Skip to main content

MINI REVIEW article

Front. Physiol., 17 November 2022
Sec. Craniofacial Biology and Dental Research
This article is part of the Research Topic Rising Stars in Craniofacial Biology and Dental Research: 2022 View all 6 articles

Potential of extracellular space for tissue regeneration in dentistry

  • Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, Kings College London, London, United Kingdom

With the proven relationship between oral and general health and the growing aging population, it is pivotal to provide accessible therapeutic approaches to regenerate oral tissues and restore clinical function. However, despite sharing many core concepts with medicine, dentistry has fallen behind the progress in precision medicine and regenerative treatments. Stem cell therapies are a promising avenue for tissue regeneration, however, ethical, safety and cost issues may limit their clinical use. With the significance of paracrine signalling in stem cell and tissue regeneration, extracellular space comprising of the cell secretome, and the extracellular matrix can serve as a potent source for tissue regeneration. Extravesicles are secreted and naturally occurring vesicles with biologically active cargo that can be harvested from the extracellular space. These vesicles have shown great potential as disease biomarkers and can be used in regenerative medicine. As a cell free therapy, secretome and extracellular vesicles can be stored and transferred easily and pose less ethical and safety risks in clinical application. Since there are currently many reviews on the secretome and the biogenesis, characterization and function of extracellular vesicles, here we look at the therapeutic potential of extracellular space to drive oral tissue regeneration and the current state of the field in comparison to regenerative medicine.

Introduction

Medicine is rapidly advancing towards precision therapy, using the latest advancements in biotechnology to provide patient-tailored treatments. In dentistry the emphasis has been placed more on enhancing the quality and aesthetics of dental fillings or impression materials and use of digital technology for diagnosis and rapid chair side treatments. With the growing aging population and the predicted increased number of people aged 65 and over by 2050, the need for a more accessible, efficient and targeted dental treatment is more palpable to regenerate dental tissues and restore clinical function (UN Population Division, 2019).

Functional tooth is comprised of innervated and vascularized dental pulp encased by dentin, cementum, and the harder enamel as the external protective layer. Periodontal or tooth supporting tissue consists of periodontal ligament, gingiva and alveolar bone. Dental caries and trauma often require restoration of the tooth by placement of filling material in the crown or endodontic therapy, to remove and replace dental pulp. Inflammation of tooth supporting tissue results in periodontitis, loss of alveolar bone, tooth mobility and ultimately tooth loss. Conventional treatment replaces the missing tooth with removable dentures, fixed restoration such as crown, bridges, or placement of an implant. All of which aim to replace a biological tissue with metal prosthesis.

Regenerative dentistry aims to replace conventional dental treatments with biologically driven approaches to induce repair of the diseased tissue, reduce inflammation and eliminate scarring. These therapies aim to be minimally invasive and can involve local delivery of active molecules into the region of interest to stimulate tissue regeneration. Regenerative approaches may be relatively easy for some dental tissues such as dental pulp since dental pulp cells can be grown and maintained in culture and subsequently applied in regenerative endodontics (Brizuela et al., 2020; Matoug-Elwerfelli et al., 2020). Similarly, conventional filling materials can be replaced by biological alternatives that stimulate stem cells and promote tissue regeneration in dentin or enamel (Neves et al., 2017; Elsharkawy et al., 2018; Birjandi et al., 2020). Treatment of edentulous patients can ultimately be achieved by replacing the whole tooth organ with bioengineered tooth or transplantation of constructed embryonic tooth primordia into the oral cavity (Yelick and Sharpe, 2019). It is however necessary to consider avenues other than cell-based approaches for regeneration of these tissues.

In regenerative medicine, Bioprinting, iPSCs and patient derived stem cells can be used for tissue repair. Similarly, Nanoparticle and Nanoclay technology are applied for drug delivery (Dutta and Dutta, 2012; Balint et al., 2013; Zhang and Zhang, 2015; Coyle et al., 2016; Dogan et al., 2017; Im, 2018; Hoveizi et al., 2019; Xie et al., 2022). New tools have been developed for minimally invasive surgical approaches, but further improvements are still required for regenerative therapies to become effective for all major diseases. Scaling up the size and speed of manufacturing, reproductivity and reliability for clinical translation are some of the matters that need tackling in regenerative medicine. Additionally, more advanced assays should be developed to better understand the cellular behaviour in these treatments and accurately predict the performance of novel therapeutics in vivo (Tucker et al., 2016). Regenerative dentistry faces additional challenges due to lack of emphasis for biologically driven therapeutics in clinics, strong marketing aspect of the profession and lack of support for clinical trials of regenerative therapies should they pass the preclinical tests.

Regenerative potential of extracellular space

Much work has been done on the development of new materials to recapitulate cellular function and promote tissue regeneration. Autologous or allogenic cell therapies can contribute to tissue regeneration, but the low proliferative potential of differentiated cells and their limited availability has restricted their use. Therefore, although stem cell therapy is a promising avenue for tissue regeneration, issues such as immune compatibility, risk of infection and tumorigenicity have prevented these treatments from becoming the mainstream regenerative approach (Maguire, 2013; Eggenhofer et al., 2014; Jiang et al., 2014; Laurencin and Nair, 2014; Turinetto et al., 2016; Menasché, 2018; Arnold et al., 2019; Zhou et al., 2019; Laurencin and Daneshmandi, 2020; Matai et al., 2020).

For years the focus of developmental biology, the base of regenerative biology, has been on the contents of the cells and the role of intracellular events in cellular behaviour and function. Today, there is an increasing appreciation of the extracellular space as a force to drive cellular function, stemness, homeostasis, healing, survival and motility (Naba et al., 2016; Laura, 2018). Furthermore, it is established that the paracrine effect of stem cells plays a major role in tissue repair and regeneration. This effect is through the extracellular space which is composed of the ECM and the cell secretome.

The ECM is a network of proteoglycans, glycosaminoglycans and proteins arranged in a 3D architecture that provides structural properties and signalling cues whilst serving as the microenvironment that diffuses nutrients. Therefore, ECM and ECM-derived biomaterials are extensively studied for their potential in tissue regeneration and as delivery vehicle for stem cell treatments (Capella-Monsonís et al., 2020). For example, Melatonin-loaded hydrogels show favourable viscoelastic properties and promote accelerated and enhanced quality of bone formation in regeneration of furcation defect in dogs (Abdelrasoul et al., 2022). Specifically, extracellular matrix derived from dental pulp stem cells can promote mineralization of human gingival fibroblasts (Nowwarote et al., 2022). Dual ECM scaffolds derived from pulp and endothelial ECM promote attachments of mesenchymal stem cells and subsequently odontogenic differentiation. Subcutaneous implantation of these scaffolds into tooth root slice model in vivo results in odontogenic differentiation of dental pulp and bone marrow derived MSC (Huang et al., 2018). ECMs are also used in disease modelling and cancer. Indeed, cell derived ECMs are great tools to study cancer microenvironment as they provide a more physiologically relevant phenotype of cancer cells in comparison to conventional 2D and 3D culture models (Serebriiskii et al., 2008; Eberle et al., 2011; Kaukonen et al., 2017; Hoshiba, 2018). This interesting application of ECM has not been tested extensively in oral dysplasia or cancer models such as squamous cell carcinoma. Further understanding and correlation of the composition and biological function of ECM is essential in regenerative approaches of complex oral tissues.

Secretome is composed of the factors that are actively or passively released by the cells. This ranges from cytokines, chemokines, growth factors, lipids to free nucleic acids and extracellular vesicles. The secretome has been shown to have various roles in tissue repair, angiogenesis and immunoregulation (Xia et al., 2019; Willis et al., 2020; Damayanti et al., 2021). Secretome can be easily harvested, freeze dried and transported. As a cell free approach, secretome therapy does not induce the adverse effects of cell therapies such as tumorgenicity and host rejection (Song et al., 2019; Mocchi et al., 2021). An important and biologically active component of the secretome are Extracellular vesicles. Extracellular vesicles (EV) are naturally occurring vesicles with biological cargo that mediate cell-cell communication. The field of extracellular vesicles is constantly evolving with the identification of new groups of nanoparticles based on their size, function and cargo such as exosomes, ectosomes, exomeres, supremeres and migrasomes (Raposo and Stoorvogel, 2013; Cocucci and Meldolesi, 2015; Zhang et al., 2018; Zhang et al., 2021). Some of the regenerative cargo within native, unmodified extracellular vesicles (EVs) are mRNA, miRNA, proteins. Therefore, EVs can be used as biomarkers of different diseases as well as cancer. A recent work has shown that EVs can contribute to cancer aggressiveness (Nigri et al., 2022). Equally EVs can modulate activity of immune cells and remodelling of ECM and subsequently contribute to tissue repair (Silva et al., 2017). As an example extracellular vesicles protect the bone and cartilage from degradation and promote bone regeneration (Cosenza et al., 2017; Jia et al., 2019). Bone marrow stem cells (BMSCs) treated with EVs promote healing of calvarial defect in mice (Li et al., 2020). Similarly, EVs derived from human Placenta MSC and human BMSC promote myelin regeneration and functional recovery (Hu et al., 2016; Clark et al., 2019). EVs can be traced back to their cells of origin and exhibit similar or even enhanced in vivo performance in comparison to their parent cells (Larssen et al., 2017; Chen and Guo, 2020). Extracellular vesicles derived from BMSCs promote liver regeneration with the same efficiency as BMSCs in a mouse model of hepatic ischemia (Anger et al., 2019). However, In a rat model of myocardial infarction, MSC derived EVs performed better than their MSC (Shao et al., 2017). Similarly iPSC-derived cardiomyocyte EVs have enhanced performance in comparison to cell injection in mice (El Harane et al., 2018). The regenerative effect of EVs can even be modified by altering the culture condition of their parent cells (Murphy et al., 2019; Ramasubramanian et al., 2019). Hypoxic condition results in increased biological activity of the cell and subsequently increased regenerative potential of their secreted EV (Hu and Li, 2018). Culturing osteoblasts in the presence of mineralizing supplements results in EVs that are capable of mineralization in MSC cultures. Similarly, priming MSCs derived from adipose tissue with TNF-a results in EV secretion and osteogenic differentiation of human primary osteoblasts (Davies et al., 2017; Lu et al., 2017; Lee and Kim, 2021; Liu and Holmes, 2021; Nagelkerke et al., 2021). EVs can also be bioengineered and enriched with favourable factors such as drugs and growth factors to serve as targeted therapies (Kim et al., 2022). All of these demonstrate the huge potential of EVs for tissue regeneration.

EVs can be derived from dental mesenchymal stem cells (DMSC). These cells have the advantage of enhanced regenerative ability and good accessibility. DMSC can be harvested from multiple sites in the oral cavity such as Dental pulp stem cells, exfoliated deciduous tooth stem cells, apical dental papilla stem cells, periodontal ligament stem cells, dental follicle stem cells, gingiva derived mesenchymal stem cells and tooth germ progenitor cells (Chouaib et al., 2022). Similar to their parent cells, conditioned medium or secretome form dental mesenchymal stem cells (DMSCs) exhibits great regenerative potential and possess greater amount of metabolic and proliferative associated proteins, neutrophils and chemokines in comparison to non-dental conditioned medium (El Moshy et al., 2020). Interestingly, DMSCs has been shown to promote tissue regeneration in liver (Hirata et al., 2016; Matsushita et al., 2017), and in many pathological conditions such as diabetes and neurological disorders (Izumoto-Akita et al., 2015; Jarmalavičiūtė et al., 2015; Mita et al., 2015; Ahmed et al., 2016; Asadi-Golshan et al., 2018; Wang et al., 2019; Chen et al., 2020), Cardiac and pulmonary lesions (Wakayama et al., 2015; Yamaguchi et al., 2015) and alveolar bone defects (Omori et al., 2015; de Cara et al., 2019; Hiraki et al., 2020; Qiu et al., 2020). Despite these discoveries, there are limited number of studies on the characterization and biological function of dental mesenchymal stem cell derived secretome or exosomes in comparison to other sources of EVs in regenerative medicine. Additionally, not all the studies using DMSC secretome have characterized the stemness and differentiation capacities of the EVs and their cell of origin in full detail. Additionally, There is also a degree of variability in factors affecting the cells of origin such as passage number, sorting of the cell and period of conditioning that subsequently affects the secretome and its biological function across different studies (Chouaib et al., 2022).

Challenges in secretome therapy for tissue regeneration

The promising results of EVs and their benefit over cell-based therapeutic approaches has resulted in them being tested in clinical settings and their clinical safety being approved (Escudier et al., 2005). As a cell free approach, secretome and extracellular vesicles do not present the ethical and safety issues associated with conventional cell therapies such as immune compatibility, risk of infection and tumorigenicity. Additionally, they can be easily harvested, stored, transferred and applied. Administration of autologous EV minimises the risk of pathogen transmission and immunological intolerances and this is can be a great advantage in assessment of EV function in clinical setting and phase I or II trials (Beer et al., 2017).

Despite these advantages, the role of EVs as sole therapy still requires further investigation. Identification of the active molecule that exerts the biological function in EVs is extremely challenging. Sensitivity of mass spectrometers should be able to cover the range of low abundance proteins over highly abundant proteins in the secretome and EVs to better characterize them (Brown et al., 2012). The purify of final EV population and their heterogeneous nature is equally important. This is because the contents and therefore biological activity or regenerative capacity of EVs may vary depending on the cell type, experimental setup and purification method. Variation in the existing methods of purification, characterization and storage of cells, their secretome and EVs are some of the other challenges in the field although many of these are currently being addressed.

To bypass some of the limitations of natural occurring EVs and avoid some of the mentioned inconsistencies, EVs can be bioengineered and enriched with favourable factors resulting in enhanced yield and better isolation but the full impact of these modifications on target cells needs to be investigated in detail (Kim et al., 2022). In clinical translation, it is yet to be determined which donor cell source can provide the most potent EV subtypes and which factors influence this (Beer et al., 2017). Mass production of EVs is also challenging. Even the golden standard of EV purification with ultracentrifuge has its own setbacks such as availability of the equipment in all labs, potential of co-isolation of non-vesicle macromolecules with EV and EV aggregation. Ultracentrifugation is also time consuming. However commercial EV purification kits that are currently available are not yet suitable for clinical application. It is therefore important to establish the manufacturing workflow for suitable EV purification according to good manufacturing practice (GMP) and enhance their storage condition and accessibility for clinical application. EV therapeutics are currently categorized as biologicals with active substance but it is still not clear if a step for pathogen reduction would be implemented in mass production of EV and if so, how that step affects the morphology and integrity of EV and subsequently the interaction with target cells and biological activity (Beer et al., 2017).

Majority of currently running EV clinical trials are aimed to monitor circulating EV as biomarkers, disease progression and cancer diagnosis and very few are addressing tissue repair and regeneration. With the proven link between the oral and general health, designing preclinical studies and clinical trials that aim to improve oral health using regenerative approaches is important. The great advantage of cell free approaches over cell-based therapies, makes secretome and EV a very attractive tool for regenerative treatment. Much work must be done to uncover the full potential of secretomics in oral health and to then develop standards for reproducibility, clinical transition and accessibility for public.

Author contributions

AB; Designed and conducted and critically revised the manuscript. PS critically reviewed the manuscript.

Conflict of interest

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

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abdelrasoul M., Abd A., Kotry G., Ramadan O., Essawy M., Kamaldin J., et al. (2022). Regeneration of critical- sized grade II furcation using a novel loaded scaffold. Oral Dis. 15, 1–16. doi:10.1111/odi.14314

PubMed Abstract | CrossRef Full Text | Google Scholar

Ahmed N. E-M. B., Murakami M., Hirose Y., Nakashima M. (2016). Therapeutic potential of dental pulp stem cell secretome for alzheimer’s disease treatment: An in vitro study. Stem Cells Int. 2016–11. doi:10.1155/2016/8102478

PubMed Abstract | CrossRef Full Text | Google Scholar

Anger F., Camara M., Ellinger E., Germer C-T., Schlegel N., Otto C., et al. (2019). Human mesenchymal stromal cell-derived extracellular vesicles improve liver regeneration after ischemia reperfusion injury in mice. Stem Cells Dev. 28 (21), 1451–1462. doi:10.1089/scd.2019.0085

PubMed Abstract | CrossRef Full Text | Google Scholar

Arnold A. M., Holt B. D., Daneshmandi L., Laurencin C. T., Sydlik S. A. (2019). Phosphate graphene as an intrinsically osteoinductive scaffold for stem cell-driven bone regeneration. Proc. Natl. Acad. Sci. U. S. A. 116 (11), 4855–4860. doi:10.1073/pnas.1815434116

PubMed Abstract | CrossRef Full Text | Google Scholar

Asadi-Golshan R., Razban V., Mirzaei E., Rahmanian A., Khajeh S., Mostafavi-Pour Z., et al. (2018). Sensory and motor behavior evidences supporting the usefulness of conditioned medium from dental pulp-derived stem cells in spinal cord injury in rats. Asian Spine J. 12 (5), 785–793. doi:10.31616/asj.2018.12.5.785

PubMed Abstract | CrossRef Full Text | Google Scholar

Balint R., Cassidy N. J., Cartmell S. H. (2013). Electrical stimulation: A novel tool for tissue engineering. Tissue Eng. Part B Rev. 19 (1), 48–57. doi:10.1089/ten.teb.2012.0183

PubMed Abstract | CrossRef Full Text | Google Scholar

Beer L., Mildner M., Ankersmit H. J. (2017). Cell secretome based drug substances in regenerative medicine: When regulatory affairs meet basic science. Ann. Transl. Med. 5 (7), 170–177. doi:10.21037/atm.2017.03.50

PubMed Abstract | CrossRef Full Text | Google Scholar

Birjandi A. A., Suzano F. R., Sharpe P. T. (2020). Drug repurposing in dentistry; towards application of small molecules in dentin repair. Int. J. Mol. Sci. 21, E6394. doi:10.3390/ijms21176394

PubMed Abstract | CrossRef Full Text | Google Scholar

Brizuela C., Meza G., Urrejola D., Quezada M. A., Concha G., Ramírez V., et al. (2020). Cell-based regenerative endodontics for treatment of periapical lesions: A randomized, controlled phase I/II clinical trial. J. Dent. Res. 99 (5), 523–529. doi:10.1177/0022034520913242

PubMed Abstract | CrossRef Full Text | Google Scholar

Brown K. J., Formolo C. A., Seol H., Marathi R. L., Duguez S., An E., et al. (2012). Advances in the proteomic investigation of the cell secretome. Expert Rev. Proteomics 9 (3), 337–345. doi:10.1586/epr.12.21

PubMed Abstract | CrossRef Full Text | Google Scholar

Capella-Monsonís H., De Pieri A., Peixoto R., Korntner S., Zeugolis D. I. (2020). Extracellular matrix-based biomaterials as adipose-derived stem cell delivery vehicles in wound healing: A comparative study between a collagen scaffold and two xenografts. Stem Cell Res. Ther. 11 (1), 510–515. doi:10.1186/s13287-020-02021-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen B., Guo L. (2020). Reply: Exosomes are comparable to source adipose stem cells in fat graft retention with up-regulating early inflammation and angiogenesis. Plast. Reconstr. Surg. 146 (4), 504e–505e. doi:10.1097/PRS.0000000000007202

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen Y-R., Lai P-L., Chien Y., Lee P-H., Lai Y-H., Ma H-I., et al. (2020). Improvement of impaired motor functions by human dental exfoliated deciduous teeth stem cell-derived factors in a rat model of Parkinson’s disease. Int. J. Mol. Sci. 21 (11), 3807. doi:10.3390/ijms21113807

PubMed Abstract | CrossRef Full Text | Google Scholar

Chouaib B., Cuisinier F., Collart-Dutilleul P. Y. (2022). Dental stem cell-conditioned medium for tissue regeneration: Optimization of production and storage. World J. Stem Cells 14, 287–302. doi:10.4252/wjsc.v14.i4.287

PubMed Abstract | CrossRef Full Text | Google Scholar

Clark K., Zhang S., Barthe S., Kumar P., Pivetti C., Kreutzberg N., et al. (2019). Placental mesenchymal stem cell-derived extracellular vesicles promote myelin regeneration in an animal model of multiple sclerosis. Cells 8 (12), 1497. doi:10.3390/cells8121497

PubMed Abstract | CrossRef Full Text | Google Scholar

Cocucci E., Meldolesi J. (2015). Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 25 (6), 364–372. doi:10.1016/j.tcb.2015.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Cosenza S., Ruiz M., Toupet K., Jorgensen C., Noël D. (2017). Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci. Rep. 7 (1), 16214. doi:10.1038/s41598-017-15376-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Coyle R., Jia J., Mei Y. (2016). Polymer microarray technology for stem cell engineering. Acta Biomater. [Internet] 34, 60–72. doi:10.1016/j.actbio.2015.10.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Damayanti R. H., Rusdiana T., Wathoni N. (2021). Mesenchymal stem cell secretome for dermatology application: A review. Clin. Cosmet. Investig. Dermatol. 14, 1401–1412. doi:10.2147/CCID.S331044

