Cargando…

Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury

Rationale: As a potentially life-threatening disorder, cerebral ischemia-reperfusion (I/R) injury is associated with significantly high mortality, especially the irreversible brain tissue damage associated with increased reactive oxygen radical production and excessive inflammation. Currently, the i...

Descripción completa

Detalles Bibliográficos
Autores principales: Luo, Li, Zang, Guangchao, Liu, Boyan, Qin, Xian, Zhang, Yuchan, Chen, Yidan, Zhang, Haijun, Wu, Wei, Wang, Guixue
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8315061/
https://www.ncbi.nlm.nih.gov/pubmed/34335979
http://dx.doi.org/10.7150/thno.60785
_version_ 1783729661889478656
author Luo, Li
Zang, Guangchao
Liu, Boyan
Qin, Xian
Zhang, Yuchan
Chen, Yidan
Zhang, Haijun
Wu, Wei
Wang, Guixue
author_facet Luo, Li
Zang, Guangchao
Liu, Boyan
Qin, Xian
Zhang, Yuchan
Chen, Yidan
Zhang, Haijun
Wu, Wei
Wang, Guixue
author_sort Luo, Li
collection PubMed
description Rationale: As a potentially life-threatening disorder, cerebral ischemia-reperfusion (I/R) injury is associated with significantly high mortality, especially the irreversible brain tissue damage associated with increased reactive oxygen radical production and excessive inflammation. Currently, the insufficiency of targeted drug delivery and “on-demand” drug release remain the greatest challenges for cerebral I/R injury therapy. Bioengineered cell membrane-based nanotherapeutics mimic and enhance natural membrane functions and represent a potentially promising approach, relying on selective interactions between receptors and chemokines and increase nanomedicine delivery efficiency into the target tissues. Methods: We employed a systematic method to synthesize biomimetic smart nanoparticles. The CXCR4-overexpressing primary mouse thoracic aorta endothelial cell (PMTAEC) membranes and RAPA@HOP were extruded through a 200 nm polycarbonate porous membrane using a mini-extruder to harvest the RAPA@BMHOP. The bioengineered CXCR4-overexpressing cell membrane-functionalized ROS-responsive nanotherapeutics, loaded with rapamycin (RAPA), were fabricated to enhance the targeted delivery to lesions with pathological overexpression of SDF-1. Results: RAPA@BMHOP exhibited a three-fold higher rate of target delivery efficacy via the CXCR4/SDF-1 axis than its non-targeting counterpart in an in vivo model. Additionally, in response to the excessive pathological ROS, nanotherapeutics could be degraded to promote “on-demand” cargo release and balance the ROS level by p-hydroxy-benzyl alcohol degradation, thereby scavenging excessive ROS and suppressing the free radical-induced focal damage and local inflammation. Also, the stealth effect of cell membrane coating functionalization on the surface resulted in extended circulation time and high stability of nanoparticles. Conclusion: The biomimetic smart nanotherapeutics with active targeting, developed in this study, significantly improved the therapeutic efficacy and biosafety profiles. Thus, these nanoparticles could be a candidate for efficient therapy of cerebral I/R injury.
format Online
Article
Text
id pubmed-8315061
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-83150612021-07-30 Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury Luo, Li Zang, Guangchao Liu, Boyan Qin, Xian Zhang, Yuchan Chen, Yidan Zhang, Haijun Wu, Wei Wang, Guixue Theranostics Research Paper Rationale: As a potentially life-threatening disorder, cerebral ischemia-reperfusion (I/R) injury is associated with significantly high mortality, especially the irreversible brain tissue damage associated with increased reactive oxygen radical production and excessive inflammation. Currently, the insufficiency of targeted drug delivery and “on-demand” drug release remain the greatest challenges for cerebral I/R injury therapy. Bioengineered cell membrane-based nanotherapeutics mimic and enhance natural membrane functions and represent a potentially promising approach, relying on selective interactions between receptors and chemokines and increase nanomedicine delivery efficiency into the target tissues. Methods: We employed a systematic method to synthesize biomimetic smart nanoparticles. The CXCR4-overexpressing primary mouse thoracic aorta endothelial cell (PMTAEC) membranes and RAPA@HOP were extruded through a 200 nm polycarbonate porous membrane using a mini-extruder to harvest the RAPA@BMHOP. The bioengineered CXCR4-overexpressing cell membrane-functionalized ROS-responsive nanotherapeutics, loaded with rapamycin (RAPA), were fabricated to enhance the targeted delivery to lesions with pathological overexpression of SDF-1. Results: RAPA@BMHOP exhibited a three-fold higher rate of target delivery efficacy via the CXCR4/SDF-1 axis than its non-targeting counterpart in an in vivo model. Additionally, in response to the excessive pathological ROS, nanotherapeutics could be degraded to promote “on-demand” cargo release and balance the ROS level by p-hydroxy-benzyl alcohol degradation, thereby scavenging excessive ROS and suppressing the free radical-induced focal damage and local inflammation. Also, the stealth effect of cell membrane coating functionalization on the surface resulted in extended circulation time and high stability of nanoparticles. Conclusion: The biomimetic smart nanotherapeutics with active targeting, developed in this study, significantly improved the therapeutic efficacy and biosafety profiles. Thus, these nanoparticles could be a candidate for efficient therapy of cerebral I/R injury. Ivyspring International Publisher 2021-07-06 /pmc/articles/PMC8315061/ /pubmed/34335979 http://dx.doi.org/10.7150/thno.60785 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Luo, Li
Zang, Guangchao
Liu, Boyan
Qin, Xian
Zhang, Yuchan
Chen, Yidan
Zhang, Haijun
Wu, Wei
Wang, Guixue
Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury
title Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury
title_full Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury
title_fullStr Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury
title_full_unstemmed Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury
title_short Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury
title_sort bioengineering cxcr4-overexpressing cell membrane functionalized ros-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8315061/
https://www.ncbi.nlm.nih.gov/pubmed/34335979
http://dx.doi.org/10.7150/thno.60785
work_keys_str_mv AT luoli bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT zangguangchao bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT liuboyan bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT qinxian bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT zhangyuchan bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT chenyidan bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT zhanghaijun bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT wuwei bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury
AT wangguixue bioengineeringcxcr4overexpressingcellmembranefunctionalizedrosresponsivenanotherapeuticsfortargetingcerebralischemiareperfusioninjury