Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Proc Natl Acad Sci U S A ; 121(27): e2314026121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917011

ABSTRACT

The fucosylation of glycoproteins regulates diverse physiological processes. Inhibitors that can control cellular levels of protein fucosylation have consequently emerged as being of high interest. One area where inhibitors of fucosylation have gained significant attention is in the production of afucosylated antibodies, which exhibit superior antibody-dependent cell cytotoxicity as compared to their fucosylated counterparts. Here, we describe ß-carbafucose, a fucose derivative in which the endocyclic ring oxygen is replaced by a methylene group, and show that it acts as a potent metabolic inhibitor within cells to antagonize protein fucosylation. ß-carbafucose is assimilated by the fucose salvage pathway to form GDP-carbafucose which, due to its being unable to form the oxocarbenium ion-like transition states used by fucosyltransferases, is an incompetent substrate for these enzymes. ß-carbafucose treatment of a CHO cell line used for high-level production of the therapeutic antibody Herceptin leads to dose-dependent reductions in core fucosylation without affecting cell growth or antibody production. Mass spectrometry analyses of the intact antibody and N-glycans show that ß-carbafucose is not incorporated into the antibody N-glycans at detectable levels. We expect that ß-carbafucose will serve as a useful research tool for the community and may find immediate application for the rapid production of afucosylated antibodies for therapeutic purposes.


Subject(s)
Cricetulus , Fucose , Fucose/metabolism , Animals , CHO Cells , Glycosylation , Humans , Trastuzumab/pharmacology , Trastuzumab/metabolism , Fucosyltransferases/metabolism , Antibody-Dependent Cell Cytotoxicity/drug effects
2.
Curr Opin Chem Biol ; 77: 102403, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37856901

ABSTRACT

Within their native milieu of the cell, the activities of enzymes are controlled by a range of factors including protein interactions and post-translational modifications. The involvement of these factors in fundamental cell biology and the etiology of diseases is stimulating interest in monitoring enzyme activities within tissues. The creation of synthetic substrates, and their use with different imaging modalities, to detect and quantify enzyme activities has great potential to propel these areas of research. Here we describe the latest developments relating to the creation of substrates for imaging and quantifying the activities of glycoside hydrolases, focusing on mammalian systems. The limitations of current tools and the difficulties within the field are summarised, as are prospects for overcoming these challenges.


Subject(s)
Glycoside Hydrolases , Mammals , Animals , Glycoside Hydrolases/metabolism , Substrate Specificity
3.
Angew Chem Int Ed Engl ; 62(40): e202309306, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37582679

ABSTRACT

Fluorogenic substrates are emerging tools that enable studying enzymatic processes within their native cellular environments. However, fluorogenic substrates that function within live cells are generally incompatible with cellular fixation, preventing their tandem application with fundamental cell biology methods such as immunocytochemistry. Here we report a simple approach to enable the chemical fixation of a dark-to-light substrate, LysoFix-GBA, which enables quantification of glucocerebrosidase (GCase) activity in both live and fixed cells. LysoFix-GBA enables measuring responses to both chemical and genetic perturbations to lysosomal GCase activity. Further, LysoFix-GBA permits simple multiplexed co-localization studies of GCase activity with subcellular protein markers. This tool will aid studying the role of GCase activity in Parkinson's Disease, creating new therapeutic approaches targeting the GCase pathway. This approach also lays the foundation for an approach to create fixable substrates for other lysosomal enzymes.


Subject(s)
Glucosylceramidase , Parkinson Disease , Humans , Glucosylceramidase/metabolism , Fluorescence , Fluorescent Dyes/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Lysosomes/metabolism , Mutation
4.
Proc Natl Acad Sci U S A ; 119(29): e2200553119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858317

ABSTRACT

Loss of activity of the lysosomal glycosidase ß-glucocerebrosidase (GCase) causes the lysosomal storage disease Gaucher disease (GD) and has emerged as the greatest genetic risk factor for the development of both Parkinson disease (PD) and dementia with Lewy bodies. There is significant interest into how GCase dysfunction contributes to these diseases, however, progress toward a full understanding is complicated by presence of endogenous cellular factors that influence lysosomal GCase activity. Indeed, such factors are thought to contribute to the high degree of variable penetrance of GBA mutations among patients. Robust methods to quantitatively measure GCase activity within lysosomes are therefore needed to advance research in this area, as well as to develop clinical assays to monitor disease progression and assess GCase-directed therapeutics. Here, we report a selective fluorescence-quenched substrate, LysoFQ-GBA, which enables measuring endogenous levels of lysosomal GCase activity within living cells. LysoFQ-GBA is a sensitive tool for studying chemical or genetic perturbations of GCase activity using either fluorescence microscopy or flow cytometry. We validate the quantitative nature of measurements made with LysoFQ-GBA using various cell types and demonstrate that it accurately reports on both target engagement by GCase inhibitors and the GBA allele status of cells. Furthermore, through comparisons of GD, PD, and control patient-derived tissues, we show there is a close correlation in the lysosomal GCase activity within monocytes, neuronal progenitor cells, and neurons. Accordingly, analysis of clinical blood samples using LysoFQ-GBA may provide a surrogate marker of lysosomal GCase activity in neuronal tissue.


Subject(s)
Gaucher Disease , Glucosylceramidase , Parkinson Disease , Gaucher Disease/enzymology , Gaucher Disease/genetics , Glucosylceramidase/analysis , Glucosylceramidase/genetics , Humans , Lewy Bodies/enzymology , Lewy Body Disease/enzymology , Lysosomes/enzymology , Mutation , Parkinson Disease/enzymology , Parkinson Disease/genetics , Substrate Specificity , alpha-Synuclein/metabolism
5.
Biochem Biophys Res Commun ; 617(Pt 1): 16-21, 2022 08 20.
Article in English | MEDLINE | ID: mdl-35667241

ABSTRACT

The CMP-sialic acid synthetase (CSS) activates free sialic acid (Sia) to CMP-Sia using CTP, and is prerequisite for the sialylation of cell surface glycoconjugates. The vertebrate CSS consists of two domains, a catalytic N-domain and a non-catalytic C-domain. Although the C-domain is not required for the CSS enzyme to synthesize CMP-Sia, its involvement in the catalytic activity remains unknown. First, the real-time monitoring of CSS-catalyzed reaction was performed by 31P NMR using the rainbow trout CSS (rtCSS). While a rtCSS lacking the C-domain (rtCSS-N) similarly activated both deaminoneuraminic acid (Kdn) and N-acetylneuraminic acid (Neu5Ac), the full-length rtCSS (rtCSS-FL) did not activate Kdn as efficiently as Neu5Ac. These results suggest that the C-domain of rtCSS affects the enzymatic activity, when Kdn was used as a substrate. Second, the enzymatic activity of rtCSS-FL and rtCSS-N was measured under various concentrations of CMP-Kdn. Inhibition by CMP-Kdn was observed only for rtCSS-FL, but not for rtCSS-N, suggesting that the inhibition was C-domain-dependent. Third, the inhibitory effect of CMP-Kdn was also investigated using the mouse CSS (mCSS). However, no inhibition was observed with mCSS even at high concentrations of CMP-Kdn. Taken together, the data demonstrated that the C-domain is involved in the CMP-Kdn-dependent inhibition of rtCSS, which is a novel regulation of the Sia metabolism in rainbow trout.


Subject(s)
N-Acylneuraminate Cytidylyltransferase , Oncorhynchus mykiss , Animals , Cytidine Monophosphate/analogs & derivatives , Mice , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/metabolism , Neuraminic Acids , Sialic Acids/metabolism
6.
Nat Chem Biol ; 18(3): 332-341, 2022 03.
Article in English | MEDLINE | ID: mdl-35210619

ABSTRACT

Understanding the function and regulation of enzymes within their physiologically relevant milieu requires quality tools that report on their cellular activities. Here we describe a strategy for glycoside hydrolases that overcomes several limitations in the field, enabling quantitative monitoring of their activities within live cells. We detail the design and synthesis of bright and modularly assembled bis-acetal-based (BAB) fluorescence-quenched substrates, illustrating this strategy for sensitive quantitation of disease-relevant human α-galactosidase and α-N-acetylgalactosaminidase activities. We show that these substrates can be used within live patient cells to precisely measure the engagement of target enzymes by inhibitors and the efficiency of pharmacological chaperones, and highlight the importance of quantifying activity within cells using chemical perturbogens of cellular trafficking and lysosomal homeostasis. These BAB substrates should prove widely useful for interrogating the regulation of glycosidases within cells as well as in facilitating the development of therapeutics and diagnostics for this important class of enzymes.


