Cargando…

Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy

INTRODUCTION: The bone regeneration of endosseous implanted biomaterials is often impaired by the host immune response, especially macrophage-related inflammation which plays an important role in the bone healing process. Thus, it is a promising strategy to design an osteo-immunomodulatory biomateri...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Yangmengfan, Guan, Ming, Ren, Ranyue, Gao, Chenghao, Cheng, Hao, Li, Yong, Gao, Biao, Wei, Yong, Fu, Jijiang, Sun, Jun, Xiong, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7102919/
https://www.ncbi.nlm.nih.gov/pubmed/32273699
http://dx.doi.org/10.2147/IJN.S242919
_version_ 1783511940410114048
author Chen, Yangmengfan
Guan, Ming
Ren, Ranyue
Gao, Chenghao
Cheng, Hao
Li, Yong
Gao, Biao
Wei, Yong
Fu, Jijiang
Sun, Jun
Xiong, Wei
author_facet Chen, Yangmengfan
Guan, Ming
Ren, Ranyue
Gao, Chenghao
Cheng, Hao
Li, Yong
Gao, Biao
Wei, Yong
Fu, Jijiang
Sun, Jun
Xiong, Wei
author_sort Chen, Yangmengfan
collection PubMed
description INTRODUCTION: The bone regeneration of endosseous implanted biomaterials is often impaired by the host immune response, especially macrophage-related inflammation which plays an important role in the bone healing process. Thus, it is a promising strategy to design an osteo-immunomodulatory biomaterial to take advantage of the macrophage-related immune response and improve the osseointegration performance of the implant. METHODS: In this study, we developed an antibacterial silver nanoparticle-loaded TiO(2) nanotubes (Ag@TiO(2)-NTs) using an electrochemical anodization method to make the surface modification and investigated the influences of Ag@TiO(2)-NTs on the macrophage polarization, osteo-immune microenvironment as well as its potential molecular mechanisms in vitro and in vivo. RESULTS: The results showed that Ag@TiO(2)-NTs with controlled releasing of ultra-low-dose Ag(+) ions had the excellent ability to induce the macrophage polarization towards the M2 phenotype and create a suitable osteo-immune microenvironment in vitro, via inhibiting PI3K/Akt, suppressing the downstream effector GLUT1, and activating autophagy. Moreover, Ag@TiO(2)-NTs surface could improve bone formation, suppress inflammation, and promote osteo-immune microenvironment compared to the TiO(2)-NTs and polished Ti surfaces in vivo. These findings suggested that Ag@TiO(2)-NTs with controlled releasing of ultra-low-dose Ag(+) ions could not only inhibit the inflammation process but also promote the bone healing by inducing healing-associated M2 polarization. DISCUSSION: Using this surface modification strategy to modulate the macrophage-related immune response, rather than prevent the host response, maybe a promising strategy for implant surgeries in the future.
format Online
Article
Text
id pubmed-7102919
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-71029192020-04-09 Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy Chen, Yangmengfan Guan, Ming Ren, Ranyue Gao, Chenghao Cheng, Hao Li, Yong Gao, Biao Wei, Yong Fu, Jijiang Sun, Jun Xiong, Wei Int J Nanomedicine Original Research INTRODUCTION: The bone regeneration of endosseous implanted biomaterials is often impaired by the host immune response, especially macrophage-related inflammation which plays an important role in the bone healing process. Thus, it is a promising strategy to design an osteo-immunomodulatory biomaterial to take advantage of the macrophage-related immune response and improve the osseointegration performance of the implant. METHODS: In this study, we developed an antibacterial silver nanoparticle-loaded TiO(2) nanotubes (Ag@TiO(2)-NTs) using an electrochemical anodization method to make the surface modification and investigated the influences of Ag@TiO(2)-NTs on the macrophage polarization, osteo-immune microenvironment as well as its potential molecular mechanisms in vitro and in vivo. RESULTS: The results showed that Ag@TiO(2)-NTs with controlled releasing of ultra-low-dose Ag(+) ions had the excellent ability to induce the macrophage polarization towards the M2 phenotype and create a suitable osteo-immune microenvironment in vitro, via inhibiting PI3K/Akt, suppressing the downstream effector GLUT1, and activating autophagy. Moreover, Ag@TiO(2)-NTs surface could improve bone formation, suppress inflammation, and promote osteo-immune microenvironment compared to the TiO(2)-NTs and polished Ti surfaces in vivo. These findings suggested that Ag@TiO(2)-NTs with controlled releasing of ultra-low-dose Ag(+) ions could not only inhibit the inflammation process but also promote the bone healing by inducing healing-associated M2 polarization. DISCUSSION: Using this surface modification strategy to modulate the macrophage-related immune response, rather than prevent the host response, maybe a promising strategy for implant surgeries in the future. Dove 2020-03-24 /pmc/articles/PMC7102919/ /pubmed/32273699 http://dx.doi.org/10.2147/IJN.S242919 Text en © 2020 Chen et al. http://creativecommons.org/licenses/by-nc/3.0/ 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, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). 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 (https://www.dovepress.com/terms.php).
spellingShingle Original Research
Chen, Yangmengfan
Guan, Ming
Ren, Ranyue
Gao, Chenghao
Cheng, Hao
Li, Yong
Gao, Biao
Wei, Yong
Fu, Jijiang
Sun, Jun
Xiong, Wei
Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy
title Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy
title_full Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy
title_fullStr Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy
title_full_unstemmed Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy
title_short Improved Immunoregulation of Ultra-Low-Dose Silver Nanoparticle-Loaded TiO(2) Nanotubes via M2 Macrophage Polarization by Regulating GLUT1 and Autophagy
title_sort improved immunoregulation of ultra-low-dose silver nanoparticle-loaded tio(2) nanotubes via m2 macrophage polarization by regulating glut1 and autophagy
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7102919/
https://www.ncbi.nlm.nih.gov/pubmed/32273699
http://dx.doi.org/10.2147/IJN.S242919
work_keys_str_mv AT chenyangmengfan improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT guanming improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT renranyue improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT gaochenghao improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT chenghao improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT liyong improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT gaobiao improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT weiyong improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT fujijiang improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT sunjun improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy
AT xiongwei improvedimmunoregulationofultralowdosesilvernanoparticleloadedtio2nanotubesviam2macrophagepolarizationbyregulatingglut1andautophagy