Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Org Lett ; 26(3): 739-744, 2024 01 26.
Article in English | MEDLINE | ID: mdl-38215221

ABSTRACT

We demonstrate the use of the symmetrical diethyl(dimethyl)difluoromethylene bisphosphonate reagent for the synthesis of terminal and unsymmetrical difluoromethylene bisphosphonates, close analogues of biologically important molecules. The difference in reactivity of the methyl and ethyl groups in the symmetrical diethyl(dimthyl)difluoromethylene bisphosphonate is exploited in a stepwise demethylation-condensation sequence to functionalize either side of the reagent to allow the generation of a series of close bioisosteres of natural pyrophosphate molecules, including ADPr, CDP-glycerol and CDP-ribitol.


Subject(s)
Diphosphonates , Hydrocarbons, Fluorinated
3.
RSC Chem Biol ; 1(1): 13-25, 2020 Apr 16.
Article in English | MEDLINE | ID: mdl-34458745

ABSTRACT

UDP-N-acetylglucosamine pyrophosphorylase (UAP1) catalyses the last step in eukaryotic biosynthesis of uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc), converting UTP and GlcNAc-1P to the sugar nucleotide. Gene disruption studies have shown that this gene is essential in eukaryotes and a possible antifungal target, yet no inhibitors of fungal UAP1 have so far been reported. Here we describe the crystal structures of substrate/product complexes of UAP1 from Aspergillus fumigatus that together provide snapshots of catalysis. A structure with UDP-GlcNAc, pyrophosphate and Mg2+ provides the first Michaelis complex trapped for this class of enzyme, revealing the structural basis of the previously reported Mg2+ dependence and direct observation of pyrophosphorolysis. We also show that a highly conserved lysine mimics the role of a second metal observed in structures of bacterial orthologues. A mechanism-inspired UTP α,ß-methylenebisphosphonate analogue (meUTP) was designed and synthesized and was shown to be a micromolar inhibitor of the enzyme. The mechanistic insights and inhibitor described here will facilitate future studies towards the discovery of small molecule inhibitors of this currently unexploited potential antifungal drug target.

4.
Nat Struct Mol Biol ; 26(11): 1071-1077, 2019 11.
Article in English | MEDLINE | ID: mdl-31695185

ABSTRACT

Modification of specific Ser and Thr residues of nucleocytoplasmic proteins with O-GlcNAc, catalyzed by O-GlcNAc transferase (OGT), is an abundant posttranslational event essential for proper animal development and is dysregulated in various diseases. Due to the rapid concurrent removal by the single O-GlcNAcase (OGA), precise functional dissection of site-specific O-GlcNAc modification in vivo is currently not possible without affecting the entire O-GlcNAc proteome. Exploiting the fortuitous promiscuity of OGT, we show that S-GlcNAc is a hydrolytically stable and accurate structural mimic of O-GlcNAc that can be encoded in mammalian systems with CRISPR-Cas9 in an otherwise unperturbed O-GlcNAcome. Using this approach, we target an elusive Ser 405 O-GlcNAc site on OGA, showing that this site-specific modification affects OGA stability.


Subject(s)
Acetylglucosamine/metabolism , N-Acetylglucosaminyltransferases/metabolism , beta-N-Acetylhexosaminidases/metabolism , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/genetics , Animals , CRISPR-Cas Systems , Glycosylation , HEK293 Cells , Humans , Mice , Models, Molecular , N-Acetylglucosaminyltransferases/chemistry , Protein Processing, Post-Translational , Substrate Specificity , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/genetics
5.
Proc Natl Acad Sci U S A ; 116(30): 14961-14970, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31296563

ABSTRACT

O-GlcNAc transferase (OGT) is an X-linked gene product that is essential for normal development of the vertebrate embryo. It catalyses the O-GlcNAc posttranslational modification of nucleocytoplasmic proteins and proteolytic maturation of the transcriptional coregulator Host cell factor 1 (HCF1). Recent studies have suggested that conservative missense mutations distal to the OGT catalytic domain lead to X-linked intellectual disability in boys, but it is not clear if this is through changes in the O-GlcNAc proteome, loss of protein-protein interactions, or misprocessing of HCF1. Here, we report an OGT catalytic domain missense mutation in monozygotic female twins (c. X:70779215 T > A, p. N567K) with intellectual disability that allows dissection of these effects. The patients show limited IQ with developmental delay and skewed X-inactivation. Molecular analyses revealed decreased OGT stability and disruption of the substrate binding site, resulting in loss of catalytic activity. Editing this mutation into the Drosophila genome results in global changes in the O-GlcNAc proteome, while in mouse embryonic stem cells it leads to loss of O-GlcNAcase and delayed differentiation down the neuronal lineage. These data imply that catalytic deficiency of OGT could contribute to X-linked intellectual disability.


Subject(s)
Catalytic Domain , Genetic Diseases, X-Linked/genetics , Intellectual Disability/genetics , Loss of Function Mutation , N-Acetylglucosaminyltransferases/genetics , Animals , Cell Line , Drosophila , Female , Genetic Diseases, X-Linked/pathology , Host Cell Factor C1/metabolism , Humans , Intellectual Disability/pathology , Mice , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Neurogenesis , Point Mutation , Twins, Monozygotic
6.
J Biol Chem ; 293(46): 17754-17768, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30224358

ABSTRACT

O-Linked GlcNAc transferase (OGT) possesses dual glycosyltransferase-protease activities. OGT thereby stably glycosylates serines and threonines of numerous proteins and, via a transient glutamate glycosylation, cleaves a single known substrate-the so-called HCF-1PRO repeat of the transcriptional co-regulator host-cell factor 1 (HCF-1). Here, we probed the relationship between these distinct glycosylation and proteolytic activities. For proteolysis, the HCF-1PRO repeat possesses an important extended threonine-rich region that is tightly bound by the OGT tetratricopeptide-repeat (TPR) region. We report that linkage of this HCF-1PRO-repeat, threonine-rich region to heterologous substrate sequences also potentiates robust serine glycosylation with the otherwise poor Rp-αS-UDP-GlcNAc diastereomer phosphorothioate and UDP-5S-GlcNAc OGT co-substrates. Furthermore, it potentiated proteolysis of a non-HCF-1PRO-repeat cleavage sequence, provided it contained an appropriately positioned glutamate residue. Using serine- or glutamate-containing HCF-1PRO-repeat sequences, we show that proposed OGT-based or UDP-GlcNAc-based serine-acceptor residue activation mechanisms can be circumvented independently, but not when disrupted together. In contrast, disruption of both proposed activation mechanisms even in combination did not inhibit OGT-mediated proteolysis. These results reveal a multiplicity of OGT glycosylation strategies, some leading to proteolysis, which could be targets of alternative molecular regulatory strategies.


Subject(s)
Endopeptidases/metabolism , Host Cell Factor C1/metabolism , N-Acetylglucosaminyltransferases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Endopeptidases/genetics , Glycosylation , Host Cell Factor C1/genetics , Humans , Molecular Dynamics Simulation , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutation , N-Acetylglucosaminyltransferases/genetics , Proteolysis , Stereoisomerism , Substrate Specificity , Uridine Diphosphate N-Acetylglucosamine/analogs & derivatives , Uridine Diphosphate N-Acetylglucosamine/metabolism
7.
Bioconjug Chem ; 29(6): 1834-1840, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29723473

ABSTRACT

O-GlcNAc transferase (OGT) is an essential glycosyltransferase that installs the O-GlcNAc post-translational modification on the nucleocytoplasmic proteome. We report the development of S-linked UDP-peptide conjugates as potent bisubstrate OGT inhibitors. These compounds were assembled in a modular fashion by photoinitiated thiol-ene conjugation of allyl-UDP and optimal acceptor peptides in which the acceptor serine was replaced with cysteine. The conjugate VTPVC(S-propyl-UDP)TA ( Ki = 1.3 µM) inhibits the OGT activity in HeLa cell lysates. Linear fusions of this conjugate with cell penetrating peptides were explored as prototypes of cell-penetrant OGT inhibitors. A crystal structure of human OGT with the inhibitor revealed mimicry of the interactions seen in the pseudo-Michaelis complex. Furthermore, a fluorophore-tagged derivative of the inhibitor works as a high affinity probe in a fluorescence polarimetry hOGT assay.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Peptides/chemistry , Peptides/pharmacology , Uridine Diphosphate/analogs & derivatives , Uridine Diphosphate/pharmacology , Drug Design , HeLa Cells , Humans , Molecular Docking Simulation , N-Acetylglucosaminyltransferases/metabolism , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology
8.
ACS Chem Biol ; 13(5): 1353-1360, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29641181

ABSTRACT

The attachment of the sugar N-acetyl-D-glucosamine (GlcNAc) to specific serine and threonine residues on proteins is referred to as protein O-GlcNAcylation. O-GlcNAc transferase (OGT) is the enzyme responsible for carrying out the modification, while O-GlcNAcase (OGA) reverses it. Protein O-GlcNAcylation has been implicated in a wide range of cellular processes including transcription, proteostasis, and stress response. Dysregulation of O-GlcNAc has been linked to diabetes, cancer, and neurodegenerative and cardiovascular disease. OGA has been proposed to be a drug target for the treatment of Alzheimer's and cardiovascular disease given that increased O-GlcNAc levels appear to exert a protective effect. The search for specific, potent, and drug-like OGA inhibitors with bioavailability in the brain is therefore a field of active research, requiring orthogonal high-throughput assay platforms. Here, we describe the synthesis of a novel probe for use in a fluorescence polarization based assay for the discovery of inhibitors of OGA. We show that the probe is suitable for use with both human OGA, as well as the orthologous bacterial counterpart from Clostridium perfringens, CpOGA, and the lysosomal hexosaminidases HexA/B. We structurally characterize CpOGA in complex with a ligand identified from a fragment library screen using this assay. The versatile synthesis procedure could be adapted for making fluorescent probes for the assay of other glycoside hydrolases.


Subject(s)
Fluorescence Polarization/methods , N-Acetylglucosaminyltransferases/metabolism , Acetylglucosamine/metabolism , Crystallography, X-Ray , Humans , N-Acetylglucosaminyltransferases/chemistry , Proof of Concept Study , Protein Conformation , Substrate Specificity
9.
Open Biol ; 7(6)2017 06.
Article in English | MEDLINE | ID: mdl-28659383

ABSTRACT

O-linked N-acetylglucosamine (O-GlcNAc) is an essential and dynamic post-translational modification found on hundreds of nucleocytoplasmic proteins in metazoa. Although a single enzyme, O-GlcNAc transferase (OGT), generates the entire cytosolic O-GlcNAc proteome, it is not understood how it recognizes its protein substrates, targeting only a fraction of serines/threonines in the metazoan proteome for glycosylation. We describe a trapped complex of human OGT with the C-terminal domain of TAB1, a key innate immunity-signalling O-GlcNAc protein, revealing extensive interactions with the tetratricopeptide repeats of OGT. Confirmed by mutagenesis, this interaction suggests that glycosylation substrate specificity is achieved by recognition of a degenerate sequon in the active site combined with an extended conformation C-terminal of the O-GlcNAc target site.


Subject(s)
N-Acetylglucosaminyltransferases/metabolism , Tetratricopeptide Repeat/physiology , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Glycosylation , Humans , N-Acetylglucosaminyltransferases/genetics , Sequence Alignment , Substrate Specificity , Tetratricopeptide Repeat/genetics
10.
Genes Dev ; 30(8): 960-72, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27056667

ABSTRACT

In complex with the cosubstrate UDP-N-acetylglucosamine (UDP-GlcNAc),O-linked-GlcNAc transferase (OGT) catalyzes Ser/ThrO-GlcNAcylation of many cellular proteins and proteolysis of the transcriptional coregulator HCF-1. Such a dual glycosyltransferase-protease activity, which occurs in the same active site, is unprecedented and integrates both reversible and irreversible forms of protein post-translational modification within one enzyme. Although occurring within the same active site, we show here that glycosylation and proteolysis occur through separable mechanisms. OGT consists of tetratricopeptide repeat (TPR) and catalytic domains, which, together with UDP-GlcNAc, are required for both glycosylation and proteolysis. Nevertheless, a specific TPR domain contact with the HCF-1 substrate is critical for proteolysis but not Ser/Thr glycosylation. In contrast, key catalytic domain residues and even a UDP-GlcNAc oxygen important for Ser/Thr glycosylation are irrelevant for proteolysis. Thus, from a dual glycosyltransferase-protease, essentially single-activity enzymes can be engineered both in vitro and in vivo. Curiously, whereas OGT-mediated HCF-1 proteolysis is limited to vertebrate species, invertebrate OGTs can cleave human HCF-1. We present a model for the evolution of HCF-1 proteolysis by OGT.


Subject(s)
Host Cell Factor C1/metabolism , N-Acetylglucosaminyltransferases/metabolism , Proteolysis , Amino Acid Motifs , Animals , Catalytic Domain , Computer Simulation , Evolution, Molecular , Humans , Invertebrates/enzymology , Models, Molecular , Mutation , Protein Processing, Post-Translational , Protein Structure, Tertiary
11.
J Am Chem Soc ; 138(10): 3325-32, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26859322

ABSTRACT

The conversion of glycoside hydrolases (GHs) into transglycosylases (TGs), i.e., from enzymes that hydrolyze carbohydrates to enzymes that synthesize them, represents a promising solution for the large-scale synthesis of complex carbohydrates for biotechnological purposes. However, the lack of knowledge about the molecular details of transglycosylation hampers the rational design of TGs. Here we present the first crystallographic structure of a natural glycosyl-enzyme intermediate (GEI) of Saccharomyces cerevisiae Gas2 in complex with an acceptor substrate and demonstrate, by means of quantum mechanics/molecular mechanics metadynamics simulations, that it is tuned for transglycosylation (ΔG(⧧) = 12 kcal/mol). The 2-OH···nucleophile interaction is found to be essential for catalysis: its removal raises the free energy barrier significantly (11 and 16 kcal/mol for glycosylation and transglycosylation, respectively) and alters the conformational itinerary of the substrate (from (4)C1 → [(4)E](⧧) → (1,4)B/(4)E to (4)C1 → [(4)H3](⧧) → (4)C1). Our results suggest that changes in the interactions involving the 2-position could have an impact on the transglycosylation activity of several GHs.


Subject(s)
Glycoside Hydrolases/chemistry , Multienzyme Complexes/chemistry , Transferases/chemistry , Crystallography, X-Ray , Glycoside Hydrolases/metabolism , Glycosylation , Multienzyme Complexes/metabolism , Quantum Theory , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Thermodynamics , Transferases/metabolism
12.
Biochem J ; 470(2): 255-262, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26348912

ABSTRACT

O-GlcNAcylation is a reversible type of serine/threonine glycosylation on nucleocytoplasmic proteins in metazoa. Various genetic approaches in several animal models have revealed that O-GlcNAcylation is essential for embryogenesis. However, the dynamic changes in global O-GlcNAcylation and the underlying mechanistic biology linking them to embryonic development is not understood. One of the limiting factors towards characterizing changes in O-GlcNAcylation has been the limited specificity of currently available tools to detect this modification. In the present study, harnessing the unusual properties of an O-GlcNAcase (OGA) mutant that binds O-GlcNAc (O-N-acetylglucosamine) sites with nanomolar affinity, we uncover changes in protein O-GlcNAcylation as a function of Drosophila development.


Subject(s)
Bacterial Proteins/metabolism , Drosophila/metabolism , beta-N-Acetylhexosaminidases/metabolism , Acylation , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Proteins/genetics , Blotting, Far-Western , Clostridium perfringens/enzymology , Drosophila/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development , HEK293 Cells , Humans , Mutation , beta-N-Acetylhexosaminidases/genetics
13.
Nat Struct Mol Biol ; 22(9): 744-750, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26237509

ABSTRACT

O-GlcNAc transferase (OGT) glycosylates a diverse range of intracellular proteins with O-linked N-acetylglucosamine (O-GlcNAc), an essential and dynamic post-translational modification in metazoans. Although this enzyme modifies hundreds of proteins with O-GlcNAc, it is not understood how OGT achieves substrate specificity. In this study, we describe the application of a high-throughput OGT assay to a library of peptides. We mapped sites of O-GlcNAc modification by electron transfer dissociation MS and found that they correlate with previously detected O-GlcNAc sites. Crystal structures of four acceptor peptides in complex with Homo sapiens OGT suggest that a combination of size and conformational restriction defines sequence specificity in the -3 to +2 subsites. This work reveals that although the N-terminal TPR repeats of OGT may have roles in substrate recognition, the sequence restriction imposed by the peptide-binding site makes a substantial contribution to O-GlcNAc site specificity.


Subject(s)
Catalytic Domain , Glycosylation , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Crystallography, X-Ray , Humans , Mass Spectrometry , Models, Molecular , Protein Conformation , Substrate Specificity
14.
J Biol Chem ; 289(33): 23020-23028, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24942743

ABSTRACT

Chitin synthases (CHS) produce chitin, an essential component of the fungal cell wall. The molecular mechanism of processive chitin synthesis is not understood, limiting the discovery of new inhibitors of this enzyme class. We identified the bacterial glycosyltransferase NodC as an appropriate model system to study the general structure and reaction mechanism of CHS. A high throughput screening-compatible novel assay demonstrates that a known inhibitor of fungal CHS also inhibit NodC. A structural model of NodC, on the basis of the recently published BcsA cellulose synthase structure, enabled probing of the catalytic mechanism by mutagenesis, demonstrating the essential roles of the DD and QXXRW catalytic motifs. The NodC membrane topology was mapped, validating the structural model. Together, these approaches give insight into the CHS structure and mechanism and provide a platform for the discovery of inhibitors for this antifungal target.


Subject(s)
Bacterial Proteins , Chitin Synthase , Chitin , Models, Biological , Models, Molecular , Sinorhizobium meliloti/enzymology , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chitin/biosynthesis , Chitin/chemistry , Chitin Synthase/chemistry , Chitin Synthase/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/metabolism
15.
Biochem J ; 457(3): 497-502, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24256146

ABSTRACT

Inhibitors of OGT (O-GlcNAc transferase) are valuable tools to study the cell biology of protein O-GlcNAcylation. We report OGT bisubstrate-linked inhibitors (goblins) in which the acceptor serine in the peptide VTPVSTA is covalently linked to UDP, eliminating the GlcNAc pyranoside ring. Goblin1 co-crystallizes with OGT, revealing an ordered C3 linker and retained substrate-binding modes, and binds the enzyme with micromolar affinity, inhibiting glycosyltransfer on to protein and peptide substrates.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Models, Molecular , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Oligopeptides/pharmacology , Uridine Diphosphate/analogs & derivatives , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glycosylation/drug effects , Humans , Interferometry , Kinetics , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine/chemistry , Uridine Diphosphate/chemistry , Uridine Diphosphate/metabolism , Uridine Diphosphate/pharmacology
16.
J Proteome Res ; 12(2): 927-36, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23301498

ABSTRACT

The post-translational modification of proteins with N-acetylglucosamine (O-GlcNAc) is involved in the regulation of a wide variety of cellular processes and associated with a number of chronic diseases. Despite its emerging biological significance, the systematic identification of O-GlcNAc proteins is still challenging. In the present study, we demonstrate a significantly improved O-GlcNAc protein enrichment procedure, which exploits metabolic labeling of cells by azide-modified GlcNAc and copper-mediated Click chemistry for purification of modified proteins on an alkyne-resin. On-resin proteolysis using trypsin followed by LC-MS/MS afforded the identification of around 1500 O-GlcNAc proteins from a single cell line. Subsequent elution of covalently resin bound O-GlcNAc peptides using selective ß-elimination enabled the identification of 185 O-GlcNAc modification sites on 80 proteins. To demonstrate the practical utility of the developed approach, we studied the global effects of the O-GlcNAcase inhibitor GlcNAcstatin G on the level of O-GlcNAc modification of cellular proteins. About 200 proteins including several key players involved in the hexosamine signaling pathway showed significantly increased O-GlcNAcylation levels in response to the drug, which further strengthens the link of O-GlcNAc protein modification to cellular nutrient sensing and response.


Subject(s)
Acetylglucosamine/metabolism , Immobilized Proteins/isolation & purification , Immobilized Proteins/metabolism , Peptide Fragments/analysis , Protein Processing, Post-Translational , Proteome/isolation & purification , Proteome/metabolism , Alkynes/chemistry , Azides/chemistry , Chromatography, Liquid , Click Chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Tandem Mass Spectrometry , Trypsin/chemistry , beta-N-Acetylhexosaminidases/antagonists & inhibitors , beta-N-Acetylhexosaminidases/metabolism
17.
Nat Chem Biol ; 8(12): 969-74, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23103942

ABSTRACT

Protein O-GlcNAcylation is an essential post-translational modification on hundreds of intracellular proteins in metazoa, catalyzed by O-linked ß-N-acetylglucosamine (O-GlcNAc) transferase (OGT) using unknown mechanisms of transfer and substrate recognition. Through crystallographic snapshots and mechanism-inspired chemical probes, we define how human OGT recognizes the sugar donor and acceptor peptide and uses a new catalytic mechanism of glycosyl transfer, involving the sugar donor α-phosphate as the catalytic base as well as an essential lysine. This mechanism seems to be a unique evolutionary solution to the spatial constraints imposed by a bulky protein acceptor substrate and explains the unexpected specificity of a recently reported metabolic OGT inhibitor.


Subject(s)
Diphosphates/metabolism , N-Acetylglucosaminyltransferases/metabolism , Nucleotides/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Catalysis , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Lysine/metabolism , Models, Molecular , Molecular Sequence Data , Phosphates/metabolism , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Stereoisomerism , Substrate Specificity , Surface Plasmon Resonance , Uridine Diphosphate Galactose/metabolism
18.
Chem Biol ; 19(2): 173-8, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22365600

ABSTRACT

Protein O-GlcNAcylation is an essential reversible posttranslational modification in higher eukaryotes. O-GlcNAc addition and removal is catalyzed by O-GlcNAc transferase and O-GlcNAcase, respectively. We report the molecular details of the interaction of a bacterial O-GlcNAcase homolog with three different synthetic glycopeptides derived from characterized O-GlcNAc sites in the human proteome. Strikingly, the peptides bind a conserved O-GlcNAcase substrate binding groove with similar orientation and conformation. In addition to extensive contacts with the sugar, O-GlcNAcase recognizes the peptide backbone through hydrophobic interactions and intramolecular hydrogen bonds, while avoiding interactions with the glycopeptide side chains. These findings elucidate the molecular basis of O-GlcNAcase substrate specificity, explaining how a single enzyme achieves cycling of the complete O-GlcNAc proteome. In addition, this work will aid development of O-GlcNAcase inhibitors that target the peptide binding site.


Subject(s)
Carbohydrates/chemistry , N-Acetylglucosaminyltransferases/chemistry , Peptides/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Glycosylation , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Kinetics , N-Acetylglucosaminyltransferases/metabolism , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Proteome/metabolism , Substrate Specificity
19.
EMBO J ; 31(6): 1394-404, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22307082

ABSTRACT

Transforming growth factor (TGF)-ß-activated kinase 1 (TAK1) is a key serine/threonine protein kinase that mediates signals transduced by pro-inflammatory cytokines such as transforming growth factor-ß, tumour necrosis factor (TNF), interleukin-1 (IL-1) and wnt family ligands. TAK1 is found in complex with binding partners TAB1-3, phosphorylation and ubiquitination of which has been found to regulate TAK1 activity. In this study, we show that TAB1 is modified with N-acetylglucosamine (O-GlcNAc) on a single site, Ser395. With the help of a novel O-GlcNAc site-specific antibody, we demonstrate that O-GlcNAcylation of TAB1 is induced by IL-1 and osmotic stress, known inducers of the TAK1 signalling cascade. By reintroducing wild-type or an O-GlcNAc-deficient mutant TAB1 (S395A) into Tab1(-/-) mouse embryonic fibroblasts, we determined that O-GlcNAcylation of TAB1 is required for full TAK1 activation upon stimulation with IL-1/osmotic stress, for downstream activation of nuclear factor κB and finally production of IL-6 and TNFα. This is one of the first examples of a single O-GlcNAc site on a signalling protein modulating a key innate immunity signalling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Interleukin-1/metabolism , Interleukin-6/metabolism , MAP Kinase Kinase Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acylation , Animals , Cells, Cultured , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , NF-kappa B/metabolism , Osmotic Pressure/physiology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Signal Transduction/physiology
20.
ACS Chem Biol ; 6(6): 648-57, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21425873

ABSTRACT

Protozoan parasites of the genus Leishmania synthesize lipophosphoglycans (LPGs), phosphoglycans and proteophosphoglycans that contain phosphosaccharide repeat units of [-6)Gal(ß1-4)Man(α1-OPO(3)H-]. The repeat structures are assembled by sequential addition of Manα1-OPO(3)H and ß-Gal. In this study, an UDP-Gal-dependent activity was detected in L. donovani and L. major membranes using synthetic phospho-oligosaccharide fragments of lipophosphoglycan as acceptor substrates. Incubation of a microsomal preparation from L. donovani or L. major parasites with synthetic substrates and UDP-[6-(3)H]Gal resulted in incorporation of radiolabel into these exogenous acceptors. The [(3)H]galactose-labeled products were characterized by degradation into radioactive, low molecular mass fragments upon hydrolysis with mild acid and treatment with ß-galactosidases. We showed that the activity detected with L. donovani membranes is the elongating ß-d-galactosyltransferase associated with LPG phosphosaccharide backbone biosynthesis (eGalT). The eGalT activity showed a requirement for the presence of at least one phosphodiester group in the substrate and it was enhanced dramatically when two or three phosphodiester groups were present. Using the same substrates we detected two types of galactosyltransferase activity in L. major membranes: the elongating ß-d-galactosyltransferase and a branching ß-d-galactosyltransferase (bGalT). Both L. major enzymes required a minimum of one phosphodiester group present in the substrate, but acceptors with two or three phosphodiester groups were found to be superior.


Subject(s)
Leishmania/enzymology , beta-N-Acetylglucosaminylglycopeptide beta-1,4-Galactosyltransferase/metabolism , Carbohydrate Conformation , Enzyme Activation , Glycosphingolipids/biosynthesis , Molecular Sequence Data , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...