4.6 Review

Recent Advances in the Modeling of Alzheimer's Disease

期刊

FRONTIERS IN NEUROSCIENCE
卷 16, 期 -, 页码 -

出版社

FRONTIERS MEDIA SA
DOI: 10.3389/fnins.2022.807473

关键词

Alzheimer's disease; amyloid; beta; amyloidosis; tau propagation; somatostatin; mouse model; non-human primate (NHP)

向作者/读者索取更多资源

This article introduces transgenic mouse models of Alzheimer's disease (AD) and discusses their limitations and applications. The first generation models have artificial phenotypes, while the second generation models have specific strengths and weaknesses. The third generation models exhibit more pathology and are suitable for preclinical studies. The article also provides a technical guide for using App knock-in mice in AD research and discusses the latest advances in using human tau knock-in mice and non-human primate models of AD.
Since 1995, more than 100 transgenic (Tg) mouse models of Alzheimer's disease (AD) have been generated in which mutant amyloid precursor protein (APP) or APP/presenilin 1 (PS1) cDNA is overexpressed (1st generation models). Although many of these models successfully recapitulate major pathological hallmarks of the disease such as amyloid beta peptide (A beta) deposition and neuroinflammation, they have suffered from artificial phenotypes in the form of overproduced or mislocalized APP/PS1 and their functional fragments, as well as calpastatin deficiency-induced early lethality, calpain activation, neuronal cell death without tau pathology, endoplasmic reticulum stresses, and inflammasome involvement. Such artifacts bring two important uncertainties into play, these being (1) why the artifacts arise, and (2) how they affect the interpretation of experimental results. In addition, destruction of endogenous gene loci in some Tg lines by transgenes has been reported. To overcome these concerns, single App knock-in mouse models harboring the Swedish and Beyreuther/Iberian mutations with or without the Arctic mutation (App(NL-G-F) and App(NL-F) mice) were developed (2nd generation models). While these models are interesting given that they exhibit A beta pathology, neuroinflammation, and cognitive impairment in an age-dependent manner, the model with the Artic mutation, which exhibits an extensive pathology as early as 6 months of age, is not suitable for investigating A beta metabolism and clearance because the A beta in this model is resistant to proteolytic degradation and is therefore prone to aggregation. Moreover, it cannot be used for preclinical immunotherapy studies owing to the discrete affinity it shows for anti-A beta antibodies. The weakness of the latter model (without the Arctic mutation) is that the pathology may require up to 18 months before it becomes sufficiently apparent for experimental investigation. Nevertheless, this model was successfully applied to modulating A beta pathology by genome editing, to revealing the differential roles of neprilysin and insulin-degrading enzyme in A beta metabolism, and to identifying somatostatin receptor subtypes involved in A beta degradation by neprilysin. In addition to discussing these issues, we also provide here a technical guide for the application of App knock-in mice to AD research. Subsequently, a new double knock-in line carrying the App(NL-F) and Psen1(P117L/WT) mutations was generated, the pathogenic effect of which was found to be synergistic. A characteristic of this 3rd generation model is that it exhibits more cored plaque pathology and neuroinflammation than the App(NL-G-F) line, and thus is more suitable for preclinical studies of disease-modifying medications targeting A beta. Furthermore, a derivative App(G-F) line devoid of Swedish mutations which can be utilized for preclinical studies of beta-secretase modifier(s) was recently created. In addition, we introduce a new model of cerebral amyloid angiopathy that may be useful for analyzing amyloid-related imaging abnormalities that can be caused by anti-A beta immunotherapy. Use of the App knock-in mice also led to identification of the alpha-endosulfine-K-ATP channel pathway as components of the somatostatin-evoked physiological mechanisms that reduce A beta deposition via the activation of neprilysin. Such advances have provided new insights for the prevention and treatment of preclinical AD. Because tau pathology plays an essential role in AD pathogenesis, knock-in mice with human tau wherein the entire murine Mapt gene has been humanized were generated. Using these mice, the carboxy-terminal PDZ ligand of neuronal nitric oxide synthase (CAPON) was discovered as a mediator linking tau pathology to neurodegeneration and showed that tau humanization promoted pathological tau propagation. Finally, we describe and discuss the current status of mutant human tau knock-in mice and a non-human primate model of AD that we have successfully created.

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

4.6
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