Skip to main content
Bioactive Materials logoLink to Bioactive Materials
. 2023 May 14;27:574–593. doi: 10.1016/j.bioactmat.2023.04.013

Epigenetic therapeutics in dental pulp treatment: Hopes, challenges and concerns for the development of next-generation biomaterials

Henry F Duncan a,, Yoshifumi Kobayashi b, Michaela Kearney a, Emi Shimizu b
PMCID: PMC10199232  PMID: 37213443

Abstract

This opinion-led review paper highlights the need for novel translational research in vital-pulp-treatment (VPT), but also discusses the challenges in translating evidence to clinics. Traditional dentistry is expensive, invasive and relies on an outmoded mechanical understanding of dental disease, rather than employing a biological perspective that harnesses cell activity and the regenerative-capacity. Recent research has focussed on developing minimally-invasive biologically-based ‘fillings’ that preserve the dental pulp; research that is shifting the paradigm from expensive high-technology dentistry, with high failure rates, to smart restorations targeted at biological processes. Current VPTs promote repair by recruiting odontoblast-like cells in a material-dependent process. Therefore, exciting opportunities exist for development of next-generation biomaterials targeted at regenerative processes in the dentin-pulp complex. This article analyses recent research using pharmacological-inhibitors to therapeutically-target histone-deacetylase (HDAC) enzymes in dental-pulp-cells (DPCs) that stimulate pro-regenerative effects with limited loss of viability. Consequently, HDAC-inhibitors have the potential to enhance biomaterial-driven tissue responses at low concentration by influencing the cellular processes with minimal side-effects, providing an opportunity to develop a topically-placed, inexpensive bio-inductive pulp-capping material. Despite positive results, clinical translation of these innovations requires enterprise to counteract regulatory obstacles, dental-industry priorities and to develop strong academic/industry partnerships. The aim of this opinion-led review paper is to discuss the potential role of therapeutically-targeting epigenetic modifications as part of a topical VPT strategy in the treatment of the damaged dental pulp, while considering the next steps, material considerations, challenges and future for the clinical development of epigenetic therapeutics or other ‘smart’ restorations in VPT.

Keywords: Epigenetics, Vital pulp treatment, Histone deacetylase inhibitors, Regenerative endodontics, Pulp capping

Highlights

  • Illustrates the need for next-generation targeted vital pulp biomaterials to improve tissue response and treatment outcome.

  • Provides evidence supporting the use of HDACi as a bioactive component to pulp capping materials to stimulate repair..

  • Discusses the implications of recent legislation on the development of ‘smart’ dental restorations that contact pulp tissue.

1. Introduction

This opinion-led review paper uses epigenetic-therapeutic dental materials as an example, to illustrate the need to develop new pulp capping materials for conservative management of the exposed pulp in vital pulp treatment (VPT), while also illustrating the obstacles to clinical translation in this area. These pulp capping materials are applied to the dental pulp, which is a dynamic connective tissue encased in health by mineralized dentin and enamel. The secretory cells of the pulp, the odontoblasts, form a peripheral layer in contact with dentin and are interconnected in what is known as the dentin-pulp complex [1]. The pulp and specifically the odontoblasts have both a formative role in tooth development (primary dentinogenesis) and throughout life (secondary dentinogenesis), as well as a protective role by acting as a biosensor and secreting new tertiary dentin after challenge by caries, trauma and microbial leakage [2,3]. The pulp is generally threatened by microbes (e.g., dental caries) and if this infection is not managed it can overwhelm the pulp, leading to a progressive pulpitis, odontoblast death and eventually pulp necrosis [4,5]. However, if the inflammation can be controlled by removing the injurious threat and placing a sealing restoration, the pulp can recover, and dentin can be repaired and regenerated [6]. The prevention and control of pulpitis and apical periodontitis forms the basis of operative dentistry; however, the difficulties of predictably treating deep caries has been highlighted in a recent international position statement [7]. The European Society of Endodontology statement [7] concluded that ‘the control of pulpitis and conservative management of the exposed pulp should be a future focus of research activity’, in order to reduce the provision of root canal treatment (RCT) and promote biomimetic solutions [7,8]. Although RCT preserves the tooth, complete removal of the irreversibly damaged pulp tissue is a destructive, technically-difficult process [7,9], which removes any inherent attributes of the pulp including perception, immunity/repair and leads to greater risk of fracture and tooth loss [10, 11]. As a result, there is an urgent need to develop our understanding of the ‘key’ mediators orchestrating pulpal repair processes and tertiary dentin formation in order to develop more predictable ‘smarter’ VPTs and immunotherapies, with the ultimate goal of creating new biologically-based dental strategies aimed at creating novel regenerative techniques that therapeutically target tertiary dentin repair and regeneration [[12], [13], [14]].

Currently, after pulp exposure, the material of choice is a hydraulic calcium silicate cement (HCSC) applied directly to the pulpal wound, which has been shown since its introduction in the early 1990s, in experimental pulp capping studies to promote the formation of thicker, more homogenous reparative tertiary dentin formation than traditional calcium hydroxide materials [6,15]. Furthermore, these histological results have translated into better clinical outcomes for management of the cariously-exposed pulp [[16], [17], [18]]. Notably, however, the reparative dentin bridge formed after exposure and primary odontoblast death rarely exhibits tubular structure and may be considered reparative rather than regenerative in nature [2,19,20]. Mechanistically, HCSC's reparative action has been attributed to a range of sources including, stimulating the release of bioactive dentin matrix components [21], high pH, increasing migration, differentiation and biomineralization of dental pulp cells (DPCs) [22,23]; however, the exact mechanism of action is unknown and not targeted to a specific pathway or biological process.

Recently, in an attempt to improve the induction and differentiation of odontoblast-like cells, several attempts to stimulate regenerative dentinogenesis using pharmacological inhibitors or activators, often already European Union (EU) or Food and Drug Administration (FDA)-approved as drugs for another indication (e.g., cancer, Alzheimer's’ disease) have been reported [24]. Proposed therapeutic strategies have included, targeting Wnt signaling with GSK-3 inhibitors such as Tideglusib [25], using the anti-cholesterol drug Simvastatin [26] or targeting acetylation of DNA-associated proteins by our group with histone deacetylase inhibitors (HDACi) [[27], [28], [29]]. These drugs have shown considerable promise in improving mineralization and regenerative processes in comparison to existing treatments in vitro and in vivo; however, translating these novel next-generation treatments to the dental clinic is challenging with obstacles, related to regulation, funding and the development of industrial partnerships needing to be addressed.

As a result, the aim of this opinion-led review paper is two-fold. Firstly, the progress in terms of epigenetic therapeutics to improve DPC mineralization processes will be highlighted, prior to secondly the obstacles, barriers and problems with the incorporation of medicinal products and the development of next-generation smart dental materials being discussed with additional reference to opinion.

2. Why do we need next-generation bioactive pulp capping materials?

2.1. To improve pulpitis management

Preservation of healthy dental pulp tissue and the subsequent prevention of apical disease, form the biological basis of operative dental care and the core of preventative endodontics. As described previously, in health, the dental pulp is naturally protected by a mineralized outer shell of enamel and dentin, while as a connective tissue it also possesses a series of defense strategies including inflammation and mineral secretion designed to protect itself against injury [2]. The pulp is principally irritated by bacteria in the form of dental caries or microleakage around restorations, but can also be challenged trauma, dental materials and as a result of restorative procedures, all of which provoke inflammatory responses in the pulp, the extent of which reflects the severity of the challenge [5,9,30]. Microbial infection by ‘leakage’ around restorations provides a considerable challenge particularly when the restoration is deep, as bacterial products diffusing through the dentinal tubules induce inflammation even when the restoration has not yet reached the pulp [31], with histological studies showing that this process intensifies when the cavity encroaches to within 0.5 mm of the pulp [4,32]. The secretory odontoblast initially reacts immunologically to the microbes by initial pathogen recognition, but other pulp cells including fibroblasts, dental pulp stem cells (DPSCs) and immune cells also contribute to the defensive response; thereafter, a complex series of antibacterial, immune and inflammatory responses is orchestrated [33,34]. If the bacterial stimulation is not managed (e.g., by placement of a new ‘sealing’ biologically-based material), the microbial biofilm will advance and the associated bacteria will invade the tissue; this aggressive bacterial challenge invariably leads to irreversible pulpits, pulp necrosis and subsequent apical periodontitis [35]. Over time the bacterial flora in the diseased pulp and the subsequent necrotic root canal system changes from comprising principally facultative anaerobic bacteria to more gram-negative, obligate anaerobic bacteria [36,37]. Pulp necrosis will necessitate remedial dental treatment, such as tooth extraction or RCT; however, removal of the bacteria and placement of a bioactive dental material can promote pulp repair and maintain vitality [5]. The choice of dental material has been shown for several years to be an important factor that determines the quality of the dental pulp defensive response [6,38,39]; however, it is accepted that no existing material is ideal [2]. Although a role for anti-inflammatory [40] or immunotherapeutic materials [41] in the treatment of pulpitis have been proposed, currently no pulp capping materials specifically target inflammatory processes except for medicaments such as Ledermix (Henry Schein, New York, USA) which contain steroids and have been used only historically for this purpose [42].

Opinion The need for improved dental restorative solutions for the management of deep caries and the exposed pulp has been highlighted by global position statements [7], as dental caries in permanent teeth remains the most prevalent disease in the world today and affects patients in all countries and social classes [43]. Although the benefits of inflammatory suppression have been advocated, no currently available commercial dental or pulp capping material has specific immunomodulatory or targeted anti-inflammatory actions.

2.2. To promote pulpal repair processes

Pulp tissue, however, has an innate ability to heal if the injurious challenge is removed and the tooth suitably restored [5]. In deep cavities, the group of strategies aimed at maintaining the vitality of the pulp are called VPT [7]. Interestingly, although limiting pulpal inflammation is critical during VPT, it appears that a controlled level of pulpitis is helpful, at least in the initial stages, to drive reparative processes [44,45]. If possible, minimally invasive, biologically-based VPTs (e.g., pulp capping and pulpotomy) that preserve at least part of the pulp are preferable to conventional RCT, which is costly, technically complex [46]; destructive of tooth tissue [47] and often poorly carried out in dental practice [48,49].

In a response to challenge and injury the dentin-pulp complex not only reacts immunologically, but also forms tertiary dentin that provides a local physical reparative barrier adjacent to the injurious challenge and contributes to restoration of tissue integrity [2]. The term tertiary dentin actually describes a heterogeneous range of secretory responses, from a regular, tubular structure in continuity with primary and orthodentin, to the secretion of a dysplastic, defective and atubular matrix [6,50,51]. The cell responsible for the secretory responses also differs with reactionary tertiary dentin formed by an upregulation of activity of the existing primary odontoblast [52], while reparative tertiary dentin is formed by a newly differentiated odontoblast-like cell [53]. This subdivision highlights not only differing reactions in response to mild and more severe external stimuli, but also the effect of severe stimuli on odontoblast survival and subsequent pulpal response (Fig. 1). In contrast to reactionary dentinogenesis, reparative dentin formation is a more complex process that will normally occur in response to stronger stimuli such as extremely deep caries [9] and will subsequently involve the recruitment of progenitor cells, which differentiate into odontoblast-like cells to form reparative dentin [53]. The reparative dentin in comparison with reactionary dentin displays a broad range of responses, which can vary in quality from tubular dentin replacement to an incomplete amorphous hard tissue bridge [2,6,51,54]. For descriptive and educational purposes, reactionary and reparative dentinogenesis are often described as separate processes, however, when the carious lesion is close to the pulp both processes are almost certain to occur simultaneously [55].

Fig. 1.

Fig. 1

Schematic of the processes of tertiary dentin formation. Reactionary and reparative dentinogenesis processes differ in the source of the secreting cell. Reactionary dentin is formed by the existing primary odontoblast with a mild stimulus (e.g. early stage of carious disease) activating the upregulation of existing odontoblast activity. During reactionary dentinogenesis the odontoblasts recognize the bacterial products and released DMCs diffusing through the dentin tubules, which increases cellular activity. Reparative dentin formation involves a more complex sequence of events in which a severe stimulus (e.g. increasing carious involvement of dentin) causes death of the primary odontoblasts, which are subsequently replaced following differentiation of progenitor or stem cells into odontoblast-like cells under the regulation of bioactive molecules (including DMCs). Although the nature of the cellular response is likely to be dependent upon the pulp environment, the mineralized tissue deposited at the pupal wound site will likely display a spectrum of dysplasia. A Enamel, B Dentin, C Pulp.

From a biological perspective, the cellular response is not only promoted by release of growth factors (GFs) and pro-mineralization cues from DPCs [56], but also orchestrated and regulated by bioactive molecules, including GFs, which are ‘fossilized’ in the dentin matrix during crown formation [[57], [58], [59]]. These bioactive dentin matrix components (DMCs) can be released by caries, irrigants and dental materials to stimulate the reparative process [[60], [61], [62], [63]]. Indeed, it has been shown that calcium hydroxide [60], the HCSC mineral trioxide aggregate (MTA) [61] and epigenetic modifying drugs such as HDACi [63] can all modify the release profile of DMCs and alter the cellular reparative response.

Opinion Continuing pulpal health and the secretion of a high-quality mineralized barrier define a successful response to a pulp capping procedure. The hard tissue response and the degree of regeneration of tubular dentin has been linked to several factors including the type of pulp capping material used. The influence of the material in modifying DMC release and the predictability, quality of the pulpal defensive response is important as it can enhance the regenerative responses in the exposed pulp, by improving not only tissue handling but also developing the biological properties of the material itself. Traditionally, however, dental filling material development has focused on physical attributes (e.g., wear and strength) and esthetic issues rather than biological properties, despite the pulp capping material coming into often direct contact with the cells of the dental pulp and the opportunity to directly target cellular processes.

2.3. To potentially regenerate dentin after pulp exposure and biomaterial placement

After pulp exposure and placement of a biomaterial in a pulp capping procedure, although hard tissue is formed it is generally considered to lack the tubular structure that defines dentin [64]. This questions whether the new secretory cell is producing dentin or simply dystrophic mineral has led to some investigators casting doubt on the veracity of the stem cell-led odontoblast-like cell theory with alternative suggestions that the formation of mineral could be attributed instead to fibroblasts [54] or fibrocytes [65]. The traditionally accepted view was that cellular differentiation of migrating progenitor cells occurred under the influence of bioactive molecules liberated from damaged dentin, which formed new odontoblast-like cells [52,62]. It is also possible that fibroblasts produce mineral in the reparative response, but this would also occur through a cyto-differentiation as these cells do not produce mineral in their basal state, but can be induced to form mineral under certain induced conditions [[66], [67], [68], [69]].

It is important to note, that the origin of the cell type that produces the repair is central to the notion that the dentin-pulp complex can be regenerated [70]. The extensive heterogeneity in hard tissue morphology may indicate that both repair and regeneration of dentin are possible with evidence that the quality of the healing response can be influenced by the environmental niche with the presence or absence of caries [71], type of capping material [6] or the operator's technical skill [9]. In order to control confounding factors, experimental pulp capping studies that histologically analyze the response of pulp cells to pulp capping use disease-free, unrestored teeth and short recall periods [72]. However, in the clinic pulpal injuries are affected by a host of events including infectious, inflammatory and operator variable, and as a result the interplay between pulp injury, infection, pulpitis and altered host defense responses over several months will be critical to the final outcome [2]. This type of experimental pulp capping studies also tends to focus on the choice of capping material [6,72,73] rather than other critical aspects including wound disinfection [74], bacterial leakage [75] or technical aspects of the procedure itself. Indeed, the bulk of studies investigating the comparative quality of pulp capping materials do not comment on the technical aspects of the procedure or verify the histological absence of microorganisms associated with failed or poor healing outcomes [76].

Opinion Although regeneration of dentin has been shown to occur after experimental pulp capping with HCSCs in ‘sound’ teeth (Fig. 2), the clinical reality is that the pulp is likely to be inflamed and tissue responses are less predictable. New improved materials could potentially improve the quality of hard tissue deposited at the wound interface, both in terms of quality and volume of dentin. Furthermore, current pulp capping and pulpotomy techniques involve placement of a setting dental material directly against the pulp tissue, which stimulates a hard tissue reparative barrier, but by its physical presence prevents any possible regeneration of dentin, as would be possible if a bioactive scaffold or collagen sponge were placed instead of hard setting material. However, the later tissue engineering-based approach would require a rethink in the way dentistry is currently provided as well as the development of a new set of approved dental biomaterials. There is little evidence at present that there is much development on commercially-developed tissue engineering strategies, in spite of evidence that there is considerable academic research activity in this area.

Fig. 2.

Fig. 2

Pulpal response to MTA capping after 3 months. Pulpal response to mineral trioxide aggregate (MTA) capping after 3 months observation. Distal microphotographic view (A) of the mesial half of a maxillary left third molar (tooth 28) shows the remnants of the restorative and capping material (CP) and a distinct hard tissue bridge (BR) across the exposed pulp (PU). The photomicrograph (B) is part of a histological section of the specimen in (A). Note the mineralized hard tissue barrier (BR), stretching across the full length of the exposed pulp (B, C). The rectangular areas demarcated in (C) and (D) are magnified in (D) and (E) respectively. Note the cuboidal pulpal cells (arrowheads) lining the bridge (BR) and absence of pulpal inflammation in (E). Original magnifications: a ·6, b ·8, c ·23, d ·200. Adapted with permission from Ref. [6].

2.4. To address the deficiencies of current pulp capping materials

Successful VPT relies upon and is influenced by the pulp capping material. Over the years, countless capping materials have been used including; gold foil [77], calcium hydroxide [78,79], resin-based composites [80], corticosteroid/antibiotic medicaments [81], resin-modified glass ionomer [82], and HCSCs [83]. Misconceptions regarding the role of a pulp capping material, have suggested that preventing microleakage is the only concern [75]; which perhaps led to the use of resin-bonded solutions that irritated pulp tissue and impaired its defensive reaction [84,85]. Although the seal of the capping material is important and should be provided by either the pulp capping material or the overlying restoration, other factors such as biocompatibility, pro-mineralization, anti-inflammatory and or anti-microbial properties are also key, as well as practical considerations related to handling, setting time, cost and radiopacity [86]. As the aim of the pulp capping material is to help maintain a viable pulp, the promotion of mineralization through tertiary reparative dentinogenesis increases the thickness of dentin between the pulp and the deepest part of any cavity and therefore the proximity of the threat to the pulp is distanced.

Calcium hydroxide materials in non-setting or hard-setting forms have been considered the gold standard material for many years [79,[87], [88], [89]]. Indeed, calcium hydroxide application directly onto the pulp can be successful clinically [90] and forms a mineral barrier [91]; however, the barrier is often porous and of poor quality [6,51]. Notably, from a mechanistic perspective the action of calcium hydroxide remains unclear [92] and generally forms low quality mineralized osteodentin when applied to the exposed pulp [6,51]. As a result, it has largely been superseded by the use of new materials such as HCSCs [83], which have demonstrated improved results histologically [15] with evidence of some tubularity over time [6], as well as clinically compared with calcium hydroxide [16- Hilton et al., 2013]. HCSC use; however, has been attributed to occurrences of postoperative tooth discoloration [93], an issue which subsequent HCSCs such as Biodentine do not pose to the same extent [94], while also favorably inducing mineralization [95] and cellular differentiation in vitro [96]. In addition to the biological effects of calcium hydroxide and HCSCs on DPCs, they also have the ability to induce the release of the aforementioned bound DMCs from dentin [60,61]. Current pulp capping materials including HCSC's, however, are limited by their lack of specificity as they are not designed to directly target regenerative or inflammatory processes, which is unfortunate as GFs [97], drugs [29] and lyophilized DMCs [98] have all been shown to improve mineralization and reparative processes in vitro and in vivo.

Opinion The introduction of HCSCs over 30 years ago has improved pulpal tissue responses and clinical outcomes in VPT. However, current VPT materials are limited by low-quality tertiary dentin formation, non-specific action, and the absence of targeted constituents focused on tissue regenerative strategies or targeting inflammation [12,24]. Therefore, although there is an urgent need to develop next-generation capping material, there is a need to consider not only the bioactivity of the material but also the ability of the material to prevent leakage in the long-term, so physical properties (although not necessarily strength) remain important.

3. Epigenetic therapeutic solutions – opportunities and challenges

As previously described, there is a need to develop smarter pulp capping biomaterials that contain pharmacological inhibitors or endogenous morphogens and are focused on pulp regenerative processes, thereby overcoming the limitations of existing materials. Several potential solutions have been investigated including the direct application of GFs [99], Wnt-signaling pathway inhibitors [100], anti-oxidants [101], anti-hypertensive drugs [26] and recently epigenetic-modifiers [27,102,103]. In the latter so-called, ‘epigenetic therapeutic approach’, drugs or epigenetic modifying agents are used to treat medical and dental conditions [104].

Targeting epigenetic machinery has exciting potential, as epigenetic modifications play an essential role in cell development and differentiation by regulating gene expression without altering the DNA sequence [105]. Recently, the critical role of epigenetic influence on embryonic stem cell (ESC) regulation and DPSC fate, as well as the therapeutic potential of orchestrating self-renewal and differentiation has been underlined [70,106,107]. The epigenetic regulatory mechanisms that have been well researched remain DNA methylation and post-translational histone modifications, although the emerging role of ncRNAs in the epigenetic regulation of gene expression has been the subject of intensive research in medicine and dentistry [108,109]. It is evident that epigenetics can control multiple transcriptional mechanisms central to the maintenance of health and response to disease, so there is an opportunity to target epigenetic modifications as diagnostic biomarkers or as part of a dental therapeutic strategy.

Dental pulp research in this area has largely focused on DNA-associated histone acetylation [103], a nuclear process balanced by histone-acetyltransferases (HATs) and histone-deacetylases (HDACs), as it has been shown that modifying this balance with pharmacological inhibitors can accelerate DPC mineralization and improve wound healing in vitro [28,29] as well as in vivo [110]. Targeting HDAC enzymes with HDACi seems to be the most promising way to therapeutically alter this balance, with a resulting modification of cellular transcription. Pan-HDACis target all Zn-dependent HDAC-isoforms and include suberoylanilide hydroxamic acid (SAHA) and Panobinostat, which are EU and FDA-approved to treat certain forms of cancer [111,112], and have been investigated in bone engineering [113] and inflammatory modification [114]; however, their use in VPT is novel [103] and not commercially approved.

Several different types of HDACis have shown considerable promise in the field of dental pulp regeneration, including Trichostatin A (TSA), SAHA and Valproic acid (VPA) (Table 1). In multiple studies low concentrations of these HDACis have been demonstrated to promote differentiation and migration in DPC with minimal toxicity in vitro, suggesting a potential in vivo role and translation towards clinical treatment of exposed dental pulp [[115], [116], [117], [118], [119]]. The first proof of principle study using a DPC culture showed that although TSA, VPA reduced DPC proliferation they also increased mineralization dose-dependently in a dental-pulp derived murine cell line only negatively affecting cell viability and cell cycle at the higher concentrations [27]. Although this work highlighted the potential of HDACi-enhanced DPC mineralization, it raised other questions relating to different responses in primary cultures, which have been demonstrated to react differently to HDACi than transformed cell lines [120,121].

Table 1.

A selection of studies that have investigated the effects of HDACis and DNMTis on mineralization and differentiation in dental pulp cell populations. TSA = Trichostatin A; SAHA= Suberyolanilide hydroxamic acid; VPA= Valproic acid; BMP= Bone morphogenetic protein; OPN= Osteopontin; ALP = Alkaline phosphatase. DMP = dentin matrix acidic phosphoprotein; DSPP = dentin sialophosphoprotein. Adapted from Ref. [109].

Reference Cell Population Specific HDACi or DNMTi Mineralization-associated Gene Expression Changes
[27] Duncan et al., 2012 DPC line (MDPC-23) TSA, VPA Up: BMP4, DMP1, TGF-β1
[115] Kwon et al., 2012 DPC line (MDPC-23) SAHA Up: DMP1, DSPP, ALP, Nestin
[28] Duncan et al., 2013 Rodent Primary DPCs TSA, VPA Up: BMP2, BMP4, DMP1, DSPP, Nestin
[110] Jin et al., 2013 Human DPSCs TSA Up: BSP, DMP1, DSPP,
Down: OCN
[116] Paino et al., 2014 Human DPSCs VPA Up: BSP, OPN
Down: OCN
[117] Liu et al., 2018 Human DPSCs LMK-235 Up: DSPP, ALP, RUNX2
[118] Lee et al., 2020 Human DPSCs MS-275 Up: DMP1, ALP, DSPP and RUNX2
[119] Man et al., 2021 Human DPSCs MI192 Up: BMP2, OCN, ALP
[102] Zhang et al., 2015 Human DPSCs 5-AZA-CdR Up: DSPP, DMP1, OSX, RUNX2, DLX5, ALP

Subsequent DPC experimentation using primary rodent DPCs [28] as well as human DPSC populations have consistently highlighted that pharmacological HDAC inhibition induced reparative cellular responses (increased cellular mineralization, pro-mineralization gene and protein markers) at concentrations which did not stimulate significant cytotoxic effects. Generally, pan-HDACi, including TSA, SAHA and VPA have been used experimentally [27,28,110,115,116], but recently isoform-specific HDACi have shown promising results targeting HDAC2/3 inhibition using MI192 [119] and HDAC4/5 inhibition using LM-235 [117], with resulting increases in calcific nodule formation in vitro. In vitro experimentation has also raised the issue of dose and concentration of HDACi with continuous HDACi administration (14 days), resulting in an inhibition of differentiation [28]. One in vivo study examined the effects of HDACis on dental pulp with the developmental effect of HDACi on dental tissues after TSA injection into the tails of pregnant mice. Subsequent histological analysis revealed that the volume of dentin deposited was thicker and the number of odontoblasts in the area was higher in the HDACi group than in the control group [110]. From a translational perspective, the stimulation of mineralization in DPCs without toxicity after short-term exposure to the HDACi supported the potential chairside application of these materials as pulp capping agents within VPT or even ostensibly as part of a tissue engineering strategy to replace lost pulp tissue (Fig. 3).

Fig. 3.

Fig. 3

Schematic diagram highlighting the therapeutic potential of HDACi in regenerative endodontics. (A) Vital pulp treatment. (i) Deep carious lesion exposes pulp tissue. (ii) HDACi topically applied to exposed pulp, potentially as a component of a dental restorative material [e.g., mineral trioxide aggregate (MTA), calcium hydroxide, resin-based composite (RBC)] promotes tissue-repair processes (mineralization, modulated inflammation and cell migration). (iii) Tooth permanently restored with amalgam and mineralized bridge formation evident under dental pulp capping material. (B) Pulp tissue engineering. (i) Pulp necrosis (ii) Necrotic tooth chemo-mechanical debrided and EDTA, NaOCl applied to dentine to release matrix components. (iii) HDACi applied within a cell or non-cell based scaffold stimulates further growth factor release, cell migration and differentiation. (iv) Tooth restored and new pulp-like tissue formed promoting continued root growth and restoring tooth tissue vitality. Reproduced from Ref. [109].

Mechanistically, researchers have attributed the action of HDACi to stimulate an increase in transcription, with one high-throughput study highlighting that SAHA promoted mineralization and cell migration in rodent DPCs by inducing the expression of matrix metalloproteinase 13 (Mmp13). Importantly, cell proliferation was not compromised when low concentrations of SAHA were applied [29). More recently, a variety of HDACis including TSA, VPA, sodium butyrate (NaB) and MS-275 were applied to a rodent odontoblast-like cell line, MDPC-23 [122]. All of the HDACi employed increased the expression of a range of mineralization-associated genes such as bone morphogenetic protein-2 (Bmp2), Bmp4, Osteocalcin (Ocn), dentin matrix acidic phosphoprotein (Dmp-1); dentin sialophosphoprotein (Dspp) and Runt-related transcription factor 2 (Runx2), albeit to varying extents. MS-275, VPA and NaB all increased mineralization of the cell line, as determined by Alizarin Red S staining. Notably, TSA did not significantly increase mineralization at any concentration ranging from 1 nM to 100 nM [122], which is consistent with the findings of the Duncan et al. [27], and supports the previously mentioned observation that transformed cell lines are less susceptible to the effects of HDACis.

To improve the clinical relevance of HDACi application specific to the tooth, the interaction between HDACis and dentin matrix was investigated in order to replicate the interface between dental restorative materials and pulp capping biomaterials after pulp exposure [63]. A range of restorative and pulp capping materials have previously been shown to release bioactive DMCs, which promote reparative events and guide tertiary reparative dentinogenesis [60]. HDACis released DMCs consisting of GFs previously identified as being released from dentin by endodontic irrigants and dental materials [21,57,60], as well as novel GFs [63]. Notably, the HDACis extracted GFs less efficiently than the well-characterized extractant EDTA for certain GFs (e.g., TGF-β1), but more effectively for others (e.g., GDF15, BDNF), while in comparison different HDACi had differing extraction profiles.

From a pulp capping perspective, the promotion of mineralization and the control of inflammation are both crucial to the success of the treatment [2]. In addition to the studies focused on the mineralization response (Table 1), HDACi are also known to exhibit anti-inflammatory properties [12], by reducing pro-inflammatory cytokine production in vitro and in vivo [114]. Indeed, lipopolysaccharide (LPS) stimulated cells cultured in the presence of SAHA exhibited reduced cytokine production for tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and interleukin-1-beta (IL-1β), in a dose-dependent manner [114]. Furthermore, several HDACi including SAHA have been shown to reduce inflammatory related pain in a rat model [123]; this is a finding which could be beneficial for a pulp capping material in the treatment of symptomatic pulpitis or classic ‘toothache’. As HDACi appear to have a pro-mineralization and anti-inflammatory effect in DPC populations, future research should focus on the effect on reparative effects under pulpitic conditions or in conditions of high glucose or oxidative stress. These environmental stressors will affect cell differentiation and likely reparative response. At present, in order to limit the influence of confounding factors current studies tend to examine mineralization or inflammation in isolation rather than examining the effect of HDACi in inflamed tissue or after carious exposure in vivo. This represents a limitation of the current literature in this area [103].

Notably, a solitary study analyzed the effects of the DNMTi 5-AZA-CdR on DPCs [102] and demonstrated that although mineralization- and dentinogenic-associated gene expression, including DSPP, DMP1, Osterix (OSX) and RUNX2, were all upregulated and calcific nodule formation was accelerated (Table 1) it was accompanied by a significant reduction in cell viability at all concentrations.

Opinion A series of studies have highlighted the potential benefit of developing new dental biomaterials targeted towards epigenetic processes. The promise of HDACi stems from their dual action of possessing pro-mineralization and anti-inflammatory properties. These effects have been demonstrated using rodent and human cells as well as in vivo models. However, in order to translate these potential next-generation therapies into clinical material for dentists, clinical trials on human subjects must be planned and funded. This has not to date been straightforward in dentistry with notable reticence of dental manufacturers to get involved in what they describe as ‘early-phase’ translational research (preferring to enter product development at a later stage) coupled with increasing regulatory hurdles, which makes the development of these smart restorations costly and time consuming. Unfortunately, these challenges risk holding back conservative treatment of carious induced disease, one of the most prevalent diseases globally.

4. What are the specific materials used for potential drug delivery in VPT?

4.1. Unintended therapeutic effects in traditional pulp-dressing materials

Historically, pulp capping materials have been an area of VPT in which new innovations have been investigated with the subsequent response to the new material analyzed [124]. As a result of the capping material directly contacting dental pulp tissue, the material represents the perfect vehicle to possess potentially beneficial pharmacological additives. Notably, traditional pulp capping materials were not doped but rather were ‘unintentionally’ equipped with the pharmacological effects that were beneficial to pulp tissue.

For example, the classic pulp capping material, calcium hydroxide, causes necrosis of the superficial layer of the pulp tissue by its high pH, which subsequently induces pulp defense and repair processes [125]. It has been postulated that the beneficial effect of calcium hydroxide is due to the calcium ions it releases [126] as well as an inherent ability to extract GFs from dentin [60], which can subsequently stimulate odontoblast-like cell differentiation of progenitor cells in the pulp tissue. MTA, which has largely replaced calcium hydroxide as the pulp capping material of choice, has succeeded and expanded the favorable characteristics of calcium hydroxide [127]. MTA application upregulates BMP expression in DPCs in vitro [128], which can stimulate subsequent matrix mineralization. MTA also induces DPC expression of the transcription factor Runx2, as well as other pro-mineralization molecules including OCN, alkaline phosphatase, dentin sialoprotein, and vascular endothelial growth factor (VEGF) [129]. Furthermore, MTA has been shown to release TGF-β1, adrenomedullin (ADM) and hepatocyte growth factor (HGF) from dentin [21,130]. Presently, a variety of bioactive molecules, including SCF, M-CSF, GM-CSF, IGF-1, IGFBP-1, NGF, and GDNF, are known to be liberated from dentin by MTA's effect [131,132]. The beneficial effects of HCSC pulp materials have been integrated into newer products, such as tricalcium silicate-based cement, Biodentine (Septodont, Saint-Maur-des-Fosses, France) or the resin-modified calcium silicate, TheraCal (Bisco, Schaumburg, IL, USA) [133- Kim et al., 2021]. Biodentine has been shown to induce TGF-β1 release from human DPCs and stimulate early dental pulp mineralization [134 – Laurent et al., 2012], while DPSCs incubated with TheraCal exhibited higher Runx2 expression [133]. Notably, although materials such as calcium hydroxide and MTA induce reparative processes, there is no controlled release of bioactive components or targeted therapeutic action.

The critical features of a vital pulp capping material are that they should provide a seal to microbes, be biocompatibility, pro-mineralization, anti-inflammatory and or anti-microbial, as well as practical considerations related to handling, setting time, cost and radiopacity. Furthermore, if possible particularly in the case of scaffolds the material should be capable of promoting regeneration [86].

4.2. Early attempts to incorporate bioactive reagents into pulp capping materials

Attempts to incorporate bioactive compounds into pulp capping materials is not new with documented experiments recorded in the 1970s. Anneroth and Bang capped the exposed pulp tissue of a Java monkey with demineralized dentin matrix and observed subsequent hard tissue formation in the teeth at the exposure site [135]. This experiment implemented the previously demonstrated ability of demineralized dentin matrix to induce ectopic bone formation [136], which later led to the discovery and purification of BMP from rat dentin [137,138]. The finding of BMPs in dentin stimulated scientists to apply these proteins to dental pulp, expecting the formation of reparative tertiary dentin. Nakashima and other groups implanted crude or purified BMPs on the surface of amputated pulp of dog or monkey and observed reparative dentin formation [[139], [140], [141]]. From a materials perspective these experiments utilized collagen matrix or the mixture with chondroitin as the carrier of BMPs and highlight the potential for scaffolds to act as vehicles to deliver bioactive components to the injury site with the tooth.

4.3. Recent attempts to incorporate drug delivery in VPT materials

Polymeric substrates, as a carrier of bioactive reagents, have distinctive advantages as for several reasons [142]: (i) they protect the bioactive reagents from degradation; (ii) they enable the localized and sustained release of active reagents, ensuring bioavailability of critical concentrations of the reagent; (iii) they serve as extracellular matrix (ECM)-mimicking scaffold supports allowing tissue ingrowth during tissue regeneration. Polymeric substrates are currently applied in various forms, such as scaffolds, hydrogels, and nanoparticles, as well as the combinations of these forms [142].

4.3.1. Naturally-derived polymeric materials

Naturally derived materials use molecules derived from the cells themselves. Their characteristics and functionality are preferable for drug delivery and occur as a result of years of natural selection. The advantage of natural materials cannot be overemphasized or readily be surpassed by any synthetic material, which can be tested for suitable characteristics over a relatively short time. A further advantage of natural materials is that they are relatively easily obtained due to their abundancy and production without strict reaction control or complicated steps. Presumably due to these advantages, naturally derived materials have been preferred for endodontic procedures and regenerative strategies for many years (Table 2). Several important natural materials are reviewed below.

Table 2.

Natural polymeric materials used for in vivo transplantation as well as human studies with multiple patients are listed. Reports using inorganic materials, in vitro research, and case reports describing single patient were not included. For individual studies with #, normal, and bold entries indicate ectopic, orthotopic transplantation, and human studies, respectively. The modification to the reagents used is briefly explained in parenthesis when necessary.

BMC = bone marrow cell; BMSC = bone marrow stem cell; BMMSC = bone marrow mesenchymal stem cell; DFSC = dental follicle stem cell; DMC = dental mesenchymal cell; DPC = dental pulp cell; DPSC = dental pulp stem cell; SCAP = stem cell from the apical papilla; SHED = stem cell from human exfoliated deciduous teeth; UCMSC = umbilical cord mesenchymal stem cell; ECM = extracellular matrix; GelMA = gelatin methacrylate; PEG = polyethylene glycol; PRP= platelet-rich plasma.

Material Reagents used Cell type First author Year Ref.
Collagen/gelatin
<fiber/membrane/sponge> BMP2, BMP4, TGF-β1 - Nakashima 1994 [143]
OP-1 (BMP7) - Rutherford 1994 [144]
BMP7 - Jepsen 1997 [180]
BMP7 - Rutherford 2000 [181]
BMP7 - Six 2002 [182]
DMP-1 - Almushayt 2006 [183]
#(collagen sponge) swine DPC Sumita 2006 [147]
#(collagen sponge) hDPSC Zhang 2006 [184]
FGF-2 - Kikuchi 2007 [185]
#DMP-1, ceramic powder hDPSC Prescott 2008 [146]
(collagen I/III sponge) dog CD31(−)/CD146(−) DPC Iohara 2009 [186]
FGF-2 - Ishimatsu 2009 [187]
(collagen sponge) - Inuyama 2010 [188]
#nano-hydroxyapatite rat DPSC Yang 2010 [189]
(collagen membrane), PRP (+collagen sponge) - Goyal 2011 [190]
SDF-1 dog CD105+ DPC Iohara 2011 [191]
SDF-1 dog CD31-/CD146- DPC, CD105+ DPC Nakashima 2011 [192]
#(chondroitin sulfate, hyaluronic acid) rat DPSC Zhang 2012 [193]
SDF-1 dog CD31 DPC, CD105+ DPC, BMC Ishizaka 2012 [194]
#BMP4, FGF2 rat DPC Srisuwan 2012 [195]
(collagen sponge) dog DPSC Wang 2013 [196]
#BMP7 hDPSC Yang 2012 [197]
G-CSF, (atelocolagen) dog DPSC, DPC Iohara 2013 [198]
#SCF hDPC Pan 2013 [199]
G-CSF, (atelocolagen) dog mobilized DPSC Iohara 2014 [200]
(intrafibrillar-silicified collagen) hDPSC Niu 2014 [201]
#(nanofibrous-gelatin), Magnesium phosphate hDPSC Qu 2014 [202]
EDTA-treated swine dentin matrix, (swine dental pulp ECM), PLGA-Gelatin sheet transduced swine DFSC Chen 2015 [164]
#(nanofibrous-gelatin) hDPSC Qu 2015 [203]
#Exosome hDPSC, hBMSC Huang 2016 [204]
G-CSF dog DPSC Iohara 2016 [205]
(gelatin sponge), simvastatin dog DPSC Jia 2016 [206]
(collagen membrane) - Sharma 2016 [207]
(collagen membrane), (gelatin foam) - Fahmy 2017 [208]
(collagen membrane) - Jiang 2017 [209]
G-CSF, (atelocollagen) mobilized hDPSC Nakashima 2017 [210]
preameloblast-conditioned medium - Bucchi 2019 [211]
leptin - Choi 2019 [212]
(collagen sponge) - Zaky 2020 [213]
(collagen sponge), β-Defensin 4 - Zhai 2020 [214]
blood clot - Shetty 2021 [215]
(collagen membrane) - Jiang 2022 [216]
#(nanofibrous gelatin) amphiregulin-treated hDPSC Li 2022 [217]
lithium chloride, Wnt3a - Sukarawan 2023 [218]
<gel> FGF-2 - Kikuchi 2007 [185]
FGF-2 - Ishimatsu 2009 [187]
#(collagen/chitosan blend) HAT-7 epithelial cell, hDPSC Ravindran 2010 [219]
#FGF-2, VEGF, PDGF, NGF, BMP-7 - Kim 2010 [220]
#SDF-1, bFGF, BMP-7 hDPC Suzuki 2011 [221]
#FGF-2, VEGF, PDGF -containing gelatin beads rat DPC Srisuwan 2013 [222]
FGF-2, VEGF, PDGF -containing gelatin beads rat DPC Srisuwan 2013 [222]
#G-CSF mobilized hDPSC, CD31 hDPC, hDPSC, hiPSC Murakami 2013 [223]
#(recombinant human collagen type I) transduced SHED Rosa 2013 [224]
#(Collagen TE®) mobilized hDPSC, hDPSC Horibe 2014 [225]
bFGF - Nagy 2014 [226]
(type I collagen) indium-111-oxine-labeled rat DPC Souron 2014 [227]
#bFGF, G-CSF - Takeuchi 2015 [228]
#SDF-1 - Zhang 2015 [229]
#PDGF-BB, NGF, BDNF - Li 2016 [230]
#(GelMA) hDPSC, HUVEC Khayat 2017 [231]
#(rat tail collagen-I) ECM-treated hDPSC Zhang 2017 [232]
(Collagen TE®) swine DPSC Zhu 2018 [233]
(collagen granule) - Mittal 2019 [234]
#bioactive glasses, FGF2 - Washio 2019 [235]
#exosome-like vehicle rat dental papilla cell Zhang 2020 [236]
(synthetic collagen) - Mittal 2021 [237]
#(GelMA) hDPSC, HUVEC Liang 2022 [148]
<microsphere/particle> FGF-2 - Kikuchi 2007 [185]
FGF-2 - Ishimatsu 2009 [187]
#FGF-2, VEGF, PDGF - Srisuwan 2012 [195]
FGF-2, VEGF, PDGF - Srisuwan 2012 [195]
#VEGF hDPSC Li 2016 [238]
#(GelMA) hDPSC Yang 2021 [239]

Alginate
<fiber/membrane/sponge> - - Machado 2020 [240]
<gel> #- rat DPC Fujiwara 2006 [241]
#- hDPC Kumabe 2006 [242]
emdogain, (propylene glycol alginate) - Orhan 2012 [243]
#- hSCAP cell line RP89 Lambricht 2014 [168]
emdogain, (propylene glycol alginate) - Matsumoto 2014 [244]
#VEGF, laponite, (RGD-alginate) hDPSC Zhang 2020 [154]
#(GelMA) hDPSC, HUVEC Liang 2022 [148]
<microsphere/particle> OP-1 (BMP7), TGF-β1 - Oliva-Rodríguez 2011 [245]
#calcium chloride, thrombin ferret DPSC Verma 2017 [246]
#TGF-β1, dexamethasone - Shrestha 2019 [247]
#VEGF, laponite, (RGD-alginate) hDPSC Zhang 2020 [154]
#(GelMA) hDPSC, HUVEC Liang 2022 [148]

Chitosan
<fiber/membrane/sponge> #(carboxymethylcellulose) - Chen 2016 [161]
#BMP7 hDPSC Yang 2020 [197]
- - Mittal 2019 [234]
dexamethazone corticosteroid, bone matrix - Abbas 2020 [248]
- - Machado 2020 [240]
#simvastatin, calcium hydroxide - Soares 2021 [249]
#ciprofloxacin, IDR-1002, (Polyvinyl alcohol/Chitosan) hSCAP cell line RP89 Gonçalves da Costa Sousa 2022 [250]
<gel> #(collagen/chitosan blend) HAT-7 epithelial cell, hDPSC Ravindran 2010 [219]
silver-doped bioactive glass - Zhu 2019 [251]
(photobiomodulation therapy) - Moreira 2021 [252]
<microsphere/particle> #TGF-β1, dexamethasone - Shrestha 2019 [247]
Acemannan
<fiber/membrane/sponge> (Acemannan) - Jittapiromsak 2010 [253]
(Acemannan) - Songsiripradubboon 2016 [254]
(Acemannan) - Songsiripradubboon 2017 [254]

Hyaluronic acid
<fiber/membrane/sponge> (dried hyaluronic acid sponge) - Inuyama 2010 [188]
<gel> #(Colgel®) hSCAP cell line RP89 Lambricht 2014 [168]
#TGF-β1 swine DMC Tan 2015 [255]
TGF-β1 swine DMC Tan 2015 [255]
#platelet lysate, cellulose crystal hDPC Silva 2018 [256]
#- swine DPSC Zhu 2018 [233]
- swine DPSC Zhu 2018 [233]

Fibrin
<fiber/membrane/sponge> #extraction of dentin matrix proteins, TGF-β1, (fibrin sealant) (homing hDPSC) Widbiller 2018 [257]
<gel> #(PEGylated fibrin) SHED Galler 2011 [258]
#- - Ruangsawasdi 2016 [259]
#SCF - Ruangsawasdi 2017 [260]
#dentin matrix proteins hDPSC Galler 2018 [261]
#extraction of dentin matrix proteins, TGF-β1 (homing hDPSC) Widbiller 2018 [257]
<microsphere/particle> #calcium chloride, thrombin ferret DPSC Verma 2017 [246]

Demineralized dentine matrix
<fiber/membrane/sponge> #- rat DPSC Zhang 2012 [193]

Small intestinal submucosa
<fiber/membrane/sponge> #- rat DPSC Zhang 2012 [193]

Hydroxyethylcellulose (Natrosol®)
<gel> Chlorhexidine gluconate, Calcium hydroxide - Nagata 2014 [262]

ECM
<fiber/membrane/sponge> #(swine dental pulp ECM) - Chen 2015 [164]
EDTA-treated swine dentin matrix, (swine dental pulp ECM), PLGA-Gelatin sheet transduced swine DFSC Chen 2015 [164]
#dentin matrix hDPSC Tran 2015 [263]
#(swine dental pulp ECM) hDPSC Hu 2017 [264]
(swine dental pulp ECM) - Alqahtani 2018 [166]
#(DPSC ECM, HUVEC ECM) hBMMSC, hDPSC Huang 2018 [265]
#(bovine dental pulp ECM) - Bakhtiar 2021 [167]
#(human amniotic membrane ECM) hDPSC Bakhtiar 2022 [266]
<gel> #(Matrigel®) SCAP cell line RP89 Lambricht 2014 [168]
#(Matrigel®) hUCMSC, HUVEC Zhang 2020 [169]
#(Matrigel®) hUCMSC, VEGF-induced hUCMSC Zhang 2020 [170]
(treated dentin matrix) - Holiel 2021 [267]

Silk fibroin
<fiber/membrane/sponge> #SDF-1α hDPSC Yang 2015 [268]
SDF-1α - Yang 2015 [268]
#bFGF hDPSC Yang 2015 [269]

Glycerin
<gel> propolis - Pagliarin 2016 [270]

Cotton pellet
<fiber/membrane/sponge> BMP9 - Li 2021 [271]
4.3.1.1. Collagen/gelatin

Collagen/gelatin has been used as a drug carrying material in endodontic research for many years [143,144]. Collagen/gelatin is the principle component of natural ECM and as a result shows low cytotoxicity [145], prominent biocompatibility [146], and low immunogenicity in humans [147]. Additionally, collagen/gelatin can be used in solid form such as a sponge or matrix, as well as hydrogel when enzymatically processed (Table 2). Recently, the usage of collagen/gelatin continues to be high; with workers employing a gelatin methacrylate (GelMA), collagen molecule modified with reactive side group, for the scaffold harboring DPSC [148]. GelMA can also be crosslinked with UV irradiation (photo-crosslink) to add a high melting temperature to the gel, overcoming the drawbacks of a gelatin gel.

4.3.1.2. Alginate

Alginate is a heteropolysaccharide mainly produced from the cell wall of brown algae. It is biocompatible, biodegradable [149] and non-toxic material [150]. Ultimately, alginate capacity to form gel is the major reason for its use in tissue engineering and wound healing [151], indeed, many endodontic studies have used alginate in the gel form (Table 2). However, they have deficiencies such as poor mechanical stiffness, uncontrolled degradation rates in vivo [152], and no ability to support cell adhesion. These drawbacks have been counteracted by adding materials to the alginate structure, producing a more robust structure [151], or by modification with cell adhering residue such as RGD [153]. Excellent results have been shown in regenerating pulp-like tissue in vivo using a scaffold based on modified alginate [154].

4.3.1.3. Chitosan

Chitosan is a deacetylated form of chitin, an amino polysaccharide polymer produced mainly by arthropods. Chitosan is completely biodegradable, non-antigenic, biocompatible, and even antimicrobial [155,156]. Furthermore, it can be readily fabricated to a desirable shape and has been shown to induce osteogenic differentiation, making it a desirable scaffold material for bone tissue engineering [157,158]. Chitosan does possess some drawbacks for use as a scaffold, with a low mechanical strength and high degradation speed in human tissue [159]; however, these deficiencies have been addressed by adding other functional materials such as hyaluronic acid or collagen [160]. In endodontics, Chen et al. added carboxymethylcellulose to chitosan scaffolds and applied them to a pulp regeneration model [161]. Addition of carboxymethylcellulose decreased the pore diameter and increased internal porosity of the scaffold. The pulp cells co-cultured in the scaffolds show improved adhesion, spreading, cell capacity, and three-dimensional configurations [161]. Da Costa Sousa et al. added poly(vinyl alcohol) to produce nanofiber, in which they incorporated the antibiotic and immunomodulatory peptide IDR-1002 [162]. This scaffold with SCAP was subsequently embedded in tooth fragment and subcutaneously implanted in mice, resulting in pulp-like tissue generation within the scaffold [162].

4.3.1.4. Extracellular matrix (ECM)

Native ECM, produced by decellularization of native tissue generally using appropriate detergents such as Triton-X, has a potential to be an ideal scaffold because it includes all functional and structural molecules to support DPC activity [163]. Furthermore, ECM scaffold is biodegradable, biocompatible, and has preferable mechanical structure for seeded DPCs. Utilizing these merits of ECM scaffold in endodontic research, decellularized dental pulp ECM has been frequently used to harbor stem cells (Table 2) [[164], [165], [166], [167]]. Also, commercially available gel-form ECM (Matrigel®) has been used for pulp regeneration [[168], [169], [170]]. One significant drawback of native ECM is its immunogenicity mainly caused by incomplete decellularization and damage-associated molecular patterns (DAMPs) [171]. To bypass this problem, apoptosis-assisted decellularization, efficient antigen removal, crosslinking to restore protein distortion caused by decellularization, have been proposed as solutions (reviewed in Ref. [171]).

4.3.2. Synthetic polymers

Design flexibility when using synthetic polymers enables their physicochemical properties, such as degradation rate, microstructure, and mechanical strength [172]. However, due to the inherent absence of bioactive components, DPCs cannot readily proliferate, differentiate, or migrate in synthetic polymers [173]. Furthermore, there remains a need to establish their harmlessness before advocating their use in human teeth. Indeed, although there have been multiple synthetic polymers investigated, only three endodontic clinical studies using synthetic polymer scaffolds have been reported so far (Table 3). Notably, chemical modification and the incorporation of bioactive molecules into synthetic polymers can allow them to mimic the native environment [173].

Table 3.

Synthetic polymeric materials used for in vivo transplantation as well as human studies with multiple patients are listed. Reports using inorganic materials, in vitro research, and case reports describing single patient were not included. For individual studies with #, normal, and bold entries indicate ectopic, orthotopic transplantation, and human studies, respectively. The modification to the reagents used is briefly explained in parenthesis when necessary. Materials approved by the FDA for some medical purpose are shown in underlined.

BMMSC = bone marrow mesenchymal stem cell; DFSC = dental follicle stem cell; DMC = dental mesenchymal cell; DPC = dental pulp cell; DPSC = dental pulp stem cell; HDMEC = human dermal microvascular endothelial cells; HUVEC = human umbilical vein endothelial cell; SCAP = stem cell from the apical papilla; SHED = stem cell from human exfoliated deciduous teeth; ECM = extracellular matrix; MTA = mineral trioxide aggregate; PEG = polyethylene glycol; PGA = polyglycolic acid; PLGA = poly(lactic-co-glycolic acid); PLLA: poly-l-lactic acid.

Material Reagents used Cell type First Author Year Ref.
PGA
<fiber/membrane/sponge> #- hDPC, human gingival fibroblast Buurma 1999 [272]
#- swine DPC Sumita 2006 [147]
#TGF-β1, (+10% PLLA) swine DMC Tan 2015 [255]
#(+3% PLLA) rat tooth bud cell Duailibi 2004 [273]
#(+3% PLLA) swine tooth bud cells Young 2002 [274]

PLLA
<fiber/membrane/sponge> #- SHED, HDMEC Cordeiro 2008 [275]
#- SHED Casagrande 2010 [276]
#- hDPSC Demarco 2010 [277]
#- SHED Sakai 2010 [278]
#dexamethasone, ascorbic acid, β-glycerophosphate, BMP7 hDPSC Wang 2010 [279]
#- hDPSC Wang 2011 [280]
#VEGF hDPSC Li 2016 [238]
#BMP2 hSCAP Wang 2016 [281]
#Matrigel® transduced hDPSC Zhang 2016 [282]
Matrigel® rat BMMSC Ito 2017 [283]
#- transduced hDPSC Silva 2017 [284]
Matrigel® rat BMMSC,rat endothelial cell Sueyama 2017 [285]
#simvastatin hDPC Soares 2018 [26]
Matrigel® LacZ-labeled rat BMMSC Kaneko 2019 [286]
Matrigel® rat DPSC Zaw 2022 [287]
#- transduced hDPSC Zhang 2022 [288]
<microsphere/particle> #- hDPSC Kuang 2015 [289]
- hDPSC Kuang 2016 [290]
#BMP2 hSCAP Wang 2016 [281]

PLGA
<fiber/membrane/sponge> #- hDPSC, hSCAP Huang 2010 [291]
#- rat DPSC Zhang 2011 [193]
#hydroxyapatite, tricalcium phosphate, calcium carbonate hydroxyapatite hDPSC, rat tooth bud cells Zheng 2011 [292]
#- hDPSC Sun 2014 [293]
EDTA-treated swine dentin matrix, (swine dental pulp ECM), PLGA-Gelatin sheet transduced swine DFSC Chen 2015 [164]
- - Sharma 2016 [207]
#- rat tooth bud cell Duailibi 2004 [273]
#- hDPSC grown in microgravity condition Li 2017 [294]
#- swine tooth bud cells Young 2002 [274]
<gel> collagen, risedronate, lornoxicam - Shamma 2017 [295]

PCL (poly(ε-caprolactone))
<fiber/membrane/sponge> SDF-1, BMP7 - Kim 2010 [296]
#nano-hydroxyapatite rat DPSC Yang 2010 [189]
MTA - Lee 2011 [297]
#NGF mouse tooth germ cells Eap 2013 [298]
#DMOG hDPC Yoo 2018 [299]

Self assembling peptide (SAP)
<gel> #FGF-2, TGF-β1, VEGF, (MMP2-cleavable peptide with RGD motif) hDPSC Galler 2011 [178]
#FGF-2, TGF-β1, VEGF, (MMP2-cleavable peptide with RGD motif) hDPSC Galler 2012 [300]
#(RADA16-I) SHED Rosa 2013 [224]
#(RADA16-I) hDPSC, HUVEC Dissanayaka 2014 [301]
(RADA16-I) swine DPC Mangione 2017 [302]
#dentin matrix proteins, (MMP2-cleavable peptide with RGD motif) hDPSC Galler 2018 [261]
extraction of dentin matrix proteins, TGF-β1, (MMP2-cleavable peptide with RGD motif) - Moon 2018 [303]
#extraction of dentin matrix proteins, TGF-β1, (MMP2-cleavable peptide with RGD motif) (homing hDPSC) Widbiller 2018 [257]
#- gene-modified hDPSC Zhu 2019 [304]
(RGD- and VEGF-mimetic peptide) hDPSC Xia 2020 [305]
#(angiogenic hydrogel), FGF4, FGF9 hCNC-like cell Kobayashi 2021 [306]
#(angiogenic hydrogel) - Siddiqui 2021 [179]
(angiogenic hydrogel) - Siddiqui 2021 [179]

PEG
<gel> Chlorhexidine gluconate - Rodríguez-Benítez 2014 [307]
metronidazole, ciprofloxacin, minocycline - Rodríguez-Benítez 2014 [307]
metronidazole, ciprofloxacin, minocycline - Pagliarin 2016 [270]
#dentin matrix proteins hDPSC Galler 2018 [261]
metronidazole, ciprofloxacin, minocycline - Neelamurthy 2018 [308]

Polydioxanone
<fiber/membrane/sponge> #VEGF - Yadlapati 2017 [309]
metronidazole, ciprofloxacin, minocycline - Bottino 2019 [310]

Poly-N-isopropylacrylamide
<fiber/membrane/sponge> #- hDPSC Itoh 2018 [311]

Polyvinyl alcohol
<fiber/membrane/sponge>
#chlorhexidine gluconate - Kalyan 2019 [312]
chlorhexidine gluconate - Kalyan 2019 [312]
#ciprofloxacin, IDR-1002, (Polyvinyl alcohol/Chitosan) hSCAP cell line RP89 da Costa Sousa 2022 [162]

Vitrogel 3D
<gel>
#SDF-1α, BMP-2 hSCAP Xiao 2019 [313]

Polyglycolic acid (PGA), poly-l-lactic acid (PLLA), polylactic-glycolic acid (PLGA), and poly(ε-caprolactone) (PCL) are repeatedly used in pulp regeneration research (Table 3). These polymers are used due to them being nontoxic, biodegradable, and their physicochemical properties such as mechanical stiffness, degradation speed, porosity, and microstructure can be controlled [174]. With these advantages, these four materials are approved by the FDA for some kind of medical use (PGA for absorbable suture, PLLA as a hydrophobic aliphatic polyester in different biomedical applications, PLGA for drug delivery, and PCL for use as an implant polymer material) [175]. However, local accumulation of acidic degradation products of these polymers can evoke a strong inflammatory response, which limits their biomedical applications [176].

Self-assembly peptides (SAP) are used in endodontic research principally as a hydrogel (Table 3). SAP monomers bind to each other via the hydrophobic sequence or by hydrogen bonds to form a nanofiber consisting of a β-sheet, β-helix, or α-helix structure, resulting in a hydrogel by way of their entanglement [177]. Modifications of the monomer sequence have added functionality to each gel; for example, the addition of RGD-motif and MMP2 digestion site, enabled the gel to be more biocompatible and biodegradable [178]. This hydrogel successfully induced hDPSC to differentiate into odontoblast-like cells [178]. Further, SAP with VEGF motif have been used to induce angiogenesis into a scaffold embedded in dog's teeth, which enhanced cell survival in the scaffold [179].

Opinion A plethora of research studies have been carried out in recent years to investigate and develop new scaffold materials principally related to a tissue engineering approach (Table 2, Table 3). These approaches allow the incorporation of bioactive components or drugs which may target aspects of the tissue inflammation and repair. Existing materials such as calcium hydroxide or MTA adopt a different approach, being solid in nature and directly contacting the exposed pulp tissue. Although these materials, exhibit pro-reparative effects they are not designed to allow tissue expansion or replacement of lost tissue, so are by their very nature not regenerative materials. By contrast synthetic and non-synthetic materials offer the prospect of tissue outgrowth and the regeneration of lost tissue. Several natural and synthetic materials have been investigated in vitro and in vivo and have the potential when doped with bioactive GFs or HDACi to improve the healing response. Notably, however, no scaffold either doped or not doped with bioactive components is currently available for dental practitioners to use. In the next section we explore what the potential reasons are for this.

5. Are epigenetic-based or other ‘smart’ next-generation dental materials likely to be developed?

Even if extensive preclinical laboratory biological testing of novel biomaterials addresses pertinent issues, for example in the case of HDACi-augmented pulp capping materials including off-target effects, delivery mechanisms, release kinetics, potentially altered mechanical properties as well as esthetic aspects relevant to dentistry [109], other ‘clinical’ hurdles have to be addressed. This testing is becoming more complex and has significant cost and time implications even for academic-industry research collaborations attempting to introduce new bioactive therapies. These regulatory obstacles can delay even strong ideas as industrial partners may question the likelihood of profit after several years of preclinical and clinical development. To take the example used throughout the current article, despite strong in vitro and in vivo biological evidence supporting the use of HDACi as part of a targeted dental restoration, much work remains to develop a next-generation dental product that is available for dentists to use. So why is this the case and is it likely to become easier for innovative dental products to evolve in the future?

Dentistry is an unusual area, because it rarely attracts the levels of funding (exchequer, international or industrial-based) evident in related medical disciplines, despite containing some of the most prevalent global human diseases including caries and periodontal disease and the 5th most prevalent human condition in dental trauma [43,314,315]. Furthermore, dental materials are the most commonly placed biomaterial in the human body with an estimated 175 million placed per annum in the United States alone [316], yet mysteriously there is not a sophisticated targeted dental biomaterial available for routine use by dentists in operative dentistry and endodontics, despite evidence to suggest that these new materials may provide better outcomes for patients [317]. Obtaining regulatory approval for a new dental material is becoming more complex, and was modified at least with Europe in May 2021, by the introduction of new regulation on medical devices [318]. Within EU law, medical devices regulation, which includes dental filling materials, has classified devices to class I, IIa, IIb and III according to their intended purpose and any potential associated-risks [318]. For pulp capping materials, which contact the dental pulp inside a tooth the categorization is clearly now stated as Class IIa without a drug substance and Class III if a drug (or likely active additive component is added). If we examine more closely the regulations as they pertain to dentistry and examine Annex VIII of the Medical Device Regulation rule 8 [318], which suggests that ‘All implantable devices and long-term surgically invasive devices are classified as class IIb unless they: are intended to be placed in the teeth, in which case they are classified as class IIa’; evidently, existing pulp capping materials such as calcium hydroxide or MTA are considered in this IIa category. Closer examination of pulp capping agents within Annex VIII rule 14 [318 - Regulation (EU) 2017/746 of the European Parliament] states that ‘All devices incorporating, as an integral part, a substance which, if used separately, can be considered to be a medicinal product, including a medicinal product derived from human blood or human plasma, and that has an action ancillary to that of the devices, are classified as class III.’ suggests that GFs, and even natural products contained within blood or extracellular matrix may be considered a drug or medicinal products and therefore class III. Note that a medicinal product has previously been defined within Europe [319] ‘as any substance or combination of substances presented as having properties for treating or preventing disease in human beings or any substance or combination of substances which may be used in or administered to human beings either with a view to restoring, correcting or modifying physiological functions by exerting a pharmacological, immunological or metabolic action, or to making a medical diagnosis’. A substance is further explained (Table 4). In the USA, a similar division exists within the FDA using three, not four categories. Generally, the higher the risk of the medical device, the higher the medical device classification with the result that Class III including pulp capping materials are considered the highest risk. With a higher classification comes more stringent data requirements to demonstrate the device's safety, effectiveness, and performance in a clinical biological environment and by extension more time to evaluate and more financial cost. One fundamental difference between class IIa and class III is the need for more clinical data for example, a clinical trial to be carried out for class III, which inevitably adds both time and cost into a project that ultimately affects a company's return to investment. For that reason, considerable legal debate is ongoing with regulators and the dental industry about the definition of what constitutes an ‘active component’, whether components are ‘leached’ out of the material as well as considerations about whether the effects of the added substances are direct or indirect.

Table 4.

Definition of substance that could be considered as a medicinal product Adapted from point 2 of Article 1 of [129 - Directive 2001/83/EC of the European Parliament].

Type of substance (any matter irrespective of origin which may be): For example:
Human Human blood and human blood products;
Animal Micro-organisms, whole animals, parts of organs, animal secretions, toxins, extracts, blood products;
Vegetable Micro-organisms, plants, parts of plants, vegetable secretions, extracts;
Chemical Elements, naturally occurring chemical materials and chemical products obtained by chemical change or synthesis;

EU Class III medical devices will involve more testing than IIa with objective clinical testing (by way of a clinical trial), an assessment of benefit and risk, safety and an analysis of whether existing clinical evidence from other medical uses can be extrapolated to the new purpose, so called ‘off-label’ use [318]. This type of regulation is difficult for academics to navigate and time consuming even for industrial partners with significant associated administrative burden. That said, the new regulation also present opportunities in that the addition of drugs or bioactive substances into a dental material will mean Class III testing rather than a separate testing route for medicinal products. There is an argument that at least for targeted pulp capping material development, this path has now become more straightforward in Europe.

The challenges of dental biomaterial development are shared in medicine where the rapid increase over the last 20 years in miRNA-related patents for diagnosis or targeted therapy has not yet resulted in an (FDA)-approved miRNA-based therapeutic [320]. This highlights not only the obstacles associated with development of a clinical product, but also regulatory issues, with product development taking time [321], albeit less time for medical devices than medicinal products [322,323]. In the past, new dental materials requiring simpler testing routes may have been prioritized over materials that require a medicinal product development for reasons of cost and expediency; however, within Europe the new legislation may imply that at least for VPT this is no longer an abbreviated route, which could provide the stimulus required for companies to support new ‘smart’ product development. Within the new EU regulation, the insertion of an epigenetic-modifying agent, the dental material remains a device, albeit now at class III. These development costs as before may have a ‘knock-on’ effect to the patient as they have to be incorporated into the product price which often in dentistry is borne by the patient rather than a third-party insurance company. So, the question manufacturers may ask themselves is, will the costs of biomaterial development result in a product that a significant number of high street dentists will avail of, albeit at a modestly but not significantly increased price?

Other groups have discussed the use of other pharmacological inhibitors, focused on the stimulation of Wnt/β-catenin pathway and used at low concentrations in VPT [317]. The use of these inhibitors, which have been prescribed therapeutically for diseases in other parts of the body, is an example of what the authors called ‘drug repurposing’ for dentistry [324], and is similar to the dental use of HDACi where FDA and EU-approval is available for treatment of myeloma [12]. The repurposing as part of a dental material offers the hope that a candidate drug can be immediately tested in vivo to investigate tertiary dentinogenesis responses [325]. Although the authors concluded that this approach bypasses the high level of economical and time investment that are usually required in novel drug discoveries, this is only partly true. In order to develop a dental material, the inhibitor will still need to undergo significant investigation in an animal pulp capping model before embarking on extensive human testing. This will require considerable funding and industrial partnerships, but potentially less than other new drugs.

Other indirect techniques within VPT have proposed using a modified glass ionomer dental restoration containing lithium to promote the Wnt-signaling pathway [326]. Unfortunately, this suggestion has not been adopted by dental materials manufacturers, partly due to the fact that glass ionomer for biological reasons is not recommended as a direct pulp capping agent [327].

Opinion Dental materials are classed as medical devices, which are subject to legal regulatory control in Europe and the United States. Recent changes in EU legislation have introduced new rules and obligations in this area, which classifies pulp capping agents as Class IIa or Class III if they contain a bioactive substance, medicinal product oDMCr drug. The definition for these substances is broad, with the result that new pulp capping materials including ‘smart’ vital pulp biomaterials including those with added ‘natural’ morphogens or in this example epigenetic modifiers will come under Class III. Although this will increase the experimental and regulatory burden for the development of these materials, it also provides genuine opportunity for new targeted material to develop. Development of such next-generation materials can only realistically occur in collaborations between industry and research partners or by ‘spin-put’ companies formed as a result of academic endeavors. Up to this point, dental companies have not been keen to fund the type of ‘early-stage’ development required to foster the type of relationships required to shift materials from proof of concept towards product development. Within the regenerative medicine space at least this type of partnership has worked in medical disciplines, but has been much less evident within dentistry. Potentially for this type of important partnership to blossom, governmental intervention and legislative change is required to incentivize companies to work with translational research units to move the discipline and eventually patient care forward.

6. Conclusions and opinion

Position statements from global international organizations have described the difficulties of predictably treating deep caries and as a result the conservative management of the exposed pulp has been designated a priority area for research activity, in order to promote better biomimetic solutions. Recent research has highlighted a range of substances, including pharmacological inhibitors or morphogens, that could supplement existing restorations or contribute bioactivity to new pulp capping dental materials to improve the regenerative healing response. These next-generation materials could encompass epigenetic therapeutic drugs such as HDACi that promote mineralization, reduce inflammation and stimulate reparative processes, with considerable volumes of recent research highlighting the potential of using these EU and FDA-approved epigenetic modifiers to work as part of a next-generation pulp capping material. Other smart restorations could include the use of antioxidants, growth factors, Wnt-signaling (GSK-3) inhibitors or simvastatin amongst other bioactive components all of which have been shown to have potential in improving biological responses in the dental pulp. In order for robust academic research to translate to the clinic, however, academic/industrial partnerships are required which aim to develop strong science into new dental products. Positively, as a result of new European legislation, the clinical development of new pulp capping materials now seems more likely, as all new pulp capping agents whether a bioactive substance is added or not will be designated in the same category. This in combination with repurposing of drugs within dentistry that are approved for other biological functions provides opportunity for new smart dental restorations in the future. The need for new pulp capping to address the deficiencies of existing restorations is evident, and the development of new targeted dental materials should be considered a priority area for dental material research in the next 10 years.

Authorship contribution statement

Henry F Duncan planned and wrote sections of and edited the manuscript. Yoshifumi Kobayashi: wrote sections of the manuscript and edited the manuscript. Michaela Kearney: wrote sections of and edited the manuscript. Emi Shimizu planned and wrote sections of and edited the manuscript.

Ethics approval and consent to participate

This opinion-led review paper did not require ethical approval. All authors consented to the final version of the manuscript.

Funding and conflict of interest

This research as supported by the following grants to Emi Shimizu, National Institute of Dental and Craniofacial Research (NIDCR), grant number R01DE025885, R01DE031812 and a Fulbright Health Impact award 2019 to Henry F. Duncan. The authors have no conflicts of interest to declare.

Data availability

The authors do not have permission to share data.

Footnotes

Peer review under responsibility of KeAi Communications Co., Ltd.

References

  • 1.Pashley D.H. Dynamics of the pulpo-dentin complex. Crit. Rev. Oral Biol. Med. 1996;7:104–133. doi: 10.1177/10454411960070020101. [DOI] [PubMed] [Google Scholar]
  • 2.Duncan H.F., Cooper P.R., Smith A.J. Dissecting dentine-pulp injury and wound healing responses: consequences for regenerative endodontics. Int. Endod. J. 2019;52:261–266. doi: 10.1111/iej.13064. [DOI] [PubMed] [Google Scholar]
  • 3.Smith A.J. Pulp responses to caries and dental repair. Caries Res. 2002;36:223–232. doi: 10.1159/000063930. [DOI] [PubMed] [Google Scholar]
  • 4.Reeves R., Stanley H.R. The relationship of bacterial penetration and pulpal pathosis in carious teeth. Oral Surg. Oral Med. Oral Pathol. 1966;22:59–65. doi: 10.1016/0030-4220(66)90143-5. [DOI] [PubMed] [Google Scholar]
  • 5.Mjör I.A., Tronstad L. The healing of experimentally induced pulpitis. Oral Surg. Oral Med. Oral Pathol. 1974;38:115–121. doi: 10.1016/0030-4220(74)90322-3. [DOI] [PubMed] [Google Scholar]
  • 6.R Nair P.N., Duncan H.F., Pitt Ford T.R., Luder H.U. Histological, ultrastructural and quantitative investigations on the response of healthy human pulps to experimental capping with mineral trioxide aggregate: a randomized controlled trial. Int. Endod. J. 2008;41:128–150. doi: 10.1111/j.1365-2591.2007.01329.x. [DOI] [PubMed] [Google Scholar]
  • 7.F Duncan H., Galler K.M., Tomson P.L., et al. European Society of Endodontology position statement: management of deep caries and the exposed pulp. Int. Endod. J. 2019;52:923–934. doi: 10.1111/iej.13080. European Society of Endodontology (ESE) developed by. [DOI] [PubMed] [Google Scholar]
  • 8.Duncan H.F., Cooper P.R. Pulp innate immune defense: translational opportunities. J. Endod. 2020;46:S10–S18. doi: 10.1016/j.joen.2020.06.019. [DOI] [PubMed] [Google Scholar]
  • 9.Bjørndal L., Simon S., Tomson P.L., Duncan H.F. Management of deep caries and the exposed pulp. Int. Endod. J. 2019;52:949–973. doi: 10.1111/iej.13128. [DOI] [PubMed] [Google Scholar]
  • 10.Paphangkorakit J., Osborn J.W. Discrimination of hardness by human teeth apparently not involving periodontal receptors. Arch. Oral Biol. 1998;43:1–7. doi: 10.1016/s0003-9969(97)00090-3. [DOI] [PubMed] [Google Scholar]
  • 11.Randow K., Glantz P.O. On cantilever loading of vital and non-vital teeth. An experimental clinical study. Acta Odontol. Scand. 1986;44:271–277. doi: 10.3109/00016358609004733. [DOI] [PubMed] [Google Scholar]
  • 12.Duncan H.F., Smith A.J., Fleming G.J., Cooper P.R. HDACi: cellular effects, opportunities for restorative dentistry. J. Dent. Res. 2011;90:1377–1388. doi: 10.1177/0022034511406919. [DOI] [PubMed] [Google Scholar]
  • 13.Duncan H.F., Kobayashi Y., Shimizu E. Growth factors and cell homing in dental tissue regeneration. Curr. Oral Health Rep. 2018;5:276–285. doi: 10.1007/s40496-018-0194-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Arora S., Cooper P.R., Friedlander L.T., et al. Potential application of immunotherapy for modulation of pulp inflammation: opportunities for vital pulp treatment. Int. Endod. J. 2021;54:1263–1274. doi: 10.1111/iej.13524. [DOI] [PubMed] [Google Scholar]
  • 15.Aeinehchi M., Eslami B., Ghanbariha M., Saffar A.S. Mineral trioxide aggregate (MTA) and calcium hydroxide as pulp-capping agents in human teeth: a preliminary report. Int. Endod. J. 2003;36:225–231. doi: 10.1046/j.13652591.2003.00652.x. [DOI] [PubMed] [Google Scholar]
  • 16.Hilton T.J., Ferracane J.L., Mancl L. Northwest practice-based research collaborative in evidence-based dentistry (NWP), comparison of CaOH with MTA for direct pulp capping: a PBRN randomized clinical trial. J. Dent. Res. 2013;92:16–22S. doi: 10.1177/0022034513484336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mente J., Hufnagel S., Leo M., et al. Treatment outcome of mineral trioxide aggregate or calcium hydroxide direct pulp capping: long-term results. J. Endod. 2014;40:1746–1751. doi: 10.1016/j.joen.2014.07.019. [DOI] [PubMed] [Google Scholar]
  • 18.Kundzina R., Stangvaltaite L., Eriksen H.M., Kerosuo E. Capping carious exposures in adults: a randomized controlled trial investigating mineral trioxide aggregate versus calcium hydroxide. Int. Endod. J. 2017;50:924–932. doi: 10.1111/iej.12719. [DOI] [PubMed] [Google Scholar]
  • 19.Simon S.R., Tomson P.L., Berdal A. Regenerative endodontics: regeneration or repair? J. Endod. 2014;40:S70–S75. doi: 10.1016/j.joen.2014.01.024. [DOI] [PubMed] [Google Scholar]
  • 20.Dammaschke T., Nowicka A., Lipski M., Ricucci D. Histological evaluation of hard tissue formation after direct pulp capping with a fast-setting mineral trioxide aggregate (RetroMTA) in humans. Clin. Oral Invest. 2019;23:4289. doi: 10.1007/s00784-019-02876-2. 4289. [DOI] [PubMed] [Google Scholar]
  • 21.Tomson P.L., Grover L.M., Lumley P.J., et al. Dissolution of bio-active dentine matrix components by mineral trioxide aggregate. J. Dent. 2007;35:636–642. doi: 10.1016/j.jdent.2007.04.008. [DOI] [PubMed] [Google Scholar]
  • 22.Zanini M., Sautier J.M., Berdal A., Simon S. Biodentine induces immortalized murine pulp cell differentiation into odontoblast-like cells and stimulates biomineralization. J. Endod. 2012;38:1220–1226. doi: 10.1016/j.joen.2012.04.018. [DOI] [PubMed] [Google Scholar]
  • 23.Parirokh M., Torabinejad M., Dummer P.M.H. Mineral trioxide aggregate and other bioactive endodontic cements: an updated overview - part I: vital pulp therapy. Int. Endod. J. 2018;51:177–205. doi: 10.1111/iej.12841. [DOI] [PubMed] [Google Scholar]
  • 24.Ferracane J.L., Cooper P.R., Smith A.J. Can interaction of materials with the dentin-pulp complex contribute to dentin regeneration? Odontology. 2010;98:2–14. doi: 10.1007/s10266-009-0116-5. [DOI] [PubMed] [Google Scholar]
  • 25.Zaugg L.K., Banu A., Walther A.R., et al. Translation approach for dentine regeneration using GSK-3 antagonists. J. Dent. Res. 2020;99:544–551. doi: 10.1177/0022034520908593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Soares D.G., Zhang Z., Mohamed F., et al. Simvastatin and nanofibrous poly(l-lactic acid) scaffolds to promote the odontogenic potential of dental pulp cells in an inflammatory environment. Acta Biomater. 2018;68:190–203. doi: 10.1016/j.actbio.2017.12.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Duncan H.F., Smith A.J., Fleming G.J., Cooper P.R. Histone deacetylase inhibitors induced differentiation and accelerated mineralization of pulp-derived cells. J. Endod. 2012;38:339–345. doi: 10.1016/j.joen.2011.12.014. [DOI] [PubMed] [Google Scholar]
  • 28.Duncan H.F., Smith A.J., Fleming G.J., Cooper P.R. Histone deacetylase inhibitors epigenetically promote reparative events in primary dental pulp cells. Exp. Cell Res. 2013;319:1534–1543. doi: 10.1016/j.yexcr.2013.02.022. [DOI] [PubMed] [Google Scholar]
  • 29.Duncan H.F., Smith A.J., Fleming G.J., et al. The histone-deacetylase-inhibitor suberoylanilide hydroxamic acid promotes dental pulp repair mechanisms through modulation of matrix metalloproteinase-13 activity. J. Cell. Physiol. 2016;231:798–816. doi: 10.1002/jcp.25128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Mjör I.A., Tronstad L. Experimentally induced pulpitis. Oral Sur. Oral Med. Oral Pathol. 1972;34:102–108. doi: 10.1016/0030-4220(72)90278-2. [DOI] [PubMed] [Google Scholar]
  • 31.Warfvinge J., Bergenholtz G. Healing capacity of human and monkey dental pulps following experimentally-induced pulpitis. Endod. Dent. Traumatol. 1986;2:256–262. doi: 10.1111/j.1600-9657.1986.tb00158.x. [DOI] [PubMed] [Google Scholar]
  • 32.Murray P.E., About I., Lumley P.J., et al. Postoperative pulpal and repair responses. J. Am. Dent. Assoc. 2000;131:321–329. doi: 10.14219/jada.archive.2000.0175. [DOI] [PubMed] [Google Scholar]
  • 33.Soden R.I., Botero T.M., Hanks C.T., Nör J.E. Angiogenic signaling triggered by cariogenic bacteria in pulp cells. J. Dent. Res. 2009;88:835–840. doi: 10.1177/0022034509341946. [DOI] [PubMed] [Google Scholar]
  • 34.Farges J.C., Alliot-Licht B., Renard E., et al. Dental pulp defence and repair mechanisms in dental caries. Mediat. Inflamm. 2015 doi: 10.1155/2015/230251. 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kakehashi S., Stanley H.R., Fitzgerald R.J. The effects of surgical exposures of dental pulps in germ-free and conventional laboratory rats. Oral Surg. Oral Med. Oral Pathol. 1965;20:340–349. doi: 10.1016/0030-4220(65)90166-0. [DOI] [PubMed] [Google Scholar]
  • 36.Fabricius L., Dahlén G., Ohman A.E., Möller A.J. Predominant indigenous oral bacteria isolated from infected root canals after varied times of closure. Scand. J. Dent. Res. 1982;90:134–144. doi: 10.1111/j.1600-0722.1982.tb01536.x. [DOI] [PubMed] [Google Scholar]
  • 37.Rôças I.N., Lima K.C., Assunção I.V., et al. Advanced caries microbiota in teeth with irreversible pulpitis. J. Endod. 2015;41:1450–1455. doi: 10.1016/j.joen.2015.05.013. [DOI] [PubMed] [Google Scholar]
  • 38.Murray P.E., Smith A.J., Garcia-Godoy F., Lumley P.J. Comparison of operative procedure variables on pulpal viability in an ex vivo model. Int. Endod. J. 2008;41:389–400. doi: 10.1111/j.1365-2591.2007.01364.x. [DOI] [PubMed] [Google Scholar]
  • 39.Pedano M.S., Li X., Yoshihara K., Landuyt K.V., Van Meerbeek B. Cytotoxicity and bioactivity of dental pulp-capping agents towards human tooth-pulp cells: a systematic review of in-vitro studies and meta-analysis of randomized and controlled clinical trials. Materials. 2002;13:2670. doi: 10.3390/ma13122670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rao A.C., Venkatesh K.V., Nandini V., D, et al. Evaluating the effect of tideglusib-loaded bioactive glass nanoparticles as a potential dentine regenerative. Material. Materials (Basel) 2022;15:4567. doi: 10.3390/ma15134567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Arora S., Cooper P.R., Friedlander L.T., et al. Potential application of immunotherapy for modulation of pulp inflammation: opportunities for vital pulp treatment. Int. Endod. J. 2021;54:1263–1274. doi: 10.1111/iej.13524. [DOI] [PubMed] [Google Scholar]
  • 42.Miyashita H., Worthington H.V., Qualtrough A., Plasschaert A. Pulp management for caries in adults: maintaining pulp vitality. Cochrane Database Syst. Rev. 2007;18:CD004484. doi: 10.1002/14651858.CD004484.pub2. [DOI] [PubMed] [Google Scholar]
  • 43.Petti S S., Andreasen J.O., Glendor U., Andersson L. The fifth most prevalent disease is being neglected by public health organisations. Lancet Global Health. 2018;6:e1070–e1071. doi: 10.1016/S2214-109X(18)30380-2. [DOI] [PubMed] [Google Scholar]
  • 44.Cooper Pr P.R., Takahashi Y., Graham L.W., et al. Inflammation-regeneration interplay in the dentine-pulp complex. J. Dent. 2010;38:687–697. doi: 10.1016/j.jdent.2010.05.016. [DOI] [PubMed] [Google Scholar]
  • 45.Duncan H.F., Cooper P.R. Pulp innate immune defense: translational opportunities. J. Endod. 2020;46:S10–S18. doi: 10.1016/j.joen.2020.06.019. 46. [DOI] [PubMed] [Google Scholar]
  • 46.Stanley H.R. Pulp capping: conserving the dental pulp - can it be done? Is it worth it? Oral Surg. Oral Med. Oral Pathol. 1989;68:628–639. doi: 10.1016/0030-4220(89)90252-1. [DOI] [PubMed] [Google Scholar]
  • 47.Reeh E.S., Messer H.H., Douglas W.H. Reduction in tooth stiffness as a result of endodontic and restorative procedures. J. Endod. 1989;15:512–516. doi: 10.1016/S0099-2399(89)80191-8. [DOI] [PubMed] [Google Scholar]
  • 48.De Moor R.J., Hommez G.M., De Boever J.G., et al. Periapical health related to the quality of root canal treatment in a Belgian population. Int. Endod. J. 2000;33:113–120. doi: 10.1046/j.1365-2591.2000.00295.x. [DOI] [PubMed] [Google Scholar]
  • 49.Di Filippo G., Sidhu S.K., Chong B.S. Apical periodontitis and the technical quality of root canal treatment in an adult sub-population in London. Br. Dent. J. 2014;216:E22. doi: 10.1038/sj.bdj.2014.404. [DOI] [PubMed] [Google Scholar]
  • 50.Kuttler Y. Classification of dentine into primary, secondary and tertiary. Oral Surg. Oral Med. Oral Pathol. 1959;12:996–999. doi: 10.1016/0030-4220(59)90207-5. [DOI] [PubMed] [Google Scholar]
  • 51.Cox C.F., Sübay R.K., Ostro E., et al. Tunnel defects in dentine bridges: their formation following direct pulp capping. Operat. Dent. 1996;21:4–11. [PubMed] [Google Scholar]
  • 52.Smith A.J., Cassidy N., Perry H., et al. Reactionary dentinogenesis. Int. J. Dev. Biol. 1995;39:273–280. [PubMed] [Google Scholar]
  • 53.Lesot H., Smith A.J., Tziafas D., et al. Biologically active molecules and dental tissue repair: a comparative review of reactionary and reparative dentinogenesis with the induction of odontoblast differentiation in vitro. Cell Mater. 1994;4:199–218. [Google Scholar]
  • 54.Ricucci D., Loghin S., Lin L.M., et al. Is hard tissue formation in the dental pulp after the death of the primary odontoblasts a regenerative or a reparative process? J. Dent. 2014;42:1156–1170. doi: 10.1016/j.jdent.2014.06.012. [DOI] [PubMed] [Google Scholar]
  • 55.Smith A.J., Duncan H.F., Diogenes A., et al. Exploiting the bioactive properties of the dentin-pulp complex in regenerative endodontics. J. Endod. 2016;42:47–56. doi: 10.1016/j.joen.2015.10.019. [DOI] [PubMed] [Google Scholar]
  • 56.Jeanneau C., Lundy F.T., El Karim I.A., About I. Potential therapeutic strategy of targeting pulp fibroblasts in dentin-pulp regeneration. J. Endod. 2017;43:S17–S24. doi: 10.1016/j.joen.2017.06.007. [DOI] [PubMed] [Google Scholar]
  • 57.Cassidy N., Fahey M., Prime S.S., Smith A.J. Comparative analysis of transforming growth factor-β isoforms 1-3 in human and rabbit dentine matrices. Arch. Oral Biol. 1997;42:219–223. doi: 10.1016/S0003-9969(96)00115-X. [DOI] [PubMed] [Google Scholar]
  • 58.Smith A.J. Vitality of the dentin-pulp complex in health and disease: growth factors as key mediators. J. Dent. Educ. 2003;67:678–689. [PubMed] [Google Scholar]
  • 59.Grando Mattuella L., Westphalen Bento L., de Figueiredo J.A., et al. Vascular endothelial growth factor and its relationship with the dental pulp. J. Endod. 2007;33:524–530. doi: 10.1016/j.joen.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 60.Graham L., Cooper P.R., Cassidy N., et al. The effect of calcium hydroxide on solubilisation of bio-active dentine matrix components. Biomaterials. 2006;27:2865–2873. doi: 10.1016/j.biomaterials.2005.12.020. [DOI] [PubMed] [Google Scholar]
  • 61.Smith A.J., Scheven B.A., Takahashi Y., et al. Dentine as a bioactive extracellular matrix. Arch. Oral Biol. 2012;57:109–121. doi: 10.1016/j.archoralbio.2011.07.008. [DOI] [PubMed] [Google Scholar]
  • 62.Galler K.M., Widbiller M., Buchalla W., et al. EDTA conditioning of dentine promotes adhesion, migration and differentiation of dental pulp stem cells. Int. Endod. J. 2016;49:581–590. doi: 10.1111/iej.12492. [DOI] [PubMed] [Google Scholar]
  • 63.Duncan H.F., Smith A.J., Fleming G.J., et al. Release of bio-active dentine extracellular matrix components by histone deacetylase inhibitors (HDACi) Int. Endod. J. 2017;50:24–38. doi: 10.1111/iej.12588. [DOI] [PubMed] [Google Scholar]
  • 64.Ricucci D., Loghin S., Niu L.N., Tay F.R. Changes in the radicular pulp-dentine complex in healthy intact teeth and in response to deep caries or restorations: a histological and histobacteriological study. J. Dent. 2018;73:76–90. doi: 10.1016/j.jdent.2018.04.007. [DOI] [PubMed] [Google Scholar]
  • 65.Yoshiba N., Edanami N., Tohma A., R, et al. Detection of bone marrow-derived fibrocytes in human dental pulp repair. Int. Endod. J. 2018;51:1187–1195. doi: 10.1111/iej.12940. [DOI] [PubMed] [Google Scholar]
  • 66.Ronchetti I., Boraldi F., Annovi G., et al. Fibroblast involvement in soft connective tissue calcification. Front. Genet. 2013;4:22. doi: 10.3389/fgene.2013.00022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Yamamoto K., Kishida T., Sato Y., et al. Direct conversion of human fibroblasts into functional osteoblasts by defined factors. Proc. Natl. Acad. Sci. U.S.A. 2015;112:6152–6157. doi: 10.1073/pnas.1420713112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mizoshiri N., Kishida T., Yamamoto K., et al. Transduction of Oct6 or Oct9 gene concomitant with Myc family gene induced osteoblast-like phenotypic conversion in normal human fibroblasts. Biochem. Biophys. Res. Commun. 2015;467:1110–1116. doi: 10.1016/j.bbrc.2015.10.098. [DOI] [PubMed] [Google Scholar]
  • 69.Yamamoto K., Kishida T., Nakai K., et al. Direct phenotypic conversion of human fibroblasts into functional osteoblasts triggered by a blockade of the transforming growth factor-β signal. Sci. Rep. 2018;8:8463. doi: 10.1038/s41598-018-26745-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Duncan H.F., Smith A.J., Fleming G.J., Cooper P.R. Epigenetic modulation of dental pulp stem cells: implications for regenerative endodontics. Int. Endod. J. 2016;49:431–446. doi: 10.1111/iej.12475. [DOI] [PubMed] [Google Scholar]
  • 71.Ricucci D., Siqueira J.F., Jr, Rôças I.N., et al. Pulp and dentine responses to selective caries excavation: a histological and histobacteriological human study. J. Dent. 2020;100 doi: 10.1016/j.jdent.2020.103430. [DOI] [PubMed] [Google Scholar]
  • 72.Fransson H., Wolf E., Petersson K. Formation of a hard tissue barrier after experimental pulp capping or partial pulpotomy in humans: an updated systematic review. Int. Endod. J. 2016;49:533–542. doi: 10.1111/iej.12480. [DOI] [PubMed] [Google Scholar]
  • 73.Aeinehchi M., Eslami B., Ghanbariha M., S Saffar A. Mineral trioxide aggregate (MTA) and calcium hydroxide as pulp-capping agents in human teeth: a preliminary report. Int. Endod. J. 2003;36:225–231. doi: 10.1046/j.1365-2591.2003.00652.x. [DOI] [PubMed] [Google Scholar]
  • 74.Ballal N.V., Duncan H.F., Wiedemeier D.B., et al. MMP-9 levels and NaOCl lavage in randomized trial on direct pulp capping. J. Dent. Res. 2022;101:414–419. doi: 10.1177/00220345211046874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cox C.F., Bergenholtz G., Heys D.R., et al. Pulp capping of dental pulp mechanically exposed to oral microflora: a 1‐2 year observation of wound healing in the monkey. J. Oral Pathol. 1985;14:156–168. doi: 10.1111/j.1600-0714.1985.tb00479.x. [DOI] [PubMed] [Google Scholar]
  • 76.Dammaschke T., Nowicka A., Lipski M., Ricucci D. Histological evaluation of hard tissue formation after direct pulp capping with a fast-setting mineral trioxide aggregate (RetroMTA) in humans. Clin. Oral Invest. 2019;23:4289. doi: 10.1007/s00784-019-02876-2. 4289. [DOI] [PubMed] [Google Scholar]
  • 77.Dammaschke T. The history of direct pulp capping. J. Hist. Dent. 2008;56:9–23. [PubMed] [Google Scholar]
  • 78.Heys D.R., Cox C.F., Heys R.J., Avery J.K. Histological considerations of direct pulp capping agents. J. Dent. Res. 1981;60:1371–1379. doi: 10.1177/00220345810600071401. [DOI] [PubMed] [Google Scholar]
  • 79.Pitt Ford T.R., Roberts G.J. Immediate and delayed direct pulp capping with the use of a new visible light-cured calcium hydroxide preparation. Oral Surg. Oral Med. Oral Pathol. 1991;71:338–342. doi: 10.1016/0030-4220(91)90311-y. [DOI] [PubMed] [Google Scholar]
  • 80.Horsted-Bindslev P., Vilkinis V., Sidlauskas A. Direct capping of human pulps with a dentin bonding system or with calcium hydroxide cement. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2003;96:591–600. doi: 10.1016/s1079-2104(03)00155-0. [DOI] [PubMed] [Google Scholar]
  • 81.Cowan A. Treatment of exposed vital pulps with a corticosteroid antibiotic agent. Br. Dent. J. 1966;120:521–532. [PubMed] [Google Scholar]
  • 82.do Nascimento A.B., Fontana U.F., Teixeira H.M., Costa C.A. Biocompatibility of a resin-modified glass ionomer cement applied as pulp capping in human teeth. Am. J. Dent. 2000;13:28–34. [PubMed] [Google Scholar]
  • 83.Torabinejad M., Chivian N. Clinical applications of mineral trioxide aggregate. J. Endod. 1999;25:197–205. doi: 10.1016/S0099-2399(99)80142-3. [DOI] [PubMed] [Google Scholar]
  • 84.Pameijer C.H., Stanley H.R. The disastrous effects of the "total etch" technique in vital pulp capping in primates. Am. J. Dent. 1998;11:S45–S54. [PubMed] [Google Scholar]
  • 85.Dammaschke T., Stratmann U., Fischer R.J., et al. A histologic investigation of direct pulp capping in rodents with dentin adhesives and calcium hydroxide. Quintessence Int. 2010;41:e62–e71. [PubMed] [Google Scholar]
  • 86.Careddu R., Duncan H.F. How does the pulpal response to Biodentine and ProRoot mineral trioxide aggregate compare in the laboratory and clinic? Br. Dent. J. 2018;225:743–749. doi: 10.1038/sj.bdj.2018.864. [DOI] [PubMed] [Google Scholar]
  • 87.Glass R.L., Zander H.A. Pulp healing. J. Dent. Res. 1949;39:97–107. doi: 10.1177/00220345490280021101. [DOI] [PubMed] [Google Scholar]
  • 88.Stanley H.R., Lundy T. Dycal therapy for pulp exposures. Oral Surg. Oral Med. Oral Pathol. 1972;34:818–827. doi: 10.1016/0030-4220(72)90300-3. [DOI] [PubMed] [Google Scholar]
  • 89.Tronstad L. Reaction of the exposed pulp to Dycal treatment. Oral Surg. Oral Med. Oral Pathol. 1974;38:945–953. doi: 10.1016/0030-4220(74)90348-x. [DOI] [PubMed] [Google Scholar]
  • 90.Brizuela C., Ormeño A., Cabrera C., et al. Direct pulp capping with calcium hydroxide, mineral trioxide aggregate, and biodentine in permanent young teeth with caries: a randomized clinical trial. J. Endod. 2017;43:1776–1780. doi: 10.1016/j.joen.2017.06.031. [DOI] [PubMed] [Google Scholar]
  • 91.Schröder U. Evaluation of healing following experimental pulpotomy of intact human teeth and capping with calcium hydroxide. Odontol. Revy. 1972;23:329–340. [PubMed] [Google Scholar]
  • 92.Sangwan P., Sangwan A., Duhan J., Rohilla A. Tertiary dentinogenesis with calcium hydroxide: a review of proposed mechanisms. Int. Endod. J. 2013;46:3–19. doi: 10.1111/j.1365-2591.2012.02101.x. [DOI] [PubMed] [Google Scholar]
  • 93.Parinyaprom N., Nirunsittirat A., Chuveera P., et al. Outcomes of direct pulp capping by using either ProRoot mineral trioxide aggregate or Biodentine in permanent teeth with carious pulp exposure in 6- to 18-year-old patients: a randomized controlled trial. J. Endod. 2018;44:341–348. doi: 10.1016/j.joen.2017.10.012. [DOI] [PubMed] [Google Scholar]
  • 94.Careddu R., Duncan H.F. A prospective clinical study investigating the effectiveness of partial pulpotomy after relating preoperative symptoms to a new and established classification of pulpitis. Int. Endod. J. 2021;54:2156–2172. doi: 10.1111/iej.13629. [DOI] [PubMed] [Google Scholar]
  • 95.Laurent P., Camps J., About I. Biodentine induces TGF-β1 release from human pulp cells and early dental pulp mineralization. Int. Endod. J. 2012;45:439–448. doi: 10.1111/j.1365-2591.2011.01995.x. [DOI] [PubMed] [Google Scholar]
  • 96.Zanini M., Sautier J.M., Berdal A., Simon S. Biodentine induces immortalized murine pulp cell differentiation into odontoblast-like cells and stimulates biomineralization. J. Endod. 2012;38:1220–1226. doi: 10.1016/j.joen.2012.04.018. [DOI] [PubMed] [Google Scholar]
  • 97.Tziafas D., Alvanou A., Papadimitriou S., Gasic J., Komnenou A. Effects of recombinant basic fibroblast growth factor, insulin-like growth factor-II and transforming growth factor-β1 on dog dental pulp cells in vivo. Arch. Oral Biol. 1998;43:431–444. doi: 10.1016/s0003-9969(98)00026-0. [DOI] [PubMed] [Google Scholar]
  • 98.Okamoto M., Takahashi Y., Komichi S., et al. Dentinogenic effects of extracted dentin matrix components digested with matrix metalloproteinases. Sci. Rep. 2018;8 doi: 10.1038/s41598-018-29112-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Tziafas D., Papadimitriou S. Role of exogenous TGF-beta in induction of reparative dentinogenesis in vivo. Eur. J. Oral Sci. 1998;106:192–196. doi: 10.1111/j.1600-0722.1998.tb02175.x. [DOI] [PubMed] [Google Scholar]
  • 100.Neves V.C., Babb R., Chandrasekaran D., Sharpe P.T. Promotion of natural tooth repair by small molecule GSK3 antagonists. Sci. Rep. 2017;7 doi: 10.1038/srep39654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Minamikawa H., Yamada M., Deyama Y., et al. Effect of N-acetylcysteine on rat dental pulp cells cultured on mineral trioxide aggregate. J. Endod. 2011;37:637–641. doi: 10.1016/j.joen.2011.02.012. [DOI] [PubMed] [Google Scholar]
  • 102.Zhang D., Li Q., Rao L., et al. Effect of 5-Aza-2'-deoxycytidine on odontogenic differentiation of human dental pulp cells. J. Endod. 2015;41:640–645. doi: 10.1016/j.joen.2014.12.006. [DOI] [PubMed] [Google Scholar]
  • 103.Yamauchi Y., Cooper P.R., Shimizu E., et al. Histone acetylation as a regenerative target in the dentine-pulp complex. Front. Genet. 2020;11:1. doi: 10.3389/fgene.2020.00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Yoo C., Jones P. Epigenetic therapy of cancer: past, present and future. Nat. Rev. Drug Discov. 2006;5:37–50. doi: 10.1038/nrd1930. [DOI] [PubMed] [Google Scholar]
  • 105.Allis C.D., Jenuwein T. The molecular hallmarks of epigenetic control. Nat. Rev. Genet. 2016;17:487–500. doi: 10.1038/nrg.2016.59. [DOI] [PubMed] [Google Scholar]
  • 106.Bayarsaihan D. Deciphering the epigenetic code in embryonic and dental pulp stem cells. Yale J. Biol. Med. 2016;89:539–563. [PMC free article] [PubMed] [Google Scholar]
  • 107.Rodas-Junco B.A., Canul-Chan M., Rojas-Herrera R.A., et al. Stem cells from dental pulp: what epigenetics can do with your tooth. Front. Physiol. 2017;8:999. doi: 10.3389/fphys.2017.00999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Peschansky V.J., Wahlestedt C. Non-coding RNAs as direct and indirect modulators of epigenetic regulation. Epigenetics. 2009;9:3–12. doi: 10.4161/epi.27473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kearney M., Cooper P.R., J Smith A., Duncan H.F. Epigenetic approaches to the treatment of dental pulp inflammation and repair: opportunities and obstacles. Front. Genet. 2018;9:311. doi: 10.3389/fgene.2018.00311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Jin H., Park J.-Y., Choi H., Choung P.-H. HDAC inhibitor Trichostatin A promotes proliferation and odontoblast differentiation of human dental pulp stem cells. Tissue Eng. 2013;19:613–624. doi: 10.1089/ten.tea.2012.0163. [DOI] [PubMed] [Google Scholar]
  • 111.Bolden J.E., Peart M.J., Johnstone R.W. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov. 2006;5:769–784. doi: 10.1038/nrd2133. [DOI] [PubMed] [Google Scholar]
  • 112.Granit A., Mishra K., Barasch D., et al. Metabolomic profiling of triple negative breast cancer cells suggests that valproic acid can enhance the anticancer effect of cisplatin. Front. Cell Dev. Biol. 2022;10 doi: 10.3389/fcell.2022.1014798. 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Boer J., De Licht R., Bongers M., et al. Inhibition of histone acetylation as a tool in bone tissue engineering. Tissue Eng. 2006;12:2927–2937. doi: 10.1089/ten.2006.12.2927. [DOI] [PubMed] [Google Scholar]
  • 114.Leoni F., Zaliani A., Bertolini G., et al. The antitumor histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits anti-inflammatory properties via suppression of cytokines. Proc. Natl. Acad. Sci. U.S.A. 2002;99:2995–3000. doi: 10.1073/pnas.052702999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kwon A., Park H.J.-, Baek K., et al. Suberoylanilide hydroxamic acid enhances odontoblast differentiation. J. Dent. Res. 2012;91:506–512. doi: 10.1177/0022034512443367. [DOI] [PubMed] [Google Scholar]
  • 116.Paino F., La Noce M., Tirino V., et al. Histone deacetylase inhibition with valproic acid downregulates osteocalcin gene expression in human dental pulp stem cells and osteoblasts: evidence for HDAC2 involvement. Stem Cell. 2014;32:279–289. doi: 10.1002/stem.1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Liu Z., Chen T., Han Q., et al. HDAC inhibitor LMK 235 promotes the odontoblast differentiation of dental pulp cells. Mol. Med. Rep. 2018;17:1445–1452. doi: 10.3892/mmr.2017.8055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lee E.C., Kim Y.M., Lim H.M., et al. The histone deacetylase inhibitor (MS-275) promotes differentiation of human dental pulp stem cells into odontoblast-like cells independent of the MAPK signaling system. Int. J. Mol. Sci. 2020;21:1–13. doi: 10.3390/IJMS21165771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Man K., Lawlor L., Jiang L.H., Yang X.B. The selective histone deacetylase inhibitor MI192 enhances the osteogenic differentiation efficacy of human dental pulp stromal cells. Int. J. Mol. Sci. 2021;22 doi: 10.3390/IJMS22105224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lee J.-H., Choy M.L., Ngo L., et al. Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc. Natl. Acad. Sci. U.S.A. 2010;107:14639–14644. doi: 10.1073/pnas.1008522107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ungerstedt J.S., Sowa Y., Xu W.-S., et al. Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc. Natl. Acad. Sci. U.S.A. 2005;102:673–678. doi: 10.1073/pnas.0408732102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Sultana S., Uehara O., Yoshida K., et al. The histone deacetylase inhibitor, entinostat (MS-275), induces the odontogenic differentiation of an odontoblast-like cell line in the absence of an osteoblast mineralization medium. Odontology. 2021;109:661–671. doi: 10.1007/s10266-020-00588-8. [DOI] [PubMed] [Google Scholar]
  • 123.Mao Y., Zhou J J., Liu X., et al. Comparison of different histone deacetylase inhibitors in attenuating inflammatory pain in rats. Pain Res. Manag. 2019;27 doi: 10.1155/2019/1648919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Hanna S.N., Perez Alfayate R., Prichard J. Vital pulp therapy an insight over the available literature and future expectations. Eur. Endod. J. 2020;5:46–53. doi: 10.14744/eej.2019.44154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Schröder U. Effects of calcium hydroxide-containing pulp-capping agents on pulp cell migration, proliferation, and differentiation. J. Dent. Res. 1985;64:541–548. doi: 10.1177/002203458506400407. [DOI] [PubMed] [Google Scholar]
  • 126.An S., Gao Y., Ling J., et al. Calcium ions promote osteogenic differentiation and mineralization of human dental pulp cells: implications for pulp capping materials. J. Mater. Sci. Mater. Med. 2012;23:789–795. doi: 10.1007/s10856-011-4531-0. [DOI] [PubMed] [Google Scholar]
  • 127.Torabinejad M. Wiley-Blackwell; London: 2014. Mineral Trioxide Aggregate: Properties and Clinical Applications. [Google Scholar]
  • 128.Yasuda Y., Ogawa M., Arakawa T., et al. The effect of mineral trioxide aggregate on the mineralization ability of rat dental pulp cells: an in vitro study. J. Endod. 2008;34:1057–1060. doi: 10.1016/j.joen.2008.06.007. [DOI] [PubMed] [Google Scholar]
  • 129.Paranjpe A., Zhang H., Johnson J.D. Effects of mineral trioxide aggregate on human dental pulp cells after pulp-capping procedures. J. Endod. 2010;36:1042–1047. doi: 10.1016/j.joen.2010.02.013. [DOI] [PubMed] [Google Scholar]
  • 130.Tomson P.L., J Lumley P., Alexander M.Y., et al. Hepatocyte growth factor is sequestered in dentine matrix and promotes regeneration-associated events in dental pulp cells. Cytokine. 2013;61:622–629. doi: 10.1016/j.cyto.2012.11.009. [DOI] [PubMed] [Google Scholar]
  • 131.Tomson P.L., Lumley P.J., Smith A.J., Cooper P.R. Growth factor release from dentine matrix by pulp-capping agents promotes pulp tissue repair-associated events. Int. Endod. J. 2017;50:281–292. doi: 10.1111/iej.12624. [DOI] [PubMed] [Google Scholar]
  • 132.Matsumura S., Quispe-Salcedo A., Schiller C.M., et al. IGF-1 mediates EphrinB1 activation in regulating tertiary dentin formation. J. Dent. Res. 2017;96:1153–1161. doi: 10.1177/0022034517708572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kim Y., Lee D., Kim H.M., et al. Biological characteristics and odontogenic differentiation effects of calcium silicate-based pulp capping materials. Materials. 2021;14:16. doi: 10.3390/ma14164661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Laurent P., Camps J., About I. Biodentine(TM) induces TGF-beta1 release from human pulp cells and early dental pulp mineralization. Int. Endod. J. 2012;45:439–448. doi: 10.1111/j.1365-2591.2011.01995.x. [DOI] [PubMed] [Google Scholar]
  • 135.Anneroth G., Bang G. The effect of allogeneic demineralized dentin as a pulp capping agent in Java monkeys. Odontol. Revy. 1972;23:315–328. [PubMed] [Google Scholar]
  • 136.Bang G., Urist M.R. Bone induction in excavation chambers in matrix of decalcified dentin. Arch. Surg. 1967;94:781–789. doi: 10.1001/archsurg.1967.01330120035008. [DOI] [PubMed] [Google Scholar]
  • 137.Urist M.R., Strates B.S. Bone morphogenetic protein. J. Dent. Res. 1971;50:1392–1406. doi: 10.1177/00220345710500060601. [DOI] [PubMed] [Google Scholar]
  • 138.Butler W.T., Mikulski A., Urist M.R., et al. Noncollagenous proteins of a rat dentin matrix possessing bone morphogenetic activity. J. Dent. Res. 1977;56:228–232. doi: 10.1177/00220345770560030601. [DOI] [PubMed] [Google Scholar]
  • 139.Nakashima M. The induction of reparative dentine in the amputated dental pulp of the dog by bone morphogenetic protein. Arch. Oral Biol. 1990;35:493–497. doi: 10.1016/0003-9969(90)90078-o. [DOI] [PubMed] [Google Scholar]
  • 140.Nakashima M. Induction of dentin formation on canine amputated pulp by recombinant human bone morphogenetic proteins (BMP)-2 and -4. J. Dent. Res. 1994;73:1515–1522. doi: 10.1177/00220345940730090601. [DOI] [PubMed] [Google Scholar]
  • 141.Rutherford R.B., Wahle J., Tucker M., et al. Induction of reparative dentine formation in monkeys by recombinant human osteogenic protein-1. Arch. Oral Biol. 1993;38:571–576. doi: 10.1016/0003-9969(93)90121-2. [DOI] [PubMed] [Google Scholar]
  • 142.Shrestha S., Kishen A. Bioactive molecule delivery systems for dentin-pulp tissue engineering. J. Endod. 2017;43:733–744. doi: 10.1016/j.joen.2016.12.020. [DOI] [PubMed] [Google Scholar]
  • 143.Nakashima M. Induction of dentine in amputated pulp of dogs by recombinant human bone morphogenetic proteins-2 and -4 with collagen matrix. Arch. Oral Biol. 1994;39:1085–1089. doi: 10.1016/0003-9969(94)90062-0. [DOI] [PubMed] [Google Scholar]
  • 144.Rutherford R.B., Spangberg L., Tucker M., et al. The time-course of the induction of reparative dentine formation in monkeys by recombinant human osteogenic protein-1. Arch. Oral Biol. 1994;39:833–838. doi: 10.1016/0003-9969(94)90014-0. [DOI] [PubMed] [Google Scholar]
  • 145.H Silver F., Pins G. Cell growth on collagen: a review of tissue engineering using scaffolds containing extracellular matrix. J. Long Term Eff. Med. Implants. 1992;2:67–80. [PubMed] [Google Scholar]
  • 146.Prescott R.S., Alsanea R., Fayad M.I., et al. In vivo generation of dental pulp-like tissue by using dental pulp stem cells, a collagen scaffold, and dentin matrix protein 1 after subcutaneous transplantation in mice. J. Endod. 2008;34:421–426. doi: 10.1016/j.joen.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Sumita Y., Honda M.J., Ohara T., et al. Performance of collagen sponge as a 3-D scaffold for tooth-tissue engineering. Biomaterials. 2006;27:3238–3248. doi: 10.1016/j.biomaterials.2006.01.055. [DOI] [PubMed] [Google Scholar]
  • 148.Liang X., Xie L., Zhang Q., et al. Gelatin methacryloyl-alginate core-shell microcapsules as efficient delivery platforms for prevascularized microtissues in endodontic regeneration. Acta Biomater. 2022;144:242–257. doi: 10.1016/j.actbio.2022.03.045. [DOI] [PubMed] [Google Scholar]
  • 149.Zhang L., Morsi Y., Wang Y., et al. Review scaffold design and stem cells for tooth regeneration. J. Dent. Sci. Res. 2013;49:14–26. doi: 10.1016/j.jdsr.2012.09.001. [DOI] [Google Scholar]
  • 150.Goh C., Heng P., Chan L. Alginates as a useful natural polymer for microencapsulation and therapeutic applications. Carbohydr. Polym. 2012;88:1–12. doi: 10.1016/j.carbpol.2011.11.012. [DOI] [Google Scholar]
  • 151.Ahmad Raus R., Wan Nawawi W.M.F., Nasaruddin R.R. Alginate and alginate composites for biomedical applications. Asian J. Pharm. Sci. 2021;16:280–306. doi: 10.1016/j.ajps.2020.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Bouhadir K.H., Lee K.Y., Alsberg E., et al. Degradation of partially oxidized alginate and its potential application for tissue engineering. Biotechnol. Prog. 2001;17:945–950. doi: 10.1021/bp010070p. [DOI] [PubMed] [Google Scholar]
  • 153.Rowley J.A., Madlambayan G., Mooney D.J. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials. 1999;20:45–53. doi: 10.1016/s0142-9612(98)00107-0. [DOI] [PubMed] [Google Scholar]
  • 154.Zhang R., Xie L., Wu H., et al. Alginate/laponite hydrogel microspheres co-encapsulating dental pulp stem cells and VEGF for endodontic regeneration. Acta Biomater. 2020;113:305–316. doi: 10.1016/j.actbio.2020.07.012. [DOI] [PubMed] [Google Scholar]
  • 155.Santos V.P., Marques N.S.S., Maia P., et al. Seafood waste as attractive source of chitin and chitosan production and their applications. Int. J. Mol. Sci. 2020;21:12. doi: 10.3390/ijms21124290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Satitsri S., Muanprasat C. Chitin and chitosan derivatives as biomaterial resources for biological and biomedical applications. Molecules. 2020;25:24. doi: 10.3390/molecules25245961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Fasolino I., Raucci M.G., Soriente A., et al. Osteoinductive and anti-inflammatory properties of chitosan-based scaffolds for bone regeneration. Mater. Sci. Eng. C. Mater. Biol. Appl. 2019;105 doi: 10.1016/j.msec.2019.110046. [DOI] [PubMed] [Google Scholar]
  • 158.Ranganathan S., Balagangadharan K., Selvamurugan N. Chitosan and gelatin-based electrospun fibers for bone tissue engineering. Int. J. Biol. Macromol. 2019;133:354–364. doi: 10.1016/j.ijbiomac.2019.04.115. [DOI] [PubMed] [Google Scholar]
  • 159.Bellich B., D'Agostino I., Semeraro S., et al. The good, the bad and the ugly" of chitosans. Mar. Drugs. 2016;14:5. doi: 10.3390/md14050099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Sukpaita T., Chirachanchai S., Pimkhaokham A., Ampornaramveth R.S. Chitosan-based scaffold for mineralized tissues regeneration. Mar. Drugs. 2021;19:10. doi: 10.3390/md19100551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Chen H., Fan M. Chitosan/carboxymethyl cellulose polyelectrolyte complex scaffolds for pulp cells regeneration. J. Bioact. Compat Polym. 2016;22:475–491. doi: 10.1177/0883911507081329. [DOI] [Google Scholar]
  • 162.Goncalves da Costa Sousa M., Conceicao de Almeida G., Martins Mota D.C., et al. Antibiofilm and immunomodulatory resorbable nanofibrous filing for dental pulp regenerative procedures. Bioact. Mater. 2022;16:173–186. doi: 10.1016/j.bioactmat.2022.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Teodori L., Costa A., Marzio R., et al. Native extracellular matrix: a new scaffolding platform for repair of damaged muscle. Front. Physiol. 2014;5:218. doi: 10.3389/fphys.2014.00218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Chen G G., Chen J J., Yang B., et al. Combination of aligned PLGA/Gelatin electrospun sheets, native dental pulp extracellular matrix and treated dentin matrix as substrates for tooth root regeneration. Biomaterials. 2015;52:56–70. doi: 10.1016/j.biomaterials.2015.02.011. [DOI] [PubMed] [Google Scholar]
  • 165.Hu L., Gao Z., Xu J., et al. Decellularized swine dental pulp as a bioscaffold for pulp regeneration. BioMed Res. Int. 2017 doi: 10.1155/2017/9342714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Alqahtani Q., Zaky S.H., Patil A., et al. Decellularized swine dental pulp tissue for regenerative root canal therapy. J. Dent. Res. 2018;97:1460–1467. doi: 10.1177/0022034518785124. [DOI] [PubMed] [Google Scholar]
  • 167.Bakhtiar H., Rajabi S., Pezeshki-Modaress M., et al. Optimizing methods for bovine dental pulp decellularization. J. Endod. 2021;47:62–68. doi: 10.1016/j.joen.2020.08.027. [DOI] [PubMed] [Google Scholar]
  • 168.Lambricht L., De Berdt P., Vanacker J., et al. The type and composition of alginate and hyaluronic-based hydrogels influence the viability of stem cells of the apical papilla. Dent. Mater. 2014;30:e349–e361. doi: 10.1016/j.dental.2014.08.369. [DOI] [PubMed] [Google Scholar]
