1
|
Bertuzzi S, Lete MG, Franconetti A, Diercks T, Delgado S, Oyenarte I, Moure MJ, Nuñez‐Franco R, Valverde P, Lenza MP, Sobczak K, Jiménez‐Osés G, Paulson JC, Ardá A, Ereño‐Orbea J, Jiménez‐Barbero J. Exploring Glycan-Lectin Interactions in Natural-Like Environments: A View Using NMR Experiments Inside Cell and on Cell Surface. Chemistry 2025; 31:e202403102. [PMID: 39588609 PMCID: PMC11833217 DOI: 10.1002/chem.202403102] [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: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 11/27/2024]
Abstract
Glycan-mediated molecular recognition events are essential for life. NMR is widely used to monitor glycan binding to lectins in solution using isolated glycans and lectins. In this context, we herein explore diverse NMR methodologies, from both the receptor and ligand perspectives, to monitor glycan-lectin interactions under experimental conditions mimicking the native milieu inside cells and on cell surface. For the NMR experiments inside cells, galectin-7 is employed as model, since most galectins are soluble and carry out their functions in the cellular micro-environment. Using Danio Rerio oocytes, the 1H-15N HMQC NMR spectrum of a folded galectin has been observed inside cell for the first time, using a glycomimetic ligand (TDG) to overcoming the natural tendency of galectins to bind to numerous galactose-containing receptors within cells. Alternatively, most lectins, other than galectins, are displayed on the cell surface, providing a multivalent presentation to bind their glycan partners in cis (at the same cell) or in trans (on other cells). In this case, ligand-based STD-NMR experiments have been successfully applied to account for the interactions of natural glycans and glycomimetics with Siglec-10. These methodologies provide the proof-of-concept to open the door to the NMR analysis of the recognition of glycans in native-like settings.
Collapse
Affiliation(s)
- Sara Bertuzzi
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Marta G. Lete
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Antonio Franconetti
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Tammo Diercks
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Sandra Delgado
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Iker Oyenarte
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Maria J. Moure
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Reyes Nuñez‐Franco
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Pablo Valverde
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Maria Pia Lenza
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Klaudia Sobczak
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - Gonzalo Jiménez‐Osés
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
| | - James C. Paulson
- Departments of Molecular Medicine and Immunology & MicrobiologyThe Scripps Research Institute10550 North Torrey Pines RoadLa Jolla, California92037USA
| | - Ana Ardá
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - June Ereño‐Orbea
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - Jesús Jiménez‐Barbero
- Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)48160Derio, BizkaiaSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Department of Organic & Inorganic ChemistryFaculty of Science and TechnologyUniversity of the Basque Country, EHU-UPV48940Leioa, BizkaiaSpain
- Centro de Investigacion Biomedica En Red de Enfermedades Respiratorias28029MadridSpain
| |
Collapse
|
2
|
Lin SY, Schmidt EN, Takahashi-Yamashiro K, Macauley MS. Roles for Siglec-glycan interactions in regulating immune cells. Semin Immunol 2024; 77:101925. [PMID: 39706106 DOI: 10.1016/j.smim.2024.101925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Cell surface complex carbohydrates, known as glycans, are positioned to be the first point of contact between two cells. Indeed, interactions between glycans with glycan-binding can modulate cell-cell interactions. This concept is particularly relevant for immune cells, which use an array of glycan-binding proteins to help in the process of differentiating 'self' from 'non-self'. This is exemplified by the sialic acid-binding immunoglobulin-type lectins (Siglecs), which recognize sialic acid. Given that sialic acid is relatively unique to vertebrates, immune cells leverage Siglecs to recognize sialic acid as a marker of 'self'. Siglecs serve many biological roles, with most of these functions regulated through interactions with their sialoglycan ligands. In this review, we provide a comprehensive update on the ligands of Siglecs and how Siglec-sialoglycan interactions help regulate immune cells in the adaptive and innate immune system.
Collapse
Affiliation(s)
- Sung-Yao Lin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Matthew S Macauley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada; Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
3
|
Sojitra M, Schmidt EN, Lima GM, Carpenter EJ, McCord KA, Atrazhev A, Macauley MS, Derda R. Measuring carbohydrate recognition profile of lectins on live cells using liquid glycan array (LiGA). Nat Protoc 2024:10.1038/s41596-024-01070-3. [PMID: 39415074 DOI: 10.1038/s41596-024-01070-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 09/05/2024] [Indexed: 10/18/2024]
Abstract
Glycans constitute a significant fraction of biomolecular diversity on cellular surfaces across all kingdoms of life. As the structure of glycans is not directly encoded by the organism's DNA, it is impossible to use high-throughput DNA technologies to study the role of cellular glycosylation or to understand how glycocalyx is recognized by glycan-binding proteins (GBPs). To address this gap, we recently described a liquid glycan array (LiGA) platform that allows profiling of glycan-GBP interactions on the surface of live cells in vitro and in vivo using next-generation sequencing. LiGA is a library of DNA-barcoded bacteriophages, where each clonal bacteriophage displays 5-1,500 copies of a glycan and the distinct DNA barcode inside each bacteriophage clone encodes the structure and density of the displayed glycans. Deep sequencing of the glycophages associated with live cells yields a glycan-binding profile of GBPs expressed on the surface of cells. This protocol provides detailed instructions for how to use LiGA to probe cell surface receptors and includes information on the preparation of glycophages, analysis by MALDI-TOF mass spectrometry, the assembly of a LiGA library and its deep sequencing. Using this protocol, we measure glycan-binding profiles of the immunomodulatory sialic acid-binding immunoglobulin-like lectins‑1, -2, -6, -7 and -9 expressed on the surface of different cell types. Compared with existing methods that require complex specialist equipment, this method allows users with basic molecular biology expertise to measure the precise glycan-binding profile of GBPs on the surface of any cell type expressing exogenous GBP within 2-3 d.
Collapse
Affiliation(s)
- Mirat Sojitra
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Guilherme M Lima
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Eric J Carpenter
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Kelli A McCord
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Alexey Atrazhev
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
4
|
Akatsu C, Naito-Matsui Y, Abdu-Allah HHM, Imamura A, Long W, Ishida H, Takematsu H, Tsubata T. Neu5Gc-mediated high-affinity interaction is dispensable for CD22 cis-ligands to regulate B cell signaling. J Biol Chem 2024; 300:107630. [PMID: 39098526 PMCID: PMC11407986 DOI: 10.1016/j.jbc.2024.107630] [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: 07/05/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/06/2024] Open
Abstract
CD22 (also known as Siglec-2) is an inhibitory receptor expressed in B cells. CD22 specifically recognizes α2,6 sialic acid and interacts with α2,6 sialylated membrane proteins expressed on the same cell (cis-ligands) and those derived from outside of the cell (trans-ligands). Previously, CD22 cis-ligands were shown to regulate the activity of CD22, thereby regulating both BCR ligation-induced signaling and low-level "tonic" signaling in the absence of BCR ligation that regulates the survival and differentiation of B cells. Mouse CD22 prefers Neu5Gc to Neu5Ac thereby binding to α2,6-linked Neu5Gc with high affinity. Although human CD22 binds to a distinct α2,6 sialylated glycan with high affinity, expression of high-affinity ligands is regulated in a conserved and stringent manner. However, how high- versus low-affinity CD22 ligands regulate B cells is poorly understood. Here we demonstrate that the interaction of CD22 with the endogenous ligands enhances BCR ligation-induced signaling but reduces tonic signaling in Cmah-/- mouse B cells deficient in Neu5Gc as well as wild-type B cells. Moreover, Cmah-/- B cells do not show alterations in the phenotypes correlated to tonic signaling. These results indicate that low-affinity interaction of the CD22 cis-ligands with CD22 is sufficient for the regulation of B cell signaling, and suggest that expression of high-affinity CD22 ligands might be involved in the regulation of B cells by competing for the binding of CD22 with exogenous trans-ligands of CD22.
Collapse
Affiliation(s)
- Chizuru Akatsu
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Naito-Matsui
- Department of Molecular Cell Biology, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | | | - Akihiro Imamura
- Department of Applied Bio-organic Chemistry, Gifu University, Gifu, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Wang Long
- Department of Pathology, Nihon University School of Dentistry, Tokyo, Japan
| | - Hideharu Ishida
- Department of Applied Bio-organic Chemistry, Gifu University, Gifu, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Hiromu Takematsu
- Department of Molecular Cell Biology, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pathology, Nihon University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
5
|
Mariuzza RA, Shahid S, Karade SS. The immune checkpoint receptor LAG3: Structure, function, and target for cancer immunotherapy. J Biol Chem 2024; 300:107241. [PMID: 38556085 PMCID: PMC11061240 DOI: 10.1016/j.jbc.2024.107241] [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: 01/29/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
Lymphocyte activation gene 3 protein (LAG3) is an immune checkpoint receptor that is highly upregulated on exhausted T cells in the tumor microenvironment. LAG3 transmits inhibitory signals to T cells upon binding to MHC class II and other ligands, rendering T cells dysfunctional. Consequently, LAG3 is a major target for cancer immunotherapy with many anti-LAG3 monoclonal antibodies (mAbs) that block LAG3 inhibitory activity in clinical trials. In this review, we examine the molecular basis for LAG3 function in light of recently determined crystal and cryoEM structures of this inhibitory receptor. We review what is known about LAG3 interactions with MHC class II, its canonical ligand, and the newly discovered ligands FGL1 and the T cell receptor (TCR)-CD3 complex, including current controversies over the relative importance of these ligands. We then address the development and mechanisms of action of anti-LAG3 mAbs in clinical trials for cancer immunotherapy. We discuss new strategies to therapeutically target LAG3 using mAbs that not only block the LAG3-MHC class II interaction, but also LAG3 interactions with FGL1 or TCR-CD3, or that disrupt LAG3 dimerization. Finally, we assess the possibility of developing mAbs that enhance, rather than block, LAG3 inhibitory activity as treatments for autoimmune diseases.
Collapse
Affiliation(s)
- Roy A Mariuzza
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA.
| | - Salman Shahid
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Sharanbasappa S Karade
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
6
|
Wißfeld J, Abou Assale T, Cuevas-Rios G, Liao H, Neumann H. Therapeutic potential to target sialylation and SIGLECs in neurodegenerative and psychiatric diseases. Front Neurol 2024; 15:1330874. [PMID: 38529039 PMCID: PMC10961342 DOI: 10.3389/fneur.2024.1330874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
Sialic acids, commonly found as the terminal carbohydrate on the glycocalyx of mammalian cells, are pivotal checkpoint inhibitors of the innate immune system, particularly within the central nervous system (CNS). Sialic acid-binding immunoglobulin-like lectins (SIGLECs) expressed on microglia are key players in maintaining microglial homeostasis by recognizing intact sialylation. The finely balanced sialic acid-SIGLEC system ensures the prevention of excessive and detrimental immune responses in the CNS. However, loss of sialylation and SIGLEC receptor dysfunctions contribute to several chronic CNS diseases. Genetic variants of SIGLEC3/CD33, SIGLEC11, and SIGLEC14 have been associated with neurodegenerative diseases such as Alzheimer's disease, while sialyltransferase ST8SIA2 and SIGLEC4/MAG have been linked to psychiatric diseases such as schizophrenia, bipolar disorders, and autism spectrum disorders. Consequently, immune-modulatory functions of polysialic acids and SIGLEC binding antibodies have been exploited experimentally in animal models of Alzheimer's disease and inflammation-induced CNS tissue damage, including retinal damage. While the potential of these therapeutic approaches is evident, only a few therapies to target either sialylation or SIGLEC receptors have been tested in patient clinical trials. Here, we provide an overview of the critical role played by the sialic acid-SIGLEC axis in shaping microglial activation and function within the context of neurodegeneration and synaptopathies and discuss the current landscape of therapies that target sialylation or SIGLECs.
Collapse
Affiliation(s)
- Jannis Wißfeld
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Tawfik Abou Assale
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - German Cuevas-Rios
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Huan Liao
- Florey Institute of Neuroscience and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
7
|
Conti G, Bärenwaldt A, Rabbani S, Mühlethaler T, Sarcevic M, Jiang X, Schwardt O, Ricklin D, Pieters RJ, Läubli H, Ernst B. Tetra- and Hexavalent Siglec-8 Ligands Modulate Immune Cell Activation. Angew Chem Int Ed Engl 2023; 62:e202314280. [PMID: 37947772 DOI: 10.1002/anie.202314280] [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: 09/26/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Carbohydrate-binding proteins are generally characterized by poor affinities for their natural glycan ligands, predominantly due to the shallow and solvent-exposed binding sites. To overcome this drawback, nature has exploited multivalency to strengthen the binding by establishing multiple interactions simultaneously. The development of oligovalent structures frequently proved to be successful, not only for proteins with multiple binding sites, but also for proteins that possess a single recognition domain. Herein we present the syntheses of a number of oligovalent ligands for Siglec-8, a monomeric I-type lectin found on eosinophils and mast cells, alongside the thermodynamic characterization of their binding. While the enthalpic contribution of each binding epitope was within a narrow range to that of the monomeric ligand, the entropy penalty increased steadily with growing valency. Additionally, we observed a successful agonistic binding of the tetra- and hexavalent and, to an even larger extent, multivalent ligands to Siglec-8 on immune cells and modulation of immune cell activation. Thus, triggering a biological effect is not restricted to multivalent ligands but could be induced by low oligovalent ligands as well, whereas a monovalent ligand, despite binding with similar affinity, showed an antagonistic effect.
Collapse
Affiliation(s)
- Gabriele Conti
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
- Chemical Biology and Drug Discovery Group, Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Anne Bärenwaldt
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, Hebelstrasse 20, 4051, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Petersgraben 4, 4051, Basel, Switzerland
| | - Said Rabbani
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Tobias Mühlethaler
- Biophysics Facility, Department Biozentrum, University of Basel, Spitalstrasse 41, 4056, Basel, Switzerland
| | - Mirza Sarcevic
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, Hebelstrasse 20, 4051, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Petersgraben 4, 4051, Basel, Switzerland
| | - Xiaohua Jiang
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Oliver Schwardt
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Daniel Ricklin
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Roland J Pieters
- Chemical Biology and Drug Discovery Group, Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, Hebelstrasse 20, 4051, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Petersgraben 4, 4051, Basel, Switzerland
| | - Beat Ernst
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| |
Collapse
|
8
|
Cao Y, Rische CH, Bochner BS, O’Sullivan JA. Interactions between Siglec-8 and endogenous sialylated cis ligands restrain cell death induction in human eosinophils and mast cells. Front Immunol 2023; 14:1283370. [PMID: 37928558 PMCID: PMC10623328 DOI: 10.3389/fimmu.2023.1283370] [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: 08/25/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a sialoside-binding receptor expressed by eosinophils and mast cells that exhibits priming status- and cell type-dependent inhibitory activity. On eosinophils that have been primed with IL-5, GM-CSF, or IL-33, antibody ligation of Siglec-8 induces cell death through a pathway involving the β2 integrin-dependent generation of reactive oxygen species (ROS) via NADPH oxidase. In contrast, Siglec-8 engagement on mast cells inhibits cellular activation and mediator release but reportedly does not impact cell viability. The differences in responses between cytokine-primed and unprimed eosinophils, and between eosinophils and mast cells, to Siglec-8 ligation are not understood. We previously found that Siglec-8 binds to sialylated ligands present on the surface of the same cell (so-called cis ligands), preventing Siglec-8 ligand binding in trans. However, the functional relevance of these cis ligands has not been elucidated. We therefore explored the potential influence of cis ligands of Siglec-8 on both eosinophils and mast cells. De-sialylation using exogenous sialidase profoundly altered the consequences of Siglec-8 antibody engagement on both cell types, eliminating the need for cytokine priming of eosinophils to facilitate cell death and enabling Siglec-8-dependent mast cell death without impacting anti-Siglec-8 antibody binding. The cell death process licensed by de-sialylation resembled that characterized in IL-5-primed eosinophils, including CD11b upregulation, ROS production, and the activities of Syk, PI3K, and PLC. These results implicate cis ligands in restraining Siglec-8 function on eosinophils and mast cells and reveal a promising approach to the selective depletion of mast cells in patients with mast cell-mediated diseases.
Collapse
Affiliation(s)
- Yun Cao
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Clayton H. Rische
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL, United States
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jeremy A. O’Sullivan
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
9
|
Hui E. Cis Interactions of Membrane Receptors and Ligands. Annu Rev Cell Dev Biol 2023; 39:391-408. [PMID: 37339682 PMCID: PMC11734567 DOI: 10.1146/annurev-cellbio-120420-103941] [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] [Indexed: 06/22/2023]
Abstract
Cell-cell communication is critical for the development and function of multicellular organisms. A crucial means by which cells communicate with one another is physical interactions between receptors on one cell and their ligands on a neighboring cell. Trans ligand:receptor interactions activate the receptor, ultimately leading to changes in the fate of the receptor-expressing cells. Such trans signaling is known to be critical for the functions of cells in the nervous and immune systems, among others. Historically, trans interactions are the primary conceptual framework for understanding cell-cell communication. However, cells often coexpress many receptors and ligands, and a subset of these has been reported to interact in cis and profoundly impact cell functions. Cis interactions likely constitute a fundamental, understudied regulatory mechanism in cell biology. Here, I discuss how cis interactions between membrane receptors and ligands regulate immune cell functions, and I also highlight outstanding questions in the field.
Collapse
Affiliation(s)
- Enfu Hui
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, USA;
| |
Collapse
|
10
|
Tsubata T. The ligand interactions of B cell Siglecs are involved in the prevention of autoimmunity to sialylated self-antigens and in the quality control of signaling-competent B cells. Int Immunol 2023; 35:461-473. [PMID: 37504378 DOI: 10.1093/intimm/dxad030] [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: 05/17/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are a family of membrane molecules that recognize sialic acid. Most of them are inhibitory receptors that inhibit immune-cell activation by recognizing sialic acid as a self-motif. Human B cells express CD22 (also known as Siglec-2), Siglec-5, Siglec-6 and Siglec-10 whereas mouse B cells express CD22 and Siglec-G (ortholog of human Siglec-10). Siglecs recognize both sialylated molecules expressed on the same cell (cis-ligands) and those expressed by other cells (trans-ligands). In Guillain-Barré syndrome (GBS), antibody production to gangliosides (which are sialic acid-containing glycolipids) expressed by neurons plays a pathogenic role. A Siglec-10 variant deficient in recognition of gangliosides is genetically associated with GBS, suggesting that Siglec-10 induces self-tolerance to gangliosides by recognizing gangliosides as trans-ligands. Recognition of the BCR as a cis-ligand by Siglec-G and CD22 suppresses BCR signaling in B-1 cells and conventional B cells, respectively. This signal suppression prevents excess expansion of B-1 cells and is involved in the quality control of signaling-competent B cells by setting a threshold for tonic signaling during B cell development. CD22 recognizes other cis-ligands including CD22 and β7 integrin. Interaction of CD22 with other CD22 molecules induces CD22 clustering that suppresses CD22-mediated signal inhibition upon BCR ligation, and interaction with β7 integrin maintains its function in the gut-homing of B cells. Taken together, interactions of B cell Siglecs with multiple trans- and cis-ligands play important roles in B cell homeostasis and immune responses.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Pathology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| |
Collapse
|
11
|
Tsubata T. Siglec cis-ligands and their roles in the immune system. Glycobiology 2023; 33:532-544. [PMID: 37154567 DOI: 10.1093/glycob/cwad038] [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: 10/20/2022] [Revised: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins are a family of membrane molecules primarily expressed in immune cells. Most of them are inhibitory receptors containing immunoreceptor tyrosine-based inhibition motifs in the cytoplasmic tail. On the cell surface, sialic acid-binding immunoglobulin-like lectins are mostly bound by sialylated glycans on membrane molecules expressed in the same cell (cis-ligands). Although ligands of sialic acid-binding immunoglobulin-like lectins are not efficiently identified by conventional methods such as immunoprecipitation, in situ labeling including proximity labeling is useful in identifying both cis-ligands and the sialylated ligands expressed by other cells (trans-ligands) of sialic acid-binding immunoglobulin-like lectins. Interaction of the inhibitory sialic acid-binding immunoglobulin-like lectins with cis-ligands including both those with and without signaling function modulates the inhibitory activity of sialic acid-binding immunoglobulin-like lectins by multiple different ways. This interaction also modulates signaling function of the cis-ligands. So far, little is known about the role of the interaction between sialic acid-binding immunoglobulin-like lectins and the cis-ligands. Nonetheless, recent studies showed that the inhibitory activity of CD22 (also known as Siglec-2) is regulated by endogenous ligands, most likely cis-ligands, differentially in resting B cells and those in which B-cell antigen receptor is ligated. This differential regulation plays a role in quality control of signaling-competent B cells and also partial restoration of B-cell antigen receptor signaling in immunodeficient B cells.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Pathology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| |
Collapse
|
12
|
Klaas M, Dubock S, Ferguson DJP, Crocker PR. Sialoadhesin (CD169/Siglec-1) is an extended molecule that escapes inhibitory cis-interactions and synergizes with other macrophage receptors to promote phagocytosis. Glycoconj J 2023; 40:213-223. [PMID: 36738392 PMCID: PMC10027830 DOI: 10.1007/s10719-022-10097-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 02/05/2023]
Abstract
Sialoadhesin (CD169/Siglec-1, Sn) is a macrophage receptor that interacts with sialic acids on both host cells and pathogens. It is a type 1 membrane protein with an unusually large number of 17 extracellular immunoglobulin (Ig)-like domains, made up of an N-terminal V-set domain that binds sialic acid and 16 adjacent C2-set domains. The potential importance of 17 Ig domains in Sn for mediating cellular interactions has not been investigated experimentally. In the present study, Chinese Hamster Ovary (CHO) cells were stably transfected with full-length or truncated forms of Sn. Using human red blood cells (RBC) as a model system, CHO cells expressing truncated forms of Sn with 4 or less Ig domains were unable to bind RBC in comparison to the full-length protein. Immunoelectron microscopy of the CHO cells indicated that full-length Sn extends ~ 33 nm from the plasma membrane compared with ~ 14 nm for a truncated form with 6 N-terminal Ig domains. Co-expresssion of Sn-expressing CHO cells with heavily glycosylated membrane proteins of differing predicted lengths resulted in selective modulation of Sn-dependent binding to RBC and supported the hypothesis that Sn has evolved 17 Ig domains to escape inhibitory cis-interactions. The functional significance of the extended length of Sn was demonstrated in experiments with macrophages showing that Sn synergizes with phagocytic receptors FcR and TIM-4 to strongly promote uptake of IgG-opsonized and eryptotic RBC respectively.
Collapse
Affiliation(s)
- Mariliis Klaas
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, Tartu, Estonia
| | - Stuart Dubock
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom
| | - David J P Ferguson
- Nuffield Division of Clinical Laboratory Sciences, Oxford University, John Radcliffe Hospital, Oxford, United Kingdom
- Department Biological & Medical Sciences, Oxford Brookes University, Oxford, United Kingdom
| | - Paul R Crocker
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, United Kingdom.
| |
Collapse
|
13
|
Abstract
Siglecs are a family of immunomodulatory cell surface receptors present on white blood cells. Binding to cell surface sialic acid-containing glycans modulates the proximity of Siglecs to other receptors that they regulate. This proximity is key to enabling signaling motifs on the cytosolic domain of Siglecs to modulate immune responses. Given the important roles that Siglecs play in helping to maintain immune homeostasis, a better understanding of their glycan ligands is needed to elucidate their roles in health and disease. A common approach for probing Siglec ligands on cells is to use soluble versions of the recombinant Siglecs in conjunction with flow cytometry. Flow cytometry has many advantages in enabling the relative levels of Siglec ligands between cell types to be rapidly quantified. Here, a step-by-step protocol is described on how to detect Siglec ligands most sensitively and accurately on cells by flow cytometry.
Collapse
Affiliation(s)
- Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
14
|
Genetically encoded chemical crosslinking of carbohydrate. Nat Chem 2023; 15:33-42. [PMID: 36216893 PMCID: PMC9840686 DOI: 10.1038/s41557-022-01059-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/02/2022] [Indexed: 01/17/2023]
Abstract
Protein-carbohydrate interactions play important roles in various biological processes, such as organism development, cancer metastasis, pathogen infection and immune response, but they remain challenging to study and exploit due to their low binding affinity and non-covalent nature. Here we site-specifically engineered covalent linkages between proteins and carbohydrates under biocompatible conditions. We show that sulfonyl fluoride reacts with glycans via a proximity-enabled reactivity, and to harness this a bioreactive unnatural amino acid (SFY) that contains sulfonyl fluoride was genetically encoded into proteins. SFY-incorporated Siglec-7 crosslinked with its sialoglycan ligand specifically in vitro and on the surface of cancer cells. Through irreversible cloaking of sialoglycan at the cancer cell surface, SFY-incorporated Siglec-7 enhanced the killing of cancer cells by natural killer cells. Genetically encoding the chemical crosslinking of proteins to carbohydrates (GECX-sugar) offers a solution to address the low affinity and weak strength of protein-sugar interactions.
Collapse
|
15
|
Cooper O, Waespy M, Chen D, Kelm S, Li Q, Haselhorst T, Tiralongo J. Sugar-decorated carbon dots: a novel tool for targeting immunomodulatory receptors. NANOSCALE ADVANCES 2022; 4:5355-5364. [PMID: 36540112 PMCID: PMC9729803 DOI: 10.1039/d2na00364c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/14/2022] [Indexed: 06/17/2023]
Abstract
Interactions between sialic acid (Sia) and sialic acid-binding immunoglobulin-like lectins (siglecs) regulate the immune system, with aberrations contributing to pathologies such as autoimmunity, infectious disease and cancer. Over the last decade, several multivalent Sia ligands have been synthesized to modulate the Sia-binding affinity of proteins/lectins. Here, we report a novel class of multivalent siglec probes through the decoration of α(2,6)-sialyllactose ligands on inherently fluorescent carbon dots (CD). We show that the preference of α(2,3)-linked Sia for siglec-1 can be altered by increasing the multivalence of Sia ligands present on the CD, and that a locally high glycan concentration can have a direct effect on linkage specificity. Additionally, micromolar (IC50 ∼ 70 μM) interaction of α(2,6)-sialyllactose-CD (6-CD) with siglec-2 (CD22) revealed it was capable of generating a significant cytotoxic effect on Burkitt's Lymphoma (BL) Daudi B cells. This phenonomen was attributed to 6-CD's ability to form trans interactions with CD22 on masked BL Daudi cells as a direct result of clustering of the Sia moiety on the CD surface. Overall, our glycoengineered carbon dots represent a novel high affinity molecular probe with multiple applications in sialoglycoscience and medicine.
Collapse
Affiliation(s)
- Oren Cooper
- Institute for Glycomics, Gold Coast Campus, Griffith University Queensland 4222 Australia
| | - Mario Waespy
- Centre for Biomolecular Interactions Bremen, Department of Biology and Chemistry, University of Bremen 28334 Bremen Germany
| | - Dechao Chen
- School of Engineering and Built Environment, Nathan Campus, Griffith University QLD 4111 Australia
| | - Sørge Kelm
- Centre for Biomolecular Interactions Bremen, Department of Biology and Chemistry, University of Bremen 28334 Bremen Germany
| | - Qin Li
- School of Engineering and Built Environment, Nathan Campus, Griffith University QLD 4111 Australia
- Queensland Micro- and Nanotechnology Centre, Australia, Nathan Campus, Griffith University QLD 4111 Australia
| | - Thomas Haselhorst
- Institute for Glycomics, Gold Coast Campus, Griffith University Queensland 4222 Australia
| | - Joe Tiralongo
- Institute for Glycomics, Gold Coast Campus, Griffith University Queensland 4222 Australia
| |
Collapse
|
16
|
Ton Tran HT, Li C, Chakraberty R, Cairo CW. NEU1 and NEU3 enzymes alter CD22 organization on B cells. BIOPHYSICAL REPORTS 2022; 2:100064. [PMID: 36425332 PMCID: PMC9680808 DOI: 10.1016/j.bpr.2022.100064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/19/2022] [Indexed: 06/16/2023]
Abstract
The B cell membrane expresses sialic-acid-binding immunoglobulin-like lectins, also called Siglecs, that are important for modulating immune response. Siglecs have interactions with sialoglycoproteins found on the same membrane (cis-ligands) that result in homotypic and heterotypic receptor clusters. The regulation and organization of these clusters, and their effect on cell activation, is not clearly understood. We investigated the role of human neuraminidase enzymes NEU1 and NEU3 on the clustering of CD22 on B cells using confocal microscopy. We observed that native NEU1 and NEU3 activity influence the cluster size of CD22. Using single-particle tracking, we observed that NEU3 activity increased the lateral mobility of CD22, which was in contrast to the effect of exogenous bacterial NEU enzymes. Moreover, we show that native NEU1 and NEU3 activity influenced cellular Ca2+ levels, supporting a role for these enzymes in regulating B cell activation. Our results establish a role for native NEU activity in modulating CD22 organization and function on B cells.
Collapse
Affiliation(s)
- Hanh-Thuc Ton Tran
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Caishun Li
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
17
|
Prescher H, Schweizer A, Frank M, Kuhfeldt E, Ring J, Nitschke L. Targeting Human CD22/Siglec-2 with Dimeric Sialosides as Novel Oligosaccharide Mimetics. J Med Chem 2022; 65:10588-10610. [PMID: 35881556 DOI: 10.1021/acs.jmedchem.2c00765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Significant interest in the development of high-affinity ligands for Siglecs exists due to the various therapeutically relevant functions of these proteins. Here, we report a new strategy to develop and design Siglec ligands as disialyl-oligosaccharide mimetics exemplified on Siglec-2 (CD22). We report insights into development of dimeric ligands with high affinity and avidity to cell surface-expressed CD22, assay development, tool compounds, structure activity relationships, and biological data on calcium flux regulation in B-cells. The binding modes of selected ligands have been modeled based on state-of-the-art molecular dynamics simulations on the microsecond timescale, providing detailed views on ligand binding and opening a new perspective on drug design efforts for Siglecs. High-avidity dimeric ligands containing a linker opening the way towards bispecifics are presented as well.
Collapse
Affiliation(s)
| | - Astrid Schweizer
- Chair of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Martin Frank
- Biognos AB, Generatorsgatan 1, 40274 Göteborg, Sweden
| | | | - Julia Ring
- Chair of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Lars Nitschke
- Chair of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| |
Collapse
|
18
|
Cummings RD. The mannose receptor ligands and the macrophage glycome. Curr Opin Struct Biol 2022; 75:102394. [PMID: 35617912 DOI: 10.1016/j.sbi.2022.102394] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/12/2022] [Accepted: 04/16/2022] [Indexed: 11/03/2022]
Abstract
A unique glycan-binding protein expressed in macrophages and some types of other immune cells is the mannose receptor (MR, CD206). It is an endocytic, transmembrane protein with multiple glycan-binding domains and different specificities in binding glycans. The mannose receptor is important as it has major roles in diverse biological processes, including regulation of circulating levels of reproductive hormones, homeostasis, innate immunity, and infections. These different functions involve the recognition of a wide range of glycans, and their nature is currently under intense study. But the mannose receptor is just one of many glycan-binding proteins expressed in macrophages, leading to an interest in the potential relationship between the macrophage glycome and how it may regulate cognate glycan-binding protein activities. This review focuses primarily on the mannose receptor and its carbohydrate ligands, as well as macrophages and their glycomes.
Collapse
Affiliation(s)
- Richard D Cummings
- Beth Israel Deaconess Medical Center, Department of Surgery, Harvard Medical School, CLS 11087 - 3 Blackfan Circle, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Harumoto T, Iwai H, Tanigawa M, Kubo T, Atsumi T, Tsutsumi K, Takashima M, Destito G, Soloff R, Tomizuka K, Nycholat C, Paulson J, Uehara K. Enhancement of Gene Knockdown on CD22-Expressing Cells by Chemically Modified Glycan Ligand-siRNA Conjugates. ACS Chem Biol 2022; 17:292-298. [PMID: 35020348 DOI: 10.1021/acschembio.1c00652] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Extrahepatic targeted delivery of oligonucleotides, such as small interfering RNA (siRNA) and antisense oligonucleotides (ASOs), is an attractive technology for the development of nucleic acid-based medicines. To target CD22-expressing B cells, several drug platforms have shown promise, including antibodies, antibody-drug conjugates, and nanoparticles, but to date CD22-targeted delivery of oligonucleotide therapeutics has not been reported. Here we report the uptake and enhancement of siRNA gene expression knockdown in CD22-expressing B cells using a chemically stabilized and modified CD22 glycan ligand-conjugated siRNA. This finding has the potential to broaden the use of siRNA technology, opening up novel therapeutic opportunities, and presents an innovative approach for targeted delivery of siRNAs to B cell lymphomas.
Collapse
Affiliation(s)
- Toshimasa Harumoto
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Hiroto Iwai
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Mari Tanigawa
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Toshiko Kubo
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Toshiyuki Atsumi
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Kyoko Tsutsumi
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Michio Takashima
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Giuseppe Destito
- Kyowa Kirin Inc., 9420 Athena Circle, La Jolla, California 92037, United States
| | - Rachel Soloff
- Kyowa Kirin Inc., 9420 Athena Circle, La Jolla, California 92037, United States
| | - Kazuma Tomizuka
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Corwin Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - James Paulson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Keiji Uehara
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
20
|
Ishida T, Nagao M, Oh T, Mori T, Hsohino Y, Miura Y. Synthesis of Glycopolymers Carrying 3’-Sialyllactose for Suppressing Inflammatory Reaction via Siglec-E. CHEM LETT 2022. [DOI: 10.1246/cl.210740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Takato Ishida
- Department of Chemical Engineering, Kyushu University, 744, Motooka, Nishi-ku, Fukuoka 819-0395
| | - Masanori Nagao
- Department of Chemical Engineering, Kyushu University, 744, Motooka, Nishi-ku, Fukuoka 819-0395
| | - Takahiro Oh
- Department of Chemical Engineering, Kyushu University, 744, Motooka, Nishi-ku, Fukuoka 819-0395
| | - Takeshi Mori
- Department of Applied Chemistry, Kyushu University, 744, Motooka, Nishi-ku, Fukuoka 819-0395
| | - Yu Hsohino
- Department of Chemical Engineering, Kyushu University, 744, Motooka, Nishi-ku, Fukuoka 819-0395
| | - Yoshiko Miura
- Department of Chemical Engineering, Kyushu University, 744, Motooka, Nishi-ku, Fukuoka 819-0395
| |
Collapse
|
21
|
van Houtum EJH, Büll C, Cornelissen LAM, Adema GJ. Siglec Signaling in the Tumor Microenvironment. Front Immunol 2021; 12:790317. [PMID: 34966391 PMCID: PMC8710542 DOI: 10.3389/fimmu.2021.790317] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are a family of receptors that recognize sialoglycans - sialic acid containing glycans that are abundantly present on cell membranes. Siglecs are expressed on most immune cells and can modulate their activity and function. The majority of Siglecs contains immune inhibitory motifs comparable to the immune checkpoint receptor PD-1. In the tumor microenvironment (TME), signaling through the Siglec-sialoglycan axis appears to be enhanced through multiple mechanisms favoring tumor immune evasion similar to the PD-1/PD-L1 signaling pathway. Siglec expression on tumor-infiltrating immune cells appears increased in the immune suppressive microenvironment. At the same time, enhanced Siglec ligand expression has been reported for several tumor types as a result of aberrant glycosylation, glycan modifications, and the increased expression of sialoglycans on proteins and lipids. Siglec signaling has been identified as important regulator of anti-tumor immunity in the TME, but the key factors contributing to Siglec activation by tumor-associated sialoglycans are diverse and poorly defined. Among others, Siglec activation and signaling are co-determined by their expression levels, cell surface distribution, and their binding preferences for cis- and trans-ligands in the TME. Siglec binding preference are co-determined by the nature of the proteins/lipids to which the sialoglycans are attached and the multivalency of the interaction. Here, we review the current understanding and emerging conditions and factors involved in Siglec signaling in the TME and identify current knowledge gaps that exist in the field.
Collapse
Affiliation(s)
- Eline J. H. van Houtum
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Christian Büll
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, Netherlands
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lenneke A. M. Cornelissen
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gosse J. Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
22
|
Jung J, Enterina JR, Bui DT, Mozaneh F, Lin PH, Nitin, Kuo CW, Rodrigues E, Bhattacherjee A, Raeisimakiani P, Daskhan GC, St. Laurent CD, Khoo KH, Mahal LK, Zandberg WF, Huang X, Klassen JS, Macauley MS. Carbohydrate Sulfation As a Mechanism for Fine-Tuning Siglec Ligands. ACS Chem Biol 2021; 16:2673-2689. [PMID: 34661385 DOI: 10.1021/acschembio.1c00501] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The immunomodulatory family of Siglecs recognizes sialic acid-containing glycans as "self", which is exploited in cancer for immune evasion. The biochemical nature of Siglec ligands remains incompletely understood, with emerging evidence suggesting the importance of carbohydrate sulfation. Here, we investigate how specific sulfate modifications affect Siglec ligands by overexpressing eight carbohydrate sulfotransferases (CHSTs) in five cell lines. Overexpression of three CHSTs─CHST1, CHST2, or CHST4─significantly enhance the binding of numerous Siglecs. Unexpectedly, two other CHSTs (Gal3ST2 and Gal3ST3) diminish Siglec binding, suggesting a new mode to modulate Siglec ligands via sulfation. Results are cell type dependent, indicating that the context in which sulfated glycans are presented is important. Moreover, a pharmacological blockade of N- and O-glycan maturation reveals a cell-type-specific pattern of importance for either class of glycan. Production of a highly homogeneous Siglec-3 (CD33) fragment enabled a mass-spectrometry-based binding assay to determine ≥8-fold and ≥2-fold enhanced affinity for Neu5Acα2-3(6-O-sulfo)Galβ1-4GlcNAc and Neu5Acα2-3Galβ1-4(6-O-sulfo)GlcNAc, respectively, over Neu5Acα2-3Galβ1-4GlcNAc. CD33 shows significant additivity in affinity (≥28-fold) for the disulfated ligand, Neu5Acα2-3(6-O-sulfo)Galβ1-4(6-O-sulfo)GlcNAc. Moreover, joint overexpression of CHST1 with CHST2 in cells greatly enhanced the binding of CD33 and several other Siglecs. Finally, we reveal that CHST1 is upregulated in numerous cancers, correlating with poorer survival rates and sodium chlorate sensitivity for the binding of Siglecs to cancer cell lines. These results provide new insights into carbohydrate sulfation as a general mechanism for tuning Siglec ligands on cells, including in cancer.
Collapse
Affiliation(s)
- Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Jhon R. Enterina
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G 2J7, Canada
| | - Duong T. Bui
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Fahima Mozaneh
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Po-Han Lin
- Departments of Chemistry and Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Nitin
- Department of Chemistry, The University of British Columbia, Kelowna, V1V 1V7, Canada
| | - Chu-Wei Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Emily Rodrigues
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | | | | | - Gour C. Daskhan
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | | | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Lara K. Mahal
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Wesley F. Zandberg
- Department of Chemistry, The University of British Columbia, Kelowna, V1V 1V7, Canada
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - John S. Klassen
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Matthew S. Macauley
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G 2J7, Canada
| |
Collapse
|
23
|
Lünemann JD, von Gunten S, Neumann H. Targeting sialylation to treat central nervous system diseases. Trends Pharmacol Sci 2021; 42:998-1008. [PMID: 34607695 DOI: 10.1016/j.tips.2021.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 02/03/2023]
Abstract
Sialic acid-binding immunoglobulin-type lectins (SIGLECs) are membrane receptors that are preferentially expressed on immune cells and recognize sialylated proteins, lipids, and RNA. Sialic acids and signaling through SIGLECs are increasingly recognized for their essential roles in immune system homeostasis as well as nervous system development, plasticity, and repair. Dysregulated sialylation and SIGLEC dysfunctions contribute to several chronic diseases of the central nervous system (CNS) in which current therapeutic options are very limited. While only a few therapies targeting SIGLECs are currently being tested in clinical trials, the area emerged as one of the most dynamic and active fields in glycobiology and drug development. This review highlights recent insights into sialic acid and SIGLEC function in CNS pathologies and illustrates opportunities and challenges for the development of sialic acid-based and SIGLEC-targeted therapies for neurological diseases.
Collapse
Affiliation(s)
- Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| | | | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
24
|
Hong S, Yu C, Rodrigues E, Shi Y, Chen H, Wang P, Chapla DG, Gao T, Zhuang R, Moremen KW, Paulson JC, Macauley MS, Wu P. Modulation of Siglec-7 Signaling Via In Situ-Created High-Affinity cis-Ligands. ACS CENTRAL SCIENCE 2021; 7:1338-1346. [PMID: 34471678 PMCID: PMC8393205 DOI: 10.1021/acscentsci.1c00064] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Indexed: 05/04/2023]
Abstract
Sialic acid-binding immunoglobulin-like lectins, also known as Siglecs, have recently been designated as glyco-immune checkpoints. Through their interactions with sialylated glycan ligands overexpressed on tumor cells, inhibitory Siglecs on innate and adaptive immune cells modulate signaling cascades to restrain anti-tumor immune responses. However, the elucidation of the mechanisms underlying these processes is just beginning. We find that when human natural killer (NK) cells attack tumor cells, glycan remodeling occurs on the target cells at the immunological synapse. This remodeling occurs through both the transfer of sialylated glycans from NK cells to target tumor cells and the accumulation of de novo synthesized sialosides on the tumor cells. The functionalization of NK cells with a high-affinity ligand of Siglec-7 leads to multifaceted consequences in modulating a Siglec-7-regulated NK-activation. At high levels of ligand, an enzymatically added Siglec-7 ligand suppresses NK cytotoxicity through the recruitment of Siglec-7 to an immune synapse, whereas at low levels of ligand an enzymatically added Siglec-7 ligand triggers the release of Siglec-7 from the cell surface into the culture medium, preventing a Siglec-7-mediated inhibition of NK cytotoxicity. These results suggest that a glycan engineering of NK cells may provide a means to boost NK effector functions for related applications.
Collapse
Affiliation(s)
- Senlian Hong
- State
Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology
Center, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chenhua Yu
- Department of Molecular Medicine, Department of Immunology
and Microbiology, The Scripps Research Institute, La Jolla 92037, California, United States
- Tianjin
Medical University Cancer Institute and Hospital, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin 300060, China
| | - Emily Rodrigues
- Department of Chemistry, Department of Medical
Microbiology and Immunology, University
of Alberta, 11227 Saskatchewan Drive NW, Edmonton AB T6G 2G2, Alberta, Canada
| | - Yujie Shi
- Department of Molecular Medicine, Department of Immunology
and Microbiology, The Scripps Research Institute, La Jolla 92037, California, United States
| | - Hongmin Chen
- State
Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology
Center, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Peng Wang
- Department of Molecular Medicine, Department of Immunology
and Microbiology, The Scripps Research Institute, La Jolla 92037, California, United States
| | - Digantkumar G. Chapla
- Complex
Carbohydrate Research Center, University
of Georgia, Athens, Georgia 30602, United States
| | - Tao Gao
- State
Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology
Center, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ruoxuan Zhuang
- State
Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology
Center, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Kelley W. Moremen
- Complex
Carbohydrate Research Center, University
of Georgia, Athens, Georgia 30602, United States
| | - James C. Paulson
- Department of Molecular Medicine, Department of Immunology
and Microbiology, The Scripps Research Institute, La Jolla 92037, California, United States
| | - Matthew S. Macauley
- Department of Chemistry, Department of Medical
Microbiology and Immunology, University
of Alberta, 11227 Saskatchewan Drive NW, Edmonton AB T6G 2G2, Alberta, Canada
| | - Peng Wu
- Department of Molecular Medicine, Department of Immunology
and Microbiology, The Scripps Research Institute, La Jolla 92037, California, United States
| |
Collapse
|
25
|
Dhar C, Sasmal A, Diaz S, Verhagen A, Yu H, Li W, Chen X, Varki A. Are sialic acids involved in COVID-19 pathogenesis? Glycobiology 2021; 31:1068-1071. [PMID: 34192318 PMCID: PMC8344891 DOI: 10.1093/glycob/cwab063] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Chirag Dhar
- Departments of Medicine and Cellular and Molecular Medicine, UC San Diego School of Medicine, La Jolla, CA.,Glycobiology Research and Training Center (GRTC), UC San Diego, La Jolla, CA
| | - Aniruddha Sasmal
- Departments of Medicine and Cellular and Molecular Medicine, UC San Diego School of Medicine, La Jolla, CA.,Glycobiology Research and Training Center (GRTC), UC San Diego, La Jolla, CA
| | - Sandra Diaz
- Departments of Medicine and Cellular and Molecular Medicine, UC San Diego School of Medicine, La Jolla, CA.,Glycobiology Research and Training Center (GRTC), UC San Diego, La Jolla, CA
| | - Andrea Verhagen
- Departments of Medicine and Cellular and Molecular Medicine, UC San Diego School of Medicine, La Jolla, CA.,Glycobiology Research and Training Center (GRTC), UC San Diego, La Jolla, CA
| | - Hai Yu
- Department of Chemistry, UC Davis, Davis, CA
| | - Wanqing Li
- Department of Chemistry, UC Davis, Davis, CA
| | - Xi Chen
- Department of Chemistry, UC Davis, Davis, CA
| | - Ajit Varki
- Departments of Medicine and Cellular and Molecular Medicine, UC San Diego School of Medicine, La Jolla, CA.,Glycobiology Research and Training Center (GRTC), UC San Diego, La Jolla, CA
| |
Collapse
|
26
|
Alborzian Deh Sheikh A, Akatsu C, Abdu-Allah HHM, Suganuma Y, Imamura A, Ando H, Takematsu H, Ishida H, Tsubata T. The Protein Tyrosine Phosphatase SHP-1 (PTPN6) but Not CD45 (PTPRC) Is Essential for the Ligand-Mediated Regulation of CD22 in BCR-Ligated B Cells. THE JOURNAL OF IMMUNOLOGY 2021; 206:2544-2551. [PMID: 33990399 DOI: 10.4049/jimmunol.2100109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/01/2021] [Indexed: 11/19/2022]
Abstract
CD22 is an inhibitory B cell coreceptor that regulates B cell development and activation by downregulating BCR signaling through activation of SH2-containing protein tyrosine phosphatase-1 (SHP-1). CD22 recognizes α2,6 sialic acid as a specific ligand and interacts with α2,6 sialic acid-containing membrane molecules, such as CD45, IgM, and CD22, expressed on the same cell. Functional regulation of CD22 by these endogenous ligands enhances BCR ligation-induced signaling and is essential for normal B cell responses to Ags. In this study, we demonstrate that CD45 plays a crucial role in CD22-mediated inhibition of BCR ligation-induced signaling. However, disruption of ligand binding of CD22 enhances CD22 phosphorylation, a process required for CD22-mediated signal inhibition, upon BCR ligation in CD45-/- as well as wild-type mouse B cells but not in mouse B cells expressing a loss-of-function mutant of SHP-1. This result indicates that SHP-1 but not CD45 is required for ligand-mediated regulation of CD22. We further demonstrate that CD22 is a substrate of SHP-1, suggesting that SHP-1 recruited to CD22 dephosphorylates nearby CD22 as well as other substrates. CD22 dephosphorylation by SHP-1 appears to be augmented by homotypic CD22 clustering mediated by recognition of CD22 as a ligand of CD22 because CD22 clustering increases the number of nearby CD22. Our results suggest that CD22 but not CD45 is an endogenous ligand of CD22 that enhances BCR ligation-induced signaling through SHP-1-mediated dephosphorylation of CD22 in CD22 clusters.
Collapse
Affiliation(s)
- Amin Alborzian Deh Sheikh
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chizuru Akatsu
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Yuki Suganuma
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Akihiro Imamura
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Hiromune Ando
- Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, Gifu, Japan.,Institute for Glyco-core Research, Gifu University, Gifu, Japan; and
| | - Hiromu Takematsu
- Faculty of Medical Technology, Fujita Health University, Toyoake, Aichi, Japan
| | - Hideharu Ishida
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, Gifu, Japan
| | - Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan;
| |
Collapse
|
27
|
Bordron A, Morel M, Bagacean C, Dueymes M, Pochard P, Harduin-Lepers A, Jamin C, Pers JO. Hyposialylation Must Be Considered to Develop Future Therapies in Autoimmune Diseases. Int J Mol Sci 2021; 22:ijms22073402. [PMID: 33810246 PMCID: PMC8036829 DOI: 10.3390/ijms22073402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune disease development depends on multiple factors, including genetic and environmental. Abnormalities such as sialylation levels and/or quality have been recently highlighted. The adjunction of sialic acid at the terminal end of glycoproteins and glycolipids is essential for distinguishing between self and non-self-antigens and the control of pro- or anti-inflammatory immune reactions. In autoimmunity, hyposialylation is responsible for chronic inflammation, the anarchic activation of the immune system and organ lesions. A detailed characterization of this mechanism is a key element for improving the understanding of these diseases and the development of innovative therapies. This review focuses on the impact of sialylation in autoimmunity in order to determine future treatments based on the regulation of hyposialylation.
Collapse
Affiliation(s)
- Anne Bordron
- Univ Brest, Inserm, LBAI, UMR1227 Brest, France; (M.M.); (C.B.); (M.D.); (C.J.); (J.-O.P.)
- Correspondence:
| | - Marie Morel
- Univ Brest, Inserm, LBAI, UMR1227 Brest, France; (M.M.); (C.B.); (M.D.); (C.J.); (J.-O.P.)
| | - Cristina Bagacean
- Univ Brest, Inserm, LBAI, UMR1227 Brest, France; (M.M.); (C.B.); (M.D.); (C.J.); (J.-O.P.)
- CHU de Brest, Laboratory of Immunolgy, 29200 Brest, France;
| | - Maryvonne Dueymes
- Univ Brest, Inserm, LBAI, UMR1227 Brest, France; (M.M.); (C.B.); (M.D.); (C.J.); (J.-O.P.)
- CHU de Brest, Laboratory of Immunolgy, 29200 Brest, France;
| | - Pierre Pochard
- CHU de Brest, Laboratory of Immunolgy, 29200 Brest, France;
| | - Anne Harduin-Lepers
- Univ. Lille, CNRS UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, 59000 Lille, France;
| | - Christophe Jamin
- Univ Brest, Inserm, LBAI, UMR1227 Brest, France; (M.M.); (C.B.); (M.D.); (C.J.); (J.-O.P.)
- CHU de Brest, Laboratory of Immunolgy, 29200 Brest, France;
| | - Jacques-Olivier Pers
- Univ Brest, Inserm, LBAI, UMR1227 Brest, France; (M.M.); (C.B.); (M.D.); (C.J.); (J.-O.P.)
- CHU de Brest, Laboratory of Immunolgy, 29200 Brest, France;
| |
Collapse
|
28
|
Yoshimura A, Asahina Y, Chang LY, Angata T, Tanaka H, Kitajima K, Sato C. Identification and functional characterization of a Siglec-7 counter-receptor on K562 cells. J Biol Chem 2021; 296:100477. [PMID: 33640457 PMCID: PMC8040268 DOI: 10.1016/j.jbc.2021.100477] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Sialic acid (Sia)-binding immunoglobulin-like lectin 7 (Siglec-7) is an inhibitory receptor primarily expressed on natural killer (NK) cells and monocytes. Siglec-7 is known to negatively regulate the innate immune system through Sia binding to distinguish self and nonself; however, a counter-receptor bearing its natural ligand remains largely unclear. Here, we identified a counter-receptor of Siglec-7 using K562 hematopoietic carcinoma cells presenting cell surface ligands for Siglec-7. We affinity-purified the ligands using Fc-ligated recombinant Siglec-7 and diSia-dextran polymer, a strong inhibitor for Siglec-7. We then confirmed the counter-receptor for Siglec-7 as leukosialin (CD43) through mass spectrometry, immunoprecipitation, and proximity labeling. Additionally, we demonstrated that the cytotoxicity of NK cells toward K562 cells was suppressed by overexpression of leukosialin in a Siglec-7-dependent manner. Taken together, our data suggest that leukosialin on K562 is a counter-receptor for Siglec-7 on NK cells and that a cluster of the Sia-containing glycan epitope on leukosialin is key as trans-ligand for unmasking the cis-ligand.
Collapse
Affiliation(s)
- Atsushi Yoshimura
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, Japan
| | - Yuki Asahina
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, Japan
| | - Lan-Yi Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Takashi Angata
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hiroshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Meguro, Tokyo, Japan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, Japan; Integrated Glyco-Biomedical Research Center (iGMed), Nagoya University, Chikusa, Nagoya, Japan; Institute for Glyco-Core Research (iGCORE), Nagoya University, Chikusa, Nagoya, Japan
| | - Chihiro Sato
- Bioscience and Biotechnology Center, Nagoya University, Chikusa, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, Japan; Integrated Glyco-Biomedical Research Center (iGMed), Nagoya University, Chikusa, Nagoya, Japan; Institute for Glyco-Core Research (iGCORE), Nagoya University, Chikusa, Nagoya, Japan.
| |
Collapse
|
29
|
Rosenstock P, Kaufmann T. Sialic Acids and Their Influence on Human NK Cell Function. Cells 2021; 10:263. [PMID: 33572710 PMCID: PMC7911748 DOI: 10.3390/cells10020263] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/14/2022] Open
Abstract
Sialic acids are sugars with a nine-carbon backbone, present on the surface of all cells in humans, including immune cells and their target cells, with various functions. Natural Killer (NK) cells are cells of the innate immune system, capable of killing virus-infected and tumor cells. Sialic acids can influence the interaction of NK cells with potential targets in several ways. Different NK cell receptors can bind sialic acids, leading to NK cell inhibition or activation. Moreover, NK cells have sialic acids on their surface, which can regulate receptor abundance and activity. This review is focused on how sialic acids on NK cells and their target cells are involved in NK cell function.
Collapse
Affiliation(s)
- Philip Rosenstock
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Hollystr. 1, D-06114 Halle/Saale, Germany;
| | | |
Collapse
|
30
|
Bachsais M, Salti S, Zaoui K, Hassan GS, Aoudjit F, Mourad W. CD154 inhibits death of T cells via a Cis interaction with the α5β1 integrin. PLoS One 2020; 15:e0235753. [PMID: 32745080 PMCID: PMC7398495 DOI: 10.1371/journal.pone.0235753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/22/2020] [Indexed: 12/30/2022] Open
Abstract
CD154 plays a major role in the pathogenesis of several autoimmune and inflammatory diseases. In addition to CD40, soluble CD154 (sCD154) binds to other receptors namely αIIbβ3, αMβ2, α5β1 and αvβ3 integrins. We have previously reported that binding of sCD154 to α5β1 integrin expressed on several human T cell lines is capable of inhibiting Fas-induced cell death. In the current study, we show that such effect of the sCD154/α5β1 interaction is not restricted to the cell death response induced by Fas but could also be exhibited toward other death signals such as TRAIL and TNF- α. We also demonstrate that sCD154 is capable of inhibiting Fas-mediated death of human activated T cells, more importantly of CD4+ than CD8+ T ones. Our data also show that membrane-bound CD154 and α5β1 integrin expressed on the surface of distinct cells failed to influence cell death responses. However, when membrane-bound CD154 and α5β1 are expressed on the surface of same cell, their interaction was capable of down regulating cell death. CD154 was shown to co-localize with the α5β1 integrin on the surface of these cells. These data strongly suggest a cis-type of interaction between CD154 and α5β1 when both are expressed on the same cell surface, rather than a trans-interaction which usually implicates the ligand and its receptor each expressed on the surface of a distinct cell. Taken together, these findings add to the list of roles through which CD154 is contributing to the pathogenesis of autoimmune-inflammatory diseases, i.e. by protecting T cells from death and enhancing their survival.
Collapse
Affiliation(s)
- Meriem Bachsais
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Suzanne Salti
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Kossay Zaoui
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Ghada S. Hassan
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
| | - Fawzi Aoudjit
- Centre de recherche du CHU de Québec-Université Laval, Québec, Québec, Canada
| | - Walid Mourad
- Laboratoire d’Immunologie Cellulaire et Moléculaire, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
31
|
Kong L, Wu L, Guo Z, Mu L, Yang Y, Bian X, Li B, Pan X, Fu S, Ye J. A Siglec-1-like lectin from Nile tilapia (Oreochromis niloticus) possesses functions of agglutination and mediation of macrophage phagocytic activity. FISH & SHELLFISH IMMUNOLOGY 2020; 102:203-210. [PMID: 32330627 DOI: 10.1016/j.fsi.2020.04.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 06/11/2023]
Abstract
Siglec-1, one of the sialic acid-binding immunoglobulin-type lectins, is closely related to the recognition of host-pathogen and cell-cell interactions in the adaptive and innate immune systems. In this communication, a Siglec-1-like gene (OnSiglec-1-like) from Nile tilapia (Oreochromis niloticus) was analyzed. Relative expression revealed that the OnSiglec-1-like was expressed in all tested tissues, and the highest expression was found in the anterior kidney. Upon Streptococcus agalactiae (S. agalactiae) infection, the expression of OnSiglec-1-like was up-regulated in anterior kidney and spleen significantly in vivo. Additionally, the same phenomenon was observed in anterior kidney leukocytes upon LPS and S. agalactiae challenges as well in vitro. Western-blotting and ELISA analyses revealed that recombinant OnSiglec-1-like protein possessed high binding activity to LTA, LPS and S. agalactiae. Further, the recombinant OnSiglec-1-like was able to agglutinate S. agalactiae. Moreover, with the digestion of specific sialidase, the phagocytic ability of macrophages to S. agalactiae was greatly enhanced. Taken together, these results indicated that the Siglec-1-like possesses conserved functions of agglutination and promotion of macrophage phagocytic activity in Nile tilapia.
Collapse
Affiliation(s)
- Linghe Kong
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Liting Wu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Zheng Guo
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China.
| | - Liangliang Mu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Yanjian Yang
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Xia Bian
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Bingxi Li
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Xunbin Pan
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Shengli Fu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Jianmin Ye
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China.
| |
Collapse
|
32
|
Sialylation of Human Natural Killer (NK) Cells is Regulated by IL-2. J Clin Med 2020; 9:jcm9061816. [PMID: 32545211 PMCID: PMC7356531 DOI: 10.3390/jcm9061816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/28/2022] Open
Abstract
Sialic acids are terminal sugars on the cell surface that are found on all cell types including immune cells like natural killer (NK) cells. The attachment of sialic acids to different glycan structures is catalyzed by sialyltransferases in the Golgi. However, the expression pattern of sialyltransferases in NK cells and their expression after activation has not yet been analyzed. Therefore, the present study determines which sialyltransferases are expressed in human NK cells and if activation with IL-2 changes the sialylation of NK cells. The expression of sialyltransferases was analyzed in the three human NK cell lines NK-92, NKL, KHYG-1 and primary NK cells. NK-92 cells were cultured in the absence or presence of IL-2, and changes in the sialyltransferase expression were measured by qPCR. Furthermore, specific sialylation was investigated by flow cytometry. In addition, polySia and NCAM were measured by Western blot analyses. IL-2 leads to a reduced expression of ST8SIA1, ST6GAL1 and ST3GAL1. α-2,3-Sialylation remained unchanged, while α-2,6-sialylation was increased after IL-2 stimulation. Moreover, an increase in the amount of NCAM and polySia was observed in IL-2-activated NK cells, whereas GD3 ganglioside was decreased. In this study, all sialyltransferases that were expressed in NK cells could be identified. IL-2 regulates the expression of some sialyltransferases and leads to changes in the sialylation of NK cells.
Collapse
|
33
|
Bandala-Sanchez E, Bediaga NG, Naselli G, Neale AM, Harrison LC. Siglec-10 expression is up-regulated in activated human CD4 + T cells. Hum Immunol 2020; 81:101-104. [PMID: 32046870 DOI: 10.1016/j.humimm.2020.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/09/2020] [Accepted: 01/24/2020] [Indexed: 02/05/2023]
Abstract
Most sialic acid-binding immunoglobulin-like lectins (Siglecs) suppress immune cell function but are expressed at low levels on human T cells. We found that soluble CD52 inhibited T cell signalling by ligating Siglec-10, but the presence of Siglec-10 on human T cells has been questioned. To address this concern, we examined the expression of Siglec-10 at the RNA and protein level in human CD4+ T cells. Analysis by RNAseq, qPCR and flow cytometry demonstrated that, in contrast to other Siglecs, after activation of CD4+ T cells Siglec-10 was selectively upregulated in a subset of cells also high for CD52 expression. This observation is consistent with a homeostatic role for Siglec-10 in human CD4+ T cells.
Collapse
Affiliation(s)
- E Bandala-Sanchez
- Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville 3052, Victoria, Australia
| | - N G Bediaga
- Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville 3052, Victoria, Australia
| | - G Naselli
- Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville 3052, Victoria, Australia
| | - A M Neale
- Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville 3052, Victoria, Australia
| | - L C Harrison
- Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville 3052, Victoria, Australia.
| |
Collapse
|
34
|
Abstract
Sialic acid-binding immunoglobulin-type lectins (Siglecs) are expressed on the majority of white blood cells of the immune system and play critical roles in immune cell signaling. Through recognition of sialic acid-containing glycans as ligands, they help the immune system distinguish between self and nonself. Because of their restricted cell type expression and roles as checkpoints in immune cell responses in human diseases such as cancer, asthma, allergy, neurodegeneration, and autoimmune diseases they have gained attention as targets for therapeutic interventions. In this review we describe the Siglec family, its roles in regulation of immune cell signaling, current efforts to define its roles in disease processes, and approaches to target Siglecs for treatment of human disease.
Collapse
Affiliation(s)
- Shiteng Duan
- Departments of Molecular Medicine, and Immunology and Microbiology, Scripps Research, La Jolla, California 92037, USA;
| | - James C Paulson
- Departments of Molecular Medicine, and Immunology and Microbiology, Scripps Research, La Jolla, California 92037, USA;
| |
Collapse
|
35
|
Suematsu R, Miyamoto T, Saijo S, Yamasaki S, Tada Y, Yoshida H, Miyake Y. Identification of lipophilic ligands of Siglec5 and -14 that modulate innate immune responses. J Biol Chem 2019; 294:16776-16788. [PMID: 31551352 DOI: 10.1074/jbc.ra119.009835] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/02/2019] [Indexed: 01/22/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are a family of cell-surface immune receptors that bind to sialic acid at terminal glycan residues. Siglecs also recognize nonsialic acid ligands, many of which remain to be characterized. Here, we found that Siglec5 and Siglec14 recognize lipid compounds produced by Trichophyton, a fungal genus containing several pathogenic species. Biochemical approaches revealed that the Siglec ligands are fungal alkanes and triacylglycerols, an unexpected finding that prompted us to search for endogenous lipid ligands of Siglecs. Siglec5 weakly recognized several endogenous lipids, but the mitochondrial lipid cardiolipin and the anti-inflammatory lipid 5-palmitic acid-hydroxystearic acid exhibited potent ligand activity on Siglec5. Further, the hydrophobic stretch in the Siglec5 N terminus region was found to be required for efficient recognition of these lipids. Notably, this hydrophobic stretch was dispensable for recognition of sialic acid. Siglec5 inhibited cell activation upon ligand binding, and accordingly, the lipophilic ligands suppressed interleukin-8 (IL-8) production in Siglec5-expressing human monocytic cells. Siglec14 and Siglec5 have high sequence identity in the extracellular region, and Siglec14 also recognized the endogenous lipids. However, unlike Siglec5, Siglec14 transduces activating signals upon ligand recognition. Indeed, the endogenous lipids induced IL-8 production in Siglec14-expressing human monocytic cells. These results indicated that Siglec5 and Siglec14 can recognize lipophilic ligands that thereby modulate innate immune responses. To our knowledge, this is the first study reporting the binding of Siglecs to lipid ligands, expanding our understanding of the biological function and importance of Siglecs in the innate immunity.
Collapse
Affiliation(s)
- Rie Suematsu
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan.,Department of Rheumatology, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Tomofumi Miyamoto
- Department of Natural Products Chemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shinobu Saijo
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | - Sho Yamasaki
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871, Japan
| | - Yoshifumi Tada
- Department of Rheumatology, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Yasunobu Miyake
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| |
Collapse
|
36
|
Enterina JR, Jung J, Macauley MS. Coordinated roles for glycans in regulating the inhibitory function of CD22 on B cells. Biomed J 2019; 42:218-232. [PMID: 31627864 PMCID: PMC6818156 DOI: 10.1016/j.bj.2019.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/19/2019] [Accepted: 07/26/2019] [Indexed: 01/17/2023] Open
Abstract
CD22 is an inhibitory B cell co-receptor that recognizes sialic acid-containing glycoconjugates as ligands. Interactions with its glycan ligands are key to regulating the ability of CD22 to modulate B cell function, the most widely explored of which is antagonizing B cell receptor (BCR) signaling. Most importantly, interactions of CD22 with ligands on the same cell (cis) control the organization of CD22 on the cell surface, which minimizes co-localization with the BCR. In contrast with the modest ability of CD22 to intrinsically dampen BCR signaling, glycan ligands presented on another cell (trans) along with an antigen drawn CD22 and the BCR together within an immunological synapse, strongly inhibiting BCR signaling. New concepts are emerging for how CD22 controls B cell function, such as changes in glycosylation at different stages of B cell differentiation, specifically on GC B cells. Related to these changes, new players, such galectin-9, have been discovered that regulate cell surface nanoclusters of CD22. Roles of glycan ligands in controlling CD22 are the primary focus of this review as we highlight the ability of CD22 to modulate B cell function.
Collapse
Affiliation(s)
- Jhon R Enterina
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Matthew S Macauley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada; Department of Chemistry, University of Alberta, Edmonton, Canada.
| |
Collapse
|
37
|
Santegoets KCM, Gielen PR, Büll C, Schulte BM, Kers-Rebel ED, Küsters B, Bossman SAJFH, Ter Laan M, Wesseling P, Adema GJ. Expression profiling of immune inhibitory Siglecs and their ligands in patients with glioma. Cancer Immunol Immunother 2019; 68:937-949. [PMID: 30953118 PMCID: PMC6529385 DOI: 10.1007/s00262-019-02332-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 03/24/2019] [Indexed: 12/22/2022]
Abstract
Gliomas appear to be highly immunosuppressive tumors, with a strong myeloid component. This includes MDSCs, which are a heterogeneous, immature myeloid cell population expressing myeloid markers Siglec-3 (CD33) and CD11b and lacking markers of mature myeloid cells including MHC II. Siglec-3 is a member of the sialic acid-binding immunoglobulin-like lectin (Siglec) family and has been suggested to promote MDSC expansion and suppression. Siglecs form a recently defined family of receptors with potential immunoregulatory functions but only limited insight in their expression on immune regulatory cell subsets, prompting us to investigate Siglec expression on MDSCs. We determined the expression of different Siglec family members on monocytic-MDSCs (M-MDSCs) and polymorphnuclear-MDSCs (PMN-MDSCs) from blood of glioma patients and healthy donors, as well as from patient-derived tumor material. Furthermore, we investigated the presence of sialic acid ligands for these Siglecs on MDSCs and in the glioma tumor microenvironment. Both MDSC subsets express Siglec-3, -5, -7 and -9, with higher levels of Siglec-3, -7 and -9 on M-MDSCs and higher Siglec-5 levels on PMN-MDSCs. Similar Siglec expression profiles were found on MDSCs from healthy donors. Furthermore, the presence of Siglec-5 and -9 was also confirmed on PMN-MDSCs from glioma tissue. Interestingly, freshly isolated glioma cells predominantly expressed sialic acid ligands for Siglec-7 and -9, which was confirmed in situ. In conclusion, our data show a distinct Siglec expression profile for M- and PMN-MDSCs and propose possible sialic acid-Siglec interactions between glioma cells and MDSCs in the tumor microenvironment.
Collapse
Affiliation(s)
- Kim C M Santegoets
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands
| | - Paul R Gielen
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands
| | - Christian Büll
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands
| | - Barbara M Schulte
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands
| | - Esther D Kers-Rebel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands
| | - Benno Küsters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sandra A J F H Bossman
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Prinses Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
38
|
Zhang T, de Waard AA, Wuhrer M, Spaapen RM. The Role of Glycosphingolipids in Immune Cell Functions. Front Immunol 2019; 10:90. [PMID: 30761148 PMCID: PMC6361815 DOI: 10.3389/fimmu.2019.00090] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/14/2019] [Indexed: 01/06/2023] Open
Abstract
Glycosphingolipids (GSLs) exhibit a variety of functions in cellular differentiation and interaction. Also, they are known to play a role as receptors in pathogen invasion. A less well-explored feature is the role of GSLs in immune cell function which is the subject of this review article. Here we summarize knowledge on GSL expression patterns in different immune cells. We review the changes in GSL expression during immune cell development and differentiation, maturation, and activation. Furthermore, we review how immune cell GSLs impact membrane organization, molecular signaling, and trans-interactions in cellular cross-talk. Another aspect covered is the role of GSLs as targets of antibody-based immunity in cancer. We expect that recent advances in analytical and genome editing technologies will help in the coming years to further our knowledge on the role of GSLs as modulators of immune cell function.
Collapse
Affiliation(s)
- Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Antonius A de Waard
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
39
|
Meyer SJ, Linder AT, Brandl C, Nitschke L. B Cell Siglecs-News on Signaling and Its Interplay With Ligand Binding. Front Immunol 2018; 9:2820. [PMID: 30559744 PMCID: PMC6286995 DOI: 10.3389/fimmu.2018.02820] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/15/2018] [Indexed: 12/11/2022] Open
Abstract
CD22 and Siglec-G are members of the Siglec family. Both are inhibitory co-receptors on the surface of B cells and inhibit B-cell receptor induced signaling, characterized by inhibition of the calcium mobilization and cellular activation. CD22 functions predominantly as an inhibitor on conventional B cells, while Siglec-G is an important inhibitor on the B1a-cell subset. These two B-cell Siglecs do not only inhibit initial signaling, but also have an important function in preventing autoimmunity, as double deficient mice develop a lupus-like phenotype with age. Siglecs are characterized by their conserved ability to bind terminal sialic acid of glycans on the cell surface, which is important to regulate the inhibitory role of Siglecs. While CD22 binds α2,6-linked sialic acids, Siglec-G can bind both α2,6-linked and α2,3-linked sialic acids. Interestingly, ligand binding is differentially regulating the ability of CD22 and Siglec-G to control B-cell activation. Within the last years, quite a few studies focused on the different functions of B-cell Siglecs and the interplay of ligand binding and signal inhibition. This review summarizes the role of CD22 and Siglec-G in regulating B-cell receptor signaling, membrane distribution with the importance of ligand binding, preventing autoimmunity and the role of CD22 beyond the naïve B-cell stage. Additionally, this review article features the long time discussed interaction between CD45 and CD22 with highlighting recent data, as well as the interplay between CD22 and Galectin-9 and its influence on B-cell receptor signaling. Moreover, therapeutical approaches targeting human CD22 will be elucidated.
Collapse
Affiliation(s)
- Sarah J Meyer
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Alexandra T Linder
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Carolin Brandl
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| |
Collapse
|
40
|
Giovannone N, Liang J, Antonopoulos A, Geddes Sweeney J, King SL, Pochebit SM, Bhattacharyya N, Lee GS, Dell A, Widlund HR, Haslam SM, Dimitroff CJ. Galectin-9 suppresses B cell receptor signaling and is regulated by I-branching of N-glycans. Nat Commun 2018; 9:3287. [PMID: 30120234 PMCID: PMC6098069 DOI: 10.1038/s41467-018-05770-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/26/2018] [Indexed: 12/29/2022] Open
Abstract
Leukocytes are coated with a layer of heterogeneous carbohydrates (glycans) that modulate immune function, in part by governing specific interactions with glycan-binding proteins (lectins). Although nearly all membrane proteins bear glycans, the identity and function of most of these sugars on leukocytes remain unexplored. Here, we characterize the N-glycan repertoire (N-glycome) of human tonsillar B cells. We observe that naive and memory B cells express an N-glycan repertoire conferring strong binding to the immunoregulatory lectin galectin-9 (Gal-9). Germinal center B cells, by contrast, show sharply diminished binding to Gal-9 due to upregulation of I-branched N-glycans, catalyzed by the β1,6-N-acetylglucosaminyltransferase GCNT2. Functionally, we find that Gal-9 is autologously produced by naive B cells, binds CD45, suppresses calcium signaling via a Lyn-CD22-SHP-1 dependent mechanism, and blunts B cell activation. Thus, our findings suggest Gal-9 intrinsically regulates B cell activation and may differentially modulate BCR signaling at steady state and within germinal centers.
Collapse
Affiliation(s)
- N Giovannone
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - J Liang
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - A Antonopoulos
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - J Geddes Sweeney
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - S L King
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - S M Pochebit
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - N Bhattacharyya
- Department of Surgery, Division of Otolaryngology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA, 02115, USA
| | - G S Lee
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA, 02115, USA
| | - A Dell
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - H R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - S M Haslam
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| | - C J Dimitroff
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
41
|
Matsubara N, Imamura A, Yonemizu T, Akatsu C, Yang H, Ueki A, Watanabe N, Abdu-Allah H, Numoto N, Takematsu H, Kitazume S, Tedder TF, Marth JD, Ito N, Ando H, Ishida H, Kiso M, Tsubata T. CD22-Binding Synthetic Sialosides Regulate B Lymphocyte Proliferation Through CD22 Ligand-Dependent and Independent Pathways, and Enhance Antibody Production in Mice. Front Immunol 2018; 9:820. [PMID: 29725338 PMCID: PMC5917077 DOI: 10.3389/fimmu.2018.00820] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 04/04/2018] [Indexed: 01/06/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are expressed in various immune cells and most of them carry signaling functions. High-affinity synthetic sialoside ligands have been developed for various Siglecs. Therapeutic potentials of the nanoparticles and compounds that contain multiple numbers of these sialosides and other reagents such as toxins and antigens have been demonstrated. However, whether immune responses can be regulated by monomeric sialoside ligands has not yet been known. CD22 (also known as Siglec-2) is an inhibitory molecule preferentially expressed in B lymphocytes (B cells) and is constitutively bound and functionally regulated by α2,6 sialic acids expressed on the same cell (cis-ligands). Here, we developed synthetic sialosides GSC718 and GSC839 that bind to CD22 with high affinity (IC50 ~100 nM), and inhibit ligand binding of CD22. When B cells are activated by B cell antigen receptor (BCR) ligation, both GSC718 and GSC839 downregulate proliferation of B cells, and this regulation requires both CD22 and α2,6 sialic acids. This result suggests that these sialosides regulate BCR ligation-induced B cell activation by reversing endogenous ligand-mediated regulation of CD22. By contrast, GSC718 and GSC839 augment B cell proliferation induced by TLR ligands or CD40 ligation, and this augmentation requires CD22 but not α2,6 sialic acids. Thus, these sialosides appear to enhance B cell activation by directly suppressing the inhibitory function of CD22 independently of endogenous ligand-mediated regulation. Moreover, GSC839 augments B cell proliferation that depends on both BCR ligation and CD40 ligation as is the case for in vivo B cell responses to antigens, and enhanced antibody production to the extent comparable to CpG oligonuleotides or a small amount of alum. Although these known adjuvants induce production of the inflammatory cytokines or accumulation of inflammatory cells, CD22-binding sialosides do not. Thus, synthetic sialosides that bind to CD22 with high-affinity modulate B cell activation through endogenous ligand-dependent and independent pathways, and carry an adjuvant activity without inducing inflammation.
Collapse
Affiliation(s)
- Naoko Matsubara
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiro Imamura
- Department of Applied Bio-Organic Chemistry, Gifu University, Gifu, Japan
| | - Tatsuya Yonemizu
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chizuru Akatsu
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hongrui Yang
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akiharu Ueki
- Department of Applied Bio-Organic Chemistry, Gifu University, Gifu, Japan
| | - Natsuki Watanabe
- Department of Applied Bio-Organic Chemistry, Gifu University, Gifu, Japan
| | - Hajjaj Abdu-Allah
- Department of Applied Bio-Organic Chemistry, Gifu University, Gifu, Japan
| | - Nobutaka Numoto
- Department of Structural Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiromu Takematsu
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Thomas F Tedder
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Jamey D Marth
- Center for Nanomedicine, University of California, Santa Barbara, CA, United States
| | - Nobutoshi Ito
- Department of Structural Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiromune Ando
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| | - Hideharu Ishida
- Department of Applied Bio-Organic Chemistry, Gifu University, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| | - Makoto Kiso
- Department of Applied Bio-Organic Chemistry, Gifu University, Gifu, Japan
| | - Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
42
|
Jones MB. IgG and leukocytes: Targets of immunomodulatory α2,6 sialic acids. Cell Immunol 2018; 333:58-64. [PMID: 29685495 DOI: 10.1016/j.cellimm.2018.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/30/2018] [Indexed: 12/27/2022]
Abstract
ST6Gal1 is a critical sialyltransferase enzyme that controls the addition of α2,6-linked sialic acids to the termini of glycans. Attachment of sialic acids to glycoproteins as a posttranslational modification influences cellular responses, and is a well-known modifier of immune cell behavior. ST6Gal1 activity impacts processes such as: effector functions of immunoglobulin G via Fc sialylation, hematopoietic capacity by hematopoietic stem and progenitor cell surface sialylation, and lymphocyte activation thresholds though CD22 engagement and inhibition of galectins. This review summarizes recent studies that suggest α2,6 sialylation by ST6Gal1 has an immunoregulatory effect on immune reactions.
Collapse
Affiliation(s)
- Mark B Jones
- Case Western Reserve University, School of Medicine, Department of Pathology, Cleveland, OH 44106, United States.
| |
Collapse
|
43
|
Daskhan GC, Tran HTT, Meloncelli PJ, Lowary TL, West LJ, Cairo CW. Construction of Multivalent Homo- and Heterofunctional ABO Blood Group Glycoconjugates Using a Trifunctional Linker Strategy. Bioconjug Chem 2018; 29:343-362. [PMID: 29237123 DOI: 10.1021/acs.bioconjchem.7b00679] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The design and synthesis of multivalent ligands displaying complex oligosaccharides is necessary for the development of therapeutics, diagnostics, and research tools. Here, we report an efficient conjugation strategy to prepare complex glycoconjugates with 4 copies of 1 or 2 separate glycan epitopes, providing 4-8 carbohydrate residues on a tetravalent poly(ethylene glycol) scaffold. This strategy provides complex glycoconjugates that approach the size of glycoproteins (15-18 kDa) while remaining well-defined. The synthetic strategy makes use of three orthogonal functional groups, including a reactive N-hydroxysuccinimide (NHS)-ester moiety on the linker to install the first carbohydrate epitope via reaction with an amine. A masked amine functionality on the linker is revealed after the removal of a fluorenylmethyloxycarbonyl (Fmoc)-protecting group, allowing the attachment to the NHS-activated poly(ethylene glycol) (PEG) scaffold. An azide group in the linker was then used to incorporate the second carbohydrate epitope via catalyzed alkyne-azide cycloaddition. Using a known tetravalent PEG scaffold (PDI, 1.025), we prepared homofunctional glycoconjugates that display four copies of lactose and the A-type II or the B-type II human blood group antigens. Using our trifunctional linker, we expanded this strategy to produce heterofunctional conjugates with four copies of two separate glycan epitopes. These heterofunctional conjugates included Neu5Ac, 3'-sialyllactose, or 6'-sialyllactose as a second antigen. Using an alternative strategy, we generated heterofunctional conjugates with three copies of the glycan epitope and one fluorescent group (on average) using a sequential dual-amine coupling strategy. These conjugation strategies should be easily generalized for conjugation of other complex glycans. We demonstrate that the glycan epitopes of heterofunctional conjugates engage and cluster target B-cell receptors and CD22 receptors on B cells, supporting the application of these reagents for investigating cellular response to carbohydrate antigens of the ABO blood group system.
Collapse
Affiliation(s)
- Gour Chand Daskhan
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta , Edmonton, Alberta T6G 2G2, Canada
| | - Hanh-Thuc Ton Tran
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta , Edmonton, Alberta T6G 2G2, Canada
| | - Peter J Meloncelli
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta , Edmonton, Alberta T6G 2G2, Canada
| | - Todd L Lowary
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta , Edmonton, Alberta T6G 2G2, Canada.,Canadian National Transplant Research Program, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - Lori J West
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta , Edmonton, Alberta T6G 2G2, Canada.,Department of Pediatrics, Surgery, Medical Microbiology and Immunology, and Laboratory Medicine and Pathology, Alberta Transplant Institute, University of Alberta Edmonton, Alberta T6G 2E1, Canada.,Canadian National Transplant Research Program, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - Christopher W Cairo
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta , Edmonton, Alberta T6G 2G2, Canada.,Canadian National Transplant Research Program, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
44
|
Proximity labeling of cis-ligands of CD22/Siglec-2 reveals stepwise α2,6 sialic acid-dependent and -independent interactions. Biochem Biophys Res Commun 2017; 495:854-859. [PMID: 29146181 DOI: 10.1016/j.bbrc.2017.11.086] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
Abstract
Lectins expressed on the cell surface are often bound and regulated by the membrane molecules containing the glycan ligands on the same cell (cis-ligands). However, molecular nature and function of cis-ligands are generally poorly understood partly because of weak interaction between lectins and glycan ligands. Cis-ligands are most extensively studied in CD22 (also known as Siglec-2), an inhibitory B lymphocyte receptor specifically recognizing α2,6 sialic acids. CD22, CD45 and IgM are suggested to be ligands of CD22. Here we labeled molecules in the proximity of CD22 in situ on B cell surface using biotin-tyramide. Molecules including CD22, CD45 and IgM were labeled in wild-type but not ST6GalI-/- B cells that lack α2,6 sialic acids, indicating that these molecules associate with CD22 by lectin-glycan interaction, and are therefore cis-ligands. In ST6GalI-/- B cells, these cis-ligands are located in a slightly more distance from CD22. Thus, the lectin-glycan interaction recruits cis-ligands already located in the relative proximity of CD22 through non-lectin-glycan interaction to the close proximity. Moreover, cis-ligands are labeled in Cmah-/- B cells that lack Neu5Gc preferred by mouse CD22 as efficiently as in wild-type B cells, indicating that very low affinity lectin-glycan interaction is sufficient for recruiting cis-ligands, and can be detected by proximity labeling. Thus, proximity labeling with tyramide appears to be a useful method to identify cis-ligands and to analyze their interaction with the lectins.
Collapse
|
45
|
Bednar KJ, Shanina E, Ballet R, Connors EP, Duan S, Juan J, Arlian BM, Kulis MD, Butcher EC, Fung-Leung WP, Rao TS, Paulson JC, Macauley MS. Human CD22 Inhibits Murine B Cell Receptor Activation in a Human CD22 Transgenic Mouse Model. THE JOURNAL OF IMMUNOLOGY 2017; 199:3116-3128. [PMID: 28972089 DOI: 10.4049/jimmunol.1700898] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/28/2017] [Indexed: 01/12/2023]
Abstract
CD22, a sialic acid-binding Ig-type lectin (Siglec) family member, is an inhibitory coreceptor of the BCR with established roles in health and disease. The restricted expression pattern of CD22 on B cells and most B cell lymphomas has made CD22 a therapeutic target for B cell-mediated diseases. Models to better understand how in vivo targeting of CD22 translates to human disease are needed. In this article, we report the development of a transgenic mouse expressing human CD22 (hCD22) in B cells and assess its ability to functionally substitute for murine CD22 (mCD22) for regulation of BCR signaling, Ab responses, homing, and tolerance. Expression of hCD22 on transgenic murine B cells is comparable to expression on human primary B cells, and it colocalizes with mCD22 on the cell surface. Murine B cells expressing only hCD22 have identical calcium (Ca2+) flux responses to anti-IgM as mCD22-expressing wild-type B cells. Furthermore, hCD22 transgenic mice on an mCD22-/- background have restored levels of marginal zone B cells and Ab responses compared with deficiencies observed in CD22-/- mice. Consistent with these observations, hCD22 transgenic mice develop normal humoral responses in a peanut allergy oral sensitization model. Homing of B cells to Peyer's patches was partially rescued by expression of hCD22 compared with CD22-/- B cells, although not to wild-type levels. Notably, Siglec-engaging antigenic liposomes formulated with an hCD22 ligand were shown to prevent B cell activation, increase cell death, and induce tolerance in vivo. This hCD22 transgenic mouse will be a valuable model for investigating the function of hCD22 and preclinical studies targeting hCD22.
Collapse
Affiliation(s)
- Kyle J Bednar
- Immunology Team, Janssen Research and Development, LLC, Raritan, NJ 08869.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Elena Shanina
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Romain Ballet
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305.,Palo Alto Veterans Institute for Research, Palo Alto, CA 94304.,The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | - Edward P Connors
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Shiteng Duan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.,Department of Immunology and Microbial Sciences, The Scripps Research Institute; La Jolla, CA 92037; and
| | - Joana Juan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Britni M Arlian
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.,Department of Immunology and Microbial Sciences, The Scripps Research Institute; La Jolla, CA 92037; and
| | - Michael D Kulis
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305.,Palo Alto Veterans Institute for Research, Palo Alto, CA 94304.,The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | | | - Tadimeti S Rao
- Immunology Team, Janssen Research and Development, LLC, Raritan, NJ 08869
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037; .,Department of Immunology and Microbial Sciences, The Scripps Research Institute; La Jolla, CA 92037; and
| | - Matthew S Macauley
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037;
| |
Collapse
|
46
|
Peng W, Paulson JC. CD22 Ligands on a Natural N-Glycan Scaffold Efficiently Deliver Toxins to B-Lymphoma Cells. J Am Chem Soc 2017; 139:12450-12458. [PMID: 28829594 DOI: 10.1021/jacs.7b03208] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
CD22 is a sialic acid-binding immunoglobulin-like lectin (Siglec) that is highly expressed on B-cells and B cell lymphomas, and is a validated target for antibody and nanoparticle based therapeutics. However, cell targeted therapeutics are limited by their complexity, heterogeneity, and difficulties in production. We describe here a chemically defined natural N-linked glycan scaffold that displays high affinity CD22 glycan ligands and outcompetes the natural ligand for the receptor, resulting in single molecule binding to CD22 and endocytosis into cells. Binding affinity is increased by up to 1500-fold compared to the monovalent ligand, while maintaining the selectivity for hCD22 over other Siglecs. Conjugates of these multivalent ligands with auristatin and saporin toxins are efficiently internalized via hCD22 resulting in killing of B-cell lymphoma cells. This single molecule ligand targeting strategy represents an alternative to antibody- and nanoparticle-mediated approaches for delivery of agents to cells expressing CD22 and other Siglecs.
Collapse
Affiliation(s)
- Wenjie Peng
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - James C Paulson
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
47
|
Leveraging Siglec-8 endocytic mechanisms to kill human eosinophils and malignant mast cells. J Allergy Clin Immunol 2017; 141:1774-1785.e7. [PMID: 28734845 DOI: 10.1016/j.jaci.2017.06.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 06/01/2017] [Accepted: 06/12/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a cell-surface protein expressed selectively on human eosinophils, mast cells, and basophils, making it an ideal target for the treatment of diseases involving these cell types. However, the effective delivery of therapeutic agents to these cells requires an understanding of the dynamics of Siglec-8 surface expression. OBJECTIVES We sought to determine whether Siglec-8 is endocytosed in human eosinophils and malignant mast cells, identify mechanisms underlying its endocytosis, and demonstrate whether a toxin can be targeted to Siglec-8-bearing cells to kill these cells. METHODS Siglec-8 surface dynamics were examined by flow cytometry using peripheral blood eosinophils, mast cell lines, and Siglec-8-transduced cells in the presence of inhibitors targeting components of endocytic pathways. Siglec-8 intracellular trafficking was followed by confocal microscopy. The ribosome-inhibiting protein saporin was conjugated to a Siglec-8-specific antibody to examine the targeting of an agent to these cells through Siglec-8 endocytosis. RESULTS Siglec-8 endocytosis required actin rearrangement, tyrosine kinase and protein kinase C activities, and both clathrin and lipid rafts. Internalized Siglec-8 localized to the lysosomal compartment. Maximal endocytosis in Siglec-8-transduced HEK293T cells required an intact immunoreceptor tyrosine-based inhibitory motif. Siglec-8 was also shuttled to the surface via a distinct pathway. Sialidase treatment of eosinophils revealed that Siglec-8 is partially masked by sialylated cis ligands. Targeting saporin to Siglec-8 consistently caused extensive cell death in eosinophils and the human mast cell leukemia cell line HMC-1.2. CONCLUSIONS Therapeutic payloads can be targeted selectively to eosinophils and malignant mast cells by exploiting this Siglec-8 endocytic pathway.
Collapse
|
48
|
Denis M, Mullaivanam Ramasamy S, Thayappan K, Munusamy A. Immune response of anti-lectin Pjlec antibody in freshwater crab Paratelphusa jacquemontii. Int J Biol Macromol 2017; 104:1212-1222. [PMID: 28690166 DOI: 10.1016/j.ijbiomac.2017.07.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 01/26/2023]
Abstract
Sialic acid specific lectin Pjlec isolated from serum of the freshwater crab Paratelphusa jacquemontii served as an antigen for the production of immunoglobulin (Ig) in Balb/c mice sera. Enzyme-linked immunosorbent assay (ELISA) of mice anti-sera with Pjlec lectin affirmed the induction and production of antibody. Anti-Pjlec antibody was isolated from the antisera of mice by Protein A Sepharose affinity chromatography and checked for purity by immunoblot with lectin. Mass spectrometry (MS/MS) of papain digethe peptide sequence of antigen binding fragment (Fab) and fragment crystallizable (Fc). Coatingsted anti-Pjlec revealed of anti-Pjlec to the target cell, rabbit erythrocyte failed to enhance in vitro phagocytosis in the crab. However, inoculation of anti-Pjlec in the hemolymph of the crab elicited in vitro phagocytosis. Proteins in hemocyte lysate supernatant (HLS) were separated by electrophoresis failed to immunoblot with Pjlec or anti-Pjlec. Peptide sequences of trypsin digested lectin protein appeared homologous to deuterostome chordate. The protostome crab that lack the ability to synthesize sialic acid however bind to sialic acid a deuterostome sugar to suggest the complexity in innate immune system of invertebrates. The application of lectin and its antibody require further study on application of pathological conditions associated with alterations in sialylated cell surface.
Collapse
Affiliation(s)
- Maghil Denis
- Laboratory of Pathobiology, Department of Zoology, University of Madras, Chennai, Tamil Nadu 600025, India.
| | | | - Karthigayani Thayappan
- Laboratory of Pathobiology, Department of Zoology, University of Madras, Chennai, Tamil Nadu 600025, India
| | - Arumugam Munusamy
- Laboratory of Pathobiology, Department of Zoology, University of Madras, Chennai, Tamil Nadu 600025, India
| |
Collapse
|
49
|
Giltiay NV, Shu GL, Shock A, Clark EA. Targeting CD22 with the monoclonal antibody epratuzumab modulates human B-cell maturation and cytokine production in response to Toll-like receptor 7 (TLR7) and B-cell receptor (BCR) signaling. Arthritis Res Ther 2017; 19:91. [PMID: 28506291 PMCID: PMC5433084 DOI: 10.1186/s13075-017-1284-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 03/24/2017] [Indexed: 12/03/2022] Open
Abstract
Background Abnormal B-cell activation is implicated in the pathogenesis of autoimmune diseases, including systemic lupus erythematosus (SLE). The B-cell surface molecule CD22, which regulates activation through the B-cell receptor (BCR), is a potential target for inhibiting pathogenic B cells; however, the regulatory functions of CD22 remain poorly understood. In this study, we determined how targeting of CD22 with epratuzumab (Emab), a humanized anti-CD22 IgG1 monoclonal antibody, affects the activation of human B-cell subsets in response to Toll-like receptor 7 (TLR7) and BCR engagement. Methods B-cell subsets were isolated from human tonsils and stimulated with F(ab′)2 anti-human IgM and/or the TLR7 agonist R848 in the presence of Emab or a human IgG1 isotype control. Changes in mRNA levels of genes associated with B-cell activation and differentiation were analyzed by quantitative PCR. Cytokine production was measured by ELISA. Cell proliferation, survival, and differentiation were assessed by flow cytometry. Results Pretreatment of phenotypically naïve CD19+CD10–CD27– cells with Emab led to a significant increase in IL-10 expression, and in some but not all patient samples to a reduction of IL-6 production in response to TLR7 stimulation alone or in combination with anti-IgM. Emab selectively inhibited the expression of PRDM1, the gene encoding B-lymphocyte-induced maturation protein 1 (Blimp-1) in activated CD10–CD27– B cells. CD10–CD27–IgD– cells were highly responsive to stimulation through TLR7 as evidenced by the appearance of blasting CD27hiCD38hi cells. Emab significantly inhibited the activation and differentiation of CD10–CD27–IgD– B cells into plasma cells. Conclusions Emab can both regulate cytokine expression and block Blimp1-dependent B-cell differentiation, although the effects of Emab may depend on the stage of B-cell development or activation. In addition, Emab inhibits the activation of CD27–IgD– tonsillar cells, which correspond to so-called double-negative memory B cells, known to be increased in SLE patients with more active disease. These data may be relevant to the therapeutic effect of Emab in vivo via modulation of the production of pro-inflammatory and anti-inflammatory cytokines by B cells. Because Blimp-1 is required by B cells to mature into antibody-producing cells, inhibition of Blimp1 may reduce autoantibody production. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1284-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Natalia V Giltiay
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA. .,Department of Immunology, University of Washington, Seattle, WA, 98109, USA.
| | - Geraldine L Shu
- Department of Immunology, University of Washington, Seattle, WA, 98109, USA
| | | | - Edward A Clark
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, 98109, USA.,Department of Immunology, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
50
|
Mikulak J, Di Vito C, Zaghi E, Mavilio D. Host Immune Responses in HIV-1 Infection: The Emerging Pathogenic Role of Siglecs and Their Clinical Correlates. Front Immunol 2017; 8:314. [PMID: 28386256 PMCID: PMC5362603 DOI: 10.3389/fimmu.2017.00314] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/06/2017] [Indexed: 11/25/2022] Open
Abstract
A better understanding of the mechanisms employed by HIV-1 to escape immune responses still represents one of the major tasks required for the development of novel therapeutic approaches targeting a disease still lacking a definitive cure. Host innate immune responses against HIV-1 are key in the early phases of the infection as they could prevent the development and the establishment of two hallmarks of the infection: chronic inflammation and viral reservoirs. Sialic acid-binding immunoglobulin-like lectins (Siglecs) belong to a family of transmembrane proteins able to dampen host immune responses and set appropriate immune activation thresholds upon ligation with their natural ligands, the sialylated carbohydrates. This immune-modulatory function is also targeted by many pathogens that have evolved to express sialic acids on their surface in order to escape host immune responses. HIV-1 envelope glycoprotein 120 (gp120) is extensively covered by carbohydrates playing active roles in life cycle of the virus. Indeed, besides forming a protecting shield from antibody recognition, this coat of N-linked glycans interferes with the folding of viral glycoproteins and enhances virus infectivity. In particular, the sialic acid residues present on gp120 can bind Siglec-7 on natural killer and monocytes/macrophages and Siglec-1 on monocytes/macrophages and dendritic cells. The interactions between these two members of the Siglec family and the sialylated glycans present on HIV-1 envelope either induce or increase HIV-1 entry in conventional and unconventional target cells, thus contributing to viral dissemination and disease progression. In this review, we address the impact of Siglecs in the pathogenesis of HIV-1 infection and discuss how they could be employed as clinic and therapeutic targets.
Collapse
Affiliation(s)
- Joanna Mikulak
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Italy; Istituto di Ricerca Genetica e Biomedica, UOS di Milano, Consiglio Nazionale delle Ricerche (UOS/IRGB/CNR), Rozzano, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Rozzano , Italy
| | - Elisa Zaghi
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Rozzano , Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| |
Collapse
|