Cargando…

Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke

BACKGROUND: Ischemic stroke is an acute and severe neurological disease, and reperfusion is an effective way to reverse brain damage after stroke. However, reperfusion causes secondary tissue damage induced by inflammatory responses, called ischemia/reperfusion (I/R) injury. Current therapeutic stra...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yanyun, Wang, Yi, Li, Shuyu, Cui, Yuliang, Liang, Xiping, Shan, Juanjuan, Gu, Wei, Qiu, Juhui, Li, Yiliang, Wang, Guixue
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8529766/
https://www.ncbi.nlm.nih.gov/pubmed/34674712
http://dx.doi.org/10.1186/s12951-021-01067-0
_version_ 1784586535587282944
author Wang, Yanyun
Wang, Yi
Li, Shuyu
Cui, Yuliang
Liang, Xiping
Shan, Juanjuan
Gu, Wei
Qiu, Juhui
Li, Yiliang
Wang, Guixue
author_facet Wang, Yanyun
Wang, Yi
Li, Shuyu
Cui, Yuliang
Liang, Xiping
Shan, Juanjuan
Gu, Wei
Qiu, Juhui
Li, Yiliang
Wang, Guixue
author_sort Wang, Yanyun
collection PubMed
description BACKGROUND: Ischemic stroke is an acute and severe neurological disease, and reperfusion is an effective way to reverse brain damage after stroke. However, reperfusion causes secondary tissue damage induced by inflammatory responses, called ischemia/reperfusion (I/R) injury. Current therapeutic strategies that control inflammation to treat I/R are less than satisfactory. RESULTS: We report a kind of shield and sword nano-soldier functionalized nanoparticles (monocyte membranes-coated rapamycin nanoparticles, McM/RNPs) that can reduce inflammation and relieve I/R injury by blocking monocyte infiltration and inhibiting microglia proliferation. The fabricated McM/RNPs can actively target and bind to inflammatory endothelial cells, which inhibit the adhesion of monocytes to the endothelium, thus acting as a shield. Subsequently, McM/RNPs can penetrate the endothelium to reach the injury site, similar to a sword, and release the RAP drug to inhibit the proliferation of inflammatory cells. In a rat I/R injury model, McM/RNPs exhibited improved active homing to I/R injury areas and greatly ameliorated neuroscores and infarct volume. Importantly, in vivo animal studies revealed good safety for McM/RNPs treatment. CONCLUSION: The results demonstrated that the developed McM/RNPs may serve as an effective and safe nanovehicles for I/R injury therapy. GRAPHIC ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-021-01067-0.
format Online
Article
Text
id pubmed-8529766
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-85297662021-10-25 Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke Wang, Yanyun Wang, Yi Li, Shuyu Cui, Yuliang Liang, Xiping Shan, Juanjuan Gu, Wei Qiu, Juhui Li, Yiliang Wang, Guixue J Nanobiotechnology Research BACKGROUND: Ischemic stroke is an acute and severe neurological disease, and reperfusion is an effective way to reverse brain damage after stroke. However, reperfusion causes secondary tissue damage induced by inflammatory responses, called ischemia/reperfusion (I/R) injury. Current therapeutic strategies that control inflammation to treat I/R are less than satisfactory. RESULTS: We report a kind of shield and sword nano-soldier functionalized nanoparticles (monocyte membranes-coated rapamycin nanoparticles, McM/RNPs) that can reduce inflammation and relieve I/R injury by blocking monocyte infiltration and inhibiting microglia proliferation. The fabricated McM/RNPs can actively target and bind to inflammatory endothelial cells, which inhibit the adhesion of monocytes to the endothelium, thus acting as a shield. Subsequently, McM/RNPs can penetrate the endothelium to reach the injury site, similar to a sword, and release the RAP drug to inhibit the proliferation of inflammatory cells. In a rat I/R injury model, McM/RNPs exhibited improved active homing to I/R injury areas and greatly ameliorated neuroscores and infarct volume. Importantly, in vivo animal studies revealed good safety for McM/RNPs treatment. CONCLUSION: The results demonstrated that the developed McM/RNPs may serve as an effective and safe nanovehicles for I/R injury therapy. GRAPHIC ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-021-01067-0. BioMed Central 2021-10-21 /pmc/articles/PMC8529766/ /pubmed/34674712 http://dx.doi.org/10.1186/s12951-021-01067-0 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/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/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://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.
spellingShingle Research
Wang, Yanyun
Wang, Yi
Li, Shuyu
Cui, Yuliang
Liang, Xiping
Shan, Juanjuan
Gu, Wei
Qiu, Juhui
Li, Yiliang
Wang, Guixue
Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke
title Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke
title_full Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke
title_fullStr Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke
title_full_unstemmed Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke
title_short Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke
title_sort functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8529766/
https://www.ncbi.nlm.nih.gov/pubmed/34674712
http://dx.doi.org/10.1186/s12951-021-01067-0
work_keys_str_mv AT wangyanyun functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT wangyi functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT lishuyu functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT cuiyuliang functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT liangxiping functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT shanjuanjuan functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT guwei functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT qiujuhui functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT liyiliang functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke
AT wangguixue functionalizednanoparticleswithmonocytemembranesandrapamycinachievesynergisticchemoimmunotherapyforreperfusioninducedinjuryinischemicstroke