Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration

Clinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and...

Full description

Bibliographic Details
Main Authors: Pan Lu, Feng Liang, Yuanlin Dong, Zhongcong Xie, Yiying Zhang
Format: Article
Language:English
Published: MDPI AG 2023-04-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/24/7/6746
_version_ 1797607758455898112
author Pan Lu
Feng Liang
Yuanlin Dong
Zhongcong Xie
Yiying Zhang
author_facet Pan Lu
Feng Liang
Yuanlin Dong
Zhongcong Xie
Yiying Zhang
author_sort Pan Lu
collection DOAJ
description Clinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and mitochondrial function. Neural progenitor cells (NPCs) migration is associated with cognitive function in developing brains. However, it is unclear whether sevoflurane can regulate NPCs migration via changes in CypD. To address this question, we treated NPCs harvested from wild-type (WT) and CypD knockout (KO) mice and young WT and CypD KO mice with sevoflurane. We used immunofluorescence staining, wound healing assay, transwell assay, mass spectrometry, and Western blot to assess the effects of sevoflurane on CypD, reactive oxygen species (ROS), doublecortin levels, and NPCs migration. We showed that sevoflurane increased levels of CypD and ROS, decreased levels of doublecortin, and reduced migration of NPCs harvested from WT mice in vitro and in WT young mice. KO of CypD attenuated these effects, suggesting that a sevoflurane-induced decrease in NPCs migration is dependent on CypD. Our findings have established a system for future studies aimed at exploring the impacts of sevoflurane anesthesia on the impairment of NPCs migration.
first_indexed 2024-03-11T05:34:06Z
format Article
id doaj.art-b1444b25d66949708244d897f6323b06
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-11T05:34:06Z
publishDate 2023-04-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-b1444b25d66949708244d897f6323b062023-11-17T16:55:28ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672023-04-01247674610.3390/ijms24076746Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells MigrationPan Lu0Feng Liang1Yuanlin Dong2Zhongcong Xie3Yiying Zhang4Department of Anesthesia, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, ChinaDepartment of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USADepartment of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USADepartment of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USADepartment of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USAClinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and mitochondrial function. Neural progenitor cells (NPCs) migration is associated with cognitive function in developing brains. However, it is unclear whether sevoflurane can regulate NPCs migration via changes in CypD. To address this question, we treated NPCs harvested from wild-type (WT) and CypD knockout (KO) mice and young WT and CypD KO mice with sevoflurane. We used immunofluorescence staining, wound healing assay, transwell assay, mass spectrometry, and Western blot to assess the effects of sevoflurane on CypD, reactive oxygen species (ROS), doublecortin levels, and NPCs migration. We showed that sevoflurane increased levels of CypD and ROS, decreased levels of doublecortin, and reduced migration of NPCs harvested from WT mice in vitro and in WT young mice. KO of CypD attenuated these effects, suggesting that a sevoflurane-induced decrease in NPCs migration is dependent on CypD. Our findings have established a system for future studies aimed at exploring the impacts of sevoflurane anesthesia on the impairment of NPCs migration.https://www.mdpi.com/1422-0067/24/7/6746sevofluranecyclophilin Dneural progenitor cellsmigrationdoublecortin
spellingShingle Pan Lu
Feng Liang
Yuanlin Dong
Zhongcong Xie
Yiying Zhang
Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration
International Journal of Molecular Sciences
sevoflurane
cyclophilin D
neural progenitor cells
migration
doublecortin
title Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration
title_full Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration
title_fullStr Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration
title_full_unstemmed Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration
title_short Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration
title_sort sevoflurane induces a cyclophilin d dependent decrease of neural progenitor cells migration
topic sevoflurane
cyclophilin D
neural progenitor cells
migration
doublecortin
url https://www.mdpi.com/1422-0067/24/7/6746
work_keys_str_mv AT panlu sevofluraneinducesacyclophilinddependentdecreaseofneuralprogenitorcellsmigration
AT fengliang sevofluraneinducesacyclophilinddependentdecreaseofneuralprogenitorcellsmigration
AT yuanlindong sevofluraneinducesacyclophilinddependentdecreaseofneuralprogenitorcellsmigration
AT zhongcongxie sevofluraneinducesacyclophilinddependentdecreaseofneuralprogenitorcellsmigration
AT yiyingzhang sevofluraneinducesacyclophilinddependentdecreaseofneuralprogenitorcellsmigration