Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Curr Opin Chem Biol ; 81: 102476, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38861851

ABSTRACT

O-GlcNAcylation is an essential protein glycosylation governed by two O-GlcNAc cycling enzymes: O-GlcNAc transferase (OGT) installs a single sugar moiety N-acetylglucosamine (GlcNAc) on protein serine and threonine residues, and O-GlcNAcase (OGA) removes them. Aberrant O-GlcNAcylation has been implicated in various diseases. However, the large repertoire of more than 1000 O-GlcNAcylated proteins and the elusive mechanisms of OGT/OGA in substrate recognition present significant challenges in targeting the dysregulated O-GlcNAcylation for therapeutic development. Recently, emerging evidence suggested that the non-catalytic domains play critical roles in regulating the functional specificity of OGT/OGA via modulating their protein interactions and substrate recognition. Here, we discuss recent studies on the structures, mechanisms, and related tools of the OGT/OGA non-catalytic domains, highlighting new opportunities for function-specific control.

2.
Proc Natl Acad Sci U S A ; 121(24): e2320867121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838015

ABSTRACT

O-GlcNAcase (OGA) is the only human enzyme that catalyzes the hydrolysis (deglycosylation) of O-linked beta-N-acetylglucosaminylation (O-GlcNAcylation) from numerous protein substrates. OGA has broad implications in many challenging diseases including cancer. However, its role in cell malignancy remains mostly unclear. Here, we report that a cancer-derived point mutation on the OGA's noncatalytic stalk domain aberrantly modulates OGA interactome and substrate deglycosylation toward a specific set of proteins. Interestingly, our quantitative proteomic studies uncovered that the OGA stalk domain mutant preferentially deglycosylated protein substrates with +2 proline in the sequence relative to the O-GlcNAcylation site. One of the most dysregulated substrates is PDZ and LIM domain protein 7 (PDLIM7), which is associated with the tumor suppressor p53. We found that the aberrantly deglycosylated PDLIM7 suppressed p53 gene expression and accelerated p53 protein degradation by promoting the complex formation with E3 ubiquitin ligase MDM2. Moreover, deglycosylated PDLIM7 significantly up-regulated the actin-rich membrane protrusions on the cell surface, augmenting the cancer cell motility and aggressiveness. These findings revealed an important but previously unappreciated role of OGA's stalk domain in protein substrate recognition and functional modulation during malignant cell progression.


Subject(s)
Cytoskeleton , LIM Domain Proteins , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Cytoskeleton/metabolism , Acetylglucosamine/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Cell Line, Tumor , Glycosylation , Hydrolysis , Mutation , Cell Movement , Antigens, Neoplasm , Hyaluronoglucosaminidase , Histone Acetyltransferases
3.
Nat Chem Biol ; 19(11): 1423-1431, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37653170

ABSTRACT

The modification of intracellular proteins with O-linked ß-N-acetylglucosamine (O-GlcNAc) moieties is a highly dynamic process that spatiotemporally regulates nearly every important cellular program. Despite its significance, little is known about the substrate recognition and regulation modes of O-GlcNAc transferase (OGT), the primary enzyme responsible for O-GlcNAc addition. In this study, we identified the intervening domain (Int-D), a poorly understood protein fold found only in metazoan OGTs, as a specific regulator of OGT protein-protein interactions and substrate modification. Using proteomic peptide phage display (ProP-PD) coupled with structural, biochemical and cellular characterizations, we discovered a strongly enriched peptide motif, employed by the Int-D to facilitate specific O-GlcNAcylation. We further show that disruption of Int-D binding dysregulates important cellular programs, including response to nutrient deprivation and glucose metabolism. These findings illustrate a mode of OGT substrate recognition and offer key insights into the biological roles of this unique domain.


Subject(s)
Proteins , Proteomics , Animals , Acetylglucosamine/metabolism , N-Acetylglucosaminyltransferases/metabolism , Peptides
4.
Res Sq ; 2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36993758

ABSTRACT

O-GlcNAcase (OGA) is the sole enzyme that hydrolyzes O-GlcNAcylation from thousands of proteins and is dysregulated in many diseases including cancer. However, the substrate recognition and pathogenic mechanisms of OGA remain largely unknown. Here we report the first discovery of a cancer-derived point mutation on the OGA's non-catalytic stalk domain that aberrantly regulated a small set of OGA-protein interactions and O-GlcNAc hydrolysis in critical cellular processes. We uncovered a novel cancer-promoting mechanism in which the OGA mutant preferentially hydrolyzed the O-GlcNAcylation from modified PDLIM7 and promoted cell malignancy by down-regulating p53 tumor suppressor in different types of cells through transcription inhibition and MDM2-mediated ubiquitination. Our study revealed the OGA deglycosylated PDLIM7 as a novel regulator of p53-MDM2 pathway, offered the first set of direct evidence on OGA substrate recognition beyond its catalytic site, and illuminated new directions to interrogate OGA's precise role without perturbing global O-GlcNAc homeostasis for biomedical applications.

5.
Res Sq ; 2023 Feb 02.
Article in English | MEDLINE | ID: mdl-36778302

ABSTRACT

The modification of intracellular proteins with O-linked ß- N -acetylglucosamine (O-GlcNAc) moieties is a highly dynamic process that spatiotemporally regulates nearly every important cellular program. Despite its significance, little is known about the substrate recognition and regulation modes of O-GlcNAc transferase (OGT), the primary enzyme responsible for O-GlcNAc addition. In this study, we have identified the intervening domain (Int-D), a poorly understood protein fold found only in metazoan OGTs, as a specific regulator of OGT protein-protein interactions and substrate modification. Utilizing an innovative proteomic peptide phage display (ProP-PD) coupled with structural, biochemical, and cellular characterizations, we discovered a novel peptide motif, employed by the Int-D to facilitate specific O-GlcNAcylation. We further show that disruption of Int-D binding dysregulates important cellular programs including nutrient stress response and glucose metabolism. These findings illustrate a novel mode of OGT substrate recognition and offer the first insights into the biological roles of this unique domain.

6.
Cancers (Basel) ; 14(20)2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36291918

ABSTRACT

The dynamic O-GlcNAc modification of intracellular proteins is an important nutrient sensor for integrating metabolic signals into vast networks of highly coordinated cellular activities. Dysregulation of the sole enzymes responsible for O-GlcNAc cycling, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), and the associated cellular O-GlcNAc profile is a common feature across nearly every cancer type. Many studies have investigated the effects of aberrant OGT/OGA expression on global O-GlcNAcylation activity in cancer cells. However, recent studies have begun to elucidate the roles of protein-protein interactions (PPIs), potentially through regions outside of the immediate catalytic site of OGT/OGA, that regulate greater protein networks to facilitate substrate-specific modification, protein translocalization, and the assembly of larger biomolecular complexes. Perturbation of OGT/OGA PPI networks makes profound changes in the cell and may directly contribute to cancer malignancies. Herein, we highlight recent studies on the structural features of OGT and OGA, as well as the emerging roles and molecular mechanisms of their aberrant PPIs in rewiring cancer networks. By integrating complementary approaches, the research in this area will aid in the identification of key protein contacts and functional modules derived from OGT/OGA that drive oncogenesis and will illuminate new directions for anti-cancer drug development.