PubMed Abstract | CrossRef Full Text | Google Scholar

Davies O. G., Cox S. C., Williams R. L., Tsaroucha D., Dorrepaal R. M., Lewis M. P., et al. (2017). Annexin-enriched osteoblast-derived vesicles act as an extracellular site of mineral nucleation within developing stem cell cultures. Sci. Rep. 7 (1), 12639. doi:10.1038/s41598-017-13027-6

PubMed Abstract | CrossRef Full Text | Google Scholar

de Cara S. P. H. M., Origassa C. S. T., de Sá Silva F., Moreira M. S. N. A., de Almeida D. C., Pedroni A. C. F., et al. (2019). Angiogenic properties of dental pulp stem cells conditioned medium on endothelial cells in vitro and in rodent orthotopic dental pulp regeneration. Heliyon 5 (4), e01560. doi:10.1016/j.heliyon.2019.e01560

PubMed Abstract | CrossRef Full Text | Google Scholar

Decuzzi P., Coclite A., Lee A., Palange A. L., Di Mascolo D., Chiappini C., et al. (2017). Nano-particles for biomedical applications. Nanotechnology, 643–91. doi:10.1007/978-3-662-54357-3_21

CrossRef Full Text | Google Scholar

Dogan A., Eser Elcin A., Murat Elcin Y. (2017). Translational applications of tissue engineering in cardiovascular medicine. Curr. Pharm. Des. 23 (6), 903. doi:10.2174/1381612823666161111141954

PubMed Abstract | CrossRef Full Text | Google Scholar

Dutta R. C., Dutta A. K. (2012). ECM analog technology a simple tool for exploring cell-ECM dynamics. Front. Biosci. E4 (3), 1043–1048. doi:10.2741/E439

PubMed Abstract | CrossRef Full Text | Google Scholar

Eberle K. E., Sansing H. A., Szaniszlo P., Resto V. A., Berrier A. L. (2011). “Carcinoma matrix controls resistance to cisplatin through talin regulation of NF-kB,”. PLoS One. Editor D-Y. Jin, 6, e21496. doi:10.1371/journal.pone.0021496

PubMed Abstract | CrossRef Full Text | Google Scholar

Eggenhofer E., Luk F., Dahlke M. H., Hoogduijn M. J. (2014). The life and fate of mesenchymal stem cells. Front. Immunol. 5. doi:10.3389/fimmu.2014.00148

PubMed Abstract | CrossRef Full Text | Google Scholar

El Harane N., Kervadec A., Bellamy V., Pidial L., Neametalla H. J., Perier M-C., et al. (2018). Acellular therapeutic approach for heart failure: In vitro production of extracellular vesicles from human cardiovascular progenitors. Eur. Heart J. 39 (20), 1835–1847. doi:10.1093/eurheartj/ehy012

PubMed Abstract | CrossRef Full Text | Google Scholar

El Moshy S., Radwan I. A., Rady D., Abbass M. M. S., El-Rashidy A. A., Sadek K. M., et al. (2020). Dental stem cell-derived secretome/conditioned medium: The future for regenerative therapeutic applications. Stem Cells Int. 2020, 7593402–7593429. doi:10.1155/2020/7593402

PubMed Abstract | CrossRef Full Text | Google Scholar

Elsharkawy S., Al-Jawad M., Pantano M. F., Tejeda-Montes E., Mehta K., Jamal H., et al. (2018). Protein disorder–order interplay to guide the growth of hierarchical mineralized structures. Nat. Commun. 9 (1), 2145. doi:10.1038/s41467-018-04319-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Escudier B., Dorval T., Chaput N., André F., Caby M. P., Novault S., et al. (2005). Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: Results of thefirst phase I clinical trial. J. Transl. Med. 3, 10–13. doi:10.1186/1479-5876-3-10

PubMed Abstract | CrossRef Full Text | Google Scholar

Hiraki T., Kunimatsu R., Nakajima K., Abe T., Yamada S., Rikitake K., et al. (2020). Stem cell-derived conditioned media from human exfoliated deciduous teeth promote bone regeneration. Oral Dis. 26 (2), 381–390. doi:10.1111/odi.13244

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirata M., Ishigami M., Matsushita Y., Ito T., Hattori H., Hibi H., et al. (2016). Multifaceted therapeutic benefits of factors derived from dental pulp stem cells for mouse liver fibrosis. Stem Cells Transl. Med. 5 (10), 1416–1424. doi:10.5966/sctm.2015-0353

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoshiba T. (2018). An extracellular matrix (ECM) model at high malignant colorectal tumor increases chondroitin sulfate chains to promote epithelial-mesenchymal transition and chemoresistance acquisition. Exp. Cell Res. 370 (2), 571–578. doi:10.1016/j.yexcr.2018.07.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoveizi E., Tavakol S., Shirian S., Sanamiri K. (2019). Electrospun nanofibers for diabetes: Tissue engineering and cell-based therapies. Curr. Stem Cell Res. Ther. [Internet] 14 (2), 152–168. doi:10.2174/1574888X13666181018150107

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu B., Chen S., Zou M., He Z., Shao S., Liu B. (2016). Effect of extracellular vesicles on neural functional recovery and immunologic suppression after rat cerebral apoplexy. Cell. Physiol. biochem. 40 (1–2), 155–162. doi:10.1159/000452533

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu C., Li L. (2018). Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J. Cell. Mol. Med. 22 (3), 1428–1442. doi:10.1111/jcmm.13492

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang C. C., Narayanan R., Warshawsky N., Ravindran S. (2018). Dual ECM biomimetic scaffolds for dental pulp regenerative applications. Front. Physiol. 9, 495–511. doi:10.3389/fphys.2018.00495

PubMed Abstract | CrossRef Full Text | Google Scholar

Im G-I. (2018). Tissue engineering in osteoarthritis: Current status and prospect of mesenchymal stem cell therapy. BioDrugs, BioDrugs [Internet] 32 (3), 183–192. doi:10.1007/s40259-018-0276-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Izumoto-Akita T., Tsunekawa S., Yamamoto A., Uenishi E., Ishikawa K., Ogata H., et al. (2015). Secreted factors from dental pulp stem cells improve glucose intolerance in streptozotocin-induced diabetic mice by increasing pancreatic β-cell function. BMJ Open Diabetes Res. Care 3 (1), e000128. doi:10.1136/bmjdrc-2015-000128

PubMed Abstract | CrossRef Full Text | Google Scholar

Jarmalavičiūtė A., Tunaitis V., Pivoraitė U., Venalis A., Pivoriūnas A. (2015). Exosomes from dental pulp stem cells rescue human dopaminergic neurons from 6-hydroxy-dopamine–induced apoptosis. Cytotherapy 17 (7), 932–939. doi:10.1016/j.jcyt.2014.07.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Jia Y., Zhu Y., Qiu S., Xu J., Chai Y. (2019). Exosomes secreted by endothelial progenitor cells accelerate bone regeneration during distraction osteogenesis by stimulating angiogenesis. Stem Cell Res. Ther. 10 (1), 12. doi:10.1186/s13287-018-1115-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang T., Deng M., James R., Nair L. S., Laurencin C. T. (2014). Micro- and nanofabrication of chitosan structures for regenerative engineering. Acta Biomater. 10 (4), 1632–1645. doi:10.1016/j.actbio.2013.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaukonen R., Jacquemet G., Hamidi H., Ivaska J. (2017). Cell-derived matrices for studying cell proliferation and directional migration in a complex 3D microenvironment. Nat. Protoc. 12 (11), 2376–2390. doi:10.1038/nprot.2017.107

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim H. Y., Kwon S., Um W., Shin S., Kim C. H., Park J. H., et al. (2022). Functional extracellular vesicles for regenerative medicine. Small 18, e2106569. doi:10.1002/smll.202106569

PubMed Abstract | CrossRef Full Text | Google Scholar

Larssen P., Wik L., Czarnewski P., Eldh M., Löf L., Ronquist K. G., et al. (2017). Tracing cellular origin of human exosomes using multiplex proximity extension assays. Mol. Cell. Proteomics 16 (3), 502–511. doi:10.1074/mcp.M116.064725

PubMed Abstract | CrossRef Full Text | Google Scholar

Laura E. (2018). Understanding the extracellular matrix to enhance stem cell-based tissue regeneration. Cell Stem Cell 22 (3), 302–305. doi:10.1016/j.stem.2018.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Laurencin C. T., Daneshmandi L. (2020). Graphene for regenerative engineering. Int. Jnl. Ceram. Engine. &amp. Sci. 2 (3), 140–143. doi:10.1002/ces2.10045

CrossRef Full Text | Google Scholar

C. T. Laurencin, and L. S. Nair (Editors) (2014). Nanotechnology and regenerative engineering. (Florida FL, USA: CRC Press). Availableat: https://www.taylorfrancis.com/books/9781466585386.

Google Scholar

Lee J. Y., Kim H. S. (2021). Extracellular vesicles in regenerative medicine: Potentials and challenges. Tissue Eng. Regen. Med. 18 (4), 479–484. doi:10.1007/s13770-021-00365-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Li X., Zheng Y., Hou L., Zhou Z., Huang Y., Zhang Y., et al. (2020). Exosomes derived from maxillary BMSCs enhanced the osteogenesis in iliac BMSCs. Oral Dis. 26 (1), 131–144. doi:10.1111/odi.13202

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu Y., Holmes C. (2021). Tissue regeneration capacity of extracellular vesicles isolated from bone marrow-derived and adipose-derived mesenchymal stromal/stem cells. Front. Cell Dev. Biol. 9, 648098. doi:10.3389/fcell.2021.648098

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu Z., Chen Y., Dunstan C., Roohani-Esfahani S., Zreiqat H. (2017). Priming adipose stem cells with tumor necrosis factor-alpha preconditioning potentiates their exosome efficacy for bone regeneration. Tissue Eng. Part A 23 (21–22), 1212–1220. doi:10.1089/ten.tea.2016.0548

PubMed Abstract | CrossRef Full Text | Google Scholar

Maguire G. (2013). Stem cell therapy without the cells. Commun. Integr. Biol. 6 (6), e26631. doi:10.4161/cib.26631

PubMed Abstract | CrossRef Full Text | Google Scholar

Matai I., Kaur G., Seyedsalehi A., McClinton A., Laurencin C. T. (2020). Progress in 3D bioprinting technology for tissue/organ regenerative engineering. Biomater. [Internet] 226, 119536. doi:10.1016/j.biomaterials.2019.119536

PubMed Abstract | CrossRef Full Text | Google Scholar

Matoug-Elwerfelli M., Nazzal H., Raif E. M., Wilshaw S-P., Esteves F., Duggal M. (2020). Ex-vivo recellularisation and stem cell differentiation of a decellularised rat dental pulp matrix. Sci. Rep. 10 (1), 21553. doi:10.1038/s41598-020-78477-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsushita Y., Ishigami M., Matsubara K., Kondo M., Wakayama H., Goto H., et al. (2017). Multifaceted therapeutic benefits of factors derived from stem cells from human exfoliated deciduous teeth for acute liver failure in rats. J. Tissue Eng. Regen. Med. 11 (6), 1888–1896. doi:10.1002/term.2086

PubMed Abstract | CrossRef Full Text | Google Scholar

Menasché P. (2018). Cell therapy trials for heart regeneration — Lessons learned and future directions. Nat. Rev. Cardiol. 15 (11), 659–671. doi:10.1038/s41569-018-0013-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Mita T., Furukawa-Hibi Y., Takeuchi H., Hattori H., Yamada K., Hibi H., et al. (2015). Conditioned medium from the stem cells of human dental pulp improves cognitive function in a mouse model of Alzheimer’s disease. Behav. Brain Res. 293, 189–197. doi:10.1016/j.bbr.2015.07.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Mocchi M., Bari E., Marrubini G., Bonda A. F., Perteghella S., Tartara F., et al. (2021). Freeze-Dried Mesenchymal stem cell-secretome pharmaceuticalization: Optimization of formulation and manufacturing process Robustness. Pharmaceutics 13 (8), 1129. doi:10.3390/pharmaceutics13081129

PubMed Abstract | CrossRef Full Text | Google Scholar

Murphy D. E., de Jong O. G., Brouwer M., Wood M. J., Lavieu G., Schiffelers R. M., et al. (2019). Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 51 (3), 1–12. doi:10.1038/s12276-019-0223-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Naba A., Clauser K. R., Ding H., Whittaker C. A., Carr S. A., Hynes R. O. (2016). The extracellular matrix: Tools and insights for the “omics” era. Matrix Biol. 49, 10–24. doi:10.1016/j.matbio.2015.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagelkerke A., Ojansivu M., van der Koog L., Whittaker T. E., Cunnane E. M., Silva A. M., et al. (2021). Extracellular vesicles for tissue repair and regeneration: Evidence, challenges and opportunities. Adv. Drug Deliv. Rev. 175, 113775. doi:10.1016/j.addr.2021.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Neves V. C. M., Babb R., Chandrasekaran D., Sharpe P. T. (2017). Promotion of natural tooth repair by small molecule GSK3 antagonists. Sci. Rep. 7, 39654. doi:10.1038/srep39654

PubMed Abstract | CrossRef Full Text | Google Scholar

Nigri J., Leca J., Tubiana S-S., Finetti P., Guillaumond F., Martinez S., et al. (2022). CD9 mediates the uptake of extracellular vesicles from cancer-associated fibroblasts that promote pancreatic cancer cell aggressiveness. Sci. Signal. 15 (745), eabg8191. doi:10.1126/scisignal.abg8191

PubMed Abstract | CrossRef Full Text | Google Scholar

Nowwarote N., Petit S., Ferre F. C., Dingli F., Laigle V., Loew D., et al. (2022). Extracellular matrix derived from dental pulp stem cells promotes mineralization. Front. Bioeng. Biotechnol. 9, 740712–740811. doi:10.3389/fbioe.2021.740712

PubMed Abstract | CrossRef Full Text | Google Scholar

Omori M., Tsuchiya S., Hara K., Kuroda K., Hibi H., Okido M., et al. (2015). A new application of cell-free bone regeneration: Immobilizing stem cells from human exfoliated deciduous teeth-conditioned medium onto titanium implants using atmospheric pressure plasma treatment. Stem Cell Res. Ther. 6 (1), 124. doi:10.1186/s13287-015-0114-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Qiu J., Wang X., Zhou H., Zhang C., Wang Y., Huang J., et al. (2020). Enhancement of periodontal tissue regeneration by conditioned media from gingiva-derived or periodontal ligament-derived mesenchymal stem cells: A comparative study in rats. Stem Cell Res. Ther. 11 (1), 42. doi:10.1186/s13287-019-1546-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramasubramanian L., Kumar P., Wang A. (2019). Engineering extracellular vesicles as nanotherapeutics for regenerative medicine. Biomolecules 10 (1), 48. doi:10.3390/biom10010048

PubMed Abstract | CrossRef Full Text | Google Scholar

Raposo G., Stoorvogel W. (2013). Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 200 (4), 373–383. doi:10.1083/jcb.201211138

PubMed Abstract | CrossRef Full Text | Google Scholar

Serebriiskii I., Castelló-Cros R., Lamb A., Golemis E. A., Cukierman E. (2008). Fibroblast-derived 3D matrix differentially regulates the growth and drug-responsiveness of human cancer cells. Matrix Biol. 27 (6), 573–585. doi:10.1016/j.matbio.2008.02.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Shao L., Zhang Y., Lan B., Wang J., Zhang Z., Zhang L., et al. (2017). MiRNA-sequence indicates that mesenchymal stem cells and exosomes have similar mechanism to enhance cardiac repair. Biomed. Res. Int. 2017, 1–9.

