251
|
Aretz J, Wratil PR, Wamhoff EC, Nguyen HG, Reutter W, Rademacher C. Fragment screening of N-acetylmannosamine kinase reveals noncarbohydrate inhibitors. CAN J CHEM 2016. [DOI: 10.1139/cjc-2015-0603] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Many biological processes from infection to tumor immune evasion are controlled by cell surface sialylation. To gather further insight into these processes, methods to alter cell surface sialylation are required. One way to achieve this is inhibiting the key enzyme of sialic acid de novo biosynthesis, the intracellular bifunctional UDP-N-acetylglucosamine epimerase/N-acetylmannosamine kinase (GNE/MNK). Here, we present low molecular weight inhibitors of MNK activity based on picolinic acid derivatives. They were identified in a fragment screening using 19F NMR and validated in a biochemical inhibition assay followed by a structure–activity relationship analysis and docking. The optimized compound 6-carbamoylpicolinic acid inhibits MNK with a double-digit micromolar affinity. Its low molecular weight (166 Da) renders this picolinic acid derivative an exquisite starting point for the development of high-affinity MNK inhibitors, which may serve as molecular probes or lead candidates in future.
Collapse
Affiliation(s)
- Jonas Aretz
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14424, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Paul Robin Wratil
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité — Universitätsmedizin Berlin, Germany
| | - Eike-Christian Wamhoff
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14424, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Hoang Giang Nguyen
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité — Universitätsmedizin Berlin, Germany
| | - Werner Reutter
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité — Universitätsmedizin Berlin, Germany
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14424, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
252
|
Allen JG, Mujacic M, Frohn MJ, Pickrell AJ, Kodama P, Bagal D, San Miguel T, Sickmier EA, Osgood S, Swietlow A, Li V, Jordan JB, Kim KW, Rousseau AMC, Kim YJ, Caille S, Achmatowicz M, Thiel O, Fotsch CH, Reddy P, McCarter JD. Facile Modulation of Antibody Fucosylation with Small Molecule Fucostatin Inhibitors and Cocrystal Structure with GDP-Mannose 4,6-Dehydratase. ACS Chem Biol 2016; 11:2734-2743. [PMID: 27434622 DOI: 10.1021/acschembio.6b00460] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The efficacy of therapeutic antibodies that induce antibody-dependent cellular cytotoxicity can be improved by reduced fucosylation. Consequently, fucosylation is a critical product attribute of monoclonal antibodies produced as protein therapeutics. Small molecule fucosylation inhibitors have also shown promise as potential therapeutics in animal models of tumors, arthritis, and sickle cell disease. Potent small molecule metabolic inhibitors of cellular protein fucosylation, 6,6,6-trifluorofucose per-O-acetate and 6,6,6-trifluorofucose (fucostatin I), were identified that reduces the fucosylation of recombinantly expressed antibodies in cell culture in a concentration-dependent fashion enabling the controlled modulation of protein fucosylation levels. 6,6,6-Trifluorofucose binds at an allosteric site of GDP-mannose 4,6-dehydratase (GMD) as revealed for the first time by the X-ray cocrystal structure of a bound allosteric GMD inhibitor. 6,6,6-Trifluorofucose was found to be incorporated in place of fucose at low levels (<1%) in the glycans of recombinantly expressed antibodies. A fucose-1-phosphonate analog, fucostatin II, was designed that inhibits fucosylation with no incorporation into antibody glycans, allowing the production of afucosylated antibodies in which the incorporation of non-native sugar is completely absent-a key advantage in the production of therapeutic antibodies, especially biosimilar antibodies. Inhibitor structure-activity relationships, identification of cellular and inhibitor metabolites in inhibitor-treated cells, fucose competition studies, and the production of recombinant antibodies with varying levels of fucosylation are described.
Collapse
Affiliation(s)
- John G. Allen
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Mirna Mujacic
- Process Development − Drug Substance
Technologies, Amgen Inc., 1201 Amgen Court W., Seattle, Washington 98119, United States
| | - Michael J. Frohn
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Alex J. Pickrell
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Paul Kodama
- Process Development − Drug Substance
Technologies, Amgen Inc., 1201 Amgen Court W., Seattle, Washington 98119, United States
| | - Dhanashri Bagal
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Tisha San Miguel
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - E. Allen Sickmier
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Steve Osgood
- Process Development − Attribute
Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Aleksander Swietlow
- Process Development − Attribute
Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Vivian Li
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - John B. Jordan
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Ki-Won Kim
- Cardiometabolic
Disorders, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Anne-Marie C. Rousseau
- Therapeutic
Innovations Unit, Amgen Inc., 1201 Amgen Court W., Seattle, Washington 98119, United States
| | - Yong-Jae Kim
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Seb Caille
- Process Development
− Drug Substance Technologies, Amgen Inc., One Amgen Center
Drive, Thousand Oaks, California 91320, United States
| | - Mike Achmatowicz
- Process Development
− Drug Substance Technologies, Amgen Inc., One Amgen Center
Drive, Thousand Oaks, California 91320, United States
| | - Oliver Thiel
- Process Development
− Drug Substance Technologies, Amgen Inc., One Amgen Center
Drive, Thousand Oaks, California 91320, United States
| | - Christopher H. Fotsch
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Pranhitha Reddy
- Process Development − Drug Substance
Technologies, Amgen Inc., 1201 Amgen Court W., Seattle, Washington 98119, United States
| | - John D. McCarter
- Therapeutic Discovery, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| |
Collapse
|
253
|
Exploring human glycosylation for better therapies. Mol Aspects Med 2016; 51:125-43. [DOI: 10.1016/j.mam.2016.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/28/2016] [Accepted: 05/06/2016] [Indexed: 01/19/2023]
|
254
|
Kanabar V, Tedaldi L, Jiang J, Nie X, Panina I, Descroix K, Man F, Pitchford SC, Page CP, Wagner GK. Base-modified UDP-sugars reduce cell surface levels of P-selectin glycoprotein 1 (PSGL-1) on IL-1β-stimulated human monocytes. Glycobiology 2016; 26:1059-1071. [PMID: 27233805 PMCID: PMC5072147 DOI: 10.1093/glycob/cww053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 04/07/2016] [Accepted: 04/22/2016] [Indexed: 12/26/2022] Open
Abstract
P-selectin glycoprotein ligand-1 (PSGL-1, CD162) is a cell-surface glycoprotein that is expressed, either constitutively or inducibly, on all myeloid and lymphoid cell lineages. PSGL-1 is implicated in cell-cell interactions between platelets, leukocytes and endothelial cells, and a key mediator of inflammatory cell recruitment and transmigration into tissues. Here, we have investigated the effects of the β-1,4-galactosyltransferase inhibitor 5-(5-formylthien-2-yl) UDP-Gal (5-FT UDP-Gal, compound 1: ) and two close derivatives on the cell surface levels of PSGL-1 on human peripheral blood mononuclear cells (hPBMCs). PSGL-1 levels were studied both under basal conditions, and upon stimulation of hPBMCs with interleukin-1β (IL-1β). Between 1 and 24 hours after IL-1β stimulation, we observed initial PSGL-1 shedding, followed by an increase in PSGL-1 levels on the cell surface, with a maximal window between IL-1β-induced and basal levels after 72 h. All three inhibitors reduce PSGL-1 levels on IL-1β-stimulated cells in a concentration-dependent manner, but show no such effect in resting cells. Compound 1: also affects the cell surface levels of adhesion molecule CD11b in IL-1β-stimulated hPBMCs, but not of glycoproteins CD14 and CCR2. This activity profile may be linked to the inhibition of global Sialyl Lewis presentation on hPBMCs by compound 1: , which we have also observed. Although this mechanistic explanation remains hypothetical at present, our results show, for the first time, that small molecules can discriminate between IL-1β-induced and basal levels of cell surface PSGL-1. These findings open new avenues for intervention with PSGL-1 presentation on the cell surface of primed hPBMCs and may have implications for anti-inflammatory drug development.
Collapse
Affiliation(s)
- Varsha Kanabar
- Sackler Institute of Pulmonary Pharmacology
- Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building, London SE1 9NH, UK
| | - Lauren Tedaldi
- Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building, London SE1 9NH, UK
- Department of Chemistry, Faculty of Natural & Mathematical Sciences, King's College London, Britannia House, 7 Trinity Street, London, SE1 1DB, UK
| | - Jingqian Jiang
- Department of Chemistry, Faculty of Natural & Mathematical Sciences, King's College London, Britannia House, 7 Trinity Street, London, SE1 1DB, UK
| | - Xiaodan Nie
- Sackler Institute of Pulmonary Pharmacology
- Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building, London SE1 9NH, UK
| | - Irina Panina
- Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building, London SE1 9NH, UK
| | - Karine Descroix
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Francis Man
- Sackler Institute of Pulmonary Pharmacology
- Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building, London SE1 9NH, UK
| | - Simon C Pitchford
- Sackler Institute of Pulmonary Pharmacology
- Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building, London SE1 9NH, UK
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology
- Institute of Pharmaceutical Science, King's College London, Franklin Wilkins Building, London SE1 9NH, UK
| | - Gerd K Wagner
- Department of Chemistry, Faculty of Natural & Mathematical Sciences, King's College London, Britannia House, 7 Trinity Street, London, SE1 1DB, UK
| |
Collapse
|
255
|
Ho WL, Hsu WM, Huang MC, Kadomatsu K, Nakagawara A. Protein glycosylation in cancers and its potential therapeutic applications in neuroblastoma. J Hematol Oncol 2016; 9:100. [PMID: 27686492 PMCID: PMC5041531 DOI: 10.1186/s13045-016-0334-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023] Open
Abstract
Glycosylation is the most complex post-translational modification of proteins. Altered glycans on the tumor- and host-cell surface and in the tumor microenvironment have been identified to mediate critical events in cancer pathogenesis and progression. Tumor-associated glycan changes comprise increased branching of N-glycans, higher density of O-glycans, generation of truncated versions of normal counterparts, and generation of unusual forms of terminal structures arising from sialylation and fucosylation. The functional role of tumor-associated glycans (Tn, sTn, T, and sLea/x) is dependent on the interaction with lectins. Lectins are expressed on the surface of immune cells and endothelial cells or exist as extracellular matrix proteins and soluble adhesion molecules. Expression of tumor-associated glycans is involved in the dysregulation of glycogenes, which mainly comprise glycosyltransferases and glycosidases. Furthermore, genetic and epigenetic mechanisms on many glycogenes are associated with malignant transformation. With better understanding of all aspects of cancer-cell glycomics, many tumor-associated glycans have been utilized for diagnostic, prognostic, and therapeutic purposes. Glycan-based therapeutics has been applied to cancers from breast, lung, gastrointestinal system, melanomas, and lymphomas but rarely to neuroblastomas (NBs). The success of anti-disialoganglioside (GD2, a glycolipid antigen) antibodies sheds light on glycan-based therapies for NB and also suggests the possibility of protein glycosylation-based therapies for NB. This review summarizes our understanding of cancer glycobiology with a focus of how protein glycosylation and associated glycosyltransferases affect cellular behaviors and treatment outcome of various cancers, especially NB. Finally, we highlight potential applications of glycosylation in drug and cancer vaccine development for NB.
Collapse
Affiliation(s)
- Wan-Ling Ho
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan.,Department of Pediatrics, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 100, Taiwan. .,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| | - Min-Chuan Huang
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan. .,Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei, 10051, Taiwan.
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | |
Collapse
|
256
|
Szabo R, Skropeta D. Advancement of Sialyltransferase Inhibitors: Therapeutic Challenges and Opportunities. Med Res Rev 2016; 37:219-270. [DOI: 10.1002/med.21407] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/14/2016] [Accepted: 08/03/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Rémi Szabo
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
| | - Danielle Skropeta
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
- Centre for Medical & Molecular Bioscience; University of Wollongong; Wollongong NSW 2522 Australia
| |
Collapse
|
257
|
Sminia TJ, Zuilhof H, Wennekes T. Getting a grip on glycans: A current overview of the metabolic oligosaccharide engineering toolbox. Carbohydr Res 2016; 435:121-141. [PMID: 27750120 DOI: 10.1016/j.carres.2016.09.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 12/16/2022]
Abstract
This review discusses the advances in metabolic oligosaccharide engineering (MOE) from 2010 to 2016 with a focus on the structure, preparation, and reactivity of its chemical probes. A brief historical overview of MOE is followed by a comprehensive overview of the chemical probes currently available in the MOE molecular toolbox and the bioconjugation techniques they enable. The final part of the review focusses on the synthesis of a selection of probes and finishes with an outlook on recent and potential upcoming advances in the field of MOE.
