Open Access

Concentrated growth factor exudate enhances the proliferation of human periodontal ligament cells in the presence of TNF‑α

  • Authors:
    • Xiaoju Li
    • Huixiao Yang
    • Zijian Zhang
    • Zhonghai Yan
    • Huling Lv
    • Yan Zhang
    • Bin Wu
  • View Affiliations

  • Published online on: December 3, 2018     https://doi.org/10.3892/mmr.2018.9714
  • Pages: 943-950
  • Copyright: © Li 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 purpose of this study was to evaluate the effects of concentrated growth factor exudate (CGFe) on human periodontal ligament cells (hPDLCs) stimulated by tumor necrosis factor (TNF)‑α. From the peripheral blood of healthy donors, CGFe was prepared according to the Sacco protocol after 7 days of incubation. The hPDLCs were cultured by a tissue explant method and identified with anti‑vimentin and anti‑cytokeratin antibodies. Cells were subjected to four different treatments: i) Control; ii) TNF‑α (10 ng/ml); iii) CGFe (concentration 50%); and iv) CGFe+TNF‑α. The proliferation of hPDLCs was measured with Cell Counting Kit‑8 assays. Osteogenic differentiation and mineralization were determined by Alizarin Red S staining, alkaline phosphatase activity, western blotting and reverse transcription‑quantitative polymerase chain reaction. CGFe enhanced cell proliferation and upregulated ALP activity, the mineralization level, and osteogenic‑associated osteocalcin, runt‑related transcription factor 2 and Osterix gene expression in hPDLCs under inflammatory conditions induced by TNF‑α. The present study demonstrated that CGFe enhanced hPDLC proliferation and osteogenic differentiation in the presence of TNF‑α‑induced inflammation. As CGFe can be obtained from the venous blood of patients, it generates no immune reaction. Thus, it is more economical and beneficial to use CGFe in clinical periodontal regeneration practice than synthetic growth factors.

Introduction

Periodontitis and chronic apical periodontitis often cause periodontal tissue destruction and severe alveolar bone defects (1). In periodontally compromised teeth, when partial or full socket wall destruction is evident, connective tissue will grow into the extraction site and lead to a deficient ridge (1). Periodontitis is widely treated with instant implant technology in clinical practice (2). However, periodontal tissue destruction and alveolar bone defects significantly weaken the stability and shorten the service life of dental implants (2). To improve the success rate of dental implants, concentrated growth factor (CGF) can be used to promote periodontal tissue regeneration and osteogenic differentiation (3). CGF is a gel-like substance, obtained by centrifugation of venous blood, which is rich in growth factors and fibrin (48), and CGF combined with bone graft material promotes immediate periodontal tissue regeneration and osteogenic differentiation (8,9).

Previous studies have reported that platelet-rich plasma (PRP) and platelet-rich fibrin (PRF) in the first and second phases promote tissue regeneration, inhibit infection, regulate inflammation and reduce postoperative reactions (914). CGF is a third-phase plasma extract, consisting of multiple growth factors (48). Due to the unique structure of CGF fibrin, it can replace the diaphragm used in guiding bone regeneration to a certain extent (8,9,15). CGF has been widely studied in the fields of oral implantation, extraction site preservation, jaw cyst treatment, and fracture healing promotion (48,16,17).

CGF exudate (CGFe) is extracted from CGF and used in research labs to study the effects of CGF. Previous studies have shown that CGFe significantly shortens the time for osteogenesis in the operational area and distinctly improves bone formation quality (8,9). CGFe has been demonstrated to stimulate the proliferation of human periodontal ligament fibroblasts (PDLFs) (17). However, these previous studies were performed without the influence of inflammatory factors, and therefore did not sufficiently reflect the clinical environment. Particularly, the effect of CGFe on enhancing the proliferation of hPDLCs in the presence of tumor necrosis factor (TNF)-α-induced inflammation has not yet been investigated, to the best of our knowledge.

TNF-α is a pro-inflammatory cytokine that is a critical pathological factor for the development of apical inflammation, which significantly inhibits osteogenic differentiation (16,1821). As TNF-α-induced inflammation is commonly encountered in clinical practice (16,20), it is of great clinical interest to evaluate the effects of CGFe in the presence of TNF-α.

In the present study, the effects of CGFe on hPDLC proliferation and osteogenic differentiation were investigated in an inflammatory environment, stimulated by TNF-α. Specifically, the effects of CGFe on alkaline phosphatase (ALP) activity, mineralization, and osteocalcin (OCN), runt-related transcription factor 2 (RUNX2), and Osterix (OSX) gene expression in hPDLCs under TNF-α-induced inflammatory conditions were examined.

Materials and methods

Preparation of CGFe

The present study was approved by the Ethics Committee of the Jilin University Health Science Center (Changchun, China). In accordance with this committee, CGFe was obtained from three healthy male donors who had visited the outpatient clinic at the Health Center of Jilin University (Changchun, China) from August 2017 to March 2018. They were non-smokers and non-drinkers aged from 22 to 30 years old, and their informed consent was obtained. The experiments described below were carried out under similar conditions and procedures as the previous study (11).

Venous blood samples from the donors were used to produce human CGF according to a previously described protocol (22). In brief, blood samples were centrifuged at 750 × g for 12 min at 4°C. Between the acellular plasma and red blood cells (RBCs), a white CGF clot formed. The CGF clot was held with sterile forceps, separated from the RBCs using scissors, placed on the grid of an endo box and compressed by the endo box cover. After 1 min of applied pressure, the CGF clot was converted into CGF membrane, and the CGF exudate (CGFe) was collected in the tray of the endo box.

The CGFe was centrifuged at 500 × g for 5 min at 4°C to remove RBCs. Then, the CGFe was precipitated and filtered using a 0.22 µm sterile syringe filter unit (Merck KGaA, Darmstadt, Germany). The pooled CGFe samples were stored at −80°C prior to use. The original concentration of CGFe was defined as 100%, and a 50% concentration of CGFe was obtained by dilution with Minimal Essential Medium-α (α-MEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and used in subsequent experiments.

Human periodontal ligament cell (hPDLC) culture

In total, 10 healthy and noncarious premolars from three male and two female donors aged from 13 to 18 years old who had received orthodontic treatment at the Oral and Maxillofacial Surgery Department of the Stomatology School of Jilin University (Changchun, China) were obtained with informed consent. Periodontal ligaments were gently scraped from the middle third of the tooth-root surface with a sharp scalpel, minced with ophthalmic scissors and rinsed with α-MEM. These explants were cultured in α-MEM supplemented with 10% fetal bovine serum (FBS; GE Healthcare Life Sciences, Chicago, IL, USA), 1% streptomycin and penicillin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in 5% CO2. Examination by inverted light microscopy (IX73; Olympus Corporation, Tokyo, Japan) at magnifications of ×20 or ×40 was performed daily, and the medium was replaced every 3 days. Once cells reached 80% confluence, they were transferred to a 75 cm2 flask, and this was defined as passage one. The same procedure was performed repeatedly to produce multiple passages. Cells prior to passage 6 were used in the present study.

Immunocytochemistry staining

hPDLCs at passage 3 were seeded into several 24-well plates at a density of 1×104/well and covered in advance with circular coverslips (diameter, 14 mm) and incubated for 48 h at 37°C. Cells were then rinsed three times with 0.01 M PBS and fixed with 4% paraformaldehyde for 20 min at room temperature. Following a further wash with PBS, 0.25% Triton X-100 was added into the 24-well plates, which were incubated at 37°C for 15 min. Endogenous peroxidase activity was eliminated by incubation with 3% H2O2 for 10 min at room temperature. Then, hPDLCs were incubated with 1% bovine serum albumin (Gibco; Thermo Fisher Scientific, Inc.) and 22.52 mg/ml glycine in PBS + 0.1% Tween-20 for 30 min at room temperature. Next, cells were incubated with anti-vimentin (1:100; cat. no. ab24525; Abcam, Cambridge, MA, USA) and anti-cytokeratin (1:200; cat. no. AM06387SU-N; OriGene Technologies, Inc., Beijing, China) primary antibodies overnight at 4°C. The SP immunohistochemistry assay kit (cat. no. SP9001; ZSGB-BIO; OriGene Technologies, Inc., Beijing, China) was used for immunocytochemical staining according to the manufacturer's instructions, and a 3,3′-diaminobenzidine was used to stain positive cells. An inverted microscope (IX73; Olympus Corporation, Tokyo, Japan) was used to view the cells at magnifications of ×20 or ×40.

Cell Counting Kit (CCK)-8 assay

The objective of this experiment was to examine the effect of CGFe on the proliferation of hPDLCs in vitro. CGFe was obtained according to a previous protocol (16). The CCK-8 assay (Dojindo Laboratories, Kumamoto, Japan) was used to determine the effects of CGFe on hPDLC proliferation.

hPDLCs (2×103/well) were seeded into a 96-well plates containing complete medium (α-MEM supplemented with 10% FBS, 1% streptomycin and penicillin) with 10% FBS and incubated for 24 h at 37°C. Then, hPDLCs were exposed to CGFe (concentration 50%), TNF-α (10 ng/ml), or CGFe+TNF-α for 24, 48 or 72 h at 37°C, respectively. There was one additional control group with no CGFe or TNF-α treatment (complete medium with 10% FBS only). Cell proliferation was determined using the CCK-8 kit at the specified time-points. Kit reagent (10 µl) was added to the culture medium in each well. After a 90 min incubation at 37°C, absorbance at 450 nm was detected using an automatic microplate reader (Infinite 200 PRO; Tecan Group Ltd., Mannedorf, Switzerland). A well containing complete medium and CCK-8 solution without seeding cells was used as a further control. The assay was performed in duplicate, and the experiments were repeated six times under the same conditions.

ALP activity assay

hPDLCs (500 µl; 1×104/well) were seeded into 24-well plates and incubated for 24 h at 37°C. Next, the cells were exposed to CGFe, TNF-α or CGFe+TNF-α for 7 or 14 days. At the given time-points, cells were lysed with 0.1% Triton X-100, and the lysates were centrifuged at 8000 × g for 10 min at 4°C. The supernatant (50 µl/well) was added to 96-well plates, and ALP activity was examined with a ALP assay kit (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) according to the manufacturer's protocols. The optical density (OD) values were read at 520 nm with an automatic microplate reader (Infinite 200 PRO; Tecan Group, Ltd.).

Osteogenic differentiation induction and Alizarin Red S staining

hPDLCs (500 µl; 1×104/well) were seeded into 24-well plates in standard medium (α-MEM supplemented with 10% FBS, 1% streptomycin and penicillin) until 60–70% confluence was reached. The medium was then replaced with four different media: i) Mineral induction medium (MM; α-MEM medium containing 10% FBS, 50 mg/ml ascorbic acid, 10 mM β-glycerolphosphate and 10−8 M dexamethasone); ii) MM with CGFe (concentration 50%); iii) MM with TNF-α (10 ng/ml); or iv) MM with CGFe+TNF-α (10 ng/ml). The medium was replaced every 3 days.

Alizarin Red staining was used to detect and quantify the formation of mineralized nodules after 21 days. This assay was performed according to a previously described protocol, with minor modifications (9,23). Specifically, the hPDLCs were fixed with 95% ethanol for 15 min at room temperature, washed twice with dH2O and stained with 0.1% Alizarin Red S solution (pH 4.1) for 20 min at room temperature. hPDLCs were observed under an inverted phase-contrast microscope (IX73; Olympus Corporation, Tokyo, Japan; magnification, ×20 or ×40). Next, the hPDLCs were washed three times with dH2O. To semi-quantify the content of mineralized matrix nodules generated from the hPDLCs, 100 mM cetyl pyridinium chloride was added to the 24-well plates to dissolve and release the calcium-combined Alizarin Red S into solution. OD values were read at 570 nm, which represented the relative quantity of mineralization nodules.

Western blotting

Cell lysates were prepared in radioimmunoprecipitation assay buffer (150 mM NaCl, 0.1% SDS, 1 mM PMSF, 10 mM Tris-Cl, pH 7.4, 1% sodium deoxycholate, 1% Triton X-100). The cells were treated with four different media (control, CGFe, TNF-α, or CGFe+TNF-α) for 14 days. The cell lysates were incubated for 30 min on ice, then clarified by centrifugation at 6,000 × g for 10 min at 4°C. Protein concentration was determined with a bicinchoninic acid assay. Lysate samples (20 µl) were denatured and resolved by 10% SDS-PAGE, transferred onto polyvinylidene difluoride membranes and run at 300 mA for 2 h. Membranes were blocked in 5% non-fat milk at room temperature for 1 h, then incubated with primary antibodies against RUNX2 (1:1,000; cat. no. 12556; Cell Signaling Technology, Inc., Danvers, MA, USA) and Osterix (1:1,000; cat. no. ab229258; Abcam) and GAPDH overnight at 4°C. Membranes were subsequently washed three times in Tris-buffered saline with 0.1% Tween 20 (TBST) and incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG (1:500; cat. no. 10285–1-AP; ProteinTech Group, Inc., Chicago, IL, USA) for 1 h at room temperature. Following three washes with TBST, the protein bands were visualized by using an Enhanced Chemiluminescence kit (GE Healthcare Life Sciences) and exposed to X-ray film. GAPDH was used as internal reference and the ImageJ (version 1.48; National Institutes of Health, Bethesda, MD, USA) was used for densitometry.

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

hPDLCs (2 ml; 1×105/well) were seeded into 6-well plates in standard medium until 60–70% confluence was reached. The cells were treated with four different media (control, CGFe, TNF-α, or CGFe+TNF-α). The control group was incubated with osteogenic induction medium [α-MEM, l50 µg/ml ascorbic acid and 10 mM β-sodium glycerophosphate (Sigma-Aldrich; Merck KGaA)]. The experimental groups were treated with osteogenic induction medium plus CGFe, osteogenic induction medium plus TNF-α (10 ng/ml), or osteogenic induction medium plus CGFe+TNF-α (10 ng/ml). RNA was extracted and reverse transcribed into cDNA using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) after 4, 7 and 14 days. cDNA synthesis was performed with 1 µg of total RNA, random hexamer primers, 5X First-Strand Buffer, and dNTP Mix (10 mM each) using SuperScript™ II RT (cat. no. 18064014; Invitrogen; Thermo Fisher Scientific, Inc.). The temperature protocol for RT was: Denaturing RNA at 65°C for 5 min, then incubation at 25 °C for 10 min and 42°C for 60 min, followed by 70°C for 15 min.

A 10 µl volume reaction system was adopted for qPCR determination of the expression of the osteogenic genes OCN, RUNX2 and OSX using the SYBR® Premix Ex Taq™ II kit (Takara Bio, Inc., Otsu, Japan). qPCR primers were designed to span an intron so that only RNA-specific amplification was possible. PCR was performed with the following thermocycling conditions: 95°C for 3 min, followed by 40 cycles of 95°C for 3 sec and 60°C for 60 sec. Each sample was tested in triplicate, and fold differences in gene expression were calculated using the 2−∆∆Cq method (24) with normalization to β-actin. The primer sequences are presented in Table I.

Table I.

Primers used for quantitative polymerase chain reaction analysis.

Table I.

Primers used for quantitative polymerase chain reaction analysis.

Primers

GeneForward (5′-3′)Reverse (5′-3′)PCR products (bp)
β-actin AGAAAATCTGGCACCACACC GGGGTGTTGAAGGTCTAAA139
Osteocalcin GGCGCTACCTGTATCAATGG TCAGCCAACTCGTCACAGTC106
RUNX2 CACCATGTCAGCAAAACTTCTT TCACGTCGCTCATTTTGC96
Osterix TGCTTGAGGAGGAAGTTCAC AGGTCACTGCCCACAGAGTA148

[i] RUNX2, runt-related transcription factor 2; bp, base pairs.

Statistical analysis

hPDLC proliferation and ALP activity assays were analyzed by one-way analysis of variance (ANOVA) followed by Tukey's post-hoc test. Western blotting and Alizarin Red staining assay data were analyzed using two-way ANOVA, followed by Bonferroni's post-hoc comparisons test for independent samples. SPSS 17.0 (SPSS, Inc., Chicago, IL, USA) was used to perform statistical analysis. P<0.05 was considered to indicate a statistically significant difference. All data were expressed as the mean ± standard deviation, and experiments were performed in triplicate.

Results

Characterization of hPDLCs

hPDLCs grew out from the tissue explant after 7 and 10 days of culture (Fig. 1A). Spindle shapes were observed, and a number of cells were distributed in a circinate pattern with rapid proliferation (Fig. 1B). The CGFe obtained was a yellowish clear fluid, and each 50 ml blood sample produced 4 ml of CGFe. The cells were vimentin positive (Fig. 1C) and keratin negative (Fig. 1D) according to immunochemistry staining, indicating that these primary cells were of mesenchymal origin.

CGFe increases hPDLC proliferation

The proliferation of hPDLCs was measured with CCK-8 assays (Fig. 2). After 24, 48 and 72 h, CGFe significantly enhanced the proliferation of hPDLCs compared with the control group (P<0.01), while TNF-α markedly inhibited the proliferation of hPDLCs (for TNF-α vs. control, P<0.05 at 24 and 48 h; P<0.01 at 72 h). In addition, the proliferation rate of the CGFe group was significantly higher than that of the CGFe+TNF-α group, and the proliferation of the CGFe group exceeded the TNF-α group substantially (for TNF-α vs. CGFe+TNF-α, P<0.05 at 24 h; P<0.01 at 48 and 72 h).

CGFe increases ALP activity

After 7 and 14 days of culture, the hPDLCs cultured in the CGFe group showed the highest levels of ALP activity compared with the other experimental groups (Fig. 3). Treatment with TNF-α significantly inhibited the ALP activity of hPDLCs, as shown by comparison of the TNF-α treatment group with the CGFe and TNF-α+CGFe groups, and the difference between these groups was significant (P<0.01) at both time-points. As expected, the ALP activity in all groups progressively increased over time.

Alizarin Red S staining and semi-quantification of mineralized matrix nodules

In order to detect the formation of mineralized matrix nodules, Alizarin Red S staining was performed (Fig. 4A-D). After 21 days of osteogenic induction, mineralized nodules were observed with an inverted phase-contrast microscope, and the number of nodules was notably higher in the CGFe group (Fig. 4C), compared with the control (Fig. 4A) or CGFe+TNF-α (Fig. 4D) groups. The TNF-α group (Fig. 4B) formed the fewest mineralized nodules.

The mineralized matrix nodules were quantified after 21 days of induction (Fig. 4E). The absorbance values at 570 nm revealed that extracellular calcium deposition in the CGFe group was significantly higher than the control or CGFe+TNF-α groups (P<0.01).

CGFe increases osteogenic-associated gene and protein expression

The gene expression of the RUNX2, OSX and OCN was determined after 4, 7 and 14 days of osteogenic induction (Fig. 5). It was observed that on days 4, 7 and 14, the expression of RUNX2 and OSX in the TNF-α (10 ng/ml) groups was decreased compared with the control group (P<0.01; excluding OSX expression on day 7, P<0.05), and the expression of these two genes in the CGFe+TNF-α group showed no significant increase compared with the control group. By contrast, on days 4, 7, and 14, expression of the RUNX2 and OSX genes in the CGFe group was significantly increased, compared with the control group (P<0.01; Fig. 5A and B). The expression of the downstream OCN gene was not significantly different between groups on day 4. By day 7, the CGFe group began to surpass the control group (P<0.01). After 14 days of culture, OCN expression in the TNF-α group was lower than that of the control group (P<0.01; Fig. 5C).

Western blotting was performed to examine the effects of CGFe on hPDLC differentiation. hPDLCs were cultured in each medium for 14 days. As shown in Fig. 6A, compared with the control group, the protein expression of RUNX2 was upregulated in the CGFe group, and downregulated in the CGFe+TNF-α and TNF-α groups (P<0.01; Fig. 6B). A similar trend was observed for OSX expression. Although the protein level of OSX was upregulated in the CGFe+TNF-α group, there was no significant difference compared with the control group (P>0.05; Fig. 6B).

Discussion

CGFe can be obtained from the venous blood of patients and thus is convenient and economical to prepare. CGFe contains high concentrations of growth factors and provokes no immune reaction (810,22,25). According to previous studies (48), the CGFe obtained from different patients in the present study contained different inventories of components, yet the major components were the same. These included epidermal growth factor, platelet-derived growth factor, transforming growth factor (TGF-β), vascular endothelium growth factor, insulin-like growth factor, bone morphogenetic protein (BMP), and fibroblast growth factor (48). In the last decade, CGFe has been widely used in the reconstruction of bone tissue in the surgical field (8,9). These growth factors have functions in accelerating the revascularization of injured tissues and inducing the migration, proliferation, and differentiation of fibroblasts and osteoblasts (810,25).

