PUBLICATION

Spliceosomal components protect embryonic neurons from R-loop-mediated DNA damage and apoptosis

Authors
Sorrells, S., Nik, S., Casey, M., Cameron, R.C., Truong, H., Toruno, C., Gulfo, M., Lowe, A., Jette, C., Stewart, R.A., Bowman, T.V.
ID
ZDB-PUB-180209-5
Date
2018
Source
Disease models & mechanisms   11(2): (Journal)
Registered Authors
Bowman, Teresa, Jette, Cicely A., Sorrells, Shelly, Stewart, Rodney A., Toruno, Cristhian, Truong, Harold
Keywords
Apoptosis, Neurons, R-loops, Radiation, Splicing, Zebrafish
MeSH Terms
  • Animals
  • Apoptosis*/radiation effects
  • Cytoprotection*/radiation effects
  • DNA Breaks, Double-Stranded
  • DNA Damage*
  • Genes, Essential
  • Mutation/genetics
  • Neurons/cytology*
  • Neurons/metabolism*
  • Neurons/radiation effects
  • Nucleic Acid Conformation*
  • RNA Splicing/genetics
  • RNA Splicing/radiation effects
  • Radiation Tolerance/genetics
  • Radiation Tolerance/radiation effects
  • Radiation, Ionizing
  • Spliceosomes/metabolism*
  • Tumor Suppressor Protein p53/metabolism
  • Zebrafish/embryology
  • Zebrafish/genetics*
  • Zebrafish Proteins/metabolism
PubMed
29419415 Full text @ Dis. Model. Mech.
Abstract
RNA splicing factors are essential for the viability of all eukaryotic cells; however, in metazoans some cell types are exquisitely sensitive to disruption of splicing factors. Neuronal cells represent one such cell type, and defects in RNA splicing factors can lead to neurodegenerative diseases. The basis for this tissue selectivity is not well understood owing to difficulties in analyzing the consequences of splicing factor defects in whole-animal systems. Here, we use zebrafish mutants to show that loss of spliceosomal components, including splicing factor 3b, subunit 1 (sf3b1), causes increased DNA double-strand breaks and apoptosis in embryonic neurons. Moreover, these mutants show a concomitant accumulation of R-loops, which are non-canonical nucleic acid structures that promote genomic instability. Dampening R-loop formation by conditional induction of ribonuclease H1 in sf3b1 mutants reduced neuronal DNA damage and apoptosis. These findings show that splicing factor dysfunction leads to R-loop accumulation and DNA damage that sensitizes embryonic neurons to apoptosis. Our results suggest that diseases associated with splicing factor mutations could be susceptible to treatments that modulate R-loop levels.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping