PUBLICATION

Knockdown of epigenetic transcriptional co-regulator Brd2a disrupts apoptosis and proper formation of hindbrain and midbrain-hindbrain boundary (MHB) region in zebrafish

Authors
Murphy, T., Melville, H., Fradkin, E., Bistany, G., Branigan, G., Olsen, K., Comstock, C.R., Hanby, H., Garbade, E., DiBenedetto, A.J.
ID
ZDB-PUB-170528-4
Date
2017
Source
Mechanisms of Development   146: 10-30 (Journal)
Registered Authors
DiBenedetto, Angela
Keywords
Bromodomain, Cell death, Cerebellum, Embryonic patterning, Engrailed, Midbrain-hindbrain boundary
MeSH Terms
  • Animals
  • Apoptosis/genetics
  • Epigenesis, Genetic/genetics
  • Gene Expression Regulation, Developmental
  • Homeodomain Proteins/genetics
  • Mesencephalon/growth & development
  • Morphogenesis/genetics*
  • Morpholinos/genetics
  • Nerve Tissue Proteins/genetics
  • Neural Tube/growth & development*
  • Protein Serine-Threonine Kinases/genetics*
  • Rhombencephalon/growth & development
  • Somites/growth & development
  • Transcription, Genetic*
  • Zebrafish/genetics
  • Zebrafish/growth & development
  • Zebrafish Proteins/genetics*
PubMed
28549975 Full text @ Mech. Dev.
Abstract
Brd2 is a member of the bromodomain-extraterminal domain (BET) family of proteins and functions as an acetyl-histone-directed transcriptional co-regulator and recruitment scaffold in chromatin modification complexes affecting signal-dependent transcription. While Brd2 acts as a protooncogene in mammalian blood, developmental studies link it to regulation of neuronal apoptosis and epilepsy, and complete knockout of the gene is invariably embryonic lethal. In Drosophila, the Brd2 homolog acts as a maternal effect factor necessary for segment formation and identity and proper expression of homeotic loci, including Ultrabithorax and engrailed. To test the various roles attributed to Brd2 in a single developmental system representing a non-mammalian vertebrate, we conducted a phenotypic characterization of Brd2a deficient zebrafish embryos produced by morpholino knockdown and corroborated by Crispr-Cas9 disruption and small molecule inhibitor treatments. brd2aMO morphants exhibit reduced hindbrain with an ill-defined midbrain-hindbrain boundary (MHB) region; irregular notochord, neural tube, and somites; and abnormalities in ventral trunk and ventral nerve cord interneuron positioning. Using whole mount TUNEL and confocal microscopy, we uncover a significant decrease, then a dramatic increase, of p53-independent cell death at the start and end of segmentation, respectively. In contrast, using qualitative and quantitative analyses of BrdU incorporation, phosphohistone H3-tagging, and flow cytometry, we detect little effect of Brd2a knockdown on overall proliferation levels in embryos. RNA in situ hybridization shows reduced or absent expression of homeobox gene eng2a and paired box gene pax2a, in the hindbrain domain of the MHB region, and an overabundance of pax2a-positive kidney progenitors, in knockdowns. Together, these results suggest an evolutionarily conserved role for Brd2 in the proper formation and/or patterning of segmented tissues, including the vertebrate CNS, where it acts as a bi-modal regulator of apoptosis, and is necessary, directly or indirectly, for proper expression of genes that pattern the MHB and/or regulate differentiation in the anterior hindbrain.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping