Previous Article in Journal
Comparing Handcrafted Radiomics Versus Latent Deep Learning Features of Admission Head CT for Hemorrhagic Stroke Outcome Prediction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Engineered Human Dental Pulp Stem Cells with Promising Potential for Regenerative Medicine

1
Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
2
Department of Pediatric Dentistry, Asahi University School of Dentistry, Gifu 501-0296, Japan
3
Department of Anatomy, Asahi University School of Dentistry, Gifu 501-0296, Japan
4
Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
5
Department of Genome Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
*
Author to whom correspondence should be addressed.
BioTech 2025, 14(4), 88; https://doi.org/10.3390/biotech14040088
Submission received: 29 September 2025 / Revised: 28 October 2025 / Accepted: 1 November 2025 / Published: 3 November 2025
(This article belongs to the Section Medical Biotechnology)

Abstract

The fields of regenerative medicine and stem cell-based tissue engineering hold great potential for treating a wide range of tissue and organ defects. Stem cells are ideal candidates for regenerative medicine because they are undifferentiated cells with the capacity for self-renewal, rapid proliferation, multilineage differentiation, and expression of pluripotency-associated genes. Human dental pulp stem cells (DPSCs) consist of various cell types (including stem cells) and possess multilineage differentiation potential. Owing to their easy isolation and rapid proliferation, DPSCs and their derivatives are considered promising candidates for repairing injured tissues. Recent advances in gene engineering have enabled cells to express specific genes of interest, leading to the secretion of medically important proteins or the alteration of cell behavior. For example, transient expression of Yamanaka’s factors in DPSCs can induce transdifferentiation into induced pluripotent stem cells (iPSCs). These gene-engineered cells represent valuable candidates for regenerative medicine, including stem cell therapies and tissue engineering. However, challenges remain in their development and application, particularly regarding safety, efficacy, and scalability. This review summarizes current knowledge on gene-engineered DPSCs and their derivatives and explores possible clinical applications, with a special focus on oral regeneration.
Key Contribution: DPSCs represent a valuable alternative to MSCs for regenerative medicine because of their easy access and minimal ethical concerns. Gene-engineered DPSCs and their derivatives (i.e., iPSCs and differentiated progeny) are also promising candidates for future cell-based therapies, including oral regeneration.

1. Introduction

Stem cells are defined as cells with the capacity to differentiate into various types of cells, a property known as multilineage differentiation or multi-differentiation, along with active proliferative activity (self-renewal) [1]. Several types of stem cells exist, including embryonic stem cells (ESCs), adult somatic stem cells (mesenchymal, hematopoietic, neuronal, and endothelial stem cells), and germ cell-specific stem cells, such as spermatogonial cells in the testis [2,3,4]. Induced pluripotent stem cells (iPSCs) and induced tissue-specific stem cells (i-TSCs), both generated by transfecting adult differentiated somatic cells with Yamanaka’s factors, are also valuable resources for regenerative medicine [2,3,4].
Bone marrow stem cells (BMSCs), derived from the bone marrow, are amongst the most widely used stem cells in clinical settings. They exhibit continuous proliferative capacity and multipotency, with the ability to differentiate into various types of cells, including osteoblasts, chondrocytes, adipocytes, tenocytes, and cardiomyocytes [5]. However, BMSCs are associated with high cost and an invasive collection procedure [6]. Dental pulp stem cells (DPSCs) are a population of cells, called dental pulp cells (DPCs), the resident mesenchymal stromal cells within the pulp cavity of impacted third molars [7] (Figure 1). Human DPSCs are ectodermal-derived stem cells originating during tooth development from ectodermal cells that migrate from the neural tube to the oral region [8,9,10]. Since their first isolation and characterization by Gronthos et al. [11], multiple reports have demonstrated the potential of DPSCs to differentiate into diverse cell types, including osteoblasts, adipocytes, chondrocytes, myocytes, neuronal and endothelial cells, hepatocytes, and melanocytes [12,13].
As mentioned above, DPSCs have potential as an alternative to BMSCs for stem cell-based therapies. However, whether DPSCs are comparable to BMSCs at the molecular level remains unclear. Table 1 presents a comparison of the characteristics of DPSCs and BMSCs based on the studies of Gimble et al. [14] and Ponnaiyan et al. [15]. Both DPSCs and BMSCs are multipotent cells that share many characteristics. However, key differences exist in their specific surface marker expression, differentiation capabilities, and proliferation rates. For example, both cell types strongly express the standard MSC-associated markers CD29 (integrinβ1 chain), CD44 (non-kinase transmembrane glycoprotein), CD73 (5′ectonucleotidase), CD90 (Thy1), and CD105 (endoglin). They typically lack expression of the hematopoietic and endothelial markers CD11b (integrin alpha-M), CD14 (lipopolysaccharide-binding protein), CD34 (transmembrane phosphoglycoprotein protein), and CD45 (lymphocyte common antigen), as well as the major histocompatibility complex class II (HLA-DR) marker. Interestingly, Kim et al. [16] used RNA sequencing (RNA-seq) to compare gene expression profiles in DPSCs and BMSCs and found distinct patterns of gene expression between the two cell types. Kumar et al. [17] demonstrated that DPSCs are a superior stem cell source for obtaining neural progenitor cells. DPSCs also exhibit greater proliferative potential than BMSCs [18,19]. Furthermore, DPSCs display stronger odontogenic capability than BMSCs, suggesting that they may be more suitable for tooth regeneration [20]. Compared with other adult stem cells, such as BMSCs, DPSCs are superior due to their easy, ethical, and non-invasive collection from extracted teeth, along with their high proliferative and multilineage differentiation capabilities, which make them a unique choice for regenerative medicine.
Engineered cells are genetically modified cells generated by adding, deleting, or altering genetic sequences at specific target sites in living cells [21]. As a result, these engineered cells may exhibit inactivation of target genes, acquisition of novel genetic traits, and correction of pathogenic gene mutations. They can also be used to explore the function of a gene of interest (GOI), introduce new characteristics (such as enhancement of proliferation or differentiation ability), secrete therapeutic proteins or small particles useful for regenerative medicine, or induce phenotypic changes, such as transdifferentiation and reprogramming into iPSCs or iTSCs [2,3,4]. These engineered cells, therefore, represent an attractive option for stem cell-based therapies.
In this review, we summarize the current knowledge on DPSCs with a special focus on the genetic engineering of primarily isolated dental cells and their use in studies related to regenerative medicine. We also discuss potential clinical applications of engineered DPSCs and their derivatives, particularly emphasizing the generation of three-dimensional (3D) bioengineered teeth embedded in various scaffold biomaterials and the subsequent grafting of these structures for regenerating damaged dental tissues.

2. Isolation and Culture of DPSCs

To date, various types of stem cells have been identified and isolated from oral tissues in the dental field, which serve as promising sources for regenerative therapies. In addition to DPSCs, which are present in the dental pulp, several other stem cells populations have been reported, including stem cells from human exfoliated deciduous teeth (SHED) [18], periodontal ligament (periodontal ligament stem cells, PDLSCs) [22], dental follicle (dental follicle stem cells, DFSCs) [23], apical papilla, (stem cells apical papilla, SCAP) [24,25], human deciduous teeth-derived DPCs (HDDPC) [26], and gingival stem cells (GING SCs) [27].

2.1. DPSCs from Enzymatic Digestion of DP Tissue (DPSC-ED) vs. DPSCs Obtained Through Outgrowth from Tissue Explants (DPSC-OG)

DPSCs can be isolated from either deciduous or permanent teeth. After tooth extraction, the tooth surface is wiped with 70% ethanol and rinsed with sterile water. The tooth is then cut to expose the pulp cavity, and dental pulp tissue is obtained by osteotomy (using osteotomy forceps or osteotomy pliers). There are two main approaches for DPSC dissociation: direct isolation from pulp tissue (DPSC-ED) and the outgrowth method using tissue explants (DPSC-OG) [28,29]. The former is a common method involving mechanical extraction of the soft pulp connective tissue, followed by maceration and enzymatic digestion with collagenase and dispase to release the cells from the dental pulp tissue. The isolated cells are then seeded onto plastic culture plates. Although the number of cells obtained tends to be low, this method allows the isolation of relatively homogenous cell populations. In contrast, the explant method allows cells to migrate out of the pulp tissue [30]. Hilkens et al. [30] reported that pulp tissues isolated are directly placed on culture plates, and cells gradually migrate outward. This approach generally requires more time than DPSC-ED. Comparisons between the two methods indicate that DPSC-ED tends to exhibit higher mineralization capacity, while both methods yield cells capable of adipogenic, chondrogenic, and osteogenic differentiation [30]. In DPSC-OG, although the number of recovered cells is higher, this method may select for specific subpopulations, thereby decreasing population homogeneity. Recently, Kiyokawa et al. [31] established a novel approach called scratch-based isolation of primary cells from human dental pulps (SCIP), which is conceptually similar to DPSC-OG. In this method, dental pulp tissue, briefly digested with proteolytic enzymes, is scratched onto a culture dish and grown in a suitable medium. By day 2, cells begin to spread radially from the pulp, and by day 10, they reach a semi-confluent state. Cells harvested by trypsinization consistently yield over 1 million cells, and upon re-cultivation, they can be propagated for more than ten passages. Importantly, their proliferative and differentiation capacities after the 10th passage remain comparable to those of early-passage cells. These isolated cells continue to express stem cell-specific and differentiation-related markers, even after extended passage. The simplicity, high success rate, and adaptability of SCIP, including its applicability to patients with genetic diseases, make it a valuable method for regenerative medicine research and clinical applications.

2.2. Optimizing Conditions for DPSCs in Cell Culture

Current techniques for expanding DPSCs require the use of fetal bovine serum (FBS). In particular, medium containing 20% FBS is often considered optimal for DPSC cultivation [29,32]. However, animal-derived reagents (such as FBS) pose safety concerns for clinical therapy, including risks of infection and immunological reactions following transplantation. In addition, the proliferation and differentiation of DPSCs vary considerably, depending on the serum batches. Expanding DPSCs in medium supplemented with human serum (HS) may overcome these problems because HS has been shown to support better proliferation of DPSCs than FBS [33]. Furthermore, inclusion of 1% HS in a chemically defined medium was also beneficial for DPSC proliferation. The type of serum also affects the differentiation potential of the cells. Khanna-Jain et al. [33] demonstrated that cultivation of DPSCs in HS-containing medium promoted more pronounced adipogenic and osteogenic differentiation, while FBS-containing medium supported more effective chondrogenic differentiation. Based on these findings, HS appears to be a suitable alternative to FBS for the safe expansion of DPSCs, although there is a possibility that serum batches can significantly affect experimental results.

2.3. Requirement for Xeno-Free Cell Culture System of DPSCs for Human Cell Therapy

Animal-derived components have traditionally been widely used in stem cell cultures. However, serious concerns remain regarding using the cultured DPSCs for human cell therapy, such as the risk of viral, bacterial, fungal, and prion contamination as well as the possible induction of immune rejection of the transplanted cells into the host [34]. For these reasons, the development of xeno-free cell culture protocols that meet Good Manufacturing Practice (GMP) standards has been continuously tested for stem cell culture [34].
Fujii et al. [7] succeeded in cultivating DPSCs using serum-free medium, STK2 [containing Iscove’s modified Dulbecco’s medium (IMDM), recombinant human albumin, hydrocortisone, and an optimal mix of growth factors, including basic fibroblast growth factor (bFGF)], a novel serum-free medium developed for human MSC multiplication. When compared with DPSCs in control medium [Dulbecco’s modified Eagle’s medium (DMEM) containing 10% FBS], those cultured in STK2 exhibited active proliferation. Furthermore, DPSCs grown in STK2 induced to undergo osteogenesis exhibited alkaline phosphatase (ALP) activity and calcification, indicating maintenance of differentiation potential in STK2. Mochizuki and Nakahara [35] employed a xenogeneic serum-free culture medium (XFM) (containing a basal medium of amino acids, vitamins, inorganic salts, and glucose, along with a supplement that replaces animal serum with human or chemically defined alternatives, growth factors, hormones, and lipids) for handling DPSCs. The DPSCs cultivated in XFM exhibited typical MSC characteristics in vitro and in vivo, including marker gene/protein expression, trilineage differentiation potential, and hard osteo-dentin tissue formation. Xiao et al. [36] established a chemically defined serum-free culture system [called serum-free Essential 8 medium (E8); provided by Thermo Fisher Scientific Co., Ltd., Waltham, MA, USA] for culturing DPSCs. Compared with serum-containing medium, E8 medium exhibited better ability to maintain cell proliferation, pluripotency, migration, and stability. However, some researchers indicated that these media do not adequately support DPSC proliferation and differentiation. On the other hand, various human blood derivatives have been proposed as alternatives to animal serum for stem cell culture, including autologous or allogenic human serum, human plasma, and human platelet lysates (PL) with their released factors. Marrazzo et al. [37] found that a low concentration of PL (1%) was able to support the growth and maintain the viability of DPSCs in vitro. Additionally, PL was shown to be a suitable option for protocols promoting osteogenic and chondrogenic differentiation of DPSCs.

2.4. Single-Cell Cloning and Single-Cell-Based RNA-Seq (scRNA-Seq)

Gronthos et al. [11] first demonstrated that human DPSCs can be isolated through DPSC-ED. The same group [38] also attempted to obtain single-colony-derived DPSC clones to determine whether DPSCs have odontogenic potential. Of the 12 individual clones analyzed, two-thirds generated abundant ectopic dentin in vivo, while only a limited amount of dentin was detected in the remaining one-third. These results suggest that DPCs are composed of various types of DPSCs.
RNA-seq analysis is a useful tool to distinguish the properties of cell types. This approach is particularly powerful for identifying cells at the single-cell level through transcript quantification in individual cells [39]. Kobayashi et al. [40] examined the transcriptome to identify new candidate marker genes in DPSCs using RNA-seq analysis of clonally isolated cells from heterogeneous multipotent human DPSCs. They first obtained 50 colony-forming single-cell-derived clones from an 11-year-old female patient. DNA microarray analysis of five representative clones with strong differentiation potentials revealed 1227 genes related to multipotency. Ninety of these genes overlapped with those involved in “stemness or differentiation”. Based on the predicted locations of expressed protein products, 14 of the 90 genes were selected as candidate DPSC markers, which are particularly associated with multipotential characteristics. These included desmoplakin (DSP), serpin family E member 1 (SERPINE1), sortilin 1 (SORT1), collagen type I alpha 2 chain (COL1A2), collagen type III alpha 1 chain (COL3A1), ATPase phospholipid transporting 8B1 (ATP8B1), intercellular adhesion molecule-1 (ICAM1), adhesion G protein-coupled receptor A2 (ADGRA2), anthrax toxin receptor 1 (ANTXR1), oxytocin receptor (OXTR), and serglycin (SRGN). These new candidate marker genes may be useful for analyzing or enriching multipotent stem cells.

2.5. Usefulness of mRNA Analysis of a Small Number of DPSCs for Creating Gene Expression Profiles

Although scRNA-seq is currently the gold standard for analyzing cell-specific expression profiles, one major limitation is the requirement for expensive, cell-sorting equipment (FACS) to exclude debris and dead or unwanted cells before sequencing. Furthermore, this approach requires pre-staining of live cells with antibodies recognizing cell surface antigens. Inada et al. [41] developed a novel method enabling mRNA analysis of a small number of fixed and immuno-stained cells, specifically expressing octamer-binding transcription factor 3/4 (OCT3/4). OCT3/4 is a transcriptional factor essential for maintaining stemness in various stem cells and is expressed in only a few HDDPCs [42]. Since OCT3/4 is localized in the nucleus (and not on the cell surface), it cannot be used with FACS, which relies largely on the isolation of live cells using cell surface antigen markers. However, isolation of 3,3′-diaminobenzidine-stained OCT3/4-expressing cells is possible by manually collecting immuno-stained cells with a mouth-controlled micropipette under a dissecting microscope. When these manually isolated cells (2~10) were subjected to RNA isolation using a whole transcriptome amplification (WTA) kit, successful amplification of OCT3/4 mRNA-derived RT-PCR products was achieved [41]. This approach is termed “RNA analysis based on a small number of manually isolated fixed cells (RNA-snMIFxC)”. Notably, a similar approach was reported by Mutisheva et al. [43], who attempted to establish a method for FACS sorting of fixed cells for scRNA-seq without compromising data. Using murine pancreatic ductal adenocarcinoma samples, in which the number of live cells is limited, they compared semi-automated mechanical/enzymatic digestion of solid tumors using the gentleMACS Dissociator with mechanical dissociation alone. This protocol enabled tissue dissociation and staining within a single day, followed by subsequent cell sorting and scRNA-seq, which represents a major advantage for processing clinical patient material.

2.6. Markers for DPSCs

As previously shown by Gornthos et al. [38] and through RNA seq analysis [44], DPSCs are a heterogeneous mixture of cell populations. This heterogeneity is further supported by the expression of stemness markers, such as OCT3/4 and SRY-related HMG-box 2 (SOX2), which are predominantly expressed in the nucleus. For example, immunocytochemical staining of HDDPC-derived DPSCs demonstrated that some cells expressed these markers, whereas others did not [42]. Notably, Kawashima et al. [45] listed CD13 (aminopeptidase N), CD146 (melanoma cell adhesion molecule; MCAM), and CD166 (activated leucocyte cell adhesion molecule; ALCAM) as new cell surface markers for DPSCs, as well as CD29 and CD44, both of which are already shown in Table 1. Furthermore, Madhoun et al. [14] reported that DPSCs express several neural stem cell markers, including nestin (NES), vimentin (VIM), β-III tubulin (TUBB3), musashi1 (MSI1), galactosylceramidase (GALC), S100 calcium-binding protein B (S100B), neurofilament heavy chain (NEFH), S100 calcium-binding proteins (S100), Notch1 (NOTCH1), CD271 (nerve growth factor receptor; NGFR), and synaptophysin (SYP).

2.7. Multilineage Differentiation Potential of DPSCs

The plasticity of DPSCs allows them to differentiate into various cell types of endodermal (respiratory and gastrointestinal tracts, liver, pancreas, thyroid, prostate, and bladder lineages), mesodermal (adipogenic, osteogenic, and chondrogenic lineages), and ectodermal (skin and neural lineages) origin [46]. By manipulating culture conditions, specific differentiation pathways can be restricted, enabling the generation of cultures enriched in particular precursors in vitro. Table 2 lists experiments on differentiation induction of human DPSCs using non-gene-engineered approaches.

2.7.1. Osteogenic (Osteoblastic) Differentiation

Osteogenic (osteoblastic) differentiation of DPSCs has been successfully induced using chemical reagents such as dexamethasone, L-ascorbic acid, and β-glycerol phosphate supplementation [49,52]. In addition to cultivation in osteogenic medium, various approaches have been reported for inducing differentiation into the osteogenic lineage, including co-cultivation of DPSCs with endothelial cells [47], cultivation in the presence of human serum [48], concentrated growth factor exudate (CGFe), and transforming growth factor beta 1 (TGF-β1) [54], or betaine (BET) [55]. Growth of DPSCs in the presence of various scaffolds [50,57,58], decellularized adipose tissue solid foams [56], or microcapsules [53] has also resulted in successful osteogenic differentiation. Alizarin red S staining is typically used to confirm matrix mineralization and calcium deposition after induction [51]. Concomitantly, expression of bone-specific proteins such as alkaline phosphatase (ALP), collagen type I (COL1), osteocalcin (OCN), osteonectin (ON), osteopontin (OPN), osterix (Osx), and runt-related transcription factor 2 (RUNX2) is also detected [49].

2.7.2. Odontoblastic Differentiation

For odontoblastic differentiation of human DPSCs, Baldión et al. [64] used odontogenic induction medium (containing TGF-β1, dexamethasone, β-glycerophosphate, and ascorbic acid) and obtained odontoblast-like cells with odontoblast markers such as dentinal sialophosphoprotein (DSPP), dentin matrix protein-1 (DMP-1), and RUNX2 transcripts. These cells exhibited mineral deposition activity, as evidenced by Alizarin Red S and von Kossa staining, along with low ALP activity. Odontogenic differentiation was also achieved by cultivating DPSCs on dentin [59], calcium silicate materials [61], or scaffolds [62,63]. Furthermore, cultivation of DPSCs in preameloblast-conditioned medium (PA-CM) [60] with endothelin-1 (ET-1) [65] or vascular endothelial growth factor A (VEGFA) [66] also led to the generation of odontoblast-like cells.

2.7.3. Differentiation into Adipogenic Cell Lineages

For differentiation into adipogenic cell lineages, DPSCs are treated with insulin, dexamethasone, indomethacin, and 3-isobutyl-1-methylxanthine (IBMX) [52,69]. Enhanced adipogenic differentiation of DPSCs has also been observed in enzymatically decellularized adipose tissue solid foams [70]. The resulting adipogenic cells contain lipid droplets in their cytoplasm, which can be visualized using Oil Red O staining [67]. The adipogenic phenotype is usually further confirmed by the expression of several adipogenic markers, such as peroxisome proliferator-activated receptor γ (PPAR-γ), glucose transporter type 4 (GLUT4), fatty acid binding protein 4 (FABP4), and lipoprotein lipase (LPL) [68].

2.7.4. Differentiation into Neurogenic Cell Lineage

DPSCs have been shown to differentiate into neuron-like cells, dopaminergic neurons, oligodendrocytes, and Schwann cells when cultivated in specific media that promote neuronal differentiation [78]. For example, Osathanon et al. [72] demonstrated that neurogenic differentiation was achieved when DPSCs were first cultured in neuroinduction medium containing B27 (a serum-free supplement optimized to promote neuronal cell differentiation), L-glutamine, bFGF, and epidermal growth factor (EGF) for 7 days. Subsequently, single cells derived from spheres were seeded onto collagen type IV-coated dishes in neuroinduction medium supplemented with retinoic acid for another 7 days. DPSCs have also been induced into neuron-like cells after incubation in commercially available neurogenic differentiation medium [73], conditioned medium of cerebrospinal fluid and retinoic acid (RA) [74], or medium containing nerve growth factor (NGF) [76]. Culturing in the presence of graphene–polycaprolactone hybrid nanofibers [75] or under high K+ stimulation (50 mM KCl) [77] can also induce transdifferentiation of DPSCs into neuron-like cells. Successful neurogenic differentiation is confirmed by the expression of several neural markers, including neuronal nuclei (NeuN), microtubule-associated protein 2 (MAP2), neural cell adhesion molecule (NCAM), growth-associated protein 43 (GAP43), glial fibrillary acid protein (GFAP), synapsin I (SYN1), and TUBB3 [96].

2.7.5. Differentiation into Hepatocytes

Hepatic differentiation of human DPSCs has been successfully demonstrated by incubating CD117-positive DPSCs in medium supplemented with hepatic growth factor, insulin-transferrin-selenium-x, dexamethasone, and oncostatin M [79]. The resulting hepatocyte-like cells exhibited substantial cytoplasmic glycogen storage and expressed hepatic markers such as alpha-fetoprotein (AFP), albumin, hepatic nuclear factor-4 alpha (HNF4α), insulin-like growth factor-1 (IGF-1), and carbamoyl phosphate synthetase (CPS) [79].

2.7.6. Differentiation into Endothelial Lineage

Endothelial differentiation (vasculogenesis) can be achieved by incubating DPSCs in a medium supplemented with a mixture of B27, heparin, and growth factors, including VEGF-A165 [83]. In particular, in vitro Matrigel assays demonstrated that DPSCs can generate vascular tubules in 3D cultures. Bento et al. [80] reported that DPSCs exposed to endothelial growth medium supplemented with VEGF differentiated into VEGF receptor 2 (VEGFR2)-positive and CD31-positive endothelial cells in vitro. They suggested that the VEGF/mitogen-activated protein kinase kinase 1 (MEK1)/extracellular signal-regulated kinase (ERK) signaling pathway acts as a key regulator of DPSC endothelial differentiation. Furthermore, co-cultivation with human umbilical vein endothelial cells (HUVECs), after encapsulation in a scaffold system of self-assembling peptide nanofibers, resulted in the generation of DPSC-derived endothelial cells [82]. Endothelial differentiation can be confirmed by the expression of several angiogenesis-related markers, including CD54/ICAM-1, CD146/MCAM, monocyte chemotactic protein-1 (MCP-1), von-Willebrand factor (VWF) (domain 1 and 2), CD31/platelet/endothelial cell adhesion molecule-1 (PECAM-1), VEGF, CD34, fetal liver kinase 1 (Flk-1)/VEGFR2, bFGF, insulin-like growth factor binding protein-3 (IGFBP3), interleukin-8 (IL-8), plasminogen activator inhibitor-1 (PAI-1), tissue inhibitors of metalloproteinase-1 (TIMP-1), and urokinase-type plasminogen activator (uPA) [81].

2.7.7. Differentiation into Cardiomyocytes

DPSCs can differentiate into cardiomyocytes when incubated in the presence of 5-azacytidine, a well-known demethylating agent, for 2 days [84]. The resulting embryoid bodies (EBs) express various precardiac markers, including myosin heavy chain 6 (MYH6) and mesoderm posterior BHLH transcription factor 1 (MESP), as well as markers for the three germ layers. When these EBs are plated onto gelatin-coated tissue culture dishes, they develop functional cardiomyocytes expressing cardiac markers such as NK-2 transcription factor related, locus 5 (Nkx2.5), and connexin 43 (Cx43).

2.7.8. Differentiation into Pancreatic Lineage

Mesenchymal stem cells can be induced into the pancreatic cell lineage using published three-step induction protocols designed to enhance pancreatic progenitor commitment and production yield. These protocols often employ growth factors, such as activin A and retinoic acid, along with other factors, like nicotinamide and laminin, to guide cell differentiation [86]. Ishkitiev et al. [86] confirmed that CD117+ DPSCs (which express CD117, a receptor tyrosine kinase encoded by the KIT gene) can differentiate into the pancreatic lineage when subjected to the three-step induction protocol. The resulting cells expressed pancreatic-specific endocrine markers, including insulin, glucagon (GCG), somatostatin (SST), pancreatic polypeptide (PPY), and exocrine marker amylase-2a (AMY2A). Additional markers such as pancreatic and duodenal homeobox 1 (PDX1), hematopoietically expressed homeobox (HHEX), motor neuron and pancreas homeobox 1 (MNX1), neurogenin 3 (NEUROG3), paired box 4 (PAX4), paired box 6 (PAX6), and NK6 homeobox 1 (NKX6-1) were also detected. Similar findings were reported by several other laboratories [85,89,90,91]. Notably, Abuarqoub et al. [92] employed PSC Definitive Endoderm Induction media (Thermo Fisher Scientific) to induce pancreatic β cell differentiation.
Islets containing insulin-producing cells (IPCs) have also been successfully generated using DPSCs. Kanafi et al. [87] used a stepwise protocol to accelerate differentiation towards the pancreatic lineage and succeeded in generating islet-like cell clusters (ICCs). These ICCs were embedded in immuno-isolatory biocompatible macro-capsules and into streptozotocin (STZ)-treated diabetic mice. The grafted mice exhibited normoglycemia 3–4 weeks after transplantation, providing an autologous and non-controversial source of human tissue for potential stem cell therapy in diabetes. Mendoza et al. [88] further demonstrated that cultivating DPSCs in a 3D culture system using a stepwise protocol generated organoid-like 3D structures resembling pancreatic islets. These structures showed enhanced insulin and C-peptide production, expressed multiple pancreatic markers, and exhibited glucose-dependent insulin secretion.

2.7.9. Differentiation into Smooth Muscle Cells (SMCs)

Song et al. [93] incubated DPSCs in bladder SMC-conditioned medium supplemented with TGF-β1 for 2 weeks. The resulting cells expressed SMC-specific markers, including alpha smooth muscle actin (α-SMA), desmin (DES), and calponin (CNN), along with myosin. Xu et al. [94] used TGF-β1 and bone morphogenetic protein 4 (BMP4) to test the induction of DPSCs into SMCs and concluded that TGF-β1, but not BMP4, is effective for that purpose. Zhang et al. [95] demonstrated that direct contact with endothelial cells in the presence of TGF-β1 is required to drive DPSCs toward SMC differentiation. Song et al. [93] suggest that human DPSCs could serve as an alternative and less invasive source of stem cells for smooth muscle regeneration.

3. Gene Engineering of DPSCs

DPSCs have shown great potential as a cell-based therapy for regenerative medicine since they can be isolated relatively easily and possess properties similar to those of bone marrow-derived MSCs. However, their utilization alone is limited due to the insufficient number of available cells and the low production of therapeutic factors needed to repair injured sites after transplantation. One approach to overcome these limitations is to generate gene-engineered DPSCs, enabling enhanced cellular differentiation (including transdifferentiation) into various cell types, production of cells that secrete specific therapeutic proteins, and generation of immature cells with high pluripotency, as exemplified by iPSCs. These genetically modified (GM) DPSCs hold promise for a wide range of tissue engineering applications as a cell-based therapy. In Figure 2, a summary of recent achievements in gene-engineered DPSCs is shown.

3.1. Development of Methods for Gene Introduction into DPSCs

To date, several methods have been developed for gene delivery into DPSCs, including transduction using lentiviral vectors [97,98,99,100,101,102], adenoviral vectors [103], retroviral vectors [104], and electroporation or chemical reagent-based transfection using plasmids or transposons [105,106,107,108,109,110,111]. In particular, “loss-of-function” experiments using RNA interference (RNAi)-based approaches, such as using siRNA and shRNA, as well as microRNA (miRs), have been widely applied. MiRs directly or indirectly regulate in vitro DPSC differentiation into various cell types and functions through different signaling pathways [112]. For example, Liu et al. [113] first applied an miR-based strategy to promote odontoblastic differentiation of mouse DPSCs and reported increased expression of DSPP and DMP-1 following lentivirus-mediated introduction of miR-145 and miR-143. Mechanistically, MiR-145 binds to the 3′-UTRs of Krüppel-like factor 4 (KLF4) and OSX, suppressing their expression. Downregulation of miR-143 partially reduces miR-145 levels, thereby releasing KLF4 and OSX expression. Upregulation of these transcription factors, in turn, enhances the expression of the odontoblast markers DSPP and DMP-1, ultimately driving odontoblast differentiation.

3.2. Immortalization

Primary cells, including DPSCs, have limited growth potential due to cellular senescence. Immortalization is a technique by which primary cells acquire continuous proliferative capacity without losing the characteristics of their parental cells [114]. Immortalization can be achieved through gene delivery of human telomerase reverse transcriptase protein (hTERT), E6/E7 proteins from human papilloma virus 16 (HPV16), or simian virus 40 large T antigen (SV40T) [115]. Inada et al. [107] immortalized HDDPCs by piggyBac-based transfection with hTERT and HPV16 genes. The resulting immortalized cells demonstrated continuous proliferation but lacked tumorigenic potential, as confirmed by transplantation under the skin of immunocompromised mice. Moreover, they retained the properties of parental DPSCs, including mineralizing capability, expression of specific markers, and multipotency. These immortalized DPSCs are now recognized as a valuable cell model for studying the mechanisms of odontoblast proliferation and differentiation [116]. Zanini et al. [117] showed that Biodentine, a new tricalcium silicate-based cement, induced differentiation of immortalized murine DPSCs into odontoblast-like cells and enhanced both cell proliferation and biomineralization. Similarly, betaine (BET) treatment promotes osteogenic differentiation in immortalized or primary-cultured human DPSCs, accompanied by increased mineral deposition and upregulation of osteogenic marker expression [52,118].

3.3. Alteration of Cell Behavior

Using these gene delivery methods, several biologically important subjects have been investigated. Recent reports on altered cell behavior and properties following gene delivery are summarized in Table 3.

3.3.1. Enhanced Differentiation into Odontoblast/Mineralization

Gene delivery experiments using DPSCs have demonstrated that several molecules enhance their differentiation potential towards odontoblasts. These include growth/differentiation factor 11 (Gdf11) [105], KLF4 [120], DNA damage-inducible transcript 3 (DDIT3) [124], inhibitor of DNA binding 1 (ID1) [125], zinc finger and BTB domain-containing 20 (ZBTB20) [126], SRY-related HMG-box 2 (SOX2) [128], platelet-derived growth factor-BB (PDGF-BB) [99], DMP-1 [134], and long noncoding RNA H19 (LncRNA H19) [135]. Notably, overexpression of the Notch ligand Jagged-1 (JAG-1) activated the Notch signaling pathway, inhibited odontoblastic differentiation, and mineralized tissue formation [119]. These results highlight the role of Notch signaling in regulating DPSC behavior. Wang et al. [121] used lentivirus-mediated knockdown for the Notch ligand Delta1 (also called delta-like canonical Notch ligand, DLL1) in DPSCs. DLL1-deficient DPSCs exhibited accelerated differentiation into odontoblasts. Furthermore, Chen et al. [123] showed that transfection with RNAi for CD44, a hyaluronan-binding cell surface signal transducing receptor, suppressed mineralization, suggesting a role of CD44 in DPSC mineralization.

3.3.2. Enhanced Differentiation into Osteogenic Lineage

Since DPSCs are considered a promising source for tissue engineering, particularly for osteogenic tissues, increasing their osteogenic capacity is crucial for their potential application in tissue engineering. Therefore, it is important to identify new therapeutic targets and elucidate the mechanisms regulating the osteogenic differentiation of DPSCs [177]. Several molecules promote osteogenic differentiation in DPSCs, including B cell lymphoma-2 (BCL-2) [127], SOX2 [138], TGF-β1 [111], Wnt family member 4 (WNT4) [139], ephrinB2 (EFNB2) [101], hsa_circ_0026827 [140], hypoxia-inducible factor-1α (HIF-1α) [141], and semaphorin 3A (SEMA3A) [142]. Growing evidence indicates that miRs influence osteogenic differentiation of DPSCs by regulating different aspects of this process. For example, Zhang et al. [178] reported that overexpression of miR-143, which is involved in metabolism, cell proliferation, inflammation, and apoptosis, inhibited osteogenic differentiation of DPSCs. Bioinformatics analysis and luciferase reporter assays showed that tumor necrosis factor-alpha (TNF-α) is a direct target of miR-143 in DPSCs, with miR-143 regulating TNF-α expression through post-transcriptional binding to its 3′UTR. Functional analysis further revealed that miR-143 inhibited TNF-α-induced osteogenic differentiation of DPSCs, suggesting that miR-143 suppresses this process by downregulating TNF-α. Conversely, inhibition of miR-143 promoted osteogenic differentiation of DPSCs through activation of the nuclear factor kappa B (NF-κB) signaling pathway.

3.3.3. Enhanced Cell Proliferation

Cell proliferation of DPSCs is promoted by several factors, including overexpression of DLL1 [121], DDIT3 [124], SOX2 [145], PDGF-BB [99], stromal cell-derived factor-1α (SDF-1α) [148], VEGF [148], Lin28 [149], TGF-β1 [111], LncRNA H19 [153], and adrenomedullin (ADM) [154]. RNAi-mediated suppression of DLL1 in DPSCs resulted in the inhibition of proliferation [121], suggesting the involvement of the Notch–Delta signaling pathway in this process. The external environment is also known to affect DPSC proliferation. Fukuyama et al. [144] demonstrated that siRNA-mediated downregulation of the catalytic alpha 1 subunit of AMP-activated protein kinase (AMPK) (AMPKα1)—a stress-responsive enzyme activated by hypoxia—led to decreased proliferation under both normoxic and hypoxic conditions, indicating that AMPK plays an important role in DPSC’s reactions to hypoxia, presumably in maintaining energy homeostasis. According to Fukuyama et al. [144], hypoxia leads to the activation of AMPK via an increase in the AMP/ATP ratio, and AMPK acts as an intercellular energy sensor, maintaining energy balance during hypoxia in a variety of cells. Consistent with this, miR-210-3p, a major hypoxia-inducible miR (also known as hypoxamir, widely expressed in many cell types), promoted proliferation and early differentiation of DPSCs under hypoxia [150]. Hara et al. [122] showed that transfection of miR-720, which targets the stem cell marker NANOG, reduced DPSC proliferation while promoting odontogenic differentiation through NANOG suppression. Similarly, Zhang et al. [150] reported that overexpression of miR-633, which targets the 3′ UTR of matrix extracellular phosphoglycoprotein (MEPE), significantly enhanced both proliferation and differentiation of DPSCs. They concluded that MEPE is a molecular biomarker regulating the differentiation of DPSCs.

3.3.4. Enhanced Angiogenic Commitment

DPSCs play a critical role in angiogenesis during dental pulp tissue repair and regeneration. The angiogenic capacity of DPSCs can be enhanced through overexpression of angiogenic factors such as PDGF-BB [99,158], SDF-1α or VEGF [148], BCL-2 [156], and Ets variant transcription factor 2 (ETV2) [157]. For example, Zhu et al. [148] demonstrated that overexpression of SDF-1α or VEGF caused enhanced cell proliferation and endothelial cell migration. Notably, culturing these engineered cells in Matrigel led to vascular-tube formation. These results suggested that a combination of VEGF-overexpressing and SDF-1α-overexpressing DPSCs could further increase the extent of vascularized dental pulp regeneration in vivo.
miRs are also known to indirectly regulate DPSC differentiation into vascular endothelial cells by modulating factors, such as hypoxia-inducible factor-1 (HIF-1) and VEGF. For instance, Liu et al. [155] identified VEGF and kinase insert domain-containing receptor (KDR) as targets of miR-424 during endothelial differentiation of DPSCs. Inhibition of miR-424 promoted endothelial differentiation, whereas its overexpression suppressed angiogenic potential. Specifically, miR-424 inhibition enhanced the secretion of angiogenic factors and upregulated receptor expression, thereby improving differentiation efficiency.

3.3.5. Enhanced Neurogenic Differentiation and Neuroprotective Effects

DPSCs can be differentiated into neural cells (neurons and glia) in vitro using growth factors such as brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), bFGF, and RA in specific induction media. The resulting neural cells can potentially be used for therapeutic treatment of neurodegenerative diseases. Differentiation of DPSCs into neural cells via gene delivery of exogenous DNA, which reprograms the cells towards a neuronal lineage, is also possible. For example, Behrouznezhad et al. [159] hypothesized that overexpression of zinc finger protein 521 (Zfp521), a transcription factor regulating several genes, in DPSCs could facilitate neural differentiation through chromatin modifications that activate neural specification genes.
A reliable method for converting DPSCs into neuronal stem cells (NSCs) has not yet been fully established. Gancheva et al. [161] employed OCT3/4 overexpression in combination with neural inductive conditions to reprogram DPSCs into NSCs. The reprogrammed DPSCs exhibited a neuronal-like phenotype, increased neural marker expression, limited sphere-forming ability, and enhanced neuronal but not glial differentiation. In vivo analysis using a developmental avian model showed that implanted DPSCs survived in the developing central nervous system (CNS) and responded to endogenous signals, displaying neuronal phenotypes. These findings indicate that OCT3/4 enhances the neural potential of DPSCs, confirming characteristics consistent with neuronal progenitors.

3.3.6. Enhanced Adipogenic Commitment

RNA-seq analysis demonstrated that several adipogenesis-related genes are influenced by complex methylation changes, leading to a differential gene expression profile in adipocytes under lean and obese conditions [179]. Argaez-Sosa et al. [180] showed that the ability of DPSCs to differentiate into adipocytes may be linked to the expression patterns of DNA methylation-related genes acquired during adipogenic induction. They again [162] showed that overexpression of ten-eleven-translocation 2 (TET2), a key regulator of DNA methylation during adipogenic induction, in DPSCs increased the expression of adipogenic marker genes and enhanced the transition of DPSCs toward adipogenic commitment.

3.3.7. Reduced Inflammatory Responses

Hepatocyte growth factor (HGF) exhibits both pro-inflammatory and anti-inflammatory effects in the context of DPSCs and inflammation, depending on the stimulus [181]. Meng et al. [164] demonstrated that overexpression of HGF in DPSCs led to reduced spleen mass in psoriatic mice and downregulation of inflammation-related factors, such as interferon-gamma (IFN-γ), TNF-α, and interleukin (IL)-17A, suggesting that DPSCs exert enhanced therapeutic effects on psoriasis by reducing inflammatory responses. Furthermore, Ni et al. [165] demonstrated that overexpression of WNT4 alleviated the inflammatory response, enhanced BCL-2 expression, and decreased apoptosis rate in DPSCs stimulated with lipopolysaccharide (LPS) by inhibiting the inhibitor of κB kinase (IKK)/nuclear factor-kappa B (NF-κB) pathway.

3.3.8. Enhanced Cell Migration

Peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1), a member of the prolyl isomerase family, specifically binds to phosphorylated Ser/Thr-pro motifs to catalytically regulate the post-phosphorylation conformation of its substrates. Kim et al. [166] investigated the role of Pin1 expression in DPSCs in regulating the P2Y1 receptor, a purinergic G protein-coupled receptor (GPCR) activated by ADP, and activating ADP-mediated P2Y1 signaling. Silencing PIN1 by siRNA reduced the migration of DPSCs expressing the P2Y1 receptor, suggesting that PIN1 is involved in P2Y1-mediated cell migration.

3.3.9. Generation of IPCs and Oligodendrocyte Progenitors (OPs) from DPSCs by Forced Expression of Exogenous Genes

In 2008, Melton and colleagues [182] first demonstrated that three transcription factors—neurogenin 3 (Ngn3; also known as Neurog3), Pdx1, and MAF bZIP transcription factor A (Mafa)—introduced in vivo could successfully reprogram differentiated pancreatic exocrine cells in adult mice into cells closely resembling β cells. This process is referred to as “direct reprogramming of hepatocytes to pancreatic cells”. The induced β cells were indistinguishable from endogenous islet β cells in size and shape, expressed genes essential for β cell function, and were able to ameliorate hyperglycemia in diabetic animal models. Since then, successful conversion of non-β cells, such as DPSCs [167,168] and keratinocytes [183] into β cells, has been reported, suggesting that reprogramming into β cells is not strictly limited to lineage-related cells. For example, Nozaki and Ohura [169] reported that miR-183 was downregulated during the direct conversion of DPSCs to IPCs based on miR array profiling. The miR-183 family, which includes miR-183, miR-96, and miR-182, is highly expressed in a variety of non-sensory diseases, including cancer, neurological, and autoimmune disorders [184]. Nozaki and Ohura [168] further demonstrated that downregulation of miR-183 in DPSCs resulted in the generation of insulin-expressing cells within 72 h after ectopic expression of an miR-183 inhibitor.
This concept broadens the potential gene-based transdifferentiation approaches in regenerative medicine, as various cells of different origins may serve as candidates. For example, the ability of DPSCs to transdifferentiate into OP was demonstrated by Askari et al. [170], who transfected DPSCs with the human oligodendrocyte transcription factor 2 (Olig2) gene. Olig2 encodes a helix–loop–helix transcription factor essential for lineage determination of oligodendrocytes and serves as an inducer for the oligodendrogenic pathway. Following differentiation, the resulting cells expressed several oligodendrogenic markers and were identified as OP cells. Notably, transplantation of these OPs into a mouse model of local sciatic nerve demyelination (induced by lysolecithin) resulted in active remyelination. These findings suggest that DPSCs can be programmed into oligodendrocyte progenitors after forced expression of Olig2, highlighting their potential as a simple and valuable source for cell therapy of neurodegenerative diseases.

3.3.10. Apoptosis

The apoptotic mechanism underlying the development of DPSCs remains unclear. To investigate this, knockdown experiments using an RNAi approach were performed. Knockdown of caspase-9 (CASP9) in DPSCs resulted in a significant reduction in apoptosis, which was associated with increased caspase-3 (CASP3) activity [171]. These results indicate that CASP9 and activated CASP3 predominantly regulate cell apoptosis in DPSCs.
miR-224-5p functions as an endogenous inhibitor of specific target genes and plays diverse roles in biological processes, including cancer cell proliferation, invasion, and metastasis [185]. In DPSCs, miR-224-5p protects cells against apoptosis by downregulating Ras-related C3 botulinum toxin substrate 1 (Rac1), a member of the Rac family of guanosine triphosphate phosphohydrolases [172]. Similar protective effects have been observed in H9C2 cardiomyocytes, a well-known immortalized rat embryonic heart-derived cell line. Li et al. [186] reported that miR-194-5p attenuated the accumulation of reactive oxygen species (ROS) and alleviated hypoxia/reoxygenation (H/R)-induced apoptosis in H9C2 cardiomyocytes.

3.3.11. Skeletal Myogenic Differentiation

miRs can promote myogenic differentiation of DPSCs. For example, Li et al. [173] first demonstrated that human adult DPSCs treated with 5-aza-2′-deoxycytidine exhibited myogenic differentiation in vitro, a process during which the expression of miR-135 and miR-143 was markedly decreased. miR-135 shows brain-specific expression and is involved in neuronal morphology, neural induction, and synaptic function [187], while miR-143 plays a role in the differentiation of stem cells into various lineages, including smooth muscle cells and adipocytes [188]. Based on these findings, Li et al. [173] co-transfected DPSCs with antisense oligonucleotides against miR-143 and miR-135. This treatment led to approximately half of the cells exhibiting distinct cardiomyocyte-like characteristics, with expression of myogenic markers such as myocyte enhancer factor 2C (MEF2C), myogenic differentiation 1 (MyoD), myogenin (MyoG), and myosin heavy chain (MyHC). These findings suggest that miRs play a decisive role in inducing the myogenic differentiation of DPSCs and provide new insights into their use for muscle regeneration therapies. Furthermore, miR-139-5p, which functions as a tumor suppressor by regulating gene expression and inhibiting cancer growth and spread [189], has also been implicated in myogenesis. Overexpression of miR-139-5p in DPSCs activated the Wnt/β-catenin signaling pathway, promoted cell growth, and induced skeletal myogenic differentiation [174]. Conversely, inhibition of miR-139-5p suppressed the Wnt/β-catenin signaling pathway, resulting in reduced cell growth and impaired skeletal myogenic differentiation in DPSCs.

3.3.12. Enhanced Pluripotency and Multilineage Differentiation Capability

OCT3/4 plays a critical role in early mammalian development by regulating the formation and maintenance of iPSCs/ESCs and the inner cell mass of the blastocyst. OCT3/4 exerts its effects by binding to DNA and often cooperates with other transcription factors, such as SOX2 and NANOG, to control gene expression and preserve the undifferentiated state of pluripotent cells [190]. It has also been reported to act as a pioneer transcription factor, with subsequent factors and environmental cues directing cell fate [191,192]. With regard to DPSCs, limited experimental evidence exists on their transcription factor-mediated reprogramming. Liu et al. [175] demonstrated that induced overexpression of OCT4A (the OCT3/4 isoform considered the key regulator of pluripotency) in DPSCs enhanced their differentiation capacity toward neural and glial lineages while also promoting the expression of other pluripotency regulators. These findings suggest a potential role for OCT4A in facilitating multipotency rather than strictly maintaining pluripotency. In other words, expression levels are critically important. Sufficient levels maintain the pluripotent state, while excessive overexpression can initiate differentiation into specific cell types.
Xeroderma pigmentosum complementation group C protein (XPC), a component of the DNA repair pathway, also interacts with OCT3/4 to modulate pluripotency. Liu et al. [176] demonstrated that overexpression of XPC in DPSCs led to significantly higher expression of the stemness markers OCT3/4, SOX2, and c-MYC. Furthermore, this treatment enhanced proliferation, reduced apoptosis, and improved multilineage differentiation, suggesting that XPC plays a crucial role in maintaining or enhancing the pluripotency of DPSCs.

3.4. Critical Evaluation of Gene-Engineering Strategies

Signaling pathways, such as Wnt/β-catenin, TGF-β/SMAD, and Notch, are critical regulators of DPSC behavior, coordinating self-renewal and differentiation for regenerative dentistry. This network of pathways controls the repair and regeneration of dentin and pulp tissue following injury or disease [193]. Gene-engineering strategies modulate these pathways to enhance the potential of DPSCs. For example, overexpression of SOX2 resulted in enhanced odontoblast differentiation with expression of odontoblast markers, such a DMP-1. This event is closely associated with the activation of the Wnt signaling pathway [128]. PDGF-BB, a homodimer of the PDGF-B protein, which is known to be involved in cell proliferation, tissue repair, and angiogenesis, caused enhanced DPSC proliferation and odontoblastic differentiation. This event is carried out through activation of the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) signaling pathway [99]. Overexpression of growth differentiation factor 15 (GDF15) caused enhanced osteogenic differentiation of DPSCs through activation of the TGF-β/SMAD signaling pathway, while knockdown of GDF15 produced the opposite effect [143].

4. Generation of Immature Cells by Forced Expression of Exogenous Genes in DPSCs

DPSCs are now known to be induced to generate iPSCs, as shown below. These cells can be obtained in large quantities in vitro, and many types of differentiated cells can subsequently be derived under specific induction conditions. In this context, iPSCs are widely considered a promising resource for regenerative medicine due to their ability to self-renew and differentiate into various cell types [4]. However, safety concerns are always associated with iPSCs when they are intended for use in regenerative medicine because iPSC-derived differentiated cells often include a mixture of immature cells (i.e., residual iPSCs) and differentiated cells. Notably, researchers have attempted to remove these immature cells after differentiation induction using chemicals, i.e., N-oleoyl serinol (S18), a ceramide analog [194], quercetin and YM155, inhibitors of survival [195], antibodies, such as keratan sulfate glycan-recognizing antibody [196], or a thymidine kinase-based cell ablation system [197], before utilizing them in regenerative medicine.

4.1. Generation of iPSCs

iPSCs can be generated from many types of dental cells, such as dental pulp, oral mucosa, gingiva, and periodontal ligament fibroblasts, making them a promising source for regenerative medicine in dentistry [198,199,200,201,202,203,204]. Tamaoki et al. [198] first attempted to isolate iPSC lines using DPSCs from young to old patients. They suggested that younger fibroblasts are more efficiently reprogrammed than older fibroblasts. Notably, non-juvenile cells, such as those from older adults (in which stem cells are not abundantly present), are generally more resistant to reprogramming with Yamanaka’s factors than juvenile cells because they have undergone more epigenetic changes associated with aging and lost part of their “reprogrammable” potential [205]. In agreement with this notion, Inada et al. [42] demonstrated that DPSCs with higher ALP activity tend to be more easily induced to generate iPSCs after transfection with Yamanaka’s factors, while DPSCs with no ALP activity failed to do so. Since ALP activity is closely associated with pluripotent cells, such as iPSCs and ESCs, DPSCs with higher ALP activity are believed to contain abundant stem cells. In other words, cells enriched with stem cells tend to be more easily induced to become iPSCs compared to those with fewer stem cells [206]. Supporting this hypothesis, repeated transfection of ALP-negative HDDPCs with Yamanaka’s factors resulted in increased endogenous ALP activity, together with acquisition of the ability to generate iPSCs [207]. These findings suggest that gene delivery of Yamanaka’s factors may increase the number of stem cells in ALP-negative cells, eventually leading to the generation of ALP-positive cells with the potential to form iPSCs.
Generally, human iPSCs and ESCs exhibit a more restricted differentiation capacity than their mouse counterparts, because they are derived from post-implantation epiblasts and represent a more committed, “primed” state of pluripotency [208]. These cells are called “epiblast stem cells (EpiSCs)” and have limited differentiation potential [209]. In contrast, mouse iPSCs and ESCs are referred to as “naïve stem cells” and have the ability to differentiate into various cells, including germ cells. Mouse iPSCs and ESCs (but not human counterparts) express a stem cell-specific antigen, stage-specific embryonic antigen-1 (SSEA-1), on their cell surface [210]. However, treatment of human iPSCs with drugs enabling conversion of EpiSCs to naïve stem cells for several days resulted in the generation of iPSCs with properties similar to naïve stem cells [211]. Concomitantly, the drug-treated human iPSCs began to express SSEA-1 [212]. Furthermore, these naïve stem cell-like human iPSCs exhibited broader differentiation potential, including the ability to form germ cells [212].

4.2. Generation of iTSCs from iPSCs

iTSCs are not fully reprogrammed to a pluripotent state like iPSCs but instead represent an intermediate stage between fully differentiated cells and pluripotent stem cells [4]. iTSCs have lost some of their original specialized functions but have not yet gained all the characteristics of pluripotency. They can be generated through partial reprogramming of various types of somatic cells, including hepatocytes, DPSCs, and pancreatic cells. To date, different types of iTSCs have been identified, including iTS-L [derived from liver cells (hepatocytes)] [213], iTS-P (derived from pancreatic cells) [214,215], and iTS-D (derived from DPSCs) [207]. iTSCs are considered to have potential applications in regenerative medicine due to their reduced tendency to form tumors (teratomas) compared to iPSCs [4,214]. More importantly, iTSCs express both stem cell and differentiated cell markers, as they are intermediate, partially reprogrammed cells. Furthermore, their marker profiles depend on the original tissue and the specific differentiation stage being captured. For example, iTS-P expresses stemness factors (Oct3/4, Sox2, Klf4, Esg-1, and Rex1) found in iPSCs/ESCs, together with Pdx1, which is not abundantly expressed in iPSCs/ESCs [214].
iTSCs can also be produced from iPSCs through cultivation under differentiation-inducing conditions. In this case, the state of human iPSCs [naïve stem cell or EpiSC state] is important for determining their fate. Kiyokawa et al. [216] demonstrated that EBs derived from naïve iPSCs—converted to a naïve state after pre-treatment with 2i (two small-molecule inhibitors, typically PD0325901 and CHIR99021)—were more efficiently induced to form pancreatic β-like cells after being subjected to an in vitro protocol compared with EBs derived from untreated iPSCs. These findings suggest that iTCS-P may be more abundantly produced from EBs derived from naïve iPSCs than from those derived from EpiSC-like iPSCs.

5. Usefulness of Feeder Cells to Maintain the Integrity of DPSCs

Feeder cells, exemplified by mitomycin-C (MMC)-treated mouse embryonic fibroblasts (MEFs), are mitotically inactivated (unable to divide) cells that provide essential nutrients, growth factors, and extracellular matrix components to support the survival and proliferation of target cells (such as stem cells) in a co-culture system. These feeder cells enable target cells to maintain their self-renewal and pluripotency by preventing spontaneous differentiation. They have been widely used for maintaining pluripotency in iPSCs and ESCs. Generally, culturing primary cells on plastic tissue culture dishes can often result in the loss of multipotency due to the inability of tissue-specific stem cells to survive in feeder-less conditions. Matrigel, a gelatinous protein mixture extracted from the Engelbreth–Holm–Swarm mouse sarcoma tumor, can be used as an alternative to feeder cells for maintaining stem cells [217], but this substance has significant limitations, including its undefined composition, variability between different production batches, and animal-derived origin. Notably, it was recently reported that culturing primary cells in medium containing feeder cells, particularly GM feeder cells expressing growth factors, may be beneficial for their survival and proliferation [218]. Ibanno et al. [219] demonstrated that GM human feeder cell-derived HDDPCs, engineered to express leukemia inhibitory factor (LIF), BMP4, and bFGF systemically, can be used to maintain the integrity of primary cultured HDDPCs. Co-culturing newly isolated HDDPCs with MMC-treated feeder cells results in enhanced cell proliferation of HDDPCs and a multipotent differentiation capability compared to HDDPCs cultured in the absence of feeder cells. Furthermore, HDDPCs co-cultivated with feeder cells continued to express stemness markers, while HDDPCs cultured in the absence of feeder cells failed to exhibit some of the stemness markers. These findings indicate that co-cultivation with feeder cells is beneficial for newly isolated HDDPCs to retain their stemness properties. However, preparing and maintaining feeder cells is a laborious task for researchers.
Notably, feeder cells can also remove contaminating bacteria (probably via phagocytosis) in a co-culture system. Saitoh et al. [220] demonstrated that STO feeder cells, which are also frequently used to maintain iPSCs and ESCs, are useful for the propagation of primarily cultured HDDPCs by eliminating contaminating bacteria and promoting cellular outgrowth.

6. Genome Editing

Periodontal disease and dental caries are common dental issues that can potentially be treated using the CRISPR/Cas9 genome editing system, a recently developed genetic engineering tool capable of cutting specific DNA sequences [221]. This system may enable personalized treatment and even the complete elimination of symptoms. The oral and craniofacial field harbors a wide range of diseases and developmental defects that require genetic models to fully exploit these genome editing techniques. For example, CRISPR/Cas9 can be used to disrupt dental-related gene functions or target Staphylococcus aureus (S. aureus) mutants, one of the major bacterial pathogens involved in tooth decay. Furthermore, genes associated with dental health—such as those involved in enamel formation or inflammatory response pathways—can be edited to reduce inflammation or genetic risk. However, the CRISPR/Cas9 system still presents challenges, particularly the risk of off-target effects, which may cause unintended gene expression and lead to occasional pathogenesis. Nonetheless, this technology represents an exciting advance that could revolutionize treatment strategies in dental health care [222,223]. In the following sections, recent reports on CRISPR/Cas9-based manipulation of virulent bacteria and genes relevant to dentistry are described.

6.1. CRISPR/Cas9-Based Manipulation of Bacterial Genes in S. aureus

Antibiotics target conserved bacterial cellular pathways or growth functions and, therefore, cannot selectively kill specific members of a complex microbial population. Bikard et al. [224] used the CRISPR/Cas9 system to destroy virulence-causing genes in pathogenic, but not non-pathogenic, S. aureus. Specifically, the Cas9/guide RNA (gRNA) complex targeted antibiotic resistance genes and successfully eliminated the staphylococcal plasmids carrying resistance determinants, thereby preventing the spread of plasmid-borne resistance. They also demonstrated that CRISPR/Cas9 antimicrobials function in vivo by killing S. aureus in a mouse skin colonization model. This technology opens opportunities to selectively manipulate complex bacterial populations as a new class of antimicrobials.
Gong et al. [225] developed a novel CRISPR/Cas9-based self-targeting gene editing strategy against glucosyltransferases (gtfs), major virulence factors involved in dental plaque biofilm. In bacteria, CRISPR/Cas9 relies on small RNAs (“spacers”) to restrict phage and plasmid infection, and it can also regulate endogenous gene expression using spacers targeting self-genes [226]. Gong et al. [225] designed self-targeting CRISPR arrays (with spacer sequences complementary to gtfB, which encodes glucosyltransferase B) and cloned them into plasmids. These plasmids were then transformed into S. mutans strain UA159—a bacterium isolated from a child with active caries and a well-recognized dental caries pathogen—as editing templates to generate desired mutants. As a result, this approach successfully edited gtfB and the gtfB-gtfC region, leading to disruption of biofilm formation.

6.2. CRISPR/Cas9-Based Manipulation of Genes Associated with Dental Health

Oxytocin (OT) is a neurohypophysial hormone that plays a role in lactation and parturition and exerts diverse biological actions via the OT receptor. Recently, OT has been suggested to promote bone formation by osteoblasts in osteoporosis. Kato and Yokose [227] hypothesized that OT can stimulate the differentiation of odontoblasts as well as osteoblasts. To test this, the Oxtr gene was knocked out by CRISPR/Cas9 in rat DPSCs. When intact rat DPSCs were treated with OT, mineralized nodule formation was induced along with increased mRNA expression of dentin sialoprotein and bone Gla protein. In contrast, Otr knockout (KO) cells showed inhibition of nodule formation and mRNA expression, which did not change after OT administration. These results suggest that OT can promote odontoblast-like cell differentiation, enhancing dentin formation, and that OT could be an important factor for dentinogenesis.
Family with sequence similarity 83 members H (Fam83h) is essential for dental enamel formation. Mutations in Fam83h cause human amelogenesis imperfecta (AI), an inherited disorder characterized by severe enamel hardness defects. Previous studies reported no enamel defects in Fam83h-KO/lacZ-knockin (KI) mice. Zhang et al. [228] generated genome-edited rabbits with a large deletion (900 bp) in the Fam83h gene using a dual single-guide RNA (sgRNA)-directed CRISPR/Cas9 system. The resulting Fam83h KO (Fam83h−/−) rabbits exhibited abnormal tooth mineralization and loose dentine. Additionally, they showed reduced hair follicle counts in dorsal skin, hair cycling dysfunction, and hair shaft differentiation. This novel Fam83h-KO rabbit provides a useful model for investigating Fam83h function and the pathogenic mechanisms of Fam83h mutations.
Hypodontia (dental agenesis) is a genetic disorder, and the mutation C175T in paired box 9 (PAX9) can cause hypodontia [229]. Liu et al. [229] investigated whether Cas9 nickase (nCas9)-mediated homology-directed repair (HDR) or the adenine base editor ABE8e could correct this PAX9 mutation. To deliver naked DNA efficiently into DPSCs, they used a chitosan hydrogel. ABEs convert A-T base pairs to G-C base pairs without requiring double-stranded DNA breaks or donor DNA templates and thus have therapeutic potential for genetic diseases [230]. ABE8e is a recently evolved ABE variant [231]. Liu et al. [229] first established DPSC lines stably carrying a PAX9 mutation and delivered either an HDR or ABE8e system into these cells. When gene correction efficiency was evaluated, ABE8e showed significantly higher efficiency in correcting C175T compared with HDR. Furthermore, corrected PAX9 enhanced DPSC viability and differentiation capacity into osteogenic and neurogenic lineages. This study demonstrates that base editor systems can be effective for correcting genes related to hypodontia in DPSCs.

7. Effect of DPSCs on Cancer Development

MSCs interact extensively with cancer cells as key components of the tumor microenvironment [232]. DPSCs and their derivatives, such as conditioned media and cell lysates, may exert anti-cancer effects by inhibiting tumor growth and promoting apoptosis in certain cancer types [233,234]. For example, He et al. [234] demonstrated that DPSC lysate significantly inhibited the growth, migration, and invasion of A549 lung cancer cells in vitro, and this effect was also confirmed in vivo. In tumor-bearing mice, treatment with DPSC lysate suppressed tumor growth and reduced tumor weight. Furthermore, the lysate increased the expression of pro-apoptotic proteins, such as CASP3 and BCL2-associated X (BAX), while decreasing the expression of anti-apoptotic protein BCL-2, thereby triggering the intrinsic, mitochondria-mediated apoptosis pathway. Based on these results, He et al. [234] concluded that DPSC lysate may serve as an alternative to cell-based therapy for lung cancer treatment. Nikkhah et al. [233] examined whether conditioned medium (CM) derived from DPSCs (DPSC-CM) could affect the growth and migration of colorectal cancer (CRC) cells. Their experiments demonstrated that DPSC-CM significantly reduced the viability and induced the apoptosis of CRC cells through the MAP kinase and apoptosis signaling pathways. Notably, the effect of DPSC-CM was concentration-dependent. Lower concentrations potentially promoted tumor cell proliferation, whereas higher concentrations inhibited it. These findings suggest that DPSCs may play a dual role in cancer development.

8. Exosomes and Cytokines Secreted from DPSCs

Stem cells secrete a complex mixture of factors called the secretome, which includes exosomes and cytokines that play key roles in regenerative therapy. Exosomes are tiny vesicles that transport a cargo of proteins, lipids, and nucleic acids (such as microRNAs) between cells, thereby mediating much of the regenerative activity of stem cells [235]. Cytokines are signaling proteins, such as growth factors, chemokines, and anti-inflammatory mediators, that act as messengers between cells to regulate immune responses and tissue repair [236]. The secretions from stem cells, particularly exosomes and cytokines, are powerful modulators of wound healing and tissue repair. They can regulate immune activity, stimulate the growth of new blood vessels (angiogenesis), and promote cell survival and regeneration [237]. Based on these findings, “cell-free” therapies that utilize stem cell–derived exosomes are now considered a promising and potentially safer alternative to whole stem cells for regenerative medicine [238]. For example, Zhang et al. [239] conducted a systematic review of the role of stem cell-derived exosomes in the repair of spinal cord injury (SCI) using animal experiments. Their results indicated that stem cell-derived exosomes significantly improved motor function in animals with SCI and increased the expression of anti-inflammatory cytokines IL-4 and IL-10, as well as anti-apoptotic protein BCL-2. In contrast, they significantly lowered the levels of pro-inflammatory cytokines IL-1β and TNF-α, as well as the expression of the pro-apoptotic protein BAX. However, the mechanism of exosome-mediated SCI repair, as well as the best source and dosage, remains unknown. Therefore, future research needs to standardize animal study protocols for consistency and explore the most effective strategies for exosome-based therapy in SCI. Key priorities include establishing unified protocols for isolating and purifying exosomes, improving methods for characterizing and producing them at scale, and designing more robust animal studies to better understand the optimal delivery, dosage, and mechanisms of action.

9. Immunomodulatory Effects of DPSCs

To date, MSCs have been shown to exhibit immunomodulatory abilities through the expression of enzymes, the production of soluble factors, and direct cell-to-cell contacts to influence both innate and adaptive immune responses [240]. In other words, MSCs suppress various immune cells, including T cells, B cells, and macrophages, and promote a balanced, less inflammatory response [241]. These immunomodulatory abilities are also seen when human DPSCs are co-cultivated with immunogenic cells. In other words, they exhibit immunomodulatory abilities by suppressing the immune response [242]. For example, co-culturing of DPSCs and phytohemagglutinin (PHA)-activated T cells inhibited T cell proliferation and induced T cell apoptosis and the formation of regulatory T cells (Tregs), which play a central role in immune homeostasis [243,244]. DPSCs suppressed the proliferation of concanavalin A (Con A)-treated peripheral blood mononuclear cells (PBMCs), which are immune cells stimulated with Con A, in co-cultures using transwells [245]. This finding suggests that DPSCs also achieve immunomodulatory effects via the expression of soluble factors that regulate the functions of immune cells. Indeed, it was reported that DPSCs upregulated the secretion of anti-inflammatory factors, such as IL-6, TGF-β, HGF, and IL-10, in T cells, whereas they downregulated the production of pro-inflammatory factors, such as IL-2, IL-12, and TNF-α [244].

10. Application of Engineered DPSCs in Regenerative Medicine

Human DPSC-based therapies using cells (including gene-engineered cells) or organoid-like 3D structures have been extensively assessed in experimental animal models as basic or pre-clinical studies for regenerative medicine [14]. For cell-based therapy, the route of cell injection is important. Several routes have been reported, including intravenous administration, intra-brain injection, transplantation into the hindlimb skeletal muscle [246], and direct injection of cells (or cells encapsulated with a scaffold) into the affected sites [247]. In cases involving transplantation of larger cell aggregates, such as organoids, direct placement of these structures onto the affected sites may be appropriate. In the following sections, more detailed explanations of these approaches will be provided.

10.1. Cell-Based Therapy Using Intact or Engineered DPSCs

DPSCs represent a promising source for cell-based therapies owing to their easy, minimally invasive surgical access, and high proliferative capacity. Research has focused on the ability of DPSCs to treat diseases using animal models. For example, El-Kersh et al. [247] first demonstrated the therapeutic potential of intravenous (IV) and intrapancreatic (IP) transplantation of human DPSCs in a rat model of STZ-induced type 1 diabetes (T1D). IP transplantation was performed by slowly injecting the cell suspension under the pancreas capsule into 10 distinct pancreatic sites using an insulin syringe with a 29-G needle. DPSCs (106 cells) labeled with the membrane-bound fluorescent marker PKH26 were injected into either the pancreas (IP) or tail vein (IV) 7 days after STZ injection, and the rats were examined about one month later. Both IV and IP transplantation of DPSCs reduced blood glucose and increased the levels of rat and human serum insulin and C-peptide. The engrafted cells survived in the STZ-injured pancreas. Islet-like clusters and scattered insulin-expressing human DPSCs, identified by anti-insulin staining and PKH26 labeling, were detected in the pancreas of diabetic rats. However, further clarification is needed regarding the possibility of an immunogenic response to the injected human cells and the transdifferentiation of human DPSCs into IPCs. El-Kersh et al. [247] concluded that DPSCs may have therapeutic potential to treat patients with long-term T1D. A similar experiment was also reported by Inada et al. [248], who generated IPCs by treating DPSCs with hydrogen sulfide in a 3D organ cultivation system and subsequently transplanted them into rat models for T1D. On the other hand, Hata et al. [246] demonstrated the usefulness of transplantation of human DPSCs into the hindlimb skeletal muscle of diabetic nude mice for the treatment of diabetic polyneuropathy. DPSC transplantation significantly reduced sensory perception thresholds, delayed nerve conduction velocity, and decreased blood flow to the sciatic nerve in diabetic mice 4 weeks post-transplantation. A subset of the transplanted DPSCs localized around the muscle bundles and expressed the human VEGF and NGF genes at the transplanted site. These results suggest that DPSC transplantation may be efficacious in treating diabetic polyneuropathy via secretion of DPSC-derived factors, such as VEGF and NGF.
Mead et al. [249] examined the therapeutic benefit of transplanting DPSCs into the vitreous body of the eye after a surgically induced optic nerve crush injury in rats. Intravitreal transplantation of DPSCs promoted neuroprotection and axon regeneration of retinal ganglion cells after optic nerve injury. Sowa et al. [250] examined the ability of HGF to provide neuroprotective effects against ischemia-induced injuries. They transplanted HGF-overexpressing DPSCs into the brains of rats following transient middle cerebral artery occlusion to determine whether the transplanted cells could attenuate brain damage associated with post-ischemia/reperfusion injury. The gene-engineered DPSCs significantly inhibited microglial activation and pro-inflammatory cytokine production, along with the suppression of neuronal degeneration. These findings suggest that the administration of HGF-overexpressing DPSCs may be useful for preventing neuronal damage in the acute phase of stroke.

10.2. Cell-Based Dental Stem Cell Therapy Using Intact DPSCs

Gronthos et al. [11] first demonstrated that DPSCs can regenerate tooth structures by transplanting them in conjunction with hydroxyapatite/tricalcium phosphate (HA/TCP) powder into immunocompromised mice. After 1.5 months post-implantation, the resulting 3D structures contained a dentin–pulp complex, expressing the dentin-specific protein DSPP and forming tubular structures within newly generated dentin, suggesting the presence of progenitor cells in DPSCs. To improve functional effectiveness in pulp-like tissue formation, specific growth factors and scaffold materials are required. For example, when DPSCs were embedded into 3D-printed hydroxyapatite scaffolds and transplanted into immunocompromised mice, vascularized pulp-like tissue was successfully obtained [251], highlighting the angiogenic potential of DPSCs. Alternatively, pulp-like tissues with rich vascularization can be generated in vivo without using scaffolds. In this approach, scaffold-free sheet-like aggregates composed of DPSCs are combined with a thermo-responsive hydrogel and transplanted subcutaneously into immunocompromised mice. After 1.5 months post-implantation, the resulting grafts developed pulp-like tissues with extensive blood vessel networks [238,252]. Notably, this method does not require additional growth factors or scaffolds.

10.3. Cell-Based Dental Stem Cell Therapy Using iPSC-Derived Dental Cells for Dentin–Pulp Complex Regeneration

iPSCs/ESCs have the potential to generate various types of differentiated cells, including neurons, adipocytes, and pancreatic β cells, as well as dental cells, such as odontoblasts and osteogenic cells. In this section, we focus on the successful production of dental cells from iPSCs/ESCs. Unlike other organs with some regenerative potential, dental tissues—particularly dental enamel—have very limited regenerative ability. To address this limitation, fabrication of dentin–pulp-like organoids using iPSC-derived cells has been explored as a strategy to regenerate damaged dental tissues. This approach is referred to as dentin–pulp complex regeneration [253]. Dentin–pulp complex regeneration requires the formation of 3D organoids, which can self-develop when seeded in an extracellular matrix (ECM)—such as Matrigel, GelMA (gelatin methacryloyl, a modified biopolymer derived from gelatin), collagen, agar gel (a natural polysaccharide polymer), or synthetic polymers, like poly(lactic-co-glycolic acid) (PLGA)—that mimics scaffolds, followed by culture in medium supplemented with growth factors. Under appropriate conditions, these organoids have been shown to contain the desired differentiated cells, similar to those in the tissue of origin. A key advantage of organoids is their ability to be serially expanded (passaged) without loss of phenotype or regenerative potential, offering a powerful tool that overcomes the limitations of cells grown on flat surfaces.
In humans, tooth development requires the interplay between epithelial and mesenchymal cells. However, studies on tooth development remain limited, as epithelial stem cells are relatively difficult to obtain and maintain. iPSCs/ESCs may be an alternative source of epithelial cells. Notably, both dental epithelial cells (DECs), derived from the ectoderm, and dental mesenchymal cells (DMCs), derived from neural crest cells, have been differentiated from iPSCs [254]. Kim et al. [255,256] established protocols for isolating DECs and DMCs through cultivation of iPSCs-derived EBs in media-specific growth factors. For iPSC-derived DECs, EBs were induced into ectodermal cells by cultivation in DMEM/F12 containing N2, BMP4, and RA. After 4 days, the medium was replaced with keratinocyte serum-free medium (K-SFM; ThermoFischer Scientific) supplemented with NOGGIN and epidermal growth factor (EGF). After another 4 days, cells were cultured in K-SFM supplemented with BMP4 and EGF. For DMC differentiation, EBs were induced into neural crest cells using DMEM/F12 supplemented with N2, B27, insulin, bFGF, and EGF. To develop bioengineered teeth using DECs and DMCs derived from human iPSCs, these differentiated cells were recombined with scaffolds, such as GelMA, collagen, and agar gel, which supported induction into osteogenic, chondro-osteogenic, and odontogenic lineages during the mineralization stage (Figure 3). These cell aggregates, encapsulated with biomaterials, were transplanted beneath the kidney capsule of mice for further maturation over 16 weeks. Kim et al. [257] also reported a novel differentiation method using a transwell membrane co-culture system to isolate DECs and DMCs from iPSCs. Additionally, DEC-like stem cells can be obtained through direct co-culture of human iPSCs with Hertwig’s epithelial root sheath/epithelial rests of the Malassez (HERS/ERM) cell line [258].
Notably, DECs can potentially regenerate dental enamel by redirecting their growth to differentiate into ameloblasts. Miao et al. [259] demonstrated that overexpression of Epiprofin (Epfn), a transcription factor specifically expressed in the dental epithelium in mouse iPSCs, led to the generation of ameloblasts, which are usually lost after tooth eruption. The presence of ameloblasts was characterized by positive staining for keratin 14 and amelogenin and Alizarin Red S staining. These results suggest that Epfn is a promising target for inducing ameloblast differentiation, which can be applied in enamel and tooth regeneration. Hemeryck et al. [260] developed a novel epithelial organoid model (enriched with cells exhibiting a tooth epithelial stemness phenotype) derived from human tooth to regenerate dental enamel. These organoids were capable of undergoing an ameloblast differentiation process, which was further enhanced by TGFβ and inhibited by TGFβ receptor blockade, thereby reproducing TGFβ’s established role in amelogenesis. Kim et al. [261] attempted to generate iPSC-derived ameloblast organoids (AOs) using a 3D culture system. The resulting AOs had similar properties to ameloblasts, forming enamel in response to calcium and undergoing mineralization through interactions with the dental mesenchyme. AOs also retained osteogenic and odontogenic differentiation potential. Furthermore, AOs demonstrated regenerative potential when combined with mouse dental mesenchyme. These findings highlight the promise of AOs not only for tooth regeneration but also for studying dental diseases that currently lack effective treatments.
Dentin–pulp complex regeneration can also be achieved in vivo. This approach focuses on injecting stem cells into disinfected root canals with an apical opening. In this case, scaffolds are required to prevent cell migration to surrounding tissues [262]. For example, dos Santos et al. [263] performed in vivo dentin–pulp complex regeneration in immunocompromised rats. After pulpectomy and canal preparation, the teeth received either DPSCs embedded in scaffolds or PBS as a control, and the cavities were sealed. After 12 weeks, the animals were euthanized, and the specimens underwent histological evaluation for intracanal connective tissue, odontoblast-like cells, intracanal mineralized tissue, and periapical inflammatory infiltrate. Transplantation of human DPSCs promoted partial pulp tissue neoformation in adult rat molars, with remnants of mineralized tissue and DMP-1-positive odontoblast-like cells.

11. Conclusions and Challenges

DPSCs contain multipotent stem cells with the potential for self-renewal, multilineage differentiation, and immunomodulatory functions. One of the intriguing aspects of making the use of DPSCs more attractive is their easy access from the discarded third molar tooth and minimal ethical concerns during procurement. In this context, these stem cells are more convenient than BMSCs. More importantly, DPSCs do not form tumors, and the use of autologous cells reduces the chances of rejection. For these reasons, DPSCs are considered a promising cell source for regenerative medicine applications, and several achievements have been made in their study. However, several challenges remain to be overcome. For example, DPSCs comprise a mixture of different cell types, which remains a key concern. Single-cell cloning or FACS-based cell sorting is sometimes needed, but these approaches are difficult due to the lack of standardized culture protocols, including optimal media and substrates. Production of immortalized DPSCs may be a promising approach to overcome these issues. The use of feeder cells may also help avoid the potential loss of DPSC integrity and their multilineage differentiation ability during continuous cultivation [219]. SCIP is another attractive method for obtaining a large number of cells through a one-step cultivation process of isolated primary DPSCs, as the cells show no overt alteration in their properties, even after the 10th cell passage [31].
The usefulness of cell-based therapy with human DPSCs has been highlighted by El-Kersh et al. [247], who first performed intravenous and intrapancreatic transplantation of human DPSCs in a rat model for T1D. Both approaches reduced blood glucose levels and increased the levels of rat and human serum insulin 28 days after STZ injection. Furthermore, islet-like clusters and scattered insulin-expressing human DPSCs were detected in the pancreas of diabetic rats. These findings suggest that transplanted human DPSCs may have been converted into functional IPCs, although the mechanism underlying this event remains to be elucidated. Concerns remain regarding how long the grafted cells can survive and continue secreting insulin and how strong the immunogenic response to the human cells may be since human cells were introduced into rats. In this context, more pre-clinical trials are still needed to establish the safety and efficacy of DPSC-based interventions in regenerative medicine.
DPSCs can be induced to differentiate into appropriate specific cell types under appropriate induction conditions. These include odontoblasts, osteoblasts, adipocytes, neuronal cells, hepatocytes, endothelial cells, cardiac cells, pancreatic β cells, and bladder smooth muscles, as shown in Table 2. Gene delivery can further enhance the multilineage differentiation ability of DPSCs, which is one of the promising strategies for future regenerative medicine. For example, as shown in Table 3, enhanced differentiation into odontoblasts, osteoblasts, cell proliferation, angio-/vasculogenic commitment, neurogenic commitment, adipogenic commitment, cell migration, enhanced production towards IPCs, and skeletal myogenic differentiation have all been achieved. Gene delivery has also been associated with effects on cell apoptosis and inflammation in DPSCs. Transplanting IPCs derived from the gene-engineered DPSCs into rat models of T1D has the potential to improve the diabetic phenotype. However, as with direct in vivo transplantation of DPSCs or their derivatives, challenges remain, including variability in IPC differentiation methods, the risk of immunogenic responses, and the need for long-term studies to ensure stable and effective outcomes.
iPSCs and their derivatives (i.e., iTSC and differentiated cells) generated from DPSCs are also promising resources for regenerative medicine, although their tumorigenic potential remains a concern. The main advantage of using iPSCs and their derivatives is the feasibility of obtaining these cells in large quantities. Moving toward clinical application, dentin–pulp complex regeneration is currently the most advanced application of dental stem cells. At present, both DECs and DMCs can be derived from iPSCs by inducing their differentiation with specific growth factors and chemicals [256] or using culture conditions that mimic natural biological processes, such as transwell cultivation methods [257]. The development of scaffolds is also critical for dentin–pulp complex regeneration, as they provide the structural support and environment necessary for cell adhesion, proliferation, and differentiation. Scaffolds can be derived from natural materials, such as collagen, agar, and Matrigel, or from synthetic polymers, such as PLGA.
In summary, DPSCs represent a valuable alternative to MSCs for regenerative medicine because of their easy access and minimal ethical concerns. Gene-engineered DPSCs and their derivatives (i.e., iPSCs and differentiated progeny) are also promising candidates for future cell-based therapies. Recent advances in genome editing technologies and 3D organ cultivation systems offer new opportunities for this field, including oral regeneration, by enabling more precise genetic corrections and the creation of complex, functional tissues.

Author Contributions

M.S. and E.I. designed and drafted the manuscript; I.S., M.T., Y.K., N.K., H.Y., K.M., and S.N. critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partly supported by a grant (no. 19K06372 to M.S.; no. 24K13200 to E.I.; no. 22H03277 to I.S.; no. 23K09441 to N.K.; no. 23K27097 to S.N.) from the Ministry of Education, Science, Sports, and Culture, Japan.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

We thank Kazusa Inada for her support with the in-house drawing of the figures. The language of the manuscript was partly edited by an external language editing service-providing company.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ABEsAdenine base editors
ADGRA2Adhesion G protein-coupled receptor A2
ADMAdrenomedullin
AFPAlpha-fetoprotein
AIAmelogenesis imperfecta
AKTProtein kinase B
ALCAMActivated leucocyte cell adhesion molecule
ALPAlkaline phosphatase
AMPKα1Catalytic alpha 1 subunit of AMP-activated protein kinase (AMPK)
AMY2AAmylase-2a
ANTXR1Anthrax toxin receptor 1
AOAmeloblast organoid
α-SMAAlpha smooth muscle actin
ATP8B1ATPase phospholipid transporting 8B1
BAXBCL2-associated X
BCL-2B cell lymphoma-2
BDNFBrain-derived neurotrophic factor
BETBetaine
bFGFBasic fibroblast growth factor
BMP4Bone morphogenetic protein 4
BMSCsBone marrow stem cells
CASP3Caspase-3
CASP9Caspase-9
CGFeConcentrated growth factor exudate
CMConditioned medium
CNNCalponin
CNSCentral nervous system
COL1Collagen type I
COL1A2Collagen type I alpha 2 chain
COL3A1Collagen type III alpha 1 chain
Con AConcanavalin A
CPSCarbamoyl phosphate synthetase
CRCColorectal cancer
Cx43Connexin 43
DDIT3DNA damage-inducible transcript 3
DECsDental epithelial cells
DESDesmin
DFSCsDental follicle stem cells
DLL1Notch ligand Delta1
DMCsDental mesenchymal cells
DMEMDulbecco’s modified Eagle’s medium
DMP-1Dentin matrix protein-1
DPCsDental pulp cells
DPSCsDental pulp stem cells
DPSC-EDDPSC from the enzymatic digestion of DP tissue
DPSC-OGDPSC obtained through the outgrowth of tissue explants
DSPDesmoplakin
DSPPDentinal sialophosphoprotein
E8Serum-free essential 8 medium
EBsEmbryoid bodies
ECMExtracellular matrix
EFNB2EphrinB2
EGFEpidermal growth factor
EpfnEpiprofin
EpiSCsEpi-stem cells
ERKExtracellular signal-regulated kinase
ESCsEmbryonic stem cells
ET-1Endothelin-1
ETV2Ets variant transcription factor 2
FABP4Fatty acid-binding protein 4
FACSFluorescence-activated cell sorting
Fam83hFamily with sequence similarity 83 members H
FBSFetal bovine serum
Flk-1Functioning legal knowledge 1
GALCGalactosyl–ceramidase
GAP43Growth-associated protein 43
GCGGlucagon
Gdf11Growth/differentiation factor 11
GDF15Growth differentiation factor 15
GDNFGlial cell-derived neurotrophic factor
GelMAGelatin methacryloyl
GFAPGlial fibrillary acid protein
GING SCsGingival stem cells
GLUT4Glucose transporter type 4
GMGenetically modified
GMPGood manufacturing practice
gtfsGlucosyltransferases
GOIGene of interest
gRNAGuide RNA
HDDPCHuman deciduous teeth-derived DPCs
HDRHomology-directed repair
HERS/ERMHertwig’s epithelial root sheath/epithelial rests of Malassez
HGFHepatocyte growth factor
HIF-1Hypoxia-inducible factor-1
HIF-1αHypoxia-inducible factor-1α
H/RHypoxia and reoxygenation
HA/TCPHydroxyapatite/tricalcium phosphate
HHEXHematopoietically expressed homeobox
HNF4αHepatic nuclear factor-4 alpha
HPV16E6/E7 proteins from human papillomavirus 16
HSHuman serum
hTERTHuman telomerase reverse transcriptase protein
HUVECsHuman umbilical vein endothelial cells
IBMX3-isobutyl-1-methylxanthine
ICAM1Intercellular adhesion molecule-1
ICCsIslet-like cell clusters
ID1Inhibitor of DNA binding 1
IFN-γInterferon-gamma
IGF-1Insulin-like growth factor-1
IGFBP3Insulin-like growth factor binding protein-3
IKKκB kinase
ILInterleukin
IL-8Interleukin-8
IMDMIscove’s modified Dulbecco’s medium
IPIntrapancreatic
IPCInsulin-producing cell
iPSCsInduced pluripotent stem cells
i-TSCsInduced tissue-specific stem cells
IVIntravenous
JAG-1Notch ligand, Jagged-1
KDRKinase insert domain-containing receptor
KIKnockin
KLF4Krüppel-like factor 4
KOKnockout
LIFLeukemia inhibitory factor
LncRNA H19Long noncoding RNA H19
LPLLipoprotein lipase
LPSLipopolysaccharide
MafaMAF bZIP transcription factor A
MAP2Microtubule-associated protein 2
MCP-1Monocyte chemotactic protein-1
MEF2CMyocyte enhancer factor 2C
MEFsMouse embryonic fibroblasts
MEK1Mitogen-activated protein kinase kinase 1
MEPEMatrix extracellular phosphoglycoprotein
MESPMesoderm posterior BHLH transcription factor 1
miRMicroRNA
MMCMitomycin-C
MNX1Motor neuron and pancreas homeobox 1
MSI1Musashi1
MYH6Myosin heavy chain 6
MyHCMyosin heavy chain
MyoDMyogenic differentiation 1
MyoGMyogenin
nCas9Cas9 nickase
MCAMMelanoma cell adhesion molecule
NCAMNeural cell adhesion molecule
NEFHNeurofilament heavy chain
NESNestin
NeuNNeuronal nuclei
NF-κBNuclear factor kappa B
NGFNerve growth factor
NGFRNerve growth factor receptor
Ngn3Neurogenin 3
Nkx2.5NK-2 transcription factor-related, locus 5
NKX6-1NK6 homeobox 1
NOTCH1Notch1
NSCsNeuronal stem cells
OCNOsteocalcin
OCT3/4Octamer-binding transcription factor 3/4
Olig2Oligodendrocyte transcription factor 2
ONOsteonectin
OPOligodendrocyte progenitors
OPNOsteopontin
OSXOsterix
OTOxytocin
OXTROxytocin receptor
PA-CMPreameloblast-conditioned medium
PAI-1Plasminogen activator inhibitor-1
PAX4Paired box 4
PAX6Paired box 6
PAX9Paired box 9
PDGF-BBPlatelet-derived growth factor BB
PDLSCsPeriodontal ligament stem cells
PDX1Pancreatic and duodenal homeobox 1
PECAM-1Platelet/endothelial cell adhesion molecule-1
PHAPhytohemagglutinin
PI3KPhosphatidylinositol 3 kinase
PIN1Peptidylprolyl cis/trans isomerase, NIMA-interacting 1
PLPlatelet lysate
PLGAPoly(lactic-co-glycolic acid)
PPAR-γPeroxisome proliferator-activated receptor γ
PPYPancreatic polypeptide
RARetinoic acid
Rac1Ras-related C3 botulinum toxin substrate 1
RNAiRNA interference
RNA-seqRNA sequencing
RNA-snMIFxCRNA analysis based on a small number of manually isolated fixed cells
ROSReactive oxygen species
RUNX2Runt-related transcription factor 2
S18N-oleoyl serinol
S100S100 calcium-binding proteins
S100BS100 calcium-binding protein B
SCAPStem cell apical papilla
SCISpinal cord injury
SCIPScratch-based isolation of primary cells from human dental pulps
scRNA-seqSingle-cell-based RNA-sequencing
SHEDStem cells from human exfoliated deciduous teeth
SDF-1αStromal cell-derived factor-1α
SEMA3ASemaphorin 3A
SERPINE1Serpin family E member 1
SMCsSmooth muscle cells
SORT1Sortilin 1
SOX2SRY-related HMG-box 2
SRGNSerglycin
SSEA-1Stage-specific embryonic antigen-1
SSTSomatostatin
STZStreptozotocin
SV40TSimian virus 40 large T antigen
SYN1Synapsin I
SYPSynaptophysin
T1DType 1 diabetes
TET2Ten-eleven-translocation 2
TGFβTransforming growth factor beta
TGF-β1Transforming growth factor beta 1
TIMP-1Tissue inhibitor of metalloproteinase-1
TNF-αTumor necrosis factor-alpha
TUBB3β-III tubulin
uPAUrokinase-type plasminogen activator
VEGFVascular endothelial growth factor
VEGFR2VEGF receptor 2
VIMVimentin
VWFvon-Willebrand factor
WNT4Wnt family member 4
WTAWhole transcriptome amplification
XFMXenogeneic serum-free culture medium
XPCXeroderma pigmentosum complementation group C protein
ZBTB20Zinc finger and BTB domain-containing 20
Zfp521Zinc finger protein 521

References

  1. Kim, K.; Evans, G. Tissue engineering: The future of stem cells. Top. Tissue Eng. 2005, 2, 1–21. [Google Scholar]
  2. Herberts, C.A.; Kwa, M.S.; Hermsen, H.P. Risk factors in the development of stem cell therapy. J. Transl. Med. 2011, 9, 29. [Google Scholar] [CrossRef]
  3. Egusa, H.; Sonoyama, W.; Nishimura, M.; Atsuta, I.; Akiyama, K. Stem cells in dentistry--part I: Stem cell sources. J. Prosthodont. Res. 2012, 56, 151–165. [Google Scholar] [CrossRef] [PubMed]
  4. Saitoh, I.; Sato, M.; Kiyokawa, Y.; Inada, E.; Iwase, Y.; Ibano, N.; Noguchi, H. Induced tissue-specific stem cells (iTSCs): Their generation and possible use in regenerative medicine. Pharmaceutics 2021, 13, 780. [Google Scholar] [CrossRef] [PubMed]
  5. Via, A.G.; Frizziero, A.; Oliva, F. Biological properties of mesenchymal stem cells from different sources. Muscles Ligaments Tendons J. 2012, 2, 154–162. [Google Scholar] [PubMed]
  6. Chu, D.T.; Phuong, T.N.T.; Tien, N.L.B.; Tran, D.K.; Thanh, V.V.; Quang, T.L.; Truong, D.T.; Pham, V.H.; Ngoc, V.T.N.; Chu-Dinh, T.; et al. An update on the progress of isolation, culture, storage, and clinical application of human bone marrow mesenchymal stem/stromal cells. Int. J. Mol. Sci. 2020, 21, 708. [Google Scholar] [CrossRef]
  7. Fujii, S.; Fujimoto, K.; Goto, N.; Abiko, Y.; Imaoka, A.; Shao, J.; Kitayama, K.; Kanawa, M.; Sosiawan, A.; Suardita, K.; et al. Characterization of human dental pulp cells grown in chemically defined serum-free medium. Biomed. Rep. 2018, 8, 350–358. [Google Scholar] [CrossRef]
  8. Aurrekoetxea, M.; Garcia-Gallastegui, P.; Irastorza, I.; Luzuriaga, J.; Uribe-Etxebarria, V.; Unda, F.; Ibarretxe, G. Dental pulp stem cells as a multifaceted tool for bioengineering and the regeneration of craniomaxillofacial tissues. Front. Physiol. 2015, 6, 289. [Google Scholar] [CrossRef]
  9. Nuti, N.; Corallo, C.; Chan, B.M.; Ferrari, M.; Gerami-Naini, B. Multipotent differentiation of human dental pulp stem cells: A literature review. Stem Cell Rev. Rep. 2016, 12, 511–523. [Google Scholar] [CrossRef]
  10. Ledesma-Martínez, E.; Mendoza-Núñez, V.M.; Santiago-Osorio, E. Mesenchymal stem cells derived from dental pulp: A review. Stem Cells Int. 2016, 2016, 4709572. [Google Scholar] [CrossRef]
  11. 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]
  12. Mayo, V.; Sawatari, Y.; Huang, C.Y.; Garcia-Godoy, F. Neural crest-derived dental stem cells--where we are and where we are going. J. Dent. 2014, 42, 1043–1051. [Google Scholar] [CrossRef]
  13. Sonoda, S.; Tomoda, E.; Tanaka, Y.; Yamaza, T. Properties and possibilities of human dental pulp-derived stem cells. Arch. Stem Cell Res. 2015, 2, 1012. [Google Scholar] [CrossRef]
  14. Gimble, J.M.; Guilak, F.; Nuttall, M.E.; Sathishkumar, S.; Vidal, M.; Bunnell, B.A. In vitro differentiation potential of mesenchymal stem cells. Transfus. Med. Hemother. 2008, 35, 228–238. [Google Scholar] [CrossRef] [PubMed]
  15. Ponnaiyan, D.; Jegadeesan, V. Comparison of phenotype and differentiation marker gene expression profiles in human dental pulp and bone marrow mesenchymal stem cells. Eur. J. Dent. 2014, 8, 307–313. [Google Scholar] [CrossRef]
  16. Kim, S.H.; Kim, Y.S.; Lee, S.Y.; Kim, K.H.; Lee, Y.M.; Kim, W.K.; Lee, Y.K. Gene expression profile in mesenchymal stem cells derived from dental tissues and bone marrow. J. Periodontal Implant. Sci. 2011, 41, 192–200. [Google Scholar] [CrossRef] [PubMed]
  17. Kumar, A.; Kumar, V.; Rattan, V.; Jha, V.; Bhattacharyya, S. Secretome cues modulate the neurogenic potential of bone marrow and dental stem cells. Mol. Neurobiol. 2017, 54, 4672–4682. [Google Scholar] [CrossRef]
  18. 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]
  19. Tamaki, Y.; Nakahara, T.; Ishikawa, H.; Sato, S. In vitro analysis of mesenchymal stem cells derived from human teeth and bone marrow. Odontology 2013, 101, 121–132. [Google Scholar] [CrossRef]
  20. Yu, J.; Wang, Y.; Deng, Z.; Tang, L.; Li, Y.; Shi, J.; Jin, Y. Odontogenic capability: Bone marrow stromal stem cells versus dental pulp stem cells. Biol. Cell 2007, 99, 465–474. [Google Scholar] [CrossRef]
  21. Xu, Y.; Li, Z. CRISPR-Cas systems: Overview, innovations and applications in human disease research and gene therapy. Comput. Struct. Biotechnol. J. 2020, 18, 2401–2415. [Google Scholar] [CrossRef]
  22. Seo, B.M.; Miura, M.; Gronthos, S.; Bartold, P.M.; Batouli, S.; Brahim, J.; Young, M.; Robey, P.G.; Wang, C.Y.; Shi, S. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004, 364, 149–155. [Google Scholar] [CrossRef]
  23. Morsczeck, C.; Götz, W.; Schierholz, J.; Zeilhofer, F.; Kühn, U.; Möhl, C.; Sippel, C.; Hoffmann, K.H. Isolation of precursor cells (PCs) from human dental follicle of wisdom teeth. Matrix Biol. 2005, 24, 155–165. [Google Scholar] [CrossRef]
  24. Sonoyama, W.; Liu, Y.; Fang, D.; Yamaza, T.; Seo, B.M.; Zhang, C.; Liu, H.; Gronthos, S.; Wang, C.Y.; Wang, S.; et al. Mesenchymal stem cell-mediated functional tooth regeneration in swine. PLoS ONE 2006, 1, e79. [Google Scholar] [CrossRef]
  25. 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]
  26. Inada, E.; Saitoh, I.; Watanabe, S.; Aoki, R.; Miura, H.; Ohtsuka, M.; Murakami, T.; Sawami, T.; Yamasaki, Y.; Sato, M. PiggyBac transposon-mediated gene delivery efficiently generates stable transfectants derived from cultured primary human deciduous tooth dental pulp cells (HDDPCs) and HDDPC-derived iPS cells. Int. J. Oral Sci. 2015, 7, 144–154. [Google Scholar] [CrossRef] [PubMed]
  27. Mitrano, T.I.; Grob, M.S.; Carrión, 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]
  28. Megat Abdul Wahab, R.; Mohamed Rozali, N.A.; Senafi, S.; Zainol Abidin, I.Z.; Zainal Ariffin, Z.; Zainal Ariffin, S.H. Impact of isolation method on doubling time and the quality of chondrocyte and osteoblast differentiated from murine dental pulp stem cells. PeerJ 2017, 5, e3180. [Google Scholar] [CrossRef] [PubMed]
  29. Rodas-Junco, B.A.; Villicaña, C. Dental pulp stem cells: Current advances in isolation, expansion and preservation. Tissue Eng. Regen. Med. 2017, 14, 333–347. [Google Scholar] [CrossRef]
  30. Hilkens, P.; Gervois, P.; Fanton, Y.; Vanormelingen, J.; Martens, W.; Struys, T.; Politis, C.; Lambrichts, I.; Bronckaers, A. Effect of isolation methodology on stem cell properties and multilineage differentiation potential of human dental pulp stem cells. Cell Tissue Res. 2013, 353, 65–78. [Google Scholar] [CrossRef]
  31. Kiyokawa, Y.; Terajima, M.; Sato, M.; Inada, E.; Hori, Y.; Bando, R.; Iwase, Y.; Kubota, N.; Murakami, T.; Tsugane, H.; et al. Scratch-based isolation of primary cells (SCIP): A novel method to obtain a large number of human dental pulp cells through one-step cultivation. J. Clin. Med. 2024, 13, 7058. [Google Scholar] [CrossRef] [PubMed]
  32. La Noce, M.; Paino, F.; Spina, A.; Naddeo, P.; Montella, R.; Desiderio, V.; De Rosa, A.; Papaccio, G.; Tirino, V.; Laino, L. Dental pulp stem cells: State of the art and suggestions for a true translation of research into therapy. J. Dent. 2014, 42, 761–768. [Google Scholar] [CrossRef] [PubMed]
  33. Khanna-Jain, R. Growth and differentiation of human dental pulp stem cells maintained in fetal bovine serum, human serum and serum-free/xeno-free culture media. J. Stem Cell Res. Ther. 2012, 2, 126. [Google Scholar] [CrossRef]
  34. Brindley, D.A.; Davie, N.L.; Culme-Seymour, E.J.; Mason, C.; Smith, D.W.; Rowley, J.A. Peak serum: Implications of serum supply for cell therapy manufacturing. Regen. Med. 2012, 7, 7–13. [Google Scholar] [CrossRef]
  35. Mochizuki, M.; Nakahara, T. Establishment of xenogeneic serum-free culture methods for handling human dental pulp stem cells using clinically oriented in-vitro and in-vivo conditions. Stem Cell Res. Ther. 2018, 9, 25. [Google Scholar] [CrossRef]
  36. Xiao, J.; Yang, D.; Li, Q.; Tian, W.; Guo, W. The establishment of a chemically defined serum-free culture system for human dental pulp stem cells. Stem Cell Res. Ther. 2018, 9, 191. [Google Scholar] [CrossRef]
  37. Marrazzo, P.; Paduano, F.; Palmieri, F.; Marrelli, M.; Tatullo, M. Highly efficient in vitro reparative behaviour of dental pulp stem cells cultured with standardised platelet lysate supplementation. Stem Cells Int. 2016, 2016, 7230987. [Google Scholar] [CrossRef]
  38. Gronthos, S.; Brahim, J.; Li, W.; Fisher, L.W.; Cherman, N.; Boyde, A.; DenBesten, P.; Robey, P.G.; Shi, S. Stem cell properties of human dental pulp stem cells. J. Dent. Res. 2002, 81, 531–535. [Google Scholar] [CrossRef]
  39. Stark, R.; Grzelak, M.; Hadfield, J. RNA sequencing: The teenage years. Nat. Rev. Genet. 2019, 20, 631–656. [Google Scholar] [CrossRef]
  40. Kobayashi, T.; Torii, D.; Iwata, T.; Izumi, Y.; Nasu, M.; Tsutsui, T.W. Characterization of proliferation, differentiation potential, and gene expression among clonal cultures of human dental pulp cells. Hum. Cell 2020, 33, 490–501. [Google Scholar] [CrossRef]
  41. Inada, E.; Saitoh, I.; Kubota, N.; Iwase, Y.; Kiyokawa, Y.; Noguchi, H.; Yamasaki, Y.; Sato, M. RNA analysis based on a small number of manually isolated fixed cells (RNA-snMIFxC) to profile stem cells from human deciduous tooth-derived dental pulp cells. Biol. Proced. Online 2021, 23, 12. [Google Scholar] [CrossRef]
  42. Inada, E.; Saitoh, I.; Kubota, N.; Soda, M.; Matsueda, K.; Murakami, T.; Sawami, T.; Kagoshima, A.; Yamasaki, Y.; Sato, M. Alkaline phosphatase and OCT-3/4 as useful markers for predicting susceptibility of human deciduous teeth-derived dental pulp cells to reprogramming factor-induced iPS cells. J. Investig. Clin. Dent. 2017, 8, e12236. [Google Scholar] [CrossRef] [PubMed]
  43. Mutisheva, I.; Robatel, S.; Bäriswyl, L.; Schenk, M. An innovative approach to tissue processing and cell sorting of fixed cells for subsequent single-cell RNA sequencing. Int. J. Mol. Sci. 2022, 23, 10233. [Google Scholar] [CrossRef] [PubMed]
  44. Ren, H.; Wen, Q.; Zhao, Q.; Wang, N.; Zhao, Y. Atlas of human dental pulp cells at multiple spatial and temporal levels based on single-cell sequencing analysis. Front. Physiol. 2022, 13, 993478. [Google Scholar] [CrossRef] [PubMed]
  45. Kawashima, N. Characterisation of dental pulp stem cells: A new horizon for tissue regeneration? Arch. Oral Biol. 2012, 57, 1439–1458. [Google Scholar] [CrossRef]
  46. Yamada, Y.; Nakamura-Yamada, S.; Kusano, K.; Baba, S. Clinical potential and current progress of dental pulp stem cells for various systemic diseases in regenerative medicine: A concise review. Int. J. Mol. Sci. 2019, 20, 1132. [Google Scholar] [CrossRef]
  47. Langenbach, F.; Handschel, J. Effects of dexamethasone, ascorbic acid and β-glycerophosphate on the osteogenic differentiation of stem cells in vitro. Stem Cell Res. Ther. 2013, 4, 117. [Google Scholar] [CrossRef]
  48. Luke, A.M.; Patnaik, R.; Kuriadom, S.; Abu-Fanas, S.; Mathew, S.; Shetty, K.P. Human dental pulp stem cells differentiation to neural cells, osteocytes and adipocytes-An in vitro study. Heliyon 2020, 6, e03054. [Google Scholar] [CrossRef]
  49. Dissanayaka, W.L.; Zhan, X.; Zhang, C.; Hargreaves, K.M.; Jin, L.; Tong, E.H. Coculture of dental pulp stem cells with endothelial cells enhances osteo-/odontogenic and angiogenic potential in vitro. J. Endod. 2012, 38, 454–463. [Google Scholar] [CrossRef]
  50. Pisciotta, A.; Riccio, M.; Carnevale, G.; Beretti, F.; Gibellini, L.; Maraldi, T.; Cavallini, G.M.; Ferrari, A.; Bruzzesi, G.; De Pol, A. Human serum promotes osteogenic differentiation of human dental pulp stem cells in vitro and in vivo. PLoS ONE 2012, 7, e50542. [Google Scholar] [CrossRef]
  51. Li, X.; Yang, H.; Zhang, Y.; Du, X.; Yan, Z.; Li, J.; Wu, B. CGFe and TGF-β1 enhance viability and osteogenic differentiation of human dental pulp stem cells through the MAPK pathway. Exp. Ther. Med. 2021, 22, 1048. [Google Scholar] [CrossRef] [PubMed]
  52. Kornsuthisopon, C.; Nantanapiboon, D.; Rochanavibhata, S.; Nowwarote, N.; Namangkalakul, W.; Osathanon, T. Betaine promotes osteogenic differentiation in immortalized human dental pulp-derived cells. BDJ Open 2022, 8, 31. [Google Scholar] [CrossRef] [PubMed]
  53. Niu, L.N.; Sun, J.Q.; Li, Q.H.; Jiao, K.; Shen, L.J.; Wu, D.; Tay, F.; Chen, J.H. Intrafibrillar-silicified collagen scaffolds enhance the osteogenic capacity of human dental pulp stem cells. J. Dent. 2014, 42, 839–849. [Google Scholar] [CrossRef] [PubMed]
  54. Bar, J.K.; Lis-Nawara, A.; Kowalczyk, T.; Grelewski, P.G.; Stamnitz, S.; Gerber, H.; Klimczak, A. Osteogenic potential of human dental pulp stem cells (hDPSCs) growing on poly L-lactide-co-caprolactone and hyaluronic acid (HYAFF-11TM) scaffolds. Int. J. Mol. Sci. 2023, 24, 16747. [Google Scholar] [CrossRef]
  55. Zhang, X.; Zhuang, J.; Wei, C.; Jin, C.; Zhu, M.; Zhao, S.; Xie, H. Enhancing osteogenic differentiation of dental pulp stem cells with covalently bonded all-carbon scaffolds. Adv. Funct. Mater. 2024, 34, 2400766. [Google Scholar] [CrossRef]
  56. Luzuriaga, J.; García-Gallastegui, P.; García-Urkia, N.; Pineda, J.R.; Irastorza, I.; Fernandez-San-Argimiro, F.J.; Briz, N.; Olalde, B.; Unda, F.; Madarieta, I.; et al. Osteogenic differentiation of human dental pulp stem cells in decellularised adipose tissue solid foams. Eur. Cell Mater. 2022, 43, 112–129. [Google Scholar] [CrossRef]
  57. Alipour, M.; Firouzi, N.; Aghazadeh, Z.; Samiei, M.; Montazersaheb, S.; Khoshfetrat, A.B.; Aghazadeh, M. The osteogenic differentiation of human dental pulp stem cells in alginate-gelatin/Nano-hydroxyapatite microcapsules. BMC Biotechnol. 2021, 21, 6. [Google Scholar] [CrossRef]
  58. Ajlan, S.A.; Ashri, N.Y.; Aldahmash, A.M.; Alnbaheen, M.S. Osteogenic differentiation of dental pulp stem cells under the influence of three different materials. BMC Oral Health 2015, 15, 132. [Google Scholar] [CrossRef]
  59. Soares, D.G.; Rosseto, H.L.; Scheffel, D.S.; Basso, F.G.; Huck, C.; Hebling, J.; de Souza Costa, C.A. Odontogenic differentiation potential of human dental pulp cells cultured on a calcium-aluminate enriched chitosan-collagen scaffold. Clin. Oral Investig. 2017, 21, 2827–2839. [Google Scholar] [CrossRef]
  60. Baldión, P.A.; Velandia-Romero, M.L.; Castellanos, J.E. Odontoblast-like cells differentiated from dental pulp stem cells retain their phenotype after subcultivation. Int. J. Cell Biol. 2018, 2018, 6853189. [Google Scholar] [CrossRef]
  61. Lee, J.H.; Lee, D.S.; Choung, H.W.; Shon, W.J.; Seo, B.M.; Lee, E.H.; Cho, J.Y.; Park, J.C. Odontogenic differentiation of human dental pulp stem cells induced by preameloblast-derived factors. Biomaterials 2011, 32, 9696–9706. [Google Scholar] [CrossRef]
  62. Liu, C.H.; Hung, C.J.; Huang, T.H.; Lin, C.C.; Kao, C.T.; Shie, M.Y. Odontogenic differentiation of human dental pulp cells by calcium silicate materials stimulating via FGFR/ERK signaling pathway. Mater. Sci. Eng. C Mater. Biol. Appl. 2014, 43, 359–366. [Google Scholar] [CrossRef]
  63. Paduano, F.; Marrelli, M.; White, L.J.; Shakesheff, K.M.; Tatullo, M. Odontogenic differentiation of human dental pulp stem cells on hydrogel scaffolds derived from decellularized bone extracellular matrix and collagen type I. PLoS ONE 2016, 11, e0148225. [Google Scholar] [CrossRef]
  64. Huang, G.T.; Shagramanova, K.; Chan, S.W. Formation of odontoblast-like cells from cultured human dental pulp cells on dentin in vitro. J. Endod. 2006, 32, 1066–1073. [Google Scholar] [CrossRef]
  65. Liu, M.; Zhao, L.; Hu, J.; Wang, L.; Li, N.; Wu, D.; Shi, X.; Yuan, M.; Hu, W.; Wang, X. Endothelial cells and endothelin-1 promote the odontogenic differentiation of dental pulp stem cells. Mol. Med. Rep. 2018, 18, 893–901. [Google Scholar] [CrossRef] [PubMed]
  66. Okada, Y.; Kawashima, N.; Noda, S.; Murano, H.; Han, P.; Hashimoto, K.; Kaneko, T.; Okiji, T. VEGFA promotes odonto/osteoblastic differentiation in dental pulp stem cells via ERK/p38 signaling. J. Dent. Sci. 2025, 20, 1590–1597. [Google Scholar] [CrossRef] [PubMed]
  67. Kraus, N.A.; Ehebauer, F.; Zapp, B.; Rudolphi, B.; Kraus, B.J.; Kraus, D. Quantitative assessment of adipocyte differentiation in cell culture. Adipocyte 2016, 5, 351–358. [Google Scholar] [CrossRef] [PubMed]
  68. Moseti, D.; Regassa, A.; Kim, W.-K. Molecular regulation of adipogenesis and potential anti-adipogenic bioactive molecules. Int. J. Mol. Sci. 2016, 17, 124. [Google Scholar] [CrossRef] [PubMed]
  69. Anitua, E.; Troya, M.; Zalduendo, M. Review Progress in the use of dental pulp stem cells in regenerative medicine. Cytotherapy 2018, 20, 479–498. [Google Scholar] [CrossRef]
  70. Garcia-Urkia, N.; Luzuriaga, J.; Uribe-Etxebarria, V.; Irastorza, I.; Fernandez-San-Argimiro, F.J.; Olalde, B.; Briz, N.; Unda, F.; Ibarretxe, G.; Madarieta, I.; et al. Enhanced adipogenic differentiation of human dental pulp stem cells in enzymatically decellularized adipose tissue solid foams. Biology 2022, 11, 1099. [Google Scholar] [CrossRef]
  71. Chang, C.-C.; Chang, K.-C.; Tsai, S.-J.; Chang, H.-H.; Lin, C.-P. Neurogenic differentiation of dental pulp stem cells to neuron-like cells in dopaminergic and motor neuronal inductive media. J. Formos. Med. Assoc. 2014, 113, 956–965. [Google Scholar] [CrossRef]
  72. Osathanon, T.; Sawangmake, C.; Nowwarote, N.; Pavasant, P. Neurogenic differentiation of human dental pulp stem cells using different induction protocols. Oral Dis. 2014, 20, 352–358. [Google Scholar] [CrossRef]
  73. Lee, J.-H.; Um, S.; Song, I.-S.; Kim, H.Y.; Seo, B.M. Neurogenic differentiation of human dental stem cells in vitro. J. Korean Assoc. Oral Maxillofac. Surg. 2014, 40, 173–180. [Google Scholar] [CrossRef] [PubMed]
  74. Haratizadeh, S.; Bojnordi, M.N.; Darabi, S.; Karimi, N.; Naghikhani, M.; Hamidabadi, H.G.; Seifi, M. Condition medium of cerebrospinal fluid and retinoic acid induces the transdifferentiation of human dental pulp stem cells into neuroglia and neural like cells. Anat. Cell Biol. 2017, 50, 107–114. [Google Scholar] [CrossRef] [PubMed]
  75. Seonwoo, H.; Jang, K.-J.; Lee, D.; Park, S.; Lee, M.; Park, S.; Lim, K.-T.; Kim, J.; Chung, J.H. Neurogenic differentiation of human dental pulp stem cells on graphene-polycaprolactone hybrid nanofibers. Nanomaterials 2018, 8, 554. [Google Scholar] [CrossRef] [PubMed]
  76. Darabi, S.; Tiraihi, T.; Bojnordi, M.N.; Hamidabadi, H.G.; Rezaei, N.; Zahiri, M.; Alizadeh, R. Trans-differentiation of human dental pulp stem cells into cholinergic-like neurons via nerve growth factor. Basic Clin. Neurosci. 2019, 10, 609–617. [Google Scholar] [CrossRef]
  77. Kogo, Y.; Seto, C.; Totani, Y.; Mochizuki, M.; Nakahara, T.; Oka, K.; Yoshioka, T.; Ito, E. Rapid differentiation of human dental pulp stem cells to neuron-like cells by high K+ stimulation. Biophys. Physicobiol. 2020, 17, 132–139. [Google Scholar] [CrossRef]
  78. Al-Maswary, A.A.; O’Reilly, M.; Holmes, A.P.; Walmsley, A.D.; Cooper, P.R.; Scheven, B.A. Exploring the neurogenic differentiation of human dental pulp stem cells. PLoS ONE 2022, 17, e0277134. [Google Scholar] [CrossRef]
  79. Ishkitiev, N.; Yaegaki, K.; Imai, T.; Tanaka, T.; Nakahara, T.; Ishikawa, H.; Mitev, V.; Haapasalo, M. High-purity hepatic lineage differentiated from dental pulp stem cells in serum-free medium. J. Endod. 2012, 38, 475–480. [Google Scholar] [CrossRef]
  80. Bento, L.W.; Zhang, Z.; Imai, A.; Nör, F.; Dong, Z.; Shi, S.; Araujo, F.B.; Nör, J.E. Endothelial differentiation of SHED requires MEK1/ERK signaling. J. Dent. Res. 2013, 92, 51–57. [Google Scholar] [CrossRef]
  81. Bronckaers, A.; Hilkens, P.; Fanton, Y.; Struys, T.; Gervois, P.; Politis, C.; Martens, W.; Lambrichts, I. Angiogenic properties of human dental pulp stem cells. PLoS ONE 2013, 8, e71104. [Google Scholar] [CrossRef]
  82. Dissanayaka, W.L.; Hargreaves, K.M.; Jin, L.; Samaranayake, L.P.; Zhang, C. The interplay of dental pulp stem cells and endothelial cells in an Injectable peptide hydrogel on angiogenesis and pulp regeneration in vivo. Tissue Eng. Part A 2014, 21, 550–563. [Google Scholar] [CrossRef]
  83. Luzuriaga, J.; Irurzun, J.; Irastorza, I.; Unda, F.; Ibarretxe, G.; Pineda, J.R. Vasculogenesis from human dental pulp stem cells grown in Matrigel with fully defined serum-free culture media. Biomedicines 2020, 8, 483. [Google Scholar] [CrossRef] [PubMed]
  84. Diomede, F.; Pizzicannella, J.; Merciaro, I.; Antonucci, I.; Guarnieri, S.; Ballerini, P.; Stuppia, L.; Trubiani, O. In vitro cardiomyogenic differentiation of human dental pulp stem cells. The role of 5-azacytidine. Ital. J. Anat. Embryol. 2014, 119 (Suppl. S1), 71. [Google Scholar] [CrossRef]
  85. Carnevalea, G.; Riccioa, M.; Pisciottaa, A.; Berettia, F.; Maraldia, T.; Zavattia, M.; Cavallinia, G.M.; La Salab, G.B.; Ferraric, A.; De Pola, A. In vitro differentiation into insulin-producing β-cells of stem cells isolated from human amniotic fluid and dental pulp. Dig. Liver Dis. 2013, 45, 669–676. [Google Scholar] [CrossRef] [PubMed]
  86. Ishkitiev, N.; Yaegaki, K.; Kozhuharova, A.; Tanaka, T.; Okada, M.; Mitev, V.; Fukuda, M.; Imai, T. Pancreatic differentiation of human dental pulp CD117+ stem cells. Regen. Med. 2013, 8, 597–612. [Google Scholar] [CrossRef] [PubMed]
  87. Kanafi, M.M.; Rajeshwari, Y.B.; Gupta, S.; Dadheech, N.; Nair, P.D.; Gupta, P.K.; Bhonde, R.R. Transplantation of islet-like cell clusters derived from human dental pulp stem cells restores normoglycemia in diabetic mice. Cytotherapy 2013, 15, 1228–1236. [Google Scholar] [CrossRef]
  88. Mendoza, H.Y.; Yokoyama, T.; Tanaka, T.; Ii, H.; Yaegaki, K. Regeneration of insulin-producing islets from dental pulp stem cells using a 3D culture system. Regen. Med. 2018, 13, 673–687. [Google Scholar] [CrossRef]
  89. Shivakumar, S.B.; Lee, H.-J.; Son, Y.-B.; Bharti, D.; Ock, S.A.; Lee, S.-L.; Kang, Y.-H.; Park, B.-W.; Rho, G.-J. In vitro differentiation of single donor derived human dental mesenchymal stem cells into pancreatic β cell-like cells. Biosci. Rep. 2019, 39, BSR20182051. [Google Scholar] [CrossRef]
  90. Kuncorojakti, S.; Rodprasert, W.; Le, Q.D.; Osathanon, T.; Pavasant, P.; Sawangmake, C. In vitro induction of human dental pulp stem cells toward pancreatic lineages. J. Vis. Exp. 2021, 175, e62497. [Google Scholar] [CrossRef]
  91. Aly, R.M.; Aglan, H.A.; Eldeen, G.N.; Ahmed, H.H. Optimization of differentiation protocols of dental tissues stem cells to pancreatic β-cells. BMC Mol. Cell Biol. 2022, 23, 41. [Google Scholar] [CrossRef]
  92. Abuarqoub, D.; Adwan, S.; Zaza, R.; Wehaibi, S.; Aslam, N.; Jafar, H.; Qinna, N.A.; Awidi, A. Effective generation of functional pancreatic β cells from human-derived dental stem cells of apical papilla and bone-marrow-derived stem cells: A comparative study. Pharmaceuticals 2023, 16, 649. [Google Scholar] [CrossRef]
  93. Song, B.; Jiang, W.; Alraies, A.; Liu, Q.; Gudla, V.; Oni, J.; Wei, X.; Sloan, A.; Ni, L.; Agarwal, M. Bladder smooth muscle cells differentiation from dental pulp stem cells: Future potential for bladder tissue engineering. Stem Cells Int. 2016, 2016, 6979368. [Google Scholar] [CrossRef]
  94. Xu, J.G.; Zhu, S.Y.; Heng, B.C.; Dissanayaka, W.L.; Zhang, C.F. TGF-β1-induced differentiation of SHED into functional smooth muscle cells. Stem Cell Res. Ther. 2017, 8, 10. [Google Scholar] [CrossRef]
  95. Zhang, Y.; Zhong, J.; Lin, S.; Hu, M.; Liu, J.; Kang, J.; Qi, Y.; Basabrain, M.S.; Zou, T.; Zhang, C. Direct contact with endothelial cells drives dental pulp stem cells toward smooth muscle cells differentiation via TGF-β1 secretion. Int. Endod. J. 2023, 56, 1092–1107. [Google Scholar] [CrossRef]
  96. Ghanavatinejad, F.; Tabrizi, Z.P.F.; Omidghaemi, S.; Sharifi, E.; Møller, S.G.; Jami, M.-S. Protein biomarkers of neural system. J. Otol. 2019, 14, 77–88. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, R.; Liu, J.; Nie, S.; Wang, X.; Li, B.; Sun, D.; Refukati, D.; Liu, Y. Transfection of stem cells derived from rat dental pulp with green fluorescent protein infection by lentiviral vector. Chin. J. Tissue Eng. Res. 2014, 53, 7299–7305. [Google Scholar] [CrossRef]
  98. Zhan, Y.; Li, X.; Gou, X.; Yuan, G.; Fan, M.; Yang, G. DLX3 inhibits the proliferation of human dental pulp cells through inactivation of canonical Wnt/β-catenin signaling pathway. Front. Physiol. 2018, 9, 1637. [Google Scholar] [CrossRef]
  99. Zhang, M.; Jiang, F.; Zhang, X.; Wang, S.; Jin, Y.; Zhang, W.; Jiang, X. The effects of platelet-derived growth factor-BB on human dental pulp stem cells mediated dentin-pulp complex regeneration. Stem Cells Transl. Med. 2017, 6, 2126–2134. [Google Scholar] [CrossRef]
  100. Guirado, E.; Zhang, Y.; George, A. Establishment of stable cell lines from primary human dental pulp stem cells. Methods Mol. Biol. 2019, 1922, 21–27. [Google Scholar] [CrossRef]
  101. Wang, W.; Yuan, C.; Geng, T.; Liu, Y.; Zhu, S.; Zhang, C.; Liu, Z.; Wang, P. EphrinB2 overexpression enhances osteogenic differentiation of dental pulp stem cells partially through ephrinB2-mediated reverse signaling. Stem Cell Res. Ther. 2020, 11, 40. [Google Scholar] [CrossRef]
  102. Janebodin, K.; Chavanachat, R.; Hays, A.; Gil, M.R. Silencing VEGFR-2 hampers odontoblastic differentiation of dental pulp stem cells. Front. Cell Dev. Biol. 2021, 9, 665886. [Google Scholar] [CrossRef]
  103. Cao, Y.; Liu, Z.; Xie, Y.; Hu, J.; Wang, H.; Fan, Z.; Zhang, C.; Wang, J.; Wu, C.-T.; Wang, S. Adenovirus-mediated transfer of hepatocyte growth factor gene to human dental pulp stem cells under good manufacturing practice improves their potential for periodontal regeneration in swine. Stem Cell Res. Ther. 2015, 6, 249. [Google Scholar] [CrossRef]
  104. Orimoto, A.; Kyakumoto, S.; Eitsuka, T.; Nakagawa, K.; Kiyono, T.; Fukuda, T. Efficient immortalization of human dental pulp stem cells with expression of cell cycle regulators with the intact chromosomal condition. PLoS ONE 2020, 15, e0229996. [Google Scholar] [CrossRef]
  105. Nakashima, M.; Mizunuma, K.; Murakami, T.; Akamine, A. Induction of dental pulp stem cell differentiation into odontoblasts by electroporation-mediated gene delivery of growth/differentiation factor 11 (Gdf11). Gene Ther. 2002, 9, 814–818. [Google Scholar] [CrossRef]
  106. Rizk, A.; Rabie, B.M. Electroporation for transfection and differentiation of dental pulp stem cells. Biores. Open Access 2013, 2, 155–162. [Google Scholar] [CrossRef]
  107. Inada, E.; Saitoh, I.; Kubota, N.; Iwase, Y.; Kiyokawa, Y.; Shibasaki, S.; Noguchi, H.; Yamasaki, Y.; Sato, M. piggyBac transposon-based immortalization of human deciduous tooth dental pulp cells with multipotency and non-tumorigenic potential. Int. J. Mol. Sci. 2019, 20, 4904. [Google Scholar] [CrossRef]
  108. Murakami, T.; Saitoh, I.; Sato, M.; Inada, E.; Soda, M.; Oda, M.; Domon, H.; Iwase, Y.; Sawami, T.; Matsueda, K.; et al. Isolation and characterization of lymphoid enhancer factor-1-positive deciduous dental pulp stem-like cells after transfection with a piggyBac vector containing LEF1 promoter-driven selection markers. Arch. Oral Biol. 2017, 81, 110–120. [Google Scholar] [CrossRef] [PubMed]
  109. Kim, I.H.; Irfan, M.; Sreekumar, S.; Phimon, A.; Kim, S.; Chung, S. CRISPR-edited DPSCs constitutively expressing BDNF enhance dentin regeneration in injured teeth. eLife 2025, 14, RP105153. [Google Scholar] [CrossRef] [PubMed]
  110. Lee, M.C.; Seonwoo, H.; Jang, K.J.; Pandey, S.; Lim, J.; Park, S.; Kim, J.E.; Choung, Y.-H.; Garg, P.; Chung, J.H. Development of novel gene carrier using modified nano hydroxyapatite derived from equine bone for osteogenic differentiation of dental pulp stem cells. Bioact. Mater. 2021, 6, 2742–2751. [Google Scholar] [CrossRef] [PubMed]
  111. Salkın, H.; Gönen, Z.B.; Ergen, E.; Bahar, D.; Çetin, M. Effects of TGF-β1 overexpression on biological characteristics of human dental pulp-derived mesenchymal stromal cells. Int. J. Stem Cells. 2019, 12, 170–182. [Google Scholar] [CrossRef] [PubMed]
  112. Cao, L.-L.; Zhang, Y.-J.; Wang, J.-W.; Tian, F.; Wang, C.-F. Studies on microRNA regulation of multidirectional differentiation of dental pulp stem cells: A narrative review. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 1816–1824. [Google Scholar] [CrossRef] [PubMed]
  113. Liu, H.; Lin, H.; Zhang, L.; Sun, Q.; Yuan, G.; Zhang, L.; Chen, S.; Chen, Z. miR-145 and miR-143 regulate odontoblast differentiation through targeting Klf4 and Osx genes in a feedback Loop. J. Biol. Chem. 2013, 288, 9261–9271. [Google Scholar] [CrossRef] [PubMed]
  114. Stacey, G.; MacDonald, C. Immortalisation of primary cells. Cell Biol. Toxicol. 2001, 17, 231–246. [Google Scholar] [CrossRef]
  115. Sato, M.; Saitoh, I.; Inada, E.; Nakamura, S.; Watanabe, S. Potential for isolation of immortalized hepatocyte cell lines by liver-directed in vivo gene delivery of transposons in mice. Stem Cells Int. 2019, 2019, 5129526. [Google Scholar] [CrossRef]
  116. Kitagawa, M.; Ueda, H.; Iizuka, S.; Sakamoto, K.; Oka, H.; Kudo, Y.; Ogawa, I.; Miyauchi, M.; Tahara, H.; Takata, T. Immortalization and characterization of human dental pulp cells with odontoblastic differentiation. Arch. Oral Biol. 2007, 52, 727–731. [Google Scholar] [CrossRef]
  117. Zanini, M.; Sautier, J.M.; Berdal, A.; Simon, S. Biodentine induces immortalized murine pulp cell differentiation into odontoblast-like cells and stimulates biomineralization. J. Endod. 2012, 38, 1220–1226. [Google Scholar] [CrossRef]
  118. Villa, I.; Senesi, P.; Montesano, A.; Ferraretto, A.; Vacante, F.; Spinello, A.; Bottani, M.; Bolamperti, S.; Rubinacci, A.; Luzi, L.; et al. Betaine promotes cell differentiation of human osteoblasts in primary culture. J. Transl. Med. 2017, 15, 132. [Google Scholar] [CrossRef]
  119. Zhang, C.; Chang, J.; Sonoyama, W.; Shi, S.; Wang, C.-Y. Inhibition of human dental pulp stem cell differentiation by Notch signaling. J. Dent. Res. 2008, 87, 250–255. [Google Scholar] [CrossRef]
  120. Lin, H.; Xu, L.; Liu, H.; Chen, Z.; Yuan, G.; Chen, Z. KLF4 promotes the odontoblastic differentiation of human dental pulp cells. J. Endod. 2011, 37, 948–954. [Google Scholar] [CrossRef]
  121. Wang, X.; He, F.; Tan, Y.; Tian, W.; Qiu, S. Inhibition of Delta1 promotes differentiation of odontoblasts and inhibits proliferation of human dental pulp stem cell in vitro. Arch. Oral Biol. 2011, 56, 837–845. [Google Scholar] [CrossRef]
  122. Hara, E.S.; Ono, M.; Eguchi, T.; Kubota, S.; Pham, H.T.; Sonoyama, W.; Tajima, S.; Takigawa, M.; Calderwood, S.K.; Kuboki, T. miRNA-720 controls stem cell phenotype, proliferation and differentiation of human dental pulp cells. PLoS ONE 2013, 8, e83545. [Google Scholar] [CrossRef]
  123. Chen, K.-L.; Huang, Y.-Y.; Lung, J.; Yeh, Y.-Y.; Yuan, K. CD44 is involved in mineralization of dental pulp cells. J. Endod. 2013, 39, 351–356. [Google Scholar] [CrossRef] [PubMed]
  124. Wu, Y.; Sun, H.; Song, F.; Fu, D.; Wang, J. DDIT3 overexpression increases odontoblastic potential of human dental pulp cells. Cell Prolif. 2014, 47, 249–257. [Google Scholar] [CrossRef] [PubMed]
  125. Maciejewska, I.; Sakowicz-Burkiewicz, M.; Pawelczyk, T. Id1 expression level determines the differentiation of human dental pulp stem cells. J. Dent. Res. 2014, 93, 576–581. [Google Scholar] [CrossRef] [PubMed]
  126. Gu, S.; Ran, S.; Qin, F.; Cao, D.; Wang, J.; Liu, B.; Liang, J. Human dental pulp stem cells via the NF-κB pathway. Cell Physiol. Biochem. 2015, 36, 1725–1734. [Google Scholar] [CrossRef]
  127. Heng, B.C.; Ye, X.; Liu, Y.; Dissanayaka, W.L.; Shun, G.; Cheung, P.; Zhang, C. Effects of recombinant overexpression of Bcl2 on the proliferation, apoptosis, and osteogenic/odontogenic differentiation potential of dental pulp stem cells. J. Endod. 2016, 42, 575–583. [Google Scholar] [CrossRef]
  128. Yang, Y.; Zhao, Y.; Liu, X.; Chen, Y.; Liu, P.; Zhao, L. Effect of SOX2 on odontoblast differentiation of dental pulp stem cells. Mol. Med. Rep. 2017, 16, 9659–9663. [Google Scholar] [CrossRef]
  129. Ou, Y.; Zhou, Y.; Liang, S.; Wang, Y. Sclerostin promotes human dental pulp cells senescence. PeerJ 2018, 6, e5808. [Google Scholar] [CrossRef]
  130. Bao, Q.; Zhang, J.; Wang, X.-X. Effects of Btbd7 knockdown on the proliferation of human dental pulp cells and expression of Dspp. Int. J. Clin. Exp. Pathol. 2018, 11, 1460–1465. [Google Scholar]
  131. Zeng, L.; Zhao, N.; Li, F.; Han, D.; Liu, Y.; Liu, H.; Sun, S.; Wang, Y.; Feng, H. miR-675 promotes odontogenic differentiation of human dental pulp cells by epigenetic regulation of DLX3. Exp. Cell Res. 2018, 367, 104–111. [Google Scholar] [CrossRef]
  132. Liu, F.; Wang, X.; Yang, Y.; Hu, R.; Wang, W.; Wang, Y. The suppressive effects of miR-508-5p on the odontogenic differentiation of human dental pulp stem cells by targeting glycoprotein non-metastatic melanomal protein B. Stem Cell Res. Ther. 2019, 10, 35. [Google Scholar] [CrossRef]
  133. Huang, X.; Liu, F.; Hou, J.; Chen, K. Inflammation-induced overexpression of microRNA-223-3p regulates odontoblastic differentiation of human dental pulp stem cells by targeting SMAD3. Int. Endod. J. 2019, 52, 491–503. [Google Scholar] [CrossRef]
  134. Guirado, E.; Chen, Y.; Ross, R.D.; Zhang, Y.; Chaussain, C.; George, A. Disrupted protein expression and altered proteolytic events in hypophosphatemic dentin can be rescued by dentin matrix protein 1. Front. Physiol. 2020, 11, 82. [Google Scholar] [CrossRef]
  135. Zhong, J.; Tu, X.; Kong, Y.; Guo, L.; Li, B.; Zhong, W.; Cheng, Y.; Jiang, Y.; Jiang, Q. LncRNA H19 promotes odontoblastic differentiation of human dental pulp stem cells by regulating miR-140-5p and BMP-2/FGF9. Stem Cell Res. Ther. 2020, 11, 202. [Google Scholar] [CrossRef]
  136. Guirado, E.; Villani, C.; Petho, A.; Chen, Y.; Maienschein-Cline, M.; Lei, Z.; Los, N.; George, A. Wnt pathway inhibitors are upregulated in XLH dental pulp cells in response to odontogenic differentiation. Int. J. Oral Sci. 2023, 15, 13. [Google Scholar] [CrossRef]
  137. Wang, X.; Zhang, J.; Sun, Z.; Zhao, S.; Deng, W. MicroRNA-15b-5p modulates the differentiation of human dental pulp stem cells into odontoblasts via targeting IGF1. J. Biol. Regul. Homeost. Agents. 2023, 37, 7191. [Google Scholar] [CrossRef]
  138. Yuan, J.; Liu, X.; Chen, Y.; Zhao, Y.; Liu, P.; Zhao, L.; Han, W. Effect of SOX2 on osteogenic differentiation of dental pulp stem cells. Cell Mol. Biol. 2017, 63, 41–44. [Google Scholar] [CrossRef] [PubMed]
  139. Zhong, T.; Gao, Y.; Qiao, H.; Zhou, H.; Liu, Y. Elevated osteogenic potential of stem cells from inflammatory dental pulp tissues by Wnt4 overexpression for treating bone defect in rats. Ann. Palliat. Med. 2020, 9, 2962–2969. [Google Scholar] [CrossRef] [PubMed]
  140. Ji, F.; Zhu, L.; Pan, J.; Shen, Z.; Yang, Z.; Wang, J.; Bai, X.; Lin, Y.; Tao, J. hsa_circ_0026827 promotes osteoblast differentiation of human dental pulp stem cells through the Beclin1 and RUNX1 signaling pathways by sponging miR-188-3p. Front. Cell Dev. Biol. 2020, 8, 470. [Google Scholar] [CrossRef]
  141. Kim, I.H.; Jeon, M.; Lee, Y.H.; Shin, J.S.; Lee, T.; Lee, S.K.; Cheon, K.; Song, J.S. HIF-1α overexpression using a protein transduction domain to increase the osteogenic potential of SHED. J. Dent. Oral Biol. 2023, 8, 1215. [Google Scholar]
  142. Wen, W.; Zheng, P.; Meng, H.; Liu, H.; Yuan, C. Overexpression of Sema3A promotes osteogenic differentiation of dental pulp stem cells and MC3T3-E1. Chin. J. Tissue Eng. Res. 2024, 28, 993–999. [Google Scholar] [CrossRef]
  143. Deng, P.; Yang, B.; Huang, C.; Li, Y.; Mei, Z.; Li, Y.; Li, J. GDF15 promotes osteogenic differentiation of human dental pulp stem cells by activating the TGF-β/SMAD signaling pathway. J. Tissue Eng. 2025, 16, 20417314251357752. [Google Scholar] [CrossRef] [PubMed]
  144. Fukuyama, Y.; Ohta, K.; Okoshi, R.; Suehara, M.; Kizaki, H.; Nakagawa, K. Hypoxia induces expression and activation of AMPK in rat dental pulp cells. J. Dent. Res. 2007, 86, 903–907. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, P.; Cai, J.; Dong, D.; Chen, Y.; Liu, X.; Wang, Y.; Zhou, Y. Effects of SOX2 on proliferation, migration and adhesion of human dental pulp stem cells. PLoS ONE 2015, 10, e0141346. [Google Scholar] [CrossRef] [PubMed]
  146. Zhang, P.; Zhu, C.; Li, S.; Zhang, L.; Wu, X.; Zhang, H.; Wang, F.; Tan, Y. MiR-633 promotes cell proliferation and differentiation by targeting MEPE in human adult dental pulp stem cells. Int. J. Clin. Exp. Pathol. 2016, 9, 8066–8074. [Google Scholar]
  147. Yang, X.; He, L.; Pan, S.; Li, Y.; Niu, Y. Effect of Satb2 overexpression on proliferation and migration of human dental pulp stem cells. J. Oral Sci. Res. 2017, 33, 597–600. [Google Scholar] [CrossRef]
  148. Zhu, L.; Dissanayaka, W.L.; Zhang, C. Dental pulp stem cells overexpressing stromal-derived factor-1α and vascular endothelial growth factor in dental pulp regeneration. Clin. Oral Investig. 2019, 23, 2497–2509. [Google Scholar] [CrossRef]
  149. Liu, Y.; Dong, N.; Miao, J.; Li, C.; Wang, X.; Ruan, J. Lin28 promotes dental pulp cell proliferation via upregulation of cyclin-dependent proteins and interaction with let-7a/IGF2BP2 pathways. Biomed. Pharmacother. 2019, 113, 108742. [Google Scholar] [CrossRef]
  150. Yuan, X.; Huan, Z.; Wang, H. Effects of microRNA-210-3p on proliferation and differentiation of human dental pulp cells under hypoxic condition. J. Oral Sci. Res. 2019, 35, 970–974. [Google Scholar] [CrossRef]
  151. Ok, C.Y.; Park, S.; Jang, H.-O.; Takata, T.; Bae, M.-K.; Kim, Y.-D.; Ryu, M.H.; Bae, S.-K. Visfatin induces senescence of human dental pulp cells. Cells 2020, 9, 193. [Google Scholar] [CrossRef]
  152. 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] [PubMed]
  153. Huang, L.; Sun, C.; Peng, R.; Liu, Z. The effect of LncRNA H19 on human dental pulp cells through tumor growth factor-β1 (Tgf-Β1)/Smad signaling pathway. J. Biomat. Tissue Eng. 2022, 12, 1247–1251. [Google Scholar] [CrossRef]
  154. Dou, W.; Xie, J.; Chen, J.; Zhou, J.; Xu, Z.; Wang, Z.; Zhu, Q. Overexpression of adrenomedullin (ADM) alleviates the senescence of human dental pulp stem cells by regulating the miR-152/CCNA2 pathway. Cell Cycle 2023, 22, 565–579. [Google Scholar] [CrossRef] [PubMed]
  155. Liu, W.; Gong, Q.; Ling, J.; Zhang, W.; Liu, Z.; Quan, J. Role of MiR-424 on angiogenic potential in human dental pulp cells. J. Endod. 2014, 40, 76–82. [Google Scholar] [CrossRef]
  156. Dissanayaka, W.L.; Han, Y.; Zhang, L.; Zou, T.; Zhang, C. Bcl-2 Overexpression and hypoxia synergistically enhance angiogenic properties of dental pulp stem cells. Int. J. Mol. Sci. 2020, 21, 6159. [Google Scholar] [CrossRef]
  157. Li, J.; Zhu, Y.; Li, N.; Wu, T.; Zheng, X.; Heng, B.C.; Zou, D.; Xu, J. Upregulation of ETV2 expression promotes endothelial differentiation of human dental pulp stem cells. Cell Transplant. 2021, 30, 963689720978739. [Google Scholar] [CrossRef]
  158. Jiang, W.; Duan, S.; Li, W.; Yan, H.; Si, C.; Xu, N.; Li, Y.; Zhang, W.; Gu, S. PDGF-BB overexpressing dental pulp stem cells improve angiogenesis in dental pulp regeneration. Front. Bioeng. Biotechnol. 2025, 13, 1578410. [Google Scholar] [CrossRef]
  159. Behrouznezhad, F.; Ejeian, F.; Emadi-Baygi, M.; Nikpour, P.; Nasr-Esfahani, M.H. Hypothesis: A challenge of overexpression Zfp521 in neural tendency of derived dental pulp stem cells. Cell J. 2019, 21, 99–102. [Google Scholar] [CrossRef]
  160. Li, J.; Diao, S.; Yang, H.; Cao, Y.; Du, J.; Yang, D. IGFBP5 promotes angiogenic and neurogenic differentiation potential of dental pulp stem cells. Dev. Growth Differ. 2019, 61, 457–465. [Google Scholar] [CrossRef]
  161. Gancheva, M.R.; Kremer, K.; Breen, J.; Arthur, A.; Hamilton-Bruce, A.; Thomas, P.; Gronthos, S.; Koblar, S. Effect of octamer-binding transcription factor 4 overexpression on the neural induction of human dental pulp stem cells. Stem Cell Rev. Rep. 2024, 20, 797–815. [Google Scholar] [CrossRef]
  162. Balam-Lara, J.A.; Carrillo-Cocom, L.M.; Rodas-Junco, B.; Villanueva-Lizama, L.; Nic-Can, G. TEN ELEVEN TRANSLOCATION 2 (TET2) improves the adipogenic potential of dental pulp stem cells. J. Mex. Che. Soc. 2023, 67, 2023. [Google Scholar] [CrossRef]
  163. Tancharoen, S.; Tengrungsun, T.; Suddhasthira, T.; Kikuchi, K.; Vechvongvan, N.; Tokuda, M.; Maruyama, I. Overexpression of receptor for advanced glycation end products and high-mobility group box 1 in human dental pulp inflammation. Mediat. Inflamm. 2014, 2014, 754069. [Google Scholar] [CrossRef]
  164. Meng, H.; Wei, F.; Zhou, Y.; Hu, L.; Ge, Z.; Jin, J.; Wang, H.; Wu, C.-T. Overexpression of hepatocyte growth factor in dental pulp stem cells ameliorates the severity of psoriasis by reducing inflammatory responses. Stem Cells Dev. 2021, 30, 876–889. [Google Scholar] [CrossRef]
  165. Ni, C.; Wu, G.; Miao, T.; Xu, J. Wnt4 prevents apoptosis and inflammation of dental pulp cells induced by LPS by inhibiting the IKK/NF-κB pathway. Exp. Thera. Med. 2023, 25, 75. [Google Scholar] [CrossRef]
  166. Kim, S.-A.; Choi, H.S.; Ahn, S.-G. Pin1 induces the ADP-induced migration of human dental pulp cells through P2Y1 stabilization. Oncotarget 2016, 7, 85381–85392. [Google Scholar] [CrossRef]
  167. Shi, J.-F.; Zhu, C.-H.; Liu, J.; Sun, J.-Y.; Rao, G.-Z.; Li, A. Recombinant hFOXA2 and hPDX1 lentivirus induced dental pulp stem cells from deciduous teeth reprogramming for insulin-producing cells. Shanghai J. Stomatal. 2013, 22, 634–642. [Google Scholar]
  168. Nozaki, T.; Ohura, K. Inhibition of miR-183 induces insulin in dental pulp cells. J. Hard Tissue Biol. 2017, 26, 319–322. [Google Scholar] [CrossRef]
  169. Nozaki, T.; Ohura, K. Regulation of miRNA during direct reprogramming of dental pulp cells to insulin-producing cells. Biochem. Biophys. Res. Commun. 2014, 444, 195–198. [Google Scholar] [CrossRef]
  170. Askari, N.; Yaghoobi, M.M.; Shamsara, M.; Esmaeili-Mahani, S. Human dental pulp stem cells differentiate into oligodendrocyte progenitors using the expression of Olig2 transcription factor. Cells Tissues Organs 2014, 200, 93–103. [Google Scholar] [CrossRef] [PubMed]
  171. Qian, H.; Huang, Q.; Chen, Y.-X.; Liu, Q.; Fang, J.-X.; Ye, M.-W. Caspase-9 was involved in cell apoptosis in human dental pulp stem cells from deciduous teeth. Mol. Med. Rep. 2018, 18, 1067–1073. [Google Scholar] [CrossRef]
  172. Qiao, W.; Li, D.; Shi, Q.; Wang, H.; Wang, H.; Guo, J. miR-224-5p protects dental pulp stem cells from apoptosis by targeting Rac1. Exp. Ther. Med. 2020, 19, 9–18. [Google Scholar] [CrossRef]
  173. Li, D.; Deng, T.; Li, H.; Li, Y. MiR-143 and miR-135 inhibitors treatment induces skeletal myogenic differentiation of human adult dental pulp stem cells. Arch. Oral Biol. 2015, 60, 1613–1617. [Google Scholar] [CrossRef] [PubMed]
  174. Xie, Y.; Shen, G. MicroRNA-139-5p elevates skeletal myogenic differentiation of human adult dental pulp stem cells through Wnt/β-catenin signaling pathway. Exp. Ther. Med. 2018, 16, 2835–2842. [Google Scholar] [CrossRef] [PubMed]
  175. Liu, L.; Wu, L.; Wei, X.; Ling, J. Induced overexpression of Oct4A in human dental pulp cells enhances pluripotency and multilineage differentiation capability. Stem Cells Dev. 2015, 24, 962–972. [Google Scholar] [CrossRef] [PubMed]
  176. Liu, L.; Peng, Z.; Xu, Z.; Wei, X. XPC promotes pluripotency of human dental pulp cells through regulation of Oct-4/Sox2/c-Myc. Stem Cells Int. 2016, 2016, 3454876. [Google Scholar] [CrossRef]
  177. Abuarqoub, D.; Theeb, L.S.; Omari, M.B.; Hamadneh, Y.I.; Alrawabdeh, J.A.; Aslam, N.; Jafar, H.; Awidi, A. The osteogenic role of biomaterials combined with human-derived dental stem cells in bone tissue regeneration. Tissue Eng. Regen. Med. 2023, 20, 251–270. [Google Scholar] [CrossRef]
  178. Zhang, P.; Yang, W.; Wang, G.; Li, Y. miR-143 suppresses the osteogenic differentiation of dental pulp stem cells by inactivation of NF-κB signaling pathway via targeting TNF-α. Arch. Oral Biol. 2018, 87, 172–179. [Google Scholar] [CrossRef]
  179. Lim, J.; Heo, Y.; Choi, S.S. Investigation of changes in DNA methylation associated with alterations in gene expression resulting in differences between lean and obese adipogenesis. Genomics 2023, 115, 110623. [Google Scholar] [CrossRef]
  180. Argaez-Sosa, A.A.; Rodas-Junco, B.A.; Carrillo-Cocom, L.M.; Rojas-Herrera, R.A.; Coral-Sosa, A.; Aguilar-Ayala, F.J.; Aguilar-Pérez, D.; Nic-Can, G. Higher expression of DNA (de)methylation-related genes reduces adipogenicity in dental pulp stem cells. Front. Cell Dev. Biol. 2022, 10, 791667. [Google Scholar] [CrossRef]
  181. Shimizu, K.; Taniyama, Y.; Sanada, F.; Iwabayashi, M.; Azuma, J.; Iekushi, K.; Katsuragi, N.; Otsu, R.; Shibata, K.; Ishikawa, Y.; et al. Novel mechanism of hepatocyte growth factor against prevention of inflammation and oxidative stress. Inflamm. Regen. 2013, 33, 136–142. [Google Scholar] [CrossRef]
  182. Zhou, Q.; Brown, J.; Kanarek, A.; Rajagopal, J.; Melton, D.A. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 2008, 455, 627–632. [Google Scholar] [CrossRef]
  183. Mauda-Havakuk, M.; Litichever, N.; Chernichovski, E.; Nakar, O.; Winkler, E.; Mazkereth, R.; Orenstein, A.; Bar-Meir, E.; Ravassard, P.; Meivar-Levy, I.; et al. Ectopic PDX-1 expression directly reprograms human keratinocytes along pancreatic insulin-producing cells fate. PLoS ONE 2011, 6, e26298. [Google Scholar] [CrossRef] [PubMed]
  184. Dambal, S.; Shah, M.; Mihelich, B.; Nonn, L. The microRNA-183 cluster: The family that plays together stays together. Nucleic Acids Res. 2015, 43, 7173–7188. [Google Scholar] [CrossRef] [PubMed]
  185. Liu, Y.; Ruan, H.; Lu, F.; Peng, H.; Luan, W. miR-224-5p acts as a tumour suppressor and reverses the resistance to BRAF inhibitor in melanoma through directly targeting PAK4 to block the MAPK pathway. Pathol. Res. Pract. 2023, 249, 154772. [Google Scholar] [CrossRef] [PubMed]
  186. Li, C.; Li, Y.; Li, Y.; Wang, Y.; Teng, Y.; Hao, Y. MicroRNA-194-5p attenuates hypoxia/reoxygenation-induced apoptosis in H9C2 cardiomyocytes by inhibiting the over-activation of RAC1 protein. Mol. Med. Rep. 2022, 27, 33. [Google Scholar] [CrossRef]
  187. Kapplingattu, S.V.; Bhattacharya, S.; Adlakha, Y.K. MiRNAs as major players in brain health and disease: Current knowledge and future perspectives. Cell Death Discov. 2025, 11, 7. [Google Scholar] [CrossRef]
  188. Kent, O.A.; McCall, M.N.; Cornish, T.C.; Halushka, M.K. Lessons from miR-143/145: The importance of cell-type localization of miRNAs. Nucleic Acids Res. 2014, 42, 7528–7538. [Google Scholar] [CrossRef]
  189. You, Y.-H.; Wang, X.-G.; Xu, M.; Zhao, J.-P. Expression and clinical significance of miR-139-5p in non-small cell lung cancer. J. Int. Med. Res. 2019, 47, 867–874. [Google Scholar] [CrossRef]
  190. Zeineddine, D.; Hammoud, A.A.; Mortada, M.; Boeuf, H. The Oct4 protein: More than a magic stemness marker. Am. J. Stem Cells 2014, 3, 74–82. [Google Scholar]
  191. Sterneckert, J.; Hoing, S.; Scholer, H.R. Concise review: Oct4 and more: The reprogramming expressway. Stem Cells 2012, 30, 15–21. [Google Scholar] [CrossRef]
  192. Mitchell, R.; Szabo, E.; Shapovalova, Z.; Aslostovar, L.; Makondo, K.; Bhatia, M. Molecular evidence for OCT4-induced plasticity in adult human fibroblasts required for direct cell fate conversion to lineage specific progenitors. Stem Cells 2014, 32, 2178–2187. [Google Scholar] [CrossRef]
  193. Edwards, P.C.; Mason, J.M. Gene-enhanced tissue engineering for dental hard tissue regeneration: (2) dentin-pulp and periodontal regeneration. Head Face Med. 2006, 2, 16. [Google Scholar] [CrossRef]
  194. Movahed, A.Y.; Bagheri, R.; Savatier, P.; Šarić, T.; Moradi, S. Elimination of tumorigenic pluripotent stem cells from their differentiated cell therapy products: An important step toward ensuring safe cell therapy. Stem Cell Rep. 2025, 20, 102543. [Google Scholar] [CrossRef] [PubMed]
  195. Lee, M.-O.; Moon, S.H.; Jeong, H.-C.; Yi, J.-Y.; Lee, T.-H.; Shim, S.H.; Rhee, Y.-H.; Lee, S.-H.; Oh, S.-J.; Lee, M.-Y.; et al. Inhibition of pluripotent stem cell-derived teratoma formation by small molecules. Proc. Natl. Acad. Sci. USA 2013, 110, E3281–E3290. [Google Scholar] [CrossRef] [PubMed]
  196. Nakao, H.; Yamaguchi, T.; Kawabata, K.; Higashi, K.; Nonaka, M.; Tuiji, M.; Nagai, Y.; Toyoda, H.; Yamaguchi, Y.; Kawasaki, N.; et al. Characterization of novel antibodies that recognize sialylated keratan sulfate and lacto-N-fucopentaose I on human induced pluripotent cells: Comparison with existing antibodies. Glycobiology 2023, 33, 150–164. [Google Scholar] [CrossRef] [PubMed]
  197. Kojima, K.; Miyoshi, H.; Nagoshi, N.; Kohyama, J.; Itakura, G.; Kawabata, S.; Ozaki, M.; Iida, T.; Sugai, K.; Ito, S.; et al. Selective ablation of tumorigenic cells following human induced pluripotent stem cell-derived neural stem/progenitor cell transplantation in spinal cord injury. Stem Cells Transl. Med. 2018, 8, 260–270. [Google Scholar] [CrossRef]
  198. Tamaoki, N.; Takahashi, K.; Tanaka, T.; Ichisaka, T.; Aoki, H.; Takeda-Kawaguchi, T.; Iida, K.; Kunisada, T.; Shibata, T.; Yamanaka, S.; et al. Dental pulp cells for induced pluripotent stem cell banking. J. Dent. Res. 2010, 89, 773–778. [Google Scholar] [CrossRef]
  199. Yan, X.; Qin, H.; Qu, C.; Tuan, R.S.; Shi, S.; Huang, G.T.-J. iPS cells reprogrammed from human mesenchymal-like stem/progenitor cells of dental tissue, origin. Stem Cells Dev. 2010, 19, 469–480. [Google Scholar] [CrossRef]
  200. Lizier, N.F.; Kerkis, I.; Wenceslau, C.V. Generation of induced pluripotent stem cells from dental pulp somatic cells. In Pluripotent Stem Cells; Bhartiya, D., Lenka, N., Eds.; InTechOpen: Rijeka, Croatia, 2013; pp. 131–149. [Google Scholar] [CrossRef]
  201. Iida, K.; Takeda-Kawaguchi, T.; Hada, M.; Yuriguchi, M.; Aoki, H.; Tamaoki, N.; Hatakeyama, D.; Kunisada, T.; Shibata, T.; Tezuka, K. Hypoxia-enhanced derivation of iPSCs from human dental pulp cells. J. Dent. Res. 2013, 92, 905–910. [Google Scholar] [CrossRef]
  202. Takeda-Kawaguchi, T.; Sugiyama, K.; Chikusa, S.; Iida, K.; Aoki, H.; Tamaoki, N.; Hatakeyama, D.; Kunisada, T.; Shibata, T.; Fusaki, N.; et al. Derivation of iPSCs after culture of human dental pulp cells under defined conditions. PLoS ONE 2014, 9, e115392. [Google Scholar] [CrossRef] [PubMed]
  203. Malhotra, N. Induced pluripotent stem (iPS) cells in dentistry: A review. Int. J. Stem Cells. 2016, 9, 176–185. [Google Scholar] [CrossRef] [PubMed]
  204. Hamano, S.; Sugiura, R.; Yamashita, D.; Tomokiyo, A.; Hasegawa, D.; Maeda, H. Current application of iPS cells in the dental tissue regeneration. Biomedicines 2022, 10, 3269. [Google Scholar] [CrossRef] [PubMed]
  205. Li, Y.; Shen, Z.; Shelat, H.; Geng, Y.-J. Reprogramming somatic cells to pluripotency: A fresh look at Yamanaka’s model. Cell Cycle 2013, 12, 3594–3598. [Google Scholar] [CrossRef]
  206. Singh, V.K.; Kalsan, M.; Kumar, N.; Saini, A.; Chandra, R. Induced pluripotent stem cells: Applications in regenerative medicine, disease modeling, and drug discovery. Front. Cell Dev. Biol. 2015, 3, 2. [Google Scholar] [CrossRef]
  207. Soda, M.; Saitoh, I.; Murakami, T.; Inada, E.; Iwase, Y.; Noguchi, H.; Shibasaki, S.; Kurosawa, M.; Sawami, T.; Terunuma, M.; et al. Repeated human deciduous tooth-derived dental pulp cell reprogramming factor transfection yields multipotent intermediate cells with enhanced iPS cell formation capability. Sci. Rep. 2019, 9, 1490. [Google Scholar] [CrossRef]
  208. Chin, M.H.; Mason, M.J.; Xie, W.; Volinia, S.; Singer, M.; Peterson, C.; Ambartsumyan, G.; Aimiuwu, O.; Richter, L.; Zhang, J.; et al. Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell Stem Cell 2009, 5, 111–123. [Google Scholar] [CrossRef]
  209. Chandrabose, S.T.; Sriram, S.; Subramanian, S.; Cheng, S.; Ong, W.K.; Rozen, S.; Kasim, N.H.A.; Sugii, S. Amenable epigenetic traits of dental pulp stem cells underlie high capability of xeno-free episomal reprogramming. Stem Cell Res. Ther. 2018, 9, 68. [Google Scholar] [CrossRef]
  210. Andrews, P.W.; Gokhale, P.J. A short history of pluripotent stem cells markers. Stem Cell Rep. 2024, 19, 1–10. [Google Scholar] [CrossRef]
  211. Russell, A.J.; Silpa, L. Chemical-induced naive pluripotency. Cell Chem. Biol. 2016, 23, 532–534. [Google Scholar] [CrossRef]
  212. Inada, E.; Saitoh, I.; Kubota, N.; Iwase, Y.; Murakami, T.; Sawami, T.; Yamasaki, Y.; Sato, M. Increased expression of cell surface SSEA-1 is closely associated with naïve-like conversion from human deciduous teeth dental pulp cells-derived iPS cells. Int. J. Mol. Sci. 2019, 20, 1651. [Google Scholar] [CrossRef]
  213. Nakashima, Y.; Miyagi-Shiohira, C.; Saitoh, I.; Watanabe, M.; Matsushita, M.; Tsukahara, M.; Noguchi, H. Induced hepatic stem cells are suitable for human hepatocyte production. iScience 2022, 25, 105052. [Google Scholar] [CrossRef]
  214. Saitoh, I.; Sato, M.; Soda, M.; Inada, E.; Iwase, Y.; Murakami, T.; Ohshima, H.; Hayasaki, H.; Noguchi, H. Tissue-specific stem cells obtained by reprogramming of non-obese diabetic (NOD) mouse-derived pancreatic cells confer insulin production in response to glucose. PLoS ONE 2016, 11, e0163580. [Google Scholar] [CrossRef]
  215. Noguchi, H.; Miyagi-Shiohira, C.; Nakashima, Y.; Onishi, Y.; Saitoh, I.; Watanabe, M. Establishment of induced pancreatic stem cells by Yes-associated protein 1. Cell Transplant. 2024, 33, 09636897241248942. [Google Scholar] [CrossRef]
  216. Kiyokawa, Y.; Sato, M.; Noguchi, H.; Inada, E.; Iwase, Y.; Kubota, N.; Sawami, T.; Terunuma, M.; Maeda, T.; Hayasaki, H.; et al. Drug-induced naïve iPS cells exhibit better performance than primed iPS cells with respect to the ability to differentiate into pancreatic β-cell lineage. J. Clin. Med. 2020, 9, 2838. [Google Scholar] [CrossRef]
  217. Lee, S.-W.; Lee, H.J.; Sung, H.H.; Ko, K.; Han, D.W.; Ko, K. Optimization of Matrigel-based culture for expansion of neural stem cells. Anim. Cells Syst. 2015, 19, 175–180. [Google Scholar] [CrossRef]
  218. Li, L.; Bennett, S.A.L.; Wang, L. Role of E-cadherin and other cell adhesion molecules in survival and differentiation of human pluripotent stem cells. Cell Adh. Migr. 2012, 6, 59–70. [Google Scholar] [CrossRef]
  219. Ibano, N.; Inada, E.; Otake, S.; Kiyokawa, Y.; Sakata, K.; Sato, M.; Kubota, N.; Noguchi, H.; Iwase, Y.; Murakami, T.; et al. The role of genetically modified human feeder cells in maintaining the integrity of primary cultured human deciduous dental pulp cells. J. Clin. Med. 2022, 11, 6087. [Google Scholar] [CrossRef] [PubMed]
  220. Saitoh, I.; Inada, E.; Iwase, Y.; Noguchi, H.; Murakami, T.; Soda, M.; Kubota, N.; Hasegawa, H.; Akasaka, E.; Matsumoto, Y.; et al. Choice of feeders is important when first establishing iPSCs derived from primarily cultured human deciduous tooth dental pulp cells. Cell Med. 2015, 8, 9–23. [Google Scholar] [CrossRef] [PubMed]
  221. Javaid, D.; Ganie, S.Y.; Hajam, Y.A.; Reshi, M.S. CRISPR/Cas9 system: A reliable and facile genome editing tool in modern biology. Mol. Biol. Rep. 2022, 49, 12133–12150. [Google Scholar] [CrossRef] [PubMed]
  222. Yu, N.; Yang, J.; Mishina, Y.; Giannobile, W.V. Genome editing: A new horizon for oral and craniofacial research. J. Dent. Res. 2019, 98, 36–45. [Google Scholar] [CrossRef]
  223. Bales, L.E. CRISPR-Cas in the Field of Dentistry: A Comprehensive Collection of the Potential Uses of CRISPR-Cas9 in Dental Health Care. Chancellor’s Honors Program Projects. 2022. Available online: https://trace.tennessee.edu/utk_chanhonoproj/2499 (accessed on 15 January 2025).
  224. Bikard, D.; Euler, C.W.; Jiang, W.; Nussenzweig, P.M.; Goldberg, G.W.; Duportet, X.; Fischetti, V.A. Exploiting CRISPR-Cas nucleases to produce sequence-specific antimicrobials. Nat. Biotechnol. 2014, 32, 1146–1150. [Google Scholar] [CrossRef]
  225. Gong, T.; Tang, B.; Zhou, X.; Zeng, J.; Lu, M.; Guo, X.; Peng, X.; Lei, L.; Gong, B.; Li, Y. Genome editing in Streptococcus mutans through self-targeting CRISPR arrays. Mol. Oral Microbiol. 2018, 33, 440–449. [Google Scholar] [CrossRef]
  226. Stern, A.; Keren, L.; Wurtzel, O.; Amitai, G.; Sorek, R. Self-targeting by CRISPR: Gene regulation or autoimmunity? Trends Genet. 2010, 26, 335–340. [Google Scholar] [CrossRef]
  227. Kato, Y.; Yokose, S. Oxytocin facilitates dentinogenesis of rat dental pulp cells. J. Endod. 2021, 47, 592–599. [Google Scholar] [CrossRef]
  228. Zhang, Y.; Yang, J.; Yao, H.; Zhang, Z.; Song, Y. CRISPR/Cas9-mediated deletion of Fam83h induces defective tooth mineralization and hair development in rabbits. J. Cell. Mol. Med. 2022, 26, 5670–5679. [Google Scholar] [CrossRef] [PubMed]
  229. Liu, B.; Zhang, C.; Zhao, H.; Gao, J.; Hu, J. Chitosan hydrogel-delivered ABE8e corrects PAX9 mutant in dental pulp stem cells. Gels 2023, 9, 436. [Google Scholar] [CrossRef] [PubMed]
  230. Molla, K.A.; Yang, Y. CRISPR/Cas-mediated base editing: Technical considerations and practical applications. Trends Biotechnol. 2019, 37, 1121–1142. [Google Scholar] [CrossRef] [PubMed]
  231. Sheriff, A.; Guri, I.; Zebrowska, P.; Llopis-Hernandez, V.; Brooks, I.R.; Tekkela, S.; Subramaniam, K.; Gebrezgabher, R.; Naso, G.; Petrova, A.; et al. ABE8e adenine base editor precisely and efficiently corrects a recurrent COL7A1 nonsense mutation. Sci. Rep. 2022, 12, 19643. [Google Scholar] [CrossRef]
  232. Rehman, A.; Panda, S.K.; Torsiello, M.; Marigliano, M.; Tufano, C.C.; Nigam, A.; Parveen, Z.; Papaccio, G.; La Noce, M. The crosstalk between primary MSCs and cancer cells in 2D and 3D cultures: Potential therapeutic strategies and impact on drug resistance. Stem Cells Transl. Med. 2024, 13, 1178–1185. [Google Scholar] [CrossRef]
  233. Nikkhah, E.; Kalalinia, F.; Rezaee, M.A.; Tayarani-Najaran, Z. Suppressive effects of dental pulp stem cells and its conditioned medium on development and migration of colorectal cancer cells through MAPKinase pathways. Iran. J. Basic Med. Sci. 2021, 24, 1292–1300. [Google Scholar] [CrossRef]
  234. He, Y.; Li, R.; She, W.; Ai, Y.; Li, K.; Kumeria, T.; Jiang, Z.; Shao, Q.; Zou, C.; Albashari, A.A.; et al. Inhibitory effects of the nanoscale lysate derived from xenogenic dental pulp stem cells in lung cancer models. J. Nanobiotechnol. 2023, 21, 488. [Google Scholar] [CrossRef] [PubMed]
  235. Hong, P.; Yang, H.; Wu, Y.; Li, K.; Tang, Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: A comprehensive review. Stem Cell Res. Ther. 2019, 10, 242. [Google Scholar] [CrossRef] [PubMed]
  236. Wong, R.S.Y.; Tan, T.; Pang, A.S.R.; Srinivasan, D.K. The role of cytokines in wound healing: From mechanistic insights to therapeutic applications. Explor. Immunol. 2025, 5, 1003183. [Google Scholar] [CrossRef]
  237. Yi, J.; Tang, Q.; Sun, S.; Xie, H.; Wang, L.; Yin, X. Exosomes in diabetic wound healing: Mechanisms, applications, and perspectives. Diabetes Metab. Syndr. Obes. 2025, 18, 2955–2976. [Google Scholar] [CrossRef] [PubMed]
  238. Roszkowski, S. Therapeutic potential of mesenchymal stem cell-derived exosomes for regenerative medicine applications. Clin. Exp. Med. 2024, 24, 46. [Google Scholar] [CrossRef] [PubMed]
  239. Zhang, C.; Deng, R.; Zhang, G.; He, X.; Chen, H.; Chen, B.; Wan, L.; Kang, X. Therapeutic effect of exosomes derived from stem cells in spinal cord injury: A systematic review based on animal studies. Front. Neurol. 2022, 13, 847444. [Google Scholar] [CrossRef]
  240. Song, N.; Scholtemeijer, M.; Shah, K. Mesenchymal stem cell immunomodulation: Mechanisms and therapeutic potential. Trends Pharmacol. Sci. 2020, 41, 653–664. [Google Scholar] [CrossRef]
  241. 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]
  242. Yoshida, S.; Tomokiyo, A.; Hasegawa, D.; Hamano, S.; Sugii, H.; Maeda, H. Insight into the role of dental pulp stem cells in regenerative therapy. Biology 2020, 9, 160. [Google Scholar] [CrossRef]
  243. 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]
  244. Demircan, P.C.; Sariboyaci, A.E.; Unal, Z.S.; Gacar, G.; Subasi, C.; Karaoz, E. Immunoregulatory effects of human dental pulp-derived stem cells on T cells: Comparison of transwell co-culture and mixed lymphocyte reaction systems. Cytotherapy 2011, 13, 1205–1220. [Google Scholar] [CrossRef]
  245. 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]
  246. Hata, M.; Omi, M.; Kobayashi, Y.; Nakamura, N.; Miyabe, M.; Ito, M.; Makino, E.; Kanada, S.; Saiki, T.; Ohno, T.; et al. Transplantation of human dental pulp stem cells ameliorates diabetic polyneuropathy in streptozotocin-induced diabetic nude mice: The role of angiogenic and neurotrophic factors. Stem Cell Res. Ther. 2020, 11, 236. [Google Scholar] [CrossRef] [PubMed]
  247. El-Kersh, A.O.F.O.; El-Akabawy, G.; Al-Serwi, R.H. Transplantation of human dental pulp stem cells in streptozotocin-induced diabetic rats. Anat. Sci. Int. 2020, 95, 523–539. [Google Scholar] [CrossRef]
  248. Inada, R.; Mendoza, H.Y.; Tanaka, T.; Horie, T.; Satomi, T. Preclinical study for the treatment of diabetes mellitus using β-like cells derived from human dental pulp stem cells. Regen. Med. 2022, 17, 905–913. [Google Scholar] [CrossRef] [PubMed]
  249. Mead, B.; Logan, A.; Berry, M.; Leadbeater, W.; Scheven, B.A. Intravitreally transplanted dental pulp stem cells promote neuroprotection and axon regeneration of retinal ganglion cells after optic nerve injury. Investig. Ophthalmol. Vis. Sci. 2013, 54, 7544–7556. [Google Scholar] [CrossRef]
  250. Sowa, K.; Nito, C.; Nakajima, M.; Ueda, M.; Kimura, K.; Okada, T. Impact of dental pulp stem cells overexpressing hepatocyte growth factor after cerebral ischemia/reperfusion in rats. Mol. Ther. Methods Clin. Dev. 2018, 10, 281–290. [Google Scholar] [CrossRef]
  251. Hilkens, P.; Bronckaers, A.; Ratajczak, J.; Gervois, P.; Wolfs, E.; Lambrichts, I. The angiogenic potential of DPSCs and SCAPs in an in vivo model of dental pulp regeneration. Stem Cells Int. 2017, 2017, 2582080. [Google Scholar] [CrossRef]
  252. Samiei, M.; Harmsen, M.C.; Abdolahinia, E.D.; Barar, J.; Petridis, X. Scaffold-free strategies in dental pulp/dentine tissue engineering: Current status and implications for regenerative biological processes. Bioengineering 2025, 12, 198. [Google Scholar] [CrossRef]
  253. Mohd, N.; Razali, M.; Fauzi, M.B.; Kasim, N.H.A. In vitro and in vivo biological assessments of 3D-bioprinted scaffolds for dental applications. Int. J. Mol. Sci. 2023, 24, 12881. [Google Scholar] [CrossRef]
  254. Lacruz, R.S.; Habelitz, S.; Wright, J.T.; Paine, M.L. Dental enamel formation and implications for oral health and disease. Physiol. Rev. 2017, 97, 939–993. [Google Scholar] [CrossRef] [PubMed]
  255. Kim, E.J.; Mai, H.N.; Lee, D.J.; Kim, K.H.; Lee, S.J.; Jung, H.S. Strategies for differentiation of hiPSCs into dental epithelial cell lineage. Cell Tissue Res. 2021, 386, 415–421. [Google Scholar] [CrossRef] [PubMed]
  256. Kim, E.-J.; Kim, K.-H.; Kim, H.-Y.; Lee, D.-J.; Li, S.; Han, M.N.; Jung, H.-S. Harnessing the dental cells derived from human induced pluripotent stem cells for hard tissue engineering. J. Adv. Res. 2023, 61, 119–131. [Google Scholar] [CrossRef] [PubMed]
  257. Kim, E.-J.; Yoon, K.-S.; Arakaki, M.; Otsu, K.; Fukumoto, S.; Harada, H.; Green, D.W.; Lee, J.-M.; Jung, H.-S. Effective differentiation of induced pluripotent stem cells into dental cells. Dev. Dyn. 2019, 248, 129–139. [Google Scholar] [CrossRef]
  258. Kim, G.-H.; Yang, J.; Jeon, D.-H.; Kim, J.-H.; Chae, G.Y.; Jang, M.; Lee, G. Differentiation and establishment of dental epithelial-like stem cells derived from human ESCs and iPSCs. Int. J. Mol. Sci. 2020, 21, 4384. [Google Scholar] [CrossRef]
  259. Miao, X.; Niibe, K.; Fu, Y.; Zhang, M.; Nattasit, P.; Ohori-Morita, Y.; Nakamura, T.; Jiang, X.; Egusa, H. Epiprofin transcriptional activation promotes ameloblast induction from mouse induced pluripotent stem cells via the BMP-Smad signaling axis. Front. Bioeng. Biotechnol. 2022, 10, 890882. [Google Scholar] [CrossRef]
  260. Hemeryck, L.; Hermans, F.; Chappell, J.; Kobayashi, H.; Lambrechts, D.; Lambrichts, I.; Bronckaers, A.; Vankelecom, H. Organoids from human tooth showing epithelial stemness phenotype and differentiation potential. Cell. Mol. Life Sci. 2022, 79, 153. [Google Scholar] [CrossRef]
  261. Kim, K.-H.; Kim, E.-J.; Kim, H.-Y.; Li, S.; Jung, H.-S. Fabrication of functional ameloblasts from hiPSCs for dental application. Front. Cell Dev. Biol. 2023, 11, 1164811. [Google Scholar] [CrossRef]
  262. Zielińska, A.; Karczewski, J.; Eder, P.; Kolanowski, T.; Szalata, M.; Wielgus, K.; Szalata, M.; Kim, D.; Shin, S.R.; Słomski, R.; et al. Scaffolds for drug delivery and tissue engineering: The role of genetics. J. Control Release 2023, 359, 207–223. [Google Scholar] [CrossRef]
  263. dos Santos, L.R.K.; Pelegrine, A.A.; da Silveira Bueno, C.E.; Ferreira, J.R.M.; Aloise, A.C.; Stringheta, C.P.; Martinez, E.F.; Pelegrine, R.A. Pulp–dentin complex regeneration with cell transplantation technique using stem cells derived from human deciduous teeth: Histological and immunohistochemical study in immunosuppressed rats. Bioengineering 2023, 10, 610. [Google Scholar] [CrossRef]
Figure 1. Structure of a human tooth. Dental pulp is enclosed in the dental cavity of each tooth, called the “pulp chamber,” and contains various types of cells, including fibroblasts, endothelial cells, neurons, odonto/osteo progenitors, and inflammatory and immune cells. Cells in the dental pulp are called “DPCs” and are highly enriched with stem cells called “DPSCs” with the capacity of self-renewal, rapid proliferation, multilineage differentiation, and pluripotent gene expression profile. The figures were created in-house using Adobe Illustrator version 12 (Illustrator CS2) (Adobe Inc., San Jose, CA, USA).
Figure 1. Structure of a human tooth. Dental pulp is enclosed in the dental cavity of each tooth, called the “pulp chamber,” and contains various types of cells, including fibroblasts, endothelial cells, neurons, odonto/osteo progenitors, and inflammatory and immune cells. Cells in the dental pulp are called “DPCs” and are highly enriched with stem cells called “DPSCs” with the capacity of self-renewal, rapid proliferation, multilineage differentiation, and pluripotent gene expression profile. The figures were created in-house using Adobe Illustrator version 12 (Illustrator CS2) (Adobe Inc., San Jose, CA, USA).
Biotech 14 00088 g001
Figure 2. Summary of recent achievements in gene-engineered DPSCs.
Figure 2. Summary of recent achievements in gene-engineered DPSCs.
Biotech 14 00088 g002
Figure 3. Schematic illustration of strategies for generating hard tissue from human iPSCs using extracellular matrix (ECM) microenvironments, such as gelatin methacryloyl (GelMA), agar, and collagen gel. Dental epithelial cells (DECs) and dental mesenchymal cells (DMCs) are first induced to differentiate from human iPSCs using specific humoral factors. To create bioengineered tooth-like constructs, these two types of cells are combined in the presence of various scaffolds, such as GelMA, agar, and collagen gel. The resulting mixture is subsequently embedded into a 3D-printed hydroxyapatite scaffold or deproteinized tooth scaffold, followed by transplantation beneath the renal capsule or under the skin of immunocompromised animals for further maturation. The figures were created in-house using Adobe Illustrator version 12 (Illustrator CS2) (Adobe Inc., San Jose, CA, USA) based on Kim et al. [256].
Figure 3. Schematic illustration of strategies for generating hard tissue from human iPSCs using extracellular matrix (ECM) microenvironments, such as gelatin methacryloyl (GelMA), agar, and collagen gel. Dental epithelial cells (DECs) and dental mesenchymal cells (DMCs) are first induced to differentiate from human iPSCs using specific humoral factors. To create bioengineered tooth-like constructs, these two types of cells are combined in the presence of various scaffolds, such as GelMA, agar, and collagen gel. The resulting mixture is subsequently embedded into a 3D-printed hydroxyapatite scaffold or deproteinized tooth scaffold, followed by transplantation beneath the renal capsule or under the skin of immunocompromised animals for further maturation. The figures were created in-house using Adobe Illustrator version 12 (Illustrator CS2) (Adobe Inc., San Jose, CA, USA) based on Kim et al. [256].
Biotech 14 00088 g003
Table 1. Comparison of the functions between DPSCs and BMSCs.
Table 1. Comparison of the functions between DPSCs and BMSCs.
FeatureDPSC (Dental Pulp Stem Cell)BMSC (Bone Marrow Stem Cells)
OriginNeural crest originates from the dental pulp tissue.Mesodermal origin from the bone marrow.
Multilineage potentialCan differentiate into multiple cell neural lineages (osteogenic, adipogenic, chondrogenic) plus specific odontogenic and neurogenic differentiation.Can differentiate into multiple cell neural lineages (osteogenic, adipogenic, chondrogenic), with a strong predisposition for osteogenic differentiation.
Surface markersTypically positive for STRO-1, CD29, CD44, CD73, CD90, CD105, CD59, and CD106.Typically positive for CD29, CD44, CD73, CD90, CD105, CD166, and CD146; lower STRO-1 expression than DPSCs.
Proliferation rateHigh proliferation rate and clonogenic potential. Slower proliferation rate compared to DPSCs.
Table 2. Summary of differentiation induction of human DPSCs by the methods based on non-gene-engineered approaches.
Table 2. Summary of differentiation induction of human DPSCs by the methods based on non-gene-engineered approaches.
Type of Differentiated Cells, Originating from DPSCsMethods for Differentiation InductionMarkers or Staining MethodReferences
Osteogenic differentiationCultivation in medium containing dexamethasone, L-ascorbic acid, and β-glycerol phosphate; co-cultivation with endothelium; cultivation in the presence of human serum, concentrated growth factor exudate (CGFe), and TGF-β1 or betaine; cultivation with scaffolds, decellularised adipose tissue solid foams, or microcapsulesAlkaline phosphatase (ALP)
Collagen type I (COL I)
Osteocalcin (OCN)
Osteonectin (ON)
Osteopontin (OPN)
Osterix (OSX)
Runt-related transcription factor 2 (RUNX2)
[47,48,49,50,51,52,53,54,55,56,57,58]
Odontogenic differentiationCultivation in medium containing TGF-β1, dexamethasone, β-glycerophosphate, and L-ascorbic acid;
cultivating on dentin, calcium silicate materials, or scaffolds; cultivation in preameloblast-conditioned medium or medium containing endothelin-1 (ET-1) or vascular endothelial growth factor A (VEGFA)
ALP
Dentin sialoprotein (DSP)
Dentin sialophosphoprotein (DSPP),
Dentin matrix protein 1 (DMP-1)
Matrix extracellular phosphoglycoprotein (MEPE)
Alizarin red S staining
[59,60,61,62,63,64,65,66]
Adipogenic differentiationCultivation in medium containing insulin, dexamethasone, indomethacin, and 3-isobutyl-1-methylxanthine (IBMX); cultivation in the presence of enzymatically decellularized adipose tissue solid foams Peroxisome proliferator-activated receptor γ (PPAR-γ)
Glucose transporter type 4 (GLUT4)
Fatty acid binding protein 4 (FABP4)
Lipoprotein lipase (LPL)
Oil red O staining
[52,67,68,69,70]
Neurogenic differentiationCultivation in neuroinduction medium containing B27, L-glutamine, basic FGF, and EGF for 7 days, and subsequently in neuroinduction medium supplemented with retinoic acid for another 7 days; cultivation in commercially available neurogenic differentiation medium, conditioned medium of cerebrospinal fluid and retinoic acid, or medium containing nerve growth factor (NGF); culturing in the presence of graphene–polycaprolactone hybrid nanofibers or highly concentrated K+ (50 mM KCl) for K+ stimulation Neuronal nuclei (NeuN)
Microtubule-associated protein 2 (MAP2)
Neural cell adhesion molecule (NCAM)
Growth-associated protein 43 (GAP43)
Glial fibrillary acid protein (GFAP)
Synapsin I (SYN1)
Neuron-specific class III beta-tubulin (TUBB3)
Gamma-aminobutyric acid (GABA receptors)
Enolase 2/neuron-specific enolase (ENO2/NSE)
Nestin (NES)
Peripherin (PRPH)
[71,72,73,74,75,76,77,78]
Hepatogenic differentiationCultivation in hepatoinduction medium containing hepatic growth factor, insulin-transferrin-selenium-x, dexamethasone, and oncostatin MAlpha fetoprotein (AFP)
Albumin (ALB)
Hepatic nuclear factor-4 alpha (HNF4α)
Insulin-like growth factor-1 (IGF-1)
Carbamoyl phosphate synthetase (CPS)
[79]
Angiogenic differentiationCultivation in a medium supplemented with a mixture of B27, heparin, and growth factors, including vascular endothelial growth factor (VEGF)-A165 or VEGF alone; co-cultivation with human umbilical vein endothelial cells (HUVECs) after encapsulation by a scaffold system, called self-assembling peptide nanofibersCD54/intercellular adhesion molecule-1 (ICAM-1)
CD146/melanoma cell adhesion molecule (MCAM)
Monocyte chemotactic protein-1 (MCP-1)
von-Willebrand factor (VWF) (domains 1 and 2)
CD31/platelet/endothelial cell adhesion molecule-1 (PECAM-1)
Vascular endothelial growth factor (VEGF)
CD34
Functioning legal knowledge 1 (Flk-1)
Vascular endothelial growth factor receptor 2 (VEGFR-2)
Fibroblast growth factor 2 (FGF-2)
Insulin-like growth factor binding protein-3 (IGFBP3)
Interleukin-8 (IL-8)
Plasminogen activator inhibitor-1 (PAI-1)
Tissue inhibitors of metalloproteinase-1 (TIMP-1)
Urokinase-type plasminogen activator (uPA)
[80,81,82,83]
Cardiogenic differentiationDPSCs are first incubated in the presence of 5-azacytidine for 2 days; then, the resulting embryoid bodies (EBs) are plated onto gelatin-coated tissue culture dishes, through which functional cardiomyocytes with cardiac markers are developedMyosin heavy chain 6 (MYH6)
Mesoderm posterior BHLH transcription factor 1 (MESP)
NK-2 transcription factor related, locus 5 (Nkx2.5)
Connexin 43 (Cx43)
[84]
Differentiation into pancreatic lineageCD117+ DPCs have the ability to differentiate into pancreatic lineage, following the 3-step induction protocol used for pancreatic cell lineage induction; cultivation of DPSCs using a stepwise protocol to generate islet-like cell clusters; cultivation of DPSCs in a 3D culture system using a stepwise protocol to generate organoid-like 3D structures that are similar to pancreatic isletsGlucose transporter 2 (GLUT2)
Pancreatic forkhead box protein A2 (Foxa2)
SRY-box transcription factor 17 (Sox17)
Insulin-like growth factor I (IGF-1)
Fibroblast growth factor 10 (FGF 10)
Pancreatic and duodenal homeobox 1 (PDX1)
Hematopoietically expressed homeobox (HHEX)
Motor neuron and pancreas homeobox 1 (MNX1)
Neurogenin 3 (NGN3)
Paired box 4 (PAX4)
Paired box 6 (PAX6)
NK6 homeobox 1 (NKX6-1)
Blood glucose
Serum insulin (INS)
c-peptide (CP)
Visfatin (VF)
Pancreatic glucagon (GC)
Somatostatin (SS)
Pancreatic polypeptide, Amylase-2a (AMY2A)
[85,86,87,88,89,90,91,92]
Differentiation into bladder smooth muscleIncubation of DPSCs in bladder smooth muscle cell-conditioned medium with transforming growth factor beta 1 (TGF-β1) for 2 weeks; incubation of DPSCs with direct contact with endothelial cells in the presence of TGF-β1Alpha smooth muscle actin (α-SMA)
Smooth muscle protein 22α (SM22α)
Smooth muscle myosin heavy chain (SM-MHC)
Desmin (DES)
Calponin (CNN1)
[93,94,95]
Table 3. Summary of experiments using gene-engineered DPSCs showing gain-of-function (via overexpression) or knockdown-of-function (via RNAi or microRNA (miR)).
Table 3. Summary of experiments using gene-engineered DPSCs showing gain-of-function (via overexpression) or knockdown-of-function (via RNAi or microRNA (miR)).
Biological SystemsType of Gene ExpressionTarget Gene or Transgene
(Method for Gene Delivery)
Cell
Species
OutcomeReferences
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)Growth/differentiation factor 11 (Gdf11)
(electroporation)
MouseIn vivo transfer of Gdf11 stimulated the reparative dentin formation during pulpal wound healing in canine teeth[105]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)Jagged canonical Notch ligand 1 (Jagged-1; JAG-1)
(retrovirus)
HumanOverexpression of JAG-1 caused activation of the Notch signaling pathway, inhibition of odontoblastic differentiation, and the formation of mineralized tissues[119]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)Krüppel-like factor 4 (KLF4)
(plasmid, FuGENE)
HumanOverexpression of KLF4 caused increased alkaline phosphatase (ALP) activity and expression of odontoblastic differentiation markers[120]
Differentiation into odontoblast/mineralizationSmall interfering RNA (siRNA)-based knockdownNotch ligand Delta1
(lentivirus)
HumanSuppression of Notch ligand Delta1 resulted in inhibition of proliferation but increased differentiation into odontoblasts[121]
Differentiation into odontoblast/mineralizationmiR-based knockdownmiR-720
(lipofectamine)
HumanmiR-720, which targets NANOG, promoted odontogenic differentiation through suppression of NANOG[122]
Differentiation into odontoblast/mineralizationsiRNA-based knockdownCD44 [HCAM (homing cell adhesion molecule)]
(lentivirus)
HumanRNAi for CD44, which is expressed in odontogenic cells, resulted in suppression of mineralization activities[123]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)DNA damage-inducible transcript 3 (DDIT3)
(lentivirus)
HumanOverexpression of DDIT3, an apoptotic transcription factor, increased calcium nodule formation related to odontoblastic differentiation[124]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)
siRNA-based knockdown
Inhibitor of DNA binding 1 (ID1)
(lentivirus)
HumanOverexpression of ID1 resulted in an enhanced odontogenic differentiation; ID1 silencing produced an opposite effect[125]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)
siRNA-based knockdown
Zinc finger and BTB domain-containing 20 (ZBTB20)
(lentivirus)
HumanInhibition of ZBTB20 reduced odontogenic differentiation, while overexpression of ZBTB20 enhanced odontogenic differentiation along with increased ALP activity[126]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)B cell lymphoma 2 gene (BCL2)
(lentivirus)
HumanOverexpression of BCL2 caused decreased osteogenic/odontogenic potential[127]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)SRY-box 2 (SOX2)
(retrovirus)
HumanOverexpression of SOX2 resulted in enhanced odontoblast differentiation, which was also associated with the activation of the Wnt signaling pathway[128]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)Platelet-derived growth factor-BB
(PDGF-BB)
(lentivirus)
HumanPDGF-BB enhanced odontoblastic differentiation; subcutaneous grafting of PDGF-BB-expressing DPSCs generated dentin-like mineralized tissue[99]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)
siRNA-based knockdown
Sclerostin (SOST)
(lentivirus)
HumanOverexpression of SOST, a protein that acts as a negative regulator of bone formation, resulted in promoted senescence-related decline of odontoblastic differentiation potential; knockdown of SOST enhances odontoblastic differentiation [129]
Differentiation into odontoblast/mineralizationsiRNA-based knockdownBTB/POZ domain-containing protein 7 (BTBD7)
(lentivirus)
HumanKnockdown of BTBD7, a regulatory gene that promotes epithelial tissue remodeling and branching morphogenesis, resulted in reduced expression of odontoblast markers[130]
Differentiation into odontoblast/mineralizationmiR-based knockdownmiR-675
(lentivirus)
HumanOverexpression of miR-675, which is involved in odontogenic differentiation, resulted in enhancement of odontogenic differentiation[131]
Differentiation into odontoblast/mineralizationmiR-based knockdownmiR-508-5p
(lipofectamine)
HumanOverexpression of miR-508-5p, which targets the glycoprotein non-metastatic melanoma protein B (GPNMB) gene, resulted in suppression of odontogenesis; ectopic expression of GPNMB (lacking 3′-UTR) rescued the effects of miR-508-5p on odontogenic differentiation[132]
Differentiation into odontoblast/mineralizationmiR-based knockdownmiR-223-3p
(lentivirus)
HumanOverexpression of miR-223-3p, one of the inflammation-induced miRs, resulted in increased production of odontogenic markers but decreased the production of SMAD family member 3 (SMAD3), a potential target of miR-223-3p[133]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)Dentin matrix protein 1 (DMP-1)
(lentivirus)
HumanOverexpression of DMP-1 in the DPSCs derived from X-linked hypophosphatemia (XLH), associated with deficient dentin formation and mineralization, restored the irregular protein processing patterns to near-physiological levels[134]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)
siRNA-based
knockdown
Long noncoding RNA H19 (LncRNA H19)
(plasmid; lentivirus)
HumanOverexpression of LncRNA H19 promoted enhanced ALP activity and increased expression of odontogenic markers via regulating the TGF-β1/Smad signaling pathway[135]
Differentiation into odontoblast/mineralizationGain-of-function (overexpression)DMP-1
(lentivirus)
HumanExpression of DMP-1, a factor stimulating canonical Wnt signaling, in the XLH-DPSCs resulted in downregulation of Wnt inhibitors and improved mineralization[136]
Differentiation into odontoblast/mineralizationmiR-based
knockdown
miR-15b-5p
(lipofectamine)
HumanMiR-15b-5p suppressed the differentiation into odontoblasts by targeting insulin-like growth factor 1 (IGF1), while the miR-15b-5p inhibitor enhanced the differentiation into odontoblasts[137]
Osteogenic differentiation Gain-of-function (overexpression)BCL2
(lentivirus)
HumanOverexpression of BCL2 resulted in enhanced cell survivability because of the inhibition of apoptosis by BCL2; it also caused decreased osteogenic/odontogenic potential[127]
Osteogenic differentiationGain-of-function (overexpression)SOX2
(retrovirus)
HumanOverexpression of SOX2 resulted in enhanced osteogenic differentiation, together with the activation of the Hippo signal pathway [138]
Osteogenic differentiationGain-of-function (overexpression)Transforming growth factor beta 1 (TGF-β1)
(plasmid; electroporation)
HumanOverexpression of TGF-β1 resulted in increased osteogenic and chondrogenic differentiation; it also resulted in increased proliferation rate and decreased apoptosis[111]
Osteogenic differentiationGain-of-function (overexpression)Wnt family member 4 (WNT4)
(lentivirus)
HumanOverexpression of WNT4 exhibited effective repair of rat bone defects, suggesting that WNT4-expressing DPSCs may be a feasible resource of seed cells for bone regeneration[139]
Osteogenic differentiationGain-of-function (overexpression)EphrinB2 (EFNB2)
(lentivirus)
Human
canine
Overexpression of EFNB2 resulted in enhanced osteogenic differentiation capacity[101]
Osteogenic differentiationGain-of-function (overexpression)
miR-based
knockdown
hsa_circ_0026827
miR-188-3p
(plasmid; lipofectamine)
HumanOverexpression of hsa_circ_0026827, one of the circular RNAs (circRNAs), resulted in increased osteoblast differentiation, while knockdown of hsa_circ_0026827 suppressed osteoblast differentiation and promoted miR-188-3p expression[140]
Osteogenic differentiationGain-of-function (overexpression)Hypoxia-inducible factor 1α (HIF-1α)
(protein transduction domains (PTDs))
(lentivirus)
HumanOverexpression of HIF-1α, a protein known to be expressed in hypoxia and affect stemness and bone differentiation, resulted in enhanced osteogenic differentiation[141]
Osteogenic differentiationGain-of-function (overexpression)Semaphorin 3A (SEMA3A)
(lentivirus)
UnknownOverexpression of Sema3A, a secretory osteoprotective factor, exhibited enhanced the osteogenic differentiation [142]
Osteogenic differentiationGain-of-function (overexpression)
siRNA-based knockdown
Growth differentiation factor 15 (GDF15)
(lentivirus; lipofectamine)
HumanOverexpression of GDF15 caused enhanced osteogenic differentiation through activation of the TGF-β/SMAD signaling pathway, while knockdown of GDF15 produced the opposite effect[143]
Enhanced cell proliferationsiRNA-based knockdownCatalytic alpha1 of AMP-activated protein kinase (AMPK alpha1)
(siRNA; lipofectamine)
RatKnockdown of AMPKalpha1, a stress-responsive enzyme that is activated by hypoxia, resulted in decreased cell proliferation under both normoxia and hypoxia[144]
Enhanced cell proliferationsiRNA-based knockdownNotch ligand Delta1
(lentivirus)
HumanKnockdown of Notch ligand Delta1 resulted in inhibition of proliferation but increased differentiation into odontoblasts[121]
Enhanced cell proliferationmiRNA-based knockdownmiR-720
(lipofectamine)
HumanmiR-720, targeting NANOG, caused decreased proliferation and promoted odontogenic differentiation through suppression of NANOG[122]
Enhanced cell proliferationGain-of-function (overexpression)DDIT3
(lentivirus)
HumanOverexpression of DDIT3, an apoptotic transcription factor, resulted in reduced cell proliferation but increased calcium nodule formation related to odontoblastic differentiation[124]
Enhanced cell proliferationGain-of-function (overexpression)SOX2
(retrovirus)
HumanSOX2 overexpression resulted in the enhancement of cell proliferation, migration, and adhesion [145]
Enhanced cell proliferationmiR-based
knockdown
MiR-633
(lentivirus)
HumanmiR-633 overexpression increased cell proliferation and differentiation through direct interaction with the 3′-UTR of matrix extracellular phosphoglycoprotein (MEPE) gene[146]
Enhanced cell proliferationGain-of-function (overexpression)Special AT-rich binding protein 2 (SATB2)
(lentivirus)
HumanOverexpression of SATB2 resulted in accelerated proliferation and cell migration [147]
Enhanced cell proliferationGain-of-function (overexpression)PDGF-BB
(lentivirus)
HumanPDGF-BB caused enhanced proliferation, odontoblastic differentiation, and cell migration via the activation of the phosphatidylinositol 3 kinase (PI3K)/Akt signaling pathway[99]
Enhanced cell proliferationGain-of-function (overexpression)Vascular endothelial growth factor (VEGF)/
stromal cell-derived factor-1α (SDF-1α)
(lentivirus)
HumanOverexpression of SDF-1α or VEGF resulted in enhanced cell proliferation, endothelial cell migration, and vascular tube formation on Matrigel in vitro; expression of both VEGF and SDF-1α resulted in enhanced vascularized dental pulp regeneration in vivo[148]
Enhanced cell proliferationGain-of-function (overexpression)Lin28
(lentivirus)
HumanOverexpression of Lin28, a conserved RNA-binding protein in eukaryotes, caused increased proliferation through interaction with let-7a/IGF2BP2 pathways [149]
Enhanced cell proliferationmiR-based
knockdown
miR-210-3p
(lentivirus)
HumanOverexpression of miR-633, targeting the 3′ UTR of the MEPE gene, caused increased cell proliferation and differentiation [150]
Enhanced cell proliferationGain-of-function (overexpression)TGF-β1
(plasmid; electroporation)
HumanOverexpression of TGF-β1 resulted in increased osteogenic and chondrogenic differentiation but decreased adipogenic differentiation; it also resulted in increased proliferation rate and decreased apoptosis[111]
Enhanced cell proliferationsiRNA-based
knockdown
Visfatin (VIS)
(unknown)
HumanKnockdown of VIS, a novel adipokine associated with cellular senescence, resulted in reduced senescence[151]
Enhanced cell proliferationGain-of-function (overexpression)Hepatocyte growth factor (HGF)
(adenovirus)
HumanOverexpression of HGF inhibited rheumatoid arthritis progression by its immunosuppressive effects, while in the late phase, HGF promoted synovitis by activating fibroblast-like synoviocytes and exhibited accelerated cell proliferation and apoptosis resistance, suggesting a dual role of HGF in RA[152]
Enhanced cell proliferationGain-of-function (overexpression)Long noncoding RNA H19 (LncRNA H19)
(plasmid; lentivirus)
HumanOverexpression of LncRNA H19 caused increased cell proliferation, enhanced ALP activity, and increased odontoblast markers via regulating the TGF-β1/Smad signaling pathway[135]
Enhanced cell proliferationGain-of-function (overexpression)LncRNA H19
(plasmid; lentivirus)
HumanOverexpression of LncRNA H19 promoted cell proliferation, enhanced ALP activity, and increased odontoblast markers via regulating the TGF-β1/Smad signaling pathway[153]
Enhanced cell proliferationGain-of-function (overexpression)Adrenomedullin (ADM)
(lentivirus)
HumanOverexpression of ADM resulted in promotion of cell cycle progression, inhibition of p53 expression, decreased reactive oxygen species (ROS) accumulation, and resistance against cellular senescence [154]
Angio-/vasculogenic commitmentmiR-based knockdownMiR-424
(lentivirus)
HumanOverexpression of miR-424 caused decreased levels of VEGF and kinase insert domain-containing receptor (KDR) protein[155]
Angio-/vasculogenic commitmentGain-of-function (overexpression)PDGF-BB
(lentivirus)
HumanPDGF-BB caused enhanced proliferation, odontoblastic differentiation, and cell migration via the activation of the PI3K/Akt signaling pathway[99]
Angio-/vasculogenic commitmentGain-of-function (overexpression)VEGF
SDF-1α
(lentivirus)
HumanOverexpression of SDF-1α or VEGF resulted in enhanced cell proliferation, endothelial cell migration, and vascular tube formation on Matrigel in vitro; expression of both VEGF and SDF-1α resulted in enhanced vascularized dental pulp regeneration in vivo[148]
Angio-/vasculogenic commitmentGain-of-function (overexpression)BCL-2/ green fluorescent protein (GFP)
(lentivirus)
HumanOverexpression of BCL-2 resulted in enhanced endothelial cell proliferation, migration, and vascular tube formation on Matrigel, suggesting that BCL-2 overexpression enhances angio-/vasculogenic properties of DPSCs[156]
Angio-/vasculogenic commitmentGain-of-function (overexpression)Ets variant transcription factor 2 (ETV2)
(lentivirus)
HumanOverexpression of ETV2 resulted in the appearance of endothelial-like morphology and increased expression of endothelial-specific genes, which also correlated with enhanced capillary-like tube formation on Matrigel in vitro[157]
Angio-/vasculogenic commitmentGain-of-function (overexpression)PDGF-BB
(lentivirus)
HumanCo-cultivation of PDGF-BB-overexpressing cells with human umbilical vein endothelial cells (HUVECs) exhibited increased formation of vascular tubes, suggesting that PDGF-BB engineering is an effective strategy to amplify DPSCs’ angiogenic potential[158]
Neurogenic commitmentGain-of-function (overexpression)zinc finger protein 521 (Zfp521)
(unknown)
unknownOverexpression of Zfp521, a transcription factor, can facilitate differentiation into neural cells through chromatin modification [159]
Neurogenic commitmentGain-of-function (overexpression)Insulin-like growth factor-binding protein 5 (IGFBP5)
(retrovirus)
HumanOverexpression of IGFBP5 prompted neurogenic differentiation potential of DPSCs, suggesting that IGFBP5 is a potential target for dental pulp–dentin functional regeneration[160]
Neurogenic commitmentGain-of-function (overexpression)OCT3/4
(lentivirus)
HumanOverexpression of OCT3/4 under neural inductive conditions caused reprogramming into the neural lineage[161]
Adipogenic commitmentGain-of-function (overexpression)Ten-eleven-translocation 2 (TET2)
(plasmid; lipofectamine)
HumanOverexpression of TET2, a key regulator of DNA methylation during adipogenic induction, resulted in increased expression of adipogenic marker genes and enhancement of the transition of DPSCs toward adipogenic commitment[162]
Inflammation commitmentsiRNA-mediated knockdownReceptor for advanced glycation end products (RAGE) (lipofectamine)HumanBinding of RAGE to high-mobility group box 1 (HMGB1), a nonhistone DNA-binding protein that promotes inflammation, is directly related to eliciting inflammation[163]
Inflammation commitmentmiR-based knockdownmiR-223-3p
(lentivirus)
HumanOverexpression of miR-223-3p, one of the inflammation-induced miRNAs, resulted in increased production of odontoblast marker genes; knockdown of Smad3 increased the level of ALP, thereby promoting odontoblast differentiation[133]
Inflammation commitmentGain-of-function (overexpression)HGF
(adenovirus)
HumanOverexpression of HGF resulted in enhanced downregulation of inflammation-related factors[164]
Inflammation commitmentGain-of-function (overexpression)WNT4
(plasmid; lipofectamine)
HumanOverexpression of WNT4 resulted in amelioration of cell inflammatory response, enhanced BCL-2 expression, and decreased apoptosis rate in the DPCs inflamed by lipopolysaccharide (LPS) [165]
Cell migrationsiRNA-based
knockdown
Peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1)
(plasmid; lipofectamine)
HumansiRNA-based silencing of PIN1 (which specifically binds to phosphorylated Ser/Thr-pro motifs to catalytically regulate the post-phosphorylation conformation of its substrates) decreased the cell migration of DPSC[166]
Cell migrationGain-of-function (overexpression)PDGF-BB
(lentivirus)
HumanPDGF-BB enhanced odontoblastic differentiation and cell migration [99]
Enhanced induction into insulin-producing cells (IPCs)Gain-of-function (overexpression)Forkhead box A2 (FOXA2)
Pancreatic and duodenal homeobox 1 (PDX1)
(lentivirus)
HumanDPSCs can be reprogrammed to generate insulin-producing cells (IPCs) by transducing them with the transcription factor genes FOXA2 and PDX1[167]
Enhanced induction into IPCsmiR-based
knockdown using miR inhibitor
miR-183 inhibitor
(FuGENE)
RatDownregulation of miR-183 through transfection with miR-183 inhibitor resulted in the generation of cells expressing insulin 72 h after transfection [168]
Enhanced induction into IPCsGain-of-function (overexpression)Pdx1
Neurogenin 3 (Neurog3)
(plasmid; FuGENE)
RatTransfection with vectors carrying transcription factors Pdx1 and Neurog3 caused direct conversion into IPCs[169]
Enhanced induction into oligodendrocyte progenitorsGain-of-function (overexpression)Oligodendrocyte transcription factor 2 (Olig2)
(plasmid; X-tremeGENE)
HumanTransfection of DPSCs with the Oligo2 gene and subsequent cultivation in the differentiation-inducing medium resulted in the generation of cells showing oligodendrogenic markers[170]
Cell apoptosisRNAi (siRNA oligo)-
mediated knockdown
Caspase-8 and caspase-9
(unknown)
HumanKnockdown of caspase-9 exhibited a reduction in apoptosis, caspase-3 expression, and its activity[171]
Cell apoptosismiR-based
knockdown
miR-224-5p
(lipofectamine)
HumanInhibition of miR-224-5p, which targets the 3′-untranslated region (3′-UTR) of the Rac family small GTPase 1 (Rac1) gene, caused increased cell apoptosis [172]
Skeletal myogenic differentiationmiR-based
knockdown
miR-143 and miR-135 inhibitors
(lipofectamine)
HumanmiR-135 and miR-143 inhibitors induce myogenic differentiation of DPSCs[173]
Skeletal myogenic differentiationmiR-based
knockdown
miRNA-139-5p
(lipofectamine)
HumanOverexpression of miR-139-5p induced skeletal myogenic differentiation via the Wnt/β-catenin signaling pathway; downregulation of miR-139-5p inhibited cell growth and reduced skeletal myogenic differentiation [174]
Pluripotency and multilineage differentiation capabilityGain-of-function (overexpression)Octamer-binding transcription factor 4A (OCT4A)
(lentivirus)
HumanOverexpression of OCT4A resulted in upregulation of expression of stemness factors, as well as increased cell proliferation, pluripotency, and multilineage differentiation potential [175]
Pluripotency and multilineage differentiation capabilityGain-of-function (overexpression)Xeroderma pigmentosum complementation group C protein (XPC)
(lentivirus)
HumanXPC, a component of the DNA repair pathway, interacts with OCT3/4 to modulate pluripotency; overexpression of XPC enhances proliferation rate, reduces apoptosis, and improves DPSC’s multilineage differentiation capabilities[176]
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

Inada, E.; Saitoh, I.; Terajima, M.; Kiyokawa, Y.; Kubota, N.; Yamaza, H.; Morohoshi, K.; Nakamura, S.; Sato, M. Engineered Human Dental Pulp Stem Cells with Promising Potential for Regenerative Medicine. BioTech 2025, 14, 88. https://doi.org/10.3390/biotech14040088

AMA Style

Inada E, Saitoh I, Terajima M, Kiyokawa Y, Kubota N, Yamaza H, Morohoshi K, Nakamura S, Sato M. Engineered Human Dental Pulp Stem Cells with Promising Potential for Regenerative Medicine. BioTech. 2025; 14(4):88. https://doi.org/10.3390/biotech14040088

Chicago/Turabian Style

Inada, Emi, Issei Saitoh, Masahiko Terajima, Yuki Kiyokawa, Naoko Kubota, Haruyoshi Yamaza, Kazunori Morohoshi, Shingo Nakamura, and Masahiro Sato. 2025. "Engineered Human Dental Pulp Stem Cells with Promising Potential for Regenerative Medicine" BioTech 14, no. 4: 88. https://doi.org/10.3390/biotech14040088

APA Style

Inada, E., Saitoh, I., Terajima, M., Kiyokawa, Y., Kubota, N., Yamaza, H., Morohoshi, K., Nakamura, S., & Sato, M. (2025). Engineered Human Dental Pulp Stem Cells with Promising Potential for Regenerative Medicine. BioTech, 14(4), 88. https://doi.org/10.3390/biotech14040088

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