唐氏综合征阿尔茨海默病的细胞模型研究

Q4 Neuroscience Neuronal signaling Pub Date : 2022-03-22 DOI:10.1042/NS20210054
Yixing Wu, N. West, A. Bhattacharyya, F. Wiseman
{"title":"唐氏综合征阿尔茨海默病的细胞模型研究","authors":"Yixing Wu, N. West, A. Bhattacharyya, F. Wiseman","doi":"10.1042/NS20210054","DOIUrl":null,"url":null,"abstract":"Abstract Down syndrome (DS) is the most common chromosomal abnormality and leads to intellectual disability, increased risk of cardiac defects, and an altered immune response. Individuals with DS have an extra full or partial copy of chromosome 21 (trisomy 21) and are more likely to develop early-onset Alzheimer’s disease (AD) than the general population. Changes in expression of human chromosome 21 (Hsa21)-encoded genes, such as amyloid precursor protein (APP), play an important role in the pathogenesis of AD in DS (DS-AD). However, the mechanisms of DS-AD remain poorly understood. To date, several mouse models with an extra copy of genes syntenic to Hsa21 have been developed to characterise DS-AD-related phenotypes. Nonetheless, due to genetic and physiological differences between mouse and human, mouse models cannot faithfully recapitulate all features of DS-AD. Cells differentiated from human-induced pluripotent stem cells (iPSCs), isolated from individuals with genetic diseases, can be used to model disease-related cellular and molecular pathologies, including DS. In this review, we will discuss the limitations of mouse models of DS and how these can be addressed using recent advancements in modelling DS using human iPSCs and iPSC-mouse chimeras, and potential applications of iPSCs in preclinical studies for DS-AD.","PeriodicalId":74287,"journal":{"name":"Neuronal signaling","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2022-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"3","resultStr":"{\"title\":\"Cell models for Down syndrome-Alzheimer’s disease research\",\"authors\":\"Yixing Wu, N. West, A. Bhattacharyya, F. Wiseman\",\"doi\":\"10.1042/NS20210054\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Abstract Down syndrome (DS) is the most common chromosomal abnormality and leads to intellectual disability, increased risk of cardiac defects, and an altered immune response. Individuals with DS have an extra full or partial copy of chromosome 21 (trisomy 21) and are more likely to develop early-onset Alzheimer’s disease (AD) than the general population. Changes in expression of human chromosome 21 (Hsa21)-encoded genes, such as amyloid precursor protein (APP), play an important role in the pathogenesis of AD in DS (DS-AD). However, the mechanisms of DS-AD remain poorly understood. To date, several mouse models with an extra copy of genes syntenic to Hsa21 have been developed to characterise DS-AD-related phenotypes. Nonetheless, due to genetic and physiological differences between mouse and human, mouse models cannot faithfully recapitulate all features of DS-AD. Cells differentiated from human-induced pluripotent stem cells (iPSCs), isolated from individuals with genetic diseases, can be used to model disease-related cellular and molecular pathologies, including DS. In this review, we will discuss the limitations of mouse models of DS and how these can be addressed using recent advancements in modelling DS using human iPSCs and iPSC-mouse chimeras, and potential applications of iPSCs in preclinical studies for DS-AD.\",\"PeriodicalId\":74287,\"journal\":{\"name\":\"Neuronal signaling\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2022-03-22\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"3\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Neuronal signaling\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1042/NS20210054\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q4\",\"JCRName\":\"Neuroscience\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Neuronal signaling","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1042/NS20210054","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q4","JCRName":"Neuroscience","Score":null,"Total":0}
引用次数: 3

摘要

唐氏综合征(DS)是最常见的染色体异常,可导致智力残疾、心脏缺陷风险增加和免疫反应改变。患有退行性痴呆的个体有一个额外的21号染色体(21三体)的完整或部分副本,比一般人群更容易患上早发性阿尔茨海默病(AD)。人类21号染色体(Hsa21)编码基因(如淀粉样前体蛋白(APP))的表达变化在DS-AD的发病机制中起重要作用。然而,对DS-AD的机制仍然知之甚少。迄今为止,已经开发了几种具有与Hsa21合成基因的额外拷贝的小鼠模型来表征ds - ad相关表型。然而,由于小鼠和人之间的遗传和生理差异,小鼠模型不能忠实地再现DS-AD的所有特征。从患有遗传性疾病的个体中分离出来的从人诱导多能干细胞(iPSCs)分化的细胞可用于模拟疾病相关的细胞和分子病理,包括退行性椎体滑移。在这篇综述中,我们将讨论DS小鼠模型的局限性,以及如何利用人类iPSCs和ipsc -小鼠嵌合体建立DS模型的最新进展来解决这些问题,以及iPSCs在DS- ad临床前研究中的潜在应用。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Cell models for Down syndrome-Alzheimer’s disease research
Abstract Down syndrome (DS) is the most common chromosomal abnormality and leads to intellectual disability, increased risk of cardiac defects, and an altered immune response. Individuals with DS have an extra full or partial copy of chromosome 21 (trisomy 21) and are more likely to develop early-onset Alzheimer’s disease (AD) than the general population. Changes in expression of human chromosome 21 (Hsa21)-encoded genes, such as amyloid precursor protein (APP), play an important role in the pathogenesis of AD in DS (DS-AD). However, the mechanisms of DS-AD remain poorly understood. To date, several mouse models with an extra copy of genes syntenic to Hsa21 have been developed to characterise DS-AD-related phenotypes. Nonetheless, due to genetic and physiological differences between mouse and human, mouse models cannot faithfully recapitulate all features of DS-AD. Cells differentiated from human-induced pluripotent stem cells (iPSCs), isolated from individuals with genetic diseases, can be used to model disease-related cellular and molecular pathologies, including DS. In this review, we will discuss the limitations of mouse models of DS and how these can be addressed using recent advancements in modelling DS using human iPSCs and iPSC-mouse chimeras, and potential applications of iPSCs in preclinical studies for DS-AD.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
4.60
自引率
0.00%
发文量
0
审稿时长
14 weeks
期刊最新文献
Neural mechanisms of dopamine function in learning and memory in Caenorhabditis elegans Cytokine activity in Parkinson's disease. Modelling Alzheimer’s disease in a Dish – Dissecting Amyloid-β Metabolism in Human Neurons Inflammation and emotion regulation: a narrative review of evidence and mechanisms in emotion dysregulation disorders Inhibition of insulin-degrading enzyme in human neurons promotes amyloid-β deposition.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1