Abstract

Extracellular vesicles (EVs) are evolutionarily conserved communication mediators that play key roles in the development of periodontal disease as well as in regeneration processes. This concise review first outlines the pathogenic mechanisms through which EVs derived from bacteria lead to the progression of periodontitis, with a focus on the enrichment of virulence factors, the amplification of immune responses, and the induction of bone destruction as key aspects influenced by bacterial EVs. This review aims to elucidate the positive effects of EVs derived from mesenchymal stem cells (MSC-EVs) on periodontal tissue regeneration. In particular, the anti-inflammatory properties of MSC-EVs and their impact on the intricate interplay between MSCs and various immune cells, including macrophages, dendritic cells, and T cells, are described. Moreover, recent advancements regarding the repair-promoting functions of MSC-EVs are detailed, highlighting the mechanisms underlying their ability to promote osteogenesis, cementogenesis, angiogenesis, and the homing of stem cells, thus contributing significantly to periodontal tissue regeneration. Furthermore, this review provides insights into the therapeutic efficacy of MSC-EVs in treating periodontitis within a clinical context. By summarizing the current knowledge, this review aims to provide a comprehensive understanding of how MSC-EVs can be harnessed for the treatment of periodontal diseases. Finally, a discussion is presented on the challenges that lie ahead and the potential practical implications for translating EV-based therapies into clinical practices for the treatment of periodontitis.

Significance statement

This review underscores the critical role of bacterial extracellular vesicles in periodontitis, detailing their pathogenic mechanisms and impact on immune responses and bone resorption. It highlights a novel insight into how bacteria manipulate host cells to enhance inflammation through cellular extracellular vesicles. Furthermore, it emphasizes the therapeutic potential of mesenchymal stem cell-derived extracellular vesicles, showcasing their anti-inflammatory properties and regenerative capabilities in periodontal therapy. By integrating these findings, the article provides a comprehensive understanding of EVs in periodontal disease, paving the way for innovative treatment strategies that could significantly improve therapeutic outcomes.

Introduction

Periodontitis is one of the most common diseases and affects 61.6% of the population, with 11.2%-12.50% of people experiencing severe manifestations,1-3 representing a substantial burden on healthcare systems and resulting in major social and economic challenges. The primary characteristic of periodontitis is the loss of periodontal tissue support, as shown by clinical attachment loss, alveolar bone loss, and the occurrence of periodontal pockets and gingival bleeding. As the leading cause of edentulism and masticatory dysfunction, periodontitis affects the nutrition, facial features, quality of life, and self-esteem of patients and results in major socioeconomic impacts and healthcare costs.4 Furthermore, increasing evidence has indicated that periodontitis is critically associated with systemic disorders, such as cardiometabolic, neurodegenerative and autoimmune diseases, and cancer.5 Moreover, periodontitis (edentulism) is linked to a greater risk of all-cause and cause-specific mortality.6 The current treatment methods for periodontitis include scaling and root planning; however, these strategies can control the development of periodontitis but cannot restore lost periodontal tissues. Guided tissue regeneration is the major method for periodontal regeneration; however, limited regenerative outcomes in the clinic are observed, which is unsatisfactory.7

Polymicrobial communities are believed to be the causative agents of periodontitis.8 These polymicrobial communities can subvert the host immune response, which can prevent death and further reinforce inflammation in a positive feedback loop. The interactions between bacteria and host cells are crucial in the pathogenesis of periodontal diseases, as noted in several excellent reviews.9,10 In these reviews, the roles of cytokines,11,12 bacterial virulence factors,13 and the direct communication of periodontal pathogens and host immune cells9 have been highlighted. Recently, accumulating findings have indicated that extracellular vesicles (EVs) released by bacteria play key roles in intercellular communication. Bacteria-derived extracellular vesicles (BEVs) are nanosized particles enclosed by lipid bilayers, that contain various substances, such as enzymes, toxins, and microbe-associated molecular patterns (MAMPs), that function as virulence factors in the host.14 Compared with parental bacteria, BEVs can harbor virulence factors and enter host immune cells, thus resulting in much stronger immune responses and exacerbating inflammation-mediated tissue destruction.15 Therefore, summarizing the recent progress in determining the effects of BEVs on the development of periodontal diseases is important.

As evolutionarily conserved communication mediators from bacteria to eukaryotic cells, EVs derived from periodontal cells also contribute to the progression of periodontal diseases and regenerative therapies for periodontal diseases. Owing to the minimal immunogenicity, high safety, and multiple bioactivities of EVs derived from mesenchymal stem cells (MSC-EVs), MSC-EVs are considered the most effective alternatives for mesenchymal stem cells (MSCs); these EVs can exert immunomodulatory effects and promote osteogenesis/cementogenesis/angiogenesis, similar to their parental MSCs. The mechanisms and clinical application potential of MSCs have been extensively described in a series of excellent reviews.16,17 Moreover, the therapeutic performance of MSC-EVs can be effectively increased by modifying their cargoes or designing local delivery systems.18 Owing to these advantages, EVs have been applied in numerous fields of medicine, including the treatment of neurodegenerative disorders (Alzheimer’s disease, central nervous system), the regeneration of damaged organs (kidney, liver and heart) and tissues(skin and dental pulp), and early diagnosis of cancers and so on.19,20 All of these compounds demonstrate great therapeutic effects and application prospects. More importantly, some investigators have also explored the translational prospects of MSC-EVs in the treatment of periodontitis. However, emerging evidence has also indicated that EVs from periodontal cells have pathogenic effects by exacerbating the host immune response and inducing tissue destruction, which cannot be ignored, and little attention has been given to this issue.

Overall, the present review summarizes the latest advancements in the application of EVs as essential mediators involved in periodontal disease and regeneration by mediating intercellular communication among bacteria and bacteria and cell-cell interactions. First, we underscore the importance of EVs in the pathogenesis of periodontitis, encompassing the pathogenic effects of both BEVs and cellular extracellular vesicles (CEVs). We subsequently explored the mechanisms through which CEVs exert anti-inflammatory effects and repair-promoting functions. Finally, we present the translational prospects of EVs in the treatment of periodontitis, along with a description of potential future challenges in this field.

Pathogenic roles of EVs in the progression of periodontitis

Extracellular vesicles derived from bacteria or cells carry virulence factors and inflammatory mediators, thereby exacerbating inflammation and accelerating tissue destruction during the pathogenesis of periodontitis. On the one hand, virulence factors can be highly enriched in EVs, which can exert even more powerful pathogenic effects than their cells of origin. On the other hand, inflammatory mediators carried by EVs can directly activate the intracellular pattern recognition receptors (PRRs) of cells, leading to an excessive proinflammatory response in cells. In this section, we delve into recent findings on the pathogenic roles of EVs in the development of periodontitis, highlighting their effects on the amplified immune response and accelerated bone resorption.

Pathogenic roles of BEVs in periodontal homeostasis

Bacteria can produce BEVs by either shedding the outer membrane or explosive cytolysis. The BEVs produced through blebbing of the outer membrane can be categorized into the following three types: outer membrane vesicles (OMVs), outer-inner membrane vesicles (OIMVs), and cytoplasmic membrane vesicles (CMVs)21 (Figure 1A). Outer-inner membrane vesicles arise from blebbing of the outer membrane due to an imbalance in peptidoglycan biosynthesis, denatured protein accumulation, or the insertion of hydrophobic molecules into the outer membrane. Intact peptidoglycans and the inner membrane can prevent the entry of cytoplasmic constituents into OMVs; therefore, the cargos of OMVs are devoid of cytoplasmic components but rich in periplasmic and outer membrane proteins.22 When the bacterial peptidoglycan layer is weakened by autolysin, the cytoplasmic contents, inner membranes, and cytoplasmic contents can be enveloped by OIMVs. CMVs may form as a result of stress-induced autolysis by gram-positive bacteria or peptidoglycan hydrolysis caused by exogenous endolysin or antibiotics that inhibit peptidoglycan biosynthesis.21 Although the biogenesis mechanisms of OMVs, OIMVs, and CMVs are substantially different, distinguishing these vesicles is difficult. In addition, pure OMVs, OIMVs, and CMVs cannot be obtained via current isolation methods. Therefore, OMVs, OIMVs, and CMVs are collectively referred to as BEVs in this study.17

Pathogenic roles of extracellular vesicles in the pathogenesis of periodontal disease. (A) The biogenesis of bacterial-derived extracellular vesicles (BEVs) containing various types of virulence factors. Outer membrane vesicles (OMVs) and outer-inner membrane vesicles (OIMVs) are generated by gram-negative bacteria, whereas cytoplasmic membrane vesicles (CMVs) originate from gram-positive bacteria. All of these BEVs are capable of transporting virulence factors. For example, OMVs from T. forsythia are known to carry BspA and sialidase, which induce inflammation by activating Toll-like receptor 2 (TLR2). Additionally, P. gingivalis-derived periplasmic vesicles (Pg EVs) contain virulence factors such as gingipain (GP) and lipopolysaccharide (LPS), which increase the expression of tumor necrosis factor-alpha (TNF-α) and interleukin-13 (IL-13) and increase the number of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-positive CD11c cells within periodontal tissues. (B) Amplification of the immune response mediated by BEVs released from periodontal pathogens. BEVs can bind to host cells (epithelial cells, neutrophils, and macrophages) through interactions with pattern recognition receptors (PRRs), such as TLR2, TLR4, NOD1, and NOD2, thereby initiating the activation of downstream proinflammatory signaling pathways. (C) BEVs induce bone resorption by activating TLR2 in bone mesenchymal stem cells (BMSCs) and osteoclasts, thereby playing dual regulatory roles in bone metabolism. BEVs inhibit osteoblast differentiation by modulating the RANKL-RANK-OPG signaling axis while promoting osteoclast differentiation by upregulating factors for osteoclast precursor cells. (D) Cellular extracellular vesicles (CEVs) originating from host cells infected by bacteria or their products exacerbate the progression of periodontitis. Host cells exposed to bacteria, BMVs, or LPS release CEVs enriched with proinflammatory cytokines and mediators, including TNF-α and interleukin-6 (IL-6). These CEVs further act on recipient cells to intensify inflammation and promote tissue destruction. Abbreviations: CM, cytoplasmic membrane; ERK, extracellular signal-regulated kinase; Fa, Fusobacterium nucleatum; FOXP3, Forkhead Box P3; G-CSF, granulocyte colony-stimulating facto; GSDMD, Gasdermin D; IL-10, interleukin-10; IL-17, interleukin-17; IL-1ra, interleukin-1ra; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; MIP-1α, macrophage inflammation protein-1α; MMP1, matrix metalloproteinase-1; MMP3, matrix metalloproteinase-3; MMP9, matrix metalloproteinase-9; NLRP3, NOD-like Receptor Family Pyrin Domain Containing 3. OM, outer membrane; OPG, osteoprotegerin; RANK, receptor activator of nuclear factorκB; RANKL, the receptor activator of nuclear factorκB ligand; RORC, Retinoic Acid Receptor-related Orphan Receptor C; SIRT1, Sirtuin-1; SPP1, Secreted Phosphoprotein 1; Tf, Treponema denticola
Figure 1.

Pathogenic roles of extracellular vesicles in the pathogenesis of periodontal disease. (A) The biogenesis of bacterial-derived extracellular vesicles (BEVs) containing various types of virulence factors. Outer membrane vesicles (OMVs) and outer-inner membrane vesicles (OIMVs) are generated by gram-negative bacteria, whereas cytoplasmic membrane vesicles (CMVs) originate from gram-positive bacteria. All of these BEVs are capable of transporting virulence factors. For example, OMVs from T. forsythia are known to carry BspA and sialidase, which induce inflammation by activating Toll-like receptor 2 (TLR2). Additionally, P. gingivalis-derived periplasmic vesicles (Pg EVs) contain virulence factors such as gingipain (GP) and lipopolysaccharide (LPS), which increase the expression of tumor necrosis factor-alpha (TNF-α) and interleukin-13 (IL-13) and increase the number of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-positive CD11c cells within periodontal tissues. (B) Amplification of the immune response mediated by BEVs released from periodontal pathogens. BEVs can bind to host cells (epithelial cells, neutrophils, and macrophages) through interactions with pattern recognition receptors (PRRs), such as TLR2, TLR4, NOD1, and NOD2, thereby initiating the activation of downstream proinflammatory signaling pathways. (C) BEVs induce bone resorption by activating TLR2 in bone mesenchymal stem cells (BMSCs) and osteoclasts, thereby playing dual regulatory roles in bone metabolism. BEVs inhibit osteoblast differentiation by modulating the RANKL-RANK-OPG signaling axis while promoting osteoclast differentiation by upregulating factors for osteoclast precursor cells. (D) Cellular extracellular vesicles (CEVs) originating from host cells infected by bacteria or their products exacerbate the progression of periodontitis. Host cells exposed to bacteria, BMVs, or LPS release CEVs enriched with proinflammatory cytokines and mediators, including TNF-α and interleukin-6 (IL-6). These CEVs further act on recipient cells to intensify inflammation and promote tissue destruction. Abbreviations: CM, cytoplasmic membrane; ERK, extracellular signal-regulated kinase; Fa, Fusobacterium nucleatum; FOXP3, Forkhead Box P3; G-CSF, granulocyte colony-stimulating facto; GSDMD, Gasdermin D; IL-10, interleukin-10; IL-17, interleukin-17; IL-1ra, interleukin-1ra; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; MIP-1α, macrophage inflammation protein-1α; MMP1, matrix metalloproteinase-1; MMP3, matrix metalloproteinase-3; MMP9, matrix metalloproteinase-9; NLRP3, NOD-like Receptor Family Pyrin Domain Containing 3. OM, outer membrane; OPG, osteoprotegerin; RANK, receptor activator of nuclear factorκB; RANKL, the receptor activator of nuclear factorκB ligand; RORC, Retinoic Acid Receptor-related Orphan Receptor C; SIRT1, Sirtuin-1; SPP1, Secreted Phosphoprotein 1; Tf, Treponema denticola

Transmission and enrichment of virulence factors

BEVs can carry and transport many toxins or virulence factors, actively participate in periodontitis-related tissue damage and deliver many effectors to host tissues that are not colonized by parental bacteria.17 Membrane-anchored toxins, including lipopolysaccharides (LPSs) and gingipains, along with bacterial surface adhesion factors, can be integrated into the membranes of OMVs and OIMVs and play key roles in the BEV-mediated pathogenic effects associated with periodontitis. Additionally, OIMVs or CMVs contain bacterial nucleic acids (eg, RNA and DNA) and cytoplasmic proteins and enzymes (eg, peptidyl arginine deiminase), which serve as virulence factors derived from intracellular components. The interplay between virulence factors and BEVs is critically important in the development of periodontal diseases. For example, the OMVs of Tannerella forsythia carry harmful substances such as BspA and sialidase, causing inflammation by activating TLR2 in macrophages.23 EVs derived from Porphyromonas gingivalis contain virulence factors, such as gingipains and LPS, which induce inflammation and increase the levels of TNF-α and interleukin-1β (IL-1β) expression in gingival tissues (Figure 1A). Furthermore, the virulence factors of BEVs are implicated in Alzheimer’s disease, colitis, and dysbiosis of the intestinal microbiota.24-26Aggregatibacter actinomycetemcomitans is implicated in aggressive forms of periodontitis, and OMVs derived from A. actinomycetemcomitans can deliver biologically active virulence factors (CDT, OmpA) into human gingival fibroblasts (HGFs) and can activate nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and 2 (NOD2), thus triggering an excessive innate immune response27-29 (Figure 1A).

Amplification of immune responses

Upon bacterial infection of the host organism, both the bacteria and their released vesicles can activate the immune response. BEVs can directly activate a broad range of innate immune PRRs, including surface Toll-like receptors (TLRs) and cytosolic NOD1 and NOD2, and then activate the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and inflammasomes, resulting in the production of proinflammatory cytokines, inflammation, and programmed cell death across various cell types, such as epithelial cells, neutrophils, monocytes, and macrophages21,30 (Figure 1B).

Neutrophils are one of the most important immune cells that defend against bacterial invasion. Gasdermin D (GSDMD), a pore-forming protein, has emerged as a key downstream effector of pyroptosis, a form of cell death triggered by intracellular LPS. After the endocytosis of OMVs by neutrophils, LPS on the surface of OMVs can induce NETosis through GSDMD.15 LPS binds to and activates caspase-4/11 within the noncanonical inflammasome complex, leading to the cleavage of GSDMD, which subsequently causes perforation of the nuclear and plasma membranes. This altered nuclear permeability allows caspase-11 to enter chromatin, leading to histone cleavage, inactivation and DNA amplification. Disruption of the plasma membrane enables neutrophils to expel their uncompressed chromatin. In addition, this process can trigger the activation of the NLRP3 inflammasome and the secretion of IL-1β and interleukin-18 (IL-18). During this process, caspase-4/11-driven NETosis prevents cytoplasmic infection, whereas the expulsion of neutrophil extracellular traps (NETs) results in the dissemination of bacteria within the host.15,31 Nevertheless, the OMVs of bacteria also facilitate bacterial evasion of the immune system. For example, P. gingivalis produces gingipains, which are cysteine proteinases that differentially affect interleukin-8 (IL-8) depending on their form. In their soluble form, gingipains first convert IL-8 into a more potent form and then gradually degrade it. However, when attached to OMVs, these molecules instantly degrade IL-8. This difference in activity can result in localized proinflammatory and anti-inflammatory responses around bacterial plaques, which may explain why the infection persists despite high levels of neutrophils at sites of periodontitis.32 In addition to IL-8, gingipains ensure bacterial survival by degrading secretory granule components with antibacterial activity produced by neutrophils, particularly the antimicrobial peptides leucine‒leucine-37 (LL-37) and myeloperoxidase (MPO).33

In addition to neutrophils, macrophages can engulf vesicles, with OMVs from P. gingivalis, Treponema denticola, and Fusobacterium nucleatum being engulfed by monocytes and macrophages in a dose-dependent manner. These OMVs also activate NF-κB and augment the release of proinflammatory cytokines such as tumor necrosis factor alpha (TNFα), IL-8, and IL-1β.34 OMVs from F. nucleatum polarize macrophages toward the M1 phenotype, thereby exacerbating the development of periodontitis.35

Bone resorption

Bone homeostasis is maintained by the coordinated action of two different types of cells: osteoblasts and osteoclasts. Osteoblasts, which originate from the mesenchymal lineage, play a crucial role in bone formation and facilitate mineralization by secreting unmineralized bone matrix and noncollagenous proteins, whereas osteoclasts, which originate from the hematopoietic lineage, are essential for bone resorption because they release hydrogen ions and lytic enzymes.36,37 In the context of periodontitis, under the dual pressures of microbial dysbiosis and an excessive immune response, the coordinated activity of osteoblasts and osteoclasts is disrupted, resulting in inhibited bone formation and increased bone resorption.38

The critical regulatory axis for bone homeostasis, namely, the receptor activator of nuclear factor κB ligand (RANKL)-receptor activator of nuclear factor κB (RANK)-osteoprotegerin (OPG) axis, is essential for periodontitis-related bone resorption. RANKL is synthesized by multiple cellular lineages, with osteoblasts and bone marrow stromal cells (BMSCs) serving as principal sources within osseous tissue.39 Throughout the lifespan of multinucleated osteoclasts, RANK expression persists on cells in the osteoclast lineage until terminal differentiation into mature entities. The interaction of RANKL and RANK not only induces osteoclastogenesis but also promotes the survival and activation of these cells, thereby facilitating osteoclastic resorption. Additionally, osteoblasts secrete OPG, which functions as a soluble decoy receptor for RANKL, competitively inhibiting the binding of RANKL to its cognate receptor RANK. This regulatory mechanism mitigates osteoclast formation while promoting the apoptosis of previously differentiated and activated osteoclasts, ultimately decreasing the extent of bone resorption. Therefore, the RANKL/OPG ratio is a crucial factor influencing the rate and extent of osteoclast-mediated bone resorption.40

BEVs derived from Filifactor alocis inhibit osteogenic differentiation by activating TLR2 and subsequently triggering the MAPK and NF-κB signaling pathways. During this process, extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) serve as the principal downstream effectors of TLR2 (Figure 1C). Additionally, BEVs augment the RANKL/OPG ratio in a TLR2-dependent manner, thereby promoting osteoclast differentiation and subsequently increasing bone resorption.14,39 In contrast, the BEVs of F. nucleatum increased the number of osteoclasts and exacerbated alveolar bone loss in a mouse model of periodontitis. BEVs from P. gingivalis and T. forsythia promote osteoclast differentiation of osteoclast precursors and increase the expression of osteoclastogenic cytokines, such as granulocyte colony-stimulating factor (G-CSF), interleukin-1ra (IL-1ra), interleukin-6 (IL-6), IFN-γ-inducible protein 10 (IP-10), macrophage inflammation protein-1α (MIP-1α), macrophage inflammation protein-1β (MIP-1β), and macrophage inflammation protein-2 (MIP-2), which are regulated upon activation and normally expressed by T cells (RANTES) and TNF-α (Figure 1C). Additionally, the osteoclastogenic effect induced by BEVs can be attenuated by lipoprotein lipase and polymyxin B (a drug that inhibits LPS). These findings indicate that BEVs from P. gingivalis and T. forsythia promote osteoclastogenesis by activating TLR2 via lipoprotein/LPS interactions41 (Figure 1C).

Pathogenic roles of CEVs in periodontal homeostasis

The influence of bacteria on periodontal homeostasis extends beyond the bacteria themselves and their released BEVs. Bacteria can also exploit host cells to secrete CEVs containing proinflammatory cytokines and mediators, thereby amplifying the inflammatory response. For example, the expression of age-related microRNAs (miRNAs), IL-1β, IL-6, TNF-α, and the P. gingivalis fimbrial adhesin protein Mfa1 in EVs increases significantly when dendritic cells (DCs) are exposed to P. gingivalis, which elicits premature senescence in neighboring DCs through paracrine signaling42,43 (Figure 1D). In the context of T. forsythia infection, macrophage-derived CEVs are rich in proinflammatory cytokines and inflammatory mediators, such as TNF, IL-1β, HLA class I histocompatibility antigen B-7 alpha chain (HLA-B) and matrix metalloproteinase-9 (MMP9), which contribute to an exaggerated inflammatory response during the progression of periodontitis.23 Under normal conditions, periodontal ligament stem cells (PDLSCs) can mitigate the inflammatory microenvironment via the T helper 17 (Th17) cell/regulatory T-cell (Treg)/miR-155-5p/Sirtuin-1 (SIRT1) regulatory network. However, EVs derived from LPS-stimulated PDLSCs presented decreased expression of miR-155-5p and elevated levels of SIRT1, resulting in an imbalance between Th17 cells and Tregs.44 Similarly, the upregulation of TNF-α and IL-6 in EVs derived from LPS-stimulated periodontal ligament fibroblasts (PDLFs) increased significantly, which inhibited alkaline phosphatase, collagen I, Runt-related transcription factor 2, and osteoprotegerin expression along with ALP activity in MG-63 osteoblasts, leading to decreased bone formation45 (Figure 1D). Moreover, EVs released from biofilm-stimulated gingival epithelial cells significantly upregulated the expression of matrix metalloproteinase-1 (MMP-1) and 3 (MMP-3) in HGFs, which promoted a tissue destruction phenotype and reduced the production of the extracellular matrix (ECM), leading to increased tissue degradation46 (Figure 1D). These findings indicate that CEVs also play key pathogenic roles in the imbalance of periodontal homeostasis.

Anti-inflammatory effects of MSC-EVs on immune cells

A balanced host immune response is the critical driver of periodontal homeostasis and health.47 MSC-EVs exert a profound and far-reaching effect on the behaviors of immune cells. These vesicles play pivotal roles in modulating macrophage polarization, steering macrophages from the M1 proinflammatory phenotype to the M2 anti-inflammatory phenotype. This transition plays a vital role in managing inflammatory responses and enhancing tissue repair. MSC-EVs also strongly impact T-cell regulation, notably affecting the differentiation of CD4 + T cells into Tregs. Additionally, MSC-EVs are pivotal in reducing the secretion of proinflammatory cytokines by DCs, decreasing their migratory capacity, and bolstering their anti-inflammatory functions. The intricate interactions between immune cells further amplify these regulatory effects; for example, MSC-EVs can indirectly influence T-cell differentiation by fostering anti-inflammatory responses in both macrophages and DCs. As a result, the modulation of immune cell behavior by MSC-EVs shows promise for therapeutic applications in managing inflammation and promoting tissue regeneration (Figure 2).

Anti-inflammatory effects of MSC-EVs on immune cells. MSC-EVs can transfer diverse cargoes (such as miRNAs and proteins) to different immune cells (including macrophages, T cells, and dendritic cells). These cargoes can activate various signaling pathways to exert anti-inflammatory effects on immune cells. Abbreviations: CCL21, C-C motif chemokine ligand 21; CCR7, chemokine receptor 7; ERK, extracellular signal-regulated kinase; GSH, glutathione; GSR, glutathione reductase; GSSG, glutathione disulfide; IL-12, interleukin-12; IL-1β, interleukin-1β; IL-6, interleukin-6; IκB, Inhibitor of κB; MyD88, Myeloid Differentiation Primary Response 88; NFAT5, Nuclear Factor of Activated T-cells 5; NF-κB, the nuclear factor kappa-light-chain-enhancer of activated B cells; PD-1, programmed cell death protein 1; PD-L1, programmed death ligand 1; ROS, reactive oxygen species; SIRT1, Sirtuin 1 SOD1, superoxide dismutase 1; TLR4, Toll-like receptor 4; TNF-α, tumor necrosis factor-alpha.
Figure 2.

Anti-inflammatory effects of MSC-EVs on immune cells. MSC-EVs can transfer diverse cargoes (such as miRNAs and proteins) to different immune cells (including macrophages, T cells, and dendritic cells). These cargoes can activate various signaling pathways to exert anti-inflammatory effects on immune cells. Abbreviations: CCL21, C-C motif chemokine ligand 21; CCR7, chemokine receptor 7; ERK, extracellular signal-regulated kinase; GSH, glutathione; GSR, glutathione reductase; GSSG, glutathione disulfide; IL-12, interleukin-12; IL-1β, interleukin-1β; IL-6, interleukin-6; IκB, Inhibitor of κB; MyD88, Myeloid Differentiation Primary Response 88; NFAT5, Nuclear Factor of Activated T-cells 5; NF-κB, the nuclear factor kappa-light-chain-enhancer of activated B cells; PD-1, programmed cell death protein 1; PD-L1, programmed death ligand 1; ROS, reactive oxygen species; SIRT1, Sirtuin 1 SOD1, superoxide dismutase 1; TLR4, Toll-like receptor 4; TNF-α, tumor necrosis factor-alpha.

Macrophages

Although there is a continual spectrum of macrophage phenotypes, macrophages can be classically divided into two phenotypes: the M1 proinflammatory phenotype and the M2 anti-inflammatory phenotype. These phenotypes are typically associated with wound healing, and the macrophage transition from the M1 phenotype to the M2 phenotype effectively alleviates inflammatory responses and promotes periodontal tissue regeneration.48 After systematic injection, MSC-EVs are engulfed mainly by macrophages, and the bioactive cargos loaded with MSC-EVs can repolarize macrophages from the M1 to the M2 phenotype through multiple signaling pathways. For example, EVs derived from dental pulp stem cells (DPSC-EVs) directly target inhibitor of nuclear factor kappa B kinase subunit beta (IKBKB) to inhibit the NF-κB and TLR signaling pathways, which inhibits the proinflammatory responses of macrophages and induces macrophage polarization toward the M2 phenotype. Moreover, these vesicles promote the release of bone morphogenetic protein 2 (BMP2) from macrophages to induce odontogenesis in dental pulp stem cells (DPSCs).49 Similarly, miR-1246 and miR-125a-3p in DPSC-EVs promoted the polarization of macrophages toward an anti-inflammatory phenotype in periodontitis models.50,51 Moreover, MSC-EVs can inhibit the pyroptosis of macrophages to inhibit inflammation, and EVs produced by PDLSCs facilitate the delivery of miR-590-3p to macrophages, resulting in the inhibition of TLR4 transcription and a subsequent reduction in macrophage pyroptosis, thereby mitigating the inflammatory damage caused by periodontitis.

In addition, pretreatment with MSCs can promote the anti-inflammatory effects of MSC-EVs in modulating macrophage function. For example, EVs derived from LPS-preconditioned dental follicle cells (DFC-EVs) inhibited reactive oxygen species (ROS) formation and promoted macrophage polarization toward the M2 phenotype through the activation of ROS/ERK signaling.52 Moreover, TNF-α pretreatment promoted the exosomal expression of CD73 to induce M2 macrophage polarization, thereby facilitating inflammation and periodontal bone preservation.53 Similarly, TNF-α-pretreated MSC-EVs decreased the levels of proinflammatory markers including IL-1β and inducible nitric oxide synthase (iNOS), while increasing the expression of reparatory markers such as Arg1 and CD206. This effect is potentially mediated by miRNAs within the EVs, promoting an immunomodulatory role.54 On the basis of these findings, Deng et al55 achieved sustained modulation of macrophage polarization by immobilizing MSC-EVs in hydrogels through biotin-avidin systems, reducing the expression of M1 polarization-associated proteins like iNOS and chemokine receptor 7 (CCR7), and increasing the levels of the M2 polarization-associated protein CD206, thereby promoting periodontal tissue repair. These studies deepen our understanding of the mechanisms by which EVs influence macrophage polarization and highlight their potential in modulating immune responses and promoting tissue repair.

T cells

T cells are among the most abundant cells in healthy and diseased periodontal tissues.56 In patients with periodontitis, the balance between Th17 cells and Tregs in the peripheral blood is disrupted, resulting in increased inflammation and alleviated tissue damage.44 MSC-EVs have considerable potential in the modulation of immune responses, particularly in the induction of Tregs and reduction of T-cell proliferation as well as related interferon gamma (IFN-γ) release.57 MSC-EVs directly influence T-cell activation and differentiation by expressing specific proteins at elevated levels. Notably, Wharton’s jelly-derived MSCs, when stimulated with IFN‐γ, increased the secretion of EVs carrying programmed death-ligand 1 (PD-L1). PD-L1 expressed by MSC-EVs interacts with programmed death receptor 1 (PD1) on T cells, thereby influencing T-cell receptor (TCR)-mediated activation and contributing to the mitigation of acute graft-versus-host disease (aGvHD).58 Furthermore, MSC-EVs express galectin-1 and transforming growth factor beta (TGF-β), which have been shown to induce the apoptosis of activated T cells and encourage the generation of Tregs.59 In contrast, MSC-EVs can indirectly modulate T-cell differentiation by modulating the cellular behavior of macrophages. In vitro studies demonstrated that EVs from human umbilical cord MSCs (HUCMSCs) could suppress the proliferation of CD4+ T cells and enhance the generation of Tregs by educating macrophages toward the M2-like phenotype. From a mechanistic perspective, miR-100-5p enriched in EVs promoted M2 macrophage polarization and diminished their ability to suppress T cells and promote Treg expansion. Knocking down miR-100-5p in MSC-sEVs partially inhibited the ability of HUCMSC-sEVs to promote M2 macrophage and Treg expansion.60

Other immune cells

Following inflammatory stimulation, MSC-EVs are also internalized more by innate immune effector cells (IECs), possibly because of their heightened expression of intercellular adhesion molecule-1 (ICAM-1), which allows MSC-EVs to bind to target cells.61 Once internalized by IECs, MSC-EVs exert immunomodulatory effects by regulating the expression of inflammatory factors. For example, treatment with MSC-EVs hindered antigen uptake by immature DCs and suppressed their maturation, leading to reduced levels of the maturation and activation markers CD83, CD38, and CD80. Moreover, this treatment results in decreased levels of the proinflammatory cytokines IL-6 and IL-12p70 while simultaneously increasing the production of the anti-inflammatory cytokine TGF-β. Additionally, MSC-EV-incubated DCs presented reduced CCR7 expression following LPS stimulation and a notably diminished ability to migrate toward the CCR7 ligand C-C motif chemokine ligand 21 (CCL21). The underlying mechanism involves the ability of miR-21-5p within MSC-EVs to degrade CCR7, and miR-21-5p mimic transfection significantly decreases the ability of DCs to migrate toward CCL21.62 Another study indicated that MSC-EVs promoted the content and secretion of IL-6 and interleukin-10 (IL-10) by DCs, resulting in a decreased population of Th17 cells and an increased number of Treg cells. Furthermore, MSC-EV-treated T cells exhibited elevated production of anti-inflammatory cytokines, including IL-10, TGF-β, and IL-6.63

Repair-promoting functions of MSC-EVs in periodontal tissue regeneration

Many studies have emphasized that MSC derivatives, such as EVs, Exos, matrix vesicles (MVs), and apoptotic bodies (ABs), can be used as alternatives to MSCs to promote periodontal tissue regeneration because of their strong repair-promoting functions.48,64-66 MSC-EVs can exert multiple bioactivities, including pro-osteogenic/cementogenic, proangiogenic, and pro-stem cell migratory, proliferative and multilineage differentiation effects, to facilitate the repair of complex periodontal tissues.67 This section summarizes the repair-promoting functions of EVs in different periodontal tissues and the underlying mechanisms (Figure 3 and Table 1).

Table 1.

Summary of EVs used in periodontal tissue regeneration.

Cell sourceEVs typeIsolation protocolCargoesEffectsMechanismsRef.
BMSCEVsGradient ultracentrifugationBgnPromote osteoblast proliferation, migration, differentiation, and mineralizationPI3K/AKT81
M2
Macrophage
ExosDifferential centrifugationLet-7f-5pPromote cementoblast mineralizationDownregulation of Ckip-1 by Let-7f-5p activates mitochondrial biogenesis118
MSCEVsDifferential centrifugationmiR-612Promote ECs migration, proliferation and angiogenesismiR-612/TP53/HIF-1α121
BMSCEVsPolymer precipitation, centrifugation techniquesBMP2Induce osteoinduction and bone regenerationPhosphorylation of Smad 1/5/895
DPSCAbsDifferential centrifugationTUFM,
TFEB
Promote ECs proliferation and angiogenesisThe TFEB-induced autophagy-lysosome pathway125
BMSCEVsPolymer precipitation, centrifugation techniquesOPGPromote PDLSCs proliferation, migration and differentiation;
Inhibit osteoclast activity; promote M2 macrophage polarization
OPG/RANKL/RANK84
PDLSCExosDifferential centrifugationmiR-31-5pInhibit osteoclast formation and reduce bone destructionmiR-31-5p/eNOS87
GMSCExosImmunoprecipitation, ultracentrifugationmiR-1260b/CD73Promote M2 macrophage polarization and enhance wound healing;
Promote osteogenesis and inhibit bone loss
Wnt5a/JNK/RANKL53
DPSCEVsUltracentrifugationmiR-1246Promote osteogenesisUpregulation of miR-1246107
SCAPEVsUltracentrifugation, differential centrifugationmiR-935Promote osteoblast differentiation of PDLSCs; inhibit inflammation and bone resorptionUpregulation of miR-935109
SHEDExosUltracentrifugation, differential centrifugationβ-catenin, Wnt3a, BMP2Promote osteoblast differentiation of PDLSCsWnt/β-catenin;
BMP/Smad (the phosphorylation of Smad 1/5/8)
98
BMSCEVsPolymer precipitation,ultrafiltrationmiR-424Promote bone formationPhosphorylation of Smad 1/5/896
BMSCExos UltracentrifugationDPT-C, miR-26aPromote osteogenesis, proliferation and migration of BMSCsmTOR pathway/Wnt/autophagy91
BMSCAbsDifferential centrifugationmiR-223-3PInhibit osteoclast activity and alleviate bone destructionTarget Itgb164
BMSC,
ASC
ExosGradient ultracentrifugationlet-7a-5p, let-7c-5p, miR-328a-5p, miR-31a-5pPromote osteoblast differentiation of BMSCsTargeted Acvr2b/Acvr1 and regulate the balance of Bmpr2/Acvr2b for Bmpr-elicited Smad1/5/9 phosphorylation97
BMSCExos UltracentrifugationmiR-130aPromote ECs migrationPTEN/AKT; Wnt/β-catenin NF-κB/AKT124
SHEDEVsDifferential centrifugationChemotactic Factors
(TGF-β1, IGF-1, FGF-2, PDGF-BB)
Promote migration of BMSCsChemotactic effect129
DPSCEVsDifferential centrifugationmiR-758-5pPromote osteoblast differentiation of PDLSCsmiR-758-5p/LMBR1/BMP2/494
PDLSCExosUltracentrifugationmiR-17-5pIncrease VEGFA and promote tube formation of ECsDownregulation of miR-17-5p122
DPSCEVsUltracentrifugationmiR-378aPromote ECs migration, proliferation, tube formation and angiogenesismiR-378a/Sufu/Hedgehog/Gli1123
Cell sourceEVs typeIsolation protocolCargoesEffectsMechanismsRef.
BMSCEVsGradient ultracentrifugationBgnPromote osteoblast proliferation, migration, differentiation, and mineralizationPI3K/AKT81
M2
Macrophage
ExosDifferential centrifugationLet-7f-5pPromote cementoblast mineralizationDownregulation of Ckip-1 by Let-7f-5p activates mitochondrial biogenesis118
MSCEVsDifferential centrifugationmiR-612Promote ECs migration, proliferation and angiogenesismiR-612/TP53/HIF-1α121
BMSCEVsPolymer precipitation, centrifugation techniquesBMP2Induce osteoinduction and bone regenerationPhosphorylation of Smad 1/5/895
DPSCAbsDifferential centrifugationTUFM,
TFEB
Promote ECs proliferation and angiogenesisThe TFEB-induced autophagy-lysosome pathway125
BMSCEVsPolymer precipitation, centrifugation techniquesOPGPromote PDLSCs proliferation, migration and differentiation;
Inhibit osteoclast activity; promote M2 macrophage polarization
OPG/RANKL/RANK84
PDLSCExosDifferential centrifugationmiR-31-5pInhibit osteoclast formation and reduce bone destructionmiR-31-5p/eNOS87
GMSCExosImmunoprecipitation, ultracentrifugationmiR-1260b/CD73Promote M2 macrophage polarization and enhance wound healing;
Promote osteogenesis and inhibit bone loss
Wnt5a/JNK/RANKL53
DPSCEVsUltracentrifugationmiR-1246Promote osteogenesisUpregulation of miR-1246107
SCAPEVsUltracentrifugation, differential centrifugationmiR-935Promote osteoblast differentiation of PDLSCs; inhibit inflammation and bone resorptionUpregulation of miR-935109
SHEDExosUltracentrifugation, differential centrifugationβ-catenin, Wnt3a, BMP2Promote osteoblast differentiation of PDLSCsWnt/β-catenin;
BMP/Smad (the phosphorylation of Smad 1/5/8)
98
BMSCEVsPolymer precipitation,ultrafiltrationmiR-424Promote bone formationPhosphorylation of Smad 1/5/896
BMSCExos UltracentrifugationDPT-C, miR-26aPromote osteogenesis, proliferation and migration of BMSCsmTOR pathway/Wnt/autophagy91
BMSCAbsDifferential centrifugationmiR-223-3PInhibit osteoclast activity and alleviate bone destructionTarget Itgb164
BMSC,
ASC
ExosGradient ultracentrifugationlet-7a-5p, let-7c-5p, miR-328a-5p, miR-31a-5pPromote osteoblast differentiation of BMSCsTargeted Acvr2b/Acvr1 and regulate the balance of Bmpr2/Acvr2b for Bmpr-elicited Smad1/5/9 phosphorylation97
BMSCExos UltracentrifugationmiR-130aPromote ECs migrationPTEN/AKT; Wnt/β-catenin NF-κB/AKT124
SHEDEVsDifferential centrifugationChemotactic Factors
(TGF-β1, IGF-1, FGF-2, PDGF-BB)
Promote migration of BMSCsChemotactic effect129
DPSCEVsDifferential centrifugationmiR-758-5pPromote osteoblast differentiation of PDLSCsmiR-758-5p/LMBR1/BMP2/494
PDLSCExosUltracentrifugationmiR-17-5pIncrease VEGFA and promote tube formation of ECsDownregulation of miR-17-5p122
DPSCEVsUltracentrifugationmiR-378aPromote ECs migration, proliferation, tube formation and angiogenesismiR-378a/Sufu/Hedgehog/Gli1123

Abbreviations: ABs, apoptotic bodies; AKT, protein kinase B; ASC(ADSC), adipose-derived stem cells; BMP2, bone morphogenetic protein 2; BMSC, bone mesenchymal stem cells; DPSC, dental pulp stem cells; ECs, endothelial cells; EVs, extracellular vesicles; Exos, exosomes; GMSC, gingival mesenchymal stem cells; HIF-1α, hypoxia-inducible factor 1-alpha; JNK, c-Jun N-terminal kinaseMSC, mesenchymal stem cells; OPG, osteoprotegerin; PDLSC, periodontal ligament stem cells; PI3K, phosphatidylinositide 3-kinase; RANK, receptor activator of nuclear factor κ; RANKL, receptor activator of nuclear factor κB ligand; SCAP, stem cells from the apical papilla; SHED, stem cells from human exfoliated deciduous teeth; TUFM, Tu translation elongation factor; VEGFA, vascular endothelial growth factor A; Wnt3a, wingless protein 3a.

Table 1.

Summary of EVs used in periodontal tissue regeneration.

Cell sourceEVs typeIsolation protocolCargoesEffectsMechanismsRef.
BMSCEVsGradient ultracentrifugationBgnPromote osteoblast proliferation, migration, differentiation, and mineralizationPI3K/AKT81
M2
Macrophage
ExosDifferential centrifugationLet-7f-5pPromote cementoblast mineralizationDownregulation of Ckip-1 by Let-7f-5p activates mitochondrial biogenesis118
MSCEVsDifferential centrifugationmiR-612Promote ECs migration, proliferation and angiogenesismiR-612/TP53/HIF-1α121
BMSCEVsPolymer precipitation, centrifugation techniquesBMP2Induce osteoinduction and bone regenerationPhosphorylation of Smad 1/5/895
DPSCAbsDifferential centrifugationTUFM,
TFEB
Promote ECs proliferation and angiogenesisThe TFEB-induced autophagy-lysosome pathway125
BMSCEVsPolymer precipitation, centrifugation techniquesOPGPromote PDLSCs proliferation, migration and differentiation;
Inhibit osteoclast activity; promote M2 macrophage polarization
OPG/RANKL/RANK84
PDLSCExosDifferential centrifugationmiR-31-5pInhibit osteoclast formation and reduce bone destructionmiR-31-5p/eNOS87
GMSCExosImmunoprecipitation, ultracentrifugationmiR-1260b/CD73Promote M2 macrophage polarization and enhance wound healing;
Promote osteogenesis and inhibit bone loss
Wnt5a/JNK/RANKL53
DPSCEVsUltracentrifugationmiR-1246Promote osteogenesisUpregulation of miR-1246107
SCAPEVsUltracentrifugation, differential centrifugationmiR-935Promote osteoblast differentiation of PDLSCs; inhibit inflammation and bone resorptionUpregulation of miR-935109
SHEDExosUltracentrifugation, differential centrifugationβ-catenin, Wnt3a, BMP2Promote osteoblast differentiation of PDLSCsWnt/β-catenin;
BMP/Smad (the phosphorylation of Smad 1/5/8)
98
BMSCEVsPolymer precipitation,ultrafiltrationmiR-424Promote bone formationPhosphorylation of Smad 1/5/896
BMSCExos UltracentrifugationDPT-C, miR-26aPromote osteogenesis, proliferation and migration of BMSCsmTOR pathway/Wnt/autophagy91
BMSCAbsDifferential centrifugationmiR-223-3PInhibit osteoclast activity and alleviate bone destructionTarget Itgb164
BMSC,
ASC
ExosGradient ultracentrifugationlet-7a-5p, let-7c-5p, miR-328a-5p, miR-31a-5pPromote osteoblast differentiation of BMSCsTargeted Acvr2b/Acvr1 and regulate the balance of Bmpr2/Acvr2b for Bmpr-elicited Smad1/5/9 phosphorylation97
BMSCExos UltracentrifugationmiR-130aPromote ECs migrationPTEN/AKT; Wnt/β-catenin NF-κB/AKT124
SHEDEVsDifferential centrifugationChemotactic Factors
(TGF-β1, IGF-1, FGF-2, PDGF-BB)
Promote migration of BMSCsChemotactic effect129
DPSCEVsDifferential centrifugationmiR-758-5pPromote osteoblast differentiation of PDLSCsmiR-758-5p/LMBR1/BMP2/494
PDLSCExosUltracentrifugationmiR-17-5pIncrease VEGFA and promote tube formation of ECsDownregulation of miR-17-5p122
DPSCEVsUltracentrifugationmiR-378aPromote ECs migration, proliferation, tube formation and angiogenesismiR-378a/Sufu/Hedgehog/Gli1123
Cell sourceEVs typeIsolation protocolCargoesEffectsMechanismsRef.
BMSCEVsGradient ultracentrifugationBgnPromote osteoblast proliferation, migration, differentiation, and mineralizationPI3K/AKT81
M2
Macrophage
ExosDifferential centrifugationLet-7f-5pPromote cementoblast mineralizationDownregulation of Ckip-1 by Let-7f-5p activates mitochondrial biogenesis118
MSCEVsDifferential centrifugationmiR-612Promote ECs migration, proliferation and angiogenesismiR-612/TP53/HIF-1α121
BMSCEVsPolymer precipitation, centrifugation techniquesBMP2Induce osteoinduction and bone regenerationPhosphorylation of Smad 1/5/895
DPSCAbsDifferential centrifugationTUFM,
TFEB
Promote ECs proliferation and angiogenesisThe TFEB-induced autophagy-lysosome pathway125
BMSCEVsPolymer precipitation, centrifugation techniquesOPGPromote PDLSCs proliferation, migration and differentiation;
Inhibit osteoclast activity; promote M2 macrophage polarization
OPG/RANKL/RANK84
PDLSCExosDifferential centrifugationmiR-31-5pInhibit osteoclast formation and reduce bone destructionmiR-31-5p/eNOS87
GMSCExosImmunoprecipitation, ultracentrifugationmiR-1260b/CD73Promote M2 macrophage polarization and enhance wound healing;
Promote osteogenesis and inhibit bone loss
Wnt5a/JNK/RANKL53
DPSCEVsUltracentrifugationmiR-1246Promote osteogenesisUpregulation of miR-1246107
SCAPEVsUltracentrifugation, differential centrifugationmiR-935Promote osteoblast differentiation of PDLSCs; inhibit inflammation and bone resorptionUpregulation of miR-935109
SHEDExosUltracentrifugation, differential centrifugationβ-catenin, Wnt3a, BMP2Promote osteoblast differentiation of PDLSCsWnt/β-catenin;
BMP/Smad (the phosphorylation of Smad 1/5/8)
98
BMSCEVsPolymer precipitation,ultrafiltrationmiR-424Promote bone formationPhosphorylation of Smad 1/5/896
BMSCExos UltracentrifugationDPT-C, miR-26aPromote osteogenesis, proliferation and migration of BMSCsmTOR pathway/Wnt/autophagy91
BMSCAbsDifferential centrifugationmiR-223-3PInhibit osteoclast activity and alleviate bone destructionTarget Itgb164
BMSC,
ASC
ExosGradient ultracentrifugationlet-7a-5p, let-7c-5p, miR-328a-5p, miR-31a-5pPromote osteoblast differentiation of BMSCsTargeted Acvr2b/Acvr1 and regulate the balance of Bmpr2/Acvr2b for Bmpr-elicited Smad1/5/9 phosphorylation97
BMSCExos UltracentrifugationmiR-130aPromote ECs migrationPTEN/AKT; Wnt/β-catenin NF-κB/AKT124
SHEDEVsDifferential centrifugationChemotactic Factors
(TGF-β1, IGF-1, FGF-2, PDGF-BB)
Promote migration of BMSCsChemotactic effect129
DPSCEVsDifferential centrifugationmiR-758-5pPromote osteoblast differentiation of PDLSCsmiR-758-5p/LMBR1/BMP2/494
PDLSCExosUltracentrifugationmiR-17-5pIncrease VEGFA and promote tube formation of ECsDownregulation of miR-17-5p122
DPSCEVsUltracentrifugationmiR-378aPromote ECs migration, proliferation, tube formation and angiogenesismiR-378a/Sufu/Hedgehog/Gli1123

Abbreviations: ABs, apoptotic bodies; AKT, protein kinase B; ASC(ADSC), adipose-derived stem cells; BMP2, bone morphogenetic protein 2; BMSC, bone mesenchymal stem cells; DPSC, dental pulp stem cells; ECs, endothelial cells; EVs, extracellular vesicles; Exos, exosomes; GMSC, gingival mesenchymal stem cells; HIF-1α, hypoxia-inducible factor 1-alpha; JNK, c-Jun N-terminal kinaseMSC, mesenchymal stem cells; OPG, osteoprotegerin; PDLSC, periodontal ligament stem cells; PI3K, phosphatidylinositide 3-kinase; RANK, receptor activator of nuclear factor κ; RANKL, receptor activator of nuclear factor κB ligand; SCAP, stem cells from the apical papilla; SHED, stem cells from human exfoliated deciduous teeth; TUFM, Tu translation elongation factor; VEGFA, vascular endothelial growth factor A; Wnt3a, wingless protein 3a.

The therapeutic effects of MSC-EVs on periodontitis. MSC-EVs deliver multiple cargoes (eg, miRNAs, proteins, etc.) to different recipient cells, including osteoblasts, cementoblasts, MSCs, and ECs. Multiple underlying mechanisms are involved in MSC-EV-mediated osteogenesis, angiogenesis, cell migration/proliferation, and cementogenesis. These signaling pathways regulate biological processes in recipient cells to exert therapeutic effects on periodontitis. Abbreviations: AKT, protein kinase B; AMP, adenosine monophosphate; AMPK, AMP-activated protein kinase; BMP2, bone morphogenetic protein 2; Ckip-1, casein kinase 2 interacting protein-1 EGF, epidermal growth factor; ERK, extracellular signal-regulated kinase; HIF-1α, hypoxia-inducible factor 1-alpha; IGFBP-3/5/6, insulin-like growth factor binding protein-3/5/6; MAPK, mitogen-activated protein kinase; NF-κB, the nuclear factor kappa-light-chain-enhancer of activated B cells; OPG, osteoprotegerin; p-AKT, the phosphorylation of AKT; p-AMPK, the phosphorylation of AMPK; PI3K, phosphoinositide 3-kinase; p-JNK, the phosphorylation of c-Jun N-terminal kinase; PKC, protein kinase C; PLC, phospholipase C; p-Smad1/5/8/9, the phosphorylation of Smad1/5/8/9; RANK, receptor activator of nuclear factor κB; RANKL, the receptor activator of nuclear factor κ ligand; STAT1, signal transducer and activator of transcription 1; TUFM, Tu translation elongation factor; VCAM-1, vascular cell adhesion molecule-1; VEGF, vascular endothelial growth factor; Wnts, wingless protein.
Figure 3.

The therapeutic effects of MSC-EVs on periodontitis. MSC-EVs deliver multiple cargoes (eg, miRNAs, proteins, etc.) to different recipient cells, including osteoblasts, cementoblasts, MSCs, and ECs. Multiple underlying mechanisms are involved in MSC-EV-mediated osteogenesis, angiogenesis, cell migration/proliferation, and cementogenesis. These signaling pathways regulate biological processes in recipient cells to exert therapeutic effects on periodontitis. Abbreviations: AKT, protein kinase B; AMP, adenosine monophosphate; AMPK, AMP-activated protein kinase; BMP2, bone morphogenetic protein 2; Ckip-1, casein kinase 2 interacting protein-1 EGF, epidermal growth factor; ERK, extracellular signal-regulated kinase; HIF-1α, hypoxia-inducible factor 1-alpha; IGFBP-3/5/6, insulin-like growth factor binding protein-3/5/6; MAPK, mitogen-activated protein kinase; NF-κB, the nuclear factor kappa-light-chain-enhancer of activated B cells; OPG, osteoprotegerin; p-AKT, the phosphorylation of AKT; p-AMPK, the phosphorylation of AMPK; PI3K, phosphoinositide 3-kinase; p-JNK, the phosphorylation of c-Jun N-terminal kinase; PKC, protein kinase C; PLC, phospholipase C; p-Smad1/5/8/9, the phosphorylation of Smad1/5/8/9; RANK, receptor activator of nuclear factor κB; RANKL, the receptor activator of nuclear factor κ ligand; STAT1, signal transducer and activator of transcription 1; TUFM, Tu translation elongation factor; VCAM-1, vascular cell adhesion molecule-1; VEGF, vascular endothelial growth factor; Wnts, wingless protein.

Pro-osteogenic/cementogenic effects

Periodontitis is a long-lasting inflammatory disease characterized by the permanent loss of tissues that support the structures of teeth. If left untreated, this condition can lead to tooth loss68; thus, restoring the lost supporting tissues is the primary objective of periodontitis treatment. Achieving complete periodontal tissue regeneration is a challenging objective, which necessitates addressing the complex anatomical structure of periodontal tissue69 and managing the uncontrolled inflammatory microenvironment.70 Notably, the delivery of bioactive cargoes, which are involved in the mediation of pro-osteogenic and cementogenic effects, to recipient cells is emerging as a promising cell-free therapeutic strategy to address this challenge. MSC-EVs have been shown to be effective in various animal models of periodontitis.54,71-78

Prior to the introduction of EVs into the clinical setting, a comprehensive understanding of the underlying mechanisms, with different pathways playing pivotal roles in the osteogenesis of PDLSCs, is essential. The phosphatidylinositide 3-kinase/protein kinase B (PI3K/AKT) pathway is known to mediate bone metabolism,79 and almost all types of EVs derived from different stem cells (eg, MSCs, PDLSCs, BMSCs and HUCMSCs) can promote the bone-generating differentiation of PDLSCs by triggering this process.80-82 For example, Zhai et al.80 demonstrated that MSC-EVs could enter MSCs via caveolin-1 and promote osteogenesis by stimulating the PI3K/AKT/MAPK signaling pathway. Similarly, adipose-derived stem cell (ADSC)-derived EVs may promote the osteogenesis of MSCs by eliminating cellular ROS through activation of the PI3K/AKT/NRF2 pathway and reducing mitochondrial changes/DNA damage.83 In addition to the PI3K/AKT pathway, the RANKL-RANK-OPG axis is another critical regulatory axis for bone homeostasis, as noted above. Numerous studies have demonstrated that EVs extracted from dental follicle stem cells (DFSCs), BMSCs, gingival mesenchymal stem cells (GMSCs) or cementocytes can inhibit osteoclast activity and promote osteoblast differentiation by decreasing the ratio of RANKL/OPG expression52,84 or inhibiting related key signals such as NF-κB,85,86 wingless protein (Wnt) 5a or the phosphorylation of JNK.53 Some miRNAs, such as those in the miR-223-3p/Itgb1 axis,64 the miR-31-5p/eNOS signaling pathway87 and the miR-503-3p/HPSE axis,88 are also involved in this process. Furthermore, miRNA-involved axes play crucial roles in promoting osteogenesis and inhibiting osteoclastogenesis; for example, BMSC-Exos delivered high levels of miR-935 to stimulate osteoblast proliferation and differentiation by inhibiting signal transducer and activator of transcription 1 (STAT1),68 and PDLSC-Exos promoted osteogenic differentiation by inhibiting TNF via Exos carrying miR-181d-5p.89 Among these osteogenesis-related pathways, Wnt signaling represents a classic pathway involved in maintaining periodontal tissue homeostasis90; however, the role of this pathway remains controversial. Most relevant experiments have demonstrated that stimulation of the Wnt/β-catenin pathway is a prerequisite for the pro-osteogenic effects of EVs from BMSCs and PDLSCs.91,92 However, a study conducted by Lei et al.93 revealed that excessive activation of Wnt signaling in PDLSCs impaired osteogenesis during periodontitis. These contradictory results may be attributed to the different origins of MSCs and varying culture conditions (with or without inflammatory factors),93 which necessitates further investigation in the future. Moreover, the bone morphogenetic protein (BMP)/Smad pathway is equally essential for osteogenesis, and EVs from the inflamed microenvironment increase the osteoblast differentiation of PDLSCs partly by inhibiting LMBR1-targeting miR-758-5p via BMP signaling.94 In addition, because of the targeting role of multicomponent EV miRNAs and proteins (eg, BMP2, let-7a-5p and miR-424), EVs derived from various types of MSCs [eg, BMSCs,95,96 ADSCs,97 and stem cells from human exfoliated deciduous teeth (SHEDs)98] can activate the phosphorylation of Smad 1/5/8/9 to promote bone regeneration, indicating that EVs share a common underlying mechanism of EV-induced osteogenesis. In addition, research progress on other mechanisms offers promising avenues for the development of bone regeneration in periodontitis, including the activation of the phospholipase C/protein kinase C/ (PLC/PKC/MAPK) pathway,69 the p38 MAPK signaling pathway,99 the adenosine monophosphate-activated protein kinase (AMPK) pathway100 and the AKT-mediating systems.101-103 In summary, MSC-derived EVs can activate various osteogenesis-related pathways, resulting in accelerated bone regeneration, which is facilitated by the release of multiple bioactive cargoes.

In addition, multiple mechanisms, including autophagy104 and mitochondrial functions,105 have been shown to participate in MSC-EV-strengthened osteogenesis. For example, autophagy is a cellular mechanism that allows cells to break down and recycle proteins and organelles to preserve the intracellular balance, is strengthened by hypoxia and then induces rapid release of BMSC-EVs.104 The transplantation of BMSC-EVs generated under hypoxic conditions significantly increased the reconstruction of defects in alveolar bone.104 Additionally, Dai et al.105 demonstrated that Exos obtained from inflammatory PDLSCs can safeguard their mitochondrial functions by increasing the mitochondrial membrane potential and reducing oxidative stress levels, thereby facilitating the osteogenic differentiation of PDLSCs. Furthermore, EVs enriched with specific miRNAs promote osteoclastogenesis by rebalancing the coordinated activities of osteoblasts and osteoclasts, which is achieved through the regulation of relevant gene expression. These altered miRNAs include upregulated miRNAs [eg, hsa-miR-29c-5p,106 miR-1246,107 miR-21-5p,108 miR-935,109 and miR-335-5p110] and downregulated miRNAs [eg, hsa-miR-31-3p, hsa-miR-31–3p, hsa-miR-221–3p, hsa-miR-183–5p and hsa-miR-503–5p,106 and hsa-miR-125b-5p111], which not only promote the expression of osteoblast-related genes (eg, RUNX, OCN, and BMP) but also inhibit the expression of osteoclast genes, including TRAP, NFATc1, and Cathepsin K. Nevertheless, the quantity of newly formed bone must also be considered. Bone remodeling is an essential aspect of bone repair and involves unhealthy bone resorption112 and the inhibition of osteogenesis.113 Numerous studies have confirmed that miRNAs and long noncoding RNAs (lncRNAs) are important posttranscriptional regulators that participate in the modulation of bone metabolism, such as the upregulation of miR-28114 and lncRNA-MALAT1110 or the downregulation of miR-124,112 which can increase osteoclast viability; high expression of miR-34c-5p can significantly suppress osteogenesis via the special AT-rich sequence-binding protein 2 (SATB2)/ERK pathway.113 Consequently, numerous and various mechanisms are involved in the pro-osteogenic/cementogenic effects of MSC-EVs, but the core of all relevant research is the inhibition of osteoclast generation and differentiation, along with the promotion of osteoblast activity. Therefore, regulating the balance between osteoclasts and osteoblasts is a promising strategy to facilitate the advancement of periodontitis treatment. Additionally, almost all of the above studies were conducted in animal models of periodontitis, but actual human periodontitis is much more complex than this situation is. Therefore, clinical trials on the ability of MSC-EVs to promote osteogenesis/cementogenesis in periodontitis patients need to be conducted in the future.

Although there is a thin layer of mineralized tissue covering the teeth roots, the cementum plays a crucial role in anchoring the teeth within the alveolar socket. This anchoring helps maintain the stability and health of the teeth, which is achieved through the embedded collagen fibers.115 Therefore, the regeneration of cementum is widely regarded as the gold standard for periodontal tissue regeneration.116 Although cementum is similar to bone in several aspects,86,90 differences in its regeneration arise from its unique anatomical position and structure.65 Cementoblasts are the principal effector cells responsible for synthesizing cementum, which plays a pivotal role in regulating cementum repair.117 Therefore, it is crucial to stimulate cementoblasts to migrate toward root surfaces and promote their cementogenic activity. Exos derived from PDLSCs were shown to increase proliferation, migration, and cementogenesis by triggering the PI3K/AKT signaling pathway.64 In addition to Exos derived from MSCs, other sources of Exos, such as M2-like macrophages, promote cementum regeneration. A study conducted by Huang et al.118 demonstrated that Exos from Ckip-1-silenced macrophages (sh-Ckip-1-Exos) effectively promoted cementogenesis and rescued P. gingivalis-suppressed cementoblast mineralization. The underlying mechanism is that let-7f-5p delivered by sh-Ckip-1-Exos targets Ckip-1, thereby promoting mitochondrial biogenesis in a PGC-1α-dependent manner.118 Nevertheless, research on cementum regeneration is lacking. Identification of accessible methods to establish an evaluable cementum-defect model and more characteristic markers to determine the regeneration outcomes of cementum in regeneration systems is urgently needed.

Proangiogenic effects

Vascularization has been demonstrated to be a prerequisite for bone defect repair.119 Mounting evidence has indicated that MSC-EVs can directly or indirectly modulate revascularization to support periodontal tissue regeneration by carrying a panel of miRNAs (eg, increased presence of miR-612 and miR-143-3p and decreased expression levels of miR-15b-5p, miR-16-5p and the let-7-5p family).120 For example, MSC-EVs carry miR-612 to promote displacement, proliferation and endothelial cell (EC) tube development by regulating the miR-612/TP53/hypoxia-inducible factor 1-alpha (HIF-1α) axis.121 Notably, vascular endothelial growth factor (VEGF) is the most potent agent that participates in the modulation of vascular ECs, with its expression being increased by the downregulation of miR-17-5p in PDLSC-Exos.122 Additionally, DPSC-EVs expressing miR-378a have been shown to decrease the expression of Sufu to promote angiogenesis by activating Hedgehog/Gli1 signaling.123 Furthermore, Exos derived from BMSCs strongly promoted the migration of ECs, which was attributed to miR-130a and its downstream effects on the PTEN/AKT pathway.124 More importantly, Exos from SHEDs augment the pro-regeneration function of ECs by increasing the levels of angiogenic molecules (eg, VEGF-A, KDR, FGF2, and SDF-1) through the regulation of the AMPK signaling pathway,100 which is similar to the AKT/eNOS pathway in PDLSC-Exos.101 Moreover, Li et al.125 demonstrated that ABs from DPSCs promoted EC autophagy by transferring mitochondrial Tu translation elongation factor (TUFM), thereby increasing the angiogenic potential of ECs. Similarly, HUCMSC-Exos119 and DPSC-EVs126 were shown to facilitate the migration and proliferation of ECs in rat alveolar bone defect models. Overall, angiogenesis has been reported to be associated with osteogenesis, in which the functions of ECs and VEGF play essential roles. Therefore, regulating the activity of ECs and VEGF might be a candidate strategy for periodontitis treatment.

Pro-stem cell migratory and proliferative effects

In addition to their roles in osteogenesis and angiogenesis, the migration and proliferation of MSCs play equally significant roles in the regeneration of tissue. As in osteogenesis and angiogenesis, various signaling pathways participate in the modulation of MSC proliferation and migration, including the PI3K/AKT pathway,81 the Wnt/β-catenin pathway,92 the RANKL/RANK/OPG axis84 and the MAPK pathway.99 For example, MSC-Exos could increase PDLSC movement and multiplication via CD73-induced activation of adenosine receptors on AKT and ERK signaling.102 Furthermore, Liu et al104 reported that a hypoxic microenvironment promoted MSC homing by increasing the production of proteins related to autophagy (eg, Beclin-1 and LC3II/LC3I), thereby facilitating the secretion of various chemokines [eg, insulin-like growth factor binding protein (IGFBP)-3, IGFBP-6, IGFBP-5, vascular cell adhesion molecule (VCAM)-1, epidermal growth factor (EGF) and VEGF]. EVs derived from various MSCs increase the quantity of specific cytokines, including miRNAs (eg, hsa-miR-29c-5p, hsa-miR-378a-5p, hsa-miR-10b-5p and hsa-miR-9-3p),106 nuclear factor I/C (NFIC)127 and chemotactic factors (eg, TGF-β1, IGF-1, FGF-2, and PDGF-BB),126 which promote the motility of their parental MSCs. In conclusion, MSC-EVs promoted cell migration and proliferation to facilitate complete periodontal tissue regeneration. Hence, the migration of MSCs cannot be ignored when developing therapeutic approaches to promote periodontal regeneration.

However, it remains unclear which types of MSC-EVs exert the most pronounced pro-osteogenic and angiogenic effects among the various types of MSCs, as numerous factors influence their efficacy, including their origin, isolation methods, and culture conditions. Research on the diverse sources of MSC-EVs is limited; however, recent findings suggest that, compared with ADSC-Exos, BMSC-Exos have greater productivity and osteogenic induction.97 Compared with GMSCs-EVs, BMSC-EVs do not demonstrate the same advantages.70 Additionally, osteoblasts and SHEDs showed superior performance compared with PDLSCs/gingival fibroblasts and DPSCs, respectively.71,72 In terms of different isolation methods and culture conditions, Yi et al69 compared the biological properties of media microvesicles (MMVs) isolated from culture supernatant and collagenase-released microvesicles (CRMVs) obtained from collagenase-digested cell suspensions. Their results revealed that CRMVs exhibited superior promotion of cell proliferation, migration, mineralization, and osteogenesis.69 Additionally, a three-dimensional (3D) culture system was shown to produce greater amounts of Exos than a traditional two-dimensional (2D) system did.68 Furthermore, compared with those derived from normoxic MSCs, EVs derived from periodontal DPSCs,126 hypoxic BMSCs55 and hypoxic MSCs121 exhibited increased potential for promoting osteogenesis and angiogenesis. Moreover, Qiu et al101 reported that Exos released by neonatal serum-incubated MSCs exhibited a more pronounced angiogenic effect than those released by adult serum-incubated MSCs. In summary, it is imperative to systematically compare the bioactivities of EVs derived from different MSC sources under uniform research conditions to expedite the clinical development of MSC-EVs for tissue regeneration.

Clinical application of EVs in periodontal tissue regeneration

The various advantages of MSC-EVs have paved the way for their use in periodontal tissue regeneration. Nevertheless, EV-based therapies are typically limited by their transient half-life and rapid clearance from the site of administration.70,128 For increased therapeutic performance of EVs, researchers must augment their bioactive properties or ensure prolonged retention of MSC-EVs at targeted sites. This phenomenon can be achieved through diverse strategies, such as preconditioning of parental MSCs, engineering of MSC-derived EVs, and the development of localized delivery systems (Figure 4). Emerging research has principally explored the potential of preconditioning MSCs to modify key metabolites and proteins in MSC-EVs linked to anti-inflammatory and immunoregulatory functions.54 For example, EVs isolated from MSCs preconditioned with TNF-α,53,54 gallic acid (GA),105 or curcumin92 or with the silencing of casein kinase 2 interacting protein-1 (Ckip-1)118 or hypoxia55,121 showed an increased ability to enhance the displacement, reproduction and osteogenic function of MSCs. In addition, the use of engineered MSC-EVs has demonstrated good potential for increasing EV bioactivity, which allows EVs to constitutively express certain molecules to promote regenerative functions. For example, Huang et al95 genetically modified BMSCs by overexpressing BMP2, consequently generating functionally engineered EVs with increased osteoinductive properties; moreover, these researchers modified MSC-EVs to have elevated levels of miR-424, which enhances the BMP2 signaling pathway, leading to improved bone regeneration.96 Moreover, polyethylenimine-engineered77 and nuclear factor I/C (NFIC)-encapsulated127 EVs have better potential for promoting angiogenesis and the migration and proliferation of MSCs, respectively. Furthermore, the targeting, regulated release, and sustained retention of EVs at the desired location have positive effects on periodontal regeneration. Our previously published studies showed that immobilizing MSC-EVs on the side chain of gelatin methacryloyl (GelMA) with the aid of a biotin-avidin system allowed the sustained release of MSC-EXs for up to 28 days. These MSC-EV-functionalized hydrogels significantly increased the ability of MSC-EVs to modulate macrophages and promote tissue repair, leading to the optimized therapeutic performance of MSC-EVs in the regrowth of complex periodontal tissues.55 In another study, novel “dual delivery” microparticles were developed to enable the controlled release of EVs in a microenvironment-sensitive manner. This process was achieved by incorporating metalloproteinases to facilitate the secretion of EVs at the impacted position and antibiotics to inhibit bacterial biofilm formation.70 This approach strategically modulates the dynamics of EVs release according to the inflammatory condition of the impacted tissue, which can vary across patients, to promote the regeneration of periodontal tissue. More importantly, the complex and dynamic nature of oral tissue healing, characterized by an abundance of inflammatory factors, underscores the need for more effective strategies (such as physical stimulation) to increase the replication and biological activity of MSC-EVs, alongside their pro-osteogenic and anti-inflammatory functions.109 In this context, low-intensity pulsed ultrasound (LIPUS)109 and cyclic stretch89 stimulation were identified as promising methods for further optimization.

The main strategies for promoting the repair-promoting functions of MSC-EVs paves the way for their clinical application in periodontitis. By preconditioning MSCs, engineering MSC-EVs through cargo loading or membrane modification, and developing local delivery systems for MSC-EVs, researchers can increase the therapeutic efficacy of MSC-EVs, rendering them potentially suitable for the treatment of periodontitis.
Figure 4.

The main strategies for promoting the repair-promoting functions of MSC-EVs paves the way for their clinical application in periodontitis. By preconditioning MSCs, engineering MSC-EVs through cargo loading or membrane modification, and developing local delivery systems for MSC-EVs, researchers can increase the therapeutic efficacy of MSC-EVs, rendering them potentially suitable for the treatment of periodontitis.

In addition to the methods mentioned above, which can increase the quantity and function of EVs, mastering the correct techniques for isolating EVs is crucial. Currently, six main strategies are used to isolate EVs, including ultracentrifugation, immunoaffinity capture, precipitation, ultrafiltration, and size-exclusion chromatography.130 Each technique has unique advantages and disadvantages, which influence its applicability in different scenarios. For example, ultracentrifugation is the most highly used method for separating EVs because of its low price and high efficiency,130 but it runs a high risk of damaging the structure of EVs.131 Other approaches, such as ultrafiltration and precipitation, can avoid disrupting EVs to some degree; however, their limitations (eg, high reagent cost, low yields, and potential contaminants) cannot be ignored.130,131 In conclusion, a standardized method for the extraction of EVs from different cell sources is lacking and needs to be developed through further research; thus, multiple processes are typically used in combination in practical applications, as shown in Table 1.

While the repair-promoting functions of EVs have been extensively investigated, their clinical applications in periodontitis therapy are still in their infancy.17 A human clinical trial was conducted by Beni-Suef University (NCT04270006, Egypt) to evaluate the effect of Exos from ADSCs in the treatment of periodontitis. Ten participants were randomly divided into normal and treatment groups on the basis of their periodontal state. However, the results of this trial have not yet been reported. Another clinical trial, conducted by Xi’an Jiaotong University (ChiCTR1900027140, China), recruited 16 participants from both the normal and treatment groups to investigate the efficacy of autologous DPSC-Exo transplantation in chronic periodontitis. Similarly, the trial results have not been published. However, nonperiodontal clinical trials using MSC-EVs were found in the Cochrane Library database. Most of these trials reported positive therapeutic effects on conditions such as Alzheimer’s disease,132 stroke,133,134 myocardial infarction,135 and skin aging.136 Furthermore, several patents have investigated regarding the production and roles of MSC-EVs in periodontal tissue regeneration, as well as their ability to be modified for clinical applications. Most of these patents focus on bone regeneration, which is essential for achieving successful periodontal regeneration. Some drugs have been developed on the basis of MSC-EVs to restore bone defects, as described in Ceioua patents (CN113559124B, CN110433289A, CN115501252B, and CN116350676B). Another critical issue to explore is how to develop a local delivery system for MSC-EVs,109 such as mineralized collagen gel (CN114642630B), nanoparticles loaded with resveratrol (CN117398361A) or biotin-avidin systems (CN116271223B).

Interestingly, many studies have highlighted not only the therapeutic effects of EVs in periodontal tissue regeneration but also their roles in the early diagnosis and immunotherapy of periodontitis.20 For example, several studies have indicated that EVs and their cargoes can serve as small biomarkers for the early detection of periodontitis.137,138 Furthermore, researchers have explored the use of EVs in the development of periodontal vaccines, leveraging the unique properties and functions of EVs.139 Although these positive effects are promising, substantial clinical evidence is currently lacking, highlighting an area where further efforts are essential in the future.

Overall, MSC-EVs have good potential in regenerative medicine because of their biocompatibility, low immunogenicity, nanoscale size, engineering versatility, and, notably, capacity to transport diverse bioactive cargoes. Nonetheless, there are still critical challenges that need to be resolved before these materials can be used in clinical treatment. First, achieving the commercialization of EVs presents a critical and challenging hurdle, as large-scale production of high-quality EVs along with a standard or harmonized method for their isolation and purification is needed.17 Furthermore, the regeneration of periodontal tissue is a complex process that involves the migration and proliferation of PDLSCs, as well as the reformation of bone, cementum, and vascular structures. However, the fundamental mechanisms governing cementogenesis, angiogenesis, and cellular migration in periodontal regeneration are not yet fully understood,73,122,126,140 impeding the realization of complete periodontal regeneration. More importantly, there are very few clinical trials on the use of EVs in periodontitis, with almost no publishable results. Even the most refined rat models of periodontitis cannot fully mimic the intricate conditions of human periodontitis, thereby necessitating additional evidence from clinical trials before clinical application is feasible. However, many unknown risks remain, such as the inability to control its local aggregation or distant dissemination once it enters the body, which may lead to adverse reactions in healthy tissues.141 Therefore, advancing the clinical translation of EVs is the direction we need to strive for in the future.

Conclusion

As discussed above, EVs derived from bacteria and cells exert both pathogenic and pro-repair effects on periodontal tissues, depending on the cargoes inherited from their parental bacteria or cells. BEVs can enrich virulence factors, amplify the immune response and induce heavy bone resorption. The development of effective ways to capture and clear the generated BEVs may help to reshape periodontal homeostasis. The repair-promoting effects of MSC-EVs in the treatment of periodontitis have been heavily explored and indicate that MSC-EVs may be good alternatives for their parental MSCs. Accumulating evidence has revealed the underlying mechanism for the excellent anti-inflammatory, pro-osteogenic/cementogenic/angiogenic, pro-stem cell migratory and proliferative effects of MSC-EVs, owing to their bioactive cargoes. Moreover, the therapeutic performance of MSC-EVs could be further optimized through various methods, such as preconditioning of MSCs, incorporation of bioactive cargoes or fabrication of local delivery systems. The increased therapeutic efficacy of modified MSC-EVs paves the way for the broad application of MSC-EVs for various biomedical applications, with a particular emphasis on periodontitis treatments. Despite the promising potential for the clinical translation of MSC-EVs in the treatment of periodontitis, several major challenges remain to be addressed. Notably, issues such as efficient storage, cost-effective production, and determination of the most appropriate sources for MSC-EVs need resolution. Furthermore, while the combination of MSC-EVs with biomaterials offers promising prospects, the optimal release profiles of MSC-EVs in periodontal defects require further investigation. Additionally, the application of sophisticated bioengineering technologies and advanced materials may limit clinical translation, necessitating careful consideration of costs, particularly as periodontitis is not a life-threatening disease.

Author contributions

Lin Jing, Hong-Yu Wang, Ning Zhang, and Wen-Jie Zhang contributed to the conception of the study, data curation, software, and writing of the original or revised draft. Yuzhe Chen contributed to the software and revised the manuscript. Dao-Kun Deng contributed to the data curation and critically revised the manuscript. Xuan Li contributed to the conception of the study and revised the manuscript. Xiao-Tao He and Fa-Ming Chen contributed to the conception of the study, supervision, and critical revision of the manuscript. All the authors read and approved the final version of the manuscript.

Funding

This work was supported by the National Natural Science Foundations of China (grant numbers 82371010 and 82301079) and the Young Elite Scientist Sponsorship Program by CAST (2022QNRC001).

Conflict of interest

The authors declared no potential conflicts of interests.

Data availability

The data underlying this article will be shared upon reasonable request to the corresponding author.

References

1.

Trindade
D
,
Carvalho
R
,
Machado
V
, et al.
Prevalence of periodontitis in dentate people between 2011 and 2020: a systematic review and meta‐analysis of epidemiological studies
.
J Clin Periodontol.
2023
;
50
:
604
-
626
. https://doi.org/

2.

Slots
J.
Primer on etiology and treatment of progressive/severe periodontitis: a systemic health perspective
.
Periodontology 2000.
2020
;
83
:
272
-
276
. https://doi.org/

3.

Genco
RJ
,
Sanz
M.
Clinical and public health implications of periodontal and systemic diseases: an overview
.
Periodontology 2000.
2020
;
83
:
7
-
13
. https://doi.org/

4.

Tonetti
MS
,
Jepsen
S
,
Jin
L
,
Otomo‐Corgel
J.
Impact of the global burden of periodontal diseases on health, nutrition and wellbeing of mankind: a call for global action
.
J Clin Periodontol.
2017
;
44
:
456
-
462
. https://doi.org/

5.

Hajishengallis
G
,
Chavakis
T.
Local and systemic mechanisms linking periodontal disease and inflammatory comorbidities
.
Nat Rev Immunol.
2021
;
21
:
426
-
440
. https://doi.org/

6.

Romandini
M
,
Baima
G
,
Antonoglou
G
, et al.
Periodontitis, edentulism, and risk of mortality: a systematic review with meta-analyses
.
J Dent Res.
2021
;
100
:
37
-
49
. https://doi.org/

7.

Petsos
H
,
Ratka‐Krüger
P
,
Neukranz
E
, et al.
Infrabony defects 20 years after open flap debridement and guided tissue regeneration
.
J Clin Periodontol.
2019
;
46
:
552
-
563
. https://doi.org/

8.

Hajishengallis
G.
Periodontitis: from microbial immune subversion to systemic inflammation
.
Nat Rev Immunol.
2015
;
15
:
30
-
44
. https://doi.org/

9.

Hajishengallis
G
,
Chavakis
T
,
Lambris
JD.
Current understanding of periodontal disease pathogenesis and targets for host-modulation therapy
.
Periodontology 2000.
2020
;
84
:
14
-
34
. https://doi.org/

10.

Joseph
S
,
Curtis
MA.
Microbial transitions from health to disease
.
Periodontology 2000.
2021
;
86
:
201
-
209
. https://doi.org/

11.

Nilsson
B-O.
Mechanisms involved in regulation of periodontal ligament cell production of pro-inflammatory cytokines: implications in periodontitis
.
J Periodontal Res.
2021
;
56
:
249
-
255
. https://doi.org/

12.

Pan
W
,
Wang
Q
,
Chen
Q.
The cytokine network involved in the host immune response to periodontitis
.
Int J Oral Sci.
2019
;
11
:
30
. https://doi.org/

13.

Xu
W
,
Zhou
W
,
Wang
H
,
Liang
S.
Roles of Porphyromonas gingivalis and its virulence factors in periodontitis
.
Adv Protein Chem Struct Biol.
2020
;
120
:
45
-
84
. https://doi.org/

14.

Kim
HY
,
Song
M-K
,
Gho
YS
,
Kim
H-H
,
Choi
B-K.
Extracellular vesicles derived from the periodontal pathogen Filifactor alocis induce systemic bone loss through Toll-like receptor 2
.
J Extracell Vesicles.
2021
;
10
:
e12157
. https://doi.org/

15.

Vanaja
SK
,
Russo
AJ
,
Behl
B
, et al.
Bacterial outer membrane vesicles mediate cytosolic localization of LPS and caspase-11 activation
.
Cell.
2016
;
165
:
1106
-
1119
. https://doi.org/

16.

Chen
L
,
Zhu
S
,
Guo
S
,
Tian
W.
Mechanisms and clinical application potential of mesenchymal stem cells-derived extracellular vesicles in periodontal regeneration
.
Stem Cell Res Ther.
2023
;
14
:
26
. https://doi.org/

17.

Deng
D-K
,
Zhang
J-J
,
Gan
D
, et al.
Roles of extracellular vesicles in periodontal homeostasis and their therapeutic potential
.
J Nanobiotechnology.
2022
;
20
:
545
. https://doi.org/

18.

Yu
T
,
Wang
J
,
Zhou
Y
, et al.
Harnessing engineered extracellular vesicles from mesenchymal stem cells as therapeutic scaffolds for bone‐related diseases
.
Adv Funct Mater.
2024
;
34
:
2402861
. https://doi.org/

19.

Miron
RJ
,
Estrin
NE
,
Sculean
A
,
Zhang
Y.
Understanding exosomes: Part 2-Emerging leaders in regenerative medicine
.
Periodontol 2000.
2024
;
94
:
257
-
414
. https://doi.org/

20.

Miron
RJ
,
Estrin
NE
,
Sculean
A
,
Zhang
Y.
Understanding exosomes: Part 3-therapeutic + diagnostic potential in dentistry
.
Periodontol 2000.
2024
;
94
:
415
-
482
. https://doi.org/

21.

Toyofuku
M
,
Schild
S
,
Kaparakis-Liaskos
M
,
Eberl
L.
Composition and functions of bacterial membrane vesicles
.
Nat Rev Microbiol.
2023
;
21
:
415
-
430
. https://doi.org/

22.

Toyofuku
M
,
Nomura
N
,
Eberl
L.
Types and origins of bacterial membrane vesicles
.
Nat Rev Microbiol.
2019
;
17
:
13
-
24
. https://doi.org/

23.

Lim
Y
,
Kim
HY
,
Han
D
,
Choi
B-K.
Proteome and immune responses of extracellular vesicles derived from macrophages infected with the periodontal pathogen Tannerella forsythia
.
J Extracell Vesicles.
2023
;
12
:
e12381
. https://doi.org/

24.

Gui
MJ
,
Dashper
SG
,
Slakeski
N
,
Chen
YY
,
Reynolds
EC.
Spheres of influence: Porphyromonas gingivalis outer membrane vesicles
.
Mol Oral Microbiol.
2016
;
31
:
365
-
378
. https://doi.org/

25.

Ma
X
,
Shin
Y-J
,
Yoo
J-W
,
Park
H-S
,
Kim
D-H.
Extracellular vesicles derived from Porphyromonas gingivalis induce trigeminal nerve-mediated cognitive impairment
.
J Adv Res.
2023
;
54
:
293
-
303
. https://doi.org/

26.

Xie
HB.
Biogenesis
and function of Porphyromonas gingivalis outer membrane vesicles
.
Future Microbiol.
1527
;
10
:
1517
. https://doi.org/

27.

Rompikuntal
PK
,
Thay
B
,
Khan
MK
, et al.
Perinuclear localization of internalized outer membrane vesicles carrying active cytolethal distending toxin from Aggregatibacter actinomycetemcomitans
.
Infect Immun.
2012
;
80
:
31
-
42
. https://doi.org/

28.

Kieselbach
T
,
Zijnge
V
,
Granström
E
,
Oscarsson
J.
Proteomics of Aggregatibacter actinomycetemcomitans outer membrane vesicles
.
PLoS One.
2015
;
10
:
e0138591
. https://doi.org/

29.

Belibasakis
GN
,
Maula
T
,
Bao
K
, et al.
Virulence and pathogenicity properties of Aggregatibacter actinomycetemcomitans
.
Pathogens.
2019
;
8
:
222
. https://doi.org/

30.

Kaparakis-Liaskos
M
,
Ferrero
RL.
Immune modulation by bacterial outer membrane vesicles
.
Nat Rev Immunol.
2015
;
15
:
375
-
387
. https://doi.org/

31.

Chen
KW
,
Monteleone
M
,
Boucher
D
, et al.
Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps
.
Sci Immunol.
2018
;
3
:
eaar6676
. https://doi.org/

32.

Mikolajczyk-Pawlinska
J
,
Travis
J
,
Potempa
J.
Modulation of interleukin‐8 activity by gingipains from Porphyromonas gingivalis: implications for pathogenicity of periodontal disease
.
FEBS Lett.
1998
;
440
:
282
-
286
. https://doi.org/

33.

du Teil Espina
M
,
Fu
Y
,
van der Horst
D
, et al.
Coating and corruption of human neutrophils by bacterial outer membrane vesicles
.
Microbiol Spectr.
2022
;
10
:
e0075322
. https://doi.org/

34.

Cecil
JD
,
O’Brien-Simpson
NM
,
Lenzo
JC
, et al.
Outer membrane vesicles prime and activate macrophage inflammasomes and cytokine secretion in vitro and in vivo
.
Front Immunol.
2017
;
8
:
1017
. https://doi.org/

35.

Chen
G
,
Sun
Q
,
Cai
Q
,
Zhou
H.
Outer membrane vesicles from Fusobacterium nucleatum switch M0-like macrophages toward the M1 phenotype to destroy periodontal tissues in mice
.
Front Microbiol.
2022
;
13
:
815638
. https://doi.org/

36.

Florencio-Silva
R
,
Sasso
GR
,
Sasso-Cerri
E
,
Simões
MJ
,
Cerri
PS.
Biology of bone tissue: structure, function, and factors that influence bone cells
.
Biomed Res Int.
2015
;
2015
:
421746
. https://doi.org/

37.

Boyce
BF.
Advances in the regulation of osteoclasts and osteoclast functions
.
J Dent Res.
2013
;
92
:
860
-
867
. https://doi.org/

38.

Xu
J
,
Yu
L
,
Ye
S
, et al.
Oral microbiota-host interaction: the chief culprit of alveolar bone resorption
.
Front Immunol.
2024
;
15
:
1254516
. https://doi.org/

39.

Song
MK
,
Kim
HY
,
Choi
BK
,
Kim
HH.
Filifactor alocis‐derived extracellular vesicles inhibit osteogenesis through TLR2 signaling
.
Mol Oral Microbiol.
2020
;
35
:
202
-
210
. https://doi.org/

40.

Kajiya
M
,
Giro
G
,
Taubman
MA
, et al.
Role of periodontal pathogenic bacteria in RANKL-mediated bone destruction in periodontal disease
.
J Oral Microbiol.
2010
;
2
:
5532
. https://doi.org/

41.

Kim
HY
,
Song
M-K
,
Lim
Y
, et al.
Effects of extracellular vesicles derived from oral bacteria on osteoclast differentiation and activation
.
Sci Rep.
2022
;
12
:
14239
. https://doi.org/

42.

Elsayed
R
,
Elashiry
M
,
Liu
Y
, et al.
Porphyromonas gingivalis provokes exosome secretion and paracrine immune senescence in bystander dendritic cells
.
Front Cell Infect Microbiol.
2021
;
11
:
669989
. https://doi.org/

43.

El-Awady
AR
,
Elashiry
M
,
Morandini
AC
,
Meghil
MM
,
Cutler
CW.
Dendritic cells a critical link to alveolar bone loss and systemic disease risk in periodontitis: immunotherapeutic implications
.
Periodontology 2000.
2022
;
89
:
41
-
50
. https://doi.org/

44.

Zheng
Y
,
Dong
C
,
Yang
J
, et al.
Exosomal microRNA‐155‐5p from PDLSCs regulated Th17/Treg balance by targeting sirtuin‐1 in chronic periodontitis
.
J Cell Physiol.
2019
;
234
:
20662
-
20674
. https://doi.org/

45.

Zhao
M
,
Dai
W
,
Wang
H
, et al.
Periodontal ligament fibroblasts regulate osteoblasts by exosome secretion induced by inflammatory stimuli
.
Arch Oral Biol.
2019
;
105
:
27
-
34
. https://doi.org/

46.

Bi
J
,
Koivisto
L
,
Owen
G
, et al.
Epithelial microvesicles promote an inflammatory phenotype in fibroblasts
.
J Dent Res.
2016
;
95
:
680
-
688
. https://doi.org/

47.

Hathaway-Schrader
JD
,
Novince
CM.
Maintaining homeostatic control of periodontal bone tissue
.
Periodontology 2000.
2021
;
86
:
157
-
187
. https://doi.org/

48.

Anvari
Y
,
Afrashteh
A
,
Pourkaveh
S
, et al.
Emerging role of mesenchymal stem cell-derived extracellular vesicles in periodontal regeneration
.
J Taibah Univ Med Sci.
2024
;
19
:
390
-
402
. https://doi.org/

49.

Zheng
J
,
Kong
Y
,
Hu
X
, et al.
MicroRNA-enriched small extracellular vesicles possess odonto-immunomodulatory properties for modulating the immune response of macrophages and promoting odontogenesis
.
Stem Cell Res Ther.
2020
;
11
:
517
. https://doi.org/

50.

Shen
Z
,
Kuang
S
,
Zhang
Y
, et al.
Chitosan hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates periodontitis in mice via a macrophage-dependent mechanism
.
Bioact Mater.
2020
;
5
:
1113
-
1126
. https://doi.org/

51.

Han
XD
,
Chen
HM
,
Li
C.
Effect of human periodontal ligament stem cell-derived extracellular vesicles on macrophage pyroptosis and periodontal inflammatory injury in periodontitis
.
Cells Tissues Organs.
2022
;
211
:
57
-
72
. https://doi.org/

52.

Huang
Y
,
Liu
Q
,
Liu
L
, et al.
Lipopolysaccharide-preconditioned dental follicle stem cells derived small extracellular vesicles treating periodontitis via reactive oxygen species/mitogen-activated protein kinase signaling-mediated antioxidant effect
.
Int J Nanomedicine.
2022
;
17
:
799
-
819
. https://doi.org/

53.

Nakao
Y
,
Fukuda
T
,
Zhang
Q
, et al.
Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss
.
Acta Biomater.
2021
;
122
:
306
-
324
. https://doi.org/

54.

Kang
M
,
Huang
C-C
,
Gajendrareddy
P
, et al.
Extracellular vesicles from TNFα preconditioned MSCs: effects on immunomodulation and bone regeneration
.
Front Immunol.
2022
;
13
:
878194
. https://doi.org/

55.

Deng
D
,
Li
X
,
Zhang
J-J
, et al.
Biotin–avidin system-based delivery enhances the therapeutic performance of MSC-derived exosomes
.
ACS Nano.
2023
;
17
:
8530
-
8550
. https://doi.org/

56.

Williams
DW
,
Greenwell-Wild
T
,
Brenchley
L
, et al. ;
NIDCD/NIDCR Genomics and Computational Biology Core
.
Human oral mucosa cell atlas reveals a stromal-neutrophil axis regulating tissue immunity
.
Cell.
2021
;
184
:
4090
-
4104.e15
. https://doi.org/

57.

Li
P
,
Ou
Q
,
Shi
S
,
Shao
C.
Immunomodulatory properties of mesenchymal stem cells/dental stem cells and their therapeutic applications
.
Cell Mol Immunol.
2023
;
20
:
558
-
569
. https://doi.org/

58.

Li
M
,
Soder
R
,
Abhyankar
S
, et al.
WJMSC-derived small extracellular vesicle enhance T cell suppression through PD-L1
.
J Extracell Vesicles.
2021
;
10
:
e12067
. https://doi.org/

59.

Del Fattore
A
,
Luciano
R
,
Pascucci
L
, et al.
Immunoregulatory effects of mesenchymal stem cell-derived extracellular vesicles on T lymphocytes
.
Cell Transplant.
2015
;
24
:
2615
-
2627
. https://doi.org/

60.

Li
N
,
Gao
Z
,
Zhao
L
, et al.
MSC-derived small extracellular vesicles attenuate autoimmune dacryoadenitis by promoting M2 macrophage polarization and inducing tregs via miR-100-5p
.
Front Immunol.
2022
;
13
:
888949
. https://doi.org/

61.

Di Trapani
M
,
Bassi
G
,
Midolo
M
, et al.
Differential and transferable modulatory effects of mesenchymal stromal cell-derived extracellular vesicles on T, B and NK cell functions
.
Sci Rep.
2016
;
6
:
24120
. https://doi.org/

62.

Reis
M
,
Mavin
E
,
Nicholson
L
, et al.
Mesenchymal stromal cell-derived extracellular vesicles attenuate dendritic cell maturation and function
.
Front Immunol.
2018
;
9
:
2538
. https://doi.org/

63.

Favaro
E
,
Carpanetto
A
,
Caorsi
C
, et al.
Human mesenchymal stem cells and derived extracellular vesicles induce regulatory dendritic cells in type 1 diabetic patients
.
Diabetologia.
2016
;
59
:
325
-
333
. https://doi.org/

64.

Li
X
,
Jiang
Y
,
Liu
X
, et al.
Mesenchymal stem cell-derived apoptotic bodies alleviate alveolar bone destruction by regulating osteoclast differentiation and function
.
Int J Oral Sci.
2023
;
15
:
51
. https://doi.org/

65.

Huang
X
,
Wang
H
,
Wang
C
,
Cao
Z.
The applications and potentials of extracellular vesicles from different cell sources in periodontal regeneration
.
Int J Mol Sci.
2023
;
24
:
5790
. https://doi.org/

66.

Mousavi
E
,
Khosravi
A
,
Sedigh
SS
, et al.
Exosomes derived from mesenchymal stem cells: heralding a new treatment for periodontitis
?
Tissue Cell.
2023
;
82
:
102070
. https://doi.org/

67.

Huang
C-C
,
Kang
M
,
Narayanan
R
, et al.
Evaluating the endocytosis and lineage-specification properties of mesenchymal stem cell derived extracellular vesicles for targeted therapeutic applications
.
Front Pharmacol.
2020
;
11
:
163
. https://doi.org/

68.

Zhang
Y
,
Chen
J
,
Fu
H
, et al.
Exosomes derived from 3D-cultured MSCs improve therapeutic effects in periodontitis and experimental colitis and restore the Th17 cell/Treg balance in inflamed periodontium
.
Int J Oral Sci.
2021
;
13
:
43
. https://doi.org/

69.

Yi
G
,
Zhang
S
,
Ma
Y
, et al.
Matrix vesicles from dental follicle cells improve alveolar bone regeneration via activation of the PLC/PKC/MAPK pathway
.
Stem Cell Res Ther.
2022
;
13
:
41
. https://doi.org/

70.

Zarubova
J
,
Hasani-Sadrabadi
MM
,
Dashtimoghadam
E
, et al.
Engineered delivery of dental stem-cell-derived extracellular vesicles for periodontal tissue regeneration
.
Adv Healthc Mater.
2022
;
11
:
e2102593
. https://doi.org/

71.

Han
P
,
Johnson
N
,
Abdal‐hay
A
, et al.
Effects of periodontal cells‐derived extracellular vesicles on mesenchymal stromal cell function
.
J Periodontal Res.
2023
;
58
:
1188
-
1200
. https://doi.org/

72.

Zheng
Y
,
Lu
H
,
Mu
Q
, et al.
Effects of sEV derived from SHED and DPSC on the proliferation, migration and osteogenesis of PDLSC
.
Regen Ther.
2023
;
24
:
489
-
498
. https://doi.org/

73.

Takeuchi
R
,
Katagiri
W
,
Endo
S
,
Kobayashi
T.
Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis
.
PLoS One.
2019
;
14
:
e0225472
. https://doi.org/

74.

Silva
FRF
,
Heredia
JE
,
Duffles
LF
, et al.
Protective effect of bovine milk extracellular vesicles on alveolar bone loss
.
Mol Nutr Food Res.
2023
;
68
:
e2300445
. https://doi.org/

75.

Shimizu
Y
,
Takeda‐Kawaguchi
T
,
Kuroda
I
, et al.
Exosomes from dental pulp cells attenuate bone loss in mouse experimental periodontitis
.
J Periodontal Res.
2021
;
57
:
162
-
172
. https://doi.org/

76.

Chen
X
,
Wan
Z
,
Yang
L
, et al.
Exosomes derived from reparative M2-like macrophages prevent bone loss in murine periodontitis models via IL-10 mRNA
.
J Nanobiotechnology.
2022
;
20
:
110
. https://doi.org/

77.

Pizzicannella
J
,
Gugliandolo
A
,
Orsini
T
, et al.
Addendum: engineered extracellular vesicles from human periodontal-ligament stem cells increase VEGF/VEGFR2 expression during bone regeneration
.
Front Physiol.
2023
;
14
:
1148929
. https://doi.org/

78.

Wei
J
,
Song
Y
,
Du
Z
, et al.
Exosomes derived from human exfoliated deciduous teeth ameliorate adult bone loss in mice through promoting osteogenesis
.
J Mol Histol.
2020
;
51
:
455
-
466
. https://doi.org/

79.

Zhao
B
,
Zhang
W
,
Xiong
Y
, et al.
Effects of rutin on the oxidative stress, proliferation and osteogenic differentiation of periodontal ligament stem cells in LPS-induced inflammatory environment and the underlying mechanism
.
J Mol Histol.
2020
;
51
:
161
-
171
. https://doi.org/

80.

Zhai
M
,
Zhu
Y
,
Yang
M
,
Mao
C.
Human mesenchymal stem cell derived exosomes enhance cell-free bone regeneration by altering their miRNAs profiles
.
Adv Sci.
2020
;
7
:
2001334
. https://doi.org/

81.

Deng
J
,
Wang
X
,
Zhang
W
, et al.
Versatile hypoxic extracellular vesicles laden in an injectable and bioactive hydrogel for accelerated bone regeneration
.
Adv Funct Mater.
2023
;
33
:
2211664
. https://doi.org/

82.

Yang
S
,
Zhu
B
,
Tian
XY
, et al.
Exosomes derived from human umbilical cord mesenchymal stem cells enhance the osteoblastic differentiation of periodontal ligament stem cells under high glucose conditions through the PI3K/AKT signaling pathway
.
Biomed Environ Sci.
2022
;
35
:
811
-
820
. https://doi.org/

83.

Fu
H
,
Sen
L
,
Zhang
F
, et al.
Mesenchymal stem cells-derived extracellular vesicles protect against oxidative stress-induced xenogeneic biological root injury via adaptive regulation of the PI3K/Akt/NRF2 pathway
.
J Nanobiotechnology.
2023
;
21
:
466
. https://doi.org/

84.

Liu
L
,
Guo
S
,
Shi
W
, et al.
Bone marrow mesenchymal stem cell-derived small extracellular vesicles promote periodontal regeneration
.
Tissue Eng Part A.
2021
;
27
:
962
-
976
. https://doi.org/

85.

Xiao
F
,
Zuo
B
,
Tao
B
, et al.
Exosomes derived from cyclic mechanical stretch-exposed bone marrow mesenchymal stem cells inhibit RANKL-induced osteoclastogenesis through the NF-κB signaling pathway
.
Ann Transl Med.
2021
;
9
:
798
. https://doi.org/

86.

Huang
X
,
Li
Y
,
Liao
H
, et al.
Research advances on stem cell-derived extracellular vesicles promoting the reconstruction of alveolar bone through RANKL/RANK/OPG pathway
.
J Funct Biomater.
2023
;
14
:
193
. https://doi.org/

87.

Lu
J
,
Yu
N
,
Liu
Q
,
Xie
Y
,
Zhen
L.
Periodontal ligament stem cell exosomes key to regulate periodontal regeneration by miR-31-5p in mice model
.
Int J Nanomedicine.
2023
;
18
:
5327
-
5342
. https://doi.org/

88.

Wang
Q
,
Shen
X
,
Chen
Y
,
Chen
J
,
Li
Y.
Osteoblasts-derived exosomes regulate osteoclast differentiation through miR-503-3p/Hpse axis
.
Acta Histochem.
2021
;
123
:
151790
. https://doi.org/

89.

Qu
F
,
Zhang
Y-F
,
Wang
Y-Y
, et al.
Cyclic stretch-induced exosomes from periodontal ligament cells promote osteoblasts osteogenic differentiation via the miR-181d-5p/TNF signaling pathway
.
Arch Oral Biol.
2024
;
157
:
105843
. https://doi.org/

90.

Li
J
,
Sakisaka
Y
,
Nemoto
E
, et al.
Cementocyte-derived extracellular vesicles regulate osteoclastogenesis and osteoblastogenesis
.
J Dent Sci.
2024
;
19
:
2236
-
2246
. https://doi.org/

91.

Lai
S
,
Deng
L
,
Liu
C
, et al.
Bone marrow mesenchymal stem cell-derived exosomes loaded with miR-26a through the novel immunomodulatory peptide DP7-C can promote osteogenesis
.
Biotechnol Lett.
2023
;
45
:
905
-
919
. https://doi.org/

92.

Lan
Q
,
Cao
J
,
Bi
X
, et al.
Curcumin-primed periodontal ligament stem cells-derived extracellular vesicles improve osteogenic ability through the Wnt/β-catenin pathway
.
Front Cell Dev Biol.
2023
;
11
:
1225449
. https://doi.org/

93.

Lei
F
,
Li
M
,
Lin
T
, et al.
Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes
.
Acta Biomater.
2022
;
141
:
333
-
343
. https://doi.org/

94.

Yan
C
,
Li
N
,
Xiao
T
, et al.
Extracellular vesicles from the inflammatory microenvironment regulate the osteogenic and odontogenic differentiation of periodontal ligament stem cells by miR-758-5p/LMBR1/BMP2/4 axis
.
J Transl Med.
2022
;
20
:
208
. https://doi.org/

95.

Huang
C-C
,
Kang
M
,
Lu
Y
, et al.
Functionally engineered extracellular vesicles improve bone regeneration
.
Acta Biomater.
2020
;
109
:
182
-
194
. https://doi.org/

96.

Huang
C-C
,
Kang
M
,
Leung
K
, et al.
Micro RNA based MSC EV engineering: targeting the BMP2 cascade for bone repair
.
Front Cell Dev Biol.
2023
;
11
:
1127594
. https://doi.org/

97.

Liu
A
,
Lin
D
,
Zhao
H
, et al.
Optimized BMSC-derived osteoinductive exosomes immobilized in hierarchical scaffold via lyophilization for bone repair through Bmpr2/Acvr2b competitive receptor-activated Smad pathway
.
Biomaterials.
2021
;
272
:
120718
. https://doi.org/

98.

Wang
M
,
Li
J
,
Ye
Y
,
He
S
,
Song
J.
SHED-derived conditioned exosomes enhance the osteogenic differentiation of PDLSCs via Wnt and BMP signaling in vitro
.
Differentiation.
2020
;
111
:
1
-
11
. https://doi.org/

99.

Ma
L
,
Rao
N
,
Jiang
H
, et al.
Small extracellular vesicles from dental follicle stem cells provide biochemical cues for periodontal tissue regeneration
.
Stem Cell Res Ther.
2022
;
13
:
92
. https://doi.org/

100.

Wu
J
,
Chen
L
,
Wang
R
, et al.
Exosomes secreted by stem cells from human exfoliated deciduous teeth promote alveolar bone defect repair through the regulation of angiogenesis and osteogenesis
.
ACS Biomater Sci Eng.
2019
;
5
:
3561
-
3571
. https://doi.org/

101.

Qiu
X
,
Liu
J
,
Zheng
C
, et al.
Exosomes released from educated mesenchymal stem cells accelerate cutaneous wound healing via promoting angiogenesis
.
Cell Prolif.
2020
;
53
:
e12830
. https://doi.org/

102.

Chew
JRJ
,
Chuah
SJ
,
Teo
KYW
, et al.
Mesenchymal stem cell exosomes enhance periodontal ligament cell functions and promote periodontal regeneration
.
Acta Biomater.
2019
;
89
:
252
-
264
. https://doi.org/

103.

Lv
P-Y
,
Gao
P-F
,
Tian
G-J
, et al.
Osteocyte-derived exosomes induced by mechanical strain promote human periodontal ligament stem cell proliferation and osteogenic differentiation via the miR-181b-5p/PTEN/AKT signaling pathway
.
Stem Cell Res Ther.
2020
;
11
:
295
. https://doi.org/

104.

Liu
Y
,
Zhang
Z
,
Ma
C
, et al.
Transplanted MSCs promote alveolar bone repair via hypoxia‐induced extracellular vesicle secretion
.
Oral Dis.
2024
;
30
(
8
):
5221
-
5231
.  https://doi.org/

105.

Dai
Z
,
Li
Z
,
Zheng
W
, et al.
Gallic acid ameliorates the inflammatory state of periodontal ligament stem cells and promotes pro-osteodifferentiation capabilities of inflammatory stem cell-derived exosomes
.
Life.
2022
;
12
:
1392
. https://doi.org/

106.

Zhang
S
,
Wang
S
,
Chen
J
, et al.
Human dental pulp stem cell-derived exosomes decorated titanium scaffolds for promoting bone regeneration
.
Colloids Surf B Biointerfaces.
2024
;
235
:
113775
. https://doi.org/

107.

Wang
L
,
Wei
X
,
He
X
, et al.
Osteoinductive dental pulp stem cell-derived extracellular vesicle-loaded multifunctional hydrogel for bone regeneration
.
ACS Nano.
2024
;
18
:
8777
-
8797
. https://doi.org/

108.

Xu
J
,
Lin
Y
,
Tian
M
, et al.
Periodontal ligament stem cell-derived extracellular vesicles enhance tension-induced osteogenesis
.
ACS Biomater Sci Eng.
2022
;
9
:
388
-
398
. https://doi.org/

109.

Zhang
T
,
Chen
Z
,
Zhu
M
, et al.
Extracellular vesicles derived from human dental mesenchymal stem cells stimulated with low-intensity pulsed ultrasound alleviate inflammation-induced bone loss in a mouse model of periodontitis
.
Genes Dis.
2023
;
10
:
1613
-
1625
. https://doi.org/

110.

Liu
Y
,
Zeng
L
,
Wang
W
, et al.
Human bone marrow mesenchymal stem cell exosome-derived miR-335-5p promotes osteogenic differentiation of human periodontal ligament stem cells to alleviate periodontitis by downregulating DKK1
.
Nan Fang Yi Ke Da Xue Xue Bao.
2023
;
43
:
420
-
427
. https://doi.org/

111.

Yu
J
,
Wu
X
,
Zhang
W
, et al.
Effect of psoralen on the regulation of osteogenic differentiation induced by periodontal stem cell-derived exosomes
.
Hum Cell.
2023
;
36
:
1389
-
1402
. https://doi.org/

112.

Cui
Y
,
Fu
S
,
Sun
D
, et al.
EPC-derived exosomes promote osteoclastogenesis through LncRNA-MALAT1
.
J Cell Mol Med.
2019
;
23
:
3843
-
3854
. https://doi.org/

113.

Lin
C
,
Yang
Y
,
Wang
Y
, et al.
Periodontal ligament fibroblasts-derived exosomes induced by PGE2 inhibit human periodontal ligament stem cells osteogenic differentiation via activating miR-34c-5p/SATB2/ERK
.
Exp Cell Res.
2022
;
419
:
113318
. https://doi.org/

114.

Zhang
X
,
Zhao
Z
,
Chen
Y
,
Han
X
,
Jie
Y.
Extracellular vesicles secreted by human periodontal ligament induced osteoclast differentiation by transporting miR-28 to osteoclast precursor cells and further promoted orthodontic tooth movement
.
Int Immunopharmacol.
2022
;
113
:
109388
. https://doi.org/

115.

Arzate
H
,
Zeichner‐David
M
,
Mercado‐Celis
G.
Cementum proteins: role in cementogenesis, biomineralization, periodontium formation and regeneration
.
Periodontol 2000.
2015
;
67
:
211
-
233
. https://doi.org/

116.

Zhao
J
,
Faure
L
,
Adameyko
I
,
Sharpe
PT.
Stem cell contributions to cementoblast differentiation in healthy periodontal ligament and periodontitis
.
Stem Cells.
2021
;
39
:
92
-
102
. https://doi.org/

117.

Li
S
,
Guan
X
,
Yu
W
, et al.
Effect of human periodontal ligament stem cell‐derived exosomes on cementoblast activity
.
Oral Dis.
2023
;
30
:
2511
-
2522
. https://doi.org/

118.

Huang
X
,
Deng
Y
,
Xiao
J
, et al.
Genetically engineered M2-like macrophage-derived exosomes for P. gingivalis-suppressed cementum regeneration: from mechanism to therapy
.
Bioact Mater.
2024
;
32
:
473
-
487
. https://doi.org/

119.

Sun
X
,
Mao
Y
,
Liu
B
, et al.
Mesenchymal stem cell-derived exosomes enhance 3D-printed scaffold functions and promote alveolar bone defect repair by enhancing angiogenesis
.
J Pers Med.
2023
;
13
:
180
. https://doi.org/

120.

Gardin
C
,
Ferroni
L
,
Erdoğan
YK
, et al.
Nanostructured modifications of titanium surfaces improve vascular regenerative properties of exosomes derived from mesenchymal stem cells: preliminary in vitro results
.
Nanomaterials (Basel, Switzerland).
2021
;
11
:
3452
. https://doi.org/

121.

Ge
L
,
Xun
C
,
Li
W
, et al.
Extracellular vesicles derived from hypoxia-preconditioned olfactory mucosa mesenchymal stem cells enhance angiogenesis via miR-612
.
J Nanobiotechnology.
2021
;
19
:
380
. https://doi.org/

122.

Zhang
Z
,
Shuai
Y
,
Zhou
F
, et al.
PDLSCs regulate angiogenesis of periodontal ligaments via VEGF transferred by exosomes in periodontitis
.
Int J Med Sci.
2020
;
17
:
558
-
567
. https://doi.org/

123.

Zhou
H
,
Li
X
,
Wu
R-X
, et al.
Periodontitis-compromised dental pulp stem cells secrete extracellular vesicles carrying miRNA-378a promote local angiogenesis by targeting Sufu to activate the Hedgehog/Gli1 signalling
.
Cell Prolif.
2021
;
54
:
e13026
. https://doi.org/

124.

Liu
L
,
Liu
Y
,
Feng
C
, et al.
Lithium-containing biomaterials stimulate bone marrow stromal cell-derived exosomal miR-130a secretion to promote angiogenesis
.
Biomaterials.
2019
;
192
:
523
-
536
. https://doi.org/

125.

Li
Z
,
Wu
M
,
Liu
S
, et al.
Apoptotic vesicles activate autophagy in recipient cells to induce angiogenesis and dental pulp regeneration
.
Mol Ther.
2022
;
30
:
3193
-
3208
. https://doi.org/

126.

Zhou
H
,
Li
X
,
Yin
Y
, et al.
The proangiogenic effects of extracellular vesicles secreted by dental pulp stem cells derived from periodontally compromised teeth
.
Stem Cell Res Ther.
2020
;
11
:
110
. https://doi.org/

127.

Yang
S
,
Liu
Q
,
Chen
S
, et al.
Extracellular vesicles delivering nuclear factor I/C for hard tissue engineering: treatment of apical periodontitis and dentin regeneration
.
J Tissue Eng.
2022
;
13
:
20417314221084095
. https://doi.org/

128.

Zhu
D
,
Li
Z
,
Huang
K
, et al.
Minimally invasive delivery of therapeutic agents by hydrogel injection into the pericardial cavity for cardiac repair
.
Nat Commun.
2021
;
12
:
1412
. https://doi.org/

129.

Luo
L
,
Avery
SJ
,
Waddington
RJ.
Exploring a chemotactic role for EVS from progenitor cell populations of human exfoliated deciduous teeth for promoting migration of naïve BMSCs in bone repair process
.
Stem Cells Int.
2021
;
2021
:
6681771
. https://doi.org/

130.

Miron
RJ
,
Zhang
Y.
Understanding exosomes: part 1-characterization, quantification and isolation techniques
.
Periodontol 2000.
2024
;
94
:
231
-
256
. https://doi.org/

131.

Yu
LL
,
Zhu
J
,
Liu
JX
, et al.
A comparison of traditional and novel methods for the separation of exosomes from human samples
.
Biomed Res Int.
2018
;
2018
:
3634563
. https://doi.org/

132.

Xie
X
,
Song
Q
,
Dai
C
, et al.
Clinical safety and efficacy of allogenic human adipose mesenchymal stromal cells-derived exosomes in patients with mild to moderate Alzheimer’s disease: a phase I/II clinical trial
.
General Psychiatry.
2023
;
36
:
e101143
. https://doi.org/

133.

Bang
OY
,
Kim
EH
,
Cho
YH
, et al.
Circulating extracellular vesicles in stroke patients treated with mesenchymal stem cells: a biomarker analysis of a randomized trial
.
Stroke.
2022
;
53
:
2276
-
2286
. https://doi.org/

134.

Dehghani
L
,
Khojasteh
A
,
Soleimani
M
, et al.
Safety of intraparenchymal injection of allogenic placenta mesenchymal stem cells derived exosome in patients undergoing decompressive craniectomy following malignant middle cerebral artery infarct, a pilot randomized clinical trial
.
Int J Prev Med.
2022
;
13
:
7
. https://doi.org/

135.

Marban
E.
Exosomes and their RNA contents as mediators of therapeutic regeneration
.
FASEB J.
2016
;
30
:
373
-
372
. https://doi.org/

136.

Park
GH
,
Kwon
HH
,
Seok
J
, et al.
Efficacy of combined treatment with human adipose tissue stem cell‐derived exosome‐containing solution and microneedling for facial skin aging: a 12‐week prospective, randomized, split‐face study
.
J Cosmet Dermatol.
2023
;
22
:
3418
-
3426
. https://doi.org/

137.

Han
P
,
Bartold
PM
,
Salomon
C
,
Ivanovski
S.
Salivary small extracellular vesicles associated miRNAs in periodontal status-a pilot study
.
Int J Mol Sci.
2020
;
21
:
2809
. https://doi.org/

138.

Nik Mohamed Kamal
NNS
,
Awang
RAR
,
Mohamad
S
,
Shahidan
WNS.
Plasma- and saliva exosome profile reveals a distinct microRNA signature in chronic periodontitis
.
Front Physiol.
2020
;
11
:
587381
. https://doi.org/

139.

Kourembanas
S.
Exosomes: vehicles of intercellular signaling, biomarkers, and vectors of cell therapy
.
Annu Rev Physiol.
2015
;
77
:
13
-
27
. https://doi.org/

140.

Ming
L
,
Qu
Y
,
Wang
Z
, et al.
Small extracellular vesicles laden oxygen-releasing thermosensitive hydrogel for enhanced antibacterial therapy against anaerobe-induced periodontitis alveolar bone defect
.
ACS Biomater Sci Eng.
2024
;
10
:
932
-
945
. https://doi.org/

141.

Zhang
XX
,
Wang
X
,
Ren
XY.
Research progress on the role of extracellular vesicles in the diagnosis and treatment of chronic periodontitis
.
Zhonghua Kou Qiang Yi Xue Za Zhi.
2021
;
56
:
385
-
389
. https://doi.org/. https://rs.yiigle.com/cmaid/1314884

Author notes

Lin Jing, Hong-Yu Wang, Ning Zhang and Wen-Jie Zhang contributed equally.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.