4.7 Article

Alzheimer's disease pathology is attenuated in a CD38-deficient mouse model

期刊

ANNALS OF NEUROLOGY
卷 78, 期 1, 页码 88-103

出版社

WILEY
DOI: 10.1002/ana.24425

关键词

-

资金

  1. US-Israel Binational Science Foundation [2007061, 2011054]
  2. Adams Super-Center for Brain Research at Tel-Aviv University
  3. National Network of Excellence in Neuroscience
  4. NIH (NIAID) [R01 AI063399]
  5. EU FP7 project LipiDiDiet [211696]
  6. HOMFORexzellent (Saarland University research grants)

向作者/读者索取更多资源

ObjectiveAlzheimer's disease (AD)-associated dementia is due to tissue damage caused by amyloid (A) deposition within the brain and by accompanying neuroinflammation. The nicotinamide adenine dinucleotide (NAD) glycohydrolase CD38, which is expressed by neurons, astrocytes, and microglial cells, regulates inflammatory and repair processes in the brain and other tissues by degrading NAD and repressing the activity of other NAD-consuming enzymes and by producing NAD-derived metabolites that regulate calcium signaling and migration of inflammatory cells. Given the role of CD38 in neuroinflammation and repair, we examined the effect of CD38 deletion on AD pathology. MethodsWe crossed APPswePS1E9 (APP.PS) mice with Cd38(-/-) mice to generate AD-prone CD38-deficient animals (APP.PS.Cd38(-/-)) and examined AD-related phenotypes in both groups. ResultsAPP.PS.Cd38(-/-) mice exhibited significant reductions in A plaque load and soluble A levels compared to APP.PS mice, and this correlated with improved spatial learning. Although CD38 deficiency resulted in decreased microglia/macrophage (MM) accumulation, the transcription profile of the Cd38(-/-) and Cd38(+/+) MM was similar, suggesting that the decreased A burden in APP.PS.Cd38(-/-) mice was not due to alterations in MM activation/function. Instead, APP.PS.Cd38(-/-) neuronal cultures secreted less A and this reduction was mimicked when APP.PS neuronal cultures were treated with inhibitors that blocked CD38 enzyme activity or the signaling pathways controlled by CD38-derived metabolites. Furthermore, - and -secretase activity was decreased in APP.PS.Cd38(-/-) mice, which correlated with decreased A production. InterpretationCD38 regulates AD pathology in the APP.PS model of AD, suggesting that CD38 may be a novel target for AD treatment. Ann Neurol 2015;78:88-103

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据