Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 603(7899): 174-179, 2022 03.
Article in English | MEDLINE | ID: mdl-35173332

ABSTRACT

Lassa virus (LASV) is a human pathogen, causing substantial morbidity and mortality1,2. Similar to other Arenaviridae, it presents a class-I spike complex on its surface that facilitates cell entry. The virus's cellular receptor is matriglycan, a linear carbohydrate that is present on α-dystroglycan3,4, but the molecular mechanism that LASV uses to recognize this glycan is unknown. In addition, LASV and other arenaviruses have a unique signal peptide that forms an integral and functionally important part of the mature spike5-8; yet the structure, function and topology of the signal peptide in the membrane remain uncertain9-11. Here we solve the structure of a complete native LASV spike complex, finding that the signal peptide crosses the membrane once and that its amino terminus is located in the extracellular region. Together with a double-sided domain-switching mechanism, the signal peptide helps to stabilize the spike complex in its native conformation. This structure reveals that the LASV spike complex is preloaded with matriglycan, suggesting the mechanism of binding and rationalizing receptor recognition by α-dystroglycan-tropic arenaviruses. This discovery further informs us about the mechanism of viral egress and may facilitate the rational design of novel therapeutics that exploit this binding site.


Subject(s)
Dystroglycans , Lassa virus , Receptors, Virus , Viral Envelope Proteins , Dystroglycans/chemistry , Dystroglycans/metabolism , Humans , Lassa Fever/virology , Lassa virus/chemistry , Lassa virus/metabolism , Protein Conformation , Protein Sorting Signals , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization
2.
Biochem Biophys Res Commun ; 703: 149656, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38364681

ABSTRACT

Dystroglycan (DG) is a cell adhesion complex that is widely expressed in tissues. It is composed by two subunits, α-DG, a highly glycosylated protein that interacts with several extracellular matrix proteins, and transmembrane ß-DG whose, cytodomain binds to the actin cytoskeleton. Glycosylation of α-DG is crucial for functioning as a receptor for its multiple extracellular binding partners. Perturbation of α-DG glycosylation is the central event in the pathogenesis of severe pathologies such as muscular dystrophy and cancer. ß-DG acts as a scaffold for several cytoskeletal and nuclear proteins and very little is known about the fine regulation of some of these intracellular interactions and how they are perturbed in diseases. To start filling this gap by identifying uncharacterized intracellular networks preferentially associated with ß-DG, HEK-293 cells were transiently transfected with a plasmid carrying the ß-DG subunit with GFP fused at its C-terminus. With this strategy, we aimed at forcing ß-DG to occupy multiple intracellular locations instead of sitting tightly at its canonical plasma membrane milieu, where it is commonly found in association with α-DG. Immunoprecipitation by anti-GFP antibodies followed by shotgun proteomic analysis led to the identification of an interactome formed by 313 exclusive protein matches for ß-DG binding. A series of already known ß-DG interactors have been found, including ezrin and emerin, whilst significant new matches, which include potential novel ß-DG interactors and their related networks, were identified in diverse subcellular compartments, such as cytoskeleton, endoplasmic reticulum/Golgi, mitochondria, nuclear membrane and the nucleus itself. Of particular interest amongst the novel identified matches, Lamina-Associated Polypeptide-1B (LAP1B), an inner nuclear membrane protein, whose mutations are known to cause nuclear envelopathies characterized by muscular dystrophy, was found to interact with ß-DG in HEK-293 cells. This evidence was confirmed by immunoprecipitation, Western blotting and immunofluorescence experiments. We also found by immunofluorescence experiments that LAP1B looses its nuclear envelope localization in C2C12 DG-knock-out cells, suggesting that LAP1B requires ß-DG for a proper nuclear localization. These results expand the role of ß-DG as a nuclear scaffolding protein and provide novel evidence of a possible link between dystroglycanopathies and nuclear envelopathies displaying with muscular dystrophy.


Subject(s)
Dystroglycans , Muscular Dystrophies , Humans , Dystroglycans/chemistry , HEK293 Cells , Proteomics , Muscular Dystrophies/metabolism , Nuclear Envelope/metabolism
3.
Glycobiology ; 31(5): 649-661, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33295623

ABSTRACT

We have carried out a comparative study of the conformational impact of modifications to threonine residues of either α-O-Man or α-O-GalNAc in the context of a sequence from the mucin-like region of α-dystroglycan. Both such modifications can coexist in this domain of the glycoprotein. Solution NMR experiments and molecular dynamics calculations were employed. Comparing the results for an unmodified peptide Ac- PPTTTTKKP-NH2 sequence from α-dystroglycan, and glycoconjugates with either modification on the Ts, we find that the impact of the α-O-Man modification on the peptide scaffold is quite limited, while that of the α-O-GalNAc is more profound. The results for the α-O-GalNAc glycoconjugate are consistent with what has been seen earlier in other systems. Further examination of the NMR-based structure and the MD results suggest a more extensive network of hydrogen bond interactions within the α-O-GalNAc-threonine residue than has been previously appreciated, which influences the properties of the protein backbone. The conformational effects are relevant to the mechanical properties of α-dystroglycan.