CrossRef Full Text | Google Scholar

Silva A. M., Teixeira J. H., Almeida M. I., Gonçalves R. M., Barbosa M. A., Santos S. G. (2017). Extracellular Vesicles: Immunomodulatory messengers in the context of tissue repair/regeneration. Eur. J. Pharm. Sci. 98, 86–95. doi:10.1016/j.ejps.2016.09.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Song P., Kwon Y., Joo J. Y., Kim D. G., Yoon J. H. (2019). Secretomics to discover regulators in diseases. Int. J. Mol. Sci. 20 (16), E3893–E3894. doi:10.3390/ijms20163893

PubMed Abstract | CrossRef Full Text | Google Scholar

Tucker G., DeSilva B., Dressman J., Ito M., Kumamoto T., Mager D., et al. (2016). Current challenges and potential opportunities for the pharmaceutical sciences to make global impact: An FIP perspective. J. Pharm. Sci. 105 (9), 2489–2497. doi:10.1016/j.xphs.2015.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Turinetto V., Vitale E., Giachino C. (2016). Senescence in human mesenchymal stem cells: Functional changes and implications in stem cell-based therapy. Int. J. Mol. Sci. 17 (7), 1164. doi:10.3390/ijms17071164

PubMed Abstract | CrossRef Full Text | Google Scholar

Wakayama H., Hashimoto N., Matsushita Y., Matsubara K., Yamamoto N., Hasegawa Y., et al. (2015). Factors secreted from dental pulp stem cells show multifaceted benefits for treating acute lung injury in mice. Cytotherapy 17 (8), 1119–1129. doi:10.1016/j.jcyt.2015.04.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang J., Zuzzio K., Walker C. L. (2019). Systemic dental pulp stem cell secretome therapy in a mouse model of amyotrophic lateral sclerosis. Brain Sci. 9 (7), 165. doi:10.3390/brainsci9070165

PubMed Abstract | CrossRef Full Text | Google Scholar

Willis C. M., Nicaise A. M., Hamel R., Pappa V., Peruzzotti-Jametti L., Pluchino S. (2020). Harnessing the neural stem cell secretome for regenerative neuroimmunology. Front. Cell. Neurosci. 14, 590960–591014. doi:10.3389/fncel.2020.590960

PubMed Abstract | CrossRef Full Text | Google Scholar

UN Population Division (2019). World Population Prospects 2019 [Internet] (Department of Economic and Social Affairs). Available at: http://www.ncbi.nlm.nih.gov/pubmed/12283219.

Google Scholar

Xia J., Minamino S., Kuwabara K., Arai S. (2019). Stem cell secretome as a new booster for regenerative medicine. Biosci. Trends 13 (4), 299–307. doi:10.5582/bst.2019.01226

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie M., Shi Y., Zhang C., Ge M., Zhang J., Chen Z., et al. (2022). In situ 3D bioprinting with bioconcrete bioink. Nat. Commun. 13 (1), 3597. doi:10.1038/s41467-022-30997-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamaguchi S., Shibata R., Yamamoto N., Nishikawa M., Hibi H., Tanigawa T., et al. (2015). Dental pulp-derived stem cell conditioned medium reduces cardiac injury following ischemia-reperfusion. Sci. Rep. 5 (1), 16295. doi:10.1038/srep16295

PubMed Abstract | CrossRef Full Text | Google Scholar

Yelick P. C., Sharpe P. T. (2019). Tooth bioengineering and regenerative dentistry. J. Dent. Res. 98, 1173–1182. doi:10.1177/0022034519861903

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang H., Freitas D., Kim H. S., Fabijanic K., Li Z., Chen H., et al. (2018). Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 20 (3), 332–343. doi:10.1038/s41556-018-0040-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang Q., Jeppesen D. K., Higginbotham J. N., Graves-Deal R., Trinh V. Q., Ramirez M. A., et al. (2021). Supermeres are functional extracellular nanoparticles replete with disease biomarkers and therapeutic targets. Nat. Cell Biol. 23 (12), 1240–1254. doi:10.1038/s41556-021-00805-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang X., Zhang Y. (2015). Tissue engineering applications of three-dimensional bioprinting. Cell biochem. Biophys. [Internet] 72 (3), 777–782. doi:10.1007/s12013-015-0531-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou Y., Yamamoto Y., Xiao Z., Ochiya T. (2019). The immunomodulatory functions of mesenchymal stromal/stem cells mediated via paracrine activity. J. Clin. Med. 8 (7), 1025. doi:10.3390/jcm8071025

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: regenerative dentistry, extracellular space, secretome, extracellular vesicles, regeneration

Citation: Birjandi AA and Sharpe P (2022) Potential of extracellular space for tissue regeneration in dentistry. Front. Physiol. 13:1034603. doi: 10.3389/fphys.2022.1034603

Received: 01 September 2022; Accepted: 24 October 2022;
Published: 17 November 2022.

Edited by:

Sachiko Iseki, Tokyo Medical and Dental University, Japan

Reviewed by:

Tsutomu Iwamoto, Tokyo Medical and Dental University, Japan

Copyright © 2022 Birjandi and Sharpe. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Paul Sharpe, Paul.sharpe@kcl.ac.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.