α-Synuclein amplifies cytoplasmic peroxide flux and oxidative stress provoked by mitochondrial inhibitors in CNS dopaminergic neurons in vivo

Convergent evidence implicates impaired mitochondrial function and α-Synuclein accumulation as critical upstream events in the pathogenesis of Parkinson's disease, but comparatively little is known about how these factors interact to provoke neurodegeneration. We previously showed that α-Synucl...

Full description

Saved in:
Bibliographic Details
Published inRedox biology Vol. 37; p. 101695
Main Authors Van Laar, Victor S., Chen, Jianming, Zharikov, Alevtina D., Bai, Qing, Di Maio, Roberto, Dukes, April A., Hastings, Teresa G., Watkins, Simon C., Greenamyre, J. Timothy, St Croix, Claudette M., Burton, Edward A.
Format Journal Article
LanguageEnglish
Published Netherlands Elsevier B.V 01.10.2020
Elsevier
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Convergent evidence implicates impaired mitochondrial function and α-Synuclein accumulation as critical upstream events in the pathogenesis of Parkinson's disease, but comparatively little is known about how these factors interact to provoke neurodegeneration. We previously showed that α-Synuclein knockdown protected rat substantia nigra dopaminergic neurons from systemic exposure to the mitochondrial complex I inhibitor rotenone. Here we show that motor abnormalities prior to neuronal loss in this model are associated with extensive α-Synuclein-dependent cellular thiol oxidation. In order to elucidate the underlying events in vivo we constructed novel transgenic zebrafish that co-express, in dopaminergic neurons: (i) human α-Synuclein at levels insufficient to provoke neurodegeneration or neurobehavioral abnormalities; and (ii) genetically-encoded ratiometric fluorescent biosensors to detect cytoplasmic peroxide flux and glutathione oxidation. Live intravital imaging of the intact zebrafish CNS at cellular resolution showed unequivocally that α-Synuclein amplified dynamic cytoplasmic peroxide flux in dopaminergic neurons following exposure to the mitochondrial complex I inhibitors MPP+ or rotenone. This effect was robust and clearly evident following either acute or prolonged exposure to each inhibitor. In addition, disturbance of the resting glutathione redox potential following exogenous hydrogen peroxide challenge was augmented by α-Synuclein. Together these data show that α-Synuclein is a critical determinant of the redox consequences of mitochondrial dysfunction in dopaminergic neurons. The findings are important because the mechanisms underlying α-Synuclein-dependent reactive oxygen species fluxes and antioxidant suppression might provide a pharmacological target in Parkinson's disease to prevent progression from mitochondrial dysfunction and oxidative stress to cell death. [Display omitted] •Extensive neuronal thiol oxidation in a rat PD model is α-Synuclein-dependent.•Peroxide flux and glutathione oxidation can be imaged in live transgenic zebrafish.•α-Synuclein amplifies cytosolic peroxide flux in dopaminergic neurons.•α-Synuclein exacerbates dynamic disturbances of the glutathione redox potential.•The underlying molecular mechanisms may provide therapeutic targets in PD.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Contributed equally and should be considered joint first authors.
ISSN:2213-2317
2213-2317
DOI:10.1016/j.redox.2020.101695