Next Article in Journal
Effects of Antifibrotic Therapy in Patients with Combined Pulmonary Fibrosis and Emphysema: A US-Based Cohort Study
Previous Article in Journal
Macrophages in Autoimmune Liver Diseases: From Immune Homeostasis to Precision-Targeted Therapy
Previous Article in Special Issue
Imbalance of Serum Bone-Metabolism-Related Factors Associated with Osteonecrosis of the Jaw
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Roles of Extracellular Vesicles in the Pathogenesis, Diagnosis, and Therapy of Periodontitis

1
Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710049, China
2
Department of Orthodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710049, China
3
Department of Periodontology, College of Stomatology, Xi’an Jiaotong University, Xi’an 710049, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2025, 13(10), 2521; https://doi.org/10.3390/biomedicines13102521
Submission received: 31 August 2025 / Revised: 29 September 2025 / Accepted: 10 October 2025 / Published: 16 October 2025
(This article belongs to the Special Issue Biomedicine in Dental and Oral Rehabilitation)

Abstract

Periodontitis is a globally prevalent oral disease and is closely associated with various systemic diseases. Periodontitis arises from dynamic and complex interactions between polymicrobial communities and host immune responses. Extracellular vesicles (EVs) are circulating subcellular particles carrying multiple signaling molecules. EVs play a key role in intercellular communication, and hold promise for diagnostic and therapeutic purposes. Bacterial extracellular vesicles (BEVs), released from oral pathogens, have been implicated in delivering virulence factors to host cells. In contrast, host cell-derived EVs (CEVs), secreted by periodontal cells, contain molecular cargo that reflect disease status. Both BEVs and CEVs contribute to periodontitis progression by exacerbating inflammation and tissue destruction, and they may also influence related systemic diseases. Moreover, the molecular components of EVs derived from saliva and gingival crevicular fluid (GCF) show potential as diagnostic biomarkers for periodontitis. In addition, mesenchymal stem cell-derived EVs (MSC-EVs) exhibit therapeutic potential in periodontitis, and engineering approaches have been developed to enhance their therapeutic efficacy and accelerate clinical translation. This review summarizes recent advances in understanding the pathogenic, diagnostic, and therapeutic roles of EVs in periodontitis and discusses current challenges and future directions toward their clinical application.

1. Introduction

Periodontitis is a highly prevalent oral disease [1], affecting nearly 62% of the global population [2]. Recent data from the Global Burden of Disease Study 2021 reported that in 2021 there were approximately 951 million cases worldwide, with the highest prevalence observed in Asia and South Asia, and particularly in low- and middle-income regions. The burden of periodontitis increases with age, peaking in the 50–59 year age group, and it accounted for 6.2 million disability-adjusted life years (DALYs) globally in 2021 [3], underscoring its substantial public health impact. Beyond the health burden, periodontitis also exerts a major economic impact, with an estimated USD 54 billion in productivity losses annually and contributing significantly to the USD 442 billion global costs of oral diseases each year [4]. Clinically, the diagnosis of periodontitis still mainly relies on visual inspection, periodontal probing, and radiographic evaluation, which lack sufficient sensitivity and specificity for assessing current disease activity or predicting future progression. In terms of therapy, conventional approaches including nonsurgical treatment, surgical interventions, and adjunctive medications can control inflammation and slow disease progression, but they are prone to recurrence, offer limited long-term efficacy, and fail to achieve functional regeneration of lost tissues [5,6,7,8]. The primary feature of periodontitis is progressive destruction of periodontal supporting tissues. As the predominant cause of tooth loss in adults, periodontitis severely impairs mastication, facial aesthetics, and quality of life [9]. Importantly, increasing evidence demonstrates that periodontitis is closely associated with systemic diseases, including cardiometabolic diseases [10], neurodegenerative disorders [11,12], autoimmune diseases [13], and cancer [14,15], suggesting its broader clinical significance.
The pathogenesis of periodontitis is driven by complex interactions between polymicrobial communities and host immune responses [16]. These interactions are largely mediated by the secretome of both host and microbial cells, among which EVs have attracted considerable attention [17]. EVs are nanoscale lipid bilayer vesicles that carry nucleic acids, proteins, lipids, and other bioactive molecules [18,19], and they are increasingly recognized as critical mediators in the progression of periodontitis as well as potential contributors to associated systemic diseases [20,21]. Notably, EVs secreted from oral pathogens can encapsulate a variety of virulence factors and deliver them into host cells, thereby modulating immune responses and aggravating tissue destruction at local and distant sites [22,23]. Additionally, the double-membrane structure of EVs protects their cargo from enzymatic degradation [24], rendering EVs derived from saliva and GCF promising candidates for periodontal diagnostics [25]. Furthermore, MSC-EVs hold therapeutic potential in periodontitis due to their low immunogenicity, high safety, and multiple bioactivities [26]. Recently, engineering approaches have been developed to increase EV yield and enhance their therapeutic efficacy, enabling sustained, precise, and effective treatment in periodontitis [27,28].
We conducted a literature search primarily in PubMed for all publications related to EVs and periodontitis up to August 2025, with a particular focus on studies published in the last 5 years. The search was performed using combinations of keywords such as “extracellular vesicles”, “exosomes”, “periodontitis”, “gingival crevicular fluid”, “saliva”, “diagnostic biomarkers”, and “therapy”. We excluded non-English publications, inaccessible full texts, duplicates, meeting abstracts, and policy papers. Eligible studies were peer-reviewed original articles and reviews addressing the pathogenic, diagnostic, or therapeutic roles of EVs in periodontitis. In this review, we outline the roles of EVs in the pathogenesis of periodontitis and related systemic diseases, encompassing both BEVs and CEVs. We then discuss the diagnostic potential of EVs derived from saliva and GCF for periodontitis. We also summarize engineering strategies to optimize the therapeutic efficacy of MSC-EVs in periodontitis. Finally, we present the key challenges and future perspectives of EV-based applications in periodontitis.

2. Pathogenic Roles of BEVs in Periodontitis and Associated Systemic Diseases

BEVs are conventionally classified as outer membrane vesicles, outer–inner membrane vesicles, and cytoplasmic membrane vesicles, depending on their biogenetic pathways including outer membrane blebbing and explosive cytolysis [29]. Structurally, BEVs are spherical nanoscale vesicles with a double-layered membrane, enriched in pathogen-associated molecular patterns such as lipopolysaccharides (LPS) [30], toxins such as gingipains [31] and leukotoxin [32], nucleic acids [33], and other virulence factors, which enable them to trigger strong inflammatory responses and promote tissue destruction [34].

3. BEVs and Periodontitis

The gingival epithelium is a physical barrier serving as a critical frontline defense against microbial invasion. Gingival epithelial cells responded strongly to Porphyromonas gingivalis (P. gingivalis) EVs, which activated MAPK (Erk1/2, JNK, p38) and STING signaling pathways, resulting in elevated production of IL-6 and IL-8 to exacerbate epithelial inflammation and compromise barrier function [35]. EVs from Filifactor alocis (F. alocis) stimulated the release of cytokines including CXCL1, IL-6, IL-8, and GM-CSF in human oral keratinocytes, mimicking whole-bacterial stimulation [36]. Oral fibroblasts, another essential component of the connective barrier, internalized P. gingivalis EVs, which dose-dependently suppressed fibroblast proliferation and growth [37].
Beyond barrier disruption, BEVs profoundly influence innate and adaptive immune defenses, driving a proinflammatory state in periodontal tissues. Neutrophils, as the first line of defense, underwent degranulation and released antimicrobial components such as LL-37 and myeloperoxidase, which were subsequently degraded by P. gingivalis EVs-bound gingipains, thereby promoting bacterial survival [38]. EVs secreted by Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) can induce the formation of neutrophil extracellular traps (NETs) [39]. Moreover, EVs shed from dental biofilms strongly induce neutrophil extracellular trap formation via a noncanonical cytosolic LPS/caspase-4/11/Gasdermin D pathway [40]. Following neutrophil activation, macrophages are another key target. Interestingly, P. gingivalis infection alone failed to trigger inflammasome activation in macrophages. In contrast, macrophages stimulated with P. gingivalis-EVs exhibited robust inflammasome activation, characterized by caspase-1 cleavage, abundant production of IL-1β and IL-18, and marked actate dehydrogenase release together with positive 7-AAD staining, indicative of pyroptotic cell death [41]. A. actinomycetemcomitans-EVs activated macrophages through the TLR-8/NF-κB signaling axis, leading to inflammasome activation and enhanced TNF-α release [42]. EVs derived from Tannerella forsythia (T. forsythia) activated macrophages through toll-like receptor 2 (TLR2) signaling and T. forsythia was also found to actively release EVs upon exposure to macrophage-derived soluble factors, suggesting a dynamic host–pathogen interaction that further amplifies periodontal inflammation [17]. EVs derived from Fusobacterium nucleatum (F. nucleatum) drove polarization toward the proinflammatory M1 phenotype. This M1-skewed response amplified local tissue destruction and accelerated alveolar bone loss [43]. F. alocis EVs markedly induced the expression of multiple inflammatory cytokines in macrophages, exhibiting immunostimulatory activity comparable to that of whole bacterial cells [36]. Further downstream, dendritic cells (DCs), as crucial antigen-presenting cells bridging innate and adaptive immunity, are highly susceptible to BEVs’ effects [44]. EVs from red complex pathogens (P. gingivalis, T. forsythia, Treponema denticola) promoted DC maturation by upregulating MHC class II and costimulatory molecules (CD80, CD86, CD40), and further skewed CD4+ T cell differentiation toward Th1 and Th17 lineages, thereby sustaining chronic inflammation and tissue degradation in periodontitis [45].
BEVs also perturb the osteoimmune balance, directly contributing to alveolar bone loss. On the osteogenic side, F. alocis EVs inhibited osteogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs) via TLR2 signaling and increased the RANKL/OPG ratio, favoring osteoclastogenesis [46]. On the osteoclastic side, EVs from P. gingivalis, T. forsythia, and Streptococcus oralis promoted osteoclast differentiation through TLR2 activation by lipoproteins/LPS [47]. The BEVs derived from periodontal pathogens and their roles in periodontitis are summarized in Table 1 and Figure 1.

4. BEVs and Associated Systemic Diseases

BEVs have been implicated as key contributors to the pathogenesis and progression of Alzheimer’s disease. Gingivally exposed P. gingivalis EVs can translocate into the brain via the trigeminal nerve and periodontal bloodstream, contributing to cognitive decline [48]. P. gingivalis EVs delivered gingipains into brain microvascular endothelial cells, where they degraded tight junction proteins ZO-1 and occludin, thereby disrupting the blood–brain barrier (BBB) [49]. EVs from A. actinomycetemcomitans, administered by intragingival injection or EV-infused gel, have been shown to penetrate the BBB and elicit neuroinflammation. Their extracellular RNA cargo can activate TLR4 and TLR8 signaling pathways along with the downstream MyD88 pathway, inducing the production of proinflammatory cytokines including IL-6 and TNF-α [50]. Consistently, another in vitro study demonstrated that A. actinomycetemcomitans EV-associated RNAs activated the TLR8–NF-κB signaling axis in macrophages, leading to TNF-α expression [42]. Meanwhile, Hong et al. reported RNAs—but not DNA cargo—within A. actinomycetemcomitans EVs activated IL-6 and NF-κB signaling in microglia [51]. Notably, the glioblastoma-specific mutation EGFRvIII has been targeted in glioma models, where engineered BEVs carrying EGFRvIII epitopes suppressed tumor growth and elicited Th1-skewed immune responses characterized by CD4+ and CD8+ T-cell infiltration [52], underscoring the translational potential of BEV-based platforms in glioma immunotherapy.
Emerging evidence also highlights the detrimental impact of BEVs on vascular health. Gingipains presented on the surface of P. gingivalis EVs degraded endothelial cell–cell adhesins such as PECAM-1, increasing vascular permeability [53]. In diabetic retinopathy, P. gingivalis EVs accelerated blood-retinal barrier damage by inducing the expression of TNF-α, MMP-9, endothelial cell death, and barrier permeability via protease-activated receptor-2 [54].
Notably, BEVs also contribute to systemic bone loss. After intraperitoneal administration, F. alocis EVs accumulated in the long bones of mice and induced proinflammatory cytokine production, osteoclastogenesis, and bone resorption via TLR2 [55]. Moreover, F. alocis EVs inhibited the osteogenic differentiation of BMSCs by activating TLR2-mediated MAPK and NF-κB signaling, and further increased the RANKL/OPG ratio in BMSCs in a TLR2-dependent manner [46].
BEVs have been implicated in other diseases as well. For instance, P. gingivalis EVs induced cell death of lung epithelial cells via caspase-3 activation, PARP cleavage, and disruption of membrane integrity, indicating a link between periodontitis and aspiration pneumonia [56]. In the liver, P. gingivalis EVs impaired insulin-induced Akt/glycogen synthase kinase-3β (GSK-3β) signaling via gingipain-dependent pathways, leading to reduced glycogen synthesis and hyperglycemia, thereby contributing to diabetes development [57]. Moreover, P. gingivalis EVs are associated with adverse pregnancy outcomes. P. gingivalis EVs can be internalized by trophoblast cells, where they can suppress glycolysis and induce a metabolically quiescent state, impairing trophoblast migration and invasion [58]. The BEVs derived from periodontal pathogens and their roles in periodontitis-related systemic diseases are summarized in Table 2 and Figure 2.

5. Pathogenic Roles of CEVs in Periodontitis and Associated Systemic Diseases

EVs are naturally secreted by virtually all cell types and are conventionally classified into endosome-derived exosomes, plasma membrane-derived microvesicles, and apoptotic bodies according to the biogenesis and release pathways [59,60]. EVs may undergo clathrin-dependent, caveolin-dependent, lipid raft–mediated endocytosis, macropinocytosis, and phagocytosis, as well as direct membrane fusion, thereby enabling their cargo to be released into the cytoplasm [61]. The cargo composition of EVs (such as proteins, transcription factors, nucleic acids, etc.) varies with the parental cell type, physiological conditions, and biogenesis [62,63,64]. Increasing evidence indicates that EVs derived from periodontal cells not only aggravate local periodontal damage (Table 3 and Figure 3) but also contribute to the progression of related systemic diseases (Table 4 and Figure 3).

6. CEVs and Periodontitis

During the progression of periodontitis, stem cell-derived EVs act as inflammatory amplifiers by aberrantly activating immune cells. In the inflamed microenvironment, EVs from Gli1+ MSCs can stimulate neutrophil activation via the CXCL1–CXCR2 axis, thereby exacerbating alveolar bone destruction [65]. LPS-stimulated periodontal ligament stem cells (PDLSCs) released EVs that drive macrophage polarization toward the proinflammatory M1 phenotype through multiple mechanisms [66]. For instance, miR-433-3p activated TLR2/TLR4/NF-κB signaling, while miR-143-3p inhibited PI3K/AKT and concomitantly activated NF-κB, collectively promoting M1 polarization, fibroblast apoptosis, and alveolar bone resorption [67,68].
In addition to modulating immune responses, EVs derived from various cell types impair osteogenesis and exacerbate bone loss [82]. Neutrophil-derived EVs inhibited the osteogenic differentiation of PDLSCs by delivering miR-223 and regulating the cGMP–PKG pathway [69]. EVs from inflammatory macrophages transferred damaged mitochondria to bone marrow mesenchymal stem cells (BMSCs), thereby impairing osteogenesis through the LCN2/OMA1/OPA1 axis [70]. Osteoclast-derived EVs enriched in miR-5134-5p suppressed JAK2/STAT3 signaling, leading to a marked reduction in osteogenic activity [71].
EVs also regulate osteoclast differentiation to modulate alveolar bone homeostasis. EVs from inflammatory PDLSCs enriched in RANKL and TNF-α strongly promoted osteoclastogenesis [72]. Osteoblast-derived EVs delivered the long noncoding RNA MALAT1, which modulated the miR-124/NFATc1 axis to enhance osteoclast differentiation [73]. P. gingivalis LPS-stimulated BMSCs secreted EVs that downregulated miR-151-3p and activated PAFAH1B1 signaling, further facilitating osteoclastogenesis [74]. In contrast, M2 macrophage-derived EVs carrying miR-1227-5p suppressed osteoclast-associated receptor (OSCAR) expression and inhibited osteoclast formation [75].

7. CEVs and Associated Systemic Diseases

EVs released from P. gingivalis LPS-stimulated periodontal cells, including macrophages and periodontal ligament fibroblasts, activated stearoyl-CoA desaturase-1 (SCD-1) and suppressed AMPK signaling in hepatocytes, thereby inducing hepatic injury and steatosis [76]. Gingival extracellular vesicles generated by polymicrobial periodontal infection disrupted adipose tissue function, as evidenced by reduced AKT phosphorylation, decreased adiponectin and leptin levels, and downregulation of genes related to adipogenesis and lipogenesis [77]. Plasma EVs from patients with periodontitis impaired insulin signaling and glucose uptake in hepatoma cells, exacerbating insulin resistance in type 2 diabetes [78,79]. Furthermore, plasma EVs enriched in miR-155-5p may increase vascular permeability and inflammation, accelerating carotid atherosclerosis [80]. In addition, EVs released by P. gingivalis infected macrophages have been shown to disrupt placental angiogenesis and downregulate VEGFR1 expression, leading to adverse pregnancy outcomes [81]. Collectively, these findings reinforce the pathological link between periodontitis and systemic diseases, and highlight periodontal cell-derived EVs as promising therapeutic targets. Current evidence indicates that in periodontitis, pathological CEVs are predominantly released in response to polymicrobial infection and bacterial components such as LPS, thereby contributing to systemic disease progression. While the possibility that BEVs directly regulate CEVs release is intriguing, current evidence is lacking, and future studies are needed to elucidate this potential interaction.

8. Diagnostic Role of EVs in Saliva and GCF

Currently, the diagnosis of periodontitis mainly depends on clinical parameters such as plaque index, gingival index, bleeding on probing, periodontal pocket depth, radiographic alveolar bone loss, and tooth mobility [9,83,84]. However, these assessments largely provide historical data on periodontal tissue rather than predicting future disease activity, thereby limiting early detection and timely intervention. The molecular content of EVs in body fluids varies depending on whether cells are in a physiological or pathological state [85,86]. GCF is in direct contact with periodontal lesions and can sensitively reflect local inflammatory and tissue-destructive changes, while saliva, as a composite oral fluid, reflects both site-specific and overall oral health status. Importantly, both fluids can be collected non-invasively and painlessly [87,88,89]. EVs derived from GCF and saliva not only mirror the periodontal microenvironment but also may capture systemic pathological alterations, highlighting their potential as powerful tools for early disease screening, staging, and risk assessment (Figure 4) [90,91].

9. EVs in GCF

Compared with healthy and gingivitis subjects, the total concentration of EVs in GCF was significantly increased in periodontitis, whereas no significant difference was observed in saliva-derived EVs. The exosomal marker CD63 was also elevated in the GCF of periodontitis patients [92]. Previous studies have also demonstrated that GCF-EVs contain host-derived and bacterial-derived components, including cytokines, proteases, and nucleic acids, which are closely associated with periodontal inflammation and tissue destruction [93,94]. In addition, GCF-EVs were increased in both gestational diabetes mellitus (GDM) and periodontitis, and their proteomic cargos included peptides of both host and bacterial origin that participated in immune–inflammatory responses, glucose metabolism, and insulin signaling [95]. Collectively, these findings highlight the potential of GCF-EVs as biomarkers for the early diagnosis and monitoring of periodontitis and its related systemic diseases.

10. EVs in Saliva

Significant alterations in salivary EV composition have been observed in periodontitis and after initial periodontal therapy, involving microRNAs (miRNAs), messenger RNAs (mRNAs), proteins, and surface markers, all demonstrating considerable diagnostic potential [96]. At the miRNA level, 33 plasma miRNAs were significantly downregulated, while 1995 salivary miRNAs (1985 downregulated and 10 upregulated) were differentially expressed in periodontitis. Among them, miR-let-7d, miR-126-3p, and miR-199a-3p in plasma, and miR-125a-3p in saliva (all AUC = 1) in a pilot study of only 16 participants (8 periodontitis vs. 8 healthy), although the authors noted that these findings require validation in larger cohorts [97]. Compared with miRNAs from whole saliva, salivary EVs-derived miR-140-5p, miR-146a-5p, and miR-628-5p were markedly upregulated in periodontitis patients, each showing strong diagnostic power (AUC > 0.9) [98]. Xie et al. reported decreased miR-223-3p in salivary EVs and proposed its role in regulating GSDMD-mediated pyroptosis via NLRP3, suggesting potential for periodontitis diagnosis [99]. At the mRNA level, salivary exosomal PD-L1 was elevated and varied across the stages of periodontitis, indicating potential for both diagnosis and staging [100]. Meanwhile, osterix (OSX) mRNA decreased and TNF-α mRNA increased in periodontitis, showing moderate discriminatory capacity (AUC > 0.72) [101]. At the protein level, proteomic analyses revealed that proteins isolated from the saliva EVs of young adults with severe periodontitis were enriched in pathways related to innate immunity, cytolysis, and complement activation [102]. Han et al. found that EVs from periodontitis patients contained higher IL-6 and IL-8, and lower IL-10, reflecting a proinflammatory state in periodontitis [103]. Moreover, global 5mC hypermethylation was elevated in periodontitis, demonstrating high diagnostic accuracy (AUC = 1) in a pilot study of 22 participants, though age and smoking status differed among groups and no external validation was performed [104]. Regarding surface markers, significantly decreased CD9 and CD81 were detected in periodontitis and correlated with disease stage, although another study reported an increase in the CD9+ subpopulation [101], suggesting that the diagnostic value of CD9 requires further validation [105].

11. Strategies to Improve Therapeutic Function of MSC-EVs in Periodontitis

Many studies have demonstrated that MSC-EVs promote the repair of complex periodontal tissues through diverse bioactivities, including enhanced cell proliferation [106,107], migration [108], angiogenesis [109], and multilineage differentiation [110]. These underlying mechanisms have been comprehensively summarized in several reviews [111,112,113]. Nevertheless, the clinical translation of EVs remains hampered by challenges such as low production efficiency, rapid clearance, limited tissue specificity, and unpredictable cargo loading [27,114,115]. To address these limitations, recent studies have investigated various engineering strategies to enhance the therapeutic efficacy of EVs (Figure 5) [116].

12. Pretreatment Approaches of Parental Cells

The yield and functional cargo of natural MSC-EVs are inherently limited. Pretreatment with biochemical agents significantly enhances the therapeutic potential of MSC-EVs. For instance, metformin increased PDLSC-EVs secretion and improved their osteogenic and anti-inflammatory activities [117]. Psoralen upregulated miR-125b-5p in PDLSC-EVs, thereby promoting osteogenic differentiation [118]. Gallic acid alleviated inflammation in PDLSCs and enhanced the pro-osteogenic effects of their EVs [119]. EVs from LPS-preconditioned dental follicle cells attenuated alveolar bone loss by modulating RANKL/OPG-mediated osteoclastogenesis and inducing M2 macrophage polarization, while also protecting periodontal ligament cells (PDLCs) against apoptosis and dysfunction through JNK and p38 signaling pathways [120,121].
Genetic modification of parental cells represents another effective strategy to improve EV bioactivity. EVs from FoxO1-overexpressing PDLSCs were enriched in FoxO1 and demonstrated enhanced osteogenic capacity while suppressing inflammation [122]. CXCR4-transfected cells generated EVs with elevated CXCR4 expression, endowing them with improved “homing” ability toward inflammatory sites [123]. Chen et al. reported that EVs derived from P2X7R-modified PDLSCs restored the function of inflammation-compromised PDLSCs by transferring miR-3679-5p, miR-6515-5p, and miR-6747-5p [124]. Additionally, the cationic antimicrobial peptide DP7-C has been applied as a transfection carrier to deliver miR-26a and miR-21b into parental cells, producing EVs enriched with these therapeutic miRNAs [125,126].
Parental cells can also be modulated through the culture microenvironment to optimize EV production. For example, dental pulp stem cells cultured in three-dimensional systems secreted EVs that restored the Th17/Treg balance in inflamed periodontal tissues via the miR-1246/Nfat5 axis [127]. Moreover, low-intensity pulsed ultrasound (LIPUS) stimulation of stem cells from apical papilla promoted the secretion of miR-935-enriched EVs with enhanced osteogenic and anti-inflammatory properties [128].

13. Direct Engineering Approaches of EVs

Recent studies have increasingly explored direct engineering strategies to load therapeutic agents into EVs for the treatment of periodontitis. Approaches such as sonication, electroporation, freeze–thaw treatments, and extrusion have been employed to enhance EV drug-loading efficiency [129,130,131]. Notably, melatonin-loaded M2 macrophage-derived EVs restored osteogenic and cementogenic differentiation of inflamed PDLCs by alleviating excessive endoplasmic reticulum stress and unfolded protein response [132]. Aspirin-loaded PDLSC-derived EVs improved the periodontal immune microenvironment by promoting oxidative phosphorylation (OXPHOS) in macrophages, thereby reducing alveolar bone loss [133]. Likewise, methotrexate-loaded EVs derived from human umbilical cord mesenchymal stem cells suppressed proinflammatory macrophage activation through acyl-CoA synthetase-1 (ACSL1) downregulation and OXPHOS restoration, ultimately improving therapeutic outcomes in periodontitis [134,135].

14. MSC-EVs Combined with Biomaterials Promote Periodontal Tissue Regeneration

The limited half-life of EVs following administration, coupled with their rapid diffusion from the site of delivery, compromises therapeutic benefits. To overcome this, hydrogels with good biocompatibility and porous architectures have been developed as promising delivery systems for EVs. Among them, Hydrogels composed of chitosan (CS) and β-sodium glycerophosphate (β-GP) are widely recognized for their injectability and ability to undergo thermosensitive gelation at physiological temperature, making them suitable for repairing irregular tissue defects. Incorporation of dental pulp stem cell-derived EVs into CS hydrogels accelerated alveolar bone and periodontal epithelial regeneration in murine models of periodontitis [136]. Sun et al. further improved CS hydrogels by adding gelatin, which shortened gelation time, while erythropoietin-stimulated EVs encapsulated in CS/β-GP/gelatin hydrogels ensured sustained release and prolonged retention in vivo [137]. To address the limited antimicrobial activity of EVs, Su et al. incorporated copper ions (Cu2+) and EVs from exfoliated deciduous teeth into hyaluronic acid hydrogels, achieving synergistic antibacterial and osteogenic effects [138]. Similarly, Xie et al. developed thermosensitive hydrogels co-loaded with BMSC-derived EVs and calcium peroxide nanoparticles, which provided sustained oxygen release, thereby alleviating anaerobic infections and promoting periodontal regeneration [139]. In addition, controlled-release systems have been designed for pathological site-specific delivery. For example, EVs derived from gingival mesenchymal stem cells immobilized on microparticles via matrix metalloproteinase (MMP)-sensitive linkers enabled targeted release at inflammatory sites, resulting in localized immunomodulation and significantly improved periodontal regeneration [140].

15. Summary and Prospect

Oral BEVs play pivotal roles in the initiation and progression of periodontitis and are increasingly recognized as contributors to related systemic diseases. Accumulating evidence indicates that BEVs can degrade connective tissue, amplify inflammatory responses, and disturb alveolar bone homeostasis, thereby exacerbating periodontal destruction. Despite these advances, the precise molecular mechanisms underlying BEV-mediated pathogenicity remain insufficiently understood and require further in-depth investigation. To enhance reproducibility and comparability across studies, it is crucial to optimize BEV isolation and purification techniques and to establish standardized criteria for their characterization. Furthermore, the application of multi-omics strategies will be instrumental in defining the detailed cargo composition and functional heterogeneity of BEVs derived from diverse bacterial species. Such approaches may ultimately facilitate the identification of specific pathogenic signatures and promote the development of innovative anti-BEV therapeutic strategies for periodontitis and related systemic diseases.
EVs derived from saliva and GCF hold great promise as non-invasive diagnostic biomarkers for periodontitis. However, several important challenges remain to be addressed before clinical translation. Methodological standardization is critical to improve reproducibility. According to the Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines from the International Society for Extracellular Vesicles (ISEV), oral EV studies should follow standardized workflows covering pre-analytical variables (sample source, collection method, storage), isolation strategies (e.g., differential ultracentrifugation, density gradients, size-exclusion chromatography, fluid flow-based separation, charge and molecular recognition-based separations), and comprehensive characterization, including particle analysis, morphology, and molecular markers. To ensure rigor, both positive markers (CD9, CD63, CD81, TSG101, ALIX, syntenin-1) and negative markers (calnexin, GM130, GRP94) should be evaluated to confirm vesicular identity and exclude contaminants. Furthermore, oral biofluids present notable pitfalls [141,142,143,144]. Oral biofluids is prone to contamination by food debris and oral bacteria, and contains endogenous enzymes such as amylase. These factors can obscure biomarker detection and reduce sensitivity and specificity by promoting rapid protein/RNA degradation and denaturation [145,146].
EVs in saliva and GCF originate from multiple host cell types and diverse bacterial species, generating substantial heterogeneity that complicates interpretation. Advanced fractionation and isolation strategies are urgently needed to accurately separate bacterial EVs from host-derived vesicles and to resolve distinct EV subpopulations. The use of immunoaffinity capture with BEV-specific markers such as LPS and lipoteichoic acid (LTA) and CEV-specific markers such as CD9, CD63, CD81, TSG101, and ALIX represents a promising approach [141,147]. In addition, emerging single-EV multi-omics technologies may uncover source-specific molecular signatures and define robust biomarker panels with high diagnostic accuracy. Importantly, the cargo of oral EVs reflects both the periodontal pathogen burden and the host inflammatory status. Therefore, integrating EV-based analyses with conventional microbiological and inflammatory biomarkers may enable a more comprehensive assessment of periodontitis activity. Finally, large-scale, multicenter clinical studies across different age groups, genders, and ethnic backgrounds are required to validate findings and establish standardized diagnostic thresholds, thereby enabling early periodontitis screening and personalized risk assessment.
MSC-EVs have emerged as a compelling alternative to conventional stem cell therapy and are increasingly recognized as a promising therapeutic modality for periodontal regeneration. The yield and functional activity of MSC-EVs are strongly influenced by parental cell type, culture conditions, and passage number [148,149]. Furthermore, large-scale production and purification of MSC-EVs remain costly. Advances in bioreactor technologies and standardized isolation protocols may improve production efficiency and cost-effectiveness [150,151]. For GMP-grade production, closed and scalable bioreactor systems with full process validation are required, and batch release testing must confirm identity, purity, sterility, potency, and safety. Storage and stability also represent critical challenges, as EVs are stable at −80 °C but prone to aggregation or degradation at higher temperatures, underscoring the need for standardized release criteria and storage conditions to ensure reproducibility [152,153].
In terms of safety, preclinical studies indicate that MSC-EVs are generally low-immunogenic. However, potential risks including off-target effects, unpredictable biodistribution, and excessive immunosuppression must be carefully addressed. Therefore, rigorous safety evaluations and long-term immunological assessments should be carefully evaluated and monitored. Systematic investigations of in vivo pharmacokinetics and pharmacodynamics are essential to clarify biodistribution, clearance profiles, and optimal dosing regimens. Considering the complexity of periodontal tissue repair, future strategies should also emphasize multifunctional and engineered approaches that integrate osteogenic, angiogenic, immunomodulatory, and antibacterial properties, possibly through the development of EV-based combinatorial biomaterials to achieve safer and more predictable therapeutic outcomes.
Translation into clinical practice will require carefully designed first-in-human trials. One possible design is a randomized, double-blind, placebo-controlled study in systemically healthy adults with periodontitis. Exclusion criteria would include smoking, diabetes, pregnancy, corticosteroid or contraceptive use, and poor oral hygiene. Patients who had completed initial periodontal therapy, such as scaling and root planing, could then be randomized to receive either local MSC-EV administration or placebo. Therapeutic efficacy would be assessed by monitoring changes in gingival inflammation, probing pocket depth, clinical attachment level, and alveolar bone level.
Collectively, EVs are pivotal in the pathogenesis, diagnosis, and therapy of periodontitis. Nonetheless, several key barriers currently limit their clinical application (Table 5). Continued efforts are essential to accelerate the clinical translation of EV-based diagnostics and therapeutics for periodontitis.

Author Contributions

Conceptualization, A.L. and R.T.; investigation, Y.F.; writing—original draft preparation, Y.F. and M.W.; writing—review and editing, Y.F. and M.W.; visualization, M.W.; supervision, A.L. and R.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (Grant No. W2411086), Key Project of Shaanxi Provincial Natural Science Foundation (2025 JC-QYXQ-045), Key R&D Program of Shaanxi Province (2025YF-04), the Shaanxi Provincial High-level Talent Program and Young Talent Support Plan of Xi’an Jiaotong University and the Shaanxi Natural Science Basic Research Program (Grant No. 2025JC-YBQN-1059).

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
EVsextracellular vesicles
GCFgingival crevicular fluid
MSC-EVsmesenchymal stem cell–derived EVs
BEVsbacterial extracellular vesicles
CEVscellular extracellular vesicles
LPSlipopolysaccharides
P. gingivalisPorphyromonas gingivalis
T. forsythiaTannerella forsythia
F. nucleatumFusobacterium nucleatum
F. alocisFilifactor alocis
A. actinomycetemcomitansAggregatibacter actinomycetemcomitans
DCsdendritic cells
IL-6interleukin-6
IL-8interleukin-8
CXCL1C-X-C motif chemokine ligand 1
GM-CSFgranulocyte–macrophage colony-stimulating factor
TLR2toll-like receptor 2
MHCmajor histocompatibility complex
BBBblood–brain barrier
ZO-1zonula occludens-1
TLR4toll-like receptor 4
TLR8toll-like receptor 8
TNF-αtumor necrosis factor-alpha
PECAM-1platelet endothelial cell adhesion molecule-1
MMP-9matrix metalloproteinase-9
GSK-3βglycogen synthase kinase-3β
CXCR2C-X-C chemokine receptor type 2
PDLSCsperiodontal ligament stem cells
SCD-1stearoyl-CoA desaturase-1
VEGFR1vascular endothelial growth factor receptor 1
GDMgestational diabetes mellitus
OSXosterix
IL-10interleukin-10
5mC5-methylcytosine
PDLCsperiodontal ligament cells
FoxO1forkhead box protein O1
CXCR4C-X-C chemokine receptor type 4
OXPHOSoxidative phosphorylation
ACSL1acyl-CoA synthetase-1
CSchitosan
β-GPβ-sodium glycerophosphate
MMPmatrix metalloproteinase

References

  1. GBD 2021 Oral Disorders Collaborators Trends in the Global, Regional, and National Burden of Oral Conditions from 1990 to 2021: A Systematic Analysis for the Global Burden of Disease Study 2021. Lancet 2025, 405, 897–910. [CrossRef]
  2. Trindade, D.; Carvalho, R.; Machado, V.; Chambrone, L.; Mendes, J.J.; Botelho, J. Prevalence of Periodontitis in Dentate People between 2011 and 2020: A Systematic Review and Meta-Analysis of Epidemiological Studies. J. Clin. Periodontol. 2023, 50, 604–626. [Google Scholar] [CrossRef]
  3. Fu, H.; Li, X.; Zhang, R.; Zhu, J.; Wang, X. Global Burden of Periodontal Diseases among the Working-Age Population from 1990–2021: Results from the Global Burden of Disease Study 2021. BMC Public Health 2025, 25, 1316. [Google Scholar] [CrossRef]
  4. Hashim, N.T.; Babiker, R.; Padmanabhan, V.; Ahmed, A.T.; Chaitanya, N.C.S.K.; Mohammed, R.; Priya, S.P.; Ahmed, A.; El Bahra, S.; Islam, M.S.; et al. The Global Burden of Periodontal Disease: A Narrative Review on Unveiling Socioeconomic and Health Challenges. Int. J. Environ. Res. Public Health 2025, 22, 624. [Google Scholar] [CrossRef]
  5. Liu, Y.; Yin, Z.; Wu, H.; Li, R.; Li, J.; Wang, C.; Yi, L.; Zeng, M.; Huang, C.; Fu, L. Antibacterial-Osteogenic Integrated Liquid Metal Nanomedicine for Periodontitis Treatment. Cell Biomater. 2025, 1, 100009. [Google Scholar] [CrossRef]
  6. Tao, L.-R.; Li, Y.; Wu, X.-Y.; Gu, Y.; Xie, Y.; Yu, X.-Y.; Lai, H.-C.; Tonetti, M.S. Deep Learning Photo Processing for Periodontitis Screening. J. Dent. Res. 2025, 220345251347508. [Google Scholar] [CrossRef]
  7. Zhang, J.; Meng, L.; Jia, Y.; Li, J.; Xu, X.; Xu, X. Development of an Injectable Salicylic Acid-Choline Eutectic Hydrogel for Enhanced Treatment of Periodontitis. Mater. Horiz. 2025, 12, 3788–3802. [Google Scholar] [CrossRef] [PubMed]
  8. Tan, M.; Cui, Z.; Li, Y.; Fang, Y.; Mei, L.; Zhao, Y.; Wu, X.; Lai, H.; Tonetti, M.S.; Shen, D. PerioAI: A Digital System for Periodontal Disease Diagnosis from an Intra-Oral Scan and Cone-Beam CT Image. Cell Rep. Med. 2025, 6, 102186. [Google Scholar] [CrossRef] [PubMed]
  9. Yin, C.; Fu, L.; Guo, S.; Liang, Y.; Shu, T.; Shao, W.; Xia, H.; Xia, T.; Wang, M. Senescent Fibroblasts Drive FAP/OLN Imbalance Through mTOR Signaling to Exacerbate Inflammation and Bone Resorption in Periodontitis. Adv. Sci. 2025, 12, e2409398. [Google Scholar] [CrossRef]
  10. Orlandi, M.; Masi, S.; Lucenteforte, E.; Bhowruth, D.; Malanima, M.A.; Darbar, U.; Patel, K.; Lim, C.; Curra, C.; Shiehfung, T.; et al. Periodontitis Treatment and Progression of Carotid Intima-Media Thickness: A Randomized Trial. Eur. Heart J. 2025, ehaf555. [Google Scholar] [CrossRef]
  11. Kong, L.; Li, J.; Gao, L.; Zhao, Y.; Chen, W.; Wang, X.; Wang, S.; Wang, F. Periodontitis-Induced Neuroinflammation Triggers IFITM3-Aβ Axis to Cause Alzheimer’s Disease-like Pathology and Cognitive Decline. Alzheimer’s Res. Ther. 2025, 17, 166. [Google Scholar] [CrossRef]
  12. Qiu, C.; Zhou, W.; Shen, H.; Wang, J.; Tang, R.; Wang, T.; Xie, X.; Hong, B.; Ren, R.; Wang, G.; et al. Profiles of Subgingival Microbiomes and Gingival Crevicular Metabolic Signatures in Patients with Amnestic Mild Cognitive Impairment and Alzheimer’s Disease. Alzheimer’s Res. Ther. 2024, 16, 41. [Google Scholar] [CrossRef] [PubMed]
  13. Li, Y.; Chen, Y.; Deng, C.; Niu, Y.; Yang, Y.; Sun, S.; Hu, Z.; Wei, Y.; Xu, M.; Huang, Y.; et al. IFN-I-Mediated Neutropoiesis Bias Drives Neutrophil Priming and Inflammatory Comorbidities. Theranostics 2025, 15, 6058–6081. [Google Scholar] [CrossRef] [PubMed]
  14. Yuan, S.; Fang, C.; Leng, W.-D.; Wu, L.; Li, B.-H.; Wang, X.-H.; Hu, H.; Zeng, X.-T. Oral Microbiota in the Oral-Genitourinary Axis: Identifying Periodontitis as a Potential Risk of Genitourinary Cancers. Mil. Med. Res. 2021, 8, 54. [Google Scholar] [CrossRef]
  15. Tan, Q.; Ma, X.; Yang, B.; Liu, Y.; Xie, Y.; Wang, X.; Yuan, W.; Ma, J. Periodontitis Pathogen Porphyromonas gingivalis Promotes Pancreatic Tumorigenesis via Neutrophil Elastase from Tumor-Associated Neutrophils. Gut Microbes 2022, 14, 2073785. [Google Scholar] [CrossRef]
  16. Kinane, D.F.; Bornstein, M.M. Introduction to the Diagnostics in Periodontology and Implant Dentistry Issue. Periodontol 2000 2024, 95, 7–9. [Google Scholar] [CrossRef]
  17. Lim, Y.; Kim, H.Y.; Han, D.; Choi, B.-K. Proteome and Immune Responses of Extracellular Vesicles Derived from Macrophages Infected with the Periodontal Pathogen Tannerella forsythia. J. Extracell. Vesicles 2023, 12, e12381. [Google Scholar] [CrossRef]
  18. Xiao, Q.; Tan, M.; Yan, G.; Peng, L. Revolutionizing Lung Cancer Treatment: Harnessing Exosomes as Early Diagnostic Biomarkers, Therapeutics and Nano-Delivery Platforms. J. Nanobiotechnol. 2025, 23, 232. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, X.; Xu, Y.; Ma, L.; Yu, K.; Niu, Y.; Xu, X.; Shi, Y.; Guo, S.; Xue, X.; Wang, Y.; et al. Essential Roles of Exosome and circRNA_101093 on Ferroptosis Desensitization in Lung Adenocarcinoma. Cancer Commun. 2022, 42, 287–313. [Google Scholar] [CrossRef]
  20. Liu, J.; Dou, G.; Zhao, W.; Hu, J.; Jiang, Z.; Wang, W.; Wang, H.; Liu, S.; Jin, Y.; Zhao, Y.; et al. Exosomes Derived from Impaired Liver Aggravate Alveolar Bone Loss via Shuttle of Fasn in Type 2 Diabetes Mellitus. Bioact. Mater. 2024, 33, 85–99. [Google Scholar] [CrossRef]
  21. Zhang, H.; Zhou, L.-Q.; Yang, S.; Dong, M.-H.; Chen, L.; Lu, Y.-L.; Zhang, L.-Y.; Zhang, L.; Chu, Y.-H.; Xu, L.-L.; et al. The Foam Cell-Derived Exosomes Exacerbate Ischemic White Matter Injury via Transmitting Metabolic Defects to Microglia. Cell Metab. 2025, 37, 1636–1654.e10. [Google Scholar] [CrossRef]
  22. Chen, G.; Gao, C.; Jiang, S.; Cai, Q.; Li, R.; Sun, Q.; Xiao, C.; Xu, Y.; Wu, B.; Zhou, H. Fusobacterium nucleatum Outer Membrane Vesicles Activate Autophagy to Promote Oral Cancer Metastasis. J. Adv. Res. 2024, 56, 167–179. [Google Scholar] [CrossRef]
  23. Wu, Z.; Long, W.; Yin, Y.; Tan, B.; Liu, C.; Li, H.; Ge, S. Outer Membrane Vesicles of Porphyromonas gingivalis: Recent Advances in Pathogenicity and Associated Mechanisms. Front. Microbiol. 2025, 16, 1555868. [Google Scholar] [CrossRef]
  24. Li, Z.; Liang, Z.; Wang, P.; Li, W.; Li, Y.; Liu, N.; Ma, Q. SnS2 QDs@MXene Ohmic Junction-Based Surface Plasmon Coupling ECL Sensor to Detect Saliva Exosome for the Diagnosis of Childhood Asthma. Nano Lett. 2024, 24, 15878–15885. [Google Scholar] [CrossRef]
  25. Lv, Z.; Fu, K.; Zhang, Q. Advances of Exosomes-Based Applications in Diagnostic Biomarkers for Dental Disease and Dental Regeneration. Colloids Surf. B Biointerfaces 2023, 229, 113429. [Google Scholar] [CrossRef]
  26. Chen, L.; Zhu, S.; Guo, S.; Tian, W. Mechanisms and Clinical Application Potential of Mesenchymal Stem Cells-Derived Extracellular Vesicles in Periodontal Regeneration. Stem Cell Res. Ther. 2023, 14, 26. [Google Scholar] [CrossRef]
  27. Niu, L.; Ou, Q.; Ren, Q.; Li, Z.; Chen, H.; Lei, F.; Mao, X.; Shi, S.; Chen, Z.; Teng, W. Engineered Foxp1high Exosomes Ameliorates Systemic Lupus Erythematosus. Adv. Sci. 2025, 12, e15712. [Google Scholar] [CrossRef] [PubMed]
  28. Wu, J.; Wu, J.; Xiang, W.; Gong, Y.; Feng, D.; Fang, S.; Wu, Y.; Liu, Z.; Li, Y.; Chen, R.; et al. Engineering Exosomes Derived from TNF-α Preconditioned IPFP-MSCs Enhance Both Yield and Therapeutic Efficacy for Osteoarthritis. J. Nanobiotechnol. 2024, 22, 555. [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. Thapa, H.B.; Passegger, C.A.; Fleischhacker, D.; Kohl, P.; Chen, Y.-C.; Kalawong, R.; Tam-Amersdorfer, C.; Gerstorfer, M.R.; Strahlhofer, J.; Schild-Prüfert, K.; et al. Enrichment of Human IgA-Coated Bacterial Vesicles in Ulcerative Colitis as a Driver of Inflammation. Nat. Commun. 2025, 16, 3995. [Google Scholar] [CrossRef] [PubMed]
  31. Huang, X.; Xie, M.; Wang, Y.; Lu, X.; Mei, F.; Zhang, K.; Yang, X.; Chen, G.; Yin, Y.; Feng, G.; et al. Porphyromonas gingivalis Aggravates Atherosclerotic Plaque Instability by Promoting Lipid-Laden Macrophage Necroptosis. Signal Transduct. Target. Ther. 2025, 10, 171. [Google Scholar] [CrossRef] [PubMed]
  32. Kato, S.; Kowashi, Y.; Demuth, D.R. Outer Membrane-like Vesicles Secreted by Actinobacillus actinomycetemcomitans Are Enriched in Leukotoxin. Microb. Pathog. 2002, 32, 1–13. [Google Scholar] [CrossRef]
  33. Wang, S.; Hu, Y.; Song, P.; Lin, Q.; Tang, X.; Gao, Q.; Wang, J.; Zhou, D.; Li, J.; Huang, D.; et al. Harnessing Extracellular Vesicles from Lactobacillus reuteri and Lactobacillus paracasei for Synergistic Osteoporosis Therapy. Compos. Part B Eng. 2025, 297, 112255. [Google Scholar] [CrossRef]
  34. Xie, J.; Li, Q.; Haesebrouck, F.; Van Hoecke, L.; Vandenbroucke, R.E. The Tremendous Biomedical Potential of Bacterial Extracellular Vesicles. Trends Biotechnol. 2022, 40, 1173–1194. [Google Scholar] [CrossRef]
  35. Uemura, Y.; Hiroshima, Y.; Tada, A.; Murakami, K.; Yoshida, K.; Inagaki, Y.; Kuwahara, T.; Murakami, A.; Fujii, H.; Yumoto, H. Porphyromonas gingivalis Outer Membrane Vesicles Stimulate Gingival Epithelial Cells to Induce Pro-Inflammatory Cytokines via the MAPK and STING Pathways. Biomedicines 2022, 10, 2643. [Google Scholar] [CrossRef]
  36. Kim, H.Y.; Lim, Y.; An, S.-J.; Choi, B.-K. Characterization and Immunostimulatory Activity of Extracellular Vesicles from Filifactor alocis. Mol. Oral Microbiol. 2020, 35, 1–9. [Google Scholar] [CrossRef]
  37. Haugsten, H.R.; Kristoffersen, A.K.; Haug, T.M.; Søland, T.M.; Øvstebø, R.; Aass, H.C.D.; Enersen, M.; Galtung, H.K. Isolation, Characterization, and Fibroblast Uptake of Bacterial Extracellular Vesicles from Porphyromonas gingivalis Strains. Microbiologyopen 2023, 12, e1388. [Google Scholar] [CrossRef]
  38. du Teil Espina, M.; Fu, Y.; van der Horst, D.; Hirschfeld, C.; López-Álvarez, M.; Mulder, L.M.; Gscheider, C.; Haider Rubio, A.; Huitema, M.; Becher, D.; et al. Coating and Corruption of Human Neutrophils by Bacterial Outer Membrane Vesicles. Microbiol. Spectr. 2022, 10, e0075322. [Google Scholar] [CrossRef] [PubMed]
  39. Fu, Y.; Trautwein-Schult, A.; Piersma, S.; Sun, C.; Westra, J.; de Jong, A.; Becher, D.; van Dijl, J.M. Characterization of Outer Membrane Vesicles of Aggregatibacter actinomycetemcomitans Serotypes a, b and c and Their Interactions with Human Neutrophils. Int. J. Med. Microbiol. 2025, 319, 151655. [Google Scholar] [CrossRef]
  40. Vitkov, L.; Krunić, J.; Dudek, J.; Bobbili, M.R.; Grillari, J.; Hausegger, B.; Mladenović, I.; Stojanović, N.; Krautgartner, W.D.; Oberthaler, H.; et al. Vesicular Messages from Dental Biofilms for Neutrophils. Int. J. Mol. Sci. 2024, 25, 3314. [Google Scholar] [CrossRef]
  41. Fleetwood, A.J.; Lee, M.K.S.; Singleton, W.; Achuthan, A.; Lee, M.-C.; O’Brien-Simpson, N.M.; Cook, A.D.; Murphy, A.J.; Dashper, S.G.; Reynolds, E.C.; et al. Metabolic Remodeling, Inflammasome Activation, and Pyroptosis in Macrophages Stimulated by Porphyromonas gingivalis and Its Outer Membrane Vesicles. Front. Cell. Infect. Microbiol. 2017, 7, 351. [Google Scholar] [CrossRef] [PubMed]
  42. Han, E.-C.; Choi, S.-Y.; Lee, Y.; Park, J.-W.; Hong, S.-H.; Lee, H.-J. Extracellular RNAs in Periodontopathogenic Outer Membrane Vesicles Promote TNF-α Production in Human Macrophages and Cross the Blood-Brain Barrier in Mice. FASEB J. 2019, 33, 13412–13422. [Google Scholar] [CrossRef]
  43. 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] [PubMed]
  44. Elsayed, R.; Elashiry, M.; Liu, Y.; El-Awady, A.; Hamrick, M.; Cutler, C.W. Porphyromonas gingivalis Provokes Exosome Secretion and Paracrine Immune Senescence in Bystander Dendritic Cells. Front. Cell. Infect. Microbiol. 2021, 11, 669989. [Google Scholar] [CrossRef]
  45. 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]
  46. Song, M.-K.; Kim, H.Y.; Choi, B.-K.; Kim, H.-H. Filifactor alocis-Derived Extracellular Vesicles Inhibit Osteogenesis through TLR2 Signaling. Mol. Oral Microbiol. 2020, 35, 202–210. [Google Scholar] [CrossRef]
  47. Kim, H.Y.; Song, M.-K.; Lim, Y.; Jang, J.S.; An, S.-J.; Kim, H.-H.; Choi, B.-K. Effects of Extracellular Vesicles Derived from Oral Bacteria on Osteoclast Differentiation and Activation. Sci. Rep. 2022, 12, 14239. [Google Scholar] [CrossRef] [PubMed]
  48. Ma, X.; Shin, Y.-J.; Yoo, J.-W.; Park, H.-S.; Kim, D.-H. Extracellular Vesicles Derived from Porphyromonas gingivalis Induce Trigeminal Nerve-Mediated Cognitive Impairment. J. Adv. Res. 2023, 54, 293–303. [Google Scholar] [CrossRef]
  49. Nonaka, S.; Kadowaki, T.; Nakanishi, H. Secreted Gingipains from Porphyromonas gingivalis Increase Permeability in Human Cerebral Microvascular Endothelial Cells through Intracellular Degradation of Tight Junction Proteins. Neurochem. Int. 2022, 154, 105282. [Google Scholar] [CrossRef]
  50. Ha, J.Y.; Seok, J.; Kim, S.-J.; Jung, H.-J.; Ryu, K.-Y.; Nakamura, M.; Jang, I.-S.; Hong, S.-H.; Lee, Y.; Lee, H.-J. Periodontitis Promotes Bacterial Extracellular Vesicle-Induced Neuroinflammation in the Brain and Trigeminal Ganglion. PLoS Pathog. 2023, 19, e1011743. [Google Scholar] [CrossRef]
  51. Ha, J.Y.; Choi, S.-Y.; Lee, J.H.; Hong, S.-H.; Lee, H.-J. Delivery of Periodontopathogenic Extracellular Vesicles to Brain Monocytes and Microglial IL-6 Promotion by RNA Cargo. Front. Mol. Biosci. 2020, 7, 596366. [Google Scholar] [CrossRef]
  52. Grandi, A.; Tomasi, M.; Zanella, I.; Ganfini, L.; Caproni, E.; Fantappiè, L.; Irene, C.; Frattini, L.; Isaac, S.J.; König, E.; et al. Synergistic Protective Activity of Tumor-Specific Epitopes Engineered in Bacterial Outer Membrane Vesicles. Front. Oncol. 2017, 7, 253. [Google Scholar] [CrossRef] [PubMed]
  53. Farrugia, C.; Stafford, G.P.; Murdoch, C. Porphyromonas gingivalis Outer Membrane Vesicles Increase Vascular Permeability. J. Dent. Res. 2020, 99, 1494–1501. [Google Scholar] [CrossRef] [PubMed]
  54. Huang, S.; Cao, G.; Dai, D.; Xu, Q.; Ruiz, S.; Shindo, S.; Nakamura, S.; Kawai, T.; Lin, J.; Han, X. Porphyromonas gingivalis Outer Membrane Vesicles Exacerbate Retinal Microvascular Endothelial Cell Dysfunction in Diabetic Retinopathy. Front. Microbiol. 2023, 14, 1167160. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, H.Y.; Song, M.-K.; Gho, Y.S.; Kim, H.-H.; Choi, B.-K. Extracellular Vesicles Derived from the Periodontal Pathogen Filifactor alocis Induce Systemic Bone Loss through Toll-like Receptor 2. J. Extracell. Vesicles 2021, 10, e12157. [Google Scholar] [CrossRef]
  56. He, Y.; Shiotsu, N.; Uchida-Fukuhara, Y.; Guo, J.; Weng, Y.; Ikegame, M.; Wang, Z.; Ono, K.; Kamioka, H.; Torii, Y.; et al. Outer Membrane Vesicles Derived from Porphyromonas gingivalis Induced Cell Death with Disruption of Tight Junctions in Human Lung Epithelial Cells. Arch. Oral Biol. 2020, 118, 104841. [Google Scholar] [CrossRef]
  57. Seyama, M.; Yoshida, K.; Yoshida, K.; Fujiwara, N.; Ono, K.; Eguchi, T.; Kawai, H.; Guo, J.; Weng, Y.; Haoze, Y.; et al. Outer Membrane Vesicles of Porphyromonas gingivalis Attenuate Insulin Sensitivity by Delivering Gingipains to the Liver. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165731. [Google Scholar] [CrossRef]
  58. Lara, B.; Loureiro, I.; Gliosca, L.; Castagnola, L.; Merech, F.; Gallino, L.; Calo, G.; Sassot, M.; Ramhorst, R.; Vota, D.; et al. Porphyromonas gingivalis Outer Membrane Vesicles Shape Trophoblast Cell Metabolism Impairing Functions Associated to Adverse Pregnancy Outcome. J. Cell. Physiol. 2023, 238, 2679–2691. [Google Scholar] [CrossRef]
  59. Luo, R.; Chang, Y.; Liang, H.; Zhang, W.; Song, Y.; Li, G.; Yang, C. Interactions between Extracellular Vesicles and Microbiome in Human Diseases: New Therapeutic Opportunities. iMeta 2023, 2, e86. [Google Scholar] [CrossRef] [PubMed]
  60. Davidson, S.M.; Boulanger, C.M.; Aikawa, E.; Badimon, L.; Barile, L.; Binder, C.J.; Brisson, A.; Buzas, E.; Emanueli, C.; Jansen, F.; et al. Methods for the Identification and Characterization of Extracellular Vesicles in Cardiovascular Studies: From Exosomes to Microvesicles. Cardiovasc. Res. 2023, 119, 45–63. [Google Scholar] [CrossRef]
  61. Qian, L.; Chen, P.; Zhang, S.; Wang, Z.; Guo, Y.; Koutouratsas, V.; Fleishman, J.S.; Huang, C.; Zhang, S. The Uptake of Extracellular Vesicles: Research Progress in Cancer Drug Resistance and Beyond. Drug Resist. Updat. 2025, 79, 101209. [Google Scholar] [CrossRef]
  62. Dixson, A.C.; Dawson, T.R.; Di Vizio, D.; Weaver, A.M. Context-Specific Regulation of Extracellular Vesicle Biogenesis and Cargo Selection. Nat. Rev. Mol. Cell Biol. 2023, 24, 454–476. [Google Scholar] [CrossRef]
  63. Zadka, Ł.; Eggerstorfer, B.; Buzalewicz, I.; Vraka, C.; Rusak, A.; Godbersen, G.M.; Opalińska, A.; Unterholzner, J.; Ulatowska-Jarża, A.; Philippe, C.; et al. Phenotyping Extracellular Vesicles and Their Serotonin Transporter Cargo in Major Depressive Disorder. J. Affect. Disord. 2025, 389, 119740. [Google Scholar] [CrossRef]
  64. Takakura, Y.; Hanayama, R.; Akiyoshi, K.; Futaki, S.; Hida, K.; Ichiki, T.; Ishii-Watabe, A.; Kuroda, M.; Maki, K.; Miura, Y.; et al. Quality and Safety Considerations for Therapeutic Products Based on Extracellular Vesicles. Pharm. Res. 2024, 41, 1573–1594. [Google Scholar] [CrossRef]
  65. Cai, X.-Y.; Zheng, C.-X.; Guo, H.; Fan, S.-Y.; Huang, X.-Y.; Chen, J.; Liu, J.-X.; Gao, Y.-R.; Liu, A.-Q.; Liu, J.-N.; et al. Inflammation-Triggered Gli1+ Stem Cells Engage with Extracellular Vesicles to Prime Aberrant Neutrophils to Exacerbate Periodontal Immunopathology. Cell. Mol. Immunol. 2025, 22, 371–389. [Google Scholar] [CrossRef]
  66. Wu, Y.; Li, J.; Liu, M.; Gao, R.; Zhou, H.; Hu, Q.; Zhao, L.; Xie, Y. Extracellular Vesicles From LPS-Treated PDLSCs Induce NLRP3 Inflammasome Activation in Periodontitis. Oral Dis. 2025, 31, 1277–1289. [Google Scholar] [CrossRef]
  67. Wang, Y.; Zhang, X.; Wang, J.; Zhang, Y.; Ye, Q.; Wang, Y.; Fei, D.; Wang, Q. Inflammatory Periodontal Ligament Stem Cells Drive M1 Macrophage Polarization via Exosomal miR-143-3p-Mediated Regulation of PI3K/AKT/NF-κB Signaling. Stem Cells 2023, 41, 184–199. [Google Scholar] [CrossRef]
  68. Cui, S.; Zhang, Z.; Cheng, C.; Tang, S.; Zhai, M.; Li, L.; Wei, F.; Ding, G. Small Extracellular Vesicles from Periodontal Ligament Stem Cells Primed by Lipopolysaccharide Regulate Macrophage M1 Polarization via miR-433-3p Targeting TLR2/TLR4/NF-κB. Inflammation 2023, 46, 1849–1858. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, Z.; Wang, P.; Zheng, Y.; Wang, M.; Chou, J.; Wang, Z. Exosomal microRNA-223 from Neutrophil-like Cells Inhibits Osteogenic Differentiation of PDLSCs through the cGMP-PKG Signaling Pathway. J. Periodontal Res. 2023, 58, 1315–1325. [Google Scholar] [CrossRef] [PubMed]
  70. Yan, J.; Yang, T.; Ma, S.; Li, D.; Hu, C.; Tan, J. Macrophage-Derived Mitochondria-Rich Extracellular Vesicles Aggravate Bone Loss in Periodontitis by Disrupting the Mitochondrial Dynamics of BMSCs. J. Nanobiotechnol. 2025, 23, 208. [Google Scholar] [CrossRef] [PubMed]
  71. Pan, L.; Zhang, C.; Zhang, H.; Ke, T.; Bian, M.; Yang, Y.; Chen, L.; Tan, J. Osteoclast-Derived Exosomal miR-5134-5p Interferes with Alveolar Bone Homeostasis by Targeting the JAK2/STAT3 Axis. Int. J. Nanomed. 2023, 18, 3727–3744. [Google Scholar] [CrossRef] [PubMed]
  72. Dai, Z.; Zheng, W.; Li, S. Receptor Activator of Nuclear Factor-κB Ligand and Tumor Necrosis Factor-α Promotes Osteoclast Differentiation through the Exosomes of Inflammatory Periodontal Ligament Stem Cells. Hua Xi Kou Qiang Yi Xue Za Zhi 2022, 40, 377–385. [Google Scholar] [CrossRef]
  73. Zhang, C.; Pan, L.; Zhang, H.; Ke, T.; Yang, Y.; Zhang, L.; Chen, L.; Tan, J. Osteoblasts-Derived Exosomal lncRNA-MALAT1 Promotes Osteoclastogenesis by Targeting the miR-124/NFATc1 Signaling Axis in Bone Marrow-Derived Macrophages. Int. J. Nanomed. 2023, 18, 781–795. [Google Scholar] [CrossRef]
  74. Dong, J.-C.; Liao, Y.; Zhou, W.; Sun, M.-J.; Zhang, H.-Y.; Li, Y.; Song, Z.-C. Porphyromonas Gingivalis LPS-Stimulated BMSC-Derived Exosome Promotes Osteoclastogenesis via miR-151-3p/PAFAH1B1. Oral Dis. 2025, 31, 206–216. [Google Scholar] [CrossRef]
  75. Chen, S.; Liu, J.; Zhu, L. M2-like Macrophage-Derived Exosomes Inhibit Osteoclastogenesis via Releasing miR-1227-5p. Immunobiology 2025, 230, 152861. [Google Scholar] [CrossRef] [PubMed]
  76. Ding, C.; Shen, Z.; Xu, R.; Liu, Y.; Xu, M.; Fan, C.; Hu, D.; Xing, T. Exosomes Derived from Periodontitis Induce Hepatic Steatosis through the SCD-1/AMPK Signaling Pathway. Biochim. Biophys. Acta Mol. Basis Dis. 2024, 1870, 167343. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, Y.; Cui, B.; Zhang, P.; Xiao, S.; Duan, D.; Ding, Y. Polymicrobial Infection Induces Adipose Tissue Dysfunction via Gingival Extracellular Vesicles. J. Dent. Res. 2024, 103, 187–196. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, F.; Peng, L.; Gu, F.; Huang, P.; Cheng, B.; Chen, G.; Meng, L.; Bian, Z. Circulating Small Extracellular Vesicles from Patients with Periodontitis Contribute to Development of Insulin Resistance. J. Periodontol. 2022, 93, 1902–1915. [Google Scholar] [CrossRef]
  79. Li, J.; Guo, Y.; Chen, Y.-Y.; Liu, Q.; Chen, Y.; Tan, L.; Zhang, S.-H.; Gao, Z.-R.; Zhou, Y.-H.; Zhang, G.-Y.; et al. miR-124-3p Increases in High Glucose Induced Osteocyte-Derived Exosomes and Regulates Galectin-3 Expression: A Possible Mechanism in Bone Remodeling Alteration in Diabetic Periodontitis. FASEB J. 2020, 34, 14234–14249. [Google Scholar] [CrossRef]
  80. Yang, W.-W.; Li, Q.-X.; Wang, F.; Zhang, X.-R.; Zhang, X.-L.; Wang, M.; Xue, D.; Zhao, Y.; Tang, L. Exosomal miR-155-5p Promote the Occurrence of Carotid Atherosclerosis. J. Cell. Mol. Med. 2024, 28, e70187. [Google Scholar] [CrossRef]
  81. Tanai, A.; Fukuhara, Y.; Eguchi, T.; Kawai, H.; Ueda, K.; Ochiai, K.; Ikegame, M.; Okamoto, K.; Okamura, H.P. Gingivalis-Infected Macrophage Extracellular Vesicles Cause Adverse Pregnancy Outcomes. J. Dent. Res. 2025, 104, 54–63. [Google Scholar] [CrossRef]
  82. Song, X.; Xue, Y.; Fan, S.; Hao, J.; Deng, R. Lipopolysaccharide-Activated Macrophages Regulate the Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells through Exosomes. PeerJ 2022, 10, e13442. [Google Scholar] [CrossRef]
  83. Liu, W.; An, J.; Jiao, C.; Guo, J.; Zhang, L.; Zhang, Z.; Liu, G.; Zhang, Y. Novel Insights into the Association of ZJU Index with Periodontitis in US Participants: A Cross-Sectional Study. Lipids Health Dis. 2025, 24, 240. [Google Scholar] [CrossRef]
  84. Chen, Z.; Deng, H.; Sun, K.; Huang, Z.; Wei, S.; Lin, Y.; Song, Z.; Liu, Y. Prevalence of Chronic Periodontitis in Patients Undergoing Peritoneal Dialysis and Its Correlation with Peritoneal Dialysis-Related Complications. BMC Nephrol. 2023, 24, 71. [Google Scholar] [CrossRef]
  85. Jafari, N.; Llevenes, P.; Denis, G.V. Exosomes as Novel Biomarkers in Metabolic Disease and Obesity-Related Cancers. Nat. Rev. Endocrinol. 2022, 18, 327–328. [Google Scholar] [CrossRef]
  86. Deng, C.; Huo, M.; Chu, H.; Zhuang, X.; Deng, G.; Li, W.; Wei, H.; Zeng, L.; He, Y.; Liu, H.; et al. Exosome circATP8A1 Induces Macrophage M2 Polarization by Regulating the miR-1-3p/STAT6 Axis to Promote Gastric Cancer Progression. Mol. Cancer 2024, 23, 49. [Google Scholar] [CrossRef]
  87. Lin, Y.; Dong, H.; Deng, W.; Lin, W.; Li, K.; Xiong, X.; Guo, Y.; Zhou, F.; Ma, C.; Chen, Y.; et al. Evaluation of Salivary Exosomal Chimeric GOLM1-NAA35 RNA as a Potential Biomarker in Esophageal Carcinoma. Clin. Cancer Res. 2019, 25, 3035–3045. [Google Scholar] [CrossRef] [PubMed]
  88. Yang, C.; Chen, J.; Zhao, Y.; Wu, J.; Xu, Y.; Xu, J.; Chen, F.; Chen, Y.; Chen, N. Salivary Exosomes Exacerbate Colitis by Bridging the Oral Cavity and Intestine. iScience 2024, 27, 111061. [Google Scholar] [CrossRef]
  89. Chai, Z.; Yu, Y.; Cheng, P.; Zhao, L.; Petrovic, B.; Li, A.; Xu, F.; Li, Y.; You, M. Recent Advances of Oral Fluids-Based Point-of-Care Testing Platforms for Oral Disease Diagnosis. Transl. Dent. Res. 2025, 1, 100003. [Google Scholar] [CrossRef]
  90. Li, K.; Lin, Y.; Luo, Y.; Xiong, X.; Wang, L.; Durante, K.; Li, J.; Zhou, F.; Guo, Y.; Chen, S.; et al. A Signature of Saliva-Derived Exosomal Small RNAs as Predicting Biomarker for Esophageal Carcinoma: A Multicenter Prospective Study. Mol. Cancer 2022, 21, 21. [Google Scholar] [CrossRef] [PubMed]
  91. Li, K.; Lin, Y.; Zhou, Y.; Xiong, X.; Wang, L.; Li, J.; Zhou, F.; Guo, Y.; Chen, S.; Chen, Y.; et al. Salivary Extracellular MicroRNAs for Early Detection and Prognostication of Esophageal Cancer: A Clinical Study. Gastroenterology 2023, 165, 932–945.e9. [Google Scholar] [CrossRef]
  92. Chaparro Padilla, A.; Weber Aracena, L.; Realini Fuentes, O.; Albers Busquetts, D.; Hernández Ríos, M.; Ramírez Lobos, V.; Pascual La Rocca, A.; Nart Molina, J.; Beltrán Varas, V.; Acuña-Gallardo, S.; et al. Molecular Signatures of Extracellular Vesicles in Oral Fluids of Periodontitis Patients. Oral Dis. 2020, 26, 1318–1325. [Google Scholar] [CrossRef]
  93. Fatima, T.; Khurshid, Z.; Rehman, A.; Imran, E.; Srivastava, K.C.; Shrivastava, D. Gingival Crevicular Fluid (GCF): A Diagnostic Tool for the Detection of Periodontal Health and Diseases. Molecules 2021, 26, 1208. [Google Scholar] [CrossRef] [PubMed]
  94. Han, P.; Bartold, P.M.; Ivanovski, S. The Emerging Role of Small Extracellular Vesicles in Saliva and Gingival Crevicular Fluid as Diagnostics for Periodontitis. J. Periodontal Res. 2022, 57, 219–231. [Google Scholar] [CrossRef]
  95. Mizgier, M.L.; Nardocci, G.; Ramírez, V.; Bendek, M.J.; Hernández, M.; Rojas, C.; Herrera, D.; Kantarci, A.; Kemp, M.W.; Illanes, S.E.; et al. Proteomic Insights Into Gingival Crevicular Extracellular Vesicles in Periodontitis and Gestational Diabetes: An Exploratory Study. J. Clin. Periodontol. 2025, 52, 225–236. [Google Scholar] [CrossRef] [PubMed]
  96. Yamaguchi, A.; Tsuruya, Y.; Igarashi, K.; Jin, Z.; Yamazaki-Takai, M.; Takai, H.; Nakayama, Y.; Ogata, Y. Changes in the Components of Salivary Exosomes Due to Initial Periodontal Therapy. J. Periodontal Implant. Sci. 2023, 53, 347–361. [Google Scholar] [CrossRef] [PubMed]
  97. Nik Mohamed Kamal, N.N.S.; Awang, R.A.R.; Mohamad, S.; Shahidan, W.N.S. Plasma- and Saliva Exosome Profile Reveals a Distinct MicroRNA Signature in Chronic Periodontitis. Front. Physiol. 2020, 11, 587381. [Google Scholar] [CrossRef]
  98. Han, P.; Bartold, P.M.; Salomon, C.; Ivanovski, S. Salivary Small Extracellular Vesicles Associated miRNAs in Periodontal Status—A Pilot Study. Int. J. Mol. Sci. 2020, 21, 2809. [Google Scholar] [CrossRef]
  99. Xia, Y.; Zhou, K.; Sun, M.; Shu, R.; Qian, J.; Xie, Y. The miR-223-3p Regulates Pyroptosis Through NLRP3-Caspase 1-GSDMD Signal Axis in Periodontitis. Inflammation 2021, 44, 2531–2542. [Google Scholar] [CrossRef]
  100. Yu, J.; Lin, Y.; Xiong, X.; Li, K.; Yao, Z.; Dong, H.; Jiang, Z.; Yu, D.; Yeung, S.-C.J.; Zhang, H. Detection of Exosomal PD-L1 RNA in Saliva of Patients with Periodontitis. Front. Genet. 2019, 10, 202. [Google Scholar] [CrossRef]
  101. Han, P.; Jiao, K.; Moran, C.S.; Liaw, A.; Zhou, Y.; Salomon, C.; Ivanovski, S. TNF-α and OSX mRNA of Salivary Small Extracellular Vesicles in Periodontitis: A Pilot Study. Tissue Eng. Part C Methods 2023, 29, 298–306. [Google Scholar] [CrossRef]
  102. Huang, X.; Hu, X.; Zhao, M.; Zhang, Q. Analysis of Salivary Exosomal Proteins in Young Adults with Severe Periodontitis. Oral Dis. 2020, 26, 173–181. [Google Scholar] [CrossRef]
  103. Liu, C.; Seneviratne, C.J.; Palma, C.; Rice, G.; Salomon, C.; Khanabdali, R.; Ivanovski, S.; Han, P. Immunoaffinity-Enriched Salivary Small Extracellular Vesicles in Periodontitis. Extracell. Vesicles Circ. Nucl. Acids 2023, 4, 698–712. [Google Scholar] [CrossRef]
  104. 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]
  105. Tobón-Arroyave, S.I.; Celis-Mejía, N.; Córdoba-Hidalgo, M.P.; Isaza-Guzmán, D.M. Decreased Salivary Concentration of CD9 and CD81 Exosome-Related Tetraspanins May Be Associated with the Periodontal Clinical Status. J. Clin. Periodontol. 2019, 46, 470–480. [Google Scholar] [CrossRef]
  106. 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]
  107. Liu, Y.; Li, H.; Li, Y.; Wang, D.; Chen, J.; Yuan, Z.; Zhang, R.; Zhang, M.; Cai, Z.; Hou, R.; et al. Giant Panda Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Promote Dermal Fibroblast Proliferation and Wound Healing. Stem Cells 2025, sxaf051. [Google Scholar] [CrossRef]
  108. Qiao, X.; Tang, J.; Dou, L.; Yang, S.; Sun, Y.; Mao, H.; Yang, D. Dental Pulp Stem Cell-Derived Exosomes Regulate Anti-Inflammatory and Osteogenesis in Periodontal Ligament Stem Cells and Promote the Repair of Experimental Periodontitis in Rats. Int. J. Nanomed. 2023, 18, 4683–4703. [Google Scholar] [CrossRef] [PubMed]
  109. Zhou, F.; Guo, J.; Dong, Z.; Zhao, W.; He, X.; Wu, M.; Li, S.; Li, B.; Zhou, M. SHED Aggregate Derived Exosomes-Shuttled miR-222 Promotes the Angiogenic Properties of Periodontal Ligament Stem Cells and Enhances Periodontal Regeneration. Transl. Dent. Res. 2025, 1, 100032. [Google Scholar] [CrossRef]
  110. Ma, S.; Li, S.; Zhang, Y.; Nie, J.; Cao, J.; Li, A.; Li, Y.; Pei, D. BMSC-Derived Exosomal CircHIPK3 Promotes Osteogenic Differentiation of MC3T3-E1 Cells via Mitophagy. Int. J. Mol. Sci. 2023, 24, 2785. [Google Scholar] [CrossRef] [PubMed]
  111. Jing, L.; Wang, H.-Y.; Zhang, N.; Zhang, W.-J.; Chen, Y.; Deng, D.-K.; Li, X.; Chen, F.-M.; He, X.-T. Critical Roles of Extracellular Vesicles in Periodontal Disease and Regeneration. Stem Cells Transl. Med. 2025, 14, szae092. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, L.; Li, X.; Zhang, B.; Li, R. Extracellular Vesicles in Periodontitis: Pathogenic Mechanisms and Therapeutic Potential. J. Inflamm. Res. 2025, 18, 1317–1331. [Google Scholar] [CrossRef] [PubMed]
  113. Cai, R.; Wang, L.; Zhang, W.; Liu, B.; Wu, Y.; Pang, J.; Ma, C. The Role of Extracellular Vesicles in Periodontitis: Pathogenesis, Diagnosis, and Therapy. Front. Immunol. 2023, 14, 1151322. [Google Scholar] [CrossRef]
  114. Teng, X.; Liu, T.; Zhao, G.; Liang, Y.; Li, P.; Li, F.; Li, Q.; Fu, J.; Zhong, C.; Zou, X.; et al. A Novel Exosome-Based Multifunctional Nanocomposite Platform Driven by Photothermal-Controlled Release System for Repair of Skin Injury. J. Control. Release 2024, 371, 258–272. [Google Scholar] [CrossRef]
  115. Zhao, H.; Li, Z.; Liu, D.; Zhang, J.; You, Z.; Shao, Y.; Li, H.; Yang, J.; Liu, X.; Wang, M.; et al. PlexinA1 (PLXNA1) as a Novel Scaffold Protein for the Engineering of Extracellular Vesicles. J. Extracell. Vesicles 2024, 13, e70012. [Google Scholar] [CrossRef] [PubMed]
  116. Mondal, J.; Pillarisetti, S.; Junnuthula, V.; Saha, M.; Hwang, S.R.; Park, I.-K.; Lee, Y.-K. Hybrid Exosomes, Exosome-like Nanovesicles and Engineered Exosomes for Therapeutic Applications. J. Control. Release 2023, 353, 1127–1149. [Google Scholar] [CrossRef]
  117. Xiang, M.; Liu, Y.; Guo, Q.; Liao, C.; Xiao, L.; Xiang, M.; Guan, X.; Liu, J. Metformin Enhances the Therapeutic Effects of Extracellular Vesicles Derived from Human Periodontal Ligament Stem Cells on Periodontitis. Sci. Rep. 2024, 14, 19940. [Google Scholar] [CrossRef]
  118. Yu, J.; Wu, X.; Zhang, W.; Chu, F.; Zhang, Q.; Gao, M.; Xu, Y.; Wu, Y. Effect of Psoralen on the Regulation of Osteogenic Differentiation Induced by Periodontal Stem Cell-Derived Exosomes. Hum. Cell 2023, 36, 1389–1402. [Google Scholar] [CrossRef]
  119. Dai, Z.; Li, Z.; Zheng, W.; Yan, Z.; Zhang, L.; Yang, J.; Xiao, J.; Sun, H.; Li, S.; Huang, W. Gallic Acid Ameliorates the Inflammatory State of Periodontal Ligament Stem Cells and Promotes Pro-Osteodifferentiation Capabilities of Inflammatory Stem Cell-Derived Exosomes. Life 2022, 12, 1392. [Google Scholar] [CrossRef]
  120. Huang, Y.; Li, M.; Liu, Q.; Song, L.; Wang, Q.; Ding, P.; Tian, W.; Guo, S. Small Extracellular Vesicles Derived from Lipopolysaccharide-Preconditioned Dental Follicle Cells Inhibit Cell Apoptosis and Alveolar Bone Loss in Periodontitis. Arch. Oral Biol. 2024, 162, 105964. [Google Scholar] [CrossRef]
  121. 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]
  122. Niu, Q.; Lin, C.; Yang, S.; Rong, S.; Wei, J.; Zhao, T.; Peng, Y.; Cheng, Z.; Xie, Y.; Wang, Y. FoxO1-Overexpressed Small Extracellular Vesicles Derived from hPDLSCs Promote Periodontal Tissue Regeneration by Reducing Mitochondrial Dysfunction to Regulate Osteogenesis and Inflammation. Int. J. Nanomed. 2024, 19, 8751–8768. [Google Scholar] [CrossRef]
  123. Luo, H.; Chen, D.; Li, R.; Li, R.; Teng, Y.; Cao, Y.; Zou, X.; Wang, W.; Zhou, C. Genetically Engineered CXCR4-Modified Exosomes for Delivery of miR-126 Mimics to Macrophages Alleviate Periodontitis. J. Nanobiotechnol. 2023, 21, 116. [Google Scholar] [CrossRef]
  124. 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]
  125. Lai, S.; Deng, L.; Liu, C.; Li, X.; Fan, L.; Zhu, Y.; Yang, Y.; Mu, Y. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Loaded with miR-26a through the Novel Immunomodulatory Peptide DP7-C Can Promote Osteogenesis. Biotechnol. Lett. 2023, 45, 905–919. [Google Scholar] [CrossRef] [PubMed]
  126. Lai, S.; Tang, N.; Guo, J.; Deng, L.; Yuan, L.; Zeng, L.; Yang, L.; Mu, Y. Immunomodulatory Peptide DP7-C Mediates Macrophage-Derived Exosomal miR-21b to Promote Bone Regeneration via the SOCS1/JAK2/STAT3 Axis. Colloids Surf. B Biointerfaces 2025, 253, 114709. [Google Scholar] [CrossRef]
  127. Zhang, Y.; Chen, J.; Fu, H.; Kuang, S.; He, F.; Zhang, M.; Shen, Z.; Qin, W.; Lin, Z.; Huang, S. Exosomes Derived from 3D-Cultured MSCs Improve Therapeutic Effects in Periodontitis and Experimental Colitis and Restore the Th17 Cell/Treg Balance in Inflamed Periodontium. Int. J. Oral Sci. 2021, 13, 43. [Google Scholar] [CrossRef]
  128. Zhang, T.; Chen, Z.; Zhu, M.; Jing, X.; Xu, X.; Yuan, X.; Zhou, M.; Zhang, Y.; Lu, M.; Chen, D.; et al. Extracellular Vesicles Derived from Human Dental Mesenchymal Stem Cells Stimulated with Low-Intensity Pulsed Ultrasound Alleviate Inflammation-Induced Bone Loss in a Mouse Model of Periodontitis. Genes. Dis. 2023, 10, 1613–1625. [Google Scholar] [CrossRef]
  129. Wu, P.; Zhang, B.; Ocansey, D.K.W.; Xu, W.; Qian, H. Extracellular Vesicles: A Bright Star of Nanomedicine. Biomaterials 2021, 269, 120467. [Google Scholar] [CrossRef]
  130. Zhao, M.; Li, Q.; Chai, Y.; Rong, R.; He, L.; Zhang, Y.; Cui, H.; Xu, H.; Zhang, X.; Wang, Z.; et al. An Anti-CD19-Exosome Delivery System Navigates the Blood-Brain Barrier for Targeting of Central Nervous System Lymphoma. J. Nanobiotechnol. 2025, 23, 173. [Google Scholar] [CrossRef]
  131. Mehryab, F.; Rabbani, S.; Shahhosseini, S.; Shekari, F.; Fatahi, Y.; Baharvand, H.; Haeri, A. Exosomes as a Next-Generation Drug Delivery System: An Update on Drug Loading Approaches, Characterization, and Clinical Application Challenges. Acta Biomater. 2020, 113, 42–62. [Google Scholar] [CrossRef] [PubMed]
  132. Cui, Y.; Hong, S.; Xia, Y.; Li, X.; He, X.; Hu, X.; Li, Y.; Wang, X.; Lin, K.; Mao, L. Melatonin Engineering M2 Macrophage-Derived Exosomes Mediate Endoplasmic Reticulum Stress and Immune Reprogramming for Periodontitis Therapy. Adv. Sci. 2023, 10, e2302029. [Google Scholar] [CrossRef]
  133. Shi, Y.; Zhang, R.; Da, N.; Wang, Y.; Yang, J.; Li, B.; He, X. Aspirin Loaded Extracellular Vesicles Inhibit Inflammation of Macrophages via Switching Metabolic Phenotype in Periodontitis. Biochem. Biophys. Res. Commun. 2023, 667, 25–33. [Google Scholar] [CrossRef]
  134. Yang, J.H.; He, X.N.; Liu, Z.; Wang, W.Z.; Li, B. Study of the methotrexate loaded extracellular vesicles in the treatment of experimental periodontitis in mice. Zhonghua Kou Qiang Yi Xue Za Zhi 2024, 59, 681–689. [Google Scholar] [CrossRef] [PubMed]
  135. Liu, Z.; Yang, J.; Tan, G.; Shi, Y.; Tao, D.; Wang, W.; Li, B.; Jin, F.; He, X. Methotrexate Loaded Extracellular Vesicles Attenuate Periodontitis by Suppressing ACSL1 and Promoting Anti-Inflammatory Macrophage. Mol. Immunol. 2025, 182, 83–95. [Google Scholar] [CrossRef]
  136. 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]
  137. Liu, S.; Wang, Z.; Li, Y.; Pan, Z.; Huang, L.; Cui, J.; Zhang, X.; Yang, M.; Zhang, Y.; Li, D.; et al. Erythropoietin-Stimulated Macrophage-Derived Extracellular Vesicles in Chitosan Hydrogel Rescue BMSCs Fate by Targeting EGFR to Alleviate Inflammatory Bone Loss in Periodontitis. Adv. Sci. 2025, 12, e2500554. [Google Scholar] [CrossRef]
  138. Yu, Y.; Li, X.; Ying, Q.; Zhang, Z.; Liu, W.; Su, J. Synergistic Effects of Shed-Derived Exosomes, Cu2+, and an Injectable Hyaluronic Acid Hydrogel on Antibacterial, Anti-Inflammatory, and Osteogenic Activity for Periodontal Bone Regeneration. ACS Appl. Mater. Interfaces 2024, 16, 33053–33069. [Google Scholar] [CrossRef]
  139. Ming, L.; Qu, Y.; Wang, Z.; Dong, L.; Li, Y.; Liu, F.; Wang, Q.; Zhang, D.; Li, Z.; Zhou, Z.; et al. Small Extracellular Vesicles Laden Oxygen-Releasing Thermosensitive Hydrogel for Enhanced Antibacterial Therapy against Anaerobe-Induced Periodontitis Alveolar Bone Defect. ACS Biomater. Sci. Eng. 2024, 10, 932–945. [Google Scholar] [CrossRef]
  140. 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. Healthc. Mater. 2022, 11, e2102593. [Google Scholar] [CrossRef] [PubMed]
  141. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal Information for Studies of Extracellular Vesicles (MISEV2023): From Basic to Advanced Approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef]
  142. Elieh-Ali-Komi, D.; Shafaghat, F.; Alipoor, S.D.; Kazemi, T.; Atiakshin, D.; Pyatilova, P.; Maurer, M. Immunomodulatory Significance of Mast Cell Exosomes (MC-EXOs) in Immune Response Coordination. Clin. Rev. Allergy Immunol. 2025, 68, 20. [Google Scholar] [CrossRef]
  143. Wang, X.; Huang, J.; Chen, W.; Li, G.; Li, Z.; Lei, J. The Updated Role of Exosomal Proteins in the Diagnosis, Prognosis, and Treatment of Cancer. Exp. Mol. Med. 2022, 54, 1390–1400. [Google Scholar] [CrossRef]
  144. Shen, H.-Y.; Xu, J.-L.; Zhang, W.; Chen, Q.-N.; Zhu, Z.; Mao, Y. Exosomal circRHCG Promotes Breast Cancer Metastasis via Facilitating M2 Polarization through TFEB Ubiquitination and Degradation. NPJ Precis. Oncol. 2024, 8, 22. [Google Scholar] [CrossRef]
  145. Cui, L.; Zheng, J.; Lu, Y.; Lin, P.; Lin, Y.; Zheng, Y.; Xu, R.; Mai, Z.; Guo, B.; Zhao, X. New Frontiers in Salivary Extracellular Vesicles: Transforming Diagnostics, Monitoring, and Therapeutics in Oral and Systemic Diseases. J. Nanobiotechnol. 2024, 22, 171. [Google Scholar] [CrossRef]
  146. Visvanathan, R.; Houghton, M.J.; Barber, E.; Williamson, G. Structure-Function Relationships in (Poly)Phenol-Enzyme Binding: Direct Inhibition of Human Salivary and Pancreatic α-Amylases. Food Res. Int. 2024, 188, 114504. [Google Scholar] [CrossRef]
  147. Hu, S.; Zhang, L.; Su, Y.; Liang, X.; Yang, J.; Luo, Q.; Luo, H. Sensitive Detection of Multiple Blood Biomarkers via Immunomagnetic Exosomal PCR for the Diagnosis of Alzheimer’s Disease. Sci. Adv. 2024, 10, eabm3088. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, Y.; Song, J.; Wang, B.; Wen, Y.; Jiang, W.; Zhang, Y.-L.; Li, Z.-L.; Yu, H.; Qin, S.-F.; Lv, L.-L.; et al. Comprehensive Comparison of Extracellular Vesicles Derived from Mesenchymal Stem Cells Cultured with Fetal Bovine Serum and Human Platelet Lysate. ACS Nano 2025, 19, 12366–12381. [Google Scholar] [CrossRef] [PubMed]
  149. Li, Z.; Bai, Y.; Wu, H.; Feng, Y.; Wang, X.; Zhao, C.; Wang, X. PTEN/PI3K/AKT Pathway Activation with Hypoxia-Induced Human Umbilical Vein Endothelial Cell Exosome for Angiogenesis-Based Diabetic Skin Reconstruction. Mater. Today Bio 2025, 32, 101651. [Google Scholar] [CrossRef] [PubMed]
  150. Yuan, W.; Wu, Y.; Huang, M.; Zhou, X.; Liu, J.; Yi, Y.; Wang, J.; Liu, J. A New Frontier in Temporomandibular Joint Osteoarthritis Treatment: Exosome-Based Therapeutic Strategy. Front. Bioeng. Biotechnol. 2022, 10, 1074536. [Google Scholar] [CrossRef]
  151. Huang, J.; Chen, H.; Li, N.; Liu, P.; Yang, J.; Zhao, Y. Emerging Technologies towards Extracellular Vesicles Large-Scale Production. Bioact. Mater. 2025, 52, 338–365. [Google Scholar] [CrossRef] [PubMed]
  152. Wiest, E.F.; Zubair, A.C. Challenges of Manufacturing Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Regenerative Medicine. Cytotherapy 2020, 22, 606–612. [Google Scholar] [CrossRef] [PubMed]
  153. Wiest, E.F.; Zubair, A.C. Generation of Current Good Manufacturing Practices-Grade Mesenchymal Stromal Cell-Derived Extracellular Vesicles Using Automated Bioreactors. Biology 2025, 14, 313. [Google Scholar] [CrossRef]
Figure 1. Pathogenic roles of BEVs in periodontitis. BEVs derived from periodontal pathogens penetrate epithelial and connective barriers, stimulating neutrophils, macrophages, and dendritic cells to induce excessive inflammatory responses, thereby sustaining chronic inflammation. Furthermore, BEVs perturb osteoimmune homeostasis by inhibiting osteogenesis and enhancing osteoclastogenesis, ultimately leading to progressive alveolar bone loss in periodontitis. Upward arrows represent upregulation, while downward arrows represent downregulation.
Figure 1. Pathogenic roles of BEVs in periodontitis. BEVs derived from periodontal pathogens penetrate epithelial and connective barriers, stimulating neutrophils, macrophages, and dendritic cells to induce excessive inflammatory responses, thereby sustaining chronic inflammation. Furthermore, BEVs perturb osteoimmune homeostasis by inhibiting osteogenesis and enhancing osteoclastogenesis, ultimately leading to progressive alveolar bone loss in periodontitis. Upward arrows represent upregulation, while downward arrows represent downregulation.
Biomedicines 13 02521 g001
Figure 2. Promotional roles of BEVs in periodontitis related systemic diseases. BEVs extend their effects beyond the oral cavity, contributing to neurodegenerative disorders, cardiovascular disease, diabetes, diabetic retinopathy, pulmonary inflammation, systemic bone loss, and adverse pregnancy outcomes. BEVs provide a mechanistic link between periodontitis and multiple systemic conditions, highlighting their significance as both pathogenic mediators and potential therapeutic targets. Upward arrows represent upregulation, while downward arrows represent downregulation.
Figure 2. Promotional roles of BEVs in periodontitis related systemic diseases. BEVs extend their effects beyond the oral cavity, contributing to neurodegenerative disorders, cardiovascular disease, diabetes, diabetic retinopathy, pulmonary inflammation, systemic bone loss, and adverse pregnancy outcomes. BEVs provide a mechanistic link between periodontitis and multiple systemic conditions, highlighting their significance as both pathogenic mediators and potential therapeutic targets. Upward arrows represent upregulation, while downward arrows represent downregulation.
Biomedicines 13 02521 g002
Figure 3. The roles of EVs derived from periodontal cells in the pathogenesis of periodontitis and related systemic diseases. CEVs modulate immune responses, impair osteogenesis, and promote osteoclastogenesis, thereby exacerbating periodontal inflammation and alveolar bone loss. Beyond local effects, CEVs influence systemic pathology by disrupting hepatic metabolism, impairing insulin signaling, increasing vascular permeability and inflammation, and inhibiting placental angiogenesis, linking periodontitis with hepatic injury, diabetes, atherosclerosis, and adverse pregnancy outcomes. Upward arrows represent upregulation, while downward arrows represent downregulation.
Figure 3. The roles of EVs derived from periodontal cells in the pathogenesis of periodontitis and related systemic diseases. CEVs modulate immune responses, impair osteogenesis, and promote osteoclastogenesis, thereby exacerbating periodontal inflammation and alveolar bone loss. Beyond local effects, CEVs influence systemic pathology by disrupting hepatic metabolism, impairing insulin signaling, increasing vascular permeability and inflammation, and inhibiting placental angiogenesis, linking periodontitis with hepatic injury, diabetes, atherosclerosis, and adverse pregnancy outcomes. Upward arrows represent upregulation, while downward arrows represent downregulation.
Biomedicines 13 02521 g003
Figure 4. EVs derived from saliva and GCF as diagnostic biomarkers for periodontitis. GCF-EVs sensitively reflect local inflammation and tissue destruction, while salivary EVs capture both site-specific and systemic changes. Their molecular cargos, including microRNAs, mRNAs, proteins, and surface markers, show significant alterations in periodontitis, highlighting their utility for early detection, staging, and risk assessment.
Figure 4. EVs derived from saliva and GCF as diagnostic biomarkers for periodontitis. GCF-EVs sensitively reflect local inflammation and tissue destruction, while salivary EVs capture both site-specific and systemic changes. Their molecular cargos, including microRNAs, mRNAs, proteins, and surface markers, show significant alterations in periodontitis, highlighting their utility for early detection, staging, and risk assessment.
Biomedicines 13 02521 g004
Figure 5. Main strategies for enhancing periodontal tissue repair with MSC-EVs, paving the way for their clinical application in periodontitis. Pretreatment, genetic modification, and microenvironmental modulation of parental cells improve EV yield and bioactivity, while sonication, electroporation, freeze–thaw, and extrusion facilitate therapeutic cargo loading. Incorporation of MSC-EVs into hydrogels enables prolonged retention, synergistic release with antibacterial agents, and stimulus-responsive delivery, thereby improving therapeutic efficiency and promoting periodontal regeneration.
Figure 5. Main strategies for enhancing periodontal tissue repair with MSC-EVs, paving the way for their clinical application in periodontitis. Pretreatment, genetic modification, and microenvironmental modulation of parental cells improve EV yield and bioactivity, while sonication, electroporation, freeze–thaw, and extrusion facilitate therapeutic cargo loading. Incorporation of MSC-EVs into hydrogels enables prolonged retention, synergistic release with antibacterial agents, and stimulus-responsive delivery, thereby improving therapeutic efficiency and promoting periodontal regeneration.
Biomedicines 13 02521 g005
Table 1. BEVs derived from periodontal pathogens and their roles in periodontitis.
Table 1. BEVs derived from periodontal pathogens and their roles in periodontitis.
BEVs and Periodontitis
EV SourceCargoTarget CellPathwayReadoutsModelStrength of EvidenceRef.
P. gingivalisGingipainsGingival epithelial cellsMAPK(Erk1/2, JNK, p38) and STING↑IL-6 and ↑IL-8In vitro gingival epithelial cells assayIn vitro[35]
