PUBLICATION

Regulation of immunity and disease resistance by commensal microbes and chromatin modifications during zebrafish development

Authors
Galindo-Villegas, J., García-Moreno, D., de Oliveira, S., Meseguer, J., and Mulero, V.
ID
ZDB-PUB-120907-10
Date
2012
Source
Proceedings of the National Academy of Sciences of the United States of America   109(39): E2605-2614 (Journal)
Registered Authors
de Oliveira, Sofia, Galindo-Villegas, Jorge, Mulero, Victor
Keywords
epigenetic, cytokines, evolution, gene regulation, live imaging
MeSH Terms
  • Animals
  • Bacteria/immunology*
  • Chromatin/genetics
  • Chromatin/immunology*
  • Chromatin/metabolism
  • Germ-Free Life/genetics
  • Germ-Free Life/immunology*
  • Histones/genetics
  • Histones/immunology
  • Histones/metabolism
  • Inflammation/genetics
  • Inflammation/immunology
  • Inflammation/metabolism
  • Inflammation/microbiology
  • Myeloid Differentiation Factor 88/genetics
  • Myeloid Differentiation Factor 88/immunology*
  • Myeloid Differentiation Factor 88/metabolism
  • Toll-Like Receptors/genetics
  • Toll-Like Receptors/immunology*
  • Toll-Like Receptors/metabolism
  • Zebrafish/classification
  • Zebrafish/embryology
  • Zebrafish/immunology*
  • Zebrafish/metabolism
  • Zebrafish Proteins/genetics
  • Zebrafish Proteins/immunology*
  • Zebrafish Proteins/metabolism
PubMed
22949679 Full text @ Proc. Natl. Acad. Sci. USA
Abstract

How fish larvae are protected from infection before the maturation of adaptive immunity, a process which may take up to several weeks in most species, has long been a matter of speculation. Using a germ-free model, we show that colonization by commensals in newly hatched zebrafish primes neutrophils and induces several genes encoding proinflammatory and antiviral mediators, increasing the resistance of larvae to viral infection. Commensal microbe recognition was found to be mediated mainly through a TLR/MyD88 signaling pathway, and professional phagocytes were identified as the source of these immune mediators. However, the induction of proinflammatory and antiviral genes, but not of antimicrobial effector genes, also required the covalent modification of histone H3 at gene promoters. Interestingly, chromatin modifications were not altered by commensal microbes or hatching. Taken together, our results demonstrate that gene-specific chromatin modifications are associated with the protection of zebrafish larvae against infectious agents before adaptive immunity has developed and prevent pathologies associated with excessive inflammation during development.

Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping