1
|
Kendler J, Wӧls F, Thapliyal S, Arcalis E, Gabriel H, Kubitschek S, Malzl D, Strobl MR, Palmberger D, Luber T, Unverzagt C, Paschinger K, Glauser DA, Wilson IBH, Yan S. N-glycan core tri-fucosylation requires Golgi α-mannosidase III activity that impacts nematode growth and behavior. J Biol Chem 2024; 300:107944. [PMID: 39481603 PMCID: PMC11697051 DOI: 10.1016/j.jbc.2024.107944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/04/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
N-glycans with complex core chitobiose modifications are observed in various free-living and parasitic nematodes but are absent in mammals. Using Caenorhabditis elegans as a model, we demonstrated that the core N-acetylglucosamine (GlcNAc) residues are modified by three fucosyltransferases (FUTs), namely FUT-1, FUT-6, and FUT-8. Interestingly, FUT-6 can only fucosylate N-glycans lacking the α1,6-mannose upper arm, indicating that a specific α-mannosidase is required to generate substrates for subsequent FUT-6 activity. By analyzing the N-glycomes of aman-3 KOs using offline HPLC-MALDI-TOF MS/MS, we observed that the absence of aman-3 abolishes α1,3-fucosylation of the distal GlcNAc of N-glycans, which suggests that AMAN-3 is the relevant mannosidase on whose action FUT-6 depends. Enzymatic characterization of recombinant AMAN-3 and confocal microscopy studies using a knock-in strain (aman-3::eGFP) demonstrated a Golgi localization. In contrast to the classical Golgi α-mannosidase II (AMAN-2), AMAN-3 displayed a cobalt-dependent α1,6-mannosidase activity toward N-glycans. Using AMAN-3 and other C. elegans glycoenzymes, we were able to mimic nematode N-glycan biosynthesis in vitro by remodeling a fluorescein conjugated-glycan and generate a tri-fucosylated structure. In addition, using a high-content computer-assisted C. elegans analysis platform, we observed that aman-3 deficient worms display significant developmental delays, morphological, and behavioral alterations in comparison to the WT. Our data demonstrated that AMAN-3 is a Golgi α-mannosidase required for core fucosylation of the distal GlcNAc of N-glycans. This enzyme is essential for the formation of the unusual tri-fucosylated chitobiose modifications in nematodes, which may play important roles in nematode development and behavior.
Collapse
Affiliation(s)
- Jonatan Kendler
- Institut für Parasitologie, Veterinärmedizinische Universität, Wien, Austria
| | - Florian Wӧls
- Department für Chemie, Universität für Bodenkultur, Wien, Austria
| | - Saurabh Thapliyal
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Elsa Arcalis
- Department für angewandte Genetik und Zellbiologie, Universität für Bodenkultur, Wien, Austria
| | - Hanna Gabriel
- Institut für Parasitologie, Veterinärmedizinische Universität, Wien, Austria
| | - Sascha Kubitschek
- Institut für Parasitologie, Veterinärmedizinische Universität, Wien, Austria
| | - Daniel Malzl
- Department für Chemie, Universität für Bodenkultur, Wien, Austria
| | - Maria R Strobl
- Department für Chemie, Universität für Bodenkultur, Wien, Austria
| | - Dieter Palmberger
- Department für Biotechnologie, Universität für Bodenkultur, Wien, Austria
| | - Thomas Luber
- Bioorganic Chemistry, University of Bayreuth, Bayreuth, Germany
| | - Carlo Unverzagt
- Bioorganic Chemistry, University of Bayreuth, Bayreuth, Germany
| | | | | | - Iain B H Wilson
- Department für Chemie, Universität für Bodenkultur, Wien, Austria
| | - Shi Yan
- Institut für Parasitologie, Veterinärmedizinische Universität, Wien, Austria.
| |
Collapse
|
2
|
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
|
3
|
Yagi H, Takagi K, Kato K. Exploring domain architectures of human glycosyltransferases: Highlighting the functional diversity of non-catalytic add-on domains. Biochim Biophys Acta Gen Subj 2024; 1868:130687. [PMID: 39097174 DOI: 10.1016/j.bbagen.2024.130687] [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: 06/21/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
Human glycosyltransferases (GTs) play crucial roles in glycan biosynthesis, exhibiting diverse domain architectures. This study explores the functional diversity of "add-on" domains within human GTs, using data from the AlphaFold Protein Structure Database. Among 215 annotated human GTs, 74 contain one or more add-on domains in addition to their catalytic domain. These domains include lectin folds, fibronectin type III, and thioredoxin-like domains and contribute to substrate specificity, oligomerization, and consequent enzymatic activity. Notably, certain GTs possess dual enzymatic functions due to catalytic add-on domains. The analysis highlights the importance of add-on domains in enzyme functionality and disease implications, such as congenital disorders of glycosylation. This comprehensive overview enhances our understanding of GT domain organization, providing insights into glycosylation mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Japan
| | - Katsuki Takagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan; Institute for Molecular Science, National Institutes of Natural Sciences, Japan
| | - Koichi Kato
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Japan; Institute for Molecular Science, National Institutes of Natural Sciences, Japan.
| |
Collapse
|
4
|
Tan J, Zhu L, Shi J, Zhang J, Kuang J, Guo Q, Zhu X, Chen Y, Zhou C, Gao X. Evaluation of drug resistance for EGFR-TKIs in lung cancer via multicellular lung-on-a-chip. Eur J Pharm Sci 2024; 199:106805. [PMID: 38763450 DOI: 10.1016/j.ejps.2024.106805] [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: 02/19/2024] [Revised: 04/10/2024] [Accepted: 05/17/2024] [Indexed: 05/21/2024]
Abstract
Drug resistance to irreversible epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) is a primary factor affecting their therapeutic efficacy in human non-small cell lung cancer (NSCLC). NSCLC cells can undergo epithelial-mesenchymal transition (EMT) induced by many factors in the tumour microenvironment (TME), which plays a crucial role in tumour drug resistance. In this study, a multicellular lung-on-a-chip that can realise the cell co-culture of the human non-small cell lung cancer cell line HCC827, human foetal lung fibroblasts (HFL-1), and human umbilical vein endothelial cells (HUVECs) is prepared. The TME was simulated on the chip combined with perfusion and other factors, and the drug evaluation of osimertinib was performed to explore the drug resistance mechanism of EGFR-TKIs. In the early stages, a two-dimensional static cell co-culture was achieved by microchip, and the results showed that HFL-1 cells could be transformed into cancer-associated fibroblasts (CAFs), and HCC827 cells could undergo EMT, both of which were mediated by Interleukin-6 (IL-6). Vimentin (VIM) and Alpha Skeletal Muscle Actin (a-SMA) expression of HFL-1 was upregulated, whereas E-cadherin (E-cad) expression of HCC827 was down-regulated. Further, N-cadherin (N-cad) expression of HCC827 was upregulated. In both the static cell co-culture and multicellular lung-on-a-chip, HCC827 cells with CAFs co-culture or IL-6 treatment developed resistance to osimertinib. Further use of the IL-6 antibody inhibitor tocilizumab could reverse EGFR-TKI resistance to a certain extent. Combination therapy with tocilizumab and EGFR-TKIs may provide a novel therapeutic strategy for overcoming EGFR-TKI resistance caused by EMT in NSCLC. Furthermore, the lung-on-a-chip can simulate complex TME and can be used for evaluating tumour resistance and exploring mechanisms, with the potential to become an important tool for personalised diagnosis, treatment, and biomedical research.
Collapse
Affiliation(s)
- Jianfeng Tan
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510030, China
| | - Leqing Zhu
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China; Shenzhen Clinical Medical College, Southern Medical University, Shenzhen,518101, China
| | - Jingyan Shi
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Jianhua Zhang
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Jun Kuang
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Quanwei Guo
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Xiaojia Zhu
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Yuliang Chen
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Chengbin Zhou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510030, China; Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, China.
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
5
|
Lv Y, Zhang Z, Wang M, Wang Y, Chen M, Jia J, Guo Y, Wang K, Li Z, Wang W, Li H. Discovery of novel FUT8 inhibitors with promising affinity and in vivo efficacy for colorectal cancer therapy. Bioorg Chem 2024; 149:107492. [PMID: 38820939 DOI: 10.1016/j.bioorg.2024.107492] [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: 03/26/2024] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024]
Abstract
As a member of glycosyltransferases, fucosyltransferase 8 (FUT8) is essential to core fucosylation and has been considered as a potential therapeutic target for malignant tumors, including colorectal cancer (CRC). Based on the identification of key binding residues and probable conformation of FUT8, an integrated strategy that combines virtual screening and chemical optimization was carried out and compound 15 was identified as a potent FUT8 inhibitor with novel chemical structure and in vitro antitumor activity. Moreover, chemical pulldown experiments and binding assays confirmed that compound 15 selectively bound to FUT8. In vivo, compound 15 showed promising anti-CRC effects in SW480 xenografts. These data support that compound 15 is a potential FUT8 inhibitor for CRC treatment and deserve further optimization studies.
Collapse
Affiliation(s)
- Yixin Lv
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Zhoudong Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mengmeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215123, China
| | - Yiyun Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jie Jia
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yueyue Guo
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Kai Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Zhi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
6
|
Tokoro Y, Nagae M, Nakano M, Harduin-Lepers A, Kizuka Y. LacdiNAc synthase B4GALNT3 has a unique PA14 domain and suppresses N-glycan capping. J Biol Chem 2024; 300:107450. [PMID: 38844136 PMCID: PMC11254600 DOI: 10.1016/j.jbc.2024.107450] [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: 04/05/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Structural variation of N-glycans is essential for the regulation of glycoprotein functions. GalNAcβ1-4GlcNAc (LacdiNAc or LDN), a unique subterminal glycan structure synthesized by B4GALNT3 or B4GALNT4, is involved in the clearance of N-glycoproteins from the blood and maintenance of cell stemness. Such regulation of glycoprotein functions by LDN is largely different from that by the dominant subterminal structure, N-acetyllactosamine (Galβ1-4GlcNAc, LacNAc). However, the mechanisms by which B4GALNT activity is regulated and how LDN plays different roles from LacNAc remain unclear. Here, we found that B4GALNT3 and four have unique domain organization containing a noncatalytic PA14 domain, which is a putative glycan-binding module. A mutant lacking this domain dramatically decreases the activity toward various substrates, such as N-glycan, O-GalNAc glycan, and glycoproteins, indicating that this domain is essential for enzyme activity and forms part of the catalytic region. In addition, to clarify the mechanism underlying the functional differences between LDN and LacNAc, we examined the effects of LDN on the maturation of N-glycans, focusing on the related glycosyltransferases upstream and downstream of B4GALNT. We revealed that, unlike LacNAc synthesis, prior formation of bisecting GlcNAc in N-glycan almost completely inhibits LDN synthesis by B4GALNT3. Moreover, the presence of LDN negatively impacted the actions of many glycosyltransferases for terminal modifications, including sialylation, fucosylation, and human natural killer-1 synthesis. These findings demonstrate that LDN has significant impacts on N-glycan maturation in a completely different way from LacNAc, which could contribute to obtaining a comprehensive overview of the system regulating complex N-glycan biosynthesis.
Collapse
Affiliation(s)
- Yuko Tokoro
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima, Japan
| | - Anne Harduin-Lepers
- University of Lille, CNRS, UMR 8576 -UGSF- Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Yasuhiko Kizuka
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| |
Collapse
|
7
|
Kizuka Y. Regulation of intracellular activity of N-glycan branching enzymes in mammals. J Biol Chem 2024; 300:107471. [PMID: 38879010 PMCID: PMC11328876 DOI: 10.1016/j.jbc.2024.107471] [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: 03/27/2024] [Revised: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 07/07/2024] Open
Abstract
Most proteins in the secretory pathway are glycosylated, and N-glycans are estimated to be attached to over 7000 proteins in humans. As structural variation of N-glycans critically regulates the functions of a particular glycoprotein, it is pivotal to understand how structural diversity of N-glycans is generated in cells. One of the major factors conferring structural variation of N-glycans is the variable number of N-acetylglucosamine branches. These branch structures are biosynthesized by dedicated glycosyltransferases, including GnT-III (MGAT3), GnT-IVa (MGAT4A), GnT-IVb (MGAT4B), GnT-V (MGAT5), and GnT-IX (GnT-Vb, MGAT5B). In addition, the presence or absence of core modification of N-glycans, namely, core fucose (included as an N-glycan branch in this manuscript), synthesized by FUT8, also confers large structural variation on N-glycans, thereby crucially regulating many protein-protein interactions. Numerous biochemical and medical studies have revealed that these branch structures are involved in a wide range of physiological and pathological processes. However, the mechanisms regulating the activity of the biosynthetic glycosyltransferases are yet to be fully elucidated. In this review, we summarize the previous findings and recent updates regarding regulation of the activity of these N-glycan branching enzymes. We hope that such information will help readers to develop a comprehensive overview of the complex system regulating mammalian N-glycan maturation.
Collapse
Affiliation(s)
- Yasuhiko Kizuka
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| |
Collapse
|
8
|
Tomida S, Nagae M, Kizuka Y. Distinctive domains and activity regulation of core fucosylation enzyme FUT8. Biochim Biophys Acta Gen Subj 2024; 1868:130561. [PMID: 38218458 DOI: 10.1016/j.bbagen.2024.130561] [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: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
BACKGROUND Core fucose, a structure added to the reducing end N-acetylglucosamine of N-glycans, has been shown to regulate various physiological and pathological processes, including melanoma metastasis, exacerbation of chronic obstructive pulmonary disease, and severe outcomes in COVID-19. SCOPE OF REVIEW Recent research has shed light on regulation of the activity and subcellular localization of a1,6-fucosyltransferase (FUT8), the glycosyltransferase responsible for core fucose biosynthesis, unraveling the mechanisms for controlling core fucosylation in vivo. MAJOR CONCLUSIONS This review summarizes the various features of FUT8, including its domains, structures, and substrate specificity. Additionally, we discuss the potential involvement of FUT8-binding proteins, such as oligosaccharyltransferase subunits, in the regulation of FUT8 activity, substrate specificity, and the secretion of FUT8. GENERAL SIGNIFICANCE We anticipate that this review will contribute to a deeper understanding of the control of core fucose levels in vivo and involvement of core fucosylation in FUT8-relevant functions and diseases.
Collapse
Affiliation(s)
- Seita Tomida
- The United Graduate School of Agricultural Science, Gifu University, Gifu 501-1193, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yasuhiko Kizuka
- The United Graduate School of Agricultural Science, Gifu University, Gifu 501-1193, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan.
| |
Collapse
|
9
|
Vicente JB, Guerreiro ACL, Felgueiras B, Chapla D, Tehrani D, Moremen KW, Costa J. Glycosyltransferase 8 domain-containing protein 1 (GLT8D1) is a UDP-dependent galactosyltransferase. Sci Rep 2023; 13:21684. [PMID: 38066107 PMCID: PMC10709319 DOI: 10.1038/s41598-023-48605-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Glycosyltransferases (GTs) are enzymes that catalyze the formation of glycosidic bonds and hundreds of GTs have been identified so far in humans. Glycosyltransferase 8 domain-containing protein 1 (GLT8D1) has been associated with central nervous system diseases and cancer. However, evidence on its enzymatic properties, including its substrates, has been scarcely described. In this paper, we have produced and purified recombinant secretory GLT8D1. The enzyme was found to be N-glycosylated. Differential scanning fluorimetry was employed to analyze the stabilization of GLT8D1 by Mn2+ and nucleotides, revealing UDP as the most stabilizing nucleotide scaffold. GLT8D1 displayed glycosyltransferase activity from UDP-galactose onto N-acetylgalactosamine but with a low efficiency. Modeling of the structure revealed similarities with other GT-A fold enzymes in CAZy family GT8 and glycosyltransferases in other families with galactosyl-, glucosyl-, and xylosyltransferase activities, each with retaining catalytic mechanisms. Our study provides novel structural and functional insights into the properties of GLT8D1 with implications in pathological processes.
Collapse
Affiliation(s)
- João B Vicente
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
| | - Ana Catarina L Guerreiro
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - Beatriz Felgueiras
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
| | - Digantkumar Chapla
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Daniel Tehrani
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Júlia Costa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal.
| |
Collapse
|
10
|
García-Alija M, van Moer B, Sastre DE, Azzam T, Du JJ, Trastoy B, Callewaert N, Sundberg EJ, Guerin ME. Modulating antibody effector functions by Fc glycoengineering. Biotechnol Adv 2023; 67:108201. [PMID: 37336296 PMCID: PMC11027751 DOI: 10.1016/j.biotechadv.2023.108201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Antibody based drugs, including IgG monoclonal antibodies, are an expanding class of therapeutics widely employed to treat cancer, autoimmune and infectious diseases. IgG antibodies have a conserved N-glycosylation site at Asn297 that bears complex type N-glycans which, along with other less conserved N- and O-glycosylation sites, fine-tune effector functions, complement activation, and half-life of antibodies. Fucosylation, galactosylation, sialylation, bisection and mannosylation all generate glycoforms that interact in a specific manner with different cellular antibody receptors and are linked to a distinct functional profile. Antibodies, including those employed in clinical settings, are generated with a mixture of glycoforms attached to them, which has an impact on their efficacy, stability and effector functions. It is therefore of great interest to produce antibodies containing only tailored glycoforms with specific effects associated with them. To this end, several antibody engineering strategies have been developed, including the usage of engineered mammalian cell lines, in vitro and in vivo glycoengineering.
Collapse
Affiliation(s)
- Mikel García-Alija
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain
| | - Berre van Moer
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium
| | - Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tala Azzam
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jonathan J Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Beatriz Trastoy
- Structural Glycoimmunology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| | - Nico Callewaert
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium.
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| |
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
|
López-Cortés R, Correa Pardo I, Muinelo-Romay L, Fernández-Briera A, Gil-Martín E. Core Fucosylation Mediated by the FucT-8 Enzyme Affects TRAIL-Induced Apoptosis and Sensitivity to Chemotherapy in Human SW480 and SW620 Colorectal Cancer Cells. Int J Mol Sci 2023; 24:11879. [PMID: 37569254 PMCID: PMC10418920 DOI: 10.3390/ijms241511879] [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: 06/26/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial cells can undergo apoptosis by manipulating the balance between pro-survival and apoptotic signals. In this work, we show that TRAIL-induced apoptosis can be differentially regulated by the expression of α(1,6)fucosyltransferase (FucT-8), the only enzyme in mammals that transfers the α(1,6)fucose residue to the pentasaccharide core of complex N-glycans. Specifically, in the cellular model of colorectal cancer (CRC) progression formed using the human syngeneic lines SW480 and SW620, knockdown of the FucT-8-encoding FUT8 gene significantly enhanced TRAIL-induced apoptosis in SW480 cells. However, FUT8 repression did not affect SW620 cells, which suggests that core fucosylation differentiates TRAIL-sensitive premetastatic SW480 cells from TRAIL-resistant metastatic SW620 cells. In this regard, we provide evidence that phosphorylation of ERK1/2 kinases can dynamically regulate TRAIL-dependent apoptosis and that core fucosylation can control the ERK/MAPK pro-survival pathway in which SW480 and SW620 cells participate. Moreover, the depletion of core fucosylation sensitises primary tumour SW480 cells to the combination of TRAIL and low doses of 5-FU, oxaliplatin, irinotecan, or mitomycin C. In contrast, a combination of TRAIL and oxaliplatin, irinotecan, or bevacizumab reinforces resistance of FUT8-knockdown metastatic SW620 cells to apoptosis. Consequently, FucT-8 could be a plausible target for increasing apoptosis and drug response in early CRC.
Collapse
Affiliation(s)
- Rubén López-Cortés
- Doctoral Program in Methods and Applications in Life Sciences, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| | - Isabel Correa Pardo
- Master Program in Advanced Biotechnology, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), CIBERONC, Travesía da Choupana, ES15706 Santiago de Compostela, Spain;
| | - Almudena Fernández-Briera
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| |
Collapse
|
13
|
Mao C, Li J, Feng L, Gao W. Beyond antibody fucosylation: α-(1,6)-fucosyltransferase (Fut8) as a potential new therapeutic target for cancer immunotherapy. Antib Ther 2023; 6:87-96. [PMID: 37077473 PMCID: PMC10108557 DOI: 10.1093/abt/tbad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Aberrant post-translational glycosylation is a well-established hallmark of cancer. Altered core fucosylation mediated by α-(1,6)-fucosyltransferase (Fut8) is one of the key changes in tumor glycan patterns that contributes to neoplastic transformation, tumor metastasis, and immune evasion. Increased Fut8 expression and activity are associated with many types of human cancers, including lung, breast, melanoma, liver, colorectal, ovarian, prostate, thyroid, and pancreatic cancer. In animal models, inhibition of Fut8 activity by gene knockout, RNA interference, and small analogue inhibitors led to reduced tumor growth/metastasis, downregulation of immune checkpoint molecules PD-1, PD-L1/2, and B7-H3, and reversal of the suppressive state of tumor microenvironment. Although the biologics field has long benefited tremendously from using FUT8 -/- Chinese hamster ovary cells to manufacture IgGs with greatly enhanced effector function of antibody-dependent cellular cytotoxicity for therapy, it is only in recent years that the roles of Fut8 itself in cancer biology have been studied. Here, we summarize the pro-oncogenic mechanisms involved in cancer development that are regulated by Fut8-mediated core fucosylation, and call for more research in this area where modifying the activity of this sole enzyme responsible for core fucosylation could potentially bring rewarding surprises in fighting cancer, infections, and other immune-related diseases.
Collapse
Affiliation(s)
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Lili Feng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, P. R. China
| | - Wenda Gao
- Antagen Pharmaceuticals, Inc., Canton, MA 02021, USA
| |
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
|
Van Coillie J, Pongracz T, Rahmöller J, Chen HJ, Geyer CE, van Vught LA, Buhre JS, Šuštić T, van Osch TLJ, Steenhuis M, Hoepel W, Wang W, Lixenfeld AS, Nouta J, Keijzer S, Linty F, Visser R, Larsen MD, Martin EL, Künsting I, Lehrian S, von Kopylow V, Kern C, Lunding HB, de Winther M, van Mourik N, Rispens T, Graf T, Slim MA, Minnaar RP, Bomers MK, Sikkens JJ, Vlaar AP, van der Schoot CE, den Dunnen J, Wuhrer M, Ehlers M, Vidarsson G. The BNT162b2 mRNA SARS-CoV-2 vaccine induces transient afucosylated IgG1 in naive but not in antigen-experienced vaccinees. EBioMedicine 2022; 87:104408. [PMID: 36529104 PMCID: PMC9756879 DOI: 10.1016/j.ebiom.2022.104408] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Afucosylated IgG1 responses have only been found against membrane-embedded epitopes, including anti-S in SARS-CoV-2 infections. These responses, intrinsically protective through enhanced FcγRIIIa binding, can also trigger exacerbated pro-inflammatory responses in severe COVID-19. We investigated if the BNT162b2 SARS-CoV-2 mRNA also induced afucosylated IgG responses. METHODS Blood from vaccinees during the first vaccination wave was collected. Liquid chromatography-Mass spectrometry (LC-MS) was used to study anti-S IgG1 Fc glycoprofiles. Responsiveness of alveolar-like macrophages to produce proinflammatory cytokines in presence of sera and antigen was tested. Antigen-specific B cells were characterized and glycosyltransferase levels were investigated by Fluorescence-Activated Cell Sorting (FACS). FINDINGS Initial transient afucosylated anti-S IgG1 responses were found in naive vaccinees, but not in antigen-experienced ones. All vaccinees had increased galactosylated and sialylated anti-S IgG1. Both naive and antigen-experienced vaccinees showed relatively low macrophage activation potential, as expected, due to the low antibody levels for naive individuals with afucosylated IgG1, and low afucosylation levels for antigen-experienced individuals with high levels of anti-S. Afucosylation levels correlated with FUT8 expression in antigen-specific plasma cells in naive individuals. Interestingly, low fucosylation of anti-S IgG1 upon seroconversion correlated with high anti-S IgG levels after the second dose. INTERPRETATION Here, we show that BNT162b2 mRNA vaccination induces transient afucosylated anti-S IgG1 responses in naive individuals. This observation warrants further studies to elucidate the clinical context in which potent afucosylated responses would be preferred. FUNDING LSBR1721, 1908; ZonMW10430012010021, 09150161910033, 10430012010008; DFG398859914, 400912066, 390884018; PMI; DOI4-Nr. 3; H2020-MSCA-ITN 721815.
Collapse
Affiliation(s)
- Julie Van Coillie
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Tamas Pongracz
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Johann Rahmöller
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany,Department of Anesthesiology and Intensive Care, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Hung-Jen Chen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Chiara Elisabeth Geyer
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
| | - Lonneke A. van Vught
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands,Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Jana Sophia Buhre
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Tonći Šuštić
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Thijs Luc Junior van Osch
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Maurice Steenhuis
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Willianne Hoepel
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands,Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Amsterdam Rheumatology and Immunology Center, Amsterdam, the Netherlands
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne Sophie Lixenfeld
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sofie Keijzer
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Federica Linty
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Remco Visser
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Mads Delbo Larsen
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Emily Lara Martin
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Inga Künsting
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Selina Lehrian
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Vera von Kopylow
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Carsten Kern
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Hanna Bele Lunding
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Menno de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Niels van Mourik
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands,Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Theo Rispens
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Tobias Graf
- Medical Department 2, University Heart Center of Schleswig-Holstein, Lübeck, Germany
| | - Marleen Adriana Slim
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands,Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Marije Kristianne Bomers
- Department of Internal Medicine, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - Jonne Jochum Sikkens
- Department of Internal Medicine, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - Alexander P.J. Vlaar
- Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - C. Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands,Corresponding author.
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany,Airway Research Center North, University of Lübeck, German Center for Lung Research (DZL), Lübeck, Germany,Corresponding author.
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Corresponding author.
| | | | | |
Collapse
|
16
|
Hsu YP, Verma D, Sun S, McGregor C, Mangion I, Mann BF. Successive remodeling of IgG glycans using a solid-phase enzymatic platform. Commun Biol 2022; 5:328. [PMID: 35393560 PMCID: PMC8990068 DOI: 10.1038/s42003-022-03257-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/11/2022] [Indexed: 12/02/2022] Open
Abstract
The success of glycoprotein-based drugs in various disease treatments has become widespread. Frequently, therapeutic glycoproteins exhibit a heterogeneous array of glycans that are intended to mimic human glycopatterns. While immunogenic responses to biologic drugs are uncommon, enabling exquisite control of glycosylation with minimized microheterogeneity would improve their safety, efficacy and bioavailability. Therefore, close attention has been drawn to the development of glycoengineering strategies to control the glycan structures. With the accumulation of knowledge about the glycan biosynthesis enzymes, enzymatic glycan remodeling provides a potential strategy to construct highly ordered glycans with improved efficiency and biocompatibility. In this study, we quantitatively evaluate more than 30 enzymes for glycoengineering immobilized immunoglobulin G, an impactful glycoprotein class in the pharmaceutical field. We demonstrate successive glycan remodeling in a solid-phase platform, which enabled IgG glycan harmonization into a series of complex-type N-glycoforms with high yield and efficiency while retaining native IgG binding affinity. A solid-phase glycan remodeling (SPGR) platform is presented. Over thirty enzymes were evaluated for successive glycoengineering of immobilized antibodies with outstanding performance in several SPGR reactions.
Collapse
Affiliation(s)
- Yen-Pang Hsu
- Analytical Research and Development, Merck & Co., Inc, Rahway, NJ, 07065, USA.,Exploratory Science Center, Merck & Co., Inc, Cambridge, MA, 02141, USA
| | - Deeptak Verma
- Computational and Structural Chemistry, Discovery Chemistry, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Shuwen Sun
- Analytical Research and Development, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Caroline McGregor
- Process Research & Development, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Ian Mangion
- Analytical Research and Development, Merck & Co., Inc, Rahway, NJ, 07065, USA
| | - Benjamin F Mann
- Analytical Research and Development, Merck & Co., Inc, Rahway, NJ, 07065, USA.
| |
Collapse
|
17
|
Pandey VK, Sharma R, Prajapati GK, Mohanta TK, Mishra AK. N-glycosylation, a leading role in viral infection and immunity development. Mol Biol Rep 2022; 49:8109-8120. [PMID: 35364718 PMCID: PMC8974804 DOI: 10.1007/s11033-022-07359-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 03/10/2022] [Indexed: 12/26/2022]
Abstract
N-linked protein glycosylation is an essential co-and posttranslational protein modification that occurs in all three domains of life; the assembly of N-glycans follows a complex sequence of events spanning the (Endoplasmic Reticulum) ER and the Golgi apparatus. It has a significant impact on both physicochemical properties and biological functions. It plays a significant role in protein folding and quality control, glycoprotein interaction, signal transduction, viral attachment, and immune response to infection. Glycoengineering of protein employed for improving protein properties and plays a vital role in the production of recombinant glycoproteins and struggles to humanize recombinant therapeutic proteins. It considers an alternative platform for biopharmaceuticals production. Many immune proteins and antibodies are glycosylated. Pathogen’s glycoproteins play vital roles during the infection cycle and their expression of specific oligosaccharides via the N-glycosylation pathway to evade detection by the host immune system. This review focuses on the aspects of N-glycosylation processing, glycoengineering approaches, their role in viral attachment, and immune responses to infection.
Collapse
Affiliation(s)
- Vijay Kant Pandey
- Department of Agriculture, Netaji Subhas University, Jamshedpur, Jharkhand, India
| | - Rajani Sharma
- Department of Biotechnology, Amity University Jharkhand, Niwaranpur, Ranchi, 834002, India.
| | | | | | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea.
| |
Collapse
|
18
|
Structural Insights in Mammalian Sialyltransferases and Fucosyltransferases: We Have Come a Long Way, but It Is Still a Long Way Down. Molecules 2021; 26:molecules26175203. [PMID: 34500643 PMCID: PMC8433944 DOI: 10.3390/molecules26175203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022] Open
Abstract
Mammalian cell surfaces are modified with complex arrays of glycans that play major roles in health and disease. Abnormal glycosylation is a hallmark of cancer; terminal sialic acid and fucose in particular have high levels in tumor cells, with positive implications for malignancy. Increased sialylation and fucosylation are due to the upregulation of a set of sialyltransferases (STs) and fucosyltransferases (FUTs), which are potential drug targets in cancer. In the past, several advances in glycostructural biology have been made with the determination of crystal structures of several important STs and FUTs in mammals. Additionally, how the independent evolution of STs and FUTs occurred with a limited set of global folds and the diverse modular ability of catalytic domains toward substrates has been elucidated. This review highlights advances in the understanding of the structural architecture, substrate binding interactions, and catalysis of STs and FUTs in mammals. While this general understanding is emerging, use of this information to design inhibitors of STs and FUTs will be helpful in providing further insights into their role in the manifestation of cancer and developing targeted therapeutics in cancer.
Collapse
|
19
|
García-García A, Serna S, Yang Z, Delso I, Taleb V, Hicks T, Artschwager R, Vakhrushev SY, Clausen H, Angulo J, Corzana F, Reichardt NC, Hurtado-Guerrero R. FUT8-Directed Core Fucosylation of N-glycans Is Regulated by the Glycan Structure and Protein Environment. ACS Catal 2021; 11:9052-9065. [PMID: 35662980 PMCID: PMC9161449 DOI: 10.1021/acscatal.1c01698] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/24/2021] [Indexed: 12/17/2022]
Abstract
FUT8 is an essential α-1,6-fucosyltransferase that fucosylates the innermost GlcNAc of N-glycans, a process called core fucosylation. In vitro, FUT8 exhibits substrate preference for the biantennary complex N-glycan oligosaccharide (G0), but the role of the underlying protein/peptide to which N-glycans are attached remains unclear. Here, we explored the FUT8 enzyme with a series of N-glycan oligosaccharides, N-glycopeptides, and an Asn-linked oligosaccharide. We found that the underlying peptide plays a role in fucosylation of paucimannose (low mannose) and high-mannose N-glycans but not for complex-type N-glycans. Using saturation transfer difference (STD) NMR spectroscopy, we demonstrate that FUT8 recognizes all sugar units of the G0 N-glycan and most of the amino acid residues (Asn-X-Thr) that serve as a recognition sequon for the oligosaccharyltransferase (OST). The largest STD signals were observed in the presence of GDP, suggesting that prior FUT8 binding to GDP-β-l-fucose (GDP-Fuc) is required for an optimal recognition of N-glycans. We applied genetic engineering of glycosylation capacities in CHO cells to evaluate FUT8 core fucosylation of high-mannose and complex-type N-glycans in cells with a panel of well-characterized therapeutic N-glycoproteins. This confirmed that core fucosylation mainly occurs on complex-type N-glycans, although clearly only at selected glycosites. Eliminating the capacity for complex-type glycosylation in cells (KO mgat1) revealed that glycosites with complex-type N-glycans when converted to high mannose lost the core Fuc. Interestingly, however, for erythropoietin that is uncommon among the tested glycoproteins in efficiently acquiring tetra-antennary N-glycans, two out of three N-glycosites obtained Fuc on the high-mannose N-glycans. An examination of the N-glycosylation sites of several protein crystal structures indicates that core fucosylation is mostly affected by the accessibility and nature of the N-glycan and not by the nature of the underlying peptide sequence. These data have further elucidated the different FUT8 acceptor substrate specificities both in vitro and in vivo in cells, revealing different mechanisms for promoting core fucosylation.
Collapse
Affiliation(s)
- 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 50018, Spain
| | - Sonia Serna
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Donostia San Sebastián 20014, Spain
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Ignacio Delso
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Víctor Taleb
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, Zaragoza 50018, Spain
| | - Thomas Hicks
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Raik Artschwager
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Donostia San Sebastián 20014, Spain
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Jesús Angulo
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.,Departamento de Química Orgánica, Universidad de Sevilla, Sevilla 41012, Spain.,Instituto de Investigaciones Químicas (CSIC-US), Avda. Américo Vespucio, 49, Seville 41092, Spain
| | - Francisco Corzana
- Departamento de Química, Universidad de La Rioja, Centro de Investigación en Síntesis Química, Logroño E-26006, Spain
| | - Niels C Reichardt
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Donostia San Sebastián 20014, Spain.,CIBER-BBN, Paseo Miramón 182, San Sebastian 20014, 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 50018, Spain.,Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen DK-2200, Denmark.,Fundación ARAID, Zaragoza 50018, Spain
| |
Collapse
|
20
|
Ihara H, Ikeda Y. The Roles of the N-terminal α-helical and C-terminal Src Homology 3 Domains in the Enzymatic Functions of FUT8. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2025.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Hideyuki Ihara
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine
| | - Yoshitaka Ikeda
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine
| |
Collapse
|
21
|
Ihara H, Ikeda Y. The Roles of the N-terminal α-helical and C-terminal Src Homology 3 Domains in the Enzymatic Functions of FUT8. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2025.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Hideyuki Ihara
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine
| | - Yoshitaka Ikeda
- Division of Molecular Cell Biology, Department of Biomolecular Sciences, Saga University Faculty of Medicine
| |
Collapse
|
22
|
Zhang R, Yang Q, Boruah BM, Zong G, Li C, Chapla D, Yang JY, Moremen KW, Wang LX. Appropriate aglycone modification significantly expands the glycan substrate acceptability of α1,6-fucosyltransferase (FUT8). Biochem J 2021; 478:1571-1583. [PMID: 33734311 PMCID: PMC8062310 DOI: 10.1042/bcj20210138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 12/16/2022]
Abstract
The α1,6-fucosyltransferase, FUT8, is the sole enzyme catalyzing the core-fucosylation of N-glycoproteins in mammalian systems. Previous studies using free N-glycans as acceptor substrates indicated that a terminal β1,2-GlcNAc moiety on the Man-α1,3-Man arm of N-glycan substrates is required for efficient FUT8-catalyzed core-fucosylation. In contrast, we recently demonstrated that, in a proper protein context, FUT8 could also fucosylate Man5GlcNAc2 without a GlcNAc at the non-reducing end. We describe here a further study of the substrate specificity of FUT8 using a range of N-glycans containing different aglycones. We found that FUT8 could fucosylate most of high-mannose and complex-type N-glycans, including highly branched N-glycans from chicken ovalbumin, when the aglycone moiety is modified with a 9-fluorenylmethyloxycarbonyl (Fmoc) moiety or in a suitable peptide/protein context, even if they lack the terminal GlcNAc moiety on the Man-α1,3-Man arm. FUT8 could also fucosylate paucimannose structures when they are on glycoprotein substrates. Such core-fucosylated paucimannosylation is a prominent feature of lysosomal proteins of human neutrophils and several types of cancers. We also found that sialylation of N-glycans significantly reduced their activity as a substrate of FUT8. Kinetic analysis demonstrated that Fmoc aglycone modification could either improve the turnover rate or decrease the KM value depending on the nature of the substrates, thus significantly enhancing the overall efficiency of FUT8 catalyzed fucosylation. Our results indicate that an appropriate aglycone context of N-glycans could significantly broaden the acceptor substrate specificity of FUT8 beyond what has previously been thought.