8.
Int J Biol Macromol ; 169: 51-59, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33333092

ABSTRACT

The essential human O-linked ß-N-acetylglucosamine (O-GlcNAc) transferase (OGT) is the sole enzyme responsible for modifying thousands of intracellular proteins with the monosaccharide O-GlcNAc. This unique modification plays crucial roles in human health and disease, but the substrate recognition of OGT remains poorly understood. Intriguingly, the only human enzyme reported to remove this modification, O-GlcNAcase (OGA), is O-GlcNAc modified. Here, we exploited a GlcNAc electrophilic probe (GEP1A) to rapidly screen OGT mutants in a fluorescence assay that can discriminate between altered OGT-sugar and -protein substrate binding to help elucidate the binding mode of OGT toward OGA protein substrate. Since OGT tetratricopeptide repeat (TPR) domain plays a key role in OGT-OGA binding, we screened 30 OGT TPR mutants, which revealed 15 "ladder like" asparagine or aspartate residues spanning TPRs 3-7 and 10-13.5 that affect OGA O-GlcNAcylation. By applying a truncated OGA construct, we found that OGA's N-terminal region or pseudo histone acetyltransferase domain is not required for its O-GlcNAcylation, suggesting OGT functionally interacts with OGA through its catalytic and/or stalk domains. This work represents the first effort to systemically investigate each OGT TPR and our findings will facilitate the development of new strategies to investigate the role of substrate-specific O-GlcNAcylation.


Subject(s)
N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , beta-N-Acetylhexosaminidases/chemistry , Acetylglucosamine/metabolism , Click Chemistry/methods , Humans , N-Acetylglucosaminyltransferases/ultrastructure , Protein Processing, Post-Translational , Proteins/metabolism , beta-N-Acetylhexosaminidases/metabolism , beta-N-Acetylhexosaminidases/ultrastructure
9.
Chemistry ; 26(53): 12086-12100, 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32207184

ABSTRACT

The O-linked ß-N-acetylglucosamine (O-GlcNAc) modification, termed O-GlcNAcylation, is an essential and dynamic post-translational modification in cells. O-GlcNAc transferase (OGT) installs this modification on serine and threonine residues, whereas O-GlcNAcase (OGA) hydrolyzes it. O-GlcNAc modifications are found on thousands of intracellular proteins involved in diverse biological processes. Dysregulation of O-GlcNAcylation and O-GlcNAc cycling enzymes has been detected in many diseases, including cancer, diabetes, cardiovascular and neurodegenerative diseases. Here, recent advances in the development of molecular tools to investigate OGT and OGA functions and substrate recognition are discussed. New chemical approaches to study O-GlcNAc dynamics and its potential roles in the immune system are also highlighted. It is hoped that this minireview will encourage more research in these areas to advance the understanding of O-GlcNAc in biology and diseases.


Subject(s)
Acetylglucosamine/chemistry , N-Acetylglucosaminyltransferases , beta-N-Acetylhexosaminidases , Acetylglucosamine/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational , Serine/chemistry , Threonine/chemistry
10.
Chem Commun (Camb) ; 55(88): 13291-13294, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31626249

ABSTRACT

O-GlcNAc transferase (OGT) glycosylates numerous proteins and is implicated in many diseases. To date, most OGT inhibitors lack either sufficient potency or characterized specificity in cells. We report the first targeted covalent inhibitor that predominantly reacts with OGT but does not affect other functionally similar enzymes. This study provides a new strategy to interrogate cellular OGT functions and to investigate other glycosyltransferases.


Subject(s)
Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , MCF-7 Cells , Models, Molecular , N-Acetylglucosaminyltransferases/metabolism
11.
Curr Opin Struct Biol ; 56: 97-106, 2019 06.
Article in English | MEDLINE | ID: mdl-30708324

ABSTRACT

Dysregulation of nuclear and cytoplasmic O-linked ß-N-acetylglucosamine (O-GlcNAc) cycling is implicated in a range of diseases including diabetes and cancer. This modification maintains cellular homeostasis by regulating several biological processes, such as cell signaling. This highly regulated cycle is governed by two sole essential enzymes, O-GlcNAc transferase and O-GlcNAcase that add O-GlcNAc and remove it from over a thousand substrates, respectively. Until recently, due to lack of structural information, the mechanism of substrate recognition has eluted researchers. Here, we review recent successes in structural characterization of these enzymes and how this information has illuminated key features essential for catalysis and substrate recognition. Additionally, we highlight recent studies which have used this information to expand our understanding of substrate specificity by each enzyme.


Subject(s)
Acetylglucosamine/metabolism , Biocatalysis , Transferases/chemistry , Transferases/metabolism , Humans
12.
Chembiochem ; 20(3): 312-318, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30199580

ABSTRACT

The O-linked N-acetylglucosamine (O-GlcNAc) modification is an essential component in cell regulation. A single pair of human enzymes conducts this modification dynamically on a broad variety of proteins: O-GlcNAc transferase (OGT) adds the GlcNAc residue and O-GlcNAcase (OGA) hydrolyzes it. This modification is dysregulated in many diseases, but its exact effect on particular substrates remains unclear. In addition, no apparent sequence motif has been found in the modified proteins, and the factors controlling the substrate specificity of OGT and OGA are largely unknown. In this minireview, we will discuss recent developments in chemical and biochemical methods toward addressing the challenge of OGT and OGA substrate recognition. We hope that the new concepts and knowledge from these studies will promote research in this area to advance understanding of O-GlcNAc regulation in health and disease.


Subject(s)
N-Acetylglucosaminyltransferases/metabolism , beta-N-Acetylhexosaminidases/metabolism , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Humans , Hydrolysis , Models, Molecular , N-Acetylglucosaminyltransferases/chemistry , Substrate Specificity , beta-N-Acetylhexosaminidases/chemistry
14.
Nat Cell Biol ; 19(11): 1358-1370, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29058718

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. Herein we discover that the key glycolytic enzyme pyruvate kinase M2 isoform (PKM2), but not the related isoform PKM1, is methylated by co-activator-associated arginine methyltransferase 1 (CARM1). PKM2 methylation reversibly shifts the balance of metabolism from oxidative phosphorylation to aerobic glycolysis in breast cancer cells. Oxidative phosphorylation depends on mitochondrial calcium concentration, which becomes critical for cancer cell survival when PKM2 methylation is blocked. By interacting with and suppressing the expression of inositol-1,4,5-trisphosphate receptors (InsP3Rs), methylated PKM2 inhibits the influx of calcium from the endoplasmic reticulum to mitochondria. Inhibiting PKM2 methylation with a competitive peptide delivered by nanoparticles perturbs the metabolic energy balance in cancer cells, leading to a decrease in cell proliferation, migration and metastasis. Collectively, the CARM1-PKM2 axis serves as a metabolic reprogramming mechanism in tumorigenesis, and inhibiting PKM2 methylation generates metabolic vulnerability to InsP3R-dependent mitochondrial functions.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carrier Proteins/metabolism , Glycolysis/physiology , Guanylate Cyclase/metabolism , Membrane Proteins/metabolism , Thyroid Hormones/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Endoplasmic Reticulum/metabolism , Female , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , MCF-7 Cells , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Neoplasm Metastasis/pathology , Oxidative Phosphorylation , Thyroid Hormone-Binding Proteins
15.
Nat Chem Biol ; 13(12): 1267-1273, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058723

ABSTRACT

O-linked ß-N-acetylglucosamine (O-GlcNAc) transferase (OGT) is an essential human glycosyltransferase that adds O-GlcNAc modifications to numerous proteins. However, little is known about the mechanism with which OGT recognizes various protein substrates. Here we report on GlcNAc electrophilic probes (GEPs) to expedite the characterization of OGT-substrate recognition. Data from mass spectrometry, X-ray crystallization, and biochemical and radiolabeled kinetic assays support the application of GEPs to rapidly report the impacts of OGT mutations on protein substrate or sugar binding and to discover OGT residues crucial for protein recognition. Interestingly, we found that the same residues on the inner surface of the N-terminal domain contribute to OGT interactions with different protein substrates. By tuning reaction conditions, a GEP enables crosslinking of OGT with acceptor substrates in situ, affording a unique method to discover genuine substrates that weakly or transiently interact with OGT. Hence, GEPs provide new strategies to dissect OGT-substrate binding and recognition.


Subject(s)
Molecular Probes/metabolism , N-Acetylglucosaminyltransferases/metabolism , Crystallography, X-Ray , Humans , Kinetics , Mass Spectrometry , Models, Molecular , Molecular Conformation , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Mutation , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/genetics , Substrate Specificity
16.
Nat Commun ; 8(1): 666, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28939839

ABSTRACT

The O-linked ß-N-acetyl glucosamine (O-GlcNAc) modification dynamically regulates the functions of numerous proteins. A single human enzyme O-linked ß-N-acetyl glucosaminase (O-GlcNAcase or OGA) hydrolyzes this modification. To date, it remains largely unknown how OGA recognizes various substrates. Here we report the structures of OGA in complex with each of four distinct glycopeptide substrates that contain a single O-GlcNAc modification on a serine or threonine residue. Intriguingly, these glycopeptides bind in a bidirectional yet conserved conformation within the substrate-binding cleft of OGA. This study provides fundamental insights into a general principle that confers the substrate binding adaptability and specificity to OGA in O-GlcNAc regulation.O-linked ß-N-acetyl glucosamine (O-GlcNAc) is an important protein modification that is hydrolyzed by O-GlcNAcase (OGA). Here the authors give insights into OGA substrate recognition by presenting four human OGA structures complexed with glycopeptide substrates containing a single O-GlcNAc modification on either a serine or threonine.


Subject(s)
beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/metabolism , Crystallography, X-Ray , Glycopeptides/chemistry , Glycopeptides/metabolism , Humans , Models, Molecular , Protein Conformation , Serine/chemistry , Substrate Specificity , Threonine/chemistry , beta-N-Acetylhexosaminidases/genetics
17.
Nat Struct Mol Biol ; 24(4): 362-369, 2017 04.
Article in English | MEDLINE | ID: mdl-28319083

ABSTRACT

Human O-GlcNAcase (hOGA) is the unique enzyme responsible for the hydrolysis of the O-linked ß-N-acetyl glucosamine (O-GlcNAc) modification, an essential protein glycosylation event that modulates the function of numerous cellular proteins in response to nutrients and stress. Here we report crystal structures of a truncated hOGA, which comprises the catalytic and stalk domains, in apo form, in complex with an inhibitor, and in complex with a glycopeptide substrate. We found that hOGA forms an unusual arm-in-arm homodimer in which the catalytic domain of one monomer is covered by the stalk domain of the sister monomer to create a substrate-binding cleft. Notably, the residues on the cleft surface afford extensive interactions with the peptide substrate in a recognition mode that is distinct from that of its bacterial homologs. These structures represent the first model of eukaryotic enzymes in the glycoside hydrolase 84 (GH84) family and provide a crucial starting point for understanding the substrate specificity of hOGA, which regulates a broad range of biological and pathological processes.


Subject(s)
beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glycosylation/drug effects , Humans , Protein Multimerization/drug effects , Protein Structure, Secondary , Substrate Specificity/drug effects , beta-N-Acetylhexosaminidases/antagonists & inhibitors
18.
ACS Chem Biol ; 12(2): 326-335, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28055183

ABSTRACT

O-GlcNAcylation is the modification of serine and threonine residues with ß-N-acetylglucosamine (O-GlcNAc) on intracellular proteins. This dynamic modification is attached by O-GlcNAc transferase (OGT) and removed by O-GlcNAcase (OGA) and is a critical regulator of various cellular processes. Furthermore, O-GlcNAcylation is dysregulated in many diseases, such as diabetes, cancer, and Alzheimer's disease. However, the precise role of this modification and its cycling enzymes (OGT and OGA) in normal and disease states remains elusive. This is partially due to the difficulty in studying O-GlcNAcylation with traditional genetic and biochemical techniques. In this review, we will summarize recent progress in chemical approaches to overcome these obstacles. We will cover new inhibitors of OGT and OGA, advances in metabolic labeling and cellular imaging, synthetic approaches to access homogeneous O-GlcNAcylated proteins, and cross-linking methods to identify O-GlcNAc-protein interactions. We will also discuss remaining gaps in our toolbox for studying O-GlcNAcylation and questions of high interest that are yet to be answered.


Subject(s)
Acetylglucosamine/metabolism , Proteins/metabolism , Glycosyltransferases/metabolism , Mass Spectrometry , Microscopy, Fluorescence , Molecular Probes/chemistry
19.
Biochemistry ; 55(7): 1149-58, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26807597

ABSTRACT

O-GlcNAcylation is a nutrient-responsive glycosylation that plays a pivotal role in transcriptional regulation. Human RNA polymerase II (Pol II) is extensively modified by O-linked N-acetylglucosamine (O-GlcNAc) on its unique C-terminal domain (CTD), which consists of 52 heptad repeats. One approach to understanding the function of glycosylated Pol II is to determine the mechanism of dynamic O-GlcNAcylation on the CTD. Here, we discovered that the Pol II CTD can be extensively O-GlcNAcylated in vitro and in cells. Efficient glycosylation requires a minimum of 20 heptad repeats of the CTD and more than half of the N-terminal domain of O-GlcNAc transferase (OGT). Under conditions of saturated sugar donor, we monitored the attachment of more than 20 residues of O-GlcNAc to the full-length CTD. Surprisingly, glycosylation on the periodic CTD follows a distributive mechanism, resulting in highly heterogeneous glycoforms. Our data suggest that initial O-GlcNAcylation can take place either on the proximal or on the distal region of the CTD, and subsequent glycosylation occurs similarly over the entire CTD with nonuniform distributions. Moreover, removal of O-GlcNAc from glycosylated CTD is also distributive and is independent of O-GlcNAcylation level. Our results suggest that O-GlcNAc cycling enzymes can employ a similar mechanism to react with other protein substrates on multiple sites. Distributive O-GlcNAcylation on Pol II provides another regulatory mechanism of transcription in response to fluctuating cellular conditions.


Subject(s)
Acetylglucosamine/metabolism , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Gene Deletion , Glycosylation , HeLa Cells , Humans , Kinetics , Molecular Weight , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Mapping , Protein Interaction Domains and Motifs , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity , Tandem Mass Spectrometry
20.
ACS Chem Biol ; 10(6): 1392-7, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25751766

ABSTRACT

O-GlcNAc transferase (OGT) is an essential mammalian enzyme that regulates numerous cellular processes through the attachment of O-linked N-acetylglucosamine (O-GlcNAc) residues to nuclear and cytoplasmic proteins. Its targets include kinases, phosphatases, transcription factors, histones, and many other intracellular proteins. The biology of O-GlcNAc modification is still not well understood, and cell-permeable inhibitors of OGT are needed both as research tools and for validating OGT as a therapeutic target. Here, we report a small molecule OGT inhibitor, OSMI-1, developed from a high-throughput screening hit. It is cell-permeable and inhibits protein O-GlcNAcylation in several mammalian cell lines without qualitatively altering cell surface N- or O-linked glycans. The development of this molecule validates high-throughput screening approaches for the discovery of glycosyltransferase inhibitors, and further optimization of this scaffold may lead to yet more potent OGT inhibitors useful for studying OGT in animal models.


Subject(s)
Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , CHO Cells , Cell Membrane Permeability , Cricetulus , Enzyme Inhibitors/chemical synthesis , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Lectins/chemistry , Lectins/metabolism , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Small Molecule Libraries/chemical synthesis , Uridine Diphosphate/chemistry , Uridine Diphosphate/metabolism , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...