1
|
Wu X, Liu J, Yin X, Ma D, Zhang S, Liu X. Protein Fusion of Biosynthetic Enzymes and a Thermo-Responsive Polypeptide Expedites Facile Access to Biocatalysts for Nucleotide Sugars. Chembiochem 2025; 26:e202401005. [PMID: 39805738 DOI: 10.1002/cbic.202401005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/16/2025]
Abstract
Nucleotide sugars (NSs) are essential building blocks for the enzymatic assembly of glycans. In this study, we established a preparation and recycling avenue to the biocatalysts for the enzymatic synthesis of NSs. This approach involves fusing two enzymes into a bifunctional chimera and using elastin-like polypeptides (ET64) as a purification tag, which allows for easy recovery of these biocatalysts without the need for chromatography. We successfully constructed and obtained five bifunctional fusion enzymes (GalK-USP-ET64, GlmU-NahK-ET64, ManC-NahK-ET64, FKP-ET64, and NanA-CSS-ET64) for the synthesis of five common NSs (UDP-Gal, UDP-GlcNAc, GDP-Man, GDP-Fuc, and CMP-Neu5Ac). These enzymes were obtained using the Inverse Transition Cycling (ITC) process in yields ranging from 60 to 124 mg per liter of fermentation. The enzymatic synthesis of NSs was carried out on a scale from hundreds of milligrams to multiple grams using these biocatalysts. Furthermore, we investigated the reusability of these biocatalysts by recycling them from the reaction solution using the ITC process. The recycling of GalK-USP-ET64, GlmU-NahK-ET64, FKP-ET64, and NanA-CSS-ET64 was effectively achieved for 15, 13, 3, and 4 times, respectively. These biocatalysts could be used not only for the enzymatic synthesis of NSs but also for the chemoenzymatic synthesis of glycan biomolecules when coupled with glycosyltransferases.
Collapse
Affiliation(s)
- Xiaocong Wu
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Glycochemistry and Glycobiology, Shandong University, Qingdao, Shandong, 266237, P. R China
| | - Jing Liu
- School of Pharmacy, Jining Medical University, 669 Xueyuan Road, Rizhao, 276826, P. R. China
| | - Xuefei Yin
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Glycochemistry and Glycobiology, Shandong University, Qingdao, Shandong, 266237, P. R China
| | - Di Ma
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Glycochemistry and Glycobiology, Shandong University, Qingdao, Shandong, 266237, P. R China
| | - Sichao Zhang
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Glycochemistry and Glycobiology, Shandong University, Qingdao, Shandong, 266237, P. R China
| | - Xianwei Liu
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Glycochemistry and Glycobiology, Shandong University, Qingdao, Shandong, 266237, P. R China
| |
Collapse
|
2
|
Kim H, Lupoli TJ. Defined Glycan Ligands for Detecting Rare l-Sugar-Binding Proteins. J Am Chem Soc 2025; 147:11693-11699. [PMID: 40167164 PMCID: PMC11987014 DOI: 10.1021/jacs.5c03251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Most cells are decorated with distinct sugar sequences that can be recognized by carbohydrate-binding proteins (CBPs), such as antibodies and lectins. While humans utilize ten monosaccharide building blocks, bacteria biosynthesize hundreds of activated sugars to assemble diverse glycans. Monosaccharides absent in mammals are termed "rare" and are enriched in deoxy l-sugars beyond the "common" sugar l-fucose (l-Fuc) found across species. While immune proteins recognize microbial surfaces, there are limited probes to identify CBPs for the many rare sugars that may mediate these interactions. Here, we devise chemoenzymatic strategies to defined glycoconjugates containing l-Fuc and its structural analog l-colitose (l-Col), a bacterial dideoxysugar believed to bind immune proteins. We report a concise synthesis of l-Col and semisynthetic routes to several activated l-sugars. Incorporation of these sugars into glycans is evaluated using bacterial and mammalian glycosyltransferases (GTs) annotated to transfer l-Col or l-Fuc, respectively. We find that each GT can transfer both l-sugars, along with the rare hexose l-galactose (l-Gal), onto various glycan acceptors. Incorporation of these l-sugars into the resulting glycoconjugates is confirmed using known CBPs. Finally, these glycan ligands are employed to detect rare sugar-binding proteins in human serum. Overall, this work reveals similarities between bacterial and mammalian GTs that may be exploited for in vitro glycoconjugate construction to unveil novel mediators of host-pathogen interactions.
Collapse
Affiliation(s)
- Hanee Kim
- Department of Chemistry, New
York University, New York, New York 10003, United States
| | - Tania J. Lupoli
- Department of Chemistry, New
York University, New York, New York 10003, United States
| |
Collapse
|
3
|
Lin SW, Ko TP, Chiang HY, Wu CG, Hsu MF, Wang AHJ, Lin CH. Structural insight into the catalytic mechanism of the bifunctional enzyme l-fucokinase/GDP-fucose pyrophosphorylase. J Biol Chem 2025; 301:108344. [PMID: 39993526 PMCID: PMC11982464 DOI: 10.1016/j.jbc.2025.108344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
The bifunctional l-fucokinase/GDP-β-l-fucose pyrophosphorylase (FKP) from Bacteroides fragilis catalyzes the conversion from l-fucose to GDP-β-l-fucose. The reaction product, representing the activated form of l-fucose, is used by all l-fucosyltransferases to incorporate l-fucose. Herein, we report the first X-ray crystal structures of FKP in complex with substrate-product, leading to the dissection of both activity domains and corresponding catalytic mechanisms. The full-length FKP (FKP-FL, 949 amino acids) exists as a tetramer in solution, but the individually prepared N-terminal domain (FKP-NTD corresponding to the sequence 1-496, also containing a SUMO tag) and C-terminal domain (FKP-CTD, the sequence 519-949) form a monomer and a dimer, respectively. FKP-NTD has a single α/β domain and a β-helix-containing domain, whereas FKP-CTD folds into two α/β domains and the linker comprises three α-helices. The β-l-fucose-1-phosphate (fucose-1-P) and GTP bound separately to the active sites of fucokinase (located at FKP-CTD) and pyrophosphorylase (FKP-NTD), and a third nucleotide-binding site is adjacent to the β-helix (also in FKP-NTD). Furthermore, Asp762 was proposed to serve as the general base in the reaction of fucokinase, to deprotonate the C1-OH of fucose in the nucleophilic attack to γ-phosphate of ATP, resulting in the formation of fucose-1-P. At the same time, Arg592 and magnesium ion stabilize the developing negative charge in the leaving group (ADP). Subsequently, in the pyrophosphorylase-catalyzed reaction, the Lys187 side chain facilitates the nucleophilic attack of fucose-1-P toward GTP, leading to the formation of GDP-fucose.
Collapse
Affiliation(s)
- Sheng-Wei Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Tzu-Ping Ko
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hung-Yu Chiang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Cheng-Guo Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Min-Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Andrew H-J Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan; PhD. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Department of Chemistry, National Taiwan University, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
4
|
Woodward AM, Guindolet D, Martinez-Carrasco R, Gabison EE, Lavker RM, Argüeso P. Low fucosylation defines the glycocalyx of progenitor cells and melanocytes in the human limbal stem cell niche. Stem Cell Reports 2025; 20:102378. [PMID: 39706176 PMCID: PMC11784483 DOI: 10.1016/j.stemcr.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024] Open
Abstract
It is widely recognized that the glycocalyx has significant implications in regulating the self-renewal and differentiation of adult stem cells; however, its composition remains poorly understood. Here, we show that the fucose-binding Aleuria aurantia lectin (AAL) binds differentially to basal cells in the stratified epithelium of the human limbus, hair follicle epithelium, and meibomian gland duct. Using fluorescence-activated cell sorting in combination with single-cell transcriptomics, we find that most epithelial progenitor cells and melanocytes in the limbus display low AAL staining (AALlow) on their cell surface, an attribute that is gradually lost in epithelial cells as they differentiate into mature corneal cells. AALlow epithelial cells were enriched in putative limbal stem cell markers and displayed high clonogenic capacity. Further analyses revealed that AALlow epithelial cells had reduced expression of GDP-mannose-4,6-dehydratase, an enzyme catalyzing the first and limiting step in the de novo biosynthesis of GDP-fucose, and that inhibition of fucosylation using a small-molecule fucose analog stimulated the proliferative potential of limbal epithelial cells ex vivo. These results provide crucial insights into the distinctive composition of the glycocalyx in adult stem cells and underscore the significance of fucose modulation in the therapeutic regeneration of the human limbal stem cell niche.
Collapse
Affiliation(s)
- Ashley M Woodward
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA
| | - Damien Guindolet
- Fondation Ophtalmologique A. de Rothschild, 25 rue Manin, 75019 Paris, France
| | - Rafael Martinez-Carrasco
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA
| | - Eric E Gabison
- Fondation Ophtalmologique A. de Rothschild, 25 rue Manin, 75019 Paris, France
| | - Robert M Lavker
- Department of Dermatology, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Pablo Argüeso
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
5
|
Adak AK, Tseng H, Chang S, Chiang Y, Lyu K, Lee Y, Lu W, Kuo W, Angata T, Lin C. Comprehensive Modular Synthesis of Ganglioside Glycans and Evaluation of their Binding Affinities to Siglec-7 and Siglec-9. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412815. [PMID: 39555730 PMCID: PMC11727393 DOI: 10.1002/advs.202412815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Indexed: 11/19/2024]
Abstract
In the present work, bacterial glycosyltransferases are utilized to construct ganglioside glycans in a convergent approach via a sugar‒nucleotide regeneration system and one-pot multienzyme reactions. Starting from β-lactoside enables the diversification of both the glycan moieties and the linkages in the lower α-arm and upper β-arm. Overall, a comprehensive panel of 24 natural a-series (GM3, GM2, GM1a, GD1a, GT1a, and fucosyl-GM1), b-series (GD3, GD2, GD1b, GT1b, and GQ1b), c-series (GT3, GT2, GT1c, GQ1c, and GP1c), α-series (GM1α, GD1aα, and GT1aα), and o-series (GA2, GA1, GM1b, GalNAc-GM1b, and GD1c) ganglioside glycans are prepared, which are suitable for biological studies and further applications. Moreover, a microarray is constructed with these synthesized ganglioside glycans to investigate their binding specificity with recombinant Fc-fused Siglec-7 and Siglec-9, which are immune checkpoint-like glycan recognition proteins on natural killer cells. The microarray binding results reveal that GD3 and GT1aα are specific ligands for Siglec-7 and Siglec-9, respectively, and this discovery can lead to the identification of appropriate ligands for investigating the roles of these Siglecs in immunomodulation.
Collapse
Affiliation(s)
- Avijit K. Adak
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Hsin‐Kai Tseng
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Shu‐Yen Chang
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Yu‐Ching Chiang
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Ke‐Hong Lyu
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Yun‐Sheng Lee
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Wen Lu
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Wen‐Hua Kuo
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
| | - Takashi Angata
- Institute of Biological ChemistryAcademia SinicaTaipei11529Taiwan
| | - Chun‐Cheng Lin
- Department of ChemistryNational Tsing Hua University101 Section 2, Kuang Fu RoadHsinchu30013Taiwan
- Department of Medicinal and Applied ChemistryKaohsiung Medical UniversityKaohsiung80708Taiwan
| |
Collapse
|
6
|
Yoon MK, Shit P, Zhang W, Meredith RJ, Kang H, Carmichael I, Serianni AS. 4hJCHOCH Spin Coupling in a Lewis x Trisaccharide as Evidence of Inter-Residue C-H···O Hydrogen Bonding in Aqueous Solution. J Am Chem Soc 2024; 146:31264-31273. [PMID: 39470652 DOI: 10.1021/jacs.4c11948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Prior studies of the solution conformation of the Lewisx (Lex) trisaccharide, αFuc-(1→3)[βGal-(1→4)]-βGlcNAc, suggest that nonclassical inter-residue C-H···O hydrogen bonding in aqueous solution contributes to the stabilization of its 3D structure and affects its biological properties. Experimental evidence for this hydrogen bond in aqueous solution has been reported in the form of a 4hJCHOCH NMR spin-coupling constant between C5'Fuc and H1″Gal measured by 2D NMR methods in unlabeled samples. A methyl glycoside of Lex (MeβLex) was prepared containing selective 13C-labeling at C5'Fuc, and the H1″Gal signal was examined in high-field 1H NMR spectra for evidence of splitting or line-broadening caused by the 13C at C5'Fuc. High-resolution 1H NMR spectra obtained at high field and at different temperatures using different FID processing parameters showed no resolved splitting of the H1″Gal signal or evidence of line-broadening. Spectral simulation showed that this splitting and/or line-broadening would be observable if the reported J-value (∼1.1 Hz) is correct. DFT calculations on MeβLex and a carbon analog (O5″Gal replaced by a CH2 group) gave very small and nearly identical calculated 4hJC5',H1″ values, suggesting that the coupling is essentially zero. DFT calculations also showed that an alternate inter-residue 3hJH5',H1″ is small. Based on NMR analyses and DFT calculations, we found that 4hJC5',H1″ in MeβLex has an upper limit of ∼0.4 Hz and that the value could be lower, possibly zero, calling into question its value as experimental proof of persistent nonclassical hydrogen bonding in aqueous solutions of MeβLex and related structures.
Collapse
Affiliation(s)
- Mi-Kyung Yoon
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556-5670, United States
- Omicron Biochemicals, 115 South Hill Street, South Bend, Indiana 46617-2701, United States
| | - Pradip Shit
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556-5670, United States
| | - Wenhui Zhang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556-5670, United States
- Omicron Biochemicals, 115 South Hill Street, South Bend, Indiana 46617-2701, United States
| | - Reagan J Meredith
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556-5670, United States
- Texas Biomedical Research Institute, San Antonio, Texas 78245-0549, United States
| | - Hannah Kang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556-5670, United States
| | - Ian Carmichael
- Radiation Laboratory, University of Notre Dame, Notre Dame, Indiana 46556-5670, United States
| | - Anthony S Serianni
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556-5670, United States
| |
Collapse
|
7
|
Liu Y, Sweet IR, Boons GJ. 2,2-Difluoro Derivatives of Fucose Can Inhibit Cell Surface Fucosylation without Causing Slow Transfer to Acceptors. JACS AU 2024; 4:3953-3963. [PMID: 39483231 PMCID: PMC11522930 DOI: 10.1021/jacsau.4c00681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 11/03/2024]
Abstract
Fucosyl transferases (FUTs) are enzymes that transfer fucose (Fuc) from GDP-Fuc to acceptor substrates, resulting in fucosylated glycoconjugates that are involved in myriad physiological and disease processes. Previously, it has been shown that per-O-acetylated 2-F-Fuc can be taken up by cells and converted into GDP-2-F-Fuc, which is a competitive inhibitor of FUTs. Furthermore, it can act as a feedback inhibitor of de novo biosynthesis of GDP-Fuc resulting in reduced glycoconjugate fucosylation. However, GDP-2-F-Fuc and several other reported analogues are slow substrates, which can result in unintended incorporation of unnatural fucosides. Here, we describe the design, synthesis, and biological evaluation of GDP-2,2-di-F-Fuc and the corresponding prodrugs as an inhibitor of FUTs. This compound lacks the slow transfer activity observed for the monofluorinated counterpart. Furthermore, it was found that GDP-2-F-Fuc and GDP-2,2-di-F-Fuc have similar Ki values for the various human fucosyl transferases, while the corresponding phosphate prodrugs exhibit substantial differences in inhibition of cell surface fucosylation. Quantitative sugar nucleotide analysis by Liquid chromatography-mass spectrometry (LC-MS) indicates that the 2,2-di-F-Fuc prodrug has substantially greater feedback inhibitory activity. It was also found that by controlling the concentration of the inhibitor, varying degrees of inhibition of the biosynthesis of different types of fucosylated N-glycan structures can be achieved. These findings open new avenues for the modulation of fucosylation of cell surface glycoconjugates.
Collapse
Affiliation(s)
- Yanyan Liu
- Chemical
Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Igor R. Sweet
- Chemical
Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Geert-Jan Boons
- Chemical
Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
- Bijvoet
Center for Biomolecular Research, Utrecht
University, 3584 CG Utrecht, The Netherlands
- Chemistry
Department, The University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
8
|
Hollingsworth K, Di Maio A, Richards SJ, Vendeville JB, Wheatley DE, Council CE, Keenan T, Ledru H, Chidwick H, Huang K, Parmeggiani F, Marchesi A, Chai W, McBerney R, Kamiński TP, Balmforth MR, Tamasanu A, Finnigan JD, Young C, Warriner SL, Webb ME, Fascione MA, Flitsch S, Galan MC, Feizi T, Gibson MI, Liu Y, Turnbull WB, Linclau B. Synthesis and screening of a library of Lewis x deoxyfluoro-analogues reveals differential recognition by glycan-binding partners. Nat Commun 2024; 15:7925. [PMID: 39271664 PMCID: PMC11399408 DOI: 10.1038/s41467-024-51081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/30/2024] [Indexed: 09/15/2024] Open
Abstract
Glycan-mediated interactions play a crucial role in biology and medicine, influencing signalling, immune responses, and disease pathogenesis. However, the use of glycans in biosensing and diagnostics is limited by cross-reactivity, as certain glycan motifs can be recognised by multiple biologically distinct protein receptors. To address this specificity challenge, we report the enzymatic synthesis of a 150-member library of site-specifically fluorinated Lewisx analogues ('glycofluoroforms') using naturally occurring enzymes and fluorinated monosaccharides. Subsequent incorporation of a subset of these glycans into nanoparticles or a microarray revealed a striking spectrum of distinct binding intensities across different proteins that recognise Lewisx. Notably, we show that for two proteins with unique binding sites for Lewisx, glycofluoroforms exhibited enhanced binding to one protein, whilst reduced binding to the other, with selectivity governed by fluorination patterns. We finally showcase the potential diagnostic utility of this approach in glycofluoroform-mediated bacterial toxin detection by lateral flow.
Collapse
Affiliation(s)
- Kristian Hollingsworth
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Antonio Di Maio
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Sarah-Jane Richards
- Department of Chemistry, University of Warwick, Coventry, UK
- Manchester Institute of Biotechnology (MIB), Department of Chemistry, University of Manchester, Manchester, UK
| | | | - David E Wheatley
- School of Chemistry, University of Southampton, Highfield, Southampton, UK
| | - Claire E Council
- School of Chemistry, University of Southampton, Highfield, Southampton, UK
| | - Tessa Keenan
- Department of Chemistry, University of York, Heslington, York, UK
| | - Hélène Ledru
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, UK
| | - Harriet Chidwick
- Department of Chemistry, University of York, Heslington, York, UK
| | - Kun Huang
- Manchester Institute of Biotechnology (MIB), Department of Chemistry, University of Manchester, Manchester, UK
| | - Fabio Parmeggiani
- Manchester Institute of Biotechnology (MIB), Department of Chemistry, University of Manchester, Manchester, UK
| | - Andrea Marchesi
- Manchester Institute of Biotechnology (MIB), Department of Chemistry, University of Manchester, Manchester, UK
| | - Wengang Chai
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ryan McBerney
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Tomasz P Kamiński
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Matthew R Balmforth
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Alexandra Tamasanu
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - James D Finnigan
- Prozomix Limited, Haltwhistle Industrial Estate, Haltwhistle, Northumberland, UK
| | - Carl Young
- Prozomix Limited, Haltwhistle Industrial Estate, Haltwhistle, Northumberland, UK
| | - Stuart L Warriner
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Michael E Webb
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | | | - Sabine Flitsch
- Manchester Institute of Biotechnology (MIB), Department of Chemistry, University of Manchester, Manchester, UK
| | - M Carmen Galan
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, UK
| | - Ten Feizi
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Matthew I Gibson
- Department of Chemistry, University of Warwick, Coventry, UK.
- Manchester Institute of Biotechnology (MIB), Department of Chemistry, University of Manchester, Manchester, UK.
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK.
| | - Yan Liu
- Glycosciences Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| | - Bruno Linclau
- School of Chemistry, University of Southampton, Highfield, Southampton, UK.
- Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
9
|
Pan Y, Xue Q, Yang Y, Shi T, Wang H, Song X, Luo Y, Liu W, Ren S, Cai Y, Nie Y, Song Z, Liu B, Li JP, Wei J. Glycoengineering-based anti-PD-1-iRGD peptide conjugate boosts antitumor efficacy through T cell engagement. Cell Rep Med 2024; 5:101590. [PMID: 38843844 PMCID: PMC11228665 DOI: 10.1016/j.xcrm.2024.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024]
Abstract
Despite the important breakthroughs of immune checkpoint inhibitors in recent years, the objective response rates remain limited. Here, we synthesize programmed cell death protein-1 (PD-1) antibody-iRGD cyclic peptide conjugate (αPD-1-(iRGD)2) through glycoengineering methods. In addition to enhancing tissue penetration, αPD-1-(iRGD)2 simultaneously engages tumor cells and PD-1+ T cells via dual targeting, thus mediating tumor-specific T cell activation and proliferation with mild effects on non-specific T cells. In multiple syngeneic mouse models, αPD-1-(iRGD)2 effectively reduces tumor growth with satisfactory biosafety. Moreover, results of flow cytometry and single-cell RNA-seq reveal that αPD-1-(iRGD)2 remodels the tumor microenvironment and expands a population of "better effector" CD8+ tumor infiltrating T cells expressing stem- and memory-associated genes, including Tcf7, Il7r, Lef1, and Bach2. Conclusively, αPD-1-(iRGD)2 is a promising antibody conjugate therapeutic beyond antibody-drug conjugate for cancer immunotherapy.
