Cargando…

Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice

Obstructive sleep apnea (OSA) patients exhibit different degrees of cognitive impairment, which is related to the activation of reactive oxygen species (ROS) production by chronic intermittent hypoxia (CIH) and the deposition of iron in the brain. As a central regulator of iron homeostasis, whether...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhao, Ya-Shuo, Tan, Miao, Song, Ji-Xian, An, Ji-Ren, Yang, Xin-Yue, Li, Wen-Ya, Guo, Ya-Jing, Ji, En-Sheng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8357469/
https://www.ncbi.nlm.nih.gov/pubmed/34394834
http://dx.doi.org/10.1155/2021/8520967
_version_ 1783737133540835328
author Zhao, Ya-Shuo
Tan, Miao
Song, Ji-Xian
An, Ji-Ren
Yang, Xin-Yue
Li, Wen-Ya
Guo, Ya-Jing
Ji, En-Sheng
author_facet Zhao, Ya-Shuo
Tan, Miao
Song, Ji-Xian
An, Ji-Ren
Yang, Xin-Yue
Li, Wen-Ya
Guo, Ya-Jing
Ji, En-Sheng
author_sort Zhao, Ya-Shuo
collection PubMed
description Obstructive sleep apnea (OSA) patients exhibit different degrees of cognitive impairment, which is related to the activation of reactive oxygen species (ROS) production by chronic intermittent hypoxia (CIH) and the deposition of iron in the brain. As a central regulator of iron homeostasis, whether hepcidin is involved in OSA-induced cognitive impairment has not been clarified. In order to simulate OSA, we established the mouse model by reducing the percentage of inspired O(2) (FiO(2)) from 21% to 5%, 20 times/h for 8 h/day. We found hepcidin was rising during CIH, along with increasing iron levels and neuron loss. Then, we constructed a mouse with astrocyte-specific knockdown hepcidin gene (shHamp). During CIH exposure, the shHamp mice showed a lower level of total iron and neuronal iron in the hippocampus, via stabilizing ferroportin 1 (FPN1) and decreasing L-ferritin (FTL) levels, when compared with wild-type (WT) mice. Furthermore, the shHamp mice showed a decrease of ROS by downregulating the elevated NADPH oxidase (NOX2) and 4-hydroxynonenal (4-HNE) levels mediated by CIH. In addition, the shHamp mice presented improved cognitive deficit by improving synaptic plasticity and BDNF expression in the hippocampus when subjected to CIH. Therefore, our data revealed that highly expressed hepcidin might promote the degradation of FPN1, resulting in neuronal iron deposition, oxidative stress damage, reduced synaptic plasticity, and impaired cognitive performance during CIH exposure.
format Online
Article
Text
id pubmed-8357469
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-83574692021-08-12 Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice Zhao, Ya-Shuo Tan, Miao Song, Ji-Xian An, Ji-Ren Yang, Xin-Yue Li, Wen-Ya Guo, Ya-Jing Ji, En-Sheng Oxid Med Cell Longev Research Article Obstructive sleep apnea (OSA) patients exhibit different degrees of cognitive impairment, which is related to the activation of reactive oxygen species (ROS) production by chronic intermittent hypoxia (CIH) and the deposition of iron in the brain. As a central regulator of iron homeostasis, whether hepcidin is involved in OSA-induced cognitive impairment has not been clarified. In order to simulate OSA, we established the mouse model by reducing the percentage of inspired O(2) (FiO(2)) from 21% to 5%, 20 times/h for 8 h/day. We found hepcidin was rising during CIH, along with increasing iron levels and neuron loss. Then, we constructed a mouse with astrocyte-specific knockdown hepcidin gene (shHamp). During CIH exposure, the shHamp mice showed a lower level of total iron and neuronal iron in the hippocampus, via stabilizing ferroportin 1 (FPN1) and decreasing L-ferritin (FTL) levels, when compared with wild-type (WT) mice. Furthermore, the shHamp mice showed a decrease of ROS by downregulating the elevated NADPH oxidase (NOX2) and 4-hydroxynonenal (4-HNE) levels mediated by CIH. In addition, the shHamp mice presented improved cognitive deficit by improving synaptic plasticity and BDNF expression in the hippocampus when subjected to CIH. Therefore, our data revealed that highly expressed hepcidin might promote the degradation of FPN1, resulting in neuronal iron deposition, oxidative stress damage, reduced synaptic plasticity, and impaired cognitive performance during CIH exposure. Hindawi 2021-08-04 /pmc/articles/PMC8357469/ /pubmed/34394834 http://dx.doi.org/10.1155/2021/8520967 Text en Copyright © 2021 Ya-Shuo Zhao et al. https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Zhao, Ya-Shuo
Tan, Miao
Song, Ji-Xian
An, Ji-Ren
Yang, Xin-Yue
Li, Wen-Ya
Guo, Ya-Jing
Ji, En-Sheng
Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice
title Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice
title_full Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice
title_fullStr Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice
title_full_unstemmed Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice
title_short Involvement of Hepcidin in Cognitive Damage Induced by Chronic Intermittent Hypoxia in Mice
title_sort involvement of hepcidin in cognitive damage induced by chronic intermittent hypoxia in mice
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8357469/
https://www.ncbi.nlm.nih.gov/pubmed/34394834
http://dx.doi.org/10.1155/2021/8520967
work_keys_str_mv AT zhaoyashuo involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice
AT tanmiao involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice
AT songjixian involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice
AT anjiren involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice
AT yangxinyue involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice
AT liwenya involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice
AT guoyajing involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice
AT jiensheng involvementofhepcidinincognitivedamageinducedbychronicintermittenthypoxiainmice