Join Us | Latest Articles | Contact

Journal Home


Editorial Board


Archive


Submit to this journal


Current issue

International Journal of Stem cell Research and Therapy





DOI: 10.23937/2469-570X/1410009



Dental Stem Cells Characterization and Bone Regenerative Potential in Oral Medicine

Ricardo Rodrigues*, Bárbara Viana, Ivo Vieira, João Tavares, Renato Lobo, Ana Portela and Mário Vasconcelos


Biomaterials Unit, Faculty of Dental Medicine, University of Porto, Portugal


*Corresponding author: Ricardo Rodrigues, Biomaterials Unit, Faculty of Dental Medicine, University of Porto, 4200-392 Porto, Portugal, Tel: +351912661070, E-mail: ricardomcrodrigues.md@gmail.com
Int J Stem Cell Res Ther, IJSCRT-2-009, (Volume 2, Issue 1), Review Article; ISSN: 2469-570X
Received: April 30, 2015 | Accepted: May 31, 2015 | Published: June 02, 2015
Citation: Rodrigues R, Viana B, Vieira I, Tavares J, Lobo R, et al. (2015) Dental Stem Cells Characterization and Bone Regenerative Potential in Oral Medicine. Int J Stem Cell Res Ther 2:009. 10.23937/2469-570X/1410009
Copyright: © 2015 Rodrigues R, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.



Abstract

The oral cavity has proven to be an important reservoir of stem cells with the advantage of being a non-invasive source. These cells potential for maxillo-facial bone regeneration has been well documented throughout the last five years. This article aims at reviewing and discussing the different types of dental stem cells available in the oral cavity, their lineage characterization and potential applications in oral medicine; cell collection, isolation and preservation; bone regenerative potential comparison between dental stem cells and bone marrow stem cells; and various factors affecting bone regeneration in the use of osteogenic cell groups.


Keywords

Dental stem cells, Osteoinduction, Gene induction, Osteointegration, Stem cells isolation, Mesenchymal stem cells, Bone regeneration, Scaffolds, Regulation factors


Abreviations

MSC: Mesenchymal Stem Cells, DSC: Dental Stem Cells, DPSCs - Dental Pulp Stem Cells, SHEDs: Stem Cells from Exfoliated Deciduous Teeth, SCAPs: Stem Cells from Apical Papilla, DLSCs: Periodontal Ligament Stem Cells, DFPCs: Dental Follicle Precursor Cells, BMMSCs: Bone Marrow Mesenchymal Stem Cells, BMP: Bone Morphogenic Protein, PLLA: Poly L-lactic Acid, ALP: Alkaline Phosphatase, BSP: Bone Sialoprotein, OSX: Ostenix, OPN: Osteopontin, PDGF: Plaquelet Derived Growth Factor, OC: Osteocalcin, Coll-1: Collagen Type I, CP23: Cementum Protein 23, FGF2: Fibroblastic Growth Factor 2, TNFα: Tumoral Necrosis Factor α, VEGF: Vascular Endothelial Growth Factor, NF-κB: Necrosis Factor κB, CEMP1: Cementum Protein 1, HIF1: Hypoxia Induction Factor 1, FGF9: Fibroblastic Growth Factor 9, HA/TCP- Hidroxiapatite + Tri-calcium phosphate


Introduction

Alveolar bone loss is a major concern, especially after tooth extraction. To overcome the alveolar bone loss and to increase support (in order to place dental implants, for example), many bone regenerative substitutes are available, such as: allografts, autografts, xenografts, synthetic biomaterials and osteoactive agents [1].

Calcium phosphate ceramics are one of the most used groups of synthetic bone substitutes [2], because of its similarity to the mineral phase of natural bone, absence of immunogenic reactions, excellent biocompatibility and osteoconductive potential [3].

Although calcium phosphate ceramics contribute to bone healing through osteoconduction, they generally lack osteoinductivity. Due to these limitations, surgeons and researchers have focused on developing alternative therapies, particularly studying the combination of osteoprogenitor cells, like mesenchymal stem cells (MSCs), with bone substitutes to improve their osteogenic properties [4].

Some studies indicate that increasing the number of grafted MSCs, associated with calcium phosphate ceramics biomaterials, result in better bone formation. This might be related to the crucial role of MSCs in osteoinduction [5].

There are many MSCs, such as: bone marrow-derived mesenchymal stem cells, umbilical cord blood-derived mesenchymal stem cells, adipose tissue-derived stem cells, muscle-derived stem cells and dental pulp stem cells [6]. Bone marrow was the most used source of MSCs. However, in recent years, dental stem cells have received extensive attention in the field of bone tissue engineering due to their easy access for dentists and oral surgeons, immunosuppressive properties, high proliferation, and the capacity to differentiate into odontoblasts, cementoblasts, osteoblasts, and other cells to be found in dental tissues [7,8].

The aim of the present review is to analyse the current state of the art about dental stem cells and their potential in oral medicine bone regeneration.


Material and Methods

A systematic search was made in the PubMed database.

The following combination of keywords was used: dental stem cells, osteoinduction, gene induction, osteointegration, stem cells isolation, mesenchymal stem cells, bone regeneration, scaffolds, and regulation factors.

Mainly review articles, clinical trials and case-reports were selected. Time frame of publication date was set from 2010 until 2015, and only papers available in English were included. The papers selection was performed by considering the presence of keywords in the title and abstract, and their relevancy to the research that has been made. Then, each paper was selected individually according to its content.


Results and Discussion

Types of dental stem cells

Dental tissues have been considered a potential source of MSC-like populations [9].

With the exception of enamel, which lacks ameloblasts or other cellular elements following tooth development, the periodontium and dentine continue to retain some regenerative or reparative capacities [10].

Dental MSCs are indicated for the regeneration of the dentin-pulp complex, bone, cartilage and neuronal tissues, among others [10,11]. Until recently, five different populations have been isolated and characterized in postnatal dental tissues and classified according to the tissue of origin: dental pulp stem cells (DPSCs), stem cells from exfoliated deciduous teeth (SHEDs), stem cells from apical papilla (SCAPs), periodontal ligament stem cells (PDLSCs) and dental follicle precursor cells (DFPCs) [9,12] (Table 1).


click here
Table 1: Therapeutic application of oral MSCs View Table 1


Dental pulp stem cells (DPSCs): DPSCs are multipotent stem cells, with ectomesenchymal origin, which have advantages for clinical applications when compared to other mesenchymal stem cells derived from bone marrow, adipose tissue, peripheral blood, and umbilical cord blood. They are easily available from discarded teeth after extraction and they can be cryopreserved, retaining their multipotent differentiation ability [9,13].

The differentiation potential of DPSCs from natal teeth to adipogenic, osteogenic, chondrogenic, myogenic and neural-glial cells lines was also shown, and an intrinsic tendency of these cells to differentiate towards osteoblasts has been demonstrated [13].

Sometimes DPSCs are referred to as odontoblastoid cells, because they appear to synthesize and secrete dentin matrix like the odontoblast that they replace [14]. When transplanted in vivo (using immunocompromised mice as hosts), the cells derived from dental pulp generated functional dental tissue in the form of complexes like dentine/pulp. This might be due to the origin of their development as neural crest derived cells [12]. When cultured in ceramic substrates, such as hydroxyapatite or tricalcium phosphate, the cells are able to form bone, dentin and cementum-like tissues [9].

Stem cells from exfoliated deciduous teeth (SHEDs): SHED is a heterogeneous multicellular population of stem cells [15] identified as highly proliferative clonogenic cells capable of differentiating into a variety of cell types including neural cells, adipocytes and odontoblasts [12]. SHED cells represent a population of multipotent stem cells that are more immature than the DPSCs and, in vivo, they seem to be able to induce bone or dentin formation [13,14].

Also, SHED cells exhibit higher proliferation rates, increased population doublings, osteoinductive capacity and ability to form sphere-like clusters when compared to DPSCs [12].

Stem cells from apical papilla (SCAPs): The cells located in the apical papilla (root foramen area) represent another unique population of dental stem cells [12]. As long as the root develops, the dental papilla's location moves in an apical direction [13]. These cells show ability to differentiate into cells of the osteogenic, odontogenic, adipogenic, and neurogenic lineages [9]. It is thought that SCAPs may be responsible for the formation of primary odontoblasts that account for the formation of root dentin, whereas DPSCs seem to be the source of replacement odontoblasts that produce reparative dentine [9,12,13]. To conclude, SCAPs are derived from a developing tissue that may represent a population of early stem/progenitor cells, which may be a superior cell source for tissue regeneration [13], since they also showed other favourable characteristics, such as higher proliferative rate, telomerase activity and improved migration capacity [14-16].

Periodontal ligament stem cells (PDLSCs): The periodontal ligament provides nourishment to the teeth, regulates periodontal homeostasis and contains a population of progenitor cells - periodontal ligament stem cells [12]. PDLSCs are capable of differentiating into cells resembling cementoblasts, osteoblasts, adipocytes, chondrocytes, and fibroblasts [9]. This population seems to be more proliferative, have a faster cell growth rate and higher clonogenic capability than bone marrow mesenchymal stem cells (BMMSCs) [9,16]. These cells maintain their tissue regenerative potential even after recovery from frozen human tissue, which suggests the possibility of cryopreserved PDLSCs from extracted teeth being used for future therapeutic purposes [12].

Dental follicle precursor cells (DFPCs): Dental follicle stem cells exist in the dental follicle (loose connective tissue sac surrounding the enamel organ and the dental papilla of the developing tooth germ before eruption) [9,12]. These cells were first isolated from the follicle of human impacted third molars [14]. In vitro, following adequate induction, DFPCs have been demonstrating osteogenic, odontogenic and cementogenic differentiation capacity [9].

Gingiva as a source of stem cell: Progenitor cells and multipotent MSC subpopulation of cells have been isolated and characterised from gingival fibroblasts. These fibroblasts are easily accessible and have recently been used to derive induced pluripotent stem cell lines [12].


Collection, isolation and preservation of dental stem cells

Collection: The main source of dental stem cells is extracted teeth. A freshly extracted tooth is transferred into a vial containing transport solution and generally hypotonic phosphate buffered saline solution. The vial is carefully sealed and placed into a thermette and then the carrier is placed into an insulated metal transport vessel. The thermette, along with the insulated transport vessel, maintains the sample in a hypothermic state during transportation. This procedure is described as sustentation. The time from harvesting to arrival at the processing storage facility should not exceed 40 hours [17].

DPSCs samples are obtained from dental pulp tissue from extracted third molars, exfoliating/extracted deciduous teeth and teeth extracted for orthodontic treatment, trauma or periodontal disease [18,19], while PDLSCs are obtained via scraping the mid third of the root in extracted teeth. SCAPs are isolated from the apical papilla tissue and can easily be removed from an extracted tooth with developing roots (very common in third molar extractions) [19-23].

Isolation: Almost every author uses a method of their own, which makes a detailed analysis of all experimental works impossible. Consequently, several studies need to be done in order to systematize laboratorial procedures [23].

The two methods most frequently used for DPSCs isolation are Enzyme-digestion and Explant outgrowth. Other methods like Magnetic activated cell sorting (MACS) and Fluorescence activated cell sorting (FACS) are also available [17,24-27].

Regardless of the chosen isolation method, common procedure dictates that the tooth surface is cleaned by washing it three times with phosphate buffered saline (PBSA) without Ca2+ and Mg2+. The next step is disinfection followed by another washing with PBSA. Pulp tissue is isolated from the pulp chamber and placed into a sterile petri dish, washed at least three times with PBSA [17,25,26].

Enzyme-digestion method: Small pieces of pulp tissues are transferred into an enzyme solution, often collagenase/dispase, for 30-60min at 37°C. Afterwards, large cell aggregates are removed and single-cell suspensions are obtained by passing cells through a cell strainer. Single-cell suspensions are centrifuged for 5 min at room temperature. Suspensions are then seeded in culture dishes containing proliferation medium and incubated at 37°C in 5% CO2. Culture medium can be substituted every three days until cell confluence is achieved [17,24-27].

Explant outgrowth method: Pulp tissue is placed in culture flasks with proliferation medium and then incubated at 37°C in 5% CO2. Medium must be changed after outgrowth is observed [17,24-27].

A modified efficient method for dental pulp stem cell isolation: Recently, an additional method has been proposed. The main difference relies on the digestion of pulp tissues and fixing them under a cover slip in the medium. This new method provided a greater in vitro expansion compared to the two methods mentioned above. Moreover, the cells were less damaged and were, therefore, healthy enough to propagate longer in vitro than with other methods [24,26].

Preservation: Once the tissue samples are obtained, they are transferred under proper conditions to a tooth bank where they are stored. The approaches used for stem cell storage are cryopreservation and magnetic freezing [17].

Cryopreservation: Cryopreservation is the process of preserving cells or whole tissues by cooling them at sub-zero temperatures. Liquid nitrogen vapour is used to preserve cells at a temperature below -150°C. Cryopreservation of stem cells maintains the viability of these cells indefinitely [17].

Magnetic freezing: This technology is referred to as cells alive system (CAS), which works on the principle of applying a weak magnetic field to water or cell tissue, which will lower the freezing point of that body by up to 6-7°C. Using CAS, Hiroshima University (first proposed this technology) claims it can increase cell's survival rate in teeth up to 83%. CAS system is a lot less expensive and more reliable than cryogenics [17].


Comparison of human mesenchymal stem cells derived from dental pulp and bone marrow

BMMSCs are a population of multipotent, non-hematopoietic marrow-derived cells that are easily expanded in culture and differentiated into cells with an osteogenic phenotype [19].

Bone marrow was the first identified source of a cell referred as a MSC. These cells were the earliest discovered and well-known stem cell population used in bone tissue engineering [28].

Although they are widely studied and have presented favourable results, there was a need to search other alternative cell sources to overcome the disadvantages that these cells present, specially the morbidity associated to their collection. The main disadvantages of BMMSCs are the low stem cell yield from aspirated bone marrow, painful and highly invasive procedure of collection, as well as the decline, over time, of the number, proliferative capacity and maximal lifespan of MSCs derived from bone marrow [29].

Dental stem cells have emerged in the recent past as an alternative source to MSCs. They are considered multipotent cells with a high proliferation rate, opportunity to be safely cryopreserved, possessing immunosuppressive properties and expressing mesenchymal markers. They also have multi-differentiation ability, easy accessibility, high viability and ready ability to be induced into multiple cell lineages, such as: odontoblasts, adipocytes, neuronal-like cells, glial cells, osteoblasts, chondrocytes, melanocytes, myotubes, and endothelial cells [8,30,31].

PSCs share similar characteristics with BMMSCs. However, it is notable that DPSCs are superior to BMMSCs in certain aspects [32]. These cells have differences in expression of pluripotent stem cell genotype when compared to other types of MSCs. This might reflect their embryonic stem cell origin. Dental pulp is made of ectomesenchymal elements, containing neural crest-derived cells, which display plasticity and multipotential capabilities [5].

Ponnaiyan D et al. concluded that MSCs derived from dental pulp proliferate much faster than MSCs from bone marrow [7]. The authors demonstrated that DPSCs exhibit a significantly higher proliferation rate than BMMSCs and are, therefore, more appropriate for cell-based therapy in clinical application. This result may be due to the age related state of respective tissues, since DPSCs were isolated from third molars (the last permanent teeth to evolve and erupt at an earlier stage of development). In contrast to BMMSCs, DPSCs were more restricted in their adipocyte differentiation capacity, while exhibiting a stronger ability for osteoblast differentiation [30].

Many studies confirm the ability of DPMSCs to undergo osteogenic differentiation and their capacity of forming vascularized adult bone. Integration between the graft and the surrounding host blood supply was achieved [5,21,32,33]. Mori et al. also successfully induced DPSCs into osteoblasts [34].

Although the majority of studies support the advantages of DPSCs over other types of stem cells, there are some which claim opposite conclusions. Asatrian G et al. advocate that while dental pulp might pose as an alternative site for MSCs isolation, the use of DMSCs is limited by the small quantity of pulp in a tooth, by the proportionally smaller DMSC yield required for their expansion by long-term culture and the corresponding costs and risks that are incurred. Thus, according to this study, DPMSCs do not supplant therapies enlisting BMMSCs [35].


Bone tissue regeneration using DSCs - recent advances

In recent years, bone tissue regeneration has proven to be achievable using MSCs from the oral cavity. This is presented as an evolutionary step from common practiced clinical techniques such as the use of rich plasma alone or scaffolds carrying BMPs. Furthermore, bone regeneration is achieved in more significant capacity using stem cells than with previous methods [36]. Therefore, it is important to review the latest developments in the research of factors which may affect stem cells' potential as a biomaterial.


Scaffold architecture and composition

Scaffolds simulate an extracellular matrix in a three dimensional model, as a carrier of signalling molecules aiming towards stimulation, differentiation and migration of cells, ultimately with the purpose of tissue regeneration [16].

Nanofibrous PLLA scaffolds are preferable to Solid Wall ones, as they exhibit higher adherence, proliferation and cellular differentiation values. Also, greater mineral deposition has been achieved in the matrix in vitro. Higher ALP activity, development of dense tissue (detected via sialoprotein concentration) and collagen levels have also been reported in vivo [37].

In recent times, Biocoral Scaffolds comprising DPSCs revealed elevated genomic expression of osteoblastic lines, along with high local values of BSP and osteocalcin. It was possible to observe DPSCs migration towards the inside of Biocoral pores, where the secretion of extracellular matrix and osteoblastic differentiation occurred [38].

Scaffolds HA/TCP carrying PDLSCs corrected furcation defects (Class II and III) combined with cementum and periodontal regeneration, after 12 weeks, in animal models. in vitro studies also verified positive results regarding osteogenesis and in vivo studies equally established favourable clinical and radiographic results. PDLSCs revealed the potential for the development of lamina-dura around titanium implants in vivo [16]. MSC Spheroids, in conjunction with osteoblastic induction agents, over the course of 7 days, have shown accelerated osteogenic properties, higher calcic deposition and bone regeneration in vivo, with significant increase in osteogenic markers concentration (OSX - 8.27x, RUNX2 - 1.57x, OPN - 1.94x, BSP - 1.33x) [39].


Genetic and nutritive regulation

Bone morphogenic proteins have long been utilized in scaffolds as stimulants for a hastened growth of bone matrix and cartilage [40]. Presently, the transfection of genetic factors to cellular populations in scaffolds, namely PDLSCs, resulted in an escalation of PDGF, with greater cementoblastic activity than reported in recombinant PDGF delivery [41].

A mutant variety of RUNX2 (a major transcription factor for bone development and osteologic differentiation) in which the C-terminal 5 amino-acid chain is missing, conveyed higher OC, OP, Coll-1 and CP23 concentrations in scaffolds prepared in vitro. Similar results have been achieved when the cells were stimulated with FGF2. Genetic regulation of cells was obtained through gene introduction via viral vectors and by incorporating the cell medium with genes in the form of recombinant proteins [42]. Bobby Sox has also been studied as a regulator of osteoblastic gene expression in DPSCs, with stem cell proliferation increasing 6.5 times over a period of 7 days when present. BBX variant 1 and 2 cDNA were previously cloned and, afterwards, overexpressed when electroporated in progenitor cells using a microporator. The knockdown of BBX using shRNA did not affect mineralization but a decrease in osteoblastic gene expression was observed. Neither procedure modulated cell proliferation [43].

A shift in miRNA expression in DSC populations undergoing osteoblastic differentiation has been detected with miRNA-218 being associated with mineralized tissue formation and downregulation of RUNX2. MiRNA-218 quantity analysis was conducted using a nanodrop spectrophotometer at baseline (day 0) when the peak expression of RUNX2 was detected [44].


Implant site determinants

Angiogenic and pro-inflammatory responses have been studied as means to regulate DPSCs' proliferation. Short-term exposure to TNFα induces cellular death through an increase in angiogenic signalling (VEGF) and NF-κB, which, once prematurely inhibited, promote mineralization. Long-term exposure prompts cellular proliferation through an increase in anti-apoptotic genes (BCL2) and Survivin [45].

Dexamethasone has been described as having an osteogenic impulse on PDLSC in vitro, vindicated by an upturn in intracellular calcium, ALP and mineralization. Two times more frequently calcified nodules were noted between days 21 and 28 of the experiment conducted, therefore leading to the question of a possible genetic induction being carried out by Dexamethasone [46].

Human serum has been studied as an option to substitute bovine serum as a support for osteomorphic inducting markers for DPSCs, solving possible disease transmission issues and xenogenic immune response, which both may occur when bovine serum is used. Human serum displayed higher cellular proliferation (13% more at each cellular cycle), greater surface expression of typical DPSC antigens, more significant mineral deposition, more ALP activity, and moreover, an increase in surrounding ossification in bone graft implanted in vivo [47].

CEMP1 has been reported as an inducer of cementoblastic phenotypes in PDLSCs. In relative hypoxic states, while the local oxygen tension is reduced, CEMP1 expression rises, leading to further mineral deposition. This mechanism may be regulated by means of HIF1 [48].

DFSCs showed maximum proliferation rates at heat-stress temperatures in the 39°C to 40°C interval, concomitant with higher expression of osteogenic markers, when compared to 37°C physiological temperature [49].


Other studies conducted

Led Nitrate (Pb2+) posed a threat to adhesion of DPSCs and PDLSCs in scaffold matrixes, with changes to cell morphology, higher osteogenic deregulation and cells expressing local markers associated to stem lineages (Oct4, Rex1). Lower proliferation and mineralization rates were also observed [50].

FGF9 induces phosphorylation of ERK1/2, therefore it inhibits osteogenic induction of DPSCs. BMP in scaffold matrix may be able to compensate for this effect, especially since FGF9 also upregulates angiogenesis (VEGF) leading to an ostegenic effect [51].


Conclusion

MSCs are available for extraction from the oral cavity without performing invasive techniques. Various cell populations express stem cell markers, diverse differentiation and clinical application potentials, such as regenerative endodontics and periodontal regeneration among others.

The most deeply studied group of cells with maxilo-facial bone regeneration capacity are DPSCs, SHEDs and PDLSCs. All of these displayed promising both in in-vitro and in-vivo results, especially when compared to other widely utilized stem cell groups such as BMMSC. However, comparisons are still controversial among the scientific community. Other oral MSCs still lack extensive research and so far have not demonstrated much bone regeneration potential. Furthermore, many factors implied in scaffold architecture and composition, gene induction, cell nutrition and general medium exposures regulate cell proliferation, mineralized matrix deposition and osteogenic markers' expression on cellular surface.

Oral stem cell research exhibits a large potential for development, with a broad spectrum of clinical applications to be considered in the coming years.


References
  1. Nazirkar G, Singh S, Dole V, Nikam A (2014) Effortless effort in bone regeneration: a review. J Int Oral Health 6: 120-124.

  2. Hannink G, Arts JJ (2011) Bioresorbability, porosity and mechanical strength of bone substitutes: what is optimal for bone regeneration? Injury 42: S22-25.

  3. Wang L, Zhang B, Bao C, Habibovic P, Hu J, et al. (2014) Ectopic osteoid and bone formation by three calcium-phosphate ceramics in rats, rabbits and dogs. PLoS One 9: e107044.

  4. Corre P, Merceron C, Vignes C, Sourice S, Masson M, et al. (2013) Determining a clinically relevant strategy for bone tissue engineering: an "all-in-one" study in nude mice. PLoS One 8: e81599.

  5. Stanko P, Kaiserova K, Altanerova V, Altaner C (2014) Comparison of human mesenchymal stem cells derived from dental pulp, bone marrow, adipose tissue, and umbilical cord tissue by gene expression. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 158: 373-377.

  6. Seong JM, Kim BC, Park JH, Kwon IK, Mantalaris A, et al. (2010) Stem cells in bone tissue engineering. Biomed Mater 5: 062001.

  7. Ponnaiyan D (2014) Do dental stem cells depict distinct characteristics? - Establishing their "phenotypic fingerprint". Dent Res J (Isfahan) 11: 163-172.

  8. Rodriguez-Lozano FJ, Moraleda JM (2011) Use of dental stem cells in regenerative dentistry: a possible alternative. Trans Res 158: 385-386.

  9. Machado E, Fernandes MH, Gomes Pde S (2012) Dental stem cells for craniofacial tissue engineering. Oral Surg Oral Med Oral Pathol Oral Radiol 113: 728-733.

  10. Han J, Menicanin D, Gronthos S, Bartold PM (2014) Stem cells, tissue engineering and periodontal regeneration. Aust Dent J 59 Suppl 1: 117-130.

  11. Xiao L, Nasu M (2014) From regenerative dentistry to regenerative medicine: progress, challenges, and potential applications of oral stem cells. Stem Cells Cloning 7: 89-99.

  12. Volponi AA, Sharpe PT (2013) The tooth-a -treasure chest of stem cells. Br Dent J 215: 353-358.

  13. Didilescu AC, Rusu MC, Nini G (2013) Dental pulp as a stem cell reservoir. Rom J Morphol Embryol 54: 473-478.

  14. Chandki R, Kala M, Banthia P, Banthia R (2012) From stem to roots: Tissue engineering in endodontics. J Clin Exp Dent 4: e66-71.

  15. Kerkis I, Caplan AI (2012) Stem cells in dental pulp of deciduous teeth. Tissue Eng Part B Rev 18: 129-138.

  16. Saito MT, Silverio KG, Casati MZ, Sallum EA, Nociti FH Jr (2015) Tooth-derived stem cells: Update and perspectives. World J Stem Cells 7: 399-407.

  17. Bansal R, Jain A (2015) Current overview on dental stem cells applications in regenerative dentistry. J Nat Sci Biol Med 6: 29-34.

  18. Shilpa PS, Kaul R, Sultana N, Bhat S (2013) Stem cells: Boon to dentistry and medicine. Dent Res J (Isfahan) 10: 149-154.

  19. Kim SH, Kim YS, Lee SY, Kim KH, Lee YM, et al. (2011) Gene expression profile in mesenchymal stem cells derived from dental tissues and bone marrow. J Periodontal Implant Sci 41: 192-200.

  20. Heo YY, Um S, Kim SK, Park JM, Seo BM (2011) Responses of periodontal ligament stem cells on various titanium surfaces. Oral Dis 17: 320-327.

  21. Verma K, Bains R, Bains VK, Rawtiya M, Loomba K, et al. (2014) Therapeutic potential of dental pulp stem cells in regenerative medicine: An overview. Dent Res J (Isfahan) 11: 302-308.

  22. Lymperi S, Ligoudistianou C, Taraslia V, Kontakiotis E, Anastasiadou E (2013) Dental Stem Cells and their Applications in Dental Tissue Engineering. Open Dent J 7: 76-81.

  23. Giordano G, La Monaca G, Annibali S, Cicconetti A, Ottolenghi L (2011) Stem cells from oral niches: a review. Ann Stomatol (Roma) 2: 3-8.

  24. Derakhshani A, Raoof M, Dabiri S, Farsinejad AR, Gorjestani H, et al. (2015) Isolation and evaluation of dental pulp stem cells from teeth with advanced periodontal disease. Arch Iran Med 18: 211-217.

  25. Karamzadeh R, Eslaminejad MB, Aflatoonian R (2012) Isolation, characterization and comparative differentiation of human dental pulp stem cells derived from permanent teeth by using two different methods. J Vis Exp.

  26. Raoof M, Yaghoobi MM, Derakhshani A, Kamal-Abadi AM, Ebrahimi B, et al. (2014) A modified efficient method for dental pulp stem cell isolation. Dent Res J (Isfahan) 11: 244-250.

  27. Tatullo M, Marrelli M, Shakesheff KM, White LJ (2014) Dental pulp stem cells: function, isolation and applications in regenerative medicine. J Tissue Eng Regen Med.

  28. Knight MN, Hankenson KD (2013) Mesenchymal Stem Cells in Bone Regeneration. Adv Wound Care (New Rochelle) 2: 306-316.

  29. Liao HT, Chen CT (2014) Osteogenic potential: Comparison between bone marrow and adipose-derived mesenchymal stem cells. World J Stem Cells 6: 288-295.

  30. Ponnaiyan D, Jegadeesan V (2014) Comparison of phenotype and differentiation marker gene expression profiles in human dental pulp and bone marrow mesenchymal stem cells. Eur J Dent 8: 307-313.

  31. Tuan RS (2011) Role of adult stem/progenitor cells in osseointegration and implant loosening. Int J Oral Maxillofac Implants 26 Suppl: 50-62.

  32. Wang Y, Yao J, Yuan M, Zhang Z, Hu W (2013) Osteoblasts can induce dental pulp stem cells to undergo osteogenic differentiation. Cytotechnology 65: 223-231.

  33. Kabir R, Gupta M, Aggarwal A, Sharma D, Sarin A, et al. (2014) Imperative role of dental pulp stem cells in regenerative therapies: a systematic review. Niger J Surg 20: 1-8.

  34. Mori G, Centonze M, Brunetti G, Ballini A, Oranger A, et al. (2010) Osteogenic properties of human dental pulp stem cells. J Biol Regul Homeost Agents 24: 167-175.

  35. Asatrian G, Pham D, Hardy WR, James AW, Peault B (2015) Stem cell technology for bone regeneration: current status and potential applications. Stem Cells Cloning 8: 39-48.

  36. Vina JA, El-Alami M, Gambini J, Borras C, Vina J, et al. (2014) Application of mesenchymal stem cells in bone regenerative procedures in oral implantology. A literature review. J Clin Exp Dent 6: e60-65.

  37. Wang J, Ma H, Jin X, Hu J, Liu X, et al. (2011) The effect of scaffold architecture on odontogenic differentiation of human dental pulp stem cells. Biomaterials 32: 7822-7830.

  38. Mangano C, Paino F, d'Aquino R, De Rosa A, Iezzi G, et al. (2011) Human dental pulp stem cells hook into biocoral scaffold forming an engineered biocomplex. PLoS One 6: e18721.

  39. Yamaguchi Y, Ohno J, Sato A, Kido H, Fukushima T (2014) Mesenchymal stem cell spheroids exhibit enhanced in-vitro and in-vivo osteoregenerative potential. BMC Biotechnol 14: 105.

  40. Kim YK, Lee J, Um IW, Kim KW, Murata M, et al. (2013) Tooth-derived bone graft material. J Korean Assoc Oral Maxillofac Surg 39: 103-111.

  41. Nguyen TT, Mui B, Mehrabzadeh M, Chea Y, Chaudhry Z, et al. (2013) Regeneration of tissues of the oral complex: current clinical trends and research advances. J Can Dent Assoc 79: d1.

  42. Kim JY, Kim MR, Kim SJ (2013) Modulation of osteoblastic/odontoblastic differentiation of adult mesenchymal stem cells through gene introduction: a brief review. J Korean Assoc Oral Maxillofac Surg 39: 55-62.

  43. Choi YA, Seol MY, Shin HI, Park EK (2014) Bobby Sox homology regulates odontoblast differentiation of human dental pulp stem cells/progenitors. Cell Commun Signal 12: 35.

  44. Gay I, Cavender A, Peto D, Sun Z, Speer A, et al. (2014) Differentiation of human dental stem cells reveals a role for microRNA-218. J Periodontal Res 49: 110-120.

  45. Boyle M, Chun C, Strojny C, Narayanan R, Bartholomew A, et al. (2014) Chronic inflammation and angiogenic signaling axis impairs differentiation of dental-pulp stem cells. PLoS One 9: e113419.

  46. Roozegar MA, Mohammadi TM, Havasian MR, Panahi J, Hashemian A, et al. (2015) in vitro Osteogenic impulse effect of Dexamethasone on periodontal ligament stem cells. Bioinformation 11: 96-100.

  47. Pisciotta A, Riccio M, Carnevale G, Beretti F, Gibellini L, et al. (2012) Human serum promotes osteogenic differentiation of human dental pulp stem cells in vitro and in vivo. PLoS One 7: e50542.

  48. Choi H, Jin H, Kim JY, Lim KT, Choung HW, et al. (2014) Hypoxia promotes CEMP1 expression and induces cementoblastic differentiation of human dental stem cells in an HIF-1-dependent manner. Tissue Eng Part A 20: 410-423.

  49. Rezai Rad M, Wise GE, Brooks H, Flanagan MB, Yao S (2013) Activation of proliferation and differentiation of dental follicle stem cells (DFSCs) by heat stress. Cell Prolif 46: 58-66.

  50. Abdullah M, Rahman FA, Gnanasegaran N, Govindasamy V, Abu Kasim NH, et al. (2014) Diverse effects of lead nitrate on the proliferation, differentiation, and gene expression of stem cells isolated from a dental origin. ScientificWorldJournal 2014: 235941.

  51. Lu J, Dai J, Wang X, Zhang M, Zhang P, et al. (2015) Effect of fibroblast growth factor 9 on the osteogenic differentiation of bone marrow stromal stem cells and dental pulp stem cells. Mol Med Rep 11: 1661-1668.

  52. Hynes K, Menicanin D, Gronthos S, Bartold PM (2012) Clinical utility of stem cells for periodontal regeneration. Periodontol 2000 59: 203-227.

  53. Yamada Y, Ito K, Nakamura S, Ueda M, Nagasaka T (2011) Promising cell-based therapy for bone regeneration using stem cells from deciduous teeth, dental pulp, and bone marrow. Cell Transplant 20: 1003-1013.

  54. Rodriguez-Lozano FJ, Insausti CL, Iniesta F, Blanquer M, Ramirez MD, et al. (2012) Mesenchymal dental stem cells in regenerative dentistry. Med Oral Patol Oral Cir Bucal 17: e1062-1067.

International Journal of Anesthetics and Anesthesiology (ISSN: 2377-4630)
International Journal of Blood Research and Disorders   (ISSN: 2469-5696)
International Journal of Brain Disorders and Treatment (ISSN: 2469-5866)
International Journal of Cancer and Clinical Research (ISSN: 2378-3419)
International Journal of Clinical Cardiology (ISSN: 2469-5696)
Journal of Clinical Gastroenterology and Treatment (ISSN: 2469-584X)
Clinical Medical Reviews and Case Reports (ISSN: 2378-3656)
Journal of Dermatology Research and Therapy (ISSN: 2469-5750)
International Journal of Diabetes and Clinical Research (ISSN: 2377-3634)
Journal of Family Medicine and Disease Prevention (ISSN: 2469-5793)
Journal of Genetics and Genome Research (ISSN: 2378-3648)
Journal of Geriatric Medicine and Gerontology (ISSN: 2469-5858)
International Journal of Immunology and Immunotherapy (ISSN: 2378-3672)
International Journal of Medical Nano Research (ISSN: 2378-3664)
International Journal of Neurology and Neurotherapy (ISSN: 2378-3001)
International Archives of Nursing and Health Care (ISSN: 2469-5823)
International Journal of Ophthalmology and Clinical Research (ISSN: 2378-346X)
International Journal of Oral and Dental Health (ISSN: 2469-5734)
International Journal of Pathology and Clinical Research (ISSN: 2469-5807)
International Journal of Pediatric Research (ISSN: 2469-5769)
International Journal of Respiratory and Pulmonary Medicine (ISSN: 2378-3516)
Journal of Rheumatic Diseases and Treatment (ISSN: 2469-5726)
International Journal of Sports and Exercise Medicine (ISSN: 2469-5718)
International Journal of Stem Cell Research & Therapy (ISSN: 2469-570X)
International Journal of Surgery Research and Practice (ISSN: 2378-3397)
Trauma Cases and Reviews (ISSN: 2469-5777)
International Archives of Urology and Complications (ISSN: 2469-5742)
International Journal of Virology and AIDS (ISSN: 2469-567X)
More Journals

Contact Us

ClinMed International Library | Science Resource Online LLC
3511 Silverside Road, Suite 105, Wilmington, DE 19810, USA
Email: contact@clinmedlib.org
 

Feedback

Get Email alerts
 
Creative Commons License
Open Access
by ClinMed International Library is licensed under a Creative Commons Attribution 4.0 International License based on a work at https://clinmedjournals.org/.
Copyright © 2017 ClinMed International Library. All Rights Reserved.