Back to Journals » Journal of Inflammation Research » Volume 18
Extracellular Vesicles in Periodontitis: Pathogenic Mechanisms and Therapeutic Potential
Authors Zhang L, Li X, Zhang B , Li R
Received 4 November 2024
Accepted for publication 18 January 2025
Published 28 January 2025 Volume 2025:18 Pages 1317—1331
DOI https://doi.org/10.2147/JIR.S504612
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Tara Strutt
Ling Zhang,1,* Xiaotong Li,2,* Bin Zhang,3 Ruiji Li4
1School of Nursing, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China; 2Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China; 3Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272001, People’s Republic of China; 4School of Pharmacy, Jining Medical University, Rizhao, Shandong, 276826, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Ruiji Li, School of Pharmacy, Jining Medical University, Xueyuan Road 669, Rizhao, Shandong, 276826, People’s Republic of China, Tel +86-15762380824, Email [email protected]
Abstract: Periodontitis is a prevalent yet frequently overlooked oral disease that is linked to a range of systemic conditions. Although basic treatment and periodontal surgery can alleviate the symptoms of periodontitis to a certain extent, the treatment of severe tissue defects or refractory cases is not effective. Extracellular vesicles (EVs) are subcellular lipid bilayer particles that come from a variety of sources and are prevalent in the biological fluids of vertebrates. They play a key role in intercellular communication by transporting multiple signaling molecules. Recent research has indicated that EVs derived from periodontal pathogens can trigger periodontitis, exacerbate the periodontal damage, and potentially disseminate to other parts of the body, leading to systemic conditions. Conversely, extracellular vesicles derived from dental stem cells (DSCs) have demonstrated the ability to regulate the local periodontal immune environment and foster the regeneration and repair of periodontal tissues, positioning them as a promising candidate for cell-free therapeutic approaches to periodontitis. This review aims to summarize the latest research on the involvement of EVs from different sources in the pathogenesis and treatment of periodontitis, especially to systematically elucidate the mechanism of EVs secreted by periodontal pathogens in periodontitis-related systemic diseases for the first time. By uncovering these complex regulatory processes, new and more effective therapeutic approaches can be explored in the battle against periodontitis and its associated systemic diseases.
Keywords: extracellular vesicles, outer membrane vesicles, dental stem cells, periodontitis, pathogenesis, therapy
Graphical Abstract:
Introduction
Periodontitis, the primary cause of tooth loss among adults, affects nearly 61.6% of the global population, with severe cases impacting roughly 23.6%.1 This condition is not only confined to oral health but also has been linked to systemic illnesses such as diabetes mellitus, Alzheimer’s disease, cardiovascular disease, and certain types of cancer.2–4 Nonsurgical periodontal treatment (NSPT) is the basis of periodontitis treatment, mainly including supragingival scaling, subgingival scaling and root planing. Surgical treatment is a supplement to nonsurgical treatment, especially when periodontitis has advanced to a more severe stage. However, these conventional treatments are not effective in treating severe tissue defects or refractory cases. Moreover, given the limitations of the current study design, the potential beneficial effects of nonsurgical periodontal therapy on arterial stiffness in patients with periodontitis still need to be validated in clinical trials.5 This strongly suggests the need to unlock new therapeutic method.
Extracellular vesicles (EVs) secreted by periodontal pathogens have been proved to be a key link in the occurrence and development of periodontitis and may also be a potential risk factor for related systemic diseases. Higher baseline levels of periodontal pathogens have been found to significantly interfere with the efficacy of one-stage full-mouth subgingival instrumentation for periodontitis.6 In addition, research has also highlighted the promise of EVs in combating inflammation-related diseases, particularly periodontitis.7 Compared with a single cell or bacteria, EVs can carry and deliver nucleic acids, proteins, lipids and other bioactive molecules for efficient information exchange between cells, thus playing a central role in a variety of physiological and pathological processes. In addition, EVs-mediated cell-free therapy effectively avoids the risk of immune rejection and tumor formation that may accompany stem cell transplantation.
Based on the above findings, this study was conducted by searching PubMed, Web of Science, Cochrane and Embase databases for all publications related to this topic up to October 2024, especially those published in the last 5 years. Non-English, non-availability of full text, duplication of publications, abstracts of meetings and policy papers were excluded. This review synthesizes current understanding of EVs’ involvement in periodontitis, emphasizing their bifurcated roles. In particular, it is the first comprehensive summary of how EVs released by periodontal pathogens are involved in the pathogenesis of periodontitis-related systemic diseases. This study was aimed to fully explore the potential of EVs in periodontitis treatment by revealing the nuances of the dual role of EVs and refining preclinical approach to EVs application to compensate for the shortcomings of conventional treatments.
Overview of EVs
EVs, subcellular particles sized 30 nm to 1 μm and enclosed by a lipid bilayer, display a cup-shaped morphology under electron microscopy.8 The membrane structure of EVs facilitates the carriage and delivery of various cellular components, including proteins, nucleic acids, and lipids. Surface-specific proteins or glycoproteins on EVs act as markers for EV type identification and parent cell tracing. Additionally, receptors or ligands on EVs engage with target cells, mediating binding and fusion.9 Changes in the sugar or protein composition of EV membrane affect vesicle tropism and physiological properties. For example, exposure to the integrin CD47 on the surface of EVs can protect EVs from phagocytosis, thereby increasing the circulation time of EVs in the bloodstream.10 In addition, the lipid membrane bilayer of EVs allows a variety of hydrophobic therapeutic ingredients to be incorporated to improve the stability and efficacy of these drugs.11 Therefore, taking full advantage of membrane modules for modification is an innovative way to design EVs. Moreover, the content of different cargos (such as proteins, transcription factors, nucleic acids, etc) in EVs will vary depending on the parental cell type, physiological conditions, and biogenesis.12 As paracrine messengers, EVs transport signaling molecules, thereby regulating a myriad of cellular pathways either in the vicinity or at a distance.
