of future research.

References

1 

Boyce BF, Rosenberg E, de Papp AE and Duong LT: The osteoclast, bone remodelling and treatment of metabolic bone disease. Eur J Clin Invest. 42:1332–1341. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Di Benedetto A, Gigante I, Colucci S and Grano M: Periodontal disease: Linking the primary inflammation to bone loss. Clin Dev Immunol. 2013:5037542013. View Article : Google Scholar : PubMed/NCBI

3 

Song B, Zhou T, Yang WL, Liu J and Shao LQ: Programmed cell death in periodontitis: Recent advances and future perspectives. Oral Dis. 23:609–619. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Nakao J, Fujii Y, Kusuyama J, Bandow K, Kakimoto K, Ohnishi T and Matsuguchi T: Low-intensity pulsed ultrasound (LIPUS) inhibits LPS-induced inflammatory responses of osteoblasts through TLR4-MyD88 dissociation. Bone. 58:17–25. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Ossola CA, Surkin PN, Pugnaloni A, Mohn CE, Elverdin JC and Fernandez-Solari J: Long-term treatment with methanandamide attenuates LPS-induced periodontitis in rats. Inflamm Res. 61:941–948. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Bandow K, Maeda A, Kakimoto K, Kusuyama J, Shamoto M, Ohnishi T and Matsuguchi T: Molecular mechanisms of the inhibitory effect of lipopolysaccharide (LPS) on osteoblast differentiation. Biochem Biophys Res Commun. 402:755–761. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Kadono H, Kido J, Kataoka M, Yamauchi N and Nagata T: Inhibition of osteoblastic cell differentiation by lipopolysaccharide extract from Porphyromonas gingivalis. Infect Immun. 67:2841–2846. 1999.PubMed/NCBI

8 

Kato H, Taguchi Y, Tominaga K, Umeda M and Tanaka A: Porphyromonas gingivalis LPS inhibits osteoblastic differentiation and promotes pro-inflammatory cytokine production in human periodontal ligament stem cells. Arch Oral Biol. 59:167–175. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Li Y, Shibata Y, Zhang L, Kuboyama N and Abiko Y: Periodontal pathogen aggregatibacter actinomycetemcomitans LPS induces mitochondria-dependent-apoptosis in human placental trophoblasts. Placenta. 32:11–19. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Thammasitboon K, Goldring SR and Boch JA: Role of macrophages in LPS-induced osteoblast and PDL cell apoptosis. Bone. 38:845–852. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Melvin KE, Miller HH and Tashjian AH Jr: Early diagnosis of medullary carcinoma of the thyroid gland by means of calcitonin assay. N Engl J Med. 285:1115–1120. 1971. View Article : Google Scholar : PubMed/NCBI

12 

Broad PM, Symes AJ, Thakker RV and Craig RK: Structure and methylation of the human calcitonin/alpha-CGRP gene. Nucleic Acids Res. 17:6999–7011. 1989. View Article : Google Scholar : PubMed/NCBI

13 

Baillie LD, Schmidhammer H and Mulligan SJ: Peripheral mu-opioid receptor mediated inhibition of calcium signaling and action potential-evoked calcium fluorescent transients in primary afferent CGRP nociceptive terminals. Neuropharmacology. 93:267–273. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Pereira RT, Costa LS, Oliveira IR, Araújo JC, Aerts M, Vigliano FA and Rosa PV: Relative distribution of gastrin-, CCK-8-, NPY- and CGRP-immunoreactive cells in the digestive tract of dorado (Salminus brasiliensis). Tissue Cell. 47:123–131. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Uddman R, Luts A and Sundler F: Occurrence and distribution of calcitonin gene-related peptide in the mammalian respiratory tract and middle ear. Cell Tissue Res. 241:551–555. 1985. View Article : Google Scholar : PubMed/NCBI

16 

Sun T, Guo Z, Liu CJ, Li MR, Li TP, Wang X and Yuan DJ: Preservation of CGRP in myocardium attenuates development of cardiac dysfunction in diabetic rats. Int J Cardiol. 220:226–234. 2016. View Article : Google Scholar : PubMed/NCBI

17 

He H, Chai J, Zhang S, Ding L, Yan P, Du W and Yang Z: CGRP may regulate bone metabolism through stimulating osteoblast differentiation and inhibiting osteoclast formation. Mol Med Rep. 13:3977–3984. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Tian G, Zhang G and Tan YH: Calcitonin gene-related peptide stimulates BMP-2 expression and the differentiation of human osteoblast-like cells in vitro. Acta Pharmacol Sin. 34:1467–1474. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Holzmann B: Modulation of immune responses by the neuropeptide CGRP. Amino Acids. 45:1–7. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Holzmann B: Antiinflammatory activities of CGRP modulating innate immune responses in health and disease. Curr Protein Pept Sci. 14:268–274. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Streilein JW: Ocular immune privilege: Therapeutic opportunities from an experiment of nature. Nat Rev Immunol. 3:879–889. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Carucci JA, Ignatius R, Wei Y, Cypess AM, Schaer DA, Pope M, Steinman RM and Mojsov S: Calcitonin gene-related peptide decreases expression of HLA-DR and CD86 by human dendritic cells and dampens dendritic cell-driven T cell-proliferative responses via the type I calcitonin gene-related peptide receptor. J Immunol. 164:3494–3499. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Harzenetter MD, Novotny AR, Gais P, Molina CA, Altmayr F and Holzmann B: Negative regulation of TLR responses by the neuropeptide CGRP is mediated by the transcriptional repressor ICER. J Immunol. 179:607–615. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Nagata S: Apoptosis by death factor. Cell. 88:355–365. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Kroeger I, Erhardt A, Abt D, Fischer M, Biburger M, Rau T, Neuhuber WL and Tiegs G: The neuropeptide calcitonin gene-related peptide (CGRP) prevents inflammatory liver injury in mice. J Hepatol. 51:342–353. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Orriss IR, Hajjawi MO, Huesa C, MacRae VE and Arnett TR: Optimisation of the differing conditions required for bone formation in vitro by primary osteoblasts from mice and rats. Int J Mol Med. 34:1201–1208. 2014. View Article : Google Scholar : PubMed/NCBI

27 

DeLeon-Pennell KY, de Castro Brás LE, Iyer RP, Bratton DR, Jin YF, Ripplinger CM and Lindsey ML: P. gingivalis lipopolysaccharide intensifies inflammation post-myocardial infarction through matrix metalloproteinase-9. J Mol Cell Cardiol. 76:218–226. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Ding PH, Darveau RP, Wang CY and Jin L: 3LPS-binding protein and its interactions with P. gingivalis LPS modulate pro-inflammatory response and toll-like receptor signaling in human oral keratinocytes. PLoS One. 12:e01732232017. View Article : Google Scholar : PubMed/NCBI

29 

Wu X, Feng X, He Y, et al: IL-4 administration exerts preventive effects via suppression of underlying inflammation and TNF-α-induced apoptosis in steroid-induced osteonecrosis. Osteoporos Int. 27:1827–1837. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Liang W, Zhuo X, Tang Z, Wei X and Li B: Calcitonin gene-related peptide stimulates proliferation and osteogenic differentiation of osteoporotic rat-derived bone mesenchymal stem cells. Mol Cell Biochem. 402:101–110. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Ma W, Zhang X, Shi S and Zhang Y: Neuropeptides stimulate human osteoblast activity and promote gap junctional intercellular communication. Neuropeptides. 47:179–186. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Millet I and Vignery A: The neuropeptide calcitonin gene-related peptide inhibits TNF-alpha but poorly induces IL-6 production by fetal rat osteoblasts. Cytokine. 9:999–1007. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Redlich K and Smolen JS: Inflammatory bone loss: Pathogenesis and therapeutic intervention. Nat Rev Drug Discov. 11:234–250. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Guo C, Yuan L, Wang JG, Wang F, Yang XK, Zhang FH, Song JL, Ma XY, Cheng Q and Song GH: Lipopolysaccharide (LPS) induces the apoptosis and inhibits osteoblast differentiation through JNK pathway in MC3T3-E1 cells. Inflammation. 37:621–631. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Favaloro B, Allocati N, Graziano V, Di Ilio C and De Laurenzi V: Role of apoptosis in disease. Aging (Albany NY). 4:330–349. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Meshcheryakova A, Mechtcheriakova D and Pietschmann P: Sphingosine 1-phosphate signaling in bone remodeling: Multifaceted roles and therapeutic potential. Expert Opin Ther Targets. 21:725–737. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Kesavalu L, Sathishkumar S, Bakthavatchalu V, Matthews C, Dawson D, Steffen M and Ebersole JL: Rat model of polymicrobial infection, immunity and alveolar bone resorption in periodontal disease. Infect Immun. 75:1704–1712. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Bai Y, Wei Y, Wu L, Wei J, Wang X and Bai Y: C/EBP β mediates endoplasmic reticulum stress regulated inflammatory response and extracellular matrix degradation in LPS-stimulated human periodontal ligament cells. Int J Mol Sci. 17:3852016. View Article : Google Scholar : PubMed/NCBI

39 

Jaedicke KM, Preshaw PM and Taylor JJ: Salivary cytokines as biomarkers of periodontal diseases. Periodontol 2000. 70:164–183. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Toyman U, Tuter G, Kurtis B, Kıvrak E, Bozkurt Ş, Yücel AA and Serdar M: Evaluation of gingival crevicular fluid levels of tissue plasminogen activator, plasminogen activator inhibitor 2, matrix metalloproteinase-3 and interleukin 1-β in patients with different periodontal diseases. J Periodontal Res. 50:44–51. 2015. View Article : Google Scholar : PubMed/NCBI

41 

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. View Article : Google Scholar : PubMed/NCBI

42 

Guo C, Wang SL, Xu ST, Wang JG and Song GH: SP600125 reduces lipopolysaccharide-induced apoptosis and restores the early-stage differentiation of osteoblasts inhibited by LPS through the MAPK pathway in MC3T3-E1 cells. Int J Mol Med. 35:1427–1434. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Kırzıoğlu FY, Tözüm Bulut M, Doğan B, Fentoğlu Ö, Özmen Ö, Çarsancaklı SA, Ergün AG, Özdem M and Orhan H: Anti-inflammatory effect of rosuvastatin decreases alveolar bone loss in experimental periodontitis. J Oral Sci. 59:247–255. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Yao Z, Lei W, Duan R, Li Y, Luo L and Boyce BF: RANKL cytokine enhances TNF-induced osteoclastogenesis independently of TNF receptor associated factor (TRAF) 6 by degrading TRAF3 in osteoclast precursors. J Biol Chem. 292:10169–10179. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Tzach-Nahman R, Mizraji G, Shapira L, Nussbaum G and Wilensky A: Oral infection with porphyromonas gingivalis induces peri-implantitis in a murine model: Evaluation of bone loss and the local inflammatory response. J Clin Periodontol. 44:739–748. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Al-Daghri NM, Aziz I, Yakout S, Aljohani NJ, Al-Saleh Y, Amer OE, Sheshah E, Younis GZ and Al-Badr FB: Inflammation as a contributing factor among postmenopausal Saudi women with osteoporosis. Medicine (Baltimore). 96:e57802017. View Article : Google Scholar : PubMed/NCBI

47 

Chen YL, Qiao YC, Xu Y, Ling W, Pan YH, Huang YC, Geng LJ, Zhao HL and Zhang XX: Serum TNF-α concentrations in type 2 diabetes mellitus patients and diabetic nephropathy patients: A systematic review and meta-analysis. Immunol Lett. 186:52–58. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Wang C, Yu X, Yan Y, Yang W, Zhang S, Xiang Y, Zhang J and Wang W: Tumor necrosis factor-alpha: a key contributor to intervertebral disc degeneration. Acta Biochim Biophys Sin (Shanghai). 49:1–13. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Centrella M, McCarthy TL and Canalis E: Tumor necrosis factor-alpha inhibits collagen synthesis and alkaline phosphatase activity independently of its effect on deoxyribonucleic acid synthesis in osteoblast-enriched bone cell cultures. Endocrinology. 123:1442–1448. 1988. View Article : Google Scholar : PubMed/NCBI

50 

Lam J, Takeshita S, Barker JE, Kanagawa O, Ross FP and Teitelbaum SL: TNF-alpha induces osteoclastogenesis by direct stimulation of macrophages exposed to permissive levels of RANK ligand. J Clin Invest. 106:1481–1488. 2000. View Article : Google Scholar : PubMed/NCBI

51 

Park J, Choi HM, Yang HI, Yoo MC and Kim KS: Increased expression of IL-1 receptors in response to IL-1β may produce more IL-6, IL-8, VEGF and PGE2 in senescent synovial cells induced in vitro than in presenescent cells. Rheumatol Int. 32:2005–2010. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Santos RS, Macedo RF, Souza EA, Soares RS, Feitosa DS and Sarmento CF: The use of systemic antibiotics in the treatment of refractory periodontitis: A systematic review. J Am Dent Assoc. 147:577–585. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou Y, Zhang H, Zhang G, He Y, Zhang P, Sun Z, Gao Y and Tan Y: Calcitonin gene‑related peptide reduces Porphyromonas gingivalis LPS‑induced TNF‑α release and apoptosis in osteoblasts. Mol Med Rep 17: 3246-3254, 2018
APA
Zhou, Y., Zhang, H., Zhang, G., He, Y., Zhang, P., Sun, Z. ... Tan, Y. (2018). Calcitonin gene‑related peptide reduces Porphyromonas gingivalis LPS‑induced TNF‑α release and apoptosis in osteoblasts. Molecular Medicine Reports, 17, 3246-3254. https://doi.org/10.3892/mmr.2017.8205
MLA
Zhou, Y., Zhang, H., Zhang, G., He, Y., Zhang, P., Sun, Z., Gao, Y., Tan, Y."Calcitonin gene‑related peptide reduces Porphyromonas gingivalis LPS‑induced TNF‑α release and apoptosis in osteoblasts". Molecular Medicine Reports 17.2 (2018): 3246-3254.
Chicago
Zhou, Y., Zhang, H., Zhang, G., He, Y., Zhang, P., Sun, Z., Gao, Y., Tan, Y."Calcitonin gene‑related peptide reduces Porphyromonas gingivalis LPS‑induced TNF‑α release and apoptosis in osteoblasts". Molecular Medicine Reports 17, no. 2 (2018): 3246-3254. https://doi.org/10.3892/mmr.2017.8205