Back to Search Start Over

NLRP3 Inflammasome-Dependent Increases in High Mobility Group Box 1 Involved in the Cognitive Dysfunction Caused by Tau-Overexpression

Authors :
Yan Zhao
Si-Wei Tan
Zhi-Zhong Huang
Fa-Bo Shan
Ping Li
Ya-Lei Ning
Shi-Yang Ye
Zi-Ai Zhao
Hao Du
Ren-Ping Xiong
Nan Yang
Yan Peng
Xing Chen
Yuan-Guo Zhou
Source :
Frontiers in Aging Neuroscience, Vol 13 (2021)
Publication Year :
2021
Publisher :
Frontiers Media S.A., 2021.

Abstract

Tau hyperphosphorylation is a characteristic alteration present in a range of neurological conditions, such as traumatic brain injury (TBI) and neurodegenerative diseases. Treatments targeting high-mobility group box protein 1 (HMGB1) induce neuroprotective effects in these neuropathologic conditions. However, little is known about the interactions between hyperphosphorylated tau and HMGB1 in neuroinflammation. We established a model of TBI with controlled cortical impacts (CCIs) and a tau hyperphosphorylation model by injecting the virus encoding human P301S tau in mice, and immunofluorescence, western blotting analysis, and behavioral tests were performed to clarify the interaction between phosphorylated tau (p-tau) and HMGB1 levels. We demonstrated that p-tau and HMGB1 were elevated in the spatial memory-related brain regions in mice with TBI and tau-overexpression. Animals with tau-overexpression also had significantly increased nucleotide-binding oligomerization domain-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome activation, which manifested as increases in apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), activating caspase-1 and interleukin 1 beta (IL-1β) levels. In addition, NLRP3–/– mice and the HMGB1 inhibitor, glycyrrhizin, were used to explore therapeutic strategies for diseases with p-tau overexpression. Compared with wild-type (WT) mice with tau-overexpression, downregulation of p-tau and HMGB1 was observed in NLRP3–/– mice, indicating that HMGB1 alterations were NLRP3-dependent. Moreover, treatment with glycyrrhizin at a late stage markedly reduced p-tau levels and improved performance in the Y- and T-mazes and the ability of tau-overexpressing mice to build nests, which revealed improvements in spatial memory and advanced hippocampal function. The findings identified that p-tau has a triggering role in the modulation of neuroinflammation and spatial memory in an NLRP3-dependent manner, and suggest that treatment with HMGB1 inhibitors may be a better therapeutic strategy for tauopathies.

Details

Language :
English
ISSN :
16634365
Volume :
13
Database :
Directory of Open Access Journals
Journal :
Frontiers in Aging Neuroscience
Publication Type :
Academic Journal
Accession number :
edsdoj.22b4664e0aef4d3ba835fa6b67a45170
Document Type :
article
Full Text :
https://doi.org/10.3389/fnagi.2021.721474