Inhibiting NLRP3 inflammasome signaling pathway promotes neurological recovery following hypoxic-ischemic brain damage by increasing p97-mediated surface GluA1-containing AMPA receptors

© 2023. BioMed Central Ltd., part of Springer Nature..

BACKGROUND: The nucleotide-binding oligomeric domain (NOD)-like receptor protein 3 (NLRP3) inflammasome is believed to be a key mediator of neuroinflammation and subsequent secondary brain injury induced by ischemic stroke. However, the role and underlying mechanism of the NLRP3 inflammasome in neonates with hypoxic-ischemic encephalopathy (HIE) are still unclear.

METHODS: The protein expressions of the NLRP3 inflammasome including NLRP3, cysteinyl aspartate specific proteinase-1 (caspase-1) and interleukin-1β (IL-1β), the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionicacid receptor (AMPAR) subunit, and the ATPase valosin-containing protein (VCP/p97), were determined by Western blotting. The interaction between p97 and AMPA glutamate receptor 1 (GluA1) was determined by co-immunoprecipitation. The histopathological level of hypoxic-ischemic brain damage (HIBD) was determined by triphenyltetrazolium chloride (TTC) staining. Polymerase chain reaction (PCR) and Western blotting were used to confirm the genotype of the knockout mice. Motor functions, including myodynamia and coordination, were evaluated by using grasping and rotarod tests. Hippocampus-dependent spatial cognitive function was measured by using the Morris-water maze (MWM).

RESULTS: We reported that the NLRP3 inflammasome signaling pathway, such as NLRP3, caspase-1 and IL-1β, was activated in rats with HIBD and oxygen-glucose deprivation (OGD)-treated cultured primary neurons. Further studies showed that the protein level of the AMPAR GluA1 subunit on the hippocampal postsynaptic membrane was significantly decreased in rats with HIBD, and it could be restored to control levels after treatment with the specific caspase-1 inhibitor AC-YVAD-CMK. Similarly, in vitro studies showed that OGD reduced GluA1 protein levels on the plasma membrane in cultured primary neurons, whereas AC-YVAD-CMK treatment restored this reduction. Importantly, we showed that OGD treatment obviously enhanced the interaction between p97 and GluA1, while AC-YVAD-CMK treatment promoted the dissociation of p97 from the GluA1 complex and consequently facilitated the localization of GluA1 on the plasma membrane of cultured primary neurons. Finally, we reported that the deficits in motor function, learning and memory in animals with HIBD, were ameliorated by pharmacological intervention or genetic ablation of caspase-1.

CONCLUSION: Inhibiting the NLRP3 inflammasome signaling pathway promotes neurological recovery in animals with HIBD by increasing p97-mediated surface GluA1 expression, thereby providing new insight into HIE therapy.

Medienart:

E-Artikel

Erscheinungsjahr:

2023

Erschienen:

2023

Enthalten in:

Zur Gesamtaufnahme - volume:21

Enthalten in:

Journal of translational medicine - 21(2023), 1 vom: 24. Aug., Seite 567

Sprache:

Englisch

Beteiligte Personen:

Chen, Yuxin [VerfasserIn]
Li, Xiaohuan [VerfasserIn]
Xiong, Qian [VerfasserIn]
Du, Yehong [VerfasserIn]
Luo, Man [VerfasserIn]
Yi, Lilin [VerfasserIn]
Pang, Yayan [VerfasserIn]
Shi, Xiuyu [VerfasserIn]
Wang, Yu Tian [VerfasserIn]
Dong, Zhifang [VerfasserIn]

Links:

Volltext

Themen:

Caspase 1
Caspase-1
EC 3.4.22.36
GluA1
Hypoxic-ischemic brain damage
Inflammasomes
Journal Article
NLR Family, Pyrin Domain-Containing 3 Protein
NLRP3
P97
Receptors, AMPA
Research Support, Non-U.S. Gov't

Anmerkungen:

Date Completed 28.08.2023

Date Revised 18.11.2023

published: Electronic

Citation Status MEDLINE

doi:

10.1186/s12967-023-04452-5

funding:

Förderinstitution / Projekttitel:

PPN (Katalog-ID):

NLM361190948