Cargando…

Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry

BACKGROUND: Astrocytes are the most abundant cells in the central nervous system and are responsible for a wide range of functions critical to normal neuronal development, synapse formation, blood-brain barrier regulation, and brain homeostasis. They are also actively involved in initiating and perp...

Descripción completa

Detalles Bibliográficos
Autores principales: Dozio, Vito, Sanchez, Jean-Charles
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6267034/
https://www.ncbi.nlm.nih.gov/pubmed/30501627
http://dx.doi.org/10.1186/s12974-018-1371-6
_version_ 1783375973974016000
author Dozio, Vito
Sanchez, Jean-Charles
author_facet Dozio, Vito
Sanchez, Jean-Charles
author_sort Dozio, Vito
collection PubMed
description BACKGROUND: Astrocytes are the most abundant cells in the central nervous system and are responsible for a wide range of functions critical to normal neuronal development, synapse formation, blood-brain barrier regulation, and brain homeostasis. They are also actively involved in initiating and perpetuating neuroinflammatory responses. However, information about their proteomic phenotypes under inflammation is currently limited. METHOD: Data-independent acquisition mass spectrometry was applied to extensively characterize the profile of more than 4000 proteins in immortalized human fetal astrocytes under distinct inflammatory conditions induced by TNF, IL-1β, and LPS, while multiplex immunoassay-based screening was used to quantify a wide range of cytokines released under these inflammatory conditions. Then, immunocytochemistry and western blotting were used to verify the activation of canonical and non-canonical NF-κB upon exposure to the different stimuli. Finally, an in vitro model of the blood-brain barrier consisting of a co-culture of primary human brain microvascular endothelial cells and primary human astrocytes was used to verify the inflammatory response of astrocytes upon LPS exposure in a more complex in vitro system. RESULTS: We reported on a set of 186 proteins whose levels were significantly modulated by TNF, IL-1β, and LPS. These three stimuli induced proteome perturbations, which led to an increased abundance of key inflammatory proteins involved in antigen presentation and non-canonical NF-κB pathways. TNF and IL-1β, but not LPS, also activated the canonical NF-κB pathway, which in turn led to an extensive inflammatory response and dysregulation of cytoskeletal and adhesion proteins. In addition, TNF and LPS, but not IL-1β, increased the abundance of several interferon-stimulated gene products. Finally, TNF and IL-1β similarly upregulated the secretion of several cytokines and chemokines, whereas LPS only induced a moderate increase in IL-8, IFN-γ, and IL-1β secretion. Upregulation of proteins associated with type I IFN and non-canonical NF-κB signaling was also observed in primary astrocytes co-cultured with primary brain microvascular endothelial cells exposed to LPS. CONCLUSIONS: The present study provides comprehensive information about the proteomic phenotypes of human astrocytes upon exposure to inflammatory stimuli both in monoculture and in co-culture with human brain microvascular endothelial cells. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s12974-018-1371-6) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6267034
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-62670342018-12-05 Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry Dozio, Vito Sanchez, Jean-Charles J Neuroinflammation Research BACKGROUND: Astrocytes are the most abundant cells in the central nervous system and are responsible for a wide range of functions critical to normal neuronal development, synapse formation, blood-brain barrier regulation, and brain homeostasis. They are also actively involved in initiating and perpetuating neuroinflammatory responses. However, information about their proteomic phenotypes under inflammation is currently limited. METHOD: Data-independent acquisition mass spectrometry was applied to extensively characterize the profile of more than 4000 proteins in immortalized human fetal astrocytes under distinct inflammatory conditions induced by TNF, IL-1β, and LPS, while multiplex immunoassay-based screening was used to quantify a wide range of cytokines released under these inflammatory conditions. Then, immunocytochemistry and western blotting were used to verify the activation of canonical and non-canonical NF-κB upon exposure to the different stimuli. Finally, an in vitro model of the blood-brain barrier consisting of a co-culture of primary human brain microvascular endothelial cells and primary human astrocytes was used to verify the inflammatory response of astrocytes upon LPS exposure in a more complex in vitro system. RESULTS: We reported on a set of 186 proteins whose levels were significantly modulated by TNF, IL-1β, and LPS. These three stimuli induced proteome perturbations, which led to an increased abundance of key inflammatory proteins involved in antigen presentation and non-canonical NF-κB pathways. TNF and IL-1β, but not LPS, also activated the canonical NF-κB pathway, which in turn led to an extensive inflammatory response and dysregulation of cytoskeletal and adhesion proteins. In addition, TNF and LPS, but not IL-1β, increased the abundance of several interferon-stimulated gene products. Finally, TNF and IL-1β similarly upregulated the secretion of several cytokines and chemokines, whereas LPS only induced a moderate increase in IL-8, IFN-γ, and IL-1β secretion. Upregulation of proteins associated with type I IFN and non-canonical NF-κB signaling was also observed in primary astrocytes co-cultured with primary brain microvascular endothelial cells exposed to LPS. CONCLUSIONS: The present study provides comprehensive information about the proteomic phenotypes of human astrocytes upon exposure to inflammatory stimuli both in monoculture and in co-culture with human brain microvascular endothelial cells. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s12974-018-1371-6) contains supplementary material, which is available to authorized users. BioMed Central 2018-11-30 /pmc/articles/PMC6267034/ /pubmed/30501627 http://dx.doi.org/10.1186/s12974-018-1371-6 Text en © The Author(s). 2018 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Dozio, Vito
Sanchez, Jean-Charles
Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry
title Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry
title_full Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry
title_fullStr Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry
title_full_unstemmed Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry
title_short Profiling the proteomic inflammatory state of human astrocytes using DIA mass spectrometry
title_sort profiling the proteomic inflammatory state of human astrocytes using dia mass spectrometry
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6267034/
https://www.ncbi.nlm.nih.gov/pubmed/30501627
http://dx.doi.org/10.1186/s12974-018-1371-6
work_keys_str_mv AT doziovito profilingtheproteomicinflammatorystateofhumanastrocytesusingdiamassspectrometry
AT sanchezjeancharles profilingtheproteomicinflammatorystateofhumanastrocytesusingdiamassspectrometry