scholarly journals Familial Alzheimer's Disease–Linked Presenilin 1 Variants Elevate Aβ1–42/1–40 Ratio In Vitro and In Vivo

Neuron ◽  
1996 ◽  
Vol 17 (5) ◽  
pp. 1005-1013 ◽  
Author(s):  
David R. Borchelt ◽  
Gopal Thinakaran ◽  
Christopher B. Eckman ◽  
Michael K. Lee ◽  
Frances Davenport ◽  
...  
2007 ◽  
Vol 1147 ◽  
pp. 248-255 ◽  
Author(s):  
Caroline Sara Hatchett ◽  
Sue Tyler ◽  
Dawn Armstrong ◽  
David Dawbarn ◽  
Shelley Jane Allen

2020 ◽  
Vol 21 (4) ◽  
pp. 1413 ◽  
Author(s):  
Takami Tomiyama ◽  
Hiroyuki Shimada

Alzheimer’s disease is believed to begin with synaptic dysfunction caused by soluble Aβ oligomers. When this oligomer hypothesis was proposed in 2002, there was no direct evidence that Aβ oligomers actually disrupt synaptic function to cause cognitive impairment in humans. In patient brains, both soluble and insoluble Aβ species always coexist, and therefore it is difficult to determine which pathologies are caused by Aβ oligomers and which are caused by amyloid fibrils. Thus, no validity of the oligomer hypothesis was available until the Osaka mutation was discovered. This mutation, which was found in a Japanese pedigree of familial Alzheimer’s disease, is the deletion of codon 693 of APP gene, resulting in mutant Aβ lacking the 22nd glutamate. Only homozygous carriers suffer from dementia. In vitro studies revealed that this mutation has a very unique character that accelerates Aβ oligomerization but does not form amyloid fibrils. Model mice expressing this mutation demonstrated that all pathologies of Alzheimer’s disease can be induced by Aβ oligomers alone. In this review, we describe the story behind the discovery of the Osaka mutation, summarize the mutant’s phenotypes, and propose a mechanism of its recessive inheritance.


2010 ◽  
Vol 19 (3) ◽  
pp. 873-884 ◽  
Author(s):  
Estrella Gómez-Tortosa ◽  
Sagrario Barquero ◽  
Manuel Barón ◽  
Eulogio Gil-Neciga ◽  
Fernando Castellanos ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document