Subject(s)
Dystroglycans/chemistry , Glycoproteins/chemistry , Dystroglycans/metabolism , Glycoproteins/metabolism , Molecular Conformation , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Processing, Post-Translational
4.
Biochem Biophys Res Commun ; 579: 8-14, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34583196

ABSTRACT

α-Dystroglycan (α-DG) is a glycoprotein specifically modified with O-mannosyl glycans bearing long polysaccharides, termed matriglycans, which comprise repeating units of glucuronic acid and xylose. The matriglycan is linked to the O-mannosyl glycan core through two ribitol phosphate units that can be replaced with glycerol phosphate (GroP) units synthesized by fukutin and fukutin-related protein that transfer GroP from CDP-Gro. Here, we found that forced expression of the bacterial CDP-Gro synthase, TagD, from Bacillus subtilis could result in the overproduction of CDP-Gro in human colon carcinoma HCT116 cells. Western blot and liquid chromatography-tandem mass spectrometry analyses indicated that α-DG prepared from the TagD-expressing HCT116 cells contained abundant GroP and lacked matriglycans. Using the GroP-containing recombinant α-DG-Fc, we developed a novel monoclonal antibody, termed DG2, that reacts with several truncated glycoforms of α-DG, including GroP-terminated glycoforms lacking matriglycans; we verified the reactivity of DG2 against various types of knockout cells deficient in the biosynthesis of matriglycans. Accordingly, forced expression of TagD in HCT116 cells resulted in the reduction of matriglycans and an increase in DG2 reactivity. Collectively, our results indicate that DG2 could serve as a useful tool to determine tissue distribution and function of α-DG lacking matriglycans under physiological and pathophysiological conditions.


Subject(s)
Antibodies, Monoclonal/chemistry , Dystroglycans/chemistry , Laminin/chemistry , Protein Isoforms/chemistry , Animals , Bacillus subtilis , CRISPR-Cas Systems , Chromatography, Liquid , DNA, Complementary/metabolism , Female , Glucuronic Acid/chemistry , Glycopeptides/chemistry , HCT116 Cells , Humans , Mass Spectrometry , Mice , Mice, Inbred BALB C , Phosphates , Polysaccharides , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Ribitol/chemistry , Xylose
5.
Molecules ; 26(21)2021 Nov 04.
Article in English | MEDLINE | ID: mdl-34771084

ABSTRACT

Bacteria contain glycerol phosphate (GroP)-containing glycans, which are important constituents of cell-surface glycopolymers such as the teichoic acids of Gram-positive bacterial cell walls. These glycopolymers comprising GroP play crucial roles in bacterial physiology and virulence. Recently, the first identification of a GroP-containing glycan in mammals was reported as a variant form of O-mannosyl glycan on α-dystroglycan (α-DG). However, the biological significance of such GroP modification remains largely unknown. In this review, we provide an overview of this new discovery of GroP-containing glycan in mammals and then outline the recent progress in elucidating the biosynthetic mechanisms of GroP-containing glycans on α-DG. In addition, we discuss the potential biological role of GroP modification along with the challenges and prospects for further research. The progress in this newly identified glycan modification will provide insights into the phylogenetic implications of glycan.


Subject(s)
Glycerophosphates/metabolism , Polysaccharides/biosynthesis , Animals , Biosynthetic Pathways , Dystroglycans/chemistry , Dystroglycans/metabolism , Extracellular Matrix/metabolism , Glycerophosphates/chemistry , Glycosylation , Humans , Laminin/metabolism , Mammals , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Polysaccharides/chemistry , Protein Binding , Structure-Activity Relationship
6.
Glycobiology ; 30(10): 817-829, 2020 09 28.
Article in English | MEDLINE | ID: mdl-32149355

