Isoflurane Disrupts Postsynaptic Density-95 Protein Interactions Causing Neuronal Synapse Loss and Cognitive Impairment in Juvenile Mice via Canonical NO-mediated Protein Kinase-G Signaling

ANESTHESIOLOGY(2022)

引用 1|浏览3
暂无评分
摘要
Background: Inhalational anesthetics are known to disrupt PDZ2 domain-mediated protein-protein interactions of the postsynaptic density (PSD)-95 protein. The aim of this study is to investigate the underlying mechanisms in response to early isoflurane exposure on synaptic PSD-95 PDZ2 domain disruption that altered spine densities and cognitive function. The authors hypothesized that activation of protein kinase-G by the components of nitric oxide (NO) signaling pathway constitutes a mechanism that prevents loss of early dendritic spines and synapse in neurons and cognitive impairment in mice in response to disruption of PDZ2 domain of the PSD-95 protein. Methods: Postnatal day 7 mice were exposed to 1.5% isoflurane for 4 h or injected with 8 mg/kg active PSD-95 wild-type PDZ2 peptide or soluble guanylyl cyclase activator YC-1 along with their respective controls. Primary neurons at 7 days in vitro were exposed to isoflurane or PSD-95 wild-type PDZ2 peptide for 4 h. Coimmunoprecipitation, spine density, synapses, cyclic guanosine monophosphate-dependent protein kinase activity, and novel object recognition memory were assessed. Results: Exposure of isoflurane or PSD-95 wild-type PDZ2 peptide relative to controls causes the following. First, there is a decrease in PSD-95 coimmunoprecipitate relative to N-methyl-d-aspartate receptor subunits NR2A and NR2B precipitate (mean +/- SD [in percentage of control]: isoflurane, 54.73 +/- 16.52, P = 0.001; and PSD-95 wild-type PDZ2 peptide, 51.32 +/- 12.93, P = 0.001). Second, there is a loss in spine density (mean +/- SD [spine density per 10 mu m]: control, 5.28 +/- 0.56 vs. isoflurane, 2.23 +/- 0.67, P < 0.0001; and PSD-95 mutant PDZ2 peptide, 4.74 +/- 0.94 vs. PSD-95 wild-type PDZ2 peptide, 1.47 +/- 0.87, P < 0.001) and a decrease in synaptic puncta (mean +/- SD [in percentage of control]: isoflurane, 41.1 +/- 14.38, P = 0.001; and PSD-95 wild-type PDZ2 peptide, 50.49 +/- 14.31, P < 0.001). NO donor or cyclic guanosine monophosphate analog prevents the spines and synapse loss and decline in the cyclic guanosine monophosphate-dependent protein kinase activity, but this prevention was blocked by soluble guanylyl cyclase or protein kinase-G inhibitors in primary neurons. Third, there were deficits in object recognition at 5 weeks (mean +/- SD [recognition index]: male, control, 64.08 +/- 10.57 vs. isoflurane, 48.49 +/- 13.41, P = 0.001, n = 60; and female, control, 67.13 +/- 11.17 vs. isoflurane, 53.76 +/- 6.64, P = 0.003, n = 58). Isoflurane-induced impairment in recognition memory was preventable by the introduction of YC-1. Conclusions: Activation of soluble guanylyl cyclase or protein kinase-G prevents isoflurane or PSD-95 wild-type PDZ2 peptide-induced loss of dendritic spines and synapse. Prevention of recognition memory with YC-1, a NO-independent activator of guanylyl cyclase, supports a role for the soluble guanylyl cyclase mediated protein kinase-G signaling in countering the effects of isoflurane-induced cognitive impairment.
更多
查看译文
关键词
cognitive impairment,mice,signaling,no-mediated
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要