Cargando…

Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs

Exosomes exhibit great potential as novel therapeutics for tissue regeneration, including cell migration and angiogenesis. However, the limited intracellular delivery efficiency of exosomes might reduce their biological effects. Here, exosomes secreted by adipose-derived mesenchymal stem cells were...

Descripción completa

Detalles Bibliográficos
Autores principales: Ma, Shengnan, Song, Lei, Bai, Yueyue, Wang, Shihao, Wang, Jiao, Zhang, Haohao, Wang, Fazhan, He, Yiyan, Tian, Chuntao, Qin, Guijun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: The Royal Society of Chemistry 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10087381/
https://www.ncbi.nlm.nih.gov/pubmed/37057265
http://dx.doi.org/10.1039/d3ra00300k
_version_ 1785022333525688320
author Ma, Shengnan
Song, Lei
Bai, Yueyue
Wang, Shihao
Wang, Jiao
Zhang, Haohao
Wang, Fazhan
He, Yiyan
Tian, Chuntao
Qin, Guijun
author_facet Ma, Shengnan
Song, Lei
Bai, Yueyue
Wang, Shihao
Wang, Jiao
Zhang, Haohao
Wang, Fazhan
He, Yiyan
Tian, Chuntao
Qin, Guijun
author_sort Ma, Shengnan
collection PubMed
description Exosomes exhibit great potential as novel therapeutics for tissue regeneration, including cell migration and angiogenesis. However, the limited intracellular delivery efficiency of exosomes might reduce their biological effects. Here, exosomes secreted by adipose-derived mesenchymal stem cells were recombined with fluorinated peptide dendrimers (FPG3) to form the fluorine-engineered exosomes (exo@FPG3), which was intended to promote the cytosolic release and the biological function of exosomes. The mass ratio of FPG3 to exosomes at 5 was used to investigate its cellular uptake efficiency and bioactivity in HUVECs, as the charge of exo@FPG3 tended to be stable even more FPG3 was applied. It was found that exo@FPG3 could enter HUVECs through a variety of pathways, in which the clathrin-mediated endocytosis played an important role. Compared with exosomes modified with peptide dendrimers (exo@PG3) and exosomes alone, the cellular uptake efficiency of exo@FPG3 was significantly increased. Moreover, exo@FPG3 significantly enhanced the angiogenesis and migration of HUVECs in vitro as compared to exo@PG3 and exosomes. It is concluded that surface fluorine modification of exosomes with FPG3 is conducive to the cellular uptake and bioactivity of the exosome, which provides a novel strategy for engineered exosomes to enhance the biological effects of exosome-based drug delivery.
format Online
Article
Text
id pubmed-10087381
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher The Royal Society of Chemistry
record_format MEDLINE/PubMed
spelling pubmed-100873812023-04-12 Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs Ma, Shengnan Song, Lei Bai, Yueyue Wang, Shihao Wang, Jiao Zhang, Haohao Wang, Fazhan He, Yiyan Tian, Chuntao Qin, Guijun RSC Adv Chemistry Exosomes exhibit great potential as novel therapeutics for tissue regeneration, including cell migration and angiogenesis. However, the limited intracellular delivery efficiency of exosomes might reduce their biological effects. Here, exosomes secreted by adipose-derived mesenchymal stem cells were recombined with fluorinated peptide dendrimers (FPG3) to form the fluorine-engineered exosomes (exo@FPG3), which was intended to promote the cytosolic release and the biological function of exosomes. The mass ratio of FPG3 to exosomes at 5 was used to investigate its cellular uptake efficiency and bioactivity in HUVECs, as the charge of exo@FPG3 tended to be stable even more FPG3 was applied. It was found that exo@FPG3 could enter HUVECs through a variety of pathways, in which the clathrin-mediated endocytosis played an important role. Compared with exosomes modified with peptide dendrimers (exo@PG3) and exosomes alone, the cellular uptake efficiency of exo@FPG3 was significantly increased. Moreover, exo@FPG3 significantly enhanced the angiogenesis and migration of HUVECs in vitro as compared to exo@PG3 and exosomes. It is concluded that surface fluorine modification of exosomes with FPG3 is conducive to the cellular uptake and bioactivity of the exosome, which provides a novel strategy for engineered exosomes to enhance the biological effects of exosome-based drug delivery. The Royal Society of Chemistry 2023-04-11 /pmc/articles/PMC10087381/ /pubmed/37057265 http://dx.doi.org/10.1039/d3ra00300k Text en This journal is © The Royal Society of Chemistry https://creativecommons.org/licenses/by-nc/3.0/
spellingShingle Chemistry
Ma, Shengnan
Song, Lei
Bai, Yueyue
Wang, Shihao
Wang, Jiao
Zhang, Haohao
Wang, Fazhan
He, Yiyan
Tian, Chuntao
Qin, Guijun
Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs
title Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs
title_full Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs
title_fullStr Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs
title_full_unstemmed Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs
title_short Improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of HUVECs
title_sort improved intracellular delivery of exosomes by surface modification with fluorinated peptide dendrimers for promoting angiogenesis and migration of huvecs
topic Chemistry
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10087381/
https://www.ncbi.nlm.nih.gov/pubmed/37057265
http://dx.doi.org/10.1039/d3ra00300k
work_keys_str_mv AT mashengnan improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT songlei improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT baiyueyue improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT wangshihao improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT wangjiao improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT zhanghaohao improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT wangfazhan improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT heyiyan improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT tianchuntao improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs
AT qinguijun improvedintracellulardeliveryofexosomesbysurfacemodificationwithfluorinatedpeptidedendrimersforpromotingangiogenesisandmigrationofhuvecs