期刊论文详细信息
Molecular Brain
Transient inhibition of microsomal prostaglandin E synthase-1 after status epilepticus blunts brain inflammation and is neuroprotective
Short Report
Madison N. Sluter1  Jianxiong Jiang1  Ying Yu1  Nelufar Yasmen1  Lexiao Li1 
[1] Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 38163, Memphis, TN, USA;
关键词: Antiseizure drugs (ASDs);    Epilepsy;    Epileptogenesis;    Reactive gliosis;    Inhibitor;    Microsomal prostaglandin E synthase-1 (mPGES-1);    Neuroinflammation;    Neuroprotection;    Prostaglandin E2 (PGE);    Seizures;   
DOI  :  10.1186/s13041-023-01008-y
 received in 2022-12-15, accepted in 2023-01-19,  发布年份 2023
来源: Springer
PDF
【 摘 要 】

Status epilepticus (SE) in humans is characterized by prolonged convulsive seizures that are generalized and often difficult to control. The current antiseizure drugs (ASDs) aim to stop seizures quickly enough to prevent the SE-induced brain inflammation, injury, and long-term sequelae. However, sole reliance on acute therapies is imprudent because prompt treatment may not always be possible under certain circumstances. The pathophysiological mechanisms underlying the devastating consequences of SE are presumably associated with neuroinflammatory reactions, where prostaglandin E2 (PGE2) plays a pivotal role. As the terminal synthase for pathogenic PGE2, the microsomal prostaglandin E synthase-1 (mPGES-1) is rapidly and robustly induced by prolonged seizures. Congenital deletion of mPGES-1 in mice is neuroprotective and blunts gliosis following chemoconvulsant seizures, suggesting the feasibility of mPGES-1 as a potential antiepileptic target. Herein, we investigated the effects of a dual species mPGES-1 inhibitor in a mouse pilocarpine model of SE. Treatment with the mPGES-1 inhibitor in mice after SE that was terminated by diazepam, a fast-acting benzodiazepine, time-dependently abolished the SE-induced PGE2 within the brain. Its negligible effects on cyclooxygenases, the enzymes responsible for the initial step of PGE2 biosynthesis, validated its specificity to mPGES-1. Post-SE inhibition of mPGES-1 also blunted proinflammatory cytokines and reactive gliosis in the hippocampus and broadly prevented neuronal damage in a number of brain areas. Thus, pharmacological inhibition of mPGES-1 by small-molecule inhibitors might provide an adjunctive strategy that can be implemented hours after SE, together with first-line ASDs, to reduce SE-provoked brain inflammation and injury.

【 授权许可】

CC BY   
© The Author(s) 2023

【 预 览 】
附件列表
Files Size Format View
RO202305112631582ZK.pdf 5917KB PDF download
Fig. 1 128KB Image download
MediaObjects/12888_2022_4508_MOESM1_ESM.pdf 173KB PDF download
41116_2022_35_Article_IEq122.gif 1KB Image download
【 图 表 】

41116_2022_35_Article_IEq122.gif

Fig. 1

【 参考文献 】
  • [1]
  • [2]
  • [3]
  • [4]
  • [5]
  • [6]
  • [7]
  • [8]
  • [9]
  • [10]
  • [11]
  • [12]
  • [13]
  • [14]
  • [15]
  • [16]
  • [17]
  • [18]
  • [19]
  • [20]
  • [21]
  • [22]
  • [23]
  • [24]
  • [25]
  • [26]
  • [27]
  • [28]
  • [29]
  • [30]
  • [31]
  • [32]
  • [33]
  • [34]
  • [35]
  • [36]
  • [37]
  • [38]
  • [39]
  • [40]
  • [41]
  • [42]
  • [43]
  • [44]
  • [45]
  • [46]
  • [47]
  • [48]
  • [49]
  • [50]
  文献评价指标  
  下载次数:1次 浏览次数:2次