PUBLICATION

Prostaglandin Gbetagamma signaling stimulates gastrulation movements by limiting cell adhesion through Snai1a stabilization

Authors
Speirs, C.K., Jernigan, K.K., Kim, S.H., Cha, Y.I., Lin, F., Sepich, D.S., DuBois, R.N., Lee, E., and Solnica-Krezel, L.
ID
ZDB-PUB-100330-14
Date
2010
Source
Development (Cambridge, England)   137(8): 1327-1337 (Journal)
Registered Authors
Kim, Seok-Hyung, Lin, Fang, Sepich, Diane, Solnica-Krezel, Lilianna, Speirs, Christina
Keywords
Prostaglandin E2, Gβγ, Gastrulation, Cell adhesion, Snail, Zebrafish
MeSH Terms
  • Animals
  • Cell Adhesion/physiology
  • DNA Primers
  • Dinoprostone/deficiency
  • Dinoprostone/metabolism
  • Embryo, Nonmammalian/physiology
  • Gastrula/physiology*
  • Gastrulation/physiology
  • In Situ Hybridization
  • Prostaglandins G/physiology*
  • RNA/genetics
  • Reverse Transcriptase Polymerase Chain Reaction
  • Signal Transduction/physiology
  • Transcription Factors/genetics
  • Zebrafish/embryology
  • Zebrafish/genetics*
  • Zebrafish/physiology
  • Zebrafish Proteins/genetics
PubMed
20332150 Full text @ Development
Abstract
Gastrulation movements form the germ layers and shape them into the vertebrate body. Gastrulation entails a variety of cell behaviors, including directed cell migration and cell delamination, which are also involved in other physiological and pathological processes, such as cancer metastasis. Decreased Prostaglandin E(2) (PGE(2)) synthesis due to interference with the Cyclooxygenase (Cox) and Prostaglandin E synthase (Ptges) enzymes halts gastrulation and limits cancer cell invasiveness, but how PGE(2) regulates cell motility remains unclear. Here we show that PGE(2)-deficient zebrafish embryos, impaired in the epiboly, internalization, convergence and extension gastrulation movements, exhibit markedly increased cell-cell adhesion, which contributes to defective cell movements in the gastrula. Our analyses reveal that PGE(2) promotes cell protrusive activity and limits cell adhesion by modulating E-cadherin transcript and protein, in part through stabilization of the Snai1a (also known as Snail1) transcriptional repressor, an evolutionarily conserved regulator of cell delamination and directed migration. We delineate a pathway whereby PGE(2) potentiates interaction between the receptor-coupled G protein betagamma subunits and Gsk3beta to inhibit proteasomal degradation of Snai1a. However, overexpression of beta-catenin cannot stabilize Snai1a in PGE(2)-deficient gastrulae. Thus, the Gsk3beta-mediated and beta-catenin-independent inhibition of cell adhesion by Prostaglandins provides an additional mechanism for the functional interactions between the PGE(2) and Wnt signaling pathways during development and disease. We propose that ubiquitously expressed PGE(2) synthesizing enzymes, by promoting the stability of Snai1a, enable the precise and rapid regulation of cell adhesion that is required for the dynamic cell behaviors that drive various gastrulation movements.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping