MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndrome

PurposeAcute respiratory distress syndrome (ARDS) is a prevalent illness in intensive care units. Extracellular vesicles and particles released from activated alveolar macrophages (AMs) assist in ARDS lung injury and the inflammatory process through mechanisms that are unclear. This study investigat...

Full description

Bibliographic Details
Main Authors: Xinyi Xu, Xu Liu, Xuecheng Dong, Yi Yang, Ling Liu
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-11-01
Series:Frontiers in Cellular and Infection Microbiology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fcimb.2022.1061790/full
_version_ 1811215603519717376
author Xinyi Xu
Xu Liu
Xuecheng Dong
Yi Yang
Ling Liu
author_facet Xinyi Xu
Xu Liu
Xuecheng Dong
Yi Yang
Ling Liu
author_sort Xinyi Xu
collection DOAJ
description PurposeAcute respiratory distress syndrome (ARDS) is a prevalent illness in intensive care units. Extracellular vesicles and particles released from activated alveolar macrophages (AMs) assist in ARDS lung injury and the inflammatory process through mechanisms that are unclear. This study investigated the role of AM-derived secretory autophagosomes (SAPs) in lung injury and microRNA (MiR)-199a-3p-regulated inflammation associated with ARDS in vitro and in a murine model.MethodsThe ARDS model in mouse was established by intratracheal LPS lipopolysaccharide (LPS) injection. The agomirs or antagomirs of MiR-199a-3p were injected into the caudal vein to figure out whether MiR-199a-3p could influence ARDS inflammation and lung injury, whereas the mimics or inhibitors of MiR-199a-3p, siRNA of Rab8a, or PAK4 inhibitor were transfected or applied to RAW264.7 cells to evaluate the mechanism of SAP release. Culture supernatants of RAW264.7 cells treated with LPS or bronchoalveolar lavage fluid from mice were collected for the isolation of SAPs.ResultsWe found that MiR-199a-3p was over-expressed in the lungs of ARDS mice. The MiR-199a-3p antagomir alleviated, whereas the MiR-199a-3p agomir exacerbated LPS-induced inflammation in mice by promoting AM-derived SAP secretion. In addition, MiR-199a-3p over-expression exacerbated LPS-induced ARDS via activating Rab8a, and Rab8a silencing significantly suppressed the promoting influence of the MiR-199a-3p mimic on SAP secretion. Furthermore, MiR-199a-3p mimic activated Rab8a by directly inhibiting PAK4 expression.ConclusionThe novel finding of this study is that MiR-199a-3p participated in the regulation of SAP secretion and the inflammatory process via targeting of PAK4/Rab8a, and is a potential therapeutic candidate for ARDS treatment.
first_indexed 2024-04-12T06:25:26Z
format Article
id doaj.art-f0ba1a38bea6435585ec4b1699f02262
institution Directory Open Access Journal
issn 2235-2988
language English
last_indexed 2024-04-12T06:25:26Z
publishDate 2022-11-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Cellular and Infection Microbiology
spelling doaj.art-f0ba1a38bea6435585ec4b1699f022622022-12-22T03:44:11ZengFrontiers Media S.A.Frontiers in Cellular and Infection Microbiology2235-29882022-11-011210.3389/fcimb.2022.10617901061790MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndromeXinyi XuXu LiuXuecheng DongYi YangLing LiuPurposeAcute respiratory distress syndrome (ARDS) is a prevalent illness in intensive care units. Extracellular vesicles and particles released from activated alveolar macrophages (AMs) assist in ARDS lung injury and the inflammatory process through mechanisms that are unclear. This study investigated the role of AM-derived secretory autophagosomes (SAPs) in lung injury and microRNA (MiR)-199a-3p-regulated inflammation associated with ARDS in vitro and in a murine model.MethodsThe ARDS model in mouse was established by intratracheal LPS lipopolysaccharide (LPS) injection. The agomirs or antagomirs of MiR-199a-3p were injected into the caudal vein to figure out whether MiR-199a-3p could influence ARDS inflammation and lung injury, whereas the mimics or inhibitors of MiR-199a-3p, siRNA of Rab8a, or PAK4 inhibitor were transfected or applied to RAW264.7 cells to evaluate the mechanism of SAP release. Culture supernatants of RAW264.7 cells treated with LPS or bronchoalveolar lavage fluid from mice were collected for the isolation of SAPs.ResultsWe found that MiR-199a-3p was over-expressed in the lungs of ARDS mice. The MiR-199a-3p antagomir alleviated, whereas the MiR-199a-3p agomir exacerbated LPS-induced inflammation in mice by promoting AM-derived SAP secretion. In addition, MiR-199a-3p over-expression exacerbated LPS-induced ARDS via activating Rab8a, and Rab8a silencing significantly suppressed the promoting influence of the MiR-199a-3p mimic on SAP secretion. Furthermore, MiR-199a-3p mimic activated Rab8a by directly inhibiting PAK4 expression.ConclusionThe novel finding of this study is that MiR-199a-3p participated in the regulation of SAP secretion and the inflammatory process via targeting of PAK4/Rab8a, and is a potential therapeutic candidate for ARDS treatment.https://www.frontiersin.org/articles/10.3389/fcimb.2022.1061790/fullsecretory autophagosomealveolar macrophagemiR-199a-3pRAB8APAK4acute respiratory distress syndrome
spellingShingle Xinyi Xu
Xu Liu
Xuecheng Dong
Yi Yang
Ling Liu
MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndrome
Frontiers in Cellular and Infection Microbiology
secretory autophagosome
alveolar macrophage
miR-199a-3p
RAB8A
PAK4
acute respiratory distress syndrome
title MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndrome
title_full MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndrome
title_fullStr MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndrome
title_full_unstemmed MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndrome
title_short MiR-199a-3p-regulated alveolar macrophage-derived secretory autophagosomes exacerbate lipopolysaccharide-induced acute respiratory distress syndrome
title_sort mir 199a 3p regulated alveolar macrophage derived secretory autophagosomes exacerbate lipopolysaccharide induced acute respiratory distress syndrome
topic secretory autophagosome
alveolar macrophage
miR-199a-3p
RAB8A
PAK4
acute respiratory distress syndrome
url https://www.frontiersin.org/articles/10.3389/fcimb.2022.1061790/full
work_keys_str_mv AT xinyixu mir199a3pregulatedalveolarmacrophagederivedsecretoryautophagosomesexacerbatelipopolysaccharideinducedacuterespiratorydistresssyndrome
AT xuliu mir199a3pregulatedalveolarmacrophagederivedsecretoryautophagosomesexacerbatelipopolysaccharideinducedacuterespiratorydistresssyndrome
AT xuechengdong mir199a3pregulatedalveolarmacrophagederivedsecretoryautophagosomesexacerbatelipopolysaccharideinducedacuterespiratorydistresssyndrome
AT yiyang mir199a3pregulatedalveolarmacrophagederivedsecretoryautophagosomesexacerbatelipopolysaccharideinducedacuterespiratorydistresssyndrome
AT lingliu mir199a3pregulatedalveolarmacrophagederivedsecretoryautophagosomesexacerbatelipopolysaccharideinducedacuterespiratorydistresssyndrome