Reducing Nav1.6 expression attenuates the pathogenesis of Alzheimer's disease by suppressing BACE1 transcription

Aberrant increases in neuronal network excitability may contribute to cognitive deficits in Alzheimer's disease (AD). However, the mechanisms underlying hyperexcitability of neurons are not fully understood. Voltage‐gated sodium channels (VGSC or Nav), which are involved in the formation of exc...

Full description

Saved in:
Bibliographic Details
Published inAging cell Vol. 21; no. 5; pp. e13593 - n/a
Main Authors Yuan, De‐Juan, Yang, Guang, Wu, Wei, Li, Qi‐Fa, Xu, De‐en, Ntim, Michael, Jiang, Chun‐Yan, Liu, Ji‐Chuan, Zhang, Yue, Wang, Ying‐Zi, Zhu, Dan‐Dan, Kundu, Supratik, Li, Ai‐Ping, Xiao, Zhi‐Cheng, Ma, Quan‐Hong, Li, Shao
Format Journal Article
LanguageEnglish
Published England John Wiley & Sons, Inc 01.05.2022
John Wiley and Sons Inc
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Aberrant increases in neuronal network excitability may contribute to cognitive deficits in Alzheimer's disease (AD). However, the mechanisms underlying hyperexcitability of neurons are not fully understood. Voltage‐gated sodium channels (VGSC or Nav), which are involved in the formation of excitable cell's action potential and can directly influence the excitability of neural networks, have been implicated in AD‐related abnormal neuronal hyperactivity and higher incidence of spontaneous non‐convulsive seizures. Here, we have shown that the reduction of VGSC α‐subunit Nav1.6 (by injecting adeno‐associated virus (AAV) with short hairpin RNA (shRNA) into the hippocampus) rescues cognitive impairments and attenuates synaptic deficits in APP/PS1 transgenic mice. Concurrently, amyloid plaques in the hippocampus and levels of soluble Aβ are significantly reduced. Interfering with Nav1.6 reduces the transcription level of β‐site APP‐cleaving enzyme 1 (BACE1), which is Aβ‐dependent. In the presence of Aβ oligomers, knockdown of Nav1.6 reduces intracellular calcium overload by suppressing reverse sodium–calcium exchange channel, consequently increasing inactive NFAT1 (the nuclear factor of activated T cells) levels and thus reducing BACE1 transcription. This mechanism leads to a reduction in the levels of Aβ in APP/PS1 transgenic mice, alleviates synaptic loss, improves learning and memory disorders in APP/PS1 mice after downregulating Nav1.6 in the hippocampus. Our study offers a new potential therapeutic strategy to counteract hippocampal hyperexcitability and subsequently rescue cognitive deficits in AD by selective blockade of Nav1.6 overexpression and/or hyperactivity. In the presence of Aβ oligomers, knockdown of Nav1.6 reduces intracellular calcium overload by suppressing reverse sodium‐calcium exchange channel, consequently increasing inactive NFAT1 (the nuclear factor of activated T cells) levels and thus reducing BACE1 transcription. This mechanism leads to a reduction in the levels of Aβ in APP/PS1 transgenic mice, alleviates synaptic loss, and improves learning and memory disorders in APP/PS1 mice after down‐regulating Nav1.6 in the hippocampus.
Bibliography:De‐Juan Yuan and Guang Yang contributed equally to this work.
Funding information
This work was supported with grants from the National Natural Sciences Foundation of China (81571061, 82101275, 82101661, 81870897, and 81901296), Liaoning Provincial Key R&D Program (2019020048‐JH2/103), Liaoning Revitalization Talents Program (XLYC1902044), Natural Science Foundation of Jiangsu Province (BK20181436), Guangdong Key Project in “Development of new tools for diagnosis and treatment of Autism” (2018B030335001) and Team Innovation Funding Program of the Second Affiliated Hospital of Soochow University (XKTJ‐TD20203)
ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:1474-9718
1474-9726
DOI:10.1111/acel.13593