Collapse
Affiliation(s)
- Tjerk J Sminia
- Laboratory of Organic Chemistry, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Han Zuilhof
- Laboratory of Organic Chemistry, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Tom Wennekes
- Laboratory of Organic Chemistry, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands; Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
258
|
Li C, Wang LX. Endoglycosidases for the Synthesis of Polysaccharides and Glycoconjugates. Adv Carbohydr Chem Biochem 2016; 73:73-116. [PMID: 27816108 DOI: 10.1016/bs.accb.2016.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent advances in glycobiology have implicated essential roles of oligosaccharides and glycoconjugates in many important biological recognition processes, including intracellular signaling, cell adhesion, cell differentiation, cancer progression, host-pathogen interactions, and immune responses. A detailed understanding of the biological functions, as well as the development of carbohydrate-based therapeutics, often requires structurally well-defined oligosaccharides and glycoconjugates, which are usually difficult to isolate in pure form from natural sources. To meet with this urgent need, chemical and chemoenzymatic synthesis has become increasingly important as the major means to provide homogeneous compounds for functional glycocomics studies and for drug/vaccine development. Chemoenzymatic synthesis, an approach that combines chemical synthesis and enzymatic manipulations, is often the method of choice for constructing complex oligosaccharides and glycoconjugates that are otherwise difficult to achieve by purely chemical synthesis. Among these, endoglycosidases, a class of glycosidases that hydrolyze internal glycosidic bonds in glycoconjugates and polysaccharides, are emerging as a very attractive class of enzymes for synthetic purposes, due to their transglycosylation activity and their capability of transferring oligosaccharide units en bloc in a single step, in contrast to the limitation of monosaccharide transfers by common glycosyltransferases. In this chapter, we provide an overview on the application of endoglycosidases for the synthesis of complex carbohydrates, including oligosaccharides, polysaccharides, glycoproteins, glycolipids, proteoglycans, and other biologically relevant polysaccharides. The scope, limitation, and future directions of endoglycosidase-catalyzed synthesis are discussed.
Collapse
Affiliation(s)
- Chao Li
- University of Maryland, College Park, MD, United States
| | - Lai-Xi Wang
- University of Maryland, College Park, MD, United States
| |
Collapse
|
259
|
Subedi GP, Barb AW. The immunoglobulin G1 N-glycan composition affects binding to each low affinity Fc γ receptor. MAbs 2016; 8:1512-1524. [PMID: 27492264 DOI: 10.1080/19420862.2016.1218586] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Immunoglobulin G1 (IgG1) is the most abundant circulating human antibody and also the scaffold for many therapeutic monoclonal antibodies (mAbs). The destruction of IgG-coated targets by cell-mediated pathways begins with an interaction between the IgG Fc region and multiple varieties of membrane-bound Fc γ receptors (FcγRs) on the surface of leukocytes. This interaction requires the presence of an asparagine-linked (N-)glycan on the Fc, and variations in the N-glycan composition can affect the affinity of CD16A binding (an FcγR). Contemporary efforts to glycoengineer mAbs focus on increasing CD16A affinity, and thus treatment efficacy, but it is unclear how these changes affect affinity for the other FcγRs. Here, we measure binding of the extracellular Fc-binding domains for human CD16A and B, CD32A, B and C, and CD64 to 6 well-defined IgG1 Fc glycoforms that cover ∼85% of the pool of human IgG1 Fc glycoforms. Core α1-6 fucosylation showed the greatest changes with CD16B (8.5-fold decrease), CD16A (3.9-fold decrease) and CD32B/C (1.8-fold decrease), but did not affect binding to CD32A. Adding galactose to the non-reducing termini of the complex-type, biantennary glycan increased affinity for all CD16s and 32s tested by 1.7-fold. Sialylation did not change the affinity of core-fucosylated Fc, but increased the affinity of afucosylated Fc slightly by an average of 1.16-fold for all CD16s and CD32s tested. The effects of fucose and galactose modification are additive, suggesting the contributions of these residues to Fc γ receptor affinity are independent.
Collapse
Affiliation(s)
- Ganesh P Subedi
- a Roy J. Carver Department of Biochemistry , Biophysics and Molecular Biology, Iowa State University , Ames , IA , USA
| | - Adam W Barb
- a Roy J. Carver Department of Biochemistry , Biophysics and Molecular Biology, Iowa State University , Ames , IA , USA
| |
Collapse
|
260
|
Wratil PR, Horstkorte R, Reutter W. Metabolic Glycoengineering with N-Acyl Side Chain Modified Mannosamines. Angew Chem Int Ed Engl 2016; 55:9482-512. [PMID: 27435524 DOI: 10.1002/anie.201601123] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Indexed: 12/14/2022]
Abstract
In metabolic glycoengineering (MGE), cells or animals are treated with unnatural derivatives of monosaccharides. After entering the cytosol, these sugar analogues are metabolized and subsequently expressed on newly synthesized glycoconjugates. The feasibility of MGE was first discovered for sialylated glycans, by using N-acyl-modified mannosamines as precursor molecules for unnatural sialic acids. Prerequisite is the promiscuity of the enzymes of the Roseman-Warren biosynthetic pathway. These enzymes were shown to tolerate specific modifications of the N-acyl side chain of mannosamine analogues, for example, elongation by one or more methylene groups (aliphatic modifications) or by insertion of reactive groups (bioorthogonal modifications). Unnatural sialic acids are incorporated into glycoconjugates of cells and organs. MGE has intriguing biological consequences for treated cells (aliphatic MGE) and offers the opportunity to visualize the topography and dynamics of sialylated glycans in vitro, ex vivo, and in vivo (bioorthogonal MGE).
Collapse
Affiliation(s)
- Paul R Wratil
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité-Universitätsmedizin Berlin, Arnimallee 22, 14195, Berlin, Germany.
| | - Rüdiger Horstkorte
- Institut für Physiologische Chemie, Martin-Luther-Universität Halle-Wittenberg, Hollystrasse 1, 06114, Halle, Germany.
| | - Werner Reutter
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité-Universitätsmedizin Berlin, Arnimallee 22, 14195, Berlin, Germany
| |
Collapse
|
261
|
Wratil PR, Horstkorte R, Reutter W. Metabolisches Glykoengineering mitN-Acyl-Seiten- ketten-modifizierten Mannosaminen. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601123] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Paul R. Wratil
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie; Charité - Universitätsmedizin Berlin; Arnimallee 22 14195 Berlin Deutschland
| | - Rüdiger Horstkorte
- Institut für Physiologische Chemie; Martin-Luther-Universität Halle-Wittenberg; Hollystraße 1 06114 Halle Deutschland
| | - Werner Reutter
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie; Charité - Universitätsmedizin Berlin; Arnimallee 22 14195 Berlin Deutschland
| |
Collapse
|
262
|
Krasnova L, Wong CH. Understanding the Chemistry and Biology of Glycosylation with Glycan Synthesis. Annu Rev Biochem 2016; 85:599-630. [DOI: 10.1146/annurev-biochem-060614-034420] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Larissa Krasnova
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037;
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037;
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, 115
| |
Collapse
|
263
|
Nieto-Garcia O, Wratil PR, Nguyen LD, Böhrsch V, Hinderlich S, Reutter W, Hackenberger CPR. Inhibition of the key enzyme of sialic acid biosynthesis by C6-Se modified N-acetylmannosamine analogs. Chem Sci 2016; 7:3928-3933. [PMID: 30155038 PMCID: PMC6013775 DOI: 10.1039/c5sc04082e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/13/2016] [Indexed: 01/11/2023] Open
Abstract
Synthetically accessible C6-analogs of N-acetylmannosamine (ManNAc) were tested as potential inhibitors of the bifunctional UDP-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE/MNK), the key enzyme of sialic acid biosynthesis. Enzymatic experiments revealed that the modification introduced at the C6 saccharide position strongly influences the inhibitory potency. A C6-ManNAc diselenide dimer showed the strongest kinase inhibition in the low μM range among all the substrates tested and successfully reduced cell surface sialylation in Jurkat cells.
Collapse
Affiliation(s)
- Olaia Nieto-Garcia
- Leibniz-Institut für Molekulare Pharmakologie , Robert-Roessle-Strasse 10 , 13125 Berlin , Germany
| | - Paul R Wratil
- Institut für Laboratoriumsmedizin , Klinische Chemie und Pathobiochemie , Charié-Universitätsmedizin Berlin , Arnimalee 22 , 14195 Berlin , Germany .
| | - Long D Nguyen
- Institut für Laboratoriumsmedizin , Klinische Chemie und Pathobiochemie , Charié-Universitätsmedizin Berlin , Arnimalee 22 , 14195 Berlin , Germany .
| | - Verena Böhrsch
- Leibniz-Institut für Molekulare Pharmakologie , Robert-Roessle-Strasse 10 , 13125 Berlin , Germany
| | - Stephan Hinderlich
- Beuth Hochschule für Technik Berlin , Department Life Sciences & Technology , Seestrase 64 , 13347 Berlin , Germany .
| | - Werner Reutter
- Institut für Laboratoriumsmedizin , Klinische Chemie und Pathobiochemie , Charié-Universitätsmedizin Berlin , Arnimalee 22 , 14195 Berlin , Germany .
| | - Christian P R Hackenberger
- Leibniz-Institut für Molekulare Pharmakologie , Robert-Roessle-Strasse 10 , 13125 Berlin , Germany
- Humboldt Universität zu Berlin , Department Chemie , Brook-Taylor-Strasse 2 , 12489 , Berlin , Germany .
| |
Collapse
|
264
|
Lin B, Wu X, Zhao H, Tian Y, Han J, Liu J, Han S. Redirecting immunity via covalently incorporated immunogenic sialic acid on the tumor cell surface. Chem Sci 2016; 7:3737-3741. [PMID: 29997860 PMCID: PMC6008587 DOI: 10.1039/c5sc04133c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 02/23/2016] [Indexed: 12/17/2022] Open
Abstract
Techniques eliciting anti-tumor immunity are of interest for immunotherapy. We herein report the covalent incorporation of a non-self immunogen into the tumor glycocalyx by metabolic oligosaccharide engineering with 2,4-dinitrophenylated sialic acid (DNPSia). This enables marked suppression of pulmonary metastasis and subcutaneous tumor growth of B16F10 melanoma cells in mice preimmunized to produce anti-DNP antibodies. Located on the exterior glycocalyx, DNPSia is well-positioned to recruit antibodies. Given the high levels of natural anti-DNP antibodies in humans and ubiquitous sialylation across many cancers, DNPSia offers a simplified route to redirect immunity against diverse tumors without recourse to preimmunization.
Collapse
Affiliation(s)
- Bijuan Lin
- State Key Laboratory for Physical Chemistry of Solid Surfaces , Department of Chemical Biology , College of Chemistry and Chemical Engineering , The Key Laboratory for Chemical Biology of Fujian Province , The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen , 361005 , China . ; Tel: +86-0592-2181728
| | - Xuanjun Wu
- State Key Laboratory for Physical Chemistry of Solid Surfaces , Department of Chemical Biology , College of Chemistry and Chemical Engineering , The Key Laboratory for Chemical Biology of Fujian Province , The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen , 361005 , China . ; Tel: +86-0592-2181728
| | - Hu Zhao
- State Key Laboratory for Physical Chemistry of Solid Surfaces , Department of Chemical Biology , College of Chemistry and Chemical Engineering , The Key Laboratory for Chemical Biology of Fujian Province , The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen , 361005 , China . ; Tel: +86-0592-2181728
| | - Yunpeng Tian
- State Key Laboratory for Physical Chemistry of Solid Surfaces , Department of Chemical Biology , College of Chemistry and Chemical Engineering , The Key Laboratory for Chemical Biology of Fujian Province , The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen , 361005 , China . ; Tel: +86-0592-2181728
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology , Innovation Center for Cell Signaling Network , School of Life Sciences , Xiamen University , Xiamen , 361005 , China
| | - Jian Liu
- State Key Laboratory for Physical Chemistry of Solid Surfaces , Department of Chemical Biology , College of Chemistry and Chemical Engineering , The Key Laboratory for Chemical Biology of Fujian Province , The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen , 361005 , China . ; Tel: +86-0592-2181728
| | - Shoufa Han
- State Key Laboratory for Physical Chemistry of Solid Surfaces , Department of Chemical Biology , College of Chemistry and Chemical Engineering , The Key Laboratory for Chemical Biology of Fujian Province , The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen , 361005 , China . ; Tel: +86-0592-2181728
| |
Collapse
|
265
|
Custódio CA, Mano JF. Cell surface engineering to control cellular interactions. CHEMNANOMAT : CHEMISTRY OF NANOMATERIALS FOR ENERGY, BIOLOGY AND MORE 2016; 2:376-384. [PMID: 30842920 PMCID: PMC6398572 DOI: 10.1002/cnma.201600047] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Cell surface composition determines all interactions of the cell with is environment, thus cell functions such as adhesion, migration and cell-cell interactions are likely to be controlled by engineering and manipulating cell membrane. Cell membranes present a rich repertoire of molecules, therefore a versatile ground for modification. However the complex and dynamic nature of the cell surface is also a major challenge for cell surface engineering that should also involve strategies compatible with cell viability. Cell surface engineering by selective chemical reactions or by the introduction of exogenous targeting ligands can be powerful tools for engineering novel interactions and control cell function. In addition to chemical conjugation and modification of functional groups, ligands of interest to modify the surface of cells include recombinant proteins, liposomes or nanoparticles. Here, we review recent efforts to perform changes to cell surface composition. We focus on the engineering of the cell surface with biological, chemical or physical methods to modulate cell functions and control cell-cell and cell-microenvironment interactions. Potential applications of cell surface engineering are also stated.
Collapse
Affiliation(s)
- Catarina A Custódio
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence of Tissue Engineering and Regenerative Medicine, Avepark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B's PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - João F Mano
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence of Tissue Engineering and Regenerative Medicine, Avepark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B's PT Government Associated Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
266
|
Natoni A, Macauley MS, O'Dwyer ME. Targeting Selectins and Their Ligands in Cancer. Front Oncol 2016; 6:93. [PMID: 27148485 PMCID: PMC4834419 DOI: 10.3389/fonc.2016.00093] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022] Open
Abstract
Aberrant glycosylation is a hallmark of cancer cells with increased evidence pointing to a role in tumor progression. In particular, aberrant sialylation of glycoproteins and glycolipids has been linked to increased immune cell evasion, drug evasion, drug resistance, tumor invasiveness, and vascular dissemination, leading to metastases. Hypersialylation of cancer cells is largely the result of overexpression of sialyltransferases (STs). Differentially, humans express twenty different STs in a tissue-specific manner, each of which catalyzes the attachment of sialic acids via different glycosidic linkages (α2-3, α2-6, or α2-8) to the underlying glycan chain. One important mechanism whereby overexpression of STs contributes to an enhanced metastatic phenotype is via the generation of selectin ligands. Selectin ligand function requires the expression of sialyl-Lewis X and its structural isomer sialyl-Lewis A, which are synthesized by the combined action of alpha α1-3-fucosyltransferases, α2-3-sialyltransferases, β1-4-galactosyltranferases, and N-acetyl-β-glucosaminyltransferases. The α2-3-sialyltransferases ST3Gal4 and ST3Gal6 are critical to the generation of functional E- and P-selectin ligands and overexpression of these STs have been linked to increased risk of metastatic disease in solid tumors and poor outcome in multiple myeloma. Thus, targeting selectins and their ligands as well as the enzymes involved in their generation, in particular STs, could be beneficial to many cancer patients. Potential strategies include ST inhibition and the use of selectin antagonists, such as glycomimetic drugs and antibodies. Here, we review ongoing efforts to optimize the potency and selectivity of ST inhibitors, including the potential for targeted delivery approaches, as well as evaluate the potential utility of selectin inhibitors, which are now in early clinical development.
Collapse
Affiliation(s)
- Alessandro Natoni
- Biomedical Sciences, National University of Ireland Galway , Galway , Ireland
| | - Matthew S Macauley
- Department of Chemical Physiology, The Scripps Research Institute , La Jolla, CA , USA
| | - Michael E O'Dwyer
- Biomedical Sciences, National University of Ireland Galway, Galway, Ireland; School of Medicine, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
267
|
Gram AM, Oosenbrug T, Lindenbergh MFS, Büll C, Comvalius A, Dickson KJI, Wiegant J, Vrolijk H, Lebbink RJ, Wolterbeek R, Adema GJ, Griffioen M, Heemskerk MHM, Tscharke DC, Hutt-Fletcher LM, Wiertz EJHJ, Hoeben RC, Ressing ME. The Epstein-Barr Virus Glycoprotein gp150 Forms an Immune-Evasive Glycan Shield at the Surface of Infected Cells. PLoS Pathog 2016; 12:e1005550. [PMID: 27077376 PMCID: PMC4831753 DOI: 10.1371/journal.ppat.1005550] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 03/14/2016] [Indexed: 12/15/2022] Open
Abstract
Cell-mediated immunity plays a key role in host control of viral infection. This is exemplified by life-threatening reactivations of e.g. herpesviruses in individuals with impaired T-cell and/or iNKT cell responses. To allow lifelong persistence and virus production in the face of primed immunity, herpesviruses exploit immune evasion strategies. These include a reduction in viral antigen expression during latency and a number of escape mechanisms that target antigen presentation pathways. Given the plethora of foreign antigens expressed in virus-producing cells, herpesviruses are conceivably most vulnerable to elimination by cell-mediated immunity during the replicative phase of infection. Here, we show that a prototypic herpesvirus, Epstein-Barr virus (EBV), encodes a novel, broadly acting immunoevasin, gp150, that is expressed during the late phase of viral replication. In particular, EBV gp150 inhibits antigen presentation by HLA class I, HLA class II, and the non-classical, lipid-presenting CD1d molecules. The mechanism of gp150-mediated T-cell escape does not depend on degradation of the antigen-presenting molecules nor does it require gp150’s cytoplasmic tail. Through its abundant glycosylation, gp150 creates a shield that impedes surface presentation of antigen. This is an unprecedented immune evasion mechanism for herpesviruses. In view of its likely broader target range, gp150 could additionally have an impact beyond escape of T cell activation. Importantly, B cells infected with a gp150-null mutant EBV displayed rescued levels of surface antigen presentation by HLA class I, HLA class II, and CD1d, supporting an important role for iNKT cells next to classical T cells in fighting EBV infection. At the same time, our results indicate that EBV gp150 prolongs the timespan for producing viral offspring at the most vulnerable stage of the viral life cycle. The human herpesvirus Epstein-Barr virus (EBV) is an important human pathogen involved in infectious mononucleosis and several malignant tumors, including lymphomas in the immunosuppressed. Upon primary infection, a balance between virus and host is established, to which EBV’s capacity to dodge T cell-mediated attack contributes. Here we identify the late protein EBV gp150 as a novel immunoevasin, frustrating antigen presentation by HLA class I, class II, and CD1d molecules. EBV gp150’s many sialoglycans create a shield impeding surface detection of presented antigen. Interestingly, exploiting glycan shielding as a mechanism to mask surface exposed proteins on infected cells could permit EBV to additionally modulate other aspects of host antiviral defense. B cells producing wild-type EBV escaped immune recognition more efficiently than those infected with a gp150-null virus, pointing towards a role for gp150 in natural infection. Our results reveal a novel, broadly active strategy by which a herpesvirus glycoprotein, EBV gp150, blocks antigen presentation to T cells through glycan shielding, a new paradigm in herpesvirus immune evasion.
Collapse
Affiliation(s)
- Anna M. Gram
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Timo Oosenbrug
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Christian Büll
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Anouskha Comvalius
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kathryn J. I. Dickson
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Joop Wiegant
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Vrolijk
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ron Wolterbeek
- Department of Medical Statistics & Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gosse J. Adema
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - David C. Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Lindsey M. Hutt-Fletcher
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | | | - Rob C. Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maaike E. Ressing
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
268
|
Nischan N, Kohler JJ. Advances in cell surface glycoengineering reveal biological function. Glycobiology 2016; 26:789-96. [PMID: 27066802 DOI: 10.1093/glycob/cww045] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/04/2016] [Indexed: 12/31/2022] Open
Abstract
Cell surface glycans are critical mediators of cell-cell, cell-ligand, and cell-pathogen interactions. By controlling the set of glycans displayed on the surface of a cell, it is possible to gain insight into the biological functions of glycans. Moreover, control of glycan expression can be used to direct cellular behavior. While genetic approaches to manipulate glycosyltransferase gene expression are available, their utility in glycan engineering has limitations due to the combinatorial nature of glycan biosynthesis and the functional redundancy of glycosyltransferase genes. Biochemical and chemical strategies offer valuable complements to these genetic approaches, notably by enabling introduction of unnatural functionalities, such as fluorophores, into cell surface glycans. Here, we describe some of the most recent developments in glycoengineering of cell surfaces, with an emphasis on strategies that employ novel chemical reagents. We highlight key examples of how these advances in cell surface glycan engineering enable study of cell surface glycans and their function. Exciting new technologies include synthetic lipid-glycans, new chemical reporters for metabolic oligosaccharide engineering to allow tandem and in vivo labeling of glycans, improved chemical and enzymatic methods for glycoproteomics, and metabolic glycosyltransferase inhibitors. Many chemical and biochemical reagents for glycan engineering are commercially available, facilitating their adoption by the biological community.
Collapse
Affiliation(s)
- Nicole Nischan
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jennifer J Kohler
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
269
|
Lee JH, Ozorowski G, Ward AB. Cryo-EM structure of a native, fully glycosylated, cleaved HIV-1 envelope trimer. Science 2016; 351:1043-8. [PMID: 26941313 DOI: 10.1126/science.aad2450] [Citation(s) in RCA: 356] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The envelope glycoprotein trimer (Env) on the surface of HIV-1 recognizes CD4(+) T cells and mediates viral entry. During this process, Env undergoes substantial conformational rearrangements, making it difficult to study in its native state. Soluble stabilized trimers have provided valuable insights into the Env structure, but they lack the hydrophobic membrane proximal external region (MPER, an important target of broadly neutralizing antibodies), the transmembrane domain, and the cytoplasmic tail. Here we present (i) a cryogenic electron microscopy (cryo-EM) structure of a clade B virus Env, which lacks only the cytoplasmic tail and is stabilized by the broadly neutralizing antibody PGT151, at a resolution of 4.2 angstroms and (ii) a reconstruction of this form of Env in complex with PGT151 and MPER-targeting antibody 10E8 at a resolution of 8.8 angstroms. These structures provide new insights into the wild-type Env structure.
Collapse
Affiliation(s)
- Jeong Hyun Lee
- Department of Integrative Structural and Computational Biology, Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, International AIDS Vaccine Initiative Neutralizing Antibody Center, and Collaboration for AIDS Vaccine Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, International AIDS Vaccine Initiative Neutralizing Antibody Center, and Collaboration for AIDS Vaccine Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, International AIDS Vaccine Initiative Neutralizing Antibody Center, and Collaboration for AIDS Vaccine Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
270
|
McCombs JE, Kohler JJ. Pneumococcal Neuraminidase Substrates Identified through Comparative Proteomics Enabled by Chemoselective Labeling. Bioconjug Chem 2016; 27:1013-22. [PMID: 26954852 DOI: 10.1021/acs.bioconjchem.6b00050] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neuraminidases (sialidases) are enzymes that hydrolytically remove sialic acid from sialylated proteins and lipids. Neuraminidases are encoded by a range of human pathogens, including bacteria, viruses, fungi, and protozoa. Many pathogen neuraminidases are virulence factors, indicating that desialylation of host glycoconjugates can be a critical step in infection. Specifically, desialylation of host cell surface glycoproteins can enable these molecules to function as pathogen receptors or can alter signaling through the plasma membrane. Despite these critical effects, no unbiased approaches exist to identify glycoprotein substrates of neuraminidases. Here, we combine previously reported glycoproteomics methods with quantitative proteomics analysis to identify glycoproteins whose sialylation changes in response to neuraminidase treatment. The two glycoproteomics methods-periodate oxidation and aniline-catalyzed oxime ligation (PAL) and galactose oxidase and aniline-catalyzed oxime ligation (GAL)-rely on chemoselective labeling of sialylated and nonsialylated glycoproteins, respectively. We demonstrated the utility of the combined approaches by identifying substrates of two pneumococcal neuraminidases in a human cell line that models the blood-brain barrier. The methods deliver complementary lists of neuraminidase substrates, with GAL identifying a larger number of substrates than PAL (77 versus 17). Putative neuraminidase substrates were confirmed by other methods, establishing the validity of the approach. Among the identified substrates were host glycoproteins known to function in bacteria adherence and infection. Functional assays suggest that multiple desialylated cell surface glycoproteins may act together as pneumococcus receptors. Overall, this method will provide a powerful approach to identify glycoproteins that are desialylated by both purified neuraminidases and intact pathogens.
Collapse
Affiliation(s)
- Janet E McCombs
- Department of Biochemistry, The University of Texas Southwestern Medical Center , Dallas, Texas 75390-9038, United States
| | - Jennifer J Kohler
- Department of Biochemistry, The University of Texas Southwestern Medical Center , Dallas, Texas 75390-9038, United States
| |
Collapse
|
271
|
Piller F, Mongis A, Piller V. Metabolic Glyco-Engineering in Eukaryotic Cells and Selected Applications. Methods Mol Biol 2016; 1321:335-59. [PMID: 26082233 DOI: 10.1007/978-1-4939-2760-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
By metabolic glyco-engineering cellular glycoconjugates are modified through the incorporation of synthetic monosaccharides which are usually analogues of naturally present sugars. In order to get incorporated, the monosaccharides need to enter the cytoplasm and to be substrates for the enzymes necessary for their transformation into activated sugars, most often nucleotide sugars. These have to be substrates for glycosyltransferases which finally catalyze their incorporation into glycans. Such pathways are difficult to reconstitute in vitro and therefore new monosaccharide analogues have to be tested in tissue culture for their suitability in metabolic glyco-engineering. For this, glycosylation mutants are the most appropriate since they are unable to synthesize specific glycans but through the introduction of the monosaccharide analogues they may express some glycans at the cell surface with the unnatural sugar incorporated. The presence of those glycans can be easily and quantitatively detected by lectin binding or by chemical methods identifying specific sugars. Monosaccharide analogues can also block the pathways leading to sugar incorporation, thus inhibiting the synthesis of glycan structures which is also easily detectable at the cell surface by lectin labeling. The most useful and most frequently employed application of metabolic glyco-engineering is the introduction of reactive groups which can undergo bio-orthogonal click reactions for the efficient labeling of glycans at the surface of live cells.
Collapse
Affiliation(s)
- Friedrich Piller
- Synthetic Protein Chemistry and Glyco-engineering Group, Centre de Biophysique Moléculaire (CNRS UPR 4301), Orléans, France
| | | | | |
Collapse
|
272
|
N-glycosylation heterogeneity and the influence on structure, function and pharmacokinetics of monoclonal antibodies and Fc fusion proteins. Eur J Pharm Biopharm 2016; 100:94-100. [DOI: 10.1016/j.ejpb.2016.01.005] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/18/2022]
|
273
|
Sialyltransferase inhibition and recent advances. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:143-53. [DOI: 10.1016/j.bbapap.2015.07.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/31/2015] [Accepted: 07/08/2015] [Indexed: 12/15/2022]
|
274
|
Subedi GP, Johnson RW, Moniz HA, Moremen KW, Barb AW. High Yield Expression of Recombinant Human Proteins with the Transient Transfection of HEK293 Cells in Suspension. J Vis Exp 2015:e53568. [PMID: 26779721 PMCID: PMC4780855 DOI: 10.3791/53568] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The art of producing recombinant proteins with complex post-translational modifications represents a major challenge for studies of structure and function. The rapid establishment and high recovery from transiently-transfected mammalian cell lines addresses this barrier and is an effective means of expressing proteins that are naturally channeled through the ER and Golgi-mediated secretory pathway. Here is one protocol for protein expression using the human HEK293F and HEK293S cell lines transfected with a mammalian expression vector designed for high protein yields. The applicability of this system is demonstrated using three representative glycoproteins that expressed with yields between 95-120 mg of purified protein recovered per liter of culture. These proteins are the human FcγRIIIa and the rat α2-6 sialyltransferase, ST6GalI, both expressed with an N-terminal GFP fusion, as well as the unmodified human immunoglobulin G1 Fc. This robust system utilizes a serum-free medium that is adaptable for expression of isotopically enriched proteins and carbohydrates for structural studies using mass spectrometry and nuclear magnetic resonance spectroscopy. Furthermore, the composition of the N-glycan can be tuned by adding a small molecule to prevent certain glycan modifications in a manner that does not reduce yield.
Collapse
Affiliation(s)
- Ganesh P Subedi
- The Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University
| | - Roy W Johnson
- Complex Carbohydrate Research Center, University of Georgia
| | | | | | - Adam W Barb
- The Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University;
| |
Collapse
|
275
|
Wu Z, Zhao G, Li T, Qu J, Guan W, Wang J, Ma C, Li X, Zhao W, Wang PG, Li L. Biochemical characterization of an α1,2-colitosyltransferase from Escherichia coli O55:H7. Glycobiology 2015; 26:493-500. [PMID: 26703456 DOI: 10.1093/glycob/cwv169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/16/2015] [Indexed: 01/17/2023] Open
Abstract
Colitose, also known as 3,6-dideoxy-L-galactose or 3-deoxy-L-fucose, is one of only five naturally occurring 3,6-dideoxyhexoses. Colitose was found in lipopolysaccharide of a number of infectious bacteria, including Escherichia coli O55 & O111 and Vibrio cholera O22 & O139. To date, no colitosyltransferase (ColT) has been characterized, probably due to the inaccessibility of the sugar donor, GDP-colitose. In this study, starting with chemically prepared colitose, 94.6 mg of GDP-colitose was prepared via a facile and efficient one-pot two-enzyme system involving an L-fucokinase/GDP-L-Fuc pyrophosphorylase and an inorganic pyrophosphatase (EcPpA). WbgN, a putative ColT from E. coliO55:H5 was then cloned, overexpressed, purified and biochemically characterized by using GDP-colitose as a sugar donor. Activity assay and structural identification of the synthetic product clearly demonstrated that wbgN encodes an α1,2-ColT. Biophysical study showed that WbgN does not require metal ion, and is highly active at pH 7.5-9.0. In addition, acceptor specificity study indicated that WbgN exclusively recognizes lacto-N-biose (Galβ1,3-GlcNAc). Most interestingly, it was found that WbgN exhibits similar activity toward GDP-l-Fuc (kcat/Km= 9.2 min(-1)mM(-1)) as that toward GDP-colitose (kcat/Km= 12 min(-1)mM(-1)). Finally, taking advantage of this, type 1 H-antigen was successfully synthesized in preparative scale.
Collapse
Affiliation(s)
- Zhigang Wu
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Guohui Zhao
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Tiehai Li
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Jingyao Qu
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Wanyi Guan
- College of Life Science, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Jiajia Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 30071, China
| | - Cheng Ma
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Xu Li
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Wei Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 30071, China
| | - Peng G Wang
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, USA State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 30071, China
| | | |
Collapse
|
276
|
Vajaria BN, Patel KR, Begum R, Patel PS. Sialylation: an Avenue to Target Cancer Cells. Pathol Oncol Res 2015; 22:443-7. [PMID: 26685886 DOI: 10.1007/s12253-015-0033-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 12/15/2015] [Indexed: 02/01/2023]
Abstract
Tumorigenesis and metastasis are frequently associated with altered structure and expression of oligosaccharides on cell surface glycoproteins and glycolipids. The expression of sialylated glycoconjugates has been shown to change during development, differentiation, disease and oncogenic transformation. Abnormal sialylation in cancer cell is a distinctive feature associated with malignant properties including invasiveness and metastatic potential. The alterations in sialylation is accompanied by changes in sialic acid, sialidase activity, sialyltransferase (ST) activity or sialoproteins. The present review summarizes the reports on alterations of sialic acid, linkage specific STs and sialoproteins, sialidase activity together with different subtypes of ST and sialidases mRNA expressions in various cancers like lung, breast, oral, cervical, ovarian, pancreatic etc. Sialic acids are widely distributed in nature as terminal sugars of oligosaccharides attached to proteins or lipids. The increase shedding of sialic acid observed in malignant tumors may be due to different types of sialidases. The amount of sialic acid is governed by levels of sialidases and STs. Various types of STs are also involved in formation of different types sialylated tumor associated carbohydrate antigens which plays important role in metastasis. The alterations associated with sialylation aids in early diagnosis, prognosis and post treatment monitoring in various cancers. Recently newer drugs targeting different interplays of sialylation have been developed, which might have profound effect in inhibiting sialylation and thus cancer metastasis and infiltration.
Collapse
Affiliation(s)
- Bhairavi N Vajaria
- Biochemistry Research Division, Department of Cancer Biology, The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, Gujarat, 380 016, India
| | - Kinjal R Patel
- Biochemistry Research Division, Department of Cancer Biology, The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, Gujarat, 380 016, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, The M. S. University of Baroda, Vadodara, Gujarat, India
| | - Prabhudas S Patel
- Biochemistry Research Division, Department of Cancer Biology, The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, Gujarat, 380 016, India.
| |
Collapse
|
277
|
A systematic analysis of acceptor specificity and reaction kinetics of five human α(2,3)sialyltransferases: Product inhibition studies illustrate reaction mechanism for ST3Gal-I. Biochem Biophys Res Commun 2015; 469:606-12. [PMID: 26692484 DOI: 10.1016/j.bbrc.2015.11.130] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 11/29/2015] [Indexed: 01/09/2023]
Abstract
Sialyltransferases (STs) catalyze the addition of sialic acids to the non-reducing ends of glycoproteins and glycolipids. In this work, we examined the acceptor specificity of five human α(2,3)sialyltransferases, namely ST3Gal -I, -II, -III, -IV and -VI. KM values for each of these enzymes is presented using radioactivity for acceptors containing Type-I (Galβ1,3GlcNAc), Type-II (Galβ1,4GlcNAc), Type-III (Galβ1,3GalNAc) and Core-2 (Galβ1,3(GlcNAcβ1,6)GalNAc) reactive groups. Several variants of acceptors inhibited ST3Gal activity emphasizing structural role of acceptor in enzyme-catalyzed reactions. In some cases, mass spectrometry was performed for structural verification. The results demonstrate human ST3Gal-I catalysis towards Type-III and Core-2 acceptors with KM = 5-50 μM and high VMax values. The KM for ST3Gal-I and ST3Gal-II was 100 and 30-fold lower, respectively, for Type-III compared to Type-I acceptors. Variants of Type-I and Type-II structures characterized ST3Gal-III, -IV and -VI for their catalytic specificity. This manuscript also estimates KM for human ST3Gal-VI using Type-I and Type-II substrates. Together, these findings built a platform for designing inhibitors of STs having therapeutic potential.
Collapse
|
278
|
Upadhyaya K, Hamidullah, Singh K, Arun A, Shukla M, Srivastava N, Ashraf R, Sharma A, Mahar R, Shukla SK, Sarkar J, Ramachandran R, Lal J, Konwar R, Tripathi RP. Identification of gallic acid based glycoconjugates as a novel tubulin polymerization inhibitors. Org Biomol Chem 2015; 14:1338-58. [PMID: 26659548 DOI: 10.1039/c5ob02113h] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A novel class of gallic acid based glycoconjugates were designed and synthesized as potential anticancer agents. Among all the compounds screened, compound 2a showed potent anticancer activity against breast cancer cells. The latter resulted in tubulin polymerization inhibition and induced G2/M cell cycle arrest, generation of reactive oxygen species, mitochondrial depolarization and subsequent apoptosis in breast cancer cells. In addition, ultraviolet-visible spectroscopy and fluorescence quenching studies of the compound with tubulin confirmed direct interaction of compounds with tubulin. Molecular modeling studies revealed that it binds at the colchicine binding site in tubulin. Further, 2a also exhibited potent in vivo anticancer activity in LA-7 syngeneic rat mammary tumor model. Current data projects its strong candidature to be developed as anticancer agent.
Collapse
Affiliation(s)
- Kapil Upadhyaya
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Dumont M, Lehner A, Bardor M, Burel C, Vauzeilles B, Lerouxel O, Anderson CT, Mollet JC, Lerouge P. Inhibition of fucosylation of cell wall components by 2-fluoro 2-deoxy-L-fucose induces defects in root cell elongation. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2015; 84:1137-51. [PMID: 26565655 DOI: 10.1111/tpj.13071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/26/2015] [Accepted: 11/03/2015] [Indexed: 05/21/2023]
Abstract
Screening of commercially available fluoro monosaccharides as putative growth inhibitors in Arabidopsis thaliana revealed that 2-fluoro 2-l-fucose (2F-Fuc) reduces root growth at micromolar concentrations. The inability of 2F-Fuc to affect an Atfkgp mutant that is defective in the fucose salvage pathway indicates that 2F-Fuc must be converted to its cognate GDP nucleotide sugar in order to inhibit root growth. Chemical analysis of cell wall polysaccharides and glycoproteins demonstrated that fucosylation of xyloglucans and of N-linked glycans is fully inhibited by 10 μm 2F-Fuc in Arabidopsis seedling roots, but genetic evidence indicates that these alterations are not responsible for the inhibition of root development by 2F-Fuc. Inhibition of fucosylation of cell wall polysaccharides also affected pectic rhamnogalacturonan-II (RG-II). At low concentrations, 2F-Fuc induced a decrease in RG-II dimerization. Both RG-II dimerization and root growth were partially restored in 2F-Fuc-treated seedlings by addition of boric acid, suggesting that the growth phenotype caused by 2F-Fuc was due to a deficiency of RG-II dimerization. Closer investigation of the 2F-Fuc-induced growth phenotype demonstrated that cell division is not affected by 2F-Fuc treatments. In contrast, the inhibitor suppressed elongation of root cells and promoted the emergence of adventitious roots. This study further emphasizes the importance of RG-II in cell elongation and the utility of glycosyltransferase inhibitors as new tools for studying the functions of cell wall polysaccharides in plant development. Moreover, supplementation experiments with borate suggest that the function of boron in plants might not be restricted to RG-II cross-linking, but that it might also be a signal molecule in the cell wall integrity-sensing mechanism.
Collapse
Affiliation(s)
- Marie Dumont
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale, EA 4358, IRIB, VASI, Normandie Université, 76821, Mont-Saint-Aignan, France
| | - Arnaud Lehner
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale, EA 4358, IRIB, VASI, Normandie Université, 76821, Mont-Saint-Aignan, France
| | - Muriel Bardor
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale, EA 4358, IRIB, VASI, Normandie Université, 76821, Mont-Saint-Aignan, France
| | - Carole Burel
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale, EA 4358, IRIB, VASI, Normandie Université, 76821, Mont-Saint-Aignan, France
| | - Boris Vauzeilles
- Institut de Chimie Moléculaire et des Matériaux d'Orsay (ICMMO) UMR CNRS 8182, Université de Paris Sud, 91405, Orsay, France
- Institut de Chimie des Substances Naturelles (ICSN) UPR CNRS 2301, 91198, Gif-sur-Yvette, France
- Click4Tag, Zone Luminy Biotech, Case 922, 163 Avenue de Luminy, 13009, Marseille, France
| | - Olivier Lerouxel
- Centre de Recherches sur les Macromolécules Végétales (CERMAV) - CNRS BP 53, 38041, Grenoble Cedex 9, France
| | - Charles T Anderson
- Department of Biology and Center for Lignocellulose Structure and Formation, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Jean-Claude Mollet
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale, EA 4358, IRIB, VASI, Normandie Université, 76821, Mont-Saint-Aignan, France
| | - Patrice Lerouge
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale, EA 4358, IRIB, VASI, Normandie Université, 76821, Mont-Saint-Aignan, France
| |
Collapse
|
280
|
Srivastava S, Makarava N, Katorcha E, Savtchenko R, Brossmer R, Baskakov IV. Post-conversion sialylation of prions in lymphoid tissues. Proc Natl Acad Sci U S A 2015; 112:E6654-62. [PMID: 26627256 PMCID: PMC4672809 DOI: 10.1073/pnas.1517993112] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sialylated glycans on the surface of mammalian cells act as part of a "self-associated molecular pattern," helping the immune system to recognize "self" from "altered self" or "nonself." To escape the host immune system, some bacterial pathogens have evolved biosynthetic pathways for host-like sialic acids, whereas others recruited host sialic acids for decorating their surfaces. Prions lack nucleic acids and are not conventional pathogens. Nevertheless, prions might use a similar strategy for invading and colonizing the lymphoreticular system. Here we show that the sialylation status of the infectious, disease-associated state of the prion protein (PrP(Sc)) changes with colonization of secondary lymphoid organs (SLOs). As a result, spleen-derived PrP(Sc) is more sialylated than brain-derived PrP(Sc). Enhanced sialylation of PrP(Sc) is recapitulated in vitro by incubating brain-derived PrP(Sc) with primary splenocytes or cultured macrophage RAW 264.7 cells. General inhibitors of sialyltranserases (STs), the enzymes that transfer sialic acid residues onto terminal positions of glycans, suppressed extrasialylation of PrP(Sc). A fluorescently labeled precursor of sialic acid revealed ST activity associated with RAW macrophages. This study illustrates that, upon colonization of SLOs, the sialylation status of prions changes by host STs. We propose that this mechanism is responsible for camouflaging prions in SLOs and has broad implications.
Collapse
Affiliation(s)
- Saurabh Srivastava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Elizaveta Katorcha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Regina Savtchenko
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Reinhard Brossmer
- Biochemistry Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ilia V Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201;
| |
Collapse
|
281
|
Henninot A, Terrier A, Charton J, Urbain R, Fontayne A, Deprez B, Beghyn T. Characterization of monoclonal antibodies by a fast and easy liquid chromatography–mass spectrometry time-of-flight analysis on culture supernatant. Anal Biochem 2015; 491:52-4. [DOI: 10.1016/j.ab.2015.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 07/22/2015] [Accepted: 08/05/2015] [Indexed: 11/29/2022]
|
282
|
Katorcha E, Klimova N, Makarava N, Savtchenko R, Pan X, Annunziata I, Takahashi K, Miyagi T, Pshezhetsky AV, d’Azzo A, Baskakov IV. Loss of Cellular Sialidases Does Not Affect the Sialylation Status of the Prion Protein but Increases the Amounts of Its Proteolytic Fragment C1. PLoS One 2015; 10:e0143218. [PMID: 26569607 PMCID: PMC4646690 DOI: 10.1371/journal.pone.0143218] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/02/2015] [Indexed: 11/29/2022] Open
Abstract
The central molecular event underlying prion diseases involves conformational change of the cellular form of the prion protein (PrPC), which is a sialoglycoprotein, into the disease-associated, transmissible form denoted PrPSc. Recent studies revealed a correlation between the sialylation status of PrPSc and incubation time to disease and introduced a new hypothesis that progression of prion diseases could be controlled or reversed by altering the sialylation level of PrPC. Of the four known mammalian sialidases, the enzymes that cleave off sialic acid residues, only NEU1, NEU3 and NEU4 are expressed in the brain. To test whether cellular sialidases control the steady-state sialylation level of PrPC and to identify the putative sialidase responsible for desialylating PrPC, we analyzed brain-derived PrPC from knockout mice deficient in Neu1, Neu3, Neu4, or from Neu3/Neu4 double knockouts. Surprisingly, no differences in the sialylation of PrPC or its proteolytic product C1 were noticed in any of the knockout mice tested as compared to the age-matched controls. However, significantly higher amounts of the C1 fragment relative to full-length PrPC were detected in the brains of Neu1 knockout mice as compared to WT mice or to the other knockout mice. Additional experiments revealed that in neuroblastoma cell line the sialylation pattern of C1 could be changed by an inhibitor of sialylatransferases. In summary, this study suggests that targeting cellular sialidases is apparently not the correct strategy for altering the sialylation levels of PrPC, whereas modulating the activity of sialylatransferases might offer a more promising approach. Our findings also suggest that catabolism of PrPC involves its α-cleavage followed by desialylation of the resulting C1 fragments by NEU1 and consequent fast degradation of the desialylated products.
Collapse
Affiliation(s)
- Elizaveta Katorcha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Nina Klimova
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Regina Savtchenko
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Xuefang Pan
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, QC, Canada
| | - Ida Annunziata
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Kohta Takahashi
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan
| | - Taeko Miyagi
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan
| | - Alexey V. Pshezhetsky
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, QC, Canada
| | - Alessandra d’Azzo
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ilia V. Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
283
|
Bai Q, Liu L, Xia Y, Long Q, Wang J, Xu J, Guo J. Prognostic significance of ST3GAL-1 expression in patients with clear cell renal cell carcinoma. BMC Cancer 2015; 15:880. [PMID: 26552809 PMCID: PMC4640103 DOI: 10.1186/s12885-015-1906-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/03/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Aberrant sialylated carbohydrate synthesis is frequently noted in various cancers. Sialyltransferase ST3GAL-1, which adds a sialic acid in an α-2,3 linkage to Gal β1,3 GalNAc, preforms an important role in modulating cellular behaviors. However, little is known about prognostic significance of ST3GAL-1 in clear cell renal cell carcinoma (ccRCC). In this study, we aimed to investigate the prognostic significance of sialyltransferase ST3GAL-1 and its correlation with clinical outcomes in patients with ccRCC. METHODS A total of 286 patients who underwent nephrectomy between 2005 and 2007 in a single academic center were recruited. Immunohistochemical staining was performed on tissue microarrays to assess the expression level. Kaplan-Meier method and Cox proportional hazard model were applied to assess the prognostic value of ST3GAL-1. Nomograms were generated as prediction model for overall survival and disease free survival at 5 and 8 years after nephrectomy. RESULTS The present results show high expression of ST3GAL-1 is associated with reduced overall survival (p = 0.013) and disease free survival (p = 0.004). In multivariate cox analyses, ST3GAL-1 was defined as an independent prognostic factor for overall survival (p = 0.006) and disease free survival (p = 0.001). After incorporation into the University of California Integrated Staging System (UISS) intermediate/high risk group for non-metastatic ccRCC, ST3GAL-1 could further distinguish patient with dismal prognosis (p = 0.015 and 0.002 for OS and DFS respectively). The nomograms revealed better predictive accuracy in predicting 5- and 8- year overall survival and disease free survival than the TNM stage alone. CONCLUSIONS ST3GAL-1 is an independent adverse prognostic factor for recurrence and survival of patients with ccRCC.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/genetics
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/surgery
- Disease-Free Survival
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Male
- Middle Aged
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/surgery
- Nephrectomy
- Prognosis
- Sialyltransferases/biosynthesis
- Sialyltransferases/genetics
- beta-Galactoside alpha-2,3-Sialyltransferase
Collapse
Affiliation(s)
- Qi Bai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yu Xia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Qilai Long
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jiajun Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
284
|
Wagner GK, Pesnot T, Palcic MM, Jørgensen R. Novel UDP-GalNAc Derivative Structures Provide Insight into the Donor Specificity of Human Blood Group Glycosyltransferase. J Biol Chem 2015; 290:31162-72. [PMID: 26527682 PMCID: PMC4692239 DOI: 10.1074/jbc.m115.681262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Indexed: 01/17/2023] Open
Abstract
Two closely related glycosyltransferases are responsible for the final step of the biosynthesis of ABO(H) human blood group A and B antigens. The two enzymes differ by only four amino acid residues, which determine whether the enzymes transfer GalNAc from UDP-GalNAc or Gal from UDP-Gal to the H-antigen acceptor. The enzymes belong to the class of GT-A folded enzymes, grouped as GT6 in the CAZy database, and are characterized by a single domain with a metal dependent retaining reaction mechanism. However, the exact role of the four amino acid residues in the specificity of the enzymes is still unresolved. In this study, we report the first structural information of a dual specificity cis-AB blood group glycosyltransferase in complex with a synthetic UDP-GalNAc derivative. Interestingly, the GalNAc moiety adopts an unusual yet catalytically productive conformation in the binding pocket, which is different from the "tucked under" conformation previously observed for the UDP-Gal donor. In addition, we show that this UDP-GalNAc derivative in complex with the H-antigen acceptor provokes the same unusual binding pocket closure as seen for the corresponding UDP-Gal derivative. Despite this, the two derivatives show vastly different kinetic properties. Our results provide a important structural insight into the donor substrate specificity and utilization in blood group biosynthesis, which can very likely be exploited for the development of new glycosyltransferase inhibitors and probes.
Collapse
Affiliation(s)
- Gerd K Wagner
- From the Department of Chemistry, King's College London, Faculty of Natural & Mathematical Sciences, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Thomas Pesnot
- the University of East Anglia, School of Pharmacy, Norwich NR47TJ, England, and
| | - Monica M Palcic
- the Carlsberg Laboratory, Gamle Carlsberg Vej 10, DK-1799, Copenhagen V, Denmark
| | - Rene Jørgensen
- the Carlsberg Laboratory, Gamle Carlsberg Vej 10, DK-1799, Copenhagen V, Denmark
| |
Collapse
|
285
|
Wands AM, Fujita A, McCombs JE, Cervin J, Dedic B, Rodriguez AC, Nischan N, Bond MR, Mettlen M, Trudgian DC, Lemoff A, Quiding-Järbrink M, Gustavsson B, Steentoft C, Clausen H, Mirzaei H, Teneberg S, Yrlid U, Kohler JJ. Fucosylation and protein glycosylation create functional receptors for cholera toxin. eLife 2015; 4:e09545. [PMID: 26512888 PMCID: PMC4686427 DOI: 10.7554/elife.09545] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022] Open
Abstract
Cholera toxin (CT) enters and intoxicates host cells after binding cell surface receptors using its B subunit (CTB). The ganglioside (glycolipid) GM1 is thought to be the sole CT receptor; however, the mechanism by which CTB binding to GM1 mediates internalization of CT remains enigmatic. Here we report that CTB binds cell surface glycoproteins. Relative contributions of gangliosides and glycoproteins to CTB binding depend on cell type, and CTB binds primarily to glycoproteins in colonic epithelial cell lines. Using a metabolically incorporated photocrosslinking sugar, we identified one CTB-binding glycoprotein and demonstrated that the glycan portion of the molecule, not the protein, provides the CTB interaction motif. We further show that fucosylated structures promote CTB entry into a colonic epithelial cell line and subsequent host cell intoxication. CTB-binding fucosylated glycoproteins are present in normal human intestinal epithelia and could play a role in cholera. DOI:http://dx.doi.org/10.7554/eLife.09545.001 Cholera is a serious diarrheal disease that can be deadly if left untreated. It is caused by eating food, or drinking water, contaminated by the bacterium Vibrio cholerae. This bacterium can survive passage through the acidic conditions of the stomach. Inside the small intestine, V. cholerae attaches to the intestinal wall and starts producing cholera toxin. The toxin enters intestinal cells, causing them to release water and ions, including sodium and chloride ions. The salt-water environment created inside the intestine can, by osmosis, draw up to a further six liters of water into the intestine each day. This results in the copious production of watery diarrhea and severe dehydration. Cholera toxin is composed of six protein subunits, including five copies of cholera toxin subunit B (CTB). CTB subunits help the uptake of the toxin by intestinal cells, and it has long been reported that CTB subunits attach to intestinal cells by binding to a cell surface molecule called GM1. CTB subunits have a high affinity for GM1, yet recent work suggests CTB may not bind exclusively to GM1; one or more additional cell surface molecules may be directly involved in cholera toxin uptake. Wands et al. now reveal that numerous cell surface molecules are recognized by CTB, and that these molecules can assist cholera toxin uptake by host cells. Glycoproteins, proteins that are marked with sugar molecules, were shown to be the primary CTB binding sites on human colon cells, and it was the glycoprotein’s sugar component, not the protein itself, that interacted with CTB. Wands et al. discovered that in particular glycoproteins containing a sugar called fucose were largely responsible for CTB binding and toxin uptake. Together these findings reveal a previously unrecognized mechanism for cholera toxin entry into host cells, and suggest that fucose-containing or fucose-mimicking molecules could be developed as new treatments for cholera. DOI:http://dx.doi.org/10.7554/eLife.09545.002
Collapse
Affiliation(s)
- Amberlyn M Wands
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Akiko Fujita
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Janet E McCombs
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jakob Cervin
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Benjamin Dedic
- Department of Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andrea C Rodriguez
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Nicole Nischan
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Michelle R Bond
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Marcel Mettlen
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - David C Trudgian
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Marianne Quiding-Järbrink
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Bengt Gustavsson
- Department of Surgery, University of Gothenburg, Gothenburg, Sweden
| | - Catharina Steentoft
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hamid Mirzaei
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Susann Teneberg
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Department of Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Jennifer J Kohler
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
286
|
Büll C, Heise T, Beurskens DMH, Riemersma M, Ashikov A, Rutjes FPJT, van Kuppevelt TH, Lefeber DJ, den Brok MH, Adema GJ, Boltje TJ. Sialic Acid Glycoengineering Using an Unnatural Sialic Acid for the Detection of Sialoglycan Biosynthesis Defects and On-Cell Synthesis of Siglec Ligands. ACS Chem Biol 2015; 10:2353-63. [PMID: 26258433 DOI: 10.1021/acschembio.5b00501] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sialoglycans play a vital role in physiology, and aberrant sialoglycan expression is associated with a broad spectrum of diseases. Since biosynthesis of sialoglycans is only partially regulated at the genetic level, chemical tools are crucial to study their function. Here, we report the development of propargyloxycarbonyl sialic acid (Ac5NeuNPoc) as a powerful tool for sialic acid glycoengineering. Ac5NeuNPoc showed strongly increased labeling efficiency and exhibited less toxicity compared to those of widely used mannosamine analogues in vitro and was also more efficiently incorporated into sialoglycans in vivo. Unlike mannosamine analogues, Ac5NeuNPoc was exclusively utilized in the sialoglycan biosynthesis pathway, allowing a genetic defect in sialic acid biosynthesis to be specifically detected. Furthermore, Ac5NeuNPoc-based sialic acid glycoengineering enabled the on-cell synthesis of high-affinity Siglec-7 ligands and the identification of a novel Siglec-2 ligand. Thus, Ac5NeuNPoc glycoengineering is a highly efficient, nontoxic, and selective approach to study and modulate sialoglycan interactions on living cells.
Collapse
Affiliation(s)
| | - Torben Heise
- Cluster
for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | | | | | | | - Floris P. J. T. Rutjes
- Cluster
for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | | | | | | | | | - Thomas J. Boltje
- Cluster
for Molecular Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| |
Collapse
|
287
|
Villalobos JA, Yi BR, Wallace IS. 2-Fluoro-L-Fucose Is a Metabolically Incorporated Inhibitor of Plant Cell Wall Polysaccharide Fucosylation. PLoS One 2015; 10:e0139091. [PMID: 26414071 PMCID: PMC4587364 DOI: 10.1371/journal.pone.0139091] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/09/2015] [Indexed: 12/29/2022] Open
Abstract
The monosaccharide L-fucose (L-Fuc) is a common component of plant cell wall polysaccharides and other plant glycans, including the hemicellulose xyloglucan, pectic rhamnogalacturonan-I (RG-I) and rhamnogalacturonan-II (RG-II), arabinogalactan proteins, and N-linked glycans. Mutations compromising the biosynthesis of many plant cell wall polysaccharides are lethal, and as a result, small molecule inhibitors of plant cell wall polysaccharide biosynthesis have been developed because these molecules can be applied at defined concentrations and developmental stages. In this study, we characterize novel small molecule inhibitors of plant fucosylation. 2-fluoro-L-fucose (2F-Fuc) analogs caused severe growth phenotypes when applied to Arabidopsis seedlings, including reduced root growth and altered root morphology. These phenotypic defects were dependent upon the L-Fuc salvage pathway enzyme L-Fucose Kinase/ GDP-L-Fucose Pyrophosphorylase (FKGP), suggesting that 2F-Fuc is metabolically converted to the sugar nucleotide GDP-2F-Fuc, which serves as the active inhibitory molecule. The L-Fuc content of cell wall matrix polysaccharides was reduced in plants treated with 2F-Fuc, suggesting that this molecule inhibits the incorporation of L-Fuc into these polysaccharides. Additionally, phenotypic defects induced by 2F-Fuc treatment could be partially relieved by the exogenous application of boric acid, suggesting that 2F-Fuc inhibits RG-II biosynthesis. Overall, the results presented here suggest that 2F-Fuc is a metabolically incorporated inhibitor of plant cellular fucosylation events, and potentially suggest that other 2-fluorinated monosaccharides could serve as useful chemical probes for the inhibition of cell wall polysaccharide biosynthesis.
Collapse
Affiliation(s)
- Jose A. Villalobos
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, Nevada, 89557, United States of America
| | - Bo R. Yi
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, Nevada, 89557, United States of America
| | - Ian S. Wallace
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, Nevada, 89557, United States of America
- * E-mail:
| |
Collapse
|
288
|
Chugh S, Gnanapragassam VS, Jain M, Rachagani S, Ponnusamy MP, Batra SK. Pathobiological implications of mucin glycans in cancer: Sweet poison and novel targets. Biochim Biophys Acta Rev Cancer 2015; 1856:211-25. [PMID: 26318196 DOI: 10.1016/j.bbcan.2015.08.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/15/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022]
Abstract
Mucins are large glycoproteins expressed on the epithelia that provide a protective barrier against harsh insults from toxins and pathogenic microbes. These glycoproteins are classified primarily as being secreted and membrane-bound; both forms are involved in pathophysiological functions including inflammation and cancer. The high molecular weight of mucins is attributed to their large polypeptide backbone that is extensively covered by glycan moieties that modulate the function of mucins and, hence, play an important role in physiological functions. Deregulation of glycosylation machinery during malignant transformation results in altered mucin glycosylation. This review describes the functional implications and pathobiological significance of altered mucin glycosylation in cancer. Further, this review delineates various factors such as glycosyltransferases and tumor microenvironment that contribute to dysregulation of mucin glycosylation during cancer. Finally, this review discusses the scope of mucin glycan epitopes as potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Seema Chugh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Vinayaga S Gnanapragassam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| |
Collapse
|
289
|
Glycosyltransferase ST6GAL1 contributes to the regulation of pluripotency in human pluripotent stem cells. Sci Rep 2015; 5:13317. [PMID: 26304831 PMCID: PMC4548446 DOI: 10.1038/srep13317] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/21/2015] [Indexed: 01/12/2023] Open
Abstract
Many studies have suggested the significance of glycosyltransferase-mediated macromolecule glycosylation in the regulation of pluripotent states in human pluripotent stem cells (hPSCs). Here, we observed that the sialyltransferase ST6GAL1 was preferentially expressed in undifferentiated hPSCs compared to non-pluripotent cells. A lectin which preferentially recognizes α-2,6 sialylated galactosides showed strong binding reactivity with undifferentiated hPSCs and their glycoproteins, and did so to a much lesser extent with differentiated cells. In addition, downregulation of ST6GAL1 in undifferentiated hPSCs led to a decrease in POU5F1 (also known as OCT4) protein and significantly altered the expression of many genes that orchestrate cell morphogenesis during differentiation. The induction of cellular pluripotency in somatic cells was substantially impeded by the shRNA-mediated suppression of ST6GAL1, partially through interference with the expression of endogenous POU5F1 and SOX2. Targeting ST6GAL1 activity with a sialyltransferase inhibitor during cell reprogramming resulted in a dose-dependent reduction in the generation of human induced pluripotent stem cells (hiPSCs). Collectively, our data indicate that ST6GAL1 plays an important role in the regulation of pluripotency and differentiation in hPSCs, and the pluripotent state in human cells can be modulated using pharmacological tools to target sialyltransferase activity.
Collapse
|
290
|
Desiderio V, Papagerakis P, Tirino V, Zheng L, Matossian M, Prince ME, Paino F, Mele L, Papaccio F, Montella R, Papaccio G, Papagerakis S. Increased fucosylation has a pivotal role in invasive and metastatic properties of head and neck cancer stem cells. Oncotarget 2015; 6:71-84. [PMID: 25428916 PMCID: PMC4381579 DOI: 10.18632/oncotarget.2698] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/06/2014] [Indexed: 12/14/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an aggressive malignancy with high mortality rates. Major challenges for OSCC management include development of resistance to therapy and early formation of distant metastases. Cancer stem cells (CSCs) have emerged as important players in both pathologic mechanisms. Increased fucosylation activity and increased expression of fucosylated polysaccharides, such as Sialyl Lewis X (SLex), are associated with invasion and metastasis. However, the role of fucosylation in CSCs has not been elucidated yet. We used the spheroid culture technique to obtain a CSC-enriched population and compared orospheres with adherent cells. We found that orospheres expressed markers of CSCs and metastasis at higher levels, were more invasive and tumorigenic, and were more resistant to cisplatin/radiation than adherent counterparts. We found fucosyltransferases FUT3 and FUT6 highly up-regulated, increased SLex expression and increased adhesion by shear flow assays in orospheres. Inhibition of fucosylation negatively affected orospheres formation and invasion of oral CSCs. These results confirm that orospheres are enriched in CSCs and that fucosylation is of paramount importance for CSC invasion. In addition, SLex may play a key role in CSC metastasis. Thus, inhibition of fucosylation may be used to block CSCs and metastatic spread.
Collapse
Affiliation(s)
- Vincenzo Desiderio
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA. Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples, Italy
| | - Petros Papagerakis
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA. Center for Organogenesis, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Virginia Tirino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples, Italy
| | - Li Zheng
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Margarite Matossian
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Mark E Prince
- Department of Otolaryngology, Head and Neck Surgery, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Francesca Paino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples, Italy
| | - Luigi Mele
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA. Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples, Italy
| | - Federica Papaccio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples, Italy
| | - Roberta Montella
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples, Italy
| | - Silvana Papagerakis
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA. Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
291
|
Rota P, Cirillo F, Piccoli M, Gregorio A, Tettamanti G, Allevi P, Anastasia L. Synthesis and biological evaluation of several dephosphonated analogues of CMP-Neu5Ac as inhibitors of GM3-synthase. Chemistry 2015; 21:14614-29. [PMID: 26397189 DOI: 10.1002/chem.201501770] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Indexed: 01/02/2023]
Abstract
Previous studies demonstrated that reducing the GM3 content in myoblasts increased the cell resistance to hypoxic stress, suggesting that a pharmacological inhibition of the GM3 synthesis could be instrumental for the development of new treatments for ischemic diseases. Herein, the synthesis of several dephosphonated CMP-Neu5Ac congeners and their anti-GM3-synthase activity is reported. Biological activity testes revealed that some inhibitors almost completely blocked the GM3-synthase activity in vitro and reduced the GM3 content in living embryonic kidney 293A cells, eventually activating the epidermal growth factor receptor (EGFR) signaling cascade.
Collapse
Affiliation(s)
- Paola Rota
- Department of Biomedical, Surgical and Dental Sciences, Institution University of Milan, Via Saldini 50, 20133 Milan (Italy)
| | - Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Milan (Italy)
| | - Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Milan (Italy)
| | - Antonio Gregorio
- Department of Biomedical, Surgical and Dental Sciences, Institution University of Milan, Via Saldini 50, 20133 Milan (Italy)
| | - Guido Tettamanti
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Milan (Italy)
| | - Pietro Allevi
- Department of Biomedical, Surgical and Dental Sciences, Institution University of Milan, Via Saldini 50, 20133 Milan (Italy)
| | - Luigi Anastasia
- Department of Biomedical Sciences for Health, Institution University of Milan, Segrate Milan (Italy). .,Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Milan (Italy).
| |
Collapse
|
292
|
Eller C, Chao TY, Singarapu KK, Ouerfelli O, Yang G, Markley JL, Danishefsky SJ, Raines RT. Human Cancer Antigen Globo H Is a Cell-Surface Ligand for Human Ribonuclease 1. ACS CENTRAL SCIENCE 2015; 1:181-190. [PMID: 26405690 PMCID: PMC4571170 DOI: 10.1021/acscentsci.5b00164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Indexed: 05/08/2023]
Abstract
Pancreatic-type ribonucleases are secretory enzymes that catalyze the cleavage of RNA. Recent efforts have endowed the homologues from cow (RNase A) and human (RNase 1) with toxicity for cancer cells, leading to a clinical trial. The basis for the selective toxicity of ribonuclease variants for cancerous versus noncancerous cells has, however, been unclear. A screen for RNase A ligands in an array of mammalian cell-surface glycans revealed strong affinity for a hexasaccharide, Globo H, that is a tumor-associated antigen and the basis for a vaccine in clinical trials. The affinity of RNase A and RNase 1 for immobilized Globo H is in the low micromolar-high nanomolar range. Moreover, reducing the display of Globo H on the surface of human breast adenocarcinoma cells with a small-molecule inhibitor of biosynthesis or a monoclonal antibody antagonist decreases the toxicity of an RNase 1 variant. Finally, heteronuclear single quantum coherence (HSQC) NMR spectroscopy showed that RNase 1 interacts with Globo H by using residues that are distal from the enzymic active site. The discovery that a systemic human ribonuclease binds to a moiety displayed on human cancer cells links two clinical paradigms and suggests a mechanism for innate resistance to cancer.
Collapse
Affiliation(s)
- Chelcie
H. Eller
- Department of Biochemistry, National Magnetic Resonance Facility
at Madison, and Department of Chemistry, University of
Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Tzu-Yuan Chao
- Department of Biochemistry, National Magnetic Resonance Facility
at Madison, and Department of Chemistry, University of
Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Kiran K. Singarapu
- Department of Biochemistry, National Magnetic Resonance Facility
at Madison, and Department of Chemistry, University of
Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Ouathek Ouerfelli
- Organic Synthesis Core
Facility and Laboratory for Bioorganic Chemistry, Memorial
Sloan Kettering Cancer Center, New York, New York 10021, United States
| | - Guangbin Yang
- Organic Synthesis Core
Facility and Laboratory for Bioorganic Chemistry, Memorial
Sloan Kettering Cancer Center, New York, New York 10021, United States
| | - John L. Markley
- Department of Biochemistry, National Magnetic Resonance Facility
at Madison, and Department of Chemistry, University of
Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Samuel J. Danishefsky
- Organic Synthesis Core
Facility and Laboratory for Bioorganic Chemistry, Memorial
Sloan Kettering Cancer Center, New York, New York 10021, United States
- Department
of Chemistry, Columbia University, New York, New York 10027, United States
| | - Ronald T. Raines
- Department of Biochemistry, National Magnetic Resonance Facility
at Madison, and Department of Chemistry, University of
Wisconsin—Madison, Madison, Wisconsin 53706, United States
- E-mail:
| |
Collapse
|
293
|
Vasconcelos-Dos-Santos A, Oliveira IA, Lucena MC, Mantuano NR, Whelan SA, Dias WB, Todeschini AR. Biosynthetic Machinery Involved in Aberrant Glycosylation: Promising Targets for Developing of Drugs Against Cancer. Front Oncol 2015; 5:138. [PMID: 26161361 PMCID: PMC4479729 DOI: 10.3389/fonc.2015.00138] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/02/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer cells depend on altered metabolism and nutrient uptake to generate and keep the malignant phenotype. The hexosamine biosynthetic pathway is a branch of glucose metabolism that produces UDP-GlcNAc and its derivatives, UDP-GalNAc and CMP-Neu5Ac and donor substrates used in the production of glycoproteins and glycolipids. Growing evidence demonstrates that alteration of the pool of activated substrates might lead to different glycosylation and cell signaling. It is already well established that aberrant glycosylation can modulate tumor growth and malignant transformation in different cancer types. Therefore, biosynthetic machinery involved in the assembly of aberrant glycans are becoming prominent targets for anti-tumor drugs. This review describes three classes of glycosylation, O-GlcNAcylation, N-linked, and mucin type O-linked glycosylation, involved in tumor progression, their biosynthesis and highlights the available inhibitors as potential anti-tumor drugs.
Collapse
Affiliation(s)
| | - Isadora A Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Miguel Clodomiro Lucena
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Natalia Rodrigues Mantuano
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Stephen A Whelan
- Department of Biochemistry, Cardiovascular Proteomics Center, Boston University School of Medicine , Boston, MA , USA
| | - Wagner Barbosa Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Adriane Regina Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| |
Collapse
|
294
|
Audfray A, Beldjoudi M, Breiman A, Hurbin A, Boos I, Unverzagt C, Bouras M, Lantuejoul S, Coll JL, Varrot A, Le Pendu J, Busser B, Imberty A. A recombinant fungal lectin for labeling truncated glycans on human cancer cells. PLoS One 2015; 10:e0128190. [PMID: 26042789 PMCID: PMC4456360 DOI: 10.1371/journal.pone.0128190] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/24/2015] [Indexed: 01/16/2023] Open
Abstract
Cell surface glycoconjugates present alterations of their structures in chronic diseases and distinct oligosaccharide epitopes have been associated with cancer. Among them, truncated glycans present terminal non-reducing β-N-acetylglucosamine (GlcNAc) residues that are rare on healthy tissues. Lectins from unconventional sources such as fungi or algi provide novel markers that bind specifically to such epitopes, but their availability may be challenging. A GlcNAc-binding lectin from the fruiting body of the fungus Psathyrella velutina (PVL) has been produced in good yield in bacterial culture. A strong specificity for terminal GlcNAc residues was evidenced by glycan array. Affinity values obtained by microcalorimetry and surface plasmon resonance demonstrated a micromolar affinity for GlcNAcβ1-3Gal epitopes and for biantennary N-glycans with GlcNAcβ1-2Man capped branches. Crystal structure of PVL complexed with GlcNAcβ1-3Gal established the structural basis of the specificity. Labeling of several types of cancer cells and use of inhibitors of glycan metabolism indicated that rPVL binds to terminal GlcNAc but also to sialic acid (Neu5Ac). Analysis of glycosyltransferase expression confirmed the higher amount of GlcNAc present on cancer cells. rPVL binding is specific to cancer tissue and weak or no labeling is observed for healthy ones, except for stomach glands that present unique αGlcNAc-presenting mucins. In lung, breast and colon carcinomas, a clear delineation could be observed between cancer regions and surrounding healthy tissues. PVL is therefore a useful tool for labeling agalacto-glycans in cancer or other diseases.