  • 169.Zhang S., Zhang W., Li Y., et al. Cotransplantation of human umbilical cord mesenchymal stem cells and endothelial cells for angiogenesis and pulp regeneration in vivo. Life Sci. 2020;255 doi: 10.1016/j.lfs.2020.117763. [DOI] [PubMed] [Google Scholar]
  • 170.Zhang S., Zhang W., Li Y., et al. Human umbilical cord mesenchymal stem cell differentiation into odontoblast-like cells and endothelial cells: a potential cell source for dental pulp tissue engineering. Front. Physiol. 2020;11:593. doi: 10.3389/fphys.2020.00593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Kasravi M., Ahmadi A., Babajani A., et al. Immunogenicity of decellularized extracellular matrix scaffolds: a bottleneck in tissue engineering and regenerative medicine. Biomater. Res. 2023;27:10. doi: 10.1186/s40824-023-00348-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Jazayeri H.E., Lee S.M., Kuhn L., et al. Polymeric scaffolds for dental pulp tissue engineering: a review. Dent. Mater. 2020;36:e47–e58. doi: 10.1016/j.dental.2019.11.005. [DOI] [PubMed] [Google Scholar]
  • 173.Wu D.T., Munguia-Lopez J.G., Cho Y.W., et al. Polymeric scaffolds for dental, oral, and craniofacial regenerative medicine. Molecules. 2021;26:22. doi: 10.3390/molecules26227043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Liu H., Lu J., Jiang Q., M, et al. Biomaterial scaffolds for clinical procedures in endodontic regeneration. Bioact. Mater. 2022;12:257–277. doi: 10.1016/j.bioactmat.2021.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Zhang Y., Wu D., Zhao X., et al. Stem cell-friendly scaffold biomaterials: applications for bone tissue engineering and regenerative medicine. Front. Bioeng. Biotechnol. 2020;8 doi: 10.3389/fbioe.2020.598607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Ulery B.D., Nair L.S., Laurencin C.T. Biomedical applications of biodegradable polymers. J. Polym. Sci. B Polym. Phys. 2011;49:832–864. doi: 10.1002/polb.22259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Fu K., Wu H., Su Z. Self-assembling peptide-based hydrogels: fabrication, properties, and applications. Biotechnol. Adv. 2021;49 doi: 10.1016/j.biotechadv.2021.107752. [DOI] [PubMed] [Google Scholar]
  • 178.Galler K.M., D'Souza R.N., Federlin M., et al. Dentin conditioning codetermines cell fate in regenerative endodontics. J. Endod. 2011;37:1536–1541. doi: 10.1016/j.joen.2011.08.027. [DOI] [PubMed] [Google Scholar]
  • 179.Siddiqui Z., Sarkar B., Kim K.K., et al. Angiogenic hydrogels for dental pulp revascularization. Acta Biomater. 2021;126:109–118. doi: 10.1016/j.actbio.2021.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Jepsen S., Albers H.K., Fleiner B., M, et al. Recombinant human osteogenic protein-1 induces dentin formation: an experimental study in miniature swine. J. Endod. 1997;23:378–382. doi: 10.1016/S0099-2399(97)80187-2. [DOI] [PubMed] [Google Scholar]
  • 181.Rutherford R.B., Gu K. Treatment of inflamed ferret dental pulps with recombinant bone morphogenetic protein-7. Eur. J. Oral Sci. 2000;108:202–206. doi: 10.1034/j.1600-0722.2000.108003202.x. [DOI] [PubMed] [Google Scholar]
  • 182.Six N., Lasfargues J.J., Goldberg M. Differential repair responses in the coronal and radicular areas of the exposed rat molar pulp induced by recombinant human bone morphogenetic protein 7 (osteogenic protein 1) Arch. Oral Biol. 2002;47:177–187. doi: 10.1016/s0003-9969(01)00100-5. [DOI] [PubMed] [Google Scholar]
  • 183.Almushayt A., Narayanan K., Zaki A.E., George A. Dentin matrix protein 1 induces cytodifferentiation of dental pulp stem cells into odontoblasts. Gene Ther. 2006;13:611–620. doi: 10.1038/sj.gt.3302687. [DOI] [PubMed] [Google Scholar]
  • 184.Zhang W., Walboomers X.F., van Kuppevelt T.H., et al. The performance of human dental pulp stem cells on different three-dimensional scaffold materials. Biomaterials. 2006;27:5658–5668. doi: 10.1016/j.biomaterials.2006.07.013. [DOI] [PubMed] [Google Scholar]
  • 185.Kikuchi N., Kitamura C., Morotomi T., et al. Formation of dentin-like particles in dentin defects above exposed pulp by controlled release of fibroblast growth factor 2 from gelatin hydrogels. J. Endod. 2007;33:1198–1202. doi: 10.1016/j.joen.2007.07.025. [DOI] [PubMed] [Google Scholar]
  • 186.Iohara K., Zheng L., Ito M., et al. Regeneration of dental pulp after pulpotomy by transplantation of CD31(-)/CD146(-) side population cells from a canine tooth. Regen. Med. 2009;4:377–385. doi: 10.2217/rme.09.5. [DOI] [PubMed] [Google Scholar]
  • 187.Ishimatsu H., Kitamura C., Morotomi T., et al. Formation of dentinal bridge on surface of regenerated dental pulp in dentin defects by controlled release of fibroblast growth factor-2 from gelatin hydrogels. J. Endod. 2009;35:858–865. doi: 10.1016/j.joen.2009.03.049. [DOI] [PubMed] [Google Scholar]
  • 188.Inuyama Y., Kitamura C., Nishihara T., et al. Effects of hyaluronic acid sponge as a scaffold on odontoblastic cell line and amputated dental pulp. J. Biomed. Mater. Res. B Appl. Biomater. 2010;92:120–128. doi: 10.1002/jbm.b.31497. [DOI] [PubMed] [Google Scholar]
  • 189.Yang X., Yang F., Walboomers X.F., et al. The performance of dental pulp stem cells on nanofibrous PCL/gelatin/nHA scaffolds. J. Biomed. Mater. Res. 2010;93:247–257. doi: 10.1002/jbm.a.32535. [DOI] [PubMed] [Google Scholar]
  • 190.Goyal B., Tewari S., Duhan J., Sehgal P.K. Comparative evaluation of platelet-rich plasma and guided tissue regeneration membrane in the healing of apicomarginal defects: a clinical study. J. Endod. 2011;37:773–780. doi: 10.1016/j.joen.2011.03.003. [DOI] [PubMed] [Google Scholar]
  • 191.Iohara K., Imabayashi K., Ishizaka R., et al. Complete pulp regeneration after pulpectomy by transplantation of CD105+ stem cells with stromal cell-derived factor-1. Tissue Eng. 2011;17:1911–1920. doi: 10.1089/ten.TEA.2010.0615. [DOI] [PubMed] [Google Scholar]
  • 192.Nakashima M., Iohara K. Regeneration of dental pulp by stem cells. Adv. Dent. Res. 2011;23:313–319. doi: 10.1177/0022034511405323. [DOI] [PubMed] [Google Scholar]
  • 193.Zhang H., Liu S., Zhou Y., et al. Natural mineralized scaffolds promote the dentinogenic potential of dental pulp stem cells via the mitogen-activated protein kinase signaling pathway. Tissue Eng. 2012;18:677–691. doi: 10.1089/ten.TEA.2011.0269. [DOI] [PubMed] [Google Scholar]
  • 194.Ishizaka R., Iohara K., Murakami M., et al. Regeneration of dental pulp following pulpectomy by fractionated stem/progenitor cells from bone marrow and adipose tissue. Biomaterials. 2012;33:2109–2118. doi: 10.1016/j.biomaterials.2011.11.056. [DOI] [PubMed] [Google Scholar]
  • 195.Srisuwan T., Tilkorn D.J., Al-Benna S., et al. Survival of rat functional dental pulp cells in vascularized tissue engineering chambers. Tissue Cell. 2012;44:111–121. doi: 10.1016/j.tice.2011.12.003. [DOI] [PubMed] [Google Scholar]
  • 196.Wang Y., Zhao Y., Jia W., et al. Preliminary study on dental pulp stem cell-mediated pulp regeneration in canine immature permanent teeth. J. Endod. 2013;39:195–201. doi: 10.1016/j.joen.2012.10.002. [DOI] [PubMed] [Google Scholar]
  • 197.Yang X., Han G., Pang X., Fan M. Chitosan/collagen scaffold containing bone morphogenetic protein-7 DNA supports dental pulp stem cell differentiation in vitro and in vivo. J. Biomed. Mater. Res. 2020;108:2519–2526. doi: 10.1002/jbm.a.34064. [DOI] [PubMed] [Google Scholar]
  • 198.Iohara K., Murakami M., Takeuchi N., et al. 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. doi: 10.5966/sctm.2012-0132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Pan S., Dangaria S., Gopinathan G., et al. SCF promotes dental pulp progenitor migration, neovascularization, and collagen remodeling - potential applications as a homing factor in dental pulp regeneration. Stem Cell Rev. Rep. 2013;9:655–667. doi: 10.1007/s12015-013-9442-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.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. doi: 10.1016/j.exger.2014.01.020. [DOI] [PubMed] [Google Scholar]
  • 201.Niu L.N., Sun J.Q., Li Q.H., et al. Intrafibrillar-silicified collagen scaffolds enhance the osteogenic capacity of human dental pulp stem cells. J. Dent. 2014;42:839–849. doi: 10.1016/j.jdent.2014.03.016. [DOI] [PubMed] [Google Scholar]
  • 202.Qu T., Jing J., Jiang Y., et al. Magnesium-containing nanostructured hybrid scaffolds for enhanced dentin regeneration. Tissue Eng. 2014;20:2422–2433. doi: 10.1089/ten.TEA.2013.0741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Qu T., Jing J., Ren Y., et al. Complete pulpodentin complex regeneration by modulating the stiffness of biomimetic matrix. Acta Biomater. 2015;16:60–70. doi: 10.1016/j.actbio.2015.01.029. [DOI] [PubMed] [Google Scholar]
  • 204.Huang C.C., Narayanan R., Alapati S., Ravindran S. Exosomes as biomimetic tools for stem cell differentiation: applications in dental pulp tissue regeneration. Biomaterials. 2016;111:103–115. doi: 10.1016/j.biomaterials.2016.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Iohara K., Fujita M., Ariji Y., et al. Assessment of pulp regeneration induced by stem cell therapy by magnetic resonance imaging. J. Endod. 2016;42:397–401. doi: 10.1016/j.joen.2015.11.021. [DOI] [PubMed] [Google Scholar]
  • 206.Jia W., Zhao Y., Yang J., et al. Simvastatin promotes dental pulp stem cell-induced coronal pulp regeneration in pulpotomized teeth. J. Endod. 2016;42:1049–1054. doi: 10.1016/j.joen.2016.03.007. [DOI] [PubMed] [Google Scholar]
  • 207.Sharma S., Neelam M. A comparative evaluation of natural and artificial scaffolds in regenerative endodontics: a clinical study. Saudi Endod. J. 2016;6:9–15. doi: 10.4103/1658-5984.171995. [DOI] [Google Scholar]
  • 208.Fahmy S.H., Hassanien E.E.S., Nagy M.M., et al. Investigation of the regenerative potential of necrotic mature teeth following different revascularisation protocols. Aust. Endod. J. 2017;43:73–82. doi: 10.1111/aej.12210. [DOI] [PubMed] [Google Scholar]
  • 209.Jiang X., Liu H., Peng C 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. doi: 10.1016/j.joen.2017.04.011. [DOI] [PubMed] [Google Scholar]
  • 210.Nakashima M., Iohara K., Murakami M., et al. Pulp regeneration by transplantation of dental pulp stem cells in pulpitis: a pilot clinical study. Stem Cell Res. Ther. 2017;8:61. doi: 10.1186/s13287-017-0506-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Bucchi C., Gimeno-Sandig A., Valdivia-Gandur I., et al. A regenerative endodontic approach in mature ferret teeth using rodent preameloblast-conditioned medium. Vivo. 2019;33:1143–1150. doi: 10.21873/invivo.11584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Choi S.H., Jang J.H., Koh J.T., et al. Effect of leptin on odontoblastic differentiation and angiogenesis: an in vivo study. J. Endod. 2019;45:1332–1341. doi: 10.1016/j.joen.2019.08.003. [DOI] [PubMed] [Google Scholar]
  • 213.Zaky S.H., AlQahtani Q., Chen J J., et al. Effect of the periapical "inflammatory plug" on dental pulp regeneration: a histologic in vivo study. J. Endod. 2020;46:51–56. doi: 10.1016/j.joen.2019.10.006. [DOI] [PubMed] [Google Scholar]
  • 214.Zhai Y., Yuan X., Zhao Y. Potential application of human beta-defensin 4 in dental pulp repair. Front. Physiol. 2020;11:1077. doi: 10.3389/fphys.2020.01077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Shetty H., Shetty S., Kakade A., et al. Three-dimensional qualitative and quantitative analyses of the effect of periradicular lesions on the outcome of regenerative endodontic procedures: a prospective clinical study. Clin. Oral Invest. 2021;25:691–700. doi: 10.1007/s00784-020-03583-z. [DOI] [PubMed] [Google Scholar]
  • 216.Jiang X., Liu H., Peng C. Continued root development of immature permanent teeth after regenerative endodontics with or without a collagen membrane: a randomized, controlled clinical trial. Int. J. Paediatr. Dent. 2022;32:284–293. doi: 10.1111/ipd.12853. [DOI] [PubMed] [Google Scholar]
  • 217.Li J., Wang Z., Wang J., et al. Amphiregulin regulates odontogenic differentiation of dental pulp stem cells by activation of mitogen-activated protein kinase and the phosphatidylinositol 3-kinase signaling pathways. Stem Cell Res. Ther. 2022;13:304. doi: 10.1186/s13287-022-02971-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Sukarawan W., Rattanawarawipa P., Yaemkleebbua K., et al. Wnt3a promotes odonto/osteogenic differentiation in vitro and tertiary dentin formation in a rat model. Int. Endod. J. 2023 doi: 10.1111/iej.13888. (Epub ahead of print) [DOI] [PubMed] [Google Scholar]
  • 219.Ravindran S., Song Y., George A. Development of three-dimensional biomimetic scaffold to study epithelial-mesenchymal interactions. Tissue Eng. 2010;16:327–342. doi: 10.1089/ten.TEA.2009.0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Kim J.Y., Xin X., Moioli E.K., et al. Regeneration of dental-pulp-like tissue by chemotaxis-induced cell homing. Tissue Eng. 2010;16:3023–3031. doi: 10.1089/ten.TEA.2010.0181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Suzuki T., Lee C.H., Chen M., et al. Induced migration of dental pulp stem cells for in vivo pulp regeneration. J. Dent. Res. 2011;90:1013–1018. doi: 10.1177/0022034511408426. [DOI] [PubMed] [Google Scholar]
  • 222.Srisuwan T., Tilkorn D.J., Al-Benna S., et al. Revascularization and tissue regeneration of an empty root canal space is enhanced by a direct blood supply and stem cells. Dent. Traumatol. 2013;29:84–91. doi: 10.1111/j.1600-9657.2012.01136.x. [DOI] [PubMed] [Google Scholar]
  • 223.Murakami M., Horibe H., Iohara K., et al. The use of granulocyte-colony stimulating factor induced mobilization for isolation of dental pulp stem cells with high regenerative potential. Biomaterials. 2013;34:9036–9047. doi: 10.1016/j.biomaterials.2013.08.011. [DOI] [PubMed] [Google Scholar]
  • 224.Rosa V., Zhang Z., Grande R.H., Nor J.E. Dental pulp tissue engineering in full-length human root canals. J. Dent. Res. 2013;92:970–975. doi: 10.1177/0022034513505772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Horibe H., Murakami M., Iohara K., et al. Isolation of a stable subpopulation of mobilized dental pulp stem cells (MDPSCs) with high proliferation, migration, and regeneration potential is independent of age. PLoS One. 2014;9 doi: 10.1371/journal.pone.0098553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Nagy M.M., Tawfik H.E., Hashem A.A., Abu-Seida A.M. Regenerative potential of immature permanent teeth with necrotic pulps after different regenerative protocols. J. Endod. 2014;40:192–198. doi: 10.1016/j.joen.2013.10.027. [DOI] [PubMed] [Google Scholar]
  • 227.Souron J.B., Petiet A., Decup F., et al. Pulp cell tracking by radionuclide imaging for dental tissue engineering. Tissue Eng. C Methods. 2014;20:188–197. doi: 10.1089/ten.TEC.2013.0148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Takeuchi N., Hayashi Y., Murakami M., et al. 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. doi: 10.1111/odi.12227. [DOI] [PubMed] [Google Scholar]
  • 229.Zhang L.X., Shen L.L., Ge S.H., et al. Systemic BMSC homing in the regeneration of pulp-like tissue and the enhancing effect of stromal cell-derived factor-1 on BMSC homing. Int. J. Clin. Exp. Pathol. 2015;8:10261–10271. [PMC free article] [PubMed] [Google Scholar]
  • 230.Li L., Wang Z. PDGF-BB, NGF and BDNF enhance pulp-like tissue regeneration via cell homing. RSC Adv. 2016;6:109519–110952. doi: 10.1039/C6RA20290J. [DOI] [Google Scholar]
  • 231.Khayat A., Monteiro N., Smith E.E., et al. GelMA-encapsulated hDPSCs and HUVECs for dental pulp regeneration. J. Dent. Res. 2017;96:192–199. doi: 10.1177/0022034516682005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Zhang X., Li H., Sun J., et al. Cell-derived micro-environment helps dental pulp stem cells promote dental pulp regeneration. Cell Prolif. 2017;50:5. doi: 10.1111/cpr.12361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Zhu X., Liu J., Yu Z., et al. A miniature swine model for stem cell-based de novo regeneration of dental pulp and dentin-like tissue. Tissue Eng. C Methods. 2018;24:108–120. doi: 10.1089/ten.tec.2017.0342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Mittal N., Parashar V. Regenerative evaluation of immature roots using PRF and artificial scaffolds in necrotic permanent teeth: a clinical study. J. Contemp. Dent. Pract. 2019;20:720–726. [PubMed] [Google Scholar]
  • 235.Washio A., Teshima H., Yokota K., et al. Preparation of gelatin hydrogel sponges incorporating bioactive glasses capable for the controlled release of fibroblast growth factor-2. J. Biomater. Sci. Polym. Ed. 2019;30:49–63. doi: 10.1080/09205063.2018.1544474. [DOI] [PubMed] [Google Scholar]
  • 236.Zhang S., Yang Y., Jia S., et al. Exosome-like vesicles derived from Hertwig's epithelial root sheath cells promote the regeneration of dentin-pulp tissue. Theranostics10. 2020:5914–5931. doi: 10.7150/thno.43156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.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. doi: 10.4103/ijdr.ijdr_253_21. [DOI] [PubMed] [Google Scholar]
  • 238.Li X., Ma C., Xie X., et al. Pulp regeneration in a full-length human tooth root using a hierarchical nanofibrous microsphere system. Acta Biomater. 2016;35:57–67. doi: 10.1016/j.actbio.2016.02.040. [DOI] [PubMed] [Google Scholar]
  • 239.Yang T., Zhang Q., Xie L., et al. hDPSC-laden GelMA microspheres fabricated using electrostatic microdroplet method for endodontic regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2021;121 doi: 10.1016/j.msec.2020.111850. [DOI] [PubMed] [Google Scholar]
  • 240.Machado M., Stafuzza T.C., Vitor L.L.R., et al. Pulp repair response after the use of a dentin-pulp biostimulation membrane (BBio) in primary teeth: study protocol for a randomized clinical trial. Trials. 2020;21:874. doi: 10.1186/s13063-020-04785-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Fujiwara S., Kumabe S., Iwai Y. Isolated rat dental pulp cell culture and transplantation with an alginate scaffold. Okajimas Folia Anat. Jpn. 2006;83:15–24. doi: 10.2535/ofaj.83.15. [DOI] [PubMed] [Google Scholar]
  • 242.Kumabe S., Nakatsuka M., Kim G.S., et al. Human dental pulp cell culture and cell transplantation with an alginate scaffold. Okajimas Folia Anat. Jpn. 2006;82:147–155. doi: 10.2535/ofaj.82.147. [DOI] [PubMed] [Google Scholar]
  • 243.Orhan E.O., Maden M., Senguuven B. Odontoblast-like cell numbers and reparative dentine thickness after direct pulp capping with platelet-rich plasma and enamel matrix derivative: a histomorphometric evaluation. Int. Endod. J. 2012;45:317–325. doi: 10.1111/j.1365-2591.2011.01977.x. [DOI] [PubMed] [Google Scholar]
  • 244.Matsumoto N., Minakami M., Hatakeyama J J., et al. Histologic evaluation of the effects of Emdogain gel on injured root apex in rats. J. Endod. 2014;40:1989–1994. doi: 10.1016/j.joen.2014.08.024. [DOI] [PubMed] [Google Scholar]
  • 245.Oliva-Rodriguez R., Perez-Urizar J., Dibildox-Alvarado E., et al. Design of a controlled release system of OP-1 and TGF-beta1 based in microparticles of sodium alginate and release characterization by HPLC-UV. In Vitro Cell. Dev. Biol. Anim. 2011;47:681–688. doi: 10.1007/s11626-011-9459-7. [DOI] [PubMed] [Google Scholar]
  • 246.Verma P., Nosrat A., Kim J.R., et al. Effect of residual bacteria on the outcome of pulp regeneration in vivo. J. Dent. Res. 2017;96:100–106. doi: 10.1177/0022034516671499. [DOI] [PubMed] [Google Scholar]
  • 247.Shrestha S., Kishen A. Temporal-controlled bioactive molecules releasing core-shell nano-system for tissue engineering strategies in endodontics. Nanomedicine. 2019;18:11–20. doi: 10.1016/j.nano.2019.02.013. [DOI] [PubMed] [Google Scholar]
  • 248.Abbas K.F., Tawfik H., Hashem A.A.R., et al. Histopathological evaluation of different regenerative protocols using Chitosan-based formulations for management of immature non-vital teeth with apical periodontitis: in vivo study. Aust. Endod. J. 2020;46:405–414. doi: 10.1111/aej.12426. [DOI] [PubMed] [Google Scholar]
  • 249.Soares D.G., Bordini E.A.F., Bronze-Uhle E.S., et al. Chitosan-calcium-simvastatin scaffold as an inductive cell-free platform. J. Dent. Res. 2021;100:1118–1126. doi: 10.1177/00220345211024207. [DOI] [PubMed] [Google Scholar]
  • 250.Goncalves da Costa Sousa M., Conceicao de Almeida G., Martins Mota D.C., et al. Antibiofilm and immunomodulatory resorbable nanofibrous filing for dental pulp regenerative procedures. Bioact. Mater. 2022;16:173–186. doi: 10.1016/j.bioactmat.2022.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Zhu N., Chatzistavrou X., Papagerakis P., et al. Silver-doped bioactive glass/chitosan hydrogel with potential application in dental pulp repair. ACS Biomater. Sci. Eng. 2019;5:4624–4633. doi: 10.1021/acsbiomaterials.9b00811. [DOI] [PubMed] [Google Scholar]
  • 252.Moreira M.S., Sarra G., Carvalho G.L., et al. Physical and biological properties of a chitosan hydrogel scaffold associated to photobiomodulation therapy for dental pulp regeneration: an in vitro and in vivo study. BioMed Res. Int. 2021;2021 doi: 10.1155/2021/6684667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Jittapiromsak N., Sahawat D., Banlunara W W., et al. Acemannan, an extracted product from Aloe vera, stimulates dental pulp cell proliferation, differentiation, mineralization, and dentin formation. Tissue Eng. 2010;16:1997–2006. doi: 10.1089/ten.TEA.2009.0593. [DOI] [PubMed] [Google Scholar]
  • 254.Songsiripradubboon S., Kladkaew S., Trairatvorakul C., et al. Stimulation of dentin regeneration by using acemannan in teeth with lipopolysaccharide-induced pulp inflammation. J. Endod. 2017;43:1097–1103. doi: 10.1016/j.joen.2017.01.037. [DOI] [PubMed] [Google Scholar]
  • 255.Tan L., Wang J., Yin S., et al. Regeneration of dentin–pulp-like tissue using an injectable tissue engineering technique. RSC Adv. 2015;5:59723–59737. doi: 10.1039/C5RA06481C. [DOI] [Google Scholar]
  • 256.Silva C.R., Babo P.S., Gulino M., et al. Injectable and tunable hyaluronic acid hydrogels releasing chemotactic and angiogenic growth factors for endodontic regeneration. Acta Biomater. 2018;77:155–171. doi: 10.1016/j.actbio.2018.07.035. [DOI] [PubMed] [Google Scholar]
  • 257.Widbiller M., Driesen R.B., Eidt A., et al. Cell homing for pulp tissue engineering with endogenous dentin matrix proteins. J. Endod. 2018;44:956–962. doi: 10.1016/j.joen.2018.02.011. [DOI] [PubMed] [Google Scholar]
  • 258.Galler K.M., Cavender A.C., Koeklue U., et al. Bioengineering of dental stem cells in a PEGylated fibrin gel. Regen. Med. 2011;6:191–200. doi: 10.2217/rme.11.3. [DOI] [PubMed] [Google Scholar]
  • 259.Ruangsawasdi N., Zehnder M., Patcas R., et al. Regenerative dentistry: animal model for regenerative Endodontology. Transfus. Med. Hemotherapy. 2016;43:359–364. doi: 10.1159/000447644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Ruangsawasdi N., Zehnder M., Patcas R., et al. Effects of stem cell factor on cell homing during functional pulp regeneration in human immature teeth. Tissue Eng. 2017;23:115–123. doi: 10.1089/ten.TEA.2016.0227. [DOI] [PubMed] [Google Scholar]
  • 261.Galler K.M., Brandl F.P., Kirchhof S., et al. Suitability of different natural and synthetic biomaterials for dental pulp tissue engineering. Tissue Eng. 2018;24:234–244. doi: 10.1089/ten.TEA.2016.0555. [DOI] [PubMed] [Google Scholar]
  • 262.Nagata J.Y., Gomes B.P., Rocha Lima T.F., et al. Traumatized immature teeth treated with 2 protocols of pulp revascularization. J. Endod. 2014;40:606–612. doi: 10.1016/j.joen.2014.01.032. [DOI] [PubMed] [Google Scholar]
  • 263.Tran Hle B., Doan V.N. Human dental pulp stem cells cultured onto dentin derived scaffold can regenerate dentin-like tissue in vivo. Cell Tissue Bank. 2015;16:559–568. doi: 10.1007/s10561-015-9503-z. [DOI] [PubMed] [Google Scholar]
  • 264.Hu L., Gao Z., Xu J., et al. Decellularized swine dental pulp as a bioscaffold for pulp regeneration. BioMed Res. Int. 2017;2017 doi: 10.1155/2017/9342714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Huang C.C., Narayanan R., Warshawsky N., Ravindran S. Dual ECM biomimetic scaffolds for dental pulp regenerative applications. Front. Physiol. 2018;9:495. doi: 10.3389/fphys.2018.00495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Bakhtiar H., Ashoori A., Rajabi S., et al. Human amniotic membrane extracellular matrix scaffold for dental pulp regeneration in vitro and in vivo. Int. Endod. J. 2022;55:374–390. doi: 10.1111/iej.13675. [DOI] [PubMed] [Google Scholar]
  • 267.Holiel A.A., Mahmoud E.M., Abdel-Fattah W.M., Kawana K.Y. Histological evaluation of the regenerative potential of a novel treated dentin matrix hydrogel in direct pulp capping. Clin. Oral Invest. 2021;25:2101–2112. doi: 10.1007/s00784-020-03521-z. [DOI] [PubMed] [Google Scholar]
  • 268.Yang J.W., Zhang Y.F., Wan C.Y., et al. Autophagy in SDF-1alpha-mediated DPSC migration and pulp regeneration. Biomaterials. 2015;44:11–23. doi: 10.1016/j.biomaterials.2014.12.006. [DOI] [PubMed] [Google Scholar]
  • 269.Yang J.W., Zhang Y.F., Sun Z.Y., et al. Dental pulp tissue engineering with bFGF-incorporated silk fibroin scaffolds. J. Biomater. Appl. 2015;30:221–229. doi: 10.1177/0885328215577296. [DOI] [PubMed] [Google Scholar]
  • 270.Pagliarin C.M., Londero Cde L., Felippe M.C., et al. Tissue characterization following revascularization of immature dog teeth using different disinfection pastes. Braz. Oral Res. 2016;30:1. doi: 10.1590/1807-3107BOR-2016.vol30.0074. [DOI] [PubMed] [Google Scholar]
  • 271.Li X., Wang L., Su Q., Ye L., et al. Potential roles of bone morphogenetic protein 9 in the odontogenic differentiation of dental pulp cells. J. Endod. 2021;47:436–443. doi: 10.1016/j.joen.2020.10.018. [DOI] [PubMed] [Google Scholar]
  • 272.Buurma B., Gu K., Rutherford R.B. Transplantation of human pulpal and gingival fibroblasts attached to synthetic scaffolds. Eur. J. Oral Sci. 1999;107:282–289. doi: 10.1046/j.0909-8836.1999.eos107408.x. [DOI] [PubMed] [Google Scholar]
  • 273.Duailibi M.T., Duailibi S.E., Young C.S., et al. Bioengineered teeth from cultured rat tooth bud cells. J. Dent. Res. 2004;83:523–528. doi: 10.1177/154405910408300703. [DOI] [PubMed] [Google Scholar]
  • 274.Young C.S., Terada S., Vacanti J.P., et al. Tissue engineering of complex tooth structures on biodegradable polymer scaffolds. J. Dent. Res. 2002;81:695–700. doi: 10.1177/154405910208101008. [DOI] [PubMed] [Google Scholar]
  • 275.Cordeiro M.M., Dong Z., Kaneko T., et al. Dental pulp tissue engineering with stem cells from exfoliated deciduous teeth. J. Endod. 2008;34:962–969. doi: 10.1016/j.joen.2008.04.009. https://doiorg/10.1016/j.joen.2008.04.009 [DOI] [PubMed] [Google Scholar]
  • 276.Casagrande L., Demarco F.F., Zhang Z., et al. Dentin-derived BMP-2 and odontoblast differentiation. J. Dent. Res. 2010;89:603–608. doi: 10.1177/0022034510364487. [DOI] [PubMed] [Google Scholar]
  • 277.Demarco F.F., Casagrande L., Zhang Z., et al. Effects of morphogen and scaffold porogen on the differentiation of dental pulp stem cells. J. Endod. 2010;36:1805–1811. doi: 10.1016/j.joen.2010.08.031. [DOI] [PubMed] [Google Scholar]
  • 278.Sakai V.T., Zhang Z., Dong Z., et al. SHED differentiate into functional odontoblasts and endothelium. J. Dent. Res. 2010;89:791–796. doi: 10.1177/0022034510368647. [DOI] [PubMed] [Google Scholar]
  • 279.Wang J., Liu X., Jin X., et al. The odontogenic differentiation of human dental pulp stem cells on nanofibrous poly(L-lactic acid) scaffolds in vitro and in vivo. Acta Biomater. 2010;6:3856–3863. doi: 10.1016/j.actbio.2010.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Wang J., Ma H., Jin X., et al. The effect of scaffold architecture on odontogenic differentiation of human dental pulp stem cells. Biomaterials. 2011;32:7822–7830. doi: 10.1016/j.biomaterials.2011.04.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Wang W., Dang M., Zhang Z., et al. Dentin regeneration by stem cells of apical papilla on injectable nanofibrous microspheres and stimulated by controlled BMP-2 release. Acta Biomater. 2016;36:63–72. doi: 10.1016/j.actbio.2016.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Zhang Z., Nor F., Oh M., et al. Wnt/beta-Catenin signaling determines the vasculogenic fate of postnatal mesenchymal stem cells. Stem Cell. 2016;34:1576–1587. doi: 10.1002/stem.2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Ito T., Kaneko T., Sueyama Y., et al. Dental pulp tissue engineering of pulpotomized rat molars with bone marrow mesenchymal stem cells. Odontology. 2017;105:392–397. doi: 10.1007/s10266-016-0283-0. [DOI] [PubMed] [Google Scholar]
  • 284.Silva G.O., Zhang Z., Cucco C., et al. Lipoprotein receptor-related protein 6 signaling is necessary for vasculogenic differentiation of human dental pulp stem cells. J. Endod. 2017;43:S25–S30. doi: 10.1016/j.joen.2017.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Sueyama Y., Kaneko T., Ito T., et al. Implantation of endothelial cells with mesenchymal stem cells accelerates dental pulp tissue regeneration/healing in pulpotomized rat molars. J. Endod. 2017;43:943–948. doi: 10.1016/j.joen.2017.01.035. [DOI] [PubMed] [Google Scholar]
  • 286.Kaneko T., Sone P.P., Zaw S.Y.M., et al. In vivo fate of bone marrow mesenchymal stem cells implanted into rat pulpotomized molars. Stem Cell Res. 2019;38 doi: 10.1016/j.scr.2019.101457. [DOI] [PubMed] [Google Scholar]
  • 287.Zaw Z.C.T., Kawashima N., Kaneko T., Okiji T. Angiogenesis during coronal pulp regeneration using rat dental pulp cells: neovascularization in rat molars in vivo and proangiogenic dental pulp cell-endothelial cell interactions in vitro. J. Dent. Sci. 2022;17:1160–1168. doi: 10.1016/j.jds.2022.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Zhang Z., Warner K.A., Mantesso A., Nor J.E. PDGF-BB signaling via PDGFR-beta regulates the maturation of blood vessels generated upon vasculogenic differentiation of dental pulp stem cells. Front. Cell Dev. Biol. 2022;10 doi: 10.3389/fcell.2022.977725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Kuang R., Zhang Z., Jin X., et al. Nanofibrous spongy microspheres enhance odontogenic differentiation of human dental pulp stem cells. Adv. Healthc. Mater. 2015;4:1993–2000. doi: 10.1002/adhm.201500308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Kuang R., Zhang Z., Jin X., et al. Nanofibrous spongy microspheres for the delivery of hypoxia-primed human dental pulp stem cells to regenerate vascularized dental pulp. Acta Biomater. 2016;33:225–234. doi: 10.1016/j.actbio.2016.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Huang G.T., Yamaza T., Shea L.D., et al. Stem/progenitor cell-mediated de novo regeneration of dental pulp with newly deposited continuous layer of dentin in an in vivo model. Tissue Eng. 2010;16:605–615. doi: 10.1089/ten.TEA.2009.0518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Zheng L., Yang F., Shen H., et al. The effect of composition of calcium phosphate composite scaffolds on the formation of tooth tissue from human dental pulp stem cells. Biomaterials. 2011;32:7053–7059. doi: 10.1016/j.biomaterials.2011.06.004. [DOI] [PubMed] [Google Scholar]
  • 293.Sun H.H., Chen B., Zhu Q.L., et al. Investigation of dental pulp stem cells isolated from discarded human teeth extracted due to aggressive periodontitis. Biomaterials. 2014;35:9459–9472. doi: 10.1016/j.biomaterials.2014.08.003. [DOI] [PubMed] [Google Scholar]
  • 294.Li Y., He L., Pan S., et al. Three-dimensional simulated microgravity culture improves the proliferation and odontogenic differentiation of dental pulp stem cell in PLGA scaffolds implanted in mice. Mol. Med. Rep. 2017;15:873–878. doi: 10.3892/mmr.2016.6042. [DOI] [PubMed] [Google Scholar]
  • 295.Shamma R.N., Elkasabgy N.A., Mahmoud A.A., et al. Design of novel injectable in-situ forming scaffolds for non-surgical treatment of periapical lesions: in-vitro and in-vivo evaluation. Int. J. Pharm. 2017;521:306–317. doi: 10.1016/j.ijpharm.2017.02.058. [DOI] [PubMed] [Google Scholar]
  • 296.Kim K., Lee C.H., Kim B.K., Mao J.J. Anatomically shaped tooth and periodontal regeneration by cell homing. J. Dent. Res. 2010;89:842–847. doi: 10.1177/0022034510370803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Lee W., Oh J.H., Park J.C., et al. Performance of electrospun poly(epsilon-caprolactone) fiber meshes used with mineral trioxide aggregates in a pulp capping procedure. Acta Biomater. 2012;8:2986–2995. doi: 10.1016/j.actbio.2012.04.032. [DOI] [PubMed] [Google Scholar]
  • 298.Eap S., Becavin T., Keller L., et al. Nanofibers implant functionalized by neural growth factor as a strategy to innervate a bioengineered tooth. Adv. Healthc. Mater. 2014;3:386–391. doi: 10.1002/adhm.201300281. [DOI] [PubMed] [Google Scholar]
  • 299.Yoo Y.J., Oh J.H., Zhang Q., et al. Dimethyloxalylglycine-embedded poly(epsilon-caprolactone) fiber meshes promote odontoblastic differentiation of human dental pulp-derived cells. J. Endod. 2018;44:98–103. doi: 10.1016/j.joen.2017.09.002. [DOI] [PubMed] [Google Scholar]
  • 300.Galler K.M., Hartgerink J.D., Cavender A.C., et al. A customized self-assembling peptide hydrogel for dental pulp tissue engineering. Tissue Eng. 2012;18:176–184. doi: 10.1089/ten.TEA.2011.0222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Dissanayaka W.L., Hargreaves K.M., Jin L., et al. The interplay of dental pulp stem cells and endothelial cells in an injectable peptide hydrogel on angiogenesis and pulp regeneration in vivo. Tissue Eng. 2015;21:550–563. doi: 10.1089/ten.TEA.2014.0154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Mangione F., EzEldeen M., Bardet C., et al. Implanted dental pulp cells fail to induce regeneration in partial pulpotomies. J. Dent. Res. 2017;96:1406–1413. doi: 10.1177/0022034517725523. [DOI] [PubMed] [Google Scholar]
  • 303.Moon C.Y., Nam O.H., Kim M., et al. Effects of the nitric oxide releasing biomimetic nanomatrix gel on pulp-dentin regeneration: pilot study. PLoS One. 2018;13 doi: 10.1371/journal.pone.0205534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Zhu L., Dissanayaka W.L., Zhang C. Dental pulp stem cells overexpressing stromal-derived factor-1alpha and vascular endothelial growth factor in dental pulp regeneration. Clin. Oral Invest. 2019;23:2497–2509. doi: 10.1007/s00784-018-2699-0. [DOI] [PubMed] [Google Scholar]
  • 305.Xia K., Chen Z., Chen J., et al. RGD- and VEGF-mimetic peptide epitope-functionalized self-assembling peptide hydrogels promote dentin-pulp complex regeneration. Int. J. Nanomed. 2020;15:6631–6647. doi: 10.2147/IJN.S253576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Kobayashi Y., Nouet J., Baljinnyam E., et al. iPSC-derived cranial neural crest-like cells can replicate dental pulp tissue with the aid of angiogenic hydrogel. Bioact. Mater. 2022;14:290–301. doi: 10.1016/j.bioactmat.2021.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Rodriguez-Benitez S., Stambolsky Guelfand C., Martin-Jimenez M., Segura-Egea J.J. Root canal disinfection of immature dog teeth with apical periodontitis: comparison of three different protocols. J. Clin. Exp. Dent. 2014;6:e357–e363. doi: 10.4317/jced.51475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Neelamurthy P.S., Kumar R.A., Balakrishnan V., et al. Revascularization in immature and mature teeth with necrotic pulp: a clinical study. J. Contemp. Dent. Pract. 2018;19:1393–1399. [PubMed] [Google Scholar]
  • 309.Yadlapati M., Biguetti C., Cavalla F., et al. Characterization of a vascular endothelial growth factor-loaded bioresorbable delivery system for pulp regeneration. J. Endod. 2017;43:77–83. doi: 10.1016/j.joen.2016.09.022. [DOI] [PubMed] [Google Scholar]
  • 310.Bottino M.C., Albuquerque M.T.P., Azabi A., et al. A novel patient-specific three-dimensional drug delivery construct for regenerative endodontics. J. Biomed. Mater. Res. B Appl. Biomater. 2019;107:1576–1586. doi: 10.1002/jbm.b.34250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Itoh Y., Sasaki J.I., Hashimoto M., et al. Pulp regeneration by 3-dimensional dental pulp stem cell constructs. J. Dent. Res. 2018;97:1137–1143. doi: 10.1177/0022034518772260. [DOI] [PubMed] [Google Scholar]
  • 312.Kalyan K., Vinay C., Arunbhupathi, et al. Preclinical evaluation and clinical trial of chlorhexidine polymer scaffold for vital pulp therapy. J. Clin. Pediatr. Dent. 2019;43:109–115. doi: 10.17796/1053-4625-43.2.7. [DOI] [PubMed] [Google Scholar]
  • 313.Xiao M., Qiu J., Kuang R., et al. Synergistic effects of stromal cell-derived factor-1alpha and bone morphogenetic protein-2 treatment on odontogenic differentiation of human stem cells from apical papilla cultured in the VitroGel 3D system. Cell Tissue Res. 2019;378:207–220. doi: 10.1007/s00441-019-03045-3. [DOI] [PubMed] [Google Scholar]
  • 314.Peres M.A., Macpherson L.M.D., Weyant R.J., et al. Oral diseases: a global public health challenge. Lancet. 2019;394:249–260. doi: 10.1016/S0140-6736(19)31146-8. [DOI] [PubMed] [Google Scholar]
  • 315.World Health Organization https://www.who.int/publications/i/item/WHO-NMH-NHD-17.12 WHO/NMH/NHD/17.12 (2017) Sugars and Decay. accessed.
  • 316.The New York Times . 2011. A Closer Look at Teeth.https://www.nytimes.com/2011/11/29/health/a-closer-look-at-teeth-may-mean-more-fillings-by-dentists.html accessed. [Google Scholar]
  • 317.Neves V.C., Babb R., Chandrasekaran D., Sharpe P.T. Promotion of natural tooth repair by small molecule GSK3 antagonists. Sci. Rep. 2017;7 doi: 10.1038/srep39654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.2017. https://eur-lex.europa.eu/eli/reg/2017/746/2017-05-05 Regulation (EU) 2017/746 of the European Parliament. accessed.
  • 319.Directive 2001/83/EC of the European Parliament. 2021. https://eur-lex.europa.eu/legal-content/en/TXT/?uri=CELEX%3A32001L0083 accessed. [Google Scholar]
  • 320.van Rooij E., Purcell A.L., Levin A.A. The art of microRNA research. Circ. Res. 2011;108:219–234. doi: 10.1161/CIRCRESAHA.110.227496. [DOI] [PubMed] [Google Scholar]
  • 321.Van Norman G.A. Drugs, devices, and the FDA: part 1: an overview of approval processes for drugs. JACC Basic Transl. Sci. 2016;1:170–179. doi: 10.1016/j.jacbts.2016.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Fargen K.M., Frei D., Fiorella D., et al. The FDA approval process for medical devices: an inherently flawed system or a valuable pathway for innovation? J. Neurointerventional Surg. 2013;5:269–275. doi: 10.1136/neurintsurg-2012-010400. [DOI] [PubMed] [Google Scholar]
  • 323.Wong C.H., Siah K.W., Lo Aw A.W. Estimation of clinical trial success rates and related parameters. Biostatistics. 2019;20:273–286. doi: 10.1093/biostatistics/kxx069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Koh Jing Jie A., Hussein M., Rao G.G., et al. Drug repurposing approaches towards defeating multidrug-resistant gram-negative pathogens: novel polymyxin/non-antibiotic combinations. Pathogens. 2022;11:1420. doi: 10.3390/pathogens11121420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Birjandi A.A., Suzano F.R., Sharpe P.T. Drug repurposing in dentistry; towards application of small molecules in dentin repair. Int. J. Mol. Sci. 2020;21:6394. doi: 10.3390/ijms21176394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Alaohali A., Brauer D.S., Gentleman E., Sharpe P.T. A modified glass ionomer cement to mediate dentine repair. Dent. Mater. 2021;37:1307–1315. doi: 10.1016/j.dental.2021.05.003. [DOI] [PubMed] [Google Scholar]
  • 327.do Nascimento A.B., Fontana U.F., Teixeira H.M., Costa C.A. Biocompatibility of a resin-modified glass-ionomer cement applied as pulp capping in human teeth. Am. J. Dent. 2000;13:28–34. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The authors do not have permission to share data.


Articles from Bioactive Materials are provided here courtesy of KeAi Publishing

RESOURCES