96 results on '"Vakhrushev, SY"'
Search Results
2. Glycoproteomics
- Author
-
Bagdonaite, I, Malaker, SA, Polasky, DA, Riley, NM, Schjoldager, K, Vakhrushev, SY, Halim, A, Aoki-Kinoshita, KF, Nesvizhskii, AI, Bertozzi, CR, Wandall, HH, Parker, BL, Thaysen-Andersen, M, Scott, NE, Bagdonaite, I, Malaker, SA, Polasky, DA, Riley, NM, Schjoldager, K, Vakhrushev, SY, Halim, A, Aoki-Kinoshita, KF, Nesvizhskii, AI, Bertozzi, CR, Wandall, HH, Parker, BL, Thaysen-Andersen, M, and Scott, NE
- Published
- 2022
3. Development of a FUT8 Inhibitor with Cellular Inhibitory Properties.
- Author
-
Manabe Y, Takebe T, Kasahara S, Hizume K, Kabayama K, Kamada Y, Asakura A, Shinzaki S, Takamatsu S, Miyoshi E, García-García A, Vakhrushev SY, Hurtado-Guerrero R, and Fukase K
- Subjects
- Humans, Molecular Structure, Structure-Activity Relationship, Fucosyltransferases antagonists & inhibitors, Fucosyltransferases metabolism, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Enzyme Inhibitors chemical synthesis
- Abstract
Core fucosylation is catalyzed by α-1,6-fucosyltransferase (FUT8), which fucosylates the innermost GlcNAc of N-glycans. Given the association of FUT8 with various diseases, including cancer, selective FUT8 inhibitors applicable to in vivo or cell-based systems are highly sought-after. Herein, we report the discovery of a compound that selectively inhibits FUT8 in cell-based assays. High-throughput screening revealed a FUT8-inhibiting pharmacophore, and further structural optimization yielded an inhibitor with a K
D value of 49 nM. Notably, this binding occurs only in the presence of GDP (a product of the enzymatic reaction catalyzed by FUT8). Mechanistic studies suggested that this inhibitor generates a highly reactive naphthoquinone methide derivative at the binding site in FUT8, which subsequently reacts with FUT8. Furthermore, prodrug derivatization of this inhibitor improved its stability, enabling suppression of core fucose expression and subsequent EGFR and T-cell signaling in cell-based assays, paving the way for the development of drugs targeting core fucosylation., (© 2024 The Authors. Angewandte Chemie International Edition published by Wiley-VCH GmbH.)- Published
- 2024
- Full Text
- View/download PDF
4. Mammalian cell-based production of glycans, glycopeptides and glycomodules.
- Author
-
Jaroentomeechai T, Karlsson R, Goerdeler F, Teoh FKY, Grønset MN, de Wit D, Chen YH, Furukawa S, Psomiadou V, Hurtado-Guerrero R, Vidal-Calvo EE, Salanti A, Boltje TJ, van den Bos LJ, Wunder C, Johannes L, Schjoldager KT, Joshi HJ, Miller RL, Clausen H, Vakhrushev SY, and Narimatsu Y
- Subjects
- Humans, Glycosylation, Animals, Glycoconjugates metabolism, Glycoconjugates chemistry, HEK293 Cells, CHO Cells, Cricetulus, Gene Editing methods, Glycopeptides metabolism, Glycopeptides chemistry, Polysaccharides metabolism, Polysaccharides chemistry
- Abstract
Access to defined glycans and glycoconjugates is pivotal for discovery, dissection, and harnessing of a range of biological functions orchestrated by cellular glycosylation processes and the glycome. We previously employed genetic glycoengineering by nuclease-based gene editing to develop sustainable production of designer glycoprotein therapeutics and cell-based glycan arrays that display glycans in their natural context at the cell surface. However, access to human glycans in formats and quantities that allow structural studies of molecular interactions and use of glycans in biomedical applications currently rely on chemical and chemoenzymatic syntheses associated with considerable labor, waste, and costs. Here, we develop a sustainable and scalable method for production of glycans in glycoengineered mammalian cells by employing secreted Glycocarriers with repeat glycosylation acceptor sequence motifs for different glycans. The Glycocarrier technology provides a flexible production platform for glycans in different formats, including oligosaccharides, glycopeptides, and multimeric glycomodules, and offers wide opportunities for use in bioassays and biomedical applications., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Altered O-glycosylation of β 1 -adrenergic receptor N-terminal single-nucleotide variants modulates receptor processing and functional activity.
- Author
-
Tuhkanen HE, Haasiomäki IJ, Lackman JJ, Goth CK, Mattila SO, Ye Z, Vakhrushev SY, Magga J, Kerkelä R, Clausen H, Schjoldager KT, and Petäjä-Repo UE
- Abstract
N-terminal nonsynonymous single-nucleotide polymorphisms (SNPs) of G protein-coupled receptors (GPCRs) are common and often affect receptor post-translational modifications. Their functional implications are, however, largely unknown. We have previously shown that the human β
1 -adrenergic receptor (β1 AR) is O-glycosylated in the N-terminal extracellular domain by polypeptide GalNAc transferase-2 that co-regulates receptor proteolytic cleavage. Here, we demonstrate that the common S49G and the rare A29T and R31Q SNPs alter these modifications, leading to distinct effects on receptor processing. This was achieved by in vitro O-glycosylation assays, analysis of native receptor N-terminal O-glycopeptides, and expression of receptor variants in cell lines and neonatal rat ventricular cardiomyocytes deficient in O-glycosylation. The SNPs eliminated (S49G) or introduced (A29T) regulatory O-glycosites that enhanced or inhibited cleavage at the adjacent sites (P52 ↓L53 and R31 ↓L32 ), respectively, or abolished the major site at R31 ↓L32 (R31Q). The inhibition of proteolysis of the T29 and Q31 variants correlated with increased full-length receptor levels at the cell surface. Furthermore, the S49 variant showed increased isoproterenol-mediated signaling in an enhanced bystander bioluminescence energy transfer β-arrestin2 recruitment assay in a coordinated manner with the common C-terminal R389G polymorphism. As Gly at position 49 is ancestral in placental mammals, the results suggest that its exchange to Ser has created a β1 AR gain-of-function phenotype in humans. This study provides evidence for regulatory mechanisms by which GPCR SNPs outside canonical domains that govern ligand binding and activation can alter receptor processing and function. Further studies on other GPCR SNPs with clinical importance as drug targets are thus warranted., (© 2024 The Author(s). The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2024
- Full Text
- View/download PDF
6. Fish-hunting cone snail disrupts prey's glucose homeostasis with weaponized mimetics of somatostatin and insulin.
- Author
-
Yeung HY, Ramiro IBL, Andersen DB, Koch TL, Hamilton A, Bjørn-Yoshimoto WE, Espino S, Vakhrushev SY, Pedersen KB, de Haan N, Hipgrave Ederveen AL, Olivera BM, Knudsen JG, Bräuner-Osborne H, Schjoldager KT, Holst JJ, and Safavi-Hemami H
- Subjects
- Animals, Fishes metabolism, Predatory Behavior drug effects, Hypoglycemia metabolism, Mollusk Venoms metabolism, Humans, Molecular Mimicry, Somatostatin metabolism, Homeostasis drug effects, Insulin metabolism, Glucose metabolism, Receptors, Somatostatin metabolism, Glucagon metabolism, Conus Snail
- Abstract
Venomous animals have evolved diverse molecular mechanisms to incapacitate prey and defend against predators. Most venom components disrupt nervous, locomotor, and cardiovascular systems or cause tissue damage. The discovery that certain fish-hunting cone snails use weaponized insulins to induce hypoglycemic shock in prey highlights a unique example of toxins targeting glucose homeostasis. Here, we show that, in addition to insulins, the deadly fish hunter, Conus geographus, uses a selective somatostatin receptor 2 (SSTR
2 ) agonist that blocks the release of the insulin-counteracting hormone glucagon, thereby exacerbating insulin-induced hypoglycemia in prey. The native toxin, Consomatin nG1, exists in several proteoforms with a minimized vertebrate somatostatin-like core motif connected to a heavily glycosylated N-terminal region. We demonstrate that the toxin's N-terminal tail closely mimics a glycosylated somatostatin from fish pancreas and is crucial for activating the fish SSTR2 . Collectively, these findings provide a stunning example of chemical mimicry, highlight the combinatorial nature of venom components, and establish glucose homeostasis as an effective target for prey capture., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
7. Global View of Domain-Specific O-Linked Mannose Glycosylation in Glycoengineered Cells.
- Author
-
Povolo L, Tian W, Vakhrushev SY, and Halim A
- Subjects
- Humans, Glycosylation, Substrate Specificity, Glycoproteins metabolism, Proteomics methods, Cell Line, Glycosyltransferases metabolism, Glycosyltransferases genetics, Protein Processing, Post-Translational, Cell Engineering methods, Mannose metabolism
- Abstract
Protein O-linked mannose (O-Man) glycosylation is an evolutionary conserved posttranslational modification that fulfills important biological roles during embryonic development. Three nonredundant enzyme families, POMT1/POMT2, TMTC1-4, and TMEM260, selectively coordinate the initiation of protein O-Man glycosylation on distinct classes of transmembrane proteins, including α-dystroglycan, cadherins, and plexin receptors. However, a systematic investigation of their substrate specificities is lacking, in part due to the ubiquitous expression of O-Man glycosyltransferases in cells, which precludes analysis of pathway-specific O-Man glycosylation on a proteome-wide scale. Here, we apply a targeted workflow for membrane glycoproteomics across five human cell lines to extensively map O-Man substrates and genetically deconstruct O-Man initiation by individual and combinatorial knockout of O-Man glycosyltransferase genes. We established a human cell library for the analysis of substrate specificities of individual O-Man initiation pathways by quantitative glycoproteomics. Our results identify 180 O-Man glycoproteins, demonstrate new protein targets for the POMT1/POMT2 pathway, and show that TMTC1-4 and TMEM260 pathways widely target distinct Ig-like protein domains of plasma membrane proteins involved in cell-cell and cell-extracellular matrix interactions. The identification of O-Man on Ig-like folds adds further knowledge on the emerging concept of domain-specific O-Man glycosylation which opens for functional studies of O-Man-glycosylated adhesion molecules and receptors., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Targeting host O-linked glycan biosynthesis affects Ebola virus replication efficiency and reveals differential GalNAc-T acceptor site preferences on the Ebola virus glycoprotein.
- Author
-
Bagdonaite I, Abdurahman S, Mirandola M, Pasqual D, Frank M, Narimatsu Y, Joshi HJ, Vakhrushev SY, Salata C, Mirazimi A, and Wandall HH
- Subjects
- Humans, HEK293 Cells, Glycosylation, Viral Envelope Proteins metabolism, Hemorrhagic Fever, Ebola virology, Hemorrhagic Fever, Ebola metabolism, N-Acetylgalactosaminyltransferases metabolism, N-Acetylgalactosaminyltransferases genetics, Glycoproteins metabolism, Polypeptide N-acetylgalactosaminyltransferase, Ebolavirus physiology, Ebolavirus metabolism, Virus Replication, Polysaccharides metabolism
- Abstract
Ebola virus glycoprotein (EBOV GP) is one of the most heavily O-glycosylated viral glycoproteins, yet we still lack a fundamental understanding of the structure of its large O-glycosylated mucin-like domain and to what degree the host O-glycosylation capacity influences EBOV replication. Using tandem mass spectrometry, we identified 47 O-glycosites on EBOV GP and found similar glycosylation signatures on virus-like particle- and cell lysate-derived GP. Furthermore, we performed quantitative differential O-glycoproteomics on proteins produced in wild-type HEK293 cells and cell lines ablated for the three key initiators of O-linked glycosylation, GalNAc-T1, -T2, and -T3. The data show that 12 out of the 47 O-glycosylated sites were regulated, predominantly by GalNAc-T1. Using the glycoengineered cell lines for authentic EBOV propagation, we demonstrate the importance of O-linked glycan initiation and elongation for the production of viral particles and the titers of progeny virus. The mapped O-glycan positions and structures allowed to generate molecular dynamics simulations probing the largely unknown spatial arrangements of the mucin-like domain. The data highlight targeting GALNT1 or C1GALT1C1 as a possible way to modulate O-glycan density on EBOV GP for novel vaccine designs and tailored intervention approaches.IMPORTANCEEbola virus glycoprotein acquires its extensive glycan shield in the host cell, where it is decorated with N-linked glycans and mucin-type O-linked glycans. The latter is initiated by a family of polypeptide GalNAc-transferases that have different preferences for optimal peptide substrates resulting in a spectrum of both very selective and redundant substrates for each isoform. In this work, we map the exact locations of O-glycans on Ebola virus glycoprotein and identify subsets of sites preferentially initiated by one of the three key isoforms of GalNAc-Ts, demonstrating that each enzyme contributes to the glycan shield integrity. We further show that altering host O-glycosylation capacity has detrimental effects on Ebola virus replication, with both isoform-specific initiation and elongation playing a role. The combined structural and functional data highlight glycoengineered cell lines as useful tools for investigating molecular mechanisms imposed by specific glycans and for steering the immune responses in future vaccine designs., Competing Interests: H.H.W. owns stocks in and is a consultant for and co-founder of EbuMab, ApS, Hemab, ApS, and GO-Therapeutics Inc. All other authors declare no conflicts of interest.
- Published
- 2024
- Full Text
- View/download PDF
9. Molecular-level architecture of Chlamydomonas reinhardtii's glycoprotein-rich cell wall.
- Author
-
Poulhazan A, Arnold AA, Mentink-Vigier F, Muszyński A, Azadi P, Halim A, Vakhrushev SY, Joshi HJ, Wang T, Warschawski DE, and Marcotte I
- Subjects
- Glycoproteins metabolism, Cell Wall metabolism, Cellulose metabolism, Water metabolism, Chlamydomonas, Chlamydomonas reinhardtii metabolism
- Abstract
Microalgae are a renewable and promising biomass for large-scale biofuel, food and nutrient production. However, their efficient exploitation depends on our knowledge of the cell wall composition and organization as it can limit access to high-value molecules. Here we provide an atomic-level model of the non-crystalline and water-insoluble glycoprotein-rich cell wall of Chlamydomonas reinhardtii. Using in situ solid-state and sensitivity-enhanced nuclear magnetic resonance, we reveal unprecedented details on the protein and carbohydrate composition and their nanoscale heterogeneity, as well as the presence of spatially segregated protein- and glycan-rich regions with different dynamics and hydration levels. We show that mannose-rich lower-molecular-weight proteins likely contribute to the cell wall cohesion by binding to high-molecular weight protein components, and that water provides plasticity to the cell-wall architecture. The structural insight exemplifies strategies used by nature to form cell walls devoid of cellulose or other glycan polymers., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Sensitive and Specific Global Cell Surface N -Glycoproteomics Shows Profound Differences Between Glycosylation Sites and Subcellular Components.
- Author
-
de Haan N, Song M, Grant OC, Ye Z, Khoder Agha F, Koed Møller Aasted M, Woods RJ, Vakhrushev SY, and Wandall HH
- Subjects
- Humans, Glycosylation, Mass Spectrometry methods, Glycoproteins chemistry, Polysaccharides chemistry
- Abstract
Cell surface glycans are essential for establishing cell communication, adhesion, and migration. However, it remains challenging to obtain cell surface-specific information about glycoconjugate structures. Acquiring this information is essential for unraveling the functional role of glycans and for exploiting them as clinical targets. To specifically analyze the N -glycoprotein forms expressed at the cell surface, we developed a C18 liquid chromatography (LC)-mass spectrometry (MS)-based glycoproteomics method in combination with highly specific cell surface protein labeling and enrichment using a biotin label. The surface-specificity of the method was validated by MS-based proteomics of subcellular component marker proteins. Using the human keratinocytes N/TERT-1 as a model system, we identified and quantified the glycosylation of hundreds of cell surface N -glycosylation sites. This approach allowed us to study the glycoforms present at the functional relevant cell surface, omitting immaturely glycosylated proteins present in the secretory pathway. Interestingly, the different stages of N -glycan processing at individual sites displayed at the cell surface were found to correlate with their accessibility for ER-residing processing enzymes, as investigated through molecular dynamics simulations. Using the new approach, we compared N -glycosylation sites of proteins expressed on the cell surface to their counterparts in a total cell lysate, showing profound differences in glycosylation between the subcellular components and indicating the relevance of the method for future studies in understanding contextual glycan functions.
- Published
- 2023
- Full Text
- View/download PDF
11. Galectin-1 induces a tumor-associated macrophage phenotype and upregulates indoleamine 2,3-dioxygenase-1.
- Author
-
Rudjord-Levann AM, Ye Z, Hafkenscheid L, Horn S, Wiegertjes R, Nielsen MAI, Song M, Mathiesen CBK, Stoop J, Stowell S, Straten PT, Leffler H, Vakhrushev SY, Dabelsteen S, Olsen JV, and Wandall HH
- Abstract
Galectins are a group of carbohydrate-binding proteins with a presumed immunomodulatory role and an elusive function on antigen-presenting cells. Here we analyzed the expression of galectin-1 and found upregulation of galectin-1 in the extracellular matrix across multiple tumors. Performing an in-depth and dynamic proteomic and phosphoproteomic analysis of human macrophages stimulated with galectin-1, we show that galectin-1 induces a tumor-associated macrophage phenotype with increased expression of key immune checkpoint protein programmed cell death 1 ligand 1 (PD-L1/CD274) and immunomodulator indoleamine 2,3-dioxygenase-1 (IDO1). Galectin-1 induced IDO1 and its active metabolite kynurenine in a dose-dependent manner through JAK/STAT signaling. In a 3D organotypic tissue model system equipped with genetically engineered tumorigenic epithelial cells, we analyzed the cellular source of galectin-1 in the extracellular matrix and found that galectin-1 is derived from epithelial and stromal cells. Our results highlight the potential of targeting galectin-1 in immunotherapeutic treatment of human cancers., Competing Interests: Unrelated to the presented work, Hans Wandall owns stock and is a consultant for and co-founder of EbuMab, ApS. and GO-Therapeutics, Inc. HL is shareholder in Galecto Biotech AB, a company that is developing galectin inhibitors, (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
12. The SHDRA syndrome-associated gene TMEM260 encodes a protein-specific O-mannosyltransferase.
- Author
-
Larsen ISB, Povolo L, Zhou L, Tian W, Mygind KJ, Hintze J, Jiang C, Hartill V, Prescott K, Johnson CA, Mullegama SV, McConkie-Rosell A, McDonald M, Hansen L, Vakhrushev SY, Schjoldager KT, Clausen H, Worzfeld T, Joshi HJ, and Halim A
- Subjects
- Animals, Glycosylation, Mammals metabolism, Mannose metabolism, Mannosyltransferases genetics, Mannosyltransferases metabolism
- Abstract
Mutations in the TMEM260 gene cause structural heart defects and renal anomalies syndrome, but the function of the encoded protein remains unknown. We previously reported wide occurrence of O-mannose glycans on extracellular immunoglobulin, plexin, transcription factor (IPT) domains found in the hepatocyte growth factor receptor (cMET), macrophage-stimulating protein receptor (RON), and plexin receptors, and further demonstrated that two known protein O-mannosylation systems orchestrated by the POMT1/2 and transmembrane and tetratricopeptide repeat-containing proteins 1-4 gene families were not required for glycosylation of these IPT domains. Here, we report that the TMEM260 gene encodes an ER-located protein O-mannosyltransferase that selectively glycosylates IPT domains. We demonstrate that disease-causing TMEM260 mutations impair O-mannosylation of IPT domains and that TMEM260 knockout in cells results in receptor maturation defects and abnormal growth of 3D cell models. Thus, our study identifies the third protein-specific O-mannosylation pathway in mammals and demonstrates that O-mannosylation of IPT domains serves critical functions during epithelial morphogenesis. Our findings add a new glycosylation pathway and gene to a growing group of congenital disorders of glycosylation.
- Published
- 2023
- Full Text
- View/download PDF
13. A universal GlycoDesign for lysosomal replacement enzymes to improve circulation time and biodistribution.
- Author
-
Chen YH, Tian W, Yasuda M, Ye Z, Song M, Mandel U, Kristensen C, Povolo L, Marques ARA, Čaval T, Heck AJR, Sampaio JL, Johannes L, Tsukimura T, Desnick R, Vakhrushev SY, Yang Z, and Clausen H
- Abstract
Currently available enzyme replacement therapies for lysosomal storage diseases are limited in their effectiveness due in part to short circulation times and suboptimal biodistribution of the therapeutic enzymes. We previously engineered Chinese hamster ovary (CHO) cells to produce α-galactosidase A (GLA) with various N-glycan structures and demonstrated that elimination of mannose-6-phosphate (M6P) and conversion to homogeneous sialylated N-glycans prolonged circulation time and improved biodistribution of the enzyme following a single-dose infusion into Fabry mice. Here, we confirmed these findings using repeated infusions of the glycoengineered GLA into Fabry mice and further tested whether this glycoengineering approach, Long-Acting-GlycoDesign (LAGD), could be implemented on other lysosomal enzymes. LAGD-engineered CHO cells stably expressing a panel of lysosomal enzymes [aspartylglucosamine (AGA), beta-glucuronidase (GUSB), cathepsin D (CTSD), tripeptidyl peptidase (TPP1), alpha-glucosidase (GAA) or iduronate 2-sulfatase (IDS)] successfully converted all M6P-containing N-glycans to complex sialylated N-glycans. The resulting homogenous glycodesigns enabled glycoprotein profiling by native mass spectrometry. Notably, LAGD extended the plasma half-life of all three enzymes tested (GLA, GUSB, AGA) in wildtype mice. LAGD may be widely applicable to lysosomal replacement enzymes to improve their circulatory stability and therapeutic efficacy., Competing Interests: A patent application has been filed by the University of Copenhagen. GlycoDisplay ApS has license rights to the patent application. ZY, WT, CK, and HC are named co-inventors, and ZY, CK, and HC have financial interests in GlycoDisplay ApS. Y-HC is an employee of GlycoDisplay ApS. RD is a Consultant to Genzyme-Sanofi and Sangamo Therapeutics, Inc. He owns founder stock in Amicus Therapeutics and options for Sangmo Therapeutics, Inc. and receives royalities from Genzyme-Sanofi. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Chen, Tian, Yasuda, Ye, Song, Mandel, Kristensen, Povolo, Marques, Čaval, Heck, Sampaio, Johannes, Tsukimura, Desnick, Vakhrushev, Yang and Clausen.)
- Published
- 2023
- Full Text
- View/download PDF
14. Identification of global inhibitors of cellular glycosylation.
- Author
-
Sørensen DM, Büll C, Madsen TD, Lira-Navarrete E, Clausen TM, Clark AE, Garretson AF, Karlsson R, Pijnenborg JFA, Yin X, Miller RL, Chanda SK, Boltje TJ, Schjoldager KT, Vakhrushev SY, Halim A, Esko JD, Carlin AF, Hurtado-Guerrero R, Weigert R, Clausen H, and Narimatsu Y
- Subjects
- Animals, Glycosylation, Glycoproteins metabolism, Polysaccharides metabolism, SARS-CoV-2 metabolism, COVID-19
- Abstract
Small molecule inhibitors of glycosylation enzymes are valuable tools for dissecting glycan functions and potential drug candidates. Screening for inhibitors of glycosyltransferases are mainly performed by in vitro enzyme assays with difficulties moving candidates to cells and animals. Here, we circumvent this by employing a cell-based screening assay using glycoengineered cells expressing tailored reporter glycoproteins. We focused on GalNAc-type O-glycosylation and selected the GalNAc-T11 isoenzyme that selectively glycosylates endocytic low-density lipoprotein receptor (LDLR)-related proteins as targets. Our screen of a limited small molecule compound library did not identify selective inhibitors of GalNAc-T11, however, we identify two compounds that broadly inhibited Golgi-localized glycosylation processes. These compounds mediate the reversible fragmentation of the Golgi system without affecting secretion. We demonstrate how these inhibitors can be used to manipulate glycosylation in cells to induce expression of truncated O-glycans and augment binding of cancer-specific Tn-glycoprotein antibodies and to inhibit expression of heparan sulfate and binding and infection of SARS-CoV-2., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Characterization of TGF-β signaling in a human organotypic skin model reveals that loss of TGF-βRII induces invasive tissue growth.
- Author
-
Ye Z, Kilic G, Dabelsteen S, Marinova IN, Thøfner JFB, Song M, Rudjord-Levann AM, Bagdonaite I, Vakhrushev SY, Brakebusch CH, Olsen JV, and Wandall HH
- Subjects
- Humans, Cell Differentiation, Cell Proliferation, Skin, Signal Transduction, Transforming Growth Factor beta1
- Abstract
Transforming growth factor-β (TGF-β) signaling regulates various aspects of cell growth and differentiation and is often dysregulated in human cancers. We combined genetic engineering of a human organotypic three-dimensional (3D) skin model with global quantitative proteomics and phosphoproteomics to dissect the importance of essential components of the TGF-β signaling pathway, including the ligands TGF-β1, TGF-β2, and TGF-β3, the receptor TGF-βRII, and the intracellular effector SMAD4. Consistent with the antiproliferative effects of TGF-β signaling, the loss of TGF-β1 or SMAD4 promoted cell cycling and delayed epidermal differentiation. The loss of TGF-βRII, which abrogates both SMAD4-dependent and SMAD4-independent downstream signaling, more strongly affected cell proliferation and differentiation than did loss of SMAD4, and it induced invasive growth. TGF-βRII knockout reduced cell-matrix interactions, and the production of matrix proteins increased the production of cancer-associated cell-cell adhesion proteins and proinflammatory mediators and increased mitogen-activated protein kinase (MAPK) signaling. Inhibiting the activation of the ERK and p38 MAPK pathways blocked the development of the invasive phenotype upon the loss of TGF-βRII. This study provides a framework for exploring TGF-β signaling pathways in human epithelial tissue homeostasis and transformation using genetic engineering, 3D tissue models, and high-throughput quantitative proteomics and phosphoproteomics.
- Published
- 2022
- Full Text
- View/download PDF
16. Global mapping of GalNAc-T isoform-specificities and O-glycosylation site-occupancy in a tissue-forming human cell line.
- Author
-
Nielsen MI, de Haan N, Kightlinger W, Ye Z, Dabelsteen S, Li M, Jewett MC, Bagdonaite I, Vakhrushev SY, and Wandall HH
- Subjects
- Humans, Glycosylation, Protein Isoforms genetics, Protein Isoforms metabolism, Cell Line, Mucins metabolism, Polysaccharides, Proteome metabolism, N-Acetylgalactosaminyltransferases genetics, N-Acetylgalactosaminyltransferases metabolism
- Abstract
Mucin-type-O-glycosylation on proteins is integrally involved in human health and disease and is coordinated by an enzyme family of 20 N-acetylgalactosaminyltransferases (GalNAc-Ts). Detailed knowledge on the biological effects of site-specific O-glycosylation is limited due to lack of information on specific glycosylation enzyme activities and O-glycosylation site-occupancies. Here we present a systematic analysis of the isoform-specific targets of all GalNAc-Ts expressed within a tissue-forming human skin cell line, and demonstrate biologically significant effects of O-glycan initiation on epithelial formation. We find over 300 unique glycosylation sites across a diverse set of proteins specifically regulated by one of the GalNAc-T isoforms, consistent with their impact on the tissue phenotypes. Notably, we discover a high variability in the O-glycosylation site-occupancy of 70 glycosylated regions of secreted proteins. These findings revisit the relevance of individual O-glycosylation sites in the proteome, and provide an approach to establish which sites drive biological functions., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
17. Role of N- Glycosylation in FcγRIIIa interaction with IgG.
- Author
-
Van Coillie J, Schulz MA, Bentlage AEH, de Haan N, Ye Z, Geerdes DM, van Esch WJE, Hafkenscheid L, Miller RL, Narimatsu Y, Vakhrushev SY, Yang Z, Vidarsson G, and Clausen H
- Subjects
- Animals, Antibody-Dependent Cell Cytotoxicity, Glycosylation, Mammals, Polysaccharides metabolism, Immunoglobulin G, Receptors, IgG metabolism
- Abstract
Immunoglobulins G (IgG) and their Fc gamma receptors (FcγRs) play important roles in our immune system. The conserved N- glycan in the Fc region of IgG1 impacts interaction of IgG with FcγRs and the resulting effector functions, which has led to the design of antibody therapeutics with greatly improved antibody-dependent cell cytotoxicity (ADCC) activities. Studies have suggested that also N- glycosylation of the FcγRIII affects receptor interactions with IgG, but detailed studies of the interaction of IgG1 and FcγRIIIa with distinct N -glycans have been hindered by the natural heterogeneity in N- glycosylation. In this study, we employed comprehensive genetic engineering of the N- glycosylation capacities in mammalian cell lines to express IgG1 and FcγRIIIa with different N- glycan structures to more generally explore the role of N- glycosylation in IgG1:FcγRIIIa binding interactions. We included FcγRIIIa variants of both the 158F and 158V allotypes and investigated the key N- glycan features that affected binding affinity. Our study confirms that afucosylated IgG1 has the highest binding affinity to oligomannose FcγRIIIa, a glycan structure commonly found on Asn162 on FcγRIIIa expressed by NK cells but not monocytes or recombinantly expressed FcγRIIIa., Competing Interests: The University of Copenhagen has filed a patent application for the cell-based display platform. GlycoDisplay Aps, Copenhagen, Denmark, has obtained a license in the field of the patent application. Authors YN, ZY, and HC are co-founders of GlycoDisplay Aps and hold ownerships in the company. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Van Coillie, Schulz, Bentlage, de Haan, Ye, Geerdes, van Esch, Hafkenscheid, Miller, Narimatsu, Vakhrushev, Yang, Vidarsson and Clausen.)
- Published
- 2022
- Full Text
- View/download PDF
18. Reference Genes across Nine Brain Areas of Wild Type and Prader-Willi Syndrome Mice: Assessing Differences in Igfbp7 , Pcsk1 , Nhlh2 and Nlgn3 Expression.
- Author
-
Kummerfeld DM, Skryabin BV, Brosius J, Vakhrushev SY, and Rozhdestvensky TS
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Brain metabolism, Disease Models, Animal, Exons, Genomic Imprinting, Humans, Mice, Proprotein Convertase 1 genetics, Proprotein Convertase 1 metabolism, RNA, Small Nucleolar genetics, RNA, Small Nucleolar metabolism, Prader-Willi Syndrome genetics, Prader-Willi Syndrome metabolism
- Abstract
Prader−Willi syndrome (PWS) is a complex neurodevelopmental disorder caused by the deletion or inactivation of paternally expressed imprinted genes at the chromosomal region 15q11−q13. The PWS-critical region (PWScr) harbors tandemly repeated non-protein coding IPW-A exons hosting the intronic SNORD116 snoRNA gene array that is predominantly expressed in brain. Paternal deletion of PWScr is associated with key PWS symptoms in humans and growth retardation in mice (PWScr model). Dysregulation of the hypothalamic−pituitary axis (HPA) is thought to be causally involved in the PWS phenotype. Here we performed a comprehensive reverse transcription quantitative PCR (RT-qPCR) analysis across nine different brain regions of wild-type (WT) and PWScr mice to identify stably expressed reference genes. Four methods (Delta Ct, BestKeeper, Normfinder and Genorm) were applied to rank 11 selected reference gene candidates according to their expression stability. The resulting panel consists of the top three most stably expressed genes suitable for gene-expression profiling and comparative transcriptome analysis of WT and/or PWScr mouse brain regions. Using these reference genes, we revealed significant differences in the expression patterns of Igfbp7, Nlgn3 and three HPA associated genes: Pcsk1, Pcsk2 and Nhlh2 across investigated brain regions of wild-type and PWScr mice. Our results raise a reasonable doubt on the involvement of the Snord116 in posttranscriptional regulation of Nlgn3 and Nhlh2 genes. We provide a valuable tool for expression analysis of specific genes across different areas of the mouse brain and for comparative investigation of PWScr mouse models to discover and verify different regulatory pathways affecting this complex disorder.
- Published
- 2022
- Full Text
- View/download PDF
19. A Bacterial Mannose Binding Lectin as a Tool for the Enrichment of C- and O-Mannosylated Peptides.
- Author
-
Hütte HJ, Tiemann B, Shcherbakova A, Grote V, Hoffmann M, Povolo L, Lommel M, Strahl S, Vakhrushev SY, Rapp E, Buettner FFR, Halim A, Imberty A, and Bakker H
- Subjects
- Glycopeptides metabolism, Glycosylation, HEK293 Cells, Humans, Lectins chemistry, Burkholderia cenocepacia chemistry, Burkholderia cenocepacia metabolism, Mannose chemistry
- Abstract
Mass spectrometry (MS) easily detects C-mannosylated peptides from purified proteins but not from complex biological samples. Enrichment of specific glycopeptides by lectin affinity prior to MS analysis has been widely applied to support glycopeptide identification but was until now not available for C-mannosylated peptides. Here, we used the α-mannose-specific Burkholderia cenocepacia lectin A (BC2L-A) and show that, in addition to its previously demonstrated high-mannose N-glycan binding capability, this lectin is able to retain C- and O-mannosylated peptides. Besides testing binding abilities to standard peptides, we applied BC2L-A affinity to enrich C-mannosylated peptides from complex samples of tryptic digests of HEK293 and MCF10A whole cell extracts, which led to the identification of novel C-mannosylation sites. In conclusion, BC2L-A enabled specific enrichment of C- and O-mannosylated peptides and might have superior properties over other mannose binding lectins for this purpose.
- Published
- 2022
- Full Text
- View/download PDF
20. Exploring the glycosylation of mucins by use of O-glycodomain reporters recombinantly expressed in glycoengineered HEK293 cells.
- Author
-
Konstantinidi A, Nason R, Čaval T, Sun L, Sørensen DM, Furukawa S, Ye Z, Vincentelli R, Narimatsu Y, Vakhrushev SY, and Clausen H
- Subjects
- Amino Acid Sequence, Animals, Glycosylation, HEK293 Cells, Humans, Polysaccharides genetics, Protein Domains, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sheep, Mucins metabolism
- Abstract
Mucins and glycoproteins with mucin-like regions contain densely O-glycosylated domains often found in tandem repeat (TR) sequences. These O-glycodomains have traditionally been difficult to characterize because of their resistance to proteolytic digestion, and knowledge of the precise positions of O-glycans is particularly limited for these regions. Here, we took advantage of a recently developed glycoengineered cell-based platform for the display and production of mucin TR reporters with custom-designed O-glycosylation to characterize O-glycodomains derived from mucins and mucin-like glycoproteins. We combined intact mass and bottom-up site-specific analysis for mapping O-glycosites in the mucins, MUC2, MUC20, MUC21, protein P-selectin-glycoprotein ligand 1, and proteoglycan syndecan-3. We found that all the potential Ser/Thr positions in these O-glycodomains were O-glycosylated when expressed in human embryonic kidney 293 SimpleCells (Tn-glycoform). Interestingly, we found that all potential Ser/Thr O-glycosites in TRs derived from secreted mucins and most glycosites from transmembrane mucins were almost fully occupied, whereas TRs from a subset of transmembrane mucins were less efficiently processed. We further used the mucin TR reporters to characterize cleavage sites of glycoproteases StcE (secreted protease of C1 esterase inhibitor from EHEC) and BT4244, revealing more restricted substrate specificities than previously reported. Finally, we conducted a bottom-up analysis of isolated ovine submaxillary mucin, which supported our findings that mucin TRs in general are efficiently O-glycosylated at all potential glycosites. This study provides insight into O-glycosylation of mucins and mucin-like domains, and the strategies developed open the field for wider analysis of native mucins., Competing Interests: Conflict of interest The University of Copenhagen has filed a patent application on the cell-based display platform. GlycoDisplay Aps, Copenhagen, Denmark, has obtained a license to the field of the patent application. Y. N. and H. C. are cofounders of GlycoDisplay Aps and hold ownerships in the company. All the other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
21. In-Depth Profiling of O -Glycan Isomers in Human Cells Using C18 Nanoliquid Chromatography-Mass Spectrometry and Glycogenomics.
- Author
-
de Haan N, Narimatsu Y, Koed Møller Aasted M, Larsen ISB, Marinova IN, Dabelsteen S, Vakhrushev SY, and Wandall HH
- Subjects
- Glycosylation, Humans, Isomerism, Mass Spectrometry, Chromatography, Polysaccharides chemistry
- Abstract
O -Glycosylation is an omnipresent modification of the human proteome affecting many cellular functions, including protein cleavage, protein folding, and cellular signaling, interactions, and trafficking. The functions are governed by differentially regulated O -glycan types and terminal structures. It is therefore essential to develop analytical methods that facilitate the annotation of O -glycans in biological material. While various successful strategies for the in-depth profiling of released O -glycans have been reported, these methods are often limitedly accessible to the nonspecialist or challenged by the high abundance of O -glycan structural isomers. Here, we developed a high-throughput sample preparation approach for the nonreductive release and characterization of O -glycans from human cell material. Reducing-end labeling allowed efficient isomer separation and detection using C18 nanoliquid chromatography coupled to Orbitrap mass spectrometry. Using the method in combination with a library of genetically glycoengineered cells displaying defined O -glycan types and structures, we were able to annotate individual O -glycan structural isomers from a complex mixture. Applying the method in a model system of human keratinocytes, we found a wide variety of O -glycan structures, including O -fucose, O -glucose, O -GlcNAc, and O -GalNAc glycosylation, with the latter carrying both elongated core1 and core2 structures and varying numbers of fucoses and sialic acids. The method, including the now well-characterized standards, provides the opportunity to study glycomic changes in human tissue and disease models using rather mainstream analytical equipment.
- Published
- 2022
- Full Text
- View/download PDF
22. Atomic and Specificity Details of Mucin 1 O -Glycosylation Process by Multiple Polypeptide GalNAc-Transferase Isoforms Unveiled by NMR and Molecular Modeling.
- Author
-
Coelho H, Rivas ML, Grosso AS, Diniz A, Soares CO, Francisco RA, Dias JS, Compañon I, Sun L, Narimatsu Y, Vakhrushev SY, Clausen H, Cabrita EJ, Jiménez-Barbero J, Corzana F, Hurtado-Guerrero R, and Marcelo F
- Abstract
The large family of polypeptide GalNAc-transferases (GalNAc-Ts) controls with precision how GalNAc O -glycans are added in the tandem repeat regions of mucins ( e.g. , MUC1). However, the structural features behind the creation of well-defined and clustered patterns of O -glycans in mucins are poorly understood. In this context, herein, we disclose the full process of MUC1 O -glycosylation by GalNAc-T2/T3/T4 isoforms by NMR spectroscopy assisted by molecular modeling protocols. By using MUC1, with four tandem repeat domains as a substrate, we confirmed the glycosylation preferences of different GalNAc-Ts isoforms and highlighted the importance of the lectin domain in the glycosylation site selection after the addition of the first GalNAc residue. In a glycosylated substrate, with yet multiple acceptor sites, the lectin domain contributes to orientate acceptor sites to the catalytic domain. Our experiments suggest that during this process, neighboring tandem repeats are critical for further glycosylation of acceptor sites by GalNAc-T2/T4 in a lectin-assisted manner. Our studies also show local conformational changes in the peptide backbone during incorporation of GalNAc residues, which might explain GalNAc-T2/T3/T4 fine specificities toward the MUC1 substrate. Interestingly, we postulate that a specific salt-bridge and the inverse γ-turn conformation of the PDTRP sequence in MUC1 are the main structural motifs behind the GalNAc-T4 specificity toward this region. In addition, in-cell analysis shows that the GalNAc-T4 isoform is the only isoform glycosylating the Thr of the immunogenic epitope PDTRP in vivo , which highlights the relevance of GalNAc-T4 in the glycosylation of this epitope. Finally, the NMR methodology established herein can be extended to other glycosyltransferases, such as C1GalT1 and ST6GalNAc-I, to determine the specificity toward complex mucin acceptor substrates., Competing Interests: The authors declare no competing financial interest., (© 2022 The Authors. Published by American Chemical Society.)
- Published
- 2022
- Full Text
- View/download PDF
23. CNOT6: A Novel Regulator of DNA Mismatch Repair.
- Author
-
Song P, Liu S, Liu D, Keijzers G, Bakula D, Duan S, de Wind N, Ye Z, Vakhrushev SY, Scheibye-Knudsen M, and Rasmussen LJ
- Subjects
- Apoptosis genetics, Humans, Male, RNA, Messenger genetics, RNA, Messenger metabolism, DNA Mismatch Repair genetics, DNA Replication
- Abstract
DNA mismatch repair (MMR) is a highly conserved pathway that corrects both base-base mispairs and insertion-deletion loops (IDLs) generated during DNA replication. Defects in MMR have been linked to carcinogenesis and drug resistance. However, the regulation of MMR is poorly understood. Interestingly, CNOT6 is one of four deadenylase subunits in the conserved CCR4-NOT complex and it targets poly(A) tails of mRNAs for degradation. CNOT6 is overexpressed in acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML) and androgen-independent prostate cancer cells, which suggests that an altered expression of CNOT6 may play a role in tumorigenesis. Here, we report that a depletion of CNOT6 sensitizes human U2OS cells to N-methyl-N'nitro-N-nitrosoguanidine (MNNG) and leads to enhanced apoptosis. We also demonstrate that the depletion of CNOT6 upregulates MMR and decreases the mutation frequency in MMR-proficient cells. Furthermore, the depletion of CNOT6 increases the stability of mRNA transcripts from MMR genes, leading to the increased expression of MMR proteins. Our work provides insight into a novel CNOT6-dependent mechanism for regulating MMR.
- Published
- 2022
- Full Text
- View/download PDF
24. Installation of O-glycan sulfation capacities in human HEK293 cells for display of sulfated mucins.
- Author
-
Sun L, Konstantinidi A, Ye Z, Nason R, Zhang Y, Büll C, Kahl-Knutson B, Hansen L, Leffler H, Vakhrushev SY, Yang Z, Clausen H, and Narimatsu Y
- Subjects
- Glycoproteins metabolism, HEK293 Cells, Humans, Kidney metabolism, Polysaccharides metabolism, Sulfotransferases metabolism, Mucins metabolism, Sulfates metabolism
- Abstract
The human genome contains at least 35 genes that encode Golgi sulfotransferases that function in the secretory pathway, where they are involved in decorating glycosaminoglycans, glycolipids, and glycoproteins with sulfate groups. Although a number of important interactions by proteins such as selectins, galectins, and sialic acid-binding immunoglobulin-like lectins are thought to mainly rely on sulfated O-glycans, our insight into the sulfotransferases that modify these glycoproteins, and in particular GalNAc-type O-glycoproteins, is limited. Moreover, sulfated mucins appear to accumulate in respiratory diseases, arthritis, and cancer. To explore further the genetic and biosynthetic regulation of sulfated O-glycans, here we expanded a cell-based glycan array in the human embryonic kidney 293 (HEK293) cell line with sulfation capacities. We stably engineered O-glycan sulfation capacities in HEK293 cells by site-directed knockin of sulfotransferase genes in combination with knockout of genes to eliminate endogenous O-glycan branching (core2 synthase gene GCNT1) and/or sialylation capacities in order to provide simplified substrates (core1 Galβ1-3GalNAcα1-O-Ser/Thr) for the introduced sulfotransferases. Expression of the galactose 3-O-sulfotransferase 2 in HEK293 cells resulted in sulfation of core1 and core2 O-glycans, whereas expression of galactose 3-O-sulfotransferase 4 resulted in sulfation of core1 only. We used the engineered cell library to dissect the binding specificity of galectin-4 and confirmed binding to the 3-O-sulfo-core1 O-glycan. This is a first step toward expanding the emerging cell-based glycan arrays with the important sulfation modification for display and production of glycoconjugates with sulfated O-glycans., Competing Interests: Conflict of interest University of Copenhagen has filed a patent application on the cell-based display platform. GlycoDisplay ApS, Copenhagen, Denmark, has obtained a license to the field of the patent application. Y. N. and H. C. are cofounders of GlycoDisplay ApS and hold ownerships in the company. H. L. is cofounder and consultant with Galecto, Inc. All other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
25. Author Correction: Community evaluation of glycoproteomics informatics solutions reveals high-performance search strategies for serum glycopeptide analysis.
- Author
-
Kawahara R, Chernykh A, Alagesan K, Bern M, Cao W, Chalkley RJ, Cheng K, Choo MS, Edwards N, Goldman R, Hoffmann M, Hu Y, Huang Y, Kim JY, Kletter D, Liquet B, Liu M, Mechref Y, Meng B, Neelamegham S, Nguyen-Khuong T, Nilsson J, Pap A, Park GW, Parker BL, Pegg CL, Penninger JM, Phung TK, Pioch M, Rapp E, Sakalli E, Sanda M, Schulz BL, Scott NE, Sofronov G, Stadlmann J, Vakhrushev SY, Woo CM, Wu HY, Yang P, Ying W, Zhang H, Zhang Y, Zhao J, Zaia J, Haslam SM, Palmisano G, Yoo JS, Larson G, Khoo KH, Medzihradszky KF, Kolarich D, Packer NH, and Thaysen-Andersen M
- Published
- 2022
- Full Text
- View/download PDF
26. Dissecting structure-function of 3-O-sulfated heparin and engineered heparan sulfates.
- Author
-
Karlsson R, Chopra P, Joshi A, Yang Z, Vakhrushev SY, Clausen TM, Painter CD, Szekeres GP, Chen YH, Sandoval DR, Hansen L, Esko JD, Pagel K, Dyer DP, Turnbull JE, Clausen H, Boons GJ, and Miller RL
- Abstract
Heparan sulfate (HS) polysaccharides are master regulators of diverse biological processes via sulfated motifs that can recruit specific proteins. 3-O-sulfation of HS/heparin is crucial for anticoagulant activity, but despite emerging evidence for roles in many other functions, a lack of tools for deciphering structure-function relationships has hampered advances. Here, we describe an approach integrating synthesis of 3-O-sulfated standards, comprehensive HS disaccharide profiling, and cell engineering to address this deficiency. Its application revealed previously unseen differences in 3-O-sulfated profiles of clinical heparins and 3- O -sulfotransferase (HS3ST)–specific variations in cell surface HS profiles. The latter correlated with functional differences in anticoagulant activity and binding to platelet factor 4 (PF4), which underlies heparin-induced thrombocytopenia, a known side effect of heparin. Unexpectedly, cells expressing the HS3ST4 isoenzyme generated HS with potent anticoagulant activity but weak PF4 binding. The data provide new insights into 3- O -sulfate structure-function and demonstrate proof of concept for tailored cell-based synthesis of next-generation heparins.
- Published
- 2021
- Full Text
- View/download PDF
27. Community evaluation of glycoproteomics informatics solutions reveals high-performance search strategies for serum glycopeptide analysis.
- Author
-
Kawahara R, Chernykh A, Alagesan K, Bern M, Cao W, Chalkley RJ, Cheng K, Choo MS, Edwards N, Goldman R, Hoffmann M, Hu Y, Huang Y, Kim JY, Kletter D, Liquet B, Liu M, Mechref Y, Meng B, Neelamegham S, Nguyen-Khuong T, Nilsson J, Pap A, Park GW, Parker BL, Pegg CL, Penninger JM, Phung TK, Pioch M, Rapp E, Sakalli E, Sanda M, Schulz BL, Scott NE, Sofronov G, Stadlmann J, Vakhrushev SY, Woo CM, Wu HY, Yang P, Ying W, Zhang H, Zhang Y, Zhao J, Zaia J, Haslam SM, Palmisano G, Yoo JS, Larson G, Khoo KH, Medzihradszky KF, Kolarich D, Packer NH, and Thaysen-Andersen M
- Subjects
- Glycosylation, Humans, Proteome metabolism, Tandem Mass Spectrometry, Glycopeptides blood, Glycoproteins blood, Informatics methods, Proteome analysis, Proteomics methods, Research Personnel statistics & numerical data, Software
- Abstract
Glycoproteomics is a powerful yet analytically challenging research tool. Software packages aiding the interpretation of complex glycopeptide tandem mass spectra have appeared, but their relative performance remains untested. Conducted through the HUPO Human Glycoproteomics Initiative, this community study, comprising both developers and users of glycoproteomics software, evaluates solutions for system-wide glycopeptide analysis. The same mass spectrometrybased glycoproteomics datasets from human serum were shared with participants and the relative team performance for N- and O-glycopeptide data analysis was comprehensively established by orthogonal performance tests. Although the results were variable, several high-performance glycoproteomics informatics strategies were identified. Deep analysis of the data revealed key performance-associated search parameters and led to recommendations for improved 'high-coverage' and 'high-accuracy' glycoproteomics search solutions. This study concludes that diverse software packages for comprehensive glycopeptide data analysis exist, points to several high-performance search strategies and specifies key variables that will guide future software developments and assist informatics decision-making in glycoproteomics., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
28. Systematic Evaluation of Fragmentation Methods for Unlabeled and Isobaric Mass Tag-Labeled O-Glycopeptides.
- Author
-
Mao Y, Wang S, Zhao Y, Konstantinidi A, Sun L, Ye Z, and Vakhrushev SY
- Subjects
- Electron Transport, Glycosylation, Ions, Glycopeptides metabolism, Tandem Mass Spectrometry
- Abstract
Dissecting site-specific functions of O-glycosylation requires simultaneous identification and quantification of differentially expressed O-glycopeptides by mass spectrometry. However, different dissociation methods have not been systematically compared in their performance in terms of identification, glycosite localization, and quantification with isobaric labeling. Here, we conducted this comparison on highly enriched unlabeled O-glycopeptides with higher-energy collision dissociation (HCD), electron-transfer/collision-induced dissociation (ETciD), and electron transfer/higher-energy collisional dissociation (EThcD), concluding that ETciD and EThcD with optimal supplemental activation resulted in superior identification of glycopeptides and unambiguous site localizations than HCD in a database search by Sequest HT. We later described a pseudo-EThcD strategy that in silico concatenates the electron transfer dissociation spectrum with the paired HCD spectrum acquired sequentially for the same precursor ions, which combines the identification advantage of ETciD/EThcD with the superior reporter ion quality of HCD. We demonstrated its improvements in identification and quantification of isobaric mass tag-labeled O-glycopeptides and showcased the discovery of the specific glycosites of GalNAc transferase 11 (GALNT11) in HepG2 cells.
- Published
- 2021
- Full Text
- View/download PDF
29. FUT8-Directed Core Fucosylation of N-glycans Is Regulated by the Glycan Structure and Protein Environment.
- Author
-
García-García A, Serna S, Yang Z, Delso I, Taleb V, Hicks T, Artschwager R, Vakhrushev SY, Clausen H, Angulo J, Corzana F, Reichardt NC, and Hurtado-Guerrero R
- Abstract
FUT8 is an essential α-1,6-fucosyltransferase that fucosylates the innermost GlcNAc of N-glycans, a process called core fucosylation. In vitro , FUT8 exhibits substrate preference for the biantennary complex N-glycan oligosaccharide (G0), but the role of the underlying protein/peptide to which N-glycans are attached remains unclear. Here, we explored the FUT8 enzyme with a series of N-glycan oligosaccharides, N-glycopeptides, and an Asn-linked oligosaccharide. We found that the underlying peptide plays a role in fucosylation of paucimannose (low mannose) and high-mannose N-glycans but not for complex-type N-glycans. Using saturation transfer difference (STD) NMR spectroscopy, we demonstrate that FUT8 recognizes all sugar units of the G0 N-glycan and most of the amino acid residues (Asn-X-Thr) that serve as a recognition sequon for the oligosaccharyltransferase (OST). The largest STD signals were observed in the presence of GDP, suggesting that prior FUT8 binding to GDP-β-l-fucose (GDP-Fuc) is required for an optimal recognition of N-glycans. We applied genetic engineering of glycosylation capacities in CHO cells to evaluate FUT8 core fucosylation of high-mannose and complex-type N-glycans in cells with a panel of well-characterized therapeutic N-glycoproteins. This confirmed that core fucosylation mainly occurs on complex-type N-glycans, although clearly only at selected glycosites. Eliminating the capacity for complex-type glycosylation in cells (KO mgat1 ) revealed that glycosites with complex-type N-glycans when converted to high mannose lost the core Fuc. Interestingly, however, for erythropoietin that is uncommon among the tested glycoproteins in efficiently acquiring tetra-antennary N-glycans, two out of three N-glycosites obtained Fuc on the high-mannose N-glycans. An examination of the N-glycosylation sites of several protein crystal structures indicates that core fucosylation is mostly affected by the accessibility and nature of the N-glycan and not by the nature of the underlying peptide sequence. These data have further elucidated the different FUT8 acceptor substrate specificities both in vitro and in vivo in cells, revealing different mechanisms for promoting core fucosylation., Competing Interests: The authors declare no competing financial interest., (© 2021 American Chemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
30. Display of the human mucinome with defined O-glycans by gene engineered cells.
- Author
-
Nason R, Büll C, Konstantinidi A, Sun L, Ye Z, Halim A, Du W, Sørensen DM, Durbesson F, Furukawa S, Mandel U, Joshi HJ, Dworkin LA, Hansen L, David L, Iverson TM, Bensing BA, Sullam PM, Varki A, Vries E, de Haan CAM, Vincentelli R, Henrissat B, Vakhrushev SY, Clausen H, and Narimatsu Y
- Subjects
- Genetic Engineering, Glycosylation, HEK293 Cells, Humans, Microbiota, Mucin-1 genetics, Mucin-1 metabolism, Mucins metabolism, Mucous Membrane metabolism, Polysaccharides genetics, Polysaccharides metabolism
- Abstract
Mucins are a large family of heavily O-glycosylated proteins that cover all mucosal surfaces and constitute the major macromolecules in most body fluids. Mucins are primarily defined by their variable tandem repeat (TR) domains that are densely decorated with different O-glycan structures in distinct patterns, and these arguably convey much of the informational content of mucins. Here, we develop a cell-based platform for the display and production of human TR O-glycodomains (~200 amino acids) with tunable structures and patterns of O-glycans using membrane-bound and secreted reporters expressed in glycoengineered HEK293 cells. Availability of defined mucin TR O-glycodomains advances experimental studies into the versatile role of mucins at the interface with pathogenic microorganisms and the microbiome, and sparks new strategies for molecular dissection of specific roles of adhesins, glycoside hydrolases, glycopeptidases, viruses and other interactions with mucin TRs as highlighted by examples.
- Published
- 2021
- Full Text
- View/download PDF
31. Quantitative characterization of O-GalNAc glycosylation.
- Author
-
Čaval T, de Haan N, Konstantinidi A, and Vakhrushev SY
- Subjects
- Glycosylation, Mass Spectrometry, Protein Processing, Post-Translational, Glycoproteins metabolism, Polysaccharides
- Abstract
O-GalNAc type glycosylation is an abundant and complex protein modification. Recent developments in mass spectrometry resulted in significant success in quantitative analysis of O-GalNAc glycosylation. The analysis of released O-GalNAc type glycans expanded our horizons of understanding the glycome of various biological models. The site-specific analysis of glycosylation micro-heterogeneity of purified proteins opened perspectives for the improved design of glycoprotein therapeutics. Advanced gene editing and chemical technologies applied to O-glycoproteomics enabled to identify O-GalNAc glycosylation at unprecedented depth. Progress in the analysis of intact glycoproteins under native and reduced conditions enabled the monitoring of glycosylation proteoform variants. Despite of the astonishing results in quantitative O-GalNAc glycoproteomics, site-specific mapping of the full O-GalNAc structural repertoire in complex samples is yet a long way off. Here, we summarize the most common quantitative strategies in O-GalNAc glycoproteomics, review recent progress and discuss benefits and limitations of the various approaches in the field., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Drosophila O -GlcNAcase Mutants Reveal an Expanded Glycoproteome and Novel Growth and Longevity Phenotypes.
- Author
-
Akan I, Halim A, Vakhrushev SY, Clausen H, and Hanover JA
- Subjects
- Animals, Body Size, Drosophila Proteins metabolism, Drosophila melanogaster anatomy & histology, Female, Gene Ontology, Histone-Lysine N-Methyltransferase metabolism, Male, Phenotype, Polytene Chromosomes metabolism, Wings, Animal enzymology, Drosophila melanogaster enzymology, Drosophila melanogaster growth & development, Glycoproteins metabolism, Longevity, Mutation genetics, Proteome metabolism, beta-N-Acetylhexosaminidases genetics
- Abstract
The reversible posttranslational O -GlcNAc modification of serine or threonine residues of intracellular proteins is involved in many cellular events from signaling cascades to epigenetic and transcriptional regulation. O -GlcNAcylation is a conserved nutrient-dependent process involving two enzymes, with O -GlcNAc transferase (OGT) adding O -GlcNAc and with O -GlcNAcase (OGA) removing it in a manner that's protein- and context-dependent. O -GlcNAcylation is essential for epigenetic regulation of gene expression through its action on Polycomb and Trithorax and COMPASS complexes. However, the important role of O -GlcNAc in adult life and health span has been largely unexplored, mainly due the lack of available model systems. Cataloging the O -GlcNAc proteome has proven useful in understanding the biology of this modification in vivo. In this study, we leveraged a recently developed oga knockout fly mutant to identify the O -GlcNAcylated proteins in adult Drosophila melanogaster . The adult O -GlcNAc proteome revealed many proteins related to cell and organismal growth, development, differentiation, and epigenetics. We identified many O -GlcNAcylated proteins that play a role in increased growth and decreased longevity, including HCF, SIN3A, LOLA, KISMET, ATX2, SHOT, and FOXO. Interestingly, oga mutant flies are larger and have a shorter life span compared to wild type flies, suggesting increased O -GlcNAc results in increased growth. Our results suggest that O -GlcNAc alters the function of many proteins related to transcription, epigenetic modification and signaling pathways that regulate growth rate and longevity. Therefore, our findings highlight the importance of O -GlcNAc in growth and life span in adult Drosophila .
- Published
- 2021
- Full Text
- View/download PDF
33. Site-Specific O-Glycosylation Analysis of SARS-CoV-2 Spike Protein Produced in Insect and Human Cells.
- Author
-
Bagdonaite I, Thompson AJ, Wang X, Søgaard M, Fougeroux C, Frank M, Diedrich JK, Yates JR 3rd, Salanti A, Vakhrushev SY, Paulson JC, and Wandall HH
- Subjects
- Amino Acid Motifs, Animals, Cell Line, Glycosylation, Humans, Insecta, Polysaccharides metabolism, SARS-CoV-2 genetics, Species Specificity, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, COVID-19 virology, SARS-CoV-2 chemistry, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus metabolism
- Abstract
Enveloped viruses hijack not only the host translation processes, but also its glycosylation machinery, and to a variable extent cover viral surface proteins with tolerogenic host-like structures. SARS-CoV-2 surface protein S presents as a trimer on the viral surface and is covered by a dense shield of N-linked glycans, and a few O-glycosites have been reported. The location of O-glycans is controlled by a large family of initiating enzymes with variable expression in cells and tissues and hence is difficult to predict. Here, we used our well-established O-glycoproteomic workflows to map the precise positions of O-linked glycosylation sites on three different entities of protein S-insect cell or human cell-produced ectodomains, or insect cell derived receptor binding domain (RBD). In total 25 O-glycosites were identified, with similar patterns in the two ectodomains of different cell origin, and a distinct pattern of the monomeric RBD. Strikingly, 16 out of 25 O-glycosites were located within three amino acids from known N-glycosites. However, O-glycosylation was primarily found on peptides that were unoccupied by N-glycans, and otherwise had low overall occupancy. This suggests possible complementary functions of O-glycans in immune shielding and negligible effects of O-glycosylation on subunit vaccine design for SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
34. The Role of Data-Independent Acquisition for Glycoproteomics.
- Author
-
Ye Z and Vakhrushev SY
- Subjects
- Animals, Glycosylation, Humans, Proteome, Glycoproteins, Proteomics methods
- Abstract
Data-independent acquisition (DIA) is now an emerging method in bottom-up proteomics and capable of achieving deep proteome coverage and accurate label-free quantification. However, for post-translational modifications, such as glycosylation, DIA methodology is still in the early stage of development. The full characterization of glycoproteins requires site-specific glycan identification as well as subsequent quantification of glycan structures at each site. The tremendous complexity of glycosylation represents a significant analytical challenge in glycoproteomics. This review focuses on the development and perspectives of DIA methodology for N- and O-linked glycoproteomics and posits that DIA-based glycoproteomics could be a method of choice to address some of the challenging aspects of glycoproteomics. First, the current challenges in glycoproteomics and the basic principles of DIA are briefly introduced. DIA-based glycoproteomics is then summarized and described into four aspects based on the actual samples. Finally, we discussed the important challenges and future perspectives in the field. We believe that DIA can significantly facilitate glycoproteomic studies and contribute to the development of future advanced tools and approaches in the field of glycoproteomics., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
35. Essential Functions of Glycans in Human Epithelia Dissected by a CRISPR-Cas9-Engineered Human Organotypic Skin Model.
- Author
-
Dabelsteen S, Pallesen EMH, Marinova IN, Nielsen MI, Adamopoulou M, Rømer TB, Levann A, Andersen MM, Ye Z, Thein D, Bennett EP, Büll C, Moons SJ, Boltje T, Clausen H, Vakhrushev SY, Bagdonaite I, and Wandall HH
- Subjects
- Gene Library, Glycoproteins genetics, Glycosylation, Humans, Skin metabolism, Skin pathology, CRISPR-Cas Systems genetics, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Epithelium physiology, Polysaccharides genetics
- Abstract
The glycome undergoes characteristic changes during histogenesis and organogenesis, but our understanding of the importance of select glycan structures for tissue formation and homeostasis is incomplete. Here, we present a human organotypic platform that allows genetic dissection of cellular glycosylation capacities and systematic interrogation of the roles of distinct glycan types in tissue formation. We used CRISPR-Cas9 gene targeting to generate a library of 3D organotypic skin tissues that selectively differ in their capacity to produce glycan structures on the main types of N- and O-linked glycoproteins and glycolipids. This tissue library revealed distinct changes in skin formation associated with a loss of features for all tested glycoconjugates. The organotypic skin model provides phenotypic cues for the distinct functions of glycoconjugates and serves as a unique resource for further genetic dissection and identification of the specific structural features involved. The strategy is also applicable to other organotypic tissue models., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
36. An atlas of O-linked glycosylation on peptide hormones reveals diverse biological roles.
- Author
-
Madsen TD, Hansen LH, Hintze J, Ye Z, Jebari S, Andersen DB, Joshi HJ, Ju T, Goetze JP, Martin C, Rosenkilde MM, Holst JJ, Kuhre RE, Goth CK, Vakhrushev SY, and Schjoldager KT
- Subjects
- Aged, Animals, Cell Line, Drug Design, Female, Glycosylation, HEK293 Cells, Humans, Male, Middle Aged, Neuropeptides metabolism, Protein Binding, Protein Conformation, alpha-Helical, Protein Stability, Rats, Swine, Peptide Hormones chemistry, Peptide Hormones metabolism, Polysaccharides chemistry, Polysaccharides metabolism
- Abstract
Peptide hormones and neuropeptides encompass a large class of bioactive peptides that regulate physiological processes like anxiety, blood glucose, appetite, inflammation and blood pressure. Here, we execute a focused discovery strategy to provide an extensive map of O-glycans on peptide hormones. We find that almost one third of the 279 classified peptide hormones carry O-glycans. Many of the identified O-glycosites are conserved and are predicted to serve roles in proprotein processing, receptor interaction, biodistribution and biostability. We demonstrate that O-glycans positioned within the receptor binding motifs of members of the neuropeptide Y and glucagon families modulate receptor activation properties and substantially extend peptide half-lives. Our study highlights the importance of O-glycosylation in the biology of peptide hormones, and our map of O-glycosites in this large class of biomolecules serves as a discovery platform for an important class of molecules with potential opportunities for drug designs.
- Published
- 2020
- Full Text
- View/download PDF
37. O-glycan initiation directs distinct biological pathways and controls epithelial differentiation.
- Author
-
Bagdonaite I, Pallesen EM, Ye Z, Vakhrushev SY, Marinova IN, Nielsen MI, Kramer SH, Pedersen SF, Joshi HJ, Bennett EP, Dabelsteen S, and Wandall HH
- Subjects
- Cell Differentiation, Epithelium metabolism, Glycosylation, Humans, Polysaccharides, Protein Processing, Post-Translational, N-Acetylgalactosaminyltransferases genetics, N-Acetylgalactosaminyltransferases metabolism
- Abstract
Post-translational modifications (PTMs) greatly expand the function and potential for regulation of protein activity, and O-glycosylation is among the most abundant and diverse PTMs. Initiation of O-GalNAc glycosylation is regulated by 20 distinct GalNAc-transferases (GalNAc-Ts), and deficiencies in individual GalNAc-Ts are associated with human disease, causing subtle but distinct phenotypes in model organisms. Here, we generate a set of isogenic keratinocyte cell lines lacking either of the three dominant and differentially expressed GalNAc-Ts. Through the ability of keratinocytes to form epithelia, we investigate the phenotypic consequences of the loss of individual GalNAc-Ts. Moreover, we probe the cellular responses through global transcriptomic, differential glycoproteomic, and differential phosphoproteomic analyses. We demonstrate that loss of individual GalNAc-T isoforms causes distinct epithelial phenotypes through their effect on specific biological pathways; GalNAc-T1 targets are associated with components of the endomembrane system, GalNAc-T2 targets with cell-ECM adhesion, and GalNAc-T3 targets with epithelial differentiation. Thus, GalNAc-T isoforms serve specific roles during human epithelial tissue formation., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
38. Correction: Discovery of O -glycans on atrial natriuretic peptide (ANP) that affect both its proteolytic degradation and potency at its cognate receptor.
- Author
-
Hansen LH, Madsen TD, Goth CK, Clausen H, Chen Y, Dzoyashvili N, Iyer SR, Sangaralingham SJ, Burnett JC Jr, Rehfeld JF, Vakhrushev SY, Schjoldager KT, and Goetze JP
- Published
- 2019
- Full Text
- View/download PDF
39. Glyco-DIA: a method for quantitative O-glycoproteomics with in silico-boosted glycopeptide libraries.
- Author
-
Ye Z, Mao Y, Clausen H, and Vakhrushev SY
- Subjects
- Glycosylation, Humans, Computational Biology methods, Computer Simulation, Glycopeptides analysis, Glycoproteins analysis, Polysaccharides analysis, Proteome analysis, Proteomics methods
- Abstract
We report a liquid chromatography coupled to tandem mass spectrometry O-glycoproteomics strategy using data-independent acquisition (DIA) mode for direct analysis of O-glycoproteins. This approach enables characterization of glycopeptides and structures of O-glycans on a proteome-wide scale with quantification of stoichiometries (though it does not allow for direct unambiguous glycosite identification). The method relies on a spectral library of O-glycopeptides; the Glyco-DIA library contains sublibraries obtained from human cell lines and human serum, and it currently covers 2,076 O-glycoproteins (11,452 unique glycopeptide sequences) and the 5 most common core1 O-glycan structures. Applying the Glyco-DIA library to human serum without enrichment for glycopeptides enabled us to identify and quantify 269 distinct glycopeptide sequences bearing up to 5 different core1 O-glycans from 159 glycoproteins in a SingleShot analysis.
- Published
- 2019
- Full Text
- View/download PDF
40. Discovery of O -glycans on atrial natriuretic peptide (ANP) that affect both its proteolytic degradation and potency at its cognate receptor.
- Author
-
Hansen LH, Madsen TD, Goth CK, Clausen H, Chen Y, Dzhoyashvili N, Iyer SR, Sangaralingham SJ, Burnett JC Jr, Rehfeld JF, Vakhrushev SY, Schjoldager KT, and Goetze JP
- Subjects
- Animals, Glycoproteins metabolism, Glycosylation, Humans, Male, Protein Stability, Rats, Sprague-Dawley, Swine, Atrial Natriuretic Factor blood, Polysaccharides metabolism, Proteolysis
- Abstract
Atrial natriuretic peptide (ANP) is a peptide hormone that in response to atrial stretch is secreted from atrial myocytes into the circulation, where it stimulates vasodilatation and natriuresis. ANP is an important biomarker of heart failure where low plasma concentrations exclude cardiac dysfunction. ANP is a member of the natriuretic peptide (NP) family, which also includes the B-type natriuretic peptide (BNP) and the C-type natriuretic peptide. The proforms of these hormones undergo processing to mature peptides, and for proBNP, this process has previously been demonstrated to be regulated by O- glycosylation. It has been suggested that proANP also may undergo post-translational modifications. Here, we conducted a targeted O- glycoproteomics approach to characterize O- glycans on NPs and demonstrate that all NP members can carry O- glycans. We identified four O- glycosites in proANP in the porcine heart, and surprisingly, two of these were located on the mature bioactive ANP itself. We found that one of these glycans is located within a conserved sequence motif of the receptor-binding region, suggesting that O -glycans may serve a function beyond intracellular processing and maturation. We also identified an O -glycoform of proANP naturally occurring in human circulation. We demonstrated that site-specific O -glycosylation shields bioactive ANP from proteolytic degradation and modifies potency at its cognate receptor in vitro Furthermore, we showed that ANP O -glycosylation attenuates acute renal and cardiovascular ANP actions in vivo The discovery of novel glycosylated ANP proteoforms reported here significantly improves our understanding of cardiac endocrinology and provides important insight into the etiology of heart failure., (© 2019 Hansen et al.)
- Published
- 2019
- Full Text
- View/download PDF
41. An Atlas of Human Glycosylation Pathways Enables Display of the Human Glycome by Gene Engineered Cells.
- Author
-
Narimatsu Y, Joshi HJ, Nason R, Van Coillie J, Karlsson R, Sun L, Ye Z, Chen YH, Schjoldager KT, Steentoft C, Furukawa S, Bensing BA, Sullam PM, Thompson AJ, Paulson JC, Büll C, Adema GJ, Mandel U, Hansen L, Bennett EP, Varki A, Vakhrushev SY, Yang Z, and Clausen H
- Subjects
- Epitopes genetics, Epitopes immunology, Glycosylation, Glycosyltransferases genetics, HEK293 Cells, Humans, Oligosaccharides genetics, Polysaccharides classification, Polysaccharides genetics, Polysaccharides immunology, Proteins immunology, Genetic Engineering, Metabolic Networks and Pathways genetics, Polysaccharides chemistry, Proteins genetics
- Abstract
The structural diversity of glycans on cells-the glycome-is vast and complex to decipher. Glycan arrays display oligosaccharides and are used to report glycan hapten binding epitopes. Glycan arrays are limited resources and present saccharides without the context of other glycans and glycoconjugates. We used maps of glycosylation pathways to generate a library of isogenic HEK293 cells with combinatorially engineered glycosylation capacities designed to display and dissect the genetic, biosynthetic, and structural basis for glycan binding in a natural context. The cell-based glycan array is self-renewable and reports glycosyltransferase genes required (or blocking) for interactions through logical sequential biosynthetic steps, which is predictive of structural glycan features involved and provides instructions for synthesis, recombinant production, and genetic dissection strategies. Broad utility of the cell-based glycan array is demonstrated, and we uncover higher order binding of microbial adhesins to clustered patches of O-glycans organized by their presentation on proteins., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
42. Exploring Regulation of Protein O-Glycosylation in Isogenic Human HEK293 Cells by Differential O-Glycoproteomics.
- Author
-
Narimatsu Y, Joshi HJ, Schjoldager KT, Hintze J, Halim A, Steentoft C, Nason R, Mandel U, Bennett EP, Clausen H, and Vakhrushev SY
- Subjects
- Glycopeptides metabolism, Glycosylation, HEK293 Cells, Hep G2 Cells, Humans, Proteome metabolism, Glycomics, Proteomics
- Abstract
Most proteins trafficking the secretory pathway of metazoan cells will acquire GalNAc-type O-glycosylation. GalNAc-type O-glycosylation is differentially regulated in cells by the expression of a repertoire of up to twenty genes encoding polypeptide GalNAc-transferase isoforms (GalNAc-Ts) that initiate O-glycosylation. These GalNAc-Ts orchestrate the positions and patterns of O-glycans on proteins in coordinated, but poorly understood ways - guided partly by the kinetic properties and substrate specificities of their catalytic domains, as well as by modulatory effects of their unique GalNAc-binding lectin domains. Here, we provide the hereto most comprehensive characterization of nonredundant contributions of individual GalNAc-T isoforms to the O-glycoproteome of the human HEK293 cell using quantitative differential O-glycoproteomics on a panel of isogenic HEK293 cells with knockout of GalNAc-T genes ( GALNT1 , T2 , T3 , T7 , T10 , or T11 ). We confirm that a major part of the O-glycoproteome is covered by redundancy, whereas distinct O-glycosite subsets are covered by nonredundant GalNAc-T isoform-specific functions. We demonstrate that the GalNAc-T7 and T10 isoforms function in follow-up of high-density O-glycosylated regions, and that GalNAc-T11 has highly restricted functions and essentially only serves the low-density lipoprotein-related receptors in linker regions (C
6 XXXTC1 ) between the ligand-binding repeats., (© 2019 Narimatsu et al.)- Published
- 2019
- Full Text
- View/download PDF
43. Different isolation approaches lead to diverse glycosylated extracellular vesicle populations.
- Author
-
Freitas D, Balmaña M, Poças J, Campos D, Osório H, Konstantinidi A, Vakhrushev SY, Magalhães A, and Reis CA
- Abstract
Extracellular vesicles (EVs) are a heterogeneous group of small secreted particles involved in intercellular communication and mediating a broad spectrum of biological functions. EVs cargo is composed of a large repertoire of molecules, including glycoconjugates. Herein, we report the first study on the impact of the isolation strategy on the EV populations' glycosylation profile. The use of different state-of-the-art protocols, namely differential ultracentrifugation (UC), total exosome isolation (TEI), OptiPrep
TM density gradient (ODG) and size exclusion chromatography (SEC) resulted in EV populations displaying different sets of glycoconjugates. The EV populations obtained by UC, ODG and SEC methods displayed similar protein and glycan profiles, whereas TEI methodology isolated the most distinct EV population. In addition, ODG and SEC isolation protocols provided an enhanced EV glycoproteins detection. Remarkably, proteins displaying the tumour-associated glycan sialyl-Tn (STn) were identified as packaged cargo into EVs independently of the isolation methodology. STn carrying EV samples isolated by UC, ODG and SEC presented a considerable set of cancer-related proteins that were not detected in EVs isolated by TEI. Our work demonstrates the impact of using different isolation methodologies in the populations of EVs that are obtained, with consequences in the glycosylation profile of the isolated population. Furthermore, our results highlight the importance of selecting adequate EV isolation protocols and cell culture conditions to determine the structural and functional complexity of the EV glycoconjugates.- Published
- 2019
- Full Text
- View/download PDF
44. Site-specific O -glycosylation of members of the low-density lipoprotein receptor superfamily enhances ligand interactions.
- Author
-
Wang S, Mao Y, Narimatsu Y, Ye Z, Tian W, Goth CK, Lira-Navarrete E, Pedersen NB, Benito-Vicente A, Martin C, Uribe KB, Hurtado-Guerrero R, Christoffersen C, Seidah NG, Nielsen R, Christensen EI, Hansen L, Bennett EP, Vakhrushev SY, Schjoldager KT, and Clausen H
- Published
- 2019
- Full Text
- View/download PDF
45. The glycosylation design space for recombinant lysosomal replacement enzymes produced in CHO cells.
- Author
-
Tian W, Ye Z, Wang S, Schulz MA, Van Coillie J, Sun L, Chen YH, Narimatsu Y, Hansen L, Kristensen C, Mandel U, Bennett EP, Jabbarzadeh-Tabrizi S, Schiffmann R, Shen JS, Vakhrushev SY, Clausen H, and Yang Z
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Disease Models, Animal, Fabry Disease drug therapy, Fabry Disease enzymology, Fabry Disease metabolism, Glycosylation, Male, Mice, Mice, Knockout, Recombinant Proteins therapeutic use, alpha-Galactosidase therapeutic use, Lysosomes enzymology
- Abstract
Lysosomal replacement enzymes are essential therapeutic options for rare congenital lysosomal enzyme deficiencies, but enzymes in clinical use are only partially effective due to short circulatory half-life and inefficient biodistribution. Replacement enzymes are primarily taken up by cell surface glycan receptors, and glycan structures influence uptake, biodistribution, and circulation time. It has not been possible to design and systematically study effects of different glycan features. Here we present a comprehensive gene engineering screen in Chinese hamster ovary cells that enables production of lysosomal enzymes with N-glycans custom designed to affect key glycan features guiding cellular uptake and circulation. We demonstrate distinct circulation time and organ distribution of selected glycoforms of α-galactosidase A in a Fabry disease mouse model, and find that an α2-3 sialylated glycoform designed to eliminate uptake by the mannose 6-phosphate and mannose receptors exhibits improved circulation time and targeting to hard-to-reach organs such as heart. The developed design matrix and engineered CHO cell lines enables systematic studies towards improving enzyme replacement therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
46. A strategy for generating cancer-specific monoclonal antibodies to aberrant O-glycoproteins: identification of a novel dysadherin-Tn antibody.
- Author
-
Steentoft C, Fuhrmann M, Battisti F, Van Coillie J, Madsen TD, Campos D, Halim A, Vakhrushev SY, Joshi HJ, Schreiber H, Mandel U, and Narimatsu Y
- Subjects
- Animals, Antibodies, Monoclonal immunology, Cell Line, Tumor, Epitopes immunology, Epitopes metabolism, Glycoproteins immunology, Humans, Mice, Neoplasms immunology, Neoplasms pathology, Antibodies, Monoclonal analysis, Antibodies, Monoclonal biosynthesis, Glycoproteins metabolism, Neoplasms metabolism
- Abstract
Successful application of potent antibody-based T-cell engaging immunotherapeutic strategies is currently limited mainly to hematological cancers. One major reason is the lack of well-characterized antigens on solid tumors with sufficient cancer specific expression. Aberrantly O-glycosylated proteins contain promising cancer-specific O-glycopeptide epitopes suitable for immunotherapeutic applications, but currently only few examples of such antibody epitopes have been identified. We previously showed that chimeric antigen receptor T-cells directed towards aberrantly O-glycosylated MUC1 can control malignant growth in a mouse model. Here, we present a discovery platform for the generation of cancer-specific monoclonal antibodies targeting aberrant O-glycoproteins. The strategy is based on cancer cell lines engineered to homogeneously express the truncated Tn O-glycoform, the so-called SimpleCells. We used SimpleCells of different cancer origin to elicit monoclonal antibodies with selectivity for aberrant O-glycoproteins. For validation we selected and characterized one monoclonal antibody (6C5) directed to a Tn-glycopeptide in dysadherin (FXYD5), known to be upregulated in cancer and promote metastasis. While dysadherin is widely expressed also in normal cells, we demonstrated that the 6C5 epitope is specifically expressed in cancer., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
47. A conserved major facilitator superfamily member orchestrates a subset of O-glycosylation to aid macrophage tissue invasion.
- Author
-
Valoskova K, Biebl J, Roblek M, Emtenani S, Gyoergy A, Misova M, Ratheesh A, Reis-Rodrigues P, Shkarina K, Larsen ISB, Vakhrushev SY, Clausen H, and Siekhaus DE
- Subjects
- Animals, Drosophila melanogaster, Gene Expression Regulation, Glycosylation, Antigens, Tumor-Associated, Carbohydrate metabolism, Cell Movement, Macrophages immunology, Protein Processing, Post-Translational
- Abstract
Aberrant display of the truncated core1 O-glycan T-antigen is a common feature of human cancer cells that correlates with metastasis. Here we show that T-antigen in Drosophila melanogaster macrophages is involved in their developmentally programmed tissue invasion. Higher macrophage T-antigen levels require an atypical major facilitator superfamily (MFS) member that we named Minerva which enables macrophage dissemination and invasion. We characterize for the first time the T and Tn glycoform O-glycoproteome of the Drosophila melanogaster embryo, and determine that Minerva increases the presence of T-antigen on proteins in pathways previously linked to cancer, most strongly on the sulfhydryl oxidase Qsox1 which we show is required for macrophage tissue entry. Minerva's vertebrate ortholog, MFSD1, rescues the minerva mutant's migration and T-antigen glycosylation defects. We thus identify a key conserved regulator that orchestrates O-glycosylation on a protein subset to activate a program governing migration steps important for both development and cancer metastasis., Competing Interests: KV, JB, MR, SE, AG, MM, AR, PR, KS, IL, SV, HC, DS No competing interests declared, (© 2019, Valoskova et al.)
- Published
- 2019
- Full Text
- View/download PDF
48. Probing the contribution of individual polypeptide GalNAc-transferase isoforms to the O -glycoproteome by inducible expression in isogenic cell lines.
- Author
-
Hintze J, Ye Z, Narimatsu Y, Madsen TD, Joshi HJ, Goth CK, Linstedt A, Bachert C, Mandel U, Bennett EP, Vakhrushev SY, and Schjoldager KT
- Subjects
- Amino Acid Sequence, Glycosylation, HEK293 Cells, Humans, Isoenzymes metabolism, Proteomics methods, Polypeptide N-acetylgalactosaminyltransferase, N-Acetylgalactosaminyltransferases metabolism, Proteome metabolism
- Abstract
The GalNAc-type O -glycoproteome is orchestrated by a large family of polypeptide GalNAc-transferase isoenzymes (GalNAc-Ts) with partially overlapping contributions to the O -glycoproteome besides distinct nonredundant functions. Increasing evidence indicates that individual GalNAc-Ts co-regulate and fine-tune specific protein functions in health and disease, and deficiencies in individual GALNT genes underlie congenital diseases with distinct phenotypes. Studies of GalNAc-T specificities have mainly been performed with in vitro enzyme assays using short peptide substrates, but recently quantitative differential O -glycoproteomics of isogenic cells with and without GALNT genes has enabled a more unbiased exploration of the nonredundant contributions of individual GalNAc-Ts. Both approaches suggest that fairly small subsets of O -glycosites are nonredundantly regulated by specific GalNAc-Ts, but how these isoenzymes orchestrate regulation among competing redundant substrates is unclear. To explore this, here we developed isogenic cell model systems with Tet-On inducible expression of two GalNAc-T genes, GALNT2 and GALNT11 , in a knockout background in HEK293 cells. Using quantitative O -glycoproteomics with tandem-mass-tag (TMT) labeling, we found that isoform-specific glycosites are glycosylated in a dose-dependent manner and that induction of GalNAc-T2 or -T11 produces discrete glycosylation effects without affecting the major part of the O -glycoproteome. These results support previous findings indicating that individual GalNAc-T isoenzymes can serve in fine-tuned regulation of distinct protein functions., (© 2018 Hintze et al.)
- Published
- 2018
- Full Text
- View/download PDF
49. Current Technologies for Complex Glycoproteomics and Their Applications to Biology/Disease-Driven Glycoproteomics.
- Author
-
Narimatsu H, Kaji H, Vakhrushev SY, Clausen H, Zhang H, Noro E, Togayachi A, Nagai-Okatani C, Kuno A, Zou X, Cheng L, Tao SC, and Sun Y
- Subjects
- Cell Line, Glycomics methods, Glycosylation, Humans, Polysaccharides analysis, Proteomics methods, Tandem Mass Spectrometry methods, Glycoproteins analysis, Proteomics trends
- Abstract
Glycoproteomics is an important recent advance in the field of glycoscience. In glycomics, glycan structures are comprehensively analyzed after glycans are released from glycoproteins. However, a major limitation of glycomics is the lack of insight into glycoprotein functions. The Biology/Disease-driven Human Proteome Project has a particular focus on biological and medical applications. Glycoproteomics technologies aimed at obtaining a comprehensive understanding of intact glycoproteins, i.e., the kind of glycan structures that are attached to particular amino acids and proteins, have been developed. This Review focuses on the recent progress of the technologies and their applications. First, the methods for large-scale identification of both N- and O-glycosylated proteins are summarized. Next, the progress of analytical methods for intact glycopeptides is outlined. MS/MS-based methods were developed for improving the sensitivity and speed of the mass spectrometer, in parallel with the software for complex spectrum assignment. In addition, a unique approach to identify intact glycopeptides using MS1-based accurate masses is introduced. Finally, as an advance of glycomics, two approaches to provide the spatial distribution of glycans in cells are described, i.e., MS imaging and lectin microarray. These methods allow rapid glycomic profiling of different types of biological samples and thus facilitate glycoproteomics.
- Published
- 2018
- Full Text
- View/download PDF
50. Viral glycoproteomes: technologies for characterization and outlook for vaccine design.
- Author
-
Bagdonaite I, Vakhrushev SY, Joshi HJ, and Wandall HH
- Subjects
- Humans, Mass Spectrometry methods, Viral Vaccines immunology, Virulence immunology, Virus Diseases immunology, Virus Diseases virology, Viruses immunology, Viruses metabolism, Viruses pathogenicity, Glycoproteins metabolism, Proteome metabolism, Proteomics methods, Viral Proteins metabolism
- Abstract
It has long been known that surface proteins of most enveloped viruses are covered with glycans. It has furthermore been demonstrated that glycosylation is essential for propagation and immune evasion for many viruses. The recent development of high-resolution mass spectrometry techniques has enabled identification not only of the precise structures but also the positions of such post-translational modifications on viruses, revealing substantial differences in extent of glycosylation and glycan maturation for different classes of viruses. In-depth characterization of glycosylation and other post-translational modifications of viral envelope glycoproteins is essential for rational design of vaccines and antivirals. In this Review, we provide an overview of techniques used to address viral glycosylation and summarize information on glycosylation of enveloped viruses representing ongoing public health challenges. Furthermore, we discuss how knowledge on glycosylation can be translated to means to prevent and combat viral infections., (© 2018 Federation of European Biochemical Societies.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.