PUBLICATION

The Nogo-C2/Nogo receptor complex regulates the morphogenesis of zebrafish lateral line primordium through modulating the expression of dkk1b, a Wnt signal inhibitor

Authors
Han, H.W., Chou, C.M., Chu, C.Y., Cheng, C.H., Yang, C.H., Hung, C.C., Hwang, P.P., Lee, S.J., Liao, Y.F., and Huang, C.J.
ID
ZDB-PUB-140321-56
Date
2014
Source
PLoS One   9(1): e86345 (Journal)
Registered Authors
Huang, Chang-Jen, Hwang, Pung Pung, Lee, Shyh-Jye, Liao, Yun-Feng
Keywords
none
MeSH Terms
  • Animals
  • Embryo, Nonmammalian/physiology
  • Embryonic Development/genetics
  • Gene Expression Regulation, Developmental/genetics
  • Intercellular Signaling Peptides and Proteins/genetics*
  • Lateral Line System/physiology*
  • Morphogenesis/genetics*
  • Myelin Proteins/genetics*
  • Receptors, Cell Surface/genetics*
  • Signal Transduction/genetics
  • Wnt Signaling Pathway/genetics*
  • Zebrafish
  • Zebrafish Proteins/genetics*
PubMed
24466042 Full text @ PLoS One
Abstract

The fish lateral line (LL) is a mechanosensory system closely related to the hearing system of higher vertebrates, and it is composed of several neuromasts located on the surface of the fish. These neuromasts can detect changes in external water flow, to assist fish in maintaining a stationary position in a stream. In the present study, we identified a novel function of Nogo/Nogo receptor signaling in the formation of zebrafish neuromasts. Nogo signaling in zebrafish, like that in mammals, involves three ligands and four receptors, as well as three co-receptors (TROY, p75, and LINGO-1). We first demonstrated that Nogo-C2, NgRH1a, p75, and TROY are able to form a Nogo-C2 complex, and that disintegration of this complex causes defective neuromast formation in zebrafish. Time-lapse recording of the CldnB::lynEGFP transgenic line revealed that functional obstruction of the Nogo-C2 complex causes disordered morphogenesis, and reduces rosette formation in the posterior LL (PLL) primordium during migration. Consistent with these findings, hair-cell progenitors were lost from the PLL primordium in p75, TROY, and Nogo-C2/NgRH1a morphants. Notably, the expression levels of pea3, a downstream marker of Fgf signaling, and dkk1b, a Wnt signaling inhibitor, were both decreased in p75, TROY, and Nogo-C2/NgRH1a morphants; moreover, dkk1b mRNA injection could rescue the defects in neuromast formation resulting from knockdown of p75 or TROY. We thus suggest that a novel Nogo-C2 complex, consisting of Nogo-C2, NgRH1a, p75, and TROY, regulates Fgf signaling and dkk1b expression, thereby ensuring stable organization of the PLL primordium.

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