Notch signaling is critical for numerous cell fate decisions in mammals. Mutations that disrupt Notch signaling cause widespread developmental defects and numerous cancers. Therefore, it is critical to understand the factors required for optimal Notch signaling. It is now clear that the glycans added to Notch receptors are key to regulating the level of Notch signaling. Recent experiments identify O-fucose and O-GlcNAc glycans of Notch receptors as those that mediate interactions with canonical Notch ligands, and thereby regulate Notch signal induction. Human diseases and mouse mutants in which O- fucose or O-GlcNAc glycans are reduced reveal that these glycans are not interchangeable. Thus it is important to determine their separate functions. The overall aim of this proposal is to identify specific cell fate decisions regulated by O-fucose versus O-GlcNAc glycans on Notch receptors. Two cell differentiation pathways will be used to address this question - hematopoietic stem cells (HSC) from which all lymphoid and myeloid cell fates develop, and intestinal stem cells (ISC) from which all intestinal crypt-villus cell fates develop. Both HSC and ISC exhibit considerable Notch signaling in the absence of O-fucose glycans. We hypothesize that O-GlcNAc glycans are responsible for this residual Notch signaling via interactions with one or more of the canonical Notch ligands. We will test this hypothesis by determining ligand-induced cell fate decisions following removal or truncation of O-fucose and/or O- GlcNAc glycans in HSC and ISC. Mutant HSC and ISC will be generated by conditionally inactivating genes that encode either the enzyme that adds O-fucose (Pofut1), or one or more Fringe enzymes that extend the fucose with GlcNAc (Lfng, Mfng and Rfng), or the enzyme that adds O-GlcNAc (Eogt).
Specific Aim 1 will generate mice with HSC lacking Pofut1 with and without Eogt, or all three Fringe genes with and without Eogt. Lymphoid and myeloid cell fates generated will be analyzed in thymus and spleen. The mechanism of altered differentiation will be investigated by defining the binding of Notch ligands Dll1, Dll4, Jag1 and Jag2 to mutant HSC, the differentiation that each ligand induces in co- culture assays, and the different gene expression consequences.
Specific Aim 2 will analyze intestinal cell fates in mice expressing a single Fringe (Lfng, Mfng or Rfng), no Fringe, or all Fringe genes, with and without Eogt. Mice that exhibit altered intestinal cell fate distributions will be investigated by lineage tracing of Lgr5+ cells. Notch ligand binding changes that induce altered cell fate distributions will be identified by binding assays, differentiation in mutant crypt organoids cultured with Dll1, Dll4, Jag1 or Jag2, and gene expression analyses. Interactions of O-GlcNAc glycans with each ligand will be identified in ISC conditionally lacking Pofut1+/-Eogt or all Fringes+/-Eogt by similar in vivo analyses and crypt organoid mechanistic strategies. Important new insights into mechanisms of O-glycan regulation of Notch signaling strength will be obtained.

Public Health Relevance

Disruption of Notch signaling causes defects in development and many cancers. Optimal Notch signaling depends on ligand interactions with O-glycans on Notch receptors, key regulators of the strength of Notch signaling. We will determine specific contributions of O-fucose versus O-GlcNAc glycans to Notch signaling.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM106417-19
Application #
9896834
Study Section
Intercellular Interactions Study Section (ICI)
Program Officer
Bond, Michelle Rueffer
Project Start
2002-04-01
Project End
2022-03-31
Budget Start
2020-04-01
Budget End
2021-03-31
Support Year
19
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Albert Einstein College of Medicine
Department
Type
DUNS #
081266487
City
Bronx
State
NY
Country
United States
Zip Code
10461
Schneider, Michael; Kumar, Vivek; Nordstrøm, Lars Ulrik et al. (2018) Inhibition of Delta-induced Notch signaling using fucose analogs. Nat Chem Biol 14:65-71
Varshney, Shweta; Stanley, Pamela (2018) Multiple roles for O-glycans in Notch signalling. FEBS Lett 592:3819-3834
Sawaguchi, Shogo; Varshney, Shweta; Ogawa, Mitsutaka et al. (2017) O-GlcNAc on NOTCH1 EGF repeats regulates ligand-induced Notch signaling and vascular development in mammals. Elife 6:
Varshney, Shweta; Stanley, Pamela (2017) EOGT and O-GlcNAc on secreted and membrane proteins. Biochem Soc Trans 45:401-408
Wang, Yidong; Wu, Bingruo; Lu, Pengfei et al. (2017) Uncontrolled angiogenic precursor expansion causes coronary artery anomalies in mice lacking Pofut1. Nat Commun 8:578
Varshney, Shweta; Stanley, Pamela (2017) Notch Ligand Binding Assay Using Flow Cytometry. Bio Protoc 7:
Stanley, Pamela (2016) What Have We Learned from Glycosyltransferase Knockouts in Mice? J Mol Biol 428:3166-3182
Song, Yinghui; Kumar, Vivek; Wei, Hua-Xing et al. (2016) Lunatic, Manic, and Radical Fringe Each Promote T and B Cell Development. J Immunol 196:232-43
Wang, Weihuan; Yu, Shuiliang; Zimmerman, Grant et al. (2015) Notch Receptor-Ligand Engagement Maintains Hematopoietic Stem Cell Quiescence and Niche Retention. Stem Cells 33:2280-93
Tashima, Yuko; Stanley, Pamela (2014) Antibodies that detect O-linked ?-D-N-acetylglucosamine on the extracellular domain of cell surface glycoproteins. J Biol Chem 289:11132-42