CC BY 4.0 · Eur J Dent 2024; 18(01): 001-013
DOI: 10.1055/s-0042-1759487
Review Article

Stem Cells in the Periodontium—Anatomically Related Yet Physiologically Diverse

1   Department of Periodontics and Oral Implantology, SRM Dental College and Hospital, Ramapuram, Chennai, Tamil Nadu, India
,
1   Department of Periodontics and Oral Implantology, SRM Dental College and Hospital, Ramapuram, Chennai, Tamil Nadu, India
,
Dhayanand John Victor
1   Department of Periodontics and Oral Implantology, SRM Dental College and Hospital, Ramapuram, Chennai, Tamil Nadu, India
,
Ganesh Shetty
2   Dental and Orthodontic Clinic, Bangalore, Karnataka, India
› Author Affiliations
 

Abstract

Periodontitis is a complex chronic disease discernible by the deterioration of periodontal tissue. The goal of periodontal therapy is to achieve complete tissue regeneration, and one of the most promising treatment options is to harness the regenerative potential of stem cells available within the periodontal complex. Periodontal ligament stem cells, gingival mesenchymal stem cells, oral periosteal stem cells, and dental follicle stem cells have structural similarities, but their immunological responses and features differ. The qualities of diverse periodontal stem cells, their immune-modulatory effects, and variances in their phenotypes and characteristics will be discussed in this review. Although there is evidence on each stem cell population in the periodontium, understanding the differences in markers expressed, the various research conducted so far on their regenerative potential, will help in understanding which stem cell population will be a better candidate for tissue engineering. The possibility of selecting the most amenable stem cell population for optimal periodontal regeneration and the development and current application of superior tissue engineering treatment options such as autologous transplantation, three-dimensional bioengineered scaffolds, dental stem cell-derived extracellular vesicles will be explored.


#

Introduction

Periodontal disease is a chronic inflammatory disease characterized by a dysregulation of balance between the native oral commensals and the pathogenic microorganisms, leading to activation of the inflammatory cascade thereby causing host mediated destruction of the periodontal soft and hard tissues.[1] Nonsurgical periodontal therapy (NSPT) primarily aims at regulating the immune-inflammatory profile by mechanical debridement. Despite this, in approximately 67% of instances, disease persists even after NSPT owing to areas of persistent pockets that do not allow complete resolution of inflammation, thereby warranting surgical treatment.[2]

Traditional surgical periodontal therapies rely on synthetic materials and biological agents for regeneration, although their effectiveness is debatable due to lack of histological evidence of regeneration.[3] Tissue engineering with a triad of cells cultivated on a scaffold with suitable biophysical and chemical cues to finally rebuild the lost tissues has been proposed for attaining optimal regeneration.[4]

Stem cells have been considered a promising approach for regeneration as they have unique properties of stemness, migration, differentiation, and immune modulation.[5] Traditionally, stem cells have been harvested from the dental pulp and the exfoliated deciduous teeth; however, recently the use of stem cells sourced from the periodontium has been advocated as they are a reservoir of highly undifferentiated cells that can migrate to regenerate the lost periodontium. Mesenchymal stem cells from periodontal tissues such as periodontal ligament stem cells (PDLSCs), gingival mesenchymal stem cells (GMSCs), oral periosteal stem cells (OPSCs), and dental follicle stem cells (DFSCs) have been tested with varying results in vitro and in vivo for regeneration. The best possible stem cell needs to be assessed and compared to aid in the selection of the candidate cell to achieve complete regeneration. This review summarizes the properties of each unique stem cell population harvested within the periodontium and their regenerative potential.


#

Origin and Distinct Phenotypes of Stem Cells within the Periodontium

The periodontal complex's postnatal root development that parallels the tooth growth process heightens the possibility of a bountiful supply of dental stem cells that are more embryonic in nature than other dental stem cell sources. The nomenclature of periodontal MSCs is strongly linked to their tissue origins ([Fig. 1]). PDLSCs are produced from ectomesenchymal cells originating from the neural crest and are principally extracted from the mid-third of the root surface post extraction of permanent teeth.[6] The stem cells isolated from root surface are termed root surface derived PDLSCs (r-PDLSCs) and the stem cells isolated from the tissue collected from the bone surface are called alveolar socket derived PDLSCs (a-PDLSCs). It has been observed that a-PDLSCs retain more proliferative capacity, high osteogenic, and adipogenic potential compared to r-PDLSCs.[7] Seo et al, 2004, found that PDLSCs have the ability to differentiate into periodontal ligament, alveolar bone, cementum, peripheral nerves, and even blood vessels. PDLSCs are found in the periodontal ligament and the developing follicle of permanent teeth.[8]

Zoom Image
Fig. 1 Origin and development of stem cells within the periodontium. (A) The neural crest ectomesenchyme and the neural crest cells—The thickening of ectomesenchyme with migration of neural epithelial cells form a tooth bud. (B) Condensation of mesenchyme to form dental papilla and follicle giving rise to dental follicle stem cells. Inner layer of follicle (dental follicle proper) gives rise to periodontal ligament cells, the denotgingival fiber system harboring periodontal ligament stem cells (PDLSCs) and alveolar bone harboring oral periosteal stem cells (OPSCs), outer layer of follicle (perifollicular mesenchyme) forms gingival lamina propria that harbors gingival mesenchymal stem cells (GMSCs). (C) Mature tooth showing location of PDLSCs, GMSCs, and OPSCs.

Gingiva, the most important periodontal organ, is the next source of stem cells that originate from the neural crest and even from the bone marrow. GMSCs are extracted from gingival tissue samples acquired during gingivectomy procedures and de-epithelialized to leave only connective tissue.[9] Neural-crest-derived GMSCs (N-GMSCs) and mesoderm-derived GMSCs (M-GMSCs) are two subpopulations of gingival mesenchymal stem/progenitor cells, with N-GMSCs having a stronger ability to develop into neural cells than M-GMSCs.[10]

Alternatively, a loose ectomesenchyme-derived connective tissue called dental follicle surrounds the enamel organ and dental papilla of the growing tooth germ. The DFSCs are comprised of a series of pluripotent stem cells formed from neural crest cells originating from the ectoderm.[11] Morsczeck was the first to discover that periodontal tissue progenitors were present in dental follicular cells, known to regulate osteoclastogenesis and osteogenesis that is vital for tooth eruption coordination, in addition to its role in periodontal development.[12] OPSCs are derived from the periosteum, a complex structure that includes undifferentiated mesenchymal cells and envelopes the bone.[13] The periosteal stem cells regulate chondrogenesis and osteogenesis that can be exploited for the maintenance of bone mass in both physiological remodeling and in periodontal surgical healing process.

Dental tissues in the craniofacial complex have become a readily available source of MSCs with multilineage differentiation capabilities comparable to bone marrow MSCs.[14] Despite the fact that there are numerous dental stem cell populations, periodontal stem cells have recently attracted a lot of attention because they are native to the periodontal complex and can be induced to accomplish regeneration.[15] MSCs from the periodontal complex have morphology and marker expression that are very similar to fibroblasts, despite the fact that they only make up around 1% of the cell culture population. There are two main methods for isolating SCs from the periodontium, and both have been successfully employed to isolate MSCs from oral tissues, including gingiva for cell culture.

The first method entails growing cells from a tissue sample in plastic-adherent fibroblast: explant culture method or by enzymatic release method.[16] The second method includes MSCs sorting from parental fibroblast cultures or cell population enzymatically liberated directly from connective tissue biopsy based on a panel of preselected cell surface markers utilized to isolate MSCs from oral and other tissues. This method of prospective separation is based on the ability of the chosen markers to accurately identify MSCs.[17] Based on these above techniques, MSCs can be identified within the periodontium and probably the biggest colonies may be identified as putative MSCs with highest potential for proliferation and self-renewal.

Distinct Phenotypes of Stem Cells within the Periodontium:

The periodontal stem cells originate from closely related tissues; however, there is wide difference in the type of markers they express, which determines the unique phenotype and differentiation capacity of each of these stem cells ([Table 1]).

Table 1

Phenotypic expression and regenerative potential of periodontal stem cells

Stem cell type

Markers expressed

In vivo tissue formation

In vitro tissue

formation

Reference

Neural crest

Embryonic

CD antigen (+) / (-)

PDLSC

Sox 10, P75NTvnR, Snail, Twist,

Sox-9, CD49d

Nanog, Sox 2, SSEA4

Oct 4,

Klf4

CD-9 + , CD-10 + , CD-13 + , CD-29 + ,

CD-44 + , CD-59 + , CD-73 + , CD-90 + , CD-105 + , CD-106 + , CD-146 + , CD-166+ CD14-, CD31-, CD34-,CD45-

Cementum, PDL, adipose, dentin, bone

Osteo, adipo, chondro, myo, neuro, cardiomyo, HLC, melanocyte

[18]

GMSC

Snail1, Twist 1, Sox 9, NES, FoxD3, PAX3

Nanog,

Sox-2

SSEA3

CD-29 + , CD-44 + , CD-73 + , CD-90 + , CD-105 + , CD-106 + , CD-146 + , CD-166+

CD34-, CD45-,CD117-

Cartilage, bone, muscle

Adiopo, chondro,

osteo, neuro, endothelial cells

.[19]

DFSC

HNK1, NES, P75NTR, Nestin, βIII-Tubulin

Oct 4,

Sox 2,

Nanog

CD-13 + ,CD-29 + , CD-9 + , CD10 + ,

CD-44 + , CD-59 + , D-53 + ,CD-73 + ,CD-90 + ,CD-105 + , CD106 + ,CD146 + , CD166 + ,CD34-, CD45-,CD31-, CD117-, CD14-

PDL like, cementum like, alveolar bone

Osteo, adipo, chondro, myo, neuro, cemento, odonto, HLCs

.[12]

OPSC

Nestin, NG2

Hox-11, Nanog

CD-13, CD-29, CD-44, CD-71, CD-73, CD-146, CD34-, CD45-, CD105-,CD166-, D117-, CD90-

Bone

Osteogenic, neurogenic chondrogenic

[20]

Abbreviations: adipo, adipocyte; cardiomyo, cardiomyocyte; CD, cluster of differentiation; cemento, cementoblast; chondro, chondrocyte; FOXD3, forkhead box D3; DFSC, dental follicle stem cell; GMSC, gingival mesenchymal stem cell; Klf4, Kruppel-like factor 4; HLC, hepatocyte like cells, myo, myoblast; HNK-1, human natural killer 1; Hox, homeobox; Nanog, nanog homeobox; NES, neuro epithelial stem cell protein; neuro, neuronal cell; NG2, neuron glial antigen 2; Nestin, neuroepithelial stem cell protein; odonto, odontoblast; OPSC, oral periosteal stem cell; osteo, osteoblast; Oct 4, octamer 4, PAX3, paired box 3; PDLSC, periodontal ligament stem cell; PDL, periodontal ligament; P75NTR, p75-neurotrophin receptor; Snail-1, Slug- Zinc finger protein; Sox-SRY-related HMG-box genes; SSEA4, stage specific embryonic antigen; Twist 1, twist related protein 1.


+ - positive expression, - negative expression; elevated expression, decreased expression.


The PDLSCs, for instance, are found to be more of adult mesenchymal-like as they express the entire range of MSC markers and less of neural crest markers apart from sharing similar phenotype to pericytes as seen by positive expression of CD146, neural/glial antigen-2, and CD140B.[21]

The majority of putative stem cell markers have been found to be expressed by GMSCs, and it has been observed that GMSCs express high levels of the embryonic stem cell markers Oct 4, Nanog, and SSEA3, which are essential for maintaining progenitor status, when cultured in three-dimensional (3D) scaffolds primed with ascorbic acid.[22] The significant expression of pluripotent markers in GMSCs indicates that gingival stem cells have the propensity for regeneration and incorporate stem cells that constitute pluripotent properties. It is discovered that the DFSCs are more embryonic in origin and express Oct 4, but only infrequently Nanog and CD90 that demonstrate the increased heterogeneity and neural crest genesis of DFSCs.[23]

The periosteum offers mechanical support while also acting as a major source of progenitor cells and growth factors for bone regeneration.[24] Further, OPSCs showed high expression of CD-73, CD-90, CD-105, and CD-29, whereas hematological markers CD-45 and CD-34 were not expressed.[25] OPSCs have a low level of CD-117 surface marker positivity, which is a stem antigen expressed on MSCs that are not yet committed to bone phenotype.[26] OPSCs are distinguished from other types of stem cells by the selective expression of cathepsin K in the periosteum as early as embryonic day 14.[27]

Overall, the stem cells of periodontal origin exhibit a wide spectrum of characteristics that can be harnessed for optimal regeneration based on a case-to-case scenario. While the GMSCs exhibit a wide range of embryonic markers, selecting a PDLSC or an OPSC for extensive periodontal regeneration would be more preferred as they have unique properties such as expression of greater osteogenic and chondrogenic differentiation markers that can achieve cementum and periodontal ligament like structures when transplanted in animal models, thereby making them a superior choice when compared to the application of GMSCs.[28] Further, the data for PDLSCs and OPSCs to form bone suggest that they can be adequately osteogenic only when they are used in a combination with suitable grafting materials until which their regenerative potentials cannot be tapped appropriately. On the other hand, the DFSCs have been shown to provide a suitable microenvironment for enhanced regeneration of PDLSCs in vivo, thereby highlighting the adjunctive role of DFSCs by acting as a scaffold in facilitating the differentiation of other types of stem cells.[29]


#
#

Inflammatory Environment and Periodontal Stem Cells—The Bidirectional Link

The behavior of stems cells in an inflammatory environment differs markedly from the healthy state as inflammation affects the stem cells and in turn the stem cells exert immune-modulatory properties in an inflammatory microenvironment.[19] [30] Understanding this bidirectional link of how periodontal inflammation can influence stem cells and how they interact in an exacerbated immune inflammatory environment can help devise future regenerative strategies ([Fig. 2]).

Zoom Image
Fig. 2 Immunomodulation and regeneration potential of periodontal stem cells. (A) Inflammation and stem cells “The Bidirectional Effect” affecting their differentiation. (B) Immunomodulatory dentistry—Regeneration of periodontal tissues like bone, cementum, and periodontal ligament. (C) Immunomodulatory medicine—regeneration into various tissues of the body.

Effect of Periodontal inflammation on Stem Cells

In periodontal disease, gram-negative bacteria predominantly Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia are key microbial regulators of the disease, causing increased expression of inflammatory mediators and adhesion molecules triggering an inflammatory cascade there by altering the immune phenotype of the stem cells in the periodontally destructed sites. This inflammatory response acts as a regulator of tissue stemness either by directly affecting periodontal tissue stem cells or by shifting differentiated cells toward a stem cell like characteristic. The balance in this inflammatory response and mediated stemness is a critical driver of either maintaining tissue integrity or promoting aberrant homeostasis and disease. PDSLCs have been studied to have scope to regenerate the supporting structures as long-term stimulation of PDLSCs by P. gingivalis lipopolysaccharide (LPS) resulted in increase in cellular cytokine release compared to GMSCs. It is also suggested that LPS restrains the osteoblast differentiation by impairment of alkaline phosphatase activity and mineral deposition in DFSCs and PDLSCs.[31] Activation of TLR2 also caused increased proliferation of OPSCs.[32] On the contrary, studies also reported that P. gingivalis LPS treatment did not alter the cell viability of both OPSCs and DFSCs. This data suggest that the activation of inflammatory cascade has a role in the stimulation of various periodontal stem cells; however, the availability of contradictory evidences questions the phenomenon of this effect thereby warranting the need of future research.


#

Immunomodulatory Effect of Periodontal Stem Cells on Inflammatory Environment

The effect of periodontal inflammation could sensitize the stem cells; however, recent data suggest that these stem cells also exert an immunomodulatory effect that explains the opposing link between SCs of periodontal origin and local inflammation. Immunomodulation is characterized by an induction, amplification, attenuation, or prevention of the functioning of the immune system by the activity of an immunomodulator such as the periodontal stem cells.

PDLSCs induce the secretion of tumor growth factor-β, indoleamine-2,3 di-oxygenase-1 and hepatocyte growth factor that have immunomodulatory effect on periodontal regeneration.[33] PDLSCs also alter the innate immune response by elevating the proliferation and diminishing the apoptotic potential of neutrophils. In addition to the inhibition of T cell proliferation by the PDLSCs, the anti-inflammatory M2 macrophage phenotype polarization is also enhanced as the PDLSCs stimulate CD-136, interleukin-10 (IL-10) and arginase 1.[34]

Similarly, GMSCs facilitate macrophage M2 polarization and also inhibit the M1 macrophages by producing prostaglandin E2 (PGE2), IL-6, and IL-10. Furthermore, they also reduce the maturation of dendritic cells that further suppresses its capability to present the antigens in a PGE2-related phenomenon, thereby dampening the inflammatory cascade.[35] The DFSCs in a similar fashion suppress bone resorption by diminishing the phagocytic activities and neutrophil extracellular trap formation and also cause M2 macrophage polarisation.[36] DFSCs also elevate the expression of anti-inflammatory cytokines such as IL-10 and suppress the concentration of the proinflammatory cytokines, thereby preventing bacterial internalisation.[37] A recent study by He et al proved that OPSCs effectively inhibited M1 polarisation.[38] The overall picture suggests that periodontal stem cells attenuate inflammation by various mechanisms as motioned above; however, the immunomodulatory capacities of these cells, which are essential participants in the modulation of immune responses to accomplish regeneration in periodontitis models, have not yet been fully explained.


#
#

Boundless Regenerative Potential of Periodontal Stem Cells

The predictability of current treatment protocols to limit the spread of periodontal disease and facilitate regeneration is questionable. However, the principles of tissue engineering can be adapted to expedite regeneration of oral and extraoral tissues tat diversifies the application of these intimately related stem cells—the periodontal stem cells ([Table 2]).[39]

Table 2

Periodontal stem cells in regenerative periodontics and regenerative medicine

Stem cell

Growth factors

Carrier/scaffold used

Cell numbers achieved In vitro in Vivo

Model used

Regenerative outcome

Reference

PDLSCs

FGF2

RhFGF-2

Chitosan conjugated Nano HA coating

5× 104

1× 107

Mice-calvarial defect

 • Osteogenic potential of PDLCs are enhanced

 • Superior hard tissue regeneration

 • Increased mineralization by Notch signaling

[40]

TGF-β3

RGD Modified alginate microspheres

1× 106

alginate solution

1× 106

Mice

Subcutaneous

 • Enhanced tendon regeneration capacity

 • Higher chondrogenic and adipogenic differentiation

[41]

rAd- BMP-2

Hydroxyapatite and bone grafts

2× 106 cells/mL

2× 106 cells/mL

Mice and canine

 • BMP-2 enhances new bone formation and promotes osteogenesis

[42]

BMP-2

BMP-9

1% collagen hydrogel

2× 106

2× 106

Canine

 • Higher osteogenic differentiation

[43]

IGF

Absorbable gelatine sponge

Gelfoam

8× 103 cells / cm2

1× 106

Mice

 • Promotes osteogenic differentiation via osteogenesis of PDL progenitor cells

[42]

1% PRP

PDLSC sheets

8× 103 cells / cm2

1× 106

Mice

 • Increases extra-cellular matrix

[44]

FGF2

Amnion

3× 105 cells

3× 105 cells

Human

 • Increased osteo, adipo differentiation

[45]

GMSCs

TGF-β3

Alginate microspheres

1× 106

alginate solution

1× 106

Mice

Subcutaneous

 • Enhanced regeneration capacity

 • Greater chondrogenic and adipogenic differentiation

[46]

BMP-2

Collagen Scaffold

2mm × 3mm

2× 106

Rats

 • Higher osteogenic differentiation

[47]

TNF-α

Exosomes

200 µg

1× 106

Mice

 • Higher chondrogenic and osteogenic differentiation

[48]

BMP-9

Hyaluronic acid synthetic ECM

250µl

5× 106

Dog

Mini

Pig

 • Enhanced adipogenic and chondrogenic lineage

[19]

BMP-2

Collagen membrane covering a scaffold with β-TCP

2 × 105

8× 106 cells / cm3

Human

 • Periodontal defects, enhanced osteogenic, adipogenic differentiation

[49]

TGF-β

Alginate based adhesive and cross-linked hydrogel

4× 106

4× 106

Rat

 • Higher osteogenic potential in repairing peri implantitis model

[50]

Hydorgel scaffold(PuraMatrix)

1 × 106

1 × 106

Rat

 • Maxillary alveolar defects-higher osteogenic bone formation

[51]

FGF2

(PLA) 3D bioengineered scaffold

Enriched with GMSCs

2× 106 cells

2× 106 cells

Rat

 • Calvarial defects enhanced regeneration into osteocytes and adipocytes

[52]

IGF-1

BMP-4

Axo guard Nerve Conduits

0.5× 106

0.5 × 106

Rat

 • Facial nerve - Enhanced neuronal and glial differentiation

[53]

DFSCs

BMP-2

BMP-9

HA powder

2 × 105

2× 105

Mice (Subcutaneous)

 • Fibrous tissue formation and cementum matrix

[54]

BMP-9

HA coated dental implant

5× 104

1× 107

Murine

 • Osteogenic differentiation and periodontal ligament like tissues

[55]

BMP-2

HA/ Collagen gel

2× 106

2× 106

Mice

 • Higher differentiation into cementum like tissues – Acellular cementum

[56]

IGF

Collagen nano HA/ phosphoserine biocomposite cryogel

1 × 106

1 × 106

Mice

 • Enhanced osteogenic differentiation to bone like tissues

[57]

FGF-2

Treated dentin matrix (TDM)

5× 104

1× 107

Canine

 • Enhanced osteogenic, cementogenic, periodontal ligament tissue formation in bony defects

[58]

FGF-2

TDM

5× 104

1× 107

Mice

 • Increased formation of periodontal tissues like cementum and alveolar bone

[59]

TGF- β1

Ceramic bovine bone

2× 106

2× 106

Mice

 • Enhanced cementogenic, osteogenic and fibroblastic potential (forms cementum-PDL complex)

[60]

BMP-2

Extra cellular matrix (ECM)

250µl

5 × 106

Rat

 • Enhanced bone regeneration and higher osteogenic differentiation

[61]

OPSCs

TGF-β

BMP-2

Periosteal cell sheets

1× 106

2× 106

Human bone defects

 • Increased bone formation by osteogenic differentiation

[62]

BMP-2

OPSC cell sheets

1× 106

2× 106

Human

 • Sinus elevation procedures- enhanced bone formation

[63]

FGF-2

VEGF

Cell sheets OPSC

1× 106

2× 106

Mice

 • Enhanced osteogenic and chondrogenic differentiation

[64]

BMP-2

HA powder

2 × 105

2× 105

Rat

 • Higher osteogenic differentiation

[65]

BMP-9

HA bone graft

2× 106

2× 106

Mice

 • Enhanced bone formation

[66]

Abbreviations: BMP, bone morphogenetic protein; DFSCs, dental follicle stem cells; FGF, fibroblast growth factor; GMSCs, gingival mesenchymal stem cells; HA, hydroxy apatite; IGF, insulin-like growth factor; OPSCs, oral periosteal stem cells; PDLSCs, periodontal ligament stem cells; PDL, periodontal ligament; PLA, polylactic acid; PRP, platelet-rich plasma; RGD, arginylglycylaspartic acid; RhFGF, recombinant human fibroblast growth factor; rAdBMP-2, recombinant adenovirus (rAd) encodingBMP-2; TCP, tricalcium phosphate; TGF, transforming growth factor; TNF, tumor necrosis factor; VEGF, vascular endothelial growth factor.


The potential of PDLSCs in tissue regeneration as explained by Seo et al suggested that PDLSCs could generate tissue similar to the cementum and periodontal ligament.[8] Coextensive research on PDLSC sheets exhibited the property of giving rise to structures similar to the periodontal tissues in a tricalcium phosphate matrix. The amalgamation of PDLSCs with chitosan-based scaffolds aided in bone regeneration in calvarial defect models.[67] Human DFSC sheets transplant was able to achieve optimal periodontal regeneration, including new periodontal ligament attachments, new alveolar bone development, and even a periodontal ligament–cementum complex structure. Further, DFSC sheets were more conducive to periodontal regeneration than PDLSC sheets, as observed in canine model, which might be attributable to DFSC's greater ability to adapt to the chronic inflammatory milieu of periodontitis.

Wang et al first demonstrated that transplanted GMSCs formed new bone in mandibular wounds and calvarial defects of nude mice and rats, which suggested that GMSCs can repair bone.[68] GMSCs exhibited a moderate osteogenic capability, similar to PDLSCs; however, GMSCs were stronger at forming mineralized nodules and differentiated into osteogenic, chondrogenic, and adipogenic lineages.[69] While some studies suggest that inflamed GMSCs have a reduced capability for osteogenic and adipogenic differentiation than healthy subjects, epigenetic variables linked to chronic periodontitis could impact the cell line's different orientations. The “real mesenchymal stem cells”, the OPSCs have the characteristics to differentiate into osteoblasts and chondroblasts.[25] Cacceralli et al also suggested OPSCs to be a valuable and precious alternative compared to other mesenchymal stem cells from bone marrow for tissue engineering applications in oral cavity. The extraoral applications of OPSCs need to be studied further; however, the other periodontal stem cells have widely studied in regenerative medicine for their varied differentiation capabilities.

The therapeutic potential of PDLSCs in medicine has been used for differentiation of corneal stromal keratinocytes as both PDLSCs and corneal cells are derived from the neural crest. In the personalized treatment of multiple sclerosis (MS), a comparison of PDLSCs obtained from systemically healthy patients and MS patients showed identical proliferative and differentiative potential of PDLSCs, thereby validating the use of PDLSCs in such auto-immune conditions.[70] Lee and Park demonstrated the transdifferentiation of PDLSCs into pancreatic islet cells thereby providing an alternative treatment strategy for diabetes.[71] Periodontal mesenchymal stem cells can also differentiate into cardiac muscles, skeletal muscles, endothelial and neuronal cells suggesting their therapeutic application in medical regenerative procedures.

DFSCs have a potential for neuronal differentiation as they differentiate into mature neurons and oligodendrocytes but not astrocytes. The application of DFSCs in myasthenia gravis was pioneered by Ulusoy et al, whereas the therapeutic effect of DFSCs in asthma was researched by coculturing DFSCs with the blood mononucleocytes of asthmatic patients in vitro.[72]

GMSCs, on the other hand, have been shown to demonstrate antiageing potentials and this property can be harnessed to develop cell free treatment strategies for ageing-related and vascular disorders.[73] GMSCs have also been known to differentiate into neuronal and glial cells and this property has been harnessed in facial nerve regeneration and the management of spinal cord injuries.[74] Ansari et al reported that GMSCs encapsulated in alginate underwent osteogenic differentiation and also have chondrogenic potential without the need of additional growth factors.[75] It has been suggested that GMSCs and OPSCs exhibit similar degree of bone regeneration in defects created in rabbit models suggesting that these stem cells maybe an useful alternative in regenerative strategies.[76]


#

Stem Cell Derivatives in Regeneration

E1. 3D Bioprinted Scaffolds—Self-Scaffolded Models

In scaffold-free tissue engineering, cells produce and arrange their own endogenous ECM to create a 3D structure. Scaffold-free tissue engineering, in contrast to conventional tissue engineering techniques, forgoes the use of an external scaffold material to create a 3D tissue. In 2007, a 3D bioengineered tooth—“organoid”—was made by combining dental epithelium and mesenchyme to form a complete tooth germ and it was proposed that combining in with biomaterials such as collagen can enhance the possibility of forming a bioengineered tooth in animal models. 3D spheroids can be used in a variety of culture methods to optimize the property and function of MSCs as they allow close cell–cell and cell–matrix interactions that closely resemble the microenvironment.[77] The various methods of culturing in 3D are using patterned microwells, floating culture for neurosphere formation, chitosan, ultra-low culture dishes, and poly-L-ornithine. These can be enhanced by adding growth factors like fibroblast like growth factors, lovastatins, spheroids, and mesenspheres.

PDLSCs have been successfully cultured to form 3D structures mimicking cementum and periodontal ligament using this technique. After in vitro cultivation, the periodontal tissue organization was evident, and it was preserved in vivo as well as after subcutaneous implantation in mice. These results show that PDLCs can self-assemble into an ordered cementum-PDL-like complex through scaffold-free tissue engineering.[78]

Self-Assembly of a 3D spheroid culture of GMSCs can enhance the differentiation and neural stem cell properties as shown by Hsu et al, where the GMSCs were found to spontaneously aggregate into 3D spheroids with enhanced stemness and increases trilineage differentiation.[79] The GMSCs cultured in patterned microwells aggregate into 3D spheroids and have higher osteogenic potential that is augmented by addition of growth factors, while floating culture technique allows for aggregation into neurospheres and elevated neural crest markers and neuronal differentiation. The possibility of reprogramming GMSCs into neural crest like cells to differentiate into nerve cells is also possible by 3D culturing.[53]

DFSCs have been seeded on 3D porous scaffolds laden with collagen-nanohydroxyapatite/phosphoserine biocompatible cryogel with osteogenic factors in the culture medium and the resultant 3D spheroids showed dynamic growth and osteogenic differentiation when implanted in mice models.[57]


#

E2. Use of Dental Stem Cell-Derived Exosomes in Regenerative Medicine Inverting the Disease Paradigm:

Extracellular vesicles (EVs) including ectosomes and exosomes are essential for intracellular communication as they can carry bioactive molecules such as lipids, nucleic acids, proteins, and metabolites. EVs are released as membrane bound agents from all types of cells and even found to be released from periodontal stem cells and used for treating diseases. EVs can be categorized as apoptotic vesicles, microvesicles, and exosomes. EVs are successful in treating a variety of disorders by encapsulating and conveying essential bioactive components (e.g., proteins and nucleic acids) to affect the phenotype of target cells.

PDLSCs produce exosomes that harbor the potential of repair and regeneration, which can induce angiogenesis, alleviate neurological diseases, and reduce the inflammatory microenvironment.[80] In periodontitis, they can be used to induce osteogenesis and enhance bone regeneration as seen by Pizzicannella et al, in lesions of the rat skull, where addition of PDLSC-derived exosomes to a 3D collagen membrane and polyethyleneimine scaffold, showed bone regeneration.[81] Further, it has been observed that human PDLSC-derived exosomes promote osteogenesis by the expression of their exosomal miRNAs in vitro.[82] Similarly, GMSC-EVs in periodontitis models in rats show periodontal regeneration by delivery of mIR-120b to inhibit osteoclastogenic activity of PDL cells by targeting the Wnt5a-mediated RANKL pathway.[48]

DFSCs-sEVs were found to greatly increase PDLSC migration, proliferation, and osteogenic differentiation and regeneration of periodontal tissue by stimulation of the p38-MAPK signaling pathway. Small EVs from DFSCs provide biochemical cues for periodontal tissue regeneration.[83] There is potential to use OPSC-derived EVs in future for bone regeneration as they are more committed to an osteogenic lineage. However, to achieve an optimal periodontal regeneration of the intricate structures in the periodontium, more research is still required to identify ideal and standardized sources of EVs, their effective concentration, frequency of treatment, and suitable scaffolds or delivery routes. Better insight into the therapeutic potential of periodontal stem cells derived-EVs would provide more reasonable options for the future treatment of periodontal diseases.


#
#

Clinical Application of Stem Cells in Human Periodontal Regeneration

The use of stem cells cultured in scaffolds has shown promising results in vitro and animal model studies. The subsequent application of these stem cells needs to be explored whether it can bring about regeneration as previous clinical trials have been performed in this regard to achieve periodontal regeneration using autologous transplanted stem cells from the periodontal complex. PDLSC sheet transplants in humans have shown no adverse effects and have generated a reduction in probing depth; however, this was not significant. Further, a clinical trial with autologous PDLSC transplants in periodontitis patients had shown improvement in periodontal parameters as seen by reduction in pocket depths and bone regeneration. Transplants of PDLSC sheets mixed with granules of β-tricalcium phosphate in bone defects have also shown no adverse effects till 6 months.[84] PDLSCs implanted in animal models show superior periodontal regeneration in the form of greater cementum, bone, and PDL formation. A recently concluded clinical trial on animal model showed that when the PDLSC was combined with collagen membrane on fenestration defects, it showed a greater cementum formation but no difference in bone formation when applied without the membrane.[85] Similarly, in furcation defects when PDLSCs were combined with hyaluronic acid sheets, it resulted in greater cementum, bone, and PDL formation than controls.[86] Further, PDLSC sheets combined with β-tricalcium phosphate in infrabony defects resulted in nearly complete regeneration of periodontal tissues.[87] Over all, it can be deciphered that the application of PDLSCs in animal models is predictable for cementum formation; however, the results are conflicting when it comes to bone regeneration.

Use of GMSC in future clinical trial seems to be a promising approach as systemically administered GMSCs have the ability to home to the injury site and differentiate into osteoblasts, cementoblasts, and periodontal ligament fibroblasts as tested in animal models. In class III furcation defect animal models, GMSC sheets significantly have enhanced the regeneration of periodontal tissues.[51] Combination of the GMSC with HA gel has also showed a significant regeneration in porcine model by exhibiting formation of newly formed bone and PDL fibres.[88] GMSC human clinical trials have also been employed in treating periodontal defects while embedding these cells in collagen scaffolds mixed with β-tricalcium phosphate, thereby reducing probing depth, attachment gains, and alveolar bone gain as seen in 6 months follow-up.[49]

DFSC sheets implanted in animal models have shown to regenerate whole periodontal tissue as observed by formation of complex–periodontal ligament like structure within a month. Implanting into periodontal irregularities in vivo, DFSCs show a better capacity for cementum and periodontal attachment healing than PDLSCs due to higher involvement of extracellular matrix.[29] [89] The DFSCs work by providing an ideal microenvironment for the growth of PDLSCs and act as a scaffold.

Human OPSCs, on the other hand, have shown promising results in vivo when transplanted in human intrabony periodontal defects and in sinus elevation procedures for forming bone.[63] There are enhanced osteogenic properties of OPSCs when transplanted with growth factors and enriched with collagen scaffolds.[62]


#

Conclusion

Multiple stem cell populations including PDLSCs, GMSCs, DFSCs, and OPSCs coexist in close proximity and still stay in function in spite of continual remodeling and inflammation in the periodontal complex. Periodontal stem cells have a strong interaction with the inflammatory milieu, as well as the ability to modulate the immune system, making them lucrative candidates for cell treatment in periodontitis and inflammatory disorders. The periodontal stem cells are one-of-a-kind, with a variety of morphologies and multipotency, both in vitro and in vivo. Each stem cell population's differentiation capacity is diverse, and it can repair bone, neurons, and tendons in addition to mesenchymal tissues in the oral cavity. The PDLSC are thought to be harboring diverse regenerative potential within the oral cavity; however, they are more differentiated cells. The DFSCs in contrast exhibit greater propensity for extraoral tissue differentiation as shown by higher expression of embryonic markers like Oct4 and Nanog. GMSCs are touted to be the stem cells with greater accessibility and higher differentiation capacity as seen by various clinical models and 3D bioprinting studies. The use of GMSCs for nerve regeneration is promising in future. The OPSCs, on the other hand, need to be studied further to understand their behavior both in vitro and in clinical human models. Although it would not be appropriate to state the superiority of one stem cell type over another, it is plausible that therapeutic application of these stem cells to regenerate hard and soft tissues and alleviate degenerative diseases may become a reality in the future. To this regard, additional prospective and long-term trials are needed to determine the true characteristics of each population and how they might be used for exogenous MSC grafting, 3D bioprinting, specific exosomal derived vesicles, and cell homing in periodontal tissue engineering.


#
#

Conflict of Interest

None.

  • References

  • 1 Cekici A, Kantarci A, Hasturk H, Van Dyke TE. Inflammatory and immune pathways in the pathogenesis of periodontal disease. Periodontol 2000 2014; 64 (01) 57-80
  • 2 Van der Weijden GAF, Dekkers GJ, Slot DE. Success of non-surgical periodontal therapy in adult periodontitis patients: a retrospective analysis. Int J Dent Hyg 2019; 17 (04) 309-317
  • 3 Liang Y, Luan X, Liu X. Recent advances in periodontal regeneration: a biomaterial perspective. Bioact Mater 2020; 5 (02) 297-308
  • 4 Wang M, Xie J, Wang C, Zhong D, Xie L, Fang H. Immunomodulatory properties of stem cells in periodontitis: current status and future prospective. Stem Cells Int 2020; 2020: 9836518 DOI: 10.1155/2020/9836518.
  • 5 Citterio F, Gualini G, Fierravanti L, Aimetti M. Stem cells and periodontal regeneration: present and future. Plast Aesthet Res 2020; 7: 41 DOI: 10.20517/2347-9264.2020.29.
  • 6 Isern J, García-García A, Martín AM. et al. The neural crest is a source of mesenchymal stem cells with specialized hematopoietic stem cell niche function. eLife 2014; 3: e03696 DOI: 10.7554/eLife.03696.
  • 7 Wang L, Shen H, Zheng W. et al. Characterization of stem cells from alveolar periodontal ligament. Tissue Eng Part A 2011; 17 (7-8): 1015-1026
  • 8 Seo BM, Miura M, Gronthos S. et al. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004; 364 (9429): 149-155
  • 9 Nugraha AP, Narmada IB, Ernawati DS. et al. Osteogenic potential of gingival stromal progenitor cells cultured in platelet rich fibrin is predicted by core-binding factor subunit-α1/Sox9 expression ratio ( in vitro). F1000 Res 2018; 7: 1134 DOI: 10.12688/f1000research.15423.1.
  • 10 Al-Qadhi G, Al-Rai S, Hafed L. The therapeutic potential of inflamed gingiva-derived mesenchymal stem cells in preclinical studies: a scoping review of a unique biomedical waste. Stem Cells Int 2021; 2021: 6619170 DOI: 10.1155/2021/6619170.
  • 11 Morsczeck C, Götz W, Schierholz J. et al. Isolation of precursor cells (PCs) from human dental follicle of wisdom teeth. Matrix Biol 2005; 24 (02) 155-165
  • 12 Zhang J, Ding H, Liu X, Sheng Y, Liu X, Jiang C. Dental follicle stem cells: tissue engineering and immunomodulation. Stem Cells Dev 2019; 28 (15) 986-994
  • 13 Saimbi CS, Gautam A, Khan MA. , Nandlal. Periosteum as a barrier membrane in the treatment of intrabony defect: a new technique. J Indian Soc Periodontol 2014; 18 (03) 331-335
  • 14 Zhou LL, Liu W, Wu YM, Sun WL, Dörfer CE, Fawzy El-Sayed KM. Oral mesenchymal stem/progenitor cells: the immunomodulatory masters. Stem Cells Int 2020; 2020: 1327405 DOI: 10.1155/2020/1327405.
  • 15 Fournier BPJ, Larjava H, Häkkinen L. Gingiva as a source of stem cells with therapeutic potential. Stem Cells Dev 2013; 22 (24) 3157-3177
  • 16 Stefańska K, Mehr K, Wieczorkiewicz M. et al. Stemness potency of human gingival cells-application in anticancer therapies and clinical trials. Cells 2020; 9 (08) E1916 DOI: 10.3390/cells9081916.
  • 17 Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell 2012; 10 (06) 709-716
  • 18 Huang GTJ, Gronthos S, Shi S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. J Dent Res 2009; 88 (09) 792-806
  • 19 Fawzy El-Sayed KM, Dörfer CE. Gingival mesenchymal stem/progenitor cells: a unique tissue engineering gem. Stem Cells Int 2016; 2016: 7154327 DOI: 10.1155/2016/7154327.
  • 20 Zhang N, Hu L, Cao Z, Liu X, Pan J. Periosteal skeletal stem cells and their response to bone injury. Front Cell Dev Biol 2022; 10: 812094 DOI: 10.3389/fcell.2022.812094.
  • 21 Kaku M, Komatsu Y, Mochida Y, Yamauchi M, Mishina Y, Ko C-C. Identification and characterization of neural crest-derived cells in adult periodontal ligament of mice. Arch Oral Biol 2012; 57 (12) 1668-1675
  • 22 Zhang Q, Nguyen AL, Shi S. et al. Three-dimensional spheroid culture of human gingiva-derived mesenchymal stem cells enhances mitigation of chemotherapy-induced oral mucositis. Stem Cells Dev 2012; 21 (06) 937-947
  • 23 Lima RL, Holanda-Afonso RC, Moura-Neto V, Bolognese AM, DosSantos MF, Souza MM. Human dental follicle cells express embryonic, mesenchymal and neural stem cells markers. Arch Oral Biol 2017; 73: 121-128
  • 24 Ichikawa Y, Watahiki J, Nampo T. et al. Differences in the developmental origins of the periosteum may influence bone healing. J Periodontal Res 2015; 50 (04) 468-478
  • 25 Ceccarelli G, Graziano A, Benedetti L. et al. Osteogenic potential of human oral-periosteal cells (PCs) isolated from different oral origin: an in vitro study. J Cell Physiol 2016; 231 (03) 607-612
  • 26 Suphanantachat S, Iwata T, Ishihara J, Yamato M, Okano T, Izumi Y. A role for c-Kit in the maintenance of undifferentiated human mesenchymal stromal cells. Biomaterials 2014; 35 (11) 3618-3626
  • 27 Debnath S, Yallowitz AR, McCormick J. et al. Discovery of a periosteal stem cell mediating intramembranous bone formation. Nature 2018; 562 (7725): 133-139
  • 28 Lee H, Park J-B. Dimethyl sulfoxide leads to decreased osteogenic differentiation of stem cells derived from gingiva via Runx2 and collagen i expression. Eur J Dent 2019; 13 (02) 131-136
  • 29 Guo S, Guo W, Ding Y. et al. Comparative study of human dental follicle cell sheets and periodontal ligament cell sheets for periodontal tissue regeneration. Cell Transplant 2013; 22 (06) 1061-1073
  • 30 Nugraha AP, Ramadhani NF, Riawan W. et al. Gingival mesenchymal stem cells metabolite decreasing TRAP, NFATc1, and sclerostin expression in LPS-associated inflammatory osteolysis in vivo. Eur J Dent 2023; 17 (03) 881-888
  • 31 Morsczeck CO, Drees J, Gosau M. Lipopolysaccharide from Escherichia coli but not from Porphyromonas gingivalis induce pro-inflammatory cytokines and alkaline phosphatase in dental follicle cells. Arch Oral Biol 2012; 57 (12) 1595-1601
  • 32 Pevsner-Fischer M, Morad V, Cohen-Sfady M. et al. Toll-like receptors and their ligands control mesenchymal stem cell functions. Blood 2007; 109 (04) 1422-1432
  • 33 Andrukhov O, Hong JSA, Andrukhova O, Blufstein A, Moritz A, Rausch-Fan X. Response of human periodontal ligament stem cells to IFN-γ and TLR-agonists. Sci Rep 2017; 7 (01) 12856 DOI: 10.1038/s41598-017-12480-7.
  • 34 Shin C, Kim M, Han JA. et al. Human periodontal ligament stem cells suppress T-cell proliferation via down-regulation of non-classical major histocompatibility complex-like glycoprotein CD1b on dendritic cells. J Periodontal Res 2017; 52 (01) 135-146
  • 35 Su WR, Zhang QZ, Shi SH, Nguyen AL, Le AD. Human gingiva-derived mesenchymal stromal cells attenuate contact hypersensitivity via prostaglandin E2-dependent mechanisms. Stem Cells 2011; 29 (11) 1849-1860
  • 36 Chen X, Yang B, Tian J. et al. Dental follicle stem cells ameliorate lipopolysaccharide-induced inflammation by secreting TGF-β3 and TSP-1 to elicit macrophage M2 polarization. Cell Physiol Biochem 2018; 51 (05) 2290-2308
  • 37 Chatzivasileiou K, Lux CA, Steinhoff G, Lang H. Dental follicle progenitor cells responses to Porphyromonas gingivalis LPS. J Cell Mol Med 2013; 17 (06) 766-773
  • 38 He F, Umrath F, von Ohle C, Reinert S, Alexander D. Analysis of the influence of jaw periosteal cells on macrophages phenotype using an innovative horizontal coculture system. Biomedicines 2021; 9 (12) 1753 DOI: 10.3390/biomedicines9121753.
  • 39 Queiroz A, Albuquerque-Souza E, Gasparoni LM. et al. Therapeutic potential of periodontal ligament stem cells. World J Stem Cells 2021; 13 (06) 605-618
  • 40 Ge S, Zhao N, Wang L, Liu H, Yang P. Effects of hydroxyapatite nanostructure on channel surface of porcine acellular dermal matrix scaffold on cell viability and osteogenic differentiation of human periodontal ligament stem cells. Int J Nanomedicine 2013; 8: 1887-1895
  • 41 Moshaverinia A, Xu X, Chen C. et al. Application of stem cells derived from the periodontal ligament or gingival tissue sources for tendon tissue regeneration. Biomaterials 2014; 35 (09) 2642-2650
  • 42 Yu B-H, Zhou Q, Wang Z-L. Periodontal ligament versus bone marrow mesenchymal stem cells in combination with Bio-Oss scaffolds for ectopic and in situ bone formation: a comparative study in the rat. J Biomater Appl 2014; 29 (02) 243-253
  • 43 Park S-Y, Kim K-H, Gwak E-H. et al. Ex vivo bone morphogenetic protein 2 gene delivery using periodontal ligament stem cells for enhanced re-osseointegration in the regenerative treatment of peri-implantitis. J Biomed Mater Res A 2015; 103 (01) 38-47
  • 44 Xu Q, Li B, Yuan L. et al. Combination of platelet-rich plasma within periodontal ligament stem cell sheets enhances cell differentiation and matrix production. J Tissue Eng Regen Med 2017; 11 (03) 627-636
  • 45 Iwasaki K, Komaki M, Yokoyama N. et al. Periodontal regeneration using periodontal ligament stem cell-transferred amnion. Tissue Eng Part A 2014; 20 (3-4): 693-704
  • 46 Moshaverinia A, Chen C, Akiyama K. et al. Encapsulated dental-derived mesenchymal stem cells in an injectable and biodegradable scaffold for applications in bone tissue engineering. J Biomed Mater Res A 2013; 101 (11) 3285-3294
  • 47 Qiu J, Wang X, Zhou H. et al. Enhancement of periodontal tissue regeneration by conditioned media from gingiva-derived or periodontal ligament-derived mesenchymal stem cells: a comparative study in rats. Stem Cell Res Ther 2020; 11 (01) 42 DOI: 10.1186/s13287-019-1546-9.
  • 48 Nakao Y, Fukuda T, Zhang Q. et al. Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater 2021; 122: 306-324
  • 49 Abdal-Wahab M, Abdel Ghaffar KA, Ezzatt OM, Hassan AAA, El Ansary MMS, Gamal AY. Regenerative potential of cultured gingival fibroblasts in treatment of periodontal intrabony defects (randomized clinical and biochemical trial). J Periodontal Res 2020; 55 (03) 441-452
  • 50 Hasani-Sadrabadi MM, Sarrion P, Pouraghaei S. et al. An engineered cell-laden adhesive hydrogel promotes craniofacial bone tissue regeneration in rats. Sci Transl Med 2020; 12 (534) eaay6853 DOI: 10.1126/scitranslmed.aay6853.
  • 51 Kandalam U, Kawai T, Ravindran G. et al. Predifferentiated gingival stem cell-induced bone regeneration in rat alveolar bone defect model. Tissue Eng Part A 2021; 27 (5-6): 424-436
  • 52 Diomede F, Gugliandolo A, Cardelli P. et al. Three-dimensional printed PLA scaffold and human gingival stem cell-derived extracellular vesicles: a new tool for bone defect repair. Stem Cell Res Ther 2018; 9 (01) 104 DOI: 10.1186/s13287-018-0850-0.
  • 53 Zhang Q, Nguyen PD, Shi S, Burrell JC, Cullen DK, Le AD. 3D bio-printed scaffold-free nerve constructs with human gingiva-derived mesenchymal stem cells promote rat facial nerve regeneration. Sci Rep 2018; 8 (01) 6634 DOI: 10.1038/s41598-018-24888-w.
  • 54 Honda MJ, Imaizumi M, Tsuchiya S, Morsczeck C. Dental follicle stem cells and tissue engineering. J Oral Sci 2010; 52 (04) 541-552
  • 55 Oshima M, Inoue K, Nakajima K. et al. Functional tooth restoration by next-generation bio-hybrid implant as a bio-hybrid artificial organ replacement therapy. Sci Rep 2014; 4: 6044 DOI: 10.1038/srep06044.
  • 56 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 (09) 632-645
  • 57 Salgado CL, Barrias CC, Monteiro FJM. Clarifying the tooth-derived stem cells behavior in a 3D biomimetic scaffold for bone tissue engineering applications. Front Bioeng Biotechnol 2020; 8: 724 DOI: 10.3389/fbioe.2020.00724.
  • 58 Yang H, Li J, Hu Y. et al. Treated dentin matrix particles combined with dental follicle cell sheet stimulate periodontal regeneration. Dent Mater 2019; 35 (09) 1238-1253
  • 59 Tian Y, Bai D, Guo W. et al. Comparison of human dental follicle cells and human periodontal ligament cells for dentin tissue regeneration. Regen Med 2015; 10 (04) 461-479
  • 60 Guo W, Gong K, Shi H. et al. Dental follicle cells and treated dentin matrix scaffold for tissue engineering the tooth root. Biomaterials 2012; 33 (05) 1291-1302
  • 61 Tsuchiya S, Ohshima S, Yamakoshi Y, Simmer JP, Honda MJ. Osteogenic differentiation capacity of porcine dental follicle progenitor cells. Connect Tissue Res 2010; 51 (03) 197-207
  • 62 Kawase T, Okuda K, Kogami H. et al. Characterization of human cultured periosteal sheets expressing bone-forming potential: in vitro and in vivo animal studies. J Tissue Eng Regen Med 2009; 3 (03) 218-229
  • 63 Nagata M, Hoshina H, Li M. et al. A clinical study of alveolar bone tissue engineering with cultured autogenous periosteal cells: coordinated activation of bone formation and resorption. Bone 2012; 50 (05) 1123-1129
  • 64 Agata H, Asahina I, Yamazaki Y. et al. Effective bone engineering with periosteum-derived cells. J Dent Res 2007; 86 (01) 79-83
  • 65 Ueno T, Honda K, Hirata A. et al. Histological comparison of bone induced from autogenously grafted periosteum with bone induced from autogenously grafted bone marrow in the rat calvarial defect model. Acta Histochem 2008; 110 (03) 217-223
  • 66 Cicconetti A, Sacchetti B, Bartoli A. et al. Human maxillary tuberosity and jaw periosteum as sources of osteoprogenitor cells for tissue engineering. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2007; 104 (05) 618.e1-618.e12
  • 67 Ge S, Zhao N, Wang L. et al. Bone repair by periodontal ligament stem cellseeded nanohydroxyapatite-chitosan scaffold. Int J Nanomedicine 2012; 7: 5405-5414
  • 68 Wang F, Yu M, Yan X. et al. Gingiva-derived mesenchymal stem cell-mediated therapeutic approach for bone tissue regeneration. Stem Cells Dev 2011; 20 (12) 2093-2102
  • 69 Al-Qadhi G, Aboushady I, Al-Sharabi N. The gingiva from the tissue surrounding the bone to the tissue regenerating the bone: a systematic review of the osteogenic capacity of gingival mesenchymal stem cells in preclinical studies. Stem Cells Int 2021; 2021: 6698100 DOI: 10.1155/2021/6698100.
  • 70 Diomede F, Rajan TS, D'Aurora M. et al. Stemness characteristics of periodontal ligament stem cells from donors and multiple sclerosis patients: a comparative study. Stem Cells Int 2017; 2017: 1606125 DOI: 10.1155/2017/1606125.
  • 71 Lee JS, An SY, Kwon IK, Heo JS. Transdifferentiation of human periodontal ligament stem cells into pancreatic cell lineage. Cell Biochem Funct 2014; 32 (07) 605-611
  • 72 Ulusoy C, Zibandeh N, Yıldırım S. et al. Dental follicle mesenchymal stem cell administration ameliorates muscle weakness in MuSK-immunized mice. J Neuroinflammation 2015; 12 (01) 231 DOI: 10.1186/s12974-015-0451-0.
  • 73 Shi HZ, Zeng JC, Shi SH, Giannakopoulos H, Zhang QZ, Le AD. Extracellular vesicles of GMSCs alleviate aging-related cell senescence. J Dent Res 2021; 100 (03) 283-292
  • 74 Subbarayan R, Barathidasan R, Raja STK. et al. Human gingival derived neuronal cells in the optimized caffeic acid hydrogel for hemitransection spinal cord injury model. J Cell Biochem 2020; 121 (03) 2077-2088
  • 75 Ansari S, Sarrion P, Hasani-Sadrabadi MM, Aghaloo T, Wu BM, Moshaverinia A. Regulation of the fate of dental-derived mesenchymal stem cells using engineered alginate-GelMA hydrogels. J Biomed Mater Res A 2017; 105 (11) 2957-2967
  • 76 Al-Qadhi G, Soliman M, Abou-Shady I, Rashed L. Gingival mesenchymal stem cells as an alternative source to bone marrow mesenchymal stem cells in regeneration of bone defects: in vivo study. Tissue Cell 2020; 63: 101325 DOI: 10.1016/j.tice.2019.101325.
  • 77 Ryu N-E, Lee S-H, Park H. Spheroid culture system methods and applications for mesenchymal stem cells. Cells 2019; 8 (12) E1620 DOI: 10.3390/cells8121620.
  • 78 Basu A, Rothermund K, Ahmed MN, Syed-Picard FN. Self-assembly of an organized cementum-periodontal ligament-like complex using scaffold-free tissue engineering. Front Physiol 2019; 10: 422 DOI: 10.3389/fphys.2019.00422.
  • 79 Hsu SH, Huang G-S, Feng F. Isolation of the multipotent MSC subpopulation from human gingival fibroblasts by culturing on chitosan membranes. Biomaterials 2012; 33 (09) 2642-2655
  • 80 Zhang Z, Shuai Y, Zhou F. et al. PDLSCs regulate angiogenesis of periodontal ligaments via VEGF transferred by exosomes in periodontitis. Int J Med Sci 2020; 17 (05) 558-567
  • 81 Pizzicannella J, Gugliandolo A, Orsini T. et al. Engineered extracellular vesicles from human periodontal-ligament stem cells increase VEGF/VEGFR2 expression during bone regeneration. Front Physiol 2019; 10: 512 DOI: 10.3389/fphys.2019.00512.
  • 82 Liu T, Hu W, Zou X. et al. Human periodontal ligament stem cell-derived exosomes promote bone regeneration by altering microRNA profiles. Stem Cells Int 2020; 2020: 8852307 DOI: 10.1155/2020/8852307.
  • 83 Ma L, Rao N, Jiang H. et al. Small extracellular vesicles from dental follicle stem cells provide biochemical cues for periodontal tissue regeneration. Stem Cell Res Ther 2022; 13 (01) 92 DOI: 10.1186/s13287-022-02767-6.
  • 84 Iwata T, Yamato M, Washio K. et al. Periodontal regeneration with autologous periodontal ligament-derived cell sheets - a safety and efficacy study in ten patients. Regen Ther 2018; 9: 38-44
  • 85 Nakahara T, Nakamura T, Kobayashi E. et al. In situ tissue engineering of periodontal tissues by seeding with periodontal ligament-derived cells. Tissue Eng 2004; 10 (3-4): 537-544
  • 86 Akbay A, Baran C, Günhan O, Ozmeriç N, Baloş K. Periodontal regenerative potential of autogenous periodontal ligament grafts in class II furcation defects. J Periodontol 2005; 76 (04) 595-604
  • 87 Chen F-M, Gao L-N, Tian B-M. et al. Treatment of periodontal intrabony defects using autologous periodontal ligament stem cells: a randomized clinical trial. Stem Cell Res Ther 2016; 7: 33 DOI: 10.1186/s13287-016-0288-1.
  • 88 Kim D, Lee AE, Xu Q, Zhang Q, Le AD. Gingiva-derived mesenchymal stem cells: potential application in tissue engineering and regenerative medicine - a comprehensive review. Front Immunol 2021; 12: 667221 DOI: 10.3389/fimmu.2021.667221.
  • 89 Ponnaiyan D, Jegadeesan V. Comparison of phenotype and differentiation marker gene expression profiles in human dental pulp and bone marrow mesenchymal stem cells. Eur J Dent 2014; 8 (03) 307-313

Address for correspondence

Roshan R. Rughwani, MDS
No. 5/3, 2nd Link Street, CIT Colony, Mylapore, Chennai – 600 004, Tamil Nadu
India   

Publication History

Article published online:
31 December 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

  • References

  • 1 Cekici A, Kantarci A, Hasturk H, Van Dyke TE. Inflammatory and immune pathways in the pathogenesis of periodontal disease. Periodontol 2000 2014; 64 (01) 57-80
  • 2 Van der Weijden GAF, Dekkers GJ, Slot DE. Success of non-surgical periodontal therapy in adult periodontitis patients: a retrospective analysis. Int J Dent Hyg 2019; 17 (04) 309-317
  • 3 Liang Y, Luan X, Liu X. Recent advances in periodontal regeneration: a biomaterial perspective. Bioact Mater 2020; 5 (02) 297-308
  • 4 Wang M, Xie J, Wang C, Zhong D, Xie L, Fang H. Immunomodulatory properties of stem cells in periodontitis: current status and future prospective. Stem Cells Int 2020; 2020: 9836518 DOI: 10.1155/2020/9836518.
  • 5 Citterio F, Gualini G, Fierravanti L, Aimetti M. Stem cells and periodontal regeneration: present and future. Plast Aesthet Res 2020; 7: 41 DOI: 10.20517/2347-9264.2020.29.
  • 6 Isern J, García-García A, Martín AM. et al. The neural crest is a source of mesenchymal stem cells with specialized hematopoietic stem cell niche function. eLife 2014; 3: e03696 DOI: 10.7554/eLife.03696.
  • 7 Wang L, Shen H, Zheng W. et al. Characterization of stem cells from alveolar periodontal ligament. Tissue Eng Part A 2011; 17 (7-8): 1015-1026
  • 8 Seo BM, Miura M, Gronthos S. et al. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004; 364 (9429): 149-155
  • 9 Nugraha AP, Narmada IB, Ernawati DS. et al. Osteogenic potential of gingival stromal progenitor cells cultured in platelet rich fibrin is predicted by core-binding factor subunit-α1/Sox9 expression ratio ( in vitro). F1000 Res 2018; 7: 1134 DOI: 10.12688/f1000research.15423.1.
  • 10 Al-Qadhi G, Al-Rai S, Hafed L. The therapeutic potential of inflamed gingiva-derived mesenchymal stem cells in preclinical studies: a scoping review of a unique biomedical waste. Stem Cells Int 2021; 2021: 6619170 DOI: 10.1155/2021/6619170.
  • 11 Morsczeck C, Götz W, Schierholz J. et al. Isolation of precursor cells (PCs) from human dental follicle of wisdom teeth. Matrix Biol 2005; 24 (02) 155-165
  • 12 Zhang J, Ding H, Liu X, Sheng Y, Liu X, Jiang C. Dental follicle stem cells: tissue engineering and immunomodulation. Stem Cells Dev 2019; 28 (15) 986-994
  • 13 Saimbi CS, Gautam A, Khan MA. , Nandlal. Periosteum as a barrier membrane in the treatment of intrabony defect: a new technique. J Indian Soc Periodontol 2014; 18 (03) 331-335
  • 14 Zhou LL, Liu W, Wu YM, Sun WL, Dörfer CE, Fawzy El-Sayed KM. Oral mesenchymal stem/progenitor cells: the immunomodulatory masters. Stem Cells Int 2020; 2020: 1327405 DOI: 10.1155/2020/1327405.
  • 15 Fournier BPJ, Larjava H, Häkkinen L. Gingiva as a source of stem cells with therapeutic potential. Stem Cells Dev 2013; 22 (24) 3157-3177
  • 16 Stefańska K, Mehr K, Wieczorkiewicz M. et al. Stemness potency of human gingival cells-application in anticancer therapies and clinical trials. Cells 2020; 9 (08) E1916 DOI: 10.3390/cells9081916.
  • 17 Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell 2012; 10 (06) 709-716
  • 18 Huang GTJ, Gronthos S, Shi S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. J Dent Res 2009; 88 (09) 792-806
  • 19 Fawzy El-Sayed KM, Dörfer CE. Gingival mesenchymal stem/progenitor cells: a unique tissue engineering gem. Stem Cells Int 2016; 2016: 7154327 DOI: 10.1155/2016/7154327.
  • 20 Zhang N, Hu L, Cao Z, Liu X, Pan J. Periosteal skeletal stem cells and their response to bone injury. Front Cell Dev Biol 2022; 10: 812094 DOI: 10.3389/fcell.2022.812094.
  • 21 Kaku M, Komatsu Y, Mochida Y, Yamauchi M, Mishina Y, Ko C-C. Identification and characterization of neural crest-derived cells in adult periodontal ligament of mice. Arch Oral Biol 2012; 57 (12) 1668-1675
  • 22 Zhang Q, Nguyen AL, Shi S. et al. Three-dimensional spheroid culture of human gingiva-derived mesenchymal stem cells enhances mitigation of chemotherapy-induced oral mucositis. Stem Cells Dev 2012; 21 (06) 937-947
  • 23 Lima RL, Holanda-Afonso RC, Moura-Neto V, Bolognese AM, DosSantos MF, Souza MM. Human dental follicle cells express embryonic, mesenchymal and neural stem cells markers. Arch Oral Biol 2017; 73: 121-128
  • 24 Ichikawa Y, Watahiki J, Nampo T. et al. Differences in the developmental origins of the periosteum may influence bone healing. J Periodontal Res 2015; 50 (04) 468-478
  • 25 Ceccarelli G, Graziano A, Benedetti L. et al. Osteogenic potential of human oral-periosteal cells (PCs) isolated from different oral origin: an in vitro study. J Cell Physiol 2016; 231 (03) 607-612
  • 26 Suphanantachat S, Iwata T, Ishihara J, Yamato M, Okano T, Izumi Y. A role for c-Kit in the maintenance of undifferentiated human mesenchymal stromal cells. Biomaterials 2014; 35 (11) 3618-3626
  • 27 Debnath S, Yallowitz AR, McCormick J. et al. Discovery of a periosteal stem cell mediating intramembranous bone formation. Nature 2018; 562 (7725): 133-139
  • 28 Lee H, Park J-B. Dimethyl sulfoxide leads to decreased osteogenic differentiation of stem cells derived from gingiva via Runx2 and collagen i expression. Eur J Dent 2019; 13 (02) 131-136
  • 29 Guo S, Guo W, Ding Y. et al. Comparative study of human dental follicle cell sheets and periodontal ligament cell sheets for periodontal tissue regeneration. Cell Transplant 2013; 22 (06) 1061-1073
  • 30 Nugraha AP, Ramadhani NF, Riawan W. et al. Gingival mesenchymal stem cells metabolite decreasing TRAP, NFATc1, and sclerostin expression in LPS-associated inflammatory osteolysis in vivo. Eur J Dent 2023; 17 (03) 881-888
  • 31 Morsczeck CO, Drees J, Gosau M. Lipopolysaccharide from Escherichia coli but not from Porphyromonas gingivalis induce pro-inflammatory cytokines and alkaline phosphatase in dental follicle cells. Arch Oral Biol 2012; 57 (12) 1595-1601
  • 32 Pevsner-Fischer M, Morad V, Cohen-Sfady M. et al. Toll-like receptors and their ligands control mesenchymal stem cell functions. Blood 2007; 109 (04) 1422-1432
  • 33 Andrukhov O, Hong JSA, Andrukhova O, Blufstein A, Moritz A, Rausch-Fan X. Response of human periodontal ligament stem cells to IFN-γ and TLR-agonists. Sci Rep 2017; 7 (01) 12856 DOI: 10.1038/s41598-017-12480-7.
  • 34 Shin C, Kim M, Han JA. et al. Human periodontal ligament stem cells suppress T-cell proliferation via down-regulation of non-classical major histocompatibility complex-like glycoprotein CD1b on dendritic cells. J Periodontal Res 2017; 52 (01) 135-146
  • 35 Su WR, Zhang QZ, Shi SH, Nguyen AL, Le AD. Human gingiva-derived mesenchymal stromal cells attenuate contact hypersensitivity via prostaglandin E2-dependent mechanisms. Stem Cells 2011; 29 (11) 1849-1860
  • 36 Chen X, Yang B, Tian J. et al. Dental follicle stem cells ameliorate lipopolysaccharide-induced inflammation by secreting TGF-β3 and TSP-1 to elicit macrophage M2 polarization. Cell Physiol Biochem 2018; 51 (05) 2290-2308
  • 37 Chatzivasileiou K, Lux CA, Steinhoff G, Lang H. Dental follicle progenitor cells responses to Porphyromonas gingivalis LPS. J Cell Mol Med 2013; 17 (06) 766-773
  • 38 He F, Umrath F, von Ohle C, Reinert S, Alexander D. Analysis of the influence of jaw periosteal cells on macrophages phenotype using an innovative horizontal coculture system. Biomedicines 2021; 9 (12) 1753 DOI: 10.3390/biomedicines9121753.
  • 39 Queiroz A, Albuquerque-Souza E, Gasparoni LM. et al. Therapeutic potential of periodontal ligament stem cells. World J Stem Cells 2021; 13 (06) 605-618
  • 40 Ge S, Zhao N, Wang L, Liu H, Yang P. Effects of hydroxyapatite nanostructure on channel surface of porcine acellular dermal matrix scaffold on cell viability and osteogenic differentiation of human periodontal ligament stem cells. Int J Nanomedicine 2013; 8: 1887-1895
  • 41 Moshaverinia A, Xu X, Chen C. et al. Application of stem cells derived from the periodontal ligament or gingival tissue sources for tendon tissue regeneration. Biomaterials 2014; 35 (09) 2642-2650
  • 42 Yu B-H, Zhou Q, Wang Z-L. Periodontal ligament versus bone marrow mesenchymal stem cells in combination with Bio-Oss scaffolds for ectopic and in situ bone formation: a comparative study in the rat. J Biomater Appl 2014; 29 (02) 243-253
  • 43 Park S-Y, Kim K-H, Gwak E-H. et al. Ex vivo bone morphogenetic protein 2 gene delivery using periodontal ligament stem cells for enhanced re-osseointegration in the regenerative treatment of peri-implantitis. J Biomed Mater Res A 2015; 103 (01) 38-47
  • 44 Xu Q, Li B, Yuan L. et al. Combination of platelet-rich plasma within periodontal ligament stem cell sheets enhances cell differentiation and matrix production. J Tissue Eng Regen Med 2017; 11 (03) 627-636
  • 45 Iwasaki K, Komaki M, Yokoyama N. et al. Periodontal regeneration using periodontal ligament stem cell-transferred amnion. Tissue Eng Part A 2014; 20 (3-4): 693-704
  • 46 Moshaverinia A, Chen C, Akiyama K. et al. Encapsulated dental-derived mesenchymal stem cells in an injectable and biodegradable scaffold for applications in bone tissue engineering. J Biomed Mater Res A 2013; 101 (11) 3285-3294
  • 47 Qiu J, Wang X, Zhou H. et al. Enhancement of periodontal tissue regeneration by conditioned media from gingiva-derived or periodontal ligament-derived mesenchymal stem cells: a comparative study in rats. Stem Cell Res Ther 2020; 11 (01) 42 DOI: 10.1186/s13287-019-1546-9.
  • 48 Nakao Y, Fukuda T, Zhang Q. et al. Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater 2021; 122: 306-324
  • 49 Abdal-Wahab M, Abdel Ghaffar KA, Ezzatt OM, Hassan AAA, El Ansary MMS, Gamal AY. Regenerative potential of cultured gingival fibroblasts in treatment of periodontal intrabony defects (randomized clinical and biochemical trial). J Periodontal Res 2020; 55 (03) 441-452
  • 50 Hasani-Sadrabadi MM, Sarrion P, Pouraghaei S. et al. An engineered cell-laden adhesive hydrogel promotes craniofacial bone tissue regeneration in rats. Sci Transl Med 2020; 12 (534) eaay6853 DOI: 10.1126/scitranslmed.aay6853.
  • 51 Kandalam U, Kawai T, Ravindran G. et al. Predifferentiated gingival stem cell-induced bone regeneration in rat alveolar bone defect model. Tissue Eng Part A 2021; 27 (5-6): 424-436
  • 52 Diomede F, Gugliandolo A, Cardelli P. et al. Three-dimensional printed PLA scaffold and human gingival stem cell-derived extracellular vesicles: a new tool for bone defect repair. Stem Cell Res Ther 2018; 9 (01) 104 DOI: 10.1186/s13287-018-0850-0.
  • 53 Zhang Q, Nguyen PD, Shi S, Burrell JC, Cullen DK, Le AD. 3D bio-printed scaffold-free nerve constructs with human gingiva-derived mesenchymal stem cells promote rat facial nerve regeneration. Sci Rep 2018; 8 (01) 6634 DOI: 10.1038/s41598-018-24888-w.
  • 54 Honda MJ, Imaizumi M, Tsuchiya S, Morsczeck C. Dental follicle stem cells and tissue engineering. J Oral Sci 2010; 52 (04) 541-552
  • 55 Oshima M, Inoue K, Nakajima K. et al. Functional tooth restoration by next-generation bio-hybrid implant as a bio-hybrid artificial organ replacement therapy. Sci Rep 2014; 4: 6044 DOI: 10.1038/srep06044.
  • 56 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 (09) 632-645
  • 57 Salgado CL, Barrias CC, Monteiro FJM. Clarifying the tooth-derived stem cells behavior in a 3D biomimetic scaffold for bone tissue engineering applications. Front Bioeng Biotechnol 2020; 8: 724 DOI: 10.3389/fbioe.2020.00724.
  • 58 Yang H, Li J, Hu Y. et al. Treated dentin matrix particles combined with dental follicle cell sheet stimulate periodontal regeneration. Dent Mater 2019; 35 (09) 1238-1253
  • 59 Tian Y, Bai D, Guo W. et al. Comparison of human dental follicle cells and human periodontal ligament cells for dentin tissue regeneration. Regen Med 2015; 10 (04) 461-479
  • 60 Guo W, Gong K, Shi H. et al. Dental follicle cells and treated dentin matrix scaffold for tissue engineering the tooth root. Biomaterials 2012; 33 (05) 1291-1302
  • 61 Tsuchiya S, Ohshima S, Yamakoshi Y, Simmer JP, Honda MJ. Osteogenic differentiation capacity of porcine dental follicle progenitor cells. Connect Tissue Res 2010; 51 (03) 197-207
  • 62 Kawase T, Okuda K, Kogami H. et al. Characterization of human cultured periosteal sheets expressing bone-forming potential: in vitro and in vivo animal studies. J Tissue Eng Regen Med 2009; 3 (03) 218-229
  • 63 Nagata M, Hoshina H, Li M. et al. A clinical study of alveolar bone tissue engineering with cultured autogenous periosteal cells: coordinated activation of bone formation and resorption. Bone 2012; 50 (05) 1123-1129
  • 64 Agata H, Asahina I, Yamazaki Y. et al. Effective bone engineering with periosteum-derived cells. J Dent Res 2007; 86 (01) 79-83
  • 65 Ueno T, Honda K, Hirata A. et al. Histological comparison of bone induced from autogenously grafted periosteum with bone induced from autogenously grafted bone marrow in the rat calvarial defect model. Acta Histochem 2008; 110 (03) 217-223
  • 66 Cicconetti A, Sacchetti B, Bartoli A. et al. Human maxillary tuberosity and jaw periosteum as sources of osteoprogenitor cells for tissue engineering. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2007; 104 (05) 618.e1-618.e12
  • 67 Ge S, Zhao N, Wang L. et al. Bone repair by periodontal ligament stem cellseeded nanohydroxyapatite-chitosan scaffold. Int J Nanomedicine 2012; 7: 5405-5414
  • 68 Wang F, Yu M, Yan X. et al. Gingiva-derived mesenchymal stem cell-mediated therapeutic approach for bone tissue regeneration. Stem Cells Dev 2011; 20 (12) 2093-2102
  • 69 Al-Qadhi G, Aboushady I, Al-Sharabi N. The gingiva from the tissue surrounding the bone to the tissue regenerating the bone: a systematic review of the osteogenic capacity of gingival mesenchymal stem cells in preclinical studies. Stem Cells Int 2021; 2021: 6698100 DOI: 10.1155/2021/6698100.
  • 70 Diomede F, Rajan TS, D'Aurora M. et al. Stemness characteristics of periodontal ligament stem cells from donors and multiple sclerosis patients: a comparative study. Stem Cells Int 2017; 2017: 1606125 DOI: 10.1155/2017/1606125.
  • 71 Lee JS, An SY, Kwon IK, Heo JS. Transdifferentiation of human periodontal ligament stem cells into pancreatic cell lineage. Cell Biochem Funct 2014; 32 (07) 605-611
  • 72 Ulusoy C, Zibandeh N, Yıldırım S. et al. Dental follicle mesenchymal stem cell administration ameliorates muscle weakness in MuSK-immunized mice. J Neuroinflammation 2015; 12 (01) 231 DOI: 10.1186/s12974-015-0451-0.
  • 73 Shi HZ, Zeng JC, Shi SH, Giannakopoulos H, Zhang QZ, Le AD. Extracellular vesicles of GMSCs alleviate aging-related cell senescence. J Dent Res 2021; 100 (03) 283-292
  • 74 Subbarayan R, Barathidasan R, Raja STK. et al. Human gingival derived neuronal cells in the optimized caffeic acid hydrogel for hemitransection spinal cord injury model. J Cell Biochem 2020; 121 (03) 2077-2088
  • 75 Ansari S, Sarrion P, Hasani-Sadrabadi MM, Aghaloo T, Wu BM, Moshaverinia A. Regulation of the fate of dental-derived mesenchymal stem cells using engineered alginate-GelMA hydrogels. J Biomed Mater Res A 2017; 105 (11) 2957-2967
  • 76 Al-Qadhi G, Soliman M, Abou-Shady I, Rashed L. Gingival mesenchymal stem cells as an alternative source to bone marrow mesenchymal stem cells in regeneration of bone defects: in vivo study. Tissue Cell 2020; 63: 101325 DOI: 10.1016/j.tice.2019.101325.
  • 77 Ryu N-E, Lee S-H, Park H. Spheroid culture system methods and applications for mesenchymal stem cells. Cells 2019; 8 (12) E1620 DOI: 10.3390/cells8121620.
  • 78 Basu A, Rothermund K, Ahmed MN, Syed-Picard FN. Self-assembly of an organized cementum-periodontal ligament-like complex using scaffold-free tissue engineering. Front Physiol 2019; 10: 422 DOI: 10.3389/fphys.2019.00422.
  • 79 Hsu SH, Huang G-S, Feng F. Isolation of the multipotent MSC subpopulation from human gingival fibroblasts by culturing on chitosan membranes. Biomaterials 2012; 33 (09) 2642-2655
  • 80 Zhang Z, Shuai Y, Zhou F. et al. PDLSCs regulate angiogenesis of periodontal ligaments via VEGF transferred by exosomes in periodontitis. Int J Med Sci 2020; 17 (05) 558-567
  • 81 Pizzicannella J, Gugliandolo A, Orsini T. et al. Engineered extracellular vesicles from human periodontal-ligament stem cells increase VEGF/VEGFR2 expression during bone regeneration. Front Physiol 2019; 10: 512 DOI: 10.3389/fphys.2019.00512.
  • 82 Liu T, Hu W, Zou X. et al. Human periodontal ligament stem cell-derived exosomes promote bone regeneration by altering microRNA profiles. Stem Cells Int 2020; 2020: 8852307 DOI: 10.1155/2020/8852307.
  • 83 Ma L, Rao N, Jiang H. et al. Small extracellular vesicles from dental follicle stem cells provide biochemical cues for periodontal tissue regeneration. Stem Cell Res Ther 2022; 13 (01) 92 DOI: 10.1186/s13287-022-02767-6.
  • 84 Iwata T, Yamato M, Washio K. et al. Periodontal regeneration with autologous periodontal ligament-derived cell sheets - a safety and efficacy study in ten patients. Regen Ther 2018; 9: 38-44
  • 85 Nakahara T, Nakamura T, Kobayashi E. et al. In situ tissue engineering of periodontal tissues by seeding with periodontal ligament-derived cells. Tissue Eng 2004; 10 (3-4): 537-544
  • 86 Akbay A, Baran C, Günhan O, Ozmeriç N, Baloş K. Periodontal regenerative potential of autogenous periodontal ligament grafts in class II furcation defects. J Periodontol 2005; 76 (04) 595-604
  • 87 Chen F-M, Gao L-N, Tian B-M. et al. Treatment of periodontal intrabony defects using autologous periodontal ligament stem cells: a randomized clinical trial. Stem Cell Res Ther 2016; 7: 33 DOI: 10.1186/s13287-016-0288-1.
  • 88 Kim D, Lee AE, Xu Q, Zhang Q, Le AD. Gingiva-derived mesenchymal stem cells: potential application in tissue engineering and regenerative medicine - a comprehensive review. Front Immunol 2021; 12: 667221 DOI: 10.3389/fimmu.2021.667221.
  • 89 Ponnaiyan D, Jegadeesan V. Comparison of phenotype and differentiation marker gene expression profiles in human dental pulp and bone marrow mesenchymal stem cells. Eur J Dent 2014; 8 (03) 307-313

Zoom Image
Fig. 1 Origin and development of stem cells within the periodontium. (A) The neural crest ectomesenchyme and the neural crest cells—The thickening of ectomesenchyme with migration of neural epithelial cells form a tooth bud. (B) Condensation of mesenchyme to form dental papilla and follicle giving rise to dental follicle stem cells. Inner layer of follicle (dental follicle proper) gives rise to periodontal ligament cells, the denotgingival fiber system harboring periodontal ligament stem cells (PDLSCs) and alveolar bone harboring oral periosteal stem cells (OPSCs), outer layer of follicle (perifollicular mesenchyme) forms gingival lamina propria that harbors gingival mesenchymal stem cells (GMSCs). (C) Mature tooth showing location of PDLSCs, GMSCs, and OPSCs.
Zoom Image
Fig. 2 Immunomodulation and regeneration potential of periodontal stem cells. (A) Inflammation and stem cells “The Bidirectional Effect” affecting their differentiation. (B) Immunomodulatory dentistry—Regeneration of periodontal tissues like bone, cementum, and periodontal ligament. (C) Immunomodulatory medicine—regeneration into various tissues of the body.