Skip to main content
  • Systematic Review
  • Open access
  • Published:

Influence of extracellular matrix scaffolds on histological outcomes of regenerative endodontics in experimental animal models: a systematic review

Abstract

Background

Decellularized extracellular matrix (dECM) from several tissue sources has been proposed as a promising alternative to conventional scaffolds used in regenerative endodontic procedures (REPs). This systematic review aimed to evaluate the histological outcomes of studies utilizing dECM-derived scaffolds for REPs and to analyse the contributing factors that might influence the nature of regenerated tissues.

Methods

The PRISMA 2020 guidelines were used. A search of articles published until April 2024 was conducted in Google Scholar, Scopus, PubMed and Web of Science databases. Additional records were manually searched in major endodontic journals. Original articles including histological results of dECM in REPs and in-vivo studies were included while reviews, in-vitro studies and clinical trials were excluded. The quality assessment of the included studies was analysed using the ARRIVE guidelines. Risk of Bias assessment was done using the (SYRCLE) risk of bias tool.

Results

Out of the 387 studies obtained, 17 studies were included for analysis. In most studies, when used as scaffolds with or without exogenous cells, dECM showed the potential to enhance angiogenesis, dentinogenesis and to regenerate pulp-like and dentin-like tissues. However, the included studies showed heterogeneity of decellularization methods, animal models, scaffold source, form and delivery, as well as high risk of bias and average quality of evidence.

Discussion

Decellularized ECM-derived scaffolds could offer a potential off-the-shelf scaffold for dentin-pulp regeneration in REPs. However, due to the methodological heterogeneity and the average quality of the studies included in this review, the overall effectiveness of decellularized ECM-derived scaffolds is still unclear. More standardized preclinical research is needed as well as well-constructed clinical trials to prove the efficacy of these scaffolds for clinical translation.

Other

The protocol was registered in PROSPERO database #CRD42023433026. This review was funded by the Science, Technology and Innovation Funding Authority (STDF) under grant number (44426).

Peer Review reports

Background

The current research focus in the field of regenerative endodontics is to reach more "predictable" outcomes regarding the success rates of this treatment modality as well as the nature of the regenerated tissues which should ideally mimic the native dentin-pulp complex [1]. Regenerative Endodontic Procedures (REPs) require three critical elements including elimination of residual bacteria and bacterial antigens, a scaffold to support and promote stem cell adhesion and proliferation, and coronal seal using a biocompatible material [2, 3].

These procedures generally have two main approaches, the cell-based approach which involves the transplantation of stem cells from an exogenous source, and the cell homing approach which aims to harness the body's own response acting on endogenous residing progenitor cells and factors bypassing the need for exogenous delivery [4, 5]. While cell-based strategies have been advocated and have demonstrated appealing results, the true value of the transplanted cells has been debatable, and their presence does not always result in predictable tissue regeneration [6]. An area of considerable interest in REPs is the scaffold type and the interplay between scaffolds and mesenchymal stem cells [7]. Synthetic scaffolds have many advantageous biological properties such as biocompatibility, controlled biodegradability and mild inflammatory response [8]. Additionally, their mechanical properties, viscosity, porosity, degradation, and releasing rates of incorporated biomolecules can be tailored [3, 9]. However, being synthetic, they lack the biochemical information that might be physiologically intrinsic in native tissues [8]. Recently, the use of naturally derived scaffolds has gained attention as they offer a more cost-effective biomimetic alternative to synthetic materials [10]. Such natural scaffolds, with innate bioactive capacities, provide structural and biochemical support for surrounding cells and can be considered a promising cell homing approach that has the ability to enhance cell recruitment, proliferation, release of signalling molecules from the surrounding microenvironment as well as stimulating cell differentiation [3, 11]. Natural endogenous scaffolds in the form of intracanal blood clot or other blood-derivatives have shown many favourable clinical outcomes such as resolution of clinical signs and symptoms, bone healing and continued root development [12,13,14]. However, several studies have reported unpredictable pattern and architecture of newly formed tissues following the use of the blood clot as a scaffold to revascularize an empty canal [15, 16].

Currently, endodontic literature focuses on the development of scaffolds that can be employed to regulate inflammation and immunologic reactions [11, 17, 18]. This has become emphasized as more studies employing regenerative endodontic procedures for mature permanent necrotic teeth are being published [19,20,21,22].

One of the recent alternatives to the blood clot in REPs is the use of decellularized extracellular matrix (dECM)-derived scaffolds [23]. Native tissue extracellular matrix (ECM) is an excellent source for the fabrication of scaffolds due to its ability to mimic the optimal natural environment for tissue regeneration [24]. Extracellular matrix contains structural proteins and growth factors that can orchestrate cellular proliferation, migration and differentiation [25]. Mild decellularization protocols can maintain ECM structure and biocompatibility whilst eliminating the cells and nucleic acids that can cause an immune reaction [24]. Indeed, dECM has been developed as a biologic scaffold for tissue engineering applications in the field of regenerative medicine. Decellularized ECM-derived scaffolds has been shown to facilitate the constructive remodelling of many different tissues in both preclinical animal studies and in human clinical applications [24]. The source from which these scaffold materials is derived includes a variety of tissues, such as heart valves [26], blood vessels [27], skin [28], skeletal muscle [29], ligaments [30], small intestinal submucosa (SIS) [31], urinary bladder [32] and liver [33]. Decellularized ECM-derived scaffolds have shown considerable success when used in vascular grafts [34], skin grafts [35] and in whole-organ regeneration [36].

Hypothetically, providing the dynamic and structural complexity of ECM will result in a microenvironment favourable for lineage specific differentiation of transplanted/recruited stem cells. Recently, decellularized ECM has been investigated as a promising scaffold for regenerative endodontic procedures [23]. However, the clinical feasibility of using decellularized ECM-derived scaffolds in regenerative endodontics is unclear regarding the source of dECM, appropriate decellularization protocol, sterilization and method of scaffold delivery. These unresolved clinical hurdles could further complicate the steps of REPs and increase the cost of treatment. Most importantly, current histological evidence following the use of dECM-derived scaffolds in REPs is scarce [23].

To address these gaps, a systematic review of the literature was conducted aiming to evaluate the role of dECM scaffolds in dentin-pulp regeneration and the potential contributing factors that could influence regenerative outcomes.

Methodology

Protocol and registration

The protocol for this review was registered in The International Prospective Register of Systematic Reviews (PROSPERO) as (CRD42023433026) and conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocol (PRISMA-P) 2020 statement [37].

Focused question

The research question was formulated as: Can decellularized ECM-derived scaffolds influence the histological outcome of regenerative endodontic applications regarding the nature and pattern of tissues?

  • Participants/population: Animal (ectopic, semi-orthotopic or orthotopic) model/ human teeth

  • Intervention: Decellularized ECM-derived scaffolds in regenerative endodontics.

  • Comparison: non-decellularized ECM-derived scaffolds in regenerative endodontics.

  • Outcome: Nature and pattern of regenerated tissues.

Search strategy

  • In June 2023, an electronic search was conducted on PubMed, Scopus, Google Scholar and Web of Science databases as well as a manual search in major endodontic journals (Journal of Endodontics and International Endodontic Journal). The search was updated April, 1st, 2024. The search strategy used a combination of keywords and Medical Subject Heading (MeSH) terms associated with the Boolean operators ‘AND’ and ‘OR’ as shown in Supplementary file 1.

  • The pool of studies was further enriched by conducting electronic search in the major endodontic journals, including Journal of Endodontics and International Endodontic Journal to search for articles that were not found in databases.

  • Articles retrieved from the search strategy were imported into Endnote X8 software (Thomson Reuters) for duplicate removal.

Study selection

Inclusion criteria

  • Original articles published until April 1st, 2023.

  • Studies reporting the histological and immunohistochemical results of scaffolds in regenerative endodontic applications.

  • Studies in all languages.

Exclusion criteria

  • Review studies.

  • In vitro studies.

  • Ex-vivo studies.

  • Ongoing trials.

  • Studies not including decellularized ECM-based scaffold in their methodology.

The articles identified were screened independently by two reviewers (H.E. and R.E.) for eligibility. Any disagreements were resolved by discussion with a third reviewer (A.A.).

Data extraction

Data were extracted from eligible articles using pre-designed data extraction tables (Microsoft Word) by two independent reviewers (H.E. and R.E.). Any disagreements were solved by discussion among them. Only the data related to in vivo experiments of the included studies were extracted for analysis. Also, in studies assessing both periodontal tissues and supporting structures regeneration as well as dentin pulp regeneration, data were extracted only for the latter. Extracted data included:

  • Characteristics of animal experiments in the included studies: Author, year of publication, animal host, experimental model, sample size, study groups, duration of experiment, evaluation of decellularization and method(s) of histological assessment (Table 1).

  • Methods of scaffold characterization, study findings and potential contributing factors to histological outcomes (Table 2).

Table 1 Characteristics of animal experiments in the included studies
Table 2 Scaffold characterization and potential contributing factors to histological outcomes

List of abbreviations mentioned in the tables is included in supplementary file 6.

Reporting quality assessment [54]

Assessment of the reporting quality for all studies included in this systematic review was performed using the Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines [54] (H.E. and R.E.). In order to evaluate the 21 items, a modified scoring system [55] was used as follows: all subitems were reported “Yes” = 2 points, not all subitems were reported “Unclear (Uc)” = 1 point, or all subitems were not reported “No” = 0 points. Afterwards, quality coefficient (QC) was generated by calculating the sum of all the points obtained for each study divided by 42 “the maximum possible points per study”. The quality of study was reported as “Excellent” when QC was 0.8–1, “Average” if QC was 0.5–0.8 and “Poor” if QC was < 0.5.

Risk of bias assessment [56]

Assessment of bias for all studies included in this systematic review was performed using SYstematic Review Centre for Laboratory animal Experimentation (SYRCLE) risk of bias (RoB) tool [56] (H.E. and R.E.). A modified scoring system [57] was used according to the total number of “yes” answers to the assigned questions (Yes = 1 point) The degree of bias was calculated as follows: High risk: 0–4, Moderate risk: 5–7 and Low risk: 8–10.

Results

Study selection

The records from the different databases were combined (n = 384) then duplicates were removed (n = 96). Then additional records identified through other sources were added (n = 3). For the initial screening of the imported records (n = 291), the titles of the papers were identified and decided whether they were relevant to the topic or not (n = 184). Reviews were excluded (n = 57) and editorials/books were excluded (n = 3). For the final screening, the abstracts of all relevant articles (n = 47) were then carefully appraised to identify eligible studies. Full texts of the relevant articles were screened. During this final screening phase, studies were excluded as: in vitro studies (n = 7), ex-vivo studies (n = 1), and studies not including dECM (n = 22). Excluded studies by full text screening (n = 30) and reasons for exclusion are listed in supplementary file 2. Total number of studies included in this systematic review was 17 studies (n = 17). The flow chart for search process and study inclusion are presented in (Fig. 1) according to PRISMA 2020 guidelines [37].

Fig. 1
figure 1

PRISMA flow diagram showing inclusion and screening process of included records

Study characteristics

Different animal models were used in the included studies. The majority used mice [38, 39, 41, 42, 45, 46, 48, 50, 51, 53] followed by rats [43, 44, 49, 52], pigs [39, 40] and beagle dogs [23, 47]. The main source of ECM used was swine [23, 39, 41, 47, 53], human [46, 48, 50] or bovine dental pulp [43,44,45]. Others used cell-generated ECM [38, 42], swine tooth bud [40], human amniotic membrane [49, 52] and rats’ submandibular glands [51]. Regarding the study model, scaffolds were ectopically transplanted in six studies [38, 39, 43, 44, 46, 52], semi-orthotopically transplanted in twelve studies [39,40,41,42, 45, 47,48,49,50,51,52,53] and orthotopically transplanted in just one study [23]. In terms of the regenerative approach, two thirds of the included studies used cell-seeded scaffolds [39,40,41,42, 45, 46, 48,49,50,51,52,53] while the rest used cell-free scaffolds [23, 38, 43, 44, 47]. Six of the former studies also compared between the two approaches [40, 45, 48, 49, 51, 52]. As for the scaffold form, this was either tissue sheets placed within either treated dentin matrix TDM [39, 42, 47, 51, 53] /tooth slices [41, 45, 48], or freeze-dried sponges [43, 49] or sheets [23, 44, 51], powder [46], hydrogel microspheres [50], injectable hydrogel [52, 53] or whole decellularized tooth bud [40]. Only four studies used crosslinked scaffolds [43, 49, 50, 52]. Regarding terminal sterilization, scaffolds were either immersed in penicillin/streptomycin [39, 41, 46, 47, 51, 53], or sterilized using ethylene oxide (EtO] [23, 50] or peracetic acid (PAA) [45]. The rest of studies did not report the method of sterilization [38, 40, 42,43,44, 48, 49, 52]. Interestingly, the concentration of ECM components was reported by only six studies [23, 43, 49, 50, 52, 53]. Regarding the methods of in vivo outcome assessment, only histological evaluation was performed in two studies [44, 46], while histological and immunohistochemical analysis were done in the rest of the studies. When ECM scaffolds were compared to other non-ECM-derived scaffolds, regeneration of both pulp-like and dentin-like tissues were reported in the ECM groups in eight studies [23, 38,39,40,41,42, 47, 50] while pulp-like tissue was solely reported in five studies [44, 46, 49, 52, 53]. Regarding the cell homing approach versus cell transplantation approach, it was noted that in cell-seeded scaffolds, pulp-like and dentin-like tissue regeneration was more likely to be detected compared to cell-free scaffolds [39,40,41,42, 46, 48, 50, 51]. Conversely, quality of pulp-like tissues in cell-seeded group was reported, in one study, to be less optimal that in cell-free groups [52].

Reporting quality assessment

All studies were analysed using Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines. Study design was described by all studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. However, none of the studies provided precalculated sample size [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. Inclusion and exclusion criteria were not properly described in any of the studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. Randomization was carried out in only three studies [45, 49, 52]. Blinding was only carried out in four studies [43,44,45, 52]. Outcome measures and experimental animals’ details were adequately described by all studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. Statistical analysis of in vivo outcomes was not performed in ten studies [23, 28, 38, 40, 42, 44, 46, 47, 52, 53] Experimental steps were unclear in all studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. Results were adequately described with statistical evaluation of each group in only five studies [41, 43, 45, 48, 51]. Data on the animal model and in vivo outcomes were properly described in abstracts in only seven studies [23, 39, 40, 45,46,47, 52, 53]. The introduction section provided adequate background on the topic in all studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53], however, none of them explained the rationale of using a specific animal model and its relevance to human biology. In all studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53], objectives were clearly described. In one study [44], the ethical statement was not provided. Housing details and animal care were reported in only three studies [23, 40, 41]. Adequate interpretation of results as well as study protocol registration were done in all studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. In one study [38], the generalizability of outcomes and clinical implications of experimental results were not stated. Data access statement was mentioned in only six studies [23, 47, 48, 50, 51, 53]. While declaration of interest statement was mentioned in all studies except for two [38, 39]. Overall, quality coefficient ranged between 0.5–0.7 indicating an “Average” grade of all the reviewed studies. The assessment criteria and their results are listed in Table 3.

Table 3 Quality assessment of included studies using Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines

Risk of bias (RoB) assessment

All studies were analysed using SYstematic Review Centre for Laboratory animal Experimentation (SYRCLE) risk of bias (RoB) tool [56]. Regarding selection bias, in all studies, allocation sequence was unclear [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. However, baseline characteristics of animals were similar among different groups in all studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. Only three studies reported concealment of sample allocation [40, 49, 52]. Investigators were blinded in only two studies [44, 45]. Outcome assessors were blinded in four studies [44, 45, 49, 52]. Random allocation of animals in study groups, random selection of animals for assessment, and addressing incomplete outcome data, were mentioned in none of the included studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. Regarding reporting bias, there was no selective reporting of outcomes in any of the studies [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. All of the reviewed studies showed high risk of bias where twelve studies scored 2/10 in RoB assessment [23, 38,39,40,41,42,43, 46,47,48, 50,51,52,53] while the remaining three studies [44, 45, 49] scored 4/10. The overall risk of bias was high in all studies. The risk of bias assessment criteria and their results are listed in Table 4.

Table 4 Risk of bias assessment of included studies using SYRCLE risk of bias tool for animal studies

Synthesis of results

Qualitative analysis of the studies was performed; however, meta-analysis was not feasible due to wide variations amongst studies in methods for assessment, decellularization protocols, animal models used, and nature and source of decellularized ECM.

Discussion

The field of regenerative endodontics has recently been challenged by a propensity of evidence that demonstrates the difficulty to regenerate true dentin-pulp tissue using clinically relevant scenarios. Currently, it is not clear that any of the conventional protocols of REPs are likely to recapitulate, histologically, the morphological and physiological characteristics of native endodontic tissues [16, 58].

The role of the scaffold has recently changed from being a passive carrier to being a bioactive material with tailored properties for guided regeneration of specific tissues. For an engineered tissue, an ideal scaffold material should mimic the physiological and physical nature of the extracellular matrix of the native target tissue [59]. The dentin-pulp organ is a unique entity with a complex microenvironment.

Normal pulp extracellular matrix (ECM) is a non-mineralized tissue, however, pulp cells express a cocktail of growth factors and cytokines with certain ratios that can regulate mineralization in response to external stimuli [60]. Pulp ECM is predominantly composed of structural proteins in the form of type I and type III collagen (Col-I and Col-III). Other specialized proteins (primarily fibronectin and laminin) and glycosaminoglycans (such as chondroitin sulphate and hyaluronic acid), are also found [61]. The dental pulp serves as a source of nutrition and sensation to dentin. Moreover, it has its own defensive and reparative functions [4].

To date, natural and synthetic biomaterials that can mimic dentin-pulp ECM to reproduce all its complex characteristics are still not available [62]. Therefore it was suggested that decellularized extracellular matrix (ECM)-derived scaffolds could offer a natural biomimetic alternative to conventional scaffolds [23]. Owing to its rich content of tissue-specific growth factors and chemical cues, ECM not only supports cell functions but also dictates cells’ commitment and guides their differentiation lineage [24]. Although many ECM components such as hyaluronic acid [63] and collagen [64] have been tested clinically, whole-tissue decellularized ECM has not been yet accepted for clinical use in regenerative endodontics [23]. It is thus imperative for clinicians and investigators to analyse, via a systematic approach, the variables observed in the preclinical protocols for dECM scaffolding techniques and their effect on the outcomes of regenerative endodontics.

Therefore, the present systematic review was conducted with the aim of studying the role of decellularized ECM-derived scaffolds in dentin-pulp regeneration. However, due to the high risk of bias and average quality of the evidence of the included studies, a meta-analysis was not feasible.

Histological outcome of using dECM-derived scaffolds for regenerative endodontic applications

The unpredictable regeneration of tissues following conventional regenerative endodontic procedures remains a major concern [65]. Several histological studies using blood-derived scaffolds have reported evidence of ectopic tissue regeneration [16, 65, 66]. Bone-like, cementum-like tissues and absence of odontoblast-like layer are often observed within regenerated tissues [67, 68]. Regenerated tissues appear to be mineralized counterparts of a mixture of tissues such as dentin, bone, cementum and periodontal tissues which have been collectively termed dentin associated mineralized tissues (DAMT) owing to their deposition and close association to the original inner canal wall dentin [69].

Results of this review indicate that decellularized ECM-derived scaffolds are novel biomimetic materials that can lead to enhanced angiogenesis and regeneration of dentin-pulp-like tissues compared to non-ECM-derived scaffolds [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51]. Notably, the included studies that employed decellularized ECM-derived scaffolds, used other naturally-derived scaffolds as controls rather than synthetic scaffolds [23, 38,39,40,41,42,43,44,45,46,47,48,49,50,51]. It was noted that thirteen of reviewed studies used IHC analysis to detect the expression levels of odontogenic/angiogenic markers. This method of evaluation is valuable for distinguishing the nature and quality of newly-formed tissues. The regenerated tissues were characterized by high expression of DSP and DPP, increased collagen deposition and neovascularization as reported by Ravidnran et al. [38] and Huang et al. [42] who both used cell-generated dECM scaffolds. These results were also in accordance with the findings of Chen et al. [39], Zhang et al. [40], Hu et al. [41], Alqahtani et al. [23] and Zheng et al. [50] who all used porcine dental pulp ECM-derived scaffolds. On the contrary, bone/cementum-like hard tissue formation was reported in the study by Fu et al. [47] in the acellular ECM group compared to high DSPP and DMP-1 expression in the laminin-coated ECM group. Formation of bone-like tissue was also observed by Bakhtiar et al. [43] when crosslinked bovine pulp dECM scaffold was used. This also coincides with the findings of Kim et al. [48] who reported hard tissue formation not resembling dentin when human dental pulp dECM was the scaffold, although they reported expression of some odontogenic markers such as DSPP and DMP-1 in the newly formed tissue. Enhanced angiogenesis and detection of pulp-like tissue were reported by Bakhtiar et al. [44, 49, 52] when either bovine pulp dECM or amniotic membrane dECM were used as scaffolds.

Generally, in the reviewed studies, it was evident that cell-seeded dECM-derived scaffolds triggered more organized tissue regeneration and robust neovascularization compared to cell-free scaffolds [51] except for one study [52]. However, the fact that those studies have used ectopic or semi-orthotopic models, cannot be overlooked. As the placement of the scaffold out of its original microenvironment may have had a significant effect on host cell recruitment and their behaviour [70]. Indeed, in the study by Alqahtani et al. [23], evidence of dentin-pulp regeneration was reported when the cell-homing approach was used in an orthotopic model which indicates the importance of providing the appropriate “niche”.

Potential contributing factors to histological outcomes

Influence of decellularization, lyophilization and terminal sterilization protocols

When dECM-derived scaffold is prepared, it must be mechanically separated from unwanted tissue structures, decellularized, often dehydrated or lyophilized then terminally sterilized [24, 71]. Each of these processing steps can alter the integrity and composition of the matrix [24].

In this review, studies that used treated dentin matrix (TDM) as the main scaffold were not included as it is not considered a decellularized tissue but rather a demineralized tissue [72]. Decellularization can be described as a procedure that aims to remove cellular contents of a tissue, leaving the extracellular matrix free of antigens as well as preserving its original three-dimensional biostructure [24].

A prominent finding to be highlighted in the included records, is the heterogeneity of decellularization and sterilization protocols. Remnants of chemicals and/or enzymes used during decellularization may result in residual cytotoxicity to host cells [24]. It was reported by Bakhtiar et al. [44] that SDS-free protocol was more biocompatible compared to SDS-containing protocols. Moreover, higher retention of GAGs and collagen contents was observed when SDS was not used [44]. Other factors to be considered, are lyophilization of the scaffolds and terminal sterilization using ethylene oxide (EtO); these steps were found to affect tissue architecture and decrease growth factor contents of dECM [23]. It can be suggested to use other methods of sterilization such as immersion in penicillin/streptomycin or peracetic acid for better preservation of native biological and structural integrity of dECM [73].

Influence of the animal model on regenerative outcomes

Another factor to be considered is the animal host used in each study. The natural response to tissue injury i.e., wound healing cascades, involves a complex sequence of events that includes vascular, cellular and humoral components; the outcome of which is either tissue necrosis and scarring or reconstruction of the tissue with return of function [74]. Decellularized ECM-derived scaffolds can modulate the wound healing response toward constructive remodelling rather than tissue destruction and scar formation [24, 71].

The use of small animal models in most of the reviewed studies might have influenced the regenerative outcomes, as the host response could differ from one species to another. Small animals are often used due to their ease of handling and economical value. Nevertheless, larger animals will eventually be needed to test conditions that highly mimic those in humans, especially regarding working inside the root canal space [75]. Large animals that have dental anatomy and tooth size comparable to humans allow for clinical simulation and evaluation of orthotopic pulp regeneration [75]. Additionally, large animal models can allow the simulation of either short or long-standing infections within the root canal space eventually leading to periapical disease [75]. Such models are critical to be employed since it has been shown that the structure of dentin previously exposed to bacterial biofilms is altered which may negatively affect the migration, attachment, proliferation and differentiation of recruited stem and progenitor cells. Furthermore, infected dentin may sequester variable levels of growth factors as compared to natural healthy dentin [76]. This is in addition to the fact that it has been shown that the larger the periapical lesion and the longer the infection, the higher the virulence of microorganisms thereby creating a more challenging environment for tissue regeneration [77]. The more severe the infection is, the more is the residual inflammatory response and hence this has been shown to have detrimental effects on regenerated tissues following REPs in animal models [78]. Such challenging conditions require the use of more effective antimicrobial strategies which in turn may further negatively affect the regenerative process [77]. The issue of selecting an appropriate model for REPs is indeed a major issue for translational research especially in the tissue engineering and regenerative medicine field.

In the current review, only one study simulated the clinical protocol for REPs [23]. It is worth mentioning that in this study, the root canals were not infected and there were no periapical lesions present prior to treatment which does not truly replicate the clinical scenario [23]. In fact, none of the included studies have used an infected model in their methodologies. Future studies should focus on not only optimizing and assessing the regenerative potential of the scaffold, but also work towards using decellularized ECM-derived scaffolds in animal models that can truly represent translational research as well as in well-executed randomized clinical trials. Moreover, future animal and clinical studies should evaluate the efficacy of dECM-derived scaffolds in the presence of microbial challenge.

Influence of scaffold source on regenerative outcome

The alignment and organization of collagen fibres and the concentration of functional proteins are dependent on the native function of the source tissue from which the ECM is derived [24, 71]. Consequently, if dentin/pulp regeneration is the case, the use of dECM of dental origin would be a more suitable choice.

Decellularized extracellular matrix derived from various human or animal tissues has been considered as a possible scaffolding medium for tissue regeneration in present studies.

Most of the reviewed studies used decellularized dental pulp-derived ECM rather than other tissues of dental and non-dental origin. However, none of the studies compared between the tissue sources (dental and non-dental) regarding their biocompatibility and their biological influence on regenerated tissues. Moreover, none of them evaluated the difference between human and xenogeneic sources of ECM scaffolds regarding their in vitro characterization and in vivo regenerative potential. Using an autologous scaffold from discarded third molars or teeth extracted for orthodontic reasons would avoid the problems of antigenicity. However, it will not be feasible in most clinical situations, hence using a xenogeneic form of ECM may be more clinically applicable [71]. Indeed, one of the main concerns of using such scaffolds, could be their possible antigenicity. However, several studies reported that following careful decellularization protocols, minimal immunological response was reported [43, 44, 47, 50, 52]. In studies assessing immunogenicity of dECM scaffolds, no systemic toxicity on major organs and no immune rejection from host tissues were detected, regardless of the source of dECM [43, 44, 47, 50, 52]. This could also be attributed to the immunomodulatory effect of dECM shifing the polarisation of the macrophage population from M1 pro-inflammatory to M2 anti-inflammatory phenotypes resulting in constructive remodelling [71, 79].

Influence of scaffold form and method of delivery

Incorporating other materials with decellularized ECM to make composite scaffolds appears to have more predictable results. In the study by Fu et al. [47], laminin-modified dECM resulted in regeneration of dentin-like tissue compared to cementum/bone-like tissue in laminin-free dECM group. likewise, dECM incorporating gelatine methacrylate (GelMA) microspheres, resulted in dentin-pulp-like tissue regeneration [50]. However, these favourable outcomes may not be only caused by the direct effect of the added materials, but also due to the modified “physical properties” of the composite scaffold [80]. It has been proven that physical characteristics of the scaffold such as the degree of stiffness and modulus of elasticity can have indirect influence on the commitment of recruited cells and nature of regenerated tissues [80,81,82]. Regeneration of organized pulp-like tissue was also reported by Bakhtiar et al. [52] following the use of cross-linked hydrogel scaffolds. It is worth mentioning that only three studies used the scaffolds as injectable hydrogel form [50, 52, 53]. Future studies should focus on optimizing the form of delivery of the scaffold, i.e., using hydrogels or biphasic scaffolds that might be more logical in regenerative endodontic applications.

Loading dECM with growth factors could also potentiate the action of dECM scaffolds. Addition of BMP4 in the study by Tan et al. [46] resulted in organized pulp-like tissue formation. However, this might not be as important in the actual clinical protocol for regenerative endodontic procedures (REPs) when EDTA or other chelating agents are used to release the sequestered growth factors from dentin [83, 84].

Limitations and future perspectives

In the reviewed studies, there were significant limitations that may have affected outcome assessment. None of the studies compared between dECM-derived scaffolds and blood derived-scaffolds currently used in REPs. There was methodological heterogeneity in the histological assessment and interpretation of results. Additionally, the duration of studies ranged from 2 to 12 weeks in most of the included studies which may not be sufficient time to assess scaffold remodelling and long-term efficacy of the scaffold on dentin-pulp regeneration. Although all included studies had both in vitro and in vivo phases, we only extracted data that was relevant to the in vivo experiments and histological outcomes. The quality and risk of bias assessments indicated that most of reviewed records are preliminary studies lacking sufficient sample size, randomization, blinding and proper statistical analyses. Other limitations of this systematic review include the small number of available studies.

Future studies should work towards optimizing decellularization and sterilization protocols with maximum preservation of ECM architecture and innate growth factors content. Moreover, evaluating the effect of adding natural or synthetic modifiers to the dECM-derived scaffolds should be also addressed, with the purpose of improving specific mechanical characteristics, such as stiffness, viscoelasticity and biodegradation. Additionally, preclinical and animal models should mimic the clinical protocol of REPs with sufficient long-term follow up. The preparation of freeze-dried hydrogel form that can be stored as an off-the-shelf product could also aid in the clinical translation of using dECM-derived scaffolds in REPs. Until this “optimal” scaffold is available, it is important not to lose sight of clinically-relevant outcomes, namely; patient-centered and clinician-centered, as these continue to be measurable and reproducible.

Conclusions

Decellularized ECM-derived scaffolds could offer a promising biomimetic alternative to current scaffolds used for regenerative endodontic procedures. These cell-free scaffolds may provide comparable histological outcomes to their cell-seeded counterparts thereby offering a potential off-the-shelf scaffold for dentin-pulp regeneration. However, due to the heterogeneity of decellularization methods, animal models, scaffold source, form and delivery, as well as the high risk of bias and average quality of the studies included in this review, the overall effectiveness of decellularized ECM-derived scaffolds still remains uncertain. Therefore, more standardized preclinical research is needed as well as well-constructed clinical trials to prove the efficacy of these scaffolds for clinical translation of organized and functional dentin-pulp regeneration.

Availability of data and materials

Data is provided within the manuscript or supplementary information files.

References

  1. Kim S, Malek M, Sigurdsson A, Lin L, Kahler B. Regenerative endodontics: a comprehensive review. Int Endod J. 2018;51(12):1367–88.

    Article  CAS  PubMed  Google Scholar 

  2. Diogenes A, Henry MA, Teixeira FB, Hargreaves KM. An update on clinical regenerative endodontics. Endod Top. 2013;28(1):2–23.

    Article  Google Scholar 

  3. Elnawam H, Abdelmougod M, Mobarak A, Hussein M, Aboualmakarem H, Girgis M, et al. Regenerative endodontics and minimally invasive dentistry: intertwining paths crossing over into clinical translation. Front Bioeng Biotechnol. 2022;10:837639.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Murray PE, Garcia-Godoy F, Hargreaves KM. Regenerative endodontics: a review of current status and a call for action. J Endod. 2007;33(4):377–90.

    Article  PubMed  Google Scholar 

  5. Ahmed GM, Abouauf EA, AbuBakr N, Fouad AM, Dörfer CE, Fawzy El-Sayed KM. Cell-based transplantation versus cell homing approaches for pulp-dentin complex regeneration. Stem cells international. 2021;2021:1–23.

    CAS  Google Scholar 

  6. Mangione F, Ezeldeen M, Bardet C, Lesieur J, Bonneau M, Decup F, et al. Implanted dental pulp cells fail to induce regeneration in partial pulpotomies. J Dent Res. 2017;96(12):1406–13.

    Article  CAS  PubMed  Google Scholar 

  7. Yu J, Shi J, Jin Y. Current approaches and challenges in making a bio-tooth. Tissue Eng Part B Rev. 2008;14(3):307–19.

    Article  CAS  PubMed  Google Scholar 

  8. Ulery BD, Nair LS, Laurencin CT. Biomedical applications of biodegradable polymers. J Polym Sci Part B Polym Phys. 2011;49(12):832–64.

    Article  CAS  Google Scholar 

  9. Athanasiou KA, Niederauer GG, Agrawal CM. Sterilization, toxicity, biocompatibility and clinical applications of polylactic acid/polyglycolic acid copolymers. Biomaterials. 1996;17(2):93–102.

    Article  CAS  PubMed  Google Scholar 

  10. Bansal R, Bansal R. Regenerative endodontics: a state of the art. Indian J Dent Res. 2011;22(1):122.

    Article  PubMed  Google Scholar 

  11. Moussa DG, Aparicio C. Present and future of tissue engineering scaffolds for dentin-pulp complex regeneration. J Tissue Eng Regen Med. 2019;13(1):58–75.

    CAS  PubMed  Google Scholar 

  12. Murray PE. Platelet-rich plasma and platelet-rich fibrin can induce apical closure more frequently than blood-clot revascularization for the regeneration of immature permanent teeth: a meta-analysis of clinical efficacy. Front Bioeng Biotechnol. 2018;6:139.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Diogenes A, Ruparel NB. Regenerative endodontic procedures: clinical outcomes. Dental Clinics. 2017;61(1):111–25.

    PubMed  Google Scholar 

  14. Yang F, Sheng K, Yu L, Wang J. Does the use of different scaffolds have an impact on the therapeutic efficacy of regenerative endodontic procedures? A systematic evaluation and meta-analysis. BMC Oral Health. 2024;24(1):1–13.

    Article  Google Scholar 

  15. Dianat O, Mashhadi Abas F, Paymanpour P, Eghbal MJ, Haddadpour S, Bahrololumi N. Endodontic repair in immature dogs’ teeth with apical periodontitis: blood clot vs plasma rich in growth factors scaffold. Dent Traumatol. 2017;33(2):84–90.

    Article  CAS  PubMed  Google Scholar 

  16. Astudillo-Ortiz E, Babo PS, Sunde PT, Galler KM, Gomez-Florit M, Gomes ME. Endodontic tissue regeneration: a review for tissue engineers and dentists. Tissue Eng Part B Rev. 2023;29(5):491–513.

    Article  CAS  PubMed  Google Scholar 

  17. Siddiqui Z, Sarkar B, Kim K-K, Kadincesme N, Paul R, Kumar A, et al. Angiogenic hydrogels for dental pulp revascularization. Acta Biomater. 2021;126:109–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Romaldini A, Ulivi V, Nardini M, Mastrogiacomo M, Cancedda R, Descalzi F. Platelet lysate inhibits NF kappa B activation and induces proliferation and an alert state in quiescent human umbilical vein endothelial cells retaining their differentiation capability. Cells. 2019;8(4):331.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Paryani K, Kim SG. Regenerative endodontic treatment of permanent teeth after completion of root development: a report of 2 cases. J Endod. 2013;39(7):929–34.

    Article  PubMed  Google Scholar 

  20. Saoud TM, Martin G, Chen YHM, Chen KL, Chen CA, Songtrakul K, et al. Treatment of mature permanent teeth with necrotic pulps and apical periodontitis using regenerative endodontic procedures: a case series. J Endod. 2016;42(1):57–65.

    Article  PubMed  Google Scholar 

  21. Nangia D, Saini A, Sharma S, Kumar V, Chawla A, Perumal V, et al. Treatment outcome of regenerative endodontic procedures in mature permanent teeth compared to nonsurgical endodontic treatment: A systematic review and meta-analysis. J Conserv Dent. 2021;24(6):530.

    Article  PubMed  Google Scholar 

  22. El-Kateb NM, El-Backly RN, Amin WM, Abdalla AM. Quantitative assessment of intracanal regenerated tissues after regenerative endodontic procedures in mature teeth using magnetic resonance imaging: a randomized controlled clinical trial. J Endod. 2020;46(5):563–74.

    Article  PubMed  Google Scholar 

  23. Alqahtani Q, Zaky S, Patil A, Beniash E, Ray H, Sfeir C. Decellularized swine dental pulp tissue for regenerative root canal therapy. J Dent Res. 2018;97(13):1460–7.

    Article  CAS  PubMed  Google Scholar 

  24. Badylak SF, Freytes DO, Gilbert TW. Extracellular matrix as a biological scaffold material: structure and function. Acta Biomater. 2009;5(1):1–13.

    Article  CAS  PubMed  Google Scholar 

  25. Khurshid Z, Alnaim AJA, Alhashim AAA, Imran E, Adanir N. Future of decellularized dental pulp matrix in regenerative endodontics. Eur J Dent. 2022;16:737–41.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Schenke-Layland K, Vasilevski O, Opitz F, König K, Riemann I, Halbhuber K, et al. Impact of decellularization of xenogeneic tissue on extracellular matrix integrity for tissue engineering of heart valves. J Struct Biol. 2003;143(3):201–8.

    Article  CAS  PubMed  Google Scholar 

  27. Conklin B, Richter E, Kreutziger K, Zhong D-S, Chen C. Development and evaluation of a novel decellularized vascular xenograft. Med Eng Phys. 2002;24(3):173–83.

    Article  CAS  PubMed  Google Scholar 

  28. Chen R-N, Ho H-O, Tsai Y-T, Sheu M-T. Process development of an acellular dermal matrix (ADM) for biomedical applications. Biomaterials. 2004;25(13):2679–86.

    Article  CAS  PubMed  Google Scholar 

  29. Borschel GH, Dennis RG, Kuzon WM Jr. Contractile skeletal muscle tissue-engineered on an acellular scaffold. Plast Reconstr Surg. 2004;113(2):595–602.

    Article  PubMed  Google Scholar 

  30. Woods T, Gratzer PF. Effectiveness of three extraction techniques in the development of a decellularized bone–anterior cruciate ligament–bone graft. Biomaterials. 2005;26(35):7339–49.

    Article  CAS  PubMed  Google Scholar 

  31. Badylak SF, Lantz GC, Coffey A, Geddes LA. Small intestinal submucosa as a large diameter vascular graft in the dog. J Surg Res. 1989;47(1):74–80.

    Article  CAS  PubMed  Google Scholar 

  32. Freytes DO, Badylak SF, Webster TJ, Geddes LA, Rundell AE. Biaxial strength of multilaminated extracellular matrix scaffolds. Biomaterials. 2004;25(12):2353–61.

    Article  CAS  PubMed  Google Scholar 

  33. Lin P, Chan WC, Badylak SF, Bhatia SN. Assessing porcine liver-derived biomatrix for hepatic tissue engineering. Tissue Eng. 2004;10(7–8):1046–53.

    Article  CAS  PubMed  Google Scholar 

  34. Kristofik NJ, Qin L, Calabro NE, Dimitrievska S, Li G, Tellides G, et al. Improving in vivo outcomes of decellularized vascular grafts via incorporation of a novel extracellular matrix. Biomaterials. 2017;141:63–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Erdag G, Morgan JR. Allogeneic versus xenogeneic immune reaction to bioengineered skin grafts. Cell Transplant. 2004;13(6):701–12.

    Article  PubMed  Google Scholar 

  36. Song JJ, Ott HC. Organ engineering based on decellularized matrix scaffolds. Trends Mol Med. 2011;17(8):424–32.

    Article  CAS  PubMed  Google Scholar 

  37. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Int J Surg. 2021;88:105906.

    Article  PubMed  Google Scholar 

  38. Ravindran S, George A. Biomimetic extracellular matrix mediated somatic stem cell differentiation: applications in dental pulp tissue regeneration. Front Physiol. 2015;6:118.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Chen G, Chen J, Yang B, Li L, Luo X, Zhang X, 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.

    Article  CAS  PubMed  Google Scholar 

  40. Zhang W, Vazquez B, Oreadi D, Yelick P. Decellularized tooth bud scaffolds for tooth regeneration. J Dent Res. 2017;96(5):516–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hu L, Gao Z, Xu J, Zhu Z, Fan Z, Zhang C, et al. Decellularized swine dental pulp as a bioscaffold for pulp regeneration. BioMed Res Int. 2017;2017:9342714.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Huang C-C, Narayanan R, Warshawsky N, Ravindran S. Dual ECM biomimetic scaffolds for dental pulp regenerative applications. Front Physiol. 2018;9:495.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Bakhtiar H, Pezeshki-Modaress M, Kiaipour Z, Shafiee M, Ellini MR, Mazidi A, et al. Pulp ECM-derived macroporous scaffolds for stimulation of dental-pulp regeneration process. Dent Mater. 2020;36(1):76–87.

    Article  CAS  PubMed  Google Scholar 

  44. Bakhtiar H, Rajabi S, Pezeshki-Modaress M, Ellini MR, Panahinia M, Alijani S, et al. Optimizing methods for bovine dental pulp decellularization. J Endod. 2021;47(1):62–8.

    Article  PubMed  Google Scholar 

  45. Alghutaimel H, Yang X, Drummond B, Nazzal H, Duggal M, Raïf E. Investigating the vascularization capacity of a decellularized dental pulp matrix seeded with human dental pulp stem cells: in vitro and preliminary in vivo evaluations. Int Endod J. 2021;54(8):1300–16.

    Article  CAS  PubMed  Google Scholar 

  46. Tan Q, Cao Y, Zheng X, Peng M, Huang E, Wang J. BMP4-regulated human dental pulp stromal cells promote pulp-like tissue regeneration in a decellularized dental pulp matrix scaffold. Odontology. 2021;109(4):895–903.

    Article  CAS  PubMed  Google Scholar 

  47. Fu J, Chen J, Li W, Yang X, Yang J, Quan H, et al. Laminin-modified dental pulp extracellular matrix for dental pulp regeneration. Front Bioeng Biotechnol. 2021;8:595096.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Kim I-H, Jeon M, Cheon K, Kim SH, Jung H-S, Shin Y, et al. In vivo evaluation of decellularized human tooth scaffold for dental tissue regeneration. Appl Sci. 2021;11(18):8472.

    Article  CAS  PubMed  Google Scholar 

  49. Bakhtiar H, Ashoori A, Rajabi S, Pezeshki-Modaress M, Ayati A, Mousavi MR, et al. Human amniotic membrane extracellular matrix scaffold for dental pulp regeneration in vitro and in vivo. Int Endod J. 2022;55(4):374–90.

    Article  PubMed  Google Scholar 

  50. Zheng L, Liu Y, Jiang L, Wang X, Chen Y, Li L, et al. Injectable decellularized dental pulp matrix-functionalized hydrogel microspheres for endodontic regeneration. Acta Biomater. 2023;156:37–48.

    Article  CAS  PubMed  Google Scholar 

  51. Shi Y, Wang Y, Shan Z, Gao Z. Decellularized rat submandibular gland as an alternative scaffold for dental pulp regeneration. Front Bioeng Biotechnol. 2023;11:1148532.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Bakhtiar H, Mousavi MR, Rajabi S, Pezeshki-Modaress M, Ayati A, Ashoori A, et al. Fabrication and characterization of a novel injectable human amniotic membrane hydrogel for dentin-pulp complex regeneration. Dent Mater. 2023;39(8):718–28.

    Article  CAS  PubMed  Google Scholar 

  53. Yuan S, Yang X, Wang X, Chen J, Tian W, Yang B. Injectable Xenogeneic Dental Pulp Decellularized Extracellular Matrix Hydrogel Promotes Functional Dental Pulp Regeneration. Int J Mol Sci. 2023;24(24):17483.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. du PercieSert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, et al. The ARRIVE guidelines 2.0: Updated guidelines for reporting animal research. J Cereb Blood Flow Metab. 2020;40(9):1769–77.

    Article  Google Scholar 

  55. Garcia-Gonzalez M, Munoz F, Gonzalez-Cantalapiedra A, Lopez-Pena M, Saulacic N. Systematic review and quality evaluation using ARRIVE 2.0 guidelines on animal models used for periosteal distraction osteogenesis. Animals. 2021;11(5):1233.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Hooijmans CR, Rovers MM, De Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE’s risk of bias tool for animal studies. BMC Med Res Methodol. 2014;14:1–9.

    Article  Google Scholar 

  57. Anagnostaki E, Mylona V, Parker S, Lynch E, Grootveld M. Systematic review on the role of lasers in endodontic therapy: valuable adjunct treatment? Dent J. 2020;8(3):63.

    Article  Google Scholar 

  58. Guo X, Li J, Wu Y, Xu L. Recent advancements in hydrogels as novel tissue engineering scaffolds for dental pulp regeneration. Int J Biol Macromol. 2024;264:130708.

    Article  CAS  PubMed  Google Scholar 

  59. Raddall G, Mello I, Leung BM. Biomaterials and scaffold design strategies for regenerative endodontic therapy. Front Bioeng Biotechnol. 2019;7:317.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Cooper PR, Holder MJ, Smith AJ. Inflammation and regeneration in the dentin-pulp complex: a double-edged sword. J Endod. 2014;40(4):S46–51.

    Article  PubMed  Google Scholar 

  61. Goldberg M, Smith AJ. Cells and extracellular matrices of dentin and pulp: a biological basis for repair and tissue engineering. Crit Rev Oral Biol Med. 2004;15(1):13–27.

    Article  PubMed  Google Scholar 

  62. Gathani KM, Raghavendra SS. Scaffolds in regenerative endodontics: a review. Dent Res J. 2016;13(5):379.

    Article  Google Scholar 

  63. Singh H, Rathee K, Kaur A, Miglani N. Pulp regeneration in an immature maxillary central incisor using hyaluronic acid hydrogel. Contemp Clin Dent. 2021;12(1):94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Jiang X, Liu H, Peng C. Clinical and radiographic assessment of the efficacy of a collagen membrane in regenerative endodontics: a randomized, controlled clinical trial. J Endod. 2017;43(9):1465–71.

    Article  PubMed  Google Scholar 

  65. Pulyodan MK, Mohan SP, Valsan D, Divakar N, Moyin S, Thayyil S. Regenerative endodontics: a paradigm shift in clinical endodontics. J Pharm Bioallied Sci. 2020;12(Suppl 1):S20.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Vatankhah M, Najary S, Dianat O. Clinical, Radiographic, and histologic outcomes of regenerative endodontic treatment in human immature teeth using different Biological Scaffolds: a systematic review and Meta-analysis. Curr Stem Cell Res Ther. 2024;19(4):611–27.

    Article  CAS  PubMed  Google Scholar 

  67. Arslan H, Şahin Y, Topçuoğlu HS, Gündoğdu B. Histologic evaluation of regenerated tissues in the pulp spaces of teeth with mature roots at the time of the regenerative endodontic procedures. J Endod. 2019;45(11):1384–9.

    Article  PubMed  Google Scholar 

  68. Zhou R, Wang Y, Chen Y, Chen S, Lyu H, Cai Z, et al. Radiographic, histologic, and biomechanical evaluation of combined application of platelet-rich fibrin with blood clot in regenerative endodontics. J Endod. 2017;43(12):2034–40.

    Article  PubMed  Google Scholar 

  69. Yamauchi N, Nagaoka H, Yamauchi S, Teixeira FB, Miguez P, Yamauchi M. Immunohistological characterization of newly formed tissues after regenerative procedure in immature dog teeth. J Endod. 2011;37(12):1636–41.

    Article  PubMed  Google Scholar 

  70. Diogenes A, Hargreaves KM. Microbial modulation of stem cells and future directions in regenerative endodontics. J Endod. 2017;43(9):S95–101.

    Article  PubMed  Google Scholar 

  71. Badylak SF. Xenogeneic extracellular matrix as a scaffold for tissue reconstruction. Transpl Immunol. 2004;12(3–4):367–77.

    Article  CAS  PubMed  Google Scholar 

  72. Bi F, Zhang Z, Guo W. Treated dentin matrix in tissue regeneration: recent advances. Pharmaceutics. 2023;15(1):91.

    Article  CAS  Google Scholar 

  73. Moradi L, Jobania BM, Jafarnezhad-Ansariha F, Ghorbani F, Esmaeil-Pour R, Zolbina MM, et al. Evaluation of different sterilization methods for decellularized kidney tissue. Tissue Cell. 2020;66:101396.

    Article  CAS  PubMed  Google Scholar 

  74. Tjäderhane L. The mechanism of pulpal wound healing. Aust Endod J. 2002;28(2):68–74.

    Article  PubMed  Google Scholar 

  75. Nakashima M, Iohara K, Bottino MC, Fouad AF, Nör JE, Huang GTJ. Animal models for stem cell-based pulp regeneration: foundation for human clinical applications. Tissue Eng Part B Rev. 2019;25(2):100–13.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Ballal NV, Narkedamalli R, Ruparel NB, Shenoy PA, Bhat VR, Belle VS. Effect of maleic acid root conditioning on release of transforming growth factor beta 1 from infected root canal dentin. J Endod. 2022;48(5):620–4.

    Article  PubMed  Google Scholar 

  77. Kim SG. Infection and pulp regeneration. Dent J. 2016;4(1):4.

    Article  Google Scholar 

  78. Iranmanesh P, Torabinejad M, Saatchi M, Toghraie D, Razavi SM, Khademi A. Effect of duration of root canal infection on the ability of dentin-pulp complex regeneration of immature permanent teeth: an animal study. J Endod. 2022;48(10):1301-7. e2.

    Article  PubMed  Google Scholar 

  79. Zaky SH, Shehabeldin M, Ray H, Sfeir C. The role of inflammation modulation in dental pulp regeneration. Eur Cell Mater. 2021;41:184–93.

    Article  CAS  PubMed  Google Scholar 

  80. Qu T, Jing J, Ren Y, Ma C, Feng JQ, Yu Q, et al. Complete pulpodentin complex regeneration by modulating the stiffness of biomimetic matrix. Acta Biomater. 2015;16:60–70.

    Article  CAS  PubMed  Google Scholar 

  81. Bertassoni LE. Progress and challenges in microengineering the dental pulp vascular microenvironment. J Endod. 2020;46(9):S90–100.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Wu DT, Munguia-Lopez JG, Cho YW, Ma X, Song V, Zhu Z, et al. Polymeric scaffolds for dental, oral, and craniofacial regenerative medicine. Molecules. 2021;26(22):7043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zeng Q, Nguyen S, Zhang H, Chebrolu HP, Alzebdeh D, Badi MA, et al. Release of growth factors into root canal by irrigations in regenerative endodontics. J Endod. 2016;42(12):1760–6.

    Article  PubMed  Google Scholar 

  84. Widbiller M, Eidt A, Hiller K-A, Buchalla W, Schmalz G, Galler K. Ultrasonic activation of irrigants increases growth factor release from human dentine. Clin Oral Invest. 2017;21:879–88.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB). This work is supported by Science, Technology & Innovation Funding Authority (STDF) under grant number (44426).

Author information

Authors and Affiliations

Authors

Contributions

H.E., A.A., and R.E. Conceptualized the work; H.E. and R.E. Developed and validated the search strategy; H.E. and R.E Performed the investigation, applied resources and software for data extraction and analysis; H.E. And R.E. Drafted the manuscript; A.A., S.N., N.M.K., and R.E. Supervised and validated the work; All authors revised, edited and approved the final version of the manuscript before submission.

Corresponding author

Correspondence to Hisham Elnawam.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Elnawam, H., Abdallah, A., Nouh, S. et al. Influence of extracellular matrix scaffolds on histological outcomes of regenerative endodontics in experimental animal models: a systematic review. BMC Oral Health 24, 511 (2024). https://doi.org/10.1186/s12903-024-04266-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12903-024-04266-x

Keywords