PUBLICATION

Topology and dynamics of the zebrafish segmentation clock core circuit

Authors
Schröter, C., Ares, S., Morelli, L.G., Isakova, A., Hens, K., Soroldoni, D., Gajewski, M., Jülicher, F., Maerkl, S.J., Deplancke, B., and Oates, A.C.
ID
ZDB-PUB-120823-3
Date
2012
Source
PLoS Biology   10(7): e1001364 (Journal)
Registered Authors
Gajewski, Martin, Oates, Andrew
Keywords
Embryos, Genetic oscillators, Morphogenic segmentation, DNA-binding proteins, Dimers (Chemical physics), Phenotypes, Zebrafish, Gene expression
MeSH Terms
  • Animals
  • Basic Helix-Loop-Helix Transcription Factors/genetics
  • Basic Helix-Loop-Helix Transcription Factors/metabolism
  • Biological Clocks/genetics*
  • Body Patterning
  • Dimerization
  • Feedback, Physiological
  • Gene Expression Regulation, Developmental*
  • Models, Biological
  • Phenotype
  • Promoter Regions, Genetic
  • Protein Interaction Mapping
  • Protein Interaction Maps
  • Protein Stability
  • RNA, Messenger/genetics
  • RNA, Messenger/metabolism
  • Repressor Proteins/genetics
  • Repressor Proteins/metabolism
  • Somites/cytology
  • Somites/embryology
  • Somites/metabolism
  • Substrate Specificity
  • Transcription Factors/genetics
  • Transcription Factors/metabolism
  • Transcription, Genetic
  • Two-Hybrid System Techniques
  • Zebrafish/embryology
  • Zebrafish/genetics*
  • Zebrafish/metabolism
  • Zebrafish Proteins/genetics
  • Zebrafish Proteins/metabolism
PubMed
22911291 Full text @ PLoS Biol.
Abstract

During vertebrate embryogenesis, the rhythmic and sequential segmentation of the body axis is regulated by an oscillating genetic network termed the segmentation clock. We describe a new dynamic model for the core pace-making circuit of the zebrafish segmentation clock based on a systematic biochemical investigation of the network's topology and precise measurements of somitogenesis dynamics in novel genetic mutants. We show that the core pace-making circuit consists of two distinct negative feedback loops, one with Her1 homodimers and the other with Her7:Hes6 heterodimers, operating in parallel. To explain the observed single and double mutant phenotypes of her1, her7, and hes6 mutant embryos in our dynamic model, we postulate that the availability and effective stability of the dimers with DNA binding activity is controlled in a “dimer cloud” that contains all possible dimeric combinations between the three factors. This feature of our model predicts that Hes6 protein levels should oscillate despite constant hes6 mRNA production, which we confirm experimentally using novel Hes6 antibodies. The control of the circuit's dynamics by a population of dimers with and without DNA binding activity is a new principle for the segmentation clock and may be relevant to other biological clocks and transcriptional regulatory networks.

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