Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
bioRxiv ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38645096

ABSTRACT

NOTCH1 (N1) is a transmembrane receptor interacting with membrane-tethered ligands on opposing cells that mediate the direct cell-cell interaction necessary for many cell fate decisions. Protein O -fucosyltransferase 1 (POFUT1) adds O -fucose to Epidermal Growth Factor (EGF)-like repeats in the NOTCH1 extracellular domain, which is required for trafficking and signaling activation. We previously showed that POFUT1 S162L caused a 90% loss of POFUT1 activity and global developmental defects in a patient; however, the mechanism by which POFUT1 contributes to these symptoms is still unclear. Compared to controls, POFUT1 S162L patient fibroblast cells had an equivalent amount of N1 on the cell surface but showed a 60% reduction of DLL1 ligand binding and a 70% reduction in JAG1 ligand binding. To determine if the reduction of O -fucose on N1 in POFUT1 S162L patient fibroblasts was the cause of these effects, we immunopurified endogenous N1 from control and patient fibroblasts and analyzed O -fucosylation using mass spectral glycoproteomics methods. N1 EGF8 to EGF12 comprise the ligand binding domain, and O -fucose on EGF8 and EGF12 physically interact with ligands to enhance affinity. Glycoproteomics of N1 from POFUT1 S162L patient fibroblasts showed WT fucosylation levels at all sites analyzed except for a large decrease at EGF9 and the complete absence of O -fucose at EGF12. Since the loss of O -fucose on EGF12 is known to have significant effects on N1 activity, this may explain the symptoms observed in the POFUT1 S162L patient.

2.
J Cell Biol ; 221(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-36053214

ABSTRACT

Biosynthesis of macromolecules requires precursors such as sugars or amino acids, originating from exogenous/dietary sources, reutilization/salvage of degraded molecules, or de novo synthesis. Since these sources are assumed to contribute to one homogenous pool, their individual contributions are often overlooked. Protein glycosylation uses monosaccharides from all the above sources to produce nucleotide sugars required to assemble hundreds of distinct glycans. Here, we demonstrate that cells identify the origin/heritage of the monosaccharide, fucose, for glycosylation. We measured the contribution of GDP-fucose from each of these sources for glycan synthesis and found that different fucosyltransferases, individual glycoproteins, and linkage-specific fucose residues identify and select different GDP-fucose pools dependent on their heritage. This supports the hypothesis that GDP-fucose exists in multiple, distinct pools, not as a single homogenous pool. The selection is tightly regulated since the overall pool size remains constant. We present novel perspectives on monosaccharide metabolism, which may have a general applicability.


Subject(s)
Fucose , Glycosylation , Guanosine Diphosphate Fucose , Fucose/metabolism , Guanosine Diphosphate Fucose/metabolism , Polysaccharides/metabolism
3.
J Biol Chem ; 298(7): 102064, 2022 07.
Article in English | MEDLINE | ID: mdl-35623385

ABSTRACT

NOTCH1 is a transmembrane receptor that initiates a cell-cell signaling pathway controlling various cell fate specifications in metazoans. The addition of O-fucose by protein O-fucosyltransferase 1 (POFUT1) to epidermal growth factor-like (EGF) repeats in the NOTCH1 extracellular domain is essential for NOTCH1 function, and modification of O-fucose with GlcNAc by the Fringe family of glycosyltransferases modulates Notch activity. Prior cell-based studies showed that POFUT1 modifies EGF repeats containing the appropriate consensus sequence at high stoichiometry, while Fringe GlcNAc-transferases (LFNG, MFNG, and RFNG) modify O-fucose on only a subset of NOTCH1 EGF repeats. Previous in vivo studies showed that each FNG affects naïve T cell development. To examine Fringe modifications of NOTCH1 at a physiological level, we used mass spectral glycoproteomic methods to analyze O-fucose glycans of endogenous NOTCH1 from activated T cells obtained from mice lacking all Fringe enzymes or expressing only a single FNG. While most O-fucose sites were modified at high stoichiometry, only EGF6, EGF16, EGF26, and EGF27 were extended in WT T cells. Additionally, cell-based assays of NOTCH1 lacking fucose at each of those O-fucose sites revealed small but significant effects of LFNG on Notch-Delta binding in the EGF16 and EGF27 mutants. Finally, in activated T cells expressing only LFNG, MFNG, or RFNG alone, the extension of O-fucose with GlcNAc in the same EGF repeats was diminished, consistent with cooperative interactions when all three Fringes were present. The combined data open the door for the analysis of O-glycans on endogenous NOTCH1 derived from different cell types.


Subject(s)
Epidermal Growth Factor , Fucose , Receptor, Notch1/metabolism , Animals , Epidermal Growth Factor/metabolism , Fucose/metabolism , Glucosyltransferases , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Mice , Polysaccharides/metabolism , Receptors, Notch/metabolism , T-Lymphocytes/metabolism
4.
Mol Aspects Med ; 79: 100938, 2021 06.
Article in English | MEDLINE | ID: mdl-33341260

ABSTRACT

The Notch receptors are a family of transmembrane proteins that mediate direct cell-cell interactions and control numerous cell-fate specifications in humans. The extracellular domains of mammalian Notch proteins contain 29-36 tandem epidermal growth factor-like (EGF) repeats, most of which have O-linked glycan modifications: O-glucose added by POGLUT1, O-fucose added by POFUT1 and elongated by Fringe enzymes, and O-GlcNAc added by EOGT. The extracellular domain is also N-glycosylated. Mutations in the glycosyltransferases modifying Notch have been identified in several diseases, including Dowling-Degos Disease (haploinsufficiency of POFUT1 or POGLUT1), a form of limb-girdle muscular dystrophy (autosomal recessive mutations in POGLUT1), Spondylocostal Dysostosis 3 (autosomal recessive mutations in LFNG), Adams-Oliver syndrome (autosomal recessive mutations in EOGT), and some cancers (amplification, gain or loss-of-function of POFUT1, Fringe enzymes, POGLUT1, MGAT3). Here we review the characteristics of these diseases and potential molecular mechanisms.


Subject(s)
Ectodermal Dysplasia , Limb Deformities, Congenital , Animals , Epidermal Growth Factor/metabolism , Glucosyltransferases , Glycosylation , Glycosyltransferases/genetics , Humans , Receptors, Notch/genetics , Receptors, Notch/metabolism
5.
J Biol Chem ; 291(26): 13743-52, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129198

ABSTRACT

Notch is a transmembrane receptor that mediates cell-cell interactions and controls various cell-fate specifications in metazoans. The extracellular domain of Notch contains multiple epidermal growth factor (EGF)-like repeats. At least five different glycans are found in distinct sites within these EGF-like repeats. The function of these individual glycans in Notch signaling has been investigated, primarily by disrupting their individual glycosyltransferases. However, we are just beginning to understand the potential functional interactions between these glycans. Monosaccharide O-fucose and O-glucose trisaccharide (O-glucose-xylose-xylose) are added to many of the Notch EGF-like repeats. In Drosophila, Shams adds a xylose specifically to the monosaccharide O-glucose. We found that loss of the terminal dixylose of O-glucose-linked saccharides had little effect on Notch signaling. However, our analyses of double mutants of shams and other genes required for glycan modifications revealed that both the monosaccharide O-glucose and the terminal dixylose of O-glucose-linked saccharides function redundantly with the monosaccharide O-fucose in Notch activation and trafficking. The terminal dixylose of O-glucose-linked saccharides and the monosaccharide O-glucose were required in distinct Notch trafficking processes: Notch transport from the apical plasma membrane to adherens junctions, and Notch export from the endoplasmic reticulum, respectively. Therefore, the monosaccharide O-glucose and terminal dixylose of O-glucose-linked saccharides have distinct activities in Notch trafficking, although a loss of these activities is compensated for by the presence of monosaccharide O-fucose. Given that various glycans attached to a protein motif may have redundant functions, our results suggest that these potential redundancies may lead to a serious underestimation of glycan functions.


Subject(s)
Drosophila Proteins/metabolism , Endoplasmic Reticulum/metabolism , Fucose/metabolism , Receptors, Notch/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Endoplasmic Reticulum/genetics , Fucose/genetics , Glucose/genetics , Glucose/metabolism , Glycosylation , Protein Transport/physiology , Receptors, Notch/genetics , Repetitive Sequences, Amino Acid , Xylose/genetics , Xylose/metabolism
6.
J Biol Chem ; 290(1): 505-19, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25378397

ABSTRACT

Notch (N) is a transmembrane receptor that mediates the cell-cell interactions necessary for many cell fate decisions. N has many epidermal growth factor-like repeats that are O-fucosylated by the protein O-fucosyltransferase 1 (O-Fut1), and the O-fut1 gene is essential for N signaling. However, the role of the monosaccharide O-fucose on N is unclear, because O-Fut1 also appears to have O-fucosyltransferase activity-independent functions, including as an N-specific chaperon. Such an enzymatic activity-independent function could account for the essential role of O-fut1 in N signaling. To evaluate the role of the monosaccharide O-fucose modification in N signaling, here we generated a knock-in mutant of O-fut1 (O-fut1(R245A knock-in)), which expresses a mutant protein that lacks O-fucosyltransferase activity but maintains the N-specific chaperon activity. Using O-fut1(R245A knock-in) and other gene mutations that abolish the O-fucosylation of N, we found that the monosaccharide O-fucose modification of N has a temperature-sensitive function that is essential for N signaling. The O-fucose monosaccharide and O-glucose glycan modification, catalyzed by Rumi, function redundantly in the activation of N signaling. We also showed that the redundant function of these two modifications is responsible for the presence of N at the cell surface. Our findings elucidate how different forms of glycosylation on a protein can influence the protein's functions.


Subject(s)
Drosophila melanogaster/metabolism , Fucose/chemistry , Glucose/chemistry , Protein Processing, Post-Translational , Signal Transduction/genetics , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Fucose/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Gene Knock-In Techniques , Glucose/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycosylation , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Folding , Protein Transport , Receptors, Notch/genetics , Receptors, Notch/metabolism , Temperature
7.
Zoolog Sci ; 30(10): 877-88, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24125651

ABSTRACT

The pearl oyster Pinctada fucata has great potential as a model system for lophotrochozoan developmental biology research. Pinctada fucata is an important commercial resource, and a significant body of primary research on this species has emphasized its basic aquaculture biology such as larval biology and growth, aquaculture, pearl formation and quality improvement, shell formation, and biomineralization. Recently, a draft genome sequence of this species was published, and many experimental resources are currently being developed, such as bioinformatics tools, embryo and larva manipulation methods, gene knockdown technique, etc. In this paper, we report the results from our genomic survey pertaining to gene families that encode developmental signaling ligands (Fgf, Hedgehog, PDGF/VEGF, TGFß, and Wnt families). We found most of the representative genes of major signaling pathways involved in axial patterning, as well as copies of the signaling molecule paralogs. Phylogenetic character mapping was used to infer a possible evolutionary scenario of the signaling molecules in the protostomes, and to reconstruct possible copy numbers of signaling molecule-coding genes for the ancestral protostome. Our reconstruction suggests that P. fucata retains the ancestral protostome gene complement, providing further justifications for the use of this taxon as a model organism for developmental genomics research.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Genome , Genomics , Pinctada/genetics , Pinctada/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Computer Simulation , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Models, Genetic , Molecular Sequence Annotation , Phylogeny , Pinctada/embryology , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Sequence Alignment , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Mech Dev ; 130(2-3): 169-80, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23041176

ABSTRACT

Animals often show left-right (LR) asymmetry in their body structures. In some vertebrates, the mechanisms underlying LR symmetry breaking and the subsequent signals responsible for LR asymmetric development are well understood. However, in invertebrates, the molecular bases of these processes are largely unknown. Therefore, we have been studying the genetic pathway of LR asymmetric development in Drosophila. The embryonic gut is the first organ that shows directional LR asymmetry during Drosophila development. We performed a genetic screen to identify mutations affecting LR asymmetric development of the embryonic gut. From this screen, we isolated pebble (pbl), which encodes a homolog of a mammalian RhoGEF, Ect2. The laterality of the hindgut was randomized in embryos homozygous for a null mutant of pbl. Pbl is a multi-functional protein required for cytokinesis and the epithelial-to-mesenchymal transition in Drosophila. Consistent with Pbl's role in cytokinesis, we found reduced numbers of cells in the hindgut epithelium in pbl homozygous embryos. The specific expression of pbl in the hindgut epithelium, but not in other tissues, rescued the LR defects and reduced cell number in embryonic pbl homozygotes. Embryos homozygous for string (stg), a mutant that reduces cell number through a different mechanism, also showed LR defects of the hindgut. However, the reduction in cell number in the pbl mutants was not accompanied by defects in the specification of hindgut epithelial tissues or their integrity. Based on these results, we speculate that the reduction in cell number may be one reason for the LR asymmetry defect of the pbl hindgut, although we cannot exclude contributions from other functions of Pbl, including regulation of the actin cytoskeleton through its RhoGEF activity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Gastrointestinal Tract/embryology , Guanine Nucleotide Exchange Factors/genetics , Alleles , Animals , Body Patterning/genetics , Cell Count , Cell Polarity , Cytokinesis , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Epithelial Cells/physiology , Epithelium/embryology , Epithelium/metabolism , Gastrointestinal Tract/cytology , Gene Deletion , Gene Expression , Guanine Nucleotide Exchange Factors/metabolism , Homozygote , Organ Specificity , Point Mutation , RNA Splice Sites , Sequence Analysis, DNA
9.
Proc Natl Acad Sci U S A ; 109(38): 15318-23, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22949680

ABSTRACT

Notch (N) is a transmembrane receptor that mediates cell-cell interactions to determine many cell-fate decisions. N contains EGF-like repeats, many of which have an O-fucose glycan modification that regulates N-ligand binding. This modification requires GDP-L-fucose as a donor of fucose. The GDP-L-fucose biosynthetic pathways are well understood, including the de novo pathway, which depends on GDP-mannose 4,6 dehydratase (Gmd) and GDP-4-keto-6-deoxy-D-mannose 3,5-epimerase/4-reductase (Gmer). However, the potential for intercellularly supplied GDP-L-fucose and the molecular basis of such transportation have not been explored in depth. To address these points, we studied the genetic effects of mutating Gmd and Gmer on fucose modifications in Drosophila. We found that these mutants functioned cell-nonautonomously, and that GDP-L-fucose was supplied intercellularly through gap junctions composed of Innexin-2. GDP-L-fucose was not supplied through body fluids from different isolated organs, indicating that the intercellular distribution of GDP-L-fucose is restricted within a given organ. Moreover, the gap junction-mediated supply of GDP-L-fucose was sufficient to support the fucosylation of N-glycans and the O-fucosylation of the N EGF-like repeats. Our results indicate that intercellular delivery is a metabolic pathway for nucleotide sugars in live animals under certain circumstances.


Subject(s)
Carbohydrate Epimerases/metabolism , Drosophila/metabolism , Gap Junctions/metabolism , Guanosine Diphosphate Fucose/chemistry , Hydro-Lyases/metabolism , Receptors, Notch/metabolism , Alleles , Animals , Epidermal Growth Factor/metabolism , Fucose/chemistry , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Homozygote , Models, Genetic , Mutation , Phenotype , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...