Loss of Endothelial Nitric Oxide Synthase Promotes p25 Generation and Tau Phosphorylation in a Murine Model of Alzheimer’s Disease

Author:

Austin Susan A.1,Katusic Zvonimir S.1

Affiliation:

1. From the Departments of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN.

Abstract

Rationale: Alzheimer’s disease has an unknown pathogenesis; however, cardiovascular risk factors are associated with a higher incidence of Alzheimer’s disease. A defining feature of endothelial dysfunction induced by cardiovascular risk factors is reduced bioavailable endothelial nitric oxide (NO). We previously demonstrated that endothelial NO acts as an important signaling molecule in neuronal tissue. Objective: We sought to determine the relationship between the loss of endothelial NO synthase (eNOS) and tau phosphorylation in neuronal tissue. Methods and Results: We used eNOS knockout ( −/− ) mice as well as an Alzheimer’s disease mouse model, amyloid precursor protein (APP)/PSEN1dE9 +/− (PS1) that lacked eNOS (APP/PS1/eNOS −/− ) to examine expression of tau kinases and tau phosphorylation. Brain tissue from eNOS −/− mice had statistically higher ratios of p25/p35, indicative of increased cyclin-dependent kinase 5 activity as compared with wild-type (n=8, P <0.05). However, tau phosphorylation was unchanged in eNOS −/− mice ( P >0.05). Next, we determined the role of NO in tau pathology in APP/PS1/eNOS −/− . These mice had significantly higher levels of p25, a higher p25/p35 ratio (n=12–14; P <0.05), and significantly higher cyclin-dependent kinase 5 activity (n=4; P <0.001). Importantly, APP/PS1/eNOS −/− mice also had significantly increased tau phosphorylation (n=4–6; P <0.05). No other changes in amyloid pathology, antioxidant pathways, or neuroinflammation were observed in APP/PS1/eNOS −/− mice as compared with APP/PS1 mice. Conclusions: Our data suggests that loss of endothelial NO plays an important role in the generation of p25 and resulting tau phosphorylation in neuronal tissue. These findings provide important new insights into the molecular mechanisms linking endothelial dysfunction with the pathogenesis of Alzheimer’s disease.

Publisher

Ovid Technologies (Wolters Kluwer Health)

Subject

Cardiology and Cardiovascular Medicine,Physiology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3