PUBLICATION

A zebrafish functional genomics model to investigate the role of human A20 variants in vivo

Authors
Cultrone, D., Zammit, N.W., Self, E., Postert, B., Han, J.Z.R., Bailey, J., Warren, J., Croucher, D.R., Kikuchi, K., Bogdanovic, O., Chtanova, T., Hesselson, D., Grey, S.T.
ID
ZDB-PUB-201120-49
Date
2020
Source
Scientific Reports   10: 19085 (Journal)
Registered Authors
Bogdanovic, Ozren, Cultrone, Daniele, Grey, Shane, Hesselson, Daniel, Kikuchi, Kazu, Postert, Benno
Keywords
none
MeSH Terms
  • Amino Acid Substitution
  • Animals
  • Animals, Genetically Modified
  • Autoimmune Diseases/etiology
  • Autoimmune Diseases/genetics
  • Conserved Sequence
  • Evolution, Molecular
  • Genetic Variation
  • Humans
  • Inflammation/etiology
  • Inflammation/genetics
  • Macrophages/immunology
  • Macrophages/metabolism
  • Models, Animal
  • Models, Genetic
  • Mutation, Missense
  • NF-kappa B/metabolism
  • Phosphorylation
  • Recombinant Proteins/genetics
  • Recombinant Proteins/metabolism
  • Tumor Necrosis Factor alpha-Induced Protein 3/deficiency
  • Tumor Necrosis Factor alpha-Induced Protein 3/genetics*
  • Tumor Necrosis Factor alpha-Induced Protein 3/physiology*
  • Zebrafish/genetics*
  • Zebrafish/physiology
  • Zebrafish Proteins/deficiency
  • Zebrafish Proteins/genetics*
  • Zebrafish Proteins/physiology*
PubMed
33154446 Full text @ Sci. Rep.
Abstract
Germline loss-of-function variation in TNFAIP3, encoding A20, has been implicated in a wide variety of autoinflammatory and autoimmune conditions, with acquired somatic missense mutations linked to cancer progression. Furthermore, human sequence data reveals that the A20 locus contains ~ 400 non-synonymous coding variants, which are largely uncharacterised. The growing number of A20 coding variants with unknown function, but potential clinical impact, poses a challenge to traditional mouse-based approaches. Here we report the development of a novel functional genomics approach that utilizes a new A20-deficient zebrafish (Danio rerio) model to investigate the impact of TNFAIP3 genetic variants in vivo. A20-deficient zebrafish are hyper-responsive to microbial immune activation and exhibit spontaneous early lethality. Ectopic addition of human A20 rescued A20-null zebrafish from lethality, while missense mutations at two conserved A20 residues, S381A and C243Y, reversed this protective effect. Ser381 represents a phosphorylation site important for enhancing A20 activity that is abrogated by its mutation to alanine, or by a causal C243Y mutation that triggers human autoimmune disease. These data reveal an evolutionarily conserved role for TNFAIP3 in limiting inflammation in the vertebrate linage and show how this function is controlled by phosphorylation. They also demonstrate how a zebrafish functional genomics pipeline can be utilized to investigate the in vivo significance of medically relevant human TNFAIP3 gene variants.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping