Control of Autophagosome Axonal Retrograde Flux by Presynaptic Activity Unveiled Using Botulinum Neurotoxin Type A
Wang, Tong *; Martin, Sally; Papadopulos, Andreas; Harper, Callista B.; Mavlyutov, Timur A.; Niranjan, Dhevahi; Glass, Nick R.; Cooper-White, Justin J.; Sibarita, Jean-Baptiste; Choquet, Daniel
2015
发表期刊JOURNAL OF NEUROSCIENCE
ISSN0270-6474
卷号35期号:15页码:6179-6194
发表状态已发表
DOI10.1523/JNEUROSCI.3757-14.2015
摘要

Botulinum neurotoxin type A (BoNT/A) is a highly potent neurotoxin that elicits flaccid paralysis by enzymatic cleavage of the exocytic machinery component SNAP25 in motor nerve terminals. However, recent evidence suggests that the neurotoxic activity of BoNT/A is not restricted to the periphery, but also reaches the CNS after retrograde axonal transport. Because BoNT/A is internalized in recycling synaptic vesicles, it is unclear which compartment facilitates this transport. Using live-cell confocal and single-molecule imaging of rat hippocampal neurons cultured in microfluidic devices, we show that the activity-dependent uptake of the binding domain of the BoNT/A heavy chain (BoNT/A-Hc) is followed by a delayed increase in retrograde axonal transport of BoNT/A-Hc carriers. Consistent with a role of presynaptic activity in initiating transport of the active toxin, activity-dependent uptake of BoNT/A in the terminal led to a significant increase in SNAP25 cleavage detected in the soma chamber compared with nonstimulated neurons. Surprisingly, most endocytosed BoNT/A-Hc was incorporated into LC3-positive autophagosomes generated in the nerve terminals, which then underwent retrograde transport to the cell soma, where they fused with lysosomes both in vitro and in vivo. Blocking autophagosome formation or acidification with wortmannin or bafilomycin A(1), respectively, inhibited the activity-dependent retrograde trafficking of BoNT/A-Hc. Our data demonstrate that both the presynaptic formation of autophagosomes and the initiation of their retrograde trafficking are tightly regulated by presynaptic activity.

;

Botulinum neurotoxin type A (BoNT/A) is a highly potent neurotoxin that elicits flaccid paralysis by enzymatic cleavage of the exocytic machinery component SNAP25 in motor nerve terminals. However, recent evidence suggests that the neurotoxic activity of BoNT/A is not restricted to the periphery, but also reaches the CNS after retrograde axonal transport. Because BoNT/A is internalized in recycling synaptic vesicles, it is unclear which compartment facilitates this transport. Using live-cell confocal and single-molecule imaging of rat hippocampal neurons cultured in microfluidic devices, we show that the activity-dependent uptake of the binding domain of the BoNT/A heavy chain (BoNT/A-Hc) is followed by a delayed increase in retrograde axonal transport of BoNT/A-Hc carriers. Consistent with a role of presynaptic activity in initiating transport of the active toxin, activity-dependent uptake of BoNT/A in the terminal led to a significant increase in SNAP25 cleavage detected in the soma chamber compared with nonstimulated neurons. Surprisingly, most endocytosed BoNT/A-Hc was incorporated into LC3-positive autophagosomes generated in the nerve terminals, which then underwent retrograde transport to the cell soma, where they fused with lysosomes both in vitro and in vivo. Blocking autophagosome formation or acidification with wortmannin or bafilomycin A(1), respectively, inhibited the activity-dependent retrograde trafficking of BoNT/A-Hc. Our data demonstrate that both the presynaptic formation of autophagosomes and the initiation of their retrograde trafficking are tightly regulated by presynaptic activity.

;

Botulinum neurotoxin type A (BoNT/A) is a highly potent neurotoxin that elicits flaccid paralysis by enzymatic cleavage of the exocytic machinery component SNAP25 in motor nerve terminals. However, recent evidence suggests that the neurotoxic activity of BoNT/A is not restricted to the periphery, but also reaches the CNS after retrograde axonal transport. Because BoNT/A is internalized in recycling synaptic vesicles, it is unclear which compartment facilitates this transport. Using live-cell confocal and single-molecule imaging of rat hippocampal neurons cultured in microfluidic devices, we show that the activity-dependent uptake of the binding domain of the BoNT/A heavy chain (BoNT/A-Hc) is followed by a delayed increase in retrograde axonal transport of BoNT/A-Hc carriers. Consistent with a role of presynaptic activity in initiating transport of the active toxin, activity-dependent uptake of BoNT/A in the terminal led to a significant increase in SNAP25 cleavage detected in the soma chamber compared with nonstimulated neurons. Surprisingly, most endocytosed BoNT/A-Hc was incorporated into LC3-positive autophagosomes generated in the nerve terminals, which then underwent retrograde transport to the cell soma, where they fused with lysosomes both in vitro and in vivo. Blocking autophagosome formation or acidification with wortmannin or bafilomycin A(1), respectively, inhibited the activity-dependent retrograde trafficking of BoNT/A-Hc. Our data demonstrate that both the presynaptic formation of autophagosomes and the initiation of their retrograde trafficking are tightly regulated by presynaptic activity.