TNF-α is a common and crucial inflammatory factor in the development of periodontal inflammation and alveolar bone resorption (20). It has been established that TNF-α significantly inhibits the proliferation of hPDLCs and mesenchymal stem cells (5,26,27). In addition, activation of the nuclear factor (NF)-κB signaling pathway inhibits the expression of RUNX2, which is a downstream transcription factor induced by the exogenous BMP2-mothers against decapentaplegic homolog (Smad) signaling pathway controlling osteoblast differentiation, thus inhibiting the formation of bone (28).

In the present study, TNF-α (10 ng/ml) was used to construct an inflammatory microenvironment. It was demonstrated TNF-α reduced ALP activity, mineralization ability and the expression of RUNX2 and OSX, compared with the control group. This result was concordant with the findings of Yang et al (18), which revealed that the bone formation efficiency of gingival mesenchymal stem cells and periodontal ligament stem cells decreased under TNF-α-induced inflammatory conditions.

Previous studies have shown that RUNX2 and OSX are two essential transcription factors in the osteogenic pathway (29,30). In particular, RUNX2 regulates cell osteogenic differentiation and serves a central role in multiple osteogenic signaling pathways (29). OSX is an osteogenic-specific transcription factor with a zinc finger structure. A previous study revealed that no bone formation occurs in mice lacking the OSX gene (30). The level of ALP activity reflects the tendency of cells to transform in the osteogenic direction midway during the osteoproductive process, while OCN is a late marker of osteogenesis. OCN is the most abundant non-procollagen protein in bone tissue and promotes osteogenic differentiation by combining with minerals (31,32). Positive Alizarin Red S staining of the mineralized nodules formed indicates the expression of OCN and osteogenesis at a later stage (31,32).

In the present study, CGFe was added to cell cultures to observe the osteogenic efficiency of hPDLCs, by detecting the expression of RUNX2, OSX and OCN genes, as well as ALP activity and mineralized nodule formation in each group of hPDLCs. The formation of mineralized nodules was used to determine the osteogenic capacity of each group of hPDLCs. It was observed that even in the presence of inflammatory cytokine stimulation, hPDLCs cultured with CGFe still exhibited osteogenic differentiation and mineralization ability. By comparing the assay and imaging results of the CGFe group with the CGFe+TNF-α group, it was demonstrated that ALP activity in the CGFe group was stronger than that of the CGFe+TNF-α group, and the formation of mineralized nodules was notable in both groups, although the CGFe group was stained more prominently. In addition, ALP activity in the CGFe+TNF-α group was significantly higher than the TNF-α group, and the CGFe+TNF-α group had deep mineralized staining with a large area and high density, while no mineralized nodule formation was observed in the TNF-α group.

The western blotting results of the present study revealed that RUNX2 and OSX expression was higher in the CGFe group, compared with the CGFe+TNF-α group, indicating that TNF-α may play an inhibitory role upstream of protein translation. By contrast, growth factors contained in the CGFe may antagonize the inhibitory effect of TNF-α on cell proliferation. For example, TGF-β1 induces the phosphorylation of Smad2/Smad3, which upregulates RUNX2 transcription (29), thereby impeding TNF-α activation of the NF-κB signaling pathway, which would result in RUNX2 inhibition.

In conclusion, CGFe not only has an osteogenic effect on hPDLCs in a normal culture, but also promotes hPDLC osteogenesis in a TNF-α-induced inflammatory microenvironment. In the present work, a single inflammatory factor, TNF-α, was used to mimic the microenvironment of periodontal disease, and this imposed certain limitations. The enhancing effect of CGFe on osteogenic differentiation of hPDLCs stimulated by other inflammatory factors will be examined in follow-up experiments. To study the effects of CGFe in the treatment of alveolar bone defects, in vivo should be performed. In addition, the exact mechanism of how CGFe enhanced the proliferation and osteogenic differentiation of hPDLCs should be determined.

Acknowledgements

Not applicable.

