Cargando…

Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling

Brain renin‐angiotensin (Ang) system (RAS) is implicated in neuroinflammation, a major characteristic of aging process. Angiotensin (Ang) II, produced by angiotensin‐converting enzyme (ACE), activates immune system via angiotensin type 1 receptor (AT1), whereas Ang(1–7), generated by ACE2, binds wit...

Descripción completa

Detalles Bibliográficos
Autores principales: Dang, Ruili, Yang, Mengqi, Cui, Changmeng, Wang, Changshui, Zhang, Wenyuan, Geng, Chunmei, Han, Wenxiu, Jiang, Pei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8520723/
https://www.ncbi.nlm.nih.gov/pubmed/34529881
http://dx.doi.org/10.1111/acel.13480
_version_ 1784584730734231552
author Dang, Ruili
Yang, Mengqi
Cui, Changmeng
Wang, Changshui
Zhang, Wenyuan
Geng, Chunmei
Han, Wenxiu
Jiang, Pei
author_facet Dang, Ruili
Yang, Mengqi
Cui, Changmeng
Wang, Changshui
Zhang, Wenyuan
Geng, Chunmei
Han, Wenxiu
Jiang, Pei
author_sort Dang, Ruili
collection PubMed
description Brain renin‐angiotensin (Ang) system (RAS) is implicated in neuroinflammation, a major characteristic of aging process. Angiotensin (Ang) II, produced by angiotensin‐converting enzyme (ACE), activates immune system via angiotensin type 1 receptor (AT1), whereas Ang(1–7), generated by ACE2, binds with Mas receptor (MasR) to restrain excessive inflammatory response. Therefore, the present study aims to explore the relationship between RAS and neuroinflammation. We found that repeated lipopolysaccharide (LPS) treatment shifted the balance between ACE/Ang II/AT1 and ACE2/Ang(1–7)/MasR axis to the deleterious side and treatment with either MasR agonist, AVE0991 (AVE) or ACE2 activator, diminazene aceturate, exhibited strong neuroprotective actions. Mechanically, activation of ACE2/Ang(1–7)/MasR axis triggered the Forkhead box class O1 (FOXO1)‐autophagy pathway and induced superoxide dismutase (SOD) and catalase (CAT), the FOXO1‐targeted antioxidant enzymes. Meanwhile, knockdown of MasR or FOXO1 in BV2 cells, or using the selective FOXO1 inhibitor, AS1842856, in animals, suppressed FOXO1 translocation and compromised the autophagic process induced by MasR activation. We further used chloroquine (CQ) to block autophagy and showed that suppressing either FOXO1 or autophagy abrogated the anti‐inflammatory action of AVE. Likewise, Ang(1–7) also induced FOXO1 signaling and autophagic flux following LPS treatment in BV2 cells. Cotreatment with AS1842856 or CQ all led to autophagic inhibition and thereby abolished Ang(1–7)‐induced remission on NLRP3 inflammasome activation caused by LPS exposure, shifting the microglial polarization from M1 to M2 phenotype. Collectively, these results firstly illustrated the mechanism of ACE2/Ang(1–7)/MasR axis in neuroinflammation, strongly indicating the involvement of FOXO1‐mediated autophagy in the neuroimmune‐modulating effects triggered by MasR activation.
format Online
Article
Text
id pubmed-8520723
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-85207232021-10-25 Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling Dang, Ruili Yang, Mengqi Cui, Changmeng Wang, Changshui Zhang, Wenyuan Geng, Chunmei Han, Wenxiu Jiang, Pei Aging Cell Original Papers Brain renin‐angiotensin (Ang) system (RAS) is implicated in neuroinflammation, a major characteristic of aging process. Angiotensin (Ang) II, produced by angiotensin‐converting enzyme (ACE), activates immune system via angiotensin type 1 receptor (AT1), whereas Ang(1–7), generated by ACE2, binds with Mas receptor (MasR) to restrain excessive inflammatory response. Therefore, the present study aims to explore the relationship between RAS and neuroinflammation. We found that repeated lipopolysaccharide (LPS) treatment shifted the balance between ACE/Ang II/AT1 and ACE2/Ang(1–7)/MasR axis to the deleterious side and treatment with either MasR agonist, AVE0991 (AVE) or ACE2 activator, diminazene aceturate, exhibited strong neuroprotective actions. Mechanically, activation of ACE2/Ang(1–7)/MasR axis triggered the Forkhead box class O1 (FOXO1)‐autophagy pathway and induced superoxide dismutase (SOD) and catalase (CAT), the FOXO1‐targeted antioxidant enzymes. Meanwhile, knockdown of MasR or FOXO1 in BV2 cells, or using the selective FOXO1 inhibitor, AS1842856, in animals, suppressed FOXO1 translocation and compromised the autophagic process induced by MasR activation. We further used chloroquine (CQ) to block autophagy and showed that suppressing either FOXO1 or autophagy abrogated the anti‐inflammatory action of AVE. Likewise, Ang(1–7) also induced FOXO1 signaling and autophagic flux following LPS treatment in BV2 cells. Cotreatment with AS1842856 or CQ all led to autophagic inhibition and thereby abolished Ang(1–7)‐induced remission on NLRP3 inflammasome activation caused by LPS exposure, shifting the microglial polarization from M1 to M2 phenotype. Collectively, these results firstly illustrated the mechanism of ACE2/Ang(1–7)/MasR axis in neuroinflammation, strongly indicating the involvement of FOXO1‐mediated autophagy in the neuroimmune‐modulating effects triggered by MasR activation. John Wiley and Sons Inc. 2021-09-16 2021-10 /pmc/articles/PMC8520723/ /pubmed/34529881 http://dx.doi.org/10.1111/acel.13480 Text en © 2021 The Authors. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Papers
Dang, Ruili
Yang, Mengqi
Cui, Changmeng
Wang, Changshui
Zhang, Wenyuan
Geng, Chunmei
Han, Wenxiu
Jiang, Pei
Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling
title Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling
title_full Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling
title_fullStr Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling
title_full_unstemmed Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling
title_short Activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling
title_sort activation of angiotensin‐converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class o1 signaling
topic Original Papers
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8520723/
https://www.ncbi.nlm.nih.gov/pubmed/34529881
http://dx.doi.org/10.1111/acel.13480
work_keys_str_mv AT dangruili activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling
AT yangmengqi activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling
AT cuichangmeng activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling
AT wangchangshui activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling
AT zhangwenyuan activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling
AT gengchunmei activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling
AT hanwenxiu activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling
AT jiangpei activationofangiotensinconvertingenzyme2angiotensin17masreceptoraxistriggersautophagyandsuppressesmicrogliaproinflammatorypolarizationviaforkheadboxclasso1signaling