Cargando…

Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation

Intermittent hypoxia (IH)-associated cognition decline is related to the neuroinflammation of microglia. SUMOylation is a post-translational modification related to multiple human diseases, which can be reversed by SENP1. Studies showed that SENP1 and PPARγ play essential roles in restricting inflam...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Hongwei, Xiong, Wei, Hang, Sitong, Wang, Yanmin, Zhang, Sisen, Liu, Song
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8221356/
https://www.ncbi.nlm.nih.gov/pubmed/34035184
http://dx.doi.org/10.18632/aging.203084
_version_ 1783711315091521536
author Wang, Hongwei
Xiong, Wei
Hang, Sitong
Wang, Yanmin
Zhang, Sisen
Liu, Song
author_facet Wang, Hongwei
Xiong, Wei
Hang, Sitong
Wang, Yanmin
Zhang, Sisen
Liu, Song
author_sort Wang, Hongwei
collection PubMed
description Intermittent hypoxia (IH)-associated cognition decline is related to the neuroinflammation of microglia. SUMOylation is a post-translational modification related to multiple human diseases, which can be reversed by SENP1. Studies showed that SENP1 and PPARγ play essential roles in restricting inflammation by blocking NF-κB activation. However, the mechanism remains unclear. Herein, we investigated the precise mechanism underlying SENP1 and PPARγ in cognitive decline after IH insult. Biochemical analysis results revealed that IH triggered the inflammatory response and neuronal apoptosis, increased the SUMOylation of PPARγ, and decreased the level of PPARγ compared to that in the normoxia group. After SENP1 downregulation, the inflammatory response, neuronal apoptosis and the SUMOylation of PPARγ were enhanced, and the level of PPARγ was further decreased in vitro and in vivo. However, the application of PPARγ agonist, GW1929, abolished the enhancement of inflammation and neuronal apoptosis in vitro. The Morris Water Maze results showed that both IH groups mice exhibited longer latency and shorter dwell-time in the goal quadrant than normoxia groups. Notably, SENP1 downregulation aggravated these alterations. Overall, these results showed that SENP1 played an essential role in IH-associated cognitive dysfunction. SENP1 depletion aggravated neuroinflammation and neuronal apoptosis via promoting the SUMOylation of PPARγ, reducing the level of PPARγ, thus exaggerating IH-induced cognitive decline.
format Online
Article
Text
id pubmed-8221356
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Impact Journals
record_format MEDLINE/PubMed
spelling pubmed-82213562021-06-26 Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation Wang, Hongwei Xiong, Wei Hang, Sitong Wang, Yanmin Zhang, Sisen Liu, Song Aging (Albany NY) Research Paper Intermittent hypoxia (IH)-associated cognition decline is related to the neuroinflammation of microglia. SUMOylation is a post-translational modification related to multiple human diseases, which can be reversed by SENP1. Studies showed that SENP1 and PPARγ play essential roles in restricting inflammation by blocking NF-κB activation. However, the mechanism remains unclear. Herein, we investigated the precise mechanism underlying SENP1 and PPARγ in cognitive decline after IH insult. Biochemical analysis results revealed that IH triggered the inflammatory response and neuronal apoptosis, increased the SUMOylation of PPARγ, and decreased the level of PPARγ compared to that in the normoxia group. After SENP1 downregulation, the inflammatory response, neuronal apoptosis and the SUMOylation of PPARγ were enhanced, and the level of PPARγ was further decreased in vitro and in vivo. However, the application of PPARγ agonist, GW1929, abolished the enhancement of inflammation and neuronal apoptosis in vitro. The Morris Water Maze results showed that both IH groups mice exhibited longer latency and shorter dwell-time in the goal quadrant than normoxia groups. Notably, SENP1 downregulation aggravated these alterations. Overall, these results showed that SENP1 played an essential role in IH-associated cognitive dysfunction. SENP1 depletion aggravated neuroinflammation and neuronal apoptosis via promoting the SUMOylation of PPARγ, reducing the level of PPARγ, thus exaggerating IH-induced cognitive decline. Impact Journals 2021-05-25 /pmc/articles/PMC8221356/ /pubmed/34035184 http://dx.doi.org/10.18632/aging.203084 Text en Copyright: © 2021 Wang 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
Wang, Hongwei
Xiong, Wei
Hang, Sitong
Wang, Yanmin
Zhang, Sisen
Liu, Song
Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation
title Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation
title_full Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation
title_fullStr Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation
title_full_unstemmed Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation
title_short Depletion of SENP1-mediated PPARγ SUMOylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation
title_sort depletion of senp1-mediated pparγ sumoylation exaggerates intermittent hypoxia-induced cognitive decline by aggravating microglia-mediated neuroinflammation
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8221356/
https://www.ncbi.nlm.nih.gov/pubmed/34035184
http://dx.doi.org/10.18632/aging.203084
work_keys_str_mv AT wanghongwei depletionofsenp1mediatedppargsumoylationexaggeratesintermittenthypoxiainducedcognitivedeclinebyaggravatingmicrogliamediatedneuroinflammation
AT xiongwei depletionofsenp1mediatedppargsumoylationexaggeratesintermittenthypoxiainducedcognitivedeclinebyaggravatingmicrogliamediatedneuroinflammation
AT hangsitong depletionofsenp1mediatedppargsumoylationexaggeratesintermittenthypoxiainducedcognitivedeclinebyaggravatingmicrogliamediatedneuroinflammation
AT wangyanmin depletionofsenp1mediatedppargsumoylationexaggeratesintermittenthypoxiainducedcognitivedeclinebyaggravatingmicrogliamediatedneuroinflammation
AT zhangsisen depletionofsenp1mediatedppargsumoylationexaggeratesintermittenthypoxiainducedcognitivedeclinebyaggravatingmicrogliamediatedneuroinflammation
AT liusong depletionofsenp1mediatedppargsumoylationexaggeratesintermittenthypoxiainducedcognitivedeclinebyaggravatingmicrogliamediatedneuroinflammation