4.7 Article

Genetic deletion of soluble epoxide hydrolase delays the progression of Alzheimer's disease

Journal

JOURNAL OF NEUROINFLAMMATION
Volume 16, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s12974-019-1635-9

Keywords

Soluble epoxide hydrolase; Alzheimer's disease; Inflammation; Astrocyte

Funding

  1. Ministry of Science and Technology of Taiwan [104-2320-B-010-041-MY3, 105-2320-B-010-036, 1052811-B-010-022, 106-2320-B-002-057-MY3, 106-2320-B-002-056, 106-2811-B-002-146, 108-2320-B-002-032-MY3]
  2. Yen Tjing Ling Medical Foundation [CI-105-15]
  3. Cheng Hsin General Hospital [CY10606, CY10708]

Ask authors/readers for more resources

Background: Soluble 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. Methods: To 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)-1 beta, IL-4, IL-6, and IL-10 and the activities of NF-kappa B 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. Results: We 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 A beta 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-1 beta, 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. Conclusion: Our 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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available