RelB controls adaptive responses of astrocytes during sterile inflammation

Glia. 2019 Aug;67(8):1449-1461. doi: 10.1002/glia.23619. Epub 2019 Apr 7.

Abstract

In response to brain injury or infections, astrocytes become reactive, undergo striking morphological and functional changes, and secrete and respond to a spectrum of inflammatory mediators. We asked whether reactive astrocytes also display adaptive responses during sterile IL-1β-induced neuroinflammation, which may limit tissue injury associated with many disorders of the central nervous system. We found that astrocytes display days-to-weeks long specific tolerance of cytokine genes, which is coordinated by NF-κB family member, RelB. However, in contrast to innate immune cells, astrocytic tolerance does not involve epigenetic silencing of the cytokine genes. Establishment of tolerance depends on persistent higher levels of RelB in tolerant astrocytes and its phosphorylation on serine 472. Mechanistically, this phosphorylation prevents efficient removal of RelB from cytokine promoters by IκBα and helps to establish tolerance. Importantly, ablation of RelB from astrocytes in mice abolishes tolerance during experimental neuroinflammation in vivo.

Keywords: IL-1β; RelB; astrocytes; neuroinflammation; tolerance.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, N.I.H., Intramural

MeSH terms

  • Adaptive Immunity / physiology*
  • Animals
  • Astrocytes / immunology*
  • Brain / immunology
  • Cytokines / metabolism
  • Epigenesis, Genetic
  • HEK293 Cells
  • Humans
  • Immune Tolerance / physiology
  • Inflammation / metabolism*
  • Mice, Transgenic
  • Neuroimmunomodulation
  • Phosphorylation
  • Sirtuin 1 / metabolism
  • Transcription Factor RelB / genetics
  • Transcription Factor RelB / metabolism*

Substances

  • Cytokines
  • RELB protein, human
  • Relb protein, mouse
  • Transcription Factor RelB
  • Sirtuin 1