Acessibilidade / Reportar erro

Low-intensity pulsed ultrasound promotes tissue regeneration in rat dental follicle cells in a porous ceramic scaffold

Abstract

The aim of this study was to investigate the effects of low-intensity pulsed ultrasound (LIPUS) on the osteogenic differentiation of dental follicle cells (DFCs) in vitro and on the regenerative effects of DFC-OsteoBoneTM complexes in vivo. DFCs were isolated and characterized. In the in vitro study, DFCs were cultured in an osteogenic medium in the presence or absence of LIPUS. The expression levels of ALP, Runx2, OSX, and COL-I mRNA were analyzed using real-time polymerase chain reaction (RT-PCR) on day 7. Alizarin red staining was performed on day 21. The state of the growth of the DFCs that were seeded on the scaffold at 3, 5, 7, and 9 days was detected by using a scanning electron microscope. In our in vivo study, 9 healthy nude mice randomly underwent subcutaneous transplantation surgery in one of three groups: group A, empty scaffold; group B, DFCs + scaffold; and group C, DFCs + scaffold + LIPUS. After 8 weeks of implantation, a histological analysis was performed by HE and Mason staining. Our results indicate that LIPUS promotes the osteogenic differentiation of DFCs by increasing the expression of the ALP, Runx2, OSX, and COL-I genes and the formation of mineralized nodules. The cells can adhere and grow on the scaffolds and grow best at 9 days. The HE and Mason staining results showed that more cells, fibrous tissue and blood vessels could be observed in the DFCs + scaffold + LIPUS group than in the other groups. LIPUS could promote the osteogenic differentiation of DFCs in vitro and promote tissue regeneration in a DFCs-scaffold complex in vivo. Further studies should be conducted to explore the underlying mechanisms of LIPUS.

Ultrasonic Waves; Dental Sac; Osteogenesis; Regeneration

Introduction

Periodontal disease is characterized by the progressive destruction of tooth-supporting tissues, such as alveolar bone, periodontal ligaments and cementum, and the subsequent loss of teeth.11. Hynes K, Menicanin D, Gronthos S, Bartold PM. Clinical utility of stem cells for periodontal regeneration. Periodontol 2000. 2012 Jun;59(1):203-27. https://doi.org/10.1111/j.1600-0757.2012.00443.x
https://doi.org/10.1111/j.1600-0757.2012...
It affects a large population worldwide.22. Jin LJ, Armitage GC, Klinge B, Lang NP, Tonetti M, Williams RC. Global oral health inequalities: task group—periodontal disease. Adv Dent Res. 2011 May;23(2):221-6. https://doi.org/10.1177/0022034511402080
https://doi.org/10.1177/0022034511402080...
, 33. Baelum V, López R. Periodontal disease epidemiology: learned and unlearned? Periodontol 2000. 2013 Jun;62(1):37-58. https://doi.org/10.1111/j.1600-0757.2012.00449.x
https://doi.org/10.1111/j.1600-0757.2012...
Conventional treatment strategies, such as oral hygiene instruction and scaling and root planing, aim to prevent the disease, halt its progress and maintain therapeutic outcomes achieved by long-term control of dental plaque accumulation,44. Racz GZ, Kadar K, Foldes A, Kallo K, Perczel-Kovach K, Keremi B, et al. Immunomodulatory and potential therapeutic role of mesenchymal stem cells in periodontitis. J Physiol Pharmacol. 2014 Jun;65(3):327-39. which has little effect on the regeneration of periodontal tissue.55. Bosshardt DD, Sculean A. Does periodontal tissue regeneration really work? Periodontol 2000. 2009;51(1):208-19. https://doi.org/10.1111/j.1600-0757.2009.00317.x
https://doi.org/10.1111/j.1600-0757.2009...
In recent years, efforts have focused on regenerating lost alveolar bone through the use of autografts (cortical/cancellous bone and bone marrow), allografts (demineralized freeze-dried/freeze-dried bone) and alloplastic materials (bioglass, polymers and hydroxyapatite).66. Lin N-H, Gronthos S, Bartod PM. Stem cells and periodontal regeneration. Aust Dent J. 2010;53(2):108-21. https://doi.org/10.1111/j.1834-7819.2008.00019.x
https://doi.org/10.1111/j.1834-7819.2008...
However, the use of such materials for periodontal regeneration has been questioned due to issues such as the variability in safety, clinical effectiveness and stability over time of these agents.77. Bartold PM, McCulloch CA, Narayanan AS, Pitaru S. Tissue engineering: a new paradigm for periodontal regeneration based on molecular and cell biology. Periodontol 2000. 2000 Oct;24(1):253-69. https://doi.org/10.1034/j.1600-0757.2000.2240113.x PMID:11276871
https://doi.org/10.1034/j.1600-0757.2000...
, 88. Becker W, Becker BE. Periodontal regeneration: a contemporary re-evaluation. Periodontol 2000. 1999 Feb;19(1):104-14. https://doi.org/10.1111/j.1600-0757.1999.tb00150.x
https://doi.org/10.1111/j.1600-0757.1999...
, 99. Karring T, Nyman S. Gottlow J, Laurell L. Development of the biological concept of guided tissue regeneration: animal and human studies. Periodontol. 2000. 2010;1:26-35. https://doi.org/10.1111/j.1600-0757.1993.tb00204.x
https://doi.org/10.1111/j.1600-0757.1993...

More recently, cell-based tissue engineering technology has emerged as a possible alternative to previously used treatments. Cell-based tissue engineering technology has been reported to supplement traditional restorative or surgical techniques when replacing injured or pathologically damaged tissues.1010. Catón J, Bostanci N, Remboutsika E, De Bari C, Mitsiadis TA. Future dentistry: cell therapy meets tooth and periodontal repair and regeneration. J Cell Mol Med. 2011 May;15(5):1054-65. https://doi.org/10.1111/j.1582-4934.2010.01251.x
https://doi.org/10.1111/j.1582-4934.2010...
This therapeutic procedure has three principal components: scaffolds, seed cells and growth factors.1111. Baldwin J, Antille M, Bonda U, De-Juan-Pardo EM, Khosrotehrani K, Ivanovski S, et al. In vitro pre-vascularisation of tissue-engineered constructs A co-culture perspective. Vasc Cell. 2014 Jun;6(1):13. https://doi.org/10.1186/2045-824X-6-13
https://doi.org/10.1186/2045-824X-6-13...
Scaffolds are temporary structures that are used to provide a three-dimensional microenvironment where cells can proliferate, differentiate and generate the desired tissue.1212. Kemppainen JM, Hollister SJ. Tailoring the mechanical properties of 3D-designed poly(glycerol sebacate) scaffolds for cartilage applications. J Biomed Mater Res A. 2010 Jul;94(1):9-18. https://doi.org/10.1002/jbm.a.32653
https://doi.org/10.1002/jbm.a.32653...
The OsteoBoneTM scaffold (Yenssen Biotech, Jiangsu, China) is an inorganic ceramic material with a porous structure that has a biomimetic 3D internal geometry that is favorable to cell seeding and new bone and blood vessel growth.1313. Sun XJ, Xia LG, Chou LL, Zhong W, Zhang XL, Wang SY, et al. Maxillary sinus floor elevation using a tissue engineered bone complex with BMP-2 gene modified bMSCs and a novel porous ceramic scaffold in rabbits. Arch Oral Biol. 2010 Mar;55(3):195-202. https://doi.org/10.1016/j.archoralbio.2010.01.006
https://doi.org/10.1016/j.archoralbio.20...
Grafting of exogenous cells has been shown to facilitate the generation of new tissue and/or the formation of a local microenvironment that is more suitable for the stimulation of endogenous progenitors, with promising results.1414. Monsarrat P, Vergnes JN, Nabet C, Sixou M, Snead ML, Planat-Bénard V, et al. Concise review: mesenchymal stromal cells used for periodontal regeneration: a systematic review. Stem Cells Transl Med. 2014 Jun;3(6):768-74. https://doi.org/10.5966/sctm.2013-0183
https://doi.org/10.5966/sctm.2013-0183...
Dental follicle cells (DFCs) are recognized as the progenitor cells of periodontal ligament cells (PDLCs), cementoblasts and osteoblasts within the dental follicle.1515. Honda MJ, Imaizumi M, Tsuchiya S, Morsczeck C. Dental follicle stem cells and tissue engineering. J Oral Sci. 2010 Dec;52(4):541-52. https://doi.org/10.2334/josnusd.52.541
https://doi.org/10.2334/josnusd.52.541...
, 1616. MI C, Garant PR. Development and general structure of the periodontium. Periodontol 2000. 2010 Oct;24(1):9-27. https://doi.org/10.1034/j.1600-0757.2000.2240102.x
https://doi.org/10.1034/j.1600-0757.2000...
These cells can develop into the components that constitute different periodontal tissues, such as periodontal ligament fibers and cementum and alveolar bone, when dental tissue injury occurs.1717. Wu Y, Feng G, Song J, Zhang Y, Yu Y, Huang L, et al. TrAmplification of human dental follicle cells by piggyBac transposon: mediated reversible immortalization system. PLoS One. 2015 Jul;10(7):e0130937. https://doi.org/10.1371/journal.pone.0130937
https://doi.org/10.1371/journal.pone.013...
Moreover, human DFCs can be isolated from impacted teeth that have been clinically discarded and can be frozen and stored for many years. Hence, DFCs have been considered ideal and promising candidate seed cells for periodontal tissue engineering. Indeed, the application of exogenous growth factors may enhance the osteogenic differentiation of cells.1818. Miao Z, Lu Z, Luo S, Lei D, He Y, Wu H, et al. Murine and Chinese cobra venom derived nerve growth factor stimulate chondrogenic differentiation of BMSCs in vitro: A comparative study. Mol Med Rep. 2018 Sep;18(3):3341-9. https://doi.org/10.3892/mmr.2018.9307
https://doi.org/10.3892/mmr.2018.9307...
, 1919. Ren J, Ward D, Chen S, Tran K, Jin P, Sabatino M, et al. Comparison of human bone marrow stromal cells cultured in human platelet growth factors and fetal bovine serum. J Transl Med. 2018 Mar;16(1):65. https://doi.org/10.1186/s12967-018-1400-3
https://doi.org/10.1186/s12967-018-1400-...
, 2020. Fujita T, Shiba H, Van Dyke TE, Kurihara H. Differential effects of growth factors and cytokines on the synthesis of SPARC, DNA, fibronectin and alkaline phosphatase activity in human periodontal ligament cells. Cell Biol Int. 2004;28(4):281-6. https://doi.org/10.1016/j.cellbi.2003.12.007
https://doi.org/10.1016/j.cellbi.2003.12...
However, the large-scale utilization of growth factors is clinically impracticable due to safety concerns.2121. Ferrandis E, Pradhananga SL, Touvay C, Kinoshita C, Wilkinson IR, Stafford K, et al. Immunogenicity, toxicology, pharmacokinetics and pharmacodynamics of growth hormone ligand-receptor fusions. Clin Sci (Lond). 2010 Aug;119(11):483-91. https://doi.org/10.1042/CS20100241
https://doi.org/10.1042/CS20100241...
Therefore, it is imperative to seek a new modality that can enhance the regenerative effect of cell-based tissue engineering.

A variety of mechanical stimuli have been actively studied to induce osteogenic differentiation, such as shock waves,2222. Hofmann A, Ritz U, Hessmann MH, Alini M, Rommens PM, Rompe JD. Extracorporeal shock wave-mediated changes in proliferation, differentiation, and gene expression of human osteoblasts. J Trauma. 2008 Dec;65(6):1402-10. https://doi.org/10.1097/TA.0b013e318173e7c2
https://doi.org/10.1097/TA.0b013e318173e...
pulsed electromagnetic fields2323. Jansen JH, van der Jagt OP, Punt BJ, Verhaar JA, van Leeuwen JP, Weinans H, et al. Stimulation of osteogenic differentiation in human osteoprogenitor cells by pulsed electromagnetic fields: an in vitro study. BMC Musculoskelet Disord. 2010 Aug;11(1):188. https://doi.org/10.1186/1471-2474-11-188
https://doi.org/10.1186/1471-2474-11-188...
and LIPUS (low intensity pulsed ultrasounds).2424. Hu B, Zhang Y, Zhou J, Li J, Deng F, Wang Z, et al. Low-intensity pulsed ultrasound stimulation facilitates osteogenic differentiation of human periodontal ligament cells. PLoS One. 2014 Apr;9(4):e95168. https://doi.org/10.1371/journal.pone.0095168
https://doi.org/10.1371/journal.pone.009...
Of these, LIPUS (intensity ranging from 30-100 mW/cm22. Jin LJ, Armitage GC, Klinge B, Lang NP, Tonetti M, Williams RC. Global oral health inequalities: task group—periodontal disease. Adv Dent Res. 2011 May;23(2):221-6. https://doi.org/10.1177/0022034511402080
https://doi.org/10.1177/0022034511402080...
) is a source of mechanical energy transmitted as acoustic pressure waves into biological tissues and subsequently evoking biochemical effects at the cellular level.2525. Rego EB, Takata T, Tanne K, Tanaka E. Current status of low intensity pulsed ultrasound for dental purposes. Open Dent J. 2012;6(1):220-5. https://doi.org/10.2174/1874210601206010220
https://doi.org/10.2174/1874210601206010...
At present, it has been widely used for the treatment of bone fractures and nonunions in the clinic.2626. Padilla F, Puts R, Vico L, Guignandon A, Raum K. Stimulation of bone repair with ultrasound. Adv Exp Med Biol. 2016 Jan; 880:385-427. https://doi.org/10.1007/978-3-319-22536-4_21
https://doi.org/10.1007/978-3-319-22536-...
As a form of non-invasive and safe mechanical stimulation, LIPUS also shows some advantages in the treatment of periodontal disease. Ikai H et al.2727. Ikai H, Tamura T, Watanabe T, Itou M, Sugaya A, Iwabuchi S, et al. Low-intensity pulsed ultrasound accelerates periodontal wound healing after flap surgery. J Periodontal Res. 2008 Apr;43(2):212-6. https://doi.org/10.1111/j.1600-0765.2007.01016.x
https://doi.org/10.1111/j.1600-0765.2007...
showed that LIPUS could enhance periodontal wound healing and bone repair by affecting osteoblasts and cells in the gingival epithelium and periodontal ligament. Studies have also reported that LIPUS can promote periodontal tissue repair and alveolar bone healing of extraction sockets by accelerating the calcium salt deposition and new bone formation in animal models.2828. Gu XQ, Li YM, Guo J, Zhang LH, Li D, Gai XD. Effect of low intensity pulsed ultrasound on repairing the periodontal bone of Beagle canines. Asian Pac J Trop Med. 2014 Apr;7(4):325-8. https://doi.org/10.1016/S1995-7645(14)60049-3
https://doi.org/10.1016/S1995-7645(14)60...
, 2929. Wang Y, Chai Z, Zhang Y, Deng F, Wang Z, Song J. Influence of low-intensity pulsed ultrasound on osteogenic tissue regeneration in a periodontal injury model: x-ray image alterations assessed by micro-computed tomography. Ultrasonics. 2014 Aug;54(6):1581-4. https://doi.org/10.1016/j.ultras.2014.03.015
https://doi.org/10.1016/j.ultras.2014.03...
, 3030. Kang KL, Kim EC, Park JB, Heo JS, Choi Y. High-frequency, low-intensity pulsed ultrasound enhances alveolar bone healing of extraction sockets in rats: a pilot study. Ultrasound Med Biol. 2016 Feb;42(2):493-502. https://doi.org/10.1016/j.ultrasmedbio.2015.10.022
https://doi.org/10.1016/j.ultrasmedbio.2...
In addition, LIPUS is expected to prevent root resorption and accelerate its repair.3131. Dalla-Bona DA, Tanaka E, Inubushi T, Oka H, Ohta A, Okada H, et al. Cementoblast response to low- and high-intensity ultrasound. Arch Oral Biol. 2008 Apr;53(4):318-23. https://doi.org/10.1016/j.archoralbio.2007.11.006
https://doi.org/10.1016/j.archoralbio.20...
Based on these findings, LIPUS has been considered as a promising therapeutic tool for periodontal regeneration.

We hypothesized that LIPUS might promote the osteogenic differentiation of DFCs and enhance the regenerative effects of the DFC-scaffold complex. Therefore, we investigated the possible effect of LIPUS on the osteogenic differentiation of DFCs in vitro and on tissue regeneration in DFC-OsteoBoneTM scaffolds implanted into the subcutaneous dorsa of the nude mouse.

Methodology

Experimental animals

Six- to 7-day-old Sprague-Dawley (SD) rats and nude mice were provided by the animal center of Chongqing Medical University, Chongqing, China. All animal experiments were approved by the Ethics Committee of Chongqing Medical University.

Separation and cultivation of rat DFC Cells

The isolation and culture of DFCs were performed as previously described.3232. Wise GE, Lin F, Fan W. Culture and characterization of dental follicle cells from rat molars. Cell Tissue Res. 1992 Mar;267(3):483-92. https://doi.org/10.1007/BF00319370
https://doi.org/10.1007/BF00319370...
Dental follicles were carefully separated from the mandibular first molars of 6- to 7-day-old SD rats under a dissecting microscope. The dental follicle tissues were digested with 1 milligram per milliliter of type I collagenase (Sigma, Shangai, China) in PBS at 37°C for 30 min with frequent gentle agitation. After they were fully digested, the dental follicles and cell suspensions were centrifuged at 800 r/min for 5 min. The supernatant was discarded, and the cell pellet was resuspended in complete medium (containing 10% fetal bovine serum (FBS)) and seeded into 25 T-flasks (Corning, Shangai, China). The cultures were incubated at 37°C in a humidified atmosphere with 5% CO2 in air. The cell culture medium was changed every 2 days. The DFCs were passaged at a 1:2 ratio until they reached approximately 70% confluence. The cells were passaged for purification.

Flow cytometric surface marker expression analysis

Flow cytometric analyses were performed to measure the expression of mesenchymal stem cell-associated surface markers. DFCs from the third passage were digested and washed twice with 1% FBS in PBS, followed by incubation with anti-rat CD34 (FITC), anti-rat CD73 (PE), anti-rat CD146 (PE), and anti-rat STRO-1 (FITC) antibodies in the dark for 20 min according to the manufacturer’s protocol (BD Biosciences, USA). After washing, flow cytometry was used to analyze the stained cells.

Osteogenic, adipogenic and chondrogenic differentiation

DFCs were seeded at a density of 1×105 cells per well in 6-well plates. After reaching 80% confluence, the DFCs were cultured in osteogenic differentiation medium (Biowit, China), adipogenic differentiation medium (Biowit, Shenzhen, China) or chondrogenic differentiation medium (Biowit, China). The cell culture medium was changed every 2 days. After 21 days of osteogenic induction, the samples were washed gently with PBS 3 times, fixed with 4% polyoxymethylene for 20 min, and then stained at room temperature for 1 h with Alizarin Red solution (Beyotime, Shangai, China) to assess mineral deposition. After 2 weeks of adipogenic induction, the differentiated cells were fixed for 20 min and then stained at room temperature for 1 h with 0.3% Oil Red O solution (Sigma, Shangai, China) to evaluate adipogenesis. After 21 days of chondrogenic induction, the cells were fixed and stained at room temperature for 30 min with 1% Alcian blue solution (Sigma. USA) to evaluate chondrogenesis.

Exposure to LIPUS

The LIPUS exposure devices were provided by the National Engineering Research Center of Ultrasound Medicine (Chongqing Medical University, Chongqing, China). As described in our previous studies,2424. Hu B, Zhang Y, Zhou J, Li J, Deng F, Wang Z, et al. Low-intensity pulsed ultrasound stimulation facilitates osteogenic differentiation of human periodontal ligament cells. PLoS One. 2014 Apr;9(4):e95168. https://doi.org/10.1371/journal.pone.0095168
https://doi.org/10.1371/journal.pone.009...
, 3333. Wang Y, Qiu Y, Li J, Zhao C, Song J. Low-intensity pulsed ultrasound promotes alveolar bone regeneration in a periodontal injury model. Ultrasonics. 2018 Nov;90:166-72. https://doi.org/10.1016/j.ultras.2018.06.015
https://doi.org/10.1016/j.ultras.2018.06...
, 3434. Yang Z, Ren L, Deng F, Wang Z, Song J. Low-intensity pulsed ultrasound induces osteogenic differentiation of human periodontal ligament cells through activation of bone morphogenetic protein-smad signaling. J Ultrasound Med. 2014 May;33(5):865-73. https://doi.org/10.7863/ultra.33.5.865
https://doi.org/10.7863/ultra.33.5.865...
the parameters for LIPUS were as follows: an intensity of 90 mW/cm22. Jin LJ, Armitage GC, Klinge B, Lang NP, Tonetti M, Williams RC. Global oral health inequalities: task group—periodontal disease. Adv Dent Res. 2011 May;23(2):221-6. https://doi.org/10.1177/0022034511402080
https://doi.org/10.1177/0022034511402080...
, a frequency of 1.5 MHz, a pulse duration of 200 µs, a repetition rate of 1.0 KHz, and a process time of 20 min/day. For the in vitro study, the induction of osteogenic differentiation in the DFCs was performed in an osteogenic medium as described above. The culture plates from the LIPUS group (LIPUS (+) group) were placed on the ultrasound transducer with a thin layer of water to maintain contact at 37°C. For the in vivo study, the mice were fixed, and the ultrasound transducer was placed into contact with the skin of the target region. To ensure favorable ultrasonic transmission, a coupling gel was used between the transducer and the skin.

Real-time polymerase chain reaction (RT-PCR)

After 7 days of treatment, the expression of osteogenesis-related genes (ALP, Runx2, OSX, and COL-I) was determined by RT-PCR. Briefly, the total RNA in the cells was isolated using a MiniBEST Universal RNA Extraction Kit (Takara, Dalian, China), and reverse transcription was performed using a PrimeScript RT Reagent Kit with gDNA Eraser (Takara, Dailian, China) to produce complementary deoxyribonucleic acid (cDNA). The RT-PCR was performed using a FX9600 Connect Real-Time PCR Detection System (Bio-Rad, Hercules, USA) under the following conditions: the reaction mixture was preheated for 15 min at 96°C to activate the Taq enzyme and denatured for 5 min at 96°C, which was followed by 40 cycles of 30 s at 96°C, 30 s at 57°C and 30 s at 72°C. For each reaction, a melting curve was generated to test for primer dimer formation and nonspecific priming. The sequences of the primers and probes are listed in Table . The quantification was performed with the 2-ΔΔCt method.

Table 1
Primer sequences used for quantitative real-time polymerase chain reaction.

Osteogenic analysis

After 21 days of treatment, the formation of mineralized nodules in the DFCs was examined by Alizarin red staining. To quantify the mineralization, the cells were stained with Alizarin Red and were then destained with 10% cetylpyridinium chloride, monohydrate (Solarbio, Beijing, China). Then, the extracted stain was transferred to a 96-well plate, and the absorbance at 562 nm was measured using a microplate reader (Perkin Elmer, Waltham, USA).

Seeding of DFCs onto the OsteoBoneTM scaffold

The DFCs were collected and seeded onto the OsteoBoneTM scaffold (Yenssen Biotech, Jiangsu, China) prior to the subsequent morphological analysis and subcutaneous implantation. Briefly, the DFCs were detached from the dishes and centrifuged to remove the supernatant. Then, the cells were resuspended in culture medium supplemented with FBS at a final density of 5×105 cells per milliliter. One milliliter of cell suspension was gently added to the OsteoboneTM scaffold and wetted with culture medium every two hours for a total of three times. The cell-scaffold complexes were further cultured in complete medium in a humidified atmosphere with 5% CO2 at 37°C.

Scanning electron microscopy (SEM)

Cell attachment and spreading on the scaffolds were visually assessed after 3, 5, 7, and 9 days. At every time point, the specimens were washed with PBS twice, fixed with 2% glutaraldehyde at 4°C for 12 h, dehydrated with a graded ethanol series, dried in a critical point dryer, coated with conductive material, and finally observed under a scanning electron microscope (SEM, Hitachi, S-3000N, Tokyo, Japan).

Histological analysis of the in vivo experiments

To evaluate the combined effect of LIPUS and the OsteoBoneTM scaffold on the osteogenic differentiation of DFCs, either the scaffold alone or the cell (DFC) scaffold complex was transplanted into the subcutaneous dorsa of a nude mouse with or without LIPUS stimulation. A total of nine nude mice were randomly assigned to one of three groups: group A, empty scaffold; group B, DFCs + scaffold; group C, DFCs + scaffold + LIPUS. All nude mice were placed under general anesthesia by intraperitoneal injection of 10% chloral hydrate prior to surgical procedures, followed by local anesthesia with 0.5 milliliter of 1% lidocaine with epinephrine (1:100000). In group A, the empty scaffold material was implanted into the subcutaneous dorsa of the nude mouse at the muscle surface and sutured. In group B, DFCs were seeded onto the OsteoBoneTM scaffold and cultured for 9 days, and the cell scaffold complex was then implanted into the subcutaneous dorsa of the nude mice. In group C, following 9 days of culture in vitro, the cell scaffold complex was implanted into the subcutaneous dorsa of the nude mice and treated with LIPUS after wound healing. The nude mice were sacrificed 8 weeks after surgery. The specimens were sectioned and fixed with 4% polyoxymethylene at 4°C for 24 h. The transplants were decalcified with 10% EDTA at pH 8.0, dehydrated with a graded ethanol series, and then embedded in paraffin. Five microliter-thick paraffin sections were prepared for HE and Mason staining. All samples were examined under a compound microscope (Olympus, Tokyo, Japan).

Statistical analysis

The data are expressed as the mean ± standard deviation. SPSS version 19 (IBM SPSS, Armonk, USA) was used to analyze the data. The statistical analysis was performed using a two-tailed Student’s t-test, and P<0.05 was considered to indicate a statistically significant difference.

Results

Morphology and identification of rat DFC Cells

After subculturing, the rat DFCs exhibited the typical spindle shape of mesenchymal cells ( Figure 1A ). The expression of markers at the DFC surface determined by flow cytometric analysis at passage 3 is presented in Figure 1B . The DFCs were positive for CD73, CD146 and STRO-1 and were negative for CD34. These data indicate that rat DFCs have characteristics of mesenchymal cells. After induction in osteogenic, adipogenic and chondrogenic media, the differentiated cells were analyzed for the formation of mineralized nodules, the accumulation of lipid clusters and the production of proteoglycan ( Figure 1C ). The formation of calcium mineralized nodules stained with Alizarin red solution was observed in adherent cell cultures of rat DFCs. The intercellular formation of lipid droplets, as indicated by Oil Red O staining, was clearly exhibited in rat DFCs; proteoglycan was also found in the DFCs.

Figure 1
A: Morphological characteristics of DFCs at the third passage were observed under a light-inverted microscope. DFCs exhibit typical fibroblast-like spindle morphology. B: Flow cytometric analysis indicated that the DFCs were of mesenchymal origin (positive for CD73 and negative for CD34) and that the DFCs were stem cells (positive for CD146 and STRO-1). C: Osteogenic (Os), adipogenic (Ad) and chondrogenic (Ch) differentiation of DFCs.

Effects of LIPUS on the osteogenic differentiation of rat DFCs

The RT-PCR results showed that rat DFCs exposed to LIPUS for 7 days expressed significantly (p < 0.05) higher levels of ALP, Runx2, OSX, and COL-I mRNA compared with those of DFCs from the non-LIPUS treatment groups ( Figure 2A ). We also found that the formation of mineralized nodules significantly increased with LIPUS stimulation, resulting in nodules with observably stronger Alizarin red staining ( Figure 2B, C ).

Figure 2
Effect of LIPUS stimulation on the osteogenic differentiation of DFCs. The cells were cultured in the presence or absence of LIPUS stimulation in an osteogenic medium. (−): control; (+): LIPUS stimulation. A: RT-PCR analysis of the expression levels of ALP, COL-1, OSX and Runx2 in DFC and BMSC cultures after 7 days of induced differentiation. B: The formation of mineralized nodules in DFCs was examined by staining with Alizarin red on day 21 of culture. C: The mineralization was quantified by destaining cells stained with alizarin red. P< 0.05 for LIPUS-treated versus control cells.

Adhesion and spreading of DFCs on the material

Under the scanning electron microscope, we observed that the DFCs could adhere and grow on the surfaces of the porous scaffolds ( Figure 3 ). After 3 days of culturing, a small number of cells was observed on the surface of the material. The number of cells increased slightly after 5 days and increased significantly after 7 days of culture. After 9 days of culture, many cells were observed to have spread on the surface and to have grown into the pore. Therefore, these results suggest that the scaffold facilitated the adhesion and spread of the DFCs onto its surface. The cell scaffold complex on day 9 of incubation was selected for in vivo implantation.

Figure 3
Scanning electron micrographs of DFCs cultured on a 3D OsteoBoneTM scaffold. The DFCs were seeded on scaffolds after 3, 5, 7, and 9 days.

Histological analyses of the in vivo experiments.

All nude mice recovered well after surgery. The effect of LIPUS on the regeneration of the OsteoBoneTM-DFC complex was evaluated after 8 weeks of subcutaneous implantation in nude mice by histological analysis using HE staining and Mason staining. In group A (empty scaffolds), we did not observe any tissue formation or vascularization inside the implants ( Figure 4A ). However, the scaffolds seeded with DFCs (group B) exhibited obvious fibers and vessels that invaded the scaffold ( Figure 4B ). Moreover, group C also showed a significant increase in newly formed fibers and vessels ( Figure 4C ).

Figure 4
Histological examination of the harvested complexes at 8 weeks. A: empty scaffold; B: DFCs + scaffold; C: DFCs + scaffold + LIPUS. C, cell; F, newly formed fibrous tissue; BV, newly formed blood vessels.

Discussion

DFCs have the capability for self-renewal and the potential to differentiate into multiple cell types, and they have been used in stem cell-based periodontal tissue engineering.3535. Chen FM, Sun HH, Lu H, Yu Q. Stem cell-delivery therapeutics for periodontal tissue regeneration. Biomaterials. 2012 Sep;33(27):6320-44. https://doi.org/10.1016/j.biomaterials.2012.05.048
https://doi.org/10.1016/j.biomaterials.2...
More importantly, DFCs are one of the most accessible sources of seed cells. Therefore, we used DFCs as seed cells in our study. Simultaneously, the OsteoBoneTMscaffold was used as a carrier for seed cells because it is an inorganic porous and biomimetic 3D scaffold that has a distribution of spatial pores of 100-300 µm in diameter and interporous channels of 350-500 µm in diameter. Its main components are calcium, silicon, and phosphorus without organic components, and the scaffold has been shown to exhibit better biocompatible and degradable properties.3636. Sun XJ, Zhang ZY, Wang SY, Gittens SA, Jiang XQ, Chou LL. Maxillary sinus floor elevation using a tissue-engineered bone complex with OsteoBone and bMSCs in rabbits. Clin Oral Implants Res. 2008 Aug;19(8):804-13. https://doi.org/10.1111/j.1600-0501.2008.01577.x
https://doi.org/10.1111/j.1600-0501.2008...
Although growth factor is one of the key components of cell-based tissue engineering technology, we did not use it in our study. Instead, we used LIPUS to promote the osteogenic differentiation of DFCs and the regeneration of the DFC-scaffold complex, as LIPUS has been widely used as a type of mechanical stimulation for therapies in various medical fields, especially for the promotion of new bone formation.3737. Suzuki A, Takayama T, Suzuki N, Sato M, Fukuda T, Ito K. Daily low-intensity pulsed ultrasound-mediated osteogenic differentiation in rat osteoblasts. Acta Biochim Biophys Sin (Shanghai). 2009 Feb;41(2):108-15. https://doi.org/10.1093/abbs/gmn012
https://doi.org/10.1093/abbs/gmn012...

Based on gene expression levels, our experiments demonstrated that the expression of the ALP, Runx2, OSX and COL-I mRNAs increased significantly after 7 days of LIPUS exposure. These findings are consistent with those of previous studies showing that LIPUS promotes osteogenic differentiation by upregulating the expression of osteogenesis-related genes in PDLCs,2424. Hu B, Zhang Y, Zhou J, Li J, Deng F, Wang Z, et al. Low-intensity pulsed ultrasound stimulation facilitates osteogenic differentiation of human periodontal ligament cells. PLoS One. 2014 Apr;9(4):e95168. https://doi.org/10.1371/journal.pone.0095168
https://doi.org/10.1371/journal.pone.009...
human alveolar-derived mesenchymal stem cells3838. Lim K, Kim J, Seonwoo H, Park SH, Choung PH, Chung JH. In vitro effects of low-intensity pulsed ultrasound stimulation on the osteogenic differentiation of human alveolar bone-derived mesenchymal stem cells for tooth tissue engineering. Biomed Res Int. 2013;2013:269724. https://doi.org/10.1155/2013/269724. Epub
https://doi.org/10.1155/2013/269724...
and even adipose-derived stem cells.3939. Zhang Z, Ma Y, Guo S, He Y, Bai G, Zhang W. Low-intensity pulsed ultrasound stimulation facilitatesin vitroosteogenic differentiation of human adipose-derived stem cells via up-regulation of heat shock protein (HSP)70, HSP90, and bone morphogenetic protein (BMP) signaling pathway. Biosci Rep. 2018 May;38(3). https://doi.org/10.1042/BSR20180087
https://doi.org/10.1042/BSR20180087...
Our findings thus provide evidence at the molecular level that explains the LIPUS-promoted osteogenesis of DFCs. Runx2 is an important transcription factor involved in the process of osteogenic differentiation because it activates osteoblast-specific genes, such as COL-I and ALP.4040. Yang Y, Yang Y, Li X, Cui L, Fu M, Rabie AB, et al. Functional analysis of core binding factor a1 and its relationship with related genes expressed by human periodontal ligament cells exposed to mechanical stress. Eur J Orthod. 2010 Dec;32(6):698-705. https://doi.org/10.1093/ejo/cjq010
https://doi.org/10.1093/ejo/cjq010...
Li et al.4141. Li L, Han M, Li S, Wang L, Xu Y. Cyclic tensile stress during physiological occlusal force enhances osteogenic differentiation of human periodontal ligament cells via ERK1/2-Elk1 MAPK pathway. DNA Cell Biol. 2013 Sep;32(9):488-97. https://doi.org/10.1089/dna.2013.2070
https://doi.org/10.1089/dna.2013.2070...
found that cyclic tensile stress promoted the osteogenic differentiation of periodontal ligament cells via activation of the ERK1/2-Elk1 MAPK pathway and the upregulation of Runx2. Previous studies by our research team found that the p38-MAPK4242. Ren L, Yang Z, Song J, Wang Z, Deng F, Li W. Involvement of p38 MAPK pathway in low intensity pulsed ultrasound induced osteogenic differentiation of human periodontal ligament cells. Ultrasonics. 2013 Mar;53(3):686-90. https://doi.org/10.1016/j.ultras.2012.10.008
https://doi.org/10.1016/j.ultras.2012.10...
pathway and the BMP-Smad3434. Yang Z, Ren L, Deng F, Wang Z, Song J. Low-intensity pulsed ultrasound induces osteogenic differentiation of human periodontal ligament cells through activation of bone morphogenetic protein-smad signaling. J Ultrasound Med. 2014 May;33(5):865-73. https://doi.org/10.7863/ultra.33.5.865
https://doi.org/10.7863/ultra.33.5.865...
pathway were involved in the process of LIPUS-induced osteogenic differentiation of periodontal ligament cells. We speculated that LIPUS probably promoted the osteogenic differentiation of DFCs via the MAPK-related and BMP-Smad pathways. However, this speculation needs to be verified, and further studies are necessary to clarify how LIPUS activates the MAPK-related and BMP-Smad pathways.

In addition, we examined the formation of mineralized nodules in the presence or absence of daily LIPUS stimulation. We found that the formation of mineralized nodules significantly increased in the presence of LIPUS stimulation, resulting in nodules with observably stronger Alizarin red staining. These results show that LIPUS stimulation may promote the formation of bone by DFCs, which is in accordance with the gene expression results.

Before implantation, we determined whether the scaffold we selected was biocompatible with DFCs. We observed that DFCs spread along the OsteoBoneTMscaffold surface and grew into the pores of the scaffold after being cultured in vitro. These results suggest that OsteoBoneTM has good biocompatibility and is favorable for the adhesion and growth of DFCs. Sun et al. observed similar results when seeding rabbit BMSCs on an OsteoBoneTMscaffold.1313. Sun XJ, Xia LG, Chou LL, Zhong W, Zhang XL, Wang SY, et al. Maxillary sinus floor elevation using a tissue engineered bone complex with BMP-2 gene modified bMSCs and a novel porous ceramic scaffold in rabbits. Arch Oral Biol. 2010 Mar;55(3):195-202. https://doi.org/10.1016/j.archoralbio.2010.01.006
https://doi.org/10.1016/j.archoralbio.20...
Furthermore, the best conditions in terms of growth and adhesion on the scaffold surface were observed after the cells were cultured for 9 days. Therefore, we selected 9 days of culture as optimal for transplantation in the subsequent animal experiments.

Finally, we conducted an in vivo study to investigate whether LIPUS can enhance the regenerative effects of DFC-OsteoBoneTM complexes implanted into the subcutaneous dorsa of nude mice. Histologically, we observed no new tissue formation in the empty scaffold group, which indicated that the OsteoBoneTM scaffold alone was not capable of promoting tissue regeneration in a subcutaneous implant model in nude mice. In the DFCs + scaffold group, we observed more obvious newly formed tissues in the scaffold than in the empty scaffold group. This result demonstrated the indispensability of seed cells in the regenerative process. As expected, the DFCs + scaffold + LIPUS group showed the most obvious new tissue formation in the scaffold. In other words, LIPUS could further promote the regenerative effects of the OsteoBoneTM-DFC complex. It is worth noting that the regenerated tissues contained some cells, fibrous tissue and blood vessels without obvious newly formed osteoid, which was not in full accordance with the results of the study that found that LIPUS may promote the osteogenic differentiation of DFCs in vitro. Therefore, further studies should be conducted to clarify the factors that influence subcutaneous heterotopic osteogenesis and to select the most optimized parameters.

Conclusion

Based on these findings, we conclude that LIPUS may promote the osteogenetic differentiation of rat DFCs by increasing the mRNA expression of osteogenesis-related genes and the formation of mineralized nodules in vitro. LIPUS may enhance the regenerative effect of a DFC-OsteoBoneTMscaffold complex implanted into the subcutaneous dorsa of a nude mouse, although obvious newly formed osteoid was not observed in the histological sections. Further studies should be conducted to clarify the underlying mechanisms of LIPUS and to select the most optimized parameters to make the best use of cell-based tissue engineering.

Acknowledgements

This research was supported by the National Natural Science Foundation of China (grants 81570981, 31700850 and 31600788), the Science and Technology Research Program of the Chongqing Education Bureau (grant KJ1500241), the Yubei District science and Technology Commission grand (2015(19) and 2017(36)), the Postgraduate Students’ Innovative Research and Development Projects of Chongqing, China (grant CYS16132), the Program for Innovation Team Building at Institutions of Higher Education in Chongqing in 2016 (grant CXTDG201602006), the Chongqing Research Program of Basic Research and Frontier Technology (grant cstc2016jcyjA0199) and the Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education.

References

  • 1
    Hynes K, Menicanin D, Gronthos S, Bartold PM. Clinical utility of stem cells for periodontal regeneration. Periodontol 2000. 2012 Jun;59(1):203-27. https://doi.org/10.1111/j.1600-0757.2012.00443.x
    » https://doi.org/10.1111/j.1600-0757.2012.00443.x
  • 2
    Jin LJ, Armitage GC, Klinge B, Lang NP, Tonetti M, Williams RC. Global oral health inequalities: task group—periodontal disease. Adv Dent Res. 2011 May;23(2):221-6. https://doi.org/10.1177/0022034511402080
    » https://doi.org/10.1177/0022034511402080
  • 3
    Baelum V, López R. Periodontal disease epidemiology: learned and unlearned? Periodontol 2000. 2013 Jun;62(1):37-58. https://doi.org/10.1111/j.1600-0757.2012.00449.x
    » https://doi.org/10.1111/j.1600-0757.2012.00449.x
  • 4
    Racz GZ, Kadar K, Foldes A, Kallo K, Perczel-Kovach K, Keremi B, et al. Immunomodulatory and potential therapeutic role of mesenchymal stem cells in periodontitis. J Physiol Pharmacol. 2014 Jun;65(3):327-39.
  • 5
    Bosshardt DD, Sculean A. Does periodontal tissue regeneration really work? Periodontol 2000. 2009;51(1):208-19. https://doi.org/10.1111/j.1600-0757.2009.00317.x
    » https://doi.org/10.1111/j.1600-0757.2009.00317.x
  • 6
    Lin N-H, Gronthos S, Bartod PM. Stem cells and periodontal regeneration. Aust Dent J. 2010;53(2):108-21. https://doi.org/10.1111/j.1834-7819.2008.00019.x
    » https://doi.org/10.1111/j.1834-7819.2008.00019.x
  • 7
    Bartold PM, McCulloch CA, Narayanan AS, Pitaru S. Tissue engineering: a new paradigm for periodontal regeneration based on molecular and cell biology. Periodontol 2000. 2000 Oct;24(1):253-69. https://doi.org/10.1034/j.1600-0757.2000.2240113.x PMID:11276871
    » https://doi.org/10.1034/j.1600-0757.2000.2240113.x
  • 8
    Becker W, Becker BE. Periodontal regeneration: a contemporary re-evaluation. Periodontol 2000. 1999 Feb;19(1):104-14. https://doi.org/10.1111/j.1600-0757.1999.tb00150.x
    » https://doi.org/10.1111/j.1600-0757.1999.tb00150.x
  • 9
    Karring T, Nyman S. Gottlow J, Laurell L. Development of the biological concept of guided tissue regeneration: animal and human studies. Periodontol. 2000. 2010;1:26-35. https://doi.org/10.1111/j.1600-0757.1993.tb00204.x
    » https://doi.org/10.1111/j.1600-0757.1993.tb00204.x
  • 10
    Catón J, Bostanci N, Remboutsika E, De Bari C, Mitsiadis TA. Future dentistry: cell therapy meets tooth and periodontal repair and regeneration. J Cell Mol Med. 2011 May;15(5):1054-65. https://doi.org/10.1111/j.1582-4934.2010.01251.x
    » https://doi.org/10.1111/j.1582-4934.2010.01251.x
  • 11
    Baldwin J, Antille M, Bonda U, De-Juan-Pardo EM, Khosrotehrani K, Ivanovski S, et al. In vitro pre-vascularisation of tissue-engineered constructs A co-culture perspective. Vasc Cell. 2014 Jun;6(1):13. https://doi.org/10.1186/2045-824X-6-13
    » https://doi.org/10.1186/2045-824X-6-13
  • 12
    Kemppainen JM, Hollister SJ. Tailoring the mechanical properties of 3D-designed poly(glycerol sebacate) scaffolds for cartilage applications. J Biomed Mater Res A. 2010 Jul;94(1):9-18. https://doi.org/10.1002/jbm.a.32653
    » https://doi.org/10.1002/jbm.a.32653
  • 13
    Sun XJ, Xia LG, Chou LL, Zhong W, Zhang XL, Wang SY, et al. Maxillary sinus floor elevation using a tissue engineered bone complex with BMP-2 gene modified bMSCs and a novel porous ceramic scaffold in rabbits. Arch Oral Biol. 2010 Mar;55(3):195-202. https://doi.org/10.1016/j.archoralbio.2010.01.006
    » https://doi.org/10.1016/j.archoralbio.2010.01.006
  • 14
    Monsarrat P, Vergnes JN, Nabet C, Sixou M, Snead ML, Planat-Bénard V, et al. Concise review: mesenchymal stromal cells used for periodontal regeneration: a systematic review. Stem Cells Transl Med. 2014 Jun;3(6):768-74. https://doi.org/10.5966/sctm.2013-0183
    » https://doi.org/10.5966/sctm.2013-0183
  • 15
    Honda MJ, Imaizumi M, Tsuchiya S, Morsczeck C. Dental follicle stem cells and tissue engineering. J Oral Sci. 2010 Dec;52(4):541-52. https://doi.org/10.2334/josnusd.52.541
    » https://doi.org/10.2334/josnusd.52.541
  • 16
    MI C, Garant PR. Development and general structure of the periodontium. Periodontol 2000. 2010 Oct;24(1):9-27. https://doi.org/10.1034/j.1600-0757.2000.2240102.x
    » https://doi.org/10.1034/j.1600-0757.2000.2240102.x
  • 17
    Wu Y, Feng G, Song J, Zhang Y, Yu Y, Huang L, et al. TrAmplification of human dental follicle cells by piggyBac transposon: mediated reversible immortalization system. PLoS One. 2015 Jul;10(7):e0130937. https://doi.org/10.1371/journal.pone.0130937
    » https://doi.org/10.1371/journal.pone.0130937
  • 18
    Miao Z, Lu Z, Luo S, Lei D, He Y, Wu H, et al. Murine and Chinese cobra venom derived nerve growth factor stimulate chondrogenic differentiation of BMSCs in vitro: A comparative study. Mol Med Rep. 2018 Sep;18(3):3341-9. https://doi.org/10.3892/mmr.2018.9307
    » https://doi.org/10.3892/mmr.2018.9307
  • 19
    Ren J, Ward D, Chen S, Tran K, Jin P, Sabatino M, et al. Comparison of human bone marrow stromal cells cultured in human platelet growth factors and fetal bovine serum. J Transl Med. 2018 Mar;16(1):65. https://doi.org/10.1186/s12967-018-1400-3
    » https://doi.org/10.1186/s12967-018-1400-3
  • 20
    Fujita T, Shiba H, Van Dyke TE, Kurihara H. Differential effects of growth factors and cytokines on the synthesis of SPARC, DNA, fibronectin and alkaline phosphatase activity in human periodontal ligament cells. Cell Biol Int. 2004;28(4):281-6. https://doi.org/10.1016/j.cellbi.2003.12.007
    » https://doi.org/10.1016/j.cellbi.2003.12.007
  • 21
    Ferrandis E, Pradhananga SL, Touvay C, Kinoshita C, Wilkinson IR, Stafford K, et al. Immunogenicity, toxicology, pharmacokinetics and pharmacodynamics of growth hormone ligand-receptor fusions. Clin Sci (Lond). 2010 Aug;119(11):483-91. https://doi.org/10.1042/CS20100241
    » https://doi.org/10.1042/CS20100241
  • 22
    Hofmann A, Ritz U, Hessmann MH, Alini M, Rommens PM, Rompe JD. Extracorporeal shock wave-mediated changes in proliferation, differentiation, and gene expression of human osteoblasts. J Trauma. 2008 Dec;65(6):1402-10. https://doi.org/10.1097/TA.0b013e318173e7c2
    » https://doi.org/10.1097/TA.0b013e318173e7c2
  • 23
    Jansen JH, van der Jagt OP, Punt BJ, Verhaar JA, van Leeuwen JP, Weinans H, et al. Stimulation of osteogenic differentiation in human osteoprogenitor cells by pulsed electromagnetic fields: an in vitro study. BMC Musculoskelet Disord. 2010 Aug;11(1):188. https://doi.org/10.1186/1471-2474-11-188
    » https://doi.org/10.1186/1471-2474-11-188
  • 24
    Hu B, Zhang Y, Zhou J, Li J, Deng F, Wang Z, et al. Low-intensity pulsed ultrasound stimulation facilitates osteogenic differentiation of human periodontal ligament cells. PLoS One. 2014 Apr;9(4):e95168. https://doi.org/10.1371/journal.pone.0095168
    » https://doi.org/10.1371/journal.pone.0095168
  • 25
    Rego EB, Takata T, Tanne K, Tanaka E. Current status of low intensity pulsed ultrasound for dental purposes. Open Dent J. 2012;6(1):220-5. https://doi.org/10.2174/1874210601206010220
    » https://doi.org/10.2174/1874210601206010220
  • 26
    Padilla F, Puts R, Vico L, Guignandon A, Raum K. Stimulation of bone repair with ultrasound. Adv Exp Med Biol. 2016 Jan; 880:385-427. https://doi.org/10.1007/978-3-319-22536-4_21
    » https://doi.org/10.1007/978-3-319-22536-4_21
  • 27
    Ikai H, Tamura T, Watanabe T, Itou M, Sugaya A, Iwabuchi S, et al. Low-intensity pulsed ultrasound accelerates periodontal wound healing after flap surgery. J Periodontal Res. 2008 Apr;43(2):212-6. https://doi.org/10.1111/j.1600-0765.2007.01016.x
    » https://doi.org/10.1111/j.1600-0765.2007.01016.x
  • 28
    Gu XQ, Li YM, Guo J, Zhang LH, Li D, Gai XD. Effect of low intensity pulsed ultrasound on repairing the periodontal bone of Beagle canines. Asian Pac J Trop Med. 2014 Apr;7(4):325-8. https://doi.org/10.1016/S1995-7645(14)60049-3
    » https://doi.org/10.1016/S1995-7645(14)60049-3
  • 29
    Wang Y, Chai Z, Zhang Y, Deng F, Wang Z, Song J. Influence of low-intensity pulsed ultrasound on osteogenic tissue regeneration in a periodontal injury model: x-ray image alterations assessed by micro-computed tomography. Ultrasonics. 2014 Aug;54(6):1581-4. https://doi.org/10.1016/j.ultras.2014.03.015
    » https://doi.org/10.1016/j.ultras.2014.03.015
  • 30
    Kang KL, Kim EC, Park JB, Heo JS, Choi Y. High-frequency, low-intensity pulsed ultrasound enhances alveolar bone healing of extraction sockets in rats: a pilot study. Ultrasound Med Biol. 2016 Feb;42(2):493-502. https://doi.org/10.1016/j.ultrasmedbio.2015.10.022
    » https://doi.org/10.1016/j.ultrasmedbio.2015.10.022
  • 31
    Dalla-Bona DA, Tanaka E, Inubushi T, Oka H, Ohta A, Okada H, et al. Cementoblast response to low- and high-intensity ultrasound. Arch Oral Biol. 2008 Apr;53(4):318-23. https://doi.org/10.1016/j.archoralbio.2007.11.006
    » https://doi.org/10.1016/j.archoralbio.2007.11.006
  • 32
    Wise GE, Lin F, Fan W. Culture and characterization of dental follicle cells from rat molars. Cell Tissue Res. 1992 Mar;267(3):483-92. https://doi.org/10.1007/BF00319370
    » https://doi.org/10.1007/BF00319370
  • 33
    Wang Y, Qiu Y, Li J, Zhao C, Song J. Low-intensity pulsed ultrasound promotes alveolar bone regeneration in a periodontal injury model. Ultrasonics. 2018 Nov;90:166-72. https://doi.org/10.1016/j.ultras.2018.06.015
    » https://doi.org/10.1016/j.ultras.2018.06.015
  • 34
    Yang Z, Ren L, Deng F, Wang Z, Song J. Low-intensity pulsed ultrasound induces osteogenic differentiation of human periodontal ligament cells through activation of bone morphogenetic protein-smad signaling. J Ultrasound Med. 2014 May;33(5):865-73. https://doi.org/10.7863/ultra.33.5.865
    » https://doi.org/10.7863/ultra.33.5.865
  • 35
    Chen FM, Sun HH, Lu H, Yu Q. Stem cell-delivery therapeutics for periodontal tissue regeneration. Biomaterials. 2012 Sep;33(27):6320-44. https://doi.org/10.1016/j.biomaterials.2012.05.048
    » https://doi.org/10.1016/j.biomaterials.2012.05.048
  • 36
    Sun XJ, Zhang ZY, Wang SY, Gittens SA, Jiang XQ, Chou LL. Maxillary sinus floor elevation using a tissue-engineered bone complex with OsteoBone and bMSCs in rabbits. Clin Oral Implants Res. 2008 Aug;19(8):804-13. https://doi.org/10.1111/j.1600-0501.2008.01577.x
    » https://doi.org/10.1111/j.1600-0501.2008.01577.x
  • 37
    Suzuki A, Takayama T, Suzuki N, Sato M, Fukuda T, Ito K. Daily low-intensity pulsed ultrasound-mediated osteogenic differentiation in rat osteoblasts. Acta Biochim Biophys Sin (Shanghai). 2009 Feb;41(2):108-15. https://doi.org/10.1093/abbs/gmn012
    » https://doi.org/10.1093/abbs/gmn012
  • 38
    Lim K, Kim J, Seonwoo H, Park SH, Choung PH, Chung JH. In vitro effects of low-intensity pulsed ultrasound stimulation on the osteogenic differentiation of human alveolar bone-derived mesenchymal stem cells for tooth tissue engineering. Biomed Res Int. 2013;2013:269724. https://doi.org/10.1155/2013/269724 Epub
    » https://doi.org/10.1155/2013/269724
  • 39
    Zhang Z, Ma Y, Guo S, He Y, Bai G, Zhang W. Low-intensity pulsed ultrasound stimulation facilitatesin vitroosteogenic differentiation of human adipose-derived stem cells via up-regulation of heat shock protein (HSP)70, HSP90, and bone morphogenetic protein (BMP) signaling pathway. Biosci Rep. 2018 May;38(3). https://doi.org/10.1042/BSR20180087
    » https://doi.org/10.1042/BSR20180087
  • 40
    Yang Y, Yang Y, Li X, Cui L, Fu M, Rabie AB, et al. Functional analysis of core binding factor a1 and its relationship with related genes expressed by human periodontal ligament cells exposed to mechanical stress. Eur J Orthod. 2010 Dec;32(6):698-705. https://doi.org/10.1093/ejo/cjq010
    » https://doi.org/10.1093/ejo/cjq010
  • 41
    Li L, Han M, Li S, Wang L, Xu Y. Cyclic tensile stress during physiological occlusal force enhances osteogenic differentiation of human periodontal ligament cells via ERK1/2-Elk1 MAPK pathway. DNA Cell Biol. 2013 Sep;32(9):488-97. https://doi.org/10.1089/dna.2013.2070
    » https://doi.org/10.1089/dna.2013.2070
  • 42
    Ren L, Yang Z, Song J, Wang Z, Deng F, Li W. Involvement of p38 MAPK pathway in low intensity pulsed ultrasound induced osteogenic differentiation of human periodontal ligament cells. Ultrasonics. 2013 Mar;53(3):686-90. https://doi.org/10.1016/j.ultras.2012.10.008
    » https://doi.org/10.1016/j.ultras.2012.10.008

Publication Dates

  • Publication in this collection
    09 Sept 2019
  • Date of issue
    2019

History

  • Received
    19 Sept 2018
  • Accepted
    8 Apr 2019
  • Reviewed
    26 Apr 2019
Sociedade Brasileira de Pesquisa Odontológica - SBPqO Av. Prof. Lineu Prestes, 2227, 05508-000 São Paulo SP - Brazil, Tel. (55 11) 3044-2393/(55 11) 9-7557-1244 - São Paulo - SP - Brazil
E-mail: office.bor@ingroup.srv.br