Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Elife ; 52016 04 29.
Article in English | MEDLINE | ID: mdl-27130732

ABSTRACT

Multiple glycosyltransferases are essential for the proper modification of alpha-dystroglycan, as mutations in the encoding genes cause congenital/limb-girdle muscular dystrophies. Here we elucidate further the structure of an O-mannose-initiated glycan on alpha-dystroglycan that is required to generate its extracellular matrix-binding polysaccharide. This functional glycan contains a novel ribitol structure that links a phosphotrisaccharide to xylose. ISPD is a CDP-ribitol (ribose) pyrophosphorylase that generates the reduced sugar nucleotide for the insertion of ribitol in a phosphodiester linkage to the glycoprotein. TMEM5 is a UDP-xylosyl transferase that elaborates the structure. We demonstrate in a zebrafish model as well as in a human patient that defects in TMEM5 result in muscular dystrophy in combination with abnormal brain development. Thus, we propose a novel structure-a ribitol in a phosphodiester linkage-for the moiety on which TMEM5, B4GAT1, and LARGE act to generate the functional receptor for ECM proteins having LG domains.


Subject(s)
Dystroglycans/chemistry , Dystroglycans/metabolism , Extracellular Matrix/metabolism , Membrane Proteins/metabolism , Polysaccharides/analysis , Animals , Humans , Mannose/analysis , Nucleotidyltransferases/metabolism , Pentosyltransferases , Protein Binding , Ribitol/analysis , Zebrafish
2.
J Proteome Res ; 13(7): 3314-29, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24884609

ABSTRACT

Inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4) is a 120 kDa acute-phase glycoprotein produced primarily in the liver, secreted into the blood, and identified in serum. ITIH4 is involved in liver development and stabilization of the extracellular matrix (ECM), and its expression is altered in liver disease. In this study, we aimed to characterize glycosylation of recombinant and serum-derived ITIH4 using analytical mass spectrometry. Recombinant ITIH4 was analyzed to optimize glycopeptide analyses, followed by serum-derived ITIH4. First, we confirmed that the four ITIH4 N-X-S/T sequons (N81, N207, N517, and N577) were glycosylated by treating ITIH4 tryptic/GluC glycopeptides with PNGaseF in the presence of (18)O water. Next, we performed glycosidase-assisted LC-MS/MS analysis of ITIH4 trypsin-GluC glycopeptides enriched via hydrophilic interaction liquid chromatography to characterize ITIH4 N-glycoforms. While microheterogeneity of N-glycoforms differed between ITIH4 protein expressed in HEK293 cells and protein isolated from serum, occupancy of N-glycosylation sites did not differ. A fifth N-glycosylation site was discovered at N274 with the rare nonconsensus NVV motif. Site N274 contained high-mannose N-linked glycans in both serum and recombinant ITIH4. We also identified isoform-specific ITIH4 O-glycoforms and documented that utilization of O-glycosylation sites on ITIH4 differed between the cell line and serum.


Subject(s)
Glycoproteins/blood , Protein Processing, Post-Translational , Proteinase Inhibitory Proteins, Secretory/blood , Amino Acid Sequence , Blood Proteins/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Glycoproteins/chemistry , Glycosylation , HEK293 Cells , Humans , Molecular Sequence Data , Protein Isoforms , Proteinase Inhibitory Proteins, Secretory/chemistry
3.
Methods Mol Biol ; 951: 229-44, 2013.
Article in English | MEDLINE | ID: mdl-23296534

ABSTRACT

As a complex post-translational event, the biosynthesis, structures, and functions of O-linked glycans have attracted research interests in various aspects. The recent development of novel technologies for the analysis of glycans and glycoproteins sheds new insights with regard to determining site occupancy, structure-function relationships, and the contributions of O-linked glycosylation to physiological and pathological processes. In this chapter, we refer to several approaches for the structural characterization and quantification of O-linked glycopeptides, with a focus on O-GlcNAc and O-Mannose modified glycoproteins.


Subject(s)
Glycopeptides/analysis , Glycopeptides/chemistry , Oxygen/chemistry , Acetylglucosamine/metabolism , Amino Acid Sequence , Binding Sites , Chromatography, Affinity , Chromatography, Liquid , Dithiothreitol/chemistry , Formates/chemistry , Glycopeptides/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycosylation , Mannose/metabolism , Mass Spectrometry , Oxidation-Reduction , Phosphoric Monoester Hydrolases/metabolism , Proteolysis , Sulfhydryl Compounds/chemistry , Tandem Mass Spectrometry , Trypsin/metabolism
4.
Cell Mol Life Sci ; 70(16): 2849-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23115008

ABSTRACT

Glycosylation of proteins is arguably the most prevalent co- and post-translational modification. It is responsible for increased heterogeneity and functional diversity of proteins. Here we discuss the importance of one type of glycosylation, specifically O-mannosylation and its relationship to a number of human diseases. The most widely studied O-mannose modified protein is alpha-dystroglycan (α-DG). Recent studies have focused intensely on α-DG due to the severity of diseases associated with its improper glycosylation. O-mannosylation of α-DG is involved in cancer metastasis, arenavirus entry, and multiple forms of congenital muscular dystrophy [1, 2]. In this review, we discuss the structural and functional characteristics of O-mannose-initiated glycan structures on α-DG, enzymes involved in the O-mannosylation pathway, and the diseases that are a direct result of disruptions within this pathway.


Subject(s)
Arenaviridae Infections/metabolism , Dystroglycans/metabolism , Mannose/metabolism , Muscular Dystrophies/metabolism , Neoplasms/metabolism , Animals , Arenaviridae Infections/genetics , Dystroglycans/chemistry , Dystroglycans/genetics , Glycosylation , Humans , Mannose/chemistry , Mannose/genetics , Muscular Dystrophies/genetics , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology
5.
J Biol Chem ; 287(25): 20967-74, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22549772

ABSTRACT

O-Linked glycosylation is a functionally and structurally diverse type of protein modification present in many tissues and across many species. α-Dystroglycan (α-DG), a protein linked to the extracellular matrix, whose glycosylation status is associated with human muscular dystrophies, displays two predominant types of O-glycosylation, O-linked mannose (O-Man) and O-linked N-acetylgalactosamine (O-GalNAc), in its highly conserved mucin-like domain. The O-Man is installed by an enzyme complex present in the endoplasmic reticulum. O-GalNAc modifications are initiated subsequently in the Golgi apparatus by the UDP-GalNAc polypeptide N-acetylgalactosaminyltransferase (ppGalNAc-T) enzymes. How the presence and position of O-Man influences the action of the ppGalNAc-Ts on α-DG and the distribution of the two forms of glycosylation in this domain is not known. Here, we investigated the interplay between O-Man and the addition of O-GalNAc by examining the activity of the ppGalNAc-Ts on peptides and O-Man-containing glycopeptides mimicking those found in native α-DG. These synthetic glycopeptides emulate intermediate structures, not otherwise readily available from natural sources. Through enzymatic and mass spectrometric methods, we demonstrate that the presence and specific location of O-Man can impact either the regional exclusion or the site of O-GalNAc addition on α-DG, elucidating the factors contributing to the glycosylation patterns observed in vivo. These results provide evidence that one form of glycosylation can influence another form of glycosylation in α-DG and suggest that in the absence of proper O-mannosylation, as is associated with certain forms of muscular dystrophy, aberrant O-GalNAc modifications may occur and could play a role in disease presentation.


Subject(s)
Acetylgalactosamine/metabolism , Dystroglycans/metabolism , Mannose/metabolism , Multienzyme Complexes/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Acetylgalactosamine/genetics , Animals , Cell Line , Dystroglycans/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Glycosylation , Humans , Mannose/genetics , Mice , Multienzyme Complexes/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , N-Acetylgalactosaminyltransferases/genetics , Uridine Diphosphate N-Acetylgalactosamine/genetics
6.
J Biol Chem ; 287(27): 22759-70, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22570487

ABSTRACT

Duchenne muscular dystrophy is an X-linked disorder characterized by loss of dystrophin, a cytoskeletal protein that connects the actin cytoskeleton in skeletal muscle cells to extracellular matrix. Dystrophin binds to the cytoplasmic domain of the transmembrane glycoprotein ß-dystroglycan (ß-DG), which associates with cell surface α-dystroglycan (α-DG) that binds laminin in the extracellular matrix. ß-DG can also associate with utrophin, and this differential association correlates with specific glycosylation changes on α-DG. Genetic modification of α-DG glycosylation can promote utrophin binding and rescue dystrophic phenotypes in mouse dystrophy models. We used high throughput screening with the plant lectin Wisteria floribunda agglutinin (WFA) to identify compounds that altered muscle cell surface glycosylation, with the goal of finding compounds that increase abundance of α-DG and associated sarcolemmal glycoproteins, increase utrophin usage, and increase laminin binding. We identified one compound, lobeline, from the Prestwick library of Food and Drug Administration-approved compounds that fulfilled these criteria, increasing WFA binding to C2C12 cells and to primary muscle cells from wild type and mdx mice. WFA binding and enhancement by lobeline required complex N-glycans but not O-mannose glycans that bind laminin. However, inhibiting complex N-glycan processing reduced laminin binding to muscle cell glycoproteins, although O-mannosylation was intact. Glycan analysis demonstrated a general increase in N-glycans on lobeline-treated cells rather than specific alterations in cell surface glycosylation, consistent with increased abundance of multiple sarcolemmal glycoproteins. This demonstrates the feasibility of high throughput screening with plant lectins to identify compounds that alter muscle cell glycosylation and identifies a novel role for N-glycans in regulating muscle cell function.


Subject(s)
Glycomics/methods , Laminin/metabolism , Myoblasts, Skeletal/metabolism , Polysaccharides/metabolism , Quadriceps Muscle/metabolism , Sarcolemma/metabolism , Animals , Cells, Cultured , Glycomics/instrumentation , Glycosylation , Lobeline/pharmacology , Male , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Nicotinic Agonists/pharmacology , Plant Lectins/pharmacology , Quadriceps Muscle/cytology , RNA, Small Interfering/genetics , Receptors, N-Acetylglucosamine , Sarcolemma/drug effects
7.
Curr Opin Struct Biol ; 21(5): 603-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21945038

ABSTRACT

Post-translational modification of polypeptides with glycans increases the diversity of the structures of proteins and imparts increased functional diversity. Here, we review the current literature on a relatively new O-glycosylation pathway, the mammalian O-mannosylation pathway. The importance of O-mannosylation is illustrated by the fact that O-mannose glycan structures play roles in a variety of processes including viral entry into cells, metastasis, cell adhesion, and neuronal development. Furthermore, mutations in the enzymes of this pathway are causal for a variety of congenital muscular dystrophies. Here we highlight the protein substrates, glycan structures, and enzymes involved in O-mannosylation as well as our gaps in understanding structure/function relationships in this biosynthetic pathway.


Subject(s)
Dystroglycans/metabolism , Mannose/metabolism , Polysaccharides/metabolism , Protein Processing, Post-Translational , Animals , Brain/abnormalities , Dystroglycans/genetics , Dystrophin/genetics , Dystrophin/metabolism , Glycosylation , Humans , Mammals , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/physiopathology , Mutation , Structure-Activity Relationship
8.
J Am Chem Soc ; 133(36): 14418-30, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21812486

ABSTRACT

Aberrant glycosylation of α-dystroglycan (α-DG) results in loss of interactions with the extracellular matrix and is central to the pathogenesis of several disorders. To examine protein glycosylation of α-DG, a facile synthetic approach has been developed for the preparation of unusual phosphorylated O-mannosyl glycopeptides derived from α-DG by a strategy in which properly protected phospho-mannosides are coupled with a Fmoc protected threonine derivative, followed by the use of the resulting derivatives in automated solid-phase glycopeptide synthesis using hyper-acid-sensitive Sieber amide resin. Synthetic efforts also provided a reduced phospho-trisaccharide, and the NMR data of this derivative confirmed the proper structural assignment of the unusual phospho-glycan structure. The glycopeptides made it possible to explore factors that regulate the elaboration of critical glycans. It was established that a glycopeptide having a 6-phospho-O-mannosyl residue is not an acceptor for action by the enzyme POMGnT1, which attaches ß(1,2)-GlcNAc to O-mannosyl moietes, whereas the unphosphorylated derivate was readily extended by the enzyme. This finding implies a specific sequence of events in determining the structural fate of the O-glycan. It has also been found that the activity of POMGnT1 is dependent on the location of the acceptor site in the context of the underlying polypeptide/glycopeptide sequence. Conformational analysis by NMR has shown that the O-mannosyl modification does not exert major conformational effect on the peptide backbone. It is, however, proposed that these residues, introduced at the early stages of glycoprotein glycosylation, have an ability to regulate the loci of subsequent O-GalNAc additions, which do exert conformational effects. The studies show that through access to discrete glycopeptide structures, it is possible to reveal complex regulation of O-glycan processing on α-DG that has significant implications both for its normal post-translational maturation, and the mechanisms of the pathologies associated with hypoglycosylated α-DG.


Subject(s)
Dystroglycans/chemistry , Glycopeptides/chemistry , Phosphoproteins/chemistry , Glycopeptides/biosynthesis , Glycopeptides/chemical synthesis , Nuclear Magnetic Resonance, Biomolecular , Phosphoproteins/biosynthesis , Phosphoproteins/chemical synthesis , Phosphorylation , Protein Conformation
9.
J Biol Chem ; 286(24): 21180-90, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21460210

ABSTRACT

Dystroglycanopathies are a subset of congenital muscular dystrophies wherein α-dystroglycan (α-DG) is hypoglycosylated. α-DG is an extensively O-glycosylated extracellular matrix-binding protein and a key component of the dystrophin-glycoprotein complex. Previous studies have shown α-DG to be post-translationally modified by both O-GalNAc- and O-mannose-initiated glycan structures. Mutations in defined or putative glycosyltransferase genes involved in O-mannosylation are associated with a loss of ligand-binding activity of α-DG and are causal for various forms of congenital muscular dystrophy. In this study, we sought to perform glycomic analysis on brain O-linked glycan structures released from proteins of three different knock-out mouse models associated with O-mannosylation (POMGnT1, LARGE (Myd), and DAG1(-/-)). Using mass spectrometry approaches, we were able to identify nine O-mannose-initiated and 25 O-GalNAc-initiated glycan structures in wild-type littermate control mouse brains. Through our analysis, we were able to confirm that POMGnT1 is essential for the extension of all observed O-mannose glycan structures with ß1,2-linked GlcNAc. Loss of LARGE expression in the Myd mouse had no observable effect on the O-mannose-initiated glycan structures characterized here. Interestingly, we also determined that similar amounts of O-mannose-initiated glycan structures are present on brain proteins from α-DG-lacking mice (DAG1) compared with wild-type mice, indicating that there must be additional proteins that are O-mannosylated in the mammalian brain. Our findings illustrate that classical ß1,2-elongation and ß1,6-GlcNAc branching of O-mannose glycan structures are dependent upon the POMGnT1 enzyme and that O-mannosylation is not limited solely to α-DG in the brain.


Subject(s)
Glycomics/methods , Muscular Dystrophies/metabolism , N-Acetylglucosaminyltransferases/genetics , Animals , Brain/metabolism , Carbohydrates/chemistry , Disease Models, Animal , Dystroglycans/chemistry , Galactosyltransferases/chemistry , Glycosylation , Mannose/chemistry , Mice , Mice, Knockout , Muscular Dystrophies/congenital , Mutation , Polysaccharides/chemistry
10.
Phytochemistry ; 71(17-18): 1963-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20970816

ABSTRACT

Understanding the biochemical mechanisms by which plants respond to microbial infection is a fundamental goal of plant science. Extracellular dermal glycoproteins (EDGPs) are widely expressed in plant tissues and have been implicated in plant defense responses. Although EDGPs are known to interact with fungal proteins, the downstream effects of these interactions are poorly understood. To gain insight into these phenomena, we used tobacco floral nectar as a model system to identify a mechanism by which the EDGP known as Nectarin IV (NEC4) functions as pathogen surveillance molecule. Our data demonstrates that the interaction of NEC4 with a fungal endoglucanase (XEG) promotes the catalytic activity of Nectarin V (NEC5), which catalyzes the conversion of glucose and molecular oxygen to gluconic acid and H(2)O(2). Significantly enhanced NEC5 activity was observed when XEG was added to nectar or nectarin solutions that contain NEC4. This response was also observed when the purified NEC4:XEG complex was added to NEC4-depleted nectarin solutions, which did not respond to XEG alone. These results indicate that formation of the NEC4:XEG complex is a key step leading to induction of NEC5 activity in floral nectar, resulting in an increase in concentrations of reactive oxygen species (ROS), which are known to inhibit microbial growth directly and activate signal transduction pathways that induce innate immunity responses in the plant.


Subject(s)
Fungal Proteins/metabolism , Glucose Oxidase/metabolism , Glycoproteins/metabolism , Nicotiana/chemistry , Plant Nectar/metabolism , Plant Proteins/metabolism , Cellulase/antagonists & inhibitors , Gene Expression Regulation, Plant , Glucans/analysis , Glucans/metabolism , Gluconates/metabolism , Glucose Oxidase/drug effects , Plant Nectar/chemistry , Reactive Oxygen Species/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Nicotiana/enzymology , Xylans/analysis , Xylans/metabolism
11.
J Biol Chem ; 285(32): 24882-91, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20507986

ABSTRACT

The main extracellular matrix binding component of the dystrophin-glycoprotein complex, alpha-dystroglycan (alpha-DG), which was originally isolated from rabbit skeletal muscle, is an extensively O-glycosylated protein. Previous studies have shown alpha-DG to be modified by both O-GalNAc- and O-mannose-initiated glycan structures. O-Mannosylation, which accounts for up to 30% of the reported O-linked structures in certain tissues, has been rarely observed on mammalian proteins. Mutations in multiple genes encoding defined or putative glycosyltransferases involved in O-mannosylation are causal for various forms of congenital muscular dystrophy. Here, we explore the glycosylation of purified rabbit skeletal muscle alpha-DG in detail. Using tandem mass spectrometry approaches, we identify 4 O-mannose-initiated and 17 O-GalNAc-initiated structures on alpha-DG isolated from rabbit skeletal muscle. Additionally, we demonstrate the use of tandem mass spectrometry-based workflows to directly analyze glycopeptides generated from the purified protein. By combining glycomics and tandem mass spectrometry analysis of 91 glycopeptides from alpha-DG, we were able to assign 21 different residues as being modified by O-glycosylation with differing degrees of microheterogeneity; 9 sites of O-mannosylation and 14 sites of O-GalNAcylation were observed with only two sites definitively exhibiting occupancy by either type of glycan. The distribution of identified sites of O-mannosylation suggests a limited role for local primary sequence in dictating sites of attachment.


Subject(s)
Dystroglycans/chemistry , Muscle, Skeletal/metabolism , Polysaccharides/chemistry , Animals , Carbohydrates/chemistry , Glycoconjugates/chemistry , Glycomics/methods , Glycoproteins/metabolism , Glycosylation , Laminin/chemistry , Mannose/chemistry , Mass Spectrometry/methods , Mice , Rabbits , Surface Plasmon Resonance/methods
12.
Science ; 327(5961): 88-92, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20044576

ABSTRACT

Alpha-dystroglycan (alpha-DG) is a cell-surface glycoprotein that acts as a receptor for both extracellular matrix proteins containing laminin-G domains and certain arenaviruses. Receptor binding is thought to be mediated by a posttranslational modification, and defective binding with laminin underlies a subclass of congenital muscular dystrophy. Using mass spectrometry- and nuclear magnetic resonance (NMR)-based structural analyses, we identified a phosphorylated O-mannosyl glycan on the mucin-like domain of recombinant alpha-DG, which was required for laminin binding. We demonstrated that patients with muscle-eye-brain disease and Fukuyama congenital muscular dystrophy, as well as mice with myodystrophy, commonly have defects in a postphosphoryl modification of this phosphorylated O-linked mannose, and that this modification is mediated by the like-acetylglucosaminyltransferase (LARGE) protein. These findings expand our understanding of the mechanisms that underlie congenital muscular dystrophy.


Subject(s)
Dystroglycans/metabolism , Laminin/metabolism , Mannose/metabolism , Animals , Carbohydrate Conformation , Cell Line , Dystroglycans/chemistry , Glycosylation , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , Muscular Dystrophy, Animal/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
13.
Glycobiology ; 20(3): 381-94, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19969597

ABSTRACT

Recent studies highlighted an emerging possibility of using Drosophila as a model system for investigating the mechanisms of human congenital muscular dystrophies, called dystroglycanopathies, resulting from the abnormal glycosylation of alpha-dystroglycan. Several of these diseases are associated with defects in O-mannosylation, one of the most prominent types of alpha-dystroglycan glycosylation mediated by two protein O-mannosyltransferases. Drosophila appears to possess homologs of all essential components of the mammalian dystroglycan-mediated pathway; however, the glycosylation of Drosophila Dystroglycan (DG) has not yet been explored. In this study, we characterized the glycosylation of Drosophila DG using a combination of glycosidase treatments, lectin blots, trypsin digestion, and mass spectrometry analyses. Our results demonstrated that DG extracellular domain is O-mannosylated in vivo. We found that the concurrent in vivo activity of the two Drosophila protein O-mannosyltransferases, Rotated Abdomen and Twisted, is required for O-mannosylation of DG. While our experiments unambiguously determined some O-mannose sites far outside of the mucin-type domain of DG, they also provided evidence that DG bears a significant amount of O-mannosylation within its central region including the mucin-type domain, and that O-mannose can compete with O-GalNAc glycosylation of DG. We found that Rotated Abdomen and Twisted could potentiate in vivo the dominant-negative effect of DG extracellular domain expression on crossvein development, which suggests that O-mannosylation can modulate the ligand-binding activity of DG. Taken together these results demonstrated that O-mannosylation of Dystroglycan is an evolutionarily ancient mechanism conserved between Drosophila and humans, suggesting that Drosophila can be a suitable model system for studying molecular and genetic mechanisms underlying human dystroglycanopathies.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/enzymology , Dystroglycans/metabolism , Mannosyltransferases/metabolism , Animals , Drosophila/metabolism , Drosophila Proteins/chemistry , Dystroglycans/chemistry , Mannosyltransferases/chemistry , Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...