关键词autophagosome axon Botulinum neurotoxin presynaptic retrograde transport synaptic vesicle autophagosome autophagosome axon axon Botulinum neurotoxin Botulinum neurotoxin presynaptic presynaptic retrograde transport retrograde transport synaptic vesicle synaptic vesicle
URL查看原文
收录类别SCI
语种英语
WOS研究方向Neurosciences & Neurology ; Neurosciences & Neurology ; Neurosciences & Neurology
WOS类目Neurosciences ; Neurosciences ; Neurosciences
WOS记录号WOS:000353055600028
WOS关键词NEUROTROPHIN RECEPTOR P75(NTR) ; SYNAPTIC VESICLES ; CHROMAFFIN CELLS ; MOTOR-NEURONS ; SEROTYPE-A ; TRANSPORT ; SNAP-25 ; PROTEIN ; TOXIN ; CARGO ; NEUROTROPHIN RECEPTOR P75(NTR) ; SYNAPTIC VESICLES ; NEUROTROPHIN RECEPTOR P75(NTR) ; CHROMAFFIN CELLS ; SYNAPTIC VESICLES ; MOTOR-NEURONS ; CHROMAFFIN CELLS ; SEROTYPE-A ; MOTOR-NEURONS ; TRANSPORT ; SEROTYPE-A ; SNAP-25 ; TRANSPORT ; PROTEIN ; SNAP-25 ; TOXIN ; PROTEIN ; CARGO ; TOXIN ; CARGO
原始文献类型Article
引用统计
文献类型期刊论文
条目标识符https://kms.shanghaitech.edu.cn/handle/2MSLDSTB/120190
专题个人在本单位外知识产出
通讯作者Meunier, Frederic A.
作者单位1.Univ Queensland, Queensland Brain Inst, Clem Jones Ctr Ageing Dementia Res, Brisbane, Qld 4072, Australia
2.Univ Queensland, Australian Inst Bioengn & Nanotechnol, Brisbane, Qld 4072, Australia
3.Univ Queensland, Sch Chem Engn, Brisbane, Qld 4072, Australia
4.Univ Wisconsin, Sch Med, Madison, WI 53706 USA
5.MRC, Mol Biol Lab, Cambridge CB2 0QH, England
6.Commonwealth Sci & Ind Res Org, Mat Sci & Engn Div, Clayton, Vic 3169, Australia
7.Univ Bordeaux, Interdisciplinary Inst Neurosci, Bordeaux, France
8.CNRS, UMR 5297, Bordeaux, France
9.Univ Bordeaux, Bordeaux Imaging Ctr, CNRS, UMS 3420,US4,INSERM, Bordeaux, France
10.Univ Sheffield, Dept Biomed Sci, Sheffield S10 2TN, S Yorkshire, England
推荐引用方式
GB/T 7714
Wang, Tong *,Martin, Sally,Papadopulos, Andreas,et al. Control of Autophagosome Axonal Retrograde Flux by Presynaptic Activity Unveiled Using Botulinum Neurotoxin Type A[J]. JOURNAL OF NEUROSCIENCE,2015,35(15):6179-6194.
APA Wang, Tong *.,Martin, Sally.,Papadopulos, Andreas.,Harper, Callista B..,Mavlyutov, Timur A..,...&Meunier, Frederic A..(2015).Control of Autophagosome Axonal Retrograde Flux by Presynaptic Activity Unveiled Using Botulinum Neurotoxin Type A.JOURNAL OF NEUROSCIENCE,35(15),6179-6194.
MLA Wang, Tong *,et al."Control of Autophagosome Axonal Retrograde Flux by Presynaptic Activity Unveiled Using Botulinum Neurotoxin Type A".JOURNAL OF NEUROSCIENCE 35.15(2015):6179-6194.
条目包含的文件 下载所有文件
文件名称/大小 文献类型 版本类型 开放类型 使用许可
个性服务
查看访问统计
谷歌学术
谷歌学术中相似的文章
[Wang, Tong *]的文章
[Martin, Sally]的文章
[Papadopulos, Andreas]的文章
百度学术
百度学术中相似的文章
[Wang, Tong *]的文章
[Martin, Sally]的文章
[Papadopulos, Andreas]的文章
必应学术
必应学术中相似的文章
[Wang, Tong *]的文章
[Martin, Sally]的文章
[Papadopulos, Andreas]的文章
相关权益政策
暂无数据
收藏/分享
文件名: 10.1523@JNEUROSCI.3757-14.2015.pdf
格式: Adobe PDF
此文件暂不支持浏览
所有评论 (0)
暂无评论
 

除非特别说明,本系统中所有内容都受版权保护,并保留所有权利。