Cargando…

Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway

Purpose: Functional impairment of endothelial progenitor cells (EPCs) is frequently observed in patients with diabetic vascular complications. Astragaloside IV (ASV) has a significant protective effect against vascular endothelial dysfunction. Thus, this study aimed to investigate the role of ASV on...

Descripción completa

Detalles Bibliográficos
Autores principales: Qian, Weibin, Cai, Xinrui, Qian, Qiuhai, Zhuang, Qianzhu, Yang, Wenjun, Zhang, Xinying, Zhao, Lijie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6677131/
https://www.ncbi.nlm.nih.gov/pubmed/31440038
http://dx.doi.org/10.2147/DDDT.S207774
_version_ 1783440892040839168
author Qian, Weibin
Cai, Xinrui
Qian, Qiuhai
Zhuang, Qianzhu
Yang, Wenjun
Zhang, Xinying
Zhao, Lijie
author_facet Qian, Weibin
Cai, Xinrui
Qian, Qiuhai
Zhuang, Qianzhu
Yang, Wenjun
Zhang, Xinying
Zhao, Lijie
author_sort Qian, Weibin
collection PubMed
description Purpose: Functional impairment of endothelial progenitor cells (EPCs) is frequently observed in patients with diabetic vascular complications. Astragaloside IV (ASV) has a significant protective effect against vascular endothelial dysfunction. Thus, this study aimed to investigate the role of ASV on oxidized low-density lipoprotein (ox-LDL)-induced EPCs dysfunction and its potential mechanisms. Methods: EPCs were isolated from the peripheral blood of mice and treated with different concentration of ASV (10, 20, 40, 60, 80, 100 and 200 µM). ox-LDL was served as a stimulus for cell model. The proliferation and migration, and improved tube formation ability of EPCs were determined. Reactive oxygen species (ROS) production and the levels of inflammatory cytokines, including interleukin 1β (IL-1β), IL-6, IL-10 and tumor necrosis factor (TNF-α) were measured. The expression oflectin-like oxidized LDL receptor (LOX-1) andNod-like receptor nucleotide-binding domain leucine rich repeat containing protein 3 (NLRP3) inflammasome were detected by Western blot analysis. Results: We found ASV treatment alleviated ox-LDL-induced cellular dysfunction, as evidenced by promoted proliferation and migration, and improved tube formation ability. Besides, ASV treatment significantly suppressed ox-LDL-induced ROS production and the levels of inflammatory cytokines. ASV inhibited ox-LDL-induced expression of LOX-1 in a concentration-dependent manner. Overexpression of LOX-1 in EPCs triggered NLRP3inflammasome activation, while inhibition of LOX-1 or treatment with ASV suppressed ox-LDL-induced NLRP3 inflammasome activation. Furthermore, overexpression of LOX-1 in ox-LDL-induced EPCs furtherly impaired cellular function, which could be ameliorated by ASV treatment. Conclusion: Our study showed that ASV may protect EPCs against ox-LDL-induced dysfunction via LOX-1/NLRP3 pathway.
format Online
Article
Text
id pubmed-6677131
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-66771312019-08-22 Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway Qian, Weibin Cai, Xinrui Qian, Qiuhai Zhuang, Qianzhu Yang, Wenjun Zhang, Xinying Zhao, Lijie Drug Des Devel Ther Original Research Purpose: Functional impairment of endothelial progenitor cells (EPCs) is frequently observed in patients with diabetic vascular complications. Astragaloside IV (ASV) has a significant protective effect against vascular endothelial dysfunction. Thus, this study aimed to investigate the role of ASV on oxidized low-density lipoprotein (ox-LDL)-induced EPCs dysfunction and its potential mechanisms. Methods: EPCs were isolated from the peripheral blood of mice and treated with different concentration of ASV (10, 20, 40, 60, 80, 100 and 200 µM). ox-LDL was served as a stimulus for cell model. The proliferation and migration, and improved tube formation ability of EPCs were determined. Reactive oxygen species (ROS) production and the levels of inflammatory cytokines, including interleukin 1β (IL-1β), IL-6, IL-10 and tumor necrosis factor (TNF-α) were measured. The expression oflectin-like oxidized LDL receptor (LOX-1) andNod-like receptor nucleotide-binding domain leucine rich repeat containing protein 3 (NLRP3) inflammasome were detected by Western blot analysis. Results: We found ASV treatment alleviated ox-LDL-induced cellular dysfunction, as evidenced by promoted proliferation and migration, and improved tube formation ability. Besides, ASV treatment significantly suppressed ox-LDL-induced ROS production and the levels of inflammatory cytokines. ASV inhibited ox-LDL-induced expression of LOX-1 in a concentration-dependent manner. Overexpression of LOX-1 in EPCs triggered NLRP3inflammasome activation, while inhibition of LOX-1 or treatment with ASV suppressed ox-LDL-induced NLRP3 inflammasome activation. Furthermore, overexpression of LOX-1 in ox-LDL-induced EPCs furtherly impaired cellular function, which could be ameliorated by ASV treatment. Conclusion: Our study showed that ASV may protect EPCs against ox-LDL-induced dysfunction via LOX-1/NLRP3 pathway. Dove 2019-07-29 /pmc/articles/PMC6677131/ /pubmed/31440038 http://dx.doi.org/10.2147/DDDT.S207774 Text en © 2019 Qian 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
Qian, Weibin
Cai, Xinrui
Qian, Qiuhai
Zhuang, Qianzhu
Yang, Wenjun
Zhang, Xinying
Zhao, Lijie
Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway
title Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway
title_full Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway
title_fullStr Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway
title_full_unstemmed Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway
title_short Astragaloside IV protects endothelial progenitor cells from the damage of ox-LDL via the LOX-1/NLRP3 inflammasome pathway
title_sort astragaloside iv protects endothelial progenitor cells from the damage of ox-ldl via the lox-1/nlrp3 inflammasome pathway
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6677131/
https://www.ncbi.nlm.nih.gov/pubmed/31440038
http://dx.doi.org/10.2147/DDDT.S207774
work_keys_str_mv AT qianweibin astragalosideivprotectsendothelialprogenitorcellsfromthedamageofoxldlviathelox1nlrp3inflammasomepathway
AT caixinrui astragalosideivprotectsendothelialprogenitorcellsfromthedamageofoxldlviathelox1nlrp3inflammasomepathway
AT qianqiuhai astragalosideivprotectsendothelialprogenitorcellsfromthedamageofoxldlviathelox1nlrp3inflammasomepathway
AT zhuangqianzhu astragalosideivprotectsendothelialprogenitorcellsfromthedamageofoxldlviathelox1nlrp3inflammasomepathway
AT yangwenjun astragalosideivprotectsendothelialprogenitorcellsfromthedamageofoxldlviathelox1nlrp3inflammasomepathway
AT zhangxinying astragalosideivprotectsendothelialprogenitorcellsfromthedamageofoxldlviathelox1nlrp3inflammasomepathway
AT zhaolijie astragalosideivprotectsendothelialprogenitorcellsfromthedamageofoxldlviathelox1nlrp3inflammasomepathway