Cargando…

Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor

BACKGROUND: Perioperative neurocognitive disorders (PNDs) are considered the most common postoperative complication in geriatric patients. However, its pathogenesis is not fully understood. Surgery-triggered neuroinflammation is a major contributor to the development of PNDs. Neuroinflammation can i...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Bo, Qin, Guangcheng, Xiao, Jingyu, Deng, Xiaoyuan, Lin, Aolei, Liu, Hongliang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9281167/
https://www.ncbi.nlm.nih.gov/pubmed/35831873
http://dx.doi.org/10.1186/s12974-022-02528-5
_version_ 1784746819919544320
author Chen, Bo
Qin, Guangcheng
Xiao, Jingyu
Deng, Xiaoyuan
Lin, Aolei
Liu, Hongliang
author_facet Chen, Bo
Qin, Guangcheng
Xiao, Jingyu
Deng, Xiaoyuan
Lin, Aolei
Liu, Hongliang
author_sort Chen, Bo
collection PubMed
description BACKGROUND: Perioperative neurocognitive disorders (PNDs) are considered the most common postoperative complication in geriatric patients. However, its pathogenesis is not fully understood. Surgery-triggered neuroinflammation is a major contributor to the development of PNDs. Neuroinflammation can influence N-methyl-D-aspartate receptor (NMDAR) expression or function which is closely associated with cognition. We, therefore, hypothesized that the persistent changes in NMDAR expression or function induced by transient neuroinflammation after surgery were involved in the development of PNDs. METHODS: Eighteen-month-old male Sprague–Dawley rats were subjected to abdominal surgery with sevoflurane anesthesia to establish the PNDs animal model. Then, we determined the transient neuroinflammation by detecting the protein levels of proinflammatory cytokines and microglia activation using ELISA, western blot, immunohistochemistry, and microglial morphological analysis from postoperative days 1–20. Persistent changes in NMDAR expression were determined by detecting the protein levels of NMDAR subunits from postoperative days 1–59. Subsequently, the dysfunction of synaptic NMDAR was evaluated by detecting the structural plasticity of dendritic spine using Golgi staining. Pull-down assay and western blot were used to detect the protein levels of Rac1-GTP, phosphor-cofilin, and Arp3, which contribute to the regulation of the structural plasticity of dendritic spine. Finally, glycyrrhizin, an anti-inflammatory agent, was administered to further explore the role of synaptic NMDAR dysfunction induced by transient neuroinflammation in the neuropathogenesis of PNDs. RESULTS: We showed that transient neuroinflammation induced by surgery caused sustained downregulation of synaptic NR2A and NR2B subunits in the dorsal hippocampus and led to a selective long-term spatial memory deficit. Meanwhile, the detrimental effect of neuroinflammation on the function of synaptic NMDARs was shown by the impaired structural plasticity of dendritic spines and decreased activity of the Rac1 signaling pathways during learning. Furthermore, anti-inflammatory treatment reversed the downregulation and hypofunction of synaptic NR2A and NR2B and subsequently rescued the long-term spatial memory deficit. CONCLUSIONS: Our results identify sustained synaptic NR2A and NR2B downregulation and hypofunction induced by transient neuroinflammation following surgery as important contributors to the development of PNDs in elderly rats. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02528-5.
format Online
Article
Text
id pubmed-9281167
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-92811672022-07-15 Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor Chen, Bo Qin, Guangcheng Xiao, Jingyu Deng, Xiaoyuan Lin, Aolei Liu, Hongliang J Neuroinflammation Research BACKGROUND: Perioperative neurocognitive disorders (PNDs) are considered the most common postoperative complication in geriatric patients. However, its pathogenesis is not fully understood. Surgery-triggered neuroinflammation is a major contributor to the development of PNDs. Neuroinflammation can influence N-methyl-D-aspartate receptor (NMDAR) expression or function which is closely associated with cognition. We, therefore, hypothesized that the persistent changes in NMDAR expression or function induced by transient neuroinflammation after surgery were involved in the development of PNDs. METHODS: Eighteen-month-old male Sprague–Dawley rats were subjected to abdominal surgery with sevoflurane anesthesia to establish the PNDs animal model. Then, we determined the transient neuroinflammation by detecting the protein levels of proinflammatory cytokines and microglia activation using ELISA, western blot, immunohistochemistry, and microglial morphological analysis from postoperative days 1–20. Persistent changes in NMDAR expression were determined by detecting the protein levels of NMDAR subunits from postoperative days 1–59. Subsequently, the dysfunction of synaptic NMDAR was evaluated by detecting the structural plasticity of dendritic spine using Golgi staining. Pull-down assay and western blot were used to detect the protein levels of Rac1-GTP, phosphor-cofilin, and Arp3, which contribute to the regulation of the structural plasticity of dendritic spine. Finally, glycyrrhizin, an anti-inflammatory agent, was administered to further explore the role of synaptic NMDAR dysfunction induced by transient neuroinflammation in the neuropathogenesis of PNDs. RESULTS: We showed that transient neuroinflammation induced by surgery caused sustained downregulation of synaptic NR2A and NR2B subunits in the dorsal hippocampus and led to a selective long-term spatial memory deficit. Meanwhile, the detrimental effect of neuroinflammation on the function of synaptic NMDARs was shown by the impaired structural plasticity of dendritic spines and decreased activity of the Rac1 signaling pathways during learning. Furthermore, anti-inflammatory treatment reversed the downregulation and hypofunction of synaptic NR2A and NR2B and subsequently rescued the long-term spatial memory deficit. CONCLUSIONS: Our results identify sustained synaptic NR2A and NR2B downregulation and hypofunction induced by transient neuroinflammation following surgery as important contributors to the development of PNDs in elderly rats. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02528-5. BioMed Central 2022-07-13 /pmc/articles/PMC9281167/ /pubmed/35831873 http://dx.doi.org/10.1186/s12974-022-02528-5 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Chen, Bo
Qin, Guangcheng
Xiao, Jingyu
Deng, Xiaoyuan
Lin, Aolei
Liu, Hongliang
Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor
title Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor
title_full Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor
title_fullStr Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor
title_full_unstemmed Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor
title_short Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor
title_sort transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic nmda receptor
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9281167/
https://www.ncbi.nlm.nih.gov/pubmed/35831873
http://dx.doi.org/10.1186/s12974-022-02528-5
work_keys_str_mv AT chenbo transientneuroinflammationfollowingsurgerycontributestolonglastingcognitivedeclineinelderlyratsviadysfunctionofsynapticnmdareceptor
AT qinguangcheng transientneuroinflammationfollowingsurgerycontributestolonglastingcognitivedeclineinelderlyratsviadysfunctionofsynapticnmdareceptor
AT xiaojingyu transientneuroinflammationfollowingsurgerycontributestolonglastingcognitivedeclineinelderlyratsviadysfunctionofsynapticnmdareceptor
AT dengxiaoyuan transientneuroinflammationfollowingsurgerycontributestolonglastingcognitivedeclineinelderlyratsviadysfunctionofsynapticnmdareceptor
AT linaolei transientneuroinflammationfollowingsurgerycontributestolonglastingcognitivedeclineinelderlyratsviadysfunctionofsynapticnmdareceptor
AT liuhongliang transientneuroinflammationfollowingsurgerycontributestolonglastingcognitivedeclineinelderlyratsviadysfunctionofsynapticnmdareceptor