Collapse
Affiliation(s)
- Aymeric Audfray
- CERMAV, UPR5301, CNRS, University Grenoble Alpes, 38041 Grenoble, France
| | - Mona Beldjoudi
- IAB, University Grenoble Alpes, F-38000 Grenoble, France
- INSERM U823, IAB, F-38000 Grenoble, France
- University El Hadj Lakhdar, 05000 Batna, Algeria
| | - Adrien Breiman
- INSERM, UMR892, 44007 Nantes, France
- CNRS, UMR6299, 44007 Nantes, France
- IRS UN, University of Nantes, Nantes, France
- Nantes University Hospital, 44000 Nantes, France
| | - Amandine Hurbin
- IAB, University Grenoble Alpes, F-38000 Grenoble, France
- INSERM U823, IAB, F-38000 Grenoble, France
| | - Irene Boos
- Bioorganische Chemie, Gebäude NW1, Universität Bayreuth, 95440 Bayreuth, Germany
| | - Carlo Unverzagt
- Bioorganische Chemie, Gebäude NW1, Universität Bayreuth, 95440 Bayreuth, Germany
| | | | - Sylvie Lantuejoul
- IAB, University Grenoble Alpes, F-38000 Grenoble, France
- INSERM U823, IAB, F-38000 Grenoble, France
- Grenoble University Hospital, F-38000 Grenoble, France
| | - Jean-Luc Coll
- IAB, University Grenoble Alpes, F-38000 Grenoble, France
- INSERM U823, IAB, F-38000 Grenoble, France
| | - Annabelle Varrot
- CERMAV, UPR5301, CNRS, University Grenoble Alpes, 38041 Grenoble, France
| | | | - Benoit Busser
- IAB, University Grenoble Alpes, F-38000 Grenoble, France
- INSERM U823, IAB, F-38000 Grenoble, France
- Grenoble University Hospital, F-38000 Grenoble, France
- * E-mail: (JLP); (BB); (AI)
| | - Anne Imberty
- CERMAV, UPR5301, CNRS, University Grenoble Alpes, 38041 Grenoble, France
- * E-mail: (JLP); (BB); (AI)
| |
Collapse
|
295
|
Effect of sialylation on EGFR phosphorylation and resistance to tyrosine kinase inhibition. Proc Natl Acad Sci U S A 2015; 112:6955-60. [PMID: 25971727 DOI: 10.1073/pnas.1507329112] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is a heavily glycosylated transmembrane receptor tyrosine kinase. Upon EGF-binding, EGFR undergoes conformational changes to dimerize, resulting in kinase activation and autophosphorylation and downstream signaling. Tyrosine kinase inhibitors (TKIs) have been used to treat lung cancer by inhibiting EGFR phosphorylation. Previously, we demonstrated that EGFR sialylation suppresses its dimerization and phosphorylation. In this report, we further investigated the effect of sialylation on the phosphorylation profile of EGFR in TKI-sensitive and TKI-resistant cells. Sialylation was induced in cancer progression to inhibit the association of EGFR with EGF and the subsequent autophosphorylation. In the absence of EGF the TKI-resistant EGFR mutant (L858R/T790M) had a higher degree of sialylation and phosphorylation at Y1068, Y1086, and Y1173 than the TKI-sensitive EGFR. In addition, although sialylation in the TKI-resistant mutants suppresses EGFR tyrosine phosphorylation, with the most significant effect on the Y1173 site, the sialylation effect is not strong enough to stop cancer progression by inhibiting the phosphorylation of these three sites. These findings were supported further by the observation that the L858R/T790M EGFR mutant, when treated with sialidase or sialyltransferase inhibitor, showed an increase in tyrosine phosphorylation, and the sensitivity of the corresponding resistant lung cancer cells to gefitinib was reduced by desialylation and was enhanced by sialylation.
Collapse
|
296
|
Brühlmann D, Jordan M, Hemberger J, Sauer M, Stettler M, Broly H. Tailoring recombinant protein quality by rational media design. Biotechnol Prog 2015; 31:615-29. [DOI: 10.1002/btpr.2089] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/04/2015] [Indexed: 02/07/2023]
Affiliation(s)
- David Brühlmann
- Merck Serono SA, Corsier-sur-Vevey, Biotech Process Sciences, Zone Industrielle B; CH-1809 Fenil-sur-Corsier Switzerland
- Dept. of Biotechnology and Biophysics; Julius-Maximilians-Universität Würzburg, Biozentrum; Am Hubland DE-97074 Würzburg Germany
| | - Martin Jordan
- Merck Serono SA, Corsier-sur-Vevey, Biotech Process Sciences, Zone Industrielle B; CH-1809 Fenil-sur-Corsier Switzerland
| | - Jürgen Hemberger
- Inst. for Biochemical Engineering and Analytics; University of Applied Sciences Giessen; Wiesenstrasse 14, DE-35390 Giessen Germany
| | - Markus Sauer
- Dept. of Biotechnology and Biophysics; Julius-Maximilians-Universität Würzburg, Biozentrum; Am Hubland DE-97074 Würzburg Germany
| | - Matthieu Stettler
- Merck Serono SA, Corsier-sur-Vevey, Biotech Process Sciences, Zone Industrielle B; CH-1809 Fenil-sur-Corsier Switzerland
| | - Hervé Broly
- Merck Serono SA, Corsier-sur-Vevey, Biotech Process Sciences, Zone Industrielle B; CH-1809 Fenil-sur-Corsier Switzerland
| |
Collapse
|
297
|
van Wijk XM, Lawrence R, Thijssen VL, van den Broek SA, Troost R, van Scherpenzeel M, Naidu N, Oosterhof A, Griffioen AW, Lefeber DJ, van Delft FL, van Kuppevelt TH. A common sugar-nucleotide-mediated mechanism of inhibition of (glycosamino)glycan biosynthesis, as evidenced by 6F-GalNAc (Ac3). FASEB J 2015; 29:2993-3002. [PMID: 25868729 DOI: 10.1096/fj.14-264226] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/12/2015] [Indexed: 12/17/2022]
Abstract
Glycosaminoglycan (GAG) polysaccharides have been implicated in a variety of cellular processes, and alterations in their amount and structure have been associated with diseases such as cancer. In this study, we probed 11 sugar analogs for their capacity to interfere with GAG biosynthesis. One analog, with a modification not directly involved in the glycosidic bond formation, 6F-N-acetyl-d-galactosamine (GalNAc) (Ac3), was selected for further study on its metabolic and biologic effect. Treatment of human ovarian carcinoma cells with 50 μM 6F-GalNAc (Ac3) inhibited biosynthesis of GAGs (chondroitin/dermatan sulfate by ∼50-60%, heparan sulfate by ∼35%), N-acetyl-d-glucosamine (GlcNAc)/GalNAc containing glycans recognized by the lectins Datura stramonium and peanut agglutinin (by ∼74 and ∼43%, respectively), and O-GlcNAc protein modification. With respect to function, 6F-GalNAc (Ac3) treatment inhibited growth factor signaling and reduced in vivo angiogenesis by ∼33%. Although the analog was readily transformed in cells into the uridine 5'-diphosphate (UDP)-activated form, it was not incorporated into GAGs. Rather, it strongly reduced cellular UDP-GalNAc and UDP-GlcNAc pools. Together with data from the literature, these findings indicate that nucleotide sugar depletion without incorporation is a common mechanism of sugar analogs for inhibiting GAG/glycan biosynthesis.
Collapse
Affiliation(s)
- Xander M van Wijk
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Roger Lawrence
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Victor L Thijssen
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Sebastiaan A van den Broek
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Ran Troost
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Monique van Scherpenzeel
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Natasha Naidu
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Arie Oosterhof
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Dirk J Lefeber
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Floris L van Delft
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Toin H van Kuppevelt
- *Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, and Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, and Glycotechnology Core, University of California San Diego, San Diego, California, USA; and Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
298
|
Du J, Hong S, Dong L, Cheng B, Lin L, Zhao B, Chen YG, Chen X. Dynamic Sialylation in Transforming Growth Factor-β (TGF-β)-induced Epithelial to Mesenchymal Transition. J Biol Chem 2015; 290:12000-13. [PMID: 25809486 DOI: 10.1074/jbc.m115.636969] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Indexed: 12/16/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a fundamental process in embryonic development and organ formation. Aberrant regulation of EMT often leads to tumor progression. Changes in cell surface sialylation have recently been implicated in mediating EMT. Herein we report the visualization of dynamic changes of sialylation and glycoproteomic analysis of newly synthesized sialylated proteins in EMT by metabolic labeling of sialylated glycans with azides, followed by click labeling with fluorophores or affinity tags. We discovered that sialylation was down-regulated during EMT but then reverted and up-regulated in the mesenchymal state after EMT, accompanied by mRNA expression level changes of genes involved in the sialic acid biosynthesis. Quantitative proteomic analysis identified a list of sialylated proteins whose biosynthesis was dynamically regulated during EMT. Sialylation of cell surface adherent receptor integrin β4 was found to be down-regulated, which may regulate integrin functions during EMT. Furthermore, a global sialylation inhibitor was used to probe the functional role of sialylation during EMT. We found that inhibition of sialylation promoted EMT. Taken together, our findings suggest the important role of sialylation in regulating EMT and imply its possible function in related pathophysiological events, such as cancer metastasis.
Collapse
Affiliation(s)
- Jun Du
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China and
| | - Senlian Hong
- the Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of the Ministry of Education, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Lu Dong
- the Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of the Ministry of Education, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Bo Cheng
- the Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of the Ministry of Education, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Liang Lin
- the Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of the Ministry of Education, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Bing Zhao
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China and
| | - Ye-Guang Chen
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China and
| | - Xing Chen
- the Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of the Ministry of Education, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
299
|
Paleček E, Tkáč J, Bartošík M, Bertók T, Ostatná V, Paleček J. Electrochemistry of nonconjugated proteins and glycoproteins. Toward sensors for biomedicine and glycomics. Chem Rev 2015; 115:2045-108. [PMID: 25659975 PMCID: PMC4360380 DOI: 10.1021/cr500279h] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Emil Paleček
- Institute
of Biophysics Academy of Science of the Czech Republic, v.v.i., Královopolská
135, 612 65 Brno, Czech Republic
| | - Jan Tkáč
- Institute
of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Martin Bartošík
- Regional
Centre for Applied Molecular Oncology, Masaryk
Memorial Cancer Institute, Žlutý kopec 7, 656 53 Brno, Czech Republic
| | - Tomáš Bertók
- Institute
of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Veronika Ostatná
- Institute
of Biophysics Academy of Science of the Czech Republic, v.v.i., Královopolská
135, 612 65 Brno, Czech Republic
| | - Jan Paleček
- Central
European Institute of Technology, Masaryk
University, Kamenice
5, 625 00 Brno, Czech Republic
| |
Collapse
|
300
|
Wang YC, Lin V, Loring JF, Peterson SE. The 'sweet' spot of cellular pluripotency: protein glycosylation in human pluripotent stem cells and its applications in regenerative medicine. Expert Opin Biol Ther 2015; 15:679-87. [PMID: 25736263 DOI: 10.1517/14712598.2015.1021329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Human pluripotent stem cells (hPSCs) promise for the future of regenerative medicine. The structural and biochemical diversity associated with glycans makes them a unique type of macromolecule modification that is involved in the regulation of a vast array of biochemical events and cellular activities including pluripotency in hPSCs. The primary focus of this review article is to highlight recent advances in stem cell research from a glycobiological perspective. We also discuss how our understanding of glycans and glycosylation may help overcome barriers hindering the clinical application of hPSC-derived cells. AREAS COVERED A literature survey using NCBI-PubMed and Google Scholar was performed in 2014. EXPERT OPINION Regenerative medicine hopes to provide novel strategies to combat human disease and tissue injury that currently lack effective therapies. Although progress in this field is accelerating, many critical issues remain to be addressed in order for cell-based therapy to become a practical and safe treatment option. Emerging evidence suggests that protein glycosylation may significantly influence the regulation of cellular pluripotency, and that the exploitation of protein glycosylation in hPSCs and their differentiated derivatives may lead to transformative and translational discoveries for regenerative medicine. In addition, hPSCs represent a novel research platform for investigating glycosylation-related disease.
Collapse
Affiliation(s)
- Yu-Chieh Wang
- The University of North Texas Health Science Center, Department of Pharmaceutical Sciences , 3500 Camp Bowie Boulevard, RES-314G, Fort Worth, TX 76107 , USA +1 817 735 2944 ; +1 817 735 2603 ;
| | | | | | | |
Collapse
|