scholarly journals Mouse WIF1 Is Only Modified with O-Fucose in Its EGF-like Domain III Despite Two Evolutionarily Conserved Consensus Sites

Biomolecules ◽  
2020 ◽  
Vol 10 (9) ◽  
pp. 1250
Author(s):  
Florian Pennarubia ◽  
Emilie Pinault ◽  
Bilal Al Jaam ◽  
Caroline E. Brun ◽  
Abderrahman Maftah ◽  
...  

The Wnt Inhibitory Factor 1 (Wif1), known to inhibit Wnt signaling pathways, is composed of a WIF domain and five EGF-like domains (EGF-LDs) involved in protein interactions. Despite the presence of a potential O-fucosylation site in its EGF-LDs III and V, the O-fucose sites occupancy has never been demonstrated for WIF1. In this study, a phylogenetic analysis on the distribution, conservation and evolution of Wif1 proteins was performed, as well as biochemical approaches focusing on O-fucosylation sites occupancy of recombinant mouse WIF1. In the monophyletic group of gnathostomes, we showed that the consensus sequence for O-fucose modification by Pofut1 is highly conserved in Wif1 EGF-LD III while it was more divergent in EGF-LD V. Using click chemistry and mass spectrometry, we demonstrated that mouse WIF1 was only modified with a non-extended O-fucose on its EGF-LD III. In addition, a decreased amount of mouse WIF1 in the secretome of CHO cells was observed when the O-fucosylation site in EGF-LD III was mutated. Based on sequence comparison and automated protein modeling, we suggest that the absence of O-fucose on EGF-LD V of WIF1 in mouse and probably in most gnathostomes, could be related to EGF-LD V inability to interact with POFUT1.

2016 ◽  
Vol 27 (5) ◽  
pp. 744-756 ◽  
Author(s):  
Yuki Niwa ◽  
Takehiro Suzuki ◽  
Naoshi Dohmae ◽  
Siro Simizu

R-spondin1 (Rspo1) is a secreted protein that enhances Wnt signaling, which has crucial functions in embryonic development and several cancers. C-mannosylation is a rare type of glycosylation and might regulate secretion, protein–protein interactions, and enzymatic activity. Although human Rspo1 contains 2 predicted C-mannosylation sites, C-mannosylation of Rspo1 has not been reported, nor have its functional effects on this protein. In this study, we demonstrate by mass spectrometry that Rspo1 is C-mannosylated at W153 and W156. Using Lec15.2 cells, which lack dolichol-phosphate-mannose synthesis activity, and mutant Rspo1-expressing cells that replace W153 and W156 by alanine residues, we observed that C-mannosylation of Rspo1 is required for its secretion. Further, the enhancement of canonical Wnt signaling by Rspo1 is regulated by C-mannosylation. Recently DPY19 was reported to be a C-mannosyltransferase in Caenorhabditis elegans, but no C-mannosyltransferases have been identified in any other organism. In gain- and loss-of-function experiments, human DPY19L3 selectively modified Rspo1 at W156 but not W153 based on mass spectrometry. Moreover, knockdown of DPY19L3 inhibited the secretion of Rspo1. In conclusion, we identified DPY19L3 as the C-mannosyltransferase of Rspo1 at W156 and found that DPY19L3-mediated C-mannosylation of Rspo1 at W156 is required for its secretion.


Oncotarget ◽  
2014 ◽  
Vol 5 (8) ◽  
pp. 2198-2207 ◽  
Author(s):  
Yozo Mitsui ◽  
Hiroaki Yasumoto ◽  
Taichi Nagami ◽  
Miho Hiraki ◽  
Naoko Arichi ◽  
...  

2011 ◽  
Vol 18 (8) ◽  
pp. 886-893 ◽  
Author(s):  
Tomas Malinauskas ◽  
A Radu Aricescu ◽  
Weixian Lu ◽  
Christian Siebold ◽  
E Yvonne Jones

Sign in / Sign up

Export Citation Format

Share Document