Cargando…

FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice

Reticulophagy regulator 1 (RETEG1, also known as FAM134B) plays a crucial role in endoplasmic reticulum autophagy. We aimed to explore the effect of FAM134B-mediated endoplasmic reticulum autophagy in sepsis myocardial injury in mice. Sepsis myocardial injury mice were established via cecal ligation...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Tong, Chen, Yongsheng, Li, Yue, Yao, Zhipeng, Liu, Wenhua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8202901/
https://www.ncbi.nlm.nih.gov/pubmed/33819192
http://dx.doi.org/10.18632/aging.202786
_version_ 1783708059341684736
author Li, Tong
Chen, Yongsheng
Li, Yue
Yao, Zhipeng
Liu, Wenhua
author_facet Li, Tong
Chen, Yongsheng
Li, Yue
Yao, Zhipeng
Liu, Wenhua
author_sort Li, Tong
collection PubMed
description Reticulophagy regulator 1 (RETEG1, also known as FAM134B) plays a crucial role in endoplasmic reticulum autophagy. We aimed to explore the effect of FAM134B-mediated endoplasmic reticulum autophagy in sepsis myocardial injury in mice. Sepsis myocardial injury mice were established via cecal ligation and puncture procedures. The expression of FAM134B and LC3-II/I was determined using immunohistochemistry. Myocardial tissue morphological changes and apoptosis were examined using hematoxylin and eosin (H&E) staining and TUNEL analysis. The effects of FAM134B knockdown or overexpression on mice with sepsis myocardial injury were also studied. The levels of TNF-α, IL-6, IL-8, and IL-10 were evaluated using enzyme-linked immunosorbent assay (ELISA). Autophagy- and apoptosis-related protein expression was detected using western blotting. The effect of FAM134B on Lipopolysaccharide (LPS) -induced cardiomyocytes was also studied. The expression of FAM134B and LC3-II/I increased in sepsis mice and lipopolysaccharide (LPS)-treated cardiomyocytes. 3-Methyladenine (3-MA) significantly inhibited FAM134B and LC3-II/I expression and promoted myocardial injury, inflammation response, and cardiomyocyte apoptosis. The overexpression of FAM134B could minimize myocardial injury, inflammation, and apoptosis, whereas FAM134B knockdown showed opposite effects. FAM134B-mediated endoplasmic reticulum autophagy had a protective effect on sepsis myocardial injury in mice by reducing inflammation and tissue apoptosis, which may provide new insights for sepsis myocardial injury therapies.
format Online
Article
Text
id pubmed-8202901
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Impact Journals
record_format MEDLINE/PubMed
spelling pubmed-82029012021-06-15 FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice Li, Tong Chen, Yongsheng Li, Yue Yao, Zhipeng Liu, Wenhua Aging (Albany NY) Research Paper Reticulophagy regulator 1 (RETEG1, also known as FAM134B) plays a crucial role in endoplasmic reticulum autophagy. We aimed to explore the effect of FAM134B-mediated endoplasmic reticulum autophagy in sepsis myocardial injury in mice. Sepsis myocardial injury mice were established via cecal ligation and puncture procedures. The expression of FAM134B and LC3-II/I was determined using immunohistochemistry. Myocardial tissue morphological changes and apoptosis were examined using hematoxylin and eosin (H&E) staining and TUNEL analysis. The effects of FAM134B knockdown or overexpression on mice with sepsis myocardial injury were also studied. The levels of TNF-α, IL-6, IL-8, and IL-10 were evaluated using enzyme-linked immunosorbent assay (ELISA). Autophagy- and apoptosis-related protein expression was detected using western blotting. The effect of FAM134B on Lipopolysaccharide (LPS) -induced cardiomyocytes was also studied. The expression of FAM134B and LC3-II/I increased in sepsis mice and lipopolysaccharide (LPS)-treated cardiomyocytes. 3-Methyladenine (3-MA) significantly inhibited FAM134B and LC3-II/I expression and promoted myocardial injury, inflammation response, and cardiomyocyte apoptosis. The overexpression of FAM134B could minimize myocardial injury, inflammation, and apoptosis, whereas FAM134B knockdown showed opposite effects. FAM134B-mediated endoplasmic reticulum autophagy had a protective effect on sepsis myocardial injury in mice by reducing inflammation and tissue apoptosis, which may provide new insights for sepsis myocardial injury therapies. Impact Journals 2021-03-26 /pmc/articles/PMC8202901/ /pubmed/33819192 http://dx.doi.org/10.18632/aging.202786 Text en Copyright: © 2021 Li et al. https://creativecommons.org/licenses/by/3.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/3.0/) (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Li, Tong
Chen, Yongsheng
Li, Yue
Yao, Zhipeng
Liu, Wenhua
FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice
title FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice
title_full FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice
title_fullStr FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice
title_full_unstemmed FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice
title_short FAM134B-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice
title_sort fam134b-mediated endoplasmic reticulum autophagy protects against sepsis myocardial injury in mice
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8202901/
https://www.ncbi.nlm.nih.gov/pubmed/33819192
http://dx.doi.org/10.18632/aging.202786
work_keys_str_mv AT litong fam134bmediatedendoplasmicreticulumautophagyprotectsagainstsepsismyocardialinjuryinmice
AT chenyongsheng fam134bmediatedendoplasmicreticulumautophagyprotectsagainstsepsismyocardialinjuryinmice
AT liyue fam134bmediatedendoplasmicreticulumautophagyprotectsagainstsepsismyocardialinjuryinmice
AT yaozhipeng fam134bmediatedendoplasmicreticulumautophagyprotectsagainstsepsismyocardialinjuryinmice
AT liuwenhua fam134bmediatedendoplasmicreticulumautophagyprotectsagainstsepsismyocardialinjuryinmice