Next Article in Journal
Brassica oleracea L. var. italica Aquaporin Reconstituted Proteoliposomes as Nanosystems for Resveratrol Encapsulation
Next Article in Special Issue
Donor Sites and Harvesting Techniques Affect miRNA Cargos of Extracellular Vesicles Released by Human Adipose-Derived Mesenchymal Stromal Cells
Previous Article in Journal
ATP-Induced Contractile Response of Esophageal Smooth Muscle in Mice
Previous Article in Special Issue
Mesenchymal Stem/Stromal Cells Induce Myeloid-Derived Suppressor Cells in the Bone Marrow via the Activation of the c-Jun N-Terminal Kinase Signaling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesenchymal Stromal Cells Derived from Dental Tissues: Immunomodulatory Properties and Clinical Potential

by
Luis Ignacio Poblano-Pérez
1,
Marta Elena Castro-Manrreza
2,
Patricia González-Alva
3,
Guadalupe R. Fajardo-Orduña
1 and
Juan José Montesinos
1,*
1
Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center (IMSS), Mexico City 06720, Mexico
2
Immunology and Stem Cells Laboratory, FES Zaragoza, National Autonomous University of Mexico (UNAM), Mexico City 09230, Mexico
3
Tissue Bioengineering Laboratory, Postgraduate Studies, Research Division, Faculty of Dentistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(4), 1986; https://doi.org/10.3390/ijms25041986
Submission received: 30 December 2023 / Revised: 30 January 2024 / Accepted: 5 February 2024 / Published: 6 February 2024

Abstract

:
Mesenchymal stem/stromal cells (MSCs) are multipotent cells located in different areas of the human body. The oral cavity is considered a potential source of MSCs because they have been identified in several dental tissues (D-MSCs). Clinical trials in which cells from these sources were used have shown that they are effective and safe as treatments for tissue regeneration. Importantly, immunoregulatory capacity has been observed in all of these populations; however, this function may vary among the different types of MSCs. Since this property is of clinical interest for cell therapy protocols, it is relevant to analyze the differences in immunoregulatory capacity, as well as the mechanisms used by each type of MSC. Interestingly, D-MSCs are the most suitable source for regenerating mineralized tissues in the oral region. Furthermore, the clinical potential of D-MSCs is supported due to their adequate capacity for proliferation, migration, and differentiation. There is also evidence for their potential application in protocols against autoimmune diseases and other inflammatory conditions due to their immunosuppressive capacity. Therefore, in this review, the immunoregulatory mechanisms identified at the preclinical level in combination with the different types of MSCs found in dental tissues are described, in addition to a description of the clinical trials in which MSCs from these sources have been applied.

1. Introduction

Mesenchymal stem/stromal cells (MSCs) were first discovered in the bone marrow (BM-MSCs) of guinea pigs more than 50 years ago by Friedenstein and his collaborators [1]. The International Society for Cell Therapy defines these cells as adherent cells with a fibroblast morphology that simultaneously express the immunophenotypic markers CD105, CD90, and CD73, present low levels of human leukocyte antigen (HLA) class I molecules and lack HLA class II molecules and markers of hematopoietic and endothelial cells. More recently, new surface molecules have been discovered, and MSCs are now recognized as cells that express STRO-1, CD106, and CD146. Finally, MSCs can differentiate when cultured in a specific inducing medium, at least in adipocytes, chondroblasts, and osteoblasts [2].
BM-MSCs are characterized by regenerative and immunomodulatory properties that can be exploited in clinical treatments for various immune diseases [3,4]. Unfortunately, obtaining them from this source presents drawbacks that reduce their feasibility for use in cell therapy as a result of the painful and invasive process involved [5]. The biological potential and number of cells may vary or decrease with the sex and age of the donor [6,7,8,9,10,11]. When cultivated in the long term, proliferation is negatively affected [12], as are the morphology, differentiation capacity, and genetic stability [13]. Finally, the anti-inflammatory potential of these cells may be compromised when they are obtained from donors with some pathological conditions, such as rheumatoid arthritis [14], which would reduce the possibility of using autologous samples. For these reasons, alternative sources to BM where these inconveniences are minimized are needed.
Currently, MSCs can be obtained from neonatal tissues, including umbilical cord blood (UCB) [15], the umbilical cord (UC), and the placenta [16], as well as adult sources, such as adipose tissue (AT) [17], synovial fluid [18], the skin [19], the lungs [20], the liver [21], peripheral blood [22,23,24,25], and dental tissues [26]. Most MSCs from dental tissues, possess similar properties to MSCs from BM; however, there are still some gaps in the knowledge of their biological characteristics.
The aim of this review is therefore to analyze the immunoregulatory mechanisms of the different types of MSCs found in dental tissues, as well as to describe the preclinical and clinical trials in which MSCs from these sources have been applied. In order to find alternative sources of MSCs to bone marrow that may be applicable in diseases where the immune system is involved, it is necessary to understand the immunological characteristics and mechanisms of D-MSCs.

2. MSCs from Dental and Periodontal Tissues

The teeth are essential multifunctional appendages for functions such as speaking or eating [27]. They are divided into two main regions: the upper part or crown and the root, which is anchored within the mandibular and maxillary bones. In humans, there are two sets of teeth: the initial deciduous or primary teeth and the successive permanent (secondary) teeth [28].
Structurally, teeth are made up of three highly mineralized tissues called enamel, dentin, and cement, whose functions include providing support, size, shape, and anchorage [29,30]. Enamel and dentin also protect a fourth nonmineralized tissue known as the dental pulp. This area serves as a reservoir for fibroblasts and is characterized by a vascular–nervous system that features a unique combination of blood vessels, nerves, odontoblasts, and an extracellular matrix [31,32]. The vascular–nervous system plays a fundamental role in tooth function, feeding it from the apex without accessory vascularization. Unlike other oral tissues adjacent to the tooth, this mechanism maintains homeostasis and provides the tooth with neurosensory function, aiding in repair processes [27,33,34]. On the other hand, teeth also rely on other tissues, including the periodontal ligament, the alveolar bone, and the gingival tissue; together, these tissues are known as periodontal tissues or simply the periodontium.
The functions of the periodontium include supporting the tooth and protecting it against invasion by oral microorganisms; it is essential in the immune response and allows the attachment of the tooth to the bone [35,36].
MSCs have been isolated from various anatomical regions of the oral cavity (Figure 1), including the dental pulp (DPSCs) [37,38], periodontal ligament (PDLSCs) [39,40], gingival tissue (GMSCs) [41,42], apical papilla (SCAPs) [43,44], dental follicle (DFSCs) [45,46], human exfoliated deciduous teeth (SHED) [47,48], alveolar bone (ABMSCs), and tooth germ (TGPCs) [34], and these are collectively defined as dental MSCs (D-MSCs) [49].
Dental tissues are frequently obtained during routine dental extraction procedures. The dental pieces and/or tissues are kept in phosphate-buffered solutions, usually supplemented with antibiotics and antifungals, and a series of washes with fresh solution are performed to remove unwanted tissues. The tissues of interest are stored at 4 °C before beginning the procedures to obtain MSCs. To access and obtain the dental pulp, the teeth are crushed or the crowns are cut. Other tissues such as the ligament, gingiva, and apical papilla can be separated mechanically by using forceps [50,51,52,53,54,55].
The most common methods used to obtain MSCs are explant and enzymatic digestion. In the first method, the tissues obtained are crushed into small pieces of 1 mm and placed on plates with ideal culture medium for the growth of these cells supplemented with fetal bovine serum and antibiotics. They are then incubated at 37 °C and 5% CO2. Additionally, in some procedures, a coverslip can be added over the explants to prevent the tissues from moving during the culture time. On the other hand, enzymatic digestion is carried out through the incubation of the crushed tissues in collagenase I and dispase II. The incubation time can vary from 30 min to 2 h at 37 °C. Once digestion has been carried out, the cells are seeded on plates with the same culture conditions mentioned above. Once the cells adhere, the medium and excess tissues are removed and fresh medium is added to continue cell expansion, and subsequently, the capacity to generate colonies, morphology, immunophenotype, and the differentiation potential characteristic of the MSCs are evaluated [50,51,52,53,54,55]. Although most protocols use enzymatic digestion to obtain D-MSCs, both methods are reliable. They can obtain a high number of cells and there is no solid evidence that either affects the biological properties of the cells. However, isolation methods continue to improve with the purpose of obtaining the greatest number of cells with the necessary quality required to be able to use them in preclinical and clinical research.
D-MSCs have several similar characteristics which are summarized in Table 1; for example, they come from easily accessible tissues, they exhibit fibroblast morphology, they provide a high number of MSCs with high proliferation rates, and, unlike MSCs from other sources, they maintain their qualities for more passages [49,56,57,58,59]. However, differences have been observed in other characteristics, such as the immunophenotype, where the expression of some markers may vary. In addition, although they have similar multipotentialities for differentiation toward chondrocytes and osteoblasts, their adipogenic capacity is lower than that of other tissues, such as BM [58,60,61,62,63,64]. Moreover, D-MSCs derived from neural crest cells possess an increased potential for differentiation into neural cells [65,66,67,68]. Taken together, these findings indicate that although all of these sources originate in related anatomical regions, the biological properties of each can vary, and of particular importance is their immunosuppressive capacity due to their relevance in clinical protocols. In the case of D-MSCs, they have great therapeutic potential for applications in tissue repair, including that of bone, dental, and soft tissues.

3. Immunomodulatory Properties of MSCs

In vitro, preclinical and clinical studies have shown that MSCs are capable of regulating inflammatory processes, an essential event for reducing tissue damage and promoting tissue repair [79]. To do this, they migrate to injured sites through the support of adhesion molecules, chemokines, and their receptors, as well as chemoattractant molecules, such as hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), transforming growth factor β (TGF-β) 1, granulocyte colony stimulating factor, and tumor necrosis factor α (TNF-α) [80,81,82,83]. Once they reach the site of inflammation, MSCs regulate the proliferation, differentiation, maturation, and production of soluble factors and the cytotoxicity of immune cells [84]. The regulatory mechanisms of MSCs include the secretion of soluble factors, the production of metabolites, cell–cell contact, and the release of extracellular vesicles (EVs).
Some of the anti-inflammatory molecules that MSCs secrete include interleukin (IL)-10 [85,86,87,88], IL-6 [89,90], TGF-β [91,92,93], prostaglandin E2 (PGE2) [94,95,96], gene-6 stimulated by tumor necrosis factor (TSG-6) [97,98], HLA-G5 [99,100], galectins [101], and chemokine ligand 2 with a CC motif (CCL2) [101]. The participation of each secreted factor in the regulation of the immune response will be explained in the following sections.
On the other hand, MSCs express intra- and extracellular enzymes that help the formation of anti-inflammatory metabolites, such as inducible nitric oxide synthase (iNOS), which allows for the generation of nitric oxide (NO) and reduces the migration of immune cells by decreasing chemoattractant molecules and the production of inflammatory cytokines [102,103,104], or indoleamine 2,3-dioxygenase (IDO), which depletes tryptophan in immune cells and generates anti-inflammatory molecules, including kinurenine and picolinic acid [105,106,107]. Finally, MSCs express the ectonucleotidases CD39 and CD73, which generate adenosine (ADO), modulating the production of inflammatory cytokines, cytotoxicity, apoptosis, and the proliferation of immune cells [108,109,110,111,112].
Among their membrane molecules, MSCs express programmed death ligands (PDLs) 1 and 2 [113,114], HLA-G1 [100], cytotoxic T lymphocyte antigen 4 (CTLA-4) [115], and intercellular adhesion molecule-1 (ICAM-1) [116]. Finally, the EVs secreted by MSCs transport and express immunomodulatory molecules and cytokines, such as microRNAs, TGF-β, galectin 1, and PDL-1 [117].
The immunomodulatory capacity of MSCs is of great relevance to the application of these cells in the clinical field. It is necessary to know in depth the biological mechanisms that allow them to sense their microenvironment to activate this property; Preclinical and clinical trials have given good results, and MSCs have been evaluated to improve adverse physiological conditions related to the immune system, which has created great expectations in cell therapy procedures.

4. Immunomodulatory Properties of D-MSCs

The anti-inflammatory characteristics of MSCs have been widely described, especially those of MSCs derived from BM, AT, and UCB [118,119]. However, recent studies have shown that D-MSCs also express immunoregulatory molecules (Table 2; Figure 2). In this regard, our workgroup has shown that, in a similar way to that of BM-MSCs, DPSCs, PDLSCs, and GMSCs reduce the proliferation of activated CD3+ T lymphocytes and the production of TNF-α and increase the production of anti-inflammatory molecules such as IL-10 and PGE2, in addition to generating regulatory CD4+ CD25+ Foxp3+ T lymphocytes. In these cocultures, some mechanisms of cell contact have also been observed, since, in MSCs, they increase the expression of PDL-1, while in T lymphocytes, the presence of CTLA-4 increases [60]. However, other mechanisms have been described by different workgroups [120]. Table 2 shows the immunoregulatory molecules that have been reported to be expressed by MSCs from different sources, the effects of which on immune cells are described below.
As discussed below, dental tissues are very promising alternative sources of MSCs to bone marrow because such tissues are usually discarded in routine dental procedures and can be obtained without significant ethical concerns. The following subsections indicate the immunomodulatory properties of the different subtypes of D-MSCs.

4.1. Dental Pulp MSCs

DPSCs are characterized by a high proliferation rate and immunomodulatory effects that are exerted through different mechanisms that affect different populations of immune cells [173,174,175]. These MSCs secrete IL-6, TGF-β, IDO, HLAG, and HGF, express PDL-1, rely on the Fas-FasL pathway, produce ADO, release EVs to reduce the proliferation of T lymphocytes, induce apoptosis, decrease the differentiation capacity of some subpopulations of proinflammatory T helper (Th) cells, such as Th17 lymphocytes, decrease the secretion of TNF-α and IL-17, and promote the generation of Tregs and the secretion of IL-10 and TGF-β [114,138,139,140,144].
On the other hand, it has been observed that hypoxia-inducible factor 1 (HIF-1) induces DPSCs to decrease the maturation of monocytes toward dendritic cells (DCs) and induce the polarization of M2 macrophages [141]. In addition, they secrete osteoprotegerin and release EVs loaded with microRNAs to inhibit the osteoclastogenesis of myeloid cells and polarize them toward M2 macrophages [142,143]. It has been shown that these types of MSCs also affect natural killer (NK) lymphocytes, since they inhibit their proliferation, induce apoptosis, and decrease their cytotoxicity and the expression of activator molecules such as NKG2D. In addition, MSCs induce the expression of CD73 in NK cells, which favors the generation of ADO [176]. In a recent study, DPSCs cultured with activated T lymphocytes from patients infected with COVID-19 decreased the secretion of several proinflammatory cytokines, including interferon γ (IFN-γ), TNF-α, IL-2, IL-5, IL-9, IL-12 (p70), IL-17A, IL-18, IL-21, IL-23, and IL-2 [177].
On the other hand, DPSCs—or EVs secreted from them—have been used in in vivo studies of induced arthritis in rodents and have been shown to be capable of reducing cartilage and bone tissue wear, the presence of TNF-α and IFN-γ, the production of metalloproteinases, and the differentiation of Th17 cells [178,179,180]. Other studies have shown that they are capable of inhibiting the activation and proliferation of microglia and, thus, reducing the production of proinflammatory cytokines in neuroinflammation caused by spinal cord injury [181]. In this regard, progenitors of DPSCs migrate from the cranial neural crest into the pharyngeal arches, and their genetic line has been shown to have a close relationship with pericytes and glial cells [182].
Moreover, a previously conducted study demonstrated that exosomes derived from lipopolysaccharide (LPS)-preconditioned DPSCs could enhance Shawn cells’ proliferation, migration, and odontogenic differentiation. Different research groups have demonstrated that stimulation of MSCs with different TLR ligands produces proinflammatory mediators, including IL-6, IL-8, and monocyte chemoattractant protein-1 (MCP-1) [183]. These findings suggest that MSCs are involved in the progression of different inflammatory diseases. However, the precise role of MSCs in oral inflammatory conditions—particularly pulpitis and periodontitis—needs further investigation. An interesting observation is that senescent DPSCs can retain an active cellular metabolism and produce functional exosomes, which can penetrate the blood barrier. This property makes exosomes useful therapeutic agents for treating various diseases, including neurological disorders, cancer, and pulmonary diseases, as well as other pathologies [184].
Finally, previously published research has demonstrated that the DPSC-derived extracellular matrix has the potential to enhance the integration of bone grafts and bone regeneration and repair. Hence, it can serve as a potential tool for enhancing the efficacy of various biomaterials (scaffolds and hydrogels) within the field of bone tissue engineering [185].

4.2. Periodontal Ligament MSCs

PDLSCs reduce the proliferation of T lymphocytes through the secretion of TGF-β, IDO, HGF, and PGE2 [145,146,147,148]. In addition, PDLSCs release EVs loaded with microRNAs and restore the balance between Tregs and Th17 lymphocytes [186]. On the other hand, the coculture of PDLSCs with DCs reduced the expression of the CD1b glycoprotein in DCs, preventing them from activating T lymphocytes appropriately [187].
PDLSCs modulate the proliferation, differentiation, migration, and production of immunoglobulins in B cells through programmed cell death protein 1 and its ligands PDL-1 and 2. In addition, the transplantation of allogenic human PDLSCs successfully suppressed the progression of bone resorption in a minipig periodontitis model [188]. Furthermore, through the secretion of the chemokines RANTES, eotaxin, IFN-γ, inducible protein 10, and MCP-1, IL-6, IL-8, and IL-1ra promoted the recruitment of neutrophils to sites of infection so that they could carry out their antimicrobial functions [149].
Several in vivo studies have shown that the administration of PDLSCs favors periodontal regeneration by inducing the polarization of M2 macrophages [189] and inhibiting the proliferation and infiltration of T lymphocytes in affected tissues [146,188,190]. On the other hand, the conditioned media (CMs) of these cells contained VEGF, growth factors (insulin-like growth factor binding protein 6), and cytokines (TGF-β 1, 2, and 3; HGF, IL-6, and MCP-1), which were related to their immunomodulatory functions and tissue regeneration. The use of CMs in vitro induced the polarization of M2 macrophages [191], while their administration in rats with periodontal defects induced the regeneration of bone tissue, decreased the presence of IL-1β, IL-6, and TNF-α and increased IL-10 [192,193].
Thorough research has proven that MSCs regulate the immune response during inflammation by producing multiple factors. This production is usually upregulated by inflammatory cytokines and TLR ligands [194,195]. PDLSCs participate in alveolar bone metabolism and cementogenesis, which are processes that are vital for maintaining periodontal tissues. Considering the importance of PDLSCs, the potential effect of LPS on the osteogenic differentiation of PDLSCs and TLR receptors—mainly TLR-4’s capacity to recognize LPS—has been extensively investigated [194,196,197].
The overall results suggest that the impact of LPS on PDLSC differentiation into bone cells depends on the concentration and, to a lesser extent, the source of LPS. Understanding such mechanisms is crucial in identifying the causes of periodontitis and will contribute to the development of effective treatments [198].

4.3. Gingival Tissue MSCs

Gingival tissue is the most accessible source of MSCs, and cells are easily isolated and have great potential for ex vivo expansion [198]. In addition to their high capacity for renewal and proliferation, these cells have been shown to have immunomodulatory properties [42]; in vitro, they decrease the proliferation and differentiation of Th17 lymphocytes through the production of PGE2, IDO, iNOS, and IL-10 and the Fas-FasL pathway [42,150,156], and they induce the polarization of M2 macrophages [159].
The immunomodulatory and protective capacities of these cells have been evaluated in animal models of immune pathologies, including experimental colitis [42], induced rheumatoid arthritis [111,152], type 1 diabetes mellitus [153], atherosclerosis [110], graft versus host disease [59,154], lupus nephritis [155], and chemotherapy-induced mucositis [141]. In these models, GMSCs migrate to inflamed regions and lymph nodes near these sites, where they decrease the proliferation and differentiation of Th1, Th2, and Th17 lymphocytes, as well as their ability to infiltrate the affected tissues; in addition, they affect the cytotoxicity of CD8+ T cells. On the other hand, they inhibit the proliferation, differentiation, maturation, and activation of B lymphocytes, decrease the migration of DCs, promote the polarization of macrophages to the M2 phenotype, inhibit osteoclastogenesis, and reduce the levels of TNF-α, IFN-γ, IL-17, and IL-4. Some of the most important immunomodulatory mechanisms of these MSCs are the adenosynergic pathway and the production of IL-10 and IDO.
In addition, the administration of GMSCs mitigated the oxidative stress-induced apoptosis of epithelial cells by regulating the activity of manganese superoxide dismutase and hypoxia-inducible factors 1 and 2α [141].
Interestingly, GMSCs induce the expression of CD39 in Tregs, which increases their anti-inflammatory capacity [59]. Additionally, GMSCs have been reported to maintain their proliferative and immunomodulatory potential longer than BM and AT MSCs [59].
In addition, GMSCs release EVs loaded with the IL-1 receptor antagonist (IL-1RA) miR1260b, and they express CD73. These vesicles induce M2 macrophage polarization, improve wound healing, and decrease osteoclastogenesis [151,157,158,159]. Likewise, the biomolecules present in the CMs of these cells decrease inflammation and improve bone regeneration in rats with periodontitis [192].

4.4. MSCs of the Apical Papilla

SCAPs have high proliferative potential and self-renewal capacity and low immunogenicity [43,199,200,201]. In vitro, they have been shown to decrease the proliferation of T lymphocytes and induce the generation of Tregs [200,202,203]. The mechanisms and immunomodulatory molecules used by these MSCs have not yet been defined; however, analysis of their secretome revealed the presence of IL-6, TGF-β 1 and 2, and galectin 1, among others [160,161,162,163].
These MSCs have been cocultivated with tissues derived from the spinal cord, and their ability to modulate microglia and mitigate neuroinflammation by increasing arginase 1 and decreasing TNF-α and NOS2, which translates into a neuroprotective effect, has been demonstrated [204,205].
In some in vivo models of experimental colitis, it has been shown that SCAPs can decrease the production of IL-1β, IL-6, and TNF-α and increase the generation of Treg lymphocytes in the lymph nodes. The Fas-FasL pathway seems to be one of the immunomodulatory mechanisms of these MSCs [206]. On the other hand, they also secrete EVs [207], which have been shown to be capable of generating Treg lymphocytes to decrease inflammation in rat models of periodontal bone defects [208].

4.5. Dental Follicle MSCs

DFSCs can be isolated from the dental follicle, a loose connective tissue around a developing tooth. They are a group of cells that play a fundamental role in the development and maintenance of the periodontal tissues during tooth development; they are accessible and abundant, have high proliferative and self-renewal potential, and have immunomodulatory properties [209]. DFSCs inhibit the expression of costimulatory molecules in monocytes, as well as the proliferation and differentiation of Th2 lymphocytes, but they allow the generation of Tregs through the secretion of TGF-β and IDO [164,210].
Chen et al. [165] stated that the CMs of these MSCs contained 42 paracrine factors related to their immunomodulatory capacity, of which TGF-β3 and thrombospondin 1 were the most abundant, and the use of these supernatants in macrophage cultures could polarize them to the M2 phenotype. On the other hand, the administration of DFSCs or their CMs to a rat model of acute lung damage similarly decreased the presence of inflammatory cytokines such as MCP-1, IL-1, IL-6, and TNF-α in bronchoalveolar lavage fluid. In addition, they promoted the secretion of IL-10 and the differentiation of M2 macrophages [165].
In a model of induced pulpitis in rats, CMs administration downregulated the ERK1/2 and NF-κB signaling pathways in active cells, which resulted in the suppression of IL-1β, IL-6, and TNF-α and promoted the expression of IL-4 and TGF-β; additionally, CM administration reduced the infiltration of immune cells into the tissues [166]. On the other hand, in a model of myasthenia gravis, its ability to decrease the proliferation of cells in the lymph nodes was observed, as was the production of IL-6, IL-12, and IgG antibodies [211].

4.6. MSCs from Human Exfoliated Deciduous Teeth

SHED are cells that have been shown to have different immunomodulatory mechanisms [212]; they reduce the secretion of IL-2, TNF-α, and IFN-γ and increase the secretion of IL-10 when they are cocultured with activated mononuclear cells (MNCs). In addition, they negatively affect the maturation of DCs or induce them to adopt a regulatory profile, which, in turn, generates Treg lymphocytes [167], inhibits the differentiation of Th17 lymphocytes [213], and polarizes macrophages toward the M2 profile [214]. On the other hand, SHED secrete EVs that carry miRNAs. Cultures of these EVs with cartilage cells decreased the production of IL-6, IL-8, and various metalloproteinases [168].
The administration of SHED in models of immune diseases, such as ovariectomy-induced osteoporosis [170], experimental autoimmune encephalitis [215], and induced arthritis [216], decreased the infiltration of T lymphocytes into tissues, the differentiation of Th1 and Th17 cells, and the presence of IFN-γ, TNF-α, IL-1β, and IL-4, which also induced the generation of Tregs. In an experimental periodontitis model, these cells were able to regenerate periodontal tissues, and this effect was attributed to their ability to polarize macrophages toward M2 [214]. Through the secretion of soluble PDL-1, these cells restored the balance between Th17 and Treg lymphocytes in a mouse model of Sjögren’s syndrome [169].
SHED-CMs also possess anti-inflammatory and protective properties; their administration in a model of multiple sclerosis decreased neuronal damage by reducing the production of inflammatory cytokines and inhibiting the infiltration and proliferation of CD4+ T lymphocytes, in addition to inducing the polarization of M2 macrophages [217]. In addition, a model of non-alcoholic steatohepatitis protected the liver and intestine through similar mechanisms [218], and it was evidenced that the protective effect of SHED-CMs was because they contained TGF-β and IL-10 [219].
EVs released by SHED have also been used in vivo. In a mouse model of lupus erythematosus, the mRNAs carried by these vesicles reached BM-MSCs, which showed a decrease in telomerase activity and, therefore, the loss of certain biological properties. With the support of EVs, these MSCs recovered their immunomodulatory capacity and, thus, managed to decrease the percentage of Th17 cells and increase the number of Tregs [220]. On the other hand, in a periodontitis model, the use of SHED-derived EVs decreased the presence of IL-6 and TNF-α in inflamed areas and improved bone regeneration [221].

4.7. MSCs from Alveolar Bone

ABMSCs were isolated for the first time by Matsubara and collaborators in 2005. These cells have the morphology, immunophenotype, and differentiation capacity of MSCs [222], and they have demonstrated the capacity for bone regeneration in vivo [223,224,225]; however, their immunomodulatory mechanisms have not been studied in depth. To date, it is only known that they inhibit the proliferation of T lymphocytes and monocytes and that they induce the differentiation of the latter towards an M2 phenotype. Its mechanisms are possibly due to its ability to produce IL-6, osteoprotegerin, and tissue inhibitors of metalloproteinase (TIMP)-1 and 2 [171,172].

5. The Induction of the Immunoregulatory Capacity of D-MSCs and the Release of Extracellular Vesicles

The importance of an inflammatory environment in the activation of MSCs has been demonstrated in numerous studies. This event allows their different immunosuppressive mechanisms to increase or be induced and is a process mediated mainly by IFN-γ and TNF-α [117], whose effect has been analyzed mainly on BM-MSCs. Regarding MSCs derived from dental tissues, it has been seen that the activation of PDLSCs with IL-1β, TNF-α, or IFN-γ increases the expression of the IDO gene at the protein level; the most pronounced effect is observed with IFN-γ, while stimulation with TNF-α or IFN-γ increases PDL1 and 2 expression at almost the same levels. However, only MSCs pre-treated with IFN-γ or IL-1β are able to affect T lymphocyte proliferation, whereas those treated with TNF-α are not [226]. The same working group observed that PDLSCs stimulated with IL-1β or IFN-γ increase IDO expression for at least 5 days, even after withdrawal of the stimulus. Interestingly, the increased IDO expression over a long period of time is observed only when PDLSCs are stimulated with IFN-γ, whereas the opposite effect is observed in MSCs treated with TNF-α [227].
On the other hand, it has been shown that the presence of bacterial molecules such as LPS or vitamins can also influence the biological properties of MSCs. It has been seen that treatment of PDLSC with IFN-γ increases IDO expression at the protein level; this effect is greater when a TLR2 agonist is added, while no such increase is observed with a TLR4 agonist [228]. Likewise, PDLSCs stimulated with TNF-α, IL-1β, or IFN-γ in the presence of vitamin D have been shown to have a greater capacity to decrease CD4+ T lymphocyte proliferation than those stimulated only with the pro-inflammatory cytokines [229]. Taken together, these data indicate that the balance between different cytokines plays an important role in the induction and maintenance of the immunoregulatory properties of MSCs, which is also influenced by the presence of other molecules.
It is currently proposed that MSCs release EVs, which are able to establish contact with immune cells and affect their proliferation, differentiation, and effector function. Interestingly, it has been observed that pro-inflammatory cytokines also modify the content of EVs released by MSCS. In this regard, it has been seen that GMSCs treated with TNF-α release exosomes enriched in CD73, which promote the polarization of macrophages towards an M2 phenotype, which correlates with reduced bone loss in a murine model of periodontitis [158]. Subsequently, the same working group demonstrated that treatment of GMSCs with TNF-α or IFN-γ, individually or in combination, does not significantly modify the CD73 expression of MSCs. However, their analysis of EVs showed a significant increase in CD73 and CD5L transport, and they determined that TNF-α and IFN-γ participate synergistically in that effect. Similarly, these EVs also favor M2-type macrophage differentiation [230]. Another conditioning method used is hypoxia, and a study carried out with MSCs derived from dental pulp shows that the release of EVs is increased in DPSCs subjected to hypoxic conditions, which has a greater capacity to favor M2 macrophage polarization, suppress osteoclast formation, and decrease bone loss in a murine model of LPS-induced inflammatory calvarial bone [231].
Due to the above, it has been proposed that EVs, by carrying immunoregulatory molecules and having an effect similar to that observed with MSCs, could be used as an alternative or complementary therapy to the use of whole cells; the possibility of using them as vehicles for drugs directed against transformed cells has also been proposed. In this regard, it has been seen that EVs released by GMSCs exposed to Paclitaxel contain the drug and are able to decrease the proliferation of transformed cell lines, including pancreatic cancer cells and squamous cell carcinoma [232]. Similar results have been observed with DPSCs which, when cultured with gemcitabine, release EVs loaded with the chemotherapeutic, which decreases the in vitro proliferation of pancreatic carcinoma cell lines [233].

6. D-MSCs in Reported Clinical Trials

The analysis of most published research reviewed in the present work shows that D-MSCs have been used for tissue engineering studies in large animals to assess their potential in preclinical applications. The results highlight their potential in dental medicine to regenerate pulp, bone, and periodontal tissues. In this regard, using natural and synthetic scaffolds with D-MSCs to regenerate entire teeth is also an active research area. Although numerous breakthroughs have been made in stem cell research, their success and applicability in clinical trials still need to be determined. Furthermore, D-MSCs possess immunomodulatory properties and have been used in preclinical studies and clinical trials for various diseases.
Due to the properties of D-MSCs, some working groups have begun to use them in clinical trials as treatments for various diseases, most of which involve accessing the oral cavity. To date, there are at least 26 published clinical trials in which D-MSCs are used as treatments (Table 3). The sources used in these tests were DPSCs, SHED, PDLSCs, and SCAPs. Most of the samples used were autologous, but some trials included allogeneic samples. Most of the studies were carried out to support the use of D-MSCs in promoting regeneration in the maxillofacial region. In evaluating the efficacy of D-MSCs for cell-based therapies, the clinical trials focused on numerous variables, including mineralization, bone regeneration, and repair, as well as periodontal parameters, such as tooth mobility, probing depth, gingival recession, the level of clinical insertion, and testing for vital pulp. Therefore, most clinical trials have evaluated the regeneration of tooth-supporting tissues and related parameters. The D-MSCs were administered intravenously, in suspension, via cell sheets, or, for the most part, via scaffolds made mainly of natural polymers, such as collagen.
Interestingly, although D-MSCs have been shown to interact effectively with immune cells in vitro and in animal models, clinical trials that have explored their immunomodulatory actions are scarce.
The first reported clinical trial was carried out by d’Aquino and collaborators in 2009. In this work, DPSCs embedded in a collagen sponge were used to repair bone defects generated after the extraction of third molars. The results revealed complete bone tissue regeneration and that the cortical level was higher in the experimental group than in the control group after three months of surgery. Additionally, the restoration of the periodontal tissue behind the second molars was also achieved with the proposed intervention. In addition, the patients did not experience symptoms of graft rejection or other complications, suggesting that DPSCs could be used safely and effectively in reconstructive and orthopedic surgery [234].
The second trial used periodontal ligament progenitors for bone regeneration in patients with periodontitis. The results revealed the regeneration of periodontal tissue, and the clinical evaluations demonstrated decreased tooth movement, a decrease in probing depth and attachment level, an increase in gingival resection, and a gain in attachment. In addition, no patients had inflammation or other complications, suggesting that the use of this cell source was safe [235]. In 2014, Shiehzadeh and collaborators used SHED and SCAPs with an injectable poly(lactide-coglycolide)-polyethylene glycol scaffold delivery system in the treatment of large periapical lesions. After a period of 4 months, both sources regenerated the apices of the root and the adjacent bone tissue from treated patients safely and without complications, confirming that these two sources are safe for use in cell therapy [239].
DPSCs are the most commonly used source in regenerative treatments. In addition to being used after the extraction of third molars [234,246,259], they have been applied to increase the floor of the maxillary sinus [236], to regenerate bone defects caused by periodontitis [237,245,247], osteoradionecrosis [238], and intrabony defects [240], mandibular bone defects caused by ameloblastomas [242] and irreversible pulpitis [243,251,258], and to regenerate teeth compromised by periodontal disease [248]. In the majority of these trials, bone regeneration was observed, the probing depth was decreased, and gingival resection was improved. In cases of pulpitis, the pulp tissue was regenerated and was functional. Finally, in the case of bone deformations due to ameloblastoma, a highly invasive and recurrent odontogenic tumor, in addition to successful bone regeneration, there was no recurrence of the tumor after DPSC transplantation.
PDLSCs have mainly been used in the treatment of bone defects caused by periodontitis [241,250,253] and have similar results to those observed with DPSCs. SHED are the second most widely used source of D-MSCs. They have been used to treat apical lesions [244,252], pulp necrosis [249], and malformations present in the palate and cleft lip [254]. These cells can generate bone tissue and functional pulp tissue. On the other hand, SHED have been used in patients suffering from type 2 diabetes mellitus, and although they cause some negative symptoms, such as fever, they are not serious; moreover, they improve glucose metabolism and the function of the pancreatic islets, suggesting that their use is safe and effective [256].
A Brazilian company evaluated the safety and tolerability of an intravenous injection of allogenic cell therapy manufactured from human SHED (Nestacell HD, Cellavita, Brazil) in patients with Huntington’s disease. Preliminary findings on the efficacy of this approach have also been assessed, providing initial evidence of allogenic SHED-based cell therapy in the treatment of this debilitating neurological disorder. A case report derived for this clinical trial evaluated whether SHED could migrate to tumors. These cells were administered intravenously to a patient with a lung nodule that was later confirmed to be an adenocarcinoma. The results showed that these cells did not graft (homing) at this point, so they did not generate conditions that favored the growth of the tumor, suggesting that these cells can be used as a treatment for other conditions even when tumors are present [257]. Finally, SHED-CMs have been used to treat erectile dysfunction, and the results showed that they improved erections in treated patients without causing adverse effects, suggesting that the use of these CMs is also safe and effective [255].
In addition to these clinical trials, three protocols have been reported for their dissemination and use in the clinic. The authors proposed the use of DPSCs as a treatment for COVID-19 [260] and cerebrovascular accidents [261,262]. All of these reports indicate that D-MSCs are safe and effective in clinical use; however, it is necessary to increase the understanding and use of these cells in other clinical scenarios that also urgently require effective treatments, such as immunological diseases. Although the immunomodulatory mechanisms of these cells have not yet been fully discerned, preclinical studies have shown promising results.
For example, cell therapy research for treating oral mucosal disorders such as oral submucous fibrosis, chronic oral ulcers, and mucositis due to chemo- or radiation therapy is limited to animal models. These conditions contain an immunological/inflammatory component, and the present-day treatment modalities consist of steroids and antioxidants, which provide only short-term and symptomatic relief and leave the patient with a certain amount of morbidity. Therefore, human clinical trials on oral mucosal disorders and D-MSCs’ immunological mechanisms are urgently required.
Some important points regarding the existent clinical trials are that only one tooth must be extracted to expand and thus obtain the number of cells needed for the treatments, and autologous samples can be used even when patients have inflammatory conditions, such as periodontitis, since the cells do not lose their proliferative and regenerative properties; these are negative points that occur in BM-MSCs [236,246,250,258,261].
One point that has not been considered relevant in clinical trials but that is convenient to emphasize is the effect of age on D-MSCs. It is known that the biological properties of BM-MSCs and other sources decrease when they are obtained from older donors [263,264,265]. In the case of DPSCs, it has been observed that cells from young and elderly donors have similar inhibitory properties on Th 1 and 2 lymphocytes and the secretion of IFN-γ and IL-4, however other characteristics are modified. For example, DPSCs derived from older adults promote the differentiation of Tregs and induce the secretion of TGF-β while DPSCs from young adults promote the differentiation of Th17 and the secretion of IL-6, IL-17a, and HGF [266]. Similar results have been reported in GMSCs. MSCs from elderly donors decrease the proliferation of MNCs and the secretion of inflammatory cytokines in a similar way to MSCs from young donors, concluding that the immunomodulatory potential is not affected by age; however, other biological properties such as differentiation capacity may be negatively altered by this parameter [267].
On the other hand, PDLSCs from elderly people do considerably decrease their biological properties, including their immunomodulatory activity [268]. In DFSCs, it has been observed in vitro that the senescence of these cells decreases their biological properties such as proliferation [269]; although it is not known whether obtaining them from elderly donors reduces the characteristics of this population, it has been observed that the total tissue suffers alterations in its metabolic pathways and has a greater risk of inflammatory infiltrate in older donors [270,271], which suggests the possibility that DPSCs could lose their anti-inflammatory potential. For this reason, it is important to further investigate the age-related effects of different sources of D-MSCs to determine which source is the most suitable for use in these clinical trials with minimal risks.

7. D-MSCs in Registered Clinical Trials

Currently, at least twenty-three clinical trials are registered on ClinicalTrials.gov (https://clinicaltrials.gov/; the URL was accessed on 28 December 2023), and three more are registered on the International Clinical Trials Registry Platform (ICTRP, https://trialsearch.who.int/; the URL was accessed on 28 December 2023) without reporting results. These protocols were registered between 2010 and 2023 (Table 4).
The predominant sources of D-MSCs are DPSCs, mentioned in thirteen trials: four treatments investigated GMSCs, six investigated SHED, two investigated PDLSCs, and one investigated MSCs obtained from oral mucosa, and one trial described the use of SCAPs. Several of these trials are in phase 1 or phase 1–2, with cells being administered via scaffolds or intravenously. Some trials aimed to administer different concentrations of cells to determine the maximum number of cells that patients could tolerate. These doses ranged from 1 × 106 to 3 × 108, and one or more doses were applied. Sometimes, depending on the protocol, several new parameters needed to be evaluated, such as the levels of inflammatory cytokines, but this depended largely on the protocol to which the use of these D-MSCs was directed. Patients were followed up for between 3 months and 1 year after the transplant.
Most of these protocols focus on dental applications, such as periodontitis, bone defects, gingival defects, and pulp necrosis. However, different research groups have aimed to evaluate their effects on other conditions, such as depression, osteoarthritis, Huntington’s disease, liver cirrhosis, and COVID-19. Although D-MSCs have proven to be safe and effective in clinical treatments carried out to date, it is crucial to continue studying their capabilities in regenerative therapy both at a clinical and preclinical level, establishing more strict and specific criteria in order to reduce potential risks and allow us to control them. However, it is necessary to begin developing more clinical protocols that focus on the other properties of these cells, such as anti-inflammatory potential. To date, preclinical studies have demonstrated the efficacy and safety of the use of these cells in some models of immunological disease, which will possibly begin to venture into the development of protocols against these diseases in humans in the near future.
The advantage of using D-MSCs in clinical trials is that tissue sources, such as the human dental pulp, gingiva, or alveolar bone, are tissues usually discarded in routine dental procedures and can be obtained without significant ethical concerns. GMSCs are also the most favorable source derived from dental tissues because of their high proliferation potential and plasticity; they are easily accessible, expandable in vitro, and exhibit immunomodulatory properties, making them effective in stem cell-based therapy. However, the analysis presented here reveals that DPSCs are the most commonly used MSCs derived from dental tissue in preclinical and clinical trials, including bone, vascular, and neural regeneration applications. There is limited information comparing the immunomodulatory properties of D-MSCs with other sources, and it is evident that further research is necessary to gain a deeper understanding of their properties and potential uses in modulating the immune response.
The heterogeneity of dental tissues has restricted the clinical application of D-MSCs; however, this complication can be addressed by controlling the relevant surface markers and epigenetic modifications. Also, a comprehensive comparative profile of D-MSCs is necessary to improve clinical decisions. Moreover, an ongoing limitation highlighted by most reviewed clinical trials is that many injectable cells are required for D-MSCs cell-based therapies, and the technical simplification of procedures is necessary to improve clinical feasibility.

8. Conclusions

The findings from the publications analyzed in this study suggest that BM-MSCs, AT-MSCs, and D-MSCs have similar properties in terms of proliferation, migration, and the ability to differentiate into three distinct types of cells: osteoblasts, chondrocytes, and adipocytes. However, BM-MSC and D-MSC are the most suitable sources for regenerating mineralized tissues in the oral region.
Clinical trials have shown that treatments with D-MSCs are effective. In addition to continuing to study their clinical safety for tissue regeneration, it is important to study other relevant aspects, such as their immunomodulatory properties, which are fundamental for the development of alternative treatments for autoimmune diseases and other inflammatory conditions. In addition, multiple biophysical cues determine the functions and biology of D-MSCs. For example, D-MSCs cultivated on certain matrices or scaffolds display characteristics that are absent in their monolayer counterparts. Hence, one way to enhance their therapeutic potential is to implant specialized constructs comprising D-MSC-laden scaffolds or hydrogels. In this regard, the use of preclinical information on this topic is encouraging. However, further exploration of the mechanisms involved in this biological property is still necessary. An important and little-explored aspect of MSCs derived from the oral cavity is the effect of an inflammatory environment on their immunomodulatory function, which is relevant for their possible clinical applications.
Another important limitation is the lack of knowledge we have about other factors that can modify the properties of these cells, such as age, which has been observed to affect the biological properties of some sources more prominently than others. These drawbacks lead us to a third limitation of the use of D-MSCs. Are there complications from using allogeneic D-MSCs samples? Although it is known that MSCs have characteristics that grant them certain immunoprivileges and therefore the risks of using allogeneic samples instead of autologous samples are considerably reduced, this must be studied in D-MSCs in greater depth to determine that the risks of using allogeneic samples are minimal.

Author Contributions

Literature research and writing, L.I.P.-P.; writing—original draft preparation, review, and editing, M.E.C.-M., P.G.-A. and G.R.F.-O.; funding acquisition, literature research, conceptualization, writing—original draft preparation, review, and editing, J.J.M. All authors have read and agreed to the published version of the manuscript.