F. alocisLipoproteins, FACIN, and autolysinsMonocytes and oral keratinocyte cells/↑CCL1, ↑CCL2, ↑MIP-1, ↑CCL5, ↑CXCL1, ↑CXCL10, ↑ICAM-1, ↑IL-1β, ↑IL-1ra, ↑IL-6, ↑IL-8, ↑MIF, ↑SerpinE, and ↑TNF-α in human monocytes and ↑CXCL1, ↑G-CSF, ↑GM-CSF, ↑IL-6, and ↑IL-8 in human oral keratinocyte cellsIn vitro monocytes and oral keratinocyte cells assayIn vitro[36]
P. gingivalisGingipainsOral fibroblasts/The internalization of Porphyromonas gingivalis bEVs by oral fibroblasts
↓Fibroblast proliferation and growth
In vitro oral gingival fibroblasts assayIn vitro[37]
P. gingivalisGingipainsNeutrophils/↑LL-37, ↑MPOIn vitro neutrophils assayIn vitro[38]
A. actinomycetemcomitans/Neutrophils/The formation of neutrophil extracellular traps (NETs)In vitro neutrophils assayIn vitro[39]
Dental biofilmLPSNeutrophilsLPS/caspase-4/11/Gasdermin DThe formation of neutrophil extracellular traps (NETs)In vitro neutrophils assay
Human periodontal tissues, blood, and biofilm samples
In vitro→Human[40]
P. gingivalis/MacrophagesCaspase-1↑TNFα, ↑IL-12p70, ↑IL-6, ↑IL-10, ↑IFNβ, ↑nitric oxide, ↑IL-1β, and ↑IL-18
The activation of the inflammasome and pyroptotic cell death pathways
In vitro macrophages assay In vitro[41]
T. forsythiaBspA, sialidase, GroEL, and various bacterial lipoproteinsMacrophagesTLR2↑Pro-inflammatory cytokines and ↑inflammatory mediatorsIn vitro macrophages assayIn vitro[17]
F. nucleatum/Macrophages and gingival fibroblasts/Polarization of macrophages toward the proinflammatory M1 phenotype
↑TNF-α,↑iNOS, and ↑LDH
In vitro macrophages and gingival fibroblasts assay
In vivo mouse periodontitis model
In vitro→Animal[43]
Red complex pathogens/Bone marrow-derived dendritic cells (BMDCs)/↑MHC class II, ↑CD80, ↑CD86, ↑CD40, ↑IL-1, ↑IL-6, ↑IL-23, and ↑IL-12p70
Maturation of BMDCs
In vitro BMDCs assayIn vitro[45]
F. alocis Bone-derived mesenchymal stromal cells (BMSCs)TLR2↑RANKL/OPGIn vitro BMSCs assayIn vitro[46]
Periodontal pathogens and oral commensalLipoproteins and LPSOsteoclastsTLR2↑Expression of osteoclastogenic cytokines
↑Osteoclast differentiation
In vitro osteoclast precursors assayIn vitro[47]
Note: ↑ indicates upregulation; ↓ indicates downregulation.
Table 2. BEVs derived from periodontal pathogens and their roles in periodontitis-related systemic diseases.
Table 2. BEVs derived from periodontal pathogens and their roles in periodontitis-related systemic diseases.
BEVs and Associated Systemic Diseases
DiseaseEV SourceCargoTarget CellPathwayReadoutsModelStrength of EvidenceRef.
Neurodegenerative diseasesP. gingivalisNeurotoxic GPs, inflammation-inducible fimbria protein and LPSBV2, SH-SY5Y, and peritoneal macrophages/↑TNF-α expression in the periodontal and hippocampus tissues
↑Hippocampal GP+Iba1+, ↑LPS+Iba1+, and ↑NF-κB+Iba1+ cell numbers
↓BDNF, ↓claudin-5, ↓N-methyl-D-aspartate receptor expression, and ↓BDNF+NeuN+ cell number
↑Blood LPS and TNF-α
In vitro BV2, SH-SY5Y, and peritoneal macrophages assay
In vivo mouse models of cognitive impairment induced by P.g or pEV
In vitro→Animal[48]
GingipainshCMEC/D3PAR2↓ZO-1 and ↓occludin
↑Permeability of hCMEC/D3 cell monolayer
In vitro hCMEC/D3 assayIn vitro[49]
A. actinomycetemcomitansRNATrigeminal ganglion (TG) neuronsTLR4, TLR8, MyD88↑IL-6 and ↑TNF-αIn vivo mouse periodontitis modelAnimal[50]
exRNAMacrophagesTLR8, NF-κB↑TNF-αIn vitro macrophages assay
In vivo mouse heart injection OMV model
In vitro→Animal[42]
MicrogliaNF-κB↑IL-6In vitro BV2 assay
In vivo imaging model after OMV injection in mice
In vitro→Animal[51]
Cardiovascular diseaseP. gingivalisGingipainsHuman microvascular endothelial cells (HMEC-1) ↓PECAM-1
↑Vascular permeability
In vitro HMEC-1 cells assay
In vivo zebrafish OMV injection model
In vitro→Animal[53]
Diabetic retinopathyP. gingivalis/Human retinal microvascular endothelial cells (HRMECs) PAR-2↑HRMECs inflammatory factors and ↑reactive oxygen species production
Mitochondrial dysfunction, apoptosis, and altered endothelial permeability
In vitro HRMECs assay
In vivo mouse diabetes model
In vitro→Animal[54]
Bone lossF. alocis/Committed osteoclast precursors (COCs)TLR2↑Proinflammatory cytokines, ↑Osteoclastogenesis, and ↑bone resorptionIn vivo mice intraperitoneal injection of OMVs modelAnimal[55]
/Bone-derived mesenchymal stromal cells (BMSCs)MAPK, NF-κB↑RANKL/OPGIn vitro BMSCs assayIn vitro[46]
PneumoniaP. gingivalis/Lung epithelial cells/Caspase-3 activation and PARP cleavage
↓Tight junction proteins
In vitro lung epithelial cellsIn vitro[56]
DiabetesP. gingivalisGingipainsHepG2Insulin-induced Akt/glycogen synthase kinase-3β (GSK-3β)↓Insulin sensitivityIn vitro HepG2 assay
In vivo mouse intraperitoneal injection of OMVs model
In vitro→Animal[57]
Adverse pregnancyP. gingivalis/Trophoblast cells/↓Glycolytic pathways in the placenta, ↓placental, ↓fetal weight and ↓GLUT1In vitro the first trimester trophoblast cells assay
In vivo mouse pregnancy model
In vitro→Animal [58]
Note: ↑ indicates upregulation; ↓ indicates downregulation.
Table 3. Roles of CEVs in the pathogenesis of periodontitis.
Table 3. Roles of CEVs in the pathogenesis of periodontitis.
CEVs and Periodontitis
EV SourceCargoTarget CellPathwayReadoutsModelStrength of EvidenceRef.
Gli1+ MSCs/NeutrophilsCXCL1–CXCR2 axis and NF-κB↑ROS
Aberrant activation of neutrophils
In vitro neutrophils assay
In vivo mouse periodontitis model
In vitro→Animal[65]
Periodontal ligament stem cells (PDLSCs)/Macrophages and periodontal ligament fibroblasts (PLFs)/Polarization of macrophages toward the proinflammatory M1 phenotype
NLRP3 inflammasome activation
In vitro macrophages and PLFs assay
In vivo mouse periodontitis model
In vitro→Animal[66]
miR-143-3pMacrophagesPI3K/AKT/NF-κBPolarization of macrophages toward the proinflammatory M1 phenotypeIn vitro macrophages assay
In vivo mouse periodontitis model
In vitro→Animal[67]
microRNA-433-3pMacrophagesTLR2/TLR4/NF-κB p65Polarization of macrophages toward the proinflammatory M1 phenotypeIn vitro macrophages assay
Human periodontal ligament stem cells isolation
In vitro→Human[68]
NeutrophilsmiR-223Periodontal ligament stem cells (PDLSCs)cGMP-PKGInhibit osteogenic differentiation of PDLSCsIn vitro PDLSCs assayIn vitro[69]
MacrophagesmitochondriaBone marrow mesenchymal stem cells (BMSCs)LCN2/OMA1/OPA1↑LCN2
Mitochondrial morphological changes in BMSCs
Osteogenesis impairment in BMSCs
In vitro BMSCs assay
In vivo mouse periodontitis model
In vitro→Animal[70]
OsteoclastsmiR-5134-5pOsteoblastsJAK2/STAT3↑miR-5134-5p
↓Runx2, ↓p-JAK2, and ↓p-STAT3
↑Inflammatory factors mRNA expression
↓BV/TV
↑Cementoenamel junction and alveolar bone crest distance
Morphological disruption of periodontal tissue
Inflammatory cell infiltration
In vitro osteoblasts assay
In vivo mouse periodontitis model
In vitro→Animal[71]
Periodontal ligament stem cells (PDLSCs)RANKL, TNF-αMacrophagesNF-κB↑Osteoclast differentiationIn vitro macrophages assayIn vitro[72]
Osteoblastslnc-MALAT1MacrophagesmiR-124/NFATc1The acceleration of the progression of osteoclastogenesisIn vitro macrophages assayIn vitro[73]
Bone marrow mesenchymal stem cells (BMSCs)miR-151-3pMacrophagesmiR-151-3p/PAFAH1B1 ↑OsteoclastogenesisIn vitro macrophages assayIn vitro[74]
M2-like macrophagesmiR-1227-5pOsteoclasts/↑miR-1227-5p
↓Osteoclast differentiation
In vitro Osteoclasts assayIn vitro[75]
Note: ↑ indicates upregulation; ↓ indicates downregulation.
Table 4. Roles of CEVs in the pathogenesis of periodontitis-related systemic diseases.
Table 4. Roles of CEVs in the pathogenesis of periodontitis-related systemic diseases.
CEVs and Associated Systemic Diseases
DiseaseEV SourceCargoTarget CellPathwayReadoutsModelStrength of EvidenceRef.
Hepatic steatosis
Macrophages and human periodontal ligament fibroblasts (hPDLFs)/HepG2SCD-1/AMPK↑SCD-1 and ↑Hepatocyte adipogenesis
↓AMPK
In vitro HepG2 assay
In vivo rat periodontitis model
In vitro→Animal[76]
Adipose tissue dysfunctionGingival cells/Adipocytes/WAT dysfunction
↓Levels of AKT phosphorylation, ↓adiponectin, ↓leptin, and ↓genes associated with adipogenesis and lipogenesis
In vivo mouse oral and intraperitoneal injection modelsAnimal[77]
DiabetesPlasma/HepG2Insulin signaling↓p-AKT, ↓p-GSK3β, and ↓hepatic glycogen contentIn vitro HepG2 assay
In vivo rat diabetic model
Human blood sample collection
In vitro→Animal→Human[78]
OsteocytesmiR-124-3p, Gal-3, and IL-6Osteoblasts/The regulation of Gal-3 expression of osteoblastsIn vitro Osteoblasts assay
In vivo rat diabetic model and periodontitis model
Human saliva collection
In vitro→Animal→Human[79]
Carotid atherosclerosisHuman umbilical vein endothelial cells (HUVECs)miR-155-5pHuman aortic endothelial cells(HAECs)/↑Angiogenesis and ↑permeability of HAECs
↑Expression of angiogenesis, ↑permeability, and ↑inflammation genes
The acceleration of the occurrence of carotid atherosclerosis
In vitro HAECs assay
In vivo mouse intravenous injection of OMVs model
Human tissue samples
In vitro→Animal→Human[80]
Adverse pregnancyMacrophages/Human umbilical vascular endothelial cells(HUVECs)/↓VEGFR1
Disoriented blood vessel alignment
Impaired angiogenesis
In vitro HUVECs assay
In vivo mouse intravenous injection of OMVs model
In vitro→Animal[81]
Note: ↑ indicates upregulation; ↓ indicates downregulation.
Table 5. Summary of key advances and controversies in EV research on periodontitis.
Table 5. Summary of key advances and controversies in EV research on periodontitis.
AspectWhat’s NewWhat’s Controversial
BEVsSystematic summary of BEVs promoting periodontitis via epithelial barrier disruption, immune-inflammatory amplification, osteogenesis inhibition, and osteoclastogenesis promotion, as well as their involvement in periodontitis-associated systemic diseases.Mechanisms of BEVs crossing biological barriers, and their contribution to periodontitis and systemic diseases remain debated.
CEVsEmphasis on periodontal cell derived EVs under inflammatory conditions, which promote periodontitis by aggravating inflammation and disturbing bone homeostasis, and further contribute to systemic disease progression.Mechanisms of CEVs-mediated effects in periodontitis and related systemic diseases are still insufficiently defined.
EVs for DiagnosisUpdated summary of recent studies on GCF- and saliva-derived EVs as non-invasive biomarkers for detection, staging, and risk assessment.The diagnostic biomarkers derived from GCF and saliva EVs for periodontitis have not yet been clearly identified.
EVs for TherapyHighlight recent engineering strategies (pretreatment of parental cells, direct EV modification, biomaterial-based delivery) to improve EVs’ therapeutic function and achieve targeting and controlled release, thereby enhancing periodontal tissue regeneration.Clinical translation of EVs is limited by low yield, rapid clearance, poor tissue specificity, and unpredictable cargo loading.
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

Fu, Y.; Wang, M.; Teng, R.; Li, A. Emerging Roles of Extracellular Vesicles in the Pathogenesis, Diagnosis, and Therapy of Periodontitis. Biomedicines 2025, 13, 2521. https://doi.org/10.3390/biomedicines13102521

AMA Style

Fu Y, Wang M, Teng R, Li A. Emerging Roles of Extracellular Vesicles in the Pathogenesis, Diagnosis, and Therapy of Periodontitis. Biomedicines. 2025; 13(10):2521. https://doi.org/10.3390/biomedicines13102521

Chicago/Turabian Style

Fu, Yiru, Mengmeng Wang, Rui Teng, and Ang Li. 2025. "Emerging Roles of Extracellular Vesicles in the Pathogenesis, Diagnosis, and Therapy of Periodontitis" Biomedicines 13, no. 10: 2521. https://doi.org/10.3390/biomedicines13102521

APA Style

Fu, Y., Wang, M., Teng, R., & Li, A. (2025). Emerging Roles of Extracellular Vesicles in the Pathogenesis, Diagnosis, and Therapy of Periodontitis. Biomedicines, 13(10), 2521. https://doi.org/10.3390/biomedicines13102521

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