Next Article in Journal
Intracellular Membrane Transport in Vascular Endothelial Cells
Next Article in Special Issue
Uncovering the Underworld of Axial Spondyloarthritis
Previous Article in Journal
Regulation of Aqueous Humor Secretion by Melatonin in Porcine Ciliary Epithelium
Previous Article in Special Issue
Update on the Role and Regulatory Mechanism of Extracellular Matrix in the Pathogenesis of Uterine Fibroids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Applications and Potentials of Extracellular Vesicles from Different Cell Sources in Periodontal Regeneration

1
The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
2
Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(6), 5790; https://doi.org/10.3390/ijms24065790
Submission received: 18 February 2023 / Revised: 8 March 2023 / Accepted: 15 March 2023 / Published: 17 March 2023

Abstract

:
Periodontitis is a chronic infectious disease worldwide that can cause damage to periodontal supporting tissues including gingiva, bone, cementum and periodontal ligament (PDL). The principle for the treatment of periodontitis is to control the inflammatory process. Achieving structural and functional regeneration of periodontal tissues is also essential and remains a major challenge. Though many technologies, products, and ingredients were applied in periodontal regeneration, most of the strategies have limited outcomes. Extracellular vesicles (EVs) are membranous particles with a lipid structure secreted by cells, containing a large number of biomolecules for the communication between cells. Numerous studies have demonstrated the beneficial effects of stem cell-derived EVs (SCEVs) and immune cell-derived EVs (ICEVs) on periodontal regeneration, which may be an alternative strategy for cell-based periodontal regeneration. The production of EVs is highly conserved among humans, bacteria and plants. In addition to eukaryocyte-derived EVs (CEVs), a growing body of literature suggests that bacterial/plant-derived EVs (BEVs/PEVs) also play an important role in periodontal homeostasis and regeneration. The purpose of this review is to introduce and summarize the potential therapeutic values of BEVs, CEVs and PEVs in periodontal regeneration, and discuss the current challenges and prospects for EV-based periodontal regeneration.

1. Introduction

Periodontitis is a chronic inflammatory disease characterized by irreversible destruction of periodontal supporting tissues, which is related to the disturbance of the local microbial community, and then immune cell infiltration, osteoclast activation and inflammatory cytokine release induced by the overactivation of host immunity. It is also associated with obesity, cardiovascular disease, Alzheimer’s disease and many other systemic diseases [1,2]. Further, periodontal disease was reported as a risk factor for infection after mandibular fracture and associated with low cognitive performance amongst older adults [3,4]. The traditional treatment for periodontitis focuses on removing calculus, plaque biofilm and diseased cementum through scaling and root planning to effectively control the inflammation. However, it is still faced with great difficulties in the reconstruction of damaged periodontal tissues [5].
Periodontal regeneration includes the regeneration of alveolar bone, cementum, PDL and gingiva. Among them, bone regeneration is the most studied and the most promising. In contrast, orderly and dense PDL regeneration and cementum regeneration on root surface are more challenging [6]. Cementum is a thin layer of mineralized tissue similar to bone tissue located on the surface of tooth root, which is very important for maintaining the stability and health of the tooth. Hence, cementum regeneration was considered to be the golden standard for evaluating a successful whole periodontal regeneration [7,8]. PDL connects alveolar bone and cementum by thick collagen bundles. PDL is a highly vascularized tissue between the bone and cementum, containing a variety of cells such as fibroblasts, endothelial cells, epithelial cells, nerve cells, and undifferentiated mesenchymal cells. Periodontal ligament cells (PDLCs) are able to differentiate into osteoblasts and cementoblasts under appropriate conditions, and are essential for maintaining the homeostasis of periodontal tissues [9].
Research in periodontal regeneration included guided tissue regeneration, platelet-rich plasma and fibrin, growth and differentiation factors, enamel matrix protein, and autologous graft, allograft and xenografts [10,11,12,13]. Nevertheless, periodontal tissue is a system containing a variety of cells and complex structures. Most of the above approaches can only exert a limited and unstable effect in specific circumstances, and need further clinical verification [14,15]. To date, cell therapy based on stem cells and immune cells has been widely concerned in periodontal tissue regeneration. A range of stem cells such as bone marrow mesenchymal stem cells (BMMSCs), periodontal ligament stem cells (PDLSCs), DPSCs (dental pulp stem cells), and a series of immune cells such as neutrophils and macrophages have been proved to play an important role in periodontal regeneration [16,17,18,19,20]. However, they both have some limitations, such as dedifferentiation during stem cell expansion, reduced regeneration efficiency after transplantation, unstable phenotypes of immune cells after transplantation, effects of the environment in different patients on immune cell phenotypes, and inconsistent standards for the production, transportation, storage, and safety of transplanted cells [16,21,22].
EVs are bilayer membranous particles secreted by cells into the extracellular space, which were originally thought to be a pathway of cellular waste excretion [23]. In addition to being the carrier of waste, it also contains a large number of bioactive materials (proteins, nucleic acids, lipids, metabolites) for cell communications [24]. The biological functions of EVs are largely determined by their source cells. More and more studies have shown that EVs derived from stem/immune cells (SCEVs/ICEVs) are the main paracrine participants account for the periodontal regeneration [19,25,26]. Compared with cell therapy, EVs has many advantages including lower immune clearance rate, long storage time, non-toxicity (avoid DMSO and other toxic substances mixed), delivery of active substances more efficiently, strong penetrability through narrow spaces such as blood–brain barrier [21,27]. So, EVs is emerging as an alternative to cell therapy for tissue regeneration [21,28].
In recent years, EV-based periodontal regeneration has become a research front. The generation of EVs is highly conserved between humans, bacteria, and plants. In addition to CEVs, a large number of studies have revealed that BEVs and PEVs are also associated with periodontal disease, homeostasis maintenance and periodontal regeneration [29,30]. There are similarities and differences between them, and some research on them is growing. Therefore, the aim of this review is to sort out and discuss the potential therapeutic values of the above three vesicles in periodontal regeneration. Further, the current challenges and prospects for EV-based periodontal regeneration were also summarized.

2. CEVs and Periodontal Regeneration

2.1. Classification and Biogenesis of CEVs

Although these vesicles have a similar structure and physical properties, they are heterogeneous in size, contents, biological function, and cell type of origin. Therefore, their naming is still controversial [27,31]. According to the classification recommendations from the International Society for Extracellular Vesicles (ISEV), CEVs can be classified into small EVs (<200 nm) and medium/large EVs (>200 nm) on the basis of their size [32]. EVs can be derived from different cell culture supernatants or body fluids. Based on the current published literature, CEVs were divided into exosomes (also known as small EVs, sEVs; <200 nm), microvesicles (50–1000 nm) and apoptotic bodies (50–5000 nm) [31].
The biogenesis of CEVs varies, mainly including the following aspects. (1) The process of exosomes formation consists of early sorting endosomes (ESEs) formed by the first plasma membrane invagination; late sorting endosomes (LSEs) formed by ESEs maturation; LSEs form multivesicular bodies (MVBs) containing multiple intraluminal vesicles (ILVs) by the second membrane invagination; MVBs can fuse with autophagosomes and lysosomes for degradation, or fuse with the plasma membrane to release ILVs (or exosomes) into the extracellular environment. MVBs can be formed through endosomal sorting complex required for transport (ESCRT)-dependent and ESCRT-independent pathways, which determines that exosomes have a variety of protein markers such as CD9, CD63, TSG101, Alix, CD81, HSP90 [31,33]. (2) Microvesicles were formed through the direct outward budding of the plasma membrane. This process requires cytoskeleton contraction, phospholipid redistribution, and Ca2+-dependent enzymatic mechanism. The typical inclusions include CD40, selectin, integrin, cytoskeleton protein, and cholesterol. ARF6 and VCAMP3 were also identified as protein markers [34,35,36]. (3) Different from the above two, apoptotic bodies were produced by programmed dying cells which undergo nuclear chromatin contraction, plasma membrane budding, and cell segmentation. Similar to microvesicles, biogenesis of apoptotic bodies also depends on actin-myosin interaction. The phosphatidylserine on their surface binding with Annexin V can be recognized, engulfed, and degraded by macrophages soon after release. The larger apoptotic bodies have more abundant cell-derived components, and phosphatidylserine, genomic DNA, Annexin V, platelet reactive protein and C3b were considered as the biomarkers of apoptotic bodies [37,38,39,40].
CEVs from multiple cells, especially SCEVs and ICEVs, play a positive role in various tissue regeneration including periodontal regeneration [21,28]. In this part, we summarized the effects of SCEVs and ICEVs on different periodontal cell functions and periodontal regeneration, as well as their potential mechanisms.

2.2. CEVs Regulate Periodontal Immunity and Inflammation

Inflammation is the response of the immune system to harmful stimuli, such as pathogens, damaged cells, toxic compounds or radiation. Periodontitis can be caused by a variety of periodontal pathogens. Controlling inflammation is a prerequisite for periodontal regeneration [1,41]. CEVs are closely related to inflammation. In terms of pathogenesis, CEVs can act as a shuttle or biomarker for inflammation, and were involved in the progression of various tissue inflammation [42]. In terms of therapy, a large number of studies have shown that SCEVs and ICEVs (sEVs were mainly discussed below) have powerful immunomodulatory and anti-inflammatory effects on various inflammatory diseases such as periodontitis [43,44,45].
There are many studies on SCEVs regulating immune cell functions and alleviating periodontal inflammation. Related research reported that stem cells from human exfoliated deciduous teeth (SHED)-derived CEVs (SHED-EVs) reduced IL-6 and TNF-α expression in lipopolysaccharide (LPS)-induced inflammatory BMMSCs, and in tissues from the defect area of periodontitis mice [46]. SCEVs could also regulate the cytokine expression in macrophages. For example, PDLC/PDLSC-derived CEVs (PDLC/PDLSC-EVs) could inhibited cell pyroptosis and the production of inflammatory cytokines (IL-1β, IL-18 and TNF-α) in macrophages stimulated with LPS through NF-κB signaling pathway or delivering miR-590-3p silencing TLR4 and its downstream signaling pathway, to modulate the periodontal microenvironment [47,48]. CEVs derived from BMMSCs (BMMSC-EVs) was demonstrated to inhibit the inflammatory response of macrophages induced by Porphyromonas gingivalis (P. gingivalis), the keystone pathogen of periodontitis, and reduce the cell infiltration in rat periodontitis tissues [49]. Compared with BMMSC-EVs, gingival mesenchymal stem cell (GMSC)-derived CEVs (GMSC-EVs) promoted the expression of IL-10, Arg-1, CD206 and inhibit the expression of TNF-α in LPS-treated monocytes more effectively. GMSC-EVs could also influence T cell functions by suppressing CD4+ T cell activation, promoting Treg cell formation and inhibiting IFN-γ production. A further periodontitis model in rats also proved its effectiveness in treating periodontitis [50,51,52]. In addition, studies have shown that TNF-α stimulation could change the contents of CEVs to enhance the anti-inflammatory effect. Nakao et al. found TNF-α pre-treatment could promote the secretion of CD73-rich GMSC-EVs to induce M2 macrophage polarization [53]. Similarly, chitosan hydrogels loaded with DPSC-derived CEVs (DPSC-EVs) reduced the expression of IL-23, IL-1α, TNF-α, IL-12, IL-1β, IL-27, and IL-17 in periodontitis tissues. By DPSC-EV-delivered miR-1246, miR-125a-3p and other miRNAs, macrophage phenotype was transformed from the pro-inflammatory M1 subtype to the anti-inflammatory M2 subtype [54,55].
On the other hand, ICEVs such as macrophage-derived CEVs (M-EVs) could also regulate M2 macrophage polarization and reduce inflammation through miRNAs such as miR-99a/146b/378a [44]. Moreover, M2 macrophage-derived CEVs (M2-EVs) changed the proportion of different types of macrophages and increase the M2/M1 ratio in bone defect models [26]. M2-EVs were reported to be abundant in IL-10, which could be delivered to target cells such as macrophages and BMMSCs to enhance their anti-inflammatory functions [56]. In addition, the role of Treg cell-derived CEVs (Treg-EVs) in immune tolerance and inflammation inhibition is also worth attention [57,58].

2.3. CEVs Promote Periodontal Bone Regeneration

Alveolar bone regeneration is the most studied part in periodontal regeneration. In terms of treatment, SCEVs and ICEVs promoted stem cell adhesion, migration, proliferation and osteogenic differentiation. The merits of CEVs in bone tissue repair and regeneration has been broadly confirmed in bone fracture, bone defect, implant bone integration, periodontal defect, periodontitis and other models [26,59,60,61,62,63,64].
It can be concluded that SCEVs mediate periodontal bone formation mainly via regulating periodontal cell function and fate, modulating immune and inflammatory microenvironment, inhibiting osteoclast formation, promoting vascularization, enhancing antibacterial effects, delivering active ingredients, and interfering with multiple signaling pathways. Relevant study showed that CD73 on the surface of mesenchymal stem cell (MSC)-derived CEVs (MSC-EVs) activated the Akt and ERK signaling pathways to promote PDLC migration and proliferation. The collagen sponge loaded with MSC-EVs facilitated new bone formation in the periodontal defect area of rats [63]. SHED-EVs were proved to promote BMMSCs proliferation, osteogenic differentiation and also angiogenesis through AMPK signaling pathway. Wnt3a and BMP2 in SHED-EVs activated Wnt/β-catenin and BMP/Smad signaling pathways, respectively, to promote the osteogenic differentiation of PDLSCs, thereby playing a protective role in periodontal bone homeostasis [46,65,66]. Mohammed et al. demonstrated that adipose-derived stem cells (ADSCs) and the CEVs derived from them (ADSC-EVs) could be applied as an adjunct therapy for periodontitis treatment in rats. Particularly, ADSC-EVs promoted PDL repair and osteoid tissue generation [62]. BMMSC-EVs promoted TGF-β1 expression, migration, proliferation, osteogenic differentiation of PDLCs. At the same time, the M2 polarization of macrophages was induced and the activity of osteoclasts was inhibited by BMMSC-EVs. The nanocomposite hydrogel integrated with BMMSC-EVs could effectively repair the bone resorption in periodontitis rats [67]. Additionally, dental pulp (stem) cell (DPC/DPSC)-derived CEVs (DPC/DPSC-EVs) could facilitate MC3T3-E1 migration, suppress osteoclast formation, transfer miR-1246 to induce M2 macrophage polarization, and alleviate bone destruction in periodontitis mice [54,68]. In addition to regulating the phenotype of macrophages, SCEVs such as GMSC-EVs could inhibit T cell activation, increase the number of Treg cells, and finally promote bone remodeling in a rat periodontal defect model [50].
Increasing evidence suggests that SCEVs from mild inflammatory cytokine pre-stimulated cells are beneficial to periodontal bone formation. For example, CEVs derived from DPSCs pretreated with a low concentration of TNF-α (TNF-α-DPSC-EVs) delivered miR-758-5p targeting LMBR1 to activate BMP2/4 and promote the osteogenic differentiation of PDLSCs [69]. Similarly, LPS-pretreated dental follicle (stem) cell (DFC/DFSC)-derived CEVs (LPS-DFC-EVs/LPS-DFSC-EVs) promoted the migration, proliferation and osteogenic differentiation of PDLSCs, and exerted an antioxidant function through ROS/MAPK signaling pathway. By down-regulating RANKL/OPG ratio and reshaping macrophage phenotype, it accelerated bone reconstruction in periodontitis and periodontal defect models of rats and Beagle dogs [70,71,72]. Further, inflammatory induction, gene modification and mechanical stimulation also endowed SCEVs the effectiveness in promoting the osteogenic differentiation of stem cells [29,73,74].
Many studies demonstrated that ICEVs favored bone regeneration through remodeling the immune microenvironment and delivering bioactive molecules such as miRNAs. Several subtypes of macrophages present in various stages of bone healing, among which osteoclasts are the typical representatives for bone resorption, and osteomacs in bone remodeling units are the resident macrophages [75]. Macrophages, especially M2 macrophages, could promote periodontal and other bone regeneration [76,77,78,79]. To a large extent, M-EVs mediated the macrophage-based bone regeneration. It was found that both M-EVs and M2-EVs could assist skull defect repair in rats and femur fracture healing in mice, between which, M2-EVs were more effective [26,80]. It has also been reported that M2-EVs were inferior to M1 macrophage-derived CEVs (M1-EVs) in osteogenic induction [81]. However, most studies confirmed that M2-EVs could promote the osteogenesis of BMMSCs by delivering multiple miRNAs, such as miR-690, miR-5106 and miR-26a-5p [80,82,83]. In terms of periodontal bone regeneration, Liao et al. pointed out that M2-EVs might also promote the osteogenic differentiation of PDLSCs through a variety of miRNAs [84]. In addition to miRNAs, Chen et al. found that M2-EVs were rich in IL-10, which promoted BMMSC osteogenic differentiation through IL-10/IL-10R signaling pathway, inhibited osteoclast formation, and reduced bone resorption in mice with periodontitis [56]. Liu et al. investigated the effect of materials on the osteoinductive activity of M-EVs. Intrafibrillarly mineralized collagen (IMC) was found to be superior than extrafibrillarly mineralized collagen (EMC) in promoting M2 macrophage formation and rat mandibular defect repairing. IMC-induced macrophage-derived CEVs (IMC-EVs) improved MSC osteogenic differentiation through BMP2/Smad5 signaling pathway [85]. In addition to the material itself, physical topography, BMP-2 and Mg2+ pre-stimulated macrophage-derived EVs all showed higher osteogenic induction capacity via delivering miR-381 or activating autophagy [64,86,87].
Similarly, CEVs derived from monocytes (MC-EVs) induced the up-regulated expression of osteogenic-related genes in MSCs [88]. Schwann cell-derived CEVs (SC-EVs) promoted BMMSC osteogenic differentiation through TGF-β1/SMAD2/3 signaling pathway, M2 macrophage polarization, vascularization, nerve repair, and ultimately bone defect restoration [89]. Inflammatory osteoclast (iOC)-derived CEVs (iOC-EVs) have been proved to reduce the Osterix ubiquitination in MC3T3-E1 cells by transferring lncRNA LIOCE, thus facilitating bone regeneration [90]. In addition, Treg cells could play a positive role in bone regeneration by inhibiting over-activated T cells, reducing the proportion of Th1 and Th17 cells, and eliminating inflammation [77,91]. However, research on Treg-EVs in the field of regeneration is still lacking. In view of its role in remodeling M2 macrophage polarization and improving inflammatory microenvironment, the future of Treg-EV-based periodontal regeneration is promising [92,93].

2.4. CEVs Promote Cementogenesis

Cementum shares similarities with bone in structure, matrix composition, source of ectodermal mesenchymal progenitor cells. They also both secret non-collagenous proteins such as BSP, OPN, OCN, vitronectin and fibronectin [7,94]. So, many studies have looked at the two together [95,96,97,98]. From this point of view, SCEVs and ICEVs, which have positive effects on osteogenesis, can theoretically promote cementogenesis. However, different from periodontal bone regeneration, there is no typical cementum regeneration model due to the special anatomical position and structure of cementum and the lack of characteristic markers. Currently, periodontal defect model was the commonly used model to identify the newly formed cementum [99]. We previously used cementoblasts combined with collagen sponges to conduct subcutaneous ectopic transplantation in nude mice. In addition, other scholars also carried out studies on the establishment of root resorption model with cementum destruction under orthodontic force. However, none of the above models is mature [100,101]. Therefore, relevant studies mainly stay at the cellular level.
A variety of stem cells have been shown to be associated with cementogenesis, including Gli1+ PDL progenitor cells, PDL(S)Cs, DPSCs, periapical follicular stem cells (PAFSCs), and BMMSCs [95,98,102,103,104]. To our knowledge, the effects of SCEVs on cementum formation have not been explored yet. Whether SCEVs are beneficial to cementum formation as much as bone formation remains to be verified.
For immune cells, macrophages especially M2 macrophages are essential for cementum formation. Li et al. found that M2 macrophages promoted the cementoblastic differentiation of PDLSCs by secreting IL-10, VEGF and other cytokines to activate Akt signaling pathway [19]. Our recent work found that M2 macrophages possessed the chemotaxis to inflammatory microenvironment with high level of CCL2 from cementoblasts, and rescued P. gingivalis-suppressed cementoblast mineralization by activating p38 signaling pathway [105]. The effects of ICEVs on cementum formation was also poorly studied. Zhao et al. proved that M2-EVs could promote cementoblast mineralization. Additionally, they used the orthodontic tooth movement strategy combined with gingival crevicular injection of LPS to construct the cementum absorption model, followed by the injection of different kinds of M-EVs. Their results indicated that M2-EVs could also significantly promote cementoblast mineralization in vivo [106]. Our unpublished study demonstrated that Ckip-1 (casein kinase 2-interacting protein 1)-silenced macrophage-derived CEVs (sh-Ckip-1-EVs) possesses the characteristic of M2-EVs in facilitating cementoblast mineralization. It can be seen that cementum regeneration based on SCEVs and ICEVs still has great exploration space.

2.5. CEVs Promote Angiogenesis and Periodontal Soft Tissue Remodeling

Angiogenesis is essential for periodontal regeneration, which can synergically enhance the osteogenic differentiation of PDLSCs and other stem cells. The growth and remodeling of the skeletal system, including alveolar bone, are often accompanied by angiogenesis [107,108]. SCEVs are closely associated with angiogenesis. An abnormal increase in vascularization in gingiva and PDL due to inflammatory cell infiltration is among the most common features leading to the progression of periodontitis, and SCEVs may play a role in it. Research has revealed that down-regulation of miR-17-5p in inflammatory PDLSCs (iPDLSCs) leads to the up-regulation of VEGFA, which could be packed into iPDLSC-derived CEVs (iPDLSC-EVs), and then delivered to human umbilical vein endothelial cells (HUVECs) to enhance angiogenesis. Similar results were also verified in inflammatory DPSC (iDPSC)-derived CEVs (iDPSC-EVs) [109,110,111,112,113].
On the other hand, various SCEVs were found to promote angiogenesis, and further the bone regeneration by bioactive factors and improved HUVEC functions [114,115,116,117]. Pizzicannella et al. constructed 3D collagen membranes loaded with PDLSCs and polyethylenimine (PEI)-coated PDLSC-EVs (PEI-PDLSC-EVs). They found the membranes promoted bone defect repair by increasing the expression of VEGF, VEGFR2, and osteogenesis-related markers in PDLSCs [116]. One related study also showed that BMMSC-EVs promoted the proliferation, migration and tube formation capacity of HUVECs through HIF-1α/VEGF signaling pathway, thereby benefiting bone reconstruction [118,119]. Similar functions were demonstrated in umbilical cord mesenchymal stem cell (UCMSC)-derived CEVs (UCMSC-EVs), ADSC-EVs and DPSC-EVs [65,120,121,122]. CEV-based angiogenesis was also studied in periodontal regeneration. For example, SHED-EVs were found to facilitate HUVEC functions, and effectively promote the angiogenesis and bone regeneration in the periodontal defect areas of rats [65]. ADSC-EVs could be used to increase the number of small blood vessels in PDL and promote its’ remodeling [62].
The relationship between ICEVs and angiogenesis has also been studied. Aharon et al. found that in addition to regulating thrombosis and endothelial cell apoptosis, MC-EVs also significantly promoted HUVECs’ tube formation capacity [123]. Osteoclast-derived CEVs (OC-EVs) could improve the functions of HUVECs and CD31 expression. This angiogenesis-promoting effect could be enhanced ulteriorly by compression force [124]. The adverse effects of M-EVs on HUVEC function under certain conditions have ever been reported. However, M2-EVs could inhibit inflammation and apoptosis of HUVECs, and promote their functions by delivering miR-221-3p and miR-21 activating related signaling pathways [89,125,126]. Therefore, the degree of vascularization of tissues or scaffolds should be taken into consideration in a successful periodontal regeneration strategy. The collaboration of CEVs to enhance periodontal blood vessel and bone regeneration is a potential entry direction for periodontal regeneration.
The process of CEV-based periodontal regeneration is more or less accompanied by the remodeling of periodontal soft tissues including gingiva and PDL. The relevant studies mainly focus on SCEVs. Huang et al. showed that DFC-EVs, especially LPS-DFC-EVs, could reduce the number of P. gingivalis in the gingival tissues of mice with periodontitis, and the expression of IL-6 and IL-1β in gingiva and PDL [127]. Liu et al. constructed a mouse model of palatal gingival defect. Based on it, they found stem cell from apical papilla (SCAP)-derived CEVs (SCAP-EVs) promoted angiogenesis, type I collagen and fibronectin expression, gingival epithelium and connective tissue regeneration through Cdc42 [128]. PDL(S)Cs are essential cell types in PDL. The above mentioned MSC-EVs, BMMSC-EVs, ADSC-EVs, DPSC-EVs, DFSC-EVs and PDLC-EVs, which have therapeutic potential in periodontal bone regeneration, could not only promote the osteogenic/cementogenic differentiation of PDL(S)Cs, but also enhance their proliferation and migration ability. Further, they could improve blood and nutrient transport and alleviate inflammatory microenvironment, which ultimately promoting periodontal epithelium healing and PDL remodeling [47,54,62,63,67,70].
In summary, CEVs represented by SCEVs and ICEVs play an important role in the regeneration of periodontal hard and soft tissues. Compared to ICEVs, the researches on SCEVs are more mature. The above studies have emphasized the positive effects of CEVs on immune modulation, inflammation clearance, cell function regulation, angiogenesis, stem cell differentiation, periodontal tissue remodeling, etc., providing an effective strategy for CEV-based periodontal regeneration (Table 1).

3. BEVs and Periodontal Regeneration

3.1. Classification and Biogenesis of BEVs

Bacteria can release spherical membrane vesicles (20 to 400 nm) with lipid layers. These membrane vesicles affect different biological processes, including virulence, horizontal gene transfer, export of cellular metabolites, phage infection, and intercellular communications. BEVs can be classified into outer membrane vesicles (OMVs), outer-inner membrane vesicles (OIMVs), cytoplasmic membrane vesicles (CMVs), and tube-shaped membranous structures (TSMSs) [129].
With no outer membrane, Gram-positive bacteria mainly secrete CMVs, which are formed by endolysin-triggered ‘bubbling cell death’. CMVs contain cytosolic components and membrane components. In Gram-negative bacteria, budding from the outer membrane gives rise to formation of OMVs, which do not contain cytoplasmic components. Explosive cell lysis is caused by phage-produced endotoxin, which degrades the peptidoglycan cell wall to produce explosive outer membrane vesicles (EOMVs) and OIMVs. OIMVs can specifically encapsulate DNA, and EOMVs randomly contain cytoplasmic components compared to blister-forming OMVs [130,131]. Many factors can influence the biogenesis of OMVs. (1) The production of OMVs increases in regions with reduced Braun’s lipoprotein–peptidoglycan (Lpp-PG) crosslinks. (2) Insertion of the Pseudomonas quinolone signal (PQS) into the outer leaflet of the outer membrane can increase membrane curvature, leading to OMVs formation. (3) Increased OMVs production can be affected by accumulation of misfolded proteins or envelope components (such as lipopolysaccharide or peptidoglycan fragments), which lead to cross-linking occurring and outer membrane bulging. (4) Some lipid microdomains of the outer membrane have the tendency to bulge outward due to their charge, and this bulging leads to increased generation of OMVs [132].
Among the above-mentioned BEVs, studies related to OMVs are the most abundant and mature. In recent years, OMVs have received more and more attention in pathogenic mechanism and nanomaterial application. In the following, we mainly summarize the pathogenicity of OMVs, and their applications and prospects in periodontal regeneration.

3.2. Pathogenicity of BEVs in Periodontal Tissues

Firstly, as a typical representative of BEVs, OMVs have a protective effect on the bacteria themselves. Bacteria can respond to threats from the external environment by regulating OMVs production and components. For example, OMVs could protect bacteria by expelling misfolded proteins or toxic substances from the periplasmic cavity [133]. Secondly, OMVs play an important role in bacterial nutrient uptake; DNA and proteins in OMVs could serve as a direct source of nitrogen and phosphorus for bacterial growth. Specially, they could also transport hydrolases and polysaccharide lytic enzymes that assist bacteria lacking these enzymes to metabolize polysaccharides for nutrients [134,135]. Thirdly, OMVs can act as pseudo-targets for some antibiotics, protecting bacteria by absorbing or binding antibiotics. OMVs of Acinetobacter baylyi could enable negative strains to acquire resistance genes through horizontal gene transfer [136]. Moreover, OMVs can deliver virulence factors into the host, compromising the host immune system and thus promoting bacterial infection [137]. In addition, the threat of OMVs to the host cannot be ignored. OMVs are closely linked to epithelial cells and innate immune cells. Epithelial cells are the first line of defense of the host. On the one hand, OMVs could be recognized by pattern recognition receptors on the cell surface and induce or inhibit the production of cytokines and chemokines by epithelial cells. On the other hand, it could disrupt the tight junctions between epithelial cells and mucosal integrity, allowing the virulence factors to enter the submucosa and contact with neutrophils, dendritic cells, and macrophages [138]. Finally, after entering into the host cells, OMVs were recognized and then degraded, causing an inflammatory response, activating dendritic cells and promoting T-cell immunity. Activated CD40 ligands on the surface of helper T cells bind to homologous receptors on the surface of B cells, facilitating B cell proliferation and differentiation into effector B cells and memory B cells, facilitating antibody production [139,140,141].
There is a complex microbial ecosystem exists in the oral cavity of periodontitis patients, with the main pathogenic bacteria “red complex” (P. gingivalis, T. forsythia and T. denticola) [142]. Other Gram-negative bacteria such as A. actinomycetemcomitans and F. nucleatum also play an essential role in the development and progression of periodontitis [143]. The pathogenicity of BEVs in periodontal disease has been extensively studied. Researchers performed genomic DNA analysis of salivary vesicles from healthy individuals and patients with periodontal diseases. It was found that P. gingivalis-derived OMVs (Pg-EVs) were significantly increased in periodontitis patients [144]. Mass spectrometry-based high-throughput proteomic analysis revealed that gingipain, leukotoxin and peptidylarginine deiminase were enriched in OMVs from periodontal pathogenic bacteria [145]. Further, small RNAs could be delivered to the host cellular immune system via OMVs of P. gingivalis, A. actinomycetemcomitans, and T. forsythia [146]. For example, Pg-EVs can target chromobox 5 (CBX5) of PDLCs through sRNA45033 in OMVs, and promote apoptosis by regulating the methylation of p53 [147]. OMVs of periodontal pathogenic bacteria can also interact with host immune cells. OMVs from the “red complex” induced maturation of bone marrow dendritic cells and differentiation of naive CD4+ T cells into Th1 or Th17 cells [148]. F. nucleatum-derived OMVs (Fn-EVs) tended to polarize M0 macrophages to M1 macrophages, thereby exacerbating the alveolar bone destruction caused by periodontitis [149]. OMVs of periodontal pathogenic bacteria also have an effect on other periodontal pathogenic bacteria. For example, Pg-EVs could down-regulate the expression of the surface adhesion proteins FadA and FomA of F. nucleatum, and thus inhibiting the invasion ability of F. nucleatum [150]. The related literature further illustrates that BEVs can disrupt periodontal homeostasis in a variety of ways, including through virulence factors, promoting plaque biofilm formation, invading the immune system, activating inflammatory responses, damaging periodontal tissues, and inducing systemic diseases [29].

3.3. Potential Therapeutic Values of BEVs in Periodontal Treatment and Regeneration

As mentioned above, the pathological roles and mechanisms of BEVs in periodontitis have been extensively studied. However, their research in periodontal treatment and regeneration is still very scarce. According to previous studies, their potential value in periodontal treatment mainly includes the following aspects.
BEVs can be used as a vaccine for periodontitis. Some researchers have proposed that Pg-EVs could be stored for a long time for its structurally and functionally stability and resistant to proteinase K. Most importantly, it could carry Pg-EVs components into the host immune system. Mice immunized intranasally with OMVs + TLR3 agonist (Poly(I:C)) showed a significant reduction in the amount of P. gingivalis in the oral cavity, and elevated levels of serum IgG (including IgG1 and IgG2a) and s-IgA in saliva. The safety of Pg-EVs was also confirmed by intracerebral injection experiments [151]. Similar studies further suggested that Pg-EVs possessed strong immunogenicity and antigenicity, which may originate from the LPS and A-LPS-modified proteins in OMVs [152]. Furthermore, other research also confirmed the potential of other OMVs in vaccine development [153]. Therefore, Pg-EVs or other OMVs of periodontal pathogenic origin are likely to be an effective vaccine strategy for periodontitis in terms of bacterial clearance, safety, low cost, high efficiency, and ease of transport [154]. Overall, a reduction in P. gingivalis, a key pathogenic bacterium in periodontitis, would be beneficial in reducing the destruction of periodontal tissues and enhancing periodontal regeneration.
BEVs can act as drug delivery vehicles to exert antibacterial effects. periodontal pathogenic bacteria such as P. gingivalis and A. actinomycetemcomitans could evade antibiotics by parasitizing human epithelial cells. High permeability of BEVs gave the possibility for drugs such as antibiotics to enter the cells and kill the bacteria efficiently [155]. Huang et al. found that the application of sublethal doses of quinolone antibiotics to bacterial culture media induced bacteria to load high doses of antibiotics in OMVs. There was an increase in the number of particles, protein content and particle size of the vesicles after antibiotic treatment, while the nanostructure of the vesicles was not affected. Such highly biocompatible antibiotic-loaded vesicles could efficiently invade bacteria and release drugs, thereby effectively killing a wide range of Gram-negative bacteria and significantly reducing the amount of bacteria in the small intestine and feces of infected mice [156]. Thus, we hypothesize that drug-loaded BEVs could likewise be an effective strategy for suppressing periodontal pathogenic bacteria. However, their effectiveness in periodontal regeneration needs to be further investigated.
Saliva-derived EVs containing BEVs can be used for early detection of periodontitis. Han et al. showed that BEVs could circulate in saliva. Saliva-derived EVs were more suitable for biomarkers for early detection of periodontal status than saliva itself. BEVs can reflect the changes in periodontal bacteria. Researchers examined saliva-derived EVs and found a high percentage of periodontal pathogenic bacteria (T. denticola, E. corrodens, P. gingivalis, and F. nucleatum)-related BEVs in periodontitis patients [144]. For the benefits of early detection and prevention of periodontitis, BEVs could be used for the assessment of oral microecological environment in periodontal regeneration state.
Lastly, BEVs act as biological shuttle systems to detect molecular transfer between microorganisms and host cells. Miriam Bittel et al. applied E. coli to express Cre recombinase and produced OMVs loaded with Cre recombinase. Then, Cre protein activated the Cre-LoxP reporter system of genetically engineered murine intestinal cells and express tdTomato fluorescent protein. This technique could enable biomolecular visualization of biomolecular transferring. In addition to the intestine, bacterial-derived Cre induced extended marker gene expression in a wide range of host tissues, including heart, liver, spleen, kidney, and brain [157]. By analogy, periodontal pathogenic bacteria-derived OMVs expressing Cre recombinase could theoretically be used to visualize periodontal-systemic disease correlations, and even facilitate the development of visualized drug delivery-based periodontal therapy and regeneration. However, compared to CEVs, prospects for translation to therapy are far off for the reason that the role of BEVs in periodontal regeneration is limited and indirect. Thus, further applications need to be explored.

4. PEVs and Periodontal Regeneration

4.1. Classification and Biogenesis of PEVs

In this review, PEVs refers to the general term for vesicular nanoparticles isolated from plants. They have also been named as plant-derived exosome-like nanoparticles (PELNs), plant-derived vesicle-like nanoparticles (PDVLNs), plant-derived nanovesicles (PDNVs), etc., including the vesicles from extracellular apoplastic fluids and from tissue crushing fluids [158]. PEVs and mammalian EVs are similar in terms of morphology, size and other physical characteristics. PEVs are also cup-shaped with different particle sizes in different plants. Even in the same plant, there are vesicles with various sizes ranging from 50 to 500 nm. Compared with mammalian EVs, the study of PEVs is still in their infancy. Due to the unstandardized means of isolation and physical characterization of PEVs, and the lack of generic or specific markers of PEVs and plant species antibodies, it is difficult to use the nomenclature for CEVs to similarly classify PEVs [32,158,159].
PEVs can be secreted into the apoplast space. Compared with CEVs, the biological mechanism of PEVs formation is less studied, containing the following secretion models. (1) MVBs is a conventional pathway similar to exosome secretion. Initially, Halperin and Jensen observed the MVBs fused with the plasma membrane of carrot cells, followed by the release of vesicle-like structures into the wall spaces. Similar phenomenon was also observed in Arabidopsis infected with turnip mosaic virus [160,161]. (2) Exocyst positive organelles (EXPOs) pathway is a plant-specific secretory form. EXPOs are spherical, bilayer structures similar to autophagosomes. It has been reported that the outer membrane of EXPOs in Arabidopsis and tobacco cells could fuse with the plasma membrane to release monolayer vesicles into the apoplast space. EXPOs were also found in other cell types, such as root hair cells, root tips, and pollen grains [162]. (3) Vacuole, the dynamic organelles of plant cells, is another unconventional secretion approach. One related study confirmed that vacuoles of Arabidopsis cells, could mediate the auto-immune process by fusing with the plasma membrane to release antibacterial and cell death-inducing proteins [163]. In addition, vacuole pathway and MVBs pathway are closely related and intersected [164]. (4) In addition, the autophagosome pathway has also been reported to potentially present in plants [165].
PEVs are rich in sources and can be isolated from the fruits, roots, stems and leaves of various plants such as ginseng, sunflower seeds, oats, Arabidopsis, ginger, broccoli, bitter melon, grape, grapefruit, lemon, blueberry and orange. Small nucleic acids, proteins, lipids and small-molecule metabolites contained in PEVs are the main functional substances [166]. PEVs have fewer ethical problems in clinical transformation due to their plant origin, with advantages of low immunogenicity and wide availability. Hence, they can be used as safe and economical therapeutic drugs and delivery carriers [167,168]. In recent years, increasing evidence suggests that PEVs possess great potential to modulate immunity, inflammation, microbiome, and tissue regeneration [169]. Here, we mainly focus on their functions to summarize and prospect the therapeutic effects of PEVs in periodontitis treatment and periodontal regeneration.

4.2. PEVs Regulate Immune and Inflammatory Responses

PEVs have been demonstrated to regulate inflammation, antioxidant factor expression, and macrophage polarization. One recent study showed that orally administrated ginger-derived PEVs (Gr-EVs) could be absorbed by mouse intestinal macrophages, then promoted the expression of HO-1, IL-10 and Nrf2, and activated the anti-inflammatory Wnt/Tcf4 signaling pathway [170]. In addition to up-regulating the expression of IL-10 and HO-1, tea leaf-derived PEVs (TL-EVs) also decreased the expression of pro-inflammatory cytokines TNF-α, IL-6, IL-12, and ROS in LPS-stimulated macrophages [171]. Gin-EVs and shiitake mushroom-derived PEVs (SM-EVs) were involved in the inhibition of NLRP3 inflammasome assembly, IL-1β and IL-18 secretion, and pyroptosis in macrophages [172,173]. Cabbage-derived PEVs (Ce-EVs) and red cabbage-derived PEVs (RCe-EVs) were reported to inhibit the M1 macrophage polarization, and the apoptosis of keratinocytes and fibroblasts [174]. Similar anti-inflammatory, antioxidant and macrophage-regulatory effects have also been confirmed in PEVs derived from Aloe saponaria (AS-EVs), aloe vera peels (AVP-EVs), and grapefruits (GF-EVs) [175,176,177].
PEVs could regulate immune and inflammatory responses through a variety of components and signaling pathways. (1) miRNAs. Aquilano et al. showed that PEVs derived from dried nuts (Nut-EVs) could deliver miR-159a and miR-156c to reduce the expression of Tnfrsf1a protein in adipocytes and inhibit the TNF-α signaling pathway, to exert anti-inflammatory effects [178]. (2) Lipids. Deng et al. reported that Broccoli-derived PEVs (Bi-EVs) activated the AMPK signaling via lipid components, inhibited dendritic cell activation, induced tolerant dendritic cell formation, and ultimately alleviated DSS-induced colitis in mice [179]. (3) Proteins. Sriwastva et al. proved that Mulberry bark derived PEVs (MB-EVs) could alleviate mice colitis by activating AhR/COPS8 signaling pathway through HSPA8 [180]. (4) Phenolic compounds. Paiotti et al. also reported that grape juice containing phenolic compounds was protective against TNBS-induced colitis [181]. The same results have been obtained in various inflammatory models such as colitis and viral pneumonia [182,183]. However, PEVs were poorly studied in periodontal inflammation with only one related study. In this study, phosphatidic acid in Gr-EVs was demonstrated to reduce P. gingivalis-induced periodontal inflammation, characterized by decreased expression of IL-1β, IL-6, IL-8 and TNF-α in periodontal tissues, and reduced inflammatory infiltration in PDL [184]. All the above studies revealed the important role of PEVs in regulating immune and inflammatory responses, which are also the manifestations of periodontitis. Their potential application values in the treatment of periodontitis and periodontal regeneration need to be further explored.

4.3. PEVs Reshape Microecology

On the other hand, periodontitis is a secondary inflammatory reaction caused by the imbalance of oral microecology. Maintaining the dynamic balance of oral microecology is vital to control the progression of periodontitis and improve the regeneration [1,2]. PEVs can mediate the interaction between plants and microorganisms. Under the stimulation of pathogens, plants themselves can secrete PEVs carrying a variety of sRNA and lipid components, playing an important role in the auto-immune process against bacteria and fungi. In addition, the mutualism between plants and microorganisms also involves the participation of PEVs [158,185]. In terms of therapy, PEVs could enhance the growth of probiotics. Lei et al. reported that lemon-derived PEVs (Ln-EVs) inhibited the production of Msp1 and Msp3 through RNaseP-mediated degradation of specific tRNA, thereby improving the bile tolerance of Lactobacillus rhamnosus GG [186]. PEVs could also inhibit the keystone periodontal pathogen. Sundaram et al. showed that the phosphatidic acid (34:2) in Gr-EVs interacted with the HBP35 on the outer membrane of P. gingivalis to mediate the internalization of Gr-EVs. The internalized Gr-EVs further inhibited the proliferation, growth, adhesion and invasion of the bacteria and reducing its virulence [184]. Therefore, regulating microecology through PEVs may potentially be an effective approach for periodontitis treatment and periodontal regeneration.

4.4. The Therapeutic Values of PEVs in Periodontal Regeneration

CEVs showed positive roles in various tissue regeneration. Given the similarity between PEVs and CEVs in structure and composition, PEVs also have potential values in tissue regeneration theoretically [187]. PEVs can enhance the proliferation and migration of various soft tissue-related cells, as well as angiogenesis, and have therapeutic effects on tissue healing and repair. AS-EVs were proved to enhance the proliferation and migration of human dermal fibroblasts (HDFs), and promote tube formation capacity of HUVECs [175]. In addition to improving HUVEC functions, GF-EVs were demonstrated to promote the proliferation, migration, and the expression of soft tissue healing-related markers such as type I collagen, fibronectin, laminin and vimentin in human immortalized epidermal cells (HaCaT cells) [176]. Similarly, cell function-promoting effects of wheat-derived PEVs (Wt-EVs) was also shown in HUVECs, HDFs and HaCaT cells [188]. In summary, these studies all revealed the potential role of PEVs in periodontal angiogenesis and soft tissue regeneration.
PEVs also regulate the function and fate of stem cells by their components. Ju et al. showed that grape-derived PEVs (Ge-EVs) contribute to the expression of growth-associated genes of Lgr5+ intestinal stem cells and their proliferation by activating the Wnt/β-catenin signaling pathway through their lipid components, thereby favoring the repair of the intestinal epithelium stimulated with DSS [183]. Strawberry-derived PEVs (Sy-EVs) promoted the early proliferation of ADSCs, and enhanced their antioxidant ability to reduce ROS production through vitamin C [189]. Ginseng-derived PEVs (Gg-EVs) could promote the neural differentiation of BMMSCs in vivo and in vitro by delivering miRNAs [190]. Therefore, it is reasonable to speculate that PEVs may also have positive regulatory effects on the proliferation, differentiation and other functions of various periodontal stem cells. Relevant works need to be further expanded.
PEVs are beneficial to periodontal hard tissue repair and regeneration. A recent study demonstrated that yam-derived PEVs (Yam-EVs) improved osteoblast mineralization for bone regeneration in ovariectomized (OVX)-induced osteoporotic mice by activating the BMP-2/p-p38-dependent Runx2 pathway [191]. As mentioned above, Sundaram et al. confirmed that Gr-EVs influenced a series of functions of P. gingivalis and reduced the periodontal inflammation through phosphatidic acid. They could also further reduce the number of Trap-positive osteoclasts, increase the number of Runx2-positive osteoblasts, and relieve the alveolar bone loss induced by P. gingivalis [184].
In conclusion, research on PEV-based periodontal regeneration is still lacking. Similar to CEVs, PEVs can also carry a variety of effective components to target cells to be involved in soft and hard tissue remodeling, which were expected to become a new strategy for periodontal regeneration.

5. Conclusions, Challenges and Prospects

EVs can be secreted by almost all cells in mammals, bacteria and plants, and the production of EVs runs through the whole life process. It seems that it is not a random event, but an active behavior of living organisms trying to change their surroundings. For example, CEVs mediate the interaction between eukaryotic cells, BEVs attack host cells, and PEVs resist the invasion of exogenous pathogens [192]. It is a great progress in medical field to be familiar with, understand and then master the characteristics and rules of EVs, and apply it in disease treatment and regenerative therapy. Periodontitis is an immune- inflammatory response caused by periodontal pathogens, involving immunity, inflammation, periodontal microecology, periodontal tissue destruction and many other aspects. EV-based periodontal regeneration is a new and promising strategy for repairing and rebuilding damaged periodontal tissues.
At present, there also exist some common issues to be improved in EV-based periodontal therapy, such as standardizing EV extraction methods, exploring new combined methods that can improve EVs yield and purity, reducing the clearance of EVs and increasing its targeting ability in vivo, and developing new multifunctional biomaterials with EVs retention capacity [29]. CEVs, BEVs and PEVs have similar physical properties such as morphology, structure, size and electric potential. They all have heterogeneity in many aspects and can carry different types of substances such as small RNAs, proteins, lipids and metabolites to target cells to perform specific functions. There are also differences. These include the functional substances, the amount and cost of access, and the possibility of clinical transformation. Compared with CEVs and BEVs, PEVs need further research in terms of extraction method and naming standardization, subtype classification, and marker discovery. In addition, research on BEVs mainly focus on the pathogenic effect in periodontitis, while CEVs and PEVs are more likely to be applied in periodontal regeneration. CEVs are maturely studied in periodontal treatment and regeneration, while PEVs are still in their infancy [29,167].
All in all, this review focuses on the multiple functions of EVs, such as immune regulation, anti-inflammation, reshaping microecology, regulating cell function and fate, and promoting soft and hard tissue regeneration. The latest research progress and potential application values of CEVs, BEVs and PEVs in periodontal regeneration are comprehensively expounded and discussed (Figure 1). In addition to CEVs, BEVs, and PEVs, some other EVs subtypes such as apoptotic cell-derived EVs (ApoEVs) and matrix vesicles (MVs) were also demonstrated to play positive roles in promoting osteogenesis, mineralization, and periodontal regeneration [193,194]. Moreover, the specific mechanisms of EV biogenesis, the development of EV-friendly scaffold materials, the synergistic function of EVs as a carrier to deliver genes and drugs, the transformation of waste into treasure (EVs in feces and urine), and the applications of engineered EVs are also the directions for further research in the field of periodontal regeneration. Although EV-based periodontal regeneration has made great progresses, we still face many unsolved problems, and its clinical transformation will be a long-term challenge. Nevertheless, it is only a matter of time. We should believe that it will have a bright future.

Author Contributions

Conceptualization, X.H. and H.W.; writing—original draft preparation, X.H. and H.W.; writing—review and editing, X.H. and C.W.; supervision, Z.C.; project administration, Z.C.; funding acquisition, C.W. and Z.C. All authors have read and agreed to the published version of the manuscript.

Funding

This review was supported by the National Natural Science Foundation of China to Zhengguo Cao (No. 82170963), and the Fundamental Research Funds for the Central Universities (No. 2042022kf1167, Wuhan University).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kinane, D.F.; Stathopoulou, P.G.; Papapanou, P.N. Periodontal diseases. Nat. Rev. Dis. Prim. 2017, 3, 17038. [Google Scholar] [CrossRef] [PubMed]
  2. Genco, R.J.; Sanz, M. Clinical and public health implications of periodontal and systemic diseases: An overview. Periodontol 2000 2020, 83, 7–13. [Google Scholar] [CrossRef]
  3. Marruganti, C.; Baima, G.; Aimetti, M.; Grandini, S.; Sanz, M.; Romandini, M. Periodontitis and low cognitive performance: A population-based study. J. Clin. Periodontol. 2023. [Google Scholar] [CrossRef] [PubMed]
  4. Chisci, G.; Gabriele, G.; Gennaro, P. Periodontal disease before and after fractures of the mandible. Br. J. Oral Maxillofac. Surg. 2023, 61, 116. [Google Scholar] [CrossRef] [PubMed]
  5. Heitz-Mayfield, L.J.A.; Lang, N.P. Surgical and nonsurgical periodontal therapy. Learned and unlearned concepts. Periodontol 2000 2013, 62, 218–231. [Google Scholar] [CrossRef]
  6. Bosshardt, D.D.; Sculean, A. Does periodontal tissue regeneration really work? Periodontol 2000 2009, 51, 208–219. [Google Scholar] [CrossRef]
  7. Arzate, H.; Zeichner-David, M.; Mercado-Celis, G. Cementum proteins: Role in cementogenesis, biomineralization, periodontium formation and regeneration. Periodontol 2000 2015, 67, 211–233. [Google Scholar] [CrossRef]
  8. Zhao, J.; Faure, L.; Adameyko, I.; Sharpe, P.T. Stem cell contributions to cementoblast differentiation in healthy periodontal ligament and periodontitis. Stem Cells 2021, 39, 92–102. [Google Scholar] [CrossRef]
  9. Nuñez, J.; Vignoletti, F.; Caffesse, R.G.; Sanz, M. Cellular therapy in periodontal regeneration. Periodontol 2000 2019, 79, 107–116. [Google Scholar] [CrossRef]
  10. Barone, A.; Toti, P.; Quaranta, A.; Alfonsi, F.; Cucchi, A.; Calvo-Guirado, J.L.; Negri, B.; Di Felice, R.; Covani, U. Volumetric analysis of remodelling pattern after ridge preservation comparing use of two types of xenografts. A multicentre randomized clinical trial. Clin. Oral Implant. Res. 2016, 27, e105–e115. [Google Scholar] [CrossRef]
  11. Sassano, P.; Gennaro, P.; Chisci, G.; Gabriele, G.; Aboh, I.V.; Mitro, V.; di Curzio, P. Calvarial onlay graft and submental incision in treatment of atrophic edentulous mandibles: An approach to reduce postoperative complications. J. Craniofac. Surg. 2014, 25, 693–697. [Google Scholar] [CrossRef] [PubMed]
  12. Majzoub, J.; Ravida, A.; Starch-Jensen, T.; Tattan, M.; Suárez-López Del Amo, F. The Influence of Different Grafting Materials on Alveolar Ridge Preservation: A Systematic Review. J. Oral Maxillofac. Res. 2019, 10, e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Chisci, G.; Fredianelli, L. Therapeutic Efficacy of Bromelain in Alveolar Ridge Preservation. Antibiotics 2022, 11, 1542. [Google Scholar] [CrossRef] [PubMed]
  14. Kassebaum, N.J.; Bernabé, E.; Dahiya, M.; Bhandari, B.; Murray, C.J.L.; Marcenes, W. Global burden of severe periodontitis in 1990-2010: A systematic review and meta-regression. J. Dent. Res. 2014, 93, 1045–1053. [Google Scholar] [CrossRef]
  15. Chen, F.-M.; Zhang, J.; Zhang, M.; An, Y.; Chen, F.; Wu, Z.-F. A review on endogenous regenerative technology in periodontal regenerative medicine. Biomaterials 2010, 31, 7892–7927. [Google Scholar] [CrossRef]
  16. Spiller, K.L.; Koh, T.J. Macrophage-based therapeutic strategies in regenerative medicine. Adv. Drug Deliv. Rev. 2017, 122, 74–83. [Google Scholar] [CrossRef]
  17. Julier, Z.; Park, A.J.; Briquez, P.S.; Martino, M.M. Promoting tissue regeneration by modulating the immune system. Acta Biomater. 2017, 53, 13–28. [Google Scholar] [CrossRef]
  18. Chen, F.-M.; Jin, Y. Periodontal tissue engineering and regeneration: Current approaches and expanding opportunities. Tissue Eng. Part B Rev. 2010, 16, 219–255. [Google Scholar] [CrossRef]
  19. Li, X.; He, X.-T.; Kong, D.-Q.; Xu, X.-Y.; Wu, R.-X.; Sun, L.-J.; Tian, B.-M.; Chen, F.-M. M2 Macrophages Enhance the Cementoblastic Differentiation of Periodontal Ligament Stem Cells via the Akt and JNK Pathways. Stem Cells 2019, 37, 1567–1580. [Google Scholar] [CrossRef]
  20. Gruber, R. Osteoimmunology: Inflammatory osteolysis and regeneration of the alveolar bone. J. Clin. Periodontol. 2019, 46, 52–69. [Google Scholar] [CrossRef] [Green Version]
  21. Lin, H.; Chen, H.; Zhao, X.; Chen, Z.; Zhang, P.; Tian, Y.; Wang, Y.; Ding, T.; Wang, L.; Shen, Y. Advances in mesenchymal stem cell conditioned medium-mediated periodontal tissue regeneration. J. Transl. Med. 2021, 19, 456. [Google Scholar] [CrossRef]
  22. Iwasaki, K.; Peng, Y.; Kanda, R.; Umeda, M.; Ishikawa, I. Stem Cell Transplantation and Cell-Free Treatment for Periodontal Regeneration. Int. J. Mol. Sci. 2022, 23, 1011. [Google Scholar] [CrossRef]
  23. Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar] [CrossRef] [PubMed]
  24. Raposo, G.; Stahl, P.D. Extracellular vesicles: A new communication paradigm? Nat. Rev. Mol. Cell Biol. 2019, 20, 509–510. [Google Scholar] [CrossRef]
  25. Kumar, L.P.; Kandoi, S.; Misra, R.; Vijayalakshmi, S.; Rajagopal, K.; Verma, R.S. The mesenchymal stem cell secretome: A new paradigm towards cell-free therapeutic mode in regenerative medicine. Cytokine Growth Factor Rev. 2019, 46, 1–9. [Google Scholar] [CrossRef]
  26. Kang, M.; Huang, C.-C.; Lu, Y.; Shirazi, S.; Gajendrareddy, P.; Ravindran, S.; Cooper, L.F. Bone regeneration is mediated by macrophage extracellular vesicles. Bone 2020, 141, 115627. [Google Scholar] [CrossRef] [PubMed]
  27. Kalluri, R.; LeBleu, V.S. The biology function and biomedical applications of exosomes. Science 2020, 367, 6977. [Google Scholar] [CrossRef]
  28. Gholami, L.; Nooshabadi, V.T.; Shahabi, S.; Jazayeri, M.; Tarzemany, R.; Afsartala, Z.; Khorsandi, K. Extracellular vesicles in bone and periodontal regeneration: Current and potential therapeutic applications. Cell Biosci. 2021, 11, 16. [Google Scholar] [CrossRef]
  29. Deng, D.-K.; Zhang, J.-J.; Gan, D.; Zou, J.-K.; Wu, R.-X.; Tian, Y.; Yin, Y.; Li, X.; Chen, F.-M.; He, X.-T. Roles of extracellular vesicles in periodontal homeostasis and their therapeutic potential. J. Nanobiotechnol. 2022, 20, 545. [Google Scholar] [CrossRef]
  30. Díaz-Garrido, N.; Badia, J.; Baldomà, L. Microbiota-derived extracellular vesicles in interkingdom communication in the gut. J. Extracell. Vesicles 2021, 10, e12161. [Google Scholar] [CrossRef]
  31. Hua, S.; Bartold, P.M.; Gulati, K.; Moran, C.S.; Ivanovski, S.; Han, P. Periodontal and Dental Pulp Cell-Derived Small Extracellular Vesicles: A Review of the Current Status. Nanomaterials 2021, 11, 1858. [Google Scholar] [CrossRef]
  32. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Babst, M. MVB vesicle formation: ESCRT-dependent, ESCRT-independent and everything in between. Curr. Opin. Cell Biol. 2011, 23, 452–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Cocucci, E.; Racchetti, G.; Meldolesi, J. Shedding microvesicles: Artefacts no more. Trends Cell Biol. 2009, 19, 43–51. [Google Scholar] [CrossRef] [PubMed]
  35. Ratajczak, M.Z.; Ratajczak, J. Extracellular microvesicles/exosomes: Discovery, disbelief, acceptance, and the future? Leukemia 2020, 34, 3126–3135. [Google Scholar] [CrossRef] [PubMed]
  36. Muralidharan-Chari, V.; Clancy, J.; Plou, C.; Romao, M.; Chavrier, P.; Raposo, G.; D’Souza-Schorey, C. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr. Biol. 2009, 19, 1875–1885. [Google Scholar] [CrossRef] [Green Version]
  37. Van Engeland, M.; Nieland, L.J.; Ramaekers, F.C.; Schutte, B.; Reutelingsperger, C.P. Annexin V-affinity assay: A review on an apoptosis detection system based on phosphatidylserine exposure. Cytometry 1998, 31, 1–9. [Google Scholar] [CrossRef]
  38. Shao, H.; Im, H.; Castro, C.M.; Breakefield, X.; Weissleder, R.; Lee, H. New Technologies for Analysis of Extracellular Vesicles. Chem. Rev. 2018, 118, 1917–1950. [Google Scholar] [CrossRef]
  39. Akers, J.C.; Gonda, D.; Kim, R.; Carter, B.S.; Chen, C.C. Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neurooncol. 2013, 113, 1–11. [Google Scholar] [CrossRef] [Green Version]
  40. Coleman, M.L.; Sahai, E.A.; Yeo, M.; Bosch, M.; Dewar, A.; Olson, M.F. Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I. Nat. Cell Biol. 2001, 3, 339–345. [Google Scholar] [CrossRef]
  41. Medzhitov, R. Inflammation 2010: New adventures of an old flame. Cell 2010, 140, 771–776. [Google Scholar] [CrossRef] [Green Version]
  42. Sanwlani, R.; Gangoda, L. Role of Extracellular Vesicles in Cell Death and Inflammation. Cells 2021, 10, 2663. [Google Scholar] [CrossRef] [PubMed]
  43. Tang, D.; Cao, F.; Yan, C.; Fang, K.; Ma, J.; Gao, L.; Sun, B.; Wang, G. Extracellular Vesicle/Macrophage Axis: Potential Targets for Inflammatory Disease Intervention. Front. Immunol. 2022, 13, 705472. [Google Scholar] [CrossRef] [PubMed]
  44. Bouchareychas, L.; Duong, P.; Covarrubias, S.; Alsop, E.; Phu, T.A.; Chung, A.; Gomes, M.; Wong, D.; Meechoovet, B.; Capili, A.; et al. Macrophage Exosomes Resolve Atherosclerosis by Regulating Hematopoiesis and Inflammation via MicroRNA Cargo. Cell Rep. 2020, 32, 107881. [Google Scholar] [CrossRef] [PubMed]
  45. Zhao, M.; Liu, S.; Wang, C.; Wang, Y.; Wan, M.; Liu, F.; Gong, M.; Yuan, Y.; Chen, Y.; Cheng, J.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Mitochondrial Damage and Inflammation by Stabilizing Mitochondrial DNA. ACS Nano 2021, 15, 1519–1538. [Google Scholar] [CrossRef]
  46. Wei, J.; Song, Y.; Du, Z.; Yu, F.; Zhang, Y.; Jiang, N.; Ge, X. Exosomes derived from human exfoliated deciduous teeth ameliorate adult bone loss in mice through promoting osteogenesis. J. Mol. Histol. 2020, 51, 455–466. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, Z.; Maruyama, K.; Sakisaka, Y.; Suzuki, S.; Tada, H.; Suto, M.; Saito, M.; Yamada, S.; Nemoto, E. Cyclic Stretch Force Induces Periodontal Ligament Cells to Secrete Exosomes That Suppress IL-1β Production Through the Inhibition of the NF-κB Signaling Pathway in Macrophages. Front. Immunol. 2019, 10, 1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Han, X.-D.; Chen, H.-M.; 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. [Google Scholar] [CrossRef]
  49. Yue, C.; Cao, J.; Wong, A.; Kim, J.H.; Alam, S.; Luong, G.; Talegaonkar, S.; Schwartz, Z.; Boyan, B.D.; Giannobile, W.V.; et al. Human Bone Marrow Stromal Cell Exosomes Ameliorate Periodontitis. J. Dent. Res. 2022, 101, 1110–1118. [Google Scholar] [CrossRef]
  50. Zarubova, J.; Hasani-Sadrabadi, M.M.; Dashtimoghadam, E.; Zhang, X.; Ansari, S.; Li, S.; Moshaverinia, A. Engineered Delivery of Dental Stem-Cell-Derived Extracellular Vesicles for Periodontal Tissue Regeneration. Adv. Health Mater. 2022, 11, e2102593. [Google Scholar] [CrossRef]
  51. Zhang, Y.; Wang, Z.; Shi, B.; Li, Y.; Wang, R.; Sun, J.; Hu, Y.; Yuan, C.; Xu, Q. Effect of gingival mesenchymal stem cell-derived exosomes on inflammatory macrophages in a high-lipid microenvironment. Int. Immunopharmacol. 2021, 94, 107455. [Google Scholar] [CrossRef]
  52. Wang, R.; Ji, Q.; Meng, C.; Liu, H.; Fan, C.; Lipkind, S.; Wang, Z.; Xu, Q. Role of gingival mesenchymal stem cell exosomes in macrophage polarization under inflammatory conditions. Int. Immunopharmacol. 2020, 81, 106030. [Google Scholar] [CrossRef]
  53. Nakao, Y.; Fukuda, T.; Zhang, Q.; Sanui, T.; Shinjo, T.; Kou, X.; Chen, C.; Liu, D.; Watanabe, Y.; Hayashi, C.; 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. [Google Scholar] [CrossRef]
  54. Shen, Z.; Kuang, S.; Zhang, Y.; Yang, M.; Qin, W.; Shi, X.; Lin, Z. 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. [Google Scholar] [CrossRef]
  55. Zheng, J.; Kong, Y.; Hu, X.; Li, Z.; Li, Y.; Zhong, Y.; Wei, X.; Ling, J. 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. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, X.; Wan, Z.; Yang, L.; Song, S.; Fu, Z.; Tang, K.; Chen, L.; Song, Y. Exosomes derived from reparative M2-like macrophages prevent bone loss in murine periodontitis models via IL-10 mRNA. J. Nanobiotechnol. 2022, 20, 110. [Google Scholar] [CrossRef] [PubMed]
  57. Rojas, C.; Campos-Mora, M.; Cárcamo, I.; Villalón, N.; Elhusseiny, A.; Contreras-Kallens, P.; Refisch, A.; Gálvez-Jirón, F.; Emparán, I.; Montoya-Riveros, A.; et al. T regulatory cells-derived extracellular vesicles and their contribution to the generation of immune tolerance. J. Leukoc Biol. 2020, 108, 813–824. [Google Scholar] [CrossRef] [PubMed]
  58. Sullivan, J.A.; Tomita, Y.; Jankowska-Gan, E.; Lema, D.A.; Arvedson, M.P.; Nair, A.; Bracamonte-Baran, W.; Zhou, Y.; Meyer, K.K.; Zhong, W.; et al. Treg-Cell-Derived IL-35-Coated Extracellular Vesicles Promote Infectious Tolerance. Cell Rep. 2020, 30, 1039–1051. [Google Scholar] [CrossRef]
  59. Wang, X.; Thomsen, P. Mesenchymal stem cell-derived small extracellular vesicles and bone regeneration. Basic Clin. Pharmacol. Toxicol. 2021, 128, 18–36. [Google Scholar] [CrossRef]
  60. Furuta, T.; Miyaki, S.; Ishitobi, H.; Ogura, T.; Kato, Y.; Kamei, N.; Miyado, K.; Higashi, Y.; Ochi, M. Mesenchymal Stem Cell-Derived Exosomes Promote Fracture Healing in a Mouse Model. Stem Cells Transl. Med. 2016, 5, 1620–1630. [Google Scholar] [CrossRef] [Green Version]
  61. Li, W.; Liu, Y.; Zhang, P.; Tang, Y.; Zhou, M.; Jiang, W.; Zhang, X.; Wu, G.; Zhou, Y. Tissue-Engineered Bone Immobilized with Human Adipose Stem Cells-Derived Exosomes Promotes Bone Regeneration. ACS Appl. Mater. Interfaces 2018, 10, 5240–5254. [Google Scholar] [CrossRef] [PubMed]
  62. Mohammed, E.; Khalil, E.; Sabry, D. Effect of Adipose-Derived Stem Cells and Their Exo as Adjunctive Therapy to Nonsurgical Periodontal Treatment: A Histologic and Histomorphometric Study in Rats. Biomolecules 2018, 8, 167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Chew, J.R.J.; Chuah, S.J.; Teo, K.Y.W.; Zhang, S.; Lai, R.C.; Fu, J.H.; Lim, L.P.; Lim, S.K.; Toh, W.S. Mesenchymal stem cell exosomes enhance periodontal ligament cell functions and promote periodontal regeneration. Acta Biomater. 2019, 89, 252–264. [Google Scholar] [CrossRef] [PubMed]
  64. Wei, F.; Li, M.; Crawford, R.; Zhou, Y.; Xiao, Y. Exosome-integrated titanium oxide nanotubes for targeted bone regeneration. Acta Biomater. 2019, 86, 480–492. [Google Scholar] [CrossRef]
  65. Wu, J.; Chen, L.; Wang, R.; Song, Z.; Shen, Z.; Zhao, Y.; Huang, S.; Lin, Z. 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. [Google Scholar] [CrossRef]
  66. 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. [Google Scholar] [CrossRef]
  67. Liu, L.; Guo, S.; Shi, W.; Liu, Q.; Huo, F.; Wu, Y.; Tian, W. Bone Marrow Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Promote Periodontal Regeneration. Tissue Eng. Part A 2021, 27, 962–976. [Google Scholar] [CrossRef]
  68. Shimizu, Y.; Takeda-Kawaguchi, T.; Kuroda, I.; Hotta, Y.; Kawasaki, H.; Hariyama, T.; Shibata, T.; Akao, Y.; Kunisada, T.; Tatsumi, J.; et al. Exosomes from dental pulp cells attenuate bone loss in mouse experimental periodontitis. J. Periodontal Res. 2022, 57, 162–172. [Google Scholar] [CrossRef]
  69. Yan, C.; Li, N.; Xiao, T.; Ye, X.; Fu, L.; Ye, Y.; Xu, T.; Yu, J. 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. [Google Scholar] [CrossRef]
  70. Ma, L.; Rao, N.; Jiang, H.; Dai, Y.; Yang, S.; Yang, H.; Hu, J. Small extracellular vesicles from dental follicle stem cells provide biochemical cues for periodontal tissue regeneration. Stem Cell Res. Ther. 2022, 13, 92. [Google Scholar] [CrossRef]
  71. Shi, W.; Guo, S.; Liu, L.; Liu, Q.; Huo, F.; Ding, Y.; Tian, W. Small Extracellular Vesicles from Lipopolysaccharide-Preconditioned Dental Follicle Cells Promote Periodontal Regeneration in an Inflammatory Microenvironment. ACS Biomater. Sci. Eng. 2020, 6, 5797–5810. [Google Scholar] [CrossRef]
  72. Huang, Y.; Liu, Q.; Liu, L.; Huo, F.; Guo, S.; Tian, W. 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. Nanomed. 2022, 17, 799–819. [Google Scholar] [CrossRef]
  73. Xu, X.-Y.; Tian, B.-M.; Xia, Y.; Xia, Y.-L.; Li, X.; Zhou, H.; Tan, Y.-Z.; Chen, F.-M. Exosomes derived from P2X7 receptor gene-modified cells rescue inflammation-compromised periodontal ligament stem cells from dysfunction. Stem Cells Transl. Med. 2020, 9, 1414–1430. [Google Scholar] [CrossRef] [PubMed]
  74. Lv, P.-Y.; Gao, P.-F.; Tian, G.-J.; Yang, Y.-Y.; Mo, F.-F.; Wang, Z.-H.; Sun, L.; Kuang, M.-J.; Wang, Y.-L. 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. [Google Scholar] [CrossRef]
  75. Schlundt, C.; Fischer, H.; Bucher, C.H.; Rendenbach, C.; Duda, G.N.; Schmidt-Bleek, K. The multifaceted roles of macrophages in bone regeneration: A story of polarization, activation and time. Acta Biomater. 2021, 133, 46–57. [Google Scholar] [CrossRef] [PubMed]
  76. Blom, A.B.; van Lent, P.L.E.M.; Holthuysen, A.E.M.; van der Kraan, P.M.; Roth, J.; van Rooijen, N.; van den Berg, W.B. Synovial lining macrophages mediate osteophyte formation during experimental osteoarthritis. Osteoarthr. Cartil. 2004, 12, 627–635. [Google Scholar] [CrossRef] [Green Version]
  77. Yang, N.; Liu, Y. The Role of the Immune Microenvironment in Bone Regeneration. Int. J. Med. Sci. 2021, 18, 3697–3707. [Google Scholar] [CrossRef] [PubMed]
  78. Pajarinen, J.; Lin, T.; Gibon, E.; Kohno, Y.; Maruyama, M.; Nathan, K.; Lu, L.; Yao, Z.; Goodman, S.B. Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials 2019, 196, 80–89. [Google Scholar] [CrossRef]
  79. Mahon, O.R.; Browe, D.C.; Gonzalez-Fernandez, T.; Pitacco, P.; Whelan, I.T.; Von Euw, S.; Hobbs, C.; Nicolosi, V.; Cunningham, K.T.; Mills, K.H.G.; et al. Nano-particle mediated M2 macrophage polarization enhances bone formation and MSC osteogenesis in an IL-10 dependent manner. Biomaterials 2020, 239, 119833. [Google Scholar] [CrossRef]
  80. Xiong, Y.; Chen, L.; Yan, C.; Zhou, W.; Yu, T.; Sun, Y.; Cao, F.; Xue, H.; Hu, Y.; Chen, D.; et al. M2 Macrophagy-derived exosomal miRNA-5106 induces bone mesenchymal stem cells towards osteoblastic fate by targeting salt-inducible kinase 2 and 3. J. Nanobiotechnology 2020, 18, 66. [Google Scholar] [CrossRef]
  81. Xia, Y.; He, X.-T.; Xu, X.-Y.; Tian, B.-M.; An, Y.; Chen, F.-M. Exosomes derived from M0, M1 and M2 macrophages exert distinct influences on the proliferation and differentiation of mesenchymal stem cells. PeerJ 2020, 8, e8970. [Google Scholar] [CrossRef] [PubMed]
  82. Li, Z.; Wang, Y.; Li, S.; Li, Y. Exosomes Derived From M2 Macrophages Facilitate Osteogenesis and Reduce Adipogenesis of BMSCs. Front. Endocrinol. 2021, 12, 680328. [Google Scholar] [CrossRef]
  83. Bin-Bin, Z.; Da-Wa, Z.X.; Chao, L.; Lan-Tao, Z.; Tao, W.; Chuan, L.; Chao-Zheng, L.; De-Chun, L.; Chang, F.; Shu-Qing, W.; et al. M2 macrophagy-derived exosomal miRNA-26a-5p induces osteogenic differentiation of bone mesenchymal stem cells. J. Orthop. Surg. Res. 2022, 17, 137. [Google Scholar] [CrossRef] [PubMed]
  84. Liao, X.-M.; Guan, Z.; Yang, Z.-J.; Ma, L.-Y.; Dai, Y.-J.; Liang, C.; Hu, J.-T. Comprehensive analysis of M2 macrophage-derived exosomes facilitating osteogenic differentiation of human periodontal ligament stem cells. BMC Oral Health 2022, 22, 647. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, A.; Jin, S.; Fu, C.; Cui, S.; Zhang, T.; Zhu, L.; Wang, Y.; Shen, S.G.F.; Jiang, N.; Liu, Y. Macrophage-derived small extracellular vesicles promote biomimetic mineralized collagen-mediated endogenous bone regeneration. Int. J. Oral Sci. 2020, 12, 33. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, Z.; Zhao, F.; Zhao, Y.; Bai, L.; Hang, R. Simultaneously enhanced osteogenesis and angiogenesis via macrophage-derived exosomes upon stimulation with titania nanotubes. Biomater. Adv. 2022, 134, 112708. [Google Scholar] [CrossRef]
  87. Zhu, Y.; Zhao, S.; Cheng, L.; Lin, Z.; Zeng, M.; Ruan, Z.; Sun, B.; Luo, Z.; Tang, Y.; Long, H. Mg2+ -mediated autophagy-dependent polarization of macrophages mediates the osteogenesis of bone marrow stromal stem cells by interfering with macrophage-derived exosomes containing miR-381. J. Orthop. Res. 2022, 40, 1563–1576. [Google Scholar] [CrossRef]
  88. Ekström, K.; Omar, O.; Granéli, C.; Wang, X.; Vazirisani, F.; Thomsen, P. Monocyte exosomes stimulate the osteogenic gene expression of mesenchymal stem cells. PLoS ONE 2013, 8, e75227. [Google Scholar] [CrossRef] [Green Version]
  89. Hao, Z.; Ren, L.; Zhang, Z.; Yang, Z.; Wu, S.; Liu, G.; Cheng, B.; Wu, J.; Xia, J. A multifunctional neuromodulation platform utilizing Schwann cell-derived exosomes orchestrates bone microenvironment via immunomodulation, angiogenesis and osteogenesis. Bioact. Mater. 2023, 23, 206–222. [Google Scholar] [CrossRef]
  90. Ren, L.; Zeng, F.; Deng, J.; Bai, Y.; Chen, K.; Chen, L.; Sun, L. Inflammatory osteoclasts-derived exosomes promote bone formation by selectively transferring lncRNA LIOCE into osteoblasts to interact with and stabilize Osterix. FASEB J. 2022, 36, e22115. [Google Scholar] [CrossRef]
  91. Liu, Y.; Wang, L.; Kikuiri, T.; Akiyama, K.; Chen, C.; Xu, X.; Yang, R.; Chen, W.; Wang, S.; Shi, S. Mesenchymal stem cell-based tissue regeneration is governed by recipient T lymphocytes via IFN-γ and TNF-α. Nat. Med. 2011, 17, 1594–1601. [Google Scholar] [CrossRef] [PubMed]
  92. Asemani, Y.; Najafi, S.; Ezzatifar, F.; Zolbanin, N.M.; Jafari, R. Recent highlights in the immunomodulatory aspects of Treg cell-derived extracellular vesicles: Special emphasis on autoimmune diseases and transplantation. Cell Biosci. 2022, 12, 67. [Google Scholar] [CrossRef] [PubMed]
  93. Hu, H.; Wu, J.; Cao, C.; Ma, L. Exosomes derived from regulatory T cells ameliorate acute myocardial infarction by promoting macrophage M2 polarization. IUBMB Life 2020, 72, 2409–2419. [Google Scholar] [CrossRef]
  94. Salmon, C.R.; Tomazela, D.M.; Ruiz, K.G.S.; Foster, B.L.; Paes Leme, A.F.; Sallum, E.A.; Somerman, M.J.; Nociti, F.H. Proteomic analysis of human dental cementum and alveolar bone. J. Proteom. 2013, 91, 544–555. [Google Scholar] [CrossRef] [Green Version]
  95. Huang, X.; Xiao, J.; Wang, X.; Cao, Z. Irisin attenuates P. gingivalis-suppressed osteogenic/cementogenic differentiation of periodontal ligament cells via p38 signaling pathway. Biochem. Biophys. Res. Commun. 2022, 618, 100–106. [Google Scholar] [CrossRef]
  96. Liu, J.; Dai, Q.; Weir, M.D.; Schneider, A.; Zhang, C.; Hack, G.D.; Oates, T.W.; Zhang, K.; Li, A.; Xu, H.H.K. Biocompatible Nanocomposite Enhanced Osteogenic and Cementogenic Differentiation of Periodontal Ligament Stem Cells In Vitro for Periodontal Regeneration. Materials 2020, 13, 4951. [Google Scholar] [CrossRef]
  97. Li, S.; Shao, J.; Zhou, Y.; Friis, T.; Yao, J.; Shi, B.; Xiao, Y. The impact of Wnt signalling and hypoxia on osteogenic and cementogenic differentiation in human periodontal ligament cells. Mol. Med. Rep. 2016, 14, 4975–4982. [Google Scholar] [CrossRef] [Green Version]
  98. Jin, H.; Xu, Y.; Qi, Y.; Wang, X.; Patel, D.K.; Deb Dutta, S.; Chen, R.; Lim, K.-T. Evaluation of Osteogenic/Cementogenic Modulating Potential of PAI-1 Transfected Media for Stem Cells. IEEE Trans. Nanobiosci. 2020, 19, 446–456. [Google Scholar] [CrossRef] [PubMed]
  99. Paknejad, M.; Eslaminejad, M.B.; Ghaedi, B.; Rokn, A.-R.; Khorsand, A.; Etemad-Moghadam, S.; Alaeddini, M.; Dehghan, M.M.; Moslemi, N.; Nowzari, H. Isolation and Assessment of Mesenchymal Stem Cells Derived From Bone Marrow: Histologic and Histomorphometric Study in a Canine Periodontal Defect. J. Oral Implantol. 2015, 41, 284–291. [Google Scholar] [CrossRef]
  100. Sun, Q.; Zhao, T.; Li, B.; Li, M.; Luo, P.; Zhang, C.; Chen, G.; Cao, Z.; Li, Y.; Du, M.; et al. FTO/RUNX2 signaling axis promotes cementoblast differentiation under normal and inflammatory condition. Biochim. Biophys. Acta Mol. Cell Res. 2022, 1869, 119358. [Google Scholar] [CrossRef]
  101. Wu, Z.; He, Y.; Chen, S.; Zhu, L.; Wang, J.; Zhang, D.; Xie, J.; Zou, S.; Zhou, C. Connective tissue growth factor promotes cementogenesis and cementum repair via Cx43/β-catenin axis. Stem Cell Res. Ther. 2022, 13, 460. [Google Scholar] [CrossRef]
  102. Cao, H.-L.; Chung, J.H.; Choung, P.-H. Allogeneic Fibrin Clot for Odontogenic/Cementogenic Differentiation of Human Dental Mesenchymal Stem Cells. Tissue Eng. Regen. Med. 2020, 17, 511–524. [Google Scholar] [CrossRef]
  103. Xie, X.; Xu, C.; Zhao, H.; Wang, J.; Feng, J.Q. A Biphasic Feature of Gli1+-Mesenchymal Progenitors during Cementogenesis That Is Positively Controlled by Wnt/β-Catenin Signaling. J. Dent. Res. 2021, 100, 1289–1298. [Google Scholar] [CrossRef]
  104. Han, P.; Xu, M.; Chang, J.; Chakravorty, N.; Wu, C.; Xiao, Y. Lithium release from β-tricalcium phosphate inducing cementogenic and osteogenic differentiation of both hPDLCs and hBMSCs. Biomater. Sci. 2014, 2, 1230–1243. [Google Scholar] [CrossRef] [PubMed]
  105. Huang, X.; Wang, X.; Ma, L.; Wang, H.; Peng, Y.; Liu, H.; Xiao, J.; Cao, Z. M2 macrophages with inflammation tropism facilitate cementoblast mineralization. J. Periodontol. 2022, 94, 290–300. [Google Scholar] [CrossRef] [PubMed]
  106. Zhao, Y.; Huang, Y.; Liu, H.; Tan, K.; Wang, R.; Jia, L.; Li, W. Macrophages with Different Polarization Phenotypes Influence Cementoblast Mineralization through Exosomes. Stem Cells Int. 2022, 2022, 4185972. [Google Scholar] [CrossRef]
  107. Rather, H.A.; Jhala, D.; Vasita, R. Dual functional approaches for osteogenesis coupled angiogenesis in bone tissue engineering. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 103, 109761. [Google Scholar] [CrossRef]
  108. Pandula, P.K.C.P.; Samaranayake, L.P.; Jin, L.J.; Zhang, C.F. Human umbilical vein endothelial cells synergize osteo/odontogenic differentiation of periodontal ligament stem cells in 3D cell sheets. J. Periodontal. Res. 2014, 49, 299–306. [Google Scholar] [CrossRef]
  109. Gurav, A.N. The implication of periodontitis in vascular endothelial dysfunction. Eur. J. Clin. Invest. 2014, 44, 1000–1009. [Google Scholar] [CrossRef] [Green Version]
  110. Zoellner, H.; Hunter, N. Vascular expansion in chronic periodontitis. J. Oral Pathol. Med. 1991, 20, 433–437. [Google Scholar] [CrossRef] [PubMed]
  111. Zoellner, H.; Chapple, C.C.; Hunter, N. Microvasculature in gingivitis and chronic periodontitis: Disruption of vascular networks with protracted inflammation. Microsc. Res. Tech. 2002, 56, 15–31. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, Z.; Shuai, Y.; Zhou, F.; Yin, J.; Hu, J.; Guo, S.; Wang, Y.; Liu, W. PDLSCs Regulate Angiogenesis of Periodontal Ligaments via VEGF Transferred by Exosomes in Periodontitis. Int. J. Med. Sci. 2020, 17, 558–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhou, H.; Li, X.; Wu, R.-X.; He, X.-T.; An, Y.; Xu, X.-Y.; Sun, H.-H.; Wu, L.-A.; Chen, F.-M. 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. [Google Scholar] [CrossRef]
  114. Xia, Y.; Ling, X.; Hu, G.; Zhu, Q.; Zhang, J.; Li, Q.; Zhao, B.; Wang, Y.; Deng, Z. Small extracellular vesicles secreted by human iPSC-derived MSC enhance angiogenesis through inhibiting STAT3-dependent autophagy in ischemic stroke. Stem Cell Res. Ther. 2020, 11, 313. [Google Scholar] [CrossRef]
  115. Cheng, P.; Cao, T.; Zhao, X.; Lu, W.; Miao, S.; Ning, F.; Wang, D.; Gao, Y.; Wang, L.; Pei, G.; et al. Nidogen1-enriched extracellular vesicles accelerate angiogenesis and bone regeneration by targeting Myosin-10 to regulate endothelial cell adhesion. Bioact. Mater. 2022, 12, 185–197. [Google Scholar] [CrossRef]
  116. Pizzicannella, J.; Gugliandolo, A.; Orsini, T.; Fontana, A.; Ventrella, A.; Mazzon, E.; Bramanti, P.; Diomede, F.; Trubiani, O. Engineered Extracellular Vesicles From Human Periodontal-Ligament Stem Cells Increase VEGF/VEGFR2 Expression During Bone Regeneration. Front. Physiol. 2019, 10, 512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Pizzicannella, J.; Diomede, F.; Gugliandolo, A.; Chiricosta, L.; Bramanti, P.; Merciaro, I.; Orsini, T.; Mazzon, E.; Trubiani, O. 3D Printing PLA/Gingival Stem Cells/ EVs Upregulate miR-2861 and -210 during Osteoangiogenesis Commitment. Int. J. Mol. Sci. 2019, 20, 3256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Zhang, L.; Jiao, G.; Ren, S.; Zhang, X.; Li, C.; Wu, W.; Wang, H.; Liu, H.; Zhou, H.; Chen, Y. Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion. Stem Cell Res. Ther. 2020, 11, 38. [Google Scholar] [CrossRef]
  119. Huang, Y.; He, B.; Wang, L.; Yuan, B.; Shu, H.; Zhang, F.; Sun, L. Bone marrow mesenchymal stem cell-derived exosomes promote rotator cuff tendon-bone healing by promoting angiogenesis and regulating M1 macrophages in rats. Stem Cell Res. Ther. 2020, 11, 496. [Google Scholar] [CrossRef]
  120. Zhang, Y.; Hao, Z.; Wang, P.; Xia, Y.; Wu, J.; Xia, D.; Fang, S.; Xu, S. Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF-1α-mediated promotion of angiogenesis in a rat model of stabilized fracture. Cell Prolif. 2019, 52, e12570. [Google Scholar] [CrossRef] [Green Version]
  121. Nan, K.; Zhang, Y.; Zhang, X.; Li, D.; Zhao, Y.; Jing, Z.; Liu, K.; Shang, D.; Geng, Z.; Fan, L. Exosomes from miRNA-378-modified adipose-derived stem cells prevent glucocorticoid-induced osteonecrosis of the femoral head by enhancing angiogenesis and osteogenesis via targeting miR-378 negatively regulated suppressor of fused (Sufu). Stem Cell Res. Ther. 2021, 12, 331. [Google Scholar] [CrossRef]
  122. Kong, F.; Wu, C.-T.; Geng, P.; Liu, C.; Xiao, F.; Wang, L.-S.; Wang, H. Dental Pulp Stem Cell-Derived Extracellular Vesicles Mitigate Haematopoietic Damage after Radiation. Stem Cell Rev. Rep. 2021, 17, 318–331. [Google Scholar] [CrossRef] [PubMed]
  123. Aharon, A.; Tamari, T.; Brenner, B. Monocyte-derived microparticles and exosomes induce procoagulant and apoptotic effects on endothelial cells. Thromb. Haemost. 2008, 100, 878–885. [Google Scholar]
  124. Wang, Y.; Zheng, Y.; Li, W. Compression loading of osteoclasts attenuated microRNA-146a-5p expression, which promotes angiogenesis by targeting adiponectin. Sci. China Life Sci. 2022, 65, 151–166. [Google Scholar] [CrossRef] [PubMed]
  125. Cheng, X.; Zhou, H.; Zhou, Y.; Song, C. M2 Macrophage-Derived Exosomes Inhibit Apoptosis of HUVEC Cell through Regulating miR-221-3p Expression. Biomed. Res. Int. 2022, 2022, 1609244. [Google Scholar] [CrossRef] [PubMed]
  126. Lyu, L.; Cai, Y.; Zhang, G.; Jing, Z.; Liang, J.; Zhang, R.; Dang, X.; Zhang, C. Exosomes derived from M2 macrophages induce angiogenesis to promote wound healing. Front. Mol. Biosci. 2022, 9, 1008802. [Google Scholar] [CrossRef]
  127. Huang, Y.; Liu, L.; Liu, Q.; Huo, F.; Hu, X.; Guo, S.; Tian, W. Dental follicle cells-derived small extracellular vesicles inhibit pathogenicity of Porphyromonas gingivalis. Oral Dis. 2022. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, Y.; Zhuang, X.; Yu, S.; Yang, N.; Zeng, J.; Liu, X.; Chen, X. Exosomes derived from stem cells from apical papilla promote craniofacial soft tissue regeneration by enhancing Cdc42-mediated vascularization. Stem Cell Res. Ther. 2021, 12, 76. [Google Scholar] [CrossRef]
  129. Toyofuku, M.; Nomura, N.; Eberl, L. Types and origins of bacterial membrane vesicles. Nat. Rev. Microbiol. 2019, 17, 13–24. [Google Scholar] [CrossRef]
  130. Sartorio, M.G.; Pardue, E.J.; Feldman, M.F.; Haurat, M.F. Bacterial Outer Membrane Vesicles: From Discovery to Applications. Annu. Rev. Microbiol. 2021, 75, 609–630. [Google Scholar] [CrossRef]
  131. Shen, H.; Aggarwal, N.; Wun, K.S.; Lee, Y.S.; Hwang, I.Y.; Chang, M.W. Engineered microbial systems for advanced drug delivery. Adv. Drug. Deliv. Rev. 2022, 187, 114364. [Google Scholar] [CrossRef] [PubMed]
  132. Schwechheimer, C.; Kuehn, M.J. Outer-membrane vesicles from Gram-negative bacteria: Biogenesis and functions. Nat. Rev. Microbiol. 2015, 13, 605–619. [Google Scholar] [CrossRef] [Green Version]
  133. McBroom, A.J.; Kuehn, M.J. Release of outer membrane vesicles by Gram-negative bacteria is a novel envelope stress response. Mol. Microbiol. 2007, 63, 545–558. [Google Scholar] [CrossRef]
  134. Toledo, A.; Coleman, J.L.; Kuhlow, C.J.; Crowley, J.T.; Benach, J.L. The enolase of Borrelia burgdorferi is a plasminogen receptor released in outer membrane vesicles. Infect. Immun. 2012, 80, 359–368. [Google Scholar] [CrossRef] [Green Version]
  135. Rakoff-Nahoum, S.; Coyne, M.J.; Comstock, L.E. An ecological network of polysaccharide utilization among human intestinal symbionts. Curr. Biol. 2014, 24, 40–49. [Google Scholar] [CrossRef] [Green Version]
  136. Fulsundar, S.; Harms, K.; Flaten, G.E.; Johnsen, P.J.; Chopade, B.A.; Nielsen, K.M. Gene transfer potential of outer membrane vesicles of Acinetobacter baylyi and effects of stress on vesiculation. Appl. Environ. Microbiol. 2014, 80, 3469–3483. [Google Scholar] [CrossRef] [Green Version]
  137. Ellis, T.N.; Kuehn, M.J. Virulence and immunomodulatory roles of bacterial outer membrane vesicles. Microbiol. Mol. Biol. Rev. 2010, 74, 81–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Kaparakis-Liaskos, M.; Ferrero, R.L. Immune modulation by bacterial outer membrane vesicles. Nat. Rev. Immunol. 2015, 15, 375–387. [Google Scholar] [CrossRef]
  139. Kaparakis, M.; Turnbull, L.; Carneiro, L.; Firth, S.; Coleman, H.A.; Parkington, H.C.; Le Bourhis, L.; Karrar, A.; Viala, J.; Mak, J.; et al. Bacterial membrane vesicles deliver peptidoglycan to NOD1 in epithelial cells. Cell Microbiol. 2010, 12, 372–385. [Google Scholar] [CrossRef] [Green Version]
  140. Vidakovics, M.L.A.P.; Jendholm, J.; Mörgelin, M.; Månsson, A.; Larsson, C.; Cardell, L.-O.; Riesbeck, K. B cell activation by outer membrane vesicles—A novel virulence mechanism. PLoS Pathog. 2010, 6, e1000724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Alaniz, R.C.; Deatherage, B.L.; Lara, J.C.; Cookson, B.T. Membrane vesicles are immunogenic facsimiles of Salmonella typhimurium that potently activate dendritic cells, prime B and T cell responses, and stimulate protective immunity in vivo. J. Immunol. 2007, 179, 7692–7701. [Google Scholar] [CrossRef] [Green Version]
  142. Holt, S.C.; Ebersole, J.L. Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia: The “red complex”, a prototype polybacterial pathogenic consortium in periodontitis. Periodontol 2000 2005, 38, 72–122. [Google Scholar] [CrossRef] [PubMed]
  143. Diaz, P.I.; Hoare, A.; Hong, B.-Y. Subgingival Microbiome Shifts and Community Dynamics in Periodontal Diseases. J. Calif. Dent. Assoc. 2016, 44, 421–435. [Google Scholar] [CrossRef] [PubMed]
  144. Han, P.; Bartold, P.M.; Salomon, C.; Ivanovski, S. Salivary Outer Membrane Vesicles and DNA Methylation of Small Extracellular Vesicles as Biomarkers for Periodontal Status: A Pilot Study. Int. J. Mol. Sci. 2021, 22, 2423. [Google Scholar] [CrossRef] [PubMed]
  145. Ma, L.; Cao, Z. Membrane vesicles from periodontal pathogens and their potential roles in periodontal disease and systemic illnesses. J. Periodontal. Res. 2021, 56, 646–655. [Google Scholar] [CrossRef]
  146. Choi, J.W.; Kim, S.C.; Hong, S.H.; Lee, H.J. Secretable Small RNAs via Outer Membrane Vesicles in Periodontal Pathogens. J. Dent. Res. 2017, 96, 458–466. [Google Scholar] [CrossRef]
  147. Fan, R.; Zhou, Y.; Chen, X.; Zhong, X.; He, F.; Peng, W.; Li, L.; Wang, X.; Xu, Y. Porphyromonas gingivalis Outer Membrane Vesicles Promote Apoptosis via msRNA-Regulated DNA Methylation in Periodontitis. Microbiol. Spectr. 2023, 11, e0328822. [Google Scholar] [CrossRef]
  148. Lim, Y.; Kim, H.Y.; An, S.-J.; Choi, B.-K. Activation of bone marrow-derived dendritic cells and CD4+ T cell differentiation by outer membrane vesicles of periodontal pathogens. J. Oral Microbiol. 2022, 14, 2123550. [Google Scholar] [CrossRef]
  149. 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. [Google Scholar] [CrossRef]
  150. Zhang, Z.; Liu, S.; Zhang, S.; Li, Y.; Shi, X.; Liu, D.; Pan, Y. Porphyromonas gingivalis outer membrane vesicles inhibit the invasion of Fusobacterium nucleatum into oral epithelial cells by downregulating FadA and FomA. J. Periodontol. 2022, 93, 515–525. [Google Scholar] [CrossRef] [PubMed]
  151. Nakao, R.; Hasegawa, H.; Dongying, B.; Ohnishi, M.; Senpuku, H. Assessment of outer membrane vesicles of periodontopathic bacterium Porphyromonas gingivalis as possible mucosal immunogen. Vaccine 2016, 34, 4626–4634. [Google Scholar] [CrossRef]
  152. Nakao, R.; Hasegawa, H.; Ochiai, K.; Takashiba, S.; Ainai, A.; Ohnishi, M.; Watanabe, H.; Senpuku, H. Outer membrane vesicles of Porphyromonas gingivalis elicit a mucosal immune response. PLoS ONE 2011, 6, e26163. [Google Scholar] [CrossRef] [Green Version]
  153. Gerritzen, M.J.H.; Martens, D.E.; Wijffels, R.H.; van der Pol, L.; Stork, M. Bioengineering bacterial outer membrane vesicles as vaccine platform. Biotechnol. Adv. 2017, 35, 565–574. [Google Scholar] [CrossRef] [PubMed]
  154. Mehanny, M.; Lehr, C.-M.; Fuhrmann, G. Extracellular vesicles as antigen carriers for novel vaccination avenues. Adv. Drug Deliv. Rev. 2021, 173, 164–180. [Google Scholar] [CrossRef]
  155. Ardila, C.M.; Granada, M.I.; Guzmán, I.C. Antibiotic resistance of subgingival species in chronic periodontitis patients. J. Periodontal. Res. 2010, 45, 557–563. [Google Scholar] [CrossRef]
  156. Huang, W.; Zhang, Q.; Li, W.; Yuan, M.; Zhou, J.; Hua, L.; Chen, Y.; Ye, C.; Ma, Y. Development of novel nanoantibiotics using an outer membrane vesicle-based drug efflux mechanism. J. Control. Release 2020, 317, 1–22. [Google Scholar] [CrossRef]
  157. Bittel, M.; Reichert, P.; Sarfati, I.; Dressel, A.; Leikam, S.; Uderhardt, S.; Stolzer, I.; Phu, T.A.; Ng, M.; Vu, N.K.; et al. Visualizing transfer of microbial biomolecules by outer membrane vesicles in microbe-host-communication in vivo. J. Extracell. Vesicles 2021, 10, e12159. [Google Scholar] [CrossRef]
  158. Pinedo, M.; de la Canal, L.; de Marcos Lousa, C. A call for Rigor and standardization in plant extracellular vesicle research. J. Extracell. Vesicles 2021, 10, e12048. [Google Scholar] [CrossRef] [PubMed]
  159. Cong, M.; Tan, S.; Li, S.; Gao, L.; Huang, L.; Zhang, H.-G.; Qiao, H. Technology insight: Plant-derived vesicles-How far from the clinical biotherapeutics and therapeutic drug carriers? Adv. Drug Deliv. Rev. 2022, 182, 114108. [Google Scholar] [CrossRef] [PubMed]
  160. Halperin, W.; Jensen, W.A. Ultrastructural changes during growth and embryogenesis in carrot cell cultures. J. Ultrastruct. Res. 1967, 18, 428–443. [Google Scholar] [CrossRef]
  161. Movahed, N.; Cabanillas, D.G.; Wan, J.; Vali, H.; Laliberté, J.-F.; Zheng, H. Turnip Mosaic Virus Components Are Released into the Extracellular Space by Vesicles in Infected Leaves. Plant. Physiol. 2019, 180, 1375–1388. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, J.; Ding, Y.; Wang, J.; Hillmer, S.; Miao, Y.; Lo, S.W.; Wang, X.; Robinson, D.G.; Jiang, L. EXPO, an exocyst-positive organelle distinct from multivesicular endosomes and autophagosomes, mediates cytosol to cell wall exocytosis in Arabidopsis and tobacco cells. Plant Cell 2010, 22, 4009–4030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Hatsugai, N.; Iwasaki, S.; Tamura, K.; Kondo, M.; Fuji, K.; Ogasawara, K.; Nishimura, M.; Hara-Nishimura, I. A novel membrane fusion-mediated plant immunity against bacterial pathogens. Genes Dev. 2009, 23, 2496–2506. [Google Scholar] [CrossRef] [Green Version]
  164. Cui, Y.; Cao, W.; He, Y.; Zhao, Q.; Wakazaki, M.; Zhuang, X.; Gao, J.; Zeng, Y.; Gao, C.; Ding, Y.; et al. A whole-cell electron tomography model of vacuole biogenesis in Arabidopsis root cells. Nat. Plants 2019, 5, 95–105. [Google Scholar] [CrossRef]
  165. Bruns, C.; McCaffery, J.M.; Curwin, A.J.; Duran, J.M.; Malhotra, V. Biogenesis of a novel compartment for autophagosome-mediated unconventional protein secretion. J. Cell Biol. 2011, 195, 979–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Urzì, O.; Raimondo, S.; Alessandro, R. Extracellular Vesicles from Plants: Current Knowledge and Open Questions. Int. J. Mol. Sci. 2021, 22, 5366. [Google Scholar] [CrossRef] [PubMed]
  167. Feng, J.; Xiu, Q.; Huang, Y.; Troyer, Z.; Li, B.; Zheng, L. Plant Derived Vesicle-Like Nanoparticles as Promising Biotherapeutic Tools: Present and Future. Adv. Mater. 2023, e2207826. [Google Scholar] [CrossRef]
  168. Fan, S.-J.; Chen, J.-Y.; Tang, C.-H.; Zhao, Q.-Y.; Zhang, J.-M.; Qin, Y.-C. Edible plant extracellular vesicles: An emerging tool for bioactives delivery. Front. Immunol. 2022, 13, 1028418. [Google Scholar] [CrossRef]
  169. Zhang, Z.; Yu, Y.; Zhu, G.; Zeng, L.; Xu, S.; Cheng, H.; Ouyang, Z.; Chen, J.; Pathak, J.L.; Wu, L.; et al. The Emerging Role of Plant-Derived Exosomes-Like Nanoparticles in Immune Regulation and Periodontitis Treatment. Front. Immunol. 2022, 13, 896745. [Google Scholar] [CrossRef]
  170. Mu, J.; Zhuang, X.; Wang, Q.; Jiang, H.; Deng, Z.-B.; Wang, B.; Zhang, L.; Kakar, S.; Jun, Y.; Miller, D.; et al. Interspecies communication between plant and mouse gut host cells through edible plant derived exosome-like nanoparticles. Mol. Nutr. Food Res. 2014, 58, 1561–1573. [Google Scholar] [CrossRef] [Green Version]
  171. Zu, M.; Xie, D.; Canup, B.S.B.; Chen, N.; Wang, Y.; Sun, R.; Zhang, Z.; Fu, Y.; Dai, F.; Xiao, B. ‘Green’ nanotherapeutics from tea leaves for orally targeted prevention and alleviation of colon diseases. Biomaterials 2021, 279, 121178. [Google Scholar] [CrossRef]
  172. Chen, X.; Zhou, Y.; Yu, J. Exosome-like Nanoparticles from Ginger Rhizomes Inhibited NLRP3 Inflammasome Activation. Mol. Pharm. 2019, 16, 2690–2699. [Google Scholar] [CrossRef] [PubMed]
  173. Liu, B.; Lu, Y.; Chen, X.; Muthuraj, P.G.; Li, X.; Pattabiraman, M.; Zempleni, J.; Kachman, S.D.; Natarajan, S.K.; Yu, J. Protective Role of Shiitake Mushroom-Derived Exosome-Like Nanoparticles in D-Galactosamine and Lipopolysaccharide-Induced Acute Liver Injury in Mice. Nutrients 2020, 12, 477. [Google Scholar] [CrossRef] [Green Version]
  174. You, J.Y.; Kang, S.J.; Rhee, W.J. Isolation of cabbage exosome-like nanovesicles and investigation of their biological activities in human cells. Bioact. Mater. 2021, 6, 4321–4332. [Google Scholar] [CrossRef] [PubMed]
  175. Kim, M.; Park, J.H. Isolation of Aloe saponaria-Derived Extracellular Vesicles and Investigation of Their Potential for Chronic Wound Healing. Pharmaceutics 2022, 14, 1905. [Google Scholar] [CrossRef]
  176. Savcı, Y.; Kırbaş, O.K.; Bozkurt, B.T.; Abdik, E.A.; Taşlı, P.N.; Şahin, F.; Abdik, H. Grapefruit-derived extracellular vesicles as a promising cell-free therapeutic tool for wound healing. Food Funct. 2021, 12, 5144–5156. [Google Scholar] [CrossRef] [PubMed]
  177. Kim, M.K.; Choi, Y.C.; Cho, S.H.; Choi, J.S.; Cho, Y.W. The Antioxidant Effect of Small Extracellular Vesicles Derived from Aloe vera Peels for Wound Healing. Tissue Eng. Regen. Med. 2021, 18, 561–571. [Google Scholar] [CrossRef]
  178. Aquilano, K.; Ceci, V.; Gismondi, A.; De Stefano, S.; Iacovelli, F.; Faraonio, R.; Di Marco, G.; Poerio, N.; Minutolo, A.; Minopoli, G.; et al. Adipocyte metabolism is improved by TNF receptor-targeting small RNAs identified from dried nuts. Commun. Biol. 2019, 2, 317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Deng, Z.; Rong, Y.; Teng, Y.; Mu, J.; Zhuang, X.; Tseng, M.; Samykutty, A.; Zhang, L.; Yan, J.; Miller, D.; et al. Broccoli-Derived Nanoparticle Inhibits Mouse Colitis by Activating Dendritic Cell AMP-Activated Protein Kinase. Mol. Ther. 2017, 25, 1641–1654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Sriwastva, M.K.; Deng, Z.-B.; Wang, B.; Teng, Y.; Kumar, A.; Sundaram, K.; Mu, J.; Lei, C.; Dryden, G.W.; Xu, F.; et al. Exosome-like nanoparticles from Mulberry bark prevent DSS-induced colitis via the AhR/COPS8 pathway. EMBO Rep. 2022, 23, e53365. [Google Scholar] [CrossRef]
  181. Paiotti, A.P.R.; Neto, R.A.; Marchi, P.; Silva, R.M.; Pazine, V.L.; Noguti, J.; Pastrelo, M.M.; Gollücke, A.P.B.; Miszputen, S.J.; Ribeiro, D.A. The anti-inflammatory potential of phenolic compounds in grape juice concentrate (G8000™) on 2,4,6-trinitrobenzene sulphonic acid-induced colitis. Br. J. Nutr. 2013, 110, 973–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Teng, Y.; Xu, F.; Zhang, X.; Mu, J.; Sayed, M.; Hu, X.; Lei, C.; Sriwastva, M.; Kumar, A.; Sundaram, K.; et al. Plant-derived exosomal microRNAs inhibit lung inflammation induced by exosomes SARS-CoV-2 Nsp12. Mol. Ther. 2021, 29, 2424–2440. [Google Scholar] [CrossRef] [PubMed]
  183. Ju, S.; Mu, J.; Dokland, T.; Zhuang, X.; Wang, Q.; Jiang, H.; Xiang, X.; Deng, Z.-B.; Wang, B.; Zhang, L.; et al. Grape exosome-like nanoparticles induce intestinal stem cells and protect mice from DSS-induced colitis. Mol. Ther. 2013, 21, 1345–1357. [Google Scholar] [CrossRef] [Green Version]
  184. Sundaram, K.; Miller, D.P.; Kumar, A.; Teng, Y.; Sayed, M.; Mu, J.; Lei, C.; Sriwastva, M.K.; Zhang, L.; Yan, J.; et al. Plant-Derived Exosomal Nanoparticles Inhibit Pathogenicity of Porphyromonas gingivalis. iScience 2019, 21, 308–327. [Google Scholar] [CrossRef] [Green Version]
  185. Cui, Y.; Gao, J.; He, Y.; Jiang, L. Plant extracellular vesicles. Protoplasma 2020, 257, 3–12. [Google Scholar] [CrossRef] [PubMed]
  186. Lei, C.; Teng, Y.; He, L.; Sayed, M.; Mu, J.; Xu, F.; Zhang, X.; Kumar, A.; Sundaram, K.; Sriwastva, M.K.; et al. Lemon exosome-like nanoparticles enhance stress survival of gut bacteria by RNase P-mediated specific tRNA decay. iScience 2021, 24, 102511. [Google Scholar] [CrossRef] [PubMed]
  187. Huang, J.; Xiong, J.; Yang, L.; Zhang, J.; Sun, S.; Liang, Y. Cell-free exosome-laden scaffolds for tissue repair. Nanoscale 2021, 13, 8740–8750. [Google Scholar] [CrossRef]
  188. Şahin, F.; Koçak, P.; Güneş, M.Y.; Özkan, İ.; Yıldırım, E.; Kala, E.Y. In Vitro Wound Healing Activity of Wheat-Derived Nanovesicles. Appl. Biochem. Biotechnol. 2019, 188, 381–394. [Google Scholar] [CrossRef] [PubMed]
  189. Perut, F.; Roncuzzi, L.; Avnet, S.; Massa, A.; Zini, N.; Sabbadini, S.; Giampieri, F.; Mezzetti, B.; Baldini, N. Strawberry-Derived Exosome-Like Nanoparticles Prevent Oxidative Stress in Human Mesenchymal Stromal Cells. Biomolecules 2021, 11, 87. [Google Scholar] [CrossRef]
  190. Xu, X.-H.; Yuan, T.-J.; Dad, H.A.; Shi, M.-Y.; Huang, Y.-Y.; Jiang, Z.-H.; Peng, L.-H. Plant Exosomes As Novel Nanoplatforms for MicroRNA Transfer Stimulate Neural Differentiation of Stem Cells In Vitro and In Vivo. Nano Lett. 2021, 21, 8151–8159. [Google Scholar] [CrossRef]
  191. Hwang, J.-H.; Park, Y.-S.; Kim, H.-S.; Kim, D.-H.; Lee, S.-H.; Lee, C.-H.; Lee, S.-H.; Kim, J.-E.; Lee, S.; Kim, H.M.; et al. Yam-derived exosome-like nanovesicles stimulate osteoblast formation and prevent osteoporosis in mice. J. Control. Release 2023, 355, 184–198. [Google Scholar] [CrossRef] [PubMed]
  192. Woith, E.; Fuhrmann, G.; Melzig, M.F. Extracellular Vesicles-Connecting Kingdoms. Int. J. Mol. Sci. 2019, 20, 5695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Li, M.; Xing, X.; Huang, H.; Liang, C.; Gao, X.; Tang, Q.; Xu, X.; Yang, J.; Liao, L.; Tian, W. BMSC-Derived ApoEVs Promote Craniofacial Bone Repair via ROS/JNK Signaling. J. Dent. Res. 2022, 101, 714–723. [Google Scholar] [CrossRef] [PubMed]
  194. Iwayama, T.; Bhongsatiern, P.; Takedachi, M.; Murakami, S. Matrix Vesicle-Mediated Mineralization and Potential Applications. J. Dent. Res. 2022, 101, 1554–1562. [Google Scholar] [CrossRef]
Figure 1. CEVs, BEVs and PEVs with versatile functions for periodontal regeneration. CEVs, eukaryocyte-derived extracellular vesicles; BEVs, bacterial-derived extracellular vesicles; PEVs, plant-derived extracellular vesicles.
Figure 1. CEVs, BEVs and PEVs with versatile functions for periodontal regeneration. CEVs, eukaryocyte-derived extracellular vesicles; BEVs, bacterial-derived extracellular vesicles; PEVs, plant-derived extracellular vesicles.
Ijms 24 05790 g001
Table 1. Application of SCEVs and ICEVs in periodontal regeneration.
Table 1. Application of SCEVs and ICEVs in periodontal regeneration.
TypeRegeneration FunctionsMechanismsScaffold/Amount of EVsLocation/Sacrifice TimeRef.
MSC-EVsImprove proliferation and migration of PDLCs, promote new bone formation in rat periodontal defectsActivate the Akt, ERK pathways via CD73collagen sponge;
40 µg
Mesial side of the upper 1st molar;
2 w, 4 w
[63]
ADSC-EVsPromote PDL repair and osteoid tissue formation in rats with periodontitis/PBS suspension;
80–150 µg
The lower incisors;
2 d, 2 w, 4 w
[62]
BMMSC-EVsImprove proliferation, migration, osteogenic differentiation of PDLCs, promote bone regeneration in rats with periodontitisPromote M2 macrophage polarization and TGF-β1 expressionNanocomposite hydrogel;
1 time/week
Right upper 2nd molar;
4 w, 8 w
[67]
SHED-EVsImprove proliferation, osteogenesis of BMMSCs, promote PDL remodeling and bone regeneration in mice with periodontitisReduce inflammationPBS suspension;
20 µg/week
The upper 1st molar;
2 w
[46]
SHED-EVsImprove the functions of HUVECs, promote osteogenesis of BMMSCs, promote angiogenesis and bone formation in rat periodontal defectsActivate the AMPK pathwayβ-tricalcium phosphate;
100 µg
Buccal side from the lower 1st molar to 3rd molar;
4 w
[65]
DFSC-EVsImprove proliferation, migration, osteogenic differentiation of PDLSCs, promote PDL remodeling and bone regeneration in rat periodontal defectsActivate p38 pathwaycollagen sponge;
/
Buccal side of the lower 1st molar;
2 w, 4 w, 8 w
[70]
LPS-DFC-EVsImprove proliferation, migration, osteogenic differentiation of PDLSCs, promote PDL remodeling and bone regeneration in periodontitis ratsDecrease the RANKL/OPG ratioNanocomposite hydrogel;
50 µg/week
Right upper 2nd molar;
4 w, 8 w
[71]
LPS-DFSC-EVsImprove proliferation, migration of PDLSCs, promote PDL remodeling and new bone formation in beagle dogs with periodontitisActivate the ROS/MAPK pathway, promote M2 macrophage polarizationHyaluronic acid hydrogel;
200 µg/week
The lower 3rd premolar;
8 w
[72]
GMSC-EVsPromote bone regeneration in rat periodontal defectsInhibit T cell activation, increase Treg cell formationPolymer particle;
≈230 μg
Mesiolingual side of the upper 1st molar;
8 w
[50]
HHH-DPC-EVsImprove migration of MC3T3-E1 cells, alleviate bone resorption in mice with periodontitisInhibit osteoclast formationSaline solution;
7.5 × 108/time, day 0, 2, 4
Right upper 2nd molar;
1 w
[68]
DPSC-EVsPromote periodontal epithelial healing and bone regeneration in mice with periodontitisDeliver miR-1246 to promote M2 macrophage polarizationChitosan hydrogel;
50 μg
Left upper 2nd molar;
4 w
[54]
M2-EVsImprove osteogenesis of BMMSCs, alleviate bone resorption in mice with periodontitisDeliver IL-10 to activate the IL-10/IL-10R pathwayPBS suspension;
3 µg/time, day 1, 4, 7
Right upper 2nd molar;
2 w
[56]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

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/10.3390/ijms24065790

AMA Style

Huang X, Wang H, Wang C, Cao Z. The Applications and Potentials of Extracellular Vesicles from Different Cell Sources in Periodontal Regeneration. International Journal of Molecular Sciences. 2023; 24(6):5790. https://doi.org/10.3390/ijms24065790

Chicago/Turabian Style

Huang, Xin, Huiyi Wang, Chuan Wang, and Zhengguo Cao. 2023. "The Applications and Potentials of Extracellular Vesicles from Different Cell Sources in Periodontal Regeneration" International Journal of Molecular Sciences 24, no. 6: 5790. https://doi.org/10.3390/ijms24065790

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop