%0 Journal Article %J Proc Natl Acad Sci U S A %D 2011 %T Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. %A Jin, Ming %A Shepardson, Nina %A Yang, Ting %A Chen, Gang %A Walsh, Dominic %A Selkoe, Dennis J %K Alzheimer Disease %K Amyloid beta-Peptides %K Animals %K Blotting, Western %K Chromatography, Gel %K Dimerization %K Genetic Vectors %K Hippocampus %K Humans %K Immunohistochemistry %K Immunoprecipitation %K In Situ Nick-End Labeling %K Lentivirus %K Microscopy, Confocal %K Microtubules %K Neurites %K Phosphorylation %K Rats %K Rats, Sprague-Dawley %K tau Proteins %X

Alzheimer disease is a major cause of cognitive failure, and a pathogenically related but more subtle process accounts for many cases of mild memory symptoms in older humans. Insoluble fibrillar plaques of amyloid β-proteins (Aβ) and neurofibrillary deposits of hyperphosphorylated tau proteins are the diagnostic lesions of AD, but their temporal mechanistic relationship has long been debated. The recent recognition that small, diffusible oligomers may be the principal bioactive form of Aβ raises the key question of whether these are sufficient to initiate cytoskeletal change and neurite degeneration. A few studies have examined the effects of oligomers of synthetic Aβ peptides of one defined length at supraphysiological concentrations, but the existence of such assemblies in the AD brain is not established. Here, we isolated Aβ dimers, the most abundant form of soluble oligomer detectable in the human brain, from the cortices of typical AD subjects and found that at subnanomolar concentrations, they first induced hyperphosphorylation of tau at AD-relevant epitopes in hippocampal neurons and then disrupted the microtubule cytoskeleton and caused neuritic degeneration, all in the absence of amyloid fibrils. Application of pure, synthetic dimers confirmed the effects of the natural AD dimers, although the former were far less potent. Knocking down endogenous tau fully prevented the neuritic changes, whereas overexpressing human tau accelerated them. Coadministering Aβ N-terminal antibodies neutralized the cytoskeletal disruption. We conclude that natural dimers isolated from the AD brain are sufficient to potently induce AD-type tau phosphorylation and then neuritic dystrophy, but passive immunotherapy mitigates this.

%B Proc Natl Acad Sci U S A %V 108 %P 5819-24 %8 2011 Apr 5 %G eng %N 14 %1 http://www.ncbi.nlm.nih.gov/pubmed/21421841?dopt=Abstract %R 10.1073/pnas.1017033108