4.8 Article

FGFR1 SUMOylation coordinates endothelial angiogenic signaling in angiogenesis

Publisher

NATL ACAD SCIENCES
DOI: 10.1073/pnas.2202631119

Keywords

SUMOylation; FGFR1; FRS2a; angiogenesis

Funding

  1. National Natural Science Foundation of China [91839104, 81770444, 81600354, 81970372, 11932017, 31800972]
  2. National Key R&D Program of China [2018YFA0800504, 2021YFA1101101]
  3. Zhejiang Provincial Natural Science Foundation of China [LZ20H020002]
  4. Fundamental Research Funds for the Central Universities of China
  5. Medical and Health Science and Technology Program of Health Commission of Zhejiang Province, China [2021KY633]

Ask authors/readers for more resources

FGFR1 SUMOylation, regulated by SENP1, controls angiogenic signaling pathway, competing with VEGFR2 to recruit FRS2 alpha, thus switching signals in hypoxia and normoxia angiogenic environments.
Angiogenesis contributes fundamentally to embryonic development, tissue homeostasis, and wound healing. Basic fibroblast growth factor (FGF2) is recognized as the first proangiogenic molecule discovered, and it facilitates angiogenesis by activating FGF receptor 1 (FGFR1) signaling in endothelial cells. However, the precise roles of FGFR and the FGF/FGFR signaling axis in angiogenesis remain unclear, especially because of the contradictory phenotypes of in vivo FGF and FGFR gene deficiency models. Our previous study results suggested a potential role of posttranslational small ubiquitin-like modifier modification (SUMOylation), with highly dynamic regulatory features, in vascular development and disorder. Here, we identified SENP1-regulated endothelial FGFR1 SUMOylation at conserved lysines responding to proangiogenic stimuli, while SENP1 functioned as the deSUMOylase. Hypoxia-enhanced FGFR1 SUMOylation restricted the tyrosine kinase activation of FGFR1 by modulating the dimerization of FGFR1 and FGFR1 binding with its phosphatase PTPRG. Consequently, it facilitated the recruitment of FRS2 alpha to VEGFR2 but limited additional recruitment of FRS2 alpha to FGFR1, supporting the activation of VEGFA/VEGFR2 signaling in endothelial cells. Furthermore, SUMOylation-defective mutation of FGFR1 resulted in exaggerated FGF2/FGFR1 signaling but suppressed VEGFA/VEGFR2 signaling and the angiogenic capabilities of endothelial cells, which were rescued by FRS2 alpha overexpression. Reduced angiogenesis and endothelial sprouting in mice bearing an endothelial-specific, FGFR1 SUMOylation-defective mutant confirmed the functional significance of endothelial FGFR1 SUMOylation in vivo. Our findings identify the reversible SUMOylation of FGFR1 as an intrinsic fine-tuned mechanism in coordinating endothelial angiogenic signaling during neovascularization; SENP1-regulated FGFR1 SUMOylation and deSUMOylation controls the competitive recruitment of FRS2 alpha by FGFR1 and VEGFR2 to switch receptor-complex formation responding to hypoxia and normoxia angiogenic environments.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available