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

J Neuroinflammation. 2018 Nov 30;15(1):331. doi: 10.1186/s12974-018-1371-6.

Abstract

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.

Keywords: Astrocytes; Blood-brain barrier; DIA MS; Inflammation; Proteomics.

MeSH terms

  • Astrocytes / metabolism*
  • Blood-Brain Barrier / drug effects
  • Blood-Brain Barrier / metabolism*
  • Cells, Cultured
  • Cluster Analysis
  • Coculture Techniques
  • Cytokines / metabolism*
  • Endothelial Cells
  • Fetus / cytology
  • Humans
  • Inflammation / chemically induced
  • Inflammation / metabolism*
  • L-Lactate Dehydrogenase / metabolism
  • Lipopolysaccharides / pharmacology
  • Mass Spectrometry / methods*
  • Models, Animal
  • Proteomics / methods*
  • Signal Transduction / drug effects
  • Signal Transduction / physiology
  • Time Factors

Substances

  • Cytokines
  • Lipopolysaccharides
  • L-Lactate Dehydrogenase