Funding

This work was supported by a grant from the People's Hospital of Longhua (Shenzhen, China) and the Health and Family Planning Commission of Shenzhen Municipality (grant no. SZFZ2018035).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XL, HY and BW conceived and designed the study. XL, YZ, and HL performed cell culture, immunostaining and proliferation analysis. XL and YZ performed the experimental procedures of osteogenic differentiation induction, RT-qPCR, and western blotting. HY, ZZ and ZY provided reagents and interpreted the data. XL, HY and BW performed data analysis and wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study was approved by the Ethics Committee of the Jilin University Health Science Center (Changchun, China). Written consent was obtained prior to experimentation.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Alsaadi G, Quirynen M, Komárek A and Van Steenberghe D: Impact of local and systemic factors on the incidence of oral implant failures, up to abutment connection. J Clin Periodontol. 34:610–617. 2010. View Article : Google Scholar

2 

Del Fabbro M, Boggian C and Taschieri S: Immediate implant placement into fresh extraction sites with chronic periapical pathologic features combined with plasma rich in growth factors: Preliminary results of single-cohort study. J Oral Maxillofac Surg. 67:2476–2484. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Qiao J, Duan J, Zhang Y, Chu Y and Sun C: The effect of concentrated growth factors in the treatment of periodontal intrabony defects. Future Sci OA. 2:FS1362016. View Article : Google Scholar : PubMed/NCBI

4 

Jang SJ, Kim JD and Cha SS: Platelet-rich plasma (PRP) injections as an effective treatment for early osteoarthritis. Eur J Orthop Surg Traumatol. 23:573–580. 2013. View Article : Google Scholar : PubMed/NCBI

5 

He L, Lin Y, Hu X, Zhang Y and Wu H: A comparative study of platelet-rich fibrin (PRF) and platelet-rich plasma (PRP) on the effect of proliferation and differentiation of rat osteoblasts in vitro. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 108:707–713. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Suba Z, Takács D, Gyulai-Gaál S and Kovács K: Facilitation of beta-tricalcium phosphate-induced alveolar bone regeneration by platelet rich plasma in beagle dogs: A histologic and histomorphometric study. Int J Oral Maxillofac Implants. 19:832–838. 2004.PubMed/NCBI

7 

Fennis JP, Stoelinga PJ and Jansen JA: Mandibular reconstruction: A histologic and histomorphomrtric study on the use of autogenous scaffolds, particulate cortico-cancellous bone grafts and platelet rich plasma in goats. Int J Oral Maxillofac Surg. 33:48–55. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Kim TH, Kim SH, Sándor GK and Kim YD: Comparison of platelet-rich plasma (PRP), platelet-rich fibrin (PRF), and concentrated growth factor (CGF) in rabbit-skull defect healing. Arch Oral Biol. 59:550–558. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Park HC, Kim SG, Oh JS, You JS, Kim JS, Lim SC, Jeong MA, Kim JS, Jung C, Kwon YS and Ji H: Early bone formation at a femur defect using CGF and PRF grafts in adult dogs: A comparative study. Implant Dent. 25:387–393. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Rodella LF, Favero G, Boninsegna R, Buffoli B, Labanca M, Scarì G, Sacco L, Batani T and Rezzani R: Growth factors, CD34 positive cells, and fibrin network analysis in concentrated growth factors fraction. Microsc Res Tech. 74:772–777. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Li XJ, Yang HX, Zhang ZJ, Yan ZH, Lv HL, Zhang Y and Wu B: Platelet-rich fibrin exudate promotes the proliferation and osteogenic differentiation of human periodontal ligament cells in vitro. Mol Med Rep. 18:4477–4485. 2018.PubMed/NCBI

12 

Dohan DM, Choukroun J, Diss A, Dohan SL, Dohan AJ, Mouhyi J and Gogly B: Platelet-rich fibrin (PRF): A second generation platelet concentrate. Part I: Technological concepts and evolution. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 101:e37–e44. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Tavassoli-Hojjati S, Sattari M, Ghasemi T, Ahmadi R and Mashayekhi A: Effect of platelet-rich plasma concentrations on the proliferation of periodontal cells: An in vitro study. Eur J Dent. 10:469–474. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Dohan DM, Choukroun J, Diss A, Dohan SL, Dohan AJ, Mouhyi J and Gogly B: Platelet-rich fibrin (PRF): A second-generation platelet concentrate. Part III: Leucocyte activation: A new feature for platelet concentrates? Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 101:e51–e55. 2006. View Article : Google Scholar

15 

Li B and Wang Y: Contour changes in human alveolar bone following tooth extraction of the maxillary central incisor. J Zhejiang Univ Sci B. 15:1064–1071. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Lacey DC, Simmons PJ, Graves SE and Hamilton JA: Proinflammatory cytokines inhibit osteogenic differentiation from stem cells: Implications for bone repair during inflammation. Osteoarthritis Cartilage. 17:735–742. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Yu B and Wang Z: Effect of concentrated growth factors on beagle periodontal ligament stem cells in vitro. Mol Med Rep. 9:235–242. 2014. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

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

20 

Li W, Yu B, Li M, Sun D, Hu Y, Zhao M, Cui CB and Hou S: NEMO-binding domain peptide promotes osteoblast differentiation impaired by tumor necrosis factor alpha. Biochem Biophys Res Commun. 391:1228–1233. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Sacco L: Lecture: International Academy of Implant Prosthesis and Osteoconnection. Lecture. 12:42006.

22 

Sohn DS, Heo JU, Kwak DH, Kim DE, Kim JM, Moon JW, Lee JH and Park IS: Bone regeneration in the maxillary sinus using an autologous fibrin-rich block with concentrated growth factors alone. Implant Dent. 20:389–395. 2011.PubMed/NCBI

23 

Yamaguchi A: Application of BMP to bone repair. Clin Calcium. 17:263–369. 2007.(In Japanese). PubMed/NCBI

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Honda H, Tamai N, Naka N, Yoshikawa H and Myoui A: Bone tissue engineering with bone marrow-derived stromal cells integrated with concentrated growth factor in Rattus norvegicus calvaria defect model. J Artif Organs. 16:305–315. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Choukroun J, Diss A, Simonpieri A, Girard MO, Schoeffler C, Dohan SL, Dohan AJ, Mouhyi J and Dohan DM: Platelet-rich fibrin (PRF): A second-generation platelet concentrate. Part V: Histologic evaluations of PRF effects on bone allograft maturation in sinus lift. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 101:299–303. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Kang YH, Jeon SH, Park JY, Chung JH, Choung YH, Choung HW, Kim ES and Choung PH: Platelet-rich fibrin is a Bioscaffold and reservoir of growth factors for tissue regeneration. Tissue Eng Part A. 17:349–359. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Clipet F, Tricot S, Alno N, Massot M, Solhi H, Cathelineau G, Perez F, De Mello G and Pellen-Mussi P: In vitro effects of Choukroun's platelet-rich fibrin conditioned medium on 3 different cell lines implicated in dental implantology. Implant Dent. 21:51–56. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Franceschi RT and Xiao G: Regulation of the osteoblast-specific transcription factor, Runx2: Responsiveness to multiple signal transduction pathways. J Cell Biochem. 88:446–454. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR and de Crombrugghe B: The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 108:17–29. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Piek E, Ju WJ, Heeyer J, Escalante-Alcalde D, Stewart CL, Weinstein M, Deng C, Kucherlapati R, Bottinger EP and Roberts AB: Functional characterization of transforming growth factor beta signaling in Smad2-and Smad3-deficient fibroblasts. J Biol Chem. 276:19945–19953. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Glowacki J, Rey C, Glimcher MJ, Cox KA and Lian J: A role for osteocalcin in osteoclast differentiation. J Cell Biochem. 45:292–302. 1991. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 19 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
Li X, Yang H, Zhang Z, Yan Z, Lv H, Zhang Y and Wu B: Concentrated growth factor exudate enhances the proliferation of human periodontal ligament cells in the presence of TNF‑α. Mol Med Rep 19: 943-950, 2019
APA
Li, X., Yang, H., Zhang, Z., Yan, Z., Lv, H., Zhang, Y., & Wu, B. (2019). Concentrated growth factor exudate enhances the proliferation of human periodontal ligament cells in the presence of TNF‑α. Molecular Medicine Reports, 19, 943-950. https://doi.org/10.3892/mmr.2018.9714
MLA
Li, X., Yang, H., Zhang, Z., Yan, Z., Lv, H., Zhang, Y., Wu, B."Concentrated growth factor exudate enhances the proliferation of human periodontal ligament cells in the presence of TNF‑α". Molecular Medicine Reports 19.2 (2019): 943-950.
Chicago
Li, X., Yang, H., Zhang, Z., Yan, Z., Lv, H., Zhang, Y., Wu, B."Concentrated growth factor exudate enhances the proliferation of human periodontal ligament cells in the presence of TNF‑α". Molecular Medicine Reports 19, no. 2 (2019): 943-950. https://doi.org/10.3892/mmr.2018.9714