Bacterial-Derived EVs
The majority of EVs released by bacteria, especially Gram-negative bacteria, are classified as bacterial outer membrane vesicles (OMVs).13 These vesicles serve as a crucial conduit for interactions within microbial communities and between microbes and their hosts. Unattached subgingival plaque is the key initiating factor of periodontitis, which is rich in gram-negative bacteria. Research has demonstrated that OMVs derived from periodontal pathogens (hereinafter referred to as pp-EVs) play a significant role in the establishment of bacterial biofilms.14 Upon interaction with pathogen-associated molecular patterns (PAMPs) present on pp-EVs, pattern recognition receptors (PRRs) on the host’s immune cells can trigger inflammatory responses and initiate EV-mediated adaptive immune responses.15 Kim et al suggested that pp-EVs accelerate bone loss through inhibiting bone formation and increasing bone resorption.16 In addition, pp-EVs can restrain the proliferation and growth of fibroblasts, resulting in chronic periodontitis.17
Dental Stem Cell-Derived EVs
Beyond the pathological impact of pp-EVs on periodontal tissues, EVs released by dental stem cells (hereinafter referred to as DSC-EVs) are instrumental in periodontitis therapy. They achieve this by modulating the immune microenvironment and fostering the regeneration of periodontal tissues.18,19 To date, seven distinct types of DSCs have been identified and isolated, encompassing tooth germ stem cells (TGSCs), dental follicle stem cells (DFSCs), stem cells from the apical papilla (SCAPs), dental pulp stem cells (DPSCs), periodontal ligament stem cells (PDLSCs), stem cells from exfoliated deciduous teeth (SHEDs), and gingival mesenchymal stem cells (GMSCs).20 Hu et al found that GMSC-derived EVs (hereinafter referred to as GMSC-EVs) effectively cross-regulate NF-κB and Wnt/β-catenin signaling pathways to promote osteogenic differentiation of PDLSCs in inflammatory environment.21 In a rat model of periodontal defect, EVs derived from DFSCs have been shown significant periodontal regeneration potential.22 On one side of the spectrum, DSC-EVs have been found to stimulate neovascularization and encourage the osteogenic differentiation of stem cells, which in turn mitigates the loss of alveolar bone.23,24 Conversely, DSC-EVs also bolster the reparative capacity of compromised periodontal tissues by augmenting the proliferation, migration, and collagen production of periodontal cells (Figure 1).25
Hence, EVs originating from various sources may exhibit distinct roles in the pathogenesis and treatment of periodontitis. The revelation of these related regulatory mechanisms may provide new strategies to tackle periodontitis.
Pathogenesis of pp-EVs in Periodontitis and Related Systemic Diseases
pp-EVs instigate inflammation and tissue destruction in periodontitis by breaching the oral epithelial barrier and perturbing the periodontal immune microenvironment. The possible pathogenesis of pp-EVs in periodontitis has been adequately described in Cai and Huang’s review and will not be repeated here.26,27 Emerging data from both epidemiological and experimental studies have demonstrated a connection between periodontitis and multiple systemic diseases, including diabetes mellitus, Alzheimer’s disease, cardiovascular disease, and osteoporosis. And the presence of periodontitis is likely to affect the prognosis and outcome of these diseases. The bacterium Porphyromonas gingivalis (P. gingivalis), a key player in periodontitis, has been identified not just in the oral lesion sites but also in distant organs such as the brain, cardiovascular system, liver and fallopian tube-ovaries.28 Beyond its nanoscale dimensions and robust structural integrity, OMVs also boasts exceptional environmental adaptability and the ability to evade immune detection. These traits facilitate its ability to surpass the localized regions of its parent bacteria, thereby enhancing its invasiveness.29 Moreover, the relatively high concentration and stable composition of pathogenic agents in OMVs enable them to establish bacteria-host dialogue and instigate disease processes even when live bacteria are not present. Hence, pp-EVs are potentially a significant nexus connecting periodontitis with its associated systemic diseases. Through in-depth understanding of the mechanisms involved, potential therapeutic intervention targets can be found in the treatment of periodontitis-related systemic diseases.
pp-EVs and Diabetes Mellitus
Diabetes mellitus is caused by defects in insulin secretion or activity. Periodontitis and diabetes mellitus have a bidirectional association, which can mutually promote their prevalence and severity.30 The mechanisms by which diabetes mellitus facilitates periodontitis have been comprehensively described by Zhao et al, including microbiome factors, host immune factors, and oxidative stress.31 Meanwhile, several studies have shown that P. gingivalis may play a major role in the development of type 2 diabetes mellitus by inducing insulin resistance. For example, P. gingivalis-induced endotoxemia can exacerbate nonalcoholic fatty liver disease by increasing insulin resistance and inhibiting glucose metabolism.32 Seyama et al reported that gingipains, an important virulence factor secreted by P. gingivalis-derived OMVs (hereinafter referred to as Pg-EVs), can inhibit glycogen synthesis in liver cells by targeting the Akt/GSK-3β signaling pathway and thus maintain a high level of blood sugar in experimental mice.33 Moreover, in an experiment by Huang, Pg-EVs were shown to release arginine gingival proteinase (Rgp) into the aqueous humor when injected into diabetic mice via venules. This process increased the level of oxidative stress in retinal microvascular endothelial cells and lead to mitochondrial dysfunction, thereby aggravating retinopathy in diabetic mice.34
pp-EVs and Alzheimer’s Disease
Alzheimer’s disease is a prevalent neurodegenerative condition in the elderly. Evidence suggests that periodontal pathogens and their virulence factors can cross the blood-brain barrier (BBB), facilitate the penetration of related pathogenic factors and cause pathological changes similar to those observed in Alzheimer’s disease.35 Nonaka found that Pg-EVs delivered gingipain to cerebral microvascular endothelial cells through the blood circulation and degrades tight junction proteins including Zonula occludens-1 (ZO-1) and occludin. Such pathological changes destroyed the BBB and promoted P. gingivalis and its virulence factors to infiltrate into the brain parenchyma.36 Consistent with these findings, Elashiry et al demonstrated for the first time in a mouse model that Pg-EVs can metastasize and cross the BBB to participate in the pathogenesis of AD.37 Actinobacillus actinomycetemcomitans (A. actinomycetemcomitans) is an equally essential periodontal pathogen, and A. actinomycetemcomitans-derived OMVs (Aa-EVs) can also penetrate the BBB and invade the brain.38 Choi and his collaborators indicated that Aa-EVs provoke neuroinflammation by delivering extracellular RNA (exRNA) to activate the proinflammatory factors IL-6 and NF-κB in brain monocyte/microglia.39
pp-EVs and Cardiovascular Disease
Disruption of intercellular junctions between endothelial cells impairs the normal function and integrity of the endothelial barrier, which further exacerbates vascular failure and cardiovascular disease.40 To determine whether pp-EVs can mediate endothelial dysfunction, Farrugia et al extracted Pg-EVs and infected human umbilical vein endothelial cells (HUVECs). Interestingly, the gingipains in pp-EVs suppress the expression of platelet-endothelial cell adhesion molecule 1 (PECAM-1) on the cell surface and loosen intercellular contact, thereby increasing the permeability of endothelial cells.41 Moreover, Yue and cooperator reported that Rho kinase (ROCK) mediates pp-EV-induced endothelial nitric oxide synthase (eNOS) suppression through ERK1/2 and p38 MAPK, thereby destroying vascular integrity and homeostasis.42 In addition, pp-EVs can also elicit cardiovascular disease by facilitating the formation of atherosclerosis. Pg-EVs upregulate the expression of Runt-related transcription factor-2 (RUNX2) by rapidly activating ERK1/2 in vascular smooth muscle cells (VSMCs), which is conducive to cell calcification and atherosclerosis.43
pp-EVs and Other Diseases
Currently, the significance of pp-EVs in other systemic diseases has been gradually emphasized. For instance, He et al proposed that Pg-EVs destroy the function of the lung epithelial barrier and inflict inflammatory lung diseases.44 EVs derived from P. gingivalis or Filifactor alocis (F. alocis) are concentrated in long bones after intraperitoneal administration and subsequently activate osteoclast precursors to yield transcription factors, ultimately leading to osteoporosis.45,46 It can be seen that pp-EVs may travel throughout the body by blood circulation and trigger or aggravate pathological changes in the distal organs by carrying multiple bioactive molecules. This realization underscores that a single treatment may not be sufficient to address the complexity of systemic diseases associated with periodontitis. When devising treatment strategies for patients grappling with periodontitis alongside associated systemic conditions, it is essential to integrate a multifaceted approach that encompasses periodontal therapy. The pathogenesis of pp-EVs in periodontitis related systemic diseases are summarized in Figure 2 and Table 1.
![]() |
Table 1 The Pathogenic Role of pp-EVs in Systemic Diseases |
Restoration Mechanisms of DSC-EVs in Periodontitis Therapy
Traditional stem cell therapy excels at facilitating tissue repair, regeneration, and addressing stubborn medical conditions. Nonetheless, it carries inherent risks, including unpredictable cell differentiation, procedural complexity, and safety concerns. In this context, EV-mediated cell-free therapy stands out for its ability to mimic parental cell functions more effectively. It is characterized by heightened safety, enhanced tissue penetration, accurate targeting, and a multiplicity of functions. These attributes render EVs a compelling option for therapeutic interventions in periodontitis.52,53 Figure 3 shows the restoration mechanisms of DSC-EVs in periodontitis therapy.
The Role of microRNAs in DSC-EVs
MicroRNAs (miRNAs) in EVs are essential for cell communication and signal transduction, affecting physiological and pathological processes. In cell-free therapies, DSC-derived EVs are key vectors for miRNA delivery and precise cell function regulation. Liu et al first identified miRNAs in PDLSC-derived EVs that promote osteogenesis in recipient cells, shedding light on miRNA regulatory mechanisms in DSC-EVs during this process.54 Similarly, Shen et al suggested that miR-1246 enrichment in DPSC-EVs protects experimental mice from alveolar bone loss, while antiagomiR-1246 reverses these effects.55 To dissect the regulatory effects of miRNAs in EVs on angiogenesis, Pizzicannella analyzed differentially expressed miRNAs in GMSC-EVs. These authors suggested that GMSC-EVs enhance the expression of VEGF, OPN, and RUNX2 in a rat skull defect model by delivering miR-2861 and miR-210.56 Furthermore, periodontal cell senescence and impaired multifunctional differentiation caused by oxidative stress are not conducive to the repair and regeneration of periodontal defects. Accordingly, delaying cell senescence may be an emerging alternative for elucidating the intrinsic repair mechanism of periodontal tissue.57 Mas-Bargues found that high oxygen tension causes DPSC senescence, but EV-derived miR-302b can delay this and restore DPSC pluripotency.58
In fact, in addition to miRNAs, there are other nucleic acid components in EVs, such as mRNAs, circRNAs, and lncRNAs. Xie et al demonstrated that elevated circLPAR1 in DPSC-EVs, by competitive binding to hsa-miR-31, enhances SATB2 expression and osteogenic differentiation in recipient cell.59 Furthermore, SHED-derived EVs (SHED-EVs) enhanced glutamate metabolism and oxidative phosphorylation activity in DPSCs through delivery of the mRNA encoding mitochondrial transcription Factor A (TFAM), which was conducive to bone regeneration in DPSCs.60
The Role of Proteins in DSC-EVs
Besides miRNAs, proteins are also essential constituents of EVs, facilitating intercellular communication and the transduction of signals through these vesicular shuttles. Furthermore, cargo proteins can be used as biomarkers for disease diagnosis and prognosis assessment. Transcriptome sequencing has revealed that GMSCs participate in regulating the processes of bone homeostasis and angiogenesis by secreting TGF-β, BMPs, VEGF and so on.61 Luo et al found that SHEDs-EVs can mobilize naïve bone marrow mesenchymal stromal cells (BMSCs) through secreting multiple growth factors, thereby promoting bone and periodontal tissue regeneration.62 Zhang et al reported a significant increase in VEGFA expression in inflamed PDLSCs, resulting in enhanced angiogenesis in HUVECs when co-cultured with inflamed PDLSCs.63 Given the above findings, EVs play a remarkable role in the treatment of periodontitis by delivering proteins to recipient cells.
Key Pathways Mediated by DSC-EVs
Although the types of molecules transported by EVs are distinctive in various cell types or in diverse growth microenvironments, key pathways in recipient cells are always mobilized by this delivery. Over the years, the pathways affected by DSC-EVs in the treatment of periodontitis have been extensively and intensively studied.
Mitogen-Activated Protein Kinase (MAPK) Pathway
MAPK pathway is a critical bridge connecting extracellular signals with intracellular responses. Co-culturing PDLSCs with DFSCs enhances PDLSC proliferation and osteogenic differentiation via the p38 MAPK pathway, and this effect is confirmed in periodontal tissue regeneration in SD rats.64 Jin et al confirmed that DPSC-EVs promoted the migration and mineralization of adipose derived stem cells (ADSCs) by activating the MAPK pathway, and significantly improved the regeneration of mandibular defects in rats.65 Similarly, Zhao et al extracted EVs from PDLSCs to incubate BMSCs and discovered that these cells could expedite cell proliferation and migration by increasing the phosphorylation of AKT and ERK1/2, subsequently accelerating bone tissue repair.66 Furthermore, DPSC-EVs were shown to promotes migration, proliferation and capillary formation of HUVECs by upregulating Cdc42/p38 MAPK signaling pathway.67
Wnt Pathway
The Wnt signaling pathway is recognized for its multifaceted role in periodontal biology. It not only stimulates the proliferation and differentiation of periodontal cells but also governs the cementum formation and development. Furthermore, it exerts a regulatory influence on the dynamics of periodontal tissue destruction and regeneration. It has been reported that GMSC-EVs improve the regenerative potential of PDLSCs in the inflammatory microenvironment by inhibiting NF-κB signaling and Wnt5a expression.68 Qian et al revealed that EVs extracted from curcumin-treated PDLSCs had an enhanced pro-osteogenic ability, which was due to upregulated p-GSK3β and β-catenin protein levels in the recipient cells.69 Consistently, after TNF-α pretreatment, miR-1260b in GMSC-EVs downregulated osteoclastogenic activity of periodontal ligament cells by inhibiting the Wnt5a/RANKL pathway.70 These results provide valuable new ideas for the treatment of periodontitis.
Janus Kinase 2/Signal Transducer and Activator of Transcription 3 (JAK2/STAT3) Pathway
JAK2/STAT3 pathway is involved in the initiation and progression of inflammatory responses and immune responses in diverse pathological processes. For instance, DPSC-EVs effectively downregulated the expression of the IL-6, p-JAK2, and p-STAT3 proteins in LPS-treated PDLSCs. These findings suggested that the JAK2/STAT3 pathway may be involved in regulating the anti-inflammatory and osteogenic effects of DPSC-EVs on PDLSCs.71 Moreover, recent studies have focused on the role of STAT3 in the regulation of autophagy. Xie et al processed macrophages with SHED-EVs and discovered that STAT3 pathway-related proteins were activated, subsequently promoting the expression of the autophagy-related protein LC3.72
Other Signaling Pathway
M1 and M2 macrophage polarization imbalance is key in periodontitis progression. LPS-stimulated PDLSCs secrete EVs that enhance M1 polarization via TLR2/TLR4/NF-κB,73 while GMSC-EVs promote M2 polarization through HIF-1α/mTOR after TNF-α and IFN-α stimulation.74 Hypoxic preconditioning has been reported to markedly increase EV release in DPSCs and promote the polarization of M2 macrophages, thus ameliorating LPS-induced inflammatory osteolysis.75 Table 2 summarizes the restoration mechanism of DSC-EVs in periodontitis therapy and tissue regeneration.
![]() |
Table 2 Restoration Mechanisms of DSC-EVs in Periodontitis Therapy and Tissue Regeneration |
Application Strategy of EVs in the Prevention and Treatment of Periodontitis
Taking Full Advantage of pp-EVs for Vaccine Development
OMVs mimic parental bacteria’s immunogenicity, elicit host adaptive responses, and are highly stable for <1 year at low temperatures.86 In 2016, the Food and Drug Administration (FDA) successfully approved an OMV-based vaccine, namely, the meningitis serotype B vaccine BEXSERO (GlaxoSmithKline, London, UK), demonstrating the viability of OMVs as a vaccine or drug delivery platform.87 Local combination of Pg-EVs and TLR3 agonist poly(I:C) on the nasal mucosa has been reported to effectively trigger a specific antibody response significantly reducing P. gingivalis levels in mice, with safety confirmed in a trial by Nakao.88 These findings confirm pp-EVs’ feasibility as a safe, efficient intranasal vaccine for periodontitis. While all Gram-negative bacteria produce OMVs with varying yields and cargos, even under different environmental conditions, thorough evaluation is essential before pp-EVs can be developed into vaccines. Maximizing the immunogenicity of pp-EVs is crucial while ensuring safety and tolerability.
Functional Modification Strategies for EVs Before Clinical Application
Preclinical studies indicate MSC-EVs’ potential in treating inflammation and promoting tissue regeneration. However, the successful clinical application of EVs still faces certain limitations, such as low yield, rapid clearance, unsatisfactory targeting ability, and uncertain loading efficiency.89 In this regard, Yang et al8 summarized the emerging technologies for EVs isolation, engineering, and delivery systems (eg, heterogeneous hydrogels, microneedle patches and micro-nanoparticles). Notably, the autonomous mobility of emerging nanomotor technology is expected to be used in the future to enhance EVs delivery efficiency and targeting.
The most common cargo loading method involves preprocessing parental cells by optimizing culture, co-incubating with therapeutics, and transfecting with RNA, peptides, and proteins. For instance, hypoxia preconditioning stimulated SHED-EVs’ potential in angiogenesis and osteogenesis by boosting EVs secretion, VEGF signaling and thyroid hormone synthesis.90 Xu et al demonstrated that PDLSCs engineered with the P2X7R gene release EVs that enhance osteogenic differentiation in inflammation via differential miRNA expression.91 Although the abovementioned three loading methods are relatively simple, the loading efficiency and drug toxicity on EV secretion cannot be controlled. Researchers suggest either direct co-incubation of therapeutic agents with EVs or temporary membrane disruption using physicochemical methods for drug loading (Figure 4). After loading BMP2 into EVs via electroporation or sonication, Yerneni et al reported that the loading efficiency of sonication increased by more than 3-fold.92 Physicochemical methods can enhance cargo loading efficiency in EVs, but they must be carefully optimized to avoid damaging or contaminating the EVs.
Additionally, EVs can be equipped with targeting ligands like antibodies, peptides, or proteins to enhance their ability to recognize and bind specific targets. For instance, CXCR4-overexpressing EVs boost macrophage targeting and M2 macrophage polarization by delivering miR-126.93 EV targeting can also be enhanced by their surface negative potential for electrostatic attraction or by conjugating them with iron oxide nanoparticles for magnetic attraction.94,95 Additionally, combining EVs with biomaterials improves targeted drug delivery, reduces EV clearance by the immune system, and enables local sustained release.96 (Figure 5).
Conclusions and Future Perspective
Rising as vital mediators of intercellular communication, EVs have been identified to influence the progression of periodontitis from multiple perspectives and seemingly playing a Janus-faced role in regulating the fate of periodontitis. Therefore, giving full play to the protective effect of EVs on periodontal tissue, such as the development of pp-EV vaccine and engineering EVs, may play an unexpected role in periodontitis prevention and periodontal regeneration treatment. Reasonably combined with conventional periodontal therapy, EVs may effectively improve quality of life in patients with periodontitis. Although EVs have shown great potential in disease diagnosis and treatment, the negative outcomes that may accompany their preclinical application have been less reported. In view of the current difficulties in the preparation of therapeutic EVs, it is necessary to further explore the safety risks caused by methodological bottlenecks. In addition, with the further study of EV-mediated systemic interactions and the continuous overcoming of methodological bottleneck, the clinical application prospect of treating periodontitis and related systemic diseases by EVs will be brighter.
Funding
This work was supported by the Natural Science Foundation of Shandong Province (ZR2021QH294; ZR2021QB124), Research Fund for Academician Lin He New Medicine (JYHL2022MS02) and Outstanding Youth Foundation (Overseas) Project of Rizhao City Natural Science Foundation (RZ2021ZR1).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Trindade D, Carvalho R, Machado V, Chambrone L, Mendes JJ, 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(5):604–626. doi:10.1111/jcpe.13769
2. Wu CZ, Yuan YH, Liu HH, et al. Epidemiologic relationship between periodontitis and type 2 diabetes mellitus. BMC Oral Health. 2020;20(1):204. doi:10.1186/s12903-020-01180-w
3. Sun YQ, Richmond RC, Chen Y, Mai XM. Mixed evidence for the relationship between periodontitis and Alzheimer’s disease: a bidirectional Mendelian randomization study. PLoS One. 2020;15(1):e0228206. doi:10.1371/journal.pone.0228206
4. Sanz M, Marco Del Castillo A, Jepsen S, et al. Periodontitis and cardiovascular diseases: consensus report. J Clin Periodontol. 2020;47(3):268–288. doi:10.1111/jcpe.13189
5. Polizzi A, Nibali L, Tartaglia GM, et al. Impact of nonsurgical periodontal treatment on arterial stiffness outcomes related to endothelial dysfunction: a systematic review and meta-analysis. J Periodontol. 2024:1–16. doi:10.1002/jper.24-0422
6. Isola G, Polizzi A, Santonocito S, et al. Effect of quadrantwise versus full-mouth subgingival instrumentation on clinical and microbiological parameters in periodontitis patients: a randomized clinical trial. J Periodontal Res. 2024;59(4):647–656. doi:10.1111/jre.13279
7. Deng DK, Zhang JJ, Gan D, et al. Roles of extracellular vesicles in periodontal homeostasis and their therapeutic potential. J Nanobiotechnology. 2022;20(1):545. doi:10.1186/s12951-022-01757-3
8. Yang C, Xue Y, Duan Y, Mao C, Wan M. Extracellular vesicles and their engineering strategies, delivery systems, and biomedical applications. J Control Release. 2024;365:1089–1123. doi:10.1016/j.jconrel.2023.11.057
9. Sato YT, Umezaki K, Sawada S, et al. Engineering hybrid exosomes by membrane fusion with liposomes. Sci Rep. 2016;6:21933. doi:10.1038/srep21933
10. Simeone P, Bologna G, Lanuti P, et al. Extracellular vesicles as signaling mediators and disease biomarkers across biological barriers. Int J Mol Sci. 2020;21(7):2514. doi:10.3390/ijms21072514
11. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16(7):748–759. doi:10.1038/s41565-021-00931-2
12. Kumar MA, Baba SK, Sadida HQ, et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct Target Ther. 2024;9(1):27. doi:10.1038/s41392-024-01735-1
13. Jalalifar S, Morovati Khamsi H, Hosseini-Fard SR, et al. Emerging role of microbiota derived outer membrane vesicles to preventive, therapeutic and diagnostic proposes. Infect Agent Cancer. 2023;18(1):3. doi:10.1186/s13027-023-00480-4
14. Duchesne P, Grenier D, Mayrand D. Demonstration of adherence properties of Porphyromonas gingivalis outer membrane vesicles using a new microassay. Oral Microbiol Immunol. 1995;10(2):76–80. doi:10.1111/j.1399-302x.1995.tb00122.x
15. Xiao M, Li G, Yang H. Microbe-host interactions: structure and functions of gram-negative bacterial membrane vesicles. Front Microbiol. 2023;14:1225513. doi:10.3389/fmicb.2023.1225513
16. Kim HY, Song MK, Gho YS, Kim HH, Choi BK. Extracellular vesicles derived from the periodontal pathogen Filifactor alocis induce systemic bone loss through Toll-like receptor 2. J Extracell Vesicles. 2021;10(12):e12157. doi:10.1002/jev2.12157
17. Haugsten HR, Kristoffersen AK, Haug TM, et al. Isolation, characterization, and fibroblast uptake of bacterial extracellular vesicles from Porphyromonas gingivalis strains. Microbiologyopen. 2023;12(5):e1388. doi:10.1002/mbo3.1388
18. Li H, Sun L, Wang Y. Inhibition of LPS‑induced NLRP3 inflammasome activation by stem cell-conditioned culture media in human gingival epithelial cells. Mol Med Rep. 2023;27(5):106. doi:10.3892/mmr.2023.12993
19. Zarubova J, Hasani-Sadrabadi MM, Dashtimoghadam E, et al. Engineered delivery of dental stem-cell-derived extracellular vesicles for periodontal tissue regeneration. Adv Healthc Mater. 2022;11(12):e2102593. doi:10.1002/adhm.202102593
20. Chen Y, Huang H, Li G, Yu J, Fang F, Qiu W. Dental-derived mesenchymal stem cell sheets: a prospective tissue engineering for regenerative medicine. Stem Cell Res Ther. 2022;13(1):38. doi:10.1186/s13287-022-02716-3
21. Hu Y, Wang Z, Fan C, et al. Human gingival mesenchymal stem cell-derived exosomes cross-regulate the Wnt/β-catenin and NF-κB signalling pathways in the periodontal inflammation microenvironment. J Clin Periodontol. 2023;50(6):796–806. doi:10.1111/jcpe.13798
22. Liang L, Wang L, Liao Z, et al. High-yield nanovesicles extruded from dental follicle stem cells promote the regeneration of periodontal tissues as an alternative of exosomes. J Clin Periodontol. 2024;51(10):1395–1407. doi:10.1111/jcpe.14036
23. Sunartvanichkul T, Arayapisit T, Sangkhamanee SS, et al. Stem cell-derived exosomes from human exfoliated deciduous teeth promote angiogenesis in hyperglycemic-induced human umbilical vein endothelial cells. J Appl Oral Sci. 2023;31:e20220427. doi:10.1590/1678-7757-2022-0427
24. Zheng Y, Lu H, Mu Q, et al. Effects of sEV derived from SHED and DPSC on the proliferation, migration and osteogenesis of PDLSC. Regen Ther. 2023;24:489–498. doi:10.1016/j.reth.2023.09.009
25. Mohammed E, Khalil E, Sabry D. Effect of adipose-derived stem cells and their exo as adjunctive therapy to nonsurgical periodontal treatment: a histologic and histomorphometric study in rats. Biomolecules. 2018;8(4):167. doi:10.3390/biom8040167
26. Cai R, Wang L, Zhang W, et al. The role of extracellular vesicles in periodontitis: pathogenesis, diagnosis, and therapy. Front Immunol. 2023;14:1151322. doi:10.3389/fimmu.2023.1151322
27. Huang X, Wang H, Wang C, Cao Z. The applications and potentials of extracellular vesicles from different cell sources in periodontal regeneration. Int J Mol Sci. 2023;24(6):5790. doi:10.3390/ijms24065790
28. Konkel JE, O’Boyle C, Krishnan S. Distal Consequences of Oral Inflammation. Front Immunol. 2019;10:1403. doi:10.3389/fimmu.2019.01403
29. Zhao G, Jones MK. Role of bacterial extracellular vesicles in manipulating infection. Infect Immun. 2023;91(5):e0043922. doi:10.1128/iai.00439-22
30. Stöhr J, Barbaresko J, Neuenschwander M, Schlesinger S. Bidirectional association between periodontal disease and diabetes mellitus: a systematic review and meta-analysis of cohort studies. Sci Rep. 2021;11(1):13686. doi:10.1038/s41598-021-93062-6
31. Zhao M, Xie Y, Gao W, Li C, Ye Q, Li Y. Diabetes mellitus promotes susceptibility to periodontitis-novel insight into the molecular mechanisms. Front Endocrinol. 2023;14:1192625. doi:10.3389/fendo.2023.1192625
32. Thalla S. Increasing risk factors for non-alcoholic fatty liver disease; an insight into chronic periodontitis and insulin resistance. Endocr Metab Immune Disord Drug Targets. 2022;22(8):807–814. doi:10.2174/1871530322666220104095534
33. Seyama M, Yoshida K, Yoshida K, 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(6):165731. doi:10.1016/j.bbadis.2020.165731
34. Huang S, Cao G, Dai D, et al. Porphyromonas gingivalis outer membrane vesicles exacerbate retinal microvascular endothelial cell dysfunction in diabetic retinopathy. Front Microbiol. 2023;14:1167160. doi:10.3389/fmicb.2023.1167160
35. Kouki MA, Pritchard AB, Alder JE, Crean S. Do periodontal pathogens or associated virulence factors have a deleterious effect on the blood-brain barrier, contributing to Alzheimer’s disease? J Alzheimers Dis. 2022;85(3):957–973. doi:10.3233/jad-215103
36. 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. doi:10.1016/j.neuint.2022.105282
37. Elashiry M, Carroll A, Yuan J, et al. Oral microbially-induced small extracellular vesicles cross the blood-brain barrier. Int J Mol Sci. 2024;25(8):4509. doi:10.3390/ijms25084509
38. Lamphere AK, Nieto VK, Kiser JR, Haddlesey CB. Potential mechanisms between periodontitis and Alzheimer’s disease: a scoping review. Can J Dent Hyg. 2023;57(1):52–60.
39. Ha JY, Choi SY, Lee JH, Hong SH, Lee HJ. Delivery of periodontopathogenic extracellular vesicles to brain monocytes and microglial IL-6 promotion by RNA cargo. Front Mol Biosci. 2020;7:596366. doi:10.3389/fmolb.2020.596366
40. Chistiakov DA, Orekhov AN, Bobryshev YV. Endothelial barrier and its abnormalities in cardiovascular disease. Front Physiol. 2015;6:365. doi:10.3389/fphys.2015.00365
41. Farrugia C, Stafford GP, Murdoch C. Porphyromonas gingivalis outer membrane vesicles increase vascular permeability. J Dent Res. 2020;99(13):1494–1501. doi:10.1177/0022034520943187
42. Jia Y, Guo B, Yang W, Zhao Q, Jia W, Wu Y. Rho kinase mediates Porphyromonas gingivalis outer membrane vesicle-induced suppression of endothelial nitric oxide synthase through ERK1/2 and p38 MAPK. Arch Oral Biol. 2015;60(3):488–495. doi:10.1016/j.archoralbio.2014.12.009
43. Yang WW, Guo B, Jia WY, Jia Y. Porphyromonas gingivalis-derived outer membrane vesicles promote calcification of vascular smooth muscle cells through ERK1/2-RUNX2. FEBS Open Bio. 2016;6(12):1310–1319. doi:10.1002/2211-5463.12151
44. He Y, Shiotsu N, Uchida-Fukuhara 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. doi:10.1016/j.archoralbio.2020.104841
45. Kim HY, Song MK, Lim Y, et al. Effects of extracellular vesicles derived from oral bacteria on osteoclast differentiation and activation. Sci Rep. 2022;12(1):14239. doi:10.1038/s41598-022-18412-4
46. Song MK, Kim HY, Choi BK, Kim HH. Filifactor alocis-derived extracellular vesicles inhibit osteogenesis through TLR2 signaling. Mol Oral Microbiol. 2020;35(5):202–210. doi:10.1111/omi.12307
47. Yoshida K, Yoshida K, Seyama M, et al. Porphyromonas gingivalis outer membrane vesicles in cerebral ventricles activate microglia in mice. Oral Dis. 2023;29(8):3688–3697. doi:10.1111/odi.14413
48. Han EC, Choi SY, Lee Y, Park JW, Hong SH, Lee HJ. 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(12):13412–13422. doi:10.1096/fj.201901575R
49. Ho MH, Guo ZM, Chunga J, Goodwin JS, Xie H. Characterization of innate immune responses of human endothelial cells induced by Porphyromonas gingivalis and their derived outer membrane vesicles. Front Cell Infect Microbiol. 2016;6:139. doi:10.3389/fcimb.2016.00139
50. Qi M, Miyakawa H, Kuramitsu HK. Porphyromonas gingivalis induces murine macrophage foam cell formation. Microb Pathog. 2003;35(6):259–267. doi:10.1016/j.micpath.2003.07.002
51. Lara B, Sassot M, Calo G, et al. Extracellular vesicles of Porphyromonas gingivalis disrupt trophoblast cell interaction with vascular and immune cells in an in vitro model of early placentation. Life. 2023;13(10):1971. doi:10.3390/life13101971
52. Mai Z, Chen H, Ye Y, et al. Translational and clinical applications of dental stem cell-derived exosomes. Front Genet. 2021;12:750990. doi:10.3389/fgene.2021.750990
53. Dostert G, Mesure B, Menu P, Velot É. How do mesenchymal stem cells influence or are influenced by microenvironment through extracellular vesicles communication? Front Cell Dev Biol. 2017;5:6. doi:10.3389/fcell.2017.00006
54. Liu T, Hu W, Zou X, et al. Human periodontal ligament stem cell-derived exosomes promote bone regeneration by altering MicroRNA profiles. Stem Cells Int. 2020;2020:8852307. doi:10.1155/2020/8852307
55. Shen Z, Kuang S, Zhang Y, et al. Chitosan hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates periodontitis in mice via a macrophage-dependent mechanism. Bioact Mater. 2020;5(4):1113–1126. doi:10.1016/j.bioactmat.2020.07.002
56. Pizzicannella J, Diomede F, Gugliandolo A, et al. 3D printing PLA/gingival stem cells/ EVs upregulate miR-2861 and −210 during osteoangiogenesis commitment. Int J Mol Sci. 2019;20(13):3256. doi:10.3390/ijms20133256
57. Ying S, Tan M, Feng G, et al. Low-intensity pulsed ultrasound regulates alveolar bone homeostasis in experimental periodontitis by diminishing oxidative stress. Theranostics. 2020;10(21):9789–9807. doi:10.7150/thno.42508
58. Mas-Bargues C, Sanz-Ros J, Román-Domínguez A, et al. Extracellular vesicles from healthy cells improves cell function and stemness in premature senescent stem cells by miR-302b and HIF-1α activation. Biomolecules. 2020;10(6):957. doi:10.3390/biom10060957
59. Xie L, Guan Z, Zhang M, et al. Exosomal circLPAR1 promoted osteogenic differentiation of homotypic dental pulp stem cells by competitively binding to hsa-miR-31. Biomed Res Int. 2020;2020:6319395. doi:10.1155/2020/6319395
60. Guo J, Zhou F, Liu Z, et al. Exosome-shuttled mitochondrial transcription factor A mRNA promotes the osteogenesis of dental pulp stem cells through mitochondrial oxidative phosphorylation activation. Cell Prolif. 2022;55(12):e13324. doi:10.1111/cpr.13324
61. Silvestro S, Chiricosta L, Gugliandolo A, et al. Extracellular vesicles derived from human gingival mesenchymal stem cells: a transcriptomic analysis. Genes. 2020;11(2):118. doi:10.3390/genes11020118
62. Luo L, Avery SJ, Waddington RJ. Exploring a chemotactic role for EVs from progenitor cell populations of human exfoliated deciduous teeth for promoting migration of naïve BMSCs in bone repair process. Stem Cells Int. 2021;2021:6681771. doi:10.1155/2021/6681771
63. Zhang Z, Shuai Y, Zhou F, et al. PDLSCs regulate angiogenesis of periodontal ligaments via VEGF transferred by exosomes in periodontitis. Int J Med Sci. 2020;17(5):558–567. doi:10.7150/ijms.40918
64. Ma L, Rao N, Jiang H, et al. Small extracellular vesicles from dental follicle stem cells provide biochemical cues for periodontal tissue regeneration. Stem Cell Res Ther. 2022;13(1):92. doi:10.1186/s13287-022-02767-6
65. Jin Q, Li P, Yuan K, et al. Extracellular vesicles derived from human dental pulp stem cells promote osteogenesis of adipose-derived stem cells via the MAPK pathway. J Tissue Eng. 2020;11:2041731420975569. doi:10.1177/2041731420975569
66. Zhao B, Chen Q, Zhao L, et al. Periodontal ligament stem cell-derived small extracellular vesicles embedded in matrigel enhance bone repair through the adenosine receptor signaling pathway. Int J Nanomed. 2022;17:519–536. doi:10.2147/ijn.S346755
67. Zhou Z, Zheng J, Lin D, Xu R, Chen Y, Hu X. Exosomes derived from dental pulp stem cells accelerate cutaneous wound healing by enhancing angiogenesis via the Cdc42/p38 MAPK pathway. Int J Mol Med. 2022;50(6). doi:10.3892/ijmm.2022.5199
68. Sun J, Wang Z, Liu P, et al. Exosomes derived from human gingival mesenchymal stem cells attenuate the inflammatory response in periodontal ligament stem cells. Front Chem. 2022;10:863364. doi:10.3389/fchem.2022.863364
69. Lan Q, Cao J, Bi X, Xiao X, Li D, Ai Y. Curcumin-primed periodontal ligament stem cells-derived extracellular vesicles improve osteogenic ability through the Wnt/β-catenin pathway. Front Cell Dev Biol. 2023;11:1225449. doi:10.3389/fcell.2023.1225449
70. Nakao Y, Fukuda T, Zhang Q, et al. Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater. 2021;122:306–324. doi:10.1016/j.actbio.2020.12.046
71. Qiao X, Tang J, Dou L, et al. 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. doi:10.2147/ijn.S420967
72. Xie Y, Yu L, Cheng Z, et al. SHED-derived exosomes promote LPS-induced wound healing with less itching by stimulating macrophage autophagy. J Nanobiotechnology. 2022;20(1):239. doi:10.1186/s12951-022-01446-1
73. Cui S, Zhang Z, Cheng C, et al. 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(5):1849–1858. doi:10.1007/s10753-023-01845-y
74. Watanabe Y, Fukuda T, Hayashi C, et al. Extracellular vesicles derived from GMSCs stimulated with TNF-α and IFN-α promote M2 macrophage polarization via enhanced CD73 and CD5L expression. Sci Rep. 2022;12(1):13344. doi:10.1038/s41598-022-17692-0
75. Tian J, Chen W, Xiong Y, et al. Small extracellular vesicles derived from hypoxic preconditioned dental pulp stem cells ameliorate inflammatory osteolysis by modulating macrophage polarization and osteoclastogenesis. Bioact Mater. 2023;22:326–342. doi:10.1016/j.bioactmat.2022.10.001
76. Liu M, Chen R, Xu Y, Zheng J, Wang M, Wang P. Exosomal miR-141-3p from PDLSCs alleviates high glucose-induced senescence of PDLSCs by activating the KEAP1-NRF2 signaling pathway. Stem Cells Int. 2023;2023:7136819. doi:10.1155/2023/7136819
77. Yu T, Mi N, Song Y, Xie W. Exosomes miR-92a-3p from human exfoliated deciduous teeth inhibits periodontitis progression via the KLF4/PI3K/AKT pathway. J Periodontal Res. 2024;59(4):771–782. doi:10.1111/jre.13262
78. Zhang Y, Chen J, Fu H, et al. Exosomes derived from 3D-cultured MSCs improve therapeutic effects in periodontitis and experimental colitis and restore the Th17 cell/Treg balance in inflamed periodontium. Int J Oral Sci. 2021;13(1):43. doi:10.1038/s41368-021-00150-4
79. Yan C, Li N, Xiao T, et al. Extracellular vesicles from the inflammatory microenvironment regulate the osteogenic and odontogenic differentiation of periodontal ligament stem cells by miR-758-5p/LMBR1/BMP2/4 axis. J Transl Med. 2022;20(1):208. doi:10.1186/s12967-022-03412-9
80. Diomede F, D’Aurora M, Gugliandolo A, et al. A novel role in skeletal segment regeneration of extracellular vesicles released from periodontal-ligament stem cells. Int J Nanomed. 2018;13:3805–3825. doi:10.2147/ijn.S162836
81. Pizzicannella J, Gugliandolo A, Orsini T, et al. Engineered extracellular vesicles from human periodontal-ligament stem cells increase VEGF/VEGFR2 expression during bone regeneration. Front Physiol. 2019;10:512. doi:10.3389/fphys.2019.00512
82. Liu Y, Zhuang X, Yu S, et al. Exosomes derived from stem cells from apical papilla promote craniofacial soft tissue regeneration by enhancing Cdc42-mediated vascularization. Stem Cell Res Ther. 2021;12(1):76. doi:10.1186/s13287-021-02151-w
83. Suwittayarak R, Klincumhom N, Ngaokrajang U, et al. Shear stress enhances the paracrine-mediated immunoregulatory function of human periodontal ligament stem cells via the ERK signalling pathway. Int J Mol Sci. 2022;23(13):7119. doi:10.3390/ijms23137119
84. Wu M, Liu X, Li Z, et al. SHED aggregate exosomes shuttled miR-26a promote angiogenesis in pulp regeneration via TGF-β/SMAD2/3 signalling. Cell Prolif. 2021;54(7):e13074. doi:10.1111/cpr.13074
85. 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. doi:10.2147/ijn.S350869
86. Arigita C, Jiskoot W, Westdijk J, et al. Stability of mono- and trivalent meningococcal outer membrane vesicle vaccines. Vaccine. 2004;22(5–6):629–642. doi:10.1016/j.vaccine.2003.08.027
87. Kim JY, Suh JW, Kang JS, Kim SB, Yoon YK, Sohn JW. Gram-negative bacteria’s outer membrane vesicles. Infect Chemother. 2023;55(1):1–9. doi:10.3947/ic.2022.0145
88. Nakao R, Hasegawa H, Dongying B, Ohnishi M, Senpuku H. Assessment of outer membrane vesicles of periodontopathic bacterium Porphyromonas gingivalis as possible mucosal immunogen. Vaccine. 2016;34(38):4626–4634. doi:10.1016/j.vaccine.2016.06.016
89. Chen H, Wang L, Zeng X, et al. Exosomes, a new star for targeted delivery. Front Cell Dev Biol. 2021;9:751079. doi:10.3389/fcell.2021.751079
90. Gao Y, Yuan Z, Yuan X, et al. Bioinspired porous microspheres for sustained hypoxic exosomes release and vascularized bone regeneration. Bioact Mater. 2022;14:377–388. doi:10.1016/j.bioactmat.2022.01.041
91. Xu XY, Tian BM, Xia Y, et al. Exosomes derived from P2X7 receptor gene-modified cells rescue inflammation-compromised periodontal ligament stem cells from dysfunction. Stem Cells Transl Med. 2020;9(11):1414–1430. doi:10.1002/sctm.19-0418
92. Yerneni SS, Adamik J, Weiss LE, Campbell PG. Cell trafficking and regulation of osteoblastogenesis by extracellular vesicle associated bone morphogenetic protein 2. J Extracell Vesicles. 2021;10(12):e12155. doi:10.1002/jev2.12155
93. Luo H, Chen D, Li R, et al. Genetically engineered CXCR4-modified exosomes for delivery of miR-126 mimics to macrophages alleviate periodontitis. J Nanobiotechnology. 2023;21(1):116. doi:10.1186/s12951-023-01863-w
94. Ridolfi A, Cardellini J, Gashi F, et al. Electrostatic interactions control the adsorption of extracellular vesicles onto supported lipid bilayers. J Colloid Interface Sci. 2023;650(Pt A):883–891. doi:10.1016/j.jcis.2023.07.018
95. Chen Y, Hou S. Recent progress in the effect of magnetic iron oxide nanoparticles on cells and extracellular vesicles. Cell Death Discov. 2023;9(1):195. doi:10.1038/s41420-023-01490-2
96. Lu Y, Yang Y, Liu S, Ge S. Biomaterials constructed for MSC-derived extracellular vesicle loading and delivery-a promising method for tissue regeneration. Front Cell Dev Biol. 2022;10:898394. doi:10.3389/fcell.2022.898394
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

Potential Pathogenetic Role of Antimicrobial Peptides Carried by Extracellular Vesicles in an in vitro Psoriatic Model
Capriotti L, Iuliano M, Lande R, Frasca L, Falchi M, Rosa P, Mangino G, Romeo G
Journal of Inflammation Research 2022, 15:5387-5399
Published Date: 16 September 2022
Extracellular Vesicles in Idiopathic Pulmonary Fibrosis: Pathogenesis, Biomarkers and Innovative Therapeutic Strategies
Yang Y, Lv M, Xu Q, Wang X, Fang Z
International Journal of Nanomedicine 2024, 19:12593-12614
Published Date: 25 November 2024