Cargando…

Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity

Background: Extracellular vesicles, including exosomes, are secreted by a variety of cell types in the central nervous system. Exosomes play a role in removing intracellular materials from the endosomal system. Alzheimer's disease (AD) is caused by an overproduction or reduced amyloid-beta (Aβ)...

Descripción completa

Detalles Bibliográficos
Autores principales: Deng, Zhiting, Wang, Jieqiong, Xiao, Yang, Li, Fei, Niu, Lili, Liu, Xin, Meng, Long, Zheng, Hairong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7977450/
https://www.ncbi.nlm.nih.gov/pubmed/33754065
http://dx.doi.org/10.7150/thno.52436
_version_ 1783667115675353088
author Deng, Zhiting
Wang, Jieqiong
Xiao, Yang
Li, Fei
Niu, Lili
Liu, Xin
Meng, Long
Zheng, Hairong
author_facet Deng, Zhiting
Wang, Jieqiong
Xiao, Yang
Li, Fei
Niu, Lili
Liu, Xin
Meng, Long
Zheng, Hairong
author_sort Deng, Zhiting
collection PubMed
description Background: Extracellular vesicles, including exosomes, are secreted by a variety of cell types in the central nervous system. Exosomes play a role in removing intracellular materials from the endosomal system. Alzheimer's disease (AD) is caused by an overproduction or reduced amyloid-beta (Aβ) peptide clearance. Increased Aβ levels in the brain may impair the exosome-mediated Aβ clearance pathway. Therapeutic ultrasound stimulation demonstrated its potential for promoting Aβ degradation efficiency in clinical trials. However, the underlying mechanism of ultrasound stimulation is still unclear. Methods: In this study, astrocytes, the most abundant glial cells in the brain, were used for exosome production. Post insonation, exosomes from ultrasound-stimulated HA cells (US-HA-Exo) were collected, nanoparticle tracking analysis and protein analysis were used to measure and characterize exosomes. Neuroprotective effect of US-HA-Exo in oligomeric Aβ(42) toxicated SH-SY5Y cells was tested. Cellular uptake and distribution of exosomes were observed by flow cytometry and confocal laser scanning microscopy. Focused ultrasound (FUS) with microbubbles was employed for blood-brain-barrier opening to achieve brain-targeted exosome delivery. After US-HA-Exo/FUS treatment, amyloid-β plaque in APP/PS1 mice were evaluated by Aβ immunostaining and thioflavin-S staining. Results: We showed that ultrasound resulted in an almost 5-fold increase in the exosome release from human astrocytes. Exosomes were rapidly internalized in SH-SY5Y cells, and colocalized with FITC-Aβ(42), causing a decreased uptake of FITC-Aβ(42). CCk-8 test results showed that US-HA-Exo could mitigate Aβ toxicity to neurons in vitro. The therapeutic potential of US-HA-Exo/FUS delivery was demonstrated by a decrease in thioflavin-S-positive amyloid plaques and Aβ immuno-staining, a therapeutic target for AD in APP/PS1 transgenic mice. The iTRAQ-based proteomic quantification was performed to gain mechanistic insight into the ultrasound effect on astrocyte-derived exosomes and their ability to alleviate Aβ neurotoxicity. Conclusion: Our results imply that US-HA-Exo have the potential to provide neuroprotective effects to reverse oligomeric amyloid-β-induced cytotoxicity in vitro and, when combined with FUS-induced BBB opening, enable the clearance of amyloid-β plaques in vivo.
format Online
Article
Text
id pubmed-7977450
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-79774502021-03-21 Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity Deng, Zhiting Wang, Jieqiong Xiao, Yang Li, Fei Niu, Lili Liu, Xin Meng, Long Zheng, Hairong Theranostics Research Paper Background: Extracellular vesicles, including exosomes, are secreted by a variety of cell types in the central nervous system. Exosomes play a role in removing intracellular materials from the endosomal system. Alzheimer's disease (AD) is caused by an overproduction or reduced amyloid-beta (Aβ) peptide clearance. Increased Aβ levels in the brain may impair the exosome-mediated Aβ clearance pathway. Therapeutic ultrasound stimulation demonstrated its potential for promoting Aβ degradation efficiency in clinical trials. However, the underlying mechanism of ultrasound stimulation is still unclear. Methods: In this study, astrocytes, the most abundant glial cells in the brain, were used for exosome production. Post insonation, exosomes from ultrasound-stimulated HA cells (US-HA-Exo) were collected, nanoparticle tracking analysis and protein analysis were used to measure and characterize exosomes. Neuroprotective effect of US-HA-Exo in oligomeric Aβ(42) toxicated SH-SY5Y cells was tested. Cellular uptake and distribution of exosomes were observed by flow cytometry and confocal laser scanning microscopy. Focused ultrasound (FUS) with microbubbles was employed for blood-brain-barrier opening to achieve brain-targeted exosome delivery. After US-HA-Exo/FUS treatment, amyloid-β plaque in APP/PS1 mice were evaluated by Aβ immunostaining and thioflavin-S staining. Results: We showed that ultrasound resulted in an almost 5-fold increase in the exosome release from human astrocytes. Exosomes were rapidly internalized in SH-SY5Y cells, and colocalized with FITC-Aβ(42), causing a decreased uptake of FITC-Aβ(42). CCk-8 test results showed that US-HA-Exo could mitigate Aβ toxicity to neurons in vitro. The therapeutic potential of US-HA-Exo/FUS delivery was demonstrated by a decrease in thioflavin-S-positive amyloid plaques and Aβ immuno-staining, a therapeutic target for AD in APP/PS1 transgenic mice. The iTRAQ-based proteomic quantification was performed to gain mechanistic insight into the ultrasound effect on astrocyte-derived exosomes and their ability to alleviate Aβ neurotoxicity. Conclusion: Our results imply that US-HA-Exo have the potential to provide neuroprotective effects to reverse oligomeric amyloid-β-induced cytotoxicity in vitro and, when combined with FUS-induced BBB opening, enable the clearance of amyloid-β plaques in vivo. Ivyspring International Publisher 2021-02-25 /pmc/articles/PMC7977450/ /pubmed/33754065 http://dx.doi.org/10.7150/thno.52436 Text en © The author(s) 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
Deng, Zhiting
Wang, Jieqiong
Xiao, Yang
Li, Fei
Niu, Lili
Liu, Xin
Meng, Long
Zheng, Hairong
Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity
title Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity
title_full Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity
title_fullStr Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity
title_full_unstemmed Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity
title_short Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity
title_sort ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7977450/
https://www.ncbi.nlm.nih.gov/pubmed/33754065
http://dx.doi.org/10.7150/thno.52436
work_keys_str_mv AT dengzhiting ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity
AT wangjieqiong ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity
AT xiaoyang ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity
AT lifei ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity
AT niulili ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity
AT liuxin ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity
AT menglong ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity
AT zhenghairong ultrasoundmediatedaugmentedexosomereleasefromastrocytesalleviatesamyloidbinducedneurotoxicity