Open Access

Tensile strength suppresses the osteogenesis of periodontal ligament cells in inflammatory microenvironments

  • Authors:
    • Chaofan Sun
    • Fen Liu
    • Shendan Cen
    • Lijiao Chen
    • Yi Wang
    • Hao Sun
    • Hui Deng
    • Rongdang Hu
  • View Affiliations

  • Published online on: May 29, 2017     https://doi.org/10.3892/mmr.2017.6644
  • Pages: 666-672
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to investigate the role of orthodontic force in osteogenesis differentiation, matrix deposition and mineralization in periodontal ligament cells (PDLCs) cells in inflammatory microenvironments. The mesenchymal origin of PDLCs was confirmed by vimentin and cytokeratin staining. PDLCs were exposed to inflammatory cytokines (5 ng/ml IL‑1β and 10 ng/ml TNF‑α) and/or tensile strength (0.5 Hz, 12% elongation) for 12, 24 or 48 h. Cell proliferation and tensile strength‑induced cytokine expression were assessed by MTT assay and ELISA, respectively. Runt‑related transcription factor 2 (RUNX2) and type I collagen (COL‑I) expression were analysed by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Additionally, alkaline phosphatase activity was measured, and the mineralization profile was evaluated by alizarin red S staining. PDLCs exposed to tensile strength in inflammatory microenvironments exhibited reduced proliferation and mineralization potential. Treatment with the inflammatory cytokines IL‑1β and TNF‑α increased RUNX2 expression levels; however, decreased COL‑I expression levels, indicating that bone formation and matrix deposition involve different mechanisms in PDL tissues. Notably, RUNX2 and COL‑I expression levels were decreased in PDLCs exposed to a combination of an inflammatory environment and loading strength. The decreased osteogenic potential in an inflammatory microenvironment under tensile strength suggests that orthodontic force may amplify periodontal destruction in orthodontic patients with periodontitis.

Introduction

The periodontal ligament (PDL) is a complex, highly specialized cellular connective tissue that supports the teeth by surrounding and anchoring them to the alveolar bone. In addition, periodontal ligament cells (PDLCs) facilitate the bone-regenerative potential of periodontium (1). The versatility of the PDL is primarily due to the ability of PDLCs to adapt to various microenvironments, including inflammatory and loading conditions (2).

An increasing number of patients with malocclusion and malpositioned teeth are opting for orthodontic therapy to correct these conditions and improve aesthetics and function. The adult periodontium is exposed to a variety of physical forces, including those caused by orthodontic therapy. Orthodontic force serves as biomechanical stress that promotes the remodelling process and metabolism of bone matrix (3). Earlier studies and models have investigated the biochemical responses evoked by biomechanical forces, including static compression (4), dynamic compression (5), centrifugal force (6) and tensile stress (7), on various oral tissues, including PDLs.

It is important to understand the physiological and homeostatic response of the periodontium to mechanical and physical forces under various conditions, including an inflammatory microenvironment. It is understood that periodontitis is associated with an increase in pro-inflammatory cytokines, including interleukin-1 beta (IL-1β) and tumour necrosis factor α (TNF-α) (8,9). IL-1β and TNF-α mediate the pathogenesis of periodontitis, and their expression promotes periodontal tissue destruction (10,11). Elevated IL-1β and TNF-α levels have previously been detected in the serum and gingival tissues of patients with periodontitis (12). Additionally, primary cells cultured from inflammatory PDL tissues were demonstrated to secrete IL-1β and TNF-α levels that were similar to those from healthy donors stimulated with exogenous cytokines (13). These cytokines are primary chemical mediators in the inflammatory microenvironment and serve as markers of a large number of inflammatory diseases, including periodontitis (14,15). Notably, there appears to be ambiguity regarding the exact effects of these cytokines in different cell types. For instance, studies on human mesenchymal stem cells (hMSCs) revealed that expression of the master transcription factor for osteoblast differentiation, runt-related transcription factor 2 (RUNX2), was enhanced in the presence of media pre-conditioned with increased pro-inflammatory cytokine levels from classically activated monocytes (16,17). By contrast, inflammatory stimuli additionally induced factors that facilitate bone resorption, including matrix metalloproteinases (MMPs) in PDL fibroblasts (18) and the anterior cruciate ligament (19). In vitro (20) and in vivo (21,22) studies have demonstrated that alveolar bone resorption is closely associated with MMP production. Notably, inflammatory cytokines appear to trigger cellular responses that facilitate bone apposition and bone resorption. Evidence regarding the mechanisms underlying the process of osteoblastogenesis in response to physical forces in inflammatory microenvironments is beginning to emerge. In particular, understanding the PDLC response to tensile strength and inflammatory status is pivotal for comprehending tooth movement in the inflammatory microenvironment.

Previously, two key factors important in PDL osteogenesis, RUNX2 and type I collagen (COL-I), have obtained increasing attention from researchers. RUNX2 is a transcription factor that serves an important role in the transcription of many osteogenesis regulating genes (23). COL-I is responsible for the formation of the primary fibril-forming collagen in the PDL, forms solid fibres located between the alveolar bone and the cementum, and functions as a cushion mitigating masticatory and orthodontic force loading (24,25). The present study investigated the combined effects of tensile strength, IL-1β and TNF-α in PDLCs. RUNX2 and COL-I mRNA and protein expression levels and alkaline phosphatase (ALP) activity were measured. The results obtained from this study will improve understanding of the risks associated with periodontitis in adult orthodontic patients, and the association between orthodontic force and periodontal inflammation.

Materials and methods

Isolation and identification of PDLCs

The present study was approved by the Ethics Committee of the School of Stomatology, Wenzhou Medical University (Wenzhou, China; no. WYKQ2013003). A total of 6 teeth were obtained from four systemically healthy patients (age, 18–28 years; two males and two females) during extraction as part of orthodontic management or tooth impaction treatment. Informed consent allowing the use of these teeth to harvest PDLCs for the current study was obtained from all patients. Extracted teeth were immediately processed for PDLC isolation. Sterile 1X PBS was used to gently clean the root surfaces of the extracted tooth and PDL tissues were gently scraped off the middle third portion of the roots. PDL tissues were digested with type I collagenase (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) solution (3 mg/ml dissolved in Hank's balanced salt solution) for 45 min at 37°C and suspended in α-minimum essential medium (α-MEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), supplemented with 100 U/ml penicillin (Hyclone; GE Healthcare Life Sciences, Logan, UT, USA), 100 U/ml streptomycin (Hyclone; GE Healthcare Life Sciences) and 100 M/ml ascorbic acid (Sigma-Aldrich; Merck KGaA). The tissues were incubated at 37°C in a humidified atmosphere of 5% CO2 for two weeks in 25 cm2 cell culture flasks (Corning Incorporated, Corning, NY, USA). The culture medium was replaced every three days and PDLCs were used for the study at passage 3 to 5.

To confirm their mesenchymal origin, PDLCs were subjected to immunocytochemical staining for vimentin and cytokeratin (Wuhan Boster Biological Technology, Ltd., Wuhan, China) according to methods previously described (26).

Cytokine treatment and application of tensile strength

PDLCs were seeded onto COL-I-coated silicone Bioflex® culture plates (Flexcell International Corporation, Burlington, NC, USA) supplemented with α-MEM medium at a density of 2×105 cells/well. When the cells reached 70–80% confluence, they were exposed to pro-inflammatory cytokines and tensile strength. As previously described (18), PDLCs were treated with 5 ng/ml IL-1β and 10 ng/ml TNF-α (Peprotec, Inc., Rocky Hill, NJ, USA), to simulate an inflammatory microenvironment in vitro. Following this, cells were subjected to uni-axial tensile strength [12% elongation (27) sinusoidal curve, 0.5 Hz (28)], using a FX-5000T Flexercell Tension Plus system (Flexcell International Corporation). Uni-axial tensile strength was designed to simulate occlusion and orthodontic tooth movement. The 12% tensile strength value was chosen according to finite element model data (29). Untreated PDLCs were incubated using the same apparatus and cultured on the same Bioflex plates. PDLCs were divided into four groups: Untreated cells [I(−)/T(−)]; cells treated with tensile strength alone [I(−)/T(+)]; cells treated with IL-1β/TNF-α alone [I(+)/T(−)]; and cells treated with tensile strength and IL-1β/TNF-α [I(+)/T(+)].

ELISA

Protein levels of IL-1β and TNF-α in the cell culture supernatants were compared using ELISA kits (cat. nos. EK101B4 and EK1824, respectively; Sunny Elisa, Hangzhou, China), according to the manufacturer's protocol. The absorbance was measured at a wavelength of 405 nm using a microplate reader (Tecan Group, Ltd., Mannedorf, Switzerland).

MTT assay

Proliferation of PDLCs following various treatments was determined by MTT assay (Amresco, LLC, Solon, OH, USA). A total of 320 µl MTT solution (5 g/l dissolved in ddH2O) was added to each well of a 6-well plate and cells were incubated at 37°C for 4 h. The MTT solution and culture medium was removed and 3,000 µl dimethyl sulfoxide was added. Complete dissolution of each sample was aided by 10 min of light-tight vibration. The absorbance was measured at a wavelength of 490 nm using a spectrophotometer. Pure culture medium without PDLCs was used as the blank control.

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

Total RNA was extracted from PDLCs following treatment using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). A PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China) was used to reverse transcribe cDNA from cellular RNA. qPCR was performed using a LightCycler® 480 Real-Time PCR Detection system (Roche Diagnostics, Basel, Switzerland) and the LightCycler® 480 SYBR-Green I Master qPCR kit (Roche Diagnostics) using the following parameters: 40 cycles of 10 sec at 95°C, 10 sec at 60°C and 20 sec at 72°C. Primer sequences of osteogenic genes, including RUNX2, COL-I and GAPDH were synthesized by Invitrogen; Thermo Fisher Scientific, Inc. GAPDH was employed as an internal reference. Primer sequences are listed in Table I.

Table I.

Primers sequences.

Table I.

Primers sequences.

GeneSequence
RUNX2F: 5′-CCCGTGGCCTTCAAGGT-3′
R: 5′-CGTTACCCGCCATGACAGTA-3′
COL-IF: 5′-CCAGAAGAACTGGTACATCAGCAA-3′
R: 5′-CGCCATACTCGAACTGGAATC-3′
GAPDHF: 5′-GAAGGTGAAGGTCGGAGTC-3′
R: 5′-GAGATGGTGATGGGATTTC-3′

[i] F, forward; R, reverse; RUNX2, runt-related transcription factor 2; COL-I, type I collagen.

Western blot analysis

PDLCs were washed three times with precooled PBS at 4°C and promptly lysed in high intensity radioimmunoprecipitation assay lysis buffer with 1 mM phenylmethylsulfonyl fluoride, then scraped from the Bioflex plates. The concentration of total protein in the lysates was assayed using the BCA Protein Quantitation kit (Fudebio, Hangzhou, China) according to the manufacturer's protocol. Total protein from cell lysates (50 µg per lane) were separated per lane by 8% SDS-PAGE and transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% skimmed milk for 2 h and subsequently incubated with primary antibodies against RUNX2 (1:1,000; cat. no. 8486; rabbit; Cell Signaling Technology, Inc., Danvers, MA, USA), COL-1 (1:10,000; cat. no. sc-8785; goat; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and GAPDH (1:10,000; cat. no. KC-5G5; rabbit; KangChen Bio-tech, Inc., Shanghai, China) overnight at 4°C. Following this, membranes were incubated for 2 h at room temperature with the following horseradish peroxidase (HRP)-conjugated secondary antibodies: Goat anti-rabbit IgG (1:5,000; cat. no. AP132P; EMD Millipore) and rabbit anti-goat IgG (1:10,000; cat. no. AP106P; EMD Millipore). Immunoreactive protein bands were detected by using Immobilon Western Chemiluminescent HRP Substrate (EMD Millipore). Protein levels were quantified using ChemiDoc XRS+ (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and Quantity One software (version 4.6.2; Bio-Rad Laboratories, Inc.).

ALP activity assay

To evaluate the impact of tensile strength and the inflammatory microenvironment on ALP activity, an ALP assay kit (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) was used according to the manufacturer's protocol. The optical density was measured at a wavelength of 405 nm using a microplate reader (Tecan Group, Ltd.).

Mineralization assay

To investigate osteogenic differentiation potential, cells were initially grown under the conditions described above for 48 h. Following this, cells were immediately cultured in commercial osteogenic media supplemented with 50 µg/ml ascorbic acid, 0.1 µM dexamethasone and 10 mM β-glycerophosphate (Cyagen Biosciences, Santa Clara, CA, USA) for 14 days (13,30). Cells were subsequently fixed in 4% formalin for 30 min at room temperature and stained with alizarin red S (Cyagen Biosciences) for 30 min. To quantify mineralization, the mineral stain was solubilized in 5% SDS in 0.5 ml 0.5 N HCl for 30 min. The absorbance was measured at a wavelength of 405 nm using a microplate reader.

Statistical analysis

All experiments were performed at least three times and all data are expressed as the mean ± standard deviation. The Student's t-test was used for comparison between two groups and one-way analysis of variance was used for multiple comparisons, followed by Tukey's post hoc test. Statistical analysis was performed using SPSS 19.0 (IBM SPSS, Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Morphology and phenotype of PDLCs

To confirm that cells derived from the PDL were of mesodermal origin, the presence of vimentin and cytokeratin was detected by staining (28). The observation that the cells were positive for vimentin (Fig. 1A) but negative for cytokeratin (Fig. 1B) was consistent with a mesodermal origin. Cells in PBS served as the negative control (Fig. 1C). PDLCs exhibited no particular morphological alterations and remained shuttle-shaped following stimulation with IL-1β+, TNF-α and/or tensile strength.

Tensile strength induces pro-inflammatory secretion with minimally increased proliferation

Tensile strength induced the secretion of pro-inflammatory cytokines in a time-dependent manner (Fig. 2) with a minimal increase in proliferation (Fig. 3). Continuous tensile strength up to 48 h led to the secretion of IL-1β (89.76±4.56 pg/ml) and TNF-α (77.52±3.51 pg/ml; Fig. 2). PDLCs treated with 5 ng/ml IL-1β and 10 ng/ml TNF-α exhibited a significant decrease in proliferative capacity compared with untreated controls. Tensile strength was demonstrated to have minimal influence on proliferation of PDLCs alone; however, it enhanced pro-inflammatory cytokine inhibition of PDLC proliferation (Fig. 3).

Tensile strength and a pro-inflammatory environment alters the osteogenic and matrix deposition potential of PDLCs

To investigate the osteogenic potential of PDLCs, the expression of osteogenic markers including RUNX2 and COL-I were determined. Tensile strength did not alter RUNX2 mRNA (Fig. 4A) or protein (Fig. 4B) expression levels in PDLCs. However, COL-I mRNA (Fig. 4C) and protein (Fig. 4D) expression levels were significantly reduced following tensile strength. Treatment with pro-inflammatory cytokines significantly increased mRNA and protein expression levels of RUNX2; however, decreased those of COL-I. The increase in RUNX2 induced by pro-inflammatory cytokines was markedly inhibited by tensile strength and was reduced below the levels observed in untreated controls. Similarly, downregulation of COL-I mRNA and protein expression levels induced by pro-inflammatory cytokines was accentuated by tensile strength. Combining tensile strength with pro-inflammatory cytokine exposure decreased COL-I mRNA and protein expression levels to markedly reduced levels compared with untreated PDLCs. Representative western blot images of COL-I and RUNX2 protein expression levels are presented in Fig. 4E.

ALP activity and mineralized nodule formation is decreased by tensile strength in an inflammatory environment in PDLCs

To determine the osteogenic and mineralization potential of PDLCs, the osteogenesis marker ALP and mineralization status were analysed. Cells were subjected to the conditions described above for 12, 24 or 48 h. ALP activity was reduced following pro-inflammatory cytokine treatment or tensile strength exposure. The greatest decrease in ALP activity was detected in cells treated with combined tensile strength and pro-inflammatory cytokines. The difference in ALP activity between the treatment groups was most marked 48 h post-exposure (Fig. 5). Increased mineralization in response to strength-induced osteogenic differentiation in the inflammatory microenvironment was measured using Alizarin Red S staining in PDLCs. A significant decrease in mineralization was observed in PDLCs treated with either pro-inflammatory cytokines or tensile strength followed by culturing in osteo-inductive medium for 14 days (Fig. 6A). Notably, a markedly significant decrease in Alizarin Red S staining was observed in cells exposed to the combination of pro-inflammatory cytokines and tensile strength (Fig. 6B).

Discussion

The demand for adult orthodontic treatment has grown exponentially in recent years (31). This increased demand has resulted in a proportional increase in the number of challenging cases, particularly for orthodontists providing services to patients with periodontal diseases (32,33). Unfortunately, the risks involved in orthodontic therapy in patients with periodontal disease remain to be fully elucidated. The present study demonstrated alterations in the osteogenic potential of PDLCs in response to tensile strength in inflammatory environments.

Cellular behaviour, including proliferation, is highly dependent on biochemical and physical stimuli in the extracellular environment. The present study revealed PDLCs were sensitive to inflammatory mediators but insensitive to tensile strength, which was consistent with previous studies. A previous study demonstrated that conditioned medium from macrophages with Porphyromonas gingivalis-lipopolysaccharide (LPS) stimulation reduced the proliferation of osteoblasts (34). Another study demonstrated that tensile force loading did not influence the viability of PDLCs (35). However, the present study revealed that suppression of PDLC proliferation was enhanced following exposure to a combination of inflammatory and tensile loading microenvironments. These findings indicated that mechanical strength may amplify the inhibitory effects of inflammatory mediators.

Orthodontic tooth movement causes bone remodelling in areas under tensile strength, and this process is coupled with an inflammatory reaction (36). Previous in vivo studies have demonstrated that orthodontic tooth movement resulted in increased levels of pro-inflammatory cytokines in periodontal tissues (37,38). The present study confirmed an increase in IL-1β and TNF-α levels following exposure to tensile strength.

These results indicated that combined IL-1β and TNF-α exposure and tensile force loading significantly decreased RUNX2 expression in PDLCs. This result is consistent with a previous PDLC study that reported that combined IL-1β and tensile strain loading decreased RUNX2 expression (39). In the present study, combined IL-1β and TNF-α exposure upregulated RUNX2 in PDLCs. The differences between this study and previous investigations may be due to the presence of TNF-α. Studies in hMSCs using LPS-stimulated conditioned media reported high levels of secreted TNF-α and a robust enhancement of RUNX2 expression (16). There is evidence that activated monocytes and T lymphocytes produce inflammatory cytokines that serve an essential role in the process of osteogenic differentiation of mesenchymal tissue-derived cells (40,41).

Notably, IL-1β and TNF-α exposure robustly enhanced RUNX2 and suppressed COL-I expression in PDLCs. Inflammatory cytokines, including IL-1β and TNF-α are involved in the up-regulation of MMPs (18,19). MMPs mediate connective tissue breakdown (11) and the degradation of numerous extracellular matrix proteins (42). The results of the present study are consistent with those of a previous study that demonstrated that the expression of COL-I was inhibited in various cell types cultured in inflammatory media (13). These results demonstrated that COL-I expression by PDLCs was markedly suppressed following combined exposure to IL-1β, TNF-α and tensile force loading. Cytokine exposure exhibited an inhibitory effect on COL-I expression, and a stimulatory effect on RUNX2 expression. This may explain the different patterns of bone formation and matrix deposition in PDL tissue. Biomechanical strength imposed on PDLCs in an inflammatory microenvironment appears to be detrimental and results in progressive downregulation of RUNX2 and COL-I.

As biomechanical strength is involved in decreased bone formation and matrix deposition under inflammatory conditions, it was hypothesized that biomechanical strength may have a negative effect on ALP activity and the mineralization process. It was observed that ALP activity was significantly suppressed in PDLCs that were exposed to tensile strength combined with pro-inflammatory cytokines. Evidence from in vitro studies revealed that PDLCs and MSCs interact and respond in different ways to cytokines or biomechanical strength. For instance, increased ALP activity in response to IL-1β and TNF-α or biomechanical strength was observed in hMSCs (43,44). On the other hand, decreased ALP activity in response to cytokines or biomechanical strength in PDLCs has been reported by numerous studies (13,15,35). ALP activity and mineralization are interdependent and associated with osteoblast differentiation. The present study revealed that the level of mineralization in the I(+)/T(+) group was significantly reduced compared with the I(+)/T(−) group. This result suggested that biomechanical strength may serve as a key stimulus influencing PDLC tolerance of inflammatory microenvironments.

In conclusion, the present study demonstrated that mechanical stresses, including tensile strength, enhanced the suppression of osteogenesis, matrix deposition and mineralization in PDLCs that is induced by an inflammatory microenvironment. Therefore, it is important to assess the periodontal health status of adults prior to orthodontic therapy.

Acknowledgements

The authors of the present study would like to thank Dr Gerald Volière from the School and Hospital of Stomatology, Wenzhou Medical University (Wenzhou, China) for reviewing and editing this manuscript. The present study was supported by grants from the National Natural Science Foundation of China (grant no. 81200795), the Zhejiang Provincial Natural Science Foundation of China (grant nos. Y207360 & LY12H14003), and the Medical and Health Science and Technology Plan of Zhejiang Province (grant no. 2015KYA149).

References

1 

Ivanovski S, Gronthos S, Shi S and Bartold PM: Stem cells in the periodontal ligament. Oral Dis. 12:358–363. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Agarwal S, Long P, Seyedain A, Piesco N, Shree A and Gassner R: A central role for the nuclear factor-kappaB pathway in anti-inflammatory and proinflammatory actions of mechanical strain. FASEB J. 17:899–901. 2003.PubMed/NCBI

3 

Liu M, Dai J, Lin Y, Yang L, Dong H, Li Y, Ding Y and Duan Y: Effect of the cyclic stretch on the expression of osteogenesis genes in human periodontal ligament cells. Gene. 491:187–193. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Lee YH, Nahm DS, Jung YK, Choi JY and Kim SG, Cho M, Kim MH, Chae CH and Kim SG: Differential gene expression of periodontal ligament cells after loading of static compressive force. J Periodontol. 78:446–452. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Nakao K, Goto T, Gunjigake KK, Konoo T, Kobayashi S and Yamaguchi K: Intermittent force induces high RANKL expression in human periodontal ligament cells. J Dent Res. 86:623–628. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Li J, Jiang L, Liao G, Chen G, Liu Y, Wang J, Zheng Y, Luo S and Zhao Z: Centrifugal forces within usually-used magnitude elicited a transitory and reversible change in proliferation and gene expression of osteoblastic cells UMR-106. Mol Biol Rep. 36:299–305. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Yang YQ, Li XT, Rabie AB, Fu MK and Zhang D: Human periodontal ligament cells express osteoblastic phenotypes under intermittent force loading in vitro. Front Biosci. 11:776–781. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Okada H and Murakami S: Cytokine expression in periodontal health and disease. Crit Rev Oral Biol Med. 9:248–266. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Gamonal J, Acevedo A, Bascones A, Jorge O and Silva A: Levels of interleukin-1 beta, -8 and -10 and RANTES in gingival crevicular fluid and cell populations in adult periodontitis patients and the effect of periodontal treatment. J Periodontol. 71:1535–1545. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Delima AJ, Oates T, Assuma R, Schwartz Z, Cochran D, Amar S and Graves DT: Soluble antagonists to interleukin-1 (IL-1) and tumor necrosis factor (TNF) inhibits loss of tissue attachment in experimental periodontitis. J Clin Periodontol. 28:233–240. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Graves DT and Cochran D: The contribution of interleukin-1 and tumor necrosis factor to periodontal tissue destruction. J Periodontol. 74:391–401. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Górska R, Gregorek H, Kowalski J, Laskus-Perendyk A, Syczewska M and Madalinski K: Relationship between clinical parameters and cytokine profiles in inflamed gingival tissue and serum samples from patients with chronic periodontitis. J Clin Periodontol. 30:1046–1052. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Liu N, Shi S, Deng M, Tang L, Zhang G, Liu N, Ding B, Liu W, Liu Y, Shi H, et al: High levels of β-catenin signaling reduce osteogenic differentiation of stem cells in inflammatory microenvironments through inhibition of the noncanonical Wnt pathway. J Bone Miner Res. 26:2082–2095. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Kim YS, Pi SH, Lee YM, Lee SI and Kim EC: The anti-inflammatory role of heme oxygenase-1 in lipopolysaccharide and cytokine-stimulated inducible nitric oxide synthase and nitric oxide production in human periodontal ligament cells. J Periodontol. 80:2045–2055. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Yang H, Gao LN, An Y, Hu CH, Jin F, Zhou J, Jin Y and Chen FM: Comparison of mesenchymal stem cells derived from gingival tissue and periodontal ligament in different incubation conditions. Biomaterials. 34:7033–7047. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Omar OM, Granéli C, Ekström K, Karlsson C, Johansson A, Lausmaa J, Wexell CL and Thomsen P: The stimulation of an osteogenic response by classical monocyte activation. Biomaterials. 32:8190–8204. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Ekström K, Omar O, Granéli C, Wang X, Vazirisani F and Thomsen P: Monocyte exosomes stimulate the osteogenic gene expression of mesenchymal stem cells. PLoS One. 8:e752272013. View Article : Google Scholar : PubMed/NCBI

18 

Rossa C Jr, Liu M, Patil C and Kirkwood KL: MKK3/6-p38 MAPK negatively regulates murine MMP-13 gene expression induced by IL-1beta and TNF-alpha in immortalized periodontal ligament fibroblasts. Matrix Biol. 24:478–488. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Wang Y, Tang Z, Xue R, Singh GK, Shi K, Lv Y and Yang L: Combined effects of TNF-α, IL-1β and HIF-1α on MMP-2 production in ACL fibroblasts under mechanical stretch: An in vitro study. J Orthop Res. 29:1008–1014. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Tsuji K, Uno K, Zhang GX and Tamura M: Periodontal ligament cells under intermittent tensile stress regulate mRNA expression of osteoprotegerin and tissue inhibitor of matrix metalloprotease-1 and -2. J Bone Miner Metab. 22:94–103. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Kusano K, Miyaura C, Inada M, Tamura T, Ito A, Nagase H, Kamoi K and Suda T: Regulation of matrix metalloproteinases (MMP-2, -3, -9 and -13) by interleukin-1 and interleukin-6 in mouse calvaria: Association of MMP induction with bone resorption. Endocrinology. 139:1338–1345. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Ma J, Kitti U, Teronen O, Sorsa T, Husa V, Laine P, Rönkä H, Salo T, Lindqvist C and Konttinen YT: Collagenases in different categories of peri-implant vertical bone loss. J Dent Res. 79:1870–1873. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Lian JB and Stein GS: Runx2/Cbfa1: A multifunctional regulator of bone formation. Curr Pharm Des. 9:2677–2685. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Matsuda N, Yokoyama K, Takeshita S and Watanabe M: Role of epidermal growth factor and its receptor in mechanical stress-induced differentiation of human periodontal ligament cells in vitro. Arch Oral Biol. 43:987–997. 1998. View Article : Google Scholar : PubMed/NCBI

25 

Kawarizadeh A, Bourauel C, Götz W and Jäger A: Early responses of periodontal ligament cells to mechanical stimulus in vivo. J Dent Res. 84:902–906. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Chen B, Sun HH, Wang HG, Kong H, Chen FM and Yu Q: The effects of human platelet lysate on dental pulp stem cells derived from impacted human third molars. Biomaterials. 33:5023–5035. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Wescott DC, Pinkerton MN, Gaffey BJ, Beggs KT, Milne TJ and Meikle MC: Osteogenic gene expression by human periodontal ligament cells under cyclic tension. J Dent Res. 86:1212–1216. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Han LM, Li S, Wang L and Xu Y: Cyclic tensile stress during physiological occlusal force enhances osteogenic differentiation of human periodontal ligament cells via ERK1/2-Elk1 MAPK pathway. DNA Cell Biol. 32:488–497. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Natali AN, Pavan PG and Scarpa C: Numerical analysis of tooth mobility: Formulation of a non-linear constitutive law for the periodontal ligament. Dent Mater. 20:623–629. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Yeh Y, Yang Y and Yuan K: Importance of CD44 in the proliferation and mineralization of periodontal ligament cells. J Periodontal Res. 49:827–835. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Pabari S, Moles DR and Cunningham SJ: Assessment of motivation and psychological characteristics of adult orthodontic patients. Am J Orthod Dentofacial Orthop. 140:e263–e272. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Hajishengallis G: Aging and its impact on innate immunity and inflammation: Implications for periodontitis. J Oral Biosci. 56:30–37. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Eke PI, Dye BA, Wei L, Thornton-Evans GO and Genco RJ: CDC Periodontal Disease Surveillance workgroup: James Beck (University of North Carolina, Chapel Hill, USA), Gordon Douglass (Past President, American Academy of Periodontology), Roy Page (University of Washin): Prevalence of periodontitis in adults in the United States: 2009 and 2010. J Dent Res. 91:914–920. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Wang Y, Wang H, Ye Q, Ye J, Xu C, Lin L, Deng H and Hu R: Co-regulation of LPS and tensile strain downregulating osteogenicity via c-fos expression. Life Sci. 93:38–43. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Yamaguchi M, Shimizu N, Shibata Y and Abiko Y: Effects of different magnitudes of tension-force on alkaline phosphatase activity in periodontal ligament cells. J Dent Res. 75:889–894. 1996. View Article : Google Scholar : PubMed/NCBI

36 

Long P, Hu J, Piesco N, Buckley M and Agarwal S: Low magnitude of tensile strain inhibits IL-1beta-dependent induction of pro-inflammatory cytokines and induces synthesis of IL-10 in human periodontal ligament cells in vitro. J Dent Res. 80:1416–1420. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Baba S, Kuroda N, Arai C, Nakamura Y and Sato T: Immunocompetent cells and cytokine expression in the rat periodontal ligament at the initial stage of orthodontic tooth movement. Arch Oral Biol. 56:466–473. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Bletsa A, Berggreen E and Brudvik P: Interleukin-1alpha and tumor necrosis factor-alpha expression during the early phases of orthodontic tooth movement in rats. Eur J Oral Sci. 114:423–429. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Nokhbehsaim M, Deschner B, Winter J, Reimann S, Bourauel C, Jepsen S, Jäger A and Deschner J: Contribution of orthodontic load to inflammation-mediated periodontal destruction. J Orofac Orthop. 71:390–402. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Rifas L: T-cell cytokine induction of BMP-2 regulates human mesenchymal stromal cell differentiation and mineralization. J Cell Biochem. 98:706–714. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Rifas L, Arackal S and Weitzmann MN: Inflammatory T cells rapidly induce differentiation of human bone marrow stromal cells into mature osteoblasts. J Cell Biochem. 88:650–659. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Nemoto T, Kajiya H, Tsuzuki T, Takahashi Y and Okabe K: Differential induction of collagens by mechanical stress in human periodontal ligament cells. Arch Oral Biol. 55:981–987. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Ding J, Ghali O, Lencel P, Broux O, Chauveau C, Devedjian JC, Hardouin P and Magne D: TNF-alpha and IL-1beta inhibit RUNX2 and collagen expression but increase alkaline phosphatase activity and mineralization in human mesenchymal stem cells. Life Sci. 84:499–504. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Zhang P, Wu Y, Jiang Z, Jiang L and Fang B: Osteogenic response of mesenchymal stem cells to continuous mechanical strain is dependent on ERK1/2-Runx2 signaling. Int J Mol Med. 29:1083–1089. 2012.PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 16 Issue 1

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
Sun C, Liu F, Cen S, Chen L, Wang Y, Sun H, Deng H and Hu R: Tensile strength suppresses the osteogenesis of periodontal ligament cells in inflammatory microenvironments. Mol Med Rep 16: 666-672, 2017
APA
Sun, C., Liu, F., Cen, S., Chen, L., Wang, Y., Sun, H. ... Hu, R. (2017). Tensile strength suppresses the osteogenesis of periodontal ligament cells in inflammatory microenvironments. Molecular Medicine Reports, 16, 666-672. https://doi.org/10.3892/mmr.2017.6644
MLA
Sun, C., Liu, F., Cen, S., Chen, L., Wang, Y., Sun, H., Deng, H., Hu, R."Tensile strength suppresses the osteogenesis of periodontal ligament cells in inflammatory microenvironments". Molecular Medicine Reports 16.1 (2017): 666-672.
Chicago
Sun, C., Liu, F., Cen, S., Chen, L., Wang, Y., Sun, H., Deng, H., Hu, R."Tensile strength suppresses the osteogenesis of periodontal ligament cells in inflammatory microenvironments". Molecular Medicine Reports 16, no. 1 (2017): 666-672. https://doi.org/10.3892/mmr.2017.6644