Abstract

BackgroundSoluble epoxide hydrolase (sEH) is a bifunctional enzyme with COOH-terminal hydrolase and NH2-terminal lipid phosphatase activities. It is expressed in various cell types in the brain and is involved in the pathogenesis of inflammatory and neurodegenerative diseases. Alzheimer’s disease (AD) is a progressive neuroinflammatory and neurodegenerative disease. However, the pathological significance of sEH and underlying molecular mechanism in AD remain unclear.MethodsTo examine the role of sEH in pathogenesis of AD, we used wild-type (WT) mice, soluble epoxide hydrolase deficient (sEH−/−) and two mouse models of AD, including amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic (APP/PS1 Tg) and APP/PS1 Tg/sEH−/− mice. Western blotting analysis and immunohistochemistry assay were performed to evaluate the protein expression. Locomotion, nesting building ability, Y-maze, and Morris water maze tests were conducted to study mouse behavior. The levels of interleukin (IL)-1β, IL-4, IL-6, and IL-10 and the activities of NF-κB and nuclear factor of activated T cells (NFAT) were measured by commercial assay kits. The quantitative protein level profiling in the brain lysate was analyzed using LC-MS/MS approaches.ResultsWe demonstrated that the level of sEH was increased in the brain and predominantly appeared in hippocampal astrocytes of APP/PS1 Tg mice. Genetic ablation of sEH in APP/PS1 Tg mice delayed the progression of AD as evidenced by the alleviation in behavior outcomes and Aβ plaque deposition. In addition, loss of the function of sEH in APP/PS1 Tg mice increased astrogliosis and the production of astrocyte-derived anti-inflammatory cytokines including IL-1β, IL-4, and IL-10, as well as the activity of NF-kB and NFAT. Moreover, analysis of gene ontology in the AD brain revealed that important signaling pathways and processes related to AD pathogenesis such as translational regulation, oxidative stress, cytoskeleton reorganization, and small GTPase signal transduction were altered in APP/PS1 Tg/sEH−/− mice compared with APP/PS1 Tg mice.ConclusionOur results suggest that sEH is a crucial regulator in the progression of AD and might be a potential therapeutic target for the treatment of AD.

Highlights

  • Soluble epoxide hydrolase, an enzyme with COOHterminal hydrolase (EH) and NH2-terminal lipid phosphatase (PT) activities, is expressed in several tissues, including the brain, heart, and kidney [1,2,3,4]

  • The level of Soluble epoxide hydrolase (sEH) is upregulated in hippocampal astrocytes during Alzheimer’s disease (AD) progression To explore the role of sEH in the pathogenesis of AD, we first investigated the levels of sEH in mouse brain from WT and amyloid precursor protein (APP)/presenilin 1 (PS1) Tg mice

  • Immunohistochemistry analysis revealed that sEH was primarily localized in endothelial cells (ECs) of vessels, hippocampal neurons, and astrocytes in WT or APP/PS1 Tg mice

Read more

Summary

Introduction

Soluble epoxide hydrolase (sEH), an enzyme with COOHterminal hydrolase (EH) and NH2-terminal lipid phosphatase (PT) activities, is expressed in several tissues, including the brain, heart, and kidney [1,2,3,4]. The precise role of sEH in the pathogenesis of inflammatory and brain diseases has been well defined [8, 9]. The role of sEH and its underlying mechanism in neurodegenerative diseases are not fully understood. Soluble epoxide hydrolase (sEH) is a bifunctional enzyme with COOH-terminal hydrolase and NH2terminal lipid phosphatase activities. It is expressed in various cell types in the brain and is involved in the pathogenesis of inflammatory and neurodegenerative diseases. The pathological significance of sEH and underlying molecular mechanism in AD remain unclear

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call