Cargando…

Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia

Recently, extracellular vesicles (EVs), such as exosomes, have been proposed to play an influential role in the cell-to-cell spread of neurodegenerative diseases, including the intercellular transmission of α-synuclein (α-syn). However, the regulation of EV biogenesis and its relation to Parkinson’s...

Descripción completa

Detalles Bibliográficos
Autores principales: Sackmann, Valerie, Sinha, Maitrayee Sardar, Sackmann, Christopher, Civitelli, Livia, Bergström, Joakim, Ansell-Schultz, Anna, Hallbeck, Martin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6724746/
https://www.ncbi.nlm.nih.gov/pubmed/31555088
http://dx.doi.org/10.3389/fnmol.2019.00200
_version_ 1783449040946462720
author Sackmann, Valerie
Sinha, Maitrayee Sardar
Sackmann, Christopher
Civitelli, Livia
Bergström, Joakim
Ansell-Schultz, Anna
Hallbeck, Martin
author_facet Sackmann, Valerie
Sinha, Maitrayee Sardar
Sackmann, Christopher
Civitelli, Livia
Bergström, Joakim
Ansell-Schultz, Anna
Hallbeck, Martin
author_sort Sackmann, Valerie
collection PubMed
description Recently, extracellular vesicles (EVs), such as exosomes, have been proposed to play an influential role in the cell-to-cell spread of neurodegenerative diseases, including the intercellular transmission of α-synuclein (α-syn). However, the regulation of EV biogenesis and its relation to Parkinson’s disease (PD) is only partially understood. The generation of EVs through the ESCRT-independent pathway depends on the hydrolysis of sphingomyelin by neutral sphingomyelinase 2 (nSMase2) to produce ceramide, which causes the membrane of endosomal multivesicular bodies to bud inward. nSMase2 is sensitive to oxidative stress, a common process in PD brains; however, little is known about the role of sphingomyelin metabolism in the pathogenesis of PD. This is the first study to show that inhibiting nSMase2 decreases the transfer of oligomeric aggregates of α-syn between neuron-like cells. Furthermore, it reduced the accumulation and aggregation of high-molecular-weight α-syn. Hypoxia, as a model of oxidative stress, reduced the levels of nSMase2, but not its enzymatic activity, and significantly altered the lipid composition of cells without affecting EV abundance or the transfer of α-syn. These data show that altering sphingolipids can mitigate the spread of α-syn, even under hypoxic conditions, potentially suppressing PD progression.
format Online
Article
Text
id pubmed-6724746
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-67247462019-09-25 Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia Sackmann, Valerie Sinha, Maitrayee Sardar Sackmann, Christopher Civitelli, Livia Bergström, Joakim Ansell-Schultz, Anna Hallbeck, Martin Front Mol Neurosci Neuroscience Recently, extracellular vesicles (EVs), such as exosomes, have been proposed to play an influential role in the cell-to-cell spread of neurodegenerative diseases, including the intercellular transmission of α-synuclein (α-syn). However, the regulation of EV biogenesis and its relation to Parkinson’s disease (PD) is only partially understood. The generation of EVs through the ESCRT-independent pathway depends on the hydrolysis of sphingomyelin by neutral sphingomyelinase 2 (nSMase2) to produce ceramide, which causes the membrane of endosomal multivesicular bodies to bud inward. nSMase2 is sensitive to oxidative stress, a common process in PD brains; however, little is known about the role of sphingomyelin metabolism in the pathogenesis of PD. This is the first study to show that inhibiting nSMase2 decreases the transfer of oligomeric aggregates of α-syn between neuron-like cells. Furthermore, it reduced the accumulation and aggregation of high-molecular-weight α-syn. Hypoxia, as a model of oxidative stress, reduced the levels of nSMase2, but not its enzymatic activity, and significantly altered the lipid composition of cells without affecting EV abundance or the transfer of α-syn. These data show that altering sphingolipids can mitigate the spread of α-syn, even under hypoxic conditions, potentially suppressing PD progression. Frontiers Media S.A. 2019-08-28 /pmc/articles/PMC6724746/ /pubmed/31555088 http://dx.doi.org/10.3389/fnmol.2019.00200 Text en Copyright © 2019 Sackmann, Sinha, Sackmann, Civitelli, Bergström, Ansell-Schultz and Hallbeck. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Neuroscience
Sackmann, Valerie
Sinha, Maitrayee Sardar
Sackmann, Christopher
Civitelli, Livia
Bergström, Joakim
Ansell-Schultz, Anna
Hallbeck, Martin
Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia
title Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia
title_full Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia
title_fullStr Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia
title_full_unstemmed Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia
title_short Inhibition of nSMase2 Reduces the Transfer of Oligomeric α-Synuclein Irrespective of Hypoxia
title_sort inhibition of nsmase2 reduces the transfer of oligomeric α-synuclein irrespective of hypoxia
topic Neuroscience
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6724746/
https://www.ncbi.nlm.nih.gov/pubmed/31555088
http://dx.doi.org/10.3389/fnmol.2019.00200
work_keys_str_mv AT sackmannvalerie inhibitionofnsmase2reducesthetransferofoligomericasynucleinirrespectiveofhypoxia
AT sinhamaitrayeesardar inhibitionofnsmase2reducesthetransferofoligomericasynucleinirrespectiveofhypoxia
AT sackmannchristopher inhibitionofnsmase2reducesthetransferofoligomericasynucleinirrespectiveofhypoxia
AT civitellilivia inhibitionofnsmase2reducesthetransferofoligomericasynucleinirrespectiveofhypoxia
AT bergstromjoakim inhibitionofnsmase2reducesthetransferofoligomericasynucleinirrespectiveofhypoxia
AT ansellschultzanna inhibitionofnsmase2reducesthetransferofoligomericasynucleinirrespectiveofhypoxia
AT hallbeckmartin inhibitionofnsmase2reducesthetransferofoligomericasynucleinirrespectiveofhypoxia