Lipopolysaccharide stimulation improves the odontoblastic differentiation of human dental pulp cells

  • Authors:
    • Yihua Huang
    • Hongwei Jiang
    • Qimei Gong
    • Xuyan Li
    • Junqi Ling
  • View Affiliations

  • Published online on: December 19, 2014     https://doi.org/10.3892/mmr.2014.3120
  • Pages: 3547-3552
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lipopolysaccharide (LPS) is one of the causative agents of pulpitis and previous studies have demonstrated that the LPS stimulation of human aortic valve interstitial cells induces inflammatory mediators and the gene expression of osteogenic factors. Therefore, in the present study, it was hypothesized that LPS affects the odontoblastic differentiation of human dental pulp cells (hDPCs). In order to investigate this, an in vitro study using hDPCs was performed. Increased alkaline phosphatase (ALP) activity was observed in the hDPCs treated with LPS, which was more marked when the cells were costimulated with odontogenic induction medium (OM). LPS also appeared to increase the gene expression levels of dentin sialophosphoprotein and dentin matrix protein‑1 and the protein expression level of dental sialoprotein in the hDPCs, particularly in combination with OM. In addition, the size and the number of nodules formed in the hDPCs exposed to OM and LPS were increased compared to those stimulated by OM alone. To determine the role of nuclear factor κB (NF‑κB) during the LPS‑induced odontoblastic differentiation of hDPCs, immunofluorescence was performed. The nuclear translocation of NF‑κB, induced by LPS was confirmed, suggesting its involvement in the LPS‑induced increase in odontoblastic differentiation of hDPCs. In conclusion, there may be an association between LPS stimulation, with or without OM, and odontoblastic differentiation.

Introduction

Dental pulp tissues have the ability to repair and regenerate following injury. In the presence of an infection within the dentin-pulp complex, oral microorganisms and their components diffuse into the pulp through the dentinal tubules. This type of stimulation induces the mixed population of pulp cells (1,2), which include stem cells and odontoblast progenitor cells, to differentiate and form a dentin-like mineralized matrix. This is marked by an increase in the expression of hard-tissue-forming proteins, including alkaline phosphatase (ALP), dentin sialophosphoprotein (DSPP) and dentin matrix protein-1 (DMP-1) (3,4).

Lipopolysaccharides (LPS) comprise a major molecular component of the outer cell wall of Gram-negative bacteria and are potent virulence factors that contribute to bacterially induced pathology, including general sepsis (5), lung disease (6), periodontal disease (7) and pulpitis (8). In several experiments, LPS has been used to form a model of inflammation and the protective inflammatory reaction evoked by LPS is considered to induce signal transduction through Toll-like receptor 4 (TLR4) (9,10). In our previous study, the mRNA and protein expression of TLR4 was observed in the cells of the odontoblast layer and pulp tissues (11). TLRs, broadly distributed pattern recognition receptors, are also involved in the pathogenesis of chronic inflammatory diseases. LPS and peptidoglycans, which are TLR4 and TLR agonists, have been observed to induce the upregulation of osteogenesis-associated factors in human aortic valve interstitial cells (hAVICs) (12,13). To date, few studies have examined the effects of LPS or TLRs on hDPC differentiation.

Furthermore, LPS is known to activate NF-κB, predominantly through the TLR4 downstream pathway (14). Increased expression levels of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 are associated with the translocation of NF-κB from the nucleus to the cytoplasm in hDPCs following incubation with LPS (15).

hDPCs can promote reparative dentin formation by odontoblastic differentiation, resulting in the production of mineralized matrix (1,3). This property of differentiation by the pulp cells is critical for its repair following injury or inflammation and is similar to the processes of osteogenic differentiation and bone formation. In addition to LPS and TLR4, NF-κB may also contribute to osteogenic differentiation (12,13) and the present study hypothesized that they may be involved in the odontoblastic differentiation of hDPCs. In the present study, hDPCs were treated with Escherichia coli LPS in growth medium and odontogenic induction medium (OM), respectively. Subsequently, an ALP enzymatic assay, reverse transcription quantitative polymerase chain reaction (RT-qPCR), western blotting and alizarin red staining were used to investigate and confirm the upregulation of mineralization indicators in the hDPCs following stimulation. The translocation of NF-κB in the cells was evaluated using immunofluorescence microscopy.

Materials and methods

hDPC cultures

Extracted premolars or third molars were collected from patients (13–25 years old) at the Department of Oral and Maxillofacial Surgery, Guanghua School and Hospital of Stomatology, Sun Yat-sen University (Guangzhou, China). Informed consent was obtained from each patient and the study was performed under the approval of the ethics committee of Guanghua School and Hospital of Stomatology, Sun Yat-sen University (Guangzhou, China). The hDPCs were isolated, as previously reported (16) and cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco-BRL, Grand Island, NY, USA) supplemented with 100 U/ml penicillin, 100 μg/ml streptomycin (Sigma-Aldrich, St Louis, MO, USA) and 10% fetal bovine serum (FBS; Gibco-BRL). The cultures were maintained at 37°C in an incubator (5% CO2/20% O2). The cells obtained between passages three and four were used in the subsequent experiments.

ALP

The hDPCs (5×105 cells per well) were seeded into 12-well culture plates. The cells in the experimental groups were treated with either 0.1, 1.0 and 10.0 μg/ml LPS (E. coli 0111:B4; Sigma) in growth medium or odontogenic induction medium (OM; 2%FBS, 10 mmol/l β-glycerophosphate, 50 μmol/l ascorbic acid and 100 nmol/l dexamethasone in DMEM) for 1, 3, 5 and 7 days, respectively, while those belonging to the control group were incubated with growth medium and OM alone. The cultures were maintained at 37°C in an incubator (5% CO2/20% O2). Following treatment at each time point, the plates were washed twice with phosphate-buffered-saline (PBS) and lysed with 200 μl 1% Triton X-100. After 16 h, 30 μl aliquots of cell lysate per well were subjected to ALP activity and protein content measurement using an ALP kit (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) and a bicinchoninic acid assay kit (Boshide Bioengineering Co., Ltd., Wuhan, China). All results were normalized by total protein content.

RT-qPCR

The cells from the experimental groups were treated with 0.1 μg/ml LPS, in growth medium and OM for 3 days, while the control groups were incubated with growth medium and OM alone. The cultures were maintained at 37°C in an incubator (5% CO2/20% O2). Following the treatment, total RNA was extracted using TRIzol (Invitrogen Life Technologies, Carlsbad, CA, USA). Quantification of the mRNA levels of DSPP, DMP-1 and GAPDH were performed based on previously described methods (17). Complementary DNA was synthesized from 2 μg total RNA using Revert AidTM First Strand cDNA Synthesis kit (Fermentas, Ottawa, Canada) according to the manufacturer’s instructions. RT-qPCR was performed to detect genes of DSPP, DMP-1 on the Chromo4 four-color Real-Time PCR system (Bio-Rad Laboratories, Inc., Hercules, CA, USA) using SYBR Green Realtime PCR Master mix (Toyobo, Osaka, Japan). The sequences of the primers used are listed in Table I. The conditions for RT-qPCR were as follows: 95°C for 10 min for activation, followed by 40 cycles of denaturation at 95°C for 15 sec and primer extension at 60°C for 1 min. All reactions were performed in triplicate for the independent experiments. The mean cycle threshold (ΔCt) value of each target gene was normalized against ΔCt value of house-keeping gene GAPDH and the relative expression calculated using the following formula: 2−(normalized average ΔCts)x104. The expression of the target gene was subsequently converted to the fold-change of the control gene.

Table I

Primer sequences used in reverse transcription quantitative polymerase chain reaction.

Table I

Primer sequences used in reverse transcription quantitative polymerase chain reaction.

GenePrimer sequenceLength (bp)
DSPPForward: 5′-GCCACTTTCAGTCTTCAAAGAGA-3′
Reverse: 5′-GCCCAAATGCAAAAATATGTAA-3′
130
DMP1Forward: 5′-TGGGCATAGATTTCCTCTTTG-3′
Reverse: 5′-TGAGCAGGATGCTGATCTTC-3′
121
GAPDHForward: 5′-AAGGTGAAGGTCGGAGTCAA-3′
Reverse: 5′-AATGAAGGGGTCATTGATGG-3′
108

[i] DSPP, dentin sialophosphoprotein; DMP1, dentin matrix protein-1; bp, base pairs.