ABSTRACT

Mutations in multiple genes required for proper O-mannosylation of α-dystroglycan are causal for congenital/limb-girdle muscular dystrophies and abnormal brain development in mammals. Previously, we and others further elucidated the functional O-mannose glycan structure that is terminated by matriglycan, [(-GlcA-ß3-Xyl-α3-)n]. This repeating disaccharide serves as a receptor for proteins in the extracellular matrix. Here, we demonstrate in vitro that HNK-1 sulfotransferase (HNK-1ST/carbohydrate sulfotransferase) sulfates terminal glucuronyl residues of matriglycan at the 3-hydroxyl and prevents further matriglycan polymerization by the LARGE1 glycosyltransferase. While α-dystroglycan isolated from mouse heart and kidney is susceptible to exoglycosidase digestion of matriglycan, the functional, lower molecular weight α-dystroglycan detected in brain, where HNK-1ST expression is elevated, is resistant. Removal of the sulfate cap by a sulfatase facilitated dual-glycosidase digestion. Our data strongly support a tissue specific mechanism in which HNK-1ST regulates polymer length by competing with LARGE for the 3-position on the nonreducing GlcA of matriglycan.


Subject(s)
Dystroglycans/metabolism , Glucuronic Acid/metabolism , Sulfotransferases/metabolism , Animals , Dystroglycans/chemistry , Glucuronic Acid/chemistry , Glycosylation , Mice , Sulfotransferases/chemistry , Sulfotransferases/isolation & purification
7.
Glycoconj J ; 37(1): 77-93, 2020 02.
Article in English | MEDLINE | ID: mdl-31823246

ABSTRACT

Dystroglycanopathies are diseases characterized by progressive muscular degeneration and impairment of patient's quality of life. They are associated with altered glycosylation of the dystrophin-glycoprotein (DGC) complex components, such as α-dystroglycan (α-DG), fundamental in the structural and functional stability of the muscle fiber. The diagnosis of dystroglycanopathies is currently based on the observation of clinical manifestations, muscle biopsies and enzymatic measures, and the available monoclonal antibodies are not specific for the dystrophic hypoglycosylated muscle condition. Thus, modified α-DG mucins have been considered potential targets for the development of new diagnostic strategies toward these diseases. In this context, this work describes the synthesis of the hypoglycosylated α-DG mimetic glycopeptide NHAc-Gly-Pro-Thr-Val-Thr[αMan]-Ile-Arg-Gly-BSA (1) as a potential tool for the development of novel antibodies applicable to dystroglycanopathies diagnosis. Glycopeptide 1 was used for the development of polyclonal antibodies and recombinant monoclonal antibodies by Phage Display technology. Accordingly, polyclonal antibodies were reactive to glycopeptide 1, which enables the application of anti-glycopeptide 1 antibodies in immune reactive assays targeting hypoglycosylated α-DG. Regarding monoclonal antibodies, for the first time variable heavy (VH) and variable light (VL) immunoglobulin domains were selected by Phage Display, identified by NGS and described by in silico analysis. The best-characterized VH and VL domains were cloned, expressed in E. coli Shuffle T7 cells, and used to construct a single chain fragment variable that recognized the Glycopeptide 1 (GpαDG1 scFv). Molecular modelling of glycopeptide 1 and GpαDG1 scFv suggested that their interaction occurs through hydrogen bonds and hydrophobic contacts involving amino acids from scFv (I51, Y33, S229, Y235, and P233) and R8 and α-mannose from Glycopeptide 1.


Subject(s)
Antibodies, Monoclonal/immunology , Dystroglycans/immunology , Glycoproteins/immunology , Mucins/immunology , Walker-Warburg Syndrome/diagnosis , Dystroglycans/chemistry , Glycoproteins/chemical synthesis , Humans , Mucins/chemistry
8.
J Chem Inf Model ; 60(6): 3145-3156, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32356985

ABSTRACT

The acetylglucosaminyltransferase-like protein LARGE1 is an enzyme that is responsible for the final steps of the post-translational modifications of dystroglycan (DG), a membrane receptor that links the cytoskeleton with the extracellular matrix in the skeletal muscle and in a variety of other tissues. LARGE1 acts by adding the repeating disaccharide unit [-3Xyl-α1,3GlcAß1-] to the extracellular portion of the DG complex (α-DG); defects in the LARGE1 gene result in an aberrant glycosylation of α-DG and consequent impairment of its binding to laminin, eventually affecting the connection between the cell and the extracellular environment. In the skeletal muscle, this leads to degeneration of the muscular tissue and muscular dystrophy. So far, a few missense mutations have been identified within the LARGE1 protein and linked to congenital muscular dystrophy, and because no structural information is available on this enzyme, our understanding of the molecular mechanisms underlying these pathologies is still very limited. Here, we generated a 3D model structure of the two catalytic domains of LARGE1, combining different molecular modeling approaches. Furthermore, by using molecular dynamics simulations, we analyzed the effect on the structure and stability of the first catalytic domain of the pathological missense mutation S331F that gives rise to a severe form of muscle-eye-brain disease.


Subject(s)
Dystroglycans , Laminin , N-Acetylglucosaminyltransferases/chemistry , Animals , Dystroglycans/chemistry , Glycosylation , Laminin/chemistry , Mice , Molecular Dynamics Simulation , Protein Processing, Post-Translational
9.
Int J Mol Sci ; 21(17)2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32824881

ABSTRACT

ß-dystroglycan (ß-DG) assembles with lamins A/C and B1 and emerin at the nuclear envelope (NE) to maintain proper nuclear architecture and function. To provide insight into the nuclear function of ß-DG, we characterized the interaction between ß-DG and emerin at the molecular level. Emerin is a major NE protein that regulates multiple nuclear processes and whose deficiency results in Emery-Dreifuss muscular dystrophy (EDMD). Using truncated variants of ß-DG and emerin, via a series of in vitro and in vivo binding experiments and a tailored computational analysis, we determined that the ß-DG-emerin interaction is mediated at least in part by their respective transmembrane domains (TM). Using surface plasmon resonance assays we showed that emerin binds to ß-DG with high affinity (KD in the nanomolar range). Remarkably, the analysis of cells in which DG was knocked out demonstrated that loss of ß-DG resulted in a decreased emerin stability and impairment of emerin-mediated processes. ß-DG and emerin are reciprocally required for their optimal targeting within the NE, as shown by immunofluorescence, western blotting and immunoprecipitation assays using emerin variants with mutations in the TM domain and B-lymphocytes of a patient with EDMD. In summary, we demonstrated that ß-DG plays a role as an emerin interacting partner modulating its stability and function.


Subject(s)
Dystroglycans/metabolism , Membrane Proteins/metabolism , Muscular Dystrophy, Emery-Dreifuss/metabolism , Nuclear Proteins/metabolism , Active Transport, Cell Nucleus , Animals , B-Lymphocytes/metabolism , Binding Sites , Cell Line , Cells, Cultured , Dystroglycans/chemistry , Dystroglycans/genetics , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation , Nuclear Envelope/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding
10.
Nature ; 503(7474): 136-40, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24132234

ABSTRACT

The dense glycan coat that surrounds every cell is essential for cellular development and physiological function, and it is becoming appreciated that its composition is highly dynamic. Post-translational addition of the polysaccharide repeating unit [-3-xylose-α1,3-glucuronic acid-ß1-]n by like-acetylglucosaminyltransferase (LARGE) is required for the glycoprotein dystroglycan to function as a receptor for proteins in the extracellular matrix. Reductions in the amount of [-3-xylose-α1,3-glucuronic acid-ß1-]n (hereafter referred to as LARGE-glycan) on dystroglycan result in heterogeneous forms of muscular dystrophy. However, neither patient nor mouse studies has revealed a clear correlation between glycosylation status and phenotype. This disparity can be attributed to our lack of knowledge of the cellular function of the LARGE-glycan repeat. Here we show that coordinated upregulation of Large and dystroglycan in differentiating mouse muscle facilitates rapid extension of LARGE-glycan repeat chains. Using synthesized LARGE-glycan repeats we show a direct correlation between LARGE-glycan extension and its binding capacity for extracellular matrix ligands. Blocking Large upregulation during muscle regeneration results in the synthesis of dystroglycan with minimal LARGE-glycan repeats in association with a less compact basement membrane, immature neuromuscular junctions and dysfunctional muscle predisposed to dystrophy. This was consistent with the finding that patients with increased clinical severity of disease have fewer LARGE-glycan repeats. Our results reveal that the LARGE-glycan of dystroglycan serves as a tunable extracellular matrix protein scaffold, the extension of which is required for normal skeletal muscle function.


Subject(s)
Dystroglycans/chemistry , Dystroglycans/metabolism , Extracellular Matrix/metabolism , Muscular Dystrophies/prevention & control , N-Acetylglucosaminyltransferases/metabolism , Polysaccharides/metabolism , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Differentiation , Cell Line , Extracellular Matrix/chemistry , Female , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Molecular Weight , Muscle Development , Muscles/metabolism , Muscles/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Myoblasts , N-Acetylglucosaminyltransferases/deficiency , N-Acetylglucosaminyltransferases/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Phenotype , Polysaccharides/chemistry
11.
Proc Natl Acad Sci U S A ; 113(33): 9280-5, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27493216

ABSTRACT

The dystrophin glycoprotein complex, which connects the cell membrane to the basement membrane, is essential for a variety of biological events, including maintenance of muscle integrity. An O-mannose-type GalNAc-ß1,3-GlcNAc-ß1,4-(phosphate-6)-Man structure of α-dystroglycan (α-DG), a subunit of the complex that is anchored to the cell membrane, interacts directly with laminin in the basement membrane. Reduced glycosylation of α-DG is linked to some types of inherited muscular dystrophy; consistent with this relationship, many disease-related mutations have been detected in genes involved in O-mannosyl glycan synthesis. Defects in protein O-linked mannose ß1,2-N-acetylglucosaminyltransferase 1 (POMGnT1), a glycosyltransferase that participates in the formation of GlcNAc-ß1,2-Man glycan, are causally related to muscle-eye-brain disease (MEB), a congenital muscular dystrophy, although the role of POMGnT1 in postphosphoryl modification of GalNAc-ß1,3-GlcNAc-ß1,4-(phosphate-6)-Man glycan remains elusive. Our crystal structures of POMGnT1 agreed with our previous results showing that the catalytic domain recognizes substrate O-mannosylated proteins via hydrophobic interactions with little sequence specificity. Unexpectedly, we found that the stem domain recognizes the ß-linked GlcNAc of O-mannosyl glycan, an enzymatic product of POMGnT1. This interaction may recruit POMGnT1 to a specific site of α-DG to promote GlcNAc-ß1,2-Man clustering and also may recruit other enzymes that interact with POMGnT1, e.g., fukutin, which is required for further modification of the GalNAc-ß1,3-GlcNAc-ß1,4-(phosphate-6)-Man glycan. On the basis of our findings, we propose a mechanism for the deficiency in postphosphoryl modification of the glycan observed in POMGnT1-KO mice and MEB patients.


Subject(s)
Dystroglycans/chemistry , N-Acetylglucosaminyltransferases/chemistry , Binding Sites , Crystallization , Glycosylation , Humans , Mannose/chemistry
12.
Genes Cells ; 22(4): 348-359, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28251761

ABSTRACT

Orchestration of the multiple enzymes engaged in O-mannose glycan synthesis provides a matriglycan on α-dystroglycan (α-DG) which attracts extracellular matrix (ECM) proteins such as laminin. Aberrant O-mannosylation of α-DG leads to severe congenital muscular dystrophies due to detachment of ECM proteins from the basal membrane. Phosphorylation at C6-position of O-mannose catalyzed by protein O-mannosyl kinase (POMK) is a crucial step in the biosynthetic pathway of O-mannose glycan. Several mis-sense mutations of the POMK catalytic domain are known to cause a severe congenital muscular dystrophy, Walker-Warburg syndrome. Due to the low sequence similarity with other typical kinases, structure-activity relationships of this enzyme remain unclear. Here, we report the crystal structures of the POMK catalytic domain in the absence and presence of an ATP analogue and O-mannosylated glycopeptide. The POMK catalytic domain shows a typical protein kinase fold consisting of N- and C-lobes. Mannose residue binds to POMK mainly via the hydroxyl group at C2-position, differentiating from other monosaccharide residues. Intriguingly, the two amino acid residues K92 and D228, interacting with the triphosphate group of ATP, are donated from atypical positions in the primary structure. Mutations in this protein causing muscular dystrophies can now be rationalized.


Subject(s)
Protein Kinases/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Dystroglycans/chemistry , Humans , Mice , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Mutation , Protein Kinases/genetics , Protein Kinases/metabolism
13.
Nat Chem Biol ; 12(10): 810-4, 2016 10.
Article in English | MEDLINE | ID: mdl-27526028

ABSTRACT

Dystroglycan is a highly glycosylated extracellular matrix receptor with essential functions in skeletal muscle and the nervous system. Reduced matrix binding by α-dystroglycan (α-DG) due to perturbed glycosylation is a pathological feature of several forms of muscular dystrophy. Like-acetylglucosaminyltransferase (LARGE) synthesizes the matrix-binding heteropolysaccharide [-glucuronic acid-ß1,3-xylose-α1,3-]n. Using a dual exoglycosidase digestion, we confirm that this polysaccharide is present on native α-DG from skeletal muscle. The atomic details of matrix binding were revealed by a high-resolution crystal structure of laminin-G-like (LG) domains 4 and 5 (LG4 and LG5) of laminin-α2 bound to a LARGE-synthesized oligosaccharide. A single glucuronic acid-ß1,3-xylose disaccharide repeat straddles a Ca(2+) ion in the LG4 domain, with oxygen atoms from both sugars replacing Ca(2+)-bound water molecules. The chelating binding mode accounts for the high affinity of this protein-carbohydrate interaction. These results reveal a previously uncharacterized mechanism of carbohydrate recognition and provide a structural framework for elucidating the mechanisms underlying muscular dystrophy.


Subject(s)
Dystroglycans/chemistry , Laminin/chemistry , Binding Sites , Models, Molecular , Molecular Structure
14.
Mol Cell Proteomics ; 15(11): 3424-3434, 2016 11.
Article in English | MEDLINE | ID: mdl-27601598

ABSTRACT

Dystroglycanopathy is a major class of congenital muscular dystrophy caused by a deficiency of functional glycans on α-dystroglycan (αDG) with laminin-binding activity. Recent advances have led to identification of several causative gene products of dystroglycanopathy and characterization of their in vitro enzymatic activities. However, the in vivo functional roles remain equivocal for enzymes such as ISPD, FKTN, FKRP, and TMEM5 that are supposed to be involved in post-phosphoryl modifications linking the GalNAc-ß3-GlcNAc-ß4-Man-6-phosphate core and the outer laminin-binding glycans. Herein, by direct nano-LC-MS2/MS3 analysis of tryptic glycopeptides derived from a truncated recombinant αDG expressed in the wild-type and a panel of mutated cells deficient in one of these enzymes, we sought to define the full extent of variable modifications on this phosphorylated core O-glycan at the functional Thr317/Thr319 sites. We showed that the most abundant glycoforms carried a phosphorylated core at each of the two sites, with and without a single ribitol phosphate (RboP) extending from terminal HexNAc. At much lower signal intensity, a novel substituent tentatively assigned as glycerol phosphate (GroP) was additionally detected. As expected, tandem RboP extended with a GlcA-Xyl unit was only identified in wild type, whereas knocking out of either ISPD or FKTN prevented formation of RboP. In the absence of FKRP, glycoforms with single but not tandem RboP accumulated, consistent with the suggested role of this enzyme in transferring the second RboP. Intriguingly, the single GroP modification also required functional FKTN whereas absence of TMEM5 significantly hindered only the addition of RboP. Our findings thus revealed additional levels of complexity associated with the core structures, suggesting functional interplay among these enzymes through their interactions. The simplified analytical workflow developed here should facilitate rapid mapping across a wider range of cell types to gain better insights into its physiological relevance.


Subject(s)
Dystroglycans/chemistry , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Walker-Warburg Syndrome/genetics , Gene Knockout Techniques , Genetic Predisposition to Disease , HCT116 Cells , HEK293 Cells , Humans , Pentosyltransferases , Phosphorylation , Proteins/genetics
15.
Angew Chem Int Ed Engl ; 57(29): 9003-9007, 2018 07 16.
Article in English | MEDLINE | ID: mdl-29802667

ABSTRACT

O-Mannose glycans account up to 30 % of total O-glycans in the brain. Previous synthesis and functional studies have only focused on the core M3 O-mannose glycans of α-dystroglycan, which are a causative factor for various muscular diseases. In this study, a highly efficient chemoenzymatic strategy was developed that enabled the first collective synthesis of 63 core M1 and core M2 O-mannose glycans. This chemoenzymatic strategy features the gram-scale chemical synthesis of five judiciously designed core structures, and the diversity-oriented modification of the core structures with three enzyme modules to provide 58 complex O-mannose glycans in a linear sequence that does not exceed four steps. The binding profiles of synthetic O-mannose glycans with a panel of lectins, antibodies, and brain proteins were also explored by using a printed O-mannose glycan array.


Subject(s)
Mannose/chemistry , Polysaccharides/chemistry , Animals , Biocatalysis , Chemistry Techniques, Synthetic , Dystroglycans/chemical synthesis , Dystroglycans/chemistry , Glycosylation , Glycosyltransferases/chemistry , Humans , Mannose/chemical synthesis , Polysaccharides/chemical synthesis
16.
J Biol Chem ; 291(47): 24618-24627, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27733679

ABSTRACT

A defect in O-mannosyl glycan is the cause of α-dystroglycanopathy, a group of congenital muscular dystrophies caused by aberrant α-dystroglycan (α-DG) glycosylation. Recently, the entire structure of O-mannosyl glycan, [3GlcAß1-3Xylα1]n-3GlcAß1-4Xyl-Rbo5P-1Rbo5P-3GalNAcß1-3GlcNAcß1-4 (phospho-6)Manα1-, which is required for the binding of α-DG to extracellular matrix ligands, has been proposed. However, the linkage of the first Xyl residue to ribitol 5-phosphate (Rbo5P) is not clear. TMEM5 is a gene product responsible for α-dystroglycanopathy and was reported as a potential enzyme involved in this linkage formation, although the experimental evidence is still incomplete. Here, we report that TMEM5 is a xylosyltransferase that forms the Xylß1-4Rbo5P linkage on O-mannosyl glycan. The anomeric configuration and linkage position of the product (ß1,4 linkage) was determined by NMR analysis. The introduction of two missense mutations in TMEM5 found in α-dystroglycanopathy patients impaired xylosyltransferase activity. Furthermore, the disruption of the TMEM5 gene by CRISPR/Cas9 abrogated the elongation of the (-3GlcAß1-3Xylα1-) unit on O-mannosyl glycan. Based on these results, we concluded that TMEM5 acts as a UDP-d-xylose:ribitol-5-phosphate ß1,4-xylosyltransferase in the biosynthetic pathway of O-mannosyl glycan.


Subject(s)
Dystroglycans/metabolism , Membrane Proteins/metabolism , Muscular Dystrophies/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism , Cell Line , Dystroglycans/chemistry , Dystroglycans/genetics , Glycosylation , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Muscular Dystrophies/genetics , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , Pentosyltransferases , Transferases (Other Substituted Phosphate Groups)/chemistry , Transferases (Other Substituted Phosphate Groups)/genetics
17.
Glycobiology ; 27(9): 806-819, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28810660

ABSTRACT

The post-translational glycosylation of select proteins by O-linked mannose (O-mannose or O-man) is a conserved modification from yeast to humans and has been shown to be necessary for proper development and growth. The most well studied O-mannosylated mammalian protein is α-dystroglycan (α-DG). Hypoglycosylation of α-DG results in varying severities of congenital muscular dystrophies, cancer progression and metastasis, and inhibited entry and infection of certain arenaviruses. Defects in the gene products responsible for post-translational modification of α-DG, primarily glycosyltransferases, are the basis for these diseases. The multitude of clinical phenotypes resulting from defective O-mannosylation highlights the biomedical significance of this unique modification. Elucidation of the various O-mannose biosynthetic pathways is imperative to understanding a broad range of human diseases and for the development of novel therapeutics. In this review, we will focus on recent discoveries delineating the various enzymes, structures and functions associated with O-mannose-initiated glycoproteins. Additionally, we discuss current gaps in our knowledge of mammalian O-mannosylation, discuss the evolution of this pathway, and illustrate the utility and limitations of model systems to study functions of O-mannosylation.


Subject(s)
Dystroglycans/chemistry , Glycosyltransferases/metabolism , Mannose/metabolism , Muscular Dystrophies/metabolism , Neoplasms/metabolism , Protein Processing, Post-Translational , Animals , Arenavirus/metabolism , Dystroglycans/genetics , Dystroglycans/metabolism , Evolution, Molecular , Glycosylation , Glycosyltransferases/genetics , Humans , Mammals , Mannose/chemistry , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Neoplasms/genetics , Neoplasms/pathology , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism
18.
Biochim Biophys Acta Gen Subj ; 1861(10): 2462-2472, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28711406

ABSTRACT

BACKGROUND: O-mannosyl glycans have been found in a limited number of glycoproteins of the brain, nerves, and skeletal muscles, particularly in α-dystroglycan (α-DG). Defects in O-mannosyl glycan on α-DG are the primary cause of a group of congenital muscular dystrophies, which are collectively termed α-dystroglycanopathy. Recent studies have revealed various O-mannosyl glycan structures, which can be classified as core M1, core M2, and core M3 glycans. Although many dystroglycanopathy genes are involved in core M3 processing, the structure and biosynthesis of core M3 glycan remains only partially understood. SCOPE OF REVIEW: This review presents recent findings about the structure, biosynthesis, and pathology of O-mannosyl glycans. MAJOR CONCLUSIONS: Recent studies have revealed that the entire structure of core M3 glycan, including ribitol-5-phosphate, is a novel structure in mammals; its unique biosynthetic pathway has been elucidated by the identification of new causative genes for α-dystroglycanopathies and their functions. GENERAL SIGNIFICANCE: O-mannosyl glycan has a novel, unique structure that is important for the maintenance of brain and muscle functions. These findings have opened up a new field in glycoscience. These studies will further contribute to the understanding of the pathomechanism of α-dystroglycanopathy and the development of glycotherapeutics. This article is part of a Special Issue entitled Neuro-glycoscience, edited by Kenji Kadomatsu and Hiroshi Kitagawa.


Subject(s)
Dystroglycans/chemistry , Muscular Dystrophies/metabolism , N-Acetylglucosaminyltransferases/chemistry , Pentosephosphates/metabolism , Protein Processing, Post-Translational , Walker-Warburg Syndrome/metabolism , Animals , Brain/metabolism , Brain/pathology , Carbohydrate Sequence , Dystroglycans/genetics , Dystroglycans/metabolism , Glycosylation , Humans , Mannose/chemistry , Mannose/metabolism , Models, Molecular , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Pentosephosphates/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/pathology
19.
Hum Mol Genet ; 23(25): 6697-711, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25082828

ABSTRACT

Mutations in dystrophin lead to Duchenne muscular dystrophy, which is among the most common human genetic disorders. Dystrophin nucleates assembly of the dystrophin-glycoprotein complex (DGC), and a defective DGC disrupts an essential link between the intracellular cytoskeleton and the basal lamina, leading to progressive muscle wasting. In vitro studies have suggested that dystrophin phosphorylation may affect interactions with actin or syntrophin, yet whether this occurs in vivo or affects protein function remains unknown. Utilizing nanoflow liquid chromatography mass spectrometry, we identified 18 phosphorylated residues within endogenous dystrophin. Mutagenesis revealed that phosphorylation at S3059 enhances the dystrophin-dystroglycan interaction and 3D modeling utilizing the Rosetta software program provided a structural model for how phosphorylation enhances this interaction. These findings demonstrate that phosphorylation is a key mechanism regulating the interaction between dystrophin and the DGC and reveal that posttranslational modification of a single amino acid directly modulates the function of dystrophin.


Subject(s)
Dystroglycans/metabolism , Dystrophin-Associated Proteins/metabolism , Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Cell Line , Cysteine/chemistry , Cysteine/metabolism , Dystroglycans/chemistry , Dystroglycans/genetics , Dystrophin/chemistry , Dystrophin/genetics , Dystrophin-Associated Proteins/chemistry , Dystrophin-Associated Proteins/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Myoblasts/cytology , Myoblasts/metabolism , Phosphorylation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Serine/metabolism , Signal Transduction
20.
J Biol Chem ; 289(40): 27585-603, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25157101

ABSTRACT

α-Neurexins (α-Nrxn) are mostly presynaptic cell surface molecules essential for neurotransmission that are linked to neuro-developmental disorders as autism or schizophrenia. Several interaction partners of α-Nrxn are identified that depend on alternative splicing, including neuroligins (Nlgn) and dystroglycan (αDAG). The trans-synaptic complex with Nlgn1 was extensively characterized and shown to partially mediate α-Nrxn function. However, the interactions of α-Nrxn with αDAG, neurexophilins (Nxph1) and Nlgn2, ligands that occur specifically at inhibitory synapses, are incompletely understood. Using site-directed mutagenesis, we demonstrate the exact binding epitopes of αDAG and Nxph1 on Nrxn1α and show that their binding is mutually exclusive. Identification of an unusual cysteine bridge pattern and complex type glycans in Nxph1 ensure binding to the second laminin/neurexin/sex hormone binding (LNS2) domain of Nrxn1α, but this association does not interfere with Nlgn binding at LNS6. αDAG, in contrast, interacts with both LNS2 and LNS6 domains without inserts in splice sites SS#2 or SS#4 mostly via LARGE (like-acetylglucosaminyltransferase)-dependent glycans attached to the mucin region. Unexpectedly, binding of αDAG at LNS2 prevents interaction of Nlgn at LNS6 with or without splice insert in SS#4, presumably by sterically hindering each other in the u-form conformation of α-Nrxn. Thus, expression of αDAG and Nxph1 together with alternative splicing in Nrxn1α may prevent or facilitate formation of distinct trans-synaptic Nrxn·Nlgn complexes, revealing an unanticipated way to contribute to the identity of synaptic subpopulations.


Subject(s)
Brain/metabolism , Dystroglycans/metabolism , Glycoproteins/metabolism , Neuropeptides/metabolism , Alternative Splicing , Animals , Dystroglycans/chemistry , Dystroglycans/genetics , Glycoproteins/genetics , Humans , Ligands , Mice , Neuropeptides/genetics , Protein Binding , Protein Structure, Tertiary , Rats , Synapses/genetics , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL