Next Article in Journal
Antibiotics Act with vB_AbaP_AGC01 Phage against Acinetobacter baumannii in Human Heat-Inactivated Plasma Blood and Galleria mellonella Models
Next Article in Special Issue
Secretome of Hypoxic Endothelial Cells Stimulates Bone Marrow-Derived Mesenchymal Stem Cells to Enhance Alternative Activation of Macrophages
Previous Article in Journal
ESR Method in Monitoring of Nanoparticle Endocytosis in Cancer Cells
Previous Article in Special Issue
Substantial Overview on Mesenchymal Stem Cell Biological and Physical Properties as an Opportunity in Translational Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Functions of Stem Cells from Oral Cavity: An Update

1
Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
2
Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Authors share co-corresponding authorship.
Int. J. Mol. Sci. 2020, 21(12), 4389; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21124389
Submission received: 1 June 2020 / Revised: 14 June 2020 / Accepted: 17 June 2020 / Published: 19 June 2020

Abstract

:
Adult stem cells have been developed as therapeutics for tissue regeneration and immune regulation due to their self-renewing, differentiating, and paracrine functions. Recently, a variety of adult stem cells from the oral cavity have been discovered, and these dental stem cells mostly exhibit the characteristics of mesenchymal stem cells (MSCs). Dental MSCs can be applied for the replacement of dental and oral tissues against various tissue-damaging conditions including dental caries, periodontitis, and oral cancers, as well as for systemic regulation of excessive inflammation in immune disorders, such as autoimmune diseases and hypersensitivity. Therefore, in this review, we summarized and updated the types of dental stem cells and their functions to exert therapeutic efficacy against diseases.

1. Introduction

Tissue-specific adult stem cells (ASCs) are the specialized cell population responsible for organ development, homeostasis, and regeneration throughout the lifetime. In general, ASCs have a great self-renew potential with lineage-specific differentiation capacity. For instance, a subset of transplanted hematopoietic stem cells can replenish the whole-blood system of lethally irradiated mice [1]. A single Lgr5+ intestinal stem cell (ISCs) can reconstitute the intestinal epithelial layer containing not only ISC itself but also other mature cell types [2]. Besides organs, connective tissues such as bone and fat are regarded as a rich source of multipotent mesenchymal stromal/stem cells (MSCs). Similar to other organ-derived ASCs, MSCs can self-renew and proliferate well. They are capable of mesenchymal lineage-specific differentiation into bone, adipose tissue, and cartilage both in vitro and in vivo; however, in addition to primary tissue replacement, MSCs are known to play pivotal roles in microenvironment regulation. MSCs contribute to stem cell niche formation and support region-specific ASCs to maintain their stemness and multipotency [3]. They communicate neighbors via direct cell-to-cell contact but also via indirect, secretory factor-dependent signaling so-called paracrine effect. According to the context, MSCs produce a plethora of bioactive molecules that can promote stem/progenitor cell proliferation, determine the direction of differentiation, enhance angiogenesis and even modulate immune responses, leading to wound healing and tissue repair [4,5]. Furthermore, it is widely accepted that MSCs are less immunogenic than other ASCs since they express a low level of MHC antigens and immune cell co-stimulatory molecules [6,7]. In these aspects, MSCs have drawn great interest in the fields of stem cell therapeutics and regenerative medicine.
As connective tissues are widely distributed throughout the body, a variety of MSCs have been described from various origins. Considering that the invasiveness of harvest procedure often limits their clinical utility, easily accessible dental- and periodontal tissues would be an attractive source for the autologous MSC isolation [8,9]. To date, researchers have successfully isolated different types of MSCs from dental specimens that are usually discarded during the treatment such as extracted teeth, attached ligament, and gingival tissue. Of note, dental development begins in the prenatal period but continues beyond the birth until the permanent teeth replace the deciduous ones. For this reason, dental MSCs can be obtained from the developing, immature tissues with higher stemness and flexibility [10]. Dental MSCs share common MSC-related features with regards to self-renewal, extensive proliferation, mesenchymal differentiation capacity, and surface marker expression, while they also exhibit distinctive biological actions depending on their origins [11]. Indeed, they seem to enhance mineral deposition during odontoblast formation and stimulate the neovascularization process within the dental pulp defect. Therefore, the inheritance properties of different dental MSCs should be considered in advance for clinical applications as well as basic science research.
In this review, we aim to provide a comprehensive overview of the general- and unique characteristics of various dental MSCs. This paper also summarizes the latest representative studies showing their regenerative- and immunomodulatory actions in non-dental immunogenic diseases, as well as dental disorders.

2. Stem Cells in the Oral Cavity: Sources, General Properties, and Therapeutic Potentials

2.1. Stem Cells in Oral Cavity

In terms of developmental view, dental and periodontal tissues are generated via continuous reciprocal actions between ectodermal epithelial cells and ectomesenchymal cells derived from the neural crest as well as the mesoderm [12,13]. While the epithelial progenitors form enamel tissue covering the crown, the ectomesenchyme is responsible for other major compartments of the teeth-dentin, cementum, and dental pulp. Surrounding tissues such as periodontal ligament and gingiva are generated from ectomesenchyme-derived progenitors, implying the presence of ectomesenchymal MSCs. Up to date, various MSC-like cells have been isolated from dental- and periodontal tissues with ectomesenchymal origins including dental pulp stem cells (DPSCs), periodontal ligament stem cells (PDLSCs), gingiva-derived mesenchymal stem cells (GMSCs), dental follicle progenitor cells (DFPCs) and stem cells from apical papilla (SCAP). The naturally exfoliated deciduous teeth also contain dental MSCs, so-called stem cells from human exfoliated deciduous teeth (SHED). Due to their developmental origin, they not only share general aspects of other MSCs but also present neurogenic capacity similar to neural crest-derived stem cells (NCSCs) [14]. In addition, they present distinctive characteristics depending on anatomical locations as summarized below (Table 1).

2.1.1. Dental Pulp Stem Cells (DPSCs)

The dental pulp is a loose connective tissue located in the innermost center part of the teeth. It consists of tiny blood vessels, nerves, and various cells including dental pulp stem cells (DPSCs) [15]. DPSCs are the first discovered MSCs among oral cavity-derived stem cells; in 2000, Gronthos et al. isolated the stem cell-like cell population from the dental pulp of the human third molars, known as “wisdom teeth” [16]. In general, obtained pulp tissue was digested using collagenase type I to generate single-cell suspensions followed by clonal expansion [17].
DPSCs express MSC markers such as CD10, CD13, CD29, CD44, CD73, CD90, CD105, CD146, and CD166 but do not express CD14, CD24, CD34, CD45, and HLA-DR [18]. In addition, they express some neural lineage markers such as Glial fibrillary acidic protein (GFAP), Microtubule-associated protein 2 (MAP2), and Nestin, as well as pluripotent markers Oct-3/4, Nanog and Sox-2 [19,20]. DPSCs can differentiate into odontoblast, osteoblasts, adipocytes, and neural crest cells [21,22]. DPSCs were reported to express the osteoblastic markers such as alkaline phosphatase (ALP), collagen type I (Col I), osteopontin (OPN), and osteocalcin (OCN) and they could differentiate into osteoblast-like cells producing mineralized matrix and give rise to bone tissue [23,24]. Also, DPSCs have superior proliferative- and osteogenic capability compared to bone marrow-derived MSCs (BM-MSCs), suggesting that DPSCs are suitable for tooth regeneration [25,26].
When dental injuries and odontoblast apoptosis occur, rapid activation of DPSCs has been observed followed by their proliferation, migration and differentiation into odontoblast cells, implying that DPSCs play key roles in tooth homeostasis and periodontal regeneration [8,27]. Among the various pathways involved in the odontoblast differentiation, Notch signaling has been reported to induce the odontogenic differentiation of DPSCs and regeneration of the dental pulp [28]. In addition, DPSCs produce neurotrophic factors and promote survival as well as neurite outgrowth of the trigeminal neurons [29]. Of note, DPSCs cultured in the neuronal inductive media condition could express neuronal marker such as nestin, beta III tubulin, neurofilament medium chain (NF-M) and neurofilament heavy chain (NF-H) and differentiate into active neurons [30].

2.1.2. Periodontal Ligament Stem Cells (PDLSCs)

Periodontal ligament (PDL) is one of the major components of the periodontium that connects the tooth-root surface and alveolar bone. The primary role of PDL is tooth support [31]. PDL tissue is composed of heterogeneous cell populations including fibroblast, undifferentiated MSCs, endothelial cells, fibroblast, epithelial cells, the rest of Malassez, and cementoblasts [32,33].
PDLSCs have been identified near the adult PDL tissue of the teeth and play a critical role in periodontal tissue regeneration [34]. The first report describing the isolation and identification of human PDL-derived MSCs was published in 2004 [35]. Since then, there have been continuous attempts to elucidate the function of PDLSCs and their interactions with other dental cells for the clinical application in regenerative periodontics. The PDL cells can be obtained from PDL tissues by enzymatic digestion into single-cell suspensions, which can form adherent clonogenic cell clusters in vitro [36]. Previous studies have reported that PDLSCs represent typical characteristics of BM-MSCs and express MSC-associated cell surface markers such as STRO-1, CD10, CD146, CD13, CD29, CD44, CD59, CD73, CD90, and CD105 while negative for CD14, CD34, CD45, CD38, CD54, or HLA-DR [35,37].
PDLSCs exhibit a self-renewing capacity and possess the multi-potency to differentiate into various types of periodontal tissue components, such as cementum, alveolar bone, and Sharpey’s fibers in vitro and in vivo [38,39]. Several bioactive factors have been reported to regulate the proliferation and differentiation of PDLSCs. A previous study has demonstrated that transforming growth factor-beta (TGF-β1) plays an important role in the fibroblastic differentiation of PDLSCs [40]. TGF-β1 was highly expressed in the entire PDL tissue, and the synthesis of TGF-β1 in PDLSCs depends on their differentiation stage. In addition, the treatment of vascular endothelial growth factor (VEGF) to PDLSCs seem to stimulate the odonto-/osteogenic differentiation in vitro and induce the mineralization of bone structure in vivo. As a powerful inducer of the progenitor cell proliferation in hard tissue regeneration, in contrast, fibroblast growth factors (FGF-2) would enhance the proliferation while inhibiting terminal differentiation of PDLSCs in vitro [41]. Finally, PDLSCs have an immunomodulatory ability comparable to other MSCs; for instance, allogeneic PDLSCs could enhance periodontal regeneration due to their low immunogenicity and immunosuppressive effects on T cell as well as B cells [42].

2.1.3. Gingiva-Derived Stem Cells (GMSCs)

As a part of the periodontium, gingiva tightly surrounds the teeth and underlying alveolar bones to support oral hygiene, providing an intact mucosal barrier against physiobiological challenges with superior wound healing capacity. This soft connective structure consists of the epithelium and lamina propria originated from the outer (perifollicular mesenchyme) and inner layer (dental follicle proper) of the dental follicles [43]. Of note, the gingiva is an easily accessible, highly renewable tissue compared to other dental components and it is often surgically excised during general dental treatment. In this aspect, gingiva has been considered as a convenient source of ASCs in the oral cavity.
Since the first successful isolation by Zhang et al., several protocols have been reported to culture the GMSCs with a description of their characteristics [43,44]. In the immunophenotypic analysis, GMSCs are positive for classical MSC markers including CD29, CD44, CD73, CD90, and CD105 while negative for hematopoietic lineage cell markers such as CD14, CD34, and CD45 [45,46]. They can self-renew while maintaining the long-term proliferative capacity with a faster population doubling time compared to BM-MSCs [47,48].
GMSCs also exhibit a multi-lineage differentiation potential into osteoblast, adipocyte, and chondrocyte as observed in other source-derived MSCs [49]. In addition, evidence of the trans-differentiative potential of GMSCs into β-tubulin+ or Glial fibrillary acidic protein (GFAP)+ neural lineage cells as well as CD31+ endothelial cells have been reported, although it is still controversial and supporting results should be followed to confirm these findings [44,50]. Notably, several attempts have been conducted to regulate their stemness and/or differentiation fate into a designate direction. Ge et al. have evaluated the effect(s) of basic fibroblast growth factor (bFGF) on GMSCs and found that bFGF could not only enhance the proliferative capacity of GMSCs but also stimulate adipogenesis while impeding osteogenesis [51]. It has shown that the encapsulation of GMSCs using hydrogels composed of alginate-gelatin methacryloyl (GelMA) could increase cell viability and control osteogenic potential depending on the hydrogel stiffness [52]. Meanwhile, conditioned medium obtained from embryonic- or postnatal apical tooth germ cell culture seems to support the osteogenic- and odontogenic induction of GMSCs, partially by providing environmental cues found in periodontal development [53,54]. In line with these findings, enamel matrix derivative (EMD) from tooth buds could stimulate cellular proliferation and mineralization during osteogenic differentiation in GMSCs [50].
Similar to other MSCs, GMSCs also play immunomodulatory roles via interacting immune cells [44,55]. Ge et al. have investigated the difference between healthy- and inflamed tissue-derived GMSCs and concluded that those cells might be functionally equivalent in terms of proliferation and differentiation [56]. Given that inflammatory signals could alter the immunomodulatory properties of MSCs, it would be necessary to further evaluate the immune-related phenotypes of GMSCs from inflamed gingiva.

2.1.4. Stem Cells from Human Exfoliated Deciduous Teeth (SHED)

Both SHEDs and DPSCs are derived from the dental pulp; however, SHEDs are derived from deciduous teeth, whereas DPSCs are derived from permanent teeth. Deciduous teeth are the official term for baby teeth, milk teeth, or primary teeth. They start developing during the embryonic stage then begin to come in about 6 months after birth in general.
As first described by Miura et al., the harvesting SHEDs is a simple and convenient procedure with little trauma [57]. Two methods are accepted to isolate and culture of SHEDs: enzymatic digestion and tissue explant. For the enzymatic digestion, the pulp tissues of the deciduous teeth are dissociated with collagenase type I and Dispase for the stem cell isolation. For the latter method, mechanically minced pulps are placed on the tissue culture dishes, allowing the clonal expansion of stem cells [58,59].
SHEDs have a typical fibroblast morphology and express MSC specific markers such as CD45, CD90, CD106, CD146, CD166, and STRO-1 but not the hematopoietic and endothelial markers CD34 and CD31 [60]. As a member of neural crest-derived stem cells, SHEDs also express neural cell markers including Nestin, beta III tubulin, and GFAP some of the pluripotent markers such as Oct4 and Nanog [13,57,61]. SHEDs present multi-lineage differentiation ability to generate osteoblasts, hepatocytes, adipocytes, and neural cells [62,63,64]. SHEDs exhibit increased gene expression patterns of osteocyte markers, including ALP, Col I, and runt-related transcription factor 2 (Runx2) than BMSCs in vitro. In addition, SHEDs can differentiate into functional odontoblasts that secrete mineralizable dentin matrices [65]. Yamaza et al. have demonstrated that the subcutaneous transplantation of SHEDs in nude mice could enhance bone repair via osteoclast inhibition in vivo [66]. SHEDs can also differentiate into vascular endothelial cells that form functional blood vessels by up-regulating the endogenous MEK1/ERK signaling [67].
Due to their origin, SHEDs generally exhibit characteristics similar to DPSCs. However, their proliferation and differentiation capacity are higher than DPSCs and BM-MSCs. Furthermore, SHEDs produce several growth factors, including FGF, TGF-β, connective tissue growth factor (CTGF), and bone morphogenic protein (BMP). In particular, CTGF derived from SHEDs contributes to the neo-dentinogenesis in human corroded teeth [68,69].

2.1.5. Dental Follicle Stem Cells (DFSCs)

Dental follicle (DF) is a loose connective tissue derived from ectomesenchymal tissue and part of the tooth germ, surrounding the enamel organ and the developing tooth [70]. The DF plays various roles during tooth development by regulating the osteoclastogenesis and osteogenesis needed for tooth eruption and tooth-root development of the periodontium [71].
Isolation of human DFSCs has been first established in 2002 [72]. Briefly, DF tissue is dissected from the upside of the dental crown and incubated with a dissociative solution containing collagenase and trypsin. Followed by cell seeding, the subset population of DFSCs would adhere to the plastic dish surface, while non-adherent cells are removed simply by the media change [73]. DFSCs express CD9, CD10, CD13, CD29, CD44, CD53, CD59, CD73, CD106, CD166, and CD271, but not hematopoietic markers including CD 117, CD31, CD34, CD45, and CD133 [14]. Also, DFSCs have putative markers of MSCs and other stem cells such as STRO-1, Oct3/4, Sox-2, Nestin, Nanog, and Notch [70,74].
It has been shown that DFSCs can differentiate into multiple lineage cell types such as osteoblasts, cementoblasts, adipocytes, and neuron-like cells [75,76]. Xiang et al. have investigated the expression pattern of Wnt5a in postnatal DF tissue and its role in the differentiation of DFSCs. They found that Wnt5a could regulate the fate of DFSCs towards differentiation into PDL cells or mineralized cementoblasts and/or alveolar bone osteoblasts [64]. In another study, it is revealed that BMP-2, a member of BMP glycoproteins playing pivotal roles in the development and regeneration of the bone, induces the differentiation of DFSCs into cementoblast/osteoblast-like cells [72]. Similarly, BMP-9 transfected DFSCs present an enhanced osteogenic capacity compared to control cells, suggesting that BMP-9 can be used as osteogenesis inducer of DFSCs [77]. In addition, long-term culture of DFSCs with dexamethasone or insulin could induce the osteoblast lineage commitment and upregulate the mRNA transcripts for osteoblast-specific genes including distal-less homeobox 3 (DLX-3) [78], BMP-2, bone sialoprotein (BSP), and OPN in a culture-duration dependent manner [79]. Yao et al. have demonstrated that dentin matrix acidic phosphoprotein 1 (DMP1) is highly expressed in the DFSCs [80]. The knockdown of DMP1 distinctly reduced the osteogenic capacity of DFSCs, whereas supplementation with mrDMP1 protein to the osteogenic induction medium profoundly increased the osteogenesis, suggesting that DMP1 plays a key role in maintaining and promoting osteogenic capability of DFSCs [80].

2.1.6. Apical Papilla Stem Cells (APSCs)

With the neighboring Hertwig epithelial root sheet, the apical papilla at the apex of developing immature teeth contributes to root development as well as regeneration, implying the presence of resident stem cells. In 2006, it was first reported that the apical papilla is another dental stem/progenitor cell source for so-called ‘stem cells from the apical papilla (SCAPs)’ [81]. SCAPs can be isolated from the apical papilla via single-cell culture after enzymatic dissociation or organotypic tissue culture method [81,82]. The established clonogenic SCAPs have typical characteristics of other dental MSCs in terms of immunophenotype, self-renewal, and differentiation capacity; however, they also exhibit various unique as well as superior properties for clinical application as described below.
First, SCAPs are highly expandable than other dental MSCs [83]. A further study has found that a telomere length of SCAPs is much longer compared to those of DPSCs and PDLSCs because of strong telomerase activity, which enables SCAPs to maintain longer lifespan without premature aging [84]. In addition, SCAPs are relatively resistant to irradiation or chronic periapical inflammation than other dental progenitors, indicating the micro-environmental differences between each stem cell niche [85,86].
Since SCAPs are derived from the immature structure, they express primitive embryonic markers such as Sox2, Oct3/4 and Nanog accompanied with typical MSC markers [81,87]. Among those markers, co-expression of CD146 and STRO-1 is found to be related to early MSCs; indeed, CD146+/STRO-1+ SCAPs exhibit superior colony-forming efficiency with increased cumulative doubling than counterpart [88]. CD24, another marker for the pluripotent population, is regarded as a representative surface marker for SCAPs because of its absence in other dental MSCs [89]. It would be worth to indicate that these three markers tend to be declined with passaging, supporting their correlation with superior stemness.
Similar to other MSCs, SCAPs are multipotent and can give rise to mesenchymal lineage cells including adipocyte and chondrocyte; however, SCAPs are known to be rather optimized for osteogenesis and odontogenesis. These findings are not surprising considering that SCAPs are regarded as the precursors for primary odontoblast in vivo [88,90]. Liang et al. found that activation of Wnt/β-catenin pathway could promote osteogenic differentiation [91], while others have reported the opposite results showing the osteogenic/odontogenic potential of Wnt inhibitory molecules such as secreted frizzled-related protein 2 (SFRP2) and Wnt inhibitory factor 1 (WIF1) [92,93]. TGF-β Signaling is another intriguing pathway to regulate SCAP differentiation and increased TGF-β1 activity is correlated to osteogenic suppression of SCAPs [94]. Of note, it is revealed that TGF-β2 is highly up-regulated during osteogenesis and recombinant TGF-β2 could induce a specific increase in odontogenic signatures of SCAPs while it simultaneously reduces osteogenic markers, indicating that each TGF-β family plays some distinctive roles in tooth development and regeneration [95]. Growth factors and hormones can affect osteogenic differentiation of SCAP; the concentrated growth factors from the culture medium, platelet-rich Fibrin, and estradiol have found to contribute to enhanced osteogenesis as well as proliferation [96,97]. Insulin-like growth factor-1 (IGF-1) is another well-described factor that potentiates the osteogenic/odontogenic differentiation in SCAPs [98].
Besides mesenchymal lineage, SCAPs can be differentiated into other lineages; interestingly, sub-population of SCAPs not only expresses neuronal cell-associated markers including βIII tubulin, GAD, nestin, and neurofilament M but also secretes some neurotrophic, neuroprotective factors to support neurogenesis and regeneration [99,100]. The use of fibrin hydrogels or nanomaterial-based scaffolds composed of Graphene and single-wall carbon nanotubes could enhance the neural differentiation capacity of SCAPs [101,102]. In addition, hepatocyte-like cells expressing hepatocyte nuclear factor-1α, alpha-fetoprotein, and albumin could be derived from SCAPs and these differentiated cells exhibit glycogen-storage function and urea production [103]. The angiogenic function of SCAPs also has been proved both in vitro and in vivo, although it is not via direct trans-differentiation of SCAPs into endothelial cells but via promoting migration and tube formation of existing vascular cells [104,105]. Collectively, SCAPs are expected to exert significant roles in tissue regeneration and repair.

2.2. Therapeutic Features of Dental MSCs

Tissue regeneration and immunomodulation are the major therapeutic benefits of dental MSCs. Tissue regenerative effect of MSCs can be achieved by (1) the replacement of defected tissues with differentiated dental MSC itself and/or (2) induction of de novo regeneration capacity of endogenous stem cells via remodeling the stem cell niche [106]. Meanwhile, MSCs exhibit an immunomodulatory function by communicating with a variety of immune cells and regulating their activity in a context-dependent manner; mostly, however, MSCs present anti-inflammatory action and stimulate the immune response resolution, which in turn leads to wound healing and tissue repair [107].

2.2.1. Tissue Regeneration

Stem cell therapeutics can be applied to the periodontal treatment for tissue regeneration, which can be achieved by direct cells/tissue replacement or enhancement of endogenous regenerative capacity [108]. During the normal to mild state of the disease, de novo oral cavity-resident stem cells can maintain the tissue homeostasis by itself; however, as the pathologic condition worsens, the exogenous substitutes such as ex vivo expanded/manipulated stem cells would be required to recover the host microenvironment and facilitate the tissue regeneration [109]. In this aspect, dental MSCs can be transplanted into impaired periodontal tissues to function as building blocks to tissue restoration, or stimulate the de novo regeneration via secretion of beneficial factors [110].
Up to date, several studies have proven that dental MSCs can induce the regeneration of not only dental tissues including enamel, dentin, tooth pulp, and PDL but also other organs that share the same developmental origins such as bone and salivary gland [65,111]. Cell modification approaches such as cell immortalization, hypoxic culture condition, and 3-dimensional spheroid formation have been applied to potentiate the stemness and differentiation capacity [111,112,113]. It is further noted that native and synthetic biomaterials can be combined with dental MSCs to provide a physical scaffold that supports engraftment and differentiation of transplanted cells. For instance, when DFSCs contact with aligned PLGA/Gelatin electrospun sheet and fabricated extracellular matrix, periodontium-like complex could be generated both in vitro and in vivo [114]. Another report has shown that transplantation of DFSCs combined with treated dentin matrix into rat alveolar fossa could regenerate the root-like tissue with pulp-dentin complex and PDL connecting cementum-like layer [115]. Huang et al. have successfully generated vascularized dental pulp-like tissue with dentin structure using DPSCs and SCAP mixture seeded in a copolymer of poly-d,l-lactide and glycolide (PLG) scaffold [116]. Collectively, these advanced protocols using modified dental MSCs and biomaterials may provide an optimal approach for regenerative periodontal therapy.

2.2.2. Immunomodulation

Dental MSCs have been reported to present immunomodulatory impact through reciprocal interactions with both innate- and adaptive immune cells as well as the microenvironment, leading to a context-specific immune response regulation [106,117]. For instance, PDLSCs can alter the immune microenvironment to enhance periodontal regeneration by inducing macrophage polarization towards the tissue repair-supportive M2 phenotype while they seem to suppress the proliferation, differentiation, and migration of B cells via enhancing programmed cell death protein-1 signaling, leading to declining in immunoglobulin production [118,119]. DPSCs could impair the viability of natural killer cells and impede their cytotoxic activity in vitro [120]. In addition, SHEDs inhibit the differentiation of T helper 17 (Th17) cells and increase the ratio of regulatory T cells (Tregs) versus Th17 in vivo [66].
Various paracrine effectors mediate the aforementioned immunomodulatory capacity of dental MSCs. When DPSCs are co-cultured with patient-derived peripheral blood mononuclear cells (PBMC), increased secretion of anti-inflammatory cytokines such as prostaglandin E2 (PGE2), Interleukin-6 (IL-6), and TGF-β could inhibit the proliferation of PBMCs [121,122]. Similarly, GMSCs and PDLSCs can reduce the expansion of PBMCs and allogeneic T cells via the expression of IL-10, IDO, iNOS, and cyclooxygenase-2 (COX-2) [123]. In another study, Fas ligand (FasL) and IL-10 have been suggested as major GMSC-derived factors of T cell inhibition, while others have revealed the importance of PGE2, a lipid immune-mediator of both innate and adaptive immune system, and other secretory factors including IL-6 and GM-CSF on immunomodulatory properties of GMSCs [124,125]. Interestingly, preconditioning with pro-inflammatory cytokine interferon-γ (IFN-γ) or hypoxic conditions could promote the immunosuppressive potential of GMSCs both in vitro and in vivo [44,126].
Finally, dental MSCs are known to have low immunogenicity due to the absence of HLA-II DR or T cell costimulatory molecules (CD80 and CD86), indicating themselves as promising candidates for the allogenic stem cell application [127].

3. Clinical Application of Dental MSCs

Due to its origins, several attempts have been conducted to apply the tissue regenerative potential of dental MSCs for the dental- and periodontal regeneration (Table 2), while the immunomodulatory capacity of dental MSCs has been evaluated in other immune-related disorders (Table 3) as well as in dentistry.

3.1. Dental Stem Cell Application in Dental and Periodontal Disease

3.1.1. Periodontitis

Periodontitis is one of the most common periodontal disease [128]. It begins in the sulcus region of the gingiva then travels to the apex as deposits of plaque and calculus induce inflammation, which slowly disrupts the attachment apparatus [129]. The primary etiology of periodontitis is an ill-defined series of microbial infections [130]. Periodontitis is a slowly developing condition but chronic inflammation induces infectious lesions of the gingiva, PDL alveolar bone, and eventually the tooth loss [131]. Tissue destruction in periodontitis is caused by the breakdown of collagen fibers of the PDL, leading to the periodontal pocket formation between the gingiva and tooth and the resorption of alveolar bone, leading to tooth mobility and subsequent tooth loss [132].
The therapeutic goal of periodontitis is reducing the pocket depth to prevent the progression of the disease and regenerating the lost deprived periodontium [133]. In patients with mild periodontitis, non-surgical therapy such as antimicrobial photodynamic therapy would be sufficient to manage the symptoms, while periodontal surgery is recommended to eliminate the residual periodontitis lesions in severe cases [134]. In general, periodontal surgery would prevent the further progress of the disease [135]; however, it would only stabilize the situations while the tissue loss is irreversible [136].
In this aspect, a multi-level cellular/biomaterial treatment strategy may provide an optimal therapeutic approach for periodontitis. The transplantation of PDLSCs to damaged periodontal tissues would help the remodeling of the local microenvironment as well as tissue by secreting various cytokines that may stimulate the function of residual progenitor cells, which in turn prevents the epithelial down-growth and induces recovery from the periodontal defects [137]. Ding et al. have transplanted cell sheets composed of allogeneic PDLSCs for the treatment of periodontitis in miniature pigs. They found that PDLSC sheets present low immunogenicity and provide appropriate therapy for periodontitis with significant periodontal tissue repair [127]. In addition, stem cells from SHEDs are likely to be a striking candidate for periodontium tissue regeneration. Fu et al. have transplanted SHEDs, composites of allogeneic stem cell sheets derived from minipig deciduous teeth (SPD) combined with hydroxyapatite/tricalcium phosphate (HA/TCP), into a periodontitis model in miniature swine to evaluate their therapeutic potential. The periodontal tissues, including new bone, cementum, and PDL, are regenerated in the periodontal defect area and 75% of the samples had successful furcation regeneration in the SHEDs group, suggesting that SHEDs can provide proper therapy for periodontitis [138]. Furthermore, xenografts of human DPSCs into immuno-deficient athymic mice have shown that DPSCs can produce periodontal cement-like materials, suggesting that DPSCs contain the potential of new therapeutic approaches for periodontal tissue regeneration [139].

3.1.2. Dental Hard Tissue and Pulpal Diseases

Dental caries, also known as tooth decay, is a chronic diseasein the dental hard tissue. Dental caries is a multifactorial disease and caused by localized destruction of dental hard tissues by several risk factors including acid-producing bacteria, fermentable carbohydrates, teeth mineral contents, and oral hygiene [146]. Dental caries arises from an impaired physiological homeostasis between tooth minerals and oral microbial biofilms leading to loss of tooth structure and discomfort [147]. In contrast, dental traumatic injuries are other dental hard tissue defects caused by exogenous damaging factors [136].
Up to now, the main treatment method of dental hard tissue defects is direct filling with amalgam or composite resins to recover the dental defects [148]; however, surgical approaches are often accompanied with remaining problems such as eliminating some healthy tissues to get retention for amalgam or shrinkage, emphasizing the needs of novel therapeutic options [149].
Of note, transplanted DPSCs for the treatment of artificial tooth defects on the premolars and the molars in dogs induced dentin and pulp capping regeneration [140]. Also, DPSCs could reduce dental tissue inflammation and exert immunomodulatory functions either via direct cell-cell contact or via paracrine effects which can prevent proliferation, maturation, cytokine/antibody production and antigen presentation by T cells, B cells, NK cells, and DCs [141].
The pulp tissue reacts in a variety of ways in the presence of periodontal disease [150], while the most common response of the pulp to injury is inflammation so-called pulpitis. In general, root canal therapy has been applied to deal with pulpitis [151]. As the substitution of the naive pulpal tissues with foreign materials leaves the teeth insensitive and changes the composition of enamel and dentin as well as the microenvironment, cell-based therapy is required to regenerate tissues [136]. In this aspect, Iohara et al. have transplanted autologous PDLSCs and progenitors into a root canal with stromal cell-derived factor (SDF-1) after pulpectomy in mature teeth with whole apical closure in dogs. The pulp CD105+ cells showed high expression of angiogenic and neurotrophic factors and exhibit complete pulp regeneration [143]. In addition, a previous study has demonstrated complete pulp regeneration by harnessing autologous DPSCs subsets transplanted with SDF-1 in a collagen scaffold into a canine pulpitis model [142].

3.1.3. Alveolar Bone Diseases

Bone loss is one of the hallmarks of periodontal diseases including loss of teeth and chronic periodontitis [152]. Conventional therapies for bone loss include the use of agents such as nonsteroidal anti-inflammatory drugs, glucocorticoids, and anti-rheumatic drugs. These immune regulating therapies may decrease the inflammation temporarily, whereas they do not provide any direct effect on controlling or reversing bone resorption, limiting their uses in the treatment of pathologic bone loss in periodontal defects [153].
Of note, the superior osteogenic regenerative capacity of DPSCs can be applied to bone loss. The treatment of DPSCs with valproic acid can enhance mineralized matrix formation and the expression of bone glycoproteins [154]. DPSCs transplanted with various scaffolds including HA/TCP [155], PLGA [156], collagen [157], nano-fiber hydrogel [158], fibroin [159], and platelet-rich plasma (PRP) [160] to the alveolar bone defects induce bone regeneration. BMP2 and autologous DPSCs promote mineralized tissue formation and osteogenesis [144]. DPSCs in dogs show the highest mean bone-implant contact values than periosteal cells and BM-MSCs [145]. Overall, DPSCs possess a high osteogenic potential and can be applied to the tissue-engineered bone regeneration.

3.1.4. Application of Dental MSC-Derived Secretome in Dental Disorders

As the paracrine effect is one of the major therapeutic mechanisms of stem cell application, the therapeutic potential of these secretomes derived from dental MSCs on dental disease has been also evaluated [161]. It is revealed that signaling components of the bone morphogenetic proteins (BMPs) and TGF-β pathway seem to be increased in dental MSC-CM. For this reason, CM obtained from dental MSCs is highly-osteogenic and stimulates odontoblast formation as well as dental MSC proliferation compared to CM from other origins [162,163,164]. The ectopic expression of angiogenic and neurogenic factors within the dental MSC-CM would contribute to dental tissue regeneration [165,166,167]. Interestingly, exosomes isolated from DPSC-CM could also exert similar beneficial actions as CM itself in terms of odontogenic induction and dental pulp regeneration [168,169]. Considering their convenience of manipulation, storage, handling, and management compared to whole CM or cells, exosomes could serve as a novel potent option in the dental MSC therapeutic field.

3.2. Dental Stem Cell Application in Non-Periodontal Immune-Related Disease

3.2.1. Rheumatoid Arthritis (RA)

Rheumatoid arthritis (RA) is the most common type of autoimmune arthritis affecting about 1% of the world’s population. RA is categorized as an autoimmune disease with unknown etiology that attacks the own tissues due to the disruption of the immune system, leading to joint destruction and deformation. It is noted that inflammatory cytokines contribute to enhanced osteoclastogenesis during RA progression [170]. Several pro-inflammatory cytokines and chemokines are abundant in RA tissues compared to normal, indicating the contribution of abnormal immune responses to the RA progress [171,172,173].
Numerous animal studies have shown that the administration of dental MSCs can improve RA symptoms. It is reported that secretory factors from SHEDs could improve arthritis symptoms and inhibited tissue damage by inducing M2 macrophage polarization and the abrogation of RANKL expression in arthritis model mice [174]. Also, the therapeutic potential of GMSCs have been also demonstrated in RA and other bone erosion related diseases; indeed, systemic infusion of GMSCs can significantly reduce the severity of experimental RA, and restore the balance of Th cell subsets via Fas ligand (FasL) and IL-10 secretion [175]. Transplantation of GMSCs into collagen-Induced arthritis (CIA) mice significantly attenuated the severity of arthritis and osteoclast activity, which in turn reduced bone erosion in vivo via anti-inflammatory CD39-CD73-adenosine signaling pathway [176].

3.2.2. Atopic Dermatitis (AD)

Atopic dermatitis (AD) is a chronic, incurable skin disease that usually begins in infancy or childhood. It is accompanied by itching, dry skin, and eczema. Although the incidence of AD has been increasing worldwide, the pathogenesis of atopic dermatitis has not been completely understood [180].
Various immune cells and related factors are known to be involved in the development of AD. In specific, AD seems to develop when the balance of the Th1/Th2 axis moved towards Th2 response than Th1, following by the B cell induction and inflammatory reactions [181,182,183]. In addition, the infiltration of eosinophils in peripheral blood is elevated in most AD patients. Activated eosinophils increase the production of IL-4, IL-5, IL-6, IL-10, and IL-13 from Th2 cells to increase IgE synthesis [182,184].
Among the dental MSCs, DFSCs show potent immunomodulatory properties which make them attractive approaches for suppression of inflammatory conditions. PBMCs isolated from atopic patients were co-cultured with DFSCs. DFSCs suppressed the expression of IL-4 and proliferation of T cells whereas the expression of IFN-γ and IL-10 was increased. Therefore, DFSCs would exert an immunomodulatory effect and therapeutic outcomes in AD [177].

3.2.3. Multiple Sclerosis (MS)

Multiple sclerosis (MS) is a disease of the central nervous system caused by an autoimmune response to the insulation around myelin in the brain, spinal cord, and optic nerves. In normal conditions, peripheral immune cells do not present in the nervous system due to the blood-brain barrier. In the case of MS, on the contrast, the impaired barrier allows peripheral immune cells to move into the brain and spinal cord of the affected patients [185]. Migrated T cells contribute to the increase of IFN-γ, which induce direct damages to the myelin sheath in the nervous system [186,187]. Eventually, MS can cause permanent damage or deterioration of the nerves.
Recently, it is reported that PDLSCs-derived conditioned medium and purified exosomes reduce pro-inflammatory cytokines such as IL-17, IFN-γ, IL-1β, IL-6, TNF-α, and induce the production of anti-inflammatory IL-10. Therefore, PDLSCs-CM and Extracellular Micro-Vesicles (EMVs) may have a modulatory role against inflammation in MS development [178].
Replacement of damaged neural tissues and cells such as oligodendrocytes are other important options for MS therapy. Of note, DPSCs express specific glial markers, including Olig2, NG2, and O4, and they can differentiate into oligodendrocyte progenitor cells (OPCs) in vitro, suggesting DPSCs as a promising alternative therapy for myelin repair [188].

3.2.4. Sjogren’s Syndrome

Sjogren’s syndrome (SjS, SS) is a chronic autoimmune disease characterized by dry mouth and eyes. The abnormal infiltration of lymphocytes in the salivary gland is often observed in the patient. Following the migration of lymphocytes into the gland, salivary epithelial cells are activated by pro-inflammatory cytokines such as IL-1β, IFN-γ, and TNF-αproduced by immune cells [189].
Both type I and II IFNs have been implicated in the pathogenesis of SS and activation of the IFN-γ receptor causes induction of IFN related genes, promoting antimicrobial protection, apoptosis, inflammation, and tissue damage [190,191]. Type I IFNs can activate B cell-activating factor (BAFF), a powerful regulator of B cell differentiation and proliferation [192]. A recent study showed increased levels of circulating BAFF in patients with SS and up-regulation of BAFF expression in inflamed salivary glands. T, leading to abnormal B cell accumulation within the gland [193,194].
Recently, the possibility of salivary gland regeneration was suggested using DPSCs [179]. In this study, the human salivary gland (HSG) cell line was co-cultured with DPSCs in vitro. It is found that HGSs co-cultured with DPSCs showed the increased expression of lysosomal-associated membrane protein-1 (LAMP-1) and CD44 and increased numbers of acinar structures compared to control. Next, only HSG or combination of HSG-murine DPSCs were subcutaneously transplanted into 2-month-old SS model Rag1 null mice for 2 weeks. Interestingly, co-transplantation of HSG and DPSCs induced significantly higher levels of FGF-7, alpha amylase-1 (AMY-1), von Willebrand factor (vWF), neurofilament-200 (NF-200), VEGFR-3, and VEGF-C than HGS transplantation, indicating that DPSCs can support salivary gland differentiation [179].

3.2.5. Inflammatory Bowel Diseases

Inflammatory bowel diseases (IBD) is a chronic disease of the gastrointestinal tract caused by an abnormal mucosal T cell response to commensal and/or pathologic bacterial antigens within the gut lumen. IBD is composed of ulcerative colitis (UC) and Crohn’s disease (CD); CD is regarded as a Th1 mediated inflammatory disorder while UC is regarded as a Th2 mediated disease [195,196]. Indeed, Th2 related cytokines such as IL-4, IL-5, IL-6, and IL-13 are increased in UC patients [197,198]. On the contrary, Th1-related cytokines such as TNF, IFN-γ, and IL-12 are increased in the colonic mucosa of CD [199,200].
Recently, the therapeutic effect of GMSCs in the mouse colitis model [44]. To explore the in vivo immunomodulation capability, GMSCs were transplanted into the subcutaneously dorsal pouches of dextran sulfate sodium (DSS)-treated IBD model mice. As a result, GMSCs significantly ameliorated the colitis symptoms and reduced inflammation via, in part, inducing regulatory T cell response and suppressing the infiltration of pro-inflammatory Th1 and Th17 cells within the colon tissue, implying that GMSCs provide beneficial impact on colitis [44].

4. Future Perspectives and Conclusions

So far, stem cells from bone marrow, adipose tissue, and an umbilical cord or cord blood have been intensively studied throughout the world and stem cells from the oral cavity have been relatively less investigated. Therefore, a large body of studies are required for tissue-specific regeneration of dental and oral tissues. Moreover, the unique characteristics and functions of dental MSCs should be precisely demonstrated based on the comparison of dental MSCs with stem cells from other tissues. In addition, future studies should focus not only on the enhancement of transplantation efficiency of differentiated cells by collaborating with biomaterials, 3D printing and drug delivery for regenerative purpose but also on the functional improvement of paracrine functions via novel strategies including priming, genetic modification, surface or conformation engineering of stem cells for immunoregulation.

Author Contributions

Conceptualization; Y.S., and H.-S.K.; writing–original draft preparation, J.W.Y., Y.Y.S., and Y.S.; writing—review and editing, H.-S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. NRF-2018R1A5A2023879).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAtopic dermatitis
ASCAdult stem cells
ALPAlkaline phosphatase
BAFFB cell-activating factor
bFGFbasic Fibroblast growth factor
BM-MSCsBone marrow-derived MSCs
BMPBone morphogenic protein
BSPBone sialo protein
cAMPcyclic Adenosine monophosphate
CDCrohn’s disease
CDCluster of differentiation
CMConditioned media
Col IType I collagen
COX-2Cyclooxygenase-2
CTGFConnective tissue growth factor
DFDental follicle
DFPCsDental follicle progenitor cells
DLXDistal-Less Homeobox
DMPDentin matrix acidic phosphoprotein
DPSCsDental pulp stem cells
EMDEnamel matrix derivative
EMVsExtracellular micro-vesicles
ESCsEmbryonic stem cells
FasLFas ligand
FGFFibroblast growth factors
GFAPGlial fibrillary acidic protein
GM-CSFGranulocyte-macrophage colony-stimulating factor
GMSCsGingiva-derived mesenchymal stem cells
HA/TCPHydroxyapatitie/tricalcium phosphate
HLAHuman leukocyte antigen
IBDInflammatory bowel diseases
IDOIndoleamine 2,3-dioxygenase
IFN-γInterferon-γ
IGFInsulin-like growth factor
ILInterleukin
IgEImmunoglobulin E
iNOSinducible Nitric oxide synthase
ISCsIntestinal stem cells
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NG2Neuron-glial antigen 2
MAP2Microtubule-associated proteins
MHCMajor histocompatibility complex
mRNAMessenger RNA
MSMultiple sclerosis
MSCsMesenchymal stromal/stem cells
MTAMineral Trioxide Aggregate
NCSCsNeural crest-derived stem cells
NFICNuclear factor I-C
O4Oligodendrocyte marker
Oct3/4Octamer-binding transcription factor 3/4
OCNOsteocalcin
Olig2Oligodendrocyte transcription factor2
OPCsOligodendrocyte progenitor cells
OPGOsteoprotegerin
OPNOpsteopontin
PBMCPeripheral blood mononuclear cells
PDLPeriodontal ligament
PDLSCsPeriodontal ligament stem cells
PGE2Postaglandin E2
PLGpoly-d,l-lactide and glycolide
PMSCsPeriodontal ligament derived mesenchymal stem cells
RARheumatoid arthritis
RANKNuclear factor κB
RANKLNuclear factor κB ligand
RETRegenerative endodontic treatment
RUNX2Runt-related transcription factor 2
SCAPStem cells from apical papilla
SDFStromal cell-derived factor
sFRPsecreted Frizzled-related protein
SHEDStem cells from human exfoliated deciduous teeth
SPDStem cell sheets derived from minipig deciduous teeth
SSSjogren’s syndrome
TGF-βTransforming growth factor β
ThT helper
TNF-αTumor necrosis factor-α
Tregsregulatory T cells
UCUlcerative colitis
VEGFVascular endothelial growth factor
WIF1Wnt inhibitory factor 1

References

  1. Laurenti, E.; Gottgens, B. From haematopoietic stem cells to complex differentiation landscapes. Nature 2018, 553, 418–426. [Google Scholar] [CrossRef]
  2. Santos, A.J.; Lo, Y.-H.; Mah, A.T.; Kuo, C.J. The intestinal stem cell niche: Homeostasis and adaptations. Trends Cell Biol. 2018, 28, 1062–1078. [Google Scholar] [CrossRef] [PubMed]
  3. Redondo, P.A.; Pavlou, M.; Loizidou, M.; Cheema, U. Elements of the niche for adult stem cell expansion. J. Tissue Eng. 2017, 8. [Google Scholar] [CrossRef] [PubMed]
  4. Regmi, S.; Pathak, S.; Kim, J.O.; Yong, C.S.; Jeong, J.H. Mesenchymal stem cell therapy for the treatment of inflammatory diseases: Challenges, opportunities, and future perspectives. Eur. J. Cell Biol. 2019, 98, 151041. [Google Scholar] [CrossRef] [PubMed]
  5. Seo, Y.; Shin, T.H.; Kim, H.S. Current strategies to enhance adipose stem cell function: An update. Int. J. Mol. Sci. 2019, 20, 3827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. El-Badri, N. Advances in stem cell therapy: Bench to bedside. In Adipose-Derived Stem Cell-Based Therapies in Regenerative Medicine; Springer: Berlin, Germany, 2016; p. 119. [Google Scholar]
  7. Turksen, K. Cell biology and translational medicine, volume 2: Approaches for diverse diseases and conditions. In Stem Cell and Obesity: Current State and Futre Perspective; Springer: Berlin, Germany, 2018; Volume 1089, p. 9. [Google Scholar]
  8. Sharpe, P.T. Dental mesenchymal stem cells. Development 2016, 143, 2273–2280. [Google Scholar] [CrossRef] [Green Version]
  9. Zheng, C.; Chen, J.; Liu, S.; Jin, Y. Stem cell-based bone and dental regeneration: A view of microenvironmental modulation. Int. J. Oral Sci. 2019, 11, 1–15. [Google Scholar] [CrossRef] [Green Version]
  10. Yu, T.; Volponi, A.A.; Babb, R.; An, Z.; Sharpe, P.T. Stem cells in tooth development, growth, repair, and regeneration. Curr. Top. Dev. Biol. 2015, 115, 187–212. [Google Scholar]
  11. Tatullo, M.; Codispoti, B.; Pacifici, A.; Palmieri, F.; Marrelli, M.; Pacifici, L.; Paduano, F. Potential use of human periapical cyst-mesenchymal stem cells (hPCy-MSCs) as a novel stem cell source for regenerative medicine applications. Front. Cell Dev. Biol. 2017, 5, 103. [Google Scholar] [CrossRef] [Green Version]
  12. Puthiyaveetil, J.S.V.; Kota, K.; Chakkarayan, R.; Chakkarayan, J.; Thodiyil, A.K.P. Epithelial–Mesenchymal interactions in tooth development and the significant role of growth factors and genes with emphasis on mesenchyme–A review. J. Clin. Diagn. Res. 2016, 10, ZE05–ZE09. [Google Scholar]
  13. Chai, Y.; Jiang, X.; Ito, Y.; Bringas, P.; Han, J.; Rowitch, D.H.; Soriano, P.; McMahon, A.P.; Sucov, H.M. Fate of the mammalian cranial neural crest during tooth and mandibular morphogenesis. Development 2000, 127, 1671–1679. [Google Scholar] [PubMed]
  14. Liu, J.; Yu, F.; Sun, Y.; Jiang, B.; Zhang, W.; Yang, J.; Xu, G.T.; Liang, A.; Liu, S. Concise reviews: Characteristics and potential applications of human dental tissue-derived mesenchymal stem cells. Stem Cells 2015, 33, 627–638. [Google Scholar] [CrossRef] [PubMed]
  15. França, C.M.; Riggers, R.; Muschler, J.L.; Widbiller, M.; Lococo, P.M.; Diogenes, A.; Bertassoni, L.E. 3D-imaging of whole neuronal and vascular networks of the human dental pulp via clarity and light sheet microscopy. Sci. Rep. 2019, 9, 10860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Gronthos, S.; Mankani, M.; Brahim, J.; Robey, P.G.; Shi, S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 13625–13630. [Google Scholar] [CrossRef] [Green Version]
  17. Rodas-Junco, B.A.; Villicana, C. Dental pulp stem cells: Current advances in isolation, expansion and preservation. Tissue Eng. Regen. Med. 2017, 14, 333–347. [Google Scholar] [CrossRef]
  18. Zavan, B.; Bressan, E. Dental stem cells: Regenerative potential. In Dental Stem Cells (DSCs): Classification and Properties; Springer: Berlin, Germany, 2016; p. 5. [Google Scholar]
  19. Karamzadeh, R.; Eslaminejad, M.B.; Sharifi-Zarchi, A. Comparative in vitro evaluation of human dental pulp and follicle stem cell commitment. Cell J. 2017, 18, 609. [Google Scholar]
  20. Van Pham, P. Stem cells: Biology and engineering. In Dental Pulp Stem Cells and Neurogenesis; Springer: Berlin, Germany, 2018; Volume 1083, p. 66. [Google Scholar]
  21. Nuti, N.; Corallo, C.; Chan, B.; Ferrari, M.; Gerami-Naini, B. Multipotent differentiation of human dental pulp stem cells: A literature review. Stem Cell Rev. Rep. 2016, 12, 511–523. [Google Scholar] [CrossRef]
  22. Ledesma-Martínez, E.; Mendoza-Núñez, V.M.; Santiago-Osorio, E. Mesenchymal stem cells derived from dental pulp: A review. Stem Cells Int. 2016, 2016, 4709572. [Google Scholar] [CrossRef] [Green Version]
  23. Paduano, F.; Marrelli, M.; White, L.J.; Shakesheff, K.M.; Tatullo, M. Odontogenic differentiation of human dental pulp stem cells on hydrogel scaffolds derived from decellularized bone extracellular matrix and collagen type I. PLoS ONE 2016, 11, e0148225. [Google Scholar] [CrossRef] [Green Version]
  24. Mortada, I.; Mortada, R. Dental pulp stem cells and osteogenesis: An update. Cytotechnology 2018, 70, 1479–1486. [Google Scholar] [CrossRef]
  25. Davies, O.; Cooper, P.; Shelton, R.; Smith, A.; Scheven, B. A comparison of the in vitro mineralisation and dentinogenic potential of mesenchymal stem cells derived from adipose tissue, bone marrow and dental pulp. J. Bone Miner. Metab. 2015, 33, 371–382. [Google Scholar] [CrossRef] [PubMed]
  26. Monterubbianesi, R.; Bencun, M.; Pagella, P.; Woloszyk, A.; Orsini, G.; Mitsiadis, T.A. A comparative in vitro study of the osteogenic and adipogenic potential of human dental pulp stem cells, gingival fibroblasts and foreskin fibroblasts. Sci. Rep. 2019, 9, 1761. [Google Scholar] [CrossRef]
  27. Chen, Y.-Y.; He, S.-T.; Yan, F.-H.; Zhou, P.-F.; Luo, K.; Zhang, Y.-D.; Xiao, Y.; Lin, M.-K. Dental pulp stem cells express tendon markers under mechanical loading and are a potential cell source for tissue engineering of tendon-like tissue. Int. J. Oral Sci. 2016, 8, 213–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Uribe-Etxebarria, V.; Luzuriaga, J.; García-Gallastegui, P.; Agliano, A.; Unda, F.; Ibarretxe, G. Notch/Wnt cross-signalling regulates stemness of dental pulp stem cells through expression of neural crest and core pluripotency factors. Eur. Cell Mater. 2017, 34, 249–270. [Google Scholar] [CrossRef] [PubMed]
  29. Zhou, J.; Zhang, Z.; Qian, G. Mesenchymal stem cells to treat diabetic neuropathy: A long and strenuous way from bench to the clinic. Cell Death Discov. 2016, 2, 1–7. [Google Scholar] [CrossRef] [Green Version]
  30. Arthur, A.; Rychkov, G.; Shi, S.; Koblar, S.A.; Gronthos, S. Adult human dental pulp stem cells differentiate toward functionally active neurons under appropriate environmental cues. Stem Cells 2008, 26, 1787–1795. [Google Scholar] [CrossRef] [Green Version]
  31. Liu, J.; Ruan, J.; Weir, M.D.; Ren, K.; Schneider, A.; Wang, P.; Oates, T.W.; Chang, X.; Xu, H.H. Periodontal bone-ligament-cementum regeneration via scaffolds and stem cells. Cells 2019, 8, 537. [Google Scholar] [CrossRef] [Green Version]
  32. Shinagawa-Ohama, R.; Mochizuki, M.; Tamaki, Y.; Suda, N.; Nakahara, T. Heterogeneous human periodontal ligament-committed progenitor and stem cell populations exhibit a unique cementogenic property under in vitro and in vivo conditions. Stem Cells Dev. 2017, 26, 632–645. [Google Scholar] [CrossRef]
  33. Kadokura, H.; Yamazaki, T.; Masuda, Y.; Kato, Y.; Hasegawa, A.; Sakagami, H.; Yokose, S. Establishment of a primary culture system of human periodontal ligament cells that differentiate into cementum protein 1-expressing cementoblast-like cells. In Vivo 2019, 33, 349–352. [Google Scholar] [CrossRef] [Green Version]
  34. Xie, K.; Peng, C. Insulin-like growth factor-1 promotes proliferation, migration and osteoblasts differentiation of periodontal ligament stem cells via activating PI3K/AKT signal pathway. Int. J. Clin. Exp. Pathol. 2017, 10, 6674–6682. [Google Scholar]
  35. Seo, B.-M.; Miura, M.; Gronthos, S.; Bartold, P.M.; Batouli, S.; Brahim, J.; Young, M.; Robey, P.G.; Wang, C.Y.; Shi, S. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004, 364, 149–155. [Google Scholar] [CrossRef]
  36. Trejo Iriarte, C.G.; Ramírez Ramírez, O.; Muñoz García, A.; Verdín Terán, S.L.; Gómez Clavel, J.F. Isolation of periodontal ligament stem cells from extracted premolars. Simplified method. Rev. Mex Biodivers 2017, 21, 13–21. [Google Scholar] [CrossRef]
  37. Bassir, S.H.; Wisitrasameewong, W.; Raanan, J.; Ghaffarigarakani, S.; Chung, J.; Freire, M.; Andrada, L.C.; Intini, G. Potential for stem cell-based periodontal therapy. J. Cell Physiol. 2016, 231, 50–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Menicanin, D.; Mrozik, K.M.; Wada, N.; Marino, V.; Shi, S.; Bartold, P.M.; Gronthos, S. Periodontal-ligament-derived stem cells exhibit the capacity for long-term survival, self-renewal, and regeneration of multiple tissue types in vivo. Stem Cells Dev. 2014, 23, 1001. [Google Scholar] [CrossRef]
  39. Tomokiyo, A.; Yoshida, S.; Hamano, S.; Hasegawa, D.; Sugii, H.; Maeda, H.J.S.c.i. Detection, characterization, and clinical application of mesenchymal stem cells in periodontal ligament tissue. Stem Cells Int. 2018, 2018. [Google Scholar] [CrossRef]
  40. Fujii, S.; Maeda, H.; Tomokiyo, A.; Monnouchi, S.; Hori, K.; Wada, N.; Akamine, A. Effects of TGF-β1 on the proliferation and differentiation of human periodontal ligament cells and a human periodontal ligament stem/progenitor cell line. Cell Tissue Res. 2010, 342, 233–242. [Google Scholar] [CrossRef]
  41. Lee, J.-H.; Um, S.; Jang, J.-H.; Seo, B.M. Effects of VEGF and FGF-2 on proliferation and differentiation of human periodontal ligament stem cells. Cell Tissue Res. 2012, 348, 475–484. [Google Scholar] [CrossRef]
  42. Wada, N.; Tomokiyo, A.; Gronthos, S.; Bartold, P.M. Immunomodulatory properties of PDLSC and relevance to periodontal regeneration. Curr. Oral Health Rep. 2015, 2, 245–251. [Google Scholar] [CrossRef] [Green Version]
  43. Fawzy El-Sayed, K.M.; Dorfer, C.E. Gingival mesenchymal stem/progenitor cells: A unique tissue engineering gem. Stem Cells Int. 2016, 2016, 7154327. [Google Scholar] [CrossRef] [Green Version]
  44. Zhang, Q.; Shi, S.; Liu, Y.; Uyanne, J.; Shi, Y.; Shi, S.; Le, A.D. Mesenchymal stem cells derived from human gingiva are capable of immunomodulatory functions and ameliorate inflammation-related tissue destruction in experimental colitis. J. Immunol. 2009, 183, 7787–7798. [Google Scholar] [CrossRef] [Green Version]
  45. Jin, S.H.; Lee, J.E.; Yun, J.H.; Kim, I.; Ko, Y.; Park, J.B. Isolation and characterization of human mesenchymal stem cells from gingival connective tissue. J. Periodontal Res. 2015, 50, 461–467. [Google Scholar] [CrossRef] [PubMed]
  46. Sun, Q.; Nakata, H.; Yamamoto, M.; Kasugai, S.; Kuroda, S. Comparison of gingiva-derived and bone marrow mesenchymal stem cells for osteogenesis. J. Cell Mol. Med. 2019, 23, 7592–7601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Al Bahrawy, M.; Ghaffar, K.; Gamal, A.; El-Sayed, K.; Iacono, V. Effect of inflammation on gingival mesenchymal stem/progenitor cells’ proliferation and migration through microperforated membranes: An in vitro study. Stem Cells Int. 2020, 5373418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Angelopoulos, I.; Brizuela, C.; Khoury, M. Gingival mesenchymal stem cells outperform haploidentical dental pulp-derived mesenchymal stem cells in proliferation rate, migration ability, and angiogenic potential. Cell Transplant. 2018, 27, 967–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Fournier, B.P.; Ferre, F.C.; Couty, L.; Lataillade, J.J.; Gourven, M.; Naveau, A.; Coulomb, B.; Lafont, A.; Gogly, B. Multipotent progenitor cells in gingival connective tissue. Tissue Eng. Part A 2010, 16, 2891–2899. [Google Scholar] [CrossRef]
  50. Wu, S.M.; Chiu, H.C.; Chin, Y.T.; Lin, H.Y.; Chiang, C.Y.; Tu, H.P.; Fu, M.M.; Fu, E. Effects of enamel matrix derivative on the proliferation and osteogenic differentiation of human gingival mesenchymal stem cells. Stem Cell Res. Ther. 2014, 5, 52. [Google Scholar] [CrossRef] [Green Version]
  51. Zeng, S.J.; Fang, Y.; Zhang, Q.B.; Peng, B.; Zhang, Z.Q.; Zhao, W.H.; Ge, L.H. Effect of basic fibroblast growth factor on the gingiva-derived mesenchymal stem cells. Int. J. Clin. Exp. Med. 2019, 12, 6347–6356. [Google Scholar]
  52. Ansari, S.; Sarrion, P.; Hasani-Sadrabadi, M.M.; Aghaloo, T.; Wu, B.M.; Moshaverinia, A. Regulation of the fate of dental-derived mesenchymal stem cells using engineered alginate-GelMA hydrogels. J. Biomed. Mater. Res. A 2017, 105, 2957–2967. [Google Scholar] [CrossRef]
  53. Chen, Y.; Liu, H. The differentiation potential of gingival mesenchymal stem cells induced by apical tooth germ cellconditioned medium. Mol. Med. Rep. 2016, 14, 3565–3572. [Google Scholar] [CrossRef] [Green Version]
  54. Gao, Y.; Zhao, G.; Li, D.; Chen, X.; Pang, J.; Ke, J. Isolation and multiple differentiation potential assessment of human gingival mesenchymal stem cells. Int. J. Mol. Sci. 2014, 15, 20982–20996. [Google Scholar] [CrossRef] [Green Version]
  55. Zhang, Q.Z.; Nguyen, A.L.; Yu, W.H.; Le, A.D. Human oral mucosa and gingiva: A unique reservoir for mesenchymal stem cells. J. Dent. Res. 2012, 91, 1011–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ge, S.; Mrozik, K.M.; Menicanin, D.; Gronthos, S.; Bartold, P.M. Isolation and characterization of mesenchymal stem cell-like cells from healthy and inflamed gingival tissue: Potential use for clinical therapy. Regen. Med. 2012, 7, 819–832. [Google Scholar] [CrossRef] [PubMed]
  57. Miura, M.; Gronthos, S.; Zhao, M.; Lu, B.; Fisher, L.W.; Robey, P.G.; Shi, S. SHED: Stem cells from human exfoliated deciduous teeth. Proc. Natl. Acad. Sci. USA 2003, 100, 5807–5812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Noda, S.; Kawashima, N.; Yamamoto, M.; Hashimoto, K.; Nara, K.; Sekiya, I.; Okiji, T. Effect of cell culture density on dental pulp-derived mesenchymal stem cells with reference to osteogenic differentiation. Sci. Rep. 2019, 9, 5430. [Google Scholar] [CrossRef]
  59. Naz, S.; Khan, F.R.; Zohra, R.R.; Lakhundi, S.S.; Khan, M.S.; Mohammed, N.; Ahmad, T. Isolation and culture of dental pulp stem cells from permanent and deciduous teeth. Pak. J. Med. Sci. 2019, 35, 997. [Google Scholar] [CrossRef] [Green Version]
  60. Zhang, N.; Chen, B.; Wang, W.; Chen, C.; Kang, J.; Deng, S.Q.; Zhang, B.; Liu, S.; Han, F. Isolation, characterization and multi-lineage differentiation of stem cells from human exfoliated deciduous teeth. Mol. Med. Rep. 2016, 14, 95–102. [Google Scholar] [CrossRef] [Green Version]
  61. Yang, X.; Ma, Y.; Guo, W.; Yang, B.; Tian, W. Stem cells from human exfoliated deciduous teeth as an alternative cell source in bio-root regeneration. Theranostics 2019, 9, 2694–2711. [Google Scholar] [CrossRef]
  62. Rosa, V.; Dubey, N.; Islam, I.; Min, K.-S.; Nör, J.E. Pluripotency of stem cells from human exfoliated deciduous teeth for tissue engineering. Stem Cells Int. 2016, 2016, 5957806. [Google Scholar] [CrossRef] [Green Version]
  63. Shi, X.; Mao, J.; Liu, Y. Pulp stem cells derived from human permanent and deciduous teeth: Biological characteristics and therapeutic applications. Stem Cells Transl. Med. 2020, 9, 445–464. [Google Scholar] [CrossRef] [Green Version]
  64. Almeida, P.N.; Cunha, K.S. Dental stem cells and their application in Dentistry: A literature review. Rev. Bras. Odontol 2016, 73, 331. [Google Scholar] [CrossRef] [Green Version]
  65. Sakai, V.T.; Zhang, Z.; Dong, Z.; Neiva, K.G.; Machado, M.A.; Shi, S.; Santos, C.F.; Nor, J.E. SHED differentiate into functional odontoblasts and endothelium. J. Dent. Res. 2010, 89, 791–796. [Google Scholar] [CrossRef] [PubMed]
  66. Yamaza, T.; Kentaro, A.; Chen, C.; Liu, Y.; Shi, Y.; Gronthos, S.; Wang, S.; Shi, S. Immunomodulatory properties of stem cells from human exfoliated deciduous teeth. Stem Cell Res. Ther. 2010, 1, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Bento, L.; Zhang, Z.; Imai, A.; Nör, F.; Dong, Z.; Shi, S.; Araujo, F.; Nör, J. Endothelial differentiation of SHED requires MEK1/ERK signaling. J. Dent. Res. 2013, 92, 51–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Wang, X.; Sha, X.-J.; Li, G.-H.; Yang, F.-S.; Ji, K.; Wen, L.-Y.; Liu, S.-Y.; Chen, L.; Ding, Y.; Xuan, K. Comparative characterization of stem cells from human exfoliated deciduous teeth and dental pulp stem cells. Arich. Oral Biol. 2012, 57, 1231–1240. [Google Scholar] [CrossRef]
  69. Nakamura, S.; Yamada, Y.; Katagiri, W.; Sugito, T.; Ito, K.; Ueda, M. Stem cell proliferation pathways comparison between human exfoliated deciduous teeth and dental pulp stem cells by gene expression profile from promising dental pulp. J. Endod. 2009, 35, 1536–1542. [Google Scholar] [CrossRef]
  70. Lucaciu, O.; Soriţău, O.; Gheban, D.; Ciuca, D.R.; Virtic, O.; Vulpoi, A.; Dirzu, N.; Câmpian, R.; Băciuţ, G.; Popa, C. Dental follicle stem cells in bone regeneration on titanium implants. BMC Biotechnol. 2015, 15, 114. [Google Scholar] [CrossRef] [Green Version]
  71. Zhou, T.; Pan, J.; Wu, P.; Huang, R.; Du, W.; Zhou, Y.; Wan, M.; Fan, Y.; Xu, X.; Zhou, X. Dental follicle cells: Roles in development and beyond. Stem Cells Int. 2019. [Google Scholar] [CrossRef] [Green Version]
  72. Zhao, M.; Xiao, G.; Berry, J.E.; Franceschi, R.T.; Reddi, A.; Somerman, M.J.; Research, M. Bone morphogenetic protein 2 induces dental follicle cells to differentiate toward a cementoblast/osteoblast phenotype. J. Bone Miner. Res. 2002, 17, 1441–1451. [Google Scholar] [CrossRef]
  73. Müller, P.; Ekat, K.; Brosemann, A.; Köntges, A.; David, R.; Lang, H. Isolation, characterization and MicroRNA-based genetic modification of human dental follicle stem cells. J. Vis. Exp. 2018, e58089. [Google Scholar] [CrossRef] [Green Version]
  74. Rezai-Rad, M.; Bova, J.F.; Orooji, M.; Pepping, J.; Qureshi, A.; Del Piero, F.; Hayes, D.; Yao, S. Evaluation of bone regeneration potential of dental follicle stem cells for treatment of craniofacial defects. Cytotherapy 2015, 17, 1572–1581. [Google Scholar] [CrossRef] [Green Version]
  75. Sowmya, S.; Chennazhi, K.; Arzate, H.; Jayachandran, P.; Nair, S.V.; Jayakumar, R. Periodontal specific differentiation of dental follicle stem cells into osteoblast, fibroblast, and cementoblast. Tissue Eng. Part C 2015, 21, 1044–1058. [Google Scholar] [CrossRef] [PubMed]
  76. Lima, R.L.; Holanda-Afonso, R.C.; Moura-Neto, V.; Bolognese, A.M.; DosSantos, M.F.; Souza, M.M. Human dental follicle cells express embryonic, mesenchymal and neural stem cells markers. Arch. Oral Biol. 2017, 73, 121–128. [Google Scholar] [CrossRef] [PubMed]
  77. Li, C.; Yang, X.; He, Y.; Ye, G.; Li, X.; Zhang, X.; Zhou, L.; Deng, F. Bone morphogenetic protein-9 induces osteogenic differentiation of rat dental follicle stem cells in P38 and ERK1/2 MAPK dependent manner. Int. J. Med. Sci. 2012, 9, 862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Morsczeck, C. Gene expression of runx2, Osterix, c-fos, DLX-3, DLX-5, and MSX-2 in dental follicle cells during osteogenic differentiation in vitro. Calcif. Tissue Int. 2006, 78, 98–102. [Google Scholar] [CrossRef]
  79. Frank, O.; Heim, M.; Jakob, M.; Barbero, A.; Schäfer, D.; Bendik, I.; Dick, W.; Heberer, M.; Martin, I. Real-time quantitative RT-PCR analysis of human bone marrow stromal cells during osteogenic differentiation in vitro. J. Cell Biochem. 2002, 85, 737–746. [Google Scholar] [CrossRef]
  80. Yao, S.; Pan, F.; Prpic, V.; Wise, G. Differentiation of stem cells in the dental follicle. J. Dent. Res. 2008, 87, 767–771. [Google Scholar] [CrossRef] [Green Version]
  81. Sonoyama, W.; Liu, Y.; Fang, D.; Yamaza, T.; Seo, B.M.; Zhang, C.; Liu, H.; Gronthos, S.; Wang, C.Y.; Wang, S.; et al. Mesenchymal stem cell-mediated functional tooth regeneration in swine. PLoS ONE 2006, 1, e79. [Google Scholar] [CrossRef] [Green Version]
  82. Rémy, M.; Ferraro, F.; Le Salver, P.; Rey, S.; Genot, E.; Djavaheri-Mergny, M.; Thébaud, N.; Boiziau, C.; Boeuf, H. Isolation and culture of human stem cells from apical papilla under low oxygen concentration highlight original properties. Cells 2019, 8, 1485. [Google Scholar] [CrossRef] [Green Version]
  83. Bakopoulou, A.; Leyhausen, G.; Volk, J.; Tsiftsoglou, A.; Garefis, P.; Koidis, P.; Geurtsen, W. Comparative analysis of in vitro osteo/odontogenic differentiation potential of human dental pulp stem cells (DPSCs) and stem cells from the apical papilla (SCAP). Arch. Oral Biol. 2011, 56, 709–721. [Google Scholar] [CrossRef]
  84. Jeon, B.G.; Kang, E.J.; Kumar, B.M.; Maeng, G.H.; Ock, S.A.; Kwack, D.O.; Park, B.W.; Rho, G.J. Comparative analysis of telomere length, telomerase and reverse transcriptase activity in human dental stem cells. Cell Transplant. 2011, 20, 1693–1705. [Google Scholar] [CrossRef]
  85. Abe, S.; Hamada, K.; Yamaguchi, S.; Amagasa, T.; Miura, M. Characterization of the radioresponse of human apical papilla-derived cells. Stem Cell Res. Ther. 2011, 2, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Chrepa, V.; Pitcher, B.; Henry, M.A.; Diogenes, A. Survival of the apical papilla and its resident stem cells in a case of advanced pulpal necrosis and apical periodontitis. J. Endod. 2017, 43, 561–567. [Google Scholar] [CrossRef] [PubMed]
  87. Lee, J.-H.; Seo, S.-J. Biomedical application of dental tissue-derived induced pluripotent stem cells. Stem Cells Int. 2016, 2016, 9762465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Nada, O.A.; El Backly, R.M. Stem Cells From the Apical Papilla (SCAP) as a Tool for Endogenous Tissue Regeneration. Front. Bioeng. Biotechnol. 2018, 6, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Kang, J.; Fan, W.; Deng, Q.; He, H.; Huang, F. Stem cells from the apical papilla: A promising source for stem cell-based therapy. BioMed. Res. Int. 2019, 2019, 6104738. [Google Scholar] [CrossRef] [Green Version]
  90. Du, J.; Lu, Y.; Song, M.; Yang, L.; Liu, J.; Chen, X.; Ma, Y.; Wang, Y. Effects of ERK/p38 MAPKs signaling pathways on MTA-mediated osteo/odontogenic differentiation of stem cells from apical papilla: A vitro study. BMC Oral Health 2020, 20, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Wang, J.; Liu, B.; Gu, S.; Liang, J. Effects of Wnt/beta-catenin signalling on proliferation and differentiation of apical papilla stem cells. Cell Prolif. 2012, 45, 121–131. [Google Scholar] [CrossRef]
  92. Yu, G.; Wang, J.; Lin, X.; Diao, S.; Cao, Y.; Dong, R.; Wang, L.; Wang, S.; Fan, Z. Demethylation of SFRP2 by histone demethylase KDM2A regulated osteo-/dentinogenic differentiation of stem cells of the apical papilla. Cell Prolif. 2016, 49, 330–340. [Google Scholar] [CrossRef]
  93. Wang, H.; Cao, Y. WIF1 enhanced dentinogenic differentiation in stem cells from apical papilla. BMC Oral Health 2019, 19, 25. [Google Scholar] [CrossRef]
  94. Thyagarajan, T.; Sreenath, T.; Cho, A.; Wright, J.T.; Kulkarni, A.B. Reduced expression of dentin sialophosphoprotein is associated with dysplastic dentin in mice overexpressing transforming growth factor-beta 1 in teeth. J. Biol. Chem. 2001, 276, 11016–11020. [Google Scholar] [CrossRef] [Green Version]
  95. Yu, S.; Li, J.; Zhao, Y.; Li, X.; Ge, L. Comparative secretome analysis of mesenchymal stem cells from dental apical papilla and bone marrow during early odonto/osteogenic differentiation: Potential role of transforming growth factor-beta2. Front. Physiol. 2020, 11, 41. [Google Scholar] [CrossRef] [PubMed]
  96. Hong, S.; Chen, W.; Jiang, B. A comparative evaluation of concentrated growth factor and platelet-rich fibrin on the proliferation, migration, and differentiation of human stem cells of the apical papilla. J. Endod. 2018, 44, 977–983. [Google Scholar] [CrossRef] [PubMed]
  97. Li, Y.; Yan, M.; Wang, Z.; Zheng, Y.; Li, J.; Ma, S.; Liu, G.; Yu, J. 17beta-estradiol promotes the odonto/osteogenic differentiation of stem cells from apical papilla via mitogen-activated protein kinase pathway. Stem Cell Res. Ther. 2014, 5, 125. [Google Scholar] [CrossRef] [Green Version]
  98. Wang, S.; Mu, J.; Fan, Z.; Yu, Y.; Yan, M.; Lei, G.; Tang, C.; Wang, Z.; Zheng, Y.; Yu, J.; et al. Insulin-like growth factor 1 can promote the osteogenic differentiation and osteogenesis of stem cells from apical papilla. Stem Cell Res. 2012, 8, 346–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. De Berdt, P.; Vanacker, J.; Ucakar, B.; Elens, L.; Diogenes, A.; Leprince, J.G.; Deumens, R.; des Rieux, A. Dental apical papilla as therapy for spinal cord injury. J. Dent. Res. 2015, 94, 1575–1581. [Google Scholar] [CrossRef] [PubMed]
  100. De Almeida, J.F.; Chen, P.; Henry, M.A.; Diogenes, A. Stem cells of the apical papilla regulate trigeminal neurite outgrowth and targeting through a BDNF-dependent mechanism. Tissue Eng. Part A 2014, 20, 3089–3100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Germain, L.; De Berdt, P.; Vanacker, J.; Leprince, J.; Diogenes, A.; Jacobs, D.; Vandermeulen, G.; Bouzin, C.; Preat, V.; Dupont-Gillain, C.; et al. Fibrin hydrogels to deliver dental stem cells of the apical papilla for regenerative medicine. Regen. Med. 2015, 10, 153–167. [Google Scholar] [CrossRef]
  102. Simonovic, J.; Toljic, B.; Nikolic, N.; Peric, M.; Vujin, J.; Panajotovic, R.; Gajic, R.; Bekyarova, E.; Cataldi, A.; Parpura, V.; et al. Differentiation of stem cells from apical papilla into neural lineage using graphene dispersion and single walled carbon nanotubes. J. Biomed. Mater. Res. A 2018, 106, 2653–2661. [Google Scholar] [CrossRef]
  103. Patil, R.; Kumar, B.M.; Lee, W.J.; Jeon, R.H.; Jang, S.J.; Lee, Y.M.; Park, B.W.; Byun, J.H.; Ahn, C.S.; Kim, J.W.; et al. Multilineage potential and proteomic profiling of human dental stem cells derived from a single donor. Exp. Cell Res. 2014, 320, 92–107. [Google Scholar] [CrossRef]
  104. Hilkens, P.; Fanton, Y.; Martens, W.; Gervois, P.; Struys, T.; Politis, C.; Lambrichts, I.; Bronckaers, A. Pro-angiogenic impact of dental stem cells in vitro and in vivo. Stem Cell Res. 2014, 12, 778–790. [Google Scholar] [CrossRef] [Green Version]
  105. Yuan, C.; Wang, P.; Zhu, L.; Dissanayaka, W.L.; Green, D.W.; Tong, E.H.; Jin, L.; Zhang, C. Coculture of stem cells from apical papilla and human umbilical vein endothelial cell under hypoxia increases the formation of three-dimensional vessel-like structures in vitro. Tissue Eng. Part A 2015, 21, 1163–1172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Andrukhov, C.B.; Blufstein, A.; Rausch-Fan, X. Immunomodulatory properties of dental tissue-derived mesenchymal stem cells: Implication in disease and tissue regeneration. World J. Stem Cells 2019, 11, 604–617. [Google Scholar] [CrossRef]
  107. Gao, F.; Chiu, S.M.; Motan, D.A.; Zhang, Z.; Chen, L.; Ji, H.L.; Tse, H.F.; Fu, Q.L.; Lian, Q. Mesenchymal stem cells and immunomodulation: Current status and future prospects. Cell Death Dis. 2016, 7, e2062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Ouchi, T.; Nakagawa, T. Mesenchymal stem cell-based tissue regeneration therapies for periodontitis. Regen. Ther. 2020, 14, 72–78. [Google Scholar] [CrossRef]
  109. Ercal, P.; Pekozer, G.G.; Kose, G.T. Dental stem cells in bone tissue engineering: Current overview and challenges. In Cell Biology and Translational Medicine; Springer: Berlin, Germany, 2018; Volume 3, pp. 113–127. [Google Scholar]
  110. Ogasawara, N.; Kano, F.; Hashimoto, N.; Mori, H.; Yao, L.; Linze, X.; Sakamaki, T.; Hibi, H.; Iwamoto, T.; Tanaka, E. Factors secreted from dental pulp stem cells show multifaceted benefits for treating experimental temporomandibular joint osteoarthritis. Osteoarthr. Cartil. 2020, 28, 831–841. [Google Scholar] [CrossRef] [PubMed]
  111. Xu, Q.L.; Furuhashi, A.; Zhang, Q.Z.; Jiang, C.M.; Chang, T.H.; Le, A.D. Induction of Salivary Gland-Like Cells from Dental Follicle Epithelial Cells. J. Dent. Res. 2017, 96, 1035–1043. [Google Scholar] [CrossRef] [PubMed]
  112. Yokoi, T.; Saito, M.; Kiyono, T.; Iseki, S.; Kosaka, K.; Nishida, E.; Tsubakimoto, T.; Harada, H.; Eto, K.; Noguchi, T.; et al. Establishment of immortalized dental follicle cells for generating periodontal ligament in vivo. Cell Tissue Res. 2007, 327, 301–311. [Google Scholar] [CrossRef]
  113. Li, Y.; He, L.; Pan, S.; Zhang, L.; Zhang, W.; Yi, H.; Niu, Y. Three-dimensional simulated microgravity culture improves the proliferation and odontogenic differentiation of dental pulp stem cell in PLGA scaffolds implanted in mice. Mol. Med. Rep. 2017, 15, 873–878. [Google Scholar] [CrossRef] [Green Version]
  114. Chen, G.; Chen, J.; Yang, B.; Li, L.; Luo, X.; Zhang, X.; Feng, L.; Jiang, Z.; Yu, M.; Guo, W.; et al. Combination of aligned PLGA/Gelatin electrospun sheets, native dental pulp extracellular matrix and treated dentin matrix as substrates for tooth root regeneration. Biomaterials 2015, 52, 56–70. [Google Scholar] [CrossRef]
  115. Guo, W.; Chen, L.; Gong, K.; Ding, B.; Duan, Y.; Jin, Y. Heterogeneous dental follicle cells and the regeneration of complex periodontal tissues. Tissue Eng. Part A 2012, 18, 459–470. [Google Scholar] [CrossRef]
  116. Huang, G.T.; Yamaza, T.; Shea, L.D.; Djouad, F.; Kuhn, N.Z.; Tuan, R.S.; Shi, S. Stem/progenitor cell-mediated de novo regeneration of dental pulp with newly deposited continuous layer of dentin in an in vivo model. Tissue Eng. Part A 2010, 16, 605–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Andrukhov, O.; Behm, C.; Blufstein, A.; Rausch-Fan, X. Immunomodulatory properties of dental-derived mesenchymal stem cells. In Periodontology and Dental Implantology; IntechOpen: London, UK, 2018; pp. 21–49. [Google Scholar]
  118. Liu, J.; Chen, B.; Bao, J.; Zhang, Y.; Lei, L.; Yan, F. Macrophage polarization in periodontal ligament stem cells enhanced periodontal regeneration. Stem Cell Res. Ther. 2019, 10, 320. [Google Scholar] [CrossRef] [PubMed]
  119. Liu, O.; Xu, J.; Ding, G.; Liu, D.; Fan, Z.; Zhang, C.; Chen, W.; Ding, Y.; Tang, Z.; Wang, S. Periodontal ligament stem cells regulate B lymphocyte function via programmed cell death protein 1. Stem Cells 2013, 31, 1371–1382. [Google Scholar] [CrossRef] [PubMed]
  120. Yan, F.; Liu, O.; Zhang, H.; Zhou, Y.; Zhou, D.; Zhou, Z.; He, Y.; Tang, Z.; Wang, S. Human dental pulp stem cells regulate allogeneic NK cells’ function via induction of anti-inflammatory purinergic signalling in activated NK cells. Cell Prolif. 2019, 52, e12595. [Google Scholar] [CrossRef] [Green Version]
  121. Ozdemir, A.T.; Ozgul Ozdemir, R.B.; Kirmaz, C.; Sariboyaci, A.E.; Unal Halbutogllari, Z.S.; Ozel, C.; Karaoz, E. The paracrine immunomodulatory interactions between the human dental pulp derived mesenchymal stem cells and CD4 T cell subsets. Cell Immunol. 2016, 310, 108–115. [Google Scholar] [CrossRef]
  122. Hossein-Khannazer, N.; Hashemi, S.M.; Namaki, S.; Ghanbarian, H.; Sattari, M.; Khojasteh, A. Study of the immunomodulatory effects of osteogenic differentiated human dental pulp stem cells. Life Sci. 2019, 216, 111–118. [Google Scholar] [CrossRef]
  123. Lu, Y.; Xu, Y.; Zhang, S.; Gao, J.; Gan, X.; Zheng, J.; Lu, L.; Zeng, W.; Gu, J. Human gingiva-derived mesenchymal stem cells alleviate inflammatory bowel disease via IL-10 signalling-dependent modulation of immune cells. Scand. J. Immunol. 2019, 90, e12751. [Google Scholar] [CrossRef]
  124. Su, W.R.; Zhang, Q.Z.; Shi, S.H.; Nguyen, A.L.; Le, A.D. Human gingiva-derived mesenchymal stromal cells attenuate contact hypersensitivity via prostaglandin E2-dependent mechanisms. Stem Cells 2011, 29, 1849–1860. [Google Scholar] [CrossRef]
  125. Zhang, Q.Z.; Su, W.R.; Shi, S.H.; Wilder-Smith, P.; Xiang, A.P.; Wong, A.; Nguyen, A.L.; Kwon, C.W.; Le, A.D. Human gingiva-derived mesenchymal stem cells elicit polarization of m2 macrophages and enhance cutaneous wound healing. Stem Cells 2010, 28, 1856–1868. [Google Scholar] [CrossRef] [Green Version]
  126. Jiang, C.M.; Liu, J.; Zhao, J.Y.; Xiao, L.; An, S.; Gou, Y.C.; Quan, H.X.; Cheng, Q.; Zhang, Y.L.; He, W.; et al. Effects of hypoxia on the immunomodulatory properties of human gingiva-derived mesenchymal stem cells. J. Dent. Res. 2015, 94, 69–77. [Google Scholar] [CrossRef]
  127. Ding, G.; Liu, Y.; Wang, W.; Wei, F.; Liu, D.; Fan, Z.; An, Y.; Zhang, C.; Wang, S. Allogeneic periodontal ligament stem cell therapy for periodontitis in swine. Stem Cells 2010, 28, 1829–1838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Loos, B.G.; Van Dyke, T.E. The role of inflammation and genetics in periodontal disease. Periodontol. 2000 2020, 83, 26–39. [Google Scholar] [CrossRef] [PubMed]
  129. Kinane, D.F.; Stathopoulou, P.G.; Papapanou, P.N. Periodontal diseases. Nat. Rev. Dis. Primers 2017, 3, 1–14. [Google Scholar] [CrossRef] [PubMed]
  130. Mira, A.; Simon-Soro, A.; Curtis, M. Role of microbial communities in the pathogenesis of periodontal diseases and caries. J. Clin. Periodontol. 2017, 44, S23–S38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Kurgan, S.; Kantarci, A. Molecular basis for immunohistochemical and inflammatory changes during progression of gingivitis to periodontitis. J. Periodontol. 2018, 76, 51–67. [Google Scholar] [CrossRef] [PubMed]
  132. Sjödin, B.; Haubek, D. Periodontal conditions. In Pediatric Dentistry: A Clinical Approach, 3rd ed.; Wiley-Blackwell: Hoboken, NJ, USA, 2017; pp. 174–192. [Google Scholar]
  133. Artzi, Z.; Sudri, S.; Platner, O.; Kozlovsky, A. Regeneration of the periodontal apparatus in aggressive periodontitis patients. Dent. J. 2019, 7, 29. [Google Scholar] [CrossRef] [Green Version]
  134. Heitz-Mayfield, L.J.; Lang, N.P. Surgical and nonsurgical periodontal therapy. Learned and unlearned concepts. Periodontology 2013, 62, 218–231. [Google Scholar] [CrossRef]
  135. Graziani, F.; Karapetsa, D.; Mardas, N.; Leow, N.; Donos, N. Surgical treatment of the residual periodontal pocket. Periodontology 2018, 76, 150–163. [Google Scholar] [CrossRef] [Green Version]
  136. Yang, B.; Qiu, Y.; Zhou, N.; Ouyang, H.; Ding, J.; Cheng, B.; Sun, J. Application of stem cells in oral disease therapy: Progresses and perspectives. Front. Physiol. 2017, 8, 197. [Google Scholar] [CrossRef] [Green Version]
  137. Liu, Y.; Zheng, Y.; Ding, G.; Fang, D.; Zhang, C.; Bartold, P.M.; Gronthos, S.; Shi, S.; Wang, S. Periodontal ligament stem cell-mediated treatment for periodontitis in miniature swine. Stem Cells 2008, 26, 1065–1073. [Google Scholar] [CrossRef] [Green Version]
  138. Fu, X.; Jin, L.; Ma, P.; Fan, Z.; Wang, S. Allogeneic stem cells from deciduous teeth in treatment for periodontitis in miniature swine. J. Periodontol. 2014, 85, 845–851. [Google Scholar] [CrossRef] [PubMed]
  139. Kinaia, B.M.; Chogle, S.M.; Kinaia, A.M.; Goodis, H.E. Regenerative therapy: A periodontal-endodontic perspective. Dent. Clin. 2012, 56, 537–547. [Google Scholar]
  140. Ji, Y.-M.; Jeon, S.H.; Park, J.-Y.; Chung, J.-H.; Choung, Y.-H.; Choung, P.-H. Dental stem cell therapy with calcium hydroxide in dental pulp capping. Tissue Eng. Part A 2010, 16, 1823–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Leprince, J.G.; Zeitlin, B.D.; Tolar, M.; Peters, O.A. Interactions between immune system and mesenchymal stem cells in dental pulp and periapical tissues. Int. Endod. J. 2012, 45, 689–701. [Google Scholar] [CrossRef] [Green Version]
  142. Nakashima, M.; Iohara, K. Regeneration of dental pulp by stem cells. Adv. Dent. Res. 2011, 23, 313–319. [Google Scholar] [CrossRef]
  143. Iohara, K.; Imabayashi, K.; Ishizaka, R.; Watanabe, A.; Nabekura, J.; Ito, M.; Matsushita, K.; Nakamura, H.; Nakashima, M. Complete pulp regeneration after pulpectomy by transplantation of CD105+ stem cells with stromal cell-derived factor-1. Tissue Eng. Part A 2011, 17, 1911–1920. [Google Scholar] [CrossRef]
  144. Liu, H.-C.; E, L.-L.; Wang, D.-S.; Su, F.; Wu, X.; Shi, Z.-P.; Lv, Y.; Wang, J.-Z. Reconstruction of alveolar bone defects using bone morphogenetic protein 2 mediated rabbit dental pulp stem cells seeded on nano-hydroxyapatite/collagen/poly (L-lactide). Tissue Eng. Part A 2011, 17, 2417–2433. [Google Scholar] [CrossRef]
  145. Ito, K.; Yamada, Y.; Nakamura, S.; Ueda, M. Osteogenic potential of effective bone engineering using dental pulp stem cells, bone marrow stem cells, and periosteal cells for osseointegration of dental implants. Int. J. Oral Maxillofac. Implant. 2011, 26, 947–954. [Google Scholar]
  146. Nyvad, B.; Takahashi, N. Integrated hypothesis of dental caries and periodontal diseases. J. Oral Microbiol. 2020, 12, 1710953. [Google Scholar] [CrossRef] [Green Version]
  147. Mosaddad, S.A.; Tahmasebi, E.; Yazdanian, A.; Rezvani, M.B.; Seifalian, A.; Yazdanian, M.; Tebyanian, H. Oral microbial biofilms: An update. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 1–15. [Google Scholar] [CrossRef]
  148. Sequeira-Byron, P.; Fedorowicz, Z.; Carter, B.; Nasser, M.; Alrowaili, E.F. Single crowns versus conventional fillings for the restoration of root-filled teeth. Cochrane Database Syst. Rev. 2012, 16, CD009109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Halaj-Mofrad, A.; Parirokh, M.; Kashi, N. Surgical replacement of huge rate of root end amalgam filling with mineral trioxide aggregate angelus: A case report. Community Dent. Oral Epidemiol. 2017, 7, 47–51. [Google Scholar]
  150. Mazur, B.; Massler, M. Influence of periodontal disease on the dental pulp. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 1964, 17, 592–603. [Google Scholar] [CrossRef]
  151. Hargreaves, K.M.; Cohen, S.; Berman, L.H.; Rotstein, I. Cohen’s Pathways of the Pulp Expert Consult; Elsevier Health Sciences: Amsterdam, The Netherlands, 2016; p. 928. [Google Scholar]
  152. Barbato, L.; Francioni, E.; Bianchi, M.; Mascitelli, E.; Marco, L.B.; Tonelli, D.P. Periodontitis and bone metabolism. Clin. Cases Miner. Bone Metab. 2015, 12, 174. [Google Scholar] [CrossRef] [PubMed]
  153. Bartold, P.M.; Cantley, M.D.; Haynes, D.R. Mechanisms and control of pathologic bone loss in periodontitis. Periodontology 2010, 53, 55–69. [Google Scholar] [CrossRef] [PubMed]
  154. Mooney, D.J.; Vandenburgh, H. Cell delivery mechanisms for tissue repair. Cell Stem Cell 2008, 2, 205–213. [Google Scholar] [CrossRef] [Green Version]
  155. Otaki, S.; Ueshima, S.; Shiraishi, K.; Sugiyama, K.; Hamada, S.; Yorimoto, M.; Matsuo, O. Mesenchymal progenitor cells in adult human dental pulp and their ability to form bone when transplanted into immunocompromised mice. Cell Biol. Int. 2007, 31, 1191–1197. [Google Scholar] [CrossRef]
  156. D’Aquino, R.; Papaccio, G.; Laino, G.; Graziano, A. Dental pulp stem cells: A promising tool for bone regeneration. Stem Cell Rev. 2008, 4, 21–26. [Google Scholar] [CrossRef]
  157. De Mendonça Costa, A.; Bueno, D.F.; Martins, M.T.; Kerkis, I.; Kerkis, A.; Fanganiello, R.D.; Cerruti, H.; Alonso, N.; Passos-Bueno, M.R. Reconstruction of large cranial defects in nonimmunosuppressed experimental design with human dental pulp stem cells. J. Craniofac. Surg. 2008, 19, 204–210. [Google Scholar] [CrossRef] [Green Version]
  158. Chan, B.; Wong, R.; Rabie, B. In vivo production of mineralised tissue pieces for clinical use: A qualitative pilot study using human dental pulp cell. Int. J. Oral Maxillofac. Surg. 2011, 40, 612–620. [Google Scholar] [CrossRef]
  159. Riccio, M.; Maraldi, T.; Pisciotta, A.; La Sala, G.B.; Ferrari, A.; Bruzzesi, G.; Motta, A.; Migliaresi, C.; De Pol, A. Fibroin scaffold repairs critical-size bone defects in vivo supported by human amniotic fluid and dental pulp stem cells. Tissue Eng. Part A 2012, 18, 1006–1013. [Google Scholar] [CrossRef] [PubMed]
  160. Yamada, Y.; Ito, K.; Nakamura, S.; Ueda, M.; Nagasaka, T. Promising cell-based therapy for bone regeneration using stem cells from deciduous teeth, dental pulp, and bone marrow. Cell Transplant. 2011, 20, 1003–1013. [Google Scholar] [CrossRef] [PubMed]
  161. El Moshy, S.; Radwan, I.A.; Rady, D.; Abbass, M.M.S.; El-Rashidy, A.A.; Sadek, K.M.; Dorfer, C.E.; Fawzy El-Sayed, K.M. Dental stem cell-derived secretome/conditioned medium: The future for regenerative therapeutic applications. Stem Cells Int. 2020, 2020, 7593402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Nakayama, H.; Iohara, K.; Hayashi, Y.; Okuwa, Y.; Kurita, K.; Nakashima, M. Enhanced regeneration potential of mobilized dental pulp stem cells from immature teeth. Oral Dis. 2017, 23, 620–628. [Google Scholar] [CrossRef]
  163. Kumar, A.; Kumar, V.; Rattan, V.; Jha, V.; Bhattacharyya, S. Secretome proteins regulate comparative osteogenic and adipogenic potential in bone marrow and dental stem cells. Biochimie 2018, 155, 129–139. [Google Scholar] [CrossRef]
  164. Diomede, F.; D’Aurora, M.; Gugliandolo, A.; Merciaro, I.; Ettorre, V.; Bramanti, A.; Piattelli, A.; Gatta, V.; Mazzon, E.; Fontana, A.; et al. A novel role in skeletal segment regeneration of extracellular vesicles released from periodontal-ligament stem cells. Int. J. Nanomed. 2018, 13, 3805–3825. [Google Scholar] [CrossRef] [Green Version]
  165. Mitsiadis, T.A.; Feki, A.; Papaccio, G.; Caton, J. Dental pulp stem cells, niches, and notch signaling in tooth injury. Adv. Dent. Res. 2011, 23, 275–279. [Google Scholar] [CrossRef]
  166. Joo, K.H.; Song, J.S.; Kim, S.; Lee, H.S.; Jeon, M.; Kim, S.O.; Lee, J.H. Cytokine Expression of Stem Cells Originating from the Apical Complex and Coronal Pulp of Immature Teeth. J. Endod. 2018, 44, 87–92. [Google Scholar] [CrossRef] [Green Version]
  167. De Cara, S.; Origassa, C.S.T.; de Sa Silva, F.; Moreira, M.; de Almeida, D.C.; Pedroni, A.C.F.; Carvalho, G.L.; Cury, D.P.; Camara, N.O.S.; Marques, M.M. Angiogenic properties of dental pulp stem cells conditioned medium on endothelial cells in vitro and in rodent orthotopic dental pulp regeneration. Heliyon 2019, 5, e01560. [Google Scholar] [CrossRef] [Green Version]
  168. Huang, C.C.; Narayanan, R.; Alapati, S.; Ravindran, S. Exosomes as biomimetic tools for stem cell differentiation: Applications in dental pulp tissue regeneration. Biomaterials 2016, 111, 103–115. [Google Scholar] [CrossRef] [Green Version]
  169. Merckx, G.; Hosseinkhani, B.; Kuypers, S.; Deville, S.; Irobi, J.; Nelissen, I.; Michiels, L.; Lambrichts, I.; Bronckaers, A. Angiogenic effects of human dental pulp and bone marrow-derived mesenchymal stromal cells and their extracellular vesicles. Cells 2020, 9, 312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Fukui, S.; Iwamoto, N.; Masuyama, R.; Kosai, K.; Yanagihara, K.; Kawakami, A. FRI0017 A novel concept of M1 and M2 monocytes in rheumatoid arthritis: Pro-inflammatory monocyte polarization imbalance, anti-citrullinated protein antibody and osteoclastogenesis. BMJ Publishing Group Ltd.: London, UK, 2017; p. 486. [Google Scholar]
  171. Noack, M.; Miossec, P. Selected cytokine pathways in rheumatoid arthritis. Semin. Immunopathol. 2017, 39, 365–383. [Google Scholar] [CrossRef] [PubMed]
  172. Avci, A.; Feist, E.; Burmester, G. Targeting GM-CSF in rheumatoid arthritis. Clin. Exp. Rheumatol. 2016, 34, 39–44. [Google Scholar] [PubMed]
  173. Narazaki, M.; Tanaka, T.; Kishimoto, T. The role and therapeutic targeting of IL-6 in rheumatoid arthritis. Expert Rev. Clin. Immunol. 2017, 13, 535–551. [Google Scholar] [CrossRef]
  174. Ishikawa, J.; Takahashi, N.; Matsumoto, T.; Yoshioka, Y.; Yamamoto, N.; Nishikawa, M.; Hibi, H.; Ishigro, N.; Ueda, M.; Furukawa, K. Factors secreted from dental pulp stem cells show multifaceted benefits for treating experimental rheumatoid arthritis. Bone 2016, 83, 210–219. [Google Scholar] [CrossRef]
  175. Gu, Y.; Shi, S. Transplantation of gingiva-derived mesenchymal stem cells ameliorates collagen-induced arthritis. Arthritis Res. Ther. 2016, 18, 262. [Google Scholar] [CrossRef] [Green Version]
  176. Luo, Y.; Wu, W.; Gu, J.; Zhang, X.; Dang, J.; Wang, J.; Zheng, Y.; Huang, F.; Yuan, J.; Xue, Y.; et al. Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritis. EBioMedicine 2019, 43, 620–631. [Google Scholar] [CrossRef] [Green Version]
  177. Zibandeh, N.; Deniz, G.; Ozgen, Z.; Duran, Y.; Kasap, N.; Goker, K.; Baris, S.; Ergun, T.; Akkoc, T. Effect of dental follicle mesenchymal stem cell on Th1 and Th2 derived naive T cells in atopic dermatitis patients. Clin. Exp. Health Sci. 2019, 9, 220–227. [Google Scholar] [CrossRef] [Green Version]
  178. Rajan, T.S.; Giacoppo, S.; Diomede, F.; Ballerini, P.; Paolantonio, M.; Marchisio, M.; Piattelli, A.; Bramanti, P.; Mazzon, E.; Trubiani, O. The secretome of periodontal ligament stem cells from MS patients protects against EAE. Sci. Rep. 2016, 6, 38743. [Google Scholar] [CrossRef] [Green Version]
  179. Janebodin, K.; Reyes, M. Neural crest-derived dental pulp stem cells function as ectomesenchyme to support salivary gland tissue formation. Dentistry 2012, 13, 5. [Google Scholar]
  180. Yamazaki, Y.; Nakamura, Y.; Núñez, G. Role of the microbiota in skin immunity and atopic dermatitis. Allergol. Int. 2017, 66, 539–544. [Google Scholar] [CrossRef]
  181. Furue, M.; Chiba, T.; Tsuji, G.; Ulzii, D.; Kido-Nakahara, M.; Nakahara, T.; Kadono, T. Atopic dermatitis: Immune deviation, barrier dysfunction, IgE autoreactivity and new therapies. Allergol. Int. 2017, 66, 398–403. [Google Scholar] [CrossRef] [PubMed]
  182. Su, C.; Yang, T.; Wu, Z.; Zhong, J.; Huang, Y.; Huang, T.; Zheng, E. Differentiation of T-helper cells in distinct phases of atopic dermatitis involves Th1/Th2 and Th17/Treg. Eur. J. Inflamm. 2017, 15, 46–52. [Google Scholar] [CrossRef] [Green Version]
  183. Heeringa, J.J.; Rijvers, L.; Arends, N.; Driessen, G.; Pasmans, S.; Van Dongen, J.; De Jongste, J.; Van Zelm, M. IgE-expressing memory B cells and plasmablasts are increased in blood of children with asthma, food allergy, and atopic dermatitis. Allergy 2018, 73, 1331–1336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Go, H.-N.; Lee, S.-H.; Cho, H.-J.; Ahn, J.-R.; Kang, M.-J.; Lee, S.-Y.; Hong, S.-J. Effects of chloromethylisothiazolinone/methylisothiazolinone (CMIT/MIT) on Th2/Th17-related immune modulation in an atopic dermatitis mouse model. Sci. Rep. 2020, 10, 1–9. [Google Scholar] [CrossRef] [Green Version]
  185. Jones, A.P.; Kermode, A.G.; Lucas, R.M.; Carroll, W.M.; Nolan, D.; Hart, P. Circulating immune cells in multiple sclerosis. Clin. Exp. Immunol. 2017, 187, 193–203. [Google Scholar] [CrossRef] [Green Version]
  186. Wu, Q.; Wang, Q.; Mao, G.; Dowling, C.A.; Lundy, S.K.; Mao-Draayer, Y. Dimethyl fumarate selectively reduces memory T cells and shifts the balance between Th1/Th17 and Th2 in multiple sclerosis patients. J. Immunol. 2017, 198, 3069–3080. [Google Scholar] [CrossRef] [Green Version]
  187. White, M.P.; Webster, G.; Leonard, F.; La Flamme, A.C. Innate IFN-γ ameliorates experimental autoimmune encephalomyelitis and promotes myeloid expansion and PDL-1 expression. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef]
  188. Moayeri, A.; Bojnordi, M.N.; Haratizadeh, S.; Esmaeilnejad-Moghadam, A.; Alizadeh, R.; Hamidabadi, H. Transdifferentiation of human dental pulp stem cells into oligoprogenitor cells. Basic Clin. Neurosci. 2017, 8, 387–394. [Google Scholar] [CrossRef] [Green Version]
  189. Bi, R.; Zeng, S.-P.; Wen, B.; Wang, C.-B.; Tan, J.-H. Study on immunological characteristics of T lymphocyte in peripheral blood from patients with Sjogren’s syndrome. TMR Integr. Med. 2019, 3, 88888-e19015. [Google Scholar]
  190. Nakamura, H.; Horai, Y.; Shimizu, T.; Kawakami, A. Modulation of apoptosis by cytotoxic mediators and cell-survival molecules in Sjögren’s syndrome. Int. J. Mol. Sci. 2018, 19, 2369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Ainola, M.; Porola, P.; Takakubo, Y.; Przybyla, B.; Kouri, V.; Tolvanen, T.; Hänninen, A.; Nordström, D. Activation of plasmacytoid dendritic cells by apoptotic particles–mechanism for the loss of immunological tolerance in Sjögren’s syndrome. Clin. Exp. Immunol. 2018, 191, 301–310. [Google Scholar] [CrossRef] [Green Version]
  192. Thompson, N.; Isenberg, D.A.; Jury, E.C.; Ciurtin, C. Exploring BAFF: Its expression, receptors and contribution to the immunopathogenesis of Sjögren’s syndrome. Rheumatology 2016, 55, 1548–1555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Verstappen, G.M.; Meiners, P.M.; Corneth, O.B.; Visser, A.; Arends, S.; Abdulahad, W.H.; Hendriks, R.W.; Vissink, A.; Kroese, F.G.; Bootsma, H. Attenuation of follicular helper T cell–dependent B cell hyperactivity by abatacept treatment in primary Sjögren’s syndrome. Arthritis Rheumatol. 2017, 69, 1850–1861. [Google Scholar] [CrossRef] [PubMed]
  194. Weißenberg, S.Y.; Szelinski, F.; Schrezenmeier, E.; Stefanski, A.-L.; Wiedemann, A.; Welle, A.; Jungmann, A.; Nordström, K.; Walter, J.; Imgenberg-Kreuz, J. Identification and characterization of post-activated B cells in systemic autoimmune diseases. Front. Immunol. 2019, 10, 2136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Dias, A.M.; Correia, A.; Pereira, M.S.; Almeida, C.R.; Alves, I.; Pinto, V.; Catarino, T.A.; Mendes, N.; Leander, M.; Oliva-Teles, M.T. Metabolic control of T cell immune response through glycans in inflammatory bowel disease. Proc. Natl. Acad. Sci. USA 2018, 115, E4651–E4660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Xia, S.; Xue, Z.; Cai, Z.; Xia, X.; Tang, Y.; Jiang, Y. Imbalance among Th1, Th2 and Th17 Cells and Crohn’s Disease. Chin. J. Dig. Dis. 2017, 22, 331–336. [Google Scholar]
  197. Hao, W.U.; Xia, S.L.; Xia, X.P.; Xu, C.L.; Hu, D.Y.; Shao, X.X.; Jiang, Y. Association of ulcerative colitis with the imbalance between Th1, Th2 and Th17 cells in the colonic tissues. Med. J. Chin. PLA 2017, 42, 793–798. [Google Scholar]
  198. Friedrich, M.; Pohin, M.; Powrie, F. Cytokine networks in the pathophysiology of inflammatory bowel disease. Immunity 2019, 50, 992–1006. [Google Scholar] [CrossRef] [Green Version]
  199. Singh, U.P.; Singh, N.P.; Murphy, E.A.; Price, R.L.; Fayad, R.; Nagarkatti, M.; Nagarkatti, P.S. Chemokine and cytokine levels in inflammatory bowel disease patients. Cytokine 2016, 77, 44–49. [Google Scholar] [CrossRef] [Green Version]
  200. Mlakar, A.S.; Hojsak, I.; Jergović, M.; Čimić, S.; Bendelja, K. Pediatric Crohn disease is characterized by Th1 in the terminal ileum and Th1/Th17 immune response in the colon. Eur. J. Pediatr 2018, 177, 611–616. [Google Scholar] [CrossRef] [PubMed]
Table 1. Main characteristics of various dental mesenchymal stromal/stem cells (MSCs).
Table 1. Main characteristics of various dental mesenchymal stromal/stem cells (MSCs).
MSCOriginFirst IsolationPrimary FunctionIn Vitro DifferentiationMain Characteristics
DPSCDental pulp of permanent teeth2000Dentin formationOsteoblast
Adipocyte
Odontoblast
Neural crest cells
Express neurogenic markers
Express pluripotent markers
Higher proliferation capacity and odontogenic capacity than BM-MSCs
PDLSCPeriodontal ligament2004Tooth supportOsteoblast
Adipocyte
Fibroblast
Cementoblast
Share similarities with pericyte
Expression of PDL-function proteins
generate the cementum-PDL structure in vivo
GMSCLaminar propria of gingiva2009Tooth supportWound healingOsteoblast
Adipocyte
Chondrocyte
Neural cells
Endothelial cells
Express pluripotent markers
Higher proliferation capacity than BM-MSCs
SHEDPulpal tissue of deciduous teeth2003Secrete mineralizable dentin matricesOsteoblast
Adipocyte
Odontoblast
Neural cells
Hepatocyte
Endothelial cells
Express neurogenic markers
Express pluripotent markers
Higher proliferation capacity than DPSCs and BM-MSCs
DFSCDental follicle2002Tooth developmentOsteoblasts
Adipocyte
Cementoblast
Neural cell
Higher immunomodulatory effects than other ASCs
Regulate the osteoclastogenesis and osteogenesis
APSCApical papilla of an immature tooth root2006The primary source of odontoblasts at root regionOsteoblast
Adipocyte
Odontoblast
Neural cells
Hepatocyte
Express pluripotent markers
Highly expandable than other dental MSCs
Table 2. Therapeutic application of dental MSCs to induce dental regeneration.
Table 2. Therapeutic application of dental MSCs to induce dental regeneration.
MSC TypeModel/ConditionInjection RouteMain EffectsRef.
DPSCImmuno-deficient athymic miceXenograftDifferentiated into cementoblast-like cells
Differentiated into adipocytes and collagen forming cells
[139]
Tooth defects in beagle dogsAutograftDentin regeneration ↑
Pulp capping regeneration ↑
[140]
Begie XIDIII nu/nu miceSC injectionDental tissue inflammation ↓
Pro-inflammatory cytokine secretion ↓
[141]
Canine pulpitis modelSC injectionDental pulp regeneration ↑
Present angiogenic/vasculogenic/neurogenic potential
[142]
Apical closure in dogsAutologous transplantationHighly expressed angiogenic/neurotrophic factors
Neovascularization ↑
Dental pulp regeneration ↑
[143]
Aveolar bone defects in New Zealand rabbitAutologous transplantationAlveolar bone regeneration ↑
Expressing STRO-1 and vimentin
[144]
Bone defects in dogsAutologous transplantationOsteogenic potential ↑[145]
PDLSCPeriodontitis in Miniature SwineFlap surgeryPeriodontal tissue regeneration ↑
Alveolar bone regeneration ↑
T Cell Proliferation ↓
[137]
[127]
SHEDPeriodontitis in Miniature SwineFlap surgeryPeriodontal bone regeneration ↑[138]
Table 3. Immunomodulatory capacity of dental MSCS targeting autoimmune diseases.
Table 3. Immunomodulatory capacity of dental MSCS targeting autoimmune diseases.
MSC TypeTarget DiseaseModelInjection RouteIn Vitro EffectsIn Vivo EffectsRef.
SHED (CM)RACIA mouseI.VM2 polarization↑
RANKL-induced Osteoclastogenesis↓
M2 polarization↑
M2 cell markers↑
RANKL expression↓
Osteoclastogenesis↓
Swelling↓
[174]
GMSCRACIA mouseI.VInduces T cell apoptosis
AnnexinV+, 7AAD+
Th1/Th17 polarization↓
Treg polarization↑
IgG, TNF-α expression↓
Immunosuppressive↑
[175]
GMSCRACIA mouseI.VNF-κB P65/P50↓
Osteoclastogenesis↓
Th1/Th17 polarization↓
TNF-α expression↓
Pro-inflammatory cytokine↓
RANKL expression↓
Osteoclastogenesis↓
CD39, CD73, TGFβR1, MAPK expression↑
[176]
DFSCADpatients PBMCsCD4+, CD8+ T cell proliferation↓
IL-4 expression↓
IL-10 expression↑
[177]
PDLSC
(CM)
MSMSI.VBody weight↑
IL-17, IFN-γ, IL-1β, IL-6, TNF-α expression↓
IL-10/TGF-β expression↑
STAT1, p53, caspase-3, Bax expression↓
[178]
DPSCSSRag1 null miceS.C.Acinar differentiation↑FGF-7/10, NF-200, VEGFR-3, VEGF-C,
AMY-1 level↑
[179]
GMSCIBDDSS modelS.C.PBMC proliferation↓
IDO, IL-10 expression↑
IL-10, FoxP3 expression↑
CD4+ T cell infiltration↓
Treg cell infiltration↑
[44]

Share and Cite

MDPI and ACS Style

Yang, J.W.; Shin, Y.Y.; Seo, Y.; Kim, H.-S. Therapeutic Functions of Stem Cells from Oral Cavity: An Update. Int. J. Mol. Sci. 2020, 21, 4389. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21124389

AMA Style

Yang JW, Shin YY, Seo Y, Kim H-S. Therapeutic Functions of Stem Cells from Oral Cavity: An Update. International Journal of Molecular Sciences. 2020; 21(12):4389. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21124389

Chicago/Turabian Style

Yang, Ji Won, Ye Young Shin, Yoojin Seo, and Hyung-Sik Kim. 2020. "Therapeutic Functions of Stem Cells from Oral Cavity: An Update" International Journal of Molecular Sciences 21, no. 12: 4389. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21124389

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop