• Medientyp: E-Artikel
  • Titel: Genetic deletion of soluble epoxide hydrolase delays the progression of Alzheimer’s disease
  • Beteiligte: Lee, Hsueh-Te; Lee, Kuan-I; Chen, Chia-Hui; Lee, Tzong-Shyuan
  • Erschienen: Springer Science and Business Media LLC, 2019
  • Erschienen in: Journal of Neuroinflammation, 16 (2019) 1
  • Sprache: Englisch
  • DOI: 10.1186/s12974-019-1635-9
  • ISSN: 1742-2094
  • Entstehung:
  • Anmerkungen:
  • Beschreibung: AbstractBackgroundSoluble 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/PS1Tg) andAPP/PS1Tg/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 ofAPP/PS1Tg mice. Genetic ablation ofsEHinAPP/PS1Tg mice delayed the progression of AD as evidenced by the alleviation in behavior outcomes and Aβ plaque deposition. In addition, loss of the function ofsEHinAPP/PS1Tg 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 inAPP/PS1Tg/sEH−/−mice compared withAPP/PS1Tg 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.
  • Zugangsstatus: Freier Zugang