Collapse
Affiliation(s)
- Yunfeng Pan
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qi Xue
- Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Yi Yang
- Glyco-therapy Biotechnology Co. Ltd., Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha Street, Qiantang District, Hangzhou, China
| | - Tao Shi
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hanbing Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xueru Song
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuting Luo
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenqi Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shiji Ren
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiran Cai
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yang Nie
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhentao Song
- Glyco-therapy Biotechnology Co. Ltd., Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha Street, Qiantang District, Hangzhou, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie P Li
- Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| | - Jia Wei
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China; Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| |
Collapse
|
10
|
Li Y, Li Y, Guo Y, Chen C, Yang L, Jiang Q, Ling P, Wang S, Li L, Fang J. Enzymatic modular synthesis of asymmetrically branched human milk oligosaccharides. Carbohydr Polym 2024; 333:121908. [PMID: 38494200 DOI: 10.1016/j.carbpol.2024.121908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/07/2024] [Accepted: 01/31/2024] [Indexed: 03/19/2024]
Abstract
Human milk oligosaccharides (HMOs) are intricate glycans that promote healthy growth of infants and have been incorporated into infant formula as food additives. Despite their importance, the limited availability of asymmetrically branched HMOs hinders the exploration of their structure and function relationships. Herein, we report an enzymatic modular strategy for the efficient synthesis of these HMOs. The key branching enzyme for the assembly of branched HMOs, human β1,6-N-acetylglucosaminyltransferase 2 (GCNT2), was successfully expressed in Pichia pastoris for the first time. Then, it was integrated with six other bacterial glycosyltransferases to establish seven glycosylation modules. Each module comprises a one-pot multi-enzyme (OPME) system for in-situ generation of costly sugar nucleotide donors, combined with a glycosyltransferase for specific glycosylation. This approach enabled the synthesis of 31 branched HMOs and 13 linear HMOs in a stepwise manner with well-programmed synthetic routes. The binding details of these HMOs with related glycan-binding proteins were subsequently elucidated using glycan microarray assays to provide insights into their biological functions. This comprehensive collection of synthetic HMOs not only serves as standards for HMOs structure identification in complex biological samples but also significantly enhances the fields of HMOs glycomics, opening new avenues for biomedical applications.
Collapse
Affiliation(s)
- Yinshuang Li
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Yi Li
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Yuxi Guo
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, United States of America
| | - Congcong Chen
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Lin Yang
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Qian Jiang
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Peixue Ling
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Shuaishuai Wang
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China.
| | - Lei Li
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30303, United States of America.
| | - Junqiang Fang
- National Glycoengineering Research Center and Shandong Key Laboratory of carbohydrate chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
11
|
Shi Y, Bashian EE, Hou Y, Wu P. Chemical immunology: Recent advances in tool development and applications. Cell Chem Biol 2024; 31:S2451-9456(24)00080-1. [PMID: 38508196 PMCID: PMC11393185 DOI: 10.1016/j.chembiol.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/01/2024] [Accepted: 02/22/2024] [Indexed: 03/22/2024]
Abstract
Immunology was one of the first biological fields to embrace chemical approaches. The development of new chemical approaches and techniques has provided immunologists with an impressive arsenal of tools to address challenges once considered insurmountable. This review focuses on advances at the interface of chemistry and immunobiology over the past two decades that have not only opened new avenues in basic immunological research, but also revolutionized drug development for the treatment of cancer and autoimmune diseases. These include chemical approaches to understand and manipulate antigen presentation and the T cell priming process, to facilitate immune cell trafficking and regulate immune cell functions, and therapeutic applications of chemical approaches to disease control and treatment.
Collapse
Affiliation(s)
- Yujie Shi
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Eleanor E Bashian
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Yingqin Hou
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Peng Wu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
12
|
Vos GM, Wu Y, van der Woude R, de Vries RP, Boons GJ. Chemo-Enzymatic Synthesis of Isomeric I-branched Polylactosamines Using Traceless Blocking Groups. Chemistry 2024; 30:e202302877. [PMID: 37909475 DOI: 10.1002/chem.202302877] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/03/2023]
Abstract
Poly-N-acetyl lactosamines (polyLacNAc) are common structural motifs of N- and O-linked glycan, glycosphingolipids and human milk oligosaccharides. They can be branched by the addition of β1,6-linked N-acetyl-glucosamine (GlcNAc) moieties to internal galactoside (Gal) residues by the I-branching enzyme beta-1,6-N-acetylglucosaminyltransferase 2 (GCNT2). I-branching has been implicated in many biological processes and is also associated with various diseases such as cancer progression. Currently, there is a lack of methods that can install, in a regioselective manner, I-branches and allows the preparation of isomeric poly-LacNAc derivatives. Here, we described a chemo-enzymatic strategy that addresses this deficiency and is based on the enzymatic assembly of an oligo-LacNAc chain that at specific positions is modified by a GlcNTFA moiety. Replacement of the trifluoroacetyl (TFA) moiety by tert-butyloxycarbonyl (Boc) gives compounds in which the galactoside at the proximal site is blocked from modification by GCNT2. After elaboration of the antennae, the Boc group can be removed, and the resulting amine acetylated to give natural I-branched structures. It is also shown that fucosides can function as a traceless blocking group that can provide complementary I-branched structures from a single precursor. The methodology made it possible to synthesize a library of polyLacNAc chains having various topologies.
Collapse
Affiliation(s)
- Gaёl M Vos
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, Netherlands
| | - Yunfei Wu
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, Netherlands
| | - Roosmarijn van der Woude
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, Netherlands
| | - Robert P de Vries
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, Netherlands
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, Netherlands
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA-30602, USA
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Chemistry Department, University of Georgia, Athens, GA-30602, USA
| |
Collapse
|
13
|
He M, Wang L, Yue Z, Feng C, Dai G, Jiang J, Huang H, Ji Q, Zhou M, Li D, Chai W. Development and validation of glycosyltransferase related-gene for the diagnosis and prognosis of head and neck squamous cell carcinoma. Aging (Albany NY) 2024; 16:1750-1766. [PMID: 38244579 PMCID: PMC10866440 DOI: 10.18632/aging.205455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/14/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a highly heterogeneous cancer characterized by difficulties in early diagnosis and outcome prediction. Aberrant glycosylated structures produced by the aberrant expression of glycosyltransferases are prevalent in HNSCC. In this study, we aim to construct glycosyltransferase-related gene signatures with diagnostic and prognostic value to better stratify patients with HNSCC and improve their diagnosis and prognosis. METHODS Bioinformatic tools were used to process data of patients with HNSCC from The Cancer Genome Atlas (TCGA) database. The prognostic model was formatted using univariate and multivariate Cox regression methods, while the diagnostic signature was constructed using support vector machine (SVM) and LASSO analysis. The results were verified using the Gene Expression Omnibus (GEO) cohort. The tumor microenvironment and benefits of immune checkpoint inhibitor (ICI) therapy in subgroups defined by glycosyltransferase-related genes were analyzed. Molecular biology experiments, including western blotting, cell counting kit (CCK)-8, colony formation, wound healing, and Transwell assays, were conducted to confirm the oncogenic function of beta-1,4-galactosyltransferase 3 (B4GALT3) in HNSCC. RESULTS We established a five-gene prognostic signature and a 15-gene diagnostic model. Based on the median risk score, patients with low risk had longer overall survival than those in the high-risk group, which was consistent with the results of the GEO cohort. The concrete results suggested that high-risk samples were related to a high tumor protein (TP)53 mutation rate, high infiltration of resting memory cluster of differentiation (CD)4 T cells, resting natural killer (NK) cells, and M0 macrophages, and benefited from ICI therapy. In contrast, the low-risk subgroup was associated with a low TP53 mutation rate; and high infiltration of naive B cells, plasma cells, CD8 T cells, and resting mast cells; and benefited less from ICI therapy. In addition, the diagnostic model had an area under curve (AUC) value of 0.997 and 0.978 in the training dataset and validation cohort, respectively, indicating the high diagnostic potential of the model. Ultimately, the depletion of B4GALT3 significantly hindered the proliferation, migration, and invasion of HNSCC cells. CONCLUSIONS We established two new biomarkers that could provide clinicians with diagnostic, prognostic, and treatment guidance for patients with HNSCC.
Collapse
Affiliation(s)
- Miao He
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Li Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Zihan Yue
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei, China
| | - Chunbo Feng
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Guosheng Dai
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Jinsong Jiang
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Hui Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Qingjun Ji
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Minglang Zhou
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Dapeng Li
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| | - Wei Chai
- Department of Otorhinolaryngology, Head and Neck Surgery, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
- Scientific Research and Experiment Center, The People’s Hospital of Bozhou, Bozhou 236000, Anhui, China
| |
Collapse
|
14
|
Huber N, Alcalá-Orozco EA, Rexer T, Reichl U, Klamt S. Model-based optimization of cell-free enzyme cascades exemplified for the production of GDP-fucose. Metab Eng 2023; 81:S1096-7176(23)00147-7. [PMID: 39492471 DOI: 10.1016/j.ymben.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/05/2024]
Abstract
Cell-free production systems are increasingly used for the synthesis of industrially relevant chemicals and biopharmaceuticals. Cell-free systems often utilize cell lysates, but biocatalytic cascades based on recombinant enzymes have emerged as a promising alternative strategy. However, implementing efficient enzyme cascades is a non-trivial task and mathematical modeling and optimization has become a key tool to improve their performance. In this work, we introduce a generic framework for the model-based optimization of cell-free enzyme cascades based on a given kinetic model of the system. We first formulate and systematize seven optimization problems relevant in the context of cell-free production processes including, for example, the maximization of productivity or product yield and the minimization of overall costs. We then present an approach that accounts for parameter uncertainties, not only during model calibration and model analysis but also when performing the actual optimization. After constructing a kinetic model of the enzyme cascade, experimental data are used to generate an ensemble of kinetic parameter sets reflecting their variabilities. For every parameter set, systems optimization is then performed and the resulting solution subsequently cross-validated for all other parameterizations to identify the solution with the highest overall performance under parameter uncertainty. We exemplify our approach for the cell-free synthesis of GDP-fucose, an important sugar nucleotide with various applications. We selected and solved three optimization problems based on a constructed dynamic model and validated two of them experimentally leading to significant improvements of the process (e.g., 50% increase of titer under identical total enzyme load). Overall, our results demonstrate the potential of model-driven optimization for the rational design and improvement of cell-free production systems. The developed approach for systems optimization under parameter uncertainty could also be relevant for the metabolic design of cell factories.
Collapse
Affiliation(s)
- Nicolas Huber
- Max Planck Institute for Dynamics of Complex Technical Systems, 39106, Magdeburg, Germany
| | | | - Thomas Rexer
- Max Planck Institute for Dynamics of Complex Technical Systems, 39106, Magdeburg, Germany; eversyn, 39106, Magdeburg, Germany
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, 39106, Magdeburg, Germany
| | - Steffen Klamt
- Max Planck Institute for Dynamics of Complex Technical Systems, 39106, Magdeburg, Germany.
| |
Collapse
|
15
|
Enzyme cascades for the synthesis of nucleotide sugars: Updates to recent production strategies. Carbohydr Res 2023; 523:108727. [PMID: 36521208 DOI: 10.1016/j.carres.2022.108727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
Nucleotide sugars play an elementary role in nature as building blocks of glycans, polysaccharides, and glycoconjugates used in the pharmaceutical, cosmetics, and food industries. As substrates of Leloir-glycosyltransferases, nucleotide sugars are essential for chemoenzymatic in vitro syntheses. However, high costs and the limited availability of nucleotide sugars prevent applications of biocatalytic cascades on a large industrial scale. Therefore, the focus is increasingly on nucleotide sugar synthesis strategies to make significant application processes feasible. The chemical synthesis of nucleotide sugars and their derivatives is well established, but the yields of these processes are usually low. Enzyme catalysis offers a suitable alternative here, and in the last 30 years, many synthesis routes for nucleotide sugars have been discovered and used for production. However, many of the published procedures shy away from assessing the practicability of their processes. With this review, we give an insight into the development of the (chemo)enzymatic nucleotide sugar synthesis pathways of the last years and present an assessment of critical process parameters such as total turnover number (TTN), space-time yield (STY), and enzyme loading.
Collapse
|
16
|
Liu W, Tang S, Peng J, Pan L, Wang J, Cheng H, Chen Z, Wang Y, Zhou H. Enhancing heterologous expression of a key enzyme for the biosynthesis of 2'-fucosyllactose. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:5162-5171. [PMID: 35289934 DOI: 10.1002/jsfa.11868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND 2'-Fucosyllactose (2'-FL) is the most abundant human milk oligosaccharide (HMO) in human milk and has important physiological functions. The market demand of 2'-FL is continuing to grow, but high production cost has limited its availability. To solve the dilemma, biosynthesis of 2'-FL has been proposed and is considered the most promising pathway for massive production. α-1,2-Fucosyltransferase is one of the key elements involved in its biosynthesis, but the limited intracellular accumulation and unstable properties of α-1,2-fucosyltransferases when expressed in host strains have become a major hurdle for the effective biosynthesis of 2'-FL. RESULTS A combinatorial engineering strategy of synergic modification of ribosome binding site, fusion peptide and enzyme gene was leveraged to enhance the soluble expression of α-1,2-fucosyltransferases and promote enzyme activity. The preferable combination was to employ an optimized ribosome binding site region to drive 3 × FLAG as a fusion partner along with the α-1,2-fucosyltransferase for expression in Escherichia coli (DE3) PlySs, and protein yield and enzyme activity were remarkably improved by 11.51-fold and 13.72-fold, respectively. CONCLUSION After finely tuning the synergy among different elements, the abundant protein yield and high enzyme activity confirmed that the drawbacks of heterologous expression in α-1,2-fucosyltransferase had been properly addressed. A suitable external environment further drives the efficient synthesis of α-1,2-fucosyltransferases. To our knowledge, this is the first report of a systematic and effective modification of α-1,2-fucosyltransferase expression, which could potentially serve as a guideline for industrial application. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wenxian Liu
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Shizhe Tang
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Jing Peng
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Lina Pan
- Ausnutria Dairy China Co. Ltd, Ausnutria Institute Food & Nutrition, Changsha, China
| | - Jiaqi Wang
- Ausnutria Dairy China Co. Ltd, Ausnutria Institute Food & Nutrition, Changsha, China
| | - Haina Cheng
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Zhu Chen
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Yuguang Wang
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Hongbo Zhou
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| |
Collapse
|
17
|
Zheng J, Xu H, Fang J, Zhang X. Enzymatic and chemoenzymatic synthesis of human milk oligosaccharides and derivatives. Carbohydr Polym 2022; 291:119564. [DOI: 10.1016/j.carbpol.2022.119564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 01/28/2023]
|
18
|
Huang K, Li C, Zong G, Prabhu SK, Chapla DG, Moremen KW, Wang LX. Site-selective sulfation of N-glycans by human GlcNAc-6-O-sulfotransferase 1 (CHST2) and chemoenzymatic synthesis of sulfated antibody glycoforms. Bioorg Chem 2022; 128:106070. [PMID: 35939855 DOI: 10.1016/j.bioorg.2022.106070] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 11/02/2022]
Abstract
Sulfation is a common modification of glycans and glycoproteins. Sulfated N-glycans have been identified in various glycoproteins and implicated for biological functions, but in vitro synthesis of structurally well-defined full length sulfated N-glycans remains to be described. We report here the first in vitro enzymatic sulfation of biantennary complex type N-glycans by recombinant human CHST2 (GlcNAc-6-O-sulfotransferase 1, GlcNAc6ST-1). We found that the sulfotransferase showed high antennary preference and could selectively sulfate the GlcNAc moiety located on the Manα1,3Man arm of the biantennary N-glycan. The glycan chain was further elongated by bacterial β1,4 galactosyltransferase from Neiserria meningitidis and human β1,4 galactosyltransferase IV(B4GALT4), which led to the formation of different sulfated N-glycans. Using rituximab as a model IgG antibody, we further demonstrated that the sulfated N-glycans could be efficiently transferred to an intact antibody by using a chemoenzymatic Fc glycan remodeling method, providing homogeneous sulfated glycoforms of antibodies. Preliminary binding analysis indicated that sulfation did not affect the apparent affinity of the antibody for FcγIIIa receptor.
Collapse
Affiliation(s)
- Kun Huang
- Department of Chemistry and Biochemistry, University of Maryland, 8051 Regents Drive, College Park, MD 20742, United States
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, 8051 Regents Drive, College Park, MD 20742, United States
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, 8051 Regents Drive, College Park, MD 20742, United States
| | - Sunaina Kiran Prabhu
- Department of Chemistry and Biochemistry, University of Maryland, 8051 Regents Drive, College Park, MD 20742, United States
| | - Digantkumar G Chapla
- Complex Carbohydrate Research Center, University of Georgia, Athens 30602, Georgia
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens 30602, Georgia
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, 8051 Regents Drive, College Park, MD 20742, United States.
| |
Collapse
|
19
|
Tong X, Ru Y, Fu J, Wang Y, Zhu J, Ding Y, Lv F, Yang M, Wei X, Liu C, Liu X, Lei L, Wu X, Guo L, Xu Y, Li J, Wu P, Gong H, Chen J, Wu D. Fucosylation Promotes Cytolytic Function and Accumulation of NK Cells in B Cell Lymphoma. Front Immunol 2022; 13:904693. [PMID: 35784355 PMCID: PMC9240281 DOI: 10.3389/fimmu.2022.904693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/11/2022] [Indexed: 12/17/2022] Open
Abstract
Natural killer (NK) cells have been demonstrated as a promising cellular therapy as they exert potent anti-tumor immune responses. However, applications of NK cells to tumor immunotherapy, especially in the treatment of advanced hematopoietic and solid malignancies, are still limited due to the compromised survival and short persistence of the transferred NK cells in vivo. Here, we observed that fucosyltransferase (FUT) 7 and 8 were highly expressed on NK cells, and the expression of CLA was positively correlated with the accumulation of NK cells in clinical B cell lymphoma development. Via enzyme-mediated ex vivo cell-surface fucosylation, the cytolytic effect of NK cells against B cell lymphoma was significantly augmented. Fucosylation also promoted NK cell accumulation in B cell lymphoma-targeted tissues by enhancing their binding to E-selectin. Moreover, fucosylation of NK cells also facilitated stronger T cell anti-tumor immune responses. These findings suggest that ex vivo fucosylation contributes to enhancing the effector functions of NK cells and may serve as a novel strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Xing Tong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuhua Ru
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jianhong Fu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Ying Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jinjin Zhu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Yiyang Ding
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Fulian Lv
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Menglu Yang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xiya Wei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Chenchen Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xin Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Lei Lei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xiaojin Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jie Li
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| | - Huanle Gong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| |
Collapse
|
20
|
Wang S, Zhang J, Wei F, Li W, Wen L. Facile Synthesis of Sugar Nucleotides from Common Sugars by the Cascade Conversion Strategy. J Am Chem Soc 2022; 144:9980-9989. [PMID: 35583341 DOI: 10.1021/jacs.2c03138] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sugar nucleotides are essential glycosylation donors in the carbohydrate metabolism. Naturally, most sugar nucleotides are derived from a limited number of common sugar nucleotides by de novo biosynthetic pathways, undergoing single or multiple reactions such as dehydration, epimerization, isomerization, oxidation, reduction, amination, and acetylation reactions. However, it is widely believed that such complex bioconversions are not practical for synthetic use due to the high preparation cost and great difficulties in product isolation. Therefore, most of the discovered sugar nucleotides are not readily available. Here, based on de novo biosynthesis mainly, 13 difficult-to-access sugar nucleotides were successfully prepared from two common sugars D-Man and sucrose in high yields, at a multigram scale, and without the need for tedious purification manipulations. This work demonstrated that de novo biosynthesis, although undergoing complex reactions, is also practical and cost-effective for synthetic use by employing a cascade conversion strategy.
Collapse
Affiliation(s)
- Shasha Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiang Su 210023, China
| | - Jiabin Zhang
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, China
| | - Fangyu Wei
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanjin Li
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liuqing Wen
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiang Su 210023, China
| |
Collapse
|
21
|
Frohnmeyer H, Rueben S, Elling L. Gram‐scale production of GDP‐β‐l‐fucose with multi‐enzyme cascades in a repetitive‐batch mode. ChemCatChem 2022. [DOI: 10.1002/cctc.202200443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Hannes Frohnmeyer
- RWTH Aachen University: Rheinisch-Westfalische Technische Hochschule Aachen Institute of Biotechnology and Helmholtz-Institute for Biomedical Engineering GERMANY
| | - Simon Rueben
- RWTH Aachen University: Rheinisch-Westfalische Technische Hochschule Aachen Institute of Biotechnology and Helmholtz-Institute for Biomedical Engineering GERMANY
| | - Lothar Elling
- RWTH Aachen University: Rheinisch-Westfalische Technische Hochschule Aachen Institute of Biotechnology and Helmholtz-Institute for Biomedical Engineering Pauwelsstr. 20 52074 Aachen GERMANY
| |
Collapse
|
22
|
Banahene N, Kavunja HW, Swarts BM. Chemical Reporters for Bacterial Glycans: Development and Applications. Chem Rev 2022; 122:3336-3413. [PMID: 34905344 PMCID: PMC8958928 DOI: 10.1021/acs.chemrev.1c00729] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bacteria possess an extraordinary repertoire of cell envelope glycans that have critical physiological functions. Pathogenic bacteria have glycans that are essential for growth and virulence but are absent from humans, making them high-priority targets for antibiotic, vaccine, and diagnostic development. The advent of metabolic labeling with bioorthogonal chemical reporters and small-molecule fluorescent reporters has enabled the investigation and targeting of specific bacterial glycans in their native environments. These tools have opened the door to imaging glycan dynamics, assaying and inhibiting glycan biosynthesis, profiling glycoproteins and glycan-binding proteins, and targeting pathogens with diagnostic and therapeutic payload. These capabilities have been wielded in diverse commensal and pathogenic Gram-positive, Gram-negative, and mycobacterial species─including within live host organisms. Here, we review the development and applications of chemical reporters for bacterial glycans, including peptidoglycan, lipopolysaccharide, glycoproteins, teichoic acids, and capsular polysaccharides, as well as mycobacterial glycans, including trehalose glycolipids and arabinan-containing glycoconjugates. We cover in detail how bacteria-targeting chemical reporters are designed, synthesized, and evaluated, how they operate from a mechanistic standpoint, and how this information informs their judicious and innovative application. We also provide a perspective on the current state and future directions of the field, underscoring the need for interdisciplinary teams to create novel tools and extend existing tools to support fundamental and translational research on bacterial glycans.
Collapse
Affiliation(s)
- Nicholas Banahene
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, United States
- Biochemistry, Cell, and Molecular Biology Program, Central Michigan University, Mount Pleasant, MI, United States
| | - Herbert W. Kavunja
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, United States
- Biochemistry, Cell, and Molecular Biology Program, Central Michigan University, Mount Pleasant, MI, United States
| | | |
Collapse
|
23
|
Heine V, Pelantová H, Bojarová P, Křen V, Elling L. Targeted fucosylation of glycans with engineered bacterial fucosyltransferase variants. ChemCatChem 2022. [DOI: 10.1002/cctc.202200037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Viktoria Heine
- Czech Academy of Sciences: Akademie ved Ceske republiky Institute of Microbiology CZECH REPUBLIC
| | - Helena Pelantová
- Czech Academy of Sciences: Akademie ved Ceske republiky Institute of Microbiology CZECH REPUBLIC
| | - Pavla Bojarová
- Czech Academy of Sciences: Akademie ved Ceske republiky Institute of Microbiology CZECH REPUBLIC
| | - Vladimír Křen
- Czech Academy of Sciences: Akademie ved Ceske republiky Institute of Microbiology CZECH REPUBLIC
| | - Lothar Elling
- RWTH Aachen University: Rheinisch-Westfalische Technische Hochschule Aachen Institute of Biotechnology and Helmholtz-Institute for Biomedical Engineering Pauwelsstr. 20 52074 Aachen GERMANY
| |
Collapse
|
24
|
Sugar nucleotide regeneration system for the synthesis of Bi- and triantennary N-glycans and exploring their activities against siglecs. Eur J Med Chem 2022; 232:114146. [DOI: 10.1016/j.ejmech.2022.114146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 11/18/2022]
|
25
|
Zheng M, Zheng M, Epstein S, Harnagel AP, Kim H, Lupoli TJ. Chemical Biology Tools for Modulating and Visualizing Gram-Negative Bacterial Surface Polysaccharides. ACS Chem Biol 2021; 16:1841-1865. [PMID: 34569792 DOI: 10.1021/acschembio.1c00341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Bacterial cells present a wide diversity of saccharides that decorate the cell surface and help mediate interactions with the environment. Many Gram-negative cells express O-antigens, which are long sugar polymers that makeup the distal portion of lipopolysaccharide (LPS) that constitutes the surface of the outer membrane. This review highlights chemical biology tools that have been developed in recent years to facilitate the modulation of O-antigen synthesis and composition, as well as related bacterial polysaccharide pathways, and the detection of unique glycan sequences. Advances in the biochemistry and structural biology of O-antigen biosynthetic machinery are also described, which provide guidance for the design of novel chemical and biomolecular probes. Many of the tools noted here have not yet been utilized in biological systems and offer researchers the opportunity to investigate the complex sugar architecture of Gram-negative cells.
Collapse
Affiliation(s)
- Meng Zheng
- Department of Chemistry, New York University, New York, 10003 New York, United States
| | - Maggie Zheng
- Department of Chemistry, New York University, New York, 10003 New York, United States
| | - Samuel Epstein
- Department of Chemistry, New York University, New York, 10003 New York, United States
| | - Alexa P. Harnagel
- Department of Chemistry, New York University, New York, 10003 New York, United States
| | - Hanee Kim
- Department of Chemistry, New York University, New York, 10003 New York, United States
| | - Tania J. Lupoli
- Department of Chemistry, New York University, New York, 10003 New York, United States
| |
Collapse
|
26
|
Council CE, Kilpin KJ, Gusthart JS, Allman SA, Linclau B, Lee SS. Enzymatic glycosylation involving fluorinated carbohydrates. Org Biomol Chem 2021; 18:3423-3451. [PMID: 32319497 DOI: 10.1039/d0ob00436g] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Fluorinated carbohydrates, where one (or more) fluorine atom(s) have been introduced into a carbohydrate structure, typically through deoxyfluorination chemistry, have a wide range of applications in the glycosciences. Fluorinated derivatives of galactose, glucose, N-acetylgalactosamine, N-acetylglucosamine, talose, fucose and sialic acid have been employed as either donor or acceptor substrates in glycosylation reactions. Fluorinated donors can be synthesised by synthetic methods or produced enzymatically from chemically fluorinated sugars. The latter process is mediated by enzymes such as kinases, phosphorylases and nucleotidyltransferases. Fluorinated donors produced by either method can subsequently be used in glycosylation reactions mediated by glycosyltransferases, or phosphorylases yielding fluorinated oligosaccharide or glycoconjugate products. Fluorinated acceptor substrates are typically synthesised chemically. Glycosyltransferases are most commonly used in conjunction with natural donors to further elaborate fluorinated acceptor substrates. Glycoside hydrolases are used with either fluorinated donors or acceptors. The activity of enzymes towards fluorinated sugars is often lower than towards the natural sugar substrates irrespective of donor or acceptor. This may be in part attributed to elimination of the contribution of the hydroxyl group to the binding of the substrate to enzymes. However, in many cases, enzymes still maintain a significant activity, and reactions may be optimised where necessary, enabling enzymes to be used more successfully in the production of fluorinated carbohydrates. This review describes the current state of the art regarding chemoenzymatic production of fluorinated carbohydrates, focusing specifically on examples of the enzymatic production of activated fluorinated donors and enzymatic glycosylation involving fluorinated sugars as either glycosyl donors or acceptors.
Collapse
Affiliation(s)
- Claire E Council
- School of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, UK.
| | | | | | | | | | | |
Collapse
|
27
|
Karimi Alavijeh M, Meyer AS, Gras SL, Kentish SE. Synthesis of N-Acetyllactosamine and N-Acetyllactosamine-Based Bioactives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7501-7525. [PMID: 34152750 DOI: 10.1021/acs.jafc.1c00384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
N-Acetyllactosamine (LacNAc) or more specifically β-d-galactopyranosyl-1,4-N-acetyl-d-glucosamine is a unique acyl-amino sugar and a key structural unit in human milk oligosaccharides, an antigen component of many glycoproteins, and an antiviral active component for the development of effective drugs against viruses. LacNAc is useful itself and as a basic building block for producing various bioactive oligosaccharides, notably because this synthesis may be used to add value to dairy lactose. Despite a significant amount of information in the literature on the benefits, structures, and types of different LacNAc-derived oligosaccharides, knowledge about their effective synthesis for large-scale production is still in its infancy. This work provides a comprehensive analysis of existing production strategies for LacNAc and important LacNAc-based structures, including sialylated LacNAc as well as poly- and oligo-LacNAc. We conclude that direct extraction from milk is too complex, while chemical synthesis is also impractical at an industrial scale. Microbial routes have application when multiple step reactions are needed, but the major route to large-scale biochemical production will likely lie with enzymatic routes, particularly those using β-galactosidases (for LacNAc synthesis), sialidases (for sialylated LacNAc synthesis), and β-N-acetylhexosaminidases (for oligo-LacNAc synthesis). Glycosyltransferases, especially for the biosynthesis of extended complex LacNAc structures, could also play a major role in the future. In these cases, immobilization of the enzyme can increase stability and reduce cost. Processing parameters, such as substrate concentration and purity, acceptor/donor ratio, water activity, and temperature, can affect product selectivity and yield. More work is needed to optimize these reaction parameters and in the development of robust, thermally stable enzymes to facilitate commercial production of these important bioactive substances.
Collapse
Affiliation(s)
- M Karimi Alavijeh
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - A S Meyer
- Protein Chemistry and Enzyme Technology Division, Department of Biotechnology and Biomedicine, Technical University of Denmark (DTU), DK-2800 Kongens Lyngby, Denmark
| | - S L Gras
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - S E Kentish
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
28
|
Xu Z, Deng Y, Zhang Z, Ma W, Li W, Wen L, Li T. Diversity-Oriented Chemoenzymatic Synthesis of Sulfated and Nonsulfated Core 2 O-GalNAc Glycans. J Org Chem 2021; 86:10819-10828. [PMID: 34254798 DOI: 10.1021/acs.joc.1c01115] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A diversity-oriented chemoenzymatic approach for the collective preparation of sulfated core 2 O-GalNAc glycans and their nonsulfated counterparts was described. A sulfated trisaccharide and a nonsulfated trisaccharide were chemically synthesized by combining flexible protected group manipulations and sequential one-pot glycosylations. The divergent enzymatic extension of these two trisaccharides, using a panel of robust glycosyltransferases that can recognize sulfated substrates and differentiating the branches with specifically designed glycosylation sequences to achieve regioselective sialylation, provided 36 structurally well-defined O-GalNAc glycans.
Collapse
Affiliation(s)
- Zhuojia Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaqi Deng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhumin Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wenjing Ma
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanjin Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liuqing Wen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tiehai Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300350, China
| |
Collapse
|
29
|
Dai Y, Hartke R, Li C, Yang Q, Liu JO, Wang LX. Synthetic Fluorinated l-Fucose Analogs Inhibit Proliferation of Cancer Cells and Primary Endothelial Cells. ACS Chem Biol 2020; 15:2662-2672. [PMID: 32930566 PMCID: PMC10901565 DOI: 10.1021/acschembio.0c00228] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Fucosylation is one of the most prevalent modifications on N- and O-glycans of glycoproteins, and it plays an important role in various cellular processes and diseases. Small molecule inhibitors of fucosylation have shown promise as therapeutic agents for sickle cell disease, arthritis, and cancer. We describe here the design and synthesis of a panel of fluorinated l-fucose analogs bearing fluorine atoms at the C2 and/or C6 positions of l-fucose as metabolic fucosylation inhibitors. Preliminary study of their effects on cell proliferation revealed that the 6,6-difluoro-l-fucose (3) and 6,6,6-trifluoro-l-fucose (6) showed significant inhibitory activity against proliferation of human colon cancer cells and human umbilical vein endothelial cells. In contrast, the previously reported 2-deoxy-2-fluoro-l-fucose (1) had no apparent effects on proliferations of all the cell lines tested. To understand the mechanism of cell proliferation inhibition by the fluorinated l-fucose analogs, we performed chemoenzymatic synthesis of the corresponding GDP-fluorinated l-fucose analogs and tested their inhibitory activities against the mammalian α1,6-fucosyltransferase (FUT8). Interestingly, the corresponding GDP derivatives of 6,6-difluoro-l-fucose (3) and 6,6,6-trifluoro-l-fucose (6), which are the stronger proliferation inhibitors, showed much weaker inhibitory activity against FUT8 than that of the 2-deoxy-2-fluoro-l-fucose (1). These results suggest that FUT8 is not the major target of the 6-fluorinated fucose analogs (3 and 6). Instead, other factors, such as the key enzymes involved in the de novo GDP-fucose biosynthetic pathway and/or other fucosyltransferases involved in the biosynthesis of tumor-associated glyco-epitopes are most likely the targets of the fluorinated l-fucose analogs to achieve cell proliferation inhibition. To our knowledge, this is the first comparative study of various fluorinated l-fucose analogs for suppressing the proliferation of human cancer and primary endothelial cells required for angiogenesis.
Collapse
Affiliation(s)
- Yuanwei Dai
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Ruth Hartke
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Qiang Yang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
30
|
Li C, Wu M, Gao X, Zhu Z, Li Y, Lu F, Qin HM. Efficient Biosynthesis of 2'-Fucosyllactose Using an In Vitro Multienzyme Cascade. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:10763-10771. [PMID: 32856455 DOI: 10.1021/acs.jafc.0c04221] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
2'-Fucosyllactose (2-FL) is a fucose-containing oligosaccharide that is found in humans and is believed to have potential nutraceutical and pharmaceutical uses. Here, a promising in vitro multienzyme cascade catalysis system (MECCS) was designed to convert L-fucose and lactose to 2-FL. The cascade comprises L-fucokinase/GDP-L-fucose phosphorylase (FKP), α-1,2-fucosyltransferase (FucT), and pyruvate kinase (PK). This MECCS was able to efficiently regenerate ATP or GTP with 5.67-fold improvement of GDP-L-fucose. To address the rate-limiting step in the MECCS, various FucT orthologues were screened, and HpFucT from Helicobacter pylori showed the highest catalytic efficiency, with a (kcat/KM) of 39.28 min-1 mM-1, while TeFucT from Thermosynechococcus elongatus showed the highest thermostability, with a melting temperature (Tm) of 48 °C. The dissociation constant (KD) of TeFucT (1.34 ± 0.41 μM) was 15-fold lower than that of HpFucT (20.24 ± 1.81 μM), suggesting that TeFucT had much higher affinity for GDP. Structural analysis of HpFucT indicated that Arg169 is part of a unique substrate-binding site that interacts with two oxygen atoms from the phosphate group of GDP-L-fucose. The 2-FL productivities of the MECCS in fed-batch reached 0.67 and 0.73 g/L/h with TeFucT and HpFucT, respectively. This research provides an alternative pathway for efficient production of 2-FL.
Collapse
Affiliation(s)
- Chao Li
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, P. R. China
| | - Mian Wu
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, P. R. China
| | - Xin Gao
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, P. R. China
| | - Zhangliang Zhu
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, P. R. China
| | - Yu Li
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, P. R. China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, P. R. China
| | - Hui-Min Qin
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science and Technology, National Engineering Laboratory for Industrial Enzymes, Tianjin 300457, P. R. China
| |
Collapse
|
31
|
Hong S, Feng L, Yang Y, Jiang H, Hou X, Guo P, Marlow FL, Stanley P, Wu P. In Situ Fucosylation of the Wnt Co-receptor LRP6 Increases Its Endocytosis and Reduces Wnt/β-Catenin Signaling. Cell Chem Biol 2020; 27:1140-1150.e4. [PMID: 32649905 DOI: 10.1016/j.chembiol.2020.06.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/14/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022]
Abstract
Wnt/β-catenin signaling regulates critical, context-dependent transcription in numerous physiological events. Among the well-documented mechanisms affecting Wnt/β-catenin activity, modification of N-glycans by L-fucose is the newest and the least understood. Using a combination of Chinese hamster ovary cell mutants with different fucosylation levels and cell-surface fucose editing (in situ fucosylation [ISF]), we report that α(1-3)-fucosylation of N-acetylglucosamine (GlcNAc) in the Galβ(1-4)-GlcNAc sequences of complex N-glycans modulates Wnt/β-catenin activity by regulating the endocytosis of low-density lipoprotein receptor-related protein 6 (LRP6). Pulse-chase experiments reveal that ISF elevates endocytosis of lipid-raft-localized LRP6, leading to the suppression of Wnt/β-catenin signaling. Remarkably, Wnt activity decreased by ISF is fully reversed by the exogenously added fucose. The combined data show that in situ cell-surface fucosylation can be exploited to regulate a specific signaling pathway via endocytosis promoted by a fucose-binding protein, thereby linking glycosylation of a receptor with its intracellular signaling.
Collapse
Affiliation(s)
- Senlian Hong
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla CA 92037, USA
| | - Lei Feng
- Department of Biochemistry, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Yi Yang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla CA 92037, USA
| | - Hao Jiang
- Department of Biochemistry, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; The School of Medicine and Pharmacy, Ocean University of China 5 Yushan Road, Qingdao 266003, China
| | - Xiaomeng Hou
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla CA 92037, USA
| | - Peng Guo
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Florence L Marlow
- Department of Cell Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pamela Stanley
- Department of Cell Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla CA 92037, USA.
| |
Collapse
|
32
|
Chen C, Wang S, Gadi MR, Zhu H, Liu F, Liu CC, Li L, Wang F, Ling P, Cao H. Enzymatic modular synthesis and microarray assay of poly-N-acetyllactosamine derivatives. Chem Commun (Camb) 2020; 56:7549-7552. [PMID: 32579622 DOI: 10.1039/d0cc03268a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A facile enzymatic modular assembly strategy for the preparative-scale synthesis of poly-N-acetyllactosamine (poly-LacNAc) glycans with varied lengths and designed sialylation and/or fucosylation patterns is described. These glycans were printed as a microarray to investigate their interactions with a panel of glycan binding proteins (GBPs). Binding affinities revealed that the avidity of GBPs could be largely affected by the length and the patterns of sialylation and fucosylation.
Collapse
Affiliation(s)
- Congcong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Tang F, Zhou M, Qin K, Shi W, Yashinov A, Yang Y, Yang L, Guan D, Zhao L, Tang Y, Chang Y, Zhao L, Yang H, Zhou H, Huang R, Huang W. Selective N-glycan editing on living cell surfaces to probe glycoconjugate function. Nat Chem Biol 2020; 16:766-775. [DOI: 10.1038/s41589-020-0551-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/20/2020] [Indexed: 12/31/2022]
|
34
|
Soroko M, Kwan DH. Enzymatic Synthesis of a Fluorogenic Reporter Substrate and the Development of a High-Throughput Assay for Fucosyltransferase VIII Provide a Toolkit to Probe and Inhibit Core Fucosylation. Biochemistry 2020; 59:2100-2110. [PMID: 32441090 DOI: 10.1021/acs.biochem.0c00286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Maxim Soroko
- Department of Chemistry and Biochemistry, Concordia University, 7141 Sherbrooke Street West, Montreal, Quebec, Canada H4B 1R6
| | - David H. Kwan
- Department of Chemistry and Biochemistry, Concordia University, 7141 Sherbrooke Street West, Montreal, Quebec, Canada H4B 1R6
- Department of Biology, Centre for Applied Synthetic Biology, and Centre for Structural and Functional Genomics, Concordia University, 7141 Sherbrooke Street West, Montreal, Quebec, Canada H4B 1R6
| |
Collapse
|
35
|
Liu Y, Hu H, Wang J, Zhou Q, Wu P, Yan N, Wang HW, Wu JW, Sun L. Cryo-EM structure of L-fucokinase/GDP-fucose pyrophosphorylase (FKP) in Bacteroides fragilis. Protein Cell 2020; 10:365-369. [PMID: 30242642 PMCID: PMC6468028 DOI: 10.1007/s13238-018-0576-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ying Liu
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Huifang Hu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jia Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qiang Zhou
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Nieng Yan
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China.,State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Hong-Wei Wang
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jia-Wei Wu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Linfeng Sun
- CAS Center for Excellence in Molecular Cell Science, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
36
|
Valverde P, Vendeville JB, Hollingsworth K, Mattey AP, Keenan T, Chidwick H, Ledru H, Huonnic K, Huang K, Light ME, Turner N, Jiménez-Barbero J, Galan MC, Fascione MA, Flitsch S, Turnbull WB, Linclau B. Chemoenzymatic synthesis of 3-deoxy-3-fluoro-l-fucose and its enzymatic incorporation into glycoconjugates. Chem Commun (Camb) 2020; 56:6408-6411. [DOI: 10.1039/d0cc02209h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A chemoenzymatic synthesis of 3-deoxy-3-fluoro-l-fucose, using a d- to l-sugar translation strategy, and its enzymatic activation and glycosylation, is reported.
Collapse
|
37
|
Gao T, Yan J, Liu CC, Palma AS, Guo Z, Xiao M, Chen X, Liang X, Chai W, Cao H. Chemoenzymatic Synthesis of O-Mannose Glycans Containing Sulfated or Nonsulfated HNK-1 Epitope. J Am Chem Soc 2019; 141:19351-19359. [PMID: 31738061 DOI: 10.1021/jacs.9b08964] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The human natural killer-1 (HNK-1) epitope is a unique sulfated trisaccharide sequence presented on O- and N-glycans of various glycoproteins and on glycolipids. It is overexpressed in the nervous system and plays crucial roles in nerve regeneration, synaptic plasticity, and neuronal diseases. However, the investigation of functional roles of HNK-1 in a more complex glycan context at the molecular level remains a big challenge due to lack of access to related structurally well-defined complex glycans. Herein, we describe a highly efficient chemoenzymatic approach for the first collective synthesis of HNK-1-bearing O-mannose glycans with different branching patterns, and for their nonsulfated counterparts. The successful strategy relies on both chemical glycosylation of a trisaccharide lactone donor for the introduction of sulfated HNK-1 branch and substrate promiscuities of bacterial glycosyltransferases that can tolerate sulfated substrates for enzymatic diversification. Glycan microarray analysis with the resulting complex synthetic glycans demonstrated their recognition by two HNK-1-specific antibodies including anti-HNK-1/N-CAM (CD57) and Cat-315, which provided further evidence for the recognition epitopes of these antibodies and the essential roles of the sulfate group for HNK-1 glycan-antibody recognition.
Collapse
Affiliation(s)
- Tian Gao
- National Glycoengineering Research Center, State Key Laboratory of Microbial Technology, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology , Shandong University , Qingdao 266237 , China.,Laboratory for Marine Drugs and Bioproducts , Pilot National Laboratory for Marine Science and Technology (Qingdao) , Qingdao 266237 , China
| | - Jingyu Yan
- Key Laboratory of Separation Science for Analytical Chemistry , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Chang-Cheng Liu
- National Glycoengineering Research Center, State Key Laboratory of Microbial Technology, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology , Shandong University , Qingdao 266237 , China.,Laboratory for Marine Drugs and Bioproducts , Pilot National Laboratory for Marine Science and Technology (Qingdao) , Qingdao 266237 , China
| | - Angelina S Palma
- UCIBIO, Department of Chemistry, Faculty of Science and Technology , NOVA University of Lisbon , Caparica 2829-516 , Portugal
| | - Zhimou Guo
- Key Laboratory of Separation Science for Analytical Chemistry , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Min Xiao
- National Glycoengineering Research Center, State Key Laboratory of Microbial Technology, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology , Shandong University , Qingdao 266237 , China
| | - Xi Chen
- Department of Chemistry , University of California , Davis , California 95616 , United States
| | - Xinmiao Liang
- Key Laboratory of Separation Science for Analytical Chemistry , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Wengang Chai
- The Glycosciences Laboratory, Faculty of Medicine , Imperial College London , London SW7 2AZ , United Kingdom
| | - Hongzhi Cao
- National Glycoengineering Research Center, State Key Laboratory of Microbial Technology, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology , Shandong University , Qingdao 266237 , China.,Laboratory for Marine Drugs and Bioproducts , Pilot National Laboratory for Marine Science and Technology (Qingdao) , Qingdao 266237 , China
| |
Collapse
|
38
|
Huang HH, Fang JL, Wang HK, Sun CY, Tsai TW, Huang YT, Kuo CY, Wang YJ, Liao CC, Yu CC. Substrate Characterization of Bacteroides fragilis α1,3/4-Fucosyltransferase Enabling Access to Programmable One-Pot Enzymatic Synthesis of KH-1 Antigen. ACS Catal 2019. [DOI: 10.1021/acscatal.9b04182] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Hsin-Hui Huang
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Jia-Lin Fang
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Hung-Kai Wang
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Chih-Yuan Sun
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Teng-Wei Tsai
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Yu-Ting Huang
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Cheng-Yu Kuo
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Yi-Jyun Wang
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| | - Chi-Chun Liao
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, 291 Zhongzheng Road, Zhonghe District, New Taipei City, 23561, Taiwan
| | - Ching-Ching Yu
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi 62102, Taiwan
| |
Collapse
|
39
|
Hevey R. Bioisosteres of Carbohydrate Functional Groups in Glycomimetic Design. Biomimetics (Basel) 2019; 4:E53. [PMID: 31357673 PMCID: PMC6784292 DOI: 10.3390/biomimetics4030053] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023] Open
Abstract
The aberrant presentation of carbohydrates has been linked to a number of diseases, such as cancer metastasis and immune dysregulation. These altered glycan structures represent a target for novel therapies by modulating their associated interactions with neighboring cells and molecules. Although these interactions are highly specific, native carbohydrates are characterized by very low affinities and inherently poor pharmacokinetic properties. Glycomimetic compounds, which mimic the structure and function of native glycans, have been successful in producing molecules with improved pharmacokinetic (PK) and pharmacodynamic (PD) features. Several strategies have been developed for glycomimetic design such as ligand pre-organization or reducing polar surface area. A related approach to developing glycomimetics relies on the bioisosteric replacement of carbohydrate functional groups. These changes can offer improvements to both binding affinity (e.g., reduced desolvation costs, enhanced metal chelation) and pharmacokinetic parameters (e.g., improved oral bioavailability). Several examples of bioisosteric modifications to carbohydrates have been reported; this review aims to consolidate them and presents different possibilities for enhancing core interactions in glycomimetics.
Collapse
Affiliation(s)
- Rachel Hevey
- Molecular Pharmacy, Department Pharmaceutical Sciences, University of Basel, Klingelbergstr. 50, 4056 Basel, Switzerland.
| |
Collapse
|
40
|
Wu HR, Anwar MT, Fan CY, Low PY, Angata T, Lin CC. Expedient assembly of Oligo-LacNAcs by a sugar nucleotide regeneration system: Finding the role of tandem LacNAc and sialic acid position towards siglec binding. Eur J Med Chem 2019; 180:627-636. [PMID: 31351394 DOI: 10.1016/j.ejmech.2019.07.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/26/2019] [Accepted: 07/15/2019] [Indexed: 11/28/2022]
Abstract
Sialosides containing (oligo-)N-acetyllactosamine (LacNAc, Galβ(1,4)GlcNAc) as core structure are known to serve as ligands for Siglecs. However, the role of tandem inner epitope for Siglec interaction has never been reported. Herein, we report the effect of internal glycan (by length and type) on the binding affinity and describe a simple and efficient chemo-enzymatic sugar nucleotide regeneration protocol for the preparative-scale synthesis of oligo-LacNAcs by the sequential use of β1,4-galactosyltransferase (β4GalT) and β1,3-N-acetylglucosyl transferase (β3GlcNAcT). Further modification of these oligo-LacNAcs was performed in one-pot enzymatic synthesis to yield sialylated and/or fucosylated analogs. A glycan library of 23 different sialosides containing various LacNAc lengths or Lac core with natural/unnatural sialylation and/or fucosylation was synthesized. These glycans were used to fabricate a glycan microarray that was utilized to screen glycan binding preferences against five different Siglecs (2, 7, 9, 14 and 15).
Collapse
Affiliation(s)
- Hsin-Ru Wu
- Department of Chemistry, National Tsing-Hua University, Hsinchu, 30013, Taiwan; Instrumentation Center of Ministry of Science and Technology at National Tsing-Hua University, Hsinchu, 30013, Taiwan
| | | | - Chen-Yo Fan
- Department of Chemistry, National Tsing-Hua University, Hsinchu, 30013, Taiwan
| | - Penk Yeir Low
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Takashi Angata
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan.
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing-Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
41
|
Gagarinov IA, Li T, Wei N, Sastre Toraño J, de Vries RP, Wolfert MA, Boons G. Protecting‐Group‐Controlled Enzymatic Glycosylation of Oligo‐
N
‐Acetyllactosamine Derivatives. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201903140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ivan A. Gagarinov
- Department of Chemical Biology and Drug DiscoveryUtrecht Institute for Pharmaceutical SciencesBijvoet Center for Biomolecular ResearchUtrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Tiehai Li
- Complex Carbohydrate Research CenterUniversity of Georgia 315 Riverbend Road Athens GA 30602 USA
| | - Na Wei
- Complex Carbohydrate Research CenterUniversity of Georgia 315 Riverbend Road Athens GA 30602 USA
| | - Javier Sastre Toraño
- Department of Chemical Biology and Drug DiscoveryUtrecht Institute for Pharmaceutical SciencesBijvoet Center for Biomolecular ResearchUtrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Robert P. de Vries
- Department of Chemical Biology and Drug DiscoveryUtrecht Institute for Pharmaceutical SciencesBijvoet Center for Biomolecular ResearchUtrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Margreet A. Wolfert
- Department of Chemical Biology and Drug DiscoveryUtrecht Institute for Pharmaceutical SciencesBijvoet Center for Biomolecular ResearchUtrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Complex Carbohydrate Research CenterUniversity of Georgia 315 Riverbend Road Athens GA 30602 USA
| | - Geert‐Jan Boons
- Department of Chemical Biology and Drug DiscoveryUtrecht Institute for Pharmaceutical SciencesBijvoet Center for Biomolecular ResearchUtrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Complex Carbohydrate Research CenterUniversity of Georgia 315 Riverbend Road Athens GA 30602 USA
- Department of ChemistryUniversity of Georgia Athens GA USA
| |
Collapse
|
42
|
Gagarinov IA, Li T, Wei N, Sastre Toraño J, de Vries RP, Wolfert MA, Boons GJ. Protecting-Group-Controlled Enzymatic Glycosylation of Oligo-N-Acetyllactosamine Derivatives. Angew Chem Int Ed Engl 2019; 58:10547-10552. [PMID: 31108002 DOI: 10.1002/anie.201903140] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/15/2019] [Indexed: 01/01/2023]
Abstract
We describe a chemoenzymatic strategy that can give a library of differentially fucosylated and sialylated oligosaccharides starting from a single chemically synthesized tri-N-acetyllactosamine derivative. The common precursor could easily be converted into 6 different hexasaccharides in which the glucosamine moieties are either acetylated (GlcNAc) or modified as a free amine (GlcNH2 ) or Boc (GlcNHBoc). Fucosylation of the resulting compounds by a recombinant fucosyl transferase resulted in only modification of the natural GlcNAc moieties, providing access to 6 selectively mono- and bis-fucosylated oligosaccharides. Conversion of the GlcNH2 or GlcNHBoc moieties into the natural GlcNAc, followed by sialylation by sialyl transferases gave 12 differently fucosylated and sialylated compounds. The oligosaccharides were printed as a microarray that was probed by several glycan-binding proteins, demonstrating that complex patterns of fucosylation can modulate glycan recognition.
Collapse
Affiliation(s)
- Ivan A Gagarinov
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, The Netherlands
| | - Tiehai Li
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Na Wei
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Javier Sastre Toraño
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, The Netherlands
| | - Robert P de Vries
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, The Netherlands
| | - Margreet A Wolfert
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, The Netherlands.,Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, The Netherlands.,Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA.,Department of Chemistry, University of Georgia, Athens, GA, USA
| |
Collapse
|
43
|
Bai J, Wu Z, Sugiarto G, Gadi MR, Yu H, Li Y, Xiao C, Ngo A, Zhao B, Chen X, Guan W. Biochemical characterization of Helicobacter pylori α1-3-fucosyltransferase and its application in the synthesis of fucosylated human milk oligosaccharides. Carbohydr Res 2019; 480:1-6. [PMID: 31132553 DOI: 10.1016/j.carres.2019.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
Fucosylated human milk oligosaccharides (HMOs) have important biological functions. Enzymatic synthesis of such compounds requires robust fucosyltransferases. A C-terminal 66-amino acid truncated version of Helicobacter pylori α1-3-fucosyltransferase (Hp3FT) is a good candidate. Hp3FT was biochemically characterized to identify optimal conditions for enzymatic synthesis of fucosides. While N-acetyllactosamine (LacNAc) and lactose were both suitable acceptors, the former is preferred. At a low guanosine 5'-diphospho-β-L-fucose (GDP-Fuc) to acceptor ratio, Hp3FT selectively fucosylated LacNAc. Based on these enzymatic characteristics, diverse fucosylated HMOs, including 3-fucosyllactose (3-FL), lacto-N-fucopentaose (LNFP) III, lacto-N-neofucopentaose (LNnFP) V, lacto-N-neodifucohexaose (LNnDFH) II, difuco- and trifuco-para-lacto-N-neohexaose (DF-paraLNnH and TF-para-LNnH), were synthesized enzymatically by varying the ratio of the donor and acceptor as well as controlling the order of multiple glycosyltransferase-catalyzed reactions.
Collapse
Affiliation(s)
- Jing Bai
- College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, 050024, China
| | - Zhigang Wu
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, 050018, China
| | - Go Sugiarto
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Madhusudhan Reddy Gadi
- Department of Chemistry, Center of Diagnostics & Therapeutics, Georgia State University, Atlanta, GA, 30303, USA
| | - Hai Yu
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Yanhong Li
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Cong Xiao
- Department of Chemistry, Center of Diagnostics & Therapeutics, Georgia State University, Atlanta, GA, 30303, USA
| | - Alice Ngo
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Baohua Zhao
- College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, 050024, China.
| | - Xi Chen
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA.
| | - Wanyi Guan
- College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, 050024, China.
| |
Collapse
|
44
|
Li J, Chen M, Liu Z, Zhang L, Felding BH, Moremen KW, Lauvau G, Abadier M, Ley K, Wu P. A Single-Step Chemoenzymatic Reaction for the Construction of Antibody-Cell Conjugates. ACS CENTRAL SCIENCE 2018; 4:1633-1641. [PMID: 30648147 PMCID: PMC6311947 DOI: 10.1021/acscentsci.8b00552] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Indexed: 05/02/2023]
Abstract
Employing live cells as therapeutics is a direction of future drug discovery. An easy and robust method to modify the surfaces of cells directly to incorporate novel functionalities is highly desirable. However, genetic methods for cell-surface engineering are laborious and limited by low efficiency for primary cell modification. Here we report a chemoenzymatic approach that exploits a fucosyltransferase to transfer bio-macromolecules, such as an IgG antibody (MW∼ 150 KD), to the glycocalyx on the surfaces of live cells when the antibody is conjugated to the enzyme's natural donor substrate GDP-Fucose. Requiring no genetic modification, this method is fast and biocompatible with little interference to cells' endogenous functions. We applied this method to construct two antibody-cell conjugates (ACCs) using both cell lines and primary cells, and the modified cells exhibited specific tumor targeting and resistance to inhibitory signals produced by tumor cells, respectively. Remarkably, Herceptin-NK-92MI conjugates, a natural killer cell line modified with Herceptin, exhibit enhanced activities to induce the lysis of HER2+ cancer cells both ex vivo and in a human tumor xenograft model. Given the unprecedented substrate tolerance of the fucosyltransferase, this chemoenzymatic method offers a general approach to engineer cells as research tools and for therapeutic applications.
Collapse
Affiliation(s)
- Jie Li
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Mingkuan Chen
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Zilei Liu
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Linda Zhang
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Brunie H. Felding
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Kelley W. Moremen
- Complex
Carbohydrate Research Center, University
of Georgia, Athens, Georgia 30602, United States
| | - Gregoire Lauvau
- Microbiology
and Immunology Department, Albert Einstein
College of Medicine, Bronx, New York 10461, United States
| | - Michael Abadier
- Division
of Inflammation Biology, La Jolla Institute
for Allergy and Immunology, La Jolla, California 92037, United States
| | - Klaus Ley
- Division
of Inflammation Biology, La Jolla Institute
for Allergy and Immunology, La Jolla, California 92037, United States
| | - Peng Wu
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| |
Collapse
|
45
|
Ahmadipour S, Beswick L, Miller GJ. Recent advances in the enzymatic synthesis of sugar-nucleotides using nucleotidylyltransferases and glycosyltransferases. Carbohydr Res 2018; 469:38-47. [PMID: 30265902 DOI: 10.1016/j.carres.2018.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 11/18/2022]
Affiliation(s)
- Sanaz Ahmadipour
- Lennard-Jones Laboratory, School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Laura Beswick
- Lennard-Jones Laboratory, School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Gavin J Miller
- Lennard-Jones Laboratory, School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| |
Collapse
|
46
|
Hunter CD, Guo T, Daskhan G, Richards MR, Cairo CW. Synthetic Strategies for Modified Glycosphingolipids and Their Design as Probes. Chem Rev 2018; 118:8188-8241. [DOI: 10.1021/acs.chemrev.8b00070] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Carmanah D. Hunter
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Tianlin Guo
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Gour Daskhan
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Michele R. Richards
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Christopher W. Cairo
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
47
|
Fang JL, Tsai TW, Liang CY, Li JY, Yu CC. Enzymatic Synthesis of Human Milk Fucosides α1,2-Fucosylpara-Lacto-N-Hexaose and its Isomeric Derivatives. Adv Synth Catal 2018. [DOI: 10.1002/adsc.201800518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jia-Lin Fang
- Department of Chemistry and Biochemistry; National Chung Cheng University; 168 University Road, Min-Hsiung Chiayi 62102 Taiwan
| | - Teng-Wei Tsai
- Department of Chemistry and Biochemistry; National Chung Cheng University; 168 University Road, Min-Hsiung Chiayi 62102 Taiwan
| | - Chin-Yu Liang
- Department of Chemistry and Biochemistry; National Chung Cheng University; 168 University Road, Min-Hsiung Chiayi 62102 Taiwan
| | - Jyun-Yi Li
- Department of Chemistry and Biochemistry; National Chung Cheng University; 168 University Road, Min-Hsiung Chiayi 62102 Taiwan
| | - Ching-Ching Yu
- Department of Chemistry and Biochemistry; National Chung Cheng University; 168 University Road, Min-Hsiung Chiayi 62102 Taiwan
| |
Collapse
|
48
|
Meng C, Sasmal A, Zhang Y, Gao T, Liu CC, Khan N, Varki A, Wang F, Cao H. Chemoenzymatic Assembly of Mammalian O-Mannose Glycans. Angew Chem Int Ed Engl 2018; 57:9003-9007. [PMID: 29802667 DOI: 10.1002/anie.201804373] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/04/2018] [Indexed: 12/27/2022]
Abstract
O-Mannose glycans account up to 30 % of total O-glycans in the brain. Previous synthesis and functional studies have only focused on the core M3 O-mannose glycans of α-dystroglycan, which are a causative factor for various muscular diseases. In this study, a highly efficient chemoenzymatic strategy was developed that enabled the first collective synthesis of 63 core M1 and core M2 O-mannose glycans. This chemoenzymatic strategy features the gram-scale chemical synthesis of five judiciously designed core structures, and the diversity-oriented modification of the core structures with three enzyme modules to provide 58 complex O-mannose glycans in a linear sequence that does not exceed four steps. The binding profiles of synthetic O-mannose glycans with a panel of lectins, antibodies, and brain proteins were also explored by using a printed O-mannose glycan array.
Collapse
Affiliation(s)
- Caicai Meng
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, 250012, China
| | - Aniruddha Sasmal
- Glycobiology Research and Training Center, University of California, San Diego, CA, 92093, USA
| | - Yan Zhang
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, 250012, China
| | - Tian Gao
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, 250012, China.,State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, China
| | - Chang-Cheng Liu
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, 250012, China.,State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, China
| | - Naazneen Khan
- Glycobiology Research and Training Center, University of California, San Diego, CA, 92093, USA
| | - Ajit Varki
- Glycobiology Research and Training Center, University of California, San Diego, CA, 92093, USA
| | - Fengshan Wang
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, 250012, China
| | - Hongzhi Cao
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, 250012, China.,State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, China
| |
Collapse
|
49
|
Meng C, Sasmal A, Zhang Y, Gao T, Liu CC, Khan N, Varki A, Wang F, Cao H. Chemoenzymatic Assembly of Mammalian O-Mannose Glycans. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201804373] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Caicai Meng
- National Glycoengineering Research Center; School of Pharmaceutical Science; Shandong University; Jinan 250012 China
| | - Aniruddha Sasmal
- Glycobiology Research and Training Center; University of California; San Diego CA 92093 USA
| | - Yan Zhang
- National Glycoengineering Research Center; School of Pharmaceutical Science; Shandong University; Jinan 250012 China
| | - Tian Gao
- National Glycoengineering Research Center; School of Pharmaceutical Science; Shandong University; Jinan 250012 China
- State Key Laboratory of Microbial Technology; Shandong University; Jinan 250100 China
| | - Chang-Cheng Liu
- National Glycoengineering Research Center; School of Pharmaceutical Science; Shandong University; Jinan 250012 China
- State Key Laboratory of Microbial Technology; Shandong University; Jinan 250100 China
| | - Naazneen Khan
- Glycobiology Research and Training Center; University of California; San Diego CA 92093 USA
| | - Ajit Varki
- Glycobiology Research and Training Center; University of California; San Diego CA 92093 USA
| | - Fengshan Wang
- National Glycoengineering Research Center; School of Pharmaceutical Science; Shandong University; Jinan 250012 China
| | - Hongzhi Cao
- National Glycoengineering Research Center; School of Pharmaceutical Science; Shandong University; Jinan 250012 China
- State Key Laboratory of Microbial Technology; Shandong University; Jinan 250100 China
| |
Collapse
|
50
|
Briard JG, Jiang H, Moremen KW, Macauley MS, Wu P. Cell-based glycan arrays for probing glycan-glycan binding protein interactions. Nat Commun 2018; 9:880. [PMID: 29491407 PMCID: PMC5830402 DOI: 10.1038/s41467-018-03245-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/30/2018] [Indexed: 12/31/2022] Open
Abstract
Glycan microarrays provide a high-throughput means of profiling the interactions of glycan-binding proteins with their ligands. However, the construction of current glycan microarray platforms is time consuming and expensive. Here, we report a fast and cost-effective method for the assembly of cell-based glycan arrays to probe glycan-glycan-binding protein interactions directly on the cell surface. Chinese hamster ovary cell mutants with a narrow and relatively homogeneous repertoire of glycoforms serve as the foundation platforms to develop these arrays. Using recombinant glycosyltransferases, sialic acid, fucose, and analogs thereof are installed on cell-surface glycans to form cell-based arrays displaying diverse glycan epitopes that can be probed with glycan-binding proteins by flow cytometry. Using this platform, high-affinity glycan ligands are discovered for Siglec-15-a sialic acid-binding lectin involved in osteoclast differentiation. Incubating human osteoprogenitor cells with cells displaying a high-affinity Siglec-15 ligand impairs osteoclast differentiation, demonstrating the utility of this cell-based glycan array technology.
Collapse
Affiliation(s)
- Jennie Grace Briard
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Hao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education and Qingdao National Laboratory for Marine Science & Technology and Shandong Provincial Key Laboratory of Glycoscience & Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 266003, Qingdao, China
| | - Kelley W Moremen
- Complex Carbohydrate Research Center and the Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Matthew Scott Macauley
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA. .,Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada.
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|