Differentiation 88 (2014) 97–105

Contents lists available at ScienceDirect

Differentiation journal homepage: www.elsevier.com/locate/diff

The difference on the osteogenic differentiation between periodontal ligament stem cells and bone marrow mesenchymal stem cells under inflammatory microenviroments Jing Zhang a,1, Zhi-Gang Li b,1, Ya-Meng Si a, Bin Chen a, Jian Meng a,n a b

Department of Stomatology, The Affiliated School of Clinical Medicine of Xuzhou Medical College, Xuzhou Central Hospital, Xuzhou, China Department of Urology, The Affiliated School of Clinical Medicine of Xuzhou Medical College, Xuzhou Central Hospital, Xuzhou, China

art ic l e i nf o

a b s t r a c t

Article history: Received 29 April 2014 Received in revised form 26 September 2014 Accepted 29 October 2014 Available online 10 December 2014

Periodontitis is a major cause of tooth loss in adults and periodontal ligament stem cells (PDLSCs) is the most favorable candidate for the reconstruction of tissues destroyed by periodontal diseases. However, pathological alterations caused by inflammatory insults might impact the regenerative capacities of these cells. Bone-marrow-derived human mesenchymal stem cells (hBMSCs) would accelerate alveolar bone regeneration by transplantation, compared to PDLSCs. Therefore, a better understanding of the osteogenic differentiation between PDLSCs and BMSCs in inflammatory microenviroments is therefore warranted. In this study, human PDLSCs were investigated for their stem cell characteristics via analysis of cell surface marker expression, colony forming unit efficiency, osteogenic differentiation and adipogenic differentiation, and compared to BMSCs. To determine the impact of both inflammation and the NF-κβ signal pathway on osteogenic differentiation, cells were challenged with TNF-α under osteogenic induction conditions and investigated for mineralization, alkaline phosphatase (ALP) activity, cell proliferation and relative genes expression. Results showed that PDLSCs exhibit weaker mineralization and ALP activity compared to BMSCs. TNF-α inhibited genes expression of osteogenic differentiation in PDLSCs, while, it stimulates gene expressions (BSP and Runx2) in BMSCs. Enhanced NF-κβ activity in PDLSCs decreases expression of Runx2 but it does not impede the osteogenic differentiation of BMSCs. Taken together, these results may suggest that the BMSCs owned the stronger immunomodulation in local microenvironment via anti-inflammatory functions, compared to PDLSCs. & 2014 International Society of Differentiation. Published by Elsevier B.V. All rights reserved.

Keywords: Periodontal ligament stem cells Bone marrow mesenchymal stem cells Inflammation Osteogenic differentiation

1. Introduction Periodontitis is a chronic infectious disease that leads to a progressive destruction of periodontal tissue. This disease is highly prevalent and can affect up to 90% of the worldwide population and it is also a major cause of tooth loss in adults (Pihlstrom et al., 2005; Chen et al., 2012; Park et al., 2011). Improving the regeneration of periodontal tissue has proven effective in the treatment of periodontitis. Human periodontal ligament (PDL) contains a novel population of multipotent stem cells that have the capacity to develop into cells with diverse phenotypes and therefore provide a unique reservoir of stem cells (Mrozik et al., 2010). In addition, periodontal ligament

n

Corresponding author. Tel.: þ 86 051683956489. E-mail address: [email protected] (J. Meng). 1 Both the authors contributed equally to this work.

stem cells (PDLSCs) have been shown to form an ectopic cementum/ ligament-like complex when transplanted in nude mice (Seo et al., 2004). Therefore, tissue regeneration mediated by human PDLSCs has the potential for use as a practical cell-based treatment for periodontal diseases (Liu et al., 2008; Tamaki et al., 2013). However, access to the periodontal ligament requires removal of teeth and the number of recoverable PDLSCs is limited due to their rarity and the small sample size. In contrast, bone-marrow-derived human mesenchymal stem cells (hBMSCs) can be harvested in much larger numbers and relative ease of acquisition (Kassem and Abdallah, 2008; Pittenger et al., 1999; Owen and Friedenstein, 1988). These cells are able to differentiate along several committed phenotypes including osteogenic, chrondogenic, adipogenic, and neurogenic and lineages in response to stimulation by multiple environmental factors (Wu et al., 2014; Lin and Hankenson, 2011; Welter et al., 2013; Haynesworth et al., 1992). Studies revealed that periodontal ligament cells and bone marrow own many similar characterists (Kramer et al., 2004). Periodontal ligament stem cells also express

http://dx.doi.org/10.1016/j.diff.2014.10.001 Join the International Society for Differentiation (www.isdifferentiation.org) 0301-4681/& 2014 International Society of Differentiation. Published by Elsevier B.V. All rights reserved.

98

J. Zhang et al. / Differentiation 88 (2014) 97–105

early mesenchymal stem cell surface markers (Menicanin et al., 2010). In addition, PDLSCs and BMSCs expressed the same surface molecules markers of bone. These results demonstrate BMSCs’ potency to develop periodontal ligament characteristics and suggest that the cells may have the potential to form other periodontal tissues. This is of potential significant since tissue regeneration is often needed in areas of an inflammatory reaction. Inflammatory processes in periodontal tissues seem to be modulated by resident PDLSCs. The features of BMSCs may differ from PDLSCs. Some researches discovered BMSCs would accelerate periodontal tissue regeneration (Chung et al., 2012; Yang et al., 2010; Kim et al., 2009). However, the differences in regulating osteogenic differentiation of BMSCs and PDLSCs in an inflammatory microenvironment remains to be elucidated. Tumor necrosis factor-α (TNF-α) is a proinflammatory cytokine released by macrophages is known for its substantial role in periodontitis mediated bone loss (Boyce et al., 2009). Elevated level of tumor necrosis factor-α (TNF-α) was confirmed to be associated with the severity of periodontal disease (Kornman et al., 1997; Soga et al., 2003; Zhang et al., 2013) and immune response (Teles et al., 2009). The transcription factor family, nuclear factor κB (NF-κB), is considered a central culprit in the pathogenesis of osteolysis in inflammatory diseases, including periodontitis, rheumatoid arthritis, lowgrade systemic inflammation, Paget’s disease of bone (PDB), and other bacterial infections (Xu et al., 2009). NF-kappaB signalling pathways are strictly regulated to maintain bone homeostasis by cytokines such as RANKL, TNF-alpha and IL-1, which differentially regulate classical and/or alternative NF-kappaB pathways in osteoclastic cells. Numerous reports have demonstrated that TNF-α activates nuclear factor (NF)-kappaB, resulting in the upregulation of several genes that regulate inflammation, proliferation, and apoptosis (Moe et al., 2014). In this study, we aimed to analyze the effects of TNF-α on the osteogenic differentiation of hPDLSCs and hBMSCs and to determine the differential expression of target genes that are related to osteogenic differentiation. In addition, we also investigated the role of NF-κβ signal pathway on the differentiation of PDLSCs and BMSCs.

2. Materials and methods 2.1. Cell culture Human PDLSCs were isolated and cultured as previously described (Yang et al., 2009). Briefly, normal premolar teeth (n¼6) extracted for orthodontic treatment from 3 individuals (at 18–22 years of age) were collected after obtaining writted informed consent. The study protocol was approved by the Xuzhou Medical College’s Ethics Committee. Both systemic and oral diseases were absent in all subjects. Periodontal ligament tissues were gently scraped from the surface of the middle part of the root, cut into 1 mm3 cubes and placed into six-well culture dishes. The tissues were grown in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% FBS, 0.292 mg/ml glutamine, 100 U/ml penicillin G, 100 mg/ml streptomycin. Cells were maintained at 37 1C in a humidified atmosphere with 5% CO2 in air and cultures obtained from each donor were processed separately. To obtain homogeneous populations of PDLSC, single cellderived colony cultures were obtained using the limiting dilution technique as previously described (Huo et al., 2010). After 2–3 weeks original culture, the single cell-derived clones were then harvested and cells were subcultured at approximately 80–90% confluence with trypsin/EDTA. Human BMSCs (n ¼4, aged from 18 to 25) were established from bone marrow samples with informed consent of the donors

and following the guidelines of the hospital’s Ethics Committee. Cells were isolated by Ficoll density gradient centrifugation, suspended in regular culture medium consisting of DMEM with 10% FBS, 0.292 mg/ml glutamine, 100 U/ml penicillin G, 100 mg/ml streptomycin. Cells were maintained at 37 1C in a humidified atmosphere with 5% CO2 in air. After 24 h the culture supernatants were replaced by fresh medium to discard the nonadherent cells. Cells were subcultured at approximately 80–90% confluence with trypsin/EDTA. 2.2. Flow cytometry analysis For identification of MSC phenotype, approximately 5  105 BMSCs and PDLSCs were washed in phosphate buffered saline (PBS) and then incubated with the following mouse anti-human monoclonal antibodies: fluorescein isothiocyanate (FITC)-conjugated CD14, CD90 (eBioscience, San Diego, CA), CD34 (Biolegend, USA), phycoerythrin (PE)-labelled CD31, CD45 (eBioscience, San Diego, CA) and CD146 (Biolegend, USA) for 30 min at 4 1C, respectively. The cell suspension was then washed twice with PBS and analyzed on a Beckman Coulter Epics XL (Beckman Coulter, Fullerton, CA). 2.3. Colony-forming assay To assesss colony forming efficiency, 1  103 PDLSCs and BMSCs were seeded into 100-mm dish and maintained for 14 days, respectively. Then they were fixed with 4% formalin and stained with 1% toluidine blue. The cells were washed twice with distilled water, and the number of colonies was counted. Aggregates of over 50 cells were counted as a colony under the microscope. Experiments were performed in triplicate. 2.4. Osteogenic and adipogenic differentiation Induction of calcification and adipogenesis were as previously reported (Platt and El-Sohemy, 2009). HBMSCs and hPDLSCs at 3rd passage were plated into 6-well culture dishes at a concentration of 1  105 cells/well. Cultures were allowed to reach 80% confluence before differentiation was initiated. Then normal DMEM medium was removed, and replaced with the osteogenic medium (OM, DMEM supplemented with 10% FBS, 50 mg/ml of ascorbic acid, 10 mM of sodium β-glycerophosphate, and 100 nM of dexamethasone) or the adipogenic medium (DMEM supplemented with 10% FBS, 0.5 mM of methylisobutylxanthine, 0.5 mM of hydrocortisone, 60 mM indomethacin, and 10 mg/ml insulin). Cells were maintained with the fresh differentiation medium every 3 or 4 days for 4 weeks. All experiments were performed in triplicate. 2.5. Alkaline phosphatase (ALP) activity assay For quantitative analysis of alkaline phosphatase (ALP) activity, single-cell suspensions of PDLSCs and BMSCs at 3rd passage were seeded at a density of 3  103 cells/well into 96-well plates and cultured in DMEM supplemented with 10% FBS. To investigate the effect of cytokine, cells were treated from day 2 with human TNFα (0.01–10 ng/ml) (Peprotech, USA) throughout the differentiation assay (DMEM supplemented with 2% FBS, 50 mg/ml of ascorbic acid, 10 mM of sodium β-glycerophosphate, and 100 nM of dexamethasone). Fresh cytokines added at every medium change. After 7 and 14 days in vitro culture, the ALP activity of each cell was detected with a commercially acailable assay kit (Zhongsheng Co, Beijing, China). In brief, cells were washed 3 times in PBS and incubated in Triton X-100 (2 ml/l in PBS) for overnight at 4 1C. One hundred microliters of p-nitrophenol phosphate substrate solutions was added to each well and the cells were incubated for 40 min at 37 1C. The addition of NaOH quenched the reaction, and

J. Zhang et al. / Differentiation 88 (2014) 97–105

99

the absorbance at 405 nm was read on a plate reader (Bio-Tek, USA). The experiment was performed in triplicate.

gene expression of β-actin was used as a reference gene expression in all applications. The assays were performed in triplicate.

2.6. Cell proliferation assays

2.9. Western blot analysis

The effect of TNF-α on cell viability was determined by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl-tetrazoliumbromide) (Sigma, St. Louis, MO) assay using Coster 96-well culture plates. Briefly, hBMSCs and hPDLSCs were plated at a density of 4  103 cells per well in DMEM medium and incubated overnight. The following day, cells were cultured as for the osteogenic differentiation assay (see above). Cells were maintained with the fresh differentiation medium every 3 days. At the indicated time points (1, 4, 7 days), 20 μl of the MTT reagent (final concentration of 5 mg/ml) was added into each well and the plates were incubated for 4 h in a humidified atmosphere (37 1C and 5% CO2). An aliquot (150 μl) of the solubilization solution (DMSO) was added to each well and plates were shaken for 10 min. The absorbance of the samples was measured at 490 nm using a microplate reader (Bio-Tek, USA). Each assay was performed in triplicate.

Cells were lysed in 1% n-octyl-p-D-glucopyranoside (OG) buffer (20 mM Tris–HCl (pH 8.0), 150 mM NaCl, 1% OG, 1 mM EDTA, 10 g/ ml leupeptin, 2 g/ml aprotinin, 1 mM PMSF). The total protein density was determined using bicinchoninic acid (BCA) protein assay kit (Pierce Biotechnology, Rockford, Illinois). Total cell lysates were subjected to 10% SDS-PAGE gels and then electrotransferred to polyvinylidene fluoride (PVDF) membranes (Bio-Rad, Richmond, CA). After blocking with 5% non-fat milk, the membranes were incubated overnight with primary antibodies and HRP-conjugated secondary antibodies (anti-mouse IgG or anti-rabbit IgG, Promega, USA) for 1 h. The membranes were washed and visualized by an enhanced chemiluminescence reagents (Pierce) according to the manufacturer’s protocol. The proteins were detected with specific antibodies against phospho-IKβ-α (monoclonal, Cell signaling technology), Runx2 (polyclonal, Millipore). All experiments were performed in triplicate.

2.7. In vitro mineralization assay To investigate the effect of cytokine on osteoblast differentiation, 1  104 PDLSCs and BMSCs per well were seeded onto a 24 well plate overnight at 37 1C in 5% CO2, respectively. The cells were washed with phosphate-buffered saline (PBS) before medium was changed into osteogenic medium (OM) with 2% FBS (see above) and cultured for 4 weeks at 37 1C in 5% CO2, with the medium being replaced every 3–4 days. Cells were also treated with human TNF-α (0.01–10 ng/ml) throughout the differentiation assay, and fresh cytokines added at every medium change. Alkaline phosphatase (ALP) staining was performed as previous described (Gronthos et al., 2000). Matrix mineralization was evaluated by Alizarin Red staining. All experiments were performed in triplicate. 2.8. RNA-isolation and real-time PCR Before RNA isolation, BMSCs and PDLSCs were seeded at 5000 cells/ cm in 25 cm flasks. Cultures were allowed to reach 80% confluence before differentiation was initiated. Then normal DMEM medium was removed, and replaced with the osteogenic medium with 2% FBS and the medium being replaced every 3 days. Cells were also treated with human TNFα (10 ng/ml) (Peprotech, USA) throughout the differentiation assay, and fresh cytokines added at every medium change. Total RNA was extracted using TRIZOL Reagent (Invitrogen) and cDNA was prepared using a Superscript II first-strand cDNA synthesis kit (Invitrogen Life Technologies) according to manufacturers’ instructions at the indicated time points (24 h, 14 days). A SYBER Premix Taq™ II Kit (Takara) was used for qRT-PCR. Relative transcript levels were measured by using the ABI PRISM 7500 sequence detection system (Applied Biosystems, Foster City, CA, USA). The expression of the osteoblast differentiation genes alkaline phosphatase (ALP), type I collagen (Col-1), bone sialoprotein (BSP), osteocalcin (OCN), the bonespecific transcription factor Runx2 and IKβ-α in PDLSCs and BMSCs was measured using qRT-PCR. Reverse transcription-polymerase chain reaction (RT-PCR) was carried out including the following primers: ALP: forward, 50 -GGACCATTCCCACGTCTTCAC-30 , reverse, 50 -CCTTGT AGCCAGGCCCATTG-30 ; Col-I: forward, 50 -CCAGAAGAACTGGTACATCAGCAA-30 , reverse, 50 -CGCCATACTCGAACTGGAATC-30 ; BSP: forward, 50 ATACAGGGTTAGCTGCAATC-30 , reverse, 50 -TCCATTGTCTCCTCCGC-30 ; OCN: forward, 50 -AGCAAAGGTGCAGCCTTTGT-30 , reverse, 50 -GCGCCTGGGTCTCTTCACT-30 ; Runx2: forward, 50 -CCCGTGGCCTTCAAGGT-30 , reverse, 50 -CGTTACCC GCCATGACAGTA-30 ; IKβ-α: forward, 50 -GCAGGACTGAGTCAGGACTCCCAC -30 , reverse, 50 GCCTTCCTCAACTTCCAGAACAACC-30 ; β-actin: forward, 50 -CAGGCTGTGCTATCCCTGTA-30 , reverse, 50 -CAT ACCCCTCGTAGATGGGC-30 . The

2.10. Statistical analysis All data are presented as the mean7SD from three independent experiments and analyzed by two-tailed Student’s t-test using SPSS software. P values less than 0.05 were considered significant.

3. Results 3.1. Culture and identification of PDLSCs and BMSCs Human PDLSCs and BMSCs showed the features of spindle shape (Fig. 1A and B) and had the ability to form adherent clonogenic cell clusters (Fig. 1C–F). The multi-differentiation potential of PDLSCs and BMSCs was determined. Under induction conditions for 4 weeks, both PDLSCs and BMSCs could formed distinct nodules as stained by Alizarin (Fig. 1G and H) and lipid droplets by Oil Red O staining in vitro (Fig. 1I and J). We used flow cytometric analysis to characterize BMSCs and PDLSCs by surface molecules. Both BMSCs and PDLSCs showed the characteristic pattern of mesenchymal surface markers including CD90, CD146 and negatively expressed endothelial cell (CD31) and hematopoietic markers CD45, CD14, CD34 (Fig. 2). 3.2. Effect of TNF-α on osteogenesis of PDLSCs and BMSCs 3.2.1. Mineralization and ALP staining Osteogenic differentiation of the PDLSCs and BMSCs were carried out, in the absence and presence of TNF-α, for 4 weeks after which time alizarin red S and ALP staining were measured. Results revealed that the inflammatory cytokine TNF-α had different effects on the osteogenic differentiation of PDLSCs and BMSCs. An inhibitory effect of TNF-α on osteogenic differentiation of PDLSCs with increasing dose. However, BMSCs were more resistant to the inflammatory cytokine compared to PDLSCs in terms of a stronger ALP activity and alizarin red staining (Fig. 3). 3.2.2. Alkaline phosphatase (ALP) activity Because alkaline phosphatase has been implicated as a marker of osteogenic differentiation, the ALP activity of PDLSCs and BMSCs were measured as shown in Fig. 4. We found that the ALP activity of PDLSCs and BMSCs treated with TNF-α was decreased increasingly on day 7. On day 14, TNF-α treatment obviously inhibited the ALP activity of PDLSCs at the maximum concentration; interestingly, in

100

J. Zhang et al. / Differentiation 88 (2014) 97–105

Fig. 1. Morphological characteristic, colony-forming and osteogenic differentiation and adipogenic differentiation capability. PDLSCs (A) and BMSCs (B) showed a typical fibroblast-like spindle appearance. PDLSCs (C and D) and BMSCs (E and F) were capable of forming a single-colony cluster at low seeding density after 2 weeks culture and exhibited typical fibroblastic morphology. Multilineage differentiation ability of PDLSCs and BMSCs (G–J). Alizarin Red Staining of PDLSCs (G) and BMSCs (H) under osteogenic medium. Oil Red O staining of PDLSCs (I) and BMSCs (J) under adipogenic induction. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

contrast, BMSCs, even at a maximum concentration of TNF-α (10 ng/ml), had a positive effect. 3.2.3. BMSCs and PDLSCs proliferation To investigate the effect of TNF-α on the proliferation of PDLSCs and BMSCs, various concentrations of TNF-α were added to the culture media at 1, 4, 7 days. The results showed that TNF-α exhibited inhibitory effects on cell proliferation, and BMSCs possessed a lower proliferative potential than PDLSCs (Fig. 5). 3.2.4. Osteoblast lineage gene expression The expression of genes involved in osteoblastic differentiation was determined by RT-PCR at day 14. The expression of the osteoblast-related genes ALP, COL-1, BSP, OCN and Runx2 is indicative of the osteogenic differentiation status of PDLSCs and BMSCs. Results revealed that TNF-α suppressed the mRNA levels of ALP, COL-1 and OCN for PDLSCs and BMSCs (Fig. 6A, B and D). The inflammatory cytokine TNF-α reduced mRNA expression of BSP and Runx2 for PDLSCs, but it significantly promoted the mRNA expression of BSP and Runx2 in BMSCs (Fig. 6C and E). The results revealed that the inflammatory cytokine TNF-α had different effects on the osteogenic differentiation of PDLSCs and BMSCs. 3.2.5. Expression characteristics of NF-κB signal in PDLSCs and BMSCs As shown in Fig. 7A, NF-κβ pathway gene (IKβ-α) was upregulated in PDLSCs and BMSCs after a 24 h stimulation. The expression level of Runx2 in PDLSCs was decreased, while, a slight increase in BMSCs was observed (Fig. 7A). Western blot analysis demonstrated that TNF-α increased expression of phospho-IKB-α in PDLSCs and BMSCs. Compared to BMSCs, the protein expression levels of Runx2 decreased for PDLSCs (Fig. 7B).

4. Discussion Mesenchymal stem cells have been widely developed and investigated nowadays as clinically applicable cell sources for tissue engineering due to their ease of isolation, in vitro expansion, differentiation potential and suppress inflammation (Matsuoka et al., 2013; Deans and Moseley, 2000; Marion and Mao, 2006; Rodríguez et al., 2004). BMSCs have been identified to have potential to transdifferentiate into periodontal ligament cells and engraft into periodontal defects which accelerating periodontal tissue regeneration both in vivo and in vitro (Yang et al., 2010; Kim et al., 2009). At present, even though the effect of BMSCs and PDLSCs on inflammation has been extensively studied, the differences of using PDLSCs or BMSCs to repair periodontal defects in an inflammatory microenvironment is poorly understood. A number of inflammatory cytokines have been shown to be associated with periodontal pathogenesis. Among these, TNF-α and IL-1β are two major cytokines that lead to a negative role in the periodontal inflammatory process. In bone metabolism, TNF-α and IL-1β have been proved to promote bone loss by activating osteoclastogenesis and decrease bone mineral density by inhibiting osteoblastic differentiation and bone formation (Schett, 2011; Zhao et al., 2012). While, Hess et al. (2009) reported that TNFalpha promotes osteogenic differentiation of human mesenchymal stem cells and Glass et al. (2011) demonstrated that TNF-alpha promotes fracture repair by augmenting the recruitment and differentiation of muscle-derived stromal cells. Lin et al. (2010) also showed that the biphasic effects of interleukin-1beta on osteoblast differentiation in vitro. Recently, Li et al. (2014) revealed that lipopolysaccharide deteriorated the osteogenic differentiation of periodontal ligament stem cells through Toll-like receptor 4 mediated nuclear factor kappaB pathway, but not for bone marrow mesenchymal stem cells. Therefore, the roles of

J. Zhang et al. / Differentiation 88 (2014) 97–105

101

Fig. 2. Flow cytometry analysis of stem cell surface markers on PDLSCs and BMSCs. Cell surface markers related to mesenchymal (CD90, and CD146), endothelial cell (CD31), or hematopoietic stem cells (CD14, CD34 and CD45).

Fig. 3. Discriminative influence of TNF-α on osteogenic differentiation of PDLSCs and BMSCs. PDLSCs and BMSCs were cultured in osteogenic differentiation medium in the absence and presence of TNF-α for 4 weeks. Osteogenic differentiation was determined by alizarin red staining (A) and ALP staining (B). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

TNF-α, IL-1β and periodontal bacteria in the osteogenic differentiation of progenitor cells and bone metabolism were complicated and biphasic.

Fig. 4. The effect of TNF-α on osteogenic differentiation in PDLSCs and BMSCs. Osteogenic differentiation was determined by ALP activity with different concentration of TNF-α at day 7, 14. (A and B) The data are shown as mean 7 SD. nP o0.05, nn Po 0.01, n¼ 3. OM: osteogenic medium.

102

J. Zhang et al. / Differentiation 88 (2014) 97–105

Fig. 5. The effect of TNF-α on the proliferation of PDLSCs and BMSCs. PDLSCs and BMSCs were cultured in osteogenic differentiation medium with or without diverse concentration TNF-α at day 1, 4, 7. (A–C) The data are shown as mean 7 SD. nPo 0.05, nnP o0.01, n¼ 3. OM: osteogenic medium.

Studies using BMSCs and TNF-α from the different species or concentration have shown divergent effects (Gilbert et al., 2002, 2000; Lu et al., 2006). Lacey et al. (2009) demonstrated that TNF-α inhibited osteogenic differentiation of murine stem cells. In contrast to that data, Ding et al. (2009)demonstrated that TNF-α were able to stimulate TNAP activity and mineralization in hBMSCs. This showed that species physiology also affected the osteogenic marker results. The results of the present study showed PDLSCs and BMSCs, although of the same mesenchymal origin, possess diverse stem cell characteristics, especially under the inflammatory microenvironments. TNF-α strongly inhibited PDLSCs mineralization and ALP activity with a dose dependently. However, TNF-α stimulated the mineralization and ALP activity of BMSCs. Studies in many cell types have shown that proliferation and differentiation are inversely correlated processes (Golding et al., 1988). Alterations in the differentiation along a cell lineage may or may not be accompanied by changes in the rate of cell proliferation and both IL-1β and TNFα can influence this parameter in other cell types (Butler et al., 1988; Lacey et al., 2003). TNF-α can influence this parameter (Mountziaris et al., 2010). We therefore monitored the effects of TNF-α for PDLSCs and BMSCs during osteogenesis. Compared to PDLSCs, our results showed that TNF-α treatment evidently inhibited BMSCs proliferation. These findings also suggested an inverse link between proliferation and differentiation. The expression of the transcription factors, Runx2 has been shown to be necessary for osteoblast differentiation at a relatively early stage (Short et al., 2003s). And it regulates the expression of osteoblast marker genes such as ALP, Col-1, BSP and OCN (Nakashima et al., 2002). Therefore, inhibition of Runx2 activity appears, confirmed by decreased expression of ALP, Col-1, BSP and

OCN, which are dependent on Runx2 activity (Ducy et al., 1997). Huang et al. (2014) revealed that treatment with TNF-α/IL-1β inhibited BMP-2-induced alkaline phosphatase activity, calcium deposition, osteogenic transcriptional factor Runx2, and the expression of osteogenic markers in C2C12 and MC3T3-E1 cells. However, Lencel et al. (2011) reported that TNF-α stimulates alkaline phosphatase and mineralization through PPARγ inhibition in human osteoblasts. Ding et al. demonstrated both TNF-alpha and IL-1beta decreased RUNX2 expression and osteocalcin secretion in human mesenchymal stem cells, suggesting that RUNX2 was not involved in mineralization. In addition, cell-specific differences in the response to cytokines cannot be excluded. Our data indicated that the suppressive effect of TNF-α on the Runx2 lead to reduced genes expression for osteogenic differentiation in PDLSCs, while, it stimulated Runx2 expression of BMSCs at 14 days. ALP and Col-1 are widely used markers of osteoblasts and both are early marker of osteogenic differentiation (Liu et al., 2013). BSP is a major bone extracellular matrix sialoprotein whose expression appears concurrently with matrix deposition and therefore is used as medium-stage marker. OCN is a marker of the late stages of osteoblast differentiation and its production denotes the onset of matrix deposition. In our study, results showed a positive effect of TNF-α on Runx2 in BMSCs during osteogenic differentiation. The expression level of ALP, Col-1 and OCN were suppressed in BMSCs, but the expression of BSP were upregulated at 14 days. Specific osteoblast phenotype genes (OPN, Col1, OCN, and BSP) appeared during active osteoblastic differentiation. Previous researches demonstrated that the sequential expression of matrix protein genes (osteonectin, osteopontin and osteocalcin) were associated with

J. Zhang et al. / Differentiation 88 (2014) 97–105

103

Fig. 6. Discriminative influence of TNF-α on osteogenic differentiation of PDLSCs and BMSCs. The expressions of osteoblast lineage gene (ALP, Col-1, BSP, OCN and Runx2) were measured by real-time PCR at day 14 (A–E). The expression level of mRNA was normalized to β-actin. The data are shown as mean 7 SD. nPo 0.05, n¼ 3. OM: osteogenic medium.

extra-cellular matrix mineralization (Nakase et al., 1994). Another study showed that a sequential and cell type-restricted expression of matrix proteins taken place during the development of the mineralized tissues (Sommer et al., 1996). The expression of matrix protein genes was spatially and temporally controlled, in relation with the biological role of their cognate proteins in epithelial–mesenchymal interactions and mineralization (Bleicher et al., 1999). Our findings suggest that TNF-α stimulates osteogenic differentiation of BMSCs at 14 days, and that this effect may reveal sequential expression of osteoblast-related genes during mineralized culture. The molecular mechanisms governing specific cytokine modulation of BMSCs and PDLSCs differentiation are complex and involve many signal pathways. Wnt signaling has been shown as an important regulatory pathway in the osteogenic differentiation of mesenchymal stem cells (Kim et al., 2013). Wnt mediated signals that comprise two main molecular pathways, namely the β-catenindependent canonical and the β-catenin-independent noncanonical Wnt pathway. Liu et al. (2011) indicate that high levels of β-catenin signaling reduce osteogenic differentiation of PDLSCs in inflammatory microenvironments through inhibition of the noncanonical Wnt pathway. However, Chen et al. (2013) identified that NF-κβ signaling might be more important for the regulation of osteogenesis in PDLSCs from periodontitis compared with β-catenin. The NFκβ signaling pathway is long known to play an important role in inflammation and control of the immune system (Ghosh et al., 1998; Liang et al., 2004; Bonizzi and Karin, 2004). In most cell types, NF-κβ proteins are sequestered in the cytoplasm by the inhibitor IκB in an inactive form (Karin and Greten, 2005). Upon stimulation, Iκβ is phosphorylated by IKK and subsequently polyubiquinated, which

triggers its rapid degradation by proteasomes (Böcker et al., 2008). Consequently, NF-κβ proteins are released and translocate into the nucleus, where they activate the expression of target genes. Chang et al. (2009) revealed that high levels of TNF-α was especially found at sites of periodontitis and they are key regulators of the canonical NF-κβ pathway. Hess et al. (2009) observed that TNF-α increased BMP-2 expression in hMSCs through the NF-κβ signaling pathway in early osteogenic differentiation. To elucidate the role of NF-κβ in osteogenesis, we have analyzed its influence on osteogenic differentiation by real-time-PCR and Western blot analysis at 24 h after cytokine treatment. Our findings indicate that enhanced NF-κβ activity in PDLSCs decreases expression of Runx2 at the mRNA and protein levels, whereas increased NF-κβ signaling did not impede the osteogenic differentiation of BMSCs. In Fig. 7B, compared to control, Runx2 protein expression in PDLSCs decreased not much. We suggested that the expression level of protein was less sensitive than gene expression. Recently, investigation revealed that periodontal ligament stem cells had impaired immunomodulatory function after exposure to an inflammatory environment, which may lead to an imbalanced immune response and the acceleration of osteoclastogenesis and inflammation related bone loss (Liu et al., 2012). While, Papadopoulou et al. (2012) showed that conditioning of the recipient with bortezomib alters the disease microenvironment enabling bone marrow-derived MSC to modulate arthritis. These results indicated that the immunomodulatory properties may play an important role in mediating BMSCs differentiation in inflammatory microenvironment. A deeper understanding of these underlying mechanisms will facilitate the development of therapies for periodontitis on the basis of stem cell application.

104

J. Zhang et al. / Differentiation 88 (2014) 97–105

Fig. 7. Impact of NF-kβ pathway on osteogenic differentiation in PDLSCs and BMSCs. The expressions of IKβ-α and Runx2 were measured by real-time PCR and Western blot at 24 h (A and B). The expression levels of mRNA and protein were normalized to β-actin. The data are shown as mean 7 SD. nPo 0.05, n¼ 3. OM: osteogenic medium.

5. Conclusion Our experiments revealed that the PDLSCs exhibited weaker osteogenic differentiation than BMSCs after treatment with inflammatory cytokine. In our study, NF-κB signaling had not obvious effect on osteogenic differentiation of BMSCs. This indicated that there may be other mechanisms involved in the regulation of the osteogenic differentiation. Recently, researches demonstrated that mesenchymal stem cells had impaired immunomodulatory function after exposure to an inflammatory environment, which may lead to an imbalanced immune response and the acceleration of osteoclastogenesis and inflammation related bone loss. We speculated that the BMSCs might own the stronger immunomodulation in local microenvironment via anti-inflammatory functions, compared to PDLSCs. Therefore, it is required to conduct a further study to compare the immunomodulation and regenerative effects of PDLSCs and BMSCs in the future.

Conflict of interest The authors declare no conflict of interest. References Böcker, W., Docheva, D., Prall, W.C., Egea, V., Pappou, E., Rossmann, O., Popov., C., Mutschler, W., Ries, C., Schieker, M., 2008. IKK-2 is required for TNF-α-induced invasion and proliferation of human mesenchymal stem cells. J. Mol. Med. (Berlin) 86, 1183–1192.

Bonizzi, G., Karin, M., 2004. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 25, 280–288. Boyce, B.F., Li, P., Yao, Z., Zhang, Q., Badell, I.R., Schwarz, E.M., O_Teles, R.P., Likhari, V., Socransky, S.S., Haffajee, A.D., 2009. Salivary cytokine levels in subjects with chronic periodontitis and in periodontally healthy individuals: A crosssectional study. J. Periodontal. Res. 44, 411–417. Bleicher, F., Couble, ML., Farges, J.C., Couble, P., Magloire, H., 1999. Sequential expression of matrix protein genes in developing rat teeth. Matrix Biol. 18, 133–143. Butler, D.M., Piccoli, D.S., Hart, P.H., Hamilton, J.A., 1988. Stimulation of human synovial fibroblast DNA synthesis by recombinant human cytokines. J. Rheumatol. 15, 1463–1470. Chang, J., Wang, Z., Tang, E., Fan, Z., McCauley, L., Franceschi, R., Guan, K., Krebsbach, P.H., Wang, C.Y., 2009. Inhibition of osteoblastic bone formation by nuclear factorkappaβ. Nat. Med 15, 682–689. Chen, F.M., Sun, H.H., Lu, H., Yu, Q., 2012. Stem cell-delivery therapeutics for periodontal tissue regeneration. Biomaterials 33, 6320–6644. Chen, X., Hu, C., Wang, G., Li, L., Kong, X., Ding, Y., Jin, Y., 2013. Nuclear factor-κβ modulates osteogenesis of periodontal ligament stem cells through competition with β-catenin signaling in inflammatory microenvironments. Cell Death Dis 4, e510. Chung, V.H., Chen, A.Y., Jeng, L.B., Kwan, C.C., Cheng, S.H., Chang, S.C., 2012. Engineered autologous bone marrow mesenchymal stem cells: alternative to cleft alveolar bone graft surgery. J. Craniofac. Surg 23, 1558–1563. Deans, R.J., Moseley, A.B., 2000. Mesenchymal stem cells: biology and potential clinical uses. Exp. Hematol. 28, 875–884. Ding, J., Ghali, O., Lencel, P., Broux, O., Chauveau, C., Devedjian, J.C., Hardouin, P., Magne, D., 2009. TNF-α and IL-1β inhibit RUNX2 and collagen expression but increase alkaline phosphatase activity and mineralization in human mesenchymal stem cells. Life Sci. 84, 499–504. Ducy, P., Zhang, R., Geoffroy, V., Ridall, A.L., Karsenty, G., 1997. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 89, 747–754. Ghosh, S., May, M.J., Kopp, E.B., 1998. NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu. Rev. Immunol. 16, 225–260. Gilbert, L., He, X., Farmer, P., Rubin, J., Drissi, H., van Wijnen, A.J., Lian, J.B., Stein, G.S., Nanes, M.S., 2002. Expression of the osteoblast differentiation factor RUNX2 (Cbfa1/AML3/Pebp2alpha A) is inhibited by tumor necrosis factor-alpha. J. Biol. Chem. 277, 2695–7201.

J. Zhang et al. / Differentiation 88 (2014) 97–105

Glass, G.E., Chan, J.K., Freidin, A., Feldmann, M., Horwood, N.J., Nanchahal, J., 2011. TNF-alpha promotes fracture repair by augmenting the recruitment and differentiation of muscle-derived stromal cells. Proc. Natl. Acad. Sci. U.S.A 108, 1585–1590. Golding, B., Pillemer, S.R., Roussou, P., Peters, E.A., Tsokos, G.C., Ballow, J.E., Hoffman, T., 1988. Inverse relationship between proliferation and differentiation in a human TNP-specific B cell line. Cell cycle dependence of antibody secretion. J. Immunol. 141, 2564–2568. Gronthos, S., Mankani, M., Brahim, J., Robey, P.G., Shi, S., 2000. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. U.S.A 97, 13625–13630. Haynesworth, S.E., Goshima, J., Goldberg, V.M., Caplan, A.I., 1992. Characterization of cells with osteogenic potential from human marrow. Bone 13, 81–88. Hess, K., Ushmorov, A., Fiedler, J., Brenner, R.E., Wirth, T., 2009. TNFα promotes osteogenic differentiation of human mesenchymal stem cells by triggering the NF-κβ signaling pathway. Bone 45, 367–376. Huang, R.L., Yuan, Y., Tu, J., Zou, G.M., Li, Q., 2014. Opposing TNF-α/IL-1β- and BMP2-activated MAPK signaling pathways converge on Runx2 to regulate BMP-2induced osteoblastic differentiation. Cell Death Dis 5, e1187. Huo, N., Tang, L., Yang, Z., Qian, H., Wang, Y., Han, C., Gu, Z., Duan, Y., Jin, Y., 2010. Differentiation of dermal multipotent cells into odontogenic lineage induced by embryonic and neonatal tooth germ cell conditioned medium. Stem Cells Dev. 19, 93–104. Karin, M., Greten, F.R., 2005. NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol. 5, 749–759. Kassem, M., Abdallah, B.M., 2008. Human bone-marrow-derived mesenchymal stem cells: biological characteristics and potential role in therapy of degenerative diseases. Cell Tissue Res. 331, 157–163. Kim, J.H., Liu, X., Wang, J., Chen, X., Zhang, H., Kim, S.H., Cui, J., Li, R., Zhang, W., Kong, Y., Zhang, J., Shui, W., Lamplot, J., Rogers, M.R., Zhao, C., Wang, N., Rajan, P., Tomal, J., Statz, J., Wu, N., Luu, H.H., Haydon, R.C., He, T.C., 2013. Wnt signaling in bone formation and its therapeutic potential for bone diseases. Ther. Adv. Musculoskelet. Dis 5, 13–31. Kim, S.H., Kim, K.H., Seo, B.M., Koo, K.T., Kim, T.I., Seol, Y.J., Ku, Y., Rhyu, I.C., Chung, C.P., Lee, Y.M., 2009. Alveolar bone regeneration by transplantation of periodontal ligament stem cells and bone marrow stem cells in a canine peri-implant defect model: a pilot study. J. Periodontol. 80, 1815–1823. Kornman, K.S., Crane, A., Wang, H.Y., di Giovine, F.S., Newman, M.G., Pirk, F.W., Wilson Jr., T.G., Higginbottom, F.L., Duff, G.W., 1997. The interleukin-1 genotype as a severity factor in adult periodontal disease. J. Clin. Periodontol. 24, 72–77. Kramer, P.R., Nares, S., Kramer, S.F., Grogan, D., Kaiser, M., 2004. Mesenchymal stem cells acquire characteristic of cells in the periodontal ligament in vitro. J. Dent. Res. 83, 27–34. Lacey, D., Sampey, A., Mitchell, R., Bucala, R., Santos, L., Leech, M., Morand, E., 2003. Control of fibroblast-like synoviocyte proliferation by macrophage migration inhibitory factor. Arthritis Rheum. 48, 103–109. Lacey, D.C., Simmons, P.J., Graves, S.E., Hamilton, J.A., 2009. Proinflammatory cytokines inhibit osteogenic differentiation from stem cells: implications for bone repair during inflammation. Osteoarthritis Cartilage 17, 735–742. Lencel, P., Delplace, S., Hardouin, P., Magne, D., 2011. TNF-α stimulates alkaline phosphatase and mineralization through PPARγ inhibition in human osteoblasts. Bone 48, 242–249. Li, C., Li, B., Dong, Z., Gao, L., He, X., Liao, L., Hu, C., Wang, Q., Jin, Y., 2014. Lipopolysaccharide differentially affects the osteogenic differentiation of periodontal ligament stem cells and bone marrow mesenchymal stem cells through Toll-like receptor 4 mediated nuclear factor kappaB pathway. Stem Cell Res. Ther 27, 67. Liang, Y., Zhou, Y., Shen, P., 2004. NF-kappaB and its regulation on the immune system. Cell. Mol. Immunol. 1, 343–350. Lin, FH., Chang, J.B., McGuire, M.H., Yee, J.A., Brigman, B.E., 2010. Biphasic effects of interleukin-1beta on osteoblast differentiation in vitro. J. Orthop. Res 28, 958–964. Lin, G.L., Hankenson, K.D., 2011. Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation. J. Cell. Biochem. 112, 3491–3501. Liu, D.D., Zhang, J.C., Zhang, Q., Wang, S.X., Yang, M.S., 2013. TGF-β/BMP signaling pathway is involved in cerium-promoted osteogenic differentiation of mesenchymal stem cells. J. Cell. Biochem. 114, 1105–1114. Liu, D., Xu, J., Liu, O., Fan, Z., Liu, Y., Wang, F., Ding, G., Wei, F, Zhang, C., Wang, S., 2012. Mesenchymal stem cells derived from inflamed periodontal ligaments exhibit impaired immunomodulation. J. Clin. Periodontol. 39, 1174–1182. Liu, N., Shi, S., Deng, M., Tang, L., Zhang, G., Liu, N., Ding, B., Liu, W., Liu, Y., Shi, H., Liu, L., Jin, Y., 2011. 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. Liu, Y., Zheng, Y., Ding, G., Fang, D., Zhang, C., Bartold, P.M., Gronthos, S., Shi, S., Wang, S., 2008. Periodontal ligament stem cell-mediated treatment for periodontitis inminiature swine. Stem Cells 26, 1065–1073. Lu, X., Gilbert, L., He, X., Rubin, J., Nanes, M.S., 2006. Transcriptional regulation of the osterix (Osx, Sp7) promoter by tumor necrosis factor identifies disparate effects of mitogen-activated protein kinase and NF kappa B pathways. J. Biol. Chem. 281, 6297–6306.

105

Marion, N.W., Mao, J.J., 2006. Mesenchymal stem cells and tissue engineering. Methods Enzymol. 420, 339–361. Matsuoka, F., Takeuchi, I., Agata, H., Kagami, H., Shiono, H., Kiyota, Y., Honda, H., Kato, R., 2013. Morphology-based prediction of osteogenic differentiation potential of human mesenchymal stem cells. PLoS One 8, e55082. Menicanin, D., Bartold, P.M., Zannettino, A.C., Gronthos, S., 2010. Identification of a common gene expression signature associated with immature clonal mesenchymal cell populations derived from bone marrow and dental tissues. Stem Cells Dev. 19, 1501–1510. Mountziaris, P.M., Tzouanas, S.N., Mikos, A.G., 2010. Dose effect of tumor necrosis factor-alpha on in vitro osteogenic differentiation of mesenchymal stem cells on biodegradable polymeric microfiber scaffolds. Biomaterials 31, 1666–1675. Moe, K.T., Khairunnisa, K., Yin, N.O., Chin-Dusting, Wong, P., Wong, M.C., 2014. Tumor necrosis factor-a-induced nuclear factor kappa B activation in human cardiomyocytes is mediated by NADPH oxidase. J. Physiol. Biochem. 70, 769–779. Mrozik, K., Gronthos, S., Shi, S., Bartold, P.M., 2010. A method to isolate, purify, and characterize human periodontal ligament stem cells. Methods Mol. Biol. 666, 269–284. Nakashima, K., Zhou, X., Kunkel, G., Zhang, Z., Deng, J.M., Behringer, R.R., de Crombrugghe, B., 2002. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 108, 17–29. Nakase, T., Takaoka, K., Hirakawa, K., Hirota, S., Takemura, T., Onoue, H., Takebayashi, K., Kitamura, Y., Nomura, S., 1994. Alterations in the expression of osteonectin, osteopontin and osteocalcin mRNAs during the development of skeletal tissues in vivo. Bone Miner 26, 109–122. Owen, M., Friedenstein, A.J., 1988. Stromal stem cells: marrow-derived osteogenic precursors. Ciba Found. Symp. 136, 42–60. Papadopoulou, A.1., Yiangou, M., Athanasiou, E., Zogas, N., Kaloyannidis, P., Batsis, I., Fassas, A., Anagnostopoulos, A., Yannaki, E., 2012. Mesenchymal stem cells are conditionally therapeutic in preclinical models of rheumatoid arthritis. Ann. Rheum. Dis. 71, 1733–1740. Park, J.Y., Jeon, S.H., Choung, P.H., 2011. Efficacy of periodontal stem cell transplantation in the treatment of advanced periodontitis. Cell Transplant. 20, 271–285. Pihlstrom, B.L., Michalowicz, B.S., Johnson, N.W., 2005. Periodontal disease. Lancet 366, 1809–1820. Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K., Douglas, R., Mosca, J.D., Moorman, M.A., Simonetti, D.W., Craig, S., Marshak, D.R., 1999. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147. Platt, I.D., El-Sohemy, A., 2009. Regulation of osteoblast and adipocyte differentiation from human mesenchymal stem cells by conjugated linoleic acid. J. Nutr. Biochem. 20, 956–964. Rodríguez, J.P., González, M., Ríos, S., Cambiazo, V., 2004. Cytoskeletal organization of human mesenchymal stem cells (MSC) changes during their osteogenic differentiation. J. Cell. Biochem. 93, 721–731. Schett, G., 2011. Effects of inflammatory and anti-inflammatory cytokines on the bone. Eur. J. Clin. Invest. 41, 1361–1366. Seo, B.M., Miura, M., Gronthos, S., Bartold, P.M., Batouli, S., Brahim, J., Young, M., Robey, P.G., Wang, C.Y., Shi, S., 2004. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 364, 149–155. Short, B., Brouard, N., Occhiodoro-Scott, T., Ramakrishnan, A., Simmons, P.J., 2003. Mesenchymal stem cells. Arch. Med. Res. 34, 565–571. Soga, Y., Nishimura, F., Ohyama, H., Maeda, H., Takashiba, S., Murayama, Y., 2003. Tumor necrosis factor-alpha gene (TNF-α)  1031/ 863,  857 singlenucleotide polymorphisms (SNPs) are associated with severe adult periodontitis in Japanese. J. Clin. Periodontol. 30, 524–531. Sommer, B., Bickel, M., Hofstetter, W., Wetterwald, A., 1996. Expression of matrix proteins during the development of mineralized tissues. Bone 19, 371–380. Tamaki, Y., Nakahara, T., Ishikawa, H., Sato, S., 2013. In vitro analysis of mesenchymal stem cells derived from human teeth and bone marrow. Odontology 101, 121–132. Welter, J.F., Penick, K.J., Solchaga, L.A., 2013. Assessing adipogenic potential of mesenchymal stem cells: a rapid three-dimensional culture screening technique. Stem Cells Int 2013, 806525. Wu, G., Cui, Y., Ma, L., Pan, X., Wang, X., Zhang, B., 2014. Repairing cartilage defects with bone marrow mesenchymal stem cells induced by CDMP and TGF-β1. Cell Tissue Bank. 15, 51–57. Xu, J., Wu, H.F., Ang, E.S., Yip, K., Woloszyn, M., Zheng, M.H., Tan, R.X., 2009. NFkappa B modulators in osteolytic bone diseases. Cytokine Growth Factor Rev. 20, 7–17. Yang, Y., Rossi, F.M., Putnins, E.E., 2010. Periodontal regeneration using engineered bone marrow mesenchymal stromal cells. Biomaterials 31, 8574–8582. Yang, Z.H., Zhang, X.J., Dang, N.N., Ma, Z.F., Xu, L., Wu, J.J., Sun, Y.J., Duan, Y.Z., Lin, Z., Jin, Y., 2009. Apical tooth germ cell- conditioned medium enhances the differentiation of periodontal ligament stem cells into cementum/periodontal ligament-like tissues. J. Periodont. Res 44, 199–210. Zhang, S., Barros, S.P., Moretti, A.J., Yu, N., Zhou, J., Preisser, J.S., Niculescu, M.D., Offenbacher, S., 2013. Epigenetic regulation of TNFA expression in periodontal disease. J. Periodontol. 84, 1606–1616. Zhao, B., Grimes, S.N., Li, S., Hu, X., Ivashkiv, L.B., 2012. TNF-induced osteoclastogenesis sand inflammatory bone resorption are inhibited by transcription factor RBP-J. J. Exp. Med. 209, 319–334.

The difference on the osteogenic differentiation between periodontal ligament stem cells and bone marrow mesenchymal stem cells under inflammatory microenviroments.

Periodontitis is a major cause of tooth loss in adults and periodontal ligament stem cells (PDLSCs) is the most favorable candidate for the reconstruc...
2MB Sizes 2 Downloads 11 Views