Published in

Wiley, Journal of Periodontal Research, 2024

DOI: 10.1111/jre.13283

Links

Tools

Export citation

Search in Google Scholar

Microbial‐ and host immune cell‐derived extracellular vesicles in the pathogenesis and therapy of periodontitis: A narrative review

Journal article published in 2024 by Jenny Wang, Chun Liu, Jason Cutler, Sašo Ivanovski, Ryan Sb Lee ORCID, Pingping Han ORCID
This paper was not found in any repository, but could be made available legally by the author.
This paper was not found in any repository, but could be made available legally by the author.

Full text: Unavailable

Green circle
Preprint: archiving allowed
Orange circle
Postprint: archiving restricted
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

AbstractPeriodontitis is a chronic inflammatory disease caused by dysbiotic biofilms and destructive host immune responses. Extracellular vesicles (EVs) are circulating nanoparticles released by microbes and host cells involved in cell‐to‐cell communication, found in body biofluids, such as saliva and gingival crevicular fluid (GCF). EVs are mainly involved in cell‐to‐cell communication, and may hold promise for diagnostic and therapeutic purposes. Periodontal research has examined the potential involvement of bacterial‐ and host‐cell‐derived EVs in disease pathogenesis, diagnosis, and therapy, but data remains scarce on immune cell‐ or microbial‐derived EVs. In this narrative review, we first provide an overview of the role of microbial and host‐derived EVs on disease pathogenesis. Recent studies reveal that Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans‐derived outer membrane vesicles (OMVs) can activate inflammatory cytokine release in host cells, while M1 macrophage EVs may contribute to bone loss. Additionally, we summarised current in vitro and pre‐clinical research on the utilisation of immune cell and microbial‐derived EVs as potential therapeutic tools in the context of periodontal treatment. Studies indicate that EVs from M2 macrophages and dendritic cells promote bone regeneration in animal models. While bacterial EVs remain underexplored for periodontal therapy, preliminary research suggests that P. gingivalis OMVs hold promise as vaccine candidates. Finally, we acknowledge the current limitations present in the field of translating immune cell derived EVs and microbial derived EVs in periodontology. It is concluded that microbial and host immune cell‐derived EVs have a role in periodontitis pathogenesis and hence may be useful for studying disease pathophysiology, and as diagnostic and treatment monitoring biomarkers.