Collapse
Affiliation(s)
- Roushu Zhang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| | - Qiang Yang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| | - Bhargavi M Boruah
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| | - Digantkumar Chapla
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| |
Collapse
|
23
|
Ohkawa Y, Harada Y, Taniguchi N. Keratan sulfate-based glycomimetics using Langerin as a target for COPD: lessons from studies on Fut8 and core fucose. Biochem Soc Trans 2021; 49:441-453. [PMID: 33616615 PMCID: PMC7924997 DOI: 10.1042/bst20200780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
Glycosylation represents one of the most abundant posttranslational modification of proteins. Glycosylation products are diverse and are regulated by the cooperative action of various glycosyltransferases, glycosidases, substrates thereof: nucleoside sugars and their transporters, and chaperons. In this article, we focus on a glycosyltransferase, α1,6-fucosyltransferase (Fut8) and its product, the core fucose structure on N-glycans, and summarize the potential protective functions of this structure against emphysema and chronic obstructive pulmonary disease (COPD). Studies of FUT8 and its enzymatic product, core fucose, are becoming an emerging area of interest in various fields of research including inflammation, cancer and therapeutics. This article discusses what we can learn from studies of Fut8 and core fucose by using knockout mice or in vitro studies that were conducted by our group as well as other groups. We also include a discussion of the potential protective functions of the keratan sulfate (KS) disaccharide, namely L4, against emphysema and COPD as a glycomimetic. Glycomimetics using glycan analogs is one of the more promising therapeutics that compensate for the usual therapeutic strategy that involves targeting the genome and the proteome. These typical glycans using KS derivatives as glycomimetics, will likely become a clue to the development of novel and effective therapeutic strategies.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Antigens, Surface/physiology
- Biomimetic Materials/chemistry
- Biomimetic Materials/therapeutic use
- Fucose/metabolism
- Fucosyltransferases/physiology
- Glycosylation
- Humans
- Keratan Sulfate/chemistry
- Lectins, C-Type/antagonists & inhibitors
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Lectins, C-Type/physiology
- Mannose-Binding Lectins/antagonists & inhibitors
- Mannose-Binding Lectins/genetics
- Mannose-Binding Lectins/metabolism
- Mannose-Binding Lectins/physiology
- Mice
- Mice, Knockout
- Molecular Targeted Therapy/methods
- Polysaccharides/chemistry
- Polysaccharides/metabolism
- Pulmonary Disease, Chronic Obstructive/drug therapy
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/metabolism
Collapse
Affiliation(s)
- Yuki Ohkawa
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan
| | - Yoichiro Harada
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan
| | - Naoyuki Taniguchi
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan
| |
Collapse
|
24
|
Bastian K, Scott E, Elliott DJ, Munkley J. FUT8 Alpha-(1,6)-Fucosyltransferase in Cancer. Int J Mol Sci 2021; 22:E455. [PMID: 33466384 PMCID: PMC7795606 DOI: 10.3390/ijms22010455] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
Aberrant glycosylation is a universal feature of cancer cells that can impact all steps in tumour progression from malignant transformation to metastasis and immune evasion. One key change in tumour glycosylation is altered core fucosylation. Core fucosylation is driven by fucosyltransferase 8 (FUT8), which catalyses the addition of α1,6-fucose to the innermost GlcNAc residue of N-glycans. FUT8 is frequently upregulated in cancer, and plays a critical role in immune evasion, antibody-dependent cellular cytotoxicity (ADCC), and the regulation of TGF-β, EGF, α3β1 integrin and E-Cadherin. Here, we summarise the role of FUT8 in various cancers (including lung, liver, colorectal, ovarian, prostate, breast, melanoma, thyroid, and pancreatic), discuss the potential mechanisms involved, and outline opportunities to exploit FUT8 as a critical factor in cancer therapeutics in the future.
Collapse
Affiliation(s)
- Kayla Bastian
- Institute of Biosciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK; (E.S.); (D.J.E.); (J.M.)
| | | | | | | |
Collapse
|