期刊论文详细信息
International Journal of Molecular Sciences
Human-Induced Neurons from Presenilin 1 Mutant Patients Model Aspects of Alzheimer’s Disease Pathology
John McDaid1  GraceE. Stutzmann1  ClarkA. Briggs1  Virginie Bottero1  Sarah Mustaly-Kalimi1  Sean Schrank1  RobertA. Marr1  Deanna Brinks2  Aiden Houcek3  Oded Singer4 
[1] Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd. North, Chicago, IL 60064, USA;Lake Forest College, Lake Forest, IL 60045, USA;Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot 76100, Israel;
关键词: ipsc;    alzheimer’s disease;    amyloid;    tau;    calcium;    fibroblast;    human;    il-18;    ryanodine receptor;   
DOI  :  10.3390/ijms21031030
来源: DOAJ
【 摘 要 】

Traditional approaches to studying Alzheimer’s disease (AD) using mouse models and cell lines have advanced our understanding of AD pathogenesis. However, with the growing divide between model systems and clinical therapeutic outcomes, the limitations of these approaches are increasingly apparent. Thus, to generate more clinically relevant systems that capture pathological cascades within human neurons, we generated human-induced neurons (HiNs) from AD and non-AD individuals to model cell autonomous disease properties. We selected an AD patient population expressing mutations in presenilin 1 (mPS1), which is linked to increased amyloid production, tau pathology, and calcium signaling abnormalities, among other features. While these AD components are detailed in model systems, they have yet to be collectively identified in human neurons. Thus, we conducted molecular, immune-based, electrophysiological, and calcium imaging studies to establish patterns of cellular pathology in this patient population. We found that mPS1 HiNs generate increased Aβ42 and hyperphosphorylated tau species relative to non-AD controls, and exaggerated ER calcium responses that are normalized with ryanodine receptor (RyR) negative allosteric modulators. The inflammasome product, interleukin-18 (IL-18), also increased PS1 expression. This work highlights the potential for HiNs to model AD pathology and validates their role in defining cellular pathogenesis and their utility for therapeutic screening.

【 授权许可】

Unknown   

  文献评价指标  
  下载次数:0次 浏览次数:1次