Neuronal heparan sulfates promote amyloid pathology by modulating brain amyloid-β clearance and aggregation in Alzheimer's disease

CC Liu, N Zhao, Y Yamaguchi, JR Cirrito… - Science translational …, 2016 - science.org
Science translational medicine, 2016science.org
Accumulation of amyloid-β (Aβ) peptide in the brain is the first critical step in the
pathogenesis of Alzheimer's disease (AD). Studies in humans suggest that Aβ clearance
from the brain is frequently impaired in late-onset AD. Aβ accumulation leads to the
formation of Aβ aggregates, which injure synapses and contribute to eventual
neurodegeneration. Cell surface heparan sulfates (HSs), expressed on all cell types
including neurons, have been implicated in several features in the pathogenesis of AD …
Accumulation of amyloid-β (Aβ) peptide in the brain is the first critical step in the pathogenesis of Alzheimer’s disease (AD). Studies in humans suggest that Aβ clearance from the brain is frequently impaired in late-onset AD. Aβ accumulation leads to the formation of Aβ aggregates, which injure synapses and contribute to eventual neurodegeneration. Cell surface heparan sulfates (HSs), expressed on all cell types including neurons, have been implicated in several features in the pathogenesis of AD including its colocalization with amyloid plaques and modulatory role in Aβ aggregation. We show that removal of neuronal HS by conditional deletion of the Ext1 gene, which encodes an essential glycosyltransferase for HS biosynthesis, in postnatal neurons of amyloid model APP/PS1 mice led to a reduction in both Aβ oligomerization and the deposition of amyloid plaques. In vivo microdialysis experiments also detected an accelerated rate of Aβ clearance in the brain interstitial fluid, suggesting that neuronal HS either inhibited or represented an inefficient pathway for Aβ clearance. We found that the amounts of various HS proteoglycans (HSPGs) were increased in postmortem human brain tissues from AD patients, suggesting that this pathway may contribute directly to amyloid pathogenesis. Our findings have implications for AD pathogenesis and provide insight into therapeutic interventions targeting Aβ-HSPG interactions.
AAAS