Skip to main content
Erschienen in: Journal of Translational Medicine 1/2022

Open Access 01.12.2022 | Review

Advances of exosomes in periodontitis treatment

verfasst von: Hongbing Lin, Huishan Chen, Xuetao Zhao, Tong Ding, Yawei Wang, Zhen Chen, Yue Tian, Peipei Zhang, Yuqin Shen

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2022

Abstract

Periodontitis is an inflammatory disease initiated by dysbiosis of the local microbial community. Periodontitis can result in destruction of tooth-supporting tissue; however, overactivation of the host immune response is the main reason for alveolar bone loss. Periodontal tissue cells, immune cells, and even further activated osteoclasts and neutrophils play pro-inflammatory or anti-inflammatory roles. Traditional therapies for periodontitis are effective in reducing the microbial quantities and improving the clinical symptoms of periodontitis. However, these methods are non-selective, and it is still challenging to achieve an ideal treatment effect in clinics using the currently available treatments and approaches. Exosomes have shown promising potential in various preclinical and clinical studies, including in the diagnosis and treatment of periodontitis. Exos can be secreted by almost all types of cells, containing specific substances of cells: RNA, free fatty acids, proteins, surface receptors and cytokines. Exos act as local and systemic intercellular communication medium, play significant roles in various biological functions, and regulate physiological and pathological processes in numerous diseases. Exos-based periodontitis diagnosis and treatment strategies have been reported to obtain the potential to overcome the drawbacks of traditional therapies. This review focuses on the accumulating evidence from the last 5 years, indicating the therapeutic potential of the Exos in preclinical and clinical studies of periodontitis. Recent advances on Exos-based periodontitis diagnosis and treatment strategies, existing challenges, and prospect are summarized as guidance to improve the effectiveness of Exos on periodontitis in clinics.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
MNPs
mononuclear phagocytes
APCs
antigen-presenting cells
SRP
scaling and root planning
MSCs
mesenchymal stem cells
DCs
dendritic cells
EVs
extracellular vesicles
SNARE
soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor
ILVs
intracavitary vesicles
MVBs
multivesicular bodies
NTA
nanoparticle tracking analysis
WB
western blotting
FC
flow cytometry
AFM
atomic force microscopy
miRNAs
microRNAs
PD-L1
programmed death-ligand 1
SP
severe periodontitis
DPSCs
dental pulp stem cells
HHH-DPCs
haplotype homo dental pulp cell lines
PDLSCs
periodontal ligament stem cells
GMSCs
gingival mesenchymal stem cells
SHED
human exfoliated deciduous teeth
BMSCs
bone marrow stromal cells
ADSCs
adipose-derived stem cells
ROS
reactive oxygen species
hUCMSCs
human umbilical cord mesenchymal stem cells
TRAF1
tumor necrosis factor receptor-associated factor 1
LECs
lymphatic endothelial cells
ECs
endothelial cells
CMS
cyclic mechanical stretch
EPCs
endothelial progenitor cells
GMP
good manufacturing practice

Background

Periodontitis is an inflammatory disease initiated by dysbiosis of the local microbial community and is characterized by the relative abundance or influence imbalance of microbial species [1]. However, overactivation of the host immune response is the main reason for the direct activation of osteoclast activity and alveolar bone loss [2]. The composition and total number of microbiota change after the colonization of "keystone" pathogen, which improves the pathogenicity of the whole community. Thus, the immune response is overactivated, resulting in immune cells infiltration, activation of osteoclast activity, and destruction of soft and hard tissues [3]. Pathological immunity of the host to dysbiotic microbes first occurs between the microbiome and host cells, which include periodontal tissue cells and other immune cells, such as mononuclear phagocytes (MNPs), antigen-presenting cells (APCs), and specific T cell subsets. Naive T cells and B cells not only differentiate into mature T cells or plasma cells but also further activate or promote osteoclasts and neutrophils to play a pro-inflammatory or anti-inflammatory role [4] (Fig. 1).
Traditional therapies for periodontitis include scaling and root planning (SRP), systemic and local administration of antibiotics, and oral antiseptics. In the short term, these therapies are usually effective in reducing the microbial quantities in the tissues and blood of periodontitis patients and improving the clinical symptoms of periodontitis. However, these methods are non-selective, and the oral and systemic effects of long-term medication need to be evaluated [5]. It is becoming increasingly clear that new strategies need to be developed to treat periodontitis more effectively.
Exos were discovered in 1987 and have been shown to be important in cell communication [6]. Other studies have confirmed that Exos are a natural nanoparticle delivery method that can treat multiple infectious and immune/inflammatory diseases [710]. Thus, in this review, we summarized studies from the last 5 years that have focused on the effect of Exos on periodontitis and host cells.

Exos

The biogenesis of Exos

Exos are secreted by almost all types of cells, including mesenchymal stem cells (MSCs), dendritic cells (DCs), B cells, T cells, and mast cells, and widely exist in many body fluids such as plasma, urine, breast milk, semen, amniotic fluid, and saliva [11]. Exos are among the three major types of extracellular vesicles (EVs). The size of the Exos is between 50 and 150 nm and the density is between 1.15 and 1.19 g / ml [12]. The biogenesis and release of Exos are different from other of other microbubbles. While other microbubbles are released directly through the plasma membrane, Exos originating from endocytosis [13]. First, the membrane of secretory cells is sunken inward to produce endocytic vesicles. Multiple endocytic vesicles combine to form early nucleosomes, and then miRNA, mRNA and DNA are packaged in the cytoplasm to form late endocytosis vesicles. The late endocytic vesicles germinate inward to form intracavitary vesicles (ILVs). The aggregation of ILVs in late endosomes create multivesicular bodies (MVBs), which are formed by the inward invagination of the endosomal limiting membrane. Finally, some MVBs with low cholesterol were degraded by lysosomes, and some MVBs, rich in cholesterol, release extracellular bodies through the fusion of the cell membrane with itself [1316]. The active formation of Exos is dependent on the endosomal sorting complex required for transport (ESCRT; ESCRT-0, I, II, III and Vps4) and its accessory proteins (Alix, TSG101, HSC70, and HSP90β). They recognize ubiquitinated transmembrane proteins and incorporate endosomal proteins into MVBs. [17, 18]. Passive formation of Exos is independent of ESCRT and involves lipids (ceramide), tetrapeptides (CD63) and heat shock proteins which induce cell membrane budding and promote MVB formation [19, 20]. In addition, certain components, such as four-transmembrane domain proteins and lipid rafts, have been reported to participate in the formation of some exosomes [21, 22]. In addition to the classic pathway, there is a much more immediate route of exosome biogenesis. T cells and erythroleukemia cell lines can release exosomes from the plasma membrane directly, and the Exos produced by these two pathways cannot be distinguished [23, 24]. Furthermore, soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor (SNARE) proteins and their effectors such as Rab GTPases (Rab27a, Rab27b and Rab35) play a significant role in exosome secretion [14].
The biogenesis of Exos is affected by many external factors, including cell type, serum conditions, cytokines, and growth factors. The heterogeneity of Exos is based on their specific morphology, content, and function [25, 26]. Encapsulated by lipid membranes derived from parental cells, Exos show inherent histocompatibility and tissue orientation mediated by surface molecules such as integrin and glycans [27]. In addition, Exos contain four transmembrane proteins (CD9, CD63, CD81 and CD82), heat shock proteins, lipoproteins, and some transport-related proteins. These proteins can not only provide markers for Exos identification but also locate Exos in specific target cells [28, 29]. Exos contain substances to mother cells, such as RNA, free fatty acids, proteins, surface receptors, and cytokines, which act as local and systemic intercellular communication media [30, 31]. Owing to these characteristics, Exos play a significant role in various biological functions and regulate many physiological and pathological processes in numerous diseases [32] (Fig. 2).

The isolation and characterization of Exos

The selection and improvement of isolation strategies should be determined according to the kind of biological fluids Exos are isolated from. Effective isolation strategies should be able to concentrate the signals of the Exos to be analyzed and avoid contamination with other molecules, such as lipoproteins, non-vesicular protein aggregates, and other EVs, which are similar to Exos in terms of size and density. At present, there are many techniques for separating and purifying Exos from biological fluids and in vitro cell cultures, including ultracentrifugation, size-based separation, Exos precipitation, immunoaffinity capture-based techniques, and microfluidic isolation [3336]. Among these, differential centrifugation is the most widely used and basic method for Exos separation, and it is a feasible strategy to combine two or more methods to improve the yield, purity, and efficiency of Exos extracrion. Various techniques based on biophysical or biological characteristics have been used to verify upstream Exos separation methods and classify Exos subgroups for downstream analysis, such as nanoparticle tracking analysis (NTA), western blotting (WB), flow cytometry (FC), and atomic force microscopy (AFM) [3537] (Fig. 3).

Exos based periodontitis diagnostic and treatment strategy

Periodontitis is a major public health problem with a high incidence rate worldwide. It could not only cause destruction of the supporting tissues of teeth but also have a negative effect on systematic disease states [38]. Therefore, effective diagnosis and treatment of periodontitis are important to reduce the risk of periodontitis.

Exos-based periodontitis diagnostic strategy

Because the components of Exos can be reprogrammed according to disease status, Exos are increasingly being evaluated as potential diagnostic biomarkers for the diagnosis and prognosis of diseases. These characteristics have made Exos a focus of oral disease research in recent years, including periodontitis [39].
At the gene level, Exos are enriched in specific microRNAs (miRNAs), that can provide disease-specific diagnostic signatures [40]. When compared to healthy controls, plasma-derived exosomal miRs (miR-1304-3p and miR-200c-3p) and snoRs (SNORD57 and SNODB1771) from periodontitis patients are differentially expressed and could be the valuable biomarkers for periodontitis diagnosis [41]. In addition, the level of programmed death-ligand 1 (PD-L1) mRNA in salivary Exos may have the potential to diagnose periodontitis and is relative to the severity of periodontitis [42].
At the protein level, detection and analysis of salivary exosomal proteins in young adults with severe periodontitis (SP) suggested that C6 proteins, which participate in the immune response during the development of periodontitis, were expressed only in the SP group [43].
In addition, levels of CD9 and CD81 Exos in periodontitis patients were significantly lower than those in the healthy controls. Because the concentration of CD9/CD81 Exos in saliva is significantly and negatively correlated with clinical measurements, it may be of great significance in the pathogenesis of periodontal disease [44]. Other advances in salivary Exos in the diagnosis of periodontitis are well-reviewed in the literature [45].

Exos-based periodontitis treatment strategy

Exos have been reported to provide a novel perspective and potential therapeutic approach for treating periodontitis and improving alveolar resorption. Exos derived from 3D-cultured MSCs restored not only the Th17 cell/Treg balance through the miR-1246/Nfat5 axis, but also the immune responses in the inflamed periodontium [46]. Dental pulp stem cell-Exo (DPSC-Exos) can facilitate the conversion of macrophages from a pro-inflammatory phenotype (M1)to an anti-inflammatory phenotype (M2) and promote the healing of alveolar bone in mice with periodontitis, the mechanism of which could be associated with miR-1246 in DPSC-Exos [47]. Exos purified from human leukocyte antigen haplotype homo dental pulp cell lines (HHH-DPCs) stimulated the migration of human DPCs and mouse osteoblastic and significantly suppressed osteoclast formation in vitro [48]. Exos secreted from healthy periodontal ligament stem cells (PDLSCs) promote osteogenic differentiation of PDLSCs derived from periodontitis tissue. Healthy PDLSC-Exos (h-PDLSC-Exos) treatment resulted in accelerated bone formation in alveolar bone defects in rat models of periodontitis. Mechanistically, h-PDLSC-Exos suppressed the overactivation of canonical Wnt signaling to recover the osteogenic differentiation capacity of inflammatory PDLSCs [49]. Exos derived from TNF-α-preconditioned gingival mesenchymal stem cells (GMSCs) could significantly regulate inflammation and osteoclastogenesis, which could provide a therapeutic approach for periodontitis [50]. Human exfoliated deciduous teeth (SHED)-derived Exos (SHED-Exos) restored bone loss in mouse periodontitis model and promoted bone marrow stromal cells (BMSCs) osteogenesis, differentiation, and bone formation [51]. SHED-Exos contribute to periodontal bone regeneration by promoting neovascularization and new bone formation, possibly through the AMPK signaling pathway [52]. Exos from reparative M2 macrophages reduced alveolar bone resorption in mice with periodontitis via the IL-10/IL-10R pathway [53]. Exos derived from adipose-derived stem cells (ADSC-Exos) represent a promising adjunctive treatment to SRP in rats [54]. Exosomal miR-25-3p in saliva contributes to the development and progression of diabetes-associated periodontitis. The discovery of other miR-25-3p targets may provide critical insights into the development of drugs to treat periodontitis by regulating γδ T cell-mediated local inflammation [55]. The in vivo effects of Exos on periodontitis are summarized in Table 1.
Table 1
Summary of in vivo results showing the Exos-based periodontitis treatment strategy used over the past 5 years
Source of Exos
Study model
Route of delivery
Dose
Duration
Outcomes
References
MSCs
Mouse experimental periodontitis model
Locally injection
50 μg per mouse
14 days
Improve the treatment by restoring the Th17 cell/Treg balance through the miR-1246/Nfat5 axis
[46]
DPSCs
Mouse experimental periodontitis model
Incorporated chitosan hydrogel
50 μg
4 weeks
Facilitate macrophages from M1 to M2 phenotype and promote alveolar bone healing
[47]
HHH-DPSCs
Mouse experimental periodontitis model
Directly applied onto the silk ligature
5 μL containing 7.5 × 108 particles
7 days
Promote the migration of both DPCs and osteoblastic cells; suppress osteoclast formation
[48]
PDLSCs
Rat periodontal bone defect model
Mixed with Matrigel
Exos (225 μg/μL): Matrigel = 2:1 (v/v)
4 weeks
Suppress overactivation Wnt signaling, recover osteogenic differentiation capacity of inflammatory PDLSCs
[49]
TNF-α-treated human GMSCs
Mouse experimental periodontitis model
Locally injection
20 μg per mouse
7 days
Regulate inflammation and osteoclastogenesis
[50]
SHED
Mouse experimental periodontitis model
Locally injection
20 μg
2 weeks
Restore bone loss, promote BMSCs osteogenesis, differentiation, and bone formation
[51]
SHED
Rat periodontal defect models
h β-TCP scaffffolds loaded with Exos
2 μg/μL Exos in 100 μL PBS
4 weeks
Contribute to periodontal bone regeneration through the AMPK signaling pathway
[52]
induced M2-like macrophages
Mouse experimental periodontitis model
Locally injection
30 μL (500 ng/ml)
2 weeks
Reduce alveolar bone resorption in mice with periodontitis via IL-10/IL-10R pathway
[53]
ADSCs
Rat experimental periodontitis model
Locally injection
80–150 µg in 200 µL PBS
4 weeks
Represent a promising adjunctive treatment to SRP
[54]
salivary Exos
Insulin resistance-associated mouse experimental periodontitis model
Locally injection
miR-25-3p inhibitors (100 μl of 8 nM)
9 days
Exosomal miR-25-3p in saliva contribute to development and progression of diabetes-associated periodontitis
[55]

Possible mechanism of Exos on host cells during periodontitis

Overactivation of the host immune response is caused by the interaction between the dysbiosis of local microbes and host cells, eventually leading to periodontal tissue destruction. The host cells include periodontal tissue cells and other immune cells, which play pro-inflammatory or anti-inflammatory roles [4]. Exos retain proteins, miRNA, mRNA, DNA, and lipids, and can transfer that cargo to distant target cells and modify the target cells. Reports have revealed the biological activities of Exos in modifying host cells (Fig. 4). Modification of host cells with Exos from different sources plays an important role in the treatment of periodontitis.

Effects of Exos on neutrophils

Neutrophils are short-lived cells in the innate immune system. They play an important role in pathogen resistance by producing reactive oxygen species (ROS). Therefore, effective strategies to improve the viability and function of neutrophils may be beneficial for treating infections and immune deficiency diseases. MSC-Exos have a protective effect on neutrophil function and lifespan [56], and could significantly reduce the terminal complement activation complex C5b-9 to inhibit neutrophils accumulation [57]. Exos isolated from ADSCs (ADSC-Exos) can decrease neutrophil apoptosis and increase phagocytosis [58]. Exos isolated from LPS-treated macrophages can induce cytokine production and neutrophils migration [59].

Effects of Exos on macrophages

MSC-Exos can modify the polarization of the pro-inflammatory phenotype (M1 macrophages) to the anti-inflammatory phenotype (M2 macrophages) via shuttling miR-182 [60]. Exos derived from BMSCs (BMSC-Exos) can increase M2 macrophages [61], and BMSC-Exos have been reported to inhibit M1 macrophages and promote M2 macrophages in a murine alveolar macrophage cell line by inhibiting cellular glycolysis [62]. FNDC5 pre-conditioned BMSC-Exos have also been confirmed to play an anti-inflammatory role and promote M2 macrophages via NF-κB signaling pathway and the Nrf2/HO-1 axis [63]. Exos from human umbilical cord mesenchymal stem cells (hUCMSC-Exos) facilitated CD163 + M2 macrophages [64] and promoted M2 macrophages in LPS-stimulated RAW 264.7 via tumor necrosis factor receptor-associated factor 1 (TRAF1) [65]. ADSC-Exos can significantly upregulate the mRNA expression of M2 macrophages [66], induce M2 macrophages through the transactivation of Arg-1 by Exos-carried active STAT3 [67], and alleviate LPS induced inflammation by regulating Nrf2/HO-1 expression (68). Exos from GMSCs (GMSC-Exos) and dental pulp stem cells (DPSC-Exos) can promote the transformation of macrophages from M1 to M2 [47, 69]. TNF-α stimulated GMSC-Exos have also been reported to induce anti-inflammatory M2 macrophage polarization [47, 50].

Effects of Exos on DCs

MSC-Exos decreased DC surface marker expression in cells treated with LPS and decreased lymphocyte proliferation in the presence of MSC-Exos treated DCs, suggesting that MSC-Exos may play a key role in DC-induced immune responses [70]. hUCMSC-Exos suppressed the maturation and activation of DC and decreased the expression of IL-23, which is particularly important for promoting the pathogenicity of Th17 cells [71]. Exos from RegDC (RegDC-Exos) suppress the maturation of DCs and promote the recruitment of Treg cells, resulting in the inhibition of bone resorptive cytokines and reduction in osteoclastic bone loss [72]. Exos from lymphatic endothelial cells (LEC-Exos) promote the directional migration of human DCs in complex tissue environments in a CX3CL1/fractalkine-dependent fashion [73].

Effects of Exos on T lymphocytes

MSC-Exos decreased T lymphocyte proliferation and the percentage of CD4 + and CD8 + T cell subsets in a dose-dependent manner while increasing Treg cell populations [74]. MSC-Exos promote the proliferation and immune-suppression capacity of Tregs by upregulating IL-10 and TGF-β1[75], and inhibit the differentiation of Th2 cells by regulating the miR-146a-5p/SERPINB2 pathway [76]. PDLSC-Exos alleviated inflammatory microenvironment and maintained Th17/Treg balance via the Th17/Treg/miR‐155‐5p/SIRT1 regulatory network [77]. CD137-modified endothelial cell-Exos (EC-Exos) promote Th17 cell differentiation via the NF-КB pathway by regulating IL-6 expression [78].

Effects of Exos on B lymphocytes

MSC-Exos had the beneficial effect of reducing plasmablasts and incresing Breg-like cells in lymph nodes [74].

Effects of Exos on osteoclasts

Exosomal miR-1260b of TNF-α-preconditioned GMSC-Exos was found to inhibit osteoclastogenic activity by targeting the Wnt5a-mediated RANKL pathway [50]. RegDC-Exos inhibit the production of bone resorptive cytokines and bone loss in osteoclasts [72]. Cyclic mechanical stretch (CMS)-treated BMSC-Exos can impair osteoclast differentiation by inhibiting the RANKL-NF-κB signaling pathway [79]. ADSC-Exos can reduce bone resorption and recover bone loss by suppressing NLRP3 inflammasome activation in osteoclasts [80] and by antagonizing osteocyte-mediated osteoclastogenesis [81]. ADSC-Exos combined with microRNA-146a (miR-146a-Exo) were reported to restrain bone resorption by inhibiting pro-inflammatory cytokine production in high glucose-treated osteoclasts [82]. Exos derived from osteoblasts can inhibit osteoclast differentiation via the miR-503-3p/Hpse axis [83]. Exos from endothelial progenitor cells (EPC-Exos) can promote bone repair by enhancing the recruitment and differentiation of osteoclast precursors via LncRNA-MALAT1 [84].
The effects of Exos on the host cells involved in periodontitis are summarized in Table 2.
Table 2
Summary of the effects of Exos on host cells
No
Source of Exos
Biological activity
References
Neutrophil
1
MSCs
Have protective effects on neutrophil function and lifespan
[56]
2
MSCs
Reduce terminal complement activation complex C5b-9 to inhibit neutrophils accumulation
[57]
3
ADSCs
Decrease neutrophils apoptosis and increased their phagocytosis capacity
[58]
4
LPS-treated macrophages
Induce cytokine production and neutrophil migration
[59]
Macrophage
1
DPSCs
Facilitate macrophages to convert from M1 phenotype to M2 phenotype
[47]
2
TNF-α induced GMSCs
Induce anti-inflammatory M2 macrophage polarization
[50]
3
MSCs
Modify the polarization of M1 macrophages to M2 macrophages via shuttling miR-182
[60]
4
BMSCs
Increase M2 macrophage polarization
[61]
5
BMSCs
Inhibit M1 polarization and promotes M2 polarization in a murine alveolar macrophage cell line by inhibiting cellular glycolysis
[62]
6
FNDC5 pre-conditioned BMSCs
Play anti-inflammation effects and promote M2 macrophage polarization via NF-κB signaling pathway and Nrf2/HO-1 axis
[63]
7
hUCMSCs
Facilitate CD163 + M2 macrophage polarization, reduced inflammation, and increases anti-inflammatory responses
[64]
8
hUCMSCs
Inhibit M1 polarization and promoted M2 polarization through tumor necrosis factor receptor-associated factor 1 (TRAF1)
[65]
9
ADSCs
Upregulate mRNA expression of M2 macrophages
[66]
10
ADSCs
Induce anti-inflammatory M2 phenotypes through the transactivation of arginase-1 by Exo-carried active STAT3
[67]
11
ADSCs
Polarize macrophage to an anti-inflammatory phenotype via regulating the Nrf2/HO-1 expression
[68]
12
GMSCs
Facilitate macrophages to convert from M1 phenotype to M2 phenotype
[69]
Dendritic cell
1
MSCs
Decrease DC surface marker expression and modulates DC-induced immune responses
[70]
2
hUCMSCs
Suppress maturation and activation of DCs, and decreases the expression level of IL-23
[71]
3
regDCs
Suppress maturation of recipient DCs resulting in inhibition of bone resorptive cytokines
[72]
4
LECs
Promote the directional migratory in a CX3CL1/fractalkine-dependent fashion
[73]
T lymphocyte
1
MSCs
Increase Treg cell populations, inhibit T lymphocyte proliferation in a dose-dependent manner and decreases the percentage of CD4 + and CD8 + T cell subsets
[74]
2
MSCs
Upregulate IL-10 and TGF-β1 to promote proliferation and immune-suppression capacity of Tregs
[75]
3
MSCs
Inhibit the differentiation of Th2 cells via the regulation of the miR-146a-5p/SERPINB2 pathway
[76]
4
PDLSCs
Alleviate inflammatory microenvironment and keep Th17/Treg balance via Th17/Treg/miR‐155‐5p/SIRT1 regulatory network
[77]
5
CD137-modified ECs
Promote Th17 cell differentiation via NF-КB pathway mediated IL-6 expression
[78]
B lymphocyte
1
MSCs
Upregulate Breg-like cells in lymph nodes
[74]
Osteoclast
1
TNF-α-preconditioned GMSCs
Inhibit osteoclastogenic activity via exosomal miR-1260b to target Wnt5a-mediated RANKL pathway and
[50]
2
regDC
Result in inhibition of bone resorptive cytokines and reduces in osteoclastic bone loss
[72]
3
CMS-treated BMSCs
Impair osteoclast differentiation via inhibiting the RANKL-induced nuclear factor kappa-B (NF-κB) signaling pathway
[79]
4
ADSCs
Suppress NLRP3 inflammasome activation in osteoclasts and reduces bone resorption and recover bone loss
[80]
5
ADSCs
Antagonize osteocyte-mediated osteoclastogenesis
[81]
6
ADSCs
Inhibit pro-inflammatory cytokines production in high glucose-treated osteoclasts and restrains bone resorption
[82]
7
osteoblast
Inhibit the osteoclast differentiation via miR-503-3p/Hpse axis
[83]
8
EPCs
Promote bone repair by enhancing recruitment and differentiation of osteoclast precursors through LncRNA-MALAT1
(84)

Summary and prospects

Exos can be secreted by almost all cell types and are the main contributor to cells efficacy. They are natural carriers of functional small RNA and proteins [85], and the constituents can be reprogrammed depending on the disease state [39]. Therefore, potential applications of Exos in the diagnosis and treatment of diseases are becoming increasingly popular. Exos derived from MSCs, with or without biomaterials, have broad application prospects in the treatment of periodontitis, especially in the cell-free treatment of tissue regeneration. Among them, Exos derived from oral stem cells are easier to collect and may show excellent characteristics of immune regulation, repair, and regeneration as well as less ethical, moral, or safety limits [12, 86]. In this review, we summarized the novel strategies using Exos in periodontitis over the last 5 years and analyze the possible mechanism of Exos in the treatment of periodontitis by summarizing the effect of Exos on host cells involved in the process of periodontitis.
Although the applications of Exos in periodontitis has been proved to be useful in animal models of preclinical research, much work needs to be done to apply it to clinics. Originally, Exos in clinical trials had to comply with good manufacturing practice (GMP), which includes the upstream of the cell culture process, the downstream of the purification process, and the quality control of Exos. The content carried by Exos varies from cell type to culture conditions and batch, which causes differences in biological functions. Therefore, it is necessary to explore more convenient and efficient technologies for the separation, purification, and storage of Exos to improve their homogeneity, purity, and repeatability. Furthermore, the corresponding role and mechanism of Exos in the diagnosis and treatment of periodontitis need to be explored more comprehensively. The critical range of differential expression of exosomes in periodontal tissue under healthy and inflammatory conditions for diagnosis and the amount and duration of safe and effective treatment need to be defined. Ultimately, the mechanisms of interaction between Exos and host cells are not clear, which makes it impossible for Exos to accurately regulate the target cells and functions. However, there is no doubt that Exos have the potential to provide personalized medical strategies for the prevention and treatment of periodontitis.

Conclusions

Exos contain specific substances in their cells and play a significant role in the diagnosis and treatment of numerous diseases, including periodontitis. Exos-based periodontitis treatment strategies have been reported to obtain the potential to overcome the drawbacks of traditional therapies and have tremendous prospect for bench-to-bed translation.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
3.
Zurück zum Zitat Hajishengallis G. 2020 New developments in neutrophil biology and periodontitis. Periodontol. 2000;82(1):78–92.CrossRef Hajishengallis G. 2020 New developments in neutrophil biology and periodontitis. Periodontol. 2000;82(1):78–92.CrossRef
6.
Zurück zum Zitat Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262(19):9412–20.PubMedCrossRef Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262(19):9412–20.PubMedCrossRef
7.
Zurück zum Zitat Kibria G, Ramos EK, Wan Y, Gius DR, Liu H. exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol Pharm. 2018;15(9):3625–33.PubMedPubMedCentralCrossRef Kibria G, Ramos EK, Wan Y, Gius DR, Liu H. exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol Pharm. 2018;15(9):3625–33.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Ahmed F, Tamma M, Pathigadapa U, Reddanna P, Yenuganti VR. Drug loading and functional efficacy of cow, buffalo, and goat milk-derived exosomes: a comparative study. Mol Pharm. 2022;19(3):763–74.PubMedCrossRef Ahmed F, Tamma M, Pathigadapa U, Reddanna P, Yenuganti VR. Drug loading and functional efficacy of cow, buffalo, and goat milk-derived exosomes: a comparative study. Mol Pharm. 2022;19(3):763–74.PubMedCrossRef
10.
Zurück zum Zitat Kanlikilicer P. Exosome-related methods and potential use as vaccines. Methods Mol Biol. 2022;2435:35–41.PubMedCrossRef Kanlikilicer P. Exosome-related methods and potential use as vaccines. Methods Mol Biol. 2022;2435:35–41.PubMedCrossRef
14.
Zurück zum Zitat Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):47.PubMedPubMedCentralCrossRef Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):47.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193–208.PubMedCrossRef Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193–208.PubMedCrossRef
16.
Zurück zum Zitat Mobius W, Ohno-Iwashita Y, van Donselaar EG, Oorschot VM, Shimada Y, Fujimoto T, et al. Immunoelectron microscopic localization of cholesterol using biotinylated and non-cytolytic perfringolysin O. J Histochem Cytochem. 2002;50(1):43–55.PubMedCrossRef Mobius W, Ohno-Iwashita Y, van Donselaar EG, Oorschot VM, Shimada Y, Fujimoto T, et al. Immunoelectron microscopic localization of cholesterol using biotinylated and non-cytolytic perfringolysin O. J Histochem Cytochem. 2002;50(1):43–55.PubMedCrossRef
18.
Zurück zum Zitat Zhang Y, Bi J, Huang J, Tang Y, Du S, Li P. Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int J Nanomedicine. 2020;15:6917–34.PubMedPubMedCentralCrossRef Zhang Y, Bi J, Huang J, Tang Y, Du S, Li P. Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int J Nanomedicine. 2020;15:6917–34.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–7.PubMedCrossRef Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–7.PubMedCrossRef
20.
Zurück zum Zitat Wei D, Zhan W, Gao Y, Huang L, Gong R, Wang W, et al. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021;31(2):157–77.PubMedCrossRef Wei D, Zhan W, Gao Y, Huang L, Gong R, Wang W, et al. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021;31(2):157–77.PubMedCrossRef
21.
Zurück zum Zitat de Gassart A, Geminard C, Fevrier B, Raposo G, Vidal M. Lipid raft-associated protein sorting in exosomes. Blood. 2003;102(13):4336–44.PubMedCrossRef de Gassart A, Geminard C, Fevrier B, Raposo G, Vidal M. Lipid raft-associated protein sorting in exosomes. Blood. 2003;102(13):4336–44.PubMedCrossRef
22.
Zurück zum Zitat Rana S, Zoller M. Exosome target cell selection and the importance of exosomal tetraspanins: a hypothesis. Biochem Soc Trans. 2011;39(2):559–62.PubMedCrossRef Rana S, Zoller M. Exosome target cell selection and the importance of exosomal tetraspanins: a hypothesis. Biochem Soc Trans. 2011;39(2):559–62.PubMedCrossRef
23.
Zurück zum Zitat Booth AM, Fang Y, Fallon JK, Yang JM, Hildreth JE, Gould SJ. Exosomes and HIV Gag bud from endosome-like domains of the T cell plasma membrane. J Cell Biol. 2006;172(6):923–35.PubMedPubMedCentralCrossRef Booth AM, Fang Y, Fallon JK, Yang JM, Hildreth JE, Gould SJ. Exosomes and HIV Gag bud from endosome-like domains of the T cell plasma membrane. J Cell Biol. 2006;172(6):923–35.PubMedPubMedCentralCrossRef
24.
26.
Zurück zum Zitat Gurunathan S, Kang MH, Kim JH. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomedicine. 2021;16:1281–312.PubMedPubMedCentralCrossRef Gurunathan S, Kang MH, Kim JH. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomedicine. 2021;16:1281–312.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Busatto S, Pham A, Suh A, Shapiro S, Wolfram J. Organotropic drug delivery: Synthetic nanoparticles and extracellular vesicles. Biomedical Microdevices. 2019;21(2):1–17.CrossRef Busatto S, Pham A, Suh A, Shapiro S, Wolfram J. Organotropic drug delivery: Synthetic nanoparticles and extracellular vesicles. Biomedical Microdevices. 2019;21(2):1–17.CrossRef
28.
Zurück zum Zitat Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.PubMedCrossRef Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.PubMedCrossRef
30.
Zurück zum Zitat Meldolesi J. Exosomes and ectosomes in intercellular communication. Curr Biol. 2018;28(8):R435–44.PubMedCrossRef Meldolesi J. Exosomes and ectosomes in intercellular communication. Curr Biol. 2018;28(8):R435–44.PubMedCrossRef
31.
Zurück zum Zitat Donoso-Quezada J, Ayala-Mar S, Gonzalez-Valdez J. The role of lipids in exosome biology and intercellular communication: Function, analytics and applications. Traffic. 2021;22(7):204–20.PubMedPubMedCentralCrossRef Donoso-Quezada J, Ayala-Mar S, Gonzalez-Valdez J. The role of lipids in exosome biology and intercellular communication: Function, analytics and applications. Traffic. 2021;22(7):204–20.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics. 2020;10(8):3684–707.PubMedPubMedCentralCrossRef Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics. 2020;10(8):3684–707.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Singh K, Nalabotala R, Koo KM, Bose S, Nayak R, Shiddiky MJA. Separation of distinct exosome subpopulations: isolation and characterization approaches and their associated challenges. Analyst. 2021;146(12):3731–49.PubMedCrossRef Singh K, Nalabotala R, Koo KM, Bose S, Nayak R, Shiddiky MJA. Separation of distinct exosome subpopulations: isolation and characterization approaches and their associated challenges. Analyst. 2021;146(12):3731–49.PubMedCrossRef
37.
Zurück zum Zitat Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):152.PubMedPubMedCentralCrossRef Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):152.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Papapanou PN, Sanz M, Buduneli N, Dietrich T, Feres M, Fine DH, et al. Periodontitis: consensus report of workgroup 2 of the 2017 world workshop on the classification of periodontal and peri-implant diseases and conditions. J Periodontol. 2018;89(Suppl 1):S173–82.PubMedCrossRef Papapanou PN, Sanz M, Buduneli N, Dietrich T, Feres M, Fine DH, et al. Periodontitis: consensus report of workgroup 2 of the 2017 world workshop on the classification of periodontal and peri-implant diseases and conditions. J Periodontol. 2018;89(Suppl 1):S173–82.PubMedCrossRef
42.
Zurück zum Zitat Yu J, Lin Y, Xiong X, Li K, Yao Z, Dong H, et al. Detection of exosomal PD-L1 RNA in saliva of patients with periodontitis. Front Genet. 2019;10:202.PubMedPubMedCentralCrossRef Yu J, Lin Y, Xiong X, Li K, Yao Z, Dong H, et al. Detection of exosomal PD-L1 RNA in saliva of patients with periodontitis. Front Genet. 2019;10:202.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Huang X, Hu X, Zhao M, Zhang Q. Analysis of salivary exosomal proteins in young adults with severe periodontitis. Oral Dis. 2020;26(1):173–81.PubMedCrossRef Huang X, Hu X, Zhao M, Zhang Q. Analysis of salivary exosomal proteins in young adults with severe periodontitis. Oral Dis. 2020;26(1):173–81.PubMedCrossRef
44.
Zurück zum Zitat Tobon-Arroyave SI, Celis-Mejia N, Cordoba-Hidalgo MP, Isaza-Guzman DM. Decreased salivary concentration of CD9 and CD81 exosome-related tetraspanins may be associated with the periodontal clinical status. J Clin Periodontol. 2019;46(4):470–80.PubMedCrossRef Tobon-Arroyave SI, Celis-Mejia N, Cordoba-Hidalgo MP, Isaza-Guzman DM. Decreased salivary concentration of CD9 and CD81 exosome-related tetraspanins may be associated with the periodontal clinical status. J Clin Periodontol. 2019;46(4):470–80.PubMedCrossRef
45.
Zurück zum Zitat Nik Mohamed Kamal NNS, Shahidan WNS. Salivary exosomes: from waste to promising periodontitis treatment. Front Physiol. 2021;12:798682.PubMedCrossRef Nik Mohamed Kamal NNS, Shahidan WNS. Salivary exosomes: from waste to promising periodontitis treatment. Front Physiol. 2021;12:798682.PubMedCrossRef
46.
Zurück zum Zitat Zhang Y, Chen J, Fu H, Kuang S, He F, Zhang M, 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.PubMedPubMedCentralCrossRef Zhang Y, Chen J, Fu H, Kuang S, He F, Zhang M, 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.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Shen Z, Kuang S, Zhang Y, Yang M, Qin W, Shi X, 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):1113–26.PubMedPubMedCentralCrossRef Shen Z, Kuang S, Zhang Y, Yang M, Qin W, Shi X, 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):1113–26.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Shimizu Y, Takeda-Kawaguchi T, Kuroda I, Hotta Y, Kawasaki H, Hariyama T, et al. Exosomes from dental pulp cells attenuate bone loss in mouse experimental periodontitis. J Periodontal Res. 2022;57(1):162–72.PubMedCrossRef Shimizu Y, Takeda-Kawaguchi T, Kuroda I, Hotta Y, Kawasaki H, Hariyama T, et al. Exosomes from dental pulp cells attenuate bone loss in mouse experimental periodontitis. J Periodontal Res. 2022;57(1):162–72.PubMedCrossRef
49.
Zurück zum Zitat Lei F, Li M, Lin T, Zhou H, Wang F, Su X. Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes. Acta Biomater. 2022;141:333–43.PubMedCrossRef Lei F, Li M, Lin T, Zhou H, Wang F, Su X. Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes. Acta Biomater. 2022;141:333–43.PubMedCrossRef
50.
Zurück zum Zitat Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, et al. Exosomes from TNF-alpha-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater. 2021;122:306–24.PubMedCrossRef Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, et al. Exosomes from TNF-alpha-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater. 2021;122:306–24.PubMedCrossRef
51.
Zurück zum Zitat Wei J, Song Y, Du Z, Yu F, Zhang Y, Jiang N, et al. Exosomes derived from human exfoliated deciduous teeth ameliorate adult bone loss in mice through promoting osteogenesis. J Mol Histol. 2020;51(4):455–66.PubMedCrossRef Wei J, Song Y, Du Z, Yu F, Zhang Y, Jiang N, et al. Exosomes derived from human exfoliated deciduous teeth ameliorate adult bone loss in mice through promoting osteogenesis. J Mol Histol. 2020;51(4):455–66.PubMedCrossRef
52.
Zurück zum Zitat Wu J, Chen L, Wang R, Song Z, Shen Z, Zhao Y, et al. Exosomes secreted by stem cells from human exfoliated deciduous teeth promote alveolar bone defect repair through the regulation of angiogenesis and osteogenesis. ACS Biomater Sci Eng. 2019;5(7):3561–71.PubMedCrossRef Wu J, Chen L, Wang R, Song Z, Shen Z, Zhao Y, et al. Exosomes secreted by stem cells from human exfoliated deciduous teeth promote alveolar bone defect repair through the regulation of angiogenesis and osteogenesis. ACS Biomater Sci Eng. 2019;5(7):3561–71.PubMedCrossRef
53.
Zurück zum Zitat Chen X, Wan Z, Yang L, Song S, Fu Z, Tang K, et al. Exosomes derived from reparative M2-like macrophages prevent bone loss in murine periodontitis models via IL-10 mRNA. J Nanobiotechnology. 2022;20(1):110.PubMedPubMedCentralCrossRef Chen X, Wan Z, Yang L, Song S, Fu Z, Tang K, et al. Exosomes derived from reparative M2-like macrophages prevent bone loss in murine periodontitis models via IL-10 mRNA. J Nanobiotechnology. 2022;20(1):110.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Byun JS, Lee HY, Tian J, Moon JS, Choi J, Lee SH, et al. Effect of salivary exosomal miR-25-3p on periodontitis with insulin resistance. Front Immunol. 2021;12: 775046.PubMedCrossRef Byun JS, Lee HY, Tian J, Moon JS, Choi J, Lee SH, et al. Effect of salivary exosomal miR-25-3p on periodontitis with insulin resistance. Front Immunol. 2021;12: 775046.PubMedCrossRef
56.
Zurück zum Zitat Taghavi-Farahabadi M, Mahmoudi M, Rezaei N, Hashemi SM. Wharton’s jelly mesenchymal stem cells exosomes and conditioned media increased neutrophil lifespan and phagocytosis capacity. Immunol Invest. 2021;50(8):1042–57.PubMedCrossRef Taghavi-Farahabadi M, Mahmoudi M, Rezaei N, Hashemi SM. Wharton’s jelly mesenchymal stem cells exosomes and conditioned media increased neutrophil lifespan and phagocytosis capacity. Immunol Invest. 2021;50(8):1042–57.PubMedCrossRef
58.
Zurück zum Zitat Mahmoudi M, Taghavi-Farahabadi M, Rezaei N, Hashemi SM. Comparison of the effects of adipose tissue mesenchymal stromal cell-derived exosomes with conditioned media on neutrophil function and apoptosis. Int Immunopharmacol. 2019;74: 105689.PubMedCrossRef Mahmoudi M, Taghavi-Farahabadi M, Rezaei N, Hashemi SM. Comparison of the effects of adipose tissue mesenchymal stromal cell-derived exosomes with conditioned media on neutrophil function and apoptosis. Int Immunopharmacol. 2019;74: 105689.PubMedCrossRef
59.
Zurück zum Zitat Murao A, Tan C, Jha A, Wang P, Aziz M. Exosome-mediated eCIRP release from macrophages to induce inflammation in sepsis. Front Pharmacol. 2021;12: 791648.PubMedPubMedCentralCrossRef Murao A, Tan C, Jha A, Wang P, Aziz M. Exosome-mediated eCIRP release from macrophages to induce inflammation in sepsis. Front Pharmacol. 2021;12: 791648.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Zhao J, Li X, Hu J, Chen F, Qiao S, Sun X, et al. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc Res. 2019;115(7):1205–16.PubMedPubMedCentralCrossRef Zhao J, Li X, Hu J, Chen F, Qiao S, Sun X, et al. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc Res. 2019;115(7):1205–16.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Shi Y, Kang X, Wang Y, Bian X, He G, Zhou M, et al. Exosomes derived from bone marrow stromal cells (BMSCs) enhance tendon-bone healing by regulating macrophage polarization. Med Sci Monit. 2020;26: e923328.PubMedPubMedCentral Shi Y, Kang X, Wang Y, Bian X, He G, Zhou M, et al. Exosomes derived from bone marrow stromal cells (BMSCs) enhance tendon-bone healing by regulating macrophage polarization. Med Sci Monit. 2020;26: e923328.PubMedPubMedCentral
62.
Zurück zum Zitat Deng H, Wu L, Liu M, Zhu L, Chen Y, Zhou H, et al. Bone marrow mesenchymal stem cell-derived exosomes attenuate LPS-induced ARDS by modulating macrophage polarization through inhibiting glycolysis in macrophages. Shock. 2020;54(6):828–43.PubMedCrossRef Deng H, Wu L, Liu M, Zhu L, Chen Y, Zhou H, et al. Bone marrow mesenchymal stem cell-derived exosomes attenuate LPS-induced ARDS by modulating macrophage polarization through inhibiting glycolysis in macrophages. Shock. 2020;54(6):828–43.PubMedCrossRef
63.
Zurück zum Zitat Ning H, Chen H, Deng J, Xiao C, Xu M, Shan L, et al. Exosomes secreted by FNDC5-BMMSCs protect myocardial infarction by anti-inflammation and macrophage polarization via NF-kappaB signaling pathway and Nrf2/HO-1 axis. Stem Cell Res Ther. 2021;12(1):519.PubMedPubMedCentralCrossRef Ning H, Chen H, Deng J, Xiao C, Xu M, Shan L, et al. Exosomes secreted by FNDC5-BMMSCs protect myocardial infarction by anti-inflammation and macrophage polarization via NF-kappaB signaling pathway and Nrf2/HO-1 axis. Stem Cell Res Ther. 2021;12(1):519.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Xin L, Lin X, Zhou F, Li C, Wang X, Yu H, et al. A scaffold laden with mesenchymal stem cell-derived exosomes for promoting endometrium regeneration and fertility restoration through macrophage immunomodulation. Acta Biomater. 2020;113:252–66.PubMedCrossRef Xin L, Lin X, Zhou F, Li C, Wang X, Yu H, et al. A scaffold laden with mesenchymal stem cell-derived exosomes for promoting endometrium regeneration and fertility restoration through macrophage immunomodulation. Acta Biomater. 2020;113:252–66.PubMedCrossRef
65.
Zurück zum Zitat Dong B, Wang C, Zhang J, Zhang J, Gu Y, Guo X, et al. Exosomes from human umbilical cord mesenchymal stem cells attenuate the inflammation of severe steroid-resistant asthma by reshaping macrophage polarization. Stem Cell Res Ther. 2021;12(1):204.PubMedPubMedCentralCrossRef Dong B, Wang C, Zhang J, Zhang J, Gu Y, Guo X, et al. Exosomes from human umbilical cord mesenchymal stem cells attenuate the inflammation of severe steroid-resistant asthma by reshaping macrophage polarization. Stem Cell Res Ther. 2021;12(1):204.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Heo JS, Lim JY, Yoon DW, Pyo S, Kim J. Exosome and melatonin additively attenuates inflammation by transferring miR-34a, miR-124, and miR-135b. Biomed Res Int. 2020;2020:1621394.PubMedPubMedCentralCrossRef Heo JS, Lim JY, Yoon DW, Pyo S, Kim J. Exosome and melatonin additively attenuates inflammation by transferring miR-34a, miR-124, and miR-135b. Biomed Res Int. 2020;2020:1621394.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Zhao H, Shang Q, Pan Z, Bai Y, Li Z, Zhang H, et al. Exosomes from adipose-derived stem cells attenuate adipose inflammation and obesity through polarizing M2 macrophages and Beiging in white adipose tissue. Diabetes. 2018;67(2):235–47.PubMedCrossRef Zhao H, Shang Q, Pan Z, Bai Y, Li Z, Zhang H, et al. Exosomes from adipose-derived stem cells attenuate adipose inflammation and obesity through polarizing M2 macrophages and Beiging in white adipose tissue. Diabetes. 2018;67(2):235–47.PubMedCrossRef
68.
Zurück zum Zitat Shen K, Jia Y, Wang X, Zhang J, Liu K, Wang J, et al. Exosomes from adipose-derived stem cells alleviate the inflammation and oxidative stress via regulating Nrf2/HO-1 axis in macrophages. Free Radic Biol Med. 2021;165:54–66.PubMedCrossRef Shen K, Jia Y, Wang X, Zhang J, Liu K, Wang J, et al. Exosomes from adipose-derived stem cells alleviate the inflammation and oxidative stress via regulating Nrf2/HO-1 axis in macrophages. Free Radic Biol Med. 2021;165:54–66.PubMedCrossRef
69.
Zurück zum Zitat Wang R, Ji Q, Meng C, Liu H, Fan C, Lipkind S, et al. Role of gingival mesenchymal stem cell exosomes in macrophage polarization under inflammatory conditions. Int Immunopharmacol. 2020;81: 106030.PubMedCrossRef Wang R, Ji Q, Meng C, Liu H, Fan C, Lipkind S, et al. Role of gingival mesenchymal stem cell exosomes in macrophage polarization under inflammatory conditions. Int Immunopharmacol. 2020;81: 106030.PubMedCrossRef
70.
Zurück zum Zitat Shahir M, Mahmoud Hashemi S, Asadirad A, Varahram M, Kazempour-Dizaji M, Folkerts G, et al. Effect of mesenchymal stem cell-derived exosomes on the induction of mouse tolerogenic dendritic cells. J Cell Physiol. 2020;235(10):7043–55.PubMedPubMedCentralCrossRef Shahir M, Mahmoud Hashemi S, Asadirad A, Varahram M, Kazempour-Dizaji M, Folkerts G, et al. Effect of mesenchymal stem cell-derived exosomes on the induction of mouse tolerogenic dendritic cells. J Cell Physiol. 2020;235(10):7043–55.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Zhang Y, Yan J, Li Z, Zheng J, Sun Q. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate psoriasis-like skin inflammation. J Interferon Cytokine Res. 2022;42(1):8–18.PubMedCrossRef Zhang Y, Yan J, Li Z, Zheng J, Sun Q. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate psoriasis-like skin inflammation. J Interferon Cytokine Res. 2022;42(1):8–18.PubMedCrossRef
72.
Zurück zum Zitat Elashiry M, Elashiry MM, Elsayed R, Rajendran M, Auersvald C, Zeitoun R, et al. Dendritic cell derived exosomes loaded with immunoregulatory cargo reprogram local immune responses and inhibit degenerative bone disease in vivo. J Extracell Vesicles. 2020;9(1):1795362.PubMedPubMedCentralCrossRef Elashiry M, Elashiry MM, Elsayed R, Rajendran M, Auersvald C, Zeitoun R, et al. Dendritic cell derived exosomes loaded with immunoregulatory cargo reprogram local immune responses and inhibit degenerative bone disease in vivo. J Extracell Vesicles. 2020;9(1):1795362.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Brown M, Johnson LA, Leone DA, Majek P, Vaahtomeri K, Senfter D, et al. Lymphatic exosomes promote dendritic cell migration along guidance cues. J Cell Biol. 2018;217(6):2205–21.PubMedPubMedCentralCrossRef Brown M, Johnson LA, Leone DA, Majek P, Vaahtomeri K, Senfter D, et al. Lymphatic exosomes promote dendritic cell migration along guidance cues. J Cell Biol. 2018;217(6):2205–21.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Cosenza S, Toupet K, Maumus M, Luz-Crawford P, Blanc-Brude O, Jorgensen C, et al. Mesenchymal stem cells-derived exosomes are more immunosuppressive than microparticles in inflammatory arthritis. Theranostics. 2018;8(5):1399–410.PubMedPubMedCentralCrossRef Cosenza S, Toupet K, Maumus M, Luz-Crawford P, Blanc-Brude O, Jorgensen C, et al. Mesenchymal stem cells-derived exosomes are more immunosuppressive than microparticles in inflammatory arthritis. Theranostics. 2018;8(5):1399–410.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Du YM, Zhuansun YX, Chen R, Lin L, Lin Y, Li JG. Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Exp Cell Res. 2018;363(1):114–20.PubMedCrossRef Du YM, Zhuansun YX, Chen R, Lin L, Lin Y, Li JG. Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Exp Cell Res. 2018;363(1):114–20.PubMedCrossRef
76.
Zurück zum Zitat Zhou J, Lu Y, Wu W, Feng Y. HMSC-derived exosome inhibited Th2 cell differentiation via regulating miR-146a-5p/SERPINB2 pathway. J Immunol Res. 2021;2021:6696525.PubMedPubMedCentral Zhou J, Lu Y, Wu W, Feng Y. HMSC-derived exosome inhibited Th2 cell differentiation via regulating miR-146a-5p/SERPINB2 pathway. J Immunol Res. 2021;2021:6696525.PubMedPubMedCentral
77.
Zurück zum Zitat Zheng Y, Dong C, Yang J, Jin Y, Zheng W, Zhou Q, et al. Exosomal microRNA-155-5p from PDLSCs regulated Th17/Treg balance by targeting sirtuin-1 in chronic periodontitis. J Cell Physiol. 2019;234(11):20662–74.PubMedCrossRef Zheng Y, Dong C, Yang J, Jin Y, Zheng W, Zhou Q, et al. Exosomal microRNA-155-5p from PDLSCs regulated Th17/Treg balance by targeting sirtuin-1 in chronic periodontitis. J Cell Physiol. 2019;234(11):20662–74.PubMedCrossRef
78.
Zurück zum Zitat Xu L, Geng T, Zang G, Bo L, Liang Y, Zhou H, et al. Exosome derived from CD137-modified endothelial cells regulates the Th17 responses in atherosclerosis. J Cell Mol Med. 2020;24(8):4659–67.PubMedPubMedCentralCrossRef Xu L, Geng T, Zang G, Bo L, Liang Y, Zhou H, et al. Exosome derived from CD137-modified endothelial cells regulates the Th17 responses in atherosclerosis. J Cell Mol Med. 2020;24(8):4659–67.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Xiao F, Zuo B, Tao B, Wang C, Li Y, Peng J, et al. Exosomes derived from cyclic mechanical stretch-exposed bone marrow mesenchymal stem cells inhibit RANKL-induced osteoclastogenesis through the NF-kappaB signaling pathway. Ann Transl Med. 2021;9(9):798.PubMedPubMedCentralCrossRef Xiao F, Zuo B, Tao B, Wang C, Li Y, Peng J, et al. Exosomes derived from cyclic mechanical stretch-exposed bone marrow mesenchymal stem cells inhibit RANKL-induced osteoclastogenesis through the NF-kappaB signaling pathway. Ann Transl Med. 2021;9(9):798.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Zhang L, Wang Q, Su H, Cheng J. Exosomes from adipose derived mesenchymal stem cells alleviate diabetic osteoporosis in rats through suppressing NLRP3 inflammasome activation in osteoclasts. J Biosci Bioeng. 2021;131(6):671–8.PubMedCrossRef Zhang L, Wang Q, Su H, Cheng J. Exosomes from adipose derived mesenchymal stem cells alleviate diabetic osteoporosis in rats through suppressing NLRP3 inflammasome activation in osteoclasts. J Biosci Bioeng. 2021;131(6):671–8.PubMedCrossRef
81.
Zurück zum Zitat Ren L, Song ZJ, Cai QW, Chen RX, Zou Y, Fu Q, et al. Adipose mesenchymal stem cell-derived exosomes ameliorate hypoxia/serum deprivation-induced osteocyte apoptosis and osteocyte-mediated osteoclastogenesis in vitro. Biochem Biophys Res Commun. 2019;508(1):138–44.PubMedCrossRef Ren L, Song ZJ, Cai QW, Chen RX, Zou Y, Fu Q, et al. Adipose mesenchymal stem cell-derived exosomes ameliorate hypoxia/serum deprivation-induced osteocyte apoptosis and osteocyte-mediated osteoclastogenesis in vitro. Biochem Biophys Res Commun. 2019;508(1):138–44.PubMedCrossRef
82.
Zurück zum Zitat Zhang L, Wang Q, Su H, Cheng J. Exosomes from adipose tissues derived mesenchymal stem cells overexpressing MicroRNA-146a alleviate diabetic osteoporosis in rats. Cell Mol Bioeng. 2022;15(1):87–97.PubMedCrossRef Zhang L, Wang Q, Su H, Cheng J. Exosomes from adipose tissues derived mesenchymal stem cells overexpressing MicroRNA-146a alleviate diabetic osteoporosis in rats. Cell Mol Bioeng. 2022;15(1):87–97.PubMedCrossRef
83.
Zurück zum Zitat Wang Q, Shen X, Chen Y, Chen J, Li Y. Osteoblasts-derived exosomes regulate osteoclast differentiation through miR-503-3p/Hpse axis. Acta Histochem. 2021;123(7): 151790.PubMedCrossRef Wang Q, Shen X, Chen Y, Chen J, Li Y. Osteoblasts-derived exosomes regulate osteoclast differentiation through miR-503-3p/Hpse axis. Acta Histochem. 2021;123(7): 151790.PubMedCrossRef
84.
Zurück zum Zitat Cui Y, Fu S, Sun D, Xing J, Hou T, Wu X. EPC-derived exosomes promote osteoclastogenesis through LncRNA-MALAT1. J Cell Mol Med. 2019;23(6):3843–54.PubMedPubMedCentralCrossRef Cui Y, Fu S, Sun D, Xing J, Hou T, Wu X. EPC-derived exosomes promote osteoclastogenesis through LncRNA-MALAT1. J Cell Mol Med. 2019;23(6):3843–54.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78.PubMedCrossRef Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78.PubMedCrossRef
Metadaten
Titel
Advances of exosomes in periodontitis treatment
verfasst von
Hongbing Lin
Huishan Chen
Xuetao Zhao
Tong Ding
Yawei Wang
Zhen Chen
Yue Tian
Peipei Zhang
Yuqin Shen
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2022
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03487-4

Weitere Artikel der Ausgabe 1/2022

Journal of Translational Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Dihydropyridin-Kalziumantagonisten können auf die Nieren gehen

30.04.2024 Hypertonie Nachrichten

Im Vergleich zu anderen Blutdrucksenkern sind Kalziumantagonisten vom Diyhdropyridin-Typ mit einem erhöhten Risiko für eine Mikroalbuminurie und in Abwesenheit eines RAS-Blockers auch für ein terminales Nierenversagen verbunden.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.