Next Article in Journal
Sex Lethal Gene Manipulates Gonadal Development of Medaka, Oryzias latipes, through Estrogenic Interventions
Next Article in Special Issue
Silver Nanoparticles Alone or in Combination with Calcium Hydroxide Modulate the Viability, Attachment, Migration, and Osteogenic Differentiation of Human Mesenchymal Stem Cells
Previous Article in Journal
The Essential Role of Prolines and Their Conformation in Allosteric Regulation of Kaiso Zinc Finger DNA-Binding Activity by the Adjacent C-Terminal Loop
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neural Regeneration in Regenerative Endodontic Treatment: An Overview and Current Trends

1
State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
2
State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
3
State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(24), 15492; https://doi.org/10.3390/ijms232415492
Submission received: 28 October 2022 / Revised: 24 November 2022 / Accepted: 1 December 2022 / Published: 7 December 2022
(This article belongs to the Special Issue Bioactive Tissue Repair and Regeneration: Focus on Endodontics)

Abstract

:
Pulpal and periapical diseases are the most common dental diseases. The traditional treatment is root canal therapy, which achieves satisfactory therapeutic outcomes—especially for mature permanent teeth. Apexification, pulpotomy, and pulp revascularization are common techniques used for immature permanent teeth to accelerate the development of the root. However, there are obstacles to achieving functional pulp regeneration. Recently, two methods have been proposed based on tissue engineering: stem cell transplantation, and cell homing. One of the goals of functional pulp regeneration is to achieve innervation. Nerves play a vital role in dentin formation, nutrition, sensation, and defense in the pulp. Successful neural regeneration faces tough challenges in both animal studies and clinical trials. Investigation of the regeneration and repair of the nerves in the pulp has become a serious undertaking. In this review, we summarize the current understanding of the key stem cells, signaling molecules, and biomaterials that could promote neural regeneration as part of pulp regeneration. We also discuss the challenges in preclinical or clinical neural regeneration applications to guide deep research in the future.

1. Introduction

Dental pulp is the only soft tissue in the tooth. It is located in the dental pulp cavity and is surrounded by dentin and connected to the periapical tissue by the narrow apical foramen [1,2]. It has several functions, including tooth formation, nutrition, sensation, and defense in physiological circumstances. When it is subjected to abnormal external stimuli such as caries or abrasion, it produces a defensive response by forming tertiary dentin to protect the pulp. Dental pulp maintains the vitality of dentin by providing oxygen and nutrients to odontoblasts and cell processes, performing sensory function through rich nerve distribution, and activating immune or inflammatory cells to participate in defense by regulating various molecular mediators [3]. The structure of dental pulp makes it difficult to restore its normal physiological state after bacterial infection or trauma. Root canal therapy (RCT) is the most effective and common treatment for pulpal and periapical diseases. By preparing, disinfecting, and filling root canals, we can control infection and accelerate the healing of periapical lesions. Nevertheless, due to the loss of vital pulp after RCT, the normal function of the pulp cannot be restored, resulting in increased fragility of the tooth, loss of defense ability, and even extraction [4].
The term “Regenerative endodontic therapy (RET)” was first proposed by Murray et al. [5] in 2007 based on tissue engineering. RET was conceived to replace inflamed/necrotic pulp tissue by regenerating pulp-like tissues [6]. At present, the most commonly used RET in clinical practice is revascularization. Revascularization is a method of recruiting undifferentiated oral stem cells and molecules from the apical region using manual files to guide a bleeding clot (BC), or using platelet-rich plasma (PRP) or platelet-rich fibrin (PRF) after adequate disinfection of the root canal in order to promote the development of immature permanent teeth. Although various studies have proven the efficacy of revascularization, many scholars suggest that revascularization may not achieve real pulp regeneration [7]. Therefore, two new strategies based on tissue engineering have been proposed: stem cell transplantation, and cell homing. Stem cell transplantation entails transplanting stem cells combined with growth factors (GFs) and scaffolds into the root canal. Further formation of a new pulp–dentin complex is accomplished by promoting stem cells’ differentiation towards odontoblasts [8]. Dental stem cells—such as dental pulp stem cells (DPSCs), stem cells from apical papilla (SCAPs), human exfoliated deciduous teeth (SHEDs), and periodontal ligament stem cells (PDLSCs)—are widely applied for stem cell transplantation owing to their excellent capability for multiple differentiation [9]. Cell homing aims to implant GFs with scaffolds into the root canal to build an environment conducive for tissue regeneration. Then, autologous stem cells are recruited from the periapical region and into the root canal to realize RET [10]. The most widely used materials for scaffolds, whether in cell transplantation or cell homing, are natural materials such as collagen [11].
The ideal pulp regeneration process should not only form the same structure as the natural dental pulp but also restore its function. Functional dental pulp should meet the following requirements [12]: (1) deposition of new dentin at an adjustable rate; (2) cell density and structure similar to those of natural dental pulp; (3) vascularization; and (4) innervation. Normal pulp is rich in nerve fibers. The number of nerve fibers in the regenerated pulp determines the degree of restoration of sensory function. Therefore, nerves are indispensable in regenerating dental pulp, and particularly important for maintaining its structure and function. This review summarizes the recent studies of successful neural regeneration in RET in vivo, analyzes the conditions and requirements for achieving neural regeneration, and discusses stem cells, GFs, and scaffolds that can promote neural regeneration. We believe that this review can provide a reference for clinical translation in guiding neural regeneration in RET in the future.

2. Function of Nerve in Dental Pulp

The nerves of the pulp are mainly derived from the maxillary and mandibular branches of the trigeminus. The sensory nerve fibers enter the pulp cavity via blood vessels through the apical foramen. The nerve fibers of the pulp are mixed fibers, since they also contain sympathetic fibers from cervical sympathetic nodes [13]. The sensory nerve fibers of pulp include myelinated Aδ fibers and unmyelinated C fibers. These are both responsible for transmitting pain impulses [14]. Because there are only nociceptors in the pulp, pain results when the pulp is stimulated by mechanical or temperature changes. The terminals of Aδ fibers are distributed in the pulp–dentin junction area, have a low stimulation threshold, and produce pain characterized by acute tingling. The terminals of the C fibers are spread throughout the pulp and have a high stimulation threshold; thus, the pain is characterized by a severe burning-like sensation [15]. The stimulation of C fibers is closely related to the pain caused by inflammation. When the pulp is inflamed, local tissue pressure increases, which excites the C fibers and transmits the sense of pain. On the other hand, inflammatory mediators such as histamine and bradykinin can excite C fibers, which also release neuropeptides such as substance P (SP) and calcitonin-gene-related peptides (CGRPs). This results in vasodilation and increased peripheral nerve sensitivity [16]. C fibers are resistant to hypoxic environments, and patients may still feel pain when the pulp is hypoxic and necrosis [17].
Sensory neurons also participate in the regulation of inflammation, immunity, and angiogenesis, mainly through the secretion of nerve growth factor (NGF) and neuropeptides (Figure 1). In dental caries, fibroblasts located directly below the caries secrete NGF to promote axon growth to the damaged area [18]. The newborn nerve terminals form processes in the pulp angle area in response to the damage of dental caries. In pulpitis, NGF is continuously upregulated to increase the density of nerve fibers [14]. Moreover, C fibers and non-nerve cells secrete neuropeptides to participate in the transmission of pain, and the level of neuropeptides significantly increases with inflammation. Furthermore, various resident cells in the pulp—such as fibroblasts, odontoblasts, inflammatory cells, and endothelial cells of the postcapillary venules—express membrane-binding receptors of sensory and sympathetic peptides [19]. During inflammation, the expression of these receptors increases significantly, indicating that neuropeptides are crucial in mediating inflammation [19]. The nerve in the pulp also participates in vascular regulation. The sympathetic autonomic nerve of the superior cervical ganglion affects the contraction of blood vessels in the pulp [15]. Moreover, the release of CGRP and SP promotes vasodilation and plasma extravasation [20]. Regarding immune system regulation, a study demonstrated that the recruitment of immunoreactive cells in the inflammatory pulp of denervated molars was significantly lower than that of innervated molars, implying that nerves are involved in the immune system processes of pulp [21]. Ho et al. [22] found that many receptors of neuronal signal molecules existed in immune cells, such as monocytes/macrophages, indicating that nerves were directly involved in the recruitment of immune cells.

3. Development of Neural Regeneration in RET

Pulp is a well-vascularized neurological tissue. After years of research and discussion, most in vivo RET experiments and clinical trials have successfully achieved the regeneration of pulp-like tissue with vessels. However, nerve formation is still problematic, and few experiments have reported nerve formation in RET. To truly achieve functional pulp regeneration, restoring innervation is indispensable.
Revascularization is the method most commonly used to promote the continuous development of roots and the closure of the apical foramen of immature permanent teeth. However, histological analysis shows that the regenerated tissue is not pulp-like, but more periodontal-like, similar to cementum. Most of the roots develop after revascularization and are positive to sensitivity tests, which are helpful to guide neurogenesis in functional pulp regeneration. A study indicated that vitality tests required periapical neuronal terminals to be located in the coronal potion of the repaired tissue, about 10-15 mm from the nearest nerve trunk [23]. Another study analyzed the tooth #45 of a 10-year-old child who underwent revascularization. The results showed that ectopic cementum and periodontal-ligament-like tissue were located in the root canal, while nerve fibers were found by immunohistochemistry [24]. Furthermore, Austah et al. [25] reported that they successfully promoted root development of necrotic pulp by revascularization and that the teeth were positive to sensitivity tests. Immunohistochemistry further indicated that there were CGRP-positive nerve fibers. These experiments further confirmed that the reconstruction of sensory fibers was successfully induced by revascularization.
During human tooth development, the innervation of the teeth is modulated by a series of molecular signals that are mainly regulated by the release of neurotrophic factors (NFs)—especially NGF and brain-derived neurotrophic factor (BDNF) [26]. The primary afferent branches accumulate around the apical papilla and then enter the dental pulp until the late cap stage to form the peripheral primary afferent branches of the pulp–dentin complex [23,27]. Therefore, it is necessary to provide the appropriate concentration of NFs for the growth of nerve fibers in RET. In addition to the exogenous provision of NFs, some mesenchymal stem cells (MSCs)—such as DPSCs, SCAPs, and SHEDs—can secrete NFs. Moreover, various studies have confirmed that dental MSCs not only have nerve differentiation potential but also secrete other molecules such as chemokines and vascular GFs, which can regulate axon growth and accelerate the recovery of nerves [28]. The reinnervation of dental pulp needs to be guided by axons to recruit free nerve terminals in the apical area. Therefore, finding suitable stem cells, NFs, and related scaffolds is particularly important.
Revascularization requires comprehensive disinfection of the root canal. The root canal can be continuously irrigated with sodium hypochlorite during the first treatment, and then calcium hydroxide or antibiotic paste can be injected into the root canal for disinfection. If there are no signs of infection during the revisit, dry the canal after removing the disinfectant and use a sterile K-file beyond the apical foramen to induce apical bleeding and fill the root canal with blood. PRP or PRF can be used instead of this step. After the blood clot in the root canal has formed, cover it with a layer of mineral trioxide aggregate (MTA)/Biodentine or other materials. The defect can then be filled with glass ionomer cement. Patients need follow-up for a long time after treatment. Stem cell transplantation and cell homing also require root canal preparation and adequate disinfection. Hydrogel scaffold materials such as collagen, which is the most widely used, can be loaded with stem cells such as DPSCs, SHEDs, bone marrow mesenchymal stem cells (BMMSCs), or adipose-derived stem cells (ADSCs), and can be injected into the canals with or without GFs, such as platelet-derived growth factor (PDGF)-BB, NGF, BDNF, granulocyte colony-stimulating factor (G-CSF), stromal-cell-derived factor-1 (SDF-1), basic fibroblast growth factor (bFGF), or bone morphogenetic protein 7 (BMP7). Regular follow-up is also necessary.
We summarized the recent successful reports of neural regeneration in animal experiments and clinical trials of pulp regeneration, as shown in Table 1 and Table 2. Our certification of successful neuroregenerative experiments included observing nerves through histological analysis or confirming classical nerve markers by immunofluorescence. Furthermore, we considered positivity for sensitivity tests, including cold tests, hot tests, and electric pulp tests (EPTs).

4. Role of Stem Cells in Neural Regeneration

As key components of tissue engineering, stem cells are essential in pulp regeneration. Stem cells commonly used for RET can be classified as dental and non-dental cells. Dental stem cells include DPSCs, SHEDs, PDLSCs, SCAPs, and dental follicle stem cells (DFSCs) [89,90,91]. Non-dental stem cells mainly include BMMSCs, ADSCs, embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs) [92,93]. Dental stem cells are the favored seed cells for pulp regeneration because they are non-invasive, have excellent proliferative activity, and possess neural and vascular differentiation ability. Compared with ESCs, dental stem cells pose fewer ethical problems. Non-dental stem cells also exhibit potential in pulp regeneration. Stem cells with neural differentiation characteristics are discussed in this session.

4.1. DPSCs

DPSCs are obtained from mature pulp tissue and have high colony-formation and proliferation rates. They are regarded as the “gold standard” for stem cells in dental tissue engineering. Gronthos et al. [94] first identified and isolated human DPSCs from human third molars. They can be obtained mainly from the third molar or orthodontic teeth that need to be extracted. DPSCs are MSCs derived from the ectoderm and originate from the cranial crest during embryonic development. They express not only MSC markers such as CD10, CD13, CD29, CD44, CD73, CD90, CD105, CD106, CD117, CD146, and STRO-1, but also neural crest markers such as p75, Snail-1 and -2, and Sry-type high-mobility-group box 9 (SOX-9) [95]. In addition, some multipotent stem cell markers—such as octamer-binding transcription factor 4 (Oct4), Nanog, SOX-2, stage-specific embryonic antigens (SSEAs), and c-Myc—are also expressed in these cells [96]. In vitro, the differentiation of DPSCs is regulated by the microenvironment. DPSCs have the potential to differentiate into multiple cell types—for example, osteoblasts, muscle cells, adipose cells, melanoma cells, chondrocytes, and corneal epithelial cells [91,97]. A lineage-tracing experiment found that peripheral-nerve-associated glial cells differentiate into dental MSCs during the development, self-renewal, and repair of teeth [98]. This confirmed the neurological origin of dental MSCs. DPSCs can differentiate into glial cells and neurons and can secrete NFs, including BDNF, GDNF, NGF, and neurotrophin-3 (NT-3) [92]. Gronthos et al. [99] explored the neural markers of DPSCs for the first time. The results were positive for neuroepithelial stem cell protein (Nestin) and glial fibrillary acidic protein (GFAP). Moreover, dental pulp cell sheets produced neurotrophins, which promoted neural regeneration and enhanced the function of an injured facial nerve in rats [100]. In addition, DPSCs protected and accelerated the regeneration of retinal ganglion cells in vivo and in vitro [101,102]. Furthermore, Kolar et al. [103] reported that axon growth inhibitors existed in completely transected spinal cord injury (SCI), but the nutritional factors secreted by human DPSCs promoted axon regeneration. The mechanism that DPSCs use to accelerate the repair and regeneration of multiple neurological diseases may be viewed from the following four perspectives: differentiated into neurological cells, paracrine, anti-inflammatory, and immunomodulatory [104].
An experiment recorded the positive staining for nerves in pulp-like tissue regenerated by DPSCs [105]. In addition, Iohara et al. [39] regenerated pulp-like tissue with dentin and dense nerve plexus in the pulp cavity of a beagle dog’s permanent teeth with collagen loaded with DPSCs and GCS-F, confirming the role of DPSCs in neural regeneration. Overall, these findings suggest that DPSC transplantation promotes axon extension and neuronal growth in pulp regeneration [8]. A CD31-/CD146- population in DPSCs found with high expression of neurotrophic and angiotrophic factors is expected to become the best candidate of stem cells for functional pulp regeneration [106].

4.2. SHEDs

SHEDs are extracted from the pulp of human deciduous primary teeth. They are similar to DPSCs and are considered to be a precious stem cell source in RET [107]. Miura et al. [108] discovered SHEDs in 2003. They showed better proliferation and self-renewal ability than DPSCs and BMMSCs. Similarly, markers of MSCs such as CD13, CD29, CD44, CD73, CD90, CD105, CD106, CD146, CD166, and STRO-1 are detected on the surface of SHEDs. However, hematopoietic stem cell markers such as CD14, CD18, CD19, CD24, CD34, and CD45 are negative [109]. SHEDs also have multiple differentiation potentials, such as osteogenesis, dentinogenesis, adipogenesis, and neurogenesis. Additionally, the osteogenic and adipogenic differentiation ability of SHEDs is more potent than that of DPSCs [110]. Because of the neural origin of pulp cells, SHEDs also express neuronal markers such as Nestin, NeuN, GFAP, Doublecortin, β III-tubulin, and glutamic acid decarboxylase (GAD) [107]. Studies have demonstrated that SHEDs form neurospheres in the culture medium and then differentiate into dopaminergic neuron clusters [111]. Additionally, SHEDs can also differentiate into Schwann cells (SCs) and accelerate axon regeneration [112]. It has been observed that SHEDs cultured in vitro produce NFs, such as NGF, BDNF, and GDNF [112]. An in vitro study reported that SHEDs expressed more neuronal markers than DPSCs and had a superior ability to differentiate into neurocytes [113].
In pulp regeneration, SHEDs and hydroxyapatite/tricalcium phosphate (HA/TCP) were combined for the first time in 2003 and demonstrated that dentin was formed in the scaffold [114]. Rosa et al. [115] implanted SHEDs with biological scaffolds (i.e., PuramatrixTM or rhCollagen type I) into full-length root canals, which were transplanted subcutaneously into the dorsal surface of immunocompromised mice. They found that SHEDs differentiated into odontoblasts with secretory functions and formed functional dental pulp and tubular dentin. In a randomized clinical trial, autologous SHED transplantation regenerated vascularized pulp-like tissue with sensory nerves in traumatic teeth in children [34]. As adult stem cells, SHEDs have unique advantages. They are derived from naturally replaced deciduous pulp; thus, they can be obtained non-invasively and involve less ethical controversy. Moreover, they can effectively avoid immune rejection when used for autografting [92,110].

4.3. PDLSCs

PDLSCs are found in the periodontal ligament (PDL) between the teeth and the alveolar fossa, where they maintain the homeostasis of the periodontal tissue. PDLSCs were first isolated from periodontal ligaments by Seo et al. in 2004 [116]. They are heterogeneous, clonal, and highly proliferative multipotent cells that produce collagen and PDL-like tissue to promote the regeneration of periodontal tissue under periodontal diseases [117]. PDLSCs can differentiate into osteoblasts, chondrocytes, adipocytes, cardiomyocytes, endothelial cells, islet cells, and corneal keratinocytes [118]. They express typical MSC markers such as CD10, CD13, CD26, CD29, CD44, CD59, CD73, CD90, CD105, CD106, CD140b, CD146, and CD166 [109]. In addition, PDLSCs can express neuron markers, glial cell markers, and early markers of neural stem cells (e.g., human natural killer 1, p75, Nestin), indicating their great potential in the repair of nervous system injury [118,119]. A study found that PDLSCs transplanted into the brains of adult mice survive, migrate, and differentiate into neuron-like cells [120]. Moreover, Fortino et al. [121] used epidermal growth factor (EGF) combined with bFGF to induce PDLSCs to differentiate into neuron cells.
PDLSCs are prone to forming directional fibers and can differentiate into glial cells and osteoblasts, but their neural differentiation ability is poorer than that of other dental stem cells [122]. However, PDLSCs mainly form PDL-like or cementum tissue in vivo, and related research on their use in RET is still in its infancy. Thus, their potential needs to be investigated further.

4.4. SCAPs

SCAPs come from the immature root apical region of permanent teeth and share the same origin as DPSCs [123,124]. Although apical papilla is a precursor tissue of root pulp, there are still significant differences in the biological characteristics of DPSCs and SCAPs [125]. Some scholars believe that SCAPs differentiate into primary odontoblasts when root dentin is formed during tooth development, while DPSCs may be the source for replacement odontoblasts to form reparative dentin [122]. Compared with DPSCs, SCAPs exhibit higher proliferation and tooth tissue regeneration ability. This may be related to the high expression of survivin and telomerase in SCAPs [126,127]. SCAPs express many odontogenic markers, but the expression levels of dentin sialophosphoprotein (DSPP), matrix extracellular phosphoglycoprotein (MEPE), transforming growth factor β RII, FGFR3, VEGFR-1, FGFR1, and CD146 in SCAPs are lower than those in DPSCs [128]. In addition, SCAPs not only express classical MSC markers but also have a SCAP-specific marker—CD24 [129]. Moreover, they are positive for neuron markers such as β III tubulin, glutamic acid decarboxylase, NeuN, Nestin, neurofilament protein M (NF-M), and neuron-specific enolase (NSE) [126]. SCAPs can differentiate into odontoblasts, osteoblasts, adipocytes, and neural cells in vitro [130]. In addition, the differentiation of SCAPs into functional odontoblasts has been verified in vivo. Research has demonstrated that SCAPs can regenerate a dentin-pulp-like complex in immunodeficient mice with appropriate scaffolds [127]. Moreover, it has been confirmed that the secretion of BDNF from SCAPs is crucial for neuronal growth in vitro [131]. Semi-quantitative RT-PCR confirmed that the gene expression levels of BDNF, GDNF, and ANGPT1 in SCAPs were much higher than those in other dental MSCs [103]. This indicates the potential of SCAPs for neural regeneration.
Sequeira et al. [132] investigated the pulp regeneration potential of SCAPs. SCAPs were embedded in PRP scaffolds and then implanted into the dorsal region of immunodeficient rats. They successfully achieved formation of pulp-like and dentin-like tissue. Since their anatomical location is near the apical foramen, SCAPs are also the most important candidate stem cells for revascularization [133]. However, in vivo analyses using SCAPs are still scarce, and the neural differentiation potential of SCAPs in RET needs further investigation.

4.5. BMMSCs

BMMSCs were the first MSCs to be discovered, and bone marrow is a widely used source of MSCs. The relevant characteristics of BMMSCs have been widely studied and applied [134]. The positive markers for BMMSCs are Stro-1, CD271, CD146, CD106, CD73, CD105, FZD9, SUSD2, LEPR, and CD90, and the negative markers are CD44, CD31, CD34, and CD45 [135]. BMMSCs are a subgroup of stromal cells. They can be isolated from compact bone, cancellous bone, and the femoral head [136]. BMMSCs can differentiate into osteoblasts, chondroblasts, adipocytes, myoblasts, and neurons [137,138]. In terms of whole-tooth regeneration, BMMSCs upregulate the expression of tooth-derived genes and recombine with embryonic oral epithelium [139]. Furthermore, C-Kit (+) BMMSCs can differentiate into odontoblasts and periodontal tissue cells [140,141,142]. Specifically, BMMSCs can differentiate into neurons, astrocytes, and SCs [143]. Experiments have also exhibited the neural differentiation potential of BMMSCs in vivo. A study demonstrated that BMMSCs differentiated into neuron-like cells after being transplanted into rats [144]. Mathot et al. [145] used BMMSCs to repair 10 mm sciatic nerve defects in rats. It was found that both differentiated and undifferentiated BMMSCs are able to promote the repair of the injured nerve without significant differences. In addition, another study combined BMMSCs and iRoot BP to explore their effects on pulp-like tissue formation. The results showed that the newborn tissue was pulp-like tissue with positive neural markers [146].

4.6. ADSCs

ADSCs are MSCs with multi-differentiation potential. They were first isolated from adipose tissue by Zuk et al. [147] in 2001. They are widely utilized in regenerative medicine and are an excellent substitute for BMMSCs in bone repair and regeneration [148,149]. ADSCs express the surface markers of MSCs, such as CD10, CD13, CD29, CD44, CD49, CD73, CD90, CD105, and CD166, but not CD11b, CD14, CD31, or HLA-DR [150]. ADSCs can protect and promote the regeneration of the central nervous system by secreting BDNF, GDNF, NGF, and insulin-like growth factors (IGFs) [151]. An in vivo study investigated the effect of silicone catheters on the repair of 7 mm facial nerve defects in rats, which contained undifferentiated ADSCs, differentiated ADSCs, or SCs [152]. Morphological quantitative analysis of the regenerated facial nerves demonstrated that undifferentiated ADSCs, differentiated ADSCs, and SCs had similar neural regeneration potential [152].
Ishizaka et al. [40] found that CD31- ADSCs induced pulp regeneration. The qualitative and quantitative patterns of mRNA expression in newborn tissue were similar to those of natural pulp. Moreover, Murakami et al. [36] successfully achieved pulp regeneration with ADSCs and BMMSCs, accompanied by angiogenesis and nerve regeneration. These results confirm the role of ADSCs in RET and neural regeneration. The extraordinary biological characteristics of ADSCs have made them vital seed cells in the field of oral regeneration, but their immunogenicity and safety after transplantation are still a concern.

5. Role of GFs in Neural Regeneration

GFs are crucial in tissue engineering. Different GFs combined with scaffolds promote endogenous tissue regeneration—especially those related to vascular, nerve, and dentin formation. GFs such as PDGF and VEGF are related to angiogenesis. SDF-1 α, bFGF, and PDGF are used for chemotaxis, while BMP-7 is used to promote odontoblast differentiation and mineralization [153,154]. Some GFs promote neuronal growth and provide protection, including NGF, BDGF, NT-3, bFGF, IGF, G-CSF, GDNF, and EGF. The following section summarizes the GFs that promote nerve growth and are widely used in RET.

5.1. NGF

NGF was the first neurotrophin found by Rita Levi-Montalcini and Viktor Hamburger [155] in murine sarcoma culture. It is a vital regulator of the fate, development, survival, and growth of neurons and non-neurons [156,157]. It has been demonstrated that NGF has two receptors: TrkA and p75NTR. The affinity between NGF and the transmembrane glycoprotein receptor p75NTR is low, and the activation of p75NTR determines cell survival or apoptosis [158]. NGF has a high affinity for TrkA, but its function is affected by p75NTR [159]. It has been found that TrkA and p75NTR are involved in cell proliferation, differentiation, and survival in bone marrow and lymphoid tissue [160]. Moreover, NGF can promote the differentiation and migration of vascular smooth muscle cells and endothelial cells, mobilize and activate endothelial progenitor cells, and promote neovascularization [161]. It has been reported that NGF induces BMMSCs to differentiate into neural cells via a neuropeptide pathway, verifying the capability of NGF for neural regeneration [162]. Chung et al. [163] found that NGF induced neurogenesis of dopaminergic cells through an ERK-driven and transcription-dependent latent process and an ERK- and PI3K-driven and transcription-independent extension process. Furthermore, NGF, in combination with other NFs, successfully induced DPSCs to differentiate into neurogenic cells [164].
In dentistry, NGF induces SCAPs’ differentiation into odontoblasts. This indicates that NGF can be applied as a mineralization stimulant [165]. Mitsiadis and Pagella et al. [166] confirmed the effects of NGF in the proliferation and differentiation of epithelial and mesenchymal cells, along with accelerated sprouting of nerve fibers in dental tissue during tooth development. Moreover, the level of NGF would increase in pulpitis to accelerate the release of neuropeptides such as SP and CGRP, resulting in pain [167]. Li and Wang [35] delivered a combination of PDGF-BB, NGF, and BDNF into teeth undergoing endodontic treatment, which were then implanted subcutaneously into the dorsum of rats. They found well-vascularized pulp-like tissue formation and positive signals of S-100, indicating that nerve fibers regenerated. Combining chemokines and NGF with cell homing can enhance the regeneration of pulp-like tissue and nerve fibers, providing a good approach for actualizing functional pulp regeneration.

5.2. BDNF

BDNF is one of the most investigated and characterized NFs in the nervous system and is also a member of the neurotrophin family. It plays a crucial role in the survival and differentiation of neurons by activating TrkB for the purpose of regulating and maintaining the normal function of the brain. In addition, BDNF can be detected in other tissues and cells—for example, bone, cartilage, tooth germ, heart tissue, and osteoblasts [168]. It was found that BDNF accelerates osteogenesis of BMMSCs through the Erk/Runx2/Osterix pathway to directly regulate endothelial cells’ survival and vascular stability. It also indirectly regulates capillary network formation through VEGF [35,169]. BDNF produced during injury or inflammation may promote axonal growth and peripheral nerve regeneration [170].
In dentistry, Nosrat et al. [171] found that the mRNA of BDNF resides mainly in the dental papilla/pulp of postnatal rats using in situ hybridization. They determined that the expression pattern was related to dental innervation. Furthermore, the complement C5a activated by fibroblasts controlled the production of BDNF in pulpitis. Thus, it could promote neuron growth to the injured area [172]. Moreover, de Almeida et al. [131] investigated SCAP co-culture with trigeminal neurons. They found that SCAPs may drive axons through the BDNF signaling pathway to regenerate nerves. Moreover, some studies have found that BDNF and NT-4/5 accelerate the migration of DPSCs in vitro [173]. Luzuriaga et al. [174] demonstrated that BDNF and NT-3 combined with StemPro MSCTM induced the partial reprogramming of ectomesenchymal human DPSCs to generate early neural crest progenitor cells. The capability of BDNF in neuroangiogenesis and the migration of DPSCs suggest its great potential for RET.

5.3. NT-3

NT-3, the third member of the neurotrophin family, was discovered by Hohn et al. [175] in 1990. Unlike NGF and BDNF, NT-3 has a higher affinity for TrkC and a lower affinity for TrkA and TrkB [176]. NT-3 has cardiovascular effects, as NT-3- and TrkC-knockout mice had severe heart defects [177,178]. In addition, Cristofaro et al. [178] revealed an angiogenic ability of NT-3 that is associated with TrkC phosphorylation and the phosphatidylinositol 3-kinase–Akt kinase–endothelial nitric oxide synthase pathway. Moreover, it has been found that NT-3 promotes the differentiation of human BMMSCs into osteoblasts in an inflammatory environment [179]. NT-3 is essential in neuronal survival and differentiation. It can promote the reconstruction of neural connections at the lesion site in SCI [180]. Yan et al. [181] cultured BMMSCs with NT-3 and found that the BMMSCs differentiated into neurons. This ameliorated the cognitive function in rats with Alzheimer’s disease. They also proved that the Wnt/β-catechin signaling pathway was involved the process. Moreover, in an SCI rat model, transplanted BMMSCs that overexpressed NT-3 promoted the recovery of motor function and neural regeneration [182]. Additionally, BDNF and NT-3 had a synergistic regulatory effect on the neural differentiation of ADSCs [183]. Given its excellent ability to promote neural regeneration and angiogenesis, NT-3 is an ideal candidate for RET in the future, but more in vivo experiments are necessary.

5.4. bFGF

bFGF, or FGF-2, is the earliest FGF to be found in pituitary extracts by Armelin [184] in 1973. As a secretory signal protein, bFGF is expressed in almost all tissues and binds to the tyrosine kinase FGF receptor on the cell membrane. It participates in cell proliferation, differentiation, migration, and angiogenesis. It is related to the activation of the RAS-MAPK, PI3K-AKT, PLC γ, and STAT signaling pathways [185,186]. bFGF is a vital regulator of bone formation and differentiation. After bFGF knockout, the bone formation capability of osteoblasts was impaired, and the accumulation of bone marrow fat increased [187]. In addition, bFGF promotes wound healing, accelerates tissue regeneration, participates in neurogenesis, and acts as an angiogenic factor [188]. bFGF increases the size of the neurospheres and the expression of neurogenic markers in DPSCs through intracellular transduction of FGF-FGFR and PLC γ [189]. Lue et al. [190] reported functional recovery in a rat model of SCI after transplanting a hydrogel containing DPSCs and bFGF. Moreover, Liang et al. [191] showed that FGF2-induced human dental pulp cells released NFs to support axon regeneration.
In dentistry, Shimabukuro et al. [192] proved that FGF-2 promotes the proliferation and migration of DPSCs and accelerates the regeneration of the dentin–pulp complex. Local regeneration of the dentin–pulp complex in rats was successfully achieved by using FGF-2, gelatin hydrogel, and a collagen sponge [193]. Additionally, Kim et al. [194] found that FGF-2 enhances the proliferation and migration of stem cells from the inflamed pulp tissue of human functional deciduous teeth (iSHFD), and that ectopic transplantation of iSHFD/FGF-2 increases the formation of dentin-like tissue. Furthermore, Sagomonyants et al. [195] found that bFGF promotes stem cells’ differentiation to odontoblasts by activating the FGFR/MEK/ERK1/2 and BMP/BMPR pathways.
Kim et al. [10] used collagen gel to deliver a variety of GFs—including bFGF—into human teeth, which were then transplanted into rats. They found newborn pulp-like tissue with angiogenesis and neurogenesis. Given the role of bFGF in cell proliferation, angiogenesis differentiation, and neurodifferentiation, as well as its potential for pulp wound healing and regeneration, it can be used as a reserve cytokine for cell homing. Moreover, bFGF has been approved for clinical use by the Japanese Pharmaceuticals and Medical Devices Agency (PMDA) and the United States Food and Drug Administration (FDA). Therefore, bFGF is safe as a candidate for the clinical application of neural regeneration in RET.

5.5. IGFBP5

IGF and its six binding proteins (IGFBPs) are crucial for cell growth, proliferation, and differentiation—especially in osteoblasts and fibroblasts [196]. IGFBP5 is related to regulation of the biological function of MSCs, especially in cell growth, differentiation, migration, senescence, and apoptosis [197]. Hao et al. [198] found that IGFBP5 promotes the differentiation of DPSCs into odontoblasts and further enhances dentin formation through the JNK and ErK signaling pathways. Saito and Ohshima et al. [199] reported that IGFBP5 was essential in regulating the survival and apoptosis of DPSCs during tooth development and pulp wound healing. Furthermore, Li et al. [200] found that the overexpression of IGFBP5 upregulates the expression of angiogenic markers (e.g., VEGF, PDGF, angiopoietin-1) in DPSCs, as well as neurogenic markers (e.g., neural cell adhesion molecule, tyrosine hydroxylase, Nestin, β-III-tubulin). These studies confirm the angiogenesis and neurogenic differentiation potential of IGFBP5, but more research is needed to verify its role in neural regeneration.

5.6. G-CSF

G-CSF is a glycoprotein belonging to the growth factor cytokine family that is used to clinically treat neutropenia [201,202]. Some investigations have exhibited the neuroprotective ability of G-CSF. In an experimental murine model of cerebral ischemia, G-CSF combined with BMMSCs produced a synergistic effect to promote cell proliferation and differentiation, resulting in early neuronal development and the reduction in cerebral infarction size [203]. Furthermore, G-CSF induces neurogenesis, increases neural plasticity, and reduces apoptosis by activating receptors [204].
In dentistry, G-CSF and dental pulp cells loaded with collagen have been used for pulp regeneration. The results were successful regeneration of pulp-like tissue after 90 days [205,206]. By comparing G-CSF and bFGF in RET, it was found that the two GFs showed similar roles in migration, proliferation, anti-apoptosis, angiogenesis, and neuronal growth in vitro [207]. However, bFGF inhibits mineralization of the DPSCs, while G-CSF promotes mineralization [207]. Meanwhile, G-CSF stimulates the migration and proliferation of endogenous stem cells, similar to SDF-1 [208]. The PMDA and FDA have approved G-CSF for clinical use, and the development of G-CSF in the field of pulp nerve regeneration can be explored further.

6. The Role of Scaffolds in Promoting Neural Regeneration

Scaffolds used in RET should not only transport stem cells into the root canal but also regulate the proliferation, differentiation, and metabolism of those stem cells [209]. The properties of biocompatibility, porosity, mechanical strength, and degradation rate should be considered when selecting a scaffold [210]. In addition, the electrical conductivity of the material and its interaction with NFs are also essential when considering the potential for neural regeneration [211].
According to the reports from in vivo and clinical trials showing successful induction of neural regeneration in RET, most used natural biomaterial scaffolds. The most common scaffold materials are autologous platelet concentrates (APCs) and collagen derivatives such as atelocollagen or collagen TE. One study created a unique sandwich structure, composed of hDPSC sheets, TDM/hDPSCs, and Matrigel/hDPSCs. TDM and Matrigel are natural biomaterials [32]. Another clinical study compared the effects of BC, PRF, collagen, and hydroxyapatite on pulp regeneration in vivo. Twelve months later, 11%, 66%, 44.4%, and 33.3% of patients in the BC, PRF, collagen, and hydroxyapatite groups, respectively, were positive to cold tests [29].
The scaffold materials used for RET and neural regeneration can be divided into natural materials, synthetic materials, and composite materials. Since most studies used natural biomaterial scaffolds, the next part of this review focuses on them.

6.1. Natural Materials

6.1.1. APC

BC, PRP, and PRF are APCs that are often used for revascularization. BCs are formed by using hand files to mechanically stimulate bleeding at the root tip [212]. PRP and PRF are obtained from peripheral blood by anticoagulation and centrifugation [213]. PRP and PRF are rich in platelets. When platelets are activated, high concentrations of GFs can be produced through degranulation to regulate cells’ chemotaxis, migration, proliferation, and differentiation, thereby promoting tissue repair [214]. A clinical study evaluated radiological results for the root length of 88 immature incisors treated with BC, PRP, PRF, and platelet granules [52]. The study found that the results of all treatment groups were similar and, after an average of 28.25 months, root healing, development, and sensitivity tests showed good results. Various research results shown in Table 2 prove the positive effects of APCs on neural regeneration, since the teeth were positive to sensitivity tests. Although APCs provide necessary GFs for tissue regeneration, further study is needed because of the unpredictability of clot formation, the challenge of obtaining PRP, and the limited curative effect [215].

6.1.2. Collagen

Collagen is not only the structural protein of human and animal connective tissue, but also the main component of extracellular matrix. The main collagen used for scaffolds is type I collagen. This is the most valuable biomaterial for tissue engineering, drug delivery, and cosmetic surgery [216,217]. Collagen has high biocompatibility and biological activity, and it can promote cell adhesion, migration, and proliferation [218]. Collagen has poor mechanical properties but is adequate for RET, and its mechanical properties can be enhanced through crosslinking with other elements or constructing composite scaffolds [219]. Collagen can carry pulp stem/progenitor cells with or without GFs and anti-inflammatory molecules to form pulp-like tissue in vivo [31,33,35,36,37,38,39,40,41]. After transplanting collagen with CD31- or CD105+ pulp cells containing SDF-1 into dogs’ root canals, pulp-like tissue was observed, along with blood vessels and nerve domination [40,41,220]. Collagen hydrogel crosslinked with cinnamaldehyde promoted the proliferation and odontogenic differentiation of human DPSCs [221]. In nerve tissue engineering, collagen can accelerate neuronal growth and neural regeneration, as well as helping to maintain the biological function of neurons [222]. However, the low mechanical strength, high water absorption, and rapid degradation of collagen are not beneficial for neural regeneration [211]. Therefore, collagen often needs further modification for this application. However, these modifications may affect the function of stem cells in pulp regeneration. How to balance the characteristics of collagen with the degree of modification in neural regeneration during RET may be worthy of further investigation.

6.1.3. Polysaccharides

Polysaccharides include HA, alginate, and chitosan. HA is a type of glycosaminoglycan in the extracellular matrix of connective tissue that is composed of repeating disaccharide, β-1, 4-methyl-D-glucuronic acid β-1, and N-methyl-acetyl-D-methyl-D-glucosamine [223]. HA is a powerful inflammatory mediator, and its unique biological and structural characteristics involve it in the regulation of wound repair and morphogenesis [224,225]. It has excellent biocompatibility, low immunogenicity and biological activity. Thus, HA has enormous potential as a scaffold for tissue regeneration [223]. It was found that HA hydrogel containing platelet lysate accelerated mineralized matrix deposition by hDPSCs, providing evidence of pulp regeneration [226]. Yang et al. [227] found that nerve conduits containing HA hydrogel contributed to the morphological and functional recovery of an injured sciatic nerve.
Chitosan is composed of N-acetyl glucosamine and glucosamine copolymer extracted from chitin [228]. The strengths of chitosan include excellent biocompatibility, biodegradability, low cytotoxicity, and low immunogenicity. It is antibacterial and exhibits outstanding mechanical properties [215,229]. A study investigated DPSCs and GFs combined with chitosan hydrogel and found that they regenerated pulp-dentin-like tissue and promoted root development in dogs [230]. The 3D porous chitosan scaffold constructed by Feng et al. [231] provided a favorable microenvironment for the attachment, survival, and neural differentiation of DPSCs. In addition, Chávez-Delgado et al. [232] successfully used chitosan combined with neurosteroids to regenerate facial nerves in rabbits.
Alginate is the polysaccharide extracted from seaweeds, with covalently linked 1mam 4 and linked β-D mannuronic acid (M) and α-L-guluronic acid (G) units [233]. It is biocompatible, non-cytotoxic, and non-immunogenic, and has been used in diabetes treatment, pancreatic transplantation, and drug/GF delivery systems [234]. One alginate gel containing TGF-β1 promoted odontoblast-like cell differentiation and upregulated dentin matrix secretion [235]. Alginate has insufficient mechanical strength and rapid degradation; thus, it usually needs to be crosslinked with other substances for neural regeneration [236].

6.1.4. Other Biomaterials

Matrigel is a bioactive soluble material derived from the basement membrane of tumor cells [237]. The function of Matrigel is similar to that of the extracellular matrix, and it is usually used as an artificial 3D surface for tissue engineering and differentiation of various cell types [238]. Luzuriaga et al. [239] achieved efficient angiogenesis in human DPSCs cultured with Matrigel. Furthermore, Jeong et al. [240] found that human DPSCs cultured in Matrigel differentiated into odontoblasts and formed pulp-like tissue. In terms of neural regeneration, it has been proven that Matrigel promotes the growth of ADSCs that successfully differentiate into dopaminergic cells [241].
Keratin is a type of natural fibrin associated with epithelial cells and skin appendages that can self-assemble into highly porous fiber scaffolds [218,242]. Gao et al. [243] reported that keratin promoted the proliferation of SCs and the extension of axons. Silk fibroin (SF) is a type of enzymatically degradable material that can be processed into a water-insoluble scaffold with excellent anticoagulant activity, regeneration ability, mechanical strength, elasticity, and a slow degradation rate [244]. An injectable silk fibroin–polydopamine composite hydrogel was fabricated and showed that it could support neuronal growth and promote SCI repair [245].

6.2. Synthetic Materials

Compared with natural materials, synthetic materials have better mechanical properties, controllable porosity, thermal stability, and a relatively cheap price. However, they are not as good as natural materials in terms of biocompatibility. This kind of material can be customized to form an ideal structure. The most common synthetic materials used for pulp regeneration are polylactic acid (PLA), poly-L-lactic acid (PLLA), polyepsiloncaprolactone (PCL), polylactic-co-glycolic (PLGA), polyglycolic acid (PGA), polydioxanone scaffolds (PDSs), and polyethylene glycol polylactic (PEG) [210]. Meanwhile, the most common biodegradable artificial materials for nerve regeneration are PGA, PLGA, PCL, and P (DLLA-co-CL) [246].

6.3. Composite Scaffolds

Overcoming the shortcomings of single scaffold materials through the combination of two or more materials is a new direction in the development of scaffold materials, such as bioceramic–polymer scaffolds, collagen–HA scaffolds, and collagen–alginate scaffolds (Figure 2). These composites can further promote cell adhesion and tissue/cell–cell interaction, thereby promoting cell proliferation, differentiation, and tissue regeneration [247].

7. Interaction between Nerve Regeneration and Maxillofacial Bone Regeneration

The preservation of teeth not only depends on the pulp but is also related to the supporting tissue, including the surrounding alveolar bone. Periapical periodontitis often leads to bone resorption around the apex, and significant bone loss eventually results in tooth extraction. Therefore, promoting the repair of jawbone defects is also key to the treatment of periapical diseases. However, the reconstruction and recovery of bone defects is still a complex matter [248,249,250,251]. An interaction between nerve and bone regeneration has recently been proposed. Brockes et al. [252] found that innervated limbs successfully regenerated while denervated limbs formed stumps after amputation in amphibians. Moreover, denervation before fingertip amputation in mice led to impaired regeneration and significant morphological defects. This emphasizes the neural dependency of bone injury repair and regeneration [253].
At present, the relationship between nerve regeneration and mandibular bone regeneration has only been preliminarily discussed. Jones et al. [254] confirmed that SCs’ paracrine signaling pathways are necessary for skeletal stem cells to promote fracture healing by constructing a mandibular denervation model. SCs, derived from the neural crest, are primary glial cells of the peripheral nervous system, which surround axons and promote impulse transmission [255]. Furthermore, SCs secrete paracrine NFs such as BDNF, NGF, and NT-3 to regulate bone regeneration upon bone injury [256,257]. BDNF mediates the effects of osteogenesis–neurogenesis–angiogenesis through the TrkB/ERK1/2 signaling pathway [258]. NGF can not only promote the differentiation, proliferation, and activity of osteoblasts, but also significantly accelerates fracture healing and the rate of bone mineralization [259,260,261]. NT-3 can induce osteogenesis, enhance vascularization, and increase the activity of osteoclasts [262,263]. Therefore, these GFs may be potential candidates to promote both bone regeneration and neural regeneration.

8. Limitations and Future Direction

Cell transplantation and cell homing have provided valuable insights into functional pulp regeneration and hold the promise to revolutionize RET and clinical options. However, there are still hurdles to address. First, although various stem cells exhibit the potential to form nerves in vitro and in vivo, the specific mechanism by which stem cells promote neural regeneration is still unknown. Thus, experiments such as lineage tracing and single-cell sequencing should be designed to explore which stem cells dominate neurogenesis during RET. Second, the most commonly used stem cells in transplantation are autologous, avoiding the problem of immunity. However, the in vitro isolation and expansion processes, the cell loss in the storage process, and the high cost are obstacles to clinical applications. It is necessary to find alternative cells and establish stem cell banks. Third, various GFs are added to regulate and promote functional pulp regeneration, and different combinations of factors will have diverse effects. Balancing the series of signals is a complex process. Other molecules that promote neurogenesis are also applicable in RET. For example, exosomes from stem cells are rich in bioactive components and have been proven to improve the repair of peripheral nerve injury [264]. Fourth, the properties of scaffolds for RET and neural regeneration requirements are dissimilar, and no innovative scaffolds have been specifically designed for neural regeneration during RET. Moreover, the scaffold should be compatible with the morphology of the root canal. Three-dimensional (3D) printing seems to be an option, with the potential to build scaffolds with a precise shape and structure to provide a suitable microenvironment for stem cells and GFs. It is expected that a functional pulp regeneration scaffold could be constructed by 3D printing in the near future with ideal biological activity and bionic vascular and nerve effects.

9. Conclusions

The field of RET has certainly made great strides over the course of recent years. Pulp revascularization has achieved satisfactory therapeutic outcomes with the restoration of sensitivity. Stem cell transplantation and cell homing are currently in the preclinical stage. The combination of dental MSCs with neurodifferentiation potential and neurotrophic factors that promote neural regeneration provides excellent conditions for the innervation of newborn dental-pulp-like tissues. Overall, we believe that the ultimate goal of functional pulp regeneration will be achieved.

Author Contributions

Y.W., P.L., X.C., Y.Y. and Z.L. collected the literature for review; Y.W., P.L., R.B. and Y.F. drafted the manuscript; Y.W., X.C., Y.Y. and Z.L. prepared the tables and figures; R.B. and Y.F. acquired the funding; Y.F. approved the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by NSFC grant 81800928, the Young Elite Scientist Sponsorship Program by CAST (No. 2018QNR001), the Sichuan Science and Technology Program (No. 2019YJ0054), Research Funding from West China School/Hospital of Stomatology Sichuan University (No. RCDWJS2021-1), and the State Key Laboratory of Oral Diseases Open Funding Grant SKLOD-R013.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that this research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Nör, J.E. Tooth regeneration in operative dentistry. Oper. Dent. 2006, 31, 633–642. [Google Scholar] [CrossRef] [PubMed]
  2. Fan, Y.; Zhou, Y.; Zhou, X.; Sun, F.; Gao, B.; Wan, M.; Zhou, X.; Sun, J.; Xu, X.; Cheng, L.; et al. MicroRNA 224 Regulates Ion Transporter Expression in Ameloblasts To Coordinate Enamel Mineralization. Mol. Cell. Biol. 2015, 35, 2875–2890. [Google Scholar] [CrossRef] [Green Version]
  3. Hashemi-Beni, B.; Khoroushi, M.; Foroughi, M.R.; Karbasi, S.; Khademi, A.A. Tissue engineering: Dentin—Pulp complex regeneration approaches (A review). Tissue Cell 2017, 49, 552–564. [Google Scholar] [CrossRef] [PubMed]
  4. Su, Y.; Wang, C.; Ye, L. Healing rate and post-obturation pain of single- versus multiple-visit endodontic treatment for infected root canals: A systematic review. J. Endod. 2011, 37, 125–132. [Google Scholar] [CrossRef] [PubMed]
  5. Murray, P.E.; Garcia-Godoy, F.; Hargreaves, K.M. Regenerative endodontics: A review of current status and a call for action. J. Endod. 2007, 33, 377–390. [Google Scholar] [CrossRef] [PubMed]
  6. Eramo, S.; Natali, A.; Pinna, R.; Milia, E. Dental pulp regeneration via cell homing. Int. Endod. J. 2018, 51, 405–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Xie, Z.; Shen, Z.; Zhan, P.; Yang, J.; Huang, Q.; Huang, S.; Chen, L.; Lin, Z. Functional Dental Pulp Regeneration: Basic Research and Clinical Translation. Int. J. Mol. Sci. 2021, 22, 8991. [Google Scholar] [CrossRef]
  8. Sui, B.; Chen, C.; Kou, X.; Li, B.; Xuan, K.; Shi, S.; Jin, Y. Pulp Stem Cell-Mediated Functional Pulp Regeneration. J. Dent. Res. 2019, 98, 27–35. [Google Scholar] [CrossRef]
  9. Hussain, A.; Tebyaniyan, H.; Khayatan, D. The Role of Epigenetic in Dental and Oral Regenerative Medicine by Different Types of Dental Stem Cells: A Comprehensive Overview. Stem Cells Int. 2022, 2022, 5304860. [Google Scholar] [CrossRef]
  10. Kim, J.Y.; Xin, X.; Moioli, E.K.; Chung, J.; Lee, C.H.; Chen, M.; Fu, S.Y.; Koch, P.D.; Mao, J.J. Regeneration of dental-pulp-like tissue by chemotaxis-induced cell homing. Tissue Eng. Part A 2010, 16, 3023–3031. [Google Scholar] [CrossRef]
  11. Hakim, L.K.; Yazdanian, M.; Alam, M.; Abbasi, K.; Tebyaniyan, H.; Tahmasebi, E.; Khayatan, D.; Seifalian, A.; Ranjbar, R.; Yazdanian, A. Biocompatible and Biomaterials Application in Drug Delivery System in Oral Cavity. Evid. Based Complement. Altern. Med. 2021, 2021, 9011226. [Google Scholar] [CrossRef] [PubMed]
  12. Fawzy El-Sayed, K.M.; Jakusz, K.; Jochens, A.; Dörfer, C.; Schwendicke, F. Stem Cell Transplantation for Pulpal Regeneration: A Systematic Review. Tissue Eng. Part B Rev. 2015, 21, 451–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ibuki, T.; Kido, M.A.; Kiyoshima, T.; Terada, Y.; Tanaka, T. An Ultrastructural Study of the Relationship between Sensory Trigeminal Nerves and Odontoblasts in Rat Dentin/Pulp as Demonstrated by the Anterograde Transport of Wheat Germ Agglutinin-Horseradish Peroxidase (WGA-HRP). J. Dent. Res. 1996, 75, 1963–1970. [Google Scholar] [CrossRef]
  14. Walton, R.E.; Ramachandran Nair, P.N. Neural elements in dental pulp and dentin. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endodontol. 1995, 80, 710–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Närhi, M. Interaction between the autonomic and sensory nerves in the dental pulp. Proc. Finn. Dent. Soc. 1989, 85, 389–393. [Google Scholar] [PubMed]
  16. Abd-Elmeguid, A.; Yu, D.C. Dental pulp neurophysiology: Part 1. Clinical and diagnostic implications. J. Can. Dent. Assoc. 2009, 75, 55–59. [Google Scholar]
  17. Longhurst, J.C.; Dittman, L.E. Hypoxia, bradykinin, and prostaglandins stimulate ischemically sensitive visceral afferents. Am. J. Physiol. 1987, 253, H556–H567. [Google Scholar] [CrossRef] [PubMed]
  18. Chmilewsky, F.; About, I.; Cooper, L.F.; Chung, S.H. C5L2 Silencing in Human Pulp Fibroblasts Enhances Nerve Outgrowth Under Lipoteichoic Acid Stimulation. J. Endod. 2018, 44, 1396–1401. [Google Scholar] [CrossRef]
  19. Zhan, C.; Huang, M.; Yang, X.; Hou, J. Dental nerves: A neglected mediator of pulpitis. Int. Endod. J. 2021, 54, 85–99. [Google Scholar] [CrossRef] [PubMed]
  20. Holzer, P. Neurogenic vasodilatation and plasma leakage in the skin. Gen. Pharmacol. Vasc. Syst. 1998, 30, 5–11. [Google Scholar] [CrossRef]
  21. Fristad, I.; Heyeraas, K.J.; Jonsson, R.; Kvinnsland, I.H. Effect of inferior alveolar nerve axotomy on immune cells and nerve fibres in young rat molars. Arch. Oral Biol. 1995, 40, 1053–1062. [Google Scholar] [CrossRef] [PubMed]
  22. Ho, W.Z.; Lai, J.P.; Zhu, X.H.; Uvaydova, M.; Douglas, S.D. Human monocytes and macrophages express substance P and neurokinin-1 receptor. J. Immunol. 1997, 159, 5654–5660. [Google Scholar] [PubMed]
  23. Diogenes, A. Trigeminal Sensory Neurons and Pulp Regeneration. J. Endod. 2020, 46, S71–S80. [Google Scholar] [CrossRef] [PubMed]
  24. Meschi, N.; Hilkens, P.; Lambrichts, I.; Van den Eynde, K.; Mavridou, A.; Strijbos, O.; De Ketelaere, M.; Van Gorp, G.; Lambrechts, P. Regenerative endodontic procedure of an infected immature permanent human tooth: An immunohistological study. Clin. Oral Investig. 2016, 20, 807–814. [Google Scholar] [CrossRef] [PubMed]
  25. Austah, O.; Joon, R.; Fath, W.M.; Chrepa, V.; Diogenes, A.; Ezeldeen, M.; Couve, E.; Ruparel, N.B. Comprehensive Characterization of 2 Immature Teeth Treated with Regenerative Endodontic Procedures. J. Endod. 2018, 44, 1802–1811. [Google Scholar] [CrossRef] [PubMed]
  26. Fried, K.; Nosrat, C.; Lillesaar, C.; Hildebrand, C. Molecular signaling and pulpal nerve development. Crit. Rev. Oral. Biol. Med. 2000, 11, 318–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Fried, K.; Lillesaar, C.; Sime, W.; Kaukua, N.; Patarroyo, M. Target finding of pain nerve fibers: Neural growth mechanisms in the tooth pulp. Physiol. Behav. 2007, 92, 40–45. [Google Scholar] [CrossRef]
  28. Caplan, A.I.; Dennis, J.E. Mesenchymal stem cells as trophic mediators. J. Cell. Biochem. 2006, 98, 1076–1084. [Google Scholar] [CrossRef]
  29. Mittal, N.; Baranwal, H.C.; Kumar, P.; Gupta, S. Assessment of pulp sensibility in the mature necrotic teeth using regenerative endodontic therapy with various scaffolds—Randomised clinical trial. Indian J. Dent. Res. 2021, 32, 216–220. [Google Scholar] [CrossRef]
  30. Guo, H.; Zhao, W.; Liu, A.; Wu, M.; Shuai, Y.; Li, B.; Huang, X.; Liu, X.; Yang, X.; Guo, X.; et al. SHED promote angiogenesis in stem cell-mediated dental pulp regeneration. Biochem. Biophys. Res. Commun. 2020, 529, 1158–1164. [Google Scholar] [CrossRef]
  31. Iohara, K.; Zayed, M.; Takei, Y.; Watanabe, H.; Nakashima, M. Treatment of Pulpectomized Teeth With Trypsin Prior to Transplantation of Mobilized Dental Pulp Stem Cells Enhances Pulp Regeneration in Aged Dogs. Front. Bioeng. Biotechnol. 2020, 8, 983. [Google Scholar] [CrossRef] [PubMed]
  32. Meng, H.; Hu, L.; Zhou, Y.; Ge, Z.; Wang, H.; Wu, C.T.; Jin, J. A Sandwich Structure of Human Dental Pulp Stem Cell Sheet, Treated Dentin Matrix, and Matrigel for Tooth Root Regeneration. Stem Cells Dev. 2020, 29, 521–532. [Google Scholar] [CrossRef] [PubMed]
  33. Iohara, K.; Utsunomiya, S.; Kohara, S.; Nakashima, M. Allogeneic transplantation of mobilized dental pulp stem cells with the mismatched dog leukocyte antigen type is safe and efficacious for total pulp regeneration. Stem Cell Res. Ther. 2018, 9, 116. [Google Scholar] [CrossRef] [Green Version]
  34. Xuan, K.; Li, B.; Guo, H.; Sun, W.; Kou, X.; He, X.; Zhang, Y.; Sun, J.; Liu, A.; Liao, L.; et al. Deciduous autologous tooth stem cells regenerate dental pulp after implantation into injured teeth. Sci. Transl. Med. 2018, 10, eaaf3227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Li, L.; Wang, Z.L. PDGF-BB, NGF and BDNF enhance pulp-like tissue regeneration via cell homing. RSC Adv. 2016, 6, 109519–109527. [Google Scholar] [CrossRef]
  36. Murakami, M.; Hayashi, Y.; Iohara, K.; Osako, Y.; Hirose, Y.; Nakashima, M. Trophic Effects and Regenerative Potential of Mobilized Mesenchymal Stem Cells From Bone Marrow and Adipose Tissue as Alternative Cell Sources for Pulp/Dentin Regeneration. Cell Transpl. 2015, 24, 1753–1765. [Google Scholar] [CrossRef]
  37. Iohara, K.; Murakami, M.; Nakata, K.; Nakashima, M. Age-dependent decline in dental pulp regeneration after pulpectomy in dogs. Exp. Gerontol. 2014, 52, 39–45. [Google Scholar] [CrossRef]
  38. Souron, J.B.; Petiet, A.; Decup, F.; Tran, X.V.; Lesieur, J.; Poliard, A.; Le Guludec, D.; Letourneur, D.; Chaussain, C.; Rouzet, F.; et al. Pulp cell tracking by radionuclide imaging for dental tissue engineering. Tissue Eng. Part C Methods 2014, 20, 188–197. [Google Scholar] [CrossRef] [Green Version]
  39. Iohara, K.; Murakami, M.; Takeuchi, N.; Osako, Y.; Ito, M.; Ishizaka, R.; Utunomiya, S.; Nakamura, H.; Matsushita, K.; Nakashima, M. A novel combinatorial therapy with pulp stem cells and granulocyte colony-stimulating factor for total pulp regeneration. Stem Cells Transl. Med. 2013, 2, 521–533. [Google Scholar] [CrossRef]
  40. Ishizaka, R.; Iohara, K.; Murakami, M.; Fukuta, O.; Nakashima, M. Regeneration of dental pulp following pulpectomy by fractionated stem/progenitor cells from bone marrow and adipose tissue. Biomaterials 2012, 33, 2109–2118. [Google Scholar] [CrossRef]
  41. Iohara, K.; Imabayashi, K.; Ishizaka, R.; Watanabe, A.; Nabekura, J.; Ito, M.; Matsushita, K.; Nakamura, H.; Nakashima, M. Complete pulp regeneration after pulpectomy by transplantation of CD105+ stem cells with stromal cell-derived factor-1. Tissue Eng. Part A 2011, 17, 1911–1920. [Google Scholar] [CrossRef] [PubMed]
  42. Sajjad, I.; Sajid, M.; Munir, B.; Akhlaq, H.; Zehra, T.; Ahmed, J. Success of Revascularization of Pulp in Necrotic Maxillary Anterior Immature Permanent Teeth. Pakiatan J. Med. Health Sci. 2022, 16, 420–422. [Google Scholar] [CrossRef]
  43. Youssef, A.; Ali, M.; ElBolok, A.; Hassan, R. Regenerative endodontic procedures for the treatment of necrotic mature teeth: A preliminary randomized clinical trial. Int. Endod. J. 2022, 55, 334–346. [Google Scholar] [CrossRef] [PubMed]
  44. Abu Zeid, S.T.; Alamoudi, R.A.; Alothmani, O.S.; Mokeem Saleh, A.A.; Siddiqui, A.Y. A Prospective Study of Long-Term Regenerative Endodontics Outcomes of Necrotic Immature Permanent Teeth: An 8-Year Follow-Up. Healthcare 2021, 9, 1670. [Google Scholar] [CrossRef] [PubMed]
  45. Brizuela, C.; Meza, G.; Urrejola, D.; Quezada, M.A.; Concha, G.; Ramírez, V.; Angelopoulos, I.; Cadiz, M.I.; Tapia-Limonchi, R.; Khoury, M. Cell-Based Regenerative Endodontics for Treatment of Periapical Lesions: A Randomized, Controlled Phase I/II Clinical Trial. J. Dent. Res. 2020, 99, 523–529. [Google Scholar] [CrossRef] [PubMed]
  46. Chrepa, V.; Joon, R.; Austah, O.; Diogenes, A.; Hargreaves, K.M.; Ezeldeen, M.; Ruparel, N.B. Clinical Outcomes of Immature Teeth Treated with Regenerative Endodontic Procedures-A San Antonio Study. J. Endod. 2020, 46, 1074–1084. [Google Scholar] [CrossRef]
  47. Mittmann, C.W.; Kostka, E.; Ballout, H.; Preus, M.; Preissner, R.; Karaman, M.; Preissner, S. Outcome of revascularization therapy in traumatized immature incisors. BMC Oral Health 2020, 20, 207. [Google Scholar] [CrossRef]
  48. Nagaveni, N.B.; Poornima, P.; Mathew, M.G.; Soni, A.J.; Khan, M.M. A Comparative Evaluation of Revascularization Done in Traumatized Immature, Necrotic Anterior Teeth with and without Platelet-rich Fibrin: A Case Report. Int. J. Clin. Pediatr. Dent. 2020, 13, 98–102. [Google Scholar] [CrossRef]
  49. Nazzal, H.; Ainscough, S.; Kang, J.; Duggal, M.S. Revitalisation endodontic treatment of traumatised immature teeth: A prospective long-term clinical study. Eur. Arch. Paediatr. Dent. 2020, 21, 587–596. [Google Scholar] [CrossRef] [Green Version]
  50. Arslan, H.; Ahmed, H.M.A.; Şahin, Y.; Doğanay Yıldız, E.; Gündoğdu, E.C.; Güven, Y.; Khalilov, R. Regenerative Endodontic Procedures in Necrotic Mature Teeth with Periapical Radiolucencies: A Preliminary Randomized Clinical Study. J. Endod. 2019, 45, 863–872. [Google Scholar] [CrossRef]
  51. Meza, G.; Urrejola, D.; Saint Jean, N.; Inostroza, C.; López, V.; Khoury, M.; Brizuela, C. Personalized Cell Therapy for Pulpitis Using Autologous Dental Pulp Stem Cells and Leukocyte Platelet-rich Fibrin: A Case Report. J. Endod. 2019, 45, 144–149. [Google Scholar] [CrossRef] [PubMed]
  52. Ulusoy, A.T.; Turedi, I.; Cimen, M.; Cehreli, Z.C. Evaluation of Blood Clot, Platelet-rich Plasma, Platelet-rich Fibrin, and Platelet Pellet as Scaffolds in Regenerative Endodontic Treatment: A Prospective Randomized Trial. J. Endod. 2019, 45, 560–566. [Google Scholar] [CrossRef] [PubMed]
  53. Lv, H.; Chen, Y.; Cai, Z.; Lei, L.; Zhang, M.; Zhou, R.; Huang, X. The efficacy of platelet-rich fibrin as a scaffold in regenerative endodontic treatment: A retrospective controlled cohort study. BMC Oral Health 2018, 18, 139. [Google Scholar] [CrossRef] [PubMed]
  54. Mustafa, M. Maturogenesis and Osseous Healing of a Necrotic Immature Premolar using Revascularization Procedure—A Case Report and Review of Literature. Ann. Med. Health Sci. Res. 2018, 8, 39–44. [Google Scholar]
  55. Nageh, M.; Ahmed, G.M.; El-Baz, A.A. Assessment of Regaining Pulp Sensibility in Mature Necrotic Teeth Using a Modified Revascularization Technique with Platelet-rich Fibrin: A Clinical Study. J. Endod. 2018, 44, 1526–1533. [Google Scholar] [CrossRef] [PubMed]
  56. Nazzal, H.; Kenny, K.; Altimimi, A.; Kang, J.; Duggal, M.S. A prospective clinical study of regenerative endodontic treatment of traumatized immature teeth with necrotic pulps using bi-antibiotic paste. Int. Endod. J. 2018, 51 (Suppl. S3), e204–e215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Neelamurthy, P.S.; Kumar, R.A.; Balakrishnan, V.; Venkatesan, S.M.; Narayan, G.S.; Karthikeyan, I. Revascularization in Immature and Mature Teeth with Necrotic Pulp: A Clinical Study. J. Contemp. Dent. Pract. 2018, 19, 1393–1399. [Google Scholar] [PubMed]
  58. Alagl, A.; Bedi, S.; Hassan, K.; AlHumaid, J. Use of platelet-rich plasma for regeneration in non-vital immature permanent teeth: Clinical and cone-beam computed tomography evaluation. J. Int. Med. Res. 2017, 45, 583–593. [Google Scholar] [CrossRef] [PubMed]
  59. Jiang, X.; Liu, H.; Peng, C. Clinical and Radiographic Assessment of the Efficacy of a Collagen Membrane in Regenerative Endodontics: A Randomized, Controlled Clinical Trial. J. Endod. 2017, 43, 1465–1471. [Google Scholar] [CrossRef]
  60. Li, L.; Pan, Y.; Mei, L.; Li, J. Clinical and Radiographic Outcomes in Immature Permanent Necrotic Evaginated Teeth Treated with Regenerative Endodontic Procedures. J. Endod. 2017, 43, 246–251. [Google Scholar] [CrossRef]
  61. Shivashankar, V.Y.; Johns, D.A.; Maroli, R.K.; Sekar, M.; Chandrasekaran, R.; Karthikeyan, S.; Renganathan, S.K. Comparison of the Effect of PRP, PRF and Induced Bleeding in the Revascularization of Teeth with Necrotic Pulp and Open Apex: A Triple Blind Randomized Clinical Trial. J. Clin. Diagn. Res. 2017, 11, Zc34–Zc39. [Google Scholar] [CrossRef] [PubMed]
  62. Ulusoy, A.T.; Cehreli, Z.C. Regenerative Endodontic Treatment of Necrotic Primary Molars with Missing Premolars: A Case Series. Pediatr. Dent. 2017, 39, 131–134. [Google Scholar] [PubMed]
  63. Farhad, A.R.; Shokraneh, A.; Shekarchizade, N. Regeneration or replacement? A case report and review of literature. Dent. Traumatol. 2016, 32, 71–79. [Google Scholar] [CrossRef] [PubMed]
  64. Nagaveni, N.B.; Pathak, S.; Poornima, P.; Joshi, J.S. Revascularization Induced Maturogenesis of Non-Vital Immature Permanent Tooth Using Platelet-Rich-Fibrin: A Case Report. J. Clin. Pediatr. Dent. 2016, 40, 26–30. [Google Scholar] [CrossRef] [PubMed]
  65. Priya, M.H.; Tambakad, P.B.; Naidu, J. Pulp and Periodontal Regeneration of an Avulsed Permanent Mature Incisor Using Platelet-rich Plasma after Delayed Replantation: A 12-month Clinical Case Study. J. Endod. 2016, 42, 66–71. [Google Scholar] [CrossRef] [PubMed]
  66. Ray, H.L., Jr.; Marcelino, J.; Braga, R.; Horwat, R.; Lisien, M.; Khaliq, S. Long-term follow up of revascularization using platelet-rich fibrin. Dent. Traumatol. 2016, 32, 80–84. [Google Scholar] [CrossRef]
  67. Subash, D.; Shoba, K.; Aman, S.; Bharkavi, S.K. Revitalization of an Immature Permanent Mandibular Molar with a Necrotic Pulp Using Platelet-Rich Fibrin: A Case Report. J. Clin. Diagn. Res. 2016, 10, Zd21–Zd23. [Google Scholar] [CrossRef]
  68. Bezgin, T.; Yilmaz, A.D.; Celik, B.N.; Kolsuz, M.E.; Sonmez, H. Efficacy of platelet-rich plasma as a scaffold in regenerative endodontic treatment. J. Endod. 2015, 41, 36–44. [Google Scholar] [CrossRef]
  69. Dudeja, P.G.; Grover, S.; Srivastava, D.; Dudeja, K.K.; Sharma, V. Pulp Revascularization- It’s your Future Whether you Know it or Not? J. Clin. Diagn. Res. 2015, 9, Zr01–Zr04. [Google Scholar] [CrossRef]
  70. Lei, L.; Chen, Y.; Zhou, R.; Huang, X.; Cai, Z. Histologic and Immunohistochemical Findings of a Human Immature Permanent Tooth with Apical Periodontitis after Regenerative Endodontic Treatment. J. Endod. 2015, 41, 1172–1179. [Google Scholar] [CrossRef]
  71. Nagaveni, N.B.; Poornima, P.; Joshi, J.S.; Pathak, S.; Nandini, D.B. Revascularization of immature, nonvital permanent tooth using platelet-rich fibrin in children. Pediatr. Dent. 2015, 37, 1–6. [Google Scholar] [PubMed]
  72. Sachdeva, G.S.; Sachdeva, L.T.; Goel, M.; Bala, S. Regenerative endodontic treatment of an immature tooth with a necrotic pulp and apical periodontitis using platelet-rich plasma (PRP) and mineral trioxide aggregate (MTA): A case report. Int. Endod. J. 2015, 48, 902–910. [Google Scholar] [CrossRef] [PubMed]
  73. Kahler, B.; Mistry, S.; Moule, A.; Ringsmuth, A.K.; Case, P.; Thomson, A.; Holcombe, T. Revascularization outcomes: A prospective analysis of 16 consecutive cases. J. Endod. 2014, 40, 333–338. [Google Scholar] [CrossRef] [PubMed]
  74. Farsi, N.; Abuzeid, S.; El Ashiry, E. Revascularization of dental pulp in human necrotic permanent teeth with immature apex: Three case reports. Life Sci. J. 2013, 10, 1516–1521. [Google Scholar]
  75. Keswani, D.; Pandey, R.K. Revascularization of an immature tooth with a necrotic pulp using platelet-rich fibrin: A case report. Int. Endod. J. 2013, 46, 1096–1104. [Google Scholar] [CrossRef]
  76. Mishra, N.; Narang, I.; Mittal, N. Platelet-rich fibrin-mediated revitalization of immature necrotic tooth. Contemp. Clin. Dent. 2013, 4, 412–415. [Google Scholar] [CrossRef]
  77. Cehreli, Z.C.; Sara, S.; Aksoy, B. Revascularization of immature permanent incisors after severe extrusive luxation injury. Tex. Dent. J. 2012, 129, 675–681. [Google Scholar]
  78. Miller, E.K.; Lee, J.Y.; Tawil, P.Z.; Teixeira, F.B.; Vann, W.F., Jr. Emerging therapies for the management of traumatized immature permanent incisors. Pediatr. Dent. 2012, 34, 66–69. [Google Scholar]
  79. Shivashankar, V.Y.; Johns, D.A.; Vidyanath, S.; Kumar, M.R. Platelet Rich Fibrin in the revitalization of tooth with necrotic pulp and open apex. J. Conserv. Dent. 2012, 15, 395–398. [Google Scholar] [CrossRef] [Green Version]
  80. Cehreli, Z.C.; Isbitiren, B.; Sara, S.; Erbas, G. Regenerative endodontic treatment (revascularization) of immature necrotic molars medicated with calcium hydroxide: A case series. J. Endod. 2011, 37, 1327–1330. [Google Scholar] [CrossRef]
  81. Iwaya, S.; Ikawa, M.; Kubota, M. Revascularization of an immature permanent tooth with periradicular abscess after luxation. Dent. Traumatol. 2011, 27, 55–58. [Google Scholar] [CrossRef] [PubMed]
  82. Torabinejad, M.; Turman, M. Revitalization of tooth with necrotic pulp and open apex by using platelet-rich plasma: A case report. J. Endod. 2011, 37, 265–268. [Google Scholar] [CrossRef] [PubMed]
  83. Petrino, J.A.; Boda, K.K.; Shambarger, S.; Bowles, W.R.; McClanahan, S.B. Challenges in regenerative endodontics: A case series. J. Endod. 2010, 36, 536–541. [Google Scholar] [CrossRef] [PubMed]
  84. Thomson, A.; Kahler, B. Regenerative endodontics--biologically-based treatment for immature permanent teeth: A case report and review of the literature. Aust. Dent. J. 2010, 55, 446–452. [Google Scholar] [CrossRef] [PubMed]
  85. Ding, R.Y.; Cheung, G.S.; Chen, J.; Yin, X.Z.; Wang, Q.Q.; Zhang, C.F. Pulp revascularization of immature teeth with apical periodontitis: A clinical study. J. Endod. 2009, 35, 745–749. [Google Scholar] [CrossRef] [PubMed]
  86. Reynolds, K.; Johnson, J.D.; Cohenca, N. Pulp revascularization of necrotic bilateral bicuspids using a modified novel technique to eliminate potential coronal discolouration: A case report. Int. Endod. J. 2009, 42, 84–92. [Google Scholar] [CrossRef] [PubMed]
  87. Banchs, F.; Trope, M. Revascularization of immature permanent teeth with apical periodontitis: New treatment protocol? J. Endod. 2004, 30, 196–200. [Google Scholar] [CrossRef]
  88. Iwaya, S.I.; Ikawa, M.; Kubota, M. Revascularization of an immature permanent tooth with apical periodontitis and sinus tract. Dent. Traumatol. 2001, 17, 185–187. [Google Scholar] [CrossRef] [Green Version]
  89. Botelho, J.; Cavacas, M.A.; Machado, V.; Mendes, J.J. Dental stem cells: Recent progresses in tissue engineering and regenerative medicine. Ann. Med. 2017, 49, 644–651. [Google Scholar] [CrossRef]
  90. Lyu, P.; Li, B.; Li, P.; Bi, R.; Cui, C.; Zhao, Z.; Zhou, X.; Fan, Y. Parathyroid Hormone 1 Receptor Signaling in Dental Mesenchymal Stem Cells: Basic and Clinical Implications. Front. Cell Dev. Biol. 2021, 9, 654715. [Google Scholar] [CrossRef]
  91. Bi, R.; Lyu, P.; Song, Y.; Li, P.; Song, D.; Cui, C.; Fan, Y. Function of Dental Follicle Progenitor/Stem Cells and Their Potential in Regenerative Medicine: From Mechanisms to Applications. Biomolecules 2021, 11, 0997. [Google Scholar] [CrossRef] [PubMed]
  92. Soudi, A.; Yazdanian, M.; Ranjbar, R.; Tebyanian, H.; Yazdanian, A.; Tahmasebi, E.; Keshvad, A.; Seifalian, A. Role and application of stem cells in dental regeneration: A comprehensive overview. Excli. J. 2021, 20, 454–489. [Google Scholar] [CrossRef] [PubMed]
  93. Cui, C.; Bi, R.; Liu, W.; Guan, S.; Li, P.; Song, D.; Xu, R.; Zheng, L.; Yuan, Q.; Zhou, X.; et al. Role of PTH1R Signaling in Prx1(+) Mesenchymal Progenitors during Eruption. J. Dent. Res. 2020, 99, 1296–1305. [Google Scholar] [CrossRef]
  94. Gronthos, S.; Mankani, M.; Brahim, J.; Robey, P.G.; Shi, S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 13625–13630. [Google Scholar] [CrossRef] [Green Version]
  95. Mattei, V.; Santacroce, C.; Tasciotti, V.; Martellucci, S.; Santilli, F.; Manganelli, V.; Piccoli, L.; Misasi, R.; Sorice, M.; Garofalo, T. Role of lipid rafts in neuronal differentiation of dental pulp-derived stem cells. Exp. Cell Res. 2015, 339, 231–240. [Google Scholar] [CrossRef] [PubMed]
  96. Venugopal, C.; Rai, K.S.; Pinnelli, V.B.; Kutty, B.M.; Dhanushkodi, A. Neuroprotection by Human Dental Pulp Mesenchymal Stem Cells: From Billions to Nano. Curr. Gene Ther. 2018, 18, 307–323. [Google Scholar] [CrossRef]
  97. Wang, L.H.; Gao, S.Z.; Bai, X.L.; Chen, Z.L.; Yang, F. An Up-To-Date Overview of Dental Tissue Regeneration Using Dental Origin Mesenchymal Stem Cells: Challenges and Road Ahead. Front. Bioeng. Biotechnol. 2022, 10, 855396. [Google Scholar] [CrossRef] [PubMed]
  98. Kaukua, N.; Shahidi, M.K.; Konstantinidou, C.; Dyachuk, V.; Kaucka, M.; Furlan, A.; An, Z.; Wang, L.; Hultman, I.; Ahrlund-Richter, L.; et al. Glial origin of mesenchymal stem cells in a tooth model system. Nature 2014, 513, 551–554. [Google Scholar] [CrossRef] [Green Version]
  99. 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]
  100. Ahmed, M.N.; Shi, D.; Dailey, M.T.; Rothermund, K.; Drewry, M.D.; Calabrese, T.C.; Cui, X.T.; Syed-Picard, F.N. Dental Pulp Cell Sheets Enhance Facial Nerve Regeneration via Local Neurotrophic Factor Delivery. Tissue Eng. Part A 2021, 27, 1128–1139. [Google Scholar] [CrossRef]
  101. Sharma, Y.; Shobha, K.; Sundeep, M.; Pinnelli, V.B.; Parveen, S.; Dhanushkodi, A. Neural Basis of Dental Pulp Stem Cells and its Potential Application in Parkinson’s Disease. CNS Neurol. Disord. Drug Targets 2022, 21, 62–76. [Google Scholar] [CrossRef] [PubMed]
  102. Mead, B.; Hill, L.J.; Blanch, R.J.; Ward, K.; Logan, A.; Berry, M.; Leadbeater, W.; Scheven, B.A. Mesenchymal stromal cell-mediated neuroprotection and functional preservation of retinal ganglion cells in a rodent model of glaucoma. Cytotherapy 2016, 18, 487–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kolar, M.K.; Itte, V.N.; Kingham, P.J.; Novikov, L.N.; Wiberg, M.; Kelk, P. The neurotrophic effects of different human dental mesenchymal stem cells. Sci. Rep. 2017, 7, 12605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Mu, X.D.; Liu, H.H.; Li, Y.F.; Xiang, L.; Hu, M. Research progress of dental pulp stem cells for peripheral nerve injury repair. Zhonghua Kou Qiang Yi Xue Za Zhi 2022, 57, 196–201. [Google Scholar] [CrossRef] [PubMed]
  105. Iohara, K.; Zheng, L.; Ito, M.; Ishizaka, R.; Nakamura, H.; Into, T.; Matsushita, K.; Nakashima, M. Regeneration of dental pulp after pulpotomy by transplantation of CD31(-)/CD146(-) side population cells from a canine tooth. Regen. Med. 2009, 4, 377–385. [Google Scholar] [CrossRef]
  106. Nakamura, S.; Yamada, Y.; Katagiri, W.; Sugito, T.; Ito, K.; Ueda, M. Stem Cell Proliferation Pathways Comparison between Human Exfoliated Deciduous Teeth and Dental Pulp Stem Cells by Gene Expression Profile from Promising Dental Pulp. J. Endod. 2009, 35, 1536–1542. [Google Scholar] [CrossRef] [PubMed]
  107. Martinez Saez, D.; Sasaki, R.T.; Neves, A.D.; da Silva, M.C. Stem Cells from Human Exfoliated Deciduous Teeth: A Growing Literature. Cells Tissues Organs 2016, 202, 269–280. [Google Scholar] [CrossRef]
  108. 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] [Green Version]
  109. Bar, J.K.; Lis-Nawara, A.; Grelewski, P.G. Dental Pulp Stem Cell-Derived Secretome and Its Regenerative Potential. Int. J. Mol. Sci. 2021, 22, 12018. [Google Scholar] [CrossRef]
  110. Oubenyahya, H. Stem Cells from Dental Pulp of Human Exfoliated Teeth: Current Understanding and Future Challenges in Dental Tissue Engineering. Chin. J. Dent. Res. 2021, 24, 9–20. [Google Scholar] [CrossRef]
  111. Kerkis, I.; Caplan, A.I. Stem cells in dental pulp of deciduous teeth. Tissue Eng. Part B Rev. 2012, 18, 129–138. [Google Scholar] [CrossRef] [PubMed]
  112. Pereira, L.V.; Bento, R.F.; Cruz, D.B.; Marchi, C.; Salomone, R.; Oiticicca, J.; Costa, M.P.; Haddad, L.A.; Mingroni-Netto, R.C.; Costa, H. Stem Cells from Human Exfoliated Deciduous Teeth (SHED) Differentiate in vivo and Promote Facial Nerve Regeneration. Cell Transpl. 2019, 28, 55–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Feng, X.; Xing, J.; Feng, G.; Sang, A.; Shen, B.; Xu, Y.; Jiang, J.; Liu, S.; Tan, W.; Gu, Z.; et al. Age-dependent impaired neurogenic differentiation capacity of dental stem cell is associated with Wnt/β-catenin signaling. Cell. Mol. Neurobiol. 2013, 33, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  114. Casagrande, L.; Cordeiro, M.M.; Nör, S.A.; Nör, J.E. Dental pulp stem cells in regenerative dentistry. Odontology 2011, 99, 1–7. [Google Scholar] [CrossRef]
  115. Rosa, V.; Zhang, Z.; Grande, R.H.; Nör, J.E. Dental pulp tissue engineering in full-length human root canals. J. Dent. Res. 2013, 92, 970–975. [Google Scholar] [CrossRef]
  116. 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]
  117. Chen, F.M.; Gao, L.N.; Tian, B.M.; Zhang, X.Y.; Zhang, Y.J.; Dong, G.Y.; Lu, H.; Chu, Q.; Xu, J.; Yu, Y.; et al. Treatment of periodontal intrabony defects using autologous periodontal ligament stem cells: A randomized clinical trial. Stem. Cell Res. Ther. 2016, 7, 33. [Google Scholar] [CrossRef] [Green Version]
  118. Tomokiyo, A.; Wada, N.; Maeda, H. Periodontal Ligament Stem Cells: Regenerative Potency in Periodontium. Stem. Cells Dev. 2019, 28, 974–985. [Google Scholar] [CrossRef]
  119. Queiroz, A.; Albuquerque-Souza, E.; Gasparoni, L.M.; de França, B.N.; Pelissari, C.; Trierveiler, M.; Holzhausen, M. Therapeutic potential of periodontal ligament stem cells. World J. Stem. Cells 2021, 13, 605–618. [Google Scholar] [CrossRef]
  120. Bueno, C.; Ramirez, C.; Rodríguez-Lozano, F.J.; Tabarés-Seisdedos, R.; Rodenas, M.; Moraleda, J.M.; Jones, J.R.; Martinez, S. Human adult periodontal ligament-derived cells integrate and differentiate after implantation into the adult mammalian brain. Cell Transpl. 2013, 22, 2017–2028. [Google Scholar] [CrossRef] [Green Version]
  121. Fortino, V.R.; Chen, R.S.; Pelaez, D.; Cheung, H.S. Neurogenesis of neural crest-derived periodontal ligament stem cells by EGF and bFGF. J. Cell Physiol. 2014, 229, 479–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zhai, Q.; Dong, Z.; Wang, W.; Li, B.; Jin, Y. Dental stem cell and dental tissue regeneration. Front. Med. 2019, 13, 152–159. [Google Scholar] [CrossRef]
  123. Janebodin, K.; Horst, O.V.; Ieronimakis, N.; Balasundaram, G.; Reesukumal, K.; Pratumvinit, B.; Reyes, M. Isolation and characterization of neural crest-derived stem cells from dental pulp of neonatal mice. PLoS ONE 2011, 6, e27526. [Google Scholar] [CrossRef] [PubMed]
  124. Smeda, M.; Galler, K.M.; Woelflick, M.; Rosendahl, A.; Moehle, C.; Lenhardt, B.; Buchalla, W.; Widbiller, M. Molecular Biological Comparison of Dental Pulp- and Apical Papilla-Derived Stem Cells. Int. J. Mol. Sci. 2022, 23, 2615. [Google Scholar] [CrossRef]
  125. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  126. Sonoyama, W.; Liu, Y.; Yamaza, T.; Tuan, R.S.; Wang, S.; Shi, S.; Huang, G.T. Characterization of the apical papilla and its residing stem cells from human immature permanent teeth: A pilot study. J. Endod. 2008, 34, 166–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Bakopoulou, A.; About, I. Stem Cells of Dental Origin: Current Research Trends and Key Milestones towards Clinical Application. Stem. Cells Int. 2016, 2016, 4209891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Huang, G.T.; Sonoyama, W.; Liu, Y.; Liu, H.; Wang, S.; Shi, S. The hidden treasure in apical papilla: The potential role in pulp/dentin regeneration and bioroot engineering. J. Endod. 2008, 34, 645–651. [Google Scholar] [CrossRef] [Green Version]
  129. Aydin, S.; Şahin, F. Stem Cells Derived from Dental Tissues. Adv. Exp. Med. Biol. 2019, 1144, 123–132. [Google Scholar] [CrossRef]
  130. Kim, S.G. A Cell-Based Approach to Dental Pulp Regeneration Using Mesenchymal Stem Cells: A Scoping Review. Int. J. Mol. Sci. 2021, 22, 4357. [Google Scholar] [CrossRef]
  131. de Almeida, J.F.; Chen, P.; Henry, M.A.; Diogenes, A. Stem cells of the apical papilla regulate trigeminal neurite outgrowth and targeting through a BDNF-dependent mechanism. Tissue Eng. Part A 2014, 20, 3089–3100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Sequeira, D.B.; Oliveira, A.R.; Seabra, C.M.; Palma, P.J.; Ramos, C.; Figueiredo, M.H.; Santos, A.C.; Cardoso, A.L.; Peça, J.; Santos, J.M. Regeneration of pulp-dentin complex using human stem cells of the apical papilla: In vivo interaction with two bioactive materials. Clin. Oral Investig. 2021, 25, 5317–5329. [Google Scholar] [CrossRef] [PubMed]
  133. Araújo, P.R.S.; Silva, L.B.; Neto, A.; Almeida de Arruda, J.A.; Álvares, P.R.; Sobral, A.P.V.; Júnior, S.A.; Leão, J.C.; Braz da Silva, R.; Sampaio, G.C. Pulp Revascularization: A Literature Review. Open. Dent. J. 2017, 10, 48–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Bernardo, M.E.; Locatelli, F.; Fibbe, W.E. Mesenchymal stromal cells. Ann. N. Y. Acad. Sci. 2009, 1176, 101–117. [Google Scholar] [CrossRef] [PubMed]
  135. Li, H.; Ghazanfari, R.; Zacharaki, D.; Lim, H.C.; Scheding, S. Isolation and characterization of primary bone marrow mesenchymal stromal cells. Ann. N. Y. Acad. Sci. 2016, 1370, 109–118. [Google Scholar] [CrossRef] [PubMed]
  136. Zhu, H.; Guo, Z.K.; Jiang, X.X.; Li, H.; Wang, X.Y.; Yao, H.Y.; Zhang, Y.; Mao, N. A protocol for isolation and culture of mesenchymal stem cells from mouse compact bone. Nat. Protoc. 2010, 5, 550–560. [Google Scholar] [CrossRef]
  137. Baksh, D.; Song, L.; Tuan, R.S. Adult mesenchymal stem cells: Characterization, differentiation, and application in cell and gene therapy. J. Cell. Mol. Med. 2004, 8, 301–316. [Google Scholar] [CrossRef]
  138. Kaneko, T.; Gu, B.; Sone, P.P.; Zaw, S.Y.M.; Murano, H.; Zaw, Z.C.T.; Okiji, T. Dental Pulp Tissue Engineering Using Mesenchymal Stem Cells: A Review with a Protocol. Stem Cell Rev. Rep. 2018, 14, 668–676. [Google Scholar] [CrossRef]
  139. Ohazama, A.; Modino, S.A.; Miletich, I.; Sharpe, P.T. Stem-cell-based tissue engineering of murine teeth. J. Dent. Res. 2004, 83, 518–522. [Google Scholar] [CrossRef]
  140. Hu, B.; Unda, F.; Bopp-Kuchler, S.; Jimenez, L.; Wang, X.J.; Haïkel, Y.; Wang, S.L.; Lesot, H. Bone Marrow Cells Can Give Rise to Ameloblast-like Cells. J. Dent. Res. 2006, 85, 416–421. [Google Scholar] [CrossRef]
  141. Yang, Y.; Rossi, F.M.; Putnins, E.E. Periodontal regeneration using engineered bone marrow mesenchymal stromal cells. Biomaterials 2010, 31, 8574–8582. [Google Scholar] [CrossRef]
  142. Hu, L.; Liu, Y.; Wang, S. Stem cell-based tooth and periodontal regeneration. Oral Dis. 2018, 24, 696–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Yi, S.; Zhang, Y.; Gu, X.; Huang, L.; Zhang, K.; Qian, T.; Gu, X. Application of stem cells in peripheral nerve regeneration. Burn. Trauma 2020, 8, tkaa002. [Google Scholar] [CrossRef]
  144. Azizi, S.A.; Stokes, D.; Augelli, B.J.; DiGirolamo, C.; Prockop, D.J. Engraftment and migration of human bone marrow stromal cells implanted in the brains of albino rats--similarities to astrocyte grafts. Proc. Natl. Acad. Sci. USA 1998, 95, 3908–3913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Mathot, F.; Saffari, T.M.; Rbia, N.; Nijhuis, T.H.J.; Bishop, A.T.; Hovius, S.E.R.; Shin, A.Y. Functional Outcomes of Nerve Allografts Seeded with Undifferentiated and Differentiated Mesenchymal Stem Cells in a Rat Sciatic Nerve Defect Model. Plast. Reconstr. Surg. 2021, 148, 354–365. [Google Scholar] [CrossRef] [PubMed]
  146. Wen, R.; Wang, X.; Lu, Y.; Du, Y.; Yu, X. The combined application of rat bone marrow mesenchymal stem cells and bioceramic materials in the regeneration of dental pulp-like tissues. Int. J. Clin. Exp. Pathol. 2020, 13, 1492–1499. [Google Scholar]
  147. Zuk, P.A.; Zhu, M.; Mizuno, H.; Huang, J.; Futrell, J.W.; Katz, A.J.; Benhaim, P.; Lorenz, H.P.; Hedrick, M.H. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Eng. 2001, 7, 211–228. [Google Scholar] [CrossRef] [Green Version]
  148. Chu, D.T.; Nguyen Thi Phuong, T.; Tien, N.L.B.; Tran, D.K.; Minh, L.B.; Thanh, V.V.; Gia Anh, P.; Pham, V.H.; Thi Nga, V. Adipose Tissue Stem Cells for Therapy: An Update on the Progress of Isolation, Culture, Storage, and Clinical Application. J. Clin. Med. 2019, 8, 917. [Google Scholar] [CrossRef] [Green Version]
  149. Fan, Y.; Hanai, J.I.; Le, P.T.; Bi, R.; Maridas, D.; DeMambro, V.; Figueroa, C.A.; Kir, S.; Zhou, X.; Mannstadt, M.; et al. Parathyroid Hormone Directs Bone Marrow Mesenchymal Cell Fate. Cell Metab. 2017, 25, 661–672. [Google Scholar] [CrossRef] [Green Version]
  150. Gronthos, S.; Franklin, D.M.; Leddy, H.A.; Robey, P.G.; Storms, R.W.; Gimble, J.M. Surface protein characterization of human adipose tissue-derived stromal cells. J. Cell. Physiol. 2001, 189, 54–63. [Google Scholar] [CrossRef]
  151. Salgado, A.J.; Reis, R.L.; Sousa, N.J.; Gimble, J.M. Adipose tissue derived stem cells secretome: Soluble factors and their roles in regenerative medicine. Curr. Stem Cell Res. Ther. 2010, 5, 103–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Watanabe, Y.; Sasaki, R.; Matsumine, H.; Yamato, M.; Okano, T. Undifferentiated and differentiated adipose-derived stem cells improve nerve regeneration in a rat model of facial nerve defect. J. Tissue Eng. Regen. Med. 2017, 11, 362–374. [Google Scholar] [CrossRef]
  153. Liang, C.; Liang, Q.; Xu, X.; Liu, X.; Gao, X.; Li, M.; Yang, J.; Xing, X.; Huang, H.; Tang, Q.; et al. Bone morphogenetic protein 7 mediates stem cells migration and angiogenesis: Therapeutic potential for endogenous pulp regeneration. Int. J. Oral. Sci. 2022, 14, 38. [Google Scholar] [CrossRef] [PubMed]
  154. Yang, J.; Yuan, G.; Chen, Z. Pulp Regeneration: Current Approaches and Future Challenges. Front. Physiol. 2016, 7, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Levi-Montalcini, R.; Hamburger, V. Selective growth stimulating effects of mouse sarcoma on the sensory and sympathetic nervous system of the chick embryo. J. Exp. Zool. 1951, 116, 321–361. [Google Scholar] [CrossRef]
  156. Tomlinson, R.E.; Li, Z.; Li, Z.; Minichiello, L.; Riddle, R.C.; Venkatesan, A.; Clemens, T.L. NGF-TrkA signaling in sensory nerves is required for skeletal adaptation to mechanical loads in mice. Proc. Natl. Acad. Sci. USA 2017, 114, E3632–E3641. [Google Scholar] [CrossRef]
  157. Tsutsui, T.W. Dental Pulp Stem Cells: Advances to Applications. Stem Cells Cloning 2020, 13, 33–42. [Google Scholar] [CrossRef] [Green Version]
  158. Gage, F.H.; Batchelor, P.; Chen, K.S.; Chin, D.; Higgins, G.A.; Koh, S.; Deputy, S.; Rosenberg, M.B.; Fischer, W.; Bjorklund, A. NGF receptor reexpression and NGF-mediated cholinergic neuronal hypertrophy in the damaged adult neostriatum. Neuron 1989, 2, 1177–1184. [Google Scholar] [CrossRef]
  159. Liu, Z.; Wu, H.; Huang, S. Role of NGF and its receptors in wound healing (Review). Exp. Ther. Med. 2021, 21, 599. [Google Scholar] [CrossRef]
  160. Vega, J.A.; García-Suárez, O.; Hannestad, J.; Pérez-Pérez, M.; Germanà, A. Neurotrophins and the immune system. J. Anat. 2003, 203, 1–19. [Google Scholar] [CrossRef]
  161. Moattari, M.; Kouchesfehani, H.M.; Kaka, G.; Sadraie, S.H.; Naghdi, M. Evaluation of nerve growth factor (NGF) treated mesenchymal stem cells for recovery in neurotmesis model of peripheral nerve injury. J. Craniomaxillofac. Surg. 2018, 46, 898–904. [Google Scholar] [CrossRef] [PubMed]
  162. Ding, J.; Cheng, Y.; Gao, S.; Chen, J. Effects of nerve growth factor and Noggin-modified bone marrow stromal cells on stroke in rats. J. Neurosci. Res. 2011, 89, 222–230. [Google Scholar] [CrossRef] [PubMed]
  163. Chung, J.; Kubota, H.; Ozaki, Y.; Uda, S.; Kuroda, S. Timing-dependent actions of NGF required for cell differentiation. PLoS ONE 2010, 5, e9011. [Google Scholar] [CrossRef] [Green Version]
  164. Zhang, J.; Lu, X.; Feng, G.; Gu, Z.; Sun, Y.; Bao, G.; Xu, G.; Lu, Y.; Chen, J.; Xu, L.; et al. Chitosan scaffolds induce human dental pulp stem cells to neural differentiation: Potential roles for spinal cord injury therapy. Cell Tissue Res. 2016, 366, 129–142. [Google Scholar] [CrossRef] [PubMed]
  165. Mizuno, N.; Shiba, H.; Xu, W.P.; Inui, T.; Fujita, T.; Kajiya, M.; Takeda, K.; Hasegawa, N.; Kawaguchi, H.; Kurihara, H. Effect of neurotrophins on differentiation, calcification and proliferation in cultures of human pulp cells. Cell Biol. Int. 2007, 31, 1462–1469. [Google Scholar] [CrossRef] [PubMed]
  166. Mitsiadis, T.A.; Pagella, P. Expression of Nerve Growth Factor (NGF), TrkA, and p75(NTR) in Developing Human Fetal Teeth. Front. Physiol. 2016, 7, 338. [Google Scholar] [CrossRef]
  167. Mitsiadis, T.A.; Magloire, H.; Pagella, P. Nerve growth factor signalling in pathology and regeneration of human teeth. Sci. Rep. 2017, 7, 1327. [Google Scholar] [CrossRef] [Green Version]
  168. Liu, Q.; Lei, L.; Yu, T.; Jiang, T.; Kang, Y. Effect of Brain-Derived Neurotrophic Factor on the Neurogenesis and Osteogenesis in Bone Engineering. Tissue Eng. Part A 2018, 24, 1283–1292. [Google Scholar] [CrossRef]
  169. Blais, M.; Lévesque, P.; Bellenfant, S.; Berthod, F. Nerve Growth Factor, Brain-Derived Neurotrophic Factor, Neurotrophin-3 and Glial-Derived Neurotrophic Factor Enhance Angiogenesis in a Tissue-Engineered In Vitro Model. Tissue Eng. Part A 2013, 19, 1655–1664. [Google Scholar] [CrossRef] [Green Version]
  170. Cen, L.P.; Ng, T.K.; Liang, J.J.; Zhuang, X.; Yao, X.; Yam, G.H.; Chen, H.; Cheung, H.S.; Zhang, M.; Pang, C.P. Human Periodontal Ligament-Derived Stem Cells Promote Retinal Ganglion Cell Survival and Axon Regeneration After Optic Nerve Injury. Stem Cells 2018, 36, 844–855. [Google Scholar] [CrossRef] [Green Version]
  171. Nosrat, C.A.; Fried, K.; Ebendal, T.; Olson, L. NGF, BDNF, NT3, NT4 and GDNF in tooth development. Eur. J. Oral Sci. 1998, 106, 94–99. [Google Scholar] [CrossRef] [PubMed]
  172. Irfan, M.; Kim, J.H.; Druzinsky, R.E.; Ravindran, S.; Chung, S. Complement C5aR/LPS-induced BDNF and NGF modulation in human dental pulp stem cells. Sci. Rep. 2022, 12, 2042. [Google Scholar] [CrossRef] [PubMed]
  173. Xiao, N.; Thor, D.; Yu, W.Y. Neurotrophins BDNF and NT4/5 accelerate dental pulp stem cell migration. Biomed. J. 2021, 44, 363–368. [Google Scholar] [CrossRef] [PubMed]
  174. Luzuriaga, J.; Pineda, J.R.; Irastorza, I.; Uribe-Etxebarria, V.; García-Gallastegui, P.; Encinas, J.M.; Chamero, P.; Unda, F.; Ibarretxe, G. BDNF and NT3 Reprogram Human Ectomesenchymal Dental Pulp Stem Cells to Neurogenic and Gliogenic Neural Crest Progenitors Cultured in Serum-Free Medium. Cell. Physiol. Biochem. 2019, 52, 1361–1380. [Google Scholar] [CrossRef] [PubMed]
  175. Hohn, A.; Leibrock, J.; Bailey, K.; Barde, Y.A. Identification and characterization of a novel member of the nerve growth factor/brain-derived neurotrophic factor family. Nature 1990, 344, 339–341. [Google Scholar] [CrossRef]
  176. Tauszig-Delamasure, S.; Bouzas-Rodriguez, J. Targeting neurotrophin-3 and its dependence receptor tyrosine kinase receptor C: A new antitumoral strategy. Expert Opin. Targets 2011, 15, 847–858. [Google Scholar] [CrossRef] [PubMed]
  177. Lin, M.I.; Das, I.; Schwartz, G.M.; Tsoulfas, P.; Mikawa, T.; Hempstead, B.L. Trk C receptor signaling regulates cardiac myocyte proliferation during early heart development in vivo. Dev. Biol. 2000, 226, 180–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Cristofaro, B.; Stone, O.A.; Caporali, A.; Dawbarn, D.; Ieronimakis, N.; Reyes, M.; Madeddu, P.; Bates, D.O.; Emanueli, C. Neurotrophin-3 is a novel angiogenic factor capable of therapeutic neovascularization in a mouse model of limb ischemia. Arter. Thromb. Vasc. Biol. 2010, 30, 1143–1150. [Google Scholar] [CrossRef] [Green Version]
  179. Zhang, S.; Jin, H.; Yao, L.; Deng, F.; Shen, L. Neurotrophin-3 enhances the osteogenesis ability of human bone marrow mesenchymal stem cells stimulated by lipopolysaccharide. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2018, 34, 47–52. [Google Scholar]
  180. Ji, W.C.; Li, M.; Jiang, W.T.; Ma, X.; Li, J. Protective effect of brain-derived neurotrophic factor and neurotrophin-3 overexpression by adipose-derived stem cells combined with silk fibroin/chitosan scaffold in spinal cord injury. Neurol. Res. 2020, 42, 361–371. [Google Scholar] [CrossRef]
  181. Yan, Z.; Shi, X.; Wang, H.; Si, C.; Liu, Q.; Du, Y. Neurotrophin-3 Promotes the Neuronal Differentiation of BMSCs and Improves Cognitive Function in a Rat Model of Alzheimer’s Disease. Front. Cell. Neurosci. 2021, 15, 629356. [Google Scholar] [CrossRef] [PubMed]
  182. Wang, L.J.; Zhang, R.P.; Li, J.D. Transplantation of neurotrophin-3-expressing bone mesenchymal stem cells improves recovery in a rat model of spinal cord injury. Acta Neurochir. 2014, 156, 1409–1418. [Google Scholar] [CrossRef] [PubMed]
  183. Ji, W.; Zhang, X.; Ji, L.; Wang, K.; Qiu, Y. Effects of brain-derived neurotrophic factor and neurotrophin-3 on the neuronal differentiation of rat adipose-derived stem cells. Mol. Med. Rep. 2015, 12, 4981–4988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Armelin, H.A. Pituitary extracts and steroid hormones in the control of 3T3 cell growth. Proc. Natl. Acad. Sci. USA 1973, 70, 2702–2706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Ornitz, D.M.; Itoh, N. The Fibroblast Growth Factor signaling pathway. WIREs Dev. Biol. 2015, 4, 215–266. [Google Scholar] [CrossRef] [Green Version]
  186. Vaseenon, S.; Chattipakorn, N.; Chattipakorn, S.C. The possible role of basic fibroblast growth factor in dental pulp. Arch. Oral Biol. 2020, 109, 104574. [Google Scholar] [CrossRef]
  187. Xiao, L.; Sobue, T.; Esliger, A.; Kronenberg, M.S.; Coffin, J.D.; Doetschman, T.; Hurley, M.M. Disruption of the Fgf2 gene activates the adipogenic and suppresses the osteogenic program in mesenchymal marrow stromal stem cells. Bone 2010, 47, 360–370. [Google Scholar] [CrossRef]
  188. Qian, J.; Jiayuan, W.; Wenkai, J.; Peina, W.; Ansheng, Z.; Shukai, S.; Shafei, Z.; Jun, L.; Longxing, N. Basic fibroblastic growth factor affects the osteogenic differentiation of dental pulp stem cells in a treatment-dependent manner. Int. Endod. J. 2015, 48, 690–700. [Google Scholar] [CrossRef]
  189. Osathanon, T.; Nowwarote, N.; Pavasant, P. Basic fibroblast growth factor inhibits mineralization but induces neuronal differentiation by human dental pulp stem cells through a FGFR and PLCγ signaling pathway. J. Cell. Biochem. 2011, 112, 1807–1816. [Google Scholar] [CrossRef]
  190. Luo, L.; Albashari, A.A.; Wang, X.; Jin, L.; Zhang, Y.; Zheng, L.; Xia, J.; Xu, H.; Zhao, Y.; Xiao, J.; et al. Effects of Transplanted Heparin-Poloxamer Hydrogel Combining Dental Pulp Stem Cells and bFGF on Spinal Cord Injury Repair. Stem Cells Int. 2018, 2018, 2398521. [Google Scholar] [CrossRef] [Green Version]
  191. Liang, C.; Liao, L.; Tian, W. Stem Cell-based Dental Pulp Regeneration: Insights From Signaling Pathways. Stem Cell Rev. Rep. 2021, 17, 1251–1263. [Google Scholar] [CrossRef] [PubMed]
  192. Shimabukuro, Y.; Ueda, M.; Ozasa, M.; Anzai, J.; Takedachi, M.; Yanagita, M.; Ito, M.; Hashikawa, T.; Yamada, S.; Murakami, S. Fibroblast growth factor-2 regulates the cell function of human dental pulp cells. J. Endod. 2009, 35, 1529–1535. [Google Scholar] [CrossRef] [PubMed]
  193. Kitamura, C.; Nishihara, T.; Terashita, M.; Tabata, Y.; Washio, A. Local regeneration of dentin-pulp complex using controlled release of fgf-2 and naturally derived sponge-like scaffolds. Int. J. Dent. 2012, 2012, 190561. [Google Scholar] [CrossRef]
  194. Kim, J.; Park, J.C.; Kim, S.H.; Im, G.I.; Kim, B.S.; Lee, J.B.; Choi, E.Y.; Song, J.S.; Cho, K.S.; Kim, C.S. Treatment of FGF-2 on stem cells from inflamed dental pulp tissue from human deciduous teeth. Oral Dis. 2014, 20, 191–204. [Google Scholar] [CrossRef] [PubMed]
  195. Sagomonyants, K.; Kalajzic, I.; Maye, P.; Mina, M. Enhanced Dentinogenesis of Pulp Progenitors by Early Exposure to FGF2. J. Dent. Res. 2015, 94, 1582–1590. [Google Scholar] [CrossRef] [Green Version]
  196. Forbes, B.E.; Blyth, A.J.; Wit, J.M. Disorders of IGFs and IGF-1R signaling pathways. Mol. Cell. Endocrinol. 2020, 518, 111035. [Google Scholar] [CrossRef]
  197. Liu, D.; Wang, Y.; Jia, Z.; Wang, L.; Wang, J.; Yang, D.; Song, J.; Wang, S.; Fan, Z. Demethylation of IGFBP5 by Histone Demethylase KDM6B Promotes Mesenchymal Stem Cell-Mediated Periodontal Tissue Regeneration by Enhancing Osteogenic Differentiation and Anti-Inflammation Potentials. Stem Cells 2015, 33, 2523–2536. [Google Scholar] [CrossRef]
  198. Hao, J.; Yang, H.; Cao, Y.; Zhang, C.; Fan, Z. IGFBP5 enhances the dentinogenesis potential of dental pulp stem cells via JNK and ErK signalling pathways. J. Oral Rehabil. 2020, 47, 1557–1565. [Google Scholar] [CrossRef]
  199. Saito, K.; Ohshima, H. The putative role of insulin-like growth factor (IGF)-binding protein 5 independent of IGF in the maintenance of pulpal homeostasis in mice. Regen. Ther. 2019, 11, 217–224. [Google Scholar] [CrossRef]
  200. 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]
  201. Ahmadi, F.; Salmasi, Z.; Mojarad, M.; Eslahi, A.; Tayarani-Najaran, Z. G-CSF augments the neuroprotective effect of conditioned medium of dental pulp stem cells against hypoxic neural injury in SH-SY5Y cells. Iran. J. Basic Med. Sci. 2021, 24, 1743–1752. [Google Scholar] [CrossRef] [PubMed]
  202. Tsai, S.T.; Chu, S.C.; Liu, S.H.; Pang, C.Y.; Hou, T.W.; Lin, S.Z.; Chen, S.Y. Neuroprotection of Granulocyte Colony-Stimulating Factor for Early Stage Parkinson’s Disease. Cell Transpl. 2017, 26, 409–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Zhang, X.M.; Du, F.; Yang, D.; Wang, R.; Yu, C.J.; Huang, X.N.; Hu, H.Y.; Liu, W.; Fu, J. Granulocyte colony-stimulating factor increases the therapeutic efficacy of bone marrow mononuclear cell transplantation in cerebral ischemia in mice. BMC Neurosci. 2011, 12, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Schneider, A.; Krüger, C.; Steigleder, T.; Weber, D.; Pitzer, C.; Laage, R.; Aronowski, J.; Maurer, M.H.; Gassler, N.; Mier, W.; et al. The hematopoietic factor G-CSF is a neuronal ligand that counteracts programmed cell death and drives neurogenesis. J. Clin. Investig. 2005, 115, 2083–2098. [Google Scholar] [CrossRef] [Green Version]
  205. Iohara, K.; Fujita, M.; Ariji, Y.; Yoshikawa, M.; Watanabe, H.; Takashima, A.; Nakashima, M. Assessment of Pulp Regeneration Induced by Stem Cell Therapy by Magnetic Resonance Imaging. J. Endod. 2016, 42, 397–401. [Google Scholar] [CrossRef]
  206. Schmalz, G.; Widbiller, M.; Galler, K.M. Signaling Molecules and Pulp Regeneration. J. Endod. 2017, 43, S7–S11. [Google Scholar] [CrossRef]
  207. Takeuchi, N.; Hayashi, Y.; Murakami, M.; Alvarez, F.J.; Horibe, H.; Iohara, K.; Nakata, K.; Nakamura, H.; Nakashima, M. Similar in vitro effects and pulp regeneration in ectopic tooth transplantation by basic fibroblast growth factor and granulocyte-colony stimulating factor. Oral Dis. 2015, 21, 113–122. [Google Scholar] [CrossRef]
  208. Yu, J.; Liu, X.L.; Cheng, Q.G.; Lu, S.S.; Xu, X.Q.; Zu, Q.Q.; Liu, S. G-CSF and hypoxic conditioning improve the proliferation, neural differentiation and migration of canine bone marrow mesenchymal stem cells. Exp. Ther. Med. 2016, 12, 1822–1828. [Google Scholar] [CrossRef] [Green Version]
  209. Smojver, I.; Katalinić, I.; Bjelica, R.; Gabrić, D.; Matišić, V.; Molnar, V.; Primorac, D. Mesenchymal Stem Cells Based Treatment in Dental Medicine: A Narrative Review. Int. J. Mol. Sci. 2022, 23, 1662. [Google Scholar] [CrossRef]
  210. Moussa, D.G.; Aparicio, C. Present and future of tissue engineering scaffolds for dentin-pulp complex regeneration. J. Tissue Eng. Regen. Med. 2019, 13, 58–75. [Google Scholar] [CrossRef] [Green Version]
  211. Pinho, A.C.; Fonseca, A.C.; Serra, A.C.; Santos, J.D.; Coelho, J.F. Peripheral Nerve Regeneration: Current Status and New Strategies Using Polymeric Materials. Adv. Healthc Mater. 2016, 5, 2732–2744. [Google Scholar] [CrossRef] [Green Version]
  212. Ramachandran, N.; Singh, S.; Podar, R.; Kulkarni, G.; Shetty, R.; Chandrasekhar, P. A comparison of two pulp revascularization techniques using platelet-rich plasma and whole blood clot. J. Conserv. Dent. 2020, 23, 637–643. [Google Scholar] [CrossRef] [PubMed]
  213. Murray, P.E. Platelet-Rich Plasma and Platelet-Rich Fibrin Can Induce Apical Closure More Frequently Than Blood-Clot Revascularization for the Regeneration of Immature Permanent Teeth: A Meta-Analysis of Clinical Efficacy. Front. Bioeng. Biotechnol. 2018, 6, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Xu, J.; Gou, L.; Zhang, P.; Li, H.; Qiu, S. Platelet-rich plasma and regenerative dentistry. Aust. Dent. J. 2020, 65, 131–142. [Google Scholar] [CrossRef] [PubMed]
  215. Raddall, G.; Mello, I.; Leung, B.M. Biomaterials and Scaffold Design Strategies for Regenerative Endodontic Therapy. Front. Bioeng. Biotechnol. 2019, 7, 317. [Google Scholar] [CrossRef] [PubMed]
  216. Cen, L.; Liu, W.; Cui, L.; Zhang, W.; Cao, Y. Collagen tissue engineering: Development of novel biomaterials and applications. Pediatr. Res. 2008, 63, 492–496. [Google Scholar] [CrossRef]
  217. Zein, N.; Harmouch, E.; Lutz, J.C.; Fernandez De Grado, G.; Kuchler-Bopp, S.; Clauss, F.; Offner, D.; Hua, G.; Benkirane-Jessel, N.; Fioretti, F. Polymer-Based Instructive Scaffolds for Endodontic Regeneration. Materials 2019, 12, 2347. [Google Scholar] [CrossRef]
  218. Abbass, M.M.S.; El-Rashidy, A.A.; Sadek, K.M.; Moshy, S.E.; Radwan, I.A.; Rady, D.; Dörfer, C.E.; Fawzy El-Sayed, K.M. Hydrogels and Dentin-Pulp Complex Regeneration: From the Benchtop to Clinical Translation. Polymers 2020, 12, 2935. [Google Scholar] [CrossRef]
  219. Dayi, B.; Bilecen, D.S.; Eröksüz, H.; Yalcin, M.; Hasirci, V. Evaluation of a collagen-bioaggregate composite scaffold in the repair of sheep pulp tissue. Eur. Oral Res. 2021, 55, 152–161. [Google Scholar] [CrossRef]
  220. Nakashima, M.; Iohara, K. Regeneration of dental pulp by stem cells. Adv. Dent. Res. 2011, 23, 313–319. [Google Scholar] [CrossRef]
  221. Kwon, Y.S.; Lee, S.H.; Hwang, Y.C.; Rosa, V.; Lee, K.W.; Min, K.S. Behaviour of human dental pulp cells cultured in a collagen hydrogel scaffold cross-linked with cinnamaldehyde. Int. Endod. J. 2017, 50, 58–66. [Google Scholar] [CrossRef] [PubMed]
  222. Li, G.; Han, Q.; Lu, P.; Zhang, L.; Zhang, Y.; Chen, S.; Zhang, P.; Zhang, L.; Cui, W.; Wang, H.; et al. Construction of Dual-Biofunctionalized Chitosan/Collagen Scaffolds for Simultaneous Neovascularization and Nerve Regeneration. Research 2020, 2020, 2603048. [Google Scholar] [CrossRef] [PubMed]
  223. Ouasti, S.; Donno, R.; Cellesi, F.; Sherratt, M.J.; Terenghi, G.; Tirelli, N. Network connectivity, mechanical properties and cell adhesion for hyaluronic acid/PEG hydrogels. Biomaterials 2011, 32, 6456–6470. [Google Scholar] [CrossRef]
  224. Ahmadian, E.; Eftekhari, A.; Dizaj, S.M.; Sharifi, S.; Mokhtarpour, M.; Nasibova, A.N.; Khalilov, R.; Samiei, M. The effect of hyaluronic acid hydrogels on dental pulp stem cells behavior. Int. J. Biol. Macromol. 2019, 140, 245–254. [Google Scholar] [CrossRef] [PubMed]
  225. Turley, E.A.; Noble, P.W.; Bourguignon, L.Y. Signaling properties of hyaluronan receptors. J. Biol. Chem. 2002, 277, 4589–4592. [Google Scholar] [CrossRef] [Green Version]
  226. Almeida, L.D.F.; Babo, P.S.; Silva, C.R.; Rodrigues, M.T.; Hebling, J.; Reis, R.L.; Gomes, M.E. Hyaluronic acid hydrogels incorporating platelet lysate enhance human pulp cell proliferation and differentiation. J. Mater. Sci. Mater. Med. 2018, 29, 88. [Google Scholar] [CrossRef] [Green Version]
  227. Yang, J.; Hsu, C.C.; Cao, T.T.; Ye, H.; Chen, J.; Li, Y.Q. A hyaluronic acid granular hydrogel nerve guidance conduit promotes regeneration and functional recovery of injured sciatic nerves in rats. Neural. Regen. Res. 2023, 18, 657–663. [Google Scholar] [CrossRef]
  228. Issa, M.M.; Köping-Höggård, M.; Artursson, P. Chitosan and the mucosal delivery of biotechnology drugs. Drug Discov. Today Technol. 2005, 2, 1–6. [Google Scholar] [CrossRef]
  229. Chang, B.; Ahuja, N.; Ma, C.; Liu, X. Injectable scaffolds: Preparation and application in dental and craniofacial regeneration. Mater. Sci. Eng. R Rep. 2017, 111, 1–26. [Google Scholar] [CrossRef] [Green Version]
  230. El Ashiry, E.A.; Alamoudi, N.M.; El Ashiry, M.K.; Bastawy, H.A.; El Derwi, D.A.; Atta, H.M. Tissue Engineering of Necrotic Dental Pulp of Immature Teeth with Apical Periodontitis in Dogs: Radiographic and Histological Evaluation. J. Clin. Pediatr. Dent. 2018, 42, 373–382. [Google Scholar] [CrossRef]
  231. Feng, X.; Lu, X.; Huang, D.; Xing, J.; Feng, G.; Jin, G.; Yi, X.; Li, L.; Lu, Y.; Nie, D.; et al. 3D porous chitosan scaffolds suit survival and neural differentiation of dental pulp stem cells. Cell. Mol. Neurobiol. 2014, 34, 859–870. [Google Scholar] [CrossRef] [PubMed]
  232. Chávez-Delgado, M.E.; Gomez-Pinedo, U.; Feria-Velasco, A.; Huerta-Viera, M.; Castañeda, S.C.; Toral, F.A.; Parducz, A.; Anda, S.L.; Mora-Galindo, J.; García-Estrada, J. Ultrastructural analysis of guided nerve regeneration using progesterone- and pregnenolone-loaded chitosan prostheses. J. Biomed. Mater. Res. B Appl. Biomater. 2005, 74, 589–600. [Google Scholar] [CrossRef] [PubMed]
  233. Drury, J.L.; Mooney, D.J. Hydrogels for tissue engineering: Scaffold design variables and applications. Biomaterials 2003, 24, 4337–4351. [Google Scholar] [CrossRef] [PubMed]
  234. Li, X.; Liu, T.; Song, K.; Yao, L.; Ge, D.; Bao, C.; Ma, X.; Cui, Z. Culture of neural stem cells in calcium alginate beads. Biotechnol. Prog. 2006, 22, 1683–1689. [Google Scholar] [CrossRef]
  235. Dobie, K.; Smith, G.; Sloan, A.J.; Smith, A.J. Effects of alginate hydrogels and TGF-beta 1 on human dental pulp repair in vitro. Connect. Tissue Res. 2002, 43, 387–390. [Google Scholar] [CrossRef]
  236. Poongodi, R.; Chen, Y.L.; Yang, T.H.; Huang, Y.H.; Yang, K.D.; Lin, H.C.; Cheng, J.K. Bio-Scaffolds as Cell or Exosome Carriers for Nerve Injury Repair. Int. J. Mol. Sci. 2021, 22, 13347. [Google Scholar] [CrossRef]
  237. Benton, G.; Arnaoutova, I.; George, J.; Kleinman, H.K.; Koblinski, J. Matrigel: From discovery and ECM mimicry to assays and models for cancer research. Adv. Drug Deliv. Rev. 2014, 79–80, 3–18. [Google Scholar] [CrossRef]
  238. Wang, J.; Chu, R.; Ni, N.; Nan, G. The effect of Matrigel as scaffold material for neural stem cell transplantation for treating spinal cord injury. Sci. Rep. 2020, 10, 2576. [Google Scholar] [CrossRef] [Green Version]
  239. 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]
  240. Jeong, S.Y.; Lee, S.; Choi, W.H.; Jee, J.H.; Kim, H.R.; Yoo, J. Fabrication of Dentin-Pulp-Like Organoids Using Dental-Pulp Stem Cells. Cells 2020, 9, 642. [Google Scholar] [CrossRef] [Green Version]
  241. Absalan, F.; Pasandi, M.S.; Ghasemi Hamidabadi, H.; Saeednia, S.; Bojnordi, M.N.; Zahiri, M.; Alizadeh, R.; Bagher, Z. Matrigel enhances differentiation of human adipose tissue-derived stem cells into dopaminergic neuron. Neurosci. Lett. 2021, 760, 136070. [Google Scholar] [CrossRef] [PubMed]
  242. Coulombe, P.A.; Bousquet, O.; Ma, L.; Yamada, S.; Wirtz, D. The ‘ins’ and ‘outs’ of intermediate filament organization. Trends Cell Biol. 2000, 10, 420–428. [Google Scholar] [CrossRef] [PubMed]
  243. Gao, J.; Zhang, L.; Wei, Y.; Chen, T.; Ji, X.; Ye, K.; Yu, J.; Tang, B.; Sun, X.; Hu, J. Human hair keratins promote the regeneration of peripheral nerves in a rat sciatic nerve crush model. J. Mater. Sci. Mater. Med. 2019, 30, 82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Park, J.Y.; Yang, C.; Jung, I.H.; Lim, H.C.; Lee, J.S.; Jung, U.W.; Seo, Y.K.; Park, J.K.; Choi, S.H. Regeneration of rabbit calvarial defects using cells-implanted nano-hydroxyapatite coated silk scaffolds. Biomater. Res. 2015, 19, 7. [Google Scholar] [CrossRef] [Green Version]
  245. Chen, S.; Liu, S.; Zhang, L.; Han, Q.; Liu, H.; Shen, J.; Li, G.; Zhang, L.; Yang, Y. Construction of injectable silk fibroin/polydopamine hydrogel for treatment of spinal cord injury. Chem. Eng. J. 2020, 399, 125795. [Google Scholar] [CrossRef]
  246. Vijayavenkataraman, S. Nerve guide conduits for peripheral nerve injury repair: A review on design, materials and fabrication methods. Acta Biomater. 2020, 106, 54–69. [Google Scholar] [CrossRef]
  247. Liu, X.; Ma, P.X. Polymeric scaffolds for bone tissue engineering. Ann. Biomed. Eng. 2004, 32, 477–486. [Google Scholar] [CrossRef] [Green Version]
  248. Fan, Y.; Bi, R.; Densmore, M.J.; Sato, T.; Kobayashi, T.; Yuan, Q.; Zhou, X.; Erben, R.G.; Lanske, B. Parathyroid hormone 1 receptor is essential to induce FGF23 production and maintain systemic mineral ion homeostasis. FASEB J. 2016, 30, 428–440. [Google Scholar] [CrossRef] [Green Version]
  249. Fan, Y.; Liu, W.; Bi, R.; Densmore, M.J.; Sato, T.; Mannstadt, M.; Yuan, Q.; Zhou, X.; Olauson, H.; Larsson, T.E.; et al. Interrelated role of Klotho and calcium-sensing receptor in parathyroid hormone synthesis and parathyroid hyperplasia. Proc. Natl. Acad. Sci. USA 2018, 115, E3749–E3758. [Google Scholar] [CrossRef] [Green Version]
  250. Fan, Y.; Cui, C.; Rosen, C.J.; Sato, T.; Xu, R.; Li, P.; Wei, X.; Bi, R.; Yuan, Q.; Zhou, C. Klotho in Osx(+)-mesenchymal progenitors exerts pro-osteogenic and anti-inflammatory effects during mandibular alveolar bone formation and repair. Signal Transduct. Target. Ther. 2022, 7, 155. [Google Scholar] [CrossRef]
  251. Fan, Y.; Cui, C.; Li, P.; Bi, R.; Lyu, P.; Li, Y.; Zhu, S. Fibrocartilage Stem Cells in the Temporomandibular Joint: Insights From Animal and Human Studies. Front. Cell Dev. Biol. 2021, 9, 665995. [Google Scholar] [CrossRef] [PubMed]
  252. Brockes, J.P. Mitogenic growth factors and nerve dependence of limb regeneration. Science 1984, 225, 1280–1287. [Google Scholar] [CrossRef]
  253. Rinkevich, Y.; Montoro, D.T.; Muhonen, E.; Walmsley, G.G.; Lo, D.; Hasegawa, M.; Januszyk, M.; Connolly, A.J.; Weissman, I.L.; Longaker, M.T. Clonal analysis reveals nerve-dependent and independent roles on mammalian hind limb tissue maintenance and regeneration. Proc. Natl. Acad. Sci. USA 2014, 111, 9846–9851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Jones, R.E.; Salhotra, A.; Robertson, K.S.; Ransom, R.C.; Foster, D.S.; Shah, H.N.; Quarto, N.; Wan, D.C.; Longaker, M.T. Skeletal Stem Cell-Schwann Cell Circuitry in Mandibular Repair. Cell Rep. 2019, 28, 2757–2766.e2755. [Google Scholar] [CrossRef]
  255. Heine, W.; Conant, K.; Griffin, J.W.; Höke, A. Transplanted neural stem cells promote axonal regeneration through chronically denervated peripheral nerves. Exp. Neurol. 2004, 189, 231–240. [Google Scholar] [CrossRef] [PubMed]
  256. Godinho, M.J.; Staal, J.L.; Krishnan, V.S.; Hodgetts, S.I.; Pollett, M.A.; Goodman, D.P.; Teh, L.; Verhaagen, J.; Plant, G.W.; Harvey, A.R. Regeneration of adult rat sensory and motor neuron axons through chimeric peroneal nerve grafts containing donor Schwann cells engineered to express different neurotrophic factors. Exp. Neurol. 2020, 330, 113355. [Google Scholar] [CrossRef]
  257. Frostick, S.P.; Yin, Q.; Kemp, G.J. Schwann cells, neurotrophic factors, and peripheral nerve regeneration. Microsurgery 1998, 18, 397–405. [Google Scholar] [CrossRef]
  258. Kajiya, M.; Shiba, H.; Fujita, T.; Ouhara, K.; Takeda, K.; Mizuno, N.; Kawaguchi, H.; Kitagawa, M.; Takata, T.; Tsuji, K.; et al. Brain-derived neurotrophic factor stimulates bone/cementum-related protein gene expression in cementoblasts. J. Biol. Chem. 2008, 283, 16259–16267. [Google Scholar] [CrossRef] [Green Version]
  259. Wang, L.; Zhou, S.; Liu, B.; Lei, D.; Zhao, Y.; Lu, C.; Tan, A. Locally applied nerve growth factor enhances bone consolidation in a rabbit model of mandibular distraction osteogenesis. J. Orthop. Res. 2006, 24, 2238–2245. [Google Scholar] [CrossRef]
  260. Grills, B.L.; Schuijers, J.A.; Ward, A.R. Topical application of nerve growth factor improves fracture healing in rats. J. Orthop. Res. 1997, 15, 235–242. [Google Scholar] [CrossRef]
  261. Sun, S.; Diggins, N.H.; Gunderson, Z.J.; Fehrenbacher, J.C.; White, F.A.; Kacena, M.A. No pain, no gain? The effects of pain-promoting neuropeptides and neurotrophins on fracture healing. Bone 2020, 131, 115109. [Google Scholar] [CrossRef] [PubMed]
  262. Shen, L.; Zeng, W.; Wu, Y.X.; Hou, C.L.; Chen, W.; Yang, M.C.; Li, L.; Zhang, Y.F.; Zhu, C.H. Neurotrophin-3 accelerates wound healing in diabetic mice by promoting a paracrine response in mesenchymal stem cells. Cell Transpl. 2013, 22, 1011–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Su, Y.W.; Chim, S.M.; Zhou, L.; Hassanshahi, M.; Chung, R.; Fan, C.; Song, Y.; Foster, B.K.; Prestidge, C.A.; Peymanfar, Y.; et al. Osteoblast derived-neurotrophin-3 induces cartilage removal proteases and osteoclast-mediated function at injured growth plate in rats. Bone 2018, 116, 232–247. [Google Scholar] [CrossRef] [PubMed]
  264. Bucan, V.; Vaslaitis, D.; Peck, C.T.; Strauß, S.; Vogt, P.M.; Radtke, C. Effect of Exosomes from Rat Adipose-Derived Mesenchymal Stem Cells on Neurite Outgrowth and Sciatic Nerve Regeneration After Crush Injury. Mol. Neurobiol. 2019, 56, 1812–1824. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Nerve function in teeth. Nerves participate in sensation, inflammation, angiogenesis, and immunity in the tooth.
Figure 1. Nerve function in teeth. Nerves participate in sensation, inflammation, angiogenesis, and immunity in the tooth.
Ijms 23 15492 g001
Figure 2. Stem cells, growth factors, and scaffolds in neural regeneration in RET. SHED: stem cell from human exfoliated deciduous teeth; DPSC: dental pulp stem cell; SCAP: apical papilla stem cell; PDLSC: periodontal ligament stem cell; BMMSC: bone marrow mesenchymal stem cell; ADSC: adipose-derived stem cell; NGF: nerve growth factor; BDNF: brain-derived neurotrophic factor; NT-3: neurotrophin-3; bFGF: basic fibroblast growth factor; IGFBP5: insulin-like growth factor-binding protein 5; G-CSF: granulocyte colony-stimulating factor.
Figure 2. Stem cells, growth factors, and scaffolds in neural regeneration in RET. SHED: stem cell from human exfoliated deciduous teeth; DPSC: dental pulp stem cell; SCAP: apical papilla stem cell; PDLSC: periodontal ligament stem cell; BMMSC: bone marrow mesenchymal stem cell; ADSC: adipose-derived stem cell; NGF: nerve growth factor; BDNF: brain-derived neurotrophic factor; NT-3: neurotrophin-3; bFGF: basic fibroblast growth factor; IGFBP5: insulin-like growth factor-binding protein 5; G-CSF: granulocyte colony-stimulating factor.
Ijms 23 15492 g002
Table 1. Summary of the characteristics of neural regeneration in vivo, including animal and clinical studies of cell homing and stem cell implantation.
Table 1. Summary of the characteristics of neural regeneration in vivo, including animal and clinical studies of cell homing and stem cell implantation.
ScaffoldStem CellsSignal Molecules Species Outcome Reference
(1) Periapical bleeding
(2) PRF
(3) Collagen
(4) Hydroxyapatite
//HumanRadiographically: all succeeded; success rate of cold test: (1) periapical bleeding: 11%; (2) PRF:66%; (3) collagen: 44.4%; (4) hydroxyapatite: 33.3%Mittal et al., 2021 [29]
/Human SHED aggregates/PigDentin-pulp-like tissue with blood vessels and nervesGuo et al., 2020 [30]
AtelocollagenDPSCG-CSFDogDentin-pulp-like tissue with blood vessels and nervesIohara et al., 2020 [31]
Human treated dentin matrix, and Matrigel Human DPSC/MiceDentin-pulp-like tissue with blood vessels and nervesMeng et al., 2020 [32]
AtelocollagenDPSCG-CSFDogDentin-pulp-like tissue with blood vessels and nervesIohara et al., 2018 [33]
/SHED aggregate/Pig;
Human
Pig: Dentin-pulp-like tissue with blood vessels and nerves;
Human: Pulp-like tissue containing an odontoblast layer, connective tissue, blood vessels, and nerves
Xuan et al., 2018 [34]
Collagen gelBMMSCPDGF-BB
NGF
BDNF
RatWell-vascularized pulp-like tissue; positive signals for S-100Li and Wang, 2016 [35]
AtelocollagenDPSC
BMMSC
ADSC
G-CSFDog Dentin-pulp-like tissue with blood vessels and nervesMurakami et al., 2015 [36]
AtelocollagenDPSCG-CSFDogDentin-pulp-like tissue with blood vessels and nervesIohara et al., 2014 [37]
Polymerizing type I collagen hydrogelIndium-111-oxine -labeled rat pulp cells/RatDentin-pulp-like tissue with blood vessels, nerves, and active fibroblastic cellsSouron et al., 2014 [38]
AtelocollagenDPSCG-CSFDogDentin-pulp-like tissue with blood vessels and nervesIohara et al., 2013 [39]
Collagen TECD31-side population cells from pulp, bone marrow, adiposeSDF-1Dog Dentin-pulp-like tissue with blood vessels and nervesIshizaka et al., 2012 [40]
Collagen TE CD105+cells form pulp or adipose SDF-1DogDentin-pulp-like tissue with blood vessels and nervesIohara et al., 2011 [41]
Collagen /bFGF
VEGF/PDGF
NGF
BMP7
Teeth from humans implanted in miceDentin-pulp-like tissue with blood vessels and nervesKim et al., 2010 [10]
Abbreviations: SDF-1: stromal-cell-derived factor-1; PDGF: platelet-derived growth factor; G-CSF: granulocyte colony-stimulating factor; bFGF: basic fibroblast growth factor; VEGF: vascular endothelial growth factor; BMP7: bone morphogenetic protein 7; BMMSC: bone marrow mesenchymal stem cell; ADSC: adipose-derived stem cell.
Table 2. Summary of the characteristics of neural regeneration in clinical revascularization.
Table 2. Summary of the characteristics of neural regeneration in clinical revascularization.
Revascularization TreatmentCharacteristics of SamplesRadiological or Pathological ResultsEvidence of Successful Neural RegenerationReference
BCImmature permanent teeth of 40 patientsIncreased root length and thickness The EPT on the sixth week demonstrated a positive responseSajjad et al., 2022 [42]
BC
PRF
Mature anterior teeth of 20 patients Periradicular healing in both groupsDifferent readings of tooth sensitivity between preoperative, 6 months, and 12 monthsYoussef et al., 2022 [43]
BC23 immature necrotic permanent teethIncreased root length and thickness 53.8% and 50% responded to EPT after 3 and 8 years, respectivelyAbu Zeid et al., 2021 [44]
Plasma-derived biomaterial with human umbilical cord MSCs Mature anterior teeth of 18 patientsAfter 12 months, the treatment showed 100% clinical efficacyIncreased positive pulp response at 12-month follow-upBrizuela et al., 2020 [45]
BC51 immature permanent teeth with pulp necrosis91.4% showed root development.54% responded positively to cold or EPT, and 30% responded to both testsChrepa et al., 2020 [46]
BC16 traumatized permanent incisors with open apices56.3% root resorptions, 31.3% ankylosis, and 92.9% discolorations81.3% of teeth regained sensitivityMittmann et al., 2020 [47]
BC
PRF
The upper front teeth of an 11-year-old childIncreased root length and thickness; closure of apexAfter 12 months, both teeth were positive to the cold test and EPTNagaveni et al., 2020) [48]
BCTraumatized upper immature anterior teeth of 15 childrenPeriapical healing, root walls thickened, and apical closure closedOne tooth was positive to the cold test; four teeth were positive to EPTNazzal et al., 2020 [49]
BC28 mature teeth92.3% favorable clinical and radiographic outcomesHalf of the teeth responded to the EPTArslan et al., 2019 [50]
MSCs
PRF
Tooth #28 of a 50-year-old manNormal under percussion and palpationDelayed response to cold test; positive to EPTMeza et al., 2019 [51]
BC
PRP
PRF
PP
88 immature incisors of 67 children86 teeth showed periapical healing and root development86 teeth were positive to sensitivity testsUlusoy et al., 2019 [52]
BC
PRF
Five patients per groupIncreased root length and thickness; closure of apexPositive to sensitivity test between 6 and 9 months of follow-upLv et al., 2018 [53]
BCAn immature permanent toothIncreased root length and thickness, and complete osseous healingPositive to cold testMustafa, 2018 [54]
PRF15 patients with mature necrotic pulpComplete healing of periapical lesionDifferent readings of tooth sensitivity between preoperative and 12 monthsNageh et al., 2018 [55]
BCImmature maxillary incisors of 15 childrenApical foramen narrowedOne-third of the teeth were responsive to EPTNazzal et al., 2018 [56]
BC15 patients with immature and mature permanent teeth10/13 root development and apical closure 2/13 were positive to EPTNeelamurthy et al., 2018 [57]
PRP
BC
30 non-vital immature permanent teethLesion size decreased; bone density increased; continued root development After 5 months, sensitivity tests elicited a delayed positive response in 23 sitesAlagl et al., 2017 [58]
Experimental: BC + Bio-Gide collagen membrane
Control: BC
46 non-vital immature teethThickening of the dentin wall EPT was achieved in 18% of the control group and 33% of the experimental groupJiang et al., 2017 [59]
BC20 teeth with dens evaginatus treatedApical diameter and root length increasedFive teeth responded to the pulp sensitivity test during the 1-year follow-up periodLi et al., 2017 [60]
PRF
PRP
BC
60 patients with immature permanent teethIncreased root length and thickness; healing of periapical wound15% in PRF, 13.30% in BC, and 15.8% in PRP were positive to the vitality testShivashankar et al., 2017 [61]
BC4 infected necrotic primary second molarsAt six months, complete periradicular healing and remained symptomlessPositive to cold testUlusoy and Cehreli, 2017 [62]
BCMaxillary left central incisor of an 8-year-old childIncreased root length and thickness; healing of periapical wound; closure of apexResponded to cold and EPT as normal teethFarhad et al., 2016 [63]
BC#45 of a 10-year-old girlPulp-like tissue with blood vesselsThe neurovascular bundles in the pulp-like tissue were NF (+)Meschi et al., 2016 [24]
PRFImmature permanent tooth of an 11-year-old boyIncreased root length and thickness; healing of periapical wound; closure of apexPositive to cold test and EPT similar to adjacent teeth after 3 monthsNagaveni et al., 2016 [64]
PRPAn avulsed mature incisor of an 11-year-old boyResolution of periapical radiolucencyPositive to thermal test and EPTPriya et al., 2016 [65]
PRFA traumatized, necrotic, immature tooth Increase in root length and an intact lamina duraRemained negative to cold test but positive to EPT at 24 and 36 months Ray et al., 2016 [66]
PRFLeft posterior tooth of a 13-year-old maleIncreased root length and thickness; resolution of periapical wound; closure of apexPositive to cold test and EPTSubash et al., 2016 [67]
PRP
BC
20 immature teeth Complete apical closure and increased root length 5 teeth in PRP and 2 teeth in BC were positive to vitality testBezgin et al., 2015 [68]
BC4 mature teethRoot formation increased and dentin wall thickened3/4 teeth responded positively to sensitivity test.Dudeja et al., 2015 [69]
BCAn immature permanent toothIncreased root length and thickness; closure of apexNeurons and nerve fibers were observedLei et al., 2015 [70]
PRFAn immature permanent toothIncreased root length and thickness; closure of apexPositive to cold test and EPTNagaveni et al., 2015 [71]
PRPMaxillary left lateral incisor of a 16-year-old maleIncreased root length and thickness; resolution of periapical wound; closure of apexNot response to cold tests; delayed positive response to EPTSachdeva et al., 2015 [72]
BC3 premolars and 13 incisorsIncomplete apical closure: 47.2%; complete apical closure: 19.4%. Change in root length varying from −2.7% to 25.3%, and change in root dentin thickness ranging from −1.9% to 72.6%5/16 teeth were positive to EPT at 18-month recallKahler et al., 2014 [73]
BC3 immature permanent central incisorsComplete root developmentPositive to pulp testFarsi et al., 2013 [74]
PRFImmature right maxillary central incisor of a childIncreased root length and thickness; closure of apexPositive to cold test and EPTKeswani and Pandey, 2013 [75]
PRFTooth #21 of an 11-year-old childResolution of periapical rarefaction, further root development, and apical closure of the toothPositive to EPT and cold testMishra et al., 2013 [76]
BCCentral incisors of an 8.5-year-old boyIncreased root length and thickness; closure of apexPositive to cold tests at 12 months and EPT at 18 monthsCehreli et al., 2012 [77]
BCAn immature permanent incisorRoot development and apical closure Positive to sensitivity tests at 1–3-monthsMiller et al., 2012) [78]
PRFImmature incisor of a 9-year-old childIncreased root length and thickness; resolution of periapical wound; closure of apexPositive to cold test and EPTShivashankar et al., 2012 [79]
BC6 immature permanent molars of childrenIncreased root length and thickness; resolution of periapical wound; closure of apexTwo teeth were positive to cold testCehreli et al., 2011 [80]
BCAn immature permanent incisor of a childRoot development and apical closurePositive to EPTIwaya et al., 2011 [81]
PRPMaxillary premolar tooth of a childIncreased root length and thickness; resolution of periapical wound; closure of apexPositive to cold test and EPTTorabinejad and Turman, 2011 [82]
BC6 immature teeth with apical periodontitisResolution of periapical lesions; continued root developmentTwo teeth positive to vitality testPetrino et al., 2010 [83]
BCA mandibular premolarIncreased root length and thickness; resolution of periapical wound; closure of apexPositive to sensibility testThomson and Kahler, 2010 [84]
BCImmature permanent teeth of 12 patientsIncreased root thickness; closure of apex 3 teeth positive to pulp testDing et al., 2009 [85]
BC2 bilateral mandibular premolars with dens evaginatus of an 11-year-old girlIncreased root length and thickness; resolution of periapical wound; closure of apexPositive to cold testReynolds et al., 2009 [86]
BCLower-right second premolar of a childIncreased root thickness; closure of apex Positive to cold testBanchs and Trope, 2004 [87]
BCAn immature premolar of a 13-year-old patientIncreased root thickness; closure of apex Positive to EPTIwaya et al., 2001 [88]
Abbreviation: EPT: electric pulp test; BC: blood clot; PRF: platelet-rich fibrin; PP: platelet pellet; PRP: platelet-rich plasma.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wei, Y.; Lyu, P.; Bi, R.; Chen, X.; Yu, Y.; Li, Z.; Fan, Y. Neural Regeneration in Regenerative Endodontic Treatment: An Overview and Current Trends. Int. J. Mol. Sci. 2022, 23, 15492. https://doi.org/10.3390/ijms232415492

AMA Style

Wei Y, Lyu P, Bi R, Chen X, Yu Y, Li Z, Fan Y. Neural Regeneration in Regenerative Endodontic Treatment: An Overview and Current Trends. International Journal of Molecular Sciences. 2022; 23(24):15492. https://doi.org/10.3390/ijms232415492

Chicago/Turabian Style

Wei, Yali, Ping Lyu, Ruiye Bi, Xinyu Chen, Yanshen Yu, Zucen Li, and Yi Fan. 2022. "Neural Regeneration in Regenerative Endodontic Treatment: An Overview and Current Trends" International Journal of Molecular Sciences 23, no. 24: 15492. https://doi.org/10.3390/ijms232415492

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