2015
DOI: 10.1016/j.celrep.2015.07.044
|View full text |Cite
|
Sign up to set email alerts
|

Amyloid-β Oligomers May Impair SNARE-Mediated Exocytosis by Direct Binding to Syntaxin 1a

Abstract: SUMMARY Alzheimer’s disease (AD) is closely associated with synaptic dysfunction and thus, current treatments often aim to stimulate neurotransmission to improve cognitive impairment. While the formation of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is essential for synaptic transmission, the correlation between SNAREs and AD neuropathology is unknown. Here we report that intracellular amyloid-β (Aβ) oligomers directly inhibit SNARE-mediated exocytosis by impairin… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

2
50
0

Year Published

2017
2017
2023
2023

Publication Types

Select...
6
2
1

Relationship

0
9

Authors

Journals

citations
Cited by 58 publications
(52 citation statements)
references
References 52 publications
(66 reference statements)
2
50
0
Order By: Relevance
“…Given the increased amount of higher order Aβ42 oligomers in rpAD[18], it is plausible to suggest that this process occurs to a greater extent in rpAD. This hypothesis is supported by a recent study that showed that Aβ oligomers bind to syntaxin 1A, which is an example key presynaptic protein that is up-regulated in rpAD plaques, and this binding prevents synaptic vesicle release and leads to synaptic impairment[87]. …”
Section: Discussionmentioning
confidence: 75%
“…Given the increased amount of higher order Aβ42 oligomers in rpAD[18], it is plausible to suggest that this process occurs to a greater extent in rpAD. This hypothesis is supported by a recent study that showed that Aβ oligomers bind to syntaxin 1A, which is an example key presynaptic protein that is up-regulated in rpAD plaques, and this binding prevents synaptic vesicle release and leads to synaptic impairment[87]. …”
Section: Discussionmentioning
confidence: 75%
“…Intracellular Aβ is enriched in both AD human brains and AD mouse models in association with dystrophic neurites and abnormal synaptic morphology (LaFerla et al, 2007; Spires-Jones and Hyman, 2014). Intracellular Aβ was proposed to induce presynaptic dysfunction, which might be one of the pathophysiological origins of early AD (Parodi et al, 2010; Yang et al, 2015). Several lines of evidence indicate that Aβ1-42 is associated with LEs or multivesicular bodies (MVBs) and AVs in AD brains (Takahashi et al, 2004, 2013, 2002; Yu et al, 2005).…”
Section: Introductionmentioning
confidence: 99%
“…An answer may be provided by a study that showed that endocytosed Aβ42 could accumulate in endosomes, increasing their membrane permeability and facilitating Aβ cytosolic accumulation and neuronal toxicity (Yang et al, 1998). Moreover, a recent study demonstrated that Aβ oligomers in vitro could inhibit the SNARE fusion complex assembly by direct binding to syntaxin-1a (Yang et al, 2015). Besides, in vitro, Aβ seems to be able to block synaptophysin complex formation with VAMP2 with relevance for SV exocytosis (Russell et al, 2012).…”
Section: Aβ-direct Impact On Synaptic Traffickingmentioning
confidence: 99%