Western blotting

The cells from the experimental groups were treated with 0.1, 1.0 and 10.0 μg/ml LPS, in growth medium and in OM for 3 days, while the control group cells were incubated with growth medium and OM alone. The cultures were maintained at 37°C in an incubator (5% CO2/20% O2). Following treatment, the hDPCs were harvested in lysis buffer containing 1% Igepal CA-630 (Sigma-Aldrich), 0.5% sodium deoxycholate and 0.1% sodium dodecylsulfate-polyacrylamide. Protease inhibitor cocktail (Merck Millipore, Darmstadt, Germany), containing AEBSF, hydrochloride, aprotinin, bovine lung, crystalline, E-64 protease inhibitor, EDTA, disodium, leupeptin and hemisulfate, was added prior to cell lysis. Western blotting was performed, as described previously (11). The proteins were detected using mouse polyclonal antibodies against human dental sialoprotein (DSP; 1:1,000; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and mouse monoclonal antibodies against human GAPDH (1:3,000; EarthOx, San Francisco, CA, USA).

Immunofluorescence

The hDPCs (5.0×105 cells per well) were seeded into 12-well culture plates and cultured overnight. The experimental group was then treated with 0.1 μg/ml LPS, with/without OM, for 16 h, while the control group was incubated with growth medium and OM alone. The cultures were maintained at 37°C in an incubator (5% CO2/20% O2). Following treatment, the cells were fixed with methanol for 15 min and washed with PBS. The cells were then permeabilized with 0.1% Triton X-100 in PBS for 10 min, incubated in 3% bovine serum albumin in PBS for 30 min and then with the primary monoclonal anti-NF-κB antibody (1:50; Cell Signaling Technology, Inc., Boston, MA, USA) and primary monoclonal anti-c-Jun antibody (1:50; Millipore, Temecula, CA, USA) simultaneously at 4°C overnight. The cells were rinsed and then incubated with fluorescein isothiocyanate-conjugated secondary antibody (1:200; Cell Signaling Technology, Inc.). Detection was then performed using a fluorescence microscope (Axio Observer Z1; Zeiss, Oberkochen, Germany).

Alizarin red stain

The hDPCs (1.0×106 cells per well) were seeded into 6-well culture plates and cultured overnight. The experimental group was then treated with 0.1 μg/ml LPS in growth medium and OM for 14 days and the control group was incubated with growth medium and OM alone. Following treatment, at each time point, the mineralized cells were washed with 1X PBS and fixed using 95% ethanol at room temperature for 10 min. Calcium accumulation was detected using 40 mM alizarin red stain solution (Sigma-Aldrich). Following washing with distilled water, images of the dried-culture dishes were captured under a fluorescence microscope (Axio Observer Z1; Zeiss). The positively stained nodules appeared orange/red.

Statistical analysis

The SPSS 16.0 software package (SPSS, Inc, Chicago, IL, USA) was used to perform statistical analyses. The ALP activity, protein expression of DSP and gene expression of DSPP in the different treatment groups were assessed using the Least Significant Difference test. The gene expression of DMP-1 was assessed using the Kruskal-Wallis and Bonferronni’s test due to heterogeneity of variance. P<0.05 and P<0.01 were considered to indicate statistically significant differences.

Results

Effect of LPS on ALP activity

The activity of ALP was examined at different time points using an ALP enzymatic assay and was found to be higher in the hDPCs treated with LPS at all concentrations in the growth medium on the 7th day (P<0.05; Fig. 1A), while no changes were observed on the 3rd and 5th days. In OM, the ALP activity of cells exposed to three concentrations of LPS increased at each time point (P<0.01; Fig. 1B) with the exception of the 1st day and at the final concentration of 1.0 μg/l LPS on the 3rd day. Exposure of the stimulated cells to 10.0 μg/l resulted in the highest ALP activity on day 7.

Effect of LPS on the gene expression levels of DSPP and DMP-1

The gene expression levels of DSPP and DMP-1 were examined using RT-qPCR on the 3rd day of treatment with 0.1 μg/ml LPS in the growth medium and OM. The expression levels of DSPP increased significantly in the hDPCs treated with LPS compared with the control group in growth medium (P<0.05) and in OM (P<0.01). The expression levels of DMP-1 were significantly higher in the hDPCs treated with LPS compared with OM alone (P<0.05; Fig. 2).

Effect of LPS on the expression of DSP

After treatment with LPS for 3 days, the protein levels of DSP were increased in the hDPCs at concentrations of 0.1, 1.0 and 10.0 μg/ml compared with the control group in growth medium and OM (P<0.01). The highest expression level of DSP was observed at a concentration of 0.1 μg/ml LPS in OM (P<0.01; Fig. 3).

LPS induces the nuclear translocation of NF-κB, but not c-Jun, in the hDPCs

The nuclear and cytosolic expression levels of NF-κB and c-Jun were detected using immunofluorescence after 16 h treatment with LPS (0.1 μg/ml) with or without OM. NF-κB was localized predominantly in the cytoplasm of the cells treated with 2% FBS in DMEM alone and in the nuclei of the cells in the experimental groups treated with LPS wit or without OM (Fig. 4). However, no change was noted in the expression of c-Jun in the cells in either the control or experimental group.

Effect of LPS on the mineralized nodule formation of hDPCs

To determine whether E. coli LPS stimulation increased cellular calcification, the cells were treated in OM with or without 0.1 μg/ml LPS for 14 days (Fig. 5). The results revealed that LPS promoted calcified nodule formation when the hDPCs were stimulated. As shown in Fig. 5, nodules of increased size and number were formed in the hDPCs exposed to OM and LPS compared with those stimulated by OM alone.

Discussion

Subsequent to injury or inflammation, reparative dentin is formed by the odontoblastic differentiation of hDPCs leading to increased production of mineralized matrix (1,3). As bacteria are one of the most important causative agents of pulpitis, LPS, which comprises a major molecular component of the outer cell wall of Gram-negative bacteria, is often used to establish a model of inflammation for the investigation of dental pulp cells (8). The present study used E. coli LPS, as it is a putative inflammation inducer, evokes a protective inflammatory reaction and is hypothesized to induce signal transduction through TLR4 (9,10). The present study hypothesized that LPS and TLR4 may be involved in the odontoblastic differentiation of hDPCs.

ALP, DSPP and DMP-1 are mineralization markers for odontoblast and osteoblast-like differentiation of hDPCs (3,18). ALP activity is higher in odontoblast-like cells compared to undifferentiated dental mesenchymal cells and is closely associated with dentin formation (19). In the present study, increased ALP activity was observed in the hDPCs treated with LPS, which was more marked when the cells were simultaneously stimulated with OM. The rise in ALP activity was time-dependent, but not dose-dependent, indicating that treatments with OM and LPS, with or without OM, may enhance odontoblastic differentiation. DSP, which promotes predentin to transform into mature dentin (20), is considered to be a specific labeled protein for odontoblastic differentiation (21). DSPP and DMP-1 are also expressed by differentiating odontoblasts. Western blot analysis revealed that the protein levels of DSP were similar to those of ALP following treatment. Similarly, the mRNA levels of DSPP and DMP-1 were reconfirmed by western blotting. Furthermore, larger and increased numbers of nodules were formed in the hDPCs exposed to OM and LPS compared with those stimulated by OM only. These observations suggested that there may be an association between LPS stimulation, with or without OM, and odontoblastic differentiation.

The results of the present study are in agreement with those of calcific aortic valve cells (hAVICs). Previous studies on calcific aortic valve stenosis have revealed that LPS stimulation of hAVICs induces inflammatory mediators and the gene expression of osteogenic factors, similar to those induced by OM (22,23). Osteogenic changes have been associated with high expression levels of TLR4 (13). In addition, the combined effects of LPS and OM have been demonstrated on the osteogenic differentiation of hAVICs (12). As fibroblasts form the major bulk of hDPCs, it they may have similarities to hAVICs, which are myofibroblasts. It has been previously reported that human dental pulp stem cells exposed to LPS, obtained from Porphyromonas gingivalis, exhibit a reduction in the expression levels of DSPP and osteocalcin and this effect can be moderated by TLR2 inhibition (24). Based on this finding and the results of the present study, it can be concluded that LPS from different bacteria, including P. gingivalis and E. coli, may not simulate the same receptor and may exert different effects with different receptors.

NF-κB activation is known to contribute to cytokine induction in response to bacterial products and is necessary for innate immunity and inflammation (25,26). It has been demonstrated that NF-κB is required for the LPS-induced expression of interleukin-8 in the hDPSCs and NF-κB is important in mediating the intracellular signaling generated by LPS, which induces the expression of pro-inflammatory genes (27). LPS has also been demonstrated to cause NF-κB intranuclear translocation and rapid phosphorylation in hAVICs (12). In the present study, the LPS-induced translocation of NF-κB was demonstrated using immunofluorescence and was in accordance with the results of western blot analysis reported in a previous study (15), indicating its role during the LPS-induced odontoblastic differentiation of hDPCs. However, LPS did not affect the translocation of c-Jun. The results suggested that NF-κB was involved in the transcriptional activation of various downstream genes and may have a similar role in the odontoblastic differentiation of hDPCs, improved by OM and LPS with or without OM.

In the present study, the expression of mineralization markers for the odontoblast and osteoblast-like differentiation of hDPCs stimulated by LPS, with or without OM, increased, suggesting the involvement of TLR4 and NF-κB. To further understand the roles of LPS, TLR4 and NF-κB in the odontoblastic differentiation of hDPCs, further studies examining the involvement of the TLR4 inhibitor, NF-κB inhibitor and the relevant signal pathways involved are required.

In addition to inducing ALP activity, LPS was observed to increase the gene expression levels of DSPP and DMP-1 and the protein expression of DSP in the hDPCs, particularly in combination with OM. The nuclear translocation of NF-κB, induced by LPS suggested that NF-κB is involved in the LPS-induced increase in odontoblastic differentiation of hDPCs.

Acknowledgements

This study was supported by the Key Clinical Program of the Ministry of Health, China (no. [2010] 439), the National Nature Science Foundation of China (nos. 81100742 and 81200775) and the Fundamental Research Funds for the Central Universities (no. 12ykpy65).

References

1 

Gronthos S, Brahim J, Li W, et al: Stem cell properties of human dental pulp stem cells. J Dent Res. 81:531–535. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Goldberg M, Farges JC, Lacerda-Pinheiro S, et al: Inflammatory and immunological aspects of dental pulp repair. Pharmacol Res. 58:137–147. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Wei X, Ling J, Wu L, Liu L and Xiao Y: Expression of mineralization markers in dental pulp cells. J Endod. 33:703–708. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Wei X, Wu L, Ling J, et al: Differentially expressed protein profile of human dental pulp cells in the early process of odontoblast-like differentiation in vitro. J Endod. 34:1077–1084. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Tobias PS, Mathison JC and Ulevitch RJ: A family of lipopolysaccharide binding proteins involved in responses to gram-negative sepsis. J Biol Chem. 263:13479–13481. 1988.PubMed/NCBI

6 

Vernooy JH, Dentener MA, van Suylen RJ, Buurman WA and Wouters EF: Long-term intratracheal lipopolysaccharide exposure in mice results in chronic lung inflammation and persistent pathology. Am J Respir Cell Mol Biol. 26:152–159. 2002. View Article : Google Scholar

7 

Hanazawa S, Nakada K, Ohmori Y, Miyoshi T, Amano S and Kitano S: Functional role of interleukin 1 in periodontal disease: induction of interleukin 1 production by Bacteroides gingivalis lipopolysaccharide in peritoneal macrophages from C3H/HeN and C3H/HeJ mice. Infect Immun. 50:262–270. 1985.PubMed/NCBI

8 

Vianna ME, Horz HP, Conrads G, Zaia AA, Souza-Filho FJ and Gomes BP: Effect of root canal procedures on endotoxins and endodontic pathogens. Oral Microbiol Immunol. 22:411–418. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Hoshino K, Takeuchi O, Kawai T, et al: Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J Immunol. 162:3749–3752. 1999.PubMed/NCBI

10 

Yang X, Coriolan D, Murthy V, Schultz K, Golenbock DT and Beasley D: Proinflammatory phenotype of vascular smooth muscle cells: role of efficient Toll-like receptor 4 signaling. Am J Physiol Heart Circ Physiol. 289:H1069–H1076. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Jiang HW, Zhang W, Ren BP, Zeng JF and Ling JQ: Expression of toll like receptor 4 in normal human odontoblasts and dental pulp tissue. J Endod. 32:747–751. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Meng X, Ao L, Song Y, et al: Expression of functional Toll-like receptors 2 and 4 in human aortic valve interstitial cells: potential roles in aortic valve inflammation and stenosis. Am J Physiol Cell Physiol. 294:C29–C35. 2008. View Article : Google Scholar

13 

Yang X, Fullerton DA, Su X, Ao L and Meng X: Pro-osteogenic phenotype of human aortic valve interstitial cells is associated with higher levels of Toll-like receptors 2 and 4 and enhanced expression of bone morphogenetic protein 2. J Am Coll Cardiol. 53:491–500. 2009. View Article : Google Scholar : PubMed/NCBI

14 

O’Neill LA and Bowie AG: The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol. 7:353–364. 2007. View Article : Google Scholar

15 

Lee JC, Yu MK, Lee R, et al: Terrein reduces pulpal inflammation in human dental pulp cells. J Endod. 34:433–437. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Wang J, Wei X, Ling J, Huang Y and Gong Q: Side population increase after simulated transient ischemia in human dental pulp cell. J Endod. 36:453–458. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Jiang HW, Ling JQ and Gong QM: The expression of stromal cell-derived factor 1 (SDF-1) in inflamed human dental pulp. J Endod. 34:1351–1354. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Yamada Y, Fujimoto A, Ito A, et al: Cluster analysis and gene expression profiles: a cDNA microarray system-based comparison between human dental pulp stem cells (hDPCs) and human mesenchymal stem cells (hMSCs) for tissue engineering cell therapy. Biomaterials. 27:3766–3781. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Shiba H, Mouri Y, Komatsuzawa H, et al: Enhancement of alkaline phosphatase synthesis in pulp cells co-cultured with epithelial cells derived from lower rabbit incisors. Cell Biol Int. 27:815–823. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Paine ML, Luo W, Wang HJ, et al: Dentin sialoprotein and dentin phosphoprotein overexpression during amelogenesis. J Biol Chem. 280:31991–31998. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Begue-Kirn C, Krebsbach PH, Bartlett JD and Butler WT: Dentin sialoprotein, dentin phosphoprotein, enamelysin and ameloblastin: tooth-specific molecules that are distinctively expressed during murine dental differentiation. Eur J Oral Sci. 106:963–970. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Babu AN, Meng X, Zou N, et al: Lipopolysaccharide stimulation of human aortic valve interstitial cells activates inflammation and osteogenesis. Ann Thorac Surg. 86:71–76. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Lopez J, Fernandez-Pisonero I, Duenas AI, et al: Viral and bacterial patterns induce TLR-mediated sustained inflammation and calcification in aortic valve interstitial cells. Int J Cardiol. 158:18–25. 2012. View Article : Google Scholar

24 

Yamagishi VT, Torneck CD, Friedman S, Huang GT and Glogauer M: Blockade of TLR2 inhibits Porphyromonas gingivalis suppression of mineralized matrix formation by human dental pulp stem cells. J Endod. 37:812–818. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Janssens S and Beyaert R: A universal role for MyD88 in TLR/IL-1R-mediated signaling. Trends Biochem Sci. 27:474–482. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Chang J, Zhang C, Tani-Ishii N, Shi S and Wang CY: NF-κB activation in human dental pulp stem cells by TNF and LPS. J Dent Res. 84:994–998. 2005. View Article : Google Scholar : PubMed/NCBI

27 

He W, Qu T, Yu Q, et al: LPS induces IL-8 expression through TLR4, MyD88, NF-kappaB and MAPK pathways in human dental pulp stem cells. Int Endod J. 46:128–136. 2013. View Article : Google Scholar

Related Articles

Journal Cover

May-2015
Volume 11 Issue 5

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
Huang Y, Jiang H, Gong Q, Li X and Ling J: Lipopolysaccharide stimulation improves the odontoblastic differentiation of human dental pulp cells. Mol Med Rep 11: 3547-3552, 2015
APA
Huang, Y., Jiang, H., Gong, Q., Li, X., & Ling, J. (2015). Lipopolysaccharide stimulation improves the odontoblastic differentiation of human dental pulp cells. Molecular Medicine Reports, 11, 3547-3552. https://doi.org/10.3892/mmr.2014.3120
MLA
Huang, Y., Jiang, H., Gong, Q., Li, X., Ling, J."Lipopolysaccharide stimulation improves the odontoblastic differentiation of human dental pulp cells". Molecular Medicine Reports 11.5 (2015): 3547-3552.
Chicago
Huang, Y., Jiang, H., Gong, Q., Li, X., Ling, J."Lipopolysaccharide stimulation improves the odontoblastic differentiation of human dental pulp cells". Molecular Medicine Reports 11, no. 5 (2015): 3547-3552. https://doi.org/10.3892/mmr.2014.3120