1
|
Mazéas L, Bouguerba-Collin A, Cock JM, Denoeud F, Godfroy O, Brillet-Guéguen L, Barbeyron T, Lipinska AP, Delage L, Corre E, Drula E, Henrissat B, Czjzek M, Terrapon N, Hervé C. Candidate genes involved in biosynthesis and degradation of the main extracellular matrix polysaccharides of brown algae and their probable evolutionary history. BMC Genomics 2024; 25:950. [PMID: 39390408 PMCID: PMC11468063 DOI: 10.1186/s12864-024-10811-3] [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: 06/07/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Brown algae belong to the Stramenopiles phylum and are phylogenetically distant from plants and other multicellular organisms. This independent evolutionary history has shaped brown algae with numerous metabolic characteristics specific to this group, including the synthesis of peculiar polysaccharides contained in their extracellular matrix (ECM). Alginates and fucose-containing sulphated polysaccharides (FCSPs), the latter including fucans, are the main components of ECMs. However, the metabolic pathways of these polysaccharides remain poorly described due to a lack of genomic data. RESULTS An extensive genomic dataset has been recently released for brown algae and their close sister species, for which we previously performed an expert annotation of key genes involved in ECM-carbohydrate metabolisms. Here we provide a deeper analysis of this set of genes using comparative genomics, phylogenetics analyses, and protein modelling. Two key gene families involved in both the synthesis and degradation of alginate were suggested to have been acquired by the common ancestor of brown algae and their closest sister species Schizocladia ischiensis. Our analysis indicates that this assumption can be extended to additional metabolic steps, and thus to the whole alginate metabolic pathway. The pathway for the biosynthesis of fucans still remains biochemically unresolved and we also investigate putative fucosyltransferase genes that may harbour a fucan synthase activity in brown algae. CONCLUSIONS Our analysis is the first extensive survey of carbohydrate-related enzymes in brown algae, and provides a valuable resource for future research into the glycome and ECM of brown algae. The expansion of specific families related to alginate metabolism may have represented an important prerequisite for the evolution of developmental complexity in brown algae. Our analysis questions the possible occurrence of FCSPs outside brown algae, notably within their closest sister taxon and in other Stramenopiles such as diatoms. Filling this knowledge gap in the future will help determine the origin and evolutionary history of fucan synthesis in eukaryotes.
Collapse
Affiliation(s)
- Lisa Mazéas
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
| | - Ahlem Bouguerba-Collin
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
| | - J Mark Cock
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
| | - France Denoeud
- Génomique Métabolique, Institut François Jacob, CEA, CNRS, Université Evry, Université Paris-Saclay, Genoscope, Evry, 91057, France
| | - Olivier Godfroy
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
| | - Loraine Brillet-Guéguen
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
- CNRS, Sorbonne Université, FR2424, ABiMS-IFB, Station Biologique, Roscoff, France
| | - Tristan Barbeyron
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
| | - Agnieszka P Lipinska
- Department of Algal Development and Evolution, Max Planck Institute for Biology, 72076, Tübingen, Germany
| | - Ludovic Delage
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
| | - Erwan Corre
- CNRS, Sorbonne Université, FR2424, ABiMS-IFB, Station Biologique, Roscoff, France
| | - Elodie Drula
- Aix Marseille Univ, CNRS, UMR 7257 AFMB, Marseille, France
- INRAE, USC 1408 AFMB, Marseille, France
- INRAE, Aix-Marseille Univ, UMR1163 BBF, Marseille, France
| | - Bernard Henrissat
- Aix Marseille Univ, CNRS, UMR 7257 AFMB, Marseille, France
- INRAE, USC 1408 AFMB, Marseille, France
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mirjam Czjzek
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France
| | - Nicolas Terrapon
- Aix Marseille Univ, CNRS, UMR 7257 AFMB, Marseille, France
- INRAE, USC 1408 AFMB, Marseille, France
| | - Cécile Hervé
- Integrative Biology of Marine Models Laboratory, Sorbonne Université, CNRS, Station Biologique de Roscoff, Roscoff, France.
| |
Collapse
|
2
|
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
|
3
|
Du Z, Zhu Y, Lu Z, Chen R, Huang Z, Chen Y, Guang C, Mu W. Combinatorial Optimization Strategies for 3-Fucosyllactose Hyperproduction in Escherichia coli. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14191-14198. [PMID: 38878091 DOI: 10.1021/acs.jafc.4c02950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
3-Fucosyllactose (3-FL), an important fucosylated human milk oligosaccharide in breast milk, offers numerous health benefits to infants. Previously, we metabolically engineered Escherichia coli BL21(DE3) for the in vivo biosynthesis of 3-FL. In this study, we initially optimized culture conditions to double 3-FL production. Competing pathway genes involved in in vivo guanosine 5'-diphosphate-fucose biosynthesis were subsequently inactivated to redirect fluxes toward 3-FL biosynthesis. Next, three promising transporters were evaluated using plasmid-based or chromosomally integrated expression to maximize extracellular 3-FL production. Additionally, through analysis of α1,3-fucosyltransferase (FutM2) structure, we identified Q126 residues as a highly mutable residue in the active site. After site-saturation mutation, the best-performing mutant, FutM2-Q126A, was obtained. Structural analysis and molecular dynamics simulations revealed that small residue replacement positively influenced helical structure generation. Finally, the best strain BD3-A produced 6.91 and 52.1 g/L of 3-FL in a shake-flask and fed-batch cultivations, respectively, highlighting its potential for large-scale industrial applications.
Collapse
Affiliation(s)
- Zhihui Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zhen Lu
- Bloomage Biotechnology Corp., Ltd., Jinan, Shandong 250010, People's Republic of China
| | - Roulin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zhaolin Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Cuie Guang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
4
|
Vannini A, Pinatel E, Costantini PE, Pelliciari S, Roncarati D, Puccio S, De Bellis G, Scarlato V, Peano C, Danielli A. (Re)-definition of the holo- and apo-Fur direct regulons of Helicobacter pylori. J Mol Biol 2024; 436:168573. [PMID: 38626867 DOI: 10.1016/j.jmb.2024.168573] [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: 12/13/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Iron homeostasis is a critical process for living organisms because this metal is an essential co-factor for fundamental biochemical activities, like energy production and detoxification, albeit its excess quickly leads to cell intoxication. The protein Fur (ferric uptake regulator) controls iron homeostasis in bacteria by switching from its apo- to holo-form as a function of the cytoplasmic level of ferrous ions, thereby modulating gene expression. The Helicobacter pylori HpFur protein has the rare ability to operate as a transcriptional commutator; apo- and holo-HpFur function as two different repressors with distinct DNA binding recognition properties for specific sets of target genes. Although the regulation of apo- and holo-HpFur in this bacterium has been extensively investigated, we propose a genome-wide redefinition of holo-HpFur direct regulon in H. pylori by integration of RNA-seq and ChIP-seq data, and a large extension of the apo-HpFur direct regulon. We show that in response to iron availability, new coding sequences, non-coding RNAs, toxin-antitoxin systems, and transcripts within open reading frames are directly regulated by apo- or holo-HpFur. These new targets and the more thorough validation and deeper characterization of those already known provide a complete and updated picture of the direct regulons of this two-faced transcriptional regulator.
Collapse
Affiliation(s)
- Andrea Vannini
- University of Bologna Department of Pharmacy and Biotechnology, Via Selmi 3, 40126 Bologna, Italy.
| | - Eva Pinatel
- Institute of Biomedical Technologies - National Research Council, Via Fratelli Cervi 93, 20054 Segrate (MI), Italy.
| | - Paolo Emidio Costantini
- University of Bologna Department of Pharmacy and Biotechnology, Via Selmi 3, 40126 Bologna, Italy.
| | - Simone Pelliciari
- Human Genetic Unit, Institute of Genetic and Cancer - University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| | - Davide Roncarati
- University of Bologna Department of Pharmacy and Biotechnology, Via Selmi 3, 40126 Bologna, Italy.
| | - Simone Puccio
- Institute of Genetics and Biomedical Research, UoS Milan - National Research Council, Via Manzoni 113, 20089 Rozzano (MI), Italy; Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (MI), Italy.
| | - Gianluca De Bellis
- Institute of Biomedical Technologies - National Research Council, Via Fratelli Cervi 93, 20054 Segrate (MI), Italy.
| | - Vincenzo Scarlato
- University of Bologna Department of Pharmacy and Biotechnology, Via Selmi 3, 40126 Bologna, Italy.
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, UoS Milan - National Research Council, Via Manzoni 113, 20089 Rozzano (MI), Italy; Human Technopole, Via Rita Levi Montalcini 1, 20157 Milan, Italy.
| | - Alberto Danielli
- University of Bologna Department of Pharmacy and Biotechnology, Via Selmi 3, 40126 Bologna, Italy.
| |
Collapse
|
5
|
Wang N, Zhu Y, Wang L, Huang Z, Li Z, Xu W, Mu W. Highly-efficient in vivo production of lacto-N-fucopentaose V by a regio-specific α1,3/4-fucosyltransferase from Bacteroides fragilis NCTC 9343. Int J Biol Macromol 2024; 266:130955. [PMID: 38499120 DOI: 10.1016/j.ijbiomac.2024.130955] [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: 10/14/2023] [Revised: 02/29/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Lacto-N-fucopentaose V (LNFP V) is a typical human milk pentasaccharide. Multi-enzymatic in vitro synthesis of LNFP V from lactose was reported, however, microbial cell factory approach to LNFP V production has not been reported yet. In this study, the biosynthetic pathway of LNFP V was examined in Escherichia coli. The previously constructed E. coli efficiently producing lacto-N-tetraose was used as the starting strain. GDP-fucose pathway module and a regio-specific glycosyltransferase with α1,3-fucosylation activity were introduced to realize the efficient synthesis of LNFP V. The α1,3/4-fucosyltransferase from Bacteroides fragilis was selected as the best enzyme for in vivo biosynthesis of LNFP V from nine candidates, with the highest titer and the lowest by-product accumulation. A beneficial variant K128D was obtained to further enhance LNFP V titer using computer-assisted site-directed mutagenesis. The final strain EW10 could produce 25.68 g/L LNFP V by fed-batch cultivation, with the productivity of 0.56 g/L·h.
Collapse
Affiliation(s)
- Ningning Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Liang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zhaolin Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zeyu Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wei Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China.
| |
Collapse
|
6
|
Okada T, Teramoto T, Ihara H, Ikeda Y, Kakuta Y. Crystal structure of mango α1,3/α1,4-fucosyltransferase elucidates unique elements that regulate Lewis A-dominant oligosaccharide assembly. Glycobiology 2024; 34:cwae015. [PMID: 38376259 DOI: 10.1093/glycob/cwae015] [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: 10/19/2023] [Revised: 01/28/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
In various organisms, α1,3/α1,4-fucosyltransferases (CAZy GT10 family enzymes) mediate the assembly of type I (Galβ1,3GlcNAc) and/or type II (Galβ1,4GlcNAc)-based Lewis structures that are widely distributed in glycoconjugates. Unlike enzymes of other species, plant orthologues show little fucosyltransferase activity for type II-based glycans and predominantly catalyze the assembly of the Lewis A structure [Galβ1,3(Fucα1,4)GlcNAc] on the type I disaccharide unit of their substrates. However, the structural basis underlying this unique substrate selectivity remains elusive. In this study, we investigated the structure-function relationship of MiFUT13A, a mango α1,3/α1,4-fucosyltransferase. The prepared MiFUT13A displayed distinct α1,4-fucosyltransferase activity. Consistent with the enzymatic properties of this molecule, X-ray crystallography revealed that this enzyme has a typical GT-B fold-type structure containing a set of residues that are responsible for its SN2-like catalysis. Site-directed mutagenesis and molecular docking analyses proposed a rational binding mechanism for type I oligosaccharides. Within the catalytic cleft, the pocket surrounding Trp121 serves as a binding site, anchoring the non-reducing terminal β1,3-galactose that belongs to the type I disaccharide unit. Furthermore, Glu177 was postulated to function as a general base catalyst through its interaction with the 4-hydroxy group of the acceptor N-acetylglucosamine residue. Adjacent residues, specifically Thr120, Thr157 and Asp175 were speculated to assist in binding of the reducing terminal residues. Intriguingly, these structural elements were not fully conserved in mammalian orthologue which also shows predominant α1,4-fucosyltransferase activity. In conclusion, we have proposed that MiFUT13A generates the Lewis A structure on type I glycans through a distinct mechanism, divergent from that of mammalian enzymes.
Collapse
Affiliation(s)
- Takahiro Okada
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Takamasa Teramoto
- Laboratory of Biophysical Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Hideyuki Ihara
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Yoshitaka Ikeda
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Yoshimitsu Kakuta
- Laboratory of Biophysical Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
7
|
Zhu Y, Chen R, Wang H, Chen Y, Huang Z, Du Z, Meng J, Zhou J, Mu W. De Novo Biosynthesis of Difucosyllactose by Artificial Pathway Construction and α1,3/4-Fucosyltransferase Rational Design in Escherichia coli. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38598361 DOI: 10.1021/acs.jafc.4c01691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Difucosyllactose (DFL) is a significant and plentiful oligosaccharide found in human breast milk. In this study, an artificial metabolic pathway of DFL was designed, focusing on the de novo biosynthesis of GDP-fucose from only glycerol. This was achieved by engineering Escherichia coli to endogenously overexpress genes manB, manC, gmd, and wcaG and heterologously overexpress a pair of fucosyltransferases to produce DFL from lactose. The introduction of α-1,2-fucosyltransferase from Helicobacter pylori (FucT2) along with α-1,3/4-fucosyltransferase (HP3/4FT) addressed rate-limiting challenges in enzymatic catalysis and allowed for highly efficient conversion of lactose into DFL. Based on these results, molecular modification of HP3/4FT was performed based on computer-assisted screening and structure-based rational design. The best-performing mutant, MH5, containing a combination of five mutated sites (F49K/Y131D/Y197N/E338D/R369A) of HP3/4FT was obtained. The best strain BLC09-58 harboring MH5 yielded 45.81 g/L of extracellular DFL in 5-L fed-batch cultures, which was the highest titer reported to date.
Collapse
Affiliation(s)
- Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Roulin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Hao Wang
- Bloomage Biotechnology Corp., Ltd., Jinan, Shandong 250010, People's Republic of China
| | - Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zhaolin Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zhihui Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Jiawei Meng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Jingwen Zhou
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
8
|
Xie Y, Wu X, Fu C, Duan H, Shi J, Blamey JM, Sun J. Rational Design of an α-1,3-Fucosyltransferase for the Biosynthesis of 3-Fucosyllactose in Bacillus subtilis ATCC 6051a via De Novo GDP-l-Fucose Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:1178-1189. [PMID: 38183288 DOI: 10.1021/acs.jafc.3c07604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2024]
Abstract
3-Fucosyllactose (3-FL) is an important oligosaccharide and nutrient in breast milk that can be synthesized in microbial cells by α-1,3-fucosyltransferase (α-1,3-FucT) using guanosine 5'-diphosphate (GDP)-l-fucose and lactose as substrates. However, the catalytic efficiency of known α-1,3-FucTs from various sources was limited due to their low solubility. To enhance the microbial production of 3-FL, the efficiencies of α-1,3-FucTs were evaluated and in Bacillus subtilis (B. subtilis) chassis cells that had been endowed with a heterologous synthetic pathway for GDP-l-fucose, revealing that the activity of FucTa from Helicobacter pylori (H. pylori) was higher than that of any of other reported homologues. To further improve the catalytic performance of FucTa, a rational design approach was employed, involving intracellular evaluation of the mutational sites of M32 obtained through directed evolution, analysis of the ligand binding site diversity, and protein structure simulation. Among the obtained variants, the FucTa-Y218 K variant exhibited the highest 3-FL yield, reaching 7.55 g/L in the shake flask growth experiment, which was 3.48-fold higher than that achieved by the wild-type enzyme. Subsequent fermentation optimization in a 5 L bioreactor resulted in a remarkable 3-FL production of 36.98 g/L, highlighting the great prospects of the designed enzyme and the strains for industrial applications.
Collapse
Affiliation(s)
- Yukang Xie
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinying Wu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Cong Fu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haiyan Duan
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Jiping Shi
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Jenny M Blamey
- Fundación Biociencia, José Domingo Cañas, 2280 Ñuñoa, Santiago, Chile
- Facultad de Química Y Biología, Universidad de Santiago de Chile, 3363 Alameda, Estación Central, Santiago, Chile
| | - Junsong Sun
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| |
Collapse
|
9
|
Yang Y, Song Z, Tian T, Zhao Z, Chen J, Hu J, Jiang X, Yang G, Xue Q, Zhao X, Sha W, Yang Y, Li JP. Trimming Crystallizable Fragment (Fc) Glycans Enables the Direct Enzymatic Transfer of Biomacromolecules to Antibodies as Therapeutics. Angew Chem Int Ed Engl 2023; 62:e202308174. [PMID: 37438983 DOI: 10.1002/anie.202308174] [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: 06/10/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/14/2023]
Abstract
Glycoengineering has provided powerful tools to construct site-specific antibody conjugates. However, only small-molecule payloads can be directly transferred to native or engineered antibodies using existing glycoengineering strategies. Herein, we demonstrate that reducing the complexity of crystallizable fragment (Fc) glycans could dramatically boost the chemoenzymatic modification of immunoglobulin G (IgG) via an engineered fucosyltransferase. In this platform, antibodies with Fc glycans engineered to a simple N-acetyllactosamine (LacNAc) disaccharide are successfully conjugated to biomacromolecules, such as oligonucleotides and nanobodies, in a single step within hours. Accordingly, we synthesized an antibody-conjugate-based anti-human epidermal growth factor receptor 2 (HER2)/ cluster of differentiation 3 (CD3) bispecific antibody and used it to selectively destroy patient-derived cancer organoids by reactivating endogenous T lymphocyte cells (T cells) inside the organoid. Our results highlight that this platform is a general approach to construct antibody-biomacromolecule conjugates with translational values.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Zhentao Song
- Glyco therapy Biotechnology Co., Ltd., 601/606 Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha street, Qiantang Distirct, Hangzhou, Zhejiang, 310058, China
| | - Tian Tian
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Zihan Zhao
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ji Chen
- Glyco therapy Biotechnology Co., Ltd., 601/606 Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha street, Qiantang Distirct, Hangzhou, Zhejiang, 310058, China
| | - Jiangping Hu
- Glyco therapy Biotechnology Co., Ltd., 601/606 Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha street, Qiantang Distirct, Hangzhou, Zhejiang, 310058, China
| | - Xin Jiang
- Glyco therapy Biotechnology Co., Ltd., 601/606 Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha street, Qiantang Distirct, Hangzhou, Zhejiang, 310058, China
| | - Guoli Yang
- Glyco therapy Biotechnology Co., Ltd., 601/606 Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha street, Qiantang Distirct, Hangzhou, Zhejiang, 310058, China
| | - Qi Xue
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Xinlu Zhao
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Wanxing Sha
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Yi Yang
- Glyco therapy Biotechnology Co., Ltd., 601/606 Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha street, Qiantang Distirct, Hangzhou, Zhejiang, 310058, China
| | - Jie P Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
10
|
Zhao M, Zhu Y, Wang H, Xu W, Zhang W, Mu W. An Overview of Sugar Nucleotide-Dependent Glycosyltransferases for Human Milk Oligosaccharide Synthesis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:12390-12402. [PMID: 37552889 DOI: 10.1021/acs.jafc.3c02895] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Human milk oligosaccharides (HMOs) have received increasing attention because of their special effects on infant health and commercial value as the new generation of core components in infant formula. Currently, large-scale production of HMOs is generally based on microbial synthesis using metabolically engineered cell factories. Introduction of the specific glycosyltransferases is essential for the construction of HMO-producing engineered strains in which the HMO-producing glycosyltransferases are generally sugar nucleotide-dependent. Four types of glycosyltransferases have been used for typical glycosylation reactions to synthesize HMOs. Soluble expression, substrate specificity, and regioselectivity are common concerns of these glycosyltransferases in practical applications. Screening of specific glycosyltransferases is an important research topic to solve these problems. Molecular modification has also been performed to enhance the catalytic activity of various HMO-producing glycosyltransferases and to improve the substrate specificity and regioselectivity. In this article, various sugar nucleotide-dependent glycosyltransferases for HMO synthesis were overviewed, common concerns of these glycosyltransferases were described, and the future perspectives of glycosyltransferase-related studies were provided.
Collapse
Affiliation(s)
- Mingli Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Hao Wang
- Bloomage Biotechnology Corp., Ltd., Jinan, Shandong 250010, People's Republic of China
| | - Wei Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
11
|
Kadirvelraj R, Boruah BM, Wang S, Chapla D, Huang C, Ramiah A, Hudson KL, Prudden AR, Boons GJ, Withers SG, Wood ZA, Moremen KW. Structural basis for Lewis antigen synthesis by the α1,3-fucosyltransferase FUT9. Nat Chem Biol 2023; 19:1022-1030. [PMID: 37202521 PMCID: PMC10726971 DOI: 10.1038/s41589-023-01345-y] [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: 07/26/2022] [Accepted: 04/20/2023] [Indexed: 05/20/2023]
Abstract
Mammalian cell surface and secreted glycoproteins exhibit remarkable glycan structural diversity that contributes to numerous physiological and pathogenic interactions. Terminal glycan structures include Lewis antigens synthesized by a collection of α1,3/4-fucosyltransferases (CAZy GT10 family). At present, the only available crystallographic structure of a GT10 member is that of the Helicobacter pylori α1,3-fucosyltransferase, but mammalian GT10 fucosyltransferases are distinct in sequence and substrate specificity compared with the bacterial enzyme. Here, we determined crystal structures of human FUT9, an α1,3-fucosyltransferase that generates Lewisx and Lewisy antigens, in complex with GDP, acceptor glycans, and as a FUT9-donor analog-acceptor Michaelis complex. The structures reveal substrate specificity determinants and allow prediction of a catalytic model supported by kinetic analyses of numerous active site mutants. Comparisons with other GT10 fucosyltransferases and GT-B fold glycosyltransferases provide evidence for modular evolution of donor- and acceptor-binding sites and specificity for Lewis antigen synthesis among mammalian GT10 fucosyltransferases.
Collapse
Affiliation(s)
- Renuka Kadirvelraj
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA, USA
| | - Bhargavi M Boruah
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Shuo Wang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Digantkumar Chapla
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Chin Huang
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Annapoorani Ramiah
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Kieran L Hudson
- Department of Biochemistry and Molecular Biology, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Stephen G Withers
- Department of Biochemistry and Molecular Biology, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zachary A Wood
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA, USA.
| | - Kelley W Moremen
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA, USA.
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| |
Collapse
|
12
|
Magdaleno JSL, Grewal RK, Medina-Franco JL, Oliva R, Shaikh AR, Cavallo L, Chawla M. Toward α-1,3/4 fucosyltransferases targeted drug discovery: In silico uncovering of promising natural inhibitors of fucosyltransferase 6. J Cell Biochem 2023; 124:1173-1185. [PMID: 37357420 DOI: 10.1002/jcb.30440] [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: 03/13/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023]
Abstract
Sialyl Lewis X (sLex ) antigen is a fucosylated cell-surface glycan that is normally involved in cell-cell interactions. The enhanced expression of sLex on cell surface glycans, which is attributed to the upregulation of fucosyltransferase 6 (FUT6), has been implicated in facilitating metastasis in human colorectal, lung, prostate, and oral cancers. The role that the upregulated FUT6 plays in the progression of tumor to malignancy, with reduced survival rates, makes it a potential target for anticancer drugs. Unfortunately, the lack of experimental structures for FUT6 has hampered the design and development of its inhibitors. In this study, we used in silico techniques to identify potential FUT6 inhibitors. We first modeled the three-dimensional structure of human FUT6 using AlphaFold. Then, we screened the natural compound libraries from the COCONUT database to sort out potential natural products (NPs) with best affinity toward the FUT6 model. As a result of these simulations, we identified three NPs for which we predicted binding affinities and interaction patterns quite similar to those we calculated for two experimentally tested FUT6 inhibitors, that is, fucose mimetic-1 and a GDP-triazole derived compound. We also performed molecular dynamics (MD) simulations for the FUT6 complexes with identified NPs, to investigate their stability. Analysis of the MD simulations showed that the identified NPs establish stable contacts with FUT6 under dynamics conditions. On these grounds, the three screened compounds appear as promising natural alternatives to experimentally tested FUT6 synthetic inhibitors, with expected comparable binding affinity. This envisages good prospects for future experimental validation toward FUT6 inhibition.
Collapse
Affiliation(s)
- Jorge Samuel Leon Magdaleno
- Department of Research and Innovation, STEMskills Research and Education Lab Private Limited, Faridabad, Haryana, India
| | - Ravneet K Grewal
- Department of Research and Innovation, STEMskills Research and Education Lab Private Limited, Faridabad, Haryana, India
| | - José L Medina-Franco
- Department of Pharmacy, DIFACQUIM Research Group, School of Chemistry, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Romina Oliva
- Department of Sciences and Technologies, University Parthenope of Naples, Naples, Italy
| | - Abdul Rajjak Shaikh
- Department of Research and Innovation, STEMskills Research and Education Lab Private Limited, Faridabad, Haryana, India
| | - Luigi Cavallo
- Physical Sciences and Engineering Division, Kaust Catalysis Center, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Mohit Chawla
- Physical Sciences and Engineering Division, Kaust Catalysis Center, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| |
Collapse
|
13
|
Yang L, Zhu Y, Meng J, Zhang W, Mu W. Recent progress in fucosylated derivatives of lacto- N-tetraose and lacto- N-neotetraose. Crit Rev Food Sci Nutr 2023; 64:10384-10396. [PMID: 37341681 DOI: 10.1080/10408398.2023.2224431] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Human milk oligosaccharides (HMOs) have attracted considerable attention owing to their unique physiological functions. Two important tetrasaccharides, lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT), are core structures of HMOs. Their safety has been evaluated and they can be added to infant formula as functional ingredients. The fucosylated derivatives of LNT and LNnT, mainly lacto-N-fucopentaose (LNFP) I, LNFP II, LNFP III, and lacto-N-difucohexaose I, exhibit prominent physiological characteristics, including modificating the intestinal microbiota, immunomodulation, anti-bacterial activities, and antiviral infection. However, they have received lesser attention than 2'-fucosyllactose. As precursors, LNT and LNnT are connected to one or two fucosyl units through α1,2/3/4 glycosidic bonds, forming a series of compounds with complex structures. These complex fucosylated oligosaccharides can be biologically synthesized using enzymatic and cell factory approaches. This review summarizes the occurrence, physiological effects, and biosynthesis of fucosylated LNT and LNnT derivatives and their future development.
Collapse
Affiliation(s)
- Longhao Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jiawei Meng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
14
|
Melamed J, Brockhausen I. Biosynthesis of the O antigen of pathogenic Escherichia coli O157:H7. Characterization of α1,4-Fuc-transferase WbdO. Glycobiology 2023; 33:165-175. [PMID: 36715215 DOI: 10.1093/glycob/cwac079] [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: 09/03/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 01/31/2023] Open
Abstract
The O157:H7 strain of Escherichia coli is responsible for frequent outbreaks of hemorrhagic colitis worldwide. Its lipopolysaccharide is a virulence factor and contains an O antigen having repeating units with the tetrasaccharide structure [2-D-PerNAcα1-3-L-Fucα1-4-D-Glcβ1-3-D-GalNAcα1-]n. Genes encoding glycosyltransferases WbdN, WbdO, and WbdP are responsible for the biosynthesis of this repeating unit. We have previously characterized the second enzyme in the pathway, WbdN, which transfers Glc in β1-3 linkage to GalNAcα-O-PO3-PO3-(CH2)11-O-Ph (GalNAc-PP-PhU). In this work, Fuc-transferase WbdO from E. coli O157:H7 expressed in BL21 bacteria was characterized using the product of WbdN as the acceptor substrate. We showed that WbdO is specific for GDP-β-L-Fuc as the donor substrate. Compounds that contained terminal Glc or Glcβ1-3GalNAc structures but lacked the diphosphate group did not serve as acceptor substrates. The structure of the WbdO product was identified by mass spectrometry and Nuclear magnetic resonance (NMR) as L-Fucα1-4-D-Glcβ1-3-D-GalNAc PP-PhU. WbdO is an unusual bivalent metal ion-dependent Fuc-transferase classified as an inverting GT2 family enzyme that has 2 conserved sequences near the N-terminus. The Asp37 residue within the 36VDGGSTD42 sequence was found to be essential for catalysis. Mutation of Asp68 to Ala within the conserved 67YDAMNK72 sequence resulted in a 3-fold increase in activity. These studies show that WbdOO157 is a highly specific Fuc-transferase with little homology to other characterized Fuc-transferases.
Collapse
Affiliation(s)
- Jacob Melamed
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L3N6, Canada
| | - Inka Brockhausen
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L3N6, Canada
| |
Collapse
|
15
|
O’Brien VP, Jackson LK, Frick JP, Rodriguez Martinez AE, Jones DS, Johnston CD, Salama NR. Helicobacter pylori Chronic Infection Selects for Effective Colonizers of Metaplastic Glands. mBio 2023; 14:e0311622. [PMID: 36598261 PMCID: PMC9973278 DOI: 10.1128/mbio.03116-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Chronic gastric infection with Helicobacter pylori can lead to progressive tissue changes that culminate in cancer, but how H. pylori adapts to the changing tissue environment during disease development is not fully understood. In a transgenic mouse gastric metaplasia model, we found that strains from unrelated individuals differed in their ability to infect the stomach, to colonize metaplastic glands, and to alter the expression of the metaplasia-associated protein TFF3. H. pylori isolates from different stages of disease from a single individual had differential ability to colonize healthy and metaplastic gastric glands. Exposure to the metaplastic environment selected for high gastric colonization by one of these strains. Complete genome sequencing revealed a unique alteration in the frequency of a variant allele of the putative adhesin sabB, arising from a recombination event with the related sialic acid binding adhesin (SabA) gene. Mutation of sabB in multiple H. pylori strain backgrounds strongly reduced adherence to both normal and metaplastic gastric tissue, and highly attenuated stomach colonization in mice. Thus, the changing gastric environment during disease development promotes bacterial adhesin gene variation associated with enhanced gastric colonization. IMPORTANCE Chronic infection with Helicobacter pylori is the primary risk factor for developing stomach cancer. As disease progresses H. pylori must adapt to a changing host tissue environment that includes induction of new cell fates in the cells that line the stomach. We tested representative H. pylori isolates collected from the same patient during early and later stages of disease in a mouse model where we can rapidly induce disease-associated tissue changes. Only the later-stage H. pylori strains could robustly colonize the diseased stomach environment. We also found that the ability to colonize the diseased stomach was associated with genetic variation in a putative cell surface adhesin gene called sabB. Additional experiments revealed that SabB promotes binding to stomach tissue and is critical for stomach colonization by the late-stage strains. Thus, H. pylori diversifies its genome during disease progression and these genomic changes highlight critical factors for bacterial persistence.
Collapse
Affiliation(s)
- V. P. O’Brien
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - L. K. Jackson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - J. P. Frick
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | | | - D. S. Jones
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - C. D. Johnston
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - N. R. Salama
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
16
|
Sugita T, Sampei S, Koketsu K. Efficient production of lacto-N-fucopentaose III in engineered Escherichia coli using α1,3-fucosyltransferase from Parabacteroides goldsteinii. J Biotechnol 2023; 361:110-118. [PMID: 36509384 DOI: 10.1016/j.jbiotec.2022.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/15/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Lacto-N-fucopentaose III (LNFP III) is a human milk oligosaccharide (HMO) with potential health benefits in infants, including in immune development and modulation of the intestinal environment. Low-cost fermentative production of various HMOs from lactose by engineered Escherichia coli has attracted attention, but few reports have investigated long-chain HMO production, such as of the pentasaccharide LNFP III. LNFP III is synthesized by α1,3-fucosyltransfer reaction to the glucosamine (GlcNAc) moiety in the lacto-N-neotetraose (LNnT) skeleton by α1,3-fucosyltransferase (α1,3-FucT). However, the known α1,3-FucTs also transfer fucose to the reducing terminal glucose moiety of LNnT or the starting material lactose, resulting in various byproducts. Here, we found a useful α1,3-FucT from Parabacteroides goldsteinii (PgsFucT), which is only reactive for GlcNAc in the N-acetyllactosamine (LacNAc) skeleton in vivo. On the basis of sequence alignment with a FucT of known structure, we also generated α1,3-FucT variants with altered reactivity for LacNAc or lactose. An E. coli strain heterologously expressing PgsFucT accumulated 3.84 g/L of LNFP III after 48 h of culture in a 3-L jar-fermenter. The amounts of various byproduct sugars were remarkably decreased compared with a strain expressing the previously characterized α1,3-fucT from Bacteroides fragilis.
Collapse
Affiliation(s)
- Tomotoshi Sugita
- Kirin Central Research Institute, Kirin Holdings Company, Limited, 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan.
| | - Sotaro Sampei
- Kirin Central Research Institute, Kirin Holdings Company, Limited, 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan.
| | - Kento Koketsu
- Kirin Central Research Institute, Kirin Holdings Company, Limited, 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
17
|
Lv Y, Zhang Z, Tian S, Wang W, Li H. Therapeutic potential of fucosyltransferases in cancer and recent development of targeted inhibitors. Drug Discov Today 2023; 28:103394. [PMID: 36223858 DOI: 10.1016/j.drudis.2022.103394] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/05/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
Fucosyltransferases (FUTs) have significant roles in various pathophysiological events. Their high expression is a signature of malignant cell transformation, contributing to many abnormal events during cancer development, such as uncontrolled cell proliferation, tumor cell invasion, angiogenesis, metastasis, immune evasion, and therapy resistance. Therefore, FUTs have evolved as an attractive therapeutic target for treating solid cancers, and many substrate analogs have been discovered with potential as FUT inhibitors for cancer therapy. Meanwhile, the development of FUT protein structures represents a significant advance in the design of FUT inhibitors with nonsubstrate structures. In this review, we summarize the role of FUTs in cancers, the resolved protein crystal structures and progress in the development of FUT inhibitors as cancer therapeutics.
Collapse
Affiliation(s)
- Yixin Lv
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhoudong Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, Jiangsu, China
| | - Sheng Tian
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, Jiangsu, China
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
18
|
Li Z, Zhu Y, Ni D, Zhang W, Mu W. Occurrence, functional properties, and preparation of 3-fucosyllactose, one of the smallest human milk oligosaccharides. Crit Rev Food Sci Nutr 2022; 63:9364-9378. [PMID: 35438024 DOI: 10.1080/10408398.2022.2064813] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human milk oligosaccharides (HMOs) are receiving wide interest and high attention due to their health benefits, especially for newborns. The HMOs-fortified products are expected to mimic human milk not only in the kinds of added oligosaccharides components but also the appropriate proportion between these components, and further provide the nutrition and physiological effects of human milk to newborns as closely as possible. In comparison to intensively studied 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL) has less attention in almost all respects. Nerveless, 3-FL naturally occurs in breast milk and increases roughly over the course of lactation with a nonnegligible content, and plays an irreplaceable role in human milk and delivers functional properties to newborns. According to the safety evaluation, 3-FL shows no acute oral toxicity, genetic toxicity, and subchronic toxicity. It has been approved as generally recognized as safe (GRAS). Biological production of 3-FL can be realized by enzymatic and cell factory approaches. The α1,3- or α1,3/4-fucosyltransferase is the key enzyme for 3-FL biosynthesis. Various metabolic engineering strategies have been applied to enhance 3-FL yield using cell factory approach. In conclusion, this review gives an overview of the recent scientific literatures regarding occurrence, bioactive properties, safety evaluation, and biotechnological preparation of 3-FL.
Collapse
Affiliation(s)
- Zeyu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Dawei Ni
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| |
Collapse
|
19
|
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
|
20
|
Tvaroška I. Glycosyltransferases as targets for therapeutic intervention in cancer and inflammation: molecular modeling insights. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-021-02026-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Fang W, Zhong K, Cheng J, Liu X, Liu C, Wang Z, Cao H. Capture‐Release
Strategy Facilitates Rapid Enzymatic Assembly of Oligosaccharides. CHINESE J CHEM 2021. [DOI: 10.1002/cjoc.202100744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Wenyuan Fang
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology Shandong University Qingdao Shandong 266237 China
| | - Kan Zhong
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology Shandong University Qingdao Shandong 266237 China
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy Ocean University of China Qingdao Shandong 266003 China
| | - Jiansong Cheng
- College of Pharmacy Nankai University Tianjin 300071 China
| | - Xian‐Wei Liu
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology Shandong University Qingdao Shandong 266237 China
| | - Chang‐Cheng Liu
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology Shandong University Qingdao Shandong 266237 China
| | - Zhongfu Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an Shaanxi 710069 China
| | - Hongzhi Cao
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology Shandong University Qingdao Shandong 266237 China
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy Ocean University of China Qingdao Shandong 266003 China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao Shandong 266237 China
| |
Collapse
|
22
|
Tomek MB, Janesch B, Braun ML, Taschner M, Figl R, Grünwald-Gruber C, Coyne MJ, Blaukopf M, Altmann F, Kosma P, Kählig H, Comstock LE, Schäffer C. A Combination of Structural, Genetic, Phenotypic and Enzymatic Analyses Reveals the Importance of a Predicted Fucosyltransferase to Protein O-Glycosylation in the Bacteroidetes. Biomolecules 2021; 11:1795. [PMID: 34944439 PMCID: PMC8698959 DOI: 10.3390/biom11121795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022] Open
Abstract
Diverse members of the Bacteroidetes phylum have general protein O-glycosylation systems that are essential for processes such as host colonization and pathogenesis. Here, we analyzed the function of a putative fucosyltransferase (FucT) family that is widely encoded in Bacteroidetes protein O-glycosylation genetic loci. We studied the FucT orthologs of three Bacteroidetes species-Tannerella forsythia, Bacteroides fragilis, and Pedobacter heparinus. To identify the linkage created by the FucT of B. fragilis, we elucidated the full structure of its nine-sugar O-glycan and found that l-fucose is linked β1,4 to glucose. Of the two fucose residues in the T. forsythia O-glycan, the fucose linked to the reducing-end galactose was shown by mutational analysis to be l-fucose. Despite the transfer of l-fucose to distinct hexose sugars in the B. fragilis and T. forsythia O-glycans, the FucT orthologs from B. fragilis, T. forsythia, and P. heparinus each cross-complement the B. fragilis ΔBF4306 and T. forsythia ΔTanf_01305 FucT mutants. In vitro enzymatic analyses showed relaxed acceptor specificity of the three enzymes, transferring l-fucose to various pNP-α-hexoses. Further, glycan structural analysis together with fucosidase assays indicated that the T. forsythia FucT links l-fucose α1,6 to galactose. Given the biological importance of fucosylated carbohydrates, these FucTs are promising candidates for synthetic glycobiology.
Collapse
Affiliation(s)
- Markus B. Tomek
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Bettina Janesch
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Matthias L. Braun
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Manfred Taschner
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Rudolf Figl
- Institute of Biochemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (R.F.); (C.G.-G.); (F.A.)
| | - Clemens Grünwald-Gruber
- Institute of Biochemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (R.F.); (C.G.-G.); (F.A.)
| | - Michael J. Coyne
- Department of Microbiology and the Duchossois Family Institute, University of Chicago, KCBD, 900 E. 57th Street, Chicago, IL 60637, USA; (M.J.C.); (L.E.C.)
| | - Markus Blaukopf
- Institute of Organic Chemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (M.B.); (P.K.)
| | - Friedrich Altmann
- Institute of Biochemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (R.F.); (C.G.-G.); (F.A.)
| | - Paul Kosma
- Institute of Organic Chemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (M.B.); (P.K.)
| | - Hanspeter Kählig
- Department of Organic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 38, A-1090 Vienna, Austria;
| | - Laurie E. Comstock
- Department of Microbiology and the Duchossois Family Institute, University of Chicago, KCBD, 900 E. 57th Street, Chicago, IL 60637, USA; (M.J.C.); (L.E.C.)
| | - Christina Schäffer
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| |
Collapse
|
23
|
Xia H, Ye J, Cao H, Liu X, Zhang Y, Liu CC. Enzymatic modular assembly of hybrid Lewis antigens. Org Biomol Chem 2021; 19:8041-8048. [PMID: 34473187 DOI: 10.1039/d1ob01579f] [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: 11/21/2022]
Abstract
The enzymatic synthesis of hybrid Lewis antigens including KH-1 (Lewis y-Lewis x-Lactose, Ley-Lex-Lac), Lewis a-Lewis x-Lactose (Lea-Lex-Lac), and Lewis b-Lewis x-Lactose (Leb-Lex-Lac) has been achieved using a facile enzymatic modular assembly strategy. Starting from a readily available tetrasaccharide, 3 complex hybrid Lewis antigens were achieved in over 40% total yields in less than 5 linear steps of sequential enzymatic glycosylation using 6 enzyme modules. The regio-selective fucosylation was achieved by simply controlling the donor-acceptor ratio. This strategy provides an easy access to these biologically important complex hybrid Lewis antigens at preparative scales.
Collapse
Affiliation(s)
- Hui Xia
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China.
| | - Jinfeng Ye
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Hongzhi Cao
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and 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
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xianwei Liu
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China.
| | - Yan Zhang
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan 250012, China.
| | - Chang-Cheng Liu
- National Glycoengineering Research Center, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, and 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
|
24
|
Mendoza F, Jaña GA. The inverting mechanism of the metal ion-independent LanGT2: the first step to understand the glycosylation of natural product antibiotic precursors through QM/MM simulations. Org Biomol Chem 2021; 19:5888-5898. [PMID: 34132308 DOI: 10.1039/d1ob00544h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Glycosyltransferases (GTs) from the GT1 family are responsible for the glycosylation of various important organic structures such as terpenes, steroids and peptide antibiotics, making it one of the most intensely studied families of GTs. The target of our study, LanGT2, is a member of the GT1 family that uses an inverting mechanism for transferring olivose from TDP-olivose, the donor substrate, to the natural product tetrangulol (Tet), the precursor of the antibiotic landomycin A. X-ray crystallography in conjunction with mutagenesis experiments has revealed the catalytic significance of 3 amino acids (Ser10, Ser219 and Asp137), suggesting Asp137 as the base catalyst. In the absence of X-ray structures that include the acceptor substrate Tet, in silico experiments and MD simulations that have modeled ternary complexes propose that Asp137 could recruit a water molecule to facilitate the nucleophilic activation of Tet, since the distance between Asp137 and the nucleophile is too long to directly deprotonate the nucleophilic moiety. So far, there is no computational evidence regarding the precise mechanism by which LanGT2 catalyzes the transfer of olivose, which raises questions such as: is a water-assisted mechanism possible? and how does this metal ion-independent GT stabilize the growing negative charge of the diphosphate leaving group? In this work, the QM/MM approach was used to unravel the catalytic mechanism of LanGT2, and to identify the role of crucial catalytic amino acids at a molecular level. Our calculations show that the minimum energy path (MEP) describes an SN2-like mechanism, identifying an oxocarbenium ion-like TS in which the olivosyl moiety adopts a 4H3 conformation. Interactions established between the diphosphate group of TDP and Ser10, Ser219, Arg220 and His283 are key to stabilize the development of charge on the leaving group. Our work also suggests that a water-mediated proton transfer mechanism is feasible, in which the water molecule is key to stabilize the phenolate ion-like nucleophile in the TS. This is the first computational insight into the inverting mechanism of an antibiotic natural product GT, and its implications may serve to guide the design of new biocatalysts for natural product glycodiversification.
Collapse
Affiliation(s)
- Fernanda Mendoza
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano, Chile.
| | - Gonzalo A Jaña
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano, Chile.
| |
Collapse
|
25
|
Zheng X, Zhu L, Li T, Xu W, Liu D, Sheng J, Cao H, Shi Y, Wang F. Improve Stability of Bioactive Peptides by Enzymatic Modular Synthesis of Peptides with O-Linked Sialyl Lewis x. ACS Catal 2021. [DOI: 10.1021/acscatal.1c00955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Xiaoju Zheng
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lin Zhu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tianlu Li
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Wenjia Xu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dongke Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Juzheng Sheng
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongzhi Cao
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Yikang Shi
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Laboratory of Carbohydrate Chemistry and Glycobiology, National Glycoengineering Research Center, Shandong University, Jinan 250012, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate Based Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
26
|
Martin KC, Tricomi J, Corzana F, García-García A, Ceballos-Laita L, Hicks T, Monaco S, Angulo J, Hurtado-Guerrero R, Richichi B, Sackstein R. Fucosyltransferase-specific inhibition via next generation of fucose mimetics. Chem Commun (Camb) 2021; 57:1145-1148. [PMID: 33411866 DOI: 10.1039/d0cc04847j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The ability to custom-modify cell surface glycans holds great promise for treatment of a variety of diseases. We propose a glycomimetic of l-fucose that markedly inhibits the creation of sLeX by FTVI and FTVII, but has no effect on creation of LeX by FTIX. Our findings thus indicate that selective suppression of sLex display can be achieved, and STD-NMR studies surprisingly reveal that the mimetic does not compete with GDP-fucose at the enzymatic binding site.
Collapse
Affiliation(s)
- Kyle C Martin
- Department of Translational Medicine, Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA. and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA and Program of Excellence in Glycoscience, Harvard Medical School, Boston, MA 02115, USA
| | - Jacopo Tricomi
- Department of Chemistry 'Ugo Schiff', University of Florence, Via della Lastruccia 13, 50019 Sesto Fiorentino, FI, Italy.
| | - Francisco Corzana
- Departamento de Química, Universidad de La Rioja, Centro de Investigación en Síntesis Química, 26006 Logroño, Spain
| | - Ana García-García
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I + D, Zaragoza, Spain
| | - Laura Ceballos-Laita
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I + D, Zaragoza, Spain
| | - Thomas Hicks
- School of Pharmacy, University of East Anglia, Norwich Research Park, NR47TJ, Norwich, UK
| | - Serena Monaco
- School of Pharmacy, University of East Anglia, Norwich Research Park, NR47TJ, Norwich, UK
| | - Jesus Angulo
- School of Pharmacy, University of East Anglia, Norwich Research Park, NR47TJ, Norwich, UK and Departamento de Química Orgánica, Universidad de Sevilla, C/Prof. García González, 1, 41012 Sevilla, Spain and Instituto de Investigaciones Químicas (CSIC-US), Avda. Américo Vespucio, 49, 41092 Sevilla, Spain
| | - Ramon Hurtado-Guerrero
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I + D, Zaragoza, Spain and Fundación ARAID, 50018, Zaragoza, Spain and Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, School of Dentistry, University of Copenhagen, Copenhagen, Denmark and Laboratorio de Microscopías Avanzada (LMA), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I + D, Zaragoza, Spain
| | - Barbara Richichi
- Department of Chemistry 'Ugo Schiff', University of Florence, Via della Lastruccia 13, 50019 Sesto Fiorentino, FI, Italy.
| | - Robert Sackstein
- Department of Translational Medicine, Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA. and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA and Program of Excellence in Glycoscience, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
27
|
Zhao L, Ma Z, Yin J, Shi G, Ding Z. Biological strategies for oligo/polysaccharide synthesis: biocatalyst and microbial cell factory. Carbohydr Polym 2021; 258:117695. [PMID: 33593568 DOI: 10.1016/j.carbpol.2021.117695] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/21/2022]
Abstract
Oligosaccharides and polysaccharides constitute the principal components of carbohydrates, which are important biomacromolecules that demonstrate considerable bioactivities. However, the variety and structural complexity of oligo/polysaccharides represent a major challenge for biological and structural explorations. To access structurally defined oligo/polysaccharides, biological strategies using glycoenzyme biocatalysts have shown remarkable synthetic potential attributed to their regioselectivity and stereoselectivity that allow mild, structurally controlled reaction without addition of protecting groups necessary in chemical strategies. This review summarizes recent biotechnological approaches of oligo/polysaccharide synthesis, which mainly includes in vitro enzymatic synthesis and cell factory synthesis. We have discussed the important factors involved in the production of nucleotide sugars. Furthermore, the strategies established in the cell factory and enzymatic syntheses are summarized, and we have highlighted concepts like metabolic flux rebuilding and regulation, enzyme engineering, and route design as important strategies. The research challenges and prospects are also outlined and discussed.
Collapse
Affiliation(s)
- Liting Zhao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, 214122, China.
| | - Zhongbao Ma
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, 214122, China.
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| | - Guiyang Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China.
| | - Zhongyang Ding
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
28
|
Chlorovirus PBCV-1 Multidomain Protein A111/114R Has Three Glycosyltransferase Functions Involved in the Synthesis of Atypical N-Glycans. Viruses 2021; 13:v13010087. [PMID: 33435207 PMCID: PMC7826918 DOI: 10.3390/v13010087] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/03/2021] [Accepted: 01/08/2021] [Indexed: 12/04/2022] Open
Abstract
The structures of the four N-linked glycans from the prototype chlorovirus PBCV-1 major capsid protein do not resemble any other glycans in the three domains of life. All known chloroviruses and antigenic variants (or mutants) share a unique conserved central glycan core consisting of five sugars, except for antigenic mutant virus P1L6, which has four of the five sugars. A combination of genetic and structural analyses indicates that the protein coded by PBCV-1 gene a111/114r, conserved in all chloroviruses, is a glycosyltransferase with three putative domains of approximately 300 amino acids each. Here, in addition to in silico sequence analysis and protein modeling, we measured the hydrolytic activity of protein A111/114R. The results suggest that domain 1 is a galactosyltransferase, domain 2 is a xylosyltransferase and domain 3 is a fucosyltransferase. Thus, A111/114R is the protein likely responsible for the attachment of three of the five conserved residues of the core region of this complex glycan, and, if biochemically corroborated, it would be the second three-domain protein coded by PBCV-1 that is involved in glycan synthesis. Importantly, these findings provide additional support that the chloroviruses do not use the canonical host endoplasmic reticulum–Golgi glycosylation pathway to glycosylate their glycoproteins; instead, they perform glycosylation independent of cellular organelles using virus-encoded enzymes.
Collapse
|
29
|
Boruah BM, Kadirvelraj R, Liu L, Ramiah A, Li C, Zong G, Bosman GP, Yang JY, Wang LX, Boons GJ, Wood ZA, Moremen KW. Characterizing human α-1,6-fucosyltransferase (FUT8) substrate specificity and structural similarities with related fucosyltransferases. J Biol Chem 2020; 295:17027-17045. [PMID: 33004438 PMCID: PMC7863877 DOI: 10.1074/jbc.ra120.014625] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Mammalian Asn-linked glycans are extensively processed as they transit the secretory pathway to generate diverse glycans on cell surface and secreted glycoproteins. Additional modification of the glycan core by α-1,6-fucose addition to the innermost GlcNAc residue (core fucosylation) is catalyzed by an α-1,6-fucosyltransferase (FUT8). The importance of core fucosylation can be seen in the complex pathological phenotypes of FUT8 null mice, which display defects in cellular signaling, development, and subsequent neonatal lethality. Elevated core fucosylation has also been identified in several human cancers. However, the structural basis for FUT8 substrate specificity remains unknown.Here, using various crystal structures of FUT8 in complex with a donor substrate analog, and with four distinct glycan acceptors, we identify the molecular basis for FUT8 specificity and activity. The ordering of three active site loops corresponds to an increased occupancy for bound GDP, suggesting an induced-fit folding of the donor-binding subsite. Structures of the various acceptor complexes were compared with kinetic data on FUT8 active site mutants and with specificity data from a library of glycan acceptors to reveal how binding site complementarity and steric hindrance can tune substrate affinity. The FUT8 structure was also compared with other known fucosyltransferases to identify conserved and divergent structural features for donor and acceptor recognition and catalysis. These data provide insights into the evolution of modular templates for donor and acceptor recognition among GT-B fold glycosyltransferases in the synthesis of diverse glycan structures in biological systems.
Collapse
Affiliation(s)
- Bhargavi M Boruah
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Renuka Kadirvelraj
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Annapoorani Ramiah
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Gerlof P Bosman
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA; Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands
| | - Zachary A Wood
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA.
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
30
|
Tvaroška I, Selvaraj C, Koča J. Selectins-The Two Dr. Jekyll and Mr. Hyde Faces of Adhesion Molecules-A Review. Molecules 2020; 25:molecules25122835. [PMID: 32575485 PMCID: PMC7355470 DOI: 10.3390/molecules25122835] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Selectins belong to a group of adhesion molecules that fulfill an essential role in immune and inflammatory responses and tissue healing. Selectins are glycoproteins that decode the information carried by glycan structures, and non-covalent interactions of selectins with these glycan structures mediate biological processes. The sialylated and fucosylated tetrasaccharide sLex is an essential glycan recognized by selectins. Several glycosyltransferases are responsible for the biosynthesis of the sLex tetrasaccharide. Selectins are involved in a sequence of interactions of circulated leukocytes with endothelial cells in the blood called the adhesion cascade. Recently, it has become evident that cancer cells utilize a similar adhesion cascade to promote metastases. However, like Dr. Jekyll and Mr. Hyde’s two faces, selectins also contribute to tissue destruction during some infections and inflammatory diseases. The most prominent function of selectins is associated with the initial stage of the leukocyte adhesion cascade, in which selectin binding enables tethering and rolling. The first adhesive event occurs through specific non-covalent interactions between selectins and their ligands, with glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains a principal strategy aimed at developing new therapies for the treatment of immune and inflammatory disorders and cancer. In this review, we will survey the significant contributions to and the current status of the understanding of the structure of selectins and the role of selectins in various biological processes. The potential of selectins and their ligands as therapeutic targets in chronic and acute inflammatory diseases and cancer will also be discussed. We will emphasize the structural characteristic of selectins and the catalytic mechanisms of glycosyltransferases involved in the biosynthesis of glycan recognition determinants. Furthermore, recent achievements in the synthesis of selectin inhibitors will be reviewed with a focus on the various strategies used for the development of glycosyltransferase inhibitors, including substrate analog inhibitors and transition state analog inhibitors, which are based on knowledge of the catalytic mechanism.
Collapse
Affiliation(s)
- Igor Tvaroška
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- Institute of Chemistry, Slovak Academy of Sciences, 84538 Bratislava, Slovak Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| | - Chandrabose Selvaraj
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
| | - Jaroslav Koča
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| |
Collapse
|
31
|
Park Y, Shin J, Yang J, Kim H, Jung Y, Oh H, Kim Y, Hwang J, Park M, Ban C, Jeong KJ, Kim SK, Kweon DH. Plasmid Display for Stabilization of Enzymes Inside the Cell to Improve Whole-Cell Biotransformation Efficiency. Front Bioeng Biotechnol 2020; 7:444. [PMID: 31998709 PMCID: PMC6967079 DOI: 10.3389/fbioe.2019.00444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/11/2019] [Indexed: 11/13/2022] Open
Abstract
Recombinant whole-cell biocatalysts are widely used for biotransformation of valuable products. However, some key enzymes involved in biotransformation processes are unstable and cannot be easily expressed in the functional form. In this study, we describe a versatile platform for enzyme stabilization inside the cell: Intracellularly Immobilized Enzyme System (IIES). A 1,2-fucosyltransferase from Pedobactor saltans (PsFL) and a 1,3-fucosyltransferase from Helicobacter pylori (HpFL), chosen as model proteins, were fused with Oct-1 DNA-binding domain, which mediated the formation of a plasmid-protein complex. Oct-1 fusion enabled both soluble and stable expression of recombinant proteins in the cytoplasm because the fusion proteins were stabilized on the plasmid like immobilized enzymes bound to solid surface. As a result, Oct-1-fusion proteins exhibited significantly greater product titer and yield than non-fusion proteins. Use of fusion proteins PsFL-Oct-1 with C-terminal Oct-1 and Oct-1-PsFL with N-terminal Oct-1 resulted in ~3- and ~2-fold higher 2'-fucosyllactose titers, respectively, than with the use of PsFL alone. When Oct-1 was fused to HpFL, which requires dimerization through heptad repeats, almost two times more 3-fucosyllactose was produced. Fucosyllactose has been used as a food additive because it has various beneficial effects on human health. We anticipate that IIES using Oct-1 fusion protein developed in this study can be applied to stabilize other unstable enzymes.
Collapse
Affiliation(s)
- Yunjeong Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Jonghyeok Shin
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Jinkyeong Yang
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Hooyeon Kim
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Younghun Jung
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Hyunseok Oh
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Yongjoon Kim
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Jaehyeon Hwang
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Myeongseo Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea
| | - Choongjin Ban
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea.,Institute of Biomolecule Control, Sungkyunkwan University, Suwon-si, South Korea
| | - Ki Jun Jeong
- Department of Chemical and Biomolecular Engineering, KAIST, Daejeon, South Korea
| | - Sun-Ki Kim
- Department of Food Science and Technology, Chung-Ang University, Anseong, South Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon-si, South Korea.,Institute of Biomolecule Control, Sungkyunkwan University, Suwon-si, South Korea.,Institute of Biologics, Sungkyunkwan University, Suwon-si, South Korea
| |
Collapse
|
32
|
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: 4.0] [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
|
33
|
Weinborn V, Li Y, Shah IM, Yu H, Dallas DC, German JB, Mills DA, Chen X, Barile D. Production of functional mimics of human milk oligosaccharides by enzymatic glycosylation of bovine milk oligosaccharides. Int Dairy J 2019; 102. [PMID: 32089591 DOI: 10.1016/j.idairyj.2019.104583] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Consumption of mothers' milk is associated with reduced incidence and severity of enteric infections, leading to reduced morbidity in breastfed infants. Fucosylated and sialylated human milk oligosaccharides (HMO) are important for both direct antimicrobial action - likely via a decoy effect - and indirect antimicrobial action through commensal growth enhancement. Bovine milk oligosaccharides (BMO) are a potential source of HMO-mimics as BMO resemble HMO; however, they have simpler and less fucosylated structures. BMO isolated at large scales from bovine whey permeate were modified by the addition of fucose and/or sialic acid to generate HMO-like glycans using high-yield and cost-effective one-pot multienzyme approaches. Quadrupole time-of-flight LC/MS analysis revealed that 22 oligosaccharides were synthesized and 9 had identical composition to known HMO. Preliminary anti-adherence activity assays indicated that fucosylated BMO decreased the uptake of enterohemorrhagic Escherichia coli O157:H7 by human intestinal epithelial Caco-2 cells more effectively than native BMO.
Collapse
Affiliation(s)
- Valerie Weinborn
- Department of Food Science and Technology, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Yanhong Li
- Glycohub, Inc., 4070 Truxel Road, Sacramento, CA 95834, USA.,Department of Chemistry, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Ishita M Shah
- Department of Food Science and Technology, University of California Davis, One Shields Avenue, Davis, CA 95616, USA.,Food for Health Institute, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Hai Yu
- Glycohub, Inc., 4070 Truxel Road, Sacramento, CA 95834, USA.,Department of Chemistry, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - David C Dallas
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - J Bruce German
- Department of Food Science and Technology, University of California Davis, One Shields Avenue, Davis, CA 95616, USA.,Food for Health Institute, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - David A Mills
- Department of Food Science and Technology, University of California Davis, One Shields Avenue, Davis, CA 95616, USA.,Food for Health Institute, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Xi Chen
- Department of Chemistry, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Daniela Barile
- Department of Food Science and Technology, University of California Davis, One Shields Avenue, Davis, CA 95616, USA.,Food for Health Institute, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
34
|
Tsai TW, Fang JL, Liang CY, Wang CJ, Huang YT, Wang YJ, Li JY, Yu CC. Exploring the Synthetic Application of Helicobacter pylori α1,3/4-Fucosyltransferase FucTIII toward the Syntheses of Fucosylated Human Milk Glycans and Lewis Antigens. ACS Catal 2019. [DOI: 10.1021/acscatal.9b03752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Tan Y, Zhang Y, Han Y, Liu H, Chen H, Ma F, Withers SG, Feng Y, Yang G. Directed evolution of an α1,3-fucosyltransferase using a single-cell ultrahigh-throughput screening method. SCIENCE ADVANCES 2019; 5:eaaw8451. [PMID: 31633018 PMCID: PMC6785251 DOI: 10.1126/sciadv.aaw8451] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/17/2019] [Indexed: 05/09/2023]
Abstract
Fucosylated glycoconjugates are involved in a variety of physiological and pathological processes. However, economical production of fucosylated drugs and prebiotic supplements has been hampered by the poor catalytic efficiency of fucosyltransferases. Here, we developed a fluorescence-activated cell sorting system that enables the ultrahigh-throughput screening (>107 mutants/hour) of such enzymes and designed a companion strategy to assess the screening performance of the system. After three rounds of directed evolution, a mutant M32 of the α1,3-FucT from Helicobacter pylori was identified with 6- and 14-fold increases in catalytic efficiency (k cat/K m) for the synthesis of Lewis x and 3'-fucosyllactose, respectively. The structure of the M32 mutant revealed that the S45F mutation generates a clamp-like structure that appears to improve binding of the galactopyranose ring of the acceptor substrate. Moreover, molecular dynamic simulations reveal that helix α5, is more mobile in the M32 mutant, possibly explaining its high fucosylation activity.
Collapse
Affiliation(s)
- Yumeng Tan
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yong Zhang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yunbin Han
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hao Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haifeng Chen
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fuqiang Ma
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, China
| | - Stephen G. Withers
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Yan Feng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guangyu Yang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Corresponding author.
| |
Collapse
|
36
|
Okada T, Ihara H, Ikeda Y. Characterization of MiFUT11 from Mangifera indica L.: A functional core α1,3-fucosyltransferase potentially involved in the biosynthesis of immunogenic carbohydrates in mango fruit. PHYTOCHEMISTRY 2019; 165:112050. [PMID: 31252202 DOI: 10.1016/j.phytochem.2019.112050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 06/09/2023]
Abstract
In higher plants, asparagine-linked oligosaccharides (N-glycans) in glycoproteins carry unique carbohydrate epitopes, namely, a core α1,3-fucose and/or a β1,2-xylose, which are common determinants responsible for the cross-reactivity of plant glycoproteins due to their strong immunogenicity. While these determinants and the relevant genes have been well characterized for herbaceous plants, information concerning whether many food plants cross-react with airborne pollens is not available. In this paper, we report on the characterization of a novel core α1,3-fucosyltransferase gene identified from Mangifera indica L., one of the major plants potentially related to food allergy. Based on sequence information of plant homologues, we amplified a candidate cDNA (MiFUT11) from pericarp tissue. An in vitro assay demonstrated that the recombinant MiFUT11 protein transfers a fucose unit onto both non-fucosylated and core α1,6-fucosylated oligosaccharides. A glycoform analysis using MALDI-TOF mass spectrometry showed that the introduction of the MiFUT11 cDNA increased the production of a core α1,3- and α1,6-fucosylated pauci-mannosidic oligosaccharide in Spodoptera Sf21 cells. Our findings suggest that MiFUT11 is a functional core α1,3-fucosyltransferase gene that is involved in the assembly of cross-reactive N-glycans in mango fruit.
Collapse
Affiliation(s)
- Takahiro Okada
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Hideyuki Ihara
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yoshitaka Ikeda
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| |
Collapse
|
37
|
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: 13] [Impact Index Per Article: 2.6] [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
|
38
|
Yu J, Shin J, Park M, Seydametova E, Jung SM, Seo JH, Kweon DH. Engineering of α-1,3-fucosyltransferases for production of 3-fucosyllactose in Escherichia coli. Metab Eng 2018; 48:269-278. [DOI: 10.1016/j.ymben.2018.05.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/12/2018] [Accepted: 05/31/2018] [Indexed: 12/23/2022]
|
39
|
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: 0.8] [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
|
40
|
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: 5.8] [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
|
41
|
Abstract
Glycosylation is a universal strategy to posttranslationally modify proteins. The recently discovered arginine rhamnosylation activates the polyproline-specific bacterial translation elongation factor EF-P. EF-P is rhamnosylated on arginine 32 by the glycosyltransferase EarP. However, the enzymatic mechanism remains elusive. In the present study, we solved the crystal structure of EarP from Pseudomonas putida. The enzyme is composed of two opposing domains with Rossmann folds, thus constituting a B pattern-type glycosyltransferase (GT-B). While dTDP-β-l-rhamnose is located within a highly conserved pocket of the C-domain, EarP recognizes the KOW-like N-domain of EF-P. Based on our data, we propose a structural model for arginine glycosylation by EarP. As EarP is essential for pathogenicity in P. aeruginosa, our study provides the basis for targeted inhibitor design. The structural and biochemical characterization of the EF-P-specific rhamnosyltransferase EarP not only provides the first molecular insights into arginine glycosylation but also lays the basis for targeted-inhibitor design against Pseudomonas aeruginosa infection.
Collapse
|
42
|
Urbanowicz BR, Bharadwaj VS, Alahuhta M, Peña MJ, Lunin VV, Bomble YJ, Wang S, Yang JY, Tuomivaara ST, Himmel ME, Moremen KW, York WS, Crowley MF. Structural, mutagenic and in silico studies of xyloglucan fucosylation in Arabidopsis thaliana suggest a water-mediated mechanism. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2017; 91:931-949. [PMID: 28670741 PMCID: PMC5735850 DOI: 10.1111/tpj.13628] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 05/17/2023]
Abstract
The mechanistic underpinnings of the complex process of plant polysaccharide biosynthesis are poorly understood, largely because of the resistance of glycosyltransferase (GT) enzymes to structural characterization. In Arabidopsis thaliana, a glycosyl transferase family 37 (GT37) fucosyltransferase 1 (AtFUT1) catalyzes the regiospecific transfer of terminal 1,2-fucosyl residues to xyloglucan side chains - a key step in the biosynthesis of fucosylated sidechains of galactoxyloglucan. We unravel the mechanistic basis for fucosylation by AtFUT1 with a multipronged approach involving protein expression, X-ray crystallography, mutagenesis experiments and molecular simulations. Mammalian cell culture expressions enable the sufficient production of the enzyme for X-ray crystallography, which reveals the structural architecture of AtFUT1 in complex with bound donor and acceptor substrate analogs. The lack of an appropriately positioned active site residue as a catalytic base leads us to propose an atypical water-mediated fucosylation mechanism facilitated by an H-bonded network, which is corroborated by mutagenesis experiments as well as detailed atomistic simulations.
Collapse
Affiliation(s)
- Breeanna R. Urbanowicz
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Vivek S. Bharadwaj
- Biosciences Division, National Renewable Energy Laboratory, Golden, Colorado 80401, USA
| | - Markus Alahuhta
- Biosciences Division, National Renewable Energy Laboratory, Golden, Colorado 80401, USA
| | - Maria J. Peña
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Vladimir V. Lunin
- Biosciences Division, National Renewable Energy Laboratory, Golden, Colorado 80401, USA
| | - Yannick J. Bomble
- Biosciences Division, National Renewable Energy Laboratory, Golden, Colorado 80401, USA
| | - Shuo Wang
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Sami T. Tuomivaara
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Michael E. Himmel
- Biosciences Division, National Renewable Energy Laboratory, Golden, Colorado 80401, USA
| | - Kelley W. Moremen
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - William S. York
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Michael F. Crowley
- Biosciences Division, National Renewable Energy Laboratory, Golden, Colorado 80401, USA
| |
Collapse
|
43
|
Han Z, Chen C, Meng C, Gao T, Peng P, Chen X, Wang F, Cao H. Chemoenzymatic synthesis of tumor-associated antigen N3 minor octasaccharide. J Carbohydr Chem 2017. [DOI: 10.1080/07328303.2017.1315123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Zhipeng Han
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, and School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Congcong Chen
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, and School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Caicai Meng
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, and School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Tian Gao
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, and School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Peng Peng
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, and School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Xi Chen
- Department of Chemistry, University of California, Davis, California, USA
| | - Fengshan Wang
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, and School of Pharmaceutical Sciences, Shandong University, Jinan, China
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, Shandong University, Jinan, China
| | - Hongzhi Cao
- National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, and School of Pharmaceutical Sciences, Shandong University, Jinan, China
- State Key Laboratory of Microbial Technology, Shandong University, Jinan, China
| |
Collapse
|
44
|
Huang D, Yang K, Liu J, Xu Y, Wang Y, Wang R, Liu B, Feng L. Metabolic engineering of Escherichia coli for the production of 2′-fucosyllactose and 3-fucosyllactose through modular pathway enhancement. Metab Eng 2017; 41:23-38. [DOI: 10.1016/j.ymben.2017.03.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/14/2017] [Accepted: 03/08/2017] [Indexed: 01/20/2023]
|
45
|
Biotechnological production of fucosylated human milk oligosaccharides: Prokaryotic fucosyltransferases and their use in biocatalytic cascades or whole cell conversion systems. J Biotechnol 2016; 235:61-83. [DOI: 10.1016/j.jbiotec.2016.03.052] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 01/29/2023]
|
46
|
Zhao X, Chen Z, Gu G, Guo Z. Recent advances in the research of bacterial glucuronosyltransferases. J Carbohydr Chem 2016. [DOI: 10.1080/07328303.2016.1205597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
47
|
Nakamura T, Tomii K. Effects of the difference in similarity measures on the comparison of ligand-binding pockets using a reduced vector representation of pockets. Biophys Physicobiol 2016; 13:139-147. [PMID: 27924268 PMCID: PMC5042158 DOI: 10.2142/biophysico.13.0_139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/06/2016] [Indexed: 12/01/2022] Open
Abstract
Comprehensive analysis and comparison of protein ligand-binding pockets are important to predict the ligands which bind to parts of putative ligand binding pockets. Because of the recent increase of protein structure information, such analysis demands a fast and efficient method for comparing ligand binding pockets. Previously we proposed a fast alignment-free method based on a simple representation of a ligand binding pocket with one 11-dimensional vector, which is suitable for such analysis. Based on that method, we conducted this study to expand and revise similarity measures of binding pockets and to investigate the effects of those modifications with two datasets for improving the ability to detect similar binding pockets. The new method exhibits higher detection performance of similar binding pockets than the previous methods and another existing accurate alignment-dependent method: APoc. Results also show that the effects of the modifications depend on the difficulty of the dataset, implying some avenues for methods of improvement.
Collapse
Affiliation(s)
- Tsukasa Nakamura
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Chiba 277-8562, Japan; Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Koto-ku, Tokyo 135-0064, Japan
| | - Kentaro Tomii
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Chiba 277-8562, Japan; Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Koto-ku, Tokyo 135-0064, Japan; Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Koto-ku, Tokyo 135-0064, Japan
| |
Collapse
|
48
|
Zhao C, Wu Y, Yu H, Shah IM, Li Y, Zeng J, Liu B, Mills DA, Chen X. The one-pot multienzyme (OPME) synthesis of human blood group H antigens and a human milk oligosaccharide (HMOS) with highly active Thermosynechococcus elongates α1-2-fucosyltransferase. Chem Commun (Camb) 2016; 52:3899-902. [PMID: 26864394 PMCID: PMC4775349 DOI: 10.1039/c5cc10646j] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A novel α1-2-fucosyltransferase from Thermosynechococcus elongatus BP-1 (Te2FT) with high fucosyltransferase activity and low donor hydrolysis activity was discovered and characterized. It was used in an efficient one-pot multienzyme (OPME) fucosylation system for the high-yield synthesis of human blood group H antigens containing β1-3-linked galactosides and an important human milk oligosaccharide (HMOS) lacto-N-fucopentaose I (LNFP I) on preparative and gram scales. LNFP I was shown to be selectively consumed by Bifidobacterium longum subsp. infantis but not Bifidobacterium animalis subsp. lactis and is a potential prebiotic.
Collapse
Affiliation(s)
- Chao Zhao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA, Tel: +1 530 754–6037; Fax: +1 530 752-8995
| | - Yijing Wu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA, Tel: +1 530 754–6037; Fax: +1 530 752-8995
| | - Hai Yu
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA, Tel: +1 530 754–6037; Fax: +1 530 752-8995
| | - Ishita M. Shah
- Department of Food Science and Technology, University of California, Davis, CA 95616, USA
| | - Yanhong Li
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA, Tel: +1 530 754–6037; Fax: +1 530 752-8995
| | - Jie Zeng
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, 453003, China
| | - Bin Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - David A. Mills
- Department of Food Science and Technology, University of California, Davis, CA 95616, USA
- Department of Viticulture and Enology, University of California, Davis, CA 95616, USA
| | - Xi Chen
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA, Tel: +1 530 754–6037; Fax: +1 530 752-8995
| |
Collapse
|
49
|
Choi YH, Kim JH, Park BS, Kim BG. Solubilization and Iterative Saturation Mutagenesis of α1,3-fucosyltransferase fromHelicobacter pylorito enhance its catalytic efficiency. Biotechnol Bioeng 2016; 113:1666-75. [DOI: 10.1002/bit.25944] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 01/29/2023]
Affiliation(s)
- Yun Hee Choi
- Interdisciplinary Program for Biochemical Engineering and Biotechnology; Institute of Molecular Biology and Genetics; Seoul National University; Seoul South Korea
| | - Jong Hoon Kim
- Interdisciplinary Program for Biochemical Engineering and Biotechnology; Institute of Molecular Biology and Genetics; Seoul National University; Seoul South Korea
| | - Bum Seok Park
- School of Chemical and Biological Engineering; Seoul National University; Seoul South Korea
| | - Byung-Gee Kim
- Interdisciplinary Program for Biochemical Engineering and Biotechnology; Institute of Molecular Biology and Genetics; Seoul National University; Seoul South Korea
- School of Chemical and Biological Engineering; Seoul National University; Seoul South Korea
- Institute of Bioengineering; Seoul National University; 151-742 Seoul South Korea
| |
Collapse
|
50
|
Abstract
We acquired molecular-resolution structures of the Golgi within its native cellular environment. Vitreous Chlamydomonas cells were thinned by cryo-focused ion beam milling and then visualized by cryo-electron tomography. These tomograms revealed structures within the Golgi cisternae that have not been seen before. Narrow trans-Golgi lumina were spanned by asymmetric membrane-associated protein arrays that had ∼6-nm lateral periodicity. Subtomogram averaging showed that the arrays may determine the narrow central spacing of the trans-Golgi cisternae through zipper-like interactions, thereby forcing cargo to the trans-Golgi periphery. Additionally, we observed dense granular aggregates within cisternae and intracisternal filament bundles associated with trans-Golgi buds. These native in situ structures provide new molecular insights into Golgi architecture and function.
Collapse
|