Funding

This article constitutes a partial fulfillment of the Postgraduate in Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico. Luis Ignacio Poblano-Pérez acknowledges the fellowship and financial support provided by the Consejo Nacional de Humanidades Ciencias y Tecnologías (CONAHCYT) (number 601731) for the training received during their studies. Grant sponsor: We are indebted to IMSS (R-2023-3602-013) for the support provided to J.J.M.M and to UNAM (PAPIIT-IN221423) for the support provided to M.E.C.-M.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Luis Ignacio Poblano-Pérez is a doctoral student from the Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), and received CONAHCYT fellowship 601731.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Friedenstein, A.J.; Chailakhjan, R.K.; Lalykina, K.S. The Development of Fibroblast Colonies in Monolayer Cultures of Guinea-Pig Bone Marrow and Spleen Cells. Cell Prolif. 1970, 3, 393–403. [Google Scholar] [CrossRef] [PubMed]
  2. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  3. Gao, G.; Fan, C.; Li, W.; Liang, R.; Wei, C.; Chen, X.; Yang, Y.; Zhong, Y.; Shao, Y.; Kong, Y.; et al. Mesenchymal Stem Cells: Ideal Seeds for Treating Diseases. Hum. Cell 2021, 34, 1585–1600. [Google Scholar] [CrossRef] [PubMed]
  4. Tyndall, A. Successes and Failures of Stem Cell Transplantation in Autoimmune Diseases. Hematol. Am. Soc. Hematol. Educ. Program. 2011, 2011, 280–284. [Google Scholar] [CrossRef] [PubMed]
  5. Haddad, R.; Saldanha-Araujo, F. Mechanisms of T-Cell Immunosuppression by Mesenchymal Stromal Cells: What Do We Know So Far? Biomed. Res. Int. 2014, 2014, 216806. [Google Scholar] [CrossRef]
  6. Siegel, G.; Kluba, T.; Hermanutz-Klein, U.; Bieback, K.; Northoff, H.; Schäfer, R. Phenotype, Donor Age and Gender Affect Function of Human Bone Marrow-Derived Mesenchymal Stromal Cells. BMC Med. 2013, 11, 146. [Google Scholar] [CrossRef]
  7. Ganguly, P.; El-Jawhari, J.J.; Burska, A.N.; Ponchel, F.; Giannoudis, P.V.; Jones, E.A. The Analysis of In Vivo Aging in Human Bone Marrow Mesenchymal Stromal Cells Using Colony-Forming Unit-Fibroblast Assay and the CD45(low) CD271(+) Phenotype. Stem Cells Int. 2019, 2019, 5197983. [Google Scholar] [CrossRef]
  8. Tokalov, S.V.; Gruner, S.; Schindler, S.; Wolf, G.; Baumann, M.; Abolmaali, N. Age-Related Changes in the Frequency of Mesenchymal Stem Cells in the Bone Marrow of Rats. Stem Cells Dev. 2007, 16, 439–446. [Google Scholar] [CrossRef] [PubMed]
  9. Li, J.; Wong, W.H.; Chan, S.; Chim, J.C.; Cheung, K.M.; Lee, T.L.; Au, W.Y.; Ha, S.Y.; Lie, A.K.; Lau, Y.L.; et al. Factors Affecting Mesenchymal Stromal Cells Yield from Bone Marrow Aspiration. Chin. J. Cancer Res. 2011, 23, 43–48. [Google Scholar] [CrossRef] [PubMed]
  10. Selle, M.; Koch, J.D.; Ongsiek, A.; Ulbrich, L.; Ye, W.; Jiang, Z.; Krettek, C.; Neunaber, C.; Noack, S. Influence of Age on Stem cells Depends on the Sex of the Bone Marrow Donor. J. Cell. Mol. Med. 2022, 26, 1594–1605. [Google Scholar] [CrossRef] [PubMed]
  11. Katsara, O.; Mahaira, L.G.; Iliopoulou, E.G.; Moustaki, A.; Antsaklis, A.; Loutradis, D.; Stefanidis, K.; Baxevanis, C.N.; Papamichail, M.; Perez, S.A. Effects of Donor Age, Gender, and In Vitro Cellular Aging on the Phenotypic, Functional, and Molecular Characteristics of Mouse Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Dev. 2011, 20, 1549–1561. [Google Scholar] [CrossRef] [PubMed]
  12. Bonab, M.M.; Alimoghaddam, K.; Talebian, F.; Ghaffari, S.H.; Ghavamzadeh, A.; Nikbin, B. Aging of Mesenchymal Stem Cell In Vitro. BMC Cell Biol. 2006, 7, 14. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, Y.-H.K.; Ogando, C.R.; Wang See, C.; Chang, T.-Y.; Barabino, G.A. Changes in Phenotype and Differentiation Potential of Human Mesenchymal Stem Cells Aging In Vitro. Stem Cell Res. Ther. 2018, 9, 131. [Google Scholar] [CrossRef] [PubMed]
  14. Lamas, J.R.; Fernandez-Gutierrez, B.; Mucientes, A.; Marco, F.; Lopiz, Y.; Jover, J.A.; Abasolo, L.; Rodriguez-Rodriguez, L. RNA Sequencing of Mesenchymal Stem Cells Reveals a Blocking of Differentiation and Immunomodulatory Activities Under Inflammatory Conditions in Rheumatoid Arthritis Patients. Arthritis Res. Ther. 2019, 21, 112. [Google Scholar] [CrossRef] [PubMed]
  15. Sibov, T.T.; Severino, P.; Marti, L.C.; Pavon, L.F.; Oliveira, D.M.; Tobo, P.R.; Campos, A.H.; Paes, A.T.; Amaro, E.; Gamarra, L.F.; et al. Mesenchymal Stem Cells from Umbilical Cord Blood: Parameters for Isolation, Characterization and Adipogenic Differentiation. Cytotechnology 2012, 64, 511–521. [Google Scholar] [CrossRef]
  16. Araújo, A.B.; Furlan, J.M.; Salton, G.D.; Schmalfuss, T.; Röhsig, L.M.; Silla, L.M.R.; Passos, E.P.; Paz, A.H. Isolation of Human Mesenchymal Stem Cells from Amnion, Chorion, Placental Decidua and Umbilical Cord: Comparison of Four Enzymatic Protocols. Biotechnol. Lett. 2018, 40, 989–998. [Google Scholar] [CrossRef]
  17. Zuk, P.A.; Zhu, M.; Ashjian, P.; Ugarte, D.A.D.; Huang, J.I.; Mizuno, H.; Alfonso, Z.C.; Fraser, J.K.; Benhaim, P.; Hedrick, M.H. Human Adipose Tissue Is a Source of Multipotent Stem Cells. Mol. Biol. Cell 2002, 13, 4279–4295. [Google Scholar] [CrossRef]
  18. Mizuno, M.; Katano, H.; Mabuchi, Y.; Ogata, Y.; Ichinose, S.; Fujii, S.; Otabe, K.; Komori, K.; Ozeki, N.; Koga, H.; et al. Specific Markers and Properties of Synovial Mesenchymal Stem Cells in the Surface, Stromal, and Perivascular Regions. Stem Cell Res. Ther. 2018, 9, 123. [Google Scholar] [CrossRef]
  19. Castro-Manrreza, M.E.; Bonifaz, L.; Castro-Escamilla, O.; Monroy-Garcia, A.; Cortes-Morales, A.; Hernandez-Estevez, E.; Hernandez-Cristino, J.; Mayani, H.; Montesinos, J.J. Mesenchymal Stromal Cells from the Epidermis and Dermis of Psoriasis Patients: Morphology, Immunophenotype, Differentiation Patterns, and Regulation of T Cell Proliferation. Stem Cells Int. 2019, 2019, 4541797. [Google Scholar] [CrossRef] [PubMed]
  20. Sabatini, F.; Petecchia, L.; Tavian, M.; Jodon de Villeroche, V.; Rossi, G.A.; Brouty-Boye, D. Human Bronchial Fibroblasts Exhibit a Mesenchymal Stem Cell Phenotype and Multilineage Differentiating Potentialities. Lab. Investig. 2005, 85, 962–971. [Google Scholar] [CrossRef] [PubMed]
  21. Najimi, M.; Khuu, D.N.; Lysy, P.A.; Jazouli, N.; Abarca, J.; Sempoux, C.; Sokal, E.M. Adult-Derived Human Liver Mesenchymal-Like Cells as a Potential Progenitor Reservoir of Hepatocytes? Cell Transplant. 2007, 16, 717–728. [Google Scholar] [CrossRef] [PubMed]
  22. Ouryazdanpanah, N.; Dabiri, S.; Derakhshani, A.; Vahidi, R.; Farsinejad, A. Peripheral Blood-Derived Mesenchymal Stem Cells: Growth Factor-Free Isolation, Molecular Characterization and Differentiation. Iran. J. Pathol. 2018, 13, 461–466. [Google Scholar] [PubMed]
  23. Lin, W.; Xu, L.; Lin, S.; Shi, L.; Wang, B.; Pan, Q.; Lee, W.Y.W.; Li, G. Characterisation of Multipotent Stem Cells from Human Peripheral Blood Using an Improved Protocol. J. Orthop. Transl. 2019, 19, 18–28. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, Y.R.; Yan, X.; Yuan, F.Z.; Ye, J.; Xu, B.B.; Zhou, Z.X.; Mao, Z.M.; Guan, J.; Song, Y.F.; Sun, Z.W.; et al. The Use of Peripheral Blood-Derived Stem Cells for Cartilage Repair and Regeneration In Vivo: A Review. Front. Pharmacol. 2020, 11, 404. [Google Scholar] [CrossRef] [PubMed]
  25. Chong, P.P.; Selvaratnam, L.; Abbas, A.A.; Kamarul, T. Human Peripheral Blood Derived Mesenchymal Stem Cells Demonstrate Similar Characteristics and Chondrogenic Differentiation Potential to Bone Marrow Derived Mesenchymal Ctem Cells. J. Orthop. Res. 2012, 30, 634–642. [Google Scholar] [CrossRef] [PubMed]
  26. Li, B.; Ouchi, T.; Cao, Y.; Zhao, Z.; Men, Y. Dental-Derived Mesenchymal Stem Cells: State of the Art. Front. Cell Dev. Biol. 2021, 9, 654559. [Google Scholar] [CrossRef] [PubMed]
  27. Morris, A.L.; Tadi, P. Anatomy, Head and Neck, Teeth. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2022. [Google Scholar]
  28. Hovorakova, M.; Lesot, H.; Peterka, M.; Peterkova, R. Early Development of the Human Dentition Revisited. J. Anat. 2018, 233, 135–145. [Google Scholar] [CrossRef]
  29. Bartlett, J.D. Dental Enamel Development: Proteinases and Their Enamel Matrix Substrates. ISRN Dent. 2013, 2013, 684607. [Google Scholar] [CrossRef]
  30. Samiei, M.; Alipour, M.; Khezri, K.; Saadat, Y.R.; Forouhandeh, H.; Abdolahinia, E.D.; Vahed, S.Z.; Sharifi, S.; Dizaj, S.M. Application of Collagen and Mesenchymal Stem Cells in Regenerative Dentistry. Curr. Stem Cell Res. Ther. 2022, 17, 606–620. [Google Scholar] [CrossRef]
  31. Goldberg, M.; Kulkarni, A.B.; Young, M.; Boskey, A. Dentin: Structure, Composition and Mineralization. Front. Biosci. 2011, 3, 711–735. [Google Scholar] [CrossRef]
  32. Ghannam, M.; Alameddine, H.; Bordoni, B. Anatomy, Head and Neck, Pulp (Tooth). In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2023. [Google Scholar]
  33. Olaru, M.; Sachelarie, L.; Calin, G. Hard Dental Tissues Regeneration-Approaches and Challenges. Materials 2021, 14, 2558. [Google Scholar] [CrossRef]
  34. Cabana-Munoz, M.E.; Pelaz Fernandez, M.J.; Parmigiani-Cabana, J.M.; Parmigiani-Izquierdo, J.M.; Merino, J.J. Adult Mesenchymal Stem Cells from Oral Cavity and Surrounding Areas: Types and Biomedical Applications. Pharmaceutics 2023, 15, 2109. [Google Scholar] [CrossRef]
  35. Torabi, S.; Soni, A. Histology, Periodontium. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2022. [Google Scholar]
  36. Koller, A.; Sapra, A. Anatomy, Head and Neck, Oral Gingiva. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2022. [Google Scholar]
  37. Agha-Hosseini, F.; Jahani, M.A.; Jahani, M.; Mirzaii-Dizgah, I.; Ali-Moghaddam, K. In Vitro Isolation of Stem Cells Derived from Human Dental Pulp. Clin. Transplant. 2010, 24, E23–E28. [Google Scholar] [CrossRef]
  38. 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]
  39. Navabazam, A.R.; Sadeghian Nodoshan, F.; Sheikhha, M.H.; Miresmaeili, S.M.; Soleimani, M.; Fesahat, F. Characterization of Mesenchymal Stem Cells from Human Dental Pulp, Preapical Follicle and Periodontal Ligament. Iran. J. Reprod. Med. 2013, 11, 235–242. [Google Scholar]
  40. Seo, B.-M.; Miura, M.; Gronthos, S.; Mark Bartold, P.; Batouli, S.; Brahim, J.; Young, M.; Gehron Robey, P.; Wang, C.Y.; Shi, S. Investigation of Multipotent Postnatal Stem Cells from Human Periodontal Ligament. Lancet 2004, 364, 149–155. [Google Scholar] [CrossRef]
  41. Mitrano, T.I.; Grob, M.S.; Carrion, F.; Nova-Lamperti, E.; Luz, P.A.; Fierro, F.S.; Quintero, A.; Chaparro, A.; Sanz, A. Culture and Characterization of Mesenchymal Stem Cells from Human Gingival Tissue. J. Periodontol. 2010, 81, 917–925. [Google Scholar] [CrossRef] [PubMed]
  42. 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] [PubMed]
  43. Sonoyama, W.; Liu, Y.; Yamaza, T.; Tuan, R.S.; Wang, S.; Shi, S.; Huang, G.T. Characterization of the Apical papilla and Its Residing Stem Cells from Human Immature Permanent Teeth: A Pilot Study. J. Endod. 2008, 34, 166–171. [Google Scholar] [CrossRef] [PubMed]
  44. Sequeira, D.B.; Oliveira, A.R.; Seabra, C.M.; Palma, P.J.; Ramos, C.; Figueiredo, M.H.; Santos, A.C.; Cardoso, A.L.; Peca, J.; Santos, J.M. Regeneration of Pulp-Dentin Complex Using Human Stem Cells of the Apical Papilla: In Vivo Interaction with Two Bioactive Materials. Clin. Oral Investig. 2021, 25, 5317–5329. [Google Scholar] [CrossRef] [PubMed]
  45. Handa, K.; Saito, M.; Tsunoda, A.; Yamauchi, M.; Hattori, S.; Sato, S.; Toyoda, M.; Teranaka, T.; Narayanan, A.S. Progenitor Cells from Dental Follicle Are Able to Form Cementum Matrix In Vivo. Connect. Tissue Res. 2002, 43, 406–408. [Google Scholar] [CrossRef]
  46. Qu, G.; Li, Y.; Chen, L.; Chen, Q.; Zou, D.; Yang, C.; Zhou, Q. Comparison of Osteogenic Differentiation Potential of Human Dental-Derived Stem Cells Isolated from Dental Pulp, Periodontal Ligament, Dental Follicle, and Alveolar Bone. Stem Cells Int. 2021, 2021, 6631905. [Google Scholar] [CrossRef]
  47. 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]
  48. Nakajima, K.; Kunimatsu, R.; Ando, K.; Ando, T.; Hayashi, Y.; Kihara, T.; Hiraki, T.; Tsuka, Y.; Abe, T.; Kaku, M.; et al. Comparison of the Bone Regeneration Ability Between Stem Cells from Human Exfoliated Deciduous Teeth, Human Dental Pulp Stem Cells and Human Bone Marrow Mesenchymal Stem Cells. Biophys. Res. Commun. 2018, 497, 876–882. [Google Scholar] [CrossRef]
  49. Sharpe, P.T. Dental Mesenchymal Stem Cells. Development 2016, 143, 2273. [Google Scholar] [CrossRef]
  50. Banavar, S.R.; Rawal, S.Y.; Paterson, I.C.; Singh, G.; Davamani, F.; Khoo, S.P.; Tan, E.L. Establishing a Technique for Isolation and Characterization of Human Periodontal Ligament Derived Mesenchymal Stem Cells. Saudi Dent. J. 2021, 33, 693–701. [Google Scholar] [CrossRef]
  51. 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, 997–1002. [Google Scholar] [CrossRef] [PubMed]
  52. Raoof, M.; Yaghoobi, M.M.; Derakhshani, A.; Kamal-Abadi, A.M.; Ebrahimi, B.; Abbasnejad, M.; Shokouhinejad, N. A Modified Efficient Method for Dental Pulp Stem Cell Isolation. Dent. Res. J. 2014, 11, 244–250. [Google Scholar]
  53. Zhang, X.; Zeng, D.; Huang, F.; Wang, J. A Protocol for Isolation and Culture of Mesenchymal stem Cells from human Gingival Tissue. Am. J. Clin. Exp. Immunol. 2019, 8, 21–26. [Google Scholar]
  54. Remy, M.; Ferraro, F.; Le Salver, P.; Rey, S.; Genot, E.; Djavaheri-Mergny, M.; Thebaud, 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] [PubMed]
  55. Morsczeck, C. Dental Follicle Stem Cells. In Stem Cell Biology and Tissue Engineering in Dental Sciences; Elsevier: Amsterdam, The Netherlands, 2015; pp. 271–277. [Google Scholar]
  56. Hernandez-Monjaraz, B.; Santiago-Osorio, E.; Monroy-Garcia, A.; Ledesma-Martinez, E.; Mendoza-Nunez, V.M. Mesenchymal Stem Cells of Dental Origin for Inducing Tissue Regeneration in Periodontitis: A Mini-Review. Int. J. Mol. Sci. 2018, 19, 944. [Google Scholar] [CrossRef]
  57. Ledesma-Martinez, E.; Mendoza-Nunez, V.M.; Santiago-Osorio, E. Mesenchymal Stem Cells Derived from Dental Pulp: A Review. Stem Cells Int. 2016, 2016, 4709572. [Google Scholar] [CrossRef] [PubMed]
  58. Dave, J.R.; Tomar, G.B. Dental Tissue–Derived Mesenchymal Stem Cells: Applications in Tissue Engineering. Crit. Rev. Biomed. Eng. 2018, 46, 429–468. [Google Scholar] [CrossRef] [PubMed]
  59. Ni, X.; Xia, Y.; Zhou, S.; Peng, H.; Wu, X.; Lu, H.; Wang, H.; Liu, R.; Blazar, B.R.; Gu, J.; et al. Reduction in Murine Acute GVHD Severity by Human Gingival Tissue-Derived Mesenchymal Stem Cells Via the CD39 Pathways. Cell Death Dis. 2019, 10, 13. [Google Scholar] [CrossRef] [PubMed]
  60. De la Rosa-Ruiz, M.d.P.; Álvarez-Pérez, M.A.; Cortés-Morales, V.A.; Monroy-García, A.; Mayani, H.; Fragoso-González, G.; Caballero-Chacón, S.; Diaz, D.; Candanedo-González, F.; Montesinos, J.J. Mesenchymal Stem/Stromal Cells Derived from Dental Tissues: A Comparative In Vitro Evaluation of Their Immunoregulatory Properties Against T cells. Cells 2019, 8, 1491. [Google Scholar] [CrossRef] [PubMed]
  61. Fracaro, L.; Senegaglia, A.C.; Herai, R.H.; Leitolis, A.; Boldrini-Leite, L.M.; Rebelatto, C.L.K.; Travers, P.J.; Brofman, P.R.S.; Correa, A. The Expression Profile of Dental Pulp-Derived Stromal Cells Supports Their Limited Capacity to Differentiate into Adipogenic Cells. Int. J. Mol. Sci. 2020, 21, 2753. [Google Scholar] [CrossRef]
  62. Deng, C.; Sun, Y.; Liu, H.; Wang, W.; Wang, J.; Zhang, F. Selective Adipogenic Differentiation of Human Periodontal Ligament Stem Cells Stimulated with High Doses of Glucose. PLoS ONE 2018, 13, e0199603. [Google Scholar] [CrossRef]
  63. Du, L.; Yang, P.; Ge, S. Isolation and Characterization of Human Gingiva-Derived Mesenchymal Stem Cells Using Limiting Dilution Method. J. Dent. Sci. 2016, 11, 304–314. [Google Scholar] [CrossRef]
  64. Dong, R.; Yao, R.; Du, J.; Wang, S.; Fan, Z. Depletion of Histone Demethylase KDM2A Enhanced the Adipogenic and Chondrogenic Differentiation Potentials of Stem Cells from Apical Papilla. Exp. Cell Res. 2013, 319, 2874–2882. [Google Scholar] [CrossRef]
  65. Koutsoumparis, A.E.; Patsiarika, A.; Tsingotjidou, A.; Pappas, I.; Tsiftsoglou, A.S. Neural Differentiation of Human Dental Mesenchymal Stem Cells Induced by ATRA and UDP-4: A Comparative Study. Biomolecules 2022, 12, 218. [Google Scholar] [CrossRef]
  66. Gao, Y.; Tian, Z.; Liu, Q.; Wang, T.; Ban, L.K.; Lee, H.H.; Umezawa, A.; Almansour, A.I.; Arumugam, N.; Kumar, R.S.; et al. Neuronal Cell Differentiation of Human Dental Pulp Stem Cells on Synthetic Polymeric Surfaces Coated with ECM Proteins. Front. Cell Dev. Biol. 2022, 10, 893241. [Google Scholar] [CrossRef] [PubMed]
  67. Rao, S.R.; Subbarayan, R.; Dinesh, M.G.; Arumugam, G.; Raja, S.T. Differentiation of Human Gingival Mesenchymal Stem Cells into Neuronal Lineages in 3D Bioconjugated Injectable Protein Hydrogel Construct for the Management of Neuronal Disorder. Exp. Mol. Med. 2016, 48, e209. [Google Scholar] [CrossRef] [PubMed]
  68. Wu, T.; Xu, W.; Chen, H.; Li, S.; Dou, R.; Shen, H.; Liu, X.; Liu, X.; Hong, Y.; He, J. Comparison of the Differentiation of Dental Pulp Stem Cells and Periodontal Ligament Stem Cells into Neuron-Like Cells and Their Effects on Focal Cerebral Ischemia. Acta Biochim. Biophys. Sin. 2020, 52, 1016–1029. [Google Scholar] [CrossRef] [PubMed]
  69. Li, N.; Dai, X.; Yang, F.; Sun, Y.; Wu, X.; Zhou, Q.; Chen, K.; Sun, J.; Bi, W.; Shi, L.; et al. Spontaneous Spheroids from Alveolar Bone-Derived Mesenchymal Stromal Cells Maintain Pluripotency of Stem Cells by Regulating Hypoxia-Inducible Factors. Biol. Res. 2023, 56, 17. [Google Scholar] [CrossRef] [PubMed]
  70. Yan, X.; Yan, F.; Mohammed, H.A.G.; Liu, O. Maxillofacial-Derived Mesenchymal Stem Cells: Characteristics and Progress in Tissue Regeneration. Stem Cells Int. 2021, 2021, 5516521. [Google Scholar] [CrossRef] [PubMed]
  71. Zhao, J.; Zhou, Y.H.; Zhao, Y.Q.; Gao, Z.R.; Ouyang, Z.Y.; Ye, Q.; Liu, Q.; Chen, Y.; Tan, L.; Zhang, S.H.; et al. Oral Cavity-Derived Stem Cells and Preclinical Models of Jaw-Bone Defects for Bone Tissue Engineering. Stem Cell Res. Ther. 2023, 14, 39. [Google Scholar] [CrossRef] [PubMed]
  72. Mosaddad, S.A.; Rasoolzade, B.; Namanloo, R.A.; Azarpira, N.; Dortaj, H. Stem Cells and Common Biomaterials in Dentistry: A Review Study. J. Mater. Sci. Mater. Med. 2022, 33, 55. [Google Scholar] [CrossRef]
  73. Zhai, Q.; Dong, Z.; Wang, W.; Li, B.; Jin, Y. Dental Stem Cell and Dental Tissue Regeneration. Front. Med. 2019, 13, 152–159. [Google Scholar] [CrossRef]
  74. 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]
  75. Aydin, S.; Sahin, F. Stem Cells Derived from Dental Tissues. Adv. Exp. Med. Biol. 2019, 1144, 123–132. [Google Scholar] [CrossRef]
  76. Santilli, F.; Fabrizi, J.; Santacroce, C.; Caissutti, D.; Spinello, Z.; Candelise, N.; Lancia, L.; Pulcini, F.; Delle Monache, S.; Mattei, V. Analogies and Differences between Dental Stem Cells: Focus on Secretome in Combination with Scaffolds in Neurological Disorders. Stem Cell Rev. Rep. 2024, 20, 159–174. [Google Scholar] [CrossRef] [PubMed]
  77. Smojver, I.; Katalinic, I.; Bjelica, R.; Gabric, D.; Matisic, V.; Molnar, V.; Primorac, D. Mesenchymal Stem Cells Based Treatment in Dental Medicine: A Narrative Review. Int. J. Mol. Sci. 2022, 23, 1662. [Google Scholar] [CrossRef] [PubMed]
  78. Gan, L.; Liu, Y.; Cui, D.; Pan, Y.; Zheng, L.; Wan, M. Dental Tissue-Derived Human Mesenchymal Stem Cells and Their Potential in Therapeutic Application. Stem Cells Int. 2020, 2020, 8864572. [Google Scholar] [CrossRef] [PubMed]
  79. Newman, R.E.; Yoo, D.; LeRoux, M.A.; Danilkovitch-Miagkova, A. Treatment of Inflammatory Diseases with Mesenchymal Stem Cells. Inflamm. Allergy Drug Targets 2009, 8, 110–123. [Google Scholar] [CrossRef] [PubMed]
  80. Li, L.; Jiang, J. Regulatory Factors of Mesenchymal Stem Cell Migration into Injured Tissues and Their Signal Transduction Mechanisms. Front. Med. 2011, 5, 33–39. [Google Scholar] [CrossRef] [PubMed]
  81. Fu, X.; Liu, G.; Halim, A.; Ju, Y.; Luo, Q.; Song, A.G. Mesenchymal Stem Cell Migration and Tissue Repair. Cells 2019, 8, 784. [Google Scholar] [CrossRef]
  82. Maeda, A. Recruitment of Mesenchymal Stem Cells to Damaged Sites by Plant-Derived Components. Front. Cell Dev. Biol. 2020, 8, 437. [Google Scholar] [CrossRef]
  83. Sohni, A.; Verfaillie, C.M. Mesenchymal Stem Cells Migration Homing and Tracking. Stem Cells Int. 2013, 2013, 130763. [Google Scholar] [CrossRef]
  84. Hass, R.; Kasper, C.; Bohm, S.; Jacobs, R. Different Populations and Sources of Human Mesenchymal Atem Cells (MSC): A Comparison of Adult and Neonatal Tissue-Derived MSC. Cell Commun. Signal 2011, 9, 1–14. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, J.; Ren, H.; Yuan, X.; Ma, H.; Shi, X.; Ding, Y. Interleukin-10 Secreted by Mesenchymal Stem Cells Attenuates Acute Liver Failure through Inhibiting Pyroptosis. Hepatol. Res. 2018, 48, E194–E202. [Google Scholar] [CrossRef] [PubMed]
  86. Nakajima, M.; Nito, C.; Sowa, K.; Suda, S.; Nishiyama, Y.; Nakamura-Takahashi, A.; Nitahara-Kasahara, Y.; Imagawa, K.; Hirato, T.; Ueda, M.; et al. Mesenchymal Stem Cells Overexpressing Interleukin-10 Promote Neuroprotection in Experimental Acute Ischemic Stroke. Mol. Ther. Methods Clin. Dev. 2017, 6, 102–111. [Google Scholar] [CrossRef]
  87. Zhang, C.; Delawary, M.; Huang, P.; Korchak, J.A.; Suda, K.; Zubair, A.C. IL-10 mRNA Engineered MSCs Demonstrate Enhanced Anti-Inflammation in an Acute GvHD Model. Cells 2021, 10, 3101. [Google Scholar] [CrossRef] [PubMed]
  88. Xiao, S.; Huang, G.; Wei, Z.; Nie, K.; Liu, Z.; Deng, C.; Wang, D. IL-10 Gene-Modified Human Amniotic Mesenchymal Stem Cells Augment Regenerative Wound Healing by Multiple Synergistic Effects. Stem Cells Int. 2019, 2019, 9158016. [Google Scholar] [CrossRef] [PubMed]
  89. Dorronsoro, A.; Lang, V.; Ferrin, I.; Fernandez-Rueda, J.; Zabaleta, L.; Perez-Ruiz, E.; Sepulveda, P.; Trigueros, C. Intracellular Role of IL-6 in Mesenchymal Stromal Cell Immunosuppression and Proliferation. Sci. Rep. 2020, 10, 21853. [Google Scholar] [CrossRef] [PubMed]
  90. Huang, P.; Zhang, C.; Delawary, M.; Korchak, J.A.; Suda, K.; Zubair, A.C. Development and Evaluation of IL-6 Overexpressing Mesenchymal Stem Cells (MSCs). J. Tissue Eng. Regen. Med. 2022, 16, 244–253. [Google Scholar] [CrossRef] [PubMed]
  91. Niu, J.; Yue, W.; Le-Le, Z.; Bin, L.; Hu, X. Mesenchymal Stem Cells Inhibit T Cell Activation by Releasing TGF-β1 from TGF-β1/GARP Complex. Oncotarget 2017, 8, 99784–99800. [Google Scholar] [CrossRef]
  92. Lynch, K.; Treacy, O.; Chen, X.; Murphy, N.; Lohan, P.; Islam, M.N.; Donohoe, E.; Griffin, M.D.; Watson, L.; McLoughlin, S.; et al. TGF-beta1-Licensed Murine MSCs Show Superior Therapeutic Efficacy in Modulating Corneal Allograft Immune Rejection In Vivo. Mol. Ther. 2020, 28, 2023–2043. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, F.; Qiu, H.; Xue, M.; Zhang, S.; Zhang, X.; Xu, J.; Chen, J.; Yang, Y.; Xie, J. MSC-Secreted TGF-beta Regulates Lipopolysaccharide-Stimulated Macrophage M2-Like Polarization Via the Akt/FoxO1 Pathway. Stem Cell Res. Ther. 2019, 10, 345. [Google Scholar] [CrossRef]
  94. Yanez, R.; Oviedo, A.; Aldea, M.; Bueren, J.A.; Lamana, M.L. Prostaglandin E2 Plays a Key Role in the Immunosuppressive Properties of Adipose and Bone Marrow Tissue-Derived Mesenchymal Stromal Cells. Exp. Cell Res. 2010, 316, 3109–3123. [Google Scholar] [CrossRef]
  95. Qi, J.; Tang, X.; Li, W.; Chen, W.; Yao, G.; Sun, L. Mesenchymal Stem Cells Inhibited the Differentiation of MDSCs Via COX2/PGE2 in Experimental Sialadenitis. Stem Cell Res. Ther. 2020, 11, 325. [Google Scholar] [CrossRef]
  96. Sun, X.; Su, Y.; Liu, X.; Liu, F.; Zhang, G.; Chen, Q.; Wang, C.; Fu, H.; Zhu, X.; Liu, K.; et al. PGE2 Dependent Inhibition of Macrophage Pyroptosis by MSCs Contributes to Alleviating aGVHD. Blood 2020, 136, 15. [Google Scholar] [CrossRef]
  97. Li, Q.; Song, W.J.; Ryu, M.O.; Nam, A.; An, J.H.; Ahn, J.O.; Bhang, D.H.; Jung, Y.C.; Youn, H.Y. TSG-6 Secreted by Human Adipose Tissue-Derived Mesenchymal Stem Cells Ameliorates Severe Acute Pancreatitis Via ER Stress Downregulation in Mice. Stem Cell Res. Ther. 2018, 9, 255. [Google Scholar] [CrossRef] [PubMed]
  98. Zhao, Y.; Zhu, X.Y.; Song, T.; Zhang, L.; Eirin, A.; Conley, S.; Tang, H.; Saadiq, I.; Jordan, K.; Lerman, A.; et al. Mesenchymal Stem Cells protect Renal Tubular Cells Via TSG-6 Regulating Macrophage Sunction and Phenotype Switching. Am. J. Physiol. Ren. Physiol. 2021, 320, F454–F463. [Google Scholar] [CrossRef] [PubMed]
  99. Selmani, Z.; Naji, A.; Zidi, I.; Favier, B.; Gaiffe, E.; Obert, L.; Borg, C.; Saas, P.; Tiberghien, P.; Rouas-Freiss, N.; et al. Human Leukocyte Antigen-G5 Secretion by Human Mesenchymal Stem Cells is Required to Suppress T Lymphocyte and Natural Killer Function and to Induce CD4+CD25highFOXP3+ Tegulatory T Cells. Stem Cells 2008, 26, 212–222. [Google Scholar] [CrossRef]
  100. Ding, D.C.; Chou, H.L.; Chang, Y.H.; Hung, W.T.; Liu, H.W.; Chu, T.Y. Characterization of HLA-G and Related Immunosuppressive Effects in Human Umbilical Cord Stroma-Derived Stem Cells. Cell Transplant. 2016, 25, 217–228. [Google Scholar] [CrossRef]
  101. Jiang, W.; Xu, J. Immune Modulation by Mesenchymal Stem Cells. Cell Prolif. 2020, 53, e12712. [Google Scholar] [CrossRef]
  102. Li, W.; Ren, G.; Huang, Y.; Su, J.; Han, Y.; Li, J.; Chen, X.; Cao, K.; Chen, Q.; Shou, P.; et al. Mesenchymal Stem Cells: A Double-Edged Sword in Regulating Immune Responses. Cell Death Differ. 2012, 19, 1505–1513. [Google Scholar] [CrossRef] [PubMed]
  103. Maria, A.T.J.; Rozier, P.; Fonteneau, G.; Sutra, T.; Maumus, M.; Toupet, K.; Cristol, J.P.; Jorgensen, C.; Guilpain, P.; Noel, D. iNOS Activity Is Required for the Therapeutic Effect of Mesenchymal Stem Cells in Experimental Systemic Sclerosis. Front. Immunol. 2018, 9, 3056. [Google Scholar] [CrossRef]
  104. Tedgui, A.; Mallat, Z. Anti-Inflammatory Mechanisms in the Vascular Wall. Circ. Res. 2001, 88, 877–887. [Google Scholar] [CrossRef]
  105. Routy, J.P.; Routy, B.; Graziani, G.M.; Mehraj, V. The Kynurenine Pathway Is a Double-Edged Sword in Immune-Privileged Sites and in Cancer: Implications for Immunotherapy. Int. J. Tryptophan Res. 2016, 9, 67–77. [Google Scholar] [CrossRef]
  106. Meesuk, L.; Tantrawatpan, C.; Kheolamai, P.; Manochantr, S. The Immunosuppressive Capacity of Human Mesenchymal Stromal Cells Derived from Amnion and Bone Marrow. Biochem. Biophys. Rep. 2016, 8, 34–40. [Google Scholar] [CrossRef]
  107. Torres Crigna, A.; Uhlig, S.; Elvers-Hornung, S.; Kluter, H.; Bieback, K. Human Adipose Tissue-Derived Stromal Cells Suppress Human, but Not Murine Lymphocyte Proliferation, via Indoleamine 2,3-Dioxygenase Activity. Cells 2020, 9, 2419. [Google Scholar] [CrossRef] [PubMed]
  108. Kerkelä, E.; Laitinen, A.; Rabina, J.; Valkonen, S.; Takatalo, M.; Larjo, A.; Veijola, J.; Lampinen, M.; Siljander, P.; Lehenkari, P.; et al. Adenosinergic Immunosuppression by Human Mesenchymal Stromal Cells Requires Co-Operation with T Cells. Stem Cells 2016, 34, 781–790. [Google Scholar] [CrossRef] [PubMed]
  109. Saldanha-Araujo, F.; Ferreira, F.I.; Palma, P.V.; Araujo, A.G.; Queiroz, R.H.; Covas, D.T.; Zago, M.A.; Panepucci, R.A. Mesenchymal Stromal Cells Up-Regulate CD39 and Increase Adenosine Production to Suppress Activated T-Lymphocytes. Stem Cell Res. 2011, 7, 66–74. [Google Scholar] [CrossRef]
  110. Zhang, X.; Huang, F.; Li, W.; Dang, J.L.; Yuan, J.; Wang, J.; Zeng, D.L.; Sun, C.X.; Liu, Y.Y.; Ao, Q.; et al. Human Gingiva-Derived Mesenchymal Stem Cells Modulate Monocytes/Macrophages and Alleviate Atherosclerosis. Front. Immunol. 2018, 9, 878. [Google Scholar] [CrossRef]
  111. 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] [PubMed]
  112. Antonioli, L.; Fornai, M.; Blandizzi, C.; Haskó, G. Adenosine Regulation of the Immune System. In The Adenosine Receptors; Borea, P.A., Varani, K., Gessi, S., Merighi, S., Vincenzi, F., Eds.; Springer International Publishing: Cham, Switzerland, 2018; pp. 499–514. [Google Scholar]
  113. Davies, L.C.; Heldring, N.; Kadri, N.; Le Blanc, K. Mesenchymal Stromal Cell Secretion of Programmed Death-1 Ligands Regulates T Cell Mediated Immunosuppression. Stem Cells 2017, 35, 766–776. [Google Scholar] [CrossRef] [PubMed]
  114. Di Tinco, R.; Bertani, G.; Pisciotta, A.; Bertoni, L.; Pignatti, E.; Maccaferri, M.; Bertacchini, J.; Sena, P.; Vallarola, A.; Tupler, R.; et al. Role of PD-L1 in Licensing Immunoregulatory Function of Dental Pulp Mesenchymal Stem Cells. Stem Cell Res. Ther. 2021, 12, 598. [Google Scholar] [CrossRef]
  115. Gaber, T.; Schonbeck, K.; Hoff, H.; Tran, C.L.; Strehl, C.; Lang, A.; Ohrndorf, S.; Pfeiffenberger, M.; Rohner, E.; Matziolis, G.; et al. CTLA-4 Mediates Inhibitory Function of Mesenchymal Stem/Stromal Cells. Int. J. Mol. Sci. 2018, 19, 2312. [Google Scholar] [CrossRef]
  116. Montesinos, J.J.; López-García, L.; Cortés-Morales, V.A.; Arriaga-Pizano, L.; Valle-Ríos, R.; Fajardo-Orduña, G.R.; Castro-Manrreza, M.E. Human Bone Marrow Mesenchymal Stem/Stromal Cells Exposed to an Inflammatory Environment Increase the Expression of ICAM-1 and Release Microvesicles Enriched in This Adhesive Molecule: Analysis of the Participation of TNF-α and IFN-γ. J. Immunol. Res. 2020, 2020, 8839625. [Google Scholar] [CrossRef]
  117. Lopez-Garcia, L.; Castro-Manrreza, M.E. TNF-alpha and IFN-gamma Participate in Improving the Immunoregulatory Capacity of Mesenchymal Stem/Stromal Cells: Importance of Cell-Cell Contact and Extracellular Vesicles. Int. J. Mol. Sci. 2021, 22, 9531. [Google Scholar] [CrossRef]
  118. Mattar, P.; Bieback, K. Comparing the Immunomodulatory Properties of Bone Marrow, Adipose Tissue, and Birth-Associated Tissue Mesenchymal Stromal Cells. Front. Immunol. 2015, 6, 560. [Google Scholar] [CrossRef] [PubMed]
  119. Song, N.; Scholtemeijer, M.; Shah, K. Mesenchymal Stem Cell Immunomodulation: Mechanisms and Therapeutic Potential. Trends Pharmacol. Sci. 2020, 41, 653–664. [Google Scholar] [CrossRef]
  120. Andrukhov, O.; Behm, C.; 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] [PubMed]
  121. Ryan, J.M.; Barry, F.; Murphy, J.M.; Mahon, B.P. Interferon-gamma Does not Break, but Promotes the Immunosuppressive Capacity of Adult Human Mesenchymal Stem Cells. Clin. Exp. Immunol. 2007, 149, 353–363. [Google Scholar] [CrossRef]
  122. Lee, R.H.; Yu, J.M.; Foskett, A.M.; Peltier, G.; Reneau, J.C.; Bazhanov, N.; Oh, J.Y.; Prockop, D.J. TSG-6 as a Biomarker to Predict Efficacy of Human Mesenchymal Stem/Progenitor Cells (hMSCs) in Modulating Sterile Inflammation In Vivo. Proc. Natl. Acad. Sci. USA 2014, 111, 16766–16771. [Google Scholar] [CrossRef]
  123. Kim, S.N.; Lee, H.J.; Jeon, M.S.; Yi, T.; Song, S.U. Galectin-9 is Involved in Immunosuppression Mediated by Human Bone Marrow-derived Clonal Mesenchymal Stem Cells. Immune Netw. 2015, 15, 241–251. [Google Scholar] [CrossRef]
  124. Sioud, M.; Mobergslien, A.; Boudabous, A.; Floisand, Y. Mesenchymal Stem Cell-Mediated T Cell Suppression Occurs Through Secreted Galectins. Int. J. Oncol. 2011, 38, 385–390. [Google Scholar] [CrossRef]
  125. Giri, J.; Das, R.; Nylen, E.; Chinnadurai, R.; Galipeau, J. CCL2 and CXCL12 Derived from Mesenchymal Stromal Cells Cooperatively Polarize IL-10+ Tissue Macrophages to Mitigate Gut Injury. Cell Rep. 2020, 30, 1923–1934. [Google Scholar] [CrossRef]
  126. Yagura, K.; Ohtaki, H.; Tsumuraya, T.; Sato, A.; Miyamoto, K.; Kawada, N.; Suzuki, K.; Nakamura, M.; Kanzaki, K.; Dohi, K.; et al. The Enhancement of CCL2 and CCL5 by Human Bone Marrow-Derived Mesenchymal Stem/Stromal Cells Might Contribute to Inflammatory Suppression and Axonal Extension After Spinal Cord Injury. PLoS ONE 2020, 15, e0230080. [Google Scholar] [CrossRef] [PubMed]
  127. Xie, Z.; Tang, J.; Chen, Z.; Wei, L.; Chen, J.; Liu, Q. Human Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles Reduce Inflammation and Pyroptosis in Acute Kidney Injury Via miR-223-3p/HDAC2/SNRK. Inflamm. Res. 2023, 72, 553–576. [Google Scholar] [CrossRef]
  128. Dabrowska, S.; Andrzejewska, A.; Strzemecki, D.; Muraca, M.; Janowski, M.; Lukomska, B. Human Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Neuroinflammation Evoked by Focal Brain Injury in Rats. J. Neuroinflamm. 2019, 16, 216. [Google Scholar] [CrossRef]
  129. Castro-Manrreza, M.E.; Mayani, H.; Monroy-Garcia, A.; Flores-Figueroa, E.; Chavez-Rueda, K.; Legorreta-Haquet, V.; Santiago-Osorio, E.; Montesinos, J.J. Human Mesenchymal Stromal Cells from Adult and Neonatal Sources: A Comparative In Vitro Analysis of Their Immunosuppressive Properties Against T Cells. Stem Cells Dev. 2014, 23, 1217–1232. [Google Scholar] [CrossRef]
  130. Stubbendorff, M.; Deuse, T.; Hua, X.; Phan, T.T.; Bieback, K.; Atkinson, K.; Eiermann, T.H.; Velden, J.; Schroder, C.; Reichenspurner, H.; et al. Immunological Properties of Extraembryonic Human Mesenchymal Stromal Cells Derived from Gestational Tissue. Stem Cells Dev. 2013, 22, 2619–2629. [Google Scholar] [CrossRef]
  131. Avanzini, M.A.; Bernardo, M.E.; Cometa, A.M.; Perotti, C.; Zaffaroni, N.; Novara, F.; Visai, L.; Moretta, A.; Del Fante, C.; Villa, R.; et al. Generation of Mesenchymal Stromal Cells in the Presence of Platelet Lysate: A Phenotypic and Functional Comparison of Umbilical Cord Blood- and Bone Marrow-Derived Progenitors. Haematologica 2009, 94, 1649–1660. [Google Scholar] [CrossRef] [PubMed]
  132. Seo, Y.; Ahn, J.S.; Shin, Y.Y.; Oh, S.J.; Song, M.H.; Kang, M.J.; Oh, J.M.; Lee, D.; Kim, Y.H.; Lee, B.C.; et al. Mesenchymal Stem Cells Target Microglia Via Galectin-1 Production to Rescue Aged Mice from Olfactory Dysfunction. Biomed. Pharmacother. 2022, 153, 113347. [Google Scholar] [CrossRef] [PubMed]
  133. Kang, J.W.; Kang, K.S.; Koo, H.C.; Park, J.R.; Choi, E.W.; Park, Y.H. Soluble Factors-Mediated Immunomodulatory Effects of Canine Adipose Tissue-Derived Mesenchymal Stem Cells. Stem Cells Dev. 2008, 17, 681–693. [Google Scholar] [CrossRef] [PubMed]
  134. Roemeling-van Rhijn, M.; Mensah, F.K.; Korevaar, S.S.; Leijs, M.J.; van Osch, G.J.; Ijzermans, J.N.; Betjes, M.G.; Baan, C.C.; Weimar, W.; Hoogduijn, M.J. Effects of Hypoxia on the Immunomodulatory Properties of Adipose Tissue-Derived Mesenchymal Stem Cells. Front. Immunol. 2013, 4, 203. [Google Scholar] [CrossRef] [PubMed]
  135. Sineh Sepehr, K.; Razavi, A.; Hassan, Z.M.; Fazel, A.; Abdollahpour-Alitappeh, M.; Mossahebi-Mohammadi, M.; Yekaninejad, M.S.; Farhadihosseinabadi, B.; Hashemi, S.M. Comparative Immunomodulatory Properties of Mesenchymal Stem Cells Derived from Human Breast Tumor and Normal Breast Adipose Tissue. Cancer Immunol. Immunother. 2020, 69, 1841–1854. [Google Scholar] [CrossRef] [PubMed]
  136. de Castro, L.L.; Xisto, D.G.; Kitoko, J.Z.; Cruz, F.F.; Olsen, P.C.; Redondo, P.A.G.; Ferreira, T.P.T.; Weiss, D.J.; Martins, M.A.; Morales, M.M.; et al. Human Adipose Tissue Mesenchymal Stromal Cells and Their Extracellular Vesicles Act Differentially on Lung Mechanics and Inflammation in Experimental Allergic Asthma. Stem Cell Res. Ther. 2017, 8, 151. [Google Scholar] [CrossRef]
  137. Heo, J.S.; Kim, S. Human Adipose Mesenchymal Stem Cells Modulate Inflammation and Angiogenesis Through Exosomes. Sci. Rep. 2022, 12, 2776. [Google Scholar] [CrossRef]
  138. Tomic, S.; Djokic, J.; Vasilijic, S.; Vucevic, D.; Todorovic, V.; Supic, G.; Colic, M. Immunomodulatory Properties of Mesenchymal Stem Cells Derived from Dental Pulp and Dental Follicle are Susceptible to Activation by Toll-Like Receptor Agonists. Stem Cells Dev. 2011, 20, 695–708. [Google Scholar] [CrossRef]
  139. 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]
  140. Ji, L.; Bao, L.; Gu, Z.; Zhou, Q.; Liang, Y.; Zheng, Y.; Xu, Y.; Zhang, X.; Feng, X. Comparison of Immunomodulatory Properties of Exosomes Derived from Bone Marrow Mesenchymal Stem Cells and Dental Pulp Stem Cells. Immunol. Res. 2019, 67, 432–442. [Google Scholar] [CrossRef] [PubMed]
  141. Martinez, V.G.; Ontoria-Oviedo, I.; Ricardo, C.P.; Harding, S.E.; Sacedon, R.; Varas, A.; Zapata, A.; Sepulveda, P.; Vicente, A. Overexpression of Hypoxia-Inducible Factor 1 Alpha Improves Immunomodulation by Dental Mesenchymal Stem Cells. Stem Cell Res. Ther. 2017, 8, 208. [Google Scholar] [CrossRef] [PubMed]
  142. Zheng, J.; Kong, Y.; Hu, X.; Li, Z.; Li, Y.; Zhong, Y.; Wei, X.; Ling, J. MicroRNA-Enriched Small Extracellular Vesicles Possess Odonto-Immunomodulatory Properties for Modulating the Immune Response of Macrophages and Promoting Odontogenesis. Stem Cell Res. Ther. 2020, 11, 517. [Google Scholar] [CrossRef]
  143. Kanji, S.; Sarkar, R.; Pramanik, A.; Kshirsagar, S.; Greene, C.J.; Das, H. Dental Pulp-Derived Stem Cells Inhibit Osteoclast Differentiation by Secreting Osteoprotegerin and Deactivating AKT Signalling in Myeloid Cells. J. Cell Mol. Med. 2021, 25, 2390–2403. [Google Scholar] [CrossRef]
  144. Ahmadi, P.; Yan, M.; Bauche, A.; Smeets, R.; Muller, C.E.; Koch-Nolte, F.; Haag, F.; Fliegert, R.; Kluwe, L.; Schulze Zur Wiesch, J.; et al. Human Dental Pulp Cells Modulate CD8(+) T Cell Proliferation and Efficiently Degrade Extracellular ATP to Adenosine In Vitro. Cell Immunol. 2022, 380, 104589. [Google Scholar] [CrossRef]
  145. Wada, N.; Menicanin, D.; Shi, S.; Bartold, P.M.; Gronthos, S. Immunomodulatory Properties of Human Periodontal Ligament Stem Cells. J. Cell Physiol. 2009, 219, 667–676. [Google Scholar] [CrossRef] [PubMed]
  146. 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]
  147. Arora, P.; Li, W.; Huang, X.; Yu, W.; Huang, R.; Jiang, Q.; Chen, C. Metabolic Reconfiguration Activates Stemness and Immunomodulation of PDLSCs. Int. J. Mol. Sci. 2022, 23, 4038. [Google Scholar] [CrossRef] [PubMed]
  148. Kim, J.H.; Jo, C.H.; Kim, H.R.; Hwang, Y.I. Comparison of Immunological Characteristics of Mesenchymal Stem Cells from the Periodontal Ligament, Umbilical Cord, and Adipose Tissue. Stem Cells Int. 2018, 2018, 8429042. [Google Scholar] [CrossRef] [PubMed]
  149. Misawa, M.Y.O.; Silverio Ruiz, K.G.; Nociti, F.H., Jr.; Albiero, M.L.; Saito, M.T.; Nobrega Stipp, R.; Condino-Neto, A.; Holzhausen, M.; Palombo, H.; Villar, C.C. Periodontal Ligament-Derived Mesenchymal Stem Cells Modulate Neutrophil Responses Via Paracrine Mechanisms. J. Periodontol. 2019, 90, 747–755. [Google Scholar] [CrossRef] [PubMed]
  150. 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] [PubMed]
  151. Sun, J.; Wang, Z.; Liu, P.; Hu, Y.; Li, T.; Yang, J.; Gao, P.; Xu, Q. Exosomes Derived from Human Gingival Mesenchymal Stem Cells Attenuate the Inflammatory Response in Periodontal Ligament Stem Cells. Front. Chem. 2022, 10, 863364. [Google Scholar] [CrossRef] [PubMed]
  152. Chen, M.; Su, W.; Lin, X.; Guo, Z.; Wang, J.; Zhang, Q.; Brand, D.; Ryffel, B.; Huang, J.; Liu, Z.; et al. Adoptive Transfer of Human Gingiva-Derived Mesenchymal Stem Cells Ameliorates Collagen-Induced Arthritis via Suppression of Th1 and Th17 Cells and Enhancement of Regulatory T Cell Differentiation. Arthritis Rheum. 2013, 65, 1181–1193. [Google Scholar] [CrossRef]
  153. Zhang, W.; Zhou, L.; Dang, J.; Zhang, X.; Wang, J.; Chen, Y.; Liang, J.; Li, D.; Ma, J.; Yuan, J.; et al. Human Gingiva-Derived Mesenchymal Stem Cells Ameliorate Streptozoticin-induced T1DM in Mice via Suppression of T effector cells and Up-regulating Treg Subsets. Sci. Rep. 2017, 7, 15249. [Google Scholar] [CrossRef] [PubMed]
  154. Huang, F.; Chen, M.; Chen, W.; Gu, J.; Yuan, J.; Xue, Y.; Dang, J.; Su, W.; Wang, J.; Zadeh, H.H.; et al. Human Gingiva-Derived Mesenchymal Stem Cells Inhibit Xeno-Graft-Versus-Host Disease Via CD39–CD73–Adenosine and IDO Signals. Front. Immunol. 2017, 8, 68. [Google Scholar] [CrossRef]
  155. Dang, J.; Xu, Z.; Xu, A.; Liu, Y.; Fu, Q.; Wang, J.; Huang, F.; Zheng, Y.; Qi, G.; Sun, B.; et al. Human Gingiva-Derived Mesenchymal Stem Cells are Therapeutic in Lupus Nephritis Through Targeting of CD39() CD73 Signaling Pathway. J. Autoimmun. 2020, 113, 102491. [Google Scholar] [CrossRef]
  156. 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]
  157. Kou, X.; Xu, X.; Chen, C.; Sanmillan, M.L.; Cai, T.; Zhou, Y.; Giraudo, C.; Le, A.; Shi, S. The Fas/Fap-1/Cav-1 Complex Regulates IL-1RA Secretion in Mesenchymal Stem Cells to Accelerate Wound Healing. Sci. Transl. Med. 2018, 10, eaai8524. [Google Scholar] [CrossRef]
  158. Nakao, Y.; Fukuda, T.; Zhang, Q.; Sanui, T.; Shinjo, T.; Kou, X.; Chen, C.; Liu, D.; Watanabe, Y.; Hayashi, C.; 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. [Google Scholar] [CrossRef]
  159. 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] [PubMed]
  160. Yu, S.; Zhao, Y.; Ma, Y.; Ge, L. Profiling the Secretome of Human Stem Cells from Dental Apical Papilla. Stem Cells Dev. 2016, 25, 499–508. [Google Scholar] [CrossRef] [PubMed]
  161. 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]
  162. Lei, T.; Zhang, X.; Chen, P.; Li, Q.; Du, H. Proteomic Profile of Human Dental Follicle Stem Cells and Apical Papilla Stem Cells. J. Proteom. 2021, 231, 103928. [Google Scholar] [CrossRef]
  163. Meneses, C.C.B.; Pizzatto, L.N.; Andrade, F.F.F.; Sipert, C.R. Prostaglandin E(2) Affects Interleukin 6 and Monocyte Chemoattractant Protein 1/CCL2 Production by Cultured Stem Cells of Apical Papilla. J. Endod. 2020, 46, 413–418. [Google Scholar] [CrossRef]
  164. Genc, D.; Zibandeh, N.; Nain, E.; Gokalp, M.; Ozen, A.O.; Goker, M.K.; Akkoc, T. Dental Follicle Mesenchymal Stem Cells Down-Regulate Th2-Mediated Immune Response in Asthmatic Patients Mononuclear Cells. Clin. Exp. Allergy 2018, 48, 663–678. [Google Scholar] [CrossRef] [PubMed]
  165. Chen, X.; Yang, B.; Tian, J.; Hong, H.; Du, Y.; Li, K.; Li, X.; Wang, N.; Yu, X.; Wei, X. Dental Follicle Stem Cells Ameliorate Lipopolysaccharide-Induced Inflammation by Secreting TGF-beta3 and TSP-1 to Elicit Macrophage M2 Polarization. Cell Physiol. Biochem. 2018, 51, 2290–2308. [Google Scholar] [CrossRef]
  166. Hong, H.; Chen, X.; Li, K.; Wang, N.; Li, M.; Yang, B.; Yu, X.; Wei, X. Dental Follicle Stem Cells Rescue the Regenerative Capacity of Inflamed Rat Dental Pulp Through a Paracrine Pathway. Stem Cell Res. Ther. 2020, 11, 333. [Google Scholar] [CrossRef]
  167. Silva Fde, S.; Ramos, R.N.; de Almeida, D.C.; Bassi, E.J.; Gonzales, R.P.; Miyagi, S.P.; Maranduba, C.P.; Sant’Anna, O.A.; Marques, M.M.; Barbuto, J.A.; et al. Mesenchymal Stem Cells Derived from Human Exfoliated Deciduous Teeth (SHEDs) Induce Immune Modulatory Profile in Monocyte-Derived Dendritic Cells. PLoS ONE 2014, 9, e98050. [Google Scholar] [CrossRef]
  168. Luo, P.; Jiang, C.; Ji, P.; Wang, M.; Xu, J. Exosomes of Stem Cells from Human Exfoliated Deciduous Teeth as an Anti-Inflammatory Agent in Temporomandibular Joint Chondrocytes Via miR-100-5p/mTOR. Stem Cell Res. Ther. 2019, 10, 216. [Google Scholar] [CrossRef] [PubMed]
  169. Yang, N.; Liu, X.; Chen, X.; Yu, S.; Yang, W.; Liu, Y. Stem Cells from Exfoliated Deciduous Teeth Transplantation Ameliorates Sjögren’s Syndrome by Secreting Soluble PD-L1. J. Leukoc. Biol. 2022, 111, 1043–1055. [Google Scholar] [CrossRef]
  170. Liu, Y.; Wang, L.; Liu, S.; Liu, D.; Chen, C.; Xu, X.; Chen, X.; Shi, S. Transplantation of SHED Prevents Bone Loss in the Early Phase of Ovariectomy-Induced Osteoporosis. J. Dent. Res. 2014, 93, 1124–1132. [Google Scholar] [CrossRef] [PubMed]
  171. Cao, C.; Tarle, S.; Kaigler, D. Characterization of the Immunomodulatory Properties of Alveolar Bone-Derived Mesenchymal Stem Cells. Stem Cell Res. Ther. 2020, 11, 102. [Google Scholar] [CrossRef] [PubMed]
  172. Cao, C.; Maska, B.; Malik, M.A.; Tagett, R.; Kaigler, D. Immunomodulatory Differences Between Mesenchymal Stem Cells from Different Oral Tissues. Heliyon 2024, 10, e23317. [Google Scholar] [CrossRef]
  173. Pierdomenico, L.; Bonsi, L.; Calvitti, M.; Rondelli, D.; Arpinati, M.; Chirumbolo, G.; Becchetti, E.; Marchionni, C.; Alviano, F.; Fossati, V.; et al. Multipotent Mesenchymal Stem Cells with Immunosuppressive Activity can be Easily Isolated from Dental Pulp. Transplantation 2005, 80, 836–842. [Google Scholar] [CrossRef]
  174. Vasandan, A.B.; Shankar, S.R.; Prasad, P.; Sowmya Jahnavi, V.; Bhonde, R.R.; Jyothi Prasanna, S. Functional Differences in Mesenchymal Stromal Cells from Human Dental Pulp and Periodontal Ligament. J. Cell Mol. Med. 2014, 18, 344–354. [Google Scholar] [CrossRef]
  175. Pisciotta, A.; Bertani, G.; Bertoni, L.; Di Tinco, R.; De Biasi, S.; Vallarola, A.; Pignatti, E.; Tupler, R.; Salvarani, C.; de Pol, A.; et al. Modulation of Cell Death and Promotion of Chondrogenic Differentiation by Fas/FasL in Human Dental Pulp Stem Cells (hDPSCs). Front. Cell Dev. Biol. 2020, 8, 279. [Google Scholar] [CrossRef]
  176. 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]
  177. Croci, S.; Bonacini, M.; Dolci, G.; Massari, M.; Facciolongo, N.; Pignatti, E.; Pisciotta, A.; Carnevale, G.; Negro, A.; Cassone, G.; et al. Human Dental Pulp Stem Cells Modulate Cytokine Production In Vitro by Peripheral Blood Mononuclear Cells from Coronavirus Disease 2019 Patients. Front. Cell. Dev. Biol. 2020, 8, 609204. [Google Scholar] [CrossRef]
  178. Cui, S.J.; Zhang, T.; Fu, Y.; Liu, Y.; Gan, Y.H.; Zhou, Y.H.; Yang, R.L.; Wang, X.D. DPSCs Attenuate Experimental Progressive TMJ Arthritis by Inhibiting the STAT1 Pathway. J. Dent. Res. 2020, 99, 446–455. [Google Scholar] [CrossRef]
  179. Zhang, Z.; Ji, J.; Dong, C.; Gu, Z. miR-21 in Exosomes Drived from Dental Pulp Stem Cells Ameliorate the Tregs/Th17 Immune Response Via Targeting Stat3 in Collagen-Induced Arthritis Mice. Ann. Rheum. Dis. 2020, 79, 250. [Google Scholar] [CrossRef]
  180. Dong, X.; Kong, F.; Liu, C.; Dai, S.; Zhang, Y.; Xiao, F.; Zhang, H.; Wu, C.T.; Wang, H. Pulp Stem Cells with Hepatocyte Growth Factor Overexpression Exhibit Dual Effects in Rheumatoid Arthritis. Stem Cell Res. Ther. 2020, 11, 229. [Google Scholar] [CrossRef]
  181. Albashari, A.; He, Y.; Zhang, Y.; Ali, J.; Lin, F.; Zheng, Z.; Zhang, K.; Cao, Y.; Xu, C.; Luo, L.; et al. Thermosensitive bFGF-Modified Hydrogel with Dental Pulp Stem Cells on Neuroinflammation of Spinal Cord Injury. ACS Omega 2020, 5, 16064–16075. [Google Scholar] [CrossRef] [PubMed]
  182. 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] [PubMed]
  183. Andrukhov, O. Toll-Like Receptors and Dental Mesenchymal Stromal Cells. Front. Oral Health 2021, 2, 648901. [Google Scholar] [CrossRef] [PubMed]
  184. Li, J.; Ju, Y.; Liu, S.; Fu, Y.; Zhao, S. Exosomes Derived from Lipopolysaccharide-Preconditioned Human Dental Pulp Stem Cells Regulate Schwann Cell Migration and Differentiation. Connect. Tissue Res. 2021, 62, 277–286. [Google Scholar] [CrossRef]
  185. Alksne, M.; Kalvaityte, M.; Simoliunas, E.; Gendviliene, I.; Barasa, P.; Rinkunaite, I.; Kaupinis, A.; Seinin, D.; Rutkunas, V.; Bukelskiene, V. Dental Pulp Stem Cell-Derived Extracellular Matrix: Autologous Tool Boosting Bone Regeneration. Cytotherapy 2022, 24, 597–607. [Google Scholar] [CrossRef]
  186. Zheng, Y.; Dong, C.; Yang, J.; Jin, Y.; Zheng, W.; Zhou, Q.; Liang, Y.; Bao, L.; Feng, G.; Ji, J.; et al. Exosomal microRNA-155-5p from PDLSCs Regulated Th17/Treg Balance by Targeting Sirtuin-1 in Phronic Periodontitis. J. Cell Physiol. 2019, 234, 20662–20674. [Google Scholar] [CrossRef]
  187. Shin, C.; Kim, M.; Han, J.A.; Choi, B.; Hwang, D.; Do, Y.; Yun, J.H. 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, 135–146. [Google Scholar] [CrossRef] [PubMed]
  188. 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]
  189. 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]
  190. Li, W.; Huang, X.; Yu, W.; Xu, Y.; Huang, R.; Park, J.; Moshaverinia, A.; Arora, P.; Chen, C. Activation of Functional Somatic Stem Cells Promotes Endogenous Tissue Regeneration. J. Dent. Res. 2022, 101, 802–811. [Google Scholar] [CrossRef] [PubMed]
  191. Liu, J.; Wang, H.; Zhang, L.; Li, X.; Ding, X.; Ding, G.; Wei, F. Periodontal Ligament Stem Cells Promote Polarization of M2 Macrophages. J. Leukoc. Biol. 2022, 111, 1185–1197. [Google Scholar] [CrossRef] [PubMed]
  192. Qiu, J.; Wang, X.; Zhou, H.; Zhang, C.; Wang, Y.; Huang, J.; Liu, M.; Yang, P.; Song, A. 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, 42. [Google Scholar] [CrossRef] [PubMed]
  193. Nagata, M.; Iwasaki, K.; Akazawa, K.; Komaki, M.; Yokoyama, N.; Izumi, Y.; Morita, I. Conditioned Medium from Periodontal Ligament Stem Cells Enhances Periodontal Regeneration. Tissue Eng. Part. A 2017, 23, 367–377. [Google Scholar] [CrossRef]
  194. Yu, B.; Li, Q.; Zhou, M. LPS-Induced Upregulation of the TLR4 Signaling Pathway Inhibits Osteogenic Differentiation of Human Periodontal Ligament Stem Cells Under Inflammatory Conditions. Int. J. Mol. Med. 2019, 43, 2341–2351. [Google Scholar] [CrossRef]
  195. Wang, W.; Yuan, C.; Geng, T.; Liu, Y.; Zhu, S.; Zhang, C.; Liu, Z.; Wang, P. Lipopolysaccharide Inhibits Osteogenic Differentiation of Periodontal Ligament Stem Cells Partially Through Toll-Like Receptor 4-Mediated EphrinB2 Downregulation. Clin. Oral Investig. 2020, 24, 3407–3416. [Google Scholar] [CrossRef]
  196. Duan, Y.; An, W.; Wu, H.; Wu, Y. Salvianolic Acid C Attenuates LPS-Induced Inflammation and Apoptosis in Human Periodontal Ligament Stem Cells via Toll-Like Receptors 4 (TLR4)/Nuclear Factor kappa B (NF-kappaB) Pathway. Med. Sci. Monit. 2019, 25, 9499–9508. [Google Scholar] [CrossRef]
  197. Chen, M.; Lin, X.; Zhang, L.; Hu, X. Effects of Nuclear Factor-KappaB Signaling Pathway on Periodontal Ligament Stem Cells Under Lipopolysaccharide-Induced Inflammation. Bioengineered 2022, 13, 7951–7961. [Google Scholar] [CrossRef] [PubMed]
  198. Paganelli, A.; Trubiani, O.; Diomede, F.; Pisciotta, A.; Paganelli, R. Immunomodulating Profile of Dental Mesenchymal Stromal Cells: A Comprehensive Overview. Front. Oral Health 2021, 2, 635055. [Google Scholar] [CrossRef] [PubMed]
  199. 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] [PubMed]
  200. Ding, G.; Liu, Y.; An, Y.; Zhang, C.; Shi, S.; Wang, W.; Wang, S. Suppression of T Cell Proliferation by Root Apical Papilla Stem Cells In Vitro. Cells Tissues Organs 2010, 191, 357–364. [Google Scholar] [CrossRef] [PubMed]
  201. Smeda, M.; Galler, K.M.; Woelflick, M.; Rosendahl, A.; Moehle, C.; Lenhardt, B.; Buchalla, W.; Widbiller, M. Molecular Biological Comparison of Dental Pulp- and Apical Papilla-Derived Stem Cells. Int. J. Mol. Sci. 2022, 23, 2615. [Google Scholar] [CrossRef]
  202. Ding, G.; Wang, W.; Liu, Y.; An, Y.; Zhang, C.; Shi, S.; Wang, S. Effect of Cryopreservation on Biological and Immunological Properties of Stem Cells from Apical Papilla. J. Cell Physiol. 2010, 223, 415–422. [Google Scholar] [CrossRef]
  203. Liu, X.M.; Liu, Y.; Yu, S.; Jiang, L.M.; Song, B.; Chen, X. Potential Immunomodulatory Effects of Stem Cells from the Apical Papilla on Treg Conversion in Tissue Regeneration for Regenerative Endodontic Treatment. Int. Endod. J. 2019, 52, 1758–1767. [Google Scholar] [CrossRef]
  204. Tatic, N.; Rose, F.; des Rieux, A.; White, L.J. Stem Cells from the Dental Apical Papilla in Extracellular Matrix Hydrogels Mitigate Inflammation of Microglial Cells. Sci. Rep. 2019, 9, 14015. [Google Scholar] [CrossRef]
  205. De Berdt, P.; Bottemanne, P.; Bianco, J.; Alhouayek, M.; Diogenes, A.; Lloyd, A.; Llyod, A.; Gerardo-Nava, J.; Brook, G.A.; Miron, V.; et al. Stem Cells from Human Apical Papilla Decrease Neuro-Inflammation and Stimulate Oligodendrocyte Progenitor Differentiation Via Activin-A Secretion. Cell Mol. Life Sci. 2018, 75, 2843–2856. [Google Scholar] [CrossRef]
  206. Li, J.; Tang, Y.; Zhu, Q.; Zhang, Y.; Zhou, P.; Gu, Y. Transplantation of Human Stem Cells from the Apical Papilla for Treating Dextran Sulfate Sodium-Induced Experimental Colitis. Chin. J. Tissue Eng. Res. 2020, 24, 1069–1075. [Google Scholar] [CrossRef]
  207. Wang, A.; Liu, J.; Zhuang, X.; Yu, S.; Zhu, S.; Liu, Y.; Chen, X. Identification and Comparison of piRNA Expression Profiles of Exosomes Derived from Human Stem Cells from the Apical Papilla and Bone Marrow Mesenchymal Stem Cells. Stem Cells Dev. 2020, 29, 511–520. [Google Scholar] [CrossRef] [PubMed]
  208. Yu, S.; Chen, X.; Liu, Y.; Zhuang, X.Y.; Wang, A.C.; Liu, X.M.; Zhu, S. Exosomes Derived from Stem Cells from the Apical Papilla Alleviate Inflammation in Rat Pulpitis by Upregulating Regulatory T Cells. Int. Endod. J. 2022, 55, 517–530. [Google Scholar] [CrossRef]
  209. Bi, R.; Lyu, P.; Song, Y.; Li, P.; Song, D.; Cui, C.; Fan, Y. Function of Dental Follicle Progenitor/Stem Cells and Their Potential in Regenerative Medicine: From Mechanisms to Applications. Biomolecules 2021, 11, 997. [Google Scholar] [CrossRef] [PubMed]
  210. Genc, D.; Zibandeh, N.; Nain, E.; Arig, U.; Goker, K.; Aydiner, E.K.; Akkoc, T. IFN-gamma Stimulation of Dental Follicle Mesenchymal Stem Cells Modulates Immune Response of CD4(+) T Lymphocytes in Der p1(+) Asthmatic Patients In Vitro. Allergol. Immunopathol. 2019, 47, 467–476. [Google Scholar] [CrossRef] [PubMed]
  211. Ulusoy, C.; Zibandeh, N.; Yildirim, S.; Trakas, N.; Zisimopoulou, P.; Kucukerden, M.; Tasli, H.; Tzartos, S.; Goker, K.; Tuzun, E.; et al. Dental Follicle Mesenchymal Stem Cell Administration Ameliorates Muscle Weakness in MuSK-Immunized Mice. J. Neuroinflamm. 2015, 12, 231. [Google Scholar] [CrossRef]
  212. Guo, R.; Yu, J. Multipotency and Immunomodulatory Benefits of Stem Cells from Human Exfoliated Deciduous Teeth. Front. Dent. Med. 2022, 3, 805875. [Google Scholar] [CrossRef]
  213. 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]
  214. Gao, X.; Shen, Z.; Guan, M.; Huang, Q.; Chen, L.; Qin, W.; Ge, X.; Chen, H.; Xiao, Y.; Lin, Z. Immunomodulatory Role of Stem Cells from Human Exfoliated Deciduous Teeth on Periodontal Regeneration. Tissue Eng. Part. A 2018, 24, 1341–1353. [Google Scholar] [CrossRef]
  215. Rossato, C.; Brandao, W.N.; Castro, S.B.R.; de Almeida, D.C.; Maranduba, C.M.C.; Camara, N.O.S.; Peron, J.P.S.; Silva, F.S. Stem Cells from Human-Exfoliated Deciduous Teeth Reduce Tissue-Infiltrating Inflammatory Cells Improving Clinical Signs in Experimental Autoimmune Encephalomyelitis. Biologicals 2017, 49, 62–68. [Google Scholar] [CrossRef]
  216. Zhang, Q.; Li, Q.; Zhu, J.; Guo, H.; Zhai, Q.; Li, B.; Jin, Y.; He, X.; Jin, F. Comparison of Therapeutic Effects of Different Mesenchymal Stem Cells on Rheumatoid Arthritis in Mice. PeerJ 2019, 7, e7023. [Google Scholar] [CrossRef]
  217. Shimojima, C.; Takeuchi, H.; Jin, S.; Parajuli, B.; Hattori, H.; Suzumura, A.; Hibi, H.; Ueda, M.; Yamamoto, A. Conditioned Medium from the Stem Cells of Human Exfoliated Deciduous Teeth Ameliorates Experimental Autoimmune Encephalomyelitis. J. Immunol. 2016, 196, 4164–4171. [Google Scholar] [CrossRef] [PubMed]
  218. Muto, H.; Ito, T.; Tanaka, T.; Yokoyama, S.; Yamamoto, K.; Imai, N.; Ishizu, Y.; Maeda, K.; Honda, T.; Ishikawa, T.; et al. Conditioned Medium from Stem Cells Derived from Human Exfoliated Deciduous Teeth Ameliorates NASH Via the Gut-Liver Axis. Sci. Rep. 2021, 11, 18778. [Google Scholar] [CrossRef] [PubMed]
  219. Muhammad, S.A.; Nordin, N.; Hussin, P.; Mehat, M.Z.; Abu Kasim, N.H.; Fakurazi, S. Protective Effects of Stem Cells from Human Exfoliated Deciduous Teeth Derived Conditioned Medium on Osteoarthritic Chondrocytes. PLoS ONE 2020, 15, e0238449. [Google Scholar] [CrossRef] [PubMed]
  220. Sonoda, S.; Murata, S.; Kato, H.; Zakaria, F.; Kyumoto-Nakamura, Y.; Uehara, N.; Yamaza, H.; Kukita, T.; Yamaza, T. Targeting of Deciduous Tooth Pulp Stem Cell-Derived Extracellular Vesicles on Telomerase-Mediated Stem Cell Niche and Immune Regulation in Systemic Lupus Erythematosus. J. Immunol. 2021, 206, 3053–3063. [Google Scholar] [CrossRef] [PubMed]
  221. Wei, J.; Song, Y.; Du, Z.; Yu, F.; Zhang, Y.; Jiang, N.; Ge, X. Exosomes Derived from Human Exfoliated Deciduous Teeth Ameliorate Adult Bone Loss in Mice Through Promoting Osteogenesis. J. Mol. Histol. 2020, 51, 455–466. [Google Scholar] [CrossRef] [PubMed]
  222. Matsubara, T.; Suardita, K.; Ishii, M.; Sugiyama, M.; Igarashi, A.; Oda, R.; Nishimura, M.; Saito, M.; Nakagawa, K.; Yamanaka, K.; et al. Alveolar Bone Marrow as a Cell Source for Regenerative Medicine: Differences Between Alveolar and Iliac Bone Marrow Stromal Cells. J. Bone Miner. Res. 2005, 20, 399–409. [Google Scholar] [CrossRef] [PubMed]
  223. Mason, S.; Tarle, S.A.; Osibin, W.; Kinfu, Y.; Kaigler, D. Standardization and Safety of Alveolar Bone-Derived Stem Cell Isolation. J. Dent. Res. 2014, 93, 55–61. [Google Scholar] [CrossRef]
  224. Wang, X.; Xing, H.; Zhang, G.; Wu, X.; Zou, X.; Feng, L.; Wang, D.; Li, M.; Zhao, J.; Du, J.; et al. Restoration of a Critical Mandibular Bone Defect Using Human Alveolar Bone-Derived Stem Cells and Porous Nano-HA/Collagen/PLA Scaffold. Stem Cells Int. 2016, 2016, 8741641. [Google Scholar] [CrossRef]
  225. Wang, F.; Zhou, Y.; Zhou, J.; Xu, M.; Zheng, W.; Huang, W.; Zhou, W.; Shen, Y.; Zhao, K.; Wu, Y.; et al. Comparison of Intraoral Bone Regeneration with Iliac and Alveolar BMSCs. J. Dent. Res. 2018, 97, 1229–1235. [Google Scholar] [CrossRef]
  226. Behm, C.; Blufstein, A.; Gahn, J.; Nemec, M.; Moritz, A.; Rausch-Fan, X.; Andrukhov, O. Cytokines Differently Define the Immunomodulation of Mesenchymal Stem Cells from the Periodontal Ligament. Cells 2020, 9, 1222. [Google Scholar] [CrossRef]
  227. Behm, C.; Blufstein, A.; Gahn, J.; Kubin, B.; Moritz, A.; Rausch-Fan, X.; Andrukhov, O. Continuing Effect of Cytokines and Toll-Like Receptor Agonists on Indoleamine-2,3-Dioxygenase-1 in Human Periodontal Ligament Stem/Stromal Cells. Cells 2020, 9, 2696. [Google Scholar] [CrossRef]
  228. Andrukhov, O.; Hong, J.S.; Andrukhova, O.; Blufstein, A.; Moritz, A.; Rausch-Fan, X. Response of Human Periodontal Ligament Stem Cells to IFN-gamma and TLR-Agonists. Sci. Rep. 2017, 7, 12856. [Google Scholar] [CrossRef]
  229. Behm, C.; Blufstein, A.; Gahn, J.; Kubin, B.; Nemec, M.; Moritz, A.; Rausch-Fan, X.; Andrukhov, O. 1,25(OH)(2)D(3) Differently Affects Immunomodulatory Activities of Mesenchymal Stem Cells Depending on the Presence of TNF-alpha, IL-1beta and IFN-gamma. J. Clin. Med. 2019, 8, 2211. [Google Scholar] [CrossRef]
  230. Watanabe, Y.; Fukuda, T.; Hayashi, C.; Nakao, Y.; Toyoda, M.; Kawakami, K.; Shinjo, T.; Iwashita, M.; Yamato, H.; Yotsumoto, K.; et al. Extracellular Vesicles Derived from GMSCs Stimulated with TNF-alpha and IFN-alpha Promote M2 Macrophage Polarization Via Enhanced CD73 and CD5L Expression. Sci. Rep. 2022, 12, 13344. [Google Scholar] [CrossRef]
  231. Tian, J.; Chen, W.; Xiong, Y.; Li, Q.; Kong, S.; Li, M.; Pang, C.; Qiu, Y.; Xu, Z.; Gong, Q.; et al. Small Extracellular Vesicles Derived from Hypoxic Preconditioned Dental Pulp Stem Cells Ameliorate Inflammatory Osteolysis by Modulating Macrophage Polarization and Osteoclastogenesis. Bioact. Mater. 2023, 22, 326–342. [Google Scholar] [CrossRef]
  232. Cocce, V.; Franze, S.; Brini, A.T.; Gianni, A.B.; Pascucci, L.; Ciusani, E.; Alessandri, G.; Farronato, G.; Cavicchini, L.; Sordi, V.; et al. In Vitro Anticancer Activity of Extracellular Vesicles (EVs) Secreted by Gingival Mesenchymal Stromal Cells Primed with Paclitaxel. Pharmaceutics 2019, 11, 61. [Google Scholar] [CrossRef]
  233. Klimova, D.; Jakubechova, J.; Altanerova, U.; Nicodemou, A.; Styk, J.; Szemes, T.; Repiska, V.; Altaner, C. Extracellular Vesicles Derived from Dental Mesenchymal Stem/Stromal Cells with Gemcitabine as a Cargo have an Inhibitory Effect on the Growth of Pancreatic Carcinoma Cell Lines In Vitro. Mol. Cell Probes 2023, 67, 101894. [Google Scholar] [CrossRef] [PubMed]
  234. d’Aquino, R.; De Rosa, A.; Lanza, V.; Tirino, V.; Laino, L.; Graziano, A.; Desiderio, V.; Laino, G.; Papaccio, G. Human mandible bone Defect Pepair by the Grafting of Dental Pulp Stem/Progenitor Cells and Collagen Sponge Biocomplexes. Eur. Cell Mater. 2009, 18, 75–83. [Google Scholar] [CrossRef] [PubMed]
  235. Feng, F.; Akiyama, K.; Liu, Y.; Yamaza, T.; Wang, T.M.; Chen, J.H.; Wang, B.B.; Huang, G.T.; Wang, S.; Shi, S. Utility of PDL Progenitors for In Vivo Tissue Regeneration: A Report of 3 Cases. Oral Dis. 2010, 16, 20–28. [Google Scholar] [CrossRef] [PubMed]
  236. Brunelli, G.; Motroni, A.; Graziano, A.; D’Aquino, R.; Zollino, I.; Carinci, F. Sinus Lift Tissue Engineering Using Autologous Pulp Micro-Grafts: A Case Report of Bone Density Evaluation. J. Indian Soc. Periodontol. 2013, 17, 644–647. [Google Scholar] [CrossRef] [PubMed]
  237. Aimetti, M.; Ferrarotti, F.; Cricenti, L.; Mariani, G.M.; Romano, F. Autologous Dental Pulp Stem Cells in Periodontal Regeneration: A Case Report. Int. J. Periodontics Restor. Dent. 2014, 34 (Suppl. S3), s27–s33. [Google Scholar] [CrossRef]
  238. Manimaran, K.; Sankaranarayanan, S.; Ravi, V.R.; Elangovan, S.; Chandramohan, M.; Perumal, S.M. Treatment of Osteoradionecrosis of Mandible with Bone Marrow Concentrate and with Dental Pulp Stem Cells. Ann. Maxillofac. Surg. 2014, 4, 189–192. [Google Scholar] [CrossRef]
  239. Shiehzadeh, V.; Aghmasheh, F.; Shiehzadeh, F.; Joulae, M.; Kosarieh, E.; Shiehzadeh, F. Healing of Large Periapical Lesions Following Delivery of Dental Stem Cells With an Injectable Scaffold: New Method and Three Case Reports. Indian J. Dent. Res. 2014, 25, 248–253. [Google Scholar] [CrossRef] [PubMed]
  240. Aimetti, M.; Ferrarotti, F.; Mariani, G.M.; Cricenti, L.; Romano, F. Use of Dental Pulp Stem Cells/Collagen Sponge Biocomplex in the Treatment of Non-Contained Intrabony Defects: A Case Series. Clin. Adv. Periodontics 2015, 5, 104–109. [Google Scholar] [CrossRef] [PubMed]
  241. Chen, F.M.; Gao, L.N.; Tian, B.M.; Zhang, X.Y.; Zhang, Y.J.; Dong, G.Y.; Lu, H.; Chu, Q.; Xu, J.; Yu, Y.; et al. Treatment of Periodontal Intrabony Defects Using Autologous Periodontal Ligament Stem Cells: A Randomized Clinical Trial. Stem Cell Res. Ther. 2016, 7, 33. [Google Scholar] [CrossRef]
  242. Manimaran, K.; Sharma, R.; Sankaranarayanan, S.; Perumal, S.M. Regeneration of Mandibular Ameloblastoma Defect with the Help of Autologous Dental Pulp Stem Cells and Buccal Pad of Fat Stromal Vascular Fraction. Ann. Maxillofac. Surg. 2016, 6, 97–100. [Google Scholar] [CrossRef]
  243. Nakashima, M.; Iohara, K.; Murakami, M.; Nakamura, H.; Sato, Y.; Ariji, Y.; Matsushita, K. Pulp Regeneration by Transplantation of Dental Pulp Stem Cells in Pulpitis: A Pilot Clinical Study. Stem Cell Res. Ther. 2017, 8, 61. [Google Scholar] [CrossRef]
  244. Prasad, M.G.S.; Ramakrishna, J.; Babu, D.N. Allogeneic Stem Cells Derived from Human Exfoliated Deciduous Teeth (SHED) for the Management of Periapical Lesions in Permanent Teeth: Two Case Reports of a Novel Biologic Alternative Treatment. J. Dent. Res. Dent. Clin. Dent. Prospect. 2017, 11, 117–122. [Google Scholar] [CrossRef]
  245. Aimetti, M.; Ferrarotti, F.; Gamba, M.N.; Giraudi, M.; Romano, F. Regenerative Treatment of Periodontal Intrabony Defects Using Autologous Dental Pulp Stem Cells: A 1-Year Follow-Up Case Series. Int. J. Periodontics Restor. Dent. 2018, 38, 51–58. [Google Scholar] [CrossRef]
  246. Barbier, L.; Ramos, E.; Mendiola, J.; Rodriguez, O.; Santamaria, G.; Santamaria, J.; Arteagoitia, I. Autologous Dental Pulp Mesenchymal Stem Cells for Inferior Third Molar Post-Extraction Socket Healing: A Split-Mouth Randomised Clinical Trial. Med. Oral Patol. Oral Cir. Bucal 2018, 23, e469–e477. [Google Scholar] [CrossRef]
  247. Ferrarotti, F.; Romano, F.; Gamba, M.N.; Quirico, A.; Giraudi, M.; Audagna, M.; Aimetti, M. Human Intrabony Defect Regeneration with Micrografts Containing Dental Pulp Stem Cells: A Randomized Controlled Clinical Trial. J. Clin. Periodontol. 2018, 45, 841–850. [Google Scholar] [CrossRef]
  248. Hernandez-Monjaraz, B.; Santiago-Osorio, E.; Ledesma-Martinez, E.; Alcauter-Zavala, A.; Mendoza-Nunez, V.M. Retrieval of a Periodontally Compromised Tooth by Allogeneic Grafting of Mesenchymal Stem Cells from Dental Pulp: A Case Report. J. Int. Med. Res. 2018, 46, 2983–2993. [Google Scholar] [CrossRef] [PubMed]
  249. Xuan, K.; Li, B.; Guo, H.; Sun, W.; Kou, X.; He, X.; Zhang, Y.; Sun, J.; Liu, A.; Liao, L.; et al. Deciduous Autologous Tooth Stem Cells Regenerate Dental Pulp After Implantation into Injured Teeth. Sci. Transl. Med. 2018, 10, eaaf3227. [Google Scholar] [CrossRef]
  250. Iwata, T.; Yamato, M.; Washio, K.; Yoshida, T.; Tsumanuma, Y.; Yamada, A.; Onizuka, S.; Izumi, Y.; Ando, T.; Okano, T.; 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. [Google Scholar] [CrossRef]
  251. Meza, G.; Urrejola, D.; Saint Jean, N.; Inostroza, C.; Lopez, V.; Khoury, M.; Brizuela, C. Personalized Cell Therapy for Pulpitis Using Autologous Dental Pulp Stem Cells and Leukocyte Platelet-rich Fibrin: A Case Report. J. Endod. 2019, 45, 144–149. [Google Scholar] [CrossRef] [PubMed]
  252. Ghana Shyam Prasad, M.; Juvva, R.; Babu Duvvi, N. Towards a New Era in the Management of Large Periapical Lesion in Permanent Tooth Using Stemcells: A 2-Year Clinical Application Report. J. Dent. 2019, 20, 137–140. [Google Scholar] [CrossRef]
  253. Sanchez, N.; Fierravanti, L.; Nunez, J.; Vignoletti, F.; Gonzalez-Zamora, M.; Santamaria, S.; Suarez-Sancho, S.; Fernandez-Santos, M.E.; Figuero, E.; Herrera, D.; et al. Periodontal Regeneration Using a Xenogeneic Bone Substitute Seeded with Autologous Periodontal Ligament-Derived Mesenchymal Stem Cells: A 12-Month Quasi-Randomized Controlled Pilot Clinical Trial. J. Clin. Periodontol. 2020, 47, 1391–1402. [Google Scholar] [CrossRef] [PubMed]
  254. Tanikawa, D.Y.S.; Pinheiro, C.C.G.; Almeida, M.C.A.; Oliveira, C.; Coudry, R.A.; Rocha, D.L.; Bueno, D.F. Deciduous Dental Pulp Stem Cells for Maxillary Alveolar Reconstruction in Cleft Lip and Palate Patients. Stem Cells Int. 2020, 2020, 6234167. [Google Scholar] [CrossRef] [PubMed]
  255. Koga, S.; Horiguchi, Y. Efficacy of a Cultured Conditioned Medium of Exfoliated Deciduous Dental Pulp Stem Cells in Erectile Dysfunction Patients. J. Cell. Mol. Med. 2021, 26, 195–201. [Google Scholar] [CrossRef]
  256. Li, W.; Jiao, X.; Song, J.; Sui, B.; Guo, Z.; Zhao, Y.; Li, J.; Shi, S.; Huang, Q. Therapeutic Potential of Stem Cells from Human Exfoliated Deciduous Teeth Infusion into Patients with Type 2 Diabetes Pepends on Basal Lipid Levels and Islet Function. Stem Cells Transl. Med. 2021, 10, 956–967. [Google Scholar] [CrossRef]
  257. da Silva, J.M.; Araldi, R.P.; Colozza-Gama, G.A.; Pagani, E.; Sid, A.; Valverde, C.W.; Kerkis, I. Human Immature Dental Pulp Stem Cells Did Not Graft into a Preexisting Human Lung Adenocarcinoma. Case Rep. Oncol. 2022, 15, 413–422. [Google Scholar] [CrossRef] [PubMed]
  258. Nakashima, M.; Fukuyama, F.; Iohara, K. Pulp Regenerative Cell Therapy for Mature Molars: A Report of 2 Cases. J. Endod. 2022, 48, 1334.e1331–1340.e1331. [Google Scholar] [CrossRef] [PubMed]
  259. Cubuk, S.; Oduncuoglu, B.F.; Alaaddinoglu, E.E. The Effect of Dental Pulp Stem Cells and L-PRF when Placed into the Extraction Sockets of Impacted Mandibular Third Molars on the Periodontal Status of Adjacent Second Molars: A Split-Mouth, Randomized, Controlled Clinical Trial. Oral Maxillofac. Surg. 2023, 27, 59–68. [Google Scholar] [CrossRef] [PubMed]
  260. Ye, Q.; Wang, H.; Xia, X.; Zhou, C.; Liu, Z.; Xia, Z.E.; Zhang, Z.; Zhao, Y.; Yehenala, J.; Wang, S.; et al. Safety and Efficacy Assessment of Allogeneic Human Dental Pulp Stem Cells to Treat Patients with Severe COVID-19: Structured Summary of a Study Protocol for a Randomized Controlled Trial (Phase I/II). Trials 2020, 21, 520. [Google Scholar] [CrossRef]
  261. Nagpal, A.; Kremer, K.L.; Hamilton-Bruce, M.A.; Kaidonis, X.; Milton, A.G.; Levi, C.; Shi, S.; Carey, L.; Hillier, S.; Rose, M.; et al. TOOTH (The Open study of dental pulp stem cell Therapy in Humans): Study Protocol for Evaluating Safety and Feasibility of Autologous Human Adult Dental Pulp Stem Cell Therapy in Patients with Chronic Disability After Stroke. Int. J. Stroke 2016, 11, 575–585. [Google Scholar] [CrossRef]
  262. Suda, S.; Nito, C.; Ihara, M.; Iguchi, Y.; Urabe, T.; Matsumaru, Y.; Sakai, N.; Kimura, K. Randomised Placebo-Controlled Multicentre Trial to Evaluate the Efficacy and Safety of JTR-161, Allogeneic Human Dental Pulp Stem Cells, in Patients with Acute Ischaemic stRoke (J-REPAIR). BMJ Open 2022, 12, e054269. [Google Scholar] [CrossRef]
  263. Massaro, F.; Corrillon, F.; Stamatopoulos, B.; Dubois, N.; Ruer, A.; Meuleman, N.; Bron, D.; Lagneaux, L. Age-Related Changes in Human Bone Marrow Mesenchymal Stromal Cells: Morphology, Gene Expression Profile, Immunomodulatory Activity and miRNA Expression. Front. Immunol. 2023, 14, 1267550. [Google Scholar] [CrossRef]
  264. Zhang, Y.; Ravikumar, M.; Ling, L.; Nurcombe, V.; Cool, S.M. Age-Related Changes in the Inflammatory Status of Human Mesenchymal Stem Cells: Implications for Cell Therapy. Stem Cell Rep. 2021, 16, 694–707. [Google Scholar] [CrossRef]
  265. Mahmoud, M.; Abdel-Rasheed, M.; Galal, E.R.; El-Awady, R.R. Factors Defining Human Adipose Stem/Stromal Cell Immunomodulation in Vitro. Stem Cell Rev. Rep. 2023, 20, 175–205. [Google Scholar] [CrossRef]
  266. Ozgul Ozdemir, R.B.; Ozdemir, A.T.; Kirmaz, C.; Eker Sariboyaci, A.; Karaoz, E.; Erman, G.; Vatansever, H.S.; Mete Gokmen, N. Age-Related Changes in the Immunomodulatory Effects of Human Dental Pulp Derived Mesenchymal Stem Cells on the CD4(+) T Cell Subsets. Cytokine 2021, 138, 155367. [Google Scholar] [CrossRef]
  267. Dave, J.R.; Chandekar, S.S.; Behera, S.; Desai, K.U.; Salve, P.M.; Sapkal, N.B.; Mhaske, S.T.; Dewle, A.M.; Pokare, P.S.; Page, M.; et al. Human Gingival Mesenchymal Stem Cells Retain their Growth and Immunomodulatory Characteristics Independent of Donor Age. Sci. Adv. 2022, 8, eabm6504. [Google Scholar] [CrossRef]
  268. Li, X.; Zhang, B.; Wang, H.; Zhao, X.; Zhang, Z.; Ding, G.; Wei, F. The Effect of Aging on the Biological and Immunological Characteristics of Periodontal Ligament Stem Cells. Stem Cell Res. Ther. 2020, 11, 326. [Google Scholar] [CrossRef]
  269. Morsczeck, C. Effects of Cellular Senescence on Dental Follicle Cells. Pharmacology 2021, 106, 137–142. [Google Scholar] [CrossRef] [PubMed]
  270. de Mello Palma, V.; Danesi, C.C.; Arend, C.F.; Venturini, A.B.; Blaya, D.S.; Neto, M.M.; Flores, J.A.; Ferrazzo, K.L. Study of Pathological Changes in the Dental Follicle of Disease-Free Impacted Third Molars. J. Maxillofac. Oral Surg. 2018, 17, 611–615. [Google Scholar] [CrossRef] [PubMed]
  271. Bastos, V.C.; Vitorio, J.G.; Martins-Chaves, R.R.; Leite-Lima, F.; Lebron, Y.A.R.; Moreira, V.R.; Duarte-Andrade, F.F.; Pereira, T.; Santos, L.V.S.; Lange, L.C.; et al. Age-Related Metabolic Pathways Changes in Dental Follicles: A Pilot Study. Front. Oral Health 2021, 2, 677731. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Locations of different sources of MSCs derived from dental tissues. Abbreviations: DPSCs, dental pulp stem cells; PDLSCs, periodontal ligament stem cells; GMSCs, gingiva-derived MSCs; SCAPs, stem cells from the apical papilla; DFSCs, dental follicle stem cells; SHED, stem cells from exfoliated deciduous teeth; ABMSCs, alveolar bone-derived MSCs; TGPCs, tooth germ progenitor cells.
Figure 1. Locations of different sources of MSCs derived from dental tissues. Abbreviations: DPSCs, dental pulp stem cells; PDLSCs, periodontal ligament stem cells; GMSCs, gingiva-derived MSCs; SCAPs, stem cells from the apical papilla; DFSCs, dental follicle stem cells; SHED, stem cells from exfoliated deciduous teeth; ABMSCs, alveolar bone-derived MSCs; TGPCs, tooth germ progenitor cells.
Ijms 25 01986 g001
Figure 2. Immunomodulatory properties of MSCs derived from dental tissues. D-MSCs modulate the immune system through different mechanisms, inhibiting proliferation, differentiation, maturation, inflammatory cytokine production, and cytotoxicity. In addition, they can induce the generation of regulatory cells that promote an anti-inflammatory response. D-MSCs can be secreted through soluble factors such as IL-10, PGE2, and TGF-β, the presence of intra- and extracellular enzymes such as IDO, CD39, and CD73, which produce anti-inflammatory molecules, the expression of membrane molecules such as PDL-1, and the secretion of EVs that carry anti-inflammatory molecules.
Figure 2. Immunomodulatory properties of MSCs derived from dental tissues. D-MSCs modulate the immune system through different mechanisms, inhibiting proliferation, differentiation, maturation, inflammatory cytokine production, and cytotoxicity. In addition, they can induce the generation of regulatory cells that promote an anti-inflammatory response. D-MSCs can be secreted through soluble factors such as IL-10, PGE2, and TGF-β, the presence of intra- and extracellular enzymes such as IDO, CD39, and CD73, which produce anti-inflammatory molecules, the expression of membrane molecules such as PDL-1, and the secretion of EVs that carry anti-inflammatory molecules.
Ijms 25 01986 g002
Table 1. Characteristics of D-MSCs.
Table 1. Characteristics of D-MSCs.
SourceEfficiency of IsolationSurface MarkersEmbryonic 
Markers
Neural MarkersDifferentiation Potential
DPSCs++++CD13, CD29, CD44, CD59, CD73, CD90, CD105, CD146STRO-1, OCT-4, Nanog, SSEA-1, SEEA-4, SOX-2β3-tubulin, NFM, Nestin, CNPase, S100, CD271Adipogenic, osteogenic, odontoblast, angiogenic, and neuronal cells
PDLSCs++++CD10, CD29, CD44, CD73, CD105SSEA-1, SSEA-3, SSEA-4, TRA-1–60, TRA-1–81, OCT-4, 
Nanog, SOX-2, REX1, ALP
Nestin, OCT-4, SSEA-4, CD271, SOX-10Adipogenic, chondrogenic, osteogenic, and neuronal cells
GMSCs++++CD73, CD90, CD105SSEA-4, OCT-4, NanogNestin, SOX10, β3-tubulin, NFM, CNPaseAdipogenic, chondrogenic, osteogenic, angiogenic, and neuronal cells
SCAPs+++CD24, CD44, CD90, CD146, STRO-1OCT-4, Nanog, NOTCH-1, SOX-2OCT-4, SOX2, NestinAdipogenic, chondrogenic osteogenic, odontogenic, and neuronal cells
DFSCs++CD13, CD29, CD44, CD56, CD59, CD90, CD105, CD106, CD166, STRO-1OCT-4, Nanog, NOTCH-1, SOX-2OCT-4, SOX2, NestinOsteogenic, odontogenic, and cementogenic
SHED+++CD29, CD73, CD90, CD146, CD166OCT-4, Nanog, SSEA-3, SSEA-4, NOTCH-1, SOX-2β3-tubulin, NFM, Nestin, CNPase, GAD, NeuN, GFAP, CD271, Vimentin, OCT-4, PAX-6, NSE, MAP-2, PSA- NCAMAdipogenic, chondrogenic, osteogenic, odontogenic, angiogenic, and neuronal cells
ABMSCs++++CD73, CD90, CD105, STRO-1Oct4, KLF4, Sox2, cMycNF-M, NeuN, GFAPAdipogenic, chondrogenic, and osteogenic
TGPCs+CD29, CD73, CD90, CD105, CD166Nanog, OCT-4, SOX-2, Klf4, c-MycNestinAdipogenic, chondrogenic, osteogenic, and neuronal cells
Reviewed in [69,70,71,72,73,74,75,76,77,78].
Table 2. Molecules involved in the immunoregulation of MSCs from different sources.
Table 2. Molecules involved in the immunoregulation of MSCs from different sources.
SourceMolecules and Mechanisms Related to Immunomodulatory EffectsReferences
BM-MSCsIDO, TGF-β1, HGF, IL-6, IL-10, HLA-G, PGE2, NO, ICAM-1, PDL-1 and 2, TSG-6, galectins, EVs[86,89,99,113,116,121,122,123,124,125,126,127,128]
UCB-MSCsIDO, TGF-β, HGF, HLA-G, PDL-1, PGE2, galectins[129,130,131,132]
AT-MSCsTGF-β, HGF, PGE2, IDO, PDL-1, IL-10, TSG-6, EVs[97,98,133,134,135,136,137]
DPSCsIL-6, TGF-β, PGE2, IDO, HLAG, HGF, HIF-1, PDL-1, Fas-FasL pathway, osteoprotegerin, EVs, ADO[60,114,138,139,140,141,142,143,144]
PDLSCsTGF-β, IDO, HGF, PGE2, RANTES, eotaxina, IFN-γ, induced protein 10, MCP-1, IL-6, IL-8, IL-1ra EVs, PD-1, PDL-1 and 2[145,146,147,148,149]
GMSCsFas-FasL pathway, PGE2, IDO, iNOS, IL-10, adenosinergic pathway (CD39, CD73, ADO), EVs[42,59,110,111,150,151,152,153,154,155,156,157,158,159]
SCAPsIL-6, IL-10, PD-1, PDL-1 y 2 TGF-β 1 y 2, galectin 1, PGE2, EVs[160,161,162,163]
DFSCsTGF-β, IDO, TGF-β3, thrombospondin 1, IL-4[164,165,166]
SHEDTGF-β, IL-10, PDL-1, EVs, Fas-FasL pathway[167,168,169,170]
ABMSCsIL-6, MCP-1, PGE2, TIMP-1 and 2, osteoprotegerin[171,172]
TGPCsNot described-
Table 3. Published clinical trials carried out with D-MSCs.
Table 3. Published clinical trials carried out with D-MSCs.
DisorderMSCs SourceAdministration RouteOutcomesReference
Mandibular bone defects after the extraction of third molarsAutologous DPSCsImplanted at the extraction sites in a collagen spongeIncreased mineralization rate, cortical bone levels, BMP-2, and VEGF[234]
Periodontal bone defectsAutologous periodontal ligament progenitorsSurgery, CALCITITE 4060-2 complex bone graft materialImproves gingival recession and decreases probing depth[235]
Sinus liftingAutologous DPSCsMicrografts in the nasal sinus on a collagen spongeBone density of the nasal sinus increased[236]
Periodontal bone defectsAutologous DPSCsSurgery, on a collagen spongeBone tissue regeneration[237]
OsteoradionecrosisAutologous DPSCsGrafted into the affected area, cells soaked in tricalcium phosphateRegeneration of mandibular bone tissue[238]
Periradicular periodontitisAutologous 
SHED and SCAPs
Implanted in a polyethylene glycol polylactic–polyglycolic acid scaffoldClosure of the apex and bone regeneration[239]
Intrabony defectsAutologous DPSCsSurgery, on a collagen spongeProbing depth decreased and bone tissue increased[240]
Periodontal bone defectsAutologous PDLSCsSurgery, on bovine bone materialsRegeneration of alveolar bone tissue[241]
Mandibular defect due to ameloblastomaAutologous DPSCsPacked inside a mesh and placed over the mandibleMandibular bone regeneration[242]
Irreversible pulpitisAutologous mobilized DPSCsInjected with atelocollagen and G-CSFThey regenerated the pulp and formed dentin[243]
Periapical lesionsAllogeneic SHEDAdministered in suspension in the root canalClosure of the apex, healing of the periapical tissue, and regeneration of the pulp[244]
Chronic periodontitisAutologous DPSCsGrafted on a collagen spongeProbing depth decreased and bone tissue increased[245]
Mandibular bone defects after extraction of third molarsAutologous DPSCsImplanted at extraction sites on collagen scaffoldsNo effect was observed[246]
Chronic periodontitisAutologous DPSCsMicrografts in a collagen spongeReduction in probing depth and bone regeneration[247]
Periodontal diseaseAllogeneic DPSCsSurgery, implantation in a lyophilized collagen–polyvinylpyrrolidone spongeProbing depth was reduced and bone tissue increased[248]
Pulp necrosisAutologous SHEDImplantation of the cells in the affected teethPulp tissue regeneration[249]
PeriodontitisAutologous PDLSCsCellular sheetsProbing depth was reduced and bone tissue was increased[250]
Irreversible pulpitisAutologous DPSCsAdministered at the affected siteRegeneration of functional pulp tissue[251]
Apical lesionsAutologous SHEDAdministration in root canalsHealing of the periapical tissue and the formation of functional pulp tissue[252]
Intrabony lesionsAutologous PDLSCsSurgery, implantation in the affected sitesReduction in probing depth[253]
Cleft lip and palateAutologous SHEDSurgery, implantation in a hydroxyapatite–collagen spongeGeneration of bone tissue[254]
Erectile dysfunctionCMs SHEDInjections into the corpora cavernosa of the penisImprovement of erections[255]
Diabetes mellitus 2Allogeneic SHEDIntravenousDecreased glycosylated hemoglobin and glycosylated serum albumin levels and caused fever, fatigue, and rash[256]
Huntington’s disease associated with primary lung adenocarcinomaAllogeneic SHEDIntravenousCells did not graft (homing) in the pre-existing tumor[257]
Irreversible pulpitisAutologous DPSCsAdministration of the cells in the root canals combined with G-CSF in atelocollagenRegeneration of functional pulp tissue[258]
Mandibular bone defects after extraction of third molarsAutologous DPSCsImplanted in the affected areasProbing depth was reduced[259]
Table 4. Registered clinical trials using D-MSCs.
Table 4. Registered clinical trials using D-MSCs.
IDStudy StatusConditionsInterventionsPhasesStudy Type
NCT04302519UnknownCOVID-19DPSCsEarly Phase 1Interventional
NCT04983225Active not recruitingPeriodontitisDPSCsPhase 1Interventional
NCT02731586UnknownEdentulous alveolar ridgeDPSCsEarly Phase 1Interventional
NCT05924373Not recruitingPeriodontitisDPSCsPhase 2Interventional
NCT05127369Not recruitingDepressionDPSCsNAInterventional
NCT02728115Active not recruitingHuntington’s diseaseDPSCsPhase 1Interventional
NCT04130100UnknownKnee osteoarthritisDPSCsEarly Phase 1Interventional
NCT03570333Active not recruitingGingival disordersGMSCsNaInterventional
NCT02728115Active not recruitingHuntington’s diseaseSHED (Cellavita HD)Phase 1Interventional
NCT04219241Active not recruitingHuntington’s diseaseSHED (Cellavita HD)Phase 2|Phase 3Interventional
NCT03252535CompletedHuntington’s diseaseSHED (Cellavita HD)Phase 2Interventional
NCT01082822UnknownPeriodontitisPDLSCsPhase 1|Phase 2Interventional
NCT04336254UnknownCOVID-19DPSCsPhase 1|Phase 2Interventional
NCT02209311UnknownAlveolar bone atrophyMSCs from oral mucosaPhase 1|Phase 2Interventional
NCT06043453RecruitingApical periodontitisDPSCs and SCAPsNAObservational
NCT03957655UnknownLiver cirrhosisSHEDEarly Phase 1Interventional
NCT01814436UnknownDental pulp necrosisSHEDNAInterventional
NCT05728346Not recruitingDental pulp necrosisSHEDNAInterventional
NCT03638154CompletedPeriodontal defectsGMSCsNAInterventional
NCT03137979UnknownPeriodontitisGMSCsPhase 1|Phase 2Interventional
NCT04434794CompletedGingival recessionGMSCsPhase 1|Phase 2Interventional
NCT02464202CompletedTooth transplantationPDLSCsNAInterventional
NCT02523651UnknownPeriodontal diseasesDPSCsPhase 1|Phase 2Interventional
ChiCTR2300073144Not recruitingPeriodontitisDPSCsPhase 2Interventional
RBR-65trt53Not recruitingCOVID-19DPSCsPhase 1|Phase 2Interventional
IRCT20140911019125N8RecruitingCOVID-19DPSCsPhase 2|Phase 3Interventional
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Poblano-Pérez, L.I.; Castro-Manrreza, M.E.; González-Alva, P.; Fajardo-Orduña, G.R.; Montesinos, J.J. Mesenchymal Stromal Cells Derived from Dental Tissues: Immunomodulatory Properties and Clinical Potential. Int. J. Mol. Sci. 2024, 25, 1986. https://doi.org/10.3390/ijms25041986

AMA Style

Poblano-Pérez LI, Castro-Manrreza ME, González-Alva P, Fajardo-Orduña GR, Montesinos JJ. Mesenchymal Stromal Cells Derived from Dental Tissues: Immunomodulatory Properties and Clinical Potential. International Journal of Molecular Sciences. 2024; 25(4):1986. https://doi.org/10.3390/ijms25041986

Chicago/Turabian Style

Poblano-Pérez, Luis Ignacio, Marta Elena Castro-Manrreza, Patricia González-Alva, Guadalupe R. Fajardo-Orduña, and Juan José Montesinos. 2024. "Mesenchymal Stromal Cells Derived from Dental Tissues: Immunomodulatory Properties and Clinical Potential" International Journal of Molecular Sciences 25, no. 4: 1986. https://doi.org/10.3390/ijms25041986

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