Subject(s)
Acetals , Lysosomes , Fluorescence , Glycoside Hydrolases , Humans , alpha-Galactosidase
7.
Org Biomol Chem ; 19(37): 8057-8062, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34494637

ABSTRACT

We report the rational design and synthesis of C2-modified DGJ analogues to improve the selective inhibition of human GALA over other glycosidases. We prepare these analogues using a concise route from non-carbohydrate materials and demonstrate the most selective inhibitor 7c (∼100-fold) can act in Fabry patient cells to drive reductions in levels of the disease-relevant glycolipid Gb3.


Subject(s)
alpha-Galactosidase
8.
J Med Chem ; 63(15): 8231-8249, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32608236

ABSTRACT

Sialin, encoded by the SLC17A5 gene, is a lysosomal sialic acid transporter defective in Salla disease, a rare inherited leukodystrophy. It also enables metabolic incorporation of exogenous sialic acids, leading to autoantibodies against N-glycolylneuraminic acid in humans. Here, we identified a novel class of human sialin ligands by virtual screening and structure-activity relationship studies. The ligand scaffold is characterized by an amino acid backbone with a free carboxylate, an N-linked aromatic or heteroaromatic substituent, and a hydrophobic side chain. The most potent compound, 45 (LSP12-3129), inhibited N-acetylneuraminic acid 1 (Neu5Ac) transport in a non-competitive manner with IC50 ≈ 2.5 µM, a value 400-fold lower than the KM for Neu5Ac. In vitro and molecular docking studies attributed the non-competitive character to selective inhibitor binding to the Neu5Ac site in a cytosol-facing conformation. Moreover, compound 45 rescued the trafficking defect of the pathogenic mutant (R39C) causing Salla disease. This new class of cell-permeant inhibitors provides tools to investigate the physiological roles of sialin and help develop pharmacological chaperones for Salla disease.


Subject(s)
Amino Acids/chemistry , Amino Acids/metabolism , Lysosomes/metabolism , Organic Anion Transporters/metabolism , Symporters/metabolism , Animals , Dose-Response Relationship, Drug , HEK293 Cells , HeLa Cells , Humans , Ligands , Molecular Docking Simulation/methods , Protein Structure, Secondary , Rats
9.
Int J Mol Sci ; 20(3)2019 Jan 31.
Article in English | MEDLINE | ID: mdl-30709055

ABSTRACT

The mammalian mono-α2,8-sialyltransferase ST8Sia VI has been shown to catalyze the transfer of a unique sialic acid residues onto core 1 O-glycans leading to the formation of di-sialylated O-glycosylproteins and to a lesser extent to diSia motifs onto glycolipids like GD1a. Previous studies also reported the identification of an orthologue of the ST8SIA6 gene in the zebrafish genome. Trying to get insights into the biosynthesis and function of the oligo-sialylated glycoproteins during zebrafish development, we cloned and studied this fish α2,8-sialyltransferase homologue. In situ hybridization experiments demonstrate that expression of this gene is always detectable during zebrafish development both in the central nervous system and in non-neuronal tissues. Intriguingly, using biochemical approaches and the newly developed in vitro MicroPlate Sialyltransferase Assay (MPSA), we found that the zebrafish recombinant enzyme does not synthetize diSia motifs on glycoproteins or glycolipids as the human homologue does. Using comparative genomics and molecular phylogeny approaches, we show in this work that the human ST8Sia VI orthologue has disappeared in the ray-finned fish and that the homologue described in fish correspond to a new subfamily of α2,8-sialyltransferase named ST8Sia VIII that was not maintained in Chondrichtyes and Sarcopterygii.


Subject(s)
Sialyltransferases/genetics , Sialyltransferases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Animals , COS Cells , Central Nervous System/metabolism , Chlorocebus aethiops , Computer Simulation , Evolution, Molecular , Gene Expression Regulation, Developmental , Glycolipids/chemistry , Glycoproteins/chemistry , HEK293 Cells , Humans , Phylogeny , Sequence Homology, Nucleic Acid , Substrate Specificity , Tissue Distribution , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
10.
Chem Sci ; 9(39): 7585-7595, 2018 Oct 21.
Article in English | MEDLINE | ID: mdl-30393518

ABSTRACT

Glycans form one of the four classes of biomolecules, are found in every living system and present a huge structural and functional diversity. As an illustration of this diversity, it has been reported that more than 50% of the human proteome is glycosylated and that 2% of the human genome is dedicated to glycosylation processes. Glycans are involved in many biological processes such as signalization, cell-cell or host pathogen interactions, immunity, etc. However, fundamental processes associated with glycans are not yet fully understood and the development of glycobiology is relatively recent compared to the study of genes or proteins. Approximately 25 years ago, the studies of Bertozzi's and Reutter's groups paved the way for metabolic oligosaccharide engineering (MOE), a strategy which consists in the use of modified sugar analogs which are taken up into the cells, metabolized, incorporated into glycoconjugates, and finally detected in a specific manner. This groundbreaking strategy has been widely used during the last few decades and the concomitant development of new bioorthogonal ligation reactions has allowed many advances in the field. Typically, MOE has been used to either visualize glycans or identify different classes of glycoproteins. The present review aims to highlight recent studies that lie somewhat outside of these more traditional approaches and that are pushing the boundaries of MOE applications.

11.
Bioconjug Chem ; 29(10): 3377-3384, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30192128

ABSTRACT

Mammalian sialyltransferases transfer sialic acids onto glycoproteins and glycolipids within the Golgi apparatus. Despite their key role in glycosylation, the study of their enzymatic activities is limited by the lack of appropriate tools. Herein, we developed a quick and sensitive sialyltransferase microplate assay based on the use of the unnatural CMP-SiaNAl donor substrate. In this assay, an appropriate acceptor glycoprotein is coated on the bottom of 96-well plate and the sialyltransferase activity is assessed using CMP-SiaNAl. The alkyne tag of SiaNAl enables subsequent covalent ligation of an azido-biotin probe via CuAAC and an antibiotin-HRP conjugated antibody is then used to quantify the amount of transferred SiaNAl by a colorimetric titration. With this test, we evaluated the kinetic characteristics and substrate preferences of two human sialyltransferases, ST6Gal I and ST3Gal I toward a panel of asialoglycoprotein acceptors, and identified cations that display a sialyltransferase inhibitory effect.


Subject(s)
Sialic Acids/metabolism , Sialyltransferases/metabolism , Biotin/chemistry , Carbon-13 Magnetic Resonance Spectroscopy , Chromatography, Liquid/methods , Colorimetry/methods , Glycoproteins/metabolism , HEK293 Cells , Horseradish Peroxidase/chemistry , Humans , Limit of Detection , Mass Spectrometry/methods , Proton Magnetic Resonance Spectroscopy , Sialyltransferases/chemistry , Substrate Specificity , beta-D-Galactoside alpha 2-6-Sialyltransferase , beta-Galactoside alpha-2,3-Sialyltransferase
12.
J Inherit Metab Dis ; 41(3): 515-523, 2018 05.
Article in English | MEDLINE | ID: mdl-29294191

ABSTRACT

The development of metabolic oligosaccharide engineering (MOE) over the past two decades enabled the bioimaging studies of glycosylation processes in physio-pathological contexts. Herein, we successfully applied the chemical reporter strategy to image the fate of sialylated glycoconjugates in healthy and sialin-deficient patient fibroblasts. This chemical glycomics enrichment is a powerful tool for tracking sialylated glycoconjugates and probing lysosomal recycling capacities. Thus, such strategies appear fundamental for the characterization of lysosomal storage diseases.


Subject(s)
Glycomics/methods , Metabolic Engineering/methods , N-Acetylneuraminic Acid/analysis , N-Acetylneuraminic Acid/metabolism , Oligosaccharides/metabolism , Single Molecule Imaging/methods , Case-Control Studies , Chemical Fractionation , Combinatorial Chemistry Techniques/methods , Humans , Lysosomal Storage Diseases/diagnosis , Lysosomal Storage Diseases/metabolism , Lysosomes/metabolism , Metabolic Networks and Pathways/physiology , Oligosaccharides/analysis , Oligosaccharides/chemistry
13.
Chembiochem ; 18(13): 1251-1259, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28395125

ABSTRACT

Sialylation of glycoproteins and glycolipids is catalyzed by sialyltransferases in the Golgi of mammalian cells, whereby sialic acid residues are added at the nonreducing ends of oligosaccharides. Because sialylated glycans play critical roles in a number of human physio-pathological processes, the past two decades have witnessed the development of modified sialic acid derivatives for a better understanding of sialic acid biology and for the development of new therapeutic targets. However, nothing is known about how individual mammalian sialyltransferases tolerate and behave towards these unnatural CMP-sialic acid donors. In this study, we devised several approaches to investigate the donor specificity of the human ß-d-galactoside sialyltransferases ST6Gal I and ST3Gal I by using two CMP-sialic acids: CMP-Neu5Ac, and CMP-Neu5N-(4pentynoyl)neuraminic acid (CMP-SiaNAl), an unnatural CMP-sialic acid donor with an extended and functionalized N-acyl moiety.


Subject(s)
Antigens, CD/metabolism , Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Cytidine Monophosphate/analogs & derivatives , Glycolipids/metabolism , Glycoproteins/metabolism , Polysaccharides/metabolism , Sialic Acids/metabolism , Sialyltransferases/metabolism , Antigens, CD/chemistry , Antigens, CD/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cloning, Molecular , Cytidine Monophosphate/chemistry , Cytidine Monophosphate/metabolism , Cytidine Monophosphate N-Acetylneuraminic Acid/chemistry , Gene Expression , Glycolipids/chemistry , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , HEK293 Cells , Humans , Kinetics , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/chemistry , Neisseria meningitidis/enzymology , Polysaccharides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sialic Acids/chemistry , Sialyltransferases/chemistry , Sialyltransferases/genetics , Substrate Specificity , beta-Galactoside alpha-2,3-Sialyltransferase
14.
Glycobiology ; 26(11): 1151-1156, 2016 11.
Article in English | MEDLINE | ID: mdl-27543325

ABSTRACT

Natural and synthetically modified cytidine monophosphate activated sialic acids (CMP-Sias) are essential research assets in the field of glycobiology: among other applications, they can be used to probe glycans, detect sialylation defects at the cell surface or carry out detailed studies of sialyltransferase activities. However, these chemical tools are notoriously unstable because of hydrolytic decomposition, and are very time-consuming and costly to obtain. They are nigh impossible to store with satisfactory purity, and their preparation requires multiple laborious purification steps that usually lead to heavy product loss. Using in situ time-resolved 31P phosphorus nuclear magnetic resonance (31P NMR), we precisely established the kinetics of formation and degradation of a number of CMP-Sias including CMP-Neu5Ac, CMP-Neu5Gc, CMP-SiaNAl and CMP-SiaNAz in several experimental conditions. 31P NMR can be carried out in undeuterated solvents and is a sensitive and nondestructive technique that allows for direct in situ monitoring and optimization of chemo-enzymatic syntheses that involve phosphorus-containing species. Thus, we showed that CMP-sialic acid derivatives can be robustly obtained in high yields using the readily available Neisseria meningitidis CMP-sialic acid synthase. This integrated workflow takes less than an hour, and the freshly prepared CMP-Sias can be directly transferred to sialylation biological assays without any purification step.


Subject(s)
Cytidine Monophosphate/chemistry , Molecular Probes/chemistry , Polysaccharides/analysis , Sialic Acids/chemistry , Cytidine Monophosphate/biosynthesis , Cytidine Monophosphate/chemical synthesis , Molecular Probes/biosynthesis , Molecular Probes/chemical synthesis , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/enzymology , Sialic Acids/biosynthesis , Sialic Acids/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL