PUBLICATION

The failure of microglia to digest developmental apoptotic cells contributes to the pathology of RNASET2-deficient leukoencephalopathy

Authors
Hamilton, N., Rutherford, H.A., Petts, J.J., Isles, H.M., Weber, T., Henneke, M., Gärtner, J., Dunning, M.J., Renshaw, S.A.
ID
ZDB-PUB-200403-184
Date
2020
Source
Glia   68(7): 1531-1545 (Journal)
Registered Authors
Renshaw, Steve A., Weber, Thomas
Keywords
RNaseT2, developmental apoptosis, leukodystrophy, lysosomal storage disorder, microglia, microglia depletion, zebrafish
Datasets
GEO:GSE138493
MeSH Terms
  • Animals
  • Apoptosis/physiology*
  • Brain/metabolism*
  • Leukoencephalopathies/metabolism
  • Leukoencephalopathies/pathology*
  • Microglia/metabolism*
  • Mutation/genetics
  • Neurodegenerative Diseases/metabolism
  • Neurodegenerative Diseases/pathology
  • Phenotype
  • Zebrafish
  • Zebrafish Proteins/metabolism
PubMed
32212285 Full text @ Glia
Abstract
The contribution of microglia in neurological disorders is emerging as a leading disease driver rather than a consequence of pathology. RNAseT2-deficient leukoencephalopathy is a severe childhood white matter disorder affecting patients in their first year of life and mimicking a cytomegalovirus brain infection. The early onset and resemblance of the symptoms to a viral infection suggest an inflammatory and embryonic origin of the pathology. There are no treatments available for this disease as our understanding of the cellular drivers of the pathology are still unknown. In this study, using a zebrafish mutant for the orthologous rnaset2 gene, we have identified an inflammatory signature in early development and an antiviral immune response in mature adult brains. Using the optical transparency and the ex utero development of the zebrafish larvae we studied immune cell behavior during brain development and identified abnormal microglia as an early marker of pathology. Live imaging and electron microscopy identified that mutant microglia displayed an engorged morphology and were filled with undigested apoptotic cells and undigested substrate. Using microglia-specific depletion and rescue experiments, we identified microglia as drivers of this embryonic phenotype and potential key cellular player in the pathology of RNAseT2-deficient leukoencephalopathy. Our zebrafish model also presented with reduced survival and locomotor defects, therefore recapitulating many aspects of the human disease. Our study therefore placed our rnaset2 mutant at the forefront of leukodystrophy preclinical models and highlighted tissue-specific approaches as future